AU5204499A - Preparation and use of superior vaccines - Google Patents

Preparation and use of superior vaccines Download PDF

Info

Publication number
AU5204499A
AU5204499A AU52044/99A AU5204499A AU5204499A AU 5204499 A AU5204499 A AU 5204499A AU 52044/99 A AU52044/99 A AU 52044/99A AU 5204499 A AU5204499 A AU 5204499A AU 5204499 A AU5204499 A AU 5204499A
Authority
AU
Australia
Prior art keywords
mrna
polynucleotide
human
homo sapiens
protein
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
AU52044/99A
Inventor
Bruce L. Roberts
Srinivas Shankara
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.)
Genzyme Corp
Original Assignee
Genzyme Corp
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Genzyme Corp filed Critical Genzyme Corp
Publication of AU5204499A publication Critical patent/AU5204499A/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
    • C07K14/52Cytokines; Lymphokines; Interferons
    • C07K14/521Chemokines
    • C07K14/523Beta-chemokines, e.g. RANTES, I-309/TCA-3, MIP-1alpha, MIP-1beta/ACT-2/LD78/SCIF, MCP-1/MCAF, MCP-2, MCP-3, LDCF-1, LDCF-2
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/04Immunostimulants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • Medicinal Chemistry (AREA)
  • Immunology (AREA)
  • General Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • General Chemical & Material Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Veterinary Medicine (AREA)
  • Toxicology (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Zoology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Investigating Or Analysing Biological Materials (AREA)

Description

WO 99/65924 PCT/US99/13800 PREPARATION AND USE OF SUPERIOR VACCINES TECHNICAL FIELD The present invention is directed to enhanced immunotherapy of human 5 malignancies such as cancers. BACKGROUND The complex relationships between the immune system and tumor cells during the course of their pathogenesis have not been thoroughly understood. 10 However, the mere fact that a host immune system has the potential to recognize and eventually eradicate tumor cells has warranted immunotherapy as one of the most promising approaches for cancer treatment. Most tumors express altered or abnormal gene products as the result of uncontrolled cell growth and malignant transformation. These abnormal gene products are often antigenic to the host 15 immune system, rendering the tumor cells potentially susceptible to immunocytolysis. Gilboa et al. (1998) Cancer Imm. Immunother. 46:82-87. Cytotoxic T lymphocyte (CTL)-mediated cellular immunity is regarded as an important weapon for a host defense system against many tumors. A variety of molecular factors determine whether a tumor cell can be recognized by the host 20 immune system and eventually lysed by CTL. Lindauer et al. (1998) J. Mol. Med. 76:32-47. Tumor associated antigens are proteolytically degraded into small peptide epitopes that compete for binding to and presentation by a finite number of major histocompatibility complex (MHC) molecules. The formed MHC-peptide complexes can be recognized by naive CTLs via their T cell 25 receptors. Further activation of the naive CTLs requires functions of costimulatory factors, most of which are associated with professional antigen presenting cells (APCs). The activated antigen-specific CTLs then differentiate into cytolytic effector cells that are capable of lysing tumor cells bearing specific tumor antigens.
WO 99/65924 PCT/US99/13800 Various tumor associated antigens have been identified so far, with the vast majority being melonoma-related. Lindauer et al. supra. Tumor associated antigens can be categorized into four classes: the differentiation antigens which are normal proteins over-expressed by tumor cells such as gplOO; viral antigens 5 such as HPV16 E6 and E7; the cancer/testes family of antigens typified by MAGE; and the mutated proteins such as ras or p53. All these tumor antigens, when processed properly and presented favorably by MHC molecules, can be the targets for recognition and binding by T cell receptors. However, mere presentation of the tumor antigen via MHC and 10 subsequent recognition by T cell receptors are not insufficient to activate a robust cytotoxic immune response that can lyse the tumor cells. Many co- factors having immunostimulatory functions are necessary for efficient CTL activation. Indeed, binding and stimulating T cell receptors in the absence of these costimulatory factors may cause the T cells to be unresponsive to further antigenic stimulation, 15 which is an anergy state potentially responsible for immune tolerance to many tumor self antigens. Costimulatory functions have primarily been associated with professional antigen-presenting cells (APC). For example, upon exposure to specific signals such as inflammatory agents, APCs have the capacity to up-regulate T cell 20 proliferation and IL-2 production, which are necessary processes for CTL activation. APCs are also known for secreting T cell growth factors to amplify antigen-specific CTL response. In addition, activated APCs can provide a favorable lymphoid environment for antigen presentation and CTL activation, either by secreting chemokines that can induce the migration of immune effector 25 cells to antigen presenting sites; or by migrating to T cell rich sites such as draining lymph nodes where favorable APC:T cell interactions can occur. Various gene based vaccines have been used to deliver transgenes encoding tumor antigen to APCs in vivo for antigen presentation, but very few are proven effective to elicite anti-tumor immunoactivity. While these gene based 30 vaccines, including genetically modified APCs, may be efficient for antigen WO 99/65924 PCTIUS99/13800 presentation, they do not provide any modulation of the functional state of the endogenous APCs such as stimulating/facilitating the activation of T-cells. The present invention addresses this limitation and provides an enhanced vaccine composition for eliciting effective anti-tumor immune responses. 5 DISCLOSURE OF THE INVENTION This invention provides an isolated population of polynucleotides comprising or corresponding to at least one polynucleotide shown in Table 1 and their respective complements. It also provides a polynucleotide encoding a ligand 10 or antibody or engineered protein that binds to a cell surface protein of an antigen presenting cell and wherein the polynucleotide comprises or corresponds to a polynucleotide shown in Table 1 or its complement. The invention further provides a polynucleotide that encodes a transcription factor and wherein the polynucleotide comprises or corresponds to a polynucleotide shown in Table 1 or 15 its complement. Further provided herein is a polynucleotide comprising a first polynucleotide comprising encoding an immunostimulatory factor that is differentially expressed in an antigen presenting cell and comprising or corresponding to a tag shown in Table 1 or its complement. In one embodiment, 20 the first polynucleotide encodes a factor selected from the group consisting of PARC, TARC, monocyte chemoattractant protein-4 (MDP-4), MDC, escalectin, MCP-2 or a biologically active fragments thereof. The polynucleotides can further comprise a first and second promoter, wherein the first and second polynucleotides are under the transcriptional control of the first and second 25 promoters, respectively. Also provided by this invention is a polynucleotide comprising a first polynucleotide comprising encoding an immunostimulatory factor that is differentially expressed in an antigen presenting cell that is differentially expressed in an antigen presenting cell and comprising or corresponding to a tag 30 shown in Table 1 and second polynucleotide that modulates the expression of the WO 99/65924 PCT/US99/13800 4 first polynucleotide. Further provided is a polynucleotide comprising a first polynucleotide encoding an antigen and a second polynucleotide that modulates the expression of a third polynucleotide which encodes an immunstimulatory factor that is differentially expressed in an antigen presenting cell, wherein the 5 third polynucleotide comprises or corresponds to a tag shown in Table 1. The first polynucleotide may encode PARC, monocyte chemoattractant protein-4 (MDP 4), MDC, escalectin, MCP-2 or a biologically active fragments thereof. Also provided herein is a polynucleotide comprising a first polynucleotide encoding an engineered protein or polypeptide that binds to a cell surface protein of antigen 10 presenting cells thereby modulating either directly or indirectly by a signal transduction pathway and a second polynucleotide encoding an immunostimulatory factor comprising or corresponding to a tag shown in Table 1. Promoters can be operatively linked to the polynucleotides to direct expression thereof. 15 The polynucleotides can be inserted within a gene delivery vehicle or a host cell. Alternatively they can be attached to a chip or within a database for computational analysis. The compositions of this invention are useful to induce an immune response in a subject. They also are useful to modulate the genotype of an antigen 20 presenting and to screen for a candidate therapeutic agent that modulates the expression of a polynucleotide differentially expressed in an antigen. BRIEF DESCRIPTION OF THE TABLE The Table depicts a series of mRNA sequences, identified by SAGE 25 analysis. Tags isolated from various populations were isolated and analyzed: tags expressed in monocytes; tags expressed in monocyte-derived immature dendritic cells; and tags expressed in monocyte-derived mature dendritic cells, that have been stimulated to mature with TNFa. The columns of the tables are as follows: "Accession" is the accession number for the EST in the public databases; "tag" is 30 the 1Omer SAGE tag; "Seq. ID No." is the corresponding Sequence ID number WO 99/65924 PCT/US99/13800 5 for the tag found at the end of the specification and claims. The description identifies the known gene or EST that corresponds to a tag. If the description section is blank or contains "NM" that identifies a novel tag as no match was found. 5 MODE(S) FOR CARRYING OUT THE INVENTION Throughout this disclosure, various publications, patents and published patent specifications are referenced by an identifying citation. The disclosures of these publications, patents and published patent specifications are hereby incorporated by reference into the present disclosure to more fully describe the 10 state of the art to which this invention pertains. The practice of the present invention employs, unless otherwise indicated, conventional techniques of molecular biology (including recombinant techniques), microbiology, cell biology, biochemistry and immunology, which are within the skill of the art. Such techniques are explained fully in the literature. 15 These methods are described in the following publications. See, e.g. Sambrook et al. MOLECULAR CLONING: A LABORATORY MANUAL, 2 n edition (1989); CURRENT PROTOCOLS IN MOLECULAR BIOLOGY (F. M. Ausubel et al. eds. (1987)); the series METHODS IN ENZYMOLOGY (Academic Press, Inc.); "PCR: A PRACTICAL APPROACH" (M. MacPherson et al. IRL Press at Oxford University Press 20 (1991)); PCR 2: A PRACTICAL APPROACH (M.J. MacPherson. B.D. Hames and G.R. Taylor eds. (1995)); ANTIBODIES, A LABORATORY MANUAL (Harlow and Lane eds. (1988)); and ANIMAL CELL CULTURE (R.I. Freshney ed. (1987)). As used herein, certain terms may have the following defined meanings. The singular form "a", "an" and "the" include plural references unless the 25 context clearly dictates otherwise. For example, the term "a cell" includes a plurality of cells, including mixtures thereof. The term "comprising" is intended to mean that the compositions and methods include the recited elements, but not excluding others. "Consisting essentially of' when used to define compositions and methods. shall mean 30 excluding other elements of any essential significance to the combination. Thus, a r. -*I L ~ T oI WO 99/65924 6 PCT/US99/13800 composition consisting essentially of the elements as defined herein would not exclude trace contaminants from the isolation and purification method and pharmaceutically acceptable carriers, such as phosphate buffered saline. preservatives, and the like. "Consisting of' shall mean excluding more than trace 5 elements of other ingredients and substantial method steps for administering the compositions of this invention. Embodiments defined by each of these transition terms are within the scope of this invention. The terms "polynucleotide" and "nucleic acid molecule" are used interchangeably to refer to polymeric forms of nucleotides of any length. The 10 polynucleotides may contain deoxyribonucleotides, ribonucleotides, and/or their analogs. Nucleotides may have any three-dimensional structure, and may perform any function, known or unknown. The term "polynucleotide" includes, for example, single-, double-stranded and triple helical molecules, a gene or gene fragment, exons, introns, mRNA, tRNA, rRNA, ribozyn es, cDNA, recombinant 15 polynucleotides, branched polynucleotides, plasmids, vectors, isolated DNA of any sequence, isolated RNA of any sequence, nucleic acid probes, and primers. A nucleic acid molecule may also comprise modified nucleic acid molecules. A "gene" refers to a polynuclec tide containing at least one open reading frame that is capable of encoding a particular polypeptide or protein after being 20 transcribed and translated. A "gene product" refers to the amino acid (e.g. peptide or polypeptide) generated when a gene is transcribed and translated. As used herein a second polynucleotide "corresponds to" another (a first) polynucleotide if it is related to the first polynucleotide by any of the following 25 relationships: 1) The second polynucleotide comprises the first polynucleotide and the second polynucleotide encodes a gene product. 2) The second polynucleotide is 5' or 3' to the first polynucleotide in cDNA, RNA, genomic DNA, or fragment of any of these polynucleotides. For 30 example, a second polynucleotide may be a fragment of a gene that includes the WO 99/65924 PCT/US99/13800 7 first and second polynucleotides. The first and second polynucleotides are related in that they are components of the gene coding for a gene product, such as a protein or antibody. However, it is not necessary that the second polynucleotide comprises or overlaps with the first polynucleotide to be encompassed within the 5 definition of "corresponding to" as used herein. For example. the first polynucleotide may be a fragment of a 3' untranslated region of the second polynucleotide, e.g., it may comprise a promoter sequence for the gene comprising the tag. The first and second polynucleotide may be fragment of a gene coding for a gene product. The second polynucleotide may be an exon of the 10 gene while the first polynucleotide may be an intron of the gene. 3) The second polynucleotide is the complement of the first polynucleotide. A "foreign polynucleotide" is a DNA sequence that is foreign to the cell, vector or position therein, wherein it is placed. 15 A "sequence tag" or "tag" or "SAGE tag" is a short oligonucleotide containing defined nucleotide sequence that occurs in a certain position of a gene transcript. The length of a tag is generally under about 20 nucleotides, preferably between 9 to 15 nucleotides, and more preferably 10 nucleotides. The tag can be used to identify the corresponding transcript and gene from which it was 20 transcribed. A tag can further comprise exogenous nucleotide sequences to facilitate the identification and utility of the tag. Such auxiliary sequences include, but are not limited to, restriction endonuclease cleavage sites and well known primer sequences for sequencing and cloning. The term "peptide" is used in its broadest sense to refer to a compound of 25 two or more subunit amino acids, amino acid analogs, or peptidomimetics. The subunits may be linked by peptide bonds. In another embodiment, the subunit may be linked by other bonds, e.g. ester, ether, etc. As used herein the term "amino acid" refers to either natural and/or unnatural or synthetic amino acids, including glycine and both the D or L optical isomers, and amino acid analogs 30 and peptidomimetics. A peptide of three or more amino acids is commonly called WO 99/65924 PCT/US99/13800 8 an oligopeptide if the peptide chain is short. If the peptide chain is long, the peptide is commonly called a polypeptide or a protein. The term "cDNAs" refers to complementary DNA, that is mRNA molecules present in a cell or organism made in to cDNA with an enzyme such as 5 reverse transcriptase. A "cDNA library" is a collection of all of the mRNA molecules present in a cell or organism, all turned into cDNA molecules with the enzyme reverse transcriptase, then inserted into "vectors". A "probe" when used in the context of polynucleotide manipulation refers to an oligonucleotide that is provided as a reagent to detect a target potentially 10 present in a sample of interest by hybridizing with the target. Usually, a probe will comprise a label or a means by which a label can be attached, either before or subsequent to the hybridization reaction. Suitable labels include, but are not limited to radioisotopes, fluorochromes, chemiluminescent compounds, dyes, and proteins, including enzymes. 15 A "primer" is a short polynucleotide, generally with a free 3' -OH group that binds to a target or "template" potentially present in a sample of interest by hybridizing with the target, and thereafter promoting polymerization of a polynucleotide complementary to the target. A "polymerase chain reaction" ("PCR") is a reaction in which replicate copies are made of a target 20 polynucleotide using a "pair of primers" or a "set of primers" consisting of an "upstream" and a "downstream" primer, and a catalyst of polymerization, such as a DNA polymerase, and typically a thermally-stable polymerase enzyme. Methods for PCR are well known in the art, and taught, for example in "PCR: A PRACTICAL APPROACH" (M. MacPherson et al., IRL Press at Oxford 25 University Press (1991)). All processes of producing replicate copies of a polynucleotide, such as PCR or gene cloning, are collectively referred to herein as "replication." A primer can also be used as a probe in hybridization reactions, such as Southern or Northern blot analyses. Sambrook et al., supra. A "promoter" is a region on a DNA molecule to which an RNA 30 polymerase binds and initiates transcription. In an operon, the promoter is usually ------------------ f__-w itMet WO 99/65924 PCTIUS99/13800 9 located at the operator end, adjacent but external to the operator. The nucleotide sequence of the promoter determines both the nature of the enzyme that attaches to it and the rate of RNA synthesis. The term "genetically modified" means containing and/or expressing a 5 foreign gene or nucleic acid sequence which in turn, modifies the genotype or phenotype of the cell or its progeny. As used herein, "expression" or "expressed" refers to the process by which polynucleotides are transcribed into mRNA or by which transcription is enhanced. In another embodiment, the RNA is translated into peptides, 10 polypeptides, or proteins. If the polynucleotide is derived from genomic DNA, expression may include splicing of the mRNA, if an appropriate eukaryotic host is selected. "Differentially expressed" as applied to a gene, refers to the differential production of the mRNA transcribed from the gene or the protein product 15 encoded by the gene. A differentially expressed gene may be overexpressed or underexpressed as compared to the expression level of a normal or control cell. In one aspect, it refers to a differential that is 3 times, preferably 5 times, or preferably 10 times higher or lower than the expression level detected in a control sample. The term "differentially expressed" also refers to nucleotide sequences in 20 a cell or tissue which are expressed where silent in a control cell or not expressed where expressed in a control cell. A "native" or "natural" antigen is a polypeptide, protein or a fragment which contains an epitope, which has been isolated from a natural biological source, and which can specifically bind to an antigen receptor, in particular a T 25 cell antigen receptor (TCR), in a subject. It also substances which are immunogenic, i.e., immunogens, as well as substances which induce immunological unresponsiveness, or anergy, i.e., anergens. A "self-antigen" also referred to herein as a native or wild-type antigen is an antigenic peptide that induces little or no immune response in the subject due WO 99/65924 PCT/US99/13800 10 to self- tolerance to the antigen. An example of a self-antigen is the human melanoma antigen gp 100. The term "tumor associated antigen" or "TAA" refers to an antigen that is associated with or specific to a tumor. Examples of known TAAs include gplOO, 5 MART and MAGE. The term "lysing" refers to the action of rupturing the cell wall and/or cell membrane of a cell through cytotoxic T-cell lymphocyte (CTL)-mediated cellular immunity. In a preferred embodiment, the lysis of cells is done to release cellular constituents from the lysed cells. For purposes of the present invention. "cellular 10 constituents" is meant any component found within a cell. Such components include, but are not limited to, proteins, lipoproteins, glycoproteins, lipids. carbohydrates, nucleic acids, steroids, prostaglandins, and combinations and complexes thereof. The components are also referred to as "endogenous antigens." 15 A "gene delivery vehicle" is defined as any molecule that can carry inserted polynucleotides into a host cell. Examples of gene delivery vehicles are liposomes, viruses, such as baculovirus, adenovirus and retrovirus, bacteriophage, cosmid, plasmid, fungal vectors and other recombination vehicles typically used in the art which have been described for expression in a variety of eukaryotic and 20 prokaryotic hosts, and may be used for gene therapy as well as for simple protein expression. A "viral vector" is defined as a recombinantly produced virus or viral particle that comprises a polynucleotide to be delivered into a host cell, either in vivo, ex vivo or in vitro. Examples of viral vectors include retroviral vectors, 25 adenovirus vectors, adeno-associated virus vectors and the like. In aspects where gene transfer is mediated by a retroviral vector, a vector construct refers to the polynucleotide comprising the retroviral genome or part thereof, and a therapeutic gene. As used herein, "retroviral mediated gene transfer" or "retroviral transduction" carries the same meaning and refers to the process by which a gene 30 or nucleic acid sequences are stably transferred into the host cell by virtue of the SB ~ ~ i~ IN n * ~ ~ f* fie% WO 99/65924 PCT/US99/13800 11 virus entering the cell and integrating its genome into the host cell genome. The virus can enter the host cell via its normal mechanism of infection or be modified such that it binds to a different host cell surface receptor or ligand to enter the cell. As used herein, retroviral vector refers to a viral particle capable of 5 introducing exogenous nucleic acid into a cell through a viral or viral-like entry mechanism. The term "isolated" means separated from constituents, cellular and otherwise, in which the polynucleotide, peptide, polypeptide, protein, antibody, or fragments thereof, are normally associated with in nature. In addition, a 10 "concentrated", "separated" or "diluted" polynucleotide, peptide, polypeptide, protein, antibody, or fragments thereof, is distinguishable from its naturally occurring counterpart in that the concentration or number of molecules per volume is greater than "concentrated" or less than "separated" than that of its naturally occurring counterpart. Although not explicitly stated for each of the 15 inventions disclosed herein, it is to be understood that all of the above embodiments for each of the compositions disclosed below and under the appropriate conditions, are provided by this invention. Thus, a non-naturally occurring polynucleotide is provided as a separate embodiment from the isolated naturally occurring polynucleotide. A protein produced in a bacterial cell is 20 provided as a separate embodiment from the naturally occurring protein isolated from a eukaryotic cell in which it is produced in nature. "Host cell" or "recipient cell" is intended to include any individual cell or cell culture which can be or have been recipients for vectors or the incorporation of exogenous nucleic acid molecules, polynucleotides and/or proteins. It also is 25 intended to include progeny of a single cell, and the progeny may not necessarily be completely identical (in morphology or in genomic or total DNA complement) to the original parent cell due to natural, accidental, or deliberate mutation. The cells may be prokaryotic or eukaryotic, and include but are not limited to bacterial cells, yeast cells. animal cells, and mammalian cells, e.g., murine, rat, simian or 30 human. ------------------ go~ 1II e WO 99/65924 PCT/US99/13800 12 A "subject" is a vertebrate, preferably a mammal, more preferably a human. Mammals include, but are not limited to, murmes, simians, humans, farm animals, sport animals, and pets. A "control" is an alternative subject or sample used in an experiment for 5 comparison purpose. A control can be "positive" or "negative. " For example, where the purpose of the experiment is to determine a correlation of an altered expression level of a proto-oncogene with a particular type of cancer, it is generally preferable to use a positive control (a subject or a sample from a subject, carrying such alteration and exhibiting syndromes characteristic of that 10 disease), and a negative control (a subject or a sample from a subject lacking the altered expression and clinical syndrome of that disease). The terms "major histocompatibility complex" or "MHC" refers to a complex of genes encoding cell-surface molecules that are required for antigen presentation to T cells and for rapid graft rejection. The proteins encoded by the 15 MHC complex are known as "MHC molecules" and are classified into class I and class II MHC molecules. Class I MHC molecules include membrane heterodimeric proteins made up of an a chain encoded in the MHC associated noncovalently with b2-microglobulin. Class I MHC molecules are expressed by nearly all nucleated cells and have been shown to function in antigen presentation 20 to CD8- T cells. Class I molecules include HLA-A, -B, and -C in humans. Class II MHC molecules also include membrane heterodimeric proteins consisting of noncovalently associated a and b chains. Class II MHC are known to participate in antigen presentation to CD4' T cells and, in humans, include HLA-DP, -DQ, and DR. The term "MHC restriction"refers to a characteristic of T cells that 25 permits them to reorganize antigen only after it is processed and the resulting antigenic peptides are displayed in association with either a self class I or class II MHC molecule. Methods of identifying and comparing MHC are well known in the art and are described in Allen, M. et al. (1994) Human Immunol. 40:25-32; Santamaria, P. et al. (1993) Human Immunol. 37:39-50 and Hurley, C.K. et al. 30 (1997) Tissue Antigens 50:401-415.
WO 99/65924 PCT/US99/13800 The term "antigen presenting cells (APC)" refers to a class of cells capable of presenting one or more antigens in the form of antigen-MHC complex recognizable by specific effector cells of the immune system. and thereby inducing an effective cellular immune response against the antigen or antigens 5 being presented. While many types of cells may be capable of presenting antigens on their cell surface for T-cell recognition, only professional APCs have the capacity to present antigens in an efficient amount and further to activate T-cells for cytotoxic T-lymphocyte (CTL) response. APCs can be intact whole cells such as macrophages, B-cells and dendritic cells; or other molecules, naturally 10 occurring or synthetic, such as purified MHC class I molecules complexed to beta2-microglobulin. The term "dendritic cells (DC)" refers to a diverse population of morphologically similar cell types found in a variety of lymphoid and non lymphoid tissues (Steinman (1991) Ann. Rev. Immunol. 9:271-296). Dendritic 15 cells constitute the most potent and preferred APCs in the organism. At least a subset, if not all, dendritic cells are derived from bone marrow progenitor cells, circulate in small numbers in the peripheral blood and appear either as immature Langerhans' cells or terminally differentiated mature cells. While the dendritic cells can be differentiated from monocytes, they possess distinct phenotypes. For 20 example, a particular differentiating marker, CD14 antigen, is either absent or present at low levels in dendritic cells, but is possessed by monocytes. Also, dendritic cells are not phagocytic, whereas the monocytes are strongly phagocytosing cells. It has been shown that DCs provide all the signals necessary for T cell activation and proliferation. 25 "Co-stimulatory molecules" are involved in the interaction between receptor-ligand pairs expressed on the surface of antigen presenting cells and T cells. Research accumulated over the past several years has demonstrated convincingly that resting T cells require at least two signals for induction of cytokine gene expression and proliferation (Schwartz R.H. (1990) Science 30 248:1349-1356 and Jenkins M.K. (1992) Immunol. Today 13:69-73). One signal, WO 99/65924 14 PCT/US99/13800 the one that confers specificity, can be produced by interaction of the TCR/CD3 complex with an appropriate MHC/peptide complex. The second signal is not antigen specific and is termed the "co-stimulatory" signal. This signal was originally defined as an activity provided by bone-marrow-derived accessory cells 5 such as macrophages and dendritic cells, the so called "professional" APCs. Several molecules have been shown to enhance co-stimulatory activity. These are heat stable antigen (HSA) (Liu Y. et al. (1992) J. Exp. Med. 175:437-445); chondroitin sulfate-modified MHC invariant chain (ICS) (Naujokas M.F. et al. (1993) Cell 74:257-268); intracellular adhesion molecule 1 (ICAM-1) (Van 10 Seventer G.A. (1990) J. Immunol. 144:4579-4586); and B7-1 and B7-2/B70 (Schwartz R.H. (1992) Cell 71:1065-1068). Co-stimulatory molecules are commercially available from a variety of sources, including, for example, Beckman Coulter. It is intended, although not always explicitly stated, that molecules having similar biological activity as wild-type or purified co 15 stimulatory molecules (e.g., recombinantly produced or muteins thereof) are intended to be used within the spirit and scope of the invention. As used herein, the term "cytokine" refers to any one of the numerous factors that exert a variety of effects on cells, for example, inducing growth or proliferation. Non-limiting examples of cytokines which may be used alone or in 20 combination in the practice of the present invention include. interleukin-2 (IL-2), stem cell factor (SCF), interleukin 3 (IL-3), interleukin 6 (IL-6), interleukin 12 (IL-12), G-CSF, granulocyte macrophage-colony stimulating factor (GM-CSF), interleukin-1 alpha (IL-1), interleukin- 11 (IL- 11), MIP- 1, leukemia inhibitory factor (LIF), c-kit ligand, thrombopoietin (TPO) and flt3 ligand. The present 25 invention also includes culture conditions in which one or more cytokine is specifically excluded from the medium. Cytokines are commercially available from several vendors such as, for example, Genentech (South San Francisco, CA), Amgen (Thousand Oaks, CA), R&D Systems (Minneapolis, MN) and Immunex (Seattle, WA). It is intended, although not always explicitly stated, that 30 molecules having similar biological activity as wild-type or purified cytokines WO 99/65924 1, PCT/US99/13800 (e.g., recombinantly produced or muteins thereof) are intended to be used within the spirit and scope of the invention. The term "culturing" refers to the in vitro propagation of cells or organisms on or in media of various kinds. It is understood that the descendants 5 of a cell grown in culture may not be completely identical (i.e., morphologically, genetically, or phenotypically) to the parent cell. By "expanded" is meant any proliferation or division of cells. A "composition" is intended to mean a combination of active agent and another compound or composition, inert (for example, a detectable agent or label) 10 or active. such as an adjuvant. A "pharmaceutical composition" is intended to include the combination of an active agent with a carrier, inert or active, making the composition suitable for diagnostic or therapeutic use in vitro, in vivo or ex vivo. An "effective amount" is an amount sufficient to effect beneficial or 15 desired results. An effective amount can be administered in one or more administrations, applications or dosages. The terms "cancer," "neoplasm," and "tumor," used interchangeably and in either the singular or plural form, refer to cells that have undergone a malignant transformation that makes them pathological to the host organism. Primary cancer 20 cells (that is, cells obtained from near the site of malignant transformation) can be readily distinguished from non-cancerous cells by well-established techniques, particularly histological examination. The definition of a cancer cell, as used herein, includes not only a primary cancer cell, but any cell derived from a cancer cell ancestor. This includes metastasized cancer cells, and in vitro cultures and 25 cell lines derived from cancer cells. When referring to a type of cancer that normally manifests as a solid tumor, a "clinically detectable" tumor is one that is detectable on the basis of tumor mass; e.g. by such procedures as CAT scan, magnetic resonance imaging (MRI), X-ray, ultrasound or palpation. Biochemical or immunologic findings alone may be insufficient to meet this definition. Tumor 30 cells often express antigens which are tumor specific. The term "tumor associated WO 99/65924 PCT/US99/13800 16 antigen" or "TAA" refers to an antigen that is associated with or specific to a tumor. As used herein, "solid phase support" is not limited to a specific type of support. Rather a large number of supports are available and are known to one of 5 ordinary skill in the art. Solid phase supports include silica gels, resins, derivatized plastic films, glass beads, cotton, plastic beads. alumina gels. A suitable solid phase support may be selected on the basis of desired end use and suitability for various synthetic protocols. For example, for peptide synthesis, solid phase support may refer to resins such as polystyrene (e.g., PAM-resin 10 obtained from Bachem Inc.. Peninsula Laboratories, etc.), POLYHIPE 0 resin (obtained from Aminotech, Canada), polyamide resin (obtained from Peninsula Laboratories), polystyrene resin grafted with polyethylene glycol (TentaGelY, Rapp Polymere, Tubingen, Germany) or polydimethylacrylamide resin (obtained from Milligen/Biosearch, California). In a preferred embodiment for peptide 15 synthesis, solid phase support refers to polydimethylacrylamide resin. A "transgenic animal" refers to a genetically engineered animal or offspring of genetically engineered animals. The transgenic animal may contain genetic material from at least one unrelated organism (such as from a bacteria, virus, plant, or other animal) or may contain a mutation which interferes with 20 expression of a gene product. The present invention provides enhanced anti-tumor vaccines in which a polypeptide or polypeptides encoding tumor antigens are linked to a polypeptide or polypeptides encoding immunostimulatory factors associated with APC functions. The co-administration of tumor antigens and APC-associated 25 stimulatory factors will not only enable adequate antigen presentation to endogenous APCs but also enhance functions of the APCs for 1) presentation of co-stimulatory signals; 2) migration to T-cell rich sites; 3) secretion of T-cell growth factors; or 4) secretion of chemokines for recruitment of immune effector cells. 30 WO 99/65924 17 PCTIUS99/13800 The immunostimulatory factors of this invention include any polypeptide factors that modulate immune responses mediated by APC and corresponding T cells. For example, co-stimulatory factors that are differentially expressed in APCs can be used directly to boost the APC functions in vivo. Co-stimulatory 5 factors have been described above and include, but not limited to, heat stable antigen (HSA); chondroitin sulfate-modified MHC invariant chain (Ii-CS); intracellular adhesion molecule 1; and B7-1 and B7-2/B70. Also, the immunostimulatory factors of the invention can be gene regulatory factors that modulate the expression and activity of the above-described APC-associated co 10 stimulatory factors. In addition, ligands of APC-associated co-stimulatory factors can be used to create an autocrine loop whereby the genetically modified APC secrets a soluble ligand which is then available to bind cell surface receptors and activate the APC. For the purpose of this invention, polypeptides and the polynucleotides 15 encoding cell-specific antigens can be, in one embodiment, previously characterized tumor-associated antigens such as melanoma-associated antigen gp 100 (Kawakami et al. (1997) Intern. Rev. Immunol. 14:173-192); MUC-1 (Henderson et al. (1996) Cancer Res. 56:3763); MART-i (Kawakami et al. (1994) Proc. Natl. Acad. Sci. 91:3515; Ribas et al. (1997) Cancer Res. 57:2865); HER 20 2/neu (U.S. Patent No. 5,550,214); MAGE (PCT/US92/04354); HPV16, 18E6 and E7 (Ressing et al. (1996) Cancer Res. 56(1):582; Restifo (1996) Current Opinion in Immunol. 8:658; Stern (1996) Adv. Cancer Res. 69:175; Tindle et al. (1995) Clin. Exp. Immunol. 101:265; van Driel et al. (1996) Annals of Medicine 28:471); CEA (U.S. Patent No. 5,274,087); PSA (Lundwall, A. (1989) Biochem. 25 Biophys. Research Communications 161:1151); prostate specific membrane antigen (PSMA) (Israeli et al. (1993) Cancer Research 53:227); tyrosinase (U.S. Patent Nos. 5,530,096 and 4,898,814; Brichard et al. (1993) J. Exp. Med. 178:489); tyrosinase related proteins 1 or 2 (TRP-1 and TRP-2); NYESO-1 (Chen et al. (1997) Proc. Natl. Acad. Sci. U.S.A. 94:1914), or the GA733 antigen (U.S. 30 Patent No. 5,185,254).
WO 99/65924 PCT/US99/13800 18 Selection of immunostimulatory factors of the invention Embodiments of the present invention include immunostimulatorv factors that are preferentially or differentially expressed in monocyte-derived dedritic cells. Many comparative gene expression analysis can be used to identify genes 5 and mRNAs preferentially or differentially expressed in monocyte-derived dedritic cells as compared to other cells such as the monocyte precursor cells. One preferred method is the SAGE analysis--Serial Analysis of Gene Expression (Velculescu, et al. (1995) Science 270:484-487 and U.S. Patent No. 5,695,937). SAGE provides the tool by which the expressed genes and the expression 10 level of the genes of a cell at any one point in the cell cycle and under various environmental stimuli are isolated, sequenced and cataloged. SAGE provides quantitative gene expression data without the prerequisite of a hybridization probe for each transcript. SAGE is based on two principles. First, a short sequence tag (9-11 base pairs) contains sufficient information to uniquely identify 15 a transcript, provided that it is derived from a defined location within that transcript. Second, many transcript tags can be concatenated into a single molecule and then sequenced, revealing the identity of multiple tags simultaneously. The expression pattern of any population of transcripts can be quantitatively evaluated by determining the abundance of individual tags and 20 identifying the gene corresponding to each tag. Velculescu. et al. (1995) supra at 484. Isolation and characterization of macromolecules of the invention In one embodiment, the present invention provides isolation and 25 characterization of costimulatory factors preferentially or differentially expressed in APCs such as monocyte-derived dendritic cells. SAGE analysis, as described above, can be used to identify a population of sequence tags corresponding to gene transcripts that are preferably or differentially expressed in dendritic cells but not in their monocyte precursor cells. In one aspect, the transcript or gene is 30 previously identified but was heretofore unknown to be preferentially or WO 99/65924 PCT/US99/13800 19 differentially expressed in monocyte-derived dendritic cells. In another embodiment, the transcript or gene disclosed herein is "novel", which means the tag or its respective complement does not comprise sequence of or correspond to a previously identified expressed sequence tag (EST) or characterized gene. 5 As one non-exclusive example, SAGE analysis has revealed that the chemokines PARC and TARC (Pulmonary and Activation-Regulated Chemokine, Hieshima, et al. (1997) J. Imm. 159(3):1140-1149 and Thymus and Activation Regulated Chemokine, Imai, et al. (1996) J. Biol. Chem. 271(35):21514-21521) known for capability to recruit activated T cells, are differentially expressed by 10 monocyte-derived immature dendritic cells, a fact that has not been appreciated prior to the present invention. In a separate embodiment of the invention, the immunostimulatory factor as claimed can be a co-stimulatory factor that is differentially expressed in monocyte-derived DCs. The costimulatory factor used herein will include at least 15 a portion of the protein sufficient to allow binding to its costimulatory ligand expressed on corresponding T cell surface. According to an alternative embodiment of the invention, genes encoding transcription factors capable of upregulating the expression and activity of above discussed costimulatory factors are used. The encoded transcription factors can be 20 naturally occurring proteins involved in gene regulation pathways for the differentially expressed costimulatory factors in dendritic cells. Examples of transcription factors include: Nuclear Factor kappa B (NFKB) [Grohmann et al., (1998), Immunity 9(3) p. 315-323], X61498; the rel family of proteins notably relB [Wu et al., (1998) Immunity 9(6) p. 839-847]; CCAAT/enhancer binding 25 protein [Yamanaka et al., (1998) Bioorganic and Medicinal Chemistry Letters 1(1) p. 213-221] Yl 1525; inteferon-stimulated gene factor 3 (ISGF-3) [Schindler et al., 1992 P.N.A.S. USA 89(16) p. 7836-7839] M97935; STAT5 [Welte et al., (1997) European Journal of Immunology 27(10) p. 2737-2740], U43185; and NFAT-X [Masuda et al., 1995 Molecular and Cellular Biology 15(5) p. 2697 30 2706] U145 10. Alternatively, the nucleotides encoding transcription factors can be WO 99/65924 20 PCT/US99/13800 engineered via recombinant DNA technology. When included in a vaccine of the present invention, these nucleotides are capable of producing transcription factors that will transactivate the expression of the endogenous genes. Thus, according to one embodiment of the invention. the 5 immunostimulatory factor as claimed can be a transcription factor regulating the gene expression of a co-stimulatory factor differentially expressed in monocyte derived DCs. Differential gene expression analysis would also be expected to reveal genes encoding cell surface proteins that are preferentially expressed by either 10 immature or mature dendritic cells as compared to monocyte precursors. Examples of such dendritic cell surface proteins that would be targets for activating ligands or engineered binding molecules (such as antibodies) include: IFN alpha/beta receptor X89814; IL-13 receptor Y09328; CD27 ligand L08096; CDlb M28826; CD151 D29963; CD53 M60871; LFA-1 M15395; and WSX1 15 cytokine receptor AF053004. These cell surface molecules may play a pivotal role in the function of dendritic cells by acting as co-stimulatory signals or modulators of DC function or migration. Naturally occurring ligands specific for these cell surface molecules or recombinant proteins (such as an antibody) generated to have specificity for these cell surface molecules might be expected to 20 interact with the cell surface protein to stimulate the function of the dendritic cells or foster the maintenance of an activated state or stimulate the migration of dendritic cells to sites rich in T cells. Thus, the present invention relates to vaccines in which a gene or genes encoding tumor antigen or antigens is linked to a gene or genes encoding secreted proteins that have the capacity to bind to and 25 modulate the activity of cell surface proteins identified as being differentially expressed in either mature or immature dendritic cells by comparative gene expression analysis (such as SAGE). In this manner, a novel autocrine loop is established whereby a genetically modified APC produces a ligand that is secreted from the APC where it can bind to cell surface receptors on that cell and 30 stimulate the genetically modified dendritic cell.
WO 99/65924 PCT/US99/13800 21 In accordance with the present invention, the polynucleotide sequence encoding a tumor antigens and the polynucleotide sequence encoding an immunostimulatory factor can be constructed as separate molecules in the vaccine composition of the invention. Alternatively, the two nucleotide sequences can be 5 covalently linked to form a single polynucleotide construct using standard recombinant DNA technology or chemical synthesis method. A recombinant DNA construct can be designed to have the linked sequences under the control of one transcriptional control region that can mediate the expression of both the tumor antigen and the immunostimulatory factor in a vaccine composition. 10 The invention also encompasses polynucleotides which differ from that of the polynucleotides described above, but encode substantially the same amino acid sequences. These altered, but phenotypically equivalent polynucleotides are referred to as "functionally equivalent nucleic acids." As used herein, "functionally equivalent nucleic acids" encompass nucleic acids characterized by 15 slight and non-consequential sequence variations that will function in substantially the same manner to produce the same protein product(s) as the nucleic acids disclosed herein (e.g. by virtue of the degeneracy of the genetic codes), or that have conservative amino acid variations. For example, conservative variations include substitution of a non-polar residue with another 20 non-polar residue, or substitution of a charged residue with a similarly charged residue. These sequence variations include those recognized by artisans in the art as those that do not substantially alter the tertiary structure of the encoded protein. The polynucleotides of the invention can comprise additional sequences, such as additional coding sequences within the same transcription unit, 25 controlling elements such as promoters, ribosome binding sites, and polyadenylation sites, additional transcription units under control of the same or a different promoter, sequences that permit cloning, expression. and transformation of a host cell, and any such construct as may be desirable to provide embodiments of this invention. 30 WO 99/65924 PCT/US99/13800 22 Indeed, this invention also provides a promoter sequence derived from cell's genome, wherein the promoter sequence corresponds to the regulatory region of a gene that is differentially transcribed in the cell as compared to a control cell. The promoters are identified and characterized by: 1) probing a 5 cDNA library with a probe corresponding to the SAGE tag sequence or generating a portion of the desired cDNA by conducting anchored PCR using primers based on the SAGE tag sequence. Examples of cell types wherein differential expression of a gene is related to promoter function include using the partial cDNA product obtained in step one above as a probe. cloning the extreme 10 5' end of the cDNA, and also by using the 5' end of the cDNA as a probe. cloning from a genomic library the promoter of the gene that encodes the cDNA. These promoters are identified using the methods described below in combination with standard molecular techniques. Functionally equivalent sequences, as defined above, are further provided by this invention. 15 In one aspect, the promoter is a sequence derived from an APC genome, wherein the promoter region corresponds to the regulatory region of a gene that is differentially transcribed in the APC. In a further aspect, the APC is a TNF-a treated dendritic cell. In a yet further aspect, the APC is an immature dendritic cell. In a still further aspect, the expression of genes from these two cell sources 20 are compared to genes expressed in monocytes from which the dendritic cell populations were derived. The promoters identified above can be operatively linked to a foreign polynucleotide to compel differential transcription of the foreign polynucleotide in the cell from which the promoter was derived. A foreign polynucleotide is 25 intended to include any sequence which encodes in whole or in part a polypeptide or protein. It also includes sequences encoding ribozymes and antisense molecules. Foreign polynucleotides also include therapeutic genes that encode dominant inhibitory oligonucleotides and peptides as well as genes that encode 30 regulatory proteins and oligonucleotides. Generally, gene therapy will involve the WO 99/65924 PCT/US99/13800 23 transfer of a single therapeutic gene although more than one gene may be necessary for the treatment of particular diseases. In one embodiment, the therapeutic gene is a dominant inhibiting mutant of the wild-type immunosuppressive agent. Alternatively, the therapeutic gene could be a wild 5 type copy of a defective gene or a functional homolog. In one aspect, a tag identified in the Table corresponds to or comprises a polynucleotide that encodes a polypeptide or protein that is biologically active as an antigen, e.g., a native antigen, an altered antigen, a self-antigen or a tumor associated antigen. Antigens are identified by noting the overexpression or cell 10 specific expression of a tag identified herein. Using the methods described below, the gene comprising or corresponding to the tag is identified. cloned and inserted into an APC. The tag corresponds to an antigen if a CTL response is raised under appropriate experimental conditions. The peptide is confirmed immunogeneic if an appropriate immune response is elecited. 15 The invention also encompasses co-administration of an immunostimulatory factor and a foreign polynucleotide, both under the control of promoters. In one embodiment, the promoter is an APC specific promoter. In alternative embodiment, the promoters are specific to tissue identified in the Table. The immunostimulatory factors of this invention include any polypeptide 20 factors that modulate immune responses mediated by APC and corresponding T cells. For example, co-stimulatory factors that are differentially expressed in APCs can be used directly to boost the APC functions in vivo. Co-stimulatory factors have been described above. The polynucleotides of the invention can be introduced and expressed in a 25 suitable host cell for generating a cell-based vaccine. These methods are described in more detail below. The polynucleotides and seqeunces identifed above can be conjugated to a detectable market, e.g., an enzymatic label or a radioisotope for detection of nucleic acid and/or expression of the gene in a cell. A wide variety of appropriate 30 detectable markers are known in the art, including fluorescent. radioactive, WO 99/65924 PCT/US99/13800 24 enzymatic or other ligands, such as avidin/biotin. which are capable of giving a detectable signal. In preferred embodiments, one will likely desire to employ a fluorescent label or an enzyme tag, such as urease, alkaline phosphatase or peroxidase, instead of radioactive or other environmental undesirable reagents. In 5 the case of enzyme tags, colorimetric indicator substrates are known which can be employed to provide a means visible to the human eye or spectrophotometrically, to identify specific hybridization with complementary nucleic acid-containing samples. The polynucleotides and sequences embodied in this invention can be 10 obtained using chemical synthesis, recombinant cloning methods, PCR, or any combination thereof. Methods of chemical polynucleotide synthesis are well known in the art and need not be described in detail herein. One of skill in the art can use the sequence data provided herein to obtain a desired polynucleotide by employing a DNA synthesizer or ordering from a commercial service. 15 The polynucleotides and sequences of this invention can be inserted into a suitable vector, and the vector in turn can be introduced into a suitable host cell for replication and/or amplification. Polynucleotides can be introduced into host cells by any means known in the art. Cells are transformed by introducing an exogenous polynucleotide by direct uptake, endocytosis, transfection, f-mating or 20 electroporation. Once introduced, the exogenous polynucleotide can be maintained within the cell as a non-integrated vector (such as a plasmid) or integrated into the host cell genome. Amplified DNA can be isolated from the host cell by standard methods. See, e.g., Sambrook et al. (1989) supra. RNA can also be obtained from transformed host cell, or it can be obtained directly from 25 the DNA by using a DNA-dependent RNA polymerase. The present invention further encompasses a variety of gene delivery vehicles comprising the polynucleotide of the present invention. Gene delivery vehicles include both viral and non-viral vectors such as naked plasmid DNA or DNA/liposome complexes. Vectors are generally categorized into cloning and 30 expression vectors. Cloning vectors are useful for obtaining replicate copies of WO 99/65924 PCT/US99/13800 25 the polynucleotides they contain, or as a means of storing the polynucleotides in a depository for future recovery. Expression vectors (and host cells containing these expression vectors) can be used to obtain polypeptides produced from the polynucleotides they contain. Suitable cloning and expression vectors include any 5 known in the art, e.g., those for use in bacterial, mammalian. yeast and insect expression systems. The polypeptides produced in the various expression systems are also within the scope of the invention and are described above When the vectors are used for gene therapy in vivo or ex vivo, a pharmaceutically acceptable vector is preferred, such as a replication-incompetent 10 retroviral or adenoviral vector. Pharmaceutically acceptable vectors containing the nucleic acids of this invention can be further modified for transient or stable expression of the inserted polynucleotide. As used herein, the term "pharmaceutically acceptable vector" includes, but is not limited to, a vector or delivery vehicle having the ability to selectively target and introduce the nucleic 15 acid into dividing cells. An example of such a vector is a "replication incompetent" vector defined by its inability to produce viral proteins, precluding spread of the vector in the infected host cell. An example of a replication incompetent retroviral vector is LNL6 (Miller A.D. et al. (1989) BioTechniques 7:980-990). The methodology of using replication-incompetent retroviruses for 20 retroviral-mediated gene transfer of gene markers is well established (Correll et al. (1989) Proc. Natl. Acad. Sci. USA 86:8912; Bordignon (1989) Proc. Natl. Acad. Sci. USA 86:8912-52; Culver K. (1991) Proc. Natl. Acad. Sci. USA 88:3155; and Rill, D.R. (1991) Blood 79(10):2694. Clinical investigations have shown that there are few or no adverse effects associated with the viral vectors, 25 see Anderson (1992) Science 256:808-13. Compositions containing the polynucleotides and sequences of this invention, in isolated form or contained within a vector or host cell are further provided herein. When these compositions are to be used pharmaceutically, they are combined with a pharmaceutically acceptable carrier. 30 '- uE vmpIt r tj*V ifIIE WO 99/65924 PCT/US99/13800 26 A vector of this invention can contain one or more poivnucleotides comprising a sequence shown in the Table or its complement. It can also contain polynucleotide sequences encoding other polypeptides that enhance, facilitate, or modulate the desired result, such as fusion components that facilitate protein 5 purification, and sequences that increase immunogenicity of the resultant protein or polypeptide. Also embodied in the present invention are host cells transformed with the vectors as described above. Both prokaryotic and eukaryotic host cells may be used. Prokaryotic hosts include bacterial cells, for example E. coli and 10 Mycobacteria. Among eukaryotic hosts are yeast, insect, avian. plant and mammalian cells. Host systems are known in the art and need not be described in detail herein. Examples of mammalian host cells include but not limited to COS, HeLa, and CHO cells, and APCs, e.g., dendritic cells. The host cells of this invention can be used, inter alia, as repositories of 15 polynucleotides differentially expressed in a cell or as vehicles for production of the polynucleotides and the encoded polypeptides. Polypeptides of the Invention This invention provides a population of proteins or polypeptides 20 expressed from the population of polynucleotides of this invention, which is intended to include wild-type and recombinantly produced polypeptides and proteins from prokaryotic and eukaryotic host cells, as well as muteins, analogs, fusions and fragments thereof. In some embodiments, the term also includes antibodies and anti-idiotypic antibodies. 25 It is understood that equivalents or variants of the wild-type polypeptide or protein also are within the scope of this invention. An "equivalent" varies from the wild-type sequence encoded by the polynucleotides of the invention by any combination of additions, deletions, or substitutions while preserving at least one functional property of the fragment relevant to the context in which it is being 30 used. For instance, an equivalent of a polypeptide of the invention may have the WO 99/65924 PCT/US99/13800 27 ability to elicit an immune response with a similar antigen specificity as that elicited by the wild-type polypeptide. As is apparent to one skilled in the art, the equivalent may also be associated with, or conjugated with, other substances or agents to facilitate, enhance, or modulate its function. 5 The invention includes modified polypeptides containing conservative or non-conservative substitutions that do not significantly affect their properties, such as the immunogenicity of the peptides or their tertiary structures. Modification of polypeptides is routine practice in the art. Amino acid residues which can be conservatively substituted for one another include but are not 10 limited to: glycine/alanine; valine/isoleucine/leucine; asparagine/glutamine; aspartic acid/glutamic acid; serine/threonine; lysine/arginine: and phenylalanine/tyrosine. These polypeptides also include glycosylated and nonglycosylated polypeptides, as well as polypeptides with other post translational modifications, such as, for example, glycosylation with different 15 sugars, acetylation, and phosphorylation. The polypeptides of the invention can also be conjugated to a chemically functional moiety. Typically, the moiety is a label capable of producing a detectable signal. These conjugated polypeptides are useful, for example, in detection systems such as imaging of breast tumor. Such labels are known in the 20 art and include, but are not limited to, radioisotopes, enzymes, fluorescent compounds, chemiluminescent compounds, bioluminescent compounds substrate cofactors and inhibitors. See, for examples of patents teaching the use of such labels, U.S. Patent Nos. 3,817,837; 3,850,752; 3,939,350; 3,996,345; 4,277,437: 4,275,149; and 4,366,241. The moieties can be covalently linked to the 25 polypeptides, recombinantly linked, or conjugated to the polypeptides through a secondary reagent, such as a second antibody, protein A, or a biotin-avidin complex. Other functional moieties include agents that enhance immunological reactivity, agents that facilitate coupling to a solid support, vaccine carriers, 30 bioresponse modifiers, paramagnetic labels and drugs. Agents that enhance WO 99/65924 28 PCT/US99/13800 immunological reactivity include, but are not limited to, bacterial superantigens. Agents that facilitate coupling to a solid support include, but are not limited to, biotin or avidin. Immunogen carriers include, but are not limited to, any physiologically acceptable buffers. 5 The invention also encompasses fusion proteins comprising polypeptides encoded by the polynucleotides disclosed herein and fragments thereof. Such fusion may be between two or more polypeptides of the invention and a related or unrelated polypeptide. Useful fusion partners include sequences that facilitate the intracellular localization of the polypeptide, or enhance immunological reactivity 10 or the coupling of the polypeptide to an immunoassay support or a vaccine carrier. For instance, the polypeptides can be fused with a bioresponse modifier. Examples of bioresponse modifiers include, but are not limited to, fluorescent proteins such as green fluorescent protein (GFP), cytokines or lymphokines such as interleukin-2 (IL-2), interleukin 4 (IL-4), GM-CSF, and a-interferon. Another 15 useful fusion sequence is one that facilitates purification. Examples of such sequences are known in the art and include those encoding epitopes such as Myc, HA (derived from influenza virus hemagglutinin), His-6, or FLAG. Other fusion sequences that facilitate purification are derived from proteins such as glutathione S-transferase (GST), maltose-binding protein (MBP), or the Fc portion of 20 immunoglobulin. For immunological purposes, tandemly repeated polypeptide segments may be used as antigens, thereby producing highly immunogenic proteins. The proteins of this invention also can be combined with various liquid phase carriers, such as sterile or aqueous solutions, pharmaceutically acceptable 25 carriers, suspensions and emulsions. Examples of non-aqueous solvents include propyl ethylene glycol, polyethylene glycol and vegetable oils. When used to prepare antibodies, the carriers also can include an adjuvant that is useful to non specifically augment a specific immune response. A skilled artisan can easily determine whether an adjuvant is required and select one. However, for the WO 99/65924 PCTIUS99/13800 29 purpose of illustration only, suitable adjuvants include, but are not limited to Freund's Complete and Incomplete, mineral salts and polynucleotides. The proteins and polypeptides of this invention are obtainable by a number of processes well known to those of skill in the art. which include 5 purification, chemical synthesis and recombinant methods. Full-length proteins can be purified from a cell derived from non-metastatic or metastatic breast tumor tissue or tissue lysate by methods such as immunoprecipitation with antibody, and standard techniques such as gel filtration, ion-exchange, reversed-phase, and affinity chromatography using a fusion protein as shown herein. For such 10 methodology, see for example Deutscher et al. (1999) GUIDE TO PROTEIN PURIFICATION: METHODS IN ENZYMOLOGY (Vol. 182, Academic Press). Accordingly, this invention also provides the processes for obtaining these proteins and polypeptides as well as the products obtainable and obtained by these processes. 15 The proteins and polypeptides also can be obtained by chemical synthesis using a commercially available automated peptide synthesizer such as those manufactured by Perkin Elmer/Applied Biosystems, Inc., Model 430A or 431A, Foster City, CA, USA. The synthesized protein or polypeptide can be precipitated and further purified, for example by high performance liquid 20 chromatography (HPLC). Accordingly, this invention also provides a process for chemically synthesizing the proteins of this invention by providing the sequence of the protein and reagents, such as amino acids and enzymes and linking together the amino acids in the proper orientation and linear sequence. Alternatively, the proteins and polypeptides can be obtained by well 25 known recombinant methods as described, for example, in Sambrook et al. (1989), supra, using the host cell and vector systems described above. Antibodies Also provided by this invention is a population of antibodies capable of 30 specifically binding to the proteins or polypeptides as described above. The WO 99/65924 PCT/US99/13800 30 antibodies of the present invention encompass polyclonal antibodies and monoclonal antibodies. They include but are not limited to mouse, rat, and rabbit or human antibodies. This invention also encompasses functionally equivalent antibodies and fragments thereof As used herein with respect to the exemplified 5 antibodies, the phrase "functional equivalent" means an antibody or fragment thereof, or any molecule having the antigen binding site (or epitope) of the antibody that cross-blocks an exemplified antibody when used in an immunoassay such as immunoblotting or immunoprecipitation. Antibody fragments include the Fab, Fab', F(ab') 2 , and Fv regions, or 10 derivatives or combinations thereof. Fab, Fab', and F(ab'), regions of an immunoglobulin may be generated by enzymatic digestion of the monoclonal antibodies using techniques well known to those skilled in the art. Fab fragments may be generated by digesting the monoclor al antibody with papain and contacting the digest with a reducing agent to reductively cleave disulfide bonds. 15 Fab' fragments may be obtained by digesting the antibody with pepsin and reductive cleavage of the fragment so produce with a reducing agent. In the absence of reductive cleavage, enzymatic digestion of the monoclonal with pepsin produces F(ab')2 fragments. It will further be appreciated that encompassed within the definition of 20 antibody fragment is single chain antibody that can be generated as described in U.S. 4,704,692, as well as chimeric antibodies and humanized antibodies (Oi et al. (1986) BioTechniques 4(3):214). Chimeric antibodies are those in which the various domains of the antibodies' heavy and light chains are coded for by DNA from more than one species. 25 As used herein with regard to the monoclonal antibody, the "hybridoma cell line" is intended to include all derivatives, progeny cells of the parent hybridoma that produce the monoclonal antibodies specific for the polypeptides of the present invention, regardless of generation of karyotypic identity. Laboratory methods for producing polyclonal antibodies and monoclonal 30 antibodies, as well as deducing their corresponding nucleic acid sequences, are e m sm e-r--a- a-T-a- e It=-r 1T0II"1 C 03MO WO 99/65924 PCT/US99/13800 31 known in the art, see Harlow and Lane (1988) supra and Sambrook et al. (1989) supra. For production of polyclonal antibodies, an appropriate host animal is selected, typically a mouse or rabbit. The substantially purified antigen. whether the whole transmembrane domain, a fragment thereof, or a polypeptide corresponding to a segment of or the entire specific loop region within the transmembrane domain, coupled or fused to another polypeptide, is presented to the immune system of the host by methods appropriate for the host. The antigen is introduced commonly by injection into the host footpads. via intramuscular, intraperitoneal, or intraderrnal routes. Peptide fragments suitable for raising 10 antibodies may be prepared by chemical synthesis, and are commonly coupled to a carrier molecule (e.g., keyhole limpet hemocyanin) and injected into a host over a period of time suitable for the production of antibodies. Alternatively, the antigen can be generated recombinantly as a fusion protein. Examples of components for these fusion proteins include, but are not limited to myc, HA, 15 FLAG, His-6, glutathione S-transferease, maltose binding protein or the Fc portion of immunoglobulin. The monoclonal antibodies of this invention refer to antibody compositions having a homogeneous antibody population. It is not intended to be limited as regards to the source of the antibody or the manner in which it is made. 20 Generally, monoclonal antibodies are biologically produced by introducing protein or a fragment thereof into a suitable host, e.g., a mouse. After the appropriate period of time, the spleens of such animal is excised and individual spleen cells fused, typically, to immortalized myeloma cells under appropriate selection conditions. Thereafter the cells are clonally separated and the 25 supernatants of each clone are tested for their production of an appropriate antibody specific for the desired region of the antigen using methods well known in the art. The isolation of other hybridomas secreting monoclonal antibodies with the specificity of the monoclonal antibodies of the invention can also be 30 accomplished by one of ordinary skill in the art by producing anti-idiotypic WO 99/65924 PCT/US99/13800 antibodies (Herlyn et al. (1986) Science 232:100). An anti-idiotypic antibody is an antibody which recognizes unique determinants present on the monoclonal antibody produced by the hybridoma of interest. Idiotypic identity between monoclonal antibodies of two hybridomas 5 demonstrates that the two monoclonal antibodies are the same with respect to their recognition of the same epitopic determinant. Thus, by using antibodies to the epitopic determinants on a monoclonal antibody it is possible to identify other hybridomas expressing monoclonal antibodies of the same epitopic specificity. It is also possible to use the anti idiotype technology to produce 10 monoclonal antibodies which mimic an epitope. For example, an anti-idiotypic monoclonal antibody made to a first monoclonal antibody will have a binding domain in the hypervariable region which is the mirror image of the epitope bound by the first monoclonal antibody. Thus, in this instance, the anti-idiotypic monoclonal antibody could be used for immunization for production of these 15 antibodies. Other suitable techniques of antibody production include, but are not limited to, in vitro exposure of lymphocytes to the antigenic polypeptides or selection of libraries of antibodies in phage or similar vectors. See Huse et al. (1989) Science 246:1275-128 1. Genetically engineered variants of the antibody 20 can be produced by obtaining a polynucleotide encoding the antibody, and applying the general methods of molecular biology to introduce mutations and translate the variant. The above described antibody "derivatives" are further provided herein. Sera harvested from the immunized animals provide a source of 25 polyclonal antibodies. Detailed procedures for purifying specific antibody activity from a source material are known within the art. Undesired activity cross-reacting with other antigens, if present, can be removed, for example, by running the preparation over adsorbants made of those antigens attached to a solid phase and eluting or releasing the desired antibodies off the antigens. If desired, the specific 30 antibody activity can be further purified by such techniques as protein A WO 99/65924 PCT/US99/13800 chromatography, ammonium sulfate precipitation, ion exchange chromatography, high-performance liquid chromatography and immunoaffinity chromatography on a column of the immunizing polypeptide coupled to a solid support. The specificity of an antibody refers to the ability of the antibody to 5 distinguish polypeptides comprising the immunizing epitope from other polypeptides. An ordinary skill in the art can readily determine without undue experimentation whether an antibody shares the same specificity as a antibody of this invention by determining whether the antibody being tested prevents an antibody of this invention from binding the polypeptide(s) with which the 10 antibody is normally reactive. If the antibody being tested competes with the antibody of the invention as shown by a decrease in binding by the antibody of this invention, then it is likely that the two antibodies bind to the same or a closely related epitope. Alternatively, one can pre-incubate the antibody of this invention with the polypeptide(s) with which it is normally reactive, and 15 determine if the antibody being tested is inhibited in its ability to bind the antigen. If the antibody being tested is inhibited, then, in all likelihood, it has the same, or a closely related, epitopic specificity as the antibody of this invention. The antibodies of the invention can be bound to many different carriers. Thus, this invention also provides compositions containing antibodies and a 20 carrier. Carriers can be active and/or inert. Examples of well-known carriers include polypropylene, polystyrene, polyethylene, dextran, nylon, amylases, glass, natural and modified celluloses, polyacrylamides, agaroses and magnetite. The nature of the carrier can be either soluble or insoluble for purposes of the invention. Those skilled in the art will know of other suitable carriers for binding 25 antibodies, or will be able to ascertain such, using routine experimentation. The antibodies of this invention can also be conjugated to a detectable agent or a hapten. The complex is useful to detect the polypeptide(s) (or polypeptide fragments) to which the antibody specifically binds in a sample, using standard immunochemical techniques such as immunohistochemistry as 30 described by Harlow and Lane (1988) supra. There are many different labels and WO 99/65924 PCT/US99/13800 34 methods of labeling known to those of ordinary skill in the art. Examples of the types of labels which can be used in the present invention include radioisotopes, enzymes, colloidal metals, fluorescent compounds, bioluminescent compounds, and chemiluminescent compounds. Those of ordinary skill in the art will know of 5 other suitable labels for binding to the antibody, or will be able to ascertain such, using routine experimentation. Furthermore, the binding of these labels to the antibody of the invention can be done using standard techniques common to those of ordinary skill in the art. Another technique which may also result in greater sensitivity consists of 10 coupling the antibodies to low molecular weight haptens. These haptens can then be specifically detected by means of a second reaction. For example, it is common to use such haptens as biotin, which reacts avidin, or dinitropherryl, pyridoxal, and fluorescein, which can react with specific anti-hapten antibodies. See Harlow and Lane (1988) supra. 15 Compositions containing the antibodies, fragments thereof or cell lines which produce the antibodies, are encompassed by this invention. When these compositions are to be used pharmaceutically, they are combined with a pharmaceutically acceptable carrier. 20 Uses of Polynucleotides, Polypeptides and Antibodies of the Invention The polynucleotides, polypeptides and antibodies embodied in this invention provide specific reagents that can be used in standard diagnostic procedures. Accordingly, one embodiment of the present invention is a method of characterizing a cell of monocyte lineage by detecting differential expression of a 25 polynucleotide comprising any one of the sequences shown in the Table or any one of the disclosed populations, or the encoded polypeptides. In assaying for an alteration in mRNA level, nucleic acid contained in the aforementioned a sample suspected of containing a dendritic cell is first extracted according to standard methods in the art. For instance, mRNA can be isolated 30 using various lytic enzymes or chemical solutions according to the procedures set WO 99/65924 PCT/US99/13800 35 forth in Sambrook et al. (1989), supra or extracted by nucleic-acid-binding resins following the accompanying instructions provided by manufactures. The mRNA contained in the extracted nucleic acid sample is then detected by hybridization (e.g. Northern blot analysis) and/or amplification procedures according to 5 methods widely known in the art or based on the methods exemplified herein. Nucleic acid molecules having at least 10 nucleotides and exhibiting sequence complementarity or homology to the polynucleotides described herein find utility as hybridization probes. It is known in the art that a "perfectly matched" probe is not needed for a specific hybridization. Minor changes in probe 10 sequence achieved by substitution, deletion or insertion of a small number of bases do not affect the hybridization specificity. In general, as much as 20% base pair mismatch (when optimally aligned) can be tolerated. Preferably, a probe useful for detecting the aforementioned mRNA that is differentially expressed in the cell type for which one is probing. These tags are identified in Table 1, below. 15 More preferably, the probe is at least 80%, or 85% identical to the corresponding gene sequence after alignment of the homologous region; even more preferably, it exhibits 90% identity. These probes can be used in hybridization reaction (e.g. Southern and Northern blot analysis) to detect, prognose, diagnose or monitor the physiological 20 states associated with the differential expression of these genes. The total size of fragment, as well as the size of the complementary stretches, will depend on the intended use or application of the particular nucleic acid segment. Smaller fragments derived from the known sequences will generally find use in hybridization embodiments, wherein the length of the complementary region may 25 be varied, such as between about 10 and about 100 nucleotides, or even full length according to the complementary sequences one wishes to detect. Nucleotide probes having complementary sequences over stretches greater than 10 nucleotides in length are generally preferred, so as to increase stability and selectivity of the hybrid, and thereby improving the specificity of particular 30 hybrid molecules obtained. More preferably, one can design nucleic acid WO 99/65924 PCT/US99/13800 36 molecules having gene-complementary stretches of more than 50 nucleotides in length, or even longer where desired. Such fragments may be readily prepared by, for example, directly synthesizing the fragment by chemical means, by application of nucleic acidreproduction technology, such as the PCRTM 5 technology with two priming oligonucleotides as described in U.S. Pat. No. 4,603,102 or by introducing selected sequences into recombinant vectors for recombinant production. A preferred probe is about 50-75 or more preferably, 50 100, nucleotides in length. In certain embodiments, it will be advantageous to employ nucleic acid 10 sequences of the present invention in combination with an appropriate means, such as a label, for detecting hybridization and therefore complementary sequences. A wide variety of appropriate indicator means are known in the art, including fluorescent, radioactive, enzymatic or other ligands, such as avidin/biotin, which are capable of giving a detectable signal. In preferred 15 embodiments, one will likely desire to employ a fluorescent label or an enzyme tag, such as urease, alkaline phosphatase or peroxidase, instead of radioactive or other environmental undesirable reagents. In the case of enzyme tags, colorimetric indicator substrates are known which can be employed to provide a means visible to the human eye or spectrophotometrically, to identify specific 20 hybridization with complementary nucleic acid-containing samples. The nucleotide probes of the present invention can also be used as primers and detection of genes or gene transcripts that are differentially expressed in certain body tissues. A preferred primer is one comprising a sequence of shown in the Table or their respective complements. Additionally, a primer useful for 25 detecting the aforementioned gene or transcript is at least about 80% identical to the homologous region of comparable size of the gene or transcript to be detected contained in the previously identified sequences. For the purpose of this invention, amplification means any method employing a primer-dependent polymerase capable of replicating a target sequence with reasonable fidelity. 30 Amplification may be carried out by natural or recombinant DNA-polymerases E.'~ ~ ~ ~~i crqr o LZr II WO 99/65924 PCTIUS99/13800 37 such as T7 DNA polymerase, Klenow fragment of E.coli DNA polymerase, and reverse transcriptase. A preferred amplification method is PCR. General procedures for PCR are taught in MacPherson et al., PCR: A PRACTICAL APPROACH, (IRL Press at 5 Oxford University Press (1991)). However, PCR conditions used for each application reaction are empirically determined. A number of parameters influence the success of a reaction. Among them are annealing temperature and time, extension time, Mg 2 ' ATP concentration, pH, and the relative concentration of primers, templates, and deoxyribonucleotides. 10 After amplification, the resulting DNA fragments can be detected by agarose gel electrophoresis followed by visualization with ethidium bromide staining and ultraviolet illumination. A specific amplification of the gene or transcript of interest can be verified by demonstrating that the amplified DNA fragment has the predicted size, exhibits the predicated restriction digestion 15 pattern, and/or hybridizes to the correct cloned DNA sequence. The probes and tags of this invention also can be attached to a solid support for use in high throughput screening assays using methods known in the art. PCT WO 97/10365 and U.S. Patent numbers 5,405,783, 5,412,087 and 5,445,934, for example, disclose the construction of high density oligonucleotide 20 chips which can contain one or more of the sequences disclosed herein. Based in the methods disclosed in U.S. Patent numbers 5,405,783, 5,412,087 and 5,445,934, the probes of this invention are synthesized on a derivatized glass surface. Photoprotected nucleoside phosphoramidites are coupled to the glass surface, selectively deprotected by photolysis through a photolithographic mask, 25 and reacted with a second protected nucleoside phosphoramidite. The coupling/deprotection process is repeated until the desired probe is complete. The expression level of a gene of interest is determined through exposure of a nucleic acid sample to the probe-modified chip. Extracted nucleic acid is labeled, for example, with a fluorescent tag, preferably during an amplification 30 step. Hybridization of the labeled sample is performed at an appropriate es us-e-- -v euccT ti ii =: jn WO 99/65924 PCTIUS99/13800 38 stringency level. The degree of probe-nucleic acid hybridization is quantitatively measured using a detection device, such as a confocal microscope. See U.S. Pat Nos. 5,578,832 and 5,631,734. The obtained measurement is directly correlated with gene expression level. 5 More specifically, the probes and high density oligonucleotide probe arrays provide an effective means of monitoring expression of a multiplicity of genes. The expression monitoring methods of this invention may be used in a wide variety of circumstances including detection of disease, identification of differential gene expression between two samples, or screening for compositions 10 that upregulate or downregulate the expression of particular genes. In another preferred embodiment, the methods of this invention are used to monitor expression of the genes which specifically hybridize to the probes of this invention in response to defined stimuli, such as a drug. In one embodiment, the hybridized nucleic acids are detected by detecting 15 one or more labels attached to the sample nucleic acids. The labels may be incorporated by any of a number of means well known to those of skill in the art. However, in one aspect, the label is simultaneously incorporated during the amplification step in the preparation of the sample nucleic acid. Thus, for example, polymerase chain reaction (PCR) with labeled primers or labeled 20 nucleotides will provide a labeled amplification product. In a separate embodiment, transcription amplification, as described above, using a labeled nucleotide (e.g. fluorescein-labeled UTP and/or CTP) incorporates a label in to the transcribed nucleic acids. Alternatively, a label may be added directly to the original nucleic acid 25 sample (e.g., mRNA, polyA, mRNA, cDNA, etc.) or to the amplification product after the amplification is completed. Means of attaching labels to nucleic acids are well known to those of skill in the art and include, for example nick translation or end-labeling (e.g. with a labeled RNA) by kinasing of the nucleic acid and subsequent attachment (ligation) of a nucleic acid linker joining the sample 30 nucleic acid to a label (e.g., a fluorophore). ~.rtg -r-r aUET OLI II WO 99/65924 PCT/US99/13800 39 The nucleic acid sample also may be modified prior to hybridization to the high density probe array in order to reduce sample complexity thereby decreasing background signal and improving sensitivity of the measurement using the methods disclosed in WO 97/10365. 5 Results from the chip assay are typically analyzed using a computer software program. See, for example, EP 0717 113 A2 and WO 95/20681. The hybridization data are read into the program, which calculates the expression level of the targeted gene(s). This figure is compared against existing data sets of gene expression levels for various cell types. 10 Expression of the genes associated characteristic of dendritic cells as compared to monocytes can also be determined by examining the protein product of the polynucleotides of the present invention. Determining the protein level involves a) providing a biological sample containing polypeptides; and (b) measuring the amount of any immunospecific binding that occurs between an 15 antibody reactive to the protein products of interest and a component in the sample, in which the amount of immunospecific binding indicates the level of the protein products. A variety of techniques are available in the art for protein analysis. They include but are not limited to radioimmunoassays, ELISA (enzyme linked 20 immunoradiometric assays), "sandwich" immunoassays, immunoradiometric assays, in situ immunoassays (using e.g., colloidal gold, enzyme or radioisotope labels), western blot analysis, immunoprecipitation assays, immunofluorescent assays, and SDS-PAGE. In addition, cell sorting analysis can be employed to detect cell surface antigens. Such analysis involves labeling target cells with 25 antibodies coupled to a detectable agent, and then separating the labeled cells from the unlabeled ones in a cell sorter. A sophisticated cell separation method is fluorescence-activated cell sorting (FACS). Cells traveling in single file in a fine stream are passed through a laser beam, and the fluorescence of each cell bound by the fluorescently labeled antibodies is then measured. 30 WO 99/65924 PCT/US99/13800 40 Antibodies that specifically recognize and bind to the protein products of interest are required for conducting the aforementioned protein analyses. These antibodies may be purchased from commercial vendors or generated and screened using methods well known in the art. See Harlow and Lane (1988) supra. and 5 Sambrook et al. (1989) supra. There are various methods available in the art for quantifying mRNA or protein level from a cell sample and indeed, any method that can quantify these levels is encompassed by this invention. For example, determination of the mRNA level of the gene may involve, in one aspect, measuring the amount of 10 mRNA in a mRNA sample isolated from the cell by hybridization or quantitative amplification using at least one oligonucleotide probe that is complementary to the mRNA. Determination of the aforementioned protein products requires measuring the amount of immunospecific binding that occurs between an antibody reactive to the product of interest. To detect and quantify the 15 immunospecific binding, or signals generated during hybridization or amplification procedures, digital image analysis systems including but not limited to those that detect radioactivity of the probes or chemiluminescence can be employed. The promoter sequences of this invention are useful for targeted 20 expression of foreign polynucleotides. The promoters can be operatively linked to foreign polynucleotides and administered to patients alone or after transduction into host cells. Because the promoters have been selected for cell-specific expression, after incorporation into host cells, either in vivo or ex vivo, targeted expression of the inserted polynucleotide can be obtained. 25 In one embodiment, the promoters preferentially express operatively linked polynucleotides in APCs, e.g., tumor associated antigens. Genes coding for immunostimulatory molecules such as cytokines or co-stimulatory molecules can be linked to the promoter sequence and gene coding for the antigen. Administration of these polynucleotides to a subject, alone or transduced into host WO 99/65924 PCT/US99/13800 41 APCs, are useful to induce an immune response by educating immune effector cells in vivo. Screening Assays 5 The present invention also provides a screen for various agents which modulate the expression of a polynucleotide associated the phenotype of a normal or pathological cell by first contacting a suitable cell with an effective amount of a potential agent, and then assaying for a change in the expression level of a polynucleotide selected from the group consisting of SEQ ID NOS: 1-207 or any 10 one of the populations from which the population was derived. For example, one would assay for a change in the expression level of any one of the polynucleotides comprising or corresponding to the sequence shown in the table in a monocyte or dendritic cell. A change in the expression level is indicative of a candidate modulating agent. In certain aspects of the invention, an agent may 15 result in phenotypic changes of the recipient cell as evidenced by an agent-induced cell apoptosis, a reduced rate of cell growth or cell motility. Altered gene expression can be detected by assaying for altered mRNA expression or protein expression using the probes, primers and antibodies as described herein. 20 To practice the method in vitro, cell cultures or tissue cultures previously identified as expressing one or more of the tags or polypeptides corresponding to the tags are first provided. The cell can be a cultured cell or a genetically modified cell in which a transcript from the Table, or their complements, or alternatively, transcripts which contain or correspond to a tag or its respective 25 complement is expressed. The cells are cultured under conditions (temperature, growth or culture medium and gas (CO 2 )) and for an appropriate amount of time to attain exponential proliferation without density dependent constraints. It also is desirable to maintain an additionalseparate cell culture; one which does not receive the agent being tested as a control. 30 ~. m~rr C%~~- tot~ iU r- 9M WO 99/65924 PCT/US99/13800 42 As is apparent to one of skill in the art, suitable cells may be cultured in microtiter plates and several agents may be assayed at the same time by noting genotypic changes and/or phenotypic changes. When the agent is a composition other than naked DNA or RNA, the 5 agent may be directly added to the cell culture or added to culture medium for addition. As is apparent to those skilled in the art, an "effective" amount must be added which can be empirically determined. When the agent is a polynucleotide, it may be introduced directly into a cell by transfection or electroporation. Alternatively, it may be inserted into the cell using a gene delivery vehicle or 10 other methods as described above. For the purposes of this invention, an "agent" is intended to include, but not be limited to a biological or chemical compound such as a simple or complex organic or inorganic molecule, a peptide, a protein (e.g. antibody) or a polynucleotide (e.g. anti-sense). A vast array of compounds can be synthesized, 15 for example polymers, such as polypeptides and polynucleotides, and synthetic organic compounds based on various core structures, and these are also included in the term "agent." In addition, various natural sources can provide compounds for screening, such as plant or animal extracts, and the like. It should be understood, although not always explicitly stated that the agent is used alone or in 20 combination with another agent, having the same or different biological activity as the agents identified by the inventive screen. The agents and methods also are intended to be combined with other therapies. The assays also can be performed in a subject. When the subject is an animal such as a rat, mouse or simian, the method provides a convenient animal 25 model system which can be used prior to clinical testing of an agent. In this system, a candidate agent is a potential drug if transcript expression is altered, i.e., upregulated (such as restoring tumor suppressor function), downregulated or eliminated as with drug resistant genes or oncogenes, or if symptoms associated or correlated to the presence of cells containing transcript expression are 30 ameliorated, each as compared to untreated, animal having the pathological cells. e., me-sr .rf- enucrrr (iiI ii 9m~ WO 99/65924 43 PCT/US99/13800 It also can be useful to have a separate negative control group of cells or animals which are healthy and not treated, which provides a basis for comparison. After administration of the agent to subject, suitable cells or tissue samples are collected and assayed for altered gene expression. 5 These agents of this invention and the above noted compounds and their derivatives can be combined with a pharmaceutically acceptable carrier for the preparation of medicaments for use in the methods described herein. The agents of the present invention can be administered to a cell or a subject by various delivery systems known in the art. Non-limiting examples 10 include encapsulation in liposomes, microparticles, microcapsules, expression by recombinant cells, receptor-mediated endocytosis (see, e.g., Wu and Wu (1987) J. Biol. Chem. 262:4429-4432), and construction of a therapeutic nucleic acid as part of a retroviral or other vector. Methods of delivery include but are not limited to transdermally, gene therapy, intra-arterial, intra-muscular, intravenous, 15 intranasal, and oral routes, and include sustained delivery systems. In a specific embodiment, it may be desirable to administer the pharmaceutical compositions of the invention locally to the area in need of treatment; this may be achieved by, for example, and not by way of limitation, local infusion during surgery, by injection, or by means of a catheter or targeted gene delivery of the sequence 20 coding for the therapeutic. Administration in vivo can be effected in one dose, continuously or intermittently throughout the course of treatment. Methods of determining the most effective means and dosage of administration are well known to those of skill in the art and will vary with the composition used for therapy, the purpose of 25 the therapy, the target cell being treated, and the subject being treated. Single or multiple administrations can be carried out with the dose level and pattern being selected by the treating physician. Suitable dosage formulations and methods of administering the agents can be found below. The agents and compositions of the present invention can be used in the 30 manufacture of medicaments and for the treatment of humans and other animals a-'2-v-, tot--,, ip, = l 1ttE WO 99/65924 44 PCTIUS99/13800 by administration in accordance with conventional procedures, such as an active ingredient in pharmaceutical compositions. The pharmaceutical compositions can be administered orally, intranasally, parenterally, transdermally or by inhalation therapy, and may take the form of 5 tablets, lozenges, granules, capsules, pills, ampoules, suppositories or aerosol form. They may also take the form of gene therapy, suspensions, solutions and emulsions of the active ingredient in aqueous or nonaqueous diluents, syrups, granulates or powders. In addition to an agent of the present invention, the pharmaceutical compositions can also contain other pharmaceutically active 10 compounds or a plurality of compounds of the invention. It should be understood that in addition to the ingredients particularly mentioned above, the formulations of this invention may include other agents conventional in the art having regard to the type of formulation in question, for example, those suitable for oral administration may include such further agents as 15 sweeteners, thickeners and flavoring agents. It also is intended that the agents, compositions and methods of this invention be combined with other suitable compositions and therapies. Genomics Applications 20 This invention also provides a process for preparing a database for the analysis of a cell's expressed genes by storing in a digital storage medium information related to the sequences of the transcriptome. Using this method, a data processing system for standardized representation of the expressed genes of a cell is compiled. The data processing system is useful to analyze gene expression 25 between two cells by first selecting a cell and then identifying and sequencing the transcriptome of the cell. This information is stored in a computer-readable storage medium as the transcriptome. The transcriptome is then compared with at least one sequence(s) of transcription fragments from a reference cell. The compared sequences are then analyzed. Uniquely expressed sequences and ~-' E'~E.Y.' ~ 1 C~~ I M WO 99/65924 PCTIUS99/13800 45 sequences differentially expressed between the reference cell and the selected cell can be identified by this method. In other words, this invention provides a computer based method for screening the homology of an unknown DNA or rnRNA sequence against one or 5 more of transcribed or expressed genes of a preselected cell by first providing the complete set of expressed genes, i.e., the transcriptome, in computer readable form and homology screening the DNA or mRNA of the unknown sequence against transcriptome and determining whether the DNA sequence of the unknown contains similarities to any portion of the transcriptome listed in the 10 computer readable form. In one embodiment, SEQ ID Nos. or polynucleotides corresponding to these sequences are the transcriptome against which test cells are compared. Thus, the information provided herein also provides a means to compare the relative abundance of gene transcripts in different biological specimens by use 15 of high-throughput sequence-specific analysis of individual RNAs or their corresponding cDNAs using a modification of the systems described in WO 95/2068, 96/23078 and 5,618,672. The tags or transcripts also can be attached to a solid support for use in high throughput screening assays. PCT WO 97/10365, for example, discloses the 20 construction of high density oligonucleotide chips. See also, U.S. Patent Nos. 5,405,783, 5,412,087 and 5,445,934. Using this method, the probes are synthesized on a derivatized glass surface. Photoprotected nucleoside phosphoramidites are coupled to the glass surface, selectively deprotected by photolysis through a photolithographic mask, and reacted with a second protected 25 nucleoside phosphoramidite. The coupling/deprotection process is repeated until the desired probe is complete. The expression level of a gene is determined through exposure of a nucleic acid sample to the probe-modified chip. Extracted nucleic acid is labeled, for example, with a fluorescent tag, preferably during an amplification step. 30 Hybridization of the labeled sample is performed at an appropriate stringency WO 99/65924 PCT/US99/13800 46 level. The degree of probe-nucleic acid hybridization is quantitatively measured using a detection device, such as a confocal microscope. See U.S. Patent Nos. 5,578,832 and 5,631,734. The obtained measurement is directly correlated with gene expression level. 5 Results from the chip assay are typically analyzed using a computer software program. See, for example, EP 0717 113 A2 and WO 95/2068 1. The hybridization data is read into the program, which calculates the expression level of the targeted gene(s). This figure is compared against existing data sets of gene expression levels 10 for that cell type. Additional utilities of the database include, but are not limited to analysis of the developmental state of a test cell, the influence of viral or bacterial infection, control of cell cycle, effect of a tumor suppressor gene or lack thereof, polymorphism within the cell type, apoptosis, and the effect of regulatory genes. 15 Non-Human Transgenic Animals In another aspect, the novel polynucleotide sequences associated with a pathological state of a cell can be used to generate transgenic animal models. In recent years, geneticists have succeeded in creating transgenic animals, for 20 example mice, by manipulating the genes of developing embryos and introducing foreign genes into these embryos. Once these genes have integrated into the genome of the recipient embryo, the resulting embryos or adult animals can be analyzed to determine the function of the gene. The mutant animals are produced to understand the function of known genes in vivo and to create animal models of 25 human diseases. (see, e.g., Chisaka et al. (1992) 355:5 1 6-520; Joyner et al. ( 1 992) in POSTIMPLANTATION DEVELOPMENT IN THE MOUSE (Chadwick and Marsh, eds., John Wiley & Sons, United Kingdom) pp:277-297; Dorin et al. (1992) Nature 359:211-215). The following examples are intended to illustrate, but not limit, the 30 invention. ~~~~'oc- totI~ itC v:~ ~II 9RI WO 99/65924 PCT/US99/13800 47 Cloning Techniques The following are several techniques available to the skilled artisan for identification and cloning of the polynucleotides corresponding to the tags having the sequences set forth in the Table. 5 1) RACE-PCR Technique One method to isolate the gene or cDNA which codes for a polypeptide or protein involves the 5'-RACE-PCR technique. In this technique, the poly-A mRNA that contains the coding sequence of particular interest is first identified 10 by hybridization to a sequence disclosed herein and then reverse transcribed with a 3'-primer comprising the sequence disclosed herein. The newly synthesized cDNA strand is then tagged with an anchor primer of a known sequence, which preferably contains a convenient cloning restriction site attached at the 5' end. The tagged cDNA is then amplified with the 3'-primer (or a nested primer sharing 15 sequence homology to the internal sequences of the coding region) and the 5' anchor primer. The amplification may be conducted under conditions of various levels of stringency to optimize the amplification specificity. 5'RACE-PCR can be readily performed using commercial kits (available from, e.g., BRL 5 Life Technologies Inc., Clontech) according to the manufacturer's instructions. 20 2) Isolation ofpartial cDNA (3 'fragment) by 3 'directed PCR reaction This procedure is a modification of the protocol described in Polyak et al. (1997) Nature 389:300. Briefly, the procedure uses SAGE tags in PCR reaction such that the resultant PCR product contains the SAGE tag of interest as well as 25 additional cDNA, the length of which is defined by the position of the tag with respect to the 3' end of the cDNA. The cDNA product derived from such a transcript driven PCR reaction can be used for many applications. RNA from a source believed to express the cDNA corresponding to a given tag is first converted to double-stranded cDNA using any standard cDNA 30 protocol. Similar conditions used to generate cDNA for SAGE library WO 99/65924 48 PCTIUS99/13800 construction can be employed except that a modified oligo-dT primer is used to derive the first strand synthesis. For example, the oligonucleotide of composition 5'-Biotin-TCC GGC GCG CCG TTT T CC CAG TCA CGA( 30 )-3' (SEQ ID NO:208), contains a poly-T stretch at the 3' end for hybridization and priming 5 from poly-A tails, an M13 priming site for use in subsequent PCR steps, a 5' Biotin label (B) for capture to strepavidin-coated magnetic beads, and an AscI restriction endonuclease site for releasing the cDNA from the streptavidin-coated magnetic beads. Theoretically, any sufficiently-sized DNA region capable of hybridizing to a PCR primer can be used as well as any other 8 base pair 10 recognizing endonuclease. cDNA constructed utilizing this or similar modified oligo-dT primer is then processed exactly as described in U.S. Patent No. 5,695,937 up until adapter ligation where only one adapter is ligated to the cDNA pool. After adapter ligation, the cDNA is released from the streptavidin-coated magnetic beads and is 15 then used as a template for cDNA amplification. Various PCR protocols can be employed using PCR priming sites within the 3' modified oligo-dT primer and the SAGE tag. The SAGE tag-derived PCR primer employed can be of varying length dictated by 5' extension of the tag into the adaptor sequence. cDNA products are now available for a variety of 20 applications. This technique can be further modified by: (1) altering the length and/or content of the modified oligo-dT primer; (2) ligating adaptors other than that previously employed within the SAGE protocol; (3) performing PCR from template retained on the streptavidin-coated magnetic beads; and (4) priming first 25 strand cDNA synthesis with non-oligo-dT based primers. 3) Isolation of cDNA using GeneTrapper or modified GeneTrapper Technology 30 The reagents and manufacturer's instructions for this technology are commercially available from Life Technologies, Inc., Gaithersburg, Maryland. '-U u,-,me.e.rqi u===== ne mt""T" IDI II C O WO 99/65924 PCT/US99/13800 Briefly a complex population of single-stranded phagemid DNA containing directional cDNA inserts is enriched for the target sequence by hybridization in solution to a biotinylated oligonucleotide probe complementary to the target sequence. The target sequence is based on the tag sequence of the present 5 invention. The hybrids are captured on streptavidin-coated paramagnetic beads. A magnet retrieves the paramagnetic beads from the solution. leaving nonhybridized single-stranded DNAs behind. Subsequently, the captured single-stranded DNA target is released from the biotinylated oligonucleotide. After release, the cDNA clone is further enriched by using a nonbiotinylated target oligonucleotide to 10 specifically prime conversion of the single-stranded target to double-stranded DNA. Following transformation and plating, typically 20% to 100% of the colonies represent the cDNA clone of interest. To identify the desired cDNA clone, the colonies may be screened by colony hybridization using the 32 P-labeled oligonucleotide as described above for solution hybridization, or alternatively by 15 DNA sequencing and alignment of all sequences obtained from numerous clones to determine a consensus sequence. 4) Isolation of cDNAs from a library by probing with the SAGE transcript or tag 20 Classical methods of constructing cDNA libraries are taught in Sambrook et al., supra. Recent procedures described in Velculescu et al. (1997) Science 270:484) can be employed to construct an expression cDNA library cloned into the ZAP Express vector. A ZAP Express cDNA synthesis kit is available from 25 Stratagene is used accordingly to the manufacturer's protocol. Plates containing 250 to 2000 plaques are hybridized as described in Rupert et al. (1988) Mol. Cell. Bio. 8:3104 to oligonucleotide probes with the same conditions previously described for standard probes except that the hybridization temperature is reduced to room temperature. Washes are performed in 6X standard-saline-citrate 0.1% 30 SDS for 30 minutes at room temperature. The probes are labeled with 32 P-ATP through use of T4 polynucleotide kinase. ~~6 ~ uq.,~LJ~F f1 It C O WO 99/65924 50 PCT/US99/13800 5) Identification of known genes or ESTs In addition, databases exist that reduce the complexity of ESTs by assembling contiguous EST sequences into tentative genes. For example, TIGR has assembled human ESTs into a database called THC for tentative human 5 consensus sequences. The THC database allows for a more definitive assignment compared to ESTs alone. Software programs exist (TIGR assembler and TIGEM EST assembly machine and contig assembly program (see Huang, X. (1996) Genomics 33:21-23)) that allow for assembling ESTs into contiguous sequences from any organism. 10 Isolation, culturing and expansion of APCs including Dendritic cells Various methods to isolate and characterize APCs including DCs have been known in the art. At least two methods have been used for the generation of human dendritic cells from hematopoictic precursor cells in peripheral blood or 15 bone marrow. One approach cultures the monocytes in GM-CSF and IL-4 as described below, where the immature portion was used for SAGE while an additional portion was treated with TNF-ca for 36 hours to encourage their maturation. This method was followed for the isolation and culturing of the cell line identified herein as TNF-ca matured dendritic cells. 20 The other method makes use of the more abundant CD34 precursor population, such as adherent peripheral blood monocytes, and stimulate them with GM-CSF plus IL-4 (see, for example, Sallusto et al. (1994), supra). In other aspects of the invention, the methods described in Romani et al. (1996), supra or Bender et al. (1996), supra are used to generate both immature 25 and mature dendritic cells from the peripheral blood mononuclear cells (PBMC) of a mammal, such as a murine, simian or human. Briefly, isolated PBMC are pre-treated to deplete T- and B-cells by means of an immunomagnetic technique. Lymphocyte-depleted PBMC are then cultured for 7 days in RPMI medium, supplemented with 1% autologous human plasma and GM-CSF/IL-4, to generate 30 dendritic cells. On day 7, non-adherent cells are harvested for further processing. ~. *.~j~ytqg r- _LI-r In21 CI 13 WO 99/65924 51 PCT/US99/13800 The dendritic cells derived from PBMC in the presence of GM-CSF and IL-4 are immature, in that they can lose den Iritic cell properties and revert back to macrophage cell fate if the cytokine stimuli are removed from the culture. A population of dendritic cells having these features were used to isolate the 5 transcripts of immature dendritic cells. The dendritic cells in an immature state are very effective in processing native protein antigens for the MHC class II restricted pathway (Romani et al. (1989) J. Exp. Med 169:1169.) Further maturation is accomplished by culturing in conditioned medium or treating with LPS or TNF-a or CD40 ligand for 3 days in a macrophage 10 conditioned medium (CM), which contains the necessary maturation factors. Mature dendritie cells are less able to capture new proteins for presentation but are much better at stimulating resting T cells (both CD4' and CD8~) to grow and differentiate. Mature dendritic cells can be identified by their change in morphology, 15 such as the formation of more motile cytoplasmic processes; by the presence of at least one of the following markers: CD83, CD68, HLA-DR or CD86; or by the loss of Fc receptors such as CD1 15 (reviewed in Steinman (1991) Ann. Rev. Immunol. 9:271.) 20 Computational Analysis This tags identified and claimed herein and populations thereof can be used in further computational analysis. The sequences and expression profiles of this invention are stored in any functionally relevant program, e.g., in Compare Report using the SAGE software (available through Dr. Ken Kinzler at Johns 25 Hopkins University). The Compare Report provides a tabulation of the polynucleotide sequences and their abundance for the samples normalized to a defined number of polynucleotides per library (say 25,000). This information can be imported into MS-ACCESS either directly or via copying the data into an Excel spreadsheet first and then from there into MS-ACCESS for additional 30 manipulations. Other programs such as SYBASE or Oracle that permit the ^2' ime---- -- t- e .LI-T I Cl 3M WO 99/65924 52 PCT/US99/13800 comparison of polynucleotide numbers could be used as alternatives to MS ACCESS. Enhancements to the software can be designed to incorporate these additional functions. These functions consist in standard Boolean, algebraic, and text search operations, applied in various combinations to reduce a large input set 5 of polynucleotides to a manageable subset of polynucleotides of specifically defined interest. Sequence information and abundance from a test sample or cell also is input into the functionally relevant program. The researcher may create groups containing one or more project(s) by 10 combining the counts of specific polynucleotides within a group (e.g., GroupNormal = Normall + Normal2, GroupTumor = PrimaryTumorl + TumorCelLine). Additional characteristic values are also calculated for each tag in the group (e.g., average count, minimum count, maximum count). The researcher may calculate individual tag count ratios between groups, for example 15 the ratio of the average GroupNormal count to the average GroupTumor count for each polynucleotide. The researcher may calculate a statistical measure of the significance of observed differences in tag counts between groups. To identify the polynucleotides within MS-ACCESS, a query to sort polynucleotide tags based on their abundance in the sample cells is run. The 20 output from the Query report lists specific polynucleotides (by sequence) that fit the sorting criteria and their abundance in the various sample cells. The sorting is based on the principle that the gene product of interest (and hence the corresponding polynucleotide) is more abundant in the samples that prominently exhibit the chosen phenotype than in samples that do not exhibit the 25 phenotype. For example, one may query to identify polynucleotides that are present at a level of 10 tags when the total number of tags per library has been normalized to a defined number in the reference sample against one or more test samples. The results of the search might reveal that 5 different polynucleotides fit the sorting 30 criteria, hence there are 5 candidates genes to be tested to determine whether they WO 99/65924 53 PCT/US99/13800 confer the phenotype when transferred into samples that do not have the phenotype. The more stringent the sorting criteria, the more efficient the sorting should be. Thus if one asked for polynucleotides that are at 5 tags when the total 5 number of tags per library has been normalized to a defined number in the reference sample and less than 5 in the test sample, a large number of candidates would be generated. However, if one can increase the differential because the samples manifest extremes of the phenotype (say >10 in the test sample and <1 in the one or more reference samples) this restricts the number of candidates that 10 will be identified. Prior knowledge of what amount of gene product (hence abundance of polynucleotides) is required to confer the phenotype is not essential as one can arbitrarily select a set of sorting parameters, run the data analysis, and identify and test candidates. If the desired candidate is not found the stringency of the 15 sorting criteria can be reduced (i.e. reduce the differential) and the new candidates that are found can be tested. Iterative cycles of sorting and testing candidates should eventually culminate in the successful recovery of the desired candidate. Knowledge of what amount of gene product (hence abundance of polynucleotide) is required to confer the phenotype will permit the rationale use 20 of stringent sorting criteria and greatly accelerate the search process as the desired gene may be captured within a handful of candidates Establishing what amount of gene product is required to confer a specific phenotype will be dependent on the specific phenotype in question and the sensitivity of assays that measure that phenotype. 25 Accordingly, one enters the individual polynucleotide sequences from the Query report into the program to determine if there is a match with any known genes or whether they are potentially novel (no match=NM). One then retrieves cDNAs corresponding to specific sequences from the Query Report and test them individually in an appropriate biological assay to 30 determine if they confer the phenotype. Of the candidates that correspond to WO 99/65924 PCT/US99/13800 known genes, it is a relatively easy task to obtain complementary DNAs for these candidates and test them individually to determine if they confer the specific phenotype in question when transferred into cells that do not exhibit the phenotype. If none of the known genes confer the phenotype, retrieve the cDNAs 5 corresponding to the No Match sequences of the Query Report by PCR cloning and test the novel cDNAs individually for their ability to confer the phenotype. If the assumptions made up to this point are sound (i.e., a single gene product can confer the phenotype; the sorting criteria are not too stringent so as to exclude the desired candidate) then a cDNA corresponding to one of the candidates of the 10 Query Report will be found to confer the phenotype and the search is over. If however none of the candidates are found to confer the phenotype then one may need to reduce the stringency of the sorting parameters to "cast a wider net" and capture more candidates to be tested as above. In one embodiment, the polynucleotide or gene sequence can also be 15 compared to a sequence database, for example, using a computer method to match a sample sequence with known sequences. Sequence identity can be determined by a sequence comparison using, i.e., sequence alignment programs that are known in the art, such as those described in CURRENT PROTOCOLS IN MOLECULAR BIOLOGY (F.M. Ausubel et al., eds., 1987) Supplement 30, 20 section 7.7.18, Table 7.7.1. A preferred alignment program is ALIGN Plus (Scientific and Educational Software, Pennsylvania), preferably using default parameters, which are as follows: mismatch = 2; open gap = 0; and extend gap = 2. Another preferred program is the BLAST program for alignment of two nucleotide sequences, using default parameters as follows: open gap = 50; 25 extension gap - 2 penalties; gap x dropoff = 0; expect = 10; word size = 11. The BLAST program is available at the following Internet address: http://www.ncbi.nlm.nih.gov. Alternatively, hybridization under conditions of high, moderate and low stringency can also indicate degree of sequence identity. 30 ~~~.'Lum - ioi*-ur U9 I~ TIIii WO 99/65924 55 PCT/US99/13800 Vaccines for Cancer Treatment and Prevention In one embodiment, the present invention comprises vaccines for cancer treatment. Recent advances in vaccine adjuvants provide effective means of administering peptides so that they impact maximally on the immune system. 5 Del-Giudice (1994) Experientia 50:1061-1066. A polynucleotide of this invention can be administered in alone or in combination with a polynucleotide encoding an antigenic peptide as a cancer vaccine. The polynucleotide can be administered as naked DNA or alternatively, in expression vectors. Therapy can be enhanced by coadministration of cytokine and/or co-stimulatory molecules which in turn, can 10 be administered as proteins or the polynucleotides encoding the proteins. Host cells comprising Antigenic Peptides of the Invention The invention further provides isolated host cells comprising the polynucleotide of the invention. In some embodiments, these host cells present 15 one or more peptides of the invention on the surface of the cell in the context of an MHC molecule, i.e., a antigenic peptide of the invention is bound to a cell surface MHC molecule such that the peptide can be recognized by an immune effector cell. Isolated host cells which present the polypeptides of this invention in the context of MHC molecules are further useful to expand and isolate a 20 population of educated, antigen-specific immune effector cells. The immune effector cells, e.g., cytotoxic T lymphocytes, are produced by culturing naYve immune effector cells with antigen-presenting cells cells which present the polypeptides in the context of MHC molecules on the surface of the APCs. The population can be purified using methods known in the art, e.g., FACS analysis or 25 FICOLTM gradient. The methods to generate and culture the immune effector cells as well as the populations produced thereby also are the inventors' contribution and invention. Pharmaceutical compositions comprising the cells and pharmaceutically acceptable carriers are useful in adoptive immunotherapy. Prior to administration in vivo, the immune effector cells are screened in vitro for their 30 ability to lyse melanoma tumor cells.
WO 99/65924 56 PCTIUS99/13800 Gene transfer Vectors useful in genetic modification In one embodiment, the present invention provides methods of eliciting efficient antigen-specific immune response in a subject by introducing to the 5 subject recombinant polynucleotides encoding antigenic peptides alone or in combination with immunostimulatory factors. Methods and materials for gene transfer are known in the art, including, for example, viral mediated gene transfer, lipofection, transformation, transfection and transduction. The polynucleotides encoding the immunostimulatory factor and target antigenic peptide can be 10 introduced ex vivo into a host cell, for example, dendritic cells. The genetically modified host cells can be introduced as a cell-based vaccine into the target subject. Alternatively, the polynucleotides encoding the immunostimulatory factor and target antigenic peptide can be introduced directly into the subject in the form of gene-based vaccine. 15 Various viral infection techniques have been developed which utilize recombinant viral vectors for gene delivery, and constitute preferred approaches to the present invention. The viral vectors which have been used in gene transfer include, but not limited to, viral sequences derived from simian virus 40 (SV40), adenovirus, adeno-associated virus (AAV), and retroviruses. 20 Vector Transduction of Cells such as APCs APCs can be transduced with viral vectors encoding a relevant polypeptides. The most common viral vectors include recombinant poxviruses such as vaccinia and fowlpox virus (Bronte et al. (1997) Proc. Natl. Acad. Sci. 25 USA 94:3183-3188; Kim et al. (1997) J. Immunother. 20:276-286) and, preferentially, adenovirus (Arthur et al. (1997) J. Immunol. 159:1393-1403; Wan et al. (1997) Human Gene Therapy 8:1355-1363; Huang et al. (1995) J. Virol. 69:2257-2263). Retrovirus also may be used for transduction of human APCs (Marin et al. (1996) J. Virol. 70:2957-2962). 30 WO 99/65924 57 PCTIUS99/13800 In vitro or ex vivo exposure of human DCs to adenovirus (Ad) vector at a multiplicity of infection (MOI) of 500 for 16-24 h in a minimal volume of serum free medium reliably gives rise to foreign polynucleotide expression in 90-100% of DCs. The efficiency of transduction of DCs or other APCs can be assessed by 5 immunofluorescence using fluorescent antibodies specific for the tumor antigen being expressed (Kim et al. (1997) J. Immunother. 20:276-286). Alternatively, the antibodies can be conjugated to an enzyme (e.g. HRP) giving rise to a colored product upon reaction with the substrate. The actual amount of antigenic polypeptides being expressed by the APCs can be evaluated by ELISA. 10 In vivo transduction of DCs, or other APCs, can be accomplished by administration of Ad (or other viral vectors) via different routes including intravenous, intramuscular, intranasal, intraperitoneal or cutaneous delivery. The preferred method is cutaneous delivery of Ad vector at multiple sites using a total dose of approximately lx1o' 0 -lx 10" i.u. Levels of in vivo transduction can be. 15 roughly assessed by co-staining with antibodies directed against APC marker(s) and the antigen being expressed. The staining procedure can be carried out on biopsy samples from the site of administration or on cells from draining lymph nodes or other organs where APCs (in particular DCs) may have migrated (Condon et al. (1996) Nature Med. 2:1122-1128; Wan et al. (1997) Human Gene 20 Therapy 8:1355-1363). The amount of antigen being expressed at the site of injection or in other organs where transduced APCs may have migrated can be evaluated by ELISA on tissue homogenates. Although viral gene delivery is more efficient, DCs can also be transduced in vitro/ex vivo by non-viral gene delivery methods such as electroporation, 25 calcium phosphate precipitation or cationic lipid/plasmid DNA complexes (Arthur et al. (1997) Cancer Gene Therapy 4:17-25). Transduced APCs can subsequently be administered to the host via an intravenous. Subcutaneous, intranasal, intramuscular or intraperitoneal route of delivery. In vivo transduction of DCs, or other APCs, can potentially be 30 accomplished by administration of cationic lipid/plasl lid DNA complexes an=====--ae -ws- es~t-o"" 1 1i C IM WO 99/65924 58 PCT/US99/13800 delivered via the intravenous, intramuscular, intranasal, intraperitoneal or cutaneous route of administration. Gene gun delivery or injection of naked plasmid DNA into the skin also leads to transduction of DCs (Condon et al. (1996) Nature Med. 2:1122-1128 and Raz et al. (1994) Proc. Natl. Acad. Sci. 5 USA 91:9519-9523). Intramuscular delivery of plasmid DNA may also be used for immunization (Rosato et al. (1997) Human Gene Therapy 8:1451-1458. The transduction efficiency and levels of foreign polynucleotide expression can be assessed as described above for viral vectors. 10 Administration of Cell-Based Vaccine to Subject Genetically modified cells can subsequently be administered to the host subject via various routes, including, for example, intravenous infusion, subcutaneous injection, intranasal, intramuscular or intraperitoneal delivery. The cells containing the recombinant polynucleotides may be used to confer immunity 15 to individuals. Administration in vivo can be effected in one dose, continuously or intermittently throughout the course of treatment. Methods of determining the most effective means and dosage of administration are well known to those of skill in the art and will vary with the composition used for therapy, the purpose of the therapy, the target cell being treated, and the subject being treated. Single or 20 multiple administrations can be carried out with the dose level and pattern being selected by the treating physician. Adoptive Immunotherapy Methods The expanded populations of antigen-specific immune effector cells and 25 APCs presenting antigens find use in adoptive immunotherapy regimes. Adoptive immunotherapy methods involve, in one aspect, administering to a subject a substantially pure population of educated, antigen-specific immune effector cells made by culturing naYve immune effector cells with APCs as described above. In some embodiments, the APCs are dendritic cells. 30 WO 99/65924 59 PCT/US99/13800 In one embodiment, the adoptive immunotherapy methods described herein are autologous. In this case, the APCs are made using parental cells isolated from a single subject. The expanded population also employs T cells isolated from that subject. Finally, the expanded population of antigen-specific 5 cells is administered tothe same patient. In a further embodiment, APCs or immune effector cells are administered with an effective amount of a stimulatory cytokine, such as IL-2 or a co stimulatory molecule. 10 Immune Effector Cells The present invention makes use of the above-described antigen presenting matrices, including APCs, to stimulate production of an enriched population of antigen-specific immune effector cells. Accordingly, the present invention provides a population of cells enriched in educated, antigen-specific 15 immune effector cells, specific for an antigenic peptide of the invention. These cells can cross-react with (bind specifically to) antigenic determinants (epitopes) on natural (endogenous) antigens. In some embodiments, the natural antigen is on the surface of tumor cells and the educated, antigen-specific immune effector cells of the invention suppress growth of the tumor cells. When APCs are used, 20 the antigen-specific immune effector cells are expanded at the expense of the APCs, which die in the culture. The process by which naive immune effector cells become educated by other cells is described essentially in Coulie (1997) Molec. Med. Today 3:261-268. An effector cell population suitable for use in the methods of the present 25 invention can be autogeneic or allogeneic, preferably autogeneic. When effector cells are allogeneic, preferably the cells are depleted of alloreactive cells before use. This can be accomplished by any known means, including, for example, by mixing the allogeneic effector cells and a recipient cell population and incubating them for a suitable time, then depleting CD69* cells, or inactivating alloreactive 30 cells, or inducing anergy in the alloreactive cell population. ~..#q~1 9r- trI~ i iDi I 13 WO 99/65924 60 PCT/US99/13800 Hybrid immune effector cells can also be used. Immune effector cell hybrids are known in the art and have been described in various publications. See, for example, International Patent Application Nos. WO 98/46785; and WO 95/16775. 5 Although the foregoing invention has been described in some detail by way of illustration and example for purposes of clarity of understanding, it will be apparent to those skilled in the art that certain changes and modifications will be practiced. Therefore, the description and examples should not be construed as limiting the scope of the invention, which is delineated by the appended claims. 10 Example 1 The Table illustrates a series of mRNAs, both known and unknown, that were found by SAGE analysis to be differentially expressed in monocyte-derived dendritic cells as compared to monocytes. SAGE analysis revealed for instance 15 that the chemokines PARC and TARC that can recruit activated T cells are differentially expressed by monocyte-derived immature dendritic cells (prepared by culturing PBMC derived monocytes in GM-CSF and IL4). Other immunostimulatory factors to mention include: monocyte chemotactic protein-4 (MCP-4) [Berkhout et al., (1997) Journal of Biological Chemistry 272(26): 20 16404-16413] U46767; macrophage-derived chemokine (MDC) [Godiska et al., (1997) Journal of Experimental Medicine 185(9): 1595-1604] U83171; ecalectin [Matsumoto et al., (1998) Journal of Biological Chemistry 273(27): 16976 16984], AB005894; and monocyte chemotactic protein-2 (MCP-2) [Proost et al., 1996 Joumla of Leukocyte Biology 59(1): 67-74] Y10802. The fact that dendritic 25 cells produce abundant levels of these chemokines has not been reported previously. The genes encoding these chemokines could be linked to the gene or genes encoding tumor antigens in a DNA based vaccine to ensure that APCs transduced with the vaccine will produce and process not only the tumor antigen, but also the stimulatory chemokines. Any cDNA or gene encoding any of the 30 mRNAs or combination of mRNAs identified by differential gene expression ~,tr%^-pr rr euccT ftim ii=9% WO 99/65924 61 PCTIUS99/13800 analysis (such as SAGE) as being differentially expressed in immature dendritic cells as shown in Table I could be linked to a tumor antigen gene or genes to prepare superior vaccines. Similarly, any cDNA or gene encoding any mRNA or combination of mRNAs identified by differential gene expression analysis (such 5 as SAGE) as being differentially expressed in mature dendritic cells could be linked to a tumor antigen gene or genes to prepare superior vaccines. Example 2 The genes encoding mRNAs that are differentially expressed in either 10 immature or mature dendritic cells are apt to be regulated by specific transcription factors. Thus, an alternative to delivering the gene encoding an mRNA identified as being differentially expressed in either mature or immature dendritic cells by comparative gene expression analysis (such as SAGE) would be to deliver a gene or genes that encode transcription factors (either naturally occurring or engineered 15 via recombinant DNA technology) that can transactivate the expression of the endogenous gene that encodes the differentially expressed mRNA. Thus, the present invention also pertains to vaccines in which a gene or genes encoding tumor antigens is linked to genes encoding transcription factors or transactivators that can upregulate the expression of mRNAs identified as being differentially 20 expressed in either mature or immature dendritic cells by comparative gene expression analysis. Example 3 Differential gene expression analysis would also be expected to reveal 25 genes encoding cell surface proteins that are preferentially expressed by either immature or mature dendritic cells as compared to monocyte precursors. These cell surface molecules may play a pivotal role in the function of dendritic cells by acting as co-stimulatory signals or modulators of DC function or migration. Naturally occurring ligands specific for these cell surface molecules or 30 recombinant proteins (such as an antibody) generated to have specificity for these ~rr eucr- r toi ii = 9R1 WO 99/65924 PCTIUS99/13800 62 cell surface molecules might be expected to interact with the cell surface protein to stimulate the function of the dendritic cells or foster the maintenance of an activated state or stimulate the migration of dendritic cells to sites rich in T cells. Thus, the present invention relates to vaccines in which a gene or genes encoding 5 tumor antigen or antigens is linked to a gene or genes encoding secreted proteins that have the capacity to bind to and modulate the activity of cell surface proteins identified as being differentially expressed in either mature or immature dendritic cells by comparative gene expression analysis (such as SAGE). In this manner, a novel autocrine loop is established whereby a genetically modified APC produces 10 a ligand that is secreted from the APC where it can bind to cell surface receptors on that cell and stimulate the genetically modified dendritic cell. Although the foregoing invention has been described in some detail by way of illustration and example for purposes of clarity of understanding, it will be apparent to those skilled in the art that certain changes and modifications will be 15 practiced. Therefore, the description and examples should not be construed as limiting the scope of the invention, which is delineated by the appended claims.
WO 99/65924 63 PCT/US99/13800 TABLE 1 TAG SEQ ID DESCRIPTION ACCESSION NO TATATTTTCT 1. Human transglutaminase mRNA, 3' M98479 untranslated region. TCTCCATACC 2. mitochondrial AGAAGTGTCC 3. Homo sapiens (HepG2) LAL mRNA for Z31690 lysosomal acid lipase. GGCACAAAGG 4. Human mRNA for chemokine, complete D43767 cds. TGGCCCCAGG 5. Human mRNA for precursor of X00570 apolipoprotein CI (apo CI). TGTACCTGTA 6. Homo sapiens (xs31) mRNA, 835bp. Z36832 AAGGGATGCT 7. Human monocyte chemoattractant U46767 protein-4 precursor (MCP-4) mRNA, GAGGGTGCCA 8. Human complement component 1, beta- M36278 chain (Cl QB) mRNA, partial cds. TCTTGATTTA 9. Human alpha-2-macroglobulin mRNA, Ml 1313 complete cds. CGACCCCACG 10. Human apolipoprotein E mRNA, M12529 complete cds. ACCCGCCGGG 11. 18s ribosomal RNA ATCTTGTTAC 12. Human mRNA for fibronectin (FN X02761 KOO precursor). GGGGGTGAAG 13. Homo sapiens mRNA for Y11525 CCAAT/enhancer binding protein alpha. CCTTGTCCTC 14. Homo sapiens mRNA for GM2 activator X62078 protein. GCCGCTACTT 15. Homo sapiens mRNA encoding RAMP 1. AJO01014 CTGGGCCTGG 16. Hs.74573: Human HU-K4 mRNA, complete cds H GACCCGCTGG 17. xxx ATATACTGTG 18. AC005102 Homo sapiens clone RG356E01, complete seq AAACTTTGCC 19. ACTATTTCCA 20. Human long transcript fructose-1,6- U47918 bisphosphatase (HUMFBPase) mRNA, ACCCAGGGTA 21. Hs.153876: ESTs Hs.153876: ESTsHs.153876: ACTGGAACGA 22. Homo sapiens NADP-dependent AF020038 isocitrate dehydrogenase (IDH) mRNA, I WO 99/65924 PCT/US99/13800 64 TAG SEQ ID DESCRIPTION ACCESSION NO I AACGGGGCCC 23. Human macrophage-derived chemokine U83 171 precursor (MDC) mRNA. complete I______ CCTGGGGTAA 24. Human mRNA for DC class X00452 KOI ________________ histocompatibility antigen alpha-chain. ______ TCACCGGTCA 25. Human mRNA for plasma gelsolin. IX04412 AATGCAGGCA 26. Human S-adenosyihomocysteine M61831 ____________ ________hydrolase (AHCY) mRNA. complete cds. CTGACCTGTG 27. Human heart mRNA for MHC class I D87665 ________HLA-Bw62, partial cds. GATCAATCAG 28. Homo sapiens mRNA for CC chemokine. AB000221 complete cds. CCCCCTGCCC 29. GTGACCACGG 30. Human N-methyl-D-aspartate receptor 2C subunit pre TGTCCCAGCC 31. Human mRNA for acid phosphatase type X14618 5 (EC 3.1.3.2). _____ ACCTTTACTG 32. GTGTGTAAAA 33. TGTTTGGGGG 34. AATATTGCAC 35. TGGCTGGCCA 36. GCGTCTGGGG 37. TATTTATTCC 38. Human mRNA for Src-like adapter D89077 prcursprotein, complete cds. CTGGGCCAGC 39. ESTs, Weakly similar to SYNAPTOBREVIN 2 [Homo sapiens CTTGATTCCC 40. Homo sapiens quiescin (Q6) mRNA U97276 hyrsAC)mNcomplete cds. CTCATAAGGA 41. Tag matches mitochondrial DNA AGAGGTGTAG 42. xxx CCTCACTACC 43. xxx AAGAAGCAGG 44. Homo sapiens unknown mRNA, AF047439 complete cds. TGGCTAGTGT 45. Human mRNA for proteasome subunit z, D38048 protein,complete cds. CTCTAAGAAG 46. Hs.9641: ESTs, Highly similar to COMPLEMENT C IQ SU ____ TGGCTGTGTG 47. ATCCTCCCTA 48. Homospiensquiescn_(Q6 _mRNA U97276 TCCTACGTGA 49. __agmatchesmitochondrialDNA TGGGAAACCT 50. GCGAAACCCA 51. several hits AGTATCTGGG 52. Homo sapiens Arp2/3 protein complex AF006084 subunit p4z-Arc (ARC41) mRNA.
WO 99/65924 PCT/US99/13800 65 TAG SEQ ID DESCRIPTION ~ ACCESSION NO CCCTCGGTCC 53. ---- GTAATCCTGC 54. TCGTAACGAG 55. ACTTAGGGAG 56. Human MrNA for LIMK (LIM kinase). D26309 complete cds. GAGGAAGAAG 57. tumor rejection antigen/tendoplasmic S74942 reticular heat shock protein TGGAAGGGCA 58. CTAGCCAGCA 59. -- TATTATTAAA 60. --- ~ GCCATCCAGA 61. Hs.107479: Homo sapiens mRNA for KIAA0738 protein, TATTGGCCTG 62. Hs.79572: Cathepsin D (lysosomal aspartyl protease GCCACCCCGT 63. Human mRNA for glucose-6-phosphate X03674 dehydrogenase (G6PD). CGCCGACGAT 64. Human interferon-inducible mRNA X02492 X02 fragment (cDNA 6-16). GCTCTGCCTC 65. Homo sapiens cathepsin X precursor. AF073890 mRNA, complete cds. CCTGTACCCC 66. Homo sapiens Sox-like transcriptional AF072836 factor mRNA, complete cds. TGTCGCTGGG 67. Hs. 154811: C4/C2 activating component of Ra-reacti AGAAGCCGTG 68. Human neutrophil adherence receptor J04145 alpha-M subunit mRNA. GGAAAATGGG 69. xxx GATACAGCCA 70. Human mRNA for lymphocyte IgE X04772 receptor (low affinity receptor Fc ATGTGCGTGG 71. Human SNC19 mRNA sequence. U20428 AAGGCGTTTC 72. GAGAACGGGG 73. CAGAATGACT 74. GGGAAACAGG 75. CTGTCTTGGG 76. GTCTGAGCTC 77. GTGGCTTCCC 78. TATCTGTCTA 79. TGCTTTGGGA 80. TCTCTGATGC 81. Homo sapiens mRNA for SH3 binding AB000462 protein, complete cds, AAATCAATAC 82. AGCCGGGATG 83. Homo sapiens RING12 mRNA. X62741 GCAGGCCATA 84.
WO 99/65924 PCT/US99/13800 66 TAG SEQ ID DESCRIPTION ACCESSION NO GGGCCCAGGA 85. TTGTCGATGG 86. Homo sapiens full length insert cDNA AF086553 clone ZE12HO5. TGGGCTCTGA 87. Human mRNA for lysosomal D12676 sialoglycoprotein, complete cds. CAGTCATTTG 88. TTTCTAGTTT 89. Human mRNA for KIAA0108 gene. D14696 complete cds. TGGATCCTAG 90. Homo sapiens NADH:ubiquinone AF067139 oxidoreductase NDUFS3 subunit mRNA. TCTCCAGGAA 91. GCTTTGCAGT 92. Homo sapiens TWIK-related acid- AF006823 sensitive K+ channel (TASK) mRNA. AGTTTCCCAA 93. Homo sapiens SULTIC sulfotransferase AF055584 (SULTIC) mRNA, complete cds. TCCACGCACC 94. CACCGCTGCA 95. GTGATGGATG 96. AGTGTATTTT 97. Human mRNA for insuline-like growth Y00285 factor II receptor. AATGACTGAA 98. GGAGATGAGG 99. TTGAGACCTC 100. Human factor XIII subunit a mRNA, 3' M14539 end. CTACACCAGT 101. GCCGTTCTTA 102. xxx TGCAGAAGAA 103. Homo sapiens mRNA for macrophage X55635 mannose receptor. CTAACTAGTT 104. xxx GCCTGCAGTC 105. Homo sapiens mRNA for hepatocyte AB006534 growth factor activator inhibitor ACATTCTTTT 106. Homo sapiens NMB mRNA. X76534 TTTGTAGATG 107. Human HepG2 3' region MboI cDNA, D17196 clone hmd3c06m3. ACAACTCAAT 108. Human HepG2 3' region cDNA, clone D16936 hmd4hl0. TCAGATAGGA 109. Human MAPKAP kinase (3pK) mRNA, U09578 complete cds. ATTAAGAGGG 110. GTGTGTCTGA 111. Human mRNA encoding major V00522 histocompatibility complex gene HLA DR CAGCATCTAA 112. ATGTAGGTGC 113. Homo sapiens mRNA for phosphatidic AB000889 WO 99/65924 PCT/US99/13800 67 TAG SEQ ID DESCRIPTION ACCESSION NO acid phosphatase 2b, complete TGAGAGCAGC 114. TTTTAGCAGG 115. Homo sapiens clone 24560 unknown AF055001 mRNA, complete cds. GGTGAAGAGG 116. Homo sapiens mRNA for hepatocyte AB000095 growth factor activator inhibitor, GAAGTCGGAA 117. CTATATTTTT 118. Homo sapiens mRNA for cathepsin C. X872 12 TCCTGCTGGC 119. GTCAGAATGG 120. Homo sapiens clone 23863 mRNA, AF035299 partial cds. GCAACGGGCC 121. Homo sapiens mRNA for brain acyl-CoA D88894 hydrolase, complete cds. CTGATCTCCA 122. AACTAATACT 123. AATGGATGAA 124. CACATCCTTA 125. CCACGCACTG 126. TAGGTCACCT 127. ATTTAGCAAG 128. CTGGATGGGC 129. TTfGGAACAAT 130. CAGGATCCAG 131. Human progesterone receptor-associated U28918 p48 protein mRNA, complete GTGACTGCCA 132. Homo sapiens clone 24722 unknown AF055020 mRNA, partial cds. GCTTGCTGGC 133. CCCTGGGCGA 134. CCCCTCCCCC 135. Human velo-cardio-facial syndrome U84524 22ql1 region mRNA sequence. GATTACCTGT 136. TCAATAAATG 137. GGCCCTCTGA 138. Human peptidyl-prolyl isomerase and U49070 essential mitotic regulator ACCAGAGGGG 139. GACGTTCACT 140. TGGCAAACGT 141. CAAGACGGGG 142. CAGGTTGTGA 143. Human mRNA for lysosomal acid X12548 phosphatase (EC 3.1.3.2). CTTGACCTGT 144. TACCCCACCC 145. Human zinc finger protein (MAZ) M94046 mRNA.
WO 99/65924 PCT/US99/13800 68 TAG SEQ ID DESCRIPTION ACCESSION NO TTACCTTTTT 146. Human beta-D-galactosidase mRNA. M27507 J05 complete cds. AGCAGCAACA 147. ACTATTAGTG 148. Homo sapiens doc-l mRNA, complete AB006077 cds. ATTGCTCTCT 149. GCCCCAGCCC 150. Human chitotriosidase precursor mRNA, U29615 complete cds. GGTGGGGAGA 151. Human chromosome l 7 q21 mRNA clone U18009 LF 113. ACCAGCCACA 152. cysteine proteinase cystatin c TGCCTCTGCG 153- Homo sapiens mRNA for CD151, D29963 complete cds. ATATAATCTG 154. Human non-integrin laminin-binding M36682 _7_ protein mRNA, complete cds. GGAGCAGACG 155. GTGCGCTGAG 156. Human mRNA for HLA class I locus C X58536 heavy chain. TTTTCTGAAA 157. Human thioredoxin (TXN) mRNA, J04026 complete cds. CTGTTAGTGT 158. Homo sapiens malate dehydrogenase U20352 (MDHA) mRNA, complete cds. GAGGACTCCG 159. Human tryptophanyl tRNA synthetase M77804 (IFNWRS) mRNA, complete cds. GAAATACAGT 160. Human cathepsin D mRNA, complete Ml 1233 cds. GGAACTTTTA 161. Hs.43857: ESTs GAAGCAGGAC 162. Homo sapiens mRNA for non-muscle X95404 type cofilin. AGCCACCGCA 163. Human mRNA upregulated during U58668 camptothecin-induced apoptosis of ATGAAACTTC 164. mitochondrial CTGGACCCGG 165. Fructose 1-6 biphosphate GTGCTATTCT 166. AGGGCAGGGA 167. Homo sapiens clone 24444 RaP2 AF055026 interacting protein 8 (RPIP8) mRNA, GGCAGCGCCC 168. GCAGTTCTGA 169. Hun mRNA fragment for class II X00700 histocompatibility antigen ______ TCCTGGGTTC 170. GGTAGAACTA 171. AGCTCCCAGA 172. Homo sapiens mRNA for ferritin L-chain. Y09188 TCCTTACTAG 17. Homo sapiens mRNA for GAIP protein. X91809 GGCCTTCCG 1 174. Human membrane-associated protein IM58285 -. ~ ~ 1- go r = ~ , . 0%&.%t WO 99/65924 PCT/US99/13800 69 TAG SEQ ID DESCRIPTION NO (HEM-I) mRNA. complete cds. ACTCAGAAGA 175. CCCAAGCTAG 176. Human clone 23827 heat shock protein ________MRNA. complete cds. CAAAATGCAA 177. GGTGGATGTG 178. TGAGAGGAGA 179. Homo sapiens NADH-ubiquinone AF047181 oxidoreductase subunit CI-SGDH ________m.RNA, AGAACAAAAC 180. Homo sapiens mRNA for proliferation- X67951 _________associated gene (pag). AGAACCTTAA 181. CAAATGCTGT 182. Homo sapiens transcription factor ISGF-3 M97935 _________mRNA, complete cds. CCCAGATGAT 183. CCTGTGTGTG 184. TGAGTCTGGC 185. CTTTTCTCTT 186. AGCAGCTGCT 187. Homo sapiens HYL tyrosine kinase X77278 ____ ____ ___ ___mRNA. CCGGGTGATG 188. TCAAAAAAAA 189. ____________ CTGTGATTGT 190. Homo sapiens FLAME- ImRNm A, AF009616 mRNA,_complete cds. AATTGCAAGC 191. Human coffin mRNA. D00682 AAAAATAAAG 192. Homo sapiens mRNA for mitochondrial X65460 associatdAT g synthase. AACTGCTTCA 193. Hs. a 1t538: Homo sapiens Arp2/3 protein RNAcomplex subu iACCACGAAT 194. Human mRNA for cathepsin H X07549 ____________(E.C.3.4.22. 16.). GAGAACGCAG 195. ___________ AGGACACCGC 196. Human .RNA for C-SRC-kinase. X59932 X71 TGGGTCATT 197. GGGCAGAATTT 198. Human mRNA for KAA37 gene, AB002368 partial cds. GGGCAAGCCA 199. Human mRNA for steroid hormone X51416 YOO _________receptor hERR I. GCCAAGGGCC 200. CTGCTAGGGG 201. ACTCCTTCCT 202. CTTTTATGTA 203. (E. CAGATTGTGA 204. CGCCCGTCGT 205.
WO 99/65924 PCTIUS99/13800 70 TAG SEQ ID DESCRIPTION V), NO ACTAACTGTG 206. Homo sapiens IEF 9306 mRNA. X71810 GAATTTTATA 207. Human peripheral benzodiazepine L21950 receptor related mRNA sequence. ATGATGCGGT 208. Homo sapiens thrombin inhibitor mRNA. Z22658 GGAAGACATC 209. GGCAGTAGGC 210. AATGAATGAA 211. TAAATCTATA 212. Homo sapiens hFcRII-C isoform mRNA X17653 for IgG Fc receptor hFcRII. TTTACAGACC 213. Homo sapiens clone 23953 mRNA AF0521 11 sequence. CTTTCAGATG 214. Human mRNA for platelet-type D25328 phosphofructokinase, complete cds. GCGTGATCCT 215. Human aldehyde reductase mRNA, J04794 complete cds. CCCTCCTGGG 216. GTGTCGGGGG 217. GGGGTAAGAA 218. Human mRNA for human homologue of D16111 rat phosphatidylethanolamine TAAACTGTTT 219. GCCCCCCCGT 220. TTGGGCACTA 221. GCTGGGGTGG 222. Homo sapiens mRNA for mediator of X84709 receptor-induced toxicity. TGCTACTGGT 223. Homo sapiens mRNA for SURF-l. Z35093 AGAGGTGGTG 224. CGGTTACTGT 225. TTCTGAAGCA 226. CAGTTTGTAC 227. Human mRNA for brain pyruvate X52709 dehydrogenase (EC 1.2.4.1). ATGTGAAGAA 228. TCTGAAAACC 229. TTAACAAACT 230. TTATTGTI'GC 231. GAAGT1TTAC 232. GTCATTATGC 233. TCTGGGAACA 234. Human clone 23828 mRNA sequence. U79285 CAGTGGGTGT 235. Homo sapiens clone 24733 mRNA AF052149 sequence. GGTTCTGTGT 236. CCCTCTGTCA 237. CCCTGGGTTC 238. Human ferritin L chain mRNA, complete MI 1147 cds. TGGGTGAGCC 239. Human cathepsin B proteinase mRNA. M14221 WO 99/65924 PCT/US99/13800 71 TAG SEQ ID DESCRIPTION I NO complete cds. CCGACGGGCG 240. CACCACGGTG 241. ATTTCAGAAG 242. Homo sapiens cellular repressor of ElA- AF084523 stimulated genes CREG mRNA, TCCGCGAGAA 243. GGGTGCTTGG 244. GACCAGAAAA 245. Human COX VIa-L mRNA for X15341 cytochrome c oxidase liver-specific GCATCCCTCC 246. GAAGATGTGG 247. TTAGTTACCT 248. TCATTGTAAT 249. CAGCGCGCCC 250. CCTGTGATCC 251. CGTGGGTGGG 252. Human mRNA for heme oxygenase. X06985 ACATCGTAGG 253. CCTGGAAGAG 254. Human thyroid hormone binding protein J02783 (p55) mRNA, complete cds. GAAAAATGGT 255. Homo sapiens mRNA for laminin- X61156 binding protein. CTCAACCCCC 256. Human mRNA for LDL-receptor related X13916 protein. TAACCAATCA 257. Human Rab5c-like protein mRNA, Ul 1293 complete cds. CTCAGTCCCC 258. Homo sapiens mRNA for ecalectin, AB005894 complete cds. CCCTGTAATA 259. TGGCGTACGG 260. CTGGCTGCAA 261. Human cytochrome c oxidase subunit Vb M19961 (coxVb) mRNA, complete cds. TGCTTGGGCA 262. Human saposin proteins A-D mRNA, M32221 complete cds. ACAGCAGCTT 263. Homo sapiens mRNA for monocyte X98306 chemoattractant protein 4. CCCTGGGGTT 264. CCAATCCTGA 265. TTCTTGCTTA 266. CCCTTAGCAA 267. ATAGGTAGAG 268. GCTGCTCCCT 269. GTGATCTCCG 270. AAGATTGGTG 271. Homo sapiens mRNA for MRP- 1. X60 111 ATGTGAAGAG 272. ,Human SPARC/osteonectin mRNA, J03040 WO 99/65924 PCTIUS99/13800 72 TAG SEQ ID DESCRIPTION--- NO _________complete cds. CGCTGTGGGG 273. ACACAGCAAG 274. CTCAGACAGT 275. CCTGGGTTCT 276. TGGGGTGAGC 277. ACTATGACAA 278. TGGACACAAG 279. AGACTGATCC 280. Homo sapiens mRNA for protein tyrosin X73568 kinase. CAATATTACA 281. CCACACCGGT 282. Human mRNA for heme oxygenase-2, D21243 complete cds. ATAGGTCAGA 283. CCATCCGCAT 284. Human lysosomal proteinase cathepsin B M13230 mRNA, 3' end. TTTCCCCGCA 285. CTTCTATGTA 286. Human mRNA for KIAAO 177 gene, D79999 partial cds. GCCCTGCTGA 287. GCGTCGGGGA 288. Homo sapiens mRNA for soluble IFN X89814 alpha/beta receptor. GGCAGAGGAC 289. Human mRNA for Nm23 protein, X17620 involved in developmental regulation CCAGTAATCC 290. TACACTACTG 291. TCTGGTCTGG 292. Human surface antigen mRNA, complete M60922 cds. CTTCTACTAA 293. TGGCACAAAC 294. TAGGCAACAC 295. GCACCTTCTG 296. GCCACTACCC 297. GACCCACCTT 298. CGCTGTGTGC 299. Human mRNA for glucocerebrosidase, D13286 complete cds. GATCT1TTGT 300. Homo sapiens mRNA for Fln29, AB007447 complete cds. TCACTGAGTT 301. ATTGTGCTTG 302. TGTAAGGCAC 303. TGAACCTCTG 304. Homo sapiens Ca2+-dependent U03090 phospholipase A2 mRNA, complete cds.
WO 99/65924 PCT/US99/13800 73 TAG SEQ ID DESCRIPTION MA sro NO CGGATAACCA 305. Human cell cycle protein p38-2G4 U59435 homolog (hG4-1) mRNA. complete GTTCAGCTGT 306. Homo sapiens porin (por) mRNA, L08666 cornplete cds and truncated cds. \AAAAACCCA 307. ATAGACATAA 308. Human pre-mRNA splicing factor M69039 SF2p32. complete sequence. CTCATAAGAA 309. CTGAGCACAA 310. TTTTGTGTGA 311. --- _- TTTTCCTGCA 312. AGTATGTATG 313. GCTGTCATCA 314. Human 26S protease (S4) regulatory L02426 subunit mRNA, complete cds. GACTGTGCCA 315. Human cytoplasmic dynein light chain I U32944 (hdlc1) mRNA, complete cds. TCACAAAAGA 316. Homo sapiens chromosome 1 beta-l.4- AF038663 galactosyltransferase mRNA, TTGCTGGAGA 317. GTCAAGACCA 318. Human beta adaptin protein mRNA, L13939 complete cds. GGAGGTGGGG 319. Homo sapiens clone 24720 epithelin 1 AF055008 and 2 mRNA, complete cds. GTGCAAAATG 320. GTGGGGGGAG 321. ACCCCAGCAA 322. CAATGTGAGC 323. CATTGTATTA 324. GCTTCCATCT 325. Homo sapiens BATI mRNA for nuclear Z37166 RNA helicase (DEAD family). ATGGGTTTGC 326. CCCAATAAAC 327. CCCTGGGCTC 328. CTGCGGTGGC 329. CATCTAAACT 330. Human mRNA for KIAA0038 gene. D26068 partial cds. TGTTTATCCT 331. Human diazepam binding inhibitor (DBI) M14200 mRNA, complete cds. ACCTCAATTA 332. Human ALASI (ALASH) mRNA for X56351 delta-aminolevulinate synthase ACTCAGGTGA 333. ACTTGAGCTT 334. Homo sapiens mRNA for cystinosin. AJ222967 AGGCTACGGG 335. GGAAGAGAAG 336. Homo sapiens mRNA for RerI protein. AJOO 1421 WO 99/65924 PCT/US99/13800 74 TAG SEQ ID DESCRIPTION NO TTTTATTAAA 337. ATACTGTCAG 338. CTTCTGCTGG 339. Homo sapiens retinal short-chain 1AF061741 dehydrogenase/reductase retSDR I TTACGAGGAA 340. Homo sapiens clone 24761 mRNA AF052155 sequence. AATCCAGGAG 341. TGGGCCTGTG 342. ATAGTAGCTT 343. Human actin bundling protein (HSN) U03057 mRNA, complete cds. TGGAAGGACC 344. AATAGAAATT 345. Homo sapiens clone 23810 osteopontin AF052124 mRNA, complete cds. TTGAGCCAGC 346. Human KH type splicing regulatory U94832 protein KSRP mRNA, complete cds. TTGGCCAGGT 347. TACTGTGATG 348. ATCTTGAAAG 349. Homo sapiens NAP (nucleosome M86667 assembly protein) mRNA, complete cds. TTAGCAATAA 350. CTGTGCGGAA 351. CCTTTGTAAG 352. ACACTACGGG 353. Homo sapiens clone CIR2 cell AF049672 immortalization-related mRNA sequence. GACGTCTTAA 354. Human mRNA for proteasome subunit D00763 HC9. TGAGGCCTCT 355. GGAGGGATCA 356. Homo sapiens integrin-linked kinase U40282 (ILK) mRNA, complete cds. GGCCCCATTT 357. Human carbonyl reductase mRNA, J04056 X51 complete cds. GGCTTTAGGG 358. GGGCCAATAA 359. Homo sapiens full length insert cDNA AF075046 YN68C05. TGAACCAAGG 360. TTTACAGCTG 361. Human diacylglycerol kinase zeta U94905 mRNA, alternatively spliced, ACTCTGCTCG 362. AGAAAGAAGG 363. GAGCAAATGT 364. AGAAGTATAG 365. Human mRNA for proteasome subunit X, D2901 I complete cds. TTTTTAATGT 366. Human H3.3 histone, class B mRNA, M11354 complete cds.
WO 99/65924 PCT/US99/13800 75 TAG SEQ ID DESCRIPTION NO AGGCTGCCT 367. Human mRNA for KIAA0249 gene. D87436 complete cds. CCATTCTCCT 368. CCCGGTGTGT 369. TTGGTTTTGT 370. GAAGTGTGTC 371. AGCCTAGGTC 372. CTTCTGTCTC 373. GCATAGTGTT 374. AGAACCAAAA 375. AAGAATCTGA 376. Homo sapiens CI-MNLL homolog gene AF054181 mRNA. complete cds. CTACAATAAA 377. CCGAGTTTTT 378. ATTTATTTAT 379. TTCCTCCACG 380. PKC- eta b=protein kinase C eta b S74620 [human, monoblastoid U937 cells, AGCACTTACA 381. Human mRNA for lipoprotein apoCIL. X00568 TTCCAAACCT 382. Human mRNA for phospholipase C. X14034 GGCCAGTAAC 383. GGCTGTACCC 384. Human cysteine-rich peptide mRNA, M33146 -complete cds. GGCCAGGTGG 385. Human mRNA for KIAA0047 gene, D38554 partial cds. GGGAAACCCT 386. Human replication factor C, 40-kDa M87338 subunit (Al) mRNA, complete cds. TGGTGGGTGT 387. GTGGGCCGCT 388. Homo sapiens heat shock protein 75 AF043254 (hsp75) mRNA, partial cds. GTCACTGCCT 389. Homo sapiens mRNA for Ribosomal AJO10119 protein kinase B (RSK-B). TACCAGTGTA 390. Human chaperonin (HSP60) mRNA, M34664 complete cds. TGGAGGGGCC 391. Homo sapiens mRNA for CLPP. Z50853 GCCTTTCTAA 392. Homo sapiens ribosomal protein S6 L07597 kinase 2 (RPS6KA2) mRNA, complete GTAGCATAAA 393. GGAACACACA 394. GAACCGTCCT 395. GTCTGGGGGA 396. Human lysophospholipase homolog (HU- U67963 K5) mRNA. complete cds. GTCTAGTCAA 397. Human mRNA for KIAA0179 gene, D8000 1 _partial cds. CACTCCTACA 398.
WO 99/65924 PCT/US99/13800 76 TAG SEQ ID DESCRIPTION NO TTTGCTCTCC 399. Human vinculin mRNA. complete cds. 1M33308 GTGTACCGGA 400. Homo sapiens mRNA for Arno protein. X99753 TAGACAATGC 401. Homo sapiens clone 23674 mRNA AF038183 sequence. TTAGATAAGC 402. Human chaperonin-like protein (HTR3) M94083 mRNA. complete cds. GTCAACTGCT 403. CAGCTCCAAA 404. TGGGCCTGGC 405. AATGAAAAGG 406. Homo sapiens testis enhanced gene AF033095 transcript protein (TEGT) mRNA., TATGTTGCTG 407. TCTTCAGGAG 408. GTTGTCCATT 409. GAAGGTGGGG 410. GCGGCGATCG 411. CCGAAGGGTC 412. GCGAAATCCT 413. TGATGCTGAT 414. GTTCACATAA 415. GATTCAAGTC 416. Homo sapiens mRNA for mitochondrial Y1 1681 ribosomal protein S12. GATCTCATCT 417. TGGAGCGATT 418. TCCCCCCCCC 419. GAGGGCCGGT 420. AGAGAAATTT 421. GAACCCTGGG 422. TGGGTGGGCA 423. CGGGTAGTAT 424. Homo sapiens GAA mRNA for Y00839 lysosomal alpha-glucosidase (acid maltase). TGTGTGTTTG 425. TTGGGTTAAT 426. TTGTTTGTAA 427. TTTGCAATAA 428. TGACCGAACA 429. GAGTGAGTGA 430. CTTCTCAGGG 431. GGGTATCCCT 432. Homo sapiens RINGI gene. Z14000 CCCTGAGTTC 433. AATATGCTTT 434. Homo sapiens mRNA for vacuolar H- X76228 ATPase E subunit.
WO 99/65924 PCT/US99/13800 77 TAG SEQ ID DESCRIPTION NO CCGCGTCCCT 435. 1Human peroxisome proliferator activated L07592 receptor mRNA, complete AAATAAAAAG 436. Homo sapiens full length insert cDNA AF085929 clone YR5IB02. CCACCTGCTT 437. CCACACACCG 438. ATACATTTAG 439. Homo sapiens mRNA for ClI protein. X81625 GGGATCAAGG 440. AATGCCGCAG 441. TCAGTTATCT 442. AAGGCAGGGC 443. AAACAGTAGT 444. CCCCATTCTT 445. GGGCGAGAGA 446. AATATCTGAC 447. Human guanine nucleotide regulatory UO 1147 protein (ABR) mRNA, complete TGATGTCCAC 448. GGGAGTAATA 449. Human protein tyrosine phosphatase M34668 (PTPase-alpha) mRNA. TCAGCTGGGG 450. CCCCAGCCAG 451. Human XPlPO ribosomal protein S3 U14990 (rpS3) mRNA, complete cds. TGGCCCTCCA 452. Hs.756 10: Human transcription factor IL 4 Stat mRN TAATAAACAG 453. Human putative 32kDa heart protein U47674 PHP32 mRNA, complete cds. TTCATTATAA 454. Human prothymosin alpha mRNA (ProT- M26708 alpha), complete cds. GTGTGTTTGT 455. Human transforming growth factor-beta M77349 induced gene product (BIGH3) TTTTGGGGGC 456. Hs.7476: Human mRNA for proton ATPase-like protein TCTCT T C 457. Homo sapiens tissue specific mRNA. X67698 CCCACACTAC 458. Human signal-transducing guanine M16538 J02 nucleotide-binding regulatory (G) TTCACAAAGG 459. Homo sapiens mRNA for macropain X61970 subunit zeta. GTGCCTAGGA 460. GGGAAACCCC 461. Human fibroblast mRNA fragment with X05126 Alu sequence (pRHF 11). CTGACTGTCC 462. Human major histocompatibility class II KO 1144 antigen gamma chain mRNA, GGGGACTGAA 463. Homo sapiens mRNA for low molecular D50369 I mass ubiquinone-binding WO 99/65924 PCT/US99/13800 78 TAG SEQ ID DESCRIPTION t ssr0 NO TACTCTTGGC 464. Human mRNA for novel heterogeneous X16135 GCACTACTCG 465. ACTGAAGGCG 466. Human metargidin precursor mRNA. U41767 ___________ ________complete cds. CTGGCGCCGA 467. GACTATAGCG 468. AGGAGCTGCT 469. Human mitochondrial NADH U65579 dehydrogenase-ubiquinone Fe-S protein _____ _____8, AAGGCCGAGT 470. ACGTTTAAGG 471. Homo sapiens ash mRNA. X62852 CCTCCAGCAG 472. ACTGGTAAAA 473. Homo sapiens F IFo-ATPase synthase f AF047436 subunit mnRNA, complete cds. AACTACATAG 474. TGCCAGCTAA 475. TACGAGGCCG 476. Homo sapiens mRNA for LAK-4p, AB002405 complete cds. CTAGCTTTTA 477. GCTTTCTCAC 478. _____________ GCTGGCTGGC 479. Homo sapiens chaperonin containing t- AF026292 complex polypeptide 1, eta GATCCCAACT 480. Human mRNA for metallothionein from V00594 cadmium-treated cells. GGGCCTGTGC 481. Homo sapiens monocarboxylate U81800 transporter (MCT3) mRNA, complete _____ ____ ____cds. TGAGGGAATA 482. Human triosephosphate isomerase M10036 M10 mRNA. complete cds. TCTCTCAAAG 483. Human cell surface antigen (CD53) M60871 mRNA, complete cds. GGAATGTACG 484. Human mitochondrial ATP synthase U098 13 subunit 9, P3 gene copy, mRNA, AGAACCTTCA 485. TTCTTGTTTT 486. Homo sapiens mRNA for prion protein. DOOO 15 NOO complete cds. ACTCCAAAAA 487. Hs.3655: Human insulinoma rig-analog mRNA encoding TCTCAGATGA 488. Homo sapiens CYP 27 mRNA for 59812 vitamin D3 25-hydroxylase. TGGCTGGGAA 489. 1 TTCTTGTTTT 4860. Human mRNA for calmodulin. complete D05N cds. cds.
WO 99/65924 PCT/US99/13800 79 TAG SEQ ID DESCRIPTION NO TGTTCATCAT 491. AAAACATTCT 492. CTAAAAAAAA 493. Human 26-kDa cell surface protein M33680 TAPA-1 mRNA, complete cds. AGATGTGTGG 494. Human mRNA for mitochondrial 3- D16481 ketoacvl-CoA thiolase beta-subunit ACGTGGTGAT 495. Homo sapiens full length insert cDNA AF086483 clone ZD92G09. CTGCCAACTT 496. Human cofilin mRNA, partial cds. JU21909 TGCTGCCTGT 497. Homo sapiens HCG IV mRNA. X81005 CGTGAGCCAC 498. TCTTGTGCAT 499. Human mRNA for lactate dehydrogenase- X02152 A (LDH-A. EC 1.1.1.27). AGCAAACTGA 500. TGGAAACAAA 501. TGTGACCCCT 502. Human ATP:D-hexose 6- U42303 phosphotransferase mRNA, partial cds. ATGTTCCTAT 503. CTTCTCACCG 504. Homo sapiens mRNA for ubiquitin- AJ002385 conjugating enzyme UBC9. AGGTGTGTCA 505. CTTCAGAAAT 506. GGTCAGTCGG 507. TGTAGGTCAT 508. Homo sapiens full length insert cDNA AF086432 clone ZD79H11. TACAGAGGGA 509. Homo sapiens zinc finger protein 216 AF062346 splice variant 1 (ZNF216) TCAAATGCAT 510. Human nuclear ribonucleoprotein particle M16342 (hnRNP) C protein mRNA, CCCAGGGAGA 511. Homo sapiens chaperonin containing t- AF026291 complex polypeptide 1, delta GCCAGACACC 512. ATCACAGTGT 513. Human nuclear-encoded mitochondrial Li 1932 serine hydroxymethyltransferase ATGGCTAAGC 514. GTAGGAGCTG 515. Human retinal protein (HRG4) mRNA, U40998 complete cds. AGTCTGATGT 516. ATCGCTTTCT 517. amyloid protein precursor {3' region. S41242 alternative polyadenylation, GTGGCACGTG 518. Human clone AZAL Alu repeat sequence. U02044 TTGGGGAAAC 519. Homo sapiens mRNA for biliverdin IX X93086 alpha reductase. CCTTGGGTTC 520.
WO 99/65924 PCT/US99/13800 80 TAG SEQ ID DESCRIPTION 1 'SI~ON NO GCAAAGAAAA 521. Human breast tumor autoantigen mRNA, U24576 complete sequence. TAAATAATGT 522. Human Grb2-associated binder-I mRNA, U43885 complete cds. GGGAGGATTA 523. Homo sapiens Tat-interacting protein AF039103 ________ TIP30 mRNA. complete cds. CACCCCTGAT 524. Human creatine kinase-B mRNA, M16364 complete cds. GAGTGGGGGC 525. GTAATTACTG 526. CGGCCACAGA 527. Human HepG2 partial cDNA, clone D16990 hmd2c12m5. AGAATATCAG 528. CCATTAACAC 529. CAACTTAGTT 530. Homo sapiens mRNA for myosin D50372 ATGCCGGA_31 regulatory light chain, complete cds. ATGCCCGTGA 531. AGAGCAAGTA 532. GTTGGTCTGT 533. TGGAGCAGTT 534. TTACCTCCTT 535. ATCAAGTTCG 536. GAACACCGTC 537. CCAAAAAAAA 538. Human interferon-induced leucine zipper U72882 protein (1FP35) rnRNA, CACAGAGTCC 539. Human alpha-2-macroglobulin receptor- M63959 associated protein mRNA, CCTGATGACC 540. TATTACTGGG 541. TGGCACTTCA 542. Human low-Mr GTP-binding protein U59878 ________(RAB32) mRNA, partial cds. CGACCGTGGC 543. CTCTCACCCT 544. Human mRNA for X13973 _________ribonuclease/angiogenin inhibitor (RA).______ GGTCCAGTGT 545. Homo sapiens phosphoglycerate mutase J04173 _______________(PGAM-B) mRNA, complete cds.______ GTGGCAGGCA 546. clone 4-3 {Alu sequences, splice acceptor S94541 sites)} [human. Pre-mRNA, ATTGTTTATG 547. Human non-histone chromosomal protein M12623 ________HMG- 17 mRNA, complete cds. GCCTGCTGGG 548. Homo sapiens GPx-4 mRNA for X71973 phospholipid hdroperoxide glutathione TTCATACACC 549. Tag matches mitochondrial DNA AAGGAAGATC 550. Human glutathione-S-transferase U90313 ribonuclease/angiogenin...r. inhbir (RAI) WO 99/65924 PCT/US99/13800 81 TAG SEQ ID DESCRIPTION ACCESSION NO homolog mRNA, complete cds. TGCAGGCCTG 551. Homo sapiens mRNA for IFN-inducible X59892 gamma2 protein. TTGTAATCGT 552. Human mRNA for ornithine D87914 decarboxylase antizyme. complete cds. CCTCTCCAAC 553. Human HLA-DMB mRNA. complete U15085 cds. ATGAGCTGAC 554. Homo sapiens cystatin B mRNA, L03558 complete cds. CTGCTAACCC 555. AGGTACTGAG 556. TGCTGTGTGC 557. Homo sapiens 15 kDa selenoprotein AF051894 mRNA, complete cds. TCAGTTTGTC 558. Human HS1 binding protein HAX-1 U68566 mRNA, nuclear gene encoding TGCTGAATCA 559. GCCCAGCAGG 560. CTGTGCATTT 561. Human 54 kDa protein mRNA, complete U02493 cds. ATGGTCTACG 562. TGAGCCTCGT 563. TGGGCCAAAC 564. ACATTTTTAA 565. AGAGGCAACC 566. AGGCAGCGAG 567. immunoglobulin epsilon chain constant S55271 region=membrane-bound form CCTGAGGGTA 568. GGTTAACGTG 569. TTTCAATAGA 570. GGTCACACTA 571. CGGCCCAACG 572. Homo sapiens mRNA for arginine Y10805 methyltransferase, splice variant, 1435 TGTGCTAATA 573. TSEI=protein kinase A regulatory S54711 __________ subunit gene [human, niRNA Partial, ____ CTGTGCAAGT 574. TTATGGGGAG 575. Human transformation-sensitive protein M86752 (IEF SSP 3521) mRNA, GTCTTTCTTG 576. GAACGCCTAA 577. Human mRNA for dihydropyrimidinase D78013 _________related protein-2, complete cds. GGGGGGTGGA 578. GCAGGTCAGC 579. Human branched chain alpha-keto acid J04474 ______________ ehydrogenase mRNA. 3' end.______ GATCATCAAG 580. Homo sapiens mRNA for monocyte 1Y10802 WO 99/65924 PCT/US99/13800 82 TAG SEQ ID DESCRIPTION ACCESSION NO chemotactic protein 2. TCGTTACGCA 581. CGCCTATAAT 582. ATCCTCTGCG 583. Homo sapiens cam kinase I mRNA. L41816 complete cds. GGCTTTGGAG 584. Homo sapiens partial mRNA: ID LG141- AJ227879 7B2. TATTTACTCT 585. TATAGGCCGA 586. TACACACACG 587. TCCTCCCTCC 588. Human mRNA for proteasome subunit D26599 HsC7-1, complete cds. TTAATTGGGA 589. Homo sapiens mRNA for novel Z35491 glucocorticoid receptor-associated GACCTATCTC 590. AGAATCACTT 591. AGGAAAAGAT 592. Human 1.1 kb mRNA upregulated in U09196 retinoic acid treated HL-60 AATGAATGTT 593. AATTCTGTAA 594. TTTTCTGCTG 595. GTGACGTGCA 596. GAGGCCACCC 597. TACTAATAAA 598. ATTAACAAAG 599. Human mRNA for coupling protein G(s) X04409 alpha-subunit (alpha-S 1) TCCTGCCCCA 600. Human parathymosin mRNA, complete M24398 cds. TCTGACAAAC 601. TGAATATACT 602. TGAGGCAGGG 603. Human syntaxin 5 mRNA. complete cds. U26648 TGATGATGTT 604. TATATCAGTG 605. TATCACTCTG 606. Human male-enhanced antigen mRNA M27937 (Mea), complete cds. ACGTCGTGTG 607. ACTATTCCAT 608. GGTCCCCTAC 609. TGGCCTAATA 610. TTGAATATTA 611. GGGGACACAG 612. TTTAGGGGGA 613. TTTTACCAGT 614. Homo sapiens reticulocyte p1Cln mRNA. AF005422 complete cds.
WO 99/65924 PCTIUS99/13800 83 TAG SEQ ID DESCRIPTION ACCESSION NO TAGCCAGTTA 615. TCCGTGTGTC 616. GTAAAACCCT 617. GTCAGGTTGA 618. Homo sapiens GOS28/P28 protein AF047438 mRNA, complete cds. GTTTGAAGGG 619. TAGAAGATGC 620. GCAAGGGCTA 621. GAAGATTGAG 622. Human signal transducer and activator of U43 185 transcription Stat5A mRNA. TCACAGACTG 623. CCTGAAGAGG 624. GGGAGCCCGG 625. Homo sapiens herpesvirus entry protein B AF058448 _ __ (HVEB) mRNA, complete cds. CTTAGAGCCC 626. Human thioredoxin mRNA, nuclear gene U78678 encoding mitochondrial TGGTTTGAGC 627. CTACCAGGAA 628. AACCTGGCCT 629. CTGACCTGGG 630. TGTGGCCTGC 631. Human glucose-6-phosphate M35604 dehydrogenase (G6PD) mRNA, 3' end. CAACGTCCTG 633. Homo sapiens full length insert cDNA AF075060 Y073E04. AGAAAGTGTC 634. ATTCAGCACC 635. TGGTGTTGAA 636. CAGGGGAGTG 637. Homo sapiens anpg mRNA. X56528 CAGTCTGGGA 638. Homo sapiens mRNA for IL13 receptor Y09328 alpha-I chain. ACAAAGTTAC 639. AGAGCCCTAG 640. Homo sapiens COX17 mRNA, complete L77701 cds. TGTGCCCTGA 641. Homo sapiens clone 24772 BDP-1 AF070616 _protein mRNA, partial cds. TTGGAGATCT 642. Human NADH:ubiquinone U94586 oxidoreductase MLRQ subunit mRNA. complete CTGCTCATCC 643. Human aldehyde dehydrogenase ALDH7 U10868 mRNA, complete cds. GATCAATGGA 644. Homo sapiens oscillin (hLn) mRNA, AF029914 complete cds. CCTGTCCTTT 645. Homo sapiens OkD protein (BC 10) AF053470 WO 99/65924 PCT/US99/13800 84 TAG SEQ ID DESCRIPTION 'ACCESSION NO mRNA. complete cds. CAAAAGGCTC 646. CGGAAAAGGA 647. GAGCAGTGCT 648. Homo sapiens RNA for c-fes. X52192 GCTACCCAAC 649. AGCACTGCTG 650. AGAGCAAACC 651. Homo sapiens lysyl hydroxylase (partial M98252 clone 2.2 Kb LH) RNA, ACTGGAGCCA 652. ACAGAAGGGA 653. Human beta-ID integrin mRNA, U28252 cytoplasmic domain, partial cds. AAGCTAATAA 654. Human prostaglandin endoperoxide M59979 synthase mRNA, complete cds. AAGGAAAGGC 655. Human manic fringe precursor mRNA, U94352 complete cds. ACCTTGTGCC 656. Human L-iditol-2 dehydrogenase mRNA, L29008 complete cds. CTTGTGTTAT 657. GCTATGAGAA 658. Human binding protein mRNA. 3'end. L23113 CTGGGTGAAG 659. GCGGGAGGGC 660. GATCCCAACA 661. Human mRNA for F1-ATPase beta X03559 subunit (F-I beta). TATATATGGG 662. GGAGCTTAGA 663. AGCTGTCCCC 664. AAGATCCAAA 665. AAGTAGAAAG 666. Homo sapiens ATF family member ATF6 AF005887 (ATF6) mRNA, complete cds. CCTTGACCAA 667. GGGGATGGGG 668. TTAAAAGTCA 669. GGTGGTGGCA 670. AACTGTGTTT 671. AAAACTGCGT 672. AGGGCTTTCA 673. GGAGCCAGGC 674. Homo sapiens GSTT I mRNA. X79389 CAACACTGTG 675. GGCAGTTAAC 676. GGGATGGAGA 677. GGAAGGGGGA 678. TCTGTTGGAC 679. TTGGATATCC 680. AAGATAATAA 681.
WO 99/65924 PCT/US99/13800 85 TAG SEQ ID DESCRIPTION NO GTAAAGCCTA 682. GTCAGATGTC 683.
~---
GTCCATCATA 684. -- GTGGAGCGGA 685. -- GTGGGTCAGC 686. ~ TTGAAACCCC 687. TAACAGAAAG 688. Homo sapiens GLI-Krupple related M77698 protein (YYl) mRNA. complete cds. GGGCCCCAAA 689. CGTGTTAATG 690. Homo sapiens sterol regulatory element- M28372 binding protein (CNBP) mRNA, GGGATGGCAG 691. Human G7a mRNA for valyl-tRNA X59303 synthetase. TGGAACCAGA 692. Homo sapiens clone 24751 unknown AF070530 mRNA. CCCTGGGTCC 693. CCAATGCACT 694. CATTCCAGAG 695. CAGCAAAAAA 696. pyruvate carboxylase [human, kidney, S72370 mRNA, 4017 nt]. CACITTGGG 697. Homo sapiens MLN50 mRNA. X82456 CACCAGGACA 698. CGGAGCCGGC 699. GTGTGAGTGT 700. TCTTCTGCCA 701. TGAGCCCGGC 702. TTGTAAAAGG 703. TGCCTTAATG 704. Homo sapiens putative tumor suppressor AF061836 protein (RDA32) mRNA, TTTACATATA 705. Homo sapiens mRNA for TIP3, complete AB000734 cds. TGGGTAAGCC 706. TGGT111GG 707. TCTCCACGAA 708. Homo sapiens Arp2/3 protein complex AF006087 subunit p20-Arc (ARC20) mRNA, TTAAAGATGG 709. TATCTATCAA 710. TTATATTGCC 711. TTCTTCTCGT 712. Homo sapiens mRNA for SMT3A X99584 protein. TTCTTCTGAA 713. GGAAGAGGGT 714. CCTATGTAAG 715. Homo sapiens mRNA gene for hnRNP G Z23064 protein. SUBSTITUTE SHEET (RULE 26) WO 99/65924 PCT/US99/13800 86 TAG SEQ ID DESCRIPTION 'AtTSTON NO TGAGGCCAGG 716. Human high mobility group box (SSRPl) M86737 mRNA. complete cds. ATCCCCCTGG 717. Homo sapiens clone 23610 mRNA AF052125 sequence. ATTGTGAACA 718. Homo sapiens calcyclin binding protein AF057356 mRNA. complete cds. TTATAAAAGA 719. ATCTTGGTAC 720. AGGGGGCAAA 721. Homo sapiens apolipoprotein L mRNA. AFO 19225 complete cds. AATCTGCGCC 722. Human interferon-induced 17-kDa/15- M13755 kDa protein mRNA, complete cds. ATACATACTG 723. AGGTGCCTCG 724. AGGTCCCTGT 725. AGGGTGGGGG 726. TGTGCTCGGG 727. Human mRNA for KIAA0088 gene. D42041 partial cds. GTGGTACAGG 728. Homo sapiens microtubule-based motor AF004426 (HsKIFC3) mRNA, complete cds. GCAAAAAAAA 729. Homo sapiens aortic carboxypeptidase- AF053944 like protein ACLP mRNA, TACCCCACCT 730. CAGTTCTCTG 731. AAAAATGGTG 732. GTTGGGACAT 733. CATTTCATAA 734. Human mitochondrial ATPase coupling M37104 factor 6 subunit (ATP5A) mRNA, AATGAAAATA 735. Homo sapiens breast cancer antiestrogen U92715 resistance 3 protein ACAGGCAGAA 736. Human tumor necrosis factor type 2 U12597 receptor associated protein ACCCTGCCTC 737. CATTGAAGGG 738. Homo sapiens clone 24433 AF070539 myelodysplasia/myeloid leukemia factor 2 ACGAGCTGGA 739. Human gene similar to Z.mays ras-like AL022729 (X63277) and Homo sapiens RAYl ACTTCCTCCT 740. AGGTGGAGGT 741. AGAACCTTTG 742. ACATATCTGG 743. AGGGTTTGCC 744. Human mRNA for HLA-A*0218, D83515 complete cds. SUBSTITUTE SHEET (RULE 26) WO 99/65924 PCT/US99/13800 87 TAG SEQ ID DESCRIPTION NO TCTTTCCAGA 745. Homo sapiens hPTPA mRNA. X73478 ATGTTTACAC 746. Human pre-T/NK cell associated protein L17329 (5A3) mRNA. CCATATACAT 747. AGTAGGTGGC 748. AGTGAGGATA 749. AGTTTTACAA 750. Homo sapiens 26S proteasome ATPase AF038965 subunit mRNA, complete cds. ATAGATGGGG 751. AGGAAAGCCA 752. Homo sapiens mRNA for Rab9 effector Z97074 p40, complete cds. ATCAAGGGGT 753. AGCCCAGGAG 754. ATGCTAGAAA 755. AGGGTGTTTT 756. Homo sapiens mRNA for MNB protein D85759 kinase. complete cds. ATTAGCAGAG 757. ATTGGAGATG 758. Homo sapiens mRNA for L-3- Y10275 phosphoserine phosphatase. CAGAGTGACT 759. Human IEF SSP 9502 mRNA, complete L07758 cds. CAGTGGGTGG 760. Human mRNA for UDP-galactose D87989 transporter related isozyme 1, AGCTGGTTTC 761. Homo sapiens Pig8 (PIG8) mRNA, AF010313 complete cds. CATTGCAGGA 762. ATATGTCAGG 763. GAGAACCGTA 764. GAGGGTTCCA 765. GATTGGTATG 766. GCCAAGACAC 767. CCCGTAATCC 768. AGAACTGGAA 769. GAATCCAACT 770. GCCCGGCTTC 771. CCTCTGGCAG 772. GCCCAGGGAA 773. CCCTCTTTGG 774. CCTTTCTGCT 775. CGAAAAAAAA 776. CGGCTCAAGT 777. CGGTTCATTG 778. CTCCATTGCC 779. CTCCTGGAAC 780. 1 SUBSTITUTE SHEET (RULE 26) WO 99/65924 PCT/US99/13800 88 TAG SEQ ID DESCRIPTION NO CTGCTAAGGT 781. GTTCTGGGTC 782. AGCGTTTCTG 783. ATCCGGGGAG 784. Homo sapiens RCL (Rcl) mRNA, AF040105 complete cds. CTGACCCGTG 785. CTGAGAGATT 786. AGGGATGGCC 787. Human putative T1/ST2 receptor binding U41804 protein precursor mRNA, GAGGGTATAC 788. Human mRNA for transcription factor X51330 TFE3 (partial). GACAGAGAAC 789. GAATGAGGAC 790. Human mRNA for reticulocalbin, D42073 complete cds. CTGTTAATAA 791. CTTTGATCAG 792. CTTTTAAAAT 793. Homo sapiens mRNA for cytochrome c. D00265 partial cds. GAAACTGAAG 794. Homo sapiens nitrilase I (NITI) mRNA. AF069987 complete cds. GAAGTCATTT 795. Homo sapiens full length insert cDNA AF086095 clone YZ88A07. CTGCAAGCGG 796. AGACAATGTG 797. ACTTGATTCA 798. Human mRNA for KIAAO 168 gene, D79990 complete cds. GCTTAATGTT 799. Human kidney mRNA for catalase. IX04076 GGGGGTCGGG 800. Homo sapiens mRNA for protein kinase. X85545 PKX1. AACTGTATAC 801. Homo sapiens TAP2E mRNA, complete Z22936 CDS. AACAATGTCA 802. TAAAAGACAA 803. GGCTCCTTGA 804. GGCCATCTCT 805. GGCTGCCCTT 806. AAGCGCTCTC 807. GCTGGCTGGG 808. TAAGGTAGAG 809. GGAGGAGCTG 810. GCTTTGCAGC 811. Human Src-like adapter protein mRNA. U44403 complete cds. GTATTGGCCT 812. TATCCTGGCT 813. SUBSTITUTE SHEET (RULE 26) WO 99/65924 PCTIUS99/13800 89 TAG SEQ ID DESCRIPTION ACCESSION I NO AAAGTGAAGA 814. GGGACGGCGC 815. GCTACCTTCT 816. GCTGCACCGG 817. AATCCCCATC 818. TAGCAATCAG 819. TAGACTTCCT 820. TAGACCCCTT 821. Human endogenous retrovirus type C M74509 oncovirus sequence. TACTGGAAGT 822. TAATAAATGC 823. Homo sapiens clone 24519 unknown AF055000 mRNA, partial cds. GTACTGTATG 824. Homo sapiens importin beta subunit L38951 mRNA, complete cds. TAACTGCCTA 825. AAGAGGAGAT 826. Human usf mRNA for late upstream X55666 transcription factor. ACCGGGGTGA 827. GGGTAATGTG 828. GTTGGACCAG 829. ACTCACCTTA 830. AAGCTTTGAG 831. GTTCACATTG 832. GTGGCACGCA 833. Homo sapiens partial mRNA; ID YG81- AJ227871 2A. GTGCTGATGA 834. TAGGAGAATC 835. Human vitamin D receptor mRNA, J03258 complete cds. GTAGCAAAAA 836. TGGGGTGGAG 837. Homo sapiens mRNA for AJO01838 maleylacetoacetate isomerase. GGGCCCCGCA 838. Human mRNA for KIAAO 123 gene, D50913 partial cds. TTCTCATAGG 839. TGACCTTACC 840. TATCCTGGTA 841. Human isolate 7 clone 10 from Graves' U09903 orbital muscle tissue, TATTTCGTAC 842. TCACTGATGG 843. TCCAAGGAAG 844. Homo sapiens DBI-related protein AF069301 mRNA, complete cds. TTTGGGGGCC 845. TCTGTAAGGG 846. Human mRNA for KIAAO 129 gene, D50919 complete cds. SUBSTITUTE SHEET (RULE 26) WO 99/65924 PCT/US99/13800 90 TAG SEQ ID DESCRIPTION d'EFSSON NO TTTCTAAACC 847. TCTTTTCAAA 848. TGGGCGCCTT 849. Human uroporphyrinogen decarboxylase M14016 mRNA. complete cds. TGATCAAAAA 850. [TGATGCGCGC 851. [TGCCTGCTCC 852. TGCTTGTCAA 853. Human butyrophilin (BTF5) mRNA. U9055 complete cds. JTGGGAAAGGG 854. TCCGCCGCCC 855. TTATGTAAAA 856. GGAGGACTCC 857. TTGATGCCCT 858. TGGGTAGCCA 859. GGAAGAGCAC 860. Homo sapiens mRNA for Gal-beta(1-3.1- X74570 4)GlcNAc TGGTTCCAAA 861. AAAGACCAAA 862. TGTGTTTGAA 863. TTTCATAAA 864. TTACAGTTAA 865. TAGCATTTTA 866. Human mRNA for KIAA0102 gene, D14658 complete cds. TTGACCGGAG 867. GCTTTCTCAA 868. TGGGAGAAGT 869. TTGCTTGTCC 870. TTGTTTAATT 871. Human capping protein alpha mRNA. U03851 partial cds. TTTATTGCAC 872. Homo sapiens mRNA for KIAA0676 ABO14576 protein, partial cds. TTAAGACTTC 873. GAACTTTTAG 874. CTCACCGCCC 875. Human cellular retinoic acid-binding M68867 protein II (CRABP) mRNA, CTAGATTCGG 876. CTACAAGAAG 877. | CGTGGAGTGG 878. CGGGCAACGT 879. Homo sapiens mRNA for rab Y08200 geranylgeranyl transferase, CGGAGGTGGG 880. Human mRNA for KIAA0 163 gene, D79985 complete cds. SUBSTITUTE SHEET (RULE 26) WO 99/65924 PCT/US99/13800 91 TAG SEQ ID DESCRIPTION CTSS[ON NO CTCATCTGAG 881. Human E2 ubiquitin conjugating enzyme U39318 UbcH5C (UBCH5C) mRNA. AAAAAGCTGG 882. GAGAGCAGAAI 883. ATGGCAAAGA 884. AAACTGTGGT 885. AATGTCATTG 886. ACCTGTAATT 887. ACGGAACAGG 888. ACTGGCCGAA 889. ATTACAAAAG 890. ACAAGGGTGA 891. AAATGTGTAA 892. AAAATGCTGA 893. ACCCCTGTTA 894. AAACTATTTG 895. Human rab2 mRNA, YPT 1-related and X12953 member of ras family. ACCCCATCGA 896. AAAGCAGTTT 897. AAAGTGAAAA 898. AAAGTTGCTA 899. AATACACAGA 900. AAATGGCTAA 901. AACTACCAAA 902. ACTGTTTGTT 903. Human mRNA for HLA-D class II X03067 antigen DPW2 beta chain. GAGAAGTTAC 904. AAACCTCAGG 905. ACTGTGGTCA 906. CTCAGCAGGA 907. CGAGTFT1 908. CTCCTGCCTT 909. CTCGGCCAGA 910. Human mRNA fragment for X00 199 apolipoprotein E (apo E). CTCGTCCGGA 911. CTCTCAGGGG 912. CTGCCTCCGT 913. CTGCTGCACT 914. ACCCTCTGTG 915. ACCAAGAGCA 916. CTGTTTCAGA 917. AATACACATC 918. AATATGGGTG 919. Human tetratricopeptide repeat protein U46571 (tpr2) mRNA. complete cds. SUBSTITUTE SHEET (RULE 26) WO 99/65924 PCT/US99/13800 92 TAG SEQ ID DESCRIPTION ACCESSION NO AATATGGTTT 920. AATCTTGCAA 921. AATGCTGGCA 922. Homo sapiens mRNA for MSJ-1, ABO 14888 complete cds. AAGGGCCGGT 923. Homo sapiens mRNA for AJ008244 immunoglobulin heavy chain. VHDJH ACATTTCAAT 924. AAGGATGCGG 925. CTCAGCAAAA 926. CAGTTACAAA 927. ATTGGTTATG 928. ATTGATCAAT 929. ATTCTGCCCA 930. GATAGGATAA 931. CACAAACCGG 932. CCACTGCAAT 933. CCTTTGTAAA 934. CCTTTAATCC 935. Homo sapiens (xs88) mRNA, 318bp. Z36845 CCTCTCTCCT 936. Homo sapiens Staf5o mRNA. X82200 CCGCCTTAAT 937. CCGAACACGG 938. CCAGAAAGAA 939. Homo sapiens TIMP3 mRNA for tissue X76227 CCCAGCCCA 940 inhibitor of metalloproteinases-3. CCCAGCCTCA 940. CAAATCCAAA 941. CAGTTAGTAA 942. CCACTCTGGC 943. Homo sapiens mRNA for processing a- X87237 glucosidase 1. CCACGCACCA 944. CCAAGGACTC 945. Human SUPT4H mRNA, complete cds. U38817 CAIT-1TCCC 946. CATTCATTGG 947. CATCAGGATA 948. ATGTACTAAA 949. Homo sapiens mRNA for TFG protein. Y07968 CCCACTGCCC 950. AACTCTGTAA 951. TGGCCAAAAA 952. CCCTATAAGC 953. CGAGAGCTGC 954. GACCCTGGGG 955. GACATAAATC 956. Human mRNA for KIAAO 113 gene, D30755 partial cds.______ GACACCTCCT 957.A CTGGAAGCTC 958. SUBSTITUTE SHEET (RULE 26) WO 99/65924 PCT/US99/13800 93 TAG SEQ ID DESCRIPTION ACCESSION NO GAATCACTGC 959. Homo sapiens ribosomal protein L33-like AF047440 protein mRNA. complete cds. CTGGGGGTCT 960. GAACCCTTCT 961. GAACAAAAAAJ 962. CTTTTTGGAA 963. CTTTTCTTTA 964. Human GTF3A mRNA for Xenopus D32257 transcription factor IIIA homologue. CTTGTAGTCC 965. ATTTCTTGCC 966. CTTCATAAGT 967. ATTTGCCTCT 968. GAATGTAAGT 969. CAATTAATAC 970. CAGCGGAAGC 971. CAGCCTTGCG 972. - CAGATGCAAA 973. ---- CAGATACCCC 974. CACGATTAAA 975. CACCGGGTAG 976. Homo sapiens nonsense-mediated mR.NA AF074016 decay trans-acting factor mRNA, CGGACAATCA 977. CACAAACACA 978. CATACACTCT 979. Homo sapiens Humig mRNA. X72755 S60 GACTAGTGCG 980. GATTTTCTGG 981. GCCGGCTCTT 982. GCAAAACTCT 983. TGTTAATGTT 984. GCTGGCAGAG 985. GGCTGAGAAT 986. GCGAACTCCG 987. GCGCGGCTAC 988. GCGCTGCTTT 989. GCGGTAAAAA 990. GCTAGTGAAA 991. GCTCAGGATG 992. TTATGGGGAT 993. GCTGCAGGGG 994. TTACTCTTTC 995. Homo sapiens mRNA for aldehyde X75425 dehydrogenase (using GCTTGTAAAA 996. GCTTGTAGCC 997. GGAATAAATT 998. Human mRNA for cytochrome c1. IX06994 SUBSTITUTE SHEET (RULE 26) WO 99/65924 94 PCT/US99/13800 TAG SEQ ID DESCRIPTION ACCESSION NO GGAATCCTGT 999. GGAGAGGGCA 1000. GGAGTAGGAA 1001. TAAACTGAAA 1002. GCTCTCCCCT 1003. GCACTCTATG 1004. Human interleukin-8 receptor type B M73969 (IL8RB) mRNA. complete cds. GATATCAAAA 1005. TTGGGGTATC 1006. GCCTCTTCCC 1007. TGGGCTACTC 1008. TGGACCAGTG 1009. TTGGTCATCC 1010. TTCAGCAGAG 1011. TTGGTGATAC 1012. TTGTAAATAG 1013. Homo sapiens Golgi complex autoantigen U51587 golgin-97 mRNA, complete TTTACAGGGT 1014. TTTATTCCCT 1015. AAGATAATGC 1016. IT-11CTGGC 1017. TCTGAAGTGG 1018. GTTTGCAAGT 1019. TGGCGTTGAG 1020. GCAAAACTTT 1021. TGTAGGAAAC 1022. TGTATGGTGG 1023. TGTATTACAG 1024. Homo sapiens mRNA for novel DNA X63071 S50 binding protein. TGTCTCCTTC 1025. TGTGAGCAGA 1026. TTCAGTGCCT 1027. TGTTCCTGAG 1028. TTCAGCGTTC 1029. TTAAAGTCAA 1030. TTAACAATTC 1031. TTACACTGGA 1032. TGTT111ATG 1033. GGTTCAGTTA 1034. GATCCGCTCT 1035. GTTGGTCCCT 1036. GTTGTTAACA 1037. Homo sapiens heparan sulfate 3-0- AF019386 sulfotransferase- 1 precursor AGGAGGGATA 1038. SUBSTITUTE SHEET (RULE 26) WO 99/65924 95 PCTIUS99/13800 TAG SEQ ID DESCRIPTION NO [GTGTCAGATA 1039. GTATATAACT 1040. GGGAAGGGGG 1041. GGGAAGTCAC 1042. Human FX protein mRNA. complete cds. 1U58766 GGGCGGGGGC 1043. Human DNA polymerase delta catalytic 1M80397 subunit mRNA, complete cds. GGGGACTCCG 1044. GTTGAGTAAC 1045. - GGTACAAATA 1046. GTTATTGAGG 1047. GTGACTCGCA 1048. GTACTTACCT 1049. GTGACCTTCT 1050. GTATCTTAAT 1051. GTATGGAAGA 1052. GTATGTAACT 1053. Human mRNA for high affinity Fc X14356 M21 receptor (FcRI). GTCTGCCTGG 1054. Homo sapiens metase (MET-I) mRNA. L23134 complete cds. GTGAAAAACA 1055. GFTTCATTC 1056. GTACATTGTA 1057. AGGCCTGCCA 1058. GGGGGCAGTG 1059. GCACACTAGC 1060. GCAACAACAC 1061. GCAACTGTGA 1062. GCAAGAA'TIT 1063. GCCTGAGGGG 1064. GCACCTATTG 1065. GGGAAGATCT 1066. Homo sapiens mRNA for ERp28 protein. X949 10 GCAGTGCCAA 1067. GCATTCGCAG 1068. GCATTTTGTG 1069. GCCAATTGGG 1070. GTTGGGTAGA 1071. GCCCTGGAAA 1072. TTTTGTTAAT 1073. GTTATAATAC 1074. Homo sapiens mRNA for putative Y10032 serine/threonine protein kinase. GTGATCATTA 1075. GTGCCCCTTC 1076. GTGCTCATTC 1077. Homo sapiens mRNA for TGF-beta AB000584 superfamily protein, complete cds. SUBSTITUTE SHEET (RULE 26) WO 99/65924 PCT/US99/13800 96 TAG SEQ ID DESCRIPTION ACCESSION NO GTGGCAGATG 1078. GTGGGTGTCC 1079. GTGGTGCGCG 1080. GTGGTGTAGG 1 1081. GGCCACCCTG 1 t082. GTTAAAACAG 1083. GTGACAGAAT 1084. Human uridine diphosphoglucose U27460 pyrophosphorylase mRNA. complete GCCCCTTGCA 1085. AGCTGCCGCA 1086. TGAATTCTAC 1087. AGAAATACCA 1088. AGAGCTCACT 1089. AGCAAGCCCC 1090. AGCACAGAGG 1091. Homo sapiens citrate synthase mRNA. AF047042 complete cds. AGCACATTCT 1092. TTGGTAAAGA 1093. AGGGCAGAGG 1094. TTGGGCAATA 1095. AGCCTTTGTT 1096. Human mRNA for collagen binding D83174 protein 2, complete cds. AGCTAAGTTT 1097. ATATGTTGAC 1098. AGCTGCTGGT 1099. AGGACAGAAG 1100. AGGATGGCGG 1101. GATAGGTCGG 1102. Homo sapiens mRNA for iron regulatory Z11559 factor. AGCCCTAGTA 1103. TGATGTGATA 1104. TGGAGTGAAG 1105. TCTTGCCTAG 1106. TCTTTTGAAT 1107. AGGGAGACCT 1108. ATATGAAGCA 1109. AAGCGAGACG 1110. AAGGAAGATT 1111. AAGGACTCCG 1112. AAATCAATAA 1113. ATCATTGTGG 1114. AGTCTCTCTT1 1115. AGTGCAAACG 1116. AGTTTCTTGA 1117. SUBSTITUTE SHEET (RULE 26) WO 99/65924 97 PCT/US99/13800 TAG SEQ ID DESCRIPTION 'ACCESSION NO ATACAATAAA 1118. Human gene for PP15 (placental protein X07315 ___ ___ ___ ___ ___ ___ __ 1 5); ACTTACATTA 1119. ATAGCCTCTT 1120. AGCCTGTTGC 1121. ACTGGGCGCC 1122. Human mRNA for KIAA0358 gene. AB002356 complete cds. ATCAGCTGCT 1123. AGGCTAGACC 1124. ATCCGCCTGC 1125. ATCGATCGCC 1126. ATCGTTGTAA 1127. Homo sapiens mRNA for ATP-dependent AB001636 RNA helicase #46, complete cds. ATGAATATTC 1128. Homo sapiens partial mRNA; single read Z50170 (clone A335 1). ATGAGCTATG 1129. ATGCGCAAGG 1130. Homo sapiens (xs 13) mRNA, 284bp. Z36785 ATACAGGTCT 1131. Homo sapiens mRINA for DNA binding X85786 regulatory factor. AGCACCAGAA 1132. Homo sapiens rnRNA for KIAA0690 ABO 14590 protein, partial cds. TATTTATATG 1133. Homo sapiens cig4l mRNA, partial AF026943 sequence. TACATACGTC 1134. TACTCCAAGC 1135. TAGATGTGAT 1136. TAGCTGAGAC 1137. Human Rchl (RCH1) mRNA, complete U09559 cds. TAGGTCCTCT 1138. TATAAATAAA 1139. Human mRNA for KIAA0 130 gene. D50920 complete cds. TATGGGTTCC 1140. Homo sapiens full length insert cDNA AF088028 clone ZCI9E11. TATTTTACGT 1141. Homo sapiens RNA polymerase II L34587 elongation factor SIII, p15 subunit TGATCACTGC 1142. TGACATTCCC 1143. TAAATACAGT 1144. TCAGTGAACG 1145. Human mRNA for motor protein, partial D21092 cds. TCAGTTCTGA 1146. TCCACCAGTT 1147. TCCGAAACCT 1148. TATATTGAGA 1149. SUBSTITUTE SHEET (RULE 26) WO 99/65924 PCT/US99/13800 98 TAG SEQ ID DESCRIPTION ACCESSION NO TTGGTGAAGA 1150. TTCCCTCGTG 1151. ITTGAACACTT 1152. TTGAATCGTG 1153. TCAGCAGGGC 1154. TTGCCTGGAT 1155. TGCGTTGAGA 1156. TGACCTGTGT 1157. TGCCTTACAG 1158. TGATGTTTGC 1159. TGCCGTGCCT 1160. TGCCCTGAGA 1161. Homo sapiens cytochrome oxidase AF026851 assembly factor (PET 112) mRNA, TGCATATCAT 1162. Homo sapiens mRNA for CRMI protein. D89729 complete cds. TGATGGGCAT 1163. TCCTTCTGTG 1164. TCCTCCCTAC 1165. TTGGAAACCT 1166. TGAGGAGCTC 1167. GAGGCCGGCC 1168. AAGCACAAAA 1169. Homo sapiens DNAX activation protein AF019562 12 (DAP12) mRNA, complete cds. GGGGCAGGGC 1170. CCCAGCTAAT 1171. Human 15-lipoxygenase mRNA, M23892 complete cds. GCTCCCAGAC 1172. Homo sapiens mRNA for synaptogyrn 2. AJ002308 AGGCGAGATC 1173. CCATTGCACT 1174. Homo sapiens full length insert cDNA AF075065 YQ02E12. TGCGAGGAGA 1175. Homo sapiens mRNA for renin-binding D10232 DO1 protein, complete cds. GTACGTCCCA 1176. Human neutral amino acid transporter B U53347 mRNA, complete cds. AATCAACTTG 1177. TGCGCGCCCT 1178. CAGGATGACG 1179. GGCGGGGACA 1180. CCACTCCTCA 1181. Human mRNA for DAD-1, complete cds. D15057 AGAGGTTGAT 1182. GGTGAGACCT 1183. neuropolypeptide h3 [human, brain. S76773 mRNA Partial, 723 nt]. CAGGAACGGG 1184. Homosapiens ERK activator kinase L 1285 (MEK2) mRNA. SUBSTITUTE SHEET (RULE 26) WO 99/65924 PCT/US99/13800 99 TAG SEQ ID DESCRIPTION ACCESSION NO CCAATTTGCA 1185. iGTTTCTTCCC 1186. ~ GGATGTGAAA 1187. Human mRNA for T-cell surface X16996 glycoprotein E2. GTGGCGCACA 1188. 26 S protease subunit 5b=50 kda subunit S79862 [human. HeLa cells, mRNA GATTAATGTG 1189. Homo sapiens ICB-1 mRNA, complete AF044896 cds. GAATTTCCCA 1190. Human mRNA for complement X04481 KO 1 component C2. TGTGAACACA 1191. Human mRNA for interferon regulatory X14454 factor 1. CGGCTGAATT 1192. GGAAGATGTT 1193. GGAAGCACGG 1194. Human antisecretory factor-I mRNA. U24704 complete cds. GGAAGGGGAG 1195. Homo sapiens mRNA for NF-kB subunit. X61498 CTAACCAGAC 1196. Human F-actin capping protein beta U03271 subunit mRNA, complete cds. TACCCCTCTC 1197. Homo sapiens phospholipase C-beta-2 M95678 mRNA, complete cds. TGGAGGCCAG 1198. GTGACAGACA 1199. Human nuclear factor NF45 mRNA, U10323 complete cds. TCACGGCAAG 1200. ATTGTGCCAC 1201. AGACAGAGTG 1202. ACCTGCTGGT 1203. Homo sapiens clone 23675 mRNA AF0521 13 sequence. CCTTACTTTA 1204. TGAACCCGCC 1205. ACCTCAGGAA 1206. Human high density lipoprotein binding M64098 M83 protein (HBP) mRNA, complete GACCCTGCCC 1207. Human FK-506 binding protein L37033 homologue (FKBP38) mRNA, complete cds. TGTGATCAGA 1208. Homo sapiens F I FO-type ATP synthase AF092124 subunit g mRNA, complete cds. TCCTTCTCCA 1209. Human mRNA for alpha-actinin. X15804 AGCCTGCAGA 1210. GCTGCCCTTG 1211. human alpha-tubulin mRNA, 3' end. K00557 ACAAACTTAG 1212. CTCGGTGATG 1213. Homo sapiens mRNA for ras-related D78132 GTP-binding protein, complete SUBSTITUTE SHEET (RULE 26) WO 99/65924 100 PCT/US99/13800 TAG SEQ ID DESCRIPTION ACCESSION NO TTCTGGCTGC 1214. 'Human mRNA for core 1 protein. D26485 complete cds. CCAGGAGGAA 1215. Hs.103424: HEAT SHOCK COGNATE 71 KD PROTEIN GTGAAGCCTC| 1216. CTGAGACACC 1217. GGGAGGGGTG 1218. TGGAGAATGT 1219. TGTGCACCCC 1220. Homo sapiens clone 24574 mRNA AF052151 sequence. TTGATTTCTT 1221. Human MHC class II HLA-DQAI M33906 mRNA, complete cds. GTCATTTGGA 1222. TGGTCTGGAG 1223. Human mRNA for KIAA0216 gene. D86970 complete cds. ATGGCACACA 1224. Homo sapiens class I cytokine receptor AF053004 (WSX1) mRNA, complete cds. AGATGAGAAA 1225. TCTGCAAATT 1226. GCAACATCAG 1227. CATCTCTAGT 1228. GCAGCCCCAA 1229. GGAATACGCA 1230. TAGAAGGTGG 1231. TCAAAGCCAT 1232. AGGGCTTCAA 1233. CGCCTCCGGG 1234. GGTAGCAGGG 1235. CCCATTTGCA 1236. AATGCTTTGT 1237. CCTGCACCCA 1238. Human Sel-1 like mRNA. complete cds. U11037 TTCCAGACCT 1239. Human HepG2 3' region Mbol cDNA, D17 clone hmdldl2m3. TTGTACAACA 1240. TTTCCACCCG 1241. GTGGCGGGCG 1242. Homo sapiens mRNA for KIAA0565 ABOl 1137 protein, complete cds. ACAGCTAACA 1243. ACCGCCTGTG 1244. ATGGTTTT1G 1245. ACGGTGATGT 1246. TCTTTGTAGG 1247. CCTTTTTAGT 1248. ACTACCACCC 1249. SUBSTITUTE SHEET (RULE 26) WO 99/65924 PCT/US99/13800 101 TAG SEQ ID DESCRIPTION ACCESSION NO TTCAAAAAGG 1250. TGTCAGAGAT 1251. TGATGTGATC 1252. Homo sapiens GT197 partial ORF L38932 mRNA. 3' end of cds. TGAAGAGAAG 1253. Human mRNA for KIAA0106 gene. D14662 complete cds. TGAAAGTGTG 1254. Homo sapiens antigen NY-CO-25 (NY- AF039695 CO-25) mRNA, partial cds. CAATACATAC 1255. TATGACTTAA 1256. Homo sapiens calcium-activated AF031815 potassium channel (KCNN3) mRNA. GTGAATGACG 1257. GTCCCAACAC 1258. Homo sapiens full length insert cDNA AF086389 clone ZD73FI 1. GGCCCTGCAG 1259. GCTTACCTTT 1260. GCCTGGGCTG 1261. GAGCTCTGAG 1262. Homo sapiens dysferlin mRNA, complete AF075575 cds. GCCTCCACAG 1263. TTACAATTTG 1264. GGCTCCTCGA 1265. Homo sapiens tapasin(NGS-17) mRNA, AF029750 _________complete cds. TCGATGTGGG 1266. TGTGCCCTGT 1267. GACACCAACT 1268. Homo sapiens deubiquninating enzyme AF017305 ___________ _______UnpEL (UTNP) mRNA, complete ACACTTACAA 1269. Homo sapiens UEV IBs (UBE2V) U97280 _____________mR.NA, alternatively spliced, partial GGAGAAGATG 1270. GACAGTCCTG 1271..__ GAGAGCTACA 1272. Human electron transfer flavoprotem J04058 I_____________ alpha-subunit mRNA, complete______ GGGTCTGTGA 1273. TATGCTGTTA 1274. GTTGTGGTTC 1275. TCATAACTGT 1276. Human mRNA for flavoprotein subunit oID30648 complex II, complete cds. GAGCCAACCC 1277. TGAGTGACAC 1278. ACCTGTGACC 1279. Homo sapiens mRNA for pro-urokinase D00244 precursor, complete cds. HTGCCTTAGTA 1280.sapiensUEls(UBE2VU TGGAGGTGGGT -1281. __________ SUBSTITUTE SHEET (RULE 26) WO 99/65924102 PCT/US99/13800 TAG SEQ ID DESCRIPTION ACCESSION NO GCGGACGAGG 1282. Homo sapiens TFAR19 mRNA. compieteiAF014955 cds. TGGGGATTAC 1283. TCAATCAAGA 1284. Human 14-3-3n protein mRNA, complete L20422 cds. GCGGACTGGG 1285. 1 GCCTCTGCCA 1286. Human mRNA for KIAA0272 gene. D87462 partial cds. GCCGAGTCCA 1287. Homo sapiens leukocyte-associated Ig- IAF013249 like receptor-I (LAIR-1) mRNA, GCCCTTGCAA 1288. GCCCTGGGTG 1289. GGCCGTGTGA 1290. TGCAGTGACT 1291. Homo sapiens mRNA for 37 kDa LIM 1X93510 domain protein. ACAGTGCTTG 1292. Human mRNA for protein phosphatase X12656 2A (beta-type). AAGTCGCTCA 1293. TAGTTGTAGG 1294. TAATAAAGAA 1295. Human mRNA for cytokeratin 15. X07696 TGGGTGTTGA 1296. GGAAGGGAGG 1297. GATGTTGTCC 1298. GGCCAGGAAG 1299. GGGAGCCGAG 1300. Human mRNA for KIAA0 169 gene, D79991 partial cds. GGGGGCTGCT 1301. GTGCACTGAA 1302. GTGGACCCCA 1303. Human siah binding protein 1 (SiahBPl) U51586 mRNA, partial cds. GTGTCTCATC 1304. Homo sapiens mRNA for 2- X84907 phosphopyruvate-hydratase-alpha enolase. GTTTAAAAGA 1305. ATGGTTAAAG 1306. CTGGAGAACA 1307. TGGCAGGTTC 1308. CTAAATATAG 1309. CGCTTTTGTA 1310. CGAATTGAGA 1311. CCTGTCCTGC 1312. CCTCTTTAAA 1313. Human mRNA for KIAAO140 gene, D50930 complete cds. CCCATCGTCT 1314. complete_____ SUBSTITUTE SHEET (RULE 26) WO 99/65924 103 PCT/US99/13800 TAG SEQ ID DESCRIPTION ' N NO ACAGCCACTG 1315. TGTGTCAAAG 1316. GAAGATTAAT 1317. Homo sapiens sorting nexin 3 (SNX3i AF034546 mRNA. complete cds. CTGGGTTGTG 1318. ATGGAAAGGA 1319. ATCCACCCAC 1320. Human telomeric repeat DNA-binding U74382 protein (PIN2) mRNA, complete AGGGAGAGGG 1321. Homo sapiens mRNA for de-ubiquitinase.IX9 1349 AGGCTGCGAC 1322. AGGATTAAAA 1323. AGGATGTGGG 1324. Human kinesin-like motor protein KIF IC U91329 mRNA, complete cds. AGCAGCCGCT 1325. AGACCAAAGT 1326. Human mRNA for heat-shock protein 40, D49547 D17 complete cds. AGAAGCCAGA 1327. CACTGTGTTG 1328. TTACCCAGGC 1329. Human UMP synthase mRNA, complete J03626 cds. CAGCGCACAGI 1330. TTGACCTGTG 1331. TTTGTGCACT 1332. TCGGGAGCTG 1333. ATGTTGTACT 1334. CTGTTGCATT 1335. CTCTGCTCGG 1336. TTTCAGGGGA 1337. TTCTCTCAAC 1338. TATTTTAAAT 1339. GTCTACCTGA 1340. GGCATTGGGG 1341. TTCTCTTTCA 1342. TGTCTAACTA 1343. TGGTGGAATG 1344. GTCCCCCCAA 1345. GTATCTTCAG 1346. GTATAATTTG 1347. TAACAAAGGAI 1348.1 TGTGAATTTT 1349. TGTGCGCGGG 1350. TGTTAGCCTG 1351. TTACAACATT 1352. TTTGTGACTG 1353. SUBSTITUTE SHEET (RULE 26) WO 99/65924 PCT/US99/13800 TAG SEQ ID DESCRIPTION ACCESSION NO GGCGCCTTCT 1354. GGGGCTGGAG1 1355. GGAGATGAAG] 1356. Human SLP-76 associated protein U93049 mRNA. complete cds. I GGAGATGCCT 1357. Human interleukin 3 receptor (hiL-3Ra) M74782 mRNA. complete cds. GTGGAGGTGC 1358. Human 100 kDa coactivator mRNA. U22055 complete cds. GGCAAAGAGG 1359. GGCACAGTAA 1360. Homo sapiens full length insert cDNA AF086406 clone ZD76B08. GCTTTCATTG 1361. ~-- GGTTACATTA 1362. TTCACTGTAG 1363. GTCTGCCTCA 1364. GGAATGAGAA 1365. GGGTTTGAAC 1366. GGAGAGTACA 1367. GGTTATCTGT 1368. GTAGATGCAA 1369. Human transcription factor (ITF-1) X52078 M30 mRNA, 3' end. GGCAGAAGAT 1370. GGGGGTCACC 1371. Human mRNA for ATP synthase subunit D13118 c encoded by P1 gene. GTGCGTGCCT 1372. TAGGTTGTCA 1373. GTTGCGGTTA 1374. GTTGGAGGCC 1375. GTTTCTCTGG 1376. GTTTTGTACA 1377. GCTGGGACAG 1378. GTGATGCTGG 1379. GTGGACTTTT 1380. TACCCGCCTC 1381. TACTAAAAAA 1382. Homo sapiens NADH-ubiquinone AF050640 oxidoreductase NDUFS2 subunit mRNA, GTTTGACAGA 1383. GCTGGCAGGC 1384. TGGGGGGTTT 1385. GGACCCTCTC 1386. Homo sapiens clone 23764 mRNA AF007133 sequence. TAACCTGCTA 1387. 1 1 ATAGCTGGGG 1388. Homosapiens ERK activator kinase L 11284 (MEK1) mRNA. SUBSTITUTE SHEET (RULE 26) WO 99/65924 105 PCT/US99/13800 TAG SEQ ID DESCRIPTION SIONI TTGATGGTGC 1389. CAATGGAGCT 1390. CAAGCTGTAA 1391. Human HepG2 3 region Mbol cDNA. D17249 clone hmd5a09m3. ATTGTCAGGG 1392. ATTGGCTGGG 1393. protein phosphatase 2C alpha [human. S87759 teratocarcinoma. mRNA, 2346 CAGGACGGGC 1394. Homo sapiens encoding CL A-l mRNA. Z22555 ATGGGAACCA 1395. Homo sapiens mRNA for GTP-binding X80754 protein. CAGGAGACAG 1396. ATATTGATGA 1397. ATATATTCAG 1398. ATATAGTCAG 1399. Human mRNA for KIAK0002 gene. D13639 complete cds. ATAAATAAGG 1400. !AGGTTTTCAT 1401. _ AGGGGCGCAG 1402. Homo sapiens mRNA for protein X99656 containing SH3 domain, SH3GL1. AGAGCCAAGT 1403. AAAGAGAAGA 1404. AAGGGTGCCA 1405. TGTTCCCTTT 1406. Human MXII mRNA, complete cds. L07648 AGCTGATCAG 1407. Human mRNA for acylamino acid- D38441 releasing enzyme, complete cds. AAGGAAAGTG 1408. Homo sapiens DEC-205 mRNA, AFl0 1333 complete cds. AACGCTGCCT 1409. CACACCAATT 1410. AAAGGTTGGT 1411. Human m.RNA for KNP-la, complete cds. D86061 ATTGACCGCT 1412. AAAAGATACT 1413. CCTTTCTCTC 1414. Human mRNA for KIAA0068 gene, D38549 partial cds. TTTTGTACCA 1415. AATGCTGTGA 1416. ATGGCGATCT 1417. CAGTCTCAGA 1418. AACAGAATAT 1419. Homo sapiens GA17 protein mRNA. AF064603 complete cds. TTTTATCTGG 1420. Homo sapiens mRNA for ITBA2 protein. X92896 TCCTTCTACG 1421. TCCATCGTCC 1422. TCATACTGAA 1423. SUBSTITUTE SHEET (RULE 26) WO 99/65924 106 PCT/US99/13800 TAG SEQ ID DESCRIPTION ACCESSION NO TCAGCAAGGG 1424. TCAAGTCACC 1425. AGGGCCCTCA 1426. TTCTCCCCCT 1427. Human mRNA for KIAA0339 gene. AB002337 complete cds. TCTGTAGCTA 1428. TTTTAAACTT 1429. AGTTGAAATT 1430. GTGTAAATGG 1431. TACTGGCTCA 1432. GTGGCGGGAG 1433. Homo sapiens mRNA for RAP74. JX64002 GTTATTCCCC 1434. TGACTGGCAG 1435. Homo sapiens mRNA for IL-l/TNF X84805 __ 7_ 1436.__ inducible EST (clone MEC-205). TGCCCAGACC 1436. AGCCACCTCA 1437. TGAACCCGTT 1438. TGGGATGACA 1439. TGGGAACCTA 1440. TGGCTGTGAG 1441. Human chromosome 17q12-21 mRNA, U18920 clone pOV-3, partial cds. TGGCCTAAAA 1442. TCTCTGCAAA 1443. TGCTTGGCTT 1444. Human mRNA for small GTP-binding D14889 protein, S10, complete cds. TCTGCATAGA 1445. TGATCTGCCT 1446. TGAGGAGCTG 1447. TCAAACTGTG 1448. TGACTGAAGC 1449. Homo sapiens 3-phosphoglycerate AF006043 dehydrogenase mRNA, complete cds. TGTAGTATT 1450. AGGCTTTAGG 1451. TGGCAAAATG 1452. CTACCAGCAC 1453. AATTGTGCAT 1454. GATGAGAAGA 1455. GCGGGAGCGG 1456. Human mRNA for KIAA0224 gene, D86977 complete cds. CCGCCCTCTA 1457. CCGGCCAGCG 1458. CCTATGGCTT 1459. CCTGGCAGTT 1460. CGAAGTGTCC 1461. SUBSTITUTE SHEET (RULE 26) WO 99/65924 107 PCT/US99/13800 TAG SEQ ID DESCRIPTION NO CGCGCACCCG 1462. CGCGTCACTA 1463. GAAGCTGCCT 1464. Homo sapiens leucocyte AF025532 immunoglobulin-like receptor-5 (LIR-5) mRNA, CGGTTTGCAG 1465. 1 GCCTGTGCTG 1466. Homo sapiens Huntington's Disease (HD) L12392 mRNA. complete cds. CGTTTTCTGA 1467. Homo sapiens protein tyrosine L39000 _ phosphatase (PRL-1) mRNA. 3' end of GCGCGGGCGAI 1468. CTCGGCGAGC 1469. CTGCAGAGTG 1470. Human putative holocytochrome c-type U36787 synthetase mRNA, complete cds. CTGCGTGATG 1471. CTGGATCTGG 1472. Human fetal brain glycogen U47025 phosphorylase B mRNA. complete cds. CTGGTCCTCC 1473. CTTAAGACTT 1474. CTTTCAAAAC 1475. CGGTGTTGAG 1476. CCTCCCCGAA 1477. TTTGTTGTAT 1478. TTTCTCTCCT 1479. Human transcription factor NFATx U14510 mRNA, complete cds. TTGTGATGTA 1480. TTGGTGAGGG 1481. CCCCTCTGAG 1482. Homo sapiens IFI-4 mRNA for type I X79448 protein. AGAAGTGTCT 1483. GCCTCCAGGG 1484. GAGCACATCC 1485. CGTCTCCACA 1486. GAGAGCTGGG 1487. Homo sapiens P2Y6 receptor, short splice AF007891 variant mRNA. complete CCAGATGTGT 1488. GATTTCTATT 1489. GAGCTTACCC 1490. CCCTTCCCCG 1491. GAGCACTGTT 1492. ACTTGATTTG 1493. Homo sapiens mRNA for KIAA0494 AB007963 protein, complete cds. ACGTGTCTAT 1494. Human clone 23612 mRNA sequence. U90902 ACCCTCCTCT 1495. SUBSTITUTE SHEET (RULE 26) WO 99/65924 10 PCT/US99/13800 TAG SEQ ID DESCRIPTION m S 10 N _ _ _ _ _ _ NO _ _ _ _ _ _ _ _ _ ACCAGAACAG 1496. _ ACCACAAATG 1497. ACACACCTGG 1498. AAGGCACAGA 1499. Homo sapiens phosphatidylinositol AFO 14807 synthase PIS) mRNA. complete GCAAAATAAC 1500. Human initiation factor 4D 9elF 4D) M23419 mRNA. complete cds. GCCCTGATTT 1501. Human interferon regulatory factor 5 U5 1127 (Humirf5) mRNA, complete cds. GCCCCACAGC 1502. GCCAAGATGC 1503. GAGATCCACG 1504. Human zinc finger protein mRNA, L16896 complete cds. GCATATTAAA 1505. Human mRNA for XP-C repair D21090 _________ 1506.__ complementing protein (p58/HHR23B). GAGGAGCCCC 1506. _________________ CTTTTAAGAA 1507. GATGAGTGGA 1508. Human adrenodoxin mRNA, complete J03548 cds. GAAACTGGAA 1509. GAGGTGGGGC 1510. GCCAAAAAAA 1511. Homo sapiens (TL7) mRNA from LNCaP X75687 cell line. TGTGTTGTCA 1512. Human mRNA for NAD-dependent X16396 methylene tetrahydrofolate GTGATGGTGT 1513. Human lupus p70 (Ku) autoantigen J046 11 protein mRNA, complete cds. TAGACTAGCA 1514. Human globin gene. M69023 GTGAGCCCAT 1515. CTGCCGCCGA 1516. GGTCACATTA 1517. CTGGGCCTGC 1518. CGATTCTGGA 1519. ACTGGTACGT 1520. AAATGCCACA 1521. CAGGGTCCTG 1522. TTGTAAAATA 1523. Homo sapiens HUMFLI-1 mRNA. X67001 S44 GACCCACTAC 1524. Human lymphocyte activation antigen J03569 4F2 large subunit mRNA. TCAATAAAGA 1525. Homo sapiens QRSHs mRNA for X76013 glutaminyl-tRNA synthetase. GGGAGCTGCG 1526. GTGTAATAAG 1527. Human hnRNP A2 protein mRNA. M29065 ICCTTCCCTGA 1528. SUBSTITUTE SHEET (RULE 26) WO 99/65924 PCT/US99/13800 TAG SEQ ID DESCRIPTION ACCESSION S~~NO __ _ _ __ TTCACAGTGC 1 1529. I CTGGAGGCAC 1530. _ GACTTTGGGA 1531. Homo sapiens mRNA for neuropathy ]AJ004832 target esterase. CGOCCGTGC 1532. Homo sapiens mRNA for Glvoxalase II. X90999 TCACCTTAGG 1533. ACTGGCGAAG 1534. Human hLON ATP-dependent protease U02389 _mRNA, nuclear gene encoding CAGTTACTTA 1535. Homo sapiens mRNA for HSl protein. X57346 CCAAGAAAGA 1536. Homo sapiens polvadenylate binding U75686 protein mRNA. complete cds. CCTGCAATCC 1537. CCTGAGCCCG 1538. GGAACAGGGG 1539. GGCCAGCCCT 1540. Human liver-type 1-phosphofructokinase X15573 (PFKL) mRNA, complete cds. TCTGCCTGGA 1541. TTTGTTCATT 1542. Homo sapiens HnRNP F protein mRNA, L28010 complete cds. TGCCTGTAGT 1543. Hum ORE (CEIS) mRNA, 3' flank. M80651 CACCTGCAAT 1544. CACTACACGG 1545. Human rapamycin-binding protein M65128 (FKBp- 13) mRNA, complete cds. GGGCAGCTGG 1546. | CAAGGATAAG 1547. GTGCCTAGGG 1548. GCAGGCTGTG 1549. Human prolidase (imidodipeptidase) J04605 mRNA. complete cds. TACATCCGAA 1550. GTATGGGCCC 1551. Human glycoprotein mRNA, complete M80927 cds. CTGAGGTGAT 1552. GAGGTCCTTC 1553. TCCTGCTGCC 1554. CATCTGTGAG 1555. Homo sapiens DAP-1 mRNA. X76105 CCTGTGGTTT 1556. Human protein p78 mRNA, complete cds. M80359 ATGATCCGGA 1557. Homo sapiens calcium-ATPase (HK1) M23114 J04 mRNA, complete cds. AGGAGTCGAC 1558. Human ubiquitin fusion-degradation U64444 protein (UFD IL) mRNA, complete TGTCCGTCAC 1559. GGCGTCCTGG 1560. CACTTGCCCT 1561. branchio-oto-renal syndrome candidate S82655 gene {3' region} [human, SUBSTITUTE SHEET (RULE 26) WO 99/65924 110 PCT/US99/13800 TAG SEQ ID DESCRIPTION ACCESSIONI NO TGTTCCACTC 1562. Homo sapiens CD39L2 (CD39L2) AF039916 mRNA. complete cds. TGTGTTAAAA 1563. AGTGCACGTG 1564. - CAAAACTGGC 1565. TCTTCATACC 1566. GGTAGCCTGG 1567. Human xeroderma pigmentosum group E U32986 UV-damaged DNA binding factor GAGTTATGTT 1568. TGGAAATAAA 1569. CTTCTGGGGA 1570.. Homo sapiens rhoG mRNA for GTPase. X61587 S38 TTTGATGTAT 1571. Human messenger RNA fragment for the V00567 J00 beta-2 microglobulin. 7 TTAATAAATG 1572. Homo sapiens Cre binding protein-like 2 AF039081 mRNA. complete cds. TGAGCCACCG 1573. GTTGGGGTTA 1574. GCCATTATAA 1575. Homo sapiens mRNA for lysosome- X77196 associated membrane protein-2. CCCTGGGTTT 1576. CTAATAAATG 1577. CTGCTATGTG 1578. Human ras-like protein mRNA, complete M31470 cds, clone TC10. TTTCATCGTA 1579. AACGTGCAGG 1580. Human mRNA for argininosuccinate X01630 synthetase. AGCTGTTCAA 1581. CCTTGGCCTC 1582. GACAGTGTGG 1583. Homo sapiens mRNA for NuMA protein. IZ 11583 CTGGCAATGA 1584. CTCAAGCACC 1585. CTGGGACTGA 1586. GATTGTGCAA 1587. Human mRNA for KIAA0 183 gene, D80005 partial cds. GTGGCTCATA 1588. GTTGCTGCCC 1589. TGAATGTCAA 1590. TGACGTCAGC 1591. TGGTTTGCGT 1592. TTGATTTCCT 1593. Homo sapiens ICERE-1 mRNA, AF007790 complete cds. GCAACGTCAG 1594. CCTTTGAACA 1595. TTTTATGGAA 1596. SUBSTITUTE SHEET (RULE 26) WO 99/65924 i PCT/US99/13800 TAG SEQ ID DESCRIPTION ACCESSION _____ NO GTCCTTTCTG 1597. Human heparin-binding EGF-like growth IM60278 factor mRNA. complete cds. TGGAGAGCAA 1598. GGGAATGTGGI 1599. 'CCTACAGGGT 1600. CTGATTTATT 1601. CTGGGTAGCA 1602. Human cGMP phosphodiesterase gamma- M36476 CCCCTTAAC 603.subunit (PDEG) mRNA, complete CCCCTTTAAC 1603. CCCGAAACCA 1604. TCTTCTCCCT 1605. Human mRNA for hepatoma-derived D16431 _____ _ 1606.growth factor, complete cds. TGGTTTTTGA 1606. CACCATTCAG 1607. CTTTTCAGCA 1608. CAGGCCTCTG 1609. CAGTCAGGCT 1610. CCAACCCATC 1611. ATGACCTGAA 1612. TGGATGGCTT 1613. GTTGGGGGTA 1614. Homo sapiens mRNA for transcription X57435 factor AP-4. CAAATAAAAA 1615. Homo sapiens (clone CD18) tumor L04270 necrosis factor receptor 2 related TCCATAAGGA 1616. CAAAGACAAT 1617. TCTTTCCCCA 1618. TCTTTGGCCT 1619. TGATTGGCTT 1620. Human alpha-N-acetylgalactosammidase M38083 TGCGACG_121 mRNA, complete cds. TGCAGAACGG 1621. ____________ TGCATCAATA 1622. GCAGCGCCTG 1623. TGCTAAAAAA 1624. GTGTCTCCCG 1625. _ CCCTACCTTC 1626. TACCACCTCC 1627. Human pregnancy-specific beta-I- M17908 J03 _________glycoprotein mRNA, complete cds. AGCACCTCCG 1628. CTGTGCTCGG 1629. Human mRNA for mitochondrial short- D13900 chain enoyl-CoA hydratase, CTGTGTCJACT 1630. Human short chain acyl-CoA M26393 dehydrogenase mRNA. complete cds. CTTAAGGATT 1631. SUBSTITUTE SHEET (RULE 26) WO 99/65924 PCT/US99/13800 112 TAG SEQ ID DESCRIPTION IACCESSIONJ NO [CTTACAACCG 1632. CTTCGGATGT 1633. Homo sapiens mRNA for SPOP. AJ000644 CTCAAAAAAA 1634. ATTAAAGTGC 1635. AACGGGCCCT 1636. AAGAGTTACG 1637. GGAGGTGGGA 1638.
AGATAATGTT 1639. Human fur mRNA for furn. X17094 TGTATTCCAC 1640.
-
AGCCTGTAGT 1641. AGGGATCCTA 1642. ATATAGGTCG 1643. ATCACGCCAC 1644. ATCCAACTTA 1645. CAGAGACGTG 1646. Human dystroglycan (DAGI) MRNA. L19711 _________cornplete cds. ATGGCTGCTG 1647. Homo sapiens nRNA for KIAA0664 AB014564 CCAATCTCIAT protein, partial cds. ATTACACCAC 1649. Homo sapiens full length insert cDNA AF086284 ________clone ZD46F04. AGAACCTTTC 1650. GAAGCCAGCC 1651. Hunan4E-binding protein 1 mRNA, L36055 _________complete cds. TGGGTCTGAA 1652. GGCATCGTTG 1653. Human nRNA for HLA-Cw*O702, D38526 ______________complete cds. GTGTGTGTGT 1654. Homo sapiens rRNA for beta 3 X70811 adrenergic receptor. GGCGTTGTCT 1655. GGGCATCTCA 1656. GGGCGAGAAC 1657. Homo sapiens huntingtin interacting AF049614 protein HYPL mRNA, partial cds. GGGGCCCCCT 1658. Homo sapiens mRNA for NA14 protein. Z96932 GGTGTGCTTG 1659. Hono sapiens clone 24736 Z FRNA AF055021 sequence. AACGGGGCCT 1660. ____________ GTGGTATGTG 1661. GCCAAGCCTG 1662. Human mRNA for protein p68. Y00097 GGATGTAGAG 1663. ___________ ACAAAGCCCC 1664. _____________ CTGGGACTGC 1665. _____________ CTGCGAGTGA 1666. ____________ CTCTTTGATT 1667. _____________ SUBSTITUTE SHEET (RULE 26) WO 99/65924 113 PCT/US99/13800 TAG SEQ ID DESCRIPTION 'ACCESSION NO ICCCACCAGGA 1668. ICTAGTATAAG 1669. iCCTTTGCACT 1670. CCTGTAATAC 1671. Homo sapiens full length insert cDNA IAF086176 clone ZB95G04. GTCTTACTTT 1672. GAGGCACTGA 1673. Human Ikaros/LyF-1 homolog (hlk-1) U40462 mRNA. complete cds. TGTTCTTTGC 1674. TTAATATGTG 1675. TTGACACACG 1676. __ TTGGCCCAGA 1677. Human IL-4-R mRNA for the interleukin X52425 4 receptor. TTTAATACAT 1678. TGCCTTGAAA 1679. Homo sapiens COX4AL mRNA, AF005888 complete cds. GGCCTTTTT 1680. Human mRNA for histone H lx, complete D64142 cds. GACATATGTA 1681. Homo sapiens coxVIIb mRNA for Z 14244 cytochrome c oxidase subunit VIIb. GAGGAATTGG 1682. GTTGATTGTA 1683. GAGGATTTTA 1684. Homo sapiens ERC-55 mRNA. X78669 CCCATCGGCC 1685. GAGGGTCTTG 1686. GATGCTAACC 1687. AAATGGCTTG 1688. TTACTAAATG 1689. TCAAAAAAAG 1690. Homo sapiens partial mRNA; ID EE2- AJ227918 16F1. GTGGATGGAC 1691. CATTTGTAAT 1692. Human HepG2 3' region cDNA, clone D16914 hmd3cl2. TGTGGCCTCC 1693. ATGTTAGGGA 1694. Homo sapiens vesicle soluble NSF AF035824 attachment protein receptor (VTI I) TTCAAAGGAA 1695. Human mRNA for KIAA0051 gene. D29640 complete cds. GCCTTTCCCT 1696. CCCAGGTGTC 1697. CAGTATGTCC 1698. CATACTTTAA 1699. CATCCTCTCT 1700. SUBSTITUTE SHEET (RULE 26) WO 99/65924 114 PCT/US99/13800 TAG SEQ ID DESCRIPTION 'ACCESSION NO CATTTATCAT 1701. Human protein tyrosine phosphatase U48296 PTPCAAXI (hPTPCAAX I) mRNA. CCAAGTGAAC 1702. CCAATTGAAG 1703. CCACTGCTGC 1704. CAGCACCTGA 1705. CCATTATTTT 1706. CAGAAGCAAA 1707. CCCAAGTGCC 1708. CACAAACAGT 1709. CCCACTTGCC 1710. GCTGAAGATG 1711. CCCATAAGGA 1712. ,CCCATTCACA 1713. ATTCCAAGGA 1714. CCACTTACGA 1715. CACCAAAAAA 1716. CTCAGCAAAC 1717. ATTGGGACAG 1718. ATTGTAAATT 1719. CAAGACTGTT 1720. Human cyclin A/CDK2-associated p19 U33760 (SkpI) mRNA, complete cds. CAAGCAAAAT 1721. Human NifU-like protein (hNifU) U47101 mRNA, partial cds. TTAAACTTAA 1722. Human mRNA for HM89. D10924 CCCACCGGTG 1723. TGTATGTGGT 1724. GGATACAGGA 1725. TTACTGATTT 1726. TTACTGTGTA 1727. Homo sapiens KIAA0410 mRNA, AB007870 complete cds. TGTGGGTATT 1728. TTGAATTCAA 1729. TTGAGTAGGA 1730. TCACAATACA 1731. Human cyclophilin-40 m.RNA, complete L 11667 cds. TCCATCAAGA 1732. Homo sapiens clone 23598 mRNA. AF035309 complete cds. TGGGGGCACC 1733. Homo sapiens I-Rel mRNA, complete M83221 cds. TGGTTCTATA 1734. TGGTTTTGGC 1735. ICCCGAGAAAG 1736. TGTAGCATCA 1737. SUBSTITUTE SHEET (RULE 26) WO 99/65924 PCT/US99/13800 115 TAG SEQ ID DESCRIPTION ACCESSIONJ CCCAACCGGT 7 1738. !TCAGTTTGAA 1739. TTAGTTAAGC 1740. Homo sapiens mRNA (clone p5) for X81198 I _ _ archain. TGTATTTATA 1741. TTGCAACCAA 1742. TTCTTCTTCT 1743. TTAGCCAGGA 1744. Human LLGL mRNA. complete cds. D50550 TTATACAGCC 1745. TGTCTTTAAA 1746. TTATCATAGC 1747. TTCTGGGGGC 1748. TACATATGGA 1749. Human mRNA for KIAA0248 gene, D87435 partial cds. CAGGCCTGGC 1750. TGTAGATGTA 1751. CCTAAAGGAG 1752. CGCGACGATG 1753. CCGTGTTAAT 1754. CCCTCTGTGA 1755. CCCTGGGTTA 1756. CCGAGGAAGG 1757. CCGATTCGTC 1758. CCGCCATCTC 1759. CCGCTTCTGC 1760. CCGGAATGTG 1761.
----
CCTTAGTTTA 1762. - CCGGGCACAG 1763.
-
CAAGGTGCAA 1764. CCGTTCTGGA 1765. CTACCTTGGT 1766. CCTCCCAAGA 1767. CCTGACGCTC 1768. CCTGATGTGG 1769. Human AHNAK nucleoprotein m.RNA, 5' M80902 end. CCTGCCAAAA 1770. Homo sapiens mRNA for serine Y08686 palmitoyltransferase, subunit II. ATGTGGGCTC 1771. Homo sapiens garp gene mRNA, Z24680 complete CDS. CCGGGACATC 1772. AGAGAATCAG 1773. AGGAGTGGTT 1774. AGCGCTGAAA 1775. AGCGCCCTGG 1776. SUBSTITUTE SHEET (RULE 26) WO 99/65924 PCT/US99/13800 116 TAG SEQ ID DESCRIPTION ACCESS NO AGCCCTACAG 1777, CCTGTAGCCC 1778. Human fgr proto-oncogene encoded 5 MI97-J03 _ _ _ _ _ _ _ _ _ _ _ _1 7 7 9 . c -fi g r p r o te in , c o m p le te c d s . CCTTGCATTC 1779. CACAAATGCT 1780. CCCTAGGGCC 1781. CACCTCCCGG 1782. LCACCTCTCCT 1783. CACCTGTCCT 1784. CACTGCCTGT 1785. CACTGTCTCA 1786. CAGAAACAGA 1787. Homo sapiens clone 24670 mRNA AF055019 ______________sequence. CAGAAATATA 1788. CACAAACGGA 1789. CGTCAAGATT 1790. Human famesyltransferase alpha-subunit L 10413 mRNA, complete cds. GCTGGGATCA 1791. CCTTCTTGAT 1792. CTATCACTAC 1793. CCTTGGGCCT 1794. CCTTGTTTAA 1795. CCTTTCTGTA 1796. CGCCGCGGCT 1797. CCCCCCTTCC 1798. CGGTCCCATT 1799. CCTGGAAGGG 1800. CGTTTTCTTG 1801. CTAAAGGAGG 1802. Homo sapiens transcription factor M83233 (HTF4A) mRNA. complete cds. CTAACAGGAT 1803. CTAATTCTTT 1804. TTAAAACAAA 1805. CCTTCTGCCA 1806. TCTGCAAAAA 1807. -_--_ TATATTTCCT 1808. -_ TAGACATTTG 1809. TAGATCCTGT 1810. ----- TAGGACCCTG 1811. Homo sapiens clone 24664 PH-20 AF070608 homology mRNA, complete cds. TAGGGAATGA 1812. TGGAGCACAG 1813. TAGTTGATGG 1814. CTCAGCCTGA 1815. Human HepG2 3' region Mbol cDNA. D17172 SUBSTITUTE SHEET (RULE 26) WO 99/65924 PCT/US99/13800 117 TAG SEQ ID DESCRIPTION ACCESSION NO clone hmd2fl0m3. CAGGTGTCTT 1816. Homo sapiens mRNA for PkB kinase. Yl5056 CACGCGCTCA 1817. Human mRNA for RPB5 (XAP4), D38251 complete cds. [TATAGCTACC 1818. ITCTGCAGGTC 1819. TACTGTATGT 1820. --- ~ GTCAGGCCTC 1821. TTTAGACTTT 1822. Homo sapiens MAD-related gene AFO 10193 SMAD7 (SMAD7) mRNA. complete cds. TGCCAATAAC 1823. TGCAGGGCCT 1824. TGCAGGCTGG 1825. TGATGGCTCC 1826. Homo sapiens arylsulphatase A mRNA, X52151 J04 ______________ complete cds._ _ _ _ _ _ TGATGCACCT 1827. TGAGATTGAG 1828. TGAAACAAGC 1829. TCTTGGCATA 1830. TCTTGATGTC 1831. Homo sapiens full length insert cDNA AF086210 clone ZC48G12. TTAAGAGGGG 1832. Homo sapiens histone-binding protein M97856 mRNA, complete cds. TATATTGCAA 1833. TAGAAAAATA 1834. Homo sapiens RNA for neuroleukin gene. X16539 TAGTGCACAT 1835. TATGTTGGGG 1836. TATGCGTTTG 1837. Homo sapiens full length insert cDNA AF086023 clone YW23E08. ACTTGCGAAT 1838. TATTAGATGT 1839. Human CC chemokine STCP-1 mRNA, U83239 complete cds. TGGTTACAAA 1840. Homo sapiens clone 23596 mRNA AF038203 sequence. TATTTATCCA 1841. Human mRNA for leukocyte-associated Y00796 molecule-I alpha subunit (LFA- 1 TATTTTGGAG 1842. TCAACAGCAG 1843. TCAAGAATCC 1844. TCACAGTGCC 1845. TACTTAATTG 1846. TATGCCCTAT 1847. GCTTTTCAGA 1848. Human VEGF related factor isoform U43368 VRF186 precursor (VRF) mRNA. SUBSTITUTE SHEET (RULE 26) WO 99/65924 PCT/US99/13800 118 TAG SEQ ID DESCRIPTION ACCESSION I NO TTGCTTCTTA 1849. TTGGGGTTTA 1850. ITTGGGTTTTC 1851. Homo sapiens can mRNA. X64228 S89 ITTGTGAAGGA 1852. TTGTGGAAAG 1853. TTTAAAACTT 1854. TACCACAGCC 1855. TACCAGAGTC 1856. Homo sapiens mRNA for serotonin Z36748 receptor. TACCAGCCAG 1857. TACCCAGGGC 1858. TACCTTTTCC 1859. TTTCTGCTAA 1860. Homo sapiens mitochondrial HSP75 L 15189 mRNA, complete cds. TCACATTCCT 1861. TGCTCTTTCC 1862. TGGGTGACCA 1863. TGGGTTAATA 1864. TGGTCCCCCT 1865. Human mRNA for KIAA0028 gene. D21851 - _ partial cds. TGGTGACAGC 1866. _ TTTTATGGGT 1867. Human HepG2 3' region cDNA, clone D16930 hmd4al2. TTTGTGGCTA 1868. TCACTCCTGG 1869. TGGAGCGCTA 1870. TGTGGTGTAG 1871. Human (clone pA3) protein disulfide J05016 isomerase related protein TGCCCGGCAG 1872. TGCCCTGGTT 1873. TGCTTTCAAA 1874. ___ ITGCGTGGCTA 1875. TGGGCCAGCC 1876. TGCTGATAAG 1877. 1 TGCTGTGAAA 1878. 1 TGTGAAGATT 1879. 1 TGTGCCACTA 1880. 1 TGCCATCAAT 1881. TTGATTTCTG 1882. TGCTGGGTAC 1883 TGTGAGCCCT 1884. _ TGTGTTCCTG 1885. TGTTCCAGAT 1886. Human syntaxin 3 mRNA, complete cds. U32315 TGTTTCCTTA 1887. 1 1____ SUBSTITUTE SHEET (RULE 26) WO 99/65924 PCT/US99/13800 119 TAG SEQ ID DESCRIPTION ACCESSION NO TGCGGGCCTG 1888. ,TCAGAACAGT 1889. Human G-rich sequence factor-I (GRSF- U0723 I _ 1) mRNA, complete cds. TCTAGCTGGA 1890. TCTAAAGAGT 1891. [TCGGGTTTAC 1892. TCGGGTCCCT 1893. TCGGAGCCCC 1894. TCCCATCATA 1895. TCCAGCTCTG 1896. TCCAATACTG 1897. Human dynamitin mRNA, complete cds. U50733 TCATCTTCAA 1898. Human autoantigen caireticulin mRNA, M84739 complete cds. TCATCTCCCT 1899. TCTGGCTGGG 1900. TCAGTGGTAG 1901. TCACTGGGGA 1902. TGGGCTCCTC 1903. TCAGAAGTTT 1904. TCAGACGCGG 1905. TCAGGCTGTT 1906. Homo sapiens mRNA for beta-centractin X82207 (PC3). TCAGTGCGCA 1907. TGCTGTGGGG 1908. TGCTTGCAAC 1909. Homo sapiens short form transcription AF055376 factor C-MAF (c-maf) mRNA, TTGCTAAAGG 1910. TGGACTGGTA 1911. TTTCATTGCC 1912. Homo sapiens full length insert cDNA AF075051 YN99C01. TGGATATGAA 1913. TTTCTCTAAG 1914. AGAGTAACTG 1915. TGGATTGCCA 1916. GAGCCCCTTG 1917. GTGGTGTGCC 1918. GTGTGGGAGA 1919. GTGTTACCCA 1920. GTGTTCTGTG 1921. GTTAACTGGG 1922. Homo sapiens mRNA for putative RNA AJ223948 helicase, 3' end. GGAGGCTGGA 1923. Human cell adhesion molecule L I M74387 (LI CAM) mRNA, complete cds. GTGGCACGCG 1924. 1 SUBSTITUTE SHEET (RULE 26) WO 99/65924 PCTIUS99/13800 120 TAG SEQ ID DESCRIPTION ACCESSION NO GATGGCAGGGI 1925. GCACAAAGGG 1926. GACACACAGA 1927. GACCACACAC 1928. GGCAGCCAGG 1929. GAGCAGGAGC 1930. Homo sapiens mRNA for KIAA0600 AB0 l1172 protein, partial cds. GTGGCTCGTG 1931. Homo sapiens (xs 130) mRNA, 260bp. Z36786 GAGGATCTGC 1932. GAGGCAAGAC 1933. GAATACGCAC 1934. GATAGAGGGA 1935. GAACTCAGGC 1936. Human HCFI gene related mRNA L20010 sequence. GCAAATATAT 1937. GCAAATCTGA 1938. GCAACGGCCC 1939. GCAACTGCAC 1940. GCAAGAAGAA 1941. AGATTTGGAA 1942. GACTCAGGGA 1943. TACACCAAGA 1944. GTTGTGGCTA 1945. GTTGTGGTAC 1946. GTTTGCCTGA 1947. TAAAAGGATG 1948. TAAAGCAGTA 1949. Homo sapiens mRNA for restin. X64838 S38 GTGAGCAAGA 1950. Human mRNA for a presumptive KDEL X55885 receptor. TAAGAAGCTT 1951. GTTATATCCA 1952. TAATCACCAG 1953. TAATGAACTA 1954. Homo sapiens mRNA for KIAA0639 ABO14539 protein, partial cds. TAATGGGAGT 1955. GTGGTGTGCA 1956. Homo sapiens RNA transcript from U17 AJ006835 small nucleolar RNA host gene, TAATTTGAA 1957. GTTCACATTT 1958. TACAGAGCCC 1959. TAAAGTGTCT 1960. GTGGTGGGCG| 1961. GTGATGTACG 1 1962. 1 SUBSTITUTE SHEET (RULE 26) WO 99/65924 121 PCT/US99/13800 TAG SEQ ID DESCRIPTION NO LcESO GTGATTTTAC 1963. Homo sapiens putative protein kinase AF062077 _________regulator mRNA. complete cds. GTGCCCAGTC 1964. Homo sapiens mRNA for KIAA06 AB014533 __________protein, partial cds. GTGCCCTTGA 1965. GTGCGCTGAC 1966. Human MHC class I HLA-CwI gene. M26429 _______________________complete cds. GTGCTAAGCG 1967. Human mRNA for collagen VI alpha-2 C- X15882 ____________ ________terminal globular domain. GTGCTGGCAG 1968. GTTCTGCCTC 1969. GAGTACCCCT 1970. TAATGGTAGC 1971. GCAGAAAGTT 1972. Homo sapiens diphthamide biosynthesis AF053003 ____________ ________protein-2 (DPH2) m.RNA,______ GCTGTAGGGG 1973. GCGAAACCTT 1974. GCTTATGTTA 1975. GCTTCCTAAG 1976. ______ _______ GCTTCTGAAC 1977. ____________ GGAAGGCAAG 1978. ___________ GGAAGGTGGA 1979. ___________ GGAAGTTTCG 1980. ___________ TACAGCACGG 1981. Homo sapiens microsomal glutathione S- AF026977 transferase 3 (MGST3) mR.NA, GCTGGTTCCT 1982. ______ _______ GCCTCCCCCA 1983. ______ _______ GCAAGGCAGA 1984. ____________ GCACTTCAAA 1985. Homo sapiens clone 24675 mRNA AF070585 sequence. GCTAGG1TTTA 1986. GCAGAGCAGT 1987. Human LL-l protein LRNA, complete M22637 complet.cds. GCAGGACCCT 1988. GCATTTAG'IT 1989. GCCAAACTTG 1990. GCGAGCTGGC 1991._ GCCGGCCGGA 1992. GCGACCAACA 1993. GCCTGCTTGG 1994. GCCTGTTGGG 1995. GCCTGTTTGG 1996. Human bilirubin UDP- M57899 glucuronosyitransferase isozyme 1 ______ ___ mRNA, SUBSTITUTE SHEET (RULE 26) WO 99/65924 PCT/US99/13800 122 TAG SEQ ID DESCRIPTION ACCESSION NO GCGAAACCGC I 1997. GCACGCGTAA 1998. CTGTTTGTCA 1999. CTGGCCTGTA [ 2000. CTCATAGGGA 2001. CTCCTACCTG 2002. CTCTCATCTC 2003. CTCTCTGTGG 2004. CTCTTCAGGA 2005. Homo sapiens phosphomevalonate kinase L77213 mRNA, complete cds. CTGAGTTAGG 2006. CTGCCCCACA 2007. Homo sapiens nuclear protein Skip U51432 mRNA, complete cds. GACAAAGCAA 2008. CTGGCCGACT 2009. Homo sapiens p160 mRNA, partial cds. U88153 GCACAGAGCC 2010. GCAATTCACC 2011. CTGTGCCAAT 2012. GCTGATCTGT 2013. CTTATGTATT 2014. CTTCTTTCCA 2015. CTTTTCAAGA 2016. Homo sapiens, gene for Membrane X59405 cofactor protein. GAAAGATTGC 2017. CTGCCCTGGG 2018. GCTGTGGTCC 2019. Human HepG2 3' region cDNA, clone D16918 hmd3d 10. GCGATGGGGG 2020. GCGCACCGCT 2021. GCGGAAACTG 2022. GCGGCCACCA 2023. GCGTGCTCTC 2024. TAAGTCTATA 2025. Homo sapiens RNA for Fc receptor, X62572 YOO PC23. CTGGGATCAT 2026. AAGGCAAAGA 2027. AGGGCTTTCC 2028. ACCTACAACG 2029. AGTTCTATGG 2030. Homo sapiens clone 23728 mRNA AF038199 ______________ _________sequence. AAAATAAACA 2031. sequence. AAGATCCTCA 2032. AAGAGCTAAT 2033. Human mRNA fragment for glutaminvl- X07466 tRNA synthetase (EC 6.1.1.18). SUBSTITUTE SHEET (RULE 26) WO 99/65924 PCT/US99/13800 123 TAG SEQ ID DESCRIPTION ACCESSION NO AAGAAACTAA 2034. Homo sapiens mRNA for telomeric DNA X93S 11 __________ 2035.__ binding protein (orfl). AACGTTCTTG 2035. _____________ AACATTGGCT 2036. AACAGCTTTA 2037. AACAATTGGG 2038. Homo sapiens EWS/CHOP chimeric X92 120 fragment. GTTGGTCCTC 2039. AAACTCGAGC 2040. AGTAAACTGA 2041. ATCTTTTCTC 2042. Human eosinophil Charcot-Leyden LO 1664 crystal (CLC) protein ATGACTGTGC 2043. ATGATACCTG 2044. ATGATTTCAG 2045. ATGGACCCCG 2046. ATGGCACCAT 2047. ATGGTGTATG 2048. AACAACTGGC 2049. NAT=CpG island-associated gene S78771 [human, mRNA, 1741 nt]. AATGTAATCA 2050. Human sorcin (SRI) mRNA, complete L12387 cds. CTCAGTGGAA 2051. ACCGTATTCC 2052. ACTGCTCATT 2053. ATCCCACTGA 2054. GCCAGCTGTG 2055. ATTCCTAGGG 2056. AGAGCAAAAA 2057. AGGCTTCTCA 2058. Human sialophorin (CD43) mRNA, J04536 complete cds. AGACCACAAC 2059. AGAATGCTGA 2060. Human myeloid progenitor inhibitory U85767 factor-I MPIF- 1 mRNA, complete AGAAGCTGTG 2061. AGAAAAAAAC 2062. ACTTCTGGAA 2063. ACTTAGGCTT 2064. ACTGTGCCAC 2065. AGGTACGGAA 2066. AGTGCCGTGT 2067. Human interferon-induced cellular M30817 resistance mediator protein (MxA) AGGTTTTCTA 2068. Homo sapiens embryonic ectoderm AF070418 development protein mRNA. partial SUBSTITUTE SHEET (RULE 26) WO 99/65924 PCT/US99/13800 124 TAG SEQ ID DESCRIPTION ACCESSION NO ATCCATCTGG 2069. ATCATTACTA 2070. ATATACTGTA 2071. ATAGGATACT 2072. ATAAGGTACA 2073. AGTTGTCCCG 2074. Homo sapiens clone 24561 unknown AF055010 mRNA. partial cds. AGGCCACCTC 2075. AGTGTCCCGG 2076. AGGCGGAGGT 2077. AGTAGTCTGC 2078. AGTACCTGTC 2079. ACCAACACAC 2080. AGAGGGTGGG 2081. GGGACGAGAA 2082. ACCGGCGTGG 2083. GTCGTTGGTG 2084. GTCTTAACTC 2085. Homo sapiens Dimip homolog (hdiml+) AF023611 mRNA, complete cds. GTGACGCCCC 2086. Homo sapiens full length insert cDNA AF086408 clone ZD76G10. CTCAACAATG 2087. GTAGATGATG 2088. GGGCCGCTCA 2089. Homo sapiens mRNA for KIAA0602 ABO 11174 protein, partial cds. GGCAGCTGGA 2090. GGCAGGCCTG 2091. GGCAGTGACT 2092. GGCCCACACC 2093. GTCCCTCAGC 2094. GGGAAATCCC 2095. GTCCCACGGG 2096. GGTCTl'CTCT 2097. GGGCCCCCAA 2098. GTGATGGGGA 2099. GGGCTCTGAG 2100. GGGGAGTAGG 2101. GGGGCAAGTG 2102. GGGGCTGTGG 2103. Human TFIIIC Box B-binding subunit U02619 mRNA, complete cds. GGGGGAAAAT 2104. GGGTGTCACT 2105. GGTAGGGGTT 2106. Homo sapiens ubiquitin conjugating AF075599 enzyme 12 (UBCI2) mRNA, complete SUBSTITUTE SHEET (RULE 26) WO 99/65924 PCT/US99/13800 125 TAG SEQ ID DESCRIPTION ACCESSION NO GGGCATTTCT 2107. AGATCTGGGA 2108. -- ~~ GGCCCATATG 2109. ----- ACAGGAAACT 2110. GTTGAGAGAG 2111. - ACATTTCAAC 2112. ACATTGGTAA 2113. AAGATGCACA 2114. Human mRNA for phosphodiesterase I D45421 alpha, complete cds. ACAAGACGGC 2115. _ AAGGAGTTCC 2116. | AATGGGAGTT 2117. AATGCTTGAT 2118. Homo sapiens IEF 7442 mRNA. X72841 AATGCCCCAC 2119. AATCAAGGTG 2120. AATAAAGCAA 2121. _ GTCCTCAAGC 2122. ACTGCCCCAA 2123. Homo sapiens full length insert cDNA AF086245 clone ZD38B07. ACCCGCCGGC 2124. GTCATACACC 2125. GTGAGGGGTG 2126. GGTGACTTCA 2127. Human DNase l-Like III protein U56814 (DNAS IL3) mRNA, complete cds. GGTGCTTATG 2128. GGTGTCTCGC 2129. GGTTAATTGA 2130. GGTTCCTGGC 2131. GGTTGGGGTA 2132. GGTTGGTGGT 2133. GGTCATTGTA 2134. | GTATAATAGC 2135. Human mRNA for U2 snRNP-specific A' X13482 protein. GGTCAGTCTC 2136. AAGGTGCATA 2137. Human Kox15 mRNA for zinc finger X52346 protein, partial. SUBSTITUTE SHEET (RULE 26)

Claims (24)

1. An isolated population of polynucleotides comprising or corresponding to at least one polynucleotide shown in Table I and their respective complements.
2. A polynucleotide encoding a ligand or antibdoy or engineered protein that 5 binds to a cell surface protein of an antigen presenting cell and wherein the polynucleotide comprises or corresponds to a polynucleotide shown in Table 1 or its complement.
3. A polynucleotide that encodes a transcription factor and wherein the polynucleotide comprises or corresponds to a polynucleotide shown in Table 1 10 or its complement.
4. A polynucleotide comprising a first polynucleotide comprising encoding an immunostimulatory factor that is differentially expressed in an antigen presenting cell and comprising or corresponding to a tag shown in Table 1 or its complement. 15
5. The polynucleotide of claim 4, wherein the first polynucleotide encodes a factor selected from the group consisting of PARC, TARC, monocyte chemoattractant protein-4 (MiDP-4), MDC, escalectin, MCP-2 or a biologically active fragments thereof
6. The polynucleotide of claim 4, further comprising a first and second promoter, 20 wherein the first and second polynucleotides are under the transcriptional control of the first and second promoters, respectively.
7. The polynucleotide of claim 4, further comprising a single promoter, wherein the first and second polynuceotides are under the transcriptional control of the single promoter. 25
8. A novel polynucleotide comprising or corresponding to a tag shown in Table 1.
9. A gene delivery vehicle comprising a polynucleotide of claims 1-8.
10. A host cell that comprises a polynucleotide of claims 1-8.
11. An array of probes comprising a polynucleotide of claims 1-8 bound to a chip. 30
12. A polynucleiotide comprising a first polynucleotide comprising encoding an immunostimulatory factor that is differentially expressed in an antigen presenting cell that is differentially expressed in an antigen presenting cell and WO 99/65924 127 PCT/US99/13800 comprising or corresponding to a tag shown in Table 1 and second polynucleotide that modulates the expression of the first polvnucleotide.
13. A polynucleotide comprising a first polynucleotide encoding an antigen and a second polynucleotide that modulates the expression of a third polvnucleotide 5 which encodes an immunstimulatory factor that is differentially expressed in an antigen presenting cell, wherein the third polynucleotide comprises or corresponds to a tag shown in Table 1.
14. The polynucleotide of claims 13 or 14, wherein the first polynucleotide encodes PARC, monocyte chemoattractant protein-4 (MDP-4), MDC, 10 escalectin, MCP-2 or a biologically active fragments thereof
15. A gene delivery vehicle comprising the polynucleotides of claims 12-14.
16. A host cell comprising the polynucleotides of claims 12-14.
17. A polynucleotide comprising a first polynucleotide encoding an engineered protein or polypeptide that binds to a cell surface protein of antigen presenting 15 cells thereby modulating either directly or indirectly by a signal transduction pathway and a second polynucleotide encoding an immunostimulatory factor comprising or corresponding to a tag shown in Table 1.
18. The polynucleotide of claim 17, further comprising a first and second promoter, wherein the first and second polynucleotides are under the 20 transcriptional control of the first and second promoters, respectively.
19. The polynucleotide of claims 17, further comprising a single promoter, wherein the first and second polynucleotides are under the transcriptional control of the single promoter.
20. A gene delivery vehcile comprising the polynucleotides of claims 17-19. 25
21. A host cell comprising th epolynucleotides of claims 17-19.
22. A emthod for inducing an immune response in a subject comprising administering an effective amount of the polynucleotide of claim 1, 12 or 17, to the subject.
23. A method of modulating the genotype of an antigen presenting cell, 30 comprising introducing into the cell a polynucleotide of claims 1, 12 or 17. SUBSTITUTE SHEET (RULE 26) WO 99/65924 128 PCTIUS99/13800
24. A method of screening for a candidate therapeutic agent that modulates the expression of a polynucleotide differentially expessed in an antigen presenting comprising contacting a cell with an effective amount of a potential agent, and assaying for a change in expression level of a polynucleotide of claim 2, 5 wherein a change in the expression level is indicative of a candidate therapeutic agent. SUBSTITUTE SHEET (RULE 26)
AU52044/99A 1998-06-19 1999-06-18 Preparation and use of superior vaccines Abandoned AU5204499A (en)

Applications Claiming Priority (59)

Application Number Priority Date Filing Date Title
US8999998P 1998-06-19 1998-06-19
US9003998P 1998-06-19 1998-06-19
US9003698P 1998-06-19 1998-06-19
US9007698P 1998-06-19 1998-06-19
US9000098P 1998-06-19 1998-06-19
US8984498P 1998-06-19 1998-06-19
US9004298P 1998-06-19 1998-06-19
US9004398P 1998-06-19 1998-06-19
US8983398P 1998-06-19 1998-06-19
US9004098P 1998-06-19 1998-06-19
US9004498P 1998-06-19 1998-06-19
US9007898P 1998-06-19 1998-06-19
US9008098P 1998-06-19 1998-06-19
US8987898P 1998-06-19 1998-06-19
US8985398P 1998-06-19 1998-06-19
US8999798P 1998-06-19 1998-06-19
US9004598P 1998-06-19 1998-06-19
US8999398P 1998-06-19 1998-06-19
US8999298P 1998-06-19 1998-06-19
US9003598P 1998-06-19 1998-06-19
US9004798P 1998-06-19 1998-06-19
US8999198P 1998-06-19 1998-06-19
US9004898P 1998-06-19 1998-06-19
US9007298P 1998-06-19 1998-06-19
US9007798P 1998-06-19 1998-06-19
US9007998P 1998-06-19 1998-06-19
US9004198P 1998-06-19 1998-06-19
US8999498P 1998-06-19 1998-06-19
US60090041 1998-06-19
US60090078 1998-06-19
US60090077 1998-06-19
US60090036 1998-06-19
US60089999 1998-06-19
US60090040 1998-06-19
US60089844 1998-06-19
US60090044 1998-06-19
US60089997 1998-06-19
US60089853 1998-06-19
US60089878 1998-06-19
US60089993 1998-06-19
US60090080 1998-06-19
US60090072 1998-06-19
US60090047 1998-06-19
US60089833 1998-06-19
US60090039 1998-06-19
US60090042 1998-06-19
US60090000 1998-06-19
US60090043 1998-06-19
US60090076 1998-06-19
US60090079 1998-06-19
US60090045 1998-06-19
US60090048 1998-06-19
US60090035 1998-06-19
US60089992 1998-06-19
US60089994 1998-06-19
US60089991 1998-06-19
US11171598P 1998-12-08 1998-12-08
US60111715 1998-12-08
PCT/US1999/013800 WO1999065924A2 (en) 1998-06-19 1999-06-18 Preparation and use of superior vaccines

Publications (1)

Publication Number Publication Date
AU5204499A true AU5204499A (en) 2000-01-05

Family

ID=27586849

Family Applications (2)

Application Number Title Priority Date Filing Date
AU48256/99A Abandoned AU4825699A (en) 1998-06-19 1999-06-17 Identification and use of differentially expressed genes and polynucleotide sequences
AU52044/99A Abandoned AU5204499A (en) 1998-06-19 1999-06-18 Preparation and use of superior vaccines

Family Applications Before (1)

Application Number Title Priority Date Filing Date
AU48256/99A Abandoned AU4825699A (en) 1998-06-19 1999-06-17 Identification and use of differentially expressed genes and polynucleotide sequences

Country Status (5)

Country Link
EP (1) EP1086215A4 (en)
JP (1) JP2002534056A (en)
AU (2) AU4825699A (en)
CA (1) CA2335452A1 (en)
WO (1) WO1999065924A2 (en)

Families Citing this family (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6825333B1 (en) 1999-08-20 2004-11-30 Chiron Corporation EGFH2 genes and gene products
US7041445B2 (en) * 1999-11-15 2006-05-09 Clontech Laboratories, Inc. Long oligonucleotide arrays
US20020072503A1 (en) * 2000-03-28 2002-06-13 Jiangchun Xu Compositions and methods for the therapy and diagnosis of ovarian cancer
US6632630B2 (en) 2000-03-29 2003-10-14 Aclara Biosciences, Inc. Monooxygenase assays
JP2006515266A (en) 2002-04-22 2006-05-25 バイオニケ ライフ サイエンシーズ インコーポレイテッド Oligonucleotide compositions and their use for modulating immune responses
WO2003101401A2 (en) 2002-06-03 2003-12-11 Chiron Corporation Use of nrg4, or inhibitors thereof, in the treatment of colon and pancreatic cancer
WO2008047128A2 (en) * 2006-10-18 2008-04-24 Nazneen Rahman Materials and methods for determining susceptibility to cancer
CN102639711A (en) * 2009-10-12 2012-08-15 医学免疫有限责任公司 Quantification of IR-A and IR-B for tumor classification
EP2322656A1 (en) * 2009-11-17 2011-05-18 Centre National de la Recherche Scientifique (C.N.R.S) Methods for diagnosing skin diseases

Family Cites Families (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE3940598A1 (en) * 1989-12-08 1991-06-13 Behringwerke Ag TOXOPLASMA GONDII-ANTIGENE, THEIR PRODUCTION AND USE
JPH03503524A (en) * 1985-10-30 1991-08-08 バイオジェン インコーポレイテッド Cleavage dimer of Mullerian canal inhibitor-like polypeptide
HUT53672A (en) * 1988-02-25 1990-11-28 Gen Hospital Corp Quick immunoselective cloning process
WO1989008254A1 (en) * 1988-03-04 1989-09-08 La Jolla Cancer Research Foundation Germ cell alkaline phosphatase gene and protein produced thereby
EP0359919B1 (en) * 1988-06-29 1996-02-28 Ml Technology Ventures, L.P. Recombinant mycoplasma hyopneumoniae antigen and uses therefor
ATE123810T1 (en) * 1988-08-23 1995-06-15 Dana Farber Cancer Inst Inc ALPHA SUBUNITY OF THE LFA-1 LEUKOCYTE ADHESION RECEPTOR.
US5191064A (en) * 1988-09-30 1993-03-02 The Research Foundation For Microbial Diseases (Osaka University) Non-a, non-b hepatitis virus antigen peptide
WO1990008160A1 (en) * 1989-01-20 1990-07-26 Imperial Cancer Research Technology Ltd. Growth factor receptor-like peptides without tyrosine-kinase activity
WO1991008217A1 (en) * 1989-12-05 1991-06-13 The Regents Of The University Of California Human intestinal mucins
US6238861B1 (en) * 1990-06-29 2001-05-29 The Regents Of The University Of Michigan Neurofibromatosis gene
US5866330A (en) * 1995-09-12 1999-02-02 The Johns Hopkins University School Of Medicine Method for serial analysis of gene expression

Also Published As

Publication number Publication date
CA2335452A1 (en) 1999-12-23
EP1086215A4 (en) 2005-10-19
EP1086215A2 (en) 2001-03-28
JP2002534056A (en) 2002-10-15
WO1999065924A2 (en) 1999-12-23
AU4825699A (en) 2000-01-05
WO1999065924A3 (en) 2000-04-13

Similar Documents

Publication Publication Date Title
US20060134682A1 (en) Preparation and use of superior vaccines
US6218529B1 (en) Biomarkers and targets for diagnosis, prognosis and management of prostate, breast and bladder cancer
KR101204241B1 (en) Compositions and Methods for the Detection, Diagnosis and Therapy of Hematological Malignancies
AU766789B2 (en) Novel uses
US20020018766A1 (en) Genes differentially expressed in cancer cells to design cancer vaccines
WO1999065928A2 (en) Polynucleotide population isolated from non-metastatic and metastatic breast tumor tissues
CZ20023567A3 (en) Compounds and methods for therapy and diagnosis of lung carcinoma
WO1998004689A1 (en) Biomarkers and targets for diagnosis, prognosis and management of prostate disease
JP2001501447A (en) Compositions and methods for treatment and diagnosis of breast cancer
JP2007518399A (en) Compositions and methods for diagnosing and treating lung cancer
JP2002533056A (en) Compounds and methods for treatment and diagnosis of lung cancer
JP2002543769A (en) Compounds and methods for treatment and diagnosis of lung cancer
KR20110015409A (en) Gene expression markers for inflammatory bowel disease
EP1127893A2 (en) Compositions and methods for the treatment and diagnosis of breast cancer
JP2010239970A (en) Composition for treatment and diagnosis of breast cancer, and method for use thereof
US6686147B1 (en) Cancer associated antigens and uses therefor
EP1536006A1 (en) Cancer antigens and utilization thereof
AU5204499A (en) Preparation and use of superior vaccines
US20020137077A1 (en) Genes regulated in activated T cells
US20020111467A1 (en) Compositions and methods for the treatment and diagnosis of breast cancer
US6635255B1 (en) CASB414:antigen overexpressed in several tumors
US20030175704A1 (en) Genes expressed in lung cancer
US20020086303A1 (en) Compositions and methods for the therapy and diagnosis of colon cancer
JP2002507425A (en) Human CASB12 polypeptide which is a serine protease
US20030194764A1 (en) Compositions and methods for the therapy and diagnosis of lung cancer

Legal Events

Date Code Title Description
MK5 Application lapsed section 142(2)(e) - patent request and compl. specification not accepted