WO2010058572A1 - Procédés pour le diagnostic ou le traitement d'un cancer de la prostate - Google Patents

Procédés pour le diagnostic ou le traitement d'un cancer de la prostate Download PDF

Info

Publication number
WO2010058572A1
WO2010058572A1 PCT/JP2009/006201 JP2009006201W WO2010058572A1 WO 2010058572 A1 WO2010058572 A1 WO 2010058572A1 JP 2009006201 W JP2009006201 W JP 2009006201W WO 2010058572 A1 WO2010058572 A1 WO 2010058572A1
Authority
WO
WIPO (PCT)
Prior art keywords
stc2
prostate cancer
gene
double
polypeptide
Prior art date
Application number
PCT/JP2009/006201
Other languages
English (en)
Inventor
Yusuke Nakamura
Hidewaki Nakagawa
Takuya Tsunoda
Original Assignee
Oncotherapy Science, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Oncotherapy Science, Inc. filed Critical Oncotherapy Science, Inc.
Priority to US13/130,284 priority Critical patent/US8420329B2/en
Priority to JP2011522180A priority patent/JP2012509058A/ja
Priority to EP09827353A priority patent/EP2367939A4/fr
Publication of WO2010058572A1 publication Critical patent/WO2010058572A1/fr
Priority to US13/830,668 priority patent/US20130302799A1/en

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6876Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
    • C12Q1/6883Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material
    • C12Q1/6886Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material for cancer
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/08Drugs for disorders of the urinary system of the prostate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • C12N15/1136Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing against growth factors, growth regulators, cytokines, lymphokines or hormones
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • G01N33/57407Specifically defined cancers
    • G01N33/57434Specifically defined cancers of prostate
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/10Type of nucleic acid
    • C12N2310/14Type of nucleic acid interfering N.A.
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/50Physical structure
    • C12N2310/53Physical structure partially self-complementary or closed
    • C12N2310/531Stem-loop; Hairpin
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/112Disease subtyping, staging or classification
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/136Screening for pharmacological compounds
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2500/00Screening for compounds of potential therapeutic value
    • G01N2500/04Screening involving studying the effect of compounds C directly on molecule A (e.g. C are potential ligands for a receptor A, or potential substrates for an enzyme A)

Definitions

  • the present invention relates to the field of biological science, more specifically to the field of cancer diagnosis and treatment.
  • the present invention relates to methods for detecting and diagnosing prostate cancer as well as methods for treating and preventing prostate cancer.
  • the present invention relates to methods for screening for a candidate compound for treating and/or preventing prostate cancer.
  • the present invention further relates to double-stranded molecules that reduce or inhibit the expression of STC2 and uses thereof.
  • PC Prostate cancer
  • Surgical and radiation therapies are effective to the localized disease, but nearly 30% of treated PC patients still suffer from the relapse of the disease (NPL 3:Feldman BJ and Feldman D, Nat Rev Cancer 2001, 1, 34-45, NPL 4:Scher HI and Sawyers CL, J Clin Oncol 2006, 23, 8253-61, and NPL 5:Han M, et al., J Urol 2001, 166, 416-9).
  • Most of the patients with relapsed or advanced disease respond well to androgen-ablation therapy (medical or surgical castration) because PCs are usually androgen-dependent at a relatively early stage. However, they often acquire castration-resistant phenotype that progresses aggressively and ultimately leads to the death of PC patients.
  • an objective of the present invention is to provide a new biomarker and new therapeutic strategies against PCs, in particular, CRPCs and aggressive PCs with high Gleason score.
  • STC2 molecular target
  • Stanniocalcin was first identified as a glycoprotein hormone secreted from specific endocrine glands (corpuscle of Stannius) in the kidney region of bony fish that is involved in calcium and phosphate homeostasis (NPL 9:Wagner GF, et al., Mol Cell Endocrinol 1991, 79, 129-38, and NPL 10:Wagner GF and Jaworski E, Mol Cell Endocrinol 1994, 99, 315-22).
  • NPL 9 Wagner GF, et al., Mol Cell Endocrinol 1991, 79, 129-38, and NPL 10:Wagner GF and Jaworski E, Mol Cell Endocrinol 1994, 99, 315-22).
  • STC1 and STC2 are related mammalian genes that are predicted to be secreted glycosylated proteins (NPL 11:Chang AC, et al., Mol Cell Endocrinol 1995, 112, 241-7, and NPL 12:Ishibashi K, et al. Biochem Biophys Res Commun 1998, 250, 252-8).
  • NPL 11:Chang AC et al.
  • Mol Cell Endocrinol 1995, 112, 241-7 et al.
  • NPL 12:Ishibashi K et al. Biochem Biophys Res Commun 1998, 250, 252-8.
  • their physiological or pathological functions in human beings and human cancers have not been clearly elucidated, and their receptor binding partners have not yet identified.
  • the present invention relates to the identification of STC2 as a molecular target useful in PC treatment and diagnosis.
  • the STC2 protein finds particular utility as a molecular target for development of novel treatments for PC.
  • Preferred PCs targeted by the prevent invention are CRPC and aggressive PC.
  • a subject-derived biological sample such as tissue, blood, or serum.
  • An increase in the level of expression of the gene as compared to a normal control level indicates that the subject suffers from or is at risk of developing PC.
  • the normal control level may be the expression level of the STC2 gene detected in a normal healthy tissue, blood, or serum of an individual or population known to be free from PC.
  • expression level of the STC2 gene in a sample can be compared to a cancerous control level of the STC2 gene.
  • a similarity between the expression level of a sample and the cancerous control level indicates that the subject suffers from or is at risk of developing PC.
  • the cancerous control level may be the expression level of the STC2 gene detected in the cancerous tissue, blood, or serum of an individual or population known to be suffering from PC. It is another object of the present invention to provide a kit for diagnosing PC, such a kit minimally containing a reagent that measures the expression level of the STC2 gene in a subject-derived biological sample.
  • the test agent binding the polypeptide or suppressing a biological activity of the polypeptide may be used to reduce symptoms of PC.
  • the present invention also provides methods for identifying agents that inhibit the expression or activity of the STC2 protein, such method involving the steps of contacting a cell expressing the STC2 protein with a test agent and determining the expression level of the STC2 gene or the activity of the gene product, the STC2 protein.
  • a decrease in the expression level of the gene or an activity of its gene product as compared to a control level in the absence of the test compound indicates that the test compound may be used to reduce symptoms of PC.
  • the present invention provides methods for identifying agents that inhibit the transcription of the STC2 gene, such methods involving the steps of contacting the cell introduced with a vector that contains the transcriptional regulatory region of the STC2 gene and a reporter gene expressed under the control of the transcriptional regulatory region and determining the expression level or activity of the reporter gene.
  • a decrease in the expression level of the gene or an activity of its gene product as compared to a control level in the absence of the test compound indicates that the test compound may be used to reduce symptoms of PC.
  • the double-stranded molecules include a sense strand and an antisense strand complementary thereto, hybridized to each other to form the double-stranded molecule and inhibit expression of the gene and cell proliferation when introduced into a cell expressing the STC2 gene.
  • the sense strand includes the sequence corresponding to a target sequence of SEQ ID NO:8 or 9.
  • Another aspect of the present invention relates to compositions for treating PC containing at least one of those siRNAs that inhibits cell proliferation of a cell expressing STC2 gene and methods for treating or preventing PC that includes the step of administering to a subject those compositions.
  • the antisense polynucleotide or siRNA can be provided as a vector expressing those.
  • One advantage of the methods and kits described herein is that the disease may be identified prior to detection of overt clinical symptoms of PC.
  • Another advantage of the methods and compositions described herein is that novel therapeutic approach with fewer adverse effect may be provided.
  • Fig. 1 demonstrates the over-expression of STC2 in CRPC cells and immunohistochemical analysis in PC tissues.
  • Part (A) depicts the results of semi-quantitative RT-PCR validating STC2 over-expression in the microdissected CRPC cells as compared to CNPC cells and normal prostatic epithelial (NP) cells, which were also microdissected.
  • ACTB was used to quantify the each of cDNA contents.
  • Part (B) depicts the results of real-time quantitative RT-PCR demonstrating over-expression of STC2 transcript in CRPC cells (samples 2-8) as compared to that of CNPC cells (samples 9-15) and NP cells (sample 1).
  • ACTB was used to quantify each of the cDNA contents, and the relative quantity (Y-axis) was calculated so that the expression in NP cells was one.
  • Real-time quantitative RT-PCR was carried out duplicated for each sample (white and black columns).
  • Part (C) depicts the results of Northern blot analysis demonstrating a high level of STC2 expression in five PC cell lines (lane 1-5), as compared to adult normal organs, including brain, heart, kidney, liver, lung, prostate and testis (lanes 6-12) wherein its expression is barely detectable.
  • Part (D) depicts the results of multiple tissue Northern blot analysis demonstrating that STC2 was expressed only in normal pancreas, with no or very low expression of STC2 in adult normal organs.
  • Parts (E)-(H) demonstrate the immunoreactivity with anti-STC2 antibody observed in CRPC and CNPC with Gleason score 10 tissues examined, with strong positive immunostaining exhibited in the cytoplasm of PC cells.
  • Part (E) is a representative picture of CRPC (x200)
  • part (F) is a representative picture of CNPC with Gleason score 10 (x200)
  • part (G) is a representative picture of CNPC with Gleason score 7 (x200).
  • Adjacent normal prostatic epithelium revealed very weak or no signal for STC2 (F and G; arrowheads).
  • Part (H) depicts the relationship between STC2 immunohistochemical score and Gleason score in CNPCs. (*p ⁇ 0.003, Mann-Whitney's U-test with Bonferroni method).
  • Fig. 2 demonstrates the knockdown of STC2 expression by siRNA attenuated PC cell viability and the promotion of cancer cell growth by STC2 over-expression.
  • Part (A) depicts the knockdown effect of siRNA on STC2 in PC-3 cells. Semi-quantitative RT-PCR was performed using cells transfected with each of shRNA-expressing vectors to STC2 (si1-3) as well as a negative control vector (siEGFP). ACTB was used to quantify RNAs.
  • Part (B) depicts the results of a colony formation assay of PC-3 cells transfected with each of indicated shRNA-expressing vectors to STC2 (si1-3) and a negative control vector (siEGFP).
  • Part (C) depicts the results of MTT assay of each of PC-3 cells transfected with indicated siRNA-expressing vectors to STC2 (si1-3) and a negative control vector (siEGFP). Each average is plotted with error bars indicating SD (standard deviation) after 10-day incubation with Geneticin.
  • ABS on Y-axis means absorbance at 490 nm, and at 630 nm as reference, measured with a microplate reader.
  • Part (D) depicts the results of Western blot analysis with anti-HA-tag antibody, demonstrating that three stable transformats (Clones 1-3) constitutively expressed an exogenous STC2. Mock was the 22Rv1-mock clone mixture. CBB stain served as a loading control.
  • Part (E) depicts the in-vitro growth curve calculated by MTT assay, showing that three stable transformants (Clones 1-3) grew more rapidly than the 22Rv1-mock clone mixture (*P ⁇ 0.01, **P ⁇ 0.05, Students' t-test).
  • ABS on Y-axis means absorbance at 490 nm, and at 630 nm as reference, measured with a microplate reader.
  • biological sample refers to a whole organism or a subset of its tissues, cells or component parts (e.g., body fluids, including but not limited to blood, mucus, lymphatic fluid, synovial fluid, cerebrospinal fluid, saliva, amniotic fluid, amniotic cord blood, urine and semen).
  • body fluids including but not limited to blood, mucus, lymphatic fluid, synovial fluid, cerebrospinal fluid, saliva, amniotic fluid, amniotic cord blood, urine and semen).
  • Biological sample further refers to a homogenate, lysate, extract, cell culture or tissue culture prepared from a whole organism or a subset of its cells, tissues or component parts, or a fraction or portion thereof.
  • biological sample refers to a medium, such as a nutrient broth or gel in which an organism has been propagated, which contains cellular components, such as proteins or polynucleotides.
  • CRPC ration-resistant prostate cancer
  • RPC hormone-refractory prostate cancers
  • isolated and purified used in relation with a substance indicates that the substance is substantially free from at least one substance that may else be included in the natural source.
  • an isolated or purified antibody refers to antibodies that are substantially free of cellular material such as carbohydrate, lipid, or other contaminating proteins from the cell or tissue source from which the protein (antibody) is derived, or substantially free of chemical precursors or other chemicals when chemically synthesized.
  • substantially free of cellular material includes preparations of a polypeptide in which the polypeptide is separated from cellular components of the cells from which it is isolated or recombinantly produced.
  • a polypeptide that is substantially free of cellular material includes preparations of polypeptide having less than about 30%, 20%, 10%, or 5% (by dry weight) of heterologous protein (also referred to herein as a "contaminating protein").
  • heterologous protein also referred to herein as a "contaminating protein”
  • the polypeptide is recombinantly produced, it is also preferably substantially free of culture medium, which includes preparations of polypeptide with culture medium less than about 20%, 10%, or 5% of the volume of the protein preparation.
  • the polypeptide is produced by chemical synthesis, it is preferably substantially free of chemical precursors or other chemicals, which includes preparations of polypeptide with chemical precursors or other chemicals involved in the synthesis of the protein less than about 30%, 20%, 10%, 5% (by dry weight) of the volume of the protein preparation.
  • That a particular protein preparation contains an isolated or purified polypeptide can be shown, for example, by the appearance of a single band following sodium dodecyl sulfate (SDS)-polyacrylamide gel electrophoresis of the protein preparation and Coomassie Brilliant Blue staining or the like of the gel.
  • SDS sodium dodecyl sulfate
  • antibodies and polypeptides of the present invention are isolated or purified.
  • An "isolated" or “purified” nucleic acid molecule, such as a cDNA molecule can be substantially free of other cellular material, or culture medium when produced by recombinant techniques, or substantially free of chemical precursors or other chemicals when chemically synthesized.
  • polypeptide polypeptide
  • peptide protein
  • protein polymer of amino acid residues.
  • the terms apply to amino acid polymers in which one or more amino acid residue is a modified residue, or a non-naturally occurring residue, such as an artificial chemical mimetic of a corresponding naturally occurring amino acid, as well as to naturally occurring amino acid polymers.
  • amino acid refers to naturally occurring and synthetic amino acids, as well as amino acid analogs and amino acid mimetics that similarly functions to the naturally occurring amino acids.
  • Naturally occurring amino acids are those encoded by the genetic code, as well as those modified after translation in cells (e.g., hydroxyproline, gamma-carboxyglutamate, and O-phosphoserine).
  • amino acid analog refers to compounds that have the same basic chemical structure (an alpha carbon bound to a hydrogen, a carboxy group, an amino group, and an R group) as a naturally occurring amino acid but have a modified R group or modified backbones (e.g., homoserine, norleucine, methionine, sulfoxide, methionine methyl sulfonium).
  • modified R group or modified backbones e.g., homoserine, norleucine, methionine, sulfoxide, methionine methyl sulfonium.
  • amino acid mimetic refers to chemical compounds that have different structures but similar functions to general amino acids. Amino acids may be referred to herein by their commonly known three letter symbols or the one-letter symbols recommended by the IUPAC-IUB Biochemical Nomenclature Commission.
  • nucleic acid molecules are used interchangeably unless otherwise specifically indicated and are similarly to the amino acids referred to by their commonly accepted single-letter codes. Similar to the amino acids, they encompass both naturally-occurring and non-naturally occurring nucleic acid polymers.
  • the polynucleotide, oligonucleotide, nucleotides, nucleic acids, or nucleic acid molecules may be composed of DNA, RNA or a combination thereof.
  • cancer refers to cancers over-expressing the STC2 gene, more particularly prostate cancers such as CRPC, more particularly pancreatic ductal adenocarcinoma and castration-resistant prostate cancer.
  • the STC2 gene or STC2 protein The present invention is based in part on the discovery of elevated expression of the STC2 gene in cells from patients of PCs. The expression of the gene was discovered to be particularly elevated in CRPC.
  • An exemplified nucleotide sequence of human STC2 gene is shown in SEQ ID NO: 11 and is also available as GenBank Accession No. NM_003714. Another nucleotide sequence data for the human STC2 gene is available as GenBank Accession No. AK075406.
  • the STC2 gene encompasses the human STC2 gene as well as those of other animals including non-human primate, mouse, rat, dog, cat, horse, and cow but are not limited thereto, and includes allelic mutants and genes found in other animals as corresponding to the STC2 gene.
  • STC2 An exemplified amino acid sequence encoded the human STC2 gene is shown in SEQ ID NO: 12 and is also available as GenBank Accession No. NP_003705.
  • STC2 the polypeptide encoded by the STC2 gene is referred to as "STC2", and sometimes as “STC2 polypeptide” or "STC2 protein".
  • functional equivalents are also included in the STC2 polypeptide.
  • a "functional equivalent" of a protein is a polypeptide that has a biological activity equivalent to the protein. Namely, any polypeptide that retains the biological ability of the STC2 protein may be used as such a functional equivalent in the present invention.
  • polypeptide may be one that includes an amino acid sequence having at least about 80% homology (also referred to as sequence identity) to the sequence of the respective proteins, more preferably at least about 90% to 95% homology, even more preferably 96% to 99% homology.
  • polypeptide can be encoded by a polynucleotide that hybridizes under stringent conditions to the naturally occurring nucleotide sequence of the STC2 gene.
  • stringent (hybridization) conditions refers to conditions under which a nucleic acid molecule will hybridize to its target sequence, typically in a complex mixture of nucleic acids, but not detectably to other sequences. Stringent conditions are sequence-dependent and will vary in different circumstances. Longer sequences hybridize specifically at higher temperatures. An extensive guide to the hybridization of nucleic acids is found in Tijssen, Techniques in Biochemistry and Molecular Biology-Hybridization with Nucleic Probes, "Overview of principles of hybridization and the strategy of nucleic acid assays” (1993). Generally, stringent conditions are selected to be about 5-10 degrees C lower than the thermal melting point (T m ) for the specific sequence at a defined ionic strength pH.
  • T m thermal melting point
  • the T m is the temperature (under defined ionic strength, pH, and nucleic concentration) at which 50% of the probes complementary to the target hybridize to the target sequence at equilibrium (as the target sequences are present in excess, at T m , 50% of the probes are occupied at equilibrium).
  • Stringent conditions may also be achieved with the addition of destabilizing agents such as formamide.
  • a positive signal is at least two times of background, preferably 10 times of background hybridization.
  • Exemplary stringent hybridization conditions can be as following: 50% formamide, 5x SSC, and 1% SDS, incubating at 42 degrees C, or, 5x SSC, 1% SDS, incubating at 65 degrees C, with wash in 0.2x SSC, and 0.1% SDS at 50 degrees C.
  • a condition of hybridization for isolating a DNA encoding a polypeptide functionally equivalent to the human STC2 protein can be routinely selected by a person skilled in the art.
  • hybridization may be performed by conducting pre-hybridization at 68 degrees C for 30 min or longer using "Rapid-hyb buffer" (Amersham LIFE SCIENCE), adding a labeled probe, and warming at 68 degrees C for 1 hour or longer.
  • the following washing step can be conducted, for example, in a low stringent condition.
  • An exemplary low stringent condition may include 42 degrees C, 2x SSC, 0.1% SDS, preferably 50 degrees C, 2x SSC, 0.1% SDS. High stringency conditions are often preferably used.
  • An exemplary high stringency condition may include washing 3 times in 2x SSC, 0.01% SDS at room temperature for 20 min, then washing 3 times in 1x SSC, 0.1% SDS at 37 degrees C for 20 min, and washing twice in 1x SSC, 0.1% SDS at 50 degrees C for 20 min.
  • 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.
  • mutated or modified proteins i.e., peptides composed of an amino acid sequence in which one, two, or several amino acid residues have been modified through substitution, deletion, insertion and/or addition
  • mutated or modified proteins have been known to retain the original biological activity (Mark et al., Proc Natl Acad Sci USA 81: 5662-6 (1984); Zoller and Smith, Nucleic Acids Res 10:6487-500 (1982); Dalbadie-McFarland et al., Proc Natl Acad Sci USA 79: 6409-13 (1982)).
  • the peptides of the present invention may have an amino acid sequence wherein one, two or even more amino acids are added, inserted, deleted, and/or substituted in the STC2 sequence. So long as the activity the protein is maintained, the number of amino acid mutations is not particularly limited. However, it is generally preferred to alter 5% or less of the amino acid sequence.
  • the number of amino acids to be mutated in such a mutant is generally 30 amino acids or less, preferably 20 amino acids or less, more preferably 10 amino acids or less, more preferably 5 or 6 amino acids or less, and even more preferably 3 or 4 amino acids or less.
  • An 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 as conservative amino acid substitution).
  • 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).
  • A, I, L, M, F, P, W, Y, V hydrophilic
  • Conservative substitution tables providing functionally similar amino acids are well known in the art. For example, the following eight groups each contain amino acids that are conservative substitutions for one another: 1) Alanine (A), Glycine (G); 2) Aspartic acid (d), Glutamic acid (E); 3) Aspargine (N), Glutamine (Q); 4) Arginine (R), Lysine (K); 5) Isoleucine (I), Leucine (L), Methionine (M), Valine (V); 6) Phenylalanine (F), Tyrosine (Y), Tryptophan (W); 7) Serine (S), Threonine (T); and 8) Cystein (C), Methionine (M) (see, e.g., Creighton, Proteins 1984).
  • Such conservatively modified polypeptides are included in the present STC2 protein.
  • the present invention is not restricted thereto and the STC2 protein includes non-conservative modifications so long as they retain at least one biological activity of the STC2 protein.
  • the modified proteins do not exclude polymorphic variants, interspecies homologues, and those encoded by alleles of these proteins.
  • the STC2 gene of the present invention encompasses polynucleotides that encode such functional equivalents of the STC2 protein.
  • a gene amplification method for example, the polymerase chain reaction (PCR) method, can be utilized to isolate a polynucleotide encoding a polypeptide functionally equivalent to the STC2 protein, using a primer synthesized based on the sequence information of the protein encoding DNA.
  • PCR polymerase chain reaction
  • High homology typically refers to a homology of 40% or higher, preferably 60% or higher, more preferably 80% or higher, even more preferably 90% to 95% or higher, even more preferably 96% to 99% or higher.
  • the homology of a particular polynucleotide or polypeptide can be determined by following the algorithm in "Wilbur and Lipman, Proc Natl Acad Sci USA 80: 726-30 (1983)".
  • I. Diagnosing cancer I-1. Method for diagnosing cancer or a predisposition for developing cancer
  • the expression of the STC2 gene was found to be specifically elevated in patients with PC, more particularly, in CRPC and aggressive PC (see Example 2).
  • the STC2 genes identified herein as well as their transcription and translation products find diagnostic utility as a marker for PC and by measuring the expression of the STC2 gene in a subject-derived biological sample, PC can be diagnosed.
  • the present invention provides a method for detecting, diagnosing and/or determining the presence of or a predisposition for developing cancer, more particulary PC in a subject by determining the expression level of the STC2 gene in the subject.
  • the term "diagnosing" is intended to encompass predictions and likelihood analysis.
  • the present method is intended to be used clinically in making decisions concerning treatment modalities, including therapeutic intervention, diagnostic criteria such as disease stages, and disease monitoring and surveillance for cancer.
  • an intermediate result for examining the condition of a subject may also be provided. Such intermediate result may be combined with additional information to assist a doctor, nurse, or other practitioner to diagnose that a subject suffers from the disease.
  • the present invention may be used to detect cancerous cells in a subject-derived tissue or biological sample and provide a doctor with useful information to diagnose that the subject suffers from the disease.
  • a subject to be diagnosed by the present method is preferably a mammal.
  • exemplary mammals include, but are not limited to, human, non-human primate, mouse, rat, dog, cat, horse, and cow. It is preferred to collect a biological sample from a subject to be diagnosed to perform the diagnosis. Any biological material can be used as the biological sample for the determination so long as it contains the objective transcription or translation product of the STC2 gene.
  • the biological samples include, but are not limited to, bodily tissues, especially some of a prostatic tissue and bodily fluids, such as blood, sputum, semen, prostatic fluid and urine.
  • the biological sample contains a cell population including an epithelial cell, more preferably a prostate epithelial cell derived from tissue suspected to be cancerous.
  • the expression level of the STC2 gene is determined in a subject-derived biological sample.
  • the expression level can be determined at the transcription (nucleic acid) product level, using methods known in the art.
  • the mRNA of the STC2 gene may be quantified using a probe by hybridization methods (e.g., Northern hybridization).
  • the detection may be carried out on a chip or an array. The use of an array is preferable for detecting the expression level of a plurarity of genes (e.g., various cancer specific genes) including the present STC2 gene.
  • probes utilizing the sequence information of the STC2 gene (for example, the sequence shown in SEQ ID NO: 11; GenBank Accession No. NM_018423 or GenBank Accession No. AK075406).
  • the cDNA of the STC2 gene may be used as the probes.
  • the probe may be labeled with a suitable label, such as dyes and isotopes, and the expression level of the gene may be detected as the intensity of the hybridized labels.
  • the transcription product of the STC2 gene may be quantified using primers by amplification-based detection methods (e.g., RT-PCR).
  • primers can also be prepared based on an available sequence information of the gene.
  • the primers used in Example 1 SEQ ID NOs: 3 and 4 may be employed for the detection by RT-PCR, but the present invention is not restricted thereto.
  • a probe or primer used for the present method hybridizes under stringent, moderately stringent, or low stringent conditions to the mRNA of the STC2 gene.
  • stringent (hybridization) conditions refers to conditions under which a probe or primer will hybridize to its target sequence, but to no other sequences.
  • the temperature of a stringent condition is selected to be about 5 degrees C lower than the thermal melting point (T m ) for a specific sequence at a defined ionic strength and pH.
  • T m is the temperature (under defined ionic strength, pH and nucleic acid concentration) at which 50% of the probes complementary to the target sequence hybridize to the target sequence at equilibrium. Since the target sequences are generally present at excess, at Tm, 50% of the probes are occupied at equilibrium.
  • stringent conditions will be those in which the salt concentration is less than about 1.0 M sodium ion, typically about 0.01 to 1.0 M sodium ion (or other salts) at pH 7.0 to 8.3 and the temperature is at least about 30 degrees C for short probes or primers (e.g., 10 to 50 nucleotides) and at least about 60 degrees C for longer probes or primers.
  • Stringent conditions may also be achieved with the addition of destabilizing agents, such as formamide.
  • the translation product may be detected for the diagnosis of the present invention.
  • the quantity of the STC2 protein may be determined.
  • a method for determining the quantity of the protein as the translation product includes immunoassay methods that use an antibody specifically recognizing the protein.
  • the antibody may be monoclonal or polyclonal.
  • any fragment or modification (e.g., chimeric antibody, scFv, Fab, F(ab') 2 , Fv, etc.) of the antibody may be used for the detection, so long as the fragment retains the binding ability to the STC2 protein.
  • Methods to prepare these kinds of antibodies for the detection of proteins are well known in the art, and any method may be employed in the present invention to prepare such antibodies and equivalents thereof.
  • the intensity of staining may be observed via immunohistochemical analysis using an antibody against the STC2 protein. Namely, the observation of strong staining indicates increased presence of the protein and at the same time high expression level of the STC2 gene.
  • STC2 protein is secreted protein. Therefore, by measuring the level of STC2 in subject-derived blood samples, the occurrence of or a predisposition to develop prostate cancer expressing STC2 in a subject can be determined.
  • any blood samples may be used for determining the level of STC2 so long as either the gene or the protein of STC2 can be detected in the samples.
  • the blood samples include whole blood, serum, and plasma. The method of determining the level of STC2 was known to those skilled in the art, like ELISA, especially sandwich method.
  • the translation product may be detected based on its biological activity.
  • the STC2 gene have been isolated as one of the mammalian gene related to STC gene that has been known to regulate calcium and phosphate uptake in different target organs (Wagner GF, et al., Mol Cell Endocrinol 1991, 79, 129-38, and Wagner GF and Jaworski E, Mol Cell Endocrinol 1994, 99, 315-22) and it was reported that the STC2 protein inhibited the phosphate uptake of a kidney cell line (Ishibashi K, et al., Biochem Biophys Res Commun 1998, Sep 18;250(2), 258-8).
  • the translation product of STC2 gene has an ability to promote PC cell growth.
  • the inhibiting activity against the phosphate uptake of a kidney cell line or the cancer cell growth promoting activity may be used as an index of the STC2 protein existing in the biological sample.
  • the expression level of other cancer-associated genes for example, genes known to be differentially expressed in CRPC, may also be determined to improve the accuracy of the diagnosis.
  • the expression level of the STC2 gene in a biological sample can be considered to be increased if it increases from the control level of the STC2 gene by, for example, 10%, 25%, or 50%; or increases to more than 1.1 fold, more than 1.5 fold, more than 2.0 fold, more than 5.0 fold, more than 10.0 fold, or more.
  • the control level may be determined at the same time with the test biological sample by using a sample(s) previously collected and stored from a subject/subjects whose disease state (cancerous or non-cancerous) is/are known. Alternatively, the control level may be determined by a statistical method based on the results obtained by analyzing previously determined expression level(s) of the STC2 gene in samples from subjects whose disease state are known.
  • control level can be a database of expression patterns from previously tested cells.
  • the expression level of the STC2 gene in a biological sample may be compared to multiple control levels, which control levels are determined from multiple reference samples. It is preferred to use a control level determined from a reference sample derived from a tissue type similar to that of the patient-derived biological sample.
  • control level determined from a reference sample derived from a tissue type similar to that of the patient-derived biological sample.
  • control level determined from a biological sample that is known to be non-cancerous or cancer-free is called “normal control level”.
  • control level is determined from a cancerous biological sample, it will be called “cancerous control level”.
  • the subject When the expression level of the STC2 gene in a biological sample derived from a subject is increased compared to the normal control level or is similar to the cancerous control level, the subject may be diagnosed to be suffering from or at a risk of developing cancer. Furthermore, in the case where the expression levels of multiple cancer-related genes are compared, a similarity in the gene expression pattern between the sample and the reference that is cancerous indicates that the subject is suffering from or at a risk of developing cancer. Difference between the expression levels of a test biological sample and the control level can be normalized to the expression level of control nucleic acids, e.g. housekeeping genes whose expression levels are known not to differ depending on the cancerous or non-cancerous state of the cell.
  • control nucleic acids e.g. housekeeping genes whose expression levels are known not to differ depending on the cancerous or non-cancerous state of the cell.
  • Exemplary housekeeping genes include, but are not limited to, beta actin, glyceraldehyde 3 phosphate dehydrogenase, and ribosomal protein P1.
  • the present invention provides a method for detecting or identifying prostate cancer cells in a subject-derived prostate tissue sample, said method including the step of determining the expression level of the STC2 gene in a subject-derived biological sample, wherein an increase in said expression level as compared to a normal control level of said gene indicates the presence or suspicion of cancer cells in the tissue.
  • the present invention may provide a doctor with useful information to diagnose a subject as afflicted with the disease.
  • the present invention when there is doubt regarding the presence of prostate cancer cells in the tissue obtained from a subject, clinical decisions can be reached by considering the expression level of the STC2 gene, plus a different aspect of the disease including tissue pathology, levels of known tumor marker(s) in blood, and clinical course of the subject, etc.
  • some well-known diagnostic prostate tumor markers in blood is PSA.
  • the outcome of the gene expression analysis serves as an intermediate result for further diagnosis of a subject's disease state.
  • the present invention provides a method for detecting a diagnostic marker of prostate cancer, said method including the step of detecting the expression of the STC2 gene in a subject-derived biological sample as a diagnostic marker of prostate cancer.
  • prostate cancer cells can be diagnosed or detected.
  • the presence of CRPC or aggressive prostate cancer cells within a prostate tissue may be diagnosed or detected.
  • aggressive prostate cancer refers to prostate cancer having Gleason score (Gleason tumor grade) of 8 or more.
  • Gleason score 8-10 is considered to be aggressive prostate cancer.
  • the present invention reveals that the immunostaining intensity of the STC2 polypeptide in a biological sample derived from a prostate cancer patient is increased as compared to the control level. Therefore, the present invention also provides a method of correlating the immunostaining intensity of the STC2 polypeptide with the presence of prostate cancer including the steps as follows: (a) determining or evaluating the immunostaining intensity of the STC2 polypeptide in a biological sample derived from a subject; and (b) correlating the immunostaining intensity of step (a) with the presence of prostate cancer.
  • the present invention provides a method of correlating the immunostaining intensity of the STC2 polypeptide with the presence of prostate cancer including the steps as follows: (a) determining or evaluating the immunostaining intensity of the STC2 polypeptide in a biological sample derived from a subject; (b) comparing the immunostaining intensity of step (a) with a control level; and (c) correlating an increase in the immunostaining intensity determined in step (b) as compared to the control level to the presence of prostate cancer.
  • a biological sample may be fixed chemically or physically.
  • the formalin fixed biological sample is preferable for evaluating the immunostaining intensity of the STC2 polypeptide
  • the biological sample can be fixed with a glutaraldehyde or osmium teraoxide.
  • a tissue derived from a subject is preferable as the biological sample.
  • a tissue in the present invention is a prostate tissue, more preferably prostate epithelial tissue.
  • sections may be prepared from tissue samples.
  • Methods for preparing the tissue section are well known in the art. For example, methods for preparing a frozen section, unfrozen section, or paraffin section are known.
  • prostate cancer of the present invention is CRPC, CNPC, or an aggressive prostate cancer, more preferably CRPC, CNPC, or an aggressive prostate cancer with Gleason score 8-10.
  • Immunostaining intensity can be determined or evaluated with a labeled anti-STC2 antibody.
  • anti-STC2 antibody may be labeled with enzymes.
  • the following enzymes are suitable for the anti-STC2 antibody labeling; alkaline phosphatase, horseradish peroxidase, beta-galactosidase, or beta-glucosidase.
  • the anti-STC2 antibody is labeled with an enzyme, the antibody can be detected or measured with a substrate which reacts with the enzyme to generate detectable signals. Substrates generating detectable signals including color change, accumulation of dyes, or pigments having color fluorescence, or luminescence are well known.
  • detectable signals may be observed with a light microscope or fluorescence microscope.
  • the antibody may be labeled with fluorescent substances.
  • the antibody bound to the biological sample may be detected or measured by a fluorescence microscope.
  • fluorescein isothiocyanate (FITC), or rhodamine is suitable as the label in the present invention.
  • the antibody may also be labeled with a gold colloid.
  • the bound antibody may be detected or measured with a light microscope or electron microscope.
  • the number of strongly or highly positive immunostained cells may be counted to determine or evaluate immunostaining intensity of the biological sample.
  • the immunostaining intensity may be determined or evaluated by comparing numbers of cells having positive and/or negative staining in the biological sample with control.
  • the immunostaining intensity of the biological sample may also be estimated or assessed in some grades. For example, three or more grades scoring or classifying of the immunostaining intensity may be applied to the present invention.
  • the positive staining of the anti-STC2 antibody may be defined as follows: (1) variable weak cytoplasmic staining, and (2) segmental and apical granular cytoplasmic staining, and (3) diffuse continuous and intense cytoplasmic staining.
  • cell population of each score is determined.
  • the sum of the population of cells with each score may be calculated for each sample as described previously (Ashida S, et al., Clin Cancer Res 2006, 2767-73).
  • the control level may be determined at the same time with the test biological sample by using a sample(s) previously collected and stored from a subject/subjects whose disease state (cancerous or non-cancerous) is/are known, as mentioned above.
  • control level may be determined by using a sample(s) previously collected and stored from a subject/subjects whose disease state is/are prostate cancer with Gleason score 2-7 or a normal healthy subject/subjects.
  • the immunostaining intensity of the STC2 polypeptide in a biological sample can be considered to be increased if it increases from the control level of the STC2 polypeptide by, for example, 10%, 25%, or 50%; or increases to more than 1.1 fold, more than 1.5 fold, more than 2.0 fold, more than 5.0 fold, more than 10.0 fold, or more.
  • the STC2 gene differentially expressed between normal and PC cells also allow for the course of treatment of PC to be monitored, and the above-described method for diagnosing cancer can be applied for assessing the efficacy of a treatment on PC.
  • the efficacy of a treatment on PC can be assessed by determining the expression level of the STC2 gene in a cell(s) derived from a subject undergoing the treatment. If desired, test cell populations are obtained from the subject at various time points, before, during, and/or after the treatment.
  • the expression level of the STC2 gene can be, for example, determined following the method described above under the item of 'I-1. Method for diagnosing cancer or a predisposition for developing cancer'.
  • control level to which the detected expression level is compared is determined from the STC2 gene expression in a cell(s) not exposed to the treatment of interest. If the expression level of the STC2 gene is compared to a normal control level, a similarity in the expression level indicates that the treatment of interest is efficacious and a difference in the expression level indicates less favorable clinical outcome or prognosis of that treatment. On the other hand, if the comparison is conducted against a cancerous control level, a difference in the expression level indicates efficacious treatment, while a similarity in the expression level indicates less favorable clinical outcome or prognosis.
  • the expression levels of the STC2 gene before and after a treatment can be compared according to the present method to assess the efficacy of the treatment.
  • the expression level detected in a subject-derived biological sample after a treatment i.e., post-treatment level
  • the expression level detected in a biological sample obtained prior to treatment onset from the same subject i.e., pre-treatment level.
  • a decrease in the post-treatment level as compared to the pre-treatment level indicates that the treatment of interest is efficacious while an increase in or similarity of the post-treatment level to the pre-treatment level indicates less favorable clinical outcome or prognosis.
  • the term "efficacious" indicates that the treatment leads to a reduction in the expression of a pathologically up-regulated gene, an increase in the expression of a pathologically down-regulated gene or a decrease in size, prevalence, or metastatic potential of carcinoma in a subject.
  • "efficacious” means that the treatment retards or prevents the forming of tumor or retards, prevents, or alleviates at least one clinical symptom of cancer.
  • Assessment of the state of tumor in a subject can be made using standard clinical protocols.
  • efficaciousness of a treatment can be determined in association with any known method for diagnosing cancer. Cancers can be diagnosed, for example, by identifying symptomatic anomalies, e.g., weight loss, abdominal pain, back pain, anorexia, nausea, vomiting and generalized malaise, weakness, and jaundice.
  • the method for diagnosing PC described above can also be used for assessing the prognosis of PC in a subject.
  • the present invention also provides a method for assessing the prognosis of a subject with PC.
  • the expression level of the STC2 gene can be, for example, determined following the method described above under the item of 'I-1.
  • Method for diagnosing cancer or a predisposition for developing cancer' For example, the expression level of the STC2 gene in biological samples derived from patients over a spectrum of disease stages can be used as control levels to be compared with the expression level of the gene detected for a subject.
  • the prognosis of the subject can be assessed.
  • the prognosis of the subject can be assessed. For example, an increase in the expression level of STC2 gene in a subject as compared to a normal control level indicates less favorable prognosis. Conversely, a similarity in the expression level as compared to normal control level indicates a more favorable prognosis for the subject.
  • kits for detecting and/or diagnosing PC which may also be useful in assessing the prognosis of cancer and/or monitoring the efficacy of a cancer therapy, i.e., kits that includes at least a reagent detecting the transcription or translation product of the STC2 gene.
  • reagents include nucleic acids that specifically bind to or identify a transcription product of the STC2 gene.
  • the nucleic acid that specifically bind to or identify a transcription product of the STC2 gene include such as oligonucleotides (e.g., probes and primers) having a sequence that is complementary to a portion of the STC2 gene transcription product.
  • antibodies can be exemplified as reagents for detecting the translation product of the gene.
  • the translation product may also be detected based on its biological activity.
  • the STC2 protein was reported to inhibit the phosphate uptake of a kidney cell line (Ishibashi K, et al., Biochem Biophys Res Commun 1998, Sep 18;250(2), 258-8).
  • the inhibiting activity against the phosphate uptake of a kidney cell line may be detected for determining the expression level of the STC2 gene.
  • labeled phosphate, or other substrate(s) of the STC2 protein may be used as a reagent for detecting the expression level of the gene.
  • the translation product of STC2 gene has an ability to promote PC ell growth.
  • a kit for detecting the expression of the STC2 gene may include a labeled phosphate and a kidney cell line, or a PC line and an appropriate medium.
  • the present kit is suited for detecting CRPC and aggressive PC with high Gleason score, in particular, Gleason score 8-10.
  • These reagents may be used for the above-described diagnosis of cancer.
  • the assay format for using the reagents may be Northern hybridization or sandwich ELISA, both of which are well-known in the art.
  • the detection reagents may be packaged together in the form of a kit.
  • the detection reagents may be packaged in separate containers.
  • the detection reagents may be packaged with other reagents necessary for the detection.
  • a kit may include a nucleic acid or antibody (either bound to a solid matrix or packaged separately with reagents for binding them to the matrix) as the detection reagent, 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 may also be included in the kit.
  • the reagents for detecting cancer may be immobilized on a solid matrix, such as a porous strip, to form at least one site for detecting cancer.
  • the measurement or detection region of the porous strip may include a plurality of sites, each containing a detection reagent (e.g., nucleic acid).
  • a test strip may also contain sites for negative and/or positive controls. Alternatively, control sites may be located on a separate strip from the test strip.
  • the different detection sites may contain different amounts of immobilized detection reagents (e.g., nucleic acid), i.e., a higher amount in the first detection site and lesser amounts in subsequent sites.
  • the number of sites displaying a detectable signal provides a quantitative indication of the expression level of the STC2 gene in the sample.
  • the detection sites may be configured in any suitably detectable shape and are typically in the shape of a bar or dot spanning the width of a test strip.
  • An agent or substance isolated by the screening method of the present invention is an agent or substance that is expected to inhibit the expression of the STC2 gene or the activity of the translation product of the gene, and thus, is a candidate for treating or preventing PC.
  • the agents or substances are expected to be particularly suited for the treatment or prevention of CRPC and aggressive PC with high Gleason score (in particular, Gleason score 8-10). Namely, the agents or substances screened through the present methods are deemed to have a clinical benefit and can be further tested for its ability to prevent cancer cell growth in animal models or test subjects.
  • agents or substances to be identified through the present screening methods may be any compound or composition including several compounds.
  • test agent or substance exposed to a cell or protein according to the screening methods of the present invention may be a single compound or a combination of compounds.
  • the compounds may be contacted sequentially or simultaneously.
  • Any test agent or substance for example, cell extracts, cell culture supernatant, products of fermenting microorganism, extracts from marine organism, plant extracts, purified or crude proteins, peptides, non-peptide compounds, synthetic micromolecular compounds (including nucleic acid constructs, such as antisense RNA, siRNA, Ribozymes, etc.) and natural compounds can be used in the screening methods of the present invention.
  • test agent or substances 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.
  • an agent or substance that binds to STC2 polypeptide and/or reduces the expression level of the STC2 gene may be used as a test agent or substance for the further screening for a candidate agent or substance that has the STC2 specific therapeutic effect.
  • test agent or substances when the screened test agent or substances is a protein, for obtaining a DNA encoding the protein, either the whole amino acid sequence of the protein may be determined to deduce the nucleic acid sequence coding for the protein, or partial amino acid sequence of the obtained protein may be analyzed to prepare an oligo DNA as a probe based on the sequence, and screen cDNA libraries with the probe to obtain a DNA encoding the protein.
  • the obtained DNA find use in preparing the test agent or substance which is a candidate for treating or preventing cancer.
  • Test agents useful in the screenings described herein can also be antibodies that specifically bind to a STC2 protein or partial peptides thereof that lack the biological activity of the original proteins in vivo.
  • test agent libraries are facilitated by knowledge of the molecular structure of compounds known to have the properties sought, and/or the molecular structure of STC2.
  • One approach to preliminary screening of test agents suitable for further evaluation utilizes computer modeling of the interaction between the test agent and its target.
  • Computer modeling technology allows for the visualization of the three-dimensional atomic structure of a selected molecule and the rational design of new compounds that will interact with the molecule.
  • the three-dimensional construct typically depends on data from x-ray crystallographic analysis or NMR imaging of the selected molecule.
  • the molecular dynamics require force field data.
  • the computer graphics systems enable prediction of how a new compound will link to the target molecule and allow experimental manipulation of the structures of the compound and target molecule to perfect binding specificity.
  • test agents may be screened using the methods of the present invention to identify test agents suited to the treatment and/or prophylaxis of cancer and/or the prevention of post-operative recurrence of cancer, particularly wherein, such as pancreatic cancer and prostate cancer.
  • Combinatorial chemical synthesis Combinatorial libraries of test agents may be produced as part of a rational drug design program involving knowledge of core structures existing in known inhibitors. This approach allows the library to be maintained at a reasonable size, facilitating high throughput screening.
  • simple, particularly short, polymeric molecular libraries may be constructed by simply synthesizing all permutations of the molecular family making up the library.
  • An example of this latter approach would be a library of all peptides six amino acids in length. Such a peptide library could include every 6 amino acid sequence permutation.
  • This type of library is termed a linear combinatorial chemical library. Preparation of combinatorial chemical libraries is well known to those of skill in the art, and may be generated by either chemical or biological synthesis.
  • Combinatorial chemical libraries include, but are not limited to, peptide libraries (see, e.g., US Patent 5,010,175; Furka, Int J Pept Prot Res 1991, 37: 487-93; Houghten et al., Nature 1991, 354: 84-6).
  • Other chemistries for generating chemical diversity libraries can also be used. Such chemistries include, but are not limited to: peptides (e.g., PCT Publication No.
  • WO 91/19735 encoded peptides (e.g., WO 93/20242), random bio-oligomers (e.g., WO 92/00091), benzodiazepines (e.g., US Patent 5,288,514), diversomers such as hydantoins, benzodiazepines and dipeptides (DeWitt et al., Proc Natl Acad Sci USA 1993, 90:6909-13), vinylogous polypeptides (Hagihara et al., J Amer Chem Soc 1992, 114: 6568), nonpeptidal peptidomimetics with glucose scaffolding (Hirschmann et al., J Amer Chem Soc 1992, 114: 9217-8), analogous organic syntheses of small compound libraries (Chen et al., J.
  • a second approach uses primarily chemical methods, of which the Geysen method (Geysen et al., Molecular Immunology 1986, 23: 709-15; Geysen et al., J Immunologic Method 1987, 102: 259-74); and the method of Fodor et al. (Science 1991, 251: 767-73) are examples.
  • Furka et al. 14th International Congress of Biochemistry 1988, Volume #5, Abstract FR:013; Furka, Int J Peptide Protein Res 1991, 37: 487-93
  • Houghten US Patent 4,631,211
  • Rutter et al. US Patent 5,010,175) describe methods to produce a mixture of peptides that can be tested as agonists or antagonists.
  • Aptamers are macromolecules composed of nucleic acid that bind tightly to a specific molecular target.
  • Tuerk and Gold discloses SELEX (Systematic Evolution of Ligands by Exponential Enrichment) method for selection of aptamers.
  • SELEX Systematic Evolution of Ligands by Exponential Enrichment
  • a large library of nucleic acid molecules e.g., 10 15 different molecules
  • the expression of the STC2 gene was suggested to be crucial for the growth and/or survival of cancer cells, in particular PC cells, more particularly CRPC cells and aggressive PC cells with high Gleason score (e.g., Gleason score 8-10). Therefore, it was considered that agents or substances that suppress the function of the polypeptide encoded by the STC2 gene would reduce or inhibit the growth and/or survival of cancer cells, and thus find use in the treatment and prevention of prostate cancer. Accordingly, the present invention provides methods of screening an agent for treating or preventing cancer, using the STC2 polypeptide.
  • fragments of the polypeptide may be used for the present screening so long as it retains at least one biological activity of the natural occuring STC2 polypeptide.
  • the polypeptide or fragments thereof may be further linked to other substances so long as the polypeptide and fragments retains at least one of its biological activity.
  • Usable substances include: peptides, lipids, sugar and sugar chains, acetyl groups, natural and synthetic polymers, etc. These kinds of modifications may be performed to confer additional functions or to stabilize the polypeptide and fragments.
  • the polypeptide or fragments used for the present method may be obtained from nature as naturally occurring proteins via conventional purification methods or through chemical synthesis based on the selected amino acid sequence.
  • conventional peptide synthesis methods that can be adopted for the synthesis include: 1) Peptide Synthesis, Interscience, New York, 1966; 2) The Proteins, Vol. 2, Academic Press, New York, 1976; 3) Peptide Synthesis (in Japanese), Maruzen Co., 1975; 4) Basics and Experiment of Peptide Synthesis (in Japanese), Maruzen Co., 1985; 5) Development of Pharmaceuticals (second volume) (in Japanese), Vol. 14 (peptide synthesis), Hirokawa, 1991; 6) WO99/67288; and 7) Barany G.
  • the protein may be obtained adopting any known genetic engineering methods for producing polypeptides (e.g., Morrison J., J Bacteriology 1977, 132: 349-51; Clark-Curtiss & Curtiss, Methods in Enzymology (eds. Wu et al.) 1983, 101: 347-62).
  • a suitable vector including a polynucleotide encoding the objective protein in an expressible form is prepared, transformed into a suitable host cell, and then the host cell is cultured to produce the protein.
  • a gene encoding the STC2 polypeptide is expressed in host (e.g., animal) cells and such by inserting the gene into a vector for expressing foreign genes, such as pSV2neo, pcDNA I, pcDNA3.1, pCAGGS, or pCD8.
  • a promoter may be used for the expression.
  • any commonly used promoters may be employed including, for example, the SV40 early promoter (Rigby in Williamson (ed.), Genetic Engineering, vol. 3. Academic Press, London, 1982, 83-141), the EF- alpha promoter (Kim et al., Gene 1990, 91:217-23), the CAG promoter (Niwa et al., Gene 1991, 108:193), the RSV LTR promoter (Cullen, Methods in Enzymology 1987, 152:684-704), the SR alpha promoter (Takebe et al., Mol Cell Biol 1988, 8:466), the CMV immediate early promoter (Seed et al., Proc Natl Acad Sci USA 1987, 84:3365-9), the SV40 late promoter (Gheysen et al., J Mol Appl Genet 1982, 1:385-94), the Adenovirus late promoter (Kaufman et al., Mol Cell Biol 1989, 9:
  • the introduction of the vector into host cells to express the STC2 gene can be performed according to any methods, for example, the electroporation method (Chu et al., Nucleic Acids Res 1987, 15:1311-26), the calcium phosphate method (Chen et al., Mol Cell Biol 1987, 7:2745-52), the DEAE dextran method (Lopata et al., Nucleic Acids Res 1984, 12:5707-17; Sussman et al., Mol Cell Biol 1985, 4:1641-3), the Lipofectin method (Derijard B, Cell 1994, 7:1025-37; Lamb et al., Nature Genetics 1993, 5:22-30; Rabindran et al., Science 1993, 259:230-4), and such.
  • the STC2 protein may also be produced in vitro adopting an in vitro translation system.
  • the STC2 polypeptide to be contacted with a test agent or substance can be, for example, a purified polypeptide, a soluble protein, or a fusion protein fused with other polypeptides.
  • test agent or substance for example, cell extracts, cell culture supernatant, products of fermenting microorganism, extracts from marine organism, plant extracts, purified or crude proteins, peptides, non-peptide compounds, synthetic micromolecular compounds (including nucleic acid constructs, such as antisense RNA, siRNA, Ribozymes, and aptamer etc.) and natural compounds can be used in the screening methods of the present invention.
  • test agent or substance for example, cell extracts, cell culture supernatant, products of fermenting microorganism, extracts from marine organism, plant extracts, purified or crude proteins, peptides, non-peptide compounds, synthetic micromolecular compounds (including nucleic acid constructs, such as antisense RNA, siRNA, Ribozymes, and aptamer etc.) and natural compounds can be used in the screening methods of the present invention.
  • test agent or substance 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.
  • biological libraries using affinity chromatography selection is limited to peptide libraries, while the other four approaches are applicable to peptide, non-peptide oligomer or small molecule libraries of compounds (Lam, Anticancer Drug Des 1997, 12: 145-67).
  • the screened test agent or substance is a protein
  • obtaining a DNA encoding the protein either the whole amino acid sequence of the protein may be determined to deduce the nucleic acid sequence coding for the protein, or partial amino acid sequence of the obtained protein may be analyzed to prepare an oligo DNA as a probe based on the sequence, and screen cDNA libraries with the probe to obtain a DNA encoding the protein.
  • the obtained DNA is confirmed it's usefulness in preparing the test agent which is a candidate for treating or preventing cancer.
  • the present invention provides a method of screening an agent for treating or preventing cancer, that includes the steps of: a) contacting a test agent or substance with the STC2 polypeptide or a fragment thereof; b) detecting the binding between the polypeptide or fragment and the test agent or substance; and c) selecting the test agent or substance that binds to the polypeptide as a candidate agent for treating or preventing cancer.
  • the present invention also provides a method of screening for a candidate agent or substance for inhibiting the cell growth or a candidate agent or substance for treating or preventing STC2 associating cancer, using the STC2 polypeptide or fragments thereof including the steps as follows: a) contacting a test agent or substance with the STC2 polypeptide or a functional fragment thereof; and b) detecting the binding level between the polypeptide or functional fragment and the test agent or substance of step (a), and c) correlating the binding level of b) with the therapeutic effect of the test agent or substance.
  • the therapeutic effect may be correlated with the binding level of STC2 polypeptide or a functional fragment thereof.
  • the test agent or substance may identified or selected as the candidate agent or substance having the therapeutic effect.
  • the test agent or substance may identified as the agent or substance having no significant therapeutic effect.
  • the binding of a test agent or substance to the STC2 polypeptide may be, for example, detected by immunoprecipitation using an antibody against the polypeptide. Therefore, for the purpose for such detection, it is preferred that the STC2 polypeptide or fragments thereof used for the screening contains an antibody recognition site.
  • the antibody used for the screening may be one that recognizes an antigenic region (e.g., epitope) of the present STC2 polypeptide which preparation methods are well known in the art, and any method may be employed in the present invention to prepare such antibodies and equivalents thereof.
  • the STC2 polypeptide or a fragment thereof may be expressed as a fusion protein having at its N- or C-terminus a recognition site (epitope) of a monoclonal antibody, whose specificity has been revealed, to the N- or C- terminus of the polypeptide.
  • a commercially available epitope-antibody system can be used (Experimental Medicine 1995, 13:85-90).
  • Vectors which can express a fusion protein with, for example, beta-galactosidase, maltose binding protein, glutathione S-transferase, green florescence protein (GFP), and such by the use of its multiple cloning sites are commercially available and can be used for the present invention.
  • epitopes composed of several to a dozen amino acids so as not to change the property of the STC2 polypeptide or fragments thereof, can also be used in the present invention.
  • epitopes composed of several to a dozen amino acids so as not to change the property of the STC2 polypeptide or fragments thereof, can also be used in the present invention. Examples include polyhistidine (His-tag), influenza aggregate HA, human c-myc, FLAG, Vesicular stomatitis virus glycoprotein (VSV-GP), T7 gene 10 protein (T7-tag), human simple herpes virus glycoprotein (HSV-tag), E-tag (an epitope on monoclonal phage), and such.
  • Glutathione S-transferase is also well-known as the counterpart of the fusion protein to be detected by immunoprecipitation.
  • GST is used as the protein to be fused with the STC2 polypeptide or fragment thereof to form a fusion protein
  • the fusion protein can be detected either with an antibody against GST or a substance specifically binding to GST, i.e., such as glutathione (e.g., glutathione-Sepharose 4B).
  • glutathione e.g., glutathione-Sepharose 4B
  • an immune complex is formed by adding an antibody (recognizing the STC2 polypeptide or a fragment thereof itself, or an epitope tagged to the polypeptide or fragment) to the reaction mixture of the STC2 polypeptide and the test agent or substances .
  • the formed immune complex will include the STC2 polypeptide, the test agent or substance, and the antibody.
  • the test agent or substance are devoid of such ability, then the formed immune complex only include the STC2 polypeptide and the antibody. Therefore, the binding ability of a test agent or substance to STC2 polypeptide can be examined by, for example, measuring the size of the formed immune complex. Any method for detecting the size of a substance can be used, including chromatography, electrophoresis, and such. For example, when mouse IgG antibody is used for the detection, Protein A or Protein G sepharose can be used for quantitating the formed immune complex. Immunoprecipitation can be performed by following or according to, for example, the methods in the literature (See, for example, Harlow et al., Antibodies, Cold Spring Harbor Laboratory publications, New York, 1988, 511-52).
  • the STC2 polypeptide or a fragment thereof used for the screening of agent or substance that bind to thereto may be bound to a carrier.
  • carriers that may be used for binding the polypeptides include insoluble polysaccharides, such as agarose, cellulose and dextran; and synthetic resins, such as polyacrylamide, polystyrene and silicon; preferably commercially available beads and plates (e.g., multi-well plates, biosensor chip, etc.) prepared from the above materials may be used. When using beads, they may be filled into a column. Alternatively, the use of magnetic beads is also known in the art, and enables to readily isolate polypeptides and agents or substances bound on the beads via magnetism.
  • binding of a polypeptide to a carrier may be conducted according to routine methods, such as chemical bonding and physical adsorption.
  • a polypeptide may be bound to a carrier via antibodies specifically recognizing the protein.
  • binding of a polypeptide to a carrier can also be conducted by means of interacting molecules, such as the combination of avidin and biotin. Screening using such carrier-bound STC2 polypeptide or fragments thereof include, for example, contacting a test agent or substance to the carrier-bound polypeptide, incubating the mixture, washing the carrier, and detecting and/or measuring the agent or substance bound to the carrier.
  • the binding may be carried out in buffer, for example, but are not limited to, phosphate buffer and Tris buffer, as long as the buffer does not inhibit the binding.
  • a screening method wherein such carrier-bound STC2 polypeptide or fragments thereof and a composition (e.g., cell extracts, cell lysates, etc.) are used as the test agent or substance such method is generally called affinity chromatography.
  • the STC2 polypeptide may be immobilized on a carrier of an affinity column, and a test agent or substance , containing a substance capable of binding to the polypeptides, is applied to the column. After loading the test agent or substance , the column is washed, and then the substance bound to the polypeptide is eluted with an appropriate buffer.
  • a biosensor using the surface plasmon resonance phenomenon may be used as a mean for detecting or quantifying the bound agent or substance in the present invention.
  • a biosensor When such a biosensor is used, the interaction between the STC2 polypeptide and a test agent or substance can be observed real-time as a surface plasmon resonance signal, using only a minute amount of the polypeptide and without labeling (for example, BIAcore, Pharmacia). Therefore, it is possible to evaluate the binding between the polypeptide and test agent or substance using a biosensor such as BIAcore.
  • a protein binding to the STC2 polypeptide can be obtained by preparing first a cDNA library from cells, tissues, organs, or cultured cells (e.g., PC cell lines) expected to express at least one protein binding to the STC2 polypeptide using a phage vector (e.g., ZAP), expressing the proteins encoded by the vectors of the cDNA library on LB-agarose, fixing the expressed proteins on a filter, reacting the purified and labeled STC2 polypeptide with the above filter, and detecting the plaques expressing proteins to which the STC2 polypeptide has bound according to the label of the STC2 polypeptide.
  • a cDNA library from cells, tissues, organs, or cultured cells (e.g., PC cell lines) expected to express at least one protein binding to the STC2 polypeptide using a phage vector (e.g., ZAP), expressing the proteins encoded by the vectors of the cDNA library on LB-agarose, fixing the expressed proteins on
  • Labeling substances such as radioisotope (e.g., 3 H, 14 C, 32 P, 33 P, 35 S, 125 I, 131 I), enzymes (e.g., alkaline phosphatase, horseradish peroxidase, beta-galactosidase, beta-glucosidase), fluorescent substances (e.g., fluorescein isothiocyanate (FITC), rhodamine) and biotin/avidin, may be used for the labeling of STC2 polypeptide in the present method.
  • radioisotope e.g., 3 H, 14 C, 32 P, 33 P, 35 S, 125 I, 131 I
  • enzymes e.g., alkaline phosphatase, horseradish peroxidase, beta-galactosidase, beta-glucosidase
  • fluorescent substances e.g., fluorescein isothiocyanate (FITC), rhodamine
  • the protein when it is labeled with an enzyme, it can be detected or measured by adding a substrate of the enzyme to detect the enzymatic change of the substrate, such as generation of color, with absorptiometer. Further, in case where a fluorescent substance is used as the label, the bound protein may be detected or measured using fluorophotometer.
  • the STC2 polypeptide bound to the protein can be detected or measured by utilizing an antibody that specifically binds to the STC2 polypeptide, or a peptide or polypeptide (for example, GST) that is fused to the STC2 polypeptide.
  • the antibody is preferably labeled with one of the labeling substances mentioned above, and detected or measured based on the labeling substance.
  • the antibody against the STC2 polypeptide may be used as a primary antibody to be detected with a secondary antibody that is labeled with a labeling substance.
  • the antibody bound to the STC2 polypeptide in the present screening may be detected or measured using protein G or protein A column.
  • two-hybrid system utilizing cells may be used ("MATCHMAKER Two-Hybrid system", “Mammalian MATCHMAKER Two-Hybrid Assay Kit”, “MATCHMAKER one-Hybrid system” (Clontech); “HybriZAP Two-Hybrid Vector System” (Stratagene); the references “Dalton et al., Cell 1992, 68:597-612” and “Fields et al., Trends Genet 1994, 10:286-92").
  • STC2 polypeptide or a fragment thereof is fused to the SRF-binding region or GAL4-binding region and expressed in yeast cells.
  • a cDNA library is prepared from cells expected to express at least one protein binding to the STC2 polypeptide, such that the library, when expressed, is fused to the VP16 or GAL4 transcriptional activation region.
  • the cDNA library is then introduced into the above yeast cells and the cDNA derived from the library is isolated from the positive clones detected (when a protein binding to the STC2 polypeptide is expressed in the yeast cells, the binding of the two activates a reporter gene, making positive clones detectable).
  • a protein encoded by the cDNA can be prepared by introducing the cDNA isolated above to E. coli and expressing the protein.
  • a reporter gene for example, Ade2 gene, lacZ gene, CAT gene, luciferase gene and such can be used in addition to the HIS3 gene.
  • the agent or substance isolated by this screening is a candidate for antagonists of the STC2 polypeptide.
  • antagonist refers to molecules that inhibit the function of the polypeptide by binding thereto.
  • suppressing the expression of STC2 reduces prostate cancer cell growth.
  • candidate compounds that find use to treat or prevent Prostate cancers can be identified.
  • the potential of these candidate substance or compounds to treat or prevent prostate cancers may be evaluated by a secondary and/or further screening to identify therapeutic agents useful in treating or preventing prostate cancers. For example, when a compound binding to STC2 protein inhibits , e.g., the proliferative activity or activity to inhibit phosphate uptake of prostate cancer cells, it may be concluded that such compound has the STC2 specific therapeutic effect.
  • STC2 gene was shown to be crucial for the growth and/or survival of cancer cells, in particular, prostate cancer cells, more particularly, CRPC cells and aggressive PC cells with high Gleason score (e.g., Gleason score 8-10). Therefore, agents or substances that suppress or inhibit the biological function of the translational product of the STC2 gene is considered to serve as candidates for treating or preventing cancer.
  • cancer cells in particular, prostate cancer cells, more particularly, CRPC cells and aggressive PC cells with high Gleason score (e.g., Gleason score 8-10). Therefore, agents or substances that suppress or inhibit the biological function of the translational product of the STC2 gene is considered to serve as candidates for treating or preventing cancer.
  • the present invention also provides a method for screening a compound for treating or preventing cancer using the STC2 polypeptide or fragments thereof including the steps as follows: a) contacting a test agent or substance with the STC2 polypeptide or a fragment thereof; and b) detecting the biological activity of the polypeptide or fragment of step (a).
  • Any polypeptide can be used for the screening so long as it has a biological activity equivalent to the STC2 polypeptide that can be used as an index in the present screening method.
  • the STC2 polypeptide has been demonstrated to be required for the growth or viability of PC cells (more specifically, CRPC cells and aggressive PC with high Grealon score).
  • the STC2 was reported to inhibit the phosphate uptake of kidney cell line. Therefore, biological activities of the STC2 polypeptide that can be used as an index for the screening include such cell growth promoting activity and the inhibiting activity against phosphate uptake of a kidney cell line.
  • a human STC2 polypeptide can be used and polypeptides functionally equivalent thereto including fragments thereof can also be used. Such polypeptides may be expressed endogenously or exogenously by suitable cells.
  • detection may be accomplished, for example, by preparing cells which express the STC2 polypeptide or a fragment thereof (e.g., 22Rv1, LNCaP, C4-2B, DU145, PC-3, etc.), culturing the cells in the presence of a test agent or substance , and determining the speed of cell proliferation, measuring the cell cycle and such, as well as by detecting wound-healing activity, conducting Matrigel invasion assay and measuring the colony forming activity.
  • STC2 polypeptide or a fragment thereof e.g., 22Rv1, LNCaP, C4-2B, DU145, PC-3, etc.
  • the screening further includes, after the above step (b), the steps of: c) comparing the biological activity of the polypeptide or fragment with the biological activity detected in the absence of the agent or substance ; and d) selecting the agent or substance that suppresses the biological activity of the polypeptide as a candidate agent for treating or preventing PC.
  • the therapeutic effect of the test agent or substance on inhibiting the cell growth or a candidate agent or substance for treating or preventing STC2 associating cancer may be evaluated.
  • the present invention also provides a method of screening for a candidate agent or substance for inhibiting the cell growth or a candidate agent or substance for treating or preventing STC2 associating cancer, using the STC2 polypeptide or fragments thereof including the steps as follows: a) contacting a test agent or substance with the STC2 polypeptide or a functional fragment thereof; and b) detecting the biological activity of the polypeptide or fragment of step (a), and c) correlating the biological activity of b) with the therapeutic effect of the test agent or substance.
  • the therapeutic effect may be correlated with the biological activity STC2 polypeptide or a functional fragment thereof.
  • the test agent or substance when the test agent or substance suppresses or inhibits the biological activity of STC2 polypeptide or a functional fragment thereof as compared to a level detected in the absence of the test agent or substance, the test agent or substance may identified or selected as the candidate agent or substance having the therapeutic effect.
  • the test agent or substance when the test agent or substance does not suppress or inhibit the biological activity of STC2 polypeptide or a functional fragment thereof as compared to a level detected in the absence of the test agent or substance, the test agent or substance may be identified as the agent or compound having no significant therapeutic effect.
  • control cells which do not express STC2 polypeptide are used.
  • the present invention also provides a method of screening for a candidate substance for inhibiting the cell growth or a candidate substance for treating or preventing STC2 associating disease, using the STC2 polypeptide or fragments thereof including the steps as follows: a) culturing cells which express a STC2 polypeptide or a functional fragment thereof, and control cells that do not express a STC2 polypeptide or a functional fragment thereof in the presence of the test substance; b) detecting the biological activity of the cells which express the protein and control cells; and c) selecting the test compound that inhibits the biological activity in the cells which express the protein as compared to the proliferation detected in the control cells and in the absence of said test substance.
  • the present invention also provides a screening method following the method described in III-1, including the steps of: a) contacting a test agent or substance with the STC2 polypeptide or a fragment thereof; b) detecting the binding between the polypeptide or fragment and the test agent or substance ; c) selecting the test agent or substance that binds to the polypeptide; d) contacting the test agent or substance selected in step c) with the STC2 polypeptide or a fragment thereof; e) comparing the biological activity of the polypeptide or fragment with the biological activity detected in the absence of the agent or substance ; and f) selecting the agent or substance that suppresses the biological activity of the polypeptide as a candidate agent for treating or preventing PC.
  • the agent or substance isolated by this screening is a candidate for an antagonist of the STC2 polypeptide, and thus, is a candidate that inhibits the in vivo interaction of the polypeptide with molecules (including nucleic acids (RNAs and DNAs) and proteins).
  • the STC2 gene plays the crucial role in the cell proliferation showing in the example 4 and 5. Therefore, a biological activity of STC2 to be detected in the present screening method may be its cell proliferation activity.
  • the present invention further provides a method for screening an agent for treating or preventing cancer.
  • An embodiment of this screening method includes the steps of: (a) contacting a cell that expresses the STC2 polypeptide or a fragment thereof with an agent or substance ; (b) detecting the inhibiting activity of the STC2 polypeptide against the phosphate uptake of a kidney cell line; (c) comparing the inhibiting activity of the STC2 polypeptide against the phosphate uptake detected in the absence of the agent or substance ; and (d) selecting the agent or substance that suppresses the inhibiting activity of the STC2 polypeptide against the phosphate uptake activity as a candidate agent or substance for treating or preventing cancer.
  • candidate compounds for suppressing the growth of PC cells especially CRPC and aggressive PC with high Gleason score (e.g., Gleason score 8-10), can be identified.
  • agents or substances by detecting expression level of the STC2 gene in a cell
  • agents or substances that may be used in the treatment or prevention of PCs in particular, CRPC and aggressive PC with high Gleason score (e.g., Gleason score 8-10) can be identified through screenings that use the expression levels of STC2 gene as indices.
  • such screening may include, for example, the following steps: a) contacting a test agent or substance with a cell expressing the STC2 gene; b) detecting the expression level of the STC2 gene; c) comparing the expression level with the expression level detected in the absence of the agent or substance ; and d) selecting the agent or substance that reduces the expression level as a candidate agent for treating or preventing cancer.
  • the therapeutic effect of the test agent or substance on inhibiting the cell growth or a candidate agent or substance for treating or preventing STC2 associating cancer may be evaluated.
  • the present invention also provides a method for screening a candidate agent or substance that suppresses the proliferation of cancer cells, and a method for screening a candidate agent or substance for treating or preventing STC2 associating cancer.
  • screening may include, for example, the following steps: a) contacting a test agent or substance with a cell expressing the STC2 gene; b) detecting the expression level of the STC2 gene; and c) correlating the expression level of b) with the therapeutic effect of the test agent or substance.
  • the therapeutic effect may be correlated with the expression level of the STC2 gene.
  • the test agent or substance when the test agent or substance reduces the expression level of the STC2 gene as compared to a level detected in the absence of the test agent or substance, the test agent or substance may identified or selected as the candidate agent or substance having the therapeutic effect.
  • the test agent or substance when the test agent or substance does not reduce the expression level of the STC2 gene as compared to a level detected in the absence of the test agent or substance, the test agent or substance may be identified as the agent or substance having no significant therapeutic effect.
  • An agent or substance that inhibits the expression of the STC2 gene or the activity of its gene product can be identified by contacting a cell expressing the STC2 gene with a test agent or substance and then determining the expression level of the STC2 gene. Naturally, the identification may also be performed using a population of cells that express the gene in place of a single cell. A decreased expression level detected in the presence of an agent or substance as compared to the expression level in the absence of the agent or substance indicates the agent as being an inhibitor of the STC2 gene, suggesting the possibility that the agent is useful for inhibiting PC, thus a candidate agent to be used for the treatment or prevention of PC, in particular, CRPC and aggressive PC (e.g., Gleason score 8-10).
  • the expression level of a gene can be estimated by methods well known to one skilled in the art.
  • the expression level of the STC2 gene can be, for example, determined following the method described above under the item of 'I-1.
  • Method for diagnosing cancer or a predisposition for developing cancer' The cell or the cell population used for such identification may be any cell or any population of cells so long as it expresses the STC2 gene.
  • the cell or population may be or contain an immortalized cell derived from a carcinoma cell, including PC cell.
  • Cells expressing the STC2 gene include, for example, cell lines established from cancers (e.g., PC cell lines such as 22Rv1, LNCaP, C4-2B, DU145, PC-3, etc.).
  • the cell or population may be or contain a cell which has been transfected with the STC2 gene.
  • the present method allows screening of various agents or substances mentioned above and is particularly suited for screening functional nucleic acid molecules including antisense RNA, siRNA, and such.
  • the present invention provides a method which includes the following steps of: a) contacting a test agent or substance with a cell into which a vector, including the transcriptional regulatory region of the STC2 gene and a reporter gene that is expressed under the control of the transcriptional regulatory region, has been introduced; b) detecting the expression or activity of said reporter gene; c) comparing the expression level or activity with the expression level or activity detected in the absence of the agent or substance ; and d) selecting the agent or substance that reduces the expression or activity of said reporter gene as a candidate agent for treating or preventing cancer.
  • the therapeutic effect of the test agent or substance on inhibiting the cell growth or a candidate agent or substance for treating or preventing STC2 associating cancer may be evaluated. Therefore, the present invention also provides a method for screening a candidate agent or substance that suppresses the proliferation of cancer cells, and a method for screening a candidate agent or compound for treating or preventing STC2 associating cancer.
  • the present invention provides a method which includes the following steps of: a) contacting a test agent or substance with a cell into which a vector, composed of the transcriptional regulatory region of the STC2 gene and a reporter gene that is expressed under the control of the transcriptional regulatory region, has been introduced; b) detecting the expression level or activity of said reporter gene; and c) correlating the expression level of b) with the therapeutic effect of the test agent or substance.
  • the therapeutic effect may be correlated with the expression level or activity of said reporter gene.
  • test agent or substance when the test agent or substance reduces the expression level or activity of said reporter gene as compared to a level detected in the absence of the test agent or substance, the test agent or substance may be identified or selected as the candidate agent or substance having the therapeutic effect.
  • the test agent or substance when the test agent or substance does not reduce the expression level or activity of said reporter gene as compared to a level detected in the absence of the test agent or substance, the test agent or substance may be identified as the agent or substance having no significant therapeutic effect.
  • reporter genes are luciferase, green florescence protein (GFP), Discosoma sp. Red Fluorescent Protein (DsRed), Chrolamphenicol Acetyltransferase (CAT), lacZ and beta-glucuronidase (GUS), and host cell is COS7, HEK293, HeLa and so on.
  • the reporter construct required for the screening can be prepared by connecting reporter gene sequence to the transcriptional regulatory region of STC2.
  • the transcriptional regulatory region of STC2 herein is the region from start codon to at least 500bp upstream, preferably 1000bp, more preferably 5000 or 10000bp upstream.
  • a nucleotide segment containing the transcriptional regulatory region can be isolated from a genome library or can be propagated by PCR.
  • the reporter construct required for the screening can be prepared by connecting reporter gene sequence to the transcriptional regulatory region of any one of these genes. Methods for identifying a transcriptional regulatory region, and also assay protocol are well known (Molecular Cloning third edition chapter 17, 2001, Cold Springs Harbor Laboratory Press).
  • the vector containing the said reporter construct is infected to host cells and the expression or activity of the reporter gene is detected by method well known in the art (e.g., using luminometer, absorption spectrometer, flow cytometer and so on).
  • "reduces the expression or activity" as defined herein are preferably at least 10% reduction of the expression or activity of the reporter gene in comparison with in absence of the compound, more preferably at least 25%, 50% or 75% reduction and most preferably at 95% reduction.
  • An agent or substance that is metabolized in a subject to act as an anti-tumor agent or substance can manifest itself by inducing a change in a gene expression pattern in the subject's cells from that characteristic of a cancerous state to a gene expression pattern characteristic of a non cancerous state.
  • the STC2 gene differentially expressed between non-cancerous prostate cells and cancerous prostate cells, in particular, CRPC cells and aggressive PC cells (e.g., Gleason score 8-10), disclosed herein allow for a putative therapeutic or prophylactic inhibitor of PC (in particular, CRPC and aggressive PC) to be tested in a test cell population from a selected subject in order to determine if the agent or substance are a suitable inhibitor of PC in the subject.
  • a test cell population from the subject is exposed to a candidate therapeutic agent or substance , and the expression of STC2 gene is determined.
  • test cell populations contain cells expressing the STC2 gene.
  • the test cell is a PC cell, more preferably, a CRPC cell or an aggressive PC cell (e.g., Gleason score 8-10).
  • a test cell population may be incubated in the presence of a candidate therapeutic agent or substance and the expression of the STC2 gene in the test cell population may be measured and compared to one or more reference profiles, e.g., a cancerous reference expression profile or a non-cancerous reference expression profile.
  • a decrease in the expression of the STC2 gene in a test cell population relative to a reference cell population containing cancerous cells indicates that the agent or substance have therapeutic potential.
  • a similarity in the expression of the STC2 gene in a test cell population relative to a reference cell population not containing cancerous cells indicates that the agent or substance have therapeutic potential.
  • Double-stranded molecules and use thereof IV-1 Double-stranded molecules Double-stranded molecules targeting the STC2 gene can inhibit the expression of the STC2 gene and cell proliferation when the molecule introduced into a cell expressing the STC2 gene (Example 4). Accordingly, present invention provides for such double-stranded molecules and methods of use the same to target the STC2 gene.
  • isolated double-stranded molecule refers to a nucleic acid molecule that inhibits expression of a target gene including, for example, short interfering RNA (siRNA; e.g., double-stranded ribonucleic acid (dsRNA) or small hairpin RNA (shRNA)) and short interfering DNA/RNA (siD/R-NA; e.g. double-stranded chimera of DNA and RNA (dsD/R-NA) or small hairpin chimera of DNA and RNA (shD/R-NA)).
  • siRNA short interfering RNA
  • dsRNA double-stranded ribonucleic acid
  • shRNA small hairpin RNA
  • siD/R-NA short interfering DNA/RNA
  • dsD/R-NA double-stranded chimera of DNA and RNA
  • shD/R-NA small hairpin chimera of DNA and RNA
  • sense strand of a target sequence is a nucleotide sequence within mRNA or cDNA sequence of a gene, which will result in suppress of translation of the whole mRNA if a double-stranded nucleic acid molecule of the present invention was introduced within a cell expressing the gene.
  • a nucleotide sequence within mRNA or cDNA sequence of a gene can be determined to be a target sequence when a double-stranded polynucleotide including a sequence corresponding to the target sequence inhibits expression of the gene in a cell expressing the gene.
  • the double stranded polynucleotide by which suppresses the gene expression may be composed of the target sequence and 3' overhang (e.g., uu).
  • siRNA refers to a double-stranded RNA molecule which prevents translation of a target mRNA. Standard techniques of introducing siRNA into the cell are used, including those in which DNA is a template from which RNA is transcribed.
  • the siRNA includes a sense nucleic acid sequence (also referred to as "sense strand") of the STC2 gene, a antisense nucleic acid sequence (also referred to as “antisense strand") of the STC2 gene or both.
  • the siRNA may be constructed such that a single transcript has both the sense and complementary antisense nucleic acid sequences of the target gene, e.g., a hairpin.
  • the siRNA may either be a dsRNA or shRNA.
  • dsRNA refers to a construct of two RNA molecules having complementary sequences to one another and that have annealed together via the complementary sequences to form a double-stranded RNA molecule.
  • the nucleotide sequence of two strands may include not only the "sense” or “antisense” RNAs selected from a protein coding sequence of target gene sequence, but also RNA molecule having a nucleotide sequence selected from non-coding rigion of the target gene.
  • shRNA refers to an siRNA having a stem-loop structure, including a first and second regions complementary to one another, i.e., sense and antisense strands.
  • the degree of complementarity and orientation of the regions are sufficient such that base pairing occurs between the regions, the first and second regions are joined by a loop region, and the loop results from a lack of base pairing between nucleotides (or nucleotide analogs) within the loop region.
  • the loop region of an shRNA is a single-stranded region intervening between the sense and antisense strands and may also be referred to as "intervening single-strand".
  • the term "siD/R-NA” refers to a double-stranded polynucleotide molecule which is composed of both RNA and DNA, and includes hybrids and chimeras of RNA and DNA and prevents translation of a target mRNA.
  • a hybrid indicates a molecule wherein a polynucleotide composed of DNA and a polynucleotied composed of RNA hybridize to each other to form the double-stranded molecule; whereas a chimera indicates that one or both of the strands composing the double stranded molecule may contain RNA and DNA.
  • Standard techniques of introducing siD/R-NA into the cell are used.
  • the siD/R-NA includes a sense nucleic acid sequence (also referred to as "sense strand") of the STC2 gene, a antisense nucleic acid sequence (also referred to as "antisense strand”) of the STC2 gene or both.
  • the siD/R-NA may be constructed such that a single transcript has both the sense and complementary antisense nucleic acid sequences from the target gene, e.g., a hairpin.
  • the siD/R-NA may either be a dsD/R-NA or shD/R-NA.
  • dsD/R-NA refers to a construct of two molecules having complementary sequences to one another and that have annealed together via the complementary sequences to form a double-stranded polynucleotide molecule.
  • the nucleotide sequence of two strands may include not only the "sense” or "antisense” polynucleotides sequence selected from a protein coding sequence of target gene sequence, but also polynucleotide having a nucleotide sequnence selected from non-coding region of the target gene.
  • dsD/R-NA refers to an siD/R-NA having a stem-loop structure, having a first and second regions complementary to one another, i.e., sense and antisense strands. The degree of complementarity and orientation of the regions are sufficient such that base pairing occurs between the regions, the first and second regions are joined by a loop region, and the loop results from a lack of base pairing between nucleotides (or nucleotide analogs) within the loop region.
  • an "isolated nucleic acid” is a nucleic acid removed from its original environment (e.g., the natural environment if naturally occurring) and thus, synthetically altered from its natural state.
  • examples of isolated nucleic acid includes DNA, RNA, and derivatives thereof.
  • the double-stranded molecule serves as a guide for identifying homologous sequences in mRNA for the RISC complex, when the double-stranded molecule is introduced into cells.
  • RNA is cleaved and degraded by the nuclease activity of Dicer, through which the double-stranded molecule eventually decreases or inhibits production (expression) of the polypeptide encoded by the RNA.
  • a double-stranded molecule of the present invention can be defined by its ability to generate a single-strand that specifically hybridizes to the mRNA of the STC2 gene under stringent conditions.
  • target sequence or “target nucleic acid” or “target nucleotide”.
  • nucleotide sequence of the "target sequence” can be shown using not only the RNA sequence of the mRNA, but also the DNA sequence of cDNA synthesized from the mRNA.
  • the target sequence of a double-stranded molecule is preferably less than 500, 200, 100, 50, or 25 base pairs in length. More preferably, the target sequences of a double-stranded is 19-25 base pairs in length.
  • the present invention provides the double-stranded molecules having a sense strand and an antisense strand, wherein the sense strand includes a nucleotide sequence corresponding to a target sequence.
  • the sense strand hybridizes with antisense strand at the target sequence to form the double-stranded molecule having between 19 and 25 nucleotide pair in length.
  • target nucleotide sequences of double-stranded molecules targeting the STC2 gene includes the nucleotide sequences of SEQ ID NO: 8 or 9.
  • the nucleotide "t” in the sequence should be replaced with "u” in RNA or derivatives thereof.
  • double-stranded molecules of the present invention can include the nucleotide sequence: 5'- GACGAACAGTCTGAGTATT -3' (SEQ ID NO: 8) or 5'- GCAGGAGCTGGTATTGTAG -3' (SEQ ID NO: 9) as a target sequence.
  • nucleotide "u”, “t " or other nucleotide can be added to the 3'end of the target sequence in the antisense strand and/or sense strand.
  • the number of nucleotides to be added is at least 2, generally 2 to 10, preferably 2 to 5.
  • the added nucleotides form a single strand at the 3'end of the antisense strand and/or sense strand of the double-stranded molecule.
  • Other target sequences of suitable double-stranded molecules for the present invention can be designed using an siRNA design computer program available from the Ambion website (www.ambion.com/techlib/ misc/siRNA_finder.html). The computer program selects target sequences for double-stranded molecules based on the following protocol.
  • Target Sites 1. Beginning with the AUG start codon of the object transcript, scan downstream for AA dinucleotide sequences. Record the occurrence of each AA and the 3' adjacent 19 nucleotides as potential siRNA target sites. Tuschl et al. Genes Cev 1999, 13(24):3191-7 don't recommend designing siRNA to the 5' and 3' untranslated regions (UTRs) and regions near the start codon (within 75 nucleotides) as these may be richer in regulatory protein binding sites. UTR-binding proteins and/or translation initiation complexes may interfere with binding of the siRNA endonuclease complex. 2.
  • the homology search can be performed using BLAST (Altschul SF et al., Nucleic Acids Res 1997, 25:3389-402; J Mol Biol 1990, 215:403-10.), which can be found on the NCBI server at: www.ncbi.nlm.nih.gov/BLAST/. 3. Select qualifying target sequences for synthesis. At Ambion, preferably several target sequences can be selected along the length of the gene to evaluate.
  • the double-stranded molecule of the present invention is directed to a single target sequence or may be directed to a plurality of target sequences.
  • a target sequence for the STC2 gene is meant a nucleotide sequence that is identical to a portion of the STC2 gene (i.e, a polynucleotide within the STC2 gene that is equal in length to and complementary to an double-stranded molecule).
  • the target sequence can include the 5' untranslated (UT) region, the open reading frame (ORF) or the 3' untranslated region of the STC2 gene.
  • the double-stranded molecule can be a nucleic acid sequence complementary to an upstream or downstream modulator of the STC2 gene expression.
  • upstream and downstream modulators include, a transcription factor that binds the STC2 gene promoter, a kinase or phosphatase that interacts with the STC2 polypeptide, a promoter or enhancer of the STC2 gene.
  • a double-stranded molecule of the present invention targeting the above-mentioned targeting sequence of the STC2 gene include isolated polynucleotides that includes any of the nucleic acid sequences of target sequences and/or complementary sequences to the target sequences.
  • Examples of polynucleotides targeting the STC2 gene include those having the sequence of SEQ ID NO: 8 or 9 and/or complementary sequences to these nucleotides.
  • the present invention is not limited to these examples, and minor modifications in the aforementioned nucleic acid sequences are acceptable so long as the modified molecule retains the ability to suppress the expression of the STC2 gene.
  • amino modification in a nucleic acid sequence indicates one, two or several substitutions, deletions, additions or insertions of nucleic acids to the sequence. Typically, a minor modification will be four or fewer, sometimes three or fewer, and often two or fewer substitutions, deletions, additions or insertions of nucleic acids to the sequence.
  • a double-stranded molecule of the present invention can be tested for its ability using the methods utilized in the Examples. In the Examples, the double-stranded molecules having sense strands or antisense strands complementary thereto of three portions of mRNA of the STC2 genes were tested in vitro for their ability to decrease production of the STC2 gene product in prostate cancer cell line according to standard methods.
  • reduction in the STC2 gene product in cells contacted with the candidate double-stranded molecule as compared to cells cultured in the absence of the candidate molecule can be detected by, e.g. RT-PCR using primers for STC2 mRNA mentioned under Example 1, item "Semi-quantitative RT-PCR”. Sequences that decrease the production of the STC2 gene product in in vitro cell-based assays can then be tested for their inhibitory effects on cell growth. Sequences that inhibit cell growth in in vitro cell-based assay can then be tested for their in vivo ability using animals with cancer, e.g. nude mouse xenograft models, to confirm decreased production of the STC2 gene product and decreased cancer cell growth.
  • the term “complementary” refers to Watson-Crick or Hoogsteen base pairing between nucleotides units of a polynucleotide
  • binding means the physical or chemical interaction between two polynucleotides.
  • the polynucleotide includes modified nucleotides and/or non-phosphodiester linkages, these polynucleotides may also bind each other as same manner.
  • complementary polynucleotide sequences hybridize under appropriate conditions to form stable duplexes containing few or no mismatches.
  • the sense strand and antisense strand of the isolated polynucleotide of the present invention can form double-stranded molecule or hairpin loop structure by the hybridization.
  • such duplexes contain no more than 1 mismatch for every 10 matches.
  • such duplexes contain no mismatches.
  • the double-stranded molecules of the present invention may contain one or more modified nucleotides and/or non-phosphodiester linkages.
  • Chemical modifications well known in the art are capable of increasing stability, availability, and/or cell uptake of the double-stranded molecule.
  • the skilled person will be aware of other types of chemical modification which may be incorporated into the present molecules (WO03/070744; WO2005/045037).
  • modifications can be used to provide improved resistance to degradation or improved uptake.
  • modifications include phosphorothioate linkages, 2'-O-methyl ribonucleotides (especially on the sense strand of a double-stranded molecule), 2'-deoxy-fluoro ribonucleotides, 2'-deoxy ribonucleotides, "universal base” nucleotides, 5'-C- methyl nucleotides, and inverted deoxyabasic residue incorporation (US20060122137).
  • modifications can be used to enhance the stability or to increase targeting efficiency of the double-stranded molecule.
  • Modifications include chemical cross linking between the two complementary strands of a double-stranded molecule, chemical modification of a 3' or 5' terminus of a strand of a double-stranded molecule, sugar modifications, nucleobase modifications and/or backbone modifications, 2 -fluoro modified ribonucleotides and 2'-deoxy ribonucleotides (WO2004/029212).
  • modifications can be used to increased or decreased affinity for the complementary nucleotides in the target mRNA and/or in the complementary double-stranded molecule strand (WO2005/044976).
  • an unmodified pyrimidine nucleotide can be substituted for a 2-thio, 5-alkynyl, 5-methyl, or 5-propynyl pyrimidine.
  • an unmodified purine can be substituted with a 7-deaza, 7-alkyl, or 7-alkenyl purine.
  • the 3'- terminal nucleotide overhanging nucleotides may be replaced by deoxyribonucleotides (Elbashir SM et al., Genes Dev 2001 Jan 15, 15(2): 188-200).
  • published documents such as US20060234970 are available.
  • the present invention is not limited to these examples and any known chemical modifications may be employed for the double-stranded molecules of the present invention so long as the resulting molecule retains the ability to inhibit the expression of the target gene.
  • the double-stranded molecules of the present invention may include both DNA and RNA, e.g., dsD/R-NA or shD/R-NA.
  • a hybrid polynucleotide of a DNA strand and an RNA strand or a DNA-RNA chimera polynucleotide shows increased stability.
  • DNA and RNA i.e., a hybrid type double-stranded molecule composed of a DNA strand (polynucleotide) and an RNA strand (polynucleotide), a chimera type double-stranded molecule including both DNA and RNA on any or both of the single strands (polynucleotides), or the like may be formed for enhancing stability of the double-stranded molecule.
  • the hybrid of a DNA strand and an RNA strand may be either where the sense strand is DNA and the antisense strand is RNA, or vice versa, so long as it has an activity to inhibit expression of the target gene when introduced into a cell expressing the gene.
  • the sense strand polynucleotide is DNA and the antisense strand polynucleotide is RNA.
  • the chimera type double-stranded molecule may be either where both of the sense and antisense strands are composed of DNA and RNA, or where any one of the sense and antisense strands is composed of DNA and RNA so long as it has an activity to inhibit expression of the target gene when introduced into a cell expressing the gene.
  • the molecule preferably contains as much DNA as possible, whereas to induce inhibition of the target gene expression, the molecule is required to be RNA within a range to induce sufficient inhibition of the expression.
  • an upstream partial region i.e., a region flanking to the target sequence or complementary sequence thereof within the sense or antisense strands
  • the upstream partial region indicates the 5' side (5'-end) of the sense strand and the 3' side (3'-end) of the antisense strand.
  • regions flanking to 5'-end of sense strand and/or 3'-end of antisense strand are referred to upstream partial region. That is, in preferable embodiments, a region flanking to the 3'-end of the antisense strand, or both of a region flanking to the 5'-end of sense strand and a region flanking to the 3'-end of antisense strand composed of RNA.
  • the chimera or hybrid type double-stranded molecule of the present invention include following combinations.
  • sense strand 5'-[---DNA---]-3' 3'-(RNA)-[DNA]-5' :antisense strand
  • sense strand 5'-(RNA)-[DNA]-3' 3'-(RNA)-[DNA]-5' :antisense strand
  • sense strand 5'-(RNA)-[DNA]-3' 3'-(---RNA---)-5' :antisense strand
  • the upstream partial region preferably is a domain composed of 9 to 13 nucleotides counted from the terminus of the target sequence or complementary sequence thereto within the sense or antisense strands of the double-stranded molecules.
  • preferred examples of such chimera type double-stranded molecules include those having a strand length of 19 to 21 nucleotides in which at least the upstream half region (5' side region for the sense strand and 3' side region for the antisense strand) of the polynucleotide is RNA and the other half is DNA. In such a chimera type double-stranded molecule, the effect to inhibit expression of the target gene is much higher when the entire antisense strand is RNA (US20050004064).
  • the double-stranded molecule may form a hairpin, such as a short hairpin RNA (shRNA) and short hairpin composed of DNA and RNA (shD/R-NA).
  • shRNA or shD/R-NA is a sequence of RNA or mixture of RNA and DNA making a tight hairpin turn that can be used to silence gene expression via RNA interference.
  • the shRNA or shD/R-NA may include the sense target sequence and the antisense target sequence on a single strand wherein the sequences are separated by a loop sequence.
  • the hairpin structure is cleaved by the cellular machinery into dsRNA or dsD/R-NA, which is then bound to the RNA-induced silencing complex (RISC).
  • RISC RNA-induced silencing complex
  • This complex binds to and cleaves mRNAs which match the target sequence of the dsRNA or dsD/R-NA.
  • a loop sequence composed of an arbitrary nucleotide sequence can be located between the sense and antisense strands in order to form a hairpin loop structure.
  • the double-stranded molecule contained in the inventive composition may take the general formula 5'-[A]-[B]-[A']-3', wherein [A] is the sense strand having a target sequence, [B] is an intervening single-strand and [A'] is the antisense strand having a complementary sequence to [A].
  • the target sequence may be selected from group composed of, for example, nucleotide sequences shown in SEQ ID NO :8 and SEQ ID NO 9.
  • the present invention is not limited to these examples, and the target sequence in [A] may be modified sequences from these examples so long as the double-stranded molecule retains the ability to suppress the expression of the targeted PKIB or NAALADL2 gene.
  • the region [A] hybridizes to [A'] to form a loop composed of the region [B].
  • the intervening single-stranded portion [B], i.e., loop sequence may be preferably 3 to 23 nucleotides in length.
  • the loop sequence for example, can be selected from group composed of following sequences (http://www.ambion.com/techlib/tb/tb_506.html).
  • loop sequence composed of 23 nucleotides also provides active siRNA (Jacque JM et al., Nature 2002 Jul 25, 418(6896): 435-8, Epub 2002 Jun 26): CCC, CCACC, or CCACACC: Jacque JM et al., Nature 2002 Jul 25, 418(6896): 435-8, Epub 2002 Jun 26; UUCG: Lee NS et al., Nat Biotechnol 2002 May, 20(5): 500-5; Fruscoloni P et al., Proc Natl Acad Sci USA 2003 Feb 18, 100(4): 1639-44, Epub 2003 Feb 10; and UUCAAGAGA: Dykxhoorn DM et al., Nat Rev Mol Cell Biol 2003 Jun, 4(6): 457-67.
  • the loop sequence can be selected from group composed of AUG, CCC, UUCG, CCACC, CTCGAG, AAGCUU, CCACACC, and UUCAAGAGA; however, the present invention is not limited thereto: 5'- GACGAACAGUCUGAGUAUU -[B]- AAUACUCAGACUGUUCGUC -3' (for target sequence of SEQ ID NO: 8); and 5'- GCAGGAGCUGGUAUUGUAG -[B]- CUACAAUACCAGCUCCUGC -3' (for target sequence of SEQ ID NO: 9).
  • the method of preparing the double-stranded molecule is not particularly limited but it is preferable to use a chemical synthetic method known in the art.
  • sense and antisense single-stranded polynucleotides are separately synthesized and then annealed together via an appropriate method to obtain a double-stranded molecule.
  • Specific example for the annealing includes wherein the synthesized single-stranded polynucleotides are mixed in a molar ratio of preferably at least about 3:7, more preferably about 4:6, and most preferably substantially equimolar amount (i.e., a molar ratio of about 5:5).
  • the mixture is heated to a temperature at which double-stranded molecules dissociate and then is gradually cooled down.
  • the annealed double-stranded polynucleotide can be purified by usually employed methods known in the art.
  • Example of purification methods include methods utilizing agarose gel electrophoresis or wherein remaining single-stranded polynucleotides are optionally removed by, e.g., degradation with appropriate enzyme.
  • the regulatory sequences flanking STC2 sequences may be identical or different, such that their expression can be modulated independently, or in a temporal or spatial manner.
  • the double-stranded molecules can be transcribed intracellularly by cloning STC2 gene templates into a vector containing, e.g., a RNA pol III transcription unit from the small nuclear RNA (snRNA) U6 or the human H1 RNA promoter.
  • snRNA small nuclear RNA
  • Vectors containing a double-stranded molecule of the present invention Also included in the invention are vector compositions containing one or more of the antisense nucleic acids or the double-stranded molecules described herein, and a cell containing the vector.
  • a vector of the present invention preferably encodes an antisense nucleic acid or a double-stranded molecule of the present invention in an expressible form.
  • the phrase "in an expressible form” indicates that the vector, when introduced into a cell, will express the molecule.
  • the vector includes regulatory elements necessary for expression of the antisense nucleic acid or the double-stranded molecule.
  • Such vectors of the present invention may be used for producing the present antisense nucleic acids or double-stranded molecules, or directly as an active ingredient for treating cancer.
  • the present invention provides vectors including each of a combination of polynucleotide having a sense strand nucleic acid and an antisense strand nucleic acid, wherein said sense strand nucleic acid has the nucleotide sequence of SEQ ID NO: 8 or 9, and said antisense strand nucleic acid has a sequence complementary to the sense strand, wherein the transcripts of said sense strand and said antisense strand hybridize to each other to form a double-stranded molecule, and wherein said vectors, when introduced into a cell expressing the STC2 gene, inhibits expression of said gene.
  • the polynucleotide is an oligonucleotide of between about 19 and 25 nucleotides pair in length (e.g., contiguous nucleotides from the nucleotide sequence of SEQ ID NO: 11). More preferably, the combination of polynucleotide includes a single oligonucleotide transcript including the sense strand and the antisense strand linked via a single-stranded nucleotide sequence.
  • the combination of polynucleotide has the general formula 5'-[A]-[B]-[A']-3', wherein [A] is the nucleotide sequence of SEQ ID NO: 8 or 9; [B] is a nucleotide sequence composed of about 3 to about 23 nucleotide; and [A'] is a nucleotide sequence complementary to [A].
  • Vectors of the present invention can be produced, for example, by cloning STC2 gene into an expression vector so that regulatory sequences are operatively-linked to STC2 gene sequence in a manner to allow expression (by transcription of the DNA molecule) of an antisense nucleic acid or both strands of a double-stranded molecule (Lee NS et al., Nat Biotechnol 2002 May, 20(5): 500-5).
  • RNA molecule that is the antisense to mRNA is transcribed by a first promoter (e.g., a promoter sequence flanking to the 3' end of the cloned DNA) and RNA molecule that is the sense strand to the mRNA is transcribed by a second promoter (e.g., a promoter sequence flanking to the 5' end of the cloned DNA).
  • a first promoter e.g., a promoter sequence flanking to the 3' end of the cloned DNA
  • RNA molecule that is the sense strand to the mRNA is transcribed by a second promoter (e.g., a promoter sequence flanking to the 5' end of the cloned DNA).
  • a second promoter e.g., a promoter sequence flanking to the 5' end of the cloned DNA
  • two vectors constructs respectively encoding the sense and antisense strands of the double-stranded molecule are utilized to respectively express the sense and anti-sense strands and then forming a double-stranded molecule construct.
  • the cloned sequence may encode a construct having a secondary structure (e.g., hairpin); namely, a single transcript of a vector contains both the sense and complementary antisense sequences of the target gene.
  • the vectors of the present invention may also be equipped so to achieve stable insertion into the genome of the target cell (see, e.g., Thomas KR & Capecchi MR, Cell 1987, 51: 503-12 for a description of homologous recombination cassette vectors).
  • DNA-based delivery technologies include "naked DNA”, facilitated (bupivicaine, polymers, peptide-mediated) delivery, cationic lipid complexes, and particle-mediated (“gene gun”) or pressure-mediated delivery (see, e.g., US Patent No. 5,922,687).
  • the vectors of the present invention may be, for example, viral or bacterial vectors.
  • expression vectors include attenuated viral hosts, such as vaccinia or fowlpox (see, e.g., US Patent No. 4,722,848). This approach involves the use of vaccinia virus, e.g., as a vector to express nucleotide sequences that encode the antisense nucleic acid or the double-stranded molecule. Upon introduction into a cell expressing the target gene, the recombinant vaccinia virus expresses the molecule and thereby suppresses the proliferation of the cell.
  • Another example of useable vector includes Bacille Calmette Guerin (BCG).
  • BCG vectors are described in Stover et al., Nature 1991, 351: 456-60.
  • a wide variety of other vectors are useful for therapeutic administration and production of the antisense nucleic acids or the double-stranded molecules; examples include adeno and adeno-associated virus vectors, retroviral vectors, Salmonella typhi vectors, detoxified anthrax toxin vectors, and the like. See, e.g., Shata et al., Mol Med Today 2000, 6: 66-71; Shedlock et al., J Leukoc Biol 2000, 68: 793-806; and Hipp et al., In Vivo 2000, 14: 571-85.
  • the present invention provides methods for inhibiting cell growth, i.e., prostate cancer cell growth, by inducing dysfunction of the STC2 gene via inhibiting the expression of the STC2 gene.
  • STC2 gene expression can be inhibited by the aforementioned double-stranded molecules of the present invention that specifically target of the STC2 gene or the vectors of the present invention that can express the double-stranded molecules.
  • the present invention provides methods to treat patients with prostate cancer by administering a double-stranded molecule against STC2 gene or a vector expressing thereof without adverse effect because that genes were hardly detected in normal organs.
  • the prostate cancer is CRPC or aggressive PC (e.g., Gleason score 8-10).
  • the growth of cells expressing a STC2 gene may be inhibited by contacting the cells with a double-stranded molecule against a STC2 gene, a vector expressing the molecule or a composition containing the same.
  • the cell may be further contacted with a transfection agent.
  • Suitable transfection agents are known in the art.
  • the phrase "inhibition of cell growth" indicates that the cell proliferates at a lower rate or has decreased viability as compared to a cell not exposed to the molecule.
  • Cell growth may be measured by methods known in the art, e.g., using the MTT cell proliferation assay.
  • patients suffering from or at risk of developing disease related to STC2 gene may be treated with an administration of at least one of the present double-stranded molecules, at least one vector expressing at least one of the molecules or at least one composition containing at least one of the molecules.
  • patients suffering from prostate cancer may be treated according to the present methods.
  • the type of cancer may be identified by standard methods according to the particular type of tumor to be diagnosed.
  • Prostate cancer may be diagnosed, for example, with PSA, as prostate cancer marker.
  • the diagnosing method of the present invention described in the item " I. Diagnosing cancer " may be used for identifying prostate cancer .
  • patients to be treated by the methods of the present invention are selected by detecting the expression level of STC2 gene in a biopsy specimen from the patient.
  • the biopsy specimen from the subject is confirmed for STC2 gene over-expression by methods known in the art, for example, immunohistochemical analysis or RT-PCR.
  • each of the molecules may have different structures but act at mRNA that matches the same target sequence of STC2 gene.
  • plural kinds of the double-stranded molecules may act on at mRNA that matches different target sequence of same gene.
  • the method may utilize double-stranded molecules directed to STC2 gene.
  • a double-stranded molecule of the present invention may be directly introduced into the cells in a form to achieve binding of the molecule with corresponding mRNA transcripts.
  • a DNA encoding the double-stranded molecule may be introduced into cells as a vector.
  • transfection-enhancing agent such as FuGENE (Roche diagnostics), Lipofectamine 2000 (Invitrogen), Oligofectamine (Invitrogen), and Nucleofector (Wako pure Chemical)
  • FuGENE FuGENE
  • Lipofectamine 2000 Invitrogen
  • Oligofectamine Invitrogen
  • Nucleofector Nucleofector
  • efficacious means that it retards or prevents cancers from forming or prevents or alleviates a clinical symptom of cancer. Efficaciousness is determined in association with any known method for diagnosing or treating the particular tumor type. To the extent that the methods and compositions of the present invention find utility in the context of "prevention” and “prophylaxis”, such terms are interchangeably used herein to refer to any activity that reduces the burden of mortality or morbidity from disease.
  • Prevention and prophylaxis can occur "at primary, secondary and tertiary prevention levels.” While primary prevention and prophylaxis avoid the development of a disease, secondary and tertiary levels of prevention and prophylaxis encompass activities aimed at the prevention and prophylaxis of the progression of a disease and the emergence of symptoms as well as reducing the negative impact of an already established disease by restoring function and reducing disease-related complications. Alternatively, prevention and prophylaxis can include a wide range of prophylactic therapies aimed at alleviating the severity of the particular disorder, e.g. reducing the proliferation and metastasis of tumors.
  • the treatment and/or prophylaxis of cancer and/or the prevention of postoperative recurrence thereof include any of the following steps, such as the surgical removal of cancer cells, the inhibition of the growth of cancerous cells, the involution or regression of a tumor, the induction of remission and suppression of occurrence of cancer, the tumor regression, and the reduction or inhibition of metastasis.
  • Effectively treating and/or the prophylaxis of cancer decreases mortality and improves the prognosis of individuals having cancer, decreases the levels of tumor markers in the blood, and alleviates detectable symptoms accompanying cancer.
  • reduction or improvement of symptoms constitutes effectively treating and/or the prophylaxis include 10%, 20%, 30% or more reduction, or stable disease.
  • a double-stranded molecule of the present invention degrades the target mRNA in sub-stoichiometric amounts. Without wishing to be bound by any theory, it is believed that the double-stranded molecule of the present invention causes degradation of the target mRNA in a catalytic manner. Thus, as compared to standard cancer therapies, the present invention requires the delivery of significantly less double-stranded molecule at or near the site of cancer in order to exert therapeutic effect.
  • an effective amount of the double-stranded molecule of the present invention can be administered to a given subject, by taking into account factors such as body weight, age, sex, type of disease, symptoms and other conditions of the subject; the route of administration; and whether the administration is regional or systemic.
  • an effective amount of the double-stranded molecule of the present invention is an intercellular concentration at or near the cancer site of from about 1 nanomolar (nM) to about 100 nM, preferably from about 2 nM to about 50 nM, more preferably from about 2.5 nM to about 10 nM. It is contemplated that greater or smaller amounts of the double-stranded molecule can be administered.
  • the precise dosage required for a particular circumstance may be readily and routinely determined by one of skill in the art.
  • the present methods can be used to inhibit the growth or metastasis of cancer expressing STC2 gene; for example prostate cancer, especially CRPC or aggressive PC (e.g., Gleason score 8-10).
  • a double-stranded molecule containing a target sequence of SEQ ID NO: 8 or 9 is particularly preferred for the treatment of prostate cancer.
  • the double-stranded molecule of the present invention can also be administered to a subject in combination with a pharmaceutical agent different from the double-stranded molecule.
  • the double-stranded molecule of the present invention can be administered to a subject in combination with another therapeutic method designed to treat cancer.
  • the double-stranded molecule of the present invention can be administered in combination with therapeutic methods currently employed for treating cancer or preventing cancer metastasis (e.g., radiation therapy, surgery and treatment using chemotherapeutic agents).
  • the double-stranded molecule can be administered to the subject either as a naked double-stranded molecule, in conjunction with a delivery reagent, or as a recombinant plasmid or viral vector which expresses the double-stranded molecule.
  • Suitable delivery reagents for administration in conjunction with the present a double-stranded molecule include the Mirus Transit TKO lipophilic reagent; lipofectin; lipofectamine; cellfectin; or polycations (e.g., polylysine), or liposomes.
  • a preferred delivery reagent is a liposome.
  • Liposomes can aid in the delivery of the double-stranded molecule to a particular tissue, such as prostate tissue, and can also increase the blood half-life of the double-stranded molecule.
  • Liposomes suitable for use in the context of the present invention may be formed from standard vesicle-forming lipids, which generally include neutral or negatively charged phospholipids and a sterol, such as cholesterol. The selection of lipids is generally guided by consideration of factors such as the desired liposome size and half-life of the liposomes in the blood stream. A variety of methods are known for preparing liposomes, for example as described in Szoka et al., Ann Rev Biophys Bioeng 1980, 9: 467; and US Pat. Nos.
  • the liposomes encapsulating the present double-stranded molecule includes a ligand molecule that can deliver the liposome to the cancer site.
  • Ligands which bind to receptors prevalent in tumor or vascular endothelial cells such as monoclonal antibodies that bind to tumor antigens or endothelial cell surface antigens, are preferred.
  • the liposomes encapsulating the present double-stranded molecule are modified so as to avoid clearance by the mononuclear macrophage and reticuloendothelial systems, for example, by having opsonization-inhibition moieties bound to the surface of the structure.
  • a liposome of the invention can include both opsonization-inhibition moieties and a ligand.
  • Opsonization-inhibiting moieties for use in preparing the liposomes of the invention are typically large hydrophilic polymers that are bound to the liposome membrane.
  • an opsonization inhibiting moiety is "bound" to a liposome membrane when it is chemically or physically attached to the membrane, e.g., by the intercalation of a lipid-soluble anchor into the membrane itself, or by binding directly to active groups of membrane lipids.
  • These opsonization-inhibiting hydrophilic polymers form a protective surface layer which significantly decreases the uptake of the liposomes by the macrophage-monocyte system ("MMS") and reticuloendothelial system ("RES"); e.g., as described in US Pat. No. 4,920,016, the entire disclosure of which is herein incorporated by reference.
  • Liposomes modified with opsonization-inhibition moieties thus remain in the circulation much longer than unmodified liposomes. For this reason, such liposomes are sometimes called "stealth” liposomes. Stealth liposomes are known to accumulate in tissues fed by porous or "leaky” microvasculature. Thus, target tissue characterized by such microvasculature defects, for example, solid tumors, will efficiently accumulate these liposomes; see Gabizon et al., Proc Natl Acad Sci USA 1988, 18: 6949-53. In addition, the reduced uptake by the RES lowers the toxicity of stealth liposomes by preventing significant accumulation in liver and spleen. Thus, liposomes of the invention that are modified with opsonization-inhibition moieties can deliver the present double-stranded molecule to prostate cancer cells.
  • Opsonization inhibiting moieties suitable for modifying liposomes are preferably water-soluble polymers with a molecular weight from about 500 to about 40,000 daltons, and more preferably from about 2,000 to about 20,000 daltons.
  • Such polymers include polyethylene glycol (PEG) or polypropylene glycol (PPG) derivatives; e.g., methoxy PEG or PPG, and PEG or PPG stearate; synthetic polymers such as polyacrylamide or poly N-vinyl pyrrolidone; linear, branched, or dendrimeric polyamidoamines; polyacrylic acids; polyalcohols, e.g., polyvinylalcohol and polyxylitol to which carboxylic or amino groups are chemically linked, as well as gangliosides, such as ganglioside GM.sub.1.
  • Copolymers of PEG, methoxy PEG, or methoxy PPG, or derivatives thereof, are also suitable.
  • the opsonization inhibiting polymer can be a block copolymer of PEG and either a polyamino acid, polysaccharide, polyamidoamine, polyethyleneamine, or polynucleotide.
  • the opsonization inhibiting polymers can also be natural polysaccharides containing amino acids or carboxylic acids, e.g., galacturonic acid, glucuronic acid, mannuronic acid, hyaluronic acid, pectic acid, neuraminic acid, alginic acid, carrageenan; aminated polysaccharides or oligosaccharides (linear or branched); or carboxylated polysaccharides or oligosaccharides, e.g., reacted with derivatives of carbonic acids with resultant linking of carboxylic groups.
  • the opsonization-inhibiting moiety is a PEG, PPG, or derivatives thereof.
  • Liposomes modified with PEG or PEG-derivatives are sometimes called "PEGylated liposomes".
  • the opsonization inhibiting moiety can be bound to the liposome membrane by any one of numerous well-known techniques.
  • an N-hydroxysuccinimide ester of PEG can be bound to a phosphatidyl-ethanolamine lipid-soluble anchor, and then bound to a membrane.
  • a dextran polymer can be derivatized with a stearylamine lipid-soluble anchor via reductive amination using Na(CN)BH. sub. 3 and a solvent mixture such as tetrahydrofuran and water in a 30:12 ratio at 60 degrees C.
  • Vectors expressing a double-stranded molecule of the present invention are discussed above. Such vectors expressing at least one double-stranded molecule of the invention can also be administered directly or in conjunction with a suitable delivery reagent, including the Mirus Transit LT1 lipophilic reagent; lipofectin; lipofectamine; cellfectin; polycations (e.g., polylysine) or liposomes.
  • a suitable delivery reagent including the Mirus Transit LT1 lipophilic reagent; lipofectin; lipofectamine; cellfectin; polycations (e.g., polylysine) or liposomes.
  • the double-stranded molecule of the present invention can be administered to the subject by any means suitable for delivering the double-stranded molecule into cancer sites.
  • the double-stranded molecule can be administered by gene gun, electroporation, or by other suitable parenteral or enteral administration routes.
  • Suitable enteral administration routes include oral, rectal, or intranasal delivery.
  • Suitable parenteral administration routes include intravesical and intravascular administration (e.g., intravenous bolus injection, intravenous infusion, intra-arterial bolus injection, intra-arterial infusion and catheter instillation into the vasculature); peri- and intra-tissue injection (e.g., peri-tumoral or intra-tumoral injection); subcutaneous injection or deposition including subcutaneous infusion (such as by osmotic pumps); direct application to the area at or near the site of cancer, for example by a catheter or other placement device (e.g., a suppository or an implant including a porous, non-porous, or gelatinous material); and inhalation. It is preferred that injections or infusions of the double-stranded molecule or vector be given at or near the site of the cancer.
  • intravascular administration e.g., intravenous bolus injection, intravenous infusion, intra-arterial bolus injection, intra-arterial infusion and catheter in
  • the double-stranded molecule of the present invention can be administered in a single dose or in multiple doses.
  • the infusion can be a single sustained dose or can be delivered by multiple infusions.
  • Injection of the agent directly into the tissue is at or near the site of cancer preferred. Multiple injections of the agent into the tissue at or near the site of cancer are particularly preferred.
  • One skilled in the art can also readily determine an appropriate dosage regimen for administering the double-stranded molecule of the invention to a given subject.
  • the double-stranded molecule can be administered to the subject once, for example, as a single injection or deposition at or near the cancer site.
  • the double-stranded molecule can be administered once or twice daily to a subject for a period of from about three to about twenty-eight days, more preferably from about seven to about ten days.
  • the double-stranded molecule is injected at or near the site of cancer once a day for seven days.
  • the effective amount of a double-stranded molecule administered to the subject can include the total amount of a double-stranded molecule administered over the entire dosage regimen.
  • compositions containing a double-stranded molecule of the present invention are provided pharmaceutical compositions that include at least one of the present double-stranded molecules or the vectors coding for the molecules.
  • the double-stranded molecules of the present invention are preferably formulated as pharmaceutical compositions prior to administering to a subject, according to techniques known in the art.
  • Pharmaceutical compositions of the present invention are characterized as being at least sterile and pyrogen-free.
  • pharmaceutical formulations include formulations for human and veterinary use. Methods for preparing pharmaceutical compositions of the invention are within the skill in the art, for example as described in Remington's Pharmaceutical Science, 17th ed., Mack Publishing Company, Easton, Pa.
  • the present pharmaceutical formulations contain at least one of the double-stranded molecules or vectors encoding them of the present invention (e.g., 0.1 to 90% by weight), or a physiologically acceptable salt of the molecule, mixed with a physiologically acceptable carrier medium.
  • Preferred physiologically acceptable carrier media are water, buffered water, normal saline, 0.4% saline, 0.3% glycine, hyaluronic acid and the like.
  • the composition may contain plural kinds of double-stranded molecules, each of molecules may be directed to the same target sequence, or different target sequences of STC2 gene.
  • compositions may contain double-stranded molecules directed to one, two or more target sequences of STC2 gene.
  • the present composition may contain a vector coding for one or plural double-stranded molecules.
  • the vector may encode one, two or several kinds of the present double-stranded molecules.
  • the present composition may contain plural kinds of vectors, each of the vectors coding for a different double-stranded molecule.
  • the present double-stranded molecules may be contained as liposomes in the present composition. See under the item of "IV-3. Methods for treating or preventing cancer using a double-stranded molecule of the present invention" for details of liposomes.
  • compositions of the present invention can also include conventional pharmaceutical excipients and/or additives.
  • Suitable pharmaceutical excipients include stabilizers, antioxidants, osmolality adjusting agents, buffers, and pH adjusting agents.
  • Suitable additives include physiologically biocompatible buffers (e.g., tromethamine hydrochloride), additions of chelants (such as, for example, DTPA or DTPA-bisamide) or calcium chelate complexes (for example calcium DTPA, CaNaDTPA-bisamide), or, optionally, additions of calcium or sodium salts (for example, calcium chloride, calcium ascorbate, calcium gluconate or calcium lactate).
  • Pharmaceutical compositions of the invention can be packaged for use in liquid form, or can be lyophilized.
  • a solid pharmaceutical composition for oral administration can include any of the carriers and excipients listed above and 10-95%, preferably 25-75%, of one or more double-stranded molecule of the present invention.
  • a pharmaceutical composition for aerosol (inhalational) administration can include 0.01-20% by weight, preferably 1-10% by weight, of one or more double-stranded molecules of the present invention encapsulated in a liposome as described above, and propellant.
  • a carrier can also be included as desired; e.g., lecithin for intranasal delivery.
  • the present composition may contain other pharmaceutical active ingredients so long as they do not inhibit the in vivo function of the present double-stranded molecules of the present invention.
  • the composition may contain chemotherapeutic agents conventionally used for treating cancers.
  • the present invention provides for the use of the double-stranded nucleic acid molecules of the present invention in manufacturing a pharmaceutical composition for treating a prostate cancer characterized by the expression of STC2 gene.
  • the present invention relates to a use of double-stranded nucleic acid molecule inhibiting the expression of STC2 gene in a cell, which molecule includes a sense strand and an antisense strand complementary thereto, hybridized to each other to form the double-stranded nucleic acid molecule and targets to a sequence selected from among SEQ ID NOs: 8 and 9, for manufacturing a pharmaceutical composition for treating prostate cancer expressing STC2 gene.
  • the present invention further provides a method or process for manufacturing a pharmaceutical composition for treating a prostate cancer characterized by the expression of STC2 gene, wherein the method or process includes a step for formulating a pharmaceutically or physiologically acceptable carrier with a double-stranded nucleic acid molecule inhibiting the expression of STC2 gene in a cell, which over-expresses the gene, which molecule includes a sense strand and an antisense strand complementary thereto, hybridized to each other to form the double-stranded nucleic acid molecule and targets to a sequence selected from among SEQ ID NOs: 8 and 9 as active ingredients.
  • the present invention provides a method or process for manufacturing a pharmaceutical composition for treating a prostate cancer characterized by the expression of STC2 gene, wherein the method or process includes a step for admixing an active ingredient with a pharmaceutically or physiologically acceptable carrier, wherein the active ingredient is a double-stranded nucleic acid molecule inhibiting the expression of STC2 gene in a cell, which over-expresses the gene, which molecule includes a sense strand and an antisense strand complementary thereto, hybridized to each other to form the double-stranded nucleic acid molecule and targets to a sequence selected from among SEQ ID NOs: 8 and 9.
  • Example 1 General Methods 1.
  • Cell lines Human PC cell lines LNCaP, 22Rv1, DU-145, and PC-3 were obtained from American Type Culture Collection (ATCC, Rockville, MD).
  • LNCaP derived androgen-independent prostate cancer cell line C4-2B was purchased from ViroMed Laboratories (Minnetonka, MN, USA). All of the cell lines were cultured as monolayers in the following medium: DMEM (Sigma-Aldrich) for 22Rv1, LNCaP, C4-2B and DU-145; and F-12 (GIBCO) for PC-3 with 10% fetal bovine serum and 1% antibiotic/ antimycotic solution (Sigma-Aldrich). Cells were maintained in incubators containing humidified air with 5% CO 2 at 37 degrees C.
  • ACTB beta-actin
  • the primer sequences employed in the context of the present invention were: ACTB forward: 5'-TTGGCTTGACTCAGGATTTA -3' (SEQ ID NO: 1), ACTB reverse: 5'-ATGCTATCACCTCCCCTGTG-3' (SEQ ID NO: 2), STC2 forward: 5'-TTACTCCATGAGCCTTCCTTTG-3' (SEQ ID NO: 3), and STC2 reverse: 5'- TCCTGTTGCCTAAATCCGTAGTA -3' (SEQ ID NO: 4).
  • RNA from five PC cell lines were extracted using RNeasy Kit (QIAGEN, Valencia, CA), and their mRNAs were purified using the mRNA Purification Kit (GE Healthcare, Piscataway, NJ, USA), according to the manufacturer's protocols.
  • mRNA Purification Kit GE Healthcare, Piscataway, NJ, USA
  • One-micro gram aliquot of each mRNA from the PC cell lines, as well as those isolated from normal human brain, heart, kidney, liver, lung, prostate and testis (BD Bioscience, Palo Alto, CA) were separated on 1% denaturing agarose gels and transferred onto nylon membranes. The membranes were hybridized for 20 hours with 32 P-labeled STC2 cDNA, which was labeled by Megaprime DNA labeling system (GE Bioscience, UK).
  • Probe cDNA of STC2 was prepared as a 551-bp PCR product by using the primers followings: 5'-AACATTTACCATTAGAGAGGGGG-3' (SEQ ID NO: 5) and 5'- CACATAGAAATGACACTCCTCCC-3' (SEQ ID NO: 6). Pre-hybridization, hybridization, and washing were performed according to the manufacturer's instruction. The blots were autoradiographed at -80 degrees C for 7 days.
  • Sections were incubated with a 1:100 diluted solution of purified anti-STC2 mAb (0.75 mg/ml), for 16 minutes.
  • the automated protocol is based on an indirect biotin-avidin system using a biotinylated universal secondary antibody and diaminobenzidine substrate with hematoxylin counterstaining. The specificity of the binding was confirmed by negative staining using mouse nonimmune serum as primary antibody.
  • Scoring of immunohistochemical staining To evaluate both the intensity of staining and proportion of the positive-stained cells, a scoring method previously reported was used (Ashida S, et al., Clin Cancer Res 2006, 2767-73). Regarding the morphology and the intensity of STC2 expression, positive staining of anti-STC2 antibody was defined as follows: score 1 for variable weak cytoplasmic staining, score 2 for segmental and apical granular cytoplasmic staining, and score 3 for diffuse continuous and intense cytoplasmic staining. For each score, the proportion of cells with the score was estimated visually.
  • shRNA short hairpin RNA
  • Plasmids designed to express shRNA were prepared by cloning of double-stranded oligonucleotides into psiU6BX vector.
  • oligonucleotide sequences of target sequences for STC2 are as followed; sense strand sequence for si1: 5'-CAACTCTTGTGAGATTCGG-3' (SEQ ID NO: 7), si2: 5'-GACGAACAGTCTGAGTATT-3' (SEQ ID NO: 8), si3: 5'- GCAGGAGCTGGTATTGTAG -3' (SEQ ID NO: 9), and siEGFP: 5'-GAAGCAGCACGACTTCTTC-3' (SEQ ID NO: 10) as a negative control.
  • PC-3 cells (2X10 6 ) which expressed STC2 in high level were seeded on 10-cm dishes, transfected with psiU6-STC2 (si1-3), or psiU6-siEGFP using FuGene6 (Roche) according to the manufacturer's instruction, and then cultured in appropriate medium containing 800 mcg/ml of Geneticin (Sigma-Aldrich) for 14 days. The cells were fixed with 100% methanol, stained with 0.1% of crystal violet-H20 for colony formation assay. In MTT assay, cell viability was measured using Cell-counting kit-8 (DOJINDO, Kumamoto, Japan) at 10 days after transfection.
  • Example 2 STC2 over expression in CRPC cells
  • STC2 was focused in this invention.
  • Semi-quantitative RT-PCR (Fig. 1A) and real-time quantitative RT-PCR (Fig. 1B) confirmed the elevated expression of STC2 in 6 out of the 7 clinical CRPC cells, comparing with CNPC cells and NP cells.
  • Example 3 Immunohistochemical analysis in clinical PC tissues To validate the over-expression of STC2 protein in CRPC cells, immunohistochemical analysis on clinical PC tissues was performed by using monoclonal antibody specific to human STC2. As shown in Fig. 1E, strong immunochemical signal for STC2 was detected predominantly in the cytoplasm of cancer cells exceptionally in CRPC cases examined. 6 of 9 CRPCs examined showed strong immunoreactivity to anti-STC2 antibody and one CNPC with Gleason score 10 also showed strong immunoreactivity (Fig. 1F), while CNPC with Gleason score 7 did not show any immunoreactivity to anti-STC2 antibody (Fig. 1G). Adjacent normal prostatic epithelium in the same patient revealed very weak or no signal for STC2.
  • Example 4 Knockdown of STC2 expression by siRNA attenuated PC cell growth
  • three vectors designed to express shRNA specifically to STC2 were constructed and transfected into PC cell lines PC-3, which expressed endogenous STC2 most strongly.
  • si2 and si3 showed the significant knockdown effect on endogenous STC2 transcript (Fig. 2A), and this transfection resulted in reduction of the numbers of colonies (Fig. 2B) as well as those of the viable cells measured by MTT assay for PC-3 cells (Fig. 2C).
  • si1 and a negative control showed no or little knockdown effect on STC2 expression and did not affect cell viability of PC-3 cells.
  • Example 5 STC2 over-expression promoted cancer cell growth
  • three stable transformants (Clones 1-3) were established from 22Rv1 cells, in which exogenous STC2 expressed constitutively.
  • control 22Rv1 cells transfected empty vector (Mock) were also prepared and compared their proliferation.
  • Western blot analysis (Fig.2D) confirmed high level of exogenous STC2 expression in three stable clones (Clones 1-3).
  • the present invention provides a novel target molecule or biomarker, STC2, for therapy development or diagnosis for PCs, especially for CRPCs and aggressive PCs.
  • PC shows relatively good prognosis, and hormone-ablation therapy or castration is effective in most of relapsed or advanced PC.
  • doxotaxel plus predonisone Tannock IF, et al., N Engl J Med 2004, 351, 1502-12, and Petrylak DP, et al., N Engl J Med 2004, 351, 1513-20
  • STC2 is a secreted protein that functions to promote PC cell viability in autocrine/paracrine manner. Accordingly, it is possible to detect STC2 in patient serum, to thus serve as a diagnostic biomarker diagnosing PC (more suitably CRPC), or predicting PC aggressiveness and Gleason score, and neutlinization of STC2 by highly specific antibody have potential as one of the therapeutic strategies against PCs, in particular, CRPCs and aggressive PCs.
  • the data provided herein add to a comprehensive understanding of PCs, facilitate development of novel diagnostic strategies, and provide clues for identification of molecular targets for therapeutic drugs and preventative agents. Such information contributes to a more profound understanding of tumorigenesis, and provide indicators for developing novel strategies for diagnosis, treatment, and ultimately prevention of PCs.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Organic Chemistry (AREA)
  • Genetics & Genomics (AREA)
  • Immunology (AREA)
  • Molecular Biology (AREA)
  • Biomedical Technology (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • Biotechnology (AREA)
  • General Health & Medical Sciences (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Analytical Chemistry (AREA)
  • Pathology (AREA)
  • Physics & Mathematics (AREA)
  • Biochemistry (AREA)
  • Microbiology (AREA)
  • General Engineering & Computer Science (AREA)
  • Urology & Nephrology (AREA)
  • Biophysics (AREA)
  • Hospice & Palliative Care (AREA)
  • Oncology (AREA)
  • Hematology (AREA)
  • Medicinal Chemistry (AREA)
  • Public Health (AREA)
  • Cell Biology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Plant Pathology (AREA)
  • Veterinary Medicine (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Endocrinology (AREA)
  • Food Science & Technology (AREA)
  • General Physics & Mathematics (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)

Abstract

La présente invention porte sur des procédés pour la détection et/ou le diagnostic d'un cancer par la détermination du niveau d'expression du gène STC2. Il a été découvert que le gène établissait une distinction entre les cellules cancéreuses et les cellules normales. En outre, la présente invention porte sur des procédés de recherche par criblage d'agents thérapeutiques utiles dans le traitement d'un cancer et sur des procédés pour le traitement d'un cancer. De plus, la présente invention porte sur des molécules double brin ciblant le gène STC2, qui donnent à penser qu'ils sont utiles dans le traitement d'un cancer. Les compositions et procédés de la présente invention trouvent une applicabilité particulière pour un cancer de la prostate, plus spécifiquement un cancer de la prostate résistant à la castration et un cancer de la prostate agressif.
PCT/JP2009/006201 2008-11-20 2009-11-18 Procédés pour le diagnostic ou le traitement d'un cancer de la prostate WO2010058572A1 (fr)

Priority Applications (4)

Application Number Priority Date Filing Date Title
US13/130,284 US8420329B2 (en) 2008-11-20 2009-11-18 Methods for diagnosing or treating prostate cancer
JP2011522180A JP2012509058A (ja) 2008-11-20 2009-11-18 前立腺癌を診断または治療するための方法
EP09827353A EP2367939A4 (fr) 2008-11-20 2009-11-18 Procédés pour le diagnostic ou le traitement d'un cancer de la prostate
US13/830,668 US20130302799A1 (en) 2008-11-20 2013-03-14 Methods for diagnosing or treating prostate cancer

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US19996408P 2008-11-20 2008-11-20
US61/199,964 2008-11-20

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US13/830,668 Division US20130302799A1 (en) 2008-11-20 2013-03-14 Methods for diagnosing or treating prostate cancer

Publications (1)

Publication Number Publication Date
WO2010058572A1 true WO2010058572A1 (fr) 2010-05-27

Family

ID=42198015

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/JP2009/006201 WO2010058572A1 (fr) 2008-11-20 2009-11-18 Procédés pour le diagnostic ou le traitement d'un cancer de la prostate

Country Status (4)

Country Link
US (2) US8420329B2 (fr)
EP (1) EP2367939A4 (fr)
JP (1) JP2012509058A (fr)
WO (1) WO2010058572A1 (fr)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN104459151A (zh) * 2014-11-26 2015-03-25 四川大学 结肠癌诊断试剂、试剂盒
CN109846884A (zh) * 2012-03-15 2019-06-07 西格诺药品有限公司 用tor激酶抑制剂治疗癌症
JP2019194257A (ja) * 2011-04-01 2019-11-07 イミュノジェン, インコーポレイテッド Folr1がん治療の有効性を増加させるための方法

Families Citing this family (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8338109B2 (en) 2006-11-02 2012-12-25 Mayo Foundation For Medical Education And Research Predicting cancer outcome
EP2806054A1 (fr) 2008-05-28 2014-11-26 Genomedx Biosciences Inc. Systèmes et procédés de discrimination basés sur l'expression d'états pathologiques cliniques distincts dans le cancer de la prostate
US10407731B2 (en) 2008-05-30 2019-09-10 Mayo Foundation For Medical Education And Research Biomarker panels for predicting prostate cancer outcomes
US10513737B2 (en) 2011-12-13 2019-12-24 Decipher Biosciences, Inc. Cancer diagnostics using non-coding transcripts
CA2881627A1 (fr) * 2012-08-16 2014-02-20 Genomedx Biosciences Inc. Diagnostic du cancer au moyen de biomarqueurs
EP3504348B1 (fr) 2016-08-24 2022-12-14 Decipher Biosciences, Inc. Utilisation de signatures génomiques en vue d'une prédiction de la réactivité de patients atteints d'un cancer de la prostate à une radiothérapie postopératoire
US11208697B2 (en) 2017-01-20 2021-12-28 Decipher Biosciences, Inc. Molecular subtyping, prognosis, and treatment of bladder cancer
WO2018165600A1 (fr) 2017-03-09 2018-09-13 Genomedx Biosciences, Inc. Sous-typage du cancer de la prostate pour prédire la réponse à une thérapie hormonale
US11078542B2 (en) 2017-05-12 2021-08-03 Decipher Biosciences, Inc. Genetic signatures to predict prostate cancer metastasis and identify tumor aggressiveness

Family Cites Families (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2002098358A2 (fr) * 2001-06-04 2002-12-12 Eos Biotechnology, Inc. Procedes de diagnostic et de traitement du cancer prostatique androgene-dependant, cancer prostatique subissant un retrait d'androgenes et cancer prostatique androgene-independant
WO2003009814A2 (fr) 2001-07-25 2003-02-06 Millennium Pharmaceuticals, Inc. Nouveaux genes, compositions, trousses et methodes d'identification, evaluation, prevention, et traitement du cancer de la prostate
EP1660677A2 (fr) 2003-08-20 2006-05-31 Oncotherapy Science, Inc. Proteine 2 inductible par l'hypoxie (hig2), nouvelle cible therapeutique potentielle de l'hypernephrome (rcc)
WO2007013479A2 (fr) 2005-07-27 2007-02-01 Oncotherapy Science, Inc. Genes et polypeptides associes aux cancers de la prostate
EP1907580A2 (fr) 2005-07-28 2008-04-09 Oncotherapy Science, Inc. Procede pour le diagnostic et le traitement d'hypernephrome

Non-Patent Citations (8)

* Cited by examiner, † Cited by third party
Title
GOO Y.A. ET AL.: "Stromal mesenchyme cell genes of the human prostate and bladder.", BMC UROL., vol. 5, no. 17, 2005, XP021005798 *
ISHIBASHI K. ET AL.: "Molecular cloning of a second human stanniocalcin homologue (STC2).", BIOCHEM. BIOPHYS. RES. COMMUN., vol. 250, 1998, pages 252 - 258, XP002167841 *
JELLINEK D.A. ET AL.: "Stanniocalcin 1 and 2 are secreted as phosphoproteins from human fibrosarcoma cells.", BIOCHEM. J., vol. 350, 2000, pages 453 - 461, XP008140348 *
LIU W. ET AL.: "cDNA microarray analysis of pigment epithelium-derived factor-regulated gene expression profile in prostate carcinoma cells.", INT. J. UROL., vol. 16, January 2009 (2009-01-01), pages 323 - 328, XP008140473 *
See also references of EP2367939A4 *
TAMURA K. ET AL.: "Molecular features of hormone-refractory prostate cancer cells by genome-wide gene expression profiles.", CANCER RES., vol. 67, 2007, pages 5117 - 5125, XP002613537 *
TAMURA K. ET AL.: "Stanniocalcin 2 overexpression in castration-resistant prostate cancer and aggressive prostate cancer.", CANCER SCI., vol. 100, February 2009 (2009-02-01), pages 914 - 919, XP008140350 *
VARGHESE R. ET AL.: "Comparative analysis of mammalian stanniocalcin genes.", ENDOCRINOLOGY, vol. 139, 1998, pages 4714 - 4725, XP008140349 *

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2019194257A (ja) * 2011-04-01 2019-11-07 イミュノジェン, インコーポレイテッド Folr1がん治療の有効性を増加させるための方法
US11135305B2 (en) 2011-04-01 2021-10-05 Immunogen, Inc. Methods for increasing efficacy of FOLR1 cancer therapy
CN109846884A (zh) * 2012-03-15 2019-06-07 西格诺药品有限公司 用tor激酶抑制剂治疗癌症
CN104459151A (zh) * 2014-11-26 2015-03-25 四川大学 结肠癌诊断试剂、试剂盒

Also Published As

Publication number Publication date
EP2367939A1 (fr) 2011-09-28
JP2012509058A (ja) 2012-04-19
US20130302799A1 (en) 2013-11-14
EP2367939A4 (fr) 2012-07-04
US20110294123A1 (en) 2011-12-01
US8420329B2 (en) 2013-04-16

Similar Documents

Publication Publication Date Title
US8420329B2 (en) Methods for diagnosing or treating prostate cancer
US8512944B2 (en) PRMT1 for target genes of cancer therapy and diagnosis
WO2011096211A1 (fr) Gènes whsc1 et whsc1l1 utilisés en tant que gènes cibles dans le cadre d'un traitement anticancéreux et d'un diagnostic associé
WO2012144220A1 (fr) Ezh2 en tant que gène cible pour une thérapie anticancéreuse et le diagnostic du cancer
WO2010095364A1 (fr) Jarid1b en tant que gène cible d'un traitement anticancéreux et d'un diagnostic de cancer
WO2012023285A1 (fr) Ehmt2 comme gène cible pour le traitement et le diagnostic du cancer
WO2010023856A1 (fr) Gène ttll4 associé au cancer du pancréas
WO2010023855A1 (fr) Gène c12orf48 servant de gène cible dans le traitement et le diagnostic du cancer
WO2011161960A1 (fr) C1orf59 pour gènes cibles de thérapie et diagnostic du cancer
WO2012023290A1 (fr) Rasef comme marqueur tumoral et cible thérapeutique pour le cancer
WO2010023864A1 (fr) Oip5 en tant que gène cible pour le traitement et le diagnostic du cancer
WO2011024441A1 (fr) Ercc6l comme gènes cibles pour le diagnostic et la thérapie du cancer
WO2012023286A1 (fr) Lrrc42 comme gène cible pour le traitement et le diagnostic du cancer
WO2012023288A1 (fr) Fam161a comme gène cible pour le traitement et le diagnostic du cancer
WO2012023287A1 (fr) Suv420h1 et suv420h2 comme gènes cibles pour le traitement et le diagnostic du cancer
WO2012023259A1 (fr) C6orf167 comme gène cible pour le traitement et le diagnostic du cancer
WO2011021386A1 (fr) Cstf2 pour des gènes cibles de thérapie et de diagnostic du cancer du poumon
WO2011018898A1 (fr) Cdc45l comme marqueur tumoral et cible thérapeutique pour le cancer

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 09827353

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 2011522180

Country of ref document: JP

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 2009827353

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 13130284

Country of ref document: US