![]() Compositions and methods relating to STOP-1
专利摘要:
公开号:AU2004232962A1 申请号:U2004232962 申请日:2004-04-16 公开日:2004-11-04 发明作者:Heidi Ackerly;Avi Ashkenazi;David Eberhard;Gretchen Frantz;Dorothy French;Germaine Fuh;Jo-Anne Hongo;Chingwei Lee;Scot Marsters;Robert Pitti;Helga Raab;Liliana Soroceanu;Evgeny Varfolomeev;Beni Wolf 申请人:Genentech Inc; IPC主号:G01N33-53
专利说明: WO 2004/094476 PCT/US2004/011793 COMPOSITIONS AND METHODS RELATING TO STOP-1 This application claims benefit from United States Provisional Application No. 60/463,656, 5 filed April 16, 2003, which application is incorporated by reference herein in its entirety. FIELD OF THE INVENTION The present invention is directed to STOP-1 polypeptides, antibodies, nucleic acid molecules, antagonists, agonists, potentiators and compositions relating to STOP-1, and 10 methods of making and using the same, including methods for diagnosing and treating of tumors in mammals. The present invention further relates to the diagnosis and treatment of disorders involving angiogenesis and vasculogenesis (e.g., cardiovascular as well as oncological disorders). 15 BACKGROUND AND INTRODUCTION OF THE INVENTION Uncontrolled cell growth is the cause of many illnesses in a variety of cell types. For example, cancer occurs when there is an increase in the number of abnormal, or neoplastic, cells derived from a normal tissue that proliferate to form a tumor mass. The tumor cells often invade the adjacent tissues and can spread via the blood or lymphatic system to regional 20 lymph nodes and to distant sites via a process called metastasis. In a cancerous growth, 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. 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)). 25 Much research has been devoted to discovering new treatments for cell proliferative disorders, such as cancer. Despite recent advances, there is a great need to identify and understand the role of new cellular targets for modulating cell proliferation and to develop alternative or more effective methods of treatment and therapeutic and diagnostic agents. There is also a need to develop alternative therapeutics and methods for treating specific cell 30 types and for treating illnesses caused by or associated with abnormal cell proliferation, such as cancers. For example, desmoplasia is the hyperplasia of fibroblasts and disproportionate formation of fibrous connective tissue, especially in the stroma of carcinomas. Desmoplasia is a hallmark of tumor invasion and malignancy. Desmoid tumors and abdominal fibroids are nodules or relatively large masses of unusually firm scarlike connective tissue resulting from 1 WO 2004/094476 PCT/US2004/011793 active proliferation of fibroblasts, occurring most frequently in the abdominal muscles of women who have borne children; the fibroblasts infiltrate surrounding muscle and fascia. In post-natal life, vasculogenesis (endothelial cells forming a primary tubular network) and angiogenesis (the growth or sprouting of new blood vessels from existing 5 vessels) play critical roles in the pathophysiology of neoplastic disorder (Semenza, G.L., (2003) Ann. Rev. Med. 54:17-28). The distinction between vasculogenesis and angiogenesis is not absolute and they overlap (Ribatti, D et al., (2001) Mech.Dev. 100:157-163). Both require endothelial cell proliferation, migration, three-dimensional reorganization of newly formed aggregates and use simlar extracellular matrix adhesive mechanisms (Ribatti, supra). 10 Use of anti-angiogenic therapies such as the antibody against vascular endothelial growth (VEGF) called Avastin have been shown to be useful in treating cancers. Another cellular protein, referred to herein as STOP-1 or UNQ762, has been shown to be overexpressed in certain tumors (e.g., WO 01/163318, WO 01/68848, WO 02/00690, WO 02/08284, WO 02/16602, WO 02/42487). Polyclonal antibodies against STOP-1 have been 15 reported (e.g., WO 02/42487). Although there has been some discussion of targeting STOP-1 to treat cancers and diseases associated with angiogenesis (e.g., WO 01/163318, WO 01/68848, WO 02/00690, WO 02/08284, WO 02/16602, WO 02/42487, WO 00/71581, WO 02/00690), there is a need to further explore the biology of the STOP-1 protein to identify alternative and more effective therapeutic agents and methods for diagnosis and treatment of 20 uncontrolled cell growth and diseases caused by, associated with or complicated by excessive and insufficient angiogenesis. The present invention addresses these needs and others by providing new STOP-1 polypeptides, antibodies, nucleic acid molecules, compositions and methods that incorporate further knowledge about the STOP-1 protein. Among other things, the present disclosure 25 shows that STOP-1 is overexpressed in the stroma of several tumor types. The present disclosure shows that overexpression of STOP-1 alone can be tumorigenic. Further, the present disclosure demonstrates that the STOP-1 protein can be secreted and that secretion is required for tumorigenesis. Still further, the present disclosure shows that the glycosylation state of STOP-1 affects whether it is secreted and that elimination of a N-glycosylation site, 30 e.g., by substituting the amino acid at position 186 (Asn) with alanine results in loss of secretion. The present disclosure shows that disulfide bonding between STOP-1 proteins can occur at a cysteine 55 in culture in the triple helix domain of STOP-1. Additionally, the present disclosure shows that the STOP-1 protein can form a complex with itself as a dimer, trimer and hexamer and that the c-terminus of the protein is sufficient for oligomerization, 2 WO 2004/094476 PCT/US2004/011793 whereas a region related to the triple helix domain of collagen is not required. The present disclosure also shows a plurality of agents that specifically bind to STOP-1, including the C terminal region and N-terminal region of the protein as well as nucleic acid and protein sequences encoding them. Further, the present disclosure shows that STOP-1 expression can 5 be modulated by overexpression of proteins in the WNT signalling pathway that are know to cause breast cancer in mice, e.g., the overexpression of WNT. Additionally, the present disclosure shows that STOP-1 can be cleaved by proteases that are overexpressed in the same tumors as STOP-1, e.g., MMP-9. Further, the present disclosure shows that a method for producing STOP-1 polypeptides by expressing the polypeptides in proteoglycan synthesis 10 deficient cell lines. The present invention shows that STOP-1 binds to the surface of cells, such as cancer cells and endothelial cells. The present invention provides antagonistic molecules that can inhibit the interaction of STOP-1 with the surface of cells. The present invention provides molecules that can potentiate the binding of STOP-1 with the surface of cells. The present invention also relates to the role of STOP-1 in angiogenesis and 15 vasculogenesis and methods and compositions for treating disorders for which treatment would be improved by modulating angiogenesis and vasculogenesis. This data and others provided herein, together with other disclosure of in present application, teach new, better and/or alternative methods for using the STOP-1 protein or compositions relating thereto. 20 SUMMARY OF THE INVENTION The present invention provides new therapeutic agents, diagnostic agents and methods for treating or preventing uncontrolled cell proliferation, including cancer, and other diseases by targeting the activity, expression and regulation of STOP-1. The present invention provides new therapeutic agents, diagnostic agents and methods for treatinga any medical 25 condition having suboptimal, excessive or inappropriate angiogenic or vasculogenic events by targeting the activity, expression and regulation of STOP-1. According to one embodiment, the present invention provides a monoclonal antibody that specifically binds to an oligomeric form of human STOP-1. According to another embodiment, the present invention provides, a monoclonal antibody that specifically binds to 30 amino acids 33-53 or 33-52 of human STOP-1. In yet another embodiment, the present invention includes a monoclonal antibody that specifically binds to amino acids 94-243 of human STOP-1. According to further embodiment, the monoclonal antibody that specifically binds to residues 94-243 of human STOP-1 or residues 33-53 or 33-52 of human STOP-1 also recognizes an oligomeric form of human STOP-1, such as the trimeric form. An 3 WO 2004/094476 PCT/US2004/011793 antibody according to this invention can be isolated. It is understood that an aforementioned antibody that specifically binds a residue within residues 33-52 or 33-53 of human STOP-1 may also bind to other residues within STOP-1 or non-human equivalents thereof. In yet another embodiment, the present invention provides monoclonal antibodies 5 having the biological characteristics of an antibody selected from the group consisting of S7 encoded by the nucleic acid molecule deposited on March 25, 2003 as designation V0350-4 S7, S4 encoded by the nucleic acid molecule deposited on March 25, 2003 as designation V0350-2b-S4, S9 encoded by the nucleic acid molecule deposited on March 25, 2003 as designation V0350-2b-S9, S16 encoded by the nucleic acid molecule deposited on March 25, 10 2003 as designation V0350-4-S16, F5 encoded by the nucleic acid molecule deposited on March 25, 2003 as designation V0350-5 and 6B 12 produced by the hybridoma cell line deposited on March 28, 2003 as designation 6B12.1.7 in the American Type Culture Collection (ATCC), 10801 University Blvd., Manassas, VA 20110-2209, USA, including the deposited antibodies, antibodies comprising a portion of those antibodies and variants 15 thereof. In another embodiment, the present invention provides antibodies that specifically bind to STOP-1, wherein the binding of the antibodies to STOP-1 can be inhibited (e.g., as observed in a competitive ELISA asay) by a second monoclonal antibody selected from one of the aforementioned deposited antibodies. The present invention also relates to antibodies having the following sequences: 20 A monoclonal antibody comprising: (a) a first amino acid sequence comprising: T - I- X1 - X2- X3-X4 25 wherein X1 is S, N or T; wherein X2 is G, N, S or A; wherein X3 is Y, S or T; and wherein X4 is D or W. 30 (b) a second amino acid sequence comprising: X1-X2-I-X3-P-X4-X5-G-X6-T-X7 (SEQ ID NO: 115) 4 WO 2004/094476 PCT/US2004/011793 wherein X1 is G or A; wherein 5 (1) X2 is an amino acid selected from the group consisting of S, T, A, and X3 is an amino acid selected from the group consisting of R, W and Y; or (2) X3 is an amino acid selected from the group consisting of S, T, A, and X2 is an amino acid selected from the group consisting of R, W and 10 Y; wherein X4 is Y or F; wherein X5 is G, S, T or A; wherein X6 is N, Y or A; and 15 wherein X7 is N, Y or D; and (c) a third amino acid sequence comprising the sequence: 20 C-X1-X2-X3-G-G-X4-X5-X6-X7-X8-X9-XO10-X11 (SEQ ID NO:116) wherein X1 is A, S or T; wherein X2 is basic amino acid; wherein X3 is any amino acid; 25 wherein X4 is a hydrophobic amino acid; wherein any one of X5- X8 can be any amino acid or can be missing, and at least one of X5-X8 is an aromatic amino acid or a hydrophobic amino acid; wherein X9 is an aromatic or hydrophobic amino acid; 30 wherein X10 is D or A; and wherein Xli1 is Y or V. 5 WO 2004/094476 PCT/US2004/011793 According to one embodiment, the monoclonal antibody comprises the light chain sequence of FIG.34. According to another embodiment, the monoclonal antibody is a full length IgG. According to one embodiment of this invention, the X1 of the first amino acid 5 sequence is S. According to another embodiment of this invention, the X2 the first amino acid sequence is G. According to yet another embodiment of this invention, X3 of the first amino acid sequence is S. According to one embodiment, the first amino acid sequence is a sequence selected from the group consisting of TISGSD, TITNSD and TISGSW. According to yet another embodiment of this invention, X3 of SEQ ID NO:115 is S or 10 A. According to yet another embodiment of this invention, X4 of SEQ ID NO: 115 is Y. According to yet another embodiment of this invention, X5 of SEQ ID NO:115 is G or A. According to yet another embodiment of this invention, X6 of SEQ ID NO: 115 is N or A. According to one embodiment, SEQ ID NO: 115 is a sequence selected from the group consisting of GRISPYGNTN, ATIYPYGGYTY and AWIAPYSGATD. 15 According to one embodiment of this invention, the X1 of SEQ ID NO:116 is A. According to another embodiment of this invention, the X2 of SEQ ID NO: 116 is R. According to yet another embodiment of this invention, X4 of SEQ ID NO: 116 is L or M. According to one preferred embodiment of this invention, the aromatic amino acid present in X5-X8 is a tryptophan residue. According to another embodiment, one amino acid of X5-X8 20 is missing. According to yet another embodiment of this invention, X9 of SEQ ID NO: 116 is F. According to one embodiment of this invention, X10 of SEQ ID NO:116 is D. According to one embodiment of this invention, X11 of SEQ ID NO: 116 is Y. According to one embodiment, the SEQ ID NO: 116 is a sequence selected from the group consisting of CARVGGLKLLFDY, CARGGGMDGYVMDY and CAREGGLYWVFDY. 25 An antibody according to this invention can comprise (a) a first amino acid sequence comprising the sequence TISGSD; (b) a second amino acid sequence comprising the sequence GRISPYGNTN; and (c) a third amino acid sequence comprising the sequence CARVGGLKLLFDY, or a variant of said antibody. Alternatively, an antibody according to this invention can comprise (a) a first amino acid sequence comprising the sequence 30 TITNSD; (b) a second amino acid sequence comprising the sequence ATIYPYGGYTY; and (c) a third amino acid sequence comprising the sequence CARGGGMDGYVMDY; or a variant of said antibody. Alternatively, an antibody according to this invention can comprise (a) a first amino acid sequence comprising the sequence TISGSW; (b) a second amino acid sequence comprising the sequence AWIAPYSGATD; and (c) a third amino acid sequence 6 WO 2004/094476 PCT/US2004/011793 comprising the sequence CAREGGLYWVFD; or a variant of said antibody. Alternatively, an antibody according to this invention can comprises (a) a first amino acid sequence comprising the sequence TISNYG; (b) a a second amino acid sequence comprising the sequence GRISPSNGSTY; and (c) a third amino acid sequence comprising the sequence 5 CAKCSVRFAY; or a variant of said antibody. Alternatively, an antibody according to this invention can comprise (a) a first amino acid sequence comprising the sequence TINNYD; (b) a second amino acid sequence comprising the sequence GYISPPSGATY; and (c) third amino acid sequence comprising the sequence CARMVGMRRGVMDY; or a variant of said antibody. 10 In a further embodiment, the first, second and third amino acid sequences described above are located in a human heavy chain wherein the first amino acid sequence is at residues 28-33 of the heavy chain according to the Kabat numbering system, the second amino acid sequence is at residues 49-58 of the heavy chain according to the Kabat numbering system and the third amino acid sequence is at residues 92-102 according to the Kabat numbering 15 system. In another embodiment, the present invention provides a monoclonal antibody comprising the amino acid sequence of: (a) the heavy chain sequence of FIG.27; (b) the heavy chain sequence of FIG.28; (c) the heavy chain sequence of FIG.29; (d) the heavy chain sequence of FIG.30; (e) the heavy chain sequence of FIG.31; or (f) the heavy chain sequence 20 of FIG.34; or variants thereof. In a further embodiment, the antibodies of this invention further comprise (a) the light chain sequence of FIG.27, (b) the light chain sequence of FIG.34; or variants thereof. In a further embodiment, the antibodies of this invention are chimeric or humanized antibodies. In another embodiment, the antibodies of this invention are antibody fragments. 25 In yet another embodiment of this invention, the antibodies are conjugated to an agent selected from the group consisting of a stromal targeting agent, a growth inhibitory agent, a cytotoxic agent, a detection agent, an agent that improves the bioavailability and an agent that improves the half-life of the antibody. In another embodiment, the antibody of this invention is a multi-specific antibody having a binding specificity for a STOP-1 polypeptide and one or 30 more binding specificities for any other antigen. According to one embodiment, the other antigen is a cell-surface protein or receptor or receptor subunit. According to one preferred embodiment, the cell-surface protein is a natural killer (NK) receptor receptor. According to a more preferred embodiment, the binding of the antibody to the NK receptor activates the natural killer cell. 7 WO 2004/094476 PCT/US2004/011793 The present invention provides variants and modifications of STOP-1 polypeptide variants. In one embodiment, the STOP-1 polypeptide variant that cannot be secreted from a cell. In another embodiment, said variant is a human STOP-1 polypeptide that is not glycosylated. In a further embodiment, the variant is a human STOP-1 polypeptide that is 5 mutated at residue 186. The present invention also provides a STOP-1 variant polypeptide comprising STOP-1 that cannot disulfide bind with another STOP-1. According to one embodiment, the variant is a human STOP-1 polypeptide that is mutated at residue 55. The present invention also provides nucleic acid molecules encoding the antibodies and polypeptides and variants thereof, vectors comprising the nucleic acid molecules, and 10 host cells comprising the nucleic acid molecules of this invention. The present invention includes compositions comprising an antibody, a polypeptide or a nucleic acid molecule of this invention. According to one embodiment, the composition further comprises a pharmaceutically acceptable carrier. In a further embodiment, the composition comprises a stromal targeting agent. In a further embodiment, the stromal 15 targeting agent is covalently linked to the monoclonal antibody or polypeptide. In yet a further embodiment, the stromal targeting agent recognizes a stromal cell of a tumor. The present invention provides methods for producing a STOP-1 polypeptide or an anti-STOP-1 antibody of this invention by culturing a cell comprising a nucleic acid according to this invention. According to one embodiment, the method for producing a 20 STOP-1 polypeptide comprises the step of culturing a mammalian cell that comprises a nucleic acid molecule encoding the STOP-1 polypeptide and that is deficient in proteoglycan synthesis. According to another embodiment, the cell line that is deficient in proteoglycan synthesis is deficient in galactosyltransferase I activity. According to one preferred embodiment, the cell line is a CHO-psbg cell line. 25 The present invention provides a method for determining the presence of a STOP-1 polypeptide in a sample comprising exposing a sample suspected of containing the STOP-1 polypeptide to an anti-STOP-1 antibody and determining binding of said antibody to a component of said sample. According to one embodiment, the antibody is a monoclonal antibody of this invention. 30 The present invention provides methods for diagnosing or monitoring a cell proliferative disorder, such as a tumor, of a patient comprising the step of comparing the expression of STOP-1 in a normal tissue to the amount of STOP-1 being tested from the patient. In one embodiment, a STOP-1 protein can be detected by an agent such as an antibody of this invention. In another embodiment, STOP-1 mRNA can be detected by an 8 WO 2004/094476 PCT/US2004/011793 agent such as a nucleic acid molecule that specifically hybridizes to the UNQ6762 mRNA. In a further embodiment, the tumor being tested has a large stromal compartment. In a further embodiment, STOP-1 detection agent is administered at or near the stromal compartment of the tissue being tested. In yet another embodiment, the method further 5 comprises the step of observing or assaying the STOP-1 protein or mRNA in the stromal compartment agent of the normal tissue and tissue being tested. In another embodiment, the antibody is a monoclonal antibody of this invention. The present invention provides a method of preventing or treating a proliferative disorder in a patient comprising the step of admininstering to the patient a 10 composition of this invention in an amount effective to inhibit the proliferation of cells in the patient. In one embodiment, the proliferative disorder is desmoplasia. The present invention also provides a method of preventing or inhibiting the growth of a tumor that overexpresses STOP-1 in a patient comprising administering to the patient an antagonist of STOP-1 in an amount effective to inhibit growth of the tumor in the patient. In a further embodiment, the 15 tumor to be treated has stromal compartments. In yet a further embodiment, the tumor having stromal compartments is selected from the group consisting of desmoid tumors, pancreatic cancer, sarcomas (e.g., hemangiosarcoma, rabdomyosarcoma) and adenocarcinomas (mammary adenocarcinomas, colon adenocarcinomas, gastrointestinal adenocarcinomas and ovarian adenocarcinomas), hepatocellular carcinoma, breast cancer, 20 colon cancer, lung cancer, ovarian cancer, glioma, endometrial cancer and vascular cancer. In a further embodiment, the antagonist is administered at or near a stroma of the tumor. In another embodiment, the tumor is a melanoma or a round cell tumor (e.g., malignant fibrous hystiocytoma). An antagonist according to this invention is any molecule that partially or fully 25 blocks, inhibits, or neutralizes a biological activity of a native STOP-1 polypeptide and that specifically binds to a native STOP-1 polypeptide, wherein the binding of the antagonistic molecule (1) is to a native STOP-1 polypeptide in oligomeric form, (2) is to residues 94-243 of native human STOP-1 and/or (3) can be inhibited (e.g., as observed in a competitive ELISA assay using STOP-1 and 6B 12) by a monoclonal antibody of this invention (e.g., a 30 deposited antibody of this invention, etc.). According to one embodiment, the deposited antibody is the 6B12 antibody. According one embodiment, the antagonist is a polypeptide. According to another embodiment, the antagonist is an antibody of this invention. In another embodiment, the STOP-1 polypeptide that the antagonist inhibits is part of a trimeric complex. 9 WO 2004/094476 PCT/US2004/011793 According to another embodiment, the biological activity that is inhibited by the antagonist is the interaction of STOP-1 with a cell that specifically binds STOP-1. According to one embodiment, the cell is a breast cancer cell. According to another embodiment, the cell is an endothelial cell. According to yet another embodiment, the antagonist has an 5 additional property selected from the group consisting of (1) capable of binding to an epitope within human STOP-1 that the 6B 12 antibody binds; (2) capable of binding to a residue within at least residues 33-52 of human STOP-1; and (3) capable being competed from binding to STOP-1 by the 6B 12 antibody (e.g., as observed in a competitive ELISA assay using STOP-1, the antagonist and the 6B 12 antibody). 10 The present invention provides a method for inhibiting the growth of a cell that overexpresses STOP-1 comprising the step of inhibiting the secretion of STOP-1 from the cell. In one embodiment, secretion is inhibited by inhibiting glycosylation of STOP-1. In another embodiment, the secretion is inhibited by overexpressing a STOP-1 protein that cannot be secreted in the cell. In a further embodiment, the secretion is inhibited by a STOP 15 1 protein that is mutated at residue 186. The present invention provides a method for preventing disulfide binding between STOP-1 molecules comprising a step selected from the group consisting of: (1) mutating STOP-I-encoding DNA molecules at residue cysteine 55; (2) expressing STOP-1 proteins that are mutated at residue cysteine 55 in the presence of naturally-occurring STOP-1 20 proteins; and (3) incubating STOP-1 proteins that are mutated at residue cysteine 55 with naturally-occurring STOP-1 proteins. The present invention provides a method for cleaving STOP-1 comprising the step of incubating STOP-1 with a protease selected from the group consisting of a matrix metalloprotease-7 (MMP-7) and a matrix metalloprotease-9 (MMP-9). In a further 25 embodiment, the method additionally comprises the step of monitoring the STOP-1 cleavage products produced. Another embodiment of the present invention is directed to a method for inhibiting the growth of a cell that overexpresses a STOP-1 polypeptide, wherein the method comprises administering an antagonist of STOP-1, wherein the antagonist specifically binds to STOP-1 30 and is optionally conjugated to one or a combination of the agent(s) selected from the group consisting of a stromal targeting agent, a growth inhibitory agent or cytotoxic agent such as a toxin, including, for example, a maytansinoid or calicheamicin, an antibiotic, a radioactive isotope and a nucleolytic enzyme. Another embodiment of the present invention is directed to a method for inhibiting the growth of a cell that overexpresses a STOP-1 polypeptide, 10 WO 2004/094476 PCT/US2004/011793 wherein the method comprises administering an agent to a stromal cell of the tumor, wherein the agent is an antagonist of STOP-1 or a nucleic acid molecule encoding a STOP polypeptide. The agent can be administered to the stromal cell directly by a patient or physician or indirectly, through the use of stromal targeting agents that can direct the agent to 5 the stromal cell. The present invention provides an article of manufacture comprising (a) a composition of matter comprising a modified STOP-1 polypeptide, a STOP-1 polypeptide variant, STOP-1 antagonist, STOP-1 agonist, STOP-1 potentiator or a nucleic acid molecule encoding a STOP-1 polypeptide conjugated to a vehicle (e.g., such as antisense therapy or RNAi therapy); (b) a container containing said composition; and (c) a label affixed to said 10 container, or a package insert included in said container referring to the use of said polypeptide variant, modified polypeptide or antagonist in the treatment of a proliferative disorder or a disease associated with abnormal angiogenesis or vasculogenesis (e.g., a package insert). According to one embodiment, the STOP-1 antagonist or potentiator is an antibody of this invention. 15 The present invention provides methods for testing the activity of STOP-1 and agonists or antagonists of STOP-1. In one embodiment, a method of inducing cell migration in vitro comprising administering to an endothelial cell a STOP-1 polypeptide in an amount effective to induce migration of said cell is provided. According to another embodiment, the present invention provides a method of testing the activity of a candidate antagonist or 20 agonist of STOP-1 comprising the steps of treating a first endothelial cell with STOP-1, treating a second endothelial cell with STOP-1 and the candidate antagonist or agonist, and comparing the migration of the first and second endothelial cells. In one preferred embodiment, the cell used in such migration assay is a HUVEC cell. The present invention also provides methods of treating a disease or condition 25 associated with excessive, inappropriate or uncontrolled angiogenesis in a mammalian subject. In one embodiment, the method comprises the step of administering to the subject a STOP-1 antagonist in an amount effective to treat the disease, wherein the STOP-1 antagonist has any property selected from the group consisting of (1) binds to residues within human STOP-1 that the 6B12 antibody binds; (2) binds to a residue within at least residues 33-52 of 30 human STOP-1; and (3) can be inhibited from binding to STOP-1 by the 6B12 antibody. The present invention also contemplates treating patients who would benefit from increased angiogeneis vasculogenesis by administering a therapeutically effective amount of a STOP-1 potentiator, a molecule that enhances STOP-1 binding to cells and/or aggregates STOP-1 on the cell surface. Such a molecule would be administered in an amount effective 11 WO 2004/094476 PCT/US2004/011793 to increase angiogenesis or vasculogenesis. In one preferred embodiment, the agonist is an anti-STOP-1 antibody that aggregates STOP-1 a cell surface. The present invention also provides agonists comprising an oligomeric form of STOP-1 polypeptide that comprises greater than three STOP-1 polypeptides. According to 5 one embodiment the agonist comprises six STOP-1 polypeptides. According to another embodiment, the STOP-1 polypeptide is part of an immunoadhesin that is used to form said agonist. The present invention provides new methods for identifying and evaluating candidate and know STOP-1 antagonists, agonist and potentiators comprising the step of observing or 10 measuring the binding of STOP-1 to a cell in the presence and absence of the antagonist, agonist or potentiator. According to one embodiment, the cell is a cancer cell. In a further embodiment the cell is a breast cancer cell. According to another embodiment, the cell is an endothelial cell. 15 BRIEF DESCRIPTION OF THE DRAWINGS FIG. 1 shows an alignment of amino acid sequences encoding STOP-1 from a wide variety of species - human (SEQ ID NO: 1), mouse (SEQ ID NO:3), rice fish (SEQ ID NO:4), zebrafish (SEQ ID NO:5) and chicken (SEQ ID NO:6). A consensus sequence is also 20 provided. The arrow indicates a signal sequence cleavage site. Red indicates residues conserved in all species. Capitalized letters in the consensus sequence indicates residues that have been conserved throughout all species. Lower case letters in the consensus sequence indicate residues that are conserved in most species. Residues that that are not conserved in those species appear as a "period." "!" indicates I or V. "$" indicates L or M." %" indicates 25 F or Y. "#" indicates B, D, E, N, Q or Z. FIG.2 shows a nucleic acid sequence encoding human STOP-1. A signal sequence is indicated by the boxed amino acids. A triple helix domain is indicated by an underline. A glycosylation site is at amino acid 186. 30 FIG.3 shows (A) the presence of human STOP-1 mRNA in certain tissue types and (B) mouse STOP-1 mRNA from different stages of mouse development. Full length human or mouse STOP-1 DNAs were radiolabelled and used to probe northern blots of tissues from adult humans or developing mouse embryos. 12 WO 2004/094476 PCT/US2004/011793 FIG.4 shows coomassie-stained human STOP-1 proteins produced by (A) CHO-DP12 or 03(B) CHO-psgb (ATCC) cells and purified by nickel-NTA affinity chromatography. The vector, pRK5, was used as a control. 5 FIG.5 shows a western blot of human histidine-tagged STOP-1 protein present in the (A) supernatant and (B) cell lysate of transient transfected CHO-psgb cells. The western blot was probed with anti-his antibody. 10 FIG.6 shows the oligomerization of human STOP-1 protein expressed using a baculoviral infection system in SF9 insect cells. STOP-1 protein and various deletion mutants were expressed from SF9 cells, separated on a size exclusion column and subjected to light scattering analysis e.g., (A) S31-K243, (B) E89-K243 and (C) L94-K243. The predicted molecular weight of the monomers appear in the left corner of each graph. The 15 numbers appearing next to several peaks refer to the average molecular weight of the complexes in the peak FIG.7 shows the oligomerization of human STOP-1 protein expressed from mammalian cells. Human STOP-1 protein and various deletion mutants were expressed from 20 CHO cells, separated on a size exclusion column and subjected to light scattering analysis, e.g., (A) M1-K243 and (B) delta-THD (residues 1-54, 94-243, plus histidine tag). The predicted molecular weight of the monomers appear in the left corner of each graph. The numbers appearing next to several peaks refer to an average molecular weight of the complexes in those peaks. Under non-reducing conditions, western blots of secreted full 25 length, his-tagged human STOP-1 protein recombinantly expressed from CHO-psgb cells presented predominantly homodimerized complexes (C). The western blots were probed with anti-his antibody. FIG.8 shows western blots of (A) cell culture media and (B) whole cell lysates from 30 CHO-psgb cells expressing human his-tagged STOP-1 WT, delta-THD, and delta-delta-THD STOP-1 (residues 1-51, 94-243, plus histidine tag) and subjected to reducing or non-reducing conditions. The western blots were probed with anti-his antibody. 13 WO 2004/094476 PCT/US2004/011793 FIG.9 shows western blots of (A) cell culture media and (B) whole cell lysates from CHO-psgb cells expressing his-tagged WT, G53A and N186A STOP-1 constructs and subjected to reducing or non-reducing conditions. The western blots were probed with anti his antibody. 5 FIG. 10 shows western blots of (A) cell culture media and (B) whole cell lysates from CHO-psgb cells expressing his-tagged WT, C55A, C93A and C109A STOP-1 constructs and subjected to reducing or non-reducing conditions. The western blots were probed with anti his antibody. 10 FIG. 11 shows that murine STOP-1 mRNA (mSTOP-1 mRNA) is expressed in breast tumors derived from MMTV-WNT1 transgenic mice but not in normal mammary epithelial cells. RNA samples were taken from breast tumor cells (marked "T1"-"T7") or C57Mg mouse normal mammary epithelial cells (marked, "N"), subjected to RT-PCR with mSTOP-1 15 primers and mRLPl9 primers. The PCR products were separated on an agarose gel. FIG.12 shows the proliferation of 3T3 cells after transfection with (A) human STOP-1 Sor (B) mouse STOP-1. FIG.12A shows the amount of 3 H-thymidine incorporation (counts per minute (cpm)) in 3T3 cells at 12, 28 and 96 hours after addition of of 3 H-thymidine. 20 FIG.12B shows the amount of 3 H-thymidine incorporation (counts per minute (cpm)) in 3T3 cells at 12, 28 and 96 hours after addition of 3 H-thymidine. Controls: transfections with vector alone (puro2 and phl). FIG. 13 shows the proliferation of 3T3 or 293 cells after infection with retrovirus 25 encoding human STOP-1. FIG.13A and B are western blots of human STOP-1 proteins expressed from 3T3 cells or 293, respectively, infected with a retrovirus encoding a control vector (Babe) or human STOP-1. STOP-1 was immunoprecipitated from whole cell lysates using the S7-IgG antibody. Western blots were probed with polyclonal anti-human STOP-1 antibodies. FIG. 13C shows the level of cell proliferation observed for the infected cell 30 populations as detected by a colorimetric Cell Titer Assay. FIG. 14 shows that mouse STOP-1 promotes tumorigenesis by 3T3 fibroblasts in a xenograft mouse model. FIG.14A shows the mean volume of tumors in mice implanted with 3T3 fibroblasts transfected with vector alone (p2 vector) or DNA encoding mouse STOP-1 or 14 WO 2004/094476 PCT/US2004/011793 RAS protein. The transfected cells were implanted into nude mice or tested for protein expression. FIG.14B and C show western blots of aliquots of the supernatants and lysates, respectively, of the transfected cells. The western blot was probed with rabbit anti-STOP-1 polyclonal antibodies. "TI" refers to the tumor incident ratio. 5 FIG.15 shows that human STOP-1 promotes tumorigenesis by 3T3 fibroblasts in a xenograft mouse model. FIG.15 shows the mean tumor volume of tumors in mice implanted with 3T3 fibroblasts transfected with vector alone or DNA encoding human STOP-1, RAS protein or LP1. 10 FIG.16 shows that recombinant STOP-1 protein potentiates SK-Mel-31 cells wound healing and motility. SK-Mel-31 cells were treated with (A) NT - no exogenous ligand treatment, (B) b762 - baculoviral produced human STOP-1 protein, (C) hrEGF - (50 ng/ml), (D) hrEGF and b762 or (E) CHO mammalian produced human STOP-1 protein. 15 FIG.17 shows that anti-human STOP-1 antibody, 6B12, binds to the N-terminal sequence of human STOP-1 between the signal sequence and triple helix domains. FIG.17A is a schematic of the his-tagged human and full length zebrafish STOP-1 proteins used in the epitope location studies of FIGs.18B and C. FIG.17B and C show western blots probed with 20 anti-his antibody and 6B 12 antibody, respectively, of extracts from cells that recombinantly expressed the proteins of FIG.17A. FIG. 18 shows the amino acid sequences of the CDRs of several phage-derived antibodies having affinity for human STOP-1. "H1," "H2" and "H3" refer to VH-CDR1, VH 25 CDR2 and VH-CDR3. The numerical header generally corresponds to amino acid positions 28-33, 49-58 and 92-102 according to the Kabat numbering system. FIG. 19 shows a graph of an ELISA assay to determine an optimal concentration of S4 and S7 Fab or IgG for use in a competition ELISA to determine the affinity of the antibodies 30 for STOP-1. "S coated" refers to a short form (#94-243) of STOP-1 coated on a microtiter plate. "F coated" refers to a full-length form of human STOP-1 coated on a microtiter plate. Approximately 90% of maximal binding was considered to be optimal for use in a competitive ELISA assay. Horse-radish peroxidase-conjugated protein G was used to detect the bound Fab and IgG. 15 WO 2004/094476 PCT/US2004/011793 FIG.20 shows a graph of the results of competitive ELISA to determine the binding affinities of the S4 and S7 Fab or IgG. The plates were coated with short form or full-length human STOP-1 and competed with short form or full length STOP-1, respectively (FIG.20A 5 and B, respectively). The calculated binding affinities are indicated in the parentheticals. FIG.21 shows a summary of the binding affinities of several phage-derived antibodies against STOP-1. "S/S" refers to an ELISA in which the microtiter plate was coated with a short form of STOP-1 and competed with a short form of STOP-1. "F/S" refers to an ELISA 10 in which the microtiter plate was coated with a full-length form of human STOP-1 and competed with a short form of human STOP-1. "F/F" refers to an ELISA in which the microtiter plate was coated with a full-length form of STOP-1 and competed with a full length form of STOP-1. The phage used in these studies were the S4-Fab phage and the S7 F(ab)' 2 phage. 15 FIG.22 shows a graph of an ELISA assay in which the plates were coated with human STOP-1, bound with S4 IgG and then competed with S4 (Fab) phage, S7 (F(ab)' 2 ) phage, S9 (Fab) phage, S16 (F(ab)' 2 ) phage and F5 (F(ab)' 2 ) phage. The Y axis refers to percentage unblocked as calculated by dividing the OD450nm value of the well that blocked S4 IgG by 20 the OD450nm value of a well without S4 IgG. FIG.23 shows a coomassie stained gel of baculovirus-expressed human STOP-1 protein cleaved by various proteases in vitro. "MMP" refers to matrix metalloprotease. 25 FIG.24A-D are schematics of phagemids encoding Fab of F(ab)' 2 phage display proteins or vectors encoding Fab or IgG proteins. FIG.24A is a schematic of a Fab-phagemid construct. The construct contains an alkaline phosphatase promoter, an STII signal sequence, a VL and CL light chain sequence, a gD tag, another STII signal sequence, a heavy chain VH and CH 1 region and a C-terminal part of the M13 bacteriophage plI coat protein (cP3). 30 FIG.24B is a schematic of a F(ab)' 2 -phagemid construct. The construct contains generally the same sequences as the Fab-phagemid, except it additionally includes a leucine zipper sequence (Zip). FIG.24C is a schematic of a nucleic acid molecule encoding a Fab protein. FIG.24D is a schematic of a nucleic acid molecule encoding an IgG protein, which IgG protein includes a CH 2 and CH 3 sequence. 16 WO 2004/094476 PCT/US2004/011793 FIG.25A-H describe amino acid sequences and a nucleic acid sequence for a phage display anti-Her-2 Fab. More specifically, FIG.25 shows an amino acid sequence comprising an anti-Her-2 Fab light chain (SEQ ID NO:86), an amino acid sequence comprising an anti 5 Her-2 Fab light chain region (SEQ ID NO:87) and the nucleic acid sequence of a phagemid encoding the amino acid sequences (SEQ ID NO:88). FIG.26A-H describe amino acid sequences and a nucleic acid sequence for a phage display anti-Her-2 F(ab)' 2 . More specifically, FIG.26 shows an amino acid sequence 10 comprising an anti-Her-2 F(ab)' 2 light chain (SEQ ID NO:89), an amino acid sequence comprising an anti-Her-2 F(ab)' 2 heavy chain region (SEQ ID NO:90) and the nucleic acid sequence of a phagemid encoding the amino acid sequences (SEQ ID NO:91). FIG.27A-C describe amino acid sequences and a nucleic acid sequence for a phage 15 display S4-Fab. More specifically, FIG.27 shows an amino acid sequence comprising an S4 Fab light chain (SEQ ID NO:92), an amino acid sequence comprising an S4-Fab heavy chain region (SEQ ID NO:93) and a nucleic acid sequence encoding the amino acid sequences (SEQ ID NO:94). 10 FIG.28A-C describe amino acid sequences and a nucleic acid sequence for a phage display S9 Fab. More specifically, FIG.28 shows an amino acid sequence comprising an S9 Fab light chain (SEQ ID NO:95), an amino acid sequence comprising an S9-Fab heavy chain region (SEQ ID NO:96) and a nucleic acid sequence encoding the amino acid sequences (SEQ ID NO:97). ,5 FIG.29A-C describe amino acid sequences and a nucleic acid sequence for a phage display S7-F(ab)' 2 . More specifically, FIG.29 shows an amino acid sequence comprising an S7-F(ab)' 2 light chain (SEQ ID NO:98), an amino acid sequence comprising an S7-F(ab)' 2 heavy chain region (SEQ ID NO:99) and a nucleic acid sequence encoding the amino acid 0 sequences (SEQ ID NO: 100). FIG.30A-C describe amino acid sequences and a nucleic acid sequence for a phage display S16-F(ab)' 2 . More specifically, FIG.30 shows an amino acid sequence comprising an S 16-F(ab)' 2 light chain (SEQ ID NO: 101), an amino acid sequence comprising an S16 17 WO 2004/094476 PCT/US2004/011793 F(ab)' 2 heavy chain region (SEQ ID NO:102) and a nucleic acid sequence encoding the amino acid sequences (SEQ ID NO:103). FIG.31 A-C describe amino acid sequences and a nucleic acid sequence for a phage 5 display F5-F(ab)' 2 . FIG.31 shows an amino acid sequence comprising a F5-F(ab)' 2 light chain (SEQ ID NO: 104), an amino acid sequence comprising an F5-F(ab)' 2 heavy chain region (SEQ ID NO: 105) and a nucleic acid sequence encoding the amino acid sequences(SEQ ID NO: 106). 10 FIG.32A-G describe amino acid sequences and a nucleic acid sequence for a S4-Fab. More specifically, FIG.32 shows an amino acid sequence comprising an S4-Fab light chain (SEQ ID NO:107), an amino acid sequence comprising an S4-Fab heavy chain region (SEQ ID NO: 108) and the nucleic acid sequence of a vector encoding the amino acid sequence (SEQ ID NO:109). 15 FIG.33A-F describe an S4 light chain sequence of an IgG protein. More specifically, FIG.33 shows an amino acid sequence comprising an S4 Light Chain (SEQ ID NO:110) and the nucleic acid sequence of a vector encoding the amino acid sequence (SEQ ID NO: 111). 20 FIG.34A-G describe an S4 heavy chain sequence of an IgG protein. More specifically, FIG.34 shows an amino acid sequence comprising an S4 Heavy Chain (SEQ ID NO:112) and the nucleic acid sequence of a vector encoding the amino acid sequence (SEQ ID NO:113). 25 FIG.35 shows a frequency of amino acids in human antibody light chain sequences from the Kabat database. FIG.36 shows one illustrative embodiment of a suitable codon set design. 30 FIG.37 is an illustrative embodiment of restricted diversity degenerate (also referred to herein as "nonrandom") codon sets for diversification of CDRs L1, L2 & L3. FIG.38 is an illustrative embodiment of restricted diversity degenerate (also referred to herein as "nonrandom") codon sets for diversification of CDRs L1, L2 & L3. 18 WO 2004/094476 PCT/US2004/011793 FIG.39 is an illustrative embodiment of restricted diversity degenerate (also referred to herein as "nonrandom") codon sets for diversification of CDR L3. 5 FIG.40 is an illustrative embodiment of a restricted diversity degenerate (also referred to herein as "nonrandom") codon sets for diversification of CDRs L1, L2 & L3. FIG.41 shows a flow cytometric analysis of populations of 293, HeLa, HT1080 or HUVEC cells treated with either (1) anti-HIS antibodies, (2) anti-HIS antibodies and STOP-1 protein or (3) anti-flag antibodies and STOP-1 protein, followed by treatment with 10 fluorescein isothiocyanate (FITC)-conjugated goat anti-mouse antibodies. A small, insignificant number of cells bound the anti-flag antibodies (i.e., the peaks at far left corner of the x-axis in the 293, HeLa and HT1080 graphs). The x-axis indicates the number of cells (log fluorescein signal intensity). The y-axis indicates the level of fluorescence emitted by the labeled cells (events). 15 FIG.42 shows a FACS analysis of populations of HT1080 cells treated with (1) anti HIS antibodies, (2) anti-HIS antibodies and STOP-1 protein, (3) anti-flag antibodies and STOP-1 protein, (3) STOP-1 protein, (4) STOP-1 protein and S7 antibodies or (5) STOP-1 and 6b12 antibodies, followed by treatment with FITC-conjugated goat anti-mouse 20 antibodies. FIG.43 charts the migration of HT1080 cells (number of cells) in a modified Boyden chemotactic chamber after treatment with bFGF or STOP-1 ("762") or a negative control. 25 FIG.44 shows a FACS analysis of STOP-1 binding to MDA435 cells in the presence and absence of an anti-STOP-1 antibody (6B 12) or an antibody control (4B7). The detection antibody, anti-flag M2 -F1TC antibody, did not effect STOP-1 binding. FIG.45 is a graph that shows the fold change in STOP-1 mRNA expression after 30 treatment (A) under hypoxic conditions for 8 and 34 hours or (B) under normnoxic conditions for 3, 8 and 34 hours, in the presence and absence of recombinant human TNFalpha. DETAILED DESCRIPTION OF THE INVENTION 19 WO 2004/094476 PCT/US2004/011793 A nucleic acid sequence coding for a STOP-1 protein according to this invention includes, e.g., SEQ ID NO: 1 and the nucleic acid molecules encoding the polypeptides of FIG.1. SEQ ID NO:1 5 GGAGAGAGGCGCGCGGGTGAAAGGCGCATTGATGCAGCCTGCGGCGGCCTCGGAGCGCGG CGGAGCCAGACGCTGACCACGTTCCTCTCCTCGGTCTCCTCCGCCTCCAGCTCCGCGCTG CCCGGCAGCCGGGAGCCATGCGACCCCAGGGCCCCGCCGCCTCCCCGCAGCGGCTCCGCG GCCTCCTGCTGCTCCTGCTGCTGCAGCTGCCCGCGCCGTCGAGCGCCTCTGAGATCCCCA 10 AGGGGAAGCAAAAGGCGCAGCTCCGGCAGAGGGAGGTGGTGGACCTGTATAATGGAATGT GCTTACAAGGGCCAGCAGGAGTGCCTGGTCGAGACGGGAGCCCTGGGGCCAATGTTATTC CGGGTACACCTGGGATCCCAGGTCGGGATGGATTCAAAGGAGAAAAGGGGGAATGTCTGA GGGAAAGCTTTGAGGAGTCCTGGACACCCAACTACAAGCAGTGTTCATGGAGTTCATTGA ATTATGGCATAGATCTTGGGAAAATTGCGGAGTGTACATTTACAAAGATGCGTTCAAATA 15 GTGCTCTAAGAGTTTTGTTCAGTGGCTCACTTCGGCTAAAATGCAGAAATGCATGCTGTC AGCGTTGGTATTTCACATTCAATGGAGCTGAATGTTCAGGACCTCTTCCCATTGAAGCTA TAATTTATTTGGACCAAGGAAGCCCTGAAATGAATTCAACAATTAATATTCATCGCACTT CTTCTGTGGAAGGACTTTGTGAAGGAATTGGTGCTGGATTAGTGGATGTTGCTATCTGGG TTGGCACTTGTTCAGATTACCCAAAAGGAGATGCTTCTACTGGATGGAATTCAGTTTCTC 20 GCATCATTATTGAAGAACTACCAAAATAAATGCTTTAATTTTCATTTGCTACCTCTTTTT TTATTATGCCTTGGAATGGTTCACTTAAATGACATTTTAAATAAGTTTATGTATACATCT GAATGAAAAGCAAAGCTAAATATGTTTACAGACCAAAGTGTGATTTCACACTGTTTTTAA ATCTAGCATTATTCATTTTGCTTCAATCAAAAGTGGTTTCAATATTTTTTTTAGTTGGTT AGAATACTTTCTTCATAGTCACATTCTCTCAACCTATAATTTGGAATATTGTTGTGGTCT 25 TTTGTTTTTTCTCTTAGTATAGCATTTTTAAAAAAATATAAAAGCTACCAATCTTTGTAC AATTTGTAAATGTTAAGAATTTTTTTTATATCTGTTAAATAAAAATTATTTCCAACA The terms "STOP-1," "STOP-1 protein," "STOP-1 polypeptide" (also refered to UNQ762 or 762) as used herein include native sequence polypeptides, polypeptide variants 30 and fragments of native sequence polypeptides and polypeptide variants (which are further defined herein), unless specified otherwise. STOP-1 proteins can be obtained from various species, e.g., humans, by using antibodies according to this invention or by recombinant or synthetic methods, including using deposited nucleic acid molecules. An oligomeric form of STOP-1 includes a human STOP-1 having only residues 94-243, or a part thereof. An 35 oligomeric form according to this invention can include a dimer, a trimer and a hexamer of STOP-1. According to one preferred embodiment, the oligomeric form of STOP-1 is a trimer. A "native sequence" polypeptide or "native" polypeptide is one which has the same amino acid sequence as a polypeptide (e.g., antibody) derived from nature. A "native sequence" polypeptide is one which has the same amino acid sequence as a polypeptide (e.g., 40 antibody) derived from nature. Such native sequence polypeptides can be isolated from nature or can be produced by recombinant or synthetic means. Thus, a native sequence polypeptide can have the amino acid sequence of a naturally occurring human polypeptide, murine polypeptide, or polypeptide from any other mammalian species. A "native sequence" STOP-1 polypeptide or a "native" STOP-1 polypeptide comprises a polypeptide having the 20 WO 2004/094476 PCT/US2004/011793 same amino acid sequence as the corresponding STOP-1 polypeptide derived from nature. For example, in one preferred embodiment, the nucleic acid sequence encoding a native sequence of human STOP-1 can be found in SEQ ID NO:2 and FIG.2. 5 SEQ ID NO:2 MRPQGPAASPQRLRGLLLLLLLQLPAPSSASEIPKGKQKAQLRQREVVDLYNGMCLQ GPAGVPGRDGSPGANVIPGTPGIPGRDGFKGEKGECLRESFEESWTPNYKQCSWSSL NYGIDLGKIAECTFTKMRSNSALRVLFSGSLRLKCRNACCQRWYFTFNGAECSGPLPI EAIIYLDQGSPEMNSTINIHRTSSVEGLCEGIGAGLVDVAlWVGTCSDYPKGDASTGW 10 NSVSRIIIEELPK Such STOP-1 polypeptides can be isolated from nature or can be produced by recombinant or synthetic means. The term "native sequence" or "native" STOP-1 polypeptide or protein specifically encompasses naturally-occurring truncated or secreted 15 forms of the STOP-1 protein, naturally-occurring variant forms (e.g., alternatively spliced forms) and naturally-occurring allelic variants of the polypeptide. In certain embodiments of the invention, the native sequence STOP-1 polypeptides disclosed herein are mature or full length native sequence polypeptides comprising the full-length amino acids sequences shown in the accompanying figures. 10 The approximate location of the "signal peptides" of the various STOP-1 polypeptides disclosed herein can be seen in the present specification and/or the accompanying figures. It is also recognized that, in some cases, cleavage of a signal sequence from a secreted polypeptide is not entirely uniform, resulting in more than one secreted species. These mature polypeptides, where the signal peptide is cleaved within no more than about 5 amino 15 acids on either side of the C-terminal boundary of the signal peptide as identified herein, and the polynucleotides encoding them, are contemplated by the present invention. "STOP-1 polypeptide variant" means a STOP-1 polypeptide having at least about 80% amino acid sequence identity with a full-length native sequence STOP-1 polypeptide sequence as disclosed herein, a STOP-1 polypeptide sequence lacking the signal peptide or ;0 triple helix domain as disclosed herein, or any other fragment of a full-length STOP-1 polypeptide sequence as disclosed herein (such as those encoded by a nucleic acid that represents only a portion of the complete coding sequence for a full-length STOP-1 polypeptide). Such STOP-1 polypeptide variants include, for instance, STOP-1 polypeptides wherein one or more amino acid residues are added, or deleted, at the N- or C-terminus of 5 the full-length native amino acid sequence. Ordinarily, a STOP-1 polypeptide variant will have at least about 80% amino acid sequence identity, alternatively at least about 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 21 WO 2004/094476 PCT/US2004/011793 or 99% amino acid sequence identity, to a full-length native sequence STOP-1 polypeptide sequence as disclosed herein, a STOP-1 polypeptide sequence lacking the signal peptide as disclosed herein, a triple helix domain of a STOP-1 polypeptide, with or without the signal peptide, as disclosed herein or any other specifically defined fragment of a full-length STOP 5 1 polypeptide sequence as disclosed herein. Ordinarily, STOP-1 variant polypeptides are at least about 10 amino acids in length, alternatively at least about 20, 30, 40, 50, 60, 70, 80, 90, 100, 110, 120, 130, 140, 150, 160, 170, 180, 190, 200, 210, 220, 230, 240, 250, 260, 270, 280, 290, 300, 310, 320, 330, 340, 350, 360, 370, 380, 390, 400, 410, 420, 430, 440, 450, 460, 470, 480, 490, 500, 510, 520, 530, 540, 550, 560, 570, 580, 590, 600 amino acids in 10 length, or more. Optionally, STOP-1 variant polypeptides will have no more than one conservative amino acid substitution as compared to the native STOP-1 polypeptide sequence, alternatively no more than 2, 3, 4, 5, 6, 7, 8, 9, or 10 conservative amino acid substitution as compared to the native STOP-1 polypeptide sequence. "Percent (%) amino acid sequence identity" with respect to the STOP-1 polypeptide 15 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 the specific STOP-1 polypeptide 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 20 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 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, 25 however, % amino acid sequence identity values are generated using the sequence comparison computer program ALIGN-2, wherein the complete source code for the ALIGN 2 program is provided in Table 1 below. The ALIGN-2 sequence comparison computer program was authored by Genentech, Inc. and the source code shown in Table 1 below has been filed with user documentation in the U.S. Copyright Office, Washington D.C., 20559, 30 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 can be compiled from the source code provided in Table 1 below. 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. 22 WO 2004/094476 PCT/US2004/011793 In situations where ALIGN-2 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 5 amino acid sequence B) is calculated as follows: 100 times the fraction X/Y where X is the number of amino acid residues scored as identical matches by the sequence 10 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 % 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 using this method, Tables 2 and 3 15 demonstrate how to calculate the % amino acid sequence identity of the amino acid sequence designated "Comparison Protein" to the amino acid sequence designated "STOP-I", wherein "STOP-I" represents the amino acid sequence of a hypothetical STOP-1 polypeptide of interest, "Comparison Protein" represents the amino acid sequence of a polypeptide against which the "STOP-I1" polypeptide of interest is being compared, and "X, "Y" and "Z" each 20 represent different hypothetical amino acid residues. Unless specifically stated otherwise, all % amino acid sequence identity values used herein are obtained as described in the immediately preceding paragraph using the ALIGN-2 computer program. "STOP-1 variant polynucleotide" or "STOP-1 variant nucleic acid sequence" means a nucleic acid molecule which encodes a STOP-1 polypeptide, preferably an active STOP-1 25 polypeptide, as defined herein and which has at least about 80% nucleic acid sequence identity with a nucleotide acid sequence encoding a full-length native sequence STOP- 1 polypeptide sequence as disclosed herein, a full-length native sequence STOP-1 polypeptide sequence lacking the signal peptide as disclosed herein, the triple helix domain of a STOP-1 polypeptide, with or without the signal peptide, as disclosed herein or any other fragment of a 30 full-length STOP-1 polypeptide sequence as disclosed herein (e.g., residues 94-243 of human STOP-1). Ordinarily, a STOP-1 variant polynucleotide will have at least about 80% nucleic acid sequence identity, alternatively at least about 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% nucleic acid sequence identity with a nucleic acid sequence encoding a full-length native sequence STOP-1 23 WO 2004/094476 PCT/US2004/011793 polypeptide sequence as disclosed herein, a full-length native sequence STOP-1 polypeptide sequence lacking the signal peptide as disclosed herein, the triple helix domain of a STOP-1 polypeptide, with or without the signal sequence, as disclosed herein or any other fragment of a full-length STOP-1 polypeptide sequence as disclosed herein. Variants do not encompass 5 the native nucleotide sequence. Ordinarily, STOP-1 variant polynucleotides are at least about 5 nucleotides in length, alternatively at least about 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 105, 110, 115, 120, 125, 130, 135, 140, 145, 150, 155, 160, 165, 170, 175, 180, 185, 190, 195, 200, 210, 10 220, 230, 240, 250, 260, 270, 280, 290, 300, 310, 320, 330, 340, 350, 360, 370, 380, 390, 400, 410, 420, 430, 440, 450, 460, 470, 480, 490, 500, 510, 520, 530, 540, 550, 560, 570, 580, 590, 600, 610, 620, 630, 640, 650, 660, 670, 680, 690, 700, 710, 720, 730, 740, 750, 760, 770, 780, 790, 800, 810, 820, 830, 840, 850, 860, 870, 880, 890, 900, 910, 920, 930, 940, 950, 960, 970, 980, 990, or 1000 nucleotides in length, wherein in this context the term 15 "about" means the referenced nucleotide sequence length plus or minus 10% of that referenced length. "Percent (%) nucleic acid sequence identity" with respect to STOP-I-encoding nucleic acid sequences identified herein is defined as the percentage of nucleotides in a candidate sequence that are identical with the nucleotides in the STOP-1 nucleic acid 2_0 sequence of interest, after aligning the sequences and introducing 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 or Megalign (DNASTAR) software. For purposes herein, however, % 15 nucleic acid sequence identity values are generated using the sequence comparison computer program ALIGN-2, wherein the complete source code for the ALIGN-2 program is provided in Table 1 below. The ALIGN-2 sequence comparison computer program was authored by Genentech, Inc. and the source code shown in Table 1 below has been filed with user documentation in the U.S. Copyright Office, Washington D.C., 20559, where it is registered ;0 under U.S. Copyright Registration No. TXU510087. The ALIGN-2 program is publicly available through Genentech, Inc., South San Francisco, California or can be compiled from the source code provided in Table 1 below. 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. 24 WO 2004/094476 PCT/US2004/011793 In situations where ALIGN-2 is employed for nucleic acid sequence comparisons, 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 comprises a certain % nucleic acid sequence identity to, with, or 5 against a given nucleic acid sequence D) 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 10 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 D, 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, Tables 4 and 5, demonstrate how to calculate the % 15 nucleic acid sequence identity of the nucleic acid sequence designated "Comparison DNA" to the nucleic acid sequence designated "STOP-1-DNA", wherein "STOP-1-DNA" represents a hypothetical STOP-l-encoding nucleic acid sequence of interest, "Comparison DNA" represents the nucleotide sequence of a nucleic acid molecule against which the "STOP-1 DNA" nucleic acid molecule of interest is being compared, and "N", "L" and "V" each 20 represent different hypothetical nucleotides. Unless specifically stated otherwise, all % nucleic acid sequence identity values used herein are obtained as described in the immediately preceding paragraph using the ALIGN-2 computer program. In other embodiments, STOP-1 variant polynucleotides are nucleic acid molecules that encode a STOP-1 polypeptide and which are capable of hybridizing, preferably under 25 stringent hybridization and wash conditions, to nucleotide sequences encoding a full-length STOP-1 polypeptide as disclosed herein. STOP-1 variant polypeptides can be those that are encoded by a STOP-1 variant polynucleotide. The term "full-length coding region" when used in reference to a nucleic acid encoding a STOP-1 polypeptide refers to the sequence of nucleotides which encode the full 30 length STOP-1 polypeptide of the invention (which is often shown between start and stop codons, inclusive thereof, in the accompanying figures). The term "full-length coding region" when used in reference to an ATCC deposited nucleic acid refers to the STOP-1 polypeptide-encoding portion of the cDNA that is inserted into the vector deposited with the 25 WO 2004/094476 PCT/US2004/011793 ATCC whichh is often shown between start and stop codons, inclusive thereof, in the accompanying figures). "Isolated," when used to describe the various STOP-1 polypeptides disclosed herein, means polypeptide that has been identified and separated and/or recovered from a component 5 of its natural environment. Contaminant components of its natural environment are materials that would typically interfere with diagnostic or therapeutic uses for the polypeptide, and can 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 10 sequenator, or (2) to homogeneity by SDS-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 STOP-1 polypeptide natural environment will not be present. Ordinarily, however, isolated polypeptide will be prepared by at least one purification step. 15 An "isolated" STOP-1 polypeptide-encoding nucleic acid or other polypeptide encoding nucleic acid 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 polypeptide-encoding nucleic acid. An isolated polypeptide-encoding nucleic acid molecule is other than in the form or setting in which it is found in nature. Isolated 0 polypeptide-encoding nucleic acid molecules therefore are distinguished from the specific polypeptide-encoding nucleic acid molecule as it exists in natural cells. However, an isolated polypeptide-encoding nucleic acid molecule includes polypeptide-encoding nucleic acid molecules contained in cells that ordinarily express the polypeptide where, for example, the nucleic acid molecule is in a chromosomal location different from that of natural cells. 15 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. 0 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 26 WO 2004/094476 PCT/US2004/011793 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 5 sites do not exist, the synthetic oligonucleotide adaptors or linkers are used in accordance with conventional practice. "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 10 temperatures for proper annealing, while shorter probes need lower temperatures. Hybridization 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 15 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, can be -0 identified by those that: (1) employ low ionic strength and high temperature for washing, for example 0.015 M sodium chloride/0.0015 M sodium citrate/0.1% sodium dodecyl sulfate at 50C; (2) employ during hybridization a denaturing agent, such as formamide, for example, 50% (v/v) formamide with 0.1% bovine serum albumin/0.1% Ficoll/0.1% polyvinylpyrrolidone/50mM sodium phosphate buffer at pH 6.5 with 750 mM sodium 15 chloride, 75 mM sodium citrate at 42C; or (3) overnight hybridization in a solution that employs 50% formamide, 5 x SSC (0.75 M NaC1, 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 gg/ml), 0.1% SDS, and 10% dextran sulfate at 42C, with a 10 minute wash at 42C in 0.2 x SSC (sodium chloride/sodium citrate) followed by a 10 minute high .0 stringency wash consisting of 0.1 x SSC containing EDTA at 55C. "Moderately stringent conditions" can 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 27 WO 2004/094476 PCT/US2004/011793 stringent conditions is overnight incubation at 37 0 C in a solution comprising: 20% formamide, 5 x SSC (150 mM NaC1, 15 mM trisodium citrate), 50 mM sodium phosphate (pH 7.6), 5 x Denhardt's solution, 10% dextran sulfate, and 20 mg/ml denatured sheared salmon sperm DNA, followed by washing the filters in 1 x SSC at about 37-50C. The skilled 5 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 STOP-1 polypeptide or anti-STOP-1 antibody fused to a "tag polypeptide". The tag polypeptide has enough residues to provide an epitope against which an antibody can be 10 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). Polypeptides and antibodies of 15 this invention that are epitope-tagged are contemplated. "Biologically active" and "biological activity" and "biological characteristics" with respect to an STOP-1 means (1) having the ability to increase cell proliferation of at least one type of mammalian cell (e.g., 3T3) in vivo or ex vivo; (2) having the ability to specifically bind STOP-1; and/or (3) having the ability to otherwise modulate STOP-1 signaling or 20 STOP-1 activity, except where specified otherwise. "Biologically active" and "biological activity" and "biological characteristics" with respect to a modified STOP-1 polypeptide or a STOP-1 polypeptide (1) having the ability to partially or fully block, inhibit or neutralize a biological activity of a native STOP-1 (either in an antagonistic or blocking manner); (2) having the ability to specifically bind STOP-1; 25 and/or (3) having the ability to modulate STOP-1 signaling or STOP-1 activity, except where specified otherwise. "Biologically active" and "biological activity" and "biological characteristics" with respect to an anti-STOP-1 antibody of this invention means (1) having the ability to partially or fully block, inhibit or neutralize a biological activity of a native STOP-1 (either in an 30 antagonistic or blocking manner); (2) having the ability to specifically bind STOP-1; and/or (3) having the ability to modulate STOP-1 signaling or STOP-1 activity, except where specified otherwise. In one preferred embodiment, an antibody of this invention binds to STOP-1 with an affinity of at least luM or less, 100nm or less, 50nm or less, 10nm or less, 5nM or less, Inm or less. As used herein, "antibody variable domain" refers to the portions 28 WO 2004/094476 PCT/US2004/011793 of the light and heavy chains of antibody molecules that include amino acid sequences of Complementary Determining Regions (CDRs; ie., CDR1, CDR2, and CDR3), and Framework Regions (FRs). VH refers to the variable domain of the heavy chain. VL refers to the variable domain of the light chain. According to the methods used in this invention, the 5 amnino acid positions assigned to CDRs and FRs are defined according to Kabat (Sequences of Proteins of Immunological Interest (National Institutes of Health, Bethesda, Md., 1987 and 1991)). Amino acid numbering of antibodies or antigen binding fragments is also according to that of Kabat. As used herein, "codon set" refers to a set of different nucleotide triplet sequences 10 used to encode desired variant amino acids. A set of oligonucleotides can be synthesized, for example, by solid phase synthesis, containing sequences that represent all possible combinations of nucleotide triplets provided by the codon set and that will encode the desired group of amino acids. A standard form of codon designation is that of the IUB code, which is known in the art and described herein. A "non-random codon set", as used herein, thus refers 15 to a codon set that encodes select amino acids that fulfill partially, preferably completely, the criteria for amino acid selection as described herein. Synthesis of oligonucleotides with selected nucleotide "degeneracy" at certain positions is known in that art, for example the TRIM approach (Knappek et al.; J. Mol. Biol. (1999), 296:57-86); Garrard & Henner, Gene (1993), 128:103). Such sets of nucleotides having certain codon sets can be synthesized 20 using commercial nucleic acid synthesizers (available from, for example, Applied Biosystems, Foster City, CA), or can be obtained commercially (for example, from Life Technologies, Rockville, MD). Therefore, a set of oligonucleotides synthesized having a particular codon set will typically include a plurality of oligonucleotides with different sequences, the differences established by the codon set within the overall sequence. 25 Oligonucleotides, as used according to the invention, have sequences that allow for hybridization to a variable domain nucleic acid template and also can, but does not necessarily, include restriction enzyme sites useful for, for example, cloning purposes. "Heterologous DNA" is any DNA that is introduced into a host cell. The DNA can be derived from a variety of sources including genomic DNA, cDNA, synthetic DNA and 30 fusions or combinations of these. The DNA can include DNA from the same cell or cell type as the host or recipient cell or DNA from a different cell type, for example, from a mammal or plant. The DNA can, optionally, include marker or selection genes, for example, antibiotic resistance genes, temperature resistance genes, etc. Host cells encoding heterologous DNAs 29 WO 2004/094476 PCT/US2004/011793 comprising the UNQ polypeptides and antibodies of this invention are contemplated as well as their use. As used herein, "highly diverse position" refers to a position of an amino acid located in the variable regions of the light and heavy chains that have a number of different amino 5 acid represented at the position when the amino acid sequences of known andlor naturally occurring antibodies or antigen binding fragments are compared. The highly diverse positions are typically in the CDR regions. In one aspect, the ability to determine highly diverse positions in known and/or naturally occurring antibodies is facilitated by the data provided by Kabat, Sequences of Proteins of Immunological Interest (National Institutes of 10 Health, Bethesda, Md., 1987 and 1991). An internet-based database located at http://immuno.bme.nwu.edu provides an extensive collection and alignment of human light and heavy chain sequences and facilitates determination of highly diverse positions in these sequences. According to the invention, an amino acid position is highly diverse if it has preferably from about 2 to about 11, preferably from about 4 to about 9, and preferably from 15 about 5 to about 7 different possible amino acid residue variations at that position. In some embodiments, an amino acid position is highly diverse if it has preferably at least about 2, preferably at least about 4, preferably at least about 6, and preferably at least about 8 different possible amino acid residue variations at that position. As used herein, "library" refers to a plurality of antibody or antibody fragment 20 sequences (for example, polypeptides of the invention), or the nucleic acids that encode these sequences, the sequences being different in the combination of variant amino acids that are introduced into these sequences according to the methods of the invention. "Phage display" is a technique by which variant polypeptides are displayed as fusion proteins to a coat protein on the surface of phage, e.g., filamentous phage, particles. A utility 25 of phage display lies in the fact that large libraries of randomized protein variants can be rapidly and efficiently sorted for those sequences that bind to a target molecule with high affinity. Display of peptide and protein libraries on phage has been used for screening millions of polypeptides for ones with specific binding properties. Polyvalent phage display methods have been used for displaying small random peptides and small proteins through 30 fusions to either gene III or gene VIII of filamentous phage. Wells and Lowman, Curr. Opin. Struct. Biol., 3:355-362 (1992), and references cited therein. In monovalent phage display, a protein or peptide library is fused to a gene III or a portion thereof, and expressed at low levels in the presence of wild type gene I protein so that phage particles display one copy or none of the fusion proteins. Avidity effects are reduced relative to polyvalent phage so that 30 WO 2004/094476 PCT/US2004/011793 sorting is on the basis of intrinsic ligand affinity, and phagemid vectors are used, which simplify DNA manipulations. Lowman and Wells, Methods: A companion to Methods in Enzymology, 3:205-0216 (1991). A "phagemid" is a plasmid vector having a bacterial origin of replication, e.g., ColE1, 5 and a copy of an intergenic region of a bacteriophage. The phagemid can be used on any known bacteriophage, including filamentous bacteriophage and lambdoid bacteriophage. The plasmid will also generally contain a selectable marker for antibiotic resistance. Segments of DNA cloned into these vectors can be propagated as plasmids. When cells harboring these vectors are provided with all genes necessary for the production of phage particles, the mode 10 of replication of the plasmid changes to rolling circle replication to generate copies of one strand of the plasmid DNA and package phage particles. The phagemid can form infectious or non-infectious phage particles. This term includes phagemids which contain a phage coat protein gene or fragment thereof linked to a heterologous polypeptide gene as a gene fusion such that the heterologous polypeptide is displayed on the surface of the phage particle. 15 The term "phage vector" means a double stranded replicative form of a bacteriophage containing a heterologous gene and capable of replication. The phage vector has a phage origin of replication allowing phage replication and phage particle formation. The phage is preferably a filamentous bacteriophage, such as an M13, fl, fd, Pf3 phage or a derivative thereof, or a lambdoid phage, such as lambda, 21, phi80, phi81, 82, 424, 434, etc., or a 20 derivative thereof. As used herein, "target amino acid" refers to an amino acid that belongs to the group of amino acids that are collectively the most commonly occurring amino acids found at a particular position of known and/or natural antibodies or antigen binding fragments. In some embodiments, the most commonly occurring amino acids" are those amino acids that are 25 found in a particular position in preferably at least about 50%, preferably at least about 70%, preferably at least about 80%, preferably at least about 90%, preferably all of sequences of known and/or natural antibodies or antigen binding fragments. In some embodiments, the most commonly occurring amino acids" are those amino acids that are found in a particular position in preferably from about 50% to about 100%, preferably from about 60% to about 30 90%, preferably from about 70% to about 85%, preferably from about 80% to about 85% of the sequences of known and/or natural antibodies or antigen binding fragments. Known antibodies or antigen biding fragments are those whose sequences are available in the art, such as those available in publicly-accessible databases, such as the database of Kabat ("Sequence of Proteins of Immunological Interest, National Institutes of Health, Bethesda, 31 WO 2004/094476 PCT/US2004/011793 MId., 198/ and 1991) and/or as located at http://immuno.bme.nwu.edu. The amino acid position is preferably a position in the CDR region. A target group of amino acids refers to a group of target amino acids for a particular position. Preferably, a target amino acid is not a cysteine residue. For positions in the light chain CDR1, CDR2, CDR3, and for heavy chain 5 CDR1 and CDR2, typically, a target group of amino acids can include from preferably about two to about eleven, preferably from about 4 to about 9, preferably from about 5 to about 7, preferably about 6 amino acids at a particular highly diverse and solvent-accessible position of the source sequence. The term "proteoglycan" refers to a molecule where at least one glycosaminoglycan 10 side chain is covalently attached to the protein core of the molecule. A proteoglycan synthesis deficient cell line according to this invention includes a cell line that is deficient in galactosyltransferase I. According to one preferred embodiment, the cell line is a CHO-psbg cell line. The term "antagonist" is any molecule that partially or fully blocks, inhibits, or [5 neutralizes a biological activity of a native STOP-1 polypeptide and that specifically binds to a native STOP-1 polypeptide, wherein the binding of the antagonist (1) is to a native STOP-1 polypeptide in oligomeric form, (2) is to residues 94-243 of native human STOP-1 and/or (3) can be inhibited (e.g., as observed in a competitive ELISA assay using STOP-1 and 6B 12) by a monoclonal antibody of this invention (e.g., a deposited antibody of this invention, etc.). ;O According to one embodiment, the antagonist is a polypeptide. According to another embodiment, the 6B 12 antibody can inhibit the binding of the antagonist to STOP-1. According to another embodiment, the antagonist binds to a residue within residues 33-52 or 33-53 of human STOP-1 or a non-human STOP-1 equivalent thereof. The term "small molecule antagonist" refers to any molecule wherein the molecular 5 weight is 1500 daltons or less and is an antagonist according to this invention. According to one embodiment the small molecule antagonist is below about 500 Daltons. According to one preferred embodiment, the antagonist blocks, inhibits or neutralizes cell proliferation in cells expressing native STOP-1. In one embodiment, the antagonist or small molecule antagonist prevents migration of a cell to which STOP-1 binds. In one ) preferred embodiment, the antagonist or small molecule antagonist specifically binds to a trimeric form of STOP-1. Suitable antagonists include antibodies, amino acid sequence variants of native STOP-1 polypeptides, peptides, of this invention etc. Methods for identifying antagonists of a STOP-1 polypeptide can comprise contacting a STOP-1 32 WO 2004/094476 PCT/US2004/011793 polypeptide with a candidate antagonist molecule and measuring a detectable change in one or more biological activities associated with the STOP-1 polypeptide. The term "potentiator" refers to any molecule that enhances a biological activity of a native STOP-1 polypeptide and that specifically binds to a native STOP-1 polypeptide, 5 wherein the potentiator can bind to an oligomeric form of a native STOP-1 polypeptide and has at least one additional activity selected from the group consisting of (1) is capable of binding to a residue in residues 94-243 of native human STOP-1, (2) is capable of aggregating STOP-1 on a cell; and (3) can be competed by a monoclonal antibody an S7 or S4 antibody of this invention (e.g., as observed in a competitive ELISA assay using STOP-1, 10 S7 or S4 and the potentiator). According to one embodiment, the potentiator increases the binding of STOP-1 to a cell (e.g., HUVEC cells, HeLa cells, and HT1080 cells). In one preferred embodiment, the agonist binds to a trimeric form of STOP-1. Methods for identifying agonists of a STOP-1 polypeptide can comprise contacting a cell that binds STOP-1 with a STOP-1 polypeptide and the candidate agonist and measuring a detectable 15 change in one or more biological activities associated with the STOP-1 polypeptide (e.g., increased binding of the STOP-1 polypeptide, cell proliferation or cell migration). "Treating" or "treatment" or "alleviation" 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 20 already with the disorder as well as those prone to have the disorder or those in whom the disorder is to be prevented. These terms indicate the therapeutic and prophylactic uses herein are successful if they ameliorate, lessen or decrease the symptoms, complications or other problems associated with a disease or ameliorate, lessen or decrease the chance of onset or frequency of the symptoms, complications or other problems associated with a disease. 25 A subject or mammal is successfully "treated" for a STOP-1 polypeptide-expressing cancer if, after receiving a therapeutic amount of an antagonist according to the methods of the present invention, the patient shows observable and/or measurable reduction in or absence of one or more of the following: reduction in the number of cancer cells or absence of the cancer cells; reduction in the tumor size; inhibition (i.e., slow to some extent and preferably 30 stop) of cancer cell infiltration into peripheral organs including the spread of cancer into soft tissue and bone; inhibition (i.e., slow to some extent and preferably stop) of tumor metastasis; inhibition, to some extent, of tumor growth; and/or relief to some extent, one or more of the symptoms associated with the specific cancer; reduced morbidity and mortality, and improvement in quality of life issues. To the extent the anti-STOP-1 antibody or STOP-1 33 WO 2004/094476 PCT/US2004/011793 binding oligopeptide can prevent growth and/or kill existing cancer cells, it can be cytostatic and/or cytotoxic. Reduction of these signs or symptoms can also be felt by the patient. A subject or mammal is successfully "treated" for abnormal angiogenesis if, after receiving a therapeutic amount of an antagonist or agonist according to the methods of the 5 present invention, the patient shows observable and/or measurable [TO BE FILLED IN]; and/or relief to some extent, one or more of the symptoms associated with the abnormal angiogenesis; and improvement in quality of life issues. The above parameters for assessing successful treatment and improvement in the disease are readily measurable by procedures familiar to a physician. For cancer therapy, 10 efficacy can be measured, for example, by assessing the time to disease progression (TTP) and/or determining the response rate (RR). Metastasis can be determined by staging tests and by bone scan and tests for calcium level and other enzymes to determine spread to the bone. CT scans can also be done to look for spread to the pelvis and lymph nodes in the area. Chest X-rays and measurement of liver enzyme levels by known methods are used to look for 15 metastasis to the lungs and liver, respectively. Other known methods for monitoring the disease include transrectal ultrasonography (TRUS) and transrectal needle biopsy (TRNB). For bladder cancer, which is a more localized cancer, methods to determine progress of disease include urinary cytologic evaluation by cystoscopy, monitoring for presence of blood in the urine, visualization of the urothelial tract by sonography or an intravenous 20 pyelogram, computed tomography (CT) and magnetic resonance imaging (MRI). The presence of distant metastases can be assessed by CT of the abdomen, chest x-rays, or radionuclide imaging of the skeleton. "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 25 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 the treatment of, alleviating the symptoms of or diagnosis of a cancer refers to any animal classified as a mammal (aka "patient"), including humans, domestic and farm animals, and zoo, sports, or pet animals, such as dogs, cats, cattle, horses, 30 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 34 WO 2004/094476 PCT/US2004/011793 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, 5 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 TWEEN®, polyethylene glycol (PEG), 10 and PLURONICS@®. By "solid phase" or "solid support" is meant a non-aqueous matrix to which an antibody, an antagonist or a polypeptide of the present invention can adhere or attach. Examples of solid phases encompassed herein include those formed partially or entirely of glass (e.g., controlled pore glass), polysaccharides (e.g., agarose), polyacrylamides, 15 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. As used herein, the term "immunoadhesin" designates antibody-like molecules that 20 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 that 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 25 molecule typically is a contiguous amino acid sequence comprising at least the binding site of a receptor or a ligand - such as a portion of a native STOP-1 protein. The immunoglobulin constant domain sequence in the immunoadhesin can 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, IgD, or IgM. 30 A "liposome" is a small vesicle composed of various types of lipids, phospholipids and/or surfactant which is useful for delivery of a drug (such as a STOP-1 polypeptide, an antibody thereto or a STOP-1 binding oligopeptide) to a mammal. The components of the liposome are commonly arranged in a bilayer formation, similar to the lipid arrangement of biological membranes. 35 WO 2004/094476 PCT/US2004/011793 A "small" molecule or "small" organic molecule is defined herein to have a molecular weight below about 500 Daltons. An "effective amount" of a polypeptide, antibody, antagonist or composition as disclosed herein is an amount sufficient to carry out a specifically stated purpose. An 5 "effective amount" can be determined empirically and by known methods relating to the stated purpose. The term "therapeutically effective amount" refers to an amount of an antibody, polypeptide or antagonist of this invention effective to "treat" a disease or disorder in a mammal (aka patient). In the case of cancer, the therapeutically effective amount of the drug 10 can reduce the number of cancer cells; reduce the tumor size; inhibit (i.e., slow to some extent and preferably stop) cancer cell infiltration into peripheral organs; inhibit (i.e., slow to some extent and preferably stop) tumor metastasis; inhibit, to some extent, tumor growth; and/or relieve to some extent one or more of the symptoms associated with the cancer. See the definition herein of "treating". To the extent the drug can prevent growth and/or kill 15 existing cancer cells, it can be cytostatic and/or cytotoxic. A "growth inhibitory amount" of a polypeptide, antibody, antagonist or composition of this invention is an amount capable of inhibiting the growth of a cell, especially tumor, e.g., cancer cell, either in vitro or in vivo. A "growth inhibitory amount" of a polypeptide, antibody, antagonist or composition of this invention for purposes of inhibiting neoplastic 20 cell growth can be determined empirically and by known methods or by examples provided herein. A "cytotoxic amount" of a polypeptide, antibody, antagonist or composition of this invention 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 polypeptide, antibody, 25 antagonist or composition of this invention for purposes of inhibiting neoplastic cell growth can be determined empirically and by methods known in the art. The term "antibody" is used in the broadest sense and specifically covers, for example, single anti-STOP-1 monoclonal antibodies (including agonist, antagonist, and neutralizing antibodies), anti-STOP- 1 antibody compositions. with polyepitopic specificity, 30 polyclonal antibodies, single chain anti-STOP-1 antibodies, and fragments of anti-STOP-1 antibodies (see below) as long as they specifically bind a native STOP-1 polypeptide and/or exhibit a biological activity or immunological activity of this invention. According to one embodiment, the antibody binds to an oligomeric form of STOP-1, e.g., a trimeric form. In a further embodiment, the antibody specifically binds to human STOP-1 between residues 94 36 WO 2004/094476 PCT/US2004/011793 243. According to another embodiment, the antibody specifically binds to STOP-1, which binding can be inhibited by a monoclonal antibody of this invention (e.g., a deposited antibody of this invention, etc.). The phrase "functional fragment or analog" of an antibody is a compound having a qualitative biological activity in common with an antibody to which 5 it is being referred. For example, a functional fragment or analog of an anti-STOP- 1 antibody can be one which can specifically bind to a STOP-1 molecule. In one embodiment, the antibody can prevent or substantially reduce the ability of a STOP-1 molecule to induce cell proliferation. The term "immunoglobulin" (Ig) is used interchangeably with "antibody" herein.According to one embodiment, an antibody of this invention does not bind to a peptide 10 having the amino acid sequence GWNSVSRIIIEELPK. An "isolated antibody" is one which has been identified and separated and/or recovered from a component of its natural environment. Contaminant components of its natural environment are materials which would interfere with diagnostic or therapeutic uses for the antibody, and can include enzymes, hormones, and other proteinaceous or 15 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 homogeneity by SDS-PAGE under reducing or nonreducing conditions using Coomassie blue 20 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 basic 4-chain antibody unit is a heterotetrameric glycoprotein composed of two identical light (L) chains and two identical heavy (H) chains (an IgM antibody consists of 5 25 of the basic heterotetramer unit along with an additional polypeptide called J chain, and therefore contain 10 antigen binding sites, while secreted IgA antibodies can polymerize to form polyvalent assemblages comprising 2-5 of the basic 4-chain units along with J chain). In the case of IgGs, the 4-chain unit is generally about 150,000 daltons. Each L chain is linked to a H chain by one covalent disulfide bond, while the two H chains are linked to each 30 other by one or more disulfide borids depending on the H chain isotype. Each H and L chain also has regularly spaced intrachain disulfide bridges. Each H chain has at the N-terminus, a variable domain (VH) followed by three constant domains (CH) for each of the a and 7 chains and four CH domains for pt and s isotypes. Each L chain has at the N-terminus, a variable domain (VL) followed by a constant domain (CL) at its other end. The VL is aligned with the 37 WO 2004/094476 PCT/US2004/011793 VH and the CL is aligned with the first constant domain of the heavy chain (CH1). Particular amino acid residues are believed to form an interface between the light chain and heavy chain variable domains. The pairing of a VH and VL together forms a single antigen-binding site. For the structure and properties of the different classes of antibodies, see, e.g., Basic and 5 Clinical Immunology, 8th edition, Daniel P. Stites, Abba I. Terr and Tristram G. Parslow (eds.), Appleton & Lange, Norwalk, CT, 1994, page 71 and Chapter 6. The L chain from any vertebrate species can be assigned to one of two clearly distinct types, called kappa and lambda, based on the amino acid sequences of their constant domains. Depending on the amino acid sequence of the constant domain of their heavy 10 chains (CH), immunoglobulins can be assigned to different classes or isotypes. There are five classes of immunoglobulins: IgA, IgD, IgE, IgG, and IgM, having heavy chains designated a, 5, F, 7, and It, respectively. The y and a classes are further divided into subclasses on the basis of relatively minor differences in CH sequence and function, e.g., humans express the following subclasses: IgG1, IgG2, IgG3, IgG4, IgA1, and IgA2. 15 The term "variable" refers to the fact that certain segments of the variable domains differ extensively in sequence among antibodies. The V domain mediates antigen binding and define specificity of a particular antibody for its particular antigen. However, the variability is not evenly distributed across the 110-amino acid span of the variable domains. Instead, the V regions consist of relatively invariant stretches called framework regions (FRs) 20 of 15-30 amino acids separated by shorter regions of extreme variability called "hypervariable regions" that are each 9-12 amino acids long. The variable domains of native heavy and light chains each comprise four FRs, largely adopting a P-sheet configuration, connected by three hypervariable regions, which form loops connecting, and in some cases forming part of, the P-sheet structure. The hypervariable regions in each chain are held 25 together in close proximity by the FRs and, with the hypervariable regions from the other chain, contribute to the formation of the antigen-binding site of antibodies (see Kabat et al., Sequences of Proteins of Inmmunological Interest, 5th Ed. Public Health Service, National Institutes of Health, Bethesda, MD. (1991)). The constant domains are not involved directly in binding an antibody to an antigen, but exhibit various effector functions, such as 30 participation of the antibody in antibody dependent cellular cytotoxicity (ADCC). 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 generally comprises amino acid residues from a "complementarity determining region" or "CDR" (e.g. around about residues 24-34 (LI), 50-56 (L2) and 89-97 (L3) in the VL, and around about 1 38 WO 2004/094476 PCT/US2004/011793 35 (H1), 50-65 (H2) and 95-102 (H113) in the VH (in one embodiment, H1 is around about 31 35); Kabat et al., Sequences of Proteins of Immunological Interest, 5th Ed. Public Health Service, National Institutes of Health, Bethesda, MD. (1991)) and/or those residues from a "hypervariable loop" (e.g. residues 26-32 (LI), 50-52 (L2) and 91-96 (L3) in the VL, and 26 5 32 (H1), 53-55 (H2) and 96-101 (H113) in the VH; Chothia and Lesk J. Mol. Biol. 196:901-917 (1987)). 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 10 can be present in minor amounts. Monoclonal antibodies are highly specific, being directed against a single antigenic site. Furthermore, in contrast to polyclonal antibody preparations which include different antibodies directed against different determinants (epitopes), each monoclonal antibody is directed against a single determinant on the antigen. In addition to their specificity, the monoclonal antibodies are advantageous in that they can be synthesized 15 uncontaminated by other antibodies. The modifier "monoclonal" is not to be construed as requiring production of the antibody by any particular method. For example, the monoclonal antibodies useful in the present invention can be prepared by the hybridoma methodology first described by Kohler et al., Nature, 256:495 (1975), or can be made using recombinant DNA methods in bacterial, eukaryotic animal or plant cells (see, e.g., U.S. Patent No. 20 4,816,567). The "monoclonal antibodies" can also be isolated from phage antibody libraries using the techniques described in Clackson et al., Nature, 352:624-628 (1991), Marks et al., J. Mol. Biol., 222:581-597 (1991), and the Examples below, for example. The monoclonal antibodies herein include "chimeric" antibodies in which a portion of the heavy and/or light chain is identical with or homologous to corresponding sequences in 25 antibodies derived from a particular species or belonging to a particular antibody class or subclass, while the remainder of the chain(s) is identical with or homologous to corresponding sequences in antibodies derived from another species or belonging to another antibody class or subclass, as well as fragments of such antibodies, so long as they exhibit a biological activity of this invention (see U.S. Patent No. 4,816,567; and Morrison et al., Proc. 30 Natl. Acad. Sci. USA, 81:6851-6855 (1984)). Chimeric antibodies of interest herein include "primatized" antibodies comprising variable domain antigen-binding sequences derived from a non-human primate (e.g. Old World Monkey, Ape etc), and human constant region sequences. 39 WO 2004/094476 PCT/US2004/011793 An "intact" antibody is one which comprises an antigen-binding site as well as a CL and at least heavy chain constant domains, CHI, CH2 and CH 3 . The constant domains can be native sequence constant domains (e.g. human native sequence constant domains) or amino acid sequence variant thereof. Preferably, the intact antibody has one or more effector 5 functions. "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(ab') 2 , and Fv fragments; diabodies; linear antibodies (see U.S. Patent No. 5,641,870, Example 2; Zapata et al., Protein Eng. 8(10): 1057-1062 [1995]); single-chain 10 antibody molecules; and multispecific antibodies formed from antibody fragments. The expression "linear antibodies" generally refers to the antibodies described in Zapata et al., Protein Eng., 8(10):1057-1062 (1995). Briefly, these antibodies comprise a pair of tandem Fd segments (VH-CH1-VH-CH1) which, together with complementary light chain polypeptides, form a pair of antigen binding regions. Linear antibodies can be bispecific or 15 monospecific. Papain digestion of antibodies produces two identical antigen-binding fragments, called "Fab" fragments, and a residual "Fc" fragment, a designation reflecting the ability to crystallize readily. The Fab fragment consists of an entire L chain along with the variable region domain of the H chain (VH), and the first constant domain of one heavy chain (CHi). 20 Each Fab fragment is monovalent with respect to antigen binding, i.e., it has a single antigen binding site. Pepsin treatment of an antibody yields a single large F(ab') 2 fragment which roughly corresponds to two disulfide linked Fab fragments having divalent antigen-binding activity and is still capable of cross-linking antigen. Fab' fragments differ from Fab fragments by having additional few residues at the carboxy terminus of the CH1 domain 25 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. 30 The Fc fragment comprises the carboxy-terminal portions of both H chains held together by disulfides. The effector ftinctions of antibodies are determined by sequences in the Fe region, which region is also the part recognized by Fe receptors (FcR) found on certain types of cells. 40 WO 2004/094476 PCT/US2004/011793 "Fv" is the minimum antibody fragment which contains a complete antigen recognition and -binding site. This fragment consists of a dimer of one heavy- and one light chain variable region domain in tight, non-covalent association. From the folding of these two domains emanate six hypervariable loops (3 loops each from the H and L chain) that 5 contribute the amino acid residues for antigen binding and 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. "Single-chain Fv" also abbreviated as "sFv" or "scFv" are antibody fragments that 10 comprise the VH and VL antibody domains connected into a single polypeptide chain. Preferably, the sFv polypeptide further comprises a polypeptide linker between the VH and VL domains which enables the sFv to form the desired structure for antigen binding. For a review of sFv, see Pluckthun in The Pharmacology of Monoclonal Antibodies, vol. 113, Rosenburg and Moore eds., Springer-Verlag, New York, pp. 269-315 (1994); Borrebaeck 15 1995, infra. The term "diabodies" refers to small antibody fragments prepared by constructing sFv fragments (see preceding paragraph) with short linkers (about 5-10 residues) between the VH and VL domains such that inter-chain but not intra-chain pairing of the V domains is achieved, resulting in a bivalent fragment, i.e., fragment having two antigen-binding sites. 20 Bispecific diabodies are heterodimers of two "crossover" sFv fragments in which the VH and VL domains of the two antibodies are present on different polypeptide chains. 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). "Humanized" forms of non-human (e.g., rodent) antibodies are chimeric antibodies 25 that contain minimal sequence derived from the non-human antibody. For the most part, humanized antibodies are human immunoglobulins (recipient antibody) in which residues from a hypervariable region of the recipient are replaced by residues from a hypervariable region of a non-human species (donor antibody) such as mouse, rat, rabbit or non-human primate having the desired antibody specificity, affinity, and capability. In some instances, 30 framework region (FR) residues of the human immunoglobulin are replaced by corresponding non-human residues. Furthermore, humanized antibodies can comprise residues that are not found in the recipient antibody or in the donor antibody. These modifications are made to further refine antibody performance. In general, the humanized antibody will comprise substantially all of at least one, and typically two, variable domains, 41 WO 2004/094476 PCT/US2004/011793 in which all or substantially all of the hypervariable loops correspond to those of a non human immunoglobulin and all or substantially all of the FRs are those of a human immunoglobulin sequence. The humanized antibody optionally also will comprise at least a portion of an immunoglobulin constant region (Fc), typically that of a human 5 immunoglobulin. For further details, see 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). A "species-dependent antibody," e.g., a mammalian anti-human IgE antibody, is an antibody which has a stronger binding affinity for an antigen from a first mammalian species 10 than it has for a homologue of that antigen from a second mammalian species. Normally, the species-dependent antibody "bind specifically" to a human antigen (i.e., has a binding affinity (Kd) value of no more than about 1 x 10 -7 M, preferably no more than about 1 x 10 8 and most preferably no more than about 1 x 10 -9 M) but has a binding affinity for a homologue of the antigen from a second non-human mammalian species which is at least about 50 fold, or 15 at least about 500 fold, or at least about 1000 fold, weaker than its binding affinity for the human antigen. The species-dependent antibody can be of any of the various types of antibodies as defined above, but preferably is a humanized or human antibody. A "STOP-1 binding oligopeptide" is an oligopeptide that binds, preferably specifically, to a STOP-1 polypeptide as described herein. STOP-1 binding oligopeptides can 20 be chemically synthesized using known oligopeptide synthesis methodology or can be prepared and purified using recombinant technology. STOP-1 binding oligopeptides are usually at least about 5 amino acids in length, alternatively at least about 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 15 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100 amino acids in length or more, wherein such oligopeptides that are capable of binding, preferably specifically, to a STOP-1 polypeptide as described herein. According to one embodiment, the STOP-1 binding oligopeptide binds to the same or overlapping region that the 6B12 antibody binds. STOP-1 ;0 binding oligopeptides can be identified without undue experimentation using known techniques. In this regard, it is noted that techniques for screening oligopeptide libraries for oligopeptides that are capable of specifically binding to a polypeptide target are known in the art (see, e.g., U.S. Patent Nos. 5,556,762, 5,750,373, 4,708,871, 4,833,092, 5,223,409, 5,403,484, 5,571,689, 5,663,143; PCT Publication Nos. WO 84/03506 and WO84/03564; 42 WO 2004/094476 PCT/US2004/011793 ieysen et al., Proc. Natl. Acad. Sci. U.S.A., 81:3998-4002 (1984); Geysen et al., Proc. Natl. Acad. Sci. U.S.A., 82:178-182 (1985); Geysen et al., in Synthetic Peptides as Antigens, 130 149 (1986); Geysen et al., J. Immunol. Meth., 102:259-274 (1987); Schoofs et al., J. Immunol., 140:611-616 (1988), Cwirla, S. E. et al. (1990) Proc. Natl. Acad. Sci. USA, 5 87:6378; Lowman, H.B. et al. (1991) Biochemistry, 30:10832; Clackson, T. et al. (1991) Nature, 352: 624; Marks, J. D. et al. (1991), J. Mol. Biol., 222:581; Kang, A.S. et al. (1991) Proc. Natl. Acad. Sci. USA, 88:8363, and Smith, G. P. (1991) Current Opin. Biotechnol., 2:668). A polypeptide, antibody, antagonist or composition of this invention "which binds" an 10 antigen of interest, e.g. a tumor-associated polypeptide antigen target such as STOP-1, is one that binds the antigen with sufficient affinity such that a polypeptide, antibody, antagonist or composition is useful as a diagnostic and/or therapeutic agent in targeting a cell or tissue expressing the antigen, and does not significantly cross-react with other proteins. In such embodiments, the extent of binding of the polypeptide, antibody, antagonist or composition 15 to a "non-target" protein will be less than about 10% of the binding of the polypeptide, antibody, antagonist or composition to its particular target protein as determined by fluorescence activated cell sorting (FACS) analysis or radioimmunoprecipitation (RIA). With regard to the binding of a polypeptide, antibody, antagonist or composition to a target molecule, the term "specific binding" or "specifically binds to" or is "specific for" a 0 particular polypeptide or an epitope on a particular polypeptide target means binding that is measurably different from a non-specific interaction. Specific binding can be measured, for example, by determining binding of a molecule compared to binding of a control molecule, which generally is a molecule of similar structure that does not have binding activity. For example, specific binding can be determined by competition with a control molecule that is 15 similar to the target, for example, an excess of non-labeled target. In this case, specific binding is indicated if the binding of the labeled target to a probe is competitively inhibited by excess unlabeled target. The term "specific binding" or "specifically binds to" or is "specific for" a particular polypeptide or an epitope on a particular polypeptide target as used herein can be exhibited, for example, by a molecule having a Kd for the target of at least ;0 about 10 -4 M, alternatively at least about 10 -5 M, alternatively at least about 10 -6 M, alternatively at least about 10 -7 M, alternatively at least about 10 -8 M, alternatively at least about 10 -9 M, alternatively at least about 10-10 M, alternatively at least about 10- 1 M, alternatively at least about 10-12 M, or greater. In one embodiment, the term "specific binding" refers to binding where a molecule binds to a particular polypeptide or epitope on a 43 WO 2004/094476 PCT/US2004/011793 particular polypeptide without substantially binding to any other polypeptide or polypeptide epitope (e.g., a non-STOP-1 protein). It is understood that an antibody that specifically binds to a human native STOP-1 polypeptide may also bind a non-human native STOP-1 polypeptide. 5 A polypeptide, antibody, antagonist or composition that "inhibits the growth" of tumor cells expressing a STOP-1 polypeptide or a "growth inhibitory" polypeptide, antibody, antagonist or composition is one which results in measurable growth inhibition of cancer cells expressing or overexpressing the appropriate STOP-1 polypeptide. Preferred growth inhibitory polypeptides, antibodies, antagonists or compositions inhibit growth of STOP-1 10 expressing tumor cells by greater than 20%, preferably from about 20% to about 50%, and even more preferably, by greater than 50% (e.g., from about 50% to about 100%) as compared to the appropriate control, the control typically being tumor cells not treated with the polypeptide, antibody, antagonist or composition being tested. In one embodiment, growth inhibition can be measured at an antibody concentration of about 0.1 to 30 ig/ml or 15 about 0.5 nM to 200 nM in cell culture, where the growth inhibition is determined 1-10 days after exposure of the tumor cells to the antibody. Growth inhibition of tumor cells in vivo can be determined in various ways such as is described in the Experimental Examples section below. The antibody is growth inhibitory in vivo if administration of the anti-STOP-1 antibody at about 1 jig/kg to about 100 mg/kg body weight results in reduction in tumor size 20 or tumor cell proliferation within about 5 days to 3 months from the first administration of the antibody, preferably within about 5 to 30 days. Antibody "effector functions" refer to those biological activities attributable to the Fc region (a native sequence Fc region or amino acid sequence variant Fc region) of an antibody, and vary with the antibody isotype. Examples of antibody effector functions include: CIq Z5 binding and complement dependent cytotoxicity; Fc receptor binding; antibody-dependent cell-mediated cytotoxicity (ADCC); phagocytosis; down regulation of cell surface receptors (e.g., B cell receptor); and B cell activation. "Antibody-dependent cell-mediated cytotoxicity" or "ADCC" refers to a form of cytotoxicity in which secreted Ig bound onto Fc receptors (FcRs) present on certain cytotoxic 10 cells (e.g., Natural Killer (NK) cells, neutrophils, and macrophages) enable these cytotoxic effector cells to bind specifically to an antigen-bearing target cell and subsequently kill the target cell with cytotoxins. The antibodies "arm" the cytotoxic cells and are absolutely required for such killing. The primary cells for mediating ADCC, NK cells, express FcyRHI only, whereas monocytes express FcyRI, FcyRII and FcyRIu. FcR expression on 44 WO 2004/094476 PCT/US2004/011793 hematopoietic cells is summarized in Table 3 on page 464 of Ravetch and Kinet, Annu. Rev. Immunol. 9:457-92 (1991). To assess ADCC activity of a molecule of interest, an in vitro ADCC assay, such as that described in US Patent No. 5,500,362 or 5,821,337 can be performed. Useful effector cells for such assays include peripheral blood mononuclear cells 5 (PBMC) and Natural Killer (NK) cells. Alternatively, or additionally, ADCC activity of the molecule of interest can be assessed in vivo, e.g., in a animal model such as that disclosed in Clynes et al. (USA) 95:652-656 (1998). "Fc receptor" or "FcR" describes a receptor that binds to the Fc region of an antibody. The preferred FcR is a native sequence human FcR. Moreover, a preferred FcR is one which 10 binds an IgG antibody (a gamma receptor) and includes receptors of the FcyRI, FcyRII and FcyREII subclasses, including allelic variants and alternatively spliced forms of these receptors. Fc'yRII receptors include FcyRIIA (an "activating receptor") and FcyRIlB (an "inhibiting receptor"), which have similar amino acid sequences that differ primarily in the cytoplasmic domains thereof. Activating receptor FcyRIIA contains an immunoreceptor 15 tyrosine-based activation motif (ITAM) in its cytoplasmic domain. Inhibiting receptor FcyRIIB contains an immunoreceptor tyrosine-based inhibition motif (ITIM) in its cytoplasmic domain. (see review M. in Dadron, Annu. Rev. Immunol. 15:203-234 (1997)). FcRs are reviewed in Ravetch and Kinet, Annu. Rev. Inmunol. 9:457-492 (1991); Capel et al., Immunomethods 4:25-34 (1994); and de Haas et al., J. Lab. Clin. Med. 126:330-41 20 (1995). Other FcRs, including those to be identified in the future, are encompassed by the term "FcR" herein. The term also includes the neonatal receptor, FcRn, which is responsible for the transfer of maternal IgGs to the fetus (Guyer et al., J. Immunol. 117:587 (1976) and Kim et al., J. Immunol. 24:249 (1994)). "Human effector cells" are leukocytes which express one or more FcRs and perform 25 effector functions. Preferably, the cells express at least FcyRII and perform ADCC effector function. Examples of human leukocytes which mediate ADCC include peripheral blood mononuclear cells (PBMC), natural killer (NK) cells, monocytes, cytotoxic T cells and neutrophils; with PBMCs and NK cells being preferred. The effector cells can be isolated from a native source, e.g., from blood. 30 "Complement dependent cytotoxicity" or "CDC" refers to the lysis of a target cell in the presence of complement. Activation of the classical complement pathway is initiated by the binding of the first component of the complement system (Clq) to antibodies (of the appropriate subclass) which are bound to their cognate antigen. To assess complement 45 WO 2004/094476 PCT/US2004/011793 activation, a CDC assay, e.g., as described in Gazzano-Santoro et al., J. Immunol. Methods 202:163 (1996), can be performed. 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 5 include, but are not limited to, carcinoma, lymphoma, blastoma, sarcoma, and leukemia or lymphoid malignancies. More particular examples of such cancers include squamous cell cancer (e.g., epithelial squamous cell cancer), lung cancer including small-cell lung cancer, non-small cell lung cancer, adenocarcinoma of the lung and squamous carcinoma of the lung, cancer of the peritoneum, hepatocellular cancer, gastric or stomach cancer including 10 gastrointestinal cancer, pancreatic cancer, glioblastoma, cervical cancer, ovarian cancer, liver cancer, bladder cancer, cancer of the urinary tract, hepatoma, breast cancer, colon cancer, rectal cancer, colorectal cancer, endometrial or uterine carcinoma, salivary gland carcinoma, kidney or renal cancer, prostate cancer, vulval cancer, thyroid cancer, hepatic carcinoma, anal carcinoma, penile carcinoma, melanoma, multiple myeloma and B-cell lymphoma, brain, as 15 well as head and neck cancer, and associated metastases. The terms "cell proliferative disorder" and "proliferative disorder" refer to disorders that are associated with some degree of abnormal cell proliferation. In one embodiment, the cell proliferative disorder is cancer. In one embodiment, the cell proliferative disorder is desmoplasia. 20 "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 term abnormal angiogenesis according to this invention occurs when new blood vessels either grow excessively, insufficiently or inappropriately (e.g., the location, timing or onset of the angiogenesis being undesired from a medical standpoint) in a diseased state or 25 such that it causes a diseased state. "Excessive, inappropriate or uncontrolled angiogenesis" occurs in a diseased state when there is new blood vessel growth that contributes to the worsening of the diseased state, such as in cancer, especially vascularized solid tumors and metastatic tumores (including colon, lung cancer (especially small-cell lung cancer), or prostate cancer), diseases caused by ocular neovascularisation, especially diabetic blindness, 30 retinopathies, primarily diabetic retinopathy or age-induced macular degeneration and rubeosis; psoriasis, haemangioblastoma such as haemangioma; inflammatory renal diseases, such as glomerulonephritis, especially mesangioproliferative glomerulonephritis, haemolytic uremic syndrome, diabetic nephropathy or hypertensive nephrosclerosis; various imflammatory diseases, such as arthritis, especially rheumatoid arthritis, inflammatory bowel 46 WO 2004/094476 PCT/US2004/011793 disease, psorsasis, sarcoidosis, arterial arteriosclerosis and diseases occurring after transplants, endometriosis or chronic asthma and more than 70 other conditions. The new blood vessels can feed the diseased tissues, destroy normal tissues, and in the case of cancer, the new vessels can allow tumor cells to escape into the circulation and lodge in other organs 5 (tumor metastases). Insufficient angiogenesis occurs when there is inadequate blood vessels growth that contributes to the worsening of a diseased state, e.g., in diseases such as coronary artery disease, stroke, and delayed wound healing. Further, ulcers, strokes, and heart attacks can result from the absence of angiogenesis that normally required for natural healing. The present invention contemplates treating those patients that are at risk of developing the 10 above-mentioned illnesses. Other patients that are candidates for receiving the STOP-1 antagonists of this invention have, or are at risk for developing, abnormal proliferation of fibrovascular tissue, acne rosacea, acquired immune deficiency syndrome, artery occlusion, atopic keratitis, bacterial ulcers, Bechets disease, blood borne tumors, carotid obstructive disease, choroidal 15 neovascularization, chronic inflammation, chronic retinal detachment, chronic uveitis, chronic vitritis, contact lens overwear, corneal graft rejection, corneal neovascularization, corneal graft neovascularization, Crohn's disease, Eales disease, epidemic keratoconjunctivitis, fungal ulcers, Herpes simplex infections, Herpes zoster infections, hyperviscosity syndromes, Kaposi's sarcoma, leukemia, lipid degeneration, Lyme's disease, 20 marginal keratolysis, Mooren ulcer, Mycobacteria infections other than leprosy, myopia, ocular neovascular disease, optic pits, Osler-Weber syndrome (Osler-Weber-Rendu, osteoarthritis, Pagets disease, pars planitis, pemphigoid, phylectenulosis, polyarteritis, post laser complications, protozoan infections, pseudoxanthoma elasticum, pterygium keratitis sicca, radial keratotomy, retinal neovascularization, retinopathy of prematurity, retrolental 25 fibroplasias, sarcoid, scleritis, sickle cell anemia, Sogrens syndrome, solid tumors, Stargarts disease, Steven's Johnson disease, superior limbic keratitis, syphilis, systemic lupus, Terrien's marginal degeneration, toxoplasmosis, trauma, tumors of Ewing sarcoma, tumors of neuroblastoma, tumors of osteosarcoma, tumors of retinoblastoma, tumors of rhabdomyosarcoma, ulcerative colitis, vein occlusion, Vitamin A deficiency and Wegeners 30 sarcoidosis, undesired angiogenesis associated with diabetes, parasitic diseases, abnormal wound healing, hypertrophy following surgery, injury or trauma, inhibition of hair growth, inhibition of ovulation and corpus luteum formation, inhibition of implantation and inhibition of embryo development in the uterus. 47 WO 2004/094476 PCT/US2004/011793 Anti-angiogenesis therapies are useful in the general treatment of graft rejection, lung inflammation, nephrotic syndrome, preeclampsia, pericardial effusion, such as that associated with pericarditis, and pleural effusion, diseases and disorders characterized by undesirable vascular permeability, e.g., edema associated with brain tumors, ascites associated with 5 malignancies, Meigs' syndrome, lung inflammation, nephrotic syndrome, pericardial effusion and pleural effusion and the like. Other angiogenesis-dependent diseases that may be treated with the compositions of this invention include angiofibroma (abnormal blood of vessels which are prone to bleeding), neovascular glaucoma (abnormal growth of blood vessels in the eye), arteriovenous 10 malformations (abnormal communication between arteries and veins), nonunion fractures (fractures that will not heal), atherosclerotic plaques (hardening of the arteries), pyogenic granuloma (common skin lesion composed of blood vessels), scleroderma (a form of connective tissue disease), hemangioma (tumor composed of blood vessels), trachoma (leading cause of blindness in the third world), hemophilic joints, vascular adhesions and 15 hypertrophic scars (abnormal scar formation). Since blood vessels play an important role in the regulation of bone turnover and growth, potentiators or agonists according to this invention may stimulate or enhance of bone and/or cartilage repair from disease or injury or by blocking inflammation or processes of tissue destruction (collagenase activity, osteoclast activity, etc.) mediated by inflammatory 20 processes. Bone injuries or diseases to be treated with STOP-1 potentiators or agonists of this invention include periodontal diseases, other tooth-repair processes, osteoporosis and fractures. A "stromal targeting agent" according to this invention is an agent that substantially recognizes and binds stromal tissue compared to other tissue. Stromal tissue is the 25 connective tissue framework of an organ, gland, or other structure, as distinguished from the tissues performing the special function of the organ or part. Examples of stromal targeting agents include antibodies that specifically bind to FAP, fascin, HSP47, mesothelin and prostate stem antigen. A polypeptide, antibody, antagonist or composition of this invention which "induces 30 cell death" is one which causes a viable cell to become nonviable. The cell is one which expresses a STOP-1 polypeptide, preferably a cell that overexpresses a STOP-1 polypeptide as compared to a normal cell of the same tissue type. Preferably, the cell is a cancer cell, e.g., a breast, ovarian, stomach, endometrial, salivary gland, lung, kidney, colon, thyroid, pancreatic or bladder cell. Cell death in vitro can be determined in the absence of 48 WO 2004/094476 PCT/US2004/011793 complement and immune effector cells to distinguish cell death induced by antibody dependent cell-mediated cytotoxicity (ADCC) or complement dependent cytotoxicity (CDC). Thus, the assay for cell death can be performed using heat inactivated serum (i.e., in the absence of complement) and in the absence of immune effector cells. To determine whether a 5 polypeptide, antibody, antagonist or composition of this invention is able to induce cell death, loss of membrane integrity as evaluated by uptake of propidium iodide (PI), trypan blue (see Moore et al. Cvtotechnology 17:1-11 (1995)) or 7AAD can be assessed relative to untreated cells. Preferred cell death-inducing polypeptides, antibodies, antagonists or compositions are those which induce PI uptake in the PI uptake assay in BT474 cells. 10 A "STOP-I-expressing cell" is a cell which expresses an endogenous or transfected STOP-1 polypeptide either on the cell surface or in a secreted form. A "STOP-1-expressing cancer" is a cancer comprising cells that have a STOP-1 polypeptide present on the cell surface or that produce and secrete a STOP-1 polypeptide. In another embodiment, a "STOP 1-expressing cancer" optionally produces and secretes sufficient levels of STOP-1 15 polypeptide, such that a polypeptide, antibody, antagonist or composition of this invention can bind thereto and have a therapeutic effect with respect to the cancer. A cancer which "overexpresses" a STOP-1 polypeptide is one which has significantly higher levels of STOP 1 polypeptide at the cell surface thereof, or produces and secretes, compared to a noncancerous cell of the same tissue type. Such overexpression can be caused by gene 20 amplification or by increased transcription or translation. STOP-1 polypeptide overexpression can be determined in a diagnostic or prognostic assay by evaluating increased levels of the STOP-1 protein present on the surface of a cell, or secreted by the cell (e.g., via an immunohistochemistry assay using anti-STOP-1 antibodies prepared against an isolated STOP-1 polypeptide which can be prepared using recombinant DNA technology from an 25 isolated nucleic acid encoding the STOP-1 polypeptide; FACS analysis, etc.). Alternatively, or additionally, one can measure levels of STOP-1 polypeptide-encoding nucleic acid or mRNA in the cell, e.g., via fluorescent in situ hybridization using a nucleic acid based probe corresponding to a STOP-l-encoding nucleic acid or the complement thereof; (FISH; see WO98/45479 published October, 1998), Southern blotting, Northern blotting, or polymerase 30 chain reaction (PCR) techniques, such as real time quantitative PCR (RT-PCR). One can also study STOP-1 polypeptide overexpression by measuring shed antigen in a biological fluid such as serum, e.g., using antibody-based assays (see also, e.g., U.S. Patent No. 4,933,294 issued June 12, 1990; WO91/05264 published April 18, 1991; U.S. Patent 5,401,638 issued March 28, 1995; and Sias et al., J. Immunol. Methods 132:73-80 (1990)). Aside from the 49 WO 2004/094476 PCT/US2004/011793 above assays, various in vivo assays are available to the skilled practitioner. For example, one can expose cells within the body of the mammal to an antibody which is optionally labeled with a detectable label, e.g., a radioactive isotope, and binding of the antibody to cells in the mammal can be evaluated, e.g., by external scanning for radioactivity or by analyzing a 5 biopsy taken from a mammal previously exposed to the antibody. The word "label" when used herein refers to a detectable compound or composition which is conjugated directly or indirectly to the polypeptide, antibody, antagonist or composition so as to generate a "labeled" a polypeptide, antibody, antagonist or composition. The label can be detectable by itself (e.g. radioisotope labels or fluorescent labels) or, in the 10 case of an enzymatic label, can catalyze chemical alteration of a substrate compound or composition which is detectable. The term "cytotoxic agent" as used herein refers to a substance that inhibits or prevents the function of cells and/or causes destruction of cells. The term is intended to include radioactive isotopes (e.g., At 211 , I3l, 1125, Y 90 , Rel 86 , Re' 88 , Sm 1 53 , Bi 21 2 , p 32 and 15 radioactive isotopes of Lu), chemotherapeutic agents e.g. methotrexate, adriamicin, vinca alkaloids (vincristine, vinblastine, etoposide), doxorubicin, melphalan, mitomycin C, chlorambucil, daunorubicin or other intercalating agents, enzymes and fragments thereof such as nucleolytic enzymes, antibiotics, and toxins such as small molecule toxins or enzymatically active toxins of bacterial, fungal, plant or animal origin, including fragments 20 and/or variants thereof, and the various antitumor or anticancer agents disclosed below. Other cytotoxic agents are described below. A tumoricidal agent causes destruction of tumor cells. A "chemotherapeutic agent" is a chemical compound useful in the treatment of cancer. Examples of chemotherapeutic agents include alkylating agents such as thiotepa and 15 CYTOXAN® cyclosphosphamide; alkyl sulfonates such as busulfan, improsulfan and piposulfan; aziridines such as benzodopa, carboquone, meturedopa, and uredopa; ethylenimines and methylamelamines including altretamine, triethylenemelamine, trietylenephosphoramide, triethiylenethiophosphoramide and trimethylolomelamine; acetogenins (especially bullatacin and bullatacinone); a camptothecin (including the synthetic ;0 analogue topotecan); bryostatin; callystatin; CC-1065 (including its adozelesin, carzelesin and bizelesin synthetic analogues); cryptophycins (particularly cryptophycin 1 and cryptophycin 8); dolastatin; duocarmycin (including the synthetic analogues, KW-2189 and CB1-TM1); eleutherobin; pancratistatin; a sarcodictyin; spongistatin; nitrogen mustards such as chlorambucil, chlornaphazine, cholophosphamide, estramustine, ifosfamide, 50 WO 2004/094476 PCT/US2004/011793 mechlorethamine, mechlorethamine oxide hydrochloride, melphalan, novembichin, phenesterine, prednimustine, trofosfamide, uracil mustard; nitrosureas such as carmustine, chlorozotocin, fotemustine, lomustine, nimustine, and ranimnustine; antibiotics such as the enediyne antibiotics (e. g., calicheamicin, especially calicheamicin gammall and 5 calicheanmicin omegall (see, e.g., Agnew, Chem Intl. Ed. Engl., 33: 183-186 (1994)); dynemicin, including dynemicin A; bisphosphonates, such as clodronate; an esperamicin; as well as neocarzinostatin chromophore and related chromoprotein enediyne antiobiotic chromophores), aclacinomysins, actinomycin, authramycin, azaserine, bleomycins, cactinomycin, carabicin, carminomycin, carzinophilin, chromomycinis, dactinomycin, 10 daunorubicin, detorubicin, 6-diazo-5-oxo-L-norleucine, ADRIAMYCIN® doxorubicin (including morpholino-doxorubicin, cyanomorpholino-doxorubicin, 2-pyrrolino-doxorubicin and deoxydoxorubicin), epirubicin, esorubicin, idarubicin, marcellomycin, mitomycins such as mitomycin C, mycophenolic acid, nogalamycin, olivomycins, peplomycin, potfiromycin, puromycin, quelamycin, rodorubicin, streptonigrin, streptozocin, tubercidin, ubenimex, 15 zinostatin, zorubicin; anti-metabolites such as methotrexate and 5-fluorouracil (5-FU); folic acid analogues such as denopterin, methotrexate, pteropterin, trimetrexate; purine analogs such as fludarabine, 6-mercaptopurine, thiamiprine, thioguanine; pyrimidine analogs such as ancitabine, azacitidine, 6-azauridine, carmofur, cytarabine, dideoxyuridine, doxifluridine, enocitabine, floxuridine; androgens such as calusterone, dromostanolone propionate, 20 epitiostanol, mepitiostane, testolactone; anti- adrenals such as aminoglutethimide, mitotane, trilostane; folic acid replenisher such as frolinic acid; aceglatone; aldophosphamide glycoside; aminolevulinic acid; eniluracil; amsacrine; bestrabucil; bisantrene; edatraxate; defofamine; demecolcine; diaziquone; elfornithine; elliptinium acetate; an epothilone; etoglucid; gallium nitrate; hydroxyurea; lentinan; lonidainine; maytansinoids such as 25 maytansine and ansamitocins; mitoguazone; mitoxantrone; mopidanmol; nitraerine; pentostatin; phenamet; pirarubicin; losoxantrone; podophyllinic acid; 2- ethylhydrazide; procarbazine; PSK® polysaccharide complex (JHS Natural Products, Eugene, OR); razoxane; rhizoxin; sizofiran; spirogermanium; tenuazonic acid; triaziquone; 2,2',2" trichlorotriethylamine; trichothecenes (especially T-2 toxin, verracurin A, roridin A and 30 anguidine); urethan; vindesine; dacarbazine; mannomustine; mitobronitol; mitolactol; pipobroman; gacytosine; arabinoside ("Ara-C"); cyclophosphamide; thiotepa; taxoids, e.g., TAXOL® paclitaxel (Bristol- Myers Squibb Oncology, Princeton, N.J.), ABRAXANET m Cremophor-free, albumin-engineered nanoparticle formulation of paclitaxel (American Pharmaceutical Partners, Schaumberg, Illinois), and TAXOTERE® doxetaxel (Rh6ne 51 WO 2004/094476 PCT/US2004/011793 Poulenc Rorer, Antony, France); chloranbucil; GEMZAR® gemcitabine; 6- thioguanine; mercaptopurine; methotrexate; platinum analogs such as cisplatin and carboplatin; vinblastine; platinum; etoposide (VP-16); ifosfamide; mitoxantrone; vincristine; NAVELBINE® vinorelbine; novantrone; teniposide; edatrexate; daunomycin; aminopterin; 5 xeloda; ibandronate; CPT-11; topoisomerase inhibitor RFS 2000; difluorometlhylornithine (DMFO); retinoids such as retinoic acid; capecitabine; and pharmaceutically acceptable salts, acids or derivatives of any of the above. Also included in this definition are anti-hormonal agents that act to regulate or inhibit hormone action on tumors such as anti-estrogens and selective estrogen receptor modulators 10 (SERMs), including, for example, tamoxifen (including NOLVADEX® tamoxifen), raloxifene, droloxifene, 4-hydroxytamoxifen, trioxifene, keoxifene, LY1 17018, onapristone, and FARESTON. toremifene; aromatase inhibitors that inhibit the enzyme aromatase, which regulates estrogen production in the adrenal glands, such as, for example, 4(5)-imidazoles, aminoglutethimide, MEGASE® megestrol acetate, AROMASIN® exemestane, formestanie, 15 fadrozole, RIVISOR@ vorozole, FEMARA® letrozole, and ARIMIDEX® anastrozole; and anti-androgens such as flutamide, nilutamide, bicalutamide, leuprolide, and goserelin; as well as troxacitabine (a 1,3-dioxolane nucleoside cytosine analog); antisense oligonucleotides, particularly those which inhibit expression of genes in signaling pathways implicated in abherant cell proliferation, such as, for example, PKC-alpha, Ralf and H-Ras; ribozymes such 20 as a VEGF expression inhibitor (e.g., ANGIOZYME@ ribozyme) and a HER2 expression inhibitor; vaccines such as gene therapy vaccines, for example, ALLOVECTIN® vaccine, LEUVECTIN® vaccine, and VAXID@ vaccine; PROLEUKIN® rTL-2; LURTOTECAN® topoisomerase 1 inhibitor; ABARELIX® rmRH; and pharmaceutically acceptable salts, acids or derivatives of any of the above. 25 A "growth inhibitory agent" when used herein refers to a compound or composition which inhibits growth of a cell, especially a STOP-1-expressing cancer cell, either in vitro or in vivo. Thus, the growth inhibitory agent can be one which significantly reduces the percentage of STOP-1-expressing cells 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 30 that induce G1 arrest and M-phase arrest. Classical M-phase blockers include the vincas (vincristine and vinblastine), taxanes, and topoisomerase II inhibitors such as doxorubicin, epimrubicin, daunorubicin, etoposide, and bleomycin. Those agents that arrest G1 also spill over into S-phase arrest, for example, DNA alkylating agents such as tamoxifen, prednisone, dacarbazine, mechlorethamine, cisplatin, methotrexate, 5-fluorouracil, and ara-C. Further 52 WO 2004/094476 PCT/US2004/011793 information can be found in The Molecular Basis of Cancer, Mendelsohn and Israel, eds., Chapter 1, entitled "Cell cycle regulation, oncogenes, and antineoplastic drugs" by Murakami et al. (WB Saunders: Philadelphia, 1995), especially p. 13. The taxanes (paclitaxel and docetaxel) are anticancer drugs both derived from the yew tree. Docetaxel (TAXOTERE®, 5 Rhone-Poulenc Rorer), derived from the European yew, is a semisynthetic analogue of paclitaxel (TAXOL®, Bristol-Myers Squibb). Paclitaxel and docetaxel promote the assembly of microtubules from tubulin dimers and stabilize microtubules by preventing depolymerization, which results in the inhibition of mitosis in cells. "Doxorubicin" is an anthracycline antibiotic. The full chemical name of doxorubicin 10 is (8S-cis)-10-[(3-amino-2,3,6-trideoxy-a-L-lyxo-hexapyranosyl)oxy]-7,8,9,10-tetrahydro 6,8,1 1-trihydroxy-8-(hydroxyacetyl)-1-methoxy-5,12-naphthacenedione. The term "package insert" is used to refer to instructions customarily included in commercial packages of therapeutic products, that contain information about the indications, usage, dosage, administration, contraindications and/or warnings concerning the use of such 15 therapeutic products. Table 1 /* * 20 * C-C increased from 12 to 15 * Z is average of EQ * B is average of ND * match with stop is M; stop-stop = 0; J (joker) match = 0 */ 25 #define _M -8 /* value of a match with a stop */ int _day[26] [26] = { /* ABCDEFGHIJKLMNOP QRSTUVWX YZ*/ /* A */ { 2, 0,-2, 0, 0,-4, 1,-1,-1, 0,-1,-2,-1, O,_M, 1, 0,-2, 1, 1, 0, 0,-6, 0,-3, 0}, 30 /* B */ { 0, 3,-4, 3, 2,-5, 0, 1,-2, 0, 0,-3,-2, 2,_M,-1, 1, 0, 0, 0, 0,-2,-5, 0,-3, 1}, /* C */ {-2,-4,15,-5,-5,-4,-3,-3,-2, 0,-5,-6,-5,-4,_M,-3,-5,-4, 0,-2, 0,-2,-8, 0, 0,-5}, /* D */ { 0, 3,-5, 4, 3,-6, 1, 1,-2, 0, 0,-4,-3, 2,_M,-1, 2,-1, 0, 0, 0,-2,-7, 0,-4, 2}, /* E */ { 0, 2,-5, 3, 4,-5, 0, 1,-2, 0, 0,-3,-2, 1,_M,-1, 2,-1, 0, 0, 0,-2,-7, 0,-4, 3}, /* F */ {-4,-5,-4,-6,-5, 9,-5,-2, 1, 0,-5, 2, 0,-4,_M,-5,-5,-4,-3,-3, 0,-1, 0, 0, 7,-5}, 53 WO 2004/094476 PCT/US2004/011793 /* G */ { 1, 0,-3, 1, 0,-5, 5,-2,-3, 0,-2,-4,-3, 0,_M,-1,-1,-3, 1, 0, 0,-1,-7, 0,-5, 0}, /* H */ {-1, 1,-3, 1, 1,-2,-2, 6,-2, 0, 0,-2,-2, 2,_M, 0, 3, 2,-1,-1, 0,-2,-3, 0, 0, 2}, /* 1*I {-1,-2,-2,-2,-2, 1,-3,-2, 5, 0,-2, 2, 2,-2, M,-2,-2,-2,-1, 0, 0, 4,-5, 0,-1,-2}, /*J*/ { 0, 0, 0, 0, 0, 0, 0, 0, 0, 0, ,0, 0, 0, 0,_M, 0, 0, 0, 0, 0, 0, 0, 0, 0, 0, 0}, 5 /* K */ {-1, 0,-5, 0, 0,-5,-2, 0,-2, 0, 5,-3, 0, 1,_M,-1, 1, 3, 0, 0, 0,-2,-3, 0,-4, 0}, /* L */ {-2,-3,-6,-4,-3, 2,-4,-2, 2, 0,-3, 6, 4,-3,_M,-3,-2,-3,-3,-1, 0, 2,-2, 0,-1,-2}, /* M */ {-1,-2,-5,-3,-2, 0,-3,-2, 2, 0, 0, 4, 6,-2,_M,-2,-1, 0,-2,-1, 0, 2,-4, 0,-2,-1}, /* N */ { 0, 2,-4, 2, 1,-4, 0, 2,-2, 0, 1,-3,-2, 2,_M,-1, 1, 0, 1, 0, 0,-2,-4, 0,-2, 1}, /* 0 */ {MM,_MM,_M,_M,_MMMM,_M,_M,M,_M,M, 10 0,_MM,_M,M,M, M,_M,_M,_M,_MM}, /* P */ { 1,-1,-3,-1,-1,-5,-1, 0,-2, 0,-1,-3,-2,-1,_M, 6, 0, 0, 1, 0, 0,-1,-6, 0,-5, 0}, /* Q */ { 0, 1,-5, 2, 2,-5,-1, 3,-2, 0, 1,-2,-1, 1,_M, 0, 4, 1,-1,-1, 0,-2,-5, 0,-4, 3}, /* R */ {-2, 0,-4,-1,-1,-4,-3, 2,-2, 0, 3,-3, 0, 0,_M, 0, 1, 6, 0,-1, 0,-2, 2, 0,-4, 0}, /* S */ { 1, 0, 0, 0, 0,-3, 1,-1,-1, 0, 0,-3,-2, 1, M, 1,-1, 0, 2, 1, 0,-1,-2, 0,-3, 0}, 15 /* T */ { 1, 0,-2, 0, 0,-3, 0,-1, 0, 0, 0,-1,-1, 0,_M, 0,-1,-1, 1, 3, 0, 0,-5, 0,-3, 0}, /*U*/ { 0, 0, 0, 0, 0, 0, 0, 0, 0, 0, 0, 0, 0, 0,_M, 0, 0, 0, 0, 00, 0, 0, 0, 0, 0, 0}, /* V */ { 0,-2,-2,-2,-2,-1,-1,-2, 4, 0,-2, 2, 2,-2, M,-1,-2,-2,-1, 0, 0, 4,-6, 0,-2,-2}, /* W */ {-6,-5,-8,-7,-7, 0,-7,-3,-5, 0,-3,-2,-4,-4,_M,-6,-5, 2,-2,-5, 0,-6,17, 0, 0,-6}, /*X*/ { 0, 0, 0, 0, 0, 0, 0, 0, 0, 0, 0, 0, 0, 0,_M, 0, 0, 0, 0, 0, 0, 0, 0, 0, 0, 0}, 20 /* Y */ {-3,-3, 0,-4,-4, 7,-5, 0,-1, 0,-4,-1,-2,-2, M,-5,-4,-4,-3,-3, 0,-2, 0, 0,10,-4}, /* Z */ { 0, 1,-5, 2, 3,-5, 0, 2,-2, 0, 0,-2,-1, 1,_M, 0, 3, 0, 0, 0, 0,-2,-6, 0,-4, 4} }; /* */ 25 #include <stdio.h> #include <ctype.h> #define MAXJMP 16 /* max jumps in a diag */ #define MAXGAP 24 /* don't continue to penalize gaps larger than this */ 30 #define JMPS 1024 /* max jmps in an path */ #define MX 4 /* save if there's at least MX-1 bases since last jmp */ #define DMAT 3 /* value of matching bases */ #define DMIS 0 /* penalty for mismatched bases */ 54 WO 2004/094476 PCT/US2004/011793 declinee DINSO 8 /* penalty for a gap */ #define DINS1 1 /* penalty per base */ #define PINSO 8 /* penalty for a gap */ #define PINS 1 4 /* penalty per residue */ 5 structjmp { short n[MAXJMP]; /* size ofjmp (neg for dely) */ unsigned short x[MAXJMP]; /* base no. ofjmp in seq x */ ; /* limits seq to 2^16 -1 */ 10 struct diag { int score; /* score at last jmp */ long offset; /* offset of prey block */ short ijmp; /* current jmp index */ 15 struct jmp jp; /* list of jmps */ 1; struct path { int spc; /* number of leading spaces */ 0 short n[JMPS]; /* size of jmp (gap) */ hit x[JMPS]; /* loc of jmp (last elem before gap) */ 1; char *ofile; /* output file name */ 5 char *namex[2]; /* seq names: getseqs() */ char *prog; /* prog name for err msgs */ char *seqx[2]; /* seqs: getseqs() */ int dmax; /* best diag: nw() */ int dmax0; /* final diag */ 0 int dna; /* set if dna: main() */ int endgaps; /* set if penalizing end gaps */ int gapx, gapy; /* total gaps in seqs */ int len0, lenl; /* seq lens */ int ngapx, ngapy; /* total size of gaps */ 55 WO 2004/094476 PCT/US2004/011793 int smax; /* max score: nw() */ int *xbm; /* bitmap for matching */ long offset; /* current offset in jmp file */ struct diag *dx; /* holds diagonals */ 5 struct path pp[2]; /* holds path for seqs */ char *calloc(), *malloc(), *index(), *strcpy(); char *getseq(), *gcalloc(); 10 /* Needleman-Wunsch alignment program * * usage: progs filel file2 * where filel and file2 are two dna or two protein sequences. * The sequences can be in upper- or lower-case an may contain ambiguity 15 * Any lines beginning with ';', '>' or '<' are ignored * Max file length is 65535 (limited by unsigned short x in the jmp struct) * A sequence with 1/3 or more of its elements ACGTU is assumed to be DNA * Output is in the file "align.out" * 20 * The program may create a tmp file in /tmp to hold info about traceback. * Original version developed under BSD 4.3 on a vax 8650 */ #include "nw.h" #include "day.h" 25 static _dbval[26] = { 1,14,2,13,0,0,4,11,0,0,12,0,3,15,0,0,0,5,6,8,8,7,9,0,10,0 }; 30 static _pbval[26] = { 1, 21(1<<('D'-'A'))I(1<<('N'-'A')), 4, 8, 16, 32, 64, 128, 256, OxFFFFFFF, 1<<10, 1<<11, 1<<12, 1<<13, 1<<14, 1<<15, 1<<16, 1<<17, 1<<18, 1<<19, 1<<20, 1<<21, 1<<22, 1<<23, 1<<24, 1<<251(1<<('E'-'A'))I(1<<('Q'-'A')) 56 WO 2004/094476 PCT/US2004/011793 }.; main(ac, av) main int ac; 5 char *av[]; { prog = av[0]; if (ac != 3) { fprintf(stderr,"usage: %s filel file2n", prog); 10 fprintf(stderr,"where filel and file2 are two dna or two protein sequences.n"); fprintf(stderr,"The sequences can be in upper- or lower-casen"); fprintf(stderr,"Any lines beginning with ';' or '<' are ignoredn"); fprintf(stderr,"Output is in the file "align.out"'n"); exit(1); 15 } namex[O] = av[1]; namex[1] = av[2]; seqx[0] = getseq(namex[0], &lenO); seqx[1] = getseq(namex[1], &lenl); 20 xbm = (dna) dbval : _pbval; endgaps = 0; /* 1 to penalize endgaps */ ofile = "align.out"; /* output file */ 25 nw(); /* fill in the matrix, get the possible jmps */ readjmps(); /* get the actualjmps */ print(); /* print stats, alignment */ cleanup(0); /* unlink any tmp files */ 30 } /* do the alignment, return best score: main() 57 WO 2004/094476 PCT/US2004/011793 * dna: values in Fitch and Smith, PNAS, 80, 1382-1386, 1983 * pro: PAM 250 values * When scores are equal, we prefer mismatches to any gap, prefer * a new gap to extending an ongoing gap, and prefer a gap in seqx 5 *to a gap in seq y. */ nw() nw { char *px, *py; /* seqs and ptrs */ 10 int *ndely, *dely; /* keep track of dely */ int ndelx, delx; /* keep track of delx */ int *tmp; /* for swapping row0, rowl */ int mis; /* score for each type */ int ins0, ins 1; /* insertion penalties */ 15 register id; /* diagonal index */ register ij; /* jmp index */ register *col0, *coll; /* score for curr, last row */ register xx, yy; /* index into seqs */ 20 dx = (struct diag *)gcalloc("to get diags", lenO+ienl+1, sizeof(struct diag)); ndely = (int *)g calloc("to get ndely", lenl+1, sizeof(int)); dely = (int *)gcalloc("to get dely", lenl+1, sizeof(int)); colO = (int *)gcalloc("to get colO", lenl+1, sizeof(int)); 25 coll = (int *)g calloc("to get coll", lenl+1, sizeof(int)); ins0 = (dna) DINSO : PINSO; insI = (dna) DINS1 : PINS1; smax = -10000; 30 if (endgaps) { for (col0[0]= dely[0] = -ins0, yy = 1; yy <= lenl; yy++) { col0[yy] = dely[yy] = col0[yy-1] - insl; ndely[yy] = yy; } 58 WO 2004/094476 PCT/US2004/011793 colO[0] = 0; /* Waterman Bull Math Biol 84 */ } else for (yy = 1; yy <= lenI; yy++) 5 dely[yy] = -ins0; /* fill in match matrix */ for (px = seqx[0], xx = 1; xx <= lenO; px++, xx++) { 10 /* initialize first entry in col */ if (endgaps) { if (xx == 1) coll [0] = delx = -(ins0+insl); 15 else coll[0] = delx = colO[O] - ins 1; ndelx = xx; } else { 20 coll[0] = 0; delx = -insO; ndelx = 0; ... nw 25 for (py = seqx[1], yy = 1; yy <= lenl; py++, yy++) { mis = col0[yy-1]; if (dna) mis += (xbm[*px-'A']&xbm[*py-'A']) DMAT: DMIS; else 30 mis += _day[*px-'A'][*py-'A']; /* update penalty for del in x seq; * favor new del over ongong del * ignore MAXGAP if weighting endgaps 59 WO 2004/094476 PCT/US2004/011793 */ if (endgaps 11 ndely[yy] < MAXGAP) { if (col0[yy] - ins0 >= dely[yy]) { dely[yy] = colO[yy] - (ins0+insl ); 5 ndely[yy] = 1; } else { dely[yy] -= ins1; ndely[yy]++; } 10 }else{ if (col0[yy] - (ins0+insl) >= dely[yy]) { dely[yy] = col0[yy] - (ins0+insl ); ndely[yy] = 1; } else 15 ndely[yy]++; } /* update penalty for del in y seq; * favor new del over ongong del 20 */ if (endgaps II ndelx < MAXGAP) { if (coll[yy-1] - ins0 >= delx) { delx = coll [yy-1] - (ins0+insl); ndelx = 1; 25 } else { delx -= insI; ndelx++; } } else { 30 if (coll[yy-1] - (ins0+insl) >= delx) { delx = coll[yy-1] - (ins0+insl); ndelx = 1; } else ndelx++; 60 WO 2004/094476 PCT/US2004/011793 } /* pick the maximum score; we're favoring * mis over any del and delx over dely 5 / ... nw id= xx- yy + len1- 1; if (mis >= delx && mis >= dely[yy]) coll [yy] = mis; 10 else if (delx >= dely[yy]) { coll[yy] = delx; ij = dx[id].ijmp; if (dx[id].jp.n[0] && (!dna | (ndelx >= MAXJMP && xx > dx[id].jp.x[ij]+MX) II mis > dx[id].score+DINSO)) { 15 dx[id].ijmp++; if (++ij >= MAXJMP) { writejmps(id); ij = dx[id].ijmp =0; dx[id].offset = offset; 20 offset += sizeof(struct jmp) + sizeof(offset); } dx[id].jp.n[ij] = ndelx; dx[id].jp.x[ij] = xx; 25 dx[id].score = delx; } else { col [yy] = dely[yy]; ij = dx[id].ijmp; 30 if (dx[id].jp.n[0] && (!dna || (ndely[yy] >= MAXJMP && xx > dx[id].jp.x[ij]+MX) I mis > dx[id].score+DINSO)) { dx[id].ijmp++; if (++ij >= MAXJMP) { writejmps(id); 61 WO 2004/094476 PCT/US2004/011793 ij = dx[id].ijmp = 0; dx[id].offset = offset; offset += sizeof(struct jmp) + sizeof(offset); } 5 } dx[id].jp.n[ij] = -ndely[yy]; dx[id].jp.x[ij] = xx; dx[id].score = dely[yy]; } 10 if (xx == lenO && yy < lenl) { /* last col */ if (endgaps) col l[yy] -= insO+insl*(lenl-yy); 15 if (coll[yy] > smax) { smax = coll[yy]; dmax = id; } } 20 } if (endgaps && xx < lenO) col [yy- 1] -= insO+ins 1 * (lenO-xx); if (coll [yy-1] > smax) { smax = coll [yy- 1]; 25 dmax = id; } trmp = colO; colO = coll; coll = tmp; } (void) free((char *)ndely); 30 (void) free((char *)dely); (void) free((char *)colO); (void) free((char *)coll); } /* * 62 WO 2004/094476 PCT/US2004/011793 * print() -- only routine visible outside this module * * static: * getmat() -- trace back best path, count matches: print() 5 * pralign( ) -- print alignment of described in array p[]: print() * dumpblock() -- dump a block of lines with numbers, stars: pralign() * nums() -- put out a number line: dumpblock() * putline( ) -- put out a line (name, [num], seq, [num]): dumpblock() * stars() - -put a line of stars: dumpblock() 10 * stripname() -- strip any path and prefix from a seqname */ #include "nw.h" 15 #define SPC 3 #define P_LINE 256 /* maximum output line */ #define P_SPC 3 /* space between name or num and seq */ extern _day[26] [26]; 20 int olen; /* set output line length */ FILE *fx; /* output file */ print() print { 25 int lx, ly, firstgap, lastgap; /* overlap */ if ((fx = fopen(ofile, "w")) == 0) { fprintf(stderr,"%s: can't write %sn", prog, ofile); cleanup(l); 30 } fprintf(fx, "<first sequence: %s (length = %d)n", namex[0], lenO); fprintf(fx, "<second sequence: %s (length = %d)n", namex[1], lenl); olen = 60; lx = lenO; 63 WO 2004/094476 PCT/US2004/011793 ly = lenl; firstgap = lastgap = 0; if (dmax < lenI - 1) { /* leading gap in x */ pp[O].spc = firstgap = lenI - dmax - 1; 5 ly -= pp[0].spc; } else if (dmax > lenI - 1) { /* leading gap in y */ pp[1].spc = firstgap = dmax - (lenl - 1); lx -= pp[1].spc; 10 } if (dmaxO < lenO - 1) { /* trailing gap in x */ lastgap = lenO - dmaxO -1; lx -= lastgap; } 15 else if (dmax0 > lenO - 1) { /* trailing gap in y */ lastgap = dmax0 - (lenO - 1); ly -= lastgap; } getmat(lx, ly, firstgap, lastgap); 20 pralign(o); } /* * trace back the best path, count matches */ 25 static getmat(lx, ly, firstgap, lastgap) getmat int lx, ly; /* "core" (minus endgaps) */ int firstgap, lastgap; /* leading trailing overlap */ { 30 int nm, iO, il, sizO, sizl; char outx[32]; double pct; register nO, nl; register char *pO, *pl; 64 WO 2004/094476 PCT/US2004/011793 /* get total matches, score */ iO = il = siz0 = sizl = 0; 5 p0 = seqx[0] + pp[1].spc; pl = seqx[1] + pp[0].spc; n0 = pp[1].spc + 1; nI = pp[0].spc + 1; 10 nm = 0; while ( *p0 && *pl ) { if (siz0) { pl++; nl++; 15 siz0--; } else if (sizl) { pO++; nO++; 20 sizl--; } else { if (xbm[*p0-'A']&xbm[*p1l-'A']) nm++; 25 if (nO++ == pp[O].x[iO]) sizO = pp[0].n[iO++]; if (nl++ == pp[1].x[il]) sizI = pp[1].n[il++]; pO++; 30 pl++; /* pct homology: 65 WO 2004/094476 PCT/US2004/011793 * if penalizing endgaps, base is the shorter seq * else, knock off overhangs and take shorter core */ if (endgaps) 5 lx = (len0 < len1) len0 : lenl; else lx = (lx < ly) lx : ly; pct = 100.*(double)nm/(double)lx; fprintf(fx, "n"); 10 fprintf(fx, "<%d match%s in an overlap of %d: %.2f percent similarityn", nm, (nm == 1) "" : "es", lx, pct); fprintf(fx, "<gaps in first sequence: %d", gapx); ...getmat if (gapx) { 15 (void) sprintf(outx, " (%d %s%s)", ngapx, (dna) "base":"residue", (ngapx == 1) "".... :"s"); fprintf(fx,"%s", outx); fprintf(fx, ", gaps in second sequence: %d", gapy); 20 if (gapy) { (void) sprintf(outx, " (%d %s%s)", ngapy, (dna) "base":"residue", (ngapy == 1) "":"s"); fprintf(fx,"%s", outx); } 25 if(dna) fprintf(fx, "n<score: %d (match = %d, mismatch = %d, gap penalty = %d + %d per base)n", smax, DMAT, DMIS, DINSO, DINS 1); 30 else fprintf(fx, "n<score: %d (Dayhoff PAM 250 matrix, gap penalty = %d + %d per residue)n", smax, PINSO, PINS1); 66 WO 2004/094476 PCT/US2004/011793 if (endgaps) fprintf(fx, "<endgaps penalized, left endgap: %d %s%s, right endgap: %d %s%sn", firstgap, (dna) "base" : "residue", (firstgap == 1) "" : "s", 5 lastgap, (dna) "base" : "residue", (lastgap == 1) "": "s"); else fprintf(fx, "<endgaps not penalizedn"); } 10 static nm; /* matches in core -- for checking */ static Imax; /* lengths of stripped file names */ static ij [2]; /* jmp index for a path */ static nc[2]; /* number at start of current line */ static ni[2]; /* current elem number -- for gapping */ 15 static siz[2]; static char *ps[2]; /* ptr to current element */ static char *po[2]; /* ptr to next output char slot */ static char out[2][PLINE]; /* output line */ static char star[PLINE]; /* set by stars() */ 20 /* * print alignment of described in struct path pp[] */ static 25 pr_align() pr align { int nn; /* char count */ int more; register i; 30 for (i = 0, Imax = 0; i < 2; i++) { nn = stripname(namex[i]); if (nn > Imax) Imax = nn; 67 WO 2004/094476 PCT/US2004/011793 nc[i] = 1; ni[i) = 1; siz[i] = ij[i] = 0; 5 ps[i] = seqx[i]; po[i] = out[i]; } for (nn = nm = 0, more = 1; more;) { ...pr align for (i = more = 0; i < 2; i++) { /* 10 * do we have more of this sequence */ if (!*psr[i]) continue; 15 more++; if (pp[i].spc) { /* leading space */ *po[i]++ =' '; pp[i].spc--; else if (siz[i]) { /* in a gap */ *po[i]++= '-'; siz[i]--; } ,5 else { /* we're putting a seq element */ *po[i] = *ps[i]; if (islower(*ps[i])) *ps[i] = toupper(*ps[i]); 0 po[i]++; ps[i]++; /* * are we at next gap for this seq 68 WO 2004/094476 PCT/US2004/011793 */ if (ni[i] == pp[i].x[ij[i]]) { /* * we need to merge all gaps 5 * at this location */ siz[i] = pp[i].n[ij[i]++]; while (ni[i] == pp[i].x[ij[i]]) siz[i] += pp[i].nl[ij [i]++]; 10 } ni[ij++; } } if (++nn == olen | !more && nn) { 15 dumpblock(); for (i= 0; i < 2; i++) po[i] = out[i]; nn = 0; 20 } /* * dump a block of lines, including numbers, stars: pr align() 25 */ static dumpblock() dumpblock { register i; 30 for (i = 0; i < 2; i++) *po[i]-- =' |