EP1309685A2 - Zusammensetzungen und verfahren für die diagnose und behandlung von angiogenese-assoziierten erkrankungen - Google Patents

Zusammensetzungen und verfahren für die diagnose und behandlung von angiogenese-assoziierten erkrankungen

Info

Publication number
EP1309685A2
EP1309685A2 EP01951036A EP01951036A EP1309685A2 EP 1309685 A2 EP1309685 A2 EP 1309685A2 EP 01951036 A EP01951036 A EP 01951036A EP 01951036 A EP01951036 A EP 01951036A EP 1309685 A2 EP1309685 A2 EP 1309685A2
Authority
EP
European Patent Office
Prior art keywords
seq
sequence
acid sequence
amino acid
polypeptide
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Withdrawn
Application number
EP01951036A
Other languages
English (en)
French (fr)
Inventor
Kevin P. Baker
Napoleone Ferrara
Hanspeter Gerber
Mary E. Gerritsen
Audrey Goddard
Paul J. Godowski
Austin L. Gurney
Kenneth J. Hillan
Scot A. Marsters
James Pan
Nicholas F. Paoni
Jean-Philippe F. Stephan
Colin K. Watanabe
P. Mickey Williams
William I. Wood
Weilan Ye
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Genentech Inc
Original Assignee
Genentech Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from PCT/US2000/020710 external-priority patent/WO2001009327A2/en
Priority claimed from US09/643,657 external-priority patent/US6642024B1/en
Priority claimed from PCT/US2000/023522 external-priority patent/WO2001016319A2/en
Priority claimed from PCT/US2000/023328 external-priority patent/WO2001016318A2/en
Priority claimed from PCT/US2000/030952 external-priority patent/WO2001049715A2/en
Priority claimed from PCT/US2000/030873 external-priority patent/WO2001040465A2/en
Priority claimed from PCT/US2000/032678 external-priority patent/WO2001040466A2/en
Priority claimed from PCT/US2000/034956 external-priority patent/WO2001046420A2/en
Priority claimed from US09/747,259 external-priority patent/US6569645B2/en
Priority claimed from US09/767,609 external-priority patent/US20020042367A1/en
Priority claimed from PCT/US2001/006520 external-priority patent/WO2001068848A2/en
Priority claimed from PCT/US2001/006666 external-priority patent/WO2001066740A2/en
Priority claimed from US09/816,744 external-priority patent/US6579520B2/en
Priority claimed from US09/828,366 external-priority patent/US20020010137A1/en
Priority claimed from US09/854,280 external-priority patent/US7115398B2/en
Priority claimed from PCT/US2001/017092 external-priority patent/WO2001092331A2/en
Priority claimed from US09/866,028 external-priority patent/US6642360B2/en
Priority claimed from US09/866,034 external-priority patent/US20030170864A1/en
Priority claimed from US09/870,574 external-priority patent/US6551799B2/en
Priority claimed from PCT/US2001/017443 external-priority patent/WO2002016611A2/en
Priority claimed from PCT/US2001/017800 external-priority patent/WO2001093983A1/en
Priority claimed from PCT/US2001/019692 external-priority patent/WO2002000690A2/en
Application filed by Genentech Inc filed Critical Genentech Inc
Publication of EP1309685A2 publication Critical patent/EP1309685A2/de
Withdrawn legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/475Growth factors; Growth regulators
    • C07K14/515Angiogenesic factors; Angiogenin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/02Drugs for dermatological disorders for treating wounds, ulcers, burns, scars, keloids, or the like
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/04Inotropic agents, i.e. stimulants of cardiac contraction; Drugs for heart failure
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/14Vasoprotectives; Antihaemorrhoidals; Drugs for varicose therapy; Capillary stabilisers

Definitions

  • the present invention relates to compositions and methods useful for the modulation (e.g., promotion or inhibition) of angiogenesis and or cardiovascularization in mammals in need of such biological effect.
  • the present invention further relates to the diagnosis and treatment of disorders involving angiogenesis (e.g. , cardiovascular as well as oncological disorders).
  • Angiogenesis defined as the growth or sprouting of new blood vessels from existing vessels, is a complex process that primarily occurs during embryonic development. Under normal physiological conditions in adults, angiogenesis takes place only in very restricted situations such as hair growth and wounding healing (Auerbach,
  • Unregulated angiogenesis has gradually been recognized to be responsible for a wide range of disorders, including, but not limited to cardiovascular disease, cancer, rheumatoid arthritis, psoriasis and diabetic retinopathy (Folkman, 1995, Nat Med 1(1):27-31; Isner, 1999, Circulation 99(13): 1653-5; Koch, 1998,ArthritisRheum4l(6) 951-62; Wahb ⁇ l999, Rheumatology (Oxford) 38(2): ⁇
  • CHF congestive heart failure
  • At least four major compensatory mechanisms are activated in the setting of heart failure to boost cardiac output, including peripheral vasoconstriction, increased heart rate, increased cardiac contractility, and increased plasma volume. These effects are mediated primarily by the sympathetic nervous system and the renin-angiotensin system. See, Eichhorn, American Journal of Medicine, 104: 163-169 (1998). Increased output from the sympathetic nervous system increases vascular tone, heart rate, and contractility.
  • Angiotensin II elevates blood pressure by 1) directly stimulating vascular smooth muscle contraction, 2) promoting plasma volume expansion by stimulating aldosterone and antidiuretic hormone secretion, 3) stimulating sympathetic-mediated vascular tone, and 4) catalyzing the degradation of bradykinin, which has vasodilatory and natriuretic activity. See, review by Brown and Vaughan. Circulation.97: 1411-1420(1998). As noted below, angiotensin II may also have directly deleterious effects on the heart by promoting myocyte necrosis (impairing systolic function) and intracardiac fibrosis (impairing diastolic and in some cases systolic function). See, Weber, Circulation. 96: 4065-4082 (1998).
  • cardiac hypertrophy an enlargement ofthe heart that is activated by both mechanical and hormonal stimuli and enables the heart to adapt to demands for increased cardiac output. Morgan and Baker, Circulation. 83: 13-25 (1991).
  • This hypertrophic response is frequently associated with a variety of distinct pathological conditions such as hypertension, aortic stenosis, myocardial infarction, cardiomyopathy, valvular regurgitation, and intracardiac shunt, all of which result in chronic hemodynamic overload.
  • Hypertrophy is generally defined as an increase in size of an organ or structure independent of natural growth that does not involve tumor formation. Hypertrophy of the heart is due either to an increase in the mass of the individual cells (myocytes), or to an increase in the number of cells making up the tissue (hyperplasia), or both. While the enlargement of an embryonic heart is largely dependent on an increase in myocyte number (which continues until shortly after birth), post-natal cardiac myocytes lose their proliferative capacity. Further growth occurs through hypertrophy ofthe individual cells.
  • Adult myocyte hypertrophy is initially beneficial as a short term response to impaired cardiac function by permitting a decrease in the load on individual muscle fibers. With severe, long-standing overload, however, the hypertrophied cells begin to deteriorate and die.
  • non-myocytes On a cellular level, the heart is composed of myocytes and surrounding support cells, generically called non-myocytes. While non-myocytes are primarily fibroblast/mesenchyrnal cells, they also include endothelial and smooth muscle cells. Indeed, although myocytes make up most ofthe adult myocardial mass, they represent only about 30% ofthe total cell numbers present in heart. In response to hormonal, physiological, hemodynamic, and pathological stimuli, adult ventricular muscle cells can adapt to increased workloads through the activation of a hypertrophic process. This response is characterized by an increase in myocyte cell size and contractile protein content of individual cardiac muscle cells, without concomitant cell division and activation of embryonic genes, including the gene for atrial natriuretic peptide (ANP).
  • APN atrial natriuretic peptide
  • non-myocyte supporting cells may additionally be involved in the development of cardiac hypertrophy, and various non-myocyte derived hypertrophic factors, such as, leukocyte inhibitory factor (LIF) and endothelin, have been identified.
  • LIF leukocyte inhibitory factor
  • Metcalf Growth Factors.
  • cardiac hypertrophy varies depending on the underlying cardiac disease.
  • Catecholamines, adrenocorticosteroids, angiotensin, prostaglandins, LIF, endothelin (including endothelin-1, -2, and -3 and big endothelin), and CT-1 are among the factors identified as potential mediators of hypertrophy.
  • beta-adrenergic receptor blocking drugs e.g., propranolol, timolol, tertalolol, carteolol, nadolol, betaxolol, penbutolol, acetobutolol, atenolol, metoprolol, carvedilol, etc.
  • verapamil have been used extensively in the treatment of hypertrophic cardiomyopathy.
  • the beneficial effects of beta-blockers on symptoms (e.g., chest pain) and exercise tolerance are largely due to a decrease in the heart rate with a consequentprolongation of diastole and increased passive ventricular filling.
  • Nifedipine and diltiazem have also been used occasionally in the treatment of hypertrophic cardiomyopathy. Lorell et al, Circulation. 65: 499-507 (1982); Betocchi et al, Am. J. Cardiol.. 78: 451-457 (1996).
  • nifedipine may be harmful, especially in patients with outflow obstruction.
  • Disopyramide has been used to relieve symptoms by virtue of its negative inotropic properties. Pollick, N. Engl. J. Med..307: 997-999 (1982). In many patients, however, the initial benefits decrease with time. Wigle et al, Circulation. 92: 1680-1692 (1995).
  • Antihypertensive drug therapy has been reported to have beneficial effects on cardiac hypertrophy associated with elevated blood pressure.
  • drugs used in antihypertensive therapy are calcium antagonists, e.g. , nitrendipine; adrenergic receptor blocking agents, e.g., those listed above; angiotensin converting enzyme (ACE) inhibitors such as quinapril, captopril, enalapril, ramipril, benazepril, fosinopril, and lisinopril; diuretics, e.g., chlorothiazide, hydrochloro iazide,hydroflumemazide,memylchlot azide,benzthiazide, dichlorphenamide, acetazolamide, and indapamide; and calcium channel blockers, e.g., diltiazem, nifedipine, verapamil, and nicardipine.
  • ACE angio
  • CHF CHF. Another treatment modality is heart transplantation, but this is limited by the availability of donor hearts.
  • Endothelin is a vasoconstricting peptide comprising 21 amino acids, isolated from swine arterial endothelial culture supernatant and structurally determined. Yanagisawa et al, Nature. 332: 411-415 (1988). Endothelin was later found to exhibit various actions, and endothelin antibodies as endothelin antagonists have proven effective in the treatment of myocardial infarction, renal failure, and other diseases. Since endothelin is present in live bodies and exhibits vasoconstricting action, it is expected to be an endogenous factor involved in the regulation of the circulatory system, and may be associated with hypertension, cardiovascular diseases such as myocardial infarction, and renal diseases such as acute renal failure.
  • Endothelin antagonists are described, for example, in U.S. Pat. No. 5,773,414; JP Pat. Publ. 3130299/1991, EP 457,195; EP 460,679; and EP 552,489.
  • a new endothelin B receptor for identifying endothelin receptor antagonists is described in U.S. Pat. No. 5,773,223.
  • ACE angiotensin-converting enzyme
  • ACE inhibitors have functional class III heart failure.
  • ACE inhibitors An alternative to ACE inhibitors is represented by specific ATI receptor antagonists.
  • Clinical studies are planned to compare the efficacy of these two modalities in the treatment of cardiovascular and renal disease.
  • animal model data suggests that the ACE/Ang II pathway, while clearly involved in cardiac hypertrophy, is not the only, or even the primary pathway active in this role.
  • Mouse genetic "knockout" models have been made to test individual components ofthe pathway. In one such model, the primary cardiac receptor for Ang II, AT sub IA, has been genetically deleted; these mice do not develop hypertrophy when Ang II is given experimentally (confirming the basic success ofthe model in eliminating hypertrophy secondary to Ang II).
  • thrombolytic agents e.g., streptokinase, urokinase, and in particular tissue plasminogen activator (t-PA) have significantly increased the survival of patients who suffered myocardial infarction.
  • t-PA tissue plasminogen activator
  • t-PA may also be administered as a single bolus, although due to its relatively short half-life, it is better suited for infusion therapy. Tebbe et al, Am. J. Cardiol., 64: 448-453 (1989).
  • TNK t-PA a T103N, NI 17Q, KHRR(296-299)AAAA t-PA variant, Keyt et al, Proc. Natl. Acad. Sci. USA.91: 3670-3674 (1994)
  • TNK t-PA a T103N, NI 17Q, KHRR(296-299)AAAA t-PA variant, Keyt et al, Proc. Natl. Acad. Sci. USA.91: 3670-3674 (1994)
  • the long-term prognosis of patient survival depends greatly on the post-infarction monitoring and treatment ofthe patients, which should include monitoring and treatment of cardiac hypertrophy.
  • FGF basic and acidic fibroblast growth factors
  • PD-ECGF latelet-derived endothelial cell growth factor
  • VEGF vascular endothelial growth factor
  • hVEGF human VEGF
  • hVEGF-related proteins Several additional cDNAs were identified in human cDNA libraries that encode 121-, 189-, and 206-amino acid isoforms of hVEGF (also collectively referred to as hVEGF-related proteins).
  • the 121 -amino acid protein differs from hVEGF by virtue ofthe deletion ofthe 44 amino acids between residues 116 and 159 in hVEGF.
  • the 189-amino acid protein differs from hVEGF by virtue of the insertion of 24 amino acids at residue 116 in hVEGF, and apparently is identical to human vascular permeability factor (hVPF).
  • the 206-amino acid protein differs from hVEGF by virtue of an insertion of 41 amino acids at residue 116 in hVEGF. Houck et al. , Mol. Endocrin.. 5: 1806 (1991); Ferrara et al, J. Cell. Biochem., 47: 211 (1991); Ferrara et al, Endocrine Reviews. 13: 18 (1992); Keck et al, Science.246: 1309 (1989); Connolly et al, J. Biol.
  • angiogenesis which involves the formation of new blood vessels from preexisting endothelium, is implicated in the patliogenesis of a variety of disorders. These include solid tumors and metastasis, atherosclerosis, retrolental fibroplasia, hemangiomas, chronic inflammation, intraocular neovascular syndromes such as proliferative retinopathies, e.g. , diabetic retinopathy, age-related macular degeneration (AMD), neovascular glaucoma, immune rejection of transplanted corneal tissue and other tissues, rheumatoid arthritis, and psoriasis. Folkmane/ ⁇ /., J. Biol. Chem., 267: 10931-10934 (1992); Klagsbrune/fl/. Annu. Rev. Physiol., 53: 217-
  • angiogenesis appears to be crucial for the transition from hyperplasia to neoplasia, and for providing nourishment for the growth and metastasis ofthe tumor.
  • Folkman et al. Nature, 339: 58 (1989).
  • Theneovascularization allows the tumor cells to acquire a growth advantage and proliferative autonomy compared to the normal cells.
  • a tumor usually begins as a single aberrant cell which can proliferate only to a size of a few cubic millimeters due to the distance from available capillary beds, and it can stay 'dormant' without further growth and dissemination for a long period of time.
  • VEGF has been shown to be a key mediator of neovascularization associated with tumors and intraocular disorders.
  • Ferrara et al Endocr. Rev., supra.
  • the VEGF mRNA is overexpressed by the majority of human tumors examined. Berkman et al, J. Clin. Invest.. 91: 153-159 (1993); Brown et al, Human Pathol.. 26: 86-91 (1995); Brown etal, Cancer Res., 53: 4727-4735 (1993); Matterne ⁇ /., Brit J. Cancer, 73: 931-934 (1996); Dvorak et al, Am. J. Pathol., 146: 1029-1039 (1995).
  • VEGF vascular endothelial growth factor
  • concentration levels of VEGF in eye fluids are highly correlated to the presence of active proliferation of blood vessels in patients with diabetic and other ischemia-related retinopathies.
  • Aiello et al N. Engl. J. Med.. 331: 1480-1487 (1994).
  • recent studies have demonstrated the localization of VEGF in choroidal neovascular membranes in patients affected by AMD. Lopez et al, Invest Ophthalmol. Vis. Sci..37: 855-868 (1996).
  • Anti- VEGF neutralizing antibodies suppress the growth of a variety of human tumor cell lines in nude mice
  • CTGF connective tissue growth factor
  • TGF- ⁇ transforming growth factor beta
  • IGFBPs insulin-like growth factor binding proteins
  • IGF insulin-like growth factor
  • vascular endothelial cell growth and angiogenesis in many diseases and disorders, it is desirable to have a means of reducing or inhibiting one or more of the biological effects causing these processes. It is also desirable to have a means of assaying for the presence of pathogenic polypeptides in normal and diseased conditions, and especially cancer. Further, in a specific aspect, as there is no generally applicable therapy for the treatment of cardiac hypertrophy, the identification of factors that can prevent or reduce cardiac myocyte hypertrophy is of primary importance in the development of new therapeutic strategies to inhibit pathophysiological cardiac growth. While there are several treatment modalities for various cardiovascular and oncologic disorders, there is still a need for additional therapeutic approaches.
  • the present invention provides compositions and methods for modulating (e.g. , promoting or inhibiting) angiogenesis and/or cardiovascularization in mammals.
  • the present invention is based on the identification of compounds ( . e. , proteins) that test positive in various cardiovascular assays that test modulation (e.g. , promotion or inhibition) of certain biological activities.
  • the compounds are believed to be useful drugs and/or drug components for the diagnosis and/or treatment (including prevention and amelioration) of disorders where such effects are desired, such as the promotion or inhibition of angiogenesis, inhibition or stimulation of vascular endothelial cell growth, stimulation of growth or proliferation of vascular endothelial cells, inhibition of tumor growth, inhibition of angiogenesis-dependent tissue growth, stimulation of angiogenesis-dependent tissue growth, inhibition of cardiac hypertrophy and stimulation of cardiac hypertrophy, e.g., for the treatment of congestive heart failure.
  • compositions and methods ofthe invention provide for the diagnostic monitoring ofpatients undergoing clinical evaluation for the treatment of angiogenesis-related disorders, for monitoring the efficacy of compounds in clinical trials and for identifying subjects who may be predisposed to such angiogenic-related disorders.
  • the present invention provides a composition comprising a PRO polypeptide, an agonist or antagonist thereof, or an anti-PRO antibody in admixture with a pharmaceutically acceptable carrier.
  • the composition comprises a therapeutically effective amount ofthe polypeptide, agonist, antagonist or antibody.
  • the composition comprises a further active ingredient, namely, a cardiovascular, endothelial or angiogenic agent or an angiostatic agent, preferably an angiogenic or angiostatic agent.
  • the composition is sterile.
  • the PRO polypeptide, agonist, antagonist or antibody may be administered in the form of a liquid pharmaceutical formulation, which may be preserved to achieve extended storage stability.
  • Preserved liquid pharmaceutical formulations might contain multiple doses of PRO polypeptide, agonist, antagonist or antibody, and might, therefore, be suitable for repeated use.
  • the composition comprises an antibody
  • the antibody is a monoclonal antibody, an antibody fragment, a humanized antibody, or a single-chain antibody.
  • the present invention provides a method for preparing such a composition useful for the treatment of a cardiovascular, endothelial or angiogenic disorder comprising admixing a therapeutically effective amount of a PRO polypeptide, agonist, antagonist or antibody with a pharmaceutically acceptable carrier.
  • the present invention provides an article of manufacture comprising:
  • composition of matter comprising a PRO polypeptide or agonist or antagonist thereof;
  • composition may comprise a therapeutically effective amount of the PRO polypeptide or the agonist or antagonist thereof.
  • present invention provides a method for identifying an agonist of a PRO polypeptide comprising:
  • the present invention provides a method for identifying an agonist of a PRO polypeptide comprising:
  • the invention provides a method for identifying a compound that inhibits the activity of a PRO polypeptide comprising contacting a test compound with a PRO polypeptide under conditions and for a time sufficient to allow the test compound and polypeptide to interact and determining whether the activity ofthe PRO polypeptide is inhibited.
  • either the test compound or the PRO polypeptide is immobilized on a solid support.
  • the non- immobilized component carries a detectable label. In a preferred aspect, this method comprises the steps of:
  • test compound (b) determining the induction of said cellular response to determine if the test compound is an effective antagonist.
  • this process comprises the steps of:
  • the invention provides a method for identifying a compound that inhibits the expression of a PRO polypeptide in cells that normally expresses the polypeptide, wherein the method comprises contacting the cells with a test compound and determining whether the expression of the PRO polypeptide is inhibited. In a preferred aspect, this method comprises the steps of:
  • the invention provides a compound that inhibits the expression of a PRO polypeptide, such as a compound that is identified by the methods set forth above.
  • Another aspect ofthe present invention is directed to an agonist or an antagonist of a PRO polypeptide which may optionally be identified by the methods described above.
  • the invention provides an isolated antibody that binds a PRO polypeptide.
  • the antibody is a monoclonal antibody, which preferably has non-human complementarity-determining-region (CDR) residues and human framework-region (FR) residues.
  • CDR non-human complementarity-determining-region
  • FR human framework-region
  • the antibody may be labeled and may be immobilized on a solid support.
  • the antibody is an antibody fragment, a single-chain antibody, or a humanized antibody.
  • the antibody specifically binds to the polypeptide.
  • the present invention provides a method for diagnosing a disease or susceptibility to a disease which is related to a mutation in a PRO polypeptide-encoding nucleic acid sequence comprising determining the presence or absence of said mutation in the PRO polypeptide nucleic acid sequence, wherein the presence or absence of said mutation is indicative ofthe presence of said disease or susceptibility to said disease.
  • the invention provides a method of diagnosing a cardiovascular, endothelial or angiogenic disorder in a mammal which comprises analyzing the level o expression of a gene encoding a PRO polypeptide (a) in a test sample of tissue cells obtained from said mammal, and (b) in a control sample of known normal tissue cells ofthe same cell type, wherein a higher or lower expression level in the test sample as compared to the control sample is indicative ofthe presence of a cardiovascular, endothelial or angiogenic disorder in said mammal.
  • the expression of a gene encoding a PRO polypeptide may optionally be accomplished by measuring the level of mRNA or the polypeptide in the test sample as compared to the control sample.
  • the present invention provides a method of diagnosing a cardiovascular, endothelial or angiogenic disorder in a mammal which comprises detecting the presence or absence of a PRO polypeptide in a test sample of tissue cells obtained from said mammal, wherein the presence or absence of said PRO polypeptide in said test sample is indicative of the presence of a cardiovascular, endothelial or angiogenic disorder in said mammal.
  • the invention provides a method of diagnosing a cardiovascular, endothelial or angiogenic disorder in a mammal comprising (a) contacting an anti-PRO antibody with a test sample of tissue cells obtained from the mammal, and (b) detecting the formation of a complex between the antibody and the PRO polypeptide in the test sample, wherein the formation of said complex is indicative of the presence of a cardiovascular, endothelial or angiogenic disorder in the mammal.
  • the detection may be qualitative or quantitative, and may be performed in comparison with monitoring the complex formation in a control sample of known normal tissue cells ofthe same cell type.
  • a larger or smaller quantity of complexes formed in the test sample indicates the presence of a cardiovascular, endothelial or angiogenic dysfunction in the mammal from which the test tissue cells were obtained.
  • the antibody preferably carries a detectable label. Complex formation can be monitored, for example, by light microscopy, flow cytometry, fluorimetry, or other techniques known in the art.
  • the test sample is usually obtained from an individual suspected to have a cardiovascular, endothelial or angiogenic disorder.
  • the invention provides a method for determining the presence of a PRO polypeptide in a sample comprising exposing a sample suspected of containing the PRO polypeptide to an anti-PRO antibody and determining binding of said antibody to a component of said sample.
  • the sample comprises a cell suspected of containing the PRO polypeptide and the antibody binds to the cell.
  • the antibody is preferably detectably labeled and/or bound to a solid support.
  • the invention provides a cardiovascular, endothelial or angiogenic disorder diagnostic kit comprising an anti-PRO antibody and a carrier in suitable packaging.
  • kit further comprises instructions for using said antibody to detect the presence of the PRO polypeptide.
  • the carrier is a buffer, for example.
  • the cardiovascular, endothelial or angiogenic disorder is cancer.
  • the present invention provides a method for treating a cardiovascular, endothelial or angiogenic disorder in a mammal comprising administering to the mammal an effective amount of a PRO polypeptide.
  • the disorder is cardiac hypertrophy, trauma such as wounds or bums, or a type of cancer.
  • the mammal is further exposed to angioplasty or a drug that treats cardiovascular, endothelial or angiogenic disorders such as ACE inhibitors or chemotherapeutic agents if the cardiovascular, endothelial or angiogenic disorder is a type of cancer.
  • the mammal is human, preferably one who is at risk of developing cardiac hypertrophy and more preferably has suffered myocardial infarction.
  • the cardiac hypertrophy is characterized by the presence of an elevated level of PGF 2o .
  • the cardiac hypertrophy may be induced by myocardial infarction, wherein preferably the administration ofthe PRO polypeptide is initiated within 48 hours, more preferably within 24 hours, following myocardial infarction.
  • the cardiovascular, endothelial or angiogenic disorder is cardiac hypertrophy and said PRO polypeptide is administered together with a cardiovascular, endothelial or angiogenic agent.
  • the preferred cardiovascular, endothelial or angiogenic agent for this purpose is selected from the group consisting of an antihypertensive drug, an ACE inhibitor, an endothelin receptor antagonist and a thrombolytic agent. If a thrombolytic agent is administered, preferably the PRO polypeptide is administered following administration of such agent. More preferably, the tlirombolytic agent is recombinant human tissue plasminogen activator.
  • the cardiovascular, endothelial or angiogenic disorder is cardiac hypertrophy and the PRO polypeptide is administered following primary angioplasty for the treatment of acute myocardial infarction, preferably wherein the mammal is further exposed to angioplasty or a cardiovascular, endothelial, or angiogenic agent.
  • the cardiovascular, endothelial or angiogenic disorder is a cancer and the PRO polypeptide is administered in combination with a chemotherapeutic agent, a growth inhibitory agent or a cytotoxic agent.
  • the invention provides a method for treating a cardiovascular, endothelial or angiogenic disorder in a mammal comprising administering to the mammal an effective amount of a PRO polypeptide agonist, antagonist or anti-PRO antibody.
  • the cardiovascular, endothelial or angiogenic disorder is cardiac hypertrophy, trauma, a cancer, or age-related macular degeneration. Also preferred is where the mammal is human, and where an effective amount of an angiogenic or angiostatic agent is administered in conjunction with the agonist, antagonist or anti-PRO antibody.
  • the invention provides a method for treating a cardiovascular, endothelial or angiogenic disorder in a mammal that suffers therefrom comprising administering to the mammal a nucleic acid molecule that codes for either (a) a PRO polypeptide, (b) an agonist of a PRO polypeptide or (c) an antagonist of a PRO polypeptide, wherein said agonist or antagonist may be an anti-PRO antibody.
  • the mammal is human.
  • the gene is administered via ex vivo gene therapy.
  • the gene is comprised within a vector, more preferably an adenoviral, adeno-associated viral, lentiviral, or retroviral vector.
  • the invention pro vides a recombinant retroviral particle comprising a retroviral vector consisting essentially of a promoter, nucleic acid encoding (a) a PRO polypeptide, (b) an agonist polypeptide of a PRO polypeptide, or (c) an antagonist polypeptide of a PRO polypeptide, and a signal sequence for cellular secretion of the polypeptide, wherein the retroviral vector is in association with retroviral structural proteins.
  • the signal sequence is from a mammal, such as from a native PRO polypeptide.
  • the invention supplies an ex vivo producer cell comprising a nucleic acid construct that expresses retroviral structural proteins and also comprises a retroviral vector consisting essentially of a promoter, nucleic acid encoding (a) a PRO polypeptide, (b) an agonist polypeptide of a PRO polypeptide or
  • the invention provides a method for inhibiting endothelial cell growth in a mammal comprising administering to the mammal (a) a PRO polypeptide, (b) an agonist of a PRO polypeptide, or
  • an antagonist of a PRO polypeptide wherein endothelial cell growth in said mammal is inhibited, and wherein said agonist or antagonist may be an anti-PRO antibody.
  • the mammal is human and the endothelial cell growth is associated with a tumor or a retinal disorder.
  • the invention provides a method for stimulating endothelial cell growth in a mammal comprising administering to the mammal (a) a PRO polypeptide, (b) an agonist of a PRO polypeptide, or (c) an antagonist of a PRO polypeptide, wherein endothelial cell growth in said mammal is stimulated, and wherein said agonist or antagonist may be an anti-PRO antibody.
  • the mammal is human.
  • the invention provides a method for inhibiting cardiac hypertrophy in a mammal comprising administering to the mammal (a) a PRO polypeptide, (b) an agonist of a PRO polypeptide, or (c) an antagonist of a PRO polypeptide, wherein cardiac hypertrophy in said mammal is inhibited, and wherein said agonist or antagonist may be an anti-PRO antibody.
  • the mammal is human and the cardiac hypertrophy has been induced by myocardial infarction.
  • the invention provides a method for stimulating cardiac hypertrophy in a mammal comprising administering to the mammal (a) a PRO polypeptide, (b) an agonist of a PRO polypeptide, or (c) an antagonist of a PRO polypeptide, wherein cardiac hypertrophy in said mammal is stimulated, and wherein said agonist or antagonist may be an anti-PRO antibody.
  • the mammal is human who suffers from congestive heart failure.
  • the invention provides a method for inliibiting angiogenesis induced by a PRO polypeptide in a mammal comprising administering a therapeutically effective amount of an anti-PRO antibody to the mammal.
  • the mammal is a human, and more preferably the mammal has a tumor or a retinal disorder.
  • the invention provides a method for stimulating angiogenesis induced by a
  • PRO polypeptide in a mammal comprising administering a therapeutically effective amount of a PRO polypeptide to the mammal.
  • the mammal is a human, and more preferably angiogenesis would promote tissue regeneration or wound healing.
  • the invention provides a method for modulating (e.g. , inhibiting or stimulating) endothelial cell growth in a mammal comprising administering to the mammal a PR021, PR0181, PRO205,
  • PR0214 PR0221, PR0229, PR0231, PR0238, PR0241, PR0247, PR0256, PR0258, PR0263, PR0265, PR0295, PR0321, PR0322, PR0337, PR0363, PR0365, PR0444, PR0533, PR0697, PRO720, PR0725, PR0771, PR0788, PR0791, PR0819, PR0827, PR0828, PR0836, PR0846, PR0865, PRO1005, PRO1006, PRO1007, PRO1025, PRO1029, PRO1054, PRO1071, PRO1075, PRO1079, PRO1080, PR01114, PROH31, PR01155, PRO1160, PROH84, PR01186, PRO1190, PROH92, PROH95, PR01244, PR01272, PR01273,
  • the invention provides amethod for modulating (e.g. , inliibiting or stimulating) smooth muscle cell growth in a mammal comprising administering to the mammal a PRO 162, PR0181, PRO 182, PR0195, PRO204, PR0221, PRO230, PR0256, PR0258, PR0533, PR0697, PR0725, PR0738, PR0826, PR0836,PR0840,PR0846,PR0865,PR0982,PR01025,PR01029,PR01071,PR01080,PR01083,PR01134,
  • a PRO 162, PR0181, PRO 182, PR0195, PRO204, PR0221, PRO230, PR0256, PR0258, PR0533, PR0697, PR0725, PR0738, PR0826, PR0836,PR0840,PR0846,PR0865,PR0982,PR01025,PR01029,PR01071,PR01080,PR01083,PR01134 a method for modulating (e
  • the invention provides a method for modulating (e.g. , inducing or reducing) cardiac hypertrophy in a mammal comprising administering to the mammal a PR021 polypeptide, agonist or antagonist thereof, wherein cardiac hypertrophy in said mammal is modulated.
  • the invention provides a method for modulating (e.g., inducing or reducing) endothelial cell apoptosis in a mammal comprising administering to the mammal a PRO4302 polypeptide, agonist or antagonist thereof, wherein cardiac hypertrophy in said mammal is modulated.
  • the invention provides a method for modulating (e.g. , stimulating or inhibiting) angiogenesis in a mammal comprising administering a therapeutically effective amount of a PRO 1376 or PRO 1449 polypeptide, agonist or antagonist thereof to the mammal, wherein said angiogenesis is modulated.
  • the invention provides a method for modulating (e.g., inducing or reducing) angiogenesis by modulating (e.g., inducing or reducing) endothelial cell tube formation in a mammal comprising administering to the mammal a PR0178, PR0195, PR0228, PRO301, PRO302, PR0532, PR0724, PRO730, PR0734,PR0793,PR0871,PR0938,PR01012,PR01120,PR01139,PR01198,PR01287,PR01361,PR01864,
  • the invention provides an isolated nucleic acid molecule comprising a nucleotide sequence that encodes a PRO polypeptide.
  • the isolated nucleic acid molecule comprises a nucleotide sequence having at least about 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97% or 98% nucleic acid sequence identity and alternatively at least about 99% nucleic acid sequence identity to (a) a DNA molecule encoding a PRO polypeptide having a full-length amino acid sequence as disclosed herein, an amino acid sequence lacking the signal peptide as disclosed herein, an extracellular domain of a transmembrane protein, with or without the signal peptide, as disclosed herein or any other specifically defined fragment ofthe full-length amino acid sequence as disclosed herein, or (b) the complement ofthe DNA molecule of (a).
  • the isolated nucleic acid molecule comprises a nucleotide sequence having at least about 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97% or 98% nucleic acid sequence identity and alternatively at least about 99% nucleic acid sequence identity to (a) a DNA molecule comprising the coding sequence of a full-length PRO polypeptide cDNA as disclosed herein, the coding sequence of a PRO polypeptide lacking the signal peptide as disclosed herein, the coding sequence of an extracellular domain of a transmembrane PRO polypeptide, with or without the signal peptide, as disclosed herein or the coding sequence of any other specifically defined fragment of the full-length amino acid sequence as disclosed herein, or (b) the complement ofthe DNA molecule of (a).
  • the invention provides an isolated nucleic acid molecule comprising a nucleotide sequence having at least about 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%,
  • nucleic acid sequence identity 94%, 95%o, 96%, 97% or 98% nucleic acid sequence identity and alternatively at least about 99% nucleic acid sequence identity to (a) a DNA molecule that encodes the same mature polypeptide encoded by any ofthe human protein cDNAs deposited with the ATCC as disclosed herein, or (b) the complement ofthe DNA molecule of (a).
  • Another aspect ofthe present invention provides an isolated nucleic acid molecule comprising a nucleotide sequence encoding a PRO polypeptide which is either transmembrane domain-deleted or transmembrane domain-inactivated, or is complementary to such encoding nucleotide sequence, wherein the transmembrane domain(s) of such polypeptide are disclosed herein. Therefore, soluble extracellular domains ofthe herein described PRO polypeptides are contemplated.
  • Another embodiment is directed to fragments of a PRO polypeptide coding sequence, or the complement thereof, that may find use as, for example, hybridization probes, for encoding fragments of a PRO polypeptide that may optionally encode a polypeptide comprising a binding site for an anti-PRO antibody or as antisense oligonucleotide probes.
  • nucleic acid fragments are usually at least about 20, 30, 40, 50, 60, 70, 80, 90, 100, 110, 120, 130, 140, 150, 160, 170, 180, 190, 200, 250, 300, 350, 400, 450, 500, 600, 700 or 800 nucleotides in length and alternatively at least about 1000 nucleotides in length, wherein in this context the term "about” means the referenced nucleotide sequence length plus or minus 10% of that referenced length.
  • novel fragments of a PRO polypeptide-encoding nucleotide sequence may be determined in a routine manner by aligning the PRO polypeptide-encoding nucleotide sequence with other known nucleotide sequences using any of a number of well known sequence alignment programs and determining which PRO polypeptide-encoding nucleotide sequence fragments) are novel. All of such PRO polypeptide-encoding nucleotide sequences are contemplated herein. Also contemplated are the PRO polypeptide fragments encoded by these nucleotide molecule fragments, preferably those PRO polypeptide fragments that comprise a binding site for an anti-PRO antibody.
  • the invention provides an isolated PRO polypeptide encoded by any ofthe isolated nucleic acid sequences hereinabove identified.
  • the invention provides an isolatedPRO polypeptide comprising an amino acid sequence having at least about 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97% or 98% amino acid sequence identity and alternatively at least about 99% amino acid sequence identity to a PRO polypeptide having a full-length amino acid sequence as disclosed herein, an amino acid sequence lacking the signal peptide as disclosed herein, an extracellular domain of a transmembrane protein, with or without the signal peptide, as disclosed herein or any other specifically defined fragment ofthe full-length amino acid sequence as disclosed herein.
  • the invention provides an isolated PRO polypeptide comprising an amino acid sequence having at least about 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%,
  • the invention provides an isolated PRO polypeptide without the N-terminal signal sequence and/or the initiating methionine and that is encoded by a nucleotide sequence that encodes such an amino acid sequence as hereinbefore described.
  • Processes forproducing the same are also herein described, wherein those processes comprise culturing a host cell comprising a vector which comprises the appropriate encoding nucleic acid molecule under conditions suitable for expression ofthe PRO polypeptide and recovering the PRO polypeptide from the cell culture.
  • Another aspect of the invention provides an isolated PRO polypeptide which is either transmembrane domain-deleted or transmembrane domain-inactivated.
  • Processes forproducing the same are also herein described, wherein those processes comprise culturing a host cell comprising a vector which comprises the appropriate encoding nucleic acid molecule under conditions suitable for expression ofthe PRO polypeptide and recovering the PRO polypeptide from the cell culture.
  • the invention provides agonists and antagonists of a native PRO polypeptide as defined herein.
  • the agonist or antagonist is an anti-PRO antibody or a small molecule.
  • the invention provides a method of identifying agonists or antagonists to a PRO polypeptide which comprise contacting the PRO polypeptide with a candidate molecule and monitoring a biological activity mediated by said PRO polypeptide.
  • the PRO polypeptide is a native PRO polypeptide.
  • the invention provides a composition of matter comprising a PRO polypeptide, or an agonist or antagonist of a PRO polypeptide as herein described, or an anti-PRO antibody, in combination with a carrier.
  • the carrier is a pharmaceutically acceptable carrier.
  • Another embodiment ofthe present invention is directed to the use of a PRO polypeptide, or an agonist or antagonist thereof as hereinbefore described, or an anti-PRO antibody, for the preparation of a medicament useful in the treatment of a condition which is responsive to the PRO polypeptide, an agonist or antagonist thereof or an anti-PRO antibody.
  • the invention provides vectors comprising DNA encoding any ofthe herein described polypeptides.
  • Host cells comprising any such vector are also provided.
  • the host cells may be CHO cells, E. coli, yeast, or Baculo virus-infected insect cells.
  • a process for producing any ofthe herein described polypeptides is further provided and comprises culturing host cells under conditions suitable for expression ofthe desired polypeptide and recovering the desired polypeptide from the cell culture.
  • the invention provides chimeric molecules comprising any ofthe herein described polypeptides fused to a heterologous polypeptide or amino acid sequence.
  • Example of such chimeric molecules comprise any of the herein described polypeptides fused to an epitope tag sequence or a Fc region of an immunoglobulin.
  • the invention provides an antibody which specifically binds to any ofthe above or below described polypeptides.
  • the antibody is a monoclonal antibody, humanized antibody, antibody fragment or single-chain antibody.
  • the invention provides oligonucleotide probes useful for isolating genomic and cDNA nucleotide sequences or as antisense probes, wherein those probes may be derived from any of the above or below described nucleotide sequences.
  • Figure 1 shows a nucleotide sequence (SEQ ID NO : 1 ) of a native sequence PRO 181 cDNA, wherein SEQ JD NO: 1 is a clone designated herein as "DNA23330-1390".
  • Figure 2 shows the amino acid sequence (SEQ ID NO:2) derived from the coding sequence of SEQ ID NO:l shown in Figure 1.
  • Figure 3 shows a nucleotide sequence (SEQ JD NO:3) of a native sequence PRO 178 cDNA, wherein SEQ ID NO:3 is a clone designated herein as "DNA23339-1130".
  • Figure 4 shows the amino acid sequence (SEQ ID NO:4) derived from the coding sequence of SEQ JD
  • Figure 5 shows a nucleotide sequence (SEQ JD NO:5) of a native sequence PR0444 cDNA, wherein SEQ ID NO:5 is a clone designated herein as "DNA26846-1397".
  • Figure 6 shows the amino acid sequence (SEQ JD NO:6) derived from the coding sequence of SEQ ID NO:5 shown in Figure 5.
  • Figure 7 shows a nucleotide sequence (SEQ ID NO:7) of a native sequence PRO 195 cDNA, wherein SEQ JD NO:7 is a clone designated herein as "DNA26847-1395".
  • Figure 8 shows the amino acid sequence (SEQ ID NO: 8) derived from the coding sequence of SEQ ID NO: 7 shown in Figure 7.
  • Figure 9 shows a nucleotide sequence (SEQ JD NO :9) of a native sequence PRO 182 cDNA, wherein SEQ
  • ID NO:9 is a clone designated herein as "DNA27865-1091".
  • Figure 10 shows the amino acid sequence (SEQ ID NO:10) derived from the coding sequence of SEQ JD NO:9 shown in Figure 9.
  • Figure 11 shows a nucleotide sequence (SEQ JD NO: 11) of a native sequence PRO205 cDNA, wherein SEQ ID NO:l 1 is a clone designated herein as "DNA30868-1156".
  • Figure 12 shows the amino acid sequence (SEQ ID NO:12) derived from the coding sequence of SEQ ID NO : 11 shown in Figure 11.
  • Figure 13 shows a nucleotide sequence (SEQ ID NO:13) of a native sequence PRO204 cDNA, wherein SEQ ID NO: 13 is a clone designated herein as "DNA30871-1157".
  • Figure 14 shows the amino acid sequence (SEQ ID NO:14) derived from the coding sequence of SEQ ID NO:13 shown in Figure 13.
  • Figure 15 shows a nucleotide sequence (SEQ ID NO: 15) of a native sequence PR01873 cDNA, wherein
  • SEQ JD N0:15 is a clone designated herein as "DNA30880".
  • Figure 16 shows the amino acid sequence (SEQ ID N0:16) derived from the coding sequence of SEQ ID NO: 15 shown in Figure 15.
  • Figure 17 shows a nucleotide sequence (SEQ ID NO: 17) of a native sequence PR0214 cDNA, wherein SEQ ID NO: 17 is a clone designated herein as "DNA32286-1191".
  • Figure 18 shows the amino acid sequence (SEQ ID NO: 18) derived from the coding sequence of SEQ JD NO: 17 shown in Figure 17.
  • Figure 19 shows a nucleotide sequence (SEQ ID NO:19) of a native sequence PR0221 cDNA, wherein SEQ JD NO:19 is a clone designated herein as "DNA33089-1132".
  • Figure 20 shows the amino acid sequence (SEQ JD NO:20) derived from the coding sequence of SEQ JD
  • Figure 21 shows a nucleotide sequence (SEQ ID NO:21) of a native sequence PR0228 cDNA, wherein SEQ ID NO:21 is a clone designated herein as "DNA33092-1202".
  • Figure 22 shows the amino acid sequence (SEQ ID NO:22) derived from the coding sequence of SEQ ID NO:21 shown in Figure 21.
  • Figure 23 shows a nucleotide sequence (SEQ JD NO:23) of a native sequence PR0229 cDNA, wherein SEQ ID N0:23 is a clone designated herein as "DNA33100-1159".
  • Figure 24 shows the amino acid sequence (SEQ JD NO:24) derived from the coding sequence of SEQ ID N0:23 shown in Figure 23.
  • Figure 25 shows a nucleotide sequence (SEQ ID NO:25) of a native sequence PRO230 cDNA, wherein
  • SEQ TD NO:25 is a clone designated herein as "DNA33223-1136".
  • Figure 26 shows the amino acid sequence (SEQ ID NO:26) derived from the coding sequence of SEQ JD NO:25 shown in Figure 25.
  • Figure 27 shows a nucleotide sequence (SEQ ID NO:27) of a native sequence PR07223 cDNA, wherein SEQ ID N0:27 is a clone designated herein as "DNA34385".
  • Figure 28 shows the amino acid sequence (SEQ ID N0:28) derived from the coding sequence of SEQ JD N0:27 shown in Figure 27.
  • Figure 29 shows a nucleotide sequence (SEQ ID NO:29) of a native sequence PR0241 cDNA, wherein SEQ ID N0:29 is a clone designated herein as "DNA34392-1170".
  • Figure 30 shows the amino acid sequence (SEQ ID NO:30) derived from the coding sequence of SEQ ID NO:29 shown in Figure 29.
  • Figure 31 shows a nucleotide sequence (SEQ ID NO:31) of a native sequence PR0263 cDNA, wherein
  • SEQ ID NO:31 is a clone designated herein as "DNA34431-1177".
  • Figure 32 shows the amino acid sequence (SEQ ID NO:32) derived from the coding sequence of SEQ JD N0:31 shown in Figure 31.
  • Figure 33 shows a nucleotide sequence (SEQ ID NO:33) of a native sequence PR0321 cDNA, wherein SEQ ID N0:33 is a clone designated herein as "DNA34433-1308".
  • Figure 34 shows the amino acid sequence (SEQ JD N0:34) derived from the coding sequence of SEQ JD N0:33 shown in Figure 33.
  • Figure 35 shows a nucleotide sequence (SEQ ID NO:35) of a native sequence PR0231 cDNA, wherein SEQ ID N0:35 is a clone designated herein as "DNA34434-1139".
  • Figure 36 shows the amino acid sequence (SEQ ID N0:36) derived from the coding sequence of SEQ JD
  • Figure 37 shows a nucleotide sequence (SEQ JD NO:37) of a native sequence PR0238 cDNA, wherein SEQ ID NO:37 is a clone designated herein as "DNA35600- 1162".
  • Figure 38 shows the amino acid sequence (SEQ ID NO:38) derived from the coding sequence of SEQ JD NO:37 shown in Figure 37.
  • Figure 39 shows a nucleotide sequence (SEQ JD NO: 39) of a native sequence PR0247 cDNA, wherein SEQ JD NO:39 is a clone designated herein as "DNA35673-1201".
  • Figure 40 shows the amino acid sequence (SEQ ID NO:40) derived from the coding sequence of SEQ JD NO:39 shown in Figure 39.
  • Figure 41 shows a nucleotide sequence (SEQ JD NO:41) of a native sequence PR0256 cDNA, wherein
  • SEQ ID NO:41 is a clone designated herein as "DNA35880-1160".
  • Figure 42 shows the amino acid sequence (SEQ ID NO:42) derived from the coding sequence of SEQ JD NO:41 shown in Figure 41.
  • Figure 43 shows a nucleotide sequence (SEQ JD NO:43) of a native sequence PR0258 cDNA, wherein SEQ JD NO:43 is a clone designated herein as "DNA35918-1174".
  • Figure 44 shows the amino acid sequence (SEQ JD NO:44) derived from the coding sequence of SEQ ID NO:43 shown in Figure 43.
  • Figure 45 shows a nucleotide sequence (SEQ JD NO:45) of a native sequence PR0265 cDNA, wherein SEQ ID N0:45 is a clone designated herein as "DNA36350-1158".
  • Figure 46 shows the amino acid sequence (SEQ ID NO :46) derived from the coding sequence of SEQ JD
  • Figure 47 shows a nucleotide sequence (SEQ ID NO:47) of a native sequence PR021 cDNA, wherein SEQ JD NO:47 is a clone designated herein as "DNA36638-1056".
  • Figure 48 shows the amino acid sequence (SEQ JD NO:48) derived from the coding sequence of SEQ JD NO:47 shown in Figure 47.
  • Figure 49 shows a nucleotide sequence (SEQ ID NO:49) of a native sequence PR0295 cDNA, wherein SEQ ID NO:49 is a clone designated herein as "DNA38268-1188".
  • Figure 50 shows the amino acid sequence (SEQ ID NO:50) derived from the coding sequence of SEQ JD NO:49 shown in Figure 49.
  • Figure 51 shows a nucleotide sequence (SEQ ID NO:51) of a native sequence PRO302 cDNA, wherein SEQ ID NO:51 is a clone designated herein as "DNA40370-1217".
  • Figure 52 shows the amino acid sequence (SEQ JD NO:52) derived from the coding sequence of SEQ ID NO:52
  • Figure 53 shows a nucleotide sequence (SEQ JD NO:53) of a native sequence PRO301 cDNA, wherein SEQ ID NO:53 is a clone designated herein as "DNA40628-1216".
  • Figure 54 shows the amino acid sequence (SEQ ID NO:54) derived from the coding sequence of SEQ ID NO:53 shown in Figure 53.
  • Figure 55 shows a nucleotide sequence (SEQ ID NO:55) of a native sequence PR0337 cDNA, wherein SEQ ID N0:55 is a clone designated herein as "DNA43316-1237".
  • Figure 56 shows the amino acid sequence (SEQ ID NO:56) derived from the coding sequence of SEQ ID NO:55 shown in Figure 55.
  • Figure 57 shows a nucleotide sequence (SEQ ID NO:57) of a native sequence PR07248 cDNA, wherein
  • SEQ ID NO:57 is a clone designated herein as "DNA44195".
  • Figure 58 shows the amino acid sequence (SEQ JD NO:58) derived from the coding sequence of SEQ JD NO:57 shown in Figure 57.
  • Figure 59 shows a nucleotide sequence (SEQ ID NO:59) of a native sequence PR0846 cDNA, wherein SEQ ID NO:59 is a clone designated herein as "DNA44196-1353".
  • Figure 60 shows the amino acid sequence (SEQ JD NO:60) derived from the coding sequence of SEQ JD NO:59 shown in Figure 59.
  • Figure 61 shows a nucleotide sequence (SEQ JD NO: 61 ) of a native sequence PRO 1864 cDNA, wherein SEQ ID NO:61 is a clone designated herein as "DNA45409-2511".
  • Figure 62 shows the amino acid sequence (SEQ ID NO:62) derived from the coding sequence of SEQ ID NO:
  • Figure 63 shows a nucleotide sequence (SEQ JD NO:63) of a native sequence PR0363 cDNA, wherein SEQ ID N0:63 is a clone designated herein as "DNA45419-1252".
  • Figure 64 shows the amino acid sequence (SEQ ID NO: 64) derived from the coding sequence of SEQ ID NO:63 shown in Figure 63.
  • Figure 65 shows a nucleotide sequence (SEQ JD NO:65) of a native sequence PRO730 cDNA, wherein SEQ JD NO:65 is a clone designated herein as "DNA45624-1400".
  • Figure 66 shows the amino acid sequence (SEQ JD NO:66) derived from the coding sequence of SEQ ID NO:65 shown in Figure 65.
  • Figure 67 shows a nucleotide sequence (SEQ ID NO:67) of a native sequence PR0365 cDNA, wherein SEQ ID NO:67 is a clone designated herein as "DNA46777-1253".
  • Figure 68 shows the amino acid sequence (SEQ ID NO:68) derived from the coding sequence of SEQ JD
  • Figure 69 shows a nucleotide sequence (SEQ JD NO:69) of a native sequence PR0532 cDNA, wherein SEQ ID NO:69 is a clone designated herein as "DNA48335".
  • Figure 70 shows the amino acid sequence (SEQ ID NO:70) derived from the coding sequence of SEQ ID NO:69 shown in Figure 69.
  • Figure 71 shows a nucleotide sequence (SEQ JD NO:71) of a native sequence PR0322 cDNA, wherein SEQ JD NO:71 is a clone designated herein as "DNA48336-1309".
  • Figure 72 shows tire amino acid sequence (SEQ JD NO:72) derived from the coding sequence of SEQ ID NO:71 shown in Figure 71.
  • Figure 73 shows a nucleotide sequence (SEQ JD NO:73) of a native sequence PROl 120 cDNA, wherein
  • SEQ JD NO:73 is a clone designated herein as "DNA48606-1479".
  • Figure 74 shows the amino acid sequence (SEQ ID NO:74) derived from the coding sequence of SEQ ID NO:73 shown in Figure 73.
  • Figure 75 shows a nucleotide sequence (SEQ JD NO:75) of a native sequence PR07261 cDNA, wherein SEQ ID NO:75 is a clone designated herein as "DNA49149".
  • Figure 76 shows the amino acid sequence (SEQ ID NO:76) derived from the coding sequence of SEQ JD NO:75 shown in Figure 75.
  • Figure 77 shows a nucleotide sequence (SEQ ID NO:77) of a native sequence PR0533 cDNA, wherein SEQ TD NO:77 is a clone designated herein as "DNA49435-1219".
  • Figure 78 shows the amino acid sequence (SEQ ID NO:78) derived from the coding sequence of SEQ ID NO:
  • Figure 79 shows a nucleotide sequence (SEQ ID NO:79) of a native sequence PR0724 cDNA, wherein SEQ ID NO:79 is a clone designated herein as "DNA49631-1328".
  • Figure 80 shows the amino acid sequence (SEQ JD NO:80) derived from the coding sequence of SEQ JD NO:79 shown in Figure 79.
  • Figure 81 shows a nucleotide sequence (SEQ JD NO:81) of a native sequence PR0734 cDNA, wherein SEQ ID NO:81 is a clone designated herein as "DNA49817" .
  • Figure 82 shows the amino acid sequence (SEQ ID NO:82) derived from the coding sequence of SEQ JD NO: 81 shown in Figure 81.
  • Figure 83 shows a nucleotide sequence (SEQ ID NO:83) of a native sequence PR0771 cDNA, wherein
  • SEQ JD NO:83 is a clone designated herein as "DNA49829-1346".
  • Figure 84 shows the amino acid sequence (SEQ JD NO:84) derived from the coding sequence of SEQ JD NO: 83 shown in Figure 83.
  • Figure 85 shows a nucleotide sequence (SEQ JD NO:85) of a native sequence PRO2010 cDNA, wherem SEQ JD NO:85 is a clone designated herein as "DNA50792".
  • Figure 86 shows the amino acid sequence (SEQ ID NO:86) derived from the coding sequence of SEQ ID NO:85 shown in Figure 85.
  • Figure 87 shows a nucleotide sequence (SEQ ID NO:87) of a native sequence PR0871 cDNA, wherein SEQ JD NO:87 is a clone designated herein as "DNA50919-1361".
  • Figure 88 shows the amino acid sequence (SEQ ID NO:88) derived from the coding sequence of SEQ JD NO:87 shown in Figure 87.
  • Figure 89 shows a nucleotide sequence (SEQ JD NO:89) of a native sequence PR0697 cDNA, wherein
  • SEQ ID NO:89 is a clone designated herein as "DNA50920-1325”.
  • Figure 90 shows the amino acid sequence (SEQ ID NO:90) derived from the coding sequence of SEQ ID NO:89 shown in Figure 89.
  • Figure 91 shows a nucleotide sequence (SEQ ID NO:91) of a native sequence PRO1083 cDNA, wherein SEQ JD NO:91 is a clone designated herein as "DNA50921-1458".
  • Figure 92 shows the amino acid sequence (SEQ JD NO:22) derived from the coding sequence of SEQ JD NO:91 shown in Figure 91.
  • Figure 93 shows a nucleotide sequence (SEQ ID NO:93) of a native sequence PR0725 cDNA, wherein SEQ JD NO:93 is a clone designated herein as "DNA52758-1399".
  • Figure 94 shows the amino acid sequence (SEQ ID NO:94) derived from the coding sequence of SEQ JD
  • Figure 95 shows a nucleotide sequence (SEQ ID NO:95) of a native sequence PRO720 cDNA, wherein SEQ JD NO:95 is a clone designated herein as "DNA53517-1366-1".
  • Figure 96 shows the amino acid sequence (SEQ ID NO:96) derived from the coding sequence of SEQ ID NO:95 shown in Figure 95.
  • Figure 97 shows a nucleotide sequence (SEQ JD NO:97) of a native sequence PR0738 cDNA, wherein SEQ ID NO:97 is a clone designated herein as "DNA53915-1258".
  • Figure 98 shows the amino acid sequence (SEQ ID NO:98) derived from the coding sequence of SEQ JD NO: 97 shown in Figure 97.
  • Figure 99 shows a nucleotide sequence (SEQ ID NO:99) of a native sequence PR0865 cDNA, wherein
  • SEQ ID NO:99 is a clone designated herein as "DNA53974-1401".
  • Figure 100 shows the amino acid sequence (SEQ JD NO: 100) derived from the coding sequence of SEQ JD NO:99 shown in Figure 99.
  • Figure 101 shows a nucleotide sequence (SEQ ID NO : 101 ) of a native sequence PRO840 cDNA, wherein SEQ JD NO: 101 is a clone designated herein as "DNA53987-1438".
  • Figure 102 shows the amino acid sequence (SEQ JD NO: 102) derived from the coding sequence of SEQ J NO:101 shown in Figure 101.
  • Figure 103 shows anucleotide sequence (SEQ ID NO: 103) of a native sequence PRO 1080 cDNA, wherein SEQ JD NO:103 is a clone designated herein as "DNA56047-1456".
  • Figure 104 shows the amino acid sequence (SEQ JD NO: 104) derived from the coding sequence of SEQ ID NO:103 shown in Figure 103.
  • Figure 105 shows a nucleotide sequence (SEQ JD NO: 105) of a native sequence PRO 1079 cDNA, wherein
  • SEQ ID NO:105 is a clone designated herein as "DNA56050-1455".
  • Figure 106 shows the amino acid sequence (SEQ ID NO: 106) derived from the coding sequence of SEQ ID NO: 105 shown in Figure 105.
  • Figure 107 shows a nucleotide sequence (SEQ JD NO : 107) of a native sequence PR0793 cDNA, wherein SEQ ID NO: 107 is a clone designated herein as "DNA56110-1437".
  • Figure 108 shows the amino acid sequence (SEQ ID NO: 108) derived from the coding sequence of SEQ JD NO: 107 shown in Figure 107.
  • Figure 109 shows anucleotide sequence (SEQ ID NO:109) of a native sequence PR0788 cDNA, wherein SEQ JD NO:109 is a clone designated herein as "DNA56405-1357”.
  • Figure 110 shows the amino acid sequence (SEQ ID NO: 110) derived from the coding sequence of SEQ
  • Figure 111 shows a nucleotide sequence (SEQ ID NO: 111) of a native sequence PR0938 cDNA, wherein SEQ ID NO:l 11 is a clone designated herein as "DNA56433-1406".
  • Figure 112 shows the amino acid sequence (SEQ ID NO: 112) derived from the coding sequence of SEQ JD NO:l 11 shown in Figure 111.
  • Figure 113 shows anucleotide sequence (SEQ ID NO: 113) of a native sequence PRO1012 cDNA, wherein SEQ JD NO: 113 is a clone designated herein as "DNA56439-1376".
  • Figure 114 shows the amino acid sequence (SEQ JD NO:l 14) derived from the coding sequence of SEQ ID NO:113 shown in Figure 113.
  • Figure 115 shows a nucleotide sequence (SEQ ID NO : 115) of a native sequence PRO 1477 cDNA, wherein
  • SEQ JD NO:l 15 is a clone designated herein as "DNA56529-1647".
  • Figure 116 shows the amino acid sequence (SEQ ID NO: 116) derived from the coding sequence of SEQ JD NO: 115 shown in Figure 115.
  • Figure 117 shows anucleotide sequence (SEQJDNO:l 17) of anative sequence PROl 134 cDNA, wherein SEQ ID NO:l 17 is a clone designated herein as "DNA56865-1491".
  • Figure 118 shows the amino acid sequence (SEQ ID NO: 118) derived from the coding sequence of SEQ JD NO: 117 shown in Figure 117.
  • Figure 119 shows a nucleotide sequence (SEQ ID NO: 119) of a native sequence PRO 162 cDNA, wherein SEQ ID NO:119 is a clone designated herein as "DNA56965-1356".
  • Figure 120 shows the amino acid sequence (SEQ JD NO:120) derived from the coding sequence of SEQ
  • Figure 121 shows a nucleotide sequence (SEQ JD NO: 121) of a native sequence PRO 1114 cDNA, wherein SEQ JD NO:121 is a clone designated herein as "DNA57033-1403-1".
  • Figure 122 shows the amino acid sequence (SEQ ID NO: 122) derived from the coding sequence of SEQ ID NO-.121 shown in Figure 121.
  • Figure 123 shows a nucleotide sequence (SEQ ID NO: 123) of a native sequence PR0828 cDNA, wherein SEQ ID NO: 123 is a clone designated herein as "DNA57037-1444".
  • Figure 124 shows the amino acid sequence (SEQ ID NO: 124) derived from the coding sequence of SEQ ID NO:123 shown in Figure 123.
  • Figure 125 shows a nucleotide sequence (SEQ ID NO: 125) of a native sequence PR0827 cDNA, wherein SEQ JD NO: 125 is a clone designated herein as "DNA57039-1402".
  • Figure 126 shows the amino acid sequence (SEQ ID NO: 126) derived from the coding sequence of SEQ
  • Figure 127 shows a nucleotide sequence (SEQ ID NO: 127) of a native sequence PRO 1075 cDNA, wherein SEQ JD NO:127 is a clone designated herein as "DNA57689-1385".
  • Figure 128 shows the amino acid sequence (SEQ JD NO:128) derived from tire coding sequence of SEQ ID NO: 127 shown in Figure 127.
  • Figure 129 shows a nucleotide sequence (SEQ ID NO: 129) of a native sequence PRO 1 07 cDNA, wherein SEQ ID NO:129 is a clone designated herein as "DNA57690-1374".
  • Figure 130 shows the amino acid sequence (SEQ ID NO: 130) derived from the coding sequence of SEQ ID NO: 129 shown in Figure 129.
  • Figure 131 shows a nucleotide sequence (SEQ ID NO: 131) of a native sequence PR0826 cDNA, wherein
  • SEQ ID NO: 131 is a clone designated herein as "DNA57694-1341".
  • Figure 132 shows the amino acid sequence (SEQ JD NO: 132) derived from the coding sequence of SEQ ID NO: 131 shown in Figure 131.
  • Figure 133 shows a nucleotide sequence (SEQ ID NO: 133) of a native sequence PR0819 cDNA, wherein SEQ ID NO: 132 is a clone designated herein as "DNA57695-1340".
  • Figure 134 shows the amino acid sequence (SEQ ID NO: 134) derived from the coding sequence of SEQ ID NO.T33 shown in Figure 133.
  • Figure 135 shows anucleotide sequence (SEQ ID NO: 135) of a native sequence PRO 1006 cDNA, wherein SEQ ID NO: 135 is a clone designated herein as "DNA57699-1412".
  • Figure 136 shows the amino acid sequence (SEQ JD NO: 136) derived from the coding sequence of SEQ
  • Figure 137 shows a nucleotide sequence (SEQ JD NO: 137) of a native sequence PR0982 cDNA, wherein SEQ ID NO:137 is a clone designated herein as 'DNA57700-1408".
  • Figure 138 shows the amino acid sequence (SEQ ID NO:138) derived from the coding sequence of SEQ JD NO:137 shown in Figure 137.
  • Figure 139 shows a nucleotide sequence (SEQ JD NO: 139) of a native sequence PRO 1005 cDNA, wherein SEQ JD NO: 139 is a clone designated herein as "DNA57708-1411".
  • Figure 140 shows the amino acid sequence (SEQ JD NO: 140) derived from the coding sequence of SEQ ID NO: 139 shown in Figure 139.
  • Figure 141 shows anucleotide sequence (SEQ JDNO:141) of anative sequence PR0791 cDNA, wherein SEQ ID NO: 141 is a clone designated herein as "DNA57838-1337".
  • Figure 142 shows the amino acid sequence (SEQ ID NO: 142) derived from the coding sequence of SEQ
  • Figure 143 shows a nucleotide sequence (SEQ ID NO : 143) of a native sequence PRO 1071 cDNA, wherein SEQ ID NO: 143 is a clone designated herein as "DNA58847-1383".
  • Figure 144 shows the amino acid sequence (SEQ ID NO: 144) derived from the coding sequence of SEQ ID NO.T43 shown in Figure 43.
  • Figure 145 shows a nucleotide sequence (SEQ ID NO : 145) of a native sequence PRO 1415 cDNA, wherein SEQ ID NO:145 is a clone designated herein as "DNA58852-1637".
  • Figure 146 shows the amino acid sequence (SEQ ID NO: 146) derived from the coding sequence of SEQ ID NO: 145 shown in Figure 145.
  • Figure 147 shows anucleotide sequence (SEQ JD NO: 147) of anative sequence PRO 1054 cDNA, wherein
  • SEQ ID NO: 147 is a clone designated herein as "DNA58853-1423".
  • Figure 148 shows the amino acid sequence (SEQ JD NO: 148) derived from the coding sequence of SEQ JD NO: 147 shown in Figure 147.
  • Figure 149 shows anucleotide sequence (SEQ IDNO:149) of anative sequence PRO 1411 cDNA, wherein SEQ ID NO:149 is a clone designated herein as "DNA59212-1627".
  • Figure 150 shows the amino acid sequence (SEQ ID NO: 150) derived from the coding sequence of SEQ ID NO: 149 shown in Figure 149.
  • Figure 151 shows a nucleotide sequence (SEQ JD NO : 151 ) of a native sequence PRO 1184 cDNA, wherein SEQ ID NO:151 is a clone designated herein as "DNA59220-1514".
  • Figure 152 shows the amino acid sequence (SEQ ID NO: 152) derived from the coding sequence of SEQ
  • Figure 153 shows a nucleotide sequence (SEQJDNO:153) of anative sequence PRO 1029 cDNA, wherein SEQ JD NO:153 is a clone designated herein as "DNA59493-1420".
  • Figure 154 shows the amino acid sequence (SEQ ID NO: 154) derived from the coding sequence of SEQ ID NO:153 shown in Figure 153.
  • Figure 155 shows anucleotide sequence (SEQ ID NO:155) of anative sequence PROl 139 cDNA, wherein SEQ ID NO: 155 is a clone designated herein as "DNA59497-1496".
  • Figure 156 shows the amino acid sequence (SEQ JD NO: 156) derived from the coding sequence of SEQ ID NO: 155 shown in Figure 155.
  • Figure 157 shows a nucleotide sequence (SEQ ID NO: 157) of a native sequence PRO 1190 cDNA, wherein
  • SEQ ID NO: 157 is a clone designated herein as "DNA59586-1520".
  • Figure 158 shows the amino acid sequence (SEQ ID NO: 158) derived from the coding sequence of SEQ JD NO:157 shown in Figure 157.
  • Figure 159 shows anucleotide sequence (SEQJDNO:159) of anative sequence PRO1309 cDNA, wherein SEQ ID NO: 159 is a clone designated herein as "DNA59588-1571".
  • Figure 160 shows the amino acid sequence (SEQ ID NO: 160) derived from the coding sequence of SEQ ID NO: 159 shown in Figure 159.
  • Figure 161 shows a nucleotide sequence (SEQ JD NO: 161 ) of a native sequence PR0836 cDNA, wherein SEQ ID N0:161 is a clone designated herein as "DNA59620-1463".
  • Figure 162 shows the amino acid sequence (SEQ ID NO: 162) derived from the coding sequence of SEQ JD NO:161 shown in Figure 161.
  • Figure 163 shows a nucleotide sequence (SEQ JD NO: 163) of a native sequence PRO 1025 cDNA, wherein
  • SEQ ID NO:163 is a clone designated herein as "DNA59622-1334".
  • Figure 164 shows the amino acid sequence (SEQ JD NO:164) derived from the coding sequence of SEQ ID NO:163 shown in Figure 163.
  • Figure 165 shows a nucleotide sequence (SEQ JD NO : 165) of a native sequence PRO 1131 cDNA, wherein SEQ ID NO: 165 is a clone designated herein as "DNA59777-1480".
  • Figure 166 shows the amino acid sequence (SEQ JD NO: 166) derived from the coding sequence of SEQ ID NO: 165 shown in Figure 165.
  • Figure 167 shows a nucleotide sequence (SEQ JD NO: 167) of a native sequence PRO 1182 cDNA, wherein SEQ JD NO: 167 is a clone designated herein as "DNA59848-1512".
  • Figure 168 shows the amino acid sequence (SEQ ID NO: 168) derived from the coding sequence of SEQ
  • Figure 169 shows a nucleotide sequence (SEQ ID NO: 169) of a native sequence PROl 155 cDNA, wherein SEQ JD NO:169 is a clone designated herein as "DNA59849-1504".
  • Figure 170 shows the amino acid sequence (SEQ JD NO.T70) derived from the coding sequence of SEQ ID NO: 169 shown in Figure 169.
  • Figure 171 shows a nucleotide sequence (SEQ ID NO: 171) of a native sequence PRO 1186 cDNA, wherein SEQ ID NO:171 is a clone designated herein as "DNA60621-1516".
  • Figure 172 shows the amino acid sequence (SEQ JD NO: 172) derived from the coding sequence of SEQ ID NO: 171 shown in Figure 171.
  • Figure 173 shows anucleotide sequence (SEQ JDNO: 173) of anative sequence PROl 198 cDNA, wherein
  • SEQ TD NO-.173 is a clone designated herein as "DNA60622-1525".
  • Figure 174 shows the amino acid sequence (SEQ JD NO:174) derived from the coding sequence of SEQ ID NO: 173 shown in Figure 173.
  • Figure 175 shows anucleotide sequence (SEQ J NO: 175) of anative sequence PR01265 cDNA, wherein SEQ JD NO:175 is a clone designated herein as "DNA60764-1533".
  • Figure 176 shows the amino acid sequence (SEQ JD NO:176) derived from the coding sequence of SEQ J NO: 175 shown in Figure 175.
  • Figure 177 showsanucleotide sequence(SEQ ID NO:177) of anative sequence PR01361 cDNA, wherein SEQ ID NO:177 is a clone designated herein as "DNA60783-1611".
  • Figure 178 shows the amino acid sequence (SEQ JD NO:178) derived from the coding sequence of SEQ JD NO: 177 shown in Figure 177.
  • Figure 179 shows anucleotide sequence (SEQ IDNO:179) of anative sequence PR01287 cDNA, wherein
  • SEQ ID NO: 179 is a clone designated herein as "DNA61755- 1554".
  • Figure 180 shows the amino acid sequence (SEQ ID NO: 180) derived from the coding sequence of SEQ ID NO: 179 shown in Figure 179.
  • Figure 181 shows anucleotide sequence (SEQ ID NO: 181) of anative sequence PRO 1308 cDNA, wherein SEQ ID NO:181 is a clone designated herein as "DNA62306-1570".
  • Figure 182 shows the amino acid sequence (SEQ JD NO.T82) derived from the coding sequence of SEQ ID NO: 181 shown in Figure 181.
  • Figure 183 shows a nucleotide sequence (SEQ ID NO: 183) of a native sequence PR04313 cDNA, wherein SEQ JD NO: 183 is a clone designated herein as "DNA62312-2558".
  • Figure 184 shows the amino acid sequence (SEQ ID NO: 184) derived from the coding sequence of SEQ
  • Figure 185 shows anucleotide sequence (SEQ JD NO: 185) of anative sequence PROl 192 cDNA, wherein SEQ ID NO:185 is a clone designated herein as "DNA62814-1521".
  • Figure 186 shows the amino acid sequence (SEQ ID NO:186) derived from the coding sequence of SEQ ID NO: 185 shown in Figure 185.
  • Figure 187 shows a nucleotide sequence (SEQ ID NO: 187) of a native sequence PRO 1160 cDNA, wherein SEQ ID NO:187 is a clone designated herein as "DNA62872-1509".
  • Figure 188 shows the amino acid sequence (SEQ ID NO: 188) derived from the coding sequence of SEQ ID NO: 187 shown in Figure 187.
  • Figure 189 shows a nucleotide sequence (SEQ ID NO: 189) of a native sequence PRO 1244 cDNA, wherein
  • SEQ ID NO:189 is a clone designated herein as "DNA64883-1526".
  • Figure 190 shows the amino acid sequence (SEQ ID NO: 190) derived from the coding sequence of SEQ ID NO: 189 shown in Figure 189.
  • Figure 191 shows a nucleotide sequence (SEQ ID NO: 191) of a native sequence PRO 1356 cDNA, wherein SEQ ID NO:191 is a clone designated herein as "DNA64886-1601".
  • Figure 192 shows the amino acid sequence (SEQ ID NO: 192) derived from the coding sequence of SEQ ID NO:191 shown in Figure 191.
  • Figure 193 shows a nucleotide sequence (SEQ JD NO: 193) of a native sequence PRO 1274 cDNA, wherein SEQ ID NO:193 is a clone designated herein as "DNA64889-1541".
  • Figure 194 shows the amino acid sequence (SEQ JD NO:194) derived from the coding sequence of SEQ
  • Figure 195 shows anucleotide sequence (SEQ JD NO: 195) of anative sequence PRO 1272 cDNA, wherein SEQ JD NO: 195 is a clone designated herein as "DNA64896-1539".
  • Figure 196 shows the amino acid sequence (SEQ JD NO: 196) derived from the coding sequence of SEQ ID NO: 195 shown in Figure 195.
  • Figure 197 shows anucleotide sequence (SEQ JD NO: 197) of anative sequence PR01412 cDNA, wherein SEQ ID NO.T97 is a clone designated herein as "DNA64897-1628".
  • Figure 198 shows the amino acid sequence (SEQ JD NO:198) derived from the coding sequence of SEQ ID NO: 197 shown in Figure 197.
  • Figure 199 shows anucleotide sequence (SEQ IDNO:199) of anative sequence PROl 286 cDNA, wherein SEQ JD NO: 199 is a clone designated herein as "DNA64903-1553".
  • Figure 200 shows the amino acid sequence (SEQ ID NO:200) derived from the coding sequence of SEQ
  • Figure 201 shows a nucleotide sequence (SEQ ID NO:201) of a native sequence PRO 1347 cDNA, wherein SEQ JD NO:201 is a clone designated herein as "DNA64950-1590".
  • Figure 202 shows the amino acid sequence (SEQ ID NO:202) derived from the coding sequence of SEQ ID NO:201 shown in Figure 201.
  • Figure 203 shows anucleotide sequence (SEQ JD NO:203) of anative sequence PR01273 cDNA, wherein SEQ ID NO:203 is a clone designated herein as "DNA65402-1540".
  • Figure 204 shows the amino acid sequence (SEQ ID NO:204) derived from the coding sequence of SEQ JD NO:203 shown in Figure 203.
  • Figure 205 shows a nucleotide sequence (SEQ JD NO:205) of anative sequence PR01283 cDNA, wherein
  • SEQ JD NO:205 is a clone designated herein as "DNA65404-15 1".
  • Figure 206 shows the amino acid sequence (SEQ JD NO:206) derived from the coding sequence of SEQ ID NO:205 shown in Figure 205.
  • Figure 207 shows a nucleotide sequence (SEQ ID NO:207) of a native sequence PRO 1279 cDNA, wherein SEQ ID NO:207 is a clone designated herein as "DNA65405-1547".
  • Figure 208 shows the amino acid sequence (SEQ ID NO:208) derived from the coding sequence of SEQ ID NO:207 shown in Figure 207.
  • Figure 209 shows anucleotide sequence (SEQ JDNO:209) of anative sequence PRO1306 cDNA, wherein SEQ JD NO:209 is a clone designated herein as "DNA65410-1569".
  • Figure 210 shows the amino acid sequence (SEQ JD NO:210) derived from the coding sequence of SEQ
  • Figure 211 shows anucleotide sequence (SEQIDNO:211) of anative sequence PROl 195 cDNA, wherein SEQ JD NO:211 is a clone designated herein as "DNA65412-1523".
  • Figure 212 shows the amino acid sequence (SEQ JD NO:212) derived from the coding sequence of SEQ ID NO:211 shown in Figure 211.
  • Figure 213 shows anucleotide sequence (SEQ IDNO:213) of anative sequence PR04995 cDNA, wherein SEQ JD NO:213 is a clone designated herein as "DNA66307-2661".
  • Figure 214 shows the amino acid sequence (SEQ JD NO:214) derived from the coding sequence of SEQ JD NO:213 shown in Figure 213.
  • Figure 215 shows a nucleotide sequence (SEQ ID NO:215) of a native sequence PRO 1382 cDNA, wherein SEQ TD NO:215 is a clone designated herein as "DNA66526-1616".
  • Figure 216 shows the amino acid sequence (SEQ ID NO:216) derived from the coding sequence of SEQ
  • Figure 217 shows a nucleotide sequence (SEQ JD NO:217) of a native sequence PRO 1325 cDNA, wherein SEQ ID NO:217 is a clone designated herein as "DNA66659-1593".
  • Figure 218 shows the amino acid sequence (SEQ ID NO:218) derived from the coding sequence of SEQ J NO:217 shown in Figure 217.
  • Figure 219 shows a nucleotide sequence (SEQ ID NO:219) of a native sequence PRO 1329 cDNA, wherein SEQ ID NO.-219 is a clone designated herein as "DNA66660-1585".
  • Figure 220 shows the amino acid sequence (SEQ ID NO:220) derived from the coding sequence of SEQ ID NO:219 shown in Figure 219.
  • Figure 221 shows a nucleotide sequence (SEQ JD NO:221 ) of a native sequence PRO 1338 cDNA, wherein
  • SEQ JD NO:221 is a clone designated herein as "DNA66667-1596".
  • Figure 222 shows the amino acid sequence (SEQ JD NO:222) derived from the coding sequence of SEQ ID NO:221 shown in Figure 221.
  • Figure 223 shows anucleotide sequence (SEQ IDNO:223) of anative sequence PR01337 cDNA, wherein SEQ JD NO:223 is a clone designated herein as "DNA66672-1586".
  • Figure 224 shows the amino acid sequence (SEQ JD NO:224) derived from the coding sequence of SEQ JD NO:223 shown in Figure 223.
  • Figure 225 shows a nucleotide sequence (SEQ ID NO:225) of a native sequence PRO 1343 cDNA, wherein SEQ JD NO:225 is a clone designated herein as "DNA66675-1587".
  • Figure 226 shows the amino acid sequence (SEQ JD NO:226) derived from the coding sequence of SEQ
  • Figure 227 shows anucleotide sequence (SEQ JDNO:227) of anative sequence PR01376 cDNA, wherein SEQ JD NO:227 is a clone designated herein as "DNA67300-1605".
  • Figure 228 shows the amino acid sequence (SEQ ID NO:228) derived from the coding sequence of SEQ ID NO:227 shown in Figure 227.
  • Figure 229 shows a nucleotide sequence (SEQ ID NO:229) of a native sequence PR01434 cDNA, wherein SEQ JD N0:229 is a clone designated herein as "DNA68818-2536".
  • Figure 230 shows the amino acid sequence (SEQ JD NO:230) derived from the coding sequence of SEQ ID NO:229 shown in Figure 229.
  • Figure 231 shows a nucleotide sequence (SEQ ID N0:231) of anative sequence PR03579 cDNA, wherein
  • SEQ JD NO:231 is a clone designated herein as "DNA68862-2546".
  • Figure 232 shows the amino acid sequence (SEQ ID NO:232) derived from the coding sequence of SEQ ID NO:231 shown in Figure 231.
  • Figure 233 shows anucleotide sequence (SEQ JDNO-.233) of anative sequence PR01387 cDNA, wherein SEQ ID NO:233 is a clone designated herein as "DNA68872-1620".
  • Figure 234 shows the amino acid sequence (SEQ ID NO:234) derived from the coding sequence of SEQ JD NO:233 shown in Figure 233.
  • Figure 235 shows a nucleotide sequence (SEQ JD NO:235) of a native sequence PRO 1419 cDNA, wherein SEQ ID NO:235 is a clone designated herein as "DNA71290-1630".
  • Figure 236 shows the amino acid sequence (SEQ ID NO:236) derived from the coding sequence of SEQ ID NO:235 shown in Figure 235.
  • Figure 237 shows anucleotide sequence (SEQ IDNO:237) of anative sequence PRO 1488 cDNA, wherein
  • SEQ ID NO:237 is a clone designated herein as "DNA73736-1657”.
  • Figure 238 shows the amino acid sequence (SEQ ID NO:238) derived from the coding sequence of SEQ ID NO:237 shown in Figure 237.
  • Figure 239 shows a nucleotide sequence (SEQ ID NO:239) of a native sequence PR01474 cDNA, wherein SEQ JD NO:239 is a clone designated herein as "DNA73739-1645".
  • Figure 240 shows the amino acid sequence (SEQ JD NO:240) derived from the coding sequence of SEQ ID NO:239 shown in Figure 239.
  • Figure 241 shows a nucleotide sequence (SEQ JD NO:241 ) of a native sequence PRO 1508 cDNA, wherein SEQ ID NO:241 is a clone designated herein as "DNA73742-1662".
  • Figure 242 shows the amino acid sequence (SEQ ID NO:242) derived from the coding sequence of SEQ
  • Figure 243 shows a nucleotide sequence (SEQ ID NO:243) of a native sequence PRO 1754 cDNA, wherein SEQ ID NO:243 is a clone designated herein as "DNA76385-1692".
  • Figure 244 shows the ammo acid sequence (SEQ ID NO:244) derived from the coding sequence of SEQ JD NO:243 shown in Figure 243.
  • Figure 245 shows a nucleotide sequence (SEQ JD NO :245) of a native sequence PRO 1550 cDNA, wherein SEQ ID NO:245 is a clone designated herein as "DNA76393-1664".
  • Figure 246 shows the amino acid sequence (SEQ JD NO:246) derived from the coding sequence of SEQ JD NO:245 shown in Figure 245.
  • Figure 247 shows a nucleotide sequence (SEQ ID NO:247) of a native sequence PRO 1758 cDNA, wherein
  • SEQ ID NO:247 is a clone designated herein as "DNA76399-1700".
  • Figure 248 shows the amino acid sequence (SEQ JD NO:248) derived from the coding sequence of SEQ ID NO:247 shown in Figure 247.
  • Figure 249 shows a nucleotide sequence (SEQ ID NO:249) of a native sequence PRO 1917 cDNA, wherein SEQ JD NO:249 is a clone designated herein as "DNA76400-2528".
  • Figure 250 shows the amino acid sequence (SEQ JD NO:250) derived from the coding sequence of SEQ JD NO:249 shown in Figure 249.
  • Figure 251 shows a nucleotide sequence (SEQ ID NO:251) of a native sequence PRO 1787 cDNA, wherein SEQ ID NO:251 is a clone designated herein as "DNA76510-2504".
  • Figure 252 shows the amino acid sequence (SEQ ID NO:252) derived from the coding sequence of SEQ ID NO-.251 shown in Figure 251.
  • Figure 253 shows a nucleotide sequence (SEQ JD NO:253) of a native sequence PROl 556 cDNA, wherein
  • SEQ ID NO:253 is a clone designated herein as "DNA76529-1666".
  • Figure 254 shows the amino acid sequence (SEQ ID NO:254) derived from the coding sequence of SEQ ID NO:253 shown in Figure 253.
  • Figure 255 shows anucleotide sequence (SEQ JD NO:255) of anative sequence PRO 1760 cDNA, wherein SEQ ID NO:255 is a clone designated herein as "DNA76532-1702".
  • Figure 256 shows the amino acid sequence (SEQ ID NO:256) derived from the coding sequence of SEQ ID NO:255 shown in Figure 255.
  • Figure 257 shows a nucleotide sequence (SEQ ID NO:257) of a native sequence PR01567 cDNA, wherein SEQ ID NO:257 is a clone designated herein as "DNA76541-1675".
  • Figure 258 shows the amino acid sequence (SEQ ID NO:258) derived from the coding sequence of SEQ
  • Figure 259 shows a nucleotide sequence (SEQ JD NO:259) of a native sequence PRO 1600 cDNA, wherein SEQ JD NO:259 is a clone designated herein as "DNA77503-1686".
  • Figure 260 shows the amino acid sequence (SEQ ID NO:260) derived from the coding sequence of SEQ JD NO:259 shown in Figure 259.
  • Figure 261 shows anucleotide sequence (SEQ ID NO:261) of anative sequence PR01868 cDNA, wherein SEQ JD NO:261 is a clone designated herein as "DNA77624-2515".
  • Figure 262 shows the amino acid sequence (SEQ ID NO:262) derived from the coding sequence of SEQ JD NO.-261 shown in Figure 261.
  • Figure 263 shows a nucleotide sequence (SEQ JD NO:263) of a native sequence PROl 890 cDNA, wherein
  • SEQ ID NO:263 is a clone designated herein as "DNA79230-2525".
  • Figure 264 shows the amino acid sequence (SEQ ID NO:264) derived from the coding sequence of SEQ ID NO:263 shown in Figure 263.
  • Fi ure 265 shows anucleotide sequence (SEQ ID O:265) of anative sequence PROl 887 cDNA, wherein SEQ JD NO:265 is a clone designated herein as "DNA79862-2522".
  • Figure 266 shows the amino acid sequence (SEQ ID NO:265) derived from the coding sequence of SEQ JD NO:265 shown in Figure 265.
  • Figure 267 shows a nucleotide sequence (SEQ JD NO:267) of anative sequence PR04353 cDNA, wherein SEQ ID NO:267 is a clone designated herein as "DNA80145-2594".
  • Figure 268 shows the amino acid sequence (SEQ ID NO:268) derived from the coding sequence of SEQ
  • Figure269 shows anucleotide sequence (SEQ IDNO:269) of anative sequence PRO 1782 cDNA, wherein SEQ ED NO-.269 is a clone designated herein as "DNA80899-2501".
  • Figure 270 shows the amino acid sequence (SEQ ID NO:270) derived from the coding sequence of SEQ ID NO:269 shown in Figure 269.
  • Figure 271 shows anucleotide sequence (SEQ ID O:271) of anative sequence PR01928 cDNA, wherein SEQ ID NO:271 is a clone designated herein as "DNA81754-2532".
  • Figure 272 shows the amino acid sequence (SEQ ID NO:272) derived from the coding sequence of SEQ ID NO-.271 shown in Figure 271.
  • Figure 273 shows a nucleotide sequence (SEQ ID NO:273) of a native sequence PROl 865 cDNA, wherein SEQ JD NO:273 is a clone designated herein as "DNA81757-2512".
  • Figure 274 shows the amino acid sequence (SEQ JD NO:274) derived from the coding sequence of SEQ
  • JD NO:273 shown in Figure 273.
  • Figure 275 shows a nucleotide sequence (SEQ JD NO:275) of a native sequence PR04341 cDNA, wherein SEQ JD NO:275 is a clone designated herein as "DNA81761-2583".
  • Figure 276 shows the amino acid sequence (SEQ ID NO:276) derived from the coding sequence of SEQ JD NO:275 shown in Figure 275.
  • Figure 277 shows anucleotide sequence (SEQ IDNO:277) of anative sequence PR06714 cDNA, wherein SEQ JD NO:277 is a clone designated herein as "DNA82358-2738".
  • Figure 278 shows the amino acid sequence (SEQ ID NO:278) derived from the coding sequence of SEQ ID NO:277 shown in Figure 277.
  • Figure 279 shows a nucleotide sequence (SEQ JD NO:279) of anative sequence PR05723 cDNA, wherein
  • SEQ ID NO:279 is a clone designated herein as "DNA82361".
  • Figure 280 shows the amino acid sequence (SEQ JD NO:280) derived from the coding sequence of SEQ JD NO.-279 shown in Figure 279.
  • Figure 281 shows anucleotide sequence (SEQ JD NO:281) of anative sequence PR03438 cDNA, wherein SEQ ID NO:281 is a clone designated herein as "DNA82364-2538".
  • Figure 282 shows the amino acid sequence (SEQ JD NO:282) derived from the coding sequence of SEQ JD NO:281 shown in Figure 281.
  • Figure 283 shows a nucleotide sequence (SEQ ID NO:283) of anative sequence PRO6071 cDNA, wherein SEQ JD NO:283 is a clone designated herein as "DNA82403-2959".
  • Figure 284 shows the amino acid sequence (SEQ ID NO:284) derived from the coding sequence of SEQ
  • JD NO:283 shown in Figure 283.
  • Figure 285 shows a nucleotide sequence (SEQ ID NO:285) of a native sequence PROl 801 cDNA, wherein SEQ JD NO:285 is a clone designated herein as "DNA83500-2506".
  • Figure 286 shows the amino acid sequence (SEQ JD NO:286) derived from the coding sequence of SEQ JD NO-.285 shown in Figure 285.
  • Figure287 shows a nucleotide sequence (SEQ JD NO:287) ofa native sequence PR04324cDNA, wherein SEQ JD NO:287 is a clone designated herein as "DNA83560-2569".
  • Figure 288 shows the amino acid sequence (SEQ ID NO:288) derived from the coding sequence of SEQ JD NO:287 shown in Figure 287.
  • Figure 289 shows anucleotide sequence (SEQ IDNO:289) of anative sequence PR04333 cDNA, wherein SEQ ID NO-.289 is a clone designated herein as "DNA84210-2576".
  • Figure 290 shows the amino acid sequence (SEQ ID NO:290) derived from the coding sequence of SEQ
  • Figure 291 shows anucleotide sequence (SEQ JDNO:291) of anative sequence PRO4405 cDNA, wherein SEQ ID NO:291 is a clone designated herein as "DNA84920-2614".
  • Figure 292 shows the amino acid sequence (SEQ ID NO:292) derived from the coding sequence of SEQ JD NO:291 shown in Figure 291.
  • Figure 293 shows a nucleotide sequence (SEQ ID NO:293) of a native sequence PR04356 cDNA, wherein SEQ ID N0:293 is a clone designated herein as "DNA86576-2595".
  • Figure 294 shows the amino acid sequence (SEQ ID NO:294) derived from the coding sequence of SEQ ID NO:293 shown in Figure 293.
  • Figure 295 shows a nucleotide sequence (SEQ ID NO:295) of a native sequence PR03444 cDNA, wherein
  • SEQ ID NO:295 is a clone designated herein as "DNA87997".
  • Figure 296 shows the amino acid sequence (SEQ ID NO:296) derived from the coding sequence of SEQ ID NO:295 shown in Figure 295.
  • Figure 297 shows anucleotide sequence (SEQ JD NO:297) of anative sequence PRO4302 cDNA, wherein SEQ ID NO:297 is a clone designated herein as "DNA92218-2554”. •
  • Figure 298 shows the amino acid sequence (SEQ ID NO:298) derived from the coding sequence of SEQ ID NO:297 shown in Figure 297.
  • Figure 299 shows a nucleotide sequence (SEQ ID NO:299) of a native sequence PR04371 cDNA, wherein SEQ JD N0:299 is a clone designated herein as "DNA92233-2599".
  • Figure 300 shows the amino acid sequence (SEQ ID NO:300) derived from the coding sequence of SEQ
  • Figure 301 shows anucleotide sequence (SEQIDNO:301) of anative sequence PR04354 cDNA, wherein SEQ ID NO:301 is a clone designated herein as "DNA92256-2596".
  • Figure 302 shows the amino acid sequence (SEQ ID NO:302) derived from the coding sequence of SEQ ID NO:301 shown in Figure 301.
  • Figure 303 shows a nucleotide sequence (SEQ ID NO:303) of a native sequence PR05725 cDNA, wherein SEQ JD NO:303 is a clone designated herein as "DNA92265-2669".
  • Figure 304 shows the amino acid sequence (SEQ ID NO:304) derived from the coding sequence of SEQ JD NO.-303 shown in Figure 303.
  • Figure 305 shows anucleotide sequence (SEQ JD NO: 305) of anative sequence PRO4408 cDNA, wherein
  • SEQ ID NO:305 is a clone designated herein as "DNA92274-2617".
  • Figure 306 shows the amino acid sequence (SEQ JD NO:306) derived from the coding sequence of SEQ JD NO:305 shown in Figure 305.
  • Figure 307 shows a nucleotide sequence (SEQ JD NO:307) of a native sequence PRO9940 cDNA, wherein SEQ ID NO:307 is a clone designated herein as "DNA92282".
  • Figure 308 shows the amino acid sequence (SEQ ID NO:308) derived from the coding sequence of SEQ JD NO:307 shown in Figure 307.
  • Figure 309 shows anucleotide sequence (SEQ JD NO:309) of a native sequence PR05737 cDNA, wherein SEQ ID NO:309 is a clone designated herein as "DNA92929-2534-1".
  • Figure 310 shows the amino acid sequence (SEQ ID NO:310) derived from the coding sequence of SEQ ID NO:309 shown in Figure 309.
  • Figure 311 shows a nucleotide sequence (SEQ ID NO:311) of a native sequence PR04425 cDNA, wherein
  • SEQ ID NO:311 is a clone designated herein as "DNA93011-2637".
  • Figure 312 shows the amino acid sequence (SEQ ID NO:312) derived from the coding sequence of SEQ JD NO:311 shown in Figure 311.
  • Figure313 shows anucleotide sequence (SEQ JDNO:313) of anative sequence PR04345 cDNA, wherein SEQ JD NO:313 is a clone designated herein as "DNA94854-2586".
  • Figure 314 shows the amino acid sequence (SEQ ID NO:314) derived from the coding sequence of SEQ JD NO.-313 shown in Figure 313.
  • Figure 315 shows a nucleotide sequence (SEQ ID NO:315) of a native sequence PR04342 cDNA, wherein SEQ JD NO:315 is a clone designated herein as "DNA96787-2534-1".
  • Figure 316 shows the amino acid sequence (SEQ JD NO:316) derived from the coding sequence of SEQ
  • Figure 317 shows a nucleotide sequence (SEQ JD NO:317) of a native sequence PR03562 cDNA, wherein SEQ ID NO:317 is a clone designated herein as "DNA96791".
  • Figure 318 shows the amino acid sequence (SEQ ID NO:318) derived from the coding sequence of SEQ ID NO:317 shown in Figure 317.
  • Figure 319 shows a nucleotide sequence (SEQ JD NO:319) of a native sequence PR04422 cDNA, wherein SEQ ID NO-.319 is a clone designated herein as "DNA96867-2620".
  • Figure 320 shows the amino acid sequence (SEQ ID NO:320) derived from the coding sequence of SEQ ID NO:319 shown in Figure 319.
  • Figure 321 shows a nucleotide sequence (SEQ JD NO:321 ) of a native sequence PR05776 cDNA, wherein
  • SEQ JD N0:321 is a clone designated herein as "DNA96872-2674".
  • Figure 322 shows the amino acid sequence (SEQ ID NO:322) derived from the coding sequence of SEQ JD NO:321 shown in Figure 321.
  • Figure 323 shows a nucleotide sequence (SEQ JD NO:323) of a native sequence PRO4430 cDNA, wherein SEQ JD NO:323 is a clone designated herein as "DNA96878-2626".
  • Figure 324 shows the amino acid sequence (SEQ ID NO:324) derived from the coding sequence of SEQ JD NO:323 shown in Figure 323.
  • Figure 325 shows anucleotide sequence (SEQ J NO:325) of anative sequence PR04499 cDNA, wherein SEQ JD NO:325 is a clone designated herein as "DNA96889-2641".
  • Figure 326 shows the amino acid sequence (SEQ JD NO:326) derived from the coding sequence of SEQ ID NO:325 shown in Figure 325.
  • Figure 327 shows a nucleotide sequence (SEQ ID NO:327) of a native sequence PRO4503 cDNA, wherein
  • SEQ ID NO:327 is a clone designated herein as "DNA100312-2645".
  • Figure 328 shows the amino acid sequence (SEQ ID NO:328) derived from the coding sequence of SEQ ID NO:327 shown in Figure 327.
  • Figure 329 shows a nucleotide sequence (SEQ ID NO:329) of a native sequence PRO10008 cDNA, wherein SEQ ID NO:329 is a clone designated herein as "DNA101921".
  • Figure 330 shows the amino acid sequence (SEQ JD NO:330) derived from the coding sequence of SEQ JD NO:329 shown in Figure 329.
  • Figure 331 shows anucleotide sequence (SEQ JD NO:331) of a native sequence PRO5730 cDNA, wherein SEQ JD NO:331 is a clone designated herein as "DNA101926".
  • Figure 332 shows the amino acid sequence (SEQ ID NO:332) derived from the coding sequence of SEQ
  • Figure 333 shows anucleotide sequence (SEQ JD NO:333) of anative sequence PRO6008 cDNA, wherein SEQ JD NO:333 is a clone designated herein as "DNA102844".
  • Figure 334 shows the amino acid sequence (SEQ JD NO:334) derived from the coding sequence of SEQ ID NO:333 shown in Figure 333.
  • Figure 335 shows a nucleotide sequence (SEQ ID NO : 335) of a native sequence PR04527 cDNA, wherein SEQ ID NO:335 is a clone designated herein as "DNA103197".
  • Figure 336 shows the amino acid sequence (SEQ ID NO:336) derived from the coding sequence of SEQ ID NO:335 shown in Figure 335.
  • Figure 337 shows a nucleotide sequence (SEQ JD NO:337) of anative sequence PR04538 cDNA, wherein
  • SEQ ID NO:337 is a clone designated herein as "DNA103208".
  • Figure 338 shows the amino acid sequence (SEQ JD NO:338) derived from the coding sequence of SEQ JD NO:337 shown in Figure 337.
  • Figure 339 shows a nucleotide sequence (SEQ JD NO:339) of a native sequence PR04553 cDNA, wherein SEQ ID NO:339 is a clone designated herein as "DNA103223".
  • Figure 340 shows the amino acid sequence (SEQ ID NO:340) derived from the coding sequence of SEQ ID NO:339 shown in Figure 339.
  • Figure 341 shows a nucleotide sequence (SEQ JD NO:341 ) of a native sequence PRO6006 cDNA, wherein SEQ JD NO:341 is a clone designated herein as "DNA105782-2693".
  • Figure 342 shows the amino acid sequence (SEQ ID NO:342) derived from the coding sequence of SEQ
  • Figure 343 shows a nucleotide sequence (SEQ JD NO:343) of a native sequence PRO6029 cDNA, wherein SEQ ID NO:343 is a clone designated herein as "DNA105849-2704".
  • Figure 344 shows the amino acid sequence (SEQ ID NO:344) derived from the coding sequence of SEQ JD NO:343 shown in Figure 343.
  • Figure 345 shows a nucleotide sequence (SEQ JD NO:345) of a native sequence PR09821 cDNA, wherein SEQ ID NO:345 is a clone designated herein as "DNA108725-2766".
  • Figure 346 shows the amino acid sequence (SEQ ID NO:346) derived from the coding sequence of SEQ ID NO:345 shown in Figure 345.
  • Figure 347 shows a nucleotide sequence (SEQ ID NO:347) of a native sequence PRO9820 cDNA, wherein SEQ ID NO:347 is a clone designated herein as "DNA108769-2765".
  • Figure 348 shows the amino acid sequence (SEQ JD NO:348) derived from the coding sequence of SEQ
  • Figure 349 shows a nucleotide sequence (SEQ JD NO:349) of anative sequence PR09771 cDNA, wherein SEQ ID N0:349 is a clone designated herein as "DNA119498-2965".
  • Figure 350 shows the amino acid sequence (SEQ JD NO:350) derived from the coding sequence of SEQ ID NO:349 shown in Figure 349.
  • Figure 351 shows anucleotide sequence (SEQ ID NO:351) of anative sequence PR07436 cDNA, wherein SEQ ID NO:351 is a clone designated herein as "DNA119535-2756".
  • Figure 352 shows the amino acid sequence (SEQ ID NO:352) derived from the coding sequence of SEQ ID NO:351 shown in Figure 351.
  • Figure 353 shows a nucleotide sequence (SEQ ID NO:353) of a native sequence PRO10096 cDNA, wherein SEQ ID NO:353 is a clone designated herein as "DNA125185-2806".
  • Fi ure 354 shows the amino acid sequence (SEQ ID NO:354) derived from the coding sequence of SEQ JD NO:353 shown in Figure 353.
  • Figure 355 shows a nucleotide sequence (SEQ JD NO:355) of a native sequence PRO19670 cDNA, wherein SEQ ID NO:355 is a clone designated herein as "DNA131639-2874".
  • Figure 356 shows the amino acid sequence (SEQ ID NO:356) derived from the coding sequence of SEQ ID NO:355 shown in Figure 355.
  • Figure 357 shows a nucleotide sequence (SEQ ID NO:357) of a native sequence PRO20044 cDNA, wherein SEQ ED NO:357 is a clone designated herein as "DNA139623-2893".
  • Figure 358 shows the amino acid sequence (SEQ ID NO:358) derived from the coding sequence of SEQ
  • JD NO:357 shown in Figure 357.
  • Figure 359 shows anucleotide sequence (SEQ JD NO:359) of anative sequence PR09873 cDNA, wherein SEQ ID NO:359 is a clone designated herein as "DNA143076-2787".
  • Figure 360 shows the amino acid sequence (SEQ JD NO:360) derived from the coding sequence of SEQ ID NO:359 shown in Figure 359.
  • Figure 361 shows a nucleotide sequence (SEQ ID NO:361) of a native sequence PR021366 cDNA, wherein SEQ ID NO:361 is a clone designated herein as "DNA143276-2975".
  • Figure 362 shows the amino acid sequence (SEQ JD NO:362) derived from the coding sequence of SEQ ID NO:361 shown in Figure 361.
  • Figure 363 shows a nucleotide sequence (SEQ ID NO:363) of a native sequence PRO20040 cDNA, wherein SEQ ID NO:363 is a clone designated herein as "DNA164625-2890".
  • Figure 364 shows the amino acid sequence (SEQ ID NO:364) derived from the coding sequence of SEQ
  • Figure 365 shows a nucleotide sequence (SEQ ID NO:365) of a native sequence PR021184 cDNA, wherein SEQ ID NO:365 is a clone designated herein as "DNA167678-2963".
  • Figure 366 shows the amino acid sequence (SEQ ID NO:366) derived from the coding sequence of SEQ ID NO:365 shown in Figure 365.
  • Figure 367 shows a nucleotide sequence (SEQ ID NO:367) of a native sequence PRO21055 cDNA, wherein SEQ ID NO:367 is a clone designated herein as "DNA170021-2923".
  • Figure 368 shows the amino acid sequence (SEQ ID NO:368) derived from the coding sequence of SEQ ID NO:367 shown in Figure 367.
  • Figure 369 shows a nucleotide sequence (SEQ ID NO:369) of a native sequence PR028631 cDNA, wherein SEQ ID NO:369 is a clone designated herein as "DNA170212-3000".
  • Figure 370 shows the amino acid sequence (SEQ ID NO:370) derived from the coding sequence of SEQ ID NO:369 shown in Figure 369.
  • Figure 371 shows a nucleotide sequence (SEQ ID NO:371) of a native sequence PR021384 cDNA, wherein SEQ JD NO:371 is a clone designated herein as "DNA177313-2982".
  • Figure 372 shows the amino acid sequence (SEQ JD NO:372) derived from the coding sequence of SEQ ED NO:371 shown in Figure 371.
  • Figure 373 shows a nucleotide sequence (SEQ ID NO:373) of a native sequence PRO 1449 cDNA, wherein SEQ ID NO:373 is a clone designated herein as "DNA64908-1163-1".
  • Figure 374 shows the amino acid sequence (SEQ ID NO:374) derived from the coding sequence of SEQ
  • Figure 375 shows wholemount in situ hybridization results on mouse embryos using a mouse orthologue of PR01449 whichhas about 78% amino acid identity with PRO 1449. The results show thatPR01449 orthologue is expressed in the developing vasculature. The cross-section further shows expression in endothelial cells and progenitors of endothelial cells.
  • Figure 376 shows that a PR01449 orthologue having about 78% amino acid identity with PR01449 is expressed in vasculature of many inflamed and diseased tissues, but is very low, or lacking, in normal adult vessels.
  • Figure 377 shows that a PR01449 orthologue having about 78% amino acid identity with PR01449 induces ectopic vessels in the eyes of chicken embryos. 5. Detailed Description ofthe Invention
  • cardiovascular, endothelial and angiogenic disorder cardiac, endothelial and angiogenic disorder
  • cardiac, endothelial and angiogenic dysfunction cardiac, endothelial or angiogenic disorder
  • cardiovascular, endothelial or angiogenic dysfunction cardiac, endothelial or angiogenic dysfunction
  • cardiac, endothelial or angiogenic dysfunction refers in part to systemic disorders that affect vessels, such as diabetes mellitus, as well as diseases ofthe vessels themselves, such as ofthe arteries, capillaries, veins, and/or lymphatics. This would include indications that stimulate angiogenesis and or cardiovascularization, and those that inhibit angiogenesis and/or cardiovascularization.
  • Such disorders include, for example, arterial disease, such as atherosclerosis, hypertension, inflammatory vasculitides, Reynaud's disease and Reynaud's phenomenon, aneurysms, and arterial restenosis; venous and lymphatic disorders such as thrombophlebitis, lymphangitis, and lymphedema; and other vascular disorders such as peripheral vascular disease, cancer such as vascular tumors, e.g.
  • hemangioma capillary and cavernous
  • glomus tumors telangiectasia
  • bacillary angiomatosis hemangioendothelioma
  • angiosarcoma haemangiopericytoma
  • Kaposi's sarcoma lymphangioma
  • lymphangiosarcoma tumor angiogenesis
  • trauma such as wounds, bu s, and other injured tissue
  • implant fixation scarring, ischemia reperfusion injury, rheumatoid arthritis
  • cerebrovascular disease renal diseases such as acute renal failure, and osteoporosis.
  • renal diseases such as acute renal failure, and osteoporosis.
  • “Hypertrophy”, as used herein, is defined as an increase in mass of an organ or structure independent of natural growth that does not involve tumor formation. Hypertrophy of an organ or tissue is due either to an increase in the mass ofthe individual cells (true hypertrophy), or to an increase in the number of cells making up the tissue
  • cardiac hypertrophy is defined as an increase in mass of the heart, which, in adults, is characterized by an increase in myocyte cell size and contractile protein content without concomitant cell division.
  • the character ofthe stress responsible for inciting the hypertrophy (e.g., increased preload, increased afterload, loss of myocytes, as in myocardial infarction, or primary depression of contractility), appears to play a critical role in determining the nature ofthe response.
  • the early stage of cardiac hypertrophy is usually characterized morphologically by increases in the size of myofibrils and mitochondria, as well as by enlargement of mitochondria and nuclei.
  • cardiac hypertrophy is used to include all stages ofthe progression of this condition, characterized by various degrees of structural damage ofthe heart muscle, regardless ofthe underlying cardiac disorder. Hence, the term also includes physiological conditions instrumental in the development of cardiac hypertrophy, such as elevated blood pressure, aortic stenosis, or myocardial infarction.
  • Heart failure refers to an abnormality of cardiac function where the heart does not pump blood at the rate needed for the requirements of metabolizing tissues.
  • the heart failure can be caused by a number of factors, including ischemic, congenital, rheumatic, or idiopathic forms.
  • CHF Congestive heart failure
  • Myocardial infarction generally results from atherosclerosis of the coronary arteries, often with superimposed coronary thrombosis. It may be divided into two major types: transmural infarcts, in which myocardial necrosis involves the full thickness ofthe ventricular wall, and subendocardial (nontransmural) infarcts, in which the necrosis involves the subendocardium, the intramural myocardium, or both, without extending all the way through the ventricular wall to the epicardium. Myocardial infarction is known to cause both a change in hemodynamic effects and an alteration in structure in the damaged and healthy zones of the heart.
  • myocardial infarction reduces the maximum cardiac output and the stroke volume of the heart. Also associated with myocardial infarction is a stimulation of the DNA synthesis occurring in the interstice as well as an increase in the formation of collagen in the areas of the heart not affected.
  • cardiac hypertrophy has long been associated with "hypertension".
  • a characteristic ofthe ventricle that becomes hypertrophic as a result of chronic pressure overload is an impaired diastolic performance. Fouad e/ ⁇ /., J. Am. Coll. Cardiol..4: 1500-1506 (1984); Smith etal, J. Am. Coll. Cardiol.,
  • hypotrophic cardiomyopathy Another complex cardiac disease associated with cardiac hypertrophy is "hypertrophic cardiomyopathy”. This condition is characterized by a great diversity of morphologic, functional, and clinical features (Maron et al, N. Engl. J. Med.. 316: 780-789 (1987); Spirito et al, N. Engl. J. Med.. 320: 749-755 (1989); Louie and Edwards,
  • Supravalvular "aortic stenosis” is an inherited vascular disorder characterized by narrowing of the ascending aorta, but other arteries, including the pulmonary arteries, may also be affected. Untreated aortic stenosis may lead to increased intracardiac pressure resulting in myocardial hypertrophy and eventually heart failure and death. The pathogenesis of this disorder is not fully understood, but hypertrophy and possibly hyperplasia of medial smooth muscle are prominent features of this disorder. It has been reported that molecular variants ofthe elastin gene are involved in the development and pathogenesis of aortic stenosis. U.S. Patent No. 5,650,282 issued July 22, 1997.
  • Valvular regurgitation occurs as a result of heart diseases resulting in disorders ofthe cardiac valves.
  • Various diseases like rheumatic fever, can cause the shrinking or pulling apart of the valve orifice, while otlier diseases may result in endocarditis, an inflammation ofthe endocardium or lining membrane ofthe atrioventricular orifices and operation ofthe heart.
  • Pefects such as the narrowing ofthe valve stenosis or the defective closing of the valve result in an accumulation of blood in the heart cavity or regurgitation of blood past the valve. If uncorrected, prolonged valvular stenosis or insufficiency may result in cardiac hypertrophy and associated damage to the heart muscle, which may eventually necessitate valve replacement.
  • cancer cardiovascular, endothelial and angiogenic disorders, which may or may not be accompanied by cardiac hypertrophy, is encompassed by the present invention.
  • cancer cancer
  • cancer cancer
  • cancer include but are not limited to, carcinoma including adenocarcinoma, lymphoma, blastoma, melanoma, sarcoma, and leukemia.
  • cancers include squamous cell cancer, small-cell lung cancer, non-small cell lung cancer, gastrointestinal cancer, Hodgkin's andnon-Hodgkin's lymphoma, pancreatic cancer, glioblastoma, cervical cancer, ovarian cancer, liver cancer such as hepatic carcinoma and hepatoma, bladder cancer, breast cancer, colon cancer, colorectal cancer, endometrial carcinoma, salivary gland carcinoma, kidney cancer such as renal cell carcinoma and Wilms' tumors, basal cell carcinoma, melanoma, prostate cancer, vulval cancer, thyroid cancer, testicular cancer, esophageal cancer, and various types of head and neck cancer.
  • cytotoxic agent 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., 131 1, 125 1, 90 Y, and 186 Re), chemotherapeutic agents, and toxins such as enzymatically active toxins of bacterial, fungal, plant, or animal origin, or fragments thereof.
  • chemotherapeutic agent is a chemical compound useful in the treatment of cancer.
  • examples of chemotherapeutic agents include alkylating agents, folic acid antagonists, anti-metabolites of nucleic acid metabolism, antibiotics, pyrimidine analogs, 5-fluorouracil, cisplatin, purine nucleosides, amines, amino acids, triazol nucleosides, or corticosteroids.
  • Adriamycin Poxorubicin, 5-Fluorouracil, Cytosine arabinoside ("Ara-C"), Cyclophosphamide, Thiotepa, Busulfan, Cytoxin, Taxol, Toxotere, Methotrexate, Cisplatin, Melphalan, Vinblastine, Bleomycin, Etoposide, Ifosfamide, Mitomycin C, Mitoxantrone, Vincreistine, Vinorelbine, Carboplatin, Teniposide, Oaunomycin, Carminomycin, Aminopterin, Pactinomycin, Mitomycins, Esperamicins (see U.S. Pat. No.4,675, 187), Melphalan, and other related nitrogen mustards. Also included in this definition are hormonal agents that act to regulate or inhibit hormone action on tumors, such as tamoxifen and onapristone.
  • a “growtli-inhibitory agent” when used herein refers to a compound or composition that inhibits growth of a cell, such as an Wnt-overexpressing cancer cell, either in vitro or in vivo.
  • the growm-inhibitory agent is one which significantly reduces the percentage of malignant cells in S phase.
  • growth-inhibitory agents include agents that block cell cycle progression (at a place other than S phase), such as agents that induce
  • M-phase blockers include the vincas (vincristine and vinblastine), taxol, and topo II inhibitors such as doxorubicin, daunorubicin, etoposide, and bleomycin.
  • vincas vincristine and vinblastine
  • topo II inhibitors such as doxorubicin, daunorubicin, etoposide, and bleomycin.
  • Those agents that arrest GI also spill over into S-phase arrest, for example, PNA alkylating agents such as tamoxifen, prednisone, dacarbazine, mechlorethamine, cisplatin, methotrexate, 5-fluorouracil, and ara-C. Further information can be found in The Molecular Basis of Cancer.
  • TNF tumor necrosis factor
  • HGF hepatocyte growth factor
  • 4P5 antibody an antibody capable of binding to HER2 receptor (WO 89/06692), such as the 4P5 antibody (and functional equivalents thereof) (e.g., WO 92/22653).
  • Treatment is an intervention performed with the intention of preventing the development or altering the pathology of a cardiovascular, endothelial, and angiogenic disorder.
  • the concept of treatment is used in the broadest sense, and specifically includes the prevention (prophylaxis), moderation, reduction, and curing of cardiovascular, endothelial, and angiogenic disorders of any stage.
  • treatment refers to both therapeutic treatment and prophylactic or preventative measures, wherein the object is to prevent or slow down (lessen) or ameliorate a cardiovascular, endothelial, and angiogenic disorder such as hypertrophy.
  • Those in need of treatment include those already with the disorder as well as those prone to have the disorder or those in whom the disorder is to be prevented.
  • the disorder may result from any cause, including idiopathic, cardiotrophic, or myotrophic causes, or ischemia or ischemic insults, such as myocardial infarction.
  • Chronic administration refers to administration ofthe agent(s) in a continuous mode as opposed to an acute mode, so as to maintain the initial effect, such as an anti-hypertrophic effect, for an extended period of time.
  • mammal for purposes of treatment refers to any animal classified as a mammal, including humans, domestic and farm animals, and zoo, sports, or pet animals, such as dogs, horses, cats, cows, sheep, pigs, etc. Preferably, the mammal is human.
  • cardiovascular, endothelial or angiogenic agents refers generically to any drag that acts in treating cardiovascular, endothelial, and angiogenic disorders.
  • cardiovascular agents are those that promote vascular homeostasis by modulating blood pressure, heart rate, heart contractility, and endothelial and smooth muscle biology, all of which factors have a role in cardiovascular disease.
  • angiotensin-II receptor antagonists include angiotensin-II receptor antagonists; endothelin receptor antagonists such as, for example, BOSENTANTM and MOXONOPINTM; interferon-gamma (IFN- ⁇ ); des-aspartate-angiotensin I; thrombolytic agents, e.g., streptokinase, urokinase, t-PA, and a t-PA variant specifically designed to have longer half-life and very high fibrin specificity, TNK t-PA (a Tl 03N, NI 17Q, KHRR(296-299)AAAA t-PA variant, Keyt et al., Proc. Natl. Acad. Sci. USA.
  • TNK t-PA a Tl 03N, NI 17Q, KHRR(296-299)AAAAAA t-PA variant
  • inotropic or hypertensive agents such as digoxigenin and ⁇ -adrenergic receptor blocking agents, e.g., propranolol, timolol, tertalolol, carteolol, nadolol, betaxolol, penbutolol, acetobutolol, atenolol, metoprolol, and carvedilol; angiotensin converting enzyme (ACE) inhibitors, e.g., quinapril, captopril, enalapril, ramipril, benazepril, fosinopril, and lisinopril; diuretics, e.g., clilorothiazide, hydrochlorothiazide, hydroflumethazide, methylchlothiazide, benzthiazide, dichlorphenamide, acetazolamide, and in
  • Angiogenic agents and “endothelial agents” are active agents that promote angiogenesis and or endothelial cell growth, or, if applicable, vasculogenesis. This would include factors that accelerate wound healing, such as growth hormone, insulin-like growth factor-I (IGF-I), VEGF, VIGF, PPGF, epidermal growth factor (EGF), CTGF and members of its family, FGF, and TGF- and TGF- ⁇ .
  • IGF-I insulin-like growth factor-I
  • VEGF VEGF
  • VIGF vascular endothelial growth factor
  • PPGF epidermal growth factor
  • CTGF epidermal growth factor
  • Angiostatic agents are active agents that inhibit angiogenesis or vasculogenesis or otherwise inhibit or prevent growth of cancer cells. Examples include antibodies or other antagonists to angiogenic agents as defined above, such as antibodies to VEGF. They additionally include cytotherapeutic agents such as cytotoxic agents, chemotherapeutic agents, growth-inhibitory agents, apoptotic agents, and other agents to treat cancer, such as anti- HER-2, anti-CP20, and other bioactive and organic chemical agents.
  • cytotherapeutic agents such as cytotoxic agents, chemotherapeutic agents, growth-inhibitory agents, apoptotic agents, and other agents to treat cancer, such as anti- HER-2, anti-CP20, and other bioactive and organic chemical agents.
  • a "therapeutically effective amount" of an active agent such as a PRO polypeptide or agonist or antagonist thereto or an anti-PRO antibody refers to an amount effective in the treatment of a cardiovascular, endothelial or angiogenic disorder in a mammal and can be determined empirically.
  • an "effective amount" of an active agent such as a PRO polypeptide or agonist or antagonist thereto or an anti-PRO antibody refers to an amount effective for carrying out a stated purpose, wherein such amounts may be determined empirically for the desired effect.
  • PRO polypeptide and "PRO” as used herein and when immediately followed by a numerical designation refer to various polypeptides, wherein the complete designation (i.e., PRO/number) refers to specific polypeptide sequences as described herein.
  • PRO/number polypeptide and “PRO/number” wherein the term “number” is provided as an actual numerical designation as used herein encompass native sequence polypeptides and polypeptide variants (which are further defined herein).
  • the PRO polypeptides described herein may be isolated from a variety of sources, such as from human tissue types or from another source, or prepared by recombinant or synthetic methods.
  • a “native sequence PRO polypeptide” comprises a polypeptide having the same amino acid sequence as the corresponding PRO polypeptide derived from nature. Such native sequence PRO polypeptides can be isolated from nature or can be produced by recombinant or synthetic means.
  • the term "native sequence PRO polypeptide” specifically encompasses naturally-occurring truncated or secreted forms ofthe specific PRO polypeptide (e.g., an extracellular domain sequence), naturally-occurring variant forms (e.g., alternatively spliced forms) and naturally-occurring allelic variants of the polypeptide.
  • the native sequence PRO polypeptides disclosed herein are mature or full-length native sequence polypeptides comprising the full-length amino acids sequences shown in the accompanying figures. Start and stop codons are shown in bold font and underlined in the figures. However, while the PRO polypeptide disclosed in the accompanying figures are shown to begin with methionine residues designated herein as amino acid position 1 in the figures, it is conceivable and possible that other methionine residues located either upstream or downstream from the amino acid position 1 in the figures may be employed as the starting amino acid residue for the PRO polypeptides.
  • the PRO polypeptide "extracellular domain” or “ECP” refers to a form ofthe PRO polypeptide which is essentially free ofthe transmembrane and cytoplasmic domains. Ordinarily, a PRO polypeptide ECD will have less than 1% of such transmembrane and or cytoplasmic domains and preferably, will have less than 0.5% of such domains. It will be understood that any transmembrane domains identified for the PRO polypeptides ofthe present invention are identified pursuant to criteria routinely employed in the art for identifying that type of hydrophobic domain. The exact boundaries of a transmembrane domain may vary but most likely by no more than about 5 amino acids at either end ofthe domain as initially identified herein.
  • an extracellular domain of a PRO polypeptide may contain from about 5 or fewer amino acids on either side of the transmembrane domain extracellular domain boundary as identified in the Examples or specification and such polypeptides, with or without the associated signal peptide, and nucleic acid encoding them, are comtemplated by the present invention.
  • the approximate location ofthe "signal peptides" ofthe various PRO polypeptides disclosed herein are shown in the present specification and or the accompanying figures.
  • the C-terminal boundary of a signal peptide may vary, but most likely by no more than about 5 amino acids on either side ofthe signal peptide C-terminal boundary as initially identified herein, wherein the C-terminal boundary of the signal peptide may be identified pursuant to criteria routinely employed in the art for identifying that type of amino acid sequence element (e.g., Nielsen et al, Prot Eng connector 10:1-6 (1997) and von Heinje et al, Nucl. Acids Res.,
  • 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 acids on either side ofthe C-terminal boundary of the signal peptide as identified herein, and the polynucleotides encoding them, are contemplated by the present invention.
  • PRO polypeptide variant means an active PRO polypeptide as defined above or below having at least about 80% amino acid sequence identity with a full-length native sequence PRO polypeptide sequence as disclosed herein, a PRO polypeptide sequence lacking the signal peptide as disclosed herein, an extracellular domain of a PRO polypeptide, with or without the signal peptide, as disclosed herein or any other fragment of a full-length PRO polypeptide sequence as disclosed herein.
  • Such PRO polypeptide variants include, for instance, PRO polypeptides wherein one or more ammo acid residues are added, or deleted, at the N- or C-terminus of the full-length native amino acid sequence.
  • a PRO polypeptide variant will have at least about 80%, 81%, 82%, 83%, 84%,
  • PRO variant polypeptides are at least about 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, 150 or 200 amino acids in length and alternatively at least about 300 amino acids in length, or more.
  • Percent (%) amino acid sequence identity with respect to the PRO polypeptide sequences identified herein is defined as the percentage of amino acid residues in a candidate sequence that are identical with the amino acid residues in a PRO 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 ofthe sequence identity. Alignment for purposes of determining percent amino acid sequence identity can be achieved in various ways that are within the skill in the art, for instance, using publicly available computer software such as BLAST, BLAST-2, ALIGN, ALIGN-2 or Megalign (DNASTAR) software. Those skilled in the art can determine appropriate parameters for measuring alignment, including any algorithms needed to achieve maximal alignment over the full-length of the sequences being compared.
  • % amino acid sequence identity values are obtained as described below by using the sequence comparison computer program ALIGN-2, wherein the complete source code for the ALIGN-2 program is provided in Table 1.
  • the ALIGN-2 sequence comparison computer program was authored by Genentech, Inc., and the source code shown in Table 1 has been filed with user documentation in the U.S. Copyright Office, Washington D.C., 20559, where it is registered under
  • % amino acid sequence identity of a given amino acid sequence A to, with, or against a given amino acid sequence B is calculated as follows:
  • % amino acid sequence identity values used herein are obtained as described above using the ALIGN-2 sequence comparison computer program.
  • % amino acid sequence identity may also be determined using the sequence comparison program NCBI-BLAST2 (Altschul et al, Nucleic Acids Res..25:3389-3402 (1997)).
  • NCBI-BLAST2 sequence comparison program may be downloaded from http://www.ncbi.nlm.nih. gov. or otherwise obtained from the National Institute of Health, Bethesda, MD.
  • % amino acid sequence identity of a given amino acid sequence A to, with, or against a given amino acid sequence B is calculated as follows:
  • a % amino acid sequence identity value is determined by dividing (a) the number of matching identical amino acids residues between the amino acid sequence ofthe PRO polypeptide of interest having a sequence derived from the native PRO polypeptide and the comparison amino acid sequence of interest (i. e.
  • the sequence against which the PRO polypeptide of interest is being compared which may be a PRO variant polypeptide) as determined by WU-BLAST-2 by (b) the total number of amino acid residues ofthe PRO polypeptide of interest.
  • the amino acid sequence A is the comparison ammo acid sequence of interest and the amino acid sequence B is the amino acid sequence of the PRO polypeptide of interest.
  • PRO variant polynucleotide or "PRO variant nucleic acid sequence” means a nucleic acid molecule which encodes an active PRO polypeptide as defined below and which has at least about 80% nucleic acid sequence identity with a nucleotide acid sequence encoding a full-length native sequence PRO polypeptide sequence as disclosed herein, a full-length native sequence PRO polypeptide sequence lacking the signal peptide as disclosed herein, an extracellular domain of a PRO polypeptide, with or without the signal peptide, as disclosed herein or any other fragment of a full-length PRO polypeptide sequence as disclosed herein.
  • a PRO variant polynucleotide will have at least about 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%,
  • nucleic acid sequence identity 93%, 94%, 95%, 96%, 97% or 98% nucleic acid sequence identity and alternatively at least about 99% nucleic acid sequence identity with a nucleic acid sequence encoding a full-length native sequence PRO polypeptide sequence as disclosed herein, a full-length native sequence PRO polypeptide sequence lacking the signal peptide as disclosed herein, an extracellular domain of a PRO polypeptide, with or without the signal sequence, as disclosed herein or any other fragment of a full-length PRO polypeptide sequence as disclosed herein. Variants do not encompass the native nucleotide sequence.
  • PRO variant polynucleotides are at least about 30, 60, 90, 120, 150, 180, 210, 240, 270, 300, 450, or 600 nucleotides in length and alternatively at least about 900 nucleotides in length, or more.
  • Percent (%) nucleic acid sequence identity with respect to the PRO polypeptide-encoding nucleic acid sequences identified herein is defined as the percentage of nucleotides in a candidate sequence that are identical with the nucleotides in a PRO polypeptide-encoding nucleic acid sequence, 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, ALIGN-2 or Megalign (DNASTAR) software.
  • ALIGN-2 sequence comparison computer program
  • Table 1 the complete source code for the ALIGN- 2 program is provided in Table 1.
  • the ALIGN-2 sequence comparison computer program was authored by Genentech, Inc., and the source code shown in Table 1 has been filed with user documentation in the U.S. Copyright
  • the ALIGN-2 program is publicly available through Genentech, Inc., South San Francisco, California or may be compiled from the source code provided in Table 1.
  • 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.
  • % nucleic acid sequence identity of a given nucleic acid sequence C to, with, or against a given nucleic acid sequence D is calculated as follows:
  • % nucleic acid sequence identity values used herein are obtained as described above using the ALIGN-2 sequence comparison computer program. However, % nucleic acid sequence identity may also be determined using the sequence comparison program NCBI-BLAST2 (Altschul et al. ,
  • % nucleic acid sequence identity of a given nucleic acid sequence C to, with, or against a given nucleic acid sequence D is calculated as follows:
  • % nucleic acid sequence identity values may also be generated using the WU-BLAST-
  • a % nucleic acid sequence identity value is determined by dividing (a) the number of matching identical nucleotides between the nucleic acid sequence ofthe PRO polypeptide-encoding nucleic acid molecule of interest having a sequence derived from the native sequence PRO polypeptide-encoding nucleic acid and the comparison nucleic acid molecule of interest (i.e., the sequence against which the PRO polypeptide-encoding nucleic acid molecule of interest is being compared which may be a variant PRO polynucleotide) as determined by WU-BLAST-2 by (b) the total number of nucleotides ofthe PRO polypeptide- encoding nucleic acid molecule of interest.
  • nucleic acid sequence A is the comparison nucleic acid molecule of interest and the nucleic acid sequence B is the nucleic acid sequence ofthe PRO polypeptide-encoding nucleic acid molecule of interest.
  • PRO variant polynucleotides are nucleic acid molecules that encode an active PRO polypeptide and which are capable of hybridizing, preferably under stringent hybridization and wash conditions, to nucleotide sequences encoding the full-length PRO polypeptide as shown in the specification herein and accompanying figures.
  • PRO variant polypeptides may be those that are encoded by a PRO variant polynucleotide.
  • isolated when used to describe the various polypeptides disclosed herein, means a polypeptide that has been identified and separated and or recovered from a component of its natural environment. Preferably, the isolated polypeptide is free of association with all components with which it is naturally associated. Contaminant components of its natural environment are materials that would typically interfere with diagnostic or therapeutic uses for the polypeptide, and may include enzymes, hormones, and other proteinaceous or non-proteinaceous solutes.
  • the polypeptide will be purified (1) to a degree sufficient to obtain at least 15 residues of N-terminal or internal amino acid sequence by use of a spinning cup sequenator, or (2) to homogeneity by SPS-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 ofthe PRO natural environment will not be present. Ordinarily, however, isolated polypeptide will be prepared by at least one purification step.
  • An "isolated" nucleic acid molecule encoding a PRO polypeptide or an “isolated” nucleic acid molecule encoding an anti-PRO antibody 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 ofthe PRO-encoding nucleic acid or the natural source ofthe anti-PRO-encoding nucleic acid.
  • the isolated nucleic acid is free of association with all components with which it is naturally associated.
  • An isolated PRO-encoding nucleic acid molecule or an isolated anti-PRO-encoding nucleic acid molecule is other than in the form or setting in which it is found in nature.
  • Isolated nucleic acid molecules therefore are distinguished from the PRO-encoding nucleic acid molecule or from the anti-PRO-encoding nucleic acid molecule as it exists in natural cells.
  • an isolated nucleic acid molecule encoding a PRO polypeptide or an isolated nucleic acid molecule encoding an anti- PRO antibody includes PRO-nucleic acid molecules or anti-PRO-nucleic acid molecules contained in cells that ordinarily express PRO polypeptides or anti-PRO antibodies where, for example, the nucleic acid molecule is in a chromosomal location different from that of natural cells.
  • 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 include a promoter, optionally an operator sequence, and a ribosome binding site.
  • Eukaryotic cells are known to utilize, for example, promoters, polyadenylation signals, and enhancers.
  • Nucleic acid is "operably linked" when it is placed into a functional relationship with another nucleic acid sequence.
  • DNA for a presequence or secretory leader is operably linked to DNA for a PRO polypeptide if it is expressed as a preprotein that participates in the secretion of the polypeptide;
  • a promoter or enhancer is operably linked to a coding sequence if it affects the transcription of the sequence; or
  • a ribosome binding site is operably linked to a coding sequence if it is positioned so as to facilitate translation.
  • "operably linked” means that the DNA sequences being linked are contiguous, and, in the case of a secretory leader, contiguous and in the same reading frame. However, enhancers do not have to be contiguous. Linking is accomplished by ligation at convenient restriction sites. If such sites do not exist, 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 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 enviromnent below their melting temperature. The higher the degree of desired homology between the probe and hybridizable sequence, the higher the relative temperature that can be used. As a result, it follows that higher relative temperatures would tend to make the reaction conditions more stringent, while lower temperatures less so. For additional details and explanation of stringency of hybridization reactions, see, Ausubel et al, Current Protocols in Molecular Biology (Wiley Interscience Publishers, 1995).
  • “Stringent conditions” or “high-stringency conditions”, as defined herein, may be identified by those that: (1) employ low ionic strength and high temperature for washing, for example, 0.015 M sodium chloride/0.0015 M sodium citrate/0.1% sodium dodecyl sulfate at 50°C; (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 chloride, 75 mM sodium citrate at 42°C; or (3) employ 50% formamide, 5 x SSC (0.75 M NaCl, 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 ⁇ g/ml), 0.1% SDS, and 10% dextran s
  • Modely-stringent conditions may be identified as describedby Sambrook et al, Molecular Cloning: A Laboratory Manual (New York: Cold Spring Harbor Press, 1989), and include the use of wasliing solution and hybridization conditions (e.g., temperature, ionic strength, and % SDS) less stringent than those described above.
  • moderately stringent conditions is overnight incubation at 37°C in a solution comprising: 20% formamide, 5 x SSC (150 mMNaCl, 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 37-50°C.
  • a solution comprising: 20% formamide, 5 x SSC (150 mMNaCl, 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 37-50°C.
  • the skilled artisan will recognize how to adjust the temperature, ionic strength, etc. as necessary to accommodate
  • epitope-tagged when used herein refers to a chimeric polypeptide comprising a PRO polypeptide fused to a "tag polypeptide".
  • the tag polypeptide has enough residues to provide an epitope against which an antibody can be made, yet is short enough such that it does not interfere with activity ofthe 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).
  • Active or “activity” in the context of PRO variants refers to form(s) of PRO proteins that retain the biologic and or immunologic activities of a native or naturally-occurring PRO polypeptide.
  • Bioactivity in the context of a molecule that antagonizes a PRO polypeptide that can be identified by the screening assays disclosed herein (e.g. , an organic or inorganic small molecule, peptide, etc.) is used to refer to the ability of such molecules to bind or complex with the PRO polypeptide identified herein, or otherwise interfere with the interaction of the PRO polypeptide with other cellular proteins or otherwise inhibits the transcription or translation of the PRO polypeptide.
  • Particularly preferred biological activity includes cardiac hypertrophy, activity that acts on systemic disorders that affect vessels, such as diabetes mellitus, as well as diseases ofthe arteries, capillaries, veins, and/or lymphatics, and cancer.
  • Antagonist is used in the broadest sense, and includes any molecule that partially or fully blocks, inhibits, or neutralizes one or more ofthe biological activities of a native PRO polypeptide disclosed herein, for example, if applicable, its mitogenic or angiogenic activity.
  • Antagonists of a PRO polypeptide may act by interfering with the binding of a PRO polypeptide to a cellular receptor, by incapacitating or killing cells that have been activated by a PRO polypeptide, or by interfering with vascular endothelial cell activation after binding of a PRO polypeptide to a cellular receptor. All such points of intervention by a PRO polypeptide antagonist shall be considered equivalent for purposes of this invention.
  • the antagonists inhibit the mitogenic, angiogenic, or other biological activity of PRO polypeptides, and thus are useful for the treatment of diseases or disorders characterized by undesirable excessive neovascularization, including by way of example tumors, and especially solid malignant tumors, rheumatoid arthritis, psoriasis, atherosclerosis, diabetic and otlier retinopathies, retrolental fibroplasia, age- related macular degeneration, neovascular glaucoma, hemangiomas, thyroid hyperplasias (including Grave's disease), corneal and other tissue transplantation, and chronic inflammation.
  • tumors and especially solid malignant tumors, rheumatoid arthritis, psoriasis, atherosclerosis, diabetic and otlier retinopathies, retrolental fibroplasia, age- related macular degeneration, neovascular glaucoma, hemangiomas, thyroid hyperplasias (including Grave's disease
  • the antagonists also are useful for the treatment of diseases or disorders characterized by undesirable excessive vascular permeability, such as edema associated with brain tumors, ascites associated with malignancies, Meigs' syndrome, lung inflammation, neplrrotic syndrome, pericardial effusion (such as that associated with pericarditis), andpleural effusion.
  • agonist is used in the broadest sense and includes any molecule that mimics a biological activity of a native PRO polypeptide disclosed herein. Suitable agonist or antagonist molecules specifically include agonist or antagonist antibodies or antibody fragments, fragments, or amino acid sequence variants of native PRO polypeptides, peptides, small organic molecules, etc.
  • PRO polypeptide receptor refers to a cellular receptor for a PRO polypeptide, ordinarily a cell-surface receptor found on vascular endothelial cells, as well as variants thereof that retain the ability to bind a PRO polypeptide.
  • Antibodies are glycoproteins having the same structural characteristics. While antibodies exhibit binding specificity to a specific antigen, immunoglobulms include both antibodies and otlier antibody-like molecules that lack antigen specificity. Polypeptides ofthe latter kind are, for example, produced at low levels by the lymph system and at increased levels by myelomas.
  • antibody is used in the broadest sense and specifically covers, without limitation, intact monoclonal antibodies, polyclonal antibodies, multispecific antibodies (e.g., bispecific antibodies) formed from at least two intact antibodies, and antibody fragments, so long as they exhibit the desired biological activity.
  • “Native antibodies” and “native immunoglobulms” are usually heterotetrameric glycoproteins of about 150,000 daltons, composed of two identical light (L) chains and two identical heavy (H) chains. Each light chain is linked to a heavy chain by one covalent disulfide bond, while the number of disulfide linkages varies among the heavy chains of different immunoglobulin isotypes. Each heavy and hght chain also has regularly spaced intrachain disulfide bridges. Each heavy chain has at one end a variable domain (V H ) followed by a number of constant domains.
  • V H variable domain
  • Each light chain has a variable domain at one end (V L ) and a constant domain at its other end; the constant domain ofthe light chain is aligned with the first constant domain ofthe heavy chain, and the light-chain variable domain is aligned with the variable domain ofthe heavy chain. Particular amino acid residues are believed to form an interface between the light- and heavy-chain variable domains.
  • the term "variable” refers to the fact that certain portions ofthe variable domains differ extensively in sequence among antibodies and are used in the binding and specificity of each particular antibody to and for its particular antigen. However, the variability is not evenly distributed throughout the variable domains of antibodies. It is concentrated in three segments called complementarity-determining regions (CDRs) or hypervariable regions both in the light-chain and the heavy-chain variable domains.
  • CDRs complementarity-determining regions
  • variable domains The more highly conserved portions of variable domains are called the framework regions (FR).
  • the variable domains of native heavy and light chains each comprise four FR regions, largely adopting a ⁇ -sheet configuration, connected by three CDRs, which form loops connecting, and in some cases forming part of, the ⁇ -sheet structure.
  • the CDRs in each chain are held together in close proximity by the FR regions and, with the CDRs from the other chain, contribute to the formation of the antigen-binding site of antibodies. See, Kabat et al, NHI Publ, No.91-3242. Vol. I, pages 647-669 (1991).
  • the constant domains are not involved directly in binding an antibody to an antigen, but exhibit various effector functions, such as participation ofthe antibody in antibody-dependent cellular toxicity.
  • Antibody fragments comprise a portion of an intact antibody, preferably the antigen-binding or variable region of the intact antibody.
  • antibody fragments include Fab, Fab', F(ab') 2 , and Fv fragments; diabodies; linear antibodies (Zapata et al. , Protein Eng..8(10): 1057-1062 (1995)); single-chain antibody molecules; and multispecific antibodies formed from antibody fragments.
  • Papain digestion of antibodies produces two identical antigen-binding fragments, called “Fab” fragments, each with a single antigen-binding site, and a residual "Fc” fragment, whose name reflects its ability to crystallize readily. Pepsin treatment yields an F(ab') 2 fragment that has two antigen-combining sites and is still capable of cross-linking antigen.
  • Fv is the minimum antibody fragment that contains a complete antigen-recognition and -binding site. This region consists of a dimer of one heavy- and one light-chain variable domain in tight, non-covalent association. It is in this configuration that the three CDRs of each variable domain interact to define an antigen-binding site on the surface of the V H -V L dimer. Collectively, the six CDRs confer antigen-binding specificity to the antibody. However, even a single variable domain (or half of an Fv comprising only three CDRs specific for an antigen) has the ability to recognize and bind antigen, although at a lower affinity than the entire binding site.
  • the Fab fragment also contains the constant domain of the light chain and the first constant domain (CHI) of the heavy chain.
  • Fab' fragments differ from Fab fragments by the addition of a few residues at the carboxy terminus ofthe heavy chain CHI domain including one or more cysteines from the antibody hinge region.
  • Fab'-SH is the designation herein for Fab' in which the cysteine residue(s) ofthe constant domains bear a free thiol group.
  • F(ab') 2 antibody fragments originally were produced as pairs of Fab' fragments that have hinge cysteines between them. Other chemical couplings of antibody fragments are also known.
  • the "light chains" of antibodies (immunoglobulms) 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.
  • hiimunoglobulins can be assigned to different classes. There are five major classes of immunoglobulms: IgA, IgD, IgE, IgG, and IgM; and several of these maybe further divided into subclasses (isotypes), e.g., IgGl , IgG2, IgG3, IgG4, IgA, and IgA2.
  • the heavy-chain constant domains that correspond to the different classes of immunoglobulms are called , ⁇ , e, ⁇ , and ⁇ , respectively.
  • the subunit stractures and three-dimensional configurations of different classes of immunoglobulms are well known.
  • 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 forpossible naturally-occurring mutations that maybe present in minor amounts. Monoclonal antibodies are highly specific, being directed against a single antigenic site. Furthermore, in contrast to conventional (polyclonal) antibody preparations that typically 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 are synthesized by the hybridoma culture, uncontaminated by other immunoglobulms.
  • the modifier "monoclonal” indicates the character ofthe antibody as being obtained from a substantially homogeneous population of antibodies, and is not to be construed as requiring production of the antibody by any particular method.
  • the monoclonal antibodies to be used in accordance with the present invention may be made by the hybridoma method first described by Kohler et al, Nature.256: 495 ( 1975), or may be made by recombinant DNA methods (see, e.g., U.S. PatentNo.4,816,567).
  • the “monoclonal antibodies” may also be isolated from phage antibody libraries using the techniques described in Clackson et al, Nature, 352: 624-628 (1991) and Marks et al, J. Mol.
  • the monoclonal antibodies herein specifically include "chimeric" antibodies (immunoglobulms) in which a portion of the heavy and/or light chain is identical with or homologous to corresponding sequences in antibodies derived from a particular species or belonging to a particular antibody class or subclass, while the remainder ofthe 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 the desired biological activity.
  • chimeric antibodies immunoglobulms in which a portion of the heavy and/or light chain is identical with or homologous to corresponding sequences in antibodies derived from a particular species or belonging to a particular antibody class or subclass, while the remainder ofthe 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 the desired biological activity.
  • Humanized forms of non-human (e.g., murine) antibodies are chimeric immunoglobulms, immunoglobulin chains, or fragments thereof (such as Fv, Fab, Fab', F(ab') 2j or other antigen-binding subsequences of antibodies) that contain minimal sequence derived from non-human immunoglobulin.
  • humanized antibodies are human immunoglobulms (recipient antibody) in which residues from a CDR of the recipient are replaced by residues from a CDR of a non-human species (donor antibody) such as mouse, rat or rabbit having the desired specificity, affinity, and capacity.
  • donor antibody such as mouse, rat or rabbit having the desired specificity, affinity, and capacity.
  • Fv FR residues of the human immunoglobulin are replaced by corresponding non-human residues.
  • humanized antibodies may comprise residues that are found neither in the recipient antibody nor in the imported CDR or framework sequences.
  • the humanized antibody will comprise substantially all of at least one, and typically two, variable domains, in which all or substantially all ofthe CDR regions correspond to those of a non-human immunoglobulin and all or substantially all ofthe FR regions are those of a human immunoglobulin sequence.
  • the humanized antibody preferably also will comprise at least aportion of an immunoglobulin constant region (Fc), typically that of a human immunoglobulin.
  • the humanized antibody includes a PRIMATIZEDTM antibody wherein the antigen-binding region ofthe antibody is derived from an antibody produced by immunizing macaque monkeys with the antigen of interest "Single-chain Fv" or "sFv” antibody fragments comprise the V H and V L domains of an antibody, wherein these domains are present in a single polypeptide chain.
  • the Fv polypeptide further comprises a polypeptide linker between the V H and V L domains that enables the sFv to form the desired structure for antigen binding.
  • a polypeptide linker between the V H and V L domains that enables the sFv to form the desired structure for antigen binding.
  • Diabodies are described more fully in, for example, EP 404,097; WO 93/11161; and Hollinger et al, Proc. Natl. Acad. Sci. USA, 90: 6444-6448 (1993).
  • an “isolated” antibody is one that has been identified and separated and/or recovered from a component of its natural environment. Contaminant components of its natural environment are materials that would interfere with diagnostic or therapeutic uses for the antibody, and may include enzymes, hormones, and other proteinaceous or nonproteinaceous solutes.
  • 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 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.
  • An antibody that "specifically binds to” or is “specific for” a particular polypeptide or an epitope on a particular polypeptide is one that binds to that particular polypeptide or epitope on a particular polypeptide without substantially binding to any other polypeptide or polypeptide epitope.
  • label when used herein refers to a detectable compound or other composition that is conjugated directly or indirectly to the antibody so as to generate a "labeled" antibody.
  • the label may be detectable by itself (e.g. , radioisotope labels or fluorescent labels) or, in the case of an enzymatic label, may catalyze chemical alteration of a substrate compound or composition that is detectable.
  • Radionuclides that can serve as detectable labels include, for example, 1-131, 1-123, 1-125, Y-90, Re-188, At-211, Cu-67, Bi-212, and Pd-109.
  • the label may also be a non- detectable entity such as a toxin.
  • solid phase is meant a non-aqueous matrix to which an antibody ofthe present invention can adhere.
  • solid phases encompassed herein include those formed partially or entirely of glass (e.g., controlled pore glass), polysaccharides (e.g., agarose), polyacrylamides, polystyrene, polyvinyl alcohol and silicones.
  • the solid phase can comprise the well of an assay plate; in others it is a purification column (e.g., an affinity chromatography column). This term also includes a discontinuous solid phase of discrete particles, such as those described in U.S. Patent No. 4,275,149.
  • a “liposome” is a small vesicle composed of various types of lipids, phospholipids and/or surfactant that is useful for delivery of a drag (such as the PRO polypeptide or antibodies thereto disclosed herein) to a mammal.
  • the components ofthe liposome are commonly arranged in a bilayer formation, similar to the lipid arrangement of biological membranes.
  • immunoadhesin designates antibody-like molecules that combine the binding specificity of a heterologous protein (an “adhesin”) with the effector functions of immunoglobulin constant domains.
  • 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 molecule typically is a contiguous amino acid sequence comprising at least the binding site of a receptor or a ligand.
  • the immunoglobulin constant domain sequence in the immunoadhesin may be obtained from any immunoglobulin, such as IgG- 1 , IgG- 2, IgG-3, or IgG-4 subtypes, IgA (including IgA-1 and IgA-2), IgE, IgD, or IgM.
  • immunoglobulin such as IgG- 1 , IgG- 2, IgG-3, or IgG-4 subtypes, IgA (including IgA-1 and IgA-2), IgE, IgD, or IgM.
  • Table 1 provides the complete source code for the ALIGN-2 sequence comparison computer program. This source code may be routinely compiled for use on a UNIX operating system to provide the ALIGN-2 sequence comparison computer program.
  • Tables 2-5 show hypothetical exemplifications for using the below described method to deteimine % amino acid sequence identity (Tables 2-3) and % nucleic acid sequence identity (Tables 4-5) using the ALIGN-2 sequence comparison computer program, wherein "PRO” represents the amino acid sequence of a hypothetical PRO polypeptide of interest, “Comparison Protein” represents the amino acid sequence of a polypeptide against which the "PRO” polypeptide of interest is being compared, “PRO-DNA” represents a hypothetical PRO-encoding nucleic acid sequence of interest, “Comparison DNA” represents the nucleotide sequence of a nucleic acid molecule against which the "PRO-DNA” nucleic acid molecule of interest is being compared, “X”, “Y”, and “Z” each represent different hypothetical amino acid residues and "N", “L” and “V” each represent different hypothetical nucleotides.
  • 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
  • the program may create a imp file in /imp to hold info about traceback.
  • dumpblock( ) dump a block of lines with numbers, stars: pr_align( )
  • stripname( ) strip any path and prefix from a seqname */
  • 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 */ static siz[2]; static char *ps[2]; /* ptr to current element */ static char *po[2]; /* ptr to next ou ⁇ ut char slot */ static char out[2][P LINE]; /* output line */ static char star[P LINE]; /* set by stars( ) */
  • *ps[i] toupper(*ps[i]); po[i] + + ; ps[i] + + ;
  • *py+ + *px; else if (islower(*px))
  • *py+ + toupper(*px); if (index("ATGCU",*(py-l))) natgc+ + ;
  • PRO variants can be prepared.
  • PRO variants can be prepared by introducing appropriate nucleotide changes into the PRO DNA, and/or by synthesis ofthe desired PRO polypeptide.
  • amino acid changes may alter post-translational processes ofthe PRO polypeptide such as changing the number or position of glycosylation sites or altering the membrane anchoring characteristics.
  • Variations in the native full-length sequence PRO polypeptide or in various domains of the PRO polypeptide described herein can be made, for example, using any of tlie techniques and guidelines for conservative and non-conservative mutations set forth, for instance, in U.S. Patent No. 5,364,934.
  • Variations may be a substitution, deletion or insertion of one or more codons encoding tlie PRO polypeptide that results in a change in tlie amino acid sequence ofthe PRO polypeptide as compared with the native sequence PRO polypetide.
  • the variation is by substitution of at least one amino acid with any other amino acid in one or more ofthe domains ofthe PRO polypeptide.
  • Guidance in determining which amino acid residue may be inserted, substituted or deleted without adversely affecting the desired activity may be found by comparing tlie sequence ofthe PRO polypeptide with that of homologous known protein molecules and minimizing the number of amino acid sequence changes made in regions of high homology.
  • Amino acid substitutions can be the result of replacing one amino acid with another amino acid having similar structural and/or chemical properties, such as the replacement of a leucine with a serine, i. e. , conservative amino acid replacements.
  • Insertions or deletions may optionally be in the range of about 1 to 5 amino acids. The variation allowed may be determined by systematically making insertions, deletions or substitutions of amino acids in the sequence and testing the resulting variants for activity exhibited by the full-length or mature native sequence.
  • conservative substitutions of interest are shown in Table 6 under the heading of preferred substitutions. If such substitutions result in a change in biological activity, then more substantial changes, denominated exemplary substitutions in Table 6, or as further described below in reference to amino acid classes, are introduced and the products screened.
  • Substantial modifications in function or immunological identity ofthe PRO polypeptide are accomplished by selecting substitutions that differ significantly in their effect on maintaining (a) the structure ofthe polypeptide backbone in the area of the substitution, for example, as a sheet or helical conformation, (b) the charge or hydrophobicity ofthe molecule at the target site, or (c) the bulk ofthe side chain.
  • Naturally occurring residues are divided into groups based on common side-chain properties:
  • hydrophobic norleucine, met, ala, val, leu, ile
  • Non-conservative substitutions will entail exchanging a member of one of these classes for another class. Such substituted residues also may be introduced into the conservative substitution sites or, more preferably, into the remaining (non-conserved) sites.
  • the variations can be made using methods known in the art such as oligonucleotide-mediated (site- directed) mutagenesis, alanine scanning, and PCR mutagenesis.
  • Site-directed mutagenesis [Carter et al, Nucl. Acids Res.. 13:4331 (1986); Zoller et al, Nucl. Acids Res.. 10:6487 (1987)]
  • cassette mutagenesis [Wells et al, Gene, 34:315 (1985)]
  • restriction selection mutagenesis [Wells etal, Philos. Trans. R. Soc. London SerA, 317:415 (1986)] or other known techniques can be performed on the cloned DNA to produce the PRO variant DNA.
  • Scanning amino acid analysis can also be employed to identify one or more amino acids along a contiguous sequence.
  • preferred scanning amino acids are relatively small, neutral amino acids.
  • amino acids include alanine, glycine, serine, and cysteine.
  • Alanine is typically a preferred scanning amino acid among this group because it eliminates the side-chain beyond the beta-carbon and is less likely to alter the main-chain conformation ofthe variant [Cunningham and Wells, Science, 244: 1081-1085 (1989)].
  • Alanine is also typically preferred because it is the most common amino acid. Further, it is frequently found in both buried and exposed positions [Creighton, The Proteins, (W.H. Freeman & Co., N.Y.); Chothia, J. Mol. Biol., 150: 1 (1976)]. If alanine substitution does not yield adequate amounts of variant, an isoteric amino acid can be used.
  • Covalent modifications of PRO polypeptides are included within the scope of this invention.
  • One type of covalent modification includes reacting targeted amino acid residues of a PRO polypeptide with an organic derivatizing agent that is capable of reacting with selected side chains or the N- or C- terminal residues ofthe PRO polypeptide.
  • Derivatization with bifunctional agents is useful, for instance, for crosslinking the PRO polypeptide to a water-insoluble support matrix or surface for use in the method for purifying anti-PRO antibodies, and vice- versa.
  • crosslinking agents include, e.g., l,l-bis(diazoacetyl)-2-phenylethane, glutaraldehyde, N- hydroxysuccinimide esters, for example, esters with 4-azidosalicylic acid, homobifunctional imidoesters, including disuccinimidyl esters such as 3,3'-dithiobis(succinimidylpropionate), bifunctional maleimides such as bis-N- maleimido-l,8-octane and agents such as methyl-3-[( ⁇ -azido ⁇ henyl)dithio]propioimidate.
  • Another type of covalent modification ofthe PRO polypeptide included within the scope of this invention comprises altering the native glycosylation pattern ofthe polypeptide.
  • "Altering the native glycosylation pattern” is intended for purposes herein to mean deleting one or more carbohydrate moieties found in the native sequence PRO polypeptide (either by removing the underlying glycosylation site or by deleting the glycosylation by chemical and/or enzymatic means) , and or adding one or more glycosylation sites that are not present in the native sequence PRO polypeptide.
  • tlie phrase includes qualitative changes in tlie glycosylation ofthe native proteins, involving a change in the nature and proportions ofthe various carbohydrate moieties present.
  • Addition of glycosylation sites to the PRO polypeptide may be accomplished by altering the amino acid sequence.
  • the alteration may be made, for example, by the addition of, or substitution by, one or more serine or threonine residues to the native sequence PRO polypeptide (for O-linked glycosylation sites).
  • the PRO amino acid sequence may optionally be altered through changes at the DNA level, particularly by mutating the DNA encoding the PRO polypeptide at preselected bases such that codons are generated that will translate into the desired amino acids.
  • Another means of increasing tlie number of carbohydrate moieties on the PRO polypeptide is by chemical or enzymatic coupling of glycosides to the polypeptide. Such methods are described in the art, e.g., in WO 87/05330 published 11 September 1987, and in Aplin and Wriston, CRC Crit. Rev. Biochem., pp.259-306 (1981). Removal of carbohydrate moieties present on tlie PRO polypeptide may be accomplished chemically or enzymatically or by mutational substitution of codons encoding for amino acid residues that serve as targets for glycosylation. Chemical deglycosylation techniques are known in the art and described, for instance, by Hakimuddin, et al, Arch. Biochem.
  • Another type of covalent modification ofthe PRO polypeptide comprises linking the PRO polypeptide to one of a variety of nonproteinaceous polymers, e.g., polyethylene glycol (PEG), polypropylene glycol, or polyoxyalkylenes, in the manner set forth in U.S. Patent Nos. 4,640,835; 4,496,689; 4,301,144; 4,670,417; 4,791,192 or 4,179,337.
  • the PRO polypeptide ofthe present invention may also be modified in a way to form a chimeric molecule comprising the PRO polypeptide fused to another, heterologous polypeptide or amino acid sequence.
  • such a chimeric molecule comprises a fusion ofthe PRO polypeptide with a protein transduction domain which targets the PRO polypeptide for delivery to various tissues and more particularly across the brain blood barrier, using, for example, the protein transduction domain of human immunodeficiency virus TAT protein (Schwarze et al, 1999, Science 285: 1569-72).
  • such a chimeric molecule comprises a fusion ofthe PRO polypeptide with a tag polypeptide which provides an epitope to which an anti-tag antibody can selectively bind.
  • the epitope tag is generally placed at the amino- or carboxyl- terminus ofthe PRO polypeptide. The presence of such epitope- tagged forms ofthe PRO polypeptide can be detected using an antibody against the tag polypeptide. Also, provision of the epitope tag enables the PRO polypeptide to be readily purified by affinity purification using an anti-tag antibody or another type of affinity matrix that binds to the epitope tag.
  • tag polypeptides and their respective antibodies are well known in the art.
  • poly-histidine poly-His
  • poly-histidine-glycine poly- His-gly
  • tag polypeptides include the Flag-peptide [Hopp etal, BioTechnology, 6:1204-1210 (1988)]: the KT3 epitope peptide [Martin etal. Science, 255:192-194 (1992)]; an ⁇ -tubulin epitope peptide [Skinner et al, J. Biol. Chem..266:15163-15166 (1991)]; and the T7 gene 10 protein peptide tag [Lutz-Freyermuth etal, Proc. Natl. Acad. Sci. USA, 87:6393-6397 (1990)].
  • the chimeric molecule may comprise a fusion ofthe PRO polypeptide with an immunoglobulin or a particular region of an immunoglobulin.
  • an immunoglobulin also referred to as an "immunoadhesin”
  • a fusion could be to the Fc region of an IgG molecule.
  • the Ig fusions preferably include the substitution of a soluble (transmembrane domain deleted or inactivated) form of a PRO polypeptide in place of at least one variable region within an Ig molecule.
  • the immunoglobulin fusion includes the hinge, CH2 and CH3, or the hinge, CHI , CH2 and CH3 regions of an IgGl molecule.
  • the present invention provides newly identified and isolated nucleotide sequences encoding polypeptides referred to in the present application as PRO polypeptides.
  • cDNAs encoding PRO polypeptides have been identified and isolated, as disclosed in further detail in the Examples below. It is noted that proteins produced in separate expression rounds may be given different PRO numbers but the UNQ number is unique for any given DNA and the encoded protein, and will not be changed.
  • PRO protein encoded by the PRO DNA as well as all further native homologues and variants included in the foregoing definition of PRO polypeptides, will be referred to as "PRO" regardless of their origin or mode of preparation.
  • Tlie description below relates primarily to production of PRO polypeptides by culturing cells transformed or transfected with a vector containing nucleic acid encoding PRO polypeptides. It is, of course, contemplated that alternative methods that are well known in the art may be employed to prepare the PRO polypeptide.
  • the PRO polypeptide sequence, or portions thereof may be produced by direct peptide synthesis using solid-phase techniques. See, e.g., Stewart etal. Solid-Phase Peptide Synthesis (W.H. Freeman Co.: San Francisco, CA, 1969); Merrif ⁇ eld, J. Am. Chem. Soc. 85: 2149-2154 (1963).
  • In vitro protein synthesis may be performed using manual techniques or by automation. Automated synthesis may be accomplished, for instance, with an Applied Biosystems
  • PRO polypeptide Synthesizer Frazier City, CA
  • Various portions ofthe PRO polypeptide may be chemically synthesized separately and combined using chemical or enzymatic methods to produce the full- length PRO polypeptide.
  • DNA encoding tlie PRO polypeptide may be obtained from a cDNA library prepared from tissue believed to possess the mRNA encoding the PRO polypeptide and to express it at a detectable level. Accordingly, DNAs encoding the human PRO polypeptide can be conveniently obtained from cDNA libraries prepared from human tissues, such as described in the Examples. The gene encoding the PRO polypeptide may also be obtained from a genomic library or by oligonucleotide synthesis. Libraries can be screened with probes (such as antibodies to the PRO polypeptide or oligonucleotides of at least about 20-80 bases) designed to identify the gene of interest or the protein encoded by it.
  • probes such as antibodies to the PRO polypeptide or oligonucleotides of at least about 20-80 bases
  • Screening the cDNA or genomic library with the selected probe may be conducted using standard procedures, such as described in Sambrook et al, supra.
  • An alternative means to isolate the gene encoding the PRO polypeptide is to use PCR methodology. Sambrook et al, supra; Dieffenbach et al, PCR Primer: A Laboratory Manual (New York: Cold Spring Harbor Laboratory Press, 1995).
  • the oligonucleotide sequences selected as probes should be of sufficient length and sufficiently unambiguous that false positives are minimized.
  • the oligonucleotide is preferably labeled such that it can be detected upon hybridization to DNA in the library being screened. Methods of labeling are well known in the art, and include the use of radio labels like 32 P-labeled ATP, biotinylation, or enzyme labeling. Hybridization conditions, including moderate stringency and high stringency, are provided in Sambrook et al, supra.
  • Sequences identified in such library screening methods can be compared and aligned to other known sequences deposited and available in public databases such as GenBank or other private sequence databases. Sequence identity (at either the amino acid or nucleotide level) within defined regions of tlie molecule or across the full-length sequence can be determined through sequence alignment using computer software programs such as
  • Nucleic acid having protein coding sequence may be obtained by screening selected cDNA or genomic libraries using the deduced amino acid sequence disclosed herein for the first time, and, if necessary, using conventional primer extension procedures as described in Sambrook et al, supra, to detect precursors and processing intermediates of mRNA that may not have been reverse-transcribed into cDNA.
  • Host cells are transfected or transformed with expression or cloning vectors described herein for PRO polypeptide production and cultured in conventional nutrient media modified as appropriate for inducing promoters, selecting transformanfs, or amplifying the genes encoding tlie desired sequences.
  • the culture conditions such as media, temperature, pH, and the like, can be selected by the skilled artisan without undue experimentation. In general, principles, protocols, and practical techniques for maximizing the productivity of cell cultures can be found in Mammalian Cell Biotechnology: A Practical Approach. M. Butler, ed. (IRL Press, 1991) and Sambrook et al, supra.
  • transfection is known to the ordinarily skilled artisan, for example, CaP0 4 treatment and electroporation.
  • transformation is performed using standard techniques appropriate to such cells.
  • the calcium treatment employing calcium chloride, as described in Sambrook et al, supra, or electroporation is generally used for prokaryotes or other cells that contain substantial cell-wall barriers.
  • Infection • withAgrobacterium tumefaciens is used for transformation of certain plant cells, as described by Shaw et al. , Gene. 23: 315 (1983) and WO 89/05859 published 29 June 1989.
  • DNA into cells such as by nuclear microinjection, electroporation, bacterial protoplast fusion with intact cells, or polycations, e.g., polybrene or polyo ⁇ uthine, may also be used.
  • polycations e.g., polybrene or polyo ⁇ uthine.
  • Suitable host cells for cloning or expressing the DNA in the vectors herein include prokaryote, yeast, or higher eukaryote cells.
  • Suitable prokaryotes include, but are not limited to, eubacteria, such as Gram-negative or Gram-positive organisms, for example, Enterobacteriaceae such as E. coli.
  • Various E. coli strains are publicly available, such as E. coliY ⁇ l strain MM294 (ATCC 31,446); £. co/ X1776 (ATCC 31,537); E. coli strain W3110 (ATCC 27,325); and K5 772 (ATCC 53,635).
  • Otlier suitable prokaryotic host cells include Enterobacteriaceae such as Escherichia, e.g., E. coli, Enterobacter, Erwinia, Klebsiella, Proteus, Salmonella, e.g., Salmonella typhimurium, Serratia, e.g., Serratia marcescans, and Shigella, as well as Bacilli such as B. subtilis and B. licheniformis (e.g.,B. licheniformis 41P disclosed in DD 266,710 published 12 April 1989), Pseudomonas such as
  • Strain W3110 is one particularly preferred host or parent host because it is a common host strain for recombinant DNA product fermentations. Preferably, the host cell secretes minimal amounts of proteolytic enzymes.
  • strain W3110 may be modified to effect a genetic mutation in the genes encoding proteins endogenous to tlie host, with examples of such hosts including E. coli W3110 strain 1 A2, which has the complete genotype tonA ; E. coli W3110 strain 9E4, which has the complete genotype tonA ptr3; E.
  • E. coli W3110 strain 27C7 (ATCC 55,244), which has the complete genotype tonAptr3phoA E15 (argF-lac)169 degP ompTkan';
  • E. coli W3110 strain 37D6 which has the complete genotype tonAptr3 phoA E15 (argF-lac)169 degP ompT rbs7 ilvG kaif;
  • E. coliW3110 strain 40B4 which is strain 37D6 with a non-kanamycin resistant degP deletion mutation; and an E. coli strain having mutant periplasmic protease disclosed in U.S. Patent No.4,946,783 issued 7 August 1990.
  • in vitro methods of cloning e.g., PCR or other nucleic acid polymerase reactions, are suitable.
  • eukaryotic microbes such as filamentous fungi or yeast are suitable cloning or expression hosts for vectors encoding the PRO polypeptide.
  • Saccharomyces cerevisiae is a commonly used lower eukaryotic host microorganism.
  • Others include Schizosaccharomyces pombe (Beach and Nurse, Nature, 290: 140 [1981]; EP 139,383 published 2 May 1985); Kluyveromyces hosts (U.S. Patent No. 4,943,529; Fleer et al,
  • Methylotropic yeasts are suitable herein and include, but are not limited to, yeast capable of growth on methanol selected from the genera consisting of Hansenula, Candida, Kloeckera, Pichia, Saccharomyces, Torulopsis, and Rhodotorula.
  • yeast capable of growth on methanol selected from the genera consisting of Hansenula, Candida, Kloeckera, Pichia, Saccharomyces, Torulopsis, and Rhodotorula.
  • yeast capable of growth on methanol selected from the genera consisting of Hansenula, Candida, Kloeckera, Pichia, Saccharomyces, Torulopsis, and Rhodotorula.
  • a list of specific species that are exemplary of this class of yeasts may be found in C. Anthony, The Biochemistry of Methylotrophs, 269 (1982).
  • Suitable host cells for the expression of nucleic acid encoding glycosylated PRO polypeptides are derived from multicellular organisms.
  • invertebrate cells include insect cells such as Drosophila S2 and Spodoptera Sf9, as well as plant cells.
  • useful mammalian host cell lines include Chinese hamster ovary
  • CHO monkey kidney CV1 line transformed by SV40 (COS-7, ATCC CRL 1651); human embryomc kidney line (293 or 293 cells subcloned for growth in suspension culture, Graham et al. . Gen. Virol, 36: 59 (1977)); Chinese hamster ovary cells/-DHFR (CHO, Urlaub and Chasin, Proc. Natl. Acad. Sci. USA. 77:4216 (1980)); mouse sertoli cells (TM4, Mather, Biol. Reprod., 23:243-251 (1980)); human lung cells (W138, ATCC CCL 75); human liver cells (Hep G2, HB 8065); and mouse mammary tumor
  • the nucleic acid e.g., cDNA or genomic DNA
  • the nucleic acid encoding the PRO polypeptide may be inserted into a replicable vector for cloning (amplification ofthe DNA) or for expression.
  • Various vectors are publicly available.
  • the vector may, for example, be in tlie form of a plasmid, cosmid, viral particle, or phage.
  • the appropriate nucleic acid sequence may be inserted into the vector by a variety of procedures. In general, DNA is inserted into an appropriate restriction endonuclease site(s) using techniques known in the art.
  • Vector components generally include, but are not limited to, one or more of a signal sequence if the sequence is to be secreted, an origin of replication, one or more marker genes, an enhancer element, a promoter, and a transcription termination sequence.
  • the PRO polypeptide may be produced recombinantly not only directly, but also as a fusion polypeptide with a heterologous polypeptide, which may be a signal sequence or other polypeptide having a specific cleavage site at tlie N-terminus of tlie mature protein or polypeptide.
  • a heterologous polypeptide which may be a signal sequence or other polypeptide having a specific cleavage site at tlie N-terminus of tlie mature protein or polypeptide.
  • the signal sequence may be a component ofthe vector, or it may be a part ofthe DNA encoding the PRO polypeptide that is inserted into tlie vector.
  • the signal sequence may be a prokaryotic signal sequence selected, for example, from the group of the alkaline phosphatase, penicillinase, lpp, or heat-stable enterotoxin II leaders.
  • the signal sequence may be, e.g., the yeast invertase leader, alpha factor leader (including Saccharomyces and Kluyveromyces a- factor leaders, the latter described in U.S. PatentNo.5,010,182), or acid phosphatase leader, the C. albicans glucoamylase leader (EP 362, 179 published 4 April 1990), or the signal described in WO 90/13646 published 15 November 1990.
  • mammalian signal sequences may be used to direct secretion ofthe protein, such as signal sequences from secreted polypeptides ofthe same or related species, as well as viral secretory leaders.
  • Both expression and cloning vectors contain a nucleic acid sequence that enables the vector to replicate in one or more selected host cells. Such sequences are well known for a variety of bacteria, yeast, and viruses.
  • the origin of replication from the plasmid pBR322 is suitable for most Gram-negative bacteria, the 2 ⁇ plasmid origin is suitable for yeast, and various viral origins (SV40, polyoma, adenovirus, VSV, or BPV) are useful for cloning vectors in mammalian cells.
  • Expression and cloning vectors will typically contain a selection gene, also termed a selectable marker.
  • Typical selection genes encode proteins that (a) confer resistance to antibiotics or other toxins, e.g., ampicillim neomycin, methotrexate, or tetracycline, (b) complement auxotrophic deficiencies, or (c) supply critical nutrients not available from complex media, e.g., the gene encoding D-alanine racemase for Bacilli.
  • suitable selectable markers for mammalian cells are those that enable the identification of cells competent to take up the nucleic acid encoding the PRO polypeptide such as DHFR or thymidine kinase.
  • An appropriate host cell when wild-type DHFR is employed is tlie CHO cell line deficient in DHFR activity, prepared and propagated as described by Urlaub et al, Proc. Natl. Acad. Sci. USA, 77: 4216 (1980).
  • a suitable selection gene for use in yeast is the trpl gene present in the yeastplasmid YRp7. Stinchcomb et al. , Nature.282: 39 (1979);
  • the trpl gene provides a selection marker for a mutant strain of yeast lacking the ability to grow in tryptophan, for example, ATCC No. 44076 or
  • Expression and cloning vectors usually contain a promoter operably linked to the nucleic acid sequence encoding the PRO polypeptide to direct mRNA synthesis. Promoters recognized by a variety of potential host cells are well known. Promoters suitable for use with prokaryotic hosts include the ⁇ -lactamase and lactose promoter systems (Chang et al, Nature, 275: 615 (1978); Goeddel et al, Nature, 281: 544 (1979)), alkaline phosphatase, a tryptophan (tip) promoter system (Goeddel, Nucleic Acids Res..
  • Promoters for use in bacterial systems also will contain a Shine-Dalgarno (S .D.) sequence operably linked to the DNA encoding the PRO polypeptide.
  • suitable promoting sequences for use with yeast hosts include tlie promoters for 3- phosphoglycerate kinase QHitzeman et al, J. Biol. Chem..255: 2073 (1980)) or other glycolytic enzymes (Hess et al. J. Adv. Enzyme Reg..7: 149 (1968); Holland, Biochemistry.17: 4900 (1978)), such as enolase, glyceraldehyde-
  • yeast promoters that are inducible promoters having tlie additional advantage of transcription controlled by growth conditions are the promoter regions for alcohol dehydrogenase 2, isocytochrome C, acid phosphatase, degradative enzymes associated with nitrogen metabolism, metallothionein, glyceraldehyde-3-phos- phate dehydrogenase, and enzymes responsible for maltose and galactose utilization. Suitable vectors and promoters for use in yeast expression are further described in EP 73,657.
  • PRO nucleic acid transcription from vectors in mammalian host cells is controlled, for example, by promoters obtained from the genomes of viruses such as polyoma virus, fowlpox virus (UK 2,211,504 published
  • adenovirus such as Adenovirus 2
  • bovine papilloma virus such as Adenovirus 2
  • bovine papilloma virus such as avian sarcoma virus
  • cytomegalovirus such as a retrovirus
  • a retrovirus such as hepatitis-B virus
  • Simian Virus 40 SV40
  • heterologous mammalian promoters e.g. , the actin promoter or an immunoglobulin promoter
  • heat-shock promoters provided such promoters are compatible with the host cell systems.
  • Enhancers are cis-acting elements of DNA, usually about from 10 to 300 bp, that act on a promoter to increase its transcription.
  • Many enhancer sequences are now known from mammalian genes (globin, elastase, albumin, ⁇ -fetoprotein, and insulin). Typically, however, one will use an enhancer from a eukaryotic cell virus.
  • Examples include the SV40 enhancer on the late side ofthe replication origin (bp 100-270), the cytomegalovirus early promoter enhancer, the polyoma enhancer on the late side ofthe replication origin, and adenovirus enhancers.
  • the enhancer may be spliced into the vector at a position 5' or 3' to the sequence coding for PRO polypeptides, but is preferably located at a site 5' from the promoter.
  • Expression vectors used in eukaryotic host cells will also contain sequences necessary for the termination of transcription and for stabilizing the mRNA. Such sequences are commonly available from the 5' and, occasionally 3', untranslated regions of eukaryotic or viral DNAs or cDNAs. These regions contain nucleotide segments transcribed as polyadenylated fragments in the untranslated portion of the mRNA encoding the PRO polypeptide.
  • Gene amplification and/or expression may be measured in a sample directly, for example, by conventional
  • Southern blotting Northern blotting to quantitate the transcription of mRNA (Thomas, Proc. Natl. Acad. Sci. USA, 77:5201-5205 (1980)), dot blotting (DNA analysis), or in situ hybridization, using an appropriately labeled probe, based on tlie sequences provided herein.
  • antibodies may be employed that can recognize specific duplexes, including DNA duplexes, RNA duplexes, and DNA-RNA hybrid duplexes or DNA-protein duplexes. The antibodies in turn may be labeled and the assay may be carried out where the duplex is bound to a surface, so that upon the formation of duplex on the surface, the presence of antibody bound to the duplex can be detected.
  • Gene expression may be measured by immunological methods, such as ihimunohistochemical staining of cells or tissue sections and assay of cell culture or body fluids, to quantitate directly the expression of gene product.
  • Antibodies useful for immunohistochemical staining and/or assay of sample fluids may be either monoclonal or polyclonal, and may be prepared in any mammal.
  • tlie antibodies may be prepared against a native-sequence PRO polypeptide or against a synthetic peptide based on the DNA sequences provided herein or against exogenous sequence fused to DNA encoding the PRO polypeptide and encoding a specific antibody epitope.
  • PRO polypeptides may be recovered from culture medium or from host cell lysates. If membrane-bound, it can be released from the membrane using a suitable detergent solution (e.g., TRITON-XTM 100) or by enzymatic cleavage. Cells employed in expression of nucleic acid encoding the PRO polypeptide can be disrupted by various physical or chemical means, such as freeze-thaw cycling, sonication, mechanical disruption, or cell-lysing agents. It may be desired to purify the PRO polypeptide from recombinant cell proteins or polypeptides.
  • a suitable detergent solution e.g., TRITON-XTM 100
  • Cells employed in expression of nucleic acid encoding the PRO polypeptide can be disrupted by various physical or chemical means, such as freeze-thaw cycling, sonication, mechanical disruption, or cell-lysing agents. It may be desired to purify the PRO polypeptide from recombinant cell proteins or polypeptides.
  • the following procedures are exemplary of suitable purification procedures: by f actionation on an ion-exchange column; ethanol precipitation; reverse phase HPLC; chromatography on silica or on a cation-exchange resin such as DEAE; chromatofocusing; SDS-PAGE; ammonium sulfate precipitation; gel filtration using, for example, Sephadex G-75; protein A Sepharose columns to remove contaminants such as IgG; and metal chelating columns to bind epitope-tagged forms ofthe PRO polypeptide.
  • Various methods of protein purification may be employed and such methods are known in the art and described, for example, in Deutscher, Methods in
  • assays can be used to test the polypeptide herein for cardiovascular, endothelial, and angiogenic activity.
  • Such assays include those provided in the Examples below.
  • Assays for testing for endothelin antagonist activity include a rat heart ventricle binding assay where the polypeptide is tested for its ability to inhibit iodinized endothelin- 1 binding in a receptor assay, an endothelin receptor binding assay testing for intact cell binding of radiolabeled endothelin- 1 using rabbit renal artery vascular smooth muscle cells, an inositol phosphate accumulation assay where functional activity is determined in Rat-1 cells by measuring infra-cellular levels of second messengers, an arachidonic acid release assay that measures tlie ability of added compounds to reduce endothelin-stimulated arachidonic acid release in cultured vascular smooth muscles, in vitro (isolated vessel) studies using endothelium from male New Zealand rabbits, and in vivo studies using male Sprague-Dawley rats.
  • Assays for tissue generation activity include, without limitation, those described in WO 95/16035 (bone, cartilage, tendon); WO 95/05846 (nerve, neuronal), and WO 91/07491 (skin, endothelium).
  • Assays for wound-healing activity include, for example, those described in Winter, Epidermal Wound Healing. Maibach, HI and Rovee, DT, eds. (Year Book Medical Publishers, Inc., Chicago), pp.71- 112, as modified by the article of Eaglstein and Mertz, J. Invest. Dermatol.. 71: 382-384 (1978).
  • An assay to screen for a test molecule relating to a PRO polypeptide that binds an endothelin B ! (ETBj) receptor polypeptide and modulates signal transduction activity involves providing a host cell transformed with a DNA encoding endothelin : receptor polypeptide, exposing the cells to the test candidate, and measuring endothelin B, receptor signal transduction activity, as described, e.g., in U.S. Pat. No. 5,773,223.
  • In vitro assays include induction of spreading of adult rat cardiac myocytes.
  • ventricular myocytes are isolated from a single (male Sprague-Dawley) rat, essentially following a modification ofthe procedure described in detail by Piper et al. , "Adult ventricular rat heart muscle cells” in Cell Culture Techniques in Heart and Vessel Research. H.M. Piper, ed. (Berlin: Springer- Verlag, 1990), pp. 36-60.
  • This procedure permits tlie isolation of adult ventricular myocytes and the long-term culture of these cells in the rod-shaped phenotype.
  • Phenylephrine and Prostaglandin F 2 ⁇ (PGF 2 « ) have been shown to induce a spreading response in these adult cells.
  • PGF 2 « or PGF 2 render analogs
  • an in vivo assay is a test for inhibiting cardiac hypertrophy induced by fluprostenol in vivo.
  • This pharmacological model tests the ability ofthe PRO polypeptide to inhibit cardiac hypertrophy induced in rats (e.g., male Wistar or Sprague-Dawley) by subcutaneous injection of fluprostenol (an agonist analog of PGF 2 till). It is known that rats with pathologic cardiac hypertrophy induced by myocardial infarction have chronically elevated levels of extractable PGF 2o in their myocardium.
  • rats with pathologic cardiac hypertrophy induced by myocardial infarction have chronically elevated levels of extractable PGF 2o in their myocardium.
  • Lai et al Am. J. Phvsiol. (Heart Circ. PhvsioL). 271: H2197- H2208 (1996).
  • the effects ofthe PRO polypeptide on cardiac hypertrophy are determined by measuring tlie weight of heart, ventricles, and left ventricle (normalized by body weight) relative to fluprostenol-treated rats not receiving the PRO polypeptide.
  • an in vivo assay is the pressure-overload cardiac hypertrophy assay.
  • pressure-overload cardiac hypertrophy assay For in vivo testing it is common to induce pressure-overload cardiac hypertrophy by constriction ofthe abdominal aorta of test animals.
  • rats e.g., male Wistar or Sprague-Dawley
  • the abdominal aorta of each rat is narrowed down just below the diaphragm. Beznak M., Can. J. Biochem. Phvsiol., 33: 985-94 (1955).
  • the aorta is exposed through a surgical incision, and a blunted needle is placed next to the vessel.
  • the aorta is constricted with a ligature of silk thread around the needle, which is immediately removed and which reduces the lumen ofthe aorta to the diameter ofthe needle.
  • This approach is described, for example, in Rossi et al, Am. Heart J.. 124: 700-709 (1992) and O'Rourke and Reibel, P.S.E.M.B.. 200: 95-100 (1992).
  • MI myocardial infarction
  • Animal models of tumors and cancers include both non-recombinant and recombinant (transgenic) animals.
  • Non-recombinant animal models include, for example, rodent, e.g. , murine models.
  • Such models can be generated by introducing tumor cells into syngeneic mice using standard techniques, e.g., subcutaneous injection, tail vein injection, spleen implantation, intraperitoneal implantation, implantation under the renal capsule, or orthopin implantation, e.g., colon cancer cells implanted in colonic tissue.
  • standard techniques e.g., subcutaneous injection, tail vein injection, spleen implantation, intraperitoneal implantation, implantation under the renal capsule, or orthopin implantation, e.g., colon cancer cells implanted in colonic tissue.
  • the autosomal recessive nu gene has been introduced into a very large number of distinct congenic strains of nude mouse, including, for example, ASW, A/He, AKR, BALB/c, B10.LP, C17, C3H, C57BL, C57, CBA, DBA, DDD, I/st, NC, NFR, NFS, NFS/N, NZB, NZC, NZW, P, RIII, and S JL.
  • a wide variety of other animals with inherited immunological defects other than tlie nude mouse have been bred and used as recipients of tumor xenografts. For further details see, e.g., The Nude Mouse in Oncology Research, E. Boven and B. Winograd, eds. (CRC Press, Inc., 1991).
  • the cells introduced into such animals can be derived from known tumor/cancer cell lines, such as any ofthe above-listed tumor cell lines, and, for example, the B104-1-1 cell line (stable NTH-3T3 cell line transfected with the neu protooncogene); r ⁇ s-transfected NTH-3T3 cells; Caco-2 (ATCC HTB-37); or a moderately well- differentiated grade II human colon adenocarcinoma cell line, HT-29 (ATCC HTB-38); or from tumors and cancers. Samples of tumor or cancer cells can be obtained from patients undergoing surgery, using standard conditions involving freezing and storing in liquid nitrogen. Karmali et al, Br. J. Cancer. 48: 689-696 (1983).
  • Tumor cells can be introduced into animals such as nude mice by a variety of procedures.
  • the subcutaneous (s.c.) space in mice is very suitable for tumor implantation.
  • Tumors can be transplanted s.c. as solid blocks, as needle biopsies by use of a frochar, or as cell suspensions.
  • a frochar or as cell suspensions.
  • tumor tissue fragments of suitable size are introduced into the s.c. space.
  • Cell suspensions are freshly prepared from primary tumors or stable tumor cell lines, and injected subcutaneously.
  • Tumor cells can also be injected as subdermal implants. In this location, the inoculum is deposited between the lower part ofthe dermal connective tissue and the s.c. tissue.
  • Animal models of breast cancer can be generated, for example, by implanting rat neuroblastoma cells (from which the neu oncogene was initially isolated), or new-transformed NIH-3T3 cells into nude mice, essentially as described by Drebin et al. Proc. Nat. Acad. Sci. USA. 83: 9129-9133 (1986).
  • animal models of colon cancer can be generated by passaging colon cancer cells in animals, e.g. , nude mice, leading to the appearance of tumors in these animals.
  • An or hotopic transplant model of human colon cancer in nude mice has been described, for example, by Wang et al, Cancer Research.54: 4726-4728 (1994) and Too et al, Cancer Research, 55: 681-684 (1995). This model is based on the so-called “METAMOUSETM” sold by AntiCancer, Inc., (San Diego, California).
  • Tumors that arise in animals can be removed and cultured in vitro. Cells from the in vitro cultures can then be passaged to animals. Such tumors can serve as targets for further testing or drug screening. Alternatively, the tumors resulting from the passage can be isolated and RNA from pre-passage cells and cells isolated after one or more rounds of passage analyzed for differential expression of genes of interest. Such passaging techniques can be performed with any known tumor or cancer cell lines.
  • Meth A, CMS4, CMS5, CMS21, and WEHI-164 are chemically induced fibrosarcomas of BALB/c female mice (DeLeo et al, J. Exp. Med.. 146: 720 (1977)), which provide a highly controllable model system for studying tlie anti-tumor activities of various agents.
  • mice are then infected subcutaneously with 10 to 100 ⁇ l of the cell suspension, allowing one to three weeks for a tumor to appear.
  • the Lewis lung (3LL) carcinoma of mice which is one of the most thoroughly studied experimental tumors, can be used as an investigational rumor model. Efficacy in this tumor model has been correlated with beneficial effects in the treatment of human patients diagnosed with small-cell carcinoma of tlie lung
  • SCCL Single cell lung cancer
  • This tumor can be introduced in normal mice upon injection of tumor fragments from an affected mouse or of cells maintained in culture. Zupi etal, Br. J. Cancer, 41: suppl.4, 30 (1980). Evidence indicates that tumors can be started from injection of even a single cell and that a very high proportion of infected tumor cells survive. For further information about this tumor model see, Zacharski, Haemostasis, 16: 300-320 (1986).
  • One way of evaluating the efficacy of a test compound in an animal model with an implanted tumor is to measure the size of the tumor before and after treatment. Traditionally, the size of implanted tumors has been measured with a slide caliper in two or three dimensions.
  • the measure limited to two dimensions does not accurately reflect the size ofthe tumor; therefore, it is usually converted into the corresponding volume by using a mathematical formula. However, the measurement of tumor size is very inaccurate.
  • the therapeutic effects of a drug candidate can be better described as treatment-induced growth delay and specific growth delay.
  • Another important variable in the description of tumor growth is the tumor volume doubling time.
  • Computer programs for the calculation and description of tumor growth are also available, such as the program reported by Rygaard and Spang-Thomsen. Proc.6th Int. Workshop on Immune-Deficient Animals, WuandShengeds. (Basel, 1989), ⁇ .301. It is noted, however, that necrosis and inflammatory responses following treatment may actually result in an increase in tumor size, at least initially. Therefore, these changes need to be carefully monitored, by a combination of a morphometric method and flow cytometric analysis.
  • recombinant (transgenic) animal models can be engineered by introducing tlie coding portion of the PRO gene identified herein into the genome of animals of interest, using standard techniques for producing transgenic animals.
  • Animals that can serve as a target for transgenic manipulation include, without limitation, mice, rats, rabbits, guinea pigs, sheep, goats, pigs, and non-human primates, e.g., baboons, chimpanzees and monkeys.
  • Techniques known in the art to introduce a transgene into such animals include pronucleic microinjection (U.S. Patent No. 4,873,191); retrovirus-mediated gene transfer into germ lines (e.g., Van der Putten et al, Proc. Natl.
  • transgenic animals include those that carry the transgene only in part of their cells ("mosaic animals").
  • the transgene can be integrated either as a single transgene, or in concatamers, e.g. , head-to-head or head-to-tail tandems. Selective introduction of a transgene into a particular cell type is also possible by following, for example, tlie technique of Lasko et al, Proc. Natl. Acad. Sci. USA.89: 6232- 636 (1992).
  • the expression ofthe transgene in transgenic animals can be monitored by standard techniques. For example, Southern blot analysis or PCR amplification can be used to verify the integration ofthe transgene.
  • the level of mRNA expression can then be analyzed using techniques such as in situ hybridization, Northern blot analysis, PCR, or immunocytochemistry. The animals are further examined for signs of tumor or cancer development.
  • "knock-out" animals can be constructed that have a defective or altered gene encoding a PRO polypeptide identified herein, as a result of homologous recombination between the endogenous gene encoding the PRO polypeptide and altered genomic DNA encoding the same polypeptide introduced into an embryonic cell ofthe animal.
  • cDNA encoding a particular PRO polypeptide can be used to clone genomic DNA encoding that polypeptide in accordance with established techniques.
  • a portion ofthe genomic DNA encoding a particular PRO polypeptide can be deleted or replaced with another gene, such as a gene encoding a selectable marker that can be used to monitor integration.
  • flanking DNA typically, several kilobases of unaltered flanking DNA (both at the 5' and 3' ends) are included in the vector. See, e.g. , Thomas and Capecchi, Cell, 51: 503 (1987) for a description of homologous recombination vectors.
  • the vector is introduced into an embryonic stem cell line (e.g., by electroporation) and cells in which the introduced DNA has homologously recombined with the endogenous DNA are selected. See, e.g., Li et al, Cell. 69: 915 (1992).
  • the selected cells are then injected into a blastocyst of an animal (e.g. , a mouse or rat) to form aggregation chimeras.
  • a chimeric embryo can then be implanted into a suitable pseudopregnant female foster animal and the embryo brought to term to create a "knock-out" animal.
  • Progeny harboring the homologously recombined DNA in their germ cells can be identified by standard techniques and used to breed animals in which all cells of the animal contain the homologously recombined DNA.
  • Knockout animals can be characterized, for instance, by their ability to defend against certain pathological conditions and by their development of pathological conditions due to absence ofthe PRO polypeptide.
  • SCC feline oral squamous cell carcinoma
  • Feline oral SCC is a highly invasive, malignant tumor that is the most common oral malignancy of cats, accounting for over 60% ofthe oral tumors reported in this species. It rarely metastasizes to distant sites, although this low incidence of metastasis may merely be a reflection of the short survival times for cats with this tumor.
  • These tumors are usually not amenable to surgery, primarily because ofthe anatomy ofthe feline oral cavity. At present, there is no effective treatment for this tumor.
  • each cat Prior to entry into the study, each cat undergoes complete clinical examination and biopsy, and is scanned by computed tomography (CT). Cats diagnosed with sublingual oral squamous cell tumors are excluded from the study. The tongue can become paralyzed as a result of such tumor, and even if the treatment kills the tumor, the animals may not be able to feed themselves.
  • CT computed tomography
  • Each cat is treated repeatedly, over a longer period of time. Photographs ofthe tumors will be taken daily during the treatment period, and at each subsequent recheck.
  • CT scans and thoracic radiograms are evaluated every 8 weeks thereafter. The data are evaluated for differences in survival, response, and toxicity as compared to control groups. Positive response may require evidence of tumor regression, preferably with improvement of quality of life and/or increased life span.
  • fibrosarcoma adenocarcinoma
  • lymphoma chondroma
  • leiomyosarcoma of dogs, cats, and baboons
  • mammary adenocarcinoma in dogs and cats is a preferred model as its appearance and behavior are very similar to those in humans.
  • the use of this model is limited by the rare occurrence of this .type of tumor in animals.
  • tlie cardiovascular, endothelial, and angiogenic assays herein can be verified by further studies, such as by determining mRNA expression in various human tissues.
  • gene amplification and/or gene expression in various tissues may be measured by conventional Southern blotting, Northern blotting to quantitate the transcription of mRNA (Thomas, Proc. Natl. Acad. Sci. USA.77:5201-5205 (1980)), dot blotting QDNA analysis), or in situ hybridization, using an appropriately labeled probe, based on the sequences provided herein.
  • antibodies may be employed that can recognize specific duplexes, including DNA duplexes, RNA duplexes, and DNA-RNA hybrid duplexes or DNA-protein duplexes.
  • Gene expression in various tissues may be measured by immunological methods, such as immmiohistochemical staining of tissue sections and assay of cell culture or body fluids, to quantitate directly tlie expression of gene product.
  • Antibodies useful for immunohistochemical staining and/or assay of sample fluids may be either monoclonal or polyclonal, and may be prepared in any mammal. Conveniently, the antibodies may be prepared against a native-sequence PRO polypeptide or against a synthetic peptide based on the DNA sequences provided herein or against exogenous sequence fused to PRO DNA and encoding a specific antibody epitope.
  • General techniques for generating antibodies, and special protocols for in situ hybridization are provided hereinbelow.
  • cardiovascular, endothelial, and angiogenic study can be further verified by antibody binding studies, in which the ability of anti-PRO antibodies to inhibit the effect of the PRO polypeptides on endothelial cells or other cells used in the cardiovascular, endothelial, and angiogenic assays is tested.
  • Exemplary antibodies include polyclonal, monoclonal, humanized, bispecific, and heteroconjugate antibodies, the preparation of which will be described hereinbelow.
  • Antibody binding studies may be carried out in any known assay method, such as competitive binding assays, direct and indirect sandwich assays, and immunoprecipitation assays. Zola, Monoclonal Antibodies: A Manual of Techniques (CRC Press, Inc., 1987), pp.147-158.
  • Competitive binding assays rely on the ability of a labeled standard to compete with the test sample analyte for binding with a limited amount of antibody.
  • the amount of target protein in the test sample is inversely proportional to the amount of standard that becomes bound to the antibodies.
  • the antibodies preferably are insolubilized before or after the competition, so that the standard and analyte that are bound to the antibodies may conveniently be separated from the standard and analyte that remain unbound.
  • Sandwich assays involve the use of two antibodies, each capable of binding to a different mimunogenic portion, or epitope, ofthe protein to be detected.
  • the test sample analyte is bound by a first antibody that is immobilized on a solid support, and thereafter a second antibody binds to the analyte, thus forming an insoluble three-part complex.
  • the second antibody may itself be labeled with a detectable moiety (direct sandwich assays) or may be measured using an anti-immunoglobulin antibody that is labeled with a detectable moiety (indirect sandwich assay) .
  • sandwich assay is an ELISA assay, in which case the detectable moiety is an enzyme.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • Medicinal Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • General Health & Medical Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Cardiology (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Vascular Medicine (AREA)
  • Hospice & Palliative Care (AREA)
  • Urology & Nephrology (AREA)
  • Ophthalmology & Optometry (AREA)
  • Pain & Pain Management (AREA)
  • Rheumatology (AREA)
  • Dermatology (AREA)
  • Toxicology (AREA)
  • Zoology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Peptides Or Proteins (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)
EP01951036A 2000-07-20 2001-07-09 Zusammensetzungen und verfahren für die diagnose und behandlung von angiogenese-assoziierten erkrankungen Withdrawn EP1309685A2 (de)

Applications Claiming Priority (73)

Application Number Priority Date Filing Date Title
US866028 1977-12-30
US854280 1997-05-09
US21955600P 2000-07-20 2000-07-20
US219556P 2000-07-20
US22062400P 2000-07-25 2000-07-25
US22066400P 2000-07-25 2000-07-25
US220624P 2000-07-25
US220664P 2000-07-25
WOPCT/US20/07107 2000-07-28
PCT/US2000/020710 WO2001009327A2 (en) 1999-07-28 2000-07-28 Method of preventing the injury or death of retinal cells and treating ocular diseases
US22269500P 2000-08-02 2000-08-02
US222695P 2000-08-02
US09/643,657 US6642024B1 (en) 1998-01-29 2000-08-17 Guanylate-binding protein
US643657 2000-08-17
PCT/US2000/023522 WO2001016319A2 (en) 1999-08-31 2000-08-23 Compositions and methods for the treatment of immune related diseases
WOPCT/US00/23522 2000-08-23
WOPCT/US00/23328 2000-08-24
PCT/US2000/023328 WO2001016318A2 (en) 1999-09-01 2000-08-24 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US23097800P 2000-09-07 2000-09-07
US230978P 2000-09-07
US66535000A 2000-09-18 2000-09-18
US66461000A 2000-09-18 2000-09-18
US665350 2000-09-18
US664610 2000-09-18
US24292200P 2000-10-24 2000-10-24
US242922P 2000-10-24
US70923800A 2000-11-08 2000-11-08
PCT/US2000/030952 WO2001049715A2 (en) 2000-01-06 2000-11-08 Methods and compositions for inhibiting neoplastic cell growth
WOPCT/US00/30952 2000-11-08
US709238 2000-11-08
PCT/US2000/030873 WO2001040465A2 (en) 1999-11-30 2000-11-10 Compositions and methods for the treatment of immune related diseases
WOPCT/US00/30873 2000-11-10
WOPCT/US00/32678 2000-12-01
PCT/US2000/032678 WO2001040466A2 (en) 1999-12-01 2000-12-01 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US09/747,259 US6569645B2 (en) 1999-05-14 2000-12-20 IL-17 homologous polypeptides and therapeutic uses thereof
PCT/US2000/034956 WO2001046420A2 (en) 1999-12-23 2000-12-20 Il-17 and il-17r homologous polypeptides and therapeutic uses thereof
WOPCT/US00/34956 2000-12-20
US747259 2000-12-20
US767609 2001-01-22
US09/767,609 US20020042367A1 (en) 1997-11-25 2001-01-22 Fibroblast growth factor-19 (FGF-19) nucleic acids and polypeptides and methods of use for the treatment of obesity and related disorders
US79649801A 2001-02-28 2001-02-28
WOPCT/US01/06520 2001-02-28
US796498 2001-02-28
PCT/US2001/006520 WO2001068848A2 (en) 2000-03-01 2001-02-28 Secreted and transmembrane polypeptides and nucleic acids encoding the same
PCT/US2001/006666 WO2001066740A2 (en) 2000-03-03 2001-03-01 Compositions and methods for the treatment of immune related diseases
WOPCT/US01/06666 2001-03-01
US80270601A 2001-03-09 2001-03-09
US802706 2001-03-09
US80868901A 2001-03-14 2001-03-14
US808689 2001-03-14
US816744 2001-03-22
US09/816,744 US6579520B2 (en) 1998-05-15 2001-03-22 IL-17 related mammalian cytokine polypeptides (IL-17E)
US828366 2001-04-05
US09/828,366 US20020010137A1 (en) 1997-09-18 2001-04-05 Methods and compositions for inhibiting neoplastic cell growth
US854208 2001-05-10
US09/854,280 US7115398B2 (en) 1998-05-15 2001-05-10 IL-17 homologous polypeptides and therapeutic uses thereof
US09/854,208 US7217412B2 (en) 1998-05-15 2001-05-10 IL-17C related mammalian cytokine polypeptides
PCT/US2001/017092 WO2001092331A2 (en) 2000-05-30 2001-05-25 Compositions and methods for the treatment of immune related diseases
US866034 2001-05-25
US09/866,028 US6642360B2 (en) 1997-12-03 2001-05-25 Secreted polypeptides that stimulate release of proteoglycans from cartilage
US09/866,034 US20030170864A1 (en) 2000-05-30 2001-05-25 Secreted and transmembrane polypeptides and nucleic acids encoding the same
WOPCT/US01/17092 2001-05-25
US09/870,574 US6551799B2 (en) 1999-12-07 2001-05-30 Interleukin-22 polypeptides, nucleic acids encoding the same and methods for the treatment of pancreatic disorders
US870574 2001-05-30
PCT/US2001/017443 WO2002016611A2 (en) 2000-08-24 2001-05-30 Interleukin-22 polypeptides, nucleic acids encoding the same and methods for the treatment of pancreatic disorders
WOPCT/US01/17443 2001-05-30
PCT/US2001/017800 WO2001093983A1 (en) 2000-06-02 2001-06-01 Secreted and transmembrane polypeptides and nucleic acids encoding the same
WOPCT/US01/17800 2001-06-01
WOPCT/US01/19692 2001-06-20
PCT/US2001/019692 WO2002000690A2 (en) 2000-06-23 2001-06-20 Compositions and methods for the diagnosis and treatment of disorders involving angiogenesis
WOPCT/US01/00000 2001-06-28
PCT/US2001/021735 WO2002008284A2 (en) 2000-07-20 2001-07-09 Compositions and methods for the diagnosis and treatment of disorders involving angiogenesis
2003-06-19

Publications (1)

Publication Number Publication Date
EP1309685A2 true EP1309685A2 (de) 2003-05-14

Family

ID=56290165

Family Applications (1)

Application Number Title Priority Date Filing Date
EP01951036A Withdrawn EP1309685A2 (de) 2000-07-20 2001-07-09 Zusammensetzungen und verfahren für die diagnose und behandlung von angiogenese-assoziierten erkrankungen

Country Status (5)

Country Link
EP (1) EP1309685A2 (de)
JP (5) JP2004516013A (de)
AU (1) AU2001271973A1 (de)
CA (1) CA2416538A1 (de)
WO (1) WO2002008284A2 (de)

Families Citing this family (75)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA2301173C (en) 1997-09-12 2015-04-14 Biogen, Inc. Cysteine rich receptors-train
US8007798B2 (en) 1997-11-21 2011-08-30 Genentech, Inc. Treatment of complement-associated disorders
US8088386B2 (en) 1998-03-20 2012-01-03 Genentech, Inc. Treatment of complement-associated disorders
US7419663B2 (en) 1998-03-20 2008-09-02 Genentech, Inc. Treatment of complement-associated disorders
US7192589B2 (en) * 1998-09-16 2007-03-20 Genentech, Inc. Treatment of inflammatory disorders with STIgMA immunoadhesins
US7771719B1 (en) 2000-01-11 2010-08-10 Genentech, Inc. Pharmaceutical compositions, kits, and therapeutic uses of antagonist antibodies to IL-17E
ATE365800T1 (de) 1998-05-15 2007-07-15 Genentech Inc Therapeutische verwendungen von il-17 homologe polypeptide
US20030190669A1 (en) 1998-12-30 2003-10-09 Genentech, Inc. Secreted and transmembrane polypeptides and nucleic acids encoding the same
US20030095967A1 (en) 1999-01-25 2003-05-22 Mackay Fabienne BAFF, inhibitors thereof and their use in the modulation of B-cell response and treatment of autoimmune disorders
US20030119113A1 (en) 1999-07-20 2003-06-26 Genentech, Inc. Secreted and transmembrane polypeptides and nucleic acids encoding the same
EP1623992A3 (de) * 1999-09-01 2006-05-10 Genentech, Inc. Ausgeschiedene und Transmembranpolypeptide und dafür kodierende Nukleinsäure
EP2258848B1 (de) * 1999-12-23 2014-03-05 Genentech, Inc. IL-17-homologe Polypeptide und ihre therapeutische Verwendung
US20040043397A1 (en) 2000-01-11 2004-03-04 Genentech, Inc. IL-17 homologous polypeptides and therapeutic uses thereof
US7718397B2 (en) 2000-03-21 2010-05-18 Genentech, Inc. Nucleic acids encoding receptor for IL-17 homologous polypeptides and uses thereof
WO2001081401A1 (fr) 2000-04-21 2001-11-01 Fuso Pharmaceutical Industries, Ltd. Nouvelles collectines
US20030096969A1 (en) 2000-06-02 2003-05-22 Genentech, Inc. Secreted and transmembrane polypeptides and nucleic acids encoding the same
CA2648051A1 (en) * 2000-06-23 2002-01-03 Genentech, Inc. Compositions and methods for the diagnosis and treatment of disorders involving angiogenesis
EP2042597B1 (de) * 2000-06-23 2014-05-07 Genentech, Inc. Zusammensetzungen und Verfahren zur Diagnose und Behandlung von Erkrankungen in Verbindung mit Angiogenese
JP2004506413A (ja) * 2000-06-23 2004-03-04 ジェネンテック・インコーポレーテッド 血管形成に関与する疾患の診断と治療のための組成物と方法
US7265210B2 (en) 2000-09-15 2007-09-04 Genentech, Inc. Anti-PRO9821 antibodies
US6630325B1 (en) * 2000-10-19 2003-10-07 Maine Medical Center Research Institute Compositions, methods and kits relating to remodel
EP1332157A2 (de) 2000-11-03 2003-08-06 The Regents of the University of California Prokineticin polypeptide, verwandte zusammensetzungen und methoden
AU2002217838A1 (en) 2000-11-14 2002-05-27 Bristol-Myers Squibb Company A human serpin secreted from lymphoid cells LSI-01
US7135334B2 (en) 2001-06-20 2006-11-14 Genentech, Inc. PRO20044 nucleic acids
US20060263770A1 (en) * 2002-03-22 2006-11-23 Marco Colonna Novel receptor trem (triggering receptor expressed on myeloid cells) and uses thereof
CA2480419A1 (en) * 2002-03-28 2003-10-09 Yamanouchi Pharmaceutical Co., Ltd. Antiopoietin-related growth factor
MXPA04010092A (es) * 2002-04-16 2004-12-13 Genentech Inc Composiciones y metodos para el diagnostico y tratamiento de tumores.
EP2305710A3 (de) 2002-06-03 2013-05-29 Genentech, Inc. Synthetische Antikörperphagenbibliotheken
AU2003261911A1 (en) * 2002-09-04 2004-03-29 Takeda Pharmaceutical Company Limited Use of disease-associated gene
EP2116616A3 (de) * 2002-09-11 2010-03-17 Genentech, Inc. In aktivierten T-Zellen differentiell exprimierte Gene und deren Verwendungen
EP2500438A3 (de) * 2002-09-25 2012-11-28 Genentech, Inc. Neue Zusammensetzungen und Verfahren zur Behandlung von Psoriasis
EP2526960A1 (de) 2003-03-12 2012-11-28 Genentech, Inc. Verwendung von BV8 und/oder EG-VEGF zum Fördern der Hämatopoiese
MXPA06000347A (es) 2003-07-08 2006-03-28 Genentech Inc Polipeptidos heterologos il-17 a/f y usos terapeuticos de los mismos.
US20100031378A1 (en) 2008-08-04 2010-02-04 Edwards Joel A Novel gene disruptions, compositions and methods relating thereto
MXPA06011851A (es) 2004-04-14 2006-12-14 Genentech Inc Composiciones y metodos para moduilar el desarrollo vascular.
PT1781682E (pt) 2004-06-24 2013-05-14 Mayo Foundation B7-h5, um polipéptido coestimulante
CA2568886A1 (en) 2004-07-02 2006-02-09 Genentech, Inc. Compositions and methods for treatment of non-hodgkin's lymphoma
JP5060293B2 (ja) 2004-08-03 2012-10-31 バイオジェン・アイデック・エムエイ・インコーポレイテッド 神経機能におけるtaj
US7906116B2 (en) 2005-09-01 2011-03-15 Parkash Gill Methods for using and identifying modulators of Delta-like 4
NZ594285A (en) 2005-11-04 2013-02-22 Genentech Inc USE OF COMPLEMENT PATHWAY INHIBITOR ANTIBODY AGAINST C5a TO TREAT OCULAR DISEASES
CA2667997C (en) 2006-11-02 2016-10-11 Genentech, Inc. Humanized anti-factor d antibodies and uses thereof
AR066660A1 (es) 2007-05-23 2009-09-02 Genentech Inc Prevencion y tratamiento de condiciones del ojo asociadas con su complemento
WO2009070243A2 (en) * 2007-11-21 2009-06-04 Amgen Inc. Wise binding agents and epitopes
CR20170001A (es) 2008-04-28 2017-08-10 Genentech Inc Anticuerpos anti factor d humanizados
JP2011519911A (ja) 2008-05-05 2011-07-14 ノヴィミュンヌ エスア 抗il17a/il−17f交差反応抗体及びその使用方法
EP3141560A3 (de) 2008-05-06 2017-05-24 Genentech, Inc. Affinitätsgereifte crig-varianten
LT2398902T (lt) 2009-02-20 2023-12-27 Astellas Pharma Inc. Vėžio diagnostikos ir gydymo būdai bei kompozicijos
CN104974250A (zh) 2009-05-05 2015-10-14 诺维莫尼公司 抗il-17f抗体及其使用方法
JP5383905B2 (ja) 2009-05-08 2014-01-08 ジェネンテック, インコーポレイテッド ヒト化抗egfl7抗体とその使用方法
AT508569A1 (de) 2009-07-23 2011-02-15 Affiris Ag Pharmaceutical compound
PT3305813T (pt) 2009-11-11 2020-04-22 Astellas Pharma Inc Anticorpos específicos para claudina 6 (cldn6)
EP2404936A1 (de) 2010-07-06 2012-01-11 Ganymed Pharmaceuticals AG Krebstherapie mittels CLDN6-zielgerichteter Antikörper im lebenden Organismus
BR112013029212B1 (pt) 2011-05-13 2020-05-19 Ganymed Pharmaceuticals Ag anticorpos para o tratamento de câncer expressando claudina-6
EP3597666A3 (de) 2011-07-01 2020-04-22 NGM Biopharmaceuticals, Inc. Zusammensetzungen, verwendungen davon und verfahren zur behandlung von stoffwechselerkrankungen und -leiden
ES2828505T3 (es) 2012-11-28 2021-05-26 Ngm Biopharmaceuticals Inc Composiciones y métodos para el tratamiento de trastornos y enfermedades metabólicos
US9290557B2 (en) 2012-11-28 2016-03-22 Ngm Biopharmaceuticals, Inc. Compositions comprising variants and fusions of FGF19 polypeptides
KR20150104579A (ko) 2012-12-27 2015-09-15 엔지엠 바이오파마슈티컬스, 아이엔씨. 담즙산 항상성의 조절 및 담즙산 질환 및 질병의 치료 방법
US9273107B2 (en) 2012-12-27 2016-03-01 Ngm Biopharmaceuticals, Inc. Uses and methods for modulating bile acid homeostasis and treatment of bile acid disorders and diseases
CA2898128A1 (en) * 2013-01-17 2014-07-24 Medizinische Hochschule Hannover Factor 1 protein, factor 2 protein and inhibitors thereof for use in treating or preventing diseases
WO2015014376A1 (en) 2013-07-31 2015-02-05 Biontech Ag Diagnosis and therapy of cancer involving cancer stem cells
CA2920666A1 (en) 2013-08-12 2015-02-19 Genentech, Inc. Compositions and method for treating complement-associated conditions
US10369199B2 (en) 2013-10-28 2019-08-06 Ngm Biopharmaceuticals, Inc. Methods of using variants of FGF19 polypeptides for the treatment of cancer
SG11201606018UA (en) 2014-01-24 2016-08-30 Ngm Biopharmaceuticals Inc Binding proteins and methods of use thereof
SG11201608868PA (en) 2014-05-01 2016-11-29 Genentech Inc Anti-factor d antibody variants and uses thereof
WO2015183890A2 (en) 2014-05-28 2015-12-03 Ngm Biopharmaceuticals, Inc. Methods and compositions for the treatment of metabolic disorders and diseases
AU2015277438B2 (en) 2014-06-16 2020-02-27 Ngm Biopharmaceuticals, Inc. Methods and uses for modulating bile acid homeostasis and treatment of bile acid disorders and diseases
SG11201702757YA (en) 2014-10-23 2017-05-30 Ngm Biopharmaceuticals Inc Pharmaceutical compositions comprising peptide variants and methods of use thereof
US10434144B2 (en) 2014-11-07 2019-10-08 Ngm Biopharmaceuticals, Inc. Methods for treatment of bile acid-related disorders and prediction of clinical sensitivity to treatment of bile acid-related disorders
WO2017019957A2 (en) 2015-07-29 2017-02-02 Ngm Biopharmaceuticals, Inc. Binding proteins and methods of use thereof
CN108289951A (zh) 2015-10-30 2018-07-17 豪夫迈·罗氏有限公司 抗-因子d抗体和缀合物
EP3368090A1 (de) 2015-10-30 2018-09-05 H. Hoffnabb-La Roche Ag Anti-faktor-d-antikörpervarianten-konjugate und verwendungen davon
EP3888672A1 (de) 2015-11-09 2021-10-06 NGM Biopharmaceuticals, Inc. Verfahren zur behandlung von gallensäurebedingten erkrankungen
CA3034399A1 (en) 2016-08-26 2018-03-01 Ngm Biopharmaceuticals, Inc. Methods of treating fibroblast growth factor 19-mediated cancers and tumors
GB201906052D0 (en) * 2019-04-30 2019-06-12 Int Centre For Genetic Engineering And Biotechnology Proteins with cardioprotective activity
JP7064179B2 (ja) 2020-11-30 2022-05-10 富士電機株式会社 水分吸着材

Family Cites Families (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2000032221A2 (en) * 1998-12-01 2000-06-08 Genentech, Inc. Promotion or inhibition of angiogenesis and cardiovascularization
AU8921598A (en) * 1997-08-29 1999-03-16 Human Genome Sciences, Inc. 29 human secreted proteins
WO1999043802A2 (en) * 1998-02-27 1999-09-02 Sagami Chemical Research Center HUMAN PROTEINS HAVING TRANSMEMBRANE DOMAINS AND DNAs ENCODING THESE PROTEINS
EP1490386B1 (de) * 1998-03-10 2008-08-13 Genentech, Inc. Neuartiges polypeptid und dessen kodierende nukleinsäuren
EP1068307A1 (de) * 1998-03-31 2001-01-17 Genetics Institute, Inc. Sekretierte proteine und für diese kodierende polynukleotide
AU5920099A (en) * 1998-09-14 2000-04-03 Genentech Inc. Promotion or inhibition of angiogenesis and cardiovascularization
AU2883600A (en) * 1999-03-08 2000-09-28 Genentech Inc. Secreted and transmembrane polypeptides and nucleic acids encoding the same
AU2399300A (en) * 1999-03-08 2000-09-28 Genentech Inc. Methods and compositions for inhibiting neoplastic cell growth
KR100543857B1 (ko) * 1999-09-01 2006-01-23 제넨테크, 인크. 혈관신생 및 심혈관형성의 촉진 또는 억제
JP2004506413A (ja) * 2000-06-23 2004-03-04 ジェネンテック・インコーポレーテッド 血管形成に関与する疾患の診断と治療のための組成物と方法

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See references of WO0208284A3 *

Also Published As

Publication number Publication date
JP2009039119A (ja) 2009-02-26
JP2009039117A (ja) 2009-02-26
JP2009039118A (ja) 2009-02-26
CA2416538A1 (en) 2002-01-31
AU2001271973A1 (en) 2002-02-05
JP2004516013A (ja) 2004-06-03
WO2002008284A2 (en) 2002-01-31
JP6033531B2 (ja) 2016-11-30
WO2002008284A3 (en) 2003-03-13
JP2012005493A (ja) 2012-01-12

Similar Documents

Publication Publication Date Title
US7135334B2 (en) PRO20044 nucleic acids
AU771751B2 (en) Promotion or inhibition of angiogenesis and cardiovascularization
WO2002008284A2 (en) Compositions and methods for the diagnosis and treatment of disorders involving angiogenesis
EP1309620A2 (de) Zusammensetzungen und verfahren zur diagnose und der behandlung von krankheiten, bei denen die angiogenese eine rolle spielt
US7265210B2 (en) Anti-PRO9821 antibodies
US20040043927A1 (en) Compositions and methods for the diagnosis and treatment of disorders involving angiogenesis
CA2373915A1 (en) Methods and compositions for inhibiting neoplastic cell growth
AU768694B2 (en) Promotion or inhibition of angiogenesis and cardiovascularization
US20030224984A1 (en) Compositions and methods for the diagnosis and treatment of disorders involving angiogenesis
EP2042597B1 (de) Zusammensetzungen und Verfahren zur Diagnose und Behandlung von Erkrankungen in Verbindung mit Angiogenese
AU2003259607B2 (en) Promotion or inhibition of angiogenesis and cardiovascularization
CA2648051A1 (en) Compositions and methods for the diagnosis and treatment of disorders involving angiogenesis
ZA200103707B (en) Promotion or inhibition of angiogenesis and cardio-vascularization.
JP2015037408A (ja) 血管形成に関連する障害を診断及び治療するための組成物と方法
NZ545534A (en) Promotion or inhibition of angiogenesis and cardiovascularization

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20030128

AK Designated contracting states

Designated state(s): AT BE CH CY DE DK ES FI FR GB GR IE IT LI LU MC NL PT SE TR

AX Request for extension of the european patent

Extension state: AL LT LV MK RO SI

17Q First examination report despatched

Effective date: 20040615

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION IS DEEMED TO BE WITHDRAWN

18D Application deemed to be withdrawn

Effective date: 20041228