AU1492601A - Sequences characteristic of bladder cancer - Google Patents

Sequences characteristic of bladder cancer Download PDF

Info

Publication number
AU1492601A
AU1492601A AU14926/01A AU1492601A AU1492601A AU 1492601 A AU1492601 A AU 1492601A AU 14926/01 A AU14926/01 A AU 14926/01A AU 1492601 A AU1492601 A AU 1492601A AU 1492601 A AU1492601 A AU 1492601A
Authority
AU
Australia
Prior art keywords
sequences
bladder cancer
gene
tcc
rna
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
AU14926/01A
Inventor
Orna Mor
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.)
Quark Pharmaceuticals Inc
Original Assignee
Quark Biotech Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Quark Biotech Inc filed Critical Quark Biotech Inc
Publication of AU1492601A publication Critical patent/AU1492601A/en
Abandoned legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6876Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
    • C12Q1/6883Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material
    • C12Q1/6886Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material for cancer
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/158Expression markers

Landscapes

  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Organic Chemistry (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Engineering & Computer Science (AREA)
  • Immunology (AREA)
  • Pathology (AREA)
  • Analytical Chemistry (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • Genetics & Genomics (AREA)
  • Hospice & Palliative Care (AREA)
  • Biochemistry (AREA)
  • Microbiology (AREA)
  • Molecular Biology (AREA)
  • Biophysics (AREA)
  • Physics & Mathematics (AREA)
  • Oncology (AREA)
  • Biotechnology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • General Engineering & Computer Science (AREA)
  • General Health & Medical Sciences (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Description

WO 01/22864 PCT/USOO/41005 SEQUENCES CHARACTERISTIC OF BLADDER CANCER BACKGROUND OF THE INVENTION FIELD OF THE INVENTION The present invention relates to the identification of polynucleotide sequences that are differentially expressed in bladder cancer. More specifically, the present invention relates to the use of the sequences and gene products for diagnosis and as probes. DESCRIPTION OF RELATED ART Bladder cancer is the second most-common genitourinary cancer in the United States, with only prostate cancer being more frequently diagnosed. Bladder cancer accounts for approximately two percent of all malignant tumors and approximately seven percent of all urinary tract malignancies in U.S. men. Over 54,000 new cases were estimated to be diagnosed in the United States in 1998, with approximately 12,500 deaths predicted [American Cancer Society, 1998]. The prevalence of bladder cancer is higher in industrialized nations, perhaps reflecting increased exposure to environmental carcinogens. Men are three times more frequently affected than women. The disease usually occurs between 60-70 years of age and the age-adjusted bladder cancer rate in white men is almost twice that of black men. Most bladder cancers (over 90%) are carcinomas of the transitional epithelium of the bladder's mucosal lining (transitional cell carcinoma (TCC)). Although 90 percent of the cases are localized at diagnosis, up to 80 percent recur. - 1- WO 01/22864 PCT/US00/41005 A number of etiological factors are associated with the development of bladder cancer, but in industrialized countries, cigarette smoking is the most significant. Specific chemicals have also been identified as causing bladder cancer, as have a number of occupational exposures to less well-defined specific agents. Treatment with cytostatic drugs, especially cyclophosphamide, is associated with increased risk of bladder cancer, as is treatment with radiotherapy for uterine cancer. Bladder cancer is a potentially preventable disease, with a significant morbidity and mortality in many parts of the world. Tumors are graded according to the degree of cellular abnormality, with the most atypical cells being designated as high-grade (i.e., G3 grade) tumors. The major prognostic factors in carcinoma of the bladder are the depth of invasion into the bladder wall and the degree of differentiation of the tumor. The higher the grade of the tumor at the diagnosis, the higher the incidence of death from the disease within two years. The stage of development of the tumor is significant in estimating disease prognosis. Most superficial, non-invasive tumors being papillary tumors which do not invade the lamina propria, and are classified as non-invasive TCC, i.e., "Ta" tumors, may recur, but nearly 70% will not progress further. A tumor which does not invade the muscle but does enter the lamina propria presents in many cases a worse prognosis. Such tumors are classified as non-invasive TCC, i.e., T1 tumors. Most superficial tumors are well differentiated and classified as G1 grade tumors. Patients in whom superficial tumors are less differentiated, large, multiple, or associated with carcinoma in situ in other areas of the bladder mucosa, (classified as G2-G3 tumors) are at greatest risk for recurrence and the development of invasive cancer. Invasive bladder tumors tend to spread rapidly to -2- WO 01/22864 PCT/US00/41005 the regional lymph nodes and then into adjacent structures. Overall, the five-year survival rate of TCC is 76 percent for whites and 55 percent for blacks. One of the management problems is the fact that carcinoma of the bladder is frequently multifocal. The entire bladder epithelium and the lining of the entire urothelial cell tract can undergo malignant change. After apparently successful treatment of a bladder lesion, new tumors may occur at the same site (recurrence) or in other urothelial cells in the bladder. Approximately 30 percent of bladder carcinomas present as multiple lesions at the time of initial diagnosis. The early diagnosis of bladder cancer is central to the effective treatment of TCC. Presently, the detection of bladder tumors relies on intravenous pyelogram or other contrast studies to rule out urothelial involvement in the kidneys or ureters, and invariably cystoscopy which remains the accepted standard for diagnosis of mucosal abnormalities. There are no presently reliable methods available to easily and specifically identify the presence of bladder cancer cells. A variety of new technologies and potential tumor markers are being studied in bladder cancer and some are being translated into clinical use. It is important to realize that all available results of the diagnostic value of tumor markers do not allow firm clinical recommendations, but tests based on biomarkers will undoubtedly influence the management of bladder cancer in the near future. Several new markers have been already identified and even approved for use (e.g. bladder tumor antigen (BTA) markers, NMP22, FDP). However, their clinical use is limited [Grossman, 1998], due to sensitivity and specificity problems in conjunction with cystoscopic examination. Furthermore, due to the high rate of disease recurrence, follow-up of TCC patients is obligatory. There is a need to eliminate the invasive cytoscopy method of diagnosis and of follow-up and replace it with a reliable and non-invasive method of diagnosis. -3- WO 01/22864 PCT/USOO/41005 Approximately 70-80 percent of patients with newly diagnosed bladder cancer will present with superficial, non-invasive bladder tumors. Those who do are often curable. Tumor patients with deeply invasive disease can sometimes be cured by complete surgical removal of the bladder, irradiation, or a combination of modalities that include chemotherapy, however the five-year survival rate is less likely for such tumors. It is therefore of major importance to detect new tools that will aid in both the initial early diagnosis and in follow-up of non-invasive TCC tumors. Adverse prognostic features associated with a greater risk of disease progression include the presence of multiple aneuploid cell lines, nuclear p53 overexpression, and expression of the Lewis-x blood group antigen [Hudson and Herr, 1995; Lacombe et al., 1996]. It has been postulated that p53 may be useful for predicting the level of aggression of the tumor and to identify patients who will not benefit from chemotherapy. However, only a very small, select group of patients with invasive disease may benefit from this approach [Ozen, 1998]. Several treatment methods (i.e., transurethral surgery, intravesical medications, and cystectomy) have been used in the management of patients with superficial tumors, and each method can be associated with five-year survival in 55-80 percent of patients treated [Hudson and Herr, 1995; Torti and Lum, 1984]. Invasive tumors that are confined to the bladder muscle on pathologic staging after radical cystectomy are associated with an approximately 75 percent, five year progression-free rate of survival. Patients with more deeply invasive tumors (which are also usually less well differentiated) experience five-year survival rates of 20-40 percent following radical cystectomy. When the patient presents with a locally extensive tumor that invades pelvic viscera or with metastases to lymph nodes or distant sites, a five-year survival rate is uncommon, but considerable symptomatic palliation can still be achieved. -4- WO 01/22864 PCT/USOO/41005 Surgery is the main treatment method. The extent of surgery is dependent on the pathological stage of the disease. Early disease is generally treated by intravesical chemotherapy and transurethral resection. Locally invasive disease can usually be managed only by radical cystectomy and urinary diversion. Definitive (curative) radiotherapy is generally reserved for bladder cancer patients who are not candidates for surgery. For superficial, low-grade disease, chemotherapy is applied intravesically (directly into the bladder) to concentrate the drug at the tumor site and eliminate any residual tumor mass after resection. Systemic chemotherapy may also be used to manage advanced bladder cancer; compete response rates of 30-50 percent have been reported. Single agent chemotherapy has demonstrated limited success. However, even following surgery and resection of non-invasive TCC tumors, frequent follow-up is required (every 3 months) in both non-invasive and invasive cases. It would therefore be useful to be able to identify early stage TCC in bladder cancer which has a significantly higher cure rate and generally does not require surgery. In addition, it would be useful to identify markers that can be employed for early diagnosis and follow-up of both non-invasive and invasive TCC, as an efficient and non-invasive alternative to cytoscopy. SUMMARY OF THE INVENTION According to the present invention, there is provided a method of diagnosing the presence of bladder cancer in a patient by analyzing a tissue sample from the patient for the presence of a least one expresses gene wherein the presence of the expressed gene is indicative of bladder cancer. Also provided by the present invention is a polynucleotide sequence whose expression is indicative of bladder cancer. A marker for bladder cancer is also provided. There are also provided methods of diagnosising bladder cancer by screening for the presenceof at least -5- WO 01/22864 PCTIUSOO/41005 one expressed gene wherein the presence of the expressed gene is indicative of bladder cancer. Methods of treating and regulating bladder cancer-associated pathologies by administering to a patient a therapeutically effective amount of a ribozyme, antisense olignucleotide, or agonist against the nucleic acids sequences of the present invention are also provided. DESCRIPTION OF THE INVENTION According to the present invention, purified, isolated and cloned nucleic acid sequences associated with bladder cancer are provided. More specifically, the sequences of the present invention are set forth in Table I or 11 or have a complementary or allelic variation sequence thereto. The nucleic acid sequences of the complementary or allelic variation sequences are provided in Tables Ill and IV, respectively. When referring to bladder cancer, both invasive and noninvasive forms are included. Bladder cancers can also be referred to as transitional cell carcinomas or "TCC". The present invention further provides a method of diagnosing the presence of bladder cancer in a patient, including the steps of analyzing a tissue sample from the patient for the presence of at least one expressed gene (up-regulated) wherein the mRNA from the expressed gene hybridizes to at least one of the sequences in Tables I or II, with hybridization occurring under conditions sufficiently stringent to require at least 95% base pairing. Further the present invention provides antibodies directed against the gene products of the sequences of the present invention. The antibodies can be either monoclonal, polyclonal or recombinant and be used in immunoassays as described in the Methods herein below. - 6- WO 01/22864 PCT/USOO/41005 By regulate or modulate or control is meant that the process is either induced or inhibited to the degree necessary to effect a change in the process and the associated disease state in the patient. Whether induction or inhibition is being contemplated is apparent from the process and disease being treated and is known to those skilled in the medical arts. The present invention identifies genes for gene therapy, diagnostics and therapeutics that have direct causal relationships between a disease and its related pathologies and up- or down regulator (responder) genes. That is, the present invention is initiated by a physiological relationship between cause and effect. The present invention identifies polynucleotide sequences named in Tables I and II, and set forth in Tables III and IV, respectively, that can be utilized diagnostically in bladder cancer. Sequences named in Table I were found to match sequences in data banks and were newly found in the present application to be upregulated in TCC. The sequences named in Table I are either genes with unknown protein product or of unknown genes. All the sequences named in both Tables I and 11 were found to be associated with TCC relative to normal bladder samples. Where the sequences are partial sequences, they are markers or probes for genes that are regulated in bladder carcinoma. By "regulated" it is meant that the genes can be either upregulated or downregulated, depending upon the specific gene. In general these partial sequences are designated "Expressed Sequence Tags" (ESTs) and are markers for the genes actually expressed in vivo and are ascertained as described herein. Generally, ESTs comprise DNA sequences corresponding to a portion of nuclear encoded mRNA. The EST has a length that allows for PCR (polymerase chain reaction), use as a hybridization probe and is a unique designation for the gene with which it hybridizes (generally under conditions sufficiently stringent to require at least 95% base pairing). -7- WO 01/22864 PCT/USOO/41005 . For a detailed description and review of ESTs and their functional utility see WO 93/00353 which is incorporated in its entirety by reference. WO 93/00353 further describes how the EST sequences can be used to identify the transcribed genes. The Example herein also describes a method of identification. The present invention also provides a method of diagnosing the presence of bladder cancer in a patient, by the expression of at least one expressed gene (up regulated) identified by the sequences of the present invention set forth in Tables I and 11. Methods of identification of hybridization results can include, but are not limited to, immunohistochemical staining of the tissue samples. Further for identification of the gene, in situ hybridization, Southern blotting, single strand conformational polymorphism, restriction endonuclease fingerprinting (REF), PCR amplification and DNA-chip analysis using nucleic acid sequence of the present invention as probes/primers can be used. Further, according to the present invention, purified, isolated and cloned bladder cancer associated genes identified by the probes and/or sequences hybridizing under stringent conditions with 95% homology and set forth herein, or a complementary or allelic variation sequence and human homologs, as relevant, thereto, are disclosed. The present invention further provides proteins encoded by the identified genes. The present invention further provides antibodies directed against these proteins. The present invention further provides transgenic animals and cell lines carrying at least one expressible gene identified by the present invention. The present invention further provides knock-out eukaryotic organisms in which at least one nucleic acid sequences as identified by the probes of the present invention and prepared as described in the Methods. The present invention provides a method of identifying bladder cancer, and particularly early stage associated pathologies in a patient. The present invention -8- WO 01/22864 PCT/USOO/41005 provides a treatment by administering to a patient a therapeutically effective amount of an antagonist of at least one protein as encoded by the nucleic acid sequences or sequences identified herein or by the probes of the present invention. Alternatively, the present invention provides a method of regulating bladder cancer-associated pathologies in a patient in need of such treatment by administering to a patient a therapeutically effective amount of at least one antisense oligonucleotide against the nucleic acid sequences or dominant negative peptide directed against the sequences or their proteins. Furthermore, the present invention provides a method of preparation of a library using proprietary methods of library preparation as follows: the SDGI method, as described in US Patent Application USSN 09/538,709 of same under the assignee, filed 30 March, 2000, and incorporated herein by reference in its entirety, the Antisense method as described in US Provisional Patent Application SN 60/157,843 of same assignee, filed 6 October, 1999, and incorporated herein by reference in its entirety, and suppressive subtraction hybridization (SSH) (Diatchenko et al., 1996). Negative dominant peptide refers to a partial cDNA sequence that encodes for a part of a protein, i.e. a peptide (see Herskowitz, 1987). This peptide can have a different function from the protein from which it was derived. It can interact with the full protein and inhibit its activity or it can interact with other proteins and inhibit their activity in response to the full protein. Negative dominant means that the peptide is able to overcome the natural proteins and fully inhibit their activity to give the cell a different characteristics like resistance or sensitization to killing. For therapeutic intervention either the peptide itself is delivered as the active ingredient of a pharmaceutical composition or the cDNA can be delivered to the cell utilizing the same methods as for antisense delivery. The antagonist or regulating agent or active ingredient is dosed and delivered in a pharmaceutically acceptable carrier as described herein below. The term antagonist or antagonizing is used in its broadest sense. Antagonism can -9- WO 01/22864 PCT/USOO/41005 include any mechanism or treatment which results in inhibition, inactivation, blocking or reduction in gene activity or gene product and for example preventing progression from non-invasive to invasive. It should be noted that the inhibition of a gene or gene product provides for an increase in a corresponding function that the gene or gene product was regulating. The antagonizing step can include blocking cellular receptors for the gene products and can include antisense treatment as discussed herein. Many reviews have covered the main aspects of antisense (AS) technology and its enormous therapeutic potential (Wright and Anazodo, 1995). There are reviews on the chemical (Crooke, 1995; Uhlmann et al, 1990), cellular (Wagner, 1994) and therapeutic (Hanania, et al, 1995; Scanlon, et al, 1995; Gewirtz, 1993) aspects of this rapidly developing technology. Within a relatively short time, ample information has accumulated about the in vitro use of AS nucleotide sequences in cultured primary cells and cell lines as well as for in vivo administration of such nucleotide sequences for suppressing specific processes and changing body functions in a transient manner. Further, enough experience is now available in vitro and in vivo in animal models and human clinical trials to predict human efficacy. Antisense intervention in the expression of specific genes can be achieved by the use of synthetic AS oligonucleotide sequences (for recent reports see Lefebvre-d'Hellencourt et al, 1995; Agrawal, 1996; Lev-Lehman et al, 1997). AS oligonucleotide sequences can be short sequences of DNA, typically 15-30 mer but can be as small as 7 mer (Wagner et al, 1996), designed to complement a target mRNA of interest and form an RNA:AS duplex. This duplex formation can prevent processing, splicing, transport or translation of the relevant mRNA. Moreover, certain AS nucleotide sequences can elicit cellular RNase H activity when hybridized with their target mRNA, resulting in mRNA degradation (Calabretta et al, 1996). In that case, RNase H cleaves the RNA component of the duplex and can release the AS to further hybridize with additional molecules of the - 10- WO 01/22864 PCT/USOO/41005 target RNA. An additional mode of action results from the interaction of AS with genomic DNA to form a triple helix which can be transcriptionally inactive. The sequence target segment for the antisense oligonucleotide is selected such that the sequence exhibits suitable energy related characteristics important for oligonucleotide duplex formation with their complementary templates, and shows a low potential for self-dimerization or self-complementation [Anazodo et al., 1996]. For example, the computer program OLIGO (Primer Analysis Software, Version 3.4), can be used to determine antisense sequence melting temperature, free energy properties, and to estimate potential self-dimer formation and self complimentary properties. The program allows the determination of a qualitative estimation of these two parameters (potential self-dimer formation and self complimentary) and provides an indication of "no potential" or "some potential" or "essentially complete potential". Using this program target segments are generally selected that have estimates of no potential in these parameters. However, segments can be used that have "some potential" in one of the categories. A balance of the parameters is used in the selection as is known to those of skill in the art. Further, the oligonucleotides are also selected as needed so that analogue substitution do not substantially affect function, as is known to those of skill in the art. Phosphorothioate antisense oligonucleotides do not normally show significant toxicity at concentrations that are effective, exhibit sufficient pharmacodynamic half-lives in animals (Agarwal et al., 1996) and are nuclease resistant. Antisense induced loss-of-function phenotypes related with cellular development were shown for the glial fibrillary acidic protein (GFAP), for the establishment of tectal plate formation in chick (Galileo et al., 1991) and for the N myc protein which is responsible for the maintenance of cellular heterogeneity in neuroectodermal cultures (ephithelial vs. neuroblastic cells, which differ in their colony forming abilities, tumorigenicity and adherence) (Rosolen et al., 1990; Whitesell et al, 1991). Antisense oligonucleotide inhibition of basic fibroblast growth factor (bFgF), having mitogenic and angiogenic properties, suppressed - 11 - WO 01/22864 PCT/USOO/41005 80% of growth in glioma cells (Morrison, 1991) in a saturable and specific manner. Being hydrophobic, antisense oligonucleotides interact well with phospholipid membranes (Akhter et al., 1991). Following their interaction with the cellular plasma membrane, they are actively (or passively) transported into living cells (Loke et al., 1989), in a saturable mechanism predicted to involve specific receptors (Yakubov et al., 1989). Instead of an antisense sequences as discussed herein above, ribozymes can be utilized. This is particularly necessary in cases where antisense therapy is limited by stoichiometric considerations (Sarver et al., 1990, Gene Regulation and Aids, pp. 305-325). Ribozymes can then be used that target the same sequence. Ribozymes are RNA molecules that possess RNA catalytic ability (see Cech for review) that cleave a specific site in a target RNA. The number of RNA molecules that are cleaved by a ribozyme is greater than the number predicted by stochiochemistry. (Hampel and Tritz, 1989; Uhlenbeck, 1987). Ribozymes catalyze the phosphodiester bond cleavage of RNA. Several ribozyme structural families have been identified including Group I introns, RNase P, the hepatitis delta virus ribozyme, hammerhead ribozymes and the hairpin ribozyme originally derived from the negative strand of the tobacco ringspot virus satellite RNA (sTRSV) (Sullivan, 1994; U.S. Patent No. 5,225,347, columns 4-5). The latter two families are derived from viroids and virusoids, in which the ribozyme is believed to separate monomers from oligomers created during rolling circle replication (Symons, 1989 and 1992). Hammerhead and hairpin ribozyme motifs are most commonly adapted for trans-cleavage of mRNAs for gene therapy (Sullivan, 1994). The ribozyme type utilized in the present invention is selected as is known by those of skill in the art. Hairpin ribozymes are now in clinical trial and are the preferred type. In general, the ribozyme is from 30-100 nucleotides in length. Modifications or analogues of nucleotides can be introduced to improve the therapeutic properties of the nucleotides. Improved properties include increased - 12- WO 01/22864 PCTIUSOO/41005 nuclease resistance and/or increased ability to permeate cell membranes. Nuclease resistance, where needed, is provided by any method known in the art that does not interfere with biological activity of the antisense oligodeoxy nucleotides, cDNA and/or ribozymes as needed for the method of use and delivery (lyer et al., 1990; Eckstein, 1985; Spitzer and Eckstein, 1988; Woolf et al., 1990; Shaw et al., 1991). Modifications that can be made to oligonucleotides in order to enhance nuclease resistance include, but are not limited to, modifying the phophorous or oxygen heteroatom in the phosphate backbone. These modifications also include preparing methyl phosphonates, phosphorothioates, phosphorodithioates and morpholino oligomers. In one embodiment, the modification is provided by having phosphorothioate bonds linking between the four to six 3'-terminus nucleotide bases. Alternatively, phosphorothioate bonds can link all the nucleotide bases. Other modifications known in the art can be used where the biological activity is retained, but the stability to nucleases is substantially increased. The present invention also includes all analogues of, or modifications to, an oligonucleotide of the invention that does not substantially affect the function of the oligonucleotide. The nucleotides can be selected from naturally occurring or synthetic modified bases. Naturally occurring bases include adenine, guanine, cytosine, thymine and uracil. Modified bases of the oligonucleotides include xanthine, hypoxanthine, 2-aminoadenine, 6-methyl, 2-propyl and other alkyl adenines, 5-halo uracil, 5-halo cytosine, 6-aza cytosine and 6-aza thymine, psuedo uracil, 4-thiuracil, 8-halo adenine, 8-aminoadenine, 8-thiol adenine, 8 thiolalkyl adenines, 8-hydroxyl adenine and other 8-substituted adenines, 8-halo guanines, 8-amino guanine, 8-thiol guanine, 8-thioalkyl guanines, 8-hydroxyl guanine and other substituted guanines, other aza and deaza adenines, other aza and deaza guanines, 5-trifluoromethyl uracil and 5-trifluoro cytosine. -13- WO 01/22864 PCTIUSOO/41005 In addition, analogues of nucleotides and/or polynucleotides can be prepared wherein the structure of the nucleotide and/or polynucleotide is fundamentally altered and that are better suited as therapeutic or experimental reagents. An example of a nucleotide analogue is a peptide nucleic acid (PNA) wherein the deoxyribose (or ribose) phosphate backbone in DNA (or RNA) is replaced with a polyamide backbone which is similar to that found in peptides. PNA analogues have been shown to be resistant to degradation by enzymes and to have extended lives in vivo and in vitro. Further, PNAs have been shown to bind stronger to a complementary DNA sequence than a DNA molecule. This observation is attributed to the lack of charge repulsion between the PNA strand and the DNA strand. Other modifications that can be made to oligonucleotides include polymer backbones, cyclic backbones, or acyclic backbones. The active ingredients of pharmaceutical compositions can include oligonucleotides that are nuclease resistant as are needed for the practice of the invention or a fragment thereof shown to have the same effect when targeted against the appropriate sequence(s) and/or ribozymes. Combinations of active ingredients as disclosed in the present invention can be used, including combinations of antisense sequences. The antisense oligonucleotides (and/or ribozymes) and cDNA of the present invention can be synthesized by any method known in the art for ribonucleic or deoxyribonucleic nucleotides. For example, an Applied Biosystems 380B DNA synthesizer can be used. When fragments are used, two or more such sequences can be synthesized and linked together for use in the present invention. The nucleotide sequences of the present invention can be delivered either directly or with viral or non-viral vectors. When delivered directly the sequences are generally rendered nuclease resistant. Alternatively the sequences can be incorporated into expression cassettes or constructs such that the sequence is expressed in the cell as discussed herein below. Generally the construct contains - 14 - WO 01/22864 PCT/USOO/41005 the proper regulatory sequence or promotor to allow the sequence to be expressed in the targeted cell. The proteins of the present invention can be produced recombinantly (see generally Marshak et al, 1996 "Strategies for Protein Purification and Characterization. A laboratory course manual.", CSHL Press) and analogues can be due to post-translational processing. The term analogue as used herein is defined as a nucleic acid sequence or protein which has some differences in their amino acid/nucleotide sequences as compared to the native sequence of the sequences disclosed herein. Ordinarily, the analogue is generally at least 70% homologous over any portion that is functionally relevant. In more preferred embodiments the homology is at least 80% and can approach 95% homology to the protein/nucleotide sequence. The amino acid or nucleotide sequence of an analog can differ from that of the primary sequence when at least one residue is deleted, inserted or substituted, but the protein or nucleic acid molecule remains functional. Differences in glycosylation can provide protein analogues. "Functionally relevant" refers to the biological property of the molecule and in this context means an in vivo effector or antigenic function or activity that is directly or indirectly performed by a naturally occurring protein or nucleic acid molecule. Effector functions include, but are not limited to include, receptor binding, any enzymatic activity or enzyme modulatory activity, any carrier binding activity, any hormonal activity, any activity in promoting or inhibiting adhesion of cells to extracellular matrix or cell surface molecules, or any structural role as well as having the nucleic acid sequence encode functional protein and can be expressible. The antigenic functions essentially mean the possession of an epitope or antigenic site that is capable of cross-reacting with antibodies raised against a naturally occurring protein. Biologically active analogues share an -15- WO 01/22864 PCT/USOO/41005 effector function of the native which can, but do not necessarily, additionally possess an antigenic function. The above discussion provides a factual basis for the use of the sequences of the present invention to identify bladder cancer-associated genes and provide diagnostic probes and markers to identify bladder cancer, particularly in the early stages of TCC. EXAMPLES METHODS A detailed description of the methods of the present invention are set forth below and are set forth in US patent application USSN 09/534,661 of the applicant company, filed on March 24, 2000 and incorporated herein by reference in its entirety. The method includes preparing cell fractionations; extracting intact total RNA from membrane bound polysomes and free polysomes; preparing cDNA probes from template RNA derived from the extracted polysomes; performing microarray-based comparison of the relative abundance of the different RNA species; analyzing the results; and identifying genes or clones encoding membranal or secreted proteins. Identification of cDNAs and genes encoding secreted or membranal coding mRNAs is of major importance in TCC. More specifically, novel genes which mark the early stages of TCC and code for secreted proteins are the ultimate markers for diagnosis and follow-up of TCC. By deriving probes from template RNA extracted from membrane-bound polysomes and free polysomes and performing microarray-based comparison of the relative abundance of different RNA species, such potentially secreted proteins can be identified. Analysis of the results of such comparison and identification of the clones encoding for membranal or secreted - 16- WO 01/22864 PCT/USOO/41005 proteins provides a valuable tool which can be used together with other gene discovery tools, and which in itself enables identification of likely targets for drug development. Since membranal and secreted proteins are both accessible and critical for transduction of numerous intra- and intercellular signals, they are generally viewed as preferred targets for pharmacological use and intervention. Therefore, the a priori classification of arrayed unknown gene sequences into those that potentially code for secreted and membranal proteins is of great value for the optimization of a high-throughput process of identifying potential drug targets. Furthermore, the identification of genes which express membranal or secreted proteins that are differentially expressed in different cellular situations is of the utmost importance in designing therapeutic or diagnostic tools for TCC. A method of identifying clones which encode membranal and secreted proteins was employed by preparing bladder cancer cell fractionations, preparing cDNA probes from template RNA derived from membrane-bound polysomes and free-polysomes, performing a microarray-based comparison of the relative abundance of different RNA species, analyzing the results and thereby identifying genes encoding for membranal and secreted proteins. Since membranal and secreted proteins are generally viewed as preferred targets for pharmacological intervention, the present invention thus provides a method of identifying likely targets for TCC diagnosis and therapy. HYBRIDIZATION AND PROBES: TCC and normal bladder hybridization: The probes were prepared from normal healthy bladder samples and from TCC tumors. Only intact RNA with a proper histological report indicating the -17- WO 01/22864 PCT/USOO/41005 existence of TCC was used. All normal and tumor material was collected from two separate clinical centers. Such approach minimizes the influence of local specific surgical bias or subjectivity of the pathological report. Forty-one hybridizations were performed. In each hybridization, two probes were used simultaneously, each labeled with either Cy3 or Cy5. These probes were as follows: Probe 1. Probe 1 was common to all hybridizations (common control probe). RNA from TCC samples was mixed with RNA from normal bladder samples. An equal amount of the RNA mixture was labeled with Cy3 and used in all hybridizations; and Probe 2. In each of the hybridizations, a different RNA sample from a single donor was used (test probe). A common control for all the hybridizations enables comparison of the results between the different hybridizations. If the common control (probe 1) hybridization results are similar in pattern in different hybridizations, comparison can be made between the results of probe 2 hybridizations and all hybridizations. Seventeen hybridizations included 16 RNA samples extracted from different control healthy bladder mucosa labeled with Cy5. Twenty-three hybridizations were performed with RNA samples derived from tumor tissues, either from non invasive Ta or from T1 stages of development. Two hybridizations were performed with RNA extracted from 2 invasive TCC samples. The hybridizations were carried out in three separate sets, but the same common control was used in all sets. Set 1 includes hybridizations 2-11 (TC2A TC11A), set 2 includes hybridizations 16-25 (TC16A-TC25A), and set 3 includes hybridizations 28-41 (TC28A-TC41A). By using three different sets of hybridizations, the possibility of technical effects related to specific hybridizations is reduced. See Tables below and related description. - 18- WO 01/22864 PCT/USOO/41005 Probe from annotation of potentially secreted proteins: TCC cell line -T24- (from ATCC) was used for cellular fractionation. Membrane-bound polysomes were separated from free polysomes using a sucrose step gradient. RNA coding for potentially secreted proteins was isolated from this microsomal-membranal fraction and separated from RNA coding for intracellular proteins. Hybridization was performed as described hereunder. The probes used were as follows: Probe 1. Free polysomal RNA fraction labeled with Cy3; and Probe 2. Membrane-bound RNA fraction with Cy5. TCC CHIP PREPARATION All hybridizations were performed on TCC designated microarray. The microarray was made up of cDNA clones derived from 3 different libraries: 1. SDGI library: (Described in US Patent Application USSN 09/538,709 of same applicant company, filed 30 March, 2000, and incorporated herein by reference in its entirety): A pool of non-invasive TCC, invasive TCC and normal bladder was used for library preparation. 4550 clones from the SDGI library were included in the TCC chip. 2. Antisense library: (Described in US Provisional Patent Application SN 60/157,843 of same applicant company, filed 6 October, 1999, and incorporated herein by reference in its entirety): The same cDNA pool used for the SDGI library was used for the preparation of a library enriched for antisense sequences. 450 clones from this library were included in the TCC chip. 3. SSH library: (Diatchenko et al., 1996). A subtraction library was made as follows. A normal bladder RNA pool was used for subtraction from non-invasive TCC RNA pool. The subtracted cDNA was used for - 19- WO 01/22864 PCTIUSOO/41005 the microarray printing. 5000 clones from the SSH library were used for printing. General methods in molecular biology: Standard molecular biology techniques known in the art and not specifically described were generally followed as in Sambrook et al., Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Laboratory Press, New York (1989), and in Ausubel et al., Current Protocols in Molecular Biology, John Wiley and Sons, Baltimore, Maryland (1989) and in Perbal, A Practical Guide to Molecular Cloning, John Wiley & Sons, New York (1988), and in Watson et al., Recombinant DNA, Scientific American Books, New York and in Birren et al (eds) Genome Analysis: A Laboratory Manual Series, Vols. 1-4 Cold Spring Harbor Laboratory Press, New York (1998) and methodology as set forth in United States patents 4,666,828; 4,683,202; 4,801,531; 5,192,659 and 5,272,057 and incorporated herein by reference. Polymerase chain reaction (PCR) was carried out generally as in PCR Protocols: A Guide To Methods And Applications, Academic Press, San Diego, CA (1990). In-situ (In-cell) PCR in combination with Flow Cytometry can be used for detection of cells containing specific DNA and mRNA sequences (Testoni et al, 1996, Blood 87:3822.) General methods in immunology: Standard methods in immunology known in the art and not specifically described are generally followed as in Stites et al.(eds), Basic and Clinical Immunology (8th Edition), Appleton & Lange, Norwalk, CT (1994) and Mishell and Shiigi (eds), Selected Methods in Cellular Immunology, W.H. Freeman and Co., New York (1980). - 20 - WO 01/22864 PCT/USOO/41005 Immunoassays In general, ELISAs where appropriate are one of the immunoassays employed to assess a specimen. ELISA assays are well known to those skilled in the art. Both polyclonal and monoclonal antibodies can be used in the assays. Where appropriate other immunoassays, such as radioimmunoassays (RIA) can be used as are known to those in the art. Available immunoassays are extensively described in the patent and scientific literature. See, for example, United States patents 3,791,932; 3,839,153; 3,850,752; 3,850,578; 3,853,987; 3,867,517; 3,879,262; 3,901,654; 3,935,074; 3,984,533; 3,996,345; 4,034,074; 4,098,876; 4,879,219; 5,011,771 and 5,281,521 as well as Sambrook et al, Molecular Cloning: A Laboratory Manual, Cold Springs Harbor, New York, 1989 Antibody Production Antibodies can be either monoclonal, polyclonal or recombinant. Conveniently, the antibodies can be prepared against the immunogen or portion thereof for example a synthetic peptide based on the sequence, or prepared recombinantly by cloning techniques or the natural gene product and/or portions thereof can be isolated and used as the immunogen. Immunogens can be used to produce antibodies by standard antibody production technology well known to those skilled in the art as described generally in Harlow and Lane, Antibodies: A Laboratory Manual, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, 1988 and Borrebaeck, Antibody Engineering - A Practical Guide, W.H. Freeman and Co., 1992. Antibody fragments can also be prepared from the antibodies and include Fab, F(ab') 2 , and Fv by methods known to those skilled in the art. For producing polyclonal antibodies a host, such as a rabbit or goat, is immunized with the immunogen or immunogen fragment, generally with an adjuvant and, if necessary, coupled to. a carrier; antibodies to the immunogen are collected from the sera. Further, the polyclonal antibody can be absorbed such -21 - WO 01/22864 PCTIUSOO/41005 that it is monospecific. That is, the sera can be absorbed against related immunogens so that no cross-reactive antibodies remain in the sera rendering it monospecific. For producing monoclonal antibodies the technique involves hyperimmunization of an appropriate donor with the immunogen, generally a mouse, and isolation of splenic antibody producing cells. These cells are fused to a cell having immortality, such as a myeloma cell, to provide a fused cell hybrid which has immortality and secretes the required antibody. The cells are then cultured, in bulk, and the monoclonal antibodies harvested from the culture media for use. For producing recombinant antibody (see generally Huston et al, 1991; Johnson and Bird, 1991; Mernaugh and Mernaugh, 1995), messenger RNAs from antibody producing B-lymphocytes of animals, or hybridoma are reverse transcribed to obtain complimentary DNAs (CDNAs). Antibody cDNA, which can be full or partial length, is amplified and cloned into a phage or a plasmid. The cDNA can be a partial length of heavy and light chain cDNA, separated or connected by a linker. The antibody, or antibody fragment, is expressed using a suitable expression system to obtain recombinant antibody. Antibody cDNA can also be obtained by screening pertinent expression libraries. The antibody can be bound to a solid support substrate or conjugated with a detectable moiety or be both bound and conjugated as is well known in the art. (For a general discussion of conjugation of fluorescent or enzymatic moieties see Johnstone & Thorpe, Immunochemistry in Practice, Blackwell Scientific Publications, Oxford, 1982.) The binding of antibodies to a solid support substrate is also well known in the art. (see for a general discussion Harlow & Lane Antibodies: A Laboratory Manual, Cold Spring Harbor Laboratory Publications, New York, 1988 and Borrebaeck, Antibody Engineering - A Practical Guide, W.H. Freeman and Co., 1992) The detectable moieties contemplated with the present - 22 - WO 01/22864 PCT/USOO/41005 invention can include, but are not limited to, fluorescent, metallic, enzymatic and radioactive markers such as biotin, gold, ferritin, alkaline phosphatase, p galactosidase, peroxidase, urease, fluorescein, rhodamine, tritium, 14C and iodination. Recombinant Protein Purification Marshak et al, "Strategies for Protein Purification and Characterization. A laboratory course manual." CSHL Press, 1996. Transgenic and Knockout Methods The present invention provides for transgenic gene and polymorphic gene animal and cellular (cell lines) models as well as for knockout models. These models are constructed using standard methods known in the art and as set forth in United States Patents 5,487,992, 5,464,764, 5,387,742, 5,360,735, 5,347,075, 5,298,422, 5,288,846, 5,221,778, 5,175,385, 5,175,384,5,175,383, 4,736,866 as well as Burke and Olson (1991), Capecchi (1989), Davies et al. (1992), Dickinson et al. (1993), Duff and Lincoln (1995), Huxley et al. (1991), Jakobovits et al. (1993), Lamb et al. (1993), Pearson and Choi (1993), Rothstein (1991), Schedl et al. (1993), Strauss et al. (1993). Further, patent applications WO 94/23049, WO 93/14200, WO 94/06908, WO 94/28123 also provide information. Further, one parent strain instead of carrying a direct human transgene can have the homologous endogenous gene modified by gene targeting such that it approximates the transgene. That is, the endogenous gene has been "humanized" and/or mutated (Reaume et al, 1996). It should be noted that if the animal and human sequence are essentially homologous a "humanized" gene is not required. The transgenic parent can also carry an overexpressed sequence, either the -23 - WO 01/22864 PCT/USOO/41005 nonmutant or a mutant sequence and humanized or not as required. The term transgene is therefore used to refer to all these possibilities. Additionally, cells can be isolated from the offspring which carry a transgene from each transgenic parent and that are used to establish primary cell cultures or cell lines as is known in the art. Where appropriate, a parent strain is homozygous for the transgene. Additionally, where appropriate, the endogenous nontransgene in the genome that is homologous to the transgene is nonexpressive. By nonexpressive is meant that the endogenous gene is not expressed and that this nonexpression is heritable in the offspring. For example, the endogenous homologous gene could be "knocked out" by methods known in the art. Alternatively, the parental strain that receives one of the transgenes could carry a mutation at the endogenous homologous gene rendering it nonexpressed. For gene therapy: By gene therapy as used herein refers to the transfer of genetic material (e.g DNA or RNA) of interest into a host to treat or prevent a genetic or acquired disease or condition phenotype. The genetic material of interest encodes a product (e.g. a protein, polypeptide, peptide, functional RNA, antisense) whose production in vivo is desired. For example, the genetic material of interest can encode a hormone, receptor, enzyme, polypeptide or peptide of therapeutic value. Alternatively, the genetic material of interest encodes a suicide gene. For a review see, in general, the text "Gene Therapy" (Advances in Pharmacology 40, Academic Press, 1997). Two basic approaches to gene therapy have evolved: (1) ex vivo and (2) in vivo gene therapy. In ex vivo gene therapy cells are removed from a patient, and while being cultured are treated in vitro. Generally, a functional replacement gene - 24 - WO 01/22864 PCT/USOO/41005 is introduced into the cell via an appropriate gene delivery vehicle/method (transfection, transduction, homologous recombination, etc.) and an expression system as needed and then the modified cells are expanded in culture and returned to the host/patient. These genetically reimplanted cells have been shown to express the transfected genetic material in situ. In in vivo gene therapy, target cells are not removed from the subject rather the genetic material to be transferred is introduced into the cells of the recipient organism in situ, that is within the recipient. In an alternative embodiment, if the host gene is defective, the gene is repaired in situ [Culver, 1998]. These genetically altered cells have been shown to express the transfected genetic material in situ. The gene expression vehicle is capable of delivery/ transfer of heterologous nucleic acid into a host cell. The expression vehicle can include elements to control targeting, expression and transcription of the nucleic acid in a cell selective manner as is known in the art. It should be noted that often the 5'UTR and/or 3'UTR of the gene can be replaced by the 5'UTR and/or 3'UTR of the expression vehicle. Therefore as used herein the expression vehicle can, as needed, not include the 5'UTR and/or 3'UTR of the actual gene to be transferred and only include the specific amino acid coding region. The expression vehicle can include a promotor for controlling transcription of the heterologous material and can be either a constitutive or inducible promotor to allow selective transcription. Enhancers that can be required to obtain necessary transcription levels can optionally be included. Enhancers are generally any non translated DNA sequence which works contiguously with the coding sequence (in cis) to change the basal transcription level dictated by the promoter. The expression vehicle can also include a selection gene as described herein below. - 25 - WO 01/22864 PCTIUSOO/41005 Vectors can be introduced into cells or tissues by any one of a variety of known methods within the art. Such methods can be found generally described in Sambrook et al., Molecular Cloning: A Laboratory Manual, Cold Springs Harbor Laboratory, New York (1989, 1992), in Ausubel et al., Current Protocols in Molecular Biology, John Wiley and Sons, Baltimore, Maryland (1989), Chang et al., Somatic Gene Therapy, CRC Press, Ann Arbor, MI (1995), Vega et al., Gene Targeting, CRC Press, Ann Arbor, MI (1995), Vectors: A Survey of Molecular Cloning Vectors and Their Uses, Butterworths, Boston MA (1988) and Gilboa et al (1986) and include, for example, stable or transient transfection, lipofection, electroporation and infection with recombinant viral vectors. In addition, see United States patent 4,866,042 for vectors involving the central nervous system and also United States patents 5,464,764 and 5,487,992 for positive-negative selection methods. Introduction of nucleic acids by infection offers several advantages over the other listed methods. Higher efficiency can be obtained due to their infectious nature. Moreover, viruses are very specialized and typically infect and propagate in specific cell types. Thus, their natural specificity can be used to target the vectors to specific cell types in vivo or within a tissue or mixed culture of cells. Viral vectors can also be modified with specific receptors or ligands to alter target specificity through receptor mediated events. A specific example of DNA viral vector for introducing and expressing recombinant sequences is the adenovirus derived vector Adenop53TK. This vector expresses a herpes virus thymidine kinase (TK) gene for either positive or negative selection and an expression cassette for desired recombinant sequences. This vector can be used to infect cells that have an adenovirus receptor which includes most cancers of epithelial origin as well as others. This vector as well as others that exhibit similar desired functions can be used to treat a mixed population of cells and can include, for example, an in vitro or ex vivo culture of cells, a tissue or a human subject. - 26 - WO 01/22864 PCTIUSOO/41005 Additional features can be added to the vector to ensure its safety and/or enhance its therapeutic efficacy. Such features include, for example, markers that can be used to negatively select against cells infected with the recombinant virus. An example of such a negative selection marker is the TK gene described above that confers sensitivity to the antibiotic gancyclovir. Negative selection is therefore a means by which infection can be controlled because it provides inducible suicide through the addition of antibiotic. Such protection ensures that if, for example, mutations arise that produce altered forms of the viral vector or recombinant sequence, cellular transformation can not occur. Features that limit expression to particular cell types can also be included. Such features include, for example, promoter and regulatory elements that are specific for the desired cell type. In addition, recombinant viral vectors are useful for in vivo expression of a desired nucleic acid because they offer advantages such as lateral infection and targeting specificity. Lateral infection is inherent in the life cycle of, for example, retrovirus and is the process by which a single infected cell produces many progeny virions that bud off and infect neighboring cells. The result is that a large area becomes rapidly infected, most of which was not initially infected by the original viral particles. This is in contrast to vertical-type of infection in which the infectious agent spreads only through daughter progeny. Viral vectors can also be produced that are unable to spread laterally. This characteristic can be useful if the desired purpose is to introduce a specified gene into only a localized number of targeted cells. As described above, viruses are very specialized infectious agents that have evolved, in many cases, to elude host defense mechanisms. Typically, viruses infect and propagate in specific cell types. The targeting specificity of viral vectors utilizes its natural specificity to specifically target predetermined cell types and thereby introduce a recombinant gene into the infected cell. The vector to be used - 27 - WO 01/22864 PCT/USOO/41005 in the methods of the invention depends on desired cell type to be targeted and is known to those skilled in the art. For example, if breast cancer is to be treated then a vector specific for such epithelial cells are used. Likewise, if diseases or pathological conditions of the hematopoietic system are to be treated, then a viral vector that is specific for blood cells and their precursors, preferably for the specific type of hematopoietic cell, is used. Retroviral vectors can be constructed to function either as infectious particles or to undergo only a single initial round of infection. In the former case, the genome of the virus is modified so that it maintains all the necessary genes, regulatory sequences and packaging signals to synthesize new viral proteins and RNA. Once these molecules are synthesized, the host cell packages the RNA into new viral particles which are capable of undergoing further rounds of infection. The vector's genome is also engineered to encode and express the desired recombinant gene. In the case of non-infectious viral vectors, the vector genome is usually mutated to destroy the viral packaging signal that is required to encapsulate the RNA into viral particles. Without such a signal, any particles that are formed do not contain a genome and therefore cannot proceed through subsequent rounds of infection. The specific type of vector depends upon the intended application. The actual vectors are also known and readily available within the art or can be constructed by one skilled in the art using well-known methodology. The recombinant vector can be administered in several ways. If viral vectors are used, for example, the procedure can take advantage of their target specificity and consequently, do not have to be administered locally at the diseased site. However, local administration can provide a quicker and more effective treatment, administration can also be performed by, for example, intravenous or subcutaneous injection into the subject. Injection of the viral vectors into a spinal fluid can also be used as a mode of administration, especially in the case of neuro-degenerative diseases. Following injection, the viral vectors circulate until they recognize host cells with the appropriate target specificity for infection. - 28 - WO 01/22864 PCTIUSOO/41005 An alternate mode of administration can be by direct inoculation locally at the site of the disease or pathological condition or by inoculation into the vascular system supplying the site with nutrients or into the spinal fluid. Local administration is advantageous because there is no dilution effect and, therefore, a smaller dose is required to achieve expression in a majority of the targeted cells. Additionally, local inoculation can alleviate the targeting requirement required with other forms of administration since a vector can be used that infects all cells in the inoculated area. If expression is desired in only a specific subset of cells within the inoculated area, then promoter and regulatory elements that are specific for the desired subset can be used to accomplish this goal. Such non-targeting vectors can be, for example, viral vectors, viral genome, plasmids, phagemids and the like. Transfection vehicles such as liposomes can also be used to introduce the non viral vectors described above into recipient cells within the inoculated area. Such transfection vehicles are known by one skilled within the art. Delivery of gene products/therapeutics (compound): The compound of the present invention is administered and dosed in accordance with good medical practice, taking into account the clinical condition of the individual patient, the site and method of administration, scheduling of administration, patient age, sex, body weight and other factors known to medical practitioners. The pharmaceutically "effective amount" for purposes herein is thus determined by such considerations as are known in the art. The amount must be effective to achieve improvement including but not limited to improved survival rate or more rapid recovery, or improvement or elimination of symptoms and other indicators as are selected as appropriate measures by those skilled in the art. In the method of the present invention, the compound of the present invention can be administered in various ways. It should be noted that it can be -29- WO 01/22864 PCT/USOO/41005 administered as the compound or as pharmaceutically acceptable salt and can be administered alone or as an active ingredient in combination with pharmaceutically acceptable carriers, diluents, adjuvants and vehicles. The compounds can be administered orally, subcutaneously or parenterally including intravenous, intraarterial, intramuscular, intraperitoneally, and intranasal administration as well as intrathecal and infusion techniques. Implants of the compounds are also useful. The patient being treated is a warm-blooded animal and, in particular, mammals including man. The pharmaceutically acceptable carriers, diluents, adjuvants and vehicles as well as implant carriers generally refer to inert, non-toxic solid or liquid fillers, diluents or encapsulating material not reacting with the active ingredients of the invention. It is noted that humans are treated generally longer than the mice or other experimental animals exemplified herein which treatment has a length proportional to the length of the disease process and drug effectiveness. The doses can be single doses or multiple doses over a period of several days, but single doses are preferred. The doses can be single doses or multiple doses over a period of several days. The treatment generally has a length proportional to the length of the disease process and drug effectiveness and the patient species being treated. When administering the compound of the present invention parenterally, it is generally formulated in a unit dosage injectable form (solution, suspension, emulsion). The pharmaceutical formulations suitable for injection include sterile aqueous solutions or dispersions and sterile powders for reconstitution into sterile injectable solutions or dispersions. The carrier can be a solvent or dispersing medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, liquid polyethylene glycol, and the like), suitable mixtures thereof, and vegetable oils. - 30 - WO 01/22864 PCT/USOO/41005 Proper fluidity can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants. Nonaqueous vehicles such a cottonseed oil, sesame oil, olive oil, soybean oil, corn oil, sunflower oil, or peanut oil and esters, such as isopropyl myristate, can also be used as solvent systems for compound compositions. Additionally, various additives which enhance the stability, sterility, and isotonicity of the compositions, including antimicrobial preservatives, antioxidants, chelating agents, and buffers, can be added. Prevention of the action of microorganisms can be ensured by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid, and the like. In many cases, it is desirable to include isotonic agents, for example, sugars, sodium chloride, and the like. Prolonged absorption of the injectable pharmaceutical form can be brought about by the use of agents delaying absorption, for example, aluminum monostearate and gelatin. According to the present invention, however, any vehicle, diluent, or additive used have to be compatible with the compounds. Sterile injectable solutions can be prepared by incorporating the compounds utilized in practicing the present invention in the required amount of the appropriate solvent with various of the other ingredients, as desired. A pharmacological formulation of the present invention can be administered to the patient in an injectable formulation containing any compatible carrier, such as various vehicle, adjuvants, additives, and diluents; or the compounds utilized in the present invention can be administered parenterally to the patient in the form of slow-release subcutaneous implants or targeted delivery systems such as monoclonal antibodies, vectored delivery, iontophoretic, polymer matrices, liposomes, and microspheres. Examples of delivery systems useful in the present invention include: 5,225,182; 5,169,383; 5,167,616; 4,959,217; 4,925,678; 4,487,603; 4,486,194; 4,447,233; 4,447,224; 4,439,196; and 4,475,196. Many other such implants, delivery systems, and modules are well known to those skilled in the art. - 31 - WO 01/22864 PCT/USOO/41005 A pharmacological formulation of the compound utilized in the present invention can be administered orally to the patient. Conventional methods such as administering the compounds in tablets, suspensions, solutions, emulsions, capsules, powders, syrups and the like are usable. Known techniques which deliver it orally or intravenously and retain the biological activity are preferred. In one embodiment, the compound of the present invention can be administered initially by intravenous injection to bring blood levels to a suitable level. The patient's levels are then maintained by an oral dosage form, although other forms of administration, dependent upon the patient's condition and as indicated above, can be used. The quantity to be administered vary for the patient being treated and vary from about 100 ng/kg of body weight to 100 mg/kg of body weight per day and preferably are from 10 ptg/kg to 10 mg/kg per day. Differential Analysis Appropriate cells, cell lines or tissues are grown under normal conditions (non-stress) or under stress conditions generally for four to sixteen hours. Alternatively two cell types are compared. The cells are harvested and RNA is prepared from the cytoplasmic extracts and from the nuclear fractions. Following the extraction of RNA, fluorescent cDNA probes are prepared. Each condition is labeled with a different fluorescent dye. For example a probe can be composed of a mixture of Cy3 -dCTP cDNA prepared from RNA extracted from stressed cells and with Cy5-dCTP cDNA prepared from RNA extracted from nonstressed cells. The probes are used for hybridization to micro-array containing individually spotted cDNA clones derived from cells that were exposed to stress. Differential expression in measured by the amount of fluorescent cDNA that hybridizes to each of the clones on the array. Genes that are up regulated under stress have more fluorescence of Cy3 than Cy5. The results generally show genes that are transcriptionally induced mRNA species that respond very fast to stress. - 32 - WO 01/22864 PCT/USOO/41005 Differential display: Reverse transcription: 2ptg of RNA are annealed with lpmol of oligo dT primer (dT)j, in a volume of 6 .5pl by heating to 700 C for five minutes and cooling on ice. 2pl reaction buffer (x5), 1pl of 10mM dNTP mix, and 0.5pt1 of SuperScript il reverse transcriptase (GibcoBRL) is added. The reaction is carried for one hour at 420C. The reaction is stopped by adding 70pl TE (10mM Tris pH=8; 0.1mM EDTA). Oligonucleotides were used for Differential display. The oligonucleotides are generally those described in the Delta RNA Fingerprinting kit (Clonetech Labs. Inc.). Amplification reactions: Each reaction is done in 20pl and contains 50ptM dNTP mix, 1 1 pM from each primer, 1x polymerase buffer, 1 unit expand Polymerase (Beohringer Mannheim), 2ptCi [c-"P]dATP and 1ptl cDNA template. Cycling conditions are generally: three minutes at 950C, then three cycles of two minutes at 940C, five minutes at 400C, five minutes at 680C. This is followed by 27 cycles of one minute at 940C, two minutes at 600C, two minutes at 680C. Reactions were terminated by a seven minute incubation at 680C and addition of 20ptl sequencing stop solution (95% formamide, 10mM NaOH, 0.025% bromophenol blue, 0.025% xylene cyanol). Gel analysis: Generally 3-4pl are loaded onto a 5% sequencing polyacrylamide gel and samples are electrophoresed at 2000 volts/40 milliamperes until the slow dye (xylene cyanol) is about 2 cm from the bottom. The gel is transferred to a filter paper, dryed under vacuum and exposed to x-ray film. Recovery of differential bands: Bands showing any a differential between the various pools are excised out of the dried gel and placed in a microcentrifuge - 33 - WO 01/22864 PCT/USO0/41005 tube. 50gl of sterile H 2 0 are added and the tubes heated to 1000C for five minutes. 1p is added to a 49I PCR reaction using the same primers used for the differential display and the samples are amplified for 30 cycles of: one minute at 940C, one minute at 600C and one minute at 680C. 10d is analyzed on agarose gel to visualize and confirm successful amplification. Representational difference analysis Reverse transcription: as above but with 2pg polyA+ selected mRNA. Preparation of double stranded cDNA: cDNA from the previous step is treated with alkali to remove the mRNA, precipitated and dissolved in 20pl H 2 0. 5p I buffer, 2ptl 10mM dATP, H 2 0 to 48pi and 2pd terminal deoxynucleotide transferase (TdT) are added. The reaction is incubated two to four hours at 370C. 5ptl oligo dT (1pg/tl) was added and incubated at 600C for five minutes. 5pl 200 mM DTT, 10 p 10x section buffer (100mM Mg C12, 900 mM Hepes, pH 6.6) 16 pl dNTPs (1 mM), and 16 U of Klenow are added and the mixture incubated overnight at room temperature to generate ds cDNA. 100pl TE is added and extracted with phenol/chloroform. The DNA is precipitated and dissolved in 50pI
H
2 0. Generation of representations: cDNA with Dpnll is digested by adding 3pl Dpnll reaction buffer 20 V and Dpnll to 2 5pl cDNA and incubated five hours at 370 C. 50ptl TE is added and extracted with phenol/chloroform. cDNA is precipitated and dissolved to a concentration of lOng/pl. Driver: 1.2pg Dpnll digested cDNA. 4[I from each oligo and 5pl ligation buffer x1O and annealed at 600C for ten minutes. 2pl ligase is added and incubated overnight at 160C. The ligation mixture is diluted by adding 140pl TE. Amplification is carried out in a volume of 200pI using appropriate primer and 2pl ligation product and repeated in twenty tubes for each sample. Before adding Taq - 34 - WO 01/22864 PCT/USOO/41005 DNA polymerase, the tubes are heated to 720C for three minutes. PCR conditions are as follows: five minutes at 720C, twenty cycles of one minute at 950C and three minutes at 72 0 C, followed by ten minutes at 72*C. Every four reactions were combined, extracted with phenol/chloroform and precipitated. Amplified DNA is dissolved to a concentration of 0.5ptg/pl and all samples are pooled. Subtraction: Tester DNA (20pg) is digested with Dpnll as above and separated on a 1.2% agarous gel. The DNA is extracted from the gel and 2 ig ligated to the appropriate oligos. The ligated Tester DNA is then diluted to 1 Ong/pl with TE. Driver DNA is digested with Dpnil and repurified to a final concentration of 0.5pg/tl. Mix 40ptg of Driver DNA with 0.4ptg of Tester DNA. Extraction is carried out with phenol/chloroform and precipitated using two washes with 70% ethanol, resuspended DNA in 4d of 30mM EPPS pH=8.0, 3mM EDTA and overlaid with 35 Il mineral oil. Denature at 980C for five minutes, cool to 670C and 1pdl of 5M NaCl added to the DNA. Incubate at 670C for twenty hours. Dilute DNA by adding 4 00p TE. Amplification: Amplification of subtracted DNA in a final volume of 200ptl as follows: Buffer, nucleotides and 20tl of the diluted DNA are added, heated to 720 C, and Taq DNA polymerase added. Incubate at 720C for five minutes and add apprpriate oligo. Ten cycles of one minute at 950C, three minutes at 700C are performed. Incubate ten minutes at 720C. The amplification is repeated in four separate tubes. The amplified DNA is extracted with phenol/chloroform, precipitated and all four tubes combined in 40pl 0.2xTE, and digested with Mung Bean Nuclease as follows: To 2 0p1 DNA 4[l buffer, 14pl H 2 0 and 2 1 pl Mung Bean Nuclease (10 units/pl) added. Incubate at 300C for thirty-five minutes + First Differential Product (DPI). - 35 - WO 01/22864 PCT/USOO/41005 Repeat subtraction hybridization and PCR amplification at driver: differential ratio of 1:400 (DPII) and 1:40,000 (DPIII) using appropriate oligonucleotides. Differential products are then cloned into a Bluescript vector at the BAM Hl site for analysis of the individual clones. DIFFERENTIAL EXPRESSION USING GENE EXPRESSION MICRO-ARRAY Messenger RNA isolated as described herein above is labeled with fluorescent dNTP's using a reverse transcription reaction to generate a labeled cDNA probe. mRNA is extracted from cells cultured in non-stress conditions and labeled with Cy3-dCTP (Amersham) and mRNA extracted from cells cultured under stress conditions is labeled with Cy5-dCTP (Amersham). The two labeled cDNA probes are then mixed and hybridized onto a microarray (Schena et al, 1996) composed of for example 2000 cDNA clones derived from a cDNA library prepared from appropriate cells cultured under the stress conditions. Following hybridization the microarray is scanned using a laser scanner and amount of fluorescence of each of the fluorescence dyes is measured for each cDNA clone on the micro-array giving an indication of the level of mRNA in each of the original mRNA populations being tested. Comparison of the fluorescence on each cDNA clone on the micro-array between the two different fluorescent dyes is a measure for the differential expression of the indicated genes between the two experimental conditions. IN SITU ANALYSIS In situ analysis as needed is performed for the candidate genes identified by the methods as described above. - 36 - WO 01/22864 PCT/USOO/41005 Cell culture protocols. C6, Hela and Jurkat cell lines (ATCC) were grown either in DMEM (C6 and HeLa) or in RPMI (Jurkat) supplemented with 10% FCS. C6 cells were grown either under normal oxygen conditions or under hypoxia for 4 or 16 hours (0.5%02 and 5%CO2). Jurkat cells were grown either at 3700 or exposed to 430C for one or four hours. HeLa cells were kept under normal tissue culture conditions. One to three hours prior to harvesting (for non-treated cells) or prior to treatment, culture medium was replaced with a fresh one. Cells were then washed with ice cold PBS-/-, harvested on ice and either directly processed to extraction of total RNA or collected by centrifugation (400g, 5 minutes). The cell pellet was immediately frozen in liquid nitrogen and kept at -70' until used for fractionation to isolate nuclei, polysomes or microsomes. Cell fractionation protocols. Preparation of nuclear and polysomal fractions. Nuclear and polysomal subcellular fractions were obtained in a single fractionation procedure as previously described 23. Briefly, frozen cell pellets were resuspended in ice-cold polysomal buffer: 25mM Tris HCI, pH 7.4-7.5; 10mM MgCl 2 ; 25mM NaCl; 0.05% Triton X-100; 0.14M sucrose, 100 tg/ml heparin, 1mM DTT and 133 pg/ml RNasin (Promega). 0.1% Triton X-100 and 0.1% deoxycholate were added and following 3' incubation on ice, the nuclei were pelleted (10,000g, 4 0 C, 3') and NUC RNA was extracted . The supernatant was further processed for isolation of the polysomal fraction. After addition of 1mg/ml of heparin it was layered on top of a sucrose gradient (0.5M 1.5M) and centrifuged (Beckman, SW41, 36K, 40C, 110'). Upon centrifugation, fractions were collected into tubes containing 0.5% SDS. The polysomal fraction was further used for extraction of RNA Preparation of membrane bound and free polysomes. For isolation of microsomes, Hela cells were grown to 50%-70% confluence and Jurkat cells were grown to cell densities 0.5 - 1X10 6 cells/ml. Cytoplasmic lysate was prepared essentially as - 37- WO 01/22864 PCT/USOO/41005 described 2 4 . Briefly, Hela or Jurkat cell pellets were thawed on ice and lysed with 0.25M sucrose lysis buffer (250mM sucrose, 50mM TEA (triethanolamine), 50mM KOAc pH 7.5, 6mM Mg(OAc)2, 1mM EDTA, 1mM DTT, 0.5mM PMSF, 200 units/ml RNasine (Promega), 0.1mg/ml heparin) - 1ml per 108 cells. Cells were homogenized on ice using a Teflon homogenizer (Heidolph, 1000 rpm). Following centrifugation (600g, 10 minutes, 40C), the supernatant was collected and kept on ice. The nuclei pellet was washed with 1ml of lysis buffer and re-centrifuged (600g, 10 minutes, 40C) to release the contaminating microsomes. The resultant supernatant was combined with the previous one. Total supernatant material was further centrifuged to precipitate mitochondria (10,000g, 10 minutes, 4*C) and heparin (1mg/ml) was added to the collected supernatant. The latter (5 ml) was layered over the two-step sucrose gradient (2.5 ml of 2.05M sucrose in lysis buffer/2.5 ml of 1.5M sucrose in lysis buffer). After centrifugation (100,000g, 3 hours, 40C), the upper phase (5 ml) containing mRNPs was discarded. Middle phase (2.5 ml) containing membrane bound polysomes (MBP) and pellet containing free polysomes (FP) were further used for RNA extraction. RNA extraction protocols Extraction of total cellular (TC) and nuclear RNA (NUC) was performed with Tri Reagent (Sigma) according to commercial protocols. Extraction of RNA from membrane bound (MBP) and free polysomes (FP). Pellet of free polysomes was dissolved in 1% SDS/0.1M NaCl. Membrane polysome fraction was diluted with TE 1:1 and SDS and proteinase K (Sigma) were added to final concentrations 1% and 0.1mg/ml, respectively. Following incubation at 370C for 30 minutes, RNA was purified from both fractions with phenol:chloroform (1:1) and precipitated with 0.3M NaOAc, 20pg/ml glycogen (Roche) and 50% isopropanol. Following centrifugation (10,000g, 20 minutes, 40C), RNA pellets were washed with 70% ethanol, dried and dissolved in H 2 0. Extraction of polysomal RNA (POL) was done as described for the MBP fraction. - 38 - WO 01/22864 PCT/USOO/41005 cDNA microarrays. C6-specific microarray. A cDNA microarray containing 1847 cDNA fragments was constructed from clones of subtracted cDNA libraries derived from C6 glioma cells enriched for hypoxia-responsive mRNAs. Specifically, mRNA prepared from C6 cells cultured under hypoxic conditions for 4 and 16 hours, respectively, was subjected to bidirectional subtraction (PCR-Select cDNA subtraction kit, Clontech) followed by cloning into pBluescript. Three sublibraries were prepared. Sublibrary A was enriched for cDNA species up-regulated after four hours of hypoxia: sublibrary B, for cDNA species up-regulated after 16 hours of hypoxia; and sublibrary C, for cDNA species down-regulated after 16 hours of hypoxia. For microarray printing, 523 cDNA clones were selected from sublibrary A, 884 from sublibrary B, and 373 from sublibrary C. The array also contained a set of control genes whose response to hypoxia in C6 glioma (and other cells) is well documented including vascular endothelial growth factor (VEGF) 8 ,9, 2 5 , glucose transporter 1(Glut1) 9
,
26 and lactate dehydrogenase (LDH) 2 7 . F2054-specific microarray. This microarray contained 1418 human cDNA clones collected from a library derived from the human F2054 fibroblast cell line. The library was prepared in pBluescript, the average insert size being about 500 bp. Human UniGEMI microarray (Incyte). This commercially available microarray that contains 9,700 human cDNA clones, 60% of them coding for known genes, was used in experiments with MBP and FP derived probes. Since proper interpretation of experimental data requires precise clone identity, only clones that were verified by sequencing in Incyte (2,800 out of 9,700) were subjected to the analysis. - 39 - WO 01/22864 PCT/USOO/41005 Probe labeling and hybridization to cDNA microarrays cDNA probes were synthesized from 50tg of RNA using reverse transcriptase (Superscript, Gibco-BRL) and 18-mer oligo-dT primer. The hybridization probe was composed of two cDNA populations, derived from two different RNA sources: one labelled with Cy3-dCTP and the other with Cy5-dCTP (Amersham), as previously described 2 8 . The following types of probes were used for hybridizations to different DNA microarrays: C6-specific microarray. This microarray was utilized for hybridizations of C6-derived probes: (1) total RNA normoxia (Cy3)/total RNA four hours hypoxia (Cy 5); (2) total RNA normoxia (Cy 3)/ total RNA 16 hours hypoxia (Cy 5); (3) nuclear RNA normoxia (Cy3)/nuclear RNA 4 hours hypoxia (Cy5); (4) nuclear RNA normoxia (Cy3)/nuclear RNA 16 hrs hypoxia (Cy5). F2054-specific microarray. This microarray was utilized for hybridization of probes derived from heat shock experiments with Jurkat cells: (1)total RNA 37 0 C(Cy3)/total RNA 1 hour 43 0 C(Cy 5); (2) total RNA 370C (Cy 3)/ total RNA four hours 430C (Cy 5); (3) nuclear RNA 37*C (Cy3)/nuclear RNA 1hr 43*C (Cy5); (4) polysomal RNA 370C (Cy3)/polysomal RNA 1 hour 430C (Cy5); (5) polysomal RNA 370C (Cy3)/polysomal RNA four hours 430C (Cy5) Unigeml(Incyte). This microarray was utilized for hybridization of probes derived from different polysomal fractions: (1) MBP RNA from Hela (Cy3)/ FP RNA from Hela (Cy5); (2) MBP RNA from Jurkat (Cy3)/ FP RNA from Jurkat (Cy5). Hybridization data processing Hybridizations, image processing and signal calculation were performed using commercial tools (Incyte, GEMTools). Overall signals were balanced by this software and differential expression values were calculated as "Cy3 signal" (P1)/"balanced Cy5 signal" (P2) (P1>P2) and "balanced Cy5 signal" (P2)/"Cy3 -40 - WO 01/22864 PCT/USOO/41005 signal" (P1) (P2>P1). For this reason, the numerical values shown in the tables may vary from the observed images. For all differentially expressed clones the image data was visually inspected. Since there was observed a certain inconsistency of hybridization results obtained with low expressed cDNA clones, in all experiments only clones that displayed an hybridization signal not lower than 500 units, at least in one of the fluorescence channels, were subjected to further analysis. Analysis of discrimination of MBP and FP cDNA populations Out of the annotated expressed clones, a random sample of 118 clones was selected and the subcellular localization of their encoded proteins was assigned. According to the literature and the SWISS-PROT database 2 9 , 29 of them code for either membranal/secreted or ribosomal proteins (MBP-resident), 63 code for cytosolic or nuclear proteins (FP-resident), and the remaining 26 genes could not be clearly assigned. Frequency histograms were built for ln(Plsignal/P2 Balanced Signal) since these values are anticipated to be approximated well by a normal distribution (while differential expression values are not). The parameters of normal distributions were then estimated for each histogram according to standard procedures, using the SPSS package, which was also used for the standard statistical tests for inequality of means of the approximated normal distributions (t test). Determination of conditional probabilities for cDNA clones to correspond to the membranal/secreted or to the cytosolic/nuclear group of proteins In order to calculate the conditional probabilities, we first estimated the a priori probability was first estimated for a selected clone to be MBP-resident (01 in equations (1,2)). This estimation is required since the relative portions of the two clone populations both on the array and in the expressed RNA population are probably not equal (more cytosolic/nuclear clones are expected), thereby biasing in advance their probability of detection. This a priori probability for a protein to be MBP-resident (a1=0.34, equation (1)) was calculated based on the assignment of -41- WO 01/22864 PCT/USOO/41005 proteins with prominent differential values >0.69 (right tail, corresponding to >+2 differential expression values) and <-0.69 (left tail, corresponding to <-2 differential expression values) of the distribution, as given in Table 2, so that a combined distribution would have these pre-determined right and left tail frequencies. The parameter al was found as a solution of the equation: (1) a -?(m-l( )) a ? -( w -ml FLI + FRI= SDI SD1 w--ml- w-m2 1wm -w-m2 al?(-Q _ )) + (1- al) ?(1 - D( -m2)) al?1(D 1 )+(1-al)?( w m SDI SD2 SDI SD2 where w = 0.69, ml,m2 - means of the two distributions, SD1,SD2 - their standard deviations, (D is standard normal distribution function (i.e. 1 Y 2 (y) Z= exp(--)dx), FL1 (FR1) - the frequency of first kind (MBP- resident) 2 _. 2 proteins to have prominent differential values less than -w (more than w), FL2 (FR2) - the frequency of second kind (FP-resident) proteins to have prominent differential values less than -w (more than w). The conditional probabilities for proteins of the first kind (encoded by the MBP resident mRNAs) and second kind (encoded by the FP-resident mRNAs) (p1 and p2, respectively) under assumption on normality of distributions were calculated according to the following equation: (2) al ?exp(- (w _m)2 p1(w)= alp( 2(SD1)2 al ?exp(- (w - m) )+(1 - al)?exp(- (w -m2) 2(SD1) 2 2(SD2) 2 where w = ln(Plsignal/P2BalancedSignal) and al is the a priori probability for a protein to be of the first kind. Bioinformatics sequence analysis of selected EST clones -42 - WO 01/22864 PCT/USOO/41005 The EST sequences were extended using QBI's propriety clustering software (manuscript in preparation). Putative coding regions for the obtained contig sequences were defined by two criteria: (1) open reading frame (ORF) longer than 50 amino acids, flanked by untranslated region(s) containing multiple stop codons in all three frames; and (2) when a potential 5' UTR was observed, the presence of an initiation methionine was required; (it was sometimes possible to define it relative to the poly-A tail). The contigs were characterized in two steps. First, homology searches (BLAST) were performed to identify homologous sequences in the nucleotide and protein non-redundant (nr) databases of Genbank. Then the contig sequences were characterized by prediction of motifs and domains using
SMART
3 0 , prediction of intracellular localization using PSORT 3 1 , searching for homologous domains in the ProDom database 32 , and searching for homologous motifs in ProSite database 3 3 . RESULTS Utilizing the methods set forth herein above, the sequences set forth herein were identified and cloned as being differentially expressed in bladder cancer. In both tables, hybridizations are compared. The sequences named in Table I are identified by clone number and accession number. This list includes sequences of known genes whose function in bladder cancer was heretofore unknown and which were now found to upregulated in bladder cancer. Nucleic acid sequences are provided in Table Ill. The sequences named in Table 11 are identified by clone number. This list includes sequences of novel genes which have no identity to known proteins or genes in the gene databases. Nucleic acid sequences are provided in Table IV. - 43 - WO 01/22864 PCT/USOO/41005 In both Tables I and II, the differential expression pattern of the different hybridization probes is provided. In both Table I and II, the genes listed were found to be upregulated in at least 60% of TCC samples and unchanged in at least 75% of the normal samples. Table I and 11 show the genes as described in biological NCBI databases, with the Genebank number of each gene as presented in the NCBI database. The location of the clone in the TCC microarray of the present invention is set forth in the tables, with their clone ID and the location of the clone in the TCC microarray. The expression differentials described in Tables I and II were calculated as follows: Since a common control probe was used for all hybridizations and the hybridizations were carried out in three separate sets, the expression differentials in each respective set were calculated as compared to one of the normal bladder samples, as a reference probe. Thus, hybridization set 1 (Columns 5-14) which includes hybridizations TC2A-TC11A, show all the results as compared to the TC7A (normal) hybridization result. The results are the calculated differential expression (see above for explanation of the calculations) of normal urothel samples (samples designated TC7A-TC11A) and of TCC samples (samples designated TC2A TC6A).. In hybridization set 2 which includes hybridizations TC16A-TC25A, all the results were calculated in comparison to the TC22A (normal) hybridization result. Set 2 is shown in columns 15-24 wherein there is shown the calculated differential expression (see above for explanation of the calculations) of normal urothel samples (samples designated TC21A-TC24A) and of TCC samples (samples designated TC16A-TC20A and TC25A). In set 3, which includes hybridizations TC28A-TC41A, all the results were calculated compared to the reference normal probe from TC47A. In set 3, as shown in Columns 25-45, the results are the calculated differential expression (see above for explanation of the calculations) of normal urothel samples (samples designated TC35A-TC38A and samples TC46A -44 - WO 01/22864 PCT/USOO/41005 TC48A) and of TCC samples (samples designated TC28A-TC34A and TC39 TC45A). The last column of Tables I and 11 show the differential expression of secreted membranal protein identification probe. The smaller the number in the result (negative number), the higher probability for such a gene to code for a secreted or membranal protein. The first set of raw data show the description of the grade of the TCC tumor i.e. G1-G3 or high / low grade (when available). The second set of raw data are for tumor material, this shows the description of the stage of the tumor i.e. Ta, T1, TIS (tumor in situ) or INV (invasive TCC). For normal urothel samples there is indicated the type of sample (e.g. normal). All the genes listed in these tables (1 and 2) can be classified as potential markers for non invasive transitional cell carcinoma. As for invasive TCC some of the genes also have diagnostic capabitily for advanced forms of invasive TCC, etc. This approach further enables the identification of potentially secreted molecules which can be used for detection of secreted proteins in urine of TCC patients. Each of the genes listed as potential marker for TCC can also be used for diagnosis of TCC in exfoliated cells in the urine by utilizing RT-PCR technology for the estimation of the level of expression of each of these genes in these cells.All the listed genes in table 1 and 2 can either be used individually or in combination (2-3 genes simultaneously) for more efficient diagnosis of all possible cases of TCC. Throughout this application, various publications, including United States patents, are referenced by author and year and patents by number. Full citations for the publications are listed below. The disclosures of these publications and patents in their entireties are hereby incorporated by reference into this application in order to more fully describe the state of the art to which this invention pertains. -45- WO 01/22864 PCT/USOO/41005 The invention has been described in an illustrative manner, and it is to be understood that the terminology which has been used is intended to be in the nature of words of description rather than of limitation. Obviously, many modifications and variations of the present invention are possible in light of the above teachings. It is, therefore, to be understood that within the scope of the described invention, the invention can be practiced otherwise than as specifically described. -46 - WO 01/22864 PCT/USOO/41005 REFERENCES AMERICAN CANCER SOCIETY, Estimating new cancer cases and deaths by sex for all sites, Cancer Facts and Figures, pg. 4 (1998) Burke and Olson, "Preparation of Clone Libraries in Yeast Artificial-Chromosome Vectors" in Methods in Enzymology, Vol. 194, "Guide to Yeast Genetics and Molecular Biology", eds. C. Guthrie and G. Fink, Academic Press, Inc., Chap. 17, pp. 251-270 (1991). Capecchi, "Altering the genome by homologous recombination" Science 244:1288-1292 (1989). Cregg JM, Vedvick TS, Raschke WC: Recent Advances in the Expression of Foreign Genes in Pichia pastoris, Bio/Technology 11:905-910, 1993 Culver, 1998. Site-Directed recombination for repair of mutations in the human ADA gene. (Abstract) Antisense DNA & RNA based therapeutics, February, 1998, Coronado, CA. Davies et al., "Targeted alterations in yeast artificial chromosomes for inter species gene transfer", Nucleic Acids Research, Vol. 20, No. 11, pp. 2693 2698 (1992). -47 - WO 01/22864 PCT/USOO/41005 Diatchenko et al., "Suppression subtractive hybridization: A method for generating differentially regulated or tissue-specific cDNA probes and libraries" Proc. Nat Acad. Sci., Vol. 93, pp. 6025-6030 (1996). Dickinson et al., "High frequency gene targeting using insertional vectors", Human Molecular Genetics, Vol. 2, No. 8, pp. 1299-1302 (1993). Duff and Lincoln, "Insertion of a pathogenic mutation into a yeast artificial chromosome containing the human APP gene and expression in ES cells", Research Advances in Alzheimer's Disease and Related Disorders, 1995. Gilboa, E, Eglitis, MA, Kantoff, PW, Anderson, WF: Transfer and expression of cloned genes using retroviral vectors. BioTechniques 4(6):504-512, 1986. GROSSMAN, HB, Semin. Urol. Oncol., 16(1):17-22 (1998) HUDSON MA, and HERR HW, Carcinoma in situ of the bladder, J. Urol., 153(3, Part 1):564-572 (1995) HUDSON, MA and HERR HW, Carcinoma in situ of the bladder, J. Urol., 153(3) Part 1):564-572 (1995) -48- WO 01/22864 PCT/USOO/41005 Huston et al, 1991 "Protein engineering of single-chain Fv analogs and fusion proteins" in Methods in Enzymology (JJ Langone, ed.; Academic Press, New York, NY) 203:46-88. Huxley et al., "The human HPRT gene on a yeast artificial chromosome is functional when transferred to mouse cells by cell fusion", Genomics, 9:742 750 (1991). Jakobovits et al., "Germ-line transmission and expression of a human-derived yeast artificial chromosome", Nature, Vol. 362, pp. 255-261 (1993). Johnson and Bird, 1991 "Construction of single-chain Fvb derivatives of monoclonal antibodies and their production in Escherichia coil in Methods in Enzymology (JJ Langone, ed.; Academic Press, New York, NY) 203:88-99. LACOMBE L et al., Overexpression of p53 protein in a high-risk population of patients with superficial bladder cancer before and after bacillus Calmette Guerin therapy; correlation to clinical outcome, J. Urol., 153(3, Part 1):564 572 (1995) Lamb et al., "Introduction and expression of the 400 kilobase precursor amyloid protein gene in transgenic mice", Nature Genetics, Vol. 5, pp. 22-29 (1993). -49- WO 01/22864 PCT/USOO/41005 Mernaugh and Mernaugh, 1995 "An overview of phage-displayed recombinant antibodies" in Molecular Methods In Plant Pathology (RP Singh and US Singh, eds.; CRC Press Inc., Boca Raton, FL) pp. 359-365. OZEN H., Curr. Opin. Oncol., 10(3):273-8 (1998) Pearson and Choi, Expression of the human f-amyloid precursor protein gene from a heast artificial chromosome in transgenic mice. Proc. NatI. Scad. Sci. USA, 1993. 90:10578-82. Rothstein, "Targeting, disruption, replacement, and allele rescue: integrative DNA transformation in yeast" in Methods in Enzymoloqy, Vol. 194, "Guide to Yeast Genetics and Molecular Biology", eds. C. Guthrie and G. Fink, Academic Press, Inc., Chap. 19, pp. 281-301 (1991). Schedl et al., "A yeast artificial chromosome covering the tyrosinase gene confers copy number-dependent expression in transgenic mice", Nature, Vol. 362, pp. 258-261 (1993). Strauss et al., "Germ line transmission of a yeast artificial chromosome spanning the murine a1 (1) collagen locus", Science, Vol. 259, pp. 1904-1907 (1993). TORTI FM and LURN BL, The biology and treatment of superficial bladder cancer, J. Clin. Oncol., 2(5):505-531 (1984) - 50 - WO 01/22864 PCTIUSOO/41005 'Al CIA!i~ i Nj 4 QR P"! 0O -n w U- Id w ell Cm C"C 4 q O O - C C to m-l: CO C9 cO N~C Ci C. )CO La w = " -7 1 '" q I - I I-~l 10 N P*C -a -C Cl a I T I I R co *~~~~1 S S7S~. i U Y ~ O N - C S N N or C~ N ID U7 C ID O ~ 5 &v -6 N8 N Wo IS * - NN -C co 7 . -M C-4 -a N2 m In -m mqN s Cq- ~ '3 S- -e L-L LL, I j-.; zz If I - I l et MC ac CL aI W,2f~ in -m _If L) U GA .85 8 L) w L) ID om -~ -. I -II' -; aClaJ s s' 2-~f -LI V T . a5 WO 01/22864 PCTUSOO/41005 Vt7 .~~~- 7 - - - -s-1 I cc P2 m V; ' ( . wi gri cn~V ea -n -3 -q -i rR I R - - - m - 'a- -cm -- TT 71.
'" ~ , 12 ad .- (.4 r- ~ 112 t2 (aR 17(1 (4(. 2 a~~ ~ 7. a7(~ ' a M.2 C4 'R '. 'R' P 1w qd Ia I - V! I W! 1- - I - r! -W c" C4 Mn aq m ~ Na C fm PI!- -__ a f-z f- a- *~C - - 11 -i a7 P .'7 17a, 72 q ... '7 - -r~ Z. C2 G ,! 1 12 .. .~ q - (a S ~'a--52 -' - a WO 0122864PCT/USOO/41005 Ldi~ -W -W 'NJ - Ir 7 I ai I~ * * ~ 111 1? 7 . - - - i ~ ~ -7' -7 1E 9D fa - ' - I 0 cc %" cmC N- m- -0 CR -1 I CM V-4 N a - n ~~~.% - C4E do. CD C12 ~ m ~ tn -D en E ;- ;7r - 1 CI :11 V - - 'C' ~ an- N II C, Iu Z3 7 L-1 i- !r 41- i z -,-Q 7Z3~~ . - m toe 9 _E a- 1Z5 -u U -u ci a n I ,w 4 Ul DIED" -53- WO 01/22864 PCTUSO0/41.005 7i 71 171 j H 7111 Lb : cc i2L ~ ~ L __ I ft v - - ~- - -qIL Ine~ e m C4 - i -. cqi .~ IQ n -T cm M N- -- I e,
-
7-" -R 1 70 -W - 1. P-. - - - - I p IN .l r4 I R Ir - -r 'I- fU -J 71 .i - r[= WO 01/22864 PCT/USOO/41005 TABLE III >40_TCC 13F11_M13F.fa TIME: Sun Sep 10 11:42:06 2000 trimming information: rawsequence:582 (high quality:29-320) sequence:97 252 [length:156] TCCGTCTCATTGAGGGTCCTGAGGAAGTTGATCTCATCATTCAGGGCATC CACCTTGGCCTCCAGCTCCACCTTGCTCATGTAGGCAGCATCCACATCCT TCTTCAGCACCACAAACTCATTCTCAGCAGCTGTGCGGCGGTTAATTTCA TCTTCG >04 TCC 94G3 M13F.TXT.fa ,constant: 15, poly A: yes AAGGCTTATTCCATCCGGACCGCATCCGCCAGTCGCAGGAGTGCCCGCGACTGAGCCGCC TCCCACCACTCCACTCCTCCAGCCACCACCCACAATCACAAGAAGATTCCCACCCCTGCC TCCCATGCCTGGTCCCAAGACAGTGAGACAGTCTGGAAAGTGATGTCAGAATAGCTTCCA ATAAAGCAGCCTCATTCTGAGGCCTGAGTGAAAAAAAAA >20 TCC 60H4 M13F.TXT.fa ,constant: -1, poly A: no CANTATATAACNAATTGGAGCTCAATNGCNCGCGGNCGCGTGTCTTCTGGGTAGAGGGAT GNGAAGGAAGGGACCCTTACCCCCGGCTCTTCTCCTGACCTGCCAATAAAAATTTATGGT CCAAGGNAAAANA >26_TCC_44C1 M13F.TXT.fa ,constant: -1, poly A: no ACTCATTGAACTTGAGCTCCGANTCCTGATTCNCATCNAAGCTCTNNATCTGCTCATCAN GAGANCCCACATCCTTGAGCAGATGGNGCANCTGCTGNTTAACCANCTCTNNGAACTCGN AGANNNTAAGGCTATCCTTCCGGNCCTCCTGCCTTGCAAAGGTGAAGAAAGTGGTGNNCA CNGTCNCAATGGANTCCTCTAGCTCTGTCAGTGGTTCTGCTGCNATTATGGAACCTGAGG CCAAAGCTGATGTCCTCAAGGGGCTAGCTGACCTTTGTCAGGGCTGACCTCTCCTCAGCG GCAGCAGGGCAGAGTGCTGAACCCAGGAACCCACAGATCCTCCCCGNTCCTGTCTCCCGG TGACAAGGGTCCTGGAACGGGGCGTCTCTGACTCCCTGCTCCAGGACGGGTTTAAGT >29_TCC_48G1 M13F.TXT.fa ,constant: -1, poly A: no ACTTTGAGAAGGCAGGACTCAAATGATGCCCTGGAGATGTCACAGATTCCTGGCAGAGCC ATGGTCCCAGGCTTCCCAAAAGTGTTTGTTGGCAATTATTCCCCTAGGCTGAGCCTGCTC ATGT >31_TCC_65B9 M13F.TXT.fa ,constant: -1, poly A: yes GACTAGAACCCACCCCCTTNCCTTCCAGCCTTTCTGTCATCATCTCCACAGNCCANCCAT CCCCTGAGCACACTAACCATCTCATGCAGGCCCCACCTGCCAATAGTAATAAAGCAATGT CACTTTGTTAAAACATGAAAAAAAAA >47 TCC 91B11 M13F.TXT.fa ,constant: -1, poly A: yes CTAGTATACACTCCNCATAGNATACGTTGCAGCTCAATTGCGCGCGGNCGCGGACGACGA CCTGCGAGGGTGTCTTCTGGGTAGAGGGATGGGAAGGAAGGGACCCTTACCCCCGGCTCT TCTCCTGACCTGCCAATAAAAATTTATGGTCCAAGGAAAAAAAAA >10 TCC 53H11 T3 TTTTTTNNATNTTATTTTGGGTATTGGTGTTNTTTCTTTTTTCCTCTTNCCTTCTTAACT CAAGACTTGTAGTGTTGTAAACCTGCCTCACAAAATACATGGTAATAACTTNTCTTTAAA AAAANAAAAAAGACAGNCTTNACACCATTTCTAATNGNANNACTATTTTTGGGCAATGTT ATGCACCACTTCAATTTCCCCATTGTGACCCCTATCACTTCATTTGATATCCCTTTTNGA CCCANCCATCTCCTTCATATATGGGCATGTCCATAGATTGACAAAGAAAGTTTACACTTT NGAATAAAGATGCAAAGTATGCAAAAACATTAATACTGATGCNAAAAAAANTANAAAAA -55 - WO 01/22864 PCT/USOO/41005 >07 TCC 57B3 M13F.TXT ,constant: -1, poly A: yes GGTACCGACGGACCTGCGGAGACTCCTGCCCTGTTGTGTATAGATGCAAGATATTTATAT ATATTTTTGGTTGCAATATTAAATACAGACACTAAGTTATAGTATATCTGGCAAGCCAAC TTGTAAATCACCACCTCACTCCTGTACTTACCTAAACAGATATAAATGGCTGGTTTTTAA GAAAAAAAAA >11_TCC_25F2 M13F.TXT ,constant: -1, poly A: no ACCCTGGGAGAGAAGTTTGAAGAAACCACAGCTGATGGCAGAAAAACTCAGACTGCTGCA ACTTTACAGATGGTGCATTGNGTCAGCATAGGAGTGAGATGGGGAAGGAAAGCACANTAA CAAGAAAATTGANAGATGNTAAATTAGTGNTGGAGTCTGTCATGAACAATGCACCTGT >25 TCC 50G5 M13F.TXT ,constant: 17, poly A: yes TAGTGTGGAAGCATAGTGAACACACTGATTAGGTTATGGTTTAATGTTACAACAACTATT TTTTAAGAAAAACATGTTTTAGAAATTTGGTTTCAAGTGACATGTGTGAAAACAATATCG ATACTACCATAGTGAGCCATGATTTTCTAAAAAAAAAA >26 TCC 50G6 M13F.TXT ,constant: 17, poly A: yes TAGTGTGGAAGCATAGTGAACACACTGATTAGGTTATGGTTTAATGTTACAACAACTATT TTTTAAGAAAAACAAGTTTTAGAAATTTGGTTCAAGTGACATGTGTGAAAACAATATTGT ATACTACCATAGTGAGCCATGATTTTCTAAAAAAAAA >26_TCC 75E3 M13F B04 032.abl.TXT ,constant: 16, poly A: yes AAAGAGGGCGGCAGGGGCCTGGAGATCCTCCTGCAGACCACGCCCGTCCTGCCTGTGGCG CCGTCTCCAGGGGCTGCTTCCTCCTGGAAATTGACGAGGGGTGTCTTGGGCAGAGCTGGC TCTGAGCCGCCCTCCATCCAAGGCCAGGTTCTCCGTTAGCTCCTGTGGCCCCACCCTGGG CCCTGGGCTGGAATCAGGAATATTTTCCAAAGAGTGATAGTCTTTTTGCTTTTTGGCAAA ACTCTACTTAATCCAATGGGTTTTTCTCTGTACAGTAGATTTTCCAAATGTAATAAACTT TAATATAAAGTAAAAAAAAA >30 TCC 76B3 M13F F04 042.abl.TXT ,constant: 16, poly A: yes AAAGTCATCCTCCGTCTACCAGAGCGTGCACTTGTGATCCTAAAATAAGCTTCATCTCCG GGCTGTGCCCCTTGGGGTGGAAGGGGCAGGATTCTGCAGCTGCTTTTGCATTTCTCTTCC TAAATTTCATTGTGTTGATTTCTTTCCTTCCCAATAGGTGATCTTAATTACTTTCAGAAT ATTTTCAAAATAGATATATTTTTAAAATCCTTAAAAAAAAA >38 TCC 56Ell M13F.TXT ,constant: -1, poly A: yes CTCTCCAGTTTGCACCTGTCCCCACCCTCCACTCAGCTGTCCTGCAGCAAACACTCCACC CTCCACCTTCCATTTTCCCCCACTACTGCAGCACCTCCAGGCCTGTTGCTATAGAGCCTA CCTGATGTCAATAAACAACAGCTGAAGCAAAAAAAAA >46 TCC 78B11 M13F F06 058.abl.TXT ,constant: 16, poly A: yes AGGAAAGGTGNGNGCTGGAAGCACTGAACCTACCTCATCCTCCTGGTGGGTGTGGCTACC CTCGCCACCCCAAATTCCATGTCATTAAAGAACAGCTAAATTCAAAAAAAAA >53 TCC 79G2 M13F E07 054.abl.TXT ,constant: 16, poly A: no TGTCCGTCTTCACCCATCCCCAAGCCTACTAGAGCAAGAAACCAGTTGTAATATAAAATG CACTGCCCTACTGTTGGTATGACTACCGTTACCTACTGTTGTCATTGTTATTACAGCTAT GGCCACTATTATTAAAGAGCTGTGTAACATCAAAAAAA >82 TCC 89G3 M13F Bli 092.abl.TXT ,constant: 16, poly A: yes CAGGAGACCATCCGCGTCACCAAGCCCTGCACCCCCAAGACCAAAGCAAAGGCCAAAGCC AAGAAAGGGAAGCGAAAGGACTAGACGCCAAGCCTGGATGCCAAGGAGCCCCTGGTGTCA CATGGGGCCTGGCCCACGCCCTCCCTCTCCCAGGCCCGAGATGTGACCCACCAGTGCCTT CTGTCTGCTCGTTAGCTTTAATCAATCATGCCCTGCCTTGTCCCTCTCACTCCCCAGCCC CACCCCTAAGTGCCCAAAGTGGGGAGGGACAAGGGATTCTGGGAAGCTTGAGCCTCCCCC AAAGCAATGTGAGTCCCAGAGCCCGCTTTTGTTCTTCCCCACAATTCCATTACTAAGAAA CACATCAAATAAACTGACTTTTTCCCCCCAAAAAAAAA - 56- WO 01/22864 PCT/USO0/41005 >35 TCC 21D6 M13F C05 037.abl.fa TIME: Wed Aug 9 12:48:31 2000 trimming information: rawsequence:889 (high quality:34-340) sequence:95-456 [length:362] CTTTGACGTGGAGAGGAACTCCTGCAATAACTTCATCTATGGAGGCTGCC GGGGCAATAAGAACAGCTACCGCTCTGAGGAGGCCTGCATGCTCCGCTGC TTCCGCCAGCAGGAGAATCCTCCCCTGCCCCTTGGCTCAAAGGTGGTGCT TCTGGCGGGGCTGTTCGTGATGGTGTTGATCCTCTTCCTGGGAGCCTCCA TGGTCTACCTGATCCGGGTGGCACGGAGGAACCAGGAGCGTGCCCTGCGC ACCGTCTGGAGCTCCGNAGATGACAAGGAGCAGCTGGTGAAGAACACATA TGTCCTGTGACCGCCCTGTCGCCAAGAGGACTGGNGAAAGGGAGGGGAGA CTATGTGTGAGC >46 TCC 27H5 M13FF06 058.abl.fa TIME: Wed Aug 9 12:48:35 2000 trimming information: raw sequence:892 (high quality:169-406) sequence:170-287 [length:118] AAAAAGAGTAAAACACTTTCAGTTTCTCCCCTTTAGCCCCTAAAACAACA TCTTACAGTCTGGATCTGGATCTACCTATACAGTCCTACATTAGCTTCTA AAATATTTGTCAGGAGGG -57- WO 01/22864 PCT/USOO/41005 TABLE IV >31 TCC 10E8_M13F.fa TIME: Sun Sep 10 11:42:01 2000 trimming information: rawsequence:549 (high quality:25-313) sequence:98 313 [length:216] CCCAAATGGAATGTTGCCCCCTTAAACACCATTTTCCCTCCAGGACCACC TTGGTTTCTAGGCACTGTGGTTCTTGGCAGGGGCTGTCTTAGGTAAAAGG GTAGTTGTGGAGCTACAGTCTGAAGAACATAGCTTGGGCTCAAGTTCAAA TGAGCCATCTTTTTCCTTTGCGTTTTTCTTGACTGAAGGTGAGATGTTAT TTGTGGCATGTGAACT >09 TCC 101C11 M13F.TXT.fa ,constant: 16, poly A: yes ACAAAGACTGCTGATAACTATCTGTGATTGATAGGAAATTTTTTTTCTTGATTTCTCTGT GAGAAATGTAATGCTGACTTTTATAAAGCCTGGACTTCTACTTTATTTAATAAATCAATG TTTGCAATGGTAAAAAAAAA >11 TCC lOlEll M13F.TXT.fa ,constant: 15, poly A: yes GCAATAAAGCTGTCCATTCAATTCCAAATACTGGTTTTAAGNGTATAGCCACTGATATTC TTTCATGTNTAGAAATTCTTTCTGTTATTATTCAAGAAAATGTTTTTAATCATGCTAATA AACTTTTTTGGAGATGAAAAAAAAA >15_TCC_57C3_M13F.TXT.fa ,constant: -1, poly A: no GGNACCACGTACCTGCTGAATGTNTCNNCGNNATGNCGNCAGGCCATGCTGTTGCTGATN TANTACTNTGAAAATANGGATATCATGATGGGAATGCATGTCATGAGGTCCAGANTCGTT CTACTGTCNATAANCTGTNTACTNGCGTTGANAANAAANGATGTCAAAGNCCCCCCGTAA AAANGTA >44 TCC 70E8 M13F.TXT.fa ,constant: 15, poly A: yes GTCCCAGTCTTCACCAGGTGTCTCTCCTCTTTTACTCAGGAGGACTTTCCCAGGAAAACC ATGCCACTAGCAAAAAAAAA >03 TCC 57E11 T7.TXT.fa ,constant: 16, poly A: yes TGAGTGTCTTCAGGCCAACCTGGTGGAAATGTTGTTCTCTGAAGATTAAGATTTTAGGAT GGCAATCATGTCTTGATGTCCTGATTTGTTCTAGTATCAATAAACTGTATACTTGCTTTG AATTCATGTTAGCAATAAATGATGTTAAAAAAAAAA >08 TCC 70E7 M13F H01 015.abl.TXT ,constant: -1, poly A: no GGATCGACGACCTGCTTCCCAGANGCGNNCNNGAGGNCCNCTTGTTNNNGNCNNGNANAC NNACCCANTTNANTTNNAGCCTTTNTGNAATAAATATACACAGGCCACCCATGCCNTGAG CACACTAACCACNTGATGCAGGCCCCACCTTGCCAATAGTAATAAAGCANTGGGACGTTT TTTA >13_TCC_71E4 M13FE02_018.abl.TXT ,constant: -1, poly A: no GGGCCAAAGCCCGNGCATCCAANCCCANGCAAGGNACAAANGANCNNGGAGAGGANNACC CAAGCANNTNNCAACCATCAAATGGAGGGCANGCCCGGGG >15 TCC 71H8 M13F G02 019.abl.TXT ,constant: -1, poly A: no GGGCCAAAGCCGNGCATCCAANCCCANCGCANGGNANAAANGANGANGGANANGGATNAC CCANGCCTNTATTAACCATCAANTGGGANGGCAAGCCCGGGGCATNTATTGATT >21 TCC 43E2 M13F.TXT ,constant: -1, poly A: no AGGACCCCTGAANACNACACAGATCTGTGNGAAACAANGGNACNTAGCGTCCCNAAAGTG CCNGGTTNNNGTANNCNNAGNGNGNGACCNGNGCNCATNT -58 - WO 01/22864 PCT/USOO/41005 >24 TCC 96C7_M13F.TXT ,constant: 16, poly A: yes ATCCAGAGACCATCAATCCTGCTAGAGTGCAGGGTGGCAAGCACCCAAGGGTGGCTGACC AAGACTGCAGAGTCTCCTCCATCTTCAGGTCCATTCAGCCTCCTGGCATTTAACTACCAG CATCCAGTGGTCCCCAAGGAATCCCTTCCTAGCCTCCTGACATGAGTCTGCTGGAAAGAG CATCCAAACAAACAAGTAATAAATAAATAAATAAACTCAAAAAAAAA >57 TCC_80C9_M13F A08 056.abl.TXT ,constant: 17, poly A: yes CTGCAGGAGTCAGCGTTCAATCTTGACCTTGAAGATGGGAAGGATGTTCTTTTTACGTAC CAATTCTTTTGTCTTTTGATATTAAAAAGAAGTACATGTTCATTGTAGAGAATTTGGAAA CTGTAGAAGAGAATCAAGAAGAAAAATAAAAATCAGCTGTTGTAATCACCTAGCAAAAAA AAA >14 TCC_9B6_M13FF02 026.abl.fa TIME: Wed Aug 9 12:48:25 2000 trimming information: rawsequence:871 (high quality:73-413) sequence:98-394 [length:297] CACGCATATGGGGCCAGTTCCACATATTTGGCAACCAGACCAGCATCCAG GACAACACAAAGTATGTTGTTTGTTGTTAGAGGGCTTGGGACATTTCACT CTTTGCCAGCCTCAGCTTAATCCAGGAGACAAAGATTATTTTCCTTATTA TCTCTTCTGCATAGGATCTGCAATCAGAACTATTGAACTTCTCCATTCAG ACCGCCACTCACACCTATGGGAAAAGGGTAATGTATCATCGGCTTAGCAA CAGGGAATACTATTCGTATGATGGAAAATGGGGACAAAAGGCTTTGG >24_TCC_12F3_M13FH03_031.abl.fa TIME: Wed Aug 9 12:48:28 2000 trimming information: raw sequence:842 (high quality:82-340) sequence:98-476 [length:379] CTATGAATAGCTTCTTGCTTTATGACTTTAGGATTAACTTGTAAAAAACA TATCCTGAACTAAGATATGCAAAATACTCATTTTCAAGTTATGGAAATGT GTTTGTGGCATATAGGACTGTGGGGTCTGTGTGTGTAGTGAGAGTGTGTA TCCACTATTATAACTGGAATTTAATTTACATTCATAAACTACTATATTTC CCATCTTGCAAATCATTTTATGTCTCATCTGTTTTTCCTTTCGGNTATAT CTTTGGNTTTGAATACCAACATTTAAAATGATGGNATTTTATCTTTTAAA CTTAAAAATTATTTAATACAGCTATATGGACCTTATAAAATTGATTTCTT ATTTATTATTAGACATTACTACTAAAAGG >26_TCC_13H10_M13FB04_032.abl.fa TIME: Wed Aug 9 12:48:29 2000 trimming information: rawsequence:874 (high quality:67-356) sequence:99-261 [length:163] CTAACCCACGATTCTGAGCCCTGAGTATGCCTGGACATTGATGCTAACAT GACCATGCTTGGGATGTCTCTAGCTGGTCTGGGGATAGCTGGAGCACTTA CTCAGGTGGCTGGTGAAATGACACCTACGAAGGAATGAGTGCTATAGAGA GGAGAGAGGAGTG >28 TCC 16D12 M13F D04_041.abl.fa TIME: Wed Aug 9 12:48:29 2000 trimming information: rawsequence:866 (high quality:71-411) sequence:95-602 [length:508] CAGCTGATGTCATGTGGTGCTGAGAAGAAAGCAGATCACACTTCATCACA GAAAGAATGCCTTGTGATTATCTTCTCCACATCTGAAATTCCTTTTGACA CCTGCATTGGGCCGACTGCCATTCCCATGACTGCTGCACCTGCGTTTTTA GAGAATGCCTCATAACCCACTGATTCTCATTCACAGAGAATGGGAATACG GAATGAAGAAAGATTCCAGCAGCTTATAGAAGGATAGCAATATTTTGGGA CAGGGAAAATCCTGTCATACCTCACCTCTTCCTCAGGAGGAGTTCTGAGC TGGTCCTGCTTTTCATAGNTGTTTCTTTTCTTCCACTTAAGAACTCATAG ATTTTTCTTACTGTCCTAAGGAAGTCCTTACCTCTGAGGTATCTCCTCAA TGAATACTGTTTTCAAGGCTGAAATAGTTCATTATGTTAATAACCTTCTT TATGTTCTCAGGGAAATGCTTAGGT GGTGTCACAAAAAGGGCCTTTTCTT TNCTTTNC -59- WO 01/22864 PCT/USOO/41005 >29 TCC 17A5 M13F E04 034.abl.fa TIME: Wed Aug 9 12:48:30 2000 trimming information: raw sequence:861 (high quality:83-477) sequence:99-187 [length:89] CTTCAAAAAGTGTATTGTCAAACATACCTAACTTTCTTGCAATAAATGCA AAAGAAACTGGAACTTGACAATTATAAATAGTAATAGTG >54 TCC 30E5 M13F F07 062.abl.fa TIME: Wed Aug 9 12:48:37 2000 trimming information: rawsequence:836 (high quality:65-394) sequence:90-235 [length:146] CAATTTGTTATAGTATAGTATCAAATTTCTATATAGATTTTATACCTCAG TGGGGAAAAATAACTGATTCCAATGACATTCATTTTGTTTTCATCTGTGA TAGTCATGGATGCTTTTATTTTCCTTGGGGTGCTGAAATTGAGCTG >59 TCC 34D5 M13F C08 065.abl.fa TIME: Wed Aug 9 12:48:39 2000 trimming information: rawsequence:875 (high quality:63-434) sequence:96-244 [length:149] CCTGCCAAAATCCTACCACAGGATAACATTACAAGCAAAAAATTTACATG TTCCAAAGTCTACCACACTCAAGAAGTTACTAAGAACTCTTGCAGAATAA AAGTCACCATTTTAGAAATGCAAACCCACTTCCAACCTTTGCACAGTCC >72 TCC 37E11 M13F_ H09079.abl.fa TIME: Wed Aug 9 12:48:43 2000 trimming information: raw_sequence:899 (high quality:35-432) sequence:97-444 [length:348] CATTTTTAGTGACATTTTAAAAGCAGTCAGATTCTATAAATGGCAAGTAA GCCTGAAGTGAGGATACTGCAATTTTCGGAGAAAAGAACAGCAGCTCTTT AAGTGTTTGCATTTTCTATTTGGGGGGCAGGGAACTGTCATTCATTTTGC ACAATTCTTGAACTGATGTCAGCACCCGAGTGGCTCCTGAATTTAAGTCT GGGACGACATCTTTTATTTTTACATGAATCTTTAAACAATTCTGTGAGCA AAGTTTGTAGCTGCTGGATTATTGTCTGTCTTTATAGCAAGTTCCAGTAA ACCACAAGTATGGCAAAGCTTATCCAATTTTATGCTTGNAGCAGTCAG -60-

Claims (16)

1. A method of diagnosing the presence of bladder cancer in a patient by analyzing a tissue sample from the patient for the presence of at least one expressed gene wherein the presence of the expressed gene is indicative of bladder cancer.
2. The method of claim 1, wherein the analyzing step further includes using mRNA from the expressed gene to hybridize to at least one of the sequences in Tables Ill or IV.
3. A polynucleotide sequence whose expression is indicative of bladder cancer.
4. The polynucleotide sequence according to claim 3, wherein said sequence is selected from the group consisting essentially of at least one of the sequences in Tables IlIl or IV.
5. A marker for bladder cancer, wherein said marker is an expressed gene wherein the presence of the expressed gene is indicative of bladder cancer.
6. The marker according to claim 5, wherein said marker is a polynucleotide sequence which is selected from the group consisting essentially of at least one of the sequences in Tables Ill or IV.
7. A method of diagnosing bladder cancer by screening for the presence of at least one expressed gene wherein the presence of the expressed gene is indicative of bladder cancer. - 61 - WO 01/22864 PCT/USOO/41005
8. The method of claim 7, wherein the analyzing step further includes using mRNA from the expressed gene to hybridize to at least one of the sequences in Tables Ill or IV.
9. Antibodies directed against the gene products of the sequences of claim 3.
10. The antibodies according to claim 9, wherein said antibodies are selected from the group consisting essentially of monoclonal, polyclonal and recombinant antibodies.
11. A method of treating bladder cancer-associated pathologies by administering to a patient a therapeutically effective amount of an antagonist of at least one protein as encoded by the nucleic acid sequences set forth in claim 3 or by the probes thereof.
12. A method of regulating bladder cancer-associated pathologies in a patient in need of such treatment by administering to a patient a therapeutically effective amount of at least one antisense oligonucleotide against the nucleic acid sequences set forth in claim 3 or dominant negative peptide directed against the sequences or proteins thereof.
13. A method of regulating bladder cancer-associated pathologies in a patient in need of such treatment by administering to a patient a therapeutically effective amount of at least one ribozyme against the nucleic acid sequences set forth in claim 3.
14. The method according to claim 13, wherein said administering step includes administering a ribozyme selected from the group consisting essentially of Group I introns, RNase P, hepatitis delta virus ribozyme, hammerhead - 62 - WO 01/22864 PCT/USOO/41005 ribozymes and a hairpin ribozyme originally derived from the negative strand of the tobacco ringspot virus satellite RNA.
15. A gene therapy vehicle for delivering the sequences as set forth in claim 3 whereby the sequences are expressed in the target cells.
16. The gene therapy vehicle according to claim 15, wherein said vehicle is selected from the group consisting essentially of viral vectors, non-viral vectors, expression cassettes, and constructs. - 63 -
AU14926/01A 1999-09-27 2000-09-27 Sequences characteristic of bladder cancer Abandoned AU1492601A (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US15615399P 1999-09-27 1999-09-27
US60156153 1999-09-27
PCT/US2000/041005 WO2001022864A2 (en) 1999-09-27 2000-09-27 Sequences characteristic of bladder cancer

Publications (1)

Publication Number Publication Date
AU1492601A true AU1492601A (en) 2001-04-30

Family

ID=22558336

Family Applications (1)

Application Number Title Priority Date Filing Date
AU14926/01A Abandoned AU1492601A (en) 1999-09-27 2000-09-27 Sequences characteristic of bladder cancer

Country Status (5)

Country Link
EP (1) EP1248855A4 (en)
AU (1) AU1492601A (en)
CA (1) CA2385893A1 (en)
IL (1) IL148798A0 (en)
WO (1) WO2001022864A2 (en)

Families Citing this family (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6998232B1 (en) 1999-09-27 2006-02-14 Quark Biotech, Inc. Methods of diagnosing bladder cancer
WO2002086084A2 (en) * 2001-04-04 2002-10-31 Quark Biotech, Inc. Sequence characteristics of bladder cancer
DK2444505T3 (en) 2004-07-23 2017-01-02 Pacific Edge Ltd Urinary markers for the detection of bladder cancer
EP1856278A2 (en) * 2005-02-10 2007-11-21 Oncotherapy Science, Inc. Method of diagnosing bladder cancer

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5856136A (en) * 1996-07-03 1999-01-05 Incyte Pharmaceuticals, Inc. Human stem cell antigens
DE19818619A1 (en) * 1998-04-21 1999-10-28 Metagen Gesellschaft Fuer Genomforschung Mbh New nucleic acid sequences expressed in bladder tumor tissue, and derived polypeptides, for treatment of bladder tumor and identification of therapeutic agents

Also Published As

Publication number Publication date
WO2001022864A3 (en) 2001-11-01
WO2001022864A9 (en) 2002-12-12
CA2385893A1 (en) 2001-04-05
EP1248855A2 (en) 2002-10-16
EP1248855A4 (en) 2004-12-22
IL148798A0 (en) 2002-09-12
WO2001022864A2 (en) 2001-04-05

Similar Documents

Publication Publication Date Title
US6998232B1 (en) Methods of diagnosing bladder cancer
US6458939B1 (en) Compositions and methods for the diagnosis, prevention, and treatment of neoplastic cell growth and proliferation
JP3399539B2 (en) Methods for diagnosing predisposition to breast and ovarian cancer
CA1341576C (en) Diagnosis of retinoblastoma
EP0826031B1 (en) Ataxia-telangiectasia gene and its genomic organization
US9909189B2 (en) Distinguishing PCA3 messenger RNA species in benign and malignant prostate tissues
KR20110084995A (en) Recurrent gene fusions in prostate cancer
EP1009753B1 (en) Hypoxia-regulated genes
CN102317470A (en) Genetic variants contributing to risk of prostate cancer
US5777093A (en) cDNAs associated with ataxia-telangiectasia
US20030148341A1 (en) Gene amplification and overexpression in cancer
JP2004505631A (en) A new member of the lysyl oxidase gene family
JP2009165473A (en) Cancer
AU1492601A (en) Sequences characteristic of bladder cancer
US8216792B2 (en) Compositions and methods for detection and treatment of proliferative abnormalities associated with overexpression of human transketolase like-1 gene
US20030175763A1 (en) Identification of an amplified gene and target for drug intervention
US20050059011A1 (en) Amplification and overexpression of oncogenes
US20030082720A1 (en) Compositions methods and kits relating to treating and diagnosing hypertension
US5728807A (en) Mutated proteins associated with ataxia-telangiectasia
JP2000500329A (en) Diagnosis and treatment of cancer susceptibility
EP1370693B1 (en) Amplified cancer gene wip1
EP1421214A2 (en) Amplified oncogenes and their involvement in cancer
US7973156B2 (en) Hypoxia-regulated genes
WO2002086084A2 (en) Sequence characteristics of bladder cancer
US20030099985A1 (en) Amplified gene involved in cancer

Legal Events

Date Code Title Description
MK1 Application lapsed section 142(2)(a) - no request for examination in relevant period