CA2390438A1 - 19 human secreted proteins - Google Patents

19 human secreted proteins Download PDF

Info

Publication number
CA2390438A1
CA2390438A1 CA002390438A CA2390438A CA2390438A1 CA 2390438 A1 CA2390438 A1 CA 2390438A1 CA 002390438 A CA002390438 A CA 002390438A CA 2390438 A CA2390438 A CA 2390438A CA 2390438 A1 CA2390438 A1 CA 2390438A1
Authority
CA
Canada
Prior art keywords
human
polypeptide
seq
sequence
polypeptides
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
CA002390438A
Other languages
French (fr)
Inventor
Steven M. Ruben
George A. Komatsoulis
Reinhard Ebner
Michele Fiscella
Ping Wei
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.)
Human Genome Sciences Inc
Original Assignee
Individual
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Individual filed Critical Individual
Publication of CA2390438A1 publication Critical patent/CA2390438A1/en
Abandoned 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
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • 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
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • 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
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/10Antimycotics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/18Antivirals for RNA viruses for HIV
    • 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
    • A61P37/00Drugs for immunological or allergic disorders
    • 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
    • A61P5/00Drugs for disorders of the endocrine system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • A61P5/14Drugs for disorders of the endocrine system of the thyroid hormones, e.g. T3, T4
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/02Antithrombotic agents; Anticoagulants; Platelet aggregation inhibitors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/04Antihaemorrhagics; Procoagulants; Haemostatic agents; Antifibrinolytic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/06Antianaemics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Diabetes (AREA)
  • Hematology (AREA)
  • Endocrinology (AREA)
  • Oncology (AREA)
  • Communicable Diseases (AREA)
  • Virology (AREA)
  • Pulmonology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Cardiology (AREA)
  • Immunology (AREA)
  • Rheumatology (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Molecular Biology (AREA)
  • Urology & Nephrology (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Biomedical Technology (AREA)
  • Toxicology (AREA)
  • Zoology (AREA)
  • Neurosurgery (AREA)
  • Pain & Pain Management (AREA)
  • AIDS & HIV (AREA)
  • Reproductive Health (AREA)
  • Dermatology (AREA)
  • Obesity (AREA)
  • Emergency Medicine (AREA)

Abstract

The present invention relates to novel human secreted proteins and isolated nucleic acids containing the coding regions of the genes encoding such proteins. Also provided are vectors, host cells, antibodies, and recombinant methods for producing human secreted proteins. The invention further relates to diagnostic and therapeutic methods useful for diagnosing and treating diseases, disorders, and/or conditions related to these novel human secreted proteins.

Description

DEMANDE OU BREVET VOLUMINEUX
LA PRESENTE PARTIE DE CETTE DEMANDE OU CE BREVET COMPREND
PLUS D'UN TOME.

NOTE : Pour les tomes additionels, veuillez contacter 1e Bureau canadien des brevets JUMBO APPLICATIONS/PATENTS
THIS SECTION OF THE APPLICATION/PATENT CONTAINS MORE THAN ONE
VOLUME

NOTE: For additional volumes, please contact the Canadian Patent Office NOM DU FICHIER / FILE NAME
NOTE POUR LE TOME / VOLUME NOTE:

19 Human Secreted Proteins Field of the Invention This invention relates to newly identified polynucleotides, polypeptides encoded by these polynucleotides, antibodies that bind these polypeptides, uses of such polynucleotides, polypeptides, and antibodies, and their production.
Background of the Invention Unlike bacterium, which exist as a single compartment surrounded by a membrane, human cells and other eucaryotes are subdivided by membranes into many functionally distinct compartments. Each membrane-bounded compartment, or organelle, contains different proteins essential for the function of the organelle. The cell uses "sorting signals," which are amino acid motifs located within the protein, to target proteins to particular cellular organelles.
One type of sorting signal, called a signal sequence, a signal peptide, or a leader sequence, directs a class of proteins to an organelle called the endoplasmic 1 S reticulum (ER). The ER separates the membrane-bounded proteins from all other types of proteins. Once localized to the ER, both groups of proteins can be further directed to another organelle called the Golgi apparatus. Here, the Golgi distributes the proteins to vesicles, including secretory vesicles, the cell membrane, lysosomes, and the other organelles.
Proteins targeted to the ER by a signal sequence can be released into the extracellular space as a secreted protein. For example, vesicles containing secreted proteins can fuse with the cell membrane and release their contents into the extracellular space - a process called exocytosis. Exocytosis can occur constitutively or after receipt of a triggering signal. In the latter case, the proteins are stored in secretory vesicles (or secretory granules) until exocytosis is triggered.
Similarly, proteins residing on the cell membrane can also be secreted into the extracellular space by proteolytic cleavage of a "linker" holding the protein to the membrane.
Despite the great progress made in recent years, only a small number of genes encoding human secreted proteins have been identified. These secreted proteins include the commercially valuable human insulin, interferon, Factor VIII, human growth hormone, tissue plasminogen activator, and erythropoeitin. Thus, in light of the pervasive role of secreted proteins in human physiology, a need exists for identifying and characterizing novel human secreted proteins and the genes that encode them. This knowledge will allow one to detect, to treat, and to prevent medical diseases, disorders, and/or conditions by using secreted proteins or the genes that encode them.
Summary of the Invention The present invention relates to novel polynucleotides and the encoded polypeptides. Moreover, the present invention relates to vectors, host cells, antibodies, and recombinant and synthetic methods for producing the polypeptides and polynucleotides. Also provided are diagnostic methods for detecting diseases, disorders, and/or conditions related to the polypeptides and polynucleotides, and therapeutic methods for treating such diseases, disorders, and/or conditions.
The invention further relates to screening methods for identifying binding partners of the polypeptides.
Detailed Description Definitions The following definitions are provided to facilitate understanding of certain terms used throughout this specification.
In the present invention, "isolated" refers to material removed from its original environment (e.g., the natural environment if it is naturally occurring), and thus is altered "by the hand of man" from its natural state. For example, an isolated polynucleotide could be part of a vector or a composition of matter, or could be contained within a cell, and still be "isolated" because that vector, composition of matter, or particular cell is not the original environment of the polynucleotide. The term "isolated" does not refer to genomic or cDNA libraries, whole cell total or mRNA preparations, genomic DNA preparations (including those separated by electrophoresis and transferred onto blots), sheared whole cell genomic DNA
preparations or other compositions where the art demonstrates no distinguishing features of the polynucleotide/sequences of the present invention.

In the present invention, a "secreted" protein refers to those proteins capable of being directed to the ER, secretory vesicles, or the extracellular space as a result of a signal sequence, as well as those proteins released into the extracellular space without necessarily containing a signal sequence. If the secreted protein is released into the extracellular space, the secreted protein can undergo extracellular processing to produce a "mature" protein. Release into the extracellular space can occur by many mechanisms, including exocytosis and proteolytic cleavage.
In specific embodiments, the polynucleotides of the invention are at least 15, at least 30, at least 50, at least 100, at least 125, at least 500, or at least 1000 continuous nucleotides but are less than or equal to 300 kb, 200 kb, 100 kb, 50 kb, 15 kb, 10 kb, 7.5 kb, 5 kb, 2.5 kb, 2.0 kb, or 1 kb, in length. In a further embodiment, polynucleotides of the invention comprise a portion of the coding sequences, as disclosed herein, but do not comprise all or a portion of any intron. In another embodiment, the polynucleotides comprising coding sequences do not contain coding sequences of a genomic flanking gene (i.e., 5' or 3' to the gene of interest in the genome). In other embodiments, the polynucleotides of the invention do not contain the coding sequence of more than 1000, 500, 250, 100, 50, 25, 20, 15, 10, 5, 4, 3, 2, or 1 genomic flanking gene(s).
As used herein, a "polynucleotide" refers to a molecule having a nucleic acid sequence contained in SEQ ID NO:X or the cDNA contained within the clone deposited with the ATCC. For example, the polynucleotide can contain the nucleotide sequence of the full length cDNA sequence, including the 5' and 3' untranslated sequences, the coding region, with or without the signal sequence, the secreted protein coding region, as well as fragments, epitopes, domains, and variants of the nucleic acid sequence. Moreover, as used herein, a "polypeptide" refers to a molecule having the translated amino acid sequence generated from the polynucleotide as broadly defined.
In the present invention, the full length sequence identified as SEQ ID NO:X
was often generated by overlapping sequences contained in multiple clones (contig analysis). A representative clone containing all or most of the sequence for SEQ ID .
NO:X was deposited with the American Type Culture Collection ("ATCC"). As shown in Table l, each clone is identified by a cDNA Clone ID (Identifier) and the ATCC Deposit Number. The ATCC is located at 10801 University Boulevard, Manassas, Virginia 20110-2209, USA. The ATCC deposit was made pursuant to the terms of the Budapest Treaty on the international recognition of the deposit of microorganisms for purposes of patent procedure.
A "polynucleotide" of the present invention also includes those polynucleotides capable of hybridizing, under stringent hybridization conditions, to sequences contained in SEQ 117 NO:X, the complement thereof, or the cDNA
within the clone deposited with the ATCC. "Stringent hybridization conditions" refers to an overnight incubation at 42 degree C in a solution comprising SO% formamide, Sx SSC
(750 mM NaCI, 75 mM trisodium citrate), 50 mM sodium phosphate (pH 7.6), Sx Denhardt's solution, 10% dextran sulfate, and 20 ~g/ml denatured, sheared salmon sperm DNA, followed by washing the filters in O.lx SSC at about 65 degree C.
Also contemplated are nucleic acid molecules that hybridize to the polynucleotides of the present invention at lower stringency hybridization conditions.
Changes in the stringency of hybridization and signal detection are primarily accomplished through the manipulation of formamide concentration (lower percentages of formamide result in lowered stringency); salt conditions, or temperature. For example, lower stringency conditions include an overnight incubation at 37 degree C in a solution comprising 6X SSPE (20X SSPE = 3M
NaCI;
0.2M NaH2P04; 0.02M EDTA, pH 7.4), 0.5% SDS, 30% formamide, 100 ug/ml salmon sperm blocking DNA; followed by washes at 50 degree C with 1XSSPE, 0.1% SDS. In addition, to achieve even lower stringency, washes performed following stringent hybridization can be done at higher salt concentrations (e.g. 5X
SSC).
Note that variations in the above conditions may be accomplished through the inclusion and/or substitution of alternate blocking reagents used to suppress background in hybridization experiments. Typical blocking reagents include Denhardt's reagent, BLOTTO, heparin, denatured salmon sperm DNA, and commercially available proprietary formulations. The inclusion of specific blocking reagents may require modification of the hybridization conditions described above, due to problems with compatibility.

Of course, a polynucleotide which hybridizes only to polyA+ sequences (such as any 3' terminal polyA+ tract of a cDNA shown in the sequence listing), or to a complementary stretch of T (or U) residues, would not be included in the definition of "polynucleotide," since such a polynucleotide would hybridize to any nucleic acid molecule containing a poly (A) stretch or the complement thereof (e.g., practically any double-stranded cDNA clone generated using oligo dT as a primer).
The polynucleotide of the present invention can be composed of any polyribonucleotide or polydeoxribonucleotide, which may be unmodified RNA or DNA or modified RNA or DNA. For example, polynucleotides can be composed of single- and double-stranded DNA, DNA that is a mixture of single- and double-stranded regions, single- and double-stranded RNA, and RNA that is mixture of single- and double-stranded regions, hybrid molecules comprising DNA and RNA
that may be single-stranded or, more typically, double-stranded or a mixture of single-and double-stranded regions. In addition, the polynucleotide can be composed of triple-stranded regions comprising RNA or DNA or both RNA and DNA. A
polynucleotide may also contain one or more modified bases or DNA or RNA
backbones modified for stability or for other reasons. "Modified" bases include, for example, tritylated bases and unusual bases such as inosine. A variety of modifications can be made to DNA and RNA; thus, "polynucleotide" embraces chemically, enzymatically, or metabolically modified forms.
The polypeptide of the present invention can be composed of amino acids joined to each other by peptide bonds or modified peptide bonds, i.e., peptide isosteres, and may contain amino acids other than the 20 gene-encoded amino acids.
The polypeptides may be modified by either natural processes, such as posttranslational processing, or by chemical modification techniques which are well known in the art. Such modifications are well described in basic texts and in more detailed monographs, as well as in a voluminous research literature.
Modifications can occur anywhere in a polypeptide, including the peptide backbone, the amino acid side-chains and the amino or carboxyl termini. It will be appreciated that the same type of modification may be present in the same or varying degrees at several sites in a given polypeptide. Also, a given polypeptide may contain many types of modifications. Polypeptides may be branched , for example, as a result of ubiquitination, and they may be cyclic, with or without branching. Cyclic, branched, and branched cyclic polypeptides may result from posttranslation natural processes or may be made by synthetic methods. Modifications include acetylation, acylation, ADP-ribosylation, amidation, covalent attachment of flavin, covalent attachment of a heme moiety, covalent attachment of a nucleotide or nucleotide derivative, covalent attachment of a lipid or lipid derivative, covalent attachment of phosphotidylinositol, cross-linking, cyclization, disulfide bond formation, demethylation, formation of covalent cross-links, formation of cysteine, formation of pyroglutamate, formylation, gamma-carboxylation, glycosylation, GPI anchor formation, hydroxylation, iodination, methylation, myristoylation, oxidation, pegylation, proteolytic processing, phosphorylation, prenylation, racemization, selenoylation, sulfation, transfer-RNA
mediated addition of amino acids to proteins such as arginylation, and ubiquitination.
(See, for instance, PROTEINS - STRUCTURE AND MOLECULAR PROPERTIES, 2nd Ed., T. E. Creighton, W. H. Freeman and Company, New York (1993);
POSTTRANSLATIONAL COVALENT MODIFICATION OF PROTEINS, B. C.
Johnson, Ed., Academic Press, New York, pgs. 1-12 (1983); Seifter et al., Meth Enzymol 182:626-646 (1990); Rattan et al., Ann NY Acad Sci 663:48-62 (1992).) "SEQ ID NO:X" refers to a polynucleotide sequence while "SEQ ID NO:Y"
refers to a polypeptide sequence, both sequences identified by an integer specified in Table 1.
"A polypeptide having biological activity" refers to polypeptides exhibiting activity similar, but not necessarily identical to, an activity of a polypeptide of the present invention, including mature forms, as measured in a particular biological assay, with or without dose dependency. In the case where dose dependency does exist, it need not be identical to that of the polypeptide, but rather substantially similar to the dose-dependence in a given activity as compared to the polypeptide of the present invention (i.e., the candidate polypeptide will exhibit greater activity or not more than about 25-fold less and, preferably, not more than about tenfold less activity, and most preferably, not more than about three-fold less activity relative to the polypeptide of the present invention.) Many proteins (and translated DNA sequences) contain regions where the amino acid composition is highly biased toward a small subset of the available residues. For example, membrane spanning domains and signal peptides (which are also membrane spanning) typically contain long stretches where Leucine (L), Valine (V), Alanine (A), and Isoleucine (I) predominate. Poly-Adenosine tracts (polyA) at the end of cDNAs appear in forward translations as poly-Lysine (poly-K) and poly-Phenylalanine (poly-F) when the reverse complement is translated. These regions are often referred to as "low complexity" regions.
Such regions can cause database similarity search programs such as BLAST to find high-scoring sequence matches that do not imply true homology. The problem is exacerbated by the fact that most weight matrices (used to score the alignments generated by BLAST) give a match between any of a group of hydrophobic amino acids (L,V and I) that are commonly found in certain low complexity regions almost as high a score as for exact matches.
In order to compensate for this, BLASTX.2 (version 2.Oa5MP-WashU) employs two filters ("seg" and "xnu") which "mask" the low complexity regions in a particular sequence. These filters parse the sequence for such regions, and create a new sequence in which the amino acids in the low complexity region have been replaced with the character "X". This is then used as the input sequence (sometimes referred to herein as "Query" and/or "Q") to the BLASTX program. While this regime helps to ensure that high-scoring matches represent true homology, there is a negative consequence in that the BLASTX program uses the query sequence that has been masked by the filters to draw alignments.
Thus, a stretch of "X"s in an alignment shown in the following application does not necessarily indicate that either the underlying DNA sequence or the translated protein sequence is unknown or uncertain. Nor is the presence of such stretches meant to indicate that the sequence is identical or not identical to the sequence disclosed in the alignment of the present invention. Such stretches may simply indicate that the BLASTX program masked amino acids in that region due to the detection of a low complexity region, as defined above. In all cases, the reference sequences) (sometimes referred to herein as "Subject", "Sbjct", and/or "S") indicated in the specification, sequence table (Table 1), and/or the deposited clone is (are) the definitive embodiments) of the present invention, and should not be construed as limiting the present invention to the partial sequence shown in an alignment, unless specifically noted otherwise herein.
Polynucleotides and Polypeptides of the Invention FEATURES OF PROTEIN ENCODED BY GENE NO: 1 The computer algorithm BLASTX has been used to determine that the translation product of this gene shares sequence homology with, as a non-limiting example, the sequence accessible through the following database accession no.
gb~AAD55357.1 ~AF125175_1 (all information available through the recited accession number is incorporated herein by reference) which is described therein as "(AF125175) angiopoietin-related protein-2 [Homo Sapiens]" A partial alignment demonstrating the observed homology is shown immediately below.
>gb~AAD55357.1~AF125175 1 (AF125175) angiopoietin-related protein-2 [Homo Sapiens] >sp~AAD55357~AAD55357 Angiopoietin-related protein-2.
Length = 493 2O Plus Strand HSPS:
Score = 315 (110.9 bits), Expect = 1.4e-26, P = 1.4e-26 Identities = 58/93 (62~), Positives = 69/93 (74~), Frame = +3 2S Q: 405 GRGQGMPGKSLAL*GAGRGFYSGERTWTLSLPSGNCALYQRGGWWYHACAHSNLNGVWHH 584 GR G G S G+ F + +R + +GNCA YQ+GGWWY+ACAHSNLNGVW+
S: 398 GRYHGNAGDSFTWHN-GKQFTTLDRDHDVY--TGNCAHYQKGGWWYNACAHSNLNGVWYR 454 Q: 585 GGHYRSRYQDGVYWAEFRGGAYSLRKAAMLIRP 683 3 O GGHYRSRYQDGVYWAEFRGG+YSL+K M+IRP
S: 455 GGHYRSRYQDGVYWAEFRGGSYSLKKWMMIRP 487 The segment of gb~AAD55357.1 ~AF125175_1 that is shown as "S" above is set out in the sequence listing as SEQ ID NO: 121. Based on the structural similarity these 35 homologous polypeptides are expected to share at least some biological activities.
Such activities are known in the art, some of which are described elsewhere herein.
Assays for determining such activities are also known in the art, some of which have been described elsewhere herein. Preferred polypeptides of the invention comprise a polypeptide having the amino acid sequence set out in the sequence listing as SEQ >I7 NO: 122 which corresponds to the Q sequence in the alignment shown above (gaps introduced in a sequence by the computer are, of course, removed).
In specific embodiments, polypeptides of the invention comprise, or alternatively consists of, an amino acid sequence selected from the group:
MSQEGVELEKSVRRLREKFHGKVSSKKAGALMRKFGSDHTGVGRSIVYGVK
QKDGQELSNDLDAQDPPEDMKQDRDIQAVATSLLPLTEANLRMFQRAQDDL
IPAVDRQFACSSCDHVWWRRVPQRKEVSRCRKCRKRYEPVPADKMWGLAE
FHCPKCRHNFRGWAQMGSPSPCYGCGFPVYPTRILPPRWDRDPDRRSTHTHS
CSAADCYNRREPHVPGTSCAHPKSRKQNHLPKVLHPSNPHISSGSTVATCLSQ
GGLLEDLDNLILEDLKEEEEEEEEVEDEEGGPRE (SEQ ID NO: 123) and LRGCWTDGRTQVGGRLSRAVRTLAGNCAAATMSQEGVELEKSVRRLREKFH
GKVSSKKAGALMRKFGSDHTGVGRSIVYGVKQKDGQELSNDLDAQDPPED
MKQDRDIQAVATSLLPLTEANLRMFQRAQDDLIPAVDRQFACS SCDHV W WR
RVPQRKEVSRCRKCRKRYEPVPADKMWGLAEFHCPKCRHNFRGWAQMGSP
SPCYGCGFPVYPTRILPPRWDRDPDRRSTHTHSCSAADCYNRREPHVPGTSCA
HPKSRKQNHLPKVLHPSNPHISSGSTVATCLSQGGLLEDLDNLILEDLKEEEEE
EEEVEDEEGGPRE (SEQ ID NO: 124). Moreover, fragments and variants of these polypeptides (such as, for example, fragments as described herein, polypeptides at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% identical to these polypeptides and polypeptides encoded by the polynucleotide which hybridizes, under stringent conditions, to the polynucleotide encoding these polypeptides ) are encompassed by the invention. Antibodies that bind polypeptides of the invention are also encompassed by the invention. Polynucleotides encoding these polypeptides are also encompassed by the invention.
This gene is expressed primarily in the following tissues/cDNA libraries:
NCI CGAP GCB1 and to a lesser extent in Soares ovary tumor NbHOT;
Soares fetal liver spleen_1NFLS S1; NCI CGAP_Brl.l; Human endometrial stromal cells-treated with progesterone; NCI CGAP-Lyml2; T-Cell PHA 24 hrs;
Ovary, Cancer: (4004576 A8); Hemangiopericytoma; Stratagene liver (#937224);
NCI CGAP_GC6; Endothelial cells-control; Soares senescent fibroblasts NbHSF;
H. Frontal cortex,epileptic,re-excision; Soares~regnant uterus NbHPU; Soares fetal liver spleen 1NFLS; H. Leukocytes, normalized cot > SOOA; S, Human foetal Adrenals tissue; Barstead prostate BPH HPLRB4 l; Kidney medulla; Thyroid Thyroiditis; Human adult lung 3' directed MboI cDNA; H. Striatum Depression, subtracted; Schiller oligodendroglioma; NCI CGAP HN4; Human promyelocyte;
Lung, Cancer (4005313 A3): Invasive Poorly Differentiated Lung Adenocarcinoma,;
5 Human Liver; NCI CGAP_Col2; NCI CGAP Ut3; Human Colon Cancer,re-excision; Hepatocellular Tumor; Human Prostate Cancer, Stage C fraction; pBMC
stimulated w/ poly I/C; Amniotic Cells - Primary Culture; H. Kidney Cortex, subtracted; Human Adipose Tissue, re-excision; H. Meningima, M1; Human Adult Small Intestine; NCI CGAP Utl; Apoptotic T-cell; Human Fetal Dura Mater;
10 Human Pancreas Tumor; NCI CGAP-Pr28; Human Placenta (re-excision); Spinal cord; NCI CGAP-Panl; Brain frontal cortex; Normal colon; NCI CGAP GC4;
Human Placenta; Human Testes, Reexcision; Smooth muscle,control; Human Bone Marrow, treated; Human Testes; Human Endometrial Tumor; Activated T-cell(12h)/Thiouridine-re-excision; Soares NFL T GBC Sl and Primary Dendritic Cells, lib 1.
Polynucleotides and polypeptides of the invention are useful as reagents for differential identification of the tissues) or cell types) present in a biological sample and for diagnosis of diseases and conditions which include but are not limited to:
neoplasms and hemapoietic, developmental, immune and inflammatory, cardiovascular and reproductive disorders. Similarly, polypeptides and antibodies directed to these polypeptides are useful in providing immunological probes for differential identification of the tissues) or cell type(s). For a number of disorders of the above tissues or cells, particularly of the immune system, expression of this gene at significantly higher or lower levels may be routinely detected in certain tissues or cell types (e.g., immune, cancerous and wounded tissues) or bodily fluids (e.g., serum, plasma, urine, synovial fluid and spinal fluid) or another tissue or sample taken from an individual having such a disorder, relative to the standard gene expression level, i.e., the expression level in healthy tissue or bodily fluid from an individual not having the disorder.
Enriched expression in fetal liver and spleen and ovarian tumor and homology to angiopoietin indicates that polynucleotides and polypeptides corresponding to this gene are useful for study and treatment of hemapoietic, developmental, immune and inflammatory, cardiovascular and reproductive disorders, and neoplasms.
The tissue distribution in immune cells (e.g., T-cells) indicates the polynucleotides and polypeptides corresponding to this gene would be useful for the diagnosis and treatment of a variety of immune system disorders.
Representative uses are described in the "Immune Activity" and "Infectious Disease" sections below, in Example 11, 13, 14, 16, 18, 19, 20, and 27, and elsewhere herein. Briefly, the expression indicates a role in regulating the proliferation; survival;
differentiation;
and/or activation of hematopoietic cell lineages, including blood stem cells.
Involvement in the regulation of cytokine production, antigen presentation, or other processes indicates a usefulness for treatment of cancer (e.g. by boosting immune responses). Expression in cells of lymphoid origin, indicates the natural gene product would be involved in immune functions. Therefore it would also be useful as an agent for immunological disorders including arthritis, asthma, immunodeficiency diseases such as AIDS, leukemia, rheumatoid arthritis, granulomatous disease, inflammatory bowel disease, sepsis, acne, neutropenia, neutrophilia, psoriasis, hypersensitivities, such as T-cell mediated cytotoxicity; immune reactions to transplanted organs and tissues, such as host-versus-graft and graft-versus-host diseases, or autoimmunity disorders, such as autoimmune infertility, lense tissue injury, demyelination, systemic lupus erythematosis, drug induced hemolytic anemia, rheumatoid arthritis, Sjogren's disease, and scleroderma. Moreover, the protein may represent a secreted factor that influences the differentiation or behavior of other blood cells, or that recruits hematopoietic cells to sites of injury. Thus, this gene product is thought to be useful in the expansion of stem cells and committed progenitors of various blood lineages, and in the differentiation and/or proliferation of various cell types.
Furthermore, the protein may also be used to determine biological activity, raise antibodies, as tissue markers, to isolate cognate ligands or receptors, to identify agents that modulate their interactions, in addition to its use as a nutritional supplement. Protein, as well as, antibodies directed against the protein may show utility as a tumor marker and/or immunotherapy targets for the above listed tissues.
FEATURES OF PROTEIN ENCODED BY GENE NO: 2 The translation product of this gene shares sequence homology with rat protocadherin, a brain-specific transmembrane receptor which is thought to be important in neuronal growth and development.
In specific embodiments, polypeptides of the invention comprise, or alternatively consists of, an amino acid sequence selected from the group:
HASAPDPLRPSCQVTAAKMGS WALLWPPLLFTGLLVRPPGTMAQAQ
YCSVNKDIFEVEENTNVTEPLVDIHVPEGQEVTLGALSTPFAFRIQGNQLFLNV
TPDYEEKSLLEAQLLCQSGGTLVTQLRVFVSVLDVNDNAPEFPFKTKEIRVEE
DTKVNSTVIPETQLQAEDRDKDDILFYTLQEMTAGASDYFSLVSVNRPALRL
DRPLDFYERPNMTFWLLVRDTPGENVEPSHTATATLVLNV VPADLRPPWFLP
CTFSDGYVCIQAQYHGAVPTGHILPSPLVLRPGPIYAEDGDRGINQPIIYSIFRG
NVNGTFIIHPDSGNLTVARSVPSPMTFLLLVKGQQADLARYSVTQVTVEAVA
AAGSPPRFPQSLYRGTVARGAGAGVVVKDAAAPSQPLRIQAQDPEFSDLNSA
ITYRITNHSHFRMEGEVVLTTTTLAQAGAFYAEVAAPRRTSASRWWIWRPW
AGCWVRCCCWLSLASPSLSTSTMAPGSSAALAKLRSPSPKALTTRRSSLTTRP
TGRPSPAPRTTPSPRRHRCPQSPHPPALPPQAVPLSPPQR PELAEAPRR (SEQ
m NO: 125) and MAALGGVLGALLLLALLGLAVLVHKHYGPRLKCCSGKAPEPQPQGFDNQAF
LPDHKANWAPVPSPTHDPKPAEAPMPAEPAPPGPASPGGAPEPPAAARAGGS
PTAVRSILTKERRPEGGYKAV WFGEDIGTEADV V VLNAPTLDVDGASDSGSG
DEGEGAGRGGGPYDAPGGDDSYI (SEQ ID NO: 155). Moreover, fragments and variants of these polypeptides (such as, for example, fragments as described herein, polypeptides at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% identical to these polypeptides and polypeptides encoded by the polynucleotide which hybridizes, under stringent conditions, to the polynucleotide encoding these polypeptides ) are encompassed by the invention. Polynucleotides encoding these polypeptides are also encompassed by the invention.
This gene is expressed primarily in the following tissues/cDNA libraries:
NCI CGAP_Kid3; Soares multiple sclerosis 2NbHMSP;
Soares-parathyroid tumor NbHPA and to a lesser extent in Stratagene neuroepithelium NT2RAMI 937234; Human Whole Brain #2 - Oligo dT > l.SKb;

Human Manic Depression Tissue; Temporal cortex-Alzheizmer, subtracted; Colon Normal III; Soaves fetal liver spleen 1NFLS; Human Fetal Brain, normalized CSOOH;
Brain Amygdala Depression; STRIATUM DEPRESSION; H. Striatum Depression, subt; Human adult small intestine,re-excision; human corpus colosum; H. Kidney Cortex, subtracted; Human Stomach,re-excision; Human Colon, re-excision; Brain Frontal Cortex, re-excision; Human Adult Small Intestine; Gessler Wilms tumor;
Merkel Cells; Liver, Hepatoma; Human Hippocampus; Spinal cord; Ulcerative Colitis; Stratagene hNT neuron (#937233); Human Liver, normal; 12 Week Old Early Stage Human; Human Substantia Nigra; Colon Normal II; NCI CGAP_CoB;
NCI CGAP_GC4; H. Frontal cortex,epileptic,re-excision; Human 8 Week Whole Embryo; Nine Week Old Early Stage Human and Soaves total fetus Nb2HF8 9w.
Polynucleotides and polypeptides of the invention are useful as reagents for differential identification of the tissues) or cell types) present in a biological sample and for diagnosis of diseases and conditions which include but are not limited to:
1 S neurological abnormalities such as Alzheimer's disease, Parkinson's disease, CNS
inflammation, cerebellar degeneration, and hearing injury. Similarly, polypeptides and antibodies directed to these polypeptides are useful in providing immunological probes for differential identification of the tissues) or cell type(s). For a number of disorders of the above tissues or cells, particularly of the nervous system, expression of this gene at significantly higher or lower levels may be routinely detected in certain tissues or cell types (e.g., neural, cancerous and wounded tissues) or bodily fluids (e.g., serum, plasma, urine, synovial fluid and spinal fluid) or another tissue or sample taken from an individual having such a disorder, relative to the standard gene expression level, i.e., the expression level in healthy tissue or bodily fluid from an individual not having the disorder.
Moreover, the tissue distribution in brain and homology to procadharin family of proteins indicates that polynucleotides and polypeptides corresponding to this gene are useful for the diagnosis and treatment of neurological abnormalities such as Alzheimer's disease, Parkinson's disease, CNS inflammation, cerebellar degeneration, and hearing injury.
The tissue distribution in brain and homology to procadharin family of proteins indicates polynucleotides and polypeptides corresponding to this gene would be useful for the detection, treatment, and/or prevention of neurodegenerative disease states, behavioral disorders, or inflammatory conditions. Representative uses are described in the "Regeneration" and "Hyperproliferative Disorders" sections below, in Example 11, 15, and 18, and elsewhere herein. Briefly, the uses include, but are not limited to the detection, treatment, and/or prevention of Alzheimer's Disease, Parkinson's Disease, multiple sclerosis, Huntington's Disease, Tourette Syndrome, meningitis, encephalitis, demyelinating diseases, peripheral neuropathies, neoplasia, trauma, congenital malformations, spinal cord injuries, ischemia and infarction, aneurysms, hemorrhages, schizophrenia, mania, dementia, paranoia, obsessive compulsive disorder, depression, panic disorder, learning disabilities, ALS, psychoses, autism, and altered behaviors, including disorders in feeding, sleep patterns, balance, and perception. In addition, elevated expression of this gene product in regions of the brain indicates it plays a role in normal neural function.
Potentially, this gene product is involved in synapse formation, neurotransmission, learning, cognition, homeostasis, or neuronal differentiation or survival.
Furthermore, the protein may also be used to determine biological activity, to raise antibodies, as tissue markers, to isolate cognate ligands or receptors, to identify agents that modulate their interactions, in addition to its use as a nutritional supplement.
Protein, as well as, antibodies directed against the protein may show utility as a tumor marker and/or immunotherapy targets for the above listed tissues.
FEATURES OF PROTEIN ENCODED BY GENE NO: 3 This gene is expressed primarily in the following tissues/cDNA libraries:
Pancreas Islet Cell Tumor and to a lesser extent in Human Endometrial Tumor;
Hemangiopericytoma; Soares fetal heart NbHHI9W; Human Synovial Sarcoma;
Nine Week Old Early Stage Human; Soares NhHMPu S1; Soares infant brain 1NIB;
NCI CGAP-Brn23; Human Hypothalamus,schizophrenia, re-excision; Human Fetal Kidney, Reexcision; Human Cerebellum; Soares-pregnant uterus NbHPU; Healing groin wound, 7.5 hours post incision; Stratagene NT2 neuronal precursor 937230;
Human Thymus; Soares breast 3NbHBst; NCI CGAP-CoB; Human Fetal Lung III;
Human 8 Week Whole Embryo; Soares placenta Nb2HP; NCI CGAP-GCB1; Soares fetal liver spleen 1NFLS; Human Spleen; Human White Fat; Human 8 Week Whole Embryo, subtracted; NCI CGAP-Pr23; Human placenta cDNA (TFujiwara); Heart;
SKIN; Frontal lobe,dementia,re-excision; Fetal Heart, re-excision; H.
Epididiymus, caput & corpus; Human Lung Cancer,re-excision; Healing groin wound - zero hr 5 post-incision (control); Human endometrial stromal cells-treated with estradiol;
Human adult (K.Okubo); Human Adipose Tissue, re-excision; Healing groin wound, 6.5 hours post incision; Breast, Normal: (4005522B2); NCI CGAP-Ut2; Stratagene lung carcinoma 937218; NCI CGAP-Pr3; Human Prostate; Gessler Wilms tumor;
Human Brain, Striatum; NCI CGAP Utl; NCI CGAP Pr28; Human Pancreas 10 Tumor, Reexcision; Human Placenta (re-excision); Spinal cord;
Soares NSF F8 9W OT PA P S1; Human Whole Six Week Old Embryo;
NCI CGAP_Co3; Human T-Cell Lymphoma; Colon Carcinoma; Human Testes Tumor; Human Ovarian Cancer Reexcision; Human Placenta; 12 Week Early Stage Human II, Reexcision; NCI CGAP-Brn25; Soares senescent fibroblasts NbHSF;
1 S Soares multiple sclerosis 2NbHMSP; H. Frontal cortex,epileptic,re-excision;
normalized infant brain cDNA; Soares fetal liver spleen-1NFLS S1;
Soares NFL T GBC_S 1; Soares testis NHT and Primary Dendritic Cells, lib 1.
Polynucleotides and polypeptides of the invention are useful as reagents for differential identification of the tissues) or cell types) present in a biological sample and for diagnosis of diseases and conditions which include but are not limited to:
pancreatic and endometrial tumors and other neoplastic growth. Similarly, polypeptides and antibodies directed to these polypeptides are useful in providing immunological probes for differential identification of the tissues) or cell type(s). For a number of disorders of the above tissues or cells, particularly of the pancreas and endometrium, expression of this gene at significantly higher or lower levels may be routinely detected in certain tissues or cell types (e.g., cancerous and wounded tissues) or bodily fluids (e.g., serum, plasma, urine, synovial fluid and spinal fluid) or another tissue or sample taken from an individual having such a disorder, relative to the standard gene expression level, i.e., the expression level in healthy tissue or bodily fluid from an individual not having the disorder.
The tissue distribution in pancreas islet cell tumor and human endometrial tumor indicates that polynucleotides and polypeptides corresponding to this gene are useful for the diagnosis and treatment of pancreatic and endometrial tumors and other neoplastic growth.
FEATURES OF PROTEIN ENCODED BY GENE NO: 4 In specific embodiments, polypeptides of the invention comprise, or alternatively consists of, the following amino acid sequence:
MGGGKKVDWHQSSDPIPSPSFERFSSMLFAHPFFRVSQEWICRKEQGGK
GSGQRWKFSGRHKSEHTCAHACTHTQTHACTHTNMHTHACTHMHMHTHT
GAHTTKLQGHGVADPLRSDLRLEAAFHRSVSFPGPVMPLILCTQCRAAGFAF
LQRHYLLMSPTTQVPFMGSLCPSEFHRPQMAISSTSHPTTHRMLVLTQPFVVA
EPWAGRISPHAVPRMCPIPSAVITFVST (SEQ ID NO: 126). Moreover, fragments and variants of these polypeptides (such as, for example, fragments as described herein, polypeptides at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99%
identical to these polypeptides and polypeptides encoded by the polynucleotide which hybridizes, under stringent conditions, to the polynucleotide encoding these polypeptides ) are encompassed by the invention. Antibodies that bind polypeptides of the invention are also encompassed by the invention. Polynucleotides encoding these polypeptides are also encompassed by the invention.
This gene is expressed primarily in the following tissues/cDNA libraries:
Neutrophils control, re-excision; Neutrophils IL-1 and LPS induced;
Neutrophils IL-1 and LPS induced; Nine Week Old Early Stage Human.
Polynucleotides and polypeptides of the invention are useful as reagents for differential identification of the tissues) or cell types) present in a biological sample and for diagnosis of diseases and conditions which include but are not limited to:
immune system disorders such as allergy, asthma, inflammation, autoimmune-diseases.. Similarly, polypeptides and antibodies directed to these polypeptides are useful in providing immunological probes for differential identification of the tissues) or cell type(s). For a number of disorders of the above tissues or cells, particularly of the immune system, expression of this gene at significantly.higher or lower levels may be routinely detected in certain tissues or cell types (e.g., cancerous and wounded tissues) or bodily fluids (e.g., serum, plasma, urine, synovial fluid and spinal fluid) or another tissue or sample taken from an individual hawing such a disorder, relative to the standard gene expression level, i.e., the expression level in healthy tissue or bodily fluid from an individual not having the disorder.
The tissue distribution highly specific in neutrophils indicates that polynucleotides and polypeptides corresponding to this gene are useful for the diagnosis and treatment of immune system disorders such as allergy, asthma, inflammation, autoimmune-diseases.
Moreover, the tissue distribution in immune cells (e.g., neutrophils) indicates the polynucleotides and polypeptides corresponding to this gene would be useful for the diagnosis and treatment of a variety of immune system disorders.
Representative uses are described in the "Immune Activity" and "Infectious Disease" sections below, in Example 11, 13, 14, 16, 18, 19, 20, and 27, and elsewhere herein. Briefly, the expression indicates a role in regulating the proliferation; survival;
differentiation;
and/or activation of hematopoietic cell lineages, including blood stem cells.
Involvement in the regulation of cytokine production, antigen presentation, or other processes indicates a usefulness for treatment of cancer (e.g. by boosting immune responses). Expression in cells of lymphoid origin, indicates the natural gene product would be involved in immune functions. Therefore it would also be useful as an agent for immunological disorders including arthritis, asthma, immunodeficiency diseases such as AIDS, leukemia, rheumatoid arthritis, granulomatous disease, inflammatory bowel disease, sepsis, acne, neutropenia, neutrophilia, psoriasis, hypersensitivities, such as T-cell mediated cytotoxicity; immune reactions to transplanted organs and tissues, such as host-versus-graft and graft-versus-host diseases, or autoimmunity disorders, such as autoimmune infertility, Tense tissue injury, demyelination, systemic lupus erythematosis, drug induced hemolytic anemia, rheumatoid arthritis, Sjogren's disease, and scleroderma. Moreover, the protein may represent a secreted factor that influences the differentiation or behavior of other blood cells, or that recruits hematopoietic cells to sites of injury. Thus, this gene product is thought to be useful in the expansion of stem cells and committed progenitors of various blood lineages, and in the differentiation and/or proliferation of various cell types.
Furthermore, the protein may also be used to determine biological activity, raise antibodies, as tissue markers, to isolate cognate ligands or receptors, to identify agents that modulate their interactions, in addition to its use as a nutritional supplement. Protein, as well as, antibodies directed against the protein may show utility as a tumor marker and/or immunotherapy targets for the above listed tissues.
FEATURES OF PROTEIN ENCODED BY GENE NO: 5 The gene encoding the disclosed cDNA is believed to reside on chromosome 15. Accordingly, polynucleotides related to this invention are useful as a marker in linkage analysis for chromosome 15.
This gene is expressed primarily in the following tissues/cDNA libraries:
Soares-placenta 8to9weeks 2NbHP8to9W and to a lesser extent in Human Bone Marrow, treated; Soares ovary tumor NbHOT; normalized infant brain cDNA;
Hypothalamus; stromal cell clone 2.5; NCI CGAP_Lul; Stromal cells 3.88; Human 1 S retina cDNA randomly primed sublibrary; Synovial hypoxia-RSF subtracted;
Breast, Cancer: (4004943 AS); Myoloid Progenitor Cell Line; wilm's tumor; H. Lymph node breast Cancer; Ovary, Cancer (15395A1F): Grade II Papillary Carcinoma;
Stratagene muscle 937209; Stratagene fetal retina 937202; Human Umbilical Vein Endothelial Cells, uninduced; NCI CLAP Pr28; Human Pancreas Tumor, Reexcision; Soares breast 2NbHBst; NCI CGAP_Co3; Colon Carcinoma; Human Placenta; Human Placenta; Human Adult Pulmonary,re-excision; Endothelial cells-control; Human Microvascular Endothelial Cells, fract. A; Soares senescent fibroblasts NbHSF;
Human Testes; Bone Marrow Cell Line (RS4,11 ); H. Frontal cortex,epileptic,re-excision; Human Endometrial Tumor; Activated T-cell(12h)/Thiouridine-re-excision;
Soares fetal lung NbHLI9W; T cell helper II; Primary Dendritic Cells, lib 1;
NCI CGAP_GCB1 and Soares fetal liver spleen 1NFLS.
Polynucleotides and polypeptides of the invention are useful as reagents for differential identification of the tissues) or cell types) present in a biological sample and for diagnosis of diseases and conditions which include but are not limited to:
hemopoietc, reproductive, metabolic and hormonal disorders. Similarly, polypeptides and antibodies directed to these polypeptides are useful in providing immunological probes for differential identification of the tissues) or cell type(s). For a number of disorders of the above tissues or cells, particularly of the immune system, expression of this gene at significantly higher or lower levels may be routinely detected in certain tissues or cell types (e.g., cancerous and wounded tissues) or bodily fluids (e.g., serum, plasma, urine, synovial fluid and spinal fluid) or another tissue or sample taken from an individual having such a disorder, relative to the standard gene expression level, i.e., the expression level in healthy tissue or bodily fluid from an individual not having the disorder.
Elevated expression in hemapoietic tissues and placenta indicates a possible utility for the diagnosis and/or treatment of hemopoietc, reproductive, metabolic and hormonal disorders.
FEATURES OF PROTEIN ENCODED BY GENE NO: 6 In specific embodiments, polypeptides of the invention comprise, or alternatively consists of, an amino acid sequence selected from the group:
GTTTKMAVARLAAVAAWVPCRSWGWAAVPFGPHRGLSVLLARIPQRAPRW
LPACRQKTSLSFLNRPDLPNLAYKKLKGKSPGIIFIPGYLSYMNGTKALAIEEF
CKSLGHACIRFDYSGVGSSDGNSEESTLGKWRKDVLSIIDDLXDGPQILVGSS
LGGWLMLXAXNCTTREGLALIG (SEQ ID NO: 127) and PPSQPESACAASVPLRXLSLSGTLQGAGTTTKMAVARLAAVAAWVPCRSWG
WAAVPFGPHRGLSVLLARIPQRAPRWLPACRQKTSLSFLNRPDLPNLAYKKL
KGKSPGIIFIPGYLSYMNGTKALAIEEFCKSLGHACIRFDYSGVGSSDGNSEES
TLGKWRKDVLSIIDDLADGPQILVGSSLGGWLMLHAAIARPEKVVALIGVAT
AADTLVTKFNQLPVELKKEVEMKGVWSMPSKYSEEGVYNVQYSFIKEAEHH
CLLHSPIPVNCPIRLLHGMKDDIVPWHTSMQVADRVLSTDVDVILRKHSDHR
MREKADIQLLVYTIDDLIDKLSTIVN (SEQ ID NO: 128). Moreover, fragments and variants of these polypeptides (such as, for example, fragments as described herein, polypeptides at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99%
identical to these polypeptides and polypeptides encoded by the polynucleotide which hybridizes, under stringent conditions, to the polynucleotide encoding these polypeptides ) are encompassed by the invention. Antibodies that bind polypeptides of the invention are also encompassed by the invention. Polynucleotides encoding these polypeptides are also encompassed by the invention.
This gene is expressed primarily in the following tissues/cDNA libraries:
Activated T-cells, 24 hrs,re-excision and to a lesser extent in Activated T-Cells, 24 5 hrs.; Soares infant brain 1NIB; Soares fetal liver spleen 1NFLS;
Soares fetal heart NbHHI9W; NCI CGAP Kidl l; Soares retina N2b4HR; Spleen, Chronic lymphocytic leukemia; Soares retina N2b5HR; B-cells (stimulated);
Soares-parathyroid tumor NbHPA; NCI CGAP_GCB1; NCI CGAP Prl l; Human Bone Marrow, re-excision; Stratagene ovarian cancer (#937219); Stratagene HeLa 10 cell s3 937216; NCI CGAP_GC6; NCI CGAP Kid3; NCI CGAP Kids;
Soares fetal lung NbHLI9W; Soares total fetus Nb2HF8 9w;
Soares NFL T GBC_S1; Soares testis NHT; NCI CGAP Kidl2;
Dermatofibrosarcoma Protuberance; NCI CGAP KidB; NCI CGAP-Lu24; Human Adult Pulmonary; NCI CGAP_Pr25; human colon cancer; Stromal Cells; Human 1 S adult (K.Okubo); Healing groin wound, 6.5 hours post incision; Mo7e Cell Line GM-CSF treated (lng/ml); NCI CGAP-Ewl; Apoptotic T-cell; Human Heart; Human Hypothalmus,Schizophrenia; Human Thymus Stromal Cells; Stratagene hNT neuron (#937233); Ovarian Tumor 10-3-95; Hepatocellular Tumor, re-excision; Human Gall Bladder; PC3 Prostate cell line; Primary Dendritic cells,frac 2; Stratagene lung 20 (#937210); Human Bone Marrow, treated; Human fetal heart, Lambda ZAP
Express;
H. Frontal cortex,epileptic,re-excision; NCI CGAP_LuS; Osteoblasts; T cell helper II; Human Cerebellum and Soares_fetal liver spleen_1NFLS S1.
Polynucleotides and polypeptides of the invention are useful as reagents for differential identification of the tissues) or cell types) present in a biological sample and for diagnosis of diseases and conditions which include but are not limited to:
hematopoietic/immune system dysfunction. Similarly, polypeptides and antibodies directed to these polypeptides are useful in providing immunological probes for differential identification of the tissues) or cell type(s). For a number of disorders of the above tissues or cells, particularly of the immune system, expression of this gene at significantly higher or lower levels may be routinely detected in certain tissues or cell types (e.g., immune, cancerous and wounded tissues) or bodily fluids (e.g., serum, plasma, urine, synovial fluid and spinal fluid) or another tissue or sample taken from an individual having such a disorder, relative to the standard gene expression level, i.e., the expression level in healthy tissue or bodily fluid from an individual not having the disorder.
The observed tissue distribution indicates that polynucleotides and polypeptides corresponding to this gene are useful for the diagnosis and/or treatment of hematopoietic/immune system dysfunction. Enriched expression in activated T-cells, spleen , bone marrow suggest a possible role in immune recognition, inflammation, and immunity, and may be useful in a variety of pathologies, including immune system dysfunction, graft-vs. host disease, tumor immunotherapy, and combating susceptibility to infections.
Moreover, the tissue distribution in immune cells indicates the polynucleotides and polypeptides corresponding to this gene would be useful for the diagnosis and treatment of a variety of immune system disorders. Representative uses are described in the "Immune Activity" and "Infectious Disease" sections below, in Example 11, 13, 14, 16, 18, 19, 20, and 27, and elsewhere herein. Briefly, the expression indicates a role in regulating the proliferation; survival; differentiation; and/or activation of hematopoietic cell lineages, including blood stem cells. Involvement in the regulation of cytokine production, antigen presentation, or other processes indicates a usefulness for treatment of cancer (e.g. by boosting immune responses). Expression in cells of lymphoid origin, indicates the natural gene product would be involved in immune functions. Therefore it would also be useful as an agent for immunological disorders including arthritis, asthma, immunodeficiency diseases such as AIDS, leukemia, rheumatoid arthritis, granulomatous disease, inflammatory bowel disease, sepsis, acne, neutropenia, neutrophilia, psoriasis, hypersensitivities, such as T-cell mediated cytotoxicity; immune reactions to transplanted organs and tissues, such as host-versus-graft and graft-versus-host diseases, or autoimmunity disorders, such as autoimmune infertility, Tense tissue injury, demyelination, systemic lupus erythematosis, drug induced hemolytic anemia, rheumatoid arthritis, Sjogren's disease, and scleroderma. Moreover, the protein may represent a secreted factor that influences the differentiation or behavior of other blood cells, or that recruits hematopoietic cells to sites of injury. Thus, this gene product is thought to be useful in the expansion of stem cells and committed progenitors of various blood lineages, and in the differentiation and/or proliferation of various cell types.
Furthermore, the protein may also be used to determine biological activity, raise antibodies, as tissue markers, to isolate cognate ligands or receptors, to identify agents that modulate their interactions, in addition to its use as a nutritional supplement. Protein, as well as, antibodies directed against the protein may show utility as a tumor marker and/or immunotherapy targets for the above listed tissues.
FEATURES OF PROTEIN ENCODED BY GENE NO: 7 Preferred polypeptides of the invention comprise a polypeptide having the amino acid sequence set out in the sequence listing as SEQ ID NO: 129, SEQ >D
NO:
130 and/or SEQ ID NO: 131. Polynucleotides encoding these polypeptides are also provided.
The polypeptide of this gene has been determined to have transmembrane domains at about amino acid positions 165-181, 110-126, 132-148, 56-72, 21-37, and 87-103 of the amino acid sequence referenced in Table 1 for this gene. Based upon these characteristics, it is believed that the protein product of this gene shares structural features to type IIIa membrane proteins.
The gene encoding the disclosed cDNA is believed to reside on chromosome 17. Accordingly, polynucleotides related to this invention are useful as a marker in linkage analysis for chromosome 17.
This gene is expressed primarily in the following tissues/cDNA libraries:
Soares breast 2NbHBst and to a lesser extent in Primary Dendritic Cells, lib l;
Human Activated T-Cells; Human Neutrophil, Activated;
Soares multiple sclerosis 2NbHMSP; Bone Marrow Cell Line (RS4,11);
Soares fetal heart NbHHI9W; Human Fetal Heart, subtracted; NCI CGAP-Lei2;
NCI CGAP-Col6; Breast, Cancer: (9806C012R); Human Aortic Endothelium;
Lung, Cancer (4005313 A3): Invasive Poorly Differentiated Lung Adenocarcinoma,;
LPS activated derived dendritic cells; NCI CGAP-Co9; Human Adipose Tissue, re-excision; NCI CGAP Lyml2; NCI CGAP-Kid6; Human Hippocampus; Human Activated T-Cells, re-excision; Ulcerative Colitis; NCI CGAP_CLL1; Rejected Kidney, lib 4; Ovarian Tumor 10-3-95; Fetal Heart; Colon Carcinoma;

NCI CGAP GC4; human tonsils; Spleen, Chronic lymphocytic leukemia; Human Bone Marrow, treated; Keratinocyte; Soares fetal lung NbHLI9W;
Soares fetal liver spleen-1NFLS_S1 and NCI CGAP_GCB1.
Polynucleotides and polypeptides of the invention are useful as reagents for differential identification of the tissues) or cell types) present in a biological sample and for diagnosis of diseases and conditions which include but are not limited to:
immune system disorders, breast cancer and other neoplastic growth. Similarly, polypeptides and antibodies directed to these polypeptides are useful in providing immunological probes for differential identification of the tissues) or cell type(s). For a number of disorders of the above tissues or cells, particularly of the immune system, expression of this gene at significantly higher or lower levels may be routinely detected in certain tissues or cell types (e.g., immune, cancerous and wounded tissues) or bodily fluids (e.g., serum, plasma, urine, synovial fluid and spinal fluid) or another tissue or sample taken from an individual having such a disorder, relative to the standard gene expression level, i.e., the expression level in healthy tissue or bodily fluid from an individual not having the disorder.
The tissue distribution in breast, hematopoietic cells and various cancers and homology to a membrane protein indicates that polynucleotides and polypeptides corresponding to this gene are useful for the diagnosis and treatment of breast cancer and other neoplastic growth. Moreover, it is a transmembrane protein that could function as a receptor for signal transduction.
The tissue distribution in immune cells (e.g., T-cells and neutrophils) indicates the polynucleotides and polypeptides corresponding to this gene would be useful for the diagnosis and treatment of a variety of immune system disorders.
Representative uses are described in the "Immune Activity" and "Infectious Disease" sections below, in Example 1 l, 13, 14, 16, 18, 19, 20, and 27, and elsewhere herein. Briefly, the expression indicates a role in regulating the proliferation; survival;
differentiation;
and/or activation of hematopoietic cell lineages, including blood stem cells.
Involvement in the regulation of cytokine production, antigen presentation, or other processes indicates a usefulness for treatment of cancer (e.g. by boosting immune responses). Expression in cells of lymphoid origin, indicates~the natural gene product would be involved in immune functions. Therefore it would also be useful as an agent for immunological disorders including arthritis, asthma, immunodeficiency diseases such as ADDS, leukemia, rheumatoid arthritis, granulomatous disease, inflammatory bowel disease, sepsis, acne, neutropenia, neutrophilia, psoriasis, hypersensitivities, such as T-cell mediated cytotoxicity; immune reactions to transplanted organs and tissues, such as host-versus-graft and graft-versus-host diseases, or autoimmunity disorders, such as autoimmune infertility, lense tissue injury, demyelination, systemic lupus erythematosis, drug induced hemolytic anemia, rheumatoid arthritis, Sjogren's disease, and scleroderma. Moreover, the protein may represent a secreted factor that influences the differentiation or behavior of other blood cells, or that recruits hematopoietic cells to sites of injury. Thus, this gene product is thought to be useful in the expansion of stem cells and committed progenitors of various blood lineages, and in the differentiation and/or proliferation of various cell types.
Furthermore, the protein may also be used to determine biological activity, raise antibodies, as tissue markers, to isolate cognate ligands or receptors, to identify agents that modulate their interactions, in addition to its use as a nutritional supplement. Protein, as well as, antibodies directed against the protein may show utility as a tumor marker and/or immunotherapy targets for the above listed tissues.
FEATURES OF PROTEIN ENCODED BY GENE NO: 8 In specific embodiments, polypeptides of the invention comprise, or alternatively consists of, the following amino acid sequence:
VARSSSELPRRLVCSKLRADPGRLTPDACTRPXMSRYLLPLSALGTVAGAAV
LLKDYVTGGACPSKATIPGKTVIVTGANTGIGKQTALELARRGGNIILACRDM
EKCEAAAKDIRGETLNHHVNARHLDLASLKSIREFAAKIIEEEERVDILINNAG

AGHIDFDDLNWQTRKYNTKAAYCQSKLAIVLFTKELSRRLQGTGALGSASLL
LYSEPRAAFP (SEQ ID NO: 132). Moreover, fragments and variants of these polypeptides (such as, for example, fragments as described herein, polypeptides at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% identical to these polypeptides and polypeptides encoded by the polynucleotide which hybridizes, under stringent conditions, to the polynucleotide encoding these polypeptides ) are encompassed by the invention. Antibodies that bind polypeptides of the invention are also encompassed by the invention. Polynucleotides encoding these polypeptides are also encompassed by the invention.
5 This gene is expressed primarily in the following tissues/cDNA libraries:
Soares placenta Nb2HP and to a lesser extent in Soares fetal heart NbHHI9W;
Soares-placenta 8to9weeks 2NbHP8to9W; NCI CGAP_GCB1;
Soares-pregnant uterus NbHPU; NCI CGAP_CLL1; Soares breast 3NbHBst; H.
Leukocytes, normalized cot > SOOA; NCI CGAP_AA1; Stratagene placenta 10 (#937225); Jurkat T-Cell, S phase; Soares breast 2NbHBst; Stratagene colon (#937204); Human Primary Breast Cancer Reexcision; NCI CGAP_Brn25;
NCI CGAP Kids; Activated T-cell(12h)/Thiouridine-re-excision; Larynx carcinoma III; NCI CGAP Pr25; Breast Lyrnph node cDNA library; Healing groin wound -zero hr post-incision (control); Human Whole Brain #2 - Oligo dT > 1.SKb;
15 NCI CGAP_CoIO; LNCAP prostate cell line; TF-1 Cell Line GM-CSF Treated;
Breast Cancer Cell line, angiogenic; NCI CGAP Pr22; Human Fetal Kidney;
Soares-pineal-gland N3HPG; Ovary, Cancer: (4004576 A8); NCI CGAP Br2;
Soares NSF F8 9W_OT PA P S1; NCI CGAP_Co3; PC3 Prostate cell line;
Human Fetal Heart; Activated T-Cell (l2hs)/Thiouridine labelledEco;
20 NCI CGAP_Kid3; NCI CGAP-Brn23; HUMAN B CELL LYMPHOMA;
NCI CGAP-LuS; Soares_parathyroid tumor NbHPA;
Soares fetal lung NbHLI9W; Soares total fetus Nb2HF8 9w and Soares NFL T GBC S 1.
Polynucleotides and polypeptides of the invention are useful as reagents for 25 differential identification of the tissues) or cell types) present in a biological sample and for diagnosis of diseases and conditions which include but are not limited to:
reproductive, metabolic and hormonal disorders. Similarly, polypeptides and antibodies directed to these polypeptides are useful in providing immunological probes for differential identification of the tissues) or cell type(s). For a number of disorders of the above tissues or cells, particularly of the reproductive system, expression of this gene at significantly higher or lower levels maybe routinely detected in certain tissues or cell types (e.g., cancerous and wounded tissues) or bodily fluids (e.g., serum, plasma, urine, synovial fluid and spinal fluid) or another tissue or sample taken from an individual having such a disorder, relative to the standard gene expression level, i.e., the expression level in healthy tissue or bodily fluid from an individual not having the disorder.
The tissue distribution and homology to putative reductase indicates that polynucleotides and polypeptides corresponding to this gene are useful for diagnosis and treatment of reproductive, metabolic and hormonal disorders.
FEATURES OF PROTEIN ENCODED BY GENE NO: 9 Preferred polypeptides of the invention comprise a polypeptide having the amino acid sequence set out in the sequence listing as SEQ ID NO: 134.
Polynucleotides encoding these polypeptides are also provided.
The polypeptide of this gene has been determined to have a transmembrane domain at about amino acid position 24-40 of the amino acid sequence referenced in Table 1 for this gene. Moreover, a cytoplasmic tail encompassing amino acids 1 of this protein has also been determined. Based upon these characteristics, it is believed that the protein product of this gene shares structural features to type II
membrane proteins.
This gene is expressed primarily in the following tissues/cDNA libraries:
Soares NFL T GBC S1 and to a lesser extent in NCI CGAP Kidl2; NTERA2, control; NCI CGAP-Kidl l; human tonsils;
Soares-placenta 8to9weeks 2NbHP8to9W; NCI CGAP_LuS; Nine Week Old Early Stage Human; Soares total fetus Nb2HF8 9w; Soares testis NHT and NCI CGAP GCB 1.
Polynucleotides and polypeptides of the invention are useful as reagents for differential identification of the tissues) or cell types) present in a biological sample and for diagnosis of diseases and conditions which include but are not limited to:
renal, immune, and developmental disorders.. Similarly, polypeptides and antibodies directed to these polypeptides are useful in providing immunological probes for differential identification of the tissues) or cell type(s). For a number of disorders of the above tissues or cells, particularly of the immune system, expression of this gene at significantly higher or lower levels may be routinely detected in certain tissues or cell types (e.g., cancerous and wounded tissues) or bodily fluids (e.g., serum, plasma, urine, synovial fluid and spinal fluid) or another tissue or sample taken from an individual having such a disorder, relative to the standard gene expression level, i.e., S the expression level in healthy tissue or bodily fluid from an individual not having the disorder.
The tissue distribution in kidney, tonsil, embryonic/fetal tissues and neutrophils indicates that polynucleotides and polypeptides corresponding to this gene are useful for the diagnosis and treatment of renal, immune, and developmental disorders.
FEATURES OF PROTEIN ENCODED BY GENE NO: 10 The computer algorithm BLASTX has been used to determine that the translation product of this gene shares sequence homology with, as a non-limiting example, the sequence accessible through the following database accession no.
emb~CAA10646.1 ~ (all information available through the recited accession number is incorporated herein by reference) which is described therein as "~ (AJ1323S6) ARS
component B precursor [Mus musculus]" A partial alignment demonstrating the observed homology is shown immediately below.
>emb~CAA10646.1~ (AJ132356) ARS component B precursor [Mus musculus]
2S >sp~Q9ZOK7~Q9ZOK7 ARS COMPONENT B PRECURSOR.
Length = 110 Plus Strand HSPs:
3 0 Score = 174 (61.3 bits), Expect = 1.4e-12, P = 1.4e-12 Identities = 34/84 (40~), Positives = 43/84 (51~), Frame = +2 Q: 65 ALRCYVCPEPTGVSDCVTIAXXXXXXXXXXXXLYSREIVYPFQGDSTVTKSCASKCKPSD 244 A RCY C +PT ++ C IA L + E +PF VT+SC+S C +D
3 S S: 22 AFRCYTCEQPTAINSCKNIAQCKMEDTACKTVLETVEAAFPFNHSPMVTRSCSSSCLATD 81 Q: 245 VDGIGQTLPVSCCNTELCNVDGAP 316 DGIG PV CC +LCN G P
S: 82 PDGIGVAHPVFCCFRDLCN-SGFP 104 The segment of emb~CAA10646.1 ~ that is shown as "S" above is set out in the sequence listing as SEQ ID NO: 135. Based on the structural similarity these homologous polypeptides are expected to share at least some biological activities.
Such activities are known in the art, some of which are described elsewhere herein.
Assays for determining such activities are also known in the art, some of which have been described elsewhere herein. Preferred polypeptides of the invention comprise a polypeptide having the amino acid sequence set out in the sequence listing as NO: 136 which corresponds to the Q sequence in the alignment shown above (gaps introduced in a sequence by the computer are, of course, removed).
This gene is expressed primarily in the following tissues/cDNA libraries:
Bone Marrow Cell Line (RS4,11 ) and to a lesser extent in Osteoblasts; Human fetal heart, Lambda ZAP Express; Human Thymus; Stratagene endothelial cell 937223;
Hemangiopericytoma; Human Fetal Lung III; Soares adult brain N2b5HB55Y;
Endothelial-induced; Soares fetal liver spleen 1NFLS; wilm's tumor; H
Macrophage (GM-CSF treated), re-excision; Anergic T-cell; Human Lung Cancer,re-excision;
Human Fetal Epithelium (Skin); T-Cell PHA 16 hrs; Apoptotic T-cell; B-cells (stimulated); Soares placenta Nb2HP; Smooth muscle, serum induced,re-exc;
breast lymph node CDNA library; Human Fetal Kidney, Reexcision; Bone marrow;
Soares fetal lung NbHLI9W; Smooth Muscle Serum Treated, Norm; HM1; Human Fetal Kidney; HUMAN JURKAT MEMBRANE BOUND POLYSOMES; Epithelial-TNFa and INF induced; Smooth muscle, serum treated; Early Stage Human Brain;
Smooth muscle,control; HUMAN B CELL LYMPHOMA; T cell helper II;
NCI CGAP_Prl O; Amniotic Cells - Primary Culture; Ovary, Cancer: (4004332 A2);
NCI CGAP Alvl; Human Pancreas Tumor; Human Fetal Brain; Resting T-Cell Library,II; Activated T-Cell (l2hs)/Thiouridine labelledEco; Colon Normal III;
Soares NhHMPu S1; Resting T-Cell, re-excision; Ovary, Cancer (15395A1F): Grade II Papillary Carcinoma; NCI CGAP-Ewl; Stratagene muscle 937209; Human Testes Tumor; HM3; Raji Cells, cyclohexamide treated; Glioblastoma; NCI CGAP_Prl2;
CD34 depleted Buffy Coat (Cord Blood); Human Uterine Cancer; Human Umbilical Vein Endothelial Cells, uninduced; Human Fetal Dura Mater; Human Activated T-Cells; Ovary, Normal: (9805C040R); Human umbilical vein endothelial cells, IL-induced; Human Whole Six Week Old Embryo; Human Gall Bladder; 12 Week Early Stage Human II, Reexcision; Soares-pregnant uterus NbHPU; HSC172 cells;
Apoptotic T-cell, re-excision; Ovary, Cancer: (4004562 B6) Papillary Serous Cystic Neoplasm, Low Malignant Pot; HL-60, PMA 4H, re-excision; Human Colon, re-excision; Jurkat T-cell G1 phase; Prostate BPH; T-Cell PHA 24 hrs; Colon Tumor;
Human Microvascular Endothelial Cells, fract. A; Spleen, Chronic lymphocytic leukemia; T Cell helper I; Soares ovary tumor NbHOT;
Soares fetal heart NbHHI9W; NCI CGAP-Lym3; NCI CGAP_Kidl;
NCI CGAP Prl 1; stomach cancer (human); Human OB HOS treated (10 nM E2) fraction I; NCI CGAP_Brl.l; Human Fetal Bone; Amniotic Cells - TNF induced;
Human Skin Tumor; Lung Carcinoma A549 TNFalpha activated; NCI CGAP-GC3;
human corpus colosum; Smooth muscle, ILlb induced; Human Osteoclastoma Stromal Cells - unamplified; Human Adipose Tissue, re-excision; Human Osteosarcoma; H. Meningima, M1; Human Prostate; NCI CGAP Pr2; Human Thymus; L428; NCI CGAP_Co3; 12 Week Old Early Stage Human; Adipocytes;
Human Synovial Sarcoma; Human Testes, Reexcision; Human Primary Breast Cancer Reexcision; CD34 depleted Buffy Coat (Cord Blood), re-excision; Human Bone Marrow, treated; Hodgkin's Lymphoma II; NCI CGAP-LuS; Activated T-cell(12h)/Thiouridine-re-excision; KMH2 cell line; Kidney Pyramids; Human Lung Cancer, subtracted; CD34+cells, II, FRACTION 2; Human Leukocytes; Activated T-Cells, 8 hrs., ligation 2; Human 7 Weeks Old Embryo, subtracted; NCI CGAP HN4;
Soares NbHFB; Liver HepG2 cell line.; NCI CGAP Pr23; HL-60, RA 4h, Subtracted; Whole 6 Week Old Embryo; Lung, Normal: (4005313 B 1 );
NCI CGAP PrB; Human OB MG63 control fraction I; stromal cell clone 2.5;
NCI CGAP-GCS; Human Colon, subtraction; NCI CGAP_Pr4.l; Human OB MG63 treated (10 nM E2) fraction I; Barstead spleen HPLRB2; Human placenta cDNA
(TFujiwara); H. Atrophic Endometrium; Lung, Cancer (4005313 A3): Invasive Poorly Differentiated Lung Adenocarcinoma,; Activated T-cells; Cem cells cyclohexamide treated; Human Normal Breast; Dendritic Cells From CD34 Cells;
STROMAL -OSTEOCLASTOMA; Stratagene placenta (#937225); Stratagene ovary (#937217); NCI CGAP-Col4; Human Osteoclastoma, re-excision; LNCAP prostate cell line; Jurkat T-Cell, S phase; Myoloid Progenitor Cell Line; NCI CGAP-Lyml2;
H. Lymph node breast Cancer; NCI .CGAP Pr22; Stratagene HeLa cell s3 937216;

Liver, Hepatoma; Human Hippocampus; Human Rhabdomyosarcoma; Bone Marrow Stromal Cell, untreated; Macrophage (GM-CSF treated); Hepatocellular Tumor, re-excision; Human Ovary; Stratagene colon (#937204); Human Substantia Nigra;
Human Eosinophils; Human Placenta; NCI CGAP-CoB; NCI CGAP-GC4; Human S Placenta; Endothelial cells-control; Stratagene lung (#937210);
Soares senescent fibroblasts NbHSF; NCI CGAP Kids; Pancreas Tumor PCA4 Tu; CD34 positive cells (Cord Blood); Soares-placenta 8to9weeks 2NbHP8to9W;
Human Testes; Human Endometrial Tumor; Human 8 Week Whole Embryo; Soares melanocyte 2NbHM; Soares testis NHT; Primary Dendritic Cells, lib 1; Human skin 10 Tumor, subtracted; Human Whole 7 Week Old Embryo (II); Human Lung Cancer;
Lymph node, abnorm. cell line (ATCC #7225); HL-60, PMA 4H, subtracted; Early Stage Human Liver, fract (II); TH1 cells; Hl-60, untreated, subtracted; Supt cells, cyclohexamide treated, differentially expressed; Human ovary tumor cell OV350721;
Human Adult Lymph Node, subtracted; Adrenal Gland,normal; NCI CGAP_Li2;
15 Smooth muscle control 2; Human Umbilical Vein Endothelial Cells, fract. B;
NCI CGAP_Br7; Placenta; Cheek Carcinoma; NCI CGAP-OvB; Colon, normal;
Tongue carcinoma; Human Fetal Liver, mixed 10 & 14 week; Human placenta polyA+ (TFujiwara); Human Normal Cartilage Fraction IV; WI 38 cells; Breast, Cancer: (4005385 A2); Human B Cell 8866; Human Thymus Tumor, subtracted;
20 Tongue Tumour; Activated T-Cells, 8 hrs, subtracted; Stratagene colon HT29 (#937221 ); Colon, Cancer: (9808C064R); NCI CGAP-Ov36; Human Colon Cancer, subtracted; Human Fetal Lung; Human epithelioid sarcoma; NCI CGAP-Br3;
Human OB HOS treated (1 nM E2) fraction I; Human Pituitary, re-excision; LNCAP
untreated; NCI CGAP_Eso2; NCI CGAP_Pr2l; NCI CGAP_LymS; Human Gall 25 Bladder, fraction II; Breast, Cancer: (9806C012R); L1 Cell line; HUMAN
TONSILS, FRACTION 2; Human Adult Spleen; NCI CGAP Pr6; Human (HCC) cell line liver (mouse) metastasis, remake; Human White Adipose; Human colon carcinoma (HCC) cell line, remake; Jia bone marrow stroma; Human Colon Carcinoma (HCC) cell line;
Human Placenta; Healing Abdomen wound,70&90 min post incision; HUMAN
30 STOMACH; NCI CGAP_Lul; Aorta endothelial cells + TNF-a; NCI CGAP-Thyl;
Lung, Cancer (4005163 B7): Invasive, Poorly Diff. Adenocarcinoma, Metastatic;
Early Stage Human Lung, subtracted; Stromal Cells; Human Soleus; Human Pineal Gland; Messangial cell, frac 2; H. Whole Brain #2, re-excision; HEL cell line;
Stomach cancer (human),re-excision; Human endometrial stromal cells-treated with estradiol; pBMC stimulated w/ poly I/C; H Female Bladder, Adult; Human endometrial stromal cells-treated with progesterone; Human Whole Brain #2 -Oligo dT > I.SKb; NCI CGAP_Ov2; B-cells (unstimulated); Human endometrial stromal cells; Synovial hypoxia; Spleen metastic melanoma; Human Chronic Synovitis;
NCI CGAP_Pr3; H. Kidney Medulla, re-excision; Mo7e Cell Line GM-CSF treated (lng/ml); Human Bone Marrow, re-excision; TF-1 Cell Line GM-CSF Treated;
Human Dermal Endothelial Cells,untreated; B-Cells; Human Brain, Striatum;
Stratagene fetal spleen (#937205); Stratagene fetal retina 937202; 12 Week Old Early Stage Human, II; Soares_pineal_gland N3HPG; Human Heart; Stromal cell TF274;
NCI CGAP_Gas4; Macrophage-oxLDL; Human Hypothalmus,Schizophrenia;
Human Pancreas Tumor, Reexcision; Human Activated Monocytes; Human Placenta (re-excision); Ovary, Cancer: (4004576 A8); NCI CGAP_Br2; Human Activated T-Cells, re-excision; Human Testes Tumor, re-excision; NCI CGAP-CLL1; Ovarian Tumor 10-3-95; CHME Cell Line,untreated; Fetal Liver, subtraction II; Brain frontal cortex; NCI CGAP_Kidl l; Dendritic cells, pooled; Ovary, Cancer(4004650 A3):
Well-Differentiated Micropapillary Serous Carcinoma; Neutrophils control, re-excision; Pancreas normal PCA4 No; Human Fetal Heart; Human Adult Pulmonary,re-excision; Human Osteoclastoma; NCI CGAP-Brn23; neutrophils control; Keratinocyte; Soares_parathyroid tumor NbHPA; Colon Tumor II; Human Cerebellum and Soares total fetus Nb2HF8 9w.
Polynucleotides and polypeptides of the invention are useful as reagents for differential identification of the tissues) or cell types) present in a biological sample and for diagnosis of diseases and conditions which include but are not limited to:
hemopoietic, immune and inflammatory defects, skeletal and blood disorders, leukemias and other neoplasms. Similarly, polypeptides and antibodies directed to these polypeptides are useful in providing immunological probes for differential identification of the tissues) or cell type(s). For a number of disorders of the above tissues or cells, particularly of the immune system, expression of this gene at significantly higher or lower levels may be routinely detected in certain tissues or cell types (e.g., immune, cancerous and wounded tissues) or bodily fluids (e.g., serum, plasma, urine, synovial fluid and spinal fluid) or another tissue or sample taken from an individual having such a disorder, relative to the standard gene expression level, i.e., the expression level in healthy tissue or bodily fluid from an individual not having the disorder.
Enriched expression in bone marrow and osteoblasts and homology to Ly-6 family antigens indicates that polynucleotides and polypeptides corresponding to this gene are useful for diagnosis, study and treatment of hemopoietic, immune and inflammatory defects, skeletal and blood disorders, leukemias and other neoplasms.
The tissue distribution in immune cells (e.g., T-cells) indicates the polynucleotides and polypeptides corresponding to this gene would be useful for the diagnosis and treatment of a variety of immune system disorders.
Representative uses are described in the "Immune Activity" and "Infectious Disease" sections below, in Example 11, 13, 14, 16, 18, 19, 20, and 27, and elsewhere herein. Briefly, the expression indicates a role in regulating the proliferation; survival;
differentiation;
and/or activation of hematopoietic cell lineages, including blood stem cells.
Involvement in the regulation of cytokine production, antigen presentation, or other processes indicates a usefulness for treatment of cancer (e.g. by boosting immune responses). Expression in cells of lymphoid origin, indicates the natural gene product would be involved in immune functions. Therefore it would also be useful as an agent for immunological disorders including arthritis, asthma, immunodeficiency diseases such as AIDS, leukemia, rheumatoid arthritis, granulomatous disease, inflammatory bowel disease, sepsis, acne, neutropenia, neutrophilia, psoriasis, hypersensitivities, such as T-cell mediated cytotoxicity; immune reactions to transplanted organs and tissues, such as host-versus-graft and graft-versus-host diseases, or autoimmunity disorders, such as autoimmune infertility, Tense tissue injury, demyelination, systemic lupus erythematosis, drug induced hemolytic anemia, rheumatoid arthritis, Sjogren's disease, and scleroderma. Moreover, the protein may represent a secreted factor that influences the differentiation or behavior of other blood cells, or that recruits hematopoietic cells to sites of injury. Thus, this gene product is thought to be useful in the expansion of stem cells and committed progenitors of various blood lineages, and in the differentiation and/or proliferation of various cell types.
Furthermore, the protein may also be used to determine biological activity, raise antibodies, as tissue markers; to isolate cognate ligands or receptors, to identify agents that modulate their interactions, in addition to its use as a nutritional supplement. Protein, as well as, antibodies directed against the protein may show utility as a tumor marker and/or immunotherapy targets for the above listed tissues.
FEATURES OF PROTEIN ENCODED BY GENE NO: 11 The gene encoding the disclosed cDNA is believed to reside on chromosome 20. Accordingly, polynucleotides related to this invention are useful as a marker in linkage analysis for chromosome 20.
In specific embodiments, polypeptides of the invention comprise, or alternatively consists of, the following amino acid sequence:
MRRLLLVTSLVVVLLWEAGAVPAPKVPIKMQVKHWPSEQDPEKAWGARW
EPPEKDDQLVVLFPVQKPKLLTTEEKPRGQGRGPILPGTKAWMETEDTLGRV
LSPEPDHDSLYHPPPEEDQGEERPRLWVMPNHQVLLGPEEDQDHIYHPQ
(SEQ ID NO: 157). Moreover, fragments and variants of these polypeptides (such as, for example, fragments as described herein, polypeptides at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% identical to these polypeptides and polypeptides encoded by the polynucleotide which hybridizes, under stringent conditions, to the polynucleotide encoding these polypeptides ) are encompassed by the invention.
Antibodies that bind polypeptides of the invention are also encompassed by the invention. Polynucleotides encoding these polypeptides (such, as for example, the polynucleotide shown in SEQ ID NO: 156) are also encompassed by the invention.
This gene is expressed primarily in the following tissues/cDNA libraries:
Soares fetal liver spleen 1NFLS and to a lesser extent in Hepatocellular Tumor, re-excision; Fetal Liver, subtraction II; Breast, Cancer:. (4004943 AS); Liver, Hepatoma;
NCI CGAP_Co3; Normal colon; Colon Tumor II; Colon, tumor; Colon Tumor;
Human Colon Cancer, subtracted; NCI CGAP-GC3; Human Epididymus;
Hepatocellular Tumor; NCI CGAP_Col O; Soares adult brain N2b4HB55Y; Ovary, Cancer (15395A1F): Grade II Papillary Carcinoma; Stratagene NT2 neuronal precursor 937230; Human Uterine Cancer; Stromal cell TF274; Human Liver, normal; Human Ovarian Cancer Reexcision; NCI CGAP GC6; NCI CGAP Kid3;

Soares multiple sclerosis 2NbHMSP; Colon Normal III and Soares infant brain 1NIB.
Polynucleotides and polypeptides of the invention are useful as reagents for differential identification of the tissues) or cell types) present in a biological sample and for diagnosis of diseases and conditions which include but are not limited to: liver and colon cancer. Similarly, polypeptides and antibodies directed to these polypeptides are useful in providing immunological probes for differential identification of the tissues) or cell type(s). For a number of disorders of the above tissues or cells, particularly of the hepatic and gastrointestinal systems, expression of this gene at significantly higher or lower levels may be routinely detected in certain tissues or cell types (e.g., cancerous and wounded tissues) or bodily fluids (e.g., serum, plasma, urine, synovial fluid and spinal fluid) or another tissue or sample taken from an individual having such a disorder, relative to the standard gene expression level, i.e., the expression level in healthy tissue or bodily fluid from an individual not having the disorder.
The tissue distribution highly enriched in fetal liver, hepatomas, normal colon and colon cancers indicates that polynucleotides and polypeptides corresponding to this gene are useful for the diagnosis and treatment of liver and colon as well as other cancers.
FEATURES OF PROTEIN ENCODED BY GENE NO: 12 In specific embodiments, polypeptides of the invention comprise, or alternatively consists of, an amino acid sequence selected from the group:
RPRKGLQGLWGAVFCKGQAALALAACGVLLGSGGPAAAWEADPRGQVWP
CPDRARTEVGGSPCAVPSSPEEAGLKPPGVAEASPCQRPKPRLGFYRCSFPST
WSPSTPSSPNSQPPFFFFLHASKVQGPQMYRSLMYHPA REPADYQAKK (SEQ
ID NO: 138) and MAKSTLSPCSYTSAPAHLLTALVLIFIHPNPIPSKAHSHRCFLLKISFNFYTAKL
CPPPGH
STLSVSLWNLSQSPMLVLSLLPATPCFWSVCSTPLEDSVPGGRTKDYTYLHFL

LWLILSRCSGSICSMNG YPSIKRALPHPYLQNDPLPDFSEPT (SEQ ID NO:
137). Moreover, fragments and variants of these polypeptides (such as, for example, fragments as described herein, polypeptides at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% identical to these polypeptides and polypeptides encoded by the 5 polynucleotide which hybridizes, under stringent conditions, to the polynucleotide encoding these polypeptides ) are encompassed by the invention. Antibodies that bind polypeptides of the invention are also encompassed by the invention.
Polynucleotides encoding these polypeptides are also encompassed by the invention.
This gene is expressed primarily in the following tissues/cDNA libraries:
10 Larynx Carcinoma Polynucleotides and polypeptides of the invention are useful as reagents for differential identification of the tissues) or cell types) present in a biological sample and for diagnosis of diseases and conditions which include but are not limited to:
cancer of the larynx and cancer, in general. Similarly, polypeptides and antibodies 15 directed to these polypeptides are useful in providing immunological probes for differential identification of the tissues) or cell type(s). For a number of disorders of the above tissues or cells, particularly of the larynx, expression of this gene at significantly higher or lower levels may be routinely detected in certain tissues or cell types (e.g., cancerous and wounded tissues) or bodily fluids (e.g., serum, plasma, 20 urine, synovial fluid and spinal fluid) or another tissue or sample taken from an individual having such a disorder, relative to the standard gene expression level, i.e., the expression level in healthy tissue or bodily fluid from an individual not having the disorder.
The tissue distribution in larynx carcinoma indicates that polynucleotides and 25 polypeptides corresponding to this gene are useful for the diagnosis and treatment of this disease and other neoplastic growth.
FEATURES OF PROTEIN ENCODED BY GENE NO: 13 In specific embodiments, polypeptides of the invention comprise, or alternatively consists of, the following amino acid sequence:

MNSKGQYPTQPTYPVQPPGNPVYPQTLHLPQAPPYTDAPPAYSELYRPSFVHP
GAA
TVPTMSAAFPGASLYLPMAQSVAVGPLGSTIPMAYYPVGPIYPPGSTVLVEGG
YDAGARFGAGATAGNIP
PPPPGCPPNAAQLAVMQGANVLVTQRKGNFFMGGSDGGYTIW (SEQ ID NO:
139). Moreover, fragments and variants of these polypeptides (such as, for example, fragments as described herein, polypeptides at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% identical to these polypeptides and polypeptides encoded by the polynucleotide which hybridizes, under stringent conditions, to the polynucleotide encoding these polypeptides ) are encompassed by the invention. Antibodies that bind polypeptides of the invention are also encompassed by the invention.
Polynucleotides encoding these polypeptides are also encompassed by the invention.
This gene is expressed primarily in the following tissues/cDNA libraries:
Primary Dendritic Cells, lib 1 and to a lesser extent in Soares fetal liver spleen 1 S 1NFLS; Soares-pregnant uterus NbHPU; Soares~arathyroid tumor NbHPA;
Soares fetal lung NbHLI9W; Human Endometrial Tumor;
Soares fetal heart NbHHI9W; Soares infant brain 1NIB; Activated T-Cells,l2 hrs,re-excision; Activated T-cell(12h)/Thiouridine-re-excision;
Soares-placenta 8to9weeks 2NbHP8to9W; NCI CGAP Ut3; Human Neutrophil;
Colon Tumor II; Soares fetal liver spleen-1NFLS_S1; HEL cell line;
NCI CGAP_Panl; Human T-Cell Lymphoma; Human Eosinophils; Colon Normal II;
NCI CGAP_CoB; Human Fetal Kidney, Reexcision; Human Fetal Heart; Human Bone Marrow, treated; Human 8 Week Whole Embryo; T cell helper II;
Soares NhHMPu S1; NCI CGAP_Lyml2; NTERA2, control; Smooth muscle, serum treated; B-cells (stimulated); Human Osteoclastoma; Human Microvascular Endothelial Cells, fract. A; NCI CGAP-Kid3; Spleen, Chronic lyrnphocytic leukemia; Nine Week Old Early Stage Human; Soares total fetus Nb2HF8 9w; H.
Striatum Depression, subtracted; Human retina cDNA Tsp509I-cleaved sublibrary;
Human Umbilical Vein Endothelial Cells, fract. A; Human Soleus; Hepatocellular Tumor; Human Prostate Cancer, Stage C fraction; NCI CGAP_CoIO;
NCI CGAP Co 14; NCI CGAP_Pr22; Human Fetal Kidney; Human Heart; Human Placenta (re-excision); Human Adipose; Human Chondrosarcoma; Pancreas Islet Cell Tumor; Dendritic cells, pooled; Primary Dendritic cells,frac 2; Endothelial-induced;
Human Primary Breast Cancer Reexcision; Stratagene lung (#937210);
NCI CGAP_Brn23; Monocyte activated; T Cell helper I; Human fetal heart, Lambda ZAP Express; H. Frontal cortex,epileptic,re-excision; Hodgkin's Lymphoma II;
NCI CGAP_LuS; normalized infant brain cDNA; Soares melanocyte 2NbHM;
Kidney medulla; NCI CGAP-Pr23; NCI CGAP_Pr9; NCI CGAP-Pr6; Human colon carcinoma (HCC) cell line, remake; Human Adult Pulmonary; eosinophil-II,S
induced; Normal Human Trabecular Bone Cells; Human Neutrophils, Activated, re-excision; Stromal Cells; B Cell lymphoma; STROMAL -OSTEOCLASTOMA;
pBMC stimulated w/ poly I/C; NCI CGAP_Co9; H Female Bladder, Adult; NTERA2 + retinoic acid, 14 days; Myoloid Progenitor Cell Line; Healing groin wound, 6.5 hours post incision; NCI CGAP Ut2; NCI CGAP_Pr3; Ovary, Cancer (15395A1F):
Grade II Papillary Carcinoma; T-Cell PHA 16 hrs; Breast Cancer Cell line, angiogenic; NCI CGAP Prl; Apoptotic T-cell; Human Umbilical Vein Endothelial Cells, uninduced; Human Activated T-Cells; T-Cell PHA 24 hrs; Human Pancreas Tumor; Stromal cell TF274; Human umbilical vein endothelial cells, IL-4 induced;
NCI CGAP_Br2; Soares NSF F8 9W_OT PA P S1; Macrophage (GM-CSF
treated); CHME Cell Line,treated 5 hrs; Rejected Kidney, lib 4; Human Adult Heart,re-excision; Resting T-Cell Library,II; Colon Carcinoma; Human Placenta;
NCI CGAP_GC4; Ovary, Cancer(4004650 A3): Well-Differentiated Micropapillary Serous Carcinoma; Human Fetal Lung III; Human Ovarian Cancer Reexcision;
Human Placenta; Bone marrow; Human Neutrophil, Activated; Human Adult Pulmonary,re-excision; Activated T-Cell (l2hs)/Thiouridine labelledEco;
Anergic T-cell; Human Amygdala; Soares senescent fibroblasts NbHSF; Smooth muscle,control; Soares multiple sclerosis 2NbHMSP; HUMAN B CELL
LYMPHOMA; Human Testes; Bone Marrow Cell Line (RS4,11); Keratinocyte;
Human Cerebellum; Soares testis NHT; NCI CGAP GCB1; Human Old Ovary, subtracted; N3HFLSK20; Human Fetal Brain, normalized c5-11-26; Salivary gland, re-excision; Human Fetal Kidney; Human Adult Small Intestine; NCI CGAP-PNSl;
Larynx Normal; Colon Tumor; Human Amygdala Depression, re-excision; Pharynx carcinoma; Human Adult Heart; Human Umbilical Vein Endothelial cells, frac B, re-excision; Human Prostate BPH, re-excision; CD34+cells, II, FRACTION 2; Human Leukocytes; Saos2 Cells, Vitamin D3 Treated; Activated T-Cells, 12 hrs.;
NCI CGAP PrB; NCI CGAP BrlS; NCI CGAP_Pr2l; Human promyelocyte;
Human Colon; Human Aortic Endothelium; NCI CGAP_KidB; Human OB HOS
treated (10 nM E2) fraction I; Adipocytes,re-excision; Human Pancreatic Carcinoma;
Human Pituitary, subtracted; NCI CGAP_Lul; Lung, Cancer (4005313 A3): Invasive Poorly Differentiated Lung Adenocarcinoma,; Breast Lyrnph node cDNA library;
Human heart cDNA (YNakamura); HSA 172 Cells; CD40 activated monocyte dendridic cells; Human Pineal Gland; Raji Cells, cyclohexamide treated; Smooth Muscle- HASTE normalized; NTERA2 teratocarcinoma cell line+retinoic acid (14 days); Human adult small intestine,re-excision; Human Lung Cancer,re-excision;
Human Tonsils, Lib 2; Dendritic Cells From CD34 Cells; Apoptotic T-cell, re-excision; Healing groin wound - zero hr post-incision (control); Palate normal;
Human Colon Cancer,re-excision; Human Hypothalamus,schizophrenia, re-excision;
human corpus colosum; Human Umbilical Vein, Endo. remake; Amniotic Cells -Primary Culture; Human Fetal Epithelium (Skin); Ovary, Cancer: (15799A1F) Poorly differentiated carcinoma; Human Stomach,re-excision; NCI CGAP_Ov2; Healing groin wound, 7.5 hours post incision; HL-60, PMA 4H, re-excision; B-cells (unstimulated); Human Osteoclastoma, re-excision; NCI CGAP Prl2; Ovary, Cancer: (4004332 A2); Prostate BPH; H. Lymph node breast Cancer; Human Adult Small Intestine; Hippocampus, Alzheimer Subtracted; H. Kidney Medulla, re-excision; CD34 depleted Buffy Coat (Cord Blood); Stratagene neuroepithelium (#937231 ); Human Bone Marrow, re-excision; Gessler Wilms tumor; B-Cells;
Human Brain, Striatum; Human Thymus; NCI CGAP Utl; Stratagene fetal retina 937202;
Human Osteoblasts II; Human Uterine Cancer; NCI CGAP-Gas4; Macrophage-oxLDL; Human Hypothalmus,Schizophrenia; Human Pancreas Tumor, Reexcision;
Human Activated Monocytes; Liver, Hepatoma; Ovary, Cancer (9809C332): Poorly differentiated adenocarcinoma; Ovary, Cancer: (4004576 A8); Human Activated T-Cells, re-excision; Ulcerative Colitis; NCI CGAP_CLLl; Ovarian Tumor 10-3-95;
Hepatocellular Tumor, re-excision; Human Ovary; NCI CGAP-Co3; Human Gall Bladder; Fetal Heart; CHME Cell Line,untreated; Colon Tumor; Human Substantia Nigra; Early Stage Human Brain; H Macrophage (GM-CSF treated), re-excision;
Human Synovial Sarcoma; Endothelial cells-control; NCI CGAP-Brn25;

NCI CGAP-Kids; Neutrophils IL-1 and LPS induced; Osteoblasts; Colon Normal III
and Soares placenta Nb2HP.
Polynucleotides and polypeptides of the invention are useful as reagents for differential identification of the tissues) or cell types) present in a biological sample and for diagnosis of diseases and conditions which include but are not limited to:
developmental diseases and immune system disorders. Similarly, polypeptides and antibodies directed to these polypeptides are useful in providing immunological probes for differential identification of the tissues) or cell type(s). For a number of disorders of the above tissues or cells, particularly of the immune system, expression of this gene at significantly higher or lower levels may be routinely detected in certain tissues or cell types (e.g., cancerous and wounded tissues) or bodily fluids (e.g., serum, plasma, urine, synovial fluid and spinal fluid) or another tissue or sample taken from an individual having such a disorder, relative to the standard gene expression level, i.e., the expression level in healthy tissue or bodily fluid from an individual not having the disorder.
The tissue distribution in embryonic/fetal tissues and hematopoietic cells indicates that polynucleotides and polypeptides corresponding to this gene are useful for the diagnosis and treatment of developmental diseases and immune system disorders.
FEATURES OF PROTEIN ENCODED BY GENE NO: 14 The gene encoding the disclosed cDNA is believed to reside on chromosome 9. Accordingly, polynucleotides related to this invention are useful as a marker in linkage analysis for chromosome 9.
This gene is expressed primarily in the following tissues/cDNA libraries:
Soares infant brain 1NIB and to a lesser extent in Human Cerebellum; Early Stage Human Brain; normalized infant brain cDNA; Soares-pregnant uterus NbHPU;
Soares breast 2NbHBst; Soares-parathyroid tumor NbHPA;
Soares NFL T GBC_S1; NCI CGAP-GC4; Soares placenta Nb2HP; Osteoblasts;
Soares fetal liver spleen 1NFLS; Soares NbHFB; Normalized infant brain, Bento Soares; Human Cerebellum, subtracted; Human Epididymus; wilm's tumor;
Stratagene lung carcinoma 937218; Human Umbilical Vein, Reexcision;
NCI CGAP_Pr28; Pancreas Islet Cell Tumor; NCI CGAP Brn25; NCI CGAP_LuS;
Soares fetal heart_NbHHI9W; Soares NhHMPu_S1; Soares testis NHT; HepG2 S Cells, lambda library; Infant brain, Bento Soares; Human Aortic Endothelium;
NCI CGAP Ov23; Lung, Cancer (4005313 A3): Invasive Poorly Differentiated Lung Adenocarcinoma,; NCI CGAP-Colt; H. Epididiymus, cauda; NCI CGAP_GC3;
NCI CGAP Ut3; Human adult small intestine,re-excision; NCI CGAP_AA1;
Human Hypothalamus,schizophrenia, re-excision; H. Kidney Cortex, subtracted;
10 Human Frontal Cortex, Schizophrenia; Human Whole Brain #2 - Oligo dT >
l.SKb;
Human Osteosarcoma; NCI CGAP_Col4; Breast, Normal: (4005522B2);
NCI CGAP_Ut2; H. Kidney Medulla, re-excision; NCI CGAP-Ewl; Human Brain, Striatum; Soares-pineal-gland N3HPG; Human Pancreas Tumor; Olfactory epithelium,nasalcavity; Ulcerative Colitis; Hemangiopericytoma; Colon Carcinoma;
15 Colon Normal II; Soares breast 3NbHBst; NCI CGAP-Co8; Human Fetal Lung III;
Human Synovial Sarcoma; NCI CGAP-GC6; Human Adult Pulmonary,re-excision;
Endothelial cells-control; NCI CGAP Kids; NCI CGAP_Brn23;
Soares multiple sclerosis 2NbHMSP; Human Testes; Soares ovary tumor NbHOT;
Keratinocyte; Nine Week Old Early Stage Human; Soares_total fetus Nb2HF8 9w 20 and Soares fetal liver spleen-1NFLS_S1.
Polynucleotides and polypeptides of the invention are useful as reagents for differential identification of the tissues) or cell types) present in a biological sample and for diagnosis of diseases and conditions which include but are not limited to:
neurological and neurodevelopmental disorders. Similarly, polypeptides and 25 antibodies directed to these polypeptides are useful in providing immunological probes for differential identification of the tissues) or cell type(s). For a number of disorders of the above tissues or cells, particularly of the nervous system, expression of this gene at significantly higher or lower levels may be routinely detected in certain tissues or cell types (e.g., cancerous and wounded tissues) or bodily fluids (e.g., 30 serum, plasma, urine, synovial fluid and spinal fluid) or another tissue or sample taken from an individual having such a disorder, relative to the standard gene expression level, i.e., the expression level in healthy tissue or bodily fluid from an individual not having the disorder.
Enriched expression in infant and fetal brain and cerebellum indicates utility for the study and treatment of neurological, neurodevelopmental and neurodegenerative, sensory and cognitive disorders and neoplasms.
Moreover, the tissue distribution in infant and fetal brain and cerebellum indicates polynucleotides and polypeptides corresponding to this gene would be useful for the detection, treatment, and/or prevention of neurodegenerative disease states, behavioral disorders, or inflammatory conditions. Representative uses are described in the "Regeneration" and "Hyperproliferative Disorders" sections below, in Example 11, 15, and 18, and elsewhere herein. Briefly, the uses include, but are not limited to the detection, treatment, and/or prevention of Alzheimer's Disease, Parkinson's Disease, Huntington's Disease, Tourette Syndrome, meningitis, encephalitis, demyelinating diseases, peripheral neuropathies, neoplasia, trauma, congenital malformations, spinal cord injuries, ischemia and infarction, aneurysms, hemorrhages, schizophrenia, mania, dementia, paranoia, obsessive compulsive disorder, depression, panic disorder, learning disabilities, ALS, psychoses, autism, and altered behaviors, including disorders in feeding, sleep patterns, balance, and perception. In addition, elevated expression of this gene product in regions of the brain indicates it plays a role in normal neural function. Potentially, this gene product is involved in synapse formation, neurotransmission, learning, cognition, homeostasis, or neuronal differentiation or survival. Furthermore, the protein may also be used to determine biological activity, to raise antibodies, as tissue markers, to isolate cognate ligands or receptors, to identify agents that modulate their interactions, in addition to its use as a nutritional supplement. Protein, as well as, antibodies directed against the protein may show utility as a tumor marker and/or immunotherapy targets for the above listed tissues.
FEATURES OF PROTEIN ENCODED BY GENE NO: 15 This gene is expressed primarily in the following tissues/cDNA libraries:
Human Rhabdomyosarcoma and to a lesser extent in NCI CGAP_Kid3; Bone Marrow Stromal Cell, untreated; Soares melanocyte 2NbHM; Human endometrial stromal cells; Human umbilical vein endothelial cells, IL-4 induced; Primary Dendritic cells,frac 2; Human Microvascular Endothelial Cells, fract. A;
Soares senescent fibroblasts NbHSF; H. hypothalamus, frac A,re-excision;
Namalwa Cells; Human Fetal Liver- Enzyme subtraction; NCI CGAP Pr23; Human Colon, subtraction; NCI CGAP_GC2; Human Umbilical Vein Endothelial Cells, fract. A; Hep G2 Cells, PCR library; Smooth Muscle- HASTE normalized;
NCI CGAP Ut3; Human adult small intestine,re-excision; NCI CGAP-AAl;
Human Epididymus; Healing groin wound, 7.5 hours post incision; Human Adipose Tissue, re-excision; T-Cell PHA 16 hrs; NCI CGAP_Ewl; Human Uterine Cancer;
Human Umbilical Vein Endothelial Cells, uninduced; Liver, Hepatoma; Human Adipose; Spinal cord; Soares NSF F8 9W OT PA P S1; Hemangiopericytoma;
NTERA2, control; Hepatocellular Tumor, re-excision; NCI CGAP-Panl; Fetal Liver, subtraction II; 12 Week Old Early Stage Human; Smooth muscle, serum treated; Human Testes Tumor; Dendritic cells, pooled; Endothelial cells-control;
Smooth muscle,control; NCI CGAP-Kids; Human Bone Marrow, treated; T Cell helper I; Hodgkin's Lymphoma II; NCI CGAP-LuS; Human 8 Week Whole Embryo;
T cell helper II; Soares total fetus Nb2HF8 9w; Soares-pregnant uterus NbHPU;
Soares fetal liver spleen_1NFLS S1 and Soares fetal heart NbHHI9W.
Polynucleotides and polypeptides of the invention are useful as reagents for differential identification of the tissues) or cell types) present in a biological sample and for diagnosis of diseases and conditions which include but are not limited to:
immune system disorders. Similarly, polypeptides and antibodies directed to these polypeptides are useful in providing immunological probes for differential identification of the tissues) or cell type(s). .For a number of disorders of the above tissues or cells, particularly of the immune system , expression of this gene at significantly higher or lower levels may be routinely detected in certain tissues or cell types (e.g., cancerous and wounded tissues) or bodily fluids (e.g., serum, plasma, urine, synovial fluid and spinal fluid) or another tissue or sample taken from an individual having such a disorder, relative to the standard gene expression level, i.e., the expression level in healthy tissue or bodily fluid from an individual not having the disorder.
Elevated expression in tumor tissue indicates a role for this gene product in proliferation. In particular, high expression in rabdomyosarcoma indicates a possible utility of this gene product in the diagnosis and/or treatment of adult and pediatric soft tissue sarcomas.
The tissue distribution in immune tissue (e.g., dendritic cells, bone marrow) indicates the polynucleotides and polypeptides corresponding to this gene would be useful for the diagnosis and treatment of a variety of immune system disorders.
Representative uses are described in the "Immune Activity" and "Infectious Disease"
sections below, in Example 11, 13, 14, 16, 18, 19, 20, and 27, and elsewhere herein.
Briefly, the expression indicates a role in regulating the proliferation;
survival;
differentiation; and/or activation of hematopoietic cell lineages, including blood stem cells. Involvement in the regulation of cytokine production, antigen presentation, or other processes indicates a usefulness for treatment of cancer (e.g. by boosting immune responses). Expression in cells of lymphoid origin, indicates the natural gene product would be involved in immune functions. Therefore it would also be useful as an agent for immunological disorders including arthritis, asthma, immunodeficiency diseases such as AIDS, leukemia, rheumatoid arthritis, granulomatous disease, inflammatory bowel disease, sepsis, acne, neutropenia, neutrophilia, psoriasis, hypersensitivities, such as T-cell mediated cytotoxicity; immune reactions to transplanted organs and tissues, such as host-versus-graft and graft-versus-host diseases, or autoimmunity disorders, such as autoimmune infertility, tense tissue injury, demyelination, systemic lupus erythematosis, drug induced hemolytic anemia, rheumatoid arthritis, Sjogren's disease, and scleroderma. Moreover, the protein may represent a secreted factor that influences the differentiation or behavior of other blood cells, or that recruits hematopoietic cells to sites of injury. Thus, this gene product is thought to be useful in the expansion of stem cells and committed progenitors of various blood lineages, and in the differentiation and/or proliferation of various cell types. Furthermore, the protein may also be used to determine biological activity, raise antibodies, as tissue markers, to isolate cognate ligands or receptors; to identify agents that modulate their interactions, in addition to its use as a nutritional supplement. Protein, as well as, antibodies directed against the protein may show utility as a tumor marker and/or immunotherapy targets for the above listed tissues.
S FEATURES OF PROTEIN ENCODED BY GENE NO: 16 The computer algorithm BLASTX has been used to determine that the translation product of this gene shares sequence homology with, as a non-limiting example, the sequence accessible through the following database accession no.
gb~AAA30690.1 ~ (all information available through the recited accession number is incorporated herein by reference) which is described therein as "~ PDI
(E.C.S.3.4.1) [Bos taurus]" A partial alignment demonstrating the observed homology is shown immediately below.
1S >gb~AAA30690.1~ PDI (E.C.5.3.4.1) [BOS taurus] >pir~A26829~ISBOSS protein disulfide-isomerase (EC 5.3.4.1) precursor - bovine >sp~P05307~PDI_BOVIN PROTEIN DISULFIDE ISOMERASE PRECURSOR (PDI) (EC 5.3.4.1) (PROLYL 4- HYDROXYLASE BETA SUBUNIT) (CELLULAR THYROID
HORMONE BINDING PROTEIN) (P55).
2 0 Length = 510 Minus Strand HSPS:
Score = 338 (119.0 bits), Expect = 1.9e-28, P = 1.9e-28 ZS Identities = 83/257 (32~), Positives = 142/257 (55~), Frame = -2 Q: 1474 KSSDGPGAAQEPTWLTDVPAAMEFIAATEVAVIGFFQDLEIPAVPILHSMVQKFPGVSFG 1295 K GP A+ L+D AA + ++EVAVIGFF+D+E + + + FG
S: 132 KKRTGPAAST----LSDGAAAEALVESSEVAVIGFFKDMESDSAKQFFLAAEVIDDIPFG 187 Q: 1294 ISTDSEVLTHYNITGNTICLFRLWNEQLNLEDEDIESIDATKLSRFIEINSLHMVTEYN 1115 I+++S+V + Y + + + LF+ D + N E E + KL FI+ N L +V E+
S: 188 ITSNSDVFSKYQLDKDGWLFKKFDEGRNNFEGE----VTKEKLLDFIKHNQLPLVIEFT 243 3 S Q: 1114 PVTVIGLFNSVIQIHLLLIMNKASPEYEENMHRYQKAAKLFQGKILFILVDSGMKENGKV 935 T +F I+ H+LL + K+ +YE + ++KAA+ F+GKILFI +DS +N ++
S: 244 EQTAPKIFGGEIKTHILLFLPKSVSDYEGKLSNFKKAAESFKGKILFIFIDSDHTDNQRI 303 4O Q~ 934 ISFFKLKESQLPALXIYQTLDDEWDTL-PTA-EVSVEHVQNFCDGFLSGKLLKE--NRES 767 + FF LK+ + PA+ + TL++E P + E++ E + FC FL GK+ ++E
S: 304 LEFFGLKKEECPAVRLI-TLEEEMTKYKPESDELTAEKITEFCHRFLEGKIKPHLMSQEL 362 Q: 766 EGKTPKVEL*LLLGTTYGQVS 704 K + +L+G + +V+
4S S: 363 PDDWDKQPVKVLVGKNFEEVA 383 The segment of gb~AAA30690.1 ~ that is shown as "S" above is set out in the sequence listing as SEQ ID NO: 144. Based on the structural similarity these homologous polypeptides are expected to share at least some biological activities.

Such activities are known in the art, some of which are described elsewhere herein.
Assays for determining such activities are also known in the art, some of which have been described elsewhere herein. Preferred polypeptides of the invention comprise a polypeptide having the amino acid sequence set out in the sequence listing as SEQ ID
5 NO: 145 which corresponds to the Q sequence in the alignment shown above (gaps introduced in a sequence by the computer are, of course, removed).
The polypeptide of this gene has been determined to have a transmembrane domain at about amino acid position 144-160 of the amino acid sequence referenced in Table 1 for this gene. Moreover, a cytoplasmic tail encompassing amino acids 1 -10 143 of this protein has also been determined. Based upon these characteristics, it is believed that the protein product of this gene shares structural features to type II
membrane proteins.
The gene encoding the disclosed cDNA is believed to reside on chromosome 12. Accordingly, polynucleotides related to this invention are useful as a marker in 15 linkage analysis for chromosome 12.
This gene is expressed primarily in the following tissues/cDNA libraries:
Pancreas Tumor PCA4 Tu; Soares fetal liver spleen 1NFLS and to a lesser extent in Pancreas normal PCA4 No; Soares~arathyroid tumor NbHPA; Ovary, Cancer:
(4004562 B6) Papillary Serous Cystic Neoplasm, Low Malignant Pot;
20 Soares fetal lung NbHLI9W; NCI CGAP_GCB1; H. Kidney Medulla, subtracted;
HPAS (human pancreas, subtracted); NCI CGAP_Thyl; Breast, Cancer: (4004943 A5); Human Pancreas Tumor; NCI CGAP-CLLl;
Soares fetal liver spleen-1NFLS S1 and Soares NhHMPu S1.
Polynucleotides and polypeptides of the invention are useful as reagents for 25 differential identification of the tissues) or cell types) present in a biological sample and for diagnosis of diseases and conditions which include but are not limited to:
breast cancer, ovarian cancer, pancreatic cancer, kidney cancer, and cancer, in general. Similarly, polypeptides and antibodies directed to these polypeptides are useful in providing immunological probes for differential identification of the 30 tissues) or cell type(s). For a number of disorders of the above tissues or cells, particularly of the endocrine, exocrine, and reproductive systems, expression of this gene at significantly higher or lower levels may be routinely detected in certain tissues or cell types (e.g., cancerous and wounded tissues) or bodily fluids (e.g., serum, plasma, urine, synovial fluid and spinal fluid) or another tissue or sample taken from an individual having such a disorder, relative to the standard gene expression level, i.e., the expression level in healthy tissue or bodily fluid from an individual not having the disorder.
The enriched expression in pancreas and other tumors and homology to protein disulpide isomerases indicates that polynucleotides and polypeptides corresponding to this gene are useful for study and treatment of hormonal, metabolic and reproductive disorders and neoplasms.
FEATURES OF PROTEIN ENCODED BY GENE NO: 17 The computer algorithm BLASTX has been used to determine that the translation product of this gene shares sequence homology with, as a non-limiting example, the sequence accessible through the following database accession no.
emb~CAA59990.1 ~ (all information available through the recited accession number is incorporated herein by reference) which is described therein as "~ elastin like protein [Drosophila melanogaster]" A partial alignment demonstrating the observed homology is shown immediately below.
>emb~CAA59990.1~ elastin like protein [Drosophila melanogaster]
>sp~Q24333~Q24333 ELASTIN LIKE PROTEIN (FRAGMENT).
Length = 110 Plus Strand HSPS:
Score = 120 (42.2 bits), Expect = 1.0e-09, Sum P(2) = 1.0e-09 Identities = 23/23 (100$), Positives = 23/23 (100$), Frame = +2 Q: 62 WSSTAVAAALELVDPPGCRNSAR 130 WSSTAVAAALELVDPPGCRNSAR
S: 1 WSSTAVAAALELVDPPGCRNSAR 23 3 5 Score = 42 (14.8 bits), Expect = 1.0e-09, Sum P(2) = 1.0e-09 Identities = 15/38 (39$), Positives = 19/38 (50$), Frame = +1 Q: 262 QKLAGPLMPDVQGPWHPAHP-PIPSAALCLLWPHCLAAP 375 Q AG P V P +PA+P P A +L P +A P
4O S: 33 QLYAGYPYPGVYLPQYPAYPQPAQFPAYGVL-PGAVAQP 70 The segments of emb~CAA59990.1 ~ that are shown as "S" above are set out in the sequence listing as SEQ ID NO: 146 and SEQ ID NO: 148. Based on the structural similarity these homologous polypeptides are expected to share at least some biological activities. Such activities are known in the art, some of which are described elsewhere herein. Assays for determining such activities are also known in the art, some of which have been described elsewhere herein. Preferred polypeptides of the invention comprise a polypeptide having the amino acid sequence set out in the sequence listing as SEQ ID NO: 147 and/or SEQ ID NO: 149 which correspond to the Q sequences in the alignment shown above (gaps introduced in a sequence by the computer are, of course, removed).
This gene is expressed primarily in the following tissues/cDNA libraries:
NCI CGAP-GCB1; Soares fetal liver spleen 1NFLS and to a lesser extent in Mo7e Cell Line GM-CSF treated (lng/ml); NCI CGAP_Co3;
Soares~arathyroid_tumor NbHPA; Soares melanocyte 2NbHM;
Soares fetal lung NbHLI9W; NCI CGAP Kidl2; Human Adult Small Intestine;
Breast, Normal: (4005522B2); Human Pancreas Tumor; Normal colon;
NCI CGAP-Kid3; HUMAN B CELL LYMPHOMA; Bone Marrow Cell Line (RS4,11); Soares total fetus Nb2HF8 9w; Soares-fetal liver spleen 1NFLS S1;
Soares placenta Nb2HP; NCI CGAP_Ovl; Stratagene cat#937212 (1992); Larynx Tumour; Liver Normal MetSNo; NCI CGAP HN4; Liver HepG2 cell line.;
HTCDL1; NCI CGAP_Ov23; NCI CGAP-Thyl; Human Liver; Human Lung;
NCI CGAP Ut4; Hepatocellular Tumor,re-excision; Glioblastoma; LNCAP prostate cell line; Human endometr~ial stromal cells; Morton Fetal Cochlea; NCI CGAP
Alvl;
NCI CGAP_Ewl; HUMAN JURKAT MEMBRANE BOUND POLYSOMES;
Human Heart; Human Placenta (re-excision); Soares NSF F8 9W_OT PA P S1;
Human Thymus Stromal Cells; NCI CGAP_Panl; Human Substantia Nigra; Human Placenta; NCI CGAP-CoB; NCI CGAP GC4; Primary Dendritic cells,frac 2;
Pancreas normal PCA4 No; Bone marrow; human tonsils; Activated T-Cell (l2hs)/Thiouridine labelledEco; CD34 depleted Buffy Coat (Cord Blood), re-excision; Human Osteoclastoma; Human Microvascular Endothelial Cells, fract.
A;
Soares senescent fibroblasts NbHSF; NCI CGAP_Kids; NCI CGAP-Brn23;
Soares multiple sclerosis 2NbHMSP; Human Bone Marrow, treated; T Cell helper I; Human fetal heart, Lambda ZAP Express; Hodgkin's Lymphoma II;
NCI CGAP_LuS; Nine Week Old Early Stage Human; Colon Tumor II; Human Cerebellum; Colon Normal III; Soares-pregnant uterus NbHPU;
Soares fetal heart NbHHI9W; Soares testis NHT and Primary Dendritic Cells, lib Polynucleotides and polypeptides of the invention are useful as reagents for .
differential identification of the tissues) or cell types) present in a biological sample and for diagnosis of diseases and conditions which include but are not limited to:
immune disorders. Similarly, polypeptides and antibodies directed to these polypeptides are useful in providing immunological probes for differential identification of the tissues) or cell type(s). For a number of disorders of the above tissues or cells, particularly of the immune system, expression of this gene at significantly higher or lower levels may be routinely detected in certain tissues or cell types (e.g., cancerous and wounded tissues) or bodily fluids (e.g., serum, plasma, urine, synovial fluid and spinal fluid) or another tissue or sample taken from an individual having such a disorder, relative to the standard gene expression level, i.e., the expression level in healthy tissue or bodily fluid from an individual not having the disorder.
Enriched expression in fetal liver and spleen and some endocrine organs indicates utility in the study and treatment of hematopoietic, immune and inflammatory, hormonal and metabolic disorders and neoplasms.
The tissue distribution in immune tissue (e.g., bone marrow) indicates the polynucleotides and polypeptides corresponding to this gene would be useful for the diagnosis and treatment of a variety of immune system disorders.
Representative uses are described in the "Immune Activity" and "Infectious Disease" sections below, in Example 11, 13, 14, 16, 18, 19, 20, and 27, and elsewhere herein.-Briefly, the expression indicates a role in regulating the proliferation; survival;
differentiation;
and/or activation of hematopoietic cell lineages, including blood stem cells.
Involvement in the regulation of cytokine production, antigen presentation, or other processes indicates a usefulness for treatment of cancer (e.g. by boosting immune responses). Expression in cells of lymphoid origin, indicates thewatural gene product would be involved in immune functions. Therefore it would also be useful as an agent for immunological disorders including arthritis, asthma, immunodeficiency diseases such as AIDS, leukemia, rheumatoid arthritis, granulomatous disease, inflammatory bowel disease, sepsis, acne, neutropenia, neutrophilia, psoriasis, hypersensitivities, such as T-cell mediated cytotoxicity; immune reactions to transplanted organs and tissues, such as host-versus-graft and graft-versus-host diseases, or autoimmunity disorders, such as autoimmune infertility, Tense tissue injury, demyelination, systemic lupus erythematosis, drug induced hemolytic anemia, rheumatoid arthritis, Sjogren's disease, and scleroderma. Moreover, the protein may represent a secreted factor that influences the differentiation or behavior of other blood cells, or that recruits hematopoietic cells to sites of injury. Thus, this gene product is thought to be useful in the expansion of stem cells and committed progenitors of various blood lineages, and in the differentiation and/or proliferation of various cell types.
Furthermore, the protein may also be used to determine biological activity, raise antibodies, as tissue markers, to isolate cognate ligands or receptors, to identify agents that modulate their interactions, in addition to its use as a nutritional supplement. Protein, as well as, antibodies directed against the protein may show utility as a tumor marker and/or immunotherapy targets for the above listed tissues.
FEATURES OF PROTEIN ENCODED BY GENE NO: 18 The computer algorithm BLASTX has been used to determine that the translation product of this gene shares sequence homology with, as a non-limiting example, the sequence accessible through the following database accession no.
emb~CAA21961.1 ~ (all information available through the recited accession number is incorporated herein by reference) which is described therein as "~ (AL033406) dna-repair protein radl8 [Schizosaccharomyces pombe]" A partial alignment demonstrating the observed homology is shown immediately below.
>emb ~ CAA21961 .1 ~ (AL033406) dna repair protein radl8 [Schizosaccharomyces pombe] >emb~CAA56900.1~ radl8 [Schizosaccharomyces pombe]
>sp~P53692~RA18_SCHPO DNA REPAIR PROTEIN RAD18.
>sp~CAA21961~CAA21961 Dna repair protein radl8.
Length = 1140 S
Plus Strand HSPs:
Score = 404 (142.2 bits), Expect = 1.3e-33, P = 1.3e-33 Identities = 116/389 (29~), Positives = 195/389 (50~), Frame = +3 Q: 105 KHNEELL-KRCQLHYKELKMKIRK-NISEIRELENIEEHQSVDIATLEDEAQENKSKMKM 278 K +E+LL ++ + K + +K R+ N E +EL ++ + I TLE E + +++
S: 747 KRDEQLLVEKIEGIKKRILLKRREVNSLESQELSVLDTEK---IQTLERRISETEKELES 803 1O Q: 279 VEEHMEQQKENMEH-LKSLKIEAENKYDAIKFKINQLSELADPLKDELN-LADSEVDNQK452 ++ K N EH ++ + + + KI ++ L+ EL+ L D +
+++

S: 804 YAGQLQDAK-NEEHRIRDNQRPVIEEIRIYREKIQTETQRLSSLQTELSRLRDEKRNSEV862 Q. 453 RGKRHYEEKQKEHLDTLNXXXXXXXXXXXXXXXXXSQARQICPERIEVEKSASILDKEIN632 IS +RH + + + L ++A C ER+ V+ S + LD EI

S: 863 DIERH-RQTVESCTNILREKEAKKVQCAQWADYTAKANTRC-ERVPVQLSPAELDNEIE920 Q: 633 RLRQKIQAEHASHG-DREEIMRQYQEARETYLDLDSKVRTLKKFIKLLGEIMEHRFKTYQ809 RL+ +I G E+ Y A+E + V L + ++ L E + R +
+

2O S: 921 RLQMQIAEWRNRTGVSVEQAAEDYLNAKEKHDQAKVLVARLTQLLQALEETLRRRNEMWT980 Q: 810 QFRRCLTLRCKLYFDNLLSQRAYCGKMNFDHKNETLSISVQPGEGNKA-AFN-------D965 +FR+ +TLR K F+ LSQR + GK+ H+ E L V P N
A A N

S: 981 KFRKLITLRTKELFELYLSQRNFTGKLVIKHQEEFLEPRWPANRNLATAHNRHEKSKVS1040 Q: 966 MRALSGGERSFSTVCFILSLWSIAESPFRCLDEFDWMDMVNRRIAMDLILKMADSQRFR 1145 ++ LSGGE+SF+T+C +LS+W P RCLDEFDV+MD VNR +++ +++ A +
S: 1041 VQGLSGGEKSFATICMLLSIWEAMSCPLRCLDEFDVFMDAVNRLVSIKMMVDSAKDSSDK 1100 3 O Q: 1146 QFILLTPQSMSSLPSSKLIRILRMSDPERGQTTLP 1250 QFI +TPQ M + K + + R+SDP + LP
S: 1101 QFIFITPQDMGQIGLDKDVWFRLSDPWSSSALP 1135 The segment of emb~CAA21961.1 ~ that is shown as "S" above is set out in the 35 sequence listing as SEQ ID NO: 151. Based on the structural similarity these homologous polypeptides are expected to share at least some biological activities.
Such activities are known in the art, some of which are described elsewhere herein.
Assays for determining such activities are also known in the art, some of which have been described elsewhere herein. Preferred polypeptides of the invention comprise a 40 polypeptide having the amino acid sequence set out in the sequence listing as SEQ ID
NO: 152 which corresponds to the Q sequence in the alignment shown above (gaps introduced in a sequence by the computer are, of course, removed).
In specific embodiments, polypeptides of the invention comprise, or alternatively consists of, the following amino acid sequence:

MKIRKNISEIRELENIEEHQSVDIATLEDEAQENKSKMKMVEEHMEQQKENM

EEKQKEI-iLDTLNKKKRELDMKEKELEEKMSQARQICPERIEVEKSASILDKEI
NRLRQKIQAEHASHGDREEIMRQYQEARETYLDLDSKVRTLKKFIKLLGEIME

HRFKTYQQFRRCLTLRCKLYFDNLLSQRAYCGKMNFDHKNETLSISVQPGEG
NKAAFNDMRALSGGERSFSTVCFILSLWSIAESPFRCLDEFDVYMDMVNRRI
AMDLILKMADSQRFRQFILLTPQSMSSLPSSKLIRILRMSDPERGQTTLPFRPV
TQEEDDDQR (SEQ ID NO: 150). Moreover, fragments and variants of these polypeptides (such as, for example, fragments as described herein, polypeptides at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% identical to these polypeptides and polypeptides encoded by the polynucleotide which hybridizes, under stringent conditions, to the polynucleotide encoding these polypeptides ) are encompassed by the invention. Antibodies that bind polypeptides of the invention are also encompassed by the invention. Polynucleotides encoding these polypeptides are also encompassed by the invention.
The gene encoding the disclosed cDNA is believed to reside on chromosome 2. Accordingly, polynucleotides related to this invention are useful as a marker in linkage analysis for chromosome 2.
This gene is expressed primarily in the following tissues/cDNA libraries:
Human Eosinophils and to a lesser extent in Spleen, Chronic lymphocytic leukemia;
NCI CGAP-GCBl; Healing groin wound - zero hr post-incision (control); Human adult (K.Okubo); Stromal cell TF274; Soares multiple sclerosis 2NbHMSP; T cell helper II; Soares NFL T GBC S1; Soares fetal liver spleen 1NFLS; Stomach Tumour; Stomach cancer (human),re-excision; NCI CGAP-Ov2; NCI CGAP-Kid6;
12 Week Old Early Stage Human, II; Stratagene HeLa cell s3 937216; Human Placenta (re-excision); NTERA2, control; NCI CGAP-GC4; Human Osteoclastoma;
NCI CGAP Kids; Hodgkin's Lymphoma II; Colon Tumor II;
Soares total fetus Nb2HF8 9w; Soares fetal liver spleen_1NFLS S1;
Soares NhHMPu S1 and Soares testis NHT.
Polynucleotides and polypeptides of the invention-are useful as reagents for differential identification of the tissues) or cell types) present in a biological sample and for diagnosis of diseases and conditions which include but are not limited to:
hematopoietic related disorders and leukemia. Similarly, polypeptides and antibodies directed to these polypeptides are useful in providing immunological probes for differential identification of the tissues) or cell type(s). For a number of disorders of the above tissues or cells, particularly of the immune system, expression of this gene at significantly higher or lower levels may be routinely detected in certain tissues or cell types (e.g., cancerous and wounded tissues) or bodily fluids (e.g., serum, plasma, urine, synovial fluid and spinal fluid) or another tissue or sample taken from an individual having such a disorder, relative to the standard gene expression level, i.e., the expression level in healthy tissue or bodily fluid from an individual not having the disorder.
The tissue distribution indicates that polynucleotides and polypeptides corresponding to this gene would be useful for the treatment and diagnosis of hematopoietic related disorders such as anemia, pancytopenia, leukopenia, thrombocytopenia or leukemia since stromal cells are important in the production of cells of hematopoietic lineages. Representative uses are described in the "Immune Activity" and "Infectious Disease" sections below, in Example 11, 13, 14, 16, 18, 19, 20, and 27, and elsewhere herein. Briefly, the uses include bone marrow cell ex-vivo culture, bone marrow transplantation, bone marrow reconstitution, radiotherapy or chemotherapy of neoplasia. The gene product may also be involved in lymphopoiesis, therefore, it can be used in immune disorders such as infection, inflammation, allergy, immunodeficiency etc. In addition, this gene product may have commercial utility in the expansion of stem cells and committed progenitors of various blood lineages, and in the differentiation and/or proliferation of various cell types.
Furthermore, the protein may also be used to determine biological activity, to raise antibodies, as tissue markers, to isolate cognate ligands or receptors, to identify agents that modulate their interactions, in addition to its use as a nutritional supplement. Protein, as well as, antibodies directed against the protein may show utility as a tumor marker and/or immunotherapy targets for the above listed tissues.
FEATURES OF PROTEIN ENCODED BY GENE NO: 19 Preferred polypeptides of the invention comprise a polypeptide having the amino acid sequence set out in the sequence listing as SEQ ID NO: 153 and/or SEQ
ID NO: 154. Polynucleotides encoding these polypeptides are also provided.

The polypeptide of this gene has been determined to have transmembrane domains at about amino acid positions 401-417, 446-465, 266-282, 340-356, 369-of the amino acid sequence referenced in Table 1 for this gene. Based upon these characteristics, it is believed that the protein product of this gene shares structural features to type IIIa membrane proteins.
This gene is expressed primarily in the following tissues/cDNA libraries:
Seven Trans Membrane Receptor Family and to a lesser extent in Soares_fetal liver spleen 1NFLS_S1; Colon Carcinoma; Soares NhHMPu_S1;
Synovial IL-1/TNF stimulated; B-cells (unstimulated); NCI CGAP_Panl;
NCI CGAP-Kidl l; NCI CGAP-CoB; NCI CGAP Kid3; Human Bone Marrow, treated; T Cell helper I; Activated T-cell(12h)/Thiouridine-re-excision; T
cell helper II; Colon Normal III; Primary Dendritic Cells, lib 1; Soares fetal liver spleen 1NFLS;
Human Pituitary, subtracted VII; CD34+ cell, I, frac II; NCI CGAP_Pr9; Human (Caco-2) cell line, adenocarcinoma, colon, remake; NCI CGAP_GCS; Human Aortic Endothelium; Fetal Heart, re-excision; Human Neutrophils, Activated, re-excision; H.
cerebellum, Enzyme subtracted; Human endometrial stromal cells-treated with estradiol; Healing groin wound, 7.5 hours post incision; Human Adipose Tissue, re-excision; Synovial hypoxia; Human Pituitary, subt IX; TF-1 Cell Line GM-CSF
Treated; NCI CGAP_Pr2; NCI CGAP Utl; Monocyte activated, re-excision;
Apoptotic T-cell; Human Adult Testes, Large Inserts, Reexcision; Ovary, Cancer (9809C332): Poorly differentiated adenocarcinoma; Ulcerative Colitis;
NCI CGAP CLL1; Human Adrenal Gland Tumor; Human adult testis, large inserts;
Human Liver, normal; Fetal Heart; H Macrophage (GM-CSF treated), re-excision;
Dendritic cells, pooled; Human Fetal Lung III; Pancreas normal PCA4 No;
NCI CGAP-Brn23; Bone Marrow Cell Line (RS4,11); Hodgkin's Lymphoma II;
NCI CGAP_LuS; Soares melanocyte 2NbHM; Soares testis NHT and Soares infant brain 1NIB.
Polynucleotides and polypeptides of the invention are useful as reagents for differential identification of the tissues) or cell types) present in a biological sample and for diagnosis of diseases and conditions which include but are not limited to:
immune system disorders. Similarly, polypeptides and antibodies directed to these polypeptides are useful in providing immunological probes for differential identification of the tissues) or cell type(s). For a number of disorders of the above tissues or cells, particularly of the immune system, expression of this gene at significantly higher or lower levels may be routinely detected in certain tissues or cell types (e.g., cancerous and wounded tissues) or bodily fluids (e.g., serum, plasma, urine, synovial fluid and spinal fluid) or another tissue or sample taken from an individual having such a disorder, relative to the standard gene expression level, i.e., the expression level in healthy tissue or bodily fluid from an individual not having the disorder.
The tissue distribution in hematopoietic cells and cancers indicates that polynucleotides and polypeptides of the invention useful for the diagnosis and treatment of a variety of immune system disorders. Representative uses are described in the "Immune Activity" and "Infectious Disease" sections below, in Example 11, 13, 14, 16, 18, 19, 20, and 27, and elsewhere herein. Briefly, the expression indicates a role in regulating the proliferation; survival; differentiation; and/or activation of hematopoietic cell lineages, including blood stem cells. Involvement in the regulation of cytokine production, antigen presentation, or other processes indicates a usefulness for treatment of cancer (e.g. by boosting immune responses). Expression in cells of lymphoid origin, indicates the natural gene product would be involved in immune functions. Therefore it would also be useful as an agent for immunological disorders including arthritis, asthma, immunodeficiency diseases such as AIDS, leukemia, rheumatoid arthritis, granulomatous disease, inflammatory bowel disease, sepsis, acne, neutropenia, neutrophilia, psoriasis, hypersensitivities, such as T-cell mediated cytotoxicity; immune reactions to transplanted organs and tissues, such as host-versus-graft and graft-versus-host diseases, or autoimmunity disorders, such as autoimmune infertility, lense tissue injury, demyelination, systemic lupus erythematosis, drug induced hemolytic anemia, rheumatoid arthritis, Sjogren's disease, and scleroderma. Moreover, the protein may represent a secreted factor that influences the differentiation or behavior of other blood cells, or that recruits hematopoietic cells to sites of injury. Thus, this gene product is thought to be useful in the expansion of stem cells and committed progenitors of various blood lineages, and in the differentiation and/or proliferation of various cell types.
Furthermore, the protein may also be used to determine biological activity, raise antibodies, as tissue SS
markers, to isolate cognate ligands or receptors, to identify agents that modulate their interactions, in addition to its use as a nutritional supplement. Protein, as well as, antibodies directed against the protein may show utility as a tumor marker and/or immunotherapy targets for the above listed tissues.

00 ~- ~n t~ ~t d_w D ~_n ~_ ~ O

O

.~ v~ N ~ ~ ~n ~ N

.b 4~ p .~, N y 00 ~O ~D ~O V~ ~n I~
y., N N N

N r",,r., N N N N

0 ~ a~ ~ ~ '~ ~n r,, ,~ ~ ~ ,_, N N N N

cue, bn O ~ N

w a ~n ' w o "' o N ~ a\

P-~ ~ ~ N ~ M V~'1Vl r~",i C%~

H
'~ ~ ~ ~ o N o, E"' ~ O N ~ ,-, ~ ~ M Q1 ~, v~
4-i N o0 l~ O I~ O 01 O U ~ 01 v0 O ~ N ~ N ~ O

w ~ C 0o O

O p ~ M ,-~ ,~ ~ ~ .~ .~ .~ M ,-.
( N
;
~

n ~

-~ M d' ~ M M 01 O
~

O ~ ~ O ~ O~ ~ N ~ M O
O ~ ~
/~
Z

C .--~~ N N N .~ ..-~N

O N ~ N M M

M .~ M M M ~ M M .-i ~ O O O O O O O

V N N ~" ss. ~" ~'"'Q" u. ~'' N

~ a Q ~ Q ~

. ~ , . .

" -d o o o o o o o o o o a, Q, o, ~ o~ a, ~ o, a, o, ~ ~ ~ ~ ~ ~ ~ ~

~ a~
a\ o, av ov o~ w ~ a\ w ~
o~ ov ~o . . . . . . .
p. N Q' ~o ~o ~ ~ ~o ~ ~o Q' Q' N ~ N ~ ~' ~' Q' Q' d Q' N N
N N N N N N N

w w w W W w w W w w O [~ [~ [~ E-~ (-i H [~ f-~ [~ (~
~ O O O O O O O O O O

z a. a.~ w a.~ a.~ a.~ w w a.~ a, .~ ,~ ,~ r., ,~ .~ r. .~ ,~ ,.., tn V7 ~ ~ ~ ~ M M M

a a W w w w W w w z v~ a ~ a Ao ~ 3 ~ 3 ~
U

~ x x x x x x x x x N N N N M M M
~z M N O ~ ~O ~ 01 ~

N M ~ ~ N ~ M N

'b 4a _ l~ ~ ~ O O ~ ~--~ l~
'n p j .~

N M M N N N N N N

~ P~

~O ,_, iM M Q\ Q1 ~, O O ,_, ~O

a w N O M .~ ~t ~ N ~D I~ M

z~

pw.~nci; ~ M ~ ~ ~ o N ~ ~ N
~

a" N
~r, H
z~ ~b N ~ ~ ~, ~ ~ ~ N

p ~ p ~ ,-, M p~ ,-, ~ O

U

p O~ oo N ~ ~ M

p O a~ ~ M M M N M ~ ~n ~ o0 cn U ~ .~ ,-. ,~ , N ,~ .-, zo o~

_ ~ .-.
i U ~

n 00 ~n o 0 ~ ~ a, N ~n ~ ~ ~ ~ ~ t~
E

r~ ~ M ~ ~ ~ ~ ~ o0 r~' /~

C N N

E~ a ~ ~D l~ ~ 00 l0 Ov O I~ ,~ N o0 M M ~ M ,~ M d' ~ ~ O O O

U N N N N N N N ~ ~ ~ N

y y ~ ~ -~ ~ Q., 0 o o o o o o o o o o 0, a, Q, a, o, o, o, o, o, a, o, ~ ~ ~ ~ ~ ~ ~ ~ ~ ~ ~
a, a1 o, o~ a1 ov o, av ~ a, a~

00 oo oo oo oo oo oo oo oo oo oo o ~ ~ ~ ~ ~ ~ ~ ~ ~ ~ ~ ~
. . . . . . . . .
~o ~ ~ ~ ~ ~o ~ ~o ~o [-~ Q.N~ Q'N Q'N Q ~N ~N Q Q ~'N ~'N ~'N Q
'N 'N 'N 'N

o E-~ H ~ H E-~ ~ ~ E-~ H E-~ ~
o o H o o H E--~o o o H
o o o o o ~ N ~ ~ ~ ~ U

~ C7 ~ ~

_ ~ ~ ~' ~' x x x x x x U ~

x x x x x t~ a\ W n ~n o0 O O

o0 ~ W d' N N

4, ~ .~", N N' ~ N N N N

d N N N

N

4~ ~ ,~ r-r Q ~ ~ ~ ~ ~t ~ N N N N

o 0 0 0 ~

_ N ~ N c' ~ ~ ~ r' ~
~

, ~
a N ~ ~ N

~ U

zwo~ ~ ~ ~ o M

~n N ~ oo ~n ~ t~ ~ tw o ov a~

z o N ~ ~ ~ ,~ ~ ~ ,~ ~
o ~

_ N N

E-~ . M O ~ ~ l~ M ~O M .--~N

z ' N i o ~ ~ ~ ~ 0 0 ~, N ~

M o, ~ ~n o vo t~ .~ o0 0, o ~t ~ ~ ~ N ~ ~ N ~ ~' v~

O~

' N N N N N N N N N N N
_.
.., .'.,~ ~ .'.,.'.,~ ... .'., t~ r~ ~ ~ s~ s~ ~ t~

-d o o o o o o o o o o o w Q, o, o, o, ~ a, a, Q, ov , U ~ ~ ~ ~ ~ ~ ~ ~ ~ ~ ~ ~ ~ ~
~ ~ ~ ~ ~ ~ ~ ~ ~ ~ o, U oo oo oo oo oo oo oo oo oo oo oo ~ ~ ~ ~ ~ ~ ~ ~ ~ ~ ~ .
. . . . . . . . . . ~o ~o ~o ~o ~o ~o ~o ~o ~o ~o ~o [-, Q.N ~'N ~'N dN ~'N ~'N ~'N dN dN ~'N dN dN
~

H H H E-~ H H H E-~ H E-~ H
o o o o o o o o o o o ~ d w r- w a, a1 w d d d ~., ~ ~

o _ ~ x O
~U

x x x ~ x ~ ~ ~

a~

00 a, a\ a, o 0 0 ,~ .~ ,~ ,-, ~ M ~ ~ O~

O 0 ~ I~

b 4, N N N O O N N N N vp U

N

~O~~ N N ~ N N ON N ~ ~ M

a dA N

~~r~o~. ~ o o ~ o o ~

~

~, z M

U 00 00 '"~ N ~ ~ ~ 01 ~ M
y.~,., p".i M N ~ M ~ ~D ~ .-r ~ V~
~ ~

'n (%~

H
zo~b ci' M N l~ ~ ~ ~n ~ o0 a1 ~ 00 ' N ~ ' ' h ~ ~ 0 ~

U o 0 N o 0 zo o~

_ .~ ~ ~, ~ ~ o ~ ~ M
U ~ N

v7 N I~ M ~ V7 ~ 00 ~ V7 00 z0 ~ N ~ ~ ~ ~ ~ O ~ O

~O o o C/~ .~ .-a ,~ O N N o N ~ N M M ~t ~ ~ ~ t~ v~

N ~1 N N ~1 v7 N V1 ~n V7 N

O

' O O O O O O

N N N N

o o o o o o o o o o o ~ ~ o~ o, ~ ov o~ o, o, o, w ~ ~ ~ ~ ~ ~ ~ ~ ~ ~ ~
Q, o, a, a~ a\ o, ~ o, v~ o~ a~

U oo oo oo oo oo oo oo oo oo oo oo s ~ ~ ~ ~ ~ ~ ~ ~ ~ ~ .
. . . . . . . . . . ~
~o ~o ~ ~o ~ ~ ~o ~ ~ ~o a. N d d d d d d d d d d d N N N N N N N N N N N

d ~ o ~ H E-~ E- H H H H H H H H
o o o o o o o o o o o ~_ M M M N N N N

a a a a w w w ~ ~ x x x x x a~
N N N M M M

O V7 tn M N D1 00 00 M r ~ M N O 0 O

~ ~ 1 ~
O

M N N -~ ~ N
.

b 4a _ o0 0o N N r r M
N v N N N

,~ ,.--.~M M N N M N

N

_ _ O ~ ~ '-' ~ ~ M M '~ '~ N N M N

M ~ ~ N V~ \O l~ M 00 ~. 01 z N

~ O ~ ~ M ~ M ~' ~ N

N N ,~ .~ o ~ o ~ p~ M
O O
N M

H
z p ~ b ~ W O 0M0 "~ ~O N

N ,~ ~ N ~ p~ M

N
z ~ ~ ~ ~ o ~ g o o ~ ow n ov r ~

c~ U .-~ ,~ ~ N ,~

r~ ~Or" V1 l0 M
p~ .
z o _O ~ .~ .-. ,~ r .~ M .-.
U ~ ~n o, ~ O 0 O ~ ~n ~ N ~ O ~ ~ 0 O ~ ~ ~ 0 z ~ r .--~.~ .~ ,-, ,-. N .~

N ~ N ~ N

~D ~ N ~

N ~ ~ ~ V
, C ~ C
r-, Q.. t1.

'd O O O O O O O O O O O
p~ p~ O~ O1 Q\ p~ p~ ~ O~ Q\ O~

U y ~ ~ r r r r r r r r r r r ~ ~ ~ ~ ~ ~ ~ ~ ~ ~ ~
~ ~ ~ ~ ~ ~ ~ ~ ~

o ~ 00 oo oo oo oo oo oo oo oo oo oo ~ . . . . ~
. ~o ~o ~o ~ .
~o ~o [~ ~..N ~'N ~'N ~N a'N ~N dN ~'N dN dN ~N dN
~

dr~zr~ ~o ~o ~o ~o ~o ~o ~o ~o ~o ~o ~o '-' ~' 00 00 00 00 M M

O O O

~ a a U U

x x x x x x x x x x x Tw o vo r r r r o0 00 0~ ov O "' b o ~

. o N

C/y r~

0 ~ N -a w , ~a~ o ~

z H

z~ ~ ~~ N
.

H
zoo ~

U

~ ~+-~ ~
a"

a~

~ 4, ~ a"

Uv~

r.

H ~

~,z~

za~zx > ~

~.

U ~

o c o d ~' ~o ~

~

z d z ~ _o UU

0 vv z Table 1 summarizes the information corresponding to each "Gene No." described above. The nucleotide sequence identified as "NT SEQ ID NO:X" was assembled from partially homologous ("overlapping") sequences obtained from the "cDNA
clone ID" identified in Table 1 and, in some cases, from additional related DNA
clones. The overlapping sequences were assembled into a single contiguous sequence of high redundancy (usually three to five overlapping sequences at each nucleotide position), resulting in a final sequence identified as SEQ >D NO:X.
The cDNA Clone m was deposited on the date and given the corresponding deposit number listed in "ATCC Deposit No:Z and Date." Some of the deposits contain multiple different clones corresponding to the same gene. "Vector"
refers to the type of vector contained in the cDNA Clone ID.
"Total NT Seq." refers to the total number of nucleotides in the contig identified by "Gene No." The deposited clone may contain all or most of these sequences, reflected by the nucleotide position indicated as "5' NT of Clone Seq."
and the "3' NT of Clone Seq." of SEQ ID NO:X. The nucleotide position of SEQ
ID
NO:X of the putative start codon (methionine) is identified as "5' NT of Start Codon."
Similarly , the nucleotide position of SEQ ID NO:X of the predicted signal sequence is identified as "5' NT of First AA of Signal Pep."
The translated amino acid sequence, beginning with the methionine, is identified as "AA SEQ ID NO:Y," although other reading frames can also be easily translated using known molecular biology techniques. The polypeptides produced by these alternative open reading frames are specifically contemplated by the present invention.
The first and last amino acid position of SEQ ID NO:Y of the predicted signal peptide is identified as "First AA of Sig Pep" and "Last AA of Sig Pep." The predicted first amino acid position of SEQ B7 NO:Y of the secreted portion is identified as "Predicted First AA of Secreted Portion." Finally, the amino acid position of SEQ ID NO:Y of the last amino acid in the open reading frame is identified as "Last AA of ORF."
SEQ ID NO:X (where X may be any of the polynucleotide sequences disclosed in the sequence listing) and the translated SEQ ID NO:Y (where Y may be any of the polypeptide sequences disclosed in the sequence listing) are sufficiently accurate and otherwise suitable for a variety of uses well known in the art and described further below. For instance, SEQ ID NO:X is useful for designing nucleic acid hybridization probes that will detect nucleic acid sequences contained in SEQ ID
NO:X or the cDNA contained in the deposited clone. These probes will also hybridize to nucleic acid molecules in biological samples, thereby enabling a variety of forensic and diagnostic methods of the invention. Similarly, polypeptides identified from SEQ ID NO:Y may be used, for example, to generate antibodies which bind specifically to proteins containing the polypeptides and the secreted proteins encoded by the cDNA clones identified in Table 1.
Nevertheless, DNA sequences generated by sequencing reactions can contain sequencing errors. The errors exist as misidentified nucleotides, or as insertions or deletions of nucleotides in the generated DNA sequence. The erroneously inserted or deleted nucleotides cause frame shifts in the reading frames of the predicted amino acid sequence. In these cases, the predicted amino acid sequence diverges from the actual amino acid sequence, even though the generated DNA sequence may be greater than 99.9% identical to the actual DNA sequence (for example, one base insertion or deletion in an open reading frame of over 1000 bases).
Accordingly, for those applications requiring precision in the nucleotide sequence or the amino acid sequence, the present invention provides not only the generated nucleotide sequence identified as SEQ ID NO:X and the predicted translated amino acid sequence identified as SEQ ID NO:Y, but also a sample of plasmid DNA containing a human cDNA of the invention deposited with the ATCC, as set forth in Table 1. The nucleotide sequence of each deposited clone can readily be determined by sequencing the deposited clone in accordance with known methods.
The predicted amino acid sequence can then be verified from such deposits.
Moreover, the amino acid sequence of the protein encoded by a particular clone can also be directly determined by peptide sequencing or by expressing the protein in a suitable host cell containing the deposited human cDNA, collecting the protein, and determining its sequence.
The present invention also relates to the genes corresponding to SEQ ID
NO:X, SEQ ID NO:Y, or the deposited clone. The corresponding gene can be isolated in accordance with known methods using the sequence information disclosed, herein. Such methods include preparing probes or primers from the disclosed sequence and identifying or amplifying the corresponding gene from appropriate sources of genomic material.
Also provided in the present invention are allelic variants, orthologs, and/or species homologs. Procedures known in the art can be used to obtain full-length genes, allelic variants, splice variants, full-length coding portions, orthologs, and/or species homologs of genes corresponding to SEQ ID NO:X, SEQ ID NO:Y, or a deposited clone, using information from the sequences disclosed herein or the clones deposited with the ATCC. For example, allelic variants and/or species homologs may be isolated and identified by making suitable probes or primers from the sequences provided herein and screening a suitable nucleic acid source for allelic variants and/or the desired homologue.
Table 2 provides preferred epitopes contained in certain embodiments of the invention and polynucleotide sequences that may be disclaimed according to certain embodiments of the invention. The first column refers to each "Gene No."
described above in Table 1. The second column provides the sequence identifier, "NT SEQ
)D
NO:X", for polynucleotide sequences disclosed in Table 1. The third column provides the sequence identifier, "AA SEQ ID NO:Y", for polypeptide sequences disclosed in Table 1. The fourth column provides a unique integer "ntA" where "ntA" is any integer between 1 and the final nucleotide minus 15 of SEQ >D NO:X, and the fifth column provides a unique integer "ntB" where "ntB" is any integer between 15 and the final nucleotide of SEQ ID NO:X, where both ntA and ntB correspond to the positions of nucleotide residues shown in SEQ ID NO:X, and where ntB is greater than or equal to a + 14. For each of the polynucleotides shown as SEQ ID NO:X, the uniquely defined integers can be substituted into the general formula of a-b, and used to describe polynucleotides which may be preferably excluded from the invention.
Column 6 lists residues comprising predicted epitopes contained in the polypeptides encoded by each of the preferred ORFs (SEQ ID NO:Y). Identification of potential immunogenic regions was performed according to the method of Jameson and Wolf ((1988) CABIOS, 4; 181-186); specifically, the-Genetics Computer~Group (GCG) implementation of this algorithm, embodied in the program PEPTIDESTRUCTURE

(Wisconsin Package v10.0, Genetics Computer Group (GCG), Madison, Wisc.). This method returns a measure of the probability that a given residue is found on the surface of the protein. Regions where the antigenic index score is greater than 0.9 over at least 6 amino acids are indicated in Table 2 as "Preferred Epitopes".
5 Polypeptides of the invention may possess one, two, three, four, five or more antigenic epitopes comprising residues described in Table 2. It will be appreciated that depending on the analytical criteria used to predict antigenic determinants, the exact address of the determinant may vary slightly.
10 Table 3 summarizes the expression profile of polynucleotides corresponding to the clones disclosed in Table 1. The first column provides a unique clone identifier, "Clone ID", for a cDNA clone related to each contig sequence disclosed in Table 1. Column 2, "Library Codes" shows the expression profile of tissue and/or cell line libraries which express the polynucleotides of the invention. Each Library Code 15 in column 2 represents a tissue/cell source identifier code corresponding to the Library Code and Library description provided in Table 4. Expression of these polynucleotides was not observed in the other tissues and/or cell libraries tested. One of skill in the art could routinely use this information to identify tissues which show a predominant expression pattern of the corresponding polynucleotide of the invention 20 or to identify polynucleotides which show predominant and/or specific tissue expression.
Table 4 provides a key to the Library Code disclosed in Table 3. Column 1 provides the Library Code disclosed in Table 3, column 2. Column 2 provides a description of the tissue or cell source from which the corresponding library was 25 derived. Library codes corresponding to diseased Tissues are indicated in column 3 with the word "disease".

Table 2 Gene NT SEQ AA SEQ nt A nt B Preferred Epitopes ID ID

# NO: X NO: Y

1 11 66 1 - 15 - Lys-10 to Phe-19 Lys-34 to His-39 Val-50 to Leu-58 Asn-60 to Asp-76 Val-118 to Pro-143 His-154 to Gly-164 Pro-189 to Ser-204 Asp-209 to His-217 Pro-226 to His-233 Leu-272 to Glu-291.

1 30 85 1 - 15 - Pro-28 to Ala-36 Arg-71 to Ala-78 Gly-89 to Thr-96 Ar -131 to Ser-142.

2 12 67 1 - 15 - Glu-40 to Val-45 Pro-84 to Ser-90 Glu-134 to Val-139 Glu-152 to Asp-157 Thr-208 to His-217 Asp-278 to Ile-283 Ser-350 to Tyr-359 Gln-388 to Asp-394 Pro-435 to Ser-441 Leu-483 to Lys-489 Leu-491 to His-526 Pro-539 to Glu-544.

2 32 87 1 - 15 - Gly-45 to Asn-51 Leu-53 to Trp-58 Ser-63 to Gln-71 Asn-73 to Phe-90 Cys-162 to Phe-171 Cys-194 to Ser-213 Ser-234 to Phe-252.

2 33 88 1 - 15 - Arg-8 to Cys-32 Thr-58 to Gly-66 Ser-96 to Asp-115 Pro-117 to T -123.

3 13 68 1 - 15 - Pro-30 to Glu-40 Arg-93 to Lys-104 Tyr-109 to Thr-119 Trp-190 to Cys-196 Arg-246 to His-262 Tyr-271 to Pro-279 His-316 to Lys-335 Ala-343 to Arg-349 Asp-403 to Asn-408 Met-427 to Gly-433 Asn-439 to Tyr-459 Ser-469 to Glu-474 Ala-488 to Thr-493.

3 34 89 1 - 15 - Pro-30 to Glu-40 Arg-93 to Lys-104 Tyr-109 to Thr-119 Trp-190 to Cys-196 Arg-246 to As -261.

3 35 90 1 - 15 - His-17 to Lys-36 Ala-44 to Arg-50 Asp-104 to Asn-109 Met-128 to Gly-134 Asn-140 to Tyr-160 Ser-170 to Glu-175 Ala-189 to Thr-194.

4 14 69 1 - 15 - Pro-96 to Arg-103 Ar -108 to Gl -114.

4 36 91 1 - 15 - Pro-96 to Arg-103.

15 70 1 - 15 - Ser-37 to Asp-60 Gln-69 to Glu-77 Gly-79 to Ser-88 Ser-123 to Glu-131 Tyr-190 to Leu-234.

5 38 93 1 - 15 - Ala-36 to Ser-43 Leu-47 to Arg-52.
6 16 71 1 - 15 - Ile-38 to Arg-44 Pro-47 to Thr-53 Lys-69 to Ser-75 Ser-118 to Thr-126 Pro-206 to T -215.

6 39 94 1 - 15 - Ile-38 to Arg-44 Pro-47 to Thr-53 Lys-69 to Ser-75 Ser-118 to Thr-126.

6 40 95 1 - 15 - Pro-60 to T -69.
7 17 72 1 - 15 - Thr-156 to L s-162.

7 41 96 1 - 15 - Trp-94 to Ser-119 GI -128 to His-134.

7 42 97 1 - 15 - Arg-11 to Ser-38 Gln-44 to Cys-49 Glu-72 to His-78 Thr-288 to L s-294.
8 18 73 1 - 15 - Phe-184 to Lys-197 Glu-213 to Gln-219.

8 43 98 1 - 15 - Pro-16 to Arg-24.
9 19 74 1 - 15 - Ser-40 to Ser-48 Gln-110 to Glu-119 Glu-139 to Met-144 Ser-242 to Asn-250 Glu-276 to Ar -283.

9 44 99 1 - 15 - Ser-40 to Ser-48 Gln-110 to Glu-119 Glu-139 to Met-144.

20 75 1 - 15 - j Thr-44 to Cys-50 Gln-64 to Val-69 C s-73 to Val-82.

46 101 1 - 15 - Thr-44 to Cys-50 Gln-64 to Val-69 Cys-73 to Val-82.
11 49 104 1 - 15 - Phe-15 to Met-28 Trp-62 to Trp-72 Pro-79 to Asp-84 Thr-100 to Arg-109 Pro-132 to Leu-139 Pro-142 to Arg-154 Pro-167 to Gln-178.

11 50 105 1 - 15 - Ar -67 to L s-72.
12 22 77 1 - 15 - Ala-29 to Val-35 Pro-39 to Gly-48 Ser-55 to Gly-61 Pro-72 to Gly-81 Ser-89 to Gln-102 Lys-113 to Gln-118 Pro-127 to Ala-132.
13 23 78 1 - 15 - Ser-32 to Ser-39.

13 53 108 1 - 15 - Ser-32 to Ser-39.

13 54 109 1 - 15 - Ser-33 to Pro-38.
14 24 79 1 - 15 - Glu-37 to Ar -48.

14 56 111 1 - 15 - Ala-16 to Trp-23 Val-53 to Gl -61.

14 57 112 1 - 15 - Asn-1 to Trp-14 Thr-36 to Leu-43 Leu-63 to Asp-69 Ar -76 to Gln-89.

25 80 1 - 15 - Pro-142 to Pro-149.
15 58 113 1 - 15 - Arg-1 to Gly-13 Pro-66 to Pro-73 Met-102 to Thr-108 Ser-122 to Pro-130 Pro-172 to Glu-183 Ala-210 to Ser-220 L s-321 to Asn-330.
16 26 81 1 - 15 - Glu-20 to Gly-25 Gly-27 to Trp-34 Ala-163 to Glu-169 Gly-193 to Gly-198 Thr-219 to Thr-226 Leu-249 to L s-262.

16 59 114 1 - 15 - Glu-20 to Gly-25 Gly-27 to Trp-34 Ala-163 to Glu-169 Gl -193 to GI
-198.
17 27 82 1 - 15 - Pro-126 to Ser-131.
1251 126_5 17 61 116 1 - 15 - Glu-21 to Gly-32 Gly-62 to Arg-68 Ile-84 to Glu-94.

17 62 117 1 - 15 - Arg-21 to His-28 Glu-55 to Phe-61 Ser-75 to Gln-84.
18 28 83 1 - 15 - Ala-54 to Phe-59 Ser-85 to Thr-92 Pro-98 to Ar -108.

18 63 118 1 - 15 - Ala-54 to Phe-59 Ser-85 to Thr-92 Pro-98 to Ar -108.
19 29 84 1 - 15 - Ser-4 to Arg-11 Ser-13 to Gly-18 Val-88 to Asp-103 Pro-105 to Ser-110 Arg-129 to Lys-135 Pro-151 to Gly-168 Ala-172 to Ser-177 Pro-185 to Lys-190 Asn-200 to Phe-205 Gln-214 to Tyr-220 Arg-245 to Asp-250 Ser-361 to Lys-366 Ser-387 to Ala-392 Gln-425 to Asp-431 Tyr-494 to Tyr-504 Leu-507 to Val-515.

19 64 119 1 - 15 - Ser-32 to Lys-37 Pro-52 to Pro-63.

19 65 120 1 - 15 - Ser-14 to Arg-21 Ser-23 to Gly-28 Val-98 to Asp-113 Pro-115 to Ser-120 Arg-139 to Lys-145 Pro-161 to Gly-178 Ala-182 to Ser-187 Pro-195 to Lys-200 Asn-210 to Phe-215 Gln-224 to Tyr-230 Arg-255 to Asp-260 Ser-371 to Lys-376 Ser-397 to Ala-402 Gln-435 to Asp-441 Tyr-504 to Tyr-514 Leu-517 to Val-525.

Table 3 CIOrie Library ID Codes H0052H0059H0063H0069H0083H0085H0087H0090HO100HOlOS

T

S

I

S

Table 4 Library Library Description Disease Code H0002 Human Adult Heart H0004 Human Adult S leen H0008 Whole 6 Week Old Embr o H0009 Human Fetal Brain H0011 Human Fetal Kidne H0012 Human Fetal Kidne H0013 Human 8 Week Whole Embr o H0014 Human Gall Bladder H0015 Human Gall Bladder, fraction II

H0022 Jurkat Cells H0024 Human Fetal Lung III

H0026 Namalwa Cells H0030 Human Placenta H0031 Human Placenta H0032 Human Prostate H0036 Human Adult Small Intestine H0037 Human Adult Small Intestine H0038 Human Testes H0039 Human Pancreas Tumor disease H0040 Human Testes Tumor disease H0041 Human Fetal Bone H0042 Human Adult Pulmonary H0046 Human Endometrial Tumor disease H0050 Human Fetal Heart H0051 Human Hi oca us H0052 Human Cerebellum H0056 Human Umbilical Vein, Endo. remake H0059 Human Uterine Cancer disease H0063 Human Th us H0068 Human Skin Tumor disease H0069 Human Activated T-Cells H0078 Human Lun Cancer disease H0079 Human Whole 7 Week Old Embryo (II

H0081 Human Fetal E ithelium (Skin) H0085 Human Colon H0086 Human epithelioid sarcoma disease H0087 Human Thymus H0090 Human T-Cell L m homa disease H0099 Human Lung Cancer, subtracted HO100 Human Whole Six Week Old Embr o HO 101 Human 7 Weeks Old Embr o, subtracted H0105 Human Fetal Heart, subtracted H0108 Human Adult L h Node, subtracted HO110 Human Old Ovar , subtracted HO113 Human skin Tumor, subtracted H0116 Human Thymus Tumor, subtracted H0123 Human Fetal Dura Mater H0124 Human Rhabdomyosarcoma disease H0125 Cem cells c clohexamide treated H0130 LNCAP untreated H0134 Ra'i Cells, c clohexamide treated H0135 Human S ovial Sarcoma H0136 Su t Cells, c clohexamide treated H0141 Activated T-Cells, 12 hrs.

H0144 Nine Week Old Early Stage Human H0150 Human E ididymus H0151 Early Stage Human Liver H0152 Earl Sta a Human Liver, fract II

H0156 Human Adrenal Gland Tumor disease H0159 Activated T-Cells, 8 hrs., ligation H0167 Activated T-Cells, 24 hrs.

H0169 Human Prostate Cancer, Sta a C fractiondisease H0170 12 Week Old Earl Stage Human H0171 12 Week Old Earl Sta a Human, II

H0179 Human Neutro hil HO 188 Human Normal Breast H0204 Human Colon Cancer, subtracted H0205 Human Colon Cancer, differential H0208 Early Stage Human Lun , subtracted H0213 Human Pituitary, subtracted H0217 Su t cells, cyclohexamide treated, differentially ex ressed H0222 Activated T-Cells, 8 hrs, subtracted H0231 Human Colon, subtraction H0244 Human 8 Week Whole Emb o, subtracted H0246 Human Fetal Liver- Enz me subtraction H0250 Human Activated Monoc tes H0251 Human Chondrosarcoma disease H0252 Human Osteosarcoma disease H0253 Human adult testis, lar a inserts H0254 Breast L h node cDNA librar H0255 breast 1 h node CDNA libra H0261 H. cerebellum, Enzyme subtracted H0263 human colon cancer disease H0264 human tonsils H0265 Activated T-Cell l2hs)/Thiouridine labelledEco H0266 Human Microvascular Endothelial Cells, fract. A

H0268 Human Umbilical Vein Endothelial Cells, fract. A

H0269 Human Umbilical Vein Endothelial Cells, fract. B

H0270 HPAS human ancreas, subtracted) H0271 Human Neutro hil, Activated H0272 HUMAN TONSILS, FRACTION 2 H0281 L h node, abnorm. cell line ATCC
#7225) H0284 Human OB MG63 control fraction I

H0286 Human OB MG63 treated 10 nM E2) fraction I

H0290 Human OB HOS treated (1 nM E2) fraction I

H0292 Human OB HOS treated (10 nM E2) fraction I

H0293 WI 38 cells H0294 Amniotic Cells - TNF induced H0295 Amniotic Cells - Primar Culture H0298 HCBB's differential consolidation H0305 CD34 positive cells (Cord Blood) H0306 CD34 de leted Buff Coat (Cord Blood H0309 Human Chronic S ovitis disease H0318 HUMAN B CELL LYMPHOMA disease H0327 human co us colosum H0328 human ovarian cancer disease H0329 Dermatofibrosarcoma Protuberance disease H0331 He atocellular Tumor disease H0333 Hemangio ericytoma disease H0341 Bone Marrow Cell Line RS4,11 H0343 stomach cancer human disease H0350 Human Fetal Liver, mixed 10 & 14 week H0351 Glioblastoma disease H0352 wilm's tumor disease H0354 Human Leukoc tes H0355 Human Liver H0359 KMH2 cell line H0369 H. Atro hic Endometrium H0370 H. Lym h node breast Cancer disease H0373 Human Heart H0375 Human Lung H0376 Human S leen H0383 Human Prostate BPH, re-excision H0390 Human Am dala De ression, re-excisiondisease H0392 H. Menin ima, M1 H0393 Fetal Liver, subtraction II

H0396 L1 Cell line H0402 CD34 de feted Buff Coat Cord Blood , re-excision H0409 H. Striatum De ression, subtracted H0411 H Female Bladder, Adult H0412 Human umbilical vein endothelial cells, IL-4 induced H0413 Human Umbilical Vein Endothelial Cells, uninduced H0415 H. Ovarian Tumor, II, OV5232 disease H0416 Human Neutro hils, Activated, re-excision H0418 Human Pituita , subtracted VII

H0421 Human Bone Marrow, re-excision H0422 T-Cell PHA 16 hrs H0423 T-Cell PHA 24 hrs H0424 Human Pituita , subt IX

H0427 Human Adi ose H0428 Human Ovar H0431 H. Kidne Medulla, re-excision H0435 Ovarian Tumor 10-3-95 H0436 Restin T-Cell Libra ,II

H0438 H. Whole Brain #2, re-excision H0441 H. Kidne Cortex, subtracted H0444 Spleen metastic melanoma disease H0445 S leen, Chronic lymphocytic leukemiadisease H0455 H. Striatum De ression, subt H0457 Human Eosino hils H0458 CD34+ cell, I, frac II

~H0459 CD34+cells, II, FRACTION 2 ~

H0461 H. Kidne Medulla, subtracted H0483 Breast Cancer cell line, MDA 36 H0484 Breast Cancer Cell line, an io enic H0486 Hodgkin's L homa II disease H0488 Human Tonsils, Lib 2 H0489 Crohn's Disease disease H0490 HI-60, untreated, subtracted H0491 HL-60, PMA 4H, subtracted H0492 HL-60, RA 4h, Subtracted H0494 Keratinoc a H0497 HEL cell line H0506 Ulcerative Colitis H0509 Liver, He atoma disease H0510 Human Liver, normal H0518 BMC stimulated w/ of I/C

H0519 NTERA2, control H0520 NTERA2 + retinoic acid, 14 da s H0521 Prima Dendritic Cells, lib 1 H0522 Primar Dendritic cells,frac 2 H0529 M oloid Progenitor Cell Line H0530 Human Dermal Endothelial Cells,untreated H0533 Human Stromal endometrial fibroblasts, treated w/ estradiol H0535 Human ovary tumor cell OV350721 disease H0538 Merkel Cells H0539 Pancreas Islet Cell Tumor disease H0542 T Cell hel er I

H0543 T cell hel er II

H0544 Human endometrial stromal cells H0545 Human endometrial stromal cells-treated with ro esterone H0546 Human endometrial stromal cells-treated with estradiol H0547 NTERA2 teratocarcinoma cell line+retinoic acid ( 14 da s H0549 H. E ididi us, ca ut & co us H0550 H. E ididi us, cauda H0551 Human Thymus Stromal Cells H0553 Human Placenta H0555 Rejected Kidney, lib 4 disease H0556 Activated T-cell(12h /Thiouridine-re-excision H0559 HL-60, PMA 4H, re-excision H0570 Human Fetal Brain, normalized C500H

H0574 He atocellular Tumor, re-excision disease H0575 Human Adult Pulmona ,re-excision H0576 Restin T-Cell, re-excision H0578 Human Fetal Th us H0580 Dendritic cells, ooled H0581 Human Bone Marrow, treated H0583 B Cell 1 phoma disease H0584 Activated T-cells, 24 hrs,re-excision H0585 Activated T-Cells,l2 hrs,re-excision H0586 Healing roin wound, 6.5 hours ost disease incision H0587 Healing groin wound, 7.5 hours ost disease incision H0590 Human adult small intestine,re-excision H0591 Human T-cell 1 m homa,re-excision disease H0592 Healing roin wound - zero hr ost-incisiondisease control) H0593 Olfactor a ithelium,nasalcavit H0594 Human Lun Cancer,re-excision disease H0595 Stomach cancer human ,re-excision disease H0596 Human Colon Cancer,re-excision H0597 Human Colon, re-excision H0598 Human Stomach,re-excision H0599 Human Adult Heart,re-excision H0600 Healin Abdomen wound,70&90 min ost disease incision H0604 Human Pituitary, re-excision H0609 H. Leukoc es, normalized cot > 500A

H0615 Human Ovarian Cancer Reexcision disease H0616 Human Testes, Reexcision H0617 Human Primar Breast Cancer Reexcisiondisease H0618 Human Adult Testes, Lar a Inserts, Reexcision H0619 Fetal Heart H0620 Human Fetal Kidne , Reexcision H0622 Human Pancreas Tumor, Reexcision disease H0624 12 Week Early Stage Human II, Reexcision H0625 Ku 812F Basophils Line H0626 Saos2 Cells, Untreated H0627 Saos2 Cells, Vitamin D3 Treated H0628 Human Pre-Differentiated Adi oc es H0631 Saos2, Dexamethosome Treated H0632 He atocellular Tumor,re-excision H0633 Lung Carcinoma A549 TNFaI ha activateddisease H0634 Human Testes Tumor, re-excision disease H0635 Human Activated T-Cells, re-excision H0637 Dendritic Cells From CD34 Cells H0638 CD40 activated monoc a dendridic cells H0641 LPS activated derived dendritic cells H0643 He G2 Cells, PCR Libra H0644 Human Placenta (re-excision) H0645 Fetal Heart, re-excision H0646 Lung, Cancer (4005313 A3): Invasive Poorly Differentiated Lun Adenocarcinoma, H0647 Lung, Cancer (4005163 B7): Invasive,disease Poorly Diff.
Adenocarcinoma, Metastatic H0648 Ovary, Cancer: (4004562 B6) Papillarydisease Serous Cystic Neo lasm, Low Mali nant Pot H0649 Lung, Normal: (4005313 B1) H0650 B-Cells H0651 Ovary, Normal: (9805C040R) H0653 Stromal Cells H0656 B-cells (unstimulated) H0657 B-cells (stimulated) H0658 Ovary, Cancer (9809C332): Poorly disease differentiated adenocarcinoma H0659 Ovary, Cancer (15395A1F): Grade II disease Pa ills Carcinoma H0660 Ovary, Cancer: (15799A1F) Poorly disease differentiated carcinoma H0661 Breast, Cancer: (4004943 A5) disease H0662 Breast, Normal: (400552282) H0663 Breast, Cancer: 4005522 A2 disease H0664 Breast, Cancer: 9806C012R disease H0665 Stromal cells 3.88 H0666 Ovar , Cancer: 4004332 A2 disease H0668 stromal cell clone 2.5 H0669 Breast, Cancer: 4005385 A2 H0670 Ovary, Cancer(4004650 A3): Well-Differentiated Micro a illar Serous Carcinoma H0672 Ova , Cancer: 4004576 A8 H0673 Human Prostate Cancer, Stage B2, re-excision H0674 Human Prostate Cancer, Stage C, re-excission H0675 Colon, Cancer: (9808C064R) H0677 TNFR degenerate oli o H0682 Ovarian cancer, Serous Pa illar Adenocarcinoma H0684 Ovarian cancer, Serous Pa illar Adenocarcinoma H0685 Adenocarcinoma of Ova , Human Cell Line, # OVCAR-3 H0686 Adenocarcinoma of Ova , Human Cell Line H0687 Human normal ovar #96106215 H0689 Ovarian Cancer H0690 Ovarian Cancer, # 97026001 H0691 Normal Ova , #97106208 H0693 Normal Prostate #ODQ3958EN

H0694 Prostate cancer adenocarcinoma H0695 mononucleocytes from anent H0696 Prostate Adenocarcinoma H0702 NK15 IL2 treated for 48 hours) H0707 Stomach Cancer(5007635) H0709 Patient#2 Acute M eloid Leukemia/SGAH

L0022 Soares infant brain 1NIB

L1290 Human rom eloc a S0001 Brain frontal cortex 50002 Monoc to activated 50003 Human Osteoclastoma disease S0007 Earl Sta a Human Brain S0010 Human Am dala S0011 STROMAL -OSTEOCLASTOMA disease S0015 Kidney medulla 50016 Kidney Pyramids 50020 Seven Trans Membrane Rece for Famil 50022 Human Osteoclastoma Stromal Cells - unam lifted S0026 Stromal cell TF274 S0027 Smooth muscle, serum treated S0028 Smooth muscle,control 50031 S final cord S0036 Human Substantia Nigra S0037 Smooth muscle, ILIb induced S0038 Human Whole Brain #2 - Oligo dT >
l.SKb S0040 Adi oc tes 50044 Prostate BPH disease 50045 Endothelial cells-control S0046 Endothelial-induced S0049 Human Brain, Striatum S0051 Human H othalmus,Schizo hrenia disease S0052 neutro hils control S0053 Neutro hils IL-1 and LPS induced S0106 STRIATUM DEPRESSION disease SO110 Brain Am dala De ression disease S0112 H othalamus 50114 Aner is T-cell S0116 Bone marrow 50118 Smooth muscle control 2 50126 Osteoblasts 50132 E ithelial-TNFa and INF induced 50134 A o totic T-cell 50140 eosino hil-ILS induced S0142 Macro ha e-oxLDL

S0144 Macro ha a GM-CSF treated) SO150 LNCAP rostate cell line S0152 PC3 Prostate cell line S0182 Human B Cell 8866 S0194 S ovial h oxia 50196 S ovial IL-1/TNF stimulated 50206 Smooth Muscle- HASTE normalized 50210 Messangial cell, frac 2 S0212 Bone Marrow Stromal Cell, untreated S0214 Human Osteoclastoma, re-excision disease S0216 Neutro hils IL-1 and LPS induced 50218 A o totic T-cell, re-excision 50220 H. h othalamus, frac A,re-excision 50222 H. Frontal cortex,e ile tic,re-excisiondisease 50242 S ovial Fibroblasts (Ill/TNF), subt 50250 Human Osteoblasts II disease S0276 S ovial h oxia-RSF subtracted 50278 H Macro hage GM-CSF treated), re-excision 50280 Human Adi ose Tissue, re-excision S0282 Brain Frontal Cortex, re-excision S0294 La tumor disease S0300 Frontallobe,dementia,re-excision S0310 Normal trachea S0314 Human osteoarthritis,fraction I disease S0328 Palate carcinoma disease S0330 Palate normal 50332 Pha x carcinoma 50336 Human Normal Cartilage Fraction IV

50342 Adi oc es,re-excision 50344 Macro hage-oxLDL, re-excision S0346 Human Am dala,re-excision S0348 Cheek Carcinoma disease 50352 Lar nx Carcinoma disease 50354 Colon Normal II

S0356 Colon Carcinoma disease 50358 Colon Normal III

S0360 Colon Tumor II disease S0366 Human Soleus S0372 Larynx carcinoma III disease S0374 Normal colon S0376 Colon Tumor disease 50378 Pancreas normal PCA4 No S0380 Pancreas Tumor PCA4 Tu disease 50384 Ton ue carcinoma disease 50388 Human H othalamus,schizo hrenia, disease re-excision S0392 Salivar Gland S0402 Adrenal Gland,normal S0406 Rectum tumour S0408 Colon, normal S0410 Colon, tumour S0412 Tem oral cortex-Alzheizmer, subtracteddisease S0414 Hi ocam us, Alzheimer Subtracted S0418 CHME Cell Line,treated 5 hrs 50420 CHME Cell Line,untreated 50422 Mo7e Cell Line GM-CSF treated lng/ml S0424 TF-1 Cell Line GM-CSF Treated 50426 Monoc a activated, re-excision 50428 Neutro hils control, re-excision S0434 Stomach Normal disease S0436 Stomach Tumour disease S0438 Liver Normal MetSNo S0440 Liver Tumour Met 5 Tu S0442 Colon Normal S0444 Colon Tumor disease S0446 Ton ue Tumour S0450 Lar x Tumour S0454 Placenta S0462 Th oid Th oiditis S0464 La Normal S0474 Human blood latelets S3012 Smooth Muscle Serum Treated, Norm S3014 Smooth muscle, serum induced,re-exc S3018 TH1 cells S6028 Human Manic De ression Tissue disease T0002 Activated T-cells T0003 Human Fetal Lung T0004 Human White Fat T0006 Human Pineal Gland T0023 Human Pancreatic Carcinoma disease T0039 HSA 172 Cells T0040 HSC172 cells T0041 Jurkat T-cell G1 hase T0042 Jurkat T-Cell, S base T0048 Human Aortic Endothelium T0049 Aorta endothelial cells + TNF-a T0060 Human White Adi ose T0067 Human Th oid T0071 Human Bone Marrow T0082 Human Adult Retina T0104 HCC cell line metastisis to liver T0109 Human (HCC) cell line liver mouse) metastasis, remake TO110 Human colon carcinoma (HCC) cell line, remake T0114 Human (Caco-2) cell line, adenocarcinoma, colon, remake TO115 Human Colon Carcinoma (HCC) cell line The polypeptides of the invention can be prepared in any suitable manner.
Such polypeptides include isolated naturally occurring polypeptides, recombinantly produced polypeptides, synthetically produced polypeptides, or polypeptides produced by a combination of these methods. Means for preparing such polypeptides are well understood in the art.
The polypeptides may be in the form of the secreted protein, including the mature form, or may be a part of a larger protein, such as a fusion protein (see below).
It.is often advantageous to include an additional amino acid sequence which contains secretory or leader sequences, pro-sequences, sequences which aid in purification , such as multiple histidine residues, or an additional sequence for stability during recombinant production.
The polypeptides of the present invention are preferably provided in an isolated form, and preferably are substantially purified. A recombinantly produced version of a polypeptide, including the secreted polypeptide, can be substantially purified using techniques described herein or otherwise known in the art, such as, for example, by the one-step method described in Smith and Johnson, Gene 67:31-40 (1988). Polypeptides of the invention also can be purified from natural, synthetic or recombinant sources using techniques described herein or otherwise known in the art, such as, for example, antibodies of the invention raised against the secreted protein.
The present invention provides a polynucleotide comprising, or alternatively consisting of, the nucleic acid sequence of SEQ >T7 NO:X, and/or a cDNA
contained in ATCC deposit Z. The present invention also provides a polypeptide comprising, or alternatively, consisting of, the polypeptide sequence of SEQ ID NO:Y and/or a polypeptide encoded by the cDNA contained in ATCC deposit Z. Polynucleotides encoding a polypeptide comprising, or alternatively consisting of the polypeptide sequence of SEQ ID NO:Y and/or a polypeptide sequence encoded by the cDNA
contained in ATCC deposit Z are also encompassed by the invention.
Signal Seguences The present invention also encompasses mature forms of the polypeptide having the polypeptide sequence of SEQ ID NO:Y and/or the polypeptide sequence encoded by the cDNA in a deposited clone. Polynucleotides encoding the mature forms (such as, for example, the polynucleotide sequence in SEQ ID NO:X and/or the polynucleotide sequence contained in the cDNA of a deposited clone) are also encompassed by the invention. According to the signal hypothesis, proteins secreted by mammalian cells have a signal or secretary leader sequence which is cleaved from the mature protein once export of the growing protein chain across the rough endoplasmic reticulum has been initiated. Most mammalian cells and even insect cells cleave secreted proteins with the same specificity. However, in some cases, cleavage of a secreted protein is not entirely uniform, which results in two or more mature species of the protein. Further, it has long been known that cleavage specificity of a secreted protein is ultimately determined by the primary structure of the complete protein, that is, it is inherent in the amino acid sequence of the polypeptide.
Methods for predicting whether a protein has a signal sequence, as well as the cleavage point for that sequence, are available. For instance, the method of McGeoch, Virus Res. 3:271-286 (1985), uses the information from a short N-terminal 1 S charged region and a subsequent uncharged region of the complete (uncleaved) protein. The method of von Heinje, Nucleic Acids Res. 14:4683-4690 (1986) uses the information from the residues surrounding the cleavage site, typically residues -13 to +2, where +1 indicates the amino terminus of the secreted protein. The accuracy of predicting the cleavage points of known mammalian secretory proteins for each of these methods is in the range of 75-80%. (von Heinje, supra.) However, the two methods do not always produce the same predicted cleavage points) for a given protein.
In the present case, the deduced amino acid sequence of the secreted polypeptide was analyzed by a computer program called SignalP (Henrik Nielsen et al., Protein Engineering 10:1-6 (1997)), which predicts the cellular location of a protein based on the amino acid sequence. As part of this computational prediction of localization, the methods of McGeoch and von Heinje are incorporated. The analysis of the amino acid sequences of the secreted proteins described herein by this program provided the results shown in Table 1.
As one of ordinary skill would appreciate, however, cleavage sites sometimes vary from organism to organism and cannot be predicted with absolute certainty.
Accordingly, the present invention provides secreted polypeptides having a sequence shown in SEQ ID NO:Y which have an N-terminus beginning within 5 residues (i.e., + or - 5 residues) of the predicted cleavage point. Similarly, it is also recognized that in some cases, cleavage of the signal sequence from a secreted protein is not entirely uniform, resulting in more than one secreted species. These polypeptides, and the polynucleotides encoding such polypeptides, are contemplated by the present invention.
Moreover, the signal sequence identified by the above analysis may not necessarily predict the naturally occurnng signal sequence. For example, the naturally occurring signal sequence may be further upstream from the predicted signal sequence. However, it is likely that the predicted signal sequence will be capable of directing the secreted protein to the ER. Nonetheless, the present invention provides the mature protein produced by expression of the polynucleotide sequence of SEQ ID
NO:X and/or the polynucleotide sequence contained in the cDNA of a deposited clone, in a mammalian cell (e.g., COS cells, as desribed below). These polypeptides, 1 S and the polynucleotides encoding such polypeptides, are contemplated by the present invention.
Polynucleotide and Polvneptide Variants The present invention is directed to variants of the polynucleotide sequence disclosed in SEQ >D NO:X, the complementary strand thereto, and/or the cDNA
sequence contained in a deposited clone.
The present invention also encompasses variants of the polypeptide sequence disclosed in SEQ ID NO:Y and/or encoded by a deposited clone.
"Variant" refers to a polynucleotide or polypeptide differing from the polynucleotide or polypeptide of the present invention, but retaining essential properties thereof. Generally, variants are overall closely similar, and, in many regions, identical to the polynucleotide or polypeptide of the present invention.
The present invention is also directed to nucleic acid molecules which comprise, or alternatively consist of, a nucleotide sequence which is at least 80%, 85%, 90%, 95%, 96%, 97%, 98% or 99% identical to, for example, the nucleotide coding sequence in SEQ ID NO:X or the complementary strand thereto, the nucleotide coding sequence contained in a deposited cDNA clone or the complementary strand thereto, a nucleotide sequence encoding the polypeptide of SEQ >D NO:Y, a nucleotide sequence encoding the polypeptide encoded by the cDNA contained in a deposited clone, and/or polynucleotide fragments of any of these nucleic acid molecules (e.g., those fragments described herein).
Polynucleotides which hybridize to these nucleic acid molecules under stringent hybridization conditions or lower stringency conditions are also encompassed by the invention, as are polypeptides encoded by these polynucleotides.
The present invention is also directed to polypeptides which comprise, or alternatively consist of, an amino acid sequence which is at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% identical to, for example, the polypeptide sequence shown in SEQ m NO:Y, the polypeptide sequence encoded by the cDNA contained in a deposited clone, and/or polypeptide fragments of any of these polypeptides (e.g., those fragments described herein).
By a nucleic acid having a nucleotide sequence at least, for example, 95%
"identical" to a reference nucleotide sequence of the present invention, it is intended that the nucleotide sequence of the nucleic acid is identical to the reference sequence except that the nucleotide sequence may include up to five point mutations per each 100 nucleotides of the reference nucleotide sequence encoding the polypeptide.
In other words, to obtain a nucleic acid having a nucleotide sequence at least 95%
identical to a reference nucleotide sequence, up to 5% of the nucleotides in the reference sequence may be deleted or substituted with another nucleotide, or a number of nucleotides up to 5% of the total nucleotides in the reference sequence may be inserted into the reference sequence. The query sequence may be an entire sequence shown inTable 1, the ORF (open reading frame), or any fragment specified as described herein.
As a practical matter, whether any particular nucleic acid molecule or polypeptide is at least 80%, 85%, 90%, 95%, 96%, 97%, 98% or 99% identical to a nucleotide sequence of the presence invention can be determined conventionally using known computer programs. A preferred method for determining the best overall match between a query sequence (a sequence of the present invention) and a subject sequence, also referred to as a global sequence alignment, can be determined using the FASTDB computer program based on the algorithm of Brutlag et al.
(Comp.

App. Biosci. 6:237-245(1990)). In a sequence alignment the query and subject sequences are both DNA sequences. An RNA sequence can be compared by converting U's to T's. The result of said global sequence alignment is in percent identity. Preferred parameters used in a FASTDB alignment of DNA sequences to 5 calculate percent identiy are: Matrix=Unitary, k-tuple=4, Mismatch Penalty=1, Joining Penalty=30, Randomization Group Length=0, Cutoff Score=1, Gap Penalty=5, Gap Size Penalty 0.05, Window Size=500 or the lenght of the subject nucleotide sequence, whichever is shorter.
If the subject sequence is shorter than the query sequence because of 5' or 3' 10 deletions, not because of internal deletions, a manual correction must be made to the results. This is because the FASTDB program does not account for 5' and 3' truncations of the subject sequence when calculating percent identity. For subject sequences truncated at the 5' or 3' ends, relative to the query sequence, the percent identity is corrected by calculating the number of bases of the query sequence that are 15 5' and 3' of the subject sequence, which are not matched/aligned, as a percent of the total bases of the query sequence. Whether a nucleotide is matched/aligned is determined by results of the FASTDB sequence alignment. This percentage is then subtracted from the percent identity, calculated by the above FASTDB program using the specified parameters, to arrive at a final percent identity score. This corrected 20 score is what is used for the purposes of the present invention. Only bases outside the 5' and 3' bases of the subject sequence, as displayed by the FASTDB alignment, which are not matched/aligned with the query sequence, are calculated for the purposes of manually adjusting the percent identity score.
For example, a 90 base subject sequence is aligned to a 100 base query 25 sequence to determine percent identity. The deletions occur at the 5' end of the subject sequence and therefore, the FASTDB alignment does not show a matched/alignment of the first 10 bases at 5' end. The 10 unpaired bases represent 10% of the sequence (number of bases at the 5' and 3' ends not matched/total number of bases in the query sequence) so 10% is subtracted from the percent identity score 30 calculated by the FASTDB program. If the remaining 90 bases were perfectly matched the final percent identity would be 90%. In another example, a 90 base subject sequence is compared with a 100 base query sequence. This time the deletions are internal deletions so that there are no bases on the 5' or 3' of the subject sequence which are not matched/aligned with the query. In this case the percent .
identity calculated by FASTDB is not manually corrected. Once again, only bases 5' and 3' of the subject sequence which are not matched/aligned with the query sequence are manually corrected for. No other manual corrections are to made for the purposes of the present invention.
By a polypeptide having an amino acid sequence at least, for example, 95%
"identical" to a query amino acid sequence of the present invention, it is intended that the amino acid sequence of the subject polypeptide is identical to the query sequence except that the subject polypeptide sequence may include up to five amino acid alterations per each 100 amino acids of the query amino acid sequence. In other words, to obtain a polypeptide having an amino acid sequence at least 95%
identical to a query amino acid sequence, up to 5% of the amino acid residues in the subject sequence may be inserted, deleted, (indels) or substituted with another amino acid.
These alterations of the reference sequence may occur at the amino or carboxy terminal positions of the reference amino acid sequence or anywhere between those terminal positions, interspersed either individually among residues in the reference sequence or in one or more contiguous groups within the reference sequence.
As a practical matter, whether any particular polypeptide is at least 80%, 85%, 90%, 95%, 96%, 97%, 98% or 99% identical to, for instance, an amino acid sequences shown in Table 1 (SEQ 117 NO:Y) or to the amino acid sequence encoded by cDNA contained in a deposited clone can be determined conventionally using known computer programs. A preferred method for determing the best overall match between a query sequence (a sequence of the present invention) and a subject sequence, also referred to as a global sequence alignment, can be determined using the FASTDB computer program based on the algorithm of Brutlag et al. (Comp.
App.
Biosci. 6:237-245(1990)). In a sequence alignment the query and subject sequences are either both nucleotide sequences or both amino acid sequences. The result of said global sequence alignment is in percent identity. Preferred parameters used in a FASTDB amino acid alignment are: Matrix=PAM 0, k-tuple=2, Mismatch Penalty=1, Joining Penalty=20, Randomization Group Length=0, Cutoff Score=1, Window Size=sequence length, Gap Penalty=5, Gap Size Penalty=0.05, Window Size=500 or the length of the subject amino acid sequence, whichever is shorter.
If the subject sequence is shorter than the query sequence due to N- or C-terminal deletions, not because of internal deletions, a manual correction must be made to the results. This is because the FASTDB program does not account for N-S and C-terminal truncations of the subject sequence when calculating global percent identity. For subject sequences truncated at the N- and C-termini, relative to the query sequence, the percent identity is corrected by calculating the number of residues of the query sequence that are N- and C-terminal of the subject sequence, which are not matched/aligned with a corresponding subject residue, as a percent of the total bases of the query sequence. Whether a residue is matched/aligned is determined by results of the FASTDB sequence alignment. This percentage is then subtracted from the percent identity, calculated by the above FASTDB program using the specified parameters, to arrive at a final percent identity score. This final percent identity score is what is used for the purposes of the present invention. Only residues to the N- and C-termini of the subject sequence, which are not matched/aligned with the query sequence,_are -considered for the purposes of manually adjusting the percent identity score. That is, only query residue positions outside the farthest N- and C-terminal residues of the subject sequence.
For example, a 90 amino acid residue subject sequence is aligned with a 100 residue query sequence to determine percent identity. The deletion occurs at the N-terminus of the subject sequence and therefore, the FASTDB alignment does not show a matching/alignment of the first 10 residues at the N-terminus. The 10 unpaired residues represent 10% of the sequence (number of residues at the N-and C-termini not matched/total number of residues in the query sequence) so 10% is subtracted from the percent identity score calculated by the FASTDB program.
If the remaining 90 residues were perfectly matched the final percent identity would be 90%. In another example, a 90 residue subject sequence is compared with a 100 residue query sequence. This time the deletions are internal deletions so there are no residues at the N- or C-termini of the subject sequence which are not matched/aligned with the query. In this case the percent identity calculated by FASTDB is not manually corrected. Once again, only residue positions outside the N- and C-terminal ends of the subject sequence, as displayed in the FASTDB alignment, which are not matched/aligned with the query sequnce are manually corrected for. No other manual corrections are to made for the purposes of the present invention.
The variants may contain alterations in the coding regions, non-coding regions, or both. Especially preferred are polynucleotide variants containing alterations which produce silent substitutions, additions, or deletions, but do not alter the properties or activities of the encoded polypeptide. Nucleotide variants produced by silent substitutions due to the degeneracy of the genetic code are preferred.
Moreover, variants in which 5-10, 1-5, or 1-2 amino acids are substituted, deleted, or added in any combination are also preferred. Polynucleotide variants can be produced for a variety of reasons, e.g., to optimize codon expression for a particular host (change codons in the human mRNA to those preferred by a bacterial host such as E.
coli).
Naturally occurring variants are called "allelic variants," and refer to one of several alternate forms of a gene occupying a given locus on a chromosome of an organism. (Genes II, Lewin, B., ed., John Wiley & Sons, New York (1985).) These allelic variants can vary at either the polynucleotide and/or polypeptide level and are included in the present invention. Alternatively, non-naturally occurring variants may be produced by mutagenesis techniques or by direct synthesis.
Using known methods of protein engineering and recombinant DNA
technology, variants may be generated to improve or alter the characteristics of the polypeptides of the present invention. For instance, one or more amino acids can be deleted from the N-terminus or C-terminus of the secreted protein without substantial loss of biological function. The authors of Ron et al., J. Biol. Chem. 268:

(1993), reported variant KGF proteins having heparin binding activity even after deleting 3, 8, or 27 amino-terminal amino acid residues. Similarly, Interferon gamma exhibited up to ten times higher activity after deleting 8-10 amino acid residues from the carboxy terminus of this protein. (Dobeli et al., J. Biotechnology 7:199-(1988).) Moreover, ample evidence demonstrates that variants often retain a biological activity similar to that of the naturally occurnng protein. For example, Gayle and coworkers (J. Biol. Chem 268:22105-22111 (1993)) conducted extensive mutational analysis of human cytokine IL-1 a. They used random mutagenesis to generate over 3,500 individual IL-1 a mutants that averaged 2.5 amino acid changes per variant over the entire length of the molecule. Multiple mutations were examined at every possible amino acid position. The investigators found that "[m]ost of the molecule could be altered with little effect on either [binding or biological activity]." (See, Abstract.) In fact, only 23 unique amino acid sequences, out of more than 3,500 nucleotide sequences examined, produced a protein that significantly differed in activity from wild-type.
Furthermore, even if deleting one or more amino acids from the N-terminus or C-terminus of a polypeptide results in modification or loss of one or more biological functions, other biological activities may still be retained. For example, the ability of a deletion variant to induce and/or to bind antibodies which recognize the secreted form will likely be retained when less than the majority of the residues of the secreted form are removed from the N-terminus or C-terminus. Whether a particular polypeptide lacking N- or C-terminal residues of a protein retains such immunogenic activities can readily be determined by routine methods described herein and otherwise known in the art.
Thus, the invention further includes polypeptide variants which show substantial biological activity. Such variants include deletions, insertions, inversions, repeats, and substitutions selected according to general rules known in the art so as have little effect on activity. For example, guidance concerning how to make phenotypically silent amino acid substitutions is provided in Bowie et al., Science 247:1306-1310 (1990), wherein the authors indicate that there are two main strategies for studying the tolerance of an amino acid sequence to change.
The first strategy exploits the tolerance of amino acid substitutions by natural selection during the process of evolution. By comparing amino acid sequences in different species, conserved amino acids can be identified. These conserved amino acids are likely important for protein function. In contrast, the amino acid positions where substitutions have been tolerated by natural selection indicates that these positions are not critical for protein function. Thus, positions tolerating amino acid substitution could be modified while still maintaining biological activity of the protein.

The second strategy uses genetic engineering to introduce amino acid changes at specific positions of a cloned gene to identify regions critical for protein function.
For example, site directed mutagenesis or alanine-scanning mutagenesis (introduction of single alanine mutations at every residue in the molecule) can be used.
(Cunningham and Wells, Science 244:1081-1085 (1989).) The resulting mutant molecules can then be tested for biological activity.
As the authors state, these two strategies have revealed that proteins are surprisingly tolerant of amino acid substitutions. The authors further indicate which amino acid changes are likely to be permissive at certain amino acid positions in the 10 protein. For example, most buried (within the tertiary structure of the protein) amino acid residues require nonpolar side chains, whereas few features of surface side chains are generally conserved. Moreover, tolerated conservative amino acid substitutions involve replacement of the aliphatic or hydrophobic amino acids Ala, Val, Leu and Ile; replacement of the hydroxyl residues Ser and Thr; replacement of the acidic 15 residues Asp and Glu; replacement of the amide residues Asn and Gln, replacement of the_basic residues_ Lys, Arg; and His; replacement of the aromatic residues Phe, Tyr, and Trp, and replacement of the small-sized amino acids Ala, Ser, Thr, Met, and Gly.
Besides conservative amino acid substitution, variants of the present invention include (i) substitutions with one or more of the non-conserved amino acid residues, 20 where the substituted amino acid residues may or may not be one encoded by the genetic code, or (ii) substitution with one or more of amino acid residues having a substituent group, or (iii) fusion of the mature polypeptide with another compound, such as a compound to increase the stability and/or solubility of the polypeptide (for example, polyethylene glycol), or (iv) fusion of the polypeptide with additional amino 25 acids, such as, for example, an IgG Fc fusion region peptide, or leader or secretory sequence, or a sequence facilitating purification or (v) fusion of the polypeptide with another compound, such as albumin (including, but not limited to, recombinant albumin (see, e.g., U.S. Patent No. 5,876,969, issued March 2, 1999, EP Patent 622, and U.S. Patent No. 5,766,883, issued June 16, 1998, herein incorporated by 30 reference in their entirety)). Such variant polypeptides are deemed to be within the scope of those skilled in the art from the teachings herein.

For example, polypeptide variants containing amino acid substitutions of charged amino acids with other charged or neutral amino acids may produce proteins with improved characteristics, such as less aggregation.. Aggregation of pharmaceutical formulations both reduces activity and increases clearance due to the aggregate's immunogenic activity. (Pinckard et al., Clin. Exp. Immunol. 2:331-(1967); Robbins et al., Diabetes 36: 838-845 (1987); Cleland et al., Crit.
Rev.
Therapeutic Drug Carrier Systems 10:307-377 (1993).) A further embodiment of the invention relates to a polypeptide which comprises the amino acid sequence of the present invention having an amino acid sequence which contains at least one amino acid substitution, but not more than 50 amino acid substitutions, even more preferably, not more than 40 amino acid substitutions, still more preferably, not more than 30 amino acid substitutions, and still even more preferably, not more than 20 amino acid substitutions. Of course, in order of ever-increasing preference, it is highly preferable for a peptide or polypeptide to have an amino acid sequence which comprises the amino acid sequence of the present invention, which contains at least one, but not more than 10, 9, 8, 7, 6, 5, 4, 3, 2 or 1 amino acid substitutions. In specific embodiments, the number of additions, substitutions, and/or deletions in the amino acid sequence of the present invention or fragments thereof (e.g., the mature form and/or other fragments described herein), is 1-5, 5-10, 5-25, 5-50, 10-50 or 50-150, conservative amino acid substitutions are preferable.
Polynucleotide and Polypeptide Fragments The present invention is also directed to polynucleotide fragments of the polynucleotides of the invention.
In the present invention, a "polynucleotide fragment" refers to a short polynucleotide having a nucleic acid sequence which: is a portion of that contained in a deposited clone, or encoding the polypeptide encoded by the cDNA in a deposited clone; is a portion of that shown in SEQ ID NO:X or the complementary strand thereto, or is a portion of a polynucleotide sequence encoding the polypeptide of SEQ
ID NO:Y. The nucleotide fragments of the invention are preferably at least about 15 Ilt, and more preferably at least about 20 nt, still more preferably at least about 30 nt, and even more preferably, at least about 40 nt, at least about 50 nt, at least about 75 nt, or at least about 150 nt in length. A fragment "at least 20 nt in length,"
for example, is intended to include 20 or more contiguous bases from the cDNA
sequence contained in a deposited clone or the nucleotide sequence shown in SEQ ID
NO:X. In this context "about" includes the particularly recited value, a value larger or smaller by several (5, 4, 3, 2, or 1) nucleotides, at either terminus or at both termini. These nucleotide fragments have uses that include, but are not limited to, as diagnostic probes and primers as discussed herein. Of course, larger fragments (e.g., 50, 150, 500, 600, 2000 nucleotides) are preferred.
Moreover, representative examples of polynucleotide fragments of the invention, include, for example, fragments comprising, or alternatively consisting of, a sequence from about nucleotide number 1-50, 51-100, 101-150, 151-200, 201-250, 251-300, 301-350, 351-400, 401-450, 451-500, 501-550, 551-600, 651-700, 701-750, 751-800, 800-850, 851-900, 901-950, 951-1000, 1001-1050, 1051-1100, 1101-1150, 1151-1200, 1201-1254, 1251-1300, 1301-1350, 1351-1400, 1401-1450, 1451-1500, 1501-1550, 1551-1600, 1601=1650, 165-1-1700, 1701-1750, 1751-1800, 1801-1850, 1851-1900, 1901-1950, 1951-2000, or 2001 to the end of SEQ ID NO:X, or the complementary strand thereto, or the cDNA contained in a deposited clone. In this context "about" includes the particularly recited ranges, and ranges larger or smaller by several (5, 4, 3, 2, or 1 ) nucleotides, at either terminus or at both termini.
Preferably, these fragments encode a polypeptide which has biological activity. More preferably, these polynucleotides can be used as probes or primers as discussed herein. Polynucleotides which hybridize to these nucleic acid molecules under stringent hybridization conditions or lower stringency conditions are also encompassed by the invention, as are polypeptides encoded by these polynucleotides.
In the present invention, a "polypeptide fragment" refers to an amino acid sequence which is a portion of that contained in SEQ DJ NO:Y or encoded by the cDNA contained in a deposited clone. Protein (polypeptide) fragments may be "free-standing," or comprised within a larger polypeptide of which the fragment forms a part or region, most preferably as a single continuous region. Representative examples of polypeptide fragments of the invention, include, for example, fragments comprising, or alternatively consisting of, from about amino acid number 1-20, 21-40, 41-60, 61-80, 81-100, 102-120, 121-140, 141-160, or 161 to the end of the coding region. Moreover, polypeptide fragments can be about 20, 30, 40, 50, 60, 70, 80, 90, 100, 110, 120, 130, 140, or 150 amino acids in length. In this context "about"
includes the particularly recited ranges or values, and ranges or values larger or smaller by several (5, 4, 3, 2, or 1 ) amino acids, at either extreme or at both extremes.
Polynucleotides encoding these polypeptides are also encompassed by the invention.
Preferred polypeptide fragments include the secreted protein as well as the mature form. Further preferred polypeptide fragments include the secreted protein or the mature form having a continuous series of deleted residues from the amino or the carboxy terminus, or both. For example, any number of amino acids, ranging from 1 60, can be deleted from the amino terminus of either the secreted polypeptide or the mature form. Similarly, any number of amino acids, ranging from 1-30, can be deleted from the carboxy terminus of the secreted protein or mature form.
Furthermore, any combination of the above amino and carboxy terminus deletions are preferred. Similarly, polynucleotides encoding these polypeptide fragments are also preferred.
Also preferred are polypeptide and polynucleotide fragments characterized by structural or functional domains, such as fragments that comprise alpha-helix and alpha-helix forming regions, beta-sheet and beta-sheet-forming regions, turn and turn-forming regions, coil and coil-forming regions, hydrophilic regions, hydrophobic regions, alpha amphipathic regions, beta amphipathic regions, flexible regions, surface-forming regions, substrate binding region, and high antigenic index regions.
Polypeptide fragments of SEQ ID NO:Y falling within conserved domains are specifically contemplated by the present invention. Moreover, polynucleotides encoding these domains are also contemplated.
Other preferred polypeptide fragments are biologically active fragments.
Biologically active fragments are those exhibiting activity similar, but not necessarily identical, to an activity of the polypeptide of the present invention. The biological activity of the fragments may include an improved desired activity, or a decreased undesirable activity. Polynucleotides encoding these polypeptide fragments are also encompassed by the invention.

Preferably, the polynucleotide fragments of the invention encode a polypeptide which demonstrates a functional activity. By a polypeptide demonstrating a "functional activity" is meant, a polypeptide capable of displaying one or more known functional activities associated with a full-length (complete) polypeptide of invention protein. Such functional activities include, but are not limited to, biological activity, antigenicity [ability to bind (or compete with a polypeptide of the invention for binding) to an antibody to the polypeptide of the invention], immunogenicity (ability to generate antibody which binds to a polypeptide of the invention), ability to form multimers with polypeptides of the invention, and ability to bind to a receptor or ligand for a polypeptide of the invention.
The functional activity of polypeptides of the invention, and fragments, variants derivatives, and analogs thereof, can be assayed by various methods.
For example, in one embodiment where one is assaying for the ability to bind or compete with full-length polypeptide of the invention for binding to an antibody of the polypeptide of the invention, various immunoassays known in the art can be used, including but not limited to, competitive and non-competitive assay systems using techniques such as radioimmunoassays, ELISA (enzyme linked immunosorbent assay), "sandwich" immunoassays, immunoradiometric assays, gel diffusion precipitation reactions, immunodiffusion assays, in situ immunoassays (using colloidal gold, enzyme or radioisotope labels, for example), western blots, precipitation reactions, agglutination assays (e.g., gel agglutination assays, hemagglutination assays), complement fixation assays, immunofluorescence assays, protein A assays, and immunoelectrophoresis assays, etc. In one embodiment, antibody binding is detected by detecting a label on the primary antibody. In another embodiment, the primary antibody is detected by detecting binding of a secondary antibody or reagent to the primary antibody. In a further embodiment, the secondary antibody is labeled. Many means are known in the art for detecting binding in an immunoassay and are within the scope of the present invention.
In another embodiment, where a ligand for a polypeptide of the invention identified, or the ability of a polypeptide fragment, variant or derivative of the invention to multimerize is being evaluated, binding can be assayed, e.g., by means well-known in the art, such as, for example, reducing and non-reducing gel chromatography, protein affinity chromatography, and affinity blotting. See generally, Phizicky, E., et al., 1995, Microbiol. Rev. 59:94-123. In another embodiment, physiological correlates of binding of a polypeptide of the invention to its substrates (signal transduction) can be assayed.
$ In addition, assays described herein (see Examples) and otherwise known in the art may routinely be applied to measure the ability of polypeptides of the invention and fragments, variants derivatives and analogs thereof to elicit related biological activity related to that of the polypeptide of the invention (either in vitro or in vivo). Other methods will be known to the skilled artisan and are within the scope 10 of the invention.
Epitopes and Antibodies The present invention encompasses polypeptides comprising, or alternatively consisting of, an epitope of the polypeptide having an amino acid sequence of SEQ ID
15 NO:Y, or an epitope of the polypeptide sequence encoded by a polynucleotide .
sequence contained in ATCC deposit No. Z or encoded by a polynucleotide that hybridizes to the complement of the sequence of SEQ ID NO:X or contained in ATCC deposit No. Z under stringent hybridization conditions or lower stringency hybridization conditions as defined supra. The present invention further encompasses 20 polynucleotide sequences encoding an epitope of a polypeptide sequence of the invention (such as, for example, the sequence disclosed in SEQ ID NO:X), polynucleotide sequences of the complementary strand of a polynucleotide sequence encoding an epitope of the invention, and polynucleotide sequences which hybridize to the complementary strand under stringent hybridization conditions or lower 25 stringency hybridization conditions defined supra.
The term "epitopes," as used herein, refers to portions of a polypeptide having antigenic or immunogenic activity in an animal, preferably a mammal, and most preferably in a human. In a preferred embodiment, the present invention encompasses a polypeptide comprising an epitope, as well as the polynucleotide 30 encoding this polypeptide. An "immunogenic epitope," as used herein, is defined as a portion of a protein that elicits an antibody response in an animal, as determined by any method known in the art, for example, by the methods for generating antibodies described infra. (See, for example, Geysen et al., Proc. Natl. Acad. Sci. USA
81:3998- 4002 (1983)). The term "antigenic epitope," as used herein, is defined as a portion of a protein to which an antibody can immunospecifically bind its antigen as determined by any method well known in the art, for example, by the immunoassays described herein. Immunospecific binding excludes non-specific binding but does not necessarily exclude cross- reactivity with other antigens. Antigenic epitopes need not necessarily be immunogenic.
Fragments which function as epitopes may be produced by any conventional means. (See, e.g., Houghten, Proc. Natl. Acad. Sci. USA 82:5131-5135 (1985), further described in U.S. Patent No. 4,631,211).
In the present invention, antigenic epitopes preferably contain a sequence of at least 4, at least 5, at least 6, at least 7, more preferably at least 8, at least 9, at least 10, at least 11, at least 12, at least 13, at least 14, at least 15, at least 20, at least 25, at least 30, at least 40, at least 50, and, most preferably, between about 15 to about 30 amino acids. Preferred polypeptides comprising immunogenic or antigenic epitopes are at least 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, or 100 amino acid residues in length. Additional non-exclusive preferred antigenic epitopes include the antigenic epitopes disclosed herein, as well as portions thereof.
Antigenic epitopes are useful, for example, to raise antibodies, including monoclonal antibodies, that specifically bind the epitope. Preferred antigenic epitopes include the antigenic epitopes disclosed herein, as well as any combination of two, three, four, five or more of these antigenic epitopes. Antigenic epitopes can be used as the target molecules in immunoassays. (See, for instance, Wilson et al., Cell 37:767-778 (1984);
Sutcliffe et al., Science 219:660-666 (1983)).
Similarly, immunogenic epitopes can be used, for example, to induce antibodies according to methods well known in the art. (See, for instance, Sutcliffe et al., supra; Wilson et al., supra; Chow et al., Proc. Natl. Acad. Sci. USA
82:910-914; and Bittle et al., J. Gen. Virol. 66:2347-2354 (1985). Preferred immunogenic epitopes include the immunogenic epitopes disclosed herein, as well as any combination of two, three, four, five or more of these immunogenic epitopes.
The polypeptides comprising one or more immunogenic epitopes may be presented for eliciting an antibody response together with a carrier protein, such as an albumin, to an animal system (such as rabbit or mouse), or, if the polypeptide is of sufficient length (at least about 25 amino acids), the polypeptide may be presented without a Garner. However, immunogenic epitopes comprising as few as 8 to 10 amino acids have been shown to be sufficient to raise antibodies capable of binding to, at the very least, linear epitopes in a denatured polypeptide (e.g., in Western blotting).
Epitope-bearing polypeptides of the present invention may be used to induce antibodies according to methods well known in the art including, but not limited to, in vivo immunization, in vitro immunization, and phage display methods. See, e.g., Sutcliffe et al., supra; Wilson et al., supra, and Bittle et al., J. Gen.
Virol., 66:2347-2354 (1985). If in vivo immunization is used, animals may be immunized with free peptide; however, anti-peptide antibody titer may be boosted by coupling the peptide to a macromolecular Garner, such as keyhole limpet hemacyanin (KLH) or tetanus toxoid. For instance, peptides containing cysteine residues may be coupled to a carrier using a linker such as maleimidobenzoyl= N-hydroxysuccinimide ester (MBS), while other peptides may be coupled to carriers using a more general linking agent such-as glutaraldehyde. Animals such as rabbits, rats and mice are immunized with either free or carrier- coupled peptides, for instance, by intraperitoneal and/or intradermal injection of emulsions containing about 100 ~g of peptide or carrier protein and Freund's adjuvant or any other adjuvant known for stimulating an immune response. Several booster injections may be needed, for instance, at intervals of about two weeks, to provide a useful titer of anti-peptide antibody which can be detected, for example, by ELISA assay using free peptide adsorbed to a solid surface. The titer of anti-peptide antibodies in serum from an immunized animal may be increased by selection of anti-peptide antibodies, for instance, by adsorption to the peptide on a solid support and elution of the selected antibodies according to methods well known in the art.
As one of skill in the art will appreciate, and as discussed above, the polypeptides of the present invention comprising an immunogenic or antigenic epitope can be fused to other polypeptide sequences. For example, the polypeptides of the present invention may be fused with the constant domain of immunoglobulins (IgA, IgE, IgG, IgM), or portions thereof (CHl, CH2, CH3, or any combination thereof and portions thereof), or albumin (including but not limited to recombinant albumin (see, e.g., U.S. Patent No. 5,876,969, issued March 2, 1999, EP Patent 622, and U.S. Patent No. 5,766,883, issued June 16, 1998, herein incorporated by reference in their entirety)), resulting in chimeric polypeptides. Such fusion proteins may facilitate purification and may increase half life in vivo. This has been shown for chimeric proteins consisting of the first two domains of the human CD4-polypeptide and various domains of the constant regions of the heavy or light chains of mammalian immunoglobulins. See, e.g., EP 394,827; Traunecker et al., Nature, 331:84-86 (1988). Enhanced delivery of an antigen across the epithelial barner to the immune system has been demonstrated for antigens (e.g., insulin) conjugated to an FcRn binding partner such as IgG or Fc fragments (see, e.g., PCT Publications WO
96/22024 and WO 99/04813). IgG Fusion proteins that have a disulfide-linked dimeric structure due to the IgG portion desulfide bonds have also been found to be more efficient in binding and neutralizing other molecules than monomeric polypeptides or fragments thereof alone. See, e.g., Fountoulakis et al., J.
Biochem., 270:3958-3964 (1995). Nucleic acids encoding the above epitopes can also be recombined with a gene of interest as an epitope tag (e.g., the hemagglutinin ("HA") tag or flag tag) to aid in detection and purification of the expressed polypeptide. For example, a system described by Janknecht et al. allows for the ready purification of non-denatured fusion proteins expressed in human cell lines (Janknecht et al., 1991, Proc. Natl. Acad. Sci. USA 88:8972- 897). In this system, the gene of interest is subcloned into a vaccinia recombination plasmid such that the open reading frame of the gene is translationally fused to an amino-terminal tag consisting of six histidine residues. The tag serves as a matrix binding domain for the fusion protein.
Extracts from cells infected with the recombinant vaccinia virus are loaded onto Ni2+
nitriloacetic acid-agarose column and histidine-tagged proteins can be selectively eluted with imidazole-containing buffers.
Additional fusion proteins of the invention may be generated through the techniques of gene-shuffling, motif shuffling, exon-shuffling, and/or codon-shuffling (collectively referred to as "DNA shuffling"). DNA shuffling may be employed to modulate the activities of polypeptides of the invention, such methods can be used to generate polypeptides with altered activity, as well as agonists and antagonists of the polypeptides. See, generally, U.S. Patent Nos. 5,605,793; 5,811,238;
5,830,721;

5,834,252; and 5,837,458, and Patten et al., Curr. Opinion Biotechnol. 8:724-(1997); Harayama, Trends Biotechnol. 16(2):76-82 (1998); Hansson, et al., J.
Mol.
Biol. 287:265-76 (1999); and Lorenzo and Blasco, Biotechniques 24(2):308- 13 (1998) (each of these patents and publications are hereby incorporated by reference in its entirety). In one embodiment, alteration of polynucleotides corresponding to SEQ
m NO:X and the polypeptides encoded by these polynucleotides may be achieved by DNA shuffling. DNA shuffling involves the assembly of two or more DNA
segments by homologous or site-specific recombination to generate variation in the polynucleotide sequence. In another embodiment, polynucleotides of the invention, or the encoded polypeptides, may be altered by being subjected to random mutagenesis by error-prone PCR, random nucleotide insertion or other methods prior to recombination. In another embodiment, one or more components, motifs, sections, parts, domains, fragments, etc., of a polynucleotide encoding a polypeptide of the invention may be recombined with one or more components, motifs, sections, parts, -domains; fragments, etc. of one or more heterologous molecules.
Antibodies Further polypeptides of the invention relate to antibodies and T-cell antigen receptors (TCR) which immunospecifically bind a polypeptide, polypeptide fragment, or variant of SEQ >D NO:Y, and/or an epitope, of the present invention (as determined by immunoassays well known in the art for assaying specific antibody-antigen binding). Antibodies of the invention include, but are not lirriited to, polyclonal, monoclonal, multispecific, human, humanized or chimeric antibodies, single chain antibodies, Fab fragments, F(ab') fragments, fragments produced by a Fab expression library, anti-idiotypic (anti-Id) antibodies (including, e.g., anti-Id antibodies to antibodies of the invention), and epitope-binding fragments of any of the above. The term "antibody," as used herein, refers to immunoglobulin molecules and immunologically active portions of immunoglobulin molecules, i.e., molecules that contain an antigen binding site that immunospecifically binds an antigen.
The immunoglobulin molecules of the invention can be of any type (e.g., IgG, IgE, IgM, IgD, IgA and IgY), class (e.g., IgGI, IgG2, IgG3, IgG4, IgAl and IgA2) or subclass of immunoglobulin molecule. In preferred embodiments, the immunoglobulin molecules of the invention are IgGl. In other preferred embodiments, the immunoglobulin molecules of the invention are IgG4.
Most preferably the antibodies are human antigen-binding antibody fragments of the present invention and include, but are not limited to, Fab, Fab' and F(ab')2, Fd, single-chain Fvs (scFv), single-chain antibodies, disulfide-linked Fvs (sdFv) and fragments comprising either a VL or VH domain. Antigen-binding antibody fragments, including single-chain antibodies, may comprise the variable regions) alone or in combination with the entirety or a portion of the following: hinge region, CH1, CH2, and CH3 domains. Also included in the invention are antigen-binding fragments also comprising any combination of variable regions) with a hinge region, CH1, CH2, and CH3 domains. The antibodies of the invention may be from any animal origin including birds and mammals. Preferably, the antibodies are human, murine (e.g., mouse and rat), donkey, ship rabbit, goat, guinea pig, camel, horse, or chicken. As used herein, "human" antibodies include antibodies having the amino acid sequence of a human immunoglobulin and include antibodies isolated from human immunoglobulin libraries or from animals transgenic for one or more human immunoglobulin and that do not express endogenous immunoglobulins, as described infra and, for example in, U.S. Patent No. 5,939,598 by Kucherlapati et al.
The antibodies of the present invention may be monospecific, bispecific, trispecific or of greater multispecificity. Multispecific antibodies may be specific for different epitopes of a polypeptide of the present invention or may be specific for both a polypeptide of the present invention as well as for a heterologous epitope, such as a heterologous polypeptide or solid support material. See, e.g., PCT
publications WO
93/17715; WO 92/08802; WO 91/00360; WO 92/05793; Tutt, et al., J. Immunol.
147:60-69 (1991); U.S. Patent Nos. 4,474,893; 4,714,681; 4,925,648; 5,573,920;
5,601,819; Kostelny et al., J. Immunol. 148:1547-1553 (1992).
Antibodies of the present invention may be described or specified in terms of the epitope(s) or portions) of a polypeptide of the present invention which they recognize or specifically bind. The epitope(s) or polypeptide portions) may be specified as described herein, e.g., by N-terminal and C-terminal positions, by size in contiguous amino acid residues, or listed in the Tables and Figures.
Antibodies which specifically bind any epitope or polypeptide of the present invention may also be excluded. Therefore, the present invention includes antibodies that specifically bind polypeptides of the present invention, and allows for the exclusion of the same.
Antibodies of the present invention may also be described or specified in terms of their cross-reactivity. Antibodies that do not bind any other analog, ortholog, or homolog of a polypeptide of the present invention are included.
Antibodies that bind polypeptides with at least 95%, at least 90%, at least 85%, at least 80%, at least 75%, at least 70%, at least 65%, at least 60%, at least 55%, and at least 50% identity (as calculated using methods known in the art and described herein) to a polypeptide of the present invention are also included in the present invention. In specific embodiments, antibodies of the present invention cross-react with murine, rat and/or rabbit homologs of human proteins and the corresponding epitopes thereof. Antibodies that do not bind polypeptides with less than 95%, less than 90%, less than 85%, less than 80%, less than 75%, less than 70%, less than 65%, less than 60%, less than 55%, and less than 50% identity (as calculated using methods known in the art and described herein) to a polypeptide of the present invention are also included in the present invention. In a specific embodiment, the above-described cross-reactivity is with respect to any single specific antigenic or immunogenic polypeptide, or combinations) of 2, 3, 4, 5, or more of the specific antigenic and/or immunogenic polypeptides disclosed herein. Further included in the present invention are antibodies which bind polypeptides encoded by polynucleotides which hybridize to a polynucleotide of the present invention under stringent hybridization conditions (as described herein). Antibodies of the present invention may also be described or specified in terms of their binding affinity to a polypeptide of the invention. Preferred binding affinities include those with a dissociation constant or Kd less than 5 X 10-z M, 10-Z M, 5 X 10-3 M, 10-3 M, 5 X 10-4 M, 10~ M, 5 X 10-S M, 10-5 M, 5 X 10-6 M, 10-6M, 5 X 10-' M, 107 M, 5 X 10-g M, 10-g M, 10-9 M, 10-9 M, 5 X 10-' ° M, 10-' ° M, 5 X 10-" M, 10-" M, 5 X
10-' 2 M, ' °-' 2 M, 5 X
10-' 3 M, 10-' 3 M, 5 X 10-' 4 M, 10-' 4 M, 5 X 10-' S M, or 10-' S M.
The invention also provides antibodies that competitively inhibit binding of an antibody to an epitope of the invention as determined by any method known in the art for determining competitive binding, for example, the immunoassays described herein. In preferred embodiments, the antibody competitively inhibits binding to the epitope by at least 95%, at least 90%, at least 85 %, at least 80%, at least 75%, at least 70%, at least 60%, or at least 50%.
Antibodies of the present invention may act as agonists or antagonists of the polypeptides of the present invention. For example, the present invention includes antibodies which disrupt the receptor/ligand interactions with the polypeptides of the invention either partially or fully. Preferrably, antibodies of the present invention bind an antigenic epitope disclosed herein, or a portion thereof. The invention features both receptor-specific antibodies and ligand-specific antibodies. The invention also features receptor-specific antibodies which do not prevent ligand binding but prevent receptor activation. Receptor activation (i.e., signaling) may be determined by techniques described herein or otherwise known in the art. For example, receptor activation can be determined by detecting the phosphorylation (e.g., tyrosine or serine/threonine) of the receptor or its substrate by immunoprecipitation followed by western blot analysis (for example, as described supra). In specific embodiments, antibodies are provided that inhibit ligand activity or receptor activity by at least 95%, at least 90%, at least 85%, at least 80%, at least 75%, at least 70%, at least 60%, or at least 50% of the activity in absence of the antibody.
The invention also features receptor-specific antibodies which both prevent ligand binding and receptor activation as well as antibodies that recognize the receptor-ligand complex, and, preferably, do not specifically recognize the unbound receptor or the unbound ligand. Likewise, included in the invention are neutralizing antibodies which bind the ligand and prevent binding of the ligand to the receptor, as well as antibodies which bind the ligand, thereby preventing receptor activation, but do not prevent the ligand from binding the receptor. Further included in the invention are antibodies which activate the receptor. These antibodies may act as receptor agonists, i.e., potentiate or activate either all or a subset of the biological activities of the ligand-mediated receptor activation, for example, by inducing dimerization of the receptor. The antibodies may be specified as agonists, antagonists or inverse agonists for biological activities comprising the specific biological activities of the peptides of the invention disclosed herein. The above antibody agonists can be made using methods known in the art. See, e.g., PCT publication WO 96/40281; U.S. Patent No.

5,811,097; Deng et al., Blood 92(6):1981-1988 (1998); Chen et al., Cancer Res.
58(16):3668-3678 (1998); Harrop et al., J. Immunol. 161(4):1786-1794 (1998);
Zhu et al., Cancer Res. 58(15):3209-3214 (1998); Yoon et al., J. Immunol.
160(7):3170-3179 (1998); Prat et al., J. Cell. Sci. 111(Pt2):237-247 (1998); Pitard et al., J.
Immunol. Methods 205(2):177-190 (1997); Liautard et al., Cytokine 9(4):233-241 (1997); Carlson et al., J. Biol. Chem. 272(17):11295-11301 (1997); Taryman et al., Neuron 14(4):755-762 (1995); Muller et al., Structure 6(9):1153-1167 (1998);
Bartunek et al., Cytokine 8(1):14-20 (1996) (which are all incorporated by reference herein in their entireties).
Antibodies of the present invention may be used, for example, but not limited to, to purify, detect, and target the polypeptides of the present invention, including both in vitro and in vivo diagnostic and therapeutic methods. For example, the antibodies have use in immunoassays for qualitatively and quantitatively measuring levels of the polypeptides of the present invention in biological samples.
See, e.g., Harlow et al., Antibodies: A Laboratory Manual, (Cold Spring Harbor Laboratory _ _Press, 2nd ed. 1988) (incorporated by reference herein in its entirety).
As discussed in more detail below, the antibodies of the present invention may be used either alone or in combination with other compositions. The antibodies may further be recombinantly fused to a heterologous polypeptide at the N- or C-terminus or chemically conjugated (including covalently and non-covalently conjugations) to polypeptides or other compositions. For example, antibodies of the present invention may be recombinantly fused or conjugated to molecules useful as labels in detection assays and effector molecules such as heterologous polypeptides, drugs, radionuclides, or toxins. See, e.g., PCT publications WO 92/08495; WO
91/14438;
WO 89/12624; U.S. Patent No. 5,314,995; and EP 396,387.
The antibodies of the invention include derivatives that are modified, i.e, by the covalent attachment of any type of molecule to the antibody such that covalent attachment does not prevent the antibody from generating an anti-idiotypic response.
For example, but not by way of limitation, the antibody derivatives include antibodies that have been modified, e.g., by glycosylation, acetylation, pegylation, phosphylation, amidation, derivatization by known protecting/blocking groups, proteolytic cleavage, linkage to a cellular ligand or other protein, etc. Any of numerous chemical modifications may be carried out by known techniques, including, but not limited to specific chemical cleavage, acetylation, formylation, metabolic synthesis of tunicamycin, etc. Additionally, the derivative may contain one or more non-classical amino acids.
The antibodies of the present invention may be generated by any suitable method known in the art. Polyclonal antibodies to an antigen-of interest can be produced by various procedures well known in the art. For example, a polypeptide of the invention can be administered to various host animals including, but not limited to, rabbits, mice, rats, etc. to induce the production of sera containing polyclonal antibodies specific for the antigen. Various adjuvants may be used to increase the immunological response, depending on the host species, and include but are not limited to, Freund's (complete and incomplete), mineral gels such as aluminum hydroxide, surface active substances such as lysolecithin, pluronic polyols, polyanions, peptides, oil emulsions, keyhole limpet hemocyanins, dinitrophenol, and potentially useful human adjuvants such as BCG (bacille Calmette-Guerin) and corynebacterium parvum. Such adjuvants are also well known in the art.
Monoclonal antibodies can be prepared using a wide variety of techniques known in the art including the use of hybridoma, recombinant, and phage display technologies, or a combination thereof. For example, monoclonal antibodies can be produced using hybridoma techniques including those known in the art and taught, for example, in Harlow et al., Antibodies: A Laboratory Manual, (Cold Spring Harbor Laboratory Press, 2nd ed. 1988); Hammerling, et al., in: Monoclonal Antibodies and T-Cell Hybridomas 563-681 (Elsevier, N.Y., 1981) (said references incorporated by reference in their entireties). The term "monoclonal antibody" as used herein is not limited to antibodies produced through hybridoma technology. The term "monoclonal antibody" refers to an antibody that is derived from a single clone, including any eukaryotic, prokaryotic, or phage clone, and not the method by which it is produced.
Methods for producing and screening for specific antibodies using hybridoma technology are routine and well known in the art and are discussed in detail in the Examples (e.g., Example 16). In a non-limiting example, mice can be immunized with a polypeptide of the invention or a cell expressing such peptide. Once an immune response is detected, e.g., antibodies specific for the antigen are detected in the mouse serum, the mouse spleen is harvested and splenocytes isolated. The splenocytes are then fused by well known techniques to any suitable myeloma cells, for example cells from cell line SP20 available from the ATCC. Hybridomas are selected and cloned by limited dilution. The hybridoma clones are then assayed by methods known in the art for cells that secrete antibodies capable of binding a polypeptide of the invention. Ascites fluid, which generally contains high levels of antibodies, can be generated by immunizing mice with positive hybridoma clones.
Accordingly, the present invention provides methods of generating monoclonal antibodies as well as antibodies produced by the method comprising culturing a hybridoma cell secreting an antibody of the invention wherein, preferably, the hybridoma is generated by fusing splenocytes isolated from a mouse immunized with an antigen of the invention with myeloma cells and then screening the hybridomas resulting from the fusion for hybridoma clones that secrete an antibody able to bind a polypeptide of the invention.
- --Antibody fragments which recognize specific epitopes may be generated by known techniques. For example, Fab and F(ab')2 fragments of the invention may be produced by proteolytic cleavage of immunoglobulin molecules, using enzymes such as papain (to produce Fab fragments) or pepsin (to produce F(ab')2 fragments).
F(ab')2 fragments contain the variable region, the light chain constant region and the CH1 domain of the heavy chain.
For example, the antibodies of the present invention can also be generated using various phage display methods known in the art. In phage display methods, functional antibody domains are displayed on the surface of phage particles which carry the polynucleotide sequences encoding them. In a particular embodiment, such phage can be utilized to display antigen binding domains expressed from a repertoire or combinatorial antibody library (e.g., human or murine). Phage expressing an antigen binding domain that binds the antigen of interest can be selected or identified with antigen, e.g., using labeled antigen or antigen bound or captured to a solid surface or bead. Phage used in these methods are typically filamentous phage including fd and M13 binding domains expressed from phage with Fab, Fv or disulfide stabilized Fv antibody domains recombinantly fused to either the phage gene III or gene VIII protein. Examples of phage display methods that can be used to make the antibodies of the present invention include those disclosed in Brinkman et al., J. Immunol. Methods 182:41-50 (1995); Ames et al., J. Immunol. Methods 184:177-186 (1995); Kettleborough et al., Eur. J. Immunol. 24:952-958 (1994);
Persic et al., Gene 187 9-18 (1997); Burton et al., Advances in Immunology 57:191-280 (1994); PCT application No. PCT/GB91/01134; PCT publications WO 90/02809;
W0 91/10737; WO 92/01047; WO 92/18619; WO 93/11236; WO 95/15982; WO
95/20401; and U.S. Patent Nos. 5,698,426; 5,223,409; 5,403,484; 5,580,717;
5,427,908; 5,750,753; 5,821,047; 5,571,698; 5,427,908; 5,516,637; 5,780,225;
5,658,727; 5,733,743 and 5,969,108; each of which is incorporated herein by reference in its entirety.
As described in the above references, after phage selection, the antibody coding regions from the phage can be isolated and used to generate whole antibodies, including human antibodies, or any other desired antigen binding fragment, and expressed in any desired host, including mammalian cells, insect cells, plant cells, yeast, and bacteria, e.g., as described in detail below. For example, techniques to recombinantly produce Fab, Fab' and F(ab')2 fragments can also be employed using methods known in the art such as those disclosed in PCT publication WO
92/22324;
Mullinax et al., BioTechniques 12(6):864-869 (1992); and Sawai et al., AJRI
34:26-34 (1995); and Better et al., Science 240:1041-1043 (1988) (said references incorporated by reference in their entireties).
Examples of techniques which can be used to produce single-chain Fvs and antibodies include those described in U.S. Patents 4,946,778 and 5,258,498;
Huston et al., Methods in Enzymology 203:46-88 (1991); Shu et al., PNAS 90:7995-7999 (1993); and Skerra et al., Science 240:1038-1040 (1988). For some uses, including in vivo use of antibodies in humans and in vitro detection assays, it may be preferable to use chimeric, humanized, or human antibodies. A chimeric antibody is a molecule in which different portions of the antibody are derived from different animal species, such as antibodies having a variable region derived from a murine monoclonal antibody and a human immunoglobulin constant region. Methods for producing chimeric antibodies are known in the art. See e.g., Morrison, Science 229:1202 (1985); Oi et al., BioTechniques 4:214 (1986); Gillies et al., (1989) J.
Immunol.

Methods 125:191-202; U.S. Patent Nos. 5,807,715; 4,816,567; and 4,816397, which are incorporated herein by reference in their entirety. Humanized antibodies are antibody molecules from non-human species antibody that binds the desired antigen having one or more complementarity determining regions (CDRs) from the non-human species and a framework regions from a human immunoglobulin molecule.
Often, framework residues in the human framework regions will be substituted with the corresponding residue from the CDR donor antibody to alter, preferably improve, antigen binding. These framework substitutions are identified by methods well known in the art, e.g., by modeling of the interactions of the CDR and framework residues to identify framework residues important for antigen binding and sequence comparison to identify unusual framework residues at particular positions.
(See, e.g., Queen et al., U.S. Patent No. 5,585,089; Riechmann et al., Nature 332:323 (1988), which are incorporated herein by reference in their entireties.) Antibodies can be humanized using a variety of techniques known in the art including, for example, CDR-grafting (EP 239,400; PCT publication WO 91/09967; U.S. Patent Nos.
5,225,539; 5~53~O;TOT; -arid 5;585,089), veneering or resurfacing (EP 592,106;
EP
519,596; Padlan, Molecular Immunology 28(4/5):489-498 (1991); Studnicka et al., Protein Engineering 7(6):805-814 (1994); Roguska. et al.,-PNAS 91:969-973 (1994)), and chain shuffling (U.5. Patent No. 5,565,332).
Completely human antibodies are particularly desirable for therapeutic treatment of human patients. Human antibodies can be made by a variety of methods known in the art including phage display methods described above using antibody libraries derived from human immunoglobulin sequences. See also, U.S. Patent Nos.
4,444,887 and 4,716,111; and PCT publications WO 98/46645, WO 98/50433, WO
98/24893, WO 98/16654, WO 96/34096, WO 96/33735, and WO 91/10741; each of which is incorporated herein by reference in its entirety.
Human antibodies can also be produced using transgenic mice which are incapable of expressing functional endogenous immunoglobulins, but which can express human immunoglobulin genes. For example, the human heavy and light chain immunoglobulin gene complexes may be introduced randomly or by homologous recombination into mouse embryonic stem cells. Alternatively, the human variable region, constant region, and diversity region may be introduced into mouse embryonic stem cells in addition to the human heavy and light chain genes.
The mouse heavy and light chain immunoglobulin genes may be rendered non-functional separately or simultaneously with the introduction of human immunoglobulin loci by homologous recombination. In particular, homozygous deletion of the JH region prevents endogenous antibody production. The modified embryonic stem cells are expanded and microinjected into blastocysts to produce chimeric mice. The chimeric mice are then bred to produce homozygous offspring which express human antibodies. The transgenic mice are immunized in the normal fashion with a selected antigen, e.g., all or a portion of a polypeptide of the invention.
Monoclonal antibodies directed against the antigen can be obtained from the immunized, transgenic mice using conventional hybridoma technology. The human immunoglobulin transgenes harbored by the transgenic mice rearrange during B
cell differentiation, and subsequently undergo class switching and somatic mutation.
Thus, using such a technique, it is possible to produce therapeutically useful IgG, IgA, IgM and IgE antibodies. For an overview of this technology for producing human _antibodies, see_Lonberg and Huszar, Int._Rev. Immunol. 13:65-93 (1995). For a detailed discussion of this technology for producing human antibodies and human monoclonal antibodies and protocols for producing such antibodies, see, e.g., PCT
publications WO 98/24893; WO 92/01047; WO 96/34096; WO 96/33735; European Patent No. 0 598 877; U.S. Patent Nos. 5,413,923; 5,625,126; 5,633,425;
5,569,825;
5,661,016; 5,545,806; 5,814,318; 5,885,793; 5,916,771; and 5,939,598, which are incorporated by reference herein in their entirety. In addition, companies such as Abgenix, Inc. (Freemont, CA) and Genpharm (San Jose, CA) can be engaged to provide human antibodies directed against a selected antigen using technology similar to that described above.
Completely human antibodies which recognize a selected epitope can be generated using a technique referred to as "guided selection." In this approach a selected non-human monoclonal antibody, e.g., a mouse antibody, is used to guide the selection of a completely human antibody recognizing the same epitope.
(Jespers et al., Biotechnology 12:899-903 (1988)).
Further, antibodies to the polypeptides of the invention can, in turn, be utilized to generate anti-idiotype antibodies that "mimic" polypeptides of the invention using techniques well known to those skilled in the art. (See, e.g., Greenspan &
Bona, FASEB J. 7(5):437-444; (1989) and Nissinoff, J. Immunol. 147(8):2429-2438 (1991)). For example, antibodies which bind to and competitively inhibit polypeptide multimerization and/or binding of a polypeptide of the invention to a ligand can be used to generate anti-idiotypes that "mimic" the polypeptide multimerization and/or binding domain and, as a consequence, bind to and neutralize polypeptide and/or its ligand. Such neutralizing anti-idiotypes or Fab fragments of such anti-idiotypes can be used in therapeutic regimens to neutralize polypeptide ligand. For example, such anti-idiotypic antibodies can be used to bind a polypeptide of the invention and/or to bind its ligands/receptors, and thereby block its biological activity.
Polynucleotides Encoding Antibodies The invention further provides polynucleotides comprising a nucleotide sequence encoding an antibody of the invention and fragments thereof. The invention also encompasses polynucleotides that hybridize under stringent or lower stringency hybridization conditions, e.g., as defined supra, to polynucleotides that encode an antibody, preferably, that specifically binds to a polypeptide of the invention, preferably, an antibody that binds to a polypeptide having the amino acid sequence of SEQ 1D NO:Y.
The polynucleotides may be obtained, and the nucleotide sequence of the polynucleotides determined, by any method known in the art. For example, if the nucleotide sequence of the antibody is known, a polynucleotide encoding the antibody may be assembled from chemically synthesized oligonucleotides (e.g., as described in Kutmeier et al., BioTechniques 17:242 (1994)), which, briefly, involves the synthesis of overlapping oligonucleotides containing portions of the sequence encoding the antibody, annealing and ligating of those oligonucleotides, and then amplification of the ligated oligonucleotides by PCR.
Alternatively, a polynucleotide encoding an antibody may be generated from nucleic acid from a suitable source. If a clone containing a nucleic acid encoding a particular antibody is not available, but the sequence of the antibody molecule is known, a nucleic acid encoding the immunoglobulin may be chemically synthesized or obtained from a suitable source (e.g., an antibody cDNA library, or a cDNA
library generated from, or nucleic acid, preferably poly A+ RNA, isolated from, any tissue or cells expressing the antibody, such as hybridoma cells selected to express an antibody of the invention) by PCR amplification using synthetic primers hybridizable to the 3' and S' ends of the sequence or by cloning using an oligonucleotide probe specific for the particular gene sequence to identify, e.g., a cDNA clone from a cDNA library that encodes the antibody. Amplified nucleic acids generated by PCR
may then be cloned into replicable cloning vectors using any method well known in the art.
Once the nucleotide sequence and corresponding amino acid sequence of the antibody is determined, the nucleotide sequence of the antibody may be manipulated using methods well known in the art for the manipulation of nucleotide sequences, e.g., recombinant DNA techniques, site directed mutagenesis, PCR, etc. (see, for example, the techniques described in Sambrook et al., 1990, Molecular Cloning, A
Laboratory Manual, 2d Ed., Cold Spring Harbor Laboratory, Cold Spring Harbor, NY and Ausubel et al., eds., 1998, Current Protocols in Molecular Biology, John Wiley-& Sons, NY, which are both incorporated by reference herein in their entireties ), to generate antibodies having a different amino acid sequence, for example to create amino acid substitutions, deletions, and/or insertions.
In a specific embodiment, the amino acid sequence of the heavy and/or light chain variable domains may be inspected to identify the sequences of the complementarity determining regions (CDRs) by methods that are well know in the art, e.g., by comparison to known amino acid sequences of other heavy and light chain variable regions to determine the regions of sequence hypervariability.
Using routine recombinant DNA techniques, one or more of the CDRs may be inserted within framework regions, e.g., into human framework regions to humanize a non-human antibody, as described supra. The framework regions may be naturally occurring or consensus framework regions, and preferably human framework regions (see, e.g., Chothia et al., J. Mol. Biol. 278: 457-479 (1998) for a listing of human framework regions). Preferably, the polynucleotide generated by the combination of the framework regions and CDRs encodes an antibody that specifically binds a polypeptide of the invention. Preferably, as discussed supra, one or more amino acid substitutions may be made within the framework regions, and, preferably, the amino acid substitutions improve binding of the antibody to its antigen.
Additionally, such methods may be used to make amino acid substitutions or deletions of one or more variable region cysteine residues participating in an intrachain disulfide bond to generate antibody molecules lacking one or more intrachain disulfide bonds.
Other alterations to the polynucleotide are encompassed by the present invention and within the skill of the art.
In addition, techniques developed for the production of "chimeric antibodies"
(Mornson et al., Proc. Natl. Acad. Sci. 81:851-855 (1984); Neuberger et al., Nature 312:604-608 (1984); Takeda et al., Nature 314:452-454 (1985)) by splicing genes from a mouse antibody molecule of appropriate antigen specificity together with genes from a human antibody molecule of appropriate biological activity can be used.
As described supra, a chimeric antibody is a molecule in which different portions are derived from different animal species, such as those having a variable region derived from a murine mAb and a human immunoglobulin constant region, e.g., humanized antibodies.
-- - - - -Alternatively, techniques described for the production of single chain antibodies (U.5. Patent No. 4,946,778; Bird, Science 242:423- 42 (1988);
Huston et al., Proc. Natl. Acad. Sci. USA 85:5879-5883 (1988); and Ward et al., Nature 334:544-54 (1989)) can be adapted to produce single chain antibodies. Single chain antibodies are formed by linking the heavy and light chain fragments of the Fv region via an amino acid bridge, resulting in a single chain polypeptide. Techniques for the assembly of functional Fv fragments in E. coli may also be used (Skerra et al., Science 242:1038- 1041 (1988)).
Methods of Producing Antibodies The antibodies of the invention can be produced by any method known in the art for the synthesis of antibodies, in particular, by chemical synthesis or preferably, by recombinant expression techniques.
Recombinant expression of an antibody of the invention, or fragment, derivative or analog thereof, (e.g., a heavy or light chain of an antibody of the invention or a single chain antibody of the invention), requires construction of an expression vector containing a polynucleotide that encodes the antibody. Once a polynucleotide encoding an antibody molecule or a heavy or light chain of an antibody, or portion thereof (preferably containing the heavy or light chain variable domain), of the invention has been obtained, the vector for the production of the antibody molecule may be produced by recombinant DNA technology using techniques well known in the art. Thus, methods for preparing a protein by expressing a polynucleotide containing an antibody encoding nucleotide sequence are described herein. Methods which are well known to those skilled in the art can be used to construct expression vectors containing antibody coding sequences and appropriate transcriptional and translational control signals. These methods include, for example, in vitro recombinant DNA techniques, synthetic techniques, and in vivo genetic recombination. The invention, thus, provides replicable vectors comprising a nucleotide sequence encoding an antibody molecule of the invention, or a heavy or light chain thereof, or a heavy or light chain variable domain, operably linked to a promoter. Such vectors may include the nucleotide sequence encoding the constant region of the antibody molecule (see, e.g., PCT Publication WO 86/05807; PCT
Publication WO 89/01036; and U.S. Patent No. 5,122,464) and the variable domain of the antibody may be cloned into such a vector for expression of the entire heavy or light chain.
The expression vector is transferred to a host cell by conventional techniques and the transfected cells are then cultured by conventional techniques to produce an antibody of the invention. Thus, the invention includes host cells containing a polynucleotide encoding an antibody of the invention, or a heavy or light chain thereof, or a single chain antibody of the invention, operably linked to a heterologous promoter. In preferred embodiments for the expression of double-chained antibodies, vectors encoding both the heavy and light chains may be co-expressed in the host cell for expression of the entire immunoglobulin molecule, as detailed below.
A variety of host-expression vector systems may be utilized to express the antibody molecules of the invention. Such host-expression systems represent vehicles by which the coding sequences of interest may be produced and subsequently purified, but also represent cells which may, when transformed or transfected with the appropriate nucleotide coding sequences, express an antibody molecule of the invention in situ. These include but are not limited to microorganisms such as bacteria (e.g., E. coli, B. subtilis) transformed with recombinant bacteriophage DNA, plasmid DNA or cosmid DNA expression vectors containing antibody coding sequences; yeast (e.g., Saccharomyces, Pichia) transformed with recombinant yeast expression vectors containing antibody coding sequences; insect cell systems infected with recombinant virus expression vectors (e.g., baculovirus) containing antibody coding sequences; plant cell systems infected with recombinant virus expression vectors (e.g., cauliflower mosaic virus, CaMV; tobacco mosaic virus, TMV) or transformed with recombinant plasmid expression vectors (e.g., Ti plasmid) containing antibody coding sequences; or mammalian cell systems (e.g., COS, CHO, BHK, 293, 3T3 cells) harboring recombinant expression constructs containing promoters derived from the genome of mammalian cells (e.g., metallothionein promoter) or from mammalian viruses (e.g., the adenovirus late promoter; the vaccinia virus 7.5K promoter). Preferably, bacterial cells such as Escherichia coli, and more preferably, eukaryotic cells, especially for the expression of whole recombinant antibody molecule, are used for the expression of a recombinant antibody molecule. For example, mammalian cells such as Chinese hamster ovary cells (CHO), in conjunction with a vector such as the major intermediate early gene promoter element from human cytomegalovirus is an effective expression system for antibodies (Foecking et al., Gene 45:101 (1986); Cockett et al., Bio/Technology 8:2 ( 1990)).
In bacterial systems, a number of expression vectors may be advantageously selected depending upon the use intended for the antibody molecule being expressed.
For example, when a large quantity of such a protein is to be produced, for the generation of pharmaceutical compositions of an antibody molecule, vectors which direct the expression of high levels of fusion protein products that are readily purified may be desirable. Such vectors include, but are not limited, to the E. coli expression vector pUR278 (Ruther et al., EMBO J. 2:1791 (1983)), in which the antibody coding sequence may be ligated individually into the vector in frame with the lac Z
coding region so that a fusion protein is produced; pIN vectors (Inouye & Inouye, Nucleic Acids Res. 13:3101-3109 (1985); Van Heeke & Schuster, J. Biol. Chem. 24:5503-5509 (1989)); and the like. pGEX vectors may also be used to express foreign polypeptides as fusion proteins with glutathione S-transferase (GST). In general, such fusion proteins are soluble and can easily be purified from lysed cells by adsorption and binding to matrix glutathione-agarose beads followed by elution in the presence of free glutathione. The pGEX vectors are designed to include thrombin or factor Xa protease cleavage sites so that the cloned target gene product can be released from the S GST moiety.
In an insect system, Autographa californica nuclear polyhedrosis virus (AcNPV) is used as a vector to express foreign genes. The virus grows in Spodoptera frugiperda cells. The antibody coding sequence may be cloned individually into non-essential regions (for example the polyhedrin gene) of the virus and placed under control of an AcNPV promoter (for example the polyhedrin promoter).
In mammalian host cells, a number of viral-based expression systems may be utilized. In cases where an adenovirus is used as an expression vector, the antibody coding sequence of interest may be ligated to an adenovirus transcription/translation control complex, e.g., the late promoter and tripartite leader sequence. This chimeric - gene-may then be inserted in the aderiovirus genome by in vitro or in vivo recombination. Insertion in a non- essential region of the viral genome (e.g., region E1 or E3) will result in a recombinant virus that is viable and capable of expressing the antibody molecule in infected hosts. (e.g., see Logan & Shenk, Proc. Natl.
Acad.
Sci. USA 81:355-359 (1984)). Specific initiation signals may also be required for efficient translation of inserted antibody coding sequences. These signals include the ATG initiation codon and adjacent sequences. Furthermore, the initiation codon must be in phase with the reading frame of the desired coding sequence to ensure translation of the entire insert. These exogenous translational control signals and initiation codons can be of a variety of origins, both natural and synthetic.
The efficiency of expression may be enhanced by the inclusion of appropriate transcription enhancer elements, transcription terminators, etc. (see Bittner et al., Methods in Enzymol. 153:51-544 (1987)).
In addition, a host cell strain may be chosen which modulates the expression of the inserted sequences, or modifies and processes the gene product in the specific fashion desired. Such modifications (e.g., glycosylation) and processing (e.g., cleavage) of protein products may be important for the function of the protein.

Different host cells have characteristic and specific mechanisms for the post-translational processing and modification of proteins and gene products.
Appropriate cell lines or host systems can be chosen to ensure the correct modification and processing of the foreign protein expressed. To this end, eukaryotic host cells which possess the cellular machinery for proper processing of the primary transcript, glycosylation, and phosphorylation of the gene product may be used. Such mammalian host cells include but are not limited to CHO, VERY, BHK, Hela, COS, MDCK, 293, 3T3, WI38, and in particular, breast cancer cell lines such as, for example, BT483, Hs578T, HTB2, BT20 and T47D, and normal mammary gland cell line such as, for example, CRL7030 and Hs578Bst.
For long-term, high-yield production of recombinant proteins, stable expression is preferred. For example, cell lines which stably express the antibody molecule may be engineered. Rather than using expression vectors which contain viral origins of replication, host cells can be transformed with DNA
controlled by 1 S appropriate expression control elements (e.g., promoter, enhancer, sequences, transcription. terminators, polyadenylation sites, etc:);-and a selectable marker.
Following the introduction of the foreign DNA, engineered cells may be allowed to grow for 1-2 days in an enriched media, and then are switched to a selective media.
The selectable marker in the recombinant plasmid confers resistance to the selection and allows cells to stably integrate the plasmid into their chromosomes and grow to form foci which in turn can be cloned and expanded into cell lines. This method may advantageously be used to engineer cell lines which express the antibody molecule.
Such engineered cell lines may be particularly useful in screening and evaluation of compounds that interact directly or indirectly with the antibody molecule.
A number of selection systems may be used, including but not limited to the herpes simplex virus thymidine kinase (Wigler et al., Cell 11:223 (1977)), hypoxanthine-guanine phosphoribosyltransferase (Szybalska & Szybalski, Proc.
Natl.
Acad. Sci. USA 48:202 (1992)), and adenine phosphoribosyltransferase (Lowy et al., Cell 22:817 (1980)) genes can be employed in tk-, hgprt- or aprt- cells, respectively.
Also, antimetabolite resistance can be used as the basis of selection for the following genes: dhfr, which confers resistance to methotrexate (Wigler et al., Natl.
Acad. Sci.
USA 77:357 (1980); O'Hare et al., Proc. Natl. Acad. Sci. USA 78:1527 (1981));
gpt, which confers resistance to mycophenolic acid (Mulligan & Berg, Proc. Natl.
Acad.
Sci. USA 78:2072 (1981)); neo, which confers resistance to the aminoglycoside G-418 Clinical Pharmacy 12:488-505; Wu and Wu, Biotherapy 3:87-95 (1991);
Tolstoshev, Ann. Rev. Pharmacol. Toxicol. 32:573-596 (1993); Mulligan, Science 260:926-932 (1993); and Morgan and Anderson, Ann. Rev. Biochem. 62:191-217 (1993); May, 1993, TIB TECH 11(5):155-215); and hygro, which confers resistance to hygromycin (Santerre et al., Gene 30:147 (1984)). Methods commonly known in the art of recombinant DNA technology may be routinely applied to select the desired recombinant clone, and such methods are described, for example, in Ausubel et al.
(eds.), Current Protocols in Molecular Biology, John Wiley & Sons, NY (1993);
Kriegler, Gene Transfer and Expression, A Laboratory Manual, Stockton Press, NY
(1990); and in Chapters 12 and 13, Dracopoli et al. (eds), Current Protocols in Human Genetics, John Wiley & Sons, NY (1994); Colberre-Garapin et al., J. Mol.
Biol. 150:1 (1981), which are incorporated by reference herein in their entireties.
The expression levels of an antibody molecule can be increased by vector _-amplification (for a review,=see Bebbington and Hentschel, The use of vectors based on gene amplification for the expression of cloned genes in mammalian cells in DNA
cloning, Vol.3. (Academic Press, New York, 1987)). When a marker in the vector system expressing antibody is amplifiable, increase in the level of inhibitor present in culture of host cell will increase the number of copies of the marker gene.
Since the amplified region is associated with the antibody gene, production of the antibody will also increase (Grouse et al., Mol. Cell. Biol. 3:257 (1983)).
The host cell may be co-transfected with two expression vectors of the invention, the first vector encoding a heavy chain derived polypeptide and the second vector encoding a light chain derived polypeptide. The two vectors may contain identical selectable markers which enable equal expression of heavy and light chain polypeptides. Alternatively, a single vector may be used which encodes, and is capable of expressing, both heavy and light chain polypeptides. In such situations, the light chain should be placed before the heavy chain to avoid an excess of toxic free heavy chain (Proudfoot, Nature 322:52 (1986); Kohler, Proc. Natl. Acad.
Sci.
USA 77:2197 (1980)). The coding sequences for the heavy and light chains may comprise cDNA or genomic DNA.

Once an antibody molecule of the invention has been produced by an animal, chemically synthesized, or recombinantly expressed, it may be purified by any method known in the art for purification of an immunoglobulin molecule, for example, by chromatography (e.g., ion exchange, affinity, particularly by affinity for the specific antigen after Protein A, and sizing column chromatography), centrifugation, differential solubility, or by any other standard technique for the purification of proteins. In addition, the antibodies of the present invention or fragments thereof can be fused to heterologous polypeptide sequences described herein or otherwise known in the art, to facilitate purification.
The present invention encompasses antibodies recombinantly fused or chemically conjugated (including both covalently and non-covalently conjugations) to a polypeptide (or portion thereof, preferably at least 10, 20, 30, 40, 50, 60, 70, 80, 90 or 100 amino acids of the polypeptide) of the present invention to generate fusion proteins. The fusion does not necessarily need to be direct, but may occur through linker sequences. The antibodies may be specific for antigens other than polypeptides _(or_portion thereof, preferably at least 10;-20, 30, 40, 50, 60, 70, 80, 90 or 100 amino acids of the polypeptide) of the present invention. For example, antibodies may be used to target the polypeptides of the present invention to particular cell types, either in vitro or in vivo, by fusing or conjugating the polypeptides of the present invention to antibodies specific for particular cell surface receptors. Antibodies fused or conjugated to the polypeptides of the present invention may also be used in in vitro immunoassays and purification methods using methods known in the art. See e.g., Harbor et al., supra, and PCT publication WO 93/21232; EP 439,095; Naramura et al., Immunol. Lett. 39:91-99 (1994); U.S. Patent 5,474,981; Gillies et al., PNAS
89:1428-1432 (1992); Fell et al., J. Immunol. 146:2446-2452(1991), which are incorporated by reference in their entireties.
The present invention further includes compositions comprising the polypeptides of the present invention fused or conjugated to antibody domains other than the variable regions. For example, the polypeptides of the present invention may be fused or conjugated to an antibody Fc region, or portion thereof. The antibody portion fused to a polypeptide of the present invention may comprise the constant region, hinge region, CH1 domain, CH2 domain, and CH3 domain or any combination of whole domains or portions thereof. The polypeptides may also be fused or conjugated to the above antibody portions to form multimers. For example, Fc portions fused to the polypeptides of the present invention can form dimers through disulfide bonding between the Fc portions. Higher multimeric forms can be made by fusing the polypeptides to portions of IgA and IgM. Methods for fusing or conjugating the polypeptides of the present invention to antibody portions are known in the art. See, e.g., U.S. Patent Nos. 5,336,603; 5,622,929; 5,359,046;
5,349,053;
5,447,851; 5,112,946; EP 307,434; EP 367,166; PCT publications WO 96/04388; WO
91/06570; Ashkenazi et al., Proc. Natl. Acad. Sci. USA 88:10535-10539 (1991);
Zheng et al., J. Immunol. 154:5590-5600 (1995); and Vil et al., Proc. Natl.
Acad. Sci.
USA 89:11337- 11341(1992) (said references incorporated by reference in their entireties).
As discussed, supra, the polypeptides corresponding to a polypeptide, polypeptide fragment, or a variant of SEQ ID NO:Y may be fused or conjugated to the above antibody portions to increase the in vivo half life of the polypeptides or for use in immunoassays using methods known in the art. Further, the polypeptides corresponding to SEQ ID NO:Y may be fused or conjugated to the above antibody portions to facilitate purification. One reported example describes chimeric proteins consisting of the first two domains of the human CD4-polypeptide and various domains of the constant regions of the heavy or light chains of mammalian immunoglobulins. (EP 394,827; Traunecker et al., Nature 331:84-86 (1988). The polypeptides of the present invention fused or conjugated to an antibody having disulfide- linked dimeric structures (due to the IgG) may also be more efficient in binding and neutralizing other molecules, than the monomeric secreted protein or protein fragment alone. (Fountoulakis et al., J. Biochem. 270:3958-3964 (1995)). In many cases, the Fc part in a fusion protein is beneficial in therapy and diagnosis, and thus can result in, for example, improved pharmacokinetic properties. (EP A
232,262). Alternatively, deleting the Fc part after the fusion protein has been expressed, detected, and purified, would be desired. For example, the Fc portion may hinder therapy and diagnosis if the fusion protein is used as an antigen for immunizations. In drug discovery, for example, human proteins, such as hIL-5, have been fused with Fc portions for the purpose of high-throughput screening assays to identify antagonists of hIL-5. (See, Bennett et al., J. Molecular Recognition 8:52-58 (1995); Johanson et al., J. Biol. Chem. 270:9459-9471 (1995).
Moreover, the antibodies or fragments thereof of the present invention can be fused to marker sequences, such as a peptide to facilitate purification. In preferred embodiments, the marker amino acid sequence is a hexa-histidine peptide, such as the tag provided in a pQE vector (QIAGEN, Inc., 9259 Eton Avenue, Chatsworth, CA, 91311), among others, many of which are commercially available. As described in Gentz et al., Proc. Natl. Acad. Sci. USA 86:821-824 (1989), for instance, hexa-histidine provides for convenient purification of the fusion protein. Other peptide tags useful for purification include, but are not limited to, the "HA" tag, which corresponds to an epitope derived from the influenza hemagglutinin protein (Wilson et al., Cell 37:767 (1984)) and the "flag" tag.
The present invention further encompasses antibodies or fragments thereof conjugated to a diagnostic or therapeutic agent. The antibodies can be used diagnostically to, for example, monitor the development or progression of a tumor as _part of a clinical testing procedure to, e.g., determine the efficacy of a given treatment regimen. Detection can be facilitated by coupling the antibody to a detectable substance. Examples of detectable substances include various enzymes, prosthetic groups, fluorescent materials, luminescent materials, bioluminescent materials, radioactive materials, positron emitting metals using various positron emission tomographies, and nonradioactive paramagnetic metal ions. The detectable substance may be coupled or conjugated either directly to the antibody (or fragment thereof) or indirectly, through an intermediate (such as, for example, a linker known in the art) using techniques known in the art. See, for example, U.S. Patent No.
4,741,900 for metal ions which can be conjugated to antibodies for use as diagnostics according to the present invention. Examples of suitable enzymes include horseradish peroxidase, alkaline phosphatase, beta-galactosidase, or acetylcholinesterase;
examples of suitable prosthetic group complexes include streptavidin/biotin and avidin/biotin; examples of suitable fluorescent materials include umbelliferone, fluorescein, fluorescein isothiocyanate, rhodamine, dichlorotriazinylamine fluorescein, dansyl chloride or phycoerythrin; an example of a luminescent material includes luminol; examples of bioluminescent materials include luciferase, luciferin, and aequorin; and examples of suitable radioactive material include 125I, 131I, 11 lIn or 99Tc.
Further, an antibody or fragment thereof may be conjugated to a therapeutic moiety such as a cytotoxin, e.g., a cytostatic or cytocidal agent, a therapeutic agent or a radioactive metal ion, e.g., alpha-emitters such as, for example, 213Bi. A
cytotoxin or cytotoxic agent includes any agent that is detrimental to cells. Examples include paclitaxol, cytochalasin B, gramicidin D, ethidium bromide, emetine, mitomycin, etoposide, tenoposide, vincristine, vinblastine, colchicin, doxorubicin, daunorubicin, dihydroxy anthracin dione, mitoxantrone, mithramycin, actinomycin D, 1-dehydrotestosterone, glucocorticoids, procaine, tetracaine, lidocaine, propranolol, and puromycin and analogs or homologs thereof. Therapeutic agents include, but are not limited to, antimetabolites (e.g., methotrexate, 6-mercaptopurine, 6-thioguanine, cytarabine, 5-fluorouracil decarbazine), alkylating agents (e.g., mechlorethamine, thioepa chlorambucil, melphalan, carmustine (BSNU) and lomustine (CCNU), 1 S cyclothosphamide, busulfan, dibromomannitol, streptozotocin, mitomycin C, and cis-dichlorodiamine platinum (II) (DDP) cisplatin), anthracyclines (e.g., daunorubicin (formerly daunomycin) and doxorubicin), antibiotics (e.g., dactinomycin (formerly actinomycin), bleomycin, mithramycin, and anthramycin (AMC)), and anti-mitotic agents (e.g., vincristine and vinblastine).
The conjugates of the invention can be used for modifying a given biological response, the therapeutic agent or drug moiety is not to be construed as limited to classical chemical therapeutic agents. For example, the drug moiety may be a protein or polypeptide possessing a desired biological activity. Such proteins may include, for example, a toxin such as abrin, ricin A, pseudomonas exotoxin, or diphtheria toxin; a protein such as tumor necrosis factor, a-interferon, 13-interferon, nerve growth factor, platelet derived growth factor, tissue plasminogen activator, an apoptotic agent, e.g., TNF-alpha, TNF-beta, AIM I (See, International Publication No. WO
97/33899), AIM II (See, International Publication No. WO 97/34911), Fas Ligand (Takahashi et al., Int. Immunol., 6:1567-1574 (1994)), VEGI (See, International Publication No. WO 99/23105), a thrombotic agent or. an anti- angiogenic agent, e.g., angiostatin or endostatin; or, biological response modifiers such as, for example, lymphokines, interleukin-1 ("IL-1 "), interleukin-2 ("IL-2"), interleukin-6 ("IL-6"), granulocyte macrophage colony stimulating factor ("GM-CSF"), granulocyte colony stimulating factor ("G-CSF"), or other growth factors.
Antibodies may also be attached to solid supports, which are particularly useful for immunoassays or purification of the target antigen. Such solid supports include, but are not limited to, glass, cellulose, polyacrylamide, nylon, polystyrene, polyvinyl chloride or polypropylene.
Techniques for conjugating such therapeutic moiety to antibodies are well known, see, e.g., Arnon et al., "Monoclonal Antibodies For Immunotargeting Of Drugs In Cancer Therapy", in Monoclonal Antibodies And Cancer Therapy, Reisfeld et al. (eds.), pp. 243-56 (Alan R. Liss, Inc. 1985); Hellstrom et al., "Antibodies For Drug Delivery", in Controlled Drug Delivery (2nd Ed.), Robinson et al. (eds.), pp.
623-53 (Marcel Dekker, Inc. 1987); Thorpe, "Antibody Garners Of Cytotoxic Agents In Cancer Therapy: A Review", in Monoclonal Antibodies '84: Biological And Clinical Applications, Pinchera et al. (eds.), pp. 475-506 (1985); "Analysis, Results, And Future Prospective Of The Therapeutic Use Of Radiolabeled Antibody In Cancer Therapy", in Monoclonal Antibodies For Cancer Detection And Therapy, Baldwin et al. (eds.), pp. 303-16 (Academic Press 1985), and Thorpe et al., "The Preparation And Cytotoxic Properties Of Antibody-Toxin Conjugates", Immunol.
Rev. 62:119-58 (1982).
Alternatively, an antibody can be conjugated to a second antibody to form an antibody heteroconjugate as described by Segal in U.S. Patent No. 4,676,980, which is incorporated herein by reference in its entirety.
An antibody, with or without a therapeutic moiety conjugated to it, administered alone or in combination with cytotoxic factors) and/or cytokine(s) can be used as a therapeutic.
Immunophenotyping The antibodies of the invention may be utilized for immunophenotyping of cell lines and biological samples. The translation product of the gene of the present invention may be useful as a cell specific marker, or more specifically as a cellular marker that is differentially expressed at various stages of differentiation and/or maturation of particular cell types. Monoclonal antibodies directed against a specific epitope, or combination of epitopes, will allow for the screening of cellular populations expressing the marker. Various techniques can be utilized using monoclonal antibodies to screen for cellular populations expressing the marker(s), and include magnetic separation using antibody-coated magnetic beads, "panning"
with antibody attached to a solid matrix (i.e., plate), and flow cytometry (See, e.g., U.S.
Patent 5,985,660; and Morrison et al., Cell, 96:737-49 (1999)).
These techniques allow for the screening of particular populations of cells, such as might be found with hematological malignancies (i.e. minimal residual disease (MRD) in acute leukemic patients) and "non-self' cells in transplantations to prevent Graft-versus-Host Disease (GVHD). Alternatively, these techniques allow for the screening of hematopoietic stem and progenitor cells capable of undergoing proliferation and/or differentiation, as might be found in human umbilical cord blood.
Assays For Antibody Binding The antibodies of the invention may be assayed for immunospecific binding by any method known in the art. The immunoassays which can be used include but are not limited to competitive and non-competitive assay systems using techniques such as western blots, radioimmunoassays, ELISA (enzyme linked immunosorbent assay), "sandwich" immunoassays, immunoprecipitation assays, precipitin reactions, gel diffusion precipitin reactions, iinmunodiffusion assays, agglutination assays, complement-fixation assays, immunoradiometric assays, fluorescent immunoassays, protein A immunoassays, to name but a few. Such assays are routine and well known in the art (see, e.g., Ausubel et al, eds, 1994, Current Protocols in Molecular Biology, Vol. 1, John Wiley & Sons, Inc., New York, which is incorporated by reference herein in its entirety). Exemplary immunoassays are described briefly below (but are not intended by way of limitation).
Immunoprecipitation protocols generally comprise lysing a population of cells in a lysis buffer such as RIPA buffer (1 % NP-40 or Triton X- 100, 1 % sodium .
deoxycholate, 0.1% SDS, 0.15 M NaCI, 0.01 M sodium phosphate at pH 7.2, 1%
Trasylol) supplemented with protein phosphatase and/or protease inhibitors (e.g., EDTA, PMSF, aprotinin, sodium vanadate), adding the antibody of interest to the cell lysate, incubating for a period of time (e.g., 1-4 hours) at 4° C, adding protein A

and/or protein G sepharose beads to the cell lysate, incubating for about an hour or more at 4° C, washing the beads in lysis buffer and resuspending the beads in SDS/sample buffer. The ability of the antibody of interest to immunoprecipitate a particular antigen can be assessed by, e.g., western blot analysis. One of skill in the art would be knowledgeable as to the parameters that can be modified to increase the binding of the antibody to an antigen and decrease the background (e.g., pre-clearing the cell lysate with sepharose beads). For further discussion regarding immunoprecipitation protocols see, e.g., Ausubel et al, eds, 1994, Current Protocols in Molecular Biology, Vol. l, John Wiley & Sons, Inc., New York at 10.16.1.
Western blot analysis generally comprises preparing protein samples, electrophoresis of the protein samples in a polyacrylamide gel (e.g., 8%- 20%
SDS-PAGE depending on the molecular weight of the antigen), transferring the protein sample from the polyacrylamide gel to a membrane such as nitrocellulose, PVDF
or nylon, blocking the membrane in blocking solution (e.g., PBS with 3% BSA or non-1 S fat milk), washing the membrane in washing buffer (e.g., PBS-Tween 20), blocking the membrane with primary antibody (the antibody of interest) diluted in blocking buffer, washing the membrane in washing buffer, blocking the membrane with a secondary antibody (which recognizes the primary antibody, e.g., an anti-human antibody) conjugated to an enzymatic substrate (e.g., horseradish peroxidase or alkaline phosphatase) or radioactive molecule (e.g., 32P or 125I) diluted in blocking buffer, washing the membrane in wash buffer, and detecting the presence of the antigen. One of skill in the art would be knowledgeable as to the parameters that can be modified to increase the signal detected and to reduce the background noise. For further discussion regarding western blot protocols see, e.g., Ausubel et al, eds, 1994, Current Protocols in Molecular Biology, Vol. 1, John Wiley & Sons, Inc., New York at 10.8.1.
ELISAs comprise preparing antigen, coating the well of a 96 well microtiter plate with the antigen, adding the antibody of interest conjugated to a detectable compound such as an enzymatic substrate (e.g., horseradish peroxidase or alkaline phosphatase) to the well and incubating for a period of time, and detecting the presence of the antigen. In ELISAs the antibody of interest does not have to be conjugated to a detectable compound; instead, a second antibody (which recognizes the antibody of interest) conjugated to a detectable compound may be added to the well. Further, instead of coating the well with the antigen, the antibody may be coated to the well. In this case, a second antibody conjugated to a detectable compound may be added following the addition of the antigen of interest to the coated well. One of skill in the art would be knowledgeable as to the parameters that can be modified to increase the signal detected as well as other variations of ELISAs known in the art. For further discussion regarding ELISAs see, e.g., Ausubel et al, eds, 1994, Current Protocols in Molecular Biology, Vol. 1, John Wiley & Sons, Inc., New York at 11.2.1.
The binding affinity of an antibody to an antigen and the off rate of an antibody-antigen interaction can be determined by competitive binding assays.
One example of a competitive binding assay is a radioimmunoassay comprising the incubation of labeled antigen (e.g., 3H or 125I) with the antibody of interest in the presence of increasing amounts of unlabeled antigen, and the detection of the antibody bound to the labeled antigen. The affinity of the antibody of interest for a particular antigen and the binding off rates can be determined from the data by scatchard plot analysis. Competition with a second antibody can also be determined using radioimmunoassays. In this case, the antigen is incubated with antibody of interest conjugated to a labeled compound (e.g., 3H or 125I) in the presence of increasing amounts of an unlabeled second antibody.
Therapeutic Uses The present invention is further directed to antibody-based therapies which involve administering antibodies of the invention to an animal, preferably a mammal, and most preferably a human, patient for treating one or more of the disclosed diseases, disorders, or conditions. Therapeutic compounds of the invention include, but are not limited to, antibodies of the invention (including fragments, analogs and derivatives thereof as described herein) and nucleic acids encoding antibodies of the invention (including fragments, analogs and derivatives thereof and anti-idiotypic antibodies as described herein). The antibodies of the invention can be used to treat, inhibit or prevent diseases, disorders or conditions associated with aberrant expression and/or activity of a polypeptide of the invention, including, but not limited to, any one or more of the diseases, disorders, or conditions described herein. The treatment and/or prevention of diseases, disorders, or conditions associated with aberrant expression and/or activity of a polypeptide of the invention includes, but is not limited to, alleviating symptoms associated with those diseases, disorders or conditions. Antibodies of the invention may be provided in pharmaceutically acceptable compositions as known in the art or as described herein.
A summary of the ways in which the antibodies of the present invention may be used therapeutically includes binding polynucleotides or polypeptides of the present invention locally or systemically in the body or by direct cytotoxicity of the antibody, e.g. as mediated by complement (CDC) or by effector cells (ADCC).
Some of these approaches are described in more detail below. Armed with the teachings provided herein, one of ordinary skill in the art will know how to use the antibodies of the present invention for diagnostic, monitoring or therapeutic purposes without undue experimentation.
The antibodies of this invention may be advantageously utilized in combination with other monoclonal or chimeric antibodies, or with lymphokines or hematopoietic growth factors (such as, e.g., IL-2, IL-3 and IL-7), for example, which serve to increase the number or activity of effector cells which interact with the antibodies.
The antibodies of the invention may be administered alone or in combination with other types of treatments (e.g., radiation therapy, chemotherapy, hormonal therapy, immunotherapy and anti-tumor agents). Generally, administration of products of a species origin or species reactivity (in the case of antibodies) that is the same species as that of the patient is preferred. Thus, in a preferred embodiment, human antibodies, fragments derivatives, analogs, or nucleic acids, are administered to a human patient for therapy or prophylaxis.
It is preferred to use high affinity and/or potent in vivo inhibiting and/or neutralizing antibodies against polypeptides or polynucleotides of the present invention, fragments or regions thereof, for both immunoassays directed to and therapy of disorders related to polynucleotides or polypeptides, including fragments thereof, of the present invention. Such antibodies, fragments, or regions, will preferably have an affinity for polynucleotides or polypeptides of the invention, including fragments thereof. Preferred binding affinities include those with a dissociation constant or Kd less than 5 X 10-Z M, 10-2 M, 5 X 10-3 M, 10-3 M, 4 M, 10-4 M, S X 10-5 M, 10-5 M, 5 X 10-~ M, 10-~ M, 5 X 10-~ M, 10-7 M, S X
10-g M, 10-8 M, 5 X 10-~ M, 10-9 M, 5 X 10-' ° M, 10-' ° M, 5 X 10-" M, 1 O-" M, 5 X 10-' Z M, 10-' 2 M, 5 X 10-' 3 M, 10-' 3 M, 5 X 10-' 4 M, 10-' 4 M, 5 X 10-' S M, and 10-' S M.
Gene Therapy In a specific embodiment, nucleic acids comprising sequences encoding antibodies or functional derivatives thereof, are administered to treat, inhibit or prevent a disease or disorder associated with aberrant expression and/or activity of a polypeptide of the invention, by way of gene therapy. Gene therapy refers to therapy performed by the administration to a subject of an expressed or expressible nucleic acid. In this embodiment of the invention, the nucleic acids produce their encoded protein that mediates a therapeutic effect.
Any of the methods for gene therapy available in the art can be used according to the present invention. Exemplary methods are described below.
For general reviews of the methods of gene therapy, see Goldspiel et al., Clinical Pharmacy 12:488-505 (1993); Wu and Wu, Biotherapy 3:87-95 (1991);
Tolstoshev, Ann. Rev. Pharmacol. Toxicol. 32:573-596 (1993); Mulligan, Science 260:926-932 (1993); and Morgan and Anderson, Ann. Rev. Biochem. 62:191-217 (1993); May, TIBTECH 11(5):155-215 (1993). Methods commonly known in the art of recombinant DNA technology which can be used are described in Ausubel et al.
(eds.), Current Protocols in Molecular Biology, John Wiley & Sons, NY (1993);
and Kriegler, Gene Transfer and Expression, A Laboratory Manual, Stockton Press, NY
(1990).
In a preferred aspect, the compound comprises nucleic acid sequences encoding an antibody, said nucleic acid sequences being part of expression vectors that express the antibody or fragments or chimeric proteins or heavy or light chains thereof in a suitable host. In particular, such nucleic acid sequences have promoters operably linked to the antibody coding region, said promoter being inducible or constitutive, and, optionally, tissue- specific. In another particular embodiment, nucleic acid molecules are used in which the antibody coding sequences and any other desired sequences are flanked by regions that promote homologous recombination at a desired site in the genome, thus providing for intrachromosomal expression of the antibody encoding nucleic acids (Koller and Smithies, Proc. Natl. Acad. Sci.
USA
86:8932-8935 (1989); Zijlstra et al., Nature 342:435-438 (1989). In specific S embodiments, the expressed antibody molecule is a single chain antibody;
alternatively, the nucleic acid sequences include sequences encoding both the heavy and light chains, or fragments thereof, of the antibody.
Delivery of the nucleic acids into a patient may be either direct, in which case the patient is directly exposed to the nucleic acid or nucleic acid- carrying vectors, or indirect, in which case, cells are first transformed with the nucleic acids in vitro, then transplanted into the patient. These two approaches are known, respectively, as in vivo or ex vivo gene therapy.
In a specific embodiment, the nucleic acid sequences are directly administered in vivo, where it is expressed to produce the encoded product. This can be accomplished by any of numerous methods known in the art, e.g., by constructing them as part of an appropriate nucleic acid expression vector and administering it so that they become intracellular, e.g., by infection using defective or attenuated retrovirals or other viral vectors (see U.S. Patent No. 4,980,286), or by direct injection of naked DNA, or by use of microparticle bombardment (e.g., a gene gun;
Biolistic, Dupont), or coating with lipids or cell-surface receptors or transfecting agents, encapsulation in liposomes, microparticles, or microcapsules, or by administering them in linkage to a peptide which is known to enter the nucleus, by administering it in linkage to a ligand subject to receptor-mediated endocytosis (see, e.g., Wu and Wu, J. Biol. Chem. 262:4429-4432 (1987)) (which can be used to target cell types specifically expressing the receptors), etc. In another embodiment, nucleic acid-ligand complexes can be formed in which the ligand comprises a fusogenic viral peptide to disrupt endosomes, allowing the nucleic acid to avoid lysosomal degradation. In yet another embodiment, the nucleic acid can be targeted in vivo for cell specific uptake and expression, by targeting a specific receptor (see, e.g., PCT
Publications WO 92/06180; WO 92/22635; W092/20316; W093/14188, WO
93/20221 ). Alternatively, the nucleic acid can be introduced intracellularly and incorporated within host cell DNA for expression, by homologous recombination (Koller and Smithies, Proc. Natl. Acad. Sci. USA 86:8932-8935 (1989); Zijlstra et al., Nature 342:435-438 (1989)).
In a specific embodiment, viral vectors that contains nucleic acid sequences encoding an antibody of the invention are used. For example, a retroviral vector can be used (see Miller et al., Meth. Enzymol. 217:581-599 (1993)). These retroviral vectors contain the components necessary for the correct packaging of the viral genome and integration into the host cell DNA. The nucleic acid sequences encoding the antibody to be used in gene therapy are cloned into one or more vectors, which facilitates delivery of the gene into a patient. More detail about retroviral vectors can be found in Boesen et al., Biotherapy 6:291-302 (1994), which describes the use of a retroviral vector to deliver the mdrl gene to hematopoietic stem cells in order to make the stem cells more resistant to chemotherapy. Other references illustrating the use of retroviral vectors in gene therapy are: Clowes et al., J. Clin. Invest.
93:644-651 (1994); Kiem et al., Blood 83:1467-1473 (1994); Salmons and Gunzberg, Human Gene Therapy 4:129-141 (1993); and Grossman and Wilson, Curr. Opin. in Genetics and Devel. 3:110-114 (1993).
Adenoviruses are other viral vectors that can be used in gene therapy.
Adenoviruses are especially attractive vehicles for delivering genes to respiratory epithelia. Adenoviruses naturally infect respiratory epithelia where they cause a mild disease. Other targets for adenovirus-based delivery systems are liver, the central nervous system, endothelial cells, and muscle. Adenoviruses have the advantage of being capable of infecting non-dividing cells. Kozarsky and Wilson, Current Opinion in Genetics and Development 3:499-503 (1993) present a review of adenovirus-based gene therapy. Bout et al., Human Gene Therapy 5:3-10 (1994) demonstrated the use of adenovirus vectors to transfer genes to the respiratory epithelia of rhesus monkeys. Other instances of the use of adenoviruses in gene therapy can be found in Rosenfeld et al., Science 252:431-434 (1991);
Rosenfeld et al., Cell 68:143- 155 (1992); Mastrangeli et al., J. Clin. Invest. 91:225-234 (1993);
PCT Publication W094/12649; and Wang, et al., Gene Therapy 2:775-783 (1995).
In a preferred embodiment, adenovirus vectors are used.

Adeno-associated virus (AAV) has also been proposed for use in gene therapy (Walsh et al., Proc. Soc. Exp. Biol. Med. 204:289-300 (1993); U.S. Patent No.
5,436,146).
Another approach to gene therapy involves transfernng a gene to cells in tissue culture by such methods as electroporation, lipofection, calcium phosphate mediated transfection, or viral infection. Usually, the method of transfer includes the transfer of a selectable marker to the cells. The cells are then placed under selection to isolate those cells that have taken up and are expressing the transferred gene.
Those cells are then delivered to a patient.
In this embodiment, the nucleic acid is introduced into a cell prior to administration in vivo of the resulting recombinant cell. Such introduction can be carried out by any method known in the art, including but not limited to transfection, electroporation, microinjection, infection with a viral or bacteriophage vector containing the nucleic acid sequences, cell fusion, chromosome-mediated gene transfer, microcell-mediated gene transfer, spheroplast fusion, etc. Numerous techniques are known in the art for the introduction of foreign genes into cells (see, e.g., Loeffler and Behr, Meth. Enzymol. 217:599-618 (1993); Cohen et al., Meth.
Enzymol. 217:618-644 (1993); Cline, Pharmac. Ther. 29:69-92m (1985) and may be used in accordance with the present invention, provided that the necessary developmental and physiological functions of the recipient cells are not disrupted.
The technique should provide for the stable transfer of the nucleic acid to the cell, so that the nucleic acid is expressible by the cell and preferably heritable and expressible by its cell progeny.
The resulting recombinant cells can be delivered to a patient by various methods known in the art. Recombinant blood cells (e.g., hematopoietic stem or progenitor cells) are preferably administered intravenously. The amount of cells envisioned for use depends on the desired effect, patient state, etc., and can be determined by one skilled in the art.
Cells into which a nucleic acid can be introduced for purposes of gene therapy encompass any desired, available cell type, and include but are not limited to epithelial cells, endothelial cells, keratinocytes, fibroblasts, muscle cells, hepatocytes;
blood cells such as Tlymphocytes"Blymphocytes, monocytes, macrophages, neutrophils, eosinophils, megakaryocytes, granulocytes; various stem or progenitor cells, in particular hematopoietic stem or progenitor cells, e.g., as obtained from bone marrow, umbilical cord blood, peripheral blood, fetal liver, etc.
In a preferred embodiment, the cell used for gene therapy is autologous to the patient.
In an embodiment in which recombinant cells are used in gene therapy, nucleic acid sequences encoding an antibody are introduced into the cells such that they are expressible by the cells or their progeny, and the recombinant cells are then administered in vivo for therapeutic effect. In a specific embodiment, stem or progenitor cells are used. Any stem and/or progenitor cells which can be isolated and maintained in vitro can potentially be used in accordance with this embodiment of the present invention (see e.g. PCT Publication WO 94/08598; Stemple and Anderson, Cell 71:973-985 (1992); Rheinwald, Meth. Cell Bio. 21A:229 (1980);
and Pittelkow and Scott, Mayo Clinic Proc. 61:771 (1986)).
In a specific embodiment, the nucleic acid to be introduced for purposes of gene therapy comprises an inducible promoter operably linked to the coding region, such that expression of the nucleic acid is controllable by controlling the presence or absence of the appropriate inducer of transcription.
Demonstration of Therapeutic or Prophylactic Activity The compounds or pharmaceutical compositions of the invention are preferably tested in vitro, and then in vivo for the desired therapeutic or prophylactic activity, prior to use in humans. For example, in vitro assays to demonstrate the therapeutic or prophylactic utility of a compound or pharmaceutical composition include, the effect of a compound on a cell line or a patient tissue sample.
The effect of the compound or composition on the cell line and/or tissue sample can be determined utilizing techniques known to those of skill in the art including, but not limited to, rosette formation assays and cell lysis assays. In accordance with the invention, in vitro assays which can be used to determine whether administration of a specific compound is indicated, include in vitro cell culture assays in which a patient tissue sample is grown in culture, and exposed to or otherwise administered a compound, and the effect of such compound upon the tissue sample is observed.

TherapeuticlProphylactic Administration and Composition The invention provides methods of treatment, inhibition and prophylaxis by administration to a subject of an effective amount of a compound or pharmaceutical composition of the invention, preferably an antibody of the invention. In a preferred aspect, the compound is substantially purified (e.g., substantially free from substances that limit its effect or produce undesired side-effects). The subject is preferably an animal, including but not limited to animals such as cows, pigs, horses, chickens, cats, dogs, etc., and is preferably a mammal, and most preferably human.
Formulations and methods of administration that can be employed when the compound comprises a nucleic acid or an immunoglobulin are described above;
additional appropriate formulations and routes of administration can be selected from among those described herein below.
Various delivery systems are known and can be used to administer a compound of the invention, e:g., encapsulation in liposomes, microparticles, microcapsules, recombinant cells capable of expressing the compound, receptor-mediated endocytosis (see, e.g., Wu and Wu, J. Biol. Chem. 262:4429-4432 (1987)), construction of a nucleic acid as part of a retroviral or other vector, etc.
Methods of introduction include but are not limited to intradermal, intramuscular, intraperitoneal, intravenous, subcutaneous, intranasal, epidural, and oral routes. The compounds or compositions may be administered by any convenient route, for example by infusion or bolus injection, by absorption through epithelial or mucocutaneous linings (e.g., oral mucosa, rectal and intestinal mucosa, etc.) and may be administered together with other biologically active agents. Administration can be systemic or local. In addition, it may be desirable to introduce the pharmaceutical compounds or compositions of the invention into the central nervous system by any suitable route, including intraventricular and intrathecal injection; intraventricular injection may be facilitated by an intraventricular catheter, for example, attached to a reservoir, such as an Ommaya reservoir. Pulmonary administration can also be employed, e.g., by use of an inhaler or nebulizer, and formulation with an aerosolizing agent.
In a specific embodiment, it may be desirable to administer the pharmaceutical compounds or compositions of the invention locally to the area in need of treatment;

this may be achieved by, for example, and not by way of limitation, local infusion during surgery, topical application, e.g., in conjunction with a wound dressing after surgery, by injection, by means of a catheter, by means of a suppository, or by means of an implant, said implant being of a porous, non-porous, or gelatinous material, including membranes, such as sialastic membranes, or fibers. Preferably, when administering a protein, including an antibody, of the invention, care must be taken to use materials to which the protein does not absorb.
In another embodiment, the compound or composition can be delivered in a vesicle, in particular a liposome (see Langer, Science 249:1527-1533 (1990);
Treat et al., in Liposomes in the Therapy of Infectious Disease and Cancer, Lopez-Berestein and Fidler (eds.), Liss, New York, pp. 353- 365 (1989); Lopez-Berestein, ibid., pp.
317-327; see generally ibid.) In yet another embodiment, the compound or composition can be delivered in a controlled release system. In one embodiment, a pump may be used (see Langer, supra; Sefton, CRC Crit. Ref. Biomed. Eng. 14:201 (1987); Buchwald et al., Surgery 88:507 (1980); Saudek et al., N. Engl. J. Med. 321:574 (1989)). In another embodiment, polymeric materials can be used (see Medical Applications of Controlled Release, Langer and Wise (eds.), CRC Pres., Boca Raton, Florida (1974);
Controlled Drug Bioavailability, Drug Product Design and Performance, Smolen and Ball (eds.), Wiley, New York (1984); Ranger and Peppas, J., Macromol. Sci.
Rev.
Macromol. Chem. 23:61 (1983); see also Levy et al., Science 228:190 (1985);
During et al., Ann. Neurol. 25:351 (1989); Howard et al., J.Neurosurg. 71:105 (1989)). In yet another embodiment, a controlled release system can be placed in proximity of the therapeutic target, i.e., the brain, thus requiring only a fraction of the systemic dose (see, e.g., Goodson, in Medical Applications of Controlled Release, supra, vol. 2, pp.
115-138 (1984)).
Other controlled release systems are discussed in the review by Langer (Science 249:1527-1533 (1990)).
In a specific embodiment where the compound of the invention is a nucleic acid encoding a protein, the nucleic acid can be administered in vivo to promote expression of its encoded protein, by constructing it as part of an appropriate nucleic acid expression vector and administering it so that it becomes intracellular, e.g., by use of a retroviral vector (see U.S. Patent No. 4,980,286), or by direct injection, or by use of microparticle bombardment (e.g., a gene gun; Biolistic, Dupont), or coating with lipids or cell-surface receptors or transfecting agents, or by administering it in linkage to a homeobox- like peptide which is known to enter the nucleus (see e.g., Joliot et al., Proc. Natl. Acad. Sci. USA 88:1864-1868 (1991)), etc.
Alternatively, a nucleic acid can be introduced intracellularly and incorporated within host cell DNA
for expression, by homologous recombination.
The present invention also provides pharmaceutical compositions. Such compositions comprise a therapeutically effective amount of a compound, and a pharmaceutically acceptable Garner. In a specific embodiment, the term "pharmaceutically acceptable" means approved by a regulatory agency of the Federal or a state government or listed in the U.S. Pharmacopeia or other generally recognized pharmacopeia for use in animals, and more particularly in humans. The term "carrier" refers to a diluent, adjuvant, excipient, or vehicle with which the therapeutic is administered. Such pharmaceutical Garners can be sterile liquids, such as water and oils, including those of petroleum, animal, vegetable or synthetic origin, such as peanut oil, soybean oil, mineral oil, sesame oil and the like. Water is a preferred carrier when the pharmaceutical composition is administered intravenously.
Saline solutions and aqueous dextrose and glycerol solutions can also be employed as liquid Garners, particularly for injectable solutions. Suitable pharmaceutical excipients include starch, glucose, lactose, sucrose, gelatin, malt, rice, flour, chalk, silica gel, sodium stearate, glycerol monostearate, talc, sodium chloride, dried skim milk, glycerol, propylene, glycol, water, ethanol and the like. The composition, if desired, can also contain minor amounts of wetting or emulsifying agents, or pH
buffering agents. These compositions can take the form of solutions, suspensions, emulsion, tablets, pills, capsules, powders, sustained-release formulations and the like. The composition can be formulated as a suppository, with traditional binders and carriers such as triglycerides. Oral formulation can include standard carriers such as pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, sodium saccharine, cellulose, magnesium carbonate, etc. Examples of suitable pharmaceutical carriers are described in "Remington's Pharmaceutical Sciences"
by E.W. Martin. Such compositions will contain a therapeutically effective amount of the compound, preferably in purified form, together with a suitable amount of carrier so as to provide the form for proper administration to the patient. The formulation should suit the mode of administration.
In a preferred embodiment, the composition is formulated in accordance with routine procedures as a pharmaceutical composition adapted for intravenous administration to human beings. Typically, compositions for intravenous administration are solutions in sterile isotonic aqueous buffer. Where necessary, the composition may also include a solubilizing agent and a local anesthetic such as lignocaine to ease pain at the site of the injection. Generally, the ingredients are supplied either separately or mixed together in unit dosage form, for example, as a dry lyophilized powder or water free concentrate in a hermetically sealed container such as an ampoule or sachette indicating the quantity of active agent. Where the composition is to be administered by infusion, it can be dispensed with an infusion bottle containing sterile pharmaceutical grade water or saline. Where the composition is administered by injection, an ampoule of sterile water for injection or saline can be provided so that the ingredients may be mixed prior to administration.
The compounds of the invention can be formulated as neutral or salt forms.
Pharmaceutically acceptable salts include those formed with anions such as those derived from hydrochloric, phosphoric, acetic, oxalic, tartaric acids, etc., and those formed with cations such as those derived from sodium, potassium, ammonium, calcium, ferric hydroxides, isopropylamine, triethylamine, 2-ethylamino ethanol, histidine, procaine, etc.
The amount of the compound of the invention which will be effective in the treatment, inhibition and prevention of a disease or disorder associated with aberrant expression and/or activity of a polypeptide of the invention can be determined by standard clinical techniques. In addition, in vitro assays may optionally be employed to help identify optimal dosage ranges. The precise dose to be employed in the formulation will also depend on the route of administration, and the seriousness of the disease or disorder, and should be decided according to the judgment of the practitioner and each patient's circumstances. Effective doses may be extrapolated from dose-response curves derived from in vitro or animal model test systems.

For antibodies, the dosage administered to a patient is typically 0.1 mg/kg to 100 mg/kg of the patient's body weight. Preferably, the dosage administered to a patient is between 0.1 mg/kg and 20 mg/kg of the patient's body weight, more preferably 1 mg/kg to 10 mg/kg of the patient's body weight. Generally, human antibodies have a longer half life within the human body than antibodies from other species due to the immune response to the foreign polypeptides. Thus, lower dosages of human antibodies and less frequent administration is often possible.
Further, the dosage and frequency of administration of antibodies of the invention may be reduced by enhancing uptake and tissue penetration (e.g., into the brain) of the antibodies by modifications such as, for example, lipidation.
The invention also provides a pharmaceutical pack or kit comprising one or more containers filled with one or more of the ingredients of the pharmaceutical compositions of the invention. Optionally associated with such containers) can be a notice in the form prescribed by a governmental agency regulating the manufacture, use or sale of pharmaceuticals or biological products, which notice reflects approval by the agency of manufacture, use or sale for human administration.
Diagnosis and Imaging Labeled antibodies, and derivatives and analogs thereof, which specifically bind to a polypeptide of interest can be used for diagnostic purposes to detect, diagnose, or monitor diseases, disorders, and/or conditions associated with the aberrant expression and/or activity of a polypeptide of the invention. The invention provides for the detection of aberrant expression of a polypeptide of interest, comprising (a) assaying the expression of the polypeptide of interest in cells or body fluid of an individual using one or more antibodies specific to the polypeptide interest and (b) comparing the level of gene expression with a standard gene expression level, whereby an increase or decrease in the assayed polypeptide gene expression level compared to the standard expression level is indicative of aberrant expression.
The invention provides a diagnostic assay for diagnosing a disorder, comprising (a) assaying the expression of the polypeptide of interest in cells or body fluid of an individual using one or more antibodies specific to the polypeptide interest and (b) comparing the level of gene expression with a standard gene expression level, whereby an increase or decrease in the assayed polypeptide gene expression level compared to the standard expression level is indicative of a particular disorder. With respect to cancer, the presence of a relatively high amount of transcript in biopsied tissue from an individual may indicate a predisposition for the development of the disease, or may provide a means for detecting the disease prior to the appearance of actual clinical symptoms. A more definitive diagnosis of this type may allow health professionals to employ preventative measures or aggressive treatment earlier thereby preventing the development or further progression of the cancer.
Antibodies of the invention can be used to assay protein levels in a biological sample using classical immunohistological methods known to those of skill in the art (e.g., see Jalkanen, et al., J. Cell. Biol. 101:976-985 (1985); Jalkanen, et al., J. Cell .
Biol. 105:3087-3096 (1987)). Other antibody-based methods useful for detecting protein gene expression include immunoassays, such as the enzyme linked immunosorbent assay (ELISA) and the radioimmunoassay (RIA). Suitable antibody assay labels are known in the art and include enzyme labels, such as, glucose oxidase;
radioisotopes, such as iodine (125I, 121I), carbon (14C), sulfur (35S), tritium (3H), indium (112In), and technetium (99Tc); luminescent labels, such as luminol;
and fluorescent labels, such as fluorescein and rhodamine, and biotin.
One aspect of the invention is the detection and diagnosis of a disease or disorder associated with aberrant expression of a polypeptide of interest in an animal, preferably a mammal and most preferably a human. In one embodiment, diagnosis comprises: a) administering (for example, parenterally, subcutaneously, or intraperitoneally) to a subject an effective amount of a labeled molecule which specifically binds to the polypeptide of interest; b) waiting for a time interval following the administering for permitting the labeled molecule to preferentially concentrate at sites in the subject where the polypeptide is expressed (and for unbound labeled molecule to be cleared to background level); c) determining background level; and d) detecting the labeled molecule in the subject, such that detection of labeled molecule above the background level indicates that the subject has a particular disease or disorder associated with aberrant expression of the polypeptide of interest. Background level can be determined by various methods including, comparing the amount of labeled molecule detected to a standard value previously determined for a particular system.
It will be understood in the art that the size of the subject and the imaging system used will determine the quantity of imaging moiety needed to produce diagnostic images. In the case of a radioisotope moiety, for a human subject, the quantity of radioactivity injected will normally range from about 5 to 20 millicuries of 99mTc. The labeled antibody or antibody fragment will then preferentially accumulate at the location of cells which contain the specific protein. In vivo tumor imaging is described in S.W. Burchiel et al., "Immunopharmacokinetics of Radiolabeled Antibodies and Their Fragments." (Chapter 13 in Tumor Imaging:
The Radiochemical Detection of Cancer, S.W. Burchiel and B. A. Rhodes, eds., Masson Publishing Inc. (1982).
Depending on several variables, including the type of label used and the mode of administration, the time interval following the administration for permitting the labeled molecule to preferentially concentrate at sites in the subject and for unbound labeled molecule to be cleared to background level is 6 to 48 hours or 6 to 24 hours or 6 to 12 hours. In another embodiment the time interval following administration is 5 to 20 days or 5 to 10 days.
In an embodiment, monitoring of the disease or disorder is carned out by repeating the method for diagnosing the disease or disease, for example, one month after initial diagnosis, six months after initial diagnosis, one year after initial diagnosis, etc.
Presence of the labeled molecule can be detected in the patient using methods known in the art for in vivo scanning. These methods depend upon the type of label used. Skilled artisans will be able to determine the appropriate method for detecting a particular label. Methods and devices that may be used in the diagnostic methods of the invention include, but are not limited to, computed tomography (CT), whole body scan such as position emission tomography (PET), magnetic resonance imaging (MRI), and sonography.
In a specific embodiment, the molecule is labeled with a radioisotope and is detected in the patient using a radiation responsive surgical instrument (Thurston et al., U.S. Patent No. 5,441,050). In another embodiment, the molecule is labeled with a fluorescent compound and is detected in the patient using a fluorescence responsive scanning instrument. In another embodiment, the molecule is labeled with a positron emitting metal and is detected in the patent using positron emission-tomography. In yet another embodiment, the molecule is labeled with a paramagnetic label and is detected in a patient using magnetic resonance imaging (MRI).
Kits The present invention provides kits that can be used in the above methods. In one embodiment, a kit comprises an antibody of the invention, preferably a purified antibody, in one or more containers. In a specific embodiment, the kits of the present invention contain a substantially isolated polypeptide comprising an epitope which is specifically immunoreactive with an antibody included in the kit. Preferably, the kits of the present invention further comprise a control antibody which does not react with the polypeptide of interest. In another specific embodiment, the kits of the present invention contain a means for detecting the binding of an antibody to a polypeptide of interest (e.g., the antibody may be conjugated to a detectable substrate such as a fluorescent compound, an enzymatic substrate, a radioactive compound or a luminescent compound, or a second antibody which recognizes the first antibody may be conjugated to a detectable substrate).
In another specific embodiment of the present invention, the kit is a diagnostic kit for use in screening serum containing antibodies specific against proliferative and/or cancerous polynucleotides and polypeptides. Such a kit may include a control antibody that does not react with the polypeptide of interest. Such a kit may include a substantially isolated polypeptide antigen comprising an epitope which is specifically immunoreactive with at least one anti-polypeptide antigen antibody. Further, such a kit includes means for detecting the binding of said antibody to the antigen (e.g., the antibody may be conjugated to a fluorescent compound such as fluorescein or rhodamine which can be detected by flow cytometry). In specific embodiments, the kit may include a recombinantly produced or chemically synthesized polypeptide antigen. The polypeptide antigen of the kit may also be attached to a solid support.
In a more specific embodiment the detecting means of the above-described kit includes a solid support to which said polypeptide antigen is attached. Such a kit may also include a non-attached reporter-labeled anti-human antibody. In this embodiment, binding of the antibody to the polypeptide antigen can be detected by binding of the said reporter-labeled antibody.
In an additional embodiment, the invention includes a diagnostic kit for use in S screening serum containing antigens of the polypeptide of the invention. The diagnostic kit includes a substantially isolated antibody specifically immunoreactive with polypeptide or polynucleotide antigens, and means for detecting the binding of the polynucleotide or polypeptide antigen to the antibody. In one embodiment, the antibody is attached to a solid support. In a specific embodiment, the antibody may be a monoclonal antibody. The detecting means of the kit may include a second, labeled monoclonal antibody. Alternatively, or in addition, the detecting means may include a labeled, competing antigen.
In one diagnostic configuration, test serum is reacted with a solid phase reagent having a surface-bound antigen obtained by the methods of the present invention. After binding with specific antigen antibody to the reagent and removing unbound serum components by washing, the reagent is reacted with reporter-labeled anti-human antibody to bind reporter to the reagent in proportion to the amount of bound anti-antigen antibody on the solid support. The reagent is again washed to remove unbound labeled antibody, and the amount of reporter associated with the reagent is determined. Typically, the reporter is an enzyme which is detected by incubating the solid phase in the presence of a suitable fluorometric, luminescent or colorimetric substrate (Sigma, St. Louis, MO).
The solid surface reagent in the above assay is prepared by known techniques for attaching protein material to solid support material, such as polymeric beads, dip sticks, 96-well plate or filter material. These attachment methods generally include non-specific adsorption of the protein to the support or covalent attachment of the protein, typically through a free amine group, to a chemically reactive group on the solid support, such as an activated carboxyl, hydroxyl, or aldehyde group.
Alternatively, streptavidin coated plates can be used in conjunction with biotinylated antigen(s).
Thus, the invention provides an assay system or kit for carrying out this diagnostic method. The kit generally includes a support with surface- bound recombinant antigens, and a reporter-labeled anti-human antibody for detecting surface-bound anti-antigen antibody.
Fusion Proteins Any polypeptide of the present invention can be used to generate fusion proteins. For example, the polypeptide of the present invention, when fused to a second protein, can be used as an antigenic tag. Antibodies raised against the polypeptide of the present invention can be used to indirectly detect the second protein by binding to the polypeptide. Moreover, because secreted proteins target cellular locations based on trafficking signals, the polypeptides of the present invention can be used as targeting molecules once fused to other proteins.
Examples of domains that can be fused to polypeptides of the present invention include not only heterologous signal sequences, but also other heterologous functional regions. The fusion does not necessarily need to be direct, but may occur through linker sequences.
Moreover, fusion proteins may also be engineered to improve characteristics of the polypeptide of the present invention. For instance, a region of additional amino acids, particularly charged amino acids, may be added to the N-terminus of the polypeptide to improve stability and persistence during purification from the host cell or subsequent handling and storage. Also, peptide moieties may be added to the polypeptide to facilitate purification. Such regions may be removed prior to final preparation of the polypeptide. The addition of peptide moieties to facilitate handling of polypeptides are familiar and routine techniques in the art.
Moreover, polypeptides of the present invention, including fragments, and specifically epitopes, can be combined with parts of the constant domain of immunoglobulins (IgA, IgE, IgG, IgM) or portions thereof (CH1, CH2, CH3, and any combination thereof, including both entire domains and portions thereof), resulting in chimeric polypeptides. These fusion proteins facilitate purification and show an increased half life in vivo. One reported example describes chimeric proteins consisting of the first two domains of the human CD4-polypeptide and various domains of the constant regions of the heavy or light chains of mammalian immunoglobulins. (EP A 394,827; Traunecker et al., Nature 331:84-86 (1988).) Fusion proteins having disulfide-linked dimeric structures (due to the IgG) can also be more efficient in binding and neutralizing other molecules, than the monomeric secreted protein or protein fragment alone. (Fountoulakis et al., J. Biochem.
270:3958-3964 (1995).) Polynucleotides comprising or alternatively consisting of nucleic acids which encode these fusion proteins are also encompassed by the invention.
Similarly, EP-A-O 464 533 (Canadian counterpart 2045869) discloses fusion proteins comprising various portions of constant region of immunoglobulin molecules together with another human protein or part thereof. In many cases, the Fc part in a fusion protein is beneficial in therapy and diagnosis, and thus can result in, for example, improved pharmacokinetic properties. (EP-A 0232 262.) Alternatively, deleting the Fc part after the fusion protein has been expressed, detected, and purified, would be desired. For example, the Fc portion may hinder therapy and diagnosis if the fusion protein is used as an antigen for immunizations. In drug discovery, for 1 S example, human proteins, such as hIL-5, have been fused with Fc portions for the purpose of high-throughput screening assays to identify antagonists of hIL-5.
(See, D. Bennett et al., J. Molecular Recognition 8:52-58 (1995); K. Johanson et al., J. Biol.
Chem. 270:9459-9471 (1995).) Moreover, the polypeptides of the present invention can be fused to marker sequences, such as a peptide which facilitates purification of the fused polypeptide.
In preferred embodiments, the marker amino acid sequence is a hexa-histidine peptide, such as the tag provided in a pQE vector (QIAGEN, Inc., 9259 Eton Avenue, Chatsworth, CA, 91311), among others, many of which are commercially available.
As described in Gentz et al., Proc. Natl. Acad. Sci. USA 86:821-824 (1989), for instance, hexa-histidine provides for convenient purification of the fusion protein.
Another peptide tag useful for purification, the "HA" tag, corresponds to an epitope derived from the influenza hemagglutinin protein. (Wilson et al., Cell 37:767 (1984).) Thus, any of these above fusions can be engineered using the polynucleotides or the polypeptides of the present invention.
Vectors, Host Cells, and Protein Production The present invention also relates to vectors containing the polynucleotide of the present invention, host cells, and the production of polypeptides by recombinant techniques. The vector may be, for example, a phage, plasmid, viral, or retroviral vector. Retroviral vectors may be replication competent or replication defective. In the latter case, viral propagation generally will occur only in complementing host cells.
The polynucleotides may be joined to a vector containing a selectable marker for propagation in a host. Generally, a plasmid vector is introduced in a precipitate, such as a calcium phosphate precipitate, or in a complex with a charged lipid.
If the vector is a virus, it may be packaged in vitro using an appropriate packaging cell line and then transduced into host cells.
The polynucleotide insert should be operatively linked to an appropriate promoter, such as the phage lambda PL promoter, the E. coli lac, trp, phoA and tac promoters, the SV40 early and late promoters and promoters of retroviral LTRs, to name a few. Other suitable promoters will be known to the skilled artisan. The expression constructs will further contain sites for transcription initiation, termination, and, in the transcribed region, a ribosome binding site for translation. The coding portion of the transcripts expressed by the constructs will preferably include a translation initiating codon at the beginning and a termination codon (UAA, UGA or UAG) appropriately positioned at the end of the polypeptide to be translated.
As indicated, the expression vectors will preferably include at least one selectable marker. Such markers include dihydrofolate reductase, 6418 or neomycin resistance for eukaryotic cell culture and tetracycline, kanamycin or ampicillin resistance genes for culturing in E. coli and other bacteria. Representative examples of appropriate hosts include, but are not limited to, bacterial cells, such as E. coli, Streptomyces and Salmonella typhimurium cells; fungal cells, such as yeast cells (e.g., Saccharomyces cerevisiae or Pichia pastoris (ATCC Accession No.
201178));
insect cells such as Drosophila S2 and Spodoptera Sf~ cells; animal cells such as CHO, COS, 293, and Bowes melanoma cells; and plant cells. Appropriate culture mediums and conditions for the above-described host cells are known in the art.
Among vectors preferred for use in bacteria include pQE70, pQE60 and pQE-9, available from QIAGEN, Inc.; pBluescript vectors, Phagescript vectors, pNH8A, pNHl6a, pNHl8A, pNH46A, available from Stratagene Cloning Systems, Inc.; and ptrc99a, pKK223-3, pKK233-3, pDR540, pRITS available from Pharmacia Biotech, Inc. Among preferred eukaryotic vectors are pWLNEO, pSV2CAT, pOG44, pXTI
and pSG available from Stratagene; and pSVK3, pBPV, pMSG and pSVL available from Pharmacia. Preferred expression vectors for use in yeast systems include, but are not limited to pYES2, pYDI, pTEFI/Zeo, pYES2/GS, pPICZ,pGAPZ, pGAPZaIph, pPIC9, pPIC3.5, pHIL-D2, pHIL-S1, pPIC3.5K, pPIC9K, and PA0815 (all available from Invitrogen, Carlbad, CA). Other suitable vectors will be readily apparent to the skilled artisan.
Introduction of the construct into the host cell can be effected by calcium phosphate transfection, DEAE-dextran mediated transfection, cationic lipid-mediated transfection, electroporation, transduction, infection, or other methods. Such methods are described in many standard laboratory manuals, such as Davis et al., Basic Methods In Molecular Biology (1986). It is specifically contemplated that the polypeptides of the present invention may in fact be expressed by a host cell lacking a recombinant vector.
A polypeptide of this invention can be recovered and purified from recombinant cell cultures by well-known methods including ammonium sulfate or ethanol precipitation, acid extraction, anion or cation exchange chromatography, phosphocellulose chromatography, hydrophobic interaction chromatography, affinity chromatography, hydroxylapatite chromatography and lectin chromatography. Most preferably, high performance liquid chromatography ("HPLC") is employed for purification.
Polypeptides of the present invention, and preferably the secreted form, can also be recovered from: products purified from natural sources, including bodily fluids, tissues and cells, whether directly isolated or cultured; products of chemical synthetic procedures; and products produced by recombinant techniques from a prokaryotic or eukaryotic host, including, for example, bacterial, yeast, higher plant, insect, and mammalian cells. Depending upon the host employed in a recombinant production procedure, the polypeptides of the present invention may be glycosylated or may be non-glycosylated. In addition, polypeptides of the invention may also include an initial modified methionine residue, in some cases as a result of host-mediated processes. Thus, it is well known in the art that the N-terminal methionine encoded by the translation initiation codon generally is removed with high efficiency from any protein after translation in all eukaryotic cells. While the N-terminal methionine on most proteins also is efficiently removed in most prokaryotes, for some proteins, this prokaryotic removal process is inefficient, depending on the nature of the amino acid to which the N-terminal methionine is covalently linked.
In one embodiment, the yeast Pichia pastoris is used to express the polypeptide of the present invention in a eukaryotic system. Pichia pastoris is a methylotrophic yeast which can metabolize methanol as its sole carbon source.
A
main step in the methanol metabolization pathway is the oxidation of methanol to formaldehyde using OZ. This reaction is catalyzed by the enzyme alcohol oxidase. In order to metabolize methanol as its sole carbon source, Pichia pastoris must generate high levels of alcohol oxidase due, in part, to the relatively low affinity of alcohol oxidase for 02. Consequently, in a growth medium depending on methanol as a main carbon source, the promoter region of one of the two alcohol oxidase genes (AOXI ) is highly active. In the presence of methanol, alcohol oxidase produced from the AOXI
gene comprises up to approximately 30% of the total soluble protein in Pichia pastoris. See, Ellis, S.B., et al., Mol. Cell. Biol. 5:1111-21 (1985); Koutz, P.J, et al., Yeast 5:167-77 (1989); Tschopp, J.F., et al., Nucl. Acids Res. 15:3859-76 (1987).
Thus, a heterologous coding sequence, such as, for example, a polynucleotide of the present invention, under the transcriptional regulation of all or part of the AOXI
regulatory sequence is expressed at exceptionally high levels in Pichia yeast grown in the presence of methanol.
In one example, the plasmid vector pPIC9K is used to express DNA encoding a polypeptide of the invention, as set forth herein, in a Pichea yeast system essentially as described in "Pichia Protocols: Methods in Molecular Biology," D.R. Higgins and J. Cregg, eds. The Humana Press, Totowa, NJ, 1998. This expression vector allows expression and secretion of a protein of the invention by virtue of the strong ADX~
promoter linked to the Pichia pastoris alkaline phosphatase (PHO) secretory signal peptide (i.e., leader) located upstream of a multiple cloning site.

Many other yeast vectors could be used in place of pPIC9K, such as, pYES2, pYDI, pTEFI/Zeo, pYES2/GS, pPICZ, pGAPZ, pGAPZalpha, pPIC9, pPIC3.5, pHIL-D2, pHIL-S1, pPIC3.5K, and PA0815, as one skilled in the art would readily appreciate, as long as the proposed expression construct provides appropriately located signals for transcription, translation, secretion (if desired), and the like, including an in-frame AUG as required.
In another embodiment, high-level expression of a heterologous coding sequence, such as, for example, a polynucleotide of the present invention, may be achieved by cloning the heterologous polynucleotide of the invention into an expression vector such as, for example, pGAPZ or pGAPZalpha, and growing the yeast culture in the absence of methanol.
In addition to encompassing host cells containing the vector constructs discussed herein, the invention also encompasses primary, secondary, and immortalized host cells of vertebrate origin, particularly mammalian origin, that have 1 S been engineered to delete or replace endogenous genetic material (e.g., coding sequence), and/or to include genetic material (e.g., heterologous polynucleotide sequences) that is operably associated with the polynucleotides of the invention, and which activates, alters, and/or amplifies endogenous polynucleotides. For example, techniques known in the art may be used to operably associate heterologous control regions (e.g., promoter and/or enhancer) and endogenous polynucleotide sequences via homologous recombination, resulting in the formation of a new transcription unit (see, e.g., U.S. Patent No. 5,641,670, issued June 24, 1997; U.S. Patent No.
5,733,761, issued March 31, 1998; International Publication No. WO 96/29411, published September 26, 1996; International Publication No. WO 94/12650, published August 4, 1994; Koller et al., Proc. Natl. Acad. Sci. USA 86:8932-(1989); and Zijlstra et al., Nature 342:435-438 (1989), the disclosures of each of which are incorporated by reference in their entireties).
In addition, polypeptides of the invention can be chemically synthesized using techniques known in the art (e.g., see Creighton, 1983, Proteins: Structures and Molecular Principles, W.H. Freeman & Co., N.Y., and Hunkapiller et al., Nature, 310:105-111 (1984)). For example, a polypeptide corresponding to a fragment of a polypeptide sequence of the invention can be synthesized by use of a peptide synthesizer. Furthermore, if desired, nonclassical amino acids or chemical amino acid analogs can be introduced as a substitution or addition into the polypeptide sequence.
Non-classical amino acids include, but are not limited to, to the D-isomers of the common amino acids, 2,4-diaminobutyric acid, a-amino isobutyric acid, 4-aminobutyric acid, Abu, 2-amino butyric acid, g-Abu, e-Ahx, 6-amino hexanoic acid, Aib, 2-amino isobutyric acid, 3-amino propionic acid, ornithine, norleucine, norvaline, hydroxyproline, sarcosine, citrulline, homocitrulline, cysteic acid, t-butylglycine, t-butylalanine, phenylglycine, cyclohexylalanine, b-alanine, fluoro-amino acids, designer amino acids such as b-methyl amino acids, Ca-methyl amino acids, Na-methyl amino acids, and amino acid analogs in general. Furthermore, the amino acid can be D (dextrorotary) or L (levorotary).
The invention encompasses polypeptides which are differentially modified during or after translation, e.g., by glycosylation, acetylation, phosphorylation, amidation, derivatization by known protecting/blocking groups, proteolytic cleavage, linkage to an antibody molecule or other cellular ligand, etc. Any of numerous chemical modifications may be carried out by known techniques, including but not limited, to specific chemical cleavage by cyanogen bromide, trypsin, chymotrypsin, papain, V8 protease, NaBH4; acetylation, formylation, oxidation, reduction;
metabolic synthesis in the presence of tunicamycin; etc.
Additional post-translational modifications encompassed by the invention include, for example, e.g., N-linked or O-linked carbohydrate chains, processing of N-terminal or C-terminal ends), attachment of chemical moieties to the amino acid backbone, chemical modifications of N-linked or O-linked carbohydrate chains, and addition or deletion of an N-terminal methionine residue as a result of procaryotic host cell expression. The polypeptides may also be modified with a detectable label, such as an enzymatic, fluorescent, isotopic or affinity label to allow for detection and isolation of the protein.
Also provided by the invention are chemically modified derivatives of the polypeptides of the invention which may provide additional advantages such as increased solubility, stability and circulating time of the polypeptide, or decreased immunogenicity (see U.S. Patent NO: 4,179,337). The chemical moieties for derivitization may be selected from water soluble polymers such as polyethylene glycol, ethylene glycol/propylene glycol copolymers, carboxymethylcellulose, dextran, polyvinyl alcohol and the like. The polypeptides may be modified at random positions within the molecule, or at predetermined positions within the molecule and may include one, two, three or more attached chemical moieties.
The polymer may be of any molecular weight, and may be branched or unbranched. For polyethylene glycol, the preferred molecular weight is between about 1 kDa and about 100 kDa (the term "about" indicating that in preparations of polyethylene glycol, some molecules will weigh more, some less, than the stated molecular weight) for ease in handling and manufacturing. Other sizes may be used, depending on the desired therapeutic profile (e.g., the duration of sustained release desired, the effects, if any on biological activity, the ease in handling, the degree or lack of antigenicity and other known effects of the polyethylene glycol to a therapeutic protein or analog). For example, the polyethylene glycol may have an average molecular weight of about 200, 500, 1000, 1500, 2000, 2500, 3000, 3500, 4000, 4500, 5000, 5500, 6000, 6500, 7000, 7500, 8000, 8500, 9000, 9500, 10,000, 10,500, 11,000, 11,500, 12,000, 12,500, 13,000, 13,500, 14,000, 14,500, 15,000, 15,500, 16,000, 16,500, 17,000, 17,500, 18,000, 18,500, 19,000, 19,500, 20,000, 25,000, 30,000, 35,000, 40,000, 50,000, 55,000, 60,000, 65,000, 70,000, 75,000, 80,000, 85,000, 90,000, 95,000, or 100,000 kDa.
As noted above, the polyethylene glycol may have a branched structure.
Branched polyethylene glycols are described, for example, in U.S. Patent No.
5,643,575; Morpurgo et al., Appl. Biochem. Biotechnol. 56:59-72 (1996);
Vorobjev et al., Nucleosides Nucleotides 18:2745-2750 (1999); and Caliceti et al., Bioconjug.
Chem. 10:638-646 (1999), the disclosures of each of which are incorporated herein by reference.
The polyethylene glycol molecules (or other chemical moieties) should be attached to the protein with consideration of effects on functional or antigenic domains of the protein. There are a number of attachment methods available to those skilled in the art, e.g., EP 0 401 384, herein incorporated by reference (coupling PEG
to G-CSF), see also Malik et al., Exp. Hematol. 20:1028-1035 (1992) (reporting pegylation of GM-CSF using tresyl chloride). For example, polyethylene glycol may be covalently bound through amino acid residues via a reactive group, such as, a free amino or carboxyl group. Reactive groups are those to which an activated polyethylene glycol molecule may be bound. The amino acid residues having a free amino group may include lysine residues and the N-terminal amino acid residues;
S those having a free carboxyl group may include aspartic acid residues glutamic acid residues and the C-terminal amino acid residue. Sulfhydryl groups may also be used as a reactive group for attaching the polyethylene glycol molecules. Preferred for therapeutic purposes is attachment at an amino group, such as attachment at the N-terminus or lysine group.
As suggested above, polyethylene glycol may be attached to proteins via linkage to any of a number of amino acid residues. For example, polyethylene glycol can be linked to a proteins via covalent bonds to lysine, histidine, aspartic acid, glutamic acid, or cysteine residues. One or more reaction chemistries may be employed to attach polyethylene glycol to specific amino acid residues (e.g., lysine, histidine, aspartic acid, glutamic acid, or cysteine) of the protein or to more than one type of amino acid residue (e.g., lysine, histidine, aspartic acid, glutamic acid, cysteine and combinations thereof) of the protein.
One may specifically desire proteins chemically modified at the N-terminus.
Using polyethylene glycol as an illustration of the present composition, one may select from a variety of polyethylene glycol molecules (by molecular weight, branching, etc.), the proportion of polyethylene glycol molecules to protein (polypeptide) molecules in the reaction mix, the type of pegylation reaction to be performed, and the method of obtaining the selected N-terminally pegylated protein.
The method of obtaining the N-terminally pegylated preparation (i.e., separating this moiety from other monopegylated moieties if necessary) may be by purification of the N-terminally pegylated material from a population of pegylated protein molecules.
Selective proteins chemically modified at the N-terminus modification may be accomplished by reductive alkylation which exploits differential reactivity of different types of primary amino groups (lysine versus the N-terminal) available for derivatization in a particular protein. Under the appropriate reaction conditions, substantially selective derivatization of the protein at the N-terminus with a carbonyl group containing polymer is achieved.

As indicated above, pegylation of the proteins of the invention may be accomplished by any number of means. For example, polyethylene glycol may be attached to the protein either directly or by an intervening linker.
Linkerless systems for attaching polyethylene glycol to proteins are described in Delgado et al., Crit. Rev.
Thera. Drug Carrier Sys. 9:249-304 (1992); Francis et al., Intern. J. of Hematol.
68:1-18 (1998); U.S. Patent No. 4,002,531; U.S. Patent No. 5,349,052; WO
95/06058;
and WO 98/32466, the disclosures of each of which are incorporated herein by reference.
One system for attaching polyethylene glycol directly to amino acid residues of proteins without an intervening linker employs tresylated MPEG, which is produced by the modification of monmethoxy polyethylene glycol (MPEG) using tresylchloride (C1SOZCHZCF3). Upon reaction of protein with tresylated MPEG, polyethylene glycol is directly attached to amine groups of the protein. Thus, the invention includes protein-polyethylene glycol conjugates produced by reacting proteins of the invention with a polyethylene glycol molecule having a 2,2,2-trifluoreothane sulphonyl group.
Polyethylene glycol can also be attached to proteins using a number of different intervening linkers. For example, U.S. Patent No. 5,612,460, the entire disclosure of which is incorporated herein by reference, discloses urethane linkers for connecting polyethylene glycol to proteins. Protein-polyethylene glycol conjugates wherein the polyethylene glycol is attached to the protein by a linker can also be produced by reaction of proteins with compounds such as MPEG-succinimidylsuccinate, MPEG activated with 1,1'-carbonyldiimidazole, MPEG-2,4,5-trichloropenylcarbonate, MPEG-p-nitrophenolcarbonate, and various MPEG-succinate derivatives. A number additional polyethylene glycol derivatives and reaction chemistries for attaching polyethylene glycol to proteins are described in WO 98/32466, the entire disclosure of which is incorporated herein by reference.
Pegylated protein products produced using the reaction chemistries set out herein are included within the scope of the invention.
The number of polyethylene glycol moieties attached to each protein of the invention (i.e., the degree of substitution) may also vary. For example, the pegylated proteins of the invention may be linked, on average, to 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 12, 15, 17, 20, or more polyethylene glycol molecules. Similarly, the average degree of substitution within ranges such as 1-3, 2-4, 3-5, 4-6, 5-7, 6-8, 7-9, 8-10, 9-11, 10-12, 11-13, 12-14, 13-15, 14-16, 15-17, 16-18, 17-19, or 18-20 polyethylene glycol moieties per protein molecule. Methods for determining the degree of substitution are discussed, for example, in Delgado et al., Crit. Rev. Thera. Drug Carrier Sys.
9:249-304 (1992).
The polypeptides of the invention may be in monomers or multimers (i.e., dimers, trimers, tetramers and higher multimers). Accordingly, the present invention relates to monomers and multimers of the polypeptides of the invention, their preparation, and compositions (preferably, Therapeutics) containing them. In specific embodiments, the polypeptides of the invention are monomers, dimers, trimers or tetramers. In additional embodiments, the multimers of the invention are at least dimers, at least trimers, or at least tetramers.
Multimers encompassed by the invention may be homomers or heteromers.
As used herein, the term homomer, refers to a multimer containing only polypeptides corresponding to the amino acid sequence of SEQ ID NO:Y or encoded by the cDNA
contained in a deposited clone (including fragments, variants, splice variants, and fusion proteins, corresponding to these polypeptides as described herein).
These homomers may contain polypeptides having identical or different amino acid sequences. In a specific embodiment, a homomer of the invention is a multimer containing only polypeptides having an identical amino acid sequence. In another specific embodiment, a homomer of the invention is a multimer containing polypeptides having different amino acid sequences. In specific embodiments, the multimer of the invention is a homodimer (e.g., containing polypeptides having identical or different amino acid sequences) or a homotrimer (e.g., containing polypeptides having identical and/or different amino acid sequences). In additional embodiments, the homomeric multimer of the invention is at least a homodimer, at least a homotrimer, or at least a homotetramer.
As used herein, the term heteromer refers to a multimer containing one or more heterologous polypeptides (i.e., polypeptides of different proteins) in addition to the polypeptides of the invention. In a specific embodiment, the multimer of the invention is a heterodimer, a heterotrimer, or a heterotetramer. In additional embodiments, the heteromeric multimer of the invention is at least a heterodimer, at least a heterotrimer, or at least a heterotetramer.
Multimers of the invention may be the result of hydrophobic, hydrophilic, ionic and/or covalent associations and/or may be indirectly linked, by for example, S liposome formation. Thus, in one embodiment, multimers of the invention, such as, for example, homodimers or homotrimers, are formed when polypeptides of the invention contact one another in solution. In another embodiment, heteromultimers of the invention, such as, for example, heterotrimers or heterotetramers, are formed when polypeptides of the invention contact antibodies to the polypeptides of the invention (including antibodies to the heterologous polypeptide sequence in a fusion protein of the invention) in solution. In other embodiments, multimers of the invention are formed by covalent associations with and/or between the polypeptides of the invention. Such covalent associations may involve one or more amino acid residues contained in the polypeptide sequence ( e.g., that recited in the sequence listing, or contained in the polypeptide encoded by a deposited clone). In one instance, the covalent associations are cross-linking between cysteine residues located within the polypeptide sequences which interact in the native (i.e., naturally occurring) polypeptide. In another instance, the covalent associations are the consequence of chemical or recombinant manipulation. Alternatively, such covalent associations may involve one or more amino acid residues contained in the heterologous polypeptide sequence in a fusion protein of the invention.
In one example, covalent associations are between the heterologous sequence contained in a fusion protein of the invention (see, e.g., US Patent Number 5,478,925). In a specific example, the covalent associations are between the heterologous sequence contained in an Fc fusion protein of the invention (as described herein). In another specific example, covalent associations of fusion proteins of the invention are between heterologous polypeptide sequence from another protein that is capable of forming covalently associated multimers, such as for example, oseteoprotegerin (see, e.g., International Publication NO: WO
98/49305, the contents of which are herein incorporated by reference in its entirety). In another embodiment, two or more polypeptides of the invention are joined through peptide linkers. Examples include those peptide linkers described in U.S. Pat. No.
5,073,627 (hereby incorporated by reference). Proteins comprising multiple polypeptides of the invention separated by peptide linkers may be produced using conventional recombinant DNA technology.
Another method for preparing multimer polypeptides of the invention involves use of polypeptides of the invention fused to a leucine zipper or isoleucine zipper polypeptide sequence. Leucine zipper and isoleucine zipper domains are polypeptides that promote multimerization of the proteins in which they are found. Leucine zippers were originally identified in several DNA-binding proteins (Landschulz et al., Science 240:1759, (1988)), and have since been found in a variety of different proteins. Among the known leucine zippers are naturally occurring peptides and derivatives thereof that dimerize or trimerize. Examples of leucine zipper domains suitable for producing soluble multimeric proteins of the invention are those described in PCT application WO 94/10308, hereby incorporated by reference. Recombinant fusion proteins comprising a polypeptide of the invention fused to a polypeptide sequence that dimerizes or trimerizes in solution are expressed in suitable host cells, and the resulting soluble multimeric fusion protein is recovered from the culture supernatant using techniques known in the art.
Trimeric polypeptides of the invention may offer the advantage of enhanced biological activity. Preferred leucine zipper moieties and isoleucine moieties are those that preferentially form trimers. One example is a leucine zipper derived from lung surfactant protein D (SPD), as described in Hoppe et al. (FEBS Letters 344:191, (1994)) and in U.S. patent application Ser. No. 08/446,922, hereby incorporated by reference. Other peptides derived from naturally occurnng trimeric proteins may be employed in preparing trimeric polypeptides of the invention.
In another example, proteins of the invention are associated by interactions between Flag~ polypeptide sequence contained in fusion proteins of the invention containing Flag~ polypeptide seuqence. In a further embodiment, associations proteins of the invention are associated by interactions between heterologous polypeptide sequence contained in Flag~ fusion proteins of the invention and anti-Flag~ antibody.
The multimers of the invention may be generated using chemical techniques known in the art. For example, polypeptides desired to be contained in the multimers of the invention may be chemically cross-linked using linker molecules and linker molecule length optimization techniques known in the art (see, e.g., US Patent Number 5,478,925, which is herein incorporated by reference in its entirety).
Additionally, multimers of the invention may be generated using techniques known in S the art to form one or more inter-molecule cross-links between the cysteine residues located within the sequence of the polypeptides desired to be contained in the multimer (see, e.g., US Patent Number 5,478,925, which is herein incorporated by reference in its entirety). Further, polypeptides of the invention may be routinely modified by the addition of cysteine or biotin to the C terminus or N-terminus of the polypeptide and techniques known in.the art may be applied to generate multimers containing one or more of these modified polypeptides (see, e.g., US Patent Number 5,478,925, which is herein incorporated by reference in its entirety).
Additionally, techniques known in the art may be applied to generate liposomes containing the polypeptide components desired to be contained in the multimer of the invention (see, e.g., US Patent Number 5,478,925, which is herein incorporated by reference in its entirety).
Alternatively, multimers of the invention may be generated using genetic engineering techniques known in the art. In one embodiment, polypeptides contained in multimers of the invention are produced recombinantly using fusion protein technology described herein or otherwise known in the art (see, e.g., US
Patent Number 5,478,925, which is herein incorporated by reference in its entirety).
In a specific embodiment, polynucleotides coding for a homodimer of the invention are generated by ligating a polynucleotide sequence encoding a polypeptide of the invention to a sequence encoding a linker polypeptide and then further to a synthetic polynucleotide encoding the translated product of the polypeptide in the reverse orientation from the original C-terminus to the N-terminus (lacking the leader sequence) (see, e.g., US Patent Number 5,478,925, which is herein incorporated by reference in its entirety). In another embodiment, recombinant techniques described herein or otherwise known in the art are applied to generate recombinant polypeptides of the invention which contain a transmembrane domain (or hyrophobic or signal peptide) and which can be incorporated by membrane reconstitution techniques into liposomes (see, e.g., US Patent Number 5,478,925, which is herein incorporated by reference in its entirety).
Uses of the Polynucleotides Each of the polynucleotides identified herein can be used in numerous ways as reagents. The following description should be considered exemplary and utilizes known techniques.
The polynucleotides of the present invention are useful for chromosome identification. There exists an ongoing need to identify new chromosome markers, since few chromosome marking reagents, based on actual sequence data (repeat polymorphisms), are presently available. Each polynucleotide of the present invention can be used as a chromosome marker.
Briefly, sequences can be mapped to chromosomes by preparing PCR primers (preferably 15-25 bp) from the sequences shown in SEQ ID NO:X. Primers can be selected using computer analysis so that primers do not span more than one predicted exon in the genomic DNA. These primers are then used for PCR screening of somatic cell hybrids containing individual human chromosomes. Only those hybrids containing the human gene corresponding to the SEQ >I7 NO:X will yield an amplified fragment.
Similarly, somatic hybrids provide a rapid method of PCR mapping the polynucleotides to particular chromosomes. Three or more clones can be assigned per day using a single thermal cycler. Moreover, sublocalization of the polynucleotides can be achieved with panels of specific chromosome fragments. Other gene mapping strategies that can be used include in situ hybridization, prescreening with labeled flow-sorted chromosomes, preselection by hybridization to construct chromosome specific-cDNA libraries and computer mapping techniques (See, e.g., Shiner, Trends Biotechnol 16:456-459 (1998) which is hereby incorporated by reference in its entirety)..
Precise chromosomal location of the polynucleotides can also be achieved using fluorescence in situ hybridization (FISH) of a metaphase chromosomal spread.
This technique uses polynucleotides as short as 500 or 600 bases; however, polynucleotides 2,000-4,000 by are preferred. For a review of this technique, see Verma et al., "Human Chromosomes: a Manual of Basic Techniques," Pergamon Press, New York (1988).
For chromosome mapping, the polynucleotides can be used individually (to mark a single chromosome or a single site on that chromosome) or in panels (for marking multiple sites and/or multiple chromosomes).
The polynucleotides of the present invention would likewise be useful for radiation hybrid mapping, HAPPY mapping, and long range restriction mapping.
For a review of these techniques and others known in the art, see, e.g., Dear, "Genome Mapping: A Practical Approach," IRL Press at Oxford University Press, London (1997); Aydin, J. Mol. Med. 77:691-694 (1999); Hacia et al., Mol. Psychiatry 3:483-492 (1998); Herrick et al., Chromosome Res. 7:409-423 (1999); Hamilton et al., Methods Cell Biol. 62:265-280 (2000); and/or Ott, J. Hered. 90:68-70 (1999) each of which is hereby incorporated by reference in its entirety.
Once a polynucleotide has been mapped to a precise chromosomal location, the physical position of the polynucleotide can be used in linkage analysis.
Linkage analysis establishes coinheritance between a chromosomal location and presentation of a particular disease. (Disease mapping data are found, for example, in V.
McKusick, Mendelian Inheritance in Man (available on line through Johns Hopkins University Welch Medical Library) .) Assuming 1 megabase mapping resolution and one gene per 20 kb, a cDNA precisely localized to a chromosomal region associated with the disease could be one of 50-S00 potential causative genes.
Thus, once coinheritance is established, differences in the polynucleotide and the corresponding gene between affected and unaffected individuals can be examined.
First, visible structural alterations in the chromosomes, such as deletions or translocations, are examined in chromosome spreads or by PCR. If no structural alterations exist, the presence of point mutations are ascertained. Mutations observed in some or all affected individuals, but riot in normal individuals, indicates that the mutation may cause the disease. However, complete sequencing of the polypeptide and the corresponding gene from several normal individuals is required to distinguish the mutation from a polymorphism. If a new polymorphism is identified, this polymorphic polypeptide can be used for further linkage analysis.

Furthermore, increased or decreased expression of the gene in affected individuals as compared to unaffected individuals can be assessed using polynucleotides of the present invention. Any of these alterations (altered expression, chromosomal rearrangement, or mutation) can be used as a diagnostic or prognostic marker.
Thus, the invention also provides a diagnostic method useful during diagnosis of a disorder, involving measuring the expression level of polynucleotides of the present invention in cells or body fluid from an individual and comparing the measured gene expression level with a standard level of polynucleotide expression level, whereby an increase or decrease in the gene expression level compared to the standard is indicative of a disorder.
In still another embodiment, the invention includes a kit for analyzing samples for the presence of proliferative and/or cancerous polynucleotides derived from a test subject. In a general embodiment, the kit includes at least one polynucleotide probe 1 S containing a nucleotide sequence that will specifically hybridize with a polynucleotide of the present invention and a suitable container. In a specific embodiment, the kit includes two polynucleotide probes defining an internal region of the polynucleotide of the present invention, where each probe has one strand containing a 31'mer-end internal to the region. In a further embodiment, the probes may be useful as primers for polymerase chain reaction amplification.
Where a diagnosis of a disorder, has already been made according to conventional methods, the present invention is useful as a prognostic indicator, whereby patients exhibiting enhanced or depressed polynucleotide of the present invention expression will experience a worse clinical outcome relative to patients expressing the gene at a level nearer the standard level.
By "measuring the expression level of polynucleotide of the present invention" is intended qualitatively or quantitatively measuring or estimating the level of the polypeptide of the present invention or the level of the mRNA encoding the polypeptide in a first biological sample either directly (e.g., by determining or estimating absolute protein level or mRNA level) or relatively (e.g., by comparing to the polypeptide level or mRNA level in a second biological sample).
Preferably, the polypeptide level or mRNA level in the first biological sample is measured or estimated and compared to a standard polypeptide level or mRNA level, the standard being taken from a second biological sample obtained from an individual not having the disorder or being determined by averaging levels from a population of individuals not having a disorder. As will be appreciated in the art, once a standard polypeptide level or mRNA level is known, it can be used repeatedly as a standard for comparison.
By "biological sample" is intended any biological sample obtained from an individual, body fluid, cell line, tissue culture, or other source which contains the polypeptide of the present invention or mRNA. As indicated, biological samples include body fluids (such as semen, lymph, sera, plasma, urine, synovial fluid and spinal fluid) which contain the polypeptide of the present invention, and other tissue sources found to express the polypeptide of the present invention. Methods for obtaining tissue biopsies and body fluids from mammals are well known in the art.
Where the biological sample is to include mRNA, a tissue biopsy is the preferred source.
The methods) provided above may preferrably be applied in a diagnostic method and/or kits in which polynucleotides and/or polypeptides are attached to a solid support. In one exemplary method, the support may be a "gene chip" or a "biological chip" as described in US Patents 5,837,832, 5,874,219, and 5,856,174.
Further, such a gene chip with polynucleotides of the present invention attached may be used to identify polymorphisms between the polynucleotide sequences, with polynucleotides isolated from a test subject. The knowledge of such polymorphisms (i.e. their location, as well as, their existence) would be beneficial in identifying disease loci for many disorders, including cancerous diseases and conditions.
Such a method is described in US Patents 5,858,659 and 5,856,104. The US Patents referenced supra are hereby incorporated by reference in their entirety herein.
The present invention encompasses polynucleotides of the present invention that are chemically synthesized, or reproduced as peptide nucleic acids (PNA), or according to other methods known in the art. The use of PNAs would serve as the preferred form if the polynucleotides are incorporated onto a solid support, or gene chip. For the purposes of the present invention, a peptide nucleic acid (PNA) is a polyamide type of DNA analog and the monomeric units for adenine, guanine, thymine and cytosine are available commercially (Perceptive Biosystems).
Certain components of DNA, such as phosphorus, phosphorus oxides, or deoxyribose derivatives, are not present in PNAs. As disclosed by P. E. Nielsen, M.
Egholm, R. H.
Berg and O. Buchardt, Science 254, 1497 (1991); and M. Egholm, O. Buchardt, L.Christensen, C. Behrens, S. M. Freier, D. A. Driver, R. H. Berg, S. K. Kim, B.
Norden, and P. E. Nielsen, Nature 365, 666 (1993), PNAs bind specifically and tightly to complementary DNA strands and are not degraded by nucleases. In fact, PNA binds more strongly to DNA than DNA itself does. This is probably because there is no electrostatic repulsion between the two strands, and also the polyamide backbone is more flexible. Because of this, PNA/DNA duplexes bind under a wider range of stringency conditions than DNA/DNA duplexes, making it easier to perform multiplex hybridization. Smaller probes can be used than with DNA due to the strong binding. In addition, it is more likely that single base mismatches can be determined with PNA/DNA hybridization because a single mismatch in a PNA/DNA 15-mer lowers the melting point (Tm) by 8°-20° C, vs. 4°-16° C for the DNA/DNA 1 S-mer duplex. Also, the absence of charge groups in PNA means that hybridization can be done at low ionic strengths and reduce possible interference by salt during the analysis.
The present invention is useful for detecting cancer in mammals. In particular the invention is useful during diagnosis of pathological cell proliferative neoplasias which include, but are not limited to: acute myelogenous leukemias including acute monocytic leukemia, acute myeloblastic leukemia, acute promyelocytic leukemia, acute myelomonocytic leukemia, acute erythroleukemia, acute megakaryocytic leukemia, and acute undifferentiated leukemia, etc.; and chronic myelogenous leukemias including chronic myelomonocytic leukemia, chronic granulocytic leukemia, etc. Preferred mammals include monkeys, apes, cats, dogs, cows, pigs, horses, rabbits and humans. Particularly preferred are humans.
Pathological cell proliferative diseases, disorders, and/or conditions are often associated with inappropriate activation of proto-oncogenes. (Gelmann, E. P.
et al., "The Etiology of Acute Leukemia: Molecular Genetics and Viral Oncology," in Neoplastic Diseases of the Blood, Vol l., Wiernik, P. H. et al. eds., 161-182 (1985)).
Neoplasias are now believed to result from the qualitative alteration of a normal cellular gene product, or from the quantitative modification of gene expression by insertion into the chromosome of a viral sequence, by chromosomal translocation of a gene to a more actively transcribed region, or by some other mechanism.
(Gelmann et al., supra) It is likely that mutated or altered expression of specific genes is S involved in the pathogenesis of some leukemias, among other tissues and cell types.
(Gelmann et al., supra) Indeed, the human counterparts of the oncogenes involved in some animal neoplasias have been amplified or translocated in some cases of human leukemia and carcinoma. (Gelmann et al., supra) For example, c-myc expression is highly amplified in the non-lymphocytic leukemia cell line HL-60. When HL-60 cells are chemically induced to stop proliferation, the level of c-myc is found to be downregulated. (International Publication Number WO
91/15580) However, it has been shown that exposure of HL-60 cells to a DNA
construct that is complementary to the 5' end of c-myc or c-myb blocks translation of the corresponding mRNAs which downregulates expression of the c-myc or c-myb proteins and causes arrest of cell proliferation and differentiation of the treated cells.
(International Publication Number WO 91/15580; Wickstrom et al., Proc. Natl.
Acad.
Sci. 85:1028 (1988); Anfossi et al., Proc. Natl. Acad. Sci. 86:3379 (1989)).
However, the skilled artisan would appreciate the present invention's usefulness would not be limited to treatment of proliferative diseases, disorders, and/or conditions of hematopoietic cells and tissues, in light of the numerous cells and cell types of varying origins which are known to exhibit proliferative phenotypes.
In addition to the foregoing, a polynucleotide can be used to control gene expression through triple helix formation or antisense DNA or RNA. Antisense techniques are discussed, for example, in Okano, J. Neurochem. 56: 560 (1991);
"Oligodeoxynucleotides as Antisense Inhibitors of Gene Expression,CRCPress, Boca Raton, FL (1988). Triple helix formation is discussed in, for instance Lee et al., Nucleic Acids Research 6: 3073 (1979); Cooney et al., Science 241: 456 (1988);
and Dervan et al., Science 251: 1360 (1991). Both methods rely on binding of the polynucleotide to a complementary DNA or RNA. For these techniques, preferred polynucleotides are usually oligonucleotides 20 to 40 bases in length and complementary to either the region of the gene involved in transcription (triple helix -see Lee et al., Nucl. Acids Res. 6:3073 (1979); Cooney et al., Science 241:456 (1988); and Dervan et al., Science 251:1360 (1991) ) or to the mRNA itself (antisense - Okano, J. Neurochem. 56:560 (1991); Oligodeoxy-nucleotides as Antisense Inhibitors of Gene Expression, CRC Press, Boca Raton, FL (1988).) Triple helix formation optimally results in a shut-off of RNA transcription from DNA, while antisense RNA hybridization blocks translation of an mRNA molecule into polypeptide. Both techniques are effective in model systems, and the information disclosed herein can be used to design antisense or triple helix polynucleotides in an effort to treat or prevent disease.
Polynucleotides of the present invention are also useful in gene therapy. One goal of gene therapy is to insert a normal gene into an.organism having a defective gene, in an effort to correct the genetic defect. The polynucleotides disclosed in the present invention offer a means of targeting such genetic defects in a highly accurate manner. Another goal is to insert a new gene that was not present in the host genome, thereby producing a new trait in the host cell.
The polynucleotides are also useful for identifying individuals from minute biological samples. The United States military, for example, is considering the use of restriction fragment length polymorphism (RFLP) for identification of its personnel.
In this technique, an individual's genomic DNA is digested with one or more restriction enzymes, and probed on a Southern blot to yield unique bands for identifying personnel. This method does not suffer from the current limitations of "Dog Tags" which can be lost, switched, or stolen, making positive identification difficult. The polynucleotides of the present invention can be used as additional DNA
markers for RFLP.
The polynucleotides of the present invention can also be used as an alternative to RFLP, by determining the actual base-by-base DNA sequence of selected portions of an individual's genome. These sequences can be used to prepare PCR primers for amplifying and isolating such selected DNA, which can then be sequenced. Using this technique, individuals can be identified because each individual will have a unique set of DNA sequences. Once an unique ID database is established for an individual, positive identification of that individual, living or dead, can be made from extremely small tissue samples.

Forensic biology also benefits from using DNA-based identification techniques as disclosed herein. DNA sequences taken from very small biological samples such as tissues, e.g., hair or skin, or body fluids, e.g., blood, saliva, semen, synovial fluid, amniotic fluid, breast milk, lymph, pulmonary sputum or surfactant,urine,fecal matter, etc., can be amplified using PCR. In one prior art technique, gene sequences amplified from polymorphic loci, such as DQa class II
HLA gene, are used in forensic biology to identify individuals. (Erlich, H., PCR
Technology, Freeman and Co. (1992).) Once these specific polymorphic loci are amplified, they are digested with one or more restriction enzymes, yielding an identifying set of bands on a Southern blot probed with DNA corresponding to the DQa class II HLA gene. Similarly, polynucleotides of the present invention can be used as polymorphic markers for forensic purposes.
There is also a need for reagents capable of identifying the source of a particular tissue. Such need arises, for example, in forensics when presented with tissue of unknown origin. Appropriate reagents can comprise, for example, DNA
probes or primers specific to particular tissue prepared from the sequences of the present invention. Panels of such reagents can identify tissue by species and/or by organ type. In a similar fashion, these reagents can be used to screen tissue cultures for contamination.
In the very least, the polynucleotides of the present invention can be used as molecular weight markers on Southern gels, as diagnostic probes for the presence of a specific mRNA in a particular cell type, as a probe to "subtract-out" known sequences in the process of discovering novel polynucleotides, for selecting and making oligomers for attachment to a "gene chip" or other support, to raise anti-DNA
antibodies using DNA immunization techniques, and as an antigen to elicit an immune response.
Uses of the Polypentides Each of the polypeptides identified herein can be used in numerous ways. The following description should be considered exemplary and utilizes known techniques.
A polypeptide of the present invention can be used to assay protein levels in a biological sample using antibody-based techniques. For example, protein expression in tissues can be studied with classical immunohistological methods.
(Jalkanen, M., et al., J. Cell. Biol. 101:976-985 (1985); Jalkanen, M., et al., J. Cell .
Biol. 105:3087-3096 (1987).) Other antibody-based methods useful for detecting protein gene expression include immunoassays, such as the enzyme linked immunosorbent assay S (ELISA) and the radioimmunoassay (RIA). Suitable antibody assay labels are known in the art and include enzyme labels, such as, glucose oxidase, and radioisotopes, such as iodine (125I, 121I), carbon (14C), sulfur (35S), tritium (3H), indium (112In), and technetium (99mTc), and fluorescent labels, such as fluorescein and rhodamine, and biotin.
In addition to assaying secreted protein levels in a biological sample, proteins can also be detected in vivo by imaging. Antibody labels or markers for in vivo imaging of protein include those detectable by X-radiography, NMR or ESR. For X-radiography, suitable labels include radioisotopes such as barium or cesium, which emit detectable radiation but are not overtly harmful to the subject. Suitable markers for NMR and ESR include those with a detectable characteristic spin, such as deuterium, which may be incorporated into the antibody by labeling of nutrients for the relevant hybridoma.
A protein-specific antibody or antibody fragment which has been labeled with an appropriate detectable imaging moiety, such as a radioisotope (for example, 131I, 1 l2ln, 99mTc), a radio-opaque substance, or a material detectable by nuclear magnetic resonance, is introduced (for example, parenterally, subcutaneously, or intraperitoneally) into the mammal. It will be understood in the art that the size of the subject and the imaging system used will determine the quantity of imaging moiety needed to produce diagnostic images. In the case of a radioisotope moiety, for a human subject, the quantity of radioactivity injected will normally range from about S
to 20 millicuries of 99mTc. The labeled antibody or antibody fragment will then preferentially accumulate at the location of cells which contain the specific protein.
In vivo tumor imaging is described in S.W. Burchiel et al., "Immunopharmacokinetics of Radiolabeled Antibodies and Their Fragments." (Chapter 13 in Tumor Imaging:
The Radiochemical Detection of Cancer, S.W. Burchiel and B. A. Rhodes, eds., Masson Publishing Inc. (1982).) Thus, the invention provides a diagnostic method of a disorder, which involves (a) assaying the expression of a polypeptide of the present invention in cells or body fluid of an individual; (b) comparing the level of gene expression with a standard gene expression level, whereby an increase or decrease in the assayed polypeptide gene expression level compared to the standard expression level is indicative of a disorder. With respect to cancer, the presence of a relatively high amount of transcript in biopsied tissue from an individual may indicate a predisposition for the development of the disease, or may provide a means for detecting the disease prior to the appearance of actual clinical symptoms. A
more definitive diagnosis of this type may allow health professionals to employ preventative measures or aggressive treatment earlier thereby preventing the development or further progression of the cancer.
Moreover, polypeptides of the present invention can be used to treat, prevent, and/or diagnose disease. For example, patients can be administered a polypeptide of the present invention in an effort to replace absent or decreased levels of the polypeptide (e.g., insulin), to supplement absent or decreased levels of a different polypeptide (e.g., hemoglobin S for hemoglobin B, SOD, catalase, DNA repair proteins), to inhibit the activity of a polypeptide (e.g., an oncogene or tumor supressor), to activate the activity of a polypeptide (e.g., by binding to a receptor), to reduce the activity of a membrane bound receptor by competing with it for free ligand (e.g., soluble TNF receptors used in reducing inflammation), or to bring about a desired response (e.g., blood vessel growth inhibition, enhancement of the immune response to proliferative cells or tissues).
Similarly, antibodies directed to a polypeptide of the present invention can also be used to treat, prevent, and/or diagnose disease. For example, administration of an antibody directed to a polypeptide of the present invention can bind and reduce overproduction of the polypeptide. Similarly, administration of an antibody can activate the polypeptide, such as by binding to a polypeptide bound to a membrane (receptor).
At the very least, the polypeptides of the present invention can be used as molecular weight markers on SDS-PAGE gels or on molecular sieve gel filtration columns using methods well known to those of skill in the art. Polypeptides can also be used to raise antibodies, which in turn are used to measure protein expression from a recombinant cell, as a way of assessing transformation of the host cell.
Moreover, the polypeptides of the present invention can be used to test the following biological activities.
Gene Therapy Methods Another aspect of the present invention is to gene therapy methods for treatingor preventing disorders, diseases and conditions. The gene therapy methods relate to the introduction of nucleic acid (DNA, RNA and antisense DNA or RNA) sequences into an animal to achieve expression of a polypeptide of the present invention. This method requires a polynucleotide which codes for a polypeptide of the invention that operatively linked to a promoter and any other genetic elements necessary for the expression of the polypeptide by the target tissue. Such gene therapy and delivery techniques are known in the art, see, for example, W090/11092, which is herein incorporated by reference.
Thus, for example, cells from a patient may be engineered with a polynucleotide (DNA or RNA) comprising a promoter operably linked to a polynucleotide of the invention ex vivo, with the engineered cells then being provided to a patient to be treated with the polypeptide. Such methods are well-known in the art. For example, see Belldegrun et al., J. Natl. Cancer Inst., 85:207-216 (1993);
Ferrantini et al., Cancer Research, 53:107-1112 (1993); Ferrantini et al., J.
Immunology 153: 4604-4615 (1994); Kaido, T., et al., Int. J. Cancer 60: 221-(1995); Ogura et al., Cancer Research 50: 5102-5106 (1990); Santodonato, et al., Human Gene Therapy 7:1-10 (1996); Santodonato, et al., Gene Therapy 4:1246-(1997); and Zhang, et al., Cancer Gene Therapy 3: 31-38 (1996)), which are herein incorporated by reference. In one embodiment, the cells which are engineered are arterial cells. The arterial cells may be reintroduced into the patient through direct injection to the artery, the tissues surrounding the artery, or through catheter injection.
As discussed in more detail below, the polynucleotide constructs can be delivered by any method that delivers injectable materials to the cells of an animal, such as, injection into the interstitial space of tissues (heart, muscle, skin, lung, liver, and the like). The polynucleotide constructs may be delivered in a pharmaceutically acceptable liquid or aqueous carrier.
In one embodiment, the polynucleotide of the invention is delivered as a naked polynucleotide. The term "naked" polynucleotide, DNA or RNA refers to sequences that are free from any delivery vehicle that acts to assist, promote or facilitate entry into the cell, including viral sequences, viral particles, liposome formulations, lipofectin or precipitating agents and the like. However, the polynucleotides of the invention can also be delivered in liposome formulations and lipofectin formulations and the like can be prepared by methods well known to those skilled in the art. Such methods are described, for example, in U.S. Patent Nos. 5,593,972, 5,589,466, and 5,580,859, which are herein incorporated by reference.
The polynucleotide vector constructs of the invention used in the gene therapy method are preferably constructs that will not integrate into the host genome nor will they contain sequences that allow for replication. Appropriate vectors include pWLNEO, pSV2CAT, pOG44, pXTI and pSG available from Stratagene;
pSVK3, pBPV, pMSG and pSVL available from Pharmacia; and pEFl/V5, pcDNA3.l, and pRc/CMV2 available from Invitrogen. Other suitable vectors will be readily apparent to the skilled artisan.
Any strong promoter known to those skilled in the art can be used for driving the expression of polynucleotide sequence of the invention. Suitable promoters include adenoviral promoters, such as the adenoviral major late promoter; or heterologous promoters, such as the cytomegalovirus (CMV) promoter; the respiratory syncytial virus (RSV) promoter; inducible promoters, such as the MMT
promoter, the metallothionein promoter; heat shock promoters; the albumin promoter;
the ApoAI promoter; human globin promoters; viral thymidine kinase promoters, such as the Herpes Simplex thymidine kinase promoter;.retroviral LTRs; the b-actin promoter; and human growth hormone promoters. The promoter also may be the native promoter for the polynucleotides of the invention.
Unlike other gene therapy techniques, one major advantage of introducing naked nucleic acid sequences into target cells is the transitory nature of the polynucleotide synthesis in the cells. Studies have shown that non-replicating DNA

sequences can be introduced into cells to provide production of the desired polypeptide for periods of up to six months.
The polynucleotide construct of the invention can be delivered to the interstitial space of tissues within the an animal, including of muscle, skin, brain, lung, liver, spleen, bone marrow, thymus, heart, lymph, blood, bone, cartilage, pancreas, kidney, gall bladder, stomach, intestine, testis, ovary, uterus, rectum, nervous system, eye, gland, and connective tissue. Interstitial space of the tissues comprises the intercellular, fluid, mucopolysaccharide matrix among the reticular fibers of organ tissues, elastic fibers in the walls of vessels or chambers, collagen fibers of fibrous tissues, or that same matrix within connective tissue ensheathing muscle cells or in the lacunae of bone. It is similarly the space occupied by the plasma of the circulation and the lymph fluid of the lymphatic channels. Delivery to the interstitial space of muscle tissue is preferred for the reasons discussed below. They may be conveniently delivered by injection into the tissues comprising these cells. They are preferably delivered to and expressed in persistent, non-dividing cells which are differentiated, although delivery and expression may be achieved in non-differentiated or less completely differentiated cells, such as, for example, stem cells of blood or skin fibroblasts. In vivo muscle cells are particularly competent in their ability to take up and express polynucleotides.
For the nakednucleic acid sequence injection, an effective dosage amount of DNA or RNA will be in the range of from about 0.05 mg/kg body weight to about mg/kg body weight. Preferably the dosage will be from about 0.005 mg/kg to about 20 mg/kg and more preferably from about 0.05 mg/kg to about 5 mg/kg. Of course, as the artisan of ordinary skill will appreciate, this dosage will vary according to the tissue site of injection. The appropriate and effective dosage of nucleic acid sequence can readily be determined by those of ordinary skill in the art and may depend on the condition being treated and the route of administration.
The preferred route of administration is by the parenteral route of injection into the interstitial space of tissues. However, other parenteral routes may also be used, such as, inhalation of an aerosol formulation particularly for delivery to lungs or bronchial tissues, throat or mucous membranes of the nose. In addition, naked DNA
constructs can be delivered to arteries during angioplasty by the catheter used in the procedure.

The naked polynucleotides are delivered by any method known in the art, including, but not limited to, direct needle injection at the delivery site, intravenous injection, topical administration, catheter infusion, and so-called "gene guns". These delivery methods are known in the art.
The constructs may also be delivered with delivery vehicles such as viral sequences, viral particles, liposome formulations, lipofectin, precipitating agents, etc.
Such methods of delivery are known in the art.
In certain embodiments, the polynucleotide constructs of the invention are complexed in a liposome preparation. Liposomal preparations for use in the instant invention include cationic (positively charged), anionic (negatively charged) and neutral preparations. However, cationic liposomes are particularly preferred because a tight charge complex can be formed between the cationic liposome and the polyanionic nucleic acid. Cationic liposomes have been shown to mediate intracellular delivery of plasmid DNA (Felgner et al., Proc. Natl. Acad. Sci.
USA , 84:7413-7416 (1987), which is herein incorporated by reference); mRNA (Malone et al., Proc. Natl. Acad. Sci. USA , 86:6077-6081 (1989), which is herein incorporated by reference); and purified transcription factors (Debs et al., J. Biol.
Chem., 265:10189-10192 (1990), which is herein incorporated by reference), in functional form.
Cationic liposomes are readily available. For example, N[1-2,3-dioleyloxy)propyl]-N,N,N-triethylammonium (DOTMA) liposomes are particularly useful and are available under the trademark Lipofectin, from GIBCO
BRL, Grand Island, N.Y. (See, also, Felgner et al., Proc. Natl Acad. Sci. USA
, 84:7413-7416 (1987), which is herein incorporated by reference). Other commercially available liposomes include transfectace (DDAB/DOPE) and DOTAP/DOPE
(Boehringer).
Other cationic liposomes can be prepared from readily available materials using techniques well known in the art. See, e.g. PCT Publication NO: WO

(which is herein incorporated by reference) for a description of the synthesis of DOTAP (1,2-bis(oleoyloxy)-3-(trimethylammonio)propane) liposomes. Preparation of DOTMA liposomes is explained in the literature, see, e.g., Felgner et al., Proc.

Natl. Acad. Sci. USA, 84:7413-7417, which is herein incorporated by reference.
Similar methods can be used to prepare liposomes from other cationic lipid materials.
Similarly, anionic and neutral liposomes are readily available, such as from Avanti Polar Lipids (Birmingham, Ala.), or can be easily prepared using readily available materials. Such materials include phosphatidyl, choline, cholesterol, phosphatidyl ethanolamine, dioleoylphosphatidyl choline (DOPC), dioleoylphosphatidyl glycerol (DOPG), dioleoylphoshatidyl ethanolamine (DOPE), among others. These materials can also be mixed with the DOTMA and DOTAP
starting materials in appropriate ratios. Methods for making liposomes using these materials are well known in the art.
For example, commercially dioleoylphosphatidyl choline (DOPC), dioleoylphosphatidyl glycerol (DOPG), and dioleoylphosphatidyl ethanolamine (DOPE) can be used in various combinations to make conventional liposomes, with or without the addition of cholesterol. Thus, for example, DOPG/DOPC vesicles can be prepared by drying SO mg each of DOPG and DOPC under a stream of nitrogen gas into a sonication vial. The sample is placed under a vacuum pump overnight and is hydrated the following day with deionized water. The sample is then sonicated for 2 hours in a capped vial, using a Heat Systems model 350 sonicator equipped with an inverted cup (bath type) probe at the maximum setting while the bath is circulated at 15EC. Alternatively, negatively charged vesicles can be prepared without sonication to produce multilamellar vesicles or by extrusion through nucleopore membranes to produce unilamellar vesicles of discrete size. Other methods are known and available to those of skill in the art.
The liposomes can comprise multilamellar vesicles (MLVs), small unilamellar vesicles (SUVs), or large unilamellar vesicles (LUVs), with SUVs being preferred.
The various liposome-nucleic acid complexes are prepared using methods well known in the art. See, e.g., Straubinger et al., Methods of Immunology , 101:512-527 (1983), which is herein incorporated by reference. For example, MLVs containing nucleic acid can be prepared by depositing a thin film of phospholipid on the walls of a glass tube and subsequently hydrating with a solution of the material to be encapsulated.
SUVs are prepared by extended sonication of MLVs to produce a homogeneous population of unilamellar liposomes. The material to be entrapped is added to a suspension of preformed MLVs and then sonicated. When using liposomes containing cationic lipids, the dried lipid film is resuspended in an appropriate solution such as sterile water or an isotonic buffer solution such as 10 mM Tris/NaCI, sonicated, and then the preformed liposomes are mixed directly with the DNA. The liposome and DNA form a very stable complex due to binding of the positively charged liposomes to the cationic DNA. SUVs find use with small nucleic acid fragments. LUVs are prepared by a number of methods, well known in the art. Commonly used methods include Ca2+-EDTA chelation (Papahadjopoulos et al., Biochim. Biophys. Acta, 394:483 (1975); Wilson et al., Cell , 17:77 (1979)); ether injection (Deamer et al., Biochim. Biophys. Acta, 443:629 (1976); Ostro et al., Biochem. Biophys. Res.
Commun., 76:836 (1977); Fraley et al., Proc. Natl. Acad. Sci. USA, 76:3348 (1979));
detergent dialysis (Enoch et al., Proc. Natl. Acad. Sci. USA , 76:145 (1979));
and reverse-phase evaporation (REV) (Fraley et al., J. Biol. Chem., 255:10431 (1980);
Szoka et al., Proc. Natl. Acad. Sci. USA , 75:145 (1978); Schaefer-Ridder et al., Science, 215:166 (1982)), which are herein incorporated by reference.
Generally, the ratio of DNA to liposomes will be from about 10:1 to about 1:10. Preferably, the ration will be from about 5:1 to about 1:5. More preferably, the ration will be about 3:1 to about 1:3. Still more preferably, the ratio will be about 1:1.
U.S. Patent NO: 5,676,954 (which is herein incorporated by reference) reports on the injection of genetic material, complexed with cationic liposomes earners, into mice. U.5. Patent Nos. 4,897,355, 4,946,787, 5,049,386, 5,459,127, 5,589,466, 5,693,622, 5,580,859, 5,703,055, and international publication NO: WO 94/9469 (which are herein incorporated by reference) provide cationic lipids for use in transfecting DNA into cells and mammals. U.S. Patent Nos. 5,589,466, 5,693,622, 5,580,859, 5,703,055, and international publication NO: WO 94/9469 (which are herein incorporated by reference) provide methods for delivering DNA-cationic lipid complexes to mammals.
In certain embodiments, cells are engineered, ex vivo or in vivo, using a retroviral particle containing RNA which comprises a sequence encoding polypeptides of the invention. Retroviruses from which the retroviral plasmid vectors may be derived include, but are not limited to, Moloney Murine Leukemia Virus, spleen necrosis virus, Rous sarcoma Virus, Harvey Sarcoma Virus, avian leukosis virus, gibbon ape leukemia virus, human immunodeficiency virus, Myeloproliferative Sarcoma Virus, and mammary tumor virus.
The retroviral plasmid vector is employed to transduce packaging cell lines to form producer cell lines. Examples of packaging cells which may be transfected include, but are not limited to, the PE501, PA317, R-2, R-AM, PA12, T19-14X, VT-19-17-H2, RCRE, RCRII', GP+E-86, GP+envAml2, and DAN cell lines as described in Miller, Human Gene Therapy , 1:5-14 (1990), which is incorporated herein by reference in its entirety. The vector may transduce the packaging cells through any means known in the art. Such means include, but are not limited to, electroporation, the use of liposomes, and CaP04 precipitation. In one alternative, the retroviral plasmid vector may be encapsulated into a liposome, or coupled to a lipid, and then administered to a host.
The producer cell line generates infectious retroviral vector particles which include polynucleotide encoding polypeptides of the invention. Such retroviral vector particles then may be employed, to transduce eukaryotic cells, either in vitro or in vivo. The transduced eukaryotic cells will express polypeptides of the invention.
In certain other embodiments, cells are engineered, ex vivo or in vivo, with polynucleotides of the invention contained in an adenovirus vector. Adenovirus can be manipulated such that it encodes and expresses polypeptides of the invention, and at the same time is inactivated in terms of its ability to replicate in a normal lytic viral life cycle. Adenovirus expression is achieved without integration of the viral DNA
into the host cell chromosome, thereby alleviating concerns about insertional mutagenesis. Furthermore, adenoviruses have been used as live enteric vaccines for many years with an excellent safety profile (Schwartzet al., Am. Rev. Respir.
Dis., 109:233-238 (1974)). Finally, adenovirus mediated gene transfer has been demonstrated in a number of instances including transfer of alpha-1-antitrypsin and CFTR to the lungs of cotton rats (Rosenfeld et al.,Science , 252:431-434 (1991);
Rosenfeld et al., Cell, 68:143-155 (1992)). Furthermore, extensive studies to attempt to establish adenovirus as a causative agent in human cancer were uniformly negative (Green et al. Proc. Natl. Acad. Sci. USA , 76:6606 (1979)).
Suitable adenoviral vectors useful in the present invention are described, for example, in Kozarsky and Wilson, Curr. Opin. Genet. Devel., 3:499-503 (1993);

Rosenfeld et al., Cell , 68:143-155 (1992); Engelhardt et al., Human Genet.
Ther., 4:759-769 (1993); Yang et al., Nature Genet., 7:362-369 (1994); Wilson et al., Nature , 365:691-692 (1993); and U.S. Patent NO: 5,652,224, which are herein incorporated by reference. For example, the adenovirus vector Ad2 is useful and can be grown in human 293 cells. These cells contain the E1 region of adenovirus and constitutively express Ela and Elb, which complement the defective adenoviruses by providing the products of the genes deleted from the vector. In addition to Ad2, other varieties of adenovirus (e.g., Ad3, AdS, and Ad7) are also useful in the present invention.
Preferably, the adenoviruses used in the present invention are replication deficient. Replication deficient adenoviruses require the aid of a helper virus and/or packaging cell line to form infectious particles. The resulting virus is capable of infecting cells and can express a polynucleotide of interest which is operably linked to a promoter, but cannot replicate in most cells. Replication deficient adenoviruses may be deleted in one or more of all or a portion of the following genes: Ela, Elb, E3, E4, E2a, or Ll through L5.
In certain other embodiments, the cells are engineered, ex vivo or in vivo, using an adeno-associated virus (AAV). AAVs are naturally occurring defective viruses that require helper viruses to produce infectious particles (Muzyczka, Curr.
Topics in Microbiol. Immunol., 158:97 (1992)). It is also one of the few viruses that may integrate its DNA into non-dividing cells. Vectors containing as little as 300 base pairs of AAV can be packaged and can integrate, but space for exogenous DNA is limited to about 4.5 kb. Methods for producing and using such AAVs are known in the art. See, for example, U.S. Patent Nos. 5,139,941, 5,173,414, 5,354,678, 5,436,146, 5,474,935, 5,478,745, and 5,589,377.
For example, an appropriate AAV vector for use in the present-invention will include all the sequences necessary for DNA replication, encapsidation, and host-cell integration. The polynucleotide construct containing polynucleotides of the invention is inserted into the AAV vector using standard cloning methods, such as those found in Sambrook et al., Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Press (1989). The recombinant AAV vector is then transfected into packaging cells which are infected with a helper virus, using any standard technique, including lipofection, electroporation, calcium phosphate precipitation, etc.
Appropriate helper viruses include adenoviruses, cytomegaloviruses, vaccinia viruses, or herpes viruses.
Once the packaging cells are transfected and infected, they will produce infectious AAV viral particles which contain the polynucleotide construct of the invention.
These viral particles are then used to transduce eukaryotic cells, either ex vivo or in vivo. The transduced cells will contain the polynucleotide construct integrated into its genome, and will express the desired gene product.
Another method of gene therapy involves operably associating heterologous control regions and endogenous polynucleotide sequences (e.g. encoding the polypeptide sequence of interest) via homologous recombination (see, e.g., U.S.
Patent NO: 5,641,670, issued June 24, 1997; International Publication NO: WO
96/29411, published September 26, 1996; International Publication NO: WO
94/12650, published August 4, 1994; Koller et al., Proc. Natl. Acad. Sci. USA, 86:8932-8935 (1989); and Zijlstra et al., Nature, 342:435-438 (1989). This method involves the activation of a gene which is present in the target cells, but which is not normally expressed in the cells, or is expressed at a lower level than desired.
Polynucleotide constructs are made, using standard techniques known in the art, which contain the promoter with targeting sequences flanking the promoter.
Suitable promoters are described herein. The targeting sequence is sufficiently complementary to an endogenous sequence to permit homologous recombination of the promoter-targeting sequence with the endogenous sequence. The targeting sequence will be sufficiently near the 5' end of the desired endogenous polynucleotide sequence so the promoter will be operably linked to the endogenous sequence upon homologous recombination.
The promoter and the targeting sequences can be amplified using PCR.
Preferably, the amplified promoter contains distinct restriction enzyme sites on the 5' and 3' ends. Preferably, the 3' end of the first targeting sequence contains the same restriction enzyme site as the 5' end of the amplified promoter and the 5' end of the second targeting sequence contains the same restriction site as the 3' end of the amplified promoter. The amplified promoter and targeting sequences are digested and ligated together.

The promoter-targeting sequence construct is delivered to the cells, either as naked polynucleotide, or in conjunction with transfection-facilitating agents, such as liposomes, viral sequences, viral particles, whole viruses, lipofection, precipitating agents, etc., described in more detail above. The P promoter-targeting sequence can be delivered by any method, included direct needle injection, intravenous injection, topical administration, catheter infusion, particle accelerators, etc. The methods are described in more detail below.
The promoter-targeting sequence construct is taken up by cells. Homologous recombination between the construct and the endogenous sequence takes place, such that an endogenous sequence is placed under the control of the promoter. The promoter then drives the expression of the endogenous sequence.
The polynucleotides encoding polypeptides of the present invention may be administered along with other polynucleotides encoding other angiongenic proteins.
Angiogenic proteins include, but are not limited to, acidic and basic fibroblast growth factors, VEGF-1, VEGF-2 (VEGF-C), VEGF-3 (VEGF-B), epidermal growth factor alpha and beta, platelet-derived endothelial cell growth factor, platelet-derived growth factor, tumor necrosis factor alpha, hepatocyte growth factor, insulin like growth factor, colony stimulating factor, macrophage colony stimulating factor, granulocyte/macrophage colony stimulating factor, and nitric oxide synthase.
Preferably, the polynucleotide encoding a polypeptide of the invention contains a secretory signal sequence that facilitates secretion of the protein.
Typically, the signal sequence is positioned in the coding region of the polynucleotide to be expressed towards or at the 5' end of the coding region. The signal sequence may be homologous or heterologous to the polynucleotide of interest and may be homologous or heterologous to the cells to be transfected. Additionally, the signal sequence may be chemically synthesized using methods known in the art.
Any mode of administration of any of the above-described polynucleotides constructs can be used so long as the mode results in the expression of one or more molecules in an amount sufficient to provide a therapeutic effect. This includes direct needle injection, systemic injection, catheter infusion, biolistic injectors, particle accelerators (i.e., "gene guns"), gelfoam sponge depots, other commercially available depot materials, osmotic pumps (e.g., Alza minipumps), oral or suppositorial solid (tablet or pill) pharmaceutical formulations, and decanting or topical applications during surgery. For example, direct injection of naked calcium phosphate-precipitated plasmid into rat liver and rat spleen or a protein-coated plasmid into the portal vein has resulted in gene expression of the foreign gene in the rat livers. (Kaneda et al., Science, 243:375 (1989)).
A preferred method of local administration is by direct injection. Preferably, a recombinant molecule of the present invention complexed with a delivery vehicle is administered by direct injection into or locally within the area of arteries.
Administration of a composition locally within the area of arteries refers to injecting the composition centimeters and preferably, millimeters within arteries.
Another method of local administration is to contact a polynucleotide construct of the present invention in or around a surgical wound. For example, a patient can undergo surgery and the polynucleotide construct can be coated on the surface of tissue inside the wound or the construct can be injected into areas of tissue inside the wound.
Therapeutic compositions useful in systemic administration, include recombinant molecules of the present invention complexed to a targeted delivery vehicle of the present invention. Suitable delivery vehicles for use with systemic administration comprise liposomes comprising ligands for targeting the vehicle to a particular site.
Preferred methods of systemic administration, include intravenous injection, aerosol, oral and percutaneous (topical) delivery. Intravenous injections can be performed using methods standard in the art. Aerosol delivery can also be performed using methods standard in the art (see, for example, Stribling et al., Proc.
Natl. Acad.
Sci. USA , 189:11277-11281 (1992), which is incorporated herein by reference).
Oral delivery can be performed by complexing a polynucleotide construct of the present invention to a carrier capable of withstanding degradation by digestive enzymes in the gut of an animal. Examples of such carriers, include plastic capsules or tablets, such as those known in the art. Topical delivery can be performed by mixing a polynucleotide construct of the present invention with a lipophilic reagent (e.g., DMSO) that is capable of passing into the skin.

Determining an effective amount of substance to be delivered can depend upon a number of factors including, for example, the chemical structure and biological activity of the substance, the age and weight of the animal, the precise condition requiring treatment and its severity, and the route of administration. The frequency of treatments depends upon a number of factors, such as the amount of polynucleotide constructs administered per dose, as well as the health and history of the subject. The precise amount, number of doses, and timing of doses will be determined by the attending physician or veterinarian. Therapeutic compositions of the present invention can be administered to any animal, preferably to mammals and birds. Preferred mammals include humans, dogs, cats, mice, rats, rabbits sheep, cattle, horses and pigs, with humans being particularly Biological Activities The polynucleotides or polypeptides, or agonists or antagonists of the present invention can be used in assays to test for one or more biological activities.
If these polynucleotides and polypeptides do exhibit activity in a particular assay, it is likely that these molecules may be involved in the diseases associated with the biological activity. Thus, the polynucleotides or polypeptides, or agonists or antagonists could be used to treat the associated disease.
Immune Activity Polynucleotides, polypeptides, antibodies, and/or agonists or antagonists of the present invention may be useful in treating, preventing, and/or diagnosing diseases, disorders, and/or conditions of the immune system, by, for example, activating or inhibiting the proliferation, differentiation, or mobilization (chemotaxis) of immune cells. Immune cells develop through a process called hematopoiesis, producing myeloid (platelets, red blood cells, neutrophils, and macrophages) and lymphoid (B
and T lymphocytes) cells from pluripotent stem cells. The etiology of these immune diseases, disorders, and/or conditions may be genetic, somatic, such as cancer and some autoimmune diseases, acquired (e.g., by chemotherapy or toxins), or infectious.
Moreover, polynucleotides, polypeptides, antibodies, and/or agonists or antagonists of the present invention can be used as a marker or detector of a particular immune system disease or disorder.
Polynucleotides, polypeptides, antibodies, and/or agonists or antagonists of the present invention may be useful in treating, preventing, and/or diagnosing diseases, disorders, and/or conditions of hematopoietic cells. Polynucleotides, polypeptides, antibodies, and/or agonists or antagonists of the present invention could be used to increase differentiation and proliferation of hematopoietic cells, including the pluripotent stem cells, in an effort to treat or prevent those diseases, disorders, and/or conditions associated with a decrease in certain (or many) types hematopoietic cells.
Examples of immunologic deficiency syndromes include, but are not limited to:
blood protein diseases, disorders, and/or conditions (e.g., agammaglobulinemia, dysgammaglobulinemia), ataxia telangiectasia, common variable immunodeficiency, Digeorge Syndrome, HN infection, HTLV-BLV infection, leukocyte adhesion deficiency syndrome, lymphopenia, phagocyte bactericidal dysfunction, severe combined immunodeficiency (SC>Ds), Wiskott-Aldrich Disorder, anemia, thrombocytopenia, or hemoglobinuria.
Moreover, polynucleotides, polypeptides, antibodies, and/or agonists or antagonists of the present invention could also be used to modulate hemostatic (the stopping of bleeding) or thrombolytic activity (clot formation). For example, by increasing hemostatic or thrombolytic activity, polynucleotides or polypeptides, and/or agonists or antagonists of the present invention could be used to treat or prevent blood coagulation diseases, disorders, and/or conditions (e.g., afibrinogenemia, factor deficiencies), blood platelet diseases, disorders, and/or conditions (e.g., thrombocytopenia), or wounds resulting from trauma, surgery, or other causes. Alternatively, polynucleotides, polypeptides, antibodies, and/or agonists or antagonists of the present invention that can decrease hemostatic or thrombolytic activity could be used to inhibit or dissolve clotting. These molecules could be important in the treatment or prevention of heart attacks (infarction), strokes, or scarring.
The polynucleotides, polypeptides, antibodies, and/or agonists or antagonists of the present invention may be useful in treating, preventing, and/or diagnosing autoimmune disorders. Many autoimmune disorders result from inappropriate recognition of self as foreign material by immune cells. This inappropriate recognition results in an immune response leading to the destruction of the host tissue.
Therefore, the administration of polynucleotides and polypeptides of the invention that can inhibit an immune response, particularly the proliferation, differentiation, or chemotaxis of T-cells, may be an effective therapy in preventing autoimmune disorders.
Autoimmune diseases or disorders that may be treated, prevented, and/or diagnosed by polynucleotides, polypeptides, antibodies, and/or agonists or antagonists of the present invention include, but are not limited to, one or more of the following:
autoimmune hemolytic anemia, autoimmune neonatal thrombocytopenia, idiopathic thrombocytopenia purpura, autoimmunocytopenia, hemolytic anemia, antiphospholipid syndrome, dermatitis, allergic encephalomyelitis, myocarditis, relapsing polychondritis, rheumatic heart disease, glomerulonephritis (e.g, IgA
nephropathy), Multiple Sclerosis, Neuritis, Uveitis Ophthalmia, Polyendocrinopathies, Purpura (e.g., Henloch-Scoenlein purpura), Reiter's Disease, Stiff Man Syndrome, Autoimmune Pulmonary Inflammation, Autism, Guillain-Barre Syndrome, insulin dependent diabetes mellitis, and autoimmune inflammatory eye, autoimmune thyroiditis, hypothyroidism (i.e., Hashimoto's thyroiditis, systemic lupus erhythematosus, Goodpasture's syndrome, Pemphigus, Receptor autoimmunities such as, for example, (a) Graves' Disease, (b) Myasthenia Gravis, and (c) insulin resistance, autoimmune hemolytic anemia, autoimmune thrombocytopenic purpura, rheumatoid arthritis, schleroderma with anti-collagen antibodies, mixed connective tissue disease, polymyositis/dermatomyositis, pernicious anemia, idiopathic Addison's disease, infertility, glomerulonephritis such as primary glomerulonephritis and IgA
nephropathy, bullous pemphigoid, Sjogren's syndrome, diabetes millitus, and adrenergic drug resistance (including adrenergic drug resistance with asthma or cystic fibrosis), chronic active hepatitis, primary biliary cirrhosis, other endocrine gland failure, vitiligo, vasculitis, post-MI, cardiotomy syndrome, urticaria, atopic dermatitis, asthma, inflammatory myopathies, and other inflammatory, granulamatous, degenerative, and atrophic disorders.
Additional autoimmune disorders (that are probable) that may be treated, prevented, and/or diagnosed with the compositions of the invention include, but are not limited to, rheumatoid arthritis (often characterized, e.g., by immune complexes in joints), scleroderma with anti-collagen antibodies (often characterized, e.g., by nucleolar and other nuclear antibodies), mixed connective tissue disease (often characterized, e.g., by antibodies to extractable nuclear antigens (e.g., ribonucleoprotein)), polymyositis (often characterized, e.g., by nonhistone ANA), pernicious anemia (often characterized, e.g., by antiparietal cell, microsomes, and intrinsic factor antibodies), idiopathic Addison's disease (often characterized, e.g., by humoral and cell-mediated adrenal cytotoxicity, infertility (often characterized, e.g., by antispermatozoal antibodies), glomerulonephritis (often characterized, e.g., by glomerular basement membrane antibodies or immune complexes), bullous pemphigoid (often characterized, e.g., by IgG and complement in basement membrane), Sjogren's syndrome (often characterized, e.g., by multiple tissue antibodies, and/or a specific nonhistone ANA (SS-B)), diabetes millitus (often characterized, e.g., by cell-mediated and humoral islet cell antibodies), and adrenergic drug resistance (including adrenergic drug resistance with asthma or cystic fibrosis) (often characterized, e.g., by beta-adrenergic receptor antibodies).
Additional autoimmune disorders (that are possible) that may be treated, prevented, and/or diagnosed with the compositions of the invention include, but are not limited to, chronic active hepatitis (often characterized, e.g., by smooth muscle antibodies), primary biliary cirrhosis (often characterized, e.g., by mitchondrial antibodies), other endocrine gland failure (often characterized, e.g., by specific tissue antibodies in some cases), vitiligo (often characterized, e.g., by melanocyte antibodies), vasculitis (often characterized, e.g., by Ig and complement in vessel walls and/or low serum complement), post-MI (often characterized, e.g., by myocardial antibodies), cardiotomy syndrome (often characterized, e.g., by myocardial antibodies), urticaria (often characterized, e.g., by IgG and IgM antibodies to IgE), atopic dermatitis (often characterized, e.g., by IgG and IgM antibodies to IgE), asthma (often characterized, e.g., by IgG and IgM antibodies to IgE), and many other inflammatory, granulamatous, degenerative, and atrophic disorders.
In a preferred embodiment, the autoimmune diseases and disorders and/or conditions associated with the diseases and disorders recited above are treated, prevented, and/or diagnosed using for example, antagonists or agonists, polypeptides or polynucleotides, or antibodies of the present invention.
In a preferred embodiment polynucleotides, polypeptides, antibodies, and/or agonists or antagonists of the present invention could be used as an agent to boost immunoresponsiveness among B cell and/or T cell immunodeficient individuals.
B cell immunodeficiencies that may be ameliorated or treated by administering the polypeptides or polynucleotides of the invention, and/or agonists thereof, include, but are not limited to, severe combined immunodeficiency (SCID)-X
linked, SCID-autosomal, adenosine deaminase deficiency (ADA deficiency), X-linked agammaglobulinemia (XLA), Bruton's disease, congenital agammaglobulinemia, X-linked infantile agammaglobulinemia, acquired agammaglobulinemia, adult onset agammaglobulinemia, late-onset agammaglobulinemia, dysgammaglobulinemia, hypogammaglobulinemia, transient hypogammaglobulinemia of infancy, unspecified hypogammaglobulinemia, agammaglobulinemia, common variable immunodeficiency (CVI) (acquired), Wiskott-Aldrich Syndrome (WAS), X-linked immunodeficiency with hyper IgM, non X-linked immunodeficiency with hyper IgM, selective IgA deficiency, IgG
subclass deficiency (with or without IgA deficiency), antibody deficiency with normal or elevated Igs, immunodeficiency with thymoma, Ig heavy chain deletions, kappa chain deficiency, B cell lymphoproliferative disorder (BLPD), selective IgM
immunodeficiency, recessive agammaglobulinemia (Swiss type), reticular dysgenesis, neonatal neutropenia, severe congenital leukopenia, thymic alymophoplasia-aplasia or dysplasia with immunodeficiency, ataxia-telangiectasia, short limbed dwarfism, X-linked lymphoproliferative syndrome (XLP), Nezelof syndrome-combined immunodeficiency with Igs, purine nucleoside phosphorylase deficiency (PNP), MHC Class II deficiency (Bare Lymphocyte Syndrome) and severe combined immunodeficiency.
T cell deficiencies that may be ameliorated or treated by administering the polypeptides or polynucleotides of the invention, and/or agonists thereof include, but are not limited to, for example, DiGeorge anomaly, thymic hypoplasia, third and fourth pharyngeal pouch syndrome, 22q11.2 deletion, chronic mucocutaneous candidiasis, natural killer cell deficiency (NK), idiopathic CD4+ T-lymphocytopenia, immunodeficiency with predominant T cell defect (unspecified), and unspecified immunodeficiency of cell mediated immunity. In specific embodiments, DiGeorge anomaly or conditions associated with DiGeorge anomaly are ameliorated or treated by, for example, administering the polypeptides or polynucleotides of the invention, or antagonists or agonists thereof.
Other immunodeficiencies that may be ameliorated or treated by administering polypeptides or polynucleotides of the invention, and/or agonists thereof, include, but are not limited to, severe combined immunodeficiency (SCID; e.g., X-linked SCID, autosomal SCID, and adenosine deaminase deficiency), ataxia-telangiectasia, Wiskott-Aldrich syndrome, short-limber dwarfism, X-linked lymphoproliferative syndrome (XLP), Nezelof syndrome (e.g., purine nucleoside phosphorylase deficiency), MHC Class II deficiency. In specific embodiments, ataxia-telangiectasia or conditions associated with ataxia-telangiectasia are ameliorated or treated by administering the polypeptides or polynucleotides of the invention, and/or agonists thereof.
In a specific preferred embodiment, rheumatoid arthritis is treated, prevented, and/or diagnosed using polynucleotides, polypeptides, antibodies, and/or agonists or antagonists of the present invention. In another specific preferred embodiment, systemic lupus erythemosus is treated, prevented, and/or diagnosed using polynucleotides, polypeptides, antibodies, and/or agonists or antagonists of the present invention. In another specific preferred embodiment, idiopathic thrombocytopenia purpura is treated, prevented, and/or diagnosed using polynucleotides, polypeptides, antibodies, and/or agonists or antagonists of the present invention. In another specific preferred embodiment IgA nephropathy is treated, prevented, and/or diagnosed using polynucleotides, polypeptides, antibodies, and/or agonists or antagonists of the present invention. In a preferred embodiment, the autoimmune diseases and disorders and/or conditions associated with the diseases and disorders recited above are treated, prevented, and/or diagnosed using antibodies against the protein of the invention.
Similarly, allergic reactions and conditions, such as asthma (particularly allergic asthma) or other respiratory problems, may also be treated, prevented, and/or diagnosed using polypeptides, antibodies, or polynucleotides of the invention, and/or agonists or antagonists thereof. Moreover, these molecules can be used to treat, prevent, and/or diagnose anaphylaxis, hypersensitivity to an antigenic molecule, or blood group incompatibility.
Moreover, inflammatory conditions may also be treated, diagnosed, and/or prevented with polynucleotides, polypeptides, antibodies, and/or agonists or antagonists of the present invention. Such inflammatory conditions include, but are not limited to, for example, respiratory disorders (such as, e.g., asthma and allergy);
gastrointestinal disorders (such as, e.g., inflammatory bowel disease);
cancers (such as, e.g., gastric, ovarian, lung, bladder, liver, and breast); CNS disorders (such as, e.g., multiple sclerosis, blood-brain barner permeability, ischemic brain injury and/or stroke, traumatic brain injury, neurodegenerative disorders (such as, e.g., Parkinson's disease and Alzheimer's disease), A>DS-related dementia, and prion disease);
cardiovascular disorders (such as, e.g., atherosclerosis, myocarditis, cardiovascular disease, and cardiopulmonary bypass complications); as well as many additional 1 S diseases, conditions, and disorders that are characterized by inflammation (such as, e.g., chronic hepatitis (B and C), rheumatoid arthritis, gout, trauma, septic shock, pancreatitis, sarcoidosis, dermatitis, renal ischemia-reperfusion injury, Grave's disease, systemic lupus erythematosis, diabetes mellitus (i.e., type 1 diabetes), and allogenic transplant rejection).
In specific embodiments, polypeptides, antibodies, or polynucleotides of the invention, and/or agonists or antagonists thereof, are useful to treat, diagnose, and/or prevent transplantation rejections, graft-versus-host disease, autoimmune and inflammatory diseases (e.g., immune complex-induced vasculitis, glomerulonephritis, hemolytic anemia, myasthenia gravis, type II collagen-induced arthritis, experimental allergic and hyperacute xenograft rejection, rheumatoid arthritis, and systemic lupus erythematosus (SLE). Organ rejection occurs by host immune cell destruction of the transplanted tissue through an immune response. Similarly, an immune response is also involved in GVHD, but, in this case, the foreign transplanted immune cells destroy the host tissues. Polypeptides, antibodies, or polynucleotides of the invention, and/or agonists or antagonists thereof, that inhibit an immune response, particularly the activation, proliferation, differentiation, or chemotaxis of T-cells, may be an effective therapy in preventing organ rejection or GVHD.

Similarly, polynucleotides, polypeptides, antibodies, and/or agonists or antagonists of the present invention may also be used to modulate and/or diagnose inflammation. For example, since polypeptides, antibodies, or polynucleotides of the invention, and/or agonists or antagonists of the invention may inhibit the activation, proliferation and/or differentiation of cells involved in an inflammatory response, these molecules can be used to treat, diagnose, or prognose, inflammatory conditions, both chronic and acute conditions, including, but not limited to, inflammation associated with infection (e.g., septic shock, sepsis, or systemic inflammatory response syndrome (SIRS)), ischemia-reperfusion injury, endotoxin lethality, arthritis, complement-mediated hyperacute rejection, nephritis, cytokine or chemokine induced lung injury, inflammatory bowel disease, Crohn's disease, and resulting from over production of cytokines (e.g., TNF or IL-1.).
Polypeptides, antibodies, polynucleotides and/or agonists or antagonists of the invention can be used to treat, detect, and/or prevent infectious agents. For example, by increasing the immune response, particularly increasing the proliferation activation and/or differentiation of B and/or T cells, infectious diseases may be treated, detected, and/or prevented. The immune response may be increased by either enhancing an existing immune response, or by initiating a new immune response.
Alternatively, polynucleotides, polypeptides, antibodies, and/or agonists or antagonists of the present invention may also directly inhibit the infectious agent (refer to section of application listing infectious agents, etc), without necessarily eliciting an immune response.
Additional preferred embodiments of the invention include, but are not limited to, the use of polypeptides, antibodies, polynucleotides and/or agonists or antagonists in the following applications:
Administration to an animal (e.g., mouse, rat, rabbit, hamster, guinea pig, pigs, micro-pig, chicken, camel, goat, horse, cow, sheep, dog, cat, non-human primate, and human, most preferably human) to boost the immune system to produce increased quantities of one or more antibodies (e.g., IgG, IgA, IgM, and IgE), to induce higher affinity antibody production (e.g., IgG, IgA, IgM, and IgE), and/or to increase an immune response.

Administration to an animal (including, but not limited to, those listed above, and also including transgenic animals) incapable of producing functional endogenous antibody molecules or having an otherwise compromised endogenous immune system, but which is capable of producing human immunoglobulin molecules by means of a reconstituted or partially reconstituted immune system from another animal (see, e.g., published PCT Application Nos. W098/24893, WO/9634096, WO/9633735, and WO/9110741.
A vaccine adjuvant that enhances immune responsiveness to specific antigen.
An adjuvant to enhance tumor-specific immune responses.
An adjuvant to enhance anti-viral immune responses. Anti-viral immune responses that may be enhanced using the compositions of the invention as an adjuvant, include virus and virus associated diseases or symptoms described herein or otherwise known in the art. In specific embodiments, the compositions of the invention are used as an adjuvant to enhance an immune response to a virus, disease, or symptom selected from the group consisting of AIDS, meningitis, Dengue, EBV, and hepatitis (e.g., hepatitis B). In another specific embodiment, the compositions of the invention are used as an adjuvant to enhance an immune response to a virus, disease, or symptom selected from the group consisting of HIV/A)DS, Respiratory syncytial virus, Dengue, Rotavirus, Japanese B encephalitis, Influenza A and B, Parainfluenza, Measles, Cytomegalovirus, Rabies, Junin, Chikungunya, Rift Valley fever, Herpes simplex, and yellow fever.
An adjuvant to enhance anti-bacterial or anti-fungal immune responses. Anti-bacterial or anti-fungal immune responses that may be enhanced using the compositions of the invention as an adjuvant, include bacteria or fungus and bacteria or fungus associated diseases or symptoms described herein or otherwise known in the art. In specific embodiments, the compositions of the invention are used as an .
adjuvant to enhance an immune response to a bacteria or fungus, disease, or symptom selected from the group consisting of: tetanus, Diphtheria, botulism, and meningitis type B. In another specific embodiment, the compositions of the invention are used as an adjuvant to enhance an immune response to a bacteria or fungus, disease, or symptom selected from the group consisting of: Vibrio cholerae, Mycobacterium leprae, Salmonella typhi, Salmonella paratyphi, Meisseria meningitidis, Streptococcus pneumoniae, Group B streptococcus, Shigella spp., Enterotoxigenic Escherichia coli, Enterohemorrhagic E. coli, Borrelia burgdorferi, and Plasmodium (malaria).
An adjuvant to enhance anti-parasitic immune responses. Anti-parasitic immune responses that may be enhanced using the compositions of the invention as an adjuvant, include parasite and parasite associated diseases or symptoms described herein or otherwise known in the art. In specific embodiments, the compositions of the invention are used as an adjuvant to enhance an immune response to a parasite. In another specific embodiment, the compositions of the invention are used as an adjuvant to enhance an immune response to Plasmodium (malaria).
As a stimulator of B cell responsiveness to pathogens.
As an activator of T cells.
As an agent that elevates the immune status of an individual prior to their receipt of immunosuppressive therapies.
As an agent to induce higher affinity antibodies.
As an agent to increase serum immunoglobulin concentrations.
As an agent to accelerate recovery of immunocompromised individuals.
As an agent to boost immunoresponsiveness among aged populations.
As an immune system enhancer prior to, during, or after bone marrow transplant and/or other transplants (e.g., allogeneic or xenogeneic organ transplantation). With respect to transplantation, compositions of the invention may be administered prior to, concomitant with, and/or after transplantation. In a specific embodiment, compositions of the invention are administered after transplantation, prior to the beginning of recovery of T-cell populations. In another specific embodiment, compositions of the invention are first administered after transplantation after the beginning of recovery of T cell populations, but prior to full recovery of B
cell populations.
As an agent to boost immunoresponsiveness among individuals having ' an acquired loss of B cell function. Conditions resulting in an acquired loss of B cell function that may be ameliorated or treated by administering the polypeptides, antibodies, polynucleotides and/or agonists or antagonists thereof, include, but are not limited to, HIV Infection, AIDS, bone marrow transplant, and B cell chronic lymphocytic leukemia (CLL).
As an agent to boost immunoresponsiveness among individuals having a temporary immune deficiency. Conditions resulting in a temporary immune deficiency that may be ameliorated or treated by administering the polypeptides, antibodies, polynucleotides and/or agonists or antagonists thereof, include, but are not limited to, recovery from viral infections (e.g., influenza), conditions associated with malnutrition, recovery from infectious mononucleosis, or conditions associated with stress, recovery from measles, recovery from blood transfusion, recovery from surgery.
As a regulator of antigen presentation by monocytes, dendritic cells, and/or B-cells. In one embodiment, polynucleotides, polypeptides, antibodies, and/or agonists or antagonists of the present invention enhance antigen presentation or antagonizes antigen presentation in vitro or in vivo. Moreover, in related embodiments, said enhancement or antagonization of antigen presentation may be useful as an anti-tumor treatment or to modulate the immune system.
As an agent to direct an individuals immune system towards development of a humoral response (i.e. TH2) as opposed to a TH1 cellular response.
As a means to induce tumor proliferation and thus make it more susceptible to anti-neoplastic agents. For example, multiple myeloma is a slowly dividing disease and is thus refractory to virtually all anti-neoplastic regimens. If these cells were forced to proliferate more rapidly their susceptibility profile would likely change.
As a stimulator of B cell production in pathologies such as AIDS, chronic lymphocyte disorder and/or Common Variable Immunodificiency.
As a therapy for generation and/or regeneration of lymphoid tissues following surgery, trauma or genetic defect.
As a gene-based therapy for genetically inherited disorders resulting in immuno-incompetence such as observed among SCII7 patients.
As an antigen for the generation of antibodies to inhibit or enhance immune mediated responses against polypeptides of the invention.
As a means of activating T cells.

As a means of activating monocytes/macrophages to defend against parasitic diseases that effect monocytes such as Leshmania.
As pretreatment of bone marrow samples prior to transplant. Such treatment would increase B cell representation and thus accelerate recover.
As a means of regulating secreted cytokines that are elicited by polypeptides of the invention.
Additionally, polypeptides or polynucleotides of the invention, and/or agonists thereof, may be used to treat or prevent IgE-mediated allergic reactions. Such allergic reactions include, but are not limited to, asthma, rhinitis, and eczema.
All of the above described applications as they may apply to veterinary medicine.
Antagonists of the invention include, for example, binding and/or inhibitory antibodies, antisense nucleic acids, or ribozymes. These would be expected to reverse many of the activities of the ligand described above as well as find clinical or practical application as:
A means of blocking various aspects of immune responses to foreign agents or self. Examples include autoimmune disorders such as lupus, and arthritis, as well as immunoresponsiveness to skin allergies, inflammation, bowel disease, injury and pathogens.
A therapy for preventing the B cell proliferation and Ig secretion associated with autoimmune diseases such as idiopathic thrombocytopenic purpura, systemic lupus erythramatosus and MS.
An inhibitor of B and/or T cell migration in endothelial cells. This activity disrupts tissue architecture or cognate responses and is useful, for example in disrupting immune responses, and blocking sepsis.
An inhibitor of graft versus host disease or transplant rejection.
A therapy for B cell and/or T cell malignancies such as ALL, Hodgkins disease, non-Hodgkins lymphoma, Chronic lymphocyte leukemia, plasmacytomas, multiple myeloma, Burkitt's lymphoma, and EBV-transformed diseases.
A therapy for chronic hypergammaglobulinemeia evident in such diseases as monoclonalgammopathy of undetermined significance (MGUS), Waldenstrom's disease, related idiopathic monoclonalgammopathies, and plasmacytomas.

A therapy for decreasing cellular proliferation of Large B-cell Lymphomas.
A means of decreasing the involvement of B cells and Ig associated with Chronic Myelogenous Leukemia.
An immunosuppressive agent(s).
S Polynucleotides, polypeptides, antibodies, and/or agonists or antagonists of the present invention may be used to modulate IgE concentrations in vitro or in vivo.
In another embodiment, administration of polypeptides, antibodies, polynucleotides and/or agonists or antagonists of the invention, may be used to treat or prevent IgE-mediated allergic reactions including, but not limited to, asthma, rhinitis, and eczema.
The agonists and antagonists may be employed in a composition with a pharmaceutically acceptable carrier, e.g., as described herein.
The agonists or antagonists may be employed for instance to inhibit polypeptide chemotaxis and activation of macrophages and their precursors, and of neutrophils, basophils, B lymphocytes and some T-cell subsets, e.g., activated and CD8 cytotoxic T cells and natural killer cells, in certain auto-immune and chronic inflammatory and infective diseases. Examples of autoimmune diseases are described herein and include multiple sclerosis, and insulin-dependent diabetes. The antagonists or agonists may also be employed to treat infectious diseases including silicosis, sarcoidosis, idiopathic pulmonary fibrosis by, for example, preventing the recruitment and activation of mononuclear phagocytes. They may also be employed to treat idiopathic hyper-eosinophilic syndrome by, for example, preventing eosinophil production and migration. The antagonists or agonists or may also be employed for treating atherosclerosis, for example, by preventing monocyte infiltration in the artery wall.
Antibodies against polypeptides of the invention may be employed to treat ARDS.
Agonists and/or antagonists of the invention also have uses in stimulating wound and tissue repair, stimulating angiogenesis, stimulating the repair of vascular or lymphatic diseases or disorders. Additionally, agonists and antagonists of the invention may be used to stimulate the regeneration of mucosal surfaces.

In a specific embodiment, polynucleotides or polypeptides, and/or agonists thereof are used to treat or prevent a disorder characterized by primary or acquired immunodeficiency, deficient serum immunoglobulin production, recurrent infections, and/or immune system dysfunction. Moreover, polynucleotides or polypeptides, S and/or agonists thereof may be used to treat or prevent infections of the joints, bones, skin, and/or parotid glands, blood-borne infections (e.g., sepsis, meningitis, septic arthritis, and/or osteomyelitis), autoimmune diseases (e.g., those disclosed herein), inflammatory disorders, and malignancies, andlor any disease or disorder or condition associated with these infections, diseases, disorders and/or malignancies) including, but not limited to, CVID, other primary immune deficiencies, HIV disease, CLL, recurrent bronchitis, sinusitis, otitis media, conjunctivitis, pneumonia, hepatitis, meningitis, herpes zoster (e.g., severe herpes zoster), and/or pneumocystis carnii.
In another embodiment, polynucleotides, polypeptides, antibodies, andlor agonists or antagonists of the present invention are used to treat, and/or diagnose an individual having common variable immunodeficiency disease ("CVID"; also known as "acquired agammaglobulinemia" and "acquired hypogammaglobulinemia") or a subset of this disease.
In a specific embodiment, polynucleotides, polypeptides, antibodies, and/or agonists or antagonists of the present invention may be used to treat, diagnose, and/or prevent (1) cancers or neoplasms and (2) autoimmune cell or tissue-related cancers or neoplasms. In a preferred embodiment, polynucleotides, polypeptides, antibodies, and/or agonists or antagonists of the present invention conjugated to a toxin or a radioactive isotope, as described herein, may be used to treat, diagnose, and/or prevent acute myelogeneous leukemia. In a further preferred embodiment, polynucleotides, polypeptides, antibodies, and/or agonists or antagonists of the present invention conjugated to a toxin or a radioactive isotope, as described herein, may be used to treat, diagnose, and/or prevent, chronic myelogeneous leukemia, multiple myeloma, non-Hodgkins lymphoma, and/or Hodgkins disease.
In another specific embodiment, polynucleotides or polypeptides, and/or agonists or antagonists of the invention may be used to treat, diagnose, prognose, and/or prevent selective IgA deficiency, myeloperoxidase deficiency, C2 deficiency, ataxia-telangiectasia, DiGeorge anomaly, common variable immunodeficiency (CVI), X-linked agammaglobulinemia, severe combined immunodeficiency (SCID), chronic granulomatous disease (CGD), and Wiskott-Aldrich syndrome.
Examples of autoimmune disorders that can be treated or detected are described above and also include, but are not limited to: Addison's Disease, hemolytic anemia, antiphospholipid syndrome, rheumatoid arthritis, dermatitis, allergic encephalomyelitis, glomerulonephritis, Goodpasture's Syndrome, Graves' Disease, Multiple Sclerosis, Myasthenia Gravis, Neuritis, Ophthalmic, Bullous Pemphigoid, Pemphigus, Polyendocrinopathies, Purpura, Reiter's Disease, Stiff Man Syndrome, Autoimmune Thyroiditis, Systemic Lupus Erythematosus, Autoimmune Pulmonary Inflammation, Guillain-Barre Syndrome, insulin dependent diabetes mellitis, and autoimmune inflammatory eye disease.
In a preferred embodiment, the autoimmune diseases and disorders and/or conditions associated with the diseases and disorders recited above are treated, prognosed, prevented, and/or diagnosed using antibodies against the polypeptide of the invention.
As an agent to boost immunoresponsiveness among B cell immunodeficient individuals, such as, for example, an individual who has undergone a partial or complete splenectomy.
Additionally, polynucleotides, polypeptides, and/or antagonists of the invention may affect apoptosis, and therefore, would be useful in treating a number of diseases associated with increased cell survival or the inhibition of apoptosis. For example, diseases associated with increased cell survival or the inhibition of apoptosis that could be treated or detected by polynucleotides, polypeptides, and/or antagonists of the invention, include cancers (such as follicular lymphomas, carcinomas with p53 mutations, and hormone-dependent tumors, including, but not limited to colon cancer, cardiac tumors, pancreatic cancer, melanoma, retinoblastoma, glioblastoma, lung cancer, intestinal cancer, testicular cancer, stomach cancer, neuroblastoma, myxoma, myoma, lymphoma, endothelioma, osteoblastoma, osteoclastoma, osteosarcoma, chondrosarcoma, adenoma, breast cancer, prostate cancer, Kaposi's sarcoma and ovarian cancer); autoimmune disorders (such as, multiple sclerosis, Sjogren's syndrome, Hashimoto's thyroiditis, biliary cirrhosis, Behcet's disease, Crohn's disease, polymyositis, systemic lupus erythematosus and immune-related glomerulonephritis and rheumatoid arthritis) and viral infections (such as herpes viruses, pox viruses and adenoviruses), inflammation, graft v. host disease, acute graft rejection, and chronic graft rejection. In preferred embodiments, polynucleotides, polypeptides, and/or antagonists of the invention are used to inhibit growth, progression, and/or metastisis of cancers, in particular those listed above.
Additional diseases or conditions associated with increased cell survival that could be treated or detected by polynucleotides, polypeptides, and/or antagonists of the invention, include, but are not limited to, progression, and/or metastases of malignancies and related disorders such as leukemia (including acute leukemias (e.g., acute lymphocytic leukemia, acute myelocytic leukemia (including myeloblastic, promyelocytic, myelomonocytic, monocytic, and erythroleukemia)) and chronic leukemias (e.g., chronic myelocytic (granulocytic) leukemia and chronic lymphocytic leukemia)), polycythemia vera, lymphomas (e.g., Hodgkin's disease and non-Hodgkin's disease), multiple myeloma, Waldenstrom's macroglobulinemia, heavy chain disease, and solid tumors including, but not limited to, sarcomas and carcinomas such as fibrosarcoma, myxosarcoma, liposarcoma, chondrosarcoma, osteogenic sarcoma, chordoma, angiosarcoma, endotheliosarcoma, lymphangiosarcoma, lymphangioendotheliosarcoma, synovioma, mesothelioma, Ewing's tumor, leiomyosarcoma, rhabdomyosarcoma, colon carcinoma, pancreatic cancer, breast cancer, ovarian cancer, prostate cancer, squamous cell carcinoma, basal cell carcinoma, adenocarcinoma, sweat gland carcinoma, sebaceous gland carcinoma, papillary carcinoma, papillary adenocarcinomas, cystadenocarcinoma, medullary carcinoma, bronchogenic carcinoma, renal cell carcinoma, hepatoma, bile duct carcinoma, choriocarcinoma, seminoma, embryonal carcinoma, Wilm's tumor, cervical cancer, testicular tumor, lung carcinoma, small cell lung carcinoma, bladder carcinoma, epithelial carcinoma, glioma, astrocytoma, medulloblastoma, craniopharyngioma, ependymoma, pinealoma, hemangioblastoma, acoustic neuroma, oligodendroglioma, menangioma, melanoma, neuroblastoma, and retinoblastoma.
Diseases associated with increased apoptosis that could be treated or detected by polynucleotides, polypeptides, and/or antagonists of the invention, include AIDS;
neurodegenerative disorders (such as Alzheimer's disease, Parkinson's disease, Amyotrophic lateral sclerosis, Retinitis pigmentosa, Cerebellar degeneration and brain tumor or prior associated disease); autoimmune disorders (such as, multiple sclerosis, Sjogren's syndrome, Hashimoto's thyroiditis, biliary cirrhosis, Behcet's disease, Crohn's disease, polymyositis, systemic lupus erythematosus and immune-related glomerulonephritis and rheumatoid arthritis) myelodysplastic syndromes (such as aplastic anemia), graft v. host disease, ischemic injury (such as that caused by myocardial infarction, stroke and reperfusion injury), liver injury (e.g., hepatitis related liver injury, ischemia/reperfusion injury, cholestosis (bile duct injury) and liver cancer); toxin-induced liver disease (such as that caused by alcohol), septic shock, cachexia and anorexia.
Hyperproliferative diseases and/or disorders that could be detected and/or treated by polynucleotides, polypeptides, and/or antagonists of the invention, include, but are not limited to neoplasms located in the: liver, abdomen, bone, breast, digestive system, pancreas, peritoneum, endocrine glands (adrenal, parathyroid, pituitary, testicles, ovary, thymus, thyroid), eye, head and neck, nervous (central and peripheral), lymphatic system, pelvic, skin, soft tissue, spleen, thoracic, and urogenital.
Similarly, other hyperproliferative disorders can also be treated or detected by polynucleotides, polypeptides, and/or antagonists of the invention. Examples of such hyperproliferative disorders include, but are not limited to:
hypergammaglobulinemia, lymphoproliferative disorders, paraproteinemias, purpura, sarcoidosis, Sezary Syndrome, Waldenstron's Macroglobulinemia, Gaucher's Disease, histiocytosis, and any other hyperproliferative disease, besides neoplasia, located in an organ system listed above.
Hyperproliferative Disorders A polynucleotides or polypeptides, or agonists or antagonists of the invention can be used to treat, prevent, and/or diagnose hyperproliferative diseases, disorders, and/or conditions, including neoplasms. A polynucleotides or polypeptides, or agonists or antagonists of the present invention may inhibit the proliferation of the disorder through direct or indirect interactions. Alternatively, a polynucleotides or polypeptides, or agonists or antagonists of the present invention may proliferate other cells which can inhibit the hyperproliferative disorder.

For example, by increasing an immune response, particularly increasing antigenic qualities of the hyperproliferative disorder or by proliferating, differentiating, or mobilizing T-cells, hyperproliferative diseases, disorders, and/or conditions can be treated, prevented, and/or diagnosed. This immune response may be increased by either enhancing an existing immune response, or by initiating a new immune response. Alternatively, decreasing an immune response may also be a method of treating, preventing, and/or diagnosing hyperproliferative diseases, disorders, and/or conditions, such as a chemotherapeutic agent.
Examples of hyperproliferative diseases, disorders, and/or conditions that can be treated, prevented, and/or diagnosed by polynucleotides or polypeptides, or agonists or antagonists of the present invention include, but are not limited to neoplasms located in the: colon, abdomen, bone, breast, digestive system, liver, pancreas, peritoneum, endocrine glands (adrenal, parathyroid, pituitary, testicles, ovary, thymus, thyroid), eye, head and neck, nervous (central and peripheral), lymphatic system, pelvic, skin, soft tissue, spleen, thoracic, and urogenital.
Similarly, other hyperproliferative diseases, disorders, and/or conditions can also be treated, prevented, and/or diagnosed by a polynucleotides or polypeptides, or agonists or antagonists of the present invention. Examples of such hyperproliferative diseases, disorders, and/or conditions include, but are not limited to:
hypergammaglobulinemia, lymphoproliferative diseases, disorders, and/or conditions, paraproteinemias, purpura, sarcoidosis, Sezary Syndrome, Waldenstron's Macroglobulinemia, Gaucher's Disease, histiocytosis, and any other hypeiproliferative disease, besides neoplasia, located in an organ system listed above.
One preferred embodiment utilizes polynucleotides of the present invention to inhibit aberrant cellular division, by gene therapy using the present invention, and/or protein fusions or fragments thereof.
Thus, the present invention provides a method for treating or preventing cell proliferative diseases, disorders, and/or conditions by inserting into an abnormally proliferating cell a polynucleotide of the present invention, wherein said polynucleotide represses said expression.
Another embodiment of the present invention provides a method of treating or preventing cell-proliferative diseases, disorders, and/or conditions in individuals comprising administration of one or more active gene copies of the present invention to an abnormally proliferating cell or cells. In a preferred embodiment, polynucleotides of the present invention is a DNA construct comprising a recombinant expression vector effective in expressing a DNA sequence encoding said polynucleotides. In another preferred embodiment of the present invention, the DNA
construct encoding the poynucleotides of the present invention is inserted into cells to be treated utilizing a retrovirus, or more preferrably an adenoviral vector (See G J.
Nabel, et. al., PNAS 1999 96: 324-326, which is hereby incorporated by reference).
In a most preferred embodiment, the viral vector is defective and will not transform non-proliferating cells, only proliferating cells. Moreover, in a preferred embodiment, the polynucleotides of the present invention inserted into proliferating cells either alone, or in combination with or fused to other polynucleotides, can then be modulated via an external stimulus (i.e. magnetic, specific small molecule, chemical, or drug administration, etc.), which acts upon the promoter upstream of said polynucleotides to induce expression of the encoded protein product. As such the beneficial therapeutic affect of the present invention may be expressly modulated (i.e.
to increase, decrease, or inhibit expression of the present invention) based upon said external stimulus.
Polynucleotides of the present invention may be useful in repressing expression of oncogenic genes or antigens. By "repressing expression of the oncogenic genes " is intended the suppression of the transcription of the gene, the degradation of the gene transcript (pre-message RNA), the inhibition of splicing, the destruction of the messenger RNA, the prevention of the post-translational modifications of the protein, the destruction of the protein, or the inhibition of the normal function of the protein.
For local administration to abnormally proliferating cells, polynucleotides of the present invention may be administered by any method known to those of skill in the art including, but not limited to transfection, electroporation, microinjection of cells, or in vehicles such as liposomes, lipofectin, or as naked polynucleotides, or any other method described throughout the specification. The polynucleotide of the present invention may be delivered by known gene delivery systems such as, but not limited to, retroviral vectors (Gilboa, J. Virology 44:845 (1982); Hocke, Nature 320:275 (1986); Wilson, et al., Proc. Natl. Acad. Sci. U.S.A. 85:3014), vaccinia virus system (Chakrabarty et al., Mol. Cell Biol. 5:3403 (1985) or other efficient DNA
delivery systems (Yates et al., Nature 313:812 (1985)) known to those skilled in the art. These references are exemplary only and are hereby incorporated by reference.
In order to specifically deliver or transfect cells which are abnormally proliferating and spare non-dividing cells, it is preferable to utilize a retrovirus, or adenoviral (as described in the art and elsewhere herein) delivery system known to those of skill in the art. Since host DNA replication is required for retroviral DNA to integrate and the retrovirus will be unable to self replicate due to the lack of the retrovirus genes needed for its life cycle. Utilizing such a retroviral delivery system for polynucleotides of the present invention will target said gene and constructs to abnormally proliferating cells and will spare the non-dividing normal cells.
The polynucleotides of the present invention may be delivered directly to cell proliferative disorder/disease sites in internal organs, body cavities and the like by use of imaging devices used to guide an injecting needle directly to the disease site. The polynucleotides of the present invention may also be administered to disease sites at the time of surgical intervention.
By "cell proliferative disease" is meant any human or animal disease or disorder, affecting any one or any combination of organs, cavities, or body parts, which is characterized by single or multiple local abnormal proliferations of cells, groups of cells, or tissues, whether benign or malignant.
Any amount of the polynucleotides of the present invention may be administered as long as it has a biologically inhibiting effect on the proliferation of the treated cells. Moreover, it is possible to administer more than one of the polynucleotide of the present invention simultaneously to the same site. By "biologically inhibiting" is meant partial or total growth inhibition as well as decreases in the rate of proliferation or growth of the cells. The biologically inhibitory dose may be determined by assessing the effects of the polynucleotides of the present invention on target malignant or abnormally proliferating cell growth in tissue culture, tumor growth in animals and cell cultures, or any other method known to one of ordinary skill in the art.

The present invention is further directed to antibody-based therapies which involve administering of anti-polypeptides and anti-polynucleotide antibodies to a mammalian, preferably human, patient for treating, preventing, and/or diagnosing one or more of the described diseases, disorders, and/or conditions. Methods for S producing anti-polypeptides and anti-polynucleotide antibodies polyclonal and monoclonal antibodies are described in detail elsewhere herein. Such antibodies may be provided in pharmaceutically acceptable compositions as known in the art or as described herein.
A summary of the ways in which the antibodies of the present invention may be used therapeutically includes binding polynucleotides or polypeptides of the present invention locally or systemically in the body or by direct cytotoxicity of the antibody, e.g. as mediated by complement (CDC) or by effector cells (ADCC).
Some of these approaches are described in more detail below. Armed with the teachings provided herein, one of ordinary skill in the art will know how to use the antibodies of the present invention for diagnostic, monitoring or therapeutic purposes without undue experimentation.
In particular, the antibodies, fragments and derivatives of the present invention are useful for treating, preventing, and/or diagnosing a subject having or developing cell proliferative and/or differentiation diseases, disorders, and/or conditions as described herein. Such treatment comprises administering a single or multiple doses of the antibody, or a fragment, derivative, or a conjugate thereof.
The antibodies of this invention may be advantageously utilized in combination with other monoclonal or chimeric antibodies, or with lymphokines or hematopoietic growth factors, for example, which serve to increase the number or activity of effector cells which interact with the antibodies.
It is preferred to use high affinity and/or potent in vivo inhibiting and/or neutralizing antibodies against polypeptides or polynucleotides of the present invention, fragments or regions thereof, for both immunoassays directed to and therapy of diseases, disorders, and/or conditions related to polynucleotides or polypeptides, including fragements thereof, of the present invention. Such antibodies, fragments, or regions, will preferably have an affinity for polynucleotides or polypeptides, including fragements thereof. Preferred binding affinities include those with a dissociation constant or Kd less than SX10-6M, 10-6M, SX10-~M, 10-~M, 8M, 10-$M, 5 X 10-~M, 10-9M, 5 X 10-' °M, 10-' °M, 5 X 10-"M, 10-"M, 5 X 10-' ZM, 10-'ZM, SX10-'3M, 10-'3M, SX10-'4M, 10-'4M, SX10-'SM, and 10-'SM.
Moreover, polypeptides of the present invention are useful in inhibiting the angiogenesis of proliferative cells or tissues, either alone, as a protein fusion, or in combination with other polypeptides directly or indirectly, as described elsewhere herein. In a most preferred embodiment, said anti-angiogenesis effect may be achieved indirectly, for example, through the inhibition of hematopoietic, tumor-specific cells, such as tumor-associated macrophages (See Joseph IB, et al. J
Natl Cancer Inst, 90(21):1648-53 (1998), which is hereby incorporated by reference).
Antibodies directed to polypeptides or polynucleotides of the present invention may also result in inhibition of angiogenesis directly, or indirectly (See Witte L, et al., Cancer Metastasis Rev. 17(2):155-61 (1998), which is hereby incorporated by reference)).
Polypeptides, including protein fusions, of the present invention, or fragments thereof may be useful in inhibiting proliferative cells or tissues through the induction of apoptosis. Said polypeptides may act either directly, or indirectly to induce apoptosis of proliferative cells and tissues, for example in the activation of a death-domain receptor, such as tumor necrosis factor (TNF) receptor-1, CD95 (Fas/APO-1), TNF-receptor-related apoptosis-mediated protein (TRAMP) and TNF-related apoptosis-inducing ligand (TRAIL) receptor-1 and -2 (See Schulze-Osthoff K, et.al., Eur J Biochem 254(3):439-59 (1998), which is hereby incorporated by reference).
Moreover, in another preferred embodiment of the present invention, said polypeptides may induce apoptosis through other mechanisms, such as in the activation of other proteins which will activate apoptosis, or through stimulating the expression of said proteins, either alone or in combination with small molecule drugs or adjuviants, such as apoptonin, galectins, thioredoxins, antiinflammatory proteins (See for example, Mutat Res 400(1-2):447-SS (1998), Med Hypotheses.50(5):423-(1998), Chem Biol Interact. Apr 24;111-112:23-34 (1998), J Mol Med.76(6):402-(1998), Int J Tissue React;20(1):3-15 (1998), which are all hereby incorporated by reference).

Polypeptides, including protein fusions to, or fragments thereof, of the present invention are useful in inhibiting the metastasis of proliferative cells or tissues.
Inhibition may occur as a direct result of administering polypeptides, or antibodies directed to said polypeptides as described elsewere herein, or indirectly, such as activating the expression of proteins known to inhibit metastasis, for example alpha 4 integrins, (See, e.g., Curr Top Microbiol Immunol 1998;231:125-41, which is hereby incorporated by reference). Such thereapeutic affects of the present invention may be achieved either alone, or in combination with small molecule drugs or adjuvants.
In another embodiment, the invention provides a method of delivering compositions containing the polypeptides of the invention (e.g., compositions containing polypeptides or polypeptide antibodes associated with heterologous polypeptides, heterologous nucleic acids, toxins, or prodrugs) to targeted cells expressing the polypeptide of the present invention. Polypeptides or polypeptide antibodes of the invention may be associated with with heterologous polypeptides, heterologous nucleic acids, toxins, or prodrugs via hydrophobic, hydrophilic, ionic and/or covalent interactions.
Polypeptides, protein fusions to, or fragments thereof, of the present invention are useful in enhancing the immunogenicity and/or antigenicity of proliferating cells or tissues, either directly, such as would occur if the polypeptides of the present invention 'vaccinated' the immune response to respond to proliferative antigens and immunogens, or indirectly, such as in activating the expression of proteins known to enhance the immune response (e.g. chemokines), to said antigens and immunogens.
Cardiovascular Disorders Polynucleotides or polypeptides, or agonists or antagonists of the invention may be used to treat, prevent, and/or diagnose cardiovascular diseases, disorders, and/or conditions, including peripheral artery disease, such as limb ischemia.
Cardiovascular diseases, disorders, and/or conditions include cardiovascular abnormalities, such as arterio-arterial fistula, arteriovenous fistula, cerebral arteriovenous malformations, congenital heart defects, pulmonary atresia, and Scimitar Syndrome. Congenital heart defects include aortic coarctation, cor triatriatum, coronary vessel anomalies, crisscross heart, dextrocardia, patent ductus arteriosus, Ebstein's anomaly, Eisenmenger complex, hypoplastic left heart syndrome, levocardia, tetralogy of fallot, transposition of great vessels, double outlet right ventricle, tricuspid atresia, persistent truncus arteriosus, and heart septal defects, such as aortopulmonary septal defect, endocardial cushion defects, Lutembacher's Syndrome, trilogy of Fallot, ventricular heart septal defects.
Cardiovascular diseases, disorders, and/or conditions also include heart disease, such as arrhythmias, carcinoid heart disease, high cardiac output, low cardiac output, cardiac tamponade, endocarditis (including bacterial), heart aneurysm, cardiac arrest, congestive heart failure, congestive cardiomyopathy, paroxysmal dyspnea, cardiac edema, heart hypertrophy, congestive cardiomyopathy, left ventricular hypertrophy, right ventricular hypertrophy, post-infarction heart rupture, ventricular septal rupture, heart valve diseases, myocardial diseases, myocardial ischemia, pericardial effusion, pericarditis (including constrictive and tuberculous), pneumopericardium, postpericardiotomy syndrome, pulmonary heart disease, rheumatic heart disease, ventricular dysfunction, hyperemia, cardiovascular pregnancy complications, Scimitar Syndrome, cardiovascular syphilis, and cardiovascular tuberculosis.
Arrhythmias include sinus arrhythmia, atrial fibrillation, atrial flutter, bradycardia, extrasystole, Adams-Stokes Syndrome, bundle-branch block, sinoatrial block, long QT syndrome, parasystole, Lown-Ganong-Levine Syndrome, Mahaim-type pre-excitation syndrome, Wolff Parkinson-White syndrome, sick sinus syndrome, tachycardias, and ventricular fibrillation. Tachycardias include paroxysmal tachycardia, supraventricular tachycardia, accelerated idioventricular rhythm, atrioventricular nodal reentry tachycardia, ectopic atrial tachycardia, ectopic functional tachycardia, sinoatrial nodal reentry tachycardia, sinus tachycardia, Torsades de Pointes, and ventricular tachycardia.
Heart valve disease include aortic valve insufficiency, aortic valve stenosis, hear murmurs, aortic valve prolapse, mitral valve prolapse, tricuspid valve prolapse, mural valve insufficiency, mitral valve stenosis, pulmonary atresia, pulmonary valve insufficiency, pulmonary valve stenosis, tricuspid atresia, tricuspid valve insufficiency, and tricuspid valve stenosis.

Myocardial diseases include alcoholic cardiomyopathy, congestive cardiomyopathy, hypertrophic cardiomyopathy, aortic subvalvular stenosis, pulmonary subvalvular stenosis, restrictive cardiomyopathy, Chagas cardiomyopathy, endocardial fibroelastosis, endomyocardial fibrosis, Kearns Syndrome, myocardial S reperfusion injury, and myocarditis.
Myocardial ischemias include coronary disease, such as angina pectoris, coronary aneurysm, coronary arteriosclerosis, coronary thrombosis, coronary vasospasm, myocardial infarction and myocardial stunning.
Cardiovascular diseases also include vascular diseases such as aneurysms, angiodysplasia, angiomatosis, bacillary angiomatosis, Hippel-Lindau Disease, Klippel-Trenaunay-Weber Syndrome, Sturge-Weber Syndrome, angioneurotic edema, aortic diseases, Takayasu's Arteritis, aortitis, Leriche's Syndrome, .arterial occlusive diseases, arteritis, enarteritis, polyarteritis nodosa, cerebrovascular diseases, disorders, and/or conditions, diabetic angiopathies, diabetic retinopathy, embolisms, thrombosis, erythromelalgia, hemorrhoids, hepatic veno-occlusive disease, hypertension, hypotension, ischemia, peripheral vascular diseases, phlebitis, pulmonary veno occlusive disease, Raynaud's disease, CREST syndrome, retinal vein occlusion, Scimitar syndrome, superior vena cava syndrome, telangiectasia, atacia telangiectasia, hereditary hemorrhagic telangiectasia, varicocele, varicose veins, varicose ulcer, vasculitis, and venous insufficiency.
Aneurysms include dissecting aneurysms, false aneurysms, infected aneurysms, ruptured aneurysms, aortic aneurysms, cerebral aneurysms, coronary aneurysms, heart aneurysms, and iliac aneurysms.
Arterial occlusive diseases include arteriosclerosis, intermittent claudication, carotid stenosis, fibromuscular dysplasias, mesenteric vascular occlusion, Moyamoya disease, renal artery obstruction, retinal artery occlusion, and thromboangiitis obliterans.
Cerebrovascular diseases, disorders, and/or conditions include carotid artery diseases, cerebral amyloid angiopathy, cerebral aneurysm, cerebral anoxia, cerebral arteriosclerosis, cerebral arteriovenous malformation, cerebral artery diseases, cerebral embolism and thrombosis, carotid artery thrombosis, sinus thrombosis, Wallenberg's syndrome, cerebral hemorrhage, epidural hematoma, subdural hematoma, subaraxhnoid hemorrhage, cerebral infarction, cerebral ischemia (including transient), subclavian steal syndrome, periventricular leukomalacia, vascular headache, cluster headache, migraine, and vertebrobasilar insufficiency.
Embolisms include air embolisms, amniotic fluid embolisms, cholesterol embolisms, blue toe syndrome, fat embolisms, pulmonary embolisms, and thromoboembolisms. Thrombosis include coronary thrombosis, hepatic vein thrombosis, retinal vein occlusion, carotid artery thrombosis, sinus thrombosis, Wallenberg's syndrome, and thrombophlebitis.
Ischemia includes cerebral ischemia, ischemic colitis, compartment syndromes, anterior compartment syndrome, myocardial ischemia, reperfusion injuries, and peripheral limb ischemia. Vasculitis includes aortitis, arteritis, Behcet's Syndrome, Churg-Strauss Syndrome, mucocutaneous lymph node syndrome, thromboangiitis obliterans, hypersensitivity vasculitis, Schoenlein-Henoch purpura, allergic cutaneous vasculitis, and Wegener's granulomatosis.
Polynucleotides or polypeptides, or agonists or antagonists of the invention, are especially effective for the treatment of critical limb ischemia and coronary disease.
Polypeptides may be administered using any method known in the art, including, but not limited to, direct needle injection at the delivery site, intravenous injection, topical administration, catheter infusion, biolistic injectors, particle accelerators, gelfoam sponge depots, other commercially available depot materials, osmotic pumps, oral or suppositorial solid pharmaceutical formulations, decanting or topical applications during surgery, aerosol delivery. Such methods are known in the art. Polypeptides of the invention may be administered as part of a Therapeutic, described in more detail below. Methods of delivering polynucleotides of the invention are described in more detail herein.
Anti-An~io~enesis Activity The naturally occurring balance between endogenous stimulators and inhibitors of angiogenesis is one in which inhibitory influences predominate.
Rastinejad et al., Cell 56:345-355 (1989). In those rare instances in which neovascularization occurs under normal physiological conditions, such as wound healing, organ regeneration, embryonic development, and female reproductive processes, angiogenesis is stringently regulated and spatially and temporally delimited. Under conditions of pathological angiogenesis such as that characterizing solid tumor growth, these regulatory controls fail. Unregulated angiogenesis becomes pathologic and sustains progression of many neoplastic and non-neoplastic diseases.
A number of serious diseases are dominated by abnormal neovascularization including solid tumor growth and metastases, arthritis, some types of eye diseases, disorders, and/or conditions, and psoriasis. See, e.g., reviews by Moses et al., Biotech. 9:630-634 (1991); Folkman et al., N. Engl. J. Med., 333:1757-1763 (1995);
Auerbach et al., J. Microvasc. Res. 29:401-411 (1985); Folkman, Advances in Cancer Research, eds. Klein and Weinhouse, Academic Press, New York, pp. 175-203 (1985); Patz, Am. J. Opthalmol. 94:715-743 (1982); and Folkman et al., Science 221:719-725 (1983). In a number of pathological conditions, the process of angiogenesis contributes to the disease state. For example, significant data have accumulated which suggest that the growth of solid tumors is dependent on angiogenesis. Folkman and Klagsbrun, Science 235:442-447 (1987).
The present invention provides for treatment of diseases, disorders, and/or conditions associated with neovascularization by administration of the polynucleotides and/or polypeptides of the invention, as well as agonists or antagonists of the present invention. Malignant and metastatic conditions which can be treated with the polynucleotides and polypeptides, or agonists or antagonists of the invention include, but are not limited to, malignancies, solid tumors, and cancers described herein and otherwise known in the art (for a review of such disorders, see Fishman et al., Medicine, 2d Ed., J. B. Lippincott Co., Philadelphia (1985)).Thus, the present invention provides a method of treating, preventing, and/or diagnosing an angiogenesis-related disease and/or disorder, comprising administering to an individual in need thereof a therapeutically effective amount of a polynucleotide, polypeptide, antagonist and/or agonist of the invention. For example, polynucleotides, polypeptides, antagonists and/or agonists may be utilized in a variety of additional methods in order to therapeutically treator prevent a cancer or tumor.
Cancers which may be treated, prevented, and/or diagnosed with polynucleotides, polypeptides, antagonists and/or agonists include, but are not limited to solid tumors, including prostate, lung, breast, ovarian, stomach, pancreas, larynx, esophagus, testes, liver, parotid, biliary tract, colon, rectum, cervix, uterus, endometrium, kidney, bladder, thyroid cancer; primary tumors and metastases; melanomas;
glioblastoma;
Kaposi's sarcoma; leiomyosarcoma; non- small cell lung cancer; colorectal cancer;
advanced malignancies; and blood born tumors such as leukemias. For example, polynucleotides, polypeptides, antagonists and/or agonists may be delivered topically, in order to treat or prevent cancers such as skin cancer, head and neck tumors, breast tumors, and Kaposi's sarcoma.
Within yet other aspects, polynucleotides, polypeptides, antagonists and/or agonists may be utilized to treat superficial forms of bladder cancer by, for example, intravesical administration. Polynucleotides, polypeptides, antagonists and/or agonists may be delivered directly into the tumor, or near the tumor site, via injection or a catheter. Of course, as the artisan of ordinary skill will appreciate, the appropriate mode of administration will vary according to the cancer to be treated. Other modes of delivery are discussed herein.
Polynucleotides, polypeptides, antagonists and/or agonists may be useful in treating, preventing, and/or diagnosing other diseases, disorders, and/or conditions, besides cancers, which involve angiogenesis. These diseases, disorders, and/or conditions include, but are not limited to: benign tumors, for example hemangiomas, acoustic neuromas, neurofibromas, trachomas, and pyogenic granulomas;
artheroscleric plaques; ocular angiogenic diseases, for example, diabetic retinopathy, retinopathy of prematurity, macular degeneration, corneal graft rejection, neovascular glaucoma, retrolental fibroplasia, rubeosis, retinoblastoma, uvietis and Pterygia (abnormal blood vessel growth) of the eye; rheumatoid arthritis; psoriasis;
delayed wound healing; endometriosis; vasculogenesis; granulations; hypertrophic scars (keloids); nonunion fractures; scleroderma; trachoma; vascular adhesions;
myocardial angiogenesis; coronary collaterals; cerebral collaterals; arteriovenous malformations;
ischemic limb angiogenesis; Osler-Webber Syndrome; plaque neovascularization;
telangiectasia; hemophiliac joints; angiofibroma; fibromuscular dysplasia;
wound granulation; Crohn's disease; and atherosclerosis.
For example, within one aspect of the present invention methods are provided for treating, preventing, and/or diagnosing hypertrophic scars and keloids, comprising the step of administering a polynucleotide, polypeptide, antagonist and/or agonist of the invention to a hypertrophic scar or keloid.
Within one embodiment of the present invention polynucleotides, polypeptides, antagonists and/or agonists are directly injected into a hypertrophic scar or keloid, in order to prevent the progression of these lesions. This therapy is of particular value in the prophylactic treatment of conditions which are known to result in the development of hypertrophic scars and keloids (e.g., burns), and is preferably initiated after the proliferative phase has had time to progress (approximately 14 days after the initial injury), but before hypertrophic scar or keloid development.
As noted above, the present invention also provides methods for treating, preventing, and/or diagnosing neovascular diseases of the eye, including for example, corneal neovascularization, neovascular glaucoma, proliferative diabetic retinopathy, retrolental fibroplasia and macular degeneration.
Moreover, Ocular diseases, disorders, and/or conditions associated with neovascularization which can be treated, prevented, and/or diagnosed with the polynucleotides and polypeptides of the present invention (including agonists and/or antagonists) include, but are not limited to: neovascular glaucoma, diabetic retinopathy, retinoblastoma, retrolental fibroplasia, uveitis, retinopathy of prematurity macular degeneration, corneal graft neovascularization, as well as other eye inflammatory diseases, ocular tumors and diseases associated with choroidal or iris neovascularization. See, e.g., reviews by Waltman et al., Am. J. Ophthal.
85:704-710 (1978) and Gartner et al., Surv. Ophthal. 22:291-312 (1978).
Thus, within one aspect of the present invention methods are provided for treating or preventing neovascular diseases of the eye such as corneal neovascularization (including corneal graft neovascularization), comprising the step of administering to a patient a therapeutically effective amount of a compound (as described above) to the cornea, such that the formation of blood vessels is inhibited.
Briefly, the cornea is a tissue which normally lacks blood vessels. In certain pathological conditions however, capillaries may extend into the cornea from the pericorneal vascular plexus of the limbus. When the cornea becomes vascularized, it also becomes clouded, resulting in a decline in the patient's visual acuity.
.Visual loss may become complete if the cornea completely opacitates. A wide variety of diseases, disorders, and/or conditions can result in corneal neovascularization, including for example, corneal infections (e.g., trachoma, herpes simplex keratrtis, leishmaniasis and onchocerciasis), immunological processes (e.g., graft rejection and Stevens-Johnson's syndrome), alkali burns, trauma, inflammation (of any cause), toxic and nutritional deficiency states, and as a complication of wearing contact lenses.
Within particularly preferred embodiments of the invention, may be prepared for topical administration in saline (combined with any of the preservatives and antimicrobial agents commonly used in ocular preparations), and administered in eyedrop form. The solution or suspension may be prepared in its pure form and administered several times daily. Alternatively, anti-angiogenic compositions, prepared as described above, may also be administered directly to the cornea.
Within preferred embodiments, the anti-angiogenic composition is prepared with a muco-adhesive polymer which binds to cornea. Within further embodiments, the anti-angiogenic factors or anti-angiogenic compositions may be utilized as an adjunct to conventional steroid therapy. Topical therapy may also be useful prophylactically in corneal lesions which are known to have a high probability of inducing an angiogenic response (such as chemical burns). In these instances the treatment, likely in combination with steroids, may be instituted immediately to help prevent subsequent complications.
Within other embodiments, the compounds described above may be injected directly into the corneal stroma by an ophthalmologist under microscopic guidance.
The preferred site of injection may vary with the morphology of the individual lesion, but the goal of the administration would be to place the composition at the advancing front of the vasculature (i.e., interspersed between the blood vessels and the normal cornea). In most cases this would involve perilimbic corneal injection to "protect" the cornea from the advancing blood vessels. This method may also be utilized shortly after a corneal insult in order to prophylactically prevent corneal neovascularization.
In this situation the material could be injected in the perilimbic cornea interspersed between the corneal lesion and its undesired potential limbic blood supply.
Such methods may also be utilized in a similar fashion to prevent capillary invasion of transplanted corneas. In a sustained-release form injections might only be required 2-3 times per year. A steroid could also be added to the injection solution to reduce inflammation resulting from the injection itself.
Within another aspect of the present invention, methods are provided for treating or preventing neovascular glaucoma, comprising the step of administering to a patient a therapeutically effective amount of a polynucleotide, polypeptide, antagonist and/or agonist to the eye, such that the formation of blood vessels is inhibited. In one embodiment, the compound may be administered topically to the eye in order to treat or prevent early forms of neovascular glaucoma. Within other embodiments, the compound may be implanted by injection into the region of the anterior chamber angle. Within other embodiments, the compound may also be placed in any location such that the compound is continuously released into the aqueous humor. Within another aspect of the present invention, methods are provided for treating or preventing proliferative diabetic retinopathy, comprising the step of administering to a patient a therapeutically effective amount of a polynucleotide, polypeptide, antagonist and/or agonist to the eyes, such that the formation of blood vessels is inhibited.
Within particularly preferred embodiments of the invention, proliferative diabetic retinopathy may be treated by injection into the aqueous humor or the vitreous, in order to increase the local concentration of the polynucleotide, polypeptide, antagonist and/or agonist in the retina. Preferably, this treatment should be initiated prior to the acquisition of severe disease requiring photocoagulation.
Within another aspect of the present invention, methods are provided for treating or preventing retrolental fibroplasia, comprising the step of administering to a patient a therapeutically effective amount of a polynucleotide, polypeptide, antagonist and/or agonist to the eye, such that the formation of blood vessels is inhibited. The compound may be administered topically, via intravitreous injection and/or via intraocular implants.
Additionally, diseases, disorders, and/or conditions which can be treated, prevented, and/or diagnosed with the polynucleotides, polypeptides, agonists and/or agonists include, but are not limited to, hemangioma, arthritis, psoriasis, angiofibroma, atherosclerotic plaques, delayed wound healing, granulations, hemophilic joints, hypertrophic scars, nonunion fractures, Osler-Weber syndrome, pyogenic granuloma, scleroderma, trachoma, and vascular adhesions.
Moreover, diseases, disorders, and/or conditions and/or states, which can be treated, prevented, and/or diagnosed with the the polynucleotides, polypeptides, agonists and/or agonists include, but are not limited to, solid tumors, blood born tumors such as leukemias, tumor metastasis, Kaposi's sarcoma, benign tumors, for example hemangiomas, acoustic neuromas, neurofibromas, trachomas, and pyogenic granulomas, rheumatoid arthritis, psoriasis, ocular angiogenic diseases, for example, diabetic retinopathy, retinopathy of prematurity, macular degeneration, corneal graft rejection, neovascular glaucoma, retrolental fibroplasia, rubeosis, retinoblastoma, and uvietis, delayed wound healing, endometriosis, vascluogenesis, granulations, hypertrophic scars (keloids), nonunion fractures, scleroderma, trachoma, vascular adhesions, myocardial angiogenesis, coronary collaterals, cerebral collaterals, arteriovenous malformations, ischemic limb angiogenesis, Osler-Webber Syndrome, plaque neovascularization, telangiectasia, hemophiliac joints, angiofibroma fibromuscular dysplasia, wound granulation, Crohn's disease, atherosclerosis, birth control agent by preventing vascularization required for embryo implantation controlling menstruation, diseases that have angiogenesis as a pathologic consequence such as cat scratch disease (Rochele minalia quintosa), ulcers (Helicobacter pylori), Bartonellosis and bacillary angiomatosis.
In one aspect of the birth control method, an amount of the compound sufficient to block embryo implantation is administered before or after intercourse and fertilization have occurred, thus providing an effective method of birth control, possibly a "morning after" method. Polynucleotides, polypeptides, agonists and/or agonists may also be used in controlling menstruation or administered as either a peritoneal lavage fluid or for peritoneal implantation in the treatment of endometriosis.
Polynucleotides, polypeptides, agonists and/or agonists of the present invention may be incorporated into surgical sutures in order to prevent stitch granulomas.
Polynucleotides, polypeptides, agonists and/or agonists may be utilized in a wide variety of surgical procedures. For example, within one aspect of the present invention a compositions (in the form of, for example, a spray or film) may be utilized to coat or spray an area prior to removal of a tumor, in order to isolate normal surrounding tissues from malignant tissue, and/or to prevent the spread of disease to surrounding tissues. Within other aspects of the present invention, compositions (e.g., in the form of a spray) may be delivered via endoscopic procedures in order to coat tumors, or inhibit angiogenesis in a desired locale. Within yet other aspects of the present invention, surgical meshes which have been coated with anti-angiogenic compositions of the present invention may be utilized in any procedure wherein a surgical mesh might be utilized. For example, within one embodiment of the invention a surgical mesh laden with an anti-angiogenic composition may be utilized during abdominal cancer resection surgery (e.g., subsequent to colon resection) in order to provide support to the structure, and to release an amount of the anti-angiogenic factor.
Within further aspects of the present invention, methods are provided for treating tumor excision sites, comprising administering a polynucleotide, polypeptide, agonist and/or agonist to the resection margins of a tumor subsequent to excision, such that the local recurrence of cancer and the formation of new blood vessels at the site is inhibited. Within one embodiment of the invention, the anti-angiogenic compound is administered directly to the tumor excision site (e.g., applied by swabbing, brushing or otherwise coating the resection margins of the tumor with the anti-angiogenic compound). Alternatively, the anti-angiogenic compounds may be incorporated into known surgical pastes prior to administration. Within particularly preferred embodiments of the invention, the anti-angiogenic compounds are applied after hepatic resections for malignancy, and after neurosurgical operations.
Within one aspect of the present invention, polynucleotides, polypeptides, agonists and/or agonists may be administered to the resection margin of a wide variety of tumors, including for example, breast, colon, brain and hepatic tumors. For example, within one embodiment of the invention, anti-angiogenic compounds may be administered to the site of a neurological tumor subsequent to excision, such that the formation of new blood vessels at the site are inhibited.
The polynucleotides, polypeptides, agonists and/or agonists of the present invention may also be administered along with other anti-angiogenic factors.
Representative examples of other anti-angiogenic factors include: Anti-Invasive Factor, retinoic acid and derivatives thereof, paclitaxel, Suramin, Tissue Inhibitor of Metalloproteinase-1, Tissue Inhibitor of Metalloproteinase-2, Plasminogen Activator Inhibitor-1, Plasminogen Activator Inhibitor-2, and various forms of the lighter "d group" transition metals.
Lighter "d group" transition metals include, for example, vanadium, molybdenum, tungsten, titanium, niobium, and tantalum species. Such transition metal species may form transition metal complexes. Suitable complexes of the above-mentioned transition metal species include oxo transition metal complexes.
Representative examples of vanadium complexes include oxo vanadium complexes such as vanadate and vanadyl complexes. Suitable vanadate complexes include metavanadate and orthovanadate complexes such as, for example, ammonium metavanadate, sodium metavanadate, and sodium orthovanadate. Suitable vanadyl complexes include, for example, vanadyl acetylacetonate and vanadyl sulfate including vanadyl sulfate hydrates such as vanadyl sulfate mono- and trihydrates.
Representative examples of tungsten and molybdenum complexes also include oxo complexes. Suitable oxo tungsten complexes include tungstate and tungsten oxide complexes. Suitable tungstate complexes include ammonium tungstate, calcium tungstate, sodium tungstate dihydrate, and tungstic acid. Suitable tungsten oxides include tungsten (IV) oxide and tungsten (VI) oxide. Suitable oxo molybdenum complexes include molybdate, molybdenum oxide, and molybdenyl complexes. Suitable molybdate complexes include ammonium molybdate and its hydrates, sodium molybdate and its hydrates, and potassium molybdate and its hydrates. Suitable molybdenum oxides include molybdenum (VI) oxide, molybdenum (VI) oxide, and molybdic acid. Suitable molybdenyl complexes include, for example, molybdenyl acetylacetonate. Other suitable tungsten and molybdenum complexes include hydroxo derivatives derived from, for example, glycerol, tartaric acid, and sugars.
A wide variety of other anti-angiogenic factors may also be utilized within the context of the present invention. Representative examples include platelet factor 4;
protamine sulphate; sulphated chitin derivatives (prepared from queen crab shells), (Murata et al., Cancer Res. 51:22-26, 1991 ); Sulphated Polysaccharide Peptidoglycan Complex (SP- PG) (the function of this compound may be enhanced by the presence of steroids such as estrogen, and tamoxifen citrate); Staurosporine;
modulators of matrix metabolism, including for example, proline analogs, cishydroxyproline, d,L-3,4-dehydroproline, Thiaproline, alpha,alpha-dipyridyl, aminopropionitrile fumarate;
4-propyl-5-(4-pyridinyl)-2(3H)-oxazolone; Methotrexate; Mitoxantrone; Heparin;
Interferons; 2 Macroglobulin-serum; ChIMP-3 (Pavloff et al., J. Bio. Chem.
267:17321-17326, 1992); Chymostatin (Tomkinson et al., Biochem J. 286:475-480, 1992); Cyclodextrin Tetradecasulfate; Eponemycin; Camptothecin; Fumagillin (Ingber et al., Nature 348:555-557, 1990); Gold Sodium Thiomalate ("GST";
Matsubara and Ziff, J. Clin. Invest. 79:1440-1446, 1987); anticollagenase-serum;
alpha2-antiplasmin (Holmes et al., J. Biol. Chem. 262(4):1659-1664, 1987);
Bisantrene (National Cancer Institute); Lobenzarit disodium (N-(2)-carboxyphenyl-4-chloroanthronilic acid disodium or "CCA"; Takeuchi et al., Agents Actions 36:312-316, 1992); Thalidomide; Angostatic steroid; AGM-1470;
carboxynaminolinidazole;
and metalloproteinase inhibitors such as BB94.
Diseases at the Cellular Level Diseases associated with increased cell survival or the inhibition of apoptosis that could be treated, prevented, and/or diagnosed by the polynucleotides or polypeptides and/or antagonists or agonists of the invention, include cancers (such as follicular lymphomas, carcinomas with p53 mutations, and hormone-dependent tumors, including, but not limited to colon cancer, cardiac tumors, pancreatic cancer, melanoma, retinoblastoma, glioblastoma, lung cancer, intestinal cancer, testicular cancer, stomach cancer, neuroblastoma, myxoma, myoma, lymphoma, endothelioma, osteoblastoma, osteoclastoma, osteosarcoma, chondrosarcoma, adenoma, breast cancer, prostate cancer, Kaposi's sarcoma and ovarian cancer); autoimmune diseases, disorders, and/or conditions (such as, multiple sclerosis, Sjogren's syndrome, Hashimoto's thyroiditis, biliary cirrhosis, Behcet's disease, Crohn's disease, polymyositis, systemic lupus erythematosus and immune-related glomerulonephritis and rheumatoid arthritis) and viral infections (such as herpes viruses, pox viruses and adenoviruses), inflammation, graft v. host disease, acute graft rejection, and chronic graft rejection. In preferred embodiments, the polynucleotides or polypeptides, and/or agonists or antagonists of the invention are used to inhibit growth, progression, and/or metasis of cancers, in particular those listed above.
Additional diseases or conditions associated with increased cell survival that could be treated, prevented or diagnosed by the polynucleotides or polypeptides, or agonists or antagonists of the invention, include, but are not limited to, progression, and/or metastases of malignancies and related disorders such as leukemia (including acute leukemias (e.g., acute lymphocytic leukemia, acute myelocytic leukemia (including myeloblastic, promyelocytic, myelomonocytic, monocytic, and erythroleukemia)) and chronic leukemias (e.g., chronic myelocytic (granulocytic) leukemia and chronic lymphocytic leukemia)), polycythemia vera, lymphomas (e.g., Hodgkin's disease and non-Hodgkin's disease), multiple myeloma, Waldenstrom's macroglobulinemia, heavy chain disease, and solid tumors including, but not limited to, sarcomas and carcinomas such as fibrosarcoma, myxosarcoma, liposarcoma, chondrosarcoma, osteogenic sarcoma, chordoma, angiosarcoma, endotheliosarcoma, lymphangiosarcoma, lymphangioendotheliosarcoma, synovioma, mesothelioma, Ewing's tumor, leiomyosarcoma, rhabdomyosarcoma, colon carcinoma, pancreatic cancer, breast cancer, ovarian cancer, prostate cancer, squamous cell carcinoma, basal cell carcinoma, adenocarcinoma, sweat gland carcinoma, sebaceous gland carcinoma, papillary carcinoma, papillary adenocarcinomas, cystadenocarcinoma, medullary carcinoma, bronchogenic carcinoma, renal cell carcinoma, hepatoma, bile duct carcinoma, choriocarcinoma, seminoma, embryonal carcinoma, Wilm's tumor, cervical cancer, testicular tumor, lung carcinoma, small cell lung carcinoma, bladder carcinoma, epithelial carcinoma, glioma, astrocytoma, medulloblastoma, craniopharyngioma, ependymoma, pinealoma, hemangioblastoma, acoustic neuroma, oligodendroglioma, menangioma, melanoma, neuroblastoma, and retinoblastoma.
Diseases associated with increased apoptosis that could be treated, prevented, and/or diagnosed by the polynucleotides or polypeptides, and/or agonists or antagonists of the invention, include AIDS; neurodegenerative diseases, disorders, and/or conditions (such as Alzheimer's disease, Parkinson's disease, Amyotrophic lateral sclerosis, Retinitis pigmentosa, Cerebellar degeneration and brain tumor or prior associated disease); autoimmune diseases, disorders, and/or conditions (such as, multiple sclerosis, Sjogren's syndrome, Hashimoto's thyroiditis, biliary cirrhosis, Behcet's disease, Crohn's disease, polymyositis, systemic lupus erythematosus and immune-related glomerulonephritis and rheumatoid arthritis) myelodysplastic syndromes (such as aplastic anemia), graft v. host disease, ischemic injury (such as that caused by myocardial infarction, stroke and reperfusion injury), liver injury (e.g., hepatitis related liver injury, ischemia/reperfusion injury, cholestosis (bile duct injury) and liver cancer); toxin-induced liver disease (such as that caused by alcohol), septic shock, cachexia and anorexia.
Wound Healing and Epithelial Cell Proliferation In accordance with yet a further aspect of the present invention, there is provided a process for utilizing the polynucleotides or polypeptides, and/or agonists or antagonists of the invention, for therapeutic purposes, for example, to stimulate epithelial cell proliferation and basal keratinocytes for the purpose of wound healing, and to stimulate hair follicle production and healing of dermal wounds.
Polynucleotides or polypeptides, as well as agonists or antagonists of the invention, may be clinically useful in stimulating wound healing including surgical wounds, excisional wounds, deep wounds involving damage of the dermis and epidermis, eye tissue wounds, dental tissue wounds, oral cavity wounds, diabetic ulcers, dermal ulcers, cubitus ulcers, arterial ulcers, venous stasis ulcers, burns resulting from heat exposure or chemicals, and other abnormal wound healing conditions such as uremia, malnutrition, vitamin deficiencies and complications associted with systemic treatment with steroids, radiation therapy and antineoplastic drugs and antimetabolites. Polynucleotides or polypeptides, and/or agonists or antagonists of the invention, could be used to promote dermal reestablishment subsequent to dermal loss The polynucleotides or polypeptides, and/or agonists or antagonists of the invention, could be used to increase the adherence of skin grafts to a wound bed and to stimulate re-epithelialization from the wound bed. The following are a non-exhaustive list of grafts that polynucleotides or polypeptides, agonists or antagonists of the invention, could be used to increase adherence to a wound bed:
autografts, artificial skin, allografts, autodermic graft, autoepdermic grafts, avacular grafts, Blair-Brown grafts, bone graft, brephoplastic grafts, cubs graft, delayed graft, dermic graft, epidermic graft, fascia graft, full thickness graft, heterologous graft, xenograft, homologous graft, hyperplastic graft, lamellar graft, mesh graft, mucosal graft, Ollier-Thiersch graft, omenpal graft, patch graft, pedicle graft, penetrating graft, split skin graft, thick split graft. The polynucleotides or polypeptides, and/or agonists or antagonists of the invention, can be used to promote skin strength and to improve the appearance of aged skin.
It is believed that the polynucleotides or polypeptides, and/or agonists or antagonists of the invention, will also produce changes in hepatocyte proliferation, and epithelial cell proliferation in the lung, breast, pancreas, stomach, small intesting, and large intestine. The polynucleotides or polypeptides, and/or agonists or antagonists of the invention, could promote proliferation of epithelial cells such as sebocytes, hair follicles, hepatocytes, type II pneumocytes, mucin-producing goblet cells, and other epithelial cells and their progenitors contained within the skin, lung, liver, and gastrointestinal tract. The polynucleotides or polypeptides, and/or agonists or antagonists of the invention, may promote proliferation of endothelial cells, keratinocytes, and basal keratinocytes.
The polynucleotides or polypeptides, and/or agonists or antagonists of the invention, could also be used to reduce the side effects of gut toxicity that result from radiation, chemotherapy treatments or viral infections. The polynucleotides or polypeptides, and/or agonists or antagonists of the invention, may have a cytoprotective effect on the small intestine mucosa. The polynucleotides or polypeptides, and/or agonists or antagonists of the invention, may also stimulate healing of mucositis (mouth ulcers) that result from chemotherapy and viral infections.
The polynucleotides or polypeptides, and/or agonists or antagonists of the invention, could further be used in full regeneration of skin in full and partial thickness skin defects, including burns, (i.e., repopulation of hair follicles, sweat glands, and sebaceous glands), treatment of other skin defects such as psoriasis. The polynucleotides or polypeptides, and/or agonists or antagonists of the invention, could be used to treat epidermolysis bullosa, a defect in adherence of the epidermis to the underlying dermis which results in frequent, open and painful blisters by accelerating reepithelialization of these lesions. The polynucleotides or polypeptides, and/or agonists or antagonists of the invention, could also be used to treat gastric and doudenal ulcers and help heal by scar formation of the mucosal lining and regeneration of glandular mucosa and duodenal mucosal lining more rapidly.
Inflamamatory bowel diseases, such as Crohn's disease and ulcerative colitis, are diseases which result in destruction of the mucosal surface of the small or large intestine, respectively. Thus, the polynucleotides or polypeptides, and/or agonists or antagonists of the invention, could be used to promote the resurfacing of the mucosal surface to aid more rapid healing and to prevent progression of inflammatory bowel disease. Treatment with the polynucleotides or polypeptides, and/or agonists or antagonists of the invention, is expected to have a significant effect on the production of mucus throughout the gastrointestinal tract and could be used to protect the intestinal mucosa from injurious substances that are ingested or following surgery.
The polynucleotides or polypeptides, and/or agonists or antagonists of the invention, could be used to treat diseases associate with the under expression of the polynucleotides of the invention.
Moreover, the polynucleotides or polypeptides, and/or agonists or antagonists of the invention, could be used to prevent and heal damage to the lungs due to various pathological states. A growth factor such as the polynucleotides or polypeptides, and/or agonists or antagonists of the invention, which could stimulate proliferation and differentiation and promote the repair of alveoli and brochiolar epithelium to prevent or treat acute or chronic lung damage. For example, emphysema, which results in the progressive loss of aveoli, and inhalation injuries, i.e., resulting from smoke inhalation and burns, that cause necrosis of the bronchiolar epithelium and alveoli could be effectively treated, prevented, and/or diagnosed using the polynucleotides or polypeptides, and/or agonists or antagonists of the invention.
Also, the polynucleotides or polypeptides, and/or agonists or antagonists of the invention, could be used to stimulate the proliferation of and differentiation of type II
pneumocytes, which may help treat or prevent disease such as hyaline membrane diseases, such as infant respiratory distress syndrome and bronchopulmonary displasia, in premature infants.
The polynucleotides or polypeptides, and/or agonists or antagonists of the invention, could stimulate the proliferation and differentiation of hepatocytes and, thus, could be used to alleviate or treat liver diseases and pathologies such as fulminant liver failure caused by cirrhosis, liver damage caused by viral hepatitis and toxic substances (i.e., acetaminophen, carbon tetraholoride and other hepatotoxins known in the art).
S In addition, the polynucleotides or polypeptides, and/or agonists or antagonists of the invention, could be used treat or prevent the onset of diabetes mellitus. In patients with newly diagnosed Types I and II diabetes, where some islet cell function remains, the polynucleotides or polypeptides, and/or agonists or antagonists of the invention, could be used to maintain the islet function so as to alleviate, delay or prevent permanent manifestation of the disease. Also, the polynucleotides or polypeptides, and/or agonists or antagonists of the invention, could be used as an auxiliary in islet cell transplantation to improve or promote islet cell function.
Neurological Diseases Nervous system diseases, disorders, and/or conditions, which can be treated, prevented, and/or diagnosed with the compositions of the invention (e.g., polypeptides, polynucleotides, and/or agonists or antagonists), include, but are not limited to, nervous system injuries, and diseases, disorders, and/or conditions which result in either a disconnection of axons, a diminution or degeneration of neurons, or demyelination. Nervous system lesions which may be treated, prevented, and/or diagnosed in a patient (including human and non-human mammalian patients) according to the invention, include but are not limited to, the following lesions of either the central (including spinal cord, brain) or peripheral nervous systems: (1) ischemic lesions, in which a lack of oxygen in a portion of the nervous system results in neuronal injury or death, including cerebral infarction or ischemia, or spinal cord infarction or ischemia; (2) traumatic lesions, including lesions caused by physical injury or associated with surgery, for example, lesions which sever a portion of the nervous system, or compression injuries; (3) malignant lesions, in which a portion of the nervous system is destroyed or injured by malignant tissue which is either a nervous system associated malignancy or a malignancy derived from non-nervous system tissue; (4) infectious lesions, in which a portion of the nervous system is destroyed or injured as a result of infection, for example, by an abscess or associated with infection by human immunodeficiency virus, herpes zoster, or herpes simplex virus or with Lyme disease, tuberculosis, syphilis; (5) degenerative lesions, in which a portion of the nervous system is destroyed or injured as a result of a degenerative process including but not limited to degeneration associated with Parkinson's disease, Alzheimer's disease, Huntington's chorea, or amyotrophic lateral sclerosis (ALS); (6) lesions associated with nutritional diseases, disorders, and/or conditions, in which a portion of the nervous system is destroyed or injured by a nutritional disorder or disorder of metabolism including but not limited to, vitamin B12 deficiency, folic acid deficiency, Wernicke disease, tobacco-alcohol amblyopia, Marchiafava-Bignami disease (primary degeneration of the corpus callosum), and alcoholic cerebellar degeneration; (7) neurological lesions associated with systemic diseases including, but not limited to, diabetes (diabetic neuropathy, Bell's palsy), systemic lupus erythematosus, carcinoma, or sarcoidosis; (8) lesions caused by toxic substances including alcohol, lead, or particular neurotoxins; and (9) demyelinated lesions in which a portion of the nervous system is destroyed or injured by a demyelinating disease including, but not limited to, multiple sclerosis, human immunodeficiency virus-associated myelopathy, transverse myelopathy or various etiologies, progressive multifocal leukoencephalopathy, and central pontine myelinolysis.
In a preferred embodiment, the polypeptides, polynucleotides, or agonists or antagonists of the invention are used to protect neural cells from the damaging effects of cerebral hypoxia. According to this embodiment, the compositions of the invention are used to treat, prevent, and/or diagnose neural cell injury associated with cerebral hypoxia. In one aspect of this embodiment, the polypeptides, polynucleotides, or agonists or antagonists of the invention are used to treat, prevent, and/or diagnose neural cell injury associated with cerebral ischemia. In another aspect of this embodiment, the polypeptides, polynucleotides, or agonists or antagonists of the invention are used to treat, prevent, and/or diagnose neural cell injury associated with cerebral infarction. In another aspect of this embodiment, the polypeptides, polynucleotides, or agonists or antagonists of the invention are used to treat, prevent, and/or diagnose or prevent neural cell injury associated with a stroke.
In a further aspect of this embodiment, the polypeptides, polynucleotides, or agonists or antagonists of the invention are used to treat, prevent, and/or diagnose neural cell injury associated with a heart attack.
The compositions of the invention which are useful for treating or preventing a nervous system disorder may be selected by testing for biological activity in promoting the survival or differentiation of neurons. For example, and not by way of limitation, compositions of the invention which elicit any of the following effects may be useful according to the invention: (1) increased survival time of neurons in culture;
(2) increased sprouting of neurons in culture or in vivo; (3) increased production of a neuron-associated molecule in culture or in vivo, e.g., choline acetyltransferase or acetylcholinesterase with respect to motor neurons; or (4) decreased symptoms of neuron dysfunction in vivo. Such effects may be measured by any method known in the art. In preferred, non-limiting embodiments, increased survival of neurons may routinely be measured using a method set forth herein or otherwise known in the art, such as, for example, the method set forth in Arakawa et al. (J. Neurosci.
10:3507-3515 (1990)); increased sprouting of neurons may be detected by methods known in the art, such as, for example, the methods set forth in Pestronk et al. (Exp.
Neurol. 70:65-82 (1980)) or Brown et al. (Ann. Rev. Neurosci. 4:17-42 (1981));
increased production of neuron-associated molecules may be measured by bioassay, enzymatic assay, antibody binding, Northern blot assay, etc., using techniques known in the art and depending on the molecule to be measured; and motor neuron dysfunction may be measured by assessing the physical manifestation of motor neuron disorder, e.g., weakness, motor neuron conduction velocity, or functional disability.
In specific embodiments, motor neuron diseases, disorders, and/or conditions that may be treated, prevented, and/or diagnosed according to the invention include, but are not limited to, diseases, disorders, and/or conditions such as infarction, infection, exposure to toxin, trauma, surgical damage, degenerative disease or malignancy that may affect motor neurons as well as other components of the nervous system, as well as diseases, disorders, and/or conditions that selectively affect neurons such as amyotrophic lateral sclerosis, and including, but not limited to, progressive spinal muscular atrophy, progressive bulbar palsy, primary lateral sclerosis, infantile and juvenile muscular atrophy, progressive bulbar paralysis of childhood (Fazio-Londe syndrome), poliomyelitis and the post polio syndrome, and Hereditary Motorsensory Neuropathy (Charcot-Marie-Tooth Disease).
Further, polypeptides or polynucleotides of the invention may play a role in neuronal survival; synapse formation; conductance; neural differentiation, etc. Thus, compositions of the invention (including polynucleotides, polypeptides, and agonists or antagonists) may be used to diagnose and/or treat or prevent diseases or disorders associated with these roles, including, but not limited to, learning and/or cognition disorders. The compositions of the invention may also be useful in the treatment or prevention of neurodegenerative disease states and/or behavioural disorders.
Such neurodegenerative disease states and/or behavioral disorders include, but are not limited to, Alzheimers Disease, Parkinsons Disease, Huntingtons Disease, Tourette Syndrome, schizophrenia, mania, dementia, paranoia, obsessive compulsive disorder, panic disorder, learning disabilities, ALS, psychoses, autism, and altered behaviors, including disorders in feeding, sleep patterns, balance, and perception. In addition, compositions of the invention may also play a role in the treatment, prevention and/or detection of developmental disorders associated with the developing embryo, or sexually-linked disorders.
Additionally, polypeptides, polynucleotides and/or agonists or antagonists of the invention, may be useful in protecting neural cells from diseases, damage, disorders, or injury, associated with cerebrovascular disorders including, but not limited to, carotid artery diseases (e.g., carotid artery thrombosis, carotid stenosis, or Moyamoya Disease), cerebral amyloid angiopathy, cerebral aneurysm, cerebral anoxia, cerebral arteriosclerosis, cerebral arteriovenous malformations, cerebral artery diseases, cerebral embolism and thrombosis (e.g., carotid artery thrombosis, sinus thrombosis, or Wallenberg's Syndrome), cerebral hemorrhage (e.g., epidural or subdural hematoma, or subarachnoid hemorrhage), cerebral infarction, cerebral ischemia (e.g., transient cerebral ischemia, Subclavian Steal Syndrome, or vertebrobasilar insufficiency), vascular dementia (e.g., multi-infarct), leukomalacia, periventricular, and vascular headache (e.g., cluster headache or migraines).
In accordance with yet a further aspect of the present invention, there is provided a process for utilizing polynucleotides or polypeptides, as well as.
agonists or antagonists of the present invention, for therapeutic purposes, for example, to stimulate neurological cell proliferation and/or differentiation. Therefore, polynucleotides, polypeptides, agonists and/or antagonists of the invention may be used to treat and/or detect neurologic diseases. Moreover, polynucleotides or polypeptides, or agonists or antagonists of the invention, can be used as a marker or detector of a particular nervous system disease or disorder.
Examples of neurologic diseases which can be treated or detected with polynucleotides, polypeptides, agonists, and/or antagonists of the present invention include brain diseases, such as metabolic brain diseases which includes phenylketonuria such as maternal phenylketonuria, pyruvate carboxylase deficiency, pyruvate dehydrogenase complex deficiency, Wernicke's Encephalopathy, brain edema, brain neoplasms such as cerebellar neoplasms which include infratentorial neoplasms, cerebral ventricle neoplasms such as choroid plexus neoplasms, hypothalamic neoplasms, supratentorial neoplasms, canavan disease, cerebellar diseases such as cerebellar ataxia which include spinocerebellar degeneration such as ataxia telangiectasia, cerebellar dyssynergia, Friederich's Ataxia, Machado-Joseph Disease, olivopontocerebellar atrophy, cerebellar neoplasms such as infratentorial neoplasms, diffuse cerebral sclerosis such as encephalitis periaxialis, globoid cell leukodystrophy, metachromatic leukodystrophy and subacute sclerosing panencephalitis.
Additional neurologic diseases which can be treated or detected with polynucleotides, polypeptides, agonists, and/or antagonists of the present invention include cerebrovascular disorders (such as carotid artery diseases which include carotid artery thrombosis, carotid stenosis and Moyamoya Disease), cerebral amyloid angiopathy, cerebral aneurysm, cerebral anoxia, cerebral arteriosclerosis, cerebral arteriovenous malformations, cerebral artery diseases, cerebral embolism and thrombosis such as carotid artery thrombosis, sinus thrombosis and Wallenberg's Syndrome, cerebral hemorrhage such as epidural hematoma, subdural hematoma and subarachnoid hemorrhage, cerebral infarction, cerebral ischemia such as transient cerebral ischemia, Subclavian Steal Syndrome and vertebrobasilar insufficiency, vascular dementia such as mufti-infarct dementia, periventricular leukomalacia, vascular headache such as cluster headache and migraine.

Additional neurologic diseases which can be treated or detected with polynucleotides, polypeptides, agonists, and/or antagonists of the present invention include dementia such as AIDS Dementia Complex, presenile dementia such as Alzheimer's Disease and Creutzfeldt-Jakob Syndrome, senile dementia such as Alzheimer's Disease and progressive supranuclear palsy, vascular dementia such as mufti-infarct dementia, encephalitis which include encephalitis periaxialis, viral encephalitis such as epidemic encephalitis, Japanese Encephalitis, St. Louis Encephalitis, tick-borne encephalitis and West Nile Fever, acute disseminated encephalomyelitis, meningoencephalitis such as uveomeningoencephalitic syndrome, Postencephalitic Parkinson Disease and subacute sclerosing panencephalitis, encephalomalacia such as periventricular leukomalacia, epilepsy such as generalized epilepsy which includes infantile spasms, absence epilepsy, myoclonic epilepsy which includes MERRF Syndrome, tonic-clonic epilepsy, partial epilepsy such as complex partial epilepsy, frontal lobe epilepsy and temporal lobe epilepsy, post-traumatic epilepsy, status epilepticus such as Epilepsia Partialis Continua, and Hallervorden-Spatz Syndrome.
Additional neurologic diseases which can be treated or detected with polynucleotides, polypeptides, agonists, and/or antagonists of the present invention include hydrocephalus such as Dandy-Walker Syndrome and normal pressure hydrocephalus, hypothalamic diseases such as hypothalamic neoplasms, cerebral malaria, narcolepsy which includes cataplexy, bulbar poliomyelitis, cerebri pseudotumor, Rett Syndrome, Reye's Syndrome, thalamic diseases, cerebral toxoplasmosis, intracranial tuberculoma and Zellweger Syndrome, central nervous system infections such as AIDS Dementia Complex, Brain Abscess, subdural empyema, encephalomyelitis such as Equine Encephalomyelitis, Venezuelan Equine Encephalomyelitis, Necrotizing Hemorrhagic Encephalomyelitis, Visna, and cerebral malaria.
Additional neurologic diseases which can be treated or detected with polynucleotides, polypeptides, agonists, and/or antagonists of the present invention include meningitis such as arachnoiditis, aseptic meningtitis such as viral meningtitis which includes lymphocytic choriomeningitis, Bacterial meningtitis which includes Haemophilus Meningtitis, Listeria Meningtitis, Meningococcal Meningtitis such as Waterhouse-Friderichsen Syndrome, Pneumococcal Meningtitis and meningeal tuberculosis, fungal meningitis such as Cryptococcal Meningtitis, subdural effusion, meningoencephalitis such as uvemeningoencephalitic syndrome, myelitis such as transverse myelitis, neurosyphilis such as tabes dorsalis, poliomyelitis which includes bulbar poliomyelitis and postpoliomyelitis syndrome, prion diseases (such as Creutzfeldt-Jakob Syndrome, Bovine Spongiform Encephalopathy, Gerstmann-Straussler Syndrome, Kuru, Scrapie), and cerebral toxoplasmosis.
Additional neurologic diseases which can be treated or detected with polynucleotides, polypeptides, agonists, and/or antagonists of the present invention include central nervous system neoplasms such as brain neoplasms that include cerebellar neoplasms such as infratentorial neoplasms, cerebral ventricle neoplasms such as choroid plexus neoplasms, hypothalamic neoplasms and supratentorial neoplasms, meningeal neoplasms, spinal cord neoplasms which include epidural neoplasms, demyelinating diseases such as Canavan Diseases, diffuse cerebral sceloris which includes adrenoleukodystrophy, encephalitis periaxialis, globoid cell leukodystrophy, diffuse cerebral sclerosis such as metachromatic leukodystrophy, allergic encephalomyelitis, necrotizing hemorrhagic encephalomyelitis, progressive multifocal leukoencephalopathy, multiple sclerosis, central pontine myelinolysis, transverse myelitis, neuromyelitis optics, Scrapie, Swayback, Chronic Fatigue Syndrome, Visna, High Pressure Nervous Syndrome, Meningism, spinal cord diseases such as amyotonia congenita, amyotrophic lateral sclerosis, spinal muscular atrophy such as Werdnig-Hoffmann Disease, spinal cord compression, spinal cord neoplasms such as epidural neoplasms, syringomyelia, Tabes Dorsalis, Stiff Man Syndrome, mental retardation such as Angelman Syndrome, Cri-du-Chat Syndrome, De Lange's Syndrome, Down Syndrome, Gangliosidoses such as gangliosidoses G(M1), Sandhoff Disease, Tay-Sachs Disease, Hartnup Disease, homocystinuria, Laurence-Moon-Biedl Syndrome, Lesch-Nyhan Syndrome, Maple Syrup Urine Disease, mucolipidosis such as fucosidosis, neuronal ceroid-lipofuscinosis, oculocerebrorenal syndrome, phenylketonuria such as maternal phenylketonuria, Prader-Willi Syndrome, Rett Syndrome, Rubinstein-Taybi Syndrome, Tuberous Sclerosis, WAGR Syndrome, nervous system abnormalities such as holoprosencephaly, neural tube defects such as anencephaly which includes hydrangencephaly, Arnold-Chain Deformity, encephalocele, meningocele, meningomyelocele, spinal dysraphism such as spina bifida cysticawand spina bifida occulta.
Additional neurologic diseases which can be treated or detected with polynucleotides, polypeptides, agonists, and/or antagonists of the present invention include hereditary motor and sensory neuropathies which include Charcot-Marie Disease, Hereditary optic atrophy, Refsum's Disease, hereditary spastic paraplegia, Werdnig-Hoffmann Disease, Hereditary Sensory and Autonomic Neuropathies such as Congenital Analgesia and Familial Dysautonomia, Neurologic manifestations (such as agnosia that include Gerstmann's Syndrome, Amnesia such as retrograde amnesia, apraxia, neurogenic bladder,. cataplexy, communicative disorders such as hearing disorders that includes deafness, partial hearing loss, loudness recruitment and tinnitus, language disorders such as aphasia which include agraphia, anomia, broca aphasia, and Wernicke Aphasia, Dyslexia such as Acquired Dyslexia, language development disorders, speech disorders such as aphasia which includes anomia, broca aphasia and Wernicke Aphasia, articulation disorders, communicative disorders such as speech disorders which include dysarthria, echolalia, mutism and stuttering, voice disorders such as aphonia and hoarseness, decerebrate state, delirium, fasciculation, hallucinations, meningism, movement disorders such as angelman syndrome, ataxia, athetosis, chorea, dystonia, hypokinesia, muscle hypotonia, myoclonus, tic, torticollis and tremor, muscle hypertonia such as muscle rigidity such as stiff man syndrome, muscle spasticity, paralysis such as facial paralysis which includes Herpes Zoster Oticus, Gastroparesis, Hemiplegia, ophthalmoplegia such as diplopia, Duane's Syndrome, Horner's 'Syndrome, Chronic progressive external ophthalmoplegia such as Kearns Syndrome, Bulbar Paralysis, Tropical Spastic Paraparesis, Paraplegia such as Brown-Sequard Syndrome, quadriplegia, respiratory paralysis and vocal cord paralysis, paresis, phantom limb, taste disorders such as ageusia and dysgeusia, vision disorders such as amblyopia, blindness, color vision defects, diplopia, hemianopsia, scotoma and subnormal vision, sleep disorders such as hypersomnia which includes Kleine-Levin Syndrome, insomnia, and somnambulism, spasm such as trismus, unconsciousness such as coma, persistent vegetative state and syncope and vertigo, neuromuscular diseases such as amyotonia congenita, amyotrophic lateral sclerosis, Lambert-Eaton Myasthenic Syndrome, motor neuron disease, muscular atrophy such as spinal muscular atrophy, Charcot-Marie Disease and Werdnig-Hoffmann Disease, Postpoliomyelitis Syndrome, Muscular Dystrophy, Myasthenia Gravis, Myotonia Atrophica, Myotonia Confenita, Nemaline Myopathy, Familial Periodic Paralysis, Multiplex Paramyloclonus, Tropical Spastic Paraparesis and Stiff Man Syndrome, peripheral nervous system diseases such as acrodynia, amyloid neuropathies, autonomic nervous system diseases such as Adie's Syndrome, Barre-Lieou Syndrome, Familial Dysautonomia, Homer's Syndrome, Reflex Sympathetic Dystrophy and Shy-Drager Syndrome, Cranial Nerve Diseases such as Acoustic Nerve Diseases such as Acoustic Neuroma which includes Neurofibromatosis 2, Facial Nerve Diseases such as Facial Neuralgia,Melkersson-Rosenthal Syndrome, ocular motility disorders which includes amblyopia, nystagmus, oculomotor nerve paralysis, ophthalmoplegia such as Duane's Syndrome, Homer's Syndrome, Chronic Progressive External Ophthalmoplegia which includes Kearns Syndrome, Strabismus such as Esotropia and Exotropia, Oculomotor Nerve Paralysis, 1 S Optic Nerve Diseases such as Optic Atrophy which includes Hereditary Optic Atrophy, Optic Disk Drusen, Optic Neuritis such as Neuromyelitis Optica, Papilledema, Trigeminal Neuralgia, Vocal Cord Paralysis, Demyelinating Diseases such as Neuromyelitis Optica and Swayback, and Diabetic neuropathies such as diabetic foot.
Additional neurologic diseases which can be treated or detected with polynucleotides, polypeptides, agonists, and/or antagonists of the present invention include nerve compression syndromes such as carpal tunnel syndrome, tarsal tunnel syndrome, thoracic outlet syndrome such as cervical rib syndrome, ulnar nerve compression syndrome, neuralgia such as causalgia, cervico-brachial neuralgia, facial neuralgia and trigeminal neuralgia, neuritis such as experimental allergic neuritis, optic neuritis, polyneuritis, polyradiculoneuritis and radiculities such as polyradiculitis, hereditary motor and sensory neuropathies such as Charcot-Marie Disease, Hereditary Optic Atrophy, Refsum's Disease, Hereditary Spastic Paraplegia and Werdnig-Hoffmann Disease, Hereditary Sensory and Autonomic Neuropathies which include Congenital Analgesia and Familial Dysautonomia, POEMS Syndrome, Sciatica, Gustatory Sweating and Tetany).

Infectious Disease A polypeptide or polynucleotide and/or agonist or antagonist of the present invention can be used to treat, prevent, and/or diagnose infectious agents.
For example, by increasing the immune response, particularly increasing the proliferation and differentiation of B and/or T cells, infectious diseases may be treated, prevented, and/or diagnosed. The immune response may be increased by either enhancing an existing immune response, or by initiating a new immune response.
Alternatively, polypeptide or polynucleotide and/or agonist or antagonist of the present invention may also directly inhibit the infectious agent, without necessarily eliciting an immune response.
Viruses are one example of an infectious agent that can cause disease or symptoms that can be treated, prevented, and/or diagnosed by a polynucleotide or polypeptide and/or agonist or antagonist of the present invention. Examples of viruses, include, but are not limited to Examples of viruses, include, but are not limited to the following DNA and RNA viruses and viral families: Arbovirus, Adenoviridae, Arenaviridae, Arterivirus, Birnaviridae, Bunyaviridae, Caliciviridae, Circoviridae, Coronaviridae, Dengue, EBV, HIV, Flaviviridae, Hepadnaviridae (Hepatitis), Herpesviridae (such as, Cytomegalovirus, Herpes Simplex, Herpes Zoster), Mononegavirus (e.g., Paramyxoviridae, Morbillivirus, Rhabdoviridae), Orthomyxoviridae (e.g., Influenza A, Influenza B, and parainfluenza), Papiloma virus, Papovaviridae, Parvoviridae, Picornaviridae, Poxviridae (such as Smallpox or Vaccinia), Reoviridae (e.g., Rotavirus), Retroviridae (HTLV-I, HTLV-II, Lentivirus), and Togaviridae (e.g., Rubivirus). Viruses falling within these families can cause a variety of diseases or symptoms, including, but not limited to: arthritis, bronchiollitis, respiratory syncytial virus, encephalitis, eye infections (e.g., conjunctivitis, keratitis), chronic fatigue syndrome, hepatitis (A, B, C, E, Chronic Active, Delta), Japanese B
encephalitis, Junin, Chikungunya, Rift Valley fever, yellow fever, meningitis, opportunistic infections (e.g., AIDS), pneumonia, Burkitt's Lymphoma, chickenpox, hemorrhagic fever, Measles, Mumps, Parainfluenza, Rabies, the common cold, Polio, leukemia, Rubella, sexually transmitted diseases, skin diseases (e.g., Kaposi's, warts), and viremia. polynucleotides or polypeptides, or agonists or antagonists of the invention, can be used to treat, prevent, and/or diagnose any of these symptoms or diseases. In specific embodiments, polynucleotides, polypeptides, or agonists or antagonists of the invention are used to treat, prevent, and/or diagnose:
meningitis, Dengue, EBV, andlor hepatitis (e.g., hepatitis B). In an additional specific embodiment polynucleotides, polypeptides, or agonists or antagonists of the invention are used to treat patients nonresponsive to one or more other commercially available hepatitis vaccines. In a further specific embodiment polynucleotides, polypeptides, or agonists or antagonists of the invention are used to treat, prevent, and/or diagnose AIDS.
Similarly, bacterial or fungal agents that can cause disease or symptoms and that can be treated, prevented, and/or diagnosed by a polynucleotide or polypeptide and/or agonist or antagonist of the present invention include, but not limited to, include, but not limited to, the following Gram-Negative and Gram-positive bacteria and bacterial families and fungi: Actinomycetales (e.g., Corynebacterium, Mycobacterium, Norcardia), Cryptococcus neoformans, Aspergillosis, Bacillaceae (e.g., Anthrax, Clostridium), Bacteroidaceae, Blastomycosis, Bordetella, Borrelia (e.g., Borrelia burgdorferi), Brucellosis, Candidiasis, Campylobacter, Coccidioidomycosis, Cryptococcosis, Dermatocycoses, E. coli (e.g., Enterotoxigenic E. coli and Enterohemorrhagic E. coli), Enterobacteriaceae (Klebsiella, Salmonella (e.g., Salmonella typhi, and Salmonella paratyphi), Serratia, Yersinia), Erysipelothrix, Helicobacter, Legionellosis, Leptospirosis, Listeria, Mycoplasmatales, Mycobacterium leprae, Vibrio cholerae, Neisseriaceae (e.g., Acinetobacter, Gonorrhea, Menigococcal), Meisseria meningitidis, Pasteurellacea Infections (e.g., Actinobacillus, Heamophilus (e.g., Heamophilus influenza type B), Pasteurella), Pseudomonas, Rickettsiaceae, Chlamydiaceae, Syphilis, Shigella spp., Staphylococcal, Meningiococcal, Pneumococcal and Streptococcal (e.g., Streptococcus pneumoniae and Group B Streptococcus). These bacterial or fungal families can cause the following diseases or symptoms, including, but not limited to:
bacteremia, endocarditis, eye infections (conjunctivitis, tuberculosis, uveitis), gingivitis, opportunistic infections (e.g., AIDS related infections), paronychia, prosthesis-related infections, Reiter's Disease, respiratory tract infections, such as Whooping Cough or Empyema, sepsis, Lyme Disease, Cat-Scratch Disease, Dysentery, Paratyphoid Fever, food poisoning, Typhoid, pneumonia, Gonorrhea, meningitis (e.g., mengitis types A and B), Chlamydia, Syphilis, Diphtheria, Leprosy, Paratuberculosis, Tuberculosis, Lupus, Botulism, gangrene, tetanus, impetigo, Rheumatic Fever, Scarlet Fever, sexually transmitted diseases, skin diseases (e.g., cellulitis, dermatocycoses), toxemia, urinary tract infections, wound infections.
Polynucleotides or polypeptides, agonists or antagonists of the invention, can be used to treat, prevent, and/or diagnose any of these symptoms or diseases. In specific embodiments, polynucleotides, polypeptides, agonists or antagonists of the invention are used to treat, prevent, and/or diagnose: tetanus, Diptheria, botulism, and/or meningitis type B.
Moreover, parasitic agents causing disease or symptoms that can be treated, prevented, and/or diagnosed by a polynucleotide or polypeptide and/or agonist or antagonist of the present invention include, but not limited to, the following families or class: Amebiasis, Babesiosis, Coccidiosis, Cryptosporidiosis, Dientamoebiasis, Dourine, Ectoparasitic, Giardiasis, Helminthiasis, Leishmaniasis, Theileriasis, Toxoplasmosis, Trypanosomiasis, and Trichomonas and Sporozoans (e.g., Plasmodium virax, Plasmodium falciparium, Plasmodium malariae and Plasmodium ovale). These parasites can cause a variety of diseases or symptoms, including, but not limited to: Scabies, Trombiculiasis, eye infections, intestinal disease (e.g., dysentery, giardiasis), liver disease, lung disease, opportunistic infections (e.g., AIDS
related), malaria, pregnancy complications, and toxoplasmosis. polynucleotides or polypeptides, or agonists or antagonists of the invention, can be used totreat, prevent, and/or diagnose any of these symptoms or diseases. In specific embodiments, polynucleotides, polypeptides, or agonists or antagonists of the invention are used to treat, prevent, and/or diagnose malaria.
Preferably, treatment or prevention using a polypeptide or polynucleotide and/or agonist or antagonist of the present invention could either be by administering an effective amount of a polypeptide to the patient, or by removing cells from the patient, supplying the cells with a polynucleotide of the present invention, and returning the engineered cells to the patient (ex vivo therapy). Moreover, the polypeptide or polynucleotide of the present invention can be used as an antigen in a vaccine to raise an immune response against infectious disease.

Regeneration A polynucleotide or polypeptide and/or agonist or antagonist of the present invention can be used to differentiate, proliferate, and attract cells, leading to the regeneration of tissues. (See, Science 276:59-87 (1997).) The regeneration of tissues could be used to repair, replace, or protect tissue damaged by congenital defects, trauma (wounds, burns, incisions, or ulcers), age, disease (e.g. osteoporosis, osteocarthritis, periodontal disease, liver failure), surgery, including cosmetic plastic surgery, fibrosis, reperfusion injury, or systemic cytokine damage.
Tissues that could be regenerated using the present invention include organs (e.g., pancreas, liver, intestine, kidney, skin, endothelium), muscle (smooth, skeletal or cardiac), vasculature (including vascular and lymphatics), nervous, hematopoietic, and skeletal (bone, cartilage, tendon, and ligament) tissue. Preferably, regeneration occurs without or decreased scarnng. Regeneration also may include angiogenesis.
Moreover, a polynucleotide or polypeptide and/or agonist or antagonist of the present invention may increase regeneration of tissues difficult to heal. For example, increased tendon/ligament regeneration would quicken recovery time after damage.
A polynucleotide or polypeptide and/or agonist or antagonist of the present invention could also be used prophylactically in an effort to avoid damage. Specific diseases that could be treated, prevented, and/or diagnosed include of tendinitis, carpal tunnel syndrome, and other tendon or ligament defects. A further example of tissue regeneration of non-healing wounds includes pressure ulcers, ulcers associated with vascular insufficiency, surgical, and traumatic wounds.
Similarly, nerve and brain tissue could also be regenerated by using a polynucleotide or polypeptide and/or agonist or antagonist of the present invention to proliferate and differentiate nerve cells. Diseases that could be treated, prevented, and/or diagnosed using this method-include central and peripheral nervous system diseases, neuropathies, or mechanical and traumatic diseases, disorders, and/or conditions (e.g., spinal cord disorders, head trauma, cerebrovascular disease, and stoke). Specifically, diseases associated with peripheral nerve injuries, peripheral neuropathy (e.g., resulting from chemotherapy or other medical therapies), localized neuropathies, and central nervous system diseases (e.g., Alzheimer's disease, Parkinson's disease, Huntington's disease, amyotrophic lateral sclerosis, and Shy-Drager syndrome), could all be treated, prevented, and/or diagnosed using the polynucleotide or polypeptide and/or agonist or antagonist of the present invention.
Chemotaxis S A polynucleotide or polypeptide and/or agonist or antagonist of the present invention may have chemotaxis activity. A chemotaxic molecule attracts or mobilizes cells (e.g., monocytes, fibroblasts, neutrophils, T-cells, mast cells, eosinophils, epithelial and/or endothelial cells) to a particular site in the body, such as inflammation, infection, or site of hyperproliferation. The mobilized cells can then fight off and/or heal the particular trauma or abnormality.
A polynucleotide or polypeptide and/or agonist or antagonist of the present invention may increase chemotaxic activity of particular cells. These chemotactic molecules can then be used to treat, prevent, and/or diagnose inflammation, infection, hyperproliferative diseases, disorders, and/or conditions, or any immune system disorder by increasing the number of cells targeted to a particular location in the body.
For example, chemotaxic molecules can be used to treat, prevent, and/or diagnose wounds and other trauma to tissues by attracting immune cells to the injured location.
Chemotactic molecules of the present invention can also attract fibroblasts, which can be used to treat, prevent, and/or diagnose wounds.
It is also contemplated that a polynucleotide or polypeptide and/or agonist or antagonist of the present invention may inhibit chemotactic activity. These molecules could also be used totreat, prevent, and/or diagnose diseases, disorders, and/or conditions. Thus, a polynucleotide or polypeptide and/or agonist or antagonist of the present invention could be used as an inhibitor of chemotaxis.
Binding Activity A polypeptide of the present invention may be used to screen for molecules that bind to the polypeptide or for molecules to which the polypeptide binds.
The binding of the polypeptide and the molecule may activate (agonist), increase, inhibit (antagonist), or decrease activity of the polypeptide or the molecule bound.
Examples of such molecules include antibodies, oligonucleotides, proteins (e.g., receptors),or small molecules.

Preferably, the molecule is closely related to the natural ligand of the polypeptide, e.g., a fragment of the ligand, or a natural substrate, a ligand, a structural or functional mimetic. (See, Coligan et al., Current Protocols in Immunology 1(2):Chapter 5 (1991).) Similarly, the molecule can be closely related to the natural S receptor to which the polypeptide binds, or at least, a fragment of the receptor capable of being bound by the polypeptide (e.g., active site). In either case, the molecule can be rationally designed using known techniques.
Preferably, the screening for these molecules involves producing appropriate cells which express the polypeptide, either as a secreted protein or on the cell membrane. Preferred cells include cells from mammals, yeast, Drosophila, or E.
coli.
Cells expressing the polypeptide (or cell membrane containing the expressed polypeptide) are then preferably contacted with a test compound potentially containing the molecule to observe binding, stimulation, or inhibition of activity of either the polypeptide or the molecule.
The assay may simply test binding of a candidate compound to the polypeptide, wherein binding is detected by a label, or in an assay involving competition with a labeled competitor. Further, the assay may test whether the candidate compound results in a signal generated by binding to the polypeptide.
Alternatively, the assay can be carned out using cell-free preparations, .
polypeptide/molecule affixed to a solid support, chemical libraries, or natural product mixtures. The assay may also simply comprise the steps of mixing a candidate compound with a solution containing a polypeptide, measuring polypeptide/molecule activity or binding, and comparing the polypeptide/molecule activity or binding to a standard.
Preferably, an ELISA assay can measure polypeptide level or activity in a sample (e.g., biological sample) using a monoclonal or polyclonal antibody.
The antibody can measure polypeptide level or activity by either binding, directly or indirectly, to the polypeptide or by competing with the polypeptide for a substrate.
Additionally, the receptor to which a polypeptide of the invention binds can be identified by numerous methods known to those of skill in the art, for example, ligand panning and FACS sorting (Coligan, et al., Current Protocols in Immun., 1(2), Chapter 5, (1991)). For example, expression cloning is employed wherein polyadenylated RNA is prepared from a cell responsive to the polypeptides, for example, NIH3T3 cells which are known to contain multiple receptors for the FGF
family proteins, and SC-3 cells, and a cDNA library created from this RNA is divided into pools and used to transfect COS cells or other cells that are not responsive to the S polypeptides. Transfected cells which are grown on glass slides are exposed to the polypeptide of the present invention, after they have been labelled. The polypeptides can be labeled by a variety of means including iodination or inclusion of a recognition site for a site-specific protein kinase.
Following fixation and incubation, the slides are subjected to auto-radiographic analysis. Positive pools are identified and sub-pools are prepared and re-transfected using an iterative sub-pooling and re-screening process, eventually yielding a single clones that encodes the putative receptor.
As an alternative approach for receptor identification, the labeled polypeptides can be photoaffmity linked with cell membrane or extract preparations that express the receptor molecule. Cross-linked material is resolved by PAGE analysis and exposed to X-ray film. The labeled complex containing the receptors of the polypeptides can be excised, resolved into peptide fragments, and subjected to protein microsequencing. The amino acid sequence obtained from microsequencing would be used to design a set of degenerate oligonucleotide probes to screen a cDNA
library to identify the genes encoding the putative receptors.
Moreover, the techniques of gene-shuffling, motif shuffling, exon-shuffling, and/or codon-shuffling (collectively referred to as "DNA shuffling") may be employed to modulate the activities of polypeptides of the invention thereby effectively generating agonists and antagonists of polypeptides of the invention. See generally, U.S. Patent Nos. 5,605,793, 5,811,238, 5,830,721, 5,834,252, and 5,837,458, and Patten, P. A., et al., Curr. Opinion Biotechnol. 8:724-33 (1997);
Harayama, S. Trends Biotechnol. 16(2):76-82 (1998); Hansson, L. O., et al., J.
Mol.
Biol. 287:265-76 (1999); and Lorenzo, M. M. and Blasco, R. Biotechniques 24(2):308-13 (1998) (each of these patents and publications are hereby incorporated by reference). In one embodiment, alteration of polynucleotides and corresponding polypeptides of the invention may be achieved by DNA shuffling. . DNA
shuffling involves the assembly of two or more DNA segments into a desired polynucleotide sequence of the invention molecule by homologous, or site-specific, recombination.
In another embodiment, polynucleotides and corresponding polypeptides of the invention may be alterred by being subjected to random mutagenesis by error-prone PCR, random nucleotide insertion or other methods prior to recombination. In S another embodiment, one or more components, motifs, sections, parts, domains, fragments, etc., of the polypeptides of the invention may be recombined with one or more components, motifs, sections, parts, domains, fragments, etc. of one or more heterologous molecules. In preferred embodiments, the heterologous molecules are family members. In further preferred embodiments, the heterologous molecule is a growth factor such as, for example, platelet-derived growth factor (PDGF), insulin-like growth factor (IGF-I), transforming growth factor (TGF)-alpha, epidermal growth factor (EGF), fibroblast growth factor (FGF), TGF-beta, bone morphogenetic protein (BMP)-2, BMP-4, BMP-5, BMP-6, BMP-7, activins A and B, decapentaplegic(dpp), 60A, OP-2, dorsalin, growth differentiation factors (GDFs), nodal, MIS, inhibin-alpha, TGF-betal, TGF-beta2, TGF-beta3, TGF-betas, and glial-derived neurotrophic factor (GDNF).
Other preferred fragments are biologically active fragments of the polypeptides of the invention. Biologically active fragments are those exhibiting activity similar, but not necessarily identical, to an activity of the polypeptide. The biological activity of the fragments may include an improved desired activity, or a decreased undesirable activity.
Additionally, this invention provides a method of screening compounds to identify those which modulate the action of the polypeptide of the present invention.
An example of such an assay comprises combining a mammalian fibroblast cell, a the polypeptide of the present invention, the compound to be screened and 3[H]
thymidine under cell culture conditions where the fibroblast cell would normally proliferate. A control assay may be performed in the absence of the compound to be screened and compared to the amount of fibroblast proliferation in the presence of the compound to determine if the compound stimulates proliferation by determining the uptake of 3[H] thymidine in each case. The amount of fibroblast cell proliferation is measured by liquid scintillation chromatography which measures the incorporation of, 3 [H] thymidine. Both agonist and antagonist compounds may be identified by this procedure.
In another method, a mammalian cell or membrane preparation expressing a receptor for a polypeptide of the present invention is incubated with a labeled polypeptide of the present invention in the presence of the compound. The ability of the compound to enhance or block this interaction could then be measured.
Alternatively, the response of a known second messenger system following interaction of a compound to be screened and the receptor is measured and the ability of the compound to bind to the receptor and elicit a second messenger response is measured to determine if the compound is a potential agonist or antagonist.
Such second messenger systems include but are not limited to, CAMP guanylate cyclase, ion channels or phosphoinositide hydrolysis.
All of these above assays can be used as diagnostic or prognostic markers.
The molecules discovered using these assays can be used to treat, prevent, and/or diagnose disease or to bring about a particular result in a patient (e.g., blood vessel growth) by activating or inhibiting the polypeptide/molecule. Moreover, the assays can discover agents which may inhibit or enhance the production of the polypeptides of the invention from suitably manipulated cells or tissues. Therefore, the invention includes a method of identifying compounds which bind to the polypeptides of the invention comprising the steps of: (a) incubating a candidate binding compound with the polypeptide; and (b) determining if binding has occurred. Moreover, the invention includes a method of identifying agonists/antagonists comprising the steps of: (a) incubating a candidate compound with the polypeptide, (b) assaying a biological activity , and (b) determining if a biological activity of the polypeptide has been altered.
Also, one could identify molecules bind a polypeptide of the invention .
experimentally by using the beta-pleated sheet regions contained in the polypeptide sequence of the protein. Accordingly, specific embodiments of the invention are directed to polynucleotides encoding polypeptides which comprise, or alternatively consist of, the amino acid sequence of each beta pleated sheet regions in a disclosed polypeptide sequence. Additional embodiments of the invention are directed to polynucleotides encoding polypeptides which comprise, or alternatively consist of, any combination or all of contained in the polypeptide sequences of the invention.
Additional preferred embodiments of the invention are directed to polypeptides which comprise, or alternatively consist of, the amino acid sequence of each of the beta pleated sheet regions in one of the polypeptide sequences of the invention.
Additional embodiments of the invention are directed to polypeptides which comprise, or alternatively consist of, any combination or all of the beta pleated sheet regions in one of the polypeptide sequences of the invention.
Targeted Delivery In another embodiment, the invention provides a method of delivering compositions to targeted cells expressing a receptor for a polypeptide of the invention, or cells expressing a cell bound form of a polypeptide of the invention.
As discussed herein, polypeptides or antibodies of the invention may be associated with heterologous polypeptides, heterologous nucleic acids, toxins, or prodrugs via hydrophobic, hydrophilic, ionic and/or covalent interactions. In one embodiment, the invention provides a method for the specific delivery of compositions of the invention to cells by administering polypeptides of the invention (including antibodies) that are associated with heterologous polypeptides or nucleic acids. In one example, the invention provides a method for delivering a therapeutic protein into the targeted cell. In another example, the invention provides a method for delivering a single stranded nucleic acid (e.g., antisense or ribozymes) or double stranded nucleic acid (e.g., DNA that can integrate into the cell's genome or replicate episomally and that can be transcribed) into the targeted cell.
In another embodiment, the invention provides a method for the specific destruction of cells (e.g., the destruction of tumor cells) by administering polypeptides of the invention (e.g., polypeptides of the invention or antibodies of the invention) in association with toxins or cytotoxic prodrugs.
By "toxin" is meant compounds that bind and activate endogenous cytotoxic effector systems, radioisotopes, holotoxins, modified toxins, catalytic subunits of toxins, or any molecules or enzymes not normally present in or. on the surface of a cell that under defined conditions cause the cell's death. Toxins that may be used according to the methods of the invention include, but are not limited to, radioisotopes known in the art, compounds such as, for example, antibodies (or complement fixing containing portions thereof) that bind an inherent or induced endogenous cytotoxic effector system, thymidine kinase, endonuclease, RNAse, alpha toxin, ricin, abrin, Pseudomonas exotoxin A, diphtheria toxin, saporin, momordin, gelonin, pokeweed antiviral protein, alpha-sarcin and cholera toxin. By "cytotoxic prodrug" is meant a non-toxic compound that is converted by an enzyme, normally present in the cell, into a cytotoxic compound. Cytotoxic prodrugs that may be used according to the methods of the invention include, but are not limited to, glutamyl derivatives of benzoic acid mustard alkylating agent, phosphate derivatives of etoposide or mitomycin C, cytosine arabinoside, daunorubisin, and phenoxyacetamide derivatives of doxorubicin.
Drug Screening Further contemplated is the use of the polypeptides of the present invention, or the polynucleotides encoding these polypeptides, to screen for molecules which modify the activities of the polypeptides of the present invention. Such a method would include contacting the polypeptide of the present invention with a selected compounds) suspected of having antagonist or agonist activity, and assaying the activity of these polypeptides following binding.
This invention is particularly useful for screening therapeutic compounds by using the polypeptides of the present invention, or binding fragments thereof, in any of a variety of drug screening techniques. The polypeptide or fragment employed in such a test may be affixed to a solid support, expressed on a cell surface, free in solution, or located intracellularly. One method of drug screening utilizes eukaryotic or prokaryotic host cells which are stably transformed with recombinant nucleic acids expressing the polypeptide or fragment. Drugs are screened against such transformed cells in competitive binding assays. One may measure, for example, the formulation of complexes between the agent being tested and a polypeptide of the present invention.
Thus, the present invention provides methods of screening for drugs or any other agents which affect activities mediated by the polypeptides of the present invention. These methods comprise contacting such an agent with a polypeptide of the present invention or a fragment thereof and assaying for the presence of a complex between the agent and the polypeptide or a fragment thereof, by methods well known in the art. In such a competitive binding assay, the agents to screen are typically labeled. Following incubation, free agent is separated from that present in bound form, and the amount of free or uncomplexed label is a measure of the ability of a particular agent to bind to the polypeptides of the present invention.
Another technique for drug screening provides high throughput screening for compounds having suitable binding affinity to the polypeptides of the present invention, and is described in great detail in European Patent Application 84/03564, published on September 13, 1984, which is incorporated herein by reference herein.
Briefly stated, large numbers of different small peptide test compounds are synthesized on a solid substrate, such as plastic pins or some other surface.
The peptide test compounds are reacted with polypeptides of the present invention and washed. Bound polypeptides are then detected by methods well known in the art.
Purified polypeptides are coated directly onto plates for use in the aforementioned drug screening techniques. In addition, non-neutralizing antibodies may be used to capture the peptide and immobilize it on the solid support.
This invention also contemplates the use of competitive drug screening assays in which neutralizing antibodies capable of binding polypeptides of the present invention specifically compete with a test compound for binding to the polypeptides or fragments thereof. In this manner, the antibodies are used to detect the presence of any peptide which shares one or more antigenic epitopes with a polypeptide of the invention.
Polvnentides of the Invention Binding Peptides and Other Molecules The invention also encompasses screening methods for identifying polypeptides and nonpolypeptides that bind polypeptides of the invention, and the polypeptide of the invention binding molecules identified thereby. These binding molecules are useful, for example, as agonists and antagonists of the polypeptides of the invention. Such agonists and antagonists can be used, in accordance with the invention, in the therapeutic embodiments described in detail, below.
This method comprises the steps of:

a. contacting a polypeptide of the invention with a plurality of molecules;
and b. identifying a molecule that binds the polypeptide of the invention.
The step of contacting the polypeptide of the invention with the plurality of molecules may be effected in a number of ways. For example, one may contemplate immobilizing the polypeptide of the invention on a solid support and bringing a solution of the plurality of molecules in contact with the immobilized polypeptide of the invention. Such a procedure would be akin to an affinity chromatographic process, with the affinity matrix being comprised of the immobilized polypeptide of the invention. The molecules having a selective affinity for the polypeptide of the invention can then be purified by affinity selection. The nature of the solid support, process for attachment of the polypeptide of the invention to the solid support, solvent, and conditions of the affinity isolation or selection are largely conventional and well known to those of ordinary skill in the art.
Alternatively, one may also separate a plurality of polypeptides into substantially separate fractions comprising a subset of or individual polypeptides. For instance, one can separate the plurality of polypeptides by gel electrophoresis, column chromatography, or like method known to those of ordinary skill for the separation of polypeptides. The individual polypeptides can also be produced by a transformed host cell in such a way as to be expressed on or about its outer surface (e.g., a recombinant phage). Individual isolates can then be "probed" by the polypeptide of the invention, optionally in the presence of an inducer should one be required for expression, to determine if any selective affinity interaction takes place between the polypeptide of the invention and the individual clone. Prior to contacting the polypeptide of the invention with each fraction comprising individual polypeptides, the polypeptides could first be transferred to a solid support for additional convenience. Such a solid support may simply be apiece of filter membrane, such as one made of nitrocellulose or nylon. In this manner, positive clones could be identified from a collection of transformed host cells of an expression library, which harbor a DNA construct encoding a polypeptide having a selective affinity for a polypeptide of the invention.
Furthermore, the amino acid sequence of the polypeptide having a selective affinity for the polypeptide of the invention can be determined directly by conventional means or the coding sequence of the DNA encoding the polypeptide can frequently be determined more conveniently. The primary sequence can then be deduced from the corresponding DNA sequence. If the amino acid sequence is to be determined from the polypeptide itself, one may use microsequencing techniques. The sequencing technique may include mass spectroscopy.
In certain situations, it may be desirable to wash away any unbound polypeptide of the invention, or alterntatively, unbound polypeptides, from a mixture of the polypeptide of the invention and the plurality of polypeptides prior to attempting to determine or to detect the presence of a selective affinity interaction.
Such a wash step may be particularly desirable when the polypeptide of the invention or the plurality of polypeptides is bound to a solid support.
The plurality of molecules provided according to this method may be provided by way of diversity libraries, such as random or combinatorial peptide or nonpeptide libraries which can be screened for molecules that specifically bind to a polypeptide of the invention. Many libraries are known in the art that can be used, e.g., chemically synthesized libraries, recombinant (e.g., phage display libraries), and in vitro translation-based libraries. Examples of chemically synthesized libraries are described in Fodor et al., 1991, Science 251:767-773; Houghten et al., 1991, Nature 354:84-86; Lam et al., 1991, Nature 354:82-84; Medynski, 1994, Bio/Technology 12:709-710;Gallop et al., 1994, J. Medicinal Chemistry 37(9):1233-1251;
Ohlmeyer et al., 1993, Proc. Natl. Acad. Sci. USA 90:10922-10926; Erb et al., 1994, Proc. Natl.
Acad. Sci. USA 91:11422-11426; Houghten et al., 1992, Biotechniques 13:412;
Jayawickreme et al., 1994, Proc. Natl. Acad. Sci. USA 91:1614-1618; Salmon et al., 1993, Proc. Natl. Acad. Sci. USA 90:11708-11712; PCT Publication No. WO
93/20242; and Brenner and Lerner, 1992, Proc. Natl. Acad. Sci. USA 89:5381-5383.
Examples of phage display libraries are described in Scott and Smith, 1990, Science 249:386-390; Devlin et al., 1990, Science, 249:404-406; Christian, R.
B., et al., 1992, J. Mol. Biol. 227:711-718); Lenstra, 1992, J. Immunol. Meth.
152:149-157;
Kay et al., 1993, Gene 128:59-65; and PCT Publication No. WO 94/18318 dated Aug.
18, 1994.
In vitro translation-based libraries include but are not limited to those described in PCT Publication No. WO 91/05058 dated Apr. 18, 1991; and Mattheakis et al., 1994, Proc. Natl. Acad. Sci. USA 91:9022-9026.

By way of examples of nonpeptide libraries, a benzodiazepine library (see e.g., Bunin et al., 1994, Proc. Natl. Acad. Sci. USA 91:4708-4712) can be adapted for use. Peptoid libraries (Simon et al., 1992, Proc. Natl. Acad. Sci. USA 89:9367-9371) can also be used. Another example of a library that can be used, in which the amide functionalities in peptides have been permethylated to generate a chemically transformed combinatorial library, is described by Ostresh et al. (1994, Proc.
Natl.
Acad. Sci. USA 91:11138-11142).
The variety of non-peptide libraries that are useful in the present invention is great. For example, Ecker and Crooke, 1995, Bio/Technology 13:351-360 list benzodiazepines, hydantoins, piperazinediones, biphenyls, sugar analogs, beta-mercaptoketones, arylacetic acids, acylpiperidines, benzopyrans, cubanes, xanthines, aminimides, and oxazolones as among the chemical species that form the basis of various libraries.
Non-peptide libraries can be classified broadly into two types: decorated monomers and oligomers. Decorated monomer libraries employ a relatively simple scaffold structure upon which a variety functional groups is added. Often the scaffold will be a molecule with a known useful pharmacological activity. For example, the scaffold might be the benzodiazepine structure.
Non-peptide oligomer libraries utilize a large number of monomers that are assembled together in ways that create new shapes that depend on the order of the monomers. Among the monomer units that have been used are carbamates, pyrrolinones, and morpholinos. Peptoids, peptide-like oligomers in which the side chain is attached to the alpha amino group rather than the alpha carbon, form the basis of another version of non-peptide oligomer libraries. The first non-peptide oligomer libraries utilized a single type of monomer and thus contained a repeating backbone.
Recent libraries have utilized more than one monomer, giving the libraries added flexibility.
Screening the libraries can be accomplished by any of a variety of commonly known methods. See, e.g., the following references, which disclose screening of peptide libraries: Parmley and Smith, 1989, Adv. Exp. Med. Biol. 251:215-218;
Scott and Smith, 1990, Science 249:386-390; Fowlkes et al., 1992; BioTechniques 13:422-427; Oldenburg et al., 1992, Proc. Natl. Acad. Sci. USA 89:5393-5397; Yu et al., 1994, Cell 76:933-945; Staudt et al., 1988, Science 241:577-580; Bock et al., 1992, Nature 355:564-566; Tuerk et al., 1992, Proc. Natl. Acad. Sci. USA 89:6988-6992;
Ellington et al., 1992, Nature 355:850-852; U.S. Pat. No. 5,096,815, U.S. Pat.
No.
5,223,409, and U.S. Pat. No. 5,198,346, all to Ladner et al.; Rebar and Pabo, 1993, Science 263:671-673; and CT Publication No. WO 94/18318.
In a specific embodiment, screening to identify a molecule that binds a polypeptide of the invention can be carned out by contacting the library members with a polypeptide of the invention immobilized on a solid phase and harvesting those library members that bind to the polypeptide of the invention. Examples of such screening methods, termed "panning" techniques are described by way of example in Parmley and Smith, 1988, Gene 73:305-318; Fowlkes et al., 1992, BioTechniques 13:422-427; PCT Publication No. WO 94/18318; and in references cited herein.
In another embodiment, the two-hybrid system for selecting interacting proteins in yeast (Fields and Song, 1989, Nature 340:245-246; Chien et al., 1991, Proc. Natl. Acad. Sci. USA 88:9578-9582) can be used to identify molecules that specifically bind to a polypeptide of the invention.
Where the polypeptide of the invention binding molecule is a polypeptide, the polypeptide can be conveniently selected from any peptide library, including random peptide libraries, combinatorial peptide libraries, or biased peptide libraries. The term "biased" is used herein to mean that the method of generating the library is manipulated so as to restrict one or more parameters that govern the diversity of the resulting collection of molecules, in this case peptides.
Thus, a truly random peptide library would generate a collection of peptides in which the probability of finding a particular amino acid at a given position of the peptide is the same for all 20 amino acids. A bias can be introduced into the library, however, by specifying, for example, that a lysine occur every fifth amino acid or that positions 4, 8, and 9 of a decapeptide library be fixed to include only arginine.
Clearly, many types of biases can be contemplated, and the present invention is not restricted to any particular bias. Furthermore, the present invention contemplates specific types of peptide libraries, such as phage displayed peptide libraries and those that utilize a DNA construct comprising a lambda phage vector with a DNA
insert.
As mentioned above, in the case of a polypeptide of the invention binding molecule that is a polypeptide, the polypeptide may have about 6 to less than about 60 amino acid residues, preferably about 6 to about 10 amino acid residues, and most preferably, about 6 to about 22 amino acids. In another embodiment, a polypeptide of the invention binding polypeptide has in the range of 15-100 amino acids, or amino acids.
The selected polypeptide of the invention binding polypeptide can be obtained by chemical synthesis or recombinant expression.
Antisense And Ribozyme (Antagonists) In specific embodiments, antagonists according to the present invention are nucleic acids corresponding to the sequences contained in SEQ ID NO:X, or the complementary strand thereof, and/or to nucleotide sequences contained a deposited clone. In one embodiment, antisense sequence is generated internally by the organism, in another embodiment, the antisense sequence is separately administered (see, for example, O'Connor, Neurochem., 56:560 (1991). Oligodeoxynucleotides as Anitsense Inhibitors of Gene Expression, CRC Press, Boca Raton, FL (1988).
Antisense technology can be used to control gene expression through antisense DNA
or RNA, or through triple-helix formation. Antisense techniques are discussed for example, in Okano, Neurochem., 56:560 (1991); Oligodeoxynucleotides as Antisense Inhibitors of Gene Expression, CRC Press, Boca Raton, FL (1988). Triple helix formation is discussed in, for instance, Lee et al., Nucleic Acids Research, 6:3073 (1979); Cooney et al., Science, 241:456 (1988); and Dervan et al., Science, 251:1300 (1991). The methods are based on binding of a polynucleotide to a complementary DNA or RNA.
For example, the use of c-myc and c-myb antisense RNA constructs to inhibit the growth of the non-lymphocytic leukemia cell line HL-60 and other cell lines was previously described. (Wickstrom et al. (1988); Anfossi et al. (1989)). These experiments were performed in vitro by incubating cells with the oligoribonucleotide.
A similar procedure for in vivo use is described in WO 91/15580. Briefly, a pair of oligonucleotides for a given antisense RNA is produced as follows: A sequence complimentary to the first 15 bases of the open reading frame is flanked by an EcoRl site on the 5 end and a HindIII site on the 3 end. Next, the pair of oligonucleotides is heated at 90°C for one minute and then annealed in 2X ligation buffer (20mM TRIS
HCl pH 7.5, lOmM MgCl2, lOMM dithiothreitol (DTT) and 0.2 mM ATP) and then ligated to the EcoRl/Hind III site of the retroviral vector PMV7 (WO
91/15580).
For example, the 5' coding portion of a polynucleotide that encodes the mature polypeptide of the present invention may be used to design an antisense RNA
oligonucleotide of from about 10 to 40 base pairs in length. A DNA
oligonucleotide is designed to be complementary to a region of the gene involved in transcription thereby preventing transcription and the production of the receptor. The antisense RNA oligonucleotide hybridizes to the mRNA in vivo and blocks translation of the mRNA molecule into receptor polypeptide.
In one embodiment, the antisense nucleic acid of the invention is produced intracellularly by transcription from an exogenous sequence. For example, a vector or a portion thereof, is transcribed, producing an antisense nucleic acid (RNA) of the invention. Such a vector would contain a sequence encoding the antisense nucleic acid of the invention. Such a vector can remain episomal or become chromosomally integrated, as long as it can be transcribed to produce the desired antisense RNA.
Such vectors can be constructed by recombinant DNA technology methods standard in the art. Vectors can be plasmid, viral, or others known in the art, used for replication and expression in vertebrate cells. Expression of the sequence encoding a polypeptide of the invention, or fragments thereof, can be by any promoter known in the art to act in vertebrate, preferably human cells. Such promoters can be inducible or constitutive. Such promoters include, but are not limited to, the SV40 early promoter region (Bernoist and Chambon, Nature, 29:304-310 (1981), the promoter contained in the 3' long terminal repeat of Rous sarcoma virus (Yamamoto et al., Cell, 22:787-797 (1980), the herpes thymidine promoter (Wagner et al., Proc.
Natl.
Acad. Sci. U.S.A., 78:1441-1445 (1981), the regulatory sequences of the metallothionein gene (Brinster et al., Nature, 296:39-42 (1982)), etc.
The antisense nucleic acids of the invention comprise a sequence complementary to at least a portion of an RNA transcript of a gene of interest.
However, absolute complementarity, although preferred, is not required. A
sequence "complementary to at least a portion of an RNA," referred to herein, means a sequence having sufficient complementarity to be able to hybridize with the RNA, forming a stable duplex; in the case of double stranded antisense nucleic acids of the invention, a single strand of the duplex DNA may thus be tested, or triplex formation may be assayed. The ability to hybridize will depend on both the degree of complementarity and the length of the antisense nucleic acid Generally, the larger the hybridizing nucleic acid, the more base mismatches with a RNA sequence of the invention it may contain and still form a stable duplex (or triplex as the case may be).
One skilled in the art can ascertain a tolerable degree of mismatch by use of standard procedures to determine the melting point of the hybridized complex.
Oligonucleotides that are complementary to the 5' end of the message, e.g., the 5' untranslated sequence up to and including the AUG initiation codon, should work most efficiently at inhibiting translation. However, sequences complementary to the 3' untranslated sequences of mRNAs have been shown to be effective at inhibiting translation of mRNAs as well. See generally, Wagner, R., Nature, 372:333-335 (1994). Thus, oligonucleotides complementary to either the 5' - or 3' -1 S non- translated, non-coding regions of a polynucleotide sequence of the invention could be used in an antisense approach to inhibit translation of endogenous mRNA.
Oligonucleotides complementary to the 5' untranslated region of the mRNA
should include the complement of the AUG start codon. Antisense oligonucleotides complementary to mRNA coding regions are less efficient inhibitors of translation but could be used in accordance with the invention. Whether designed to hybridize to the 5' -, 3' - or coding region of mRNA, antisense nucleic acids should be at least six nucleotides in length, and are preferably oligonucleotides ranging from 6 to about SO
nucleotides in length. In specific aspects the oligonucleotide is at least 10 nucleotides, at least 17 nucleotides, at least 25 nucleotides or at least 50 nucleotides.
The polynucleotides of the invention can be DNA or RNA or chimeric mixtures or derivatives or modified versions thereof, single-stranded or double-stranded. The oligonucleotide can be modified at the base moiety, sugar moiety, or phosphate backbone, for example, to improve stability of the molecule, hybridization, etc. The oligonucleotide may include other appended groups such as peptides (e.g., for targeting host cell receptors in vivo), or agents facilitating transport across the cell membrane (see, e.g., Letsinger et al., Proc. Natl. Acad. Sci. U.S.A. 86:6553-(1989); Lemaitre et al., Proc. Natl. Acad. Sci., 84:648-652 (1987); PCT
Publication NO: W088/09810, published December 15, 1988) or the blood-brain barrier (see, e.g., PCT Publication NO: W089/10134, published April 25, 1988), hybridization-triggered cleavage agents. (See, e.g., Krol et al., BioTechniques, 6:958-976 (1988)) or intercalating agents. (See, e.g., Zon, Pharm. Res., 5:539-549 (1988)). To this end, the oligonucleotide may be conjugated to another molecule, e.g., a peptide, hybridization triggered cross-linking agent, transport agent, hybridization-triggered cleavage agent, etc.
The antisense oligonucleotide may comprise at least one modified base moiety which is selected from the group including, but not limited to, 5-fluorouracil, S-bromouracil, 5-chlorouracil, S-iodouracil, hypoxanthine, xantine, 4-acetylcytosine, 5-(carboxyhydroxylmethyl) uracil, 5-carboxymethylaminomethyl-2-thiouridine, S-carboxyrnethylaminomethyluracil, dihydrouracil, beta-D-galactosylqueosine, inosine, N6-isopentenyladenine, 1-methylguanine, 1-methylinosine, 2,2-dimethylguanine, 2-methyladenine, 2-methylguanine, 3-methylcytosine, S-methylcytosine, N6-adenine, 7-methylguanine, 5-methylaminomethyluracil, S-methoxyaminomethyl-2-thiouracil, beta-D-mannosylqueosine, 5'-methoxycarboxymethyluracil, 5-methoxyuracil, 2-methylthio-N6-isopentenyladenine, uracil-5-oxyacetic acid (v), wybutoxosine, pseudouracil, queosine, 2-thiocytosine, 5-methyl-2-thiouracil, 2-thiouracil, 4-thiouracil, S-methyluracil, uracil-5-oxyacetic acid methylester, uracil-S-oxyacetic acid (v), 5-methyl-2-thiouracil, 3-(3-amino-3-N-2-carboxypropyl) uracil, (acp3)w, and 2,6-diaminopurine.
The antisense oligonucleotide may also comprise at least one modified sugar moiety selected from the group including, but not limited to, arabinose, 2-fluoroarabinose, xylulose, and hexose.
In yet another embodiment, the antisense oligonucleotide comprises at least one modified phosphate backbone selected from the group including, but not limited to, a phosphorothioate, a phosphorodithioate, a phosphoramidothioate, a phosphoramidate, a phosphordiamidate, a methylphosphonate, an alkyl phosphotriester, and a formacetal or analog thereof.
In yet another embodiment, the antisense oligonucleotide is an a-anomeric oligonucleotide. An a-anomeric oligonucleotide forms specific double-stranded hybrids with complementary RNA in which, contrary to the usual b-units, the strands run parallel to each other (Gautier et al., Nucl. Acids Res., 15:6625-6641 (1987)).
The oligonucleotide is a 2-0-methylribonucleotide (moue et al., Nucl. Acids Res., 15:6131-6148 (1987)), or a chimeric RNA-DNA analogue (moue et al., FEBS Lett.
S 215:327-330 (1987)).
Polynucleotides of the invention may be synthesized by standard methods known in the art, e.g. by use of an automated DNA synthesizer (such as are commercially available from Biosearch, Applied Biosystems, etc.). As examples, phosphorothioate oligonucleotides may be synthesized by the method of Stein et al.
(Nucl. Acids Res., 16:3209 (1988)), methylphosphonate oligonucleotides can be prepared by use of controlled pore glass polymer supports (Sarin et al., Proc.
Natl.
Acad. Sci. U.S.A., 85:7448-7451 (1988)), etc.
While antisense nucleotides complementary to the coding region sequence of the invention could be used, those complementary to the transcribed untranslated 1 S region are most preferred.
Potential antagonists according to the invention also include catalytic RNA, or a ribozyme (See, e.g., PCT International Publication WO 90/11364, published October 4, 1990; Sarver et al, Science, 247:1222-1225 (1990). While ribozymes that cleave mRNA at site specific recognition sequences can be used to destroy mRNAs corresponding to the polynucleotides of the invention, the use of hammerhead ribozymes is preferred. Hammerhead ribozymes cleave mRNAs at locations dictated by flanking regions that form complementary base pairs with the target mRNA.
The sole requirement is that the target mRNA have the following sequence of two bases:
5' -UG-3' . The construction and production of hammerhead ribozymes is well known in the art and is described more fully in Haseloff and Gerlach, Nature, 334:585-591 (1988). There are numerous potential hammerhead ribozyme cleavage sites within each nucleotide sequence disclosed in the sequence listing.
Preferably, the ribozyme is engineered so that the cleavage recognition site is located near the 5' end of the mRNA corresponding to the polynucleotides of the invention; i.e., to increase efficiency and minimize the intracellular accumulation of non-functional mRNA transcripts.

As in the antisense approach, the ribozymes of the invention can be composed of modified oligonucleotides (e.g. for improved stability, targeting, etc.) and should be delivered to cells which express the polynucleotides of the invention in vivo.
DNA constructs encoding the ribozyme may be introduced into the cell in the same manner as described above for the introduction of antisense encoding DNA. A
preferred method of delivery involves using a DNA construct "encoding" the ribozyme under the control of a strong constitutive promoter, such as, for example, pol III or pol II promoter, so that transfected cells will produce sufficient quantities of the ribozyme to destroy endogenous messages and inhibit translation. Since ribozymes unlike antisense molecules, are catalytic, a lower intracellular concentration is required for efficiency.
Antagonist/agonist compounds may be employed to inhibit the cell growth and proliferation effects of the polypeptides of the present invention on neoplastic cells and tissues, i.e. stimulation of angiogenesis of tumors, and, therefore, retard or prevent abnormal cellular growth and proliferation, for example, in tumor formation or growth.
The antagonist/agonist may also be employed to prevent hyper-vascular diseases, and prevent the proliferation of epithelial lens cells after extracapsular cataract surgery. Prevention of the mitogenic activity of the polypeptides of the present invention may also be desirous in cases such as restenosis after balloon angioplasty.
The antagonist/agonist may also be employed to prevent the growth of scar tissue during wound healing.
The antagonist/agonist may also be employed to treat, prevent, and/or diagnose the diseases described herein.
Thus, the invention provides a method of treating or preventing diseases, disorders, and/or conditions, including but not limited to the diseases, disorders, and/or conditions listed throughout this application, associated with overexpression of a polynucleotide of the present invention by administering to a patient (a) an antisense molecule directed to the polynucleotide of the present invention, and/or (b) a ribozyme directed to the polynucleotide of the present invention.

invention, and/or (b) a ribozyme directed to the polynucleotide of the present invention Other Activities The polypeptide of the present invention, as a result of the ability to stimulate vascular endothelial cell growth, may be employed in treatment for stimulating re vascularization of ischemic tissues due to various disease conditions such as thrombosis, arteriosclerosis, and other cardiovascular conditions. These polypeptide may also be employed to stimulate angiogenesis and limb regeneration, as discussed above.
The polypeptide may also be employed for treating wounds due to injuries, burns, post-operative tissue repair, and ulcers since they are mitogenic to various cells of different origins, such as fibroblast cells and skeletal muscle cells, and therefore, facilitate the repair or replacement of damaged or diseased tissue.
The polypeptide of the present invention may also be employed stimulate neuronal growth and to treat, prevent, and/or diagnose neuronal damage which occurs in certain neuronal disorders or neuro-degenerative conditions such as Alzheimer's disease, Parkinson's disease, and A>DS-related complex. The polypeptide of the invention may have the ability to stimulate chondrocyte growth, therefore, they may be employed to enhance bone and periodontal regeneration and aid in tissue transplants or bone grafts.
The polypeptide of the present invention may be also be employed to prevent skin aging due to sunburn by stimulating keratinocyte growth.
The polypeptide of the invention may also be employed for preventing hair loss, since FGF family members activate hair-forming cells and promotes melanocyte growth. Along the same lines, the polypeptides of the present invention may be employed to stimulate growth and differentiation of hematopoietic cells and bone marrow cells when used in combination with other cytokines.
The polypeptide of the invention may also be employed to maintain organs before transplantation or for supporting cell culture of primary tissues.
The polypeptide of the present invention may also be employed for inducing tissue of mesodermal origin to differentiate in early embryos.

The polypeptide or polynucleotides and/or agonist or antagonists of the present invention may also increase or decrease the differentiation or proliferation of embryonic stem cells, besides, as discussed above, hematopoietic lineage.
The polypeptide or polynucleotides and/or agonist or antagonists of the present invention may also be used to modulate mammalian characteristics, such as body height, weight, hair color, eye color, skin, percentage of adipose tissue, pigmentation, size, and shape (e.g., cosmetic surgery). Similarly, polypeptides or polynucleotides and/or agonist or antagonists of the present invention may be used to modulate mammalian metabolism affecting catabolism, anabolism, processing, utilization, and storage of energy.
Polypeptide or polynucleotides and/or agonist or antagonists of the present invention may be used to change a mammal's mental state or physical state by influencing biorhythms, caricadic rhythms, depression (including depressive diseases, disorders, and/or conditions), tendency for violence, tolerance for pain, reproductive capabilities (preferably by Activin or Inhibin-like activity), hormonal or endocrine levels, appetite, libido, memory, stress, or other cognitive qualities.
Polypeptide or polynucleotides and/or agonist or antagonists of the present invention may also be used as a food additive or preservative, such as to increase or decrease storage capabilities, fat content, lipid, protein, carbohydrate, vitamins, minerals, cofactors or other nutritional components.
Other Preferred Embodiments Other preferred embodiments of the claimed invention include an isolated nucleic acid molecule comprising a nucleotide sequence which is at least 95%
identical to a sequence of at least about 50 contiguous nucleotides in the nucleotide sequence of SEQ ID NO:X wherein X is any integer as defined in Table 1.
Also preferred is a nucleic acid molecule wherein said sequence of contiguous nucleotides is included in the nucleotide sequence of SEQ ID NO:X in the range of positions beginning with the nucleotide at about the position of the 5' Nucleotide of the Clone Sequence and ending with the nucleotide at about the position of the 3' Nucleotide of the Clone Sequence as defined for SEQ ID NO:X in Table 1.
Also preferred is a nucleic acid molecule wherein said sequence of contiguous nucleotides is included in the nucleotide sequence of SEQ ID NO:X in the range of positions beginning with the nucleotide at about the position of the 5' Nucleotide of the Start Codon and ending with the nucleotide at about the position of the 3' Nucleotide of the Clone Sequence as defined for SEQ ID NO:X in Table 1.
Similarly preferred is a nucleic acid molecule wherein said sequence of contiguous nucleotides is included in the nucleotide sequence of SEQ LD NO:X
in the range of positions beginning with the nucleotide at about the position of the 5' Nucleotide of the First Amino Acid of the Signal Peptide and ending with the nucleotide at about the position of the 3' Nucleotide of the Clone Sequence as defined for SEQ ID NO:X in Table 1.
Also preferred is an isolated nucleic acid molecule comprising a nucleotide sequence which is at least 95% identical to a sequence of at least about 150 contiguous nucleotides in the nucleotide sequence of SEQ ID NO:X.
Further preferred is an isolated nucleic acid molecule comprising a nucleotide.
sequence which is at least 95% identical to a sequence of at least about S00 contiguous nucleotides in the nucleotide sequence of SEQ ID NO:X.
A further preferred embodiment is a nucleic acid molecule comprising a nucleotide sequence which is at least 95% identical to the nucleotide sequence of SEQ
117 NO:X beginning with the nucleotide at about the position of the 5' Nucleotide of the First Amino Acid of the Signal Peptide and ending with the nucleotide at about the position of the 3' Nucleotide of the Clone Sequence as defined for SEQ ID
NO:X
in Table 1.
A further preferred embodiment is an isolated nucleic acid molecule comprising a nucleotide sequence which is at least 95% identical to the complete nucleotide sequence of SEQ ID NO:X.
Also preferred is an isolated nucleic acid molecule which hybridizes under stringent hybridization conditions to a nucleic acid molecule, wherein said nucleic acid molecule which hybridizes does not hybridize under stringent hybridization conditions to a nucleic acid molecule having a nucleotide sequence consisting of only A residues or of only T residues.
Also preferred is a composition of matter comprising a DNA molecule which comprises a human cDNA clone identified by a cDNA Clone Identifier in Table 1, S which DNA molecule is contained in the material deposited with the American Type Culture Collection and given the ATCC Deposit Number shown in Table 1 for said cDNA Clone Identifier.
Also preferred is an isolated nucleic acid molecule comprising a nucleotide sequence which is at least 95% identical to a sequence of at least 50 contiguous nucleotides in the nucleotide sequence of a human cDNA clone identified by a cDNA
Clone Identifier in Table 1, which DNA molecule is contained in the deposit given the ATCC Deposit Number shown in Table 1.
Also preferred is an isolated nucleic acid molecule, wherein said sequence of at least 50 contiguous nucleotides is included in the nucleotide sequence of the complete open reading frame sequence encoded by said human cDNA clone.
Also preferred is an isolated nucleic acid molecule comprising a nucleotide sequence which is at least 95% identical to sequence of at least 150 contiguous nucleotides in the nucleotide sequence encoded by said human cDNA clone.
A further preferred embodiment is an isolated nucleic acid molecule comprising a nucleotide sequence which is at least 95% identical to sequence of at least 500 contiguous nucleotides in the nucleotide sequence encoded by said human cDNA clone.
A further preferred embodiment is an isolated nucleic acid molecule comprising a nucleotide sequence which is at least 95% identical to the complete nucleotide sequence encoded by said human cDNA clone.
A further preferred embodiment is a method for detecting in a biological sample a nucleic acid molecule comprising a nucleotide sequence which is at least 95% identical to a sequence of at least 50 contiguous nucleotides in a sequence selected from the group consisting of: a nucleotide sequence of SEQ ID NO:X
wherein X is any integer as defined in Table 1; and a nucleotide sequence encoded by a human cDNA clone identified by a cDNA Clone Identifier in Table 1 and contained in the deposit with the ATCC Deposit Number shown for said cDNA clone in Table l; which method comprises a step of comparing a nucleotide sequence of at least one nucleic acid molecule in said sample with a sequence selected from said group and determining whether the sequence of said nucleic acid molecule in said sample is at least 95% identical to said selected sequence.
Also preferred is the above method wherein said step of comparing sequences comprises determining the extent of nucleic acid hybridization between nucleic acid molecules in said sample and a nucleic acid molecule comprising said sequence selected from said group. Similarly, also preferred is the above method wherein said step of comparing sequences is performed by comparing the nucleotide sequence determined from a nucleic acid molecule in said sample with said sequence selected from said group. The nucleic acid molecules can comprise DNA molecules or RNA
molecules.
A further preferred embodiment is a method for identifying the species, tissue or cell type of a biological sample which method comprises a step of detecting nucleic 1 S acid molecules in said sample, if any, comprising a nucleotide sequence that is at least 95% identical to a sequence of at least 50 contiguous nucleotides in a sequence selected from the group consisting of: a nucleotide sequence of SEQ ID NO:X
wherein X is any integer as defined in Table 1; and a nucleotide sequence encoded by a human cDNA clone identified by a cDNA Clone Identifier in Table 1 and contained in the deposit with the ATCC Deposit Number shown for said cDNA clone in Table 1.
The method for identifying the species, tissue or cell type of a biological sample can comprise a step of detecting nucleic acid molecules comprising a nucleotide sequence in a panel of at least two nucleotide sequences, wherein at least one sequence in said panel is at least 95% identical to a sequence of at least contiguous nucleotides in a sequence selected from said group.
Also preferred is a method for diagnosing in a subject a pathological condition associated with abnormal structure or expression of a gene encoding a secreted protein identified in Table 1, which method comprises a step of detecting in a biological sample obtained from said subject nucleic acid molecules, if any, comprising a nucleotide sequence that is at least 95% identical to a sequence of at least 50 contiguous nucleotides in a sequence selected from the group consisting of: a nucleotide sequence of SEQ ID NO:X wherein X is any integer as defined in Table 1;
and a nucleotide sequence encoded by a human cDNA clone identified by a cDNA
Clone Identifier in Table 1 and contained in the deposit with the ATCC Deposit Number shown for said cDNA clone in Table 1.
The method for diagnosing a pathological condition can comprise a step of detecting nucleic acid molecules comprising a nucleotide sequence in a panel of at least two nucleotide sequences, wherein at least one sequence in said panel is at least 95% identical to a sequence of at least 50 contiguous nucleotides in a sequence selected from said group.
Also preferred is a composition of matter comprising isolated nucleic acid molecules wherein the nucleotide sequences of said nucleic acid molecules comprise a panel of at least two nucleotide sequences, wherein at least one sequence in said panel is at least 95% identical to a sequence of at least 50 contiguous nucleotides in a sequence selected from the group consisting of: a nucleotide sequence of SEQ
ID
NO:X wherein X is any integer as defined in Table 1; and a nucleotide sequence encoded by a human cDNA clone identified by a cDNA Clone Identifier in Table 1 and contained in the deposit with the ATCC Deposit Number shown for said cDNA
clone in Table 1. The nucleic acid molecules can comprise DNA molecules or RNA
molecules.
Also preferred is an isolated polypeptide comprising an amino acid sequence at least 90% identical to a sequence of at least about 10 contiguous amino acids in the amino acid sequence of SEQ ID NO:Y wherein Y is any integer as defined in Table 1.
Also preferred is a polypeptide, wherein said sequence of contiguous amino acids is included in the amino acid sequence of SEQ ID NO:Y in the range of positions beginning with the residue at about the position of the First Amino Acid of the Secreted Portion and ending with the residue at about the Last Amino Acid.
of the Open Reading Frame as set forth for SEQ ID NO:Y in Table 1.
Also preferred is an isolated polypeptide comprising an amino acid sequence at least 95% identical to a sequence of at least about 30 contiguous amino acids in the amino acid sequence of SEQ ID NO:Y.

Further preferred is an isolated polypeptide comprising an amino acid sequence at least 95% identical to a sequence of at least about 100 contiguous amino acids in the amino acid sequence of SEQ ID NO:Y.
Further preferred is an isolated polypeptide comprising an amino acid sequence at least 95% identical to the complete amino acid sequence of SEQ ID
NO:Y.
Further preferred is an isolated polypeptide comprising an amino acid sequence at least 90% identical to a sequence of at least about 10 contiguous amino acids in the complete amino acid sequence of a secreted protein encoded by a human cDNA clone identified by a cDNA Clone Identifier in Table 1 and contained in the deposit with the ATCC Deposit Number shown for said cDNA clone in Table 1.
Also preferred is a polypeptide wherein said sequence of contiguous amino acids is included in the amino acid sequence of a secreted portion of the secreted protein encoded by a human cDNA clone identified by a cDNA Clone Identifier in 1 S Table 1 and contained in the deposit with the ATCC Deposit Number shown for said cDNA clone in Table 1.
Also preferred is an isolated polypeptide comprising an amino acid sequence at least 95% identical to a sequence of at least about 30 contiguous amino acids in the amino acid sequence of the secreted portion of the protein encoded by a human cDNA
clone identified by a cDNA Clone Identifier in Table 1 and contained in the deposit with the ATCC Deposit Number shown for said cDNA clone in Table 1.
Also preferred is an isolated polypeptide comprising an amino acid sequence at least 95% identical to a sequence of at least about 100 contiguous amino acids in the amino acid sequence of the secreted portion of the protein encoded by a human cDNA clone identified by a cDNA Clone Identifier in Table 1 and contained in the deposit with the ATCC Deposit Number shown for said cDNA clone in Table 1.
Also preferred is an isolated polypeptide comprising an amino acid sequence at least 95% identical to the amino acid sequence of the secreted portion of the protein encoded by a human cDNA clone identified by a cDNA Clone Identifier in Table 1 and contained in the deposit with the ATCC Deposit Number shown for said cDNA
clone in Table 1.

Further preferred is an isolated antibody which binds specifically to a polypeptide comprising an amino acid sequence that is at least 90% identical to a sequence of at least 10 contiguous amino acids in a sequence selected from the group consisting o~ an amino acid sequence of SEQ ID NO:Y wherein Y is any integer as defined in Table 1; and a complete amino acid sequence of a protein encoded by a human cDNA clone identified by a cDNA Clone Identifier in Table 1 and contained in the deposit with the ATCC Deposit Number shown for said cDNA clone in Table 1.
Further preferred is a method for detecting in a biological sample a polypeptide comprising an amino acid sequence which is at least 90% identical to a sequence of at least 10 contiguous amino acids in a sequence selected from the group consisting of an amino acid sequence of SEQ ID NO:Y wherein Y is any integer as defined in Table 1; and a complete amino acid sequence of a protein encoded by a human cDNA clone identified by a cDNA Clone Identifier in Table 1 and contained in the deposit with the ATCC Deposit Number shown for said cDNA clone in Table 1; which method comprises a step of comparing an amino acid sequence of at least one polypeptide molecule in said sample with a sequence selected from said group and determining whether the sequence of said polypeptide molecule in said sample is at least 90% identical to said sequence of at least 10 contiguous amino acids.
Also preferred is the above method wherein said step of comparing an amino acid sequence of at least one polypeptide molecule in said sample with a sequence selected from said group comprises determining the extent of specific binding of polypeptides in said sample to an antibody which binds specifically to a polypeptide comprising an amino acid sequence that is at least 90% identical to a sequence of at least 10 contiguous amino acids in a sequence selected from the group consisting o~
an amino acid sequence of SEQ ID NO:Y wherein Y is any integer as defined in Table 1; and a complete amino acid sequence of a protein encoded by a human cDNA
clone identified by a cDNA Clone Identifier in Table 1 and contained in the deposit with the ATCC Deposit Number shown for said cDNA clone in Table 1.
Also preferred is the above method wherein said step of comparing sequences is performed by comparing the amino acid sequence determined from a polypeptide molecule in said sample with said sequence selected from said group.

Also preferred is a method for identifying the species, tissue or cell type of a biological sample which method comprises a step of detecting polypeptide molecules in said sample, if any, comprising an amino acid sequence that is at least 90%
identical to a sequence of at least 10 contiguous amino acids in a sequence selected from the group consisting of: an amino acid sequence of SEQ ID NO:Y wherein Y
is any integer as defined in Table 1; and a complete amino acid sequence of a secreted protein encoded by a human cDNA clone identified by a cDNA Clone Identifier in Table 1 and contained in the deposit with the ATCC Deposit Number shown for said cDNA clone in Table 1.
Also preferred is the above method for identifying the species, tissue or cell type of a biological sample, which method comprises a step of detecting polypeptide molecules comprising an amino acid sequence in a panel of at least two amino acid sequences, wherein at least one sequence in said panel is at least 90%
identical to a sequence of at least 10 contiguous amino acids in a sequence selected from the above group.
Also preferred is a method for diagnosing in a subject a pathological condition associated with abnormal structure or expression of a gene encoding a secreted protein identified in Table 1, which method comprises a step of detecting in a biological sample obtained from said subject polypeptide molecules comprising an amino acid sequence in a panel of at least two amino acid sequences, wherein at least one sequence in said panel is at least 90% identical to a sequence of at least contiguous amino acids in a sequence selected from the group consisting of an amino acid sequence of SEQ ID NO:Y wherein Y is any integer as defined in Table 1;
and a complete amino acid sequence of a secreted protein encoded by a human cDNA
clone identified by a cDNA Clone Identifier in Table 1 and contained in the deposit with the ATCC Deposit Number shown for said cDNA clone in Table 1.
In any of these methods, the step of detecting said polypeptide molecules includes using an antibody.
Also preferred is an isolated nucleic acid molecule comprising a nucleotide sequence which is at least 95% identical to a nucleotide sequence encoding a polypeptide wherein said polypeptide comprises an amino acid sequence that is at least 90% identical to a sequence of at least 10 contiguous amino acids in a sequence selected from the group consisting of: an amino acid sequence of SEQ ID NO:Y
wherein Y is any integer as defined in Table 1; and a complete amino acid sequence of a secreted protein encoded by a human cDNA clone identified by a cDNA Clone Identifier in Table 1 and contained in the deposit with the ATCC Deposit Number S shown for said cDNA clone in Table 1.
Also preferred is an isolated nucleic acid molecule, wherein said nucleotide sequence encoding a polypeptide has been optimized for expression of said polypeptide in a prokaryotic host.
Also preferred is an isolated nucleic acid molecule, wherein said polypeptide comprises an amino acid sequence selected from the group consisting of: an amino acid sequence of SEQ ID NO:Y wherein Y is any integer as defined in Table 1;
and a complete amino acid sequence of a secreted protein encoded by a human cDNA
clone identified by a cDNA Clone Identifier in Table 1 and contained in the deposit with the ATCC Deposit Number shown for said cDNA clone in Table 1.
Further preferred is a method of making a recombinant vector comprising inserting any of the above isolated nucleic acid molecule into a vector. Also preferred is the recombinant vector produced by this method. Also preferred is a method of making a recombinant host cell comprising introducing the vector into a host cell, as well as the recombinant host cell produced by this method.
Also preferred is a method of making an isolated polypeptide comprising culturing this recombinant host cell under conditions such that said polypeptide is expressed and recovering said polypeptide. Also preferred is this method of making an isolated polypeptide, wherein said recombinant host cell is a eukaryotic cell and said polypeptide is a secreted portion of a human secreted protein comprising an amino acid sequence selected from the group consisting of: an amino acid sequence of SEQ )D NO:Y beginning with the residue at the position of the First Amino Acid of the Secreted Portion of SEQ ID NO:Y wherein Y is an integer set forth in Table 1 and said position of the First Amino Acid of the Secreted Portion of SEQ ID NO:Y
is defined in Table 1; and an amino acid sequence of a secreted portion of a protein encoded by a human cDNA clone identified by a cDNA Clone Identifier in Table 1 and contained in the deposit with the ATCC Deposit Number shown for said cDNA
clone in Table 1. The isolated polypeptide produced by this method is also preferred.

Also preferred is a method of treatment of an individual in need of an increased level of a secreted protein activity, which method comprises administering to such an individual a pharmaceutical composition comprising an amount of an isolated polypeptide, polynucleotide, or antibody of the claimed invention effective to increase the level of said protein activity in said individual.
The above-recited applications have uses in a wide variety of hosts. Such hosts include, but are not limited to, human, m \ \e, rabbit, goat, guinea pig, camel, horse, mouse, rat, hamster, pig, micro-pig, chicken, goat, cow, sheep, dog, cat, non-human primate, and human. In specific embodiments, the host is a mouse, rabbit, goat, guinea pig, chicken, rat, hamster, pig, sheep, dog or cat. In preferred embodiments, the host is a mammal. In most preferred embodiments, the host is a human.
In specific embodiments of the invention, for each "Contig ID" listed in the fourth column of Table S, preferably excluded are one or more polynucleotides comprising, or alternatively consisting of, a nucleotide sequence referenced in the fifth column of Table S and described by the general formula of a-b, whereas a and b are uniquely determined for the corresponding SEQ ID NO:X referred to in column 3:
of Table 5. Further specific embodiments are directed to polynucleotide sequences excluding one, two, three, four, or more of the specific polynucleotide sequences referred to in the fifth column of Table 5. In no way is this listing meant to encompass all of the sequences which may be excluded by the general formula, it is just a representative example. All references available through these accessions are hereby incorporated by reference in their entirety.

Gene cDNA CloneNT Contig Public Accession Numbers ID

No. ID SEQ

ID

NO:

X

1 HSICQ15 11 1044613 T72762, T72891, T85361, T85459, N94575, W30865, AA001748, AA001818, AA035092, AA035091, AA588811, AA614723, AA766000, AA806770, AA827880, AA857415, AA643901, AA649144, AA283120, AA402880, AA456589, AA703179, 219869, AA918225, D20926, D45688, AI272999, AI350879, AI358117, AI434378, AI500055, AI624383, AI650392, AI656664, AI659886, AI253545, and AI272334.

1 HSICQ15 30 897684 AI862559, AW237168, AA827880, AA001818, AI926952, AI692205, AA703179, AI924406, AI971032, AA649144, AA919135, AI332587, AI624383, AI394424, AI806111, AW139416, AA035091, AA588811, AI358117, AI253545, AA001748, AW 177634, N94575, AA806770, AI350879, AA857415, AA402880, AW304315, AA035092, AW028905, D52736, AA614723, T72891, AI659886, AA643901, AA766000, AI272999, T72762, AA456589, AI272334, T85459, W30865, AA918225, AA343076, T85361, AA294984, AW263985, AA283120, AW 169654, AW014329, 219869, AI500055, AI825434, AI689134, AI650392, AI924675, D20926, D45688, AI434378, C14331, D80022, C14389, D80391, D59787, D80024, C14429, D80196, D80164, D50995, D80043, D58283, D80166, D80195, D59859, D51423, D59619, D80210, D80240, D59502, D80188, D59467, D81030, D51799, D59275, D80253, D80227, D59927, AA305409, D80212, D80269, D80366, D80219, C15076, C14014, D57483, D80038, D80193, D50979, D51060, D59889, D59610, D80378, D80045, D51022, D80241, T03269, AW 178893, AW 179328, AW 177440, C75259, D80248, AA305578, D81026, AA514186, D80133, D80522, AA514188, AW378532, AW360811, AW178775, D80251, D80247, AW 178762, AA402847, AA809122, D80134, AW377671, AW375405, D51250, D80168, C05695, D80268, D80439, AW 177505, D80132, AI557751, AW 177501, AW 177511, D59695, AW378540, AW352158, AW366296, AW360844, D80302, AW360817, D52291, AW375406, AW378534, AW 179332, AW377672, D58253, AW179023, AW178905, AW352117, AW 176467, D80949, D59373, AW 178906, AW 177731, AW352171, AW377676, AI910186, AW352170, AI905856, AW 178907, AW 179019, AW179024, D51103, AW360841, D80157, AW 179020, AW 178909, AW
177456, AW 179329, AW360834, AW 178980, AW 177733, AW378528, AW 178908, AW 178754, AW 179018, AW352163, D51759, AW177722, C06015, AW352120, AW179004, AW367967, AW 179012, AW 177728, AW178914, AW178774, AW378543, AW378525, T48593, C14344, D59653, AW 179009, AW 178781, AW
178911, AI535686, D45260, AI525923, D51097, C03092, H67866, H67854, T11417, D81111, C 14407, F 13647, C 14227, D59627, AW367950, AA299583, D80258, C14077, D51221, C14973, T03116, D58246, AI525917, D60214, D59503, AL137694, A74398, AF090883, A84916, A62298, A62300, AR018138, AJ132110, Y17188, AB028859, AR008278, AF058696, X67155, A82595, D26022, A25909, X82626, A67220, D89785, A78862, D34614, A30438, 282022, D88547, Y12724, AR060385, AB002449, A94995, AR025207, AR054175, Y17187, AR008443, U46128, AR016691, AR016690, I50126, I50132, I50128, I50133, AR008277, AR008281, AR066488, AR016514, AR060138, A45456, A26615, AR052274, AB012117, X68127, Y09669, A43192, A43190, AR038669, I14842, AR066487, X64588, A63261, A63887, A85396, AR066482, AR066490, A44171, D50010, A85477, I19525, A86792, I18367, AR062872, A70867, X93549, AR008408, A64136, A68321, I79511, D13509, D88507, AR060133, AF123263, and AI656664.

2 HWLEZ82 12 1056336 AI640262, AI691066, AI215163, AI762251, AW449055, AI867211, AI222536, AI640454, AI222484, AA911069, AA910552, AA994834, AI948684, AI762130, AA377131, AI942346, N64302, N58101, AW268261, AW148738, AI345677, AI619581, AA807088, AW084097, AW 193872, AI697099, AI890806, AI345253, AA070777, AI368816, AI494201, AI745485, AI357599, AW078729, AW020419, AI560625, AI636619, AA555145, AI349933, AI345608, AI310606, AI799195, AA659314, AA811406, AI620093, AW058233, AI345471, AI819970, AI955906, AI805769, AW073898, AI887775, AI582871, AI805688, AI249946, AI801556, AI400725, AI348847, AI343091, AI335363, AI623736, AI345527, AI223980, AI608805, AI560679, AI627714, AW022102, AI539153, N75771, AI784214, AI801404, AI349814, AI252789, AW084056, AW079432, AI050666, AI953765, AI539771, F26535, AI612875, AI873638, AI537643, AI343059, AI608711, AI249877, AI924621, AI888621, AW079334, AI922110, AA806719, AL138386, AI499104, AI669639, 840432, AW 192375, AW006302, AI560545, AI540606, AI915295, AI922665, AI680539, AW191844, AI696626, AW068845, AI589428, AI366968, AI950664, AI886055, AI521596, AL041115, AI366959, AW 168828, AA848053, AW089006, AW 183621, AA738052, AI912474, AW081383, AI539781, AI573026, AI648699, AI539707, AW025279, AI251458, AI379711, AI251830, AW075642, AI570872, AL120854, AW172723, AI366992, AI439020, AI349276, AI918554, AI570966, AI472566, AI282652, AI539089, AW265004, AW085373, AI866608, AW082600, AW168031, AI611743, AW263979, AI349269, AW083804, AW022636, AA830821, N71180, AI951222, AI349246, AI589993, AW149876, AI267185, AI365256, AI446405, AI419440, AI345735, AW085786, AI929108, AL046463, AW 118508, AI472536, AW082623, AI333638, AI677797, AI421087, W46547, AI874151, AI345737, AI348897, AI307210, AI470293, AW130398, AI886594, AI925404, AI244380, AW051048, AW059713, AA835966, AI345736, AI307494, AI310575, AI313320, AI564188, AI336495, AI434242, AI636788, AI313352, AI250627, AL036652, AI446373, AI340627, AI349622, AI307736, AW021373, AI953438, AI446124, AA514684, AW 189933, AI445620, AI334893, AI452857, AI752007, AI699857, AI537677, AI340533, AI683939, AI345370, AI348969, AI583578, AI690472, AI312146, AW409775, N98606, AI312339, AI866786, AI922577, AI352184, AI348854, AI345258, AI348995, AA579232, AI310571, AI553669, N74355, AW022682, AL040694, AW403717, AI589267, AI311604, N29398, AI312428, AI922216, AW022093, AI784028, AI538881, AI689614, AI343131, AW 168700, AI370392, AI345396, AI680498, AI242736, AI345347, AI567582, AI828574, AI866461, AI866082, U77594, E03349, AF061573, E03348, AL133077, U92068, U87620, A18777, AL137665, AF151109, S69510, AL049283, AR011880, AF114168, 579832, AF022363, I48978, I89947, AL050277, A08913, S77771, AL117394, I89931, L13297, A08912, A08910, A08911, I49625, A08909, AL133098, L40363, M86826, AL117629, AL122121, AR038854, A08907, A57389, A08908, AL117585, S76508, I89934, AC003032, X96540, AF067420, AF030165, I42402, AL137705, I29004, X66417, AF 120268, AL080060, AF200464, U68233, I92592, AF104032, Y16258, Y16257, E02756, Y16256, I09360, AF097996, Y14314, AL050146, AL050092, AF081197, AF081195, AF113691, AL022170, AL137556, AL137574, AR068182, AF085809, AR034821, I68732, AF035161, D55641, AL050280, X52128, AF113699, E08631, AF118064, X60786, AL117626, AF162782, A08916, AF094480, AL137660, AF137367, I89944, AL137429, X66871, AF090886, AL137712, U66274, AL080137, AF180525, AF117959, E15582, AB026995, Y10936, AF051325, AR059958, X84990, AB007812, AF016271, AL137300, AF111112, AJ001838, AL133067, X66862, AF118070, U67958, AL049460, AL049314, AL137538, U42031, U96683, AF205861, X59414, AF125949, U57352, 568736, AF090896, X72387, AL110224, U75370, AL137521, AJ006417, AF113013, I00734, AF089818, AF114818, AB019565, AF078844, AR068751, AF119337, AF012536, U00763, AL133104, E02253, AF065135, AR019470, AL137656, X62580, 272491, AL133081, AL122111, AL080127, X57084, E00617, E00717, E00778, AB029065, X62773, A93016, AL137539, Y10655, AF162270, 575997, AF113694, AL049464, AF028823, AL137283, AL049339, E04233, AL137478, AF118090, AF115392, AF106827, AJ242859, I17767, A27171, AF061795, AL050149, AF151685, AF015958, AL137658, U72621, AL133031, A03736, AF182215, X83508, AL137476, AC006112, D83989, X76228, AL133054, AF022813, AF036941, Y11587, AL049430, AF132676, AL137523, AF061836, M30514, AL080110, E01812, X99257, X92070, AL137547, AL133014, AL137527, A12297, AL133606, X63574, AL137480, U91329, X72889, AL096744, E01314, S61953, AL080086, AL122049, AL133080, AL122050, E12579, AB025103, AF176651, AF076633, AL049466, ALI 10221, AF199027, A90832, AF030513, M92439, AC004227, I41145, AF067790, AF113689, AL137526, AL137557, A65340, X87582, E05822, AF215669, AR000496, AL049382, U39656, AL122106, AF111851, AF143957, AR038969, L30117, X06146, A94751, X93495, X55446, AF109155, AL137711, and E07108.

2 HWLEZ82 32 906027 AW247289, AI808090, AW250783, N47945, AA861657, N77555, AA084165, AI038716, AW246922, AI040877, D60694, AI023144, AA694591, AA084138, AL120454, AW023630, AI984284, AA199586, N49154, and AJ001454.

2 HWLEZ82 33 896895 AI640262, A1691066, AI215163, AI762251, AI867211, AI222536, AI640454, AI222484, AA910552, AA911069, AA994834, AI948684, AI762130, AI942346, N64302, N58101, AW449055, AW268261, AW 148738, AI345677, AI619581, AI368816, AI499104, AI697099, AW084097, AI221076, AI348847, AI345253, AA807088, AI310606, AI345527, AW193872, AI494201, AI357599, AI223980, AI745485, AI815239, AW020419, AI560625, AI829990, AA555145, AI801404, AI912474, AI379711, AA811406, AI635158, AI345471, AI805769, AW009624, AI887775, AI582871, AI819970, AW 168828, AI924621, AI249946, AA659314, AI343091, AI335363, AA806160, AI623736, AI680539, AI801556, AI926593, AW022102, N75771, AI784214, AA575874, AI758694, AW004595, AI608805, AI400725, AI203903, AI349814, AW404239, AW078729, AI885943, AW079432, AW074365, AI050666, F26535, AI636619, AI401699, AI702540, AI627714, AI612875, AI888621, AW090238, AI866419, AW079334, AI252789, AA836606, 840432, AI932739, AI608711, AI452857, AI560545, AI540606, AI611728, AI915295, AW 191844, AI696626, AW068845, AI922665, AI589428, AW192375, AI609195, AW006302, AI886055, AI890806, AI640370, AI366959, AA848053, AI521596, AI868200, AW081383, AA837430, AI335476, AI539781, AI539707, AI648699, AA738052, AA693331, AA743274, AL041115, AW 183621, AI500113, AI241799, AI805688, AI694190, AW 172723, AW 150351, AI285514, AW 157250, AI469516, AW055065, AI366992, AI287476, AI401193, AI345608, AL120854, AI922110, AI349276, AI918554, AI344931, AI679261, AW148882, AW075642, AI570872, AW265004, AI801175, AI225248, AI336512, AW082600, AI439020, AI539089, AI349269, AI625464, N71180, AI285417, AW020381, AI570800, AI349246, AI589993, AW 149876, AI267185, AI365256, AI630947, AW073710, AI446405, AW005772, AW085786, N22406, AW263979, AI537837, AI472536, AI360195, AI584118, AI251458, AI677797, AW 148303, W46547, AI349933, AI333638, AI421087, AI244380, AI344935, AW059713, AA835966, AI345736, AI307494, AW089293, AW073843, AW082066, AI310575, AI313320, AI247298, AI336495, AI383804, AI434242, AI636788, AI313352, AI250627, AI564188, AI310582, AI307503, AA207067, AI340627, AW 130398, AI307736, AI349622, AA504514, AW021373, AI446124, AA514684, AW 189933, AI334893, AI340533, AI349266, AI334452, AI349787, AI865289, AI345370, AI348969, AI340653, AI583578, AW089936, AW073898, AA665587, AW074702, AI312146, N98606, AI476147, AI312339, AI309431, AI340537, AI866786, AA838230, AI700164, AI953438, AI610318, AI348854, AI345258, AI340552, AI434760, AW058233, AI348995, AA729782, U77594, AF054831, E12579, E03349, E12580, AF102166, AL049283, AF114168, AF117959, U92068, AF093119, AL133098, E03348, AL133099, U87620, X56530, A18777, AL137665, 569510, AF151109, M64936, AR011880, AF030635, A94751, S79832, AF022363, AF035161, Y14314, AL050277, AF162782, L13297, 577771, AL117394, AF130470, D00174, L40363, M86826, I73428, AL122121, AL117629, E02756, Y16256, A57389, ALl 17585, AR068182, AC003032, Y11435, AF180525, AF067420, AF030165, I42402, U51124, AL137705, AB026995, AC009286, I29004, X66417, AR005195, AF120268, E15568, AL137659, AL080060, AF200464, X52128, U68233, I92592, AL080129, AF085809, AF104032, AL035407, Y16258, Y16257, I09360, AF097996, A90844, AL050092, AF081197, AF081195, AC004686, AL022170, AF113691, A27171, U75378, A92311, AL137556, AF110417, AL137574, E04257, AF106934, AC007390, D55641, AF094480, AF113699, X57084, E08631, AP000130, AP000208, AP000247, AC006039, AF026008, AFI 18064, X60786, ALI 17626, AL137660, AF137367, AF200416, AF148129, AL137429, X61399, AF089818, AL049423, I48978, AF090886, AL137712, U66274, AL080137, AF029750, AB028451, Y10936, AF051325, AR059958, X84990, AB007812, AF016271, AJ012582, U58653, X93495, AC005353, AC005091, AL034417, AC008014, AC006112, AL137300, AF111112, A32826, A30330, A32827, A30331, AL133559, E15582, AJ001838, AL133067, AF131773, X66862, U70981, U89906, AF118070, U67958, AF109906, AL049314, AF131821, AL137538, U42031, U96683, AL049460, AF205861, AF125949, U57352, S68736, AF090896, Y14634, AL110224, U75370, AF040723, U36585, AJ006417, AL137268, X62773, AF016628, X59414, AC005723, L78810, AF113013, X72387, I00734, AB019565, AF078844, AF119337, U89295, AF012536, AL133104, L10353, AR068751, E02253, AL133636, X99257, AF065135, AR019470, X62580, 272491, AL133081, AL122111, AL133053, AF114818, AL080127, E00617, E00717, E00778, M27260, AC004837, AL110228, AL096728, AF047443, AB029065, A93016, and U00763.

3 HISEN93 13 911934 AI827230, AA152023, AI346417, N48299, W84430, AI304351, AI334148, W72005, AW338914, AI127763, AI922951, AI077680, AI191873, AI138802, AI568416, AA152097, AA358674, AW 138291, AI589070, AI088745, AI271659, AA622339, AA724257, AI144150, AW418696, AI401749, AI375884, AI880453, AW 151953, AI619746, AI225114, AI801268, AI419063, AI554474, AI093946, AA302424, AI803839, AI611143, H10756, AI699306, H39784, AA733204, AA496797, AI991589, AI361531, H44939, AI123010, AW169313, AA232382, AA278226, H10757, AI081626, AA513457, AW384209, AA297418, 817581, AA127464, AA278720, T07824, 842918, 830801, AA361091, AW235771, W76603, AI499891, AA370719, T34425, AA358448, AI686346, AA326110, T32000, 810703, 827637, AA651675, AI681496, AA095560, 810795, AA2339S0, AW382078, 857461, AA228006, C19020, 242382, W32704, C01061, T19926, AW370992, AI801263, and AL110199.

3 HISEN93 34 906792 H10756, AA733204, AA232382, 817581, AW384209, T34425, AA326110, T32000, AA095560, 810795, AW382078, C19020, W76603, 242382, AA358448, AA297418, T19926, 299396, AW392670, U463S1, AL119497, AW384394, AW372827, AL119522, AW363220, AL119457, AL119355, AL119324, AL119319, AL119483, AL036418, AL038837, AL119443, U46349, AL119484, AL119363, AL119391, AL037051, AL036725, AA631969, U46350, U46341, AL119341, AL119335, AL036858, AL134902, AL119396, AL119418, AL119496, U46347, ALI 19444, AL039074, AL036924, AL042984, AL037205, U46346, AL119439, AL119401, AL134538, AL042614, AL043033, AL042965, AL042975, AL038509, AL134536, AL119399, AL134525, U46345, AL042450, AL037085, AI142131, AL043029, AL042551, AL037094, AL043019, AL037526, AL036196, AL042544, AL043011, AL037639, AL134542, AL036190, AL042542, AL037082, AL036767, AL119464, AL038520, AL037077, AL043003, AL036268, AL036774, AL036998, AL038851, AL038447, AL036238, AL036733, AL037027, AL037178, AL037615, AL036719, AL036679, AL036765, AL036191, AR060234, A81671, AR066494, AR023813, AR064707, AR069079, AB026436, and AR054110.

3 HISEN93 35 906791 AI827230, AA152023, AI346417, N48299, bl84430, AI304351, AI334148, W72005, AW338914, AI127763, AI9229S1, AI077680, AI191873, AI138802, AA152097, AI568416, AW 138291, AA358674, AI589070, AI088745, AI271659, AA622339, AA724257, AI144150, AW418696, AI401749, AI375884, AW 151953, AI880453, AI619746, AI225114, AI419063, AI801268, AIS54474, AA302424, AI093946, AI611143, AI803839, AI699306, H39784, H10757, AI991589, AA496797, AI361531, H44939, AI123010, AW169313, AA278226, A1081626, AA513457, AW384209, AA297418, AA127464, AA278720, T07824, 842918, W76603, 830801, AW235771, AA361091, A1499891, AA370719, AA358448, AI686346, 810703, 827637, AA651675, AI681496, AA233950, 810795, 857461, AA228006, AA326110, W32704, C01061, AW370992, AI801263, and AL110199.

HPMGR66 15 926706 AI870969, AI640516, AW406868, AI739668, AI992044, AW328099, N40146, AI815435, AI399682, W31098, AA287199, AW401694, H94757, AA458743, N26914, AI825889, W28375, AA622936, AA251057, F36573, AA425786, AW 136132, AA424896, AI191142, 891711, AI927998, 848085, F30656, T09141, AA284495, N39872, AA514473, AW269371, AW 150630, AI743905, H03820, H88046, AI797604, AA337690, AI623328, 242009, F11282, AI087945, AI816373, AW364488, 865904, 881078, AA211331, AI621024, AA460198, AA211286, AA740753, AA741052, AW009770, AW264967, AL036802, AI611728, AI307477, AW020419, AI800185, AL119836, AA888196, N49165, AI282655, AA746607, AI682106, AI249389, AA743430, AI560545, AI609331, AL036150, AW022102, AI678989, AA928539, AW088793, AI445620, AI590020, AW 195253, AL036736, N42321, AI801523, AA613907, AI313346, AW268072, AI281773, AI345415, AI923989, AI366991, AI621341, AI538885, AI699865, AI679724, AL036240, AI491710, AI364135, F37450, AI955945, AW020693, AI312428, AI436429, AI564716, AL040459, AA736984, AI872910, AI440263, AI889168, AI340603, AW051088, AI912434, AA568405, AW083340, AW075382, AL048323, A1471429, AA575874, AL038069, AL048340, AI340519, AI619820, AI537677, AW055252, AI434731, AI114703, AI349246, AI401697, AL119049, AI474646, AL047344, AA420722, AA555145, AI497733, AL036247, AI349957, AI307210, AI699823, AI301046, AW078574, AI307569, AI927233, AI290677, AI313320, AI955441, AI313352, AI648699, AI349622, N99092, AW008253, AI312146, AI312339, AI345258, AI572676, AI906328, AI539260, AI345005, AI612885, AI866131, AI554444, AI625444, AI311604, AI950664, AI334893, AI815855, AI349269, AI627714, AI335411, N71180, AI890907, AA614183, AI439745, AI886355, N22276, AW071380, AI345608, AI348897, AI584118, AW151451, AL042595, AW020397, AI310575, AI801325, AI335363, AI310582, AA806160, AW059568, AI349937, AW020095, AL037013, AI349186, AI334884, AI307543, AI340533, AI926593, AI254226, AW071412, AI307734, AW021464, AI307708, AI597748, AI312325, AL120854, AI345745, AI096771, AI340659, AL049085, AI345471, AI333104, AA640779, AI334895, AW068845, N75771, AI345612, AI344785, AI334930, AI309443, AI538850, AI312353, AW 162194, AI307520, AI581033, AL045979, AI345416, AI225000, AW080076, AI345739, AI702301, AI868204, AI312143, AI656270, AL036638, AI287476, AI698391, AI818728, AI537643, AI521799, AI349955, AW075093, AL042944, AI494201, AI891102, AI312357, AA127565, AI308032, AI500659, AW 152559, AW079432, AI934035, AL120695, AW059828, AA176980, D42039, AL109672, I48978, AF113694, AF026008, AF195092, X72889, AF082324, AF090943, AF159148, A65341, I48979, AL137479, AL050146, AF112208, AL050278, I30339, I30334, AL117457, I33392, X87582, AF200416, ' Y16258, Y16257, E02756, Y16256, I89947, E01812, A08910, A08909, AL050277, AL035458, X83544, AL133565, A57389, A08908, AF013214, E01614, E13364, AF061943, AF114168, AL137459, A08913, AR038854, AR020905, L13297, S63521, A08912, U68387, AF177401, A08911, Y16645, X79812, AL049430, E08631, AF004162, AL080126, AF057300, AF057299, AR068753, AL137537, E02349, AL080234, AL080127, AL049938, S76508, AF054289, I33391, AF028823, X66871, I00734, I89931, AL122098, E00617, E00717, E00778, AL137476, I49625, AB019565, AF078844, AF118094, AL133640, AL133557, AL133016, AL122093, AL137660, AF026816, AC002564, AL137526, AL049339, AF097996, Y11254, AF118090, AL117583, X52128, 577771, AL096744, AL133113, A08916, A08907, A08456, AF017437, AF118070, A76335, E12579, AP000697, L31396, 568736, AL133568, AL117394, Y09972, U42766, L31397, AL122123, S78214, A65965, A18777, AL049464, X60786, X06146, I17767, AF026124, AL110158, A65943, AF213396, I89934, AF119337, AL133560, AL137627, AB016226, AF119336, AL117626, AF111851, AL050172, E12580, U87620, AF017790, 237987, AL122100, AL137533, AF199027, AF146568, AR034821, AJ006417, AF140224, U49908, AF022813, M19658, AL050280, AF162270, AL049300, A65340, AL049283, E05822, X70685, AF069506, AF098484, AL137538, U96683, AL137529, I09499, AF125948, AF044323, U80742, AF055917, AR068466, AL133031, U91329, AF102578, M79462, AF182215, X98834, AL133665, A58524, A58523, AF091084, AL049324, AL137557, X80340, AF106827, AL122111, AF111849, AL137281, AL050116, AC006112, 583440, AF158248, AJ010277, S79832, I32738, AF139986, AL050393, AF022363, AF032666, 297214, AR011880, AL080124, AL137429, AF113690, AL133558, A76337, Y13350, M92439, AF200464, X75295, E07108, U78525, AJ012755, AR053103, AF003737, AF100931, U00763, AL122050, X61399, I18355, AR029490, AF210052, L30117, AF153205, AL117460, AL080139, I03321, AB008792, AL133606, AL137521, AL122121, D16301, X57961, AB008791, AF113013, I34392, AF118064, AL137478, AF087943, S82852, A91160, AF180525, A91162, X83508, AF081197, AF081195, AL049465, AF113689, AR034830, I96214, and X56039.

HPMGR66 38 897505 AI870969, AI640516, AW406868, AI992044, AI739668, N40146, AI815435, AI399682, AW328099, W31098, AA287199, AW401694, H94757, AA458743, N26914, AI825889, W28375, AA622936, AA251057, F36573, AA425786, AW 136132, AA424896, AI191142, 891711, 848085, AI927998, F30656, T09141, N39872, AA284495, AA514473, AW 150630, AI743905, H03820, H88046, AI797604, AA337690, AW269371, AI623328, 242009, F11282, AI087945, AA211331, AI816373, AW364488, 865904, 881078, AI621024, AA460198, AA211286, AA740753, AA741052, AW009770, AW264967, AL036802, AI343030, AI611728, AI307477, AW020419, AI800185, AL119836, AA888196, N49165, AI282655, AA746607, AI682106, AI249389, AA743430, AI313346, AI560545, AL036736, AI609331, AL036150, AW022102, AI678989, AA928539, AW088793, AI445620, AI590020, AW 195253, N42321, AI801523, AA613907, AW268072, AI281773, AI345415, AI923989, AI366991, AI621341, AI538885, AI699865, AI679724, AL036240, AI491710, AI364135, F37450, AW411235, AI955945, AW020693, AI312428, AI436429, AI564716, AL040459, AA736984, AI872910, AI440263, AI889168, AI340511, AI340603, AW051088, AA127565, AI309431, AI912434, AI307550, AA568405, AW411351, AI310927, AW083340, AI307578, AW075382, AL048323, AI471429, AW411265, AA575874, AI310582, AL048340, AL038069, AI340519, A1619820, AI537677, AW055252, AI434731, AW021464, AI114703, AI349246, AI401697, AA853213, AI474646, AL119049, AL047344, AA420722, AA555145, AI497733, AI349957, AI307210, AI699823, AL036247, AI301046, AW078574, AI307569, AI927233, AI290677, AI313320, AI955441, AI313352, AI648699, AI349622, N99092, AW008253, AI312146, AI312339, AI345258, AI539260, AA100772, AI572676, AI345005, AI906328, AI612885, AA420758, AI866131, AW265004, AI625444, AI311604, AI950664, AI554444, AI334893, AI815855, AI349269, AA853539, AI627714, N71180, AI890907, AI335411, AA614183, AI439745, AI886355, N22276, AW071380, AI345608, AW238688, AI348897, AW020592, AI584118, AW410969, AW 151451, AL042595, AW020397, AI310575, AI335363, AI801325, AW410902, AI312261, AA806160, AW059568, AI349937, AW020095, AL037013, AI349186, AI334884, AI307543, AI340533, AI926593, AW071412, AI254226, AI307734, AI307708, AI597748, AI312325, AL120854, AI345745, AI096771, AI340659, AW409775, AI345471, AI333104, AA640779, AI334895, AW068845, AL049085, N75771, AI345612, AI376474, AI344785, AI334930, AI309443, AI538850, AI312353, AW 162194, AI307520, AW410696, AI581033, AL045979, AW411043, AI345416, AI225000, AW080076, AI345739, AI702301, D42039, AL109672, I48978, AF113694, AF026008, AF195092, X72889, AF082324, AF090943, AF159148, A65341, I48979, AL137479, AF112208, E12579, AL050278, I30339, I30334, AL117457, I33392, X87582, AF200416, Y16258, Y16257, E02756, Y16256, I89947, E01812, A08910, A18777, A08909, AL050277, A08908, X83544, AL035458, AL133565, A57389, AF013214, AL050146, E01614, E13364, AF061943, AF114168, AL137459, A08913, AR038854, AR020905, L13297, S63521, A08912, U68387, AF177401, A08911, Y16645, X79812, AL049430, E08631, AF004162, AL080126, AF057300, AF057299, AR068753, AL137537, AP000697, E02349, AL080234, AL080127, S76508, AL049938, AF054289, I33391, AF028823, X66871, Y11254, S68736, A93016, I00734, I89931, AR068751, AC002564, AL122098, A91160, E00617, E00717, E00778, AL137476, I49625, AB019565, AF078844, AF118094, AF065135, A76337, AL133640, A76335, AL133557, I92592, S77771, AL133016, AL122093, AL137660, AF026816, AL137526, AL049339, AF097996, AF118090, AL117583, X52128, AL096744, AL133113, A08916, A08907, A08456, AF017437, AF118070, L31396, AL133568, AL117394, U42766, L31397, AL122123, Y09972, S78214, A65965, AL049464, X60786, X06146, I17767, AF026124, AL110158, A65943, AF213396, I89934, AL133560, AF119337, AL137627, AB016226, AF119336, AL117626, AF111851, AL050172, E12580, AF017790, 237987, AL122100, AL137533, AF199027, 087620, AF146568, AR034821, AJ006417, AF140224, 049908, M19658, AF022813, AF162270, AL050280, AL049300, A65340, AL049283, E05822, X70685, AF069506, AL137538, 096683, AL137529, I09499, AF125948, AF098484, AF044323, 080742, AR068466, AL133031, 091329, AF102578, M79462, AF182215, X98834, AL133665, AF055917, A58524, A58523, AF091084, AL049324, AL137557, X80340, AF106827, AL122111, AF111849, AL137281, AF043493, AL050116, 583440, AF158248, AJ010277, 579832, I32738, AF139986, AL050393, AC006112, AF022363, AF032666, 297214, AR011880, AL080124, AL137429, AF113690, AL133558, Y13350, AF200464, X75295, E07108, M92439, 078525, AJ012755, AR053103, AF003737, AF100931, 000763, AL122050, X61399, AR029490, AF210052, L30117, AF153205, AL117460, I18355, AL080139, I03321, AB008792, AL133606, AL137521, AL122121, D16301, X57961, AB008791, AF113013, I34392, AF118064, AL137478, AF087943, S82852, AF180525, A91162, X83508, AF081197, and AF081195.

6 HTSEK75 16 1085443 AW242493, AI521709, AI525773, W93033, AI640543, AW271801, AA057558, AI651741, AW 139938, AI708135, AI126211, AI371164, AI654038, AW302215, AI480086, W94365, AA947140, AA423865, AI913648, AI808861, AI684050, N76519, AI459337, AA774295, AA644036, AW070975, AI807293, AI206906, AI765015, AI243281, AA488990, AA625569, W37585, N92011, N69457, AI347792, N81012, AI144532, T64459, H19348, W94474, AI308172, AA724261, AA722607, AA970325, AI434819, AA173902, AA423936, AW051693, AAl 13264, H06985, AA021498, AA552460, AI032182, AA054077, W03287, AI334612, AA736787, AW 105083, AI261450, W37460, 841884, AA515975, N63866, H83472, AI240794, AA021497, H05439, AI435002, H86036, AI289946, AA854625, AA113410, H68008, 817269, W07840, AW 194804, C20615, H18909, AA812404, AA057557, AA325158, H83473, AI478425, H38795, AA642403, H67686, AW389618, AA315372, AL135145, H85900, AA172214, AA355951, W92922, T65731, AA247124, AA381082, AI824557, AW087534, AI624279, AL042628, AI886206, AI554218, AI247193, AW087462, AI340582, AI621209, AI280747, AL119863, AI925156, AI824648, AI491775, AL036146, AI564719, AW268122, AI521244, AI348854, AW302988, AI499263, AI570781, AI335426, AI348777, AI636456, AW269097, AL045266, AW074993, AI349614, AI919345, AA225339, AI686817, AI537515, AL040243, AI349256, AI312152, AL038445, AW075084, AI349937, AI680226, AI348897, AI307708, AI312325, AI269862, AI567993, AL135661, AI307520, AI364788, AL045500, AI569583, AI366992, AL079963, AA494167, AW302992, AA427700, AI274508, AI318569, AI888661, AI677796, AI480118, AI433037, AI612759, AL121286, AW083175, AI349645, AA572758, AI802826, AW161579, AI539687, AL041772, AI521100, AL134999, AI340603, AI554245, AW071417, AI334884, AI433976, AI689420, AI308035, AI433023, AI284509, AL120736, AI873644, AI345347, AW15057$, AI620284, AI340627, AW268220, AL036403, AI334450, AW059837, AI582932, AW089572, AI934035, AI889189, AI349598, AI433157, AI623396, AI866573, AI439087, AI923768, AI312428, AL045620, AI539771, AW103371, AW020693, AI469532, AI499986, AI537677, AW083804, AI288285, AI500659, AI869367, AI801325, AI500523, AW148320, AI567351, AI608936, AW071380, AI343112, AI613017, AL036548, AI284517, AI500706, AI445237, AI491776, AW151138, AW268302, AW301300, AI521560, AI500662, AI343059, AW075207, AL121328, AI815855, AW301409, AA508692, AI633493, 259328, AF152363, AF090896, AF126403, AC002457, AF113013, AF061943, I03321, AL133016, I48979, AL049452, I48978, I89947, A08916, A08913, AL050393, A08910, A08909, AL137459, AL050146, U91329, I89931, AL133565, I49625, AL117583, AF078844, AF113699, S78214, AF017152, AL080124, AJ242859, AL117585, AL049466, U00763, AL117435, AL133557, AF111851, AL050277, AL050116, U35846, AF090943, AL049430, AL133640, X96540, AF158248, AL117394, AL122123, AF017437, Y16645, AL117457, AL133560, AL050108, AF113694, AF113690, AF118094, AL110196, ALI 17460, U42766, AF079765, AF091084, AR011880, AL133113, AL137271, AL133080, AL110221, AF113676, E07361, AF097996, Y11587, Y11254, AL122050, AF125949, AB019565, AL133606, AL080060, AF090934, AF113689, AL137557, AL049300, E07108, AL137550, AJ000937, X70685, AL049314, AL137648, AL137463, AL122093, A93016, AL137538, AJ238278, L31396, AF090901, A03736, L31397, AF113019, X82434, AF113677, A77033, A77035, E02349, AF183393, AF090903, AF125948, U80742, AL122110, X72889, A58524, A58523, AF118064, A65341, AL049464, AL050024, AF118070, I33392, AL049382, 282022, AL080127, AF177401, AL137527, X65873, X63574, AF104032, AL133093, AF113691, AF090900, S68736, AF146568, AL050149, AF106862, AL122121, E03348, AR059958, AL133075, AL110225, AL080137, AF087943, U72620, X84990, AL122098, X93495, AL133072, AL049938, AC006221, AL096744, AL050138, A08912, AL137283, AL049283, AC005341, AL137521, U67958, AL080159, I09360, AF067728, I42402, AF026124, AJ012755, A12297, E15569, AL122049, X98834, AF119337, AL137560, A93350, I26207, AL110197, AF111112, AW472863, and AW611570.

6 HT5EK75 39 896931 AI525773, AA021497, H83472, H86036, AA057557, AA315372, AA355951, AA381082, and 259328.

6 HT5EK75 40 896932 AW242493, AI521709, W93033, AI640543, AW271801, AA057558, AI651741, AW 139938, AI708135, AI126211, AI371164, AI654038, AW302215, AI480086, W94365, AA947140, AA423865, AI913648, AI808861, AI684050, N76519, AI459337, AA774295, AA644036, AW070975, AI807293, AI206906, AI765015, AI243281, AA488990, AA625569, N92011, N69457, W37585, AI347792, N81012, T64459, AI144532, H19348, W94474, AA724261, AI308172, AA722607, AA970325, AI434819, AA173902, AA423936, H06985, AW051693, AA113264, AA021498, AA552460, AI032182, AA054077, W03287, AI334612, AA736787, AW 105083, AI261450, W37460, 841884, AA515975, N63866, AI240794, H05439, AI435002, AI289946, AA854625, AA113410, H68008, 817269, W07840, AW194804, C20615, H18909, AA812404, AA325158, H83473, H38795, AI478425, AA642403, H67686, AW389618, AL135145, H85900, AA172214, W92922, T65731, AA247124, AI525773, AI824557, AW087534, AI624279, AL042628, AI886206, AI554218, AI247193, AW087462, AI340582, AI621209, AI280747, AL119863, AI925156, AI491775, AI824648, AL036146, AI564719, AW268122, AI521244, AI348854, AW302988, AI636456, AI499263, AI570781, AI686817, AI335426, AI348777, AW269097, AL045266, AW074993, AI349614, AI919345, AA225339, AI537515, AL040243, AI349256, AI312152, AL038445, AW075084, AI349937, AI680226, AI348897, AI307708, AI312325, AI269862, AI567993, AL135661, AI307520, AI364788, AL045500, AI569583, AI366992, AL079963, AA494167, AW302992, AA427700, AI274508, AI318569, AI888661, AI677796, AI480118, AI433037, AI612759, AL121286, AW083175, AI349645, AA572758, AI433023, AI802826, AW 161579, AI539687, AL041772, AI521100, AL134999, AI340603, AI554245, AW071417, AI334884, AI433976, AI689420, AI308035, AI284509, AL120736, AI873644, AI345347, AW150578, AI340627, AI620284, AW268220, AL036403, AI334450, AI610086, AW059837, AI582932, AW089572, AI934035, AI889189, AI349598, AI433157, AI623396, AI866573, AI439087, AI923768, AI312428, AL045620, AI539771, AW103371, AI469532, AW020693, AI499986, AI537677, AW083804, AI500659, AI288285, AI869367, AI801325, AI500523, AW 148320, AI567351, AI608936, AL036548, AW071380, AI343112, AI613017, AI284517, AI500706, AI445237, AI491776, AW151138, AW268302, AW301300, AI521560, AI500662, AI343059, AL121328, AW075207, AW301409, AI815855, AA508692, AI633493, AI434256, AI745713, AI345735, AI285735, AI284513, AI800453, AF152363, AF126403, AF090896, AC002457, AF113013, AF061943, I03321, AL133016, I48979, AL049452, I48978, I89947, A08916, A08913, A08910, A08909, AL137459, AL050146, U91329, I89931, AL133565, I49625, AL117583, AF078844, AF113699, S78214, AF017152, AL080124, AJ242859, AL117585, AL049466, U00763, AL117435, AL133557, AF111851, AL050277, X96540, AL050116, U35846, AF090943, AL049430, AL133640, AF158248, AL117394, AL122123, AF017437, Y16645, AL133560, AL050108, AF113694, AF118094, AL110196, AL133113, AL117460, AL133080, U42766, AF079765, AF091084, AR011880, AL137271, AF113690, AL110221, AF113676, AL050393, E07361, AF113689, AF097996, Y11587, Y11254, AL122050, AL049300, AF125949, AB019565, AL133606, AL080060, AF090934, AL137557, E07108, AL137550, AJ000937, X70685, AL049314, AL117457, AL137648, AL137463, AL122093, AL122110, A93016, AL137538, AJ238278, L31396, AF090901, A03736, L31397, AF113019, X82434, AF113677, A77033, A77035, E02349, AF183393, AF090903, AF125948, U80742, X72889, A58524, A58523, AF118064, A65341, AL049464, AL050024, AF118070, I33392, AL049382, 282022, AL080127, AF177401, AL137527, X65873, X63574, AF104032, AL133093, AF113691, AF090900, S68736, AF146568, AF106862, AL050149, AL122121, E03348, AR059958, AL133075, AL110225, AL080137, AF087943, AC006221, X84990, AL122098, X93495, AL096744, AL133072, AL049938, AL050138, A08912, AL137283, AL049283, AL137521, U67958, AL080159, I09360, AC005341, AJ012755, A12297, AF067728, I42402, AF026124, U72620, E15569, AC004808, AL122049, X98834, AF119337, AL137560, AL110197, and A93350.

7 HCRNC80 17 975367 AW003857, AI820045, AI523911, AW084836, AW 188197, AI983096, AI333432, AW386991, AI433769, AI379333, AW438811, N41047, AW387003, AA927750, AI435307, 871675, 850080, AA678318, AA748856, AA301830, AW243869, 871707, AI337423, AI382805, AA577365, AI926335, AA568182, 847922, AA057770, 848030, AA047722, 850081, AI539624, AI698267, AI224055, AI365443, N47137, AA903242, AA581759, AI687947, AW081661, AI683568, AI282865, AI887659, AW 152615, AI094749, AI524774, AA928869, AI274452, AW083572, AW 195253, AI831333, AI584130, AI373276, AI673140, AI690748, AI423631, AI815018, AI525669, AI922607, AI678324, AI963564, AI884459, AI277325, AW080346, AI619631, AA019328, AL080126, AF161527, AF131792, AL133655, I79592, AF118558, AR020905, I52013, AL079293, AF118070, X80340, AL137534, AL137463, S75997, D44497, and AL137558.

7 HCRNC80 42 905116 AW003857, AI820045, AI523911, AW084836, AI983096, AI333432, AW 188197, AW386991, AI433769, AI379333, AW438811, N41047, AW387003, AA927750, AI435307, 871675, 850080, AA678318, AA748856, AA301830, 871707, AI337423, AW243869, AI382805, AA577365, AI926335, AA568182, 847922, AA057770, 848030, AA047722, 850081, AI539624, AI698267, AI224055, AI365443, N47137, AA903242, AA581759, 299396, AL119457, AL119399, AL119324, AL134524, AL042544, AL038837, AI541205, AL037051, AL036725, AL036418, AA631969, AL039074, AI557082, AI541321, AI525500, AI557602, AL036924, AL039564, AL039085, AL036858, AW392670, AI557533, AI557731, AL119443, AL039156, AI557808, AL039108, AL039109, AL039128, AL038509, AW372827, AI525556, T18597, AI557238, AI525856, AL037094, AW384394, AI540890, AL039659, AL038531, AL036196, AL119497, AW363220, AW022727, AL119319, AL039625, AL039648, AI541346, AL045337, AL036767, AL036190, AL037082, AW022571, AL119496, AL037526, AL037639, AI557084, AW022456, AL119363, AW020543, AL039678, AL039629, AL119418, AL119335, AI557262, AI541027, AL039423, U46341, AL036238, AL119341, AL038447, AL039150, AI557697, AL119484, AL119391, AL119483, U46350, AL119355, AL040992, AL119522, AL042909, AW023232, AW022086, AI557285, U46349, AW020592, AL119396, AW022874, U46351, AI557258, AL037077, AL119444, AW023469, AW021729, AW020666, AW021930, AI541048, AL039386, AL037726, AL038520, AW023235, AW021777, AW021585, AL038851, U46346, AL036268, AW023351, AI557234, AW021121, AW020196, H65400, AW021400, AW021281, AW021693, AW022983, AL042614, AL039410, U46347, AW021890, AL037085, AW022981, AW021182, AL134518, AI541075, AW023863, AW021178, AW022826, AW021816, U46345, AL036998, AL036733, AL037615, AL119439, AL037205, AW022593, AW022308, AL134528, AW020480, AR060234, AR066494, A81671, AR023813, AR064707, AR069079, AR050070, A62298, Y08991, Y11505, S68736, AB026436, AR054110, and A91160.

8 HPJCK10 18 899642 AI769632, AA312179, T49469, AI829069, AI871379, AI762594, AW300271, 865687, 831122, 832052, AW058189, AI740633, AW237763, AW381197, AI867197, AI380503, AI830241, AW301155, AA937166, AI765531, AI588959, and AI744631.

8 HPJCK10 43 897549 AI479628, AA535256, AI361644, AA612947, AI380503, AI867197, AI744631, AI830241, AI588959, AI765531, W68057, AI740633, AI871379, AA115759, AI924506, AA877095, AA306658, AI927423, AI969915, AI457940, AW237763, AI928256, AW301155, AW316942, AW058189, AW300271, AI762594, AA937166, AA722992, AA688120, AW340134, AI363133, AI990886, AI800708, AI300806, AA447952, AW071230, AI829069, AW 183777, AA133534, AI188687, AA565318, AI017958, AI198517, AI393839, AI760828, 869557, AA953355, AA488884, AI361494, AA053347, H15912, AA604764, AA828477, AA505286, AI189961, T49470, AW138552, AA134945, AI870155, 879824, H94537, AI660160, AA810978, AA767789, H01309, H16019, 869427, AA448886, H44971, AI092045, 865688, AA040351, 832005, AI144157, AA804639, H94448, AW196042, AA025221, AI266135, H28671, AI003662, 832020, 832072, AI475652, AA917021, AW291031, AA040350, AW072896, AA026009, AA207090, AA654222, 831122, AA855110, 865687, AA135022, 879630, AW137350, 883503, 883518, AA204689, H45024, AI769632, and H01355.

9 HE9PF45 19 1017309 AI688462, AI767570, AI798848, AI335004, AI214475, AI201227, AI298943, AA442147, AW118792, AA827126, AI220068, AI262105, AI220061, AI684409, AI279679, AA805448, AW001737, AW341083, AI761004, AW236044, AI984943, AW341978, AA715814, AI733856, AW069227, AW270385, AI923052, AW023111, AA847499, AA410788, AA714011, AW304580, AA669155, AL135377, AW303196, AI801505, AW274349, AI620992, AI792575, AI634187, AI056177, AI696793, AW301350, AI278972, N57681, H73550, AI281818, AI457313, AW265688, AA833875, AA833896, AI801482, AI912401, AW188742, AA831638, AW410354, AA357878, AA832145, AW419389, AI669421, N23504, AI494417, AI923050, AI797998, AI587583, AI587565, AA493226, F24745, AA829036, AA574442, AA120920, AA640430, AI914747, AL044701, AA228778, AL138182, AW162288, AI149537, AW268329, AI432298, AI076228, AW270255, AI360521, AI434037, AL037910, AI635440, T47138, AI358712, AA746911, AA315361, AI538491, T74524, AW303008, H79633, AI689198, AA502991, AA640410, AI280504, H27020, AI246796, AA720702, AI627614, AA534047, AL134418, AC002528, 283844, AL035659, AC007216, AC004491, AC005049, AL031683, AL049780, AC004686, U80017, AL049856, AL109801, AC006241, AF064861, AL033392, AL080243, AC012039, AF129756, AC003963, AC005620, 285986, AC005476, AL135783, AC005409, AL035071, AC002425, AC007308, U95742, AC005237, AL008719, AL031680, U63721, AC006211, AC016830, AC005224, D88270, AC007298, AC008372, AC005399, AC002470, AC004583, AF001550, AC003982, AC003108, AC002544, AC005921, AC007425, D88268, AC007226, AC006121, AF047825, AF134726, U78027, AC007172, AC005057, D86992, AL031584, AC005031, AC004648, AC004832, 298742, AC005225, AC004253, AC004895, AC008033, AC007151, AF207550, AC006023, AF051976, AL031668, AL023553, AC004099, U62293, AC016027, AC006111, AC003041, AC005632, AF001549, AC002350, AP000694, AL117258, AC004765, AC007421, AC007283, AL050318, AC007934, 285996, AL121973, AL035422, AC005736, AC007327, AC004447, AL020993, AC007637, AP000565, AC006077, AL035400, AC005808, U91326, AC001644, AL034549, AB000882, AC004217, AC005753, 282203, AC005781, AC004551, AC007312, AC002316, AC005058, AC006011, AC005071, AL078611, AC004232, AP000045, AP000505, AP000502, AC005548, 293023, AC000159, AL021808, AL023575, AC005599, AC003104, 295113, AC005874, AF134471, AC005261, AC004216, AC007536, AL035588, 284483, AP000112, AL009181, AC005015, AL021453, U96629, Y18000, AL031282, AL135960, AJ131016, AC004408, AL133246, AP000299, AC006312, AC007114, AL109627, AC005730, AC010205, AC004771, AL117694, AL022323, AL031597, Y14768, AC000003, AC004647, AC005209, AC007055, AC004890, 282182, AL137100, AC007227, AL121652, AC006360, AP000501, AC005274, AC007537, 294801, 284466, AC005340, AC005988, AC000353, AC006317, AP000113, AD001527, AC004796, 283840, AL121658, AC004417, AC005251, AC005821, AC005777, U52112, AL049776, AC005899, AL109952, AC004477, AC005940, AC005081, AC006512, AC006539, AC005042, U91321, AJ246003, AC007041, AC000025, AC004106, AL139054, AL035587, AD001502, AB003151, AC007993, AC006480, AC002306, AC002996, AB023048, AC006509, AL031427, AC006468, AC004534, AL031291, AC005102, AP000044, AC005704, AC006130, AC004805, 269917, AC004020, AL049569, AL1-36295, AF196969, 294056, U85195, AC005619, AL031767, AP000555, AP000704, AL121825, AL109628, AC006942, AC005280, AC002310, AC005562, AL049692, AL049631, AF095901, AC004659, AC007023, U62292, AC004813, AP000691, AP000212, AP000134, AF205588, AP000248, AC005519, M13792, AL031577, and AC008056.

9 HE9PF45 44 892844 AI688462, AI767570, AI798848, AW 118792, AI262105, AI279679, AI220068, AW001737, AI220061, AI761004, AI684409, AA827126, AW341083, AW236044, AA805448, AI984943, AA827653, AA860689, AI809154, and AC002528.

9 HE9PF45 45 896960 AI335004, AI214475, AI298943, AI201227, AI936047, and AC002528.

HFEBP27 47 897470 AW328077, AI133550, AI830010, AI929068, AW196817, AI537251, AW328236, AA488252, AW 170187, AA628078, AA191721, AI922228, AI417100, AI672827, AI000225, AI433577, AI349404, AA583448, AI262556, AI926539, AA488698, AA650542, C17501, AW248173, AA310571, AI683576, AA890377, AI096911, AA583817, AI126388, AA488195, AI796647, AA600904, AA689373, AI865678, AI521468, AW327827, AI431656, AA155929, AA995746, AA155840, AI096915, AA857102, AA513694, AA651744, AI340304, AI092349, AI245945, AA147988, AI700512, AA410242, AI191496, AA505299, AA025257, AA643371, AA988782, AI307119, AI200589, AI871751, AA961846, N79603, AA053681, AA187038, AA603098, AA081292, AW247136, AI626108, AI934754, AA862676, AI144335, AA564163, C18629, AA314215, F28451, AI681675, AI720549, AA191372, AA668301, AA375955, AI720137, AA442822, AW000808, AI338096, AA857302, AI707541, AI991119, AA128636, AI459906, AW131547, W19070, AI273284, AA136372, F28489, H68829, AA313519, AI379756, AI718697, AA985257, AI832773, AA187495, AI953967, AI829972, AA603211, AA233036, AI541256, AA436334, AI878866, N69961, F21693, AA180306, AI082281, AI421239, AA232908, AA148016, AI471544, F29877, AI433371, AA948197, AA916947, AI621207, AA890305, AA643938, AA650600, AA928212, F30263, AA916012, AW261952, AA316415, AI557642, AA604035, AA187494, AI582128, 835226, H20011, AA181208, AA436153, AI480366, AA128769, AA908412, AA604060, AI280877, AA457164, D78886, AW026267, AA383221, AA179953, AA603636, AA316539, H19064, N31531, D55798, AI824691, AI347785, AA436279, AI802709, AI749780, AA582088, D55799, AA487574, H18286, AA984880, AA642929, T50340, AA197199, AA197084, AA970976, H90436, AW022039, AA722251, AA305904, F33366, AI459651, H20081, H43525, 807057, H90492, 876129, 831456, AA931749, D55821, AW385239, AI015503, AA025256, AA131595, AW105270, AA328786, 831455, AI804467, 827285, AA306350, AI471350, AI277461, H18967, AI925814, F29153, 807019, AA354246, T80961, AI131296, F34385, F33784, AA599993, D55094, AI307121, AA664612, AA358465, AI191093, F33690, H41291, AA484132, AW021223, F33618, D58254, AI223990, F35873, 876081, AI240337, H46038, AI133637, AA557823, F25933, N63970, AW263906, F34063, AI915175, AA532948, 828643, F17113, AA729613, AA665440, AA657566, F21169, AA081117, AA508438, F22094, AA552461, AA228286, F29455, AA229978, AA527611, AI942257, F17454, AA379932, AI523067, AA484159, AA513058, AA300009, AW 198186, M60854, AR062871, A25909, AR038855, Y09813, U94592, 232836, AF082186, AC005913, X81969, AJ244005, Y16359, D50010, D13509, AJ244004, A20702, AR062872, A43189, AR062873, A43188, A20700, D78345, A85395, A70872, A85476, AR017907, AJ244003, AR037157, A98420, A98423, A98432, A98436, A98417, A98427, AF006072, A98767, A93963, A93964, I63120, AR054723, A62298, 230183, AR038762, AR008429, AR050070, A82595, A82593, X55486, I19525, I44681, A86792, X82786, X83865, A84772, A84776, A84773, A84775, A84774, AR054109, AR067731, AR067732, A58522, D28392, A91750, A93016, AJ243486, X76012, A18053, A60212, A60209, A60210, A60211, I13349, AB025273, I62368, I05558, A35537, A35536, A02136, A04664, A02135, A04663, I84553, I84554, AR031358, AR031365, I05845, X82834, E03627, I48927, A90655, D13316, E 17098, A02712, A77094, A77095, A81878, A95051, I06859, A18050, A23334, A75888, I70384, A64973, A60111, A23633, AR007512, A58524, A58523, AR031566, I00682, A11245, A11624, A11623, E00609, E13740, A11178, E01007, A10361, AR035975, AR035977, AR043601, AJ244006, A22738, A22739, AR017826, A62300, AR063812, M28262, AF149828, Y14219, AJ230972, I15718, I03331, S60422, I01995, E12615, A02710, AR035193, A92133, E14304, A07700, A13393, A13392, AR031488, I13521, I52048, A27396, I25027, AR027100, I49890, I44531, I28266, I21869, I26929, I44515, I26928, I26930, I26927, A91965, I44516, A70040, E16678, A82653, I08051, E16636, I15717, A16035, A22734, A24783, A24782, AR003381, A95117, AJ230935, A06631, AJ231028, AJ230845, AR038066, AJ230951, AJ244007, AJ230902, I66495, I66494, I60241, I60242, I66498, I66497, I66496, I66486, I66487, AF213384, AJ230867, I33632, AR035974, AR035976, AR035978, AJ231009, E03654, AR051957, A20699, E00696, E00697, E03813, I66482, AR009151, I66485, I66483, I66484, AR027099, AR051651, AR051652, AJ231011, T39461, T40600, T52852, T52853, T80960, T81088, T81136, 805273, 805330, 824087, 827479, 832576, H18246, H45187, H45204, H46124, H68463, N21513, N98543, W07431, W42687, W45243, AA053701, AA079081, AA081424, AA083633, AA131659, AA194786, AA197012, AA480768, AA483516, AA484271, AA493174, AA501841, AA513465, AA523046, AA523142, AA524275, AA528406, AA528729, AA531519, AA533546, AA534255, AA535560, AA535776, AA551554, AA554987, AA557881, F15507, F16162, F17276, F17399, AA602024, AA603553, AA570604, AA730722, AA730769, AA745094, AA746807, AA846965, AA886587, AA888702, AA937509, AA995294, F18059, F18559, N83641, N85034, N85277, N86025, N86064, N89022, N89332, N89491, AA090558, AA090768, AA091238, AA092105, AA093142, AA094380, AA095031, AA644078, AA652334, AA248200, AA293341, AA401712, F20560, F21316, F21434, D11523, D11939, AA609480, AA550886, AI265916, AI339402, AI355627, AI185597, and AI280340.

11 HODEA51 21 926697 AA827620, AI888385, AA865660, AI052527, AI208929, AA167720, AI670003, AA507543, AW027137, AA569368, AI888464, AI183691, 888418, AA227188, 860508, N59805, AW270444, AI546863, AI825719, AI341077, AA528030, AA738076, AW339924, AW299746, W72816, AA146582, AA988494, AW411517, AA994601, AI668917, AA992763, AI796034, AA400652, AW085903, AA909932, AA992759, AI278646, AW087489, AA579599, 892489, AA037146, AW 196801, AI682481, AA669170, AC000118, and L02547.

11 HODEA51 48 892841 AC000118.

11 HODEA51 49 904952 AA502331, AW444616, AA568450, AA503839, AI017393, T85589, T78178, T85588, T72043, AI699382, AA299977, T86494, AA335186, AA551860, and AR027051.

11 HODEA51 50 896910 AI888385, N59805, AI208929, AA865660, AA827620, AA167720, AI670003, AA507543, AW027137, AA569368, AI052527, AI183691, AI888464, 888418, 892489, AA227188, 860508, AW270444, AI546863, AA528030, AI825719, AI341077, AA738076, AW339924, AW299746, W72816, AA146582, AA988494, AA994601, AI668917, AW411517, AA992763, AI796034, AA400652, AW085903, AA909932, AA992759, AI278646, AW087489, AA579599, AA037146, AW196801, AI682481, AA669170, AC000118, and L02547.

12 HDRMB11 22 946790 AI610814, AA569565, AI275989, AA199578, F34376, AC003043, AL031276, AC004000, AC006441, AL080243, AC005695, AC006312, AC005084, 295152, AC004650, L78810, AC006241, AC006509, 299716, AC008080, AL021392, AF111169, AC005971, AL109628, AL096701, AL049795, AC005746, and AC006978.

12 HDRMB11 52 896765 AI610814, AA569565, AI275989, AA199578, F34376, AC003043, AL031276, AC006441, AL031281, AC006312, AC008080, AF111169, 295152, AL021392, 299716, AF109907, AL049697, AC005695, AC005971, AL080243, AF196971, AL139054, AL049795, AC006241, AC004650, AC005746, AC004000, and AL080250.

13 HUSJN32 23 1002995 AA932920, AI819491, AW247213, AW247181, AL134701, AW247822, AW275836, AL035940, AI762461, AI831752, AA452294, AW245750, AW027153, AL036958, AI922810, AW248210, AW 160492, AA313277, AI635885, AI338956, AW080446, AI937174, AL035943, AW 162109, AW 152468, AI274816, AW189480, AI499576, AA033735, AW247594, AI346362, AI826792, AA923542, AA777539, AI193128, AI421813, N93432, AW130786, AW402276, AA947271, AI342634, AW393392, AW393396, AI149040, AI673711, AI376896, AI088535, AW130071, AA039878, AI921927, AA771987, AI692812, AI817199, AA670088, AI028464, AI721243, AI986105, AW 188412, AA702460, AA033734, AI572997, AI434254, AA533043, AI445268, AA041522, AI347935, AA531357, AI091679, AW043959, AW401713, AI355407, AI589603, AA595023, AI754889, AI811721, AI364942, AI784537, AI744867, AA204794, AI027683, AW402174, AI262844, AA917745, AI354754, N57425, AA622027, AA313360, AI124096, N39837, AI061258, AI628289, AA912276, AI089942, AI057417, AW073102, W32437, AA603574, AI560758, AW371395, AI697403, AA035560, AI281891, N32217, AA009590, AA025019, AI471961, W22735, W46373, AA838774, AA025020, AA313912, AA595060, N32330, AI093049, AI635968, AI275701, AI493745, AW403145, AA612888, AA622191, AA635666, AA029145, N57316, AA603430, AA834866, AW337459, AA028897, AW 149153, AA877890, AI371000, H24124, AW 130638, AI250444, AA057160, AI039270, AW 194080, AA846644, AA035291, N93855, AA028898, N95223, AA934603, AA291213, AA890551, W22385, AA630581, AA987484, AI685700, AI433483, W70020, AA502870, 858895, W 17327, AI247122, AA858042, AA325636, AI014396, AA494328, AA911083, W87004, AI913912, AI340855, AA129826, AA129827, AI382278, T08768, W21052, AI865726, AW371396, AA564158, AA429968, AI436338, AW405965, AI140087, AW245395, AI539147, AA653092, AI582447, AI368898, W40348, AI187408, AA340869, 243415, AI569165, T63126, N23611, T70293, T93167, AW 190981, AA493458, AA991275, AI034239, AI619598, AI623815, AI033895, AI475369, N23610, AA361071, AA773406, N43920, N90161, AA853509, N43923, 894161, AA569997, AA340025, AW188445, AI123224, 842582, AI274683, AI862871, N34846, AA854692, 894160, AA226649, W21269, AA996376, AW438841, 857583, N55688, AA333832, T72713, T80749, AA340448, AI702045, AI264576, AI702392, W19964, AA492350, AW402184, N74030, AW 102956, 818379, T80869, AA853510, 837242, W46199, H91309, T23934, AI289955, N93031, W87477, T98974, T93076, AI697834, AI304645, AA311029, AW293952, N34850, D31767, AF085348, U77594, I48979, Y11587, A65341, AR013797, AL117460, AL137292, 282022, AL117649, I48978, E05822, AF113699, AL133640, I66342, AL050155, AL133080, A45787, AF132676, AF061836, AF090886, X82434, X80340, M30514, X72889, AL096744, AF113694, AL137548, AL080163, I89947, A08913, AL133560, U00763, A08912, AF090934, AF 126247, AF097996, A77033, A77035, AR029490, A18777, Y16645, A08916, AF090900, Y09972, I09499, AB007812, AL117457, AF113013, A03736, AL137521, AF106862, A08910, AL122110, AL133067, AF100931, A08909, AL049382, I42402, AL117583, AL137550, AL133665, 237987, AL122121, AJ012755, AL137479, AL137557, A08908, AF090903, AF030513, D83032, AR011880, AL137271, E02349, AF111851, X84990, AL080158, AL110222, AF081197, AF081195, A93350, AF090896, AL117435, AR038854, AL049466, AL122050, X83508, 561953, AL133113, E03348, I89931, X52128, AL133557, AL133075, AL050116, AL137273, AL122093, U42766, I00734, AF032666, AL080124, AL122100, I49625, Y11254, AL110196, I33392, X70685, AL137459, AL133016, E00617, E00717, E00778, AL137526, AF153205, AF061795, AF151685, AL023657, AF177401, AL133568, AL137560, U49434, AF067728, AF079763, AF158248, AL122118, AL080148, X63574, AF102578, AF111112, AL137429, AF017437, X87582, X62580, AL049465, AL050146, AL117440, S68736, AL050393, AL050277, AL049938, E03349, AL049314, AR059958, AF090901, AJ005690, AL137478, AL133645, AF026124, AL110221, AF125948, AL096720, U80742, AL133565, AF104032, E12747, AL137463, AF119337, AF113690, AF113677, AL137495, AF090943, AL110296, AF111849, Y14314, AJ003118, AL137294, AF061573, X96540, AL137476, AR020905, AF118094, I03321, AL122106, AL133098, AL137538, E08631, AL050138, AR068751, A08911, AF091084, L19437, AL049430, AL137533, AL137656, 578214, AJ006417, AL110218, AL050108, I89934, I89944, A21103, AF067790, AF113019, AF028823, AL050024, U67958, AF210052, AL080127, E07108, AL137480, AB016226, S36676, AJ000937, AL080154, AL117416, D16301, AL137529, AF146568, S76508, X98834, AL080086, AL117585, U96683, U87620, AF113676, AF185576, U78525, AL133606, S83456, AL133104, A65340, AL137640, E06743, AL049452, AL133081, AF159615, AL122098, Y10080, A07647, AL050092, AL096751, AL133072, U35846, AF008439, AL122123, U88966, AF078844, Y10655, A08907, AL049283, AL080159, AF087943, A90832, AF106697, AF061981, AF106657, Y07905, AF139986, and AF113691.

13 HUSJN32 53 897305 AW247213, AA932920, AL035940, AI819491, AW247181, AL134701, AW245750, AL036958, AW247822, AW275836, AI762461, AA452294, AI831752, AL035943, AW247594, AW027153, AA313277, AW 160492, AI922810, AW248210, AA670088, AI635885, AI338956, AI937174, AW080446, AW 162109, AW 152468, AI274816, AW 189480, AA033735, AI346362, AA923542, AI499576, AI826792, AA777539, AI193128, AI421813, N93432, AW402276, AW 130786, AI149040, AI376896, AA947271, AI342634, AW393392, AW393396, AI673711, AI088535, AW130071, AA039878, AI921927, AA771987, AI817199, AA313360, AI692812, AI028464, N57425, AI721243, AI986105, AW 188412, AA702460, AA033734, AA533043, AI572997, AI434254, AI445268, AA041522, AI347935, AA531357, AI091679, AI589603, AW401713, AW043959, AI355407, AI754889, AI811721, AI364942, AI784537, AI744867, AA204794, AA595023, AI027683, AW402174, AI262844, AA917745, AI354754, AA622027, N39837, AI124096, AI061258, AI628289, AA912276, AI089942, AI057417, AI560758, AW073102, W32437, AA603574, AW371395, AI697403, N57316, AI471961, N32217, AI281891, AA009590, AA025019, W22735, AA035560, W46373, AA025020, AA838774, AA313912, AA595060, N32330, AI093049, AI635968, AI493745, AW403145, AI275701, AA612888, AA622191, AA029145, AA635666, T08768, AA603430, AA834866, AW337459, AW149153, AA028897, AA877890, AI371000, H24124, AI039270, AW 130638, AI250444, AA057160, W22385, AW 194080, W40348, AA035291, N93855, AA846644, AA028898, N95223, AA291213, AA934603, AA890551, AA630581, AA987484, AI433483, W70020, AA502870, AI685700, 858895, W 17327, AI247122, AA858042, AA325636, AI014396, AA911083, AA494328, W87004, 243415, AI913912, AA129826, AI340855, AA129827, AI382278, AW371396, W21052, AI865726, AA564158, AA429968, AI436338, AW405965, AI140087, AW245395, AI539147, AA653092, AI582447, AI368898, N55688, AA340869, AI187408, AI569165, T72713, T63126, N23611, T93167, T70293, AA493458, AW190981, AA991275, AI034239, AI619598, AA361071, N23610, 894160, AI623815, AW402184, W21269, AI033895, AI475369, N43920, AA773406, N90161, AA853509, N43923, 894161, AA340025, AA569997, W19964, AI123224, AA378756, 842582, AI274683, AI862871, AW 188445, AA854692, AA226649, N34846, AA996376, W46199, AW438841, 857583, AA333832, T80749, AA340448, AI702045, AI264576, AI702392, AA492350, AA853510, N74030, AA314671, 818379, AW102956, T80869, 837242, H91309, T23934, AI289955, N93031, 819889, AI697834, W87477, T98974, T93076, AI304645, D31767, AF085348, T72645, 805378, H91405, AA035050, AA056951, C03918, and AA090565.

13 HUSJN32 54 897306 AW024669, AI970941, AI955231, AI857681, AW026331, AI206079, AA993576, W72349, AI094928, AI151201, AA976654, AI298126, AI206570, AI970732, AI279251, AA827346, N21610, AI690486, AA629364, AA565249, W73999, AW083823, AA219179, AA371908, AA947014, AA872651, N23803, H11095, N28847, AA652504, H11186, AW130920, AA652632, AA629373, AA483793, AA757756, 801128, N24852, N31786, W25720, H87388, AA923072, 805750, AA658547, AA505979, AA902241, AA878186, AA092752, 847466, AI207999, N42643, 853032, AA678984, AA219178, AW273692, AA190207, AI581834, AW009129, AI298127, AJ005895, AF077039, AF034790, AF207550, AR069088, AF196971, AF106621, T80869, 818379, 877494, 894160, N23610, W19964, W21269, H93636, AI017701, N70688, N99827, W22385, AA853510, AA773406, AI033895, AI034239, and 243415.

14 HCE4L28 24 1055555 AW299604, AA910649, AI815089, AA701287, H17354, AI338026, AA725780, AW440253, AW295635, AW292781, AA580304, AA059396, AW304383, AI268730, AI761274, AA442825, 856735, AW183032, AI864641, D56341, AL046919, AA099547, AI890959, AI961584, AI625445, AI148478, AI362708, AI285897, AI827547, AI613048, H16611, AA464784, 861616, AI334362, AA488171, AI720129, H75829, AA299333, H10767, AA133786, 239481, AI627407, 243409, W39643, T09408, AI570588, AA902408, AA488225, AA972881, AI362313, H17326, F02924, AA135871, H19243, H04883, AA932084, AA922133, H19244, 243600, AW242187, AA133785, H10768, AA299334, 856889, AI911499, AA491517, AI474042, AA737781, AI355457, AI937442, AI270750, AA897754, T31105, H75830, H16569, 869373, AA137176, W 15236, T16709, AI659107, AA678973, AA436997, 860948, H26851, 836218, H04785, F02239, T31008, 819887, D59033, 869372, AI869489, 848022, 845109, F06672, 836219, AI351348, 847916, AA857732, AA099546, T31831, AW375745, AA938012, AI540377, T30366, AA947105, AA464191, T31084, N78459, AL046920, and 242770.

14 HCE4L28 55 894130 AW440253, AW292781, AA737781, AA857732, AA464191, AW299604, D80227, D80269, AW369651, AW360811, AW177440, C14389, D51799, T03269, D59619, D80210, D80240, D59859, D80164, D80188, D80219, D57483, D80166, D81030, D80253, D59467, D51423, AW375405, D59889, D80378, D58283, D59502, D80212, D80022, C15076, C14331, D80195, D80038, T11417, AW378533, D81111, D80391, D59275, D80366, D80043, D59787, D81026, D58246, D80196, D80045, C14227, D59927, AW377671, D80024, AA305409, D80064, AA305578, D50979, D59503, T03116, D51022, D50995, D80193, D80133, D80248, C05763, D58101, C14014, AA514188, 221582, D80014, D80522, D59610, AA514186, AI525920, C03092, AA809122, D80268, H67866, D80439, D80241, C14407, AW 179328, D80251, AW378534, D80258, AW 179019, D80157, AW375406, D80247, C06015, F13647, AW179332, AW377672, AW 179023, AW 178905, AI525917, H67854, D51759, AW177731, D45260, C14298, AW 179020, D51079, D51103, AW 178893, AW378532, D80168, AW352171, C14344, D50981, D80302, D51221, AA514184, D59474, AW352170, D59317, D51250, C14973, AW377676, AW178762, AA285331, AW 179024, AI525235, AI525912, AW360817, AI525227, D59551, T02974, D59695, D80949, AIS35686, AW360834, AW 178980, AI525923, AI557751, AIS25215, AW178986, AW378528, AI525237, AI525242, AI525925, C14046, AW178906, D45273, AW 178907, AW 178908, AI525903, H67858, 233452, T48593, AI557774, D59627, AIS25222, AW378542, AW367950, AW179018, C16955, AW360855, AW178914, D80228, T03048, T02868, AW
178774, AW 177733, AI525238, AI525928, AI525907, AW177456, AI525216, AW378543, 230160, AW 178781, AW 178911, AW378525, D51213, AW378540, AW352163, AI525239, AI525228, C13958, AW177728, D31458, AR008278, A62298, A84916, AR018138, A62300, AB028859, AJ132110, AF058696, AR054175, A82595, AR060385, AB002449, I14842, I50133, I50126, I50132, I50128, Y17188, AR008277, AR008281, D26022, X67155, A67220, D89785, Y12724, A78862, D34614, A25909, A70867, AR016514, Y17187, D88547, AR060138, A45456, A94995, A26615, AR052274, A43192, A63261, A43190, AR038669, AR062872, A64136, A68321, AR066488, Y09669, AR066487, I79511, A30438, AR008443, X68127, AR016691, AR016690, U46128, and AF123263.

14 HCE4L28 56 896965 AA910649, AI815089, AA701287, H17354, AI338026, AA725780, AW295635, AA580304, AA059396, AW304383, AI268730, AI761274, AA442825, 856735, AI864641, D56341, AW183032, AA099547, AL046919, AI890959, AI961584, AI625445, AI148478, AI362708, AI285897, AI827547, AI613048, H16611, AA464784, 861616, AA488171, AI334362, AI720129, H75829, AA299333, AA133786, 239481, AI627407, 243409, W39643, AI570588, AA902408, AA972881, AI362313, AA488225, H17326, F02924, H19243, AA135871, H04883, AA932084, AA922133, 856889, H19244, 243600, H10768, AA133785, AW242187, AA299334, AI911499, AA491517, AI474042, AI355457, AI937442, AI270750, AA897754, H75830, H16569, 869373, T31105, AA137176, W15236, AI659107, T16709, AA436997, 860948, H26851, 836218, H04785, F02239, 819887, T31008, 869372, D59033, AI869489, H10767, 845109, 848022, F06672, 836219, AI351348, 847916, AA099546, T31831, AW375745, AA938012, AI540377, T30366, AA947105, T31084, N78459, T09408, AL046920, 242770, AI535660, T18597, D50992, AI525757, AI557262, AI535639, AI525556, 233559, AI525500, AI557084, 232887, AI541205, D59751, AI557533, AA058620, C14228, AI540903, H65400, AI536138, AI557082, AI557864, AI525666, 233585, AI525302, AI557312, C14322, AI541075, AI525852, AI526078, AI525878, AI557731, AI557250, AI525316, AI546829, AI541034, N71206, AI557238, AI541321, AI541365, AI5413S6, AI557474, AI525661, AI557258, AI5258S6, AI557602, AIS57809, AI535813, AI557317, AI557155, AI557041, AI541353, AI541154, A62298, 230183, U45328, A82595, A82593, AR050070, U94592, A62300, AF006072, and AR025466.

14 HCE4L28 57 896666 AI031666, AI708012, AA993284, AA604630, W67139, AI553737, AI223395, AW 137493, T16280, AW269130, 843011, H42219, AW269370, W67140, T85029, AA772604, AI034220, AI942425, 850218, F 10147, AA922793, T15843, T87232, AI474372, T31885, 854689, 241583, AA922794, AA029120, AI817793, and RS0164.

15 HFVGM16 25 897053 N28008, AA362394, and T57136.

1S HFVGM16 58 897054 AW293248, AI678037, AI269883, AA894746, AI424848, AI493776, AA778869, AA525497, AA622403, AI095265, N21347, AA564674, AI268502, AA995849, AI249680, AA894745, AW087844, N72839, AI244187, AI300762, AI089147, AI368934, AI339842, AI740804, AI335796, AW192649, AI095231, N28008, AI951011, AW393151, AW393138, AI127890, AW090182, W76593, AA362394, AI906642, TS7136, AA533658, AI797686, AW001850, AW 167918, AI758583, AI638644, AI345688, AW243886, N36182, AA806720, AI700358, AI698391, AI799189, AI582912, AI281867, AI758694, AI446597, AL046466, AI564723, AI590043, AI540821, AI583578, AW 128834, AI554821, AW081383, AW 166870, AW 169684, AI934011, AI473799, AI685005, AI884318, AI570807, AI539800, AI440239, AI539771, AI950664, AI956080, AW 193027, AA916133, AW 166583, AA019328, AI889189, AI621341, AI633125, AI634345, AI289310, AI580451, AI962922, AW081231, AI802542, AL046595, AW084151, AW103928, AIS84130, AI589428, AI684369, AI860348, AW 163834, AI254731, AW087934, AI270706, AI702073, AI916419, AI499570, AI933992, AI270183, AI613038, AI699823, AI249946, AL044192, AI624693, AI538564, AI915291, AW152182, AI445829, AI950729, AI678446, AI885982, AI473536, AI823719, AI446139, AI554344, AI569521, AI824746, AI824576, AI520862, AI611743, AW 198090, AW262552, AI473208, AI610362, AI277008, AI651840, AI922707, AI623905, AW191844, AI125015, AI873604, AI612913, AW 129230, AI478723, AI572096, AI261589, AI610115, AI274745, AI636588, AI811603, AW 198112, AI446775, AW 149311, AI433157, AI685798, AI610446, AI799183, AI687127, AI538850, AW079572, AI499890, AI961278, AI866040, AI951950, AI307285, AI927233, AI567582, AA825548, AI371243, AI932966, AL036705, AI954080, 841605, AI056328, AI567513, AW051088, AW 151893, AW 129659, AI474146, AI521095, AI270055, AI866469, AI561356, AI270039, AW079119, AW 169604, AW151714, AI685517, AI096481, AI567769, AI800341, AL037582, AL037602, AI623867, AA464646, AI648509, AI445611, AW024594, AI352497, AW102794, AI499963, AI536638, AI619817, AI590020, AA641818, AA279795, AW 172982, AI925404, AI798456, AI673363, AI697236, AI472566, AW020419, AI627745, AI637521, AL040243, AI249877, AI537408, AW084219, AI536601, AA788861, AW080746, AI280747, AW 170700, AI470293, AI357273, AI587606, AI884469, AI670009, AI275609, AI281757, AW008226, AI423982, AI280689, AI537677, AI591228, AW078895, AI624293, AW104151, AW078945, AW243451, AI701097, AI798373, N33175, AI627893, AW020876, AI580957, AI567846, AW089844, 832821, AI890223, AA417129, AI308037, AI375420, AW170725, AI250627, AF086351, I89947, AL122045, AL137256, AF159615, AL050366, AF100931, AL133075, AF061981, AL133665, AL133637, A77033, A77035, AF102578, I48978, AL080159, AR038854, AF031147, AF057300, AF057299, S36676, X80340, AL080148, 297214, AF106697, A65341, AL117587, AF026816, AL049324, 213966, AL133619, A15345, AL137276, AF044323, U35846, AL137533, I33392, AF090900, AL050116, AL133113, AL137558, AL080126, AL117435, AR034821, A21103, AF069506, AL080118, 282022, AL137268, A65340, I33391, AL133067, Y16645, U58996, U37312, A18777, AL110280, U92992, AL133558, AL117416, AB007812, AL122100, AF177401, AL133049, L13297, AF051325, 148979, M27260, Y14314, AL137547, X83508, AF082526, A76335, AL110197, S77771, AR066486, AL110222, AF067790, AR013797, AF114170, AL049382, AF141289, AL117460, I09499, AF030513, AL137429, AL122050, AF115392, AF192557, U37359, AF061573, AL137557, AF215669, AL137459, AF047443, AL133557, AL050155, AL137480, E01314, A18788, AB025103, A08913, AL137529, AF004162, AF000145, I89931, A86558, E05822, A08912, AF205861, AL137488, AL050138, X93495, AL080163, A08910, I68732, A08911, AL035458, I49625, AB019565, A08909, X82434, AL133084, 578453, AF126247, AF065135, AL049460, A58545, X57084, AL050149, AF139986, AJ005690, X81464, U75932, A08907, L04849, AL050015, AF113019, A08908, AF118070, AL137478, M30514, X06146, L31396, AF106657, L31397, 576508, 235309, I89934, AC004227, AL050170, AL137463, A30330, A30331, U89906, AF087943, AL137530, AF150103, X59812, AF047716, AL133014, U91329, AR068753, AL110296, AF183393, AL137574, AL137656, AF119358, AL137555, AL137550, E03348, X63410, S83456, AB016226, AL050190, AF107847, AF111851, AL050172, E12579, AF079763, AF159148, AL137538, X84990, AF094480, AF017790, AL137281, AR059883, A45787, AF015958, AF158248, S68736, AL096728, I32738, AL137548, A03736, AL137292, AC002471, AC005374, AL137300, AF119336, AL050277, E03671, E03349, AL137271, AL050280, S73498, E07108, U87620, AL110225, A52563, E02221, AF032666, AF182215, A58524, A58523, AC006197, Y11587, AL110171, AF118094, X66871, AL117463, AL122104, U90884, AL133081, AF153205, AF111849, 582852, AL137254, E01963, AF199027, AJ003118, AF039138, AF039137, AF090896, X93328, AL122121, A32826, A32827, AL137657, AL049464, Y10823, AJ242859, U55017, X67688, AL122098, AF126488, 237987, AL023657, AL133062, and AL137658.

16 HPASD51 26 919903 AA492506, AI380126, AA772096, AI051248, AI796046, N78680, AI923995, AW104821, AW 194429, AI082180, N73147, AA468796, W32940, W86908, AA214384, W15186, W35375, AI640542, T82084, AA294934, 892821, AW050468, AW170216, AA211695, T81773, AI905700, AA354566, AL046681, and AC007655.

16 HPASD51 59 896016 AA492506, AI380126, AA772096, AI051248, AI796046, N78680, AI923995, AW104821, AW 194429, AI082180, N73147, AA468796, W32940, W86908, AA214384, W35375, W 15186, AI640542, T82084, AA294934, 892821, AW050468, AW 170216, AA211695, T81773, AI905700, AA354566, C14331, C14389, C14429, D80195, AI557751, D59859, D59889, D58283, D51423, D80022, D80043, D80166, D59275, D80253, D59467, D59619, D80210, D51799, D80391, D80164, D80240, D59787, D80227, D81030, D59502, D80038, AA305409, D80366, D57483, D80269, D80212, D80196, D80188, D80219, C15076, D59927, D51022, D59610, D80024, D50979, D80193, D50995, D80378, D80241, D80248, D80045, AW 177440, D51060, AA305578, D80522, AA514188, D80133, T03269, . D81026, AW 178893, AW 179328, C14014, C75259, C14407, AW360811, AW378532, AA514186, D80251, D80302, AW178762, AW377671, AW375405, D51250, AW 178775, D58253, AW 177501, AW 177511, D80268, D80134, AW366296, AW360844, AW360817, AW375406, AW378534, D80247, AW 179332, AW377672, AW 179023, AW 178905, AW369651, D80439, AW352117, C05695, AA809122, AW 176467, AW 177505, AW352158, AW352170, D80132, D51079, D59373, AW352171, AW377676, F13647, AW 178906, AW 177731, AW
178907, AW 179019, AW 179024, AI910186, D80168, AW360841, AW 179020, AW 178909, AW 177456, AW 179329, AW
178980, AI905856, AW177733, AW378528, AW 178908, AW 178754, AW
179018, C 14227, D51103, AW378540, D81111, D80157, C14298, AW179004, AW367967, D80064, AW 179012, AW 178914, AW378525, AW352163, D51759, T11417, C06015, AW352174, AW 177722, AW 177728, AW 179009, AW360834, AW 178983, AW178774, AW178911, AW378543, C14344, T48593, 221582, AW352120, D59503, AW178781, D45260, D58246, D59653, C14077, AI535686, D51097, D58101, AA285331, AW177723, H67854, D59627, C03092, H67866, D80258, AW367950, C14975, D80014, AI535850, C14973, AI525923, AW378533, AW178986, AW177508, D51213, AI535961, T03116, AI525917, D59317, AW 177497, AW 177734, D60010, D51221, N66429, AI525235, AC007655, A62298, AR018138, A84916, A62300, Y17188, AJ132110, X82626, AF058696, AR008278, A82595, AB028859, A30438, X68127, X67155, D26022, A25909, X93549, A67220, D89785, A78862, D34614, Y12724, D88547, U79457, I82448, A94995, AR025207, Y17187, AR060385, AB002449, AR008443, I50126, I50132, I50128, I50133, AR008277, AR008281, AR066488, AR016514, X64588, U87247, AR060138, A45456, A26615, AR052274, AB012117, Y09669, A43192, A43190, AR038669, AR054175, and AR066487.

17 HTOIZ28 ~ 919915 AA507824, H44786, AA526779, 27 AA630672, AL120343, AW023111, AA483710, AA829065, AA378682, AA502991, AA904137, AA600368, AW407578, AA362348, AA937427, AP000359, U95090, AL021392, AL034420, AC007878, AP000134, AP000212, AL035587, AC003110, AC004883, AC006515, AC002430, AC005412, AF217403, AC007384, AC005280, AL035458, 283844, AC005726, AP000251, AC006023, AL080243, AC005300, AC007845, AC004983, AL034423, AC007899, AC005971, AP000030, AP000555, AC007051, AC004815, AC005529, AL049557, AC005932, AC006480, AC005826, AC006323, AC004890, AL109654, AL021393, AC009464, AC006312, AC002312, AC005375, AC005899, AC006430, AL031589, AC005081, AC005531, AL022238, AL022328, AC000025, AC004531, AL021878, AL031670, AC008372, AC006130, AC002477, AP000114, AP000046, AC004858, AL035071, U29953, AC004675, 284469, AC006380, AL031295, AL096699, AL022578, AC006064, AC008079, AL121653, AL050321, AP000088, AC007055, AC005089, AL031005, AC007686, AC005095, D84394, AC007388, AL049777, AF008191, AF031078, AC009247, AC005933, 299716, AL031848, AF001549, AC006530, AC008033, AC000353, AC003029, AF088219, AF109907, AC002531, AC004030, AC008984, AC005696, AF030876, AC006449, AC004799, AC007226, AC000062, AL121825, AC009516, AL031311, AC005015, AC007731, AP000692, AC004686, AL022327, AC004231, AC005839, AC005500, AC002470, AP001065, AC001234, AC004167, AC002395, AL035422, 298051, AL022322, U85195, AL031120, AC007707, AC006071, AF060568, U95742, AC005229, 293017, AL049539, AC006486, AL031447, AC002301, AL049569, AC005901, AC005031, AL031668, ACOOSS27, AL021707, AC007564, AC007193, AC005231, AC006511, AC005520, AL021939, AC016025, AP000558, AL121652, AC004448, AC004638, AL024507, AF053356, AL022320, AL021546, AC004019, U91322, AE000658, AC007216, AC007308, 269917, AL034379, AF111169, and AP001052.

17 HTOIZ28 60 892657 AL045848, AI590406, AA501655, AA483710, AA757743, 299426, AA507824, AA133942, AL120343, AA321849, AA323026, T69214, AC004983, AC003110, AL022328, AC005599, AC005280, AP000359, AL021392, AF172277, 298946, AC004890, AL021393, AL034420, AC007276, AF008191, AC006023, AC008372, U95742, AC004706, AF217403, AC006430, AL049780, AC007216, AC007899, AC005531, AP000555, AC000062, AL035587, AC003029, AC005089, AC006130, AC006111, AC005660, 283845, 298051, AC004675, AC007746, AC005562, AC007686, AL034423, AC005971, AC007707, AL031670, AF088219, AP000269, AF134726, AL022327, AL109654, AL035072, AL035458, U29953, AC006312, AC005899, AC005300, AP000103, AP000032, 298950, AC002395, AC000353, AC016025, AL121652, AL035422, AL080243, AC005932, AC006285, AL121825, AL031668, AC005768, AC004231, AF031078, AC007731, AL035451, AC002563, AC007226, AL022578, AC007055, AC005500, AL031589, AC009247, AC005924, AC004448, AL133163, AL031296, AF030876, AF053356, AC004167, AL049557, AC003070, AC004125, D86995, AC004583, AC002117, AL050321, AF196779, AC005347, AC004141, AC005364, AC005946, AC004034, D88547, U80017, AC005225, AF198095, AF001551, and AF 165926.

17 HTOIZ28 61 905877 AI084656, AI660894, AI553885, AI193090, AW026119, AI760275, AI031824, AI814413, AA702018, AI760530, AA706342, AW009035, AW025468, AI770168, AI191049, AA452020, AW083870, AA448320, AA524455, AW374049, AI097278, N66745, N76262, AA934770, AI573184, AI091711, AI281532, AW014097, AI796502, AA524281, AA482767, AI934622, AI308130, AA278854, AI023711, AI051144, AA857349, N21163, N29122, AI368740, T36289, AI735627, AA736812, AW008869, AW087422, AW404882, W72625, AA812709, AI419780, AA470689, AI306712, AA844548, AI244213, AA813855, AI027398, AA977302, AA680364, AA581092, AA429790, AI696520, AW 150629, AI334596, AA809560, AA731117, AA385937, AA864529, AW405923, AA613462, N54932, AI581932, AI193486, AW392663, AA046651, 877543, AA090041, AI193334, H97672, AA085289, AI193948, AA834444, AW392666, AI919095, AI273855, AA906635, 836186, H12497, T98268, 836091, N47535, AI831810, N55998, N44558, AA278421, D11812, T26341, AW392672, AA085356, AI831820, T98322, F13775, and W76533.

17 HTOIZ28 62 897559 N77289, AA904274, AA078536, AA662926, DEMANDE OU BREVET VOLUMINEUX
LA PRESENTE PARTIE DE CETTE DEMANDE OU CE BREVET COMPREND
PLUS D'UN TOME.

NOTE : Pour les tomes additionels, veuillez contacter 1e Bureau canadien des brevets JUMBO APPLICATIONS/PATENTS
THIS SECTION OF THE APPLICATION/PATENT CONTAINS MORE THAN ONE
VOLUME

NOTE: For additional volumes, please contact the Canadian Patent Office NOM DU FICHIER / FILE NAME
NOTE POUR LE TOME / VOLUME NOTE:

Claims (23)

What Is Claimed Is:
1. An isolated nucleic acid molecule comprising a polynucleotide having a nucleotide sequence at least 95% identical to a sequence selected from the group consisting of:

(a) a polynucleotide fragment of SEQ ID NO:X or a polynucleotide fragment of the cDNA sequence included in ATCC Deposit No:Z, which is hybridizable to SEQ ID NO:X;

(b) a polynucleotide encoding a polypeptide fragment of SEQ ID NO:Y or a polypeptide fragment encoded by the cDNA sequence included in ATCC Deposit No:Z, which is hybridizable to SEQ ID NO:X;

(c) a polynucleotide encoding a polypeptide domain of SEQ ID NO:Y or a polypeptide domain encoded by the cDNA sequence included in ATCC Deposit No:Z, which is hybridizable to SEQ ID NO:X;

(d) a polynucleotide encoding a polypeptide epitope of SEQ ID NO:Y or a polypeptide epitope encoded by the cDNA sequence included in ATCC Deposit No:Z, which is hybridizable to SEQ ID NO:X;

(e) a polynucleotide encoding a polypeptide of SEQ ID NO:Y or the cDNA
sequence included in ATCC Deposit No:Z, which is hybridizable to SEQ ID NO:X, having biological activity;

(f) a polynucleotide which is a variant of SEQ ID NO:X;

(g) a polynucleotide which is an allelic variant of SEQ ID NO:X;

(h) a polynucleotide which encodes a species homologue of the SEQ ID
NO:Y;

(i) a polynucleotide capable of hybridizing under stringent conditions to any one of the polynucleotides specified in (a)-(h), wherein said polynucleotide does not hybridize under stringent conditions to a nucleic acid molecule having a nucleotide sequence of only A residues or of only T residues.
2. The isolated nucleic acid molecule of claim 1, wherein the polynucleotide fragment comprises a nucleotide sequence encoding a secreted protein.
3. The isolated nucleic acid molecule of claim 1, wherein the polynucleotide fragment comprises a nucleotide sequence encoding the sequence identified as SEQ ID NO:Y or the polypeptide encoded by the cDNA sequence included in ATCC Deposit No:Z, which is hybridizable to SEQ ID NO:X.
4. The isolated nucleic acid molecule of claim 1, wherein the polynucleotide fragment comprises the entire nucleotide sequence of SEQ ID
NO:X
or the cDNA sequence included in ATCC Deposit No:Z, which is hybridizable to SEQ ID NO:X.
5. The isolated nucleic acid molecule of claim 2, wherein the nucleotide sequence comprises sequential nucleotide deletions from either the C-terminus or the N-terminus.
6. The isolated nucleic acid molecule of claim 3, wherein the nucleotide sequence comprises sequential nucleotide deletions from either the C-terminus or the N-terminus.
7. A recombinant vector comprising the isolated nucleic acid molecule of claim 1.
8. A method of making a recombinant host cell comprising the isolated nucleic acid molecule of claim 1.
9. A recombinant host cell produced by the method of claim 8.
10. The recombinant host cell of claim 9 comprising vector sequences.
11. An isolated polypeptide comprising an amino acid sequence at least 95% identical to a sequence selected from the group consisting of:

(a) a polypeptide fragment of SEQ ID NO:Y or the encoded sequence included in ATCC Deposit No:Z;

(b) a polypeptide fragment of SEQ ID NO:Y or the encoded sequence included in ATCC Deposit No:Z, having biological activity;

(c) a polypeptide domain of SEQ ID NO:Y or the encoded sequence included in ATCC Deposit No:Z;

(d) a polypeptide epitope of SEQ ID NO:Y or the encoded sequence included in ATCC Deposit No:Z;

(e) a secreted form of SEQ ID NO:Y or the encoded sequence included in ATCC Deposit No:Z;

(f) a full length protein of SEQ ID NO:Y or the encoded sequence included in ATCC Deposit No:Z;

(g) a variant of SEQ ID NO:Y;

(h) an allelic variant of SEQ ID NO:Y; or (i) a species homologue of the SEQ ID NO:Y.
12. The isolated polypeptide of claim 11, wherein the secreted form or the full length protein comprises sequential amino acid deletions from either the C-terminus or the N-terminus.
13. An isolated antibody that binds specifically to the isolated polypeptide of claim 11.
14. A recombinant host cell that expresses the isolated polypeptide of claim 11.
15. A method of making an isolated polypeptide comprising:

(a) culturing the recombinant host cell of claim 14 under conditions such that said polypeptide is expressed; and (b) recovering said polypeptide.
16. The polypeptide produced by claim 15.
17. A method for preventing, treating, or ameliorating a medical condition, comprising administering to a mammalian subject a therapeutically effective amount of the polypeptide of claim 11 or the polynucleotide of claim 1.
18. A method of diagnosing a pathological condition or a susceptibility to a pathological condition in a subject comprising:

(a) determining the presence or absence of a mutation in the polynucleotide of claim 1; and (b) diagnosing a pathological condition or a susceptibility to a pathological condition based on the presence or absence of said mutation.
19. A method of diagnosing a pathological condition or a susceptibility to a pathological condition in a subject comprising:

(a) determining the presence or amount of expression of the polypeptide of claim 11 in a biological sample; and (b) diagnosing a pathological condition or a susceptibility to a pathological condition based on the presence or amount of expression of the polypeptide.
20. A method for identifying a binding partner to the polypeptide of claim 11 comprising:

(a) contacting the polypeptide of claim 11 with a binding partner; and (b) determining whether the binding partner effects an activity of the polypeptide.
21. The gene corresponding to the cDNA sequence of SEQ ID NO:Y.
22. A method of identifying an activity in a biological assay, wherein the method comprises:

(a) expressing SEQ ID NO:X in a cell;

(b) isolating the supernatant;

(c) detecting an activity in a biological assay; and (d) identifying the protein in the supernatant having the activity.
23. The product produced by the method of claim 20.
CA002390438A 1999-11-12 2000-11-08 19 human secreted proteins Abandoned CA2390438A1 (en)

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US16475099P 1999-11-12 1999-11-12
US60/164,750 1999-11-12
US21512800P 2000-06-30 2000-06-30
US60/215,128 2000-06-30
PCT/US2000/030674 WO2001034800A1 (en) 1999-11-12 2000-11-08 19 human secreted proteins

Publications (1)

Publication Number Publication Date
CA2390438A1 true CA2390438A1 (en) 2001-05-17

Family

ID=26860817

Family Applications (1)

Application Number Title Priority Date Filing Date
CA002390438A Abandoned CA2390438A1 (en) 1999-11-12 2000-11-08 19 human secreted proteins

Country Status (5)

Country Link
EP (1) EP1230367A4 (en)
JP (1) JP2003521242A (en)
AU (1) AU1916101A (en)
CA (1) CA2390438A1 (en)
WO (1) WO2001034800A1 (en)

Families Citing this family (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6391311B1 (en) * 1998-03-17 2002-05-21 Genentech, Inc. Polypeptides having homology to vascular endothelial cell growth factor and bone morphogenetic protein 1
US20030199674A1 (en) * 1997-10-17 2003-10-23 Genentech, Inc. Secreted and transmembrane polypeptides and nucleic acids encoding the same
US7081521B2 (en) 1998-05-06 2006-07-25 Genentech, Inc. Anti-PRO788 antibodies
ATE348108T1 (en) * 1999-03-08 2007-01-15 Genentech Inc METHOD FOR DETECTING TUMORS
AU2001293846A1 (en) * 2000-09-28 2002-04-08 Bayer Aktiengesellschaft Regulation of a human serine protease
US7084257B2 (en) 2001-10-05 2006-08-01 Amgen Inc. Fully human antibody Fab fragments with human interferon-gamma neutralizing activity
EP1314784A1 (en) * 2001-11-27 2003-05-28 Bayer Ag G-protein coupled receptor LUSTR2 and uses thereof
EP2383278A1 (en) * 2002-07-08 2011-11-02 Genentech, Inc. Method to determine B cell mediated diseases
JP2010151800A (en) * 2008-11-06 2010-07-08 Kumamoto Univ Diagnostic agent for lifestyle-related diseases and/or cancers
US20110189097A1 (en) * 2009-11-09 2011-08-04 Dritan Agalliu Use of WNT inhibitor to inhibit angiogenesis in the CNS

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1999040100A1 (en) * 1998-02-09 1999-08-12 Human Genome Sciences, Inc. 45 human secreted proteins
US6057435A (en) * 1997-09-19 2000-05-02 Genentech, Inc. Tie ligand homologues
AU2589399A (en) * 1998-02-17 1999-08-30 Incyte Pharmaceuticals, Inc. Human short-chain tnf-receptor family protein
EP1074617A3 (en) * 1999-07-29 2004-04-21 Research Association for Biotechnology Primers for synthesising full-length cDNA and their use

Also Published As

Publication number Publication date
WO2001034800A1 (en) 2001-05-17
AU1916101A (en) 2001-06-06
EP1230367A1 (en) 2002-08-14
EP1230367A4 (en) 2003-10-22
JP2003521242A (en) 2003-07-15

Similar Documents

Publication Publication Date Title
US20050239099A1 (en) 27 human secreted proteins
CA2389722A1 (en) 15 human secreted proteins
US6936691B2 (en) Secreted protein HCE3C63
EP1224285A2 (en) 27 human secreted proteins
EP1234030A2 (en) 18 human secreted proteins
US7078186B2 (en) Apoptosis related polynucleotides, polypeptides, and antibodies
CA2390438A1 (en) 19 human secreted proteins
US20030036646A1 (en) Uncoupling protein polynucleotides, polypeptides, and antibodies
US20050010042A1 (en) 26 human secreted proteins
US7247442B1 (en) Antibodies to HHPEN62 polypeptide
CA2385884A1 (en) 26 human secreted proteins
CA2389305A1 (en) 28 human secreted proteins
CA2390421A1 (en) 24 human secreted proteins
WO2001023402A1 (en) 43 human secreted proteins
CA2384584A1 (en) 38 human secreted proteins
EP1235844A1 (en) 28 human secreted proteins
EP1230255A1 (en) 25 human secreted proteins
US20060099575A9 (en) 67 human secreted proteins
EP1556401A2 (en) 22 human secreted proteins
CA2387754A1 (en) 10 human secreted proteins
CA2395815A1 (en) Nucleic acids, proteins, and antibodies
EP1233975A1 (en) 29 human secreted proteins
EP1220942A1 (en) 41 human secreted proteins

Legal Events

Date Code Title Description
FZDE Dead