WO2000072679A1 - Therapie genique toxique a base d'osteonectine destinee au traitement de tumeurs et de tissus calcifies - Google Patents

Therapie genique toxique a base d'osteonectine destinee au traitement de tumeurs et de tissus calcifies Download PDF

Info

Publication number
WO2000072679A1
WO2000072679A1 PCT/US2000/014482 US0014482W WO0072679A1 WO 2000072679 A1 WO2000072679 A1 WO 2000072679A1 US 0014482 W US0014482 W US 0014482W WO 0072679 A1 WO0072679 A1 WO 0072679A1
Authority
WO
WIPO (PCT)
Prior art keywords
osn
gene
cells
therapeutic
expression
Prior art date
Application number
PCT/US2000/014482
Other languages
English (en)
Inventor
Leland W. K. Chung
Chia-Ling Hsieh
Kenneth S. Koeneman
Fan Yeung
Original Assignee
University Of Virginia Patent Foundation
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 University Of Virginia Patent Foundation filed Critical University Of Virginia Patent Foundation
Priority to CA002374141A priority Critical patent/CA2374141A1/fr
Priority to JP2000620801A priority patent/JP2003500422A/ja
Priority to EP00937791A priority patent/EP1182929A4/fr
Priority to AU52918/00A priority patent/AU5291800A/en
Priority to IL14679400A priority patent/IL146794A0/xx
Publication of WO2000072679A1 publication Critical patent/WO2000072679A1/fr

Links

Classifications

    • 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
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • 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
    • C12N2830/00Vector systems having a special element relevant for transcription
    • C12N2830/008Vector systems having a special element relevant for transcription cell type or tissue specific enhancer/promoter combination
    • 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
    • C12N2830/00Vector systems having a special element relevant for transcription
    • C12N2830/80Vector systems having a special element relevant for transcription from vertebrates
    • C12N2830/85Vector systems having a special element relevant for transcription from vertebrates mammalian

Definitions

  • the present invention relates to promoters, enhancers and other regulatory elements that direct expression within tumor and tissue cells with calcification potential.
  • it relates to compositions comprising nucleotide sequences from the 5' regulatory region, and transcriptionally active fragments thereof, that control expression of an osteonectin ("OSN").
  • OSN osteonectin
  • transgenic animals wherein an OSN regulatory region is capable of controlling expression of a heterologous coding sequence, over-expressing an endogenous OSN coding sequence or an inhibitor of a pathological process or knocking out expression of a specific gene believed to be important for a calcification-related disease in tumor and tissue cells with calcification potential.
  • the invention also relates to methods for using said vectors, cells and animals for
  • the present invention further relates to compositions and methods for modulating expression of compounds within tumor and tissue cells with calcification potential.
  • the invention further relates to screening compounds that modulate expression
  • the invention further relates to methods of treating tumors and other diseases and disorders involving tumor and tissue cells with calcification potential.
  • Somatic cell gene therapy is a strategy in which a nucleic acid, typically in the form of DNA, is administered to alter the genetic repertoire of target cells for therapeutic purposes.
  • a nucleic acid typically in the form of DNA
  • Gene therapy was originally conceived of as a specific gene replacement therapy for correction of heritable defects to deliver functionally active therapeutic genes
  • ex vivo gene therapy e.g., target cells are removed from the body, transfected or infected with vectors carrying recombinant genes and re- implanted into the body ("autologous cell transfer").
  • ex vivo gene therapy e.g., target cells are removed from the body, transfected or infected with vectors carrying recombinant genes and re- implanted into the body ("autologous cell transfer").
  • transfection techniques are currently available and used to transfer DNA in vitro into cells; including calcium
  • endothelial cells WO89/05345
  • hepatocytes WO89/07136
  • Wilson and Mulligan WO89/07136; Wilson et al., 1990, Proc. Natl. Acad. Sci. 87:8437-8441
  • fibroblasts Palmer et al., 1987, Proc. Natl. Acad. Sci. USA 84:1055- 1059; Anson et al., 1987, Mol. Biol. Med. 4:11-20; Rosenberg et al., 1988, Science
  • tissue-specific promoters The concept of delivery and expression of therapeutic toxic genes to tumor cells through the use of tissue-specific promoters has been well recognized. This approach decreases the toxic effect of therapeutic genes on neighboring normal cells when vector (virus, liposome, etc.) gene delivery results in the infection of the normal cells as well as the cancerous cells. Examples include the uses of -fetoprotein promoter to target hepatoma cells (Koryama, et al., 1991, Cell Struct.
  • CEA carcinoembryonic antigen
  • tissue-specific or tumor-restricted promoters which, in some instances, may be inducible by a hormone, vitamin, an antibiotic, drug or heavy metal
  • ii) the selection of therapeutic (or toxic) genes iii) the appropriate vectors, such as retrovirus, adenovirus, liposomes, etc.
  • a promoter Key to targeting the appropriate tumor tissue while sparing the normal host tissue is a promoter that can home the therapeutic genes to only those tissues which use the chosen promoter.
  • osteotropic tumors such as breast, osteosarcoma and prostate which have metastacised is a major challenge.
  • These seemingly unrelated diseases unite through a molecular analysis of the gene(s) that may be overexpressed in these forms of cancer during disease progression.
  • OSN also known as BM-40, basement membrane-40; SPARC, secreted protein acidic rich in Lysteine
  • BM-40 basement membrane-40
  • SPARC secreted protein acidic rich in Lysteine
  • This protein is synthesized and secreted by some fetal and adult tissues and by a wide spectrum of human cancers.
  • the gene is located at human chromosome 5q31-33.
  • the size of the OSN gene is 26.5 kb with 10 exons separated by 9 introns.
  • the OSN promoter was previously cloned and found to have no TATA or CAAT box. There are two GGA boxes in the human OSN promoter and three GGA boxes in the murine OSN promoter that are 0 important for transcriptional initiation.
  • Exon 1 of the OSN promoter is untranslated.
  • CCTG element that was identified at exon 1 , which is important for AP-2 transcriptional factor binding.
  • the large intron 1 (>10 kb) contains numerous regulatory motifs, including retinoic acid, c-AMP, heat shock 5 proteins, metal ions and growth hormone.
  • OSN is involved in tumor invasion and metastasis. Through their interaction with OSN, tumor cells can be activated to secrete proteolytic enzymes to degrade the basement membrane. OSN-tumor interactions can result in the disruption of cellular adhesion, modulation of cellular proliferation, enhanced permeability and increased cell 0 migration and chemotaxis (Nature Med. 3:144, 1997). Other studies have demonstrated that OSN overexpression was found in most invasive malignant tumors, including those of mammary, ovarian, colon, squamous, endometrium, renal, hepatocellular, gall bladder, pulmonary and prostate cancers.
  • sarcomas such as malignant fibrohistosarcoma, chondrosarcoma, angiosarcoma, hemangioendothelioma, osteosarcoma, 5 giant cell tumor of bone, neuroendocrine tumors such as carcinoids and small cell carcinoma of the lung, glioma, B cell lymphoma, seminoma, and melanoma also have overexpression of OSN.
  • OSN overexpression correlates with neoplastic progression of breast cancer, colorectal cancer and myeloma.
  • suppression of OSN expression by antisense RNA strategy abrogates the tumorigenicity of human melanoma cells (Nature Med. 3:17 1, 1997).
  • the invention disclosed herein provides a model for osteotropic-specific gene transcription.
  • the invention is based in part on the identification of a novel therapeutic o agent for treating, curing and/or ameliorating tumors with calcification potential, including, but not limited to, localized or disseminated osteosarcoma, lung, colon, melanoma, thyroid, brain, multiple myeloma, gastric, ovarian, and especially including, without limitation, breast and prostate cancers.
  • the invention specifically targets sites of metastases of the above mentioned osteotropic tumors, and where applicable, their supporting osseous stroma 5 in the metastatic environment.
  • the present invention also relates to therapeutic agents which may also be applicable to benign conditions, such as benign prostatic hyperplasia (BPH) or arterial sclerotic conditions where calcification occurs.
  • benign prostatic hyperplasia BPH
  • arterial sclerotic conditions where calcification occurs.
  • the osteonectin (“OSN”) promoter represents a novel sequence which has high activity in osteotropic tumors, and can be used as a powerful tool to direct the action of 0 a chosen therapeutic gene to these osteotropic tumors in a tumor and tissue-restricted fashion.
  • the best studied therapeutic gene is herpes simplex virus thymidine kinase (HSVTK or TK) gene.
  • Herpes simplex virus-TK converts the pro-drug ACV (or related drug) to a phosphorylated form that is cytotoxic to dividing cells (Moolten, F.L., 1996, Cancer Research, 46: 5276-5281).
  • a number of methods of vector delivery can be implemented, including injection, subcutaneous delivery, intraperitoneal delivery, intratumor delivery, intralesion delivery, intravenous delivery, intraosseous delivery, delivery by inhalation or loco-regionally by perfusion.
  • Central to 5 effective gene therapy is the choice of a tumor-specific promoter. Because of the poor response rate of previously treated patients with relapsed prostate cancer (or other osteotropic tumors) to conventional radiotherapy, surgery, or chemotherapy, it is important to develop new therapeutic approaches that can be applied either independently or in conjunction with current or other novel treatment modalities.
  • the current invention provides the major advance of identifying a novel therapeutic gene that drives expression of therapeutic or toxic genes in a tumor and tissue-specific manner.
  • the instant invention provides, for the first time, ter alia, the identification of a novel therapeutic gene comprising the OSN promoter to direct osteotropic-specific expression, both in vitro in cultured osteotropic cells, and in vivo in transgenic animals.
  • the present invention provides a novel therapeutic composition comprising an OSN promoter that drives the expression of a therapeutic or toxic gene, for example herpes simplex virus thymidine kinase (TK) which is delivered by a vector, such as a recombinant adenovirus (Ad), to a variety of human tumors or benign tissues that exhibit the ability to calcify either in the primary or at metastatic sites.
  • TK herpes simplex virus thymidine kinase
  • Ad recombinant adenovirus
  • osteosarcoma tumors includes any osteotropic aggressive metastatic tumor such as, for example, localized or disseminated osteosarcoma, lung, colon, melanoma, thyroid, brain, multiple myeloma, gastric, ovarian, and especially including, without limitation, breast and prostate cancers.
  • osteotropic aggressive metastatic tumor such as, for example, localized or disseminated osteosarcoma, lung, colon, melanoma, thyroid, brain, multiple myeloma, gastric, ovarian, and especially including, without limitation, breast and prostate cancers.
  • Non-tumor cells which have the ability to calcify and thus express OSN also are able to express high levels of the recombinant reporter or therapeutic genes of the present invention.
  • the present invention also provides a method of treating osteosarcoma or prostate cancer or other osteotropic tumors that are able to use the OSN promoter by the above routes of recombinant adenovirus Ad-OSN-TK administration in combination with a prodrug, most commonly acyclovir (ACV), although the OSN promoter-driven therapy is not limited to a specific vector or therapeutic gene. Indeed, conceivably, many forms of vector or toxic gene can be generated and combined with this novel OSN promoter-driven strategy to obtain a desired antitumor effect.
  • a prodrug most commonly acyclovir
  • the present invention will effectively eliminate prostate and osteosarcoma or other osteotropic tumors, including, but not limited to, lung, colon, melanoma, thyroid, brain, multiple myeloma, and breast cancers growth both in vitro, and in vivo as localized and as osseous metastatic deposits.
  • the present invention provides compositions and methods for screening compounds that modulate expression within osteotropic cells and tissues.
  • compositions comprising nucleotides from the human OSN promoter, and transcriptionally active fragments thereof, as well as nucleic acids that hybridize under highly stringent conditions to such nucleotides, that control the expression of an osteotropic- specific gene.
  • expression vectors comprising the OSN promoter, and transcriptionally active fragments thereof, operably associated to a heterologous reporter gene, e.g., luciferase, and host cells and transgenic animals containing such vectors.
  • the invention also provides methods for using such vectors, cells and animals for screening candidate molecules for agonists and antagonists of osteotropic-related disorders. Methods for using molecules and compounds identified by the screening assays for therapeutic treatments also are provided.
  • a composition comprising a reporter gene is operatively linked to an OSN promoter.
  • the OSN driven reporter gene is expressed as a transgene in animals.
  • the transgenic animal, and cells derived from osteotropic cells of such transgenic animal can be used to screen compounds for candidates useful for modulating osteotropic-related disorders.
  • such compounds are likely to interfere with the function of trans-acting factors, such as transcription factors, cis-acting elements, such as promoters and enhancers, as well as any class of post-transcriptional, translational or post-translational compounds involved in osteotropic-related disorders.
  • the invention provides methods for high throughput screening of compounds that modulate specific expression of genes within osteotropic cells and tissues.
  • cells from osteotropic-tissues are removed from the transgenic animal and cultured in vitro.
  • the expression of the reporter gene is used to monitor osteotropic-specific gene activity.
  • luciferase is the reporter gene.
  • the transgenic animal models of the invention can be used for in vivo screening to test the mechanism of action of candidate drugs for their effect on osteotropic-related disorders.
  • the effects of the drugs on osteotropic-related disorders including, but not limited to, localized or disseminated osteosarcoma, lung, colon, melanoma, thyroid, brain, multiple myeloma, gastric, ovarian, and especially including, without limitation, breast and prostate cancers, and benign conditions, such as BPH or arterial sclerotic conditions where calcification occurs, can be assayed.
  • a gene therapy method for treating and/or preventing osteotropic-related disorders comprises OSN promoter sequences are used to drive osteotropic-specific expression of toxic or therapeutic molecules and introduced in the osteotropic cells.
  • the method comprises introducing an OSN promoter sequence operatively associated with a nucleic acid encoding a toxic or therapeutic molecule into osteotropic cells.
  • the invention provides a preventative gene therapy method comprising introducing an OSN promoter sequence operatively associated with a nucleic acid encoding a toxic or therapeutic molecule into osteotropic cells to delay and/or prevent an osteotropic-related disorder.
  • the invention provides a gene therapy method for treatment of cancer or other proliferative disorder, including, but not limited to, localized or disseminated osteosarcoma, lung, colon, melanoma, thyroid, brain, multiple myeloma, gastric, ovarian, and especially including, without limitation, breast and prostate cancers.
  • the OSN promoter sequence is used to direct the expression of one or more proteins specifically in the osteotropic-tumor cells of a patient.
  • therapeutic and/or toxic agents are effective not only when administered via direct application, such as by injection, but also when administered systemically to the body via intravenous administration, intra-arterial administration, intra-tumoral administration, perfusion, oral administration or the like, because gene expression will be limited and localized to specific cell and tissue types.
  • therapeutic and/or toxic agents of the invention exhibit pleiotropic effects, expression of the therapeutic and/or toxic agents in only specifically targeted cells is essential in order to prevent numerous, harmful side effects.
  • the present invention encompasses vectors using inducible promoters.
  • Inducible promoters have the advantage that they can be switched on and off, depending on the clinical state of the patient. Therefore, if a cell is stably transfected with a therapeutic transgene under the control of an inducible promoter, its expression could be controlled over the life-time of an individual.
  • the invention further provides methods for screening for novel transcription factors that modulate the OSN promoter sequence.
  • novel transcription factors identified by this method can be used as targets for treating osteotropic-related disorders.
  • TK thymidine kinase
  • OSN osteonectin
  • FBS fetal bovine serum
  • Beta-gal. beta-galactosidase
  • CMV cytomegalovirus
  • ROS 17/2.7 rat osteoblastic osteosarcoma
  • MG 63 human osteosarcoma
  • NIH 3T3 embryonic mouse fibroblast
  • P69 human "normal" prostate cell type without any tumorogenic or metastatic ability
  • LNCaP human androgen dependent prostate cancer
  • C4-2 human androgen independent highly tumorogenic/metastasizing prostate cancer
  • PC-3M human androgen independent highly metastatic prostate cancer
  • ArCaP human androgen independent prostate cancer
  • Saos-2 human osteosarcoma
  • Dl mouse embryonic pluripotent bone marrow cell
  • MCF-7 human breast cancer
  • U-97 human brain cancer of the gioblastoma multiform type
  • A547 human lung cancer
  • DMEM Dulbeco's Modified Eagle Media
  • T media prostate cancer cell optimal growth media
  • RLU relative luciferase units
  • FIG. 1 Schematic Representation of the Strategy used to Construct OSN deletion constructs.
  • FIG. 3 Promoter Activity of OSN Deletion Constructs in Prostatic and Non-prostatic Cell Lines.
  • FIG. 4 Western Blot Revealing OSN Protein Expression in various Prostatic and Non-prostatic Cell Lines.
  • FIG. 5 Northern Blot Revealing OSN Expression (ON) in Various Prostate Cancer Cell Lines.
  • Figure 7 Schematic Representation of a 2.3 kb Human Osteonectin Gene Promoter.
  • Figure 8 Promoter Activity of OSN Deletion Constructs in Various Cancer Cell Lines.
  • Figure 10A-10B In Vitro Cytotoxicity Assay with AD-522E-TK.
  • Figure 10A represents the assay with PC3M cells.
  • Figure 9B represents the assay with MG63 cells.
  • Figure 11 OSN Regulatory Region Sequence from -522 to +62.
  • FIG. 12 PC3M Subcutanous Tumor Growth in Athymic Mice Injected with Ad-522E-TK. 5 DETAILED DESCRIPTION OF THE INVENTION
  • the present invention provides promoters, enhancers and other regulatory elements that direct expression within osteotropic cells, comprising nucleotide sequences from the 5' regulatory region, and transcriptionally active fragments thereof, that control expression of an OSN.
  • expression vectors, host cells and transgenic animals wherein an OSN regulatory region is capable of controlling expression of a heterologous coding sequence, over-expressing an endogenous OSN gene or an inhibitor of a pathological process or knocking out expression of a specific gene believed to be important for a calcification-related disease in tumor and tissue cells with calcification potential.
  • the invention also provides methods for using said vectors, cells and animals for screening candidate molecules for agonists and antagonists of disorders involving tumor and tissue cells with calcification potential.
  • the invention provides compositions and methods for modulating expression of compounds within tumor and tissue cells with calcification potential, and to screening compounds that modulate expression within tumor and tissue cells with calcification potential. Methods for using the molecules and compounds identified by the screening assays for therapeutic treatments also are provided.
  • the invention further provides methods of treating and/or ameliorating tumors and other diseases and disorders with calcification potential, including, but not limited to, localized or disseminated osteosarcoma, lung, colon, melanoma, thyroid, brain, multiple myeloma, breast and prostate cancers.
  • the invention specifically targets sites of metastases of the above mentioned osteotropic tumors, and where applicable, their supporting osseous stroma in the metastatic environment.
  • the present invention provides therapeutic agents which may be applicable to benign conditions such as benign prostatic hyperplasia (BPH) or arterial sclerotic conditions where calcification occurs.
  • the invention is based, in part, on the discovery that nucleotide sequences encoding toxic and/or therapeutic coding sequences contained within vectors (i.e. viral vectors) can be administered in a cell and tissue specific manner, with the use of promoters which allow for tissue specific expression of the nucleotide sequences. Further, because the vectors of the invention utilize these promoters to control the expression of toxic and/or therapeutic coding sequences, the vectors of the invention are effective therapeutic agents not only when administered via direct application, but also when administered systemically to the body, because the toxic and/or therapeutic coding sequences will be expressed only in specifically targeted cells, i.e., within tumor and tissue cells with calcification potential.
  • vectors i.e. viral vectors
  • the methods of the present invention are designed to efficiently transfer one or more DNA molecules encoding therapeutic agents to a site where the therapeutic agent is necessary.
  • the methods involve the administration of a vector containing DNA encoding translational products (i.e. therapeutic proteins) or transcriptional products (i.e. antisense or ribozymes) within a mammalian host to a site where the translational product is necessary.
  • translational products i.e. therapeutic proteins
  • transcriptional products i.e. antisense or ribozymes
  • the present invention relates also to pharmaceutical compositions comprising vectors containing DNA for use in treating and/or ameliorating osteotropic- related disorders, including, but not limited to, localized or disseminated osteosarcoma, lung, colon, melanoma, thyroid, brain, multiple myeloma, breast and prostate cancers, and benign conditions, such as, for example, benign prostatic hyperplasia (BPH) or arterial sclerotic conditions where calcification occurs.
  • the compositions of the invention generally are comprised of a bio-compatible material containing the vector containing DNA encoding a therapeutic protein of interest, i.e., thymidine kinase, growth factors, etc.
  • a bio- compatible composition is one that is in a form that does not produce an allergic, adverse or other untoward reaction when administered to a mammalian host.
  • the invention overcomes shortcomings specifically associated with current recombinant protein therapies for treating and/or ameliorating osteotropic diseases.
  • direct gene transfer is a rational strategy that allows transfected cells to (a) make physiological amounts of therapeutic protein, modified in a tissue- and context-specific manner, and (b) deliver this protein to the appropriate cell surface signaling receptor under the approp ⁇ ate circumstances. Exogenous delivery of such molecules is expected to be associated with significant dosing and delivery problems.
  • repeated administration while possible, is not required with the methods of the invention because various promoters, including inducible promoters, can be used to control the level of expression of the therapeutic protein of interest. Further, integration of transfected DNA can be associated with long term recombinant protein expression.
  • Sections 5.1 and 5.2 are nucleotide sequences of the OSN regulatory region, and expression vectors, host cells and transgenic animals wherein the expression of a heterologous coding sequence is controlled by the OSN regulatory region.
  • Section 5.3 methods for using such polynucleotides (i.e., regulatory regions of the OSN gene) and fusion protein products, for screening compounds that interact with the regulatory region of the OSN gene are described.
  • This Section describes both in vivo and in vitro assays to screen small molecules, compounds, recombinant proteins, peptides, nucleic acids, antibodies, etc. which bind to or modulate the activity of the OSN regulatory region.
  • Section 5.4 describes methods for the use of the compositions of the invention, identified agonists and antagonists for drug delivery or gene therapy.
  • pharmaceutical compositions are described for using such compositions, agonists and antagonists to modulate osteotropic-related disorders.
  • Methods and compositions are provided for treating various osteotropic-related disorders, including, but not limited to, localized or disseminated osteosarcoma, lung, colon, melanoma, thyroid, o brain, multiple myeloma, breast and prostate cancers, and benign conditions, including, but not limited to, benign prostatic hyperplasia (BPH) or arterial sclerotic conditions where calcification occurs.
  • BPH benign prostatic hyperplasia
  • the present invention encompasses polynucleotide sequences comprising the
  • the present invention provides polynucleotides comprising about 2.3 kb, 1.5 kb, 1.1 kb, 0.6 and 0.2 kb sequences that are located within an OSN gene.
  • the polynucleotides comprise -1409 bp through +904 bp, -1409 bp through +73 bp, -120 bp 0 through +904 bp and -120 bp through +73 bp of the OSN sequence shown in Figure 1 and -522 bp through +39, -522 bp through +62 and -522 bp through +73 of the OSN sequence shown in Figure 11.
  • the polynucleotide may be 5000, 4000, 3000, 2000, 1000 and preferably approximately 500 bp in length.
  • the invention further provides probes, primers and fragments of the OSN 5 regulatory region.
  • purified nucleic acids consisting of at least 8 nucleotides (i.e., a hybridizable portion) of an OSN gene sequence are provided; in other embodiments, the nucleic acids consist of at least 20 (contiguous) nucleotides, 25 nucleotides, 50 nucleotides, 100 nucleotides, 200 nucleotides, 500, 1000, 2000, 3000, 4000 or 5000 nucleotides of an OSN sequence. Methods which are well known to those skilled in 0 the art can be used to construct these sequences, either in isolated form or contained in expression vectors.
  • nucleic acids are smaller than 20, 25, 35, 200 or 500 nucleotides in length. Nucleic acids can be single or double stranded.
  • the invention also encompasses nucleic acids hybridizable to or complementary to the foregoing sequences.
  • nucleic acids are provided which comprise a sequence complementary to at least 10, 20, 25, 50, 100, 200, 500 nucleotides or the entire regulatory region of an OSN gene.
  • the probes, primers and fragments of the OSN regulatory region provided by the present invention can be used by the research community for various purposes. They can be used as molecular weight markers on Southern gels; as chromosome markers or tags o (when labeled) to identify chromosomes or to map related gene positions; to compare with endogenous DNA sequences in patients to identify potential genetic disorders; as probes to hybridize and thus discover novel, related DNA sequences; as a source of information to derive PCR primers for genetic fingerprinting; and as a probe to "subtract-out" known sequences in the process of discovering other novel polynucleotides. Methods for 5 performing the uses listed above are well known to those skilled in the art.
  • nucleotide sequences of the invention also include nucleotide sequences that have at least 65%, 70%, 75%, 80%, 85%, 90%, 95%, 98% or more nucleotide sequence identity to the nucleotide sequence depicted in Figure 1 , and/or transcriptionally active fragments thereof, which are capable of driving expression specifically within tumor and tissue cells with calcification potential. 5
  • the sequences are aligned for optimal comparison purposes (e.g., gaps can be introduced in the sequence of a first amino acid or nucleic acid sequence for optimal alignment with a second amino or nucleic acid sequence).
  • the amino acid residues or nucleotides at corresponding amino acid positions or nucleotide positions are then 0 compared.
  • a position in the first sequence is occupied by the same amino acid residue or nucleotide as the corresponding position in the second sequence, then the molecules are identical at that position.
  • the two -> sequences are the same length.
  • the determination of percent identity between two sequences also can be accomplished using a mathematical algorithm.
  • a preferred, non-limiting example of a mathematical algorithm utilized for the comparison of two sequences is the algorithm of Kariin and Altschul (1990) Proc.
  • Gapped BLAST can be utilized as described in Altschul et al. (1997) Nucleic Acids i?e.s.25:3389-3402.
  • PSI-Blast can be used to perform an iterated search which detects distant relationships between molecules (Id.).
  • the default parameters of the respective programs e.g., XBLAST and NBLAST
  • Another preferred, non-limiting example of a mathematical algorithm utilized for the comparison of sequences is the algorithm of Myers and Miller, (1988) CABIOS :11-17.
  • alignments can be obtained using the NA_MULTIPLE_ALIGNMENT 1.0 program, using a Gap Weight of 5 and a GapLength Weight of 1.
  • the percent identity between two sequences can be determined using techniques similar to those described above, with or without allowing gaps. In calculating percent identity, typically only exact matches are counted.
  • the invention also encompasses:
  • DNA expression vectors that contain any of the foregoing OSN regulatory element sequences operatively associated with a heterologous gene, such as a reporter gene; and (c) genetically engineered host cells that contain any of the foregoing OSN regulatory element sequences operatively associated with a heterologous gene such that the OSN regulatory element directs the expression of the heterologous gene in the host cell.
  • heterologous gene such as a reporter gene
  • genetically engineered host cells that contain any of the foregoing OSN regulatory element sequences operatively associated with a heterologous gene such that the OSN regulatory element directs the expression of the heterologous gene in the host cell.
  • various transcriptionally active fragments of this regulatory region are also encompassed within the scope of the invention.
  • a “transcriptionally active" or “transcriptionally functional” fragment of the sequence depicted in Figure 1 according to the present invention refers to a polynucleotide comprising a fragment of said polynucleotide which is functional as a regulatory region for expressing a recombinant polypeptide or a recombinant polynucleotide in a recombinant cell host.
  • a nucleic acid or polynucleotide is "transcriptionally active" as a regulatory region for expressing a recombinant polypeptide or a recombinant polynucleotide if said regulatory polynucleotide contains nucleotide sequences which contain transcriptional information, and such sequences are operably associated to nucleotide sequences which encode the desired polypeptide or the desired polynucleotide.
  • the transcriptionally active fragments of the OSN regulatory region of the present invention encompass those fragments that are of sufficient length to promote transcription of a heterologous gene, such as a reporter gene, when operatively linked to the OSN regulatory sequence and transfected into tumor and tissue cells with calcification potential.
  • the regulatory region is placed immediately 5' to, and is operatively associated with the coding sequence.
  • the term "operatively associated” refers to the placement of the regulatory sequence immediately 5' (upstream) of the reporter gene, such that trans-acting factors required for initiation of transcription, such as transcription factors, polymerase subunits and accessory proteins, can assemble at this region to allow RNA polymerase dependent transcription initiation of the reporter gene.
  • the polynucleotide sequence chosen may further comprise other nucleotide sequences, either from the OSN gene, or from a heterologous gene.
  • multiple copies of a promoter sequence, or a fragment thereof may be linked to each other.
  • the promoter sequence, or a fragment thereof may be linked to another copy of the promoter sequence, or another fragment thereof, in a head to tail, head to head, or tail to tail orientation.
  • an osteotropic-specific enhancer may be operatively linked to the OSN regulatory sequence, or fragment thereof, and used to enhance transcription from the construct containing the OSN regulatory sequence.
  • nucleotide sequence without substantially affecting its transcriptional activities. Such modifications include additions, deletions and substitutions.
  • nucleotide sequence that selectively hybridizes to the complement of the sequence depicted in Figure 1 under stringent conditions, and is capable of activating the expression of a coding sequence specifically within tumor and tissue cells with calcification potential is encompassed by the invention.
  • Exemplary moderately stringent hybridization conditions are as follows: prehybridization of filters containing DNA is carried out for 8 hours to overnight at 65 °C in buffer composed of 6X SSC, 50 mM Tris-HCl (pH 7.5), 1 mM EDTA, 0.02% PVP, 0.02% Ficoll, 0.02%o BSA, and 500 ⁇ g/m(> denatured salmon sperm DNA. Filters are hybridized for 48 hours at 65 °C in prehybridization mixture containing 100 ⁇ g/mC denatured salmon sperm DNA and 5-20 X 10 6 cpm of 32 P-labeled probe.
  • exemplary conditions of high stringency are as follows: e.g., hybridization to filter-bound DNA in 0.5 M NaHPO 4 , 7% sodium dodecyl sulfate (SDS), 1 mM EDTA at 65 °C, and washing in 0.1xSSC/0.1% SDS at 68 °C (Ausubel F.M. et al., eds., 1989, Current Protocols in Molecular Biology, Vol. I.
  • TM melting temperature
  • hybridization is carried out at about 20-25 degrees below Tm (for DNA-DNA hybrids) or 10-15 degrees below Tm (for RNA- DNA hybrids).
  • the OSN regulatory region, or transcriptionally functional fragments thereof is preferably derived from a mammalian organism. Screening procedures which rely on nucleic acid hybridization make it possible to isolate gene sequences from various organisms.
  • the isolated polynucleotide sequence disclosed herein, or fragments thereof may be labeled and used to screen a cDNA library constructed from mRNA obtained from appropriate cells or tissues (e.g., calcified tissue) derived from the organism of interest.
  • the hybridization conditions used should be of a lower stringency when the cDNA library is derived from an organism different from the type of organism from which the labeled sequence was derived.
  • mammalian OSN regulatory region homologues may be isolated from, for example, bovine or other non-human nucleic acid, by performing polymerase chain reaction (PCR) amplification using two primer pools designed on the basis of the nucleotide sequence of the OSN regulatory region disclosed herein.
  • the template for the reaction may be cDNA obtained by reverse transcription of the mRNA prepared from, for example, bovine or other non-human cell lines, or tissue known to express the OSN gene. For guidance regarding such conditions, see, e.g., Innis et al.
  • Promoter sequences within the 5 ' non-coding regions of the OSN gene may be further defined by constructing nested 5 ' and or 3 ' deletions using conventional techniques such as exonuclease III or appropriate restriction endonuclease digestion. The resulting deletion fragments can be inserted into the promoter reporter vector to determine whether the deletion has reduced or obliterated promoter activity, such as described, for example, by Coles et al. (Hum. Mol. Genet., 7:791-800, 1998).
  • promoters may be defined.
  • potential individual regulatory sites within the promoter may be identified using site directed mutagenesis or linker scanning to obliterate potential transcription factor binding sites within the promoter individually or in combination.
  • the effects of these mutations on transcription levels may be determined by inserting the mutations into cloning sites in promoter reporter vectors.
  • assays are well known to those skilled in the art (WO 97/17359, US 5,374,544, EP 582 796, US 5,698,389, US 5,643,746, US5,502,176, and US 5,266,488).
  • OSN regulatory regions and transcriptionally functional fragments thereof, and the fragments and probes described herein which serve to identify OSN regulatory regions and fragments thereof may be produced by recombinant DNA technology using techniques well known in the art. Methods which are well known to those skilled in the art can be used to construct these sequences, either in isolated form or contained in expression vectors. These methods include, for example, in vitro recombinant DNA techniques, synthetic techniques and in vivo genetic recombination. See, e.g., the techniques described in Sambrook et al. , 1989, supra, and Ausabel et al. , 1989, supra; also see the techniques described in "Oligonucleotide Synthesis", 1984, Gait M.J. ed., IRL Press,
  • Alterations in the regulatory sequences can be generated using a variety of chemical and enzymatic methods which are well known to those skilled in the art. For example, regions of the sequences defined by restriction sites can be deleted.
  • Oligonucleotide-directed mutagenesis can be employed to alter the sequence in a defined way and/or to introduce restriction sites in specific regions within the sequence.
  • deletion mutants can be generated using DNA nucleases such as Bal31 ,
  • ExoIII or SI nuclease.
  • Progressively larger deletions in the regulatory sequences are generated by incubating the DNA with nucleases for increased periods of time (see, e.g.,
  • altered sequences are evaluated for their ability to direct expression of heterologous coding sequences in appropriate host cells. It is within the scope of the present invention that any altered regulatory sequences which retain their ability to direct expression of a coding sequence be incorporated into recombinant expression vectors for further use.
  • the OSN regulatory region shows selective tissue and cell-type specificity; i- e. , i induces gene expression in osteotropic cells.
  • the regulatory region, and transcriptionally active fragments thereof, of the present invention may be used to induce expression of a heterologous coding sequence specifically in osteotropic cells.
  • the present invention provides for the use of the OSN regulatory region to achieve tissue specific expression of a target coding sequence.
  • the activity and the specificity of the OSN regulatory region can further be assessed by monitoring the expression level of a detectable polynucleotide operably associated with the OSN promoter in different types of cells, tissues and cell lines engineered to contain the OSN promoter.
  • the detectable polynucleotide may be either a polynucleotide that specifically hybridizes with a predefined oligonucleotide probe, or a polynucleotide encoding a detectable protein.
  • the regulatory polynucleotides according to the invention may be advantageously part of a recombinant expression vector that may be used to express a coding sequence, or reporter gene, in a desired host cell or host organism.
  • the OSN regulatory region of the present invention, and transcriptionally active fragments thereof, may be used to direct the expression of a heterologous coding sequence.
  • the present invention encompasses mammalian OSN regulatory regions.
  • transcriptionally active fragments of the OSN regulatory region encompass those fragments of the region which are of sufficient length to promote transcription of a reporter coding sequence to which the fragment is operatively linked.
  • reporter gene sequences well known to those of skill in the art can be utilized, including, but not limited to, genes encoding fluorescent proteins such as green fluorescent protein (GFP), enzymes (e.g. CAT, beta-galactosidase, luciferase) or antigenic markers.
  • GFP green fluorescent protein
  • enzymes e.g. CAT, beta-galactosidase, luciferase
  • antigenic markers e.g. CAT, beta-galactosidase, luciferase
  • enzymatic reporters and light-emitting reporters analyzed by colorometric or fluorometric assays are preferred for the screening assays of the
  • a bioluminescent, chemiluminescent or fluorescent protein can be used as a light-emitting reporter in the invention.
  • Types of light- emitting reporters include, but are not limited to the wild-type green fluorescent protein (GFP) of Victoria aequoria (Chalfie et al, 1994,
  • reporter gene Another type of reporter gene that may be used are enzymes that require cofactor(s) to emit light, including but not limited to, Renilla luciferase.
  • Other sources of luciferase also are well known in the art, including, but not limited to, the bacterial luciferase (luxAB gene product) of Vibrio harveyi (Karp, 1989, Biochim. Biophys. Acta
  • reporter genes that can be analyzed using colorimetric analysis include, but are not limited to, ⁇ -galactosidase (Nolan et al. 1988, Proc. Natl. Acad. Sci. USA 85:2603- 07), ⁇ -glucuronidase (Roberts et al. 1989, Curr. Genet. 15:177-180), luciferase (Miyamoto et al, 1987, J. Bacteriol. 169:247-253), or ⁇ -lactamase.
  • the reporter gene sequence comprises a nucleotide sequence which encodes a LacZ gene product, ⁇ -
  • the enzyme is very stable and has a broad specificity so as to allow the use of different histochemical, chromogenic or fluorogenic substrates, such as, but not limited to, 5-bromo-4-chloro-3-indoyl- ⁇ -D-galactoside (X-gal), lactose 2,3,5-triphenyl-2H- tetrazolium (lactose-tetrazolium) and fluorescein galactopyranoside (see Nolan et al, 1988, supra).
  • X-gal 5-bromo-4-chloro-3-indoyl- ⁇ -D-galactoside
  • lactose 2,3,5-triphenyl-2H- tetrazolium lactose-tetrazolium
  • fluorescein galactopyranoside see Nolan et al, 1988, supra.
  • the product of the E. coli ⁇ -glucuronidase gene can be used as a reporter gene (Roberts et al. 1989, Curr. Genet. 15:177-180).
  • GUS activity can be detected by various histochemical and fluorogenic substrates, such as X- glucuronide (Xgluc) and 4-methylumbelliferyl glucuronide.
  • reporter gene sequences such as those described above, which provide convenient colorimetric responses
  • other reporter gene sequences such as, for o example, selectable reporter gene sequences
  • the coding sequence for chloramphenicol acetyl transferase (CAT) can be utilized, leading to OSN regulatory region-dependent expression of chloramphenicol resistant cell growth.
  • CAT chloramphenicol acetyl transferase
  • Other selectable reporter gene sequences 5 also can be utilized and include, but are not limited to, gene sequences encoding polypeptides which confer zeocin (Hegedus et al. 1998, Gene 207:241-249) or kanamycin resistance (Friedrich & Soriano, 1991, Genes. Dev. 5:1513-1523).
  • the detectable reporter polynucleotide may be either a polynucleotide that specifically hybridizes with a predefined oligonucleotide probe, or a polynucleotide encoding a detectable protein, including an OSN polypeptide or a fragment or a variant thereof. This type of assay is well known to those skilled in the art (US 5,502,176 and US 5,266,488).
  • OSN promoter driven reporter constructs can be constructed according to standard recombinant DNA techniques (see, e.g., Methods in Enzymology, 1987, volume 154, Academic Press; Sambrook et al. 1989, Molecular Cloning - A Laboratory Manual, 2nd Edition, Cold Spring Harbor Press, New York; and Ausubel et al. Current Protocols in Molecular Biology, Greene Publishing Associates and Wiley Interscience, New York, each 0 of which is incorporated herein by reference in its entirety).
  • reporter gene for example, green fluorescent protein, luciferase, ⁇ -galactosidase or chloramphenicol acetyl transferase
  • the reporter gene for example, green fluorescent protein, luciferase, ⁇ -galactosidase or chloramphenicol acetyl transferase
  • Genomic sequences located upstream of the first exon of the gene may be cloned into any suitable promoter reporter vector.
  • a number of commercially available vectors can be engineered to insert the OSN regulatory region of the invention for expression in mammalian host cells.
  • Non-limiting examples of such vectors are pSEAPBasic, pSEAP-Enhancer, p ⁇ gal-Basic, p ⁇ gal-Enhancer, or pEGFP-1 Promoter Reporter vectors (Clontech, Palo Alto, CA) or pGL2-basic or pGL3-basic promoterless luciferase reporter gene vector (Promega, Madison, WI).
  • Each of these promoter reporter vectors include multiple cloning sites positioned upstream of a reporter gene encoding a readily assayable protein such as secreted alkaline phosphatase, green fluorescent protein, luciferase or ⁇ -galactosidase.
  • the regulatory sequences of the OSN gene are inserted into the cloning sites upstream of the reporter gene in both orientations and introduced into an appropriate host cell.
  • the level of reporter protein is assayed and compared to the level obtained with a vector lacking an insert in the cloning site. The presence of an elevated expression level in the vector containing the insert with respect the control vector indicates the presence of a promoter in the insert.
  • Expression vectors that comprise an OSN regulatory region may further contain a gene encoding a selectable marker.
  • a number of selection systems may be used, including but not limited to, the herpes simplex virus thymidine kinase (Wigler et al, 1977, Cell 11 :223), hypoxanthine-guanine phosphoribosyltransferase (Szybalska & Szybalski, 1962, Proc. Natl. Acad. Sci. USA 48:2026) and adenine phosphoribosyltransferase (Lowy et al, 1980, Cell 22:817) genes, which can be employed in tk " , hgprt " or aprt " cells, respectively.
  • antimetabolite resistance can be used as the basis of selection for dhfr, which confers resistance to methotrexate (Wigler et al, 1980, Proc. Natl. Acad. Sci. USA 77:3567; O'Hare et al, 1981, Proc. Natl. Acad. Sci. USA 78:1527); gpt, which confers resistance to mycophenolic acid (Mulligan & Berg, 1981, Proc. Natl. Acad. Sci. USA 78:2072); neo, which confers resistance to the aminoglycoside G-418 (Colberre-Garapin et al, 1981, J. Mol. Biol.
  • hygro which confers resistance to hygromycin (Santerre et al, 1984, Gene 30:147) genes.
  • Additional selectable genes include trpB, which allows cells to utilize indole in place of tryptophan; hisD, which allows cells to utilize histinol in place of histidine (Hartman & Mulligan, 1988, Proc. Natl. Acad. Sci.
  • ODC ornithine decarboxylase
  • DFMO McConlogue L., 1987, In: Current Communications in Molecular Biology, Cold Spring Harbor Laboratory ed.
  • glutamine synthetase Bebbington et al. , 1992, Biotech 10:169.
  • a fusion construct comprising an OSN regulatory region, or a fragment thereof, can be assayed for transcriptional activity.
  • the transcriptional start point (+1 site) of the osteotropic-specific gene under study has to be determined using primer extension assay and/or RNAase protection assay, following standard methods (Sambrook et ⁇ /.,1989, Molecular Cloning: A Laboratory Manual, Cold Spring Harbor, Cold Spring Harbor Press).
  • the DNA sequence upstream of the +1 site is generally considered as the promoter region responsible for gene regulation. However, o downstream sequences, including sequences within introns, also may be involved in gene regulation.
  • a -3 kb to +3 kb region may be cloned upstream of the reporter gene coding region.
  • Two or more additional reporter gene constructs also may be made which contain 5' and/or 3' truncated versions of the regulatory region to aid in identification of the region responsible 5 for osteotropic-specific expression. The choice of the type of reporter gene is made based on the application.
  • a GFP reporter gene construct is used.
  • the application of green fluorescent protein (GFP) as a reporter is particularly useful in the study of osteotropic-specific gene promoters.
  • GFP green fluorescent protein
  • a major advantage of using GFP as a reporter lies 0 in the fact that GFP can be detected in freshly isolated tumor and tissue cells with calcification potential without the need for substrates.
  • a Lac Z reporter construct is used.
  • the Lac Z gene product, ⁇ -galactosidase is extremely stable and has a broad specificity so as to allow the use of different histochemical, chromogenic or fluorogenic substrates, such 5 as, but not limited to, 5-bromo-4-chloro-3-indoyl- ⁇ -D-galactoside (X-gal), lactose 2,3,5- triphenyl-2H-tetrazolium (lactose-tetrazolium) and fluorescein galactopyranoside (see Nolan et al, 1988, supra).
  • GFP that has been optimized for expression in mammalian cells
  • the promoterless cloning vector pEGFPl 0 (Clontech, Palo Alto, CA) encodes a red shifted variant of the wild-type GFP which has been optimized for brighter fluorescence and higher expression in mammalian cells (Cormack et al. , 1996, Gene 173 :33; Haas et al. , 1996, Curr. Biol. 6: 315).
  • EGFP enhanced GFP
  • filter sets such as fluorescein isothiocyanate (FITC) optics which illuminate at 450-500 5 nm can be used to visualize GFP fluorescence.
  • FITC fluorescein isothiocyanate
  • pEGFPl proved to be useful as a reporter vector for promoter analysis in transgenic mice (Okabe et al, 1997, FEBS Lett. 407: 313).
  • transgenic mice containing transgenes with an OSN regulatory region upstream of a luciferase reporter gene are utilized.
  • Putative promoter fragments can be prepared (usually from a parent phage clone containing 8-10 kb genomic DNA including the promoter region) for cloning using methods known in the art.
  • promoter fragments are cloned into the multiple cloning site of a luciferase reporter vector.
  • restriction endonucleases are used to excise the regulatory region fragments to be inserted into the reporter vector.
  • the feasibility of this method depends on the availability of proper restriction endonuclease sites in the regulatory fragment.
  • the required promoter fragment is amplified by polymerase chain reaction (PCR; Saiki et al, 1988, Science 239:487) using oligonucleotide primers bearing the appropriate sites for restriction endonuclease cleavage.
  • the sequence necessary for restriction cleavage is included at the 5' end of the forward and reverse primers which flank the regulatory fragment to be amplified.
  • the appropriate ends are generated by restriction digestion of the PCR product.
  • the promoter fragments, generated by either method, are then ligated into the multiple cloning site of the reporter vector following standard cloning procedures (Sambrook et ⁇ /.,1989, supra).
  • the DNA sequence of the PCR generated promoter fragments in the constructs be verified prior to generation of transgenic animals.
  • the resulting reporter gene construct will contain the putative promoter fragment located upstream of the reporter gene open reading frame, e.g., GFP or luciferase cDNA.
  • the following protocol is used. Fifty to 100 pg of the reporter gene construct is digested using appropriate restriction endonucleases to release the transgene fragment.
  • the restriction endonuclease cleaved products are resolved in a 1 % (w/v) agarose gel containing 0.5 ug/ml ethidium bromide and TAE buffer (IX: 0.04 M Triacetate, 0.001 M EDTA, pH 8.0) at 5-6 V/cm.
  • the transgene band is located by size using a UV transilluminator, preferably using long- wavelength UV lamp to reduce nicking of DNA, and the gel piece containing the required band carefully excised.
  • the gel slice and 1 ml of 0.5 X TAE buffer is added to a dialysis bag, which has been boiled in 1 mM EDTA, pH 8.0 for 10 minutes (Sambrook et ⁇ /.,1989, supra) and the ends are fastened.
  • the dialysis bag containing the gel piece is submerged in a horizontal gel electrophoresis chamber containing 0.5 X TAE buffer, and electrophoresed at 5-6 V/cm for 45 minutes.
  • the current flow in the electrophoresis chamber is reversed for one minute before stopping the run to release the DNA which may be attached to the wall of the dialysis tube.
  • the TAE buffer containing the electroeluted DNA from the dialysis bag is collected in a fresh eppendorf tube.
  • the gel piece may be observed on the UV transilluminator to ascertain that the electroelution of the DNA is complete.
  • the electroeluted DNA sample is further purified by passing through Elutip D columns.
  • the matrix of the column is prewashed with 1-2 ml of High salt buffer (1.0 M NaCI, 20mM Tris. Cl, 1.0 mM EDTA, pH 7.5), followed by a wash with 5 ml of Low salt buffer (0.2 M NaCI, 20 mM Tris. Cl, 1.0 mM EDTA, pH 7.5).
  • a 5 ml syringe is used to apply solutions to the Elutip D column, avoiding reverse flow.
  • the solution containing the electroeluted DNA is loaded slowly.
  • the column is washed with 2-3 ml of Low salt buffer and the DNA is eluted in 0.4 ml of High salt buffer.
  • the DNA is resuspend in the injection buffer (lOmM TM, 0.1 mM EDTA, pH 7.5 prepared 5 with Milli-Q quality water).
  • the mammalian OSN regulatory region can be used to direct expression of, ter alia, a reporter coding sequence, a homologous gene or a heterologous gene in transgenic animals specifically within tumor and tissue cells with calcification potential.
  • Animals of any species including, but not limited to, mice, rats, rabbits, guinea pigs, pigs, 5 micro-pigs, goats, sheep, and non-human primates, e.g., baboons, monkeys and chimpanzees may be used to generate transgenic animals.
  • transgenic refers to non-human animals expressing OSN gene sequences from a different species (e.g., mice expressing human OSN sequences), as well as animals that have been genetically engineered to over-express endogenous (i.e., same species) OSN sequences or 0 animals that have been genetically engineered to knock-out specific sequences.
  • the present invention provides for transgenic animals that carry a transgene such as a reporter gene, therapeutic and/or toxic coding sequence under the control of the OSN regulatory region, or transcriptionally active fragments thereof, in all their cells, as well as animals that carry the transgene in some, but not all their 5 cells, i.e., mosaic animals.
  • the transgene may be integrated as a single transgene or in concatamers, e.g., head-to-head tandems or head-to-tail tandems.
  • the transgene may also be selectively introduced into and activated in a particular cell type by following, for example, the teaching of Lasko et al. (1992, Proc. Natl. Acad. Sci. USA 89:6232-6236).
  • transgene When it is desired that the transgene be integrated into the chromosomal site of the endogenous corresponding gene, gene targeting is preferred.
  • vectors containing some nucleotide sequences homologous to the endogenous gene are designed for the purpose of integrating, via homologous recombination with chromosomal sequences, into and disrupting the function of the nucleotide sequence of the endogenous gene.
  • Any technique known in the art may be used to introduce a transgene under the control of the OSN regulatory region into animals to produce the founder lines of transgenic animals.
  • Such techniques include, but are not limited to, pronuclear microinjection (Hoppe & Wagner, 1989, U.S. Patent No. 4,873,191); nuclear transfer into enucleated oocytes of nuclei from cultured embryonic, fetal or adult cells induced to quiescence (Campbell et al, 1996, Nature 380:64-66; Wilmut et al, Nature 385:810-813); retrovirus gene transfer into germ lines (Van der Putten et al. , 1985, Proc. Natl. Acad.
  • the transgene containing the regulatory region, the reporter gene and the polyadenylation signals, is excised from the reporter gene construct.
  • the transgene may be gel purified by methods known in the art, for example, by the electroelution method. Following electroelution of gel fragments, any traces of impurities are further removed by passing through Elutip D column (Schleicher & Schuell, Dassel, Germany).
  • the purified transgene fragment is microinjected into the male pronuclei of fertilized eggs obtained from B6 CBA females by standard methods (Hogan, 1986, Manipulating the Mouse Embryo, A Laboratory Manual. Cold Spring Harbor Laboratory Press, Cold Spring Harbor, NY). Mice are analyzed transiently at several embryonic stages or by establishing founder lines that allow more detailed analysis of transgene expression throughout development and in adult animals. Transgene presence is analyzed by PCR using genomic DNA purified from placentas (transients) or tail clips (founders) according to the method of Vemet et al, Methods Enzymol 1993;225:434-451.
  • the PCR reaction is carried out in a volume of 100 ⁇ l containing 1 ⁇ g of genomic DNA, in IX reaction buffer supplemented with 0.2 mM dNTPs, 2 mM MgCl 2 , 600 ⁇ M each of primer, and 2.5 units of Taq polymerase (Promega, Madison, WI).
  • Each of the 30 PCR cycles consists of denaturation at 94°C for 1 min, annealing at 54°C for 1 min, and extension at 72 °C for 1 min.
  • the founder mice are then mated with C57B1 partners to generate transgenic F, lines of mice.
  • Compounds that interfere with the abnormal function and/or growth of tumor and tissue cells with calcification potential can provide therapies targeting defects in osteotropic-related disorders including, but not limited to, localized or disseminated osteosarcoma, lung, colon, melanoma, thyroid, brain, multiple myeloma, breast and prostate o cancers, and benign conditions, such as benign prostatic hyperplasia (BPH) or arterial sclerotic conditions where calcification occurs.
  • BPH benign prostatic hyperplasia
  • Compounds that stimulate or inhibit promoter activity also may be used to ameliorate symptoms of osteotropic-related disorders.
  • transgenic mice can be used as systems for the screening of agents that modulate OSN regulatory region activity.
  • Such transgenic mice provide an experimental model in vivo (or can be used as a source of primary cells or cell lines for use in vitro ) which can be used to develop new methods of 0 treating osteotropic-related disorders by targeting therapeutic and/or toxic agents to cause arrest in the progression of such disorders.
  • the present invention encompasses screening assays designed to identify compounds that modulate activity of the OSN regulatory region.
  • the present invention encompasses in vitro and cell-based assays, as well as in vivo assays in transgenic animals. 5
  • compounds to be tested may include, but are not limited to, oligonucleotides, peptides, proteins, small organic or inorganic compounds, antibodies, etc.
  • Examples of compounds may include, but are not limited to, peptides, such as, for example, soluble peptides, including, but not limited to, Ig-tailed fusion peptides, and members of random peptide libraries; (see, e.g., Lam, et al, 1991, Nature 354:82-84; 0 Houghten, et al, 1991, Nature 354:84-86), and combinatorial chemistry-derived molecular library made of D- and/or L- configuration amino acids, phosphopeptides (including, but not limited to members of random or partially degenerate, directed phosphopeptide libraries; see, e.g., Songyang, et al, 1993, Cell 72:767-778), antibodies (including, but not limited to, polyclonal, monoclonal, humanized, anti-idiotypic, chimeric or single chain 5 antibodies, and FAb, F(ab') 2 and FAb expression library fragments, and epitope-binding fragments thereof), and
  • Such compounds may further comprise compounds, in particular drugs or members of classes or families of drugs, known to ameliorate the symptoms of an osteotropic-related disorder.
  • Such compounds include, but are not limited to, families of anti depressants such as lithium salts, carbamazepine, valproic acid, lysergic acid diethylamide (LSD), p- chlorophenylalanine, >-propyldopacetamide dithiocarbamate derivatives e.g., FLA 63; anti- anxiety drugs, e.g., diazepam; monoamine oxidase (MAO) inhibitors, e.g., iproniazid, clorgyline, phenelzine and isocarboxazid; biogenic amine uptake blockers, e.g., tricyclic antidepressants such as desipramine, imipramine and ami tripty line; serotonin reuptake inhibitors e.g., fluoxetine; antipsychotic drugs such as phenothiazine derivatives (e.g., chlorpromazine (thorazine) and trifluopromaz
  • genetically engineered cells, cell lines or primary cultures of germ and/or somatic cells containing a mammalian OSN regulatory region operatively linked to a heterologous gene are used to develop assay systems to screen for compounds which can inhibit sequence-specific DNA-protein interactions.
  • Such methods comprise contacting a compound to a cell that expresses a gene under the control of an OSN regulatory region, or a transcriptionally active fragment thereof, measuring the level of the gene expression or gene product activity and comparing this level to the level of gene expression or gene product activity produced by the cell in the absence of the compound, such that if the level obtained in the presence of the compound differs from that obtained in its absence, a compound capable of modulating the expression of the mammalian OSN regulatory region has been identified.
  • Alterations in gene expression levels may be by any number of methods known to those of skill in the art e.g., by assaying for reporter gene activity, assaying cell lysates for mRNA transcripts, e.g. by Northern analysis or using other methods known in the art for assaying for gene products expressed by the cell.
  • microdissection and transillumination can be used. These techniques offer a rapid assay for monitoring effects of putative drugs on osteotropic cells in transgenic animals containing an OSN regulatory region-driven reporter gene.
  • a test agent is delivered to the transgenic animal by any of a variety of methods.
  • Methods of introducing a test agent may include oral, intradermal, intramuscular, intraperitoneal, intravenous, subcutaneous, intranasal and via scarification (scratching through the top layers of skin, e.g., using a bifurcated needle) or any other standard routes of drug delivery.
  • the effect of such test compounds on the osteotropic cells can be analyzed by the microdissection and transillumination of the osteoblastic cells. If the level of reporter gene expression observed or measured in the presence of the compound differs from that obtained in its absence, a compound capable of modulating the expression of the mammalian OSN regulatory region has been identified.
  • compounds that may be used in screens for modulators of osteotropic-related disorders include peptides, small molecules, both naturally occurring and/or synthetic (e.g., libraries of small molecules or peptides), cell-bound or soluble molecules, organic, non-protein molecules and recombinant molecules that may have OSN regulatory region binding capacity and, therefore, may be candidates for pharmaceutical agents.
  • the proteins and compounds include endogenous cellular components which interact with OSN regulatory region sequences in vivo.
  • Cell lysates or tissue homogenates may be screened for proteins or other compounds which bind to the OSN regulatory region, or fragment thereof.
  • Such endogenous components may provide new targets for pharmaceutical and therapeutic interventions.
  • libraries can be screened.
  • Many libraries are known in the art that can be used, e.g. , peptide libraries, chemically synthesized libraries, recombinant (e.g., phage display libraries), and in vitro translation-based libraries.
  • peptide libraries may be used to screen for agonists or antagonists of OSN-linked reporter expression.
  • Diversity libraries such as random or combinatorial peptide or non-peptide libraries can be screened for molecules that specifically modulate OSN regulatory region activity. Random peptide libraries consisting of all possible combinations of amino acids attached to a solid phase support may be used to identify peptides that are able to activate or inhibit OSN regulatory region activities (Lam, K.S. et al, 1991, Nature 354: 82-84).
  • the screening of peptide libraries may have therapeutic value in the discovery of pharmaceutical agents that stimulate or inhibit the expression of OSN regulatory regions. Examples of chemically synthesized libraries are described in Fodor et al,
  • a benzodiazepine library (see e.g., Bunin et al, 1994, Proc. Natl. Acad. Sci. USA 91 :4708-4712) can be adapted for use.
  • Peptoid libraries (Simon et al, 1992, Proc. Natl. Acad. Sci. USA 89:9367-9371) also can be used.
  • the OSN regulatory region-reporter vector is used to generate transgenic mice from which primary cultures of OSN regulatory region-reporter vector germ cells are established. About 10,000 cells per well are plated in 96-well plates in total volume of 100 ⁇ l, using - > medium appropriate for the cell line. Candidate inhibitors of the OSN regulatory region are added to the cells. The effect of the inhibitors of the OSN regulatory region can be determined by measuring the response of the reporter gene driven by the OSN regulatory region. This assay could easily be set up in a high-throughput screening mode for evaluation of compound libraries in a 96-well format that reduce (or increase) reporter gene
  • tumor or tissue cells with calcification potential derived from transgenic mice can be transplanted into mice with a normal or other desired phenotype (Brinster et al, 1994, Proc. Natl. Acad. Sci. USA 91 : 11298-302; Ogawa et al, 1997, Int. J. Dev. Biol. 41 :1 11-12).
  • mice can then be used to test the effect of compounds and other various factors on osteotropic-related disorders.
  • such mice can be used to assay factors or conditions that can be difficult to test using other methods, such as dietary effects, internal pH, temperature, etc.
  • a compound may then be tested in an animal-based assay to determine if the compound exhibits the ability to act as a drug to ameliorate and/or prevent symptoms of an osteotropic-related disorder, including, but not limited to, localized or disseminated osteosarcoma, lung, colon, melanoma, thyroid, brain, multiple myeloma, breast and prostate cancers, and benign conditions, such as benign prostatic hyperplasia (BPH) or arterial 5 sclerotic conditions where calcification occurs.
  • an osteotropic-related disorder including, but not limited to, localized or disseminated osteosarcoma, lung, colon, melanoma, thyroid, brain, multiple myeloma, breast and prostate cancers, and benign conditions, such as benign prostatic hyperplasia (BPH) or arterial 5 sclerotic conditions where calcification occurs.
  • the assays of the present invention may be first optimized on a small scale (i.e., in test tubes), and then scaled up for high-throughput assays.
  • the screening assays of the present invention may be performed in vitro, i.e., in test tubes, using purified components or cell lysates.
  • the screening assays of the present invention may also be 0 carried out in intact cells in culture and in animal models.
  • test compounds which are shown to modulate the activity of the OSN regulatory region in vitro, as described herein will further be assayed in vivo in cultured cells and animal models to determine if the test compound has the similar effects in vivo and to determine the effects of the test compound on osteotropic-related disorders. 5
  • OSN regulatory regions or transcriptionally active fragments thereof, can be used to treat and/or prevent diseases, conditions or disorders that can be ameliorated by 0 modifying the level or the expression of OSN, or a heterologous gene linked to an OSN regulatory region, in an osteotropic-specific manner. Described herein are methods for such therapeutic treatments.
  • the OSN regulatory region may be used to achieve tissue specific expression in gene therapy protocols. In cases where such cells are tumor cells, the induction of a 5 cytotoxic product by the OSN regulatory region may be used in the form of cancer gene therapy specifically targeted to tumor cells with calcification potential which contain trans- acting factors required for OSN expression. In this way, the OSN regulatory region may serve as a delivery route for a gene therapy approach to cancers involving tumor cells with calcification potential. Additionally, antisense, antigene or aptameric oligonucleotides may be delivered to cells using the presently described expression constructs. Ribozymes or single-stranded R A also can be expressed in a cell to inhibit the expression of a target gene of interest. The target genes for these antisense or ribozyme molecules should be those encoding gene products that are essential for cell maintenance.
  • the OSN regulatory region, and transcriptionally active fragments thereof, of the present invention may be used for a wide variety of purposes, e.g., to down regulate OSN gene expression, or, alternatively, to achieve osteotropic-specific expression of heterologous coding sequences.
  • the endogenous OSN regulatory region may be targeted to specifically down-regulate expression of the OSN gene.
  • oligonucleotides complementary to the regulatory region may be designed and delivered to the cells. Such oligonucleotides may anneal to the regulatory sequence and prevent transcription activation.
  • the regulatory sequence, or portions thereof may be delivered to cells in saturating concentrations to compete for transcription factor binding.
  • a gene therapy method for ameliorating osteotropic- related disorders comprises introducing an OSN regulatory region sequence operatively associated with a drug or toxin gene into the osteotropic cells.
  • the invention provides a gene therapy method for treatment of cancer or other proliferative disorders.
  • the OSN regulatory region is used to direct the expression of one or more proteins specifically in osteotropic tumor cells of a patient.
  • proteins may be, for example, tumor suppressor genes, thymidine kinase (used in combination with acyclovir), toxins or proteins involved in cell killing, such as proteins involved in the apoptosis pathway.
  • the invention provides for a therapeutic agent comprising an OSN promoter which is useful for toxic gene therapy.
  • This method includes a eukaryotic delivery vector and a toxic gene.
  • the vector is adenovirus (Ad) and the gene is thymidine kinase (TK).
  • Ad-OSN-TK adenovirus
  • TK thymidine kinase
  • the DNA encoding the translational or transcriptional products of interest may be engineered recombinantly into a variety of vector systems that provide for replication of the DNA in large scale for the preparation of the vectors of the invention. These vectors can be designed to contain the necessary elements for directing the transcription and/or translation of the DNA sequence taken up by the osteotropic cells.
  • Vectors that may be used include, but are not limited to, those derived from recombinant bacteriophage DNA, plasmid DNA or cosmid DNA.
  • plasmid vectors such as pBR322, pUC 19/18, pUC 118, 119 and the M13 mp series of vectors may be used.
  • Bacteriophage vectors may include ⁇ gtlO, ⁇ gtl 1, ⁇ gtl8-23, ⁇ ZAP/R and the EMBL series of bacteriophage vectors.
  • Cosmid vectors that may be utilized include, but are not limited to, pJB8, pCV 103, pCV 107, pCV 108, pTM, pMCS, pNNL, pHSG274, COS202, COS203, pWE15, pWE16 and the charomid 9 series of vectors.
  • Vectors that allow for the in vitro transcription of RNA such as SP6 vectors, also may be used to produce large quantities of RNA that may be incorporated into viral vectors.
  • recombinant replication competent or incompetent viral vectors including, but not limited to, those derived from viruses such as herpes virus, retroviruses, vaccinia viruses, adenoviruses, adeno-associated viruses or bovine papilloma virus may be engineered. While integrating vectors may be used, non-integrating systems, which do not transmit the gene product to daughter cells for many generations, are preferred for non-disease related repair and regeneration. In this way, the gene product is expressed during the repair process, and as the gene is diluted out in progeny generations, the amount of expressed gene product is diminished.
  • tissue specific promoters to drive therapeutic gene expression would decrease further a toxic effect of the therapeutic gene on neighboring normal cells when virus-mediated gene delivery results in the infection of the normal cells. This would be important especially in diseases where systemic administration could be utilized to deliver a therapeutic vector throughout the body, while maintaining transgene expression to a limited and specific number of cell types. Moreover, since many bone growth factors, such as TGF- ⁇ , have pleiotropic effects, numerous, harmful side effects likely would be exhibited if the growth factor genes are expressed in all cells.
  • the promoter elements may be constitutive or inducible promoters and can be used under the appropriate conditions to direct high level or regulated expression of the gene of interest. Expression of genes under the control of constitutive promoters does not require the presence of a specific substrate to induce gene expression and will occur under all conditions of cell growth. In contrast, expression of genes controlled by inducible promoters is responsive to the presence or absence of an inducing agent. For example, if a cell is stably transfected with a therapeutic, inducible transgene, its expression could be controlled over the life-time of the individual. In fact, the OSN promoter, itself, is induced by glucocorticoids and ascorbic acid.
  • Specific initiation signals also are required for sufficient translation of inserted protein coding sequences. These signals include the ATG initiation codon and adjacent sequences. In cases where the entire coding sequence, including the initiation codon and adjacent sequences, are inserted into the appropriate expression vectors, no additional translational control signals may be needed. However, in cases where only a portion of the coding sequence is inserted, exogenous translational control signals, including the ATG initiation codon, must be provided. Furthermore, the initiation codon must be in phase with the reading frame of the protein coding sequences to ensure translation of the entire insert. These exogenous translational control signals and initiation codons can be of a variety of origins, both natural and synthetic.
  • a method for treating osteotropic tumors comprising delivering a therapeutic agent to the tumor.
  • the therapeutic agent comprises a recombinant adenovirus vector (Ad) containing an OSN promoter driven toxic thymidine kinase (Tk).
  • Ad adenovirus vector
  • Tk OSN promoter driven toxic thymidine kinase
  • An additional aspect of the present invention provides a method of regulating expression of Tk with the addition of a suitable prodrug including, but not limited to, acyclovir (AcV).
  • the therapeutic agent containing the OSN promoter-driven toxic gene therapy in the presence of a suitable prodrug, can be administered to osteosarcoma tumors, and prostate cancer tumors, and their metastases, and many other osteotropic tumors, including, but not limited to, colon, brain, lung, breast, multiple, myeloma, thyroid, and melanoma.
  • the present therapeutic invention comprising Ad-OSN-TK, or other vectors containing OSN promoter-driven activity, is provided to target cancers that are osteotropic, thus possessing bone-like and bone homing capabilities and eliciting an osteoblastic or osteolytic phenotype when in association with bone tissue.
  • the OSN regulatory region may code for a variety of factors that promote bone repair including extracellular, cell surface and intracellular RNAs and proteins. These therapeutic constructs would be useful for, inter alia, aging and certain degenerative conditions. Examples of extracellular proteins include growth factors, cytokines, therapeutic proteins, hormones and peptide fragments of hormones, inhibitors of cytokines, peptide growth and differentiation factors, interleukins, chemokines, interferons, colony stimulating factors and angiogenic factors.
  • TGF- ⁇ proteins include, but are not limited to, the superfamily of TGF- ⁇ molecules, including the five TGF- ⁇ isoforms and bone morphogenetic proteins (BMP), latent TGF- ⁇ binding proteins, LTBP; keratinocyte growth factor (KGF); hepatocyte growth factor (HGF); platelet derived growth factor (PDGF); insulin-like growth factor (IGF); the basic fibroblast growth factors (FGF-1, FGF-2, etc.), vascular endothelial growth factor (VEGF); Factor VIII and Factor IX; erythropoietin (EPO); tissue plasminogen activator (TPA) and activins and inhibins.
  • BMP bone morphogenetic proteins
  • KGF keratinocyte growth factor
  • HGF hepatocyte growth factor
  • PDGF platelet derived growth factor
  • IGF insulin-like growth factor
  • FGF-1, FGF-2, etc. the basic fibroblast growth factors
  • VEGF vascular endotheli
  • Hormones which may be used in the practice of the invention include, for example, growth hormone (GH) and parathyroid hormone (PTH).
  • extracellular proteins also include the extracellular matrix proteins such as collagen, laminin and fibronectin.
  • cell surface proteins include the family of cell adhesion molecules (e.g., the mteg ⁇ ns, selectins, Ig family members such as N-CAM and LI and cadherins); cytokine signaling receptors such as the type I and type II TGF- ⁇ receptors and the FGF receptor and non-signaling co-receptors such as betaglycan and syndecan.
  • RNAs and proteins examples include the family of signal transducing kinases, cytoskeletal proteins such as talin and vinculin, cytokine binding proteins such as the family of latent TGF- ⁇ binding proteins and nuclear trans acting proteins such as transcription factors and enhancing factors.
  • the method comprises introducing an OSN regulatory region sequence operatively associated with a nucleic acid encoding a therapeutic compound that promotes bone synthesis and/or repair.
  • the tissue specificity of the OSN promoter will allow for specific expression of the therapeutic compounds in osteotropic cells of interest.
  • the use of the OSN promoter to drive therapeutic gene expression would decrease further a toxic effect of the therapeutic gene on neighboring normal cells when virus-mediated gene delivery results in the infection of the normal cells. This would be important especially in diseases where systemic administration could be utilized to deliver a therapeutic vector throughout the body, while maintaining transgene expression to a limited and specific number of cell types.
  • many therapeutic growth factors, such as TGF- ⁇ have pleiotropic effects, numerous, harmful side effects likely would be exhibited if the growth factor genes are expressed in all cells.
  • the OSN regulatory region may code for a variety of genes with immune modulatory functions, e.g. for cytokines such as interleukins 1 to 15 inclusive, especially for example IL2, IL12, gamma-interferon, tumour necrosis factor, GMCSF, and/or other genes, e.g. those mentioned in specifications WO 88/00971 (CSIRO, Australian National University: Ramshaw et al) and WO 94/16716 (Virogenetics Corp; Paoletti et al).
  • cytokines such as interleukins 1 to 15 inclusive, especially for example IL2, IL12, gamma-interferon, tumour necrosis factor, GMCSF, and/or other genes, e.g. those mentioned in specifications WO 88/00971 (CSIRO, Australian National University: Ramshaw et al) and WO 94/16716 (Virogenetics Corp; Paoletti et al).
  • genes for interferons alpha, beta or gamma genes for interferons alpha, beta or gamma; tumour necrosis factor; granulocyte-macrophage colony-stimulating factor (GM-CSF), granulocyte colony-stimulating factor (G-CSF), macrophage colony-stimulating factor (N-CSF), chemokines such as neutrophil activating protein NAP, macrophage chemoattractant and activating factor MCAF, RANTES, macrophage inflammatory peptides MIP- 1 a and MlP-lb, complement components and their receptors, accessory molecules such as 87.1, 87.2, ICAM-1.2 or 3 or cytokine receptors.
  • nucleotide sequences encoding more than one immunomodulating protein are inserted, they may comprise more than one cytokine or may represent a combination of cytokine and accessory molecule(s).
  • the types of conditions, disorders, or diseases involving tumor and tissue cells with calcification potential which may be prevented, delayed, or rescued by modulating osteotropic-specifc gene expression by using an OSN regulatory region in conjunction with well-known antisense, gene "knock-out,” ribozyme and/or triple helix methods, are described.
  • Such molecules may be designed to modulate, reduce or inhibit either unimpaired, or if appropriate, mutant osteotropic gene activity. Techniques for the production and use of such molecules are well known to those of skill in the art.
  • Antisense RNA and DNA molecules act to directly block the translation of mRNA by hybridizing to targeted mRNA and preventing protein translation.
  • Antisense approaches involve the design of oligonucleotides which are complementary to an mRNA sequence.
  • the antisense oligonucleotides will bind to the complementary mRNA sequence transcripts and prevent translation. Absolute complementarity, although preferred, is not required.
  • a sequence "complementary" to a portion of an RNA means a sequence having sufficient complementarity to be able to hybridize with the RNA, forming a stable duplex; in the case of double-stranded antisense nucleic acids, a single strand of the duplex DNA may thus be tested, or triplex formation may be assayed.
  • the ability to hybridize will depend on both the degree of complementarity and the length of the antisense nucleic acid.
  • oligonucleotides complementary to non-coding regions of the sequence of interest could be used in an antisense approach to inhibit translation of endogenous mRNA.
  • Antisense nucleic acids should be at least six nucleotides in length, and are preferably oligonucleotides ranging from 6 to about 50 nucleotides in length. In specific aspects, the oligonucleotide is at least 10 nucleotides, at least 17 nucleotides, at least 25 nucleotides or at least 50 nucleotides.
  • in vitro studies are first performed to quantitate the ability of the antisense oligonucleotide to inhibit sequence expression. It is preferred that these studies utilize controls that distinguish between antisense gene inhibition and nonspecific biological effects of oligonucleotides. Additionally, it is envisioned that results obtained using the antisense oligonucleotide are compared with those obtained using a control oligonucleotide. It is preferred that the control oligonucleotide is of approximately the same length as the test oligonucleotide and that the nucleic acid of the oligonucleotide differs from the antisense sequence no more than is necessary to prevent specific hybridization to the target sequence.
  • the oligonucleotides can be DNA or RNA or chimeric mixtures or derivatives or modified versions thereof, single-stranded or double-stranded.
  • the oligonucleotide can be modified at the base moiety, sugar moiety, or phosphate backbone, for example, to improve stability of the molecule, hybridization, etc.
  • the oligonucleotide may include other appended groups such as peptides (e.g., for targeting host cell receptors in vivo), or agents facilitating transport across the cell membrane (see, e.g., Letsinger, et al, 1989, Proc. Natl Acad. Sci. U.S.A.
  • the oligonucleotide may be conjugated to another molecule, e.g., a peptide, hybridization triggered cross-linking agent, transport agent, hybridization-triggered cleavage agent, etc.
  • the antisense oligonucleotide may comprise at least one modified base moiety which is selected from the group including but not limited to 5-fluorouracil, 5-bromouracil, 5-chlorouracil, 5-iodouracil, hypoxanthine, xanthine, 4-acetylcytosine, 5-(carboxyhydroxylmethyl) uracil, 5-carboxymethylaminomethyl-2-thiouridine, 5-carboxymethylaminomethyluracil, dihydrouracil, beta-D-galactosylqueosine, inosine, N6-isopentenyladenine, 1 -methylguanine, 1 -methylinosine, 2,2-dimethylguanine, 2-methyladenine, 2-methylguanine, 3-methylcytosine, 5-methylcytosine, N6-adenine, 7-methylguanine, 5-methylaminomethyluracil, 5-methoxyaminomethyl-2-thiouracil, beta- D-mannos
  • the antisense oligonucleotide may also comprise at least one modified sugar moiety selected from the group including but not limited to arabinose, 2-fluoroarabinose, xylulose, and hexose.
  • the antisense oligonucleotide comprises at least one modified phosphate backbone selected from the group consisting of a phosphorothioate, a phosphorodithioate, a phosphoramidothioate, a phosphoramidate, a phosphordiamidate, a methylphosphonate, an alkyl phosphotriester, and a formacetal or analog thereof.
  • the antisense oligonucleotide is an ⁇ -anomeric oligonucleotide.
  • An ⁇ -anomeric oligonucleotide forms specific double-stranded hybrids with complementary RNA in which, contrary to the usual ⁇ -units, the strands run parallel to each other (Gautier, et al, 1987, Nucl Acids Res. 15:6625-6641).
  • the oligonucleotide is a 2'-0-methylribonucleotide (Inoue, et al, 1987, Nucl. Acids Res. 15:6131-6148), or a chimeric RNA-DNA analogue (Inoue, et al, 1987, FEBS Lett. 215:327-330).
  • Oligonucleotides of the invention may be synthesized by standard methods known in the art, e.g., by use of an automated DNA synthesizer (such as are commercially available from Biosearch, Applied Biosystems, etc.).
  • an automated DNA synthesizer such as are commercially available from Biosearch, Applied Biosystems, etc.
  • phosphorothioate oligonucleotides may be synthesized by the method of Stein, et al. (1988, Nucl. Acids Res. 16:3209)
  • methylphosphonate oligonucleotides can be prepared by use of controlled pore glass polymer supports (Sarin, et al, 1988, Proc. Natl. Acad. Sci. U.S.A. 85:7448-7451), etc. While antisense nucleotides complementary to the osteotropic specific coding region sequence could be used, those complementary to the transcribed, untranslated region are most preferred.
  • Antisense molecules should be delivered to cells that express the osteotropic sequence in vivo.
  • a number of methods have been developed for delivering antisense DNA or RNA to cells; e.g., antisense molecules can be injected directly into the tissue site, or modified antisense molecules, designed to target the desired cells (e.g., antisense linked to peptides or antibodies which specifically bind receptors or antigens expressed on the target cell surface) can be administered systemically.
  • a preferred approach to achieve intracellular concentrations of the antisense sufficient to suppress translation of endogenous mRNAs utilizes a recombinant DNA construct in which the antisense oligonucleotide is placed under the control of a strong pol III or pol II promoter.
  • the use of such a construct to transfect target cells in the patient will result in the transcription of sufficient amounts of single stranded RNAs which will form 5 complementary base pairs with the endogenous sequence transcripts and thereby prevent translation of the mRNA.
  • a vector can be introduced e.g., such that it is taken up by a cell and directs the transcription of an antisense RNA.
  • Such a vector can remain episomal or become chromosomally integrated, as long as it can be transcribed to produce the desired antisense RNA.
  • Such vectors can be constructed by recombinant DNA
  • Vectors can be plasmid, viral, or others known in the art, used for replication and expression in mammalian cells.
  • Expression of the sequence encoding the antisense RNA can be by any promoter known in the art to act in mammalian, preferably human cells.
  • Such promoters can be inducible or constitutive.
  • Such promoters include but are not limited to: the SV40 early promoter region (Bernoist and Chambon,
  • viral vectors can be used that selectively infect the desired tissue, in which case administration may be accomplished by another route (e.g. , systemically).
  • Ribozyme molecules designed to catalytically cleave target gene mRNA transcripts can also be used to prevent translation of target gene mRNA and, therefore,
  • Ribozymes are enzymatic RNA molecules capable of catalyzing the specific cleavage of RNA.
  • the mechanism of ribozyme action involves sequence specific hybridization of the ribozyme molecule to complementary target RNA, followed by an endonucleolytic cleavage event.
  • the composition of ribozyme molecules must include one or more sequences complementary to the target gene mRNA, and must include the well known catalytic sequence responsible for mRNA cleavage. For this sequence, see, e.g., U.S. Patent No. 5,093,246, which is incorporated herein by reference in its entirety.
  • ribozymes that cleave mRNA at site specific recognition sequences can be used to destroy target gene mRNAs
  • the use of hammerhead ribozymes is preferred.
  • Hammerhead ribozymes cleave mRNAs at locations dictated by flanking regions which form complementary base pairs with the target mRNA. The sole requirement is that the target mRNA have the following sequence of two bases: 5'-UG-3'.
  • the ribozyme is engineered so that the cleavage recognition site is located near the 5' end of the target gene mRNA, i.e., to increase efficiency and minimize the intracellular accumulation of non-functional mRNA transcripts.
  • the ribozymes of the present invention also include RNA endoribonucleases (hereinafter "Cech-type ribozymes”) such as the one which occurs naturally in Tetrahymena thermophi/a (known as the IVS, or L-19 IVS RNA) and which has been extensively described by Thomas Cech and collaborators (Zaug, et al, 1984, Science, 224:574-578; Zaug and Cech, 1986, Science, 231 :470-475; Zaug, et al, 1986, Nature, 324:429-433; published International patent application No. WO 88/04300 by University Patents Inc.; Been and Cech, 1986, Cell, Al. -201 -2X6).
  • Cech-type ribozymes such as the one which occurs naturally in Tetrahymena thermophi/a (known as the IVS, or L-19 IVS RNA) and which has been extensively described by Thomas Cech and collaborators (Zaug, et al, 1984, Science,
  • the Cech-type ribozymes have an eight base pair active site that hybridizes to a target RNA sequence whereafter cleavage of the target RNA takes place.
  • the invention encompasses those Cech-type ribozymes that target eight base- pair active site sequences that are present in the target gene.
  • the ribozymes can be composed of modified oligonucleotides (e.g., for improved stability, targeting, etc.) and should be delivered to cells that express the target gene in vivo.
  • a preferred method of delivery involves using a DNA construct "encoding" the ribozyme under the control of a strong constitutive pol III or pol II promoter, so that transfected cells will produce sufficient quantities of the ribozyme to destroy endogenous target gene messages and inhibit translation. Because ribozymes, unlike antisense molecules, are catalytic, a lower intracellular concentration is required for efficiency.
  • Endogenous target gene expression can also be reduced by inactivating or "knocking out” the target gene or its promoter using targeted homologous recombination (e.g., see Smithies, et al, 1985, Nature 317:230-234; Thomas and Capecchi, 1987, Cell 51 :503-512; Thompson, et al, 1989, Cell 5:313-321 ; each of which is incorporated by reference herein in its entirety).
  • targeted homologous recombination e.g., see Smithies, et al, 1985, Nature 317:230-234; Thomas and Capecchi, 1987, Cell 51 :503-512; Thompson, et al, 1989, Cell 5:313-321 ; each of which is incorporated by reference herein in its entirety).
  • a mutant, non-functional target gene flanked by DNA homologous to the endogenous target gene (either the coding regions or regulatory regions of the target gene) can be used, with or without a selectable marker and/or a negative selectable marker, to transfect cells which express the target gene in vivo. Insertion of the DNA construct, via targeted homologous recombination, results in inactivation of the target gene.
  • ES embryonic stem
  • Such approaches are particularly suited in the agricultural field where modifications to ES (embryonic stem) cells can be used to generate animal offspring with an inactive target gene (e.g. , see Thomas and Capecchi, 1987 and Thompson, 1989, supra).
  • this approach can be adapted for use in humans provided the recombinant DNA constructs are directly administered or targeted to the required site in vivo using appropriate viral vectors.
  • endogenous target gene expression can be reduced by targeting deoxyribonucleotide sequences complementary to the regulatory region of the target gene (i.e., the target gene promoter and/or enhancers) to form triple helical structures which prevent transcription of the target gene in target cells in the body.
  • deoxyribonucleotide sequences complementary to the regulatory region of the target gene i.e., the target gene promoter and/or enhancers
  • triple helical structures which prevent transcription of the target gene in target cells in the body.
  • Nucleic acid molecules to be used in triple helix formation for the inhibition of transcription should be single stranded and composed of deoxynucleotides.
  • the base composition of these oligonucleotides must be designed to promote triple helix formation via Hoogsteen base pairing rules, which generally require sizable stretches of either purines or pyrimidines to be present on one strand of a duplex.
  • Nucleic acids may be pyrimidine- based, which will result in TAT and CGC + triplets across the three associated strands of the resulting triple helix.
  • the pyrimidine-rich molecules provide base complementarity to a purine-rich region of a single strand of the duplex in a parallel orientation to that strand.
  • nucleic acid molecules may be chosen which are purine-rich, for example, contain a stretch of G residues.
  • Switchback molecules will form a triple helix with a DNA duplex that is rich in GC pairs, in which the majority of the purine residues are located on a single strand of the targeted duplex, resulting in GGC triplets across the three strands in the triplex.
  • the potential sequences that can be targeted for triple helix formation may be increased by creating a so-called "switchback" nucleic acid molecule.
  • Switchback molecules are synthesized in an alternating 5'-3', 3'-5' manner, such that they base pair with first one strand of a duplex and then the other, eliminating the necessity for a sizable stretch of either purines or pyrimidines to be present on one strand of a duplex.
  • the technique may so efficiently reduce or inhibit the transcription (triple helix) and/or translation (antisense, ribozyme) of mRNA produced by normal target gene alleles which the possibility may arise wherein the concentration of normal target gene product present may be lower than is necessary for a normal phenotype.
  • nucleic acid molecules which encode and express target gene polypeptides exhibiting normal target gene activity may be introduced into cells via gene therapy methods such as those described, below, in Section 5.4.2 which do not contain sequences susceptible to whatever antisense, ribozyme, or triple helix treatments are being utilized.
  • the target gene encodes an extracellular protein
  • Anti-sense RNA and DNA, ribozyme, and triple helix molecules of the invention may be prepared by any method known in the art for the synthesis of DNA and RNA molecules, as discussed above.
  • RNA molecules may be generated by in vitro and in vivo transcription of DNA sequences encoding the antisense RNA molecule.
  • DNA sequences may be incorporated into a wide variety of vectors that incorporate suitable RNA polymerase promoters such as the T7 or SP6 polymerase promoters.
  • antisense cDNA constructs that synthesize antisense RNA constitutively or inducibly, depending on the promoter used, can be introduced stably into cell lines.
  • nucleic acid sequences of the invention can be utilized for transferring recombinant nucleic acid sequences to cells and expressing said sequences in recipient cells.
  • Such techniques can be used, for example, in marking cells or for the treatment of a disorder involving tumor or tissue cells with calcification potential.
  • Such treatment can be in the form of gene replacement therapy.
  • one or more copies of a normal gene or a portion of the gene that directs the production of a gene product exhibiting normal gene function may be inserted into the appropriate cells within a patient, using vectors that include, but are not limited to adenovirus, adeno-associated virus, herpes simplex virus and retrovirus vectors, in addition to other particles that introduce DNA into cells, such as liposomes.
  • nucleic acid is directly administered in vivo into a target cell or a transgenic mouse that expresses an OSN regulatory region operably linked to a heterologous coding sequencee.
  • This can be o accomplished by any method known in the art, e.g. , by constructing it as part of an appropriate nucleic acid expression vector and administering it so that it becomes intracellular, e.g., by infection using a defective or attenuated retroviral or other viral vector (see U.S. Patent No.
  • microparticle bombardment e.g., a gene gun; Biolistic, Dupont
  • lipids or cell-surface 5 receptors or transfecting agents by encapsulation in liposomes, microparticles, or microcapsules, by administering it in linkage to a peptide which is known to enter the nucleus or by administering it in linkage to a ligand subject to receptor-mediated endocytosis (see e.g., Wu and Wu, 1987, J. Biol. Chem. 262:4429-4432), which can be used to target cell types specifically expressing the receptors.
  • a nucleic 0 acid-ligand complex can be formed in which the ligand comprises a fusogenic viral peptide to disrupt endosomes, allowing the nucleic acid to avoid lysosomal degradation.
  • the nucleic acid can be targeted in vivo for cell specific uptake and expression, by targeting a specific receptor (see, e.g., PCT Publications WO 92/06180 dated April 16, 1992; WO 92/22635 dated December 23, 1992; WO92/20316 dated November 26, 5 1992; WO93/14188 dated July 22, 1993; WO 93/20221 dated October 14, 1993).
  • the nucleic acid can be introduced intracellularly and incorporated within host cell DNA for expression, by homologous recombination (Koller and Smithies, 1989, Proc. Natl. Acad. Sci. USA 86:8932-8935; Zijlstra et ⁇ l, 1989, Nature 342:435-438).
  • techniques for delivery involve direct administration, 0 e.g., by stereotactic delivery of such gene sequences to the site of the cells in which the gene sequences are to be expressed.
  • Additional methods that may be utilized to increase the overall level of gene expression and/or gene product activity include using targeted homologous recombination methods, as discussed above, to modify the expression characteristics of an endogenous 5 gene in a cell or microorganism by inserting a heterologous DNA regulatory element such that the inserted regulatory element is operatively linked with the endogenous gene in question.
  • Targeted homologous recombination can thus be used to activate transcription of an endogenous gene that is "transcriptionally silent", i.e., is not normally expressed or is normally expressed at very low levels, or to enhance the expression of an endogenous gene
  • target gene-expressing cells preferably autologous cells
  • Such cells may be either
  • the cells to be administered are non-autologous cells, they can be administered using well known techniques that prevent a host immune response against the introduced cells from developing.
  • the cells may be introduced in an encapsulated form which, while allowing for an exchange of components with the
  • compounds such as those identified via techniques such as those described above that are capable of modulating activity of an OSN regulatory region can be administered using standard techniques that are well known to those of skill in the
  • the compounds that are determined to modify OSN regulatory regionlinger activity or gene product activity can be administered to a patient at therapeutically effective doses to treat or ameliorate a disorder involving tumor or tissue cells with calcification potential.
  • a therapeutically effective dose refers to that amount of the compound sufficient to result in amelioration of symptoms of such a disorder.
  • Toxicity and therapeutic efficacy of such compounds can be determined by standard pharmaceutical procedures in cell cultures or experimental animals, e.g., for determining the LD 50 (the dose lethal to 50% of the population) and the ED 50 (the dose therapeutically effective in 50%> of the population).
  • the dose ratio between toxic and - - therapeutic effects is the therapeutic index and it can be expressed as the ratio LD 50 /ED 50 .
  • Compounds that exhibit large therapeutic indices are preferred. While compounds that exhibit toxic side effects may be used, care should be taken to design a delivery system that targets such compounds to the site of affected tissue in order to minimize potential damage to uninfected cells and, thereby, reduce side effects.
  • the data obtained from the cell culture assays and animal studies can be used in formulating a range of dosage for use in humans.
  • the dosage of such compounds lies preferably within a range of circulating concentrations that include the ED 50 with little or no toxicity.
  • the dosage may vary within this range depending upon the dosage form employed and the route of administration utilized.
  • the therapeutically effective dose can be estimated initially from cell culture
  • a dose may be formulated in animal models to achieve a circulating plasma concentration range that includes the IC 50 (i.e., the concentration of the test compound that achieves a half-maximal inhibition of symptoms) as determined in cell culture. Such information can be used to more accurately determine useful doses in humans. Levels in plasma may be measured, for example, by high performance liquid chromatography.
  • compositions for use in accordance with the present invention may be formulated in conventional manner using one or more physiologically acceptable carriers or excipients.
  • the compounds and their physiologically acceptable salts and solvates may be formulated for administration by inhalation or insufflation (either through the mouth or the nose) or oral, buccal, parenteral or rectal administration.
  • the pharmaceutical compositions may take the form of, for example, tablets or capsules prepared by conventional means with pharmaceutically acceptable excipients such as binding agents (e.g., pregelatinised maize starch, polyvinylpyrrolidone or hydroxypropyl methylcellulose); fillers (e.g., lactose, microcrystalline cellulose or calcium hydrogen phosphate); lubricants (e.g., magnesium stearate, talc or silica); disintegrants (e.g., potato starch or sodium starch glycolate); or wetting agents (e.g., sodium lauryl sulphate).
  • binding agents e.g., pregelatinised maize starch, polyvinylpyrrolidone or hydroxypropyl methylcellulose
  • fillers e.g., lactose, microcrystalline cellulose or calcium hydrogen phosphate
  • lubricants e.g., magnesium stearate, talc or silica
  • disintegrants e.g., potato starch
  • Liquid preparations for oral administration may take the form of, for example, solutions, syrups or suspensions, or they may be presented as a dry product for constitution with water or other suitable vehicle before use.
  • Such liquid preparations may be prepared by conventional means with pharmaceutically acceptable additives such as suspending agents (e.g., sorbitol syrup, cellulose derivatives or hydrogenated edible fats);
  • JJ emulsifying agents e.g., lecithin or acacia
  • non-aqueous vehicles e.g., almond oil, oily esters, ethyl alcohol or fractionated vegetable oils
  • preservatives e.g., methyl or propyl-p-hydroxybenzoates or sorbic acid
  • the preparations may also contain buffer salts, flavoring, coloring and sweetening agents as appropriate.
  • Preparations for oral administration may be suitably formulated to give controlled release of the active compound.
  • compositions may take the form of tablets or lozenges formulated in conventional manner.
  • the compounds for use according to the present invention are conveniently delivered in the form of an aerosol spray presentation from pressurized packs or a nebuliser, with the use of a suitable propellant, e.g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas.
  • a suitable propellant e.g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas.
  • the dosage unit may be determined by providing a valve to deliver a metered amount.
  • Capsules and cartridges of e.g., gelatin for use in an inhaler or insufflator may be formulated containing a powder mix of the compound and a suitable powder base such as lactose or starch.
  • the compounds may be formulated for parenteral administration by injection, e.g., by bolus injection or continuous infusion.
  • Formulations for injection may be presented in unit dosage form, e.g., in ampoules or in multi-dose containers, with an added preservative.
  • the compositions may take such forms as suspensions, solutions or emulsions in oily or aqueous vehicles, and may contain formulatory agents such as suspending, stabilizing and/or dispersing agents.
  • the active ingredient may be in powder form for constitution with a suitable vehicle, e.g., sterile pyrogen-free water, before use.
  • the compounds may also be formulated in rectal compositions such as suppositories or retention enemas, e.g., containing conventional suppository bases such as cocoa butter or other glycerides.
  • compositions of the invention may be desirable to administer locally to the area in need of treatment.
  • This may be achieved by, for example, and not by way of limitation, local infusion during surgery, topical application, e.g., in conjunction with a wound dressing after surgery, by injection, by means of a catheter, by means of a suppository, or by means of an implant, said implant being of a porous, non-porous, or gelatinous material, including membranes, such as sialastic membranes, or fibers.
  • administration can be by direct injection at the site (or former site) of a malignant tumor or neoplastic or pre-neoplastic tissue.
  • the compounds may be combined with a carrier so that an effective dosage is delivered, based on the desired activity.
  • the compounds also may be formulated as a depot preparation. Such long acting formulations may be administered by implantation (for example subcutaneously or intramuscularly) or by intramuscular injection.
  • the compounds may be formulated with suitable polymeric or hydrophobic materials (for example as an emulsion in an acceptable oil) or ion exchange resins, or as sparingly soluble derivatives, for example, as a sparingly soluble salt.
  • suitable polymeric or hydrophobic materials for example as an emulsion in an acceptable oil
  • ion exchange resins for example as sparingly soluble derivatives, for example, as a sparingly soluble salt.
  • the compositions may, if desired, be presented in a pack or dispenser device that may contain one or more unit dosage forms containing the active ingredient.
  • the pack may for example comprise metal or plastic foil, such as a blister pack.
  • the pack or dispenser device may be accompanied by instructions for administration.
  • LNCaP, C4-2, C4-2B, PC3, PC3M, P69, DU145, MG, MG63, 9069E, ROS and WH cells were maintained in T medium supplemented with 5%> FBS.
  • cells were growth in phenol-red free serum-free RPMI 1640 (Gibco BRL, MD).
  • 293 cell line was purchased from Microbix Biosystems Inc. (Toronto, Ontario, Canada) and maintained in Minimal Eagles Medium (MEM) supplemented with 10% fetal calf serum and glutamine (Gibco BRL).
  • All promoter constructs were generated by TOPO TA cloning system (Invitrogene, CA) and subsequently digested using appropriate restriction sites in the poly linker to allow insertion into the vector pGL3 -basic (Promega) containing the coding region of the firefly luciferase gene.
  • the OSN 2.3, OSN 1.5, OSN 1.1 and OSN 0.2 promoter constructs were prepared using the primer sequences PI, P2, P3 and P4 as shown in Figure 1. Additional OSN promoter deletion constructs starting at -522 were prepared.
  • constructs which also contain a deletion of a spacer bewteen GGA box 1 and 2, contain -522 to +39, -522 to +62 and -522 to +73 of the OSN sequence (See Figures 1 and 11).
  • PCR was performed utilizing the following primers: 522-N: (5 ⁇ CTAGTAGCAGCTTGTCTTGTC3'), spdel-C: (5'CTTCTCCCCTGTCTCTGTCTT3'); and spdel-N:
  • Intron-C (5'TACCTCAGTGGCAGGCAGGCAG3')
  • Exon-C (5'CAGGCAGGCAGGCGGCAG')
  • Hafner-C (5'GCGCGCTCTCCGGGCAGTCTG3') to construct hON-5221, hON-522E and hON- Hafner, respectively, and the genomic DNA isolated from DU145 cells as template. All constructs, including the PCR-generated DNA fragments, were confirmed by sequencing.
  • Figure 1 represents an OSN regulatory region from -1409 to +904. Also shown on Figure 1 are the GGA box-1, GGA box-2 and the boundary between exon 1 and intron 1. Similarly, Figure 11 represents a human osteonectin promoter sequence from -522 to +62 lacking a spacer between GGA box 1 and 2.
  • FIG. 1 is a schematic representation of the strategy used to construct the OSN promoter-mediated luciferase plasmids.
  • Figure 7 is a schematic representation of the OSN sequence which was used to generate the various deletion constructs.
  • ROS, LNCaP, C4-2 and C4-2B cells were seeded at 3.3 xlO 5 cells/well in 6- well plates 2 days before transfection.
  • PC3, PC3M, DU145, MG63, 9069E and WH cells were plated at 2 xlO 5 cells/well in 6-well plates 1 day prior to transfection.
  • Plasmid DNA was introduced into cells by DOTAP reagent following the supplier's protocol (Boehringer Mannheim, IN). Briefly, the medium was replaced by 1 ml of serum-free and phenol-red free RPMI 1640 medium, and the cells were incubated for 6 h with a DOTAP-DNA mixture containing 3 ⁇ g DNA and 10 ⁇ l of DOTAP.
  • the medium was then replaced by normal growth medium.
  • the cells were cotransfected in a 5:1 molar ratio with the vector pCMV- ⁇ -gal encoding the reporter gene ⁇ -galactosidase.
  • luciferase activity assay 20 ⁇ l of supernatant was mixed with 100 ⁇ l of luciferase substrate (Promega, WI) and luciferase activity was measured by a luminometer (Monolight 2010, Analytical luminescence Laboratory, MD).
  • ⁇ -gal activity assay 10 100 ⁇ l of supernatant was mixed with an equal volume of 2X ⁇ -gal substrate (Promega, WI) and then incubated at 37°C for 15-30 minutes. The ⁇ -gal activity was determined by a microplate reader at 405 nm wavelength. Luciferase activity was calculated relative to the ⁇ -galactosidase activity. As a reference control, the basic promoter vector pGL3-TATA was used. At least three independent transfections were performed in duplicate for each 15 construct.
  • OSN-promoter deletion constructs were analyzed in both prostatic (LNCaP, C4-2, C4-2B, PC-3, P69) and non-prostatic (WH, MG) human epithelial
  • Figure 9 further shows a comparison of hON522E activity in different prostate cell lines, with PC3M cells having the highest activity.
  • a novel aspect of the present invention is that the expression of OSN in a cell type-specific manner was observed upon the removal of intron 1 and by extending the OSN promoter from GGA box 1 to include the entire exon 1 ( Figure 1).
  • inhibitory cis-elements that exist in the OSN promoter e.g. intron 1
  • intron 1 inhibitory cis-elements that exist in the OSN promoter
  • TEL-Test INC, TX
  • 2.5 ⁇ l of the RT reaction was adjusted to contain 25 ng of each pair of human osteonectin-specific primers (5 CCACCACCCTGTTGCTGT3', sense; and 5'CTCCAGGCGCTTCTCATT3', 5 antisense) or glyceraldehyde-3-phosphate dehydrogenase (GAPDH)-specific primers (5' ACCACAGTCCATGCCATCA3', sense; 5 CCACCACCCTGTTGCTGT3', antisense); an additional buffer was added to a total volume of 25 ⁇ l.
  • human osteonectin-specific primers 5 CCACCACCCTGTTGCTGT3', sense; and 5'CTCCAGGCGCTTCTCATT3', 5 antisense
  • GPDH glyceraldehyde-3-phosphate dehydrogenase
  • PCR was then performed for 50 cycles (94°C, 30 second; 55°C, 30 second; and 72°C, 1 min) for detecting the expression of osteonectin and 25 cycles for detecting the expression of GAPDH as an internal control. 0 Finally, the amplified products were separated on a 1.5%> agarose gel.
  • Figure 5 shows OSN expression in various prostate cancer cell lines, including LNCaP, C4-2, C4-2B, DU145, PC3, PC3M and MG63. GAPDH expression was 5 also monitored as an internal control.
  • Figure 6 shows OSN expression in primary prostate cancer tissues. Samples that revealed detectable OSN mRNA are in lanes 1, 2 and 5-7. Again, as with Figure 5, GAPDH expression was monitored as an internal control.
  • Ad-522E-TK adenovirus type 5
  • the plasmid p522E-TK containing a human osteonectin promoter and herpes simplex virus TK gene was constructed by inserting the 5 expression cassette into the El A deleted region of Ad5 adeno viral shuttle vector, p ⁇
  • Ad522E-TK adenovirus was generated in 293 cells by co-transfecting these cells with both the expression shuttle plasmid and a circular Ad genome plasmid (pJM17) using the standard calcium-phosphate precipitation method. Briefly, low passage ( ⁇ 40) 293 cells were seeded in 60-mm dishes to be about 70-80% confluent at the time of use.
  • Ad-522E-TK virus was obtained by plaque purification according to the method of Graham and Prevec (Molecular Biotechnology 3: 207, 1995). Plaques were picked up and isolated from transfected cultures and infected 293 cells. Cells were harvested 36-48 hrs after infection, pelleted, resuspended in PBS, and lysed. The virus in the cell lysate was purified by CsCl 2 gradient centrifugation. After dialysis, the concentrated virus was evaluated by particle count as determined by optical density measurement of DNA, and stored at -80°C before experimentation.
  • PC3M and MG63 cells were seeded onto 24-well plates at a density of 2x 10 4 cells/well. After 24 hr, the cells were infected with PBS or increasing m.o.i. of Ad-522E- TK in 1 ml of growth medium. After overnight infection, the virus was removed and cells were incubated in fresh medium containing 0 or 10 ⁇ g/ml of GCV for an additional 5 days. Duplicate plates were fixed and stained with crystal violet.
  • Figure 10A shows that PC3M cells with virus plus GCV did not proliferate as well as the cells without GCV.
  • Figure 10B shows the results using the MG-63 cell line. Similar to the results with PC3M, the virus plus GCV treated cells did not proliferate as much as the cells in the control group (without GCV). 10 Example: Inhibition of Tumor Growth In Vivo
  • mice Male athymic nu/nu mice (Harlan Co., TX), 5-6 weeks of age, were inoculated subcutaneously with 5x10 5 PC3M cells in 50 ⁇ l PBS into both sites of the flank. When the tumor became palpable (3-4 mm in diameter), the animals were randomly assigned to 4 experimental groups: group 1, PBS treatment; group 2, GCV only; group 3, Ad-522E-TK plus PBS; and group 4, Ad-522E-TK plus GCV. Fifty ⁇ l of Ad-522E-TK (2xl0 9 pfu) in PBS-10% glycerol was intratumorally injected on days 8, 11 and 14.
  • the data shown in Figure 12 demonstrate that the virus plus prodrug treated group had minimal tumor growth over the 21 day period of the experiment while the three control groups had quite a large increase in tumor growth.
  • the data demonstrate that the Ad-522E-TK viral vector is effective at ameliorating and/or treating tumors with calcification potential in vivo.
  • the present invention unexpectedly demonstrates that OSN promoter-driven genes, when infected into susceptible osteotropic tumors and cells of osteoblastic lineage, such as osteosarcomas (ROS, MG63, Saos-2), prostate (LNCaP, C4-2, C4-2B4 PC-3 PC3M, Du-145, ARCaP), colon (Lovo), lung (A547), brain (U-87), and breast (MCF-7) express very high levels of reporter gene, and thus are expected also to efficiently express high levels of chosen toxic or therapeutic gene when in combination with known delivery vector, including, but not limited to, Ad-OSN-TK.
  • the invention demonstrates that addition of a suitable prodrug, such as GCV, when combined with Ad-OSN-TK system, will effect the cessation, inhibition and cytotoxicity of said osteotropic tumors and their associated osteoblastic supporting stroma.
  • a suitable prodrug such as GCV
  • GCV Ad-OSN-TK
  • the present invention unexpectedly teaches that OSN promoter constructs constitute an essential tumor-specific gene therapy that can inhibit the growth of certain osteotropic tumors (osteosarcoma, prostate, etc.) while sparing the surrounding normal tissues, or non-osteoblastic or non-osteolytic lineage cells of significant damage, and when applied systemically can prevent destruction of inappropriate tissue, while still obtaining the desired destructive effect on tumor growth.
  • the OSN promoter-driven therapeutic gene therapy is superior to conventional gene therapies for osteotropic tumors when the therapeutic gene expression is driven by universal promoters such as cytomegalovirus (CMV) and the long terminal repeat promoters of Rous Sarcoma Virus (RSV), because these universal promoters cannot distinguish the specific o targeted tumor, and thus may cause inappropriate damage to nonselected tissues by expression of toxic gene in normal cells.
  • CMV cytomegalovirus
  • RSV Rous Sarcoma Virus
  • osteotroblastic cells may 5 synthesize and secrete products that are able to stimulate the growth, adhesion, and migration of osteotropic tumors such as prostate and breast cancer, and thus the reciprocal relationship between foreign tumor epithelium (cancer) and supporting bone stroma provides a favorable environment (soil) for accelerated tumor growth.
  • the proliferation and migration of prostate or breast tumor cells may also themselves secrete paracrine growth 0 factors that stimulate osteoblast or osteoclast bone cell growth at sites of bone metastases that results in induction of predominantly osteoblastic (e.g. prostate) or osteolytic (e.g.
  • OSN driven toxic gene therapy most commonly in the form of Ad-OSN-TK, exhibits various therapeutic 0 implications: a) OSN promoter driven gene therapy (Ad-OSN-TK) affects the expression of toxic compounds to osteosarcoma and prostate cancer cells and also eradicates osteoblastic cells that may be required to maintain the survival of osseous metastatic deposits of certain osteotropic tumors.
  • Ad-OSN-TK OSN promoter driven gene therapy
  • OSN promoter driven gene application can express high levels of therapeutic target genes in many calcified tumors, and is a reasonable choice for eradicating these tumors primary focus and its metastatic deposits in conjunction with the supporting stroma; c) OSN promoter-driven therapeutic gene treatments in conjunction with appropriate vehicles may be used in conjunction with conventional chemotherapy, surgery or radiation techniques or other novel therapies in reducing tumor burden and associated morbidity in local and metastatic deposits associated with various susceptible osteotropic human or eukaryotic tumors; d) Long lasting anti-tumor immunity might be elected against the remaining osteoblastic cells and tumor cells from OSN promoter-driven killing of tumor cells; e) OSN-promoter driven constructs may be used for the delivery and expression of therapeutic genes for the treatment of benign diseases such as BPH, arterial sclerosis, etc.
  • benign diseases such as BPH, arterial sclerosis, etc.
  • Prostate cancer metastasizes primarily to the skeleton.
  • the present invention demonstrates that OSN is expressed highly in a wide variety of prostate cancer cells, and that the OSN promoter can efficiently drive expression of chosen reporter gene and thus also drive a toxic and/or therapeutic gene when in conjunction with this promoter, and a gene delivery vehicle. This expression includes both osteoblastic prostate cancer cells and osteolytic prostate cancer cells.
  • the present invention will also be useful in other tumors, including, but not limited to, melanoma, thyroid, gastric, ovarian, osteosarcoma, colon, lung, breast, and brain tumors.
  • a novel therapeutic agent comprising an OSN promoter linked with an appropriate delivery vector and therapeutic (or toxic) gene is conceived to be generated, including, but not limited to, Ad-OSN-TK.
  • Ad-OSN-TK an appropriate delivery vector and therapeutic (or toxic) gene
  • This recombinant, novel system will efficiently express therapeutic action and selectively target and induce the killing of osteoblast lineage cells and a wide spectrum of tumors or other susceptible benign tissues that have acquired the potential ability to calcify.
  • a new recombinant therapy agent such as, but not limited to Ad-
  • OSN-TK will be available to patients afflicted not only with osteosarcoma or prostate cancer, but also lung, breast, thyroid, myeloma, melanoma, colon, brain, gastric, ovarian and other calcifying tumors, or susceptible benign tissues.
  • toxins or therapeutic genes may be used with the OSN promoter- driven therapy instead of the mentioned Tk. These include genes for cytosine deaminase, tumor suppresser genes, cyclic regulatory proteins, including various cytokines, growth or differentiation factors and others all can be ligated to OSN promoter in place of the mentioned Tk gene. In addition, other vector delivery systems can be generated and combined with the OSN promoter to effectively elicit the desired therapeutic response.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Organic Chemistry (AREA)
  • Genetics & Genomics (AREA)
  • General Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Medicinal Chemistry (AREA)
  • Public Health (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Animal Behavior & Ethology (AREA)
  • Biotechnology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Engineering & Computer Science (AREA)
  • Zoology (AREA)
  • Molecular Biology (AREA)
  • Wood Science & Technology (AREA)
  • Biomedical Technology (AREA)
  • Plant Pathology (AREA)
  • Biophysics (AREA)
  • Physics & Mathematics (AREA)
  • Microbiology (AREA)
  • Epidemiology (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Biochemistry (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
  • Investigating Or Analysing Biological Materials (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
  • Medicinal Preparation (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

La présente invention concerne des promoteurs, des activateurs et d'autres éléments régulateurs qui régulent l'expression dans des cellules de tumeurs et de tissus possédant un potentiel de calcification. L'invention concerne spécifiquement des vecteurs d'expression, des cellules hôtes et des animaux transgéniques comportant une séquence régulatrice de l'ostéonectine (OSN).
PCT/US2000/014482 1999-05-28 2000-05-25 Therapie genique toxique a base d'osteonectine destinee au traitement de tumeurs et de tissus calcifies WO2000072679A1 (fr)

Priority Applications (5)

Application Number Priority Date Filing Date Title
CA002374141A CA2374141A1 (fr) 1999-05-28 2000-05-25 Therapie genique toxique a base d'osteonectine destinee au traitement de tumeurs et de tissus calcifies
JP2000620801A JP2003500422A (ja) 1999-05-28 2000-05-25 石灰化した腫瘍および組織を治療するためのオステオネクチンに基づく毒性遺伝子治療
EP00937791A EP1182929A4 (fr) 1999-05-28 2000-05-25 Therapie genique toxique a base d'osteonectine destinee au traitement de tumeurs et de tissus calcifies
AU52918/00A AU5291800A (en) 1999-05-28 2000-05-25 Osteonectin based toxic gene therapy for the treatment of calcified tumors and tissues
IL14679400A IL146794A0 (en) 1999-05-28 2000-05-25 Osteonectin based toxic gene therapy for the treatment of calcified tumors and tissues

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US13644099P 1999-05-28 1999-05-28
US60/136,440 1999-05-28

Publications (1)

Publication Number Publication Date
WO2000072679A1 true WO2000072679A1 (fr) 2000-12-07

Family

ID=22472869

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2000/014482 WO2000072679A1 (fr) 1999-05-28 2000-05-25 Therapie genique toxique a base d'osteonectine destinee au traitement de tumeurs et de tissus calcifies

Country Status (6)

Country Link
EP (1) EP1182929A4 (fr)
JP (1) JP2003500422A (fr)
AU (1) AU5291800A (fr)
CA (1) CA2374141A1 (fr)
IL (1) IL146794A0 (fr)
WO (1) WO2000072679A1 (fr)

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1308517A1 (fr) * 2001-10-31 2003-05-07 Aventis Pharmacueticals Products Inc. Vecteurs pour l'expression de multiples transgenes
EP1644530A2 (fr) * 2003-06-24 2006-04-12 Johns Hopkins University Biomarqueurs geniques methyles servant a detecter le cancer
US7060275B2 (en) 2001-10-17 2006-06-13 Agy Therapeutics, Inc. Use of protein biomolecular targets in the treatment and visualization of brain tumors
WO2007127347A3 (fr) * 2006-04-28 2008-01-03 Inis Biotech Llc Fragment d'adn isolé du promoteur humain du gène sparc et son utilisation
US7638272B2 (en) 2003-01-14 2009-12-29 Dana-Farber Cancer Institute Cancer therapy sensitizer
US8420603B2 (en) 2004-05-14 2013-04-16 Abraxis Bioscience, Llc SPARC and methods of use thereof
US9671406B2 (en) 2006-03-31 2017-06-06 Abraxis Bioscience, Llc SPARC and methods of use thereof
US10987308B2 (en) 2014-09-03 2021-04-27 Genesegues, Inc. Therapeutic nanoparticles and related compositions, methods and systems
CN114533959A (zh) * 2022-04-02 2022-05-27 山东隽秀生物科技股份有限公司 一种肌腱修复材料、制备方法及在制备肌腱修复产品中的应用

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5416017A (en) * 1990-05-18 1995-05-16 The Scripps Research Institute Cholera toxin gene regulated by tissue-specific promoters

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0950097A2 (fr) * 1996-12-27 1999-10-20 Instituto de Investigaciones Bioquimicas Fundacion Campomar Compositions et methodes destines au traitement de tumeurs
US5772993A (en) * 1997-01-21 1998-06-30 The University Of Virginia Patent Foundation Osteocalcin promoter-based toxic gene therapy for the treatment of calcified tumors and tissues

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5416017A (en) * 1990-05-18 1995-05-16 The Scripps Research Institute Cholera toxin gene regulated by tissue-specific promoters

Non-Patent Citations (5)

* Cited by examiner, † Cited by third party
Title
ANDERSON W.F.: "Human gene therapy", NATURE, vol. 392, 30 April 1998 (1998-04-30), pages 25 - 30, XP002924246 *
DATABASE CAPLUS ON STN DAMJONVSKI ET AL.: "Regulation of SPARC expression during early xenopus development: evolutionary divergence and conservation of DNA regulatory elements between amphibians and mammals", XP002924285 *
DEV. GENES EVOL., vol. 207, no. 7, 1998, pages 453 - 461 *
See also references of EP1182929A4 *
VERMA ET AL.: "Gene therapy- promises, problems and prospects", NATURE, vol. 389, 18 September 1997 (1997-09-18), pages 239 - 242, XP002924266 *

Cited By (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7060275B2 (en) 2001-10-17 2006-06-13 Agy Therapeutics, Inc. Use of protein biomolecular targets in the treatment and visualization of brain tumors
EP1308517A1 (fr) * 2001-10-31 2003-05-07 Aventis Pharmacueticals Products Inc. Vecteurs pour l'expression de multiples transgenes
US7638272B2 (en) 2003-01-14 2009-12-29 Dana-Farber Cancer Institute Cancer therapy sensitizer
US8071294B2 (en) 2003-01-14 2011-12-06 Dana-Farber Cancer Institute Cancer therapy sensitizer
US7879542B2 (en) 2003-06-24 2011-02-01 The Johns Hopkins University Methylated gene biomarkers for detecting cancer
EP1644530A4 (fr) * 2003-06-24 2006-10-04 Univ Johns Hopkins Biomarqueurs geniques methyles servant a detecter le cancer
EP1644530A2 (fr) * 2003-06-24 2006-04-12 Johns Hopkins University Biomarqueurs geniques methyles servant a detecter le cancer
US8420603B2 (en) 2004-05-14 2013-04-16 Abraxis Bioscience, Llc SPARC and methods of use thereof
US9671406B2 (en) 2006-03-31 2017-06-06 Abraxis Bioscience, Llc SPARC and methods of use thereof
WO2007127347A3 (fr) * 2006-04-28 2008-01-03 Inis Biotech Llc Fragment d'adn isolé du promoteur humain du gène sparc et son utilisation
US8436160B2 (en) 2006-04-28 2013-05-07 Consejo Nacional De Investigaciones Cientificas Y Tecnicas (Conicet) Isolated DNA fragment of the SPARC human promoter and its use for driving the expression of an heterologous gene in tumor cells
AU2007243252B2 (en) * 2006-04-28 2013-06-20 Consejo Nacional De Investigaciones Cientificas Y Tecnicas (Conicet) An isolated DNA fragment of the SPARC human promoter and its use
US10987308B2 (en) 2014-09-03 2021-04-27 Genesegues, Inc. Therapeutic nanoparticles and related compositions, methods and systems
CN114533959A (zh) * 2022-04-02 2022-05-27 山东隽秀生物科技股份有限公司 一种肌腱修复材料、制备方法及在制备肌腱修复产品中的应用
CN114533959B (zh) * 2022-04-02 2022-12-09 山东隽秀生物科技股份有限公司 一种肌腱修复材料、制备方法及在制备肌腱修复产品中的应用

Also Published As

Publication number Publication date
EP1182929A1 (fr) 2002-03-06
JP2003500422A (ja) 2003-01-07
AU5291800A (en) 2000-12-18
IL146794A0 (en) 2002-07-25
CA2374141A1 (fr) 2000-12-07
EP1182929A4 (fr) 2003-04-16

Similar Documents

Publication Publication Date Title
KR20010030912A (ko) 종양 특이적인 세포독성을 유도할 수 있는 방법 및 조성물
WO2000072679A1 (fr) Therapie genique toxique a base d'osteonectine destinee au traitement de tumeurs et de tissus calcifies
Tsuruta et al. Combination effect of adenovirus-mediated pro-apoptotic bax gene transfer with cisplatin or paclitaxel treatment in ovarian cancer cell lines
KR20120132594A (ko) 마이크로rna를 이용한 조절을 통한 암 특이적 유전자 치료제
AU769773B2 (en) Bone sialoprotein based toxic gene therapy for the treatment of calcified tumors and tissues
JP2002543792A (ja) ベクターによるトランスポゾン配列の供給及び組込み
EP0999278B1 (fr) Eléments de régulation de spécificité vasculaire de la région d'encadrement 5' du gène de la desmine
US20030213006A1 (en) Beta-hcg promoter based tumor restrictive gene expression for cancer theraphy
WO2001090344A1 (fr) $g(b)-hcg promoter based tumor-restrictive gene expression for cancer gene therapy
EP1594549B1 (fr) Plasmide vegf inductible par l'hypoxie utile pour traiter une maladie ischemique
US20030219412A1 (en) Somatic gene therapy to suppress secondary cataract formation following eye surgery
WO2000024254A1 (fr) Compositions et procedes pour moduler l'expression a l'interieur des cellules de muscles lisses
EP1287362A2 (fr) Compositions et methodes destinees a la modulation de l'expression specifique des tumeurs
US6825035B1 (en) Compositions and methods for modulating expression within smooth muscle cells
US20040038232A1 (en) Compositions and methods for modulating tumor specific expression
US20030078224A1 (en) Gene expression directed by a super-PSA promoter
JP2003521895A (ja) スーパーpsaプロモーターにより指令された遺伝子発現
WO2001070175A2 (fr) Replication d'adenovirus sous controle de promoteur d'osteocalcine, utilisee a des fins therapeutiques
JP2002519025A (ja) ***形成を調節するための方法および組成物
WO2002059270A2 (fr) Procedes et compositions pour l'expression de polynucleotides specifiquement dans des cellules de muscle lisse in vivo
US20040101840A1 (en) Identification, cloning and characterization of a novel pc-1 gene differentially expressed in prostate cells and tissues
WO2003006621A2 (fr) Super agent promoteur de l'osteocalcine pour traiter les tumeurs calcifiees et les tissus calcifies
WO2001081547A2 (fr) Identification, clonage et caracterisation d'un nouveau gene pc-1 exprime differemment dans des cellules et des tissus de la prostate
JP2004504846A (ja) 癌治療用p972遺伝子をコードする発現ベクターおよびそれを生産するアデノウイルス

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BY CA CH CN CR CU CZ DE DK DM DZ EE ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX NO NZ PL PT RO RU SD SE SG SI SK SL TJ TM TR TT TZ UA UG US UZ VN YU ZA ZW

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): GH GM KE LS MW MZ SD SL SZ TZ UG ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE BF BJ CF CG CI CM GA GN GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
ENP Entry into the national phase

Ref document number: 2374141

Country of ref document: CA

Ref country code: CA

Ref document number: 2374141

Kind code of ref document: A

Format of ref document f/p: F

ENP Entry into the national phase

Ref country code: JP

Ref document number: 2000 620801

Kind code of ref document: A

Format of ref document f/p: F

WWE Wipo information: entry into national phase

Ref document number: 2000937791

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 52918/00

Country of ref document: AU

WWP Wipo information: published in national office

Ref document number: 2000937791

Country of ref document: EP

REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

WWW Wipo information: withdrawn in national office

Ref document number: 2000937791

Country of ref document: EP