CA2382743A1 - 48 human secreted proteins - Google Patents

48 human secreted proteins Download PDF

Info

Publication number
CA2382743A1
CA2382743A1 CA002382743A CA2382743A CA2382743A1 CA 2382743 A1 CA2382743 A1 CA 2382743A1 CA 002382743 A CA002382743 A CA 002382743A CA 2382743 A CA2382743 A CA 2382743A CA 2382743 A1 CA2382743 A1 CA 2382743A1
Authority
CA
Canada
Prior art keywords
human
seq
sequence
polypeptide
soares
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
CA002382743A
Other languages
French (fr)
Inventor
Craig A. Rosen
Steven M. Ruben
George A. Komatsoulis
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 CA2382743A1 publication Critical patent/CA2382743A1/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
    • 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
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • A61P15/10Drugs for genital or sexual disorders; Contraceptives for impotence
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • 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
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • A61P25/16Anti-Parkinson drugs
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/18Antipsychotics, i.e. neuroleptics; Drugs for mania or schizophrenia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/24Antidepressants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • 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
    • 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
    • A61P31/12Antivirals
    • 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
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • 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
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/04Immunostimulants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • 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
    • A61P7/04Antihaemorrhagics; 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
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy

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

DEMANDES OU BREVETS VOLUMINEUX
LA PRESENTE PARTIE DE CETTE DEMANDE OU CE BREVETS
COMPRI~:ND PLUS D'UN TOME.
CECI EST L,E TOME 1 DE 2 NOTE: Pour les tomes additionels, veillez contacter 1e Bureau Canadien des Brevets.
JUMBO APPLICATIONS / PATENTS
THIS SECTION OF THE APPLICATION / PATENT CONTAINS MORE
THAN ONE VOLUME.

NOTE: For additional valumes please contact the Canadian Patent Office.

48 Human Secreted Proteins Field of the Invention This invention relates to newly identified polynucleotides and the polypeptides encoded by these polynucleotides, uses of such polynucleotides and polypeptides, 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 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 S' 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 1, 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 ID 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 50% 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 ~cg/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.OaSMP-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 drew 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 Poly~eptides 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.
dbjIBAA20827.11 (all information available through the recited accession number is incorporated herein by reference) which is described therein as "KIAA0372 [Homo Sapiens]". A partial alignment demonstrating the observed homology is shown S immediately below.

IS
>dbj~BAA20827.1~ (AB002370) KIAA0372 [Homo Sapiens] >sp~015077~015077 KIAA0372.
Length = 1564 Plus Strand HSPs:
Score = 3739 (1316.2 bits), Expect = 0.0, Sum P(2) = 0.0 Identities = 729/754 (96~), Positives = 731/754 (96~), Frame = +2 Q: 8 VHCLKKAVRLDSNNHLYWNALGWACYSGIGNYALAQHCFIKSIQSEQINAVAWTNLGVL 187 +HCLKKAVRLDSNNHLYWNALGWACYSGIGNYALAQHCFIKSIQSEQINAVAWTNLGVL
S: 811 LHCLKKAVRLDSNNHLYWNALGWACYSGIGNYALAQHCFIKSIQSEQINAVAWTNLGVL 870 2O Q: 188 YLTNENIEQAHEAFKMAQSLDPSYLMCWIGQALIAEAVGSYDTMDLFRHTTELNMHTEGA 367 YLTNENIEQAHEAFKMAQSLDPSYLMCWIGQALIAEAVGSYDTMDLFRHTTELNMHTEGA
S: 871 YLTNENIEQAHEAFKMAQSLDPSYLMCWIGQALIAEAVGSYDTMDLFRHTTELNMHTEGA 930 Q: 368 LGYAYWVCTTLQDKSNRETELYQYNILQMNAIPAAQVILNKYVERIQNYAPAFTMLGYLN 547 S: 931 LGYAYWVCTTLQDKSNRETELYQYNILQMNAIPAAQVILNKYVERIQNYAPAFTMLGYLN 990 Q: 548 EHLQLKKEAANAYQRAILLLQTAEDQDTYNVAIRNYGRLLCSTGEYDKAIQAFKSTPLEV 727 EHLQLKKEAANAYQRAILLLQTAEDQDTYNVAIRNYGRLLCSTGEYDKAIQAFKSTPLEV
3O S: 991 EHLQLKKEAANAYQRAILLLQTAEDQDTYNVAIRNYGRLLCSTGEYDKAIQAFKSTPLEV 1050 Q: 728 LEDIIGFALALFMKGLYKESSKAYERALSIVESEQDKAHILTALAITEYKQGKTDVAKTL 907 LEDIIGFALAI~FMKGLYKESSKAYERALSIVESEQDKAHILTALAITEYKQGKTDVAKTL
S: 1051 LEDIIGFALALFMKGLYKESSKAYERALSIVESEQDKAHILTALAITEYKQGKTDVAKTL 1110 Q: 908 LFKCSILKEPTTESLQALCAI~GLAMQDATLSKAALNELLKHIKHKDSNYQRCLLTSAIYA 1087 LFKCSILKEPTTESLQALCALGLAMQDATLSKAALNELLKHIKHKDSNYQRCLLTSAIYA
S: 1111 LFKCSILKEPTTESLQALCALGLAMQDATLSKAALNELLKHIKHKDSNYQRCLLTSAIYA 1170 4O Q: 1088 LQGRSVAVQKQISKAVHSNPGDPALWSLLSRWAQYAQRNAKGGWAGNVAHILDSNHGK 1267 LQGRSVAVQKQISKAVHSNPGDPALWSLLSRWAQYAQRNAKGGWAGNVAHILDSNHGK
S: 1171 LQGRSVAVQKQISKAVHSNPGDPALWSLLSRWAQYAQRNAKGGWAGNVAHILDSNHGK 1230 Q: 1268 KALLYTAVNQLAMGSSSAEDEKNTALKTIQKAALLSPGDPAIWAGLMAACHADDKLALMN 1447 4S KALLYTAVNQLAMGSSSAEDEKNTALKTIQKAALLSPGDPAIWAGLMAACHADDKLAL+N
S: 1231 KALLYTAVNQLAMGSSSAEDEKNTALKTIQKAALLSPGDPAIWAGLMAACHADDKLALVN 1290 Q: 1448 NTQPKRIDLYXXXXXXXXXXIKDEKFFENYNQSLEKWSLSQAVTGLIDTGRISEAETLCT 1627 NTQPKRIDLY IKDEKFFENYNQSLEKWSLSQAVTGLIDTGRISEAETLCT
SO S: 1291 NTQPKRIDLYLALLSAVSASIKDEKFFENYNQSLEKWSLSQAVTGLIDTGRISEAETLCT 1350 Q: 1628 KNLKSNPDQPAVILLLRQVQCKPLPESQKPLPDAVLEELQKTVMSNSTSVPAWQWLAHVY 1807 KNLKSNPDQPAVILLLRQVQCKPL ESQKPLPDAVLEELQKTVMSNSTSVPAWQWLAHVY
S: 1351 KNLKSNPDQPAVILLLRQVQCKPLLESQKPLPDAVLEELQKTVMSNSTSVPAWQWLAHVY 1410 S Q: 1808 QSQGMMRAAEMCYRKSLQLASQRGSWSGKXXXXXXXXXXXXKVCMANISNDHWPSLVQEA 1987 QSQGMMRAAEMCYRKSLQLASQRGSWSGK KVCMANISNDHWPSLVQEA
S: 1411 QSQGMMRAAEMCYRKSLQLASQRGSWSGKLSSLLRLALLALKVCMANISNDHWPSLVQEA 1470 Q: 1988 TTEALKLCFCPLAVLLQALLQFKRKMGARETRRLLERWYQPGYPKSIASTARWYLLRHL 2167 IO TTEALKLCFCPLAVLLQALLQFKRKMGARETRRLLERWYQPGYPKSIASTARWYLLRHL
S: 1471 TTEALKLCFCPLAVLLQALLQFKRKMGARETRRLLERWYQPGYPKSIASTARWYLLRHL 1530 Q: 2168 YAKDDYELIDVLVNNAKTHGDTRALELNQRLSSQ 2269 YAKDDYELIDVLVNNAKTHGDTRALELNQRLSSQ
IS S: 1531 YAKDDYELIDVLVNNAKTHGDTRALELNQRLSSQ 1564 Score = 93 (32.7 bits), Expect = 1.5, Sum P(3) = 0.79 Identities = 23/69 (33$), Positives = 35/69 (508), Frame = +2 2O Q: 116 QHCFIKSI-QSEQINAVAWTNLGVLYLTNENIEQAHEAFKMAQSLDPSYLMCWIGQALIA 292 +HC K++ + E+ N AW +GV E +QA A+K A L+P L+ W G A +
S: 27 KHC--KTVLKQEKNNYNAWVFIGVAAAELEQPDQAQSAYKKAAELEPDQLLAWQGLANLY 84 Q: 293 EAVGSYDTMD 322 2S E + D
S: 85 EKYNHINAKD 94 The segments of dbjIBAA20827.11 that are shown as "S" above are set out in the sequence listing as SEQ ID NO. 109 and SEQ ID NO. 111 . Based on the 30 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 3S amino acid sequence set out in the sequence listing as SEQ ID NO. 110 and/or SEQ
ID NO. 112 which correspond to the "Q" sequences in the alignment shown above (gaps introduced in a sequence by the computer are, of course, removed).
It has been discovered that this gene is expressed primarily in the following tissues/cDNA libraries: Soares placenta Nb2HP and to a lesser extent in Soares infant 40 brain 1NIB; Soares fetal liver spleen 1NFLS; Stratagene ovarian cancer (#937219);

Soares NFL T GBC_S1; Soares fetal liver spleen_1NFLS S1; normalized infant brain cDNA; Osteoblasts; Soares fetal heart NbHHI9W; Human T-cell lymphoma,re-excision; Normal Human Trabecular Bone Cells; Human Umbilical Vein, Reexcision; 12 Week Old Early Stage Human, II; NCI CGAP_Ewl;
NCI CGAP_GCB1; Human Umbilical Vein Endothelial Cells, uninduced; Human Hypothalmus,Schizophrenia; 12 Week Old Early Stage Human; Adipocytes; Soares melanocyte 2NbHM; Human aorta polyA+ (TFujiwara); Human Osteoclastoma; H.
Frontal cortex,epileptic,re-excision; Keratinocyte; Human 8 Week Whole Embryo;
Infant brain, LLNL array of Dr. M. Soares 1NIB; Messangial cell, frac 1;
Stromal Cells; NCI CGAP_GCB1; Ku 812F Basophils Line; H. Striatum Depression, subt;
HUMAN TONSILS, FRACTION 2; Human Umbilical Vein Endothelial Cells, fract.
A; HSA 172 Cells; Pancreas Tumor PCA4 Tu; Human Colon Cancer,re-excision;
Synovial IL-1/TNF stimulated; Human Umbilical Vein, Endo. remake; NTERA2 +
retinoic acid, 14 days; Synovial hypoxia-RSF subtracted; Healing groin wound, 7.5 hours post incision; Human Adipose Tissue, re-excision; Myoloid Progenitor Cell Line; Spinal Cord, re-excision; H. Kidney Medulla, re-excision; Mo7e Cell Line GM-CSF treated (lng/ml); L428; NCI CGAP_Prl2; NCI CGAP_Schl;
NCI CGAP_Thy 1; Human Pancreas Tumor; Stromal cell TF274; Macrophage-oxLDL; Human Ovary; Spinal cord; Synovial Fibroblasts (control); Human Fetal Brain; Human Adrenal Gland Tumor; Fetal Liver, subtraction II; Hepatocellular Tumor, re-excision; Macrophage-oxLDL, re-excision; Colon Carcinoma; Dendritic cells, pooled; Soares NhHMPu_S1; Primary Dendritic cells~rac 2; 12 Week Early Stage Human II, Reexcision; Human Adult Pulmonary,re-excision; HUMAN B
CELL LYMPHOMA; Spleen, Chronic lymphocytic leukemia; Bone Marrow Cell Line (RS4,11); NCI CGAP_Gas4; NCI CGAP Pr28; Hodgkin's Lymphoma II;

NCI CGAP_Ewl; NCI CGAP GC4; NCI CGAP_Prl2; Nine Week Old Early Stage Human; NCI CGAP_GC4; NCI CGAP_Kidl l and T cell helper II.
Preferred polypeptides of the present invention comprise immunogenic epitopes shown in SEQ ID NO: 60 as residues: Ser-22 to Asn-29, Pro-37 to Thr-48.
Polynucleotides encoding said polypeptides are also encompassed by the invention.
Antibodies that bind polypeptides of the invention are encompassed by the invention, particularly antibodies that inhibit activity.
The secreted protein can 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, and as nutritional supplements. It may also have a very wide range of biological activities. Representative uses are described in the "Chemotaxis" and "Binding Activity" sections below, in Examples 11, 12, 13, 14, 15, 16, 18, 19, and 20, and elsewhere herein. Briefly, the protein may possess the following activities: cytokine, cell proliferation/differentiation modulating activity or induction of other cytokines; immunostimulating/immunosuppressant activities (e.g.
for treating human immunodeficiency virus infection, cancer, autoimmune diseases and allergy); regulation of hematopoiesis (e.g. for treating anemia or as adjunct to chemotherapy); stimulation or growth of bone, cartilage, tendons, ligaments and/or nerves (e.g. for treating wounds, stimulation of follicle stimulating hormone (for control of fertility); chemotactic and chemokinetic activities (e.g. for treating infections, tumors); hemostatic or thrombolytic activity (e.g. for treating hemophilia, cardiac infarction etc.); anti-inflammatory activity (e.g. for treating septic shock, Crohn's disease); as antimicrobials; for treating psoriasis or other hyperproliferative diseases; for regulation of metabolism, and behavior. Also contemplated is the use of the corresponding nucleic acid in gene therapy procedures. 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.
Many polynucleotide sequences, such as EST sequences, are publicly available and accessible through sequence databases. Some of these sequences are related to SEQ ID NO:11 and may have been publicly available prior to conception of the present invention. Preferably, such related polynucleotides are specifically excluded from the scope of the present invention. To list every related sequence would be cumbersome. Accordingly, preferably excluded from the present invention are one or more polynucleotides comprising a nucleotide sequence described by the general formula of a-b, where a is any integer between 1 to 2997 of SEQ ID
NO:11, b is an integer of 15 to 3011, where both a and b correspond to the positions of nucleotide residues shown in SEQ ID NO:11, and where b is greater than or equal to a + 14.
FEATURES OF PROTEIN ENCODED BY GENE NO: 2 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.
gbIAAA42125.11 (all information available through the recited accession number is incorporated herein by reference; for example, Cell 71, 489-505 (1992)) which is described therein as "homologue to sec61 [Rattus rattus]". A partial alignment demonstrating the observed homology is shown immediately below.
>gb~AAA42125.1~ homologue to sec61 [Rattus rattusl Length = 476 Plus Strand HSPs:
S
Score = 1766 (621.7 bits), Expect = 1.1e-214, Sum P(2) = 1.1e-214 Identities = 348/356 (97$), Positives = 348/356 (97$), Frame = +1 Q: 163 IKFLEVIKPFCVILPEIQKPERKIQFKEKVLWTAITLFIFLVCCQIPLFGIMSSDSADPF 342 IKFLEVIKPFCVILPEIQKPERKIQFKEKVLWTAITLFIFLVCCQIPLFGIMSSDSADPF
S: 3 IKFLEVIKPFCVILPEIQKPERKIQFKEKVLWTAITLFIFLVCCQIPLFGIMSSDSADPF 62 1O Q: 343 YWMRVILASNRGTLMELGISPIVTSGLIMQLLAGAKIIEVGDTPKDRALFNGAQKLFGMI 522 YWMRVILASNRGTLMELGISPIVTSGLIMQLLAGAKIIEVGDTPKDRALFNGAQKLFGMI
S: 63 YWMRVILASNRGTLMELGISPIVTSGLIMQLLAGAKIIEVGDTPKDRALFNGAQKLFGMI 122 Q: 523 ITIGQSIVYVMTGMYGDPSEMGAGICLLITIQLFVAGLIVLLLDELLQKGYGLGSGISLF 702 IS ITIGQSIVYVMTGMYGDPSEMGAGICLLITIQLFVAGLIVLLLDELLQKGYGLGSGISLF
S: 123 ITIGQSIVYVMTGMYGDPSEMGAGICLLITIQLFVAGLIVLLLDELLQKGYGLGSGISLF 182 Q: 703 IATNICETIVWKAFSPTTVNTGRGMEFEGAIIALFHLLATRTDKVRALREAFYRQNLPNL 882 IATNICETIVWKAFSPTTVNTGRGMEFEGAIIALFHLLATRTDKVRALREAFYRQNLPNL
2O S: 183 IATNICETIVWKAFSPTTVNTGRGMEFEGAIIALFHLLATRTDKVRALREAFYRQNLPNL 242 Q: 883 MNLIATIFVFAWIYFQGFRVDLPIKSARYRGQYNTYPIKLFYTSNIPIILQSALVSNLY 1062 MNLIATIFVFAWIYFQGFRVDLPIKSARYRGQYNTYPIKLFYTSNIPIILQSALVSNLY
S: 243 MNLIATIFVFAWIYFQGFRVDLPIKSARYRGQYNTYPIKLFYTSNIPIILQSALVSNLY 302 Q: 1063 VISQMLSARFSGNLLVSLLGTWSDTSSGGPXRAYPVGGLCYYLSPPWSMNSTGTSP 1230 VISQMLSARFSGNLLVSLLGTWSDTSSGGP RAYPVGGLCYYLSPP S S P
S: 303 VISQMLSARFSGNLLVSLLGTWSDTSSGGPARAYPVGGLCYYLSPPESFGSVLEDP 358 3O Score = 338 (119.0 bits), Expect = 1.1e-214, Sum P(2) = 1.1e-214 Identities = 68/68 (100$), Positives = 68/68 (100$), Frame = +3 Q: 1200 MVHELNRYIPTAAAFGGLCIGALSVLADFLGAIGSGTGILLAVTIIYQYFEIFVKEQSEV 1379 MVHELNRYIPTAAAFGGLCIGALSVLADFLGAIGSGTGILLAVTIIYQYFEIFVKEQSEV
3S S: 409 MVHELNRYIPTAAAFGGLCIGALSVLADFLGAIGSGTGILLAVTIIYQYFEIFVKEQSEV 468 Q: 1380 GSMGALLF 1403 GSMGALLF
S: 469 GSMGALLF 476 The segments of gbIAAA4212S.11 that are shown as "S" above are set out in the sequence listing as SEQ ID NO. 113 and SEQ ID NO. 11S . 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 4S 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. 114 and/or SEQ
ID NO. 116 which correspond to the "Q" sequences in the alignment shown above (gaps introduced in a sequence by the computer are, of course, removed).
It has been discovered that this gene is expressed primarily in the following tissues/cDNA libraries: Human Chondrosarcoma; Primary Dendritic Cells, lib 1 and to a lesser extent in Human Pancreas Tumor, Reexcision; Human Adult Pulmonary,re-excision; Human endometrial stromal cells-treated with estradiol;
NCI CGAP_GCB1; T Cell helper I; T cell helper II; Soares ovary tumor NbHOT;
Human OB MG63 treated (10 nM E2) fraction I; Activated T-cells, 24 hrs,re-excision; TF-1 Cell Line GM-CSF Treated; Human Activated T-Cells; Human umbilical vein endothelial cells, IL-4 induced; Human Thymus Stromal Cells;
Soares melanocyte 2NbHM; Soares_placenta_8to9weeks 2NbHP8to9W; Colon Normal III;
Hodgkin's Lymphoma II; Normal Human Trabecular Bone Cells; Human Liver;
Human endometrial stromal cells-treated with progesterone; Human endometrial stromal cells; HUMAN JURKAT MEMBRANE BOUND POLYSOMES;
NCI CGAP_GC4; Human Whole Six Week Old Embryo; normalized infant brain cDNA; Smooth muscle, serum treated; Human Placenta; Human Testes Tumor;
Human Synovial Sarcoma; Soares infant brain 1NIB; CD34+ cell, I, frac II;
Human Umbilical Vein Endothelial Cells, fract. A; Human Placenta; Human Lung;
Stratagene ovary (#937217); Messangial cell, frac 2; STROMAL -OSTEOCLASTOMA; Smooth muscle, ILlb induced;
Soares_pregnant uterus NbHPU; Human Umbilical Vein, Reexcision; Monocyte activated, re-excision; Human Osteoblasts II; NCI CGAP_Co2; NCI CGAP_CoB;
NCI CGAP_ColO; NCI CGAP Pr22; Human Umbilical Vein Endothelial Cells, uninduced; Human Pancreas Tumor; T-Cell PHA 24 hrs; Liver, Hepatoma; Human Adipose; Ulcerative Colitis; Human Testes Tumor, re-excision; Rejected Kidney, lib 4; Ovarian Tumor 10-3-95; Stratagene liver (#937224); Stratagene corneal stroma (#937222); Fetal Heart; CHME Cell Line,untreated; Soares NhHMPu_S1; Stratagene HeLa cell s3 937216; NCI CGAP_Kids; Soares_parathyroid tumor NbHPA;
Human Fetal Kidney, Reexcision; Soares fetal heart NbHHI9W;
Soares senescent fibroblasts NbHSF; Activated T-Cell (l2hs)/Thiouridine labelledEco; NCI CGAP_Panl; Human Primary Breast Cancer Reexcision;
Monocyte activated; HUMAN B CELL LYMPHOMA; Human Bone Marrow, treated; NCI CGAP_Ut3; NCI CGAP_Ov23; NCI CGAP Brn23;
NCI CGAP_Brn25; Bone Marrow Cell Line (RS4,11); NCI CGAP_Utl;
NCI CGAP_CLL1; NCI CGAP_Col4; NCI CGAP_Pr28; Activated T-cell(12h)/Thiouridine-re-excision; Human 8 Week Whole Embryo; Nine Week Old Early Stage Human; CD34 positive cells (Cord Blood); PRMIX; Brain, Kozak;
Human Osteoarthritic Cartilage Fraction IV; Human Astrocyte; Infant brain, Bento Soares; Soares testis NHT; Soares_pineal_gland_N3HPG;
Soares_parathyroid tumor NbHPA; Soares fetal liver spleen_1NFLS S1;
Stratagene hNT neuron (#937233); Sinus piniformis Tumour; Human Osteoarthritic Cartilage Fraction III; Human Normal Cartilage Fraction IV; Tongue Normal;
Human Microvascular Endothelial Cells, fract. B; Hypothalamus; Human OB HOS treated (1 nM E2) fraction I; Human OB MG63 control fraction I; Saos2 Cells, Untreated;
Human Adult Liver, subtracted; LNCAP untreated; Human Aortic Endothelium;
Human Cerebellum, subtracted; Human Primary Breast Cancer,re-excision;
Hodgkin's Lymphoma I; Healing Abdomen wound,70&90 min post incision; Human Pituitary, subtracted; Smooth muscle-ILb induced; H. Epididiymus, caput &
corpus;
NTERA2 teratocarcinoma cell line+retinoic acid (14 days); Human Normal Breast;
Human Fetal Epithelium (Skin); Soares fetal lung NbHLI9W; H. Kidney Cortex, subtracted; Salivary Gland, Lib 2; Human Osteosarcoma; Human Colon, re-excision;
Jurkat T-Cell, S phase; H. Meningima, Ml; Spleen metastic melanoma; Human Adult Small Intestine; Human Infant Brain; Human Neutrophil; b4HB3MA-Cotl8-Bio;
b4HB3MA-Cot0.38-HAP-Ft-6; normalized infant brain cDNA; Human Thymus;
Human Brain, Striatum; NCI CGAP_Pr9; NCI CGAP PrlO; NCI CGAP_Pr22;
Human Fetal Kidney; NCI CGAP_AA1; NCI CGAP_Br2; NCI CGAP Br3;
NCI CGAP_Co3; NCI CGAP_Co9; NCI CGAP_HN4; NCI CGAP_Lul; Human Fetal Dura Mater; Human Adult Testes, Large Inserts, Reexcision; Human Heart;
Merkel Cells; Soares testis NHT; Human Hippocampus; Olfactory epithelium,nasalcavity; Human Placenta (re-excision); Human Activated Monocytes;
Human Rhabdomyosarcoma; Soares NhHMPu_S1; Human Activated T-Cells, re-excision; Soares testis NHT; Synovial Fibroblasts (control);
Hemangiopericytoma;
CHME Cell Line,treated 5 hrs; NTERA2, control; normalized infant brain cDNA;
Smooth muscle, serum induced,re-exc; Macrophage-oxLDL, re-excision; Resting T-Cell Library,II; 12 Week Old Early Stage Human; Human Eosinophils;
NCI CGAP_Co9; NCI CGAP_Pr22; NCI CGAP_Pr25; Colon Carcinoma; Colon Normal II; Adipocytes; H Macrophage (GM-CSF treated), re-excision;
NCI CGAP_BfL; Colon Tumor II; Soares fetal lung NbHLI9W;
Soares multiple sclerosis 2NbHMSP; Human Fetal Lung III; 12 Week Early Stage Human II, Reexcision; Human Fetal Heart; Endothelial cells-control;
NCI CGAP_Ut4; NCI CGAP_Brn23; Soares fetal heart_NbHHI9W;
Soares_placenta 8to9weeks 2NbHP8to9W; CD34 depleted Buffy Coat (Cord Blood), re-excision; Human Microvascular Endothelial Cells, fract. A; Smooth muscle,control; HM1; Human retina cDNA randomly primed sublibrary;
NCI CGAP_GC3; Spleen, Chronic lymphocytic leukemia; NCI CGAP_CoB;
NCI CGAP_GC6; NCI CGAP_Ut2; NCI CGAP CLL1; NCI CGAP_Kid3;

NCI CGAP_Kids; NCI CGAP_KidB; NCI CGAP_Gas4; NCI CGAP_Panl;
NCI CGAP_Brn25; H. Frontal cortex,epileptic,re-excision; Human Endometrial Tumor; Keratinocyte; Soares_pregnant uterus NbHPU; Stratagene neuroepithelium (#937231); Human Cerebellum; Soares fetal liver spleen 1NFLS and Stratagene neuroepithelium (#937231).
Preferred polypeptides of the present invention comprise immunogenic epitopes shown in SEQ ID NO: 61 as residues: Ile-19 to Ile-26, Asp-104 to Arg-109, Phe-234 to Leu-239, Lys-268 to Tyr-279. Polynucleotides encoding said polypeptides are also encompassed by the invention. Antibodies that bind polypeptides of the invention are encompassed by the invention, particularly antibodies that inhibit activity.
The tissue distribution in dendritic cells indicates the protein product of this clone is 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 of this gene product indicates a role in regulating the proliferation; survival; differentiation; and/or activation of hematopoietic cell lineages, including blood stem cells. This gene product is involved in the regulation of cytokine production, antigen presentation, or other processes suggesting a usefulness in the treatment of cancer (e.g. by boosting immune responses).
Since the gene is expressed in cells of lymphoid origin, the natural gene product is involved in immune functions. Therefore it is also used 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 5 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.
Moreover, the protein is useful for the treatment, detection, and/or prevention of developmental, 10 reproductive, skeletal, and proliferative diseases and/or disorders. Based upon the homology of this protein to sec6l, it is contemplated by the present invention that this protein is useful in correcting developmental defects associated with improper protein secretion and/or maturation. The protein, can be used in combination with other proteins or chemicals in a pharamaceutical composition. Such a use would be 15 beneficial in the treatment of certain diseases, particularly those amenable to gene therapy based regimens. 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 20 show utility as a tumor marker and/or immunotherapy targets for the above listed tissues.
Many polynucleotide sequences, such as EST sequences, are publicly available and accessible through sequence databases. Some of these sequences are related to SEQ ID N0:12 and may have been publicly available prior to conception of the present invention. Preferably, such related polynucleotides are specifically excluded from the scope of the present invention. To list every related sequence would be cumbersome. Accordingly, preferably excluded from the present invention are one or more polynucleotides comprising a nucleotide sequence described by the general formula of a-b, where a is any integer between 1 to 2411 of SEQ ID
N0:12, b is an integer of 1S to 2425, where both a and b correspond to the positions of S nucleotide residues shown in SEQ ID N0:12, and where b is greater than or equal to a + 14.
FEATURES OF PROTEIN ENCODED BY GENE NO: 3 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.
gbIAAA67890.11 (all information available through the recited accession number is incorporated herein by reference) which is described therein as "~Rabin3 [Rattus norvegicus]". A partial alignment demonstrating the observed homology is shown 1S immediately below.

>gb~AAA67890.1~ Rabin3 [Rattus norvegicus] >pir~I57546~I57546 Rabin3 - rat >sp~Q62739~Q62739 RABIN3.
Length = 460 Plus Strand HSPs:
Score = 1363 (479.8 bits), Expect = 3.7e-214, Sum P(2) = 3.7e-214 Identities = 272/321 (84$), Positives = 285/321 (88~), Frame = +2 Q: 152 MANDPLEGFHEVNLASPTSPDLLGVYESGTQEQTTSPSVIYRPHPSALSSVPIQANALDV 331 MANDPLEGFHEVNLASPTSPDLLGV + GTQEQTTSPSVIYRPHPS L S IQANAL++
S: 1 MANDPLEGFHEVNLASPTSPDLLGVCDPGTQEQTTSPSVIYRPHPSTLCSATIQANALNL 60 3O Q: 332 SELPTQPVYSSPRRLNCAEISSISFHVTDPAPCSTSGVTAGLTKLTTRKDNYNAEREFLQ 511 S+LpTQPVYSSPR LNCAEIS+IS HV +pA S V AGLT+ T+RKD+ NAEREFLQ
S: 61 SDLPTQPVYSSPRHLNCAEISNISIHVPEPASSVASEVAAGLTRFTSRKDSCNAEREFLQ 120 Q: 512 GATITEACDGSDDIFGLSTDSLSRLRSPSVLEVREKGYERLKEELAKAQRELKLKDEECE 691 3S GAT+TEA G+DDIFGLSTDSLSRLRSPSVLEVREKGYERLKEELAKAQRELKLKDEECE
S: 121 GATVTEASAGNDDIFGLSTDSLSRLRSPSVLEVREKGYERLKEELAKAQRELKLKDEECE 180 Q: 692 RLSKVRDQLGQELEELTASLFEEAHKMVREANIKQATAEKQLKEAQGKIDVLQAEVAALK 871 RLSKVRDQLGQELEELTASLFEEAHKMVREAN+KQATAEKQLKEAQGKIDVLQAEVAALK

S: 181 RLSKVRDQLGQELEELTASLFEEAHKMVREANVKQATAEKQLKEAQGKIDVLQAEVAALK 240 Q: 872 TLVLXXXXXXXXQEPLPGGKTPFKKGHTRNKSTSSAMSGSHQDLSVIQPIVKDCKEADLS 1051 TLVL QEPL GKTPFK GHTRNKSTSSAMSGSHQD S IQ IVKDC+EADLS
S S: 241 TLVLSSSPTSPTQEPLADGKTPFKGGHTRNKSTSSAMSGSHQDFSAIQAIVKDCREADLS 300 Q: 1052 LYNEFRLWKDEPTMDRTVSFL 1114 LYNEFR WKDEPTMDRT FL
S: 301 LYNEFRSWKDEPTMDRTCPFL 321 Score = 736 (259.1 bits), Expect = 3.7e-214, Sum P(2) = 3.7e-214 Identities = 140/149 (93~), Positives = 142/149 (95$), Frame = +3 Q: 1086 PQWTGPCPFLDKIYQEDIFPCLTFSKSELASAVLEAVENNTLSIEPVGLQPIRFVKASAV 1265 IS P CPFLDKIYQEDIFPCLTF+KSELASAVLEAVENNTLSIEPVGLQPIRFVKASAV
S: 312 PTMDRTCPFLDKIYQEDIFPCLTFAKSELASAVLEAVENNTLSIEPVGLQPIRFVKASAV 371 Q: 1266 ECGGPKKCALTGQSKSCKHRIKLGDSSNYYYISPFCRYRITSVCNFFTYIRYIQQGLVKQ 1445 ECGGPKKCALTGQSK CKHRIKLGDSS+YYYISP CRYRITSVCNFFTYIRYIQQGLVKQ
2O S: 372 ECGGPKKCALTGQSKPCKHRIKLGDSSSYYYISPVCRYRITSVCNFFTYIRYIQQGLVKQ 431 Q: 1446 QDVDQMFWEVMQLRKEMSLAKLGYFKEEL 1532 QDVDQMFWEVMQLRKEMSLAKLGYFKEEL
S: 432 QDVDQMFWEVMQLRKEMSLAKLGYFKEEL 460 The segments of gbIAAA67890.11 that are shown as "S" above are set out in the sequence listing as SEQ ID NO. 117 and SEQ ID NO. 119 . 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 30 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. 118 and/or SEQ
ID NO. 120 which correspond to the "Q" sequences in the alignment shown above 3S (gaps introduced in a sequence by the computer are, of course, removed).
It has been discovered that this gene is expressed primarily in the following tissues/cDNA libraries: NCI CGAP GCBl and to a lesser extent in NCI CGAP_Co9; Human Ovarian Cancer Reexcision; Soares testis NHT; Human Testes; Human Fetal Bone; Soares retina N2b4HR; Brain Frontal Cortex, re-excision;

H. Kidney Medulla, re-excision; Human Hypothalmus,Schizophrenia; CHME Cell Line,treated 5 hrs; NCI CGAP_CoB; NCI CGAP_Kid3; PC3 Prostate cell line;
Human Substantia Nigra; Soares NFL T GBC_S1;
Soares_parathyroid tumor NbHPA; Endothelial cells-control; Anergic T-cell;
Bone Marrow Cell Line (RS4,11); H. Frontal cortex,epileptic,re-excision;
NCI CGAP_Co3; NCI CGAP Ewl; NCI CGAP GC4; NCI CGAP_Prl2;
Hodgkin's Lymphoma II and Human 8 Week Whole Embryo.
The tissue distribution in testicular tissue indicates that polynucleotides and polypeptides corresponding to this gene are useful for the treatment and diagnosis of conditions concerning proper testicular function (e.g. endocrine function, sperm maturation), as well as cancer. Therefore, this gene product is useful in the treatment of male infertility and/or impotence. This gene product is also useful in assays designed to identify binding agents, as such agents (antagonists) are useful as male contraceptive agents. Similarly, the protein is believed to be useful in the treatment and/or diagnosis of testicular cancer. The testes are also a site of active gene expression of transcripts that is expressed, particularly at low levels, in other tissues of the body. Therefore, this gene product is expressed in other specific tissues or organs where it may play related functional roles in other processes, such as hematopoiesis, inflammation, bone formation, and kidney function, to name a few possible target indications. The protein is also useful for the treatment, detection, and/or prevention of reproductive diseases and/or disorders. 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.

Many polynucleotide sequences, such as EST sequences, are publicly available and accessible through sequence databases. Some of these sequences are related to SEQ ID N0:13 and may have been publicly available prior to conception of the present invention. Preferably, such related polynucleotides are specifically excluded from the scope of the present invention. To list every related sequence would be cumbersome. Accordingly, preferably excluded from the present invention are one or more polynucleotides comprising a nucleotide sequence described by the general formula of a-b, where a is any integer between 1 to 1862 of SEQ ID
N0:13, b is an integer of 15 to 1876, where both a and b correspond to the positions of nucleotide residues shown in SEQ ID N0:13, and where b is greater than or equal to a + 14.
FEATURES OF PROTEIN ENCODED BY GENE NO: 4 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.
dbjIBAA20825.11 (all information available through the recited accession number is incorporated herein by reference) which is described therein as "KIAA0370 [Homo Sapiens]". A partial alignment demonstrating the observed homology is shown immediately below.
>dbj~BAA20825.1~ (AB002368) KIAA0370 [Homo sapiens] >sp~015076~015076 (FRAGMENT). >gb~AAC06276.1~ (AC003010) KIAA0370 [Homo Sapiens]
~suB
192-801}
Length = 801 Plus Strand HSPs:
Score = 3354 (1180.7 bits), Expect = 0.0, P = 0.0 Identities = 672/732 (91~), Positives = 672/732 (91$), Frame = +3 Q: 408 CL-SCSYIEKFTDFLRLFVSVHLRRIESYSQFPVVEFLTLLFKYTFHQPTHEGYFSCLDI 584 CL S SYIEKFTDFLRLFVSVHLRRIESYSQFPVVEFLTLLFKYTFHQPTHEGYFSCLDI
S: 17 CLVSFSYIEKFTDFLRLFVSVHLRRIESYSQFPVVEFLTLLFKYTFHQPTHEGYFSCLDI 76 S Q: 585 WTLFLDYLTSKIKSRLGDKEAVLNRYEDALVLLLTEVLNRIQFRYNQAXXXXXXXXXXXX 764 WTLFLDYLTSKIKSRLGDKEAVLNRYEDALVLLLTEVLNRIQFRYNQA
S: 77 WTLFLDYLTSKIKSRLGDKEAVLNRYEDALVLLLTEVLNRIQFRYNQAQLEELDDETLDD 136 Q: 765 XXXXXWQRYLRQSLEWAKVMELLPTHAFSTLFPVLQDNLEVYLGLQQFIVTSGSGHRLN 944 IO WQRYLRQSLEWAKVMELLPTHAFSTLFPVLQDNLEVYLGLQQFIVTSGSGHRLN
S: 137 DQQTEWQRYLRQSLEWAKVMELLPTHAFSTLFPVLQDNLEVYLGLQQFIVTSGSGHRLN 196 Q: 945 ITAENDCRRLHCSLRDLSSLLQAVGRLAEYFIGDVFAARFNDALTVVERLVKVTLYGSQI 1124 ITAENDCRRLHCSLRDLSSLLQAVGRLAEYFIGDVFAARFNDALTVVERLVKVTLYGSQI
IS S: 197 ITAENDCRRLHCSLRDLSSLLQAVGRLAEYFIGDVFAARFNDALTVVERLVKVTLYGSQI 256 Q: 1125 KLYNIETAVPSVLKPDLIDVHAQSLAALQAYSHWLAQYCSEVHRQNTQQFVTLISTTMDA 1304 KLYNIETAVPSVLKPDLIDVHAQSLAALQAYSHWLAQYCSEVHRQNTQQFVTLISTTMDA
S: 257 KLYNIETAVPSVLKPDLIDVHAQSLAALQAYSHWLAQYCSEVHRQNTQQFVTLISTTMDA 316 Q: 1305 ITPLISTKVQDKXXXXXXXXXXXXXTTVRPVFLISIPAVQKVFNRITDASALRLVDKAQV 1484 ITPLISTKVQDK TTVRPVFLISIPAVQKVFNRITDASALRLVDKAQV
S: 317 ITPLISTKVQDKLLLSACHLLVSLATTVRPVFLISIPAVQKVFNRITDASALRLVDKAQV 376 2S Q: 1485 LVCRAXXXXXXXXXXXXXXXEQQWPVRSINHASLISALSRDYRNLKPSAVAPQRKMPLDD 1664 LVCRA EQQWPVRSINHASLISALSRDYRNLKPSAVAPQRKMPLDD
S: 377 LVCRALSNILLLPWPNLPENEQQWPVRSINHASLISALSRDYRNLKPSAVAPQRKMPLDD 436 Q: 1665 TKLIIHQTLSVLEDIVENISGESTKSRQICYQSLQESVQVSLALFPAFIHQSDVTDEMLS 1844 S: 437 TKLIIHQTLSVLEDIVENISGESTKSRQICYQSLQESVQVSLALFPAFIHQSDVTDEMLS 496 Q: 1845 FFLTLFRGLRVQMGVPFTEQIIQTFLNMFTREQLAESILHEGSTGCRVVEKFLKILQVW 2024 FFLTLFRGLRVQMGVPFTEQIIQTFLNMFTREQLAESILHEGSTGCRWEKFLKILQVW
3S S: 497 FFLTLFRGLRVQMGVPFTEQIIQTFLNMFTREQLAESILHEGSTGCRWEKFLKILQVW 556 Q: 2025 QEPGQVFKPFLPSIIALCMEQVYPIIAERPSPDVKAELFELLFRTLHHNWRYFFKSTVLA 2204 QEPGQVFKPFLPSIIALCMEQVYPIIAERPSPDVKAELFELLFRTLHHNWRYFFKSTVLA
S: 557 QEPGQVFKPFLPSIIALCMEQVYPIIAERPSPDVKAELFELLFRTLHHNWRYFFKSTVLA 616 Q: 2205 SVQRGIAEEQMENEPQFSAIMQAFGQSFLQPDIHLFKQNLFYLETLNTKQKLYHKKIFRT 2384 SVQRGIAEEQMENEPQFSAIMQAFGQSFLQPDIHLFKQNLFYLETLNTKQKLYHKKIFRT
S: 617 SVQRGIAEEQMENEPQFSAIMQAFGQSFLQPDIHLFKQNLFYLETLNTKQKLYHKKIFRT 676 4S Q: 2385 AMXXXXXXXXXXXXXHKSHDLLQEEIGIAIYNMASVDFDGFFAAFLPEFLTSCDGVDANQ 2564 AM HKSHDLLQEEIGIAIYNMASVDFDGFFAAFLPEFLTSCDGVDANQ
S: 677 AMLFQFVNVLLQVLVHKSHDLLQEEIGIAIYNMASVDFDGFFAAFLPEFLTSCDGVDANQ 736 Q: 2565 KSVLGRNFKMDR 2600 SO KSVLGRNFKMDR
S: 737 KSVLGRNFKMDR 748 The segment of dbjIBAA20825.11 that is shown as "S" above is set out in the sequence listing as SEQ ID NO. 121 . 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. 122 which corresponds to the "Q" sequence in the alignment shown above (gaps introduced in a sequence by the computer are, of course, removed).
It has been discovered that this gene is expressed primarily in the following tissues/cDNA libraries: Human Neutrophil, Activated and to a lesser extent in Activated T-cell(12h)/Thiouridine-re-excision; NCI CGAP_GCB1; Soares adult brain N2b4HB55Y; Soares breast 2NbHBst; Soares breast 3NbHBst; Keratinocyte;
Human Neutrophils, Activated, re-excision; Soares_pregnant uterus NbHPU;
Human Neutrophil; Human Adult Testes, Large Inserts, Reexcision;
Soares fetal liver spleen_1NFLS S1; Human Pancreas Tumor, Reexcision;
Hemangiopericytoma; NTERA2, control; Pancreas Islet Cell Tumor; Fetal Heart;
Soares melanocyte 2NbHM; T Cell helper I; T cell helper II; Primary Dendritic Cells, lib 1; CD34+ cell, I, frac II; NCI CGAP_GC6;
Soares_placenta_8to9weeks 2NbHP8to9W; Healing groin wound - zero hr post-incision (control); Jurkat T-cell G1 phase; Soares testis NHT; Stratagene HeLa cell s3 937216; Soares adult brain N2b5HB55Y; Stratagene liver (#937224); Smooth muscle, serum induced,re-exc; Soares_pregnant uterus NbHPU; HUMAN B CELL
LYMPHOMA; NCI CGAP LuS; NCI CGAP_Ewl; Osteoblasts; Adrenal Gland,normal; Human Lung Cancer, subtracted; Kidney cancer; H. hypothalamus, frac A; Ku 812F Basophils Line; Human Colon Cancer, subtracted; HepG2 Cells, lambda library; Resting T-Cell; Frontal Lobe, Dementia; H. Striatum Depression, subt; Human OB HOS control fraction I; Human OB MG63 treated (10 nM E2) fraction I; Adipocytes,re-excision; HSC172 cells; H. Epididiymus, caput &
corpus;
Human Primary Breast Cancer; NCI CGAP_Brn25; Soares NhHMPu_S1;
Soares testis NHT; Soares_pregnant uterus NbHPU; Invasive poorly differentiated lung adenocarcinoma, metastatic; Human Normal Breast; STROMAL -OSTEOCLASTOMA; Hepatocellular Tumor,re-excision; Hepatocellular Tumor;
Smooth muscle, ILlb induced; Salivary Gland, Lib 2; Human endometrial stromal cells; Jurkat T-Cell, S phase; H. Meningima, Ml; Human Umbilical Vein, Reexcision; KMH2; Human Brain, Striatum; Apoptotic T-cell; Merkel Cells; Human Hypothalmus,Schizophrenia; Gessler Wilms tumor; Human pancreatic islet; Human adult lung 3' directed MboI cDNA; Soares NhHMPu_S 1; Soares NFL T GBC S 1;
Soares fetal heart-NbHHI9W; Soares NSF F8 9W_OT PA P S1;
Soares total fetus Nb2HF8 9w; Soares_parathyroid tumor NbHPA; Liver, Hepatoma; Spinal cord; Human Chondrosarcoma; Soares_fetal heart NbHHI9W;
Epithelial-TNFa and INF induced; Macrophage-oxLDL, re-excision; Human Gall Bladder; KG1-a Lambda Zap Express cDNA library; Soares NhHMPu_S1;
Stratagene NT2 neuronal precursor 937230; CHME Cell Line,untreated; Human Eosinophils; breast lymph node CDNA library; Colon Normal II; Human Placenta;
Adipocytes; Human Testes Tumor; 12 Week Early Stage Human II, Reexcision;
Human Testes, Reexcision; Human Placenta; Human Fetal Heart; Human Primary Breast Cancer Reexcision; CD34 depleted Buffy Coat (Cord Blood), re-excision;
Anergic T-cell; Soares_pregnant uterus NbHPU; Smooth muscle,control; Human Bone Marrow, treated; NCI CGAP_Ut3; NCI CGAP_Ut4; NCI CGAP_Kid3;
NCI CGAP_Panl; NCI CGAP_Pr28; NCI CGAP Brn25; H. Frontal cortex,epileptic,re-excision; Human Endometrial Tumor; neutrophils control;
Human fetal brain (TFujiwara); NCI CGAP_Br2; NCI CGAP_GC1; NCI CGAP Lul;
NCI CGAP_Col2; NCI CGAP_Brl.l; Human 8 Week Whole Embryo; Human Cerebellum; Stratagene pancreas (#937208) and Soares infant brain 1NIB.
Preferred polypeptides of the present invention comprise immunogenic epitopes shown in SEQ ID NO: 63 as residues: Arg-23 to Arg-30. Polynucleotides encoding said polypeptides are also encompassed by the invention. Antibodies that bind polypeptides of the invention are encompassed by the invention, particularly antibodies that inhibit activity.
The secreted protein can 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, and as nutritional supplements. It may also have a very wide range of biological activities. Representative uses are described in the "Chemotaxis" and "Binding Activity" sections below, in Examples 11, 12, 13, 14, 15, 16, 18, 19, and 20, and elsewhere herein. Briefly, the protein may possess the following activities: cytokine, cell proliferation/differentiation modulating activity or induction of other cytokines; immunostimulating/immunosuppressant activities (e.g.
for treating human immunodeficiency virus infection, cancer, autoimmune diseases and allergy); regulation of hematopoiesis (e.g. for treating anemia or as adjunct to chemotherapy); stimulation or growth of bone, cartilage, tendons, ligaments and/or nerves (e.g. for treating wounds, stimulation of follicle stimulating hormone (for control of fertility); chemotactic and chemokinetic activities (e.g. for treating infections, tumors); hemostatic or thrombolytic activity (e.g. for treating hemophilia, cardiac infarction etc.); anti-inflammatory activity (e.g. for treating septic shock, Crohn's disease); as antimicrobials; for treating psoriasis or other hyperproliferative diseases; for regulation of metabolism, and behavior. Also contemplated is the use of the corresponding nucleic acid in gene therapy procedures. 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.
Many polynucleotide sequences, such as EST sequences, are publicly available and accessible through sequence databases. Some of these sequences are related to SEQ ID N0:14 and may have been publicly available prior to conception of the present invention. Preferably, such related polynucleotides are specifically excluded from the scope of the present invention. To list every related sequence would be cumbersome. Accordingly, preferably excluded from the present invention are one or more polynucleotides comprising a nucleotide sequence described by the general formula of a-b, where a is any integer between 1 to 3289 of SEQ ID
N0:14, b is an integer of 15 to 3303, where both a and b correspond to the positions of nucleotide residues shown in SEQ ID N0:14, and where b is greater than or equal to a + 14.
FEATURES OF PROTEIN ENCODED BY GENE NO: 5 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.
dbjIBAAl3399.11 (all information available through the recited accession number is incorporated herein by reference) which is described therein as "Similar to Volbox carteri extensin (S22697) [Homo Sapiens]". A partial alignment demonstrating the observed homology is shown immediately below.

>dbj~BAA13399.1~ Similar to Volbox carteri extensin (S22697) [Homo sapiens]
Length = 559 Plus Strand HSPs:
Score = 1306 (459.7 bits), Expect = 4.0e-211, Sum P(3) = 4.0e-211 Identities = 249/260 (95~), Positives = 249/260 (95$), Frame = +3 Q: 321 MPLVKRNIDPRHLCHTALPRGIKNELECVTNISLANIIRQLSSLSKYAEDIFGELFNEAH 500 MPLVKRNIDPRHLCHTALPRGIKNELECVTNISLANIIRQLSSLSKYAEDIFGELFNEAH
S: 1 MPLVKRNIDPRHLCHTALPRGIKNELECVTNISLANIIRQLSSLSKYAEDIFGELFNEAH 60 IS Q: 501 SFSFRVNSLQERVDRLSVSVTQLDPKEEELSLQDITMRKAFRSSTIQDQQLFDRKTLPIP 680 SFSFRVNSLQERVDRLSVSVTQLDPKEEELSLQDITMRKAFRSSTIQDQQLFDRKTLPIP
S: 61 SFSFRVNSLQERVDRLSVSVTQLDPKEEELSLQDITMRKAFRSSTIQDQQLFDRKTLPIP 120 Q: 681 LQETYDVCEQPPPLNILTPYRDDGKEGLKFYTNPSYFFDLWKEKMLQDTEDXXXXXXXXX 860 S: 121 LQETYDVCEQPPPLNILTPYRDDGKEGLKFYTNPSYFFDLWKEKMLQDTEDKRKEKRKQK 180 Q: 861 XXNLDRPHEPEKVPRAPHDRRREWQKLAQGPELAEDDANLLHKHIEVANGPASHFETRPQ 1040 NLDRPHEPEKVPRAPHDRRREWQKLAQGPELAEDDANLLHKHIEVANGPASHFETRPQ
2S S: 181 QKNLDRPHEPEKVPRAPHDRRREWQKLAQGPELAEDDANLLHKHIEVANGPASHFETRPQ 240 Q: 1041 TYVDHMDGSYSLSALPFSQM 1100 TYVDHMDGSYSLSALPFSQM
S: 241 TYVDHMDGSYSLSALPFSQM 260 Score = 518 (182.3 bits), Expect = 4.0e-211, Sum P(3) = 4.0e-211 Identities = 112/158 (70~), Positives = 112/158 (708), Frame = +1 Q: 1525 RAAPVCETVPVHPLPQGEVQGXXXXXXXXXXXXXGIRPSSPVTVTALAHPPSGLHPTPST 1704 S: 402 RAAPVCETVPVHPLPQGEVQGLPPPPPPPPLPPPGIRPSSPVTVTALAHPPSGLHPTPST 461 Q: 1705 AXXXXXXXXXXXXXXXXXXXXEPKRHPSTLPVISDARSVLLEAIRKGIQLRKVEEQREQE 1884 A EPKRHPSTLPVISDARSVLLEAIRKGIQLRKVEEQREQE
S: 462 APGPHVPLMPPSPPSQVIPASEPKRHPSTLPVISDARSVLLEAIRKGIQLRKVEEQREQE 521 Q: 1885 AKHERIENDVATILSRRIAVEYXXXXXXXXXXXXXWLE 1998 AKHERIENDVATILSRRIAVEY WLE
S: 522 AKHERIENDVATILSRRIAVEYSDSEDDSEFDEVDWLE 559 The segments of dbjIBAA13399.11 that are shown as "S" above are set out in the sequence listing as SEQ ID NO. 123 and SEQ ID NO. 12S . 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. 124 and/or SEQ
ID NO. 126 which correspond to the "Q" sequences in the alignment shown above (gaps introduced in a sequence by the computer are, of course, removed).
It has been discovered that this gene is expressed primarily in the following tissues/cDNA libraries: Human Brain, Striatum and to a lesser extent in Human Testes, Reexcision; Osteoblasts; human corpus colosum; Bone Marrow Stromal Cell, untreated; H. Frontal cortex,epileptic,re-excision; Keratinocyte; Human 8 Week Whole Embryo; Nine Week Old Early Stage Human; Soares infant brain 1NIB;
KMH2 cell line; Human 8 Week Whole Embryo, subtracted; Human Fetal Brain;
Human T-cell lymphoma,re-excision; Human Skin Tumor; Human adult small intestine,re-excision; H. Whole Brain #2, re-excision; Human Hypothalamus,schizophrenia, re-excision; NTERA2 + retinoic acid, 14 days; H.
Meningima, M1; Human Manic Depression Tissue; Spinal Cord, re-excision; Human Infant Brain; human ovarian cancer; Human Ovarian Cancer Reexcision; Brain frontal cortex; Human Synovial Sarcoma; Human Amygdala; Human Testes; Human fetal brain (TFujiwara); NCI CGAP_GC6; Soares testis NHT;
Soares NFL_T GBC_S1; normalized infant brain cDNA and Stratagene neuroepithelium NT2RAMI 937234.
The gene encoding the disclosed cDNA is believed to reside on chromosome 6. Accordingly, polynucleotides related to this invention are useful as a marker in linkage analysis for chromosome 6.
The tissue distribution in human brain indicates the protein product of this clone is 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 Alzheimeris Disease, Parkinsonis Disease, Huntingtonis 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.
Many polynucleotide sequences, such as EST sequences, are publicly available and accessible through sequence databases. Some of these sequences are related to SEQ ID NO:15 and may have been publicly available prior to conception of the present invention. Preferably, such related polynucleotides are specifically excluded from the scope of the present invention. To list every related sequence would be cumbersome. Accordingly, preferably excluded from the present invention are one or more polynucleotides comprising a nucleotide sequence described by the general formula of a-b, where a is any integer between 1 to 2041 of SEQ ID
NO:1S, b is an integer of 1S to 20SS, where both a and b correspond to the positions of nucleotide residues shown in SEQ ID NO:1S, and where b is greater than or equal to a + 14.
S
FEATURES OF PROTEIN ENCODED BY GENE NO: 6 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.
embICAASS026.11 (all information available through the recited accession number is incorporated herein by reference) which is described therein as "ribosomal protein L1S [Rattus norvegicus]". A partial alignment demonstrating the observed homology is shown immediately below.
1S >emb~CAA55026.1~ ribosomal protein L15 [Rattus norvegicus]
>pir~JC2369~JC2369 ribosomal protein L15 - rat Length = 204 Plus Strand HSPS:
Score = 969 (341.1 bits), Expect = 7.5e-97, P = 7.5e-97 Identities = 183/204 (89$), Positives = 183/204 (89~), Frame = +2 2S Q: 110 MGAYKYIQELWRKKQSDVMRFLLRVRCWQYRQLSALHRAPRPTRPDKARRLGYKAKQXXX 289 MGAYKYIQELWRKKQSDVMRFLLRVRCWQYRQLSALHRAPRPTRPDKARRLGYKAKQ
S: 1 MGAYKYIQELWRKKQSDVMRFLLRVRCWQYRQLSALHRAPRPTRPDKARRLGYKAKQGYV 60 Q: 290 XXXXXXXXXXXXXXXXXXATYGKPVHHGVNQLKFARSLQSVAEERAGRHCGALRVLNSYW 469 S: 61 IYRIRVRRGGRKRPVPKGATYGKPVHHGVNQLKFARSLQSVAEERAGRHCGALRVLNSYW 120 Q: 470 VGEDSTYKFFEVILIDPFHKAIRRNPDTQWITKPVHKHREMRGLTSAGRKSRGLGKGHKF 649 VGEDSTYKFFEVILIDPFHKAIRRNPDTQWITKPVHKHREMRGLTSAGRKSRGLGKGHKF
3S S: 121 VGEDSTYKFFEVILIDPFHKAIRRNPDTQWITKPVHKHREMRGLTSAGRKSRGLGKGHKF 180 Q: 650 HHTIGGSRRAAWRRRNTLQLHRYR 721 HHTIGGSRRAAWRRRNTLQLHRYR
S: 181 HHTIGGSRRAAWRRRNTLQLHRYR 204 The segment of embICAA55026.11 that is shown as "S" above is set out in the sequence listing as SEQ ID NO. 127. 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. 128 which corresponds to the "Q" sequence in the alignment shown above (gaps introduced in a sequence by the computer are, of course, removed).
It has been discovered that this gene is expressed primarily in the following tissues/cDNA libraries: Soares fetal liver spleen 1NFLS and to a lesser extent in Bone Marrow Cell Line (RS4,11); Osteoblasts; Human Fetal Kidney, Reexcision;
Soares fetal lung NbHLI9W; Human Lung Cancer,re-excision; Smooth muscle, serum induced,re-exc; Endothelial-induced; Keratinocyte; T cell helper II;
Nine Week Old Early Stage Human; Hodgkin's Lymphoma II; Human Fetal Kidney; Human Fetal Lung III; Human Microvascular Endothelial Cells, fract. A; Human 8 Week Whole Embryo; Human Fetal Dura Mater; Human Testes Tumor; Smooth muscle,control; Soares placenta Nb2HP; Human Fetal Brain; Smooth muscle, serum treated; Endothelial cells-control; Early Stage Human Brain; Activated T-cell(12h)/Thiouridine-re-excision; Human Synovial Sarcoma; Human Endometrial Tumor; Human Fetal Heart; Smooth Muscle- HASTE normalized; wilm's tumor;
Human umbilical vein endothelial cells, IL-4 induced; Soares_pineal_gland N3HPG;
Human Thymus; Soares breast 3NbHBst; Hemangiopericytoma; Macrophage-oxLDL, re-excision; Colon Normal III; Raji Cells, cyclohexamide treated; Human Thymus; HUMAN JURKAT MEMBRANE BOUND POLYSOMES; Soares melanocyte 2NbHM; HUMAN B CELL LYMPHOMA; Amniotic Cells - Primary Culture; Jurkat T-Cell, S phase; T-Cell PHA 24 hrs; Hepatocellular Tumor, re-excision; Adipocytes; Human fetal heart, Lambda ZAP Express;
Soares_parathyroid tumor NbHPA; Soares fetal heart NbHHI9W; Human 5 Cerebellum; Soares infant brain 1NIB; Early Stage Human Lung, subtracted;
HSA
172 Cells; Human Gall Bladder; CHME Cell Line,untreated; Colon Tumor; 12 Week Old Early Stage Human; Dendritic cells, pooled; Anergic T-cell; Human Bone Marrow, treated; Spleen, Chronic lymphocytic leukemia; Cem cells cyclohexamide treated; Smooth muscle, ILIb induced; Human Fetal Epithelium (Skin); HL-60, PMA
10 4H, re-excision; Human Umbilical Vein Endothelial Cells, uninduced; Resting T-Cell Library,II; Activated T-Cells, 12 hrs, subtracted; Apoptotic T-cell, re-excision;
Human Prostate Cancer, Stage C fraction; Human Uterine Cancer; Human Activated Monocytes; Human Activated T-Cells, re-excision; breast lymph node CDNA
library;
12 Week Early Stage Human II, Reexcision; Human OB MG63 control fraction I;
15 LNCAP untreated; Amniotic Cells - TNF induced; Stratagene placenta (#937225);
Prostate BPH; Macrophage (GM-CSF treated); H Macrophage (GM-CSF treated), re-excision; Human Placenta; Stratagene endothelial cell 937223; T Cell helper I;
Human Testes; Soares_parathyroid tumor NbHPA; Human OB HOS control fraction I; Supt Cells, cyclohexamide treated; Human Fetal Bone; Human endometrial stromal 20 cells-treated with estradiol; LNCAP prostate cell line; Jurkat T-cell G1 phase;
Apoptotic T-cell; NCI CGAP_Pr2; Human Heart; Human Ovary; Human Testes Tumor, re-excision; Synovial Fibroblasts (control); Human Adult Pulmonary,re-excision; Activated T-Cell (l2hs)/Thiouridine labelledEco; Stratagene endothelial cell 937223; Soares senescent fibroblasts NbHSF; Primary Dendritic Cells, lib 1; H.
25 Atrophic Endometrium; HSC172 cells; Smooth muscle, control, re-excision;
Smooth muscle-ILb induced; Messangial cell, frac 2; Human Colon Cancer,re-excision;

Human endometrial stromal cells-treated with progesterone; Stratagene endothelial cell 937223; T-Cell PHA 16 hrs; Stratagene fetal spleen (#937205); Macrophage-oxLDL; Epithelial-TNFa and INF induced; NCI CGAP Pr2l; Bone Marrow Stromal Cell, untreated; Human Whole Six Week Old Embryo; Human Ovarian Cancer Reexcision; Human fetal heart, Lambda ZAP Express;
Soares senescent fibroblasts_NbHSF; Colon Carcinoma;
Soares fetal lung NbHLI9W; Colon Tumor II; Human fetal heart, Lambda ZAP
Express; Soares testis NHT; Soares senescent fibroblasts_NbHSF; Human Testes, Reexcision; Soares fetal lung NbHLI9W; Supt cells, cyclohexamide treated, subtracted; Human B Cell 8866; LNCAP + o.3nM 81881; Resting T-Cell; Human Adult Retina; HEL cell line; Glioblastoma; Human Stomach,re-excision; Human Osteosarcoma; Human endometrial stromal cells; Healing groin wound, 6.5 hours post incision; normalized infant brain cDNA; Human Activated T-Cells; Human Pancreas Tumor; Human Placenta (re-excision); CHME Cell Line,treated 5 hrs;
Fetal Liver, subtraction II; Fetal Heart; Normal colon; Bone marrow; Human Amygdala;
Activated T-Cells, 8 hrs., ligation 2; Human (HCC) cell line liver (mouse) metastasis, remake; Human Aortic Endothelium; Hodgkin's Lymphoma I; Fetal Heart, re-excision; Activated T-cells; Human Skin Tumor; Human Adult Heart,re-excision;
Resting T-Cell, re-excision; NTERA2 teratocarcinoma cell line+retinoic acid (14 days); Stomach cancer (human),re-excision; Pancreas Tumor PCA4 Tu;
Hepatocellular Tumor; NTERA2 + retinoic acid, 14 days; Human Amygdala,re-excision; Human Prostate; KMH2; Human Dermal Endothelial Cells,untreated;
L428;
12 Week Old Early Stage Human, II; NCI CGAP_Alvl; Stromal cell TF274; Spinal cord; Ulcerative Colitis; NTERA2, control; Stratagene liver (#937224);
NCI CGAP_Prl; Human T-Cell Lymphoma; Human Substantia Nigra; Colon Normal II; NCI CGAP GC3; NCI CGAP LuS; Soares fetal lung NbHLI9W;

Primary Dendritic cells,frac 2; human tonsils; NCI_CGAP Prl2; Stratagene colon (#937204); Stratagene NT2 neuronal precursor 937230;
Soares fetal heart NbHHI9W; Soares_parathyroid tumor NbHPA; Activated T
Cells, l2hrs, differentially expressed; Bone Marrow Stroma, TNF&LPS ind; Lung Mesothelium; Colon, tumour; Human Normal Cartilage Fraction IV; Human OB
HOS treated (1 nM E2) fraction I; Human (Caco-2) cell line, adenocarcinoma, colon, remake; stomach cancer (human); Human colon carcinoma (HCC) cell line, remake;
Human Colon Carcinoma (HCC) cell line; HUMAN STOMACH; Smooth Muscle Serum Treated, Norm; Human Lung; Human Quadriceps; Human T-cell lymphoma,re-excision; Human Soleus; Stratagene ovary (#937217); Human Pineal Gland; STROMAL -OSTEOCLASTOMA; Hepatocellular Tumor,re-excision; pBMC
stimulated w/ poly I/C; human corpus colosum; Healing groin wound, 7.5 hours post incision; Human Colon, re-excision; Myoloid Progenitor Cell Line; H.
Meningima, .
M1; Human Manic Depression Tissue; H. Lymph node breast Cancer; Brain Frontal Cortex, re-excision; Stratagene endothelial cell 937223; Stratagene ovarian cancer (#937219); Spinal Cord, re-excision; Human Chronic Synovitis; Mo7e Cell Line GM-CSF treated (lng/ml); TF-1 Cell Line GM-CSF Treated; human ovarian cancer;
NCI CGAP_Br3; NCI CGAP_Pr24; Human Pancreas Tumor, Reexcision; Human Hippocampus; NCI CGAP_GC3; NCI CGAP_Lym3; NCI CGAP_Prl l;
NCI CGAP_Brl.l; NCI CGAP_GC4; Stratagene NT2 neuronal precursor 937230;
Human Liver, normal; Barstead spleen HPLRB2; Human fetal lung;
NCI CGAP_Kid6; Soares fetal heart NbHHI9W;
Soares senescent fibroblasts_NbHSF; Human Osteoclastoma;
Soares fetal heart_NbHHI9W; Monocyte activated; CD34 positive cells (Cord Blood); H. Frontal cortex,epileptic,re-excision; NCI CGAP_Kid6;
Soares fetal lung NbHLI9W; NCI CGAP_Lym3;

Soares_placenta_8to9weeks 2NbHP8to9W; Activated T-Cells, 12 hrs., ligation 2;
Hemangiopericytoma; Human Cardiomyopathy, diff exp; Human colon cancer, metaticized to liver, subtraction; Activated T-Cells, Ohrs, subtracted;
CD34+cells, II;
Human Osteoarthritic Cartilage Fraction III; Human Adult Heart, subtracted;
Human Infant Adrenal Gland; Human osteoarthritic,fraction II; Human Umbilical Vein Endothelial cells, frac B, re-excision; Human Prostate Cancer, Stage B2; Raji cells, cyclohexamide treated, subtracted; Normal Ovary, Premenopausal; Activated T-Cells, 8 hrs, subtracted; LNCAP + 30nM 81881; H. Frontal Cortex, Epileptic; Ku 812F
Basophils Line; K562 + PMA (36 hrs),re-excision; Colorectal Tumor; Saos2, Dexamethosome Treated; Human,Prostate Cancer, Stage B2 fraction; H. Normalized Fetal Liver, II; Human Colon, subtraction; HL-60, PMA 4H; H. Striatum Depression, subt; Human Adult Spleen; Human Cerebellum, subtracted; Human Cardiomyopathy, subtracted; Human Adult Pulmonary; Adipocytes,re-excision; Human Fetal Spleen;
Human Pancreatic Carcinoma; human colon cancer; CD40 activated monocyte dendridic cells; H. Epididiymus, caput & corpus; H. Epididiymus, cauda; Soares retina N2b4HR; Stratagene fibroblast (#937212); Soares_pineal_gland_N3HPG; H.
Whole Brain #2, re-excision; Human fetal heart, Lambda ZAP Express; Healing groin wound - zero hr post-incision (control); B Cell lymphoma; Synovial IL-1/TNF
stimulated; Human Whole Brain #2 - Oligo dT > I.SKb; Human Adipose Tissue, re-excision; Stratagene colon (#937204); Stratagene lung carcinoma 937218; Spleen metastic melanoma; Human Bone Marrow, re-excision; Human Osteoblasts II;
NCI CGAP_Co3; NCI CGAP GC3; NCI CGAP_Lul; NCI CGAP_Thyl; Merkel Cells; Human Adipose; Human Rhabdomyosarcoma; Human Chondrosarcoma;
Soares fetal heart_NbHHI9W; Human fetal heart, Lambda ZAP Express;
NCI CGAP_Co2; NCI CGAP Co3; NCI CGAP_PrlO; NCI CGAP_Prl2; Human Thymus Stromal Cells; Human Adrenal Gland Tumor; Rejected Kidney, lib 4;

Ovarian Tumor 10-3-95; Stratagene lung (#937210); Stratagene fetal retina 937202;
Stratagene lung carcinoma 937218; NCI CGAP_Pr2; NCI CGAP_Lym3;
Soares fetal heart NbHHI9W; Soares_pregnant uterus NbHPU; PC3 Prostate cell line; Human pancreatic islet; Soares testis NHT;
Soares_parathyroid tumor NbHPA; Stratagene hNT neuron (#937233); Stratagene NT2 neuronal precursor 937230; Soares fetal heart_NbHHI9W; Human Placenta;
Human heart cDNA (YNakamura); NCI CGAP GC4;
Soares fetal heart NbHHI9W; Human Neutrophil, Activated; Human Primary Breast Cancer Reexcision; CD34 depleted Buffy Coat (Cord Blood), re-excision;
NCI CGAP_LuS; neutrophils control; 22 week old human fetal liver cDNA library;
Stratagene muscle 937209; Stratagene HeLa cell s3 937216; Stratagene hNT
neuron (#937233); Stratagene neuroepithelium NT2RAMI 937234;
Soares_pineal_gland N3HPG; Human Fetal Brain; Human Namalwa Membrane Bound Polysomes; HCBB's Subtractive (- mito genes); Human Fetal Heart, Differential (Adult-Specific); Human Pancreas Tumor; HL-60, PMA 1d, subtracted;
Human Skin Fibroblasts, normal; C7MCF7 cell line, estrogen treated, subtraction;
HeLa cell line; Human Thymus, subtracted; H Amygdala Depression, subtracted;
Human Adult Lymph Node; HCC cell line metastisis to liver; Prostate/LNCAP, subtraction I; Human Brain, striatum, re-excision; Human Osteoarthritic Cartilage . Fraction IV; Human Astrocyte; Pericardium; Larynx normal #10 261-273; Human Adult Spleen, fractionII; L428 cell line; Thyroid Thyroiditis; Human Macrophage, subtracted; Human Infant Adrenal Gland, Subtracted; Soares ovary tumor NbHOT;
Cheek Carcinoma; Palate carcinoma; Rectum tumour; Larynx Normal; Larynx Carcinoma; Colon Tumor; Liver Tumour Met 5 Tu; 7 Week Old Early Stage Human, subtracted; CD34+ cell, I; Larynx tumor; Tongue carcinoma; Human Gastrocnemius;
Human Tonsils, lib I; Tongue Normal; Larynx Normal; K562 + PMA (36 hrs);

Human Microvascular Endothelial Cells, fract. B; Prostate; Human Pre-Differentiated Adipocytes; Human Thymus Tumor, subtracted; Human Prostate BPH, re-excision;
CD34+cells, II, FRACTION 2; Human Leukocytes; Human osteoarthritis,fraction I;
H Umbilical Vein Endothelial Cells, frac A, re-excision; Human Fetal Lung;
Human 5 epithelioid sarcoma; HL-60, RA 4h, Subtracted; Human Adult Liver, subtracted;
prostate-edited; A1-CELL LINE; L1 Cell line; HUMAN TONSILS, FRACTION 2;
Human Fetal Brain; Human Colon; Human OB HOS treated (10 nM E2) fraction I;
Healing Abdomen wound,70&90 min post incision; Human Pituitary, subtracted;
Human Neutrophils, Activated, re-excision; Aorta endothelial cells + TNF-a;
Invasive 10 poorly differentiated lung adenocarcinoma, metastatic; H. cerebellum, Enzyme subtracted; Human Liver; Stratagene corneal stroma (#937222);
Soares total fetus Nb2HF8 9w; Human Normal Breast; Human Tonsils, Lib 2;
Dendritic Cells From CD34 Cells; Human Hypothalamus,schizophrenia, re-excision;
Human Synovium; Soares_pineal_gland N3HPG; Soares total fetus Nb2HF8 9w;
15 H Female Bladder, Adult; Human Osteoclastoma Stromal Cells - unamplified;
Human Frontal Cortex, Schizophrenia; Salivary Gland, Lib 2; Human Osteoclastoma, re-excision; Pancreas normal PCA4 No; Synovial hypoxia; Stratagene NT2 neuronal precursor 937230; Stratagene endothelial cell 937223; Stratagene pancreas (#937208); Human Infant Brain; H. Kidney Medulla, re-excision; Human Umbilical 20 Vein, Reexcision; STRATAGENE Human skeletal muscle cDNA library, cat.
#936215.; Breast Cancer Cell line, angiogenic; Human Brain, Striatum;
NCI CGAP_Co2; NCI CGAP_GCS; NCI CLAP Prl; NCI CGAP_Pr3;
NCI CGAP_Pr6; NCI CGAP_SS1; NCI CGAP_ColO; NCI CGAP_Col2;
NCI CGAP_Kid6; NCI CGAP_PrlO; NCI CGAP Prl2; NCI CGAP_Pr23;
25 NCI CGAP_Brl.l; Normal Human Trabecular Bone Cells; HMI; Human cerebral cortex; Human pancreatic islet; NCI CGAP_Pr22; NCI CGAP_Pr25;

NCI CGAP_Co4; NCI CGAP_Ewl; NCI CGAP_Lul; NCI CGAP_Pr3;
NCI CGAP_Pr4; NCI CGAP_PrB; NCI CGAP Coll; NCI CGAP_HSC1;
NCI CGAP_Pr4.l; Soares breast 2NbHBst; Jia bone marrow stroma;
Soares testis NHT; NCI CGAP CoB; NCI CGAP_Kids; NCI CGAP_Prl2;
Stratagene hNT neuron (#937233); NCI CGAP Ov6; NCI CGAP PrlO;
NCI CGAP_Pr24; Soares total fetus Nb2HF8 9w; KG1-a Lambda Zap Express cDNA library; Human promyelocyte; Soares fetal heart NbHHI9W; Brain frontal cortex; Barstead pancreas HPLRB1; Human retina cDNA randomly primed sublibrary; Liver HepG2 cell line.; Stratagene cDNA library Human fibroblast, cat#937212; Soares fetal lung_NbHLI9W; NCI CGAP_HN3; NCI CGAP Col2;
HTCDL1; NCI CGAP_CoB; NCI CGAP_GC4; NCI CGAP_LuS;
NCI CGAP_Kids; NCI CGAP_Prl l; NCI CGAP Pr22; NCI CGAP_Pr25;
Stratagene neuroepithelium (#937231); Soares multiple sclerosis 2NbHMSP and Infant brain, LLNL array of Dr. M. Soares 1NIB.
The tissue distribution in fetal/liver spleen and bone marrow cells and tissues indicates the protein product of this clone is 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. The protein, in addition to antogonists and antibodies directed against the present invention, is useful in the treatment, detection, and/or prevention of proliferative diseases and/or disorders.
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.
Many polynucleotide sequences, such as EST sequences, are publicly available and accessible through sequence databases. Some of these sequences are related to SEQ ID N0:16 and may have been publicly available prior to conception of the present invention. Preferably, such related polynucleotides are specifically excluded from the scope of the present invention. To list every related sequence would be cumbersome. Accordingly, preferably excluded from the present invention are one or more polynucleotides comprising a nucleotide sequence described by the general formula of a-b, where a is any integer between 1 to 819 of SEQ ID
N0:16, b is an integer of 15 to 833, where both a and b correspond to the positions of nucleotide residues shown in SEQ ID N0:16, and where b is greater than or equal to a + 14.
FEATURES OF PROTEIN ENCODED BY GENE NO: 7 It has been discovered that this gene is expressed primarily in the following tissues/cDNA libraries: prostate-edited; Human Bone Marrow, re-excision;
Primary Dendritic cells,frac 2.
Many polynucleotide sequences, such as EST sequences, are publicly available and accessible through sequence databases. Some of these sequences are related to SEQ ID N0:17 and may have been publicly available prior to conception of the present invention. Preferably, such related polynucleotides are specifically excluded from the scope of the present invention. To list every related sequence would be cumbersome. Accordingly, preferably excluded from the present invention are one or more polynucleotides comprising a nucleotide sequence described by the general formula of a-b, where a is any integer between 1 to 1213 of SEQ ID
N0:17, b is an integer of 15 to 1227, where both a and b correspond to the positions of nucleotide residues shown in SEQ ID N0:17, and where b is greater than or equal to a + 14.
FEATURES OF PROTEIN ENCODED BY GENE NO: 8 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.
spIP561341ATPK HUMAN (all information available through the recited accession number is incorporated herein by reference) which is described therein as "ATP
SYNTHASE F CHAIN, MITOCHONDRIAL (EC 3.6.1.34).". A partial alignment demonstrating the observed homology is shown immediately below.
>sp~P56134~ATPK HUMAN ATP SYNTHASE F CHAIN, MITOCHONDRIAL (EC 3.6.1.34).
Length = 87 Plus Strand HSPs:
Score = 460 (161.9 bits), Expect = 6.3e-43, P = 6.3e-43 Identities = 87/87 (1000 , Positives = 87/87 (1000 , Frame = +2 Q: 23 ASVVPVKDKKLLEVKLGELPSWILMRDFSPSGIFGAFQRGYYRYYNKYINVKKGSISGIT 202 ASVVPVKDKKLLEVKLGELPSWILMRDFSPSGIFGAFQRGYYRYYNKYINVKKGSISGIT
3O S: 1 ASVVPVKDKKLLEVKLGELPSWILMRDFSPSGIFGAFQRGYYRYYNKYINVKKGSISGIT 60 Q: 203 MVLACYVLFSYSFSYKHLKHERLRKYH 283 MVLACYVLFSYSFSYKHLKHERLRKYH
S: 61 MVLACYVLFSYSFSYKHLKHERLRKYH 87 The segment of spIP561341ATPK_HUMAN that is shown as "S" above is set out in the sequence listing as SEQ ID NO. 129 . 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. 130 which corresponds to the "Q" sequence in the alignment shown above (gaps introduced in a sequence by the computer are, of course, removed).
It has been discovered that this gene is expressed primarily in the following tissues/cDNA libraries: Soares placenta Nb2HP and to a lesser extent in Human Fetal Brain; Early Stage Human Brain; Bone Marrow Cell Line (RS4,11); Soares fetal liver spleen 1NFLS; Soares_parathyroid tumor NbHPA; Macrophage-oxLDL, re-excision; HUMAN B CELL LYMPHOMA; H Macrophage (GM-CSF treated), re-excision; Soares_pregnant uterus NbHPU; HSA 172 Cells; Raji Cells, cyclohexamide treated; Human Heart; Stromal cell TF274;
Soares fetal lung NbHLI9W; Soares fetal heart NbHHI9W;
Hemangiopericytoma; Macrophage (GM-CSF treated); Endothelial-induced;
Soares NhHMPu_S1; Stratagene fibroblast (#937212);
Soares fetal heart NbHHI9W; Human Thymus; Soares_fetal heart_NbHHI9W;
Keratinocyte, lib 3; Brain pons; Supt cells, cyclohexamide treated, subtracted;
Soares fetal lung NbHLI9W; Human B Cell 8866; H. Frontal Cortex, Epileptic;
K562 + PMA (36 hrs),re-excision; Human Colon, subtraction; H. Striatum Depression, subt; stomach cancer (human); Human (HCC) cell line liver (mouse) metastasis, remake; Human Adult Retina; SKIN; Adipocytes,re-excision; HSC172 cells; Frontal lobe,dementia,re-excision; Activated T-cells; Human Liver;
Amniotic Cells - TNF induced; Apoptotic T-cell, re-excision; Human Prostate Cancer, Stage C
fraction; Glioblastoma; Human Stomach,re-excision; Human endometrial stromal cells-treated with progesterone; Human Frontal Cortex, Schizophrenia; wilm's tumor;
5 Stratagene endothelial cell 937223; Brain Frontal Cortex, re-excision;
Breast Cancer Cell line, angiogenic; Human Thymus; Human Brain, Striatum; human ovarian cancer; 12 Week Old Early Stage Human, II; Human Fetal Dura Mater; Human Hypothalmus,Schizophrenia; Spinal cord; Ulcerative Colitis; Human Thymus;
Epithelial-TNFa and INF induced; Human Gall Bladder; 12 Week Old Early Stage 10 Human; Colon Tumor II; human tonsils; Human Adult Pulmonary,re-excision;
Endothelial cells-control; Human Amygdala; Human Microvascular Endothelial Cells, fract. A; Soares fetal lung NbHLI9W; H. Frontal cortex,epileptic,re-excision;
Human 8 Week Whole Embryo and Human Cerebellum.
Ther protein, in addition to antagonists and/or antibodies directed to the 15 invention, is useful for the treatment and/or prevention of proliferative and developmental diseases and/or disorders. 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 20 against the protein may show utility as a tumor marker and/or immunotherapy targets for the above listed tissues.
Many polynucleotide sequences, such as EST sequences, are publicly available and accessible through sequence databases. Some of these sequences are related to SEQ ID N0:18 and may have been publicly available prior to conception of 25 the present invention. Preferably, such related polynucleotides are specifically excluded from the scope of the present invention. To list every related sequence would be cumbersome. Accordingly, preferably excluded from the present invention are one or more polynucleotides comprising a nucleotide sequence described by the general formula of a-b, where a is any integer between 1 to 430 of SEQ ID
N0:18, b is an integer of 15 to 444, where both a and b correspond to the positions of nucleotide residues shown in SEQ ID N0:18, and where b is greater than or equal to a + 14.
FEATURES OF PROTEIN ENCODED BY GENE NO: 9 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.
dbjIBAA24864.11 (all information available through the recited accession number is incorporated herein by reference; for example, Biochem. Biophys. Res. Commun.

(1), 268-274 (1998); in addition, the Geneseq Accession No.W87644 is also incorporated herein by referecence) which is described therein as "aquaporin 9 [Homo Sapiens]". A partial alignment demonstrating the observed homology is shown immediately below.
>dbj~BAA24864.1~ (AB008775) aquaporin 9 [Homo Sapiens] >pir~JC5973~JC5973 aquaporin 9 - Human >sp~043315~AQP9 HUMAN AQUAPORIN 9.
Length = 295 Plus Strand HSPs:
Score = 888 (312.6 bits), Expect = 3.2e-88, P = 3.2e-88 Identities = 174/177 (98~), Positives = 174/177 (98~), Frame = +3 Q: 18 TPLGIXYDGLMSFAGGKLLIVGENATAHIFATYPAPYLSLANAFADQWATMILLIIVFA 197 T GI YDGLMSFAGGKLLIVGENATAHIFATYPAPYLSLANAFADQWATMILLIIVFA
3O S: 119 TVFGIYYDGLMSFAGGKLLIVGENATAHIFATYPAPYLSLANAFADQWATMILLIIVFA 178 Q: 198 IFDSRNLGAPRGLEPIAIGLLIIVIASSLGLNSGCAMNPARDLSPRLFTALAGWGFEVFR 377 IFDSRNLGAPRGLEPIAIGLLIIVIASSLGLNSGCAMNPARDLSPRLFTALAGWGFEVFR
S: 179 IFDSRNLGAPRGLEPIAIGLLIIVIASSLGLNSGCAMNPARDLSPRLFTALAGWGFEVFR 238 Q: 378 AGNNFWWIPWGPLVGAVIGGLIYVLVIEIHHPEPDSVFKAEQSEDKPEKYELSVIM 548 AGNNFWWIPWGPLVGAVIGGLIYVLVIEIHHPEPDSVFKAEQSEDKPEKYELSVIM
S: 239 AGNNFWWIPWGPLVGAVIGGLIYVLVIEIHHPEPDSVFKAEQSEDKPEKYELSVIM 295 The segment of dbjIBAA24864.11 that is shown as "S" above is set out in the sequence listing as SEQ ID NO. 131 . 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. 132 which corresponds to the "Q" sequence in the alignment shown above (gaps introduced in a sequence by the computer are, of course, removed).
It has been discovered that this gene is expressed primarily in the following tissues/cDNA libraries: Hepatocellular Tumor, re-excision and to a lesser extent in Human Neutrophil, Activated; Hepatocellular Tumor,re-excision; Neutrophils control, re-excision; Hepatocellular Tumor; neutrophils control; Macrophage-oxLDL;
Human Liver, normal; Monocyte activated; Neutrophils IL-1 and LPS induced;
Human Gall Bladder; Human Adult Liver, subtracted; Human Neutrophils, Activated, re-excision; Human Activated T-Cells; Liver, Hepatoma; Human Activated T-Cells, re-excision; Human Testes Tumor, re-excision; H Macrophage (GM-CSF treated), re-excision; Crohn's Disease; Activated T-Cells, 8 hrs, subtracted; Human Adult Pulmonary; Human Lung; NCI CGAP_Brn23; pBMC stimulated w/ poly I/C;
NCI CGAP_Li2; Monocyte activated, re-excision; Soares NhHMPu_Sl;
Macrophage-oxLDL, re-excision; Neutrophils IL-1 and LPS induced; Human Fetal Lung III; Human Bone Marrow, treated; Spleen, Chronic lymphocytic leukemia and Human Endometrial Tumor.

Preferred polypeptides of the present invention comprise immunogenic epitopes shown in SEQ ID NO: 68 as residues: Met-1 to Phe-6. Polynucleotides encoding said polypeptides are also encompassed by the invention. Antibodies that bind polypeptides of the invention are encompassed by the invention, particularly antibodies that inhibit activity.
The tissue distribution in immune cells and tissues, combined with the homology to an aquaporin protein, indicates the protein product of this clone is 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 of this gene product indicates a role in regulating the proliferation; survival; differentiation; and/or activation of hematopoietic cell lineages, including blood stem cells. This gene product is involved in the regulation of cytokine production, antigen presentation, or other processes suggesting a usefulness in the treatment of cancer (e.g. by boosting immune responses).
Since the gene is expressed in cells of lymphoid origin, the natural gene product is involved in immune functions. Therefore it is also used 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 S 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.
Many polynucleotide sequences, such as EST sequences, are publicly available and accessible through sequence databases. Some of these sequences are related to SEQ ID N0:19 and may have been publicly available prior to conception of the present invention. Preferably, such related polynucleotides are specifically excluded from the scope of the present invention. To list every related sequence would be cumbersome. Accordingly, preferably excluded from the present invention are one or more polynucleotides comprising a nucleotide sequence described by the general formula of a-b, where a is any integer between 1 to 2371 of SEQ ID
N0:19, b is an integer of 15 to 2385, where both a and b correspond to the positions of nucleotide residues shown in SEQ ID N0:19, and where b is greater than or equal to a + 14.
FEATURES OF PROTEIN ENCODED BY GENE NO: 10 It has been discovered that this gene is expressed primarily in the following tissues/cDNA libraries: Human Cerebellum and to a lesser extent in Soares fetal liver spleen 1NFLS; Human Adrenal Gland Tumor; 12 Week Early Stage Human II, Reexcision; Human Testes Tumor; H. Frontal cortex,epileptic,re-excision; Nine Week Old Early Stage Human; 12 Week Old Early Stage Human; Human Whole Brain #2 -Oligo dT > 1.SKb; 12 Week Old Early Stage Human, II; Human Fetal Brain; Human Fetal Kidney, Reexcision; NCI CGAP_LuS; Soares infant brain 1NIB; H.
cerebellum, Enzyme subtracted; Soares retina N2b4HR; H. Kidney Cortex, subtracted; NCI CGAP_LuS; Human Substantia Nigra; Human 8 Week Whole 5 Embryo; Human Fetal Liver- Enzyme subtraction; Human Fetal Liver, subtracted;
Human 8 Week Whole Embryo, subtracted; Frontal lobe,dementia,re-excision; H.
Whole Brain #2, re-excision; Human Infant Brain; NCI CGAP_AR1; Human Hippocampus; Human Thymus; Human Whole Six Week Old Embryo; Human Placenta; Soares fetal heart NbHHI9W; Human Pediatric Kidney; Brain, normal;
10 Placenta; Human Fetal Liver, subtracted, neg clone; STRIATUM DEPRESSION;
Human Pituitary, re-excision; Human Cerebellum, subtracted; Human Umbilical Vein Endothelial Cells, fract. A; Human Placenta; Human Fetal Bone; Messangial cell, frac 2; Human Whole Brain, re-excision; Soares adult brain N2b4HB55Y; Human Hypothalamus,schizophrenia, re-excision; Alzheimers, spongy change; Human 15 Amygdala,re-excision; Myoloid Progenitor Cell Line; Human Manic Depression Tissue; Soares fetal liver spleen_1NFLS S1; Human Umbilical Vein, Reexcision;
human ovarian cancer; Human Osteoblasts II; NCI CGAP_Ewl; NCI CGAP_Pr4.l;
Human Adult Testes, Large Inserts, Reexcision; Olfactory epithelium,nasalcavity;
Spinal cord; Soares adult brain N2b5HB55Y; Bone Marrow Stromal Cell, untreated;
20 Human Thymus Stromal Cells; Fetal Liver, subtraction II; Human Gall Bladder;
Brain frontal cortex; Human Placenta; Early Stage Human Brain; Human adult (K.Okubo); Human fetal heart, Lambda ZAP Express; Soares NFL T GBC_S1;
Soares NSF F8 9W_OT PA P S1; Soares fetal liver spleen_1NFLS S1;
Stratagene schizo brain S11; Pancreatic Islet; Human Synovial Sarcoma; Human Fetal 25 Lung III; Endothelial-induced; Soares fetal liver spleen_1NFLS S1; Human Microvascular Endothelial Cells, fract. A; NCI CGAP_Brn35 and Soares placenta Nb2HP.
Many polynucleotide sequences, such as EST sequences, are publicly available and accessible through sequence databases. Some of these sequences are related to SEQ ID N0:20 and may have been publicly available prior to conception of the present invention. Preferably, such related polynucleotides are specifically excluded from the scope of the present invention. To list every related sequence would be cumbersome. Accordingly, preferably excluded from the present invention are one or more polynucleotides comprising a nucleotide sequence described by the general formula of a-b, where a is any integer between 1 to 2743 of SEQ ID
N0:20, b is an integer of 15 to 2757, where both a and b correspond to the positions of nucleotide residues shown in SEQ ID N0:20, and where b is greater than or equal to a + 14.
FEATURES OF PROTEIN ENCODED BY GENE NO: 11 It has been discovered that this gene is expressed primarily in the following tissues/cDNA libraries: Soares placenta Nb2HP; Soares infant brain 1NIB and to a lesser extent in Soares_pregnant uterus NbHPU; NCI CGAP_GCBI; Primary Dendritic Cells, lib 1; Soares melanocyte 2NbHM;
Soares multiple sclerosis 2NbHMSP; Human colon carcinoma (HCC) cell line, remake; Stratagene ovary (#937217); pBMC stimulated w/ poly I/C; Stratagene HeLa cell s3 937216; Stratagene fetal spleen (#937205);
Soares senescent fibroblasts NbHSF; Soares testis NHT;
Soares NFL T_GBC Sl; Colon Tumor; Soares_placenta_8to9weeks 2NbHP8to9W;
Human Osteoclastoma; NCI CGAP Br2; NCI CGAP_LuS; Human Bone Marrow, treated; Nine Week Old Early Stage Human; Soares_pregnant uterus NbHPU;

Human Pancreas Tumor; Soares ovary tumor NbHOT; Cheek Carcinoma; Rectum tumour; Human Fetal Spleen; Activated T-cells; Human Lung; Early Stage Human Lung, subtracted; Cem cells cyclohexamide treated; normalized infant brain cDNA;
NTERA2 teratocarcinoma cell line+retinoic acid (14 days); Human Tonsils, Lib 2;
Human endometrial stromal cells; Synovial Fibroblasts (Ill/TNF), subt; Mo7e Cell Line GM-CSF treated (lng/ml); Human Heart; Soares_parathyroid tumor NbHPA;
Soares multiple sclerosis 2NbHMSP; Human umbilical vein endothelial cells, IL-induced; Ulcerative Colitis; Bone Marrow Stromal Cell, untreated; Human colorectal cancer; Soares_pregnant uterus NbHPU; Soares total fetus Nb2HF8 9w; 12 Week Old Early Stage Human; Human T-Cell Lymphoma; Human Placenta; Human Testes Tumor; Endothelial-induced; normalized infant brain cDNA; Anergic T-cell;
NCI CGAP_Co3; NCI CGAP CoB; NCI CGAP_Kids;
Soares multiple sclerosis 2NbHMSP; T cell helper II and Stratagene NT2 neuronal precursor 937230.
Many polynucleotide sequences, such as EST sequences, are publicly available and accessible through sequence databases. Some of these sequences are related to SEQ ID N0:21 and may have been publicly available prior to conception of the present invention. Preferably, such related polynucleotides are specifically excluded from the scope of the present invention. To list every related sequence would be cumbersome. Accordingly, preferably excluded from the present invention are one or more polynucleotides comprising a nucleotide sequence described by the general formula of a-b, where a is any integer between 1 to 1429 of SEQ ID
N0:21, b is an integer of 15 to 1443, where both a and b correspond to the positions of nucleotide residues shown in SEQ ID N0:21, and where b is greater than or equal to a + 14.

FEATURES OF PROTEIN ENCODED BY GENE NO: 12 It has been discovered that this gene is expressed primarily in the following tissues/cDNA libraries: Human Eosinophils; Human Bone Marrow, treated and to a lesser extent in eosinophil-ILS induced; Bone marrow.
Preferred polypeptides of the present invention comprise immunogenic epitopes shown in SEQ ID NO: 71 as residues: Ser-5 to Thr-10, Cys-36 to Glu-51.
Polynucleotides encoding said polypeptides are also encompassed by the invention.
Antibodies that bind polypeptides of the invention are encompassed by the invention, particularly antibodies that inhibit activity.
Many polynucleotide sequences, such as EST sequences, are publicly available and accessible through sequence databases. Some of these sequences are related to SEQ ID N0:22 and may have been publicly available prior to conception of the present invention. Preferably, such related polynucleotides are specifically excluded from the scope of the present invention. To list every related sequence would be cumbersome. Accordingly, preferably excluded from the present invention are one or more polynucleotides comprising a nucleotide sequence described by the general formula of a-b, where a is any integer between 1 to 925 of SEQ ID
N0:22, b is an integer of 15 to 939, where both a and b correspond to the positions of nucleotide residues shown in SEQ ID N0:22, and where b is greater than or equal to a + 14.
FEATURES OF PROTEIN ENCODED BY GENE NO: 13 It has been discovered that this gene is expressed primarily in Synovial hypoxia-RSF subtracted.
Preferred polypeptides of the present invention comprise immunogenic epitopes shown in SEQ ID NO: 72 as residues: Cys-26 to Lys-31. Polynucleotides encoding said polypeptides are also encompassed by the invention. Antibodies that bind polypeptides of the invention are encompassed by the invention, particularly antibodies that inhibit activity.
Many polynucleotide sequences, such as EST sequences, are publicly available and accessible through sequence databases. Some of these sequences are related to SEQ ID N0:23 and may have been publicly available prior to conception of the present invention. Preferably, such related polynucleotides are specifically excluded from the scope of the present invention. To list every related sequence would be cumbersome. Accordingly, preferably excluded from the present invention are one or more polynucleotides comprising a nucleotide sequence described by the general formula of a-b, where a is any integer between 1 to 1846 of SEQ ID
N0:23, b is an integer of 15 to 1860, where both a and b correspond to the positions of nucleotide residues shown in SEQ ID N0:23, and where b is greater than or equal to a + 14.
FEATURES OF PROTEIN ENCODED BY GENE NO: 14 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.
gbIAAA88038.11 (all information available through the recited accession number is incorporated herein by reference) which is described therein as "Homo sapiens protein". A partial alignment demonstrating the observed homology is shown immediately below.
>gb~AAA88038.1~ unknown protein [Homo Sapiens] >sp~Q14288~Q14288 HYPOTHETICAL
PROTEIN (FRAGMENT).
Length = 641 Minus Strand HSPs:
Score = 447 (157.4 bits), Expect = 1.8e-41, P = 1.8e-41 Identities = 82/86 (958), Positives = 82/86 (95$), Frame = -1 Q: 969 KDTCTRMFIXALFTIAKTWNQPKCPTMIDWIKKMWHIYTMEYYAAIKNDEFMSFVGTWMK 790 KDTCTRMFI ALFTIAKTWNQPKCPTMIDWIKKMWHIYTMEYYAAIKNDEFMSFVGTWMK
S: 556 KDTCTRMFIAALFTIAKTWNQPKCPTMIDWIKKMWHIYTMEYYAAIKNDEFMSFVGTWMK 615 1O Q: 789 LEXIILSKLSQXQKTKHRXFSLIGGN 712 LE IILSKLSQ QKTKHR FSLIGGN
S: 616 LETIILSKLSQEQKTKHRIFSLIGGN 641 The segment of gbIAAA88038.11 that is shown as "S" above is set out in the 15 sequence listing as SEQ ID NO. 133 . 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.
20 Preferred polypeptides of the invention comprise a polypeptide having the amino acid sequence set out in the sequence listing as SEQ ID NO. 134 which corresponds to the "Q" sequence in the alignment shown above (gaps introduced in a sequence by the computer are, of course, removed).
It has been discovered that this gene is expressed primarily in the following 25 tissues/cDNA libraries: Soares_parathyroid tumor NbHPA and to a lesser extent in Neutrophils control, re-excision; 12 Week Old Early Stage Human; Stratagene colon (#937204); Soares_parathyroid_tumor NbHPA; Neutrophils IL-1 and LPS induced;
neutrophils control; NCI CGAP_GCB1; Human Eosinophils; NCI CGAP_Lu26;
Human Cerebellum; Soares fetal liver spleen 1NFLS; human ovarian cancer; PC3 30 Prostate cell line; Colon Tumor; Colon Normal II; Soares melanocyte 2NbHM;
Human Neutrophil, Activated; Monocyte activated; Human Uterus, normal; SKIN;
Brain-medulloblastoma; Human Fetal Spleen; Fetal Heart, re-excision; Human fetal heart, Lambda ZAP Express; Stomach cancer (human),re-excision; Human Frontal Cortex, Schizophrenia; NCI CGAP_Prl; NCI CGAP_GCB1; NCI CGAP_Pr4.l;
Monocyte activated, re-excision; Stromal cell TF274; Soares fetal heart_NbHHI9W;
Soares fetal heart_NbHHI9W; Soares_pregnant uterus NbHPU;
Soares total fetus Nb2HF8 9w; normalized infant brain cDNA; NCI CGAP_Ov2;
NCI CGAP_Kid3; Neutrophils IL-1 and LPS induced; Stratagene fetal retina 937202; Stratagene lung carcinoma 937218; Brain frontal cortex; HM1;
Soares fetal lung NbHLI9W; Anergic T-cell; Spleen, Chronic lymphocytic leukemia; Nine Week Old Early Stage Human; NCI CGAP_Kid3;
NCI CGAP_Ov32; T cell helper II; Bone Cancer, re-excision; Liver, normal;
Whole brain; Soares ovary tumor NbHOT; H. Male bladder, adult; human pleural cancer;
Normal Ovary, Premenopausal; Human Fibrosarcoma; Human Fetal Brain, random primed; prostate-edited; stomach cancer (human); Adipocytes,re-excision; Human Placenta; Human Liver; Clontech human aorta polyA+ mRNA (#6572);
NCI CGAP_Pr22; Soares retina N2b4HR; Soares fetal heart- NbHHI9W;
Stratagene colon (#937204); Stratagene pancreas (#937208); Human Normal Breast;
Human Tonsils, Lib 2; Healing groin wound - zero hr post-incision (control);
Human Synovium; Stratagene placenta (#937225); normalized infant brain cDNA; Human Stomach,re-excision; Human Adipose Tissue, re-excision; Human Osteosarcoma;
Pancreas normal PCA4 No; Prostate BPH; TF-1 Cell Line GM-CSF Treated; KMH2;
NCI CGAP_Br2; NCI CGAP_Co3; NCI CGAP PrB; NCI CGAP_Col l;
NCI CGAP_Brl.l; Human Fetal Dura Mater; Human Activated T-Cells; Human Pancreas Tumor; Human Hippocampus; Human Chondrosarcoma; PERM TF274;
Soares_parathyroid tumor NbHPA; Human Testes Tumor, re-excision; Atrium cDNA library Human heart; Infant brain, Bento Soares; Soares testis NHT;
Stratagene lung (#937210); Stratagene lung carcinoma 937218; Stratagene liver (#937224); Human Gall Bladder; NCI CGAP AA1; NCI CGAP_HN3;
NCI CGAP_LuS; NCI CGAP_Lu6; NCI CGAP_Pr3; NCI CGAP_Kids;
NCI CGAP_Kid6; NCI CGAP_Ov26; Human Ovarian Cancer Reexcision; Human T-Cell Lymphoma; Stratagene endothelial cell 937223; Stratagene neuroepithelium NT2RAMI 937234; Colon Carcinoma; Human fetal heart, Lambda ZAP Express;
Pancreatic Islet; Soares senescent fibroblasts_NbHSF;
Soares_placenta 8to9weeks_2NbHP8to9W; Human Fetal Heart; Human Microvascular Endothelial Cells, fract. A; T Cell helper I; H. Frontal cortex,epileptic,re-excision; Activated T-cell(12h)/Thiouridine-re-excision;
NCI CGAP_AAI; NCI CGAP HN4; NCI CGAP_LuS; NCI CGAP_Ut3;
NCI CGAP_Brl6; NCI CGAP_Eso2; NCI CGAP HSC2; NCI CGAP Ov31;
NCI CGAP_Ov34; Soares infant brain 1NIB and Soares fetal liver spleen_1NFLS S1.
Many polynucleotide sequences, such as EST sequences, are publicly available and accessible through sequence databases. Some of these sequences are related to SEQ ID N0:24 and may have been publicly available prior to conception of the present invention. Preferably, such related polynucleotides are specifically excluded from the scope of the present invention. To list every related sequence would be cumbersome. Accordingly, preferably excluded from the present invention are one or more polynucleotides comprising a nucleotide sequence described by the general formula of a-b, where a is any integer between 1 to 1063 of SEQ ID
N0:24, b is an integer of 15 to 1077, where both a and b correspond to the positions of nucleotide residues shown in SEQ ID N0:24, and where b is greater than or equal to a + 14.

FEATURES OF PROTEIN ENCODED BY GENE NO: 15 It has been discovered that this gene is expressed primarily in Brain frontal cortex.
Preferred polypeptides of the present invention comprise immunogenic epitopes shown in SEQ ID NO: 74 as residues: Gly-60 to Asp-65. Polynucleotides encoding said polypeptides are also encompassed by the invention. Antibodies that bind polypeptides of the invention are encompassed by the invention, particularly antibodies that inhibit activity.
Many polynucleotide sequences, such as EST sequences, are publicly available and accessible through sequence databases. Some of these sequences are related to SEQ ID N0:25 and may have been publicly available prior to conception of the present invention. Preferably, such related polynucleotides are specifically excluded from the scope of the present invention. To list every related sequence would be cumbersome. Accordingly, preferably excluded from the present invention are one or more polynucleotides comprising a nucleotide sequence described by the general formula of a-b, where a is any integer between 1 to 1365 of SEQ ID
N0:25, b is an integer of 15 to 1379, where both a and b correspond to the positions of nucleotide residues shown in SEQ ID N0:25, and where b is greater than or equal to a + 14.
FEATURES OF PROTEIN ENCODED BY GENE NO: 16 It has been discovered that this gene is expressed primarily in the following tissues/cDNA libraries: H. Epididiymus, cauda; Brain Frontal Cortex, re-excision.
Many polynucleotide sequences, such as EST sequences, are publicly available and accessible through sequence databases. Some of these sequences are related to SEQ ID N0:26 and may have been publicly available prior to conception of the present invention. Preferably, such related polynucleotides are specifically excluded from the scope of the present invention. To list every related sequence would be cumbersome. Accordingly, preferably excluded from the present invention are one or more polynucleotides comprising a nucleotide sequence described by the general formula of a-b, where a is any integer between 1 to 726 of SEQ ID
N0:26, b is an integer of 15 to 740, where both a and b correspond to the positions of nucleotide residues shown in SEQ ID N0:26, and where b is greater than or equal to a + 14.
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.
dbjIBAA13387.11 (all information available through the recited accession number is incorporated herein by reference) which is described therein as "Similar to a C.elegans protein encoded in cosmid C27F2 (U40419) [Homo Sapiens]". A partial alignment demonstrating the observed homology is shown immediately below.
>dbj~BAA13387.1~ Similar to a C.elegans protein encoded in cosmid C27F2 (U40419) [Homo Sapiens] >sp~Q92545~Q92545 MYELOBLAST KIAA0257 (FRAGMENT).
Length = 1805 Plus Strand HSPs:
Score = 1221 (429.8 bits), Expect = 1.2e-208, Sum P(3) = 1.2e-208 Identities = 253/366 (69$), Positives = 256/366 (69~), Frame = +1 Q: 1303 APLAHPSHPERASSARHSSEDSDITSLIEAMDKDFDHHDSPALEVFTEQXXXXXXXXXXX 1482 S: 1246 APLAHPSHPERASSARHSSEDSDITSLIEAMDKDFDHHDSPALEVFTEQPPSPLPKSKGK 1305 Q: 1483 XXXXQRKVXXXXXXXXXXXXXXXXXXXDELKXXXXXXXXXXXXXXXXXXXXEPLLKEDTE 1662 QRKV DELK EPLLKEDTE
3S S: 1306 GKPLQRKVKPPKKQEEKEKKGKGKPQEDELKDSLADDDSSSTTTETSNPDTEPLLKEDTE 1365 Q: 1663 KQKGKQAMPEKHESEMSQVXXXXXXXXXXXXEIPTDVKPSSLELPYTPPLESKQRRNLPS 1842 KQKGKQAMPEKHESEMSQV EIPTDVKPSSLELPYTPPLESKQRRNLPS
S: 1366 KQKGKQAMPEKHESEMSQVKQKSKKLLNIKKEIPTDVKPSSLELPYTPPLESKQRRNLPS 1425 S Q: 1843 KIPLPTAMTSGSKSRNAQKTKGTSKLVDNRPPALAKFLPNSQELGNTSSSEGEKDSPPPE 2022 KIPLPTAMTSGSKSRNAQKTKGTSKLVDNRPPALAKFLPNSQELGNTSSSEGEKDSPPPE
S: 1426 KIPLPTAMTSGSKSRNAQKTKGTSKLVDNRPPALAKFLPNSQELGNTSSSEGEKDSPPPE 1485 Q: 2023 WDSVPVHKPGSSTDSLYKLSLQTLNADIFLKQRQXXXXXXXXXXXXXXCPFVARGXXXXX 2202 IO WDSVPVHKPGSSTDSLYKLSLQTLNADIFLKQRQ CPFVARG
S: 1486 WDSVPVHKPGSSTDSLYKLSLQTLNADIFLKQRQTSPTPASPSPPAAPCPFVARGSYSSI 1545 Q: 2203 XXXXXXXDPKIKQPNGSKHKLTKAASLPGKNGNPXFAAXTAXLRQEPRXGMALLKVSXKQ 2382 DPKIKQPNGSKHKLTKAASLPGKNGNP FAA TA + P G KVS +
IS S: 1546 VNSSSSSDPKIKQPNGSKHKLTKAASLPGKNGNPTFAAVTAGYDKSP-GGNGFAKVSSNK 1604 Q: 2383 NRFXXA 2400 F +
S: 1605 TGFSSS 1610 The segment of dbjIBAA13387.11 that is shown as "S" above is set out in the sequence listing as SEQ ID NO. 13S . 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.
2S 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. 136 which corresponds to the "Q" sequence in the alignment shown above (gaps introduced in a sequence by the computer are, of course, removed).
It has been discovered that this gene is expressed primarily in the following tissues/cDNA libraries: Human Umbilical Vein, Endo. remake and to a lesser extent in Smooth muscle, serum treated; Human Placenta; Human 8 Week Whole Embryo;
Soares infant brain 1NIB; Stratagene neuroepithelium (#937231); normalized infant 3S brain cDNA; Human Pre-Differentiated Adipocytes; Weizmann Olfactory Epithelium; Human Tonsils, Lib 2; Human Pancreas Tumor; Human Placenta (re-excision); Ulcerative Colitis; Bone Marrow Stromal Cell, untreated; Rejected Kidney, lib 4; Human Whole Six Week Old Embryo; PC3 Prostate cell line; Human Testes Tumor; Dendritic cells, pooled; Human Testes, Reexcision; Human Fetal Heart;
Osteoblasts; NCI CGAP_CLLl; Keratinocyte and Nine Week Old Early Stage Human.
Preferred polypeptides of the present invention comprise immunogenic epitopes shown in SEQ ID NO: 76 as residues: Ala-31 to Val-38, Pro-63 to Gly-68, Gly-74 to Cys-87, Pro-93 to Asp-104, Arg-109 to Ser-121, Gln-128 to Thr-133, Thr-148 to Ser-163, Pro-170 to Leu-179, Val-183 to Glu-188. Polynucleotides encoding said polypeptides are also encompassed by the invention. Antibodies that bind polypeptides of the invention are encompassed by the invention, particularly antibodies that inhibit activity.
Many polynucleotide sequences, such as EST sequences, are publicly available and accessible through sequence databases. Some of these sequences are related to SEQ ID N0:27 and may have been publicly available prior to conception of the present invention. Preferably, such related polynucleotides are specifically excluded from the scope of the present invention. To list every related sequence would be cumbersome. Accordingly, preferably excluded from the present invention are one or more polynucleotides comprising a nucleotide sequence described by the general formula of a-b, where a is any integer between 1 to 2429 of SEQ ID
N0:27, b is an integer of 15 to 2443, where both a and b correspond to the positions of nucleotide residues shown in SEQ ID N0:27, and where b is greater than or equal to a + 14.

FEATURES OF PROTEIN ENCODED BY GENE NO: 18 It has been discovered that this gene is expressed primarily in the following tissues/cDNA libraries: PERM TF274 and to a lesser extent in Soares senescent fibroblasts NbHSF; Human Liver, normal; normalized infant brain cDNA and Fetal Heart.
Preferred polypeptides of the present invention comprise immunogenic epitopes shown in SEQ ID NO: 77 as residues: Met-1 to Arg-8. Polynucleotides encoding said polypeptides are also encompassed by the invention. Antibodies that bind polypeptides of the invention are encompassed by the invention, particularly antibodies that inhibit activity.
Many polynucleotide sequences, such as EST sequences, are publicly available and accessible through sequence databases. Some of these sequences are related to SEQ ID N0:28 and may have been publicly available prior to conception of the present invention. Preferably, such related polynucleotides are specifically excluded from the scope of the present invention. To list every related sequence would be cumbersome. Accordingly, preferably excluded from the present invention are one or more polynucleotides comprising a nucleotide sequence described by the general formula of a-b, where a is any integer between 1 to 1957 of SEQ ID
N0:28, b is an integer of 15 to 1971, where both a and b correspond to the positions of nucleotide residues shown in SEQ ID N0:28, and where b is greater than or equal to a + 14.
FEATURES OF PROTEIN ENCODED BY GENE NO: 19 The computer algorithm BLASTX has been used to determine that the translation product of this gene shares sequence homology with, as a non-liW
iting example, the sequence accessible through the following database accession no.

gbIAAA88036.11 (all information available through the recited accession number is incorporated herein by reference) which is described therein as "Homo Sapiens protein". A partial alignment demonstrating the observed homology is shown immediately below.
' >gb~AAA88036.1~ unknown protein [Homo sapiens] >sp~Q14287~Q14287 HYPOTHETICAL

PROTEIN (FRAGMENT).
Length = 157 Minus Strand HSPs:
Score = 147 (51.7 bits), Expect = 1.1e-08, P = 1.1e-08 Identities = 33/55 (60$), Positives = 37/55 (67$), Frame = -2 Q: 1968 LLGIYPQKMKSIC*RDI*TLMIIARLFTIAKIGNQPKYPSTDEEIKIM*HIYTTE 1804 LLGIYP + KS C +D T M IA LFTIAK NQPK P+ + IK M HIYT E
S: 58 LLGIYPNEYKSCCYKDTCTRMFIAALFTIAKTWNQPKCPTMIDWIKKMWHIYTME 112 The segment of gbIAAA88036.11 that is shown as "S" above is set out in the sequence listing as SEQ ID NO. 137 . 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. 138, 163, and/or 170 which corresponds to the "Q" sequence in the alignment shown above (gaps introduced in a sequence by the computer are, of course, removed).
It has been discovered that this gene is expressed primarily in the following tissues/cDNA libraries: Soares fetal liver spleen 1NFLS; Human Hippocampus;
Human Fetal Lung III.

Preferred polypeptides of the present invention comprise immunogenic epitopes shown in SEQ ID NO: 78 as residues: Gly-25 to Asn-34. Polynucleotides encoding said polypeptides are also encompassed by the invention. Antibodies that bind polypeptides of the invention are encompassed by the invention, particularly antibodies that inhibit activity.
Many polynucleotide sequences, such as EST sequences, are publicly available and accessible through sequence databases. Some of these sequences are related to SEQ ID N0:29 and may have been publicly available prior to conception of the present invention. Preferably, such related polynucleotides are specifically excluded from the scope of the present invention. To list every related sequence would be cumbersome. Accordingly, preferably excluded from the present invention are one or more polynucleotides comprising a nucleotide sequence described by the general formula of a-b, where a is any integer between 1 to 2027 of SEQ ID
N0:29, b is an integer of 15 to 2041, where both a and b correspond to the positions of nucleotide residues shown in SEQ ID N0:29, and where b is greater than or equal to a + 14.
FEATURES OF PROTEIN ENCODED BY GENE NO: 20 It has been discovered that this gene is expressed primarily in the following tissues/cDNA libraries: Soares NhHMPu_S1 and to a lesser extent in Soares placenta Nb2HP; Soares melanocyte 2NbHM; NCI CGAP_Co3;
Soares_pregnant uterus NbHPU; Soares fetal liver spleen 1 NFLS; STRIATUM
DEPRESSION; Activated T-cells; Human Amygdala,re-excision; Jurkat T-cell G1 phase; Fetal brain, Stratagene; NCI CGAP_Thyl; Soares fetal heart NbHHI9W;
Soares_parathyroid tumor NbHPA; Stratagene ovarian cancer (#937219); H.

Meningima, M1; Human Brain, Striatum; Hemangiopericytoma;
Soares fetal liver spleen_1NFLS S1 and Human 8 Week Whole Embryo.
Many polynucleotide sequences, such as EST sequences, are publicly available and accessible through sequence databases. Some of these sequences are 5 related to SEQ ID N0:30 and may have been publicly available prior to conception of the present invention. Preferably, such related polynucleotides are specifically excluded from the scope of the present invention. To list every related sequence would be cumbersome. Accordingly, preferably excluded from the present invention are one or more polynucleotides comprising a nucleotide sequence described by the 10 general formula of a-b, where a is any integer between 1 to 2004 of SEQ ID
N0:30, b is an integer of 15 to 2018, where both a and b correspond to the positions of nucleotide residues shown in SEQ ID N0:30, and where b is greater than or equal to a + 14.
15 FEATURES OF PROTEIN ENCODED BY GENE NO: 21 It has been discovered that this gene is expressed primarily in the following tissues/cDNA libraries: CD34 depleted Buffy Coat (Cord Blood), re-excision;
Activated T-cell(12h)/Thiouridine-re-excision and to a lesser extent in LNCAP
+
o.3nM 81881; Human Prostate Cancer, Stage C fraction; Activated T-Cell 20 (l2hs)/Thiouridine labelledEco; Monocyte activated; Soares NFL T GBC_S1 and HUMAN B CELL LYMPHOMA.
Many polynucleotide sequences, such as EST sequences, are publicly available and accessible through sequence databases. Some of these sequences are related to SEQ ID N0:31 and may have been publicly available prior to conception of 25 the present invention. Preferably, such related polynucleotides are specifically excluded from the scope of the present invention. To list every related sequence would be cumbersome. Accordingly, preferably excluded from the present invention are one or more polynucleotides comprising a nucleotide sequence described by the general formula of a-b, where a is any integer between 1 to 2114 of SEQ ID
N0:31, b is an integer of 15 to 2128, where both a and b correspond to the positions of nucleotide residues shown in SEQ ID N0:31, and where b is greater than or equal to a + 14.
FEATURES OF PROTEIN ENCODED BY GENE NO: 22 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.
embICAA05867.11 (all information available through the recited accession number is incorporated herein by reference) which is described therein as "doublecortin [Homo sapiens]". A partial alignment demonstrating the observed homology is shown immediately below.
>emb~CAA05867.1~ (AJ003112) doublecortin [Homo Sapiens] >embICAA06617.1~
(AJ005592) doublecortin [Homo Sapiens] >gb~AAC31696.1~
(AF040255) doublecortin; lissencephalin-X; LISX [Homo Sapiens]
>gb~AAC52037.1 (AF034634) doublecortin [Homo Sapiens] >sp~043911~043911 LISSENCEPHALIN-X. >emb~CAA19966.11 (AL031117) DCX
(doublecortin) [Homo Sapiens] {SUB 236-360}
Length = 360 Plus Strand HSPs:
Score = 173 (60.9 bits), Expect = 1.6e-09, P = 1.6e-09 Identities = 31/35 (88$), Positives = 32/35 (91~), Frame = +2 Q: 650 QVTCLQDFFGDDDVFIACGPEKFRYAQDDFVLDHS 754 QVTCL DFFGDDDVFIACGPEKFRYAQDDF LD +
3S S: 235 QVTCLHDFFGDDDVFIACGPEKFRYAQDDFSLDEN 269 The segment of embICAA05867.11 that is shown as "S" above is set out in the sequence listing as SEQ ID NO. 139 . 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. 140 which corresponds to the "Q" sequence in the alignment shown above (gaps introduced in a sequence by the computer are, of course, removed).
It has been discovered that this gene is expressed primarily in the following tissues/cDNA libraries: Soares retina N2b5HR; Monocyte activated; Soares fetal liver spleen 1NFLS and to a lesser extent in Morton Fetal Cochlea; Stratagene placenta (#937225); Dendritic cells, pooled; CD34 depleted Buffy Coat (Cord Blood), re-excision; Soares_pregnant uterus NbHPU; Human Bone Marrow, treated; Spleen, Chronic lymphocytic leukemia and neutrophils control.
Many polynucleotide sequences, such as EST sequences, are publicly available and accessible through sequence databases. Some of these sequences are related to SEQ ID N0:32 and may have been publicly available prior to conception of the present invention. Preferably, such related polynucleotides are specifically excluded from the scope of the present invention. To list every related sequence would be cumbersome. Accordingly, preferably excluded from the present invention are one or more polynucleotides comprising a nucleotide sequence described by the general formula of a-b, where a is any integer between 1 to 1583 of SEQ ID
N0:32, b is an integer of 15 to 1597, where both a and b correspond to the positions of nucleotide residues shown in SEQ ID N0:32, and where b is greater than or equal to a + 14.
FEATURES OF PROTEIN ENCODED BY GENE NO: 23 It has been discovered that this gene is expressed primarily in the following tissues/cDNA libraries: NCI CGAP GC2 and to a lesser extent in H. Whole Brain #2, re-excision; Human Hypothalamus,schizophrenia, re-excision; Human umbilical vein endothelial cells, IL-4 induced; Bone Marrow Stromal Cell, untreated;
breast lymph node CDNA library; Early Stage Human Brain; NCI CGAP_CoB;
NCI CGAP_GCS; NCI CGAP_Kid3; NCI CGAP_Brn35 and Colon Tumor II.
Preferred polypeptides of the present invention comprise immunogenic epitopes shown in SEQ ID NO: 82 as residues: Pro-39 to Tyr-45, Pro-47 to Pro-54.
Polynucleotides encoding said polypeptides are also encompassed by the invention.
Antibodies that bind polypeptides of the invention are encompassed by the invention, particularly antibodies that inhibit activity.
Many polynucleotide sequences, such as EST sequences, are publicly available and accessible through sequence databases. Some of these sequences are related to SEQ ID N0:33 and may have been publicly available prior to conception of the present invention. Preferably, such related polynucleotides are specifically excluded from the scope of the present invention. To list every related sequence would be cumbersome. Accordingly, preferably excluded from the present invention are one or more polynucleotides comprising a nucleotide sequence described by the general formula of a-b, where a is any integer between 1 to 1458 of SEQ ID
N0:33, b is an integer of 15 to 1472, where both a and b correspond to the positions of nucleotide residues shown in SEQ ID N0:33, and where b is greater than or equal to a + 14.

FEATURES OF PROTEIN ENCODED BY GENE NO: 24 It has been discovered that this gene is expressed primarily in the following tissues/cDNA libraries: neutrophils control and to a lesser extent in NCI CGAP_GCB1; Breast Lymph node cDNA library; CD34 depleted Buffy Coat (Cord Blood), re-excision; Soares placenta Nb2HP; Human Primary Breast Cancer;
Prostate,BPH, Lib 2; Infant brain, Bento Soares; NCI CGAP_Lul; Larynx Normal;
prostate-edited; Breast Lymph node cDNA library; Jurkat T-Cell, S phase; Human Thymus; Human Adult Testes, Large Inserts, Reexcision; Human Thymus; Fetal Liver, subtraction II; Human Eosinophils and Human Neutrophil, Activated.
Preferred polypeptides of the present invention comprise immunogenic epitopes shown in SEQ ID NO: 83 as residues: Ser-21 to Ser-39, Gln-45 to Gln-61, Cys-124 to Gly-130. Polynucleotides encoding said polypeptides are also encompassed by the invention. Antibodies that bind polypeptides of the invention are encompassed by the invention, particularly antibodies that inhibit activity.
Many polynucleotide sequences, such as EST sequences, are publicly available and accessible through sequence databases. Some of these sequences are related to SEQ ID N0:34 and may have been publicly available prior to conception of the present invention. Preferably, such related polynucleotides are specifically excluded from the scope of the present invention. To list every related sequence would be cumbersome. Accordingly, preferably excluded from the present invention are one or more polynucleotides comprising a nucleotide sequence described by the general formula of a-b, where a is any integer between 1 to 1397 of SEQ ID
N0:34, b is an integer of 15 to 1411, where both a and b correspond to the positions of nucleotide residues shown in SEQ ID N0:34, and where b is greater than or equal to a + 14.

FEATURES OF PROTEIN ENCODED BY GENE NO: 25 It has been discovered that this gene is expressed primarily in neutrophils control .
5 Many polynucleotide sequences, such as EST sequences, are publicly available and accessible through sequence databases. Some of these sequences are related to SEQ ID N0:35 and may have been publicly available prior to conception of the present invention. Preferably, such related polynucleotides are specifically excluded from the scope of the present invention. To list every related sequence 10 would be cumbersome. Accordingly, preferably excluded from the present invention are one or more polynucleotides comprising a nucleotide sequence described by the general formula of a-b, where a is any integer between 1 to 1086 of SEQ ID
N0:35, b is an integer of 15 to 1100, where both a and b correspond to the positions of nucleotide residues shown in SEQ ID N0:35, and where b is greater than or equal to a 15 + 14.
FEATURES OF PROTEIN ENCODED BY GENE NO: 26 The computer algorithm BLASTX has been used to determine that the translation product of this gene shares sequence homology with, as a non-limiting 20 example, the sequence accessible through the following database accession no.
dbjIBAA22931.11 (all information available through the recited accession number is incorporated herein by reference) which is described therein as "CCA2 protein [Rattus norvegicus]". A partial alignment demonstrating the observed homology is shown immediately below.
>dbj~BAA22931.1~ (AB000199) CCA2 protein [Rattus norvegicus]
>sp~035048~035048 CCA2 PROTEIN.

Length = 338 Plus Strand HSPs:
Score = 544 (191.5 bits), Expect = 8.6e-52, P = 8.6e-52 Identities = 103/156 (66~), Positives = 109/156 (69$), Frame = +1 Q: 10 GGWLFRAIPASVEHGRVYVGNVAWMHVXXXXXXXXXXXXMGGQVYFCYDGSPYRSYEDFN 189 GG LFRAIPASVEHGRVYVGNVAWMH+ MGGQVYFCYD SPY+SYEDFN
1O S: 182 GGRLFRAIPASVEHGRVYVGNVAWMHILVARELEQRAALMGGQVYFCYDKSPYKSYEDFN 241 Q: 190 MEFLGPCGLRLVGARPLLPYWXXXXXXXXXXXXXXXXXXXXXXXXXXNPYTLAVANTTFT 369 MEFL PCGLRL+G PLLPYW NPYTLAVANTTFT
S: 242 MEFLSPCGLRLIGTHPLLPYWLLVLLTALNALLQWLLRPLVLYTPLLNPYTLAVANTTFT 301 Q: 370 VSTDKAQRHFGYEPLFSWEDSRTRTILWVQAATGSA 477 VST+KAQRHFGY+PLFSWE+SR RTI WVQA GSA
S: 302 VSTNKAQRHFGYKPLFSWEESRARTIHWVQAMEGSA 337 The segment of dbjIBAA22931.11 that is shown as "S" above is set out in the sequence listing as SEQ ID NO. 141 . 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. 142 which corresponds to the "Q" sequence in the alignment shown above (gaps introduced in a sequence by the computer are, of course, removed).
It has been discovered that this gene is expressed primarily in the following tissues/cDNA libraries: Soares placenta Nb2HP and to a lesser extent in Soares fetal heart_NbHHI9W; normalized infant brain cDNA; LNCAP untreated;
Human Primary Breast Cancer,re-excision; Human Synovium; Human endometrial stromal cells-treated with estradiol; Pancreatic Islet; Human Fetal Epithelium (Skin);
Stratagene HeLa cell s3 937216; Synovial hypoxia-RSF subtracted; KMH2; Human Uterine Cancer; Macrophage-oxLDL; Human Rhabdomyosarcoma;
Hemangiopericytoma; Macrophage (GM-CSF treated); Human Adult Pulmonary,re-excision; Hodgkin's Lymphoma II; neutrophils control and Human Cerebellum.
Preferred polypeptides of the present invention comprise immunogenic epitopes shown in SEQ ID NO: 85 as residues: Asp-10 to Asn-21, Thr-84 to Gly-92, Glu-100 to Arg-105. Polynucleotides encoding said polypeptides are also encompassed by the invention. Antibodies that bind polypeptides of the invention are encompassed by the invention, particularly antibodies that inhibit activity.
Many polynucleotide sequences, such as EST sequences, are publicly available and accessible through sequence databases. Some of these sequences are related to SEQ ID N0:36 and may have been publicly available prior to conception of the present invention. Preferably, such related polynucleotides are specifically excluded from the scope of the present invention. To list every related sequence would be cumbersome. Accordingly, preferably excluded from the present invention are one or more polynucleotides comprising a nucleotide sequence described by the general formula of a-b, where a is any integer between 1 to 1459 of SEQ ID
N0:36, b is an integer of 15 to 1473, where both a and b correspond to the positions of nucleotide residues shown in SEQ ID N0:36, and where b is greater than or equal to a + 14.
FEATURES OF PROTEIN ENCODED BY GENE NO: 27 It has been discovered that this gene is expressed primarily in the following tissues/cDNA libraries: normalized infant brain cDNA and to a lesser extent in Activated T-cell(12h)/Thiouridine-re-excision; Keratinocyte; Human Cerebellum;
H
Macrophage (GM-CSF treated), re-excision; Soares melanocyte 2NbHM; Nine Week Old Early Stage Human; Soares fetal liver spleen 1NFLS; Raji cells, cyclohexamide treated, subtracted; Saos2 Cells, Untreated; Smooth muscle, ILlb induced; CHME
Cell Line,treated 5 hrs; Stratagene HeLa cell s3 937216; Fetal Liver, subtraction II;
Ovarian Tumor 10-3-95; Adipocytes; Soares total fetus Nb2HF8 9w; Activated T-Cell (l2hs)/Thiouridine labelledEco; Osteoblasts; Human 8 Week Whole Embryo;
Prostate; H.Leukocytes, normalized cot SOA3; Human Umbilical Vein Endothelial Cells, fract. B; Normal Prostate; Tongue Tumour; Bone Cancer; H. Frontal Cortex, Epileptic; Supt Cells, cyclohexamide treated; Smooth muscle, control, re-excision;
Smooth muscle-ILb induced; Aorta endothelial cells + TNF-a; H. cerebellum, Enzyme subtracted; Amniotic Cells - TNF induced; Soares retina N2b4HR; Cem cells cyclohexamide treated; Apoptotic T-cell, re-excision; B Cell lymphoma;
Hepatocellular Tumor; Human endometrial stromal cells; Synovial Fibroblasts (Ill/TNF), subt; Human Prostate; Human Bone Marrow, re-excision;
Soares total fetus Nb2HF8 9w; KMH2; HUMAN JURKAT MEMBRANE
BOUND POLYSOMES; Human Fetal Dura Mater; Human Placenta (re-excision);
Epithelial-TNFa and INF induced; Human fetal heart, Lambda ZAP Express;
Stratagene fibroblast (#937212); Human Testes Tumor, re-excision; Bone Marrow Stromal Cell, untreated; Human Fetal Brain; Stratagene ovarian cancer (#937219);
Macrophage (GM-CSF treated); Soares_parathyroid tumor NbHPA;
Soares senescent fibroblasts NbHSF; Stratagene lung (#937210); Smooth muscle, serum induced,re-exc; Macrophage-oxLDL, re-excision; PC3 Prostate cell line;
Human T-Cell Lymphoma; Soares NhHMPu_S1; Human Fetal Kidney, Reexcision;
Human Synovial Sarcoma; Human Fetal Lung III; Monocyte activated; Neutrophils IL-1 and LPS induced; Bone Marrow Cell Line (RS4,11); NCI CGAP_Co3;
NCI CGAP_Co9; NCI CGAP_GCB1; NCI CGAP_Kid3; Hodgkin's Lymphoma II;
Soares NhHMPu_S1; Stratagene NT2 neuronal precursor 937230; T cell helper II;
Primary Dendritic Cells, lib 1 and Soares infant brain 1NIB.

Many polynucleotide sequences, such as EST sequences, are publicly available and accessible through sequence databases. Some of these sequences are related to SEQ ID N0:37 and may have been publicly available prior to conception of the present invention. Preferably, such related polynucleotides are specifically excluded from the scope of the present invention. To list every related sequence would be cumbersome. Accordingly, preferably excluded from the present invention are one or more polynucleotides comprising a nucleotide sequence described by the general formula of a-b, where a is any integer between 1 to 939 of SEQ ID
N0:37, b is an integer of 15 to 953, where both a and b correspond to the positions of nucleotide residues shown in SEQ ID N0:37, and where b is greater than or equal to a + 14.
FEATURES OF PROTEIN ENCODED BY GENE NO: 28 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.
gbIAAD00107.11 (all information available through the recited accession number is incorporated herein by reference) which is described therein as "Tic [Homo sapiens]".
A partial alignment demonstrating the observed homology is shown immediately below.
>gb~AAD00107.1~ Tic [Homo sapiens] >sp~AAD00107~AAD00107 Tic.
Length = 1056 Plus Strand HSPs:
Score = 710 (249.9 bits), Expect = 2.6e-102, Sum P(2) = 2.6e-102 Identities = 145/202 (71~), Positives = 147/202 (72~), Frame = +2 3O Q: 185 QEAHVFQLRTADWRLYLFXAXTAKEMSSWIARINLAAATHSAPPFPAAVGSQRRFVRPIL 364 ++ HVFQLRTADWRLYLF A TAKEMSSWIARINLAAATHSAPPFPAAVGSQRRFVRPIL
S: 855 KKPHVFQLRTADWRLYLFQAPTAKEMSSWIARINLAAATHSAPPFPAAVGSQRRFVRPIL 914 Q: 365 PVGPAQSSLEEQHRSHENCXXXXXXXXXXXQXXXXXXXXXXXXXXXXXXXXXXXXXXKTR 544 PVGPAQSSLEEQHRSHENC Q KTR
S: 915 PVGPAQSSLEEQHRSHENCLDAAADDLLDLQRNLPERRGRGRELEEHRLRKEYLEYEKTR 974 S Q: 545 YETYVQLLVARLHCPSDALDLWEEQLGREAGGTREXXXXXXXXXXXXXXXQDEAPTTAKV 724 YETYVQLLVARLHCPSDALDLWEEQLGREAGGTRE QDEAPTTAKV
S: 975 YETYVQLLVARLHCPSDALDLWEEQLGREAGGTREPKLSLKKSHSSPSLHQDEAPTTAKV 1034 Q: 725 KRNISERRTYRKIIPKRNRNQL 790 S: 1035 KRNISERRTYRKIIPKRNRNQL 1056 The segment of gbIAAD00107.11 that is shown as "S" above is set out in the sequence listing as SEQ ID NO. 143 . Based on the structural similarity, these 1S 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 20 amino acid sequence set out in the sequence listing as SEQ ID NO. 144 which corresponds to the "Q" sequence in the alignment shown above (gaps introduced in a sequence by the computer are, of course, removed).
It has been discovered that this gene is expressed primarily in the following tissues/cDNA libraries: Soares fetal heart NbHHI9W and to a lesser extent in 2S Activated T-Cells, 12 hrs, subtracted; Human Primary Breast Cancer Reexcision;
Soares fetal liver spleen 1NFLS; Soares fetal liver spleen_1NFLS S1; Colon Carcinoma; human tonsils; Human Colon, differential expression;
Soares_parathyroid tumor NbHPA; Human Fetal Dura Mater; Soares breast 2NbHBst; Human Eosinophils; breast lymph node CDNA library; Colon Normal II;
30 Anergic T-cell; Activated T-cell(12h)/Thiouridine-re-excision; Primary Dendritic Cells, lib 1; Human Colon, subtraction; NCI CGAP_CoB; NCI CGAP_LuS;

NCI CGAP_Col6; NCI CGAP_Kids; NCI CGAP Lyml2; Fetal Heart, re-excision;
Dendritic Cells From CD34 Cells; Human Osteosarcoma; Jurkat T-cell G1 phase;
Soares NFL T GBC_S1; Soares NSF F8 9W OT PA P S1;
Soares_pregnant uterus NbHPU; Monocyte activated, re-excision; Ulcerative Colitis; Human Thymus; Human Thymus Stromal Cells; Stratagene liver (#937224);
Soares breast 3NbHBst; Dendritic cells, pooled; Human Testes, Reexcision;
Human Fetal Heart; NCI CGAP_Prl2; Human Bone Marrow, treated; Neutrophils IL-1 and LPS induced; Bone Marrow Cell Line (RS4,11); NCI CGAP_Panl; H. Frontal cortex,epileptic,re-excision; Human Endometrial Tumor; Soares NhHMPu_S1;
Hodgkin's Lymphoma II; Keratinocyte; T cell helper II; NCI CGAP GCB 1 and NCI CGAP Kid3.
Preferred polypeptides of the present invention comprise immunogenic epitopes shown in SEQ ID NO: 87 as residues: Gln-21 to Trp-30. Polynucleotides encoding said polypeptides are also encompassed by the invention. Antibodies that bind polypeptides of the invention are encompassed by the invention, particularly antibodies that inhibit activity.
Many polynucleotide sequences, such as EST sequences, are publicly available and accessible through sequence databases. Some of these sequences are related to SEQ ID N0:38 and may have been publicly available prior to conception of the present invention. Preferably, such related polynucleotides are specifically excluded from the scope of the present invention. To list every related sequence would be cumbersome. Accordingly, preferably excluded from the present invention are one or more polynucleotides comprising a nucleotide sequence described by the general formula of a-b, where a is any integer between 1 to 2317 of SEQ ID
N0:38, b is an integer of 15 to 2331, where both a and b correspond to the positions of nucleotide residues shown in SEQ ID N0:38, and where b is greater than or equal to a + 14.
FEATURES OF PROTEIN ENCODED BY GENE NO: 29 It has been discovered that this gene is expressed primarily in H. Ovarian Tumor, II, OV5232.
Preferred polypeptides of the present invention comprise immunogenic epitopes shown in SEQ ID NO: 88 as residues: Gln-26 to Thr-35. Polynucleotides encoding said polypeptides are also encompassed by the invention. Antibodies that bind polypeptides of the invention are encompassed by the invention, particularly antibodies that inhibit activity.
Many polynucleotide sequences, such as EST sequences, are publicly available and accessible through sequence databases. Some of these sequences are related to SEQ ID N0:39 and may have been publicly available prior to conception of the present invention. Preferably, such related polynucleotides are specifically excluded from the scope of the present invention. To list every related sequence would be cumbersome. Accordingly, preferably excluded from the present invention are one or more polynucleotides comprising a nucleotide sequence described by the general formula of a-b, where a is any integer between 1 to 886 of SEQ ID
N0:39, b is an integer of 15 to 900, where both a and b correspond to the positions of nucleotide residues shown in SEQ ID N0:39, and where b is greater than or equal to a + 14.
FEATURES OF PROTEIN ENCODED BY GENE NO: 30 It has been discovered that this gene is expressed primarily in the following tissues/cDNA libraries: PERM TF274.

Preferred polypeptides of the present invention comprise immunogenic epitopes shown in SEQ ID NO: 89 as residues: Gln-20 to Leu-25. Polynucleotides encoding said polypeptides are also encompassed by the invention. Antibodies that bind polypeptides of the invention are encompassed by the invention, particularly antibodies that inhibit activity.
When tested against K562 leukemia cell lines, supernatants removed from cells containing this gene activated the ISRE assay. Thus, it is likely that this gene activates leukemia cells through the Jak-STAT signal transduction pathway. The interferon-sensitive response element is apromoter element found upstream of many genes which are involved in the Jak-STAT pathway. The Jak-STAT pathway is a large, signal transduction pathway involved in the differentiation and proliferation of cells. Therefore, activation of the Jak-STAT pathway, reflected by the binding of the ISRE element, can be used to indicate proteins involved in the proliferation and differentiation of cells.
Many polynucleotide sequences, such as EST sequences, are publicly available and accessible through sequence databases. Some of these sequences are related to SEQ ID N0:40 and may have been publicly available prior to conception of the present invention. Preferably, such related polynucleotides are specifically excluded from the scope of the present invention. To list every related sequence would be cumbersome. Accordingly, preferably excluded from the present invention are one or more polynucleotides comprising a nucleotide sequence described by the general formula of a-b, where a is any integer between 1 to 808 of SEQ ID
N0:40, b is an integer of 15 to 822, where both a and b correspond to the positions of nucleotide residues shown in SEQ ID N0:40, and where b is greater than or equal to a + 14.

FEATURES OF PROTEIN ENCODED BY GENE NO: 31 It has been discovered that this gene is expressed primarily in .
Many polynucleotide sequences, such as EST sequences, are publicly available and accessible through sequence databases. Some of these sequences are related to SEQ ID N0:41 and may have been publicly available prior to conception of the present invention. Preferably, such related polynucleotides are specifically excluded from the scope of the present invention. To list every related sequence would be cumbersome. Accordingly, preferably excluded from the present invention are one or more polynucleotides comprising a nucleotide sequence described by the general formula of a-b, where a is any integer between 1 to 2057 of SEQ ID
N0:41, b is an integer of 15 to 2071, where both a and b correspond to the positions of nucleotide residues shown in SEQ ID N0:41, and where b is greater than or equal to a + 14.
FEATURES OF PROTEIN ENCODED BY GENE NO: 32 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.
gbIAAC46766.11 (all information available through the recited accession number is incorporated herein by reference) which is described therein as "No definition line found [Caenorhabditis elegans]". A partial alignment demonstrating the observed homology is shown immediately below.
>gb~AAC46766.1~ No definition line found [Caenorhabditis elegans]
Length = 477 Plus Strand HSPS:
Score = 184 (64.8 bits), Expect = 4.2e-15, Sum P(2) = 4.2e-15 Identities = 36/76 (47$), Positives = 51/76 (67$), Frame = +2 Q: 23 ASFVFNGFLDFILRPDDPRAQTLRRLFVFKLIPMLNPDGWRGHYRTDSRGVNLNRQYLK 202 +S V +G ++F++ DD RAQ LR+++ FK+IPMLNPDGV G+YR G +LNR +
S: 210 SSHVMHGIIEFLVSKDD-RAQKLRKVYCFKIIPMLNPDGVFLGNYRCSLMGHDLNRMWRT 268 Q: 203 PDAVLHPAIYGAKAVL 250 P HP+IY K +L
S: 269 PSDWAHPSIYAVKNLL 284 10 The segment of gbIAAC46766.11 that is shown as "S" above is set out in the sequence listing as SEQ ID NO. 145. 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 15 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. 146 which corresponds to the "Q" sequence in the alignment shown above (gaps introduced in a sequence by the computer are, of course, removed).
20 It has been discovered that this gene is expressed primarily in the following tissues/cDNA libraries: Soares_pregnant uterus NbHPU; Soares fetal liver spleen 1NFLS and to a lesser extent in NTERA2 teratocarcinoma cell line+retinoic acid (14 days); Soares_pregnant uterus NbHPU; Human Primary Breast Cancer Reexcision;
Human 8 Week Whole Embryo; Soares infant brain 1NIB; Soares ovary tumor 25 NbHOT; Testis, normal; Human OB MG63 treated (10 nM E2) fraction I; Human Thyroid; Soares NhHMPu_S1; Stratagene muscle 937209; Human endometrial stromal cells-treated with estradiol; Human Hippocampus; Hemangiopericytoma;
Rejected Kidney, lib 4; Human adult testis, large inserts; Smooth muscle, serum treated; Adipocytes; Soares melanocyte 2NbHM; Soares fetal heart NbHHI9W;

Soares senescent fibroblasts_NbHSF; Human Fetal Lung III; T Cell helper I;
Osteoblasts; Nine Week Old Early Stage Human and T cell helper II.
Many polynucleotide sequences, such as EST sequences, are publicly available and accessible through sequence databases. Some of these sequences are related to SEQ ID N0:42 and may have been publicly available prior to conception of the present invention. Preferably, such related polynucleotides are specifically excluded from the scope of the present invention. To list every related sequence would be cumbersome. Accordingly, preferably excluded from the present invention are one or more polynucleotides comprising a nucleotide sequence described by the general formula of a-b, where a is any integer between 1 to 1482 of SEQ ID
N0:42, b is an integer of 15 to 1496, where both a and b correspond to the positions of nucleotide residues shown in SEQ ID N0:42, and where b is greater than or equal to a + 14.
FEATURES OF PROTEIN ENCODED BY GENE NO: 33 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.
gbIAAC69227.11 (all information available through the recited accession number is incorporated herein by reference) which is described therein as "No definition line found [Caenorhabditis elegans]". A partial alignment demonstrating the observed homology is shown immediately below.
>gb~AAC69227.1~ (AF101315) No definition line found [Caenorhabditis elegans) >sp~AAC69227~AAC69227 T22H9.2 protein.
Length = 880 Plus Strand HSPs:

Score = 398 (140.1 bits), Expect = 1.7e-32, Sum P(2) = 1.7e-32 Identities = 78/186 (418), Positives = 112/186 (60$), Frame = +1 Q: 70 WSLYGRCYLRHFNELEHELQSRLNRGYKPASKYMNCFLSPLLTLLAKNGAFFAGSILAVL 249 S W+ + LRHFNEL+HEL +RLNR + A+ YM+ F SP+L + AKN F A ++ VL
S: 421 WNEKIQYLLRHFNELDHELSARLNRSHIYAAAYMDQFFSPVLEIAAKNITFIAAAVFGVL 480 Q: 250 IALTIYDEDXXXXXXXXXXXXXXXXXXXXCRSFIPDQHMVFCPEQLLRVILAHIHYMPDH 429 L+ +DED CR IPD+++V+ PE L+ + + +HY+P
1O S: 481 TILSAWDEDVLQVEHVITVLTICGIVVLVCRGMIPDENLVWQPEILMTHVTSELHYLPST 540 Q: 430 WQGNAHRSQTRDEFAQLFQYKAVFILEELLSPIVTPLILIFCLRPRALEIIDFFRNFTVE 609 W+G AH + R EF QLFQ K +F + EL SPI TP +L+F LRPR ++ +FF ++T
S: 541 WKGKAHTTGVRHEFDQLFQMKWMFFVLELTSPIFTPFVLLFWLRPRCSQLANFFHDYTER 600 Q: 610 WGVGD 627 V G+GD
S: 601 VDGLGD 606 20 Score = 42 (14.8 bits), Expect = 1.7e-32, Sum P(2) = 1.7e-32 Identities = 20/65 (30$), Positives = 30/65 (46~), Frame = +2 Q: 1628 MEATAALPSRLPAMGRLL*VFPGPTCVVFVCLCLAKGGANTGLATAPGEEFGA*EPRAHG 1807 + A+ P R P +G L GP F + AK G+ LA+ E+ PRA
2S S: 669 ISASQVAPGRHPLIGDGLHRIDGPVGNAFQGIQGAKLGSGGVLASLYQEQ-----PRAAE 723 Q: 1808 TLASS 1822 +L++S
S: 724 SLSNS 728 The segments of gbIAAC69227.11 that are shown as "S" above are set out in the sequence listing as SEQ ID NO. 147 and SEQ ID NO. 149. 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 3S 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. 148 and/or SEQ
ID NO. 1S0 and/or SEQ ID NO. 16S and/or SEQ ID N0.166which correspond to the "Q" sequences in the alignment shown above (gaps introduced in a sequence by the computer are, of course, removed).

It has been discovered that this gene is expressed primarily in the following tissues/cDNA libraries: Soares ovary tumor NbHOT; Human Adult Testes, Large Inserts, Reexcision and to a lesser extent in Soares placenta Nb2HP; Human adult testis, large inserts; Soares fetal liver spleen 1NFLS; Soares infant brain 1NIB;
Soares testis NHT; Human Cerebellum; Human Fetal Brain, normalized A5002F;
Soares testis NHT; Glioblastoma; NCI CGAP_Pr9; T-Cell PHA 24 hrs;
Macrophage-oxLDL; Soares fetal liver spleen_1NFLS S1; Human Adult Skeletal Muscle; Bone Cancer; Human 7 Weeks Old Embryo, subtracted; H. Adipose Tissue;
Saos2 Cells, Untreated; Human Cerebellum, subtracted; Human Placenta;
Stratagene neuroepithelium (#937231); Smooth Muscle Serum Treated, Norm; CD40 activated monocyte dendridic cells; H. Epididiymus, caput & corpus; Human Liver; Breast Lymph node cDNA library; H. Epididiymus, cauda; Human Quadriceps; Human Soleus; Human Colon Cancer,re-excision; NTERA2 + retinoic acid, 14 days; Human Fetal Epithelium (Skin); Human Adipose Tissue, re-excision; Human Pituitary, subt IX; Prostate BPH; H. Lymph node breast Cancer; HUMAN JURKAT MEMBRANE
BOUND POLYSOMES; NCI CGAP_Brn23; Olfactory epithelium,nasalcavity;
Human Adipose; Bone Marrow Stromal Cell, untreated; Human Thymus Stromal Cells; Macrophage (GM-CSF treated); Human Whole Six Week Old Embryo;
Hepatocellular Tumor, re-excision; Human Liver, normal; Smooth muscle, serum induced,re-exc; Human Gall Bladder; Smooth muscle, serum treated; Human Placenta; Dendritic cells, pooled; HM3; Infant brain, Bento Soares;
Soares NFL T_GBC S1; Stratagene lung carcinoma 937218; Stratagene cDNA
library Human heart, cat#936208; Human Fetal Kidney, Reexcision; Endothelial-induced; Endothelial cells-control; Smooth muscle,control; T Cell helper I;
Human Testes; NCI CGAP Ut2; H. Frontal cortex,epileptic,re-excision; Activated T-cell(12h)/Thiouridine-re-excision; Keratinocyte; Nine Week Old Early Stage Human;
HM1; NCI CGAP_Br3; NCI CGAP_ColO and NCI CGAP_Kid6.
Preferred polypeptides of the present invention comprise immunogenic epitopes shown in SEQ ID NO: 92 as residues: Pro-85 to Asp-99, Arg-163 to Arg-170, Gln-183 to Thr-189, Pro-201 to Ser-209, Ser-216 to Gly-222.
Polynucleotides encoding said polypeptides are also encompassed by the invention. Antibodies that bind polypeptides of the invention are encompassed by the invention, particularly antibodies that inhibit activity.
Many polynucleotide sequences, such as EST sequences, are publicly available and accessible through sequence databases. Some of these sequences are related to SEQ ID N0:43 and may have been publicly available prior to conception of the present invention. Preferably, such related polynucleotides are specifically excluded from the scope of the present invention. To list every related sequence would be cumbersome. Accordingly, preferably excluded from the present invention are one or more polynucleotides comprising a nucleotide sequence described by the general formula of a-b, where a is any integer between 1 to 2690 of SEQ ID
N0:43, b is an integer of 15 to 2704, where both a and b correspond to the positions of nucleotide residues shown in SEQ ID N0:43, and where b is greater than or equal to a + 14.
FEATURES OF PROTEIN ENCODED BY GENE NO: 34 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.
gbIAAA81099.11 (all information available through the recited accession number is incorporated herein by reference) which is described therein as "weak similarity to E.

coli 30S ribosomal protein S16 (SP:P02372) [Caenorhabditis elegans]". A
partial alignment demonstrating the observed homology is shown immediately below.
>gb~AAA81099.1~ weak similarity to E. coli 30S ribosomal protein S16 (SP:P02372) [Caenorhabditis elegans]
Length = 152 Plus Strand HSPs:
10 Score = 222 (78.1 bits), Expect = 1.2e-17, P = 1.2e-17 Identities = 48/103 (468), Positives = 66/103 (64$), Frame = +2 Q: 116 GHLTIRLALGGCTNRPFYRIVAAHNKC-PR--DGRFVEQLGSYDPLPNSHGEKLVALNLD 286 G +I LAL GCTNRPFY + ++ R +G +EQ+G++DPLPN EKLVALN
IS S: 11 GRPSIGLALFGCTNRPFYHVCVFPDRALGRRYEGNILEQVGTFDPLPNQKNEKLVALNFG 70 Q: 287 RIRHWIG-CGAHLSKPMEKLLG-----LAGFFPLHPMMITNAE 397 R+++WIG AH+S P+ +LLG L+G FP+HP A+
S: 71 RLKYWIGERNAHISVPVLELLGERRNCLSGLFPIHPKSFIRAK 113 The segment of gbIAAA81099.11 that is shown as "S" above is set out in the 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 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).
It has been discovered that this gene is expressed primarily in the following tissues/cDNA libraries: NCI CGAP_GCB1 and to a lesser extent in Stratagene fetal spleen (#937205); Stratagene endothelial cell 937223; Endothelial-induced;
Osteoblasts; Keratinocyte; Human OB MG63 treated (10 nM E2) fraction I; Human Colon Carcinoma (HCC) cell line; Smooth muscle-ILb induced; Aorta endothelial cells + TNF-a; Human Primary Breast Cancer; Soares total fetus Nb2HF8_9w;
Human Lung; Raji Cells, cyclohexamide treated; Myoloid Progenitor Cell Line;
Jurkat T-Cell, S phase; Human Adult Small Intestine; Human Brain, Striatum;
HUMAN JURKAT MEMBRANE BOUND POLYSOMES; Human Adult Testes, Large Inserts, Reexcision; Stromal cell TF274; NCI CGAP_Br2; NCI CGAP_Col;
NCI CGAP_Co9; NCI CGAP_GC4; NCI CGAP_LuS; NCI CGAP_Pr3;
NCI CGAP_GCBO; Human umbilical vein endothelial cells, IL-4 induced; Rejected Kidney, lib 4; Macrophage (GM-CSF treated); NTERA2, control;
NCI CGAP_GCB 1; Soares NhHMPu_S 1; Pancreas Islet Cell Tumor;
NCI CGAP_Brn23; Stratagene fetal retina 937202; PC3 Prostate cell line; Colon Tumor; Colon Normal II; Human Synovial Sarcoma; Activated T-Cell (l2hs)/Thiouridine labelledEco; Endothelial cells-control; Monocyte activated and Primary Dendritic Cells, lib 1.
Preferred polypeptides of the present invention comprise immunogenic epitopes shown in SEQ ID NO: 93 as residues: Cys-26 to Phe-31, Asn-39 to Gly-4.5, Asp-55 to Gly-62, Glu-108 to Ala-115, Gln-123 to Thr-137. Polynucleotides encoding said polypeptides are also encompassed by the invention. Antibodies that bind polypeptides of the invention are encompassed by the invention, particularly antibodies that inhibit activity.
Many polynucleotide sequences, such as EST sequences, are publicly available and accessible through sequence databases. Some of these sequences are related to SEQ ID N0:44 and may have been publicly available prior to conception of the present invention. Preferably, such related polynucleotides are specifically excluded from the scope of the present invention. To list every related sequence would be cumbersome. Accordingly, preferably excluded from the present invention are one or more polynucleotides comprising a nucleotide sequence described by the general formula of a-b, where a is any integer between 1 to 603 of SEQ ID
N0:44, b is an integer of 15 to 617, where both a and b correspond to the positions of nucleotide residues shown in SEQ ID N0:44, and where b is greater than or equal to a + 14.
FEATURES OF PROTEIN ENCODED BY GENE NO: 35 It has been discovered that this gene is expressed primarily in the following tissues/cDNA libraries: Primary Dendritic Cells, lib 1 and to a lesser extent in NCI CGAP_GCB1; Human Activated T-Cells; Human T-cell lymphoma,re-excision;
Human T-Cell Lymphoma; Hodgkin's Lymphoma II; Human Umbilical Vein Endothelial Cells, fract. A; pBMC stimulated w/ poly I/C; Human Osteoclastoma;
Activated T-cell(12h)/Thiouridine-re-excision; Morton Fetal Cochlea; Mo7e Cell Line GM-CSF treated (lng/ml); Human Pancreas Tumor; Human Neutrophil, Activated; NCI CGAP_Br2; Anergic T-cell; Soares placenta Nb2HP; Human Stomach,re-excision; Myoloid Progenitor Cell Line; T-Cell PHA 16 hrs;
Soares_NhHMPu_S1; NCI CGAP_Brn23; Apoptotic T-cell; T-Cell PHA 24 hrs;
Human Pancreas Tumor, Reexcision; Soares_pregnant uterus NbHPU; Ulcerative Colitis; Macrophage (GM-CSF treated); Human Whole Six Week Old Embryo;
Soares multiple sclerosis 2NbHMSP; Resting T-Cell Library,II; Colon Normal II;
Activated T-Cell (l2hs)/Thiouridine labelledEco; Human aorta polyA+
(TFujiwara);
Human Bone Marrow, treated; Spleen, Chronic lymphocytic leukemia; T Cell helper I; Human 8 Week Whole Embryo; Human Kidney Medulla - unamplified; Human Activated T-Cells (II); Human Adult Heart; Ku 812F Basophils Line; Human Uterus, normal; Human Fetal Lung; Human Adult Spleen; Human Fetal Brain; Human White Adipose; H. Epididiymus, caput & corpus; Soares NhHMPu_S1; Cem cells cyclohexamide treated; Human Hypothalamus,schizophrenia, re-excision; Synovial Fibroblasts (Ill/TNF), subt; Spinal Cord, re-excision; Soares NFL T GBC_S1;
Soares_pregnant uterus NbHPU; Soares_parathyroid tumor NbHPA;
Soares senescent fibroblasts NbHSF; NCI CGAP LuS; NCI CGAP_Gas4;
NCI CGAP_Panl; Temporal cortex-Alzheizmer, subtracted; Human Bone Marrow, re-excision; Monocyte activated, re-excision; Soares multiple sclerosis 2NbHMSP;
Human Hypothalmus,Schizophrenia; Soares_pineal_gland N3HPG; Human Placenta (re-excision); Human Chondrosarcoma; Rejected Kidney, lib 4;
Soares fetal heart NbHHI9W; Smooth muscle, serum induced,re-exc; Macrophage-oxLDL, re-excision; Human Eosinophils; Human Testes Tumor; Normal colon;
Primary Dendritic cells,frac 2; Human Fetal Lung III; 12 Week Early Stage Human II, Reexcision; human tonsils; NCI CGAP_LuS; NCI CGAP_Kid3;
NCI CGAP_Kid6; NCI CGAP_Lei2; NCI CGAP_Brn23; Human Microvascular Endothelial Cells, fract. A; Monocyte activated; CD34 positive cells (Cord Blood);
HUMAN B CELL LYMPHOMA; Human fetal heart, Lambda ZAP Express;
Osteoblasts; Nine Week Old Early Stage Human and T cell helper II.
Many polynucleotide sequences, such as EST sequences, are publicly available and accessible through sequence databases. Some of these sequences are related to SEQ ID N0:45 and may have been publicly available prior to conception of the present invention. Preferably, such related polynucleotides are specifically excluded from the scope of the present invention. To list every related sequence would be cumbersome. Accordingly, preferably excluded from the present invention are one or more polynucleotides comprising a nucleotide sequence described by the general formula of a-b, where a is any integer between 1 to 267 of SEQ ID
N0:45, b is an integer of 15 to 281, where both a and b correspond to the positions of nucleotide residues shown in SEQ ID N0:45, and where b is greater than or equal to a + 14.

FEATURES OF PROTEIN ENCODED BY GENE NO: 36 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.
pirIB253131GNLRL1 (all information available through the recited accession number is incorporated herein by reference) which is described therein as "retrovirus-related reverse transcriptase pseudogene - slow loris". A partial alignment demonstrating the observed homology is shown immediately below.
>pirIB253131GNLRL1 retrovirus-related reverse transcriptase pseudogene -slow loris >spIP085481LIN1 NYCCO LINE-1 REVERSE TRANSCRIPTASE
HOMOLOG.
Length = 1260 Minus Strand HSPs:
Score = 195 (68.6 bits), Expect = 3.2e-23, Sum P(5) = 3.2e-23 Identities = 54/123 (43$), Positives = 73/123 (59~), Frame = -3 Q: 362 NFPWVNHGEIKTLNTLIITKGIKAVIKNLSAKKNPGPENFTGKFC*TLKE-LTSIFLKQL 186 + P ++ E++ LN I + I + I+NL KK+PGP+ FT +F T KE L I L
S: 436 HLPRLSQKEVEMLNRPISSSEIASTIQNLPKKKSPGPDGFTSEFYQTFKEELVPILLNLF 495 Q: 185 SSKELKQREHFSNTF*EACISLTPKPDKD-TRKLQ------TNIPDAKILNRTLANGIKM 27 + +++ NTF EA I+L PKP KD TRK NI DAKILN+ L N I+
S: 496 QN--IEKEGILPNTFYEANITLIPKPGKDPTRKENYRPISLMNI-DAKILNKILTNRIQQ 552 3~ Q: 26 HIKRII 9 HIK+II
S: 553 HIKKII 558 The segment of pirIB253131GNLRL1 that is shown as "S" above is set out in the sequence listing as SEQ ID NO. 153 . 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. 154, SEQ ID
5 N0.167, and/or SEQ ID N0:168 which corresponds to the "Q" sequence in the alignment shown above (gaps introduced in a sequence by the computer are, of course, removed).
It has been discovered that this gene is expressed primarily in Anergic T-cell.
Many polynucleotide sequences, such as EST sequences, are publicly 10 available and accessible through sequence databases. Some of these sequences are related to SEQ ID N0:46 and may have been publicly available prior to conception of the present invention. Preferably, such related polynucleotides are specifically excluded from the scope of the present invention. To list every related sequence would be cumbersome. Accordingly, preferably excluded from the present invention 15 are one or more polynucleotides comprising a nucleotide sequence described by the general formula of a-b, where a is any integer between 1 to 1478 of SEQ ID
N0:46, b is an integer of 15 to 1492, where both a and b correspond to the positions of nucleotide residues shown in SEQ ID N0:46, and where b is greater than or equal to a + 14.
20 FEATURES OF PROTEIN ENCODED BY GENE NO: 37 It has been discovered that this gene is expressed primarily in the following tissues/cDNA libraries: Activated T-cells, 24 hrs,re-excision and to a lesser extent in Soares fetal liver spleen 1NFLS; Soares breast 2NbHBst; Soares breast 3NbHBst;
Human Epididymus; Early Stage Human Brain; Human Cerebellum; T Cell helper I;
25 Soares testis NHT; Activated T-Cells, 24 hrs.; Human Fetal Brain; Human Lung Cancer,re-excision; LNCAP prostate cell line; Jurkat T-cell G1 phase; Human Fetal Dura Mater; Colon Carcinoma; Human Primary Breast Cancer Reexcision;
Soares fetal heart NbHHI9W; Soares NFL_T GBC_S1; Keratinocyte; Soares infant brain 1NIB; Human Esophagus, Cancer; Human Pituitary, subtracted VIII;
Colon, normal; Prostate; Human Whole 6 Week Old Embryo (II), subt; Normal Ovary, Premenopausal; Bone Cancer; Hypothalamus; Human Prostate Cancer, Stage B2 fraction; Whole 6 Week Old Embryo; Human OB HOS treated ( 10 nM E2) fraction I; Human Aortic Endothelium; HUMAN STOMACH; Supt Cells, cyclohexamide treated; Amniotic Cells - TNF induced; Human Pineal Gland;
STROMAL -OSTEOCLASTOMA; Human endometrial stromal cells-treated with estradiol; H Female Bladder, Adult; Human Frontal Cortex, Schizophrenia; Human Osteoclastoma, re-excision; Myoloid Progenitor Cell Line; wilm's tumor;
Prostate BPH; CD34 depleted Buffy Coat (Cord Blood); Human Thymus; Human Fetal Kidney; 12 Week Old Early Stage Human, II; Human Uterine Cancer;
Soares total fetus Nb2HF8 9w; Human Heart; Human Hippocampus; Soares adult brain N2b5HB55Y; Hemangiopericytoma; Rejected Kidney, lib 4; Human Whole Six Week Old Embryo; Human adult testis, large inserts; Human Liver, normal;
Pancreas Islet Cell Tumor; Adipocytes; H Macrophage (GM-CSF treated), re-excision;
Human Testes Tumor; Primary Dendritic cells,frac 2;
Soares_placenta 8to9weeks 2NbHP8to9W; Human Fetal Lung III; Bone marrow;
Endothelial-induced; Human Adult Pulmonary,re-excision; Endothelial cells-control;
CD34 depleted Buffy Coat (Cord Blood), re-excision; Anergic T-cell; Monocyte activated; H. Frontal cortex,epileptic,re-excision; Hodgkin's Lymphoma II;
Human heart cDNA (YNakamura); NCI CGAP_ColO; NCI CGAP_Kid3;
NCI CGAP_Kids; Soares NhHMPu_S1 and Nine Week Old Early Stage Human.
Many polynucleotide sequences, such as EST sequences, are publicly available and accessible through sequence databases. Some of these sequences are related to SEQ ID N0:47 and may have been publicly available prior to conception of the present invention. Preferably, such related polynucleotides are specifically excluded from the scope of the present invention. To list every related sequence would be cumbersome. Accordingly, preferably excluded from the present invention are one or more polynucleotides comprising a nucleotide sequence described by the general formula of a-b, where a is any integer between 1 to 885 of SEQ ID
N0:47, b is an integer of 15 to 899, where both a and b correspond to the positions of nucleotide residues shown in SEQ ID N0:47, and where b is greater than or equal to a + 14.
FEATURES OF PROTEIN ENCODED BY GENE NO: 38 It has been discovered that this gene is expressed primarily in the following tissues/cDNA libraries: 12 Week Old Early Stage Human, II; Spleen/normal;
Anergic T-cell.
Many polynucleotide sequences, such as EST sequences, are publicly available and accessible through sequence databases. Some of these sequences are related to SEQ ID N0:48 and may have been publicly available prior to conception of the present invention. Preferably, such related polynucleotides are specifically excluded from the scope of the present invention. To list every related sequence would be cumbersome. Accordingly, preferably excluded from the present invention are one or more polynucleotides comprising a nucleotide sequence described by the general formula of a-b, where a is any integer between 1 to 1228 of SEQ ID
N0:48, b is an integer of 15 to 1242, where both a and b correspond to the positions of nucleotide residues shown in SEQ ID N0:48, and where b is greater than or equal to a + 14.

FEATURES OF PROTEIN ENCODED BY GENE NO: 39 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.
gbIAAD02337.11 (all information available through the recited accession number is incorporated herein by reference) which is described therein as "MM4b [Homo sapiens]". A partial alignment demonstrating the observed homology is shown immediately below.
>gb~AAD02337.1~ (AF044671) MM46 [Homo sapiens] >sp~AAD02337~AAD02337 MM46.
Length = 117 Plus Strand HSPS:
Score = 142 (50.0 bits), Expect = 3.5e-09, P = 3.5e-09 Identities = 26/26 (100$), Positives = 26/26 (100$), Frame = +2 Q: 8 GQLYQEHHEEDFFLYIAYSDESVYGL 85 GQLYQEHHEEDFFLYIAYSDESVYGL
2O S: 92 GQLYQEHHEEDFFLYIAYSDESVYGL 117 The segment of gbIAAD02337.11 that is shown as "S" above is set out in the sequence listing as SEQ ID NO. 155 . 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. 156 which corresponds to the "Q" sequence in the alignment shown above (gaps introduced in a sequence by the computer are, of course, removed).

It has been discovered that this gene is expressed primarily in the following tissues/cDNA libraries: Soares breast 3NbHBst and to a lesser extent in Early Stage Human Brain; Soares fetal liver spleen 1NFLS; Soares placenta Nb2HP; Human Normal Breast; Human Epididymus; CD34 depleted Buffy Coat (Cord Blood);
Soares fetal heart NbHHI9W; Soares melanocyte 2NbHM;
Soares fetal heart_NbHHI9W; Soares retina N2b4HR;
Soares_pregnant uterus NbHPU; Human Pineal Gland; wilm's tumor; 12 Week Old Early Stage Human; breast lymph node CDNA library; CD34 depleted Buffy Coat (Cord Blood), re-excision; Stratagene endothelial cell 937223; Human Prostate Cancer, Stage C fraction; Soares_pregnant uterus NbHPU; Human Chronic Synovitis; Human Prostate; Human Fetal Dura Mater; Macrophage-oxLDL; Human Thymus; Hemangiopericytoma; Human Fetal Brain; Human Fetal Lung III; Human Brain; Human Pituitary, subtracted VIII; Messangial cell, frac 1; Human Placenta, subtracted; Human Infant Adrenal Gland; Duodenum; WI 38 cells; Human White Fat;
H. Adipose Tissue; Human Uterus, normal; Human epithelioid sarcoma; Whole 6 Week Old Embryo; Morton Fetal Cochlea; Soares fetal liver spleen_1NFLS S1;
Frontal Lobe, Dementia; H. Striatum Depression, subt; Human White Adipose;
Human Adult Pulmonary; eosinophil-ILS induced; Human Fetal Spleen; Human Placenta; Aorta endothelial cells + TNF-a; Human Primary Breast Cancer; Breast Lymph node cDNA library; Soares_pineal_gland N3HPG;
Soares fetal heart NbHHI9W; Stratagene lung carcinoma 937218; Smooth Muscle-HASTE normalized; Human endometrial stromal cells-treated with estradiol;
human corpus colosum; Human Fetal Epithelium (Skin); H. Kidney Cortex, subtracted;
Human Osteoclastoma, re-excision; T-Cell PHA 16 hrs; Human Thymus; Human Brain, Striatum; Apoptotic T-cell; Human Uterine Cancer;
Soares fetal lung NbHLI9W; Soares_pregnant uterus NbHPU; Human Hypothalmus,Schizophrenia; Human umbilical vein endothelial cells, IL-4 induced;
Soares adult brain N2b5HB55Y; Soares breast 2NbHBst; Human Adrenal Gland Tumor; Macrophage (GM-CSF treated); Human adult testis, large inserts; Smooth muscle, serum induced,re-exc; Human Placenta;
5 Soares_placenta 8to9weeks_2NbHP8to9W; Human Placenta; Bone marrow; Human Fetal Heart; Endothelial-induced; Anergic T-cell; Neutrophils IL-1 and LPS
induced;
Human Testes and Osteoblasts.
Many polynucleotide sequences, such as EST sequences, are publicly available and accessible through sequence databases. Some of these sequences are 10 related to SEQ ID N0:49 and may have been publicly available prior to conception of the present invention. Preferably, such related polynucleotides are specifically excluded from the scope of the present invention. To list every related sequence would be cumbersome. Accordingly, preferably excluded from the present invention are one or more polynucleotides comprising a nucleotide sequence described by the 15 general formula of a-b, where a is any integer between 1 to 519 of SEQ ID
N0:49, b is an integer of 15 to 533, where both a and b correspond to the positions of nucleotide residues shown in SEQ ID N0:49, and where b is greater than or equal to a + 14.
20 FEATURES OF PROTEIN ENCODED BY GENE NO: 40 It has been discovered that this gene is expressed primarily in the following tissues/cDNA libraries: Human Primary Breast Cancer Reexcision; Human Cerebellum and to a lesser extent in Soares ovary tumor NbHOT; Activated T-Cells, Ohrs, subtracted; Soares_pregnant uterus NbHPU; Nine Week Old Early Stage 25 Human; normalized infant brain cDNA; NCI CGAP_HSC2; Olfactory epithelium,nasalcavity; Rejected Kidney, lib 4; Human adult testis, large inserts;

NTERA2, control; breast lymph node CDNA library; Soares melanocyte 2NbHM;
NCI CGAP_GCB1; NCI CGAP_Pr22; Bone Marrow Cell Line (RS4,11);
Osteoblasts; Human 8 Week Whole Embryo; T cell helper II; Soares infant brain 1NIB; normalized infant brain cDNA; Infant brain, Bento Soares;
Soares testis NHT; Thyroid Tumour; Stomach Tumour; CD34+ cell, I, frac II;
Human OB HOS treated (1 nM E2) fraction I; Human Fetal Brain, random primed;
LNCAP untreated; Breast Lymph node cDNA library; Resting T-Cell, re-excision;
human corpus colosum; Human endometrial stromal cells-treated with progesterone;
Human endometrial stromal cells; Jurkat T-Cell, S phase; Healing groin wound, 6.5 hours post incision; L428; Human Osteoblasts II; Human Pancreas Tumor;
NCI_CGAP Lu26; Human Activated T-Cells, re-excision; NCI CGAP_Kid3;
NCI CGAP_Brn25; Human Testes Tumor, re-excision; CHME Cell Line,treated 5 hrs; Hepatocellular Tumor, re-excision; Pancreas Islet Cell Tumor; PC3 Prostate cell line; Colon Tumor; Smooth muscle, serum treated; Human Placenta;
NCI CGAP_Br2; NCI CGAP_GC4; NCI CGAP_Kid3; NCI CGAP_Kid6;
NCI CGAP_Prl2; Colon Tumor II; Human Synovial Sarcoma; Human fetal lung; 12 Week Early Stage Human II, Reexcision; NCI CGAP_Pr28; NCI CGAP_Brn25;
NCI CGAP_Kidll; Soares fetal heart_NbHHI9W;
Soares senescent fibroblasts NbHSF; NCI CGAP_Prl l; NCI CGAP_Pr23; Colon Normal III; Monocyte activated; Human Bone Marrow, treated; H. Frontal cortex,epileptic,re-excision and Keratinocyte.
Many polynucleotide sequences, such as EST sequences, are publicly available and accessible through sequence databases. Some of these sequences are related to SEQ ID NO:50 and may have been publicly available prior to conception of the present invention. Preferably, such related polynucleotides are specifically excluded from the scope of the present invention. To list every related sequence would be cumbersome. Accordingly, preferably excluded from the present invention are one or more polynucleotides comprising a nucleotide sequence described by the general formula of a-b, where a is any integer between 1 to 2252 of SEQ ID
NO:SO, b is an integer of 1S to 2266, where both a and b correspond to the positions of S nucleotide residues shown in SEQ ID NO:SO, and where b is greater than or equal to a + 14.
FEATURES OF PROTEIN ENCODED BY GENE NO: 41 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.
gbIAAA46S 11.11 (all information available through the recited accession number is incorporated herein by reference) which is described therein as "pol polyprotein [Murine leukemia virus]". A partial alignment demonstrating the observed homology 1S is shown immediately below.
>gb~AAA46511.1~ pol polyprotein [Murine leukemia virus] >pir~A26103~A26103 pol polyprotein - Cas-Br-E murine leukemia virus (fragment) 2O >sp~P08361~POL MLVCB POL POLYPROTEIN [CONTAINS: REVERSE
TRANSCRIPTASE (EC 2.7.7.49)] (FRAGMENT).
Length = 282 Plus Strand HSPS:

Score = 215 (75.7 bits), Expect = 1.6e-26, Sum P(2) = 1.6e-26 Identities = 50/105 (47~), Positives = 67/105 (63~), Frame = +3 Q: 630 LQACATARFEKAIEVSKLLLKEIIPRVGLPKSLQSDNGSPFTATVTRNTSSALGIQCRLD 809 3O ++A T + E A V+K LL+EI PR G+p+ L +DNG F + V++ + LGI +L
S: 27 IEAFPTKK-ETAKWTKKLLEEIFPRFGMPQVLGTDNGPAFVSKVSQTVADLLGIDWKLH 85 Q: 810 SARRPQSLGKVERANQTLKRTLAKLCQET-SETWRSLLPVALLRVR 944 A RPQS G+VER N+T+K TL KL T S W LLP+AL R R
3S S: 86 CAYRPQSSGQVERMNRTIKETLTKLTLATGSRDWVLLLPLALYRAR 131 The segment of gbIAAA46511.11 that is shown as "S" above is set out in the sequence listing as SEQ ID NO. 157 . 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. 158 which corresponds to the "Q" sequence in the alignment shown above (gaps introduced in a sequence by the computer are, of course, removed).
It has been discovered that this gene is expressed primarily in the following tissues/cDNA libraries: Supt Cells, cyclohexamide treated; HUMAN JURKAT
MEMBRANE BOUND POLYSOMES.
Preferred polypeptides of the present invention comprise immunogenic epitopes shown in SEQ ID NO: 100 as residues: Ala-17 to Cys-25, Arg-34 to Leu-40, Ser-57 to Gly-63. Polynucleotides encoding said polypeptides are also encompassed by the invention. Antibodies that bind polypeptides of the invention are encompassed by the invention, particularly antibodies that inhibit activity.
Many polynucleotide sequences, such as EST sequences, are publicly available and accessible through sequence databases. Some of these sequences are related to SEQ ID NO:51 and may have been publicly available prior to conception of the present invention. Preferably, such related polynucleotides are specifically excluded from the scope of the present invention. To list every related sequence would be cumbersome. Accordingly, preferably excluded from the present invention are one or more polynucleotides comprising a nucleotide sequence described by the general formula of a-b, where a is any integer between 1 to 1306 of SEQ ID
NO:51, b is an integer of 15 to 1320, where both a and b correspond to the positions of nucleotide residues shown in SEQ ID NO:51, and where b is greater than or equal to a + 14.
FEATURES OF PROTEIN ENCODED BY GENE NO: 42 It has been discovered that this gene is expressed primarily in the following tissues/cDNA libraries: 12 Week Old Early Stage Human; NCI CGAP_Lu5 and to a lesser extent in Soares NhHMI'u_S1; normalized infant brain cDNA; Human Whole Brain #2 - Oligo dT > l.SKb; Soares breast 3NbHBst; CD34 depleted Buffy Coat (Cord Blood), re-excision; Human Testes and Soares infant brain 1NIB.
Many polynucleotide sequences, such as EST sequences, are publicly available and accessible through sequence databases. Some of these sequences are related to SEQ ID N0:52 and may have been publicly available prior to conception of the present invention. Preferably, such related polynucleotides are specifically excluded from the scope of the present invention. To list every related sequence would be cumbersome. Accordingly, preferably excluded from the present invention are one or more polynucleotides comprising a nucleotide sequence described by the general formula of a-b, where a is any integer between 1 to 2186 of SEQ ID
N0:52, b is an integer of 15 to 2200, where both a and b correspond to the positions of nucleotide residues shown in SEQ ID N0:52, and where b is greater than or equal to a + 14.
FEATURES OF PROTEIN ENCODED BY GENE NO: 43 It has been discovered that this gene is expressed primarily in the following tissues/cDNA libraries: Signal trap,Femur Bone Marrow,pooled and to a lesser extent in normalized infant brain cDNA; Soares melanocyte 2NbHM; Soares ovary tumor NbHOT; Human Osteoclastoma; Soares fetal liver spleen 1NFLS; Soares infant brain 1NIB; Morton Fetal Cochlea; Human Pineal Gland; Spleen, Chronic lymphocytic leukemia; Weizmann Olfactory Epithelium; Human Osteoclastoma Stromal Cells -unamplified; Soares_pregnant uterus NbHPU; Human Fetal Kidney; Human Ovary;
Human Testes Tumor, re-excision; Human fetal heart, Lambda ZAP Express;
Hodgkin's Lymphoma II; Soares placenta Nb2HP;
Soares_parathyroid tumor NbHPA; Human Colon Cancer, differential;
Prostate,BPH, Lib 2; Bone marrow stroma,treated; NCI CGAP_Co9;
NCI CGAP_Prl ; NCI CGAP_Pr2; NCI CGAP Alv 1; NCI CGAP_GCB 1;
NCI CGAP_Kid6; Weizmann Olfactory; Hodgkin's Lymphoma I; Early Stage Human Lung, subtracted; H. Epididiymus, cauda; Messangial cell, frac 2; Human Prostate Cancer, Stage C fraction; Amniotic Cells - Primary Culture; Synovial hypoxia-RSF subtracted; Healing groin wound, 6.5 hours post incision; Synovial Fibroblasts (Ill/TNF), subt; T-Cell PHA 16 hrs; Soares fetal heart NbHHI9W;
Soares total fetus Nb2HF8 9w; Stratagene fetal retina 937202; Temporal cortex-Alzheizmer, subtracted; Human Thymus; Human Activated T-Cells; Stromal cell TF274; Human umbilical vein endothelial cells, IL-4 induced; NCI CGAP GCB1;
Soares NFL T_GBC S 1; Bone Marrow Stromal Cell, untreated; NTERA2, control;
Human Gall Bladder; Human T-Cell Lymphoma; Smooth muscle, serum treated;
Dendritic cells, pooled; Human Testes, Reexcision; Human Adult Pulmonary,re-excision; CD34 depleted Buffy Coat (Cord Blood), re-excision; Human placenta polyA+ (TFujiwara); NCI CGAP Kids; HUMAN B CELL LYMPHOMA; Human Bone Marrow, treated; Human Testes; NCI CGAP_GCB1 and Human 8 Week Whole Embryo.
Many polynucleotide sequences, such as EST sequences, are publicly available and accessible through sequence databases. Some of these sequences are related to SEQ ID NO:S3 and may have been publicly available prior to conception of the present invention. Preferably, such related polynucleotides are specifically excluded from the scope of the present invention. To list every related sequence would be cumbersome. Accordingly, preferably excluded from the present invention S are one or more polynucleotides comprising a nucleotide sequence described by the general formula of a-b, where a is any integer between 1 to 2011 of SEQ ID
NO:S3, b is an integer of 1S to 2025, where both a and b correspond to the positions of nucleotide residues shown in SEQ ID NO:S3, and where b is greater than or equal to a + 14.
FEATURES OF PROTEIN ENCODED BY GENE NO: 44 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.
1S dbjIBAA37146.11 (all information available through the recited accession number is incorporated herein by reference) which is described therein as "SOX30 protein [Homo Sapiens]". A partial alignment demonstrating the observed homology is shown immediately below.
>dbj~BAA37146.1~ (AB022083) SOX30 protein [Homo sapiens]
>sp~BAA371461BAA37146 SOX30 protein.
Length = 753 2S Plus Strand HSPs:
Score = 687 (241.8 bits), Expect = 6.7e-98, Sum P(3) = 6.7e-98 Identities = 140/170 (82$), Positives = 146/170 (85$), Frame = +2 3O Q: 242 QFRPDLRLLLPPASSEGAPCRPELHPVQPRALHVKAKK*ELGACLDASVRPRGAVETGRR 421 QFRPDLRLL PP +S+GA RPELHPVQP ALHVKAKK +LG LD SV PRGAVETG R
S: 102 QFRPDLRLLQPPTASDGATSRPELHPVQPLALHVKAKKQKLGPSLDQSVGPRGAVETGPR 161 Q: 422 ASRAVKLEGLGPALDYFRGNEKGKLEAEEVMRDAMQGGEGKSSAAIREGVIKTEEPERLL 601 3S ASR VKLEG GPAL YFRG+EKGKLEAEEVMRD+MQGG GKS AAIREGVIKTEEPERLL

S: 162 ASRVVKLEGPGPALGYFRGDEKGKLEAEEVMRDSMQGGAGKSPAAIREGVIKTEEPERLL 221 Q: 602 EDCRLGAEPASNGPARGSAEVILAPTSGAFGPHQQDLRIPLDSPHCFPWG 751 EDCRLGAEPASNG GSAEVILAPTSGAFGPHQQDLRIPL + H P G
S S: 222 EDCRLGAEPASNGLVHGSAEVILAPTSGAFGPHQQDLRIPL-TLHTVPPG 270 The segment of dbjIBAA37146.11 that is shown as "S" above is set out in the sequence listing as SEQ ID NO. 1S9 . 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. 160 and/or SEQ
1S ID NO. 169 which corresponds to the "Q" sequence in the alignment shown above (gaps introduced in a sequence by the computer are, of course, removed).
It has been discovered that this gene is expressed primarily in the following tissues/cDNA libraries: Human Testes, Reexcision; Human adult testis, large inserts and to a lesser extent in Human Adult Testes, Large Inserts, Reexcision; Human Testes.
Preferred polypeptides of the present invention comprise immunogenic epitopes shown in SEQ ID NO: 103 as residues: Pro-7S to Ser-93.
Polynucleotides encoding said polypeptides are also encompassed by the invention. Antibodies that bind polypeptides of the invention are encompassed by the invention, particularly 2S antibodies that inhibit activity.
Many polynucleotide sequences, such as EST sequences, are publicly available and accessible through sequence databases. Some of these sequences are related to SEQ ID NO:S4 and may have been publicly available prior to conception of the present invention. Preferably, such related polynucleotides ace specifically excluded from the scope of the present invention. To list every related sequence would be cumbersome. Accordingly, preferably excluded from the present invention are one or more polynucleotides comprising a nucleotide sequence described by the general formula of a-b, where a is any integer between 1 to 868 of SEQ ID
N0:54, b is an integer of 15 to 882, where both a and b correspond to the positions of nucleotide residues shown in SEQ ID N0:54, and where b is greater than or equal to a + 14.
FEATURES OF PROTEIN ENCODED BY GENE NO: 45 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.
dbjIBAA32700.11 (all information available through the recited accession number is incorporated herein by reference) which is described therein as "beta-spectrin III
[Homo Sapiens]". A partial alignment demonstrating the observed homology is shown immediately below.
>dbj~BAA32700.1~ (AB008567) beta-spectrin III [Homo Sapiens]
>sp~015020~015020 BETA-SPECTRIN III. >gb~AAC79502.11 (AF026487) beta III
spectrin [Homo sapiens] {SUB 1900-2390} >gb~AAC79503.1~ (AF026488) beta III
spectrin [Homo Sapiens] {SUB 2138-2390} >gb~AAC80006.1 (AF079569) beta III spectrin [Homo Sapiens] {SUB 1-34}
>sp~G248095~G248095 SPECTRIN BETA SUBUNIT=28 KDA FRAGMENT. {SUB 50-69}
Length = 2390 Plus Strand HSPS:
Score = 760 (267.5 bits), Expect = 4.4e-73, P = 4.4e-73 Identities = 148/148 (100$), Positives = 148/148 (100 0 , Frame = +3 Q: 9 RKQEMEAFGKKAANRSWQNVYCVLRRGSLGFYKDAKAASAGVPYHGEVPVSLARAQGSVA 188 RKQEMEAFGKKAANRSWQNVYCVLRRGSLGFYKDAKAASAGVPYHGEVPVSLARAQGSVA
S: 2227 RKQEMEAFGKKAANRSWQNVYCVLRRGSLGFYKDAKAASAGVPYHGEVPVSLARAQGSVA 2286 S Q: 189 FDYRKRKHVFKLGLQDGKEYLFQAKDEAEMSSWLRVVNAAIATASSASGEPEEPWPSTT 368 FDYRKRKHVFKLGLQDGKEYLFQAKDEAEMSSWLRVVNAAIATASSASGEPEEPWPSTT
S: 2287 FDYRKRKHVFKLGLQDGKEYLFQAKDEAEMSSWLRVVNAAIATASSASGEPEEPWPSTT 2346 Q: 369 RGMTRAMTMPPVSPVGAEGPVVLRSKDG 452 S: 2347 RGMTRAMTMPPVSPVGAEGPWLRSKDG 2374 The segment of dbjIBAA32700.11 that is shown as "S" above is set out in the sequence listing as SEQ ID NO. 161. Based on the structural similarity, these 1S 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 20 amino acid sequence set out in the sequence listing as SEQ ID NO. 162 which corresponds to the "Q" sequence in the alignment shown above (gaps introduced in a sequence by the computer are, of course, removed).
It has been discovered that this gene is expressed primarily in the following tissues/cDNA libraries: Human adult testis, large inserts and to a lesser extent in 2S Human Adult Testes, Large Inserts, Reexcision; Soares adult brain N2bSHBSSY;
Human Fetal Heart; Soares testis NHT; Merkel Cells; Soares infant brain 1NIB;
Human Fetal Heart, Differential (Fetal-Specific); H. Striatum Depression, subt; H.
cerebellum, Enzyme subtracted; Soares adult brain N2b4HBSSY; Human Whole Brain #2 - Oligo dT > 1.SKb; Jurkat T-Cell, S phase; CD34 depleted Buffy Coat 30 (Cord Blood); TF-1 Cell Line GM-CSF Treated;
Soares fetal liver spleen_1NFLS S1; Stratagene pancreas (#937208); Human Hypothalmus,Schizophrenia; Human Fetal Brain; Human Primary Breast Cancer Reexcision; H. Frontal cortex,epileptic,re-excision and Human Cerebellum.
Preferred polypeptides of the present invention comprise immunogenic epitopes shown in SEQ ID NO: 104 as residues: Ser-56 to Arg-67.
Polynucleotides encoding said polypeptides are also encompassed by the invention. Antibodies that bind polypeptides of the invention are encompassed by the invention, particularly antibodies that inhibit activity.
Many polynucleotide sequences, such as EST sequences, are publicly available and accessible through sequence databases. Some of these sequences are related to SEQ ID NO:55 and may have been publicly available prior to conception of the present invention. Preferably, such related polynucleotides are specifically excluded from the scope of the present invention. To list every related sequence would be cumbersome. Accordingly, preferably excluded from the present invention are one or more polynucleotides comprising a nucleotide sequence described by the general formula of a-b, where a is any integer between 1 to 1115 of SEQ ID
NO:55, b is an integer of 15 to 1129, where both a and b correspond to the positions of nucleotide residues shown in SEQ ID NO:55, and where b is greater than or equal to a + 14.
FEATURES OF PROTEIN ENCODED BY GENE NO: 46 It has been discovered that this gene is expressed primarily in the following tissues/cDNA libraries: human tonsils; CD34 depleted Buffy Coat (Cord Blood), re-excision.
Many polynucleotide sequences, such as EST sequences, are publicly available and accessible through sequence databases. Some of these sequences are related to SEQ ID N0:56 and may have been publicly available prior to conception of the present invention. Preferably, such related polynucleotides are specifically excluded from the scope of the present invention. To list every related sequence would be cumbersome. Accordingly, preferably excluded from the present invention are one or more polynucleotides comprising a nucleotide sequence described by the general formula of a-b, where a is any integer between 1 to 1368 of SEQ ID
N0:56, b is an integer of 15 to 1382, where both a and b correspond to the positions of nucleotide residues shown in SEQ ID N0:56, and where b is greater than or equal to a + 14.
FEATURES OF PROTEIN ENCODED BY GENE NO: 47 It has been discovered that this gene is expressed primarily in the following tissues/cDNA libraries: NCI CGAP_GCB1; Soares breast 3NbHBst; Dendritic cells, pooled; Human Bone Marrow, treated and to a lesser extent in NCI CGAP_Pr6;
NCI CGAP_PrB; Tongue Normal; Hepatocellular Tumor; Human Stomach,re-excision; H. Meningima, M1; Atrium cDNA library Human heart;
NCI CGAP_GCB1; Human Umbilical Vein Endothelial Cells, uninduced; Soares adult brain N2b5HBS5Y; Human Thymus Stromal Cells; Smooth muscle, serum induced,re-exc; Resting T-Cell Library,II; Soares fetal lung NbHLI9W; Human Eosinophils; Human Placenta; Human Testes Tumor; Soares melanocyte 2NbHM;
Endothelial-induced; Human Adult Pulmonary,re-excision;
Soares senescent fibroblasts NbHSF; Monocyte activated; Stratagene hNT neuron (#937233); Spleen, Chronic lymphocytic leukemia and T cell helper II.
Many polynucleotide sequences, such as EST sequences, are publicly available and accessible through sequence databases. Some of these sequences are related to SEQ ID N0:57 and may have been publicly available prior to conception of the present invention. Preferably, such related polynucleotides are specifically excluded from the scope of the present invention. To list every related sequence would be cumbersome. Accordingly, preferably excluded from the present invention are one or more polynucleotides comprising a nucleotide sequence described by the general formula of a-b, where a is any integer between 1 to 2350 of SEQ ID
N0:57, b is an integer of 15 to 2364, where both a and b correspond to the positions of nucleotide residues shown in SEQ ID N0:57, and where b is greater than or equal to a + 14.
FEATURES OF PROTEIN ENCODED BY GENE NO: 48 It has been discovered that this gene is expressed primarily in the following tissues/cDNA libraries: HUMAN JURKAT MEMBRANE BOUND POLYSOMES
and to a lesser extent in Human Eosinophils; Soares breast 3NbHBst; Primary Dendritic Cells, lib 1; Human promyelocyte; CD34+ cell, I; H Umbilical Vein Endothelial Cells, frac A, re-excision; H Female Bladder, Adult; Myoloid Progenitor Cell Line; NCI CGAP_GCB1; Human Manic Depression Tissue; Human Activated Monocytes; Resting T-Cell Library,II; NCI CGAP_GC4; Dendritic cells, pooled;
Primary Dendritic cells,frac 2; Human Neutrophil, Activated; Spleen, Chronic lymphocytic leukemia and neutrophils control.
Preferred polypeptides of the present invention comprise immunogenic epitopes shown in SEQ ID NO: 107 as residues: Asp-33 to Arg-40.
Polynucleotides encoding said polypeptides are also encompassed by the invention. Antibodies that bind polypeptides of the invention are encompassed by the invention, particularly antibodies that inhibit activity.
Many polynucleotide sequences, such as EST sequences, are publicly available and accessible through sequence databases. Some of these sequences are related to SEQ ID N0:58 and may have been publicly available prior to conception of the present invention. Preferably, such related polynucleotides are specifically excluded from the scope of the present invention. To list every related sequence would be cumbersome. Accordingly, preferably excluded from the present invention are one or more polynucleotides comprising a nucleotide sequence described by the general formula of a-b, where a is any integer between 1 to 2562 of SEQ ID
N0:58, b is an integer of 15 to 2576, where both a and b correspond to the positions of nucleotide residues shown in SEQ ID N0:58, and where b is greater than or equal to a + 14.

I~ l/7 M ~ -~ 01 M

O O ~ ~ I~ .~ M N ,~ M

'L3 4~

O

N ~1 N ~ N N M

d ~ 0., N N o0 ~O ~ N 01 d O ~ ~ N ~ N ~ N N N

.~.,n d c+-, b p Q., _ w zo~~~~ ~ o M

~

p.., .-..~ ,~ ,~ N '-.
d v~

E ~ ~n ~ ~ ~, O
-~ ~ p ~ M V7 l~ M N r, N
r~ ~ y~ 'O o av .-. ,~ ,~ N

~n ~ CT' ' ~ 01 M

4r ,~ N ~ p\ ~ ~ I~ V' r~ O O ~ ' O <f ~ 00 N O ~ ~ ~

M N N ~ M N N

N
~ 4-a ~ CS' ~ ~ N ~O
-. , r 00 0o . ~ . ., r l~ ~O M ~ ~ ~
H ~ ~

+, .~ N I C~ O ~ N 00 z O

H ~ N c~ ~ M N ~ ~ ~ N
~

N 01 M ~ ~ ~O I~ 00 Q1 za~zx ~C ?C x X x ~C ~C x x d d d ~ d d d d d N N N N N N N N N

~ oho ~ ~ oar U v ~ ~ ~

, . a ~
a . .

p H is. N '~ d Q Q Q, d Q, Q, Q Q d o o o o o o o o o o ~'' 0.o o o o o o o o o 0 ~' ~ ~ ~ ~ ~ ~ ~

a a ., n. a .., ..~ n a .. . a , a a . ., M
N ~ ~ O

~ ~ p p w ~ U ~ c~ U a ~ o ~ ~ a x x x x x x x x x N N M ~ ~n ~D l~ oo O~

O ~ ~ M V'1M ~ ~ O

M V1 M M \O N ~ V1 Y

~O M 01 V1 ~D O~ ~ M

N N -r N N

d '+v ~ ~ v~ N oo ~ v~ oo ~n N

-a d' ~ N N ~ N N

m 4-a ~d d a ~ ~ ,..~r., .~ .~ r., .~ ~ r.. ,~

~ ~ ~ ~ ~ ~ ~

_~
O

4r ~ ~ ~ ~t N ~ O~ 00 V7 00 ~ ' ~
N

~ ~D V1 O ~ d l ~--~M

~, ~ C~ '_' N '-'M I~ N

z ~ ~ o ~b o ~

N .~ M l~ N

'f" p-' O~ ~ O

M ~ ~D l~ ~ l~ M

0o O ~ ~ N Q> O

z oo ~
U ~

n l~ M O l~ 01 M .~
~

O (~ ~ ~ 00 O M ~ ~ Ov O
~

~-, z C/~ N .-i ~ .~ .-.r,~ ~ (~l .~ N

ON N N N N N N N N N

r'~C~C ~ ~ ~G' d d ,'~G'o ,'~' y, . . d C/~
O p O

d d O ~ cy--,c d d '~ ~-.~ '' ~

N N ~ ~ N ~ ~ N ~ N
~' o, ~ .--,.-- r- ~ ~ .--~~ .--U W o o, o, a, o~ o, o, o, o, o~
~ ~ o o ~ ~
o o , ~ a. o, ~ ~ ~ ~ a, ~ , ~ , ~ , o, o, o, o, U
' H H N H ~ H H H H H
~ ~ ~

o ~ ~ ~ ~ ~ ~ o d ~1 z A..,c1. A." a. a. a. a. a. p..,c~.., O o o o o o O o o o ~ o d W w U ~

~

~ ~ ~ 0 0 ~ x ~ ~ x _ " U

x x x x x x x a~

O O --~ N M ~ ~n \O t~ 00 O~

N
d 4., ~ ~ .-. tm n t~ W n t~

~' O O .~ M M 00 ~ M , ~ M ~O
~, "G

N

\D 00 N 01 O 00 ~ 00 M .--~N

d 4-r ~ O" ~!1 l~ ~ 00 01 I~ M l~

o ~ ~ ,~ r. r, .-, ,~ .~

an , w z ~ ~ ~ ~ M
o~
~~

~ M M O N ~ O

_ ~ M M N
d C/~ ~

~-' ~'' _ .-r z M
"

~ ~ V1 ~ M M O N ~ ~ O
O

M M N

U

H ~ 00 00 I~ ~O ~ O M -z o o ~ ~ N o, oo .~ O ~ ,M N o O ~ ~ ' -~ ~

. M ~ M
M U C/~ N N .~ .-~ .~ ,~ .~ ~ N

z o o ~ o, ~ .~ .-..
N N

00 00 l~ N ~ O M M - O

O z ~ O ~ ~n ~ ~ ~ ~ ~ M

E-~ C/~ N N .~ .-~ ,~ ,~ .~ N

O O

M M M M M M M M M M

O ~ ~ ~ ~
O
'"

d d d d d r N N ~ N N N N N N

~

u, ~~ ~~ ~'.'~~ ~~ ~~ ~~ ~~ U

~ o, ~ ~ ~ -~ ~ ~ ~ ~
~ o~ oo o~ o, o~ o~ o~ o, o, o.
U . 00 a 00 00 00 00 00 00 00 00 0, 0, 0 0 0 0, 0 0 0 ~ . , , , , , , c~ aJ

U o a H W a H a W a H a ' ~

o -~ ~ o -~ ~ -~ o -,, ~ E
o ~ ~ ~ -~
E E E E o p..,C~, A.. ~ ci.,A." ci.a.. a. p..~
o o o O O O o O O O

~l1 N ,~ 00 ~l N ~ ~

z ~ ~

v ~ .7 C ~ d ~ ~ ~ C .7 r~ ~ c~ c~ c~ x d w o _ x ~ ~ ~ ~ x x x x a~

O

N N N N N N N N N N
G~
z ~O D1 ~ ~

N l~ ~O

"CS

N

O N N ~ N N

N ~ c N -~ O~ v n N N M ~ N M

4, ~ ~ ~N .-, .~ .--r.-.-~,~ r., w ~~ ~ ~ ~ ~
zo~

d ~ ~ ~ N N

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

~

z 4'..' ~n ct ~ ~ O N
b N .~ .~ ~ v1 N ,_, U

~D ~ N N
O ~
z ~

p _ N p ~ ~D '_" 0o ~ p~ N M

M U ~ ~ N .~ N ~O N ,-, 00 ~,, ~n N

z~
o _ U ~

~ E""' ~ N l~ O~ O ~ ~ ~ ~ ~

M
O ~

~ oo N ~O N ~ 00 ~ ~n x x x x x x x o o N u, s~.
o o ,~ N ~ ~ N N N N N N
~ M ~'' '~

r- ~ .-- r- ~ '- --~ ~ ~ .--U ' c~ o, o~ a, a, o~ a~ o~ o, ov ~ o, ~ , a , o , , v, ~ ~ ~ ~ ~ \. ~ ' ~ ~
~ a, o o, o ~ o ~ a, o U o ~' . . . . . . . . .
~ r~ ~ t~ ~ ~ ~ t~ r- H
a H H H a a a H a ~

d E o ~ o E- F ~ o E
-yy ~ -~ o -~
~ ~ E- ~

C~ Z o., 0. p~ a" a, ci.,p..,ci.,a., A.., O O O O O O O O O O

O 01 ~ 00 ~.,~~' 00 ~ .

~ d ~

Z ~ v w r~_o o' ~ ~ ~ c~ o ~ ~ Q
~

~ x x x x x x x x x x O .~ N M ~ ~n ~O t~ 00 01 M M M M M M M M M M

~ M ~1 V1 v~ N

O O N o ~ ~
o N
O N

~ ~ ~ v1 ,~ .~ p d V ~ N N N M N
~

' d ~ a,, ~ cue, b0 O O O Q\

N

~, on ~.

..,.~
w ~

~ ~ 0 0 0 0 0 0 0 0 ~o~

z H o ~ ~ o ~ o zo~
~

d O v0 o0 CU

_ r., N N ,-, ~ N
d C/~

' O ~

z ~ ~ ~ O ~ o0 N
b N N ~ ~ N

p N
~ N

z~ON M O N pp N O N
O

N M N O w-~ M M ~
~ ~

c~ (~ N ~ N N ,~ .~ N N

Z a o ~ M .., ~ -. . , a, .

~ E-~ ~' ~ O N N

N O N N O
Z O O

E-r N ~ N N ~ .~ ~ N N
~

za~zx ~C x X ~C x >C ~C x N

d d d d d d d d N N N N N N N N

...'.,: : .:, ~. ,~ .~ ~. r..r.
U'v o o~ o~ o~ o~ o, o~
~ o ~ o , , ~o~ ~o. ~ ~o, ~o~.~ ~o, ~o ' ~ ~ , ~

U
E-~ Q., N d Q d d d d d d d c~ o o o o o o o o o ' o ~ H Wo H E- Wo E-' E-' Wo H
~ ~ ~ D ~ ~

p..,A. A..,A..,0., rs. p, Ci.ci.
o o o 0 o O O o o ~ M

z ~ ~

~ Q

r~ o ~ ~ x a ~ o ~ ~ x x x x x x x x x ~

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 ID NO:X.
The cDNA Clone ID 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 1D 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 I.
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.
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 ID 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 Seduences 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 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 1D 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 occurring 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 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, and the polynucleotides encoding such polypeptides, are contemplated by the present invention.

Polvnucleotide and Poly~eptide Variants The present invention is directed to variants of the polynucleotide sequence disclosed in SEQ ID 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 ID 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 ID 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 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' 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 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 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 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°Io 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 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 S' 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 ID 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-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, I-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 occurring protein. For example, Gayle and coworkers (J. Biol. Chem 268:22105-22111 (1993)) conducted extensive mutational analysis of human cytokine IL-la. They used random mutagenesis to generate over 3,500 individual IL-la 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 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 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, 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 acids, such as, for example, an IgG Fc fusion region peptide, or leader or secretory sequence, or a sequence facilitating purification. 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 Poly~e~tide 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 nt, 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, 1201-1250, 1251-1300, 1301-1350, 1351-1400, 1401-1450, 1451-1500, 1501-1550, 1551-1600, 1601-1650, 1651-1700, 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 ID 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 (S, 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.
1n 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 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
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 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 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 S 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. lmmunospecific 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).
1n 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 carrier. 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 carrier, 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 maleimidobenzoy(- 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 y~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, lgE, IgG, IgM), or portions thereof (CH1, CH2, CH3, or any combination thereof and portions thereof) 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 barrier 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 1gG 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
ID 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 ID 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 limited 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-ld 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, lgE, IgM, lgD, IgA and IgY), class (e.g., IgGI, IgG2, lgG3, IgG4, IgAI and IgA2) or subclass of immunoglobulin molecule.
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 102 M, 10-2 M, 5 X 10-3 M, 10-3 M, 5 X 10-4 M, 10-4 M, 5 X 10-5 M, 10-5 M, 5 X 10-6 M, 10-6M, 5 X 10-' M, 10' M, 5 X 10-8 M, 10-g M, 5 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;
7; 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; 5,530,101; and 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.S. 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 S 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.
Pol~nucleotides 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 ID 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 5' 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"
(Morrison 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.S. 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 GST moiety.
In an insect system, Autographs 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 adenovirus 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 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 S 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 1251, 131I, 111In 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, S-fluorouracil decarbazine), alkylating agents (e.g., mechlorethamine, thioepa chlorambucil, melphalan, carmustine (BSNU) and lomustine (CCNU), 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 ("1L-2"), interleukin-6 ("1L-6"), r 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 Carriers 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, immunodiffusion 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. 1, 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-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 l71 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 10-2 M, 10-2 M, 5 X 10-3 M, 10-3 M, M, 10-4 M, 5 X 10-5 M, 10-5 M, 5 X 10-6 M, 10-6 M, 5 X 10-' M, 10-' M, 5 X
10~g M, 10-g M, S X 10-9 M, 10-9 M, 5 X 10-'° M, 10-'° M, S X 10-"~ M, 10-1' M, 5 X 10-'Z M, 10~
1z M, 5 X 10-13 M, 10-'3 M, 5 X 10-'4 M, 10-14 M, 5 X 10-'S M, and 10-15 M.
Gene Theranv 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 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.
1n 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 transferring 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, S 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.
Tn 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.
Therapeutic/Prophylactic 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 carrier. 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 carriers 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 carriers, 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 S 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 lm~ina 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 (R1A). Suitable antibody assay labels are known in the art and include enzyme labels, such as, glucose oxidase;
radioisotopes, such as iodine (125I, 1211), 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 carried 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. 1n 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 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 po(ypeptide 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).) 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 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 Sf9 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, lnc.; pBluescript vectors, Phagescript vectors, pNHBA, pNH 16a, pNH 18A, pNH46A, available from Stratagene Cloning Systems, lnc.; and ptrc99a, pKK223-3, pKK233-3, pDR540, pRITS available from Pharmacia Biotech, Inc. Among preferred eukaryotic vectors are pWLNEO, pSV2CAT, pOG44, pXT 1 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-S 1, 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 canon 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-s 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 O2. 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 AOXl 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 AOXl 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 AOXI
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, pTEFl/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 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).
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;
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.

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.
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, 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 20l 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 occurring 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 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 Polvnucleotides 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 ID 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, and preselection by hybridization to construct chromosome specific-cDNA libraries.
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). Preferred polynucleotides correspond to the noncoding regions of the cDNAs because the coding sequences are more likely conserved within gene families, thus increasing the chance of cross hybridization during chromosomal mapping.
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 50-500 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 not 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 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 IS 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 15-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 riiammals. 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 1., 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 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 Poly~eptides 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 (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 (1251, 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, 1311, 112In, 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 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).) 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 S 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 pEFI/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 IS 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 50 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 S on the injection of genetic material, complexed with cationic liposomes carriers, into mice. U.S. 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 (fines 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, RCRIP, 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 L1 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 a 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 The polynucleotides or polypeptides, 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 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 or some autoimmune diseases, disorders,and/or conditions, acquired (e.g., by chemotherapy or toxins), or infectious.
Moreover, a polynucleotides or polypeptides, or agonists or antagonists of the present invention can be used as a marker or detector of a particular immune system disease or disorder.
A polynucleotides or polypeptides, 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. A polynucleotides or polypeptides, 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, HIV infection, HTLV-BLV infection, leukocyte adhesion deficiency syndrome, lymphopenia, phagocyte bactericidal dysfunction, severe combined immunodeficiency (SCIDs), Wiskott-Aldrich Disorder, anemia, thrombocytopenia, or hemoglobinuria.
Moreover, a polynucleotides or polypeptides, 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, a polynucleotides or polypeptides, 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, a polynucleotides or polypeptides, 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.
A polynucleotides or polypeptides, or agonists or antagonists of the present invention may also be useful in treating, preventing, and/or diagnosing autoimmune diseases, disorders, and/or conditions. Many autoimmune diseases, disorders, and/or conditions 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 a polynucleotides or polypeptides, or agonists or antagonists of the present invention that inhibits an immune response, particularly the proliferation, differentiation, or chemotaxis of T-cells, may be an effective therapy in preventing autoimmune diseases, disorders, and/or conditions.
Examples of autoimmune diseases, disorders, and/or conditions that can be treated, prevented, and/or diagnosed or detected by the present invention 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.
Similarly, allergic reactions and conditions, such as asthma (particularly allergic asthma) or other respiratory problems, may also be treated, prevented, and/or diagnosed by polynucleotides or polypeptides, or agonists or antagonists of the present invention. Moreover, these molecules can be used to treat anaphylaxis, hypersensitivity to an antigenic molecule, or blood group incompatibility.
A polynucleotides or polypeptides, or agonists or antagonists of the present invention may also be used to treat, prevent, and/or diagnose organ rejection or graft-s versus-host disease (GVHD). 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. The administration of a polynucleotides or polypeptides, or agonists or antagonists of the present invention that inhibits an immune response, particularly the proliferation, differentiation, or chemotaxis of T-cells, may be an effective therapy in preventing organ rejection or GVHD.
Similarly, a polynucleotides or polypeptides, or agonists or antagonists of the present invention may also be used to modulate inflammation. For example, the polypeptide or polynucleotide or agonists or antagonist may inhibit the proliferation and differentiation of cells involved in an inflammatory response. These molecules can be used to treat, prevent, and/or diagnose inflammatory conditions, both chronic and acute conditions, including chronic prostatitis, granulomatous prostatitis and malacoplakia, 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, or resulting from over production of cytokines (e.g., TNF or IL-1.) Hvnerproliferative 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 hyperproliferative 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 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, 5X10-'M, 10~'M, gM, 10-8M, SX10-9M, 10-9M, 5X10-'°M, 10-'°M, 5X10-"M, 10-"M, SX10-'ZM, 10~'ZM, 5X10-'3M, 10-'3M, 5X10-'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 atone or in combination with small molecule drugs or adjuviants, such as apoptonin, galectins, thioredoxins, antiinflammatory proteins (See for example, Mutat Res 400(1-2):447-55 (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 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.
lschemia 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-Angio,~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 keratitis, leishmaniasis and onchocerciasis), immunological processes (e.g., graft rejection and DEMANDES OU BREVETS VOLUMINEUX
LA PRESENTE PARTIE I)E CETTE DEMANDE OU CE BREVETS
COMPRI~:ND PLUS D'UN TOME.
CECI EST ~.E TOME 1 DE 2 NOTE: Pour les tomes additionels, veillez contacter 1e Bureau Canadien des Brevets.
JUMBO APPLICATIONS / PATENTS
THIS SECTION OF THE APPLICATION / PATENT CONTAINS MORE
THAN ONE VOLUME.

NOTE: For additional vohxmes please contact the Canadian Patent Oi~ice.

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.
CA002382743A 1999-06-11 2000-06-01 48 human secreted proteins Abandoned CA2382743A1 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US13862799P 1999-06-11 1999-06-11
US60/138,627 1999-06-11
PCT/US2000/014964 WO2000077023A1 (en) 1999-06-11 2000-06-01 48 human secreted proteins

Publications (1)

Publication Number Publication Date
CA2382743A1 true CA2382743A1 (en) 2000-12-21

Family

ID=22482883

Family Applications (1)

Application Number Title Priority Date Filing Date
CA002382743A Abandoned CA2382743A1 (en) 1999-06-11 2000-06-01 48 human secreted proteins

Country Status (5)

Country Link
EP (1) EP1192171A4 (en)
JP (1) JP2003508020A (en)
AU (1) AU5308300A (en)
CA (1) CA2382743A1 (en)
WO (1) WO2000077023A1 (en)

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6511834B1 (en) 2000-03-24 2003-01-28 Millennium Pharmaceuticals, Inc. 32142,21481,25964,21686, novel human dehydrogenase molecules and uses therefor
US6627423B2 (en) 2000-03-24 2003-09-30 Millennium Pharmaceuticals, Inc. 21481, a novel dehydrogenase molecule and uses therefor
US6613554B2 (en) 2001-03-26 2003-09-02 Applera Corporation Isolated human enzyme, nucleic acid molecules encoding human enzyme, and uses thereof

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0679716A4 (en) * 1993-11-12 1999-06-09 Kenichi Matsubara Gene signature.
WO1998054963A2 (en) * 1997-06-06 1998-12-10 Human Genome Sciences, Inc. 207 human secreted proteins
DE10021834A1 (en) * 2000-05-06 2001-11-15 Lynx Therapeutics Gmbh New mRNA indicative of T cell activation and functional status, useful for diagnosis and therapy e.g. of autoimmunity or transplant rejection

Also Published As

Publication number Publication date
WO2000077023A1 (en) 2000-12-21
JP2003508020A (en) 2003-03-04
EP1192171A1 (en) 2002-04-03
EP1192171A4 (en) 2003-05-07
AU5308300A (en) 2001-01-02

Similar Documents

Publication Publication Date Title
CA2362423A1 (en) 47 human secreted proteins
CA2383041A1 (en) 49 human secreted proteins
CA2382185A1 (en) 48 human secreted proteins
CA2370489A1 (en) 49 human secreted proteins
CA2364209A1 (en) 49 human secreted proteins
CA2387696A1 (en) 48 human secreted proteins
CA2365223A1 (en) 46 human secreted proteins
CA2370767A1 (en) 49 human secreted proteins
CA2368281A1 (en) 50 human secreted proteins
CA2391364A1 (en) 47 human secreted proteins
CA2368467A1 (en) 50 human secreted proteins
CA2383828A1 (en) 26 human secreted proteins
CA2365522A1 (en) 48 human secreted proteins
CA2364635A1 (en) 47 human secreted proteins
CA2368302A1 (en) 49 human secreted proteins
CA2391361A1 (en) 47 human secreted proteins
CA2371172A1 (en) 50 human secreted proteins
CA2390946A1 (en) 49 human secreted proteins
CA2382743A1 (en) 48 human secreted proteins
CA2365247A1 (en) 49 human secreted proteins
CA2365905A1 (en) 48 human secreted proteins
CA2370189A1 (en) 48 human secreted proteins
CA2383048A1 (en) 49 human secreted proteins
CA2370562A1 (en) 49 human secreted proteins
CA2368916A1 (en) 49 human secreted proteins

Legal Events

Date Code Title Description
FZDE Dead