METHOD FOR DIAGNOSING NON-SMALL CELL LUNG CANCERS BY tRNA-DIHYDROURIDINE SYNTHASE ACTIVITY OF URLC8

Nakamura; Yusuke ;   et al.

Patent Application Summary

U.S. patent application number 12/782641 was filed with the patent office on 2011-01-20 for method for diagnosing non-small cell lung cancers by trna-dihydrouridine synthase activity of urlc8. This patent application is currently assigned to Oncotherapy Science, Inc.. Invention is credited to Yataro Daigo, Yusuke Nakamura, Shuichi Nakatsuru.

Application Number20110014615 12/782641
Document ID /
Family ID35559267
Filed Date2011-01-20

United States Patent Application 20110014615
Kind Code A1
Nakamura; Yusuke ;   et al. January 20, 2011

METHOD FOR DIAGNOSING NON-SMALL CELL LUNG CANCERS BY tRNA-DIHYDROURIDINE SYNTHASE ACTIVITY OF URLC8

Abstract

The present invention features a method for determining t-RNA dihydrouridine-synthase activity of a polypeptide and screening for modulators of t-RNA dihydrouridine-synthase activity. The present invention further provides methods or pharmaceutical compositions for preventing and/or treating non-small cell lung cancer (NSCLC) using such modulators. Furthermore, the present invention provides methods for diagnosing non-small cell lung cancer (NSCLC) using the t-RNA dihydrouridine-synthase activity of IMS-E21 (URLC8) protein as an index. The present invention further provides methods for predicting and prognosing lung squamous-cell carcinoma (SCC).


Inventors: Nakamura; Yusuke; (Tokyo, JP) ; Daigo; Yataro; (Tokyo, JP) ; Nakatsuru; Shuichi; (Kanagawa, JP)
Correspondence Address:
    TOWNSEND AND TOWNSEND AND CREW, LLP
    TWO EMBARCADERO CENTER, EIGHTH FLOOR
    SAN FRANCISCO
    CA
    94111-3834
    US
Assignee: Oncotherapy Science, Inc.
Kawasaki-shi
JP

Family ID: 35559267
Appl. No.: 12/782641
Filed: May 18, 2010

Related U.S. Patent Documents

Application Number Filing Date Patent Number
11575812 Oct 8, 2007
PCT/JP05/17915 Sep 21, 2005
12782641
60612937 Sep 24, 2004

Current U.S. Class: 435/6.11 ; 435/6.13
Current CPC Class: G01N 2333/99 20130101; G01N 33/57423 20130101; C12Q 1/6886 20130101; G01N 2500/02 20130101; A61P 11/00 20180101; C12Q 2600/118 20130101; C12Q 1/533 20130101; C12Q 2600/136 20130101; A61P 35/00 20180101
Class at Publication: 435/6
International Class: C12Q 1/68 20060101 C12Q001/68

Claims



1.-9. (canceled)

10. A method of identifying an agent that modulates t-RNA dihydrouridine-synthase activity, said method comprising the steps of: a. incubating in the presence of a test compound under conditions suitable for the synthesis of t-RNA dihydrouridine a polypeptide selected from the group consisting of: i. a polypeptide comprising the amino acid sequence of SEQ ID NO: 2 (URLC8); ii. a polypeptide comprising the amino acid sequence of SEQ ID NO: 2 wherein one or more amino acids are substituted, deleted, or inserted, provided said polypeptide has a biological activity equivalent to the polypeptide consisting of the amino acid sequence of SEQ ID NO: 2; iii. a polypeptide encoded by a polynucleotide that hybridizes under stringent conditions to a polynucleotide consisting of the nucleotide sequence of SEQ ID NO: 1, provided the polypeptide has a biological activity equivalent to a polypeptide consisting of the amino acid sequence of SEQ ID NO: 2; b. detecting a t-RNA dihydrouridine synthesis level; and c. comparing the t-RNA dihydrouridine synthesis level to a control level, wherein an increase or decrease in the t-RNA dihydrouridine synthesis level as compared to said control level indicates that the test compound modulates t-RNA dihydrouridine-synthase activity.

11. A method of screening for a compound for treating and/or preventing non-small cell lung cancer (NSCLC), said method comprising the steps of: a. identifying a test compound that modulates t-RNA dihydrouridine-synthase activity by the method of claim 10, and b. selecting a compound that decreases the t-RNA dihydrouridine synthesis level as compared to a control level.

12.-20. (canceled)

21. The method of claim 10, wherein the polypeptide comprising the amino acid sequence of SEQ ID NO:2 wherein 25 or less of amino acids are substituted, deleted, or inserted, provided said polypeptide has a t-RNA dihydrouridine-synthase activity.

22. The method of claim 21, wherein DSRM (double-strand RNA binding motif) in the amino acid sequence of the polypeptide is conserved.

23. The method of claim 10, wherein the polypeptide encoded by a polynucleotide that hybridizes under stringent conditions to a polynucleotide consisting of the nucleotide sequence of SEQ ID NO: 1, provided the polypeptide has a t-RNA dihydrouridine-synthase activity, wherein the stringent condition comprises washing 3 times in 2.times.SSC, 0.01% SDS at room temperature for 20 minutes, then washing 3 times in 1.times.SSC, 0.1% SDS at 37.degree. C. for 20 minutes, and washing twice in 1.times.SSC, 0.1% SDS at 50.degree. C. for 20 minutes.

24. The method of claim 23, wherein DSRM (double-strand RNA binding motif) in the amino acid sequence of the polypeptide is conserved.
Description



CROSS-REFERENCES TO RELATED APPLICATIONS

[0001] This application is a division of U.S. application Ser. No. 11/575,812, filed Oct. 8, 2007, which is a U.S. National Stage of PCT/JP2005/017915, filed Sep. 21, 2005, which claims the benefit of U.S. Provisional Application Ser. No. 60/612,937, filed on Sep. 24, 2004, the contents of which are incorporated by reference herein in their entirety.

FIELD OF THE INVENTION

[0002] The present invention relates to lung cancer, more particularly non-small cell lung cancer, and the diagnosis and treatment thereof.

BACKGROUND OF THE INVENTION

[0003] Lung cancer is one of the most common causes of cancer death worldwide, and non-small cell lung cancer (NSCLC) accounts for nearly 80% of those cases (Greenlee, R. T., et al., (2001) CA Cancer J Clin, 51: 15-36.). Many genetic alterations associated with the development and progression of lung cancer have been reported, but the precise molecular mechanisms remain unclear (Sozzi, G. Eur J Cancer, (2001) 37 Suppl 7: S63-73.). Over the last decade, newly developed cytotoxic agents, including paclitaxel, docetaxel, gemcitabine, and vinorelbine, have emerged to offer multiple therapeutic choices for patients with advanced NSCLC; however, each of the new regimens can provide only modest survival benefits as compared to cisplatin-based therapies (Schiller, J. H. et al. (2002) N Engl J Med, 346: 92-98.; Kelly, K., et al. (2001) J Clin Oncol, 19: 3210-3218.). Hence, the development of new therapeutic strategies, such as molecular-targeted agents and antibodies, and cancer vaccines, are eagerly awaited.

[0004] Systematic analysis of expression levels of thousands of genes on cDNA microarrays is an effective approach to identifying unknown molecules involved in pathways of carcinogenesis, and can reveal candidate targets for development of novel therapeutics and diagnostics. The present inventors have been attempting to isolate novel molecular targets for diagnosis, treatment and prevention of NSCLC by analyzing genome-wide expression profiles of NSCLC cells on a cDNA microarray containing 23,040 genes, using pure populations of tumor cells prepared from 37 cancer tissues by laser-capture microdissection (Kikuchi, T. et al., Oncogene, (2003) 22: 2192-2205.; Zembutsu, H. et al. (2003) Int J Oncol, 23: 29-39.; Kakiuchi, S. et al. Mol Cancer Res, (2003) 1: 485-499.; Suzuki, C. et al., (2003) Cancer Res, 63: 7038-7041.). Through this genome wide cDNA microarray analysis, 642 up-regulated genes and 806 down-regulated genes have been identified as diagnostic markers and therapeutic targets for NSCLC (WO 2004/31413).

BRIEF SUMMARY OF THE INVENTION

[0005] To verify the biological and clinicopathological significance of the respective gene products, tumor-tissue microarray analysis of clinical lung-cancer materials have been performed. This systematic approach revealed that a novel gene, tentatively named IMS-E21 (also known as URLC8: up-regulated in lung cancer 8, Accession No. AB101210; formerly FLJ20399), was frequently over-expressed in primary NSCLCs.

[0006] The URLC8 gene encodes a double-strand RNA binding motif (DSRM) domain. The lowered expression of this gene in normal tissues, elevated expression in NSCLCs, and reduced growth, proliferation and/or survival of transfected cells by the suppression of this gene together suggest that this gene might be useful as a novel diagnostic marker and target for new drugs and immunotherapy (WO2004/31413).

[0007] The URLC8 gene has been assigned to chromosome 16q22.2 and encodes a protein of 493 amino acids with 30% homology to S. cerevisiae Dus1 (dihydrouridine synthase 1), a member of UPF0034 (unclassified protein family 0034), which catalyses the reduction of the 5,6-double bond of a uridine residue on D-loop in tRNA (Xing, F. et al., RNA, (2002) 8: 370-381.), and contains conserved DSRM (double-strand RNA binding motif).

[0008] 5,6-Dihydrouridine is a modified base found abundantly in the D-loops of tRNA from Archaea, Bacteria, and Eukarya. 5,6-Dihydrouridine is thought to be formed post-transcriptionally by the reduction of uridines in tRNA transcripts. The role of dihydrouridine in tRNA was presumed to increase the conformational flexibility of the tRNA. Interestingly, an increase in the level of dihydrouridine was previously reported in tumor-specific tRNA.sup.Phe purified from human malignant tissues (Kuchino, Y. and Borek, E. (1978) Nature, 271: 126-129.), however, its precise mechanism and biological contribution to tumorigenesis remain unclear (Dalluge, J. J. et al., (1996) Nucleic Acids Res, 24: 1073-1079.).

[0009] Herein the present inventors report the identification and functional characterization of a novel human tRNA-dihydrouridine synthase (DUS) protein, URLC8, in human lung tumors. The results herein suggest that overexpression of URLC8 plays a significant role in development/progression of lung cancer and that this molecule represents a potential target for development of novel therapeutic drugs.

[0010] The present invention is based in part on the discovery of the t-RNA dihydrouridine-synthase activity of URLC8, a polypeptide which is involved in the proliferation of lung cancer cells.

[0011] Accordingly, the present invention provides a method of diagnosing non-small cell lung cancer or a predisposition to developing non-small cell lung cancer in a subject, comprising determining a level of t-RNA dihydrouridine-synthase activity and/or t-RNA dihydrouridine in a biological sample derived from the subject, wherein an increase in said level as compared to a normal control level indicates that said subject suffers from or is at risk of developing non-small cell lung cancer.

[0012] The present invention also provides methods of predicting a lung squamous-cell carcinoma (SCC) prognosis. In some embodiments, the method comprises the steps of:

[0013] a. detecting a URLC8 expression level in a specimen collected from a subject whose SCC prognosis is to be predicted, and

[0014] b. indicating a poor prognosis when an elevated level of URLC8 expression is detected.

[0015] In a further embodiment, the present invention features a method of measuring t-RNA dihydrouridine-synthase activity by incubating a polypeptide under conditions suitable for synthesis of t-RNA dihydrouridine and detecting the t-RNA dihydrouridine synthesis level. The polypeptide is a URLC8 polypeptide or functional equivalent thereof. For example, the polypeptide may comprise the amino acid sequence of SEQ ID NO: 2. Alternatively, the polypeptide may comprise an amino acid sequence of SEQ ID NO: 2 where one or more amino acids are substituted, deleted, or inserted, so long as the resulting polypeptide retains the biological activity of the polypeptide of SEQ ID NO: 2. Biological activities of the polypeptide of SEQ ID No: 2 include, for example, the promotion of cell proliferation and the synthesis of t-RNA dihydrouridine. Additionally, the polypeptide may comprise a 493-amino acid protein encoded by the open reading frame of SEQ. ID. NO. 1, or a polynucleotide that hybridizes under stringent conditions, e.g., low or high, to the nucleotide sequence of SEQ ID NO: 1, so long as the resulting polynucleotide encodes a protein that retains the biological activity of the polypeptide of SEQ ID NO: 2. Examples of suitable low stringency conditions include, for example, 42.degree. C., 2.times.SSC, 0.1% SDS, or preferably 50.degree. C., 2.times.SSC, 0.1% SDS. Preferably, a high stringency condition is used. An example of a suitable high stringency condition includes, for example, washing 3 times in 2.times.SSC, 0.01% SDS at room temperature for 20 min, then washing 3 times in 1.times.SSC, 0.1% SDS at 37.degree. C. for 20 min, and washing twice in 1.times.SSC, 0.1% SDS at 50.degree. C. for 20 min. However, several factors, such as temperature and salt concentration, can influence the stringency of hybridization and one skilled in the art can suitably select the factors to achieve the requisite stringency.

[0016] In the context of the present invention, t-RNA dihydrouridine-synthase activity is defined as the catalysis of the reduction of the 5,6-double bond of a uridine residue on tRNA (FIG. 1c.). Synthesis of t-RNA dihydrouridine may be detected by conventional methods, such as those using a radioactive tRNA substrate or derivatives thereof. The substrate may be any compound capable of reduction of the 5,6-double bond of a uridine residue. An exemplary substrate is a tRNA having D-loop such as a tRNA.sup.Phe or other tRNA. The co-factor, e.g., the hydrogen donor, may be any compound capable of donating a hydrogen atom. For example, the co-factor may be a reduced form of nicotinamide adenine dinucleotide (NADH) or nicotinamide adenine dinucleotide phosphate (NADPH). Suitable conditions for synthesis include, for example, basic buffer conditions know in the art such as Tris-HCl.

[0017] The present invention further provides methods of identifying an agent that modulates (e.g., increases or decreases) t-RNA dihydrouridine-synthase activity by incubating a polypeptide under conditions suitable for synthesis of t-RNA dihydrouridine in the presence of a test agent and determining the t-RNA dihydrouridine synthesis level. A decrease in the level of synthesis as compared to a normal control synthesis level indicates that the test agent is an inhibitor of t-RNA dihydrouridine-synthase activity. Compounds that inhibit (e.g., decreases) t-RNA dihydrouridine-synthase activity are useful for treating, preventing or alleviating a symptom of lung cancer. For example, such compounds may inhibit the proliferation of lung cancer cells. Alternatively, an increase in the level or activity as compared to a normal control level indicates that the test agent is an enhancer of t-RNA dihydrouridine-synthase activity. Herein, the phrase normal control level refers to a level of t-RNA dihydrouridine synthesis detected in the absence of the test compound.

[0018] The present invention also encompasses compositions and methods for treating or preventing of lung cancer by contacting a lung cancer cell with a compound identified as described above. In a further embodiment, the present invention provides for the use of a compound identified as described above, for manufacturing a pharmaceutical composition suitable for treating or preventing lung cancer. For example, a method of treating lung cancer may involve the step of administering to a mammal, e.g. a human patient having been diagnosed with such a disease state, with a composition containing a pharmaceutically effective amount of a compound identified as described above and a pharmaceutical carrier.

[0019] The present invention also provides a kit for the detecting the t-RNA dihydrouridine-synthase activity of a compound with a t-RNA dihydrouridine-synthase polypeptide or living cell expressing the polypeptide and reagent for detecting a t-RNA dihydrouridine-synthase activity. The reagents are preferably packaged together in the form of a kit. The reagents may be packaged in separate containers and may include, for example, a t-RNA dihydrouridine-synthase polypeptide, reagent for detecting a t-RNA dihydrouridine-synthase activity, a control reagent (positive and/or negative), and/or a detectable label. Instructions (e.g., written, tape, VCR, CD-ROM, etc.) for carrying out the assay are preferably included in the kit. The assay format of the kit may comprise any t-RNA dihydrouridine-synthase assay known in the art.

[0020] In a further embodiment, the present method may include the step of administering to a subject a small interfering RNA (siRNA) composition. In the context of the present invention, the siRNA composition reduces the expression of URLC8.

[0021] These and other objects and features of the invention will become more fully apparent when the following detailed description is read in conjunction with the accompanying figures and examples. However, it is to be understood that both the foregoing summary of the invention and the following detailed description are of a preferred embodiment, and not restrictive of the invention or other alternate embodiments of the invention.

[0022] Unless otherwise defined, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. Although methods and materials similar or equivalent to those described herein can be used in the practice or testing of the present invention, suitable methods and materials are described below. In case of conflict, the present specification, including definitions, will control. In addition, the materials, methods, and examples are illustrative only and not intended to be limiting.

BRIEF DESCRIPTION OF THE DRAWINGS

[0023] FIG. 1 depicts the structure of IMS-E21 (URLC8) and structural features of tRNA-dihydrouridine.

[0024] a, Schematic structure of IMS-E21 (URLC8) protein. A member of UPF0034 (unclassified protein family 0034), Dus domain at N-terminal end, and DSRM (double-strand RNA binding motif) at C-terminal end were conserved in its protein structure, showing 30% homology to S. cerevisiae Dihydrouridine synthase 1 (DUS1), which catalyses the reduction of the 5,6-double bond of a uridine residue on tRNA. b, Alignment of the predicted amino acid sequences of the IMS-E21 (URLC8). Shading indicates homologues residues. c, The structure of dihydrouridine in tRNA. d, Two-dimensional representation of a generic tRNA with the D-loop nucleotides.

[0025] FIG. 2 depicts the expression of IMS-E21 (URLC8) in lung tumors and normal tissues.

[0026] a, Expression of IMS-E21 (URLC8) in clinical samples of NSCLC and corresponding normal lung tissues, examined by semi-quantitative RT-PCR. b, Expression of IMS-E21 (URLC8) in lung-cancer cell lines by semi-quantitative RT-PCR. c, Expression of IMS-E21 (URLC8) in human normal tissues, detected by multiple-tissue Northern-blot analysis.

[0027] FIG. 3 depicts the results of the immunohistochemical study of IMS-E21 (URLC8) expression in NSCLC and inhibition of growth of NSCLC cells by siRNA against IMS-E21 (URLC8).

[0028] a, Immunohistochemical evaluation of representative samples from surgically-resected NSCLC tissues using anti-IMS-E21 (URLC8) polyclonal antibody on tissue microarrays. b, Kaplan-Meier analysis of tumor specific survival in lung SCC patients according to IMS-E21 (URLC8) expression. c, mRNA knock down effect in response to si-IMS-E21 (URLC8) or control siRNAs in LC319 cells, (left panels) analyzed by semi-quantitative RT-PCR (upper panels) and endogenous IMS-E21 (URLC8) protein knock down effect by western blotting (middle panels), and (lower panels) colony-formation assays. (right panels) MTT assays of LC319 cells transfected with specific siRNAs or control plasmids (EGFP, Scramble, or Luciferase).

[0029] FIG. 4 depicts the characterization of IMS-E21 (URLC8) function in NSCLC cells.

[0030] a, Subcellular localization of endogenous IMS-E21 (URLC8) in LC319 cells; staining is visible mainly in cytoplasm (Rhodamine). Co-localization of myc-tagged-IMS-E21 (URLC8) (Rhodamine) and endogenous PDI (Protein Disulfide Isomerase), which was an abundant protein of endoplasmic reticulum (ER) (FITC) was observed. b, Identification of EPRS as an IMS-E21 (URLC8) interacting protein. Reciprocal co-immunoprecipitation of IMS-E21 (URLC8) and EPRS, both of which had been transfected into LC319 cell extracts. Western-blot analysis of the cell extracts immunoprecipitated with anti-FLAG M2 monoclonal antibodies, detected myc-tagged-IMS-E21 (URLC8) protein in the immunoprecipitate. Western-blot of extracts immunoprecipitated with anti-myc antibodies, detected FLAG-tagged EPRS protein in the immunoprecipitate. c, Co-localization of myc-tagged-IMS-E21 (URLC8) and FLAG-tagged EPRS in LC319 cells. d, Reduction of tRNA dihydrouridine content in A549 and LC319 cells treated with siRNAs against IMS-E21 (URLC8).

DETAILED DESCRIPTION OF THE INVENTION

Overview:

[0031] Although advances have been made in development of molecular-targeting drugs for cancer therapy, the ranges of tumor types that respond as well as the effectiveness of the treatments are still very limited (Ranson, M., et al. (2002) J Clin Oncol, 20: 2240-2250.; Blackledge, G. and Averbuch, S. (2004) Br J Cancer, 90: 566-572.). Hence, it is urgent to develop new anti-cancer agents highly specific to malignant cells with minimal or no adverse reactions. A powerful strategy toward these ends would combine screening of up-regulated genes in cancer cells on the basis of genetic information obtained on cDNA microarrays with high-throughput screening of their effect on cell growth, by inducing loss-of-function phenotypes with RNAi systems, and with validation of the potential drug targets by analyzing hundreds of clinical samples on tissue microarray (Sauter, G., et al. (2003) Nat Rev Drug Discov, 2: 962-972.; Kononen, J., et al. (1998) Nat Med, 4: 844-847.). By pursuing such a strategy, the present inventors have demonstrated herein that IMS-E21 (URLC8) is not only frequently co-over-expressed in clinical NSCLC samples and cell lines, but also that the high levels of expression of the gene products are indispensable for the disease progression as well as growth of NSCLC cells.

[0032] IMS-E21 (URLC8) is assigned to chromosome 16q22.2 and encodes a protein of 493 amino acids with 30% homology to S. cerevisiae Dihydrouridine synthase 1 (DUS1), a member of UPF0034 (unclassified protein family 0034), which catalyses the reduction of the 5,6-double bond of a uridine residue on tRNA (Xing, F., et al. (2002) RNA, 8: 370-381.), and with conserved double-strand RNA binding motif, DSRM. Several studies have revealed that tRNA modification enhances structural stabilization (Bjork, G. R., et al., (1987) Annu Rev Biochem, 56: 263-287.). In contrast, much less attention has been paid to modifications that potentially decrease regional stability and promote conformational flexibility of individual nucleotide residues. Dihydrouridine is the single most common form of post-translational modification in tRNA from bacteria and eukaryotes (Sprinzl, M., et al., (1998) Nucleic Acids Res, 26: 148-153.). The widespread presence of 5,6-dihydrouridine in the D-loop of tRNA has been known for decades, and some DUS enzymes have been identified in S. cerevisiae and E. coli, but the DUS enzyme with, double-strand RNA binding motif in mammalian cells has not been previously identified (Xing, F., et al., (2002) RNA, 8: 370-381.; Kuchino, Y. and Borek, E. (1978) Nature, 271: 126-129.).

[0033] With regards to dihydrouridine in tRNA in tumor cells, interestingly, an increase in the level of dihydrouridine was previously reported in tumor-specific tRNA.sup.Phe purified from human malignant tissues. The role of dihydrouridine in tRNA may increase conformational flexibility of the tRNA, but its precise function remains unclear. To elucidate the biological function of IMS-E21 (URLC8) and its contribution to lung carcinogenesis, the sub-cellular localization of IMS-E21 (URLC8) protein in LC319 cells was examined and found to be localized in ER. To clarify whether native IMS-E21 (URLC8) protein is required for tRNA-DUS activity in human NSCLC cells, IMS-E21 (URLC8)-siRNA vectors were transfected to A549 and LC319 cell lines in which the IMS-E21 (URLC8) gene is highly expressed. In those lung-cancer cells, endogenous IMS-E21 (URLC8) expression was suppressed significantly by siRNA, and tRNA-DUS activity was reduced, resulted in suppression of growth of the cancer cells. The results herein suggest that IMS-E21 (URLC8) is likely to play an important role in synthesis of tRNA-dihydrouridine of NSCLC cells, being possibly essential for translation processes and, when over-expressed, for growth and survival of NSCLC cells. The data herein strongly imply the possibility of designing new anti-cancer drugs specifically inhibit IMS-E21 (URLC8). Targeting the IMS-E21 (URLC8) enzyme activity and/or the IMS-E21 (URLC8)-tRNA synthetase complex may be a promising therapeutic and diagnostic strategy for treatment of lung-cancer patients.

URLC8 and Its Functional Equivalents and Uses Thereof:

[0034] Herein, the words "a", "an", and "the" as used herein mean "at least one" unless otherwise specifically indicated.

[0035] As noted above, the present invention is based in part on the discovery of a novel t-RNA dihydrouridine-synthase, URLC8, which is involved in the proliferation of lung cancer cells. The present invention is also based on the finding that a high expression level of URLC8 is associated with poor prognosis in lung squamous-cell carcinoma (SCC) patients. In view of the evidence provided herein, that URLC8 expression is associated with poor prognosis of cancer patients, the present invention thus provides methods for determining a prognosis for cancer patients. An example of such a method comprises the steps of:

[0036] a. detecting a URLC8 expression level in a specimen collected from a subject whose SCC prognosis is to be predicted, and

[0037] b. indicating a poor prognosis when an elevated level of URLC8 expression is detected.

[0038] In the context of the present invention, when the URLC8 expression level detected in a test specimen is higher than a control level, then the test specimen is deemed to have an elevated level of URLC8 expression. An example of a useful control level in the context of the present invention may comprise a standard value of URLC8 expression level taken from a group associated with good prognosis. The standard value may be obtained by any method known in the art. For example, a range of mean.+-.2 S.D. or mean.+-.3 S.D. may be used as the standard value. Alternatively, poor prognosis can be determined, when strong staining is observed by immunohistochemical analysis of sample tissue.

[0039] In the context of the present invention, an expression level of URLC8 may be detected by any one of the method selected from the group consisting of:

[0040] (a) detecting an mRNA encoding the amino acid sequence of SEQ ID NO: 2,

[0041] (b) detecting a protein comprising the amino acid sequence of SEQ ID NO: 2, and

[0042] (c) detecting the biological activity of a protein comprising the amino acid sequence of SEQ ID NO: 2.

[0043] In the context of the present invention, the mRNA, the `protein, or biological activity of the protein may be detected by any method. Methods for detecting a given protein, mRNA or biological activity thereof are well known to those skilled in the art. For example, mRNA may be detected using known PCR or hybridization based technologies. Alternatively, any immunoassay format may be applied for detection of a protein. Furthermore, the biological activity of URLC8, e.g. t-RNA dihydrouridine-synthase, or interactions between EPRS, may also detected using any suitable assay method, such as those described herein.

[0044] In the context of the present invention, determination of a poor prognosis may be used to determine further treatment, e.g., to stop further treatments that reduce quality of life, to treat the cancer in a different manner than previously used, or to treat the cancer more aggressively. In other words, the prediction of prognosis by URLC8 enables clinicians to choose in advance the most appropriate treatment for an individual SCC patient without even the information of conventional clinical staging of the disease, using only routine procedures for tissue-sampling.

[0045] Further, the methods of the present invention may be used to assess the efficacy of a course of treatment. For example, in a mammal with cancer from which a biological sample is found to have an elevated level of URLC8 expression, the efficacy of an anti-cancer treatment can be assessed by monitoring the URLC8 expression level over time. For example, a decrease in URLC8 expression level in a biological sample taken from a mammal following a course of treatment, as compared to a level observed in a sample taken from the mammal before treatment onset, or earlier in, the treatment, may be indicative of efficacious treatment.

[0046] As noted above, the present invention also provides kits for predicting lung squamous-cell carcinoma (SCC) prognosis, comprising any one component selected from the group consisting of:

[0047] (a) a reagent for detecting an mRNA encoding the amino acid sequence of SEQ ID NO: 2,

[0048] (b) a reagent for detecting a protein comprising the amino acid sequence of SEQ ID NO: 2, and

[0049] (c) a reagent for detecting a biological activity of the protein comprising the amino acid sequence of SEQ ID NO: 2.

[0050] URLC8 has t-RNA dihydrouridine-synthase activity, and its expression level is markedly elevated in lung cancer cells as compared to non-lung cancer tissues. Thus, URLC8-mediated t-RNA dihydrouridine-synthase activity is useful as a diagnostic parameter of lung cancer, e.g. non-small cell lung cancer. Accordingly, the present invention provides a method of diagnosing non-small cell lung cancer or a predisposition to developing non-small cell lung cancer in a subject, comprising the step of determining an level of t-RNA dihydrouridine-synthase activity and/or t-RNA dihydrouridine in a biological sample derived from the subject, wherein an increase in said level, as compared to a normal control level, indicates that said subject suffers from or is at risk of developing non-small cell lung cancer. Accumulation of t-RNA dihydrouridine in a cell reflects the presence of t-RNA dihydrouridine-synthase activity. Thus, the cellular t-RNA dihydrouridine-synthase activity can be evaluated through the determination of t-RNA dihydrouridine in a cell. The t-RNA dihydrouridine may be determined by any method known in the art. For example, t-RNA dihydrouridine can be determined through colorimetric assay using N-phenyl-p-phenylenediamine and 2,3-butadione monoxime (Jacobson, M. and Hedgcoth, C. (1970) Anal Biochem, 34: 459-469.). In the present invention any sample derived from a patient to be diagnosed may be used. An example of a preferred sample for use in the context of the present invention is a lung tissue obtained by biopsy or surgical-resection.

[0051] The present invention also provides a kit for detection of t-RNA dihydrouridine-synthase of URLC8. Examples of components of such kits include, t-RNA having D-loop as substrate, e.g., t-RNA.sup.Phe or other t-RNAs (excluding t-RNA.sup.Tyr and t-RNA.sup.Glu), an antibody that binds to URLC8, and a detectable label for detecting the antibody in a cell.

[0052] The URLC8 cDNA consists of 2,020 nucleotides that contain an open reading frame of 1,479 nucleotides as set forth in SEQ. ID. NO.: 1. The open reading frame encodes a 493-amino acid protein having amino acid sequence as set forth in SEQ. ID. NO.: 2. The amino acid sequence shows a 30% homology to S. cerevisiae Dus1 (dihydrouridine synthase 1), is member of UPF0034 (unclassified protein family 0034), and contains a conserved DSRM (double-strand RNA binding motif). S. cerevisiae Dus1 catalyses the reduction of the 5,6-double bond of a uridine residue on D-loop in tRNA (Xing, F., et al., (2002) RNA, 8: 370-381.). URLC8 localized mainly in cytoplasm and co-localized with ER-abundant protein PDI.

[0053] The present invention is also based on the finding that URLC8 has t-RNA dihydrouridine-synthase activity. To that end, one aspect of the invention involves identifying test compounds that regulate URLC8-mediated t-RNA dihydrouridine-synthase activity. Accordingly, the present invention provides novel methods for identifying compounds that slow or arrest the progression of, e.g., non-small cell lung cancer, by inhibiting URLC8-mediated t-RNA dihydrouridine-synthase activity.

[0054] The invention thus provides a method of screening for a compound that modulates URLC8 t-RNA dihydrouridine-synthase activity. The method is practiced by contacting a URLC8, or a functional equivalent thereof having t-RNA dihydrouridine-synthase activity, with one or more candidate compounds, and assaying t-RNA dihydrouridine-synthase activity of the contacted URLC8 or the functional equivalent. A compound that modulates t-RNA dihydrouridine-synthase activity of the URLC8 or functional equivalent is thereby identified.

[0055] In the context of the present invention, the term "functionally equivalent" means that the subject protein has t-RNA dihydrouridine-synthase activity. Whether or not a subject protein has the target activity can be determined in accordance with the present invention. For example, t-RNA dihydrouridine-synthase activity can be determined by incubating a polypeptide under conditions suitable for synthesis of t-RNA dihydrouridine and detecting the t-RNA dihydrouridine synthesis level.

[0056] Methods for preparing proteins functionally equivalent to a given protein are well known to those skilled in the art and include conventional methods of introducing mutations into the protein. For example, one skilled in the art can prepare proteins functionally equivalent to the human URLC8 protein by introducing an appropriate mutation in the amino acid sequence of the human URLC8 protein via site-directed mutagenesis (Hashimoto-Gotoh, T. et al. (1995), Gene 152, 271-275; Zoller, M J, and Smith, M. (1983), Methods Enzymol. 100, 468-500; Kramer, W. et al. (1984), Nucleic Acids Res. 12, 9441-9456; Kramer W, and Fritz H J. (1987) Methods. Enzymol. 154, 350-367; Kunkel, T A (1985), Proc. Natl. Acad. Sci. USA. 82, 488-492; Kunkel (1991), Methods Enzymol. 204, 125-139). Amino acid mutations can occur in nature, too. The proteins suitable for use in the context present invention include those proteins having the amino acid sequences of the human URLC8 protein in which one or more amino acids are mutated, provided the resulting mutated proteins are functionally equivalent to the human URLC8 protein. The number of amino acids to be mutated in such a mutant is generally 25 amino acids or less, preferably 10 to 15 amino acids or less, more preferably 5 to 6 amino acids or less, and even more preferably 2 to 3 amino acids or less. To maintain t-RNA dihydrouridine-synthase activity, it is preferable to conserve the DSRM (double-strand RNA binding motif) in the amino acid sequence of the mutated protein.

[0057] Mutated or modified proteins, proteins having amino acid sequences modified by deleting, adding and/or replacing one or more amino acid residues of a certain amino acid sequence, are known to retain the original biological activity (Mark, D. F. et al., Proc. Natl. Acad. Sci. USA (1984) 81, 5662-5666, Zoller, M. J. & Smith, M., Nucleic Acids Research (1982) 10, 6487-6500, Wang, A. et al., Science (1984) 224, 1431-1433, Dalbadie-McFarland, G. et al., Proc. Natl. Acad. Sci. USA (1982) 79, 6409-6413).

[0058] The amino acid residue to be mutated is preferably mutated into a different amino acid in which the properties of the amino acid side-chain are conserved (a process known in the art as "conservative amino acid substitution"). Examples of properties of amino acid side chains are hydrophobic amino acids (A, I, L, M, F, P, W, Y, V), hydrophilic amino acids (R, D, N, C, E, Q, G, H, K, S, T), and side chains having the following functional groups or characteristics in common: an aliphatic side-chain (G, A, V, L, I, P); a hydroxyl group containing side-chain (S, T, Y); a sulfur atom containing side-chain (C, M); a carboxylic acid and amide containing side-chain (D, N, E, Q); a base containing side-chain (R, K, H); and an aromatic containing side-chain (H, F, Y, W). Note, the parenthetic letters indicate the one-letter codes of amino acids.

[0059] An example of a protein to which one or more amino acids residues are added to the amino acid sequence of human URLC8 protein (SEQ ID NO: 2) is a fusion protein containing the human URLC8 protein. Fusion proteins suitable for use in the context of the present invention include, for example, fusions of the human URLC8 protein and other peptides or proteins. Fusion proteins can be made using techniques well known to those skilled in the art, for example by linking the DNA encoding the human URLC8 protein of the invention with DNA encoding other peptides or proteins, so that the frames match, inserting the fusion DNA into an expression vector and expressing it in a host. There is no restriction as to the peptides or proteins to be fused to the protein of the present invention.

[0060] Known peptides that can be used as peptides that are fused to the URLC8 protein include, for example, FLAG (Hopp, T. P. et al., (1988) Biotechnology 6, 1204-1210), 6.times. His containing six His (histidine) residues, 10.times. His, Influenza agglutinin (HA), human c-myc fragment, VSP-GP fragment, p18HIV fragment, T7-tag, HSV-tag, E-tag, SV4OT antigen fragment, lck tag, .alpha.-tubulin fragment, B-tag, Protein C fragment, and the like. Examples of proteins that may be fused to a protein of the invention include GST (glutathione-S-transferase), Influenza agglutinin (HA), immunoglobulin constant region, .beta.-galactosidase, MBP (maltose-binding protein), and such.

[0061] Fusion proteins can be prepared by fusing commercially available DNA, encoding the fusion peptides or proteins discussed above, with the DNA encoding a protein of the present invention and expressing the fused DNA prepared.

[0062] An alternative method known in the art to isolate functional equivalent proteins is, for example, the method using a hybridization technique (Sambrook, J. et al., (1989) Molecular Cloning 2nd ed. 9.47-9.58, Cold Spring Harbor Lab. Press). One skilled in the art can readily isolate a DNA having high homology with a whole or part of the URLC8 DNA sequence (e.g., SEQ ID NO: 1) encoding the human URLC8 protein, and isolate functional equivalent proteins to the human URLC8 protein from the isolated DNA. The proteins used for the present invention include those that are encoded by DNA that hybridize with a whole or part of the DNA sequence encoding the human URLC8 protein and are functional equivalent to the human URLC8 protein. These proteins include mammal homologues corresponding to the protein derived from human or rat (for example, a protein encoded by a monkey, mouse, rabbit and bovine gene). In isolating a cDNA highly homologous to the DNA encoding the human URLC8 protein from animals, it is particularly preferable to use tissues from testis or lung cancer.

[0063] The conditions of hybridization for isolating a DNA encoding a protein functionally equivalent to the human URLC8 protein can be routinely selected by a person skilled in the art. For example, hybridization may be performed by conducting pre-hybridization at 68.degree. C. for 30 min or longer using "Rapid-hyb buffer" (Amersham LIFE SCIENCE), adding a labeled probe, and warming at 68.degree. C. for 1 hour or longer. The following washing step can be conducted, for example, for a low stringency condition. Exemplary low stringency conditions include, for example, 42.degree. C., 2.times.SSC, 0.1% SDS, or preferably 50.degree. C., 2.times.SSC, 0.1% SDS. More preferably, high stringency conditions are selected. Exemplary high stringency conditions include, for example, washing 3 times in 2.times.SSC, 0.01% SDS at room temperature for 20 min, then washing 3 times in 1.times.SSC, 0.1% SDS at 37.degree. C. for 20 min, and washing twice in 1.times.SSC, 0.1% SDS at 50.degree. C. for 20 min. However, several factors, such as temperature and salt concentration, can influence the stringency of hybridization. Selection of the factors necessary to achieve a requisite level of stringency constitutes routine optimization that is well within the purview of one skilled in the art.

[0064] In place of hybridization, a gene amplification method, for example, the polymerase chain reaction (PCR) method, can be utilized to isolate a DNA encoding a protein functionally equivalent to the human URLC8 protein, using a primer synthesized based on the sequence information of the DNA (SEQ ID NO: 1) encoding the human URLC8 protein (SEQ ID NO: 2).

[0065] Proteins that are functionally equivalent to the human URLC8 protein, encoded by the DNA isolated through the above hybridization techniques or gene amplification techniques, normally have a high homology to the amino acid sequence of the human URLC8 protein. In the context of the present invention, the term "high homology" refers to a homology of 40% or higher, preferably 60% or higher, more preferably 80% or higher, even more preferably 95% or higher. The homology of a protein can be determined by following the algorithm in "Wilbur, W. J. and Lipman, D. J. (1983) Proc. Natl. Acad. Sci. USA 80, 726-730".

[0066] A protein useful in the context of the present invention may have variations in amino acid sequence, molecular weight, isoelectric point, the presence or absence of sugar chains, or form, depending on the cell or host used to produce it or the purification method utilized. Nevertheless, so long as it has a function equivalent to that of a human URLC8 protein (SEQ ID NO: 2), it is useful in the context of the present invention.

[0067] The proteins useful in the context of the present invention can be prepared as recombinant proteins or natural proteins, by methods well known to those skilled in the art. A recombinant protein can be prepared, for example, by inserting a DNA encoding a protein of the present invention (for example, the DNA comprising the nucleotide sequence of SEQ ID NO: 1) into an appropriate expression vector, introducing the vector into an appropriate host cell, obtaining the extract, and purifying the protein by subjecting the extract to chromatography, for example, ion exchange chromatography, reverse phase chromatography, gel filtration, or affinity chromatography utilizing a column to which antibodies against the protein of the present invention are fixed, or by combining more than one of aforementioned columns.

[0068] In addition, when a protein useful in the context of the present invention is expressed within host cells (for example, animal cells and E. coli) as a fusion protein with glutathione-S-transferase protein or as a recombinant protein supplemented with multiple histidines, the expressed recombinant protein can be purified using a glutathione column or nickel column.

[0069] After purifying the fusion protein, it is also possible to exclude regions, other than the objective protein, by cutting with thrombin or factor-Xa as required.

[0070] A natural protein can be isolated by methods known to those skilled in the art, for example, by contacting an affinity column, in which antibodies binding to the URLC8 protein described below are bound, with the extract of tissues or cells expressing a protein of the present invention. The antibodies can be polyclonal antibodies or monoclonal antibodies.

[0071] In the present invention, t-RNA dihydrouridine-synthase activity of URLC8 or its functional equivalent can be determined by methods known in the art. For example, URLC8 and a substrate, e.g. tRNA having D-loop can be incubated with a hydrogen donor, under suitable assay conditions for t-RNA dihydrouridine synthesis. In the present invention, exemplary conditions for the synthesis of t-RNA dihydrouridine include the steps of contacting the substrate, hydrogen donor, with t-RNA dihydrouridine-synthase or sample, and incubating them. A tRNA.sup.Phe and reduced form of nicotinamide adenine dinucleotide (NADH) or nicotinamide adenine dinucleotide phosphate (NADPH) are examples of suitable substrates and hydrogen donors, respectively. Preferably, the substrate or hydrogen atom of the donor is labeled for tracing t-RNA dihydrouridine. Specifically, in the context of the present invention, a radio labeled tRNA is the preferred substrate. The reduced radio-labeled-tRNA (t-RNA dihydrouridine) may be detected by any suitable method. Increase of molecular mass of tRNA by addition of hydrogen atoms can be detected, for example by suitable chromatography, e.g.

[0072] thin layer chromatography. For example, suitable conditions for t-RNA dihydrouridine synthesis are set forth below:

[0073] Reaction mixture:

[0074] 100 mM Tris-HCl (pH 8.0),

[0075] 100 mM ammonium acetate,

[0076] 5 mM MgCl.sub.2,

[0077] 2 mM DTT,

[0078] 0.1 mM EDTA,

[0079] 1 mM NADPH,

[0080] 1 mM NADH, and

[0081] 50,000 cpm of labeled transcript of tRNA (6 fmol)

[0082] The reaction mixture is mixed with a sample containing t-RNA dihydrouridine-synthase to be determined, and incubated for 30 min. at 30`0 C. In titration assays, extracts and purified proteins are preferably diluted in buffer containing 50 mM Tris-HCl, pH 8.0, 250 .mu.g/mL bovine serum albumin, and 2 mM DTT. In some assays, 250 .mu.M of flavin adenine dinucleotide (FAD) may also be included. Following the incubation, RNA is extracted from the mixture, and treated with P1 nuclease. Then nucleotides are resolved by thin layer chromatography using either cellulose plates developed in one dimension with solvent containing ammonium sulfate (74 g/100 mL H.sub.2O, pH 3.5):H.sub.2O:isopropanol (80:18:2, v/v/v), or using PEI-cellulose plates developed in two dimensions with 1 M acetic acid, pH 3.5, for the first dimension, and in buffer containing 74 g ammonium sulfate/100 mL H.sub.2O (adjusted to pH 3.5 with H.sub.2SO.sub.4) for the second dimension (Bochner & Ames, (1982) J Biol Chem 257:9759-9769). In titration assays, 1 U of activity is defined as the amount of protein required to convert half of the tRNA.

[0083] Alternatively, t-RNA dihydrouridine-synthase activity of URLC8 can be estimated based on t-RNA dihydrouridine accumulation. t-RNA dihydrouridine can be detected by a colorimetric method. Furthermore, t-RNA dihydrouridine-synthase activity of URLC8 can be determined by consumed NADH and/or NADPH during the reaction. Suitable methods for determination of NADH and/or NADPH are well known to a person skilled in the art. Alternatively, following the reaction, either or both of tRNA and reduced tRNA can be detected with mass spectrometry, e.g. MALDI-TOF-MS.

[0084] Various low-throughput and high-throughput enzyme assay formats are known in the art and can be readily adapted for detection or measuring of the t-RNA dihydrouridine-synthase activity of URLC8. For high-throughput assays, the tRNA substrate is preferably immobilized on a solid support, such as a multi-well plate, slide or chip. Following the reaction, the reduced product (t-RNA dihydrouridine) can be detected on the solid support. Alternatively, the t-RNA dihydrouridine-synthase reaction can take place in solution, after which the t-RNA dihydrouridine can be immobilized on a solid support, and detected. In order to detect the reduction of tRNA, for example, H.sup.3 labeled NADH and/or NADPH may be used as a hydrogen donor. The reduced product (t-RNA dihydrouridine) can be traced with radioactive H.sup.3. To facilitate such assays, the solid support may be coated with streptavidin and the t-RNA labeled with biotin. The skilled person can determine suitable assay formats depending on the desired throughput capacity of the screen.

[0085] Any test compound, including, but not limited to, cell extracts, cell culture supernatant, products of fermenting microorganisms, extracts from marine organisms, plant extracts, purified or crude proteins, peptides, non-peptide compounds, synthetic micromolecular compounds and natural compounds, can be used in the screening methods of the present invention. The test compound of the present invention can be also obtained using any of the numerous approaches in combinatorial library methods known in the art, including (1) biological libraries, (2) spatially addressable parallel solid phase or solution phase libraries, (3) synthetic library methods requiring deconvolution, (4) the "one-bead, one-compound" library method and (5) synthetic library methods using affinity chromatography selection. The biological library methods using affinity chromatography selection are limited to peptide libraries, while the other four approaches are applicable to peptide, non-peptide oligomer or small molecule libraries of compounds (Lam (1997) Anticancer Drug Des. 12: 145). Examples of methods for the synthesis of molecular libraries can be found in the art (DeWitt et al. (1993) Proc. Natl. Acad. Sci. USA 90: 6909; Erb et al. (1994) Proc. Natl. Acad. Sci. USA 91: 11422; Zuckermann et al. (1994) J. Med. Chem. 37: 2678; Cho et al. (1993) Science 261: 1303; Carell et al. (1994) Angew. Chem. Int. Ed. Engl. 33: 2059; Carell et al. (1994) Angew. Chem. Int. Ed. Engl. 33: 2061; Gallop et al. (1994) J. Med. Chem. 37: 1233). Libraries of compounds may be presented in solution (see Houghten (1992) Bio/Techniques 13: 412) or on beads (Lam (1991) Nature 354: 82), chips (Fodor (1993) Nature 364: 555), bacteria (US Pat. No. 5,223,409), spores (U.S. Pat. No. 5,571,698; 5,403,484, and 5,223,409), plasmids (Cull et al. (1992) Proc. Natl. Acad. Sci. USA 89: 1865) or phage (Scott and Smith (1990) Science 249: 386; Devlin (1990) Science 249: 404; Cwirla et al. (1990) Proc. Natl. Acad. Sci. USA 87: 6378; Felici (1991) J. Mol. Biol. 222: 301; US Pat. Application 2002103360).

[0086] A compound isolated by the screening method of the present invention is a candidate for drugs that inhibit the t-RNA dihydrouridine-synthase activity of URLC8 and can be applied to the treatment or prevention of NSCLC.

[0087] Moreover, a compound in which a part of the structure of the compound inhibiting the t-RNA dihydrouridine-synthase activity of URLC8 is converted by addition, deletion and/or replacement are also included in the compounds obtainable by the screening method of the present invention.

Treating and Preventing Lung Cancer:

[0088] The present invention provides compositions for treating or preventing NSCLC comprising any of the compounds selected by the screening methods of the present invention.

[0089] When administrating a compound isolated by a method of the present invention as a pharmaceutical for humans and other mammals, such as mice, rats, guinea-pigs, rabbits, cats, dogs, sheep, pigs, cattle, monkeys, baboons, and chimpanzees, the isolated compound can be directly administered or, alternatively, can be formulated into a dosage form using conventional pharmaceutical preparation methods. For example, according to the need, the drugs can be taken orally, as sugar-coated tablets, capsules, elixirs and microcapsules, or non-orally, in the form of injections of sterile solutions or suspensions with water or any other pharmaceutically acceptable liquid. For example, the compound can be mixed with pharmaceutically acceptable carriers or media, specifically, sterilized water, physiological saline, plant-oils, emulsifiers, suspending agents, surfactants, stabilizers, flavoring agents, excipients, vehicles, preservatives, binders, and such, in a unit dose form required for generally accepted drug implementation. The amount of active ingredients in these preparations makes a suitable dosage within the indicated range acquirable.

[0090] Examples of additives that can be mixed to form tablets and capsules include, for example, binders, such as gelatin, corn starch, tragacanth gum and arabic gum; excipients, such as crystalline cellulose; swelling agents, such as corn starch, gelatin and alginic acid; lubricants, such as magnesium stearate; sweeteners, such as sucrose, lactose or saccharin; and flavoring agents, such as peppermint, Gaultheria adenothrix oil and cherry. When the unit-dose form is a capsule, a liquid carrier, such as an oil, can also be further included in the above ingredients. Sterile composites for injections can be formulated following normal drug implementations using vehicles such as distilled water used for injections.

[0091] Physiological saline, glucose, and other isotonic liquids, including adjuvants, such as D-sorbitol, D-mannose, D-mannitol, and sodium chloride, can be used as aqueous solutions for injections. These can be used in conjunction with suitable solubilizers, such as alcohol, specifically ethanol, polyalcohols such as propylene glycol and polyethylene glycol, non-ionic surfactants, such as Polysorbate 80.TM. and HCO-50.

[0092] Sesame oil and soy-bean oil are examples of suitable oleaginous liquids and may be used in conjunction with benzyl benzoate or benzyl alcohol as solubilizers. They may be further formulated with a buffer, such as phosphate buffer or sodium acetate buffer; a pain-killer, such as procaine hydrochloride; a stabilizer, such as benzyl alcohol or phenol; and an anti-oxidant. The prepared injection may be filled into a suitable ampule.

[0093] Methods well known to those skilled in the art may be used to administer a pharmaceutical composition of the present invention to patients, for example, as intra-arterial, intravenous, or percutaneous injections and also as intranasal, transbronchial, intramuscular or oral administrations. The dosage and method of administration may vary according to the body-weight and age of the patient and the selected administration method; however, one skilled in the art can routinely select a suitable method of administration and dosage. If said compound is encodable by a DNA, the DNA can be inserted into a vector for gene therapy and the vector can be administered to a patient to perform the therapy. The dosage and method of administration may again vary according to the body-weight, age, and symptoms of the patient; however, one skilled in the art can suitably select them.

[0094] For example, although the dose of a compound that binds to URLC8 and regulates its activity depends on the symptoms, a suitable dose is generally about 0.1 mg to about 100 mg per day, preferably about 1.0 mg to about 50 mg per day and more preferably about 1.0 mg to about 20 mg per day, when administered orally to a normal adult (weight 60 kg).

[0095] When administering parenterally, in the form of an injection to a normal adult (weight 60 kg), although there are some differences according to the patient, target organ, symptoms and method of administration, it is convenient to intravenously inject a dose of about 0.01 mg to about 30 mg per day, preferably about 0.1 to about 20 mg per day and more preferably about 0.1 to about 10 mg per day. Also, in the case of other animals too, it is possible to administer an amount converted to 60 kg of body-weight.

[0096] The present invention further provides a method for treating a NSCLC in a subject. Administration can be prophylactic or therapeutic to a subject at risk of (or susceptible to) a disorder or having a disorder associated with aberrant the t-RNA dihydrouridine-synthase activity of URLC8. The method includes decreasing the function of URLC8 in a non-small cell lung cancer (NSCLC) cell. Function can be inhibited through the administration of a compound obtained by the screening method of the present invention.

[0097] Also, an siRNA against a URLC8 gene can be used to reduce the expression level. Herein, term "siRNA" refers to a double stranded RNA molecule that prevents translation of a target mRNA. Standard techniques for introducing siRNA into a cell are used, including those in which DNA is a template from which siRNA is transcribed. In the context of the present invention, the siRNA comprises a sense nucleic acid sequence and an anti-sense nucleic acid sequence against URLC8. The siRNA method of the present invention can be used to alter the expression in a cell of an up-regulated NSCLC gene, e.g., up-regulation resulting from the malignant transformation of the cells. Binding of an siRNA to a transcript corresponding to URLC8 in a target cell results in a reduction in the protein production by the cell. The length of the oligonucleotide is preferably at least 10 nucleotides and may be as long as the naturally-occurring the transcript. Preferably, the oligonucleotide is about 19-25 nucleotides in length. More preferably, the oligonucleotide is less than 75, less than 50, or less than 25 nucleotides in length. Examples of URLC8 siRNA oligonucleotides which inhibited the expression in A549 and LC319 cells include the target sequence containing SEQ ID NO: 11.

[0098] An siRNA of the present invention can be constructed such that a single transcript has both the sense and complementary antisense sequences from the target gene, e.g., as a hairpin.

[0099] An siRNA of URLC8 hybridizes to a target mRNA and thereby decreases or inhibits the production of URLC8 polypeptides by associating with the normally single-stranded mRNA transcript, thereby interfering with translation and thus, expression of the protein. In order to enhance the inhibition activity of an siRNA, nucleotide "u" can be added to 3'end of the antisense strand of the target sequence. The number of "u"s to be added is at least 2, generally 2 to 10, preferably 2 to 5. The added "u''s form single strand at the 3'end of the antisense strand of the siRNA.

[0100] An siRNA of URLC8 can be directly introduced into the cells in a form that is capable of binding to the mRNA transcripts. Alternatively, a DNA encoding the siRNA may be carried in a vector.

[0101] Vectors may be produced, for example, by cloning an URLC8 gene target sequence into an expression vector having operatively-linked regulatory sequences flanking the sequence in a manner that allows for expression (by transcription of the DNA molecule) of both strands (Lee, N. S., et al., (2002) Nature Biotechnology 20 : 500-505.). An RNA molecule that is antisense to mRNA of URLC8 is transcribed by a first promoter (e.g., a promoter sequence 3' of the cloned DNA) and an RNA molecule that is the sense strand for the mRNA of URLC8 gene is transcribed by a second promoter (e.g., a promoter sequence 5' of the cloned DNA). The sense and antisense strands hybridize in vivo to generate siRNA constructs for silencing of the URLC8 gene. Alternatively, the two constructs can be utilized to create the sense and anti-sense strands of an siRNA construct. Cloned URLC8 gene can encode a construct having secondary structure, e.g., hairpins, wherein a single transcript has both the sense and complementary antisense sequences from the target gene.

[0102] A loop sequence consisting of an arbitrary nucleotide sequence can be located between the sense and antisense sequence in order to form the hairpin loop structure. Thus, the present invention also provides siRNA having the general formula 5'-[A]-[B]-[A']-3', wherein [A] is a ribonucleotide sequence corresponding to a sequence selected from the group consisting of nucleotides of SEQ ID NO: 11.

[0103] [B] is a ribonucleotide sequence consisting of 3 to 23 nucleotides, and

[0104] [A'] is a ribonucleotide sequence consisting of the complementary sequence of [A]. The region [A] hybridizes to [A'], and then a loop consisting of region [B] is formed. The loop sequence may be preferably 3 to 23 nucleotide in length. The loop sequence, for example, can be selected from group consisting of following sequences (http://www.ambion.com/techlibitb/tb.sub.--506.html). Furthermore, loop sequence consisting of 23 nucleotides also provides active siRNA (Jacque, J. M., et al., (2002) Nature 418: 435-438.).

[0105] CCC, CCACC or CCACACC: Jacque, J. M, et al., (2002) Nature 418: 435-438.

[0106] UUCG: Lee, N. S., et al., (2002) Nature Biotechnology 20: 500-505. Fruscoloni, P., et al., (2003) Proc. Natl. Acad. Sci. USA 100: 1639-1644.

[0107] UUCAAGAGA: Dykxhoorn, D. M., (2003) Nature Reviews Molecular Cell Biology 4: 457-467.

[0108] Examples of preferred siRNAs having hairpin loop structure of the present invention are shown below. In the following structure, the loop sequence can be selected from group consisting of, CCC, UUCG, CCACC, CCACACC, and UUCAAGAGA. The preferred loop sequence is UUCAAGAGA ("ttcaagaga" in DNA). Exemplary hairpin siRNA suitable for use in the context of the present invention include:

TABLE-US-00001 ugaggugcucagcacagug-[b]-cacugugcugagcaccuca (for target sequence of SEQ ID NO: 10) guuggcacagccuguguau-[b]-auacacaggcugugccaac (for target sequence of SEQ ID NO: 11)

[0109] The regulatory sequences flanking the URLC8 genes can be identical or different, such that their expression can be modulated independently, or in a temporal or spatial manner. siRNAs are transcribed intracellularly by cloning the URLC8 gene template into a vector containing, e.g., a RNA polymerase III transcription unit from the small nuclear RNA (snRNA) U6 or the human III RNA promoter. For introducing the vector into the cell, transfection-enhancing agent can be used. FuGENE6 (Roche diagnostics), Lipofectamine 2000 (Invitrogen), Oligofectamine (Invitrogen), and Nucleofector (Wako pure Chemical) are useful as the transfection-enhancing agent.

[0110] The siRNA of the present invention inhibits the expression of a polypeptide of the invention and is thereby useful for suppressing the biological activity of the polypeptide of the invention. Also, expression-inhibitors, comprising siRNA of the present invention, are useful in that they can inhibit the biological activity of a polypeptide of the invention. Therefore, a composition comprising an antisense oligonucleotide of the present invention, such as an siRNA, is useful in treating NSCLC.

[0111] Thus, the present invention provides a composition for treating or preventing non-small cell lung cancer (NSCLC), comprising a pharmaceutically effective amount of small interfering RNA (siRNA) against a URLC8 gene, wherein said small interfering RNA comprises the nucleotide sequence 5'-TGAGGTGCTCAGCACAGTG-3' (SEQ ID NO.10) and 5'-GTTGGCACAGCCTGTGTAT-3' (SEQ ID NO.11) as the target sequence. Alternatively, the present invention further provides a method for treating or preventing NSCLC, said method comprising the step of administering a pharmaceutically effective amount of the small interfering RNA.

[0112] Patients with tumors characterized as over-expressing URLC8 may be treated by administering URLC8-siRNA. siRNA therapy is used to inhibit expression of URLC8 in patients suffering from or at risk of developing non-small cell lung cancer (NSCLC). Such patients are identified by standard methods of the particular tumor type; for example, non-small cell lung cancer (NSCLC) is typically diagnosed by tomography, ultrasound or biopsy.

[0113] Treatment is efficacious if the treatment leads to a clinical benefit, such as a reduction in the expression of URLC8, or a decrease in size, prevalence, or metastatic potential of the tumor in the subject. When treatment is applied prophylactically, "efficacious" means that the treatment retards or prevents tumors from forming or prevents or alleviates a clinical symptom of the tumor. Efficaciousness may be determined in association with any known method for diagnosing or treating the particular tumor type.

[0114] siRNA therapy is carried out by administering to a patient an siRNA using standard vectors and/or gene delivery systems. Suitable gene delivery systems may include liposomes, receptor-mediated delivery systems, and viral vectors, such as herpes viruses, retroviruses, adenoviruses and adeno-associated viruses, among others. A reduction in URLC8 production results in a decrease in URLC8 protein expression. A therapeutic nucleic acid composition is preferably formulated with a pharmaceutically acceptable carrier. The therapeutic composition may also include a gene delivery system as described above. Pharmaceutically acceptable carriers are biologically compatible vehicles which are suitable for administration to an animal, e.g., physiological saline. A therapeutically effective amount of a compound is an amount which is capable of producing a medically desirable result, such as reduced production of a URLC8 gene product, reduction of cell growth, e.g., proliferation, or reduction in tumor growth in a treated animal.

[0115] Parenteral administration, such as intravenous, subcutaneous, intramuscular, and intraperitoneal delivery routes, may be used to deliver URLC8-siRNA compositions.

[0116] Dosages for any one patient depends upon many factors, including the patient's size, body surface area, age, the particular nucleic acid to be administered, sex, time and route of administration, general health, and other drugs being administered concurrently. Suitable dosage for intravenous administration of nucleic acids ranges from approximately 10.sup.6 to 10.sup.22 copies of the polynucleotide.

[0117] The polynucleotides of the present invention may be administered by standard methods, such as by injection into the interstitial space of tissues such as muscles or skin, introduction into the circulation or into body cavities or by inhalation or insufflation. Polynucleotides are preferably injected or otherwise delivered to the animal in combination with a pharmaceutically acceptable liquid carrier, e.g., a liquid carrier, which is aqueous or partly aqueous. The polynucleotides of the present invention may also beassociated with a liposome (e.g., a cationic or anionic liposome). The polynucleotides of the present invention preferably include the genetic information necessary for expression by a target cell, such as promoters.

[0118] In another aspect, the present invention includes pharmaceutical, or therapeutic, compositions containing one or more therapeutic compounds described herein. Pharmaceutical formulations may include those suitable for oral, rectal, nasal, topical (including buccal and sub-lingual), vaginal or parenteral (including intramuscular, subcutaneous and intravenous) administration, or for administration by inhalation or insufflation. The formulations may, where appropriate, be conveniently presented in discrete dosage units and may be prepared by any of the methods conventional in the art of pharmacy. All such pharmacy methods include the steps of bringing into association the active compound with liquid carriers or finely divided solid carriers or both as needed and then, if necessary, shaping the product into the desired formulation.

[0119] Pharmaceutical formulations suitable for oral administration may conveniently be presented as discrete units, such as capsules, cachets or tablets, each containing a predetermined amount of the active ingredient; as a powder or granules; or as a solution, a suspension or as an emulsion. The active ingredient may also be presented as a bolus electuary or paste, and be in a pure form, i.e., without a carrier. Tablets and capsules for oral administration may contain conventional excipients, such as binding agents, fillers, lubricants, disintegrant or wetting agents. A tablet may be made by compression or molding, optionally with one or more formulational ingredients. Compressed tablets may be prepared by compressing in a suitable machine the active ingredients in a free-flowing form, such as a powder or granules, optionally mixed with a binder, lubricant, inert diluent, lubricating, surface active or dispersing agent. Molded tablets may be made by molding in a suitable machine a mixture of the powdered compound moistened with an inert liquid diluent. The tablets may be coated according to methods well known in the art. Oral fluid preparations may be in the form of, for example, aqueous or oily suspensions, solutions, emulsions, syrups or elixirs, or may be presented as a dry product for constitution with water or other suitable vehicle before use. Such liquid preparations may contain conventional additives, .such as suspending agents, emulsifying agents, non-aqueous vehicles (which may include edible oils), or preservatives. Furthermore, the tablets may optionally be formulated so as to provide slow or controlled release of the active ingredient therein.

[0120] Formulations for parenteral administration include aqueous and non-aqueous sterile injection solutions which may contain anti-oxidants, buffers, bacteriostats and solutes which render the formulation isotonic with the blood of the intended recipient; and aqueous and non-aqueous sterile suspensions which may include suspending agents and thickening agents. The formulations may be presented in unit dose or multi-dose containers, for example, sealed ampoules and vials, and may be stored in a freeze-dried (lyophilized) condition requiring only the addition of the sterile liquid carrier, for example, saline, water-for-injection, immediately prior to use. Alternatively, the formulations may be presented for continuous infusion. Extemporaneous injection solutions and suspensions may be prepared from sterile powders, granules and tablets of the kind previously described.

[0121] Formulations for rectal administration may be presented as a suppository with the usual carriers, such as cocoa butter or polyethylene glycol. Formulations for topical administration in the mouth, for example, buccally or sublingually, include lozenges, comprising the active ingredient in a flavored base, such as sucrose and acacia or tragacanth, and pastilles comprising the active ingredient in a base, such as gelatin and glycerin or sucrose and acacia. For intra-nasal administration, the compounds of the present invention may be used as a liquid spray or dispersible powder or in the form of drops. Drops may be formulated with an aqueous or non-aqueous base also comprising one or more dispersing agents, solubilizing agents or suspending agents. Liquid sprays are conveniently delivered from pressurized packs.

[0122] For administration by inhalation, the compounds of the present invention are conveniently delivered from an insufflator, nebulizer, pressurized pack or other convenient aerosol spray delivery means. Pressurized packs may comprise a suitable propellant, such as dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas. In the case of a pressurized aerosol, the dosage unit may be determined by providing a valve to deliver a metered amount.

[0123] Alternatively, for administration by inhalation or insufflation, the compounds of the present invention may take the form of a dry powder composition, for example a powder mix of the compound and a suitable powder base, such as lactose or starch. The powder composition may be presented in a unit dosage form, in for example, capsules, cartridges, gelatin or blister packs from which the powder may be administered with the aid of an inhalator or insufflators.

[0124] When desired, the above-described formulations, adapted to give sustained release of the active ingredient, may be employed. The pharmaceutical compositions of the present invention may also contain other active ingredients, such as antimicrobial agents, immunosuppressants or preservatives.

[0125] It should be understood that in addition to the ingredients particularly mentioned above, the formulations of the present invention may include other agents conventional in the art, having regard to the type of formulation in question; for example, those suitable for oral administration may include flavoring agents.

[0126] Preferred unit dosage formulations are those containing an effective dose, as recited below, or an appropriate fraction thereof, of the active ingredient.

[0127] For each of the aforementioned conditions, the compositions may be administered orally or via injection at a dose ranging from about 0.1 to about 250 mg/kg per day. The dose range for adult humans is generally from about 5 mg to about 17.5 g/day, preferably about 5 mg to about 10 g/day, and most preferably about 100 mg to about 3 g/day. Tablets or other unit dosage forms of presentation provided in discrete units may conveniently contain an amount which is effective at such dosage or as a multiple of the same, for instance, units containing about 5 mg to about 500 mg, usually from about 100 mg to about 500 mg.

[0128] The pharmaceutical composition preferably is administered orally or by injection (intravenous or subcutaneous), and the precise amount administered to a subject will be the responsibility of the attendant physician. However, the dose employed will depend upon a number of factors, including the age and sex of the subject, the precise disorder being treated, and its severity. In addition, the route of administration may vary depending upon the condition and its severity.

Examples

Example 1

Materials and Methods

(a) Lung Cancer Cell Lines and Tissue Samples.

[0129] The 19 human NSCLC and 4 SCLC cell lines used in this study were as follows: lung ADC A427, A549, LC319, PC3, PC9, PC14, and NCI-H1373; bronchioloalveolar cell carcinomas (BAC) NCI-H1666 and NCI-H1781; lung adenosquamous carcinoma (ASC) NCI-H226 and NCI-H647; lung SCC RERF-LC-AI, SK-MES-1, EBC-1, LU61, NCI-H520, NCI-H1703, and NCI-H2170; a lung large-cell carcinoma (LCC) LX1; and SCLC DMS114, DMS273, SBC-3, and SBC-5. All cells were grown in monolayers in appropriate medium supplemented with 10% fetal calf serum (FCS) and were maintained at 37.degree. C. in an atmosphere of humidified air with 5% CO2. Human small airway epithelial cells, SAEC were grown in optimized medium (SAGM) purchased from Cambrex Bio Science Inc. (Walkersville, Md.).

[0130] Primary NSCLC samples, of which 22 were classified as ADCs, 14 as SCCs, and one as ASC, were originally obtained from 37 patients, with written informed consent, for a study described elsewhere (Kikuchi, T. et al. (2003) Oncogene, 22: 2192-2205.). An independent set of fourteen additional primary NSCLCs, including seven ADCs and seven SCCs, were obtained along with adjacent normal lung tissue samples from patients undergoing surgery.

[0131] A total of 292 NSCLC and adjacent normal lung tissue samples used for immunostaining on tissue microarray and additional statistical analysis were also obtained from patients who underwent surgery.

(b) Semi-Quantitative RT-PCR Analysis.

[0132] Total RNA was extracted from cultured cells and clinical tissues using the Trizol reagent (Life Technologies, Inc.) according to the manufacturer's protocol. Extracted RNAs and normal human tissue poly(A) RNAs were treated with DNase I (Nippon Gene) and were reverse-transcribed using oligo (dT) primer and SuperScript II reverse transcriptase (Invitrogen). Semi-quantitative RT-PCR experiments were carried out with the following synthesized gene-specific primers or with ACTB-specific primers as an internal control:

(c) IMS-E21 (URLC8).

[0133] The following primers were used to isolated IMS-E21 (URLC8):

TABLE-US-00002 5'-GACCACATCCAACAGTATTCG-3' (SEQ ID NO. 3) and 5'-TGCCAGGACATCTAACTTCTG-3'; (SEQ ID NO. 4) ACTB, 5'-GAGGTGATAGCATTGCTTTCG-3' (SEQ ID NO. 5) and 5'-CAAGTCAGTGTACAGGTAAGC-3'. (SEQ ID NO. 6)

[0134] PCR reactions were optimized for the number of cycles to ensure product intensity within the logarithmic phase of amplification.

(d) Northern-Blot Analysis.

[0135] Human multiple-tissue blots (BD Biosciences Clontech) were hybridized with a .sup.32P-labeled PCR product of IMS-E21 (URLC8). The cDNA probes of IMS-E21 (URLC8) were prepared by RT-PCR using primers with the same above. Pre-hybridization, hybridization, and washing were performed according to the supplier's recommendations. The blots were autoradiographed with intensifying BAS screens (BIO-RAD, Hercules, Calif.) at room temperature for 30 hours.

(e) Cloning of a Full-Length IMS-E21 (URLC8) cDNA and DNA Sequencing.

[0136] Firstly, the IMS-E21 (URLC8) sequence was searched against the EST database by BLAST program and found to be highly homologous to an EST clone (FLJ20399, NM.sub.--017803). cDNA was generated from total RNA derived from 12 normal organ mixed sample by random priming using SuperScript II reverse transcriptase (Invitrogen) according to the manufacturer's instructions. RT-PCR product was cloned into the pcDNA3.1-myc-His vector (Invitrogen) and confirmed by sequencing.

(f) Generation of Anti-IMS-E21 (URLC8) Antibodies.

[0137] Plasmids expressing IMS-E21 (URLC8) (full length) that contain His-tagged epitope at the NH.sub.2-terminal of the individual proteins were prepared using pET28 vector (Novagen, Madison, Wis.). The recombinant protein was expressed in Escherichia coli, BL21 codon-plus strain (Stratagene, LaJolla, Calif.), and purified using TALON resin (BD Bioscience) according to the supplier's protocol. The protein extracted in SDS-PAGE gel was inoculated into rabbits, and the immune sera were purified on affinity columns according to the standard methodology. Affinity-purified anti-IMS-E21 (URLC8) antibodies were used for western-blot analysis, immunoprecipitation, and immunostaining.

(g) Co-Immunoprecipitation and MALDI-TOF-MS Mapping of IMS-E21 (URLC8)-Associated Proteins.

[0138] Cell extracts from lung-cancer cell line LC319 transfected with the plasmids expressing N-terminal FLAG-tagged and C-terminal HA-tagged-pCAGGS-IMS-E21 (URLC8) vector or mock vector (control) were pre-cleared by incubation at 4.degree. C. for 1 hour with 100 .mu.L of protein G-agarose beads in a final volume of 2 ml of IP-buffer (0.5% NP-40, 50 mM Tris-HCl, 150 mM NaCl) in the presence of proteinase inhibitor. After centrifugation at 1000 rpm for 5 min at 4.degree. C., the supernatant was incubated at 4.degree. C. with anti-Flag M2 agarose for 2 hours. The beads were then collected by centrifugation at 5000 rpm for 2 min and washed six times with 1 ml of each immunoprecipitation buffer. Then, the washed beads were eluted by Flag-peptide (Sigma-Aldrich Co., St. Louis, Mo.). The elution was incubated at 4.degree. C. with anti-HA agarose for 2 hours. The washed beads were resuspended in 50 .mu.L of Laemmli sample buffer and boiled for 5 min, and the proteins were separated by 5-10% SDS PAGE gels (BIO RAD). After electrophoresis, the gels were stained with silver. Protein bands specifically found in IMS-E21 (URLC8)-transfected extracts were excised and served for matrix-assisted laser desorption/ionization-time of flight mass spectrometry (MALDI-TOF-MS) analysis (AXIMA-CFR plus, SHIMADZU BIOTECH, Kyoto, Japan).

(h) RNA Interference Assay.

[0139] A vector-based RNA interference (RNAi) system, psiH1BX3.0, was previously established to direct the synthesis of siRNAs in mammalian cells (Suzuki, C. et al. (2003) Cancer Res, 63: 7038-7041.). Herein, 10 .mu.g of siRNA-expression vector were transfected, using 30 .mu.l of Lipofectamine 2000 (Invitrogen), into NSCLC cell lines A549 and LC319, both of which over-expressed IMS-E21 (URLC8) endogenously. More than 80% of the transfected cells expressed the synthetic siRNA, and in those cells endogenous expression of the IMS-E21 (URLC8) genes was effectively suppressed. The transfected cells were cultured for seven days in the presence of appropriate concentrations of geneticin (G418), after which cell numbers and viability were measured by Giemsa staining and triplicate MTT assays. The target sequences of the synthetic oligonucleotides for RNAi were as follows:

TABLE-US-00003 control 1 (EGFP: enhanced green fluorescent protein (GFP) gene, a mutant of Aequorea victoria GFP), 5'-GAAGCAGCACGACTTCTTC-3'; (SEQ ID NO. 7) control 2 (Luciferase: Photinus pyralis luciferase gene), 5'-CGTACGCGGAATACTTCGA-3'; (SEQ ID NO. 8) control 3 (Scramble: chloroplast Euglena gracilis gene coding for 5S and 16S rRNAs), 5'-GCGCGCTTTGTAGGATTCG-3'; (SEQ ID NO. 9) siRNA-IMS-E21-#2 (si-IMS-E21-#2), 5'-TGAGGTGCTCAGCACAGTG-3'; (SEQ ID NO. 10) siRNA-IMS-E21-#3 (si-IMS-E21-#3), 5'-GTTGGCACAGCCTGTGTAT-3'. (SEQ ID NO. 11)

[0140] The insert sequences of siRNA expression vectors were as follows:

TABLE-US-00004 control 1 (EGFP); (SEQ ID NO. 12) 5'-TCCCGAAGCAGCACGACTTCTTCTTCAAGAGAGAAGAAGTCGTGC TGCTTC-3'; (SEQ ID NO. 13) 5'-AAAAGAAGCAGCACGACTTCTTCTCTCTTGAAGAAGAAGTCGTGC TGCTTC-3'; control 2 (Luciferase); (SEQ ID NO. 15) 5'-TCCCCGTACGCGGAATACTTCGATTCAAGAGATCGAAGTATTCCG CGTACG-3'; (SEQ ID NO. 16) 5'-AAAACGTACGCGGAATACTTCGATCTCTTGAAATCGAAGTATTCC GCGTACG-3'; control 3 (Scramble); (SEQ ID NO. 18) 5'-TCCCGCGCGCTTTGTAGGATTCGTTCAAGAGACGAATCCTACAAA GCGCGC-3'; (SEQ ID NO. 19) 5'-AAAAGCGCGCTTTGTAGGATTCGTCTCTTGAACGAATCCTACAAA GCGCGC-3'; siRNA-IMS-E21-#2 (si-IMS-E2-#2); (SEQ ID NO. 21) 5'-TCCCTGAGGTGCTCAGCACAGTGTTCAAGAGACACTGTGCTGAGC ACCTCA-3'; (SEQ ID NO. 22) 5'-AAAATGAGGTGCTCAGCACAGTGTCTCTTGAACACTGTGCTGAGC ACCTCA-3'; siRNA-IMS-E21-#3 (si-IMS-E21-#3); (SEQ ID NO. 24) 5'-TCCCGTTGGCACAGCCTGTGTATTCAAGAGAATACACAGGCTGTG CCAAC-3'; (SEQ ID NO. 25) 5'-AAAAGTTGGCACAGCCTGTGTATCTCTTGAAATACACAGGCTGTG CCAAC-3';

(i) Immunohistochemistry and Tissue Microarray Analysis.

[0141] The tumor tissue microarrays using formalin-fixed NSCLCs were constructed as published elsewhere. The tissue area for sampling was selected based on a visual alignment with the corresponding HE-stained section on a slide. Three, four, or five tissue cores (diameter 0.6 mm; height 3-4 mm) taken from the donor tumor blocks were placed into a recipient paraffin block using a tissue microarrayer (Beecher Instruments, Sun Prairie, Wis.). A core of normal tissue was punched from each case. 5-.mu.m sections of the resulting microarray block were used for immunohistochemical analysis. IMS-E21 (URLC8) positivity were assessed semi-quantitatively, recording staining intensity as absent (scored as 0), weak (scored as 1+) or strongly positive (scored as 2+), by three independent investigators without prior knowledge of the clinical follow-up data. Cases were accepted only as positive if reviewers independently defined them thus.

[0142] To investigate the presence of IMS-E21 (URLC8) protein in clinical tissue samples, the sections were stained using ENVISION+ Kit/HRP (DakoCytomation). Affinity-purified anti-IMS-E21 (URLC8) antibody was added after blocking endogenous peroxidase and proteins, and the sections were incubated with HRP-labeled anti-rabbit IgG as the secondary antibody. Substrate-chromogen was added and the specimens were counterstained with hematoxylin.

Example 2

Cloning of IMS-E21 (URLC8) Gene and Its Expression in Lung Tumors, Cell Lines, and Normal Tissues

[0143] To obtain novel target molecules for development of therapeutic agents and/or diagnostic markers for NSCLC, genes that showed 5-fold higher expression in more than 50% of 37 NSCLCs analyzed by cDNA microarray (Kikuchi, et.al. (2003) Oncogene, 22: 2192-2205.) were screened. Among 23,040 genes screened, one EST transcript (FLJ20399, NM.sub.--017803) was identified as frequently over-expressed in NSCLCs; its overexpression was confirmed--in eight representative cases by semi-quantitative RT-PCR experiments. To obtain a full-length clone of this transcript, the FASTA database was screened using the cDNA fragment originally isolated by cDNA microarray experiment and identified several ESTs. Assembling DNA sequences of these clones, the entire coding nucleotide sequence of this gene was determined. The cDNA consisted of 2,020 nucleotides, including an open reading frame of 1,479 nucleotides that encoded a 493 amino acid peptide. A homology search using the FASTA program revealed that this predicted protein was homologous to proteins belonging to the family of tRNA-dihydrouridine synthase (DUS), especially S. cerevisiae Dus1 (dihydrouridine synthase 1) which catalyses the reduction of the 5,6-double bond of a uridine residue on tRNA (30% identity in amino acids, respectively; FIG. 1, a-c). Hence, this gene was tentatively designated IMS-E21 (also referred to as URLC8: up-regulated in lung cancer 8, Accession No. AB101210). Two motifs were identified using SMART program (http://smart.embl-heidelberg.de/) (Letunic I, et al., (2004) Nucleic Acids Res.;32(Database issue):D142-4; Schultz J, et al., (1998) Proc Natl Acad Sci USA.;95:5857-64.) within the sequence of IMS-E21 (URLC8); N-terminal Dus domain and C-terminal DSRM, respectively (FIG. 1, a). The human IMS-E21 (URLC8) appeared to conserve 90% identity in amino acids with the murine (M. musculus and R. Norvegicus) products, 48% with the D. melanogaster protein, and 40% with C. elegans protein (FIG. 1, b).

[0144] Increased IMS-E21 (URLC8) expression was further confirmed in 11 of 14 additional NSCLC cases (4 of 7 adenocarcinomas (ADCs); 7 of 7 squamous-cell carcinomas (SCCs) (FIG. 2, a), and documented up-regulation of IMS-E21 (URLC8) in 21 of the 23 NSCLC and small-cell lung cancer (SCLC) cell lines examined, but no expression was found in SAEC cells derived from normal bronchial epithelium (FIG. 2, b).

[0145] Northern blotting with IMS-E21 (URLC8) cDNA as a probe identified a 2.4-kb transcript as a band, mainly seen in testis, among the 23 normal human tissues examined (FIG. 2, c).

Example 3

Overexpression of IMS-E21 (URLC8) Protein is Associated with a Worse Outcome in SCC

[0146] Immunohistochemical analysis was performed with affinity-purified anti-IMS-E21 (URLC8) polyclonal antibodies in tissue microarrays of available 292 NSCLCs. The study showed that the number of cases that showed positive staining of IMS-E21 (URLC8) in the cytoplasm was 254 of 292 (87%) NSCLC total cases; 132 of 158 (84%) ADC cases, 89 of 99 (90%) SCC cases, 19 of 21 (90%) LCC cases, 10 of 10 (100%) BAC cases, and 4 of 4 (100%) ASC cases, respectively. All of those tumors were surgically-resectable NSCLCs, and no staining was observed in any of their adjacent normal lung tissues (FIG. 3, a). A pattern of IMS-E21 (URLC8) expression was classified on the tissue array, ranging from absent/weak (scored as 0.about.1+) to strong (scored as 2+). Next, the association of IMS-E21 (URLC8) expression with clinical outcome was examined. Statistical analysis revealed no significant correlation of any levels of IMS-E21 (URLC8) expression with pT- or pN-factors among the lung-cancer patients examined. However, the expression of IMS-E21 (URLC8) in SCC was found to be significantly associated with tumor specific 5 year-survival using Kaplan-Meier method (P=0.0091 by the Log-rank test) (FIG. 3, b). By univariate analysis pT, pN, gender, and IMS-E21 (URLC8) expression (P=0.0111) were each significantly related to a poor tumor-specific survival among SCC patients. Furthermore, IMS-E21 (URLC8) staining was determined to be an independent prognostic factors by multivariate analysis using Cox proportional hazard model of SCC patients (P=0.0234).

Example 4

Inhibition of Growth of NSCLC Cells by Specific siRNA Against IMS-E21 (URLC8)

[0147] To assess whether IMS-E21 (URLC8) is essential for growth or survival of lung-cancer cells, plasmids were designed and constructed to express siRNA against IMS-E21 (URLC8) (si-IMS-E21) and control plasmids (siRNAs for EGFP, Scramble, and Luciferase) and transfected them into A549 and LC319 cells to suppress expression of endogenous IMS-E21 (URLC8). The amount of IMS-E21 (URLC8) transcript and protein level in the cells transfected with si-IMS-E21-#2, and -#3 were significantly decreased in comparison with cells transfected with the control (FIG. 3, c, left panels); transfection of si-IMS-E21-#2, and -#3 also resulted in significant decreases of colony number in colony-formation assays (FIG. 3, c, lower panels) and colony numbers measured by MTT cell viability assay (FIG. 3, c, right panel).

Example 5

IMS-E21 (URLC8) Localized Mainly in Cytoplasm and Co-Localized with ER-Abundant Protein PDI

[0148] To determine the subcellular localization of endogenous IMS-E21 (URLC8) in lung cancer-cells, immunocytochemistry was performed using affinity-purified anti-IMS-E21 (URLC8) polyclonal antibodies. Confocal microscopy revealed the distribution of the IMS-E21 (URLC8) protein in the cytoplasm of A549 cells (data not shown). To further investigate the sub-cellular localization of IMS-E21 (URLC8) proteins, we transfected LC319 cells with plasmids that contained c-myc-His-epitope sequences (LDEESILKQE-HHHHHH) at the C-terminal of the human IMS-E21 (URLC8) protein. Co-immunostaining using anti-c-myc antibodies and antibodies against endogenous Protein Disulfide Isomerase (PDI), which are abundantly expressed in endoplasmic reticulum (ER) suggested that IMS-E21 (URLC8) protein mainly distribute at ER (FIG. 4, a).

Example 6

Identification of EPRS as a Protein Interacting with IMS-E21 (URLC8)

[0149] To elucidate the function of IMS-E21 (URLC8) in lung-cancer cells, protein(s) interacting with IMS-E21 (URLC8) were identified. Lysate of LC319 cells transfected with N-terminal FLAG-tagged and C-terminal HA-tagged-pCAGGS-IMS-E21 vector or mock vector (control) were extracted and immunoprecipitated with anti-FLAG M2 monoclonal antibody followed by immunoprecipitation with anti-HA monoclonal antibody. The protein complex including IMS-E21 (URLC8) was stained with SilverQuest (Invitrogen) on the SDS-PAGE gel. A 180-kDa band, which was seen in immunoprecipitates of cell lysates transfected with IMS-E21 (URLC8) expressing plasmids, but not seen in those with mock plasmids was extracted and determined to be EPRS (glutamyl-prolyl tRNA synthetase), by MALDI-TOF mass spectrometric sequencing (data not shown). The interaction between endogenous IMS-E21 (URLC8) and EPRS was confirmed by co-immunoprecipitation and immunocytochemistry (FIG. 4, b and c), suggesting the possibility of existence of IMS-E21 (URLC8)-aminoacyl tRNA synthetase complex in an ER-dependent translation.

Example 7

tRNA-DUS Activity of IMS-E21 (URLC8) in Lung Cancer Cells

[0150] To test the hypothesis that IMS-E21 (URLC8) encodes a family of dihydrouridine synthase enzyme, the effects of suppression of IMS-E21 (URLC8) gene by siRNA on DUS activity in NSCLC cell line were investigated. Total tRNA was purified from total RNA of A549 and LC319 cells transfected with siRNA constructs by HPLC technique and analyzed the samples for dihydrouridine content of each cell line (Jacobson, M. and Hedgcoth, C. (1970) Anal Biochem, 34: 459-469. Hunninghake, D. and Grisolia, S. (1966) Anal Biochem, 16: 200-205.). Most effective IMS-E21 (URLC8) siRNA constructs (si-IMS-E21-#2), which could suppress transcript levels of IMS-E21 (URLC8) as stated above, also lead to reduce levels of tRNA-dihydrouridine content when compared with their corresponding control siRNA construct (si-EGFP) (FIG. 4, d). The deficiencies of tRNA-dihydrouridine induced by si-IMS-E21-#2 in these tumor cells were closely related to the tumor growth suppression (FIG. 3, c), implicating that tRNA-DUS activity of IMS-E21 (URLC8) is indispensable for tumor cell survival and/or progression.

INDUSTRIAL APPLICABILITY

[0151] The present inventors have shown that URLC8 has t-RNA dihydrouridine-synthase activity, and the suppression of the activity leads to the inhibition of cell proliferation of lung cancer cells. Thus, agents that inhibit the t-RNA dihydrouridine-synthase activity of URLC8 find therapeutic utility as anti-cancer agents for the treatment of lung cancer, such as NSCLC.

[0152] In addition, treatment of NSCLC cells with siRNA against URLC8 suppressed its expression and also the activity of tRNA-DUS, and suppressed growth of the cancer cells. Additional experiments revealed that the URLC8 protein physically interacts with EPRS (glutamyl-prolyl tRNA synthetase) in NSCLC cells. These data imply that up-regulation of URLC8 function and increased tRNA-DUS activity are common features of pulmonary carcinogenesis. Accordingly, the selective suppression of URLC8 enzyme activity and/or the formation of the URLC8-tRNA synthetase complex may be a promising therapeutic strategy for treatment of lung-cancer patients.

[0153] Alternatively, lung cancer can be detected using t-RNA dihydrouridine-synthase activity of URLC8 as a diagnostic index.

[0154] Furthermore, the present inventors revealed that a high level of URLC8 (IMS-E21) expression was significantly associated with poor prognosis for patients with lung squamous-cell carcinoma (SCC). Accordingly, a prognosis of lung cancer can be predicted by measurement of URLC8 (IMS-E21) expression level.

[0155] All patents, patent applications, and publications cited herein are incorporated by reference in their entirety. Furthermore, while the invention has been described in detail and with reference to specific embodiments thereof, it will be apparent to one skilled in the art that various changes and modifications can be made therein without departing from the spirit and scope of the invention.

Sequence CWU 1

1

3312020DNAHomo sapiensCDS(171)...(1652) 1gctcagtacg gtgtgtggag ctggagcacc gtgaggaaga agcgaggttc tttttaagag 60ttcagctgcg agatatcaaa caaagaatta ctctgtacaa agccagaaca catatatcaa 120agtaatcctg aagtatcaga acaaaataat aggctgtaac agaggaggaa atg att 176 Met Ile 1ttg aat agc ctc tct ctg tgt tac cat aat aag cta atc ctg gcc cca 224Leu Asn Ser Leu Ser Leu Cys Tyr His Asn Lys Leu Ile Leu Ala Pro 5 10 15atg gtt cgg gta ggg act ctt cca atg agg ctg ctg gcc ctg gat tat 272Met Val Arg Val Gly Thr Leu Pro Met Arg Leu Leu Ala Leu Asp Tyr 20 25 30gga gcg gac att gtt tac tgt gag gag ctg atc gac ctc aag atg att 320Gly Ala Asp Ile Val Tyr Cys Glu Glu Leu Ile Asp Leu Lys Met Ile35 40 45 50cag tgc aag aga gtt gtt aat gag gtg ctc agc aca gtg gac ttt gtc 368Gln Cys Lys Arg Val Val Asn Glu Val Leu Ser Thr Val Asp Phe Val 55 60 65gcc cct gat gat cga gtt gtc ttc cgc acc tgt gaa aga gag cag aac 416Ala Pro Asp Asp Arg Val Val Phe Arg Thr Cys Glu Arg Glu Gln Asn 70 75 80agg gtg gtc ttc cag atg ggg act tca gac gca gag cga gcc ctt gct 464Arg Val Val Phe Gln Met Gly Thr Ser Asp Ala Glu Arg Ala Leu Ala 85 90 95gtg gcc agg ctt gta gaa aat gat gtg gct ggt att gat gtc aac atg 512Val Ala Arg Leu Val Glu Asn Asp Val Ala Gly Ile Asp Val Asn Met 100 105 110ggc tgt cca aaa caa tat tcc acc aag gga gga atg gga gct gcc ctg 560Gly Cys Pro Lys Gln Tyr Ser Thr Lys Gly Gly Met Gly Ala Ala Leu115 120 125 130ctg tca gac cct gac aag att gag aag atc ctc agc act ctt gtt aaa 608Leu Ser Asp Pro Asp Lys Ile Glu Lys Ile Leu Ser Thr Leu Val Lys 135 140 145ggg aca cgc aga cct gtg acc tgc aag att cgc atc ctg cca tcg cta 656Gly Thr Arg Arg Pro Val Thr Cys Lys Ile Arg Ile Leu Pro Ser Leu 150 155 160gaa gat acc ctg agc ctt gtg aag cgg ata gag agg act ggc att gct 704Glu Asp Thr Leu Ser Leu Val Lys Arg Ile Glu Arg Thr Gly Ile Ala 165 170 175gcc atc gca gtt cat ggg agg aag cgg gag gag cga cct cag cat cct 752Ala Ile Ala Val His Gly Arg Lys Arg Glu Glu Arg Pro Gln His Pro 180 185 190gtc agc tgt gaa gtc atc aaa gcc att gct gat acc ctc tcc att cct 800Val Ser Cys Glu Val Ile Lys Ala Ile Ala Asp Thr Leu Ser Ile Pro195 200 205 210gtc ata gcc aac gga gga tct cat gac cac atc caa cag tat tcg gac 848Val Ile Ala Asn Gly Gly Ser His Asp His Ile Gln Gln Tyr Ser Asp 215 220 225ata gag gac ttt cga caa gcc acg gca gcc tct tcc gtg atg gtg gcc 896Ile Glu Asp Phe Arg Gln Ala Thr Ala Ala Ser Ser Val Met Val Ala 230 235 240cga gca gcc atg tgg aac cca tct atc ttc ctc aag gag ggt ctg cgg 944Arg Ala Ala Met Trp Asn Pro Ser Ile Phe Leu Lys Glu Gly Leu Arg 245 250 255ccc ctg gag gag gtc atg cag aaa tac atc aga tac gcg gtg cag tat 992Pro Leu Glu Glu Val Met Gln Lys Tyr Ile Arg Tyr Ala Val Gln Tyr 260 265 270gac aac cac tac acc aac acc aag tac tgc ttg tgc cag atg cta cga 1040Asp Asn His Tyr Thr Asn Thr Lys Tyr Cys Leu Cys Gln Met Leu Arg275 280 285 290gaa cag ctg gag tcg ccc cag gga agg ttg ctc cat gct gcc cag tct 1088Glu Gln Leu Glu Ser Pro Gln Gly Arg Leu Leu His Ala Ala Gln Ser 295 300 305tcc cgg gaa att tgt gag gcc ttt ggc ctt ggt gcc ttc tat gag gag 1136Ser Arg Glu Ile Cys Glu Ala Phe Gly Leu Gly Ala Phe Tyr Glu Glu 310 315 320acc aca cag gag ctg gat gcc cag cag gcc agg ctc tca gcc aag act 1184Thr Thr Gln Glu Leu Asp Ala Gln Gln Ala Arg Leu Ser Ala Lys Thr 325 330 335tca gag cag aca ggg gag cca gct gaa gat acc tct ggt gtc att aag 1232Ser Glu Gln Thr Gly Glu Pro Ala Glu Asp Thr Ser Gly Val Ile Lys 340 345 350atg gct gtc aag ttt gac cgg aga gca tac cca gcc cag atc acc cct 1280Met Ala Val Lys Phe Asp Arg Arg Ala Tyr Pro Ala Gln Ile Thr Pro355 360 365 370aag atg tgc cta cta gag tgg tgc cgg agg gag aag ttg gca cag cct 1328Lys Met Cys Leu Leu Glu Trp Cys Arg Arg Glu Lys Leu Ala Gln Pro 375 380 385gtg tat gaa acg gtt caa cgc cct cta gat cgc ctg ttc tcc tct att 1376Val Tyr Glu Thr Val Gln Arg Pro Leu Asp Arg Leu Phe Ser Ser Ile 390 395 400gtc acc gtt gct gaa caa aag tat cag tct acc ttg tgg gac aag tcc 1424Val Thr Val Ala Glu Gln Lys Tyr Gln Ser Thr Leu Trp Asp Lys Ser 405 410 415aag aaa ctg gcg gag cag gct gca gcc atc gtc tgt ctg cgg agc cag 1472Lys Lys Leu Ala Glu Gln Ala Ala Ala Ile Val Cys Leu Arg Ser Gln 420 425 430ggc ctc cct gag ggt cgg ctg ggt gag gag agc cct tcc ttg cac aag 1520Gly Leu Pro Glu Gly Arg Leu Gly Glu Glu Ser Pro Ser Leu His Lys435 440 445 450cga aag agg gag gct cct gac caa gac cct ggg ggc ccc aga gct cag 1568Arg Lys Arg Glu Ala Pro Asp Gln Asp Pro Gly Gly Pro Arg Ala Gln 455 460 465gag cta gca caa cct ggg gat ctg tgc aag aag ccc ttt gtg gcc ttg 1616Glu Leu Ala Gln Pro Gly Asp Leu Cys Lys Lys Pro Phe Val Ala Leu 470 475 480gga agt ggt gaa gaa agc ccc ctg gaa ggc tgg tga ctactcttcc 1662Gly Ser Gly Glu Glu Ser Pro Leu Glu Gly Trp 485 490tgccttagtc acccctccat gggcctggtg ctaaggtggc tgtggatgcc acagcatgaa 1722ccagatgccg ttgaacagtt tgctggtctt gcctggcaga agttagatgt cctggcaggg 1782gccatcagcc tagagcatgg accaggggcc gcccaggggt ggatcctggc ccctttggtg 1842gatctgagtg acagggtcaa gttctctttg aaaacaggag cttttcaggt ggtaactccc 1902caacctgaca ttggtactgt gcaataaaga caccccctac cctcaaaaaa aaaaaaaaaa 1962aaaaaaaaaa aaaaaaaaaa aaaaaaaaaa aaaaaaaaaa aaaaaaaaaa aaaaaaaa 20202493PRTHomo sapiens 2Met Ile Leu Asn Ser Leu Ser Leu Cys Tyr His Asn Lys Leu Ile Leu1 5 10 15Ala Pro Met Val Arg Val Gly Thr Leu Pro Met Arg Leu Leu Ala Leu 20 25 30Asp Tyr Gly Ala Asp Ile Val Tyr Cys Glu Glu Leu Ile Asp Leu Lys 35 40 45Met Ile Gln Cys Lys Arg Val Val Asn Glu Val Leu Ser Thr Val Asp 50 55 60Phe Val Ala Pro Asp Asp Arg Val Val Phe Arg Thr Cys Glu Arg Glu65 70 75 80Gln Asn Arg Val Val Phe Gln Met Gly Thr Ser Asp Ala Glu Arg Ala 85 90 95Leu Ala Val Ala Arg Leu Val Glu Asn Asp Val Ala Gly Ile Asp Val 100 105 110Asn Met Gly Cys Pro Lys Gln Tyr Ser Thr Lys Gly Gly Met Gly Ala 115 120 125Ala Leu Leu Ser Asp Pro Asp Lys Ile Glu Lys Ile Leu Ser Thr Leu 130 135 140Val Lys Gly Thr Arg Arg Pro Val Thr Cys Lys Ile Arg Ile Leu Pro145 150 155 160Ser Leu Glu Asp Thr Leu Ser Leu Val Lys Arg Ile Glu Arg Thr Gly 165 170 175Ile Ala Ala Ile Ala Val His Gly Arg Lys Arg Glu Glu Arg Pro Gln 180 185 190His Pro Val Ser Cys Glu Val Ile Lys Ala Ile Ala Asp Thr Leu Ser 195 200 205Ile Pro Val Ile Ala Asn Gly Gly Ser His Asp His Ile Gln Gln Tyr 210 215 220Ser Asp Ile Glu Asp Phe Arg Gln Ala Thr Ala Ala Ser Ser Val Met225 230 235 240Val Ala Arg Ala Ala Met Trp Asn Pro Ser Ile Phe Leu Lys Glu Gly 245 250 255Leu Arg Pro Leu Glu Glu Val Met Gln Lys Tyr Ile Arg Tyr Ala Val 260 265 270Gln Tyr Asp Asn His Tyr Thr Asn Thr Lys Tyr Cys Leu Cys Gln Met 275 280 285Leu Arg Glu Gln Leu Glu Ser Pro Gln Gly Arg Leu Leu His Ala Ala 290 295 300Gln Ser Ser Arg Glu Ile Cys Glu Ala Phe Gly Leu Gly Ala Phe Tyr305 310 315 320Glu Glu Thr Thr Gln Glu Leu Asp Ala Gln Gln Ala Arg Leu Ser Ala 325 330 335Lys Thr Ser Glu Gln Thr Gly Glu Pro Ala Glu Asp Thr Ser Gly Val 340 345 350Ile Lys Met Ala Val Lys Phe Asp Arg Arg Ala Tyr Pro Ala Gln Ile 355 360 365Thr Pro Lys Met Cys Leu Leu Glu Trp Cys Arg Arg Glu Lys Leu Ala 370 375 380Gln Pro Val Tyr Glu Thr Val Gln Arg Pro Leu Asp Arg Leu Phe Ser385 390 395 400Ser Ile Val Thr Val Ala Glu Gln Lys Tyr Gln Ser Thr Leu Trp Asp 405 410 415Lys Ser Lys Lys Leu Ala Glu Gln Ala Ala Ala Ile Val Cys Leu Arg 420 425 430Ser Gln Gly Leu Pro Glu Gly Arg Leu Gly Glu Glu Ser Pro Ser Leu 435 440 445His Lys Arg Lys Arg Glu Ala Pro Asp Gln Asp Pro Gly Gly Pro Arg 450 455 460Ala Gln Glu Leu Ala Gln Pro Gly Asp Leu Cys Lys Lys Pro Phe Val465 470 475 480Ala Leu Gly Ser Gly Glu Glu Ser Pro Leu Glu Gly Trp 485 490321DNAArtificial SequenceAn artificially synthesized primer sequence for RT-PCR 3gaccacatcc aacagtattc g 21421DNAArtificial SequenceAn artificially synthesized primer sequence for RT-PCR 4tgccaggaca tctaacttct g 21521DNAArtificial SequenceAn artificially synthesized primer sequence for RT-PCR 5gaggtgatag cattgctttc g 21621DNAArtificial SequenceAn artificially synthesized primer sequence for RT-PCR 6caagtcagtg tacaggtaag c 21719DNAArtificial SequenceAn artificially synthesized target sequence for siRNA 7gaagcagcac gacttcttc 19819DNAArtificial SequenceAn artificially synthesized target sequence for siRNA 8cgtacgcgga atacttcga 19919DNAArtificial SequenceAn artificially synthesized target sequence for siRNA 9gcgcgctttg taggattcg 191019DNAArtificial SequenceAn artificially synthesized target sequence for siRNA 10tgaggtgctc agcacagtg 191119DNAArtificial SequenceAn artificially synthesized target sequence for siRNA 11gttggcacag cctgtgtat 191251DNAArtificial SequenceAn artificially synthesized oligonucleotide sequence for siRNA 12tcccgaagca gcacgacttc ttcttcaaga gagaagaagt cgtgctgctt c 511351DNAArtificial SequenceAn artificially synthesized oligonucleotide sequence for siRNA 13aaaagaagca gcacgacttc ttctctcttg aagaagaagt cgtgctgctt c 511447DNAArtificial SequenceAn artificially synthesized hairpin sequence for siRNA 14gaagcagcac gacttcttct tcaagagaga agaagtcgtg ctgcttc 471551DNAArtificial SequenceAn artificially synthesized oligonucleotide sequence for siRNA 15tccccgtacg cggaatactt cgattcaaga gatcgaagta ttccgcgtac g 511652DNAArtificial SequenceAn artificially synthesized oligonucleotide sequence for siRNA 16aaaacgtacg cggaatactt cgatctcttg aaatcgaagt attccgcgta cg 521747DNAArtificial SequenceAn artificially synthesized hairpin sequence for siRNA 17cgtacgcgga atacttcgat tcaagagatc gaagtattcc gcgtacg 471851DNAArtificial SequenceAn artificially synthesized oligonucleotide sequence for siRNA 18tcccgcgcgc tttgtaggat tcgttcaaga gacgaatcct acaaagcgcg c 511951DNAArtificial SequenceAn artificially synthesized oligonucleotide sequence for siRNA 19aaaagcgcgc tttgtaggat tcgtctcttg aacgaatcct acaaagcgcg c 512047DNAArtificial SequenceAn artificially synthesized hairpin sequence for siRNA 20gcgcgctttg taggattcgt tcaagagacg aatcctacaa agcgcgc 472151DNAArtificial SequenceAn artificially synthesized oligonucleotide sequence for siRNA 21tccctgaggt gctcagcaca gtgttcaaga gacactgtgc tgagcacctc a 512251DNAArtificial SequenceAn artificially synthesized oligonucleotide sequence for siRNA 22aaaatgaggt gctcagcaca gtgtctcttg aacactgtgc tgagcacctc a 512347DNAArtificial SequenceAn artificially synthesized hairpin sequence for siRNA 23tgaggtgctc agcacagtgt tcaagagaca ctgtgctgag cacctca 472450DNAArtificial SequenceAn artificially synthesized oligonucleotide sequence for siRNA 24tcccgttggc acagcctgtg tattcaagag aatacacagg ctgtgccaac 502550DNAArtificial SequenceAn artificially synthesized oligonucleotide sequence for siRNA 25aaaagttggc acagcctgtg tatctcttga aatacacagg ctgtgccaac 502646DNAArtificial SequenceAn artificially synthesized hairpin sequence for siRNA 26gttggcacag cctgtgtatt caagagaata cacaggctgt gccaac 4627493PRTMus musculus 27Met Ile Val Asn Ser Leu Ser Leu Cys Tyr His Asn Lys Leu Ile Leu1 5 10 15Ala Pro Met Val Arg Val Gly Thr Leu Pro Met Arg Leu Leu Ala Leu 20 25 30Asp Tyr Gly Ala Asp Ile Val Tyr Cys Glu Glu Leu Ile Asp Leu Lys 35 40 45Met Leu Gln Cys Lys Arg Val Val Asn Glu Val Leu Ser Thr Val Asp 50 55 60Phe Val Ala Pro Asp Asp Arg Val Val Phe Arg Thr Cys Glu Arg Glu65 70 75 80Gln Ser Arg Val Val Phe Gln Met Gly Thr Ser Asp Ala Glu Arg Ala 85 90 95Leu Ala Val Ala Arg Leu Val Glu Asn Asp Val Ala Gly Ile Asp Val 100 105 110Asn Met Gly Cys Pro Lys Glu Tyr Ser Thr Lys Gly Gly Met Gly Ala 115 120 125Ala Leu Leu Ser Asp Pro Asp Lys Ile Glu Lys Ile Leu Ser Thr Leu 130 135 140Val Lys Gly Thr His Arg Pro Val Thr Cys Lys Ile Arg Ile Leu Pro145 150 155 160Ser Leu Glu Asp Thr Leu Asn Leu Val Lys Arg Ile Glu Arg Thr Gly 165 170 175Ile Ser Ala Ile Ala Val His Gly Arg Asn Arg Asp Glu Arg Pro Gln 180 185 190His Pro Val Ser Cys Glu Val Ile Arg Ala Ile Ala Glu Thr Leu Ser 195 200 205Ile Pro Val Ile Ala Asn Gly Gly Ser His Asp His Ile Gln Gln His 210 215 220Val Asp Ile Glu Asp Phe Arg Gln Ala Thr Ala Ala Ser Ser Val Met225 230 235 240Val Ala Arg Ala Ala Met Trp Asn Pro Ser Ile Phe Leu Lys Asp Gly 245 250 255Leu Arg Pro Leu Glu Glu Val Met Gln Lys Tyr Ile Arg Tyr Ala Val 260 265 270Gln Tyr Asp Asn His Tyr Thr Asn Thr Lys Tyr Cys Leu Cys Gln Met 275 280 285Leu Arg Glu Gln Leu Glu Ser Pro Gln Gly Arg Leu Leu His Ala Ala 290 295 300Gln Ser Ser Gln Glu Ile Cys Glu Ala Phe Gly Leu Gly Ala Phe Tyr305 310 315 320Glu Glu Thr Ile Arg Glu Leu Asp Ala Arg Arg Ala Asp Leu Leu Ala 325 330 335Lys Thr Pro Glu Ala Val Glu Glu Pro Ala Glu Asp Thr Ser Gly Ile 340 345 350Ile Lys Met Ala Ile Arg Phe Asp Arg Arg Ala Tyr Pro Pro Gln Ile 355 360 365Thr Pro Lys Met Cys Leu Leu Glu Trp Cys Arg Arg Glu Lys Leu Pro 370 375 380Gln Pro Val Tyr Glu Thr Val Gln Arg Thr Ile Asp Arg Met Phe Cys385 390 395 400Ser Val Val Thr Val Ala Glu Gln Lys Tyr Gln Ser Thr Leu Trp Asp 405 410 415Lys Ser Lys Lys Leu Ala Glu Gln Thr Ala Ala Ile Val Cys Leu Arg 420 425 430Ser Gln Gly Leu Pro Glu Gly Arg Leu Gly Glu Glu Ser Pro Ser Leu 435 440 445Asn Lys Arg Lys Arg Glu Ala Pro Asp Gln Asp Pro Gly Gly Pro Arg 450 455 460Val Gln Glu Pro Ala Leu Pro Gly Glu Ile Cys Lys Lys Pro Phe Val465 470 475 480Thr Leu Asp Ser Ser Glu Glu Asn Leu Leu Glu Gly Cys 485 49028531PRTRattus norvegicus 28Met Ile Val Asn Ser Leu Ser Leu Cys Tyr His Asn Lys Leu Ile Leu1 5 10 15Ala Pro Met Val Arg Val Gly Thr Leu Pro Met Arg Leu Leu Ala Leu

20 25 30Asp Tyr Gly Ala Asp Ile Val Tyr Cys Glu Glu Leu Ile Asp Leu Lys 35 40 45Met Leu Gln Cys Arg Arg Val Val Asn Glu Val Leu Ser Thr Val Asp 50 55 60Phe Val Ala Pro Asp Asp Arg Val Val Phe Arg Thr Cys Glu Arg Glu65 70 75 80Gln Ser Arg Val Val Phe Gln Met Gly Thr Ser Asp Ala Glu Arg Ala 85 90 95Leu Ala Val Ala Arg Leu Val Glu Asn Asp Val Ala Gly Ile Asp Val 100 105 110Asn Met Gly Cys Pro Lys Glu Tyr Ser Thr Lys Gly Gly Met Gly Ala 115 120 125Ala Leu Leu Ser Asp Pro Asp Lys Ile Glu Lys Ile Leu Ser Thr Leu 130 135 140Val Lys Gly Thr His Arg Pro Val Thr Cys Lys Ile Arg Ile Leu Pro145 150 155 160Ser Leu Glu Asp Thr Leu Asn Leu Val Lys Arg Ile Glu Arg Thr Gly 165 170 175Ile Ser Ala Ile Ala Val His Gly Arg Asn Arg Asp Glu Arg Pro Gln 180 185 190His Pro Val Ser Cys Glu Val Ile Arg Ala Ile Ala Glu Thr Leu Ser 195 200 205Ile Pro Val Ile Ala Lys Val Leu Ile Val Glu Gly Leu Leu Lys Leu 210 215 220Thr Asp Asn Glu Arg Gln Arg Ser Ser Gly Asn Thr Gly Arg Phe His225 230 235 240Tyr Gly Ile Leu Pro Asn Pro Leu Leu Leu Phe Ser Gly Gly Ser His 245 250 255Asp His Ile Gln Gln His Leu Asp Ile Glu Asp Phe Arg Gln Ala Thr 260 265 270Ala Ala Ser Ser Val Met Val Ala Arg Ala Ala Met Trp Asn Pro Ser 275 280 285Ile Phe Leu Lys Asp Gly Leu Arg Pro Leu Glu Glu Val Met Gln Lys 290 295 300Tyr Ile Arg Tyr Ala Val Gln Tyr Asp Asn His Tyr Thr Asn Thr Lys305 310 315 320Tyr Cys Leu Cys Gln Met Leu Arg Glu Gln Leu Glu Ser Pro Gln Gly 325 330 335Arg Met Leu His Ala Ala Gln Ser Ser Gln Glu Ile Cys Glu Ala Phe 340 345 350Gly Leu Gly Thr Phe Tyr Glu Asp Thr Ile Arg Glu Leu Asp Ala Arg 355 360 365Arg Ala Asp Leu Leu Ala Lys Thr Pro Glu Ala Val Glu Glu Pro Ala 370 375 380Glu Asp Thr Ser Gly Ile Ile Lys Met Ala Ile Arg Phe Asp Arg Arg385 390 395 400Ala Tyr Pro Pro Gln Ile Thr Pro Lys Thr Cys Leu Leu Glu Trp Cys 405 410 415Arg Arg Glu Lys Leu Pro Gln Pro Val Tyr Glu Thr Val Gln Arg Pro 420 425 430Ile Asp Arg Met Phe Cys Ser Val Val Thr Val Ala Glu Gln Lys Tyr 435 440 445Gln Ser Thr Leu Trp Asp Lys Ser Lys Lys Leu Ala Glu Gln Thr Ala 450 455 460Ala Ile Val Cys Leu Arg Ser Gln Gly Leu Pro Glu Gly Arg Leu Gly465 470 475 480Glu Glu Asn Pro Ser Leu Asn Lys Arg Lys Arg Glu Ala Pro Asn Gln 485 490 495Asp Pro Gly Gly Pro Arg Val Gln Glu Thr Ala Leu Pro Gly Glu Ile 500 505 510Cys Lys Lys Pro Phe Val Thr Leu Glu Ser Ser Glu Glu Asn Leu Leu 515 520 525Glu Gly Cys 53029473PRTDrosophila melanogaster 29Met Leu Arg Leu Pro Thr Ile Leu Arg Lys Ser Phe Ser Met Lys Thr1 5 10 15Arg Gln Arg Leu Asp Tyr Arg Asn Lys Leu Ile Leu Ala Pro Met Val 20 25 30Arg Val Gly Thr Leu Pro Met Arg Leu Leu Ala Leu Glu Met Gly Ala 35 40 45Asp Ile Val Tyr Thr Glu Glu Leu Val Asp Ile Lys Leu Ile Lys Ser 50 55 60Ile Arg Arg Pro Asn Pro Ala Leu Gly Thr Val Asp Phe Val Asp Pro65 70 75 80Ser Asp Gly Thr Ile Val Phe Arg Thr Cys Ala Gln Glu Thr Ser Arg 85 90 95Leu Val Leu Gln Met Gly Thr Ser Asp Ala Gly Arg Ala Leu Ala Val 100 105 110Gly Lys Leu Leu Gln Arg Asp Ile Ser Gly Leu Asp Ile Asn Met Gly 115 120 125Cys Pro Lys Glu Phe Ser Thr Lys Gly Gly Met Gly Ala Ala Leu Leu 130 135 140Ala Asp Pro Asp Lys Ala Ala His Ile Leu Arg Thr Leu Cys Ser Gly145 150 155 160Leu Asp Ile Pro Val Thr Cys Lys Ile Arg Ile Leu Pro Asp Val Glu 165 170 175Gly Thr Ile Asp Leu Val Gln Lys Leu Ala Ala Thr Gly Ile Ala Ala 180 185 190Ile Gly Val His Ala Arg Thr Arg Asp Glu Arg Pro Gln His Pro Ala 195 200 205His Pro Glu Val Leu Arg Ala Val Ala Gln Ala Val Asp Ile Pro Ile 210 215 220Ile Ala Asn Gly Gly Ser Lys Asn Met His Cys Tyr Asp Asp Leu Arg225 230 235 240Lys Phe Gln Met Glu Cys Gly Ala Asp Ser Val Met Val Ala Arg Ala 245 250 255Ala Gln Ile Asn Val Ser Ile Phe Arg Pro Glu Gly Leu Leu Pro Met 260 265 270Asp Glu Leu Ile Glu Lys Tyr Leu Arg Leu Cys Val Asp Tyr Asp Asn 275 280 285Ala Pro His Asn Ala Lys Tyr Cys Val Gln Ser Ile Leu Arg Glu Leu 290 295 300Gln Glu Thr Pro Arg Gly Lys Arg Phe Leu Gln Cys Gln Thr Leu Gln305 310 315 320Gln Ile Cys Glu Ile Trp Glu Leu Gly Asp Tyr Cys Arg Arg Lys Gln 325 330 335Arg Glu Leu Lys Thr Met Gly Asn Ser Gly Arg Ala Glu Val Glu Pro 340 345 350Pro Glu Ala Leu Ala Lys Arg Gln Lys Leu Glu Asp Ala Ala Ile Ala 355 360 365Ile Thr Asp Glu Tyr Asp Gly Ile Ile Cys Arg His Met Pro Phe Leu 370 375 380Arg Ser Thr Tyr Pro Ser Asp Asn His Leu Pro Lys Thr Gln Leu Tyr385 390 395 400Val His Ala Val Lys Thr Gly Lys Ser Pro Pro Ala Tyr Glu Thr Gln 405 410 415Gln Cys Asp Lys Leu Phe Arg Ser Ile Cys Thr Tyr Asp Gly Gln Arg 420 425 430Phe Ser Ser Ser Phe Trp Glu Lys Asn Lys Lys Gln Ala Glu Gln Gly 435 440 445Ala Ala Leu Val Ala Leu Leu His Leu Gly Gln Leu Glu Ala Glu Val 450 455 460Leu Arg Asp Asn Gly Ser Leu Leu Asn465 47030436PRTCaenorhabditis elegans 30Met Ser Asp Leu Tyr Arg Asn Lys Lys Ile Leu Ala Pro Met Val Arg1 5 10 15Ala Gly Arg Thr Pro Leu Arg Leu Leu Cys Leu Lys Tyr Gly Ala Asp 20 25 30Leu Cys Tyr Thr Glu Glu Ile Val Asp Lys Lys Leu Ile Glu Ala Thr 35 40 45Arg Val Val Asn Glu Ala Leu Gly Thr Ile Asp Tyr Arg Asn Gly Asp 50 55 60Asp Ile Ile Leu Arg Leu Ala Pro Glu Glu Lys Gly Arg Cys Ile Leu65 70 75 80Gln Ile Gly Thr Asn Ser Gly Glu Lys Ala Ala Lys Ile Ala Gln Ile 85 90 95Val Gly Asp Asp Val Ala Gly Ile Asp Val Asn Met Gly Cys Pro Lys 100 105 110Pro Phe Ser Ile His Cys Gly Met Gly Ala Ala Leu Leu Thr Gln Thr 115 120 125Glu Lys Ile Val Asp Ile Leu Thr Ser Leu Lys Ser Ala Ala Lys Val 130 135 140Pro Val Thr Cys Lys Ile Arg Val Leu Asp Asp Pro Glu Asp Thr Leu145 150 155 160Lys Leu Val Gln Glu Ile Glu Lys Cys Arg Val Ser Ala Leu Gly Val 165 170 175His Gly Arg Arg Arg Asp Glu Arg Gln Pro Asp Lys Cys Arg Ile Asp 180 185 190Glu Ile Arg Asp Val Ala Gln Ala Val Gln Ser Ile Pro Val Ile Ser 195 200 205Asn Gly Leu Ser Asp Glu Ile Glu Gln Tyr Ser Asp Phe Glu Lys Tyr 210 215 220Gln Leu Leu Thr Glu Thr Ser Ser Thr Met Ile Ala Arg Lys Ala Leu225 230 235 240Ser Thr Pro Ser Ile Phe Arg Arg Glu Gly Cys Leu Asp Lys Tyr Glu 245 250 255Asp Ile Arg Asn Phe Leu Glu Leu Ala Cys Gln Tyr Asp Glu Ser Tyr 260 265 270Thr Met Thr Lys Tyr Val Val Gln Arg Ile Leu Gly Ala Asp Gln Glu 275 280 285Tyr Asp Pro Arg Gly Lys Ala Thr Val Ala Ala Gly Ser Val Leu Gln 290 295 300Ile Cys Lys Ala Phe Gly Met Glu Asp Val Tyr Asp Lys Trp Arg Asp305 310 315 320Glu Arg Lys Lys Lys Gln Ser Lys Lys Arg Ala Arg Val Asp Asp Asp 325 330 335Gly Val Tyr Asn Ile Glu Val Ser Phe Pro Leu Lys Arg Leu Lys Asn 340 345 350Ser Val Gly Phe Ser Pro Thr Pro Lys Met Val Leu His Asp Tyr Cys 355 360 365Val Glu Thr Lys Ile Pro Lys Ala Thr Tyr Glu Val Val Lys Arg Asp 370 375 380Asp Lys Arg Phe Val Ala Thr Ala Cys Ile Gly Asp Lys Lys Tyr Arg385 390 395 400Ser Gly Ile Gly Gln Pro Asn Leu Arg Met Ala Glu Gln Val Ala Ala 405 410 415Leu Ala Ala Leu His Gly Met Asn Ile Arg Asn Leu Leu Val Gly Asn 420 425 430Trp Glu Glu Glu 43531384PRTSaccharomyces cerevisiae 31Met Val Thr Tyr Ala Gly Lys Leu Val Leu Ala Pro Met Val Arg Ala1 5 10 15Gly Glu Leu Pro Thr Arg Leu Met Ala Leu Ala His Gly Ala Asp Leu 20 25 30Val Trp Ser Pro Glu Ile Ile Asp Lys Lys Leu Ile Gln Cys Val Arg 35 40 45Lys Glu Asn Thr Ala Leu Gln Thr Val Asp Tyr Val Val Pro Ser Lys 50 55 60Val Gln Thr Arg Pro Glu Thr Leu Val Phe Arg Thr Tyr Pro Lys Leu65 70 75 80Glu Ser Ser Lys Leu Ile Phe Gln Ile Gly Ser Ala Ser Pro Ala Leu 85 90 95Ala Thr Gln Ala Ala Leu Lys Val Ile Asn Asp Val Ser Gly Ile Asp 100 105 110Val Asn Met Gly Cys Pro Lys His Phe Ser Ile His Ser Gly Met Gly 115 120 125Ser Ala Leu Leu Arg Thr Pro Asp Thr Leu Cys Leu Ile Leu Lys Glu 130 135 140Leu Val Lys Asn Val Gly Asn Pro His Ser Lys Pro Ile Ser Val Lys145 150 155 160Ile Arg Leu Leu Asp Thr Lys Gln Asp Thr Leu Gln Leu Val Lys Arg 165 170 175Leu Cys Ala Thr Gly Ile Thr Asn Leu Thr Val His Cys Arg Lys Thr 180 185 190Glu Met Arg Asn Arg Glu Gln Pro Ile Thr Asp Tyr Ile Ala Glu Ile 195 200 205Tyr Glu Ile Cys Gln Ala Asn Asn Val Ser Leu Ile Val Asn Gly Ala 210 215 220Ile Arg Asp Arg Ser His Phe His Asp Leu Gln Ala Asn His Trp Lys225 230 235 240Asn Thr Asn Ile Gly Gly Met Ile Ala Glu Cys Ala Glu Arg Asp Pro 245 250 255Thr Val Phe Asp His Thr Ser Lys Pro Ser Glu Asp Gly Pro Ser Trp 260 265 270Val Val Ala Cys Arg Glu Phe Ile Gln Trp Ala Thr Lys Phe Asp Asn 275 280 285His Ile Gly Asn Thr Lys Tyr Met Leu Ser Arg Ile Val Pro Gly Lys 290 295 300Ser Val Phe Phe Gln Tyr Phe Ala Arg Cys Lys Ser Pro Glu Glu Val305 310 315 320Ser Phe Val Leu Lys Gln Leu Asn Asp Asp Gly Ser Ala Gln Thr Asp 325 330 335Pro Ser Glu Tyr Leu Glu Asn Cys Arg Ala Gln Glu Lys Ala Leu Lys 340 345 350Asn Ala Asn Ala Ile Ala Lys Gln Lys Arg Lys Gln Thr Asp His Ile 355 360 365Gly Ser Asp Thr Lys Lys Gln Lys Val Val Pro Leu Pro Thr Asp Ile 370 375 38032423PRTSaccharomyces cerevisiae 32Met Thr Glu Pro Ala Leu Ser Ser Ala Asn Asn Ala Leu Met Gln Lys1 5 10 15Leu Thr Gly Arg Gln Leu Phe Asp Lys Ile Gly Arg Pro Thr Arg Ile 20 25 30Val Ala Pro Met Val Asp Gln Ser Glu Leu Ala Trp Arg Ile Leu Ser 35 40 45Arg Arg Tyr Gly Ala Thr Leu Ala Tyr Thr Pro Met Leu His Ala Lys 50 55 60Leu Phe Ala Thr Ser Lys Lys Tyr Arg Glu Asp Asn Trp Ser Ser Leu65 70 75 80Asp Gly Ser Ser Val Asp Arg Pro Leu Val Val Gln Phe Cys Ala Asn 85 90 95Asp Pro Glu Tyr Leu Leu Ala Ala Ala Lys Leu Val Glu Asp Lys Cys 100 105 110Asp Ala Val Asp Leu Asn Leu Gly Cys Pro Gln Gly Ile Ala Lys Lys 115 120 125Gly His Tyr Gly Ser Phe Leu Met Glu Glu Trp Asp Leu Ile His Asn 130 135 140Leu Ile Asn Thr Leu His Lys Asn Leu Lys Val Pro Val Thr Ala Lys145 150 155 160Ile Arg Ile Phe Asp Asp Cys Glu Lys Ser Leu Asn Tyr Ala Lys Met 165 170 175Val Leu Asp Ala Gly Ala Gln Phe Leu Thr Val His Gly Arg Val Arg 180 185 190Glu Gln Lys Gly Gln Lys Thr Gly Leu Ala Asn Trp Glu Thr Ile Lys 195 200 205Tyr Leu Arg Asp Asn Leu Pro Lys Glu Thr Val Phe Phe Ala Asn Gly 210 215 220Asn Ile Leu Tyr Pro Glu Asp Ile Ser Arg Cys Met Glu His Ile Gly225 230 235 240Ala Asp Ala Val Met Ser Ala Glu Gly Asn Leu Tyr Asn Pro Gly Val 245 250 255Phe Asn Val Gly Gln Thr Lys Asn Lys Glu Lys Ile Phe Pro Arg Val 260 265 270Asp Lys Ile Ile Arg Glu Tyr Phe Gln Ile Val Lys Glu Cys Gln Glu 275 280 285Ser Lys Ala Ser Lys Thr Ala Met Lys Ser His Phe Phe Lys Ile Leu 290 295 300Arg Pro Phe Leu Pro His His Thr Asp Ile Arg Ser Thr Leu Ala Thr305 310 315 320Met Asn Ala Lys Ala Thr Trp Glu Glu Trp Glu Glu Gln Val Val Lys 325 330 335Pro Val Glu Lys Val Val Gln Glu Ile Phe Glu Gln Pro Asp Ile Ala 340 345 350Ile Lys Asp Glu Ile Thr Ile Gly Glu Lys Gln Ser Trp Gly Gly Ser 355 360 365Tyr Arg Thr Val Pro Tyr Trp Arg Cys Gln Pro Tyr Phe Arg Pro Val 370 375 380Asn Gly Ile Thr Gly Asp Lys Arg Val Met Gln Gly Leu Ile Asp Glu385 390 395 400Ser Val Asn Lys Lys Arg Lys Ala Asp Val Pro Leu Glu Ser Ala Asp 405 410 415Lys Lys Lys Asp Val Lys Ala 4203310PRTArtificial Sequencec-Myc-His epitope tag 33Leu Asp Glu Glu Ser Ile Leu Lys Gln Glu1 5 10

* * * * *

References


uspto.report is an independent third-party trademark research tool that is not affiliated, endorsed, or sponsored by the United States Patent and Trademark Office (USPTO) or any other governmental organization. The information provided by uspto.report is based on publicly available data at the time of writing and is intended for informational purposes only.

While we strive to provide accurate and up-to-date information, we do not guarantee the accuracy, completeness, reliability, or suitability of the information displayed on this site. The use of this site is at your own risk. Any reliance you place on such information is therefore strictly at your own risk.

All official trademark data, including owner information, should be verified by visiting the official USPTO website at www.uspto.gov. This site is not intended to replace professional legal advice and should not be used as a substitute for consulting with a legal professional who is knowledgeable about trademark law.

© 2024 USPTO.report | Privacy Policy | Resources | RSS Feed of Trademarks | Trademark Filings Twitter Feed