CA2448073A1 - Therapeutic polypeptides, nucleic acids encoding same, and methods of use - Google Patents

Therapeutic polypeptides, nucleic acids encoding same, and methods of use Download PDF

Info

Publication number
CA2448073A1
CA2448073A1 CA002448073A CA2448073A CA2448073A1 CA 2448073 A1 CA2448073 A1 CA 2448073A1 CA 002448073 A CA002448073 A CA 002448073A CA 2448073 A CA2448073 A CA 2448073A CA 2448073 A1 CA2448073 A1 CA 2448073A1
Authority
CA
Canada
Prior art keywords
polypeptide
novx
nucleic acid
protein
cell
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
CA002448073A
Other languages
French (fr)
Inventor
David W. Anderson
Jason C. Baumgartner
Ferenc L. Boldog
Stacie J. Casman
Schlomit R. Edinger
Esha A. Gangolli
Valerie L. Gerlach
Linda Gorman
Xiaojia Guo
Tord Hjalt
Ramesh Kekuda
Li Li
John R. Macdougall
Uriel M. Malyankar
Isabelle Millet
Muralidhara Padigaru
Meera Patturajan
Carol E. A. Pena
Luca Rastelli
Richard A. Shimkets
David J. Stone
Kimberly A. Spytek
Corine A. M. Vernet
Edward Z. Voss
Bryan D. Zerhusen
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.)
CuraGen Corp
Original Assignee
Curagen Corporation
David W. Anderson
Jason C. Baumgartner
Ferenc L. Boldog
Stacie J. Casman
Schlomit R. Edinger
Esha A. Gangolli
Valerie L. Gerlach
Linda Gorman
Xiaojia Guo
Tord Hjalt
Ramesh Kekuda
Li Li
John R. Macdougall
Uriel M. Malyankar
Isabelle Millet
Muralidhara Padigaru
Meera Patturajan
Carol E. A. Pena
Luca Rastelli
Richard A. Shimkets
David J. Stone
Kimberly A. Spytek
Corine A. M. Vernet
Edward Z. Voss
Bryan D. Zerhusen
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from US10/162,335 external-priority patent/US7034132B2/en
Application filed by Curagen Corporation, David W. Anderson, Jason C. Baumgartner, Ferenc L. Boldog, Stacie J. Casman, Schlomit R. Edinger, Esha A. Gangolli, Valerie L. Gerlach, Linda Gorman, Xiaojia Guo, Tord Hjalt, Ramesh Kekuda, Li Li, John R. Macdougall, Uriel M. Malyankar, Isabelle Millet, Muralidhara Padigaru, Meera Patturajan, Carol E. A. Pena, Luca Rastelli, Richard A. Shimkets, David J. Stone, Kimberly A. Spytek, Corine A. M. Vernet, Edward Z. Voss, Bryan D. Zerhusen filed Critical Curagen Corporation
Publication of CA2448073A1 publication Critical patent/CA2448073A1/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
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; CARE OF BIRDS, FISHES, INSECTS; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2217/00Genetically modified animals
    • A01K2217/05Animals comprising random inserted nucleic acids (transgenic)
    • 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
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • 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

Disclosed herein are nucleic acid sequences that encode G-coupled protein-receptor related polypeptides. Also disclosed are polypeptides encoded by these nucleic acid sequences, and antibodies, which immunospecifically-bind to the polypeptide, as well as derivatives, variants, mutants, or fragments of the aforementioned polypeptide, polynucleotide, or antibody. The invention further discloses therapeutic, diagnostic and research methods for diagnosis, treatment, and prevention of disorders involving any one of these novel human nucleic acids and proteins.

Description

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

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

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

THERAPEUTIC POLYPEPTIDES, NUCLEIC ACIDS
ENCODING SAME, AND METHODS OF USE
FIELD OF THE INVENTION
The present invention relates to novel polypeptides, and the nucleic acids encoding them, having properties related to stimulation of biochemical or physiological responses in a cell, a tissue, an organ or an organism. More particularly, the novel polypeptides are gene products of novel genes, or are specified biologically active fragments or derivatives thereof. Methods of use encompass diagnostic and prognostic assay procedures as well as methods of treating diverse pathological conditions.
BACKGROUND OF THE INVENTION
Eukaryotic cells are characterized by biochemical and physiological processes, which under normal conditions are exquisitely balanced to achieve the preservation and propagation of the cells. When such cells are components of multicellular organisms such as vertebrates or, more particularly, organisms such as mammals, the regulation of the biochemical and physiological processes involves intricate signaling pathways.
Frequently, such signaling pathways include constituted of extracellular signaling proteins, cellular receptors that bind the signaling proteins and signal transducing components located within the cells.
Signaling proteins may be classified as endocrine effectors, paracrine effectors or autocrine effectors. Endocrine effectors are signaling molecules secreted by a given organ into the circulatory system, which are then transported to a distant target organ or tissue.
The target cells include the receptors for the endocrine effector, and when the endocrine effector binds, a signaling cascade is induced. Paracrine effectors involve secreting cells ' and receptor cells in close proximity to each other, such as two different classes of cells in the same tissue or organ. Qne class of cells secretes the paracrine effector, which then reaches the second class of cells, for example by diffusion through the extracellular fluid.
The second class of cells contains the receptors for the paracrine effector;
binding of the effector results in induction of the signaling cascade that elicits the corresponding biochemical or physiological effect. Autocrine effectors are highly analogous to paracrine efFectors, except that the same cell type that secretes the autocrine effector also contains the receptor. Thus the autocrine effector binds to receptors on the same cell, or on identical neighboring cells. The binding process then elicits the characteristic biochemical or physiological effect.
Signaling processes may elicit a variety of effects on cells and tissues including, by way of nonlimiting example, induction of cell or tissue proliferation, suppression of growth or proliferation, induction of differentiation or maturation of a cell or tissue, and suppression of differentiation or maturation of a cell or tissue.
Many pathological conditions involve dysregulation of expression of important effector proteins. In certain classes of pathologies the dysregulation is manifested as diminished or suppressed level of synthesis and secretion of protein effectors. In other classes of pathologies the dysregulation is manifested as increased or up-regulated level of synthesis and secretion of protein effectors. In a clinical setting a subject may be suspected of suffering from a condition brought on by altered or mis-regulated levels of a protein effector of interest. Therefore there is a need to assay for the level of the protein effector of interest in a biological sample from such a subject, and to compare the level with that characteristic of a nonpathological condition. There also is a need to provide the protein effector as a product of manufacture. Administration of the effector to a subject in need thereof is useful in treatment of the pathological condition.
Accordingly, there is a
2 need for a method of treatment of a pathological condition brought on by a diminished or suppressed levels of the protein effector of interest. In addition, there is a need for a method of treatment of a pathological condition brought on by a increased or up-regulated levels of the protein effector of interest.
SUMMARY OF THE INVENTION
The invention is based in part upon the discovery of isolated polypeptides including amino acid sequences selected from mature forms of the amino acid sequences selected from the group consisting of SEQ ID N0:2n, wherein n is an integer between 1 and 54. The invention also is based in part upon variants of a mature form of the amino acid sequence selected from the group consisting of SEQ ID N0:2n, wherein n is an integer between 1 and 54, wherein any amino acid in the mature form is changed to a different amino acid, provided that no more than 15% of the amino acid residues in the sequence of the mature form are so changed. In another embodiment, the invention includes the amino acid sequences selected from the group consisting of SEQ ID
NO:2n, wherein n is an integer between 1 and 54. In another embodiment, the invention also comprises variants of the amino acid sequence selected from the group consisting of SEQ
ID N0:2n, wherein n is an integer between 1 and 54 wherein any amino acid specified in the chosen sequence is changed to a different amino acid, provided that no more than 15%
of the amino acid residues in the sequence are so changed. The invention also involves fragments of any of the mature forms of the amino acid sequences selected from the group consisting of SEQ ID N0:2n, wherein n is an integer between 1 and 54, or any other amino acid sequence selected from this group. The invention also comprises fragments from these groups in which up to 15% of the residues are changed.
In another embodiment, the invention encompasses polypeptides that are naturally occurring allelic variants of the sequence selected from the group consisting of SEQ ID
N0:2n, wherein n is an integer between 1 and 54. These allelic variants include amino acid sequences that are the translations of nucleic acid sequences differing by a single nucleotide from nucleic acid sequences selected from the group consisting of SEQ ID
NOS: 2n-l, wherein n is an integer between 1 and 54. T'he variant polypeptide where any amino acid changed in the chosen sequence is changed to provide a conservative substitution.
In another embodiment, the invention comprises a pharmaceutical composition involving a polypeptide with an amino acid sequence selected from the group consisting of
3 SEQ ID N0:2n, wherein n is an integer between 1 and 54 and a pharmaceutically acceptable carrier. In another embodiment, the invention involves a kit, including, in one or more containers, this pharmaceutical composition.
In another embodiment, the invention includes the use of a therapeutic in the manufacture of a medicament for treating a syndrome associated with a human disease, the disease being selected from a pathology associated with a polypeptide with an amino acid sequence selected from the group consisting of SEQ ID N0:2n, wherein n is an integer between 1 and 54 wherein said therapeutic is the polypeptide selected from this group.
In another embodiment, the invention comprises a method for determining the presence or amount of a polypeptide with an amino acid sequence selected from the group consisting of SEQ ID NO:2n, wherein n is an integer between 1 and 54 in a sample, the method involving providing the sample; introducing the sample to an antibody that binds immunospecifically to the polypeptide; and determining the presence or amount of antibody bound to the polypeptide, thereby determining the presence or amount of polypeptide in the sample.
In another embodiment, the invention includes a method for determining the presence of or predisposition to a disease associated with altered levels of a polypeptide with an amino acid sequence selected from the group consisting of SEQ ID
N0:2n, wherein n is an integer between 1 and 54 in a first mammalian subject, the method involving measuring the level of expression of the polypeptide in a sample from the first mammalian subject; and comparing the amount of the polypeptide in this sample to the amount of the polypeptide present in a control sample from a second mammalian subject known not to have, or not to be predisposed to, the disease, wherein an alteration in the expression level of the polypeptide in the first subject as compared to the control sample indicates the presence of or predisposition to the disease.
In another embodiment, the invention involves a method of identifying an agent that binds to a polypeptide with an amino acid sequence selected from the group consisting of SEQ ID N0:2n, wherein n is an integer between 1 and 54, the method including introducing the polypeptide to the agent; and determining whether the agent binds to the polypeptide. The agent could be a cellular receptor or a downstream effector.
In another embodiment, the invention involves a method for identifying a potential therapeutic agent for use in treatment of a pathology, wherein the pathology is related to aberrant expression or aberrant physiological interactions of a polypeptide with an amino
4 acid sequence selected from the group consisting of SEQ ID N0:2n, wherein n is an integer between 1 and 54, the method including providing a cell expressing the polypeptide of the invention and having a property or function ascribable to the polypeptide; contacting the cell with a composition comprising a candidate substance; and determining whether the substance alters the property or function ascribable to the polypeptide; whereby, if an alteration observed in the presence of the substance is not observed when the cell is contacted with a composition devoid of the substance, the substance is identified as a potential therapeutic agent.
In another embodiment, the invention involves a method for screening for a modulator of activity or of latency or predisposition to a pathology associated with a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID
N0:2n, wherein n is an integer between 1 and 54, the method including administering a test compound to a test animal at increased risk for a pathology associated with the polypeptide of the invention, wherein the test animal recombinantly expresses the polypeptide of the invention; measuring the activity of the polypeptide in the test animal after administering the test compound; and comparing the activity of the protein in the test animal with the activity of the polypeptide in a control animal not administered the polypeptide, wherein a change in the activity of the polypeptide in the test animal relative to the control animal indicates the test compound is a modulator of latency of, or predisposition to, a pathology associated with the polypeptide of the invention. The recombinant test animal could express a test protein transgene or express the transgene under the control of a promoter at an increased level relative to a wild-type 'test animal The promoter may or may not b the native gene promoter of the transgene.
In another embodiment, the invention involves a method for modulating the activity of a polypeptide with an amino acid sequence selected from the group consisting of SEQ ID NO:2n, wherein n is an integer between 1 and 54, the method including introducing a cell sample expressing the polypeptide with a compound that binds to the polypeptide in an amount sufficient to modulate the activity of the polypeptide.
In another embodiment, the invention involves a method of treating or preventing a pathology associated with a polypeptide with an amino acid sequence selected from the group consisting of SEQ ID N0:2n, wherein n is an integer between l and 54, the method including administering the polypeptide to a subject in which such treatment or prevention is desired in an amount sufficient to treat or prevent the pathology in the subject. The subject could be human.

In another embodiment, the invention involves a method of treating a pathological state in a mammal, the method including administering to the mammal a polypeptide in an amount that is sufficient to alleviate the pathological state, wherein the polypeptide is a polypeptide having an amino acid sequence at least 95% identical to a polypeptide having the amino acid sequence selected from the group consisting of SEQ ID N0:2n, wherein n is an integer between 1 and 54 or a biologically active fragment thereof.
In another embodiment, the invention involves an isolated nucleic acid molecule comprising a nucleic acid sequence encoding a polypeptide having an amino acid sequence selected from the group consisting of a mature form of the amino acid sequence given SEQ ID N0:2n, wherein n is an integer between 1 and 54; a variant of a mature form of the amino acid sequence selected from the group consisting of SEQ ID
N0:2n, wherein n is an integer between 1 and 54 wherein any amino acid in the mature form of the chosen sequence is changed to a different amino acid, provided that no more than 15%
of the amino acid residues in the sequence of the mature form are so changed;
the amino acid sequence selected from the group consisting of SEQ ID N0:2n, wherein n is an integer between 1 and 54; a variant of the amino acid sequence selected from the group consisting of SEQ ID N0:2n, wherein n is an integer between 1 and 54, in which any amino acid specified in the chosen sequence is changed to a different amino acid, provided that no more than 15% of the amino acid residues in the sequence are so changed; a nucleic acid fragment encoding at least a portion of a polypeptide comprising the amino acid sequence selected from the group consisting of SEQ ID N0:2n, wherein n is an integer between 1 and 54 or any variant of the polypeptide wherein any amino acid of the chosen sequence is changed to a different amino acid, provided that no more than 10% of the amino acid residues in the sequence are so changed; and the complement of any of the nucleic acid molecules.
In another embodiment, the invention comprises an isolated nucleic acid molecule having a nucleic acid sequence encoding a polypeptide comprising an amino acid sequence selected from the group consisting of a mature form of the amino acid sequence given SEQ ID N0:2n, wherein n is an integer between 1 and 54, wherein the nucleic acid molecule comprises the nucleotide sequence of a naturally occurring allelic nucleic acid variant.
In another embodiment, the invention involves an isolated nucleic acid molecule including a nucleic acid sequence encoding a polypeptide having an amino acid sequence selected from the group consisting of a mature form of the amino acid sequence given SEQ ID N0:2n, wherein n is an integer between 1 and 54 that encodes a variant polypeptide, wherein the variant polypeptide has the polypeptide sequence of a naturally occurring polypeptide variant.
In another embodiment, the invention comprises an isolated nucleic acid molecule S having a nucleic acid sequence encoding a polypeptide comprising an amino acid sequence selected from the group consisting of a mature form of the amino acid sequence given SEQ ID N0:2n, wherein n is an integer between 1 and 54, wherein the nucleic acid molecule differs by a single nucleotide from a nucleic acid sequence selected from the group consisting of SEQ ID NOS: 2n-1, wherein n is an integer between l and 54.
In another embodiment, the invention includes an isolated nucleic acid molecule having a nucleic acid sequence encoding a polypeptide including an amino acid sequence selected from the group consisting of a mature form of the amino acid sequence given SEQ ID N0:2n, wherein n is an integer between 1 and 54, wherein the nucleic acid molecule comprises a nucleotide sequence selected from the group consisting of the nucleotide sequence selected from the group consisting of SEQ ID NO:2n-1, wherein n is an integer between 1 and 54; a nucleotide sequence wherein one or more nucleotides in the nucleotide sequence selected from the group consisting of SEQ ID NO:2n-1, wherein n is an integer between 1 and 54 is changed from that selected from the group consisting of the chosen sequence to a different nucleotide provided that no more than 15% of the nucleotides are so changed; a nucleic acid fragment of the sequence selected from the group consisting of SEQ ID NO:2n-1, wherein n is an integer between 1 and 54;
and a nucleic acid fragment wherein one or more nucleotides in the nucleotide sequence selected from the group consisting of SEQ ID N0:2n-1, wherein n is an integer between 1 and 54 is changed from that selected from the group consisting of the chosen sequence to a different nucleotide provided that no more than 15% of the nucleotides are so changed.
In another embodiment, the invention includes an isolated nucleic acid molecule having a nucleic acid sequence encoding a polypeptide including an amino acid sequence selected from the group consisting of a mature form of the amino acid sequence given SEQ ID N0:2n, wherein n is an integer between 1 and 54, wherein the nucleic acid molecule hybridizes under stringent conditions to the nucleotide sequence selected from the group consisting of SEQ ID N0:2n-1, wherein n is an integer between l and 54, or a complement of the nucleotide sequence.
In another embodiment, the invention includes an isolated nucleic acid molecule having a nucleic acid sequence encoding a polypeptide including an amino acid sequence selected from the group consisting of a mature form of the amino acid sequence given SEQ ID N0:2n, wherein n is an integer between 1 and 54, wherein the nucleic acid molecule has a nucleotide sequence in which any nucleotide specified in the coding sequence of the chosen nucleotide sequence is changed from that selected from the group consisting of the chosen sequence to a different nucleotide provided that no more than 15% of the nucleotides in the chosen coding sequence are so changed, an isolated second polynucleotide that is a complement of the first polynucleotide, or a fragment of any of them.
In another embodiment, the invention includes a vector involving the nucleic acid molecule having a nucleic acid sequence encoding a polypeptide including an amino acid sequence selected from the group consisting of a mature form of the amino acid sequence given SEQ ID N0:2n, wherein n is an integer between 1 and 54. This vector can have a promoter operably linked to the nucleic acid molecule. This vector can be located within a cell.
In another embodiment, the invention involves a method for determining the presence or amount of a nucleic acid molecule having a nucleic acid sequence encoding a polypeptide including an amino acid sequence selected from the group consisting of a mature form of the amino acid sequence given SEQ ID N0:2n, wherein n is an integer between 1 and 54 in a sample, the method including providing the sample;
introducing the sample to a probe that binds to the nucleic acid molecule; and determining the presence or amount of the probe bound to the nucleic acid molecule, thereby determining the presence or amount of the nucleic acid molecule in the sample. The presence or amount of the nucleic acid molecule is used as a marker for cell or tissue type. The cell type can be cancerous.
In another embodiment, the invention involves a method for determining the presence of or predisposition for a disease associated with altered levels of a nucleic acid molecule having a nucleic acid sequence encoding a polypeptide including an amino acid sequence selected from the group consisting of a mature form of the amino acid sequence given SEQ ID N0:2n, wherein n is an integer between 1 and 54 in a first mammalian subject, the method including measuring the amount of the nucleic acid in a sample from the first mammalian subject; and comparing the amount of the nucleic acid in the sample of step (a) to the amount of the nucleic acid present in a control sample from a second mammalian subject known not to have or not be predisposed to, the disease;
wherein an alteration in the level of the nucleic acid in the first subject as compared to the control sample indicates the presence of or predisposition to the disease.
Unless otherwise defined, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. Although methods and materials similar or equivalent to those described herein can be used in the practice or testing of the present invention, suitable methods and materials are described below. All publications, patent applications, patents, and other references mentioned herein are incorporated by reference in their entirety. In the case of conflict, the present specification, including definitions, will control. In addition, the materials, methods, and examples are illustrative only and not intended to be limiting.
Other features and advantages of the invention will be apparent from the following detailed description and claims.
DETAILED DESCRIPTION OF THE INVENTION
The present invention provides novel nucleotides and polypeptides encoded thereby. Included in the invention are the novel nucleic acid sequences, their encoded polypeptides, antibodies, and other related compounds. The sequences are collectively referred to herein as "NOVX nucleic acids" or "NOVX polynucleotides" and the corresponding encoded polypeptides are referred to as "NOVX polypeptides" or "NOVX
proteins." Unless indicated otherwise, "NOVX" is meant to refer to any of the novel sequences disclosed herein. Table 1 provides a summary of the NOVX nucleic acids and their encoded polypeptides.
TABLE 1. Sequences and Corresponding SEQ ID Numbers SEQ ID SEQ
NOVX Internal NO ID Homology AssignmentIdentification(nucleicNO
acid (amino acid Novla CG100488-O11 2 Elastase 2B like homo sa iens Novlb CG100488-063 4 Elastase 2B like homo sapiens Novlc CG100488-07S 6 Elastase 2B like homo sa iens Novld CG100488-087 8 Elastase 2B like homo sa iens Novle CG100488-099 10 Elastase 2B like homo sa iens Novlf 198353297 11 12 Elastase 2B like homo sa iens Novl 198353301 13 14 Elastase 2B like homo sa iens Novlh 198353319 15 16 Elastase 2B like homo sa iens Novli 198362547 17 18 Elastase 2B like homo sa iens Novl' 198362642 19 20 Elastase 2B like homo sa iens Nov2a CG100560-Ol 21 22 Leucine Rich Repeat like homo sa iens Nov2b. CG100560-02 23 24 Leucine Rich Repeat like homo Sapiens Nov3a CG101012-01 25 26 Gonadotrophin beta-subunit like homo sa iens Nov4a CG101584-Ol 27 2g odorant binding protein like homo sa iens NovSa CG101707-O1 29 30 Com lement Clq Nov6a CG101836-O1 31 32 Cathe sin F like homo sa iens Nov6b CG101836-02 33 34 Cathe sin F like homo sa iens Nov7a CG102221-O1 35 36 netrin G1 like homo sa iens NovBa CG102325-O1 37 38 Secreted reprolysin Nov9a CG102832-O1 39 40 CAC37763 like homo Sapiens Ig domain-containing Nov9b CG102832-02 41 42 transmembrane protein like homo sa iens Ig domain-containing Nov9c 197195425 43 44 transmembrane protein like homo sa iens Ig domain-containing Nov9d 197192431 45 46 transmembrane protein like homo sapiens Ig domain-containing Nov9e 197192437 47 48 transmembrane protein like homo sa iens Ig domain-containing Nov9f 197192443 49 50 transmembrane protein like homo sa iens Ig domain-containing Nov9g 197192448 51 52 transmembrane protein like homo sa iens NovlOa CG102942-O1 53 54 Ii ocalin 2 like homo sapiens Neutrophil Gelatinase-NovlOb CG102942-03 55 56 Associated lipocalin like homo sapiens Neutrophil Gelatinase-NovlOc 237376776 57 58 Associated lipocalin like homo Sapiens Novl CG104016-O1 59 60 DENN domain containing la rotein like homo Sapiens Novl 197208336 61 62 DENN domain containing 1b rotein like homo sa iens Novl 197306179 63 64 DENN domain containing lc rotein like homo sa iens Novl 219903686 65 66 DENN domain containing 1d rotein like homo sapiens Novl 219903690 67 6g DENN domain containing 1e rotein like homo sa iens Novl2a CG104903-O1 69 70 Kininogen Precursor like homo sapiens Novl2b CG104903-02 71 73 Kininogen Precursor like homo sa iens Novl2c CG104903-03 73 74 Kininogen Precursor like homo Sapiens Novl2d CG104903-OS 75 76 Kininogen Precursor like homo sa iens Novl2e CG104903-06 77 7g Kininogen Precursor like homo sa iens Novl2f CG104903-07 79 g0 Kininogen Precursor like homo sa iens Novl2g CG104903-O8 81 g2 Kininogen Precursor ' like homo sapiens Novl2h CG104903-09 83 g4 Kininogen Precursor like homo Sapiens Novl3a CG105982-O1 85 g6 Serine Protease-CUB
Domain Protein like homo sa iens Novl4a CG107614-02 87 88 Hemopexin-like NovlSa CG109445-O1 89 90 neuronal leucine-rich repeat protein like homo Sapiens Novl6a CG109496-O1 91 g2 neuronal leucine-rich repeat protein like homo sa iens Immunoglobulin domains Novl7a CG109532-O1 93 94 containing protein like homo sapiens Immunoglobulin domains Novl7b 207775340 95 96 containing protein like homo sa iens Immunoglobulin domains Novl7c 207775361 97 98 containing protein like homo sapiens Immunoglobulin domains Novl7d 207775365 99 100 containing protein like homo sa iens small inducible cytokine Novl CG50213-O1 101 102 subfamily B member 8a 14 (BRAK) small inducible cytokine Novl8b CG50213-02 103 104 subfamily B member (BRAK) small inducible cytokine Novl8c CG50213-03 105 106 subfamily B member (BRAK) BETA-NEOENDORPHIN-Novl9a CG88912-02 107 108 DYNORPHIN PRECURSOR

like homo sa iens Table 1 indicates homology of NOVX nucleic acids to known protein families.
Thus, the nucleic acids and polypeptides, antibodies and related compounds according to the invention corresponding to a NOVX as identified in column 1 of Table 1 will be useful in therapeutic and diagnostic applications implicated in, for example, pathologies and disorders associated with the known protein families identified in column 5 of Table 1.
NOVX nucleic acids and their encoded polypeptides are useful in a variety of applications and contexts. The various NOVX nucleic acids and polypeptides according to the invention are useful as novel members of the protein families according to the presence of domains and sequence relatedness to previously described proteins.
Additionally, NOVX nucleic acids and polypeptides can also be used to identify proteins that are members of the family to which the NOVX polypeptides belong.
Consistent with other known members of the family of proteins, identified in column 5 of Table 1, the NOVX polypeptides of the present invention show homology to, and contain domains that are characteristic of, other members of such protein families.
Details of the sequence relatedness and domain analysis for each NOVX are presented in Example A.
The NOVX nucleic acids and polypeptides can also be used to screen for molecules, which inhibit or enhance NOVX activity or function. Specifically, the nucleic acids and polypeptides according to the invention may be used as targets for the identification of small molecules that modulate or inhibit diseases associated with the protein families listed in Table 1.
The NOVX nucleic acids and polypeptides are also useful for detecting specific cell types. Details of the expression analysis for each NOVX are presented in Example C.
Accordingly, the NOVX nucleic acids, polypeptides, antibodies and related compounds according to the invention will have diagnostic and therapeutic applications in the detection of a variety of diseases with differential expression in normal vs.
diseased tissues, e.g. a variety of cancers.
Additional utilities for NOVX nucleic acids and polypeptides according to the invention are disclosed herein.
NOVX clones NOVX nucleic acids and their encoded polypeptides are useful in a variety of applications and contexts. The various NOVX nucleic acids and polypeptides according to the invention are useful as novel members of the protein families according to the presence of domains and sequence relatedness to previously described proteins.
Additionally, NOVX nucleic acids and polypeptides can also be used to identify proteins that are members of the family to which the NOVX polypeptides belong.
The NOVX genes and their corresponding encoded proteins are useful for preventing, treating or ameliorating medical conditions, e.g., by protein or gene therapy.
Pathological conditions can be diagnosed by determining the amount of the new protein in a sample or by determining the presence of mutations in the new genes.
Specific uses are described for each of the NOVX genes, based on the tissues in which they are most highly expressed. Uses include developing products for the diagnosis or treatment of a variety of diseases and disorders.
The NOVX nucleic acids and proteins of the invention are useful in potential diagnostic and therapeutic applications and as a research tool. These include serving as a specific or selective nucleic acid or protein diagnostic and/or prognostic marker, wherein .
the presence or amount of the nucleic acid or the protein are to be assessed, as well as potential therapeutic applications such as the following: (i) a protein therapeutic, (ii) a small molecule drug target, (iii) an antibody target (therapeutic, diagnostic, drug targeting/cytotoxic antibody), (iv) a nucleic acid useful in gene therapy (gene delivery/gene ablation), and (v) a composition promoting tissue regeneration in vitro and in vivo (vi) biological defense weapon.
In one specific embodiment, the invention includes an isolated polypeptide comprising an amino acid sequence selected from the group consisting of-. (a) a mature form of the amino acid sequence selected from the group consisting of SEQ ID
N0:2n, wherein n is an integer between 1 and 54; (b) a variant of a mature form of the amino acid sequence selected from the group consisting of SEQ ID N0:2n, wherein n is an integer between I and 54, wherein any amino acid in the mature form is changed to a different amino acid, provided that no more than 15% of the amino acid residues in the sequence of the mature form are so changed; (c) an amino acid sequence selected from the group consisting of SEQ ID NO:2n, wherein n is an integer between 1 and 54; (d) a variant of the amino acid sequence selected from the group consisting of SEQ ID N0:2n, wherein n is an integer between 1 and 54 wherein any amino acid specified in the chosen sequence is changed to a different amino acid, provided that no more than 15% of the amino acid residues in the sequence are so changed; and (e) a fragment of any of (a) through (d).

In another specific embodiment, the invention includes an isolated nucleic acid molecule comprising a nucleic acid sequence encoding a polypeptide comprising an amino acid sequence selected from the group consisting of (a) a mature form of the amino acid sequence given SEQ ID N0:2n, wherein n is an integer between 1 and 54; (b) a variant of a mature form of the amino acid sequence selected from the group consisting of SEQ ID
N0:2n, wherein n is an integer between 1 and 54 wherein any amino acid in the mature form of the chosen sequence is changed to a different amino acid, provided that no more than 15% of the amino acid residues in the sequence of the mature form are so changed;
(c) the amino acid sequence selected from the group consisting of SEQ ID
N0:2n, wherein n is an integer between 1 and 54; (d) a variant of the amino acid sequence selected from the group consisting of SEQ ID N0:2n, wherein n is an integer between 1 and 54, in which any amino acid specified in the chosen sequence is changed to a different amino acid, provided that no more than 15% of the amino acid residues in the sequence are so changed; (e) a nucleic acid fragment encoding at least a portion of a polypeptide comprising the amino acid sequence selected from the group consisting of SEQ
ID N0:2n, wherein n is an integer between 1 and 54 or any variant of said polypeptide wherein any amino acid of the chosen sequence is changed to a different amino acid, provided that no more than 10% of the amino acid residues in the sequence are so changed; and (f) the complement of any of said nucleic acid molecules.
In yet another specific embodiment, the invention includes an isolated nucleic acid molecule, wherein said nucleic acid molecule comprises a nucleotide sequence selected from the group consisting of (a) the nucleotide sequence selected from the group consisting of SEQ ID N0:2n-l, wherein n is an integer between 1 and 54; (b) a nucleotide sequence wherein one or more nucleotides in the nucleotide sequence selected from the group consisting of SEQ ID N0:2n-1, wherein n is an integer between 1 and 54 is changed from that selected from the group consisting of the chosen sequence to a different nucleotide provided that no more than 15% of the nucleotides are so changed;
(c) a nucleic acid fragment of the sequence selected from the group consisting of SEQ ID
N0:2n-1, wherein n is an integer between 1 and 54; and (d) a nucleic acid fragment wherein one or more nucleotides in the nucleotide sequence selected from the group consisting of SEQ ID NO:2n-1, wherein n is an integer between 1 and 54 is changed from that selected from the group consisting of the chosen sequence to a different nucleotide provided that no more than 15% of the nucleotides are so changed.

NOVX Nucleic Acids and Polypeptides One aspect of the invention pertains to isolated nucleic acid molecules that encode NOVX polypeptides or biologically active portions thereof. Also included in the invention are nucleic acid fragments sufficient for use as hybridization probes to identify NOVX-encoding nucleic acids (e.g., NOVX mRNAs) and fragments for use as PCR
primers for the amplification and/or mutation of NOVX nucleic acid molecules.
As used herein, the term "nucleic acid molecule" is intended to include DNA molecules (e.g., cDNA or genomic DNA), RNA molecules (e.g., mRNA), analogs of the DNA or RNA
generated using nucleotide analogs, and derivatives, fragments and homologs thereof. The nucleic acid molecule may be single-stranded or double-stranded, but preferably is comprised double-stranded DNA.
A NOVX nucleic acid can encode a mature NOVX polypeptide. As used herein, a "mature" form of a polypeptide or protein disclosed in the present invention is the product of a naturally occurring polypeptide, precursor form, or proprotein. The naturally occurring polypeptide, precursor or proprotein includes, by way of nonlimiting example, the full-length gene product encoded by the corresponding gene. Alternatively, it may be defined as the polypeptide, precursor or proprotein encoded by an ORF
described herein.
The product "mature" form arises, by way of nonlimiting example, as a result of one or more naturally occurring processing steps that may take place within the cell (host cell) in which the gene product arises. Examples of such processing steps leading to a "mature"
form of a polypeptide or protein include the cleavage of the N-terminal methionine residue encoded by the initiation codon of an ORF or the proteolytic cleavage of a signal peptide or leader sequence. Thus a mature form arising from a precursor polypeptide or protein that has residues 1 to N, where residue 1 is the N-terminal methionine, would have residues 2 through N remaining after removal of the N-terminal methionine.
Alternatively, a mature form arising from a precursor polypeptide or protein having residues 1 to N, in which an N-terminal signal sequence from residue 1 to residue M is cleaved, would have the residues from residue M+1 to residue N remaining.
Further as used herein, a "mature" form of a polypeptide or protein may arise from a post-translational modification other than a proteolytic cleavage event. Such additional processes include, by way of non-limiting example, glycosylation, myristoylation or phosphorylation. In general, a mature polypeptide or protein may result from the operation of only one of these processes, or a combination of any of them.
IS

The term "probe", as utilized herein, refers to nucleic acid sequences of variable length, preferably between at least about 10 nucleotides (nt), and 100 nt, or as many as approximately, e.g., 6,000 nt, depending upon the specific use. Probes are used in the detection of identical, similar,~or complementary nucleic acid sequences.
Longer length probes are generally obtained from a natural or recombinant source, are highly specific, and much slower to hybridize than shorter-length oligomer probes. Probes may be single-or double-stranded and designed to have specificity in PCR, membrane-based hybridization technologies, or ELISA-like technologies.
The term "isolated" nucleic acid molecule, as used herein, is a nucleic acid which is separated from other nucleic acid molecules which are present in the natural source of the nucleic acid. Preferably, an "isolated" nucleic acid is free of sequences which naturally flank the nucleic acid (i.e., sequences located at the 5'- and 3'-termini of the nucleic acid) in the genomic DNA of the organism from which the nucleic acid is derived.
For example, in various embodiments, the isolated NOVX nucleic acid molecules can contain less than about 5 kb, 4 kb, 3 kb, 2 kb, 1 kb, 0.5 kb, 0.1 kb, or less of nucleotide sequences which naturally flank the nucleic acid molecule in genomic DNA of the cellltissue from which the nucleic acid is derived (e.g., brain, heart, liver, spleen, etc.).
Moreover, an "isolated" nucleic acid molecule, such as a cDNA molecule, can be substantially free of other cellular material, culture medium, or of chemical precursors or other chemicals.
A nucleic acid molecule of the invention, e.g., a nucleic acid molecule having the nucleotide sequence SEQ ID NOS: 2n-1, wherein n is an integer between l and 54, or a complement of this nucleotide sequence, can be isolated using standard molecular biology techniques and the sequence information provided herein. Using all or a portion of the nucleic acid sequence of SEQ ID NOS:2n-l, wherein n is an integer between 1 and 54, as a hybridization probe, NOVX molecules can be isolated using standard hybridization and cloning techniques (e.g., as described in Sambrook, et al., (eds.), MOLECULAR
CLOI~TING: A
LABORATORY MAt~tuAL 2nd Ed., Cold Spring Harbor Laboratory Press, Cold Spring Harbor, NY, 1959; and Ausubel, et al., (eds.), CURRENT PROTOCOLS IN MOLECULAR
BIOLOGY, John Wiley & Sons, New York, NY, 1993).
A nucleic acid of the invention can be amplified using cDNA, mRNA or, alternatively, genomic DNA as a template with appropriate oligonucleotide primers according to standard PCR amplification techniques. The nucleic acid so amplified can be cloned into an appropriate vector and characterized by DNA sequence analysis.

Furthermore, oligonucleotides corresponding to NOVX nucleotide sequences can be prepared by standard synthetic techniques, e.g., using an automated DNA
synthesizer.
As used herein, the term "oligonucleotide" refers to a series of linked nucleotide residues. A short oligonucleotide sequence may be based on, or designed from, a genomic or cDNA sequence and is used to amplify, confirm, or reveal the presence of an identical, similar or complementary DNA or RNA in a particular cell or tissue.
Oligonucleotides comprise a nucleic acid sequence having about 10 nt, 50 nt, or 100 nt in length, preferably about 15 nt to 30 nt in length. In one embodiment of the invention, an oligonucleotide comprising a nucleic acid molecule less than 100 nt in length would further comprise at least 6 contiguous nucleotides of SEQ ID NOS:2n-1, wherein n is an integer between.l and 54, or a complement thereof. Oligonucleotides may be chemically synthesized and may also be used as probes.
In another embodiment, an isolated nucleic acid molecule of the invention comprises a nucleic acid molecule that is a complement of the nucleotide sequence shown in SEQ ID NOS:2n-l, wherein n is an integer between 1 and 54, or a portion of this nucleotide sequence (e.g., a fragment that can be used as a probe or primer or a fragment encoding a biologically-active portion of A NOVX polypeptide). A nucleic acid molecule that is complementary to the nucleotide sequence shown SEQ ID NOS:2n-1, wherein n is an integer between 1 and 54,is one that is sufficiently complementary to the nucleotide sequence shown SEQ ID NOS:2n-l, wherein n is an integer between 1 and 54,that it can hydrogen bond with few or no mismatches to the nucleotide sequence shown SEQ
ID
NOS:2n-1, wherein n is an integer between 1 and 54, thereby forming a stable duplex.
As used herein, the term "complementary" refers to Watson-Crick or Hoogsteen base pairing between nucleotides units of a nucleic acid molecule, and the term "binding"
means the physical or chemical interaction between two polypeptides or compounds or associated polypeptides or compounds or combinations thereof. Binding includes ionic, non-ionic, van der Waals, hydrophobic interactions, and the like. A physical interaction can be either direct or indirect. Indirect interactions may be through or due to the effects of another polypeptide or compound. Direct binding refers to interactions that do not take place through, or due to, the effect of another polypeptide or compound, but instead are without other substantial chemical intermediates.
"Fragments" provided herein are defined as sequences of at least 6 (contiguous) nucleic acids or at least 4 (contiguous) amino acids, a length sufficient to allow for specific hybridization in the case of nucleic acids or for specific recognition of an epitope in the case of amino acids, and are at most some portion less than a full length sequence.
Fragments may be derived from any contiguous portion of a nucleic acid or amino acid sequence of choice.
A full-length NOVX clone is identified as containing an ATG translation start codon and an in-frame stop codon. Any disclosed NOVX nucleotide sequence lacking an ATG start codon therefore encodes a truncated C-terminal fragment of the respective NOVX polypeptide, and requires that the corresponding full-length cDNA extend in the 5' direction of the disclosed sequence. Any disclosed NOVX nucleotide sequence lacking an in-frame stop codon similarly encodes a truncated N-terminal fragment of the respective NOVX polypeptide, and requires that the corresponding full-length cDNA extend in the 3' direction of the disclosed sequence.
"Derivatives" are nucleic acid sequences or amino acid sequences formed from the native compounds either directly, by modification, or by partial substitution.
"Analogs"
are nucleic acid sequences or amino acid sequences that have a structure similar to, but not identical to, the native compound, e.g. they differ from it in respect to certain components or side chains. Analogs may be synthetic or derived from a different evolutionary origin and may have a similar or opposite metabolic activity compared to wild type.
Homologs are nucleic acid sequences or amino acid sequences of a particular gene that are derived from different species.
Derivatives and analogs may be full length or other than full length.
Derivatives or analogs of the nucleic acids or proteins of the invention include, but are not limited to, molecules comprising regions that are substantially homologous to the nucleic acids or proteins of the invention, in various embodiments, by at least about 70%, 80%, or 95%
identity (with a preferred identity of 80-95%) over a nucleic acid or amino acid sequence of identical size or when compared to an aligned sequence in which the alignment is done by a computer homology program known in the art, or whose encoding nucleic acid is capable of hybridizing to the complement of a sequence encoding the proteins of the invention under stringent, moderately stringent, or low stringent conditions.
See e.g.
Ausubel, et al., CURRENT PROTOCOLS IN MOLECULAR BIOLOGY, John Wiley & Sons, New York, NY, 1993, and below.
A "homologous nucleic acid sequence" or "homologous amino acid sequence," or variations thereof, refer to sequences characterized by a homology at the nucleotide level or amino acid level as discussed above. Homologous nucleotide sequences include those sequences coding for isoforms of NOVX polypeptides. Isoforms can be expressed in different tissues of the same organism as a result of, for example, alternative splicing of RNA. Alternatively, isoforms can be encoded by different genes. In the invention, homologous nucleotide sequences include nucleotide sequences encoding for A
NOVX
polypeptide of species other than humans, including, but not limited to vertebrates, and thus can include, e.g., frog, mouse, rat, rabbit, dog, cat, cow, horse, and other organisms.
Homologous nucleotide sequences also include, but axe not limited to, naturally occurring allelic variations and mutations of the nucleotide sequences set forth herein.
A
homologous nucleotide sequence does not, however, include the exact nucleotide sequence encoding a human NOVX protein. Homologous nucleic acid sequences include those nucleic acid sequences that encode conservative amino acid substitutions (see below) in SEQ ID NOS:2n-1, wherein n is an integer between 1 and 54, as well as a polypeptide possessing NOVX biological activity. Various biological activities of the NOVX proteins are described below.
A NOVX polypeptide is encoded by the open reading frame ("ORF") of a NOVX
1S nucleic acid. An ORF corresponds to a nucleotide sequence that could potentially be translated into a polypeptide. A stretch of nucleic acids comprising an ORF is uninterrupted by a stop colon. An ORF that represents the coding sequence for a full protein begins with an ATG "start" colon and terminates with one of the three "stop"
colons, namely, TAA, TAG, or TGA. For the purposes of this invention, an ORF
may be any part of a coding sequence, with or without a start colon, a stop colon, or both. For an ORF to be considered as a good candidate fox coding for a bona frde cellular protein, a minimum size requirement is often set, e.g., a stretch of DNA that would encode a protein of 50 amino acids or more.
The nucleotide sequences determined from the cloning of the human NOVX genes allows for the generation of probes and primers designed for use in identifying and/or cloning NOVX homologues in other cell types, e.g. from other tissues, as well as NOVX
homologues from other vertebrates. The probe/primer typically comprises a substantially purified oligonucleotide. The oligonucleotide typically comprises a region of nucleotide sequence that hybridizes under stringent conditions to at least about 12, 25, 50, 100, 150, 200, 250, 300, 350 or 400 consecutive sense strand nucleotide sequence of SEQ
ID
NOS:2n-1, wherein n is an integer between l and 54; or an anti-sense strand nucleotide sequence of SEQ ID NOS:2n-1, wherein n is an integer between 1 and 54; or of a naturally occurring mutant of SEQ ID NOS:2n-1, wherein n is an integer between 1 and 54.

Probes based on the human NOVX nucleotide sequences can be used to detect transcripts or genomic sequences encoding the same or homologous proteins. In various embodiments, the probe has a detectable label attached, e.g. the label can be a radioisotope, a fluorescent compound, an enzyme, or an enzyme co-factor. Such probes can be used as a part of a diagnostic test kit for identifying cells or tissues which mis-express A NOVX protein, such as by measuring a level of A NOVX-encoding nucleic acid in a sample of cells from a subject e.g., detecting NOVX mRNA levels or determining whether a genomic NOVX gene has been mutated or deleted.
"A polypeptide having a biologically-active portion of A NOVX polypeptide"
refers to polypeptides exhibiting activity similar, but not necessarily identical, an activity of a polypeptide of the invention, including mature forms, as measured in a particular biological assay, with or without dose dependency. A nucleic acid fragment encoding a "biologically-active portion of NOVX" can be prepared by isolating a portion SEQ ID
NOS:2n-l, wherein n is an integer between 1 and 54, that encodes a polypeptide having A
NOVX biological activity (the biological activities of the NOVX proteins are described below), expressing the encoded portion of NOVX protein (e.g., by recombinant expression in vitr~) and assessing the activity of the encoded portion of NOVX.
NOVX Nucleic Acid and PolypepHde Variants The invention further encompasses nucleic acid molecules that differ from the nucleotide sequences shown in SEQ ID NOS:2n-1, wherein n is an integer between 1 and 54, due to degeneracy of the genetic code and thus encode the same NOVX
proteins as that encoded by the nucleotide sequences shown in SEQ ID NOS:2n-1, wherein n is an integer between 1 and 54. In another embodiment, an isolated nucleic acid molecule of the invention has a nucleotide sequence encoding a protein having an amino acid sequence shown in SEQ ID NOS:2n, wherein n is an integer between 1 and 54.
In addition to the human NOVX nucleotide sequences shown in SEQ ID
NOS:2n-1, wherein n is an integer between l and 54, it will be appreciated by those skilled in the art that DNA sequence polymorphisms that lead to changes in the amino acid sequences of the NOVX polypeptides may exist within a population (e.g., the human population). Such genetic polymorphism in the NOVX genes may exist among individuals within a population due to natural allelic variation. As used herein, the terms "gene" and "recombinant gene" refer to nucleic acid molecules comprising an open reading frame (ORF) encoding A NOVX protein, preferably a vertebrate NOVX
protein.

Such natural allelic variations can typically result in I-5% variance in the nucleotide sequence of the NOVX genes. Any and all such nucleotide variations and resulting amino acid polymorphisms in the NOVX polypeptides, which are the result of natural allelic variation and that do not alter the functional activity of the NOVX
polypeptides, are intended to be within the scope of the invention.
Moreover, nucleic acid molecules encoding NOVX proteins from other species, and thus that have a nucleotide sequence that differs from the human SEQ ID
NOS:2n-I, wherein n is an integer between 1 and 54, are intended to be within the scope of the invention. Nucleic acid molecules corresponding to natural allelic variants and homologues of the NOVX cDNAs of the invention can be isolated based on their homology to the human NOVX nucleic acids disclosed herein using the human cDNAs, or a portion thereof, as a hybridization probe according to standard hybridization techniques under stringent hybridization conditions.
Accordingly, in another embodiment, an isolated nucleic acid molecule of the invention is at least 6 nucleotides in length and hybridizes under stringent conditions to the nucleic acid molecule comprising the nucleotide sequence of SEQ ID NOS:2n-l, wherein n is an integer between 1 and 54. In another embodiment, the nucleic acid is at least 10, 25, 50, 100, 250, 500, 750, 1000, 1500, 2000 or more nucleotides in length. In yet another embodiment, an isolated nucleic acid molecule of the invention hybridizes to the coding region. As used herein, the term "hybridizes under stringent conditions" is intended to describe conditions for hybridization and washing under which nucleotide sequences at least about 65% homologous to each other typically remain hybridized to each other.
Homologs (i.e., nucleic acids encoding NOVX proteins derived from species other than human) or other related sequences (e.g., paralogs) can be obtained by low, moderate or high stringency hybridization with all or a portion of the particular human sequence as a probe using methods well known in the art for nucleic acid hybridization and cloning.
As used herein, the phrase "stringent hybridization conditions" refers to conditions under which a probe, primer or oligonucleotide will hybridize to its target sequence, but to no other sequences. Stringent conditions are sequence-dependent and will be different in different circumstances. Longer sequences hybridize specifically at higher temperatures than shorter sequences. Generally, stringent conditions are selected to be about 5 °C lower than the thermal melting point (Tm) for the specific sequence at a defined ionic strength and pH. The Tm is the temperature (under defined ionic strength, pH and nucleic acid concentration) at which 50% of the probes complementary to the target sequence hybridize to the target sequence at equilibrium. Since the target sequences are generally present at excess at Tm, 50% of the probes are occupied at equilibrium.
Typically, stringent conditions will be those in which the salt concentration is less than about 1.0 M
sodium ion, typically about 0.01 to 1.0 M sodium ion (or other salts) at pH 7.0 to 8.3 and the temperature is at least about 30 °C for short probes, primers or oligonucleotides (e.g., 10 nt to 50 nt) and at least about 60 °C for longer probes, primers and oligonucleotides. Stringent conditions may also be achieved with the addition of destabilizing agents, such as formamide.
Stringent conditions are known to those skilled in the art and can be found in Ausubel, et al., (eds.), CURRENT PROTOCOLS IN MOLECULAR BIOLOGY, John Wiley &
Sons, N.Y. (1989), 6.3.1-6.3.6. Preferably, the conditions are such that sequences at least about 65%, 70%, 75%, 85%, 90%, 95%, 98%, or 99% homologous to each other typically remain hybridized to each other. A non-limiting example of stringent hybridization conditions are hybridization in a high salt buffer comprising 6X SSC, 50 mM
Tris-HCl (pH 7.5), 1 mM EDTA, 0.02% PVP, 0.02% Ficoll, 0.02% BSA, and 500 mg/ml denatured salmon sperm DNA at 65 °C, followed by one or more washes in 0.2X SSC, 0.01% BSA
at 50 °C. An isolated nucleic acid molecule of the invention that hybridizes under stringent conditions to the sequences SEQ ID NOS:2n-l, wherein n is an integer between 1 and 54, corresponds to a naturally-occurring nucleic acid molecule. As used herein, a "naturally-occurring" nucleic acid molecule refers to an RNA or DNA molecule having a nucleotide sequence that occurs in nature (e.g., encodes a natural protein).
In a second embodiment, a nucleic acid sequence that is hybridizable to the nucleic acid molecule comprising the nucleotide sequence of SEQ ID NOS:2n-1, wherein n is an integer between 1 and 54, or fragments, analogs or derivatives thereof, under conditions of moderate stringency is provided. A non-limiting example of moderate stringency hybridization conditions are hybridization in 6X SSC, SX Denhardt's solution, 0.5% SDS
and 100 mg/ml denatured salmon sperm DNA at 55 °C, followed by one or more washes in 1X SSC, 0.1% SDS at 37 °C. Other conditions of moderate stringency that may be used are well-known within the art. See, e.g., Ausubel, et al. (eds.), 1993, CURRENT PROTOCOLS
IN MOLECULAR BIOLOGY, John Wiley & Sons, NY, and Kriegler, 1990; GENE TRANSFER
AND ExPRESSION, A LABORATORY MANUAL, Stockton Press, NY.
In a third embodiment, a nucleic acid that is hybridizable to the nucleic acid .
molecule comprising the nucleotide sequences SEQ ID NOS:2n-l, wherein n is an integer between 1 and S4, or fragments, analogs or derivatives thereof, under conditions of low stringency, is provided. A non-limiting example of low stringency hybridization conditions are hybridization in 35% formamide, SX SSC, 50 mM Tris-HCl (pH
7.5), 5 mM EDTA, 0.02% PVP, 0.02% Ficoll, 0.2% BSA, 100 mglml denatured salmon sperm DNA, 10% (wt/vol) dextran sulfate at 40 °C, followed by one or more washes in 2X SSC, 25 mM Tris-HCl (pH 7.4), 5 mM EDTA, and 0.1% SDS at 50 °C. Other conditions of low stringency that may be used are well known in the art (e.g., as employed for cross-species hybridizations). See, e.g., Ausubel, et al. (eds.), 1993, CURRENT PROTOCOLS IN
MOLECULAR BIOLOGY, John Wiley ~z Sons, NY, and Kriegler, 1990, GENE TRANSFER
AND
EXPRESSION, A LABORATORY MANUAL, Stockton Press, NY; Shilo and Weinberg, 1981.
Proc Natl Acad Sci USA 78: 6789-6792.
Conservative Mutations In addition to naturally-occurring allelic variants of NOVX sequences that may exist in the population, the skilled artisan will further appreciate that changes can be introduced by mutation into the nucleotide sequences SEQ ID NOS:2n-l, wherein n is an integer between 1 and 54, thereby leading to changes in the amino acid sequences of the encoded NOVX proteins, without altering the functional ability of the NOVX
proteins.
For example, nucleotide substitutions leading to amino acid substitutions at "non-essential" amino acid residues can be made in the sequence SEQ ID NOS:2n, wherein n is an integer between 1 and 54. A "non-essential" amino acid residue is a residue that can be altered from the wild-type sequences of the NOVX proteins without altering their biological activity, whereas an "essential" amino acid residue is required for such biological activity. For example, amino acid residues that are conserved among the NOVX proteins of the invention are predicted to be particularly non-amenable to alteration. Amino acids for which conservative substitutions can be made are well known within the art.
Another aspect of the invention pertains to nucleic acid molecules encoding NOVX proteins that contain changes in amino acid residues that are not essential for activity. Such NOVX proteins differ in amino acid sequence from SEQ ID NOS:2n-1, wherein n is an integer between 1 and 54, yet retain biological activity. In one embodiment, the isolated nucleic acid molecule comprises a nucleotide sequence encoding a protein, wherein the protein comprises an amino acid sequence at least about homologous to the amino acid sequences SEQ ID NOS:2n, wherein n is an integer between 1 and 54. Preferably, the protein encoded by the nucleic acid molecule is at least about 60% homologous to SEQ ID NOS:2n, wherein n is an integer between 1 and 54;
more preferably at least about 70% homologous SEQ ID NOS:2n, wherein n is an integer between 1 and 54; still more preferably at least about 80% homologous to SEQ
ID
NOS:2n, wherein n is an integer between 1 and 54; even more preferably at least about 90% homologous to SEQ ID NOS:2n, wherein n is an integer between 1 and 54; and most preferably at least about 95% homologous to SEQ ID NOS:2n, wherein n is an integer between 1 and 54.
An isolated nucleic acid molecule encoding A NOVX protein homologous to the protein of SEQ ID NOS:2n, wherein n is an integer between 1 and 54, can be created by introducing one or more nucleotide substitutions, additions or deletions into the nucleotide sequence of SEQ ID NOS:2n-l, wherein n is an integer between 1 and 54, such that one or more amino acid substitutions, additions or deletions are introduced into the encoded protein.
Mutations can be introduced into SEQ ID NOS:2n-l, wherein n is an integer between 1 and 54, by standard techniques, such as site-directed mutagenesis and PCR-mediated mutagenesis. Preferably, conservative amino acid substitutions are made at one or more predicted, non-essential amino acid residues. A "conservative amino acid substitution" is one in which the amino acid residue is replaced with an amino acid residue w having a similar side chain. Families of amino acid residues having similar side chains have been defined within the art. These families include amino acids with basic side chains (e.g., lysine, arginine, histidine), acidic side chains (e.g., aspartic acid, glutamic acid), uncharged polar side chains (e.g., glycine, asparagine, glutamine, serine, threonine, tyrosine, cysteine), nonpolar side chains (e.g:, alanine, valine, leucine, isoleucine, proline, phenylalanine, methionine, tryptophan), beta-branched side chains (e.g., threonine, valine, isoleucine) and aromatic side chains (e.g., tyrosine, phenylalanine, tryptophan, histidine).
Thus, a predicted non-essential amino acid residue in the NOVX protein is replaced with another amino acid residue from the same side chain family. Alternatively, in another embodiment, mutations can be introduced randomly along all or part of A NOVX
coding sequence, such as by saturation mutagenesis, and the resultant mutants can be screened for NOVX biological activity to identify mutants that retain activity. Following mutagenesis SEQ ID NOS:2n-1, wherein n is an integer between 1 and 54, the encoded protein can be expressed by any recombinant technology known in the art and the activity of the protein can be determined.

The relatedness of amino acid families may also be determined based on side chain interactions. Substituted amino acids may be fully conserved "strong" residues or fully conserved "weak" residues. The "strong" group of conserved amino acid residues may be any one of the following groups: STA, NEQK, NHQK, NDEQ, QHRK, MILV, MILF, HY, FYW, wherein the single letter amino acid codes are grouped by those amino acids that may be substituted for each other. Likewise, the "weak" group of conserved residues may be any one of the following: CSA, ATV, SAG, STNK, STPA, SGND, SNDEQK, NDEQHK, NEQHRK, HFY, wherein the letters within each group represent the single letter amino acid code.
In one embodiment, a mutant NOVX protein can be assayed for (r~ the ability to form protein:protein interactions with other NOVX proteins, other cell-surface proteins, or biologically-active portions thereof, (irk complex formation between a mutant NOVX
protein and A NOVX ligand; or (iir~ the ability of a mutant NOVX protein to bind to an intracellular target protein or biologically-active portion thereof; (e.g.
avidin proteins).
In yet another embodiment, a mutant NOVX protein can be assayed for the ability to regulate a specific biological function (e.g., regulation of insulin release).
Antisense Nucleic Acids Another aspect of the invention pertains to isolated antisense nucleic acid molecules that are hybridizable to or complementary to the nucleic acid molecule comprising the nucleotide sequence of SEQ ID NOS:2n-1, wherein n is an integer between 1 and 54, or fragments, analogs or derivatives thereof. An "antisense" nucleic acid comprises a nucleotide sequence that is complementary to a "sense" nucleic acid encoding a protein (e.g., complementary to the coding strand of a double-stranded cDNA
molecule or complementary to an mRNA sequence). In specific aspects, antisense nucleic acid molecules are provided that comprise a sequence complementary to at least about 10, 25, 50, 100, 250 or 500 nucleotides or an entire NOVX coding strand, or to only a portion thereof. Nucleic acid molecules encoding fragments, homologs, derivatives and analogs of A NOVX protein of SEQ ID NOS:2n, wherein n is an integer between 1 and 54, or antisense nucleic acids complementary to A NOVX nucleic acid sequence of SEQ
ID
NOS:2n-l, wherein n is an integer between 1 and 54, are additionally provided.
In one embodiment, an antisense nucleic acid molecule is antisense to a "coding region" of the coding strand of a nucleotide sequence encoding A NOVX protein.
The term "coding region" refers to the region of the nucleotide sequence comprising codons, which are translated into amino acid residues. In another embodiment, the antisense nucleic acid molecule is antisense to a "noncoding region" of the coding strand of a nucleotide sequence encoding the NOVX protein. The term "noncoding region"
refers to
5' and 3' sequences, which flank the coding region that are not translated into amino acids (i.e., also referred to as 5' and 3' untranslated regions).
Given the coding strand sequences encoding the NOVX protein disclosed herein, antisense nucleic acids of the invention can be designed according to the rules of Watson and Crick or Iioogsteen base pairing. The antisense nucleic acid molecule can be complementary to the entire coding region of NOVX mRNA, but more preferably is an oligonucleotide that is antisense to only a portion of the coding or noncoding region of NOVX mRNA. For example, the antisense oligonucleotide can be complementary to the region surrounding the translation start site of NOVX mRNA. An antisense oligonucleotide can be, for example, about 5, 10, 15, 20, 25, 30, 35, 40, 45 or 50 nucleotides in length. An antisense nucleic acid of the invention can be constructed using chemical synthesis or enzymatic ligation reactions using procedures known in the art. For example, an antisense nucleic acid (e.g., an antisense oligonucleotide) can be chemically synthesized using naturally occurring nucleotides or variously modified nucleotides designed to increase the biological stability of the molecules or to increase the physical stability ofthe duplex formed between the antisense and sense nucleic acids (e.g., phosphorothioate derivatives and acridine substituted nucleotides can be used).
Examples of modified nucleotides that can be used to generate the antisense nucleic acid include: 5-fluorouracil, 5-bromouracil, 5-chlorouracil, 5-iodouracil, hypoxanthine, xanthine, 4-acetylcytosine, 5-(carboxyhydroxylmethyl) uracil, beta-D-mannosylqueosine, 5-carboxymethylaminomethyl-2-thiouridine, 5-carboxymethylaminomethyluracil, dihydrouracil, beta-D-galactosylqueosine, inosine, N6-isopentenyladenine, 1-methylguanine, 1-methylinosine, 2,2-dimethylguanine, 2-methyladenine, 2-methylguanine, 3-methylcytosine, 5-methylcytosine, N6-adenine, 7-methylguanine, 5-methylaminomethyluracil, 5-methoxyaminomethyl-2-thiouracil, 5'-methoxycarboxymethyluracil, 5-methoxyuracil, 2-methylthio-N6-isopentenyladenine, uracil-5-oxyacetic acid (v), wybutoxosine, pseudouracil, queosine, 2-thiocytosine, 5-methyl-2-thiouracil, 2-thiouracil, 4-thiouracil, 5-methyluracil, uracil-5-oxyacetic acid methylester, uracil-5-oxyacetic acid (v), 5-methyl-2-thiouracil, 3-(3-amino-3-N-2-carboxypropyl) uracil, (acp3)w, and 2,6-diaminopurine.
Alternatively, the antisense nucleic acid can be produced biologically using an expression vector into
6 PCT/US02/17428 which a nucleic acid has been subcloned in an antisense orientation (i.e., RNA
transcribed from the inserted nucleic acid will be of an antisense orientation to a target nucleic acid of interest, described further in the following subsection).
The antisense nucleic acid molecules of the invention are typically administered to a subject or generated in situ such that they hybridize with or bind to cellular mRNA
andlor genomic DNA encoding A NOVX protein to thereby inhibit expression of the protein (e.g., by inhibiting transcription and/or translation). The hybridization can be by conventional nucleotide complementarity to form a stable duplex, or, for example, in the case of an antisense nucleic acid molecule that binds to DNA duplexes, through specific interactions in the major groove of the double helix. An example of a route of administration of antisense nucleic acid molecules of the invention includes direct injection at a tissue site. Alternatively, antisense nucleic acid molecules can be modified to target selected cells and then administered systemically. For example, for systemic administration, antisense molecules can be modified such that they specifically bind to receptors or antigens expressed on a selected cell surface (e.g., by linking the antisense nucleic acid molecules to peptides or antibodies that bind to cell surface receptors or antigens). The antisense nucleic acid molecules can also be delivered to cells using the vectors described herein. To achieve sufficient nucleic acid molecules, vector constructs in which the antisense nucleic acid molecule is placed under the control of a strong pol II
or pol III promoter are preferred.
In yet another embodiment, the antisense nucleic acid molecule of the invention is an cc-anomeric nucleic acid molecule. A oc-anomeric nucleic acid molecule forms specific double-stranded hybrids with complementary RNA in which, contrary to the usual (3-units, the strands run parallel to each other. See, e.g., Gaultier, et al., 1987.
Nucl. Acids Res. 15:
6625-6641. The antisense nucleic acid molecule can also comprise a 2'-o-methylribonucleotide (See, e.g., moue, et al. 1987. Nuct. Acids Res. 15:
6131-6148) or a chimeric RNA-DNA analogue (See, e.g., moue, et al., 1987. FEBS Lett. 215:
327-330.

Ribozymes and PNA Moieties Nucleic acid modifications include, by way of non-limiting example, modified bases, and nucleic acids whose sugar phosphate backbones are modified or derivatized.
These modifications are carried out at least in part to enhance the chemical stability of the modified nucleic acid, such that they may be used, for example, as antisense binding nucleic acids in therapeutic applications in a subject.
In one embodiment, an antisense nucleic acid of the invention is a ribozyme.
Ribozymes are catalytic RNA molecules with ribonuclease activity that are capable of cleaving a single-stranded nucleic acid, such as an mRNA, to which they have a complementary region. Thus, ribozymes (e.g., hammerhead ribozymes as described in Haselhoff and Gerlach 1988. Nature 334: 585-591) can be used to catalytically cleave NOVX mRNA transcripts to thereby inhibit translation of NOVX mRNA. A ribozyme having specificity for a NOVX-encoding nucleic acid can be designed based upon the nucleotide sequence of A NOVX cDNA disclosed herein (i.e., SEQ ID NOS:2n-1, wherein n is an integer between 1 and 54). For example, a derivative of a Tetrahymena L-19 IVS RNA can be constructed in which the nucleotide sequence of the active site is complementary to the nucleotide sequence to be cleaved in a NOVX-encoding mRNA.
See, e.g., U.S. Patent 4,987,071 to Cech, et al. and U.S. Patent 5,116,742 to Cech, et al.
NOVX mRNA can also be used to select a catalytic RNA having a specific ribonuclease activity from a pool of RNA molecules. See, e.g., Bartel et al., (1993) Science 261:1411-1418.
Alternatively, NOVX gene expression can be inhibited by targeting nucleotide sequences complementary to the regulatory region of the NOVX nucleic acid (e.g., the NOVX promoter and/or enhancers) to form triple helical structures that prevent transcription of the NOVX gene in target cells. See, e.g., Helene, 1991.
Anticancer Drug Des. 6: 569-84; Helene, et al. 1992. Ann. N. Y. Acad. Sci. 660: 27-36; Maher, 1992.
Bioassays 14: 807-15.
In various embodiments, the NOVX nucleic acids can be modified at the base moiety, sugar moiety or phosphate backbone to improve, e.g., the stability, hybridization, or solubility of the molecule. For example, the deoxyribose phosphate backbone of the nucleic acids can be modified to generate peptide nucleic acids. See, e.g., Hyrup, et al., 1996. Bioorg Med Chem 4: 5-23. As used herein, the terms "peptide nucleic acids" or "PNAs" refer to nucleic acid mimics (e.g , DNA mimics) in which the deoxyribose phosphate backbone is replaced by a pseudopeptide backbone and only the four natural nucleobases are retained. The neutral backbone of PNAs has been shown to allow for specific hybridization to DNA and RNA under conditions of low ionic strength.
The synthesis of PNA oligomers can be performed using standard solid phase peptide synthesis protocols as described in Hyrup, et al., 1996. supra; Perry-O'Keefe, et al., 1996. Proc.
Natl. Acad. Sci. USA 93: 14670-14675.
PNAs of NOVX can be used in therapeutic and diagnostic applications. For example, PNAs can be used as antisense or antigene agents for sequence-specific modulation of gene expression by, e.g., inducing transcription or translation arrest or inhibiting replication. PNAs of NOVX can also be used, for example, in the analysis of single base pair mutations in a gene (e.g., PNA directed PCR clamping; as artificial restriction enzymes when used in combination with other enzymes, e.g., S~
nucleases (See, Hyrup, et al., 1996.supra); or as probes or primers for DNA sequence and hybridization (See, Hyrup, et al., 1996, supra; Perry-O'Keefe, et al., 1996. supra).
In another embodiment, PNAs of NOVX can be modified, e.g., to enhance their stability or cellular uptake, by attaching lipophilic or other helper groups to PNA, by the formation of PNA-DNA chimeras, or by the use of liposomes or other techniques of drug delivery known in the art. For example, PNA-DNA chimeras of NOVX can be generated that may combine the advantageous properties of PNA and DNA. Such chimeras allow DNA recognition enzymes (e.g., RNase H and DNA polymerases) to interact with the DNA portion while the PNA portion would provide high binding affinity and specificity.
PNA-DNA chimeras can be linked using linkers of appropriate lengths selected in terms of base stacking, number of bonds between the nucleobases, and orientation (see, Hyrup, et al., 1996. supra). The synthesis of PNA-DNA chimeras can be performed as described in Hyrup, et al., 1996. supra and Finn, et al., 1996. Nucl Acids Res 24: 3357-3363. For example, a DNA chain can be synthesized on a solid support using standard phosphoramidite coupling chemistry, and modified nucleoside analogs, e.g., 5'-(4-methoxytrityl)amino-5'-deoxy-thymidine phosphoramidite, can be used between the PNA and the 5' end of DNA. See, e.g., Mag, et al., 1989. Nucl Acid Res 17:
5973-5988.
PNA monomers are then coupled in a stepwise manner to produce a chimeric molecule with a 5' PNA segment and a 3' DNA segment. See, e.g., Finn, et al., 1996.
supra.
Alternatively, chimeric molecules can be synthesized with a 5' DNA segment and a 3' PNA segment. See, e.g., Petersen, et al., 1975. Bioorg. Med. Chem. Lett. 5:
1119-11124.
In other embodiments, the oligonucleotide may include other appended groups such as peptides (e.g:, for targeting host cell receptors in vivo), or agents facilitating transport across the cell membrane (see, e.g., Letsinger, et al., 1989. Proc.
Natl. Acad. Sci.
U.S.A. 86: 6553-6556; Lemaitre, et al., 1987. Proc. Natl. Acad. Sci. 84: 648-652; PCT
Publication No. W088/09810) or the blood-brain barrier (see, e.g., PCT
Publication No.
WO 89/10134). In addition, oligonucleotides can be modified with hybridization triggered cleavage agents (see, e.g., Krol, et al., 1988. BioTechniques 6:958-976) or intercalating agents (see, e.g., Zon, 1988. Pharm. Res. 5: 539-549). To this end, the oligonucleotide may be conjugated to another molecule, e.g., a peptide, a hybridization triggered cross-linking agent, a transport agent, a hybridization-triggered cleavage agent, and the like.
NOVX Polypeptides A polypeptide according to the invention includes a polypeptide including the amino acid sequence of NOVX polypeptides whose sequences are provided in SEQ
ID
NOS:2n, wherein n is an integer between 1 and 54. The invention also includes a mutant or variant protein any of whose residues may be changed from the corresponding residues shown in SEQ ID NOS:2n, wherein n is an integer between 1 and 54, while still encoding a protein that maintains its NOVX activities and physiological functions, or a functional fragment thereof.
In general, A NOVX variant that preserves NOVX-like function includes any variant in which residues at a particular position in the sequence have been substituted by other amino acids, and further include the possibility of inserting an additional residue or residues between two residues of the parent protein as well as the possibility of deleting . one or more residues from the parent sequence. Any amino acid substitution, insertion, or deletion is encompassed by the invention. In favorable circumstances, the substitution is a conservative substitution as defined above.
One aspect of the invention pertains to isolated NOVX proteins, and biologically-active portions thereof, or derivatives, fragments, analogs or homologs thereof. Also provided are polypeptide fragments suitable for use as immunogens to raise anti-NOVX antibodies. In one embodiment, native NOVX proteins can be isolated from cells or tissue sources by an appropriate purification scheme using standard protein purification techniques. In another embodiment, NOVX proteins are produced by recombinant DNA techniques. Alternative to recombinant expression, A NOVX
protein or polypeptide can be synthesized chemically using standard peptide synthesis techniques.

An "isolated" or "purified" polypeptide or protein or biologically-active portion thereof is substantially free of cellular material or other contaminating proteins from the cell or tissue source from which the NOVX protein is derived, or substantially free from chemical precursors or other chemicals when chemically synthesized. The language "substantially free of cellular material" includes preparations of NOVX
proteins in which the protein is separated from cellular components of the cells from which it is isolated or recombinantly-produced. In one embodiment, the language "substantially free of cellular material" includes preparations of NOVX proteins having less than about 30%
(by dry weight) of non-NOVX proteins (also referred to herein as a "contaminating protein"), more preferably less than about 20% of non-NOVX proteins, still more preferably less than about 10% of non-NOVX proteins, and most preferably less than about 5% of non-NOVX proteins. When the NOVX protein or biologically-active portion thereof is recombinantly-produced, it is also preferably substantially free of culture medium, i.e., culture medium represents less than about 20%, more preferably less than about 10%, and most preferably less than about 5% of the volume of the NOVX protein preparation.
The language "substantially free of chemical precursors or other chemicals"
includes preparations of NOVX proteins in which the protein is separated from chemical precursors or other chemicals that are involved in the synthesis of the protein. In one embodiment, the language "substantially free of chemical precursors or other chemicals"
includes preparations of NOVX proteins having less than about 30% (by dry weight) of chemical precursors or non-NOVX chemicals, more preferably less than about 20%
chemical precursors or non-NOVX chemicals, still more preferably less than about 10%
chemical precursors or non-NOVX chemicals, and most preferably less than about 5%
chemical precursors or non-NOVX chemicals.
Biologically-active portions of NOVX proteins include peptides comprising amino acid sequences sufficiently homologous to or derived from the amino acid sequences of the NOVX proteins (e.g., the amino acid sequence shown in SEQ ID NOS:2n, wherein n is an integer between 1 and 54) that include fewer amino acids than the full-length NOVX
proteins, and exhibit at least one activity of A NOVX protein. Typically, biologically-active portions comprise a domain or motif with at least one activity of the NOVX protein. A biologically-active portion of A NOVX protein can be a polypeptide which is, for example, 10, 25, 50, 100 or more amino acid residues in length.

Moreover, other biologically-active portions, in which other regions of the protein are deleted, can be prepared by recombinant techniques and evaluated for one or more of the functional activities of a native NOVX protein.
In an embodiment, the NOVX protein has an amino acid sequence shown SEQ ID
NOS:2n, wherein n is an integer between 1 and 54. In other embodiments, the NOVX
protein is substantially homologous to SEQ ID NOS:2n, wherein n is an integer between 1 and 54, and retains the functional activity of the protein of SEQ ID NOS:2n, wherein n is an integer between 1 and 54, yet differs in amino acid sequence due to natural allelic variation or mutagenesis, as described in detail, below. Accordingly, in another embodiment, the NOVX protein is a protein that comprises an amino acid sequence at least about 45% homologous to the amino acid sequence SEQ ID NOS:2n, wherein n is an integer between 1 and 54, and retains the functional activity of the NOVX
proteins of SEQ ID NOS:2n, wherein n is an integer between 1 and 54.

To determine the percent homology of two amino acid sequences or of two nucleic acids, the sequences are aligned for optimal comparison purposes (e.g., gaps can be introduced in the sequence of a first amino acid or nucleic acid sequence for optimal alignment with a second amino or nucleic acid sequence). The amino acid residues or nucleotides at corresponding amino acid positions or nucleotide positions are then compared. When a position in the first sequence is occupied by the same amino acid residue or nucleotide as the corresponding position in the second sequence, then the molecules are homologous at that position (i.e., as used herein amino acid or nucleic acid "homology" is equivalent to amino acid or nucleic acid "identity").
The nucleic acid sequence homology may be determined as the degree of identity between two sequences. The homology may be determined using computer programs known in the art, such as GAP software provided in the GCG program package.
See, Needleman and Wunsch,1970. JMoI Biol 48: 443-453. Using GCG GAP software with the following settings for nucleic acid sequence comparison: GAP creation penalty of 5.0 and GAP extension penalty of 0.3, the coding region of the analogous nucleic acid sequences referred to above exhibits a degree of identity preferably of at least 70%, 75%, 80%, 85%, 90%, 95%, 98%, or 99%, with the CDS (encoding) part of the DNA
sequence shown in SEQ lD NOS:2n-1, wherein n is an integer between 1 and 54.

The term "sequence identity" refers to the degree to which two polynucleotide or polypeptide sequences are identical on a residue-by-residue basis over a particular region of comparison. The term "percentage of sequence identity" is calculated by comparing two optimally aligned sequences over that region of comparison, determining the number of positions at which the identical nucleic acid base (e.g., A, T, C, G, U, or I, in the case of nucleic acids) occurs in both sequences to yield the number of matched positions, dividing the number of matched positions by the total number of positions in the region of comparison (i.e., the window size), and multiplying the result by 100 to yield the percentage of sequence identity. The term "substantial identity" as used herein denotes a characteristic of a polynucleotide sequence, wherein the polynucleotide comprises a sequence that has at least 80 percent sequence identity, preferably at least 85 percent identity and often 90 to 95 percent sequence identity, more usually at least 99 percent sequence identity as compared to a reference sequence over a comparison region.
CHIMERIC AND FUSION PROTEINS
The invention also provides NOVX chimeric or fusion proteins. As used herein, A
NOVX "chimeric protein" or "fusion protein" comprises A NOVX polypeptide operatively-linked to a non-NOVX polypeptide. An "NOVX polypeptide" refers to a polypeptide having an amino acid sequence corresponding to A NOVX protein SEQ
ID
NOS:2n, wherein n is an integer between l and 54, whereas a "non-NOVX
polypeptide"
refers to a polypeptide having an amino acid sequence corresponding to a protein that is not substantially homologous to the NOVX protein, e.g., a protein that is different from the NOVX protein and that is derived from the same or a different organism.
Within A
NOVX fusion protein the NOVX polypeptide can correspond to all or a portion of A
NOVX protein. In one. embodiment, A NOVX fusion protein comprises at least one biologically active portion of A NOVX protein. In another embodiment, A NOVX
fusion protein comprises at least two biologically active portions of A NOVX protein.
In yet another embodiment, A NOVX fusion protein comprises at least three biologically active portions of A NOVX protein. Within the fusion protein, the term "operatively-linked" is intended to indicate that the NOVX polypeptide and the non-NOVX polypeptide are fused in-frame with one another. The non-NOVX polypeptide can be fused to the N-terminus or C-terminus of the NOVX polypeptide.
In one embodiment, the fusion protein is a GST-NOVX fusion protein in which the NOVX sequences are fused to the C-terminus of the GST (glutathione S-transferase) sequences. Such fusion proteins can facilitate the purification of recombinant NOVX
polypeptides.
In another embodiment, the fusion protein is A NOVX protein containing a heterologous signal sequence at its N-terminus. In certain host cells (e.g., mammalian host cells), expression and/or secretion of NOVX can be increased through use of a heterologous signal sequence.
In yet another embodiment, the fusion protein is a NOVX-immunoglobulin fusion protein in which the NOVX sequences are fused to sequences derived from a member of the immunoglobulin protein family. The NOVX-immunoglobulin fusion proteins of the invention can be incorporated into pharmaceutical compositions and administered to a subject to inhibit an interaction between A NOVX ligand and A NOVX protein on the surface of a cell, to thereby suppress NOVX-mediated signal transduction in vivo. The NOVX-immunoglobulin fusion proteins can be used to affect the bioavailability of A
NOVX cognate ligand. Inhibition of the NOVX ligand/NOVX interaction may be useful 1 S therapeutically for both the treatment of proliferative and differentiative disorders, as well as modulating (e.g. promoting or inhibiting) cell survival. Moreover, the NOVX-immunoglobulin fusion proteins of the invention can be used as immunogens to produce anti-NOVX antibodies in a subject, to purify NOVX ligands, and in screening assays to identify molecules that inhibit the interaction of NOVX with A NOVX
ligand.
A NOVX chimeric or fusion protein of the invention can be produced by standard recombinant DNA techniques. For example, DNA fragments coding for the different polypeptide sequences are ligated together in-frame in accordance with conventional techniques, e.g., by employing blunt-ended or stagger-ended termini for ligation, restriction enzyme digestion to provide for appropriate termini, filling-in of cohesive ends as appropriate, alkaline phosphatase treatment to avoid undesirable joining, and enzymatic ligation. In another embodiment, the fusion gene can be synthesized by conventional techniques including automated DNA synthesizers. Alternatively, PCR
amplification of gene fragments can be carried out using anchor primers that give rise to complementary overhangs between two consecutive gene fragments that can subsequently be annealed and reamplified to generate a chimeric gene sequence (see, e.g., Ausubel, et al.
(eds.) CURRENT PROTOCOLS 1N MOLECULAR BIOLOGY, John Wiley & Sons, 1992). Moreover, many expression vectors are commercially available that already encode a fusion moiety (e.g., a GST polypeptide). A NOVX-encoding nucleic acid can be cloned into such an expression vector such that the fusion moiety is linked in-frame to the NOVX
protein.

NOVX AGONISTS AND ANTAGONISTS
The invention also pertains to variants of the NOVX proteins that function as either NOVX agonists (i.e., mimetics) or as NOVX antagonists. Variants of the NOVX
protein can be generated by mutagenesis (e.g., discrete point mutation or truncation of the NOVX
protein). An agonist of the NOVX protein can retain substantial 1y the same, or a subset of, the biological activities of the naturally occurring form of the NOVX protein.
An antagonist of the NOVX protein,can inhibit one or more of the activities of the naturally occurring form of the NOVX protein by, for example, competitively binding to a downstream or upstream member of a cellular signaling cascade, which includes the NOVX protein. Thus, specific biological effects can be elicited by treatment with a variant of limited function. In one embodiment, treatment of a subject with a variant having a subset of the biological activities of the naturally occurring form of the protein has fewer side effects in a subject relative to treatment with the naturally occurring form of the NOVX proteins.
Variants of the NOVX proteins that function as either NOVX agonists (i.e., mimetics) or as NOVX antagonists can be identified by screening combinatorial libraries of mutants (e.g., truncation mutants) of the NOVX proteins for NOVX protein agonist or antagonist activity. In one embodiment, a variegated library of NOVX variants is generated by combinatorial mutagenesis at the nucleic acid level and is encoded by a variegated gene library. A variegated library of NOVX variants can be produced by, for example, enzymatically ligating a mixture of synthetic oligonucleotides into gene sequences such that a degenerate set of potential NOVX sequences is expressible as individual polypeptides, or alternatively, as a set of larger fusion proteins (e.g., for phage display) containing the set of NOVX sequences therein. There are a variety of methods, which can be used to produce libraries of potential NOVX variants from a degenerate oligonucleotide sequence. Chemical synthesis of a degenerate gene sequence can be performed in an automatic DNA synthesizer, and the synthetic gene then ligated into an appropriate expression vector. Use of a degenerate set of genes allows for the provision, in one mixture, of all of the sequences encoding the desired set of potential NOVX
sequences. Methods for synthesizing degenerate oligonucleotides are well known within the art. See, e.g., Narang, 1983. Tetrahedron 39: 3; Itakura, et al., 1984.
Annu. Rev.
Biochem. 53: 323; Itakura, et al., 1984. Science 198: 1056; Ike, et al., 1983.
Nucl. Acids Res. 11: 477.

POLYPEPTIDE LIBRARIES
In addition, libraries of fragments of the NOVX protein coding sequences can be used to generate a variegated population of NOVX fragments for screening and subsequent selection of variants of A NOVX protein. In one embodiment, a library of coding sequence fragments can be generated by treating a double stranded PCR
fragment of A NOVX coding sequence with a nuclease under conditions wherein nicking occurs only about once per molecule, denaturing the double stranded DNA, renaturing the DNA
to form double-stranded DNA that can include sense/antisense pairs from different nicked products, removing single stranded portions from reformed duplexes by treatment with Sl nuclease, and ligating the resulting fragment library into an expression vector. By this method, expression libraries can be derived which encodes N-terminal and internal fragments of various sizes of the NOVX proteins.
Various techniques are known in the art for screening gene products of combinatorial libraries made by point mutations or truncation, and for screening cDNA
libraries for gene products having a selected property. Such techniques are adaptable for rapid screening of the gene libraries generated by the combinatorial mutagenesis of NOVX proteins. The most widely used techniques, which are amenable to high throughput analysis, for screening large gene libraries typically include cloning the gene library into replicable expression vectors, transforming appropriate cells with the resulting library of vectors, and expressing the combinatorial genes under conditions in which detection of a desired activity facilitates isolation of the vector encoding the gene whose product was detected. Recursive ensemble mutagenesis (REM), a new technique that enhances the frequency of functional mutants in the libraries, can be used in combination with the screening assays to identify NOVX variants. See, e.g., Arkin and Yourvan, 1992.
Proc. Natl. Acad. Sci. USA 89: 7811-7815; Delgrave, et al., 1993. Protein Engineering 6:327-331.
NOVX Antibodies The term "antibody" as used herein refers to immunoglobulin molecules and immunologically active portions of immunoglobulin (Ig) molecules, i.e., molecules that contain an antigen-binding site that specifically binds (immunoreacts with) an antigen.
Such antibodies include, but are not limited to, polyclonal, monoclonal, chimeric, single chain, Fab, Fab° and F(ab~2 fragments, and an Fab expression library.
In general, antibody molecules obtained from humans relates to any of the classes IgG, IgM, IgA, IgE and IgD, which differ from one another by the nature of the heavy chain present in the molecule.
Certain classes have subclasses as well, such as IgG~, IgG2, and others.
Furthermore, in humans, the light chain may be a kappa chain or a lambda chain. Reference herein to antibodies includes a reference to all such classes, subclasses and types of human antibody species.
An isolated protein of the invention intended to serve as an antigen, or a portion or fragment thereof, can be used as an immunogen to generate antibodies that immunospecifically bind the antigen, using standard techniques for polyclonal and monoclonal antibody preparation. The full-length protein can be used or, alternatively, the invention provides antigenic peptide fragments of the antigen for use as immunogens. An antigenic peptide fragment comprises at least 6 amino acid residues of the amino acid sequence of the full length protein, such as an amino acid sequence shown in SEQ ID
NOs: 2n, wherein n is an integer between 1 and 54, and encompasses an epitope thereof such that an antibody raised against the peptide forms a specific immune complex with the full length protein or with any fragment that contains the epitope.
Preferably, the antigenic peptide comprises at least 10 amino acid residues, or at least 15 amino acid residues, or at least 20 amino acid residues, or at least 30 amino acid residues. Preferred epitopes encompassed by the antigenic peptide are regions of the protein that are located on its surface; commonly these are hydrophilic regions.
In certain embodiments of the invention, at least one epitope encompassed by the antigenic peptide is a region of NOVX that is located on the surface of the protein, e.g., a hydrophilic region. A hydrophobicity analysis of the human NOVX protein sequence will indicate which regions of a NOVX polypeptide are particularly hydrophilic and, therefore, are likely to encode surface residues useful for targeting antibody production. As a means for targeting antibody production, hydropathy plots showing regions of hydrophilicity and hydrophobicity may be generated by any method well known in the art, including, for example, the Kyte Doolittle or the Hopp Woods methods, either with or without Fourier transformation. See, e.g., Hopp and Woods, 1981, Proc. Nat. Acad. Sci. USA 78:
3824-3828; Kyte and Doolittle 1982, J. Mol. Biol. 157: 105-1.42, each incorporated herein by reference in their entirety. Antibodies that are specific for one or more domains within an antigenic protein, or derivatives, fragments, analogs or homologs thereof, are also provided herein.

A protein of the invention, or a derivative, fragment, analog, homolog or ortholog thereof, may be utilized as an immunogen in the generation of antibodies that immunospecifically bind these protein components.
Various procedures known within the art may be used for the production of polyclonal or monoclonal antibodies directed against a protein of the invention, or against derivatives, fragments, analogs homologs or orthologs thereof (see, for example, Antibodies: A Laboratory Manual, Harlow E, and Lane D, 1988, Cold Spring Harbor i Laboratory Press, Cold Spring Harbor, NY, incorporated herein by reference).
Some of these antibodies are discussed below.
Polyclonal Antibodies For the production of polyclonal antibodies, various suitable host animals (e.g., rabbit, goat, mouse or other mammal) may be immunized by one or more injections with the native protein, a synthetic variant thereof, or a derivative of the foregoing. An appropriate immunogenic preparation can contain, for example, the naturally occurring immunogenic protein, a chemically synthesized polypeptide representing the immunogenic protein, or a recombinantly expressed immunogenic protein.
Furthermore, the protein may be conjugated to a second protein known to be immunogenic in the mammal being immunized. Examples of such immunogenic proteins include but are not limited to keyhole limpet hemocyanin, serum albumin, bovine thyroglobulin, and soybean trypsin inhibitor. The preparation can further include an adjuvant. Various adjuvants used to increase the immunological response include, but are not limited to, Freund's (complete and incomplete), mineral gels (e.g., aluminum hydroxide), surface active substances (e.g., lysolecithin, pluronic polyols, polyanions, peptides; oil emulsions, dinitrophenol, etc.), adjuvants usable in humans such as Bacille Calmette-Guerin and Corynebacterium parvum, or similar immunostimulatory agents. Additional examples of adjuvants which can be employed include MPL-TDM adjuvant (monophosphoryl Lipid A, synthetic trehalose dicorynomycolate).
The polyclonal antibody molecules directed against the immunogenic protein can be isolated from the mammal (e.g., from the blood) and further purified by well known techniques, such as affinity chromatography using protein A or protein G, which provide primarily the IgG fraction of immune serum. Subsequently, or alternatively, the specific antigen which is the target of the immunoglobulin sought, or an epitope thereof, may be immobilized on a column to purify the immune specific antibody by immunoa~nity chromatography. Purification of immunoglobulins is discussed, for example, by D.
Wilkinson (The Scientist, published by The Scientist, Inc., Philadelphia PA, Vol. 14, No. 8 (April 17, 2000), pp. 25-28).
Monoclonal Antibodies The term "monoclonal antibody" (MAb) or "monoclonal antibody composition", as used herein, refers to a population of antibody molecules that contain only one molecular species of antibody molecule consisting of a unique light chain gene product and a unique heavy chain gene product. In particular, the complementarity determining regions (CDRs) of the monoclonal antibody are identical in all the molecules of the population. MAbs thus contain an antigen binding site capable of immunoreacting with a particular epitope of the antigen characterized by a unique binding affinity for it.
Monoclonal antibodies can be prepared using hybridoma methods, such as those described by Kohler and Milstein, Nature. 256:495 (1975). In a hybridoma method, a mouse, hamster, or other appropriate host animal, is typically immunized with an immunizing agent to elicit lymphocytes that produce or are capable of producing antibodies that will specifically bind to the immunizing agent. Alternatively, the lymphocytes can be immunized in vitro.
The immunizing agent will typically include the protein antigen, a fragment thereof or a fusion protein thereof. Generally, either peripheral blood lymphocytes are used if cells of human origin are desired, or spleen cells or lymph node cells are used if non-human mammalian sources are desired. The lymphocytes are then fused with an immortalized cell line using a suitable fusing agent, such as polyethylene glycol, to form a hybridoma cell [coding, Monoclonal Antibodies: Principles and Practice, Academic Press, (1986) pp. 59-103. Immortalized cell lines are usually transformed mammalian cells, particularly myeloma cells of rodent, bovine and human origin. Usually, rat or mouse myeloma cell lines are employed. The hybridoma cells can be cultured in a suitable culture medium that preferably contains one or more substances that inhibit the growth or survival of the unfused, immortalized cells. For example, if the parental cells lack the enzyme hypoxanthine guanine phosphoribosyl transferase (HGPRT or HPRT), the culture medium for the hybridomas typically will include hypoxanthine, aminopterin, and thymidine ("HAT medium"), which substances prevent the growth of HGPRT-deficient cells.

Preferred immortalized cell lines are those that fuse efficiently, support stable high level expression of antibody by the selected antibody-producing cells, and are sensitive to a medium such as HAT medium. More preferred immortalized cell lines are murine myeloma lines, which can be obtained, for instance, from the Salk Institute Cell Distribution Center, San Diego, California and the American Type Culture Collection, Manassas, Virginia. Human myeloma and mouse-human heteromyeloma cell lines also have been described for the production of human monoclonal antibodies [Kozbor, J.
Immunol., 133:3001 (1984); Brodeur et al., Monoclonal Antibody Production Techniques an~lications, Marcel Dekker, Inc., New York, (1987) pp. 51-63].
The culture medium in which the hybridoma cells are cultured can then be assayed for the presence of monoclonal antibodies directed against the antigen.
Preferably, the binding specificity of monoclonal antibodies produced by the hybridoma cells is determined by immunoprecipitation or by an in vitro binding assay, such as radioimmunoassay (RIA) or enzyme-linked immunoabsorbent assay (ELISA). Such techniques and assays are known in the art. The binding affinity of the monoclonal antibody can, for example, be determined by the Scatchard analysis of Munson and Pollard, Anal. Biochem., 107:220 (1980). It is an objective, especially important in therapeutic applications of monoclonal antibodies, to identify antibodies having a high degree of specificity and a high binding affinity for the target antigen.
A$er the desired hybridoma cells are identified; the clones can be subcloned by limiting dilution procedures and grown by standard methods (Goding,1986).
Suitable culture media for this purpose include, for example, Dulbecco's Modified Eagle's Medium and RPMI-1640 medium. Alternatively, the hybridoma cells can be grown in vivo as ascites in a mammal.
The monoclonal antibodies secreted by the subclones can be isolated or purified from the culture medium or ascites fluid by conventional immunoglobulin purification procedures such as, for example, protein A-Sepharose, hydroxylapatite chromatography, gel electrophoresis, dialysis, or affinity chromatography.
The monoclonal antibodies can also be made by recombinant DNA methods, such as those described in U.S. Patent No. 4,816,567. DNA encoding the monoclonal antibodies of the invention can be readily isolated and sequenced using conventional procedures (e.g., by using oligonucleotide probes that are capable of binding specifically to genes encoding the heavy and light chains of murine antibodies). The hybridoma cells of the invention serve as a preferred source of such DNA. Once isolated, the DNA can be placed into expression vectors, which are then transfected into host cells such as simian COS cells, Chinese hamster ovary (CHO) cells, or myeloma cells that do not otherwise produce immunoglobulin protein, to obtain the synthesis of monoclonal antibodies in the recombinant host cells. The DNA also can be modified, for example, by substituting the coding sequence for human heavy and light chain constant domains in place of the homologous murine sequences (U.S. Patent No. 4,816,567; Morrison, Nature 368, (1994)) or by covalently joining to the immunoglobulin coding sequence all or part of the coding sequence for a non-immunoglobulin polypeptide. Such a non-immunoglobulin polypeptide can be substituted for the constant domains of an antibody of the invention, or can be substituted for the variable domains of one antigen-combining site of an antibody of the invention to create a chimeric bivalent antibody.
Humanized Antibodies The antibodies directed against the protein antigens of the invention can further comprise humanized antibodies or human antibodies. These antibodies are suitable for administration to humans without engendering an immune response by the human against the administered immunoglobulin. Humanized forms of antibodies are chimeric immunoglobulins, immunoglobulin chains or fragments thereof (such as Fv, Fab, Fab', F(ab')~ or other antigen-binding subsequences of antibodies) that are principally comprised of the sequence of a human immunoglobulin, and contain minimal sequence derived from a non-human immunoglobulin. Humanization can be performed following the method of Winter and co-workers (Jones et al., Nature, 321:522-525 (1986); Riechmann et al., Nature, 332:323-327 (1988); Verhoeyen et al., Science, 239:1534-1536 (1988)), by substituting rodent CDRs or CDR sequences for the corresponding sequences of a human antibody. (See also U.S. Patent No. 5,225,539.) In some instances, Fv framework residues of the human immunoglobulin are replaced by corresponding non-human residues. Humanized antibodies can also comprise residues which are found neither in the recipient antibody nor in the imported CDR or framework sequences. In general, the humanized antibody will comprise substantially all of at least one, and typically two, variable domains, in which all or substantially all of the CDR regions correspond to those of a non-human immunoglobulin and all or substantially all of the framework regions are those of a human immunoglobulin consensus sequence. The humanized antibody optimally also will comprise at least a portion of an immunoglobulin constant region (Fc), typically that of a human immunoglobulin (Jones et al., 1986; Riechmann et al., 1988; and Presta, Curr. Op. Struct. Biol.. 2:593-596 (1992)).
Human Antibodies Fully human antibodies essentially relate to antibody molecules in which the entire sequence of both the light chain and the heavy chain, including the CDRs, arise from human genes. Such antibodies are termed "human antibodies", or "fully human antibodies" herein. Human monoclonal antibodies can be prepared by the trioma technique; the human B-cell hybridoma technique (see I~ozbor, et al., 1983 Immunol Today 4: 72) and the EBV hybridoma technique to produce human monoclonal antibodies (see Cole, et al., 1985 In: MONOCLONAL ANTIBODIES AND CANCER THERAPY, Alan R.
Liss, Inc., pp. 77-96). Human monoclonal antibodies may be utilized in the practice of the present invention and may be produced by using human hybridomas (see Cote, et al., 1983. Proc Natl Acad Sci USA 80: 2026-2030) or by transforming human B-cells with Epstein Barr Virus in vitro (see Cole, et al., 1985 In: MONOCLONAL ANTIBODIES
AND
CANCER THERAPY, Alan R. Liss, Inc., pp. 77-96).
In addition, human antibodies can also be produced using additional techniques, including phage display libraries (Hoogenboom and Winter, 3. Mol. Biol., 227:381 (1991);
Marks et al., J. Mol. Biol.. 222:581 (1991)). Similarly, human antibodies can be made by introducing human immunoglobulin loci into transgenic animals, e.g., mice in which the endogenous immunoglobulin genes have been partially or completely inactivated.
Upon challenge, human antibody production is observed, which closely resembles that seen in humans in all respects, including gene rearrangement, assembly, and antibody repertoire.
This approach is described, for example, in U.S. Patent Nos. 5,545,807;
5,545,806;
5,569,825; 5,625,126; 5,633,425; 5,661,016, and in Marks et al.
(Bio/Technolo~v 10, 779-783 (1992)); Lonberg et al. ature 368 856-859 (1994)); Morrison ( Nature 368, 812-13 (1994)); Fishwild et al,( Nature Biotechnolo~.y 4 845-51 (1996));
Neuberger (Nature Biotechnology 4 826 (1996)); and Lonberg and Huszar (Intern. Rev.
Immunol.
13 65-93 (1995)).
Human antibodies may additionally be produced using transgenic nonhuman animals which are modified so as to produce fully human antibodies rather than the animal's endogenous antibodies in response to challenge by an antigen. (See PCT
publication W094/02602). The endogenous genes encoding the heavy and light immunoglobulin chains in the nonhuman host have been incapacitated, and active loci encoding human heavy and light chain imrnunoglobulins are inserted into the host's genome. The human genes are incorporated, for example, using yeast artificial chromosomes containing the requisite human DNA segments. An animal which provides all the desired modifications is then obtained as progeny by crossbreeding intermediate transgenic animals containing fewer than the full complement of the modifications. The preferred embodiment of such a nonhuman animal is a mouse, and is termed the XenomouseTM as disclosed in PCT publications WO 96/33735 and WO 96/34096. This animal produces B cells which secrete fully human immunoglobulins. The antibodies can be obtained directly from the animal after immunization with an immunogen of interest, as, for example, a preparation of a polyclonal antibody, or alternatively from immortalized B cells derived from the animal, such as hybridomas producing monoclonal antibodies.
Additionally, the genes encoding the immunoglobulins with human variable regions can be recovered and expressed to obtain the antibodies directly, or can be further modified to obtain analogs of antibodies such as, for example, single chain Fv molecules.
An example of a method of producing a nonhuman host, exemplified as a mouse, lacking expression of an endogenous immunoglobulin heavy chain is disclosed in U.S.
Patent No. 5,939,598. It can be obtained by a method including deleting the J
segment genes from at least one endogenous heavy chain locus in an embryonic stem cell to prevent rearrangement of the locus and to prevent formation of a transcript of a rearranged immunoglobulin heavy chain locus, the deletion being effected by a targeting vector containing a gene encoding a selectable marker; and producing from the embryonic stem cell a transgenic mouse whose somatic and germ cells contain the gene encoding the selectable marker.
A method for producing an antibody of interest, such as a human antibody, is disclosed in U.S. Patent No. 5,916,771. It includes introducing an expression vector that contains a nucleotide sequence encoding a heavy chain into one mammalian host cell in culture, introducing an expression vector containing a nucleotide sequence encoding a light chain into another mammalian host cell, and fusing the two cells to form a hybrid cell. The hybrid cell expresses an antibody containing the heavy chain and the light chain.
In a further improvement on this procedure, a method for identifying a clinically relevant epitope on an immunogen, and a correlative method for selecting an antibody that binds immunospecifically to the relevant epitope with high affinity, are disclosed in PCT
publication WO 99/53049.

Fab Fragments and Single Chain Antibodies According to the invention, techniques can be adapted for the production of single-chain antibodies specific to an antigenic protein of the invention (see e.g., U.S.
Patent No. 4,946,778). In addition, methods can be adapted for the construction of Fab expression libraries (see e.g., Huse, et al., 1989 Science 246: 1275-1281) to allow rapid and effective identification of monoclonal Fab fragments with the desired specificity for a protein or derivatives, fragments, analogs or homologs thereof. Antibody fragments that contain the idiotypes to a protein antigen may be produced by techniques known in the art including, but not limited to: (i) an F~ab~)2 fragment produced by pepsin digestion of an antibody molecule; (ii) an Fab fragment generated by reducing the disulfide bridges of an F(ab')z fragment; (iii) an Fab fragment generated by the treatment of the antibody molecule with papain and a reducing agent and (iv) F,, fragments.
Bispecific Antibodies Bispecific antibodies are monoclonal, preferably human or humanized, antibodies that have binding specificities for at least two different antigens. In the present case, one of the binding specificities is for an antigenic protein of the invention. The second binding target is any other antigen, and advantageously is a cell-surface protein or receptor or receptor subunit.
Methods for making bispecific antibodies are known in the art. Traditionally, the recombinant production of bispecific antibodies is based on the co-expression of two immunoglobulin heavy-chain/light-chain pairs, where the two heavy chains have different specificities (Milstein and Cuello, Nature. 305:537-539 (1983)). Because ofthe random assortment of immunoglobulin heavy and light chains, these hybridomas (quadromas) produce a potential mixture of ten different antibody molecules, of which only one has the correct bispecific structure. The purification of the correct molecule is usually accomplished by affinity chromatography steps. Similar procedures are disclosed in WO
93/08829, published 13 May 1993, and in Traunecker et al., EMBO J.. 10:3655-(1991).
Antibody variable domains with the desired binding specificities (antibody-antigen combining sites) can be fused to immunoglobulin constant domain sequences. The fusion preferably is with an immunoglobulin heavy-chain constant domain, comprising at least part of the hinge, CH2, and CH3 regions. It is preferred to have the first heavy-chain constant region (CHl) containing the site necessary for light-chain binding present in at least one of the fusions. DNAs encoding the immunoglobulin heavy-chain fusions and, if desired, the immunoglobulin light chain, are inserted into separate expression vectors, and are co-transfected into a suitable host organism. For further details of generating bispecific antibodies see, for example, Suresh et al., Methods in Enzvmoloay, 121:210 (1986).
According to another approach described in WO 96/27011, the interface between a pair of antibody molecules can be engineered to maximize the percentage of heterodimers which are recovered from recombinant cell culture. The preferred interface comprises at least a part of the CH3 region of an antibody constant domain. In this method, one or more small amino acid side chains from the interface of the first antibody molecule are replaced with larger side chains (e.g. tyrosine or tryptophan). Compensatory "cavities" of identical or similar size to the large side chains) are created on the interface of the second antibody molecule by replacing large amino acid side chains with smaller ones (e.g.
alanine or threonine). This provides a mechanism for increasing the yield of the heterodimer over other unwanted end-products such as homodimers.
Bispecific antibodies can be prepared as full length antibodies or antibody fragments (e.g. F(ab')2 bispecific antibodies). Techniques for generating bispecific antibodies from antibody fragments have been described in the literature., For example, bispecific antibodies can be prepared using chemical linkage. Brennan et al., Science 229:81 (1985) describe a procedure wherein intact antibodies are proteolytically cleaved to generate F(ab')a fragments. These fragments are reduced in the presence of the dithiol complexing agent sodium arsenite to stabilize vicinal dithiols and prevent intermolecular disulfide formation. The Fab' fragments generated are then converted to thionitrobenzoate (TNB) derivatives. One of the Fab'-TNB derivatives is then reconverted to the Fab'-thiol by reduction with mercaptoethylamine and is mixed with an equimolar amount of the other Fab'-T'NB derivative to form the bispecific antibody. The bispecific antibodies produced can be used as agents for the selective immobilization of enzymes.
Additionally, Fab' fragments can be directly recovered from E. coli and chemically coupled to form bispecific antibodies. Shalaby et al., J. Exp. Med. 175:217-225 (1992) describe the production of a fully humanized bispecific antibody F(ab')2 molecule. Each Fab' fragment was separately secreted from E. coli and subjected to directed chemical coupling in vitro to form the bispecific antibody. The bispecific antibody thus formed was able to bind to cells overexpressing the ErbB2 receptor and normal human T
cells, as well as trigger the lytic activity of human cytotoxic lymphocytes against human breast tumor targets.
Various techniques for making and isolating bispecific antibody fragments directly"from recombinant cell culture have also been described. For example, bispecific antibodies have been produced using leucine zippers. Kostelny et al., J.
Immunol.
148(5):1547-1553 (1992). The leucine zipper peptides from the Fos and Jun proteins were linked to the Fab' portions of two different antibodies by gene fusion. The antibody homodimers were reduced at the hinge region to form monomers and then re-oxidized to form the antibody heterodimers. This method can also be utilized for the production of antibody homodimers. The "diabody" technology described by Hollinger et al., Proc.
Natl. Acad. Sci. USA 90:6444-6448 (1993) has provided an alternative mechanism for making bispecific antibody fragments. The fragments comprise a heavy-chain variable domain (VH) connected to a light-chain variable domain (VL) by a linker which is too short to allow pairing between the two domains on the same chain. Accordingly, the VH and VL
domains of one fragment are forced to pair with the complementary VL and VH
domains of another fragment, thereby forming two antigen-binding sites. Another strategy for making bispecific antibody fragments by the use of single-chain Fv (sFv) dimers has also been reported. See, Gruber et al., J. Immunol. 152:5368 (1994).
Antibodies with more than two valencies are contemplated. For example, trispecific antibodies can be prepared. Tutt et al., J. Immunol. 147:60 (1991).
Exemplary bispecific antibodies can bind to two different epitopes, at least one of which originates in the protein antigen of the invention. Alternatively, an anti-antigenic arm of an immunoglobulin molecule can be combined with an arm which binds to a triggering molecule on a leukocyte such as a T-cell receptor molecule (e.g.
CD2, CD3, CD28, or B7), or Fc receptors for IgG (FcYR), such as FcYRI (CD64), FcyRII
(CD32) and FcyRIII (CD16) so as to focus cellular defense mechanisms to the cell expressing the particular antigen. Bispecific antibodies can also be used to direct cytotoxic agents to cells which express a particular antigen. These antibodies possess an antigen-binding arm and an arm which binds a cytotoxic agent or a radionuclide chelator, such as EOTUBE, DPTA, DOTA, or TETA. Another bispecific antibody of interest binds the protein antigen described herein and further binds tissue factor (TF).
Heteroconjugate Antibodies Heteroconjugate antibodies are also within the scope of the present invention.
I-Ieteroconjugate antibodies are composed of two covalently joined antibodies.
Such antibodies have, for example, been proposed to target immune system cells to unwanted cells (U.S. Patent No. 4,676,980), and for treatment of HIV infection (WO
91/00360; WO
92/200373; EP 03089). It is contemplated that the antibodies can be prepared in vitro using known methods in synthetic protein chemistry, including those involving crosslinking agents. For example, immunotoxins can be constructed using a disulfide exchange reaction or by forming a thioether bond. Examples of suitable reagents for this purpose include iminothiolate and methyl-4-mercaptobutyrimidate and those disclosed, for example, in U.S. Patent No. 4,676,980.
Effector Function Engineering It can be desirable to modify the antibody of the invention with respect to effector function, so as to enhance, e.g., the effectiveness of the antibody in treating cancer. For example, cysteine residues) can be introduced into the Fc region, thereby allowing interchain disulfide bond formation in this region. The homodimeric antibody thus generated can have improved internalization capability and/or increased complement-mediated cell killing and antibody-dependent cellular cytotoxicity (ADCC).
See Caron et al., J. Exp Med., 176: 1191-1195 (1992) and Shopes, J. Immunol., 148:
2918-2922 (1992). Homodimeric antibodies with enhanced anti-tumor activity can also be prepared using heterobifunctional cross-linkers as described in Wolff et al.
Cancer Research, 53: 2560-2565 (1993). Alternatively, an antibody can be engineered that has dual Fc regions and can thereby have enhanced complement lysis and ADCC
capabilities.
See Stevenson et al., Anti-Cancer Drag_Desi~n, 3: 219-230 (1989).
Immunoconjugates The invention also pertains to immunoconjugates comprising an antibody conjugated to a cytotoxic agent such as a chemotherapeutic agent, toxin (e.g., an enzymatically active toxin of bacterial, fungal, plant, or animal origin, or fragments thereof), or a radioactive isotope (i.e., a radioconjugate).
Chemotherapeutic agents useful in the generation of such immunoconjugates have been described above. Enzymatically active toxins and fragments thereof that can be used include diphtheria A chain, nonbinding active fragments of diphtheria toxin, exotoxin A
chain (from Pseudomonas aeruginosa), ricin A chain, abrin A chain, modeccin A
chain, alpha-sarcin, Aleurites fordii proteins, dianthin proteins, Phytolaca americana proteins (PAPI, PAPII, and PAP-S), momordica charantia inhibitor, curcin, crotin, sapaonaria officinalis inhibitor, gelonin, mitogellin, restrictocin, phenomycin, enomycin, and the tricothecenes. A variety of radionuclides are available for the production of radioconjugated antibodies. Examples include Z~ZBi, i3~I, ~3lln, 9°Y, and 186Re.
Conjugates of the antibody and cytotoxic agent are made using a variety of bifunctional protein-coupling agents such as N-succinimidyl-3-(2-pyridyldithiol) propionate (SPDP), iminothiolane (IT), bifunctional derivatives of imidoesters (such as dimethyl adipimidate HCL), active esters (such as disuccinimidyl suberate), aldehydes (such as glutareldehyde), bis-azido compounds (such as bis (p-azidobenzoyl) hexanediamine), bis-diazonium derivatives (such as bis-(p-diazoniumbenzoyl)-ethylenediamine), diisocyanates (such as tolyene 2,6-diisocyanate), and bis-active fluorine compounds (such as 1,5-difluoro-2,4-dinitrobenzene). For example, a ricin immunotoxin can be prepared as described in Vitetta et al., Science, 238: 1098 (1987). Carbon-14-labeled 1-isothiocyanatobenzyl-3-methyldiethylene triaminepentaacetic acid (MX-DTPA) is an exemplary chelating agent for conjugation of radionucleotide to the antibody.
See W094/ 11026.
In another embodiment, the antibody can be conjugated to a "receptor" (such streptavidin) for utilization in tumor pretargeting wherein the antibody-receptor conjugate is administered to the patient, followed by removal of unbound conjugate from the circulation using a clearing agent and then administration of a "ligand"
(e.g., avidin) that is in turn conjugated to a cytotoxic agent.
Immunoliposomes The antibodies disclosed herein can also be formulated as immunoliposomes.
Liposomes containing the antibody are prepared by methods known in the art, such as described in Epstein et al., Proc. Natl. Acad. Sci. USA, 82: 3688 (1985);
Hwang et al., Proc. Natl Acad. Sci. USA. 77: 4030 (1980); and U.S. Pat. Nos. 4,485,045 and 4,544,545.
Liposomes with enhanced circulation time are disclosed in U.S. Patent No.
5,013,556.
Particularly useful liposomes can be generated by the reverse-phase evaporation method with a lipid composition comprising phosphatidylcholine, cholesterol, and PEG-derivatized phosphatidylethanolamine (PEG-PE). Liposomes are extruded through f lters of defined pore size to yield liposomes with the desired diameter.
Fab' fragments of the antibody of the present invention can be conjugated to the liposomes as described in Martin et al ., J. Biol. Chem.. 257: 286-288 (1982) via a disulfide-interchange reaction. A
chemotherapeutic agent (such as Doxorubicin) is optionally contained within the liposome. See Gabizon et al., J. National Cancer Inst., 81 (19): 1484 ( 1989).
Diagnostic Applications of Antibodies Directed Against the Proteins of the Invention Antibodies directed against a protein of the invention may be used in methods known within the art relating to the localization and/or quantitation of the protein (e.g., for use in measuring levels of the protein within appropriate physiological samples, for use in diagnostic methods, for use in imaging the protein, and the like). In a given embodiment, antibodies against the proteins, or derivatives, fragments, analogs or homologs thereof, that contain the antigen binding domain, are utilized as pharmacologically-active compounds (see below).
An antibody specific for a protein of the invention can be used to isolate the protein by standard techniques, such as immunoa~nity chromatography or immunoprecipitation. Such an antibody can facilitate the purification of the natural protein antigen from cells and of recombinantly produced antigen expressed in host cells.
Moreover, such an antibody can be used to detect the antigenic protein (e.g., in a cellular lysate or cell supernatant) in order to evaluate the abundance and pattern of expression of the antigenic protein. Antibodies directed against the protein can be used diagnostically to monitor protein levels in tissue as part of a clinical testing procedure, e.g., to, for example, determine the efficacy of a given treatment regimen. Detection can be facilitated by coupling (i.e., physically linking) the antibody to a detectable substance.
Examples of detectable substances include various enzymes, prosthetic groups, fluorescent materials, 2S luminescent materials, bioluminescent materials, and radioactive materials.
Examples of suitable enzymes include horseradish peroxidase, alkaline phosphatase, (3-galactosidase, or acetylcholinesterase; examples of suitable prosthetic group complexes include streptavidinlbiotin 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 ~aSI,'3'I, ssS
or 3H.
Antibody Therapeutics Antibodies of the invention, including polyclonal, monoclonal, humanized and fully human antibodies, may used as therapeutic agents. Such agents will generally be employed to treat or prevent a disease or pathology in a subject. An antibody preparation, preferably one having high specificity and high affinity for its target antigen, is administered to the subject and will generally have an effect due to its binding with the target. Such an effect may be one of two kinds, depending on the specific nature of the interaction between the given antibody molecule and the target antigen in question. In the first instance, administration of the antibody may abrogate or inhibit the binding of the target with an endogenous ligand to which it naturally binds. In this case, the antibody binds to the target and masks a binding site of the naturally occurring ligand, wherein the ligand serves as an effector molecule. Thus the receptor mediates a signal transduction pathway for which ligand is responsible.
Alternatively, the effect may be one in which the antibody elicits a physiological result by virtue of binding to an effector binding site on the target molecule. In this case the target, a receptor having an endogenous ligand which may be absent or defective in the disease or pathology, binds the antibody as a surrogate effector ligand, initiating a receptor-based signal transduction event by the receptor.
A therapeutically effective amount of an antibody of the invention relates generally to the amount needed to achieve a therapeutic objective. As' noted above, this may be a binding interaction between the antibody and its target antigen that, in certain cases, interferes with the functioning of the target, and in other cases, promotes a physiological response. The amount required to be administered will furthermore depend on the binding affinity of the antibody for its specific antigen, and will also depend on the rate at which an administered antibody is depleted from the free volume other subject to which it is administered. Common ranges for therapeutically effective dosing of an antibody or antibody fragment of the invention may be, by way of nonlimiting example, from about 0.1 mg/kg body weight to about 50 mg/kg body weight.-Common dosing frequencies may range, for example, from twice daily to once a week.
Pharmaceutical Compositions of Antibodies Antibodies specifically binding a protein of the invention; as well as other molecules identified by the screening assays disclosed herein, can be administered for the treatment of various disorders in the form of pharmaceutical compositions.
Principles and considerations involved in preparing such compositions, as well as guidance in the choice of components are provided, for example, in Remington : The Science And Practice Of Pharmacy 19th ed. (Alfonso R. Gennaro, et al., editors) Mack Pub. Co., Easton, Pa. : 1995;
Drug Absorption Enhancement : Concepts, Possibilities, Limitations, And Trends, Harwood Academic Publishers, Langhorne, Pa., 1994; and Peptide And Protein Drug Delivery (Advances In Parenteral Sciences, Vol. 4), 1991, M. Dekleer, New York.
If the antigenic protein is intracellular and whole antibodies are used as inhibitors, internalizing antibodies are preferred. However, liposomes can also be used to deliver the antibody, or an antibody fragment, into cells. Where antibody fragments are used, the smallest inhibitory fragment that specifically binds to the binding domain of the target protein is preferred. For example, based upon the variable-region sequences of an antibody, peptide molecules can be designed that retain the ability to bind the target protein sequence. Such peptides can be synthesized chemically and/or produced by recombinant DNA technology. See, e.g., Marasco et al., Proc. Natl. Acad. Sci.
USA, 90:
7889-7893 (1993). The formulation herein can also contain more than one active 1 S compound as necessary for the particular indication being treated, preferably those with complementary activities that do not adversely affect each other.
Alternatively, or in addition, the composition can comprise an agent that enhances its function, such as, for example, a cytotoxic agent, cytokine, chemotherapeutic agent, or growth-inhibitory agent.
Such molecules are suitably present in combination in amounts that are effective for the purpose intended.
The active ingredients can also be entrapped in microcapsules prepared, for example, by coacervation techniques or by interfacial polymerization, for example, hydroxymethylcellulose or gelatin-microcapsules and poly-(methylmethacrylate) microcapsules, respectively, in colloidal drug delivery systems (for example, liposomes, albumin microspheres, microemulsions, nano-particles, and nanocapsules) or in macroemulsions.
The formulations to be used for in vivo administration must be sterile. This is readily accomplished by filtration through sterile filtration membranes.
Sustained-release preparations can be prepared. Suitable examples of sustained-release preparations include semipermeable matrices of solid hydrophobic polymers containing the antibody, which matrices are in the form of shaped articles, e.g., films, or microcapsules. Examples of sustained-release matrices include polyesters, hydrogels (for example, poly(2-hydroxyethyl-methacrylate), or poly(vinylalcohol)), polylactides (L1.S. Pat. No. 3,773,919), copolymers of L-glutamic acid and y ethyl-L-glutamate, non-degradable ethylene-vinyl acetate, degradable lactic acid-glycolic acid copolymers such as the LUPRON DEPOT ~ (injectable microspheres composed of lactic acid-glycolic acid copolymer and leuprolide acetate), and poly-D-(-)-3-hydroxybutyric acid. While polymers such as ethylene-vinyl acetate and lactic acid-glycolic acid enable release of molecules for over 100 days, certain hydrogels release proteins for shorter time periods.
ELISA Assay An agent for detecting an analyte protein is an antibody capable of binding to an analyte protein, preferably an antibody with a detectable label. Antibodies can be polyclonal, or more preferably, monoclonal. An intact antibody, or a fragment thereof (e.g., Fab or F~ab~2) can be used. The term "labeled", with regard to the probe or antibody, is intended to encompass direct labeling of the probe or antibody by coupling (i.e., physically linking) a detectable substance to the probe or antibody, as well as indirect labeling of the probe or antibody by reactivity with another reagent that is directly labeled.
Examples of indirect labeling include detection of a primary antibody using a fluorescently-labeled secondary antibody and end-labeling of a DNA probe with biotin such that it can be detected with fluorescently-labeled streptavidin. The term "biological sample" is intended to include tissues, cells and biological fluids isolated from a subject, as well as tissues, cells and fluids present within a subject. Included within the usage of the term "biological sample", therefore, is blood and a fraction or component of blood including blood serum, blood plasma, or lymph. That is, the detection method of the invention can be used to detect an analyte mRNA, protein, or genomic DNA in a biological sample in vitro as well as in vivo. For example, in vitro techniques for detection of an analyte mRNA include Northern hybridizations and in situ hybridizations.
In vitro techniques for detection of an analyte protein include enzyme linked immunosorbent assays (ELISAs), Western blots, immunoprecipitations, and immunofluorescence.
1~ vitro techniques for detection of an analyte genomic DNA include Southern hybridizations.
Procedures for conducting immunoassays are described, for example in "ELISA:
Theory and Practice: Methods in Molecular Biology", Vol. 42, J. R. Crowther (Ed.) Human Press, Totowa, NJ, 1995; "Immunoassay", E. Diamandis and T. Christopoulus, Academic Press, Inc., San Diego, CA, 1996; and "Practice and Thory of Enzyme Immunoassays", P.
Tijssen, Elsevier Science Publishers, Amsterdam, 1985. Furthermore, in vivo techniques for detection of an analyte protein include introducing into a subject a labeled anti-an analyte protein antibody. For example, the antibody can be labeled with a radioactive marker whose presence and location in a subject can be detected by standard imaging techniques.
NOVX Recombinant Expression Vectors and Host Cells Another aspect of the invention pertains to vectors, preferably expression vectors, containing a nucleic acid encoding A NOVX protein, or derivatives, fragments;
analogs or homologs thereof. As used herein, the term "vector" refers to a nucleic acid molecule capable of transporting another nucleic acid to which it has been linked. One type of vector is a "plasmid", which refers to a circular double stranded DNA loop into which additional DNA segments can be ligated. Another type of vector is a viral vector, wherein additional DNA segments can be ligated into the viral genome. Certain vectors are capable of autonomous replication in a host cell into which they are introduced (e.g., bacterial vectors having a bacterial origin of replication and episomal mammalian vectors). Other vectors (e.g., non-episomal mammalian vectors) are integrated into the genome of a host cell upon introduction into the host cell, and thereby are replicated along with the host genome. Moreover, certain vectors are capable of directing the expression of genes to which they are operatively-linked. Such vectors are referred to herein as "expression vectors". In general, expression vectors of utility in recombinant DNA
techniques are often in the form of plasmids. In the present specification, "plasmid" and "vector" can be used interchangeably as the plasmid is the most commonly used form of vector. However, the invention is intended to include such other forms of expression vectors, such as viral vectors (e.g., replication defective retroviruses, adenoviruses and adeno-associated viruses), which serve equivalent functions.
The recombinant expression vectors of the invention comprise a nucleic acid of the invention in a form suitable for expression of the nucleic acid in a host cell, which means that the recombinant expression vectors include one or more regulatory sequences, selected on the basis of the host cells to be used for expression, that is operatively-linked to the nucleic acid sequence to be expressed. Within a recombinant expression vector, "operably-linked" is intended to mean that the nucleotide sequence of interest is linked to the regulatory sequences) in a manner that allows for expression of the nucleotide sequence (e.g., in an in vitro transcription/translation system or in a host cell when the vector is introduced into the host cell).

The term "regulatory sequence" is intended to includes promoters, enhancers and other expression control elements (e.g., polyadenylation signals). Such regulatory sequences are described, for example, in Goeddel, GENE EXPRESSION TECHNOLOGY:
METHODS IN ENZYMOLOGY 185, Academic Press, San Diego, Calif. (1990).
Regulatory sequences include those that direct constitutive expression of a nucleotide sequence in many types of host cell and those that direct expression of the nucleotide sequence only in certain host cells (e.g., tissue-specific regulatory sequences). It will be appreciated by those skilled in the art that the design of the expression vector can depend on such factors as the choice of the host cell to be transformed, the level of expression of protein desired, etc. The expression vectors of the invention can be introduced into host cells to thereby produce proteins or peptides, including-fusion proteins or peptides, encoded by nucleic acids as described herein (e.g., NOVX proteins, mutant forms of NOVX proteins, fusion proteins, etc.).
The recombinant expression vectors of the invention can be designed for expression of NOVX proteins in prokaryotic or eukaryotic cells. For example, NOVX
proteins can be expressed in bacterial cells such as Escherichia cola, insect cells (using baculovirus expression vectors) yeast cells or mammalian cells. Suitable host cells are discussed further in Goeddel, GENE EXPRESSION TECHNOLOGY: METHODS IN
ENZYMOLOGY 185, Academic Press, San Diego, Calif. (1990). Alternatively, the recombinant expression vector can be transcribed and translated in vitro, for example using T7 promoter regulatory sequences and T7 polymerase.
Expression of proteins in prokaryotes is most often carried out in Escherichia coli with vectors containing constitutive or inducible promoters directing the expression of either fusion or non-fusion proteins. Fusion vectors add a number of amino acids to a protein encoded therein, usually to the amino terminus of the recombinant protein. Such fusion vectors typically serve three purposes: (i) to increase expression of recombinant protein; (ir'to increase the solubility of the recombinant protein; and (iiy to aid in the purification of the recombinant protein by acting as a ligand in affinity purification.
Often, in fusion expression vectors, a proteolytic cleavage site is introduced at the junction of the fusion moiety and the recombinant protein to enable separation of the recombinant protein from the fusion moiety subsequent to purification of the fusion protein. Such enzymes, and their cognate recognition sequences, include Factor Xa, thrombin and enterokinase. Typical fusion expression vectors include pGEX (Pharmacia Biotech Inc;
Smith and Johnson, 1988. Gene 67: 31-40), pMAL (New England Biolabs, Beverly, Mass.) and pRITS (Pharmacia, Piscataway, N.J.) that fuse glutathione S-transferase (GST), maltose E binding protein, or protein A, respectively, to the target recombinant protein.
Examples of suitable inducible non-fusion E. coli expression vectors include pTrc (Amrann et al., (1988) Gene 69:301-315) and pET l 1d (Studier et al., GENE
E3iPRESSION
TECHNOLOGY: METHODS IN ENZYMOLOGY 185, Academic Press, San Diego, Calif.
(1990) 60-89).
One strategy to maximize recombinant protein expression in E. coli is to express the protein in a host bacteria with an impaired capacity to proteolytically cleave the recombinant protein. See, e.g., Gottesman, GENE EXPRESSION TECHNOLOGY: METHODS
IN
ENZYMOLOGY 185, Academic Press, San Diego, Calif. (1990) 119-128. Another strategy is to alter the nucleic acid sequence of the nucleic acid to be inserted into an expression vector so that the individual codons for each amino acid are those preferentially utilized in E. coli (see, e.g:, Wada, et al., 1992. Nucl. Acids Res. 20: 2111-2118). Such alteration of nucleic acid sequences of the invention can be carried out by standard DNA
synthesis techniques.
In another embodiment, the NOVX expression vector is a yeast expression vector.
Examples of vectors for expression in yeast Saccharomyces cerivisae include pYepSec 1 (Baldari, et al., 1987. EMBO J. 6: 229-234), pMFa (Kurjan and Herskowitz, 1982. Cell 30: 933-943), pJRY88 (Schultz et al., 1987. Gene 54: 113-123), pYES2 (Invitrogen Corporation, San Diego, Calif.), and picZ (InVitrogen Corp, San Diego, Calif.).
Alternatively, NOVX can be expressed in insect cells using baculovirus expression vectors. Baculovirus vectors available for expression of proteins in cultured insect cells (e.g., SF9 cells) include the pAc series (Smith, et al., 1983. Mol. Cell.
Biol. 3: 2156-2165) and the pVL series (Lucklow and Summers, 1989. Virology 170: 31-39).
In yet another embodiment, a nucleic acid of the invention is expressed in mammalian cells using a mammalian expression vector. Examples of mammalian expression vectors include pCDM8 (Seed, 1987. Nature 329: 840) and pMT2P~
(Kaufinan, et al., 1987. EMBO J. 6: 187-195). When used in mammalian cells, the expression vector's control functions are often provided by viral regulatory elements. For example, commonly used promoters are derived from polyoma, adenovirus 2, cytomegalovirus, and simian virus 40. For other suitable expression systems for both prokaryotic and eukaryotic cells see, e.g., Chapters 16 and 17 of Sambrook, et al., MOLECULAR CLONING: A LABORATORY MANUAL. 2nd ed., Cold Spring Harbor Laboratory, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y., 1989.
In another embodiment, the recombinant mammalian expression vector is capable of directing expression of the nucleic acid preferentially in a particular cell Type (e.g., tissue-specific regulatory elements are used to express the nucleic acid).
Tissue-specific regulatory elements are known in the art. Non-limiting examples of suitable tissue-specific promoters include the albumin promoter (liver-specific;
Pinkert, et al., 1987. Genes Dev. 1: 268-277), lymphoid-specific promoters (Calame and Eaton, 1988.
Adv. Immunol. 43: 235-275), in particular promoters of T cell receptors (Winoto and Baltimore, 1989. EMBO J. 8: 729-733) and imrnunoglobulins (Banerji, et al., 1983. Cell 33: 729-740; Queen and Baltimore, 1983. Cell 33: 741-748), neuron-specific promoters (e.g., the neurofilament promoter; Byrne and Ruddle, 1989. Proc. Natl. Acad.
Sci. USA 86:
5473-5477), pancreas-specific prombters (Edlund, et al., 1985. Science 230:
912-916), and mammary gland-specific promoters (e.g., milk whey promoter; U.S. Pat. No.
4,873,316 and European Application Publication No. 264,166). Developmentally-regulated promoters are also encompassed, e.g., the murine hox promoters (Kessel and Gruss, 1990.
Science 249: 374-379) and the a-fetoprotein promoter (Campes and Tilghman, 1989.
Genes Dev. 3: 537-546).
The invention further provides a recombinant expression vector comprising a DNA
molecule of the invention cloned into the expression vector in an antisense orientation.
That is, the DNA molecule is operatively-linked to a regulatory sequence in a manner that allows for expression (by transcription of the DNA molecule) of an RNA
molecule that is antisense to NOVX mRNA. Regulatory sequences operatively linked to a nucleic acid cloned in the antisense orientation can be chosen that direct the continuous expression of the antisense RNA molecule in a variety of cell types, for instance viral promoters and/or enhancers, or regulatory sequences can be chosen that direct constitutive, tissue specific or cell type specific expression of antisense RNA. The antisense expression vector can be in the form of a recombinant plasmid, phagemid or attenuated virus in which antisense i nucleic acids are produced under the control of a high efficiency regulatory region, the activity of which can be determined by the cell type into which the vector is introduced.
For a discussion of the regulation of gene expression using antisense genes see, e.g., Weintraub, et al., "Antisense RNA as a molecular tool for genetic analysis,"
Reviews-Trends in Genetics, Vol. 1(1) 1986.

Another aspect of the invention pertains to host cells into which a recombinant expression vector of the invention has been introduced. The terms "host cell"
and "recombinant host cell" are used interchangeably herein. It is understood that such terms refer not only to the particular subject cell but also to the progeny or potential progeny of such a cell. Because certain modifications may occur in succeeding generations due to either mutation or environmental influences, such progeny may not, in fact, be identical to the parent cell, but are still included within the scope ofthe term as used herein.
A host cell can be any prokaryotic or eukaryotic cell. For example, NOVX
protein can be expressed in bacterial cells such as E. coli, insect cells, yeast or mammalian cells (such as Chinese hamster ovary cells (CHO) or COS cells). Other suitable host cells are known to those skilled in the art.
Vector DNA can be introduced into prokaryotic or eukaryotic cells via conventional transformation or transfection techniques. As used herein, the terms "transformation" and "transfection" are intended to refer to a variety of art-recognized techniques for introducing foreign nucleic acid (e.g., DNA) into a host cell, including calcium phosphate or calcium chloride co-precipitation, DEAE-dextran-mediated transfection, lipofection, or electroporation. Suitable methods for transforming or transfecting host cells can be found in Sambrook, et al. (lVIoLECULAR CLONING:
A
LABORATORY MANUAL. 2nd ed., Cold Spring Harbor Laboratory, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y., I 989), and other laboratory manuals.
For stable transfection of mammalian cells, it is known that, depending upon the expression vector and transfection technique used, only a small fraction of cells may integrate the foreign DNA into their genome. In order to identify and select these integrants, a gene that encodes a selectable marker (e.g., resistance to antibiotics) is generally introduced into the host cells along with the gene of interest.
Various selectable markers include those that confer resistance to drugs, such as 6418, hygromycin and methotrexate. Nucleic acid encoding a selectable marker can be introduced into a host cell on the same vector as that encoding NOVX or can be introduced on a separate vector.
Cells stably transfected with the introduced nucleic acid can be identified by drug selection (e.g., cells that have incorporated the selectable marker gene will survive, while the other cells die).
A host cell of the invention, such as a prokaryotic or eukaryotic host cell in culture, can be used to produce (i.e., express) NOVX protein. Accordingly, the invention further provides methods for producing NOVX protein using the host cells of the invention. In 5?

one embodiment, the method comprises culturing the host cell of invention (into which a recombinant expression vector encoding NOVX protein has been introduced) in a suitable medium such that NOVX protein is produced. In another embodiment, the method further comprises isolating NOVX protein from the medium or the host cell.
Transgenic NOVX Animals The host cells of the invention can also be used to produce non-human transgenic animals. For example, in one embodiment, a host cell of the invention is a fertilized oocyte or an embryonic stem cell into which NOVX protein-coding sequences have been introduced. Such host cells can then be used to create non-human transgenic animals in which exogenous NOVX sequences have been introduced into their genome or homologous recombinant animals in which endogenous NOVX sequences have been altered. Such animals are useful for studying the function and/or activity of NOVX
protein and for identifying and/or evaluating modulators of NOVX protein activity. As used herein, a "transgenic animal" is a non-human animal, preferably a mammal, more preferably a rodent such as a rat or mouse, in which one or more of the cells of the animal includes a transgene. Other examples of transgenic animals include non-human primates, sheep, dogs, cows, goats, chickens, amphibians, etc. A transgene is exogenous DNA that is integrated into the genome of a cell from which a transgenic animal develops and that remains in the genome of the mature animal, thereby directing the expression of an encoded gene product in one or more cell types or tissues of the transgenic animal. As used herein, a "homologous recombinant animal" is a non-human animal, preferably a mammal, more preferably a mouse, in which an endogenous NOVX gene has been altered by homologous recombination between the endogenous gene and an exogenous DNA
molecule introduced into a cell of the animal, e.g., an embryonic cell of the animal, prior to development of the animal.
A transgenic animal of the invention can be created by introducing NOVX-encoding nucleic acid into the male pronuclei of a fertilized oocyte (e.g., by microinjection, retroviral infection) and allowing the oocyte to develop in a pseudopregnant female foster animal. The human NOVX cDNA sequences SEQ ID
NOS:2n-l, wherein n is an integer between l and 54, can be introduced as a transgene into the genome of a non-human animal. Alternatively, a non-human homologue of the human NOVX gene, such as a mouse NOVX gene, can be isolated based on hybridization to the human NOVX cDNA (described further supra) and used as a transgene. Intronic SS

sequences and polyadenylation signals can also be included in the transgene to increase the efficiency of expression of the transgene. A tissue-specific regulatory sequences) can be operably-linked to the NOVX transgene to direct expression of NOVX protein to particular cells. Methods for generating transgenic animals via embryo manipulation and microinjection, particularly animals such as mice, have become conventional in the art and are described, for example, in U.S. Patent Nos. 4,736,866; 4,870,009; and 4,873,191; and Hogan, 1986. In: MANIPULATING THE MousE EMBRYO, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y. Similar methods are used for production of other transgenic animals. A transgenic founder animal can be identified based upon the presence of the NOVX transgene in its genome and/or expression of NOVX mIZNA
in tissues or cells of the animals. A transgenic founder animal can then be used to breed additional animals carrying the transgene. Moreover, transgenic animals carrying a transgene-encoding NOVX protein can further be bred to other transgenic animals carrying other transgenes.
To create a homologous recombinant animal, a vector is prepared which contains at least a portion of A NOVX gene into which a deletion, addition or substitution has been introduced to thereby alter, e.g., functionally disrupt, the NOVX gene. The NOVX gene can be a human gene (e.g., the cDNA of SEQ ID NOS:2n-l, wherein n is an integer between 1 and 54), but more preferably, is a non-human homologue of a human NOVX .
gene. For example, a mouse homologue of human NOVX gene of SEQ ID NOS:2n-1, wherein n is an integer between 1 and 54, can be used to construct a homologous recombination vector suitable for altering an endogenous NOVX gene in the mouse genome. In one embodiment, the vector is designed such that, upon homologous recombination, the endogenous NOVX gene is functionally disrupted (i.e., no longer encodes a functional protein; also referred to as a "knock out" vector).
Alternatively, the vector can be designed such that, upon homologous recombination, the endogenous NOVX gene is mutated or otherwise altered but still encodes functional protein (e.g., the upstream regulatory region can be altered to thereby alter the expression of the endogenous NOVX protein). In the homologous recombination vector, the altered portion of the NOVX gene is flanked at its 5'- and 3'-termini by additional nucleic acid of the NOVX gene to allow for homologous recombination to occur between the exogenous NOVX gene carried by the vector and an endogenous NOVX gene in an embryonic stem cell. The additional flanking NOVX nucleic acid is of sufficient length for successful homologous recombination with the endogenous gene.

Typically, several kilobases of flanking DNA (both at the 5'- and 3'-termini) are included in the vector. See, e.g., Thomas, et al., 1987. Cell 51: 503 for a description of homologous recombination vectors. The vector is ten introduced into an embryonic stem cell line (e.g, by electroporation) and cells in which the introduced NOVX gene has homologously-recombined with the endogenous NOVX gene are selected. See, e.g., Li, et al., 1992. Cell 69: 915.
The selected cells are then injected into a blastocyst of an animal (e.g., a mouse) to form aggregation chimeras. See, e.g., Bradley, 1987. In: TERATOCARCINOMAS AND
EMBRYONIC STEM CELLS: A PRACTICAL APPROACH, Robertson, ed. IRL, Oxford, pp.
113-152. A chimeric embryo can then be implanted into a suitable pseudopregnant female foster animal and the embryo brought to term. Progeny harboring the homologously-recombined DNA in their germ cells can be used to breed animals in which all cells of the animal contain the homologously-recombined DNA by germline transmission of the transgene. Methods for constructing homologous recombination I 5 vectors and homologous recombinant animals are described further in Bradley, 1991.
Curr. ~pin. Biotechhol. 2: 823-829; PCT International Publication Nos.: WO
90/11354;
WO 91/01140; WO 92/0968; and WO 93/04169.
In another embodiment, transgenic non-humans animals can be produced that contain selected systems that allow for regulated expression of the transgene.
One example of such a system is the cre/IoxP recombinase system of bacteriophage Pl. For a description of the cre/IoxP recombinase system, See, e.g., Lakso, et al., 1992. Proc. Natl.
Acad. Sci. USA 89: 6232-6236. Another example of a recombinase system is the FLP
recombinase system of Saccharomyces cerevisiae. See, O'Gorman, et al., 1991.
Science 251:1351-1355. If a cre/IoxP recombinase system is used to regulate expression of the transgene, animals containing transgenes encoding both the Cre recombinase and a selected protein are required. Such animals can be provided through the construction of "double" transgenic animals, e.g., by mating two transgenic animals, one containing a transgene encoding a selected protein and the other containing a transgene encoding a recombinase. .
Clones of the non-human transgenic animals described herein can also be produced according to the methods described in Wilmut, et al., 1997. Nature 385: 810-813. In brief, a cell (e.g., a somatic cell) from the transgenic animal can be isolated and induced to exit the growth cycle and enter Go phase. The quiescent cell can then be fused, e.g., through the use of electrical pulses, to an enucleated oocyte from an animal of the same species from which the quiescent cell is isolated. The reconstructed oocyte is then cultured such that it develops to morula or blastocyte and then transferred to pseudopregnant female foster animal. The offspring borne of this female foster animal will be a clone of the animal from which the cell (e.g., the somatic cell) is isolated.
Pharmaceutical Compositions The NOVX nucleic acid molecules, NOVX proteins, and anti-NOVX antibodies (also referred to herein as "active compounds") of the invention, and derivatives, fragments, analogs and homologs thereof, can be incorporated into pharmaceutical compositions suitable for administration. Such compositions typically comprise the nucleic acid molecule, protein, or antibody and a pharmaceutically acceptable carrier. As used herein, "pharmaceutically acceptable carrier" is intended to include any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like, compatible with pharmaceutical administration.
Suitable carriers are described in the most recent edition of Remington's Pharmaceutical Sciences, a standard reference text in the field, which is incorporated herein by reference.
Preferred examples of such carriers or diluents include, but are not limited to, water, saline, finger's solutions, dextrose solution, and 5% human serum albumin.
Liposomes and non-aqueous vehicles such as fixed oils may also be used. The use of such media and agents for pharmaceutically active substances is well known in the art. Except insofar as any conventional media or agent is incompatible with the active compound, use thereof in the compositions is contemplated. Supplementary active compounds can also be incorporated into the compositions.
A pharmaceutical composition of the invention is formulated to be compatible with its intended route of administration. Examples of routes of administration include parenteral, e.g., intravenous, intradermal, 'subcutaneous, oral (e.g., inhalation), transdermal (i.e., topical), transmucosal, and rectal administration. Solutions or suspensions used for parenteral, intradermal, or subcutaneous application can include the following components: a sterile diluent such as water for injection, saline solution, fixed oils, polyethylene glycols, glycerine, propylene glycol or other synthetic solvents;
antibacterial agents such as benzyl alcohol or methyl parabens; antioxidants such as ascorbic acid or sodium bisulfate; chelating agents such as ethylenediaminetetraacetic acid (EDTA);
buffers such as acetates, citrates or phosphates, and agents for the adjustment of tonicity such as sodium chloride or dextrose. The pH can be adjusted with acids or bases, such as hydrochloric acid or sodium hydroxide. The parenteral preparation can be enclosed in ampoules, disposable syringes or multiple dose vials made of glass or plastic.
Pharmaceutical compositions suitable for injectable use include sterile aqueous solutions (where water soluble) or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersion. For intravenous administration, suitable carriers include physiological saline, bacteriostatic water, Cremophor ELT"
(BASF, Parsippany, N.J.) or phosphate buffered saline (PBS). In all cases, the composition must be sterile and should be fluid to the extent that easy syringeability exists. It must be stable under the conditions of manufacture and storage and must be preserved against the contaminating action of microorganisms such as bacteria and fungi.
The carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, and liquid polyethylene glycol, and the like), and suitable mixtures thereof. The proper fluidity can be maintained, for example, by the use of a coating~such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants. Prevention of the action of microorganisms can be achieved by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, ascorbic acid, thimerosal, and the like. In many cases, it will be preferable to include isotonic agents, for example, sugars, polyalcohols such as manitol, sorbitol, sodium chloride in the composition. Prolonged absorption of the injectable compositions can be brought about by including in the composition an agent which delays absorption, for example, aluminum monostearate and gelatin.
Sterile injectable solutions can be prepared by incorporating the active compound (e.g., A NOVX protein or anti-NOVX antibody) in the required amount in an appropriate solvent with one or a combination of ingredients enumerated above, as required, followed by filtered sterilization. Generally, dispersions are prepared by incorporating the active compound into a sterile vehicle that contains a basic dispersion medium and the required other ingredients from those enumerated above. In the case of sterile powders for the preparation of sterile injectable solutions, methods of preparation are vacuum drying and freeze-drying that yields a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof.
Oral compositions generally include an inert diluent or an edible carrier.
They can be enclosed in gelatin capsules or compressed into tablets. For the purpose of oral therapeutic administration, the active compound can be incorporated with excipients and used in the form of tablets, troches, or capsules. Oral compositions can also be prepared using a fluid carrier for use as a mouthwash, wherein the compound in the fluid carrier is applied orally and swished and expectorated or swallowed. Pharmaceutically compatible binding agents, and/or adjuvant materials can be included as part of the composition. The tablets, pills, capsules, troches and the like can contain any of the following ingredients, or compounds of a similar nature: a binder such as microcrystalline cellulose, gum tragacanth or gelatin; an excipient such as starch or lactose, a disintegrating agent such as alginic acid, Primogel, or corn starch; a lubricant such as magnesium stearate or Sterotes; a glidant such as colloidal silicon dioxide; a sweetening agent such as sucrose or saccharin;
or a flavoring agent such as peppermint, methyl salicylate, or orange flavoring.
For administration by inhalation, the compounds are delivered in the form of an aerosol spray from pressured container or dispenser which contains a suitable propellant, e.g., a gas such as carbon dioxide, or a nebulizer.
Systemic administration can also be by transmucosal or transdermal means. For transmucosal or transdermal administration, penetrants appropriate to the barrier to be permeated are used in the formulation. Such penetrants are generally known in the art, and include, for example, for transmucosal administration, detergents, bile salts, and fusidic acid derivatives. Transmucosal administration can be accomplished through the use of nasal sprays or suppositories. For transdermal administration, the active compounds are formulated into ointments, salves, gels, or creams as generally known in the art.
The compounds can also be prepared in the form of suppositories (e.g., with conventional suppository bases such as cocoa butter and other glycerides) or retention enemas for rectal delivery.
In one embodiment, the active compounds are prepared with carriers that will protect the compound against rapid elimination from the body, such as a controlled release formulation, including implants and microencapsulated delivery systems.
Biodegradable, biocompatible polymers can be used, 'such as ethylene vinyl acetate, polyanhydrides, polyglycolic acid, collagen, polyorthoesters, and polylactic acid. Methods for preparation of such formulations will be apparent to those skilled in the art. The materials can also be obtained commercially from Alza Corporation and Nova Pharmaceuticals, Inc.
Liposomal suspensions (including liposomes targeted to infected cells with monoclonal antibodies to viral antigens) can also be used as pharmaceutically acceptable carriers.
These can be prepared according to methods known to those skilled in the art, for example, as described in U.S. Patent No. 4,522,811.
It is especially advantageous to formulate oral or parenteral compositions in dosage unit form for ease of administration and uniformity of dosage. Dosage unit form as used herein refers to physically discrete units suited as unitary dosages for the subject to be treated; each unit containing a predetermined quantity of active compound calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier. The specification for the dosage unit forms of the invention are dictated by and directly dependent on the unique characteristics of the active compound and the particular therapeutic effect to be achieved, and the limitations inherent in the art of compounding such an active compound for the treatment of individuals.
The nucleic acid molecules of the invention can be inserted into vectors and used as gene therapy vectors. Gene therapy vectors can be delivered to a subject by, for example, intravenous injection, local administration (see, e.g., U.S. Patent No. 5,328,470) or by stereotactic injection (see, e.g., Chen, et al., 1994. Proc. Natl. Acad.
Sei. USA 91:
3054-3057). The pharmaceutical preparation of the gene therapy vector can include the gene therapy vector in an acceptable diluent, or can comprise a slow release matrix in which the gene delivery vehicle is imbedded. Alternatively, where the complete gene delivery vector can be produced intact from recombinant cells, e.g., retroviral vectors, the pharmaceutical preparation can include one or more cells that produce the gene delivery system.
The pharmaceutical compositions can be included in a container, pack, or dispenser together with instructions for administration.
Screening and Detection Methods The isolated nucleic acid molecules of the invention can be used to express NOVX
protein (e.g., via a recombinant expression vector in a host cell in gene therapy applications), to detect NOVX mRNA (e.g., in a biological sample) or a genetic lesion in A NOVX gene, and to modulate NOVX activity, as described further, below. In addition, the NOVX proteins can be used to screen drugs or compounds that modulate the NOVX
protein activity or expression as well as to treat disorders characterized by insufficient or excessive production of NOVX protein or production of NOVX protein forms that have decreased or aberrant activity compared to NOVX wild-type protein (e.g.;
diabetes (regulates insulin release); obesity (binds and transport lipids); metabolic disturbances associated with obesity, the metabolic syndrome X as well as anorexia and wasting disorders associated with chronic diseases and various cancers, and infectious disease(possesses anti-microbial activity) and the various dyslipidemias. In addition, the anti-NOVX antibodies of the invention can be used to detect and isolate NOVX
proteins and modulate NOVX activity. In yet a further aspect, the invention can be used in methods to influence appetite, absorption of nutrients and the disposition of metabolic substrates in both a positive and negative fashion.
The invention further pertains to novel agents identified by the screening assays described herein and uses thereof for treatments as described, supra.
Screening Assays The invention provides a method (also referred to herein as a "screening assay") for identifying modulators, i.e., candidate or test compounds or agents (e.g., peptides, peptidomimetics, small molecules or other drugs) that bind to NOVX proteins or have a stimulatory or inhibitory effect on, e.g., NOVX protein expression or NOVX
protein activity. The invention also includes compounds identified in the screening assays described herein.
In one embodiment, the invention provides assays for screening candidate or test compounds which bind to or modulate the activity of the membrane-bound form of A
NOVX protein or polypeptide or biologically-active portion thereof. The test compounds of the invention can be obtained using any of the numerous approaches in combinatorial library methods known in the art, including: biological libraries; spatially addressable parallel solid phase or solution phase libraries; synthetic library methods requiring deconvolution; the "one-bead one-compound" library method; and synthetic library methods using affinity chromatography selection. The biological library approach is limited to peptide libraries, while the other four approaches are applicable to peptide, non-peptide oligomer or small molecule libraries of compounds. See, e.g., Lam, 1997.
Anticancer Drug Design 12: 145.
A "small molecule" as used herein, is meant to refer to a composition that has a molecular weight of less than about 5 kD and most preferably less than about 4 kD. Small molecules can be, e.g., nucleic acids, peptides, polypeptides, peptidomimetics, carbohydrates, lipids or other organic or inorganic molecules. Libraries of chemical and/or biological mixtures, such as fungal, bacterial, or algal extracts, are known in the art and can be screened with any of the assays of the invention.

Examples of methods for the synthesis of molecular libraries can be found in the art, for example in: DeWitt, et al., 1993. Proc. Natl. Acad. Sci. U.S.A. 90:
6909; Erb, et al., 1994. Proc. Natl. Acad. Sci. U.S.A. 91: 11422; Zuckermann, et al., 1994. J.
Med. Chem.
37: 2678; Cho, et al., 1993. Science 261: 1303; Carrell, et al., 1994. Angew.
Chem. Int. Ed.
Engl. 33: 2059; Carell, et al., 1994. Angew. Chem. Int. Ed. Engl. 33: 2061;
and Gallop, et al., 1994. J. Med. Chem. 37: 1233.
Libraries of compounds may be presented in solution (e.g., Houghten, 1992.
Biotechniques 13: 412-421), or on beads (Lam, 1991. Nature 354: 82-84), on chips (Fodor, 1993. Nature 364: 555-556), bacteria (Ladner, U.S. Patent No.
5,223,409), spores (Ladner, U.S. Patent 5,233,409), plasmids (Cull, et al., 1992. Proc. Natl.
Acad. Sci. USA
89: 1865-1869) or on phage (Scott and Smith, 1990. Science 249: 386-390;
Devlin, 1990.
Science 249: 404-406; Cwirla, et al., 1990. Proc. Natl. Acad. Sci. U.S.A. 87:
6378-6382;
Felici, 1991. J. Mol. Biol. 222: 301-310; Ladner, U.S. Patent No. 5,233,409.).
In one embodiment, an assay is a cell-based assay in which a cell which expresses a membrane-bound form of NOVX protein, or a biologically-active portion thereof, on the cell surface is contacted with a test compound and the ability of the test compound to bind to A NOVX protein determined. The cell, for example, can of mammalian origin or a yeast cell. Determining the ability of the test compound to bind to the NOVX
protein can be accomplished, for example, by coupling the test compound with a radioisotope or enzymatic label such that binding of the test compound to the NOVX protein or biologically-active portion thereof can be determined by detecting the labeled compound in a complex. For example, test compounds can be labeled with ~aSI, 35S,14C, or 3H, either directly or indirectly, and the radioisotope detected by direct counting of radioemission or by scintillation counting. Alternatively, test compounds can be enzymatically-labeled with, for example, horseradish peroxidase, alkaline phosphatase, or luciferase, and the enzymatic label detected by determination of conversion of an appropriate substrate to product. In one embodiment, the assay comprises contacting a cell which expresses a membrane-bound form of NOVX protein, or a biologically-active portion thereof, on the cell surface with a known compound which binds NOVX to form an assay mixture, contacting the assay mixture with a test compound, and determining the ability of the test compound to interact with A NOVX protein, wherein determining the ability of the test compound to interact with A NOVX protein comprises determining the ability of the test compound to preferentially bind to NOVX protein or a biologically-active portion thereof as compared to the known compound.

In another embodiment, an assay is a cell-based assay comprising contacting a cell expressing a membrane-bound form of NOVX protein, or a biologically-active portion thereof, on the cell surface with a test compound and determining the ability of the test compound to modulate (e.g., stimulate or inhibit) the activity of the NOVX
protein or biologically-active portion thereof. Determining the ability of the test compound to modulate the activity of NOVX or a biologically-active portion thereof can be accomplished, for example, by determining the ability of the NOVX protein to bind to or interact with A NOVX target molecule. As used herein, a "target molecule" is a molecule with which A NOVX protein binds or interacts in nature, for example, a molecule on the surface of a cell which expresses A NOVX interacting protein, a molecule on the surface of a second cell, a molecule in the extracellular milieu, a molecule associated with the internal surface of a cell membrane or a cytoplasmic molecule. A NOVX target molecule can be a non-NOVX molecule or A NOVX protein or polypeptide of the invention.
In one embodiment, A NOVX target molecule is a component of a signal transduction pathway that facilitates transduction of an extracellular signal (e.g. a signal generated by binding of a compound to a membrane-bound NOVX molecule) through the cell membrane and into the cell. The target, for example, can be a second intercellular protein that has catalytic activity or a protein that facilitates the association of downstream signaling molecules with NOVX.
Determining the ability of the NOVX protein to bind to or interact with A NOVX
target molecule can be accomplished by one of the methods described above for determining direct binding. In one embodiment, determining the ability of the NOVX
protein to bind to or interact with A NOVX target molecule can be accomplished by determining the activity of the target molecule. For example, the activity of the target molecule can be determined by detecting induction of a cellular second messenger of the target (i.e. intracellular Ca2+, diacylglycerol, IP3, etc.), detecting catalytic/enzymatic activity of the target an appropriate substrate, detecting the induction of a reporter gene (comprising A NOVX-responsive regulatory element operatively linked to a nucleic acid encoding a detectable marker, e.g., luciferase), or detecting a cellular response, for example, cell survival, cellular differentiation, or cell proliferation.
In yet another embodiment, an assay of the invention is a cell-free assay comprising contacting A NOVX protein or biologically-active portion thereof with a test compound and determining the ability of the test compound to bind to the NOVX
protein or biologically-active portion thereof. Binding of the test compound to the NOVX protein can be determined either directly or indirectly as described above. In one such embodiment, the assay comprises contacting the NOVX protein or biologically-active portion thereof with a known compound which binds NOVX to form an assay mixture, contacting the assay mixture with a test compound, and determining the ability of the test compound to interact with A NOVX protein, wherein determining the ability of the test compound to interact with A NOVX protein comprises determining the ability of the test compound to preferentially bind to NOVX or biologically-active portion thereof as compared to the known compound.
In still another embodiment, an assay is a cell-free assay comprising contacting NOVX protein or biologically-active portion thereof with a test compound and determining the ability of the test compound to modulate (e.g. stimulate or inhibit) the activity of the NOVX protein or biologically-active portion thereof.
Determining the ability of the test compound to modulate the activity of NOVX can be accomplished, for example, by determining the ability of the NOVX protein to bind to A NOVX
target molecule by one of the methods described above for determining direct binding.
In an alternative embodiment, determining the ability of the test compound to modulate the activity of NOVX protein can be accomplished by determining the ability of the NOVX
protein further modulate A NOVX target molecule. For example, the catalytic/enzymatic activity of the target molecule on an appropriate substrate can be determined as described, supra.
In yet another embodiment, the cell-free assay comprises contacting the NOVX
protein or biologically-active portion thereof with a known compound which binds NOVX
protein to form an assay mixture, contacting the assay mixture with a test compound, and determining the ability ofthe test,compound to interact with A NOVX protein, wherein determining the ability of the test compound to interact with A NOVX protein comprises determining the ability of the NOVX protein to preferentially bind to or modulate the activity of A NOVX target molecule.
The cell-free assays of the invention are amenable to use of both the soluble form or the membrane-bound form of NOVX protein. In the case of cell-free assays comprising the membrane-bound form of NOVX protein, it may be desirable to utilize a solubilizing agent such that the membrane-bound form of NOVX protein is maintained in solution.
Examples of such solubilizing agents include non-ionic detergents such as n-octylglucoside, n-dodecylglucoside, n-dodecylmaltoside, octanoyl-N-methylglucamide, decanoyl-N-methylglucamide, Triton~ X-100, Triton~ X-114, Thesit~, Isotridecypoly(ethylene glycol ether)", N-dodecyl--N,N-dimethyl-3-ammonio-1-propane sulfonate, 3-(3-cholamidopropyl) dimethylamminiol-1-propane sulfonate (CHAPS), or 3-(3-cholamidopropyl)dimethylamminiol-2-hydroxy-1-propane sulfonate (CHAPSO).
In more than one embodiment of the above assay methods of the invention, it may be desirable to immobilize either NOVX protein or its target molecule to facilitate separation of complexed from uncomplexed forms of one or both of the proteins, as well as to accommodate automation of the assay. Binding of a test compound to NOVX
protein, or interaction of NOVX protein with a target molecule in the presence and absence of a candidate compound, can be accomplished in any vessel suitable for containing the reactants. Examples of such vessels include microtiter plates, test tubes, and micro-centrifuge tubes. In one embodiment, a fusion protein can be provided that adds a domain that allows one or both of the proteins to be bound to a matrix.
For example, GST-NOVX fusion proteins or GST-target fusion proteins can be adsorbed onto glutathione sepharose beads (Sigma Chemical, St. Louis, MO) or glutathione derivatized microtiter plates, that are then combined with the test compound or the test compound and either the non-adsorbed target protein or NOVX protein, and the mixture is incubated under conditions conducive to complex formation (e.g., at physiological conditions for salt and pH). Following incubation, the beads or microtiter plate wells are washed to remove any unbound components, the matrix immobilized in the case of beads, complex determined either directly or indirectly, for example, as described, supra.
Alternatively, the complexes can be dissociated from the matrix, and the level ofNOVX protein binding or activity determined using standard techniques.
Other techniques for immobilizing proteins on matrices can also be used in the screening assays of the invention. For example, either the NOVX protein or its target molecule can be immobilized utilizing conjugation of biotin and streptavidin.
Biotinylated NOVX protein or target molecules can be prepared from biotin-NHS
(N-hydroxy-succinimide) using techniques well-known within the art (e.g., biotinylation kit, Pierce Chemicals, Rockford, Ill.), and immobilized in the wells of streptavidin-coated 96 well plates (Pierce Chemical). Alternatively, antibodies reactive with NOVX
protein or target molecules, but which do not interfere with binding of the NOVX
protein to its target molecule, can be derivatized to the wells of the plate, and unbound target or NOVX
protein trapped in the wells by antibody conjugation. Methods for detecting such complexes, in addition to those described above for the GST-immobilized complexes, include immunodetection of complexes using antibodies reactive with the NOVX
protein or target molecule, as well as enzyme-linked assays that rely on detecting an enzymatic activity associated with the NOVX protein or target molecule.
In another embodiment, modulators of NOVX protein expression are identified in a method wherein a cell is contacted with a candidate compound and the expression of NOVX mRNA or protein in the cell is determined. The level of expression of NOVX
mRNA or protein in the presence of the candidate compound is compared to the level of expression of NOVX mRNA or protein in the absence of the candidate compound.
The candidate compound can then be identified as a modulator of NOVX mRNA or protein expression based upon this comparison. For example, when expression of NOVX
mRNA
or protein is greater (i.e., statistically significantly greater) in the presence of the candidate compound than in its absence, the candidate compound is identified as a stimulator of NOVX mRNA or protein expression. Alternatively, when expression of NOVX mRNA
or protein is less (statistically significantly less) in the presence of the candidate compound than in its absence, the candidate compound is identified as an inhibitor of NOVX mRNA
or protein expression. The level of NOVX mRNA or protein expression in the cells can be determined by methods described herein for detecting NOVX mRNA or protein.
In yet another aspect of the invention, the NOVX proteins can be used as "bait proteins" in a two-hybrid assay or three hybrid assay (see, e.g., U.S. Patent No. 5,283,317;
Zervos, et al., 1993. Cell 72: 223-232; Madura, et al., 1993. J. Biol. Chem.
268:
12046-12054; Bartel, et al., 1993. Biotechniques 14: 920-924; Iwabuchi, et al., 1993.
Oncogene 8: 1693-1696; and Brent WO 94/10300), to identify other proteins that bind to or interact with NOVX ("NOVX-binding proteins" or "NOVX-by") and modulate NOVX
activity. Such NOVX-binding proteins are also likely to be involved in the propagation of signals by the NOVX proteins as, for example, upstream or downstream elements of the NOVX pathway.
The two-hybrid system is based on the modular nature of most transcription factors, which consist of separable DNA-binding and activation domains.
Briefly, the assay utilizes two different DNA constructs. In one construct, the gene that codes for NOVX is fused to a gene encoding the DNA binding domain of a known transcription factor (e.g., GAL-4). In the other construct, a DNA sequence, from a library of DNA
sequences, that encodes an unidentified protein ("prey" or "sample") is fused to a gene that codes for the activation domain of the known transcription factor. If the "bait" and the "prey" proteins are able to interact, in vivo, forming A NOVX-dependent complex, the DNA-binding and activation domains of the transcription factor are brought into close proximity. This proximity allows transcription of a reporter gene (e.g., LacZ) that is operably linked to a transcriptional regulatory site responsive to the transcription factor.
Expression of the reporter gene can be detected and cell colonies containing the functional transcription factor can be isolated and used to obtain the cloned gene that encodes the protein which interacts with NOVX.
The invention further pertains to novel agents identified by the aforementioned screening assays and uses thereof for treatments as described herein.
Detection Assays Portions or fragments of the cDNA sequences identified herein (and the corresponding complete gene sequences) can be used in numerous ways as polynucleotide reagents. By way of example, and not of limitation, these sequences can be used to: (i~
map their respective genes on a chromosome; and, thus, locate gene regions associated with genetic disease; (ii) identify an individual from a minute biological sample (tissue I S typing); and (iiy aid in forensic identification of a biological sample.
Some of these applications are described in the subsections, below.
Chromosome Mapping Once the sequence (or a portion of the sequence) of a gene has been isolated, this sequence can be used to map the location of the gene on a chromosome. This process is called chromosome mapping. Accordingly, portions or fragments of the NOVX
sequences, SEQ ID NOS:2n-1, wherein n is an integer between 1 and 54, or fragments or derivatives thereof, can be used to map the location of the NOVX genes, respectively, on a chromosome. The mapping of the NOVX sequences to chromosomes is an important first step in correlating these sequences with genes associated with disease.
Briefly, NOVX genes can be mapped to chromosomes by preparing PCR primers (preferably 15-25 by in length) from the NOVX sequences. Computer analysis of the NOVX, sequences can be used to rapidly select primers that do not span more than one exon in the genomic DNA, thus complicating the amplification process. These primers can then be used for PCR screening of somatic cell hybrids containing individual human chromosomes. Only those hybrids containing the human gene corresponding to the NOVX sequences will yield an amplified fragment.
Somatic cell hybrids are prepared by fusing somatic cells from different mammals (e.g., human and mouse cells). As hybrids of human and mouse cells grow and divide, they gradually lose human chromosomes in random order, but retain the mouse chromosomes. By using media in which mouse cells cannot grow, because they lack a particular enzyme, but in which human cells can, the one human chromosome that contains the gene encoding the needed enzyme will be retained. By using various media, panels of hybrid cell lines can be established. Each cell line in a panel contains either a single human chromosome or a small number of human chromosomes, and a full set of mouse chromosomes, allowing easy mapping of individual genes to specific human chromosomes. See, e.g., D'Eustachio, et al., 1983. Science 220: 919-924.
Somatic cell hybrids containing only fragments of human chromosomes can also be produced by using human chromosomes with translocations and deletions.
PCR mapping of somatic cell hybrids is a rapid procedure for assigning a particular sequence to a particular chromosome. Three or more sequences can be assigned per day using a single thermal cycler. Using the NOVX sequences to design oligonucleotide primers, sub-localization can be achieved with panels of fragments from specific chromosomes.
Fluorescence in situ hybridization (FISH) of a DNA sequence to a metaphase chromosomal spread can further be used to provide a precise chromosomal location in one step. Chromosome spreads can be made using cells whose division has been blocked in metaphase by a chemical like colcemid that disrupts the mitotic spindle. The chromosomes can be treated briefly with trypsin, and then stained with Giemsa.
A pattern of light and dark bands develops on each chromosome, so that the chromosomes can be identified individually. The FISH technique can be used with a DNA sequence as short as 500 or 600 bases. However, clones larger than 1,000 bases have a higher likelihood of binding to a unique chromosomal location with sufficient signal intensity for simple detection. Preferably 1,000 bases, and more preferably 2,000 bases, will suffice to get good results at a reasonable amount of time. For a review of this technique, see, Verma, et al., HUMAN CHROMOSOMES: A MANUAL OF BASIC TECHNIQUES (Pergamon Press, New York 1988).
Reagents for chromosome mapping can be used individually to mark a single chromosome or a single site on that chromosome, or panels of reagents can be used for marking multiple sites and/or multiple chromosomes. Reagents corresponding to noncoding regions of the genes actually are preferred for mapping purposes.
Coding sequences are more likely to be conserved within gene families, thus increasing the chance of cross hybridizations during chromosomal mapping.

Once a sequence has been mapped to a precise chromosomal location, the physical position of the sequence on the chromosome can be correlated with genetic map data.
Such data are found, e.g., in McKusick, MENDELIAN INHERITANCE IN MAN, available on-line through Johns Hopkins University Welch Medical Library). The relationship between genes and disease, mapped to the same chromosomal region, can then be identified through linkage analysis (co-inheritance of physically adjacent genes), described in, e.g., Egeland, et al., 1987. Nature, 325: 783-787.
Moreover, differences in the DNA sequences between individuals affected and unaffected with a disease associated with the NOVX gene, can be determined. If a mutation is observed in some or all of the affected individuals but not in any unaffected individuals, then the mutation is likely to be the causative agent of the particular disease.
Comparison of affected and unaffected individuals generally involves first looking for structural alterations in the chromosomes, such as deletions or translocations that are visible from chromosome spreads or detectable using PCR based on that DNA
sequence.
Ultimately, complete sequencing of genes from several individuals can be performed to confirm the presence of a mutation and to distinguish mutations from polymorphisms.
Tissue Typing The NOVX sequences of the invention can also be used to identify individuals from minute biological samples. 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 identification. The sequences of the invention are useful as additional DNA markers for RFLP ("restriction fragment length polymorphisins," described in U.S.
Patent No. 5,272,057).
Furthermore, the sequences of the invention can be used to provide an alternative technique that determines the actual base-by-base DNA sequence of selected portions of an individual's genome. Thus, the NOVX sequences described herein can be used to prepare two PCR primers from the S'- and 3'-termini of the sequences. These primers can then be used to amplify an individual's DNA and subsequently sequence it.
Panels of corresponding DNA sequences from individuals, prepared in this manner, can provide unique individual identifications, as each individual will have a unique set of such DNA sequences due to allelic differences. The sequences of the invention can be used to obtain such identification sequences from individuals and from tissue. The NOVX sequences of the invention uniquely represent portions of the human genome. Allelic variation occurs to some degree in the coding regions of these sequences, and to a greater degree in the noncoding regions. It is estimated that allelic variation between individual humans occurs with a frequency of about once per each 500 bases.
Much of the allelic variation is due to single nucleotide polymorphisms (SNPs), which include restriction fragment length polymorphisms (RFLPs).
Each of the sequences described herein can, to some degree, be used as a standard against which DNA from an individual can be compared for identification purposes.
Because greater numbers of polymorphisms occur in the noncoding regions, fewer sequences are necessary to differentiate individuals. The noncoding sequences can comfortably provide positive individual identification with a panel of perhaps 10 to 1,000 primers that each yield a noncoding amplified sequence of 100 bases. If predicted coding sequences, such as those in SEQ ID NOS:2n-1, wherein n is an integer between 1 and 54, are used, amore appropriate number of primers for positive individual identification would be S00-2,000.
Predictive Medicine The invention also pertains to the field of predictive medicine in which diagnostic assays, prognostic assays, pharmacogenomics, and monitoring clinical trials are used for prognostic (predictive) purposes to thereby treat an individual prophylactically.
Accordingly, one aspect of the invention relates to diagnostic assays for determining NOVX protein and/or nucleic acid expression as well~as NOVX activity, in the context of a biological sample (e.g., blood, serum, cells, tissue) to thereby determine whether an individual is afflicted with a disease or disorder, or is at risk of developing a disorder, associated with aberrant NOVX expression or activity. The disorders include metabolic disorders, diabetes, obesity, infectious disease, anorexia, cancer-associated cachexia, cancer, neurodegenerative disorders, Alzheimer's Disease, Parkinson's Disorder, immune disorders, and hematopoietic disorders, and the various dyslipidemias, metabolic disturbances associated with obesity, the metabolic syndrome X and wasting disorders associated with chronic diseases and various cancers. The invention also provides for prognostic (or predictive) assays for determining whether an individual is at risk of developing a disorder associated with NOVX protein, nucleic acid expression or activity.
For example, mutations in A NOVX gene can be assayed in a biological sample.
Such assays can be used for prognostic or predictive purpose to thereby prophylactically treat an individual prior to the onset of a disorder characterized by or associated with NOVX
protein, nucleic acid expression, or biological activity.
Another aspect of the invention provides methods for determining NOVX protein, nucleic acid expression or activity in an individual to thereby select appropriate therapeutic or prophylactic agents for that individual (referred to herein as "pharmacogenomics"). Pharmacogenomics allows for the selection of agents (e.g., drugs) for therapeutic or prophylactic treatment of an individual based on the genotype of the individual (e.g., the genotype of the individual examined to determine the ability of the individual to respond to a particular agent.) Yet another aspect of the invention pertains to monitoring the influence of agents (e.g., drugs, compounds) on the expression or activity of NOVX in clinical trials.
These and other agents are described in further detail in the following sections.
DIAGNOSTIC ASSAYS
An exemplary method for detecting the presence or absence of NOVX in a biological sample involves obtaining a biological sample from a test subject and contacting the biological sample with a compound or an agent capable of detecting NOVX
protein or nucleic acid (e.g., mRNA, genomic DNA) that encodes NOVX protein such that the presence of NOVX is detected in the biological sample. An agent for detecting NOVX
mRNA or genomic DNA is a labeled nucleic acid probe capable of hybridizing to NOVX
mIRNA or genomic DNA. The nucleic acid probe can be, for example, a full-length NOVX nucleic acid, such as the nucleic acid of SEQ ID NOS:2n-1, wherein n is an integer between 1 and 54, or a portion thereof, such as an oligonucleotide of at least 15, 30, 50, 100, 250 or 500 nucleotides in length and sufficient to specifically hybridize under stringent conditions to NOVX mRNA or genomic DNA. Other suitable probes for use in the diagnostic assays of the invention are described herein.
An agent for detecting NOVX protein is an antibody capable of binding to NOVX
protein, preferably an antibody with a detectable label. Antibodies can be polyclonal, or more preferably, monoclonal. An intact antibody, or a fragment thereof (e.g., Fab or F(ab')2) can be used. The term "labeled", with regard to the probe or antibody, is intended to encompass direct labeling of the probe or antibody by coupling (i.e., physically linking) a detectable substance to the probe or antibody, as well as indirect labeling of the probe or antibody by reactivity with another reagent that is directly labeled. Examples of indirect labeling include detection of a primary antibody using a fluorescently-labeled secondary antibody and end-labeling of a DNA probe with biotin such that it can be detected with fluorescently-labeled streptavidin. The term "biological sample" is intended to include tissues, cells and biological fluids isolated from a subject, as well as tissues, cells and fluids present within a subject. That is, the detection method of the invention can be used to detect NOVX mRNA, protein, or genomic DNA in a biological sample in vitro as well as in vivo. For example, in vitro techniques for detection of NOVX mRNA
include Northern hybridizations and in situ hybridizations. In vitro techniques for detection of NOVX protein include enzyme linked immunosorbent assays (ELISAs), Western blots, immunoprecipitations, and immunofluorescence. In vitro techniques for detection of NOVX genomic DNA include Southern hybridizations. Furthermore, in vivo techniques for detection of NOVX protein include introducing into a subject a labeled anti-NOVX
antibody. For example, the antibody can be labeled with a radioactive marker whose presence and location in a subject can be detected by standard imaging techniques.
In one embodiment, the biological sample contains protein molecules from the test subject. Alternatively, the biological sample can contain mRNA molecules from the test subject or genomic DNA molecules from the test subject. A preferred biological sample is a peripheral blood leukocyte sample isolated by conventional means from a subject.
In another embodiment, the methods further involve obtaining a control biological sample from a control subject, contacting the control sample with a compound or agent capable of detecting NOVX protein, mRNA, or genomic DNA, such that the presence of NOVX protein, mRNA or genomic DNA is detected in the biological sample, and comparing the presence of NOVX protein, rinRNA or genomic DNA in the control sample with the presence of NOVX protein, mRNA or genomic DNA in the test sample.
The invention also encompasses kits for detecting the presence of NOVX in a biological sample. For example, the kit can comprise: a labeled compound or agent capable of detecting NOVX protein or mRNA in a biological sample; means for determining the amount of NOVX in the sample; and means for comparing the amount of NOVX in the sample with a standard. The compound or agent can be packaged in a suitable container. The kit can further comprise instructions for using the kit to detect NOVX protein or nucleic acid.
PROGNOSTIC ASSAYS
The diagnostic methods described herein can furthermore be utilized to identify subjects having or at risk of developing a disease or disorder associated with aberrant NOVX expression or activity. For example, the assays described herein, such as the preceding diagnostic assays or the following assays, can be utilized to identify a subject having or at risk of developing a disorder associated with NOVX protein, nucleic acid expression or activity. Alternatively, the prognostic assays can be utilized to identify a subject having or at risk for developing a disease or disorder. Thus, the invention provides a method for identifying a disease or disorder associated with aberrant NOVX
expression or activity in which a test sample is obtained from a subject and NOVX protein or nucleic acid (e.g., mRNA, genomic DNA) is detected, wherein the presence of NOVX
protein or nucleic acid is diagnostic for a subject having or at risk of developing a disease or disorder associated with aberrant NOVX expression or activity. As used herein, a "test sample"
refers to a biological sample obtained from a subject of interest. For example, a test sample can be a biological fluid (e.g., serum), cell sample, or tissue.
Furthermore, the prognostic assays described herein can be used to determine whether a subject can be administered an agent (e.g., an agonist, antagonist, peptidomimetic, protein, peptide, nucleic acid, small molecule, or other drug candidate) to treat a disease or disorder associated with aberrant NOVX expression or activity. For example, such methods can be used to determine whether a subject can be effectively treated with an agent for a disorder. Thus, the invention provides methods for determining whether a subject can be effectively treated with an agent for a disorder associated with aberrant NOVX expression or activity in which a test sample is obtained and NOVX
protein or nucleic acid is detected (e.g., wherein the presence of NOVX
protein or nucleic acid is diagnostic for a subject that can be administered the agent to treat a disorder associated with aberrant NOVX expression or activity).
The methods of the invention can also be used to detect genetic lesions in A
NOVX gene, thereby determining if a subject with the lesioned gene is at risk for a disorder characterized by aberrant cell proliferation and/or differentiation.
In various embodiments, the methods include detecting, in a sample of cells from the subject, the presence or absence of a genetic lesion characterized by at least one of an alteration affecting the integrity of a gene encoding A NOVX-protein, or the misexpression of the NOVX gene. For example, such genetic lesions can be detected by ascertaining the existence of at least one of (r~ a deletion of one or more nucleotides from A
NOVX gene;
(ii) an addition of one or more nucleotides to A NOVX gene; (iir~ a substitution of one or more nucleotides of A NOVX gene, (iv) a chromosomal rearrangement of A NOVX
gene;
(v) an alteration in the level of a messenger RNA transcript of A NOVX gene, (vr) aberrant modification of A NOVX gene, such as of the methylation pattern of the genomic DNA, (viy the presence of a non-wild-type splicing pattern of a messenger RNA
transcript of A
NOVX gene, (viiT~ a non-wild-type level of A NOVX protein, (ix) allelic loss of A NOVX
gene, and (x) inappropriate post-translational modification of A NOVX protein.
As described herein, there are a large number of assay techniques known in the art which can be used for detecting lesions in A NOVX gene. A preferred biological sample is a peripheral blood leukocyte sample isolated by conventional means from a subject.
However, any biological sample containing nucleated cells may be used, including, for example, buccal mucosal cells.
In certain embodiments, detection of the lesion involves the use of a probe/primer in a polymerase chain reaction (PCR) (see, e.g., U.S. Patent Nos. 4,683,195 and 4,683,202), such as anchor PCR or RACE PCR, or, alternatively, in a ligation chain reaction (LCR) (see, e.g., Landegran, et al., 1988. Science 241: 1077-1080;
and Nakazawa, et al., 1994. Proc. Natl. Acad. Sci. USA 91: 360-364), the latter of which can be particularly useful for detecting point mutations in the NOVX-gene (see, Abravaya, et al., 1995. Nuel. Acids Res. 23: 675-682). This method can include the steps of collecting a sample of cells from a patient, isolating nucleic acid (e.g., genomic, mRNA or both) from the cells of the sample, contacting the nucleic acid sample with one or more primers that specifically hybridize to A NOVX gene under conditions such that hybridization and amplification of the NOVX gene (if present) occurs, and detecting the presence or absence of an amplification product, or detecting the size of the amplification product and comparing the length to a control sample. It is anticipated that PCR and/or LCR may be desirable to use as a preliminary amplification step in conjunction with any of the techniques used for detecting mutations described herein.
Alternative amplification methods include: self sustained sequence replication (see, Guatelli, et al., 1990. Proc. Natl. Acad. Sci. USA 87: 1874-1878), transcriptional amplification system (see, Kwoh, et al., 1989. Pr~c. Natl. Acad. Sci. USA 86:
1173-1177);
Q/3 Replicase (see, Lizardi, et al, 1988. BioTechreology 6: 1197), or any other nucleic acid amplification method, followed by the detection of the amplified molecules using techniques well known to those of skill in the art. These detection schemes are especially useful for the detection of nucleic acid molecules if such molecules are present in very low numbers.

In an alternative embodiment, mutations in A NOVX gene from a sample cell can be identified by alterations in restriction enzyme cleavage patterns. For example, sample and control DNA is isolated, amplified (optionally), digested with one or more restriction endonucleases, and fragment length sizes are determined by gel electrophoresis and compared. Differences in fragment length sizes between sample and control DNA
indicates mutations in the sample DNA. Moreover, the use of sequence specific ribozymes (see, e.g., U.S. Patent No. 5,493,531) can be used to score for the presence of specific mutations by development or loss of a ribozyme cleavage site.
In other embodiments, genetic mutations in NOVX can be identified by hybridizing a sample and control nucleic acids, e.g., DNA or RNA, to high-density arrays containing hundreds or thousands of oligonucleotides probes. See, e.g., Cronin, et al., 1996. Human Mutation 7: 244-255; Kozal, et al., 1996. Nat. Med. 2: 753-759.
For example, genetic mutations in NOVX can be identified in two dimensional arrays containing light-generated DNA probes as described in Cronin, et al., supra.
Briefly, a first hybridization array of probes can be used to scan through long stretches of DNA in a sample and control to identify base changes between the sequences by making linear arrays of sequential overlapping probes. This step allows the identification of point mutations. This is followed by a second hybridization array that allows the characterization of specific mutations by using smaller, specialized probe arrays complementary to all variants or mutations detected. Each mutation array is composed of parallel probe sets, one complementary to the wild-type gene and the other complementary to the mutant gene.
In yet another embodiment, any of a variety of sequencing reactions known in the art can be used to directly sequence the NOVX gene and detect mutations by comparing the sequence of the sample NOVX with the corresponding wild-type (control) sequence.
Examples of sequencing reactions include those based on techniques developed by Maxim and Gilbert, 1977. Proc. Natl. Acad. Sci. USA 74: 560 or Sanger, 1977. Proc.
Natl. Acad.
Sci. USA 74: 5463. It is also contemplated that any of a variety of automated sequencing procedures can be utilized when performing the diagnostic assays (see, e.g., Naeve, et al., 1995. Biotechniques 19: 448), including sequencing by mass spectrometry (see, e.g., PCT
International Publication No. WO 94/16101; Cohen, et al., 1996. Adv.
Chromatography 36: 127-162; and Griffin, et al., 1993. Appl. Biochem. Biotechnol. 38: 147-159).
Other methods for detecting mutations in the NOVX gene include methods in which protection from cleavage agents is used to detect mismatched bases in RNA/RNA

or RNA/DNA heteroduplexes. See, e.g., Myers, et al., 1985. Science 230: 1242.
In general, the art technique of "mismatch cleavage" starts by providing heteroduplexes of formed by hybridizing (labeled) RNA or DNA containing the wild-type NOVX
sequence with potentially mutant RNA or DNA obtained from a tissue sample. The double-stranded duplexes are treated with an agent that cleaves single-stranded regions of the duplex such as which will exist due to basepair mismatches between the control and sample strands.
For instance, RNA/DNA duplexes can be treated with RNase and DNA/DNA hybrids treated with S~ nuclease to enzymatically digesting the mismatched regions. In other embodiments, either DNA/DNA or RNA/DNA duplexes can be treated with hydroxylamine or osmium tetroxide and with piperidine in order to digest mismatched regions. After digestion of the mismatched regions, the resulting material is then separated by size on denaturing polyacrylamide gels to determine the site of mutation.
See, e.g., Cotton, et al., 1988. Proc. Natl. Acad. Sci. USA 85: 4397; Saleeba, et al., 1992.
Methods Enzymol. 217: 286-295. In an embodiment, the control DNA or RNA can be labeled for detection.
In still another embodiment, the mismatch cleavage reaction employs one or more proteins that recognize mismatched base pairs in double-stranded DNA (so called "DNA
mismatch repair" enzymes) in defined systems for detecting and mapping point mutations in NOVX cDNAs obtained from samples of cells. For example, the mutt enzyme of E.
coli cleaves A at G/A mismatches and the thymidine DNA glycosylase from HeLa cells cleaves T at G/T mismatches. See, e.g., Hsu, et al., 1994. Carcinogenesis 15:
1657-1662.
According to an exemplary embodiment, a probe based on A NOVX sequence, e.g., a wild-type NOVX sequence, is hybridized to a cDNA or other DNA product from a test cell(s). The duplex is treated with a DNA mismatch repair enzyme, and the cleavage products, if any, can be detected from electrophoresis protocols or the like.
See, e.g., U.S.
Patent No. 5,459,039.
In other embodiments, alterations in electrophoretic mobility will be used to identify mutations in NOVX genes. For example, single strand conformation polymorphism (SSCP) may be used to detect differences in electrophoretic mobility between mutant and wild type nucleic acids. See, e.g., Orita, et al., 1989.
Proc. Natl.
Acad. Sci. USA: 86: 2766; Cotton, 1993. Mutat. Res. 285: 125-144; Hayashi, 1992. Genet.
Anal. Tech. Appl. 9: 73-79. Single-stranded DNA fragments of sample and control NOVX
nucleic acids will be denatured and allowed to renature. The secondary structure of single-stranded nucleic acids varies according to sequence, the resulting alteration in electrophoretic mobility enables the detection of even a single base change.
The DNA
fragments may be labeled or detected with labeled probes. The sensitivity of the assay may be enhanced by using RNA (rather than DNA), in which the secondary structure is more sensitive to a change in sequence. In one embodiment, the subject method utilizes heteroduplex analysis to separate double stranded heteroduplex molecules on the basis of changes in electrophoretic mobility. See, e.g., Keen, et al., 1991. Trends Genet. 7: 5.
In yet another embodiment, the movement of mutant or wild-type fragments in polyacrylamide gels containing a gradient of denaturant is assayed using denaturing gradient gel electrophoresis (DGGE). See, e.g., Myers, et al., 1985. Nature 313: 495.
When DGGE is used as the method of analysis, DNA will be modified to insure that it does not completely denature, for example by adding a GC clamp of approximately 40 by of high-melting GC-rich DNA by PCR. In a further embodiment, a temperature gradient is used in place of a denaturing gradient to identify differences in the mobility of control and sample DNA. See, e.g., Rosenbaum and Reissner, 1987. Biophys. Chem. 265:
12753.
Examples of other techniques for detecting point mutations include, but are not limited to, selective oligonucleotide hybridization, selective amplification, or selective primer extension. For example, oligonucleotide primers may be prepared in which the known mutation is placed centrally and then hybridized to target DNA under conditions that permit hybridization only if a perfect match is found. See, e.g., Saiki, et al., 1986.
Nature 324: 163; Saiki, et al., 1989. Proc. Natl. Acad. Sci. USA 86: 6230.
Such allele specific oligonucleotides are hybridized to PCR amplified target DNA or a number of different mutations when the oligonucleotides are attached to the hybridizing membrane and hybridized with labeled target DNA.
Alternatively, allele specific amplification technology that depends on selective PCR amplification may be used in conjunction with the instant invention.
Oligonucleotides used as primers for specific amplification may carry the mutation of interest in the center of the molecule (so that amplification depends on differential hybridization; see, e.g., Gibbs, et al., 1989. Nucl. Acids Res. 17: 2437-2448) or at the extreme 3'-terminus of one primer where, under appropriate conditions, mismatch can prevent, or reduce polymerase extension (see, e.g., Prossner, 1993. Tibtech.
11: 238). In addition it may be desirable to introduce a novel restriction site in the region of the mutation to create cleavage-based detection. See, e.g., Gasparini, et al., 1992. Mol. Cell Probes 6: 1. It is anticipated that in certain embodiments amplification may also be performed using Taq ligase for amplification. See, e.g., Barany, 1991. Proc.
Natl. Acad.

Sci. USA 88: 189. In such cases, ligation will occur only if there is a perfect match at the 3'-terminus of the 5' sequence, making it possible to detect the presence of a known mutation at a specific site by looking for the presence or absence of amplification.
The methods described herein may be performed, for example, by utilizing pre-packaged diagnostic kits comprising at least one probe nucleic acid or antibody reagent described herein, which may be conveniently used, e.g., in clinical settings to diagnose patients exhibiting symptoms or family history of a disease or illness involving A
NOVX gene.
Furthermore, any cell type or tissue, preferably peripheral blood leukocytes, in which NOVX is expressed may be utilized in the prognostic assays described herein.
However, any biological sample containing nucleated cells may be used, including, for example, buccal mucosal cells.
PHARMACOGENOMICS
Agents, or modulators that have a stimulatory or inhibitory effect on NOVX
activity (e.g., NOVX gene expression), as identified by a screening assay described herein can be administered to individuals to treat (prophylactically or therapeutically) disorders (The disorders include metabolic disorders, diabetes, obesity, infectious disease, anorexia, cancer-associated cachexia, cancer, neurodegenerative disorders, Alzheimer's Disease, Parkinson's Disorder, immune disorders, and hematopoietic disorders, and the various dyslipidemias, metabolic disturbances associated with obesity, the metabolic syndrome X
and wasting disorders associated with chronic diseases and various cancers.) In conjunction with such treatment, the pharmacogenomics (i.e., the study of the relationship between an individual's genotype and that individual's response to a foreign compound or drug) of the individual may be considered. Differences in metabolism of therapeutics can lead to severe toxicity or therapeutic failure by altering the relation between dose and blood concentration of the pharmacologically active drug. Thus, the pharmacogenomics of the individual permits the selection of effective agents (e.g., drugs) for prophylactic or therapeutic treatments based on a consideration of the individual's genotype.
Such pharmacogenomics can further be used to determine appropriate dosages and therapeutic regimens. Accordingly, the activity of NOVX protein, expression of NOVX
nucleic acid, or mutation content of NOVX genes in an individual can be determined to thereby select appropriate agents) for therapeutic or prophylactic treatment of the individual.

Pharmacogenomics deals with clinically significant hereditary variations in the response to drugs due to altered drug disposition and abnormal action in affected persons.
See e.g., Eichelbaum, 1996. Clin. Exp. Pharmacol. Physiol., 23: 983-985;
Linder, 1997.
Clin. Chem., 43: 254-266. In general, two types of pharmacogenetic conditions can be differentiated. Genetic conditions transmitted as a single factor altering the way drugs act on the body (altered drug action) or genetic conditions transmitted as single factors altering the way the body acts on drugs (altered drug metabolism). These pharmacogenetic conditions can occur either as rare defects or as polymorphisms. For example, glucose-6-phosphate dehydrogenase (G6PD) deficiency is a common inherited enzymopathy in which the main clinical complication is hemolysis after ingestion of oxidant drugs (anti-malarials, sulfonamides, analgesics, nitrofurans) and consumption of fava beans.
As an illustrative embodiment, the activity of drug metabolizing enzymes is a major determinant of both the intensity and duration of drug action. The discovery of genetic polymorphisms of drug metabolizing enzymes (e.g., N-acetyltransferase 2 (NAT
2) and cytochrome Pregnancy Zone Protein Precursor enzymes CYP2D6 and CYP2C19) has provided an explanation as to why some patients do not obtain the expected drug effects or show exaggerated drug response and serious toxicity after taking the standard and safe dose of a drug. These polymorphisms are expressed in two phenotypes in the population, the extensive metabolizer (EM) and poor metabolizer (PM). The prevalence of PM is different among different populations. For example, the gene coding for CYP2D6 is highly polymorphic and several mutations have been identified in PM, which all lead to the absence of functional CYP2D6. Poor metabolizers of CYP2D6 and CYP2C19 quite frequently experience exaggerated drug response and side effects when they receive standard doses. If a metabolite is the active therapeutic moiety, PM show no therapeutic response, as demonstrated for the analgesic effect of codeine mediated by its CYP2D6-formed metabolite morphine. At the other extreme are the so called ultra-rapid metabolizers who do not respond to standard doses. Recently, the molecular basis of ultra-rapid metabolism has been identified to be due to CYP2D6 gene amplification.
Thus, the activity of NOVX protein, expression of NOVX nucleic acid, or mutation content of NOVx genes in an individual can be determined to thereby select appropriate agents) for therapeutic or prophylactic treatment of the individual. In addition, pharmacogenetic studies can be used to apply genotyping of polymorphic alleles encoding drug-metabolizing enzymes to the identification of an individual's drug responsiveness phenotype. This knowledge, when applied to dosing or drug selection, can avoid adverse reactions or therapeutic failure and thus enhance therapeutic or prophylactic efficiency when treating a subject with A NOVX modulator, such as a modulator identified by one of the exemplary screening assays described herein.
MONITORING OF EFFECTS DURING CLINICAL TRIALS
Monitoring the influence of agents (e.g., drugs, compounds) on the expression or activity of NOVX (e.g., the ability to modulate aberrant cell proliferation and/or differentiation) can be applied not only in basic drug screening, but also in clinical trials.
For example, the effectiveness of an agent determined by a screening assay as described herein to increase NOVX gene expression, protein levels, or upregulate NOVX
activity, can be monitored in clinical trails of subjects exhibiting decreased NOVX gene expression, protein levels, or downregulated NOVX activity. Alternatively, the effectiveness of an agent determined by a screening assay to decrease NOVX
gene expression, protein levels, or downregulate NOVX activity, can be monitored in clinical trails of subjects exhibiting increased NOVX gene expression, protein levels, or upregulated NOVX activity. In such clinical trials, the expression or activity of NOVX
and, preferably, other genes that have been implicated in, for example, a cellular proliferation or immune disorder can be used as a "read out" or markers of the immune responsiveness of a particular cell.
By way of example, and not of limitation, genes, including NOVX, that are modulated in cells by treatment with an agent (e.g., compound, drug or small molecule) that modulates NOVX activity (e.g., identified in a screening assay as described herein) can be identified. Thus, to study the effect of agents on cellular proliferation disorders, for example, in a clinical trial, cells can be isolated and RNA prepared and analyzed for the levels of expression of NOVX and other genes implicated in the disorder. The levels of gene expression (i.e., a gene expression pattern) can be quantified by Northern blot analysis or RT-PCR, as described herein, or alternatively by measuring the amount of protein produced, by one of the methods as described herein, or by measuring the levels of activity of NOVX or other genes. In this manner, the gene expression pattern can serve as a marker, indicative of the physiological response of the cells to the agent.
Accordingly, this response state may be determined before, and at various points during, treatment of the individual with the agent.

In one embodiment, the invention provides a method for monitoring the effectiveness of treatment of a subject with an agent (e.g., an agonist, antagonist, protein, peptide, peptidomimetic, nucleic acid, small molecule, or other drug candidate identified by the screening assays described herein) comprising the steps of (T~
obtaining a pre-administration sample from a subject prior to administration of the agent;
(iy detecting the level of expression of A NOVX protein, mRNA, or genomic DNA in the preadministration sample; (iiy obtaining one or more post-administration samples from the subject; (iv) detecting the level of expression or activity of the NOVX
protein, mRNA, or genomic DNA in the post-administration samples; (v) comparing the level of expression or activity of the NOVX protein, mRNA, or genomic DNA in the pre-administration sample with the NOVX protein, mRNA, or genomic DNA in the post administration sample or samples; and (vt~ altering the administration of the agent to the subject accordingly. For example, increased administration of the agent may be desirable to increase the expression or activity of NOVX to higher levels than detected, i.e., to increase the effectiveness of the agent. Alternatively, decreased administration of the agent may be desirable to decrease expression or activity ofNOVX to lower levels than detected, i.e., to decrease the effectiveness of the agent.
Methods of Treatment The invention provides for both prophylactic and therapeutic methods of treating a subject at risk of (or susceptible to) a disorder or having a disorder associated with aberrant NOVX expression or activity. The disorders include cardiomyopathy, atherosclerosis, hypertension, congenital heart defects, aortic stenosis, atrial septal defect (ASD), atrioventricular (A-V) canal defect, ductus arteriosus, pulmonary stenosis, subaortic stenosis, ventricular septal defect (VSD), valve diseases, tuberous sclerosis, scleroderma, obesity, transplantation, adrenoleukodystrophy, congenital adrenal hyperplasia, prostate cancer, neoplasm; adenocarcinoma, lymphoma, uterus cancer, fertility, hemophilia, hypercoagulation, idiopathic thrombocytopenic purpura, immunodeficiencies, graft versus host disease, A)DS, bronchial asthma, Crohn's disease;
multiple sclerosis, treatment of Albright Hereditary Ostoeodystrophy, and other diseases, disorders and conditions of the like.
These methods of treatment will be discussed more fully, below.

DISEASES AND DISORDERS
Diseases and disorders that are characterized by increased (relative to a subject not suffering from the disease or disorder) levels or biological activity may be treated with Therapeutics that antagonize (i.e., reduce or inhibit) activity. Therapeutics that antagonize activity may be administered in a therapeutic or prophylactic manner.
Therapeutics that may be utilized include, but are not limited to: (i) an aforementioned peptide, or analogs, derivatives, fragments or homologs thereof; (ii) antibodies to an aforementioned peptide;
(iir~ nucleic acids encoding an aforementioned peptide; (iv) administration of antisense nucleic acid and nucleic acids that are "dysfunctional" (i.e., due to a heterologous insertion within the coding sequences of coding sequences to an aforementioned peptide) that are utilized to "knockout" endogenous function of an aforementioned peptide by homologous recombination (see, e.g., ~apecchi, 1989. Science 244: 1288-1292); or (v) modulators i.e., inhibitors, agonists and antagonists, including additional peptide mimetic of the 1 S invention or antibodies specific to a peptide of the invention) that alter the interaction between an aforementioned peptide and its binding partner.
Diseases and disorders that are characterized by decreased (relative to a subject not suffering from the disease or disorder) levels or biological activity may be treated with Therapeutics that increase (i.e., are agonists to) activity. Therapeutics that upregulate activity may be administered in a therapeutic or prophylactic manner.
Therapeutics that may be utilized include, but are not limited to, an aforementioned peptide, or analogs, derivatives, fragments or homologs thereof; or an agonist that increases bioavailability.
Increased or decreased levels can be readily detected by quantifying peptide and/or RNA, by obtaining a patient tissue sample (e.g., from biopsy tissue) and assaying it in vitro for RNA or peptide levels, structure and/or activity of the expressed peptides (or mRNAs of an aforementioned peptide). Methods that are well-known within the art include, but are not limited to, immunoassays (e.g., by Western blot analysis, immunoprecipitation followed by sodium dodecyl sulfate (SDS) polyacrylamide gel electrophoresis, immunocytochemistry, etc.) and/or hybridization assays to detect expression of mRNAs (e.g., Northern assays, dot blots, in situ hybridization, and the like).
PROPHYLACTIC METHODS
In one aspect, the invention provides a method for preventing, in a subject, a disease or.condition associated with an aberrant NOVX expression or activity, by administering to the subject an agent that modulates NOVX expression or at least one NOVX activity. Subjects at risk for a disease that is caused or contributed to by aberrant NOVX expression or activity can be identified by, for example, any or a combination of diagnostic or prognostic assays as described herein. Administration of a prophylactic agent can occur prior to the manifestation of symptoms characteristic of the NOVX
aberrancy, such that a disease or disorder is prevented or, alternatively, delayed in its progression. Depending upon the type of NOVX aberrancy, for example, A NOVX
agonist or NOVX antagonist agent can be used for treating the subject. The appropriate agent can be determined based on screening assays described herein. The prophylactic methods of the invention are further discussed in the following subsections.
Therapeutic Methods Another aspect of the invention pertains to methods of modulating NOVX
expression or activity for therapeutic purposes. The modulatory method of the invention involves contacting a cell with an agent that modulates one or more of the activities of NOVX protein activity associated with the cell. An agent that modulates NOVX
protein activity can be an agent as described herein, such as a nucleic acid or a protein, a naturally-occurring cognate ligand of A NOVX protein, a peptide, A NOVX
peptidomimetic, or other small molecule. In one embodiment, the agent stimulates one or more NOVX protein activity. Examples of such stimulatory agents include active NOVX
protein and a nucleic acid molecule encoding NOVX that has been introduced into the cell. In another embodiment, the agent inhibits one or more NOVX protein activity.
Examples of such inhibitory agents include antisense NOVX nucleic acid molecules and anti-NOVX antibodies. These modulatory methods can be performed i~ vitro (e.g., by culturing the cell with the agent) or, alternatively, ih vivo (e.g., by administering the agent to a subject). As such, the invention provides methods of treating an individual afflicted with a disease or disorder characterized by aberrant expression or activity of A NOVX
protein or nucleic acid molecule. In one embodiment, the method involves administering an agent (e.g., an agent identified by a screening assay described herein), or combination of agents that modulates (e.g., up-regulates or down-regulates) NOVX
expression or activity. In another embodiment, the method involves administering A NOVX
protein or nucleic acid molecule as therapy to compensate for reduced or aberrant NOVX
expression or activity.

Stimulation of NOVX activity is desirable in situations in which NOVX is abnormally downregulated and/or in which increased NOVX activity is likely to have a beneficial effect. One example of such a situation is where a subject has a disorder characterized by aberrant cell proliferation and/or differentiation (e.g., cancer or immune associated disorders). Another example of such a situation is where the subject has a gestational disease (e.g., preclampsia).
Determination of the Biological Effect of the Therapeutic In various embodiments of the invention, suitable in vitro or in vivo assays are performed to determine the effect of a specific Therapeutic and whether its administration is indicated for treatment of the affected tissue.
In various specific embodiments, in vitro assays may be performed with representative cells of the types) involved in the patient's disorder, to determine if. a given Therapeutic exerts the desired effect upon the cell type(s). Compounds for use in therapy may be tested in suitable animal model systems including, but not limited to rats, mice, chicken, cows, monkeys, rabbits, and the like, prior to testing in human subjects.
Similarly, for in vivo testing, any of the animal model system known in the art may be used prior to administration to human subjects.
Prophylactic and Therapeutic Uses of the Compositions of the Invention The NOVX nucleic acids and proteins of the invention are useful in potential prophylactic and therapeutic applications implicated in a variety of disorders including, but not limited to: metabolic disorders, diabetes, obesity, infectious disease, anorexia, cancer-associated cancer, neurodegenerative disorders, Alzheimer's Disease, Parkinson's Disorder, immune disorders, hematopoietic disorders, and the various dyslipidemias, metabolic disturbances associated with obesity, the metabolic syndrome X and wasting disorders associated with chronic diseases and various cancers.
As an example, a cDNA encoding the NOVX protein of the invention may be useful in gene therapy, and the protein may be useful~when administered to a subject in need thereof. By way of non-limiting example, the compositions of the invention will have efficacy for treatment of patients suffering from: metabolic disorders, diabetes, obesity, infectious disease, anorexia, cancer-associated cachexia, cancer, neurodegenerative disorders, Alzheimer's Disease, Parkinson's Disorder, immune disorders, hematopoietic disorders, and the various dyslipidemias.

Both the novel nucleic acid encoding the NOVX protein, and the NOVX protein of the invention, or fragments thereof, may also be useful in diagnostic applications, wherein the presence or amount of the nucleic acid or the protein are to be assessed.
A further use could be as an anti-bacterial molecule (i.e., some peptides have been found to possess anti-bacterial properties). These materials are further useful in the generation of antibodies, which immunospecifically-bind to the novel substances of the invention for use in therapeutic or diagnostic methods.
The invention will be further described in the following examples, which do not limit the scope of the invention described in the claims.

EXAMPLES
Example A: Polynucleotide And Polypepfide Sequences, And Homology Data EXAMPLE 1.
The NOV 1 clone was analyzed, and the nucleotide and encoded polypeptide sequences are shown in Table 1A.
Table 1A. NOVl Sequence Analysis SEQ ID NO: 1 1239 by NOVla, AACCAGGGCCTTATCCAGGGCCACGCTTACAGAACTCCCACGGACACACCATGATTAG

CG100488-OlGACCCTGCTGCTGTCCACTTTGGTGGCCCTCAGTTGTGGGGTCTCCACTTACGCGCCT

DNA GATATGTCTAGGATGCTTGGAGGTGAAGAAGCGAGGCCCAACAGCTGGCCCTGGCAGG

TGAGTCTGCAGTACAGCTCCAATGGCCAGTGGTACCACACCTGCGGAGGGTCCCTGAT

Sequence AGCCAACAGCTGGGTCCTGACGGCTGCCCACTGCATCAGCTCCTCCGGGATCTACCGC

GTGATGCTGGGCCAGCATAACCTCTACGTTGCAGAGTCCGGCTCGCTGGCCGTCAGTG

TCTCTAAGATTGTGGTGCACAAGGACTGGAACTCCGACCAGGTCTCCAAAGGGAACGA

CATTGCCCTGCTCAAACTGGCTAACCCCGTCTCCCTCACCGACAAGATCCAGCTGGCC

TGCCTCCCTCCTGCCGGCACCATTCTACCCAACAACTACCCCTGCTACGTCACGGGCT

GGGGAAGGCTGCAGAGTAACGGGGCTCTCCCTGATGACCTGAAGCAGGGCCAGTTGCT

GGTTGTGGACTATGCCACCTGCTCCAGCTCTGGCTGGTGGGGCAGCACCGTGAAGACG

AATATGATCTGTGCTGGGGGTGATGGCGTGATATGCACCTGCAACGGAGACTCCGGTG

GGCCGCTGAACTGTCAGGCATCTGACGGCCGGTGGGAGGTGCATGGCATCGGCAGCCT

CACGTCGGTCCTTGGTTGCAACTACTACTACAAGCCCTCCATCTTCACGCGGGTCTCC

AACTACAACGACTGGATCAATTCGGTAAGAACCGGAGCAGCCCTGAGCCCCAAGGCAC

TGACCTGCTCACCTGGCCTCGGGAGTGCCATGCCCACCTGGCGACTGAGAACCCCCTC

CTTCCTCTTGAGAGCTAGATGGGAACCCCTTGGAGGAGGCTGCAGACCTTGGCAACTG

CTGAGTCCCCCATGGGTCCCCAAAATTTCTGTGTGGGTAAAGCTGAGTGAAAAGGAAC

ATGAGAGTATGGCCTTGTCCAAAGACGTTGGACACTCCTCAGGTACGTTAAGAGTGAG

TTCCACAGGAATGATTTTATTTTTGTGTATTTGTGTGTGGCCCAGACTCTACCATCCA

GTGCTATAAATGGGTATATGTCTGCAAAACCCAAAACCTGATACTTTGAGACCCCCAT

AGCATTAATTATTGGAAATTA

ORF Start: ATG at 51 ORF Stop: TAA at 1167 SEQ ID NO: 2 372 as MW at 40287.81cD

NOVla, MIRTLLLSTLVALSCGVSTYAPDMSRMLGGEEARPNSWPWQVSLQYSSNGQWYHTCGG

Protein GNDIALLKLANPVSLTDKIQLACLPPAGTILPNNYPCYVTGWGRLQSNGALPDDLKQG

QLLVVDYATCSSSGWWGSTVKTNMICAGGDGVICTCNGDSGGPLNCQASDGRWEVHGI

SBquenC e GSLTSVLGCNYYYKPSIFTRVSNYNDWINSVRTGAALSPKALTCSPGLGSAMPTWRLR

TPSF.LLRARWEPLGGGCRPWQLLSPPWVPKISVWVKLSEKEHESMALSKDVGHSSGTL

RVSSTGMILFLCICVWPRLYHPVL

SEQ ID NO: 3 1188 by NOVlb, ATGATTAGGACCCTGCTGCTGTCCACTTTGGTGGCTGGAGCCCTCAGTTGTGGGGTCT

DNA C'rGGCCCTGGCAGGTGAGTCTGCAGTACAGCTCCAATGGCCAGTGGTACCACACCTGC

GGAGGGTCCCTGATAGCCAACAGCTGGGTCCTGACGGCTGCCCACTGCATCAGCTCCT

Sequence CCGGGATCTACCGCGTGATGCTGGGCCAGCATAACCTCTACGTTGCAGAGTCCGGCTC

GCTGGCCGTCAGTGTCTCTAAGATTGTGGTGCACAAGGACTGGAACTCCGACCAGGTC

TCCAAAGGGAACGACATTGCCCTGCTCAAACTGGCTAACCCCGTCTCCCTCACCGACA

AGATCCAGCTGGCCTGCCTCCCTCCTGCCGGCACCATTCTACCCAACAACTACCCCTG

CTACGTCACGGGCTGGGGAAGGCTGCAGACCAACGGGGCTCTCCCTGATGACCTGAAG

CAGGGCCAGTTGCTGGTTGTGGACTATGCCACCTGCTCCAGCTCTGGCTGGTGGGGCA

GCACCGTGAAGACGAATATGATCTGTGCTGGGGGTAATGGCGTGATATGCACCTGCAA

CGGAGACTCTGGCGGGCCACTGAACTGTCAGGCGTCTGACGGCCGGTGGCAGGTGCAC

GGCATCGTCAGCTTCGGGTCTCGCCTCGGCTGCAACTACTACCACAAGCCCTCCGTCT

TCACGCGGGTCTCCAATTACATCGACTGGATCAATTCGGTAAGAACCGGACCAGCCTT

GAGCCCCAAGGCACTACCCTGCTCACCTGGCCTCGGGAGTGCCATGCCCACCTGGTGA

CTGAGAATCCCCTCCTTCCTCTTGAGAGCTAGATGGGAACCCCTTGGAGGAGGCTGCA

GACCTGAGTAACTGCTGGGCCTGCCATGGGTCCCCCAAATTTCTGTGTGGATAAAGCT

GAGTGAAAAGGAACATAGAGGGTGGCCTTGTCCAAAGAGGTTGGACACTCCTCAGGCA

TATGAAGAGTGAGTTCCGCTGGGCGCCGTGGCTCATGCCTGTAATCCCAGCTCTTTGG

GAGGCCAAGGCGGGCAGATCACGAGGTCAGAAGTTCAAGACCAGCCTGACCAACCTGG

CAAAACCCCATGTCTACTAAAAAAATCC

ORF Start: ATG at 1 ORF Stop: TGA at 868 SEQ ID NO: 4 289 as MW at 30820.8kD

NOVlb, MIRTLLLSTLVAGALSCGVSTYAPDMSRMLGGEEARPNSWPWQVSLQYSSNGQWYHTC

PrOtelri SKGNDIALLKLANPVSLTDKIQLACLPPAGTILPNNYPCYVTGWGRLQTNGALPDDLK

QGQLLWDYATCSSSGWWGSTVKTNMICAGGNGVICTCNGDSGGPLNCQASDGRWQVH

S8ClileriCBGIVSFGSRLGCNYYHKPSVFTRVSNYIDWINSVRTGPALSPKALPCSPGLGSAMPTW

SEQ ID NO: 5 889 by NOV1C, ATGATTAGGACCCTGCTGCTGTCCACTTTGGTGGCTGGAGCCCTCAGTTGTGGGGTCT

DNA CTGGCCCTGGCAGGTGAGTCTGCAGTACAGCTCCAATGGCCAGTGGTACCACACCTGC

GGAGGGTCCCTGATAGCCAACAGCTGGGTCCTGACGGCTGCCCACTGCATCAGCTCCT

SeCltleriCeCCGGGATCTACCGCGTGATGCTGGGCCAGCATAACCTCTACGTTGCAGAGTCCGGCTC

GCTGGCCGTCAGTGTCTCTAAGATTGTGGTGCACAAGGACTGGAACTCCGACCAGGTC

TCCAAAGGGAACGACATTGCCCTGCTCAAACTGGCTAACCCCGTCTCCCTCACCGACA

AGATCCAGCTGGCCTGCCTCCCTCCTGCCGGCACCATTCTACCCAACAACTACCCCTG

CTACGTCACGGGCTGGGGAAGGCTGCAGACCAACGGGGCTCTCCCTGATGACCTGAAG

CAGGGCCAGTTGCTGGTTGTGGACTATGCCACCTGCTCCAGCTCTGGCTGGTGGGGCA

GCACCGTGAAGACGAATATGATCTGTGCTGGGGGTAATGGCGTGATATGCACCTGCAA

CGGAGACTCTGGCGGGCCACTGAACTGTCAGGCGTCTGACGGCCGGTGGCAGGTGCAC

GGCATCGTCAGCTTCGGGTCTCGCCTCGGCTGCAACTACTACCACAAGCCCTCCGTCT

TCACGCGGGTCTCCAATTACATCGACTGGATGATTGCAAATAACTAACCAAAAGAAGT

CCCTGGGACTGTTTCAGACTTGGAAAGGTCACGGAAGGAAAATAATATAATAAAGTGG

CAACTATGCAAAAAAAAAA

ORF Start: ATG at 1 ORF Stop: TAA at 799 SEQ ID NO: 6 266 as MW at 28573.2kD

NOV1C, MIRTLLLSTLVAGALSCGVSTYAPDMSRMLGGEEARPNSWPWQVSLQYSSNGQWYHTC

PrOtelri SKGNDIALLKLANPVSLTDKIQLACLPPAGTILPNNYPCYVTGWGRLQTNGALPDDLK

QGQLLWDYATCSSSGWWGSTVKTNMICAGGNGVICTCNGDSGGPLNCQASDGRWQVH

SeC111eriCeGIVSFGSRLGCNYYHKPSVFTRVSNYIDWMIANN

SEQ ID NO: 7 ~ 1188 by NOV1C1, ATGATTAGGACCCTGCTGCTGTCCACTTTGGTGGCTGGAGCCCTCAGTTGTGGGGACC

CG1OO488-O8C~'CTTACCCACCTTATGTGACTAGGGTGGTTGGCGGTGAAGAAGCGAGGCCCAACAG

DNA CTGGCCCTGGCAGGTGAGTCTGCAGTACAGCTCCAATGGCCAGTGGTACCACACCTGC

GGAGGGTCCCTGATAGCCAACAGCTGGGTCCTGACGGCTGCCCACTGCATCAGCTCCT

SeCllleriCBCCGGGATCTACCGCGTGATGCTGGGCCAGCATAACCTCTACGTTGCAGAGTCCGGCTC

GCTGGCCGTCAGTGTCTCTAAGATTGTGGTGCACAAGGACTGGAACTCCGACCAGGTC

TCCAAAGGGAACGACATTGCCCTGCTCAAACTGGCTAACCCCGTCTCCCTCACCGACA

AGATCCAGCTGGCCTGCCTCCCTCCTGCCGGCACCATTCTACCCAACAACTACCCCTG

CTACGTCACGGGCTGGGGAAGGCTGCAGGCCAACGGGGCTCTCCCTGATGACCTGAAG

CAGGGCCAGTTGCTGGTTGTGGACTATGCCACCTGCTCCAGCTCTGGCTGGTGGGGCA

GCACCGTGAAGACGAATATGATCTGTGCTGGGGGTAATGGCGTGATATGCACCTGCAA

CGGAGACTCTGGCGGGCCACTGAACTGTCAGGCGTCTGACGGCCGGTGGCAGGTGCAC

GGCATCGTCAGCTTCGGGTCTCGCCTCGGCTGCAACTACTACCACAAGCCCTCCGTCT

TCACGCGGGTCTCCAA'~'TACATCGACTGGATCAATTCGGTAAGAACCGGACCAGCCTT

GAGCCCCAAGGCACTACCCTGCTCACCTGGCCTCGGGAGTGCCATGCCCACCTGGTGA
CTGAGAATCCCCTCCTTCCTCTTGAGAGrTAGATGre~AArrrrmm~rnr_rnnnrmr_ra GAGGCCAAGGCGGGCAGA
CAAAACCCCATGTCTACT.
ORF Start: ATG at 1 ~ ORF Stop: TGA at 868 SEQ ID NO: 8 289 aa~MW at 30826.8kD
NOV1CI, MIRTLLLSTLVAGALSCGDPTYPPYVTRWGGEEARPNSWPVJQVSLQYSSNGQWYHTC

Protein SKGNDIALLKLANPVSLTDKIQLACLPPAGTILPNNYPCYVTGWGRLQANGALPDDLK
.

QGQLLVVDYATCSSSGWWGSTVKTNMICAGGNGVICTCNGDSGGPLNCQASDGRWQVH

11CriCe q GIVSFGSRLGCNYYHKPSVFTRVSNYIDWINSVRTGPALSPKALPCSPGLGSAMPTW

SEQ ID NO: 9 889 by NOVle, ATGATTAGGACCCTGCTGCTGTCCACTTTGGTGGCTGGAGCCCTCAGTTGTGGGGACC
CG100488-09~CCACTTACCCACCTTATGTGACTAGGGTGGTTGGCGGTGAAGAAGCGAGGCCCAACAG
DNA CTGGCCCTGGCAGGTGAGTCTGCAGTACAGCTCCAATGGCCAGTGGTACCACACCTGC
I GGAGGGTCCCTGATAGCCAACAGCTGGGTCCTGACGGCTGCCCACTGCATCAGCTCCT
!Sequence CCGGGATCTACCGCGTGATGCTGGGCCAGCATAACCTCTACGTTGCAGAGTCCGGCTC
GCTGGCCGTCAGTGTCTCTAAGATTGTGGTGCACAAGGACTGGAACTCCGACCAGGTC
TCCAAAGGGAACGACATTGCCCTGCTCAAACTGGCTAACCCCGTCTCCCTCACCGACA
CTACGTCACGGGCTGGGGAAGGCTGCAGGCCAACGGGGCTCTCCCTGATGACCTGAAG
CAGGGCCAGTTGCTGGTTGTGGACTATGCCACCTGCTCCAGCTCTGGCTGGTGGGGCA
GCACCGTGAAGACGAATATGATCTGTGCTGGGGGTAATGGCGTGATATGCACCTGCAA
CGGAGACTCTGGCGGGCCACTGAACTGTCAGGCGTCTGACGGCCGGTGGCAGGTGCAC
GGCATCGTCAGCTTCGGGTCTCGCCTCGGCTGCAACTACTACCACAAGCCCTCCGTCT
TCACGCGGGTCTCCAATTACATCGACTGGATGATTGCAAATAACTAACCAAAAGAAGT
CCCTGGGACTGTTTCAGACTTGGAAAGGTCACGGAAGGAAAATAATATAATAAAGTGG
CAACTATGC
ORF Start: ATG at 1 ORF Stop: TAA at 799 SEQ ID NO: 10 266 as MW at 28579.2kD
~NOVIC, MIRTLLLSTLVAGALSCGDPTYPPYVTRVVGGEEARPNSWPWQVSLQYSSNGQWYHTC

'CG100488-09GGSLIANSWVLTAAHCISSSGIYRVMLGQHNLYVAESGSLAVSVSKIVVHKDWNSDQV

'PIOtBlriSKGNDIALLKLANPVSLTDKIQLACLPPAGTILPNNYPCYVTGWGRLQANGALPDDLK

QGQLLVVDYATCSSSGWWGSTVKTNMICAGGNGVICTCNGDSGGPLNCQASDGRWQVH
SeqlIeriCC
' , GIVSFGSRLGCNYYHKPSVFTRVSNYIDWMIANN

SEQ ID NO: 11 ~ 1080 by NOVIf, GGATCCGTCTCCACTTACGCGCCTGATATGTCTAGGATGCTTGGAGGTGAAGAAGCGA

DNA CCACACCTGCGGAGGGTCCCTGATAGCCAACAGCTGGGTCCTGACGGCTGCCCACTGC

ATCAGCTCCTCCGGGATCTACCGCGTGATGCTGGGCCAGCATAACCTCTACGTTGCAG
Seq11e11Ce AGTCCGGCTCGCTGGCCGTCAGTGTCTCTAArammrmrnmr__rarnaccarmr_r_TTnm~

CGACCAGGTCTCCAAAGGGAACGACATTGCCCTGCTCAAACTGGCTAACCCCGTCTCC
CTCGCCGACAAGATCCAGCTGGCCTGCCTCCCTCCTGCCGGCACCATTCTACCCAACA
ACTACCCCTGCTACGTCACGGGCTGGGGAAGGCTGCAGACCAACGGGGCTCTCCCTGA
TGACCTGAAGCAGGGCCGGTTGCTGGTTGTGGACTATGCCACCTGCTCCAGCTCTGGC
TGGTGGGGCAGCACCGTGAAGACGAATATGATCTGTGCTGGGGGTGATGGCGTGATAT
GCACCTGCAACGGAGACTCCGGTGGGCCGCTGAACTGTCAGGCATCTGACGGCCGGTG
GGAGGTGCATGGCATCGGCAGCCTCACGTCGGTCCTTGGTTGCAACTACTACTACAAG
CCCTCCATCTTCACGCGGGTCTCCAACTACAACGACTGGATCAATTCGGTAAGAACCG
GAGCAGCCCTGAGCCCCAAGGCACTGACCTGCTCACCTGGCCTCGGGAGTGCCATGCC
CACCTGGCGACTGAGAACCCCCTCCTTCCTCTTGAGAGCTAGATGGGAACCCCTTGGA

GGGTAAAGCTGAGTGAAAAGGAACATGAGAGTATGGCCTTGTCCAAAGACGTTGGACA
CTCCTCAGGTACGTTAAGAGTGAGTTCCACAGGAATGATTTTATTTTTGTGTATTTGT
GTGTGGCCCAGACTCTACCATCCAGTGCTACTCGAG
ORF Start: at 1 . ORF Stop: end of sequence SEQ ID NO: 12 360 as MW at 39027.2kD
NOVIf, GSVSTYAPDMSRMLGGEEARPNSWPWQISLQYSSNGQWYHTCGGSLIANSWVLTAAHC

P1'Oteln L~KIQLACLPPAGTILPNNYPCYVTGWGRLQTNGALPDDLKQGRLLVVDYATCSSSG
WWGSTVKTNMICAGGDGVICTCNGDSGGPLNCQASDGRWEVHGIGSLTSVLGCNYYYK
Sequence pSIFTRVSNYNDWINSVRTGAALSPKALTCSPGLGSAMPTWRLRTPSFLLRARWEPLG
GGCRPWQLLSPPWVPKISVWVKLSEKEHESMALSKDVGHSSGTLRVSSTGMILFLCIC
VWPRLYHPVLLE
SEQ ID NO: 13 1080 by NOVIg, GGATCCGTCTCCACTTACGCGCCTGATATGTCTAGGATGCGTGGAGGTGAAGAAGCGA

DNA CCACACCTGCGGAGGGTCCCTGATAGCCAACAGCTGGGTCCTGACGGCTGCCCACTGC
ATCAGCTCCTCCGGGATCTACCGCGTGATGCTGGGCCAGCATAACCTCTACGTTGCAG
Sequence AGTCCGGCTCGCTGGCCGTCAGTGTCTCTAAGATTGTGGTGCACAAGGACTGGAACTC
CGACCAGGTCTCCAAAGGGAACGACATTGCCCTGCTCAAACTGGCTAACCCCGTCTCC
CTCACCGACAAGATCCAGCTGGCCTGCCTCCCTCCTGCCGGCACCATTCTACCCAACA
ACTACCCCTGCTACGTCACGGGCTGGGGAAGGCTGCAGACCAACGGGGCTCTCCCTGA
TGACCTGAAGCAGGGCCGGTTGCTGGTTGTGGACTATGCCACCTGCTCCAGCTCTGGC
TGGTGGGGCAGCACCGTGAAGACGAATATGATCTGTGCTGGGGGTGATGGCGTGATAT
GCACCTGCAACGGAGACTCCGGTGGGCCGCTGAACTGCCAGGCATCTGACGGCCGGTG
GGAGGTGCATGGCATCGGCAGCCTCACGTCGGTCCTTGGTTGCAACTACTACTACAAG
CCCTCCATCTTCACGCGGGTCTCCAACTACAACGACTGGATCAATTCGGTAAGAACCG
GAGCAGCCCTGAGTCCCAAGGCACTGCCCTGCTCACCTGGCCTCGGGAGTGCCATGCC
CACCTGGCGACTGAGAACCCCCTCCTTCCTCTTGAGAGCTAGATGGGAACCCCTTGGA
GGAGGCTGCAGACCTTGGCAACTGCTGAGTCCCCCATGGGTCCCCAAAATTTCTGTGT
GGGTAAAGCTGAGTGAAAAGGAACATGAGAGTATGGCCTTGTCCAAAGACGTTGGACA
CTCCTCAGGTATGTTAAGAGTGAGTTCCACAGGAATGATTTTATTTTTGTGTATTTGT
GAATGGCCCAGACTCTACCATCCAGTGCTACTCGAG
ORF Start: at 1 ORF Stop: end of sequence SEQ ID NO: 14 360 as MW at 39142.3kD
NOVIg, GSVSTYAPDMSRMRGGEEARPNSWPWQVSLQYSSNGQWYHTCGGSLIANSWVLTAAHC

P1'Oteln LTDKIQLACLPPAGTILPNNYPCYVTGWGRLQTNGALPDDLKQGRLLVVDYATCSSSG
WWGSTVKTNMICAGGDGVICTCNGDSGGPLNCQASDGRWEVHGIGSLTSVLGCNYYYK
SeqUenCe PSIFTRVSNYNDWINSVRTGAALSPKALPCSPGLGSAMPTWRLRTPSFLLRARWEPLG
GGCRPWQLLSPPWVPKISVWVKLSEKEHESMALSKDVGHSSGMLRVSSTGMILFLCIC
EWPRLYHPVLLE
SEQ ID NO: 15 1080 by NOV1I7, GGATCCGTCTCCACTTACGCGCCTGATATGTCTAGGATGCTTGGAGGTGAAGAAGCGA

DNA ~ CCACACCTGCGGAGGGTCCCTGATAGCCAACAGCTGGGTCCTGACGGCTGCCCACTGC
ATCAGCTCCTCCAGGATCTACCGCGTGATGCTGGGCCAGCATAACCTCTACGTTGCAG
Sequence AGTCCGGCTCGCTAGCCGTCAGTGTCTCTAAGATTGTGGTGCACAAGGACTGGAACTC
CAACCAGGTCTCCAAAGGGAACGACATTGCCCTGCTCAAACTGGCTAACCCCGTCTCC
CTCACCGACAAGATCCAGCTGGCCTGCCTCCCTCCTGCCGGCACCATTCTACCCAACA
ACTACCCCTGCTACGTCACAGGCTGGGGAAGGCTGCAGACCAACGGGGCTCTCCCTGA
TGACCTGAAGCAGGGCCGGTTGCTGGTTGTGGACTATGCCACCTGCTCCAGCTCTGGC
TGGTGGGGCAGCACCGTGAAGACGAATATGATTTGTGCTGGGGGTGATGGCGTGATAT
GCACCTGCAACGGAGACTCCGGTGGGCCGCTGAACTGTCAGGCATCTGACGGCCGGTG
GGAGGTGCATGGCATCGGCAGCCTCACGTCGGTCCTTGGTTGCAACTACTACTACAAG
CCCTCCATCTTCACGCGGGTCTCCAACTACAACGACTGGATCAATTCGGTAAGAACCG

GAGCAGCCCTGAGCCCCAAGGCACTGACCTGCTCACCTGGCCTCGGGAGTGCCATGCC

CACCTGGCGACTGAGAACCCCCTCCTTCCTCTTGAGAGCTAGATGGGAACCCCTTGGA

GGAGGCTGCAGACCTTGGCAACTGCTGAGTCCCCCATGGGTCCCCAAAATTTCTGTGT

GGGTAAAGCTGAGTGAAAAGGAACATGAGAGTATGGCCTTGTCCAAAGACGTTGGACA

CTCCTCAGGTATGTTAAGAGTGAGTTCCACAGGAATGATTTTATTTTTGTGTATTTGT

GTGTGGCCCAGACTCTACCATCCAGTGCTACTCGAG

ORF Start: at 1 ORF
Stop: end of sequence SEQ ID NO: 16 360 as 'MW at 39171.4kD

NOV1I1, GSVSTYAPDMSRMLGGEEARPNSWPWQVSLQYSSNGQWYHTCGGSLIANSWVLTAAHC

PrOteln LTDKIQLACLPPAGTILPNNYPCYVTGWGRLQTNGALPDDLKQGRLLVVDYATCSSSG

WWGSTVKTNMICAGGDGVICTCNGDSGGPLNCQASDGRWEVHGIGSLTSVLGCNYYYK
SeqLlenCe pSIFTRVSNYNDWINSVRTGAALSPKALTCSPGLGSAMPTWRLRTPSFLLRARWEPLG

GGCRPWQLLSPPWVPKISVWVKLSEKEHESMALSKDVGHSSGMLRVSSTGMILFLCIC

VWPRLYHPVLLE

SEQ ID NO: 17 1080 by NOV11, GGATCCGTCTCCACTTACGCGCCTGATATGTCTAGGATGCTTGGAGGTGAAGAAGCGA

DNA CCACACCTGCGGAGGGTCCCTGATAGCCAACAGCTGGGTCCTGACGGCTGCCCACTGC

ATCAGCTCCTCCGGGATCTACCGCGTGATGCTGGGCCAGCATAACCTCTACGTTGCAG

Sequenc e AGTCCGGCTCGCTGGCCGTCAGTGTCTCTAAGATTGTGGTGCACAAGGACTGGAACTC

CGACCAGGTCTCCAAAGGGAACGACATTGCCCTGCTCAAACTGGCTAACCCCGTCTCC

CTCACCGACAAGATCCAGCTGGCCTGCCTCCCTCCTGCCGGCACCATTCTACCCAACA

ACTACCCCTGCTACGTCACGGGCTGGGGAAGGCTGCAGACCAACGGGGCTCTCCCTGA

TGACCTGAAGCAGGGCCGGTTGCTGGTTGTGGACTATGCCACCTGCTCCAGCTCTGGC

TGGTGGGGCAGCACCGTGAAGACGAATATGATCTGTGCTGGGGGTGATGGCGTGATAT

GCACCTGCAACGGAGACTCCGGTGGGCCGCTGAACTGTCAGGCATCTGACGGCCGGTG

GGAGGTGCATGGCATCGGCAGCCTCACGTCGGTCCTTGGTTGCAACTACTACTACAAG

CCCTCCATCTTCACGCGGGTCTCCAACTACAACGACTGGATCAATTCGGTAAGAACCG

GAGCAGCCCTGAGCCCCAAGGCACTGCCCTGCTCACCTGGCCTCGGGAGTGCCATGCC

CACCTGGCGACTGAGAACCCCCTCCTTCCTCTTGAGAGCTAGATGGGAACCCCTTGGA

GGAGGCTGCAGACCTTGGCAACTGCTGAGTCCCCCATGGGTCCCCAAAATTTCTGTGT

GGGTAAAGCTGAGTGAAAAGGAACATGAGAGTATGGCCTTGTCCAAAGACGTTGGACA

CTCCTCAGGTATGTTAAGAGTGAGTTCCACAGGAATGATTTTATTTTTGTGTATTTGT

GTGTGGCCCAGACTCTACCATCCAGTGCTACTCGAG

ORF Start: at 1 ORF
Stop:
end of sequence SEQ ID NO: 18 360 MW at 39069.3kD
as NOVI1, GSVSTYAPDMSRMLGGEEARPNSWPWQVSLQYSSNGQWYHTCGGSLIANSWVLTAAHC

PT'Otel ri LTDKIQLACLPPAGTILPNNYPCWTGWGRLQTNGALPDDLKQGRLLVVDYATCSSSG

WWGSTVKTNMICAGGDGVICTCNGDSGGPLNCQASDGRWEVHGIGSLTSVLGCNYYYK
SeC~llenCe pSIFTRVSNYNDWINSVRTGAALSPKALPCSPGLGSAMPTWRLRTPSFLLRARWEPLG

GGCRPWQLLSPPWVPKISVWVKLSEKEHESMALSKDVGHSSGMLRVSSTGMILFLCIC

VWPRLYHPVLLE

SEQ ID NO: 19 1023 by NOV1~, GGATCCGTCTCCACTTACGCGCCTGATATGTCTAGGATGCTTGGAGGTGAAGAAGCGA

DNA CCACACCTGCGGAGGGTCCCTGATAGCCAACAGCTGGGTCCTGACGGCTGCCCACTGC

ATCAGCTCCTCCGGGATCTACCGCGTGATGCTGGGCCAGCATAACCTCTACGTTGCAG
Sequence AGTCCGGCTCGCTGGCCGTCAGTGTCTCTAAGATTGTGGTGCACAAGGACTGGAACTC

CGACCAGGTCTCCAAAGGGAACGACATTGCCCTGCTCAAACTGGCTAACCCCGTCTCC

CTCACCGACAAGATCCAGCTGGCCTGCCTCCCTCCTGCCGGCACCATTCTACCCAACA

ACTACCCCTGCTACGTCACGGGCTGGGGAAGGCTGCAGACCAACGGGGCTCTCCCTGA

TGACCTGAAGCAGGGCCGGTTGCTGGTTGTGGACTATGCCACCTGCTCCAACTCTGGC

TGGTGGGGCAGCACCGTGAAGACGAATATGATCTGTGCTGGGGGTGATGGCGTGATAT

GCACCTGCAACGGAGACTCCGGTGGGCCGCTGAACTGTCAGGCATCTGACGGCCGGTG
GGAGGTGCATGGCATCGGCAGCCTCACGTCGGTCCTTGGTTGCAACTACTACTACAAG
CCCTCCATCTTCACGCGGGTCTCCAACTACAACGACTGGATCAATTCGGTAAGAACCG
GAGCAGCCCTGAGCCCCAAGGCACTGACCTGCTCACCTGGCCTCGGGAGTGCCATGCC
CACCTGGCGACTGAGAACCCCCTCCTTCCTCTTGAGAACTAGATGGGAACCCCTTGGA
GGAGGCTGCAGACCTTGGCAACTGCTGAGTCCCCCATGGGTCCCCAAAATTTCTGTGT
_ GGGTAAAGCTGAGTGAAAAGGAACATGAGAGTATGGCCTTGTCCAAAGACGTTGGACA
CTCCTCAGGTACGTTAAGAGTGAGTTCCACACTCGAG
ORF Start: at 1 ORF Stop: end of sequence SEQ ID NO: 20 341 as MW at 36814.4kD
NOV1J, GSVSTYAPDMSRMLGGEEARPNSRPWQVSLQYSSNGQWYHTCGGSLIANSWVLTAAHC

LTDKIQLACLPPAGTILPNNYPCYVTGWGRLQTNGALPDDLKQGRLLVVDYATCSNSG
PrOtelri ~r,7GSTVKTNMICAGGDGVICTCNGDSGGPLNCQASDGRWEVHGIGSLTSVLGCNYYYK
Sequence pSIFTRVSNYNDWINSVRTGAALSPKALTCSPGLGSAMPTWRLRTPSFLLRTRWEPLG
GGCRPWQLLSPPWVPKISVWVKLSEKEHESMA'GSKDVGHSSGTLRVSSTLE
Sequence comparison of the above protein sequences yields the following sequence relationships shown in Table 1B.

Table 1S.
Comparison of NOVIa against NOVlb through NOVlj.

Protein NOVla Residues/Identities/

Sequence Match ResiduesSimilarities for the Matched Region NOVlb 1..287 275/289 (95%) I ..289 ~ 280/289 (96%) NOV 1 c 1..260 249/262 (95%) 1..262 255/262 (97%) NOV 1 d 1..287 2671289 (92%) 1..289 276/289 (95%) NOV 1 a 1..260 241 /262 (91 %) 1..262 251/262 (94%) NOV 1 f 17..372 352/356 (98%) 3..358 355/356 (98%) NOVIg 17..372 350/356 (98%) 3..358 352/356 (98%) NOVlh 17..372 351/356 (98%) 3..358 354/356 (98%) NOVli 17..372 352/356 (98%) 3..358 354/356 (98%) NOVlj 17..353 ~ 332/337 (98%) 3..339 3351337 (98%) Further analysis of the NOV 1 a protein yielded the following properties shown in Table 1 C.
Table 1C. Protein Sequence Properties NOVla PSort 0.5469 probability located in outside; 0.1000 probability located in analysis: endoplasmic reticulum (membrane); 0.1000 probability located in endoplasmic reticulum (lumen); 0.1000 probability located in lysosome (lumen) SignalP Cleavage site between residues 17 and 18 analysis:
A search of the NOV 1 a protein against the Geneseq database, a proprietary database that contains sequences published in patents and patent publication, yielded several homologous proteins shown in Table 1D.

Table 1D. Geneseq Results for NOVIa NOVla Identities/

Geneseq Protein/Organism/Length Residues/SimilaritiesExpect for Identifier[Patent #, Date] Match the MatchedValue ResiduesRegion AAM78338 Human protein SEQ ID 1..277 261/279 e-156 NO 1000 -~ (93%) Homo Sapiens, 1052 aa. 1..279 268/279 ~ (95%) [W0200157190-A2, 09-AUG-2001 ]

AAP70760 Human pancreas elastase-21..263 259/265 e-155 - Sus (97%) scrofa, 269 aa. [JP62000276-A,1..265 262/265 06- (98%) JAN-1987]

AAP60059 Sequence of human pancreatic15..263 245/249 e-149 ~ (98%) elastase IIB - Homo Sapiens,1..249 248/249 253 (99%) aa. [EP198645-A, 22-OCT-1986]

AAP60062 Sequence of human pancreatic1..263 230/265 e-137 ' (86%) elastase IIA encoded 1..265 248/265 on pH2E2 - ~ (92%) Homo Sapiens, 269 aa.
[EP198645-A, 22-OCT-1986]

AAP61723 Human elastase II - Homo1..263 2291265 e-135 Sapiens, (86%) 269 aa. [JP61192288-A, 1..265 246/265 26-AUG- (92%) ~
1986]

In a BLAST
search of public sequence datbases, the NOV
1 a protein was found to have homology to the proteins shown in the BLASTP data in Table 1E.

Table 1E. Public BLASTP Results for NOVla Protein NOVIa Identities/

AccessionProtein/Organism/LengthResidues!SimilaritiesExpect for Number Match the Matched Value Residues Portion Q96QV5 BA265F14.3 (ELASTASE 1..263 262/265 (98%)e-157 2B) -Homo Sapiens (Human), 1..265 263/265 (98%) 269 aa.

P08218 Elastase 2B precursor 1..263 259/265 (97%)e-155 (EC

3.4.21.71) - Homo Sapiens1..265 262/265 (98%) (Human), 269 aa.

P08217 Elastase 2A precursor 1..263 230/265 (86%)e-137 (EC

3.4.21.71) -Homo Sapiens1..265 248/265 (92%) (Human), 269 aa.

P08419 Elastase 2 precursor 1..263 204/265 (76%)e-122 (EC

3.4.21.71) - Sus scrofa1..265 230/265 (85%) (Pig), 269 aa.

Q29461 Elastase 2 precursor 1..263 202/265 (76%)e-121 (EC

3.4.21.71) - Bos taurus1..265 231/265 (86%) (Bovine), 269 aa.

PFam analysis predicts that the NOV
1 a protein contains the domains shown in the Table 1F.
Table 1F. Domain Analysis of NOVla Identities/
Pfam Domain NOVla Match Region Similarities Expect Value for the Matched Region trypsin 27..260 120/261 (46%) 1.3e-87 1921261 (74%) S EXAMPLE 2.
The NOV2 clone was analyzed, and the nucleotide and encoded polypeptide sequences are shown in Table 2A.
Table 2A. NOV2 Sequence Analysis SEQ ID NO: 21 1800 by NOV2a, GAGCCTCTCTTCACCATGTGCTTCGTCCCCCTGGTGTGCTGGGTGGTGTGTACCTGCC

DNA AGTGTACATGCTCTACCTGCTGAGTCTGATGCAACCCAAGCCGGGGGCCCCGCGCCTC

CAGCCCCCACCCAACCAGAGAGGGTTGTGCTCCTTGGCGGCAGATGGGCTCTGGAATC

Sequence AGAAAATCCTATTTGAGGAGCAGGACCTCCGGAAGCACGGCCTAGACGGC'GAAGAC'CT

CTCTGCCTTCCTCAACATGAACATCTTCCAGAAGGACATCAACTGTGAGAGGTACTAC

AGGGGGAGGGCGGGGCAGGCCCAGACCAGGACGTGACCAGGCTGTTGACCGAGTACGC
GTTTTCTGAAAGGAGCTTCCTGGCACTCACCAGCCGCTTCCTGTTTGGACTCCTGAAC
GAGGAGACCAGGAGCCACCTGGAGAAGAGTCTCTGCTGGAAGGTCTCGCCGCACATCA
AGATGGACCTGTTGCAGTGGATCCAAAGCAAAGCTCAGAGCGACGGCTCCACCCTGCA
GCAGGGCTCCTTGGAGTTCTTCAGCTGCTTGTACGAGATCCAGGAGGAGGAGTTTATC
CAGCAGGCCCTGAGCCACTTCCAGGTGATCGTGGTCAGCAACATTGCCTCCAAGATGG
AGCACATGGTCTCCTCGTTCTGTCTGAAGCGCTGCAGGAGCGCCCAGGTGCTGCACTT
GTATGGCGCCACCTACAGCGCGGACGGGGAAGACCGCGCGAGGTGCTCCGCAGGAGCG
CACACGCTGTTGGTGCAGCTGAGACCAGAGAGGACCGTTCTGCTGGACGCCTACAGTG
AACATCTGGCAGCGGCCCTGTGCACCAATCCAAACCTGATAGAGCTGTCTCTGTACCG
AAATGCCCTGGGCAGCCGGGGGGTGAAGCTGCTCTGTCAAGGACTCAGACACCCCAAC
TGCAAACTTCAGAACCTGAGGAGGCTGAAGAGGTGCCGCATCTCCAGCTCAGCCTGCG
AGGACCTCTCTGCAGCTCTCATAGCCAATAAGAATTTGACAAGGATGGATCTCAGTGG
CAACGGCGTTGGATTCCCAGGCATGATGCTGCTTTGCGAGGGCCTGCGGCATCCCCAG
TGCAGGCTGCAGATGATTCAGTTGAGGAAGTGTCAGCTGGAGTCCGGGGCTTGTCAGG
AGATGGCTTCTGTGCTCGGCACCAACCCACATCTGGTTGAGTTGGACCTGACAGGAAA
TGCACTGGAGGATTTGGGCCTGAGGTTACTATGCCAGGGACTGAGGCACCCAGTCTGC
AGACTACGGACTTTGTGGTGCAGGCTGAAGATCTGCCGCCTCACTGCTGCTGCCTGTG
ACGAGCTGGCCTCAACTCTCAGTGTGAACCAGAGCCTGAGAGAGCTGGACCTGAGCCT
GAATGAGCTGGGGGACCTCGGGGTGCTGCTGCTGTGTGAGGGCCTCAGGCATCCCACG
TGCAAGCTCCAGACCCTGCGGAGGTTGGGCATCTGCCGGCTGGGCTCTGCCGCCTGTG
AGGGTCTTTCTGTGGTGCTCCAGGCCAACCACAACCTCCGGGAGCTGGACTTGAGTTT
CAACGACCTGGGAGACTGGGGCCTGTGGTTGCTGGCTGAGGGGCTGCAACATCCCGCC
TGCAGACTCCAGAAACTGTGGTGAGCATCGGGGAGTGACGGGGTGGCAGTGGTCACGT
TT
ORF Start: ATG at 16 ORF Stop: TGA at 1762 SEQ ID NO: 22 S82 as MW at 65280.8kD
NOV2a, MCFVPLVCWVVCTCLQQQLEGGGLLRQTSRTTTAVYMLYLLSLMQPKPGAPRLQPPPN

PPOtelri SFQEFFAAMYYILDEGEGGAGPDQDVTRLLTEYAFSERSFLALTSRFLFGLLNEETRS
HLEKSLCWKVSPHIKMDLLQWIQSKAQSDGSTLQQGSLEFFSCLYEIQEEEFIQQALS
SeCI118riC8 HFQVIVVSNIASKMEHMVSSFCLKRCRSAQVLHLYGATYSADGEDRARCSAGAHTLLV
QLRPERTVLLDAYSEHLAAALCTNPNLIELSLYRNALGSRGVKLLCQGLRHPNCKLQN
LRRLKRCRISSSACEDLSAALIANKNLTRMDLSGNGVGFPGMMLLCEGLRHPQCRLQM
IQLRKCQLESGACQEMASVLGTNPHLVELDLTGNALEDLGLRLLCQGLRHPVCRLRTL
WCRLKICRLTAAACDELASTLSVNQSLRELDLSLNELGDLGVLLLCEGLRHPTCKLQT
LRRLGICRLGSAACEGLSVVLQANHNLRELDLSFNDLGDWGLWLLAEGLQHPACRLQK
LW
SEQ ID NO: 23 1683 by NOV2b, GCGCGCCTCTCTTCACCATGTGCTTCGTCCCCCTGGTGTGCTGGGTGGTGTGTACCTG

DNA GCAGTGTACATGCTCTACCTGCTGAGTCTGATGCAACCCAAGCCGGGGGCCCCGCGCC
TCCAGCCCCCACCCAACCAGAGAGGGTTGTGCTCCTTGGCGGCAGATGGGCTCTGGAA
SeC111eriCe TCAGAAAATCCTATTTGAGGAGCAGGACCTCCGGAAGCACGGCCTAGACGGGGAAGAC
GTCTCTGCCTTCCTCAACATGAACATCTTCCAGAAGGACATCAACTGTGAGAGGTACT
ACAGCTTCATCCACTTGAGTTTCCAGGAATTCTTTGCAGCTATGTACTATATCCTGGA
CGAGGGGGAGGGCGGGGCAGGCCCAGACCAGGACGTGACCAGGCTGTTGACCGAGTAC
GCGTTTTCTGAAAGGAGCTTCCTGGCACTCACCAGCCGCTTCCTGTTTGGACTCCTGA
ACGAGGAGACCAGGAGCCACCTGGAGAAGAGTCTCTGCTGGAAGGTCTCGCCGCACAT
CAAGATGGACCTGTTGCAGTGGATCCAAAGCAAAGCTCAGAGCGACGGCTCCACCCTG
CAGCAGGGCTCCTTGGAGTTCTTCAGCTGCTTGTACGAGATCCAGGAGGAGGAGTTTA
TCCAGCAGGCCCTGAGCCACTTCCAGGTGATCGTGGTCAGCAACATTGCCTCCAAGAT
GGAGCACATGGTCTCCTCGTTCTGTCTGAAGCGCTGCAGGAGCGCCCAGGTGCTGCAC
TTGTATGGCGCCACCTACAGCGCGGACGGGGAAGACCGCGCGAGGTGCTCCGCAGGAG
CGCACACGCTGTTGGTGCAGCTCAGACCAGAGAGGACCGTTCTGCTGGACGCCTACAG
TGAACATCTGGCAGCGGCCCTGTGCACCAATCCAAACCTGATAGAGCTGTCTCTGTAC
CGAAATGCCCTGGGCAGCCGGGGGGTGAAGCTGCTCTGTCAAGGACTCAGACACCCCA
ACTGCAAACTTCAGAACCTGAGGCTGAAGAGGTGCCGCATCTCCAGCTCAGCCTGCGA

GGACCTCTCTGCAGCTCTCATAGCCAATAAGAATTTGACAAGGATGGATCTCAGTGGC
AACGGCGTTGGATTCCCAGGCATGATGCTGCTTTGCGAGGGCCTGCGGCATCCCCAAT
GCAGGCTGCAGATGATTCAGCTGAAGATCTGCCGCCTCACTGCTGCTGCCTGTGACGA
GCTGGCCTCAACTCTCAGTGTGAACCAGAGCCTGAGAGAGCTGGACCTGAGCCTGAAT
GAGCTGGGGGACCTCGGGGTGCTGCTGCTGTGTGAGGGCCTCAGGCATCCCACGTGCA
AGCTCCAGACCCTGCGGTTGGGCATCTGCCGGCTGGGCTCTGCCGCCTGTGAGGGTCT
TTCTGTGGTGCTCCAGGCCAACCACAACCTCCGGGAGCTGGACTTGAGTTTCAACGAC
CTGGGAGACTGGGGCCTGTGGTTGCTGGCTGAGGGGCTGCAACATCCCGCCTGCAGAC
TCCAGAAACTGTGGTGAGCATCGGGGAGTGACGGGGTGGCAGTGGTCACGTTTGGACA
GTGGAAGCGCCTTCTCATCCTTCATTTTTCTATTTATGAACTATCCTGCTTCACTACA
A
OIZF Start: ATG at 18 OItF Stop: TGA at 1 S81 SEQ ID NO: 24 S21 as MW at 58384.71cD
NOV2b, MCFVPLVCWWCTCLQQQLEGGGLLRQTSRTTTAVYMLYLLSLMQPKPGAPRLQPPPN

P1'Oteln SFQEFFAAMYYILDEGEGGAGPDQDVTRLLTEYAFSERSFLALTSRFLFGLLNEETRS
HLEKSLCWKVSPHIKMDLLQWIQSKAQSDGSTLQQGSLEFFSCLYEIQEEEFIQQALS
SequeriCe HFQVIWSNIASKMEHMVSSFCLKRCRSAQVLHLYGATYSADGEDRARCSAGAHTLLV
QLRPERTVLLDAYSEHLAAALCTNPNLIELSLYRNALGSRGVKLLCQGLRHPNCKLQN
LRLKRCRISSSACEDLSAALIANKNLTRMDLSGNGVGFPGMMLLCEGLRHPQCRLQMI
QLKICRLTAAACDELASTLSVNQSLRELDLSLNELGDLGVLLLCEGLRHPTCKLQTLR
LGICRLGSAACEGLSWLQANHNLRELDLSFNDLGDWGLWLLAEGLQHPACRLQKLW
Sequence comparison of the above protein sequences yields the following sequence relationships shown in Table 2B.
Table 2B. Comparison of NOV2a against NOV2b.
Protein Sequence ~ NOV2a Residues/ ~ Identities!
Match Residues Similarities for the Matched Region NOV2b ~ 1..523 4S0/S23 (86%) 1..520 464/S23 (88%) Further analysis of the NOV2a protein yielded the following properties shown in Table 2C.
Table 2C. Protein Sequence Properties NOV2a PSort 0.3700 probability located in outside; 0.1900 probability located in lysosome analysis: (lumen); 0.1000 probability located in endoplasmic reticulum (membrane);
0.1000 probability located in endoplasmic reticulum (lumen) SignalP Cleavage site between residues SO and S1 analysis:
S A search of the NOV2a protein against the Geneseq database, a proprietary database that contains sequences published in patents and patent publication, yielded several homologous proteins shown in Table 2D.

Table 2D. Geneseq Results for NOV2a NOV2a Identities/

Geneseq Protein/Organism/Length Residues/SimilaritiesExpect [Patent for Identifier#, Date] Match the MatchedValue ResiduesRegion AAU01067Human secreted protein 1..581 275/602 e-143 sequence (45%) encoded by gene #28 - 1..596 380/602 Homo (62%) Sapiens, 630 aa. [W0200123402-A1, OS-APR-2001]

AAE07514Human PYRIN-1 protein I ..581 275/602 e-143 - Homo (45%) Sapiens, 1034 aa. [W0200161005-406..1001380/602 (62%) A2, 23-AUG-2001]

AAU01096Gene 28 Human secreted 1..523 236/532 e-123 protein (44%) homologous amino acid 1..482 324/532 sequence - (60%) Homo Sapiens, 484 aa.

[W0200123402-A1, OS-APR-2001]

AAY39778CBDAKDO1 protein sequence1..464 210/487 e-106 - (43%) Homo Sapiens, 514 aa. 1..481 298/487 (61%) [W09946290-A1, 16-SEP-1999]

ABG04570Novel human diagnostic 156..324167/169 Se-90 protein (98%) #4561 - Homo Sapiens, 1..168 167/169 168 aa. (98%) [W0200175067-A2, 11-OCT-2001]

In a BLAST
search of public sequence datbases, the NOV2a protein was found to have homology to the proteins shown in the BLASTP data in Table 2E.

Table 2E. Public BLASTP Results for NOV2a Protein NOV2a Identities/

AccessionProtein/Organism/Length Residues/SimilaritiesExpect for Number Match the MatchedValue ResiduesPortion AAH28069 HYPOTHETICAL 120.2 KDA 1..582 577/582 0.0 (99%) PROTEIN - Homo Sapiens 400..976577/582 (Human), (99%) 1061 aa.

Q96P20 Cold autoinflammatory 1..581 275/602 e-143 syndrome 1 (45%) protein (Cryopyrin) (NACHT-,406..1001380/602 LRR- (62%) and PYD-containing protein 3) (PYRIN-containing APAF
I -like protein 1) (Angiotensin/vasopressin receptor AII/AVP-like) - Homo Sapiens (Human), 1034 aa.

AAL78632 NALP3 LONG ISOFORM - Homo1..581 275/602 e-143 (45%) Sapiens (Human), 1036 408..1003379/602 aa. (62%) AAL90874 MAST CELL MATURATION I ..581 274/603 e-140 (45%) INDUCIBLE PROTEIN 1 - 404..1000372/603 Mus (61%) musculus (Mouse), 1033 aa.

AAL12498 CRYOPYRIN - Homo Sapiens 1..464 220/485 e-115 (45%) (Human), 920 aa. 406..887310/485 (63%) PFam analysis he predicts domains that shown the NOV2a in protein the contains t Table 2F.
Table 2F. Domain Analysis of NOV2a Identities/
Pfam Domain NOV2a Match Region Similarities Expect Value for the Matched Region EXAMPLE 3.
The NOV3 clone was analyzed, and the nucleotide and encoded polypeptide sequences are shown in Table 3A.
Table 3A. NOV3 Sequence SEQ ID NO: 25 X481 NUV3a, CTTGTCTTGTTCCAGTTCTCAGAGGGAATGCTTTCAATTTTTCTCTATTCAGTATTAT

DNA G~GCCAGGCTGCAGGGGCCTTCGGATCACCACGGATGCCTGCTGGGGTCGCTGTGAG

ACCTTCTATCTATGGGGACAGAAACCCATTCTGGAACCCCCCTATATTGAAGCCCATC
Sequence ATCGAGTCTGTACCTACAACGAGACCAAACAGGTGACTGTCAAGCTGCCCAACTGTGC

CCCGGGAGTCGACCCCTTCTACACCTATCCCGTGGCCATCCGCTGTGACTGCGGAGCC

ORF Start: ATG at 28 IOIZF Stop: TGA at 382 SEQ ID NO: 26 ~ 118 as BMW at 13491.6kD
V3a, MLSIFLYSVLCWLWVCHRLCAVREFTFLAKKPGCRGLRITTDACWGRCETFYLWGQKP
101012-O1~II'EPPYIEAHHRVCTYNETKQVTVKLPNCAPGVDPFYTYPVAIRCDCGACSTATTECE
Sequence Further analysis of the NOV3a protein yielded the following properties shown in Table 3B.
Table 3B. Protein Sequence Properties NOV3a PSort 0.5500 probability located in endoplasmic reticulum (membrane); 0.1900 analysis: probability located in lysosome (lumen); 0.1449 probability located in microbody (peroxisome); 0.1000 probability located in endoplasmic reticulum (lumen) SignalP Cleavage site between residues 22 and 23 analysis:
A search of the NOV3a protein against the Geneseq database, a proprietary database that contains sequences published in patents and patent publication, yielded several homologous proteins shown in Table 3C.

Table 3C. Geneseq Results for NOV3a NOV3a Identities/

Geneseq Protein/Organism/Length Residues/SimilaritiesExpect [Patent Identifier#, Date] Match for the Value Residues Matched Region AAU10366Human beta-like glycoprotein20..118 93/99 (93%)7e-53 hormone, BetalO - Homo 36..130 95/99 (95%) Sapiens, 130 aa. [W0200173034-A2, OCT-2001 ]

AAG64065Human anterior pituitary 20..118 93/99 (93%)7e-53 hormone-related polypeptide #2 12..106 95/99 (95%) - Homo sapiens, 106 aa. (W0200144475-A1, 21-JUN-2001 ]

AAG64064Human anterior pituitary 20..118 93/99 (93%)7e-53 hormone-related polypeptide - 36..130 95/99 (95%) Homo Sapiens, 130 aa. [W0200144475-Al, JUN-2001 ]

AAG63211Amino acid sequence of 20..118 93/99 (93%)~ 7e-53 a human cystine knot polypeptide 36..130 95/99 (95%) - Homo sapiens, 130 aa. [W0200153346-A1, 26-JUL-2001 ]

AAE09440Human sbghGTa protein 20..118 93/99 (93%)7e-53 - Homo sapiens, 130 aa. [W0200160850-Al,36..130 95/99 (95%) 23-AUG-2001 ]

In a BLAST
search of public sequence datbases, the NOV3a protein was found to have homology to the proteins shown in the BLASTP data in Table 3D.

Table 3D. Public BLASTP Results for NOV3a NOV3a Identities!

Protein Similarities Residues/ Expect AccessionProtein/Organism/Length Match for the Value Number ResiduesMatched Portion Q9I997 FOLLICLE-STIMULATING 2..118 49/119 8e-20 (41%) HORMONE PRECURSOR - 3..116 66/119 (55%) Acipenser baerii (Siberian sturgeon), 128 aa.

Q98849 Gonadotropin beta-II chain4..115 431117 8e-16 precursor (36%) (GTH-II-beta) (Luteinizing9..120 63/117 hormone- (53%) like GTH) - Carassius auratus (Goldfish), 140 aa.

Q90ZK1 ~ FOLLICLE STIMULATING 5..115 46/112 2e-15 (41%) HORMONE BETA SUBUNIT 1..108 59/112 (52%) PRECURSOR - Rana ridibunda (Laughing frog) (Marsh frog), 123 as (fragment).

P01235 Gonadotropin beta-II chain26..115 37/91 (40%)2e-15 precursor (GTH-II-beta) (Luteinizing39..124 54/91 (58%) hormone-like GTH) - Cyprinus carpio (Common carp), 144 aa.

Q98TY3 LUTEINIZING HORMONE BETA 1..115 44/118 2e-15 (37%) SUBUNIT - Mylopharyngodon 8..120 62/118 (52%) piceus, 140 aa.

PFam analysis predicts that the NOV3a protein contains the domains shown in the Table 3E.
Table 3E. Domain Analysis of NOV3a Identities!
Pfam Domain NOV3a Match Region Similarities Expect Value fox the Matched Region Cys knot 29..116 36/92 (39%) 2.5e-14 62/92 (67%) S EXAMPLE 4.
The NOV4 clone was analyzed, and the nucleotide and encoded polypeptide sequences are shown in Table 4A.

Table 4A. NOV4 Sequence Analysis SEQ ID NO: 27 682 by NOV~Ia, ATGAAGACCCTGTTCCTGGGTGTCACGCTCGGCCTGGCCGCTGCCCTGTCCTTCACCC

DNA TCATGTACCTGCAGGAGCTGCCCAGGAGGGACCACTACATCTTTTACTGCAAAGACCA
GCACCATGGGGGCCTGCTCCACATGGGAAAGCTTGTGGGTGCTCCCTGCAGGGCCGTG
Sequence CCGCTGTCCCCACGTCGGCTCACCTGGCCACCTCACCTGCAGGTAGGAATTCTGATAC
CAACCGGGAGGCCCTGGAAGAATTTAAGAAATTGGTGCAGCGCAAGGGACTCTCGGAG
GAGGACATTTTCACGCCCCTGCAGACGGGTGAGGATGGCTGTGCCCAGTCCCCTGTGT
CCCTCTGCTGTGTCTGTCTGCTATCTCCAGTGTCCCATGACCCCCATGTCCTCCCATG
TCCCCCGCATTCCCCATGTGCCCCGAGTCTCCTCGCAGGGGCTCCCGGGCCCTGTTTA
GCGTCCTCCTCATTGGAGGCTCTGTGCTCTGGGCTGCGATGGGGTCTGGGGCTCCGCG
CTCTGGGCTGCGATGGGGTCTGGGGCTCCGCACTCTGGGCTGCGATGGGGTCTGGGGC
TCCGCGCTCTGGGCTGCGATGGGCTCTGGGGCTCTGAGCTCTGG
ORF Start: ATG at 1 ORF Stop: TGA at 673 SEQ ID NO: 28 224 as MW at 23172.11cD
NOV4a, MKTLFLGVTLGLAAALSFTLEEEDVHPEENPDAEWGQEAHVPAGAAQEGPLHLLLQRP

PrOteln EDTFTPLQTGEDGCAQSPVSLCCVCLLSPVSHDPHVLPCPPHSPCAPSLLAGAPGPCL
ASSSLEALCSGLRWGLGLRALGCDGVWGSALWAAMGSGAPRSGLRWALGL
Sequence Further analysis of the NOV4a protein yielded the following properties shown in Table 4B.
Table 4B. Protein Sequence Properties NOV4a PSort 0.7571 probability located in outside; 0.1000 probability located in analysis: endoplasmic reticulum (membrane); 0.1000 probability located in endoplasmic reticulum (lumen); 0.1000 probability located in microbody (peroxisome) SignaIP Cleavage site between residues 16 and 17 analysis:
A search of the NOV4a protein against the Geneseq database, a proprietary database that contains sequences published in patents and patent publication, yielded several homologous proteins shown in Table 4C.

Table 4C. Geneseq Results for NOV4a N0~4a Identities/

Geneseq Protein/Organism/Length Residues/SimilaritiesExpect [Patent for Identifier#, Date] Match the Matched Value ResiduesRegion AAB67741? Amino acid sequence 1..126 115/141 (81%)1e-56 of odorant binding polypeptide OBPIIa-delta1..141 119/141 (83%) -Homo Sapiens, 147 aa.

[W0200112806-A2, 22-FEB-2001 AAB67744Amino acid sequence of 1..71 70/85 (82%) 7e-33 odorant binding polypeptide OBPIIb-1..85 71/85 (83%) gamma - Homo Sapiens, 85 aa.

[WO200112806-A2, 22-FEB-2001]

AAB67743~ Amino acid sequence 31..71 38/41 (92%) 1e-17 of odorant binding polypeptide OBPIIb-beta139..17938/41 (92%) -Homo Sapiens, 179 aa.

[W0200112806-A2, 22-FEB-2001]

ABG11867- Novel human diagnostic 92..15446/74 (62%) 3e-13 protein #11858 - Homo Sapiens, 130..19847/74 (63%) 200 aa.

[W0200175067-A2, 11-OCT-2001]

ABG11867Novel human diagnostic 92..15446/74 (62%) 3e-13 protein #11858 - Homo Sapiens, 130..19847/74 (63%) 200 aa.

[W0200175067-A2, 11-OCT-2001]

In a BLAST
search of public sequence datbases, the NOV4a protein was found to have homology to the proteins shown in the BLASTP data in Table 4D.

Table 4D. Public BLASTP Results for NOV4a Protein NOV4a Identities/

AccessionProteinlOrganism/Length Residues!SimilaritiesExpect for Number Match the Matched Value ResiduesPortion Q9NY54 PUTATIVE ODORANT 1..126 1151141 (81%)3e-56 BINDING PROTEIN AD - 1..141 119/141 (83%) Homo Sapiens (Human), 147 aa.

Q9NY51 PUTATIVE ODORANT 1..71 70/85 (82%) 2e-32 BINDING PROTEIN BG - 1..85 71/85 (83%) Homo Sapiens (Human), 85 aa.

CAC33327 SEQUENCE 11 FROM PATENT 31..71 38/41 (92%) 3e-17 W00112806 - Homo Sapiens139..17938/41 (92%) ~

(Human), 179 as (fragment). I

Q9NY52 PUTATIVE ODORANT- 31..71 38/41 (92%) 3e-17 BINDING PROTEIN BB - 125..16538/41 (92%) Homo Sapiens (Human), 165 aa.

Q9NPH6 Odorant-binding protein 92..126 35/35 (100%)3e-12 2b precursor (OBPIIb) - 130..16435/35 (100%) Homo Sapiens (Human), 170 aa.

PFam analysis predicts that the NOV4a protein contains the domains shown in the Table 4E.
Table 4E. Domain Analysis of NOV4a Identities/
Pfam Domain NOV4a Match Region Similarities Expect Value for the Matched Region lipocalin 92..128 13/37 (35%) 0.00022 31/37 (84%) EXAMPLE 5.
The NOVS clone was analyzed, and the nucleotide and encoded polypeptide sequences are shown in Table SA.
Table SA. NOVS Sequence Analysis SEQ ID NO: 29 ~ 1178 by VSa _GGGATGGGAAAACTATGCCTGGGGCCGACGCTCTGCCCGGCTGCTGCCGCTGAGGAAA
GCCGGGACGCGGAGCCCCGCCGAGAGCTTCTTTGCTCCGGACGCCCCTGGACGTGGCG

GGCAGCCGCGAGGGTAACCACCATGATCCCCTGGGTGCTCCTGGCCTGTGCCCTCCCC
CTCAACTGGTCTGCAGCCTGCCTGGCCCCCAGGGCCCACCCGGCCCCCCAGGAGCCCC
AGGGCCCTCAGGAATGATGGGACGAATGGGCTTTCCTGGCAAAGACGGCCAAGATGGA

CACGACGGCGACCGGGGGGACAGCGGAGAGGAAGGTCCACCTGGCCGGACAGGTAACC

GGGGAAAGCCAGGACCAAAGGGCAAAGCCGGGGCCATTGGGCGGGCTGGCCCCCGTGG

CCCCAAGGGGGTCAACGGTACCCCCGGGAAGCATGGCACACCAGGCAAGAAGGGGCCC

AAGGGCAAGAAGGGGGAGCCAGGCCTCCCAGGCCCCTGCAGCTGTGGCAGTGGCCATA

CCAAGTCAGCTTTCTCGGTGGCAGTGACCAAGAGCTACCCACGGGAGCGGCTGCCCAT

CAAGTTTGACAAGATTCTGATGAACGAGGGTGGCCACTACAATGCTTCCAGCGGCAAG

TTCGTCTGCGGCGTGCCTGGGATCTACTACTTCACCTACGACATCACGCTGGCCAACA

AGCACCTGGCCATCGGCCTGGTGCACAACGGCCAGTACCGCATCCGGACCTTTGATGC

CAACACCGGCAACCACGATGTGGCCTCAGGCTCCACCATCCTGGCTCTCAAGCAGGGT

GACGAAGTTTGGCTGCAGATCTTCTACTCAGAGCAGAACGGGCTCTTCTATGACCCTT

ACTGGACAGACAGCCTCTTTACGGGCTTCCTAATCTATGCCGACCAGGATGACCCCAA

CGAGGTATAGACATGCCACGGCGGTCCTCCAGGCAGGGAACAAGCTTCTGGACTTGGG

CTTACAGAGCAAGACCCCACAACTGTAGGCTGGGGGTGGGGGGTCGAGTGAGCGGTTC

TAGCCTCAGGCTCACCTCCTCCGCCTCTTTTTTTTCCCCTTCATTAAATCCAAACCTT

TTTATTCATCCAAAAAAA

ORF Start: ATG at 4 ORF Stop: TAG at 994 SEQ ID NO: 30 330 as MW at 34833.2kD

NOVSa, MGKLCLGPTLCPAAAAEESRDAEPRRELLCSGRPWTWRAAARVTTMIPWVLLACALPC

CG101707-Ol~PLLGAFARRDFRKGSPQLVCSLPGPQGPPGPPGAPGPSGMMGRMGFPGKDGQDGH

DGDRGDSGEEGPPGRTGNRGKPGPKGKAGAIGRAGPRGPKGVNGTPGKHGTPGKKGPK
PrOtelri GKKGEPGLPGPCSCGSGHTKSAFSVAVTKSYPRERLPIKFDKILMNEGGHYNASSGKF

Sequence VCGVPGIYYFTYDITLANKHLAIGLVHNGQYRIRTFDANTGNHDVASGSTILALKQGD

EVWLQIFYSEQNGLFYDPYWTDSLFTGFLIYADQDDPNEV

Further analysis of the NOVSa protein yielded the following properties shown in Table SB.
Table SB. Protein Sequence Properties NOVSa PSort 0.7900 probability located in plasma membrane; 0.4043 probability located in analysis: microbody (peroxisome); 0.3000 probability located in Golgi body;
0.2000 probability located in endoplasmic reticulum (membrane) SignalP Cleavage site between residues 61 and 62 analysis:
A search of the NOVSa protein against the Geneseq database, a proprietary database that contains sequences published in patents and patent publication, yielded several homologous proteins shown in Table SC.

Table SC. Geneseq Results for NOVSa NOVSa Identities/

Geneseq Protein/Organism/Length Residues/SimilaritiesExpect for Identifier[Patent #, Date) Match the Matched Value ResiduesRegion AAU19557Human diagnostic and 1..330 3291330 (99%)0.0 therapeutic polypeptide (DITHP) #1432..331 329/330 (99%) - Homo Sapiens, 331 aa. [W0200162927-A2, 30-AUG-2001]

AAB50374Human adipocyte complement46..330 285/285 (100!)e-177 related protein homologue1..285 285/285 (100%) zacrp2 -Homo sapiens, 285 aa.

[WO200U73448-Al, 07-DEC-2000]

AAY54321A polypeptide designated46..330 285/285 (100%)e-177 ACRP30R1L which is a 1..285 285/285 (100%) homologue of ACRP30 -Homo Sapiens, 285 aa. [W09959618-A1, 25-NOV-1999]

AAB30232Human adipocyte complement46..330 285/285 (100%)e-177 related protein homologue1..285 285!285 (100%) zacrp2 -Homo Sapiens, 285 aa.

[W0200063376-A1, 26-OCT-2000]

ABB72178Rat protein isolated 42..330 271/289 (93%)e-168 from skin cells SEQ ID NO: 294 - Rattus 6..294 278/289 (95%) sp, 294 aa. [W0200190357-Al, 2001]

In a BLAST search of public sequence datbases, the NOVSa protein was found to have homology to the proteins shown in the BLASTP data in Table 5D.

Table SD. Public BLASTP Results for NOVSa Protein NOVSa Identities/

AccessionProtein/Organism/LengthResidues/SimilaritiesExpect for Number Match the Matched Value ResiduesPortion Q9BXJ5 Complement-clq tumor 46..330 285/285 (100%)e-177 necrosis factor-related protein 1..285 285/285 (100%) 2 precursor - Homo sapiens (Human), 285 aa.

Q9D8U4 1810033KOSRIK PROTEIN 42..330 272/289 (94%)e-168 - $

Mus musculus (Mouse), 6..294 279/289 (96%) 294 aa.

CAC21967 SEQUENCE 14 FROM PATENT76..325 160/250 (64%)1e-96 ~

W00073448 - Mus musculus29..278 191/250 (76%) (Mouse), 289 aa.

CAC21966 SEQUENCE 1 FROM PATENT 76..325 159/250 (63%)3e-96 ~

W00073448 - Homo sapiens43..292 192/250 (76%) (Human), 303 aa.

Q9BXJ2 Complement-clq tumor 76..325 159/250 (63%)3e-96 necrosis factor-related protein 29..278 192/250 (76%)
7 precursor - Homo Sapiens (Human), 289 aa.

PFam analysis predicts that the NOVSa protein contains the domains shown in the Table SE.
Table 5E. Domain Analysis of NOVSa Identities/

Pfam DomainNOVSa Match RegionSimilarities Expect Value for the Matched Region Collagen 83..141 32/60 (53%) 3.3e-05 .
40/60 (67%) Collagen 142..201 23160 (38%) 0.0014 37/60 (62%) Clq 196..320 45/138 (33%) 2.3e-38 93/138 (67%) EXAMPLE 6.
The NOV6 clone was analyzed, and the nucleotide and encoded polypeptide sequences are shown in Table 6A.

_ Table 6A. NOV6 Sequence Analysis SEQ ID NO: 31 1611 by NOV6a, GGAGCGTCTGTTGGGTCCGGGCCGCCGGCTTCGCCCTCGCCATGGCGCCCTGGCTGCA

DNA GCCGCCAGCTTTCAGGCCTGGGGGCCGCCGTCCCCGCAGCTGCTGGCGCCCACCCGCT

TCGCGCTGGAGATGTTCAACCGCGGCCGGGCTGCGGGGACGCCGGCCGTGCTGGGCCT

SBqileriCeTGTGCGCGACCGTCCGCGCCTCACCTACTCCTCTCTCCAGGCGGGCCAGGGGTCGCTG

TACTCCCTGGAGGCCACCCTGGAGGAGCCACCCTGCAACGACCCCATGGTGTGCCGGC

TCCCCGTGTCCAAGAAAACCCTGGTGACTTTCAAAGTCCTGGATGAGCTCGGGGGGCG

CGTGCTGCTGCGGAAGGACTGTGGCCCAGTGGACACCAAGGTTCCAGGTGCTGGGGAG

CCCAAGTCAGCCTTCACTCAGGGCTCAGCCATGATTTCTTCTCTGTCCCAAAACCATC

CAGACAACAGAAACGAGACTTTCAGCTCAGTCATTTCCCTGTTGAATGAGGATCCCCT

GTCCCAGGACTTGCCTGTGAAGATGGCTTCAATCTTCAAGAACTTTGTCATTACCTAT

AACCGGACATATGAGTCAAAGGAAGAAGCCCGGTGGCGCCTGTCCGTCTTTGTCAATA

ACATGGTGCGAGCACAGAAGATCCAGGCCCTGGACCGTGGCACAGCTCAGTATGGAGT

CACCAAGTTCAGTGATCTCACAGAGGAGGAGTTCCGCACTATCTACCTGAATACTCTC

CTGAGAAAAGAGCCTGGCAACAAGATGAAGCAAGCCAAGTCTGTGGGTGACCTCGCCC

CACCTGAATGGGACTGGAGGAGTAAGGGGGCTGTCACAAAAGTCAAAGACCAGGGCAT

GTGTGGCTCCTGCTGGGCCTTCTCAGTCACAGGCAATGTGGAGGGCCAGTGGTTTCTC

AACCAGGGGACCCTGCTCTCCCTCTCTGAACAGGAGCTCTTGGACTGTGACAAGATGG

ACAAGGCCTGCATGGGCGGCTTGCCCTCCAATGCCTACTCGGCCATAAAGAATTTGGG

AGGGCTGGAGACAGAGGATGACTACAGCTACCAGGGTCACATGCAGTCCTGCAACTTC

TCAGCAGAGAAGGCCAAGGTCTACATCAATGACTCCGTGGAGCTGAGCCAGAACGAGC

AGGAGCTGGCAGCCTGGCTGGCCAAGAGAGGCCCAATCTCCGTGGCCATCAATGCCTT

TGGCATGCAGTTTTACCGCCACGGGATCTCCCGCCCTCTCCGGCCCCTCTGCAGCCCT

TGCGTCATTGACCATGCGGTGTTGCTTGTGGGCTACGGAACCGTGAGTTCTGACGTTC

CCTTTTGGGCCATCAAGAACAGCTGGGGCACTGACTGGGGTGAGAAGGGTTACTACTA

CTTGCATCGCGGGTCCGGGGCATGTGGCGTGAACACCATGGCCAGCTCGGCGGTGGTG

GACTGAAGAGGGGCCCCCAGCTCGGGACCTGGTGCTGATCAGAGTGGCTGCTGCCCCA

GCCTGACATGTGTCCAGGCCCCTCCCCGGGAGGTACAGCTGGCAG

ORF Start: ATG at 42 ORF Stop:
TGA at 1512 SEQ ID NO: 32 490 as MW at 53794.7kD

NOV6a, MAPWLQLLSLLGLLPGAVAAPAQPRAASFQAWGPPSPQLLAPTRFALEMFNRGRAAGT

CG101836-O1~~'GLVRDRPRLTYSSLQAGQGSLYSLEATLEEPPCNDPMVCRLPVSKKTLVTFKVL

P1'OtelriDELGGRVLLRKDCGPVDTKVPGAGEPKSAFTQGSAMISSLSQNHPDNRNETFSSVISL

LNEDPLSQDLPVKMASIFKNFVITYNRTYESKEEARWRLSVFVNNMVRAQKIQALDRG

S8qi1C11CeTAQYGVTKFSDLTEEEFRTIYLNTLLRKEPGNKMKQAKSVGDLAPPEWDWRSKGAVTK

VKDQGMCGSCWAFSVTGNVEGQWFLNQGTLLSLSEQELLDCDKMDKACMGGLPSNAYS

AIKNLGGLETEDDYSYQGHMQSCNFSAEKAKVYINDSVELSQNEQELAAWLAKRGPIS

VAINAFGMQFYRHGISRPLRPLCSPCVIDHAVLLVGYGTVSSDVPFWATKNSWGTDWG

EKGYYYLHRGSGACGVNTMASSAVVD
~ ~~

~ EQ ID NO: 33 ~ ~~ 1226 by NOV6b, GCTTCGCCCTCGCCATGGCGCCCTGGCTGCAGCTCCTGTCGCTGCTGGGGCTGCTCCC

DNA CTGCTGCGGAAGGACTGTGGCCCAGTGGACACCAAGGTTCCAGGTGCTGGGGAGCCCA

AGTCAGCCTTCACTCAGGGCTCAGCCATGATTTCTTCTCTGTCCCAAAACCATCCAGA

S8C~11eriCeCAACAGAAACGAGACTTTCAGCTCAGTCATTTCCCTGTTGAATGAGGATCCCCTGTCC

CAGGACTTGCCTGTGAAGATGGCTTCAATCTTCAAGAACTTTGTCATTACCTATAACC

GGACATATGAGTCAAAGGAAGAAGCCCGGTGGCGCCTGTCCGTCTTTGTCAATAACAT

GGTGCGAGCACAGAAGATCCAGGCCCTGGACCGTGGCACAGCTCAGTATGGAGTCACC

AAGTTCAGTGATCTCACAGAGGAGGAGTTCCGCACTATCTACCTGAATACTCTCCTGA

GGAAAGAGCCTGGCAACAAGATGAAGCAAGCCAAGTCTGTGGGTGACCTCGCCCCACC

TGAATGGGACTGGAGGAGTAAGGGGGCTGTCACAAAAGTCAAAGACCAGGGCATGTGT

GGCTCCTGCTGGGCCTTCTCAGTCACAGGCAATGTGGAGGGCCAGTGGTTTCTCAACC

AGGGGACCCTGCTCTCCCTCTCTGAACAGGAGCTCTTGGACTGTGACAAGATGGACAA

GGCCTGCATGGGCGGCTTGCCCTCCAATGCCTACTCGGCCATAAAGAATTTGGGAGGG

CTGGAGACAGAGGATGACTACAGCTACCAGGGTCACATGCAGTCCTGCAACTTCTCAG

CAGAGAAGGCCAAGGTCTACATCAATGACTCCGTGGAGCTGAGCCAGAACGAGCAGAA
GCTGGCAGCCTGGCTGGCCAAGAGAGGCCCAATCTCCGTGGCCATCAATGCCTTTGGC
ATGCAGTTTTACCGCCACGGGATCTCCCGCCCTCTCCGGCCCCTCTGCAGCCCTTGGC
TCATTGACCATGCGGTGTTGCTTGTGGGCTACGGCAACCGCTCTGACGTTCCCTTTTG
GGCCATCAAGAACAGCTGGGGCACTGACTGGGGTGAGAAGGGTTACTACTACTTGCAT
CGCGGGTCCGGGGCCTGTGGCGTGAACACCATGGCCAGCTCGGCGGTGGTGGACTGA_A
GAGGGGCC
ORF Start: ATG at 15 ORF Stop: TGA at 1215 SEQ.ID NO: 34 400 as MW at 44237.8kD
NOV6b, MAPWLQLLSLLGLLPGAVAAPAQPQVLDELGRHVLLRKDCGPVDTKVPGAGEPKSAFT

PT'Oteln ~EARWRLSVFVNNMVRAQKIQALDRGTAQYGVTKFSDLTEEEFRTIYLNTLLRKEPG
NKMKQAKSVGDLAPPEWDWRSKGAVTKVKDQGMCGSCWAFSVTGNVEGQWFLNQGTLL
SeqllenCe SLSEQELLDCDKMDKACMGGLPSNAYSAIKNLGGLETEDDYSYQGHMQSCNFSAEKAK
VYINDSVELSQNEQKLAAWLAKRGPISVAINAFGMQFYRHGISRPLRPLCSPWLIDHA
VLLVGYGNRSDVPFWAIKNSWGTDWGEKGYYYLHRGSGACGVNTMASSAVVD
Sequence comparison of the above protein sequences yields the following sequence relationships shown in Table 6B.
Table 6B. Comparison of NOV6a against NOV6b.
Protein Sequence NOV6a Residues/ Identities/
Match Residues Similarities for the Matched Region NOV6b 117..490 366/374 (97%) 28..400 368/3?4 (9?%) Further analysis of the NOV6a protein yielded the following properties shown in Table 6C.
Table 6C. Protein Sequence Properties NOV6a PSort 0.4514 probability located in outside; 0.1900 probability located in lysosome analysis: (lumen); 0.1000 probability located in endoplasmic reticulum (membrane);
0.1000 probability located in endoplasmic reticulum (lumen) SignaIP Cleavage site between residues 20 and 21 analysis:
A search of the NOV6a protein against the Geneseq database, a proprietary database that contains sequences published in patents and patent publication, yielded several homologous proteins shown in Table 6D.

Table 6D. Geneseq Results for NOV6a NOV6a Identities/

Geneseq Protein/Organism/Length Residues/SimilaritiesExpect [Patent for Identifier#, Date) Match the MatchedValue ResiduesRegion AAB 11960Human cathepsin Y - Homo1..490 469/491 0.0 (95%) sapiens, 484 aa. [JP2000157263-A,1..484 476/491 (96%) 13-JUN-2000]

AAW53200 Human spleen-derived 99..490 381/393 0.0 cysteine (96%) protease - Homo sapiens,1..392 386/393 392 aa. (97%) [JP10099084-A, 21-APR-1998]

AAW37957 Amino acid sequence of 99..490 378/393 0.0 human ' (96t) cathepsin polypeptide-1 1..392 384/393 - Homo (97%) Sapiens, 392 aa. [W09813484-Al, 02-APR-1998]

AAY45041 Human Apop2 protein - 152..490333/339 0.0 Homo (98%) Sapiens, 338 aa. [W0200007545-1..338 335/339 (98%) A2, 17-FEB-2000]

AAB51802 Gene 26 human secreted 256..490229/235 e-135 protein (97%) homologous amino acid 1..234 231/235 sequence (97%) #131 - Homo Sapiens, 234 aa.

[W0200061625-A1, 19-OCT-2000]

In a BLAST search of public sequence datbases, the NOV6a protein was found to have homology to the proteins shown in the BLASTP data in Table 6E.

Table 6E. Public BLASTP Results for NOV6a Protein NOV6a Identities) AccessionProtein/Organism/LengthResidues/Similarities Expect for Number Match the Matched Value ResiduesPortion Q9UBX1 Cathepsin F precursor 1..490 469/491 (95%)0.0 (EC

3.4.22.41) (CATSF) - 1..484 476/491 (96%) Homo sapiens (Human), 484 aa.

Q9R013 Cathepsin F precursor 1..490 350/493 (70%)0.0 (EC

3.4.22.41) - Mus musculus1..462 401/493 (80%) (Mouse), 462 aa.

Q9ES93 CATHEPSIN F - Mus musculusI ..490 348/493 (70%)0.0 (Mouse), 462 aa. 1..462 399/493 (80%) T46294 hypothetical protein 166..444276/279 (98%)~ e-161 DI~FZp434F0610.1 - human,I ..279 278/279 (98%) 308 ' as (fragment).

Q99KQ9 SIMILAR TO CATHEPSIN 188..490249/303 (82%)e-153 F -Mus musculus (Mouse), 1..302 281/303 (92%) 302 aa.

PFam analysis predicts that the NOV6a protein contains the domains shown in the Table 6F.
Table 6F. Domain Analysis of NOV6a Identities/
Pfam Domain NOV6a Match Region Similarities Expect Value for the Matched Region gpdh 404..413 5/10 (50%) 0.35
8/10 (80%) Peptidase C1 276..488 102/337 (30%) 1.8e-104 1841337 (55%) E~PAMI'LE 7.
The NOV7 clone was analyzed, and the nucleotide and encoded polypeptide sequences are shown in Table 7A.

DNA TGGGTGACCACAGATGAGGGCCCCACCTGGGAGTTCTACGCCTGCCAGCCCAAGGTGA
Sequence TGCGCCTGAAGGACTACGTCAAGGTGAAGGTGGAGCCCTCAGGCATCACATGTGGAGA
CCCCCCTGAGAGGTTCTGCTCCCATCCCTACCTATGCAGCAACGAGTGTGACGCCTCC
AACCCGGACCTGGCCCACCCGCCCAGGCTCATGTTCGACAAGGAGGAGGAGGGCCTGG
CCACCTACTGGCAGAGCATCACCTGGAGCCGCTACCCCAGCCCGCTGGAAGCCAACAT
CACCCTTTCGTGGAACAAGACCGTGGAGCTGACCGACGACGTGGTGATGACCTTCGAG
TACGGCCGGCCCACGGTCATGGTCCTGGAGAAGTCCCTGGACAACGGGCGCACCTGGC
AGCCCTACCAGTTCTACGCCGAGGACTGCATGGAGGCCTTCGGTATGTCCGCCCGCCG
GGCCCGCGACATGTCATCCTCCAGCGCGCACCGCGTGCTCTGCACCGAGGAGTACTCG
CGCTGGGCAGGCTCCAAGAAGGAGAAGCACGTGCGCTTCGAGGTGCGGGACCGCTTCG
CCATCTTTGCCGGCCCCGACCTGCGCAACATGGACAACCTCTACACGCGGCTGGAGAG
CGCCAAGGGCCTCAAGGAGTTCTTCACCCTCACCGACCTGCGCATGCGGCTGCTGCGC
CCGGCGCTGGGCGGCACCTATGTGCAGCGGGAGAACCTCTACAAGTACTTCTACGCCA
TCTCCAACATCGAGGTCATCGGCAGGTGCAAGTGCAACCTGCACGCCAACCTGTGCTC
CATGCGCGAGGGCAGCCTGCAGTGCGAGTGCGAGCACAACACCACCGGCCCCGACTGC
GGCAAGTGCAAGAAGAATTTCCGCACCCGGTCCTGGCGGGCCGGCTCCTACCTGCCGC
TGCCCCATGGCTCTCCCAACGCCTGTGACTGCGAATGCTACGGTCACTCCAACCGCTG
CAGCTACATTGACTTCCTGAATGTGGTGACCTGCGTCAGCTGCAAGCACAACACGCGA
GGTCAGCACTGCCAGCACTGCCGGCTGGGCTACTACCGCAACGGCTCGGCAGAGCTGG
ATGATGAGAACGTCTGCATTGAGTGTAACTGCAACCAGATAGGCTCCGTGCACGACCG
GTGCAACGAGACCGGCTTCTGCGAGTGCCGCGAGGGCGCGGCGGGCCCCAAGTGCGAC
GACTGCCTCCCCACGCACTACTGGCGCCAGGGCTGCTACCCCAACGTGTGCGACGACG
GGCGGTCTGGACTGCGACCGCGCGCCCGGGGCCGCCCCGCGCCCCGCCACCCTGCTCG
GCTGCCTGCTGCTGCTGGGGCTGGCCGCCCGCCTGGGCCGCTGAGCCCCGCCCGGAGG
ORF Start: ATG at 103 ORF Stop: TGA at 1666 ~SEQ~ID~N0:~~36~~~ 521 as MW at 58964.O1cD
NOV7a, MLHLLALFLHCLPLASGDYDICKSWVTTDEGPTWEFYACQPKVMRLKDYVKVKVEPSG

PPOteln LEANITLSWNKTVELTDDVVMTFEYGRPTVMVLEKSLDNGRTWQPYQFYAEDCMEAFG
MSARRARDMSSSSAHRVLCTEEYSRWAGSKKEKHVRFEVRDRFAIFAGPDLRNMDNLY
Sequence TRLESAKGLKEFFTLTDLRMRLLRPALGGTYVQRENLYKYFYAISNIEVIGRCKCNLH
ANLCSMREGSLQCECEHNTTGPDCGKCKKNFRTRSWRAGSYLPLPHGSPNACDCECYG
HSNRCSYIDFLNWTCVSCKHNTRGQHCQHCRLGYYRNGSAELDDENVCIECNCNQIG
SVHDRCNETGFCECREGAAGPKCDDCLPTHYWRQGCYPNVCDDDQLLCQNGGTCLQNQ
Further analysis of the NOV7a protein yielded the following properties shown in Table 7B.
Table 7B. Protein Sequence Properties NOV7a PSort 0.7000 probability located in plasma membrane; 0.3000 probability located in analysis: microbody (peroxisome); 0.2000 probability located in endoplasmic reticulum (membrane); 0.1000 probability located in mitochondrial inner membrane SignalP Cleavage site between residues 18 and 19 analysis:

A search of the NOV7a protein against the Geneseq database, a proprietary database that contains sequences published in patents and patent publication, yielded several homologous proteins shown in Table 7C.
Table 7C. Geneseq Results for NOV7a NOV7a Identities/

Geneseq Protein/Organism/Length Residues/SimilaritiesExpect [Patent ' for Identifier#, Date] Match the Matched Value ResiduesRegion ABB53284 Human polypeptide #24 1..521 5211533 (97%)0.0 - Homo Sapiens, 533 aa. [W0200181363-1..533 521/533 (97%) A1, O1-NOV-2001] ~

ABB05418 Mouse membrane bound 17..519 308/515 (59%)0.0 type netrin ~

protein SEQ ID NO:8 - 28..537 379/515 (72%) Mus musculus, 539 aa. [JP2001327289-A, 27-NOV-2001 ABB53283 Human polypeptide #23 1..284 284/286 (99%)e-170 - Homo Sapiens, 286 aa. [W0200181363-1..286 284/286 (99%) A1, O1-NOV-2001]

ABB05419 Mouse membrane bound 17..427 220/438 (50%)e-124 type netrin protein SEQ ID NO:10 28..461 277/438 (63%) - Mus ~

musculus, 483 aa., []P2001327289-' A, 27-NOV-2001]

AAB65181 Human PR01133 (LTNQ571) 13..427 210/419 (50%)e-123 protein sequence SEQ 24..416 274/419 (65%) ID N0:129 -Homo Sapiens, 438 aa.

[WO200073454-Al, 07-DEC-2000]

In a BLAST search of public sequence datbases, the NOV7a protein was found to have homology to the proteins shown in the BLASTP data in Table 7D.

Table 7D. Public BLASTP Results for NOV7a Protein NOV7a Identities/

AccessionProtein/Organism/LengthResidues/SimilaritiesExpect for Number Match the Matched Value Residues Portion BAB47486 KI:AA1857 PROTEIN - 1..521 520/530 (98%)0.0 Homo sapiens (Human), 549 20..549 521/530 (98%) as (fragment).

Q96CW9 HYPOTHETICAL 59.8 KDA 1..521 520/530 (98%)0.0 PROTEIN - Homo Sapiens1..530 521/530 (98%) (Human), 530 aa.

Q8VIP8 NETRIN-G2A - Mus musculus1..519 493/529 (93%)0.0 (Mouse), 530 aa. ~ 505/529 (95%) I ..528 AAL84788 LAMINET-2A - Mus musculus1..519 492/529 (93%)0.0 domesticus (western 1..528 5041529 (95%) European house mouse), 530 aa.

Q96JH0 I~IAA1857 PROTEIN - 1..342 342/344 (99%)0.0 Homo Sapiens (Human), 438 20..363 342/344 (99%) as (fragment).

PFam analysis predicts that the NOV7a protein contains the domains shown in the Table 7E.
Table 7E. Domain Analysis of NOV7a Identities/

Pfam DomainNOV7a Match RegionSimilarities Expect Value for the Matched Region laminin_Nterm39..283 79/282 (28%) 5.9e-12 134/282 (48%) laminin_EGF285..342 15/68 (22%) 1.5e-06 .

38/68 (56%) laminin_EGF344..397 18/63 (29%) 0.00013 ~ 39/63 (62%) laminin_EGF400..442 20/59 (34%) 8.3e-09 35/59 (59%) EGF 447..477 16/47 (34%) 0.00014 22/47 (47%) EXAMPLE 8.
The NOV8 clone was analyzed, and the nucleotide and encoded polypeptide sequences are shown in Table 8A.
Table 8A. NOV8 SEQ ID NO: 37 X910 by NOVga, GGTCCGGGGGGGCTGCCGGTCCCGGGTACCATGTGTGACGGCGCCCTGCTGCCTCCGC
CG102325-OlTCGTCCTGCCCGTGCTGCTGCTGCTGGTTTGGGGACTGGACCCGGGCACAGCTGTCGG
DNA CGACGCGGCGGCCGACGTGGAGGTGGTGCTCCCGTGGCGGGTGCGCCCCGACGACGTG
CACCTGCCGCCGCTGCCCGCAGCCCCCGGGCCCCGACGGCGGCGACGCCCCCGCACGC
Sequence CCCCAGCCGCCCCGCGCGCCCGGCCCGGAGAGCGCGCCCTGCTGCTGCACCTGCCGGC
CTTCGGGCGCGACCTGTACCTTCAGCTGCGCCGCGACCTGCGCTTCCTGTCCCGAGGC
TCTACTCGGGCCGTGTGCTCGGCCACCCCGGCTCCCTCGTCTCGCTCAGCGCCTGCGG
CGCCGCCGGCGGCCTGGTACTGCCCGCGCCACCTCCGGGTCGGCCCGTCCGGTCTGTT
GCGACGCAGAGTGGTCGCCGTGGAGGGTGGGGGTGGGGCGCCTCTGCTGGAAGTCCAG
CCTCCAGGGGAACCGGAGGGAACCCCCTGCCTTTCCACCTCTCCCCATCCCCCACCCC
GGCCTTCGGTACCCTCTATAGGCAAAGGGGGTGGGAGGGGCAGCATCCCAGTCCAGCG
CCTCTGCAGCCCGTGGAACCCGCGCGGAGCTGGGGTTGCGTGGGGGTATACGCCGCCC
GCTCTAGGGAGCGCAGATCTGGCAGGGATGAAACTGTCAGGGCCCTGGACAGAGGCGC
CTTGGCCCCAATGTAGAGAACACTGCATCTGCACCGCCGTGTCAAAGTGTATGTCACG
GGAGTACCTGTGTACGTGTAGGTGTTATGTTCTTGGACTT
ORF Start: ATG at 31 OLtF Stop: TAG at 826 SEQ ID NO: 38 265 a~MW at 28223.O1cD
NOVBa, MCDGALLPPLVLPVLLLLVWGLDPGTAVGDAAADVEWLPWRVRPDDVHLPPLPAAPG
CG102325-OlPRRRRRPRTPPAAPRARPGERALLLHLPAFGRDLYLQLRRDLRFLSRGFEVEEAGAAR
PrOteln RRGRPAELCFYSGRVLGHPGSLVSLSACGAAGGLVLPAPPPGRPVRSVATQSGRRGGW
GWGASAGSPASRGTGGNPLPFHLSPSPTPAFGTLYRQRGWEGQHPSPAPLQPVEPARS
Sequence WGCVGWAARSRERRSGRDETVRALDRGALAPM
Further analysis of the NOV8a protein yielded the following properties shown in Table 8B.
Table 8B. Protein Sequence Properties NOVBa PSort 0.8200 probability located in outside; 0.1000 probability located in analysis: endoplasmic reticulum (membrane); 0.1000 probability located in endoplasmic reticulum (lumen); 0.1000 probability located in lysosome (lumen) SignalP Cleavage site between residues 28 and 29 analysis:
A search of the NOV 8a protein against the Geneseq database, a proprietary database that contains sequences published in patents and patent publication, yielded several homologous proteins shown in Table 8C.

Table 8C. Geneseq Results for NOVBa NOVBa Identities/

Geneseq Protein/Organism/Length Residues/SimilaritiesExpect for Identifier[Patent #, Date] Match the Matched Value ResiduesRegion AAE10350 Human ADAMTS-J1.4 variant1..151 151/151 (100%)6e-85 protein - Homo Sapiens, 1..151 151/151 (100%) 891 aa.

[EP1134286-A2, 19-SEP-2001]

AAE10348 Human ADAMTS-J1.2 variant1..151 151/151 (100%)6e-85 protein - Homo Sapiens, 1..151 151/151 (100%) 635 aa.

[EP1134286-A2, 19-SEP-2001]

AAE10347 Human ADAMTS-JI.1 variant1..151 151/151 (100%)6e-85 protein - Homo Sapiens, 1..151 151/151 (100%) 745 aa.

[EP1134286-A2, 19-SEP-2001]

AAU72894 Human metalloprotease 27..151 125/125 (100%)1e-68 partial protein sequence #6 - 434:.558125/125 (100%) Homo Sapiens, 1428 aa. [W0200183782-A2, 08-NOV-2001]

AAU72900 Human metalloprotease 51..151 52/112 (46%)3e-15 partial protein sequence #12 142..24459/112 (52%) - Homo Sapiens, 1094 aa. [W0200183782- .
, A2, 08-NOV-2001]

In a BLAST search of public sequence datbases, the NOV8a protein was found to have homology to the proteins shown in the BLASTP data in Table 8D.

Table 8D. Public BLASTP Results for NOVBa NOVBa Identities/
Protein Residues/SimilaritiesExpect AccessionProteiia/Organism/Length for Match the MatchedValue Number ' ResiduesPortion CAC86016METALLOPROTEASE 1..151 151/151 1e-84 (100%) DIS1NTEGRIN 17, WITH 1..151 151/151 (100%) THROMBOSPONDIN DOMAINS

- Homo Sapiens (Human), 1095 aa.

CPrC84565ADAMTS-19 - Homo Sapiens 51..151 51/112 (45%)1e-14 (Human), 1207 aa. 142..24459/112 (52%) CAC86014METALLOPROTEASE 25..259 72/248 (29%)Se-08 DISINTEGRIN I S WITH 13..218 100/248 (40%) THROMBOSPONDIN DOMAINS

- Homo sapiens (Human), 950 aa.

Q9WUQ1 ADAMTS-1 precursor (EC 69..149 35/90 (38%)1e-06 3.4.24.-) ~

(A disintegrin and metalloproteinase66..153 46/90 (50%) with thrombospondin motifs 1 ) (ADAM-TS 1 ) (ADAM-TS
I ) -Rattus norvegicus (Rat), 967 aa.
i Q9UP79 ADAMTS-8 precursor (EC 52..187 52/167 (31%)3e-06 3.4.24.-) (A disintegrin and metalloproteinase3..165 65/167 (38%) ~
with thrombospondin motifs 8) (ADAM-TS 8) (ADAM-TS8) (METH-2) (METH-8) - Homo Sapiens (Human), 890 aa.

PFam analysis predicts that the NOVBa protein contains the domains shown in the Table 8E.
Table 8E. Domain Analysis of NOV8a Identities/
Pfam Domain NOVBa Match Region Similarities Expect Value for the Matched Region Pep Ml2B~ropep ~ 95..192 261119 (22%) 0.021 601119 (50%) EXAMPLE 9.
The NOV9 clone was analyzed, and the nucleotide and encoded polypeptide sequences are shown in Table 9A.
Table 9A. NOV9 Sequence Analysis SEQ ID NO: 39 958 by NOV9a, GCAGCACCCGCAGCCAGAGCCGCGCTCGGCATGATGCCCGGGGCGCCGCTCCTGCGGC

DNA '~TGCCCCCCACCACGGGGGACGCCACCCTGGCCTTCGTCTTCGACGTCACCGGCTCC
ATGTGGGACGAACTGATGCAGGTGATCGATGGCGCCTCGCGCATTCTGGAACGCAGTC
Sequence TGAGCCGCCGCAGCCAGGCCATCGCCAACTACGCGCTGGTGCCCTTCCACGACCCAGA
TATTGGCCCAGTGACCCTCACGGCGGACCCCACAGTGTTTCAGAGGGAGCTGAGAGAA
CTCTACGTGCAGGGAGGTGGTGACTGCCCGGAGATGAGTGTGGGGGCCATTAAGGCTG
CCGTGGAGGTTGCCAACCCCGGATCCTTCATCTACGTCTTTTCGGATGCCCGCGCCAA
AGACTATCACAAGAAGGAAGAGCTGCTGCGGCTCCTGCAGCTCAAGCAATCACAGGTG
GTCTTTGTGCTGACGGGGGACTGTGGCGACCGCACCCATCCTGGCTACCTGGCTTATG
AGGAGATCGCTGCCACCAGCTCTGGGCAGGTGTTCCACCTGGACAAGCAGCAAGTGAC
AGAGGTGCTGAAGTGGGTGGAGTCAGCGATCCAGGCCTCCAAGGTGCACCTGCTGTCC
ACAGACCACGAGGAGGAGGGGGAGCACACATGGAGACTCCCCTTTGACCCCAGCCTGA
AGGAGGTCACCATCTCATTGAGTGGGCCAGGGCCTGAGATTGAAGTCCAAGATCCGCT
GGGTATGGACCACCCCGGGGCTGGCCTCCTCTTTGGCCCCAAGACTGAGGTGGAAGCC
CAGGATGGGACAAAGAAAGAGACCAAGGGTGACAGGGCTTCAGACATGAGGCTCCAGG
AATAGGGAAATATGGGGTGGGGGGGACACG
ORF Start: ATG at 31 ORF Stop: TAG at 931 SEQ ID NO: 40 300 as MW at 32481.S1cD
NOV9a, MMPGAPLLRLLTAVSAAVAVAVAGAPGTVMPPTTGDATLAFVFDVTGSMWDELMQVID

PrOteln EMSVGAIKAAVEVANPGSFIWFSDARAKDYHKKEELLRLLQLKQSQWFVLTGDCGD
RTHPGYLAYEEIAATSSGQVFHLDKQQVTEVLKWVESAIQASKVHLLSTDHEEEGEHT
Sequence WRLPFDPSLKEVTISLSGPGPEIEVQDPLGMDHPGAGLLFGPKTEVEAQDGTKKETKG
DRASDMRLQE
SEQ ID NO: 41 2916 by NOV9b, GCAGCACCCGCAGCCAGAGCCGCGCTCGGCATGATGCCCGGGGCGCCGCTCCTGCGGC

DNA ~TGCCCCCCACCACGGGGGACGCCACCCTGGCCTTCGTCTTCGACGTCACCGGCTCC
ATGTGGGACGAACTGATGCAGGTGATCGATGGCGCCTCGCGCATTCTGGAACGCAGTC
Sequence TGAGCCGCCGCAGCCAGGCCATCGCCAACTACGCGCTGGTGCCCTTCCACGACCCAGA
TATTGGCCCAGTGACCCTCACGGCGGACCCCACAGTGTTTCAGAGGGAGCTGAGAGAA
CTCTACGTGCAGGGAGGTGGTGACTGCCCGGAGATGAGTGTGGGGGCCATTAAGGCTG
CCGTGGAGGTTGCCAACCCCGGATCCTTCATCTACGTCTTTTCGGATGCCCGCGCCAA
AGACTATCACAAGAAGGAAGAGCTGCTGCGGCTCCTGCAGCTCAAGCAATCACAGGTG
GTCTTTGTGCTGACGGGGGACTGTGGCGACCGCACCCATCCTGGCTACCTGGCTTATG
AGGAGATCGCTGCCACCAGCTCTGGGCAGGTGTTCCACCTGGACAAGCAGCAAGTGAC
AGAGGCAGGTGCTTCCGTGTTTCCAGGCAAAATTGTGCAGGAGCACAGGATCCTTTCA
GGGGCCAGCTGGGAAATGATGAACAACGCTCTCTCTGGAAAGGACAAGCACACCCATT
TCCGTGGTATAAATGCTCCCACCTCGGCTGATTCCAAGTCAGAGTTGGGAAGTGACGC
TGACACTCAGCTTTCCGGAGCCTACACAAGTGGCTCCCACACACCACTGGATCCCGCA
CAGGCACCTCTCACCGCCAGTTGGGTTAACGAGAGCCCCTACCTGGTGCTGAAGTGGG
TGGAGTCAGCGATCCAGGCCTCCAAGGTGCACCTGCTGTCCACAGACCACGAGGAGGA
GGGGGAGCACACATGGAGACTCCCCTTTGACCCCAGCCTGAAGGAGGTCACCATCTCA
TTGAGTGGGCCAGGGCCTGAGATTGAAGTCCAAGATCCGCTGGGTATGGACCACCCCG
GGGCTGGCCTCCTCTTTGGCCCCAAGACTGAGGTGGAAGCCCAGGATGGGACAAAGAA
AGAGACCAAGGGGAGGATCCTGCAGGAGGACGAGGGCCTCAACGTGCTTCTCAACATC

CCTGACTCGGCCAAGGTCGTAGCCTTTAAGCCTGAGCATCCGGGGCTGTGGTCCATCA

AGGTCTATAGCAGTGGCCGCCATTCAGTGAGGATCACAGGCGTCAGCAACATTGACTT

CCGAGCCGGCTTCTCCACTCAGCCCTTGCTGGACCTCAACCACACCCTCGAGTGGCCC

TTGCAAGGAGTCCCCATCTCCCTGGTGATCAATTCCACGGGCCTGAAGGCACCCGGCC

GCCTAGACTCGGTGGAGCTGGCACAAAGCTCAGGGAAGCCCCTCCTGACTCTGCCCAC

GAAGCCCCTCTCCAATGGCTCCACCCATCAGCTGTGGGGCGGGCCGCCCTTCCACACC

CCCAAGGAGCGCTTCTACCTCAAGGTGAAGGGCAAGGACCATGAGGGAAACCCCCTCC

TTCGTGTCTCTGGAGTGTCCTACAGTGGGGTGGCCCCAGGCGCTCCCCTCGTCAGCAT

GGCCCCCAGGATCCATGGCTACCTGCACCAGCCCCTGCTGGTCTCCTGCTCGGTGCAC

AGTGCCCTTCCCTTCCGGCTGCAGCTGCGGCGAGGTGAAGCCAGGCTGGGCGAAGAGA

GGCACTTTCAGGAGTCGGGAAACAGCAGCTGGGAGATCCTGCGGGCCTCCAAGGCCGA

GGAGGGCACGTACGAGTGCACAGCCGTCAGCAGGGCTGGGACCGGGCGAGCAAAGGCC

CAGATTGTTGTCACCCTGCACCTCAGGGTGGGGTTCGGGGCAGCACCAGGGCTTGCAC

GAAGACCCCCTCCCTTGCCTCAGCTCCTTGGTTCCTCCTGTGCTCATGTCCCTGCAGA

CCCCCCGCCGCAGCTGGTCCCTGCTCCCAACGTGACCGTGTCCCCAGGGGAGACTGCC

GTCCTATCCTGCCGGGTCCTAGGCGAGGCCCCCTACAACCTGACGTGGGTCCGGGACT

GGCGAGTCCTGCCGGCCTCGACGGGCCGAGTTGCCCAGCTGGCTGACCTGTCCCTGGA

GATCAGTGGCATCATCCCCACAGACGGCGGGAGGTACCAGTGTGTGGCCAGCAATGCC

AATGGGGTCACAAGGGCATCCGTCTGGCTCCTGGTGCGAGAGGCCCCACAGGTCAGCA

TCCACACCAGCTCCCAGCACTTCTCCCAAGGTGTGGAGGTGAAGGTCAGCTGCTCAGC

CTCTGGATACCCCACACCCCACATCTCCTGGAGCCGTGAGAGCCAAGCCCTACAAGAG

GACAGCAGAATCCATGTGGACGCACAGGGAACCCTGATTATTCAGGGGGTAGCCCCAG

AGGATGCTGGGAATTACAGCTGCCAGGCGACTAATGAGGTTGGCACTGACCAGGAGAC

GGTCACCCTCTACTACACAGACCCACCGTCGGTCTCTGCTGTAAATGCCGTGGTGCTG

GTGGCCGTTGGGGAGGAGGCTGTGTTGGTGTGTGAGGCATCTGGGGTTCCCCCGCCCC

GAGTCATCTGGTATCGAGGGGGTCTTGAAATGATCCTGGCCCCTGAGGGCTCCAGCTC

TGGGAAGCTGCGGATCCCGGCGGCTCAGGAGAGGGATGCTGGCACCTACACCTGCCGG

GCTGTCAATGAGTTGGGTGACGCCTCTGCAGAAATCCAGCTGGCGGTTGGACATGCGC

CCCAGCTGACGGAGCTGCCCCGGGATGTCACTGTGGAACTGGGGAGGAGTGCCCAGCT

GCGGCGTGGGACTTAA

ORF Start: ATG at 31 ORF Stop: TAA
at 2914 SEQ ID NO: 42 961 as MW at 102789.2kD

NOV9b, MMPGAPLLRLLTAVSAAVAVAVAGAPGTVMPPTTGDATLAFVFDWGSMWDELMQVID

PTOteIri EMSVGAIKAAVEVANPGSF~YVFSDARAKDYHKKEELLRLLQLKQSQWFVLTGDCGD

RTHPGYLAYEEIAATSSGQVFHLDKQQVTEAGASVFPGKIVQEHRILSGASWEMMNNA
SeCILIeriCeL

SGKDKHTHFRGINAPTSADSKSELGSDADTQLSGAYTSGSHTPLDPAQAPLTASWVN

ESPYLVLKWVESAIQASKVHLLSTDHEEEGEHTWRLPFDPSLKEVTISLSGPGPEIEV

QDPLGMDHPGAGLLFGPKTEVEAQDGTKKETKGRILQEDEGLNVLLNIPDSAKWAFK

PEHPGLWSIKVYSSGRHSVRITGVSNIDFRAGFSTQPLLDLNHTLEWPLQGVPISLVI

NSTGLKAPGRLDSVELAQSSGKPLLTLPTKPLSNGSTHQLWGGPPFHTPKERFYLKVK

GKDHEGNPLLRVSGVSYSGVAPGAPLVSMAPRIHGYLHQPLLVSCSVHSALPFRLQLR

RGEARLGEERHFQESGNSSWEILRASKAEEGTYECTAVSRAGTGRAKAQIWTLHLRV

GFGAAPGLARRPPPLPQLLGSSCAHVPADPPPQLVPAPNVTVSPGETAVLSCRVLGEA

PYNLTWVRDWRVLPASTGRVAQLADLSLEISGIIPTDGGRYQCVASNANGVTRASVWL

LVREAPQVSIHTSSQHFSQGVEVKVSCSASGYPTPHISWSRESQALQEDSRIHVDAQG

TLIIQGVAPEDAGNYSCQATNEVGTDQETVTLYYTDPPSVSAVNAWLVAVGEEAVLV

CEASGVPPPRVIWYRGGLEMILAPEGSSSGKLRIPAAQERDAGTYTCRAVNELGDASA

EIQLAVGHAPQLTELPRDVTVELGRSAQLRRGT
' SEQ ID NO: 43 1023 by NOV9C, CTCGAGTGTGGAACTCACTCTTAACGTACCTGAGGAGTGTCCAACGTCTTTGGACAAG

DNA ATGGGGGACTCAGCAGTTGCCAAGGTCTGCAGCCTCCTCCAAGGGGTTCCCATCTAGT

TCTCAAGAGGAAGGAGGGGGTTCTCAGTCGCCAGGTGGGCATGGCACTCCCGAGGCCA

SeC1L18riCeGGTGAGCAGGTCAGTGCCTTGGGG

CTCAGGGCTGCTCCGGTTCTTACCGAATTGATCC

AGTCGTTGTAGTTGGAGACCCGCGTGAAGATGGAGGGCTTGTAGTAGTAGTTGCAACC

AAGGACCGACGTGAGGCTGCCGATGCCATGCACCTCCCACCGGCCGTCAGATGCCTGA

CAGTTCAGCGGCCCACCGGAGTCTCCGTTGCAGGTGCATATCACGCCATCACCCCCAG

CACAGATCATATTCGTCTTCACGGTGCTGCCCCACCAGCCAGAGTTGGAGCAGGTGGC
ATAGTCCACAACCAGCAACCGGCCCTGCTTCAGGTCATCAGGGAGAGCCCCGTTGGTC
TGCAGCCTTCCCCAGCCCGTGACGTAGCAGGGGTAGTTGTTGGGTAGAATGGTGCCGG
CAGGAGGGAGGCAGGCCAGCTGGATCTTGTCGGTGAGGGAGACGGGGTTAGCCAGTTT
GAGCAGGGCAATGTCGTTCCCTTTGGAGACCTGGTCGGAGTTCCAGTCCTTGTGCACC
ACAATCTTAGAGACACTGACGGCCAGCGAGCCGGACTCTGCAACGTAGAGGTTATGCT
GGCCCAGCATCACGCGGTAGATCCCGGAGGAGCTGATGCAGTGGGCAGCCGTCAGGAC
CCAGCTGTTGGCTATCAGGGACCCTCCGCAGGTGTGGTACCACTGGCCATTGGAGCTG
TACTGCAGGGAGACCTGCCAGGGCCGGCTGTTGGGCCTCGCTTCTTCACCTCCAAGCA
TCCTAGACATATCAGGCGCGTAAGTGGAGACGGATCC
ORF Start: at 628 ORF Stop: end of sequence SEQ ID NO: 44 132 as MW at 13513.O1eD
NOV9C' NGAGRREAGQLDLVGEGDGVSQFEQGNWPFGDLVGVPVLVHFiNLRDTDGQRAGLCNV
197195425 EVMI'p'QHHAVDPGGADAVGSRQDPAVGYQGPSAGWPLAIGAVLQGDLPGPAVGPRFF
PrOteln TSKHPRHIRRVSGDGS
Sequence SEQ ID NO: 45 2058 by NOV9d, AAGCTTGTGGCAGTGGCCGGGGCGCCCGGGACGGTAATGCCCCCCACCACGGGGGACG

DNA GATCGATGGCGCCTCGCGCATTCTGGAACGCAGTCTGAGCCGCCGCAGCCAGGCCATC
GCCAACTACGCGCTGGTGCCCTTCCACGACCCAGATATTGGCCCAGTGACCCTCACGG
Sequence CGGACCCCACAGTGTTTCAGAGGGAGCTGAGAGAACTCTACGTGCAGGGAGGTGGTGA
CTGCCCGGAGATGAGTGTGGGGGCCATTAAGGCTGCCGTGGAGGTTGCCAACCCCGGA
TCCTTCATCTACGTCTTTTCGGATGCCCGCGCCAAAGACTATCACAAGAAGGAAGAGC
TGCTGCGGCTCCTGCAGCTCAAGCAATCACAGGTGGTCTTTGTGCTGACGGGGGACTG
TGGCGACCACACCCATCCTGGCTACCTGGCTTATGAGGAGATCGCTGCCACCAGCTCT
GGGCAGGTGTTCCACCTGGACAAGCAGCAAGTGACAGAGGTGCTGAAGTGGGTGGAGT
CAGCGATCCAGGCCTCCAAGGTGCACCTGCTGTCCACAGACCACGAGGAGGAGGGGGA
GCACACATGGAGACTCCCCTTTGACCCCAGCCTGAAGGAGGTCACCATCTCATTGAGT
GGGCCAGGGCCTGAGATTGAAGTCCAAGATCCGCTGGGGAGGATCCTGCAGGAGGACG
AGGGCCTCAACGTGCTTCTCAACATCCCTGACTCGGCCAAGGTCGTAGCCTTTAAGCC
TGAGCATCCGGGGCTGTGGTCCATCAAGGTCTATAGCAGTGGCCGCCATTCAGTGAGG
ATCACAGGCGTCAGCAACATTGACTTCCGAGCCGGCTTCTCCACTCAGCCCTTGCTGG
ACCTCAACCACACCCTCGAGTGGCCCTTGCAAGGAGTCCCCATCTCCCTGGTGATCAA
TTCCACGGGCCTGAAGGCACCCGGCCGCCTAGACTCGGTGGAGCTGGCACAAAGCTCA
GGGAAGCCCCTCCTGACTCTGCCCACGAAGCCCCTCTCCAATGGCTCCACCCATCAGC
TGTGGGGCGGGCCACCCTTCCACACCCCCAAGGAGCGCTTCTACCTCAAGGTGAAGGG
CAAGGACCATGAGGGAAACCCCCTCCTTCGTGTCTCTGGAGTGTCCTACAGTGGGGTG
GCCCCAGGCGCTCCCCTCGTCAGCATGGTCCCCAGGATCCATGGCTACCTGCACCAGC
CCCTGCTGGTCTCCTGCTCGGTGCACAGTGCCCTTCCCTTCCGGCTGCAGCTGCGGCG
AGGTGAAGCCAGGCTGGGCGAAGAGAGGCACTTTCAGGAGTCGGGAAACAGTAGCTGG
GAGATCCTGCGGGCCTCCAAGGCCGAGGAGGGCACGTACGAGTGCACAGCCGTCAGCA
GGGCTGGGACCGGGCGAGCAAAGGCCCAGATTGTTGTCACAGACCCCCCGCCGCAGCT
GGTCCCTGCTCCCAACGTGACCGTGTCCCCAGGGGAGACTGCCGTCCTATCCTGCCGG
GTCCTAGGCGAGGCCCCCTACAACCTGACGTGGGTCCGGGACTGGCGAGTCCTGCCGG
CCTCGACGGGCCGAGTTGCCCAGCTGGCTGACCTGTCCCTGGAGATCAGTGGCATCAT
CCCCACAGACGGCGGGAGGTACCAGTGTGTGGCCAGCAATGCCAATGGGGTCACAAGG
GCATCCGTCTGGCTCCTGGTGCGAGAGGTCCCACAGGTCAGCATCCACACCAGCTCCC
AGCACTTCTCCCAAGGTGTGGAGGTGAAGGTCAGCTGCTCAGCCTCTGGATACCCCAC
ACCCCACATCTCCTGGAGCCGTGAGAGCCAAGCCCTACAAGAGGACAGCAGAATCCAT
GTGGACGCACAGGGAACCCTGATTATTCAGGGGGTAGCCCCAGAGGATGCTGGGAATT
ACAGCTGCCAGGCGACTAATGAGGTTGGCACTGACCAGGAGACGGTCACCCTCTACTA
CACAGACCCACCGTCGGTCTCTGTCGAC
ORF Start: at I ORF Stop: end of sequence SEQ ID NO: 46 ~ 686 as MW at 74318.2kD

NOV9d, KLVAVAGAPGTVMPPTTGDATLAFVFDVTGSMWDELMQVIDGASRILERSLSRRSQAI

197192431~~'VPFHDPDIGPWLTADPTVFQRELRELWQGGGDCPEMSVGAIKAAVEVANPG

PrOteln SFIYVFSDARAKDYHKKEELLRLLQLKQSQWFVLTGDCGDHTHPGYLAYEEIAATSS

GQVFHLDKQQVTEVLKWVESAIQASKVHLLSTDHEEEGEHTWRLPFDPSLKEWISLS

Sequence GPGPEIEVQDPLGRILQEDEGLNVLLNIPDSAKWAFKPEHPGLWSIKWSSGRHSVR

ITGVSNIDFRAGFSTQPLLDLNHTLEWPLQGVPISLVINSTGLKAPGRLDSVELAQSS

GKPLLTLPTKPLSNGSTHQLWGGPPFHTPKERFYLKVKGKDHEGNPLLRVSGVSYSGV

APGAPLVSMVPRIHGYLHQPLLVSCSVHSALPFRLQLRRGEARLGEERHFQESGNSSW

EILRASKAEEGTYECTAVSRAGTGRAKAQIWTDPPPQLVPAPNVTVSPGETAVLSCR

VLGEAPYNLTWVRDWRVLPASTGRVAQLADLSLEISGIIPTDGGRYQCVASNANGWR

ASVWLLVREVPQVSIHTSSQHFSQGVEVKVSCSASGYPTPHISWSRESQALQEDSRIH

VDAQGTLIIQGVAPEDAGNYSCQATNEVGTDQETVTLYYTDPPSVSVb SEQ ID NO: 47 2058 by NOV9e, AAGCTTGTGGCAGTGGCCGGGGCGCCCGGGACGGTAATGCCCCCCACCACGGGGGACG

DNA GATCGATGGCGCCTCGCGCATTCTGGAACGCAGTCTGAGCCGCCGCAGCCAGGCCATC

GCCAACTACGCGCTGGTGCCCTTCCACGACCCAGATATTGGCCCAGTGACCCTCACGG

SeqLlenCeCGGACCCCACAGTGTTTCAGAGGGAGCTGAGAGAACTCTACGTGCAGGGAGGTGGTGA

CTGCCCGGAGATGAGTGTGGGGGCCATTAAGGCTGCCGTGGAGGTTGCCAACCCCGGA

TCCTTCATCTACGTCTTTTCGGATGCCCGCGCCAAAGACTATCACAAGAAGGAAGAGC

TGCTGCGGCTCCTGCAGCTCAAGCAATCACAGGTGGTCTTTGTGCTGACGGGGGACTG

TGGCGACCACACCCATCCTGGCTACCTGGCTTATGAGGAGATCGCTGCCACCAGCTCT

GGGCAGGTGTTCCACCTGGACAAGCAGCAAGTGACAGAGGTGCTGAAGTGGGTGGAGT

CAGCGATCCAGGCCTCCAAGGTGCACCTGCTGTCCACAGACCACGAGGAGGAGGGGGA

GCACACATGGAGACTCCCCTTTGACCCCAGCCTGAAGGAGGTCACCATCTCATTGAGT

GGGCCAGGGCCTGAGATTGAAGTCCAAGATCCGCTGGGGAGGATCCTGCAGGAGGACG

AGGGCCTCAACGTGCTTCTCAACATCCCTGACTCGGCCAAGGTCGTAGCCTTTAAGCC

TGAGCATCCGGGGCTGTGGTCCATCAAGGTCTATAGCAGTGGCCGCCATTCAGTGAGG

ATCACAGGCGTCAGCAACATTGACTTCCGAGCCGGCTTCTCCACTCAGCCCTTGCTGG

ACCTCAACCACACCCTCGAGTGGCCCTTGCAAGGAGTCCCCATCTCCCTGGTGATCAA

TTCCACGGGCCTGAAGGCACCCGGCCGCCTAGACTCGGTGGAGCTGGCACAAAGCTCA

GGGAAGCCCCTCCTGACTCTGCCCACGAAGCCCCTCTCCAATGGCTCCACCCATCAGC

TGTGGGGCGGGCCGCCCTTCCACACCCCCAAGGAGCGCTTCTACCTCAAGGTGAAGGG

CAAGGACCATGAGGGAAACCCCCTCCTTCGTGTCTCTGGAGTGTCCTACAGTGGGGTG

GCCCCAGGCGCTCCCCTCGTCAGCATGGCCCCCAGGATCCATGGCTACCTGCACCAGC

CCCTGCTGGTCTCCTGCTCGGTGCACAGTGCCCTTCCCTTCCGGCTGCAGCTGCGGCG

AGGTGAAGCCAGGCTGGGCGAAGAGAGGCACTTTCAGGAGTCGGGAAACAGCAGCTGG

GAGATCCTGCGGGCCTCCAAGGCCGAGGAGGGCACGTACGAGTGCACAGCCGTCAGCA

GGGCTGGGACCGGGCGAGCAAAGGCCCAGATTGTTGTCACAGACCCCCCGCCGCAGCT

GGTCCCTGCTCCCAACGTGACCGTGTCCCCAGGGGAGACTGCCGTCCTATCCTGCCGG

GTCCTAGGCGAGGCCCCCTACAACCTGACGTGGGTCCGGGACTGGCGAGTCCTGCCGG

CCTCGACGGGCCGAGTTGCCCAGCTGGCTGACCTGTCCCTGGAGATCAGTGGCATCAT

CCCCACAGACGGCGGGAGGTACCAGTGTGTGGCCAGCAATGCCAATGGGGTCACAAGG

GCATCCGTCTGGCTCCTGGTGCGAGAGGCCCCACAGGTCAGCATCCACACCAGCTCCC

AGCACTTCTCCCAAGGTGTGGAGGTGAAGGTCAGCTGCTCAGCCTCTGGATACCCCAC

ACCCCACATCTCCTGGAGCCGTGAGAGCCAAGCCCTACAAGAGGACAGCAGAATCCAT

GTGGACGCACAGGGAACCCTGATTATTCAGGGGGTAGCCCCAGAGGATGCTGGGAATT

ACAGCTGCCAGGCGACTAATGAGGTTGGCACTGACCAGGAGACGGTCACCCTCTACGA

CACAGACCCACCGTCGGTCTCTGTCGAC

ORF Start: at 1 ORF
Stop: end of sequence SEQ ID NO: 48 686 as MW at 74214.OkD

NOV9e, KLVAVAGAPGTVMPPTTGDATLAFVFDVTGSMWDELMQVIDGASRILERSLSRRSQAI

197192437~~'VPFHDPDIGPVTLTADPTVFQRELRELYVQGGGDCPEMSVGAIKAAVEVANPG

PPOteln SFIYVFSDARAKDYHKKEELLRLLQLKQSQWFVLTGDCGDHTHPGYLAYEEIAATSS

GQVFHLDKQQVTEVLKWVESAIQASKVHLLSTDHEEEGEHTWRLPFDPSLKEWISLS

Sequence GPGPEIEVQDPLGRILQEDEGLNVLLNIPDSAKWAFKPEHPGLWSIKWSSGRHSVR

ITGVSNIDFRAGFSTQPLLDLNHTLEWPLQGVPISLVINSTGLKAPGRLDSVELAQSS

GKPLLTLPTKPLSNGSTHQLWGGPPFHTPKERFYLKVKGKDHEGNPLLRVSGVSYSGV

APGAPLVSMAPRIHGYLHQPLLVSCSVHSALPFRLQLRRGEARLGEERHFQESGNSSW
EILRASKAEEGTYECTAVSRAGTGRAKAQIVVTDPPPQLVPAPNVTVSPGETAVLSCR
VLGEAPYNLTWVRDWRVLPASTGRVAQLADLSLEISGIIPTDGGRYQCVASNANGVTR
ASVWLLVREAPQVSIHTSSQHFSQGVEVKVSCSASGYPTPHISWSRESQALQEDSRIH
VDAQGTLIIQGVAPEDAGNYSCQATNEVGTDQEWTLYDTDPPSVSVD
ID NO: 49 X2058 by NOV9f, ~AAGCTTGTGGCAGTGGCCGGGGCGCCCGGGACGGTAATGCCCCCCACCACGGGGGACG

DNA GATCGATGGCGCCTCGCGCATTCTGGAACGCAGTCTGAGCCGCCGCAGCCAGGCCATC

GCCAACTACGCGCTGGTGCCCTTCCACGACCCAGATATTGGCCCAGTGACCCTCACGG

SeqllenCeCGGACCCCACAGTGTTTCAGAGGGAGCTGAGAGAACTCTACGTGCAGGGAGGTGGTGA

CTGCCCGGAGATGAGTGTGGGGGCCATTAAGGCTGCCGTGGAGGTTGCCAACCCCGGA

TCCTTCATCTACGTCTTTTCGGATGCCCGCGCCAAAGACTATCACAAGAAGGAAGAGC

TGCTGCGGCTCCTGCAGCTCAAGCAATCACAGGTGGTCTTTGTGCTGACGGGGGACTG

TGGCGACCACACCCATCCTGGCTACCTGGCTTATGAGGAGATCGCTGCCACCAGCTCT

GGGCAGGTGTTCCACCTGGACAAGCAGCAAGTGACAGAGGTGCTGAAGTGGGTGGAGT

CAGCGATCCAGGCCTCCAAGGTGCACCTGCTGTCCACAGACCACGAGGAGGAGGGGGA

GCACACATGGAGACTCCCCTTTGACCCCAGCCTGAAGGAGGTCACCATCTCATTGAGT

~GGGCCAGGGCCTGAGATTGAAGTCCAAGATCCGCTGGGGAGGATCCTGCAGGAGGACG

TGAGCATCCGGGGCTGTGGTCCATCAAGGTCTATAGCAGTGGCCGCCATTCAGTGAGG
ATCACAGGCGTCAGCAACATTGACTTCCGAGCCGGCTTCTCCACTCAGCCCTTGCTGG
ACCTCAACCACACCCTCGAGTGGCCCTTGCAAGGAGTCCCCATCTCCCTGGTGATCAA
TTCCACGGGCCTGAAGGCACCCGGCCGCCTAGACTCGGTGGAGCTGGCACAAAGCTCA
GGGAAGCCCCTCCTGACTCTGCCCACGAAGCCCCTCTCCAATGGCTCCACCCATCAGC
TGTGGGGCGGGCCGCCCTTCCACACCCCCAAGGAGCGCTTCTACCTCAAGGTGAAGGG
CAAGGACCATGAGGGAAACCCCCTCCTTCGTGTCTCTGGAGTGTCCTACAGTGGGGTG
GCCCCAGGCGCTCCCCTCGTCAGCATGGCCCCCAGGATCCATGGCTACCTGCACCAGC
CCCTGCTGGTCTCCTGCTCGGTGCACAGTGCCCTTCCCTTCCGGCTGCAGCTGCGGCG
AGGTGAAGCCAGGCTGGGCGAAGAGAGGCACTTTCAGGAGTCGGGAAACAGCAGCTGG
GAGATCCTGCGGGCCTCCAAGGCCGAGGAGGGCACGTACGAGTGCACAGCCGTCAGCA
GGGCTGGGACCGGGCGAGCAAAGGCCCAGATTGTTGTCACAGACCCCCCGCCGCAGCT
GGTCCCTGCTCCCAACGTGACCGTGTCCCCAGGGGAGGCTGCCGTCCTATCCTGCCGG
GTCCTAGGCGAGGCCCCCTACAACCTGACGTGGGTCCGGGACTGGCGAGTCCTGCCGG
CCTCGACGGGCCGAGTTGCCCAGCTGGCTGACCTGTCCCTGGAGATCAGTGGCATCAT
CCCCACAGACGGCGGGAGGTACCAGTGTGTGGCCAGCAATGCCAATGGGGTCACAAGG
GCATCCGTCTGGCTCCTGGTGCGAGAGGCCCCACAGGTCAGCATCCACACCAGCTCCC
AGCACTTCTCCCAAGGTGTGGAGGTGAAGGTCAGCTGCTCAGCCTCTGGATACCCCAC
GTGGACGCACAGGGAACCCTGATTATTCAGGGGGTAGCCCCAGAGGATGCTGGGAATT
CACAGACCCACCGTCGGTCTCTGTCGAC
ORF Start: at 1 ORF Stop: end of sequence SEQ ID NO: 50 686 as ~ MW at 74232.OkD
NOV9f, KLVAVAGAPGTVMPPTTGDATLAFVFDVTGSMWDELMQVIDGASRILERSLSRRSQAI

197192443~~'VPFHDPDIGPWLTADPTVFQRELRELWQGGGDCPEMSVGAIKAAVEVANPG

PrOteln SFIYVFSDARAKDYHKKEELLRLLQLKQSQWFVLTGDCGDHTHPGYLAYEEIAATSS

GQVFHLDKQQVTEVLKWVESAIQASKVHLLSTDHEEEGEHTWRLPFDPSLKEVTISLS

SeqilenCeGPGPEIEVQDPLGRILQEDEGLNVLLNIPDSAKWAFKPEHPGLWSIKVYSSGRHSVR

ITGVSNIDFRAGFSTQPLLDLNHTLEWPLQGVPISLVINSTGLKAPGRLDSVELAQSS

GKPLLTLPTKPLSNGSTHQLWGGPPFHTPKERFYLKVKGKDHEGNPLLRVSGVSYSGV

APGAPLVSMAPRIHGYLHQPLLVSCSVHSALPFRLQLRRGEARLGEERHFQESGNSSW

EILRASKAEEGTYECTAVSRAGTGRAKAQIWTDPPPQLVPAPNVTVSPGEAAVLSCR

VLGEAPYNLTWVRDWRVLPASTGRVAQLADLSLEISGIIPTDGGRYQCVASNANGVTR

ASVWLLVREAPQVSIHTSSQHFSQGVEVKVSCSASGYPTPHISWSRESQALQEDSRIH

VDAQGTLIIQGVAPEDAGNYSCQATNEVGTDQETVTLYYTDPPSVSVD

SEQ ID NO: 51 X2058 by DNA GATCGATGGCGCCTCGCGCATTCTGGAACGCAGTCTGAGCCGCCGCAGCCAGGCCATC

GCCAACTACGCGCTGGTGCCCTTCCACGACCCAGATATTGGCCCAGTGACCCTCACGG

SeqLlenCeCGGACCCCACAGTGTTTCAGAGGGAGCTGAGAGAACTCTACGTGCAGGGAGGTGGTGA

CTGCCCGGAGATGAGTGTGGGGGCCATTAAGGCTGCCGTGGAGGTTGCCAACCCCGGA

TCCTTCATCTACGTCTTTTCGGATGCCCGCGCCAAAGACTATCACAAGAAGGAAGAGC

TGCTGCGGCTCCTGCAGCTCAAGCAATCACAGGTGGTCTTTGTGCTGACGGGGGACTG

TGGCGACCACACCCATCCTGGCTACCTGGCTTATGAGGAGATCGCTGCCACCAGCTCT

GGGCAGGTGTTCCACCTGGACAAGCAGCAAGTGACAGAGGTGCTGAAGTGGGTGGAGT

CAGCGATCCAGGCCTCCAAGGTGCACCTGCTGTCCACAGACCACGAGGAGGAGGGGGA

GCACACATGGAGACTCCCCTTTGACCCCAGCCTGAAGGAGGTCACCATCTCATTGAGT

GGGCCAGGGCCTGAGATTGAAGTCCAAGATCCGCTGGGGAGGATCCTGCAGGAGGACG

AGCAGTGGCCGCCATTCAGTGAGG
TTCCACGGGCCTGAAGGCACCCGGCCGCCTAGACTCGGTGGAGCTGGCACAAAGCTCA
GGGAAGCCCCTCCTGACTCTGCCCACGAAGCCCCTCTCCAATGGCTCCACCCATCAGC
TGTGGGGCGGGCCGCCCTTCCACACCCCCAAGGAGCGCTTCTACCTCAAGGTGAAGGG
CAAGGACCATGAGGGAAACCCCCTCCTTCGTGTCTCTGGAGTGTCCTACAGTGGGGTG
GCCCCAGGCGCTCCCCTCGTCAGCATGGCCCCCAGGATCCATGGCTACCTGCACCAGC
CCCTGCTGGTCTCCTGCTCGGTGCACAGTGCCCTTCCCTTCCGGCTGCAGCTGCGGCG
AGGTGAAGCCAGGCTGGGCGAAGAGAGGCACTTTCAGGAGTCGGGAAACAGCAGCTGG
GAGATCCTGCGGGCCTCCAAGGCCGAGGAGGGCACGTACGAGTGCACAGCCGTCAGCA
GGGCTGGGACCGGGCGAGCAAAGGCCCAGATTGTTGTCACAGACCCCCCGCCGCAGCT
GGTCCCTGCTCCCAACGTGACCGTGTCCCCAGGGGAGACTGCCGTCCTATCCTGCCGG
GTCCTAGGCGAGGCCCCCTACAACCTGACGTGGGTCCGGGACTGGCGAGTCCTGCCGG
CCTCGACGGGCCGAGTTGCCCAGCTGGCTGACCTGTCCCTGGAGATCAGTGGCATCAT
CCCCACAGACGGCGGGAGGTACCAGTGTGTGGCCAGCAATGCCAATGGGGTCACAAGG
ACCCCAC
ACAAGAGGACAGCAGAATCCAT
ACAGCTGCCAGGCGACTAATGAGGTTGGCACTGACCAGGAGACGGTCACCCTCTACTA
CACAGACCCACCGTCGGTCTCTGTCGAC
ORF Start: at 1 ORF Stop: end of sequence SEQ ID NO: 52 686 as MW at 74292.1 kD
NOV9g, KLVAVAGAPGTVMPPTTGDATLAFVFDVTGSMWDELMQVIDGASRILERSLSRRSQAI

197192448~~'VPFHDPDIGPVTLTADPTVFQRELRELWQGGGDCPEMSVGAIKAAVEVANPG

PPOteln SFIWFSDARAKDYHKKEELLRLLQLKQSQWFVLTGDCGDHTHPGYLAYEEIAATSS

GQVFHLDKQQVTEVLKWESAIQASKVHLLSTDHEEEGEHTWRLPFDPSLKEVTISLS

SeqllenCeGPGPEIEVQDPLGRILQEDEGLNVLLNIPDSAKWAFKPEHPGLWSIKVYSSGRHSVR

ITGVSNIDFRAGFSTQPLLDLNHTLEWPLQGVPISLVINSTGLKAPGRLDSVELAQSS

GKPLLTLPTKPLSNGSTHQLWGGPPFHTPKERFYLKVKGKDHEGNPLLRVSGVSYSGV

APGAPLVSMAPRIHGYLHQPLLVSCSVHSALPFRLQLRRGEARLGEERHFQESGNSSW

EILRASKAEEGTYECTAVSRAGTGRAKAQIVVTDPPPQLVPAPNVTVSPGETAVLSCR

VLGEAPYNLTWVRDWRVLPASTGRVAQLADLSLEISGIIPTDGGRYQCVASNANGVTR

TSVWLLVREAPQVSIHTSSQHFSQGVEVKVSCSASGYPTPHISWSRESQALQEDSRIH

WAQGTLIIQGVAPEDAGNYSCQATNEVGTDQETVTLWTDPPSVSVD

Sequence comparison of the above protein sequences yields the following sequence relationships shown in Table 9B.
Table 9B. Comparison of NOV9a against NOV9b through NOV9g.
NOV9a Residues/ ~Y~~,~"",~"".",.y,r"~,~"~, Identities/ ,I,.,.~~""~", Protein Sequence Match Residues Similarities for the Matched Region NOV9b 1..204 166/204 (81%) 1..204 166/204 (81 %) NOV9c 80..133 15/54 (27%) 28..71 21/54 (38%) NOV9d 20..262 217/243 (89%) 3..245 217/243 (89%) NOV9e 20..262 217/243 (89%) 3..245 217/243 (89%) NOV9f 20..262 217/243 (89%) 3..245 217/243 (89%) NOV9g 20..262 217/243 (89%) ~

3..245 217/243 (89%) Further analysis of the NOV9a protein yielded the following properties shown in Table 9C.
Table 9C. Protein Sequence Properties NOV9a PSort 0.8200 probability located in outside; 0.1000 probability located in analysis: endoplasmic reticulum (membrane); 0.1000 probability located in endoplasmic reticulum (lumen); 0.1000 probability located in lysosome (lumen) SignalP Cleavage site between residues 17 and 18 analysis:
A search of the NOV9a protein against the Geneseq database, a proprietary database that contains sequences published in patents and patent publication, yielded several homologous proteins shown in Table 9D.
Table 9D. Geneseq Results for NOV9a NOV9a Identities/

Geneseq Protein/Organism/LengthResidues/SimilaritiesExpect for Identifier(Patent #, Date] Match ~ the MatchedValue ResiduesRegion AAB83147 Rat secreted factor 1..271 231/272 (84%)e-129 encoded by clone P00210D09 - Rattus1..2?2 241/272 (87%) sp, 275 aa. [W0200123419-A2, OS-APR-AAY53667 Sequence gi/3328186 34..262 119/234 (50%)1e-64 from an alignment with protein 32..265 168/234 (70%) Unidentified, 3117 aa.

[W09960164-A1, 25-NOV-1999]

AAU75886 ~ g~ adhesion molecule 1e-21 protei 1 AD4/AAD21820.1 - Homo 311..547119/239 (49%) Sapiens, 852 aa. [W0200208423-A2, 31-JAN-2002]

AAU75884 Human adhesion molecule34..263 72/239 (30%) 1e-21 protein AD2/G7c - Homo Sapiens, 536 aa. 13..249 119/239 (49%) [W0200208423-A2, 31-JAN-2002]

AAM79854 Human protein SEQ ID 34..263 72/239 (30%) 1e-21 Homo sapiens, 836 aa. 311..547,119/239 (49%) [W0200157190-A2, 09-AUG-2001 ]

In a BLAST search of public sequence datbases, the NOV9a protein was found to have homology to the proteins shown in the BLASTP data in Table 9E.

Table 9E. Public BLASTP Results for NOV9a Protein NOV9a Identities/

Accession Protein/Organism/LengthResidues/SimilaritiesExpect for Number Match the Matched Value Residues Portion CAC37763 SEQUENCE 2 FROM PATENT1..271 231/272 (84%)e-128 W00123419 - Rattus 1..272 241 /272 norvegicus (87%) (Rat), 275 aa.

Q96RW7 HEMICENTIN - Homo sapiens35..262 169/228 (74%)3e-97 (Human), 5636 aa. 38..265 199/228 (87%) T20992 hypothetical protein 34..262 119/234 (50%)3e-64 F15G9.4a -Caenorhabditis elegans,32..265 168/234 (70%) 5175 aa.

076518 HEMICENTIN PRECURSOR 34..262 119/234 (50%)3e-64 -~ ~ 168/234 (70%) Caenorhabditis elegans,32..265 5198 aa.

Q96QC8 G7C PROTEIN - Homo 34..263 72/239 (30%)3e-21 sapiens (Human), 852 aa. 311..547 119/239 (49%) PFam analysis predicts that the NOV9a protein contains the domains shown in the Table 9F.
Table 9F. Domain Analysis of NOV9a Identities/
Pfam Domain NOV9a Match Region Similarities ' Expect Value for the Matched Region EXAMPLE 10.
The NOV 10 clone was analyzed, and the nucleotide and encoded polypeptide sequences are shown in Table 10A.
Table 10A. NOV10 Sequence Analysis SEQ ID NO: 53 X621 NOVlOa, A_TCATGCCCCTAGGTCTCCTGTGGCTGGGCCTAGCCCTGTTGGGGGCTCTGCATGCCC

CG102942-01'a'GGCCCAGGACTCCACCTCAGACCTGATCCCAGCCCCACCTCTGAGCAAGGTCCCTCT

DNA GCAGCAGAACTTCCAGGACAACCAATTCCAGGGGAAGTGGTATGTGGTAGGCCTGGCA

GGGAATGCAATTCTCAGAGAAGACAAAGACCCGCAAAAGATGTATGCCACCATCTATG

Sequence AGCTGAAAGAAGACAAGAGCTACAATGTCACCTCCGTCCTGTTTAGGAAAAAGAAGTG

TGACTACTGGATCAGGACTTTTGTTCCAGGTTGCCAGCCCGGCGAGTTCACGCTGGGC

AACATTAAGAGTTACCCTGGATTAACGAGTTACCTCGTCCGAGTGGTGAGCACCAACT

GATCACCCTCTACGGTAGAACCAAGGAGCTGACTTCGGAACTAAAGGAGAACTTCATC

CGCTTCTCCAAATCTCTGGGCCTCCCTGAAAACCACATCGTCTTCCCTGTCCCAATCG

GTAATGGCCAGTCTGGATGAGGGGACGGGGACATGGGGACT

ORF Start: ATG at 4 ORF
Stop:
TGA
at SEQ ID NO: 54 198 MW at 22456.71cD
as NOVIOa, MPLGLLWLGLALLGALHAQAQDSTSDLIPAPPLSKVPLQQNFQDNQFQGKWYWGLAG

PPOteln IKSYPGLTSYLVRWSTNYNQHAMVFFKKVSQNREYFKITLYGRTKELTSELKENFIR

FSKSLGLPENHIVFPVPIGNGQSG

Sequence SEQ ID NO: 55 609 by NOVIOb, A_TCATGCCCCTAGGTCTCCTGTGGCTGGGCCTAGCCCTGTTGGGGGCTCTGCATGCCC

DNA GCAGCAGAACTTCCAGGACAACCAATTCCAGGGGAAGTGGTATGTGGTAGGCCTGGCA

GGGAATGCAATTCTCAGAGAAGACAAAGACCCGCAAAAGATGTATGCCACCATCTATG

Sequence AGCTGAAAGAAGACAAGAGCTACAATGTCACCTCCGTCCTGTTTAGGAAAAAGAAGTG

TGACTACTGGATCAGGACTTTTGTTCCAGGTTGCCAGCCCGGCGAGTTCACGCTGGGC

AACATTAAGAGTTACCCTGGATTAACGAGTTACCTCGTCCGAGTGGTGAGCACCAACT

ACAACCAGCATGCTATGGTGTTCTTCAAGAAAGTTTCTCAAAACAGGGAGTACTTCAA

GATCACCCTCTACGGGAGAACCAAGGAGCTGACTTCGGAACTAAAGGAGAACTTCATC

CGCTTCTCCAAATCTCTGGGCCTCCCTGAAAACCACATCGTCTTCCCTGTCCCAATCG

GTAATGGCCAGTCTGGATGAGGGGACGGG

ORF Start: ATG at 4 ORF
Stop:
TGA
at SEQ ID NO: 56 198 as MW
at 22456.71cD

NOVIOb, MPLGLLWLGLALLGALHAQAQDSTSDLIPAPPLSKVPLQQNFQDNQFQGKWYWGLAG

IKSYPGLTSYLVRWSTNYNQHAMVFFKKVSQNREYFKITLYGRTKELTSELKENFIR
PrOteln FSKSLGLPENHIVFPVPIGNGQSG

Sequence SEQ ID NO: 57 477 by NOV1OC, CGCGGATCCCAATTCCAGGGGAAGTGGTATGTGGTAGGCCTGGCAGGGAATGCAATTC

DNA C~GAGCTACAATGTCACCTCCGTCCTGTTTAGGAAAAAGAAGTGTGACTACTGGATC

AGGACTTTTGTTCCAGGTTGCCAGCCCGGCGAGTTCACGCTGGGCAACATTAAGAGTT

SeqLlenCeACCCTGGATTAACGAGTTACCTCGTCCGAGTGGTGAGCACCAACTACAACCAGCATGC

TATGGTGTTCTTCAAGAAAGTTTCTCAAAACAGGGAGTACTTCAAGATCACCCTCTAC

GGGAGAACCAAGGAGCTGACTTCGGAACTAAAGGAGAACTTCATCCGCTTCTCCAAAT

CTCTGGGCCTCCCTGAAAACCACATCGTCTTCCCTGTCCCAATCGGTAATGGCCAGTC

TGGACTCGAGGCG

ORF Start: at I ORF
Stop: end of sequence SEQ ID NO: 58 159 as MW at 18222.81cD

NOV1OC, RGSQFQGKWYWGLAGNAILRGDKDPQKMYATIYELKEDKSYNVTSVLFRKKKCDYWI

GRTKELTSELKENFIRFSKSLGLPENHIVFPVPIGNGQSGLEA
Protein Sequence Sequence comparison of the above protein sequences yields the following sequence relationships shown in Table IOB.

Table 10B. Comparison of NOVlOa against NOVlOb and NOVlOc.
Protein Sequence NOVlOa Residues/ Identities/
Match Residues Similarities for the Matched Region NOVlOb 19..198 180/180 (100%) 19..198 180/180 (100%) NOV l Oc 45..198 1521154 (98%) 3..156 ~ 1531154 (98%) Further analysis of the NOV 10a protein yielded the following properties.shown in Table IOC.
Table 10C. Protein Sequence Properties NOVlOa PSort 0.4658 probability located in outside; 0.1134 probability located in analysis: microbody (peroxisome); 0.1000 probability located in endoplasmic reticulum (membrane); 0.1000 probability located in endoplasmic reticulum (lumen) SignalP ~ Cleavage site between residues 21 and 22 analysis:
A search of the NOV 10a protein against the Geneseq database, a proprietary database that contains sequences published in patents and patent publication, yielded several homologous proteins shown in Table l OD.

Table !OD. Geneseq Results for NOVlOa NOVlOa Identities/

Geneseq Protein/OrganismlLengthResidues/SimilaritiesEzpect for Identifier[Patent #, Date] Match the Matched Value ResiduesRegion AAG74315 Human colon cancer antigen1..192 192/192 (100%)e-110 protein SEQ ID N0:5079 57..248 192/192 (100%) - Homo Sapiens, 254 aa. [W0200122920-A2, OS-APR-2001]

AAY71470 Human neutrophil gelatinase1..192 1921192 (100%)e-110 associated protein (NGAL)1..192 192/192 (100%) -Homo sapiens, 198 aa.

[W0200029576-A1, 25-MAY-2000]

AAB43668 Human cancer associatedI ..192 192/192 (100%)e-110 protein sequence SEQ ID NO:l 57..248 1921192 (100%) Homo Sapiens, 254 aa.

[W0200055350-A1, 21-SEP-2000]

AAW49088 Human NGAL protein - I ..192 1891192 (98%)e-107 Homo Sapiens, 197 aa. [W09830907-AI,1..191 190/192 (98%)~

16-JUL-1998]

AAW 18203Human NGAL protein - I ..192 189/192 (98%)e-107 Homo Sapiens, 197 aa. [US5627034-A,1..191 1901192 (98%) 06-MAY-1997]

In a BLAST search of public sequence datbases, the NOV I Oa protein was found to have homology to the proteins shown in the BLASTP data in Table 10E.

Table 10E. Public BLASTP Results for NOVlOa Protein NOVlOa Identities/

AccessionProtein/Organism/Length Residues/SimilaritiesExpect for Number Match the Matched Value ResiduesPortion P80188 Neutrophil gelatinase-associated1..192 192/192 (100%)e-110 lipocalin precursor (NGAL)1..192 192/192 (100%) (P25) (25 lcDa alpha-2-microglobulin-related subunit of MMP-9) (Lipocalin 2) (Oncogene 24p3) -Homo sapiens (Human), 198 aa.

JC2339 neutrophil gelatinase-associated1..192 189/192 (98%)e-107 lipocalin precursor - 1..191 190/192 (98%) human, 197 aa.

Q9QVP7 NEU-RELATED LIPOCALIN 1..191 121/191 (63%)1e-66 -Rattus sp, 198 aa. 1..191 150/191 (78%) P30152 Neutrophil gelatinase-associated1..191 120/191 (62%)1e-65 lipocalin precursor (NGAL)1..191 148/191 (76%) (P25) (Alpha-2-microglobulin-related protein) (Alpha-2U globulin-related protein) (Lipocalin 2) - Rattus norvegicus (Rat), 198 aa.

Q60842 CHROMOSOME 24P3 - Mus 1..194 119/196 (60%)3e-64 musculus (Mouse), 283 8..203 154/196 (77%) as (fragment)., PFam analysis predicts that the NOV 10a protein contains the domains shown in the Table !OF.
Table !OF. Domain Analysis of NOVlOa Identities/
Pfam Domain NOVlOa Match Region Similarities Expect Value for the Matched Region lipocalin 46..189 42/152 (28%) 5.4e-34 115/152 (76%) Eh'AMPLE 11.
The NOV 11 clone was analyzed, and the nucleotide and encoded polypeptide sequences are shown in Table 11 A.
Table 11A. NOVll Sequence Analysis SEQ ID NO: 592210 by NOVlla, ATGACAATTTTAAGAGTGTTTAACCAAGACTGTTCCTTTAAATGTGTTCTTTTGCTGC

CG1O4O16-OlI'rGTTTAATTATACATGTCAATTATTTACAGATCCTGTGGTATTGTGGAAATTCCCAGA
DNA GGACTTTGGAGACCAGGAAATACTACAGAGTGTGCCAAAGTTCTGTTTTCCCTTTGAC
SeCllleriCe GTTGAAAGGTACAGTATAAGTCAAGTTGGACAGCACTTTACCTTTGTACTGACAGACA
TTGAAAGTAAACAGAGATTTGGATTCTGCAGACTGACGTCAGGAGGCACAATTTGTTT
ATGCATCCTTAGTTACCTTCCCTGGTTTGAAGTGTATTACAAGCTTCTAAATACTCTT
GCAGATTACTTGGCTAAGCATTCCTACTTCATTGCCCCTGATGTAACTGGACTCCCAA
CAATACCCGAGAGTAGAAATCTTACAGAATATTTTGTTGCCGTGGATGTGAACAACAT
GCTGCAGCTGTATGCCAGTATGCTGCATGAAAGGCGCATCGTGATTATCTCGAGCAAA
TTAAGCACTTTAACTGCCTGTATCCATGGATCAGCTGCTCTTCTATACCCAATGTATT
GGCAACACATATACATCCCAGTGCTTCCTCCACACCTGCTGGACTACTGCAGTGCCCC
AATGCCATACCTGATTGGAATACACTCCAGCCTCATAGAGAGAGTGAAAAACAAATCA
TTGGAAGATGTTGTTATGTTAAATGTTGATACAAACACATTAGAATCACCATTTAGTG
ACTTGAACAACCTACCAAGTGATGTGGTAAGTGCCTTGAAAAATAAACTGAAGAAGCA
GTCTACAGCTACGGGTGATGGAGTAGCTAGGGCCTTTCTTAGAGCACAGGCTGCTTTG
TTTGGATCCTACAGAGATGCACTGAGATACAAACCTGGTGAGCCCATCACTTTCTGTG
AGGAGAGTTTTGTAAAGCACCGCTCAAGCGTGATGAAACAGTTCCTGGAAACTGCCAT
TAACCTCCAGCTTTTTAAGCAGGTATTTATCGATGGTCGACTGGCAAAACTAAATGCA
GGAAGGGGTTTCTCTGATGTATTTGAAGAAGAGATCACTTCAGGTGGCTTTTGTGGAG
GTAAAGACAAGTTACAATATAAATATGTTTCTGTTTTTCTTTTGCAGAAAGGAGGTGC
ACTGTTCAACACAGCAATGACCAAAGCAACCCCTGCTGTACGGACAGCATATAAATTT
GCAAAAAATCATGCAAAGCTGGGACTAAAGGAAGTGAAGAGTAAACTAAAACACAAGG
AAAATGAAGAAGATTATGGGACCTGTTCTAGTTCTGTACAATATACACCAGTTTACAA
ATTACACAATGAAAAGGGAGGAAACTCAGAAAAGCGTAAGCTTGCTCAGGCACGCTTA
AAAAGGCCTCTTAAGAGCCTTGATGGTGCTCTATATGATGATGAAGATGATGATGACA
TTGAAAGAGCAAGCAAGTTATCTTCTGAAGATGGTGAAGAAGCTTCTGCTTATCTCTA
TGAGAGTGATGACTCTGTTGAAACAAGAGTGAAGACTCCTTACTCAGGTGAAATGGAC
TTACTAGGAGAGATTCTTGATACATTGAGCACACACAGCTCAGATCAGGGGAAGCTGG
CAGCTGCAAAGAGCTTGGATTTCTTTAGATCAATGGATGACATTGATTACAAACCTAC
GAATAAATCTAATGCTCCTAGTGAGAATAACCTGGCTTTCCTCTGTGGTGGTTCTGGT
GACCAAGCAGAGTGGAATCTTGGGCAAGACGATAGTGCCCTCCATGGCAAACACCTCC
CTCCATCTCCTAGGAAGCGGGTTTCCTCTAGTGGTTTGACAGATTCTCTGTTTATCCT
GAGAGAGGAAAACAGTAACAAGCACCTCGGTGCTGACAATGTGAGTGACCCTACTTCA
GGACTGGATTTCCAACTCACTTCCCCTGAAGTTTCCCAGACTGATAAAGGAAAAACAG
AAAAGAGGGAAACACTAAGCCAGATTTCAGATGATCTGCTTATACCCGGTCTTGGGCG
GCATTCATCGACTTTTGTTCCTTGGGAGAAAGAAGGGAAAGAAGCCAAAGAGACTTCA
GAAGATATTGGACTGCTCCATGAAGTAGTGTCATTATGTCATATGACATCTGACTTCC
AACAAAGCTTGAACATTTCAGACAAAAACACAAATGGAAACCAAACTTAAATCTTGCA
ORF Start: ATG at 1 ORF Stop: TAA at 219_4 SEQ ID NO: 60 ~ 731 as MW at 81769.SkD
NOVlla, MTILRVFNQDCSFKCVLLLLFNYTCQLFTDPWLWKFPEDFGDQEILQSVPKFCFPFD

PrOteIri ~YLAKHSYFIAPDVTGLPTIPESRNLTEYFVAVDVNNMLQLYASMLHERRIVIISSK
LSTLTACIHGSAALLYPMYWQHIYIPVLPPHLLDYCSAPMPYLIGIHSSLIERVKNKS
SeC1L12riCe LEDVVMLNVDTNTLESPFSDLNNLPSDWSALKNKLKKQSTATGDGVARAFLRAQAAL
FGSYRDALRYKPGEPITFCEESFVKHRSSVMKQFLETAINLQLFKQVFIDGRLAKLNA
GRGFSDVFEEEITSGGFCGGKDKLQYKYVSVFLLQKGGALFNTAMTKATPAVRTAYKF
KRPLKSLDGALYDDEDDDDIERASKLSSEDGEEASAYLYESDDSVETRVKTPYSGEMD
LLGEILDTLSTHSSDQGKLAAAKSLDFFRSMDDIDYKPTNKSNAPSENNLAFLCGGSG
DQAEWNLGQDDSALHGKHLPPSPRKRVSSSGLTDSLFILREENSNKHLGADNVSDPTS
GLDFQLTSPEVSQTDKGKTEKRETLSQISDDLLIPGLGRHSSTFVPWEKEGKEAKETS
EDIGLLHEWSLCHMTSDFQQSLNISDKNTNGNQT
SEQ ID NO: 61 X2256 by NOVllb, AGATCTGATCCTGTGGTATTGTGGAAATTCCCAGAGGACTTTGGAGACCAGGAAATAC

DNA AGTTGGACAGCACTTTACCTTTGTACTGACAGACATTGAAAGTAAACAGAGATTTGGA
TTCTGCAGACTGACGTCAGGAGGCACAATTTGTTTATGCATCCTTAGTTACCTTCCCT

SeqLlenCeGGTTTGAAGTGTATTACAAGCTTCTAAATACTCTTGCAGATTACTTGGCTAAGGAACT

GGAAAATGATTTGAATGAAACTCTCAGATCACTGTATAACCACCCAGTACCAAAGGCA

AATACTCCTGTAAATTTGAGTGTGAACCAAGAGATATTTATTACCTGTGAGCAAGTTC

TGAAAGATCAGCCTGCTCTACTACCGCATTCCTACTTCATTGCCCCTGATGTAACTGG

ACTCCCAACAATACCCGAGAGTAGAAATCTTACAGAATATTTTGTTGCCGTGGATGTG

AACAACATGCTGCAGCTGTATGCCAGTATGCTGCATGAAAGGCGCATCGTGATTATCT

CGAGCAAATTAAGCACTTTAACTGCCTGTATCCATGGATCAGCTGCTCTTCTATACCC

AATGTATTGGCAACACATATACATCCCAGTGCTTCCTCCACACCTGCTGGACTACTGC

TGTGCCCCAATGCCATACCTGATTGGAATACACTCCAGCCTCATAGAGAGAGTGAAAA

ACAAATCATTGGAAGATGTTGTTATGTTAAATGTTGATACAAACACATTAGAATCACC

ATTTAGTGACTTGAACAACCTACCAAGTGATGTGGTCTCGGCCTTGAAAAATAAACTG

AAGAAGCAGTCTACAGCTACGGGTGATGGAGTAGCTAGGGCCTTTCTTAGAGCACAGG

CTGCTTTGTTTGGATCCTACAGAGATGCACTGAGATACAAACCTGGTGAGCCCATCAC

TTTCTGTGAGGAGAGTTTTGTAAAGCACCGCTCAAGCGTGATGAAACAGTTCCTGGAA

ACTGCCATTAACCTCCAGCTTTTTAAGCAGTTTATCGATGGTCGACTGGCAAAACTAA

ATGCAGGAAGGGGTTTCTCTGATGTATTTGAAGAAGAGATCACTTCAGGTGGCTTTTG

TGGAGGGAACCCGAGGTCATATCAACAATGGGTGCATACAGTCAAGAAAGGAGGTGCA

CTGTTCAACACAGCAATGACCAAAGCAACCCCTGCTGTACGGACAGCATATAAATTTG

CAAAP.AATCATGCAAAGCTGGGACTAAAGGAAGTGAAGAGTAAACTAAAACACAAGGA

AAATGAAGAAGATTATGGGACCTGTTCTAGTTCTGTACAATATACACCAGTTTACAAA

TTACACAATGAAAAGGGAGGAAACTCAGAAAAGCGTAAGCTTGCTCAGGCACGCTTAA

AAAGGCCTCTTAAGAGCCTTGATGGTGCTCTATATGATGATGAAGATGATGATGACAT

TGAAAGAGCAAGCAAGTTATCTTCTGAAGATGGTGAAGAAGCTTCTGCTTATCTCTAT

GAGAGTGATGACTCTGTTGAAACAAGAGTGAAGACTCCTTACTCAGGTGAAATGGACT

TACTAGGAGAGATTCTTGATACATTGAGCACACACAGCTCAGATCAGGGGAGGCTGGC

AGCTGCAAAGAGCTTGGATTTCTTTAGATCAATGGACGACATTGATTACAAACCTACG

AATAAATCTAATGCTCCTAGTGAGAATAACCTGGCTTTCCTCTGTGGTGGTTCTGGTG

ACCAAGCAGAGTGGAATCTTGGGCAAGACGATAGTGCCCTCCATGGCAAACACCTCCC

TCCATCTCCTAGGAAGCGGGTTTCCTCTAGTGGTTTGACAGATTCTCTGTTTATCCTG

AAAGAGGAAAACAGTAACAAGCACCTCGGTGCTGACAATGTGAGTGACCCTACTTCAG

GACTGGATTTCCAACTCACTTCCCCTGAAGTTTCCCAGACTGATAAAGGAAAAACAGA

AAAGAGGGAAACACTAAGCCAGATTTCAGATGATCTGCTTATACCCGGTCTTGGGCGG

CATTCATCGACTTTTGTTCCTTGGGAGAAAGAAGGGAAAGAAGCCAAAGAGACTTCAG

AAGATATTGGACTGCTCCATGAAGTAGTGTCATTATGTCATATGACATCTGACTTCCA

ACAAAGCTTGAACATTTCAGACAAAAACACAAATGGAAACCAAACTAGATCT

ORF Start: at 1 ORF Stop:
end of sequence SEQ ID NO: 62 752 as MW at 84005.6kD

NOVllb, RSDPVVLWKFPEDFGDQEILQSVPKFCFPFDVERVSQNQVGQHFTFVLTDIESKQRFG

NTPVNLSVNQEIFITCEQVLKDQPALLPHSYFIAPDVTGLPTIPESRNLTEYFVAVDV
PrOteln ~L,QLYASMLHERRIVIISSKLSTLTACIHGSAALLYPMYWQHIYIPVLPPHLLDYC

SeqLlenCeCAPMPYLIGIHSSLIERVKNKSLEDVVMLNVDTNTLESPFSDLNNLPSDWSALKNKL

KKQSTATGDGVARAFLRAQAALFGSYRDALRYKPGEPITFCEESFVKHRSSVMKQFLE

TAINLQLFKQFIDGRLAKLNAGRGFSDVFEEEITSGGFCGGNPRSYQQWVHTVKKGGA

LFNTAMTKATPAVRTAYKFAKNHAKLGLKEVKSKLKHKENEEDYGTCSSSVQYTPVYK

LHNEKGGNSEKRKLAQARLKRPLKSLDGALYDDEDDDDIERASKLSSEDGEEASAYLY

ESDDSVETRVKTPYSGEMDLLGEILDTLSTHSSDQGRLAAAKSLDFFRSMDDIDYKPT

NKSNAPSENNLAFLCGGSGDQAEWNLGQDDSALHGKHLPPSPRKRVSSSGLTDSLFIL

KEENSNKHLGADNVSDPTSGLDFQLTSPEVSQTDKGKTEKRETLSQISDDLLIPGLGR

HSSTFVPWEKEGKEAKETSEDIGLLHEVVSLCHMTSDFQQSLNISDKNTNGNQTRS

SEQ ID NO: 63 2256 by NOVIIC, AGATCTGATCCTGTGGTATTGTGGAAATTCCCAGAGGACTTTGGAGACCAGGAAATAC

DNA AGTTGGACAGCACTTTACCTTTGTACTGACAGACATTGAAAGTAAACAGAGATTTGGA

TTCTGCAGACTGACGTCAGGAGGCACAATTTGTTTATGCATCCTTAGTTACCTTCCCT

SequenceGGTTTGAAGTGTATTACAAGCTTCTAAATACTCTTGCAGATTACTTGGCTAAGGAACT

GGAAAATGATTTGAATGAAACTCTCAGATCACTGTATAACCACCCAGTACCAAAGGCA

AATACTCCTGTAAATTTGAGTGTGAACCAAGAGATATTTATTACCTGTGAGCAAGTTC

TGAAAGATCAGCCTGCTCTACTACCGCATTCCTACTTCATTGCCCCTGATGTAACTGG
ACTCCCAACAATACCCGAGAGTAGAAATCTTACAGAATATTTTGTTGCCGTGGATGTG
AACAACATGCTGCAGCTGTATGCCAGTATGCTGCATGAAAGGCGCATCGTGATTATCT
CGAGCAAATTAAGCACTTTAACTGCCTGTATCCATGGATCAGCTGCTCTTCTATACCC
AATGTATTGGCAACACATATACATCCCAGTGCTTCCTCCACACCTGCTGGACTACTGC
TGTGCCCCAATGCCATACCTGATTGGAATACACTCCAGCCTCATAGAGAGAGTGAAAA
ACAAATCATTGGAAGATGTTGTTATGTTAAATGTTGATACAAACACATTAGAATCACC
ATTTAGTGACTTGAACAACCTACCAAGTGATGTGGTCTCGGCCTTGAAAAATAAACTG
AAGAAGCAGTCTACAGCTACGGGTGATGGAGTAGCTAGGGCCTTTCTTAGAGCACAGG
CTGCTTTGTTTGGATCCTACAGAGATGCACTGAGATACAAACCTGGTGAGCCCATCAC
TTTCTGTGAGGAGAGTTTTGTAAAGCACCGCTCAAGCGTGATGAAACAGTTCCTGGAA
ACTGCCATTAACCTCCAGCTTTTTAAGCAGTTTATCGATGGTCGACTGGCAAAACTAA
ATGCAGGAAGGGGTTTCTCTGATGTATTTGAAGAAGAGATCACTTCAGGTGGCTTTTG
TGGAGGGAACCCGAGGTCATATCAACAATGGGTGCATACAGTCAAGAAAGGAGGTGCA
CTGTTCAACACAGCAATGACCAAAGCAACCCCTGCTGTACGGACAGCATATAAATTTG
CAAAAAATCATGCAAAGCTGGGACTAAAGGAAGTGAAGAGTAAACTAAAACACAAGGA
AAATGAAGAAGATTATGGGACCTGTTCTAGTTCTGTACAATATACACCAGTTTACAAA
TTACACAATGAAAAGGGAGGAAACTCAGAAAAGCGTAAGCTTGCTCAGGCACGCTTAA
AAAGGCCTCTTAAGAGCCTTGATGGTGCTCTATATGATGATGAAGATGATGATGACAT
TGAAAGAGCAAGCAAGTTATCTTCTGAAGATGGTGAAGAAGCTTCTGCTTATCTCTAT
GAGAGTGATGACTCTGTTGAAACAAGAGTGAAGACTCCTTACTCAGGTGAAATGGACT
TACTAGGAGAGATTCTTGATACATTGAGCACACACAGCTCAGATCAGGGGAGGCTGGC
AGCTGCAAAGAGCTTGGATTTCTTTAGATCAATGGACGACATTGATTACAAACCTACG
AATAAATCTAATGCTCCTAGTGAGAATAACCTGGCTTTCCTCTGTGGTGGTTCTGGTG
ACCAAGCAGAGTGGAATCTTGGGCAAGACGATAGTGCCCTCCATGGCAAACACCTCCC
TCCATCTCCTAGGAAGCGGGTTTCCTCTAGTGGTTTGACAGATTCTCTGTTTATCCTG
AAAGAGGAAAACAGTAACAAGCACCTCGGTGCTGACAATGTGAGTGACCCTACTTCAG
GACTGGATTTCCAACTCACTTCCCCTGAAGTTTCCCAGACTGATAAAGGAAAAACAGA
AAAGAGGGAAACACTAAGCCAGATTTCAGATGATCTGCTTATACCCGGTCTTGGGCGG
CATTCATCGACTTTTGTTCCTTGGGAGAAAGAAGGGAAAGAAGCCAAAGAGACTTCAG
AAGATATTGGACTGCTCCATGAAGTAGTGTCATTATGTCATATGACATCTGACTTCCA
ACAAAGCTTGAACATTTCAGACAAAAACACAAATGGAAACCAAACTAGATCT
ORF Start: at 1 ORF Stop: end of sequence SEQ ID NO: 64 752 as ~MW at 84005.6kD
NOV11C, RSDPWLWKFPEDFGDQEILQSVPKFCFPFDVERVSQNQVGQHFTFVLTDIESKQRFG

NTPVNLSVNQEIFITCEQVLKDQPALLPHSYFIAPDVTGLPTIPESRNLTEYFVAVDV
PrOteln ~I,QLYASMLHERRIVIISSKLSTLTACIHGSAALLYPMYWQHIYIPVLPPHLLDYC
Sequence CAPMPYLIGIHSSLIERVKNKSLEDVVMLNVDTNTLESPFSDLNNLPSDWSALKNKL
KKQSTATGDGVARAFLRAQAALFGSYRDALR1'KPGEPITFCEESFVKHRSSVMKQFLE
TAINLQLFKQFIDGRLAKLNAGRGFSDVFEEEITSGGFCGGNPRSYQQWVHTVKKGGA
LFNTAMTKATPAVRTAYKFAKNHAKLGLKEVKSKLKHKENEEDYGTCSSSVQYTPWK
LHNEKGGNSEKRKLAQARLKRPLKSLDGALYDDEDDDDIERASKLSSEDGEEASAYLY
ESDDSVETRVKTPYSGEMDLLGEILDTLSTHSSDQGRLA.AAKSLDFFRSMDDIDYKPT
NKSNAPSENNLAFLCGGSGDQAEWNLGQDDSALHGKHLPPSPRKRVSSSGLTDSLFIL
KEENSNKHLGADNVSDPTSGLDFQLTSPEVSQTDKGKTEKRETLSQISDDLLIPGLGR
HSSTFVPWEKEGKEAKETSEDIGLLHEWSLCHMTSDFQQSLNISDKNTNGNQTRS
SEQ ID NO: 65 2259 by NOVlld, AGATCTGATCCTGTGGTATTGTGGAAATTCCCAGAGGACTTTGGAGACCAGGAAATAC

DNA AGTTGGACAGCACTTTACCTTTGTACTGACAGACATTGAAAGTAAACAGAGATTTGGA
TTCTGCAGACTGACGTCAGGAGGCACAATTTGTTTATGCATCCTTAGTTACCTTCCCT
SeqlleriCe GGTTTGAAGTGTATTACAAGCTTCTAAATACTCTTGCAGATTACTTGGCTAAGGAACT
GGAAAATGATTTGAATGAAACTCTCAGATCACTGTATAACCACCCAGTACCAAAGGCA
AATACTCCTGTAAATTTGAGTGTGAACCAAGAGATATTTATTGCCTGTGAGCAAGTTC
TGAAAGATCAGCCTGCTCTAGTACCGCATTCCTACTTCATTGCCCCTGATGTAACTGG
ACTCCCAACAATACCCGAGAGTAGAAATCTTACAGAATATTTTGTTGCCGTGGATGTG
AACAACATGCTGCAGCTGTATGCCAGTATGCTGCATGAAAGGCGCATCGTGATTATCT

CGAGCAAATTAAGCACTTTAACTGCCTGTATCCATGGATCAGCTGCTCTTCTATACCC

AATGTATTGGCAACACATATACATCCCAGTGCTTCCTCCACACCTGCTGGACTACTGC

TGTGCCCCAATGCCATACCTGATTGGAATACACTCCAGCCTCATAGAGAGAGTGAAAA

ACAAATCATTGGAAGATGTTGTTATGTTAAATGTTGATACAAACACATTAGAATCACC

ATTTAGTGACTTGAACAACCTACCAAGTGATGTGGTCTCGGCCTTGAAAAATAAACTG

AAGAAGCAGTCTACAGCTACGGGTGATGGAGTAGCTAGGGCCTTTCTTAGAGCACAGG

CTGCTTTGTTTGGATCCTACAGAGATGCACTGAGATACAAACCTGGTGAGCCCATCAC

TTTCTGTGAGGAGAGTTTTGTAAAGCACCGCTCAAGCGTGATGAAACAGTTCCTGGAA

ACTGCCATTAACCTCCAGCTTTTTAAGCAGTTTATCGATGGTCGACTGGCAAAACTAA

ATGCAGGAAGGGGTTTCTCTGATGTATTTGAAGAAGAGATCACTTCAGGTGGCTTTTG

TGGAGGGP:ACCCGAGGTCATATCAACAATGGGTGCATACAGTCAAGAAAGGAGGTGCA

CTGTTCAACACAGCAATGACCAAAGCAACCCCTGCTGTACGGACAGCATATAAATTTG

CAAAAAATCATGCAAAGCTGGGACTAAAGGAAGTGAAGAGTAAACTAAAACACAAGGA

AAATGAAGAAGATTATGGGACCTGTTCTAGTTCTGTACAATATACACCAGTTTACAAA

TTACACAATGAAAAGGGAGGAAACTCAGAAAAGCGTAAGCTTGCTCAGGCACGCTTAA

AAAGGCCTCTTAAGAGCCTTGATGGTGCTCTATATGATGATGAAGATGATGATGACAT

TGAAAGAGCAAGCAAGTTATCTTCTGAAGATGGTGAAGAAGCTTCTGCTTATCTCTAT

GAGAGTGATGACTCTGTTGAAACAAGAGTGAAGACTCCTTACTCAGGTGAAATGGACT

TACTAGGAGAGATTCTTGATACATTGAGCACACACAGCTCAGATCAGGGGAAGCTGGC

AGCTGCAAAGAGCTTGGATTTCTTTAGATCAATGGATGACATTGATTACAAACCTACG

AATAAATCTAATGCTCCTAGTGAGAATAACCTGGCTTTCCTCTGTAGTGGTTCTGGTG

ACCAAGCAGAGTGGAATCTTGGGCAAGACGATAGTGCCCTCCATGGCAAACACCTCCC

TCCATCTCCTAGGAAGCGGGTTTCCTCTAGTGGTTTGACAGATTCTCTGTCTATCCTG

AAAGAGGAAAACAGTAACAAGCACCTCGGTGCTGACAATGTGAGTGACCCTACTTCAG

GACTGGATTTCCAACTCACTTCCCCTGAAGTTTCCCAGACTGATAAAGGAAAAACAGA

AAAGAGGGAAACACTAAGCCAGATTTCAGATGATCTGCTTATACCCGGTCTTGGGCGG

CATTCATCGACTTTTGTTCCTTGGGAGAAAGAAGGGAAAGAAGCCAAAGAGACTTCAG

AAGATATTGGACTGCTCCATGAAGTAGTGTCATTATGTCATATGACATCTGACTTCCA

AGCTAAAGCTTGGAACATTTCAGACAAAAACACAAATGGAAACCAAACTAGATCT

ORF Start: at 1 ORF
Stop: end of sequence SEQ ID NO: 66 753 as MW at 84031.7kD

NOVlld, RSDPWLWKFPEDFGDQEILQSVPKFCFPFDVERVSQNQVGQHFTFVLTDIESKQRFG

PTOteln NTPVNLSVNQEIFIACEQVLKDQPALVPHSYFIAPDVTGLPTIPESRNLTEYFVAVDV

NNMLQLYASMLHERRIVIISSKLSTLTACIHGSAALLYPMYWQHIYIPVLPPHLLDYC

Sequence CAPMPYLIGIHSSLIERVKNKSLEDVVMLNVDTNTLESPFSDLNNLPSDWSALKNKL

KKQSTATGDGVARAFLRAQAALFGSYRDALRYKPGEPITFCEESFVKHRSSVMKQFLE

TAINLQLFKQFIDGRLAKLNAGRGFSDVFEEEITSGGFCGGNPRSYQQWVHTVKKGGA

LFNTAMTKATPAVRTAYKFAKNHAKLGLKEVKSKLKHKENEEDYGTCSSSVQYTPWK

LHNEKGGNSEKRKLAQARLKRPLKSLDGALYDDEDDDDIERASKLSSEDGEEASAYLY

ESDDSVETRVKTPYSGEMDLLGEILDTLSTHSSDQGKLAAAKSLDFFRSMDDIDYKPT

NKSNAPSENNLAFLCSGSGDQAEWNLGQDDSALHGKHLPPSPRKRVSSSGLTDSLSIL

KEENSNKHLGADNVSDPTSGLDFQLTSPEVSQTDKGKTEKRETLSQISDDLLIPGLGR

HSSTFVPWEKEGKEAKETSEDIGLLHEWSLCHMTSDFQAKAWNISDKNTNGNQTRS

SEQ ID NO: 67 2256 by NOVIle, AGATCTGATCCTGTGGTATTGTGGAAATTCCCAGAGGACTTTGGAGACCAGGAAATAC

DNA AGTTGGACAGCACTTTACCTTTGTACTGACAGACATTGAAAGTAAACAGAGATTTGGA

TTCTGCAGACTGACGTCAGGAGGCACAATTTGTTTATGCATCCTTAGTTACCTTCCCT

SeqllenCeGGTTTGAAGTGTATTACAAGCTTCTAAATACTCTTGCAGATTACTTGGCTAAGGAACT

GGAAAATGATTTGAATGAAACTCTCAGATCACTGTATAACCACCCAGTACCAAAGGCA

AATACTCCTGTAAATTTGAGTGTGAACCAAGAGATATTTATTGCCTGTGAGCAAGTTC

TGAAAGATCAGCCTGCTCTAGTACCGCATTCCTACTTCATTGCCCCTGATGTAACTGG

ACTCCCAACAATACCCGAGAGTAGAAATCTTACAGAATATTTTGTTGCCGTGGATGTG

AACAACATGCTGCAGCTGTATGCCAGTATGCTGCATGAAAGGCGCATCGTGATTATCT

CGAGCAAATTAAGCACTTTAACTGCCTGTATCCATGGATCAGCTGCTCTTCTATACCC

AATGTATTGGCAACACATATACATCCCAGTGCTTCCTCCACACCTGCTGGACTACTGC

TGTGCCCCAATGCCATACCTGATTGGAATACACTCCAGCCTCATAGAGAGAGTGAAAA

ACAAATCATTGGAAGATGTTGTTATGTTAAATGTTGATACAAACACATTAGAATCACC
ATTTAGTGACTTGAACAACCTACCAAGTGATGTGGTCTCGGCCTTGAAAAATAAACTG
AAGAAGCAGTCTACAGCTACGGGTGATGGAGTAGCTAGGGCCTTTCTTAGAGCACAGG
CTGCTTTGTTTGGATCCTACAGAGATGCACTGAGATACAAACCTGGTGAGCCCATCAC
TTTCTGTGAGGAGAGTTTTGTAAAGCACCGCTCAAGCGTGATGAAACAGTTCCTGGAA
ACTGCCATTAACCTCCAGCTTTTTAAGCAGTTTATCGATGGTCGACTGGCAAAACTAA
ATGCAGGAAGGGGTTTCTCTGATGTATTTGAAGAAGAGATCACTTCAGGTGGCTTTTG
TGGAGGGAACCCGAGGTCATATCAACAATGGGTGCATACAGTCAAGAAAGGAGGTGCA
CTGTTCAACACAGCAATGACCAAAGCAACCCCTGCTGTACGGACAGCATATAAATTTG
CAAAAAATCATGCAAAGCTGGGACTAAAGGAAGTGAAGAGTAAACTAAAACACAAGGA
AAATGAAGAAGATTATGGGACCTGTTCTAGTTCTGTACAATATACACCAGTTTACAAA
TTACACAATGAAAAGGGAGGAAACTCAGAAAAGCGTAAGCTTGCTCAGGCACGCTTAA
AAAGGCCTCTTAAGAGCCTTGATGGTGCTCTATATGATGATGAAGATGATGATGACAT
TGAAAGAGCAAGCAAGTTATCTTCTGAAGATGGTGAAGAAGCTTCTGCTTATCTCTAT
GAGAGTGATGACTCTGTTGAAACAAGAGTGAAGACTCCTTACTCAGGTGAAATGGACT
TACTAGGAGAGATTCTTGATACATTGAGCACACACAGCTCAGATCAGGGGAAGCTGGC
AGCTGCAAAGAGCTTGGATTTCTTTAGATCAATGGATGACATTGATTACAAACCTACG
AATAAATCTAATGCTCCTAGTGAGAATAACCTGGCTTTCCTCTGTGGTGGTTCTGGTG
ACCAAGCAGAGTGGAATCTTGGGCAAGACGATAGTGCCCTCCATGGCAAACACCTCCC
TCCATCTCCTAGGAAGCGGGTTTCCTCTAGTGGTTTGACAGATTCTCTGTTTATCCTG
GACTGGATTTCCAACTCACTTCCCCTGAAGTTTCCCAGACTGATAAAGGAAAAACAGA
AAAGAGGGAAACACTAAGCCAGATTTCAGATGATCTGCTTATACCCGGTCTTGGGCGG
CATTCATCGACTTTTGTTCCTTGGGAGAAAGAAGGGAAAGAAGCCAAAGAGACTTCAG
AAGATATTGGACTGCTCCATGAAGTAGTGTCATTATGTCATATGACATCTGACTTCCA
ACAAAGCTTGAACATTTCAGACAAAAACACAAATGGAAACCAAACTAGATCT
ORF Start: at 1 _ ORF Stop: end of sequence ~~

SEQ II) NO: 68 752 as MW at 83933.6kD

NOVIIe, RSDPWLWKFPEDFGDQEILQSVPKFCFPFDVERVSQNQVGQHFTFVLTDIESKQRFG

PT'OtelnNTPVNLSVNQEIFIACEQVLKDQPALVPHSYFIAPDVTGLPTIPESRNLTEYFVAVDV

NNMLQLYASMLHERRIVIISSKLSTLTACIHGSAALLYPMYWQHIYIPVLPPHLLDYC

SeqllenCeCAPMPYLIGIHSSLIERVKNKSLEDVVMLNVDTNTLESPFSDLNNLPSDWSALKNKL

KKQSTATGDGVARAFLRAQAALFGSYRDALRYKPGEPITFCEESFVKHRSSVMKQFLE

TAINLQLFKQFIDGRLAKLNAGRGFSDVFEEEITSGGFCGGNPRSYQQWVHTVKKGGA

LFNTAMTKATPAVRTAYKFAKNHAKLGLKEVKSKLKHKENEEDYGTCSSSVQYTPWK

LHNEKGGNSEKRKLAQARLKRPLKSLDGALYDDEDDDDIERASKLSSEDGEEASAYLY

ESDDSVETRVKTPYSGEMDLLGEILDTLSTHSSDQGKLAAAKSLDFFRSMDDIDYKPT

NKSNAPSENNLAFLCGGSGDQAEWNLGQDDSALHGKHLPPSPRKRVSSSGLTDSLFIL

KEENSNKHLGADNVSDPTSGLDFQLTSPEVSQTDKGKTEKRETLSQISDDLLIPGLGR

HSSTFVPWEKEGKEAKETSEDIGLLHEWSLCHMTSDFQQSLNISDKNTNGNQTRS

Sequence comparison of the above protein sequences yields the following sequence relationships shown in Table 11B.
Table 11B. Comparison of NOVlla against NOVllb through NOVlle.
Protein SequenceNOVlla Residues/Identities/

Match ResiduesSimilarities for the Matched Region NOVIIb 29..731 ~ 662/752 (88%) 2..750 671/752 (89%) NOV 11 c 29..731 662/752 (88%) 2..750 671/752 (89%) NOVlld 29..731 658/753 (87%) 2..751 ~ 668/753 (88%) NOV 11 a 29..731 663/752 (88%) 2..750...... ..... ... . . 671/752.
_ . ~ (89%)..........

Further analysis of the NOV 11 a protein yielded the following properties shown in Table 11C.
Table 11C. Protein Sequence Properties NOVlla PSort 0.3700 probability located in outside; 0.1900 probability located in lysosome analysis: (lumen); 0.1304 probability located in microbody (peroxisome);
0.1000 probability located in endoplasmic reticulum (membrane) SignalP Cleavage site between residues 30 and 31 analysis:
A search of the NOV 11 a protein against the Geneseq database, a proprietary database that contains sequences published in patents and patent publication, yielded several homologous proteins shown in Table 11 D.

Table 11D. Geneseq Results for NOVlla NOVlla Identities/

Geneseq Protein/Organism/Length Residues/SimilaritiesExpect for Identifier[Patent #, Date] Match the Matched Value ResiduesRegion AAU82007Human secreted protein 8..430 289/454 (63%)e-163 Homo sapiens, 559 aa. 8..457 351/454 (76%) [W0200198353-A2, 27-DEC-2001 ]

AAM3971 Human polypeptide SEQ 8..430 289/454 (63%)e-163 S ID NO

2860 - Homo Sapiens, 8..457 351/454 (76%) 559 aa.

[W0200153312-Al, 26-JUL-2001 ]

AAM41501Human polypeptide SEQ 8..406 275/430 (63%)e-154 ID NO

6432 - Homo Sapiens, 13..438 330/430 (75%) 545 aa.

[W0200153312-A1, 26-JUL-2001]

ABG03235Novel human diagnostic 188..378137/192 (71%)4e-75 protein #3226 - Homo sapiens, I ..190 164/192 (85%) 196 aa.

[W0200175067-A2, 11-OCT-2001 ]

ABG03235Novel human diagnostic 188..378137/192 (71%)4e-75 protein #3226 - Homo sapiens, I .. 164/192 (85%) 196 aa. I 90 [W0200175067-A2, 11-OCT-2001]

In a BLAST search of public sequence datbases, the NOV 11 a protein was found to have homology to the proteins shown in the BLASTP data in Table I !E.

Table 11E. Public BLASTP Results for NOVlIa NOVlIa Identities/

Protein Residues/Similarities Expect for AccessionProteinlOrganism/LengthMatch the Matched Value Number ResiduesPortion Q9NXU2 CDNA FLJ20054 FIS, 393..731339/339 (100%)0.0 CLONE

COL00849 - Homo sapiens1..339 339/339 (100%) (Human), 339 aa.

AAH22561 HYPOTHETICAL 45.0 KDA 27..376 340/379 (89%)0.0 PROTEIN - Homo sapiens15..392 344/379 (90%) (Human), 396 aa.

Q9DSB9 4930571B16RIK PROTEIN 337..731298/407 (73%)e-167 -Mus musculus (Mouse), 96..499 337/407 (82%) 499 aa.

AAH27786 SIMILAR TO KIAA1608 8..623 342/680 (50%)e-166 PROTEIN - Mus musculus8..676 439/680 (64%) (Mouse), 1016 aa.

Q9H796 CDNA: FLJ21129 FIS, 8..426 2881450 (64%)e-162 CLONE

CAS06266 - Homo sapiens8..453 349/450 (77%) (Human), 559 aa.

PFam analysis predicts that the NOV1 la protein contains the domains shown in the Table 11F.
Table 11F. Domain Analysis of NOVlla Identities/
Pfam Domain NOVlla Match Region Similarities Expect Value for the Matched Region DENN 129..244 48/120 (40%) 6.4e 35 84/12 °
. .. _... ~.~70%). ~ .
EXAMPLE 12.
The NOV 12 clone was analyzed, and the nucleotide and encoded polypeptide sequences are shown in Table 12A.
Table 12A. NOV12 Sequence Analysis SEQ ID NO: 69 ~ 1357 NOVl2a, ATGAAACTAATTACCATCCTTTTCCTCTGCTCCAGGCTACTACTAAGTTTAACCCAGG
CG104903-Ol ~T~~GTCCGAGGAAATTGATGACTGCAATGACAAGGATTTATTTAAAGCTGTGGA
DNA TGCTGCTCTGAAGAAATATAACAGTCAAAACCAAAGTAACAACCAGTTTGTATTGTAC
CGCAAAACCTGGCAGGACTGTGAGTACAAGGATGCTGCAAAAGCAGCCACTGGAGAAT
Seguence ~GCACAGCAACCGTGGGGAAGAGGAGCAGTACGAAATTCTCCGTGGCTACCCAGACCTG

CCAGATTACTCCAGCCGAGGGCCCTGTGGTGACAGCCCAGTACGACTGCCTCGGCTGT
GTGCATCCTATATCAACGCAGAGCCCAGACCTGGAGCCCATTCTGAGACACGGCATTC
AGTACTTTAACAACAACACTCAACATTCCTCCCTCTTCACGCTTAATGAAGTAAAACG
GGCCCAAAGACAGGTGGTGGCTGGATTGAACTTTCGAATTACCTACTCAATTGTGCAA
ACGAATTGTTCCAAAGAGAATTTTCTGTTCTTAACTCCAGACTGCAAGTCCCTTTGGA
ATGGTGATACCGGTGAATGTACAGATAATGCATACATCGATATTCAGCTACGAATTGC
TTCCTTCTCACAGAACTGTGACATTTATCCAGGGAAGGATTTTGTACAACCACCTACC
AAGATTTGCGTGGGCTGCCCCAGAGATATACCCACCAACAGCCCAGAGCTGGAGGAGA
CACTGACTCACACCATCACAAAGCTTAATGCAGAGAATAACGCAACTTTCTATTTCAA
GATTGACAATGTGF.AAAAAGCAAGAGTACAGGTGGTGGCTGGCAAGAAATATTTTATT
GACTTCGTGGCCAGGGAAACCACATGTTCCAAGGAAAGTAATGAAGAGTTGACCGAAA
GCTGTGAGACCAAAAAACTTGGCCAAAGCCTAGATTGCAACGCTGAAGTTTATGTGGT
ACCCTGGGAGAAAAAAATTTACCCTACTGTCAACTGTCAACCACTGGGAATGATCTCA
CTGATGAAAAGGCCTCCAGGTTTTTCACCTTTCCGATCATCACGAATAGGGGAAATAA
AAGAAGAAACAACTAGTCACCTAAGGTCCTGCGAGTACAAGGGTCGACCCCCAAAGGC
AGGGGCAGAGCCAGCATCTGAGAGGGAGGTCTCTTGACCAATGGGCAGAATCTTCACT
CCAGGCACATAGCCCCAACCACCTCTGCCAGCAACCTTGAGAGGAAGGACAAGAAGAA
AGATGGGATAGAATTTAAATAGAGAAGAATGCCATTTTATCACTCTGCCTCTGGGTGA
AATAAAGATCAGTCTTGATGTTC
ORF Start: ATG at 1 ORF Stop: TGA at 1195 SEQ ID NO: 70 398 as , ~MW at 44684.1kD
NOVl2a, MKLITILFLCSRLLLSLTQESQSEEIDDCNDKDLFKAVDAALKKYNSQNQSNNQFVLY
CG104903-Ol ~'KTWQDCEYKDAAKAATGECTATVGKRSSTKFSVATQTCQITPAEGPVVTAQYDCLGC
P1'otelri ~PISTQSPDLEPILRHGIQYFNNNTQHSSLFTLNEVKRAQRQWAGLNFRITYSIVQ
TNCSKENFLFLTPDCKSLWNGDTGECTDNAYIDIQLRIASFSQNCDIYPGKDFVQPPT
SeCjllBriCe KICVGCPRDIPTNSPELEETLTHTITKLNAENNATFYFKIDNVKKARVQVVAGKKYFI
DFVARETTCSKESNEELTESCETKKLGQSLDCNAEVYWPWEKKIYPTVNCQPLGMIS
LMKRPPGFSPFRSSRIGEIKEETTSHLRSCEYKGRPPKAGAEPASEREVS
SEQ ID NO: 71 1848 by NOVI~b, ATGAAACTAATTACCATCCTTTTCCTCTGCTCCAGGCTACTACTAAGTTTAACCCAGG
CG104903-02 ~TCACAGTCCGAGGAAATTGATGACTGCAATGACAAGGATTTATTTAAAGCTGTGGA
DNA TGCTGCTCTGAAGAAATATAACAGTCAAAACCAAAGTAACAACCAGTTTGTATTGTAC
CGCAAAACCTGGCAGGACTGTGAGTACAAGGATGCTGCAAAAGCAGCCACTGGAGAAT
SeC~LleriCe GCACAGCAACCGTGGGGAAGAGGAGCAGTACGAAATTCTCCGTGGCTACCCAGACCTG
CCAGATTACTCCAGCCGAGGGCCCTGTGGTGACAGCCCAGTACGACTGCCTCGGCTGT
GTGCATCCTATATCAACGCAGAGCCCAGACCTGGAGCCCATTCTGAGACACGGCATTC
AGTACTTTAACAACAACACTCAACATTCCTCCCTCTTCACGCTTAATGAAGTAAAACG
GGCCCAAAGACAGGTGGTGGCTGGATTGAACTTTCGAATTACCTACTCAATTGTGCAA
ACGAATTGTTCCAAAGAGAATTTTCTGTTCTTAACTCCAGACTGCAAGTCCCTTTGGA
ATGGTGATACCGGTGAATGTACAGATAATGCATACATCGATATTCAGCTACGAATTGC
TTCCTTCTCACAGAACTGTGACATTTATCCAGGGAAGGATTTTGTACAACCACCTACC
AAGATTTGCGTGGGCTGCCCCAGAGATATACCCACCAACAGCCCAGAGCTGGAGGAGA
CACTGACTCACACCATCACAAAGCTTAATGCAGAGAATAACGCAACTTTCTATTTCAA
GATTGACAATGTGAAAAAAGCAAGAGTACAGGTGGTGGCTGGCAAGAAATATTTTATT
GACTTCGTGGCCAGGGAAACCACATGTTCCAAGGAAAGTAATGAAGAGTTGACCGAAA
GCTGTGAGACCAAAAAACTTGGCCAAAGCCTAGATTGCAACGCTGAAGTTTATGTGGT
ACCCTGGGAGAAAAAAATTTACCCTACTGTCAACTGTCAACCACTGGGAATGATCTCA
CTGATGAAAAGGCCTCCAGGTTTTTCACCTTTCCGATCATCACGAATAGGGGAAATAA
AAGAAGAAACAACTGTAAGTCCACCCCACACTTCCATGGCACCTGCACAAGATGAAGA
GCGGGATTCAGGAAAAGAACAAGGGCATACTCGTAGACATGACTGGGGCCATGAAAAA
CAAAGAAAACATAATCTTGGCCATGGCCATAAACATGAACGTGACCAAGGGCATGGGC
ACCAAAGAGGACATGGCCTTGGCCATGGACACGAACAACAGCATGGTCTTGGTCATGG
ACATAAGTTCAAACTTGATGATGATCTTGAACACCAAGGGGGCCATGTCCTTGACCAT
GGACATAAGCATAAGCATGGTCATGGCCACGGAAAACATAAAAATAAAGGCAAAAAGA
ATGGAAAGCACAATGGTTGGAAAACAGAGCATTTGGCAAGCTCTTCTGAAGACAGTAC
TACACCTTCTGCACAGACACAAGAGAAGACAGAAGGGCCAACACCCATCCCTTCCCTA
GCCAAGCCAGGTGTAACAGTTACCTTTTCTGACTTTCAGGACTCTGATCTCATTGCAA
CTATGATGCCTCCTATATCACCAGCTCCCATACAGAGTGATGACGATTGGATCCCTGA

TATCCAGACAGACCCAAATGGCCTTTCATTTAACCCAATATCAGATTTTCCAGACACG

ACCTCCCCAAAATGTCCTGGACGCCCCTGGAAGTCAGTTAGTGAAATTAATCCAACCA

CACAAATGAAAGAATCTTATTATTTCGATCTCACTGATGGCCTTTCTTAA

ORF Start: ATG at 1 ORF Stop: TAA at 1846 SEQ ID NO: 72 615 as MW at 68746.1kD

NOVl2b, MKLITILFLCSRLLLSLTQESQSEEIDDCNDKDLFKAVDAALKKYNSQNQSNNQFVLY

P1'Oteln VHPISTQSPDLEPILRHGIQYFNNNTQHSSLFTLNEVKRAQRQVVAGLNFRITYSIVQ

TNCSKENFLFLTPDCKSLWNGDTGECTDNAYIDIQLRIASFSQNCDIYPGKDFVQPPT

SequeriCeKICVGCPRDIPTNSPELEETLTHTITKLNAENNATFYFKIDNVKKARVQVVAGKKYFI

DFVARETTCSKESNEELTESCETKKLGQSLDCNAEVYWPWEKKIYPTVNCQPLGMIS

LMKRPPGFSPFRSSRIGEIKEETTVSPPHTSMAPAQDEERDSGKEQGHTRRHDWGHEK

QRKHNLGHGHKHERDQGHGHQRGHGLGHGHEQQHGLGHGHKFKLDDDLEHQGGHVLDH

GHKHKHGHGHGKHKNKGKKNGKHNGWKTEHLASSSEDSTTPSAQTQEKTEGPTPIPSL

AKPGVTVTFSDFQDSDLIATMMPPISPAPIQSDDDWIPDIQTDPNGLSFNPISDFPDT

TSPKCPGRPWKSVSEINPTTQMKESYYFDLTDGLS

SEQ ID NO: 73 1981 by NOV12C, AA'rTCCGGTTGAAACCATCCCTCAGCTCCTAGAGGGAGATTGTTAGATCATGAAACTA

CG104903-03~'TTACCATCCTTTTCCTCTGCTCCAGGCTACTACTAAGTTTAACCCAGGAATCACAGT

DNA CCGAGGAAATTGACTGCAATGACAAGGATTTATTTAAAGCTGTGGATGCTGCTCTGAA

GAAATATAACAGTCAAAACCAAAGTAACAACCAGTTTGTATTGTACCGCATAACTGAA

Sequence GCCACTAAGACGGTTGGCTCTGACACGTTTTATTCCTTCAAGTACGAAATCAAGGAGG

GGGATTGTCCTGTTCAAAGTGGCAAAACCTGGCAGGACTGTGAGTACAAGGATGCTGC

AAAAGCAGCCACTGGAGAATGCACGGCAACCGTGGGGAAGAGGAGCAGTACGAAATTC

TCCGTGGCTACCCAGACCTGCCAGATTACTCCAGCCGAGGGCCCTGTGGTGACAGCCC

AGTACGACTGCCTCGGCTGTGTGCATCCTATATCAACGCAGAGCCCAGACCTGGAGCC

CATTCTGAGACACGGCATTCAGTACTTTAACAACAACACTCAACATTCCTCCCTCTTC

ATGCTTAATGAAGTAAAACGGGCCCAAAGACAGGTGGTGGCTGGATTGAACTTTCGAA

TTACCTACTCAATTGTGCAAACGAATTGTTCCAAAGAGAATTTTCTGTTCTTAACTCC

AGACTGCAAGTCCCTTTGGAATGGTGATACCGGTGAATGTACAGATAATGCATACATC

GATATTCAGCTACGAATTGCTTCCTTCTCACAGAACTGTGACATTTATCCAGGGAAGG

ATTTTGTACAACCACCTACCAAGATTTGCGTGGGCTGCCCCAGAGATATACCCACCAA

CAGCCCAGAGCTGGAGGAGACACTGACTCACACCATCACAAAGCTTAATGCAGAGAAT

AACGCAACTTTCTATTTCAAGATTGACAATGTGAAAAAAGCAAGAGTACAGGTGGTGG

CTGGCAAGAAATATTTTATTGACTTCGTGGCCAGGGAAACCACATGTTCCAAGGAAAG

TAATGAAGAGTTGACCGAAAGCTGTGAGACCAAAAAACTTGGCCAAAGCCTAGATTGC

AACGCTGAAGTTTATGTGGTACCCTGGGAGAAAAAAATTTACCCTACTGTCAACTGTC

AACCACTGGGAATGATCTCACTGATGAAAAGGCCTCCAGGTTTTTCACCTTTCCGATC

ATCACGAATAGGGGAAATAAAAGAAGAAACAACTGTAAGTCCACCCCACACTTCCATG

GCACCTGCACAAGATGAAGAGCGGGATTCAGGAAAAGAACAAGGGCATACTCGTAGAC

ATGACTGGGGCCATGAAAAACAAAGAAAACATAATCTTGGCCATGGCCATAAACATGA

ACGTGACCAAGGGCATGGGCACCAAAGAGGACATGGCCTTGGCCATGGACACGAACAA

CAGCATGGTCTTGGTCATGGACATAAGTTCAAACTTGATGATGATCTTGAACACCAAG

GGGGCCATGTCCTTGACCATGGACATAAGCATAAGCATGGTCATGGCCACGGAAAACA

TAAAAATAAAGGCAAAAAGAATGGAAAGCACAATGGTTGGAAAACAGAGCATTTGGCA

AGCTCTTCTGAAGACAGTACTACACCTTCTGCACAGACACAAGAGAAGACAGAAGGGC

CAACACCCATCCCTTCCCTAGCCAAGCCAGGTGTAACAGTTACCTTTTCTGACTTTCA

GGACTCTGATCTCATTGCAACTATGATGCCTCCTATATCACCAGCTCCCATACAGAGT

GATGACGATTGGATCCCTGATATCCAGATAGACCCAAATGGCCTTTCATTTAACCCAA

TATCAGATTTTCCAGACACGACCTCCCCAAAATGTCCTGGACGCCCCTGGAAGTCAGT

TAGTGAAATTAATCCAACCACACAAATGAAAGAATCTTATTATTTCGATCTCACTGAT

GGCCTTTCT

ORF Start: ATG at 50 ORF Stop: end of sequence SEQ ID NO: 74 644 as MW at 71956.8kD
NOV12C, MKLITILFLCSRLLLSLTQESQSEEIDCNDKDLFKAVDAALKKYNSQNQSNNQFVLYR

PrOtelri TKFSVATQTCQITPAEGPWTAQYDCLGCVHPISTQSPDLEPILRHGIQYFNNNTQHS

S8CILIeriCeSLFMLNEVKRAQRQWAGLNFRITYSIVQTNCSKENFLFLTPDCKSLWNGDTGECTDN

AYIDIQLRIASFSQNCDIYPGKDFVQPPTKICVGCPRDIPTNSPELEETLTHTITKLN

AENNATFYFKIDNVKKARVQWAGKKYFIDFVARETTCSKESNEELTESCETKKLGQS

LDCNAEVYWPWEKKIYPTVNCQPLGMISLMKRPPGFSPFRSSRIGEIKEETTVSPPH

TSMAPAQDEERDSGKEQGHTRRHDWGHEKQRKHNLGHGHKHERDQGHGHQRGHGLGHG

HEQQHGLGHGHKFKLDDDLEHQGGHVLDHGHKHKHGHGHGKHKNKGKKNGKHNGWKTE

HLASSSEDSTTPSAQTQEKTEGPTPIPSLAKPGVTVTFSDFQDSDLIATMMPPISPAP

IQSDDDWIPDIQIDPNGLSFNPTSDFPDTTSPKCPGRPWKSVSEINPTTQMKESYYFD

LTDGLS

SEQ ID NO: 75 1297 by NOV12CI, AATTCCGGTTGAAACCATCCCTCAGCTCCTAGAGGGAGATTGTTAGATCATGAAACTA

DNA CCGAGGAAATTGACTGCAATGACAAGGATTTATTTAAAGCTGTGGATGCTGCTCTGAA

GAAATATAACAGTCAAAACCAAAGTAACAACCAGTTTGTATTGTACCGCATAACTGAA

SeChlBriCeGCCACTAAGACGGTTGGCTCTGACACGTTTTATTCCTTCAAGTACGAAATCAAGGAGG

GGGATTGTCCTGTTCAAAGTGGCAAAACCTGGCAGGACTGTGAGTACAAGGATGCTGC

AAAAGCAGCCACTGGAGAATGCACAGCAACCGTGGGGAAGAGGAGCAGTACGAAATTC

TCCGTGGCTACCCAGACCTGCCAGATTACTCCAGCCGAGGGCCCTGTGGTGACAGCCC

AGTACGACTGCCTCGGCTGTGTGCATCCTATATCAACGCAGAGCCCAGGTTTTTCP.CC

TTTCCGATCATCACGAATAGGGGAAATAAAAGAAGAAACAACTGTAAGTCCACCCCAC

ACTTCCATGGCACCTGCACAAGATGAAGAGCGGGATTCAGGAAAAGAACAAGGGCATA

CTCGTAGACATGACTGGGGCCATGAAAAACAAAGAAAACATAATCTTGGCCATGGCCA

TAAACATGAACGTGACCAAGGGCATGGGCACCAAAGAGGACATGGCCTTGGCCATGGA

CACGAACAACAGCATGGTCTTGGTCATGGACATAAGTTCAAACTTGATGATGATCTTG

AACACCAAGGGGGCCATGTCCTTGACCATGGACATAAGCATAAGCATGGTCATGGCCA

CGGAAAACATAAAAATAAAGGCAAAAAGAATGGAAAGCACAATGGTTGGAAAACAGAG

CATTTGGCAAGCTCTTCTGAAGACAGTACTACACCTTCTGCACAGACACAAGAGAAGA

CAGAAGGGCCAACACCCATCCCTTCCCTAGCCAAGCCAGGTGTAACAGTTACCTTTTC

TGACTTTCAGGACTCTGATCTCATTGCAACTATGATGCCTCCTATATCACCAGCTCCC

ATACAGAGTGATGACGATTGGATCCCTGATATCCAGATAGACCCAAATGGCCTTTCAT

TTAACCCAATATCAGATTTTCCAGACACGACCTCCCCAAAATGTCCTGGACGCCCCTG

GAAGTCAGTTAGTGAAATTAATCCAACCACACAAATGAAAGAATCTTATTATTTCGAT

CTCACTGATGGCCTTTCTTAA

ORF Start: ATG at 50 ORF
Stop:
TAA
at SEQ ID NO: 76 415 MW at 45897.3kD
as NOV12C~, MKLITILFLCSRLLLSLTQESQSEEIDCNDKDLFKAVDAALKKYNSQNQSNNQFVLYR

PIOtelri TKFSVATQTCQITPAEGPWTAQYDCLGCVHPISTQSPGFSPFRSSRIGEIKEETTVS

PPHTSMAPAQDEERDSGKEQGHTRRHDWGHEKQRKHNLGHGHKHERDQGHGHQRGHGL

SCCjlleriCCGHGHEQQHGLGHGHKFKLDDDLEHQGGHVLDHGHKHKHGHGHGKHKNKGKKNGKHNGW

KTEHLASSSEDSTTPSAQTQEKTEGPTPIPSLAKPGVTVTFSDFQDSDLIATMMPPIS

PAPIQSDDDWIPDIQIDPNGLSFNPTSDFPDTTSPKCPGRPWKSVSEINPTTQMKESY

YFDLTDGLS

SEQ ID NO: 77 1892 by NOVl2e, AATTCCGGTTGAAACCATCCCTCAGCTC_CTAGAGGGAGATTGTTAGATCATGAAACTA

DNA CCGAGGAAATTGACTGCAATGACAAGGATTTATTTAAAGCTGTGGATGCTGCTCTGAA

GAAATATAACAGTCAAAACCAAAGTAACAACCAGTTTGTATTGTACCGCATAACTGAA

SeC1L18riCeGCCACTAAGACGGTTGGCTCTGACACGTTTTATTCCTTCAAGTACGAAATCAAGGAGG

GGGATTGTCCTGTTCAAAGTGGCAAAACCTGGCAGGACTGTGAGTACAAGGATGCTGC

AAAAGCAGCCACTGGAGAATGCACAGCAACCGTGGGAAGAGGAGCAGTACGAAATTCT

CCGTGGCTACCCAGACCTGGAGCCCATTCTGAGACACGGCATTCAGTACTTTAACAAC

AACACTCAACATTCCTCCCTCTTCACGCTTAATGAAGTAAAACGGGCCCAAAGACAGG

TGGTGGCTGGATTGAACTTTCGAATTACCTACTCAATTGTGCAAACGAATTGTTCCAA

AGAGAATTTTCTGTTCTTAACTCCAGACTGCAAGTCCCTTTGGAATGGTGATACCGGT

GAATGTACAGATAATGCATACATCGATATTCAGCTACGAATTGCTTCCTTCTCACAGA

ACTGTGACATTTATCCAGGGAAGGATTTTGTACAACCACCTACCAAGATTTGCGTGGG
CTGCCCCAGAGATATACCCACCAACAGCCCAGAGCTGGAGGAGACACTGACTCACACC
ATCACAAAGCTTAATGCAGAGAATAACGCAACTTTCTATTTCAAGATTGACAATGTGA
ACCCT
GTAAGTCCACCCCACACTTCCATGGCACCTGCACAAGATGAAGAGCGGGATTCAGGAA
GGCCTTGGCCATGGACACGAACAACAGCATGGTCTTGGTCATGGACATAAGTTCAAAC
TTGATGATGATCTTGAACACCAAGGGGGCCATGTCCTTGACCATGGACATAAGCATAA
GCATGGTCATGGCCACGGAAAACATAAAAATAAAGGCAAAAAGAATGGAAAGCACAAT
CAAATGGCCTTTCATTTAACCCAATATCAGATTTTCCAGACACGACCTCCCCAAAATG
TCCTGGACGCCCCTGGAAGTCAGTTAGTGAAATTAATCCAACCACACAAATGAAAGAA
TCTTATTATTTCGATCTCACTGATGGCCTTTCTTAA
ORF Start: ATG at 50 ORF Stop: TAA at 458 SEQ ID NO: 78 136 as MW at I 5218.9kD
VI2e, MKLITILFLCSRLLLSLTQESQSEEIDCNDKDLFKAVDAALKKYNSQNQSNNQFVLYR

RNSPWLPRPGAHSETRHSVL
Sequence SEQ ID NO: 79 670 NOVl2f, ~ATGAAACTAATTACCATCCTTTTCCTCTGCTCCAGGCTACTACTAAGTTTAACCCAGG
CG104903-07 ~'TCACAGTCCGAGGAAATTGATGACTGCAATGACAAGGATTTATTTAAAGCTGTGGA
DNA TGCTGCTCTGAAGAAATATAACAGTCAAAACCAAAGTAACAACCAGTTTGTATTGTAC
CGCAAAACCTGGCAGGACTGTGAGTACAAGGATGCTGCAAAAGCAGCCACTGGAGAAT
Sequence GCACAGCAACCGTGGGGAAGAGGAGCAGTACGAAATTCTCCGTGGCTACCCAGACCTG
CCAGATTACTCCAGCCGAGGGCCCTGTGGTGACAGCCCAGTACGACTGCCTCGGCTGT
GTGCATCCTATATCAACGCAGAGCCCAGGTTTTTCACCTTTCCGATCATCACGAATAG
~GGGAAATAAAAGAAGAAACAACTAGTCACCTAAGGTCCTGCGAGTACAAGGGTCGACC
CCCAAAGGCAGGGGCAGAGCCAGCATCTGAGAGGGAGGTCTCTTGACCAATGGGCAGA
ORF Start: ATG at 1 ORF Stop:.TGA at 508_ SEQ ID NO: 80 w~ 169 aa~MW at 18654.7kD
Vl2f, MKLITILFLCSRLLLSLTQESQSEEIDDCNDKDLFKAVDAALKKYNSQNQSNNQFVLY

VHPISTQSPGFSPFRSSRTGEIKEETTSALRSCEYKGRPPKAGAEPASEREVS
Sequence SEQ ID NO: 81 1193 Vl2g, ATGAAACTAATTACCATCCTTTTCCTCTGCTCCAGGCTACTACTAAGTTTAACCCAGG
104903-08'~T~CAGTCCGAGGAAATTGACTGCAATGACAAGGATTTATTTAAAGCTGTGGATGC
ATAACTGAAGCCACTAAGACGGCCACTGGAGAATGCACGGCAACCGTGGGGAAGAGGA
GCAGTACGAAATTCTCCGTGGCTACCCAGACCTGCCAGATTACTCCAGCCGAGGGCCC
CCAGACCTGGAGCCCATTCTGAGACACGGCATTCAGTACTTTAACAACAACACTCAAC

ATTCCTCCCTCTTCACGCTTAATGAAGTAAAACGGGCCCAAAGACAGGTGGTGGCTGG
ATTGAACTTTCGAATTACCTACTCAATTGTGCAAACGAATTGTTCCAAAGAGAATTTT
CTGTTCTTAACTCCAGACTGCGAGTCCCTTTGGAATGGTGATACCGGTGAATGTACAG
ATAATGCATACATCGATATTCAGCTACGAATTGCTTCCTTCTCACAGAACTGTGACAT
TTATCCAGGGAAGGATTTTGTACAACCACCTACCAAGATTTGCGTGGGCTGCCCCAGA
GATATACCCACCAACAGCCCAGAGCTGGAGGAGACACTGACTCACACCATCACAAAGC
TTAATGCAGAGAATAACGCAACTTTCTATTTCAAGATTGACAATGTGAAAAAAGCAAG
AGTACAGGTGGTGGCTGGCAAGAAATATTTTATTGACTTCGTGGCCAGGGAAACCACA
TGTTCCAAGGAAAGTAATGAAGAGTTGACCGAAAGCTGTGAGACCAAAAAACTTGGCC
AAAGCCTAGATTGCAACGCTGAAGTTTATGTGGTACCCTGGGAGP~AAAAAATTTACCC
TACTGTCAACTGTCAACCACTGGGAATGATCTCACTGATGAAAAGGCCTCCAGGTTTT
TCACCTTTCCGATCATCACGAATAGGGGAAATAAAAGAAGAAACAACTAGTCACCTAA
GGTCCTGCGAGTACAAGGGTCGACCCCCAAAGGCAGGGGCAGAGCCAGTATCTGAGAG
GGAGGTCTCTTGACCAATGGGCAGAATCTTCAC
ORF Start: ATG at 1 ORF Stop: TGA at 1171 SEQ ID NO: 82 390 as MW at 43704.OkD
NOVl2g, MKLITILFLCSRLLLSLTQESQSEEIDCNDKDLFKAVDAALKKYNSQNQSNNQFVLYR

Pi'Otelri PDLEPILRHGIQYFNNNTQHSSLFTLNEVKRAQRQWAGLNFRITYSIVQTNCSKENF
LFLTPDCESLWNGDTGECTDNAYIDIQLRIASFSQNCDIYPGKDFVQPPTKICVGCPR
SeqlleriC~ DIPTNSPELEETLTHTITKLNAENNATFYFKIDNVKKARVQWAGKKYFIDFVARETT
CSKESNEELTESCETKKLGQSLDCNAEVYWPWEKKIYPTVNCQPLGMISLMKRPPGF
SPFRSSRIGEIKEETTSHLRSCEYKGRPPKAGAEPVSEREVS
SEQ ID NO: 83 1984 by NOVl2h, AATTCCGGTTGAAACCATCCCTCAGCTCCTAGAGGGAGATTGTTAGATCATGAAACTA

DNA CCGAGGAAATTGACTGCAATGACAAGGATTTATTTAAAGCTGTGGATGCTGCTCTGAA
GAAATATAACAGTCAAAACCAAAGTAACAACCAGTTTGTATTGTACCGCATAACTGAA
SeqLleriCe GCCACTAAGACGGTTGGCTCTGACACGTTTTATTCCTTCAAGTACGAAATCAAGGAGG
GGGATTGTCCTGTTCAAAGTGGCAAAACCTGGCAGGACTGTGAGTACAAGGATGCTGC
AAAAGCAGCCACTGGAGAATGCACGGCAACCGTGGGGAAGAGGAGCAGTACGAAATTC
TCCGTGGCTACCCAGACCTGCCAGATTACTCCAGCCGAGGGCCCTGTGGTGACAGCCC
AGTACGACTGCCTCGGCTGTGTGCATCCTATATCAACGCAGAGCCCAGACCTGGAGCC
CATTCTGAGACACGGCATTCAGTACTTTAACAACAACACTCAACATTCCTCCCTCTTC
ATGCTTAATGAAGTAAAACGGGCCCAAAGACAGGTGGTGGCTGGATTGAACTTTCGAA
TTACCTACTCAATTGTGCAAACGAATTGTTCCAAAGAGAATTTTCTGTTCTTAACTCC
AGACTGCAAGTCCCTTTGGAATGGTGATACCGGTGAATGTACAGATAATGCATACATC
GATATTCAGCTACGAATTGCTTCCTTCTCACAGAACTGTGACATTTATCCAGGGAAGG
ATTTTGTACAACCACCTACCAAGATTTGCGTGGGCTGCCCCAGAGATATACCCACCAA
CAGCCCAGAGCTGGAGGAGACACTGACTCACACCATCACAAAGCTTAATGCAGAGAAT
AACGCAACTTTCTATTTCAAGATTGACAATGTGAAAAAAGCAAGAGTACAGGTGGTGG
CTGGCAAGAAATATTTTATTGACTTCGTGGCCAGGGAAACCACATGTTCCAAGGAAAG
TAATGAAGAGTTGACCGAAAGCTGTGAGACCAAAAAACTTGGCCAAAGCCTAGATTGC
AACGCTGAAGTTTATGTGGTACCCTGGGAGAAAAAAATTTACCCTACTGTCAACTGTC
AACCACTGGGAATGATCTCACTGATGAAAAGGCCTCCAGGTTTTTCACCTTTCCGATC
ATCACGAATAGGGGAAATAAAAGAAGAAACAACTGTAAGTCCACCCCACACTTCCATG
GCACCTGCACAAGATGAAGAGCGGGATTCAGGAAAAGAACAAGGGCATACTCGTAGAC
ATGACTGGGGCCATGAAAAACAAAGAAAACATAATCTTGGCCATGGCCATAAACATGA
ACGTGACCAAGGGCATGGGCACCAAAGAGGACATGGCCTTGGCCATGGACACGAACAA
CAGCATGGTCTTGGTCATGGACATAAGTTCAAACTTGATGATGATCTTGAACACCAAG
GGGGCCATGTCCTTGACCATGGACATAAGCATAAGCATGGTCATGGCCACGGAAAACA
TAAAAATAAAGGCAAAAAGAATGGAAAGCACAATGGTTGGAAAACAGAGCATTTGGCA
AGCTCTTCTGAAGACAGTACTACACCTTCTGCACAGACACAAGAGAAGACAGAAGGGC
CAACACCCATCCCTTCCCTAGCCAAGCCAGGTGTAACAGTTACCTTTTCTGACTTTCA
GGACTCTGATCTCATTGCAACTATGATGCCTCCTATATCACCAGCTCCCATACAGAGT
GATGACGATTGGATCCCTGATATCCAGATAGACCCAAATGGCCTTTCATTTAACCCAA
TATCAGATTTTCCAGACACGACCTCCCCAAAATGTCCTGGACGCCCCTGGAAGTCAGT
TAGTGAAATTAATCCAACCACACAAATGAAAGAATCTTATTATTTCGATCTCACTGAT

GGCCTTTCTTAA
ORF Start: ATG at 50 IORF Stob: TAA at 1982 SEQ ID NO: 84 X644 as BMW at 71956.81cD
NOVl2h, ~MKLITILFLCSRLLLSLTQESQSEEIDCNDKDLFKAVDAALKKYNSQNQSNNQFVLYR

PrOteln TKFSVATQTCQITPAEGPWTAQYDCLGCVHPISTQSPDLEPILRHGIQYFNNNTQHS

SLFMLNEVKRAQRQWAGLNFRITYSIVQTNCSKENFLFLTPDCKSLWNGDTGECTDN

SeqllenCeAYIDIQLRIASFSQNCDIYPGKDFVQPPTKICVGCPRDIPTNSPELEETLTHTITKLN

AENNATFYFKIDNVKKARVQWAGKKYFIDFVARETTCSKESNEELTESCETKKLGQS

~LDCNAEVYWPWEKKIYPTVNCQPLGMISLMKRPPGFSPFRSSRIGEIKEETTVSPPH

HLASSSEDSTTPSAQTQEKTEGPTPIPSLAKPGVTVTFSDFQDSDLIATMMPPISPAP
IQSDDDWIPDIQIDPNGLSFNPISDFPDTTSPKCPGRPWKSVSEINPTTQMKESYYFD
LTDGLS
Sequence comparison of the above protein sequences yields the following sequence relationships shown in Table 12B.

Table 12B. Comparison of NOVl2a against NOVl2b through NOVl2h.
Protein SequenceNOVl2a Residues/Identities/

Match ResiduesSimilarities for the Matched Region ~

NOV 12b 26..396 343/371 (92%) 26..387 344/371 (92%) NOV 12c 28..396 3401399 (85%) 27..416 341/399 (85%) NOV 12d 28..129 90/132 (68%) 27..158 90/132 (68%) NOV 12e 28..84 47/87 (54%) 27..113 48/87 (55%) NOV 12f 26..129 ~ 92/104 (88%) 26:.129 92/104 (88%) NOV 12g 28..398 349/371 (94%) 27..390 351/371 (94%) NOV 12h 28..396 340/399 (85%) 2?..416 341/399 (85%) Further analysis of the NOV 12a protein yielded the following properties shown in Table 12C.
Table 12C. Protein Sequence Properties NOVl2a PSort 0.5135 probability located in outside; 0.1900 probability located in lysosome analysis: (lumen); 0.1000 probability located in endoplasmic reticulum (membrane);
0.1000 probability located in endoplasmic reticulum (lumen) SignalP Cleavage site between residues 24 and 25 analysis:
A search of the NOV 12a protein against the Geneseq database, a proprietary database that contains sequences published in patents and patent publication, yielded several homologous proteins shown in Table 12D.

Table 12D. Geneseq Results for NOVl2a NOVl2a Identities/

Geneseq Protein/Organism/Length Residues/SimilaritiesExpect for Identifier[Patent #, Date] Match the Matched Value ResiduesRegion ABG21101Novel human diagnostic 1..396 375/426 (88%)0.0 protein #21092 - Homo Sapiens, 1..416 376/426 (88%) 644 aa.

[W0200175067-A2, I1-OCT-2001 ]

ABG21101Novel human diagnostic 1..396 375/426 (88%)0.0 protein #21092 - Homo Sapiens, 1..416 376/426 (88%) 644 aa.

[W02001?5067-A2, I l-OCT- ' 2001]

ABG21105Novel human diagnostic 1..398 377/435 (86%)0.0 protein #21096 - Homo Sapiens, 2..435 380/435 (86%) 435 aa.

[W0200175067-A2, I1-OCT-2001 ]

ABG21105Novel human diagnostic 1..398 377/435 (86%)0.0 protein #21096 - Homo sapiens, 2..435 3801435 (86%}
435 aa.

[W0200175067-A2, l 1-OCT-AAP40257Bradykinin protein precursor:1..398 297/428 (69%)e-174 type I (pKGl3, pK.G59), 436 1..426 343/428 (79%) aa.

[JP59125896-A, 20-JUL-1984]

In a BLAST search of public sequence datbases, the NOV 12a protein was found to have homology to the proteins shown in the BLASTP data in Table 12E.

Table 12E. Public BLASTP Results for NOVl2a NOVl2a Identities/

Protein Residues/SimilaritiesExpect for AccessionProtein/Organism/Length Match the Matched Value Number ResiduesPortion KGHULI kininogen, LMW precursor1..398 3961428 (92%)0.0 [validated] - human, 1..427 396/428 (92%) 427 aa.

P01042 Kininogen precursor (Alpha-2-thiol1..396 375/426 (88%)0.0 proteinase inhibitor) 1..416 376/426 (88%) [Contains:

Bradykinin] - Homo sapiens (Human), 644 aa.

P01046 Kininogen, LMW I precursor1..398 297/428 (69%)e-173 (Thiol proteinase inhibitor)1..426 343/428 (79%) [Contains: Bradykinin]
- Bos taurus (Bovine), 436 aa.

P01047 Kininogen, LMW II precursor1..398 292/428 (68%), e-170 (Thiol proteinase inhibitor)1..424 340/428 (79%) [Contains: Bradykinin]
- Bos taurus (Bovine), 434 aa.

P01044 Kininogen, HMW I precursor1..375 280/405 (69%)e-I61 (Thiol proteinase inhibitor)1..403 321/405 (79%) [Contains: Bradykinin]
- Bos taurus (Bovine), 621 aa. .

PFam analysis predicts that the NOV 12a protein contains the domains shown in the Table 12F.
Table 12F. Domain Analysis of NOVl2a Identities/

Pfam DomainNOVl2a Match RegionSimilarities Expect Value for the Matched Region cystatin 21..59 11/40 (28%) 1.9e-06 35/40 (88%) cystatin 60..97 14/40 (35%) 4e-07 30/40 (75%) cystatin 115..219 28/113 (25%) Se-35 92/113 (81%) cystatin 237..341 32/113 (28%) 3.4e-39 94/113 (83%) EXAMPLE 13.
The NOV 13 clone was analyzed, and the nucleotide and encoded polypeptide sequences are shown in Table 13A.
Table 13A. NOV13 Sequence Analysis SEQ ID NO: 85 ~ 1272 by NOVl3a, ~CTTCCCCAGGACTCCAGGAGACATAAAACTTGAAACGGGAGACTTCGTGCAAATCCTG

DNA GGCGAATGTGACTCTCCTCCTGTTCACCCACAAGGCTGATTTTTCCGTGTTCCTCCTC

TGGAAAGAGCATTGCTTTCTCTCTTCCAGCACTTTACCTACATTCATGTCTTTCAGGT

Sequence GGCTGCTTCTCTATTATGCTCTGTGCTTCTCCCTGTCAAAGGCTTCAGCCCACACCGT

GGAGCTAAACAATATGTTTGGCCAGATCCAGTCGCCTGGTTATCCAGACTCCTATCCC

AGTGATTCAGAGGTGACTTGGAATATCACTGTCCCAGATGGGTTTCGGATCAAGCTTT

ACTTCATGCACTTCAACTTGGAATCCTCCTACCTTTGTGAATATGACTATGTGAAGGT

CTCTCTGCGTCCTGGATCCTCACAGCAGCTCATGTGCTGCGCTCCCAGCGTAGAGACA
CCACGGTGATACCAGTCTCCAAGGAGCATGTCACCGTCTACCTGGGCTTGCATGATGT
GCGAGACAAATCGGGGGCAGTCAACAGCTCAGCTGCCCGAGTGGTGCTCCACCCAGAC
TTCAACATCCAAAACTACAACCACGATATAGCTCTGGTGCAGCTGCAGGAGCCTGTGC
CCCTGGGACCCCACGTTATGCCTGTCTGCCTGCCAAGGCTTGAGCCTGAAGGCCCGGC
CCCCCACATGCTGGGCCTGGTGGCCGGCTGGGGCATCTCCAATCCCAATGTGACAGTG
GATGAGATCATCAGCAGTGGCACACGGACCTTGTCAGATGTCCTGCAGTATGTCAAGT
TACCCGTGGTGCCTCACGCTGAGTGCAAAACTAGCTATGAGTCCCGCTCGGGCAATTA
CAGCGTCACGGAGAACATGTTCTGTGCTGGCTACTACGAGGGCGGCAAAGACACGTGC
CTTGGAGATAGCGGTGGGGCCTTTGTCATCTTTGATGACTTGAGCCAGCGCTGGGTGG
TGCAAGGCCTGGTGTCCTGGGGGGGACCTGAAGAATGCGGCAGCAAGCAGGTCTATGG
AGTCTACACAAAGGTCTCCAATTACGTGGACTGGGTGTGGGAGCAGATGGGCTTACCA
CAAAGTGTTGTGGAGCCCCAGGTGGAACGGTGAGCTGACTTACTTCCTCGCGGG
012F Start: ATG at 220 ORF Stop: TGA at 1249 SEQ ID NO: 86 343 as MW at 38275.9kD
NOVl3a, MSFRWLLLWALCFSLSKASAHTVELNNMFGQIQSPGYPDSYPSDSEVTWNIWPDGF
CG105982-O1RIKI'YFMHFNLESSYLCEYDYVKVETEDTSRVPNDKWFGSGALLSASWILTAAHVLRS
PrOteln QRRDTTVIPVSKEHVTVYLGLHDVRDKSGAVNSSAARWLHPDFNIQNYNHDIALVQL
QEPVPLGPHVMPVCLPRLEPEGPAPHMLGLVAGWGISNPNVTVDEIISSGTRTLSDVL
Sequence QWKLPWPHAECKTSYESRSGNYSWENMFCAGYYEGGKDTCLGDSGGAFVIFDDLS
~QRWWQGLVSWGGPEECGSKQVYGVYTKVSNYVDWVWEQMGLPQSWEPQVER
Further analysis of the NOV 13a protein yielded the following properties shown in Table 13B.
Table 13B. Protein Sequence Properties NOVl3a PSort 0.3700 probability located in outside; 0.1900 probability located in lysosome analysis: (lumen); 0.1000 probability located in endoplasmic reticulum (membrane);
0.1000 probability located in endoplasmic reticulum (lumen) SignalP Cleavage site between residues 22 and 23 analysis:

A search of the NOV 13a protein against the Geneseq database, a proprietary database that contains sequences published in patents and patent publication, yielded several homologous proteins shown in Table 13C.
Table 13C. Geneseq Results for NOVl3a NOVl3a Identities/

Geneseq Protein/Organism/Length Residues/SimilaritiesExpect [Patent for Identifier#, Date) Match the Matched Value ResiduesRegion AAB85060 Human serine protease 82..343 259/262 (98%)e-154 polypeptide - Homo Sapiens,467..728260/262 (98%) aa. [W0200140451-A2, AAB47559 Protease PRTS-1 - Homo 82..343 258/262 (98%)e-153 Sapiens, 728 aa. [W0200171004-A2,467..728259/262 (98%) SEP-2001 ]

AAB84203 Amino acid sequence of 82..332 248/251 (98%)e-148 a human serine protease designated19..269 249/251 (98%) Zfaixl -Homo Sapiens, 269 aa.

[W0200138501-A2, 31-MAY-2001]

AAG00221 Human secreted protein, 4..82 79/79 (100%)3e-42 SEQ ID

NO: 4302 - Homo Sapiens,2..80 79179 (100%) 97 aa.

[EP1033401-A2, 06-SEP-2000]

AAB60935 Horseshoe crab recombinant92..326 90/244 (36%)2e-37 Factor C #2 - Carcinoscorpius 787..1015127/244 (51 %) rotundicauda, 1019 aa.

[WO200127289-A2, 19-APR-2001]

In a BLAST search of public sequence datbases, the NOV 13a protein was found to have homology to the proteins shown in the BLASTP data in Table 13D.

THIS PAGE IS LEFT BLANK.

Table 13D. Public BLASTP Results for NOVl3a NOVl3a Identities/
Protein Residues/SimilaritiesExpect AccessionProtein/Organism/Length for Match the MatchedValue Number ResiduesPortion :

CAC42682SEQUENCE 1 FROM PATENT 82..343 259/262 e-154 (98%) W00140451 - Homo Sapiens 467..728260/262 (98%) (Human), 728 aa.

Q96RS4 COMPLEMENT FACTOR MASP- 82..343 259/262 e-154 (98%) 3 - Homo Sapiens (Human),~ 260/262 728 aa. 467..728(98%) CAC42545~ SEQUENCE 1 FROM PATENT 82..332 248/251 e-147 (98%) W00138501 - Homo Sapiens 19..269 249/251 (98%) (Human), 269 as (fragment).

Q920S0 MBL-ASSOCIATED SERINE 82..343 236/262 e-141 (90%) PROTEASE-3 - Mus musculus472..7332471262 f (94%) (Mouse), 733 aa.

Q9PVY2 MANNOSE-BINDING LECTIN- 82..330 158/251 9e-93 (62%) ASSOCIATED SERINE 465..714198/251 (77%) PROTEASE - Triakis scyllium (Leopard shark) (Triakis scyllia), ~ 719 aa.

PFam analysis predicts that the NOVl3a protein contains the domains shown in the Table 13E.
Table 13E. Domain Analysis of NOVl3a Identities/
Pfam Domain NOVl3a Match Region Similarities Expect Value for the Matched Region CUB 22..134 37/127 (29%) l.Se-OS
75/127 (59%) Trypsin 94..326 86/258 (33%) 2.2e-66 192/258 (74%) EXAMPLE 14.
The NOV 14 clone was analyzed, and the nucleotide and encoded polypeptide sequences are shown in Table 14A.
Table 14A. NOV14 Sequence Analysis SEQ ID NO: 87 861 by NOVl4a, CAGCTCAGCATGGCTAGGGTACTGGGAGCACCCGTTGCACTGGGGTTGTGGAGCCTAT

DNA TGAAGGCGAGACCAAGCCAGACCCAGACGTGACTGAACGCTGCTCAGATGGCTGGAGC

SeClllenCeTTTGATGCTACCACCCTGGATGACAATGGAACCATGCTGTTTTTTAAAGGGACCCACT

ACTGGCGTCTGGACACCAGCCGGGATGGCTGGCATAGCTGGCCCATTGCTCATCAGTG

GCCCCAGGGTCCTTCAGCAGTGGATGCTGCCTTTTCCTGGGAAGAAAAACTCTATCTG

GTCCAGGGCACCCAGGTATATGTCTTCCTGACAAAGGGAGGCTATACCCTAGTAAGCG

GTTATCCGAAGCGGCTGGAGAAGGAAGTCGGGACCCCTCATGGGATTATCCTGGACTC

TGTGGATGCGGCCTTTATCTGCCCTGGGTCTTCTCGGCTCCATATCATGGCAGGACGG

CGGCTGTGGTGGCTGGACCTGAAGTCAGGAGCCCAAGCCACGTGGACAGAGCTTCCTT

GGCCCCATGAGAAGGTAGACGGAGCCTTGTGTATGGAAAAGTCCCTTGGCCCTAACTC

ATGTTCCGCCAATGGTCCCGGCTTGTACCTCATCCATGGTCCCAATTTGTACTGCTAC

AGTGATGTGGAGAAACTGAATGCAGCCAAGGCCCTTCCGCAACCCCAGAATGTGACCA

GTCTCCTGGGCTGCACTCACTGAGGGGCCTTCTGACATGAGTCTGGCCTGGCCCCACC

TCCTAGTTCCTCATAATAAAGACAGATTGCTTCTTCGCTTCTCACTGAG

ORF Start: ATG at 10 ORF Stop:
TGA at 775 SEQ ID NO: 88 255 as MW at 27921.4kD

NOVl4a, MARVLGAPVALGLWSLCWSLAIATPLPPTSAIiGNVAEGETKPDPDVTERCSDGWSFDA

PrOteln TQ~FLTKGGYTLVSGYPKRLEKEVGTPHGIILDSVDAAFICPGSSRLHIMAGRRLW

WLDLKSGAQATWTELPWPHEKVDGALCMEKSLGPNSCSANGPGLYLIHGPNLYCYSDV

Sequence EKLNAAKALPQPQNVTSLLGCTH

Further analysis of the NOV 14a protein yielded the following properties shown in Table 14B.
Table 14B. Protein Sequence Properties NOVl4a PSort 0.4586 probability located in lysosome (lumen); 0.4323 probability located in analysis: outside; 0.3077 probability located in microbody (peroxisome);
0.1000 probability located in endoplasmic reticulum (membrane) SignalP Cleavage site between residues 32 and 33 analysis:
A search of the NOV 14a protein against the Geneseq database, a proprietary database that contains sequences published in patents and patent publication, yielded several homologous proteins shown in Table 14C.

Table 14C. Geneseq Results for NOVl4a NOVl4a Identities/
~

Geneseq Protein/Organism/LengthResidues!SimilaritiesExpect for Identifier[Patent #, Date] Match the MatchedValue Residues Region AAM23933 Human EST encoded protein. 30..255197/226 e-116 SEQ (87%) ID NO: 1458 - Homo sapiens,242..462 201!226 462 (88%) aa. [W0200154477-A2, 2001 ]

AAG00304 Human secreted protein,1..77 73/77 (94%)Se-39 SEQ ID

NO: 4385 - Homo sapiens,1..77 74/77 (95%) 83 aa.

[EP1033401-A2, 06-SEP-2000]

AAP93630 Sequence of rat transin43..179 45/142 (31 2e-08 - Rattus %) rattus, 463 aa. [GB2209526-A,270..401 68/142 (47%) MAY-1989]

AAM48977 Human matrix metalloproteinase30..177 38/150 (25%)4e-07 13 (collagenase 3) - 264..406 68!150 (45%) Homo Sapiens, 471 aa. [W0200206294-A2, JAN-2002]

AAB84615 Amino acid sequence 30..177 38/150 (25%)4e-07 ofmatrix metalloproteinase-13 264..406 68/150 (45%) - Homo Sapiens, 471 aa. [W0200149309-A2, 12-JUL-2001]

In a BLAST search of public sequence datbases, the NOV 14a protein was found to have homology to the proteins shown in the BLASTP data in Table 14D.

Table 14D. Public BLASTP Results for NOVl4a Protein NOVl4a Identities/

AccessionProteinlOrganism/LengthResidues/SimilaritiesExpect for Number Match the Matched Value Residues Portion P02790 Hemopexin precursor 30..255 197/226 (87%)e-116 (Beta-1B-glycoprotein) - Homo 242..462 201/226 (88%) Sapiens (Human), 462 aa. , OQRB hemopexin precursor 28..255 167/228 (73%)e-100 - rabbit, 459 aa. 233..459 186/228 (81%) P200S8 Hemopexin precursor 28..255 167/228 (73%)e-100 -Oryctolagus cuniculus 234..460 186/228 (81%) (Rabbit), 460 aa.

P20059 Hemopexin precursor 30..254 1 591225 ?e-96 - Rattus (70%) norvegicus (Rat), 460 242..459 183/225 (80%) aa.

PS0828 Hemopexin precursor 48..253 152/206 (73%)2e-9S

(Hyaluronidase) (EC 248..453 175/206 (84%) 3.2.1.35) -Sus scrofa (Pig), 459 aa.

PFam analysis predicts that the NOV 14a protein contains the domains shown in the Table 14E.
Table 14E. Domain Analysis of NOVl4a Identities/
Pfam Domain NOVl4a Match Region Similarities Expect Value for the Matched Region hemopexin 56..99 ' 17/50 (34%) 1.4e-09 31/50 (62%) , hemopexin 101..146 14/50 (28%) 4.5e-07 37/50 (74%) S EXAMPLE 15.
The NOV 1 S clone was analysed, and the nucleotide and encoded polypeptide sequences are shown in Table 1 SA.
Table 15A. NOV15 Sequence Analysis SEQ ID NO: 89 2671 by NOVISa, CCCGCCGGGCGAGCATGGGGCGCCTGGCCTCGAGGCCGCTGCTGCTGGCGCTCCTGTC

DNA GTGGGCACTGAGCTGGTCATCCCCTGCAACGTCAGTGACTATGATGGCCCCAGCGAGC

AAAACTTTGACTGGAGCTTCTCATCTTTGGGGAGCAGCTTTGTGGAGCTTGCAAGCAC

Sequence CTGGGAGGTGGGGTTCCCAGCCCAACTGTACCAGGAGCGGCTGCAGAGGGGCGAGATC

!CTGTTAAGGCGGACTGCCAACGACGCCGTGGAGCTCCACATAAAGAACGTCCAGCCTT
CAGACCAAGGCCACTACAAATGTTCAACCCCCAGCACAGATGCCACTGTCCAGGGAAA
CTATGAGGACACAGTGCAGGTTAAAGTGCTGGCCGACTCCCTGCACGTGGGCCCCAGC
'GCGCGGCCCCCGCCGAGCCTGAGCCTGCGGGAGGGGGAGCCCTTCGAGCTGCGCTGCA
',CCGCCGCCTCCGCCTCGCCGCTGCACACGCACCTGGCGCTGCTGTGGGAGGTGCACCG
CGGCCCGGCCAGGCGGAGCGTCCTCGCCCTGACCCACGAGGGCAGGTTCCACCCGGGC
',CTGGGGTACGAGCAGCGCTACCACAGTGGGGACGTGCGCCTCGACACCGTGGGCAGCG
',ACGCCTACCGCCTCTCAGTGTCCCGGGCTCTGTCTGCCGACCAGGGCTCCTACAGGTG
'TATCGTCAGCGAGTGGATCGCCGAGCAGGGCAACTGGCAGGAAATCCAAGAAA.AGGCC
GTGGAAGTTGCCACCGTGGTGATCCAGCCGACAGTTCTGCGAGCAGCCGTGCCCAAGA
ATGTGTCTGTGGCTGAAGGAAAGGAACTGGACCTGACCTGTAACATCACAACAGACCG
AGCCGATGACGTCCGGCCCGAGGTGACGTGGTCCTTCAGCAGGATGCCTGACAGCACC
CTACCTGGCTCCCGCGTGTTGGCGCGGCTTGACCGTGATTCCCTGGTGCACAGCTCGC
CTCATGTTGCTTTGAGTCATGTGGATGCACGCTCCTACCATTTACTGGTTCGGGATGT
TAGCAAAGAAAACTCTGGCTACTATTACTGCCACGTGTCCCTGTGGGCACCCGGACAC
AACAGGAGCTGGCACAAAGTGGCAGAGGCCGTGTCTTCCCCAGCTGGTGTGGGTGTGA
CCTGGCTAGAACCAGACTACCAGGTGTACCTGAATGCTTCCAAGGTCCCCGGGTTTGC
GGATGACCCCACAGAGCTGGCATGCCGGGTGGTGGACACGAAGAGTGGGGAGGCGAAT
GTCCGATTCACGGTTTCGTGGTACTACAGGATGAACCGGCGCAGCGACAATGTGGTGA
CCAGCGAGCTGCTTGCAGTCATGGACGGGGACTGGACGCTAAAATATGGAGAGAGGAG
CAAGCAGCGGGCCCAGGATGGAGACTTTATTTTTTCTAAGGAACATACAGACACGTTC
AATTTCCGGATCCAAAGGACTACAGAGGAAGACAGAGGCAATTATTACTGTGTTGTGT
CTGCCTGGACCAAACAGCGGAACAACAGCTGGGTGAAAAGCAAGGATGTCTTCTCCAA
GCCTGTTAACATATTTTGGGCATTAGAAGATTCCGTGCTTGTGGTGAAGGCGAGGCAG
CCAAAGCCTTTCTTTGCTGCCGGAAATACATTTGAGATGACTTGCAAAGTATCTTCCA
AGAATATTAAGTCGCCACGCTACTCTGTTCTCATCATGGCTGAGAAGCCTGTCGGCGA
CCTCTCCAGTCCCAATGAAACGAAGTACATCATCTCTCTGGACCAGGATTCTGTGGTG
AAGCTGGAGAATTGGACAGATGCATCACGGGTGGATGGCGTTGTTTTAGAAAAAGTGC
AGGAGGATGAGTTCCGCTATCGAATGTACCAGACTCAGGTCTCAGACGCAGGGCTGTA
CCGCTGCATGGTGACAGCCTGGTCTCCTGTCAGGGGCAGCCTTTGGCGAGAAGCAGCA
ACCAGTCTCTCCAATCCTATTGAGATAGACTTCCAAACCTCAGGTCCTATATTTAATG
CTTCTGTGCATTCAGACACACCATCAGTAATTCGGGGAGATCTGATCAAATTGTTCTG
TATCATCACTGTCGAGGGAGCAGCACTGGATCCAGATGACATGGCCTTTGATGTGTCC
TGGTTTGCGGTGCACTCTTTTGGCCTGGACAAGGCTCCTGTGCTCCTGTCTTCCCTGG
ATCGGAAGGGCATCGTGACCACCTCCCGGAGGGACTGGAAGAGCGACCTCAGCCTGGA
GCGCGTGAGTGTGCTGGAATTCTTGCTGCAAGTGCATGGCTCCGAGGACCAGGACTTT
GGCAACTACTACTGTTCCGTGACTCCATGGGTGAAGTCACCAACAGGTTCCTGGCAGA
AGGAGGCAGAGATCCACTCCAAGCCCGTTTTTATAACTGTGAAGATGGATGTGCTGAA
CGCCTTCAAGTATCCCTTGCTGATCGGCGTCGGTCTGTCCACGGTCATCGGGCTCCTG
TCCTGTCTCATCGGGTACTGCAGCTCCCACTGGTGTTGTAAGAAGGAGGTTCAGGAGA
CACGGCGCGAGCGCCGCAGGCTCATGTCGATGGAGATGGACTAGGCTGGCCCGGGAGG
GGA
ORF Start: ATG at 1 S ~ ORF Stop: TAG at 2652 SFQ~ID NO: 90 879 aa~~MW at ~98569.4kD
NOVISa, MGRLASRPLLLALLSLALCRGRWRVPTATLVRWGTELVIPCNVSDYDGPSEQNFDW

PIOtE'.lll YKCSTPSTDATVQGNYEDTVQVKVLADSLHVGPSARPPPSLSLREGEPFELRCTAASA
SPLHTHLALLWEVHRGPARRSVLALTHEGRFHPGLGYEQRYHSGDVRLDTVGSDAYRL
SeC111e11Ce SVSRALSADQGSYRCIVSEWIAEQGNWQEIQEKAVEVATWIQPTVLRAAVPKNVSVA
EGKELDLTCNITTDRADDVRPEVTWSFSRMPDSTLPGSRVLARLDRDSLVHSSPHVAL
SHVDARSYHLLVRDVSKENSGYYYCHVSLWAPGHNRSWHKVAEAVSSPAGVGVTWLEP
DYQWLNASKVPGFADDPTELACRVVDTKSGEANVRFTVSWYYRMNRRSDNVVTSELL
AVMDGDWTLKYGERSKQRAQDGDFIFSKEHTDTFNFRIQRTTEEDRGNYYCWSAWTK
QRNNSWVKSKDVFSKPVNIFWALEDSVLWKARQPKPFFAAGNTFEMTCKVSSKNIKS
PRYSVLIMAEKPVGDLSSPNETKYIISLDQDSWKLENWTDASRVDGVVLEKVQEDEF
RYRMYQTQVSDAGLYRCMVTAWSPVRGSLWREAATSLSNPIEIDFQTSGPIFNASVHS
DTPSVIRGDLIKLFCIITVEGAALDPDDMAFDVSWFAVHSFGLDKAPVLLSSLDRKGI
VTTSRRDWKSDLSLERVSVLEFLLQVHGS'EDQDFGNYYCSVTPWVKSPTGSWQKEAEI
HSKPVFITVKMDVLNAFKYPLLIGVGLSTVIGLLSCLIGYCSSHWCCKKEVQETRRER

RRLMSMEMD
Further analysis of the NOV 15a protein yielded the following properties shown in Table 15B.
Table 15B. Protein Sequence Properties NOVlSa PSort 0.6800 probability located in lysosome (membrane); 0.5140 probability analysis: located in plasma membrane; 0.1760 probability located in microbody (peroxisome); 0.1000 probability located in endoplasmic reticulum (membrane) SignalP Cleavage site between residues 26 and 27 analysis:
A search of the NOV 1 Sa protein against the Geneseq database, a proprietary database that contains sequences published in patents and patent publication, yielded several homologous proteins shown in Table 15C.
Table 15C. Geneseq Results for NOVlSa NOVlSa Identities) Geneseq Protein/Organism/Length Residues!SimilaritiesExpect [Patent for Identifier#, Date] Match the MatchedValue ResiduesRegion AAM93277Human polypeptide, SEQ 1..863 8621863 0.0 ID NO: (99%) 2751 - Homo Sapiens, 863 1..863 8621863 aa. (99%) [EP1130094-A2, OS-SEP-2001]

ABBl Human PG F2a receptor 236..372131/137 7e-70 1196 regulator (95%) homologue, SEQ ID N0:15662..138 1321137 - (95%) Homo sapiens, 138 aa.

[W0200157188-A2, 09-AUG-ABB10996Human prostaglandin receptor500..625117/126 3e-60 (92%) regulator homologue, SEQ 1..126 118/126 ID (92%) NO:1366 - Homo sapiens, 126 aa.

[W0200157188-A2, 09-AUG-2001]

AAB90544Human secreted protein; 6..542 163/565 2e-59 SEQ ID (28%) NO: 82 - Homo Sapiens, 12..571 2601565 613 aa. (45%) [W0200121658-Al, 29-MAR-2001]

AAM24248H~ EST encoded protein 2e-59 ID NO: 1773 - Homo Sapiens, 613 12..571 260/565 (45%) aa. [W0200154477-A2, 02-AUG-2001 ~
In a BLAST search of public sequence datbases, the NOV 15a protein was found to have homology to the proteins shown in the BLASTP
data in Table 15D.

Table 15D. Public BLASTP
Results for NOVlSa Protein NOVlSa Identities/

AccessionProteinlOrganismlLength Residues/SimilaritiesExpect for Number Match the Matched Value ResiduesPortion Q9P2B2 KIAA1436 PROTEIN - Homo 1..879 878/879 (99%)0.0 Sapiens (Human), 924 46..924 879/879 (99%) as (fragment).

Q9WV91 F2 ALPHA PROSTOGLANDIN 1..879 786/879 (89%)0.0 REGULATORY PROTEIN - I ..879 830/879 (94%) Mus musculus (Mouse), 879 aa.

Q62786 Prostaglandin F2-alpha 1..879 784/879 (89%)0.0 receptor regulatory protein precursor1..879 834/879 (94%) (Prostaglandin F2-alpha receptor associated protein) -Rattus norvegicus (Rat), 879 aa.

Q9H3U3 SMAP-6 - Homo sapiens 694..879186/186 (100%)e-106 (Human), 186 as (fragment). 1..186 186/186 (100%) 002834 ADIPOCYTE MEMBRANE 690..879184/190 (96%)e-105 PROTEIN - Sus scrofa 1..190 186/190 (97%) (Pig), 190 as (fragment).

PFam analysis predicts that the NOV
1 Sa protein contains the domains shown in the Table 15E.
Table 15E. Domain Analysis of NOVlSa Identities/

Pfam DomainNOVlSa Match RegionSimilarities Expect Value for the Matched Region ig 36..121 15/87 (17%) 0.0013 52/87 (60%) ig 162..249 13/89 (15%) 0.00048 60/89 (67%) ig 292..375 16/85 (19%) 5.8e-07 58/85 (68%) ig 422..517 16/97 (16Z) 2.3e-06 72/97 (74%) ig 564..657 12/97 (12%) 1.2e-05 64/97 (66%) ig 704..795 11/93 (12%) ~ 0.19 55/93 (59%) EXAMPLE 16.
The NOV 16 clone was analyzed, and the nucleotide and encoded polypeptide sequences are shown in Table 16A.
Table 16A. NOV16 Sequence Analysis SEQ ID NO: 91 E 1565 by NOVl6a, ~GGAATGCTCTCCCGCCTGAGCCTGCTCCAGGAATTGGACCTCAGCTACAACCAGCTCT
CG109496-O1C~CCCTTGAGCCTGGGGCCTTCCATGGCCTACAAAGCCTACTCACCCTGAGGCTGCA

DNA GGGCAATCGGCTCAGAATCATGGGGCCTGGGGTCTTCTCAGGCCTCTCTGCTCTGACC

CTGCTGGACCTCCGCCTCAACCAGATTGTTCTCTTCCTAGATGGAGCTTTTGGGGAGC

SeqllenCeTAGGCAGCCTCCAGAAGCTGGAGGTTGGGGACAACCACCTGGTATTTGTGGCTCCGGG

GGCCTTTGCAGGGCTAGCCAAGTTGAGCACCCTCACCCTGGAGCGCTGCAACCTCAGC

ACAGTGCCTGGCCTAGCCCTTGCCCGTCTCCCGGCACTAGTGGCCCTAAGGCTTAGAG

AACTGGATATTGGGAGGCTGCCAGCTGGGGCCCTGCGGGGGCTGGGGCAGCTCAAGGA

GCTGGAGATCCACCTCTGGCCATCTCTGGAGGCTCTGGACCCTGGGAGCCTGGTTGGG

CTCAATCTCAGCAGCCTGGCCATCACTCGCTGCAATCTGAGCTCGGTGCCCTTCCAAG

CACTGTACCACCTCAGCTTCCTCAGGGTCCTGGATCTGTCCCAGAATCCCATCTCAGC

CATCCCAGCCCGAAGGCTCAGCCCCCTGGTGCGGCTCCAGGAGCTACGCCTGTCAGGG

GCATGCCTCACCTCCATTGCTGCCCATGCCTTCCATGGCTTGACTGCCTTCCACCTCC

TGGATGTGGCAGATAACGCCCTTCAGACACTAGAGGAAACAGCTTTCCCTTCTCCAGA

CAAACTGGTCACCTTGAGGCTGTCTGGCAACCCCCTAACCTGTGACTGCCGCCTCCTC

TGGCTGCTCCGGCTCCGCCGCCACCTGGACTTTGGCATGTCCCCCCCTGCCTGTGCTG

GCCCCCATCATGTCCAGGGGAAGAGCCTGAAGGAGTTTTCAGACATCCTGCCTCCAGG

GCACTTCACCTGCAAACCAGCCCTGATCCGAAAGTCGGGGCCTCGATGGGTCATTGCA

GAGGAGGGCGGGCATGCGGTTTTCTCCTGCTCTGGAGATGGAGACCCAGCCCCCACTG

TCTCCTGGATGAGGCCTCATGGGGCTTGGCTGGGCAGGGCTGGGAGAGTAAGGGTCCT

AGAGGATGGGACACTGGAGATCCGCTCAGTGCAGCTACGGGACAGAGGGGCCTATGTC

TGTGTGGTTAGCAATGTCGCTGGGAATGACTCCCTGAGGACCTGGCTGGAAGTCATCC

AGGTGGAACCACCAAACGGCACACTTTCTGACCCCAACATCACCGTGCCAGGGATCCC

AGGGCCTTTTTTTCTGGATAGCAGAGGTGTGGCCATGGTGCTGGCAGTCGGCTTCCTC

CCCTTCCTCACCTCAGTGACCCTCTGCTTTGGCCTGATTGCCCTTTGGAGCAAGGGCA

AAGGTCGGGTCAAACATCACATGACCTTTGACTTTGTGGCACCTCGGCCCTCTGGGGA

TAAAAACTCTGGGGGTAACCGGGTCACTGCCAAGCTCTTCTGACCTTTCCTTCCCCA

ORF Start: ATG at 4 ORF Stop:
TGA at 1549 .

SEQ ID NO: 92 51 S as MW at 55659.OkD

NOVl6a, MLSRLSLLQELDLSYNQLSTLEPGAFHGLQSLLTLRLQGNRLRIMGPGVFSGLSALTL

P1'Oteln VPGLALARLPALVALRLRELDIGRLPAGALRGLGQLKELEIHLWPSLEALDPGSLVGL
NLSSLAITRCNLSSVPFQALYHLSFLRVLDLSQNPISAIPARRLSPLVRLQELRLSGA
Sequence CLTSIAAHAFHGLTAFHLLDVADNALQTLEETAFPSPDKLVTLRLSGNPLTCDCRLLW
LLRLRRHLDFGMSPPACAGPHHVQGKSLKEFSDILPPGHFTCKPALIRKSGPRWVIAE
EGGHAVFSCSGDGDPAPTVSWMRPHGAWLGRAGRVRVLEDGTLEIRSVQLRDRGAYVC
VVSNVAGNDSLRTWLEVIQVEPPNGTLSDPNITVPGIPGPFFLDSRGVAMVLAVGFLP
FLTSVTLCFGLIALWSKGKGRVKHHMTFDFVAPRPSGDKNSGGNRVTAKLF
Further analysis of the NOV 16a protein yielded the following properties shown in Table 16B.

Table 16B. Protein Sequence Properties NOVl6a PSort 0.7000 probability located in plasma membrane; 0.5204 probability located in analysis: mitochondria) inner membrane; 0.4430 probability located in microbody (peroxisome); 0.2217 probability located in mitochondria) intermembrane space SignaIP No Known Signal Sequence Predicted analysis:
A search of the NOV 16a protein against the Geneseq database, a proprietary database that contains sequences published in patents and patent publication, yielded several homologous proteins shown in Table 16C.
Table 16C. Geneseq Results for NOVl6a NOVl6a Identities/

Geneseq ProteinlOrganism/Length Residues/SimilaritiesExpect for Identifier[Patent #, Date] Match the Matched Value ~

ResiduesRegion AAW84596 Amino acid sequence of 8..500 229/509 (44%)e-116 the human .

Tango-79 protein - Homo 91..596 3121509 (60%) Sapiens, 3 614 aa. [W09906427-A1, 1999]

AAB74705 Human membrane associated8..500 228/509 (44%)e-116 protein MEMAP-11 - Homo 97..602 312/509 (60%) Sapiens, 620 aa. [WO200112662-A2, 22-FEB-2001]

AAB80225 Human PR0227 protein 8..500 227/509 (44%)e-115 - Homo ;

Sapiens, 620 aa. [W0200104311-97..602 311/509 (60%) A1, 18-JAN-2001]

AAU12333 Human PR0227 polypeptide8..500 227/509 (44%)e-115 sequence - Homo Sapiens,97..602 311/509 (60%) 620 aa.

[W0200140466-A2, 07-JUN-2001 ]

AAY13357 Amino acid sequence ofprotein8..500 227/509 (44%)e=115 ~

PR0227 - Homo Sapiens, 97..602 311/509 (60%) 620 aa.

[W09914328-A2, 25-MAR-1999]

In a BLAST search of public sequence datbases, the NOV 16a protein was found to have homology to the proteins shown in the BLASTP data in Table 16D.

Table 16D. Public SLASTP Results for NOVl6a NOVl6a Identities/
Protein Residues/SimilaritiesExpect AccessionProtein/Organism/Length for Match the MatchedValue Number ResiduesPortion Q9N008 HYPOTHETICAL 69.2 KDA 8..500 228/509 e-115 (44%) PROTEIN - Macaca fascicularis91..596 312/509 (60%) (Crab eating macaque) (Cynomolgus monkey), 614 aa.

Q96FE5 UNKNOWN (PROTEIN FOR 8..500 228/509 e-115 (44%) MGC:17422) - Homo sapiens91..596 312/509 (60%) (Human), 614 aa.

Q9D1T0 ADULT MALE TESTIS CDNA, 8..500 228/509 e-115 (44%) RIKEN FULL-LENGTH 91..596 312/509 (60%) ENRICHED LIBRARY, CLONE:4930471K13, FULL

INSERT SEQUENCE - Mus musculus (Mouse), 614 aa.

Q9BZ20 BA438B23.1 (NEURONAL 7..501 224/507 e-113 (44%) LEUCINE-RICH REPEAT 82..588 311/507 (61%) PROTEIN) (CDNA FLJ31810 FIS, CLONE NT2RI2009289, WEAKLY

SIMILAR TO

KDA

CHAIN) - Homo Sapiens (Human), 606 aa.

CAC34918SEQUENCE 1 FROM PATENT 7..501 197/505 3e-89 (39%) W00075358 - Homo Sapiens 82..530 278/505 (55%) (Human), 548 aa.

PFam analysis predicts that the NOV 16a protein contains the domains shown in the Table 16E.
Table 16E. Domain Analysis of NOVl6a Identities/

Pfam DomainNOVl6a Match RegionSimilarities Expect Value for the Matched Region LRR 7..30 13/25 (52%) 3.8e-05 22/25 (88%) LRR 31..54 8/25 (32%) 0.64 19/25 (76%) LRR 199..222 ' 9125 (36%) 0.27 16/25 (64%) LltR 223..246 ~ 9/25 (36%) 0.32 19/25 (76%) LRRCT 280..333 ~ 19/58 (33%) 3.1e-07 42158 (72%) 1g 350..408 19/62 (31 %) 9. l e-09 41/62 (66%) EXAMPLE 17.
The NOV 17 clone was analyzed, and the nucleotide and encoded polypeptide sequences are shown in Table 17A.
Table 17A. NOV17 Sequence Analysis SEQ ID NO: 93 ~ 1780 by NOVl7a, ~CCAACCCTCTGCCCGGCCGGTGCCCATGCTTCTGTGGCTGCTGCTGCTGATCCTGACT

DNA GGTCCACAGCCTTCAAAGGAGAAAAAGTGGCTCTCATATGCAGCAGCATATCACATTC

CCTAGCCCAGGGAGACACATATTGGTATCACGATGAGAAGTTGTTGAAAATAAAACAT

SeqllenCeGACAAGATCCAAATTACAGAGCCTGGAAATTACCAATGTAAGACCCGAGGATCCTCCC

TCAGTGATGCCGTGCATGTGGAATTTTCACCTGACTGGCTGATCCTGCAGGCTTTACA

TCCTGTCTTTGAAGGAGACAATGTCATTCTGAGATGTCAGGGGAAAGACAACAAAAAC.

ACTCATCAAAAGGTTTACTACAAGGATGGAAAACAGCTTCCTAATAGTTATAATTTAG

AGAAGATCACAGTGAATTCAGTCTCCAGGGATAATAGCAAATATCATTGTACTGCTTA

TAGGAAGTTTTACATACTTGACATTGAAGTAACTTCAAAACCCCTAAATATCCAAGTT

CAAGAGCTGTTTCTACATCCTGTGCTGAGAGCCAGCTCTTCCACGCCCATAGAGGGGA

GTCCCATGACCCTGACCTGTGAGACCCAGCTCTCTCCACAGAGGCCAGATGTCCAGCT

GCAATTCTCCCTCTTCAGAGATAGCCAGACCCTCGGATTGGGCTGGAGCAGGTCCCCC

ACGTGTACAGAG
GGAGAAAATATGGTCCTTATTTGCTCAGTAGCCCAGGGTTCAGGGACTGTCACATTCT
CCTGGCACAAAGAAGGAAGAGTAAGAAGCCTGGGTAGAAAGACCCAGCGTTCCCTGTT
GGCAGAGCTGCATGTTCTCACCGTGAAGGAGAGTGATGCAGGGAGATACTACTGTGCA
GCTGATAACGTTCACAGCCCCATCCTCAGCACGTGGATTCGAGTCACCGTGAGAATTC

CGGTATCTCACCCTGTCCTCACCTTCAGGGCTCCCAGGGCCCACACTGTGGTGGGGGA

CCTGCTGGAGCTTCACTGTGAGTCCCTGAGAGGCTCTCCCCCGATCCTGTACCGATTT

TATCATGAGGATGTCACCCTGGGGAACAGCTCAGCCCCCTCTGGAGGAGGAGCCTCCT

TCAACCTCTCTCTGACTGCAGAACATTCTGGAAACTACTCCTGTGATGCAGACAATGG

CCTGGGGGCCCAGCACAGTCATGGAGTGAGTCTCAGGGTCACAGTTCCGGTGTCTCGC

CCCGTCCTCACCCTCAGGGCTCCCGGGGCCCAGGCTGTGGTGGGGGACCTGCTGGAGC

TTCACTGTGAGTCCCTGAGAGGCTCCTTCCCGATCCTGTACTGGTTTTATCACGAGGA

TGACACCTTGGGGAACATCTCGGCCCACTCTGGAGGAGGGGCATCCTTCAACCTCTCT

CTGACTACAGAACATTCTGGAAACTACTCATGTGAGGCTGACAATGGCCTGGGGGCCC

AGCACAGTAAAGTGGTGACACTCAATGTTACAGGTGTGTTAATAGTACCTGGGCTAGA

GGTCACAGTTATGGTAAATAAAATAGTTATCTGACAGATT

ORF Start: ATG at 26 ORF Stop:
TGA at 1772 SEQ ID NO: 94 582 as MW at 64270.SkD

NOVl7a, MLLWLLLLILTPGREQSGVAPKAVLLLNPPWSTAFKGEKVALICSSISHSLAQGDTYW

PrOteIri ILRCQGKDNKNTHQKWYKDGKQLPNSYNLEKITVNSVSRDNSKYHCTAYRKFYILDI

EVTSKPLNIQVQELFLHPVLRASSSTPIEGSPMTLTCETQLSPQRPDVQLQFSLFRDS

Sequence QTLGLGWSRSPRLQIPAMWTEDSGSYWCEVETVTHSIKKRSLRSQIRVQRVPVSNVNL

EIRPTGGQLIEGENMVLICSVAQGSGTWFSWHKEGRVRSLGRKTQRSLLAELHVLTV

KESDAGRYYCAADNVHSPILSTWIRVTVRIPVSHPVLTFRAPRAHTWGDLLELHCES

LRGSPPILYRFYHEDVTLGNSSAPSGGGASFNLSLTAEHSGNYSCDADNGLGAQHSHG

VSLRVTVPVSRPVLTLRAPGAQAWGDLLELHCESLRGSFPILYWFYHEDDTLGNISA

HSGGGASFNLSLTTEHSGNYSCEADNGLGAQHSKVVTLNVTGVLIVPGLEVTVMVNKI

VI

5EQ ID NO: 95 1263 by NOVl7b, AAGCTTGGAGAAAAAGTGGCTCTCATATGCAGCAGCATATCACATTCCCTAGCCCAGG

DNA ~TTACAGAGCCTGGAAATTACCAATGTAAGACCCGAGGATCCTCCCTCAGTGATGCC

GTGCATGTGGAATTTTCACCTGACTGGCTGATCCTGCAGGCTTTACATCCTGTCTTTG

SequeriCeppGGAGACAATGTCATTCTGAGATGTCAGGGGAAAGACAACAAAAACACTCATCAAAA

GGTTTACTACAAGGATGGAAAACAGCTTCCTAATAGTTATAATTTAGAGAAGATCACA

GTGAATTCAGTCTCCAGGGATAATAGCAAATATCATTGTACTGCTTATAGGAAGTTTT

ACATACTTGACATTGAAGTAACTTCAAAACCCCTAAATATCCAAGTTCAAGAGCTGTT

TCTACATCCTGTGCTGAGAGCCAGCTCTTCCACGCCCATAGAGGGGAGTCCCATGACC

CTGACCTGTGAGACCCAGCTCTCTCCACAGAGGCCAGATGTCCAGCTGCAATTCTCCC

TCTTCAGAGATAGCCAGACCCTCGGATTGGGCTGGAGTAGGTCCCCCAGACTCCAGAT

CCCTGCCATGTGGACTGAAGACTCAGGGTCTTACTGGTGTGAGGTGGAGACAGTGACT

CACAGCATCAAAAAAAGGAGCCTGAGATCTCAGATACGTGTACAGAGAGTCCCTGTGT

CTAATGTGAATCTAGAGATCCGGCCCACCGGAGGGCAGCTGATTGAAGGAGAAAATAT

GGTCCTTATTTGCTCAGTAGCCCAGGGTTCAGGGACTGTCACATTCTCCTGGCACAAA

GAAGGAAGAGTAAGAAGCCTGGGTAGAAAGACCCAGCGTTCCCTGTTGGCAGAGCTGC

ATGTTCTCACCGTGAAGGAGAGTGATGCAGGGAGATACTACTGTGCAGCTGATAACGT

TCACAGCCCCATCCTCAGCACGTGGATTCGAGTCACCGTGAGAATTCCGGTATCTCAC

CCTGTCCTCACCTTCAGGGCTCCCAGGGCCCACACTGTGGTGGGGGACCTGCTGGAGC

TTCACTGTGAGTCCCTGAGAGGCTCTCCCCCGATCCTGTACCGATTTTATCATGAGGA

TGTCACCCTGGGGAACAGCTCAGCCCCCTCTGGAGGAGGAGCCTCCTTCAACCTCTCT

CTGACTGCAGAACATTCTGGAAACTACTCATGTGAGGCTCTCGAG

ORF Start: at 1 ORF Stop: end of sequence SEQ ID NO: 96 421 as MW at 47243.1 kD

NOVl7b, KLGEKVALICSSISHSLAQGDTYWYHDEKLLKIKHDKIQITEPGNYQCKTRGSSLSDA

207775340V~EFSPDWLILQALHPVFEGDNVILRCQGKDNKNTHQKVYYKDGKQLPNSYNLEKIT

PTOtelri ~'TSVSRDNSKYHCTAYRKFYILDIEVTSKPLNIQVQELFLHPVLRASSSTPIEGSPMT

LTCETQLSPQRPDVQLQFSLFRDSQTLGLGWSRSPRLQIPAMWTEDSGSYWCEVETVT

SequeriCeHSIKKRSLRSQIRVQRVPVSNVNLEIRPTGGQLIEGENMVLICSVAQGSGTVTFSWHK

EGRVRSLGRKTQRSLLAELHVLTVKESDAGRYYCAADNVHSPILSTWIRVTVRIPVSH

PVLTFRAPRAHTWGDLLELHCESLRGSPPILYRFYHEDVTLGNSSAPSGGGASFNLS

LTAEHSGNYSCEALE
SEQ ID NO: 97 1263 by NOV17C, AAGCTTGGAGAAAAAGTGGCTCTCATATGCAGCAGCATATCACATTCCCTAGCCCAGG

DNA ~TTACAGAGCCTGGAAATTACCAATGTAAGACCCGAGGATCCTCCCTCAGTGATGCC
GTGCATGTGGAATTTTCACCTGACTGGCTGATCCTGCAGGCTTTACATCCTGTCTTTG
SeqtlenCe AAGGAGACAATGTCATTCTGAGATGTCAGGGGAAAGACAACAAAAACACTCATCAAAA
GGTTTACTACAAGGATGGAAAACAGCTTCCTAATAGTTATAATTTAGAGAAGATCACA
GTGAATTCAGTCTCCAGGGATAATAGCAAATATCATTGTACTGCTTATAGGAAGTTTT
ACATACTTGACATTGAAGTAACTTCAAAACCCCTAAATATCCAAGTTCAAGAGCTGTT
TCTACATCCTGTGCTGAGAGCCAGCTCTTCCACGCCCATAGAGGGGAGTCCCATGACC
CTGACCTGTGAGACCCAGCTCTCTCCACAGAGGCCAGATGTCCAGCTGCAATTCTCCC
TCTTCAGAGATAGCCAGACCCTCGGATTGGGCTGGAGCAGGTCCCCCAGACTCCAGAT
CCCTGCCATGTGGACTGAAGACTCAGGGTCTTACTGGTGTGAGGTGGAGACAGTGACT
CACAGCATCAAAAAAAGGAGCCTGAGATCTCAGATACGTGTACAGAGAGTCCCTGTGT
CTAATGTGAATCTAGAGATCCGGCCCACCGGAGGGCAGCTGATTGAAGGAGAAAATAT
GGTCCTTATTTGCTCAGTAGCCCAGGGTTCAGGGACTGTCACATTCTCCTGGCACAAA
GAAGGAAGAGTAAGAAGCCTGGGTAGAAAGACCCAGCGTTCCCTGTTGGCAGAGCTGC
ATGTTCTCACCGTGAAGGAGAGTGATGCAGGGAGATACTACTGTGCAGCTGATAACGT
TCACAGCCCCATCCTCAGCACGTGGATTCGAGTCACCGTGAGAATTCCGGTATCTCAC
CCTGTCCTCACCTTCAGGGCTCCCAGGGCCCACACTGTGGTGGGGGACCTGCTGGAGC
TTCACTGTGAGTCCCTGAGAGGCTCTCCCCCGATCCTGTACCGATTTTATCATGAGGA
TGTCACCCTGGGGAACAGCTCAGCCCCCTCTGGAGGAGGAGCCTCCTTCAACCTCTCT
CTGACTGCAGAACATTCTGGAAACTACTCATGTGAGGCTCTCGAG
ORF Start: at 1 ORF Stop: end of sequence SEQ ID NO: 98 421 as MW at 47243.1kD
NOV17C, KLGEKVALICSSISHSLAQGDTYWYHDEKLLKIKHDKIQITEPGNYQCKTRGSSLSDA
207775361 ~EFSPDWLILQALHPVFEGDNVILRCQGKDNKNTHQKVYYKDGKQLPNSYNLEKIT
Protein ~SVSRDNSKYHCTAYRKFYILDIEVTSKPLNIQVQELFLHPVLRASSSTPIEGSPMT
LTCETQLSPQRPDVQLQFSLFRDSQTLGLGWSRSPRLQTPAMWTEDSGSYWCEVETVT
Sequence HSIKKRSLRSQIRVQRVPVSNVNLEIRPTGGQLIEGENMVLICSVAQGSGTVTFSWHK
EGRVRSLGRKTQRSLLAELHVLTVKESDAGRYYCAADNVHSPILSTWIRVTVRIPVSH
PVLTFRAPRAHTVVGDLLELHCESLRGSPPILYRFYHEDVTLGNSSAPSGGGASFNLS
LTAEHSGNYSCEALE
SEQ ID NO: 99 1263 by NOVl7d, ~GCTTGGAGAAAAAGTGGCTCTCATATGCAGCAGCATATCACATTCCCTAGCCCAGG

DNA ~Z'TACAGAGCCTGGAAATTACCAATGTAAGACCCGAGGATCCTCCCTCAGTGATGCC
GTGCATGTGGAATTTTCACCTGACTGGCTGATCCTGCAGGCTTTACATCCTGTCTTTG
Sequence AAGGAGACAATGTCATTCTGAGATGTCAGGGGAAAGACAACAAAAACACTCATCAAAA
GGTTTACTACAAGGATGGAAAACAGCTTCCTAATAGTTATAATTTAGAGAAGATCACA
GTGAATTCAGTCTCCAGGGATAATAGCAAATATCATTGTACTGCTTATAGGAAGTTTT
ACATACTTGACATTGAAGTAACTTCAAAACCCCTAAATATCCAAGTTCAGGAGCTGTT
TCTACATCCTGTGCTGAGAGCCAGCTCTTCCACGCCCATAGAGGGGAGTCCCATGACC
CTGACCTGTGAGACCCAGCTCTCTCCACAGAGGCCAGATGTCCAGCTGCAATTCTCCC
TCTTCAGAGATAGCCAGACCCCCGGATTGGGCTGGAGCAGGTCCCCCAGACTCCAGAT
CCCTGCCATGTGGACTGAAGACTCAGGGTCTTACTGGTGTGAGGTGGAGACAGTGACT
CACAGCATCAAAAAAAGGAGCCTGAGATCTCAGATACGTGTACAGAGAGTCCCTGTGT
CTAATGTGAATCTAGAGATCCGGCCCACCGGAGGGCAGCTGATTGAAGGAGAAAATAT
GGTCCTTATTTGCTCAGTAGCCCAGGGTTCAGGGACTGTCACATTCTCCTGGCACAAA
GAAGGAAGAGTAAGAAGCCTGGGTAGAAAGACCCAGCGTTCCCTGTTGGCAGAGCTGC
ATGTTCTCACCGTGAAGGAGAGTGATGCAGGGAGATACTACTGTGCAGCTGATAACGT
TCACAGCCCCATCCTCAGCACGTGGATTCGAGTCACCGTGAGAATTCCGGTATCTCAC
CCTGTCCCCACCTTCAGGGCTCCCAGGGCCCACACTGTGGTGGGGGACCTGCTGGAGC
TTCACTGTGAGTCCCTGAGAGGCTCTCCCCCGATCCTGTACCGATTTTATCATGAGGA
TGTCACCCTGGGGAACAGCTCAGCCCCCTCTGGAGGAGGAGACTCCTTCAACCTCTCT
CTGACTGCAGAACATTCTGGAAACTACTCATGTGAGGCTCTCGAG

ORF Start: at 1 OIZF Stop: end o_f sequence SEQ ID NO: 100 421 as MW at 47255.OkD
NOVl7d, ~KLGEKVALICSSISHSLAQGDTYWYHDEKLLKIKHDKIQITEPGNYQCKTRGSSLSDA

PTOteln VNSVSRDNSKYHCTAYRKFYILDIEVTSKPLNIQVQELFLHPVLRASSSTPIEGSPMT

LTCETQLSPQRPDVQLQFSLFRDSQTPGLGWSRSPRLQIPAMWTEDSGSYWCEVETVT

SequenceHSIKKRSLRSQIRVQRVPVSNVNLEIRPTGGQLIEGENMVLICSVAQGSGTVTFSWHK

EGRVRSLGRKTQRSLLAELHVLTVKESDAGRYYCAADNVHSPILSTWIRVTVRIPVSH

PVPTFRAPRAHTVVGDLLELHCESLRGSPPILYRFYHEDVTLGNSSAPSGGGDSFNLS

LTAEHSGNYSCEALE

Sequence comparison of the above protein sequences yields the following sequence relationships shown in Table 17B.
Table 17B. Comparison of NOVl7a against NOVl7b through NOVl7d.
Protein SequenceNOVl7a Residues/Identities/

Match ResiduesSimilarities for the Matched Region NOVl7b 37..453 404/417 (96%) 3..419 . 405/417 (96%) NOV 17c 37..453 404/417 (96%) ~ 3..419 4051417 (96%) NOV 17d 37..453 413/417 (99%) 3..419 414/417 (99%) Further analysis of the NOV 17a protein yielded the following properties shown in Table 17C.
Table 17C. Protein Sequence Properties NOVl7a PSort 0.5374 probability located in outside; 0.1900 probability located in lysosome analysis: (lumen); 0.1000 probability located in endoplasmic reticulum (membrane);
0.1000 probability located in endoplasmic reticulum (lumen) SignalP Cleavage site between residues 18 and 19 analysis:

A search of the NOV 17a protein against the Geneseq database, a proprietary database that contains sequences published in patents and patent publication, yielded several homologous proteins shown in Table 17D.
Table 17D. Geneseq Results for NOVl7a NOVl7a Identities/

Geneseq Protein/Organism/LengthResidues/SimilaritiesExpect for Identifier. [Patent #, Date] Match the Matched Value Residues Region AAB82316Human immunoglobulin 1..564 564/564 (100%)0.0 receptor IRTA3 protein - Homo 1..564 564/564 (100%) sapiens, 734 aa. [W0200138490-A2, MAY-2001 ]

AAB82314Human immunoglobulin 1..564 259/568 (45%)e-130 receptor isoform IRTA2b - Homo 1..561 3341568 (58%) Sapiens, 592 aa. [W0200138490-A2, AAB82315Human immunoglobulin 1..575 261/579 (45%)e-129 receptor isoform IRTA2c - Homo 1..572 338/579 (58%) sapiens, 977 aa. [W0200138490-A2, MAY-2001 ]

AAB82313Human immunoglobulin 1..575 261/579 (45%)e-129 receptor isoform IRTA2a - Homo 1..572 338/579 (58%) Sapiens, 759 aa. [W0200138490-A2, MAY-2001]

AAB82317Human immunoglobulin 100..472 227/374 (60%)e-129 receptor IRTA4 protein - Homo 18..389 280/374 (74%) sapiens, 508 aa. [W0200138490-A2, In a BLAST search of public sequence datbases, the NOV 17a protein was found to have homology to the proteins shown in the BLASTP data in Table 17E.
Table 17E. Public BLASTP Results for NOVl7a Protein NOVl7a Identities/

AccessionProtein/Organism/Length Residues/SimilaritiesExpect for Number Match the Matched Value ResiduesPortion Q96LA4 FC RECEPTOR-LIKE PROTEIN1..564 564/564 (100%)0.0 3 - Homo sapiens (Human),1..564 564/564 (100%) 734 aa.

Q96P31 SH2 DOMAIN-CONTAINING 1..564 564/564 (100%)0.0 PHOSPHATASE ANCHOR 1..564 564/564 (100%) PROTEIN 2A - Homo Sapiens (Human), 734 aa.

Q96P29 SH2 DOMAIN-CONTAINING 1..564 5631570 (98%)0.0 PHOSPHATASE ANCHOR I ..570 564/570 (98%) PROTEIN 2C - Homo sapiens (Human), 740 aa.

CAC05323BA367J7.2.1 (NOVEL 1..548 548/548 (100%)0.0 IMMUNOGLOBUL1N DOMAINS 1..548 548/548 (100%) CONTAINING PROTEIN

(ISOFORM 1)) - Homo Sapiens (Human), 548 as (fragment).

Q96P30 SH2 DOMAIN-CONTAINING 11 I 318/457 (69%)e-167 ..564 PHOSPHATASE ANCHOR 35..469 347/457 (75%) PROTEIN 2B - Homo Sapiens (Human), 639 aa.

PFam analysis predicts that the NOV 17a protein contains the domains shown in the Table 17F.
Table 17F. Domain Analysis of NOVl7a Identities/

Pfam DomainNOVl7a Match RegionSimilarities Expect Value for the Matched Region ig 37..84 12/52 (23%) 0.84 29/52 (56%) ig I 13..165 12157 (21 %) 0.52 38/57 (67%) ig 204..262 ~ 18/61 (30%) 2.3e-08 43161 (70%) ig 302..360 15/61 (25%) 8.2e-10 ~ 46/61 (75%) ig 397..453 ~ 13/59 (22%) 0.0004 ~ 45/59 (76%) ig 490..546 13/60 (22%) 1.7e-05 43/60 (72%) EXAMPLE 18.
The NOV 18 clone was analyzed, and the nucleotide and encoded polypeptide sequences axe shown in Table 18A.
Table 18A. NOV18 Sequence Analysis SEQ ID NO: 101 360 by ~....,~~

NOVl8a, _CGCTGCTCCTGCTGCTGCTGGCGCTGTACACCGCGCGTGTGGACGGGTCCAAATGCAA

DNA CCAAAGTACCCGCACTGCGAGGAGAAGATGGTTATCATCACCACCAAGAGCGTGTCCA

GGTACCGAGGTCAGGAGCACTGCCTGCACCCCAAGCTGCAGAGCACCAAGCGCTTCAT
Sequence CAAGTGGTACAACGCCTGGAACGAGAAGCGCAGGGTCTACGAAGAATAGGGTGAAAAA

CCTCAGAAGGGAAAACTCCAAACCAGTTGGGAGACTTGTGCPrAAGGACTTTGCAGATT

ORF Start: at 3 ORF
Stop:
TAG
at SEQ ID NO: 102 92 MW at 11045.O1cD
as NOVlBa, LLLLLLALYTARVDGSKCKCSRKGPKIRYSDVKKLEMKPKYPHCEEKMVIITTKSVSR

CG50213-O1'~RGQEHCLHPKLQSTKRFIKWYNAWNEKRRVYEE

Protein Sequence SEQ ID NO: 103 228 by NOVlBb, AAATGCAAGTGCTCCCGGAAGGGACCCAAGATCCGCTACAGCGACGTGAAGAAGCTGG

CG50213-02~'TGAAGCCAAAGTACCCGCACTGCGAGGAGAAGATGGTTATCATCACCACCAAGAG

DNA CGTGTCCAGGTACCGAGGTCAGGAGCACTGCCTGCACCCCAAGCTGCAGAGCACCAAG

CGCTTCATCAAGTGGTACAACGCCTGGAACGAGAAGCGCAGGGTCTACGAAGAA

Sequence ORF Start: at 1 ORF
Stop: end of sequence SEQ ID NO: 104 76 as MW at 9331.9kD

NOVlBb, KCKCSRKGPKIRYSDVKKLEMKPKYPHCEEKMVIITTKSVSRYRGQEHCLHPKLQSTK

Protein Sequence SEQ ID NO: 105 228 by NOV18C, AAATGCAAGTGCTCCCGGAAGGGACCCAAGATCCGCTACAGCGACGTGAAGAAGCTGG

CG50213-03~'TGAP'GCCAAAGTACCCGCACTGCGAGGAGAAGATGGTTATCATCACCACCAAGAG

DNA CGTGTCCAGGTACCGAGGTCAGGAGCACTGCCTGCACCCCAAGCTGCAGAGCACCAAG

CGCTTCATCAAGTGGTACAACGCCTGGAACGAGAAGCGCAGGGTCTACGAAGAA
Sequence ORF Start: at 1 ORF
Stop: end of sequence SEQ ID NO: 106 76 as MW at 9331.9kD

NOV18C, KCKCSRKGPKIRYSDVKKLEMKPKYPHCEEKMVIITTKSVSRYRGQEHCLHPKLQSTK

Protein Sequence Sequence comparison of the above protein sequences yields the following sequence relationships shown in Table 18B.
Table 18B. Comparison of NOVl8a against NOVl8b and NOVl8c.
Protein Sequence NOVl8a Residues/ Identities/
Match Residues Similarities for the Matched Region NOVl8b 17..92 76/76 (100%) 1..76 76/76 (100%) NOVl8c 17..92 76/76 (100%) 1..76 76/76 (100%) Further analysis of the NOV 18a protein yielded the following properties shown in Table 18C.
Table 18C. Protein Sequence Properties NOVl8a PSort 0.3700 probability located in outside; 0.1800 probability located in nucleus;
analysis: 0.1000 probability located in endoplasmic reticulum (membrane);
0.1000 probability located in endoplasmic reticulum (lumen) SignalP Cleavage site between residues 16 and 17 analysis:

A search of the NOV 18a protein against the Geneseq database, a proprietary database that contains sequences published in patents and patent publication, yielded several homologous proteins shown in Table 18D.

Table 18D. Geneseq Results for NOVl8a NOVl8a Identities/

Geneseq Protein/OrganismlLength Residues!SimilaritiesExpect [Patent for Identifier#, Date] Match the MatchedValue ResiduesRegion ABB72228 Human protein isolated 1..92 92/92 (100%)!e-50 from skin cells SEQ ID NO: 344 4..95 92/92 (100%) - Homo Sapiens, 95 aa. [W0200190357-Al, 29-NOV-2001 ]

AAB56028 Skin cell protein, SEQ 1..92 92/92 (100%)!e-50 ID NO: 344 -Homo Sapiens, 95 aa. 4..95 92/92 (1001) [W0200069884-A2, 23 NOV-2000]

AAB88478 Human membrane or secretory1..92 92/92 (100%)!e-50 protein clone PSEC0212 20..111 92/92 (100%) - Homo Sapiens, 111 aa. [EP1067182-A2, 10-JAN-2001 ]

AAE05371 Human huKSl protein - 1..92 92192 (100%)!e-50 Homo Sapiens, 95 aa. [W0200148192-Al,4..95 92/92 (100%) OS-JUL-2001]

AAY76089 Human CXC chemokine homologue1..92 92/92 (100%)!e-50 huKSl, SEQ ID N0:344 4..95 92!92 (100%) - Homo Sapiens, 95 aa. [W09955865-A1, 04-NOV-1999]

In a BLAST search of public sequence datbases, the NOV 18a protein was found to have homology to the proteins shown in the BLASTP data in Table I 8E.
Table 18E. Public BLASTP Results for NOVlBa Protein NOVl8a Identities/

AccessionProtein/Organism/Length Residues/SimilaritiesExpect for Number Match the MatchedValue ResiduesPortion JG0182 chemokine BRAK - human, 1..92 92/92 (100%)2e-50 99 aa.

8..99 ~
92/92 (100%) Q9BTR1 SMALL INDUCIBLE CYTOKINE 1..92 92/92 (100%)2e-SO

SUBFAMILY B (CYS-X-CYS), 20..1 92/92 ( I 1 100%) , MEMBER 14 (BRAK) - Homo sapiens (Human), 111 aa.

095715 Srriall inducible cytokine1..92 92/92 (100%)2e-50 B14 ~ 8 , precursor (Chemokine BRAK)..99 92/92 ( - 100%) Homo sapiens (Human), 99 aa.

Q9NS21 CHEMOKINE MIP-2 GAMMA 1..92 91 /92 (98%)9e-50 -Homo Sapiens (Human), 20..111 91 /92 (98%) 111 aa. . ~

Q91 V02 MIP2GAMMA - Mus musculus 1..92 87/92 (94%)7e-48 (Mouse), 95 as (fragment).4..95 90/92 (97%) PFam analysis predicts that the NOV 18a protein contains the domains shown in the Table 18F.
Table 18F. Domain Analysis of NOVlBa Identities/
Pfam Domain NOVl8a Match Region ~ Similarities Expect Value for the Matched Region EXAMPLE 19.
The NOV 19 clone was analyzed, and the nucleotide and encoded polypeptide sequences are shown in Table 19A.
Table 19A. NOV19 Sequence Analysis SEQ ID NO: 107 619 by NOVl9a, GCTGCCTGCCTCCTCATGTTCCCCTCCACCACAGCGGACTGCCTGTCGCGGTGCTCCT

DNA G~TGCCAGGCTGCCCTGCTGCCCTCTGAGGAATGGGAGAGATGCCAGAGCTTTCTG
TCTTTTTTCACCCCCTCCACCCTTGGGCTCAATGACAAGGAGGACTTGGGGAGCAAGT

Sequence CGGTTGGGGAAGGGCCCTACAGTGAGCTGGCCAAGCTCTCTGGGTCATTCCTGAAGGA
GCTGAACGATGGTGCCATGGAGACTGGCACACTCTATCTCGCTGAGGAGGACCCCAAG
GAGCAGGTCAAACGCTATGGGGGCTTTTTGCGCAAATACCCCAAGAGGAGCTCAGAGG
TGGCTGGGGAGGGGGACGGGGATAGCATGGGCCATGAGGACCTGTACAAACGCTATGG
GGGCTTCTTGCGGCGCATTCGTCCCAAGCTCAAGTGGGACAACCAGAAGCGCTATGGC
GGTTTTCTCCGGCGCCAGTTCAAGGTGGTGACTCGGTCTCAGGAAGATCCGAATGCTT
ACTCTGGAGAGCTTTTTGATGCATAAGCACTTCTTTTCA
OItF Start: at 1 ORF Stop: TAA at 604 SEQ 1D NO: 108 201 as MW at 22447.11cD
NOVl9a, AACLLMFPSTTADCLSRCSLCAVKTQDGPKPINPLICSLQCQAALLPSEEWERCQSFL

P1'Oteln EQVKRYGGFLRKYPKRSSEVAGEGDGDSMGAEDLYKRYGGFLRRIRPKLKWDNQKRYG
GFLRRQFKVVTRSQEDPNAYSGELFDA
Sequence Further analysis of the NOV 19a protein yielded the following properties shown in Table 19B.
Table 19B. Protein Sequence Properties NOVl9a PSort 0.7562 probability located in mitochondria) matrix space; 0.4352 probability analysis: located in mitochondria) inner membrane; 0.4352 probability located in mitochondria) intermembrane space; 0.4352 probability located in mitochondria) outer membrane SignalP Cleavage site between residues 13 and 14 analysis: , A search of the NOV 19a protein against the Geneseq database, a proprietary database that contains sequences published in patents and patent publication, yielded several homologous proteins shown in Table 19C.

Table 19C. Geneseq Results for NOVl9a NOVl9a Identities/

Geneseq Protein/Organism/Length Residues/SimilaritiesExpect [Patent for Identifier#, Date] Match the MatchedValue ResiduesRegion AAM79544 Human protein SEQ ID 1..201 201/246 e-110 NO 3190 - (81%) Homo Sapiens, 256 aa. 11..256 201/246 (81%) [W0200157190-A2, 09-AUG-2001]

AAM78560 Human protein SEQ ID 1..201 201/246 e-110 NO 1222 - (81%) Homo Sapiens, 254 aa. 9..254 201/246 (81%) [W0200157190-A2, 09-AUG-2001 ]

AAM05438 Peptide #4120 encoded 135..20167/67 (100%)2e-34 by probe for measuring breast 1..67 67/67 (100%) gene expression - Homo sapiens, 67 aa, [W0200157270-A2, 09-AUG-2001 ]

AAM30301 Peptide #4338 encoded 135..20167/67 (100%)2e-34 by probe for measuring placental 1..67 67/67 (100%) gene expression - Homo Sapiens, 67 aa.

[W0200157272-A2, 09-AUG-2001]

AAM1779I Peptide #4225 encoded 135..20167/67 (100%)2e-34 by probe for measuring cervical I ..67 67/67 (100%) gene expression - Homo sapiens, 67 aa.

[W0200157278-A2, 09-AUG-2001]

In a BLAST search of public sequence datbases, the NOV 19a protein was found to have homology to the proteins shown in the BLASTP data in Table 19D.

Table 19D. Public BLASTP Results for NOVl9a Protein NOVl9a Identities!

AccessionProtein/Organism/Length Residues/SimilaritiesExpect for Number Match the MatchedValue ResiduesPortion P41213 Beta-neoendorphin-dynorphin1..201 201/246 e-110 (81%) precursor (Proenkephalin 9..254 201/246 B) (81%) (Preprodynorphin) [Contains:
Beta-neoendorphin; Dynorphin;
Leu-Enkephalin; Rimorphin;
Leumorphin]

- Homo Sapiens (Human), 254 aa.

P01214 Beta-ne~endorphin-dynorphin1..200 164/247 2e-84 (66%) precursor (Proenkephalin 9..255 171/247 B) (68%) (Preprodynorphin) [Contains:
Beta-neoendorphin; Dynorphin;
Leu-Enkephalin; Rimorphin; , LeumorphinJ

- Sus scrofa (Pig), 256 aa.

Q95104 Beta-neoendorphin-dynorphin1..200 155/249 7e-79 (62%) precursor (Proenkephalin 9..257 170/249 B) (68%) (Preprodynorphin) [Contains:
Beta-neoendorphin; Dynorphin;
Leu-Enkephalin; Rimorphin;
Leumorphin]

- Bos taurus (Bovine), 258 aa.

Q60478 Beta-neoendorphin-dynorphin1..200 153/238 3e-77 (64%) precursor (Proenkephalin 9..244 165/238 B) (69%) (Preprodynorphin) [Contains:
Beta-neoendorphin; Dynorphin;
Leu-Enkephalin; Rimorphin;
Leumorphin]

- Cavia porcellus (Guinea pig), 245 aa.

035852 PREPRODYNORPHIN - Mus 1..198 140/238 4e-69 {58%) rriusculus (Mouse), 248 9..246 157/238 as (65%) (fragment).

PFam analysis predicts that the NOVl9a protein contains the domains shown in the Table 19E.
Table 19E. Domain Analysis of NOVl9a Identities/
Pfam Domain NOVl9a Match Region Similarities Expect Value for the Matched Region Opiods_neuropep 1..201 145/267 (54%) 1.3e-115 197/267 (74%) Example B: Sequencing Methodology and Identification of NOVX Clones 1. GeneCalling~ Technology: This is a proprietary method of performing differential gene expression profiling between two or more samples developed at CuraGen and described by Shimkets, et al., "Gene expression analysis by transcript profiling coupled to a gene database query" Nature Biotechnology 17:198-803 (1999). cDNA
was derived from various human samples representing multiple tissue types, normal and diseased states, physiological states, and developmental states from different donors.
Samples were obtained as whole tissue, primary cells or tissue cultured primary cells or cell lines. Cells and cell lines may have been treated with biological or chemical agents that regulate gene expression, for example, growth factors, chemokines or steroids. The cDNA thus derived was then digested with up to as many as 120 pairs of restriction enzymes and pairs of linker-adaptors specific for each pair of restriction enzymes were ligated to the appropriate end. The restriction digestion generates a mixture of unique cDNA gene fragments. Limited PCR amplification is performed with primers homologous to the linker adapter sequence where one primer is biotinylated and the other is fluorescently labeled. The doubly labeled material is isolated and the fluorescently labeled single strand is resolved by capillary gel electrophoresis. A computer algorithm compares the electropherograms from an experimental and control group for each of the restriction digestions. This and additional sequence-derived information is used to predict the identity of each differentially expressed gene fragment using a variety of genetic databases. The identity of the gene fragment is confirmed by additional, gene-specific competitive PCR or by isolation and sequencing of the gene fragment.
2. SeqCallingTM Technology: cDNA was derived from various human samples representing multiple tissue types, normal and diseased states, physiological states, and developmental states from different donors. Samples were obtained as whole tissue, primary cells or tissue cultured primary cells or cell lines. Cells and cell lines may have been treated with biological or chemical agents that regulate gene expression, for example, growth factors, chemokines or steroids. The cDNA thus derived was then sequenced using CuraGen's proprietary SeqCalling technology. Sequence traces were evaluated manually and edited for corrections if appropriate. cDNA sequences from all samples were assembled together, sometimes including public human sequences, using bioinformatic programs to produce a consensus sequence for each assembly. Each assembly is included in CuraGen Corporation's database. Sequences were included as components for assembly when the extent of identity with another component was at least 95% over SO
bp. Each assembly represents a gene or portion thereof and includes information on variants, such as splice forms single nucleotide polymorphisms (SNPs), insertions, deletions and other sequence variations.
3. PathCallingTM Technology:
The NOVX nucleic acid sequences are derived by laboratory screening of cDNA
library by the two-hybrid approach. cDNA fragments covering either the full length of the DNA sequence, or part of the sequence, or both, are sequenced. In silico prediction was based on sequences available in CuraGen Corporation's proprietary sequence databases or in the public human sequence databases, and provided either the full length DNA
sequence, or some portion thereof.
The laboratory screening was performed using the methods summarized below:
cDNA libraries were derived from various human samples representing multiple tissue types, normal and diseased states, physiological states, and developmental states from different donors. Samples were obtained as whole tissue, primary cells or tissue cultured primary cells or cell lines. Cells and cell lines may have been treated with biological or chemical agents that regulate gene expression, for example, growth factors, chemokines or steroids. The cDNA thus derived was then directionally cloned into the appropriate two-hybrid vector (Gal4-activation domain (Gal4-AD) fusion). Such cDNA
libraries as well as commercially available cDNA libraries from Clontech (Palo Alto, CA) were then transferred from E.coli into a CuraGen Corporation proprietary yeast strain (disclosed in LT. S. Patents 6,057,101 and 6,083,693, incorporated herein by reference in their entireties).
Gal4-binding domain (Gal4-BD) fusions of a CuraGen Corportion proprietary library of human sequences was used to screen multiple Gal4-AD fusion cDNA
libraries resulting in the selection of yeast hybrid diploids in each of which the Gal4-AD fusion contains an individual cDNA. Each sample was amplified using the polymerase chain reaction (PCR) using non-specific primers at the cDNA insert boundaries. Such PCR
product was sequenced; sequence traces were evaluated manually and edited for corrections if appropriate. cDNA sequences from all samples were assembled together, sometimes including public human sequences, using bioinformatic programs to produce a consensus sequence for each assembly. Each assembly is included in CuraGen Corporation's database. Sequences were included as components for assembly when the extent of identity with another component was at least 95% over 50 bp. Each assembly represents a gene or portion thereof and includes information on variants, such as splice forms single nucleotide polymorphisms (SNPs), insertions, deletions and other sequence variations.
Physical clone: the cDNA fragment derived by the screening procedure, covering the entire open reading frame is, as a recombinant DNA, cloned into pACT2 plasmid (Clontech) used to make the cDNA library. The recombinant plasmid is inserted into the host and selected by the yeast hybrid diploid generated during the screening procedure by the mating of both CuraGen Corporation proprietary yeast strains N106' and YULH (U. S.
Patents 6,057,101 and 6,083,693).
4. RACE: Techniques based on the polymerase chain reaction such as rapid amplification of cDNA ends (RACE), were used to isolate or complete the predicted sequence of the cDNA of the invention. Usually multiple clones were sequenced from one or more human samples to derive the sequences for fragments. Various human tissue samples from different donors were used for the RACE reaction. The sequences derived from these procedures were included in the SeqCalling Assembly process described in preceding paragraphs.
5. Exon Linking: The NOVX target sequences identified in the present invention were subjected to the exon linking process to confirm the sequence. PCR
primers were designed by starting at the most upstream sequence available, for the forward primer, and at the most downstream sequence available for the reverse primer. In each case, the sequence was examined, walking inward from the respective termini toward the coding sequence, until a suitable sequence that is either unique or highly selective was encountered, or, in the case of the reverse primer, until the stop codon was reached. Such primers were designed based on in silico predictions for the full length cDNA, part (one or more exons) of the DNA or protein sequence of the target sequence, or by translated homology of the predicted exons to closely related human sequences from other species.
These primers were then employed in PCR amplification based on the following pool of human cDNAs: adrenal gland, bone marrow, brain - amygdala, brain - cerebellum, brain -hippocampus, brain - substantia nigra, brain - thalamus, brain -whole, fetal brain, fetal kidney, fetal liver, fetal lung, heart, kidney, lymphoma - Raji, mammary gland, pancreas, pituitary gland, placenta, prostate, salivary gland, skeletal muscle, small intestine, spinal cord, spleen, stomach, testis, thyroid, trachea, uterus. Usually the resulting amplicons were gel purified, cloned and sequenced to high redundancy. The PCR product derived from exon linking was cloned into the pCR2.1 vector from Invitrogen. The resulting bacterial clone has an insert covering the entire open reading frame cloned into the pCR2.1 vector. The resulting sequences from all clones were assembled with themselves, with other fragments in CuraGen Corporation's database and with public ESTs.
Fragments and ESTs were included as components for an assembly when the extent of their identity with another component of the assembly was at least 95% over 50 bp. In addition, sequence traces were evaluated manually and edited for corrections if appropriate.
These procedures provide the sequence reported herein.
6. Physical Clone: Exons were predicted by homology and the intron/exon boundaries were determined using standard genetic rules. Exons were further selected and refined by means of similarity determination using multiple BLAST (for example, tBIastN, BIastX, and BlastN) searches, and, in some instances, GeneScan and Grail.
Expressed sequences from both public and proprietary databases were also added when available to further define and complete the gene sequence. The DNA sequence was then manually corrected for apparent inconsistencies thereby obtaining the sequences encoding the full-length protein.
The PCR product derived by exon linking, covering the entire open reading frame, was cloned into the pCR2.1 vector from Invitrogen to provide clones used for expression and screening purposes.
Example C: Quantitative expression analysis of clones in various cells and tissues The quantitative expression of various clones was assessed using microtiter plates containing RNA samples from a variety of normal and pathology-derived cells, cell lines and tissues using real time quantitative PCR (RTQ PCR). RTQ PCR was performed on an Applied Biosystems ABI PRISM~ 7700 or an ABI PRISM~ 7900 HT Sequence Detection System. Various collections of samples are assembled on the plates, and referred to as Panel 1 (containing normal tissues and cancer cell lines), Panel 2 (containing samples derived from tissues from normal and cancer sources), Panel 3 (containing cancer cell lines), Panel 4 (containing cells and cell lines from normal tissues and cells related to inflammatory conditions), Panel 5D/5I (containing human tissues and cell lines with an emphasis on metabolic diseases), AI comprehensive~anel (containing normal tissue and samples from autoinflammatory diseases), Panel CNSD.O1 (containing samples from normal and diseased brains) and CNS neurodegeneration~anel (containing samples from normal and Alzheimer's diseased brains).
RNA integrity from all samples is controlled for quality by visual assessment of agarose gel electropherograms using 28S and 18S ribosomal RNA staining intensity ratio as a guide (2:1 to 2.5:1 28s:18s) and the absence of low molecular weight RNAs that would be indicative of degradation products. Samples are controlled against genomic DNA contamination by RTQ PCR reactions run in the absence of reverse transcriptase using probe and primer sets designed to amplify across the span of a single exon.
First, the RNA samples were normalized to reference nucleic acids such as constitutively expressed genes (for example, (3-actin and GAPDH). Normalized RNA (5 u1) was converted to cDNA and analyzed by RTQ-PCR using One Step RT-PCR Master Mix Reagents (Applied Biosystems; Catalog No. 4309169) and gene-specific primers according to the manufacturer's instructions.
In other cases, non-normalized RNA samples were converted to single strand cDNA (sscDNA) using Superscript II (Invitrogen Corporation; Catalog No. 18064-147) and random hexamers according to the manufacturer's instructions. Reactions containing up to 10 g.g of total RNA were performed in a volume of 20 p1 and incubated for 60 minutes at 42°C. This reaction can be scaled up to 50 pg of total RNA
in a final volume of 100 p1. sscDNA samples are then normalized to reference nucleic acids as described previously, using 1X TaqMan~ Universal Master mix (Applied Biosystems; catalog No.
4324020), following the manufacturer's instructions.
Probes and primers were designed for each assay according to Applied Biosystems Primer Express Software package (version I for Apple Computer's Macintosh Power PC) or a similar algorithm using the target sequence as input. Default settings were used for reaction conditions and the following parameters were set before selecting primers: primer concentration = 250 nM, primer melting temperature (Tm) range = 5 8°-60°C, primer optimal Tm = 59°C, maximum primer difference = 2°C, probe does not have 5'G, probe Tm must be 10°C greater than primer Tm, amplicon size 75bp to 100bp.
The probes and primers selected (see below) were synthesized by Synthegen (Houston, TX, USA).
Probes were double purified by HPLC to remove uncoupled dye and evaluated by mass spectroscopy to verify coupling of reporter and quencher dyes to the 5' and 3' ends of the probe, respectively. Their final concentrations were: forward and reverse primers, 900nM
each, and probe, 200nM.
PCR conditions: When working with RNA samples, normalized RNA from each tissue and each cell line was spotted in each well of either a 96 well or a 384-well PCR
plate (Applied Biosystems). PCR cocktails included either a single gene specific probe and primers set, or two multiplexed probe and primers sets (a set specific for the target clone and another gene-specific set multiplexed with the target probe). PCR
reactions were set up using TaqMan~ One-Step RT-PCR Master Mix (Applied Biosystems, Catalog No.
4313803) following manufacturer's instructions. Reverse transcription was performed at 48°C for 30 minutes followed by amplification/PCR cycles as follows:
95°C 10 min, then 40 cycles of 95°C for 15 seconds, 60°C for 1 minute. Results were recorded as CT values (cycle at which a given sample crosses a threshold level of fluorescence) using a log scale, with the difference in RNA concentration between a given sample and the sample with the lowest CT value being represented as 2 to the power of delta CT. The percent relative expression is then obtained by taking the reciprocal of this RNA difference and multiplying by 100. ' When working with sscDNA samples, normalized sscDNA was used as described previously for RNA samples. PCR reactions containing one or two sets of probe and primers were set up as described previously, using 1X TaqMan~ Universal Master mix (Applied Biosystems; catalog No. 4324020), following the manufacturer's instructions.
PCR amplification was performed as follows: 95°C 10 min, then 40 cycles of 95°C for 15 seconds, 60°C for 1 minute. Results were analyzed and processed as described previously.
Panels 1,1.1,1.2, and 1.3D
The plates for Panels 1, 1.1, I .2 and 1.3D include 2 control wells (genomic DNA
control and chemistry control) and 94 wells containing cDNA from various samples. The samples in these panels are broken into 2 classes: samples derived from cultured cell lines and samples derived from primary normal tissues. The cell lines are derived from cancers of the following types: lung cancer, breast cancer, melanoma, colon cancer, prostate cancer, CNS cancer, squamous cell carcinoma, ovarian cancer, liver cancer, renal cancer, gastric cancer and pancreatic cancer. Cell lines used in these panels are widely available through the American Type Culture Collection (ATCC), a repository for cultured cell lines, and were cultured using the conditions recommended by the ATCC. The normal tissues found on these panels are comprised of samples derived from all major organ systems from single adult individuals or fetuses. These samples are derived from the following organs: adult skeletal muscle, fetal skeletal muscle, adult heart, fetal heart, adult kidney, fetal kidney, adult liver, fetal liver, adult lung, fetal lung, various regions of the brain, the spleen, bone marrow, lymph node, pancreas, salivary gland, pituitary gland, adrenal gland, spinal cord, thymus, stomach, small intestine, colon, bladder, trachea, breast, ovary, uterus, placenta, prostate, testis and adipose.
In the results for Panels l, 1.1, 1.2 and 1.3D, the following abbreviations are used:
ca. = carcinoma, * = established from metastasis, met = metastasis, s cell var = small cell variant, non-s = non-sm = non-small, squam = squamous, p1. eff = p1 effusion = pleural effusion, glio = glioma, astro = astrocytoma, and neuro = neuroblastoma.
General screening_panel v1.4, v1.5 and v1.6 The plates for Panels 1.4, 1.5, and 1.6 include 2 control wells (genomic DNA
control and chemistry control) and 94 wells containing cDNA from various samples. The samples in Panels 1.4, 1.5, and 1.6 are broken into 2 classes: samples derived from cultured cell lines and samples derived from primary normal tissues. The cell lines are derived from cancers of the following types: lung cancer, breast cancer, melanoma, colon cancer, prostate cancer, CNS cancer, squamous cell carcinoma, ovarian cancer, liver cancer, renal cancer, gastric cancer and pancreatic cancer. Cell lines used in Panels 1.4, 1.5, and I .6 are widely available through the American Type Culture Collection (ATCC), a repository for cultured cell lines, and were cultured using the conditions recommended by the ATCC. The normal tissues found on Panels 1.4, 1.5, and 1.6 are comprised of pools of samples derived from all major organ systems from 2 to 5 different adult individuals or fetuses. These samples are derived from the following organs: adult skeletal muscle, fetal skeletal muscle, adult heart, fetal heart, adult kidney, fetal kidney, adult liver, fetal liver, adult lung, fetal lung, various regions of the brain, the spleen, bone marrow, lymph node, pancreas, salivary gland, pituitary gland, adrenal gland, spinal cord, thymus, stomach, small intestine, colon, bladder, trachea, breast, ovary, uterus, placenta, prostate, testis and adipose. Abbreviations are as described for Panels 1, I .I, 1.2, and 1.3D.
Panels 2D, 2.2, 2.3 and 2.4 1~4 The plates for Panels 2D, 2.2, 2.3 and 2.4 generally include 2 control wells and 94 test samples composed of RNA or cDNA isolated from human tissue procured by surgeons working in close cooperation with the National Cancer Institute's Cooperative Human Tissue Network (CHTN) or the National Disease Research Initiative (NDRI) or from Ardais or Clinomics). The tissues are derived from human malignancies and in cases where indicated many malignant tissues have "matched margins" obtained from noncancerous tissue just adjacent to the tumor. These are termed normal adjacent tissues and are denoted "NAT" in the results below. The tumor tissue and the "matched margins"
are evaluated by two independent pathologists (the surgical pathologists and again by a pathologist at NDRI/ CHTN/Ardais/Clinomics). Unmatched RNA samples from tissues without malignancy (normal tissues) were also obtained from Ardais or Clinomics. This analysis provides a gross histopathological assessment of tumor differentiation grade.
Moreover, most samples include the original surgical pathology report that provides information regarding the clinical stage of the patient. These matched margins are taken from the tissue surrounding (i.e. immediately proximal) to the zone of surgery (designated "NAT", for normal adjacent tissue, in Table RR). In addition, RNA and cDNA
samples were obtained from various human tissues derived from autopsies performed on elderly people or sudden death victims (accidents, etc.). These tissues were ascertained to be free of disease and were purchased from various commercial sources such as Clontech (Palo Alto, CA), Research Genetics, and Invitrogen.
HASS Panel v 1.0 The HASS panel v 1.0 plates are comprised of 93 cDNA samples and two controls.
Specifically, 81 of these samples are derived from cultured human cancer cell lines that had been.subjected to serum starvation, acidosis and anoxia for different time periods as well as controls for these treatments, 3 samples of human primary cells, 9 samples of malignant brain cancer (4 medulloblastomas and 5 glioblastomas) and 2 controls. The human cancer cell lines are obtained from ATCC (American Type Culture Collection) and fall into the following tissue groups: breast cancer, prostate cancer, bladder carcinomas, pancreatic cancers and CNS cancer cell lines. These cancer cells are all cultured under standard recommended conditions. The treatments used (serum starvation, acidosis and anoxia) have been previously published in the scientific literature. The primary human cells were obtained from Clonetics (Walkersville, MD) and were grown in the media and conditions recommended by Clonetics. The malignant brain cancer samples are obtained as part of a collaboration (Henry Ford Cancer Center) and are evaluated by a pathologist prior to CuraGen receiving the samples . RNA was prepared from these samples using the standard procedures. The genomic and chemistry control wells have been described previously.
Panel 3D and 3.1 The plates of Panel 3D and 3.1 are comprised of 94 cDNA samples and two control samples. Specifically, 92 of these samples are derived from cultured human cancer cell lines, 2 samples of human primary cerebellar tissue and 2 controls. The human cell lines are generally obtained from ATCC (American Type Culture Collection), NCI
or the German tumor cell bank and fall into the following tissue groups: Squamous cell carcinoma of the tongue, breast cancer, prostate cancer, melanoma, epidennoid carcinoma, sarcomas, bladder carcinomas, pancreatic cancers, kidney cancers, leukemias/lymphomas, ovarian/uterine/cervical, gastric, colon, lung and CNS cancer cell lines. In addition, there are two independent samples of cerebellum. These cells are all cultured under standard recommended conditions and RNA extracted using the standard procedures. The cell lines in panel 3D, 3.1 and 1.3D are of the most common cell lines used in the scientific literature.
Panels 4D, 4R, and 4.1D
Panel 4 includes samples on a 96 well plate (2 control wells, 94 test samples) composed of RNA (Panel 4R) or cDNA (Panels 4D14.1D) isolated from various human cell lines or tissues related to inflammatory conditions. Total RNA from control normal tissues such as colon and lung (Stratagene, La Jolla, CA) and thymus and kidney (Clontech) was employed. Total RNA from liver tissue from cirrhosis patients and kidney from lupus patients was obtained from BioChain (Biochain Institute, Inc., Hayward, CA).
Intestinal tissue for RNA preparation from patients diagnosed as having Crohn's disease and ulcerative colitis was obtained from the National Disease Research Interchange (NDRI) (Philadelphia, PA).
Astrocytes, lung fibroblasts, dermal fibroblasts, coronary artery smooth muscle cells, small airway epithelium, bronchial epithelium, microvascular dermal endothelial cells, microvascular lung endothelial cells, human pulmonary aortic endothelial cells, human umbilical vein endothelial cells were all purchased from Clonetics (Walkersville, MD) and grown in the media supplied for these cell types by Clonetics. These primary cell types were activated with various cytokines or combinations of cytokines for 6 andlor 12-14 hours, as indicated. The following cytokines were used; IL-1 beta at approximately I-Sng/ml, TNF alpha at approximately 5-l Onglml, IFN gamma at approximately 20-SOng/ml, IL-4 at approximately S-IOng/ml, IL-9 at approximately 5-lOnglml, IL-13 at approximately 5-l Ong/ml. Endothelial cells were sometimes starved for various times by culture in the basal media from Clonetics with 0.1% serum.
Mononuclear cells were prepared from blood of employees at CuraGen Corporation, using Ficoll. LAK cells were prepared from these cells by culture in DMEM
5% FCS (Hyclone), 100~,M non essential amino acids (Gibco/Life Technologies, Rockville, MD), 1mM sodium pyruvate (Gibco), mercaptoethanol S.SxlO-SM
(Gibco), and I OmM Hepes (Gibco) and Interleukin 2 for 4-6 days. Cells were then either activated with 10-20ng/ml PMA and 1-2pg/ml ionomycin, IL-12 at 5-l Ong/ml, IFN gamma at 20-SOng/ml and IL-18 at 5-lOng/ml for 6 hours. In some cases, mononuclear cells were cultured for 4-5 days in DMEM 5% FCS (Hyclone), I OOpM non essential amino acids (Gibco), 1mM sodium pyruvate (Gibco), mercaptoethanol S.SxlO'SM (Gibco), and lOmM
Hepes (Gibco) with PHA (phytohemagglutinin) or PWM (pokeweed mitogen) at approximately Spg/ml. Samples were taken at 24, 48 and 72 hours for RNA
preparation.
MLR (mixed lymphocyte reaction) samples were obtained by taking blood from two donors, isolating the mononuclear cells using Ficoll and mixing the isolated mononuclear cells 1:1 at a final concentration of approximately 2x106cells/ml in DMEM 5%
FCS
(Hyclone), IOOpM non essential amino acids (Gibco), ImM sodium pyruvate (Gibco), mercaptoethanol (S.SxlO-SM) (Gibco), and l OmM Hepes (Gibco). The MLR was cultured and samples taken at various time points ranging from 1- 7 days for RNA
preparation.
Monocytes were isolated from mononuclear cells using CD14 Miltenyi Beads, +ve VS selection columns and a Vario Magnet according to the manufacturer's instructions:
Monocytes were differentiated into dendritic cells by culture in DMEM 5% fetal calf serum (FCS) (Hyclone, Logan, UT), 100p.M non essential amino acids (Gibco), 1mM
sodium pyruvate (Gibco), mercaptoethanol S.SxlO'SM (Gibco), and l OmM Hepes (Gibco), SOng/ml GMCSF and Sng/ml IL-4 for 5-7 days. Macrophages were prepared by culture of monocytes for 5-7 days in DMEM 5% FCS (Hyclone), 100pM non essential amino acids (Gibco), 1mM sodium pyruvate (Gibco), mercaptoethanol S.SxlO'SM (Gibco), lOmM
Hepes (Gibco) and 10% AB Human Serum or MCSF at approximately SOng/ml.
Monocytes, macrophages and dendritic cells were stimulated for 6 and 12-14 hours with lipopolysaccharide (LPS) at 100ng/ml. Dendritic cells were also stimulated with anti-CD40 monoclonal antibody (Pharmingen) at lOpg/ml for 6 and 12-14 hours.
CD4 lymphocytes, CD8 lymphocytes and NK cells were also isolated from mononuclear cells using CD4, CD8 and CD56 Miltenyi beads, positive VS
selection columns and a Vario Magnet according to the manufacturer's instructions.
CD45RA and CD45R0 CD4 lymphocytes were isolated by depleting mononuclear cells of CDB, CD56, CD14 and CD19 cells using CDB, CD56, CD14 and CD19 Miltenyi beads and positive selection. CD45R0 beads were then used to isolate the CD45R0 CD4 lymphocytes with the remaining cells being CD45RA CD4 lymphocytes. CD45RA CD4, CD45R0 CD4 and CD8 lymphocytes were placed in DMEM 5% FCS (Hyclone), 100p,M non essential amino acids (Gibco), 1mM sodium pyruvate (Gibco), mercaptoethanol S.SxlO-SM (Gibco), and l OmM Hepes (Gibco) and plated at 10&cells/ml onto Falcon 6 well tissue culture plates that had been coated overnight with O.S~,g/ml anti-CD28 (Pharmingen) and 3ug/ml anti-CD3 (OKT3, ATCC) in PBS. After 6 and 24 hours, the cells were harvested for RNA
preparation. To prepare chronically activated CD8 lymphocytes, we activated the isolated CD8 lymphocytes for 4 days on anti-CD28 and anti-CD3 coated plates and then harvested the cells and expanded them in DMEM 5% FCS (Hyclone), 100pM non essential amino acids (Gibco), 1mM sodium pyruvate (Gibco), mercaptoethanol S.SxlO'SM (Gibco), and l OmM Hepes (Gibco) and IL-2. The expanded CD8 cells were then activated again with plate bound anti-CD3 and anti-CD28 for 4 days and expanded as before. RNA was isolated 6 and 24 hours after the second activation and after 4 days of the second expansion culture. The isolated NK cells were cultured in DMEM 5% FCS
(Hyclone), 100pM non essential amino acids (Gibco), 1mM sodium pyruvate (Gibco), mercaptoethanol S.SxlO-SM (Gibco), and l OmM Hepes (Gibco) and IL-2 for 4-6 days before RNA was prepared.
To obtain B cells, tonsils were procured from NDRI. The tonsil was cut up with sterile dissecting scissors and then passed through a sieve. Tonsil cells were then spun down and resupended at 106cells/ml in DMEM 5% FCS (Hyclone), 100~M non essential amino acids (Gibco), 1mM sodium pyruvate (Gibco), mercaptoethanol S.SxlO-SM
(Gibco), and lOmM Hepes (Gibco). To activate the cells, we used PWM at Spg/ml or anti-(Pharmingen) at approximately l Opg/ml and IL-4 at 5-l Onglml. Cells were harvested for RNA preparation at 24,48 and 72 hours.
To prepare the primary and secondary Thl/T'h2 and Tr1 cells, six-well Falcon plates were coated overnight with l Opg/ml anti-CD28 (Pharmingen) and 2pg/ml (ATCC), and then washed twice with PBS. Umbilical cord blood CD4 lymphocytes (Poietic Systems, German Town, MD) were cultured at 105-106cells/ml in DMEM S%
FCS (Hyclone), 100pM non essential amino acids (Gibco), 1mM sodium pyruvate (Gibco), mercaptoethanol S.SxlO'SM (Gibco), IOmM Hepes (Gibco) and IL-2 (4ng/ml).
IL-12 (Sng/ml) and anti-IL4 (1 pg/ml) were used to direct to Thl, while IL-4 (Snglml) and anti-IFN gamma (1 pg/ml) were used to direct to Th2 and IL-10 at Sng/ml was used to direct to Trl . After 4-S days, the activated Thl, Th2 and Trl lymphocytes were washed once in DMEM and expanded for 4-7 days in DMEM S% FCS (Hyclone), 100pM non essential amino acids (Gibco), 1mM sodium pyruvate (Gibco), mercaptoethanol S.SxlO' SM (Gibco), lOmM Hepes (Gibco) and IL-2 (lng/ml). Following this, the activated Thl, Th2 and Trl lymphocytes were re-stimulated for S days with anti-CD28/OKT3 and cytokines as described above, but with the addition of anti-CD9SL (1 pg/ml) to prevent apoptosis. After 4-S days, the Thl, Th2 and Trl lymphocytes were washed and then expanded again with IL-2 for 4-7 days. Activated Thl and Th2 lymphocytes were maintained in this way for a maximum of three cycles. RNA was prepared from primary and secondary Thl, T'h2 and Trl after 6 and 24 hours following the second and third activations with plate bound anti-CD3 and anti-CD28 mAbs and 4 days into the second and third expansion cultures in Interleukin 2.
The following leukocyte cells lines were obtained from the ATCC: Ramos, EOL-1, KU-812. EOL cells were further differentiated by culture in O.lmM dbcAMP at Sx105cells/ml for 8 days, changing the media every 3 days and adjusting the cell concentration to Sxl Oscells/ml. For the culture of these cells, we used DMEM
or RPMI (as recommended by the ATCC), with the addition of S% FCS (Hyclone), 100~,M non essential amino acids (Gibco), 1mM sodium pyruvate (Gibco), mercaptoethanol S.SxlO' SM (Gibco), l OmM Hepes (Gibco). RNA was either prepared from resting cells or cells activated with PMA at l Ong/ml and ionomycin at 1 p.g/ml for 6 and 14 hours.
Keratinocyte line CCD106 and an airway epithelial tumor line NCI-H292 were also obtained from the ATCC. Both were cultured in DMEM S% FCS (Hyclone), 100p.M non essential amino acids (Gibco), 1mM sodium pyruvate (Gibco), mercaptoethanol S.SxlO'SM (Gibco), and lOmM Hepes (Gibco). CCD1106 cells were activated for 6 and 14 hours with approximately S ng/ml TNF alpha and lng/ml IL-1 beta, while NCI-H292 cells were activated for 6 and 14 hours with the following cytokines: Sng/ml IL-4, Sng/ml IL-9, Sng/ml IL-13 and 2Sng/ml IFN gamma.

For these cell lines and blood cells, RNA was prepared by lysing approximately l0~cells/ml using Trizol (Gibco BRL). Briefly, 1/10 volume of bromochloropropane (Molecular Research Corporation) was added to the RNA sample, vortexed and after 10 minutes at room temperature, the tubes were spun at 14,000 rpm in a Sorvall SS34 rotor.
The aqueous phase was removed and placed in a 15m1 Falcon Tube. An equal volume of isopropanol was added and left at -20°C overnight. The precipitated RNA
was spun down at 9,000 rpm for 15 min in a Sorvall SS34 rotor and washed in 70% ethanol. The pellet was redissolved in 300p1 of RNAse-free water and 35p1 buffer (Promega) Sp.l DTT, 7p,1 RNAsin and 8p1 DNAse were added. The tube was incubated at 37°C for 30 minutes to remove contaminating genomic DNA, extracted once with phenol chloroform and re-precipitated with 1/10 volume of 3M sodium acetate and 2 volumes of 100%
ethanol. The RNA was spun down and placed in RNAse free water. RNA was stored at -80°C.
AI comprehensive panel v1.0 The plates for AI comprehensive panel v1.0 include two control wells and 89 test samples comprised of cDNA isolated from surgical and postmortem human tissues obtained from the Backus Hospital and Clinomics (Frederick, MD). Total RNA was extracted from tissue samples from the Backus Hospital in the Facility at CuraGen. Total RNA from other tissues was obtained from Clinomics.
Joint tissues including synovial fluid, synovium, bone and cartilage were obtained from patients undergoing total knee or hip replacement surgery at the Backus Hospital.
Tissue samples were immediately snap frozen in liquid nitrogen to ensure that isolated RNA was of optimal quality and not degraded. Additional samples of osteoarthritis and rheumatoid arthritis joint tissues were obtained from Clinomics. Normal control tissues were supplied by Clinomics and were obtained during autopsy of trauma victims.
Surgical specimens of psoriatic tissues and adjacent matched tissues were provided as total RNA by Clinomics. Two male and two female patients were selected between the ages of 25 and 47. None of the patients were taking prescription drugs at the time samples were isolated.
Surgical specimens of diseased colon from patients with ulcerative colitis and Crohns disease and adjacent matched tissues were obtained from Clinomics.
Bowel tissue from three female and three male Crohn's patients between the ages of 41-69 were used.
Two patients were not on prescription medication while the others were taking dexamethasone, phenobarbital, or tylenol. Ulcerative colitis tissue was from three male and four female patients. Four of the patients were taking lebvid and two were on Phenobarbital.
Total RNA from post mortem lung tissue from trauma victims with no disease or with emphysema, asthma or COPD was purchased from Clinomics. Emphysema patients ranged in age from 40-70 and all were smokers, this age range was chosen to focus on patients with cigarette-linked emphysema and to avoid those patients with alpha-1 anti-trypsin deficiencies. Asthma patients ranged in age from 36-75, and excluded smokers to prevent those patients that could also have COPD. COPD patients ranged in age from 35-80 and included both smokers and non-smokers. Most patients were taking corticosteroids, and bronchodilators.
In the labels employed to identify tissues in the AI comprehensive panel v1.0 panel, the following abbreviations are used:
AI = Autoimmunity Syn = Synovial Normal = No apparent disease Rep22 /Rep20 = individual patients RA = Rheumatoid arthritis Backus = From Backus Hospital OA = Osteoarthritis (SS) (BA) (MF) = Individual patients Adj = Adjacent tissue Match control = adjacent tissues -M = Male -F = Female COPD = Chronic obstructive pulmonary disease Panels SD and SI
The plates for Panel SD and SI include two control wells and a variety of cDNAs isolated from human tissues and cell lines with an emphasis on metabolic diseases.
Metabolic tissues were obtained from patients enrolled in the Gestational Diabetes study.
Cells were obtained during different stages in the differentiation of adipocytes from human mesenchymal stem cells. Human pancreatic islets were also obtained.
In the Gestational Diabetes study subjects are young (18 - 40 years), otherwise healthy women with and without gestational diabetes undergoing routine (elective) Caesarean section. After delivery ofthe infant, when the surgical incisions were being repairedlclosed, the obstetrician removed a small sample (<1 cc) of the exposed metabolic tissues during the closure of each surgical level. The biopsy material was rinsed in sterile saline, blotted and fast frozen within 5 minutes from the time of removal. The tissue was then flash frozen in liquid nitrogen and stored, individually, in sterile screw-top tubes and kept on dry ice for shipment to or to be picked up by CuraGen. The metabolic tissues of interest include uterine wall (smooth muscle), visceral adipose, skeletal muscle (rectos) and subcutaneous adipose. Patient descriptions are as follows:
Patient 2: Diabetic Hispanic, overweight, not on insulin Patient 7-9: Nondiabetic Caucasian and obese (BMI>30) Patient 10: Diabetic Hispanic, overweight, on insulin Patient 11: Nondiabetic African American and overweight Patient 12: Diabetic Hispanic on insulin Adiocyte differentiation was induced in donor progenitor cells obtained from Osirus (a division of Clonetics/BioWhittaker) in triplicate, except for Donor 3U which had only two replicates. Scientists at Clonetics isolated, grew and differentiated human mesenchymal stem cells (HuMSCs) for CuraGen based on the published protocol found in Mark F. Pittenger, et al., Multilineage Potential of Adult Human Mesenchymal Stem Cells Science Apr 2 1999: 143-147. Clonetics provided Trizol lysates or frozen pellets suitable for mRNA isolation and ds cDNA production. A general description of each donor is as follows:
Donor 2 and 3 U: Mesenchymal Stem cells, Undifferentiated Adipose Donor 2 and 3 AM: Adipose, AdiposeMidway Differentiated Donor 2 and 3 AD: Adipose, Adipose Differentiated Human cell lines were generally obtained from ATCC (American Type Culture Collection), NCI or the German tumor cell bank and fall into the following tissue groups:
kidney proximal convoluted tubule, uterine smooth muscle cells, small intestine, liver HepG2 cancer cells, heart primary stromal cells, and adrenal cortical adenoma cells. These cells are all cultured under standard recommended conditions and RNA extracted using the standard procedures. All samples were processed at CuraGen to produce single stranded cDNA.
Panel SI contains all samples previously described with the addition of pancreatic islets from a 58 year old female patient obtained from the Diabetes Research Institute at the University of Miami School of Medicine. Islet tissue was processed to total RNA at an outside source and delivered to CuraGen for addition to panel SI.
In the labels employed to identify tissues in the SD and SI panels, the following abbreviations are used:
GO Adipose = Greater Omentum Adipose SK = Skeletal Muscle UT = Uterus PL = Placenta AD = Adipose Differentiated AM = Adipose Midway Differentiated U = Undifferentiated Stem Cells Panel CNSD.Ol The plates for Panel CNSD.O1 include two control wells and 94 test samples comprised of cDNA isolated from postmortem human brain tissue obtained from the Harvard Brain Tissue Resource Center. Brains are removed from calvaria of donors between 4 and 24 hours after death, sectioned by neuroanatomists, and frozen at -80°C in liquid nitrogen vapor. All brains are sectioned and examined by neuropathologists to confirm diagnoses with clear associated neuropathology.
Disease diagnoses are taken from patient records. The panel contains two brains from each of the following diagnoses: Alzheimer's disease, Parkinson's disease, Huntington's disease, Progressive Supernuclear Palsy, Depression, and "Normal controls".
Within each of these brains, the following regions are represented: cingulate gyrus, temporal pole, globus palladus, substantia nigra, Brodman Area 4 (primary motor strip), Brodman Area 7 (parietal cortex), Brodman Area 9 (prefrontal cortex), and Brodman area 17 (occipital cortex). Not all brain regions are represented in all cases;
e.g., Huntington's disease is characterized in part by neurodegeneration in the globus palladus, thus this region is impossible to obtain from confirmed Huntington's cases. Likewise Parkinson's disease is characterized by degeneration of the substantia nigra making this region more difficult to obtain. Normal control brains were examined for neuropathology and found to be free of any pathology consistent with neurodegeneration.
In the labels employed to identify tissues in the CNS panel, the following abbreviations are used:
PSP = Progressive supranuclear palsy Sub Nigra = Substantia nigra Glob Palladus= Globus palladus Temp.Pole = Temporal pole Cing Gyr = Cingulate gyrus BA 4 = Brodman Area 4 Panel CNS Neurodegeneration_V1.0 The plates for Panel CNS Neurodegeneration V 1.0 include two control wells and 47 test samples comprised of cDNA isolated from postmortem human brain tissue obtained from the Harvard Brain Tissue Resource Center (McLean Hospital) and the Human Brain and Spinal Fluid Resource Center (VA Greater Los Angeles Healthcare System). Brains are removed from calvaria of donors between 4 and 24 hours after death, sectioned by neuroanatomists, and frozen at -80°C in liquid nitrogen vapor. All brains are sectioned and examined by neuropathologists to confirm diagnoses with clear associated neuropathology.
Disease diagnoses are taken from patient records. The panel contains six brains from Alzheimer's disease (AD) patients, and eight brains from "Normal controls" who showed no evidence of dementia prior to death. The eight normal control brains are divided into two categories: Controls with no dementia and no Alzheimer's like pathology (Controls) and controls with no dementia but evidence of severe Alzheimer's like pathology, (specifically senile plaque load rated as level 3 on a scale of 0-3; 0 = no evidence of plaques, 3 = severe AD senile plaque load). Within each of these brains, the following regions are represented: hippocampus, temporal cortex (Brodman Area 21 ), parietal cortex (Brodman area 7), and occipital cortex (Brodman area 17).
These regions were chosen to encompass all levels of neurodegeneration in AD. The hippocampus is a region of early and severe neuronal loss in AD; the temporal cortex is known to show neurodegeneration in AD after the hippocampus; the parietal cortex shows moderate neuronal death in the late stages of the disease; the occipital cortex is spared in AD and therefore acts as a "control" region within AD patients. Not all brain regions are represented in all cases.
In the labels employed to identify tissues in the CNS Neurodegeneration V 1.0 panel, the following abbreviations are used:
AD = Alzheimer's disease brain; patient was demented and showed AD-like pathology upon autopsy Control = Control brains; patient not demented, showing no neuropathology Control (Path) = Control brains; pateint not demented but showing sever AD-like pathology SupTemporal Ctx = Superior Temporal Cortex Inf Temporal Ctx = Inferior Temporal Cortex A. NOV2A and NOV2B: LRR Protein Expression of gene NOV2A and full length physical clone NOV2B was assessed using the primer-probe sets Ag4180, Ag6318, Ag6602, Ag6659 and Ag6702, described in Tables AA, AB, AC, AD and AE. Please note that NOV2A is recognized by primer-probe set Ag4180 only. Results of the RTQ-PCR runs are shown in Tables AF, AG, AH, AI, AJ
and AK.
Table AA. Probe Name Ag4180 Primers ~'.."'~'y,~~~'~uSequences Length Start SEQ
ID

PositionNo Forward 5'-tcttccagaaggacatcaactg-3'22 1347 109 Probe T~~~-cagcttcatccacttgagtttccagg-3'-26 1309 110 _....._. .... .................__._..............
.._....._.........._.............. ._.....
..............._..........._..........._...._. _ ........................_...._ _..........._................... .
........_.................._...._.....
._ _ _..

Reverse 5'-cccctcgtccaggatatagtac-3'22 1271 111 Table AB. Probe Name Ag6318 Primers .~..~...~...",~"."."~".".. ~",Length"..,..,StartSEQ
Sequences ~~.~~~~,"~."m.~",."._ ~ ID

PositionNo Forward 5'-gtagtgaagcaggatagttcataaatagaa-3'30 3 112 P1'Obe TET-5'-agtggaagcgccttctcatccttcat-3'-26 35 113 TAMRA ....... ...... . _... ... . .

Reverse 5'-gcagtggtcacgtttgga-3' 18 62 114 Table AC. Probe Name Ag6602 Primers Sequences ~LengthStart~~ SEQ
ID

1 PositionNo Forward 5'-gtgaggcggcagatcttc-3' 18 426 115 Probe T~~~-agctgaatcatctgcagcctgcatt-3'-25 444 116 Reverse 5'-attcccaggcatgatgct-3' 18 495 117 Table AD. Probe Name Ag6659 Primers .. .. ... . . . Se uences Leng Start SEQ
Y~.. _ . ....._.._......_...._........._..;..... ID
th q PositionNo _....~....._........ ...
_ ......
_........_..._....................._........_._...._....._.....................
.._....._..__..._.............._........._...._........._._........_...........
.........._......._........... . ......._._........
_........................__ _........_..... 118 Forward5 -gtgaggcggcagatcttc-3' 18 ._._....._._..........

_......._.............................._....._.................................
.......
__..............._.............................................................
.....__...

Probe T~~~-agctgaatcatctgcagcctgcatt-3'-25 444 119 Reverse 5'-attcccaggcatgatgct-3' 18 495 120 Table AE. Probe Name Ag6702 SEQ
Primers Len t m Se uences ~~ h. ~
, ~~ S ' Posi No tion _...._.'______~~...
..:~_.~_.._._._.__..:;:,..~._...._y~_......~~..................._..._......_.._ .~;,~..___......................._..._....._...................._..............
..................._.....;...~:...-~i :-.._ atcttc-3 ....~,_...__.._.............._...........
Forward 5 gtgaggcggcag ... .. .. ........1.8........ ....
.... .. .. . ._ ...... . ....... _. :. 121 ... .. .... ... ...._.. . .. ....4~6 .......................
...... ...
...

Probe '1'ET-5'-agctgaatcatctgcagcctgcatt-3'-25 444 122 _. .__._. .TAMRA . . _.

Reverse 5'-attcccaggcatgatgct-3' 18 495 123 Table AF. AI comprehensive panel v1.0 Rel. Ezp.(%) Rel. Ezp.(%).. ... ,~:",~"::,:~,.....ReI.Exp.(%) Rel.
Egp.(%) Tissue Name Ag6318, Run Ag6659, Run me Ag6318, Tissue Na Run ~ Ag6659, Run - -..._ _ 12427 Match 5.2 0.0 Control 1 0.5 0.0 F

Psoriasis-F
_.........._.._.......... __.._.... ..... ._..........._ ._.. ._._. _ ... . _.......... _ . . _._ . _..
..... _...._._.. _..._.. ... - ' ._.._....._ ....._...
.._ _...

~ 1 .
12418 :

, y 0 1.7 0 0.5 0.0 psoriasis M ~ ., 110968 COPD- 112723 Match M 2.5 1.6 Control 0.0 0.0 Psoriasis-M

4 4.9 10 S.1 M . psoriasis-M .

110989 112424 Match Emphysema-F 5.4 2.1 Control 5.2 0.0 Psoriasis-M

1.4 0.0 8.4 0 Emphysema.-F._..... _ .. ..'Psoriasis-M_.........
......... ~... . _._._........ .....

110993 112425 Match Emphysema-F 2.9 0.0 ~ps r ~ i 2.0 0.0 ~

_............_._......................_..........._.._..............._.........
...._..._.. ........._......._.s-M
......_.__......................._....._.........._.........._..._.........
....._ .....................__....:..........._...................
.....

._.
110994 1 104689 (MF) Emphysema-F 6.1 0.0 -OA Bone- 3.9 18.4 Backus x _.. _.... ...._......_....
.................................._........................._ 1046.............................................._ ...
..... . ........

2.3 ~ 0.0 IAdj "Normal"5.5 5.4 Em h sema-F Bone-Backus p y 104691 (MF) 110996 4.2 0.0 OA 8.9 19.3 Emphysema-F Synovium-:

Backus I 10997 ~ 104692 (BA) Asthma-M 2.2 0.0 ;OA Cartilage-1.3 0.0 ~Backus 111001 104694 (BA) Asthma-F 4~7 2.1 OA Bone- 6.7 4.5 Backus 111002 104695(BA) . 6.3 0.0 Adj "Normal" 7.1 10.8 Asthma-F

Bone-Backus 104696 (BA) 111003 Atopic4 0.0 ~O'~ 5 15 ~ 5 0 5 Asthma-F , Synovium- . .
~

.................. ............~Backus. ............... _ . .. ........

104700(SS) 111004 Atopic 5.4 0.0 OA Bone- 100.0 100.0 Asthma-F

. ......... .._ ...._..._.._. ... Backus.............._........_...
.. . .... .. .. .....~._. ._........._............._..._ ..............._......_ ... _ ...... . ... ...
..... _. .................... .. ..
..._.._...

. . ...._.
111005 Atopic 104701 (SS) ... .. .
.
.......
_ .

Asthma-F 1.3 0.0 r, 8~5 0.0 ~ ~

_.._......_ ...............__..................._._._......___....__............_.....__Bon e-Ba kus .._...._......_........._........_.._.
....................._.. __._............. ____ _....... _.._......._.__....._.
_._.__. _..._._._......._.
..._.._ . ....._.__......_ 104702 (SS) _ _..._.__.
._..__ 111006 Atopic OA

p.0 0.0 7.0 3.0 Asthma-F Synovium-Backus 111417 ~ 117093 OA

Allergy-M 1.5 0.0 Cartilage 0.8 0.0 Rep7 112347 0.5 0.0 112672 OA 8.1 7 Allergy-M Bon_e5 _ .
~
~

l F ormal 0.3 0.0 syllovium5 9'7 2.3 Lung-Normal Lung- Synovial Fluid8.2 1.9 ~ 6.7 ~

F 1 ~e11s5 .4 ..................................._...................._......................
... ..._................
........_.__........... _...... ...
_ _..................._.. _.......
12354 . .. ...._......_. ...
..
.
~
_........
.........

OA

Normal Lung-2.9 0.0 Cartilage 1.8 0.0 Rep 14 ~

_........._.........~....
..

~ 9.4 ~ 3.g 112756 OA 0 Crohns F .... .... _.................... . .
_........................ .
...............Bone9,..........................................................
.. _...._...
.-.... ._....... ............................. ..........
. _....................... ..__.
.... .

112389 Match 112757 OA

Control 1.2 0.0 6.3 . 0.0 ~ Synovium9 Crohns-F

112375 2.1 ~ 2.6 lSynovial 6.1 6.5 Fluid Crohns-F , Cells9 112732 Match ~ 117125 RA

Control 1.3 0.0 Cartilage 2.9 0.0 Crohns-F Rep2 I 12725 3.5 0.0 113492 Bone2 18 5 Crohns-M RA . .

112387 Match 113493 Control 0.7 0.0 Synovium2 9.0 2.2 ~

Crohns-M RA
~

112378 113494 Syn Crohns-M 1.3 0.0 Fluid Cells 15.5 0.0 ~ ~

~ __. ~ _. ., ~,.._._..._._.___ . ..
_.. .. _.__._.___~ ~~ _ __......
_ ..

112390 Match 1l 13499 Control 5.1 0.0 ~ 14 1 Crohns-M -Cartilage4 . .
~ RA ~
~

112726 0,6 0.0 113500 Bone4 16 0 Crohns-M, . . . _.. .... ...~'........ .
. .... ._ ... .. ._ . ...............
... ~ ..... ..... . .. . ..... .. ..
. ~. . ._ ............ _ .._ .

.
112731 Match 113501 .

6.2 0.0 ovium4 14.0 0.0 C ohns-M.......__._..._... ... ~ .. . .._~~' _.... .. . ......__ _ . ._ .........__.... ~ ..
..... ... .....

.... ._.
112380 Ulcer 113502 Syn Col-F 0.0 0.0 d Cells4 6 0.0 ~ ~ ~F 2 ..__.__.........__..__........._.._. .__ ~l ......._...__ .._ .._. __.__ ._...._._.. _ . .. .. .. .. _.. _ ..._ .._.. _ _.

_ .... ._...
I 12734 17,2 22.8 113495 16.2 .
Match ._....

Control Cartilage3 .
Ulcer RA

Col-F

_ 112384 Ulcer7.2 0 I 13496 Bone320.0 4 Col-F . RA .

112737 Match ~ I 13497 Control Ulcer3.0 0.0 Synovium3 10.4 1.6 Col-F RA _ 112386 Ulcer 113498 Syn Col-F 5.3 0.0 Cells3 3.9 ~ 8.3 ~F 1 a .. ...... . .. .... . .... .
. ._ _......... . .... ........
.. ~ ..
.....
..

112738 Match 11710 6 Control Ulcer0.7 1 0.0 ~ Normal 1.1 0 Col-F ge .

..........._.__..__.........._......................................._.....R
p2~ _.
... .....__...__....._.._................._ _....__.. "

112381 Ulcerp 0 X113663 Bone30 0 ' 0 0 0 0 Col-M....__......._..... . Normal,. . .._....
_............._........_..._...........
......_.........~.__.............___..._..........._...._ .........
_ .... .........._.......... ..............
....... ._.._....

~ . .
112735 Match 113664 ..._..._.........

Control Ulcer0.5 0.0 ;Synovium3 0.0 0.0 ~

Col-M Normal 112382 Ulcer 113665 Syn Col-M 2.6 0.0 Fluid Cells30.9 0.0 Normal 112394 Match 117107 Control Ulcer2.2 ~ 0.0 ~N~al 0.8 0.0 Col-M 7C~ilage Rep22 112383 Ulcer0.7 0 113667 Bone4I .4 0 Col-M . Normal .

112736 Match 113668 Control Ulcer1.6 0.0 Synovium4 4.4 0.0 Col-M Normal 112423 113669 Syn Psoriasis-F 16.71.9 Fluid Gells42.0 0.0 Normal Table AG.
CNS neurodegeneration v1.0 Tissue Name Rel. Tissue Name ~ Rel.
Exp.(%) Exp.(%) Ag4180, Ag4180, Run 215539679 Run AD 1 Hippo 7.1 Control (Path) 0.0 Temporal Ctx AD 2 Hippo 0.0 Control (Path) 45.4 Temporal Y Ctx AD 3 0.0 ~ I Occipital 0.0 Hippo AD 4 Hippo 0.0 ~ 2 Occipital 0.0 Ctx (Missing) AD 5 hippo 52.1 AD 3 Occipital 13.3 Ctx AD 6 Hippo 57.0 C~ 4 Occipital 15.4 Control 2 Hippo17.4 ~ 5 Occipital 33.0 Control 4 Hippo14.3 ~ 6 Occipital 40.9 _ _ __ t _ . _._ ~ _ _. __ ._ _ Control (Path) Control 1 Hippo ' Occipital Ctx .

AD 1 Temporal 16.7 Control 2 0.0 Ctx Occipital Ctx AD 2 Temporal 0.0 Control 3 19.3 Ctx occipital. Ctx,..._.... . .. ...
.. . . ..

AD 3 Temporal 0.0 Control 4 19.1 Ctx Occipital Ctx _......... ..... ..~.... ...................
.. .. ....
.
.
...........
. .. ...

AD 4 Temporal 0.0 Oocip 3 C~ 1 8 ) .4 jP

__....................it ..................
Ctac ....................
......................... ... ............
............... .
. : .. ............
_.... .........
. ....~.........

AD S Inf Temporal Control (Path) 25.0 2 0.0 ~ Occipital Ctx AD 5 SupTemporal Control (Path) 51.4 3 0.0 Occipital Ctx _ _.._ ....._...
,, . . _...................._..
_.._..__..~~InfTemworal-..........._.....
p .......
Ctx ~~~......._..........~.,..._........._......__.................

Control Path~,4....
,-__~ ..~,_.._.....-,~_,.,-~......~,~,~..
0.0 .p.
( ) 0.0 Occi ital Ctx _......
D_._.__...................._.e~._........................._..........._........
.._..._...._.._.............._.........._:........................oj____.._.._.
....__~....._....................~
._.............. .l...Parietal ................_ .. ......_. ~' 6 Sup T poral 58.6 C~ 0.0 _.... ...._.
_, . ...._..... ....._........ ...
trol l .Temporal~;,..... .C~ ~ol 2 Parietal_..........._.........
... _..... ................7 ...
..._... ~ _.
......

C~ 7.9 ..._......_.. 0.0 _.. ..... __............._........._.___........................
.._.__...._._.__...._...._...._.._.................._..
_...........__.........................__ ...._. ..._ ..._.

p 0Ø Control 3 Parietal0.0 Control 2 Tem oral _......._......._..._............._....__..........._._...........__...........
_................._.__._... .. C~......__...._._._..._...__..._.
....._. ..._.___._._...._._... _...._............._....._.._........
.. . . .........................

Control 3 Temporal Control (Path) 19 12 1 '3 _.._._.. ............ parietal Ctx _..._._... ....
.._...._..._..._........._...._....._........._................................
_....._....... _........... ..
.. .... ...._ _._.._..._......_._._ _..
_......_..........._........._..__....
.... ...__...
._._..
_... __...

Control 4 Tem ; o oral 0 Control (Path) p 0 2 C~ . Parietal....C~.....
.........._ ...... ......._. .
_... _........ . _...._..... _ ... .. .
..........................................._......._.....
......................... . ......... ..........
_.._.... . . ...... _ . ..
....
........_ ..

_ --Control (Path) ,~,_~ Control (Path) 1 __ 3 6.7 _ _ Temporal Ctx . parietal Ctx Control (Path) Control (Path) 2 0'0 4 100 Temporal Ctx Parietal Ctx .

Table AH. General screening_panel v1.4 Rel. Exp.(%)' ~ Rel. Exp.(%) ....

Tissue Name Ag4180, Tissue Name Ag4180, Run Run Adipose 5.1 Renal ca. TK-10 0.9_ Melanoma*
0.0 Bladder 0.0 Hs688(A).T

Melanoma* Gastric ca. (liver 0'4 met.) 0 Hs688(B).T NCI-N87 .

Melanoma* M14 0.0 Gastric ca. KATO III 0.3 Melanoma* 0.0 Colon ca. SW-948 1.2 LOXIMVI

Melanoma* SK- 0.0 Colon ca. SW480 3.3 MEL-5_ _ _ _. __ . _ _ Squamous cell ~ Colon ca.* (SW480 0'0 0 carcinoma SCC-4 met) SW_620 .
' Testis Pool 8.0 Colon ca. HT29 0.0 ~

Prostate ca.* 1.0 Colon ca. HCT-116 1 (bone 1 met) PC-3 .

Prostate Po_ 0.9 Colon ca. CaCo_-2 36.3 of ~y ~

Placenta 6.2 Colon cancer tissue 6.3 Uterus Pool 0.0 Colon ca. SW1116 0.0 Ovarian ca.
5.0 'Colon ca. Colo-205 0.0 Ovarian ca. SK- 1.1 Colon ca. SW-48 0 OV-3 .

Ovarian ca. 2.4 Colon Pool 3.3 Ovarian ca.
1.0 Small Intestine Pool 0 OVCAR-5 _ .. . . . ......_ .
_. _ . _ _ ...... .. ~.. . . ___ Ovarian ca. 1.1 Stomach Pool ~ 2.0 IGROV-1 _ ._ _ __ _ ..___._._...._....._._.
...

Ovarian ca. 1 Bone Marrow Pool 0.8 OVCAR-8 .

Ovary 2.1 Fetal Heart 3.9 Breast ca. MCF-70.0 Heart Pool _ _ 0.0 Breast ca. MDA- 0.0 ~Lyrnph Node Pool 1.8 MB-231 _ p ~ -~~y Breast ca. 0.9 _ 0.5 BT 549 Fetal Skeletal Muscle-~ m Breast ca. T47D 1.3 ~ Skeletal Musc_ 1e 0.0 _ _ Pool ~

Breast ca. MDA-N1.2 Spleen Pool 18.4 .

Breast Pool 1.7 Thymus Pool 3.5 Trachea 0.0 CNS cancer 100 (glio/astro) U87-MG .

g CNS cancer Lun 0.0 0.0 (glio/astro) U-118-MG

CNS cancer Fetal Lung 12.9 0.0 (neuro;met) SK-N-AS
.

Lung ca. NCI-N4170.0 ~ 9S cancer (astro) 0.0 SF-Lung ca. LX-1 0.0 CNS cancer (astro) 0.5 Lung ca. NCI-H1460,0 CNS cancer (glio) 0.5 _ SNB-19 Lung ca. SHP-770.0 29 S cancer (glio) 7.0 SF-Brain (Amygdala) Lung ca. A549 1.0 0.0 pool Lung ca. NCI-H5260.0 Brain (cerebellum) 0.0 Lung ca. NCI-H230.7 Brain (fetal) 4.5 Lung ca. NCI-H4600.0 Brain (Hippocampus)0.0 Pool Lung ca. HOP-620.0 Cerebral Cortex 0.0 Pool Lung ca. NCI-H5222.2 Brain (Substantia 1.0 nigra) Pool Liver 3.0 Brain (Thalamus) 0.0 Pool Fetal Liver 3.1 Brain (whole) 0.0 Liver ca. HepG20.0 ~ Spinal Cord Pool 0.0 Kidney Pool 5.7 Adrenal Gland 4.1 Fetal Kidney 2.8 Pituitary gland 0.0 Pool Renal ca. 786-00.0 Salivary Gland 0.6 Renal ca. A498 0.9 Thyroid (female) 7.5 Pancreatic ca.
Renal ca. ACHN 0.0 0.0 Renal ca. U0-310.8 Pancreas Pool 3.0 Table AI. General screening~anel v1.5 Rel. Exp.(%).. .._.
.,._~:.":".....,.~~,........................".~J,.......,~"...Rel.
..... ,~ Exp.(%).

Tissue Name Ag6318, Tissue Name Ag6318, Run Run 259139880 _ 259139880 ....... _..................._.__.._._....._._..- _ ...
......... ._. .........' . . . .._ . _...._ Adipose 0.0 Renal ca. TK-10 0.0 .._......_................... ......._................_.....
....._.. .. ...._._...._._.............._.............._..._._._... ..
.... ._........ _ ... ......... _....
..... . ...._ _ ...
._.._ ...._........
._.__..._._.
_.... _._._._.

Melanoma:x _,, 0 ladder .
_ .

88(A) ~. B .
Hs6 .T
~

. ...... ....._._.. _ . . ..
. ................._.._.. _~..~............
............................................._...................._. o._..
.........
........... i~'T_-~...~__._. . ........ _.. ...............
Melanoma . _ 0.
Hs688(B).T u~~ 0'0 , Gastric ca. liner _........ ~ ......_...._.._._.....met.
- .. NCI N87 ( _.._...__. _............._....._....__..........._.
................_....__.___.._.....___._...._......
........__..._._......

................ _..........
Melanoma* M14 .. . _...._._. 0.0 ... . Gastric ca. KATO
0.0 III
_ ..

Melanoma* 0 Colon ca 0 LOXIMVI . . .
_. .._. ... .. . . ...
........ _....
'.. ...

* .. ._...._._0Ø...... .... __ _........
~ ......._ Colon ca w. .... .... . ...
. . ... ..._ .. ... ... . .. . _. .. _...
Melanoma SK . . .... _. . ._...... .
~ ~ . Swq.gO ~ . ...
2.8 ' Squamous cell Colon ca.* (SW480 carcinoma SCC-4' met) SW620 .
~ .... ....._.... . .. _ ..... ................_ .. ... ........_ _. _.............. .. . ..... _ _ ..._.................._..............................
._. .... _. .............__ ......... . .._......
_........ ... ..._........_...... ....... . ....
.. _...._. ......
. . _ Testis Pool 1.0 __ _ Colon ca. HT29 0.0 y _ Prostate ca.* 0 Colon ca 0 (bone 0 HCT-116 0 ' met) PC-3 , . .

Prostate Pool 0.5 ~ Colon ca. CaCo-2 77.9 ~~

Placenta 1.7 Colon cancer tissue 0.4 Uterus Pool 0.7 Colon ca. SW1116 100.0 Ovarian ca. 0.0 Colon ca. Colo-205 0.0 Ovarian ca. SK- 1 Colon ca. SW-48 0.0 OV-3 .

Ovarian ca. 0.0 , Colon Pool 1.2 Ovarian ca. ~ 0.0 Small Intestine Pool 0.0 Ovarian ca. 0.0 Stomach Pool ~ 0.0 ~

_ Ovarian ca. 0.0 Bone Marrow Pool 0.0 Ovary _ _0.0 Fetal Heart 0.0 _ Breast ca. MCF-70.0 W Heart Pool 0.0 Breast ca. MDA- ~ 0.0 Lymph Node Pool ~ 0.0 ~

Breast ca. BT 0.0 Fetal Skeletal Muscle0.4 Breast ca. T47D 0.0 Skeletal Muscle Pool 0.0 Breast ca. MDA-N0.0 Spleen Pool 0.0 Breast Pool 0.0 Thymus Pool 0.5 Trachea 0.0 CNS cancer 2.9 (glio/astro) U87-MG

g CNS cancer Lun 0.0 0.0 (glio/astro) U-118-MG

Fetal Lung ~ I .4 CNS cancer 0.0 (neuro;met) SK-N-AS

Lung ca. NCI-N4170.0 5 9S cancer (astro) 0.0 SF-Lung ca. LX-1 O.p CNS cancer (astro) 0.0 _ . SNB-75 Lung ca. NCI-H1460.0 CNS cancer (glio) 0.0 _ SNB-19 Lung ca. SHP-77 0.0 ~ SS cancer (glio) 0.0 SF-' -Lung ca. A5~49 0.0 Brain (Amygdala) p.0 _... Pool _ . . . ....
_ _. .. _.
_ Lung ca. NCI-H5260.0 Brain (cerebellum) 0.0 Lung ca. NCI-H230.0 Brain (fetal) 0.5 Lung ca. NCI-H4600.0 Brain (Hippocampus) 0.0 Pool Lung ca. HOP-62 0.0 Cerebral Cortex Pool 1.0 Table AJ. Panel 4.1D
..~." Rel. Exp.(%) Rel. Exp.(%) Rel. Exp.(%) Tissue Name IAg4180, Run Ag6318, Run Ag6602, Run 1?3607813259196823 274219626 Secondary Th 0.0 0.0 0.0 1 act Secondary Th2 ~ 0.0 ' 0.0 0.0 act Secondary Trl 0.0 0.0 0.0 act ........................_. __ .............._....
.... .. ...... ..
.. .............................
. .
..
.
.
..

Secondary o 0. .
Thl o . ........_ rest : . _.._.
. ._ . ~.0 ...
~

.... ........................_..........._....................
Secondary Th2 .. .._................_.........................._......_..
rest ~ ....._......................Ø0 0.0 _. ._ . 0.0 . .............
... .. ... .... _........._..
..... .................
. ... _._....._ . _.
_ .

~y _. .....
Second Trl 0.0 0.0 .. _.
rest ~ ........__........ .. ....
_...... ...... .... _ 0.0 ..... _ _......... __ ...
.._ _.... .. _. _......_...
...
....

: 0.0 O.0 .
~ 1 _ ...._ .___..._ _.
act .... . O .__._ Prima . ..... _. . . ... _. ... ............
.. ............ . _ . . .0 ... ... .._ ._ .. _ __ .

Primary Th2 0.0 .. ~ 0.0 ..
act ....... 0.0 __._._._-....._.._.._._._...... .. _.__ .. ......._..._._ .. ..
.___._ Primary 0.0 . O.0_______..._.
Trl .. ........1.7 ......
act . . ._... . ....
... . ... .
... .
.

best 0.0 O.0 .
l ............ .Ø0 Primary Th .............................._.......... .._........
'........ .. ..............._._ . .. ..... ...
- .. ... .
..

Primary .. .
Th2 ~~~. ~~0 .
rest ......_.........
~.. ~.0 .. .. ... ..........
Primary T_rl 0.2 0 0.0 rest e_. .0 .. __ . -_... _ ~ ._ _ 0 CD4512A CD4 0 _ 0T-_ ~ 0 0~

lymphocyte . . .
act' _.__..
s_. _ _... ._.__ -.._.___ ._..~_.

CD45R0 CD4 p 0.0 0 lymphocyte . .
act _ _._. .. .. . _ . .
_. ... .... . . ......... .
. .. .
.
......._...
.

CD8lymphocyte O. 0 ~ 0 0.0 0 act ..... _...........

_............... ......._ Secondary CD8 . . . .. . _. _.._ ~ ... ... .... ....
p 0.0 ~ _ ..........
0 ~ _....._.

lymphocyte . ... . .
.rest ............ _.. ..
. ... ......... ...........
....._...... _..._.....
... ....
. ...
...
.

Secondary CD8 0 0.0 0 lymphocyte . .
act .. .. ~ .
. _..
.. ......

CD4lymphocyte 0.0 0.0 0.0 none try Thl/Th2/Tr1_anti-0.0 0.0 0.0 LAK cells rest4.8 ~ 3.1 0.6 LAK cells IL-20.0 0.0 0.0 LAK cells IL-2+IL-0,0 0 0 1~ ___ . .
_ _. _ . .___._ .____ LAK cells IL- 0.1 ... .
_ ._.
. 0 0 ~

2+IFN gamma . .
___ LAK cells IL-2+
0.0 0 0 IL-18 . .
- ~~
~

LA K 1 'J 0 cells 1.1 0 0 PMA/ionomycin . .

NK Cells IL-2 0.3 ' 0.0 0.0 rest __ __ . _.__.__ _ _ __._ _.. .
_..._ __ ..

Two Way MLR .
3 0.4 1 0 ~ 4 0 ~

day - . .
' ~ _....
-. .._...

Two Way ML R ~ O
0.0 0.0 p ~

day..... .... ......... .... ... .
... .. ...... . .. . .. .....
... . ..._. ....
....... ..............
. ....... .

ay Way MLR p 0 0 7 ~ 0 0 ~ 0 . . .
_.....

PBMC rest 23.7 3.l 3.3 . .. . . . ........ . . ..
........ ........._..... _ .... .

PBMC PWM 0.0 0.0 0.0 ..
. . ....
_....
~

_PBMC PHA-L 0.2 0.0 0.0 ~

Ramos (B cell) 0.0 0.0 0.0 none ~

Ramos (B cell) 0.0 ~ ...._ ~.......___ _-0.0 O.0 ionomycin _ __ . _. .
. _._........
__ _ B~phocytes 0.0 ~ 0 ~ 0 ... ...... ~~ .... .
..........~

. __....... . ....
B lymphocytes 0.0 ...... .
.. . ....
.
....

CD40L and IL-4 .

.. ..
EOL-1 dbcAMP ... 2.8 0.4 5.7 EOL-1 dbcAMP _ ~. . _ PMA/ionomycin 0.0 ~ 1.3 ~ 0 0 Dendritic cells4.1 0.6 2.3 none Dendritic cells0.3 0.0 0.0 LPS

Dendritic cells anti- a~4 0.0 0 CD40 ~ .

Monocytes rest100.0 _ 11.0 , 7.4.
. .. , _ Monocytes LPS 6.4 0.0 1.4 ~

Macrophages 0.9 0.0 0.0~
rest Macrophages 0.0 0.0 0.0 LPS

HUVEC none 0.0 0.0 0.0 HLTVEC starved0.0 0.0 0.0 HUVEC IL-lbeta 0.0 0.0 0.0 HUVEC IFN 0.0 0.0 0.7 gamma HLTVEC TNF alpha0,0 0.0 0.0 + IFN gamma HUVEC TNF alpha0,0 0.0 0 IL4 .

HUVEC IL-11 0.0 0.0 0.0 Lung Microvascular 0.0 0.0 0.0 EC

none Lung Microvascular EC 0.0 0 0 TNFalpha + IL- . .

1 beta Microvascular 0.0 0 0 Dermal EC none . .

Microsvasular Dermal EC 0.0 0 0 lpha + IL . .
~

b to ..._.............................
. ....._....................................
......._. .........
. .....

. .
Bronchia1 . . .
....

epithelium ' 0.0 0 0 NFalpha + . .

ILlbeta _...... ........._..._....._..._ .
_...._..._.............._........_.. .
_........_..........._.....

. ,~... .,~"__ ._.,~..
0.0 0 ..
Small a~ 0 ._ ~ay ......
_...
~ 0 m none . .
epitheliu . .............__......................_............ _ .. .....
__.._ ... ___........_....... ._............_...._.....__.__..._._....._ . _.._.......... _...._...._...........
........

Small airway epithelium 0.0 0 0 TNFalpha + IL- . .

1 beta Coronery artery0.0 0 0 SMC rest . .
-Coronery artery SMC TNFalpha 0.3 0.0 0.0 +

IL-1 beta Astrocytes rest0.0 0.0 0.0 Astrocytes TNFalpha + IL- 0.0 1.9 0.0 1 beta KU-812 (Basophil)0,0 0.0 0.0 rest KU-812 (Basophil)0,0 2 0 PMA/ionomycin . .

CCD1106 0.0 0.0 0.0 (Keratinocytes) none (Keratinocytes) TNFalpha + IL- . . .

1 beta Liver cirrhosis0.2 1.0 0.0 .......

NCI H292 none 0.0 0.0 o . .0 NCI-H292IL-4 0.0 0.0 0:0 NCI-H292IL-9 0.0 2.7 0.0 NCI-H292IL-13 0.2 0.0 0.0 NCI-H292IFN 0.0 0.0 0.0 gamma ~

HPAEC none 0.0 ~ 0.0 0.0 HPAEC TNF alpha0.0 ~ 1.4 0.0 + IL-1 be_ta_ p' _ Lung fibroblast 0.0 0.0 0.0 none Lung fibroblast a alpha + IL-1 0.1 ~ 0.0 ~ 0.0 ~

bet . _.... .. ...........
....... _............ . ...............
........... ............
...... ...

~ 0,0 0.0 0 Lung 0 fibroblast IL

........ .. ........... ..._..............................
. ............. _... . .. ...._.........
......_..........
...............

Lung fbroblast 0 0.0 0 . .

Lung fibroblast0 0 0 13 , . .
. ....... ..
...... ........

g 0,0 ~ 0.0 ~ 0.0 g u n abroblast IFN~

a m ....__....~_.... :......_.....
........ _~~.~~_... _..._,y,:....................
.................._u,~u~.._..~...- ..._.
Dermal fibroblast ~ 0 ~ 0 0 6 ~ 0 CCD1070 rest , . .
.
,__._:..~_.............._.............._._...__......._._......_...............
.................
..... .~ .
_ ....... ................
...... ..._ ~ ... ...._ ...

, ~ ~,~;_::,___ ~ : ~__ ~~ ~~ ~
~;
Dermal fibroblast CCD 1070 TNF 0.0 0.0 ~ 0.0 alpha....................~..................
:. ......._........_....._........... ..............._.._....._........
. ~... ..
...
............_ Dermal fibroblast0 0.0 0 CCD.1070. IL-1 , ~ ._ _....._.._.....
beta .... ......
......
. .. _ . ..

Dermal fibroblast1 0.0 0.0
9 IFN gamma _...._...... ... _... ................................_....
..... _. ._...._..__ ...
......._..

Dermal fibroblast4,5 0.0 0 IL-4 .

Dermal Fibroblasts1.2 0.0 0.0 rest Neutrophils 23 11.6 17 TNFa+LPS . .

Neutrophils 68.8 100.4 100.0 rest Table AK. General oncology screening panel v 2.4 Rel. E~p.(%) ~ Rel. Exp.(%) Tissue Name Ag4180, Run Tissue Name Ag4180, Run Colon cancer 46.0 Bladder cancer 0.0 Colon cancer 9.1 Bladder cancer 0 ~ NAT 3 0 NAT 1 .

Colon cancer 55.9 Bladder cancer 0.0 Colon cancer Adenocarcinoma 11'9 of the 0.0 ~

NAT 2 prostate 1 Colon cancer 22.4 Adenocarcinoma 4.1 3 of the prostate 2 Colon cancer Adenocarcinoma 6'7 ~ of the 0 NAT 3 prostate 3 .

Colon malignant49,7 Adenocarcinoma 1.7 of the cancer 4 prostate 4 Colon normal 0 Prostate cancer 0 0 NAT 5 ' 0 adjacent tissue. .

Lung cancer 100.0 Adenocarcinoma 0.0 1 of the prostate 6 Lung NAT 1 13.8 Adenocarcinoma 3.5 of the prostate 7 Lung cancer 11.9 Adenocarcinoma 0.0 2 ~ of the ~

prostate 8 _ Adenocarcinoma Lung NAT 2 5.7 of the 16.0 ~ ~

prostate 9 y _ .

Squamous cell Prostate cancer
10 NAT . 0 carcinoma . 10 .

Lung NAT 3 6.3 Kidney cancer 1 44.1 metastatic 32.8 KidneyNAT 1 3.9 melanoma 1 Melanoma 2 4.1 Kidney cancer 2 65.1 Melanoma 3 2.7 Kidney NAT 2 2.1 metastatic 48.3 Kidney cancer 3 0.0 melanoma 4 metastatic 51.8 Kidney NAT 3 0.0 melanoma 5 Bladder cancer 1 9.2 Kidney cancer 4 47.6 Bladder cancer 0.0 Kidney NAT 4 ~/ 33.0 Bladder cancer 2 0.0 AI comprehensive panel v1.0 Summary: Ag6318lAg6659 Two experiments with two different probe and primer sets that are specific to the NOV2B
variant produce results that are in reasonable agreement. Highest expression of this gene is seen in bone from an OA patient (CTs=30-34). Expression levels in the other samples in the Ag6659 experiment are below the threshold of reliable detection. In the experiment using probe and primer set Ag6318, low but significant levels of expression are seen in many of the samples on this panel, including bone, synovium, synovial fluid and cartilage from OA
and RA patients. These results confirm expression of this gene in samples related to the autoimmune response. Thus, therapeutic modulation of the expression or function of this gene or gene product may be useful in the treatment of OA.
CNS neurodegeneration v1.0 Summary: Ag4180 This panel does not show differential expression of this gene in Alzheimer's disease. However, this expression profile confirms the presence of this gene in the brain. This gene encodes a leucine-rich repeat protein. Leucine rich repeats (LRR) mediate reversible protein-protein interactions and have diverse cellular functions, including cellular adhesion and signaling. Several of these proteins, such as connectin, slit, chaoptin, and Toll have pivotal roles in neuronal development in Drosophila and may play significant but distinct roles in neural development and in the adult nervous system of humans (Battye R. (2001) J.
Neurosci. 21:
4290-4298. Itoh A. (1998) Brain Res. Mol. Brain Res. 62: 175-186). In Drosophilia, the LRR region of axon guidance proteins has been shown to be critical for their function (especially in axon repulsion). Since the leucine-rich-repeat protein encoded by this gene shows high expression in the cerebral cortex, it is an excellent candidate neuronal guidance protein for axons, dendrites and/or growth cones in general.
Therefore, therapeutic modulation of the levels of this protein, or possible signaling via this protein, may be of utility in enhancing/directing compensatory synaptogenesis and fiber growth in the CNS in response to neuronal death (stroke, head trauma), axon lesion (spinal cord injury), or neurodegeneration (Alzheimer's, Parkinson's, Huntington's, vascular dementia or any neurodegenerative disease). A second experiment with Ag6318 shows low/undetectable levels of expression in all samples on this panel. (CTS>35).
(Data not shown.) General screening-panel v1.4 Summary: Ag4180 Highest expression ofthis gene is seen in a brain cancer cell line (CT=30.8). Low but significant expression is also seen in colon cancer. Thus, expression of this gene could be used to differentiate between these samples and other samples on this panel and as a marker to detect the presence of these cancers. Members of the leucine rich superfamily have been shown to be upregulated in some brain cancers (Almeida A, Oncogene 1998 Jun 11;16(23):2997-3002) Therefore, therapeutic modulation of the expression or function of this gene may be effective in the treatment of brain and colon cancer.
Low but significant expression is also seen in the thyroid. The extracellular domains of receptors for glycoprotein hormones that influence the development and function of the thyroid are members of the leucine-rich repeat (LItR) protein superfamily and are responsible for the high-affinity binding. (Jiang X. (1995) Structure 3: 1341-1353.) Thus, therapeutic modulation of this gene product may aid in the treatment of metabolic and neuroendocrine disorders.
General screening-panel v1.5 Summary: Ag6318 This probe and primer set is specific for the NOV2B variant only and produces a different expression profile than in Panel 1.4. In this panel, expression is exclusive to colon cancer cell lines (CTs=32). Thus, expression of this gene could be used to differentiate between this sample and other samples on this panel and as a marker to detect the presence of colon cancer.
Furthermore, therapeutic modulation of the expression or function of this gene may be effective in the treatment of colon cancer.
General screening~anel v1.6 Summary: Ag6702 Expression is low/undetectable in all samples on this panel. (CTs>35). (Data not shown.) Panel 4.1D Summary: Ag4180 Expression of this gene is highest in resting monocytes (CT=28.7). Moderate levels of expression are seen in resting PBMCs, resting neutrophils (CT=29.2), TNF-a and LPS treated neutrophils (CT=30.7), and normal kidney.
Low but significant levels of expression are seen in activated dermal fibroblasts, resting LAK cells, LPS treated monocytes, eosinophils and treated dendritic cells.
Two experiments with the probe and primer sets Ag6318 and Ag6602, both specific to NOV2B, show expression in resting neutrophils only (CTs=31-32).
The expression of this transcript in LPS treated monocytes, cells that play a crucial role in linking innate immunity to adaptive immunity, suggests a role for this gene product in initiating inflammatory reactions. Therefore, modulation of the expression or activity of the NOV2A gene may reduce or prevent early stages of inflammation and reduce the severity of inflammatory diseases such as psoriasis, asthma, inflammatory bowel disease, rheumatoid arthritis, osteoarthritis and other lung inflammatory diseases.
General oncology screening panel v 2.4 Summary: Ag4180 Highest expression of this gene is seen in lung cancer (CT=33.5). In addition, expression is higher in lung, colon and kidney cancers when compared to expression in the corresponding normal adjacent tissue. Thus, expression of this gene could be as a marker to detect the presence of these cancers. Furthermore, therapeutic modulation of the expression or function of this gene may be effective in the treatment of lung, colon and kidney cancer.
B. NOV3A: Gonadotrophin beta-subunit Expression of gene NOV3A was assessed using the primer-probe sets Ag338 and Ag74, described in Tables BA and BB. Results of the RTQ-PCR runs are shown in Table BC.
Table BA. Probe Name Ag338 PrimersSequences ~~ Length y~Start SEQ )~
y~~~~~

_ _ _ _ _ PositionNo y Forward5'- t 23 248 124 acaacgagaccaaacaggtgact-3' ProbeT~~~-tcaagctgcccaactgtgcccc-3'~~22 272 125 _..........._..._...,_,_.._ ........_...._......._ .........__.._.......__...
_..._..... _............_..........._..._.... . ....__....._._.....
.......... _ ..... ............ .
........_ . .._....._ .
.

.
Reverse5'-ggccacgggataggtgtaga-3' 20 308 .
.
.
.__ _._..

Table BB. Probe Name Ag74 . ~-, ~ SEQ m Primers Se uences ~
q n~h _ Positi I on No . _ _._ Forward acgagaccaaacaggtgact-3 7 : ~

_ _ ___ _ _ _ . _........_........_ -_-................_........._........................_........_........._......_.
..........._.............._........._............._..................
................._...__..............._...............
TET-5'-caact t ccccgggagtcgac-3'- 2 .. .. ..........._.........
Probe ~ .......
~ 282 ~ 12 g ~4L~_ ~~

T~R _ 2 _. __ _ _ _ ~.__... .__. _.. _ . A~__. ~.. ._ ___. .
_.__._ . g.
. _._ _ . _ _. __ ReVerSe 5'-ggccacgggataggtgtaga-3' 20 308 129 Table BC. Panel 1 ~~~..........._......_ Tissue Name Rel. Exp.(%) Ag338, Rel. Exp.(%) Ag74, Run 97805375 Run 87354633 Endothelial cells 0.0 0.0 ... . .
.

0.0 _.....................
Endothelial cells (treated) _ ... . . .....Ø0 . . ......_ .. ...... .
..
. . .:.. . . .
..

Pancreas p.0 : 0.0 Pancreatic ca. CAPAN 2 0.0 ~ 0.0 -Adrenal gland 0.0 0.0 Thyroid 0.0 0.0 Salivary gland 0.0 0.0 Pituitary gland 0.0 0.0 Brain (fetal) 0.0 0.0 Brain (whole) 0.5 0.0 Brain (amygdala) 0.6 0.0 Brain (cerebellum) 0.3 24.8 Brain (hippocampus) 0.0 0.0 Brain (substantia nigra) 0.0 0.0 Brain (thalamus) 0.1 0.0 Brain (hypothalamus) 0.0 0.0 Spinal cord 0.0 0.0 glio/astro U87-MG 0.0 0.0 glio/astro U-118-MG 0.0 0.0 _.................... ............... .._................
_..........................................................................
.................~........
............ ......................................
.... ..... ........__...

astrocytoma SW1783 0.0 ~ 0.0 _.._............._._.._.........___....._..........._................_._.......
.........._..................................._._......_.......................
........... ~ .
_ ......... . ............._._....................................._.
....._......._._._.._ _......

neuro*; met..SK..N AS......._..........., 0.Ø........Ø0 . _....... ...................
_._............... .................
_ .... _..........._.................._............._................
..........

astrocytoma SF-539 0.0 ~ 0.0 ...._._........_.........._.................._...__................._..._......
......_..............._.........................................
....................._....._.............................................__....
...,~,~
- _ . . ~,._~_ .~_~
~____,._._......................_...._...._......_............_._........_.....
.......__..._....._.
. . . ..
....

yt 0.0 ~ 0.0 astroc oma SNB 75 ........_............................._ ......,............
. ...._...._.........
..
.

lioma SNB-19 0.0 .
g ...._ ..... . .
.. . .. . .. . _.. ... .. ~ 0.0 .. .. .... ..

lioma 0.0 0.0 g U251 . .. .. . ........
.. ................... ....._........ ... ..
. _..... . ..
..

glioma 0.0 0.0 SF-295 _...... _........
'................ ... . ......... ... .
.................

Hea_rt 0.0 0.0 N. ....._ .. _ .
.
....

Skeletal muscle Y p.0 _ _._ _ .._._..._...__... 0.0 .

Bone marrow 0.0 0.0 Thymus 0.0 0.0 Spleen 0.0 0.0 Lymph node 0.0 0.0 Colon (ascending) 23.2 40.3 Stomach 0.0 p.0 Small intestine ~ 0.0 ~ 0.0 Colon ca. SW480 0.0 0.0 Colon ca.* SW620 (SW480 0.0 __ 0.0 met) . ~~v Colon ca. HT29 0.0 0.0 Colon ca. HCT-116 0.0 0.0 Colon ca. CaCo-2 0.0 0.0 Colon ca. HCT-15 1.7 0.0 Colon ca. HCC-2998 0.0 0.0 Gastric ca. * (liver met)0.0 0.0 ........................._.......__....._..._____.__..___..._.._..._.._._....__ ...._.
__......__..._........_.._..._........__..... ._..._._...____ _......__ _. ___...._... ............._......__.._..._.._..._......
_._...___.._........_..
......_..._............ _.._ Bladder 0.0 _..
.....__...._ .. .... ..._....._........_ .........._0.0 . ...... ........... . .............. _ .
........ ......... _.... ._......._.. _._..._..
............_. .._.. .._. .. _.. .... ... . ...
........
.... ... ...

Trachea 0.0 O.0 _.._.._... _...__. . ._..._._........_....__._. ..
.._...__. __ _...__..... ................_.._ ._... ... .__ _ . __._... ._......._......._.........___._ __ .. __._. .. _.___.
.._ . ...... __.

Kidney _.__....... . . ._...... _._ .._.. .. _ _...... ...Ø0....Ø0., .. _ Kidney (fetal) .. .....
0.0 _ ...._ 0.0 Renal ca. 786-0 0.0 ~~ 0.0 Renal ca. A498 0.0 0.0 Renal ca. RXF 393 0.0 0.0 Renal ca. ACHN 0.0 0.0 Renal ca. U0-31 0.0 0.0 Renal ca. TK-10 0.0 0.0 Liver 0.0 0.0 Liver (fetal) 0.0 0.0 Liver ca. (hepatoblast) 0.0 0.0 HepG2 Lung 0.0 0.0 Lung (fetal) 0.0 ~ 0.0 Lung ca. (small cell) 0.0 0.0 LX-1 . ,., ............~.........._......~, "~, ~.._...__. . .. . .- , Lung ca. (small cell) 8.4 . ~.6 NCI-H69 .............................................._....
_.................._ _............... ....
.......,.................

Lun ca. s.cell var. SHP 0.0 0.0 _..................._............._......_..................._..._.............
...._.._...._................................_ g.... ( ) _ _..__................_.....
............_........__.._........._ ._....._.._..__......_............__.._.........__...._.._.........
. .
_.. ,~

Lung ca. (large cell)NCI ,~~_ ~ O
H460~ 0.0 .0 Lum................................_........~..................................
.....549............................_..........._O.4'.........................~
~~~_..........................._........................._.....................
.....
.... _........ .........Ø0 ca. non sm. cell A
g ~ ) .. . . ._......_.... ..........._..............
........ ....... _..........._............................ .. .. ...
..._.................. _......... ...........a........................._ ..
.............._.
. .. g. ..... -... _( ........__,;~ ~
............................_........
_........._....,-........................~............._....-~ .~ ~
~.0 - __~
H23 ~
Lun v s.celI
NCI
non ~a.

.
...............__......._......................................................
._........
., . ......... ... .
. .............
,......
. ... . . .. ..
.....

Lun ca. non s.cell HOP 0.0 0.0 .
..
......

g a o ( ............................
................................ ......
~ ~Ø. ................ v ..
~ . _.... ...
Lun.... Via. non s.cl . . .
.NCI H522 ..... ... ...............

Lung ca. (squam.) SW 900 0.0 , 0.0 .. ..... _....._......._...._................ ..
..........
... . ... . . .. ... .. .. .............
.. .. .. ..
.. ...........

, g ~ 0.0 Lung i ' ca. (squam.) ~-~

Mamma 0. ~
gland ~
i'3' _ _ __ ~._ .~ ' _ . _.._...._._..._.-0.0 __.._._~ 0.0 _ .__ Breast ca.* (pl.ef) MCF-7 Breast ca.* (pl.ef) MDA-MB-2310.0 0.0 Breastca.* (p1. ef) T47D 1.l l8.6 ~ ~~ -~~
a~-Breast ca. BT-549 ~ _ ~~~~ 0.0 0.0 ~ ~

Breast ca. MDA-N 0.0 0.0 Ovary 0.0 0.0 Ovarian ca. OVC_A_R-3 0.0 , 0.0 ~ ~~ ~w~~
y~~

_ Ovarian ca.OVCAR-4~ 0.0 i ~~ 0.0 Ovarian ca. OVCAR-5 10.3 ~ 4.8 ~ ~ _ ~

Ovarian ca. OVCAR-8 _0.0 0.0 ~~

Ovarian ca. IGROV-1 _ ~-~~~~~~H 0.0 1.5 "-Ovarian ca. (ascites) 0.0 0.0 Uterus 0.0 0.0 Placenta 0.0 0.0 Prostate 0.0 0.0 _............._.............._..._._........._..__._..__.._...__._._..._.......
_....._................_..._............_........__.........____......__...__._ _._..._.._.._._.......
_....._..._..._.._.__._.........._ _........__._......_._...
..._ ._...___..._......._ Prostate ca.* (bone met) 0.5 0.0 PC-3 _.._ ......_....................... ......
... ..... ........._ _...._........ .
... _..... ...
. .
. _. . _ .....

. . . ..... . ............
_ _ .. .._......_ ..._........._ . _ _ 5.8 .._......_......_..
Testis ..........._. . ...
._. __._. _...._. _.. ~ _._....__ 100.0 ..._ -. - _ ._. _. _ .._._. ....
.. _ Melanoma ,Hs688(A).T .. . ..., y O.0 . . .. p..p . . .

Melanoma* (met) Hs688(B).T0.0 0.0 with the NOV3A gene are not included. The amp plot indicates that there were experimental difficulties with this run.
Panel 1 Summary: Ag338 Highest expression of the NOV3A gene is detected in a melanoma SK-MEL-28 cell line (CT=27.8). Thus, expression of this gene may be used to distinguish this sample from other samples used in this panel. In addition, low to moderate expression of this gene is also seen in lung cancer, breast cancer and ovarian cancer cell lines. Therefore, therapeutic modulation of this gene product may be useful in the treatment of these cancers.
Low expression of this gene is also seen in testis and colon. Therefore, therapeutic modulation of this gene product may be useful in the treatment of diseases associated testis and colon such as fertility, hypogonadism, inflammatory bowel diseases, cancers.
Ag74 Highest expression of the NOV3A gene is detected in testis (CT=31.4).
Thus, expression of this gene can be used to distinguish this sample from other samples in this panel. In addition, moderate expression of this gene is also seen in colon and brain (cerebellum). Therefore, therapeutic modulation of this gene product may be useful in the treatment of neurological disorders and diseases associated with testis and colon.
Panel 4D Summary: Ag338 Expression of the NOV3A gene is lowlundetectable (CTs > 35) across all of the samples on this panel (data not shown).
General oncology screening panel v 2.4 Summary: Ag338 Expression of the NOV3A gene is low/undetectable (CTs > 35) across all of the samples on this panel (data not shown).
C. NOV4A: Odorant binding protein Expression of gene NOV4A was assessed using the primer-probe sets Ag4218 and Ag4261, described in Tables CA and CB. Results of the RTQ-PCR runs are shown in Tables GC and CD.
Table CA. Probe Name Ag4218 Sequences Length Start SEQ ID
Position No CNS neurodegeneration v1.0 Summary: Ag338 Results from one experiment Forward 5' ggaggaggacatttt-3 ~. 21 339 -130 -actctc ~

Probe T~~'-cagtcccctgtgtccctctgctg-3'- 23 394 131 Reverse 5'-cactggagatagcagacagaca-3' 22 417 132 Table CB.
Probe Name Ag4261 Primers .~ Sequences ~~ 'Length Start ~-1~ SEQ
ID

Position No Forward 5'-actctcggaggaggacatttt-3' 21 339 TET-5'-ca gtcccctgtgtccctctgctg-3'-Probe ' 134 . T~M~ _....._._......... ....
.......23........... .....

...
Reverse 5'-cactggagatagcagacagaca-3' 22 417 Table CC. v1.4 General screening~anel Rel. Exp.(%) ~ Rel.
Exp.(%) Tissue Name Ag4261, Run Tissue Ag4261, Name Run Adipose 0.0 Renal 0.0 ca.

.

Melanoma*

Hs688(A).T 0.0 Bladder 0.0 Melanoma* Gastric 0'0 ca. 2 (liver 5 met.) Hs688(B).T NCI N87 .

Melanoma* 0.0 Gastric 100.0 M14 ca.
KATO
III

Melanoma*

LOXIMVI 0.0 Colon 1.5 ca.

Melanoma*
SK-0.0 Colon 0.0 MEL-5 ca.

Squamous cell Colon ca.*
(SW480 carcinoma 0'0 met) 0.0 Testis Pool 0.0 Colon 0.0 ca.

Prostate ca.*
(bone 0.0 Colon 0.0 met) PC-3 ca.

Prostate Pool0.0 Colon 0.0 ca.
CaCo-2 Placenta 0.0 Colon 0.0 cancertissue Uterus Pool _0.0 Colon 0.0 ~~ ca.
S
W_l 116 Ovarian ca. _ p,0 Colon 0.0 OVCAR-3 ca.
Colo-205 Ovarian ca.
SK-13.4 Colon 0.0 OV-3 ca.

Ovarian ca.

O.p Colon 7.0 OVCAR-4 Pool Ovarian ca. 0.0 Small 5.7 Intestine Pool Ovarian ca. 0.0 Stomach 0.9 Pool Ovarian ca. 0.0 Bone 8.4 Marrow Pool ~

Ovary 0.0 Fetal Heart 0.4 Breast ca. MCF-7 0.0 Heart Pool 0.0 Breast ca. MDA- 0.0 Lymph Node Pool 0.0 Breast ca. BT 549 0.0 Fetal Skeletal Muscle 0.0 Breast ca. T47D 0.0 Skeletal Muscle Pool 0.0 Breast ca. MDA-N 0.0 Spleen Pool 0.0 Breast Pool 0.0 Thymus Pool 0.0 Trachea 0.0 CNS cancer 0.0 (glio/astro) U87-MG

g CNS cancer Lun 0.0 0.0 (glio/astro) U-118-MG

Fetal Lung 2.6 CNS cancer 0.0 (neuro;met) SK-N-AS

CNS cancer (astro) Lung ca. NCI-N417 0.0 SF- 0.0 Lung ca. LX-1 4.2 CNS cancer (astro) 0.0 Lung ca. NCI-H146 0.0 CNS cancer (glio) 0.0 Lung ca. SHP-77 0.0 ~9 S cancer (glio) 0.0 SF-Lung ca. A549 0.0 Brain (Amygdala) 0.0 Pool Lung ca. NCI-H526 0.0 Brain (cerebellum) 0.0 Lung ca. NCI-H23 0.0 Brain (fetal) 0.0 Brain (Hippocampus) Lung ca. NCI-H460 0.0 0.0 pool Lung ca. HOP-62 0.0 ' Cerebral Cortex Pool0.0 Lung ca. NCI-H522 0.0 Brain (Substantia 0.0 nigra) Pool Liver 0.0 . Brain (Thalamus) 0.0 Pool Fetal Liver 0.0 Brain (whole) 0.0 Liver ca. HepG2 0.0 Spinal Cord Pool 0.0 Kidney Pool 0.0 Adrenal Gland 0.0 Fetal Kidney 1.9 Pituitary gland Pool 0.0 Renal ca. 786-0 0.0 Salivary Gland 0.0 Renal ca. A498 0.0 Thyroid (female) 0.0 Pancreatic ca.
Renal ca. ACHN 0 0 . CAPAN2 .

Renal ca. U0-31 0.0 Pancreas Pool 0.0 Table CD. Panel 4.1 D

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

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

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

Claims (45)

  1. We claim:
    An isolated polypeptide comprising the mature form of an amino acid sequenced selected from the group consisting of SEQ ID N0:2n, wherein n is an integer between 1 and 54
  2. 2. An isolated polypeptide comprising an amino acid sequence selected from the group consisting of SEQ ID N0:2n, wherein n is an integer between l and 54.
  3. 3. An isolated polypeptide comprising an amino acid sequence which is at least 95%
    identical to an amino acid sequence selected from the group consisting of SEQ
    ID
    N0:2n, wherein n is an integer between 1 and 54.
  4. 4. An isolated polypeptide, wherein the polypeptide comprises an amino acid sequence comprising one or more conservative substitutions in the amino acid sequence selected from the group consisting of SEQ ID N0:2n, wherein n is an integer between 1 and 54.
  5. 5. The polypeptide of claim 1 wherein said polypeptide is naturally occuring.
  6. 6. A composition comprising the polypeptide of claim 1 and a carrier.
  7. 7. A kit comprising, in one or more containers, the composition of claim 6.
  8. 8. The use of a therapeutic in the manufacture of a medicament for treating a syndrome associated with a human disease, the disease selected from a pathology associated with the polypeptide of claim 1, wherein the therapeutic comprises the polypeptide of claim 1.
  9. 9. A method for determining the presence or amount of the polypeptide of claim 1 in a sample, the method comprising:
    (a) providing said sample;
    (b) introducing said sample to an antibody that binds immunospecifically to the polypeptide; and (c) determining the presence or amount of antibody bound to said polypeptide, thereby determining the presence or amount of polypeptide in said sample.
  10. 10. A method for determining the presence of or predisposition to a disease associated with altered levels of expression of the polypeptide of claim 1 in a first mammalian subject, the method comprising:
    a) measuring the level of expression of the polypeptide in a sample from the first mammalian subject; and b) comparing the expression of said polypeptide in the sample of step (a) to the expression of the polypeptide present in a control sample from a second mammalian subject known not to have, or not to be predisposed to, said disease, wherein an alteration in the level of expression of the polypeptide in the first subject as compared to the control sample indicates the presence of or predisposition to said disease.
  11. 11. A method of identifying an agent that binds to the polypeptide of claim 1, the method comprising:
    (a) introducing said polypeptide to said agent; and (b) determining whether said agent binds to said polypeptide.
  12. 12. The method of claim 11 wherein the agent is a cellular receptor or a downstream effector.
  13. 13. A method for identifying a potential therapeutic agent for use in treatment of a pathology, wherein the pathology is related to aberrant expression or aberrant physiological interactions of the polypeptide of claim 1, the method comprising:
    (a) providing a cell expressing the polypeptide of claim 1 and having a properly or function ascribable to the polypeptide;
    (b) contacting the cell with a composition comprising a candidate substance;
    and (c) determining whether the substance alters the property or function ascribable to the polypeptide;

    whereby, if an alteration observed in the presence of the substance is not observed when the cell is contacted with a composition in the absence of the substance, the substance is identified as a potential therapeutic agent.
  14. 14. A method for screening for a modulator of activity of or of latency or predisposition to a pathology associated with the polypeptide of claim 1, said method comprising:
    (a) administering a test compound to a test animal at increased risk for a pathology associated with the polypeptide of claim 1, wherein said test animal recombinantly expresses the polypeptide of claim 1;
    (b) measuring the activity of said polypeptide in said test animal after administering the compound of step (a); and (c) comparing the activity of said polypeptide in said test animal with the activity of said polypeptide in a control animal not administered said polypeptide, wherein a change in the activity of said polypeptide in said test animal relative to said control animal indicates the test compound is a modulator activity of or latency or predisposition to, a pathology associated with the polypeptide of claim 1 .
  15. 15. The method of claim 14, wherein said test animal is a recombinant test animal that expresses a test protein transgene or expresses said transgene under the control of a promoter at an increased level relative to a wild-type test animal, and wherein said promoter is not the native gene promoter of said transgene.
  16. 16. A method for modulating the activity of the polypeptide of claim l, the method comprising contacting a cell sample expressing the polypeptide of claim 1 with a compound that binds to said polypeptide in an amount sufficient to modulate the activity of the polypeptide.
  17. 17. A method of treating or preventing a pathology associated with the polypeptide of claim 1, the method comprising administering the polypeptide of claim 1 to a subject in which such treatment or prevention is desired in an amount sufficient to treat or prevent the pathology in the subject.
  18. 18. The method of claim 17, wherein the subject is a human.
  19. 19. A method of treating a pathological state in a mammal, the method comprising - administering to the mammal a polypeptide in an amount that is sufficient to alleviate the pathological state, wherein the polypeptide is a polypeptide having an amino acid sequence at least 95% identical to a polypeptide comprising the amino acid sequence selected from the group consisting of SEQ ID N0:2n, wherein n is an integer between 1 and 54 or a biologically active fragment thereof.
  20. 20. An isolated nucleic acid molecule comprising a nucleic acid sequence selected from the group consisting of SEQ ID N0:2n-l, wherein n is an integer between 1 and 54.
  21. 21. The nucleic acid molecule of claim 20, wherein the nucleic acid molecule is naturally occurring.
  22. 22. A nucleic acid molecule, wherein the nucleic acid molecule differs by a single nucleotide from a nucleic acid sequence selected from the group consisting of SEQ
    ID NO: 2n-1, wherein n is an integer between 1 and 54.
  23. 23. An isolated nucleic acid molecule encoding the mature form of a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID
    N0:2n, wherein n is an integer between l and 54.
  24. 24. An isolated nucleic acid molecule comprising a nucleic acid selected from the group consisting of 2n-1, wherein n is an integer between 1 and 54.
  25. 25. The nucleic acid molecule of claim 20, wherein said nucleic acid molecule hybridizes under stringent conditions to the nucleotide sequence selected from the group consisting of SEQ ID NO: 2n-1, wherein n is an integer between l and 54, or a complement of said nucleotide sequence.
  26. 26. A vector comprising the nucleic acid molecule of claim 20.
  27. 27. The vector of claim 26, further comprising a promoter operably linked to said nucleic acid molecule.
  28. 28. A cell comprising the vector of claim 26.
  29. 29. An antibody that immunospecifically binds to the polypeptide of claim 1.
  30. 30. The antibody of claim 29, wherein the antibody is a monoclonal antibody.
  31. 31. The antibody of claim 29, wherein the antibody is a humanized antibody.
  32. 32. A method for determining the presence or amount of the nucleic acid molecule of claim 20 in a sample, the method comprising:
    (a) providing said sample;
    (b) introducing said sample to a probe that binds to said nucleic acid molecule;
    and (c) determining the presence or amount of said probe bound to said nucleic acid molecule, thereby determining the presence or amount of the nucleic acid molecule in said sample.
  33. 33. The method of claim 32 wherein presence or amount of the nucleic acid molecule is used as a marker for cell or tissue type.
  34. 34. The method of claim 33 wherein the cell or tissue type is cancerous.
  35. 35. A method for determining the presence of or predisposition to a disease associated with altered levels of expression of the nucleic acid molecule of claim 20 in a first mammalian subject, the method comprising:
    a) measuring the level of expression of the nucleic acid in a sample from the first mammalian subject; and b) comparing the level of expression of said nucleic acid in the sample of step (a) to the level of expression of the nucleic acid present in a control sample from a second mammalian subject known not to have or not be predisposed to, the disease;

    wherein an alteration in the level of expression of the nucleic acid in the first subject as compared to the control sample indicates the presence of or predisposition to the disease.
  36. 36. A method of producing the polypeptide of claim 1, the method comprising culturing a cell under conditions that lead to expression of the polypeptide, wherein said cell comprises a vector comprising an isolated nucleic acid molecule comprising a nucleic acid sequence selected from the group consisting of SEQ
    ID
    NO:2n-1, wherein n is an integer between 1 and 54.
  37. 37. The method of claim 36 wherein the cell is a bacterial cell.
  38. 38. The method of claim 36 wherein the cell is an insect cell.
  39. 39. The method of claim 36 wherein the cell is a yeast cell.
  40. 40. The method of claim 36 wherein the cell is a mammalian cell.
  41. 41. A method of producing the polypeptide of claim 2, the method comprising culturing a cell under conditions that lead to expression of the polypeptide, wherein said cell comprises a vector comprising an isolated nucleic acid molecule comprising a nucleic acid sequence selected from the group consisting of SEQ
    ID
    NO:2n-1, wherein n is an integer between 1 and 54.
  42. 42. The method of claim 41 wherein the cell is a bacterial cell.
  43. 43. The method of claim 41 wherein the cell is an insect cell.
  44. 44. The method of claim 41 wherein the cell is a yeast cell.
  45. 45. The method of claim 41 wherein the cell is a mammalian cell.
CA002448073A 2001-06-04 2002-06-04 Therapeutic polypeptides, nucleic acids encoding same, and methods of use Abandoned CA2448073A1 (en)

Applications Claiming Priority (39)

Application Number Priority Date Filing Date Title
US29560701P 2001-06-04 2001-06-04
US29566101P 2001-06-04 2001-06-04
US60/295,607 2001-06-04
US60/295,661 2001-06-04
US29641801P 2001-06-06 2001-06-06
US29640401P 2001-06-06 2001-06-06
US60/296,404 2001-06-06
US60/296,418 2001-06-06
US29828501P 2001-06-14 2001-06-14
US60/298,285 2001-06-14
US29855601P 2001-06-15 2001-06-15
US60/298,556 2001-06-15
US29994901P 2001-06-21 2001-06-21
US60/299,949 2001-06-21
US30088301P 2001-06-26 2001-06-26
US60/300,883 2001-06-26
US30155001P 2001-06-28 2001-06-28
US60/301,550 2001-06-28
US31197201P 2001-08-13 2001-08-13
US60/311,972 2001-08-13
US31507101P 2001-08-27 2001-08-27
US60/315,071 2001-08-27
US31566001P 2001-08-29 2001-08-29
US60/315,660 2001-08-29
US32229301P 2001-09-14 2001-09-14
US60/322,293 2001-09-14
US32270601P 2001-09-17 2001-09-17
US60/322,706 2001-09-17
US34118601P 2001-12-14 2001-12-14
US60/341,186 2001-12-14
US36118902P 2002-02-28 2002-02-28
US60/361,189 2002-02-28
US36367302P 2002-03-12 2002-03-12
US36367602P 2002-03-12 2002-03-12
US60/363,676 2002-03-12
US60/363,673 2002-03-12
US10/162,335 2002-06-03
US10/162,335 US7034132B2 (en) 2001-06-04 2002-06-03 Therapeutic polypeptides, nucleic acids encoding same, and methods of use
PCT/US2002/017428 WO2002099116A2 (en) 2001-06-04 2002-06-04 Therapeutic polypeptides, nucleic acids encoding same, and methods of use

Publications (1)

Publication Number Publication Date
CA2448073A1 true CA2448073A1 (en) 2002-12-12

Family

ID=28047119

Family Applications (1)

Application Number Title Priority Date Filing Date
CA002448073A Abandoned CA2448073A1 (en) 2001-06-04 2002-06-04 Therapeutic polypeptides, nucleic acids encoding same, and methods of use

Country Status (3)

Country Link
EP (1) EP1401858A4 (en)
CA (1) CA2448073A1 (en)
WO (1) WO2002099116A2 (en)

Families Citing this family (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6815181B2 (en) 2001-07-09 2004-11-09 Applera Corporation Nucleic acid molecules encoding human secreted hemopexin-related proteins
SI1606409T1 (en) 2003-03-19 2011-01-31 Biogen Idec Inc Nogo receptor binding protein
EP1776136B1 (en) 2004-06-24 2012-10-03 Biogen Idec MA Inc. Treatment of conditions involving demyelination
EP2478917A1 (en) 2005-07-08 2012-07-25 Biogen Idec MA Inc. SP35 antibodies and uses thereof
ES2674710T3 (en) 2007-05-16 2018-07-03 Gene Signal International Sa Drug, medication, antitumor composition and use thereof
EP1992694A1 (en) * 2007-05-16 2008-11-19 Gene Signal International Sa Anti-tumor drug, medicament, composition, and use thereof
JP2011527572A (en) 2008-07-09 2011-11-04 バイオジェン・アイデック・エムエイ・インコーポレイテッド Composition comprising a LINGO antibody or fragment
JP2015518829A (en) 2012-05-14 2015-07-06 バイオジェン・エムエイ・インコーポレイテッドBiogen MA Inc. LINGO-2 antagonist for treatment of conditions involving motor neurons
WO2016112270A1 (en) 2015-01-08 2016-07-14 Biogen Ma Inc. Lingo-1 antagonists and uses for treatment of demyelinating disorders

Family Cites Families (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPS5951221A (en) * 1982-09-17 1984-03-24 Eisai Co Ltd Remedy for osteoarthritis deformans
US4643991A (en) * 1984-12-18 1987-02-17 The University Of Kentucky Research Foundation Peptide elastase inhibitors and methods
US5212068A (en) * 1985-04-05 1993-05-18 Sankyo Company Limited Human pancreatic elastase
KR940003653B1 (en) * 1985-04-05 1994-04-25 상꾜가부시끼가이샤 Method of producing human pancreatic elastase
JPH0673456B2 (en) * 1986-04-26 1994-09-21 三共株式会社 Human / Pancreas Elastase ▲ I ▼
US4746729A (en) * 1986-07-30 1988-05-24 Kuettner Klaus E Cartilage-derived leukocyte elastase-inhibitor
AU7835400A (en) * 1999-09-27 2001-04-30 Scios Inc. Secreted factors

Also Published As

Publication number Publication date
EP1401858A4 (en) 2005-12-21
WO2002099116A3 (en) 2003-11-20
WO2002099116A8 (en) 2003-09-25
WO2002099116A2 (en) 2002-12-12
EP1401858A2 (en) 2004-03-31

Similar Documents

Publication Publication Date Title
US20060063200A1 (en) Therapeutic polypeptides, nucleic acids encoding same, and methods of use
US20050287564A1 (en) Therapeutic polypeptides, nucleic acids encoding same, and methods of use
US20030185815A1 (en) Novel antibodies that bind to antigenic polypeptides, nucleic acids encoding the antigens, and methods of use
US20040002120A1 (en) Therapeutic polypeptides, nucleic acids encoding same, and methods of use
CA2448073A1 (en) Therapeutic polypeptides, nucleic acids encoding same, and methods of use
US20030219823A1 (en) Therapeutic polypeptides, nucleic acids encoding same, and methods of use
US20040006205A1 (en) Therapeutic polypeptides, nucleic acids encoding same, and methods of use
US20030236188A1 (en) Novel human proteins, polynucleotides encoding them and methods of using the same
CA2465239A1 (en) Therapeutic polypeptides, nucleic acids encoding same, and methods of use
US20040052806A1 (en) Proteins and nucleic acids encoding same
CA2446437A1 (en) Novel antibodies that bind to antigenic polypeptides, nucleic acids encoding the antigens, and methods of use
CA2448540A1 (en) Therapeutic polypeptides, nucleic acids encoding same, and methods of use
US20040014053A1 (en) Novel proteins and nucleic acids encoding same
US20060211031A1 (en) Novel proteins and nucleic acids encoding same
US20040018594A1 (en) Novel antibodies that bind to antigenic polypeptides, nucleic acids encoding the antigens, and methods of use
US20030212257A1 (en) Novel human proteins, polynucleotides encoding them and methods of using the same
US20040029790A1 (en) Novel human proteins, polynucleotides encoding them and methods of using the same
WO2003064589A2 (en) Therapeutic polypeptides, nucleic acids encoding same, and methods of use
CA2471480A1 (en) Therapeutic polypeptides, nucleic acids encoding same, and methods of use
US20030235821A1 (en) Novel Human proteins, polynucleotides encoding them and methods of using the same
WO2002072770A2 (en) Novel human proteins, polynucleotides encoding them and methods of using the same
EP1661998A2 (en) Antigenic polypeptides, antibodies binding thereto, nucleic acids encoding the antigens, and methods of use
US20060210559A1 (en) Novel antibodies that bind to antigenic polypeptides, nucleic acids encoding the antigens, and methods of use
US20040076967A1 (en) Novel human proteins, polynucleotides encoding them and methods of using the same
AU2002312258A1 (en) Therapeutic polypeptides, nucleic acids encoding same, and methods of use

Legal Events

Date Code Title Description
FZDE Discontinued