WO1997011170A1 - Antisense oligonucleotide chemotherapy for benign hyperplasia or cancer of the prostate - Google Patents

Antisense oligonucleotide chemotherapy for benign hyperplasia or cancer of the prostate Download PDF

Info

Publication number
WO1997011170A1
WO1997011170A1 PCT/US1996/015081 US9615081W WO9711170A1 WO 1997011170 A1 WO1997011170 A1 WO 1997011170A1 US 9615081 W US9615081 W US 9615081W WO 9711170 A1 WO9711170 A1 WO 9711170A1
Authority
WO
WIPO (PCT)
Prior art keywords
seq
oligonucleotides
oligonucleotide
gene
group
Prior art date
Application number
PCT/US1996/015081
Other languages
French (fr)
Inventor
Paul A. Zamecnik
Original Assignee
Worcester Foundation For Biomedical Research
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 Worcester Foundation For Biomedical Research filed Critical Worcester Foundation For Biomedical Research
Priority to EP96935879A priority Critical patent/EP0851919A1/en
Priority to AU73662/96A priority patent/AU7366296A/en
Publication of WO1997011170A1 publication Critical patent/WO1997011170A1/en

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
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • C12N15/1136Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing against growth factors, growth regulators, cytokines, lymphokines or hormones
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • C12N15/1138Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing against receptors or cell surface proteins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/31Chemical structure of the backbone
    • C12N2310/315Phosphorothioates
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/35Nature of the modification
    • C12N2310/351Conjugate
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/50Physical structure
    • C12N2310/53Physical structure partially self-complementary or closed

Definitions

  • the present invention relates to the field of chemotherapy for hyperplasias and cancers and, in particular, to chemotherapy for benign hyperplasia or cancer ofthe prostate.
  • the invention relates to the field of antisense oligonucleotides and their use in human hyperplasia and cancer therapy.
  • BPH Bacillus subtilis
  • This condition may be a precursor to full blown prostatic cancer or may continue for decades without evolving into the deadly carcinoma.
  • treatment may range from "watchful waiting” to more aggressive approaches employing anti-androgen hormonal therapy, transurethral resection, or radical prostatectomy (see, e.g., Catalona (1994)).
  • the androgen receptor (AR) binds the male hormone testosterone and, acting at the transcriptional level, regulates the growth of normal prostatic cells.
  • a cDNA for the human AR was disclosed by Lubahn et al. (1988).
  • anti-androgen or estrogen hormonal therapy including physical or chemical castration, may be effective against early stage prostate cancer but, after a period of roughly 18 months, the patient becomes refractory to the hormonal therapy. The relapse is believed to be the result ofthe development or clonal selection of androgen-independent tumor cells in which the AR has mutated or been lost (see, e.g., Taplin, et al. (1995); Klocker, et al. (1994).
  • transfection with an AR cDNA has been shown to inhibit growth in the presence of testosterone (Suzuki, et al. (1994)).
  • the acidic fibroblast growth factor also known as the heparin binding growth factor type one (HBGF-1)
  • HBGF-1 heparin binding growth factor type one
  • An mRNA sequence for a human allele of ⁇ FGF was disclosed in Harris, et al. (1991). Mansson. et al. (1989) found that ⁇ FGF was expressed in normal immature rat prostate but not in normal mature rat prostate. In cancerous rat prostatic cell lines, they found ⁇ FGF expression similar to that in immature rat prostate.
  • the present invention provides methods for treating a patient diagnosed as having benign prostatic hype ⁇ lasia or a prostatic cancer.
  • the methods include administering to the patient a therapeutically effective amount of a composition comprising an antisense oligonucleotide which selectively hybridizes to an AR or ⁇ FGF gene or mRNA sequence ofthe patient, thereby inhibiting the expression ofthe AR or ⁇ FGF gene or mRNA sequence.
  • This inhibition of the AR or ⁇ FGF genes or mRNAs by antisense oligonucleotides results in a significant inhibition ofthe growth or survival of prostatic cells.
  • the methods provide a useful new means of treating benign prostatic hype ⁇ lasia and prostatic cancer.
  • the methods are particularly useful in treating prostate cancer patients who have become refractory to anti-androgen hormonal therapy.
  • the AR antisense oligonucleotides may comprise at least 10 consecutive bases from SEQ ID NO.: 1, at least 10 consecutive bases from a genomic sequence corresponding to SEQ ID NO.: 1, or oligonucleotides that hybridize to the complements of these sequences under physiological conditions. More preferably, the antisense oligonucleotides comprise at least 15 consecutive bases, and most preferably, 20-30 consecutive bases from the above-described sequences.
  • the ⁇ FGF antisense oligonucleotides may comprise at least 10 consecutive bases from any one of SEQ ID NO.: 2, SEQ ID NO.: 3 or SEQ ID NO.: 4, at least 10 consecutive bases from the joined exons of SEQ ID NO.: 2, SEQ ID NO.: 3 and SEQ ID NO.: 4; or oligonucleotides that hybridize to the complements of these sequences under physiological conditions. More preferably, the antisense oligonucleotides comprise at least 15 consecutive bases, and most preferably, 20-30 consecutive bases from the above-described sequences.
  • sequences ofthe invention include, but are not limited to, those disclosed as SEQ ID NO.: 5, SEQ ID NO.: 6, SEQ ID NO.: 7, and SEQ ID NO.: 8.
  • all ofthe above-described oligonucleotides are modified oligonucleotides.
  • the modified oligonucleotide includes at least one synthetic internucleoside linkage such as a phosphorothioate, alkylphosphonate, phosphorodithioate, phosphate ester, alkylphosphonothioate, phosphoramidate, carbamate, carbonate, phosphate triester, acetamidate, or carboxymethyl ester.
  • the modified oligonucleotide has at least one low molecular weight organic group covalently bound to a phosphate group of said oligonucleotide. In another set of embodiments, the modified oligonucleotide has at least one low molecular weight organic group covalently bound to a 2' position of a ribose of said oligonucleotide.
  • Such low molecular weight organic groups include lower alkyl chains or aliphatic groups (e.g., methyl, ethyl, propyl, butyl), substituted alkyl and aliphatic groups (e.g., aminoethyl, aminopropyl, aminohydroxyethyl, aminohydroxypropyl), small saccharides or glycosyl groups.
  • the modified oligonucleotide has covalently attached thereto a prostate-targeting compound such as an androgen, androgen derivative, estrogen, estrogen derivative, estramustine, emcyt or estracyt.
  • a prostate-targeting compound such as an androgen, androgen derivative, estrogen, estrogen derivative, estramustine, emcyt or estracyt.
  • the antisense oligonucleotides are administered intravenously at a dosage between 1.0 ⁇ g and 100 mg per kg body weight of the patient.
  • the present invention also provides for any or all ofthe above-described antisense oligonucleotides, including the various modified oligonucleotides, in a pharmaceutical composition.
  • the antisense oligonucleotides are admixed with a sterile pharmaceutically acceptable carrier in a therapeutically effective amount such that the isolated antisense oligonucleotide selectively hybridizes to the AR or ⁇ FGF gene or mRNA sequence when administered to a patient.
  • a pharmaceutical kit is also provided in which such a pharmaceutical composition is combined with a pharmaceutically acceptable carrier for intravenous administration.
  • the methods and products ofthe present invention further include antisense oligonucleotides, as described above, directed at a PSA gene, a probasin gene, an estrogen receptor gene, a telomerase gene, a prohibitin gene, a src gene, a ras gene, a myc gene, a blc-2 gene, a protein kinase-A gene, a plasminogen activator urokinase gene and a methyl transferase gene.
  • the present invention provides new methods for the treatment of cancer ofthe prostate and pharmaceutical compositions useful therefor. It is now disclosed that antisense oligonucleotides complementary to genes which are expressed predominantly or strongly in prostatic cells are effective for inhibiting the growth of and/or killing hype ⁇ lastic or cancerous cells of prostatic origin.
  • the present invention provides oligonucleotides, including modified oligonucleotides. which have antisense homology to a sufficient portion of either the AR or ⁇ FGF gene such that they inhibit the expression of that gene. Su ⁇ risingly, inhibition of either of these genes, even in androgen-resistant prostatic cancer cells, inhibits the growth of these cells. Because the antisense oligonucleotides ofthe invention can be administered systemically but selectively inhibit prostate cells, the present invention has particular utility in late stage prostate cancer which has metastasized.
  • AR refers to the androgen receptor well known in the art and described in the various references cited herein.
  • a cDNA sequence ofthe human AR gene was disclosed in Lubahn et al. (1988). The Lubahn et al. (1988)sequence is available on GenBank (Accession number J03180) and is reproduced here as SEQ. ID NO.: 1. The translation initiation codon of this gene is found at base positions 363-365 and the stop codon is at positions 3120-3122 of SEQ ID NO. : 1. As will be obvious to one of ordinary skill in the art.
  • ⁇ FGF refers to the ⁇ FGF protein known in the art and described in the various references cited herein.
  • the genomic DNA of one allele ofthe human ⁇ FGF gene has been partially sequenced and was disclosed in Wang et al. (1989). The Wang et al.(1989) sequences cover the three exons ofthe ⁇ FGF gene as well as some 5', 3' and intron sequences.
  • GenBank accesion numbers M23017, M23086 and M23087
  • SEQ. ID NO.: 2 SEQ ID NO.: 3
  • SEQ ID NO.: 4 A partial cDNA sequence for a human ⁇ FGF gene also may be found in Harris et al. (1991). The locations ofthe exons are located in the sequence listings. The translation initiation codon is found at positions 602-604 of SEQ ID NO.: 2 and the stop codon is found at positions 496-498.
  • ⁇ FGF gene including other human alleles and homologues from other mammalian species, encoding an ⁇ FGF protein and hybridizing to one or more of SEQ ID NO.: 2, SEQ ID NO.: 3 or SEQ ID NO.: 4 under stringent hybridization conditions, will exist in natural populations and are embraced by the term " ⁇ FGF gene" as used herein.
  • antisense oligonucleotide or “antisense” describes an oligonucleotide that is an oligoribonucleotide, oligodeoxyribonucleotide, modified oligoribonucleotide, or modified oligodeoxyribonucleotide which hybridizes under physiological conditions to DNA comprising a particular gene or to an mRNA transcript of that gene and, thereby, inhibits the transcription of that gene and/or the translation of that mRNA.
  • an "AR-antisense oligonucleotide” and by an “ ⁇ FGF-antisense oligonucleotide” are meant oligonucleotides which hybridize under physiological conditions to the AR gene/mRNA or ⁇ FGF gene/mRNA and, thereby, inhibit transcription/translation ofthe AR and ⁇ FGF genes/mRNAs, respectively.
  • the antisense molecules are designed so as to interfere with transcription or translation of AR or ⁇ FGF upon hybridization with the target.
  • the antisense oligonucleotide be selected so as to hybridize selectively with the target under physiological conditions, i.e., to hybridize substantially more to the target sequence than to any other sequence in the target cell under physiological conditions.
  • Stringent hybridization conditions means hybridization conditions from 30°C-60°C and from 5x to 0.1 x SSC. Highly stringent hybridization conditions are at 45 °C and O.lx SSC. "Stringent hybridization conditions" is a term of art understood by those of ordinary skill in the art.
  • stringent hybridization conditions are those conditions of temperature and buffer solution which will permit hybridization of that nucleic acid sequence to its complementary sequence and not to substantially different sequences.
  • the exact conditions which constitute "stringent” conditions depend upon the length ofthe nucleic acid sequence and the frequency of occurrence of subsets of that sequence within other non-identical sequences.
  • the present invention depends, in part, upon the discovery that the selective inhibition of the expression of AR or ⁇ FGF by antisense oligonucleotides in prostatic cells effectively inhibits cell growth and/or causes cell death.
  • antisense oligonucleotides Based upon SEQ ID NO.: 1, SEQ ID NO.: 2, SEQ ID NO.: 3 and SEQ ID NO.: 4, or upon allelic or homologous genomic or cDNA sequences, one of skill in the art can easily choose and synthesize any of a number of appropriate antisense molecules for use in accordance with the present invention.
  • such antisense oligonucleotides should comprise at least 10 and, more preferably, at least 15 consecutive bases which are complementary to the AR or ⁇ FGF mRNA transcripts. Most preferably, the antisense oligonucleotides comprise a complementary sequence of 20-30 bases.
  • oligonucleotides may be chosen which are antisense to any region of the AR or ⁇ FGF genes or mRNA transcripts, in preferred embodiments the antisense oligonucleotides correspond to N-terminal or 5' upstream sites such as translation initiation, transcription initiation or promoter sites. In addition, 3'-untranslated regions or telomerase sites may be targeted. Targeting to mRNA splicing sites has also been used in the art but may be less preferred if alternative mRNA splicing occurs.
  • the AR or ⁇ FGF antisense is, preferably, targeted to sites in which mRNA secondary structure is not expected (see, e.g., Sainio et al.
  • SEQ ID NO.: 1 discloses a cDNA sequence
  • SEQ ID NO.: 2 disclose genomic DNA sequences
  • one of ordinary skill in the art may easily derive the genomic DNA corresponding to the cDNA of SEQ ID NO.: 1 and may easily obtain the cDNA sequence corresponding to SEQ ID NO.: 2, SEQ ID NO.:3 and SEQ ID NO.: 4.
  • the present invention also provides for antisense oligonucleotides which are complementary to the genomic DNA corresponding to SEQ ID NO.: 1 and the cDNA corresponding to SEQ ID NO.: 2, SEQ ID NO.: 3 and SEQ ID NO.: 4.
  • antisense to allelic or homologous cDNAs and genomic DNAs are enabled without undue experimentation.
  • the antisense oligonucleotides of the present invention need not be perfectly complementary to the AR or ⁇ FGF genes or mRNA transcripts in order to be effective. Rather, some degree of mismatches will be acceptable if the antisense oligonucleotide is of sufficient length. In all cases, however, the oligonucleotides should have sufficient length and complementarity so as to hybridize to an AR or ⁇ FGF transcript under physiological conditions. Preferably, of course, mismatches are absent or minimal.
  • the antisense oligonucleotides may have one or more non-complementary sequences of bases inserted into an otherwise complementary antisense oligonucleotide sequence.
  • Such non-complementary sequences may "loop" out of a duplex formed by an AR or ⁇ FGF transcript and the bases flanking the non-complementary region. Therefore, the entire oligonucleotide may retain an inhibitory effect despite an apparently low percentage of complementarity.
  • self-stabilized or hai ⁇ in oligonucleotides are examples of self-stabilized or hai ⁇ in oligonucleotides.
  • Such oligonucleotides, or modified oligonucleotides have a sequence at the 5' and/or 3' end which is capable of folding over and forming a duplex with itself.
  • the duplex region which is preferably at least 4-6 bases joined by a loop of 3-6 bases, stabilizes the oligonucleotide against degradation.
  • These self-stabilized oligonucleotides are easily designed by adding the inverted complement of a 5 ' or 3' AR or ⁇ FGF sequence to the end of the oligonucleotide (see, e.g., Table 1, SEQ ID NO.: 6 and SEQ ID NO.: 7; Tang, J.-Y., et al. (1993) Nucleic Acids Res. 21 :2729-2735).
  • the AR and ⁇ FGF antisense oligonucleotides ofthe invention may be composed of "natural" deoxyribonucleotides, ribonucleotides, or any combination thereof. That is, the 5' end of one nucleotide and the 3' end of another nucleotide may be covalently linked, as in natural systems, via a phosphodiester intemucleoside linkage.
  • These oligonucleotides may be prepared by art recognized methods which may be carried out manually or by an automated synthesizer. In preferred embodiments, however, the antisense oligonucleotides ofthe invention also may include "modified" oligonucleotides.
  • modified oligonucleotide describes an oligonucleotide in which (1) at least two of its nucleotides are covalently linked via a synthetic intemucleoside linkage (i.e., a linkage other than a phosphodiester linkage between the 5' end of one nucleotide and the 3' end of another nucleotide) and/or (2) a chemical group not normally associated with nucleic acids has been covalently attached to the oligonucleotide.
  • a synthetic intemucleoside linkage i.e., a linkage other than a phosphodiester linkage between the 5' end of one nucleotide and the 3' end of another nucleotide
  • Preferred synthetic intemucleoside linkages are phosphorothioates, alkylphosphonates, phosphorodithioates, phosphate esters, alkylphosphonothioates, phosphoramidates, carbamates, carbonates, phosphate triesters, acetamidate, and carboxymethyl esters. Further, one or more of the 5'-3' phosphate group may be covalently joined to a low molecular weight (e.g., 15-500 Da) organic group.
  • a low molecular weight e.g. 15-500 Da
  • Such low molecular weight organic groups include lower alkyl chains or aliphatic groups (e.g., methyl, ethyl, propyl, butyl), substituted alkyl and aliphatic groups (e.g., aminoethyl, aminopropyl, aminohydroxyethyl, aminohydroxypropyl), small saccharides or glycosyl groups.
  • Other low molecular weight organic modifications include additions to the intemucleoside phosphate linkages such as cholesteryl or diamine compounds with varying numbers of carbon residues between the amino groups and terminal ribose.
  • Oligonucleotides with these linkages or other modifications can be prepared according to known methods (see, e.g., Agrawal and Goodchild (1987); Agrawal et al. (1988); Uhlmann et al. (1990); Agrawal et al. (1992); Agrawal (1993); and U.S. Pat. No. 5,149,798).
  • modified oligonucleotide also encompasses oligonucleotides with a covalently modified base and/or sugar.
  • modified oligonucleotides include oligonucleotides having backbone sugars which are covalently attached to low molecular weight organic groups other than a hydroxyl group at the 3' position and other than a phosphate group at the 5' position.
  • modified oligonucleotides may include a 2'-O-alkylated ribose group such as a 2'-O-methylated ribose.
  • modified oligonucleotides may include sugars such as arabinose instead of ribose.
  • the modified oligonucleotides may be branched oligonucleotides.
  • Unoxidized or partially oxidized oligonucleotides having a substitution in one or more nonbridging oxygen per nucleotide in the molecule are also considered to be modified oligonucleotides.
  • modified oligonucleotides are oligonucleotides having prostate- targeting, nuclease resistance-conferring, or other bulky substituents and/or various other stmctural modifications not found in vivo without human intervention.
  • the androgen receptor and other hormonal receptor sites on prostate cells allow for targeting antisense oligonucleotides specifically or particularly to prostatic cells.
  • Attachment ofthe antisense oligonucleotides by a molecular "tether" e.g., an alkyl chain
  • Estramustine targets particularly to the ventral prostate (Forsgren, et al. (1979)).
  • chemotherapeutic agents e.g., dexamethasone, vinblastine, etoposide
  • modified oligonucleotides are hybrid or chimeric oligonucleotides in which some but not all ofthe phosphodiester linkages, bases or sugars have been modified.
  • the currently most preferred modified oligonucleotides are 2'-O- methylated hybrid oligonucleotides. Since degradation occurs mainly at the 3' end, secondarily at the 5' end, and less in the middle, unmodified oligonucleotides located at this position can activate RNase H, and yet are degraded slowly. Furthermore, the T m of such a 27-mer is approximately 20 °C higher than that of a 27-mer all phosphorothioate oligodeoxynucleotide. This greater affinity for the targeted genomic area can result in greater inhibiting efficacy.
  • the number of synthetic linkages at the termini need not be ten and synthetic linkages may be combined with other modifications, such as alkylation of a 5' or 3' phosphate, or 2'-O- alkylation.
  • synthetic linkages may be combined with other modifications, such as alkylation of a 5' or 3' phosphate, or 2'-O- alkylation.
  • one may produce a modified oligonucleotide with the following stmcture, where B represents any base, R is an alkyl, aliphatic or other substituent, the subscript S represents a synthetic (e.g. phosphorothioate) linkage, and each n is an independently chosen integer from 1 to about 20:
  • the methods ofthe present invention represent new and useful additions to the field of benign prostate hype ⁇ lasia or prostate cancer therapy.
  • the methods ofthe present invention are especially useful for late stage prostate cancer in which metastases have occurred and in which the cells have become resistant to estrogen or anti-androgen therapy.
  • the methods may, however, also be used in benign prostate hype ⁇ lasia or early stage prostate cancer and may provide a substitute for more radical procedures such as transurethral resection, radical prostatectomy, or physical or chemical castration.
  • the products of the present invention include the isolated antisense oligonucleotides described above.
  • the term "isolated" as applied to an antisense oligonucleotide means not covalently bound to and physically separated from the 5' and 3' sequences which flank the corresponding antisense sequence in nature.
  • Administration ofthe AR or ⁇ FGF antisense oligonucleotides may be oral, intravenous,
  • parenteral cutaneous or subcutaneous.
  • the administration also may be localized to the prostate or to the region ofthe tumor by injection to or perfusion ofthe site.
  • AR or ⁇ FGF antisense oligonucleotides may be administered as part of a pharmaceutical composition.
  • a pharmaceutical composition may include the antisense oligonucleotides in combination with any standard physiologically and/or pharmaceutically acceptable carriers which are known in the art.
  • the compositions should be sterile and contain a therapeutically effective amount ofthe antisense oligonucleotides in a unit of weight or volume suitable for administration to a patient.
  • pharmaceutically acceptable means a non-toxic material that does not interfere with the effectiveness ofthe biological activity ofthe active ingredients.
  • physiologically acceptable refers to a non-toxic material that is compatible with a biological system such as a cell, cell culture, tissue, or organism.
  • the characteristics ofthe carrier will depend on the route of administration.
  • Physiologically and pharmaceutically acceptable carriers include diluents, fillers, salts, buffers, stabilizers, solubilizers, and other materials which are well known in the art.
  • the pharmaceutical composition ofthe invention may also contain other active factors and/or agents which inhibit prostate cell growth or increase cell death. Such additional factors and/or agents may be included in the pharmaceutical composition to produce a synergistic effect or to minimize side-effects caused.
  • the pharmaceutical composition ofthe invention may be in the form of a liposome in which the AR or ⁇ FGF antisense oligonucleotides are combined, in addition to other pharmaceutically acceptable carriers, with amphipathic agents such as lipids which exist in aggregated form as micelles, insoluble monolayers, liquid crystals, or lamellar layers which are in aqueous solution.
  • Suitable lipids for liposomal formulation include, without limitation, monoglycerides, diglycerides, sulfatides, lysolecithin, phospholipids, saponin, bile acids, and the like. Preparation of such liposomal formulations is within the level of skill in the art, as disclosed, for example, in U.S. Pat. No. 4,235,871 ; U.S. Pat. No. 4,501,728; U.S. Pat. No. 4,837,028; and U.S. Pat. No. 4,737,323.
  • the pharmaceutical composition ofthe invention may further include compounds such as cyclodextrins and the like which enhance delivery of oligonucleotides into cells.
  • cationic detergents e.g. Lipofectin
  • the oligonucleotides will be in the form of a tablet, capsule, powder, solution or elixir.
  • the pharmaceutical composition ofthe invention may additionally contain a solid carrier such as a gelatin or an adjuvant.
  • the tablet, capsule, and powder may contain from about 5 to 95% ofthe AR and/or ⁇ FGF antisense oligonucleotides and preferably from about 25 to 90% ofthe oligonucleotides.
  • a liquid carrier such as water, petroleum, oils of animal or plant origin such as peanut oil, mineral oil, soybean oil, sesame oil, or synthetic oils may be added.
  • the liquid form ofthe pharmaceutical composition may further contain physiological saline solution, dextrose or other saccharide solution, or glycols such as ethylene glycol, propylene glycol or polyethylene glycol.
  • the pharmaceutical composition may contain from about 0.5 to 90% by weight of an AR and or ⁇ FGF antisense oligonucleotide and preferably from about 1 to 50% ofthe oligonucleotide.
  • the oligonucleotides When a therapeutically effective amount of an AR or ⁇ FGF antisense oligonucleotide is administered by intravenous, cutaneous or subcutaneous injection, the oligonucleotides will be in the form of a pyrogen-free, parenterally acceptable aqueous solution.
  • parenterally acceptable solutions having due regard to pH, isotonicity, stability, and the like, is within the skill in the art.
  • a preferred pharmaceutical composition for intravenous, cutaneous, or subcutaneous injection should contain, in addition to the antisense oligonucleotides, an isotonic vehicle such as Sodium Chloride Injection, Ringer's Injection, Dextrose Injection, Dextrose and Sodium Chloride Injection, Lactated Ringer's Injection, or another vehicle as known in the art.
  • an isotonic vehicle such as Sodium Chloride Injection, Ringer's Injection, Dextrose Injection, Dextrose and Sodium Chloride Injection, Lactated Ringer's Injection, or another vehicle as known in the art.
  • the pharmaceutical composition ofthe present invention may also contain stabilizers, preservatives, buffers, antioxidants, or other additives known to those of skill in the art.
  • administration of the antisense oligonucleotides is localized to the region ofthe targeted cells in order to maximize the delivery ofthe antisense and to minimize the amount of antisense needed per treatment.
  • administration is by direct injection at or perfusion ofthe site ofthe targeted cells, such as a tumor.
  • the antisense oligonucleotides may be adhered to small particles (e.g., microscopic gold beads) which are impelled through the membranes of the target cells (see, e.g., U.S. Pat. No. 5,149,655).
  • a recombinant gene is constmcted which encodes an AR or ⁇ FGF antisense oligonucleotide and this gene is introduced within the targeted cells on a vector.
  • an AR or ⁇ FGF antisense gene may, for example, consist ofthe normal AR or ⁇ FGF sequence, or a subset ofthe normal sequences, operably joined in reverse orientation to a promoter region.
  • An operable antisense gene may be introduced on an integration vector or may be introduced on an expression vector. In order to be most effective, it is preferred that the antisense sequences be operably joined to a strong eukaryotic promoter which is inducible or constitutively expressed.
  • the AR and/or ⁇ FGF antisense oligonucleotides are administered in therapeutically effective amounts.
  • therapeutically effective amount means that amount of antisense which, under the conditions of administration, including mode of administration and presence of other active components, is sufficient to result in a meaningful patient benefit, i.e., the killing or inhibition ofthe growth of target cells.
  • the amount of AR and/or ⁇ FGF antisense oligonucleotides in the pharmaceutical composition ofthe present invention will depend not only upon the potency ofthe antisense but also upon the nature and severity ofthe condition being treated, and on the nature of prior treatments which the patient has undergone. Ultimately, the attending physician will decide the amount of antisense with which to treat each individual patient. Initially, the attending physician will administer low doses ofthe inhibitor and observe the patient's response. Larger doses of antisense may be administered until the optimal therapeutic effect is obtained for the patient. and at that point the dosage is not increased further. In preferred embodiments, it is contemplated that the various pharmaceutical compositions used to practice the method of the present invention should contain about 1.0 ⁇ g to about 100 mg of oligonucleotide per kg body weight.
  • the duration of intravenous therapy using the pharmaceutical compositions ofthe present invention will vary, depending on the severity ofthe disease being treated and the condition and potential idiosyncratic response of each individual patient. Because a bolus of oligonucleotides, particularly highly negatively-charged phosphorothioate modified oligonucleotides, may have adverse side effects (e.g., rapid lowering of blood pressure), slow intravenous administration is preferred. Thus, intravenous administration of therapeutically effective amounts over a 12-24 hour period are contemplated. Ultimately the attending physician will decide on the appropriate duration of intravenous therapy using the pharmaceutical composition ofthe present invention.
  • antisense oligonucleotides substantially complementary to subsets of SEQ ID NO.: 1, SEQ ID NO.: 2.
  • SEQ ID NO.: 3 or SEQ ID NO.: 4 and chemical modifications ofthe same which do not prevent hybridization under physiological conditions are contemplated as equivalents ofthe examples presented below.
  • the use of prostate specific antisense oligonucleotides is contemplated as a method of selectively inhibiting the growth of or killing prostatic cells.
  • antisense oligonucleotides to the estrogen receptor, PSA, probasin, telomerase, prohibitin, src, ras, myc, blc-2, protein kinase- A, pla ' sminogenctivator urokinase and methyl transferase genes is contemplated for the treatment of benign prostatic hype ⁇ lasia or prostatic cancer.
  • the PC3-1435 permanent cell line of human prostatic cancer obtained from the American Type Culture Collection, was grown in monolayer culture: The PC3-1435 cells are from an osseous metastasis and are androgen-insensitive. Cells were grown in Dulbecco's medium supplemented with 10 percent fetal calf semm, glutamate, pymvate, penicillin and streptomycin, in 25-150 cm flasks, incubated at 37°C in 6 percent CO 2 -air.
  • SEQ ID NO.: 5 is antisense to positions 927-953 ofthe AR gene (SEQ ID NO.: 1).
  • SEQ ID NO.: 6 is a self-stabilized or hai ⁇ in oligonucleotide. The first 21 bases are complementary to positions 916-936 ofthe AR gene. The remaining eight are identical to positions 920-927 ofthe gene, allowing formation of a 3' hai ⁇ in.
  • SEQ ID NO.: 7 is another self-stabilized antisense oligonucleotide.
  • SEQ ID NO.: 8 is an antisense sequence corresponding to positions 611-635 of the ⁇ FGF gene.
  • modified oligonucleotides were tested in which just the terminal two phosphodiester linkages at each end had been replaced by phosphorothioate synthetic linkages (shown as a subscript S between nucleotides in Table 1) and/or in which small organic chemical groups (e.g., 2-hydroxy-3-amino- propyl, propylamine) were added to the 3' terminal phosphate or the penultimate 3' phosphate.
  • small organic chemical groups e.g., 2-hydroxy-3-amino- propyl, propylamine
  • ADDRESSEE WOLF, GREENFIELD & SACKS, P.C.
  • B STREET: 600 ATLANTIC AVENUE
  • TAATAACTCA GTTCTTATTT GCACCTACTT CAGTGGACAC TGAATTTGGA AGGTGGAGGA 60 TTTTGTTTTT TTCTTTTAAG ATCTGGGCAT CTTTTGAATC TACCCTTCAA GTATTAAGAG 120
  • AAG CCC ATC TAT TTC CAC ACC CAG TGAAGCATTG GAAACCCTAT TTCCCCACCC 3149 Lys Pro He Tyr Phe His Thr Gin 915 920 CAGCTCATGC CCCCTTTCAG ATGTCTTCTG CCTGTTATAA CTCTGCACTA CTCCTCTGCA 3209
  • MOLECULE TYPE DNA (genomic)
  • CTCTATGGCA CCCCCCTTCC CTTTCTGACA TCTTCTGTAG TCAAGGTGGG AGGAAGGTGC 840
  • MOLECULE TYPE DNA (genomic)
  • CTGCCGGGTC CTATCGGCAA AAGCGTAGTG TTTATTTACT TTTGCTCGTG TTATTTTTAT 180 TCCAGTTCAG CTGCAGCTCA GTGCGGAAAG CGTGGGGGAG GTGTATATAA AGAGTACCGA 240
  • GCTGACATGC TTCCAGACGT TGGCCAAGGT TTGAGGTTTC CAGAAATCTT GTTACATGGA 360
  • MOLECULE TYPE DNA (genomic)
  • TAGCAGACAC CAAATGAGGA ATGTTTGTTC CTGGAAAGGC TGGAGGAGAA CCATTACAAC 360

Abstract

Methods of selectively inhibiting the growth of or killing prostatic cells, using antisense oligonucleotides to prostate specific genes, are disclosed. The oligonucleotides may have natural nucleic acid structures or may be modified oligonucleotides with enhanced stability or tissue specific targeting. The prostate specific genes to which the antisense may be directed include the AR and the αFGF gene. Pharmaceutical compositions including such antisense oligonucleotides are also described for use in the methods. The methods and products are of particular utility in the treatment of benign prostatic hyperplasia or prostate cancer.

Description

ANTISENSE OLIGONUCLEOTIDE CHEMOTHERAPY FOR BENIGN HYPERPLASIA OR CANCER OF THE PROSTATE
Field of the Invention The present invention relates to the field of chemotherapy for hyperplasias and cancers and, in particular, to chemotherapy for benign hyperplasia or cancer ofthe prostate. In addition, the invention relates to the field of antisense oligonucleotides and their use in human hyperplasia and cancer therapy.
Background of the Invention
Treatment of carcinoma ofthe prostate was one ofthe first successes of cancer chemotherapy, using the therapeutic program of castration and/or anti-androgen hormonal treatments introduced by Charles Huggins in the 1940s. A remarkable relief of symptoms and objective regression of bony metastases occurs under this endocrine therapeutic program. Unfortunately, after a "golden period" which lasts roughly 18 months, regrowth of the prostate cancer cells occurs and, in the later stages ofthe disease, sensitivity to and repression by anti- androgen hormonal therapy ceases. The conventional regimen of combined chemotherapeutic agents also is typically ineffective after the golden period, and a downhill clinical course follows, terminating in death. A key problem had been the silent onset of cancer ofthe prostate, with growth beyond its capsule and metastasis to bone too frequently occurring before the first visit to a physician. During the last half dozen years, there has been increasing recognition ofthe importance of early diagnosis and significant improvements in the available tests. As a consequence of early diagnosis, detection of prostatic cancer still contained within its capsule has become more frequent. For this situation, radical prostatectomy has largely supplanted the traditional castration/estrogen therapy. Radiation targeted to the prostate itself and to any proximal capsular infiltration has also become a prominent modality of therapy. When these two therapeutic approaches fail to halt progression ofthe disease, which is all too often (see, e.g., Gittes (1991); and Catalona (1994)), the prospect of benefit from available chemotherapy is gloomy. Less severe but more common than prostatic cancer is benign prostatic hyperplasia
(BPH). This condition may be a precursor to full blown prostatic cancer or may continue for decades without evolving into the deadly carcinoma. Depending upon the degree of hypertrophy and the age ofthe patient, treatment may range from "watchful waiting" to more aggressive approaches employing anti-androgen hormonal therapy, transurethral resection, or radical prostatectomy (see, e.g., Catalona (1994)).
The androgen receptor (AR) binds the male hormone testosterone and, acting at the transcriptional level, regulates the growth of normal prostatic cells. A cDNA for the human AR was disclosed by Lubahn et al. (1988). As noted above, anti-androgen or estrogen hormonal therapy, including physical or chemical castration, may be effective against early stage prostate cancer but, after a period of roughly 18 months, the patient becomes refractory to the hormonal therapy. The relapse is believed to be the result ofthe development or clonal selection of androgen-independent tumor cells in which the AR has mutated or been lost (see, e.g., Taplin, et al. (1995); Klocker, et al. (1994). Interestingly, in murine androgen-independent prostatic cancer cells, transfection with an AR cDNA has been shown to inhibit growth in the presence of testosterone (Suzuki, et al. (1994)).
The acidic fibroblast growth factor (αFGF), also known as the heparin binding growth factor type one (HBGF-1), is an androgen-regulated mitogen produced by prostatic cells. An mRNA sequence for a human allele of αFGF was disclosed in Harris, et al. (1991). Mansson. et al. (1989) found that αFGF was expressed in normal immature rat prostate but not in normal mature rat prostate. In cancerous rat prostatic cell lines, they found αFGF expression similar to that in immature rat prostate.
Summary of the Invention The present invention provides methods for treating a patient diagnosed as having benign prostatic hypeφlasia or a prostatic cancer. The methods include administering to the patient a therapeutically effective amount of a composition comprising an antisense oligonucleotide which selectively hybridizes to an AR or αFGF gene or mRNA sequence ofthe patient, thereby inhibiting the expression ofthe AR or αFGF gene or mRNA sequence. This inhibition of the AR or αFGF genes or mRNAs by antisense oligonucleotides results in a significant inhibition ofthe growth or survival of prostatic cells. As a result, the methods provide a useful new means of treating benign prostatic hypeφlasia and prostatic cancer. The methods are particularly useful in treating prostate cancer patients who have become refractory to anti-androgen hormonal therapy. The AR antisense oligonucleotides may comprise at least 10 consecutive bases from SEQ ID NO.: 1, at least 10 consecutive bases from a genomic sequence corresponding to SEQ ID NO.: 1, or oligonucleotides that hybridize to the complements of these sequences under physiological conditions. More preferably, the antisense oligonucleotides comprise at least 15 consecutive bases, and most preferably, 20-30 consecutive bases from the above-described sequences.
The αFGF antisense oligonucleotides may comprise at least 10 consecutive bases from any one of SEQ ID NO.: 2, SEQ ID NO.: 3 or SEQ ID NO.: 4, at least 10 consecutive bases from the joined exons of SEQ ID NO.: 2, SEQ ID NO.: 3 and SEQ ID NO.: 4; or oligonucleotides that hybridize to the complements of these sequences under physiological conditions. More preferably, the antisense oligonucleotides comprise at least 15 consecutive bases, and most preferably, 20-30 consecutive bases from the above-described sequences.
Examples of sequences ofthe invention include, but are not limited to, those disclosed as SEQ ID NO.: 5, SEQ ID NO.: 6, SEQ ID NO.: 7, and SEQ ID NO.: 8.
In preferred embodiments, all ofthe above-described oligonucleotides are modified oligonucleotides. In one set of embodiments, the modified oligonucleotide includes at least one synthetic internucleoside linkage such as a phosphorothioate, alkylphosphonate, phosphorodithioate, phosphate ester, alkylphosphonothioate, phosphoramidate, carbamate, carbonate, phosphate triester, acetamidate, or carboxymethyl ester.
In other embodiments with modified oligonucleotides, the modified oligonucleotide has at least one low molecular weight organic group covalently bound to a phosphate group of said oligonucleotide. In another set of embodiments, the modified oligonucleotide has at least one low molecular weight organic group covalently bound to a 2' position of a ribose of said oligonucleotide. Such low molecular weight organic groups include lower alkyl chains or aliphatic groups (e.g., methyl, ethyl, propyl, butyl), substituted alkyl and aliphatic groups (e.g., aminoethyl, aminopropyl, aminohydroxyethyl, aminohydroxypropyl), small saccharides or glycosyl groups.
In another set of embodiments the modified oligonucleotide has covalently attached thereto a prostate-targeting compound such as an androgen, androgen derivative, estrogen, estrogen derivative, estramustine, emcyt or estracyt. In preferred embodiments, the antisense oligonucleotides are administered intravenously at a dosage between 1.0 μg and 100 mg per kg body weight of the patient. The present invention also provides for any or all ofthe above-described antisense oligonucleotides, including the various modified oligonucleotides, in a pharmaceutical composition. The antisense oligonucleotides are admixed with a sterile pharmaceutically acceptable carrier in a therapeutically effective amount such that the isolated antisense oligonucleotide selectively hybridizes to the AR or αFGF gene or mRNA sequence when administered to a patient. A pharmaceutical kit is also provided in which such a pharmaceutical composition is combined with a pharmaceutically acceptable carrier for intravenous administration.
The methods and products ofthe present invention further include antisense oligonucleotides, as described above, directed at a PSA gene, a probasin gene, an estrogen receptor gene, a telomerase gene, a prohibitin gene, a src gene, a ras gene, a myc gene, a blc-2 gene, a protein kinase-A gene, a plasminogen activator urokinase gene and a methyl transferase gene.
Detailed Description of the Invention
The present invention provides new methods for the treatment of cancer ofthe prostate and pharmaceutical compositions useful therefor. It is now disclosed that antisense oligonucleotides complementary to genes which are expressed predominantly or strongly in prostatic cells are effective for inhibiting the growth of and/or killing hypeφlastic or cancerous cells of prostatic origin. In particular, the present invention provides oligonucleotides, including modified oligonucleotides. which have antisense homology to a sufficient portion of either the AR or αFGF gene such that they inhibit the expression of that gene. Suφrisingly, inhibition of either of these genes, even in androgen-resistant prostatic cancer cells, inhibits the growth of these cells. Because the antisense oligonucleotides ofthe invention can be administered systemically but selectively inhibit prostate cells, the present invention has particular utility in late stage prostate cancer which has metastasized.
Definitions In order to describe more clearly and concisely the subject matter ofthe present invention, the following definitions are provided for specific terms used in the claims appended hereto:
AR. As used herein, the abbreviation "AR" refers to the androgen receptor well known in the art and described in the various references cited herein. A cDNA sequence ofthe human AR gene was disclosed in Lubahn et al. (1988). The Lubahn et al. (1988)sequence is available on GenBank (Accession number J03180) and is reproduced here as SEQ. ID NO.: 1. The translation initiation codon of this gene is found at base positions 363-365 and the stop codon is at positions 3120-3122 of SEQ ID NO. : 1. As will be obvious to one of ordinary skill in the art. other alleles ofthe AR gene, including other human alleles and homologues from other mammalian species, encoding an AR protein and hybridizing to SEQ ID NO.: 1 under stringent hybridization conditions, will exist in natural populations and are embraced by the term "AR gene" as used herein. αFGF. As used herein, the term "αFGF" refers to the αFGF protein known in the art and described in the various references cited herein. The genomic DNA of one allele ofthe human αFGF gene has been partially sequenced and was disclosed in Wang et al. (1989). The Wang et al.(1989) sequences cover the three exons ofthe αFGF gene as well as some 5', 3' and intron sequences. These sequences are available on GenBank (Accession numbers M23017, M23086 and M23087) and are reproduced here as SEQ. ID NO.: 2, SEQ ID NO.: 3 and SEQ ID NO.: 4. A partial cDNA sequence for a human αFGF gene also may be found in Harris et al. (1991). The locations ofthe exons are located in the sequence listings. The translation initiation codon is found at positions 602-604 of SEQ ID NO.: 2 and the stop codon is found at positions 496-498. In addition, as will be obvious to one of ordinary skill in the art, other alleles ofthe αFGF gene, including other human alleles and homologues from other mammalian species, encoding an αFGF protein and hybridizing to one or more of SEQ ID NO.: 2, SEQ ID NO.: 3 or SEQ ID NO.: 4 under stringent hybridization conditions, will exist in natural populations and are embraced by the term "αFGF gene" as used herein.
Antisense Oligonucleotides. As used herein, the term "antisense oligonucleotide" or "antisense" describes an oligonucleotide that is an oligoribonucleotide, oligodeoxyribonucleotide, modified oligoribonucleotide, or modified oligodeoxyribonucleotide which hybridizes under physiological conditions to DNA comprising a particular gene or to an mRNA transcript of that gene and, thereby, inhibits the transcription of that gene and/or the translation of that mRNA. In particular, by an "AR-antisense oligonucleotide" and by an "αFGF-antisense oligonucleotide" are meant oligonucleotides which hybridize under physiological conditions to the AR gene/mRNA or αFGF gene/mRNA and, thereby, inhibit transcription/translation ofthe AR and αFGF genes/mRNAs, respectively. The antisense molecules are designed so as to interfere with transcription or translation of AR or αFGF upon hybridization with the target. Those skilled in the art will recognize that the exact length ofthe antisense oligonucleotide and its degree of complementarity will depend upon the specific target selected, including the sequence of the target and the particular bases which comprise that sequence. It is preferred that the antisense oligonucleotide be selected so as to hybridize selectively with the target under physiological conditions, i.e., to hybridize substantially more to the target sequence than to any other sequence in the target cell under physiological conditions. Stringent hybridization conditions. As used herein, the term "stringent hybridization conditions" means hybridization conditions from 30°C-60°C and from 5x to 0.1 x SSC. Highly stringent hybridization conditions are at 45 °C and O.lx SSC. "Stringent hybridization conditions" is a term of art understood by those of ordinary skill in the art. For any given nucleic acid sequence, stringent hybridization conditions are those conditions of temperature and buffer solution which will permit hybridization of that nucleic acid sequence to its complementary sequence and not to substantially different sequences. The exact conditions which constitute "stringent" conditions, depend upon the length ofthe nucleic acid sequence and the frequency of occurrence of subsets of that sequence within other non-identical sequences. By varying hybridization conditions from a level of stringency at which no hybridization occurs to a level at which hybridization is first observed, one of ordinary skill in the art can, without undue experimentation, determine conditions which will allow a given sequence to hybridize only with identical sequences. Suitable ranges of such stringency conditions are described in Krause. M.H.. and S.A. Aaronson, Methods in Enzymology. 200:546-556 (1991). As used herein with respect to in vivo hybridization conditions, the term "physiological conditions" is considered functionally equivalent to the in vitro stringent hybridization conditions. I. Design of AR and αFGF Antisense Oligonucleotides
The present invention depends, in part, upon the discovery that the selective inhibition of the expression of AR or αFGF by antisense oligonucleotides in prostatic cells effectively inhibits cell growth and/or causes cell death.
Based upon SEQ ID NO.: 1, SEQ ID NO.: 2, SEQ ID NO.: 3 and SEQ ID NO.: 4, or upon allelic or homologous genomic or cDNA sequences, one of skill in the art can easily choose and synthesize any of a number of appropriate antisense molecules for use in accordance with the present invention. In order to be sufficiently selective and potent for AR or αFGF inhibition, such antisense oligonucleotides should comprise at least 10 and, more preferably, at least 15 consecutive bases which are complementary to the AR or αFGF mRNA transcripts. Most preferably, the antisense oligonucleotides comprise a complementary sequence of 20-30 bases. Although oligonucleotides may be chosen which are antisense to any region of the AR or αFGF genes or mRNA transcripts, in preferred embodiments the antisense oligonucleotides correspond to N-terminal or 5' upstream sites such as translation initiation, transcription initiation or promoter sites. In addition, 3'-untranslated regions or telomerase sites may be targeted. Targeting to mRNA splicing sites has also been used in the art but may be less preferred if alternative mRNA splicing occurs. In addition, the AR or αFGF antisense is, preferably, targeted to sites in which mRNA secondary structure is not expected (see, e.g., Sainio et al. (1994)) and at which proteins are not expected to bind. Finally, although, SEQ ID NO.: 1 discloses a cDNA sequence and SEQ ID NO.: 2, SEQ ID NO.:3 and SEQ ID NO.: 4 disclose genomic DNA sequences, one of ordinary skill in the art may easily derive the genomic DNA corresponding to the cDNA of SEQ ID NO.: 1 and may easily obtain the cDNA sequence corresponding to SEQ ID NO.: 2, SEQ ID NO.:3 and SEQ ID NO.: 4. Thus, the present invention also provides for antisense oligonucleotides which are complementary to the genomic DNA corresponding to SEQ ID NO.: 1 and the cDNA corresponding to SEQ ID NO.: 2, SEQ ID NO.: 3 and SEQ ID NO.: 4. Similarly, antisense to allelic or homologous cDNAs and genomic DNAs are enabled without undue experimentation.
As will be understood by one of ordinary skill in the art, the antisense oligonucleotides of the present invention need not be perfectly complementary to the AR or αFGF genes or mRNA transcripts in order to be effective. Rather, some degree of mismatches will be acceptable if the antisense oligonucleotide is of sufficient length. In all cases, however, the oligonucleotides should have sufficient length and complementarity so as to hybridize to an AR or αFGF transcript under physiological conditions. Preferably, of course, mismatches are absent or minimal. In addition, although it is not recommended, the antisense oligonucleotides may have one or more non-complementary sequences of bases inserted into an otherwise complementary antisense oligonucleotide sequence. Such non-complementary sequences may "loop" out of a duplex formed by an AR or αFGF transcript and the bases flanking the non-complementary region. Therefore, the entire oligonucleotide may retain an inhibitory effect despite an apparently low percentage of complementarity. Of particular importance in this respect is the use of self-stabilized or haiφin oligonucleotides. Such oligonucleotides, or modified oligonucleotides, have a sequence at the 5' and/or 3' end which is capable of folding over and forming a duplex with itself. The duplex region, which is preferably at least 4-6 bases joined by a loop of 3-6 bases, stabilizes the oligonucleotide against degradation. These self-stabilized oligonucleotides are easily designed by adding the inverted complement of a 5 ' or 3' AR or αFGF sequence to the end of the oligonucleotide (see, e.g., Table 1, SEQ ID NO.: 6 and SEQ ID NO.: 7; Tang, J.-Y., et al. (1993) Nucleic Acids Res. 21 :2729-2735).
In one set of embodiments, the AR and αFGF antisense oligonucleotides ofthe invention may be composed of "natural" deoxyribonucleotides, ribonucleotides, or any combination thereof. That is, the 5' end of one nucleotide and the 3' end of another nucleotide may be covalently linked, as in natural systems, via a phosphodiester intemucleoside linkage. These oligonucleotides may be prepared by art recognized methods which may be carried out manually or by an automated synthesizer. In preferred embodiments, however, the antisense oligonucleotides ofthe invention also may include "modified" oligonucleotides. That is, the oligonucleotides may be modified in a number of ways which do not prevent them from hybridizing to their target but which enhance their stability or targeting to prostatic cells or which otherwise enhance their therapeutic effectiveness. The term "modified oligonucleotide" as used herein describes an oligonucleotide in which (1) at least two of its nucleotides are covalently linked via a synthetic intemucleoside linkage (i.e., a linkage other than a phosphodiester linkage between the 5' end of one nucleotide and the 3' end of another nucleotide) and/or (2) a chemical group not normally associated with nucleic acids has been covalently attached to the oligonucleotide.
Preferred synthetic intemucleoside linkages are phosphorothioates, alkylphosphonates, phosphorodithioates, phosphate esters, alkylphosphonothioates, phosphoramidates, carbamates, carbonates, phosphate triesters, acetamidate, and carboxymethyl esters. Further, one or more of the 5'-3' phosphate group may be covalently joined to a low molecular weight (e.g., 15-500 Da) organic group. Such low molecular weight organic groups include lower alkyl chains or aliphatic groups (e.g., methyl, ethyl, propyl, butyl), substituted alkyl and aliphatic groups (e.g., aminoethyl, aminopropyl, aminohydroxyethyl, aminohydroxypropyl), small saccharides or glycosyl groups. Other low molecular weight organic modifications include additions to the intemucleoside phosphate linkages such as cholesteryl or diamine compounds with varying numbers of carbon residues between the amino groups and terminal ribose. Oligonucleotides with these linkages or other modifications can be prepared according to known methods (see, e.g., Agrawal and Goodchild (1987); Agrawal et al. (1988); Uhlmann et al. (1990); Agrawal et al. (1992); Agrawal (1993); and U.S. Pat. No. 5,149,798).
The term "modified oligonucleotide" also encompasses oligonucleotides with a covalently modified base and/or sugar. For example, modified oligonucleotides include oligonucleotides having backbone sugars which are covalently attached to low molecular weight organic groups other than a hydroxyl group at the 3' position and other than a phosphate group at the 5' position. Thus modified oligonucleotides may include a 2'-O-alkylated ribose group such as a 2'-O-methylated ribose. In addition, modified oligonucleotides may include sugars such as arabinose instead of ribose. Altematively, the modified oligonucleotides may be branched oligonucleotides. Unoxidized or partially oxidized oligonucleotides having a substitution in one or more nonbridging oxygen per nucleotide in the molecule are also considered to be modified oligonucleotides.
Also considered as modified oligonucleotides are oligonucleotides having prostate- targeting, nuclease resistance-conferring, or other bulky substituents and/or various other stmctural modifications not found in vivo without human intervention. The androgen receptor and other hormonal receptor sites on prostate cells allow for targeting antisense oligonucleotides specifically or particularly to prostatic cells. Attachment ofthe antisense oligonucleotides by a molecular "tether" (e.g., an alkyl chain) to estramustine, emcyt or estracyt (Sheridan and Tew (1991)), for example, may provide prostatic targeting and the possibility of covalent alkylation of host prostatic DNA. Estramustine targets particularly to the ventral prostate (Forsgren, et al. (1979)). Similarly, one may covalently attach androgen, estrogen, androgen or estrogen derivatives, or other prostate cell ligands to antisense oligonucleotides using tethers and conjugating linkages for prostatic targeting. Finally, one may of course covalently attach other chemotherapeutic agents (e.g., dexamethasone, vinblastine, etoposide) to the antisense oligonucleotides for enhanced effect.
The most preferred modified oligonucleotides are hybrid or chimeric oligonucleotides in which some but not all ofthe phosphodiester linkages, bases or sugars have been modified. Hybrid modified antisense oligonucleotides may be composed, for example, of stretches of ten 2'-O-alkyl nucleotides or ten phosphorothioate synthetic linkages at the 5' and/or 3' ends, and a segment of seven unmodified oligodeoxynucleotides in the center, or of similar terminal segments of alkyl phosphonates, with central P=S or P=O oligonucleotides (Agrawal, et al. (1990); Metelev, et al. (1994)). The currently most preferred modified oligonucleotides are 2'-O- methylated hybrid oligonucleotides. Since degradation occurs mainly at the 3' end, secondarily at the 5' end, and less in the middle, unmodified oligonucleotides located at this position can activate RNase H, and yet are degraded slowly. Furthermore, the Tm of such a 27-mer is approximately 20 °C higher than that of a 27-mer all phosphorothioate oligodeoxynucleotide. This greater affinity for the targeted genomic area can result in greater inhibiting efficacy. Obviously, the number of synthetic linkages at the termini need not be ten and synthetic linkages may be combined with other modifications, such as alkylation of a 5' or 3' phosphate, or 2'-O- alkylation. Thus, merely as another example, one may produce a modified oligonucleotide with the following stmcture, where B represents any base, R is an alkyl, aliphatic or other substituent, the subscript S represents a synthetic (e.g. phosphorothioate) linkage, and each n is an independently chosen integer from 1 to about 20:
OH
5(Bs)nBBBB- ... -BBBB(BS)„B— P=O3
I O— R
II. Products and Methods of Treatment for BPH and Prostate Cancer
The methods ofthe present invention represent new and useful additions to the field of benign prostate hypeφlasia or prostate cancer therapy. In particular, the methods ofthe present invention are especially useful for late stage prostate cancer in which metastases have occurred and in which the cells have become resistant to estrogen or anti-androgen therapy. The methods may, however, also be used in benign prostate hypeφlasia or early stage prostate cancer and may provide a substitute for more radical procedures such as transurethral resection, radical prostatectomy, or physical or chemical castration. The products of the present invention include the isolated antisense oligonucleotides described above. As used herein, the term "isolated" as applied to an antisense oligonucleotide means not covalently bound to and physically separated from the 5' and 3' sequences which flank the corresponding antisense sequence in nature.
Administration ofthe AR or αFGF antisense oligonucleotides may be oral, intravenous,
77454.1 parenteral, cutaneous or subcutaneous. For BPH or when the site of a prostatic tumor is known, the administration also may be localized to the prostate or to the region ofthe tumor by injection to or perfusion ofthe site.
AR or αFGF antisense oligonucleotides may be administered as part of a pharmaceutical composition. Such a pharmaceutical composition may include the antisense oligonucleotides in combination with any standard physiologically and/or pharmaceutically acceptable carriers which are known in the art. The compositions should be sterile and contain a therapeutically effective amount ofthe antisense oligonucleotides in a unit of weight or volume suitable for administration to a patient. The term "pharmaceutically acceptable" means a non-toxic material that does not interfere with the effectiveness ofthe biological activity ofthe active ingredients. The term "physiologically acceptable" refers to a non-toxic material that is compatible with a biological system such as a cell, cell culture, tissue, or organism. The characteristics ofthe carrier will depend on the route of administration. Physiologically and pharmaceutically acceptable carriers include diluents, fillers, salts, buffers, stabilizers, solubilizers, and other materials which are well known in the art. The pharmaceutical composition ofthe invention may also contain other active factors and/or agents which inhibit prostate cell growth or increase cell death. Such additional factors and/or agents may be included in the pharmaceutical composition to produce a synergistic effect or to minimize side-effects caused.
The pharmaceutical composition ofthe invention may be in the form of a liposome in which the AR or αFGF antisense oligonucleotides are combined, in addition to other pharmaceutically acceptable carriers, with amphipathic agents such as lipids which exist in aggregated form as micelles, insoluble monolayers, liquid crystals, or lamellar layers which are in aqueous solution. Suitable lipids for liposomal formulation include, without limitation, monoglycerides, diglycerides, sulfatides, lysolecithin, phospholipids, saponin, bile acids, and the like. Preparation of such liposomal formulations is within the level of skill in the art, as disclosed, for example, in U.S. Pat. No. 4,235,871 ; U.S. Pat. No. 4,501,728; U.S. Pat. No. 4,837,028; and U.S. Pat. No. 4,737,323.
The pharmaceutical composition ofthe invention may further include compounds such as cyclodextrins and the like which enhance delivery of oligonucleotides into cells. When the composition is not administered systemically but, rather, is injected at the site ofthe target cells, cationic detergents (e.g. Lipofectin) may be added to enhance uptake. When a therapeutically effective amount of AR or αFGF antisense oligonucleotides is administered orally, the oligonucleotides will be in the form of a tablet, capsule, powder, solution or elixir. When administered in tablet form, the pharmaceutical composition ofthe invention may additionally contain a solid carrier such as a gelatin or an adjuvant. The tablet, capsule, and powder may contain from about 5 to 95% ofthe AR and/or αFGF antisense oligonucleotides and preferably from about 25 to 90% ofthe oligonucleotides. When administered in liquid form, a liquid carrier such as water, petroleum, oils of animal or plant origin such as peanut oil, mineral oil, soybean oil, sesame oil, or synthetic oils may be added. The liquid form ofthe pharmaceutical composition may further contain physiological saline solution, dextrose or other saccharide solution, or glycols such as ethylene glycol, propylene glycol or polyethylene glycol. When administered in liquid form, the pharmaceutical composition may contain from about 0.5 to 90% by weight of an AR and or αFGF antisense oligonucleotide and preferably from about 1 to 50% ofthe oligonucleotide.
When a therapeutically effective amount of an AR or αFGF antisense oligonucleotide is administered by intravenous, cutaneous or subcutaneous injection, the oligonucleotides will be in the form of a pyrogen-free, parenterally acceptable aqueous solution. The preparation of such parenterally acceptable solutions, having due regard to pH, isotonicity, stability, and the like, is within the skill in the art. A preferred pharmaceutical composition for intravenous, cutaneous, or subcutaneous injection should contain, in addition to the antisense oligonucleotides, an isotonic vehicle such as Sodium Chloride Injection, Ringer's Injection, Dextrose Injection, Dextrose and Sodium Chloride Injection, Lactated Ringer's Injection, or another vehicle as known in the art. The pharmaceutical composition ofthe present invention may also contain stabilizers, preservatives, buffers, antioxidants, or other additives known to those of skill in the art.
In preferred embodiments, when the target cells are readily accessible, administration of the antisense oligonucleotides is localized to the region ofthe targeted cells in order to maximize the delivery ofthe antisense and to minimize the amount of antisense needed per treatment. Thus, in one preferred embodiment, administration is by direct injection at or perfusion ofthe site ofthe targeted cells, such as a tumor. Altematively, the antisense oligonucleotides may be adhered to small particles (e.g., microscopic gold beads) which are impelled through the membranes of the target cells (see, e.g., U.S. Pat. No. 5,149,655).
In another series of embodiments, a recombinant gene is constmcted which encodes an AR or αFGF antisense oligonucleotide and this gene is introduced within the targeted cells on a vector. Such an AR or αFGF antisense gene may, for example, consist ofthe normal AR or αFGF sequence, or a subset ofthe normal sequences, operably joined in reverse orientation to a promoter region. An operable antisense gene may be introduced on an integration vector or may be introduced on an expression vector. In order to be most effective, it is preferred that the antisense sequences be operably joined to a strong eukaryotic promoter which is inducible or constitutively expressed.
In all ofthe above-described methods of treatment, the AR and/or αFGF antisense oligonucleotides are administered in therapeutically effective amounts. As used herein, the term "therapeutically effective amount" means that amount of antisense which, under the conditions of administration, including mode of administration and presence of other active components, is sufficient to result in a meaningful patient benefit, i.e., the killing or inhibition ofthe growth of target cells.
The amount of AR and/or αFGF antisense oligonucleotides in the pharmaceutical composition ofthe present invention will depend not only upon the potency ofthe antisense but also upon the nature and severity ofthe condition being treated, and on the nature of prior treatments which the patient has undergone. Ultimately, the attending physician will decide the amount of antisense with which to treat each individual patient. Initially, the attending physician will administer low doses ofthe inhibitor and observe the patient's response. Larger doses of antisense may be administered until the optimal therapeutic effect is obtained for the patient. and at that point the dosage is not increased further. In preferred embodiments, it is contemplated that the various pharmaceutical compositions used to practice the method of the present invention should contain about 1.0 μg to about 100 mg of oligonucleotide per kg body weight.
The duration of intravenous therapy using the pharmaceutical compositions ofthe present invention will vary, depending on the severity ofthe disease being treated and the condition and potential idiosyncratic response of each individual patient. Because a bolus of oligonucleotides, particularly highly negatively-charged phosphorothioate modified oligonucleotides, may have adverse side effects (e.g., rapid lowering of blood pressure), slow intravenous administration is preferred. Thus, intravenous administration of therapeutically effective amounts over a 12-24 hour period are contemplated. Ultimately the attending physician will decide on the appropriate duration of intravenous therapy using the pharmaceutical composition ofthe present invention.
77454 ! The following examples ofthe use of AR and αFGF antisense are presented merely to illustrate some ofthe oligonucleotides, including modified oligonucleotides, that may be employed according to the present invention. The particular oligonucleotides used, therefore, should not be constmed as limiting ofthe invention but, rather, as indicative ofthe wide range of oligonucleotides which may be employed. As will be obvious to one of ordinary skill in the art in light ofthe present disclosure, a great many equivalents to the presently disclosed antisense oligonucleotides and disclosed methods are now available. In particular, other antisense oligonucleotides substantially complementary to subsets of SEQ ID NO.: 1, SEQ ID NO.: 2. SEQ ID NO.: 3 or SEQ ID NO.: 4 and chemical modifications ofthe same which do not prevent hybridization under physiological conditions, are contemplated as equivalents ofthe examples presented below. In general, the use of prostate specific antisense oligonucleotides is contemplated as a method of selectively inhibiting the growth of or killing prostatic cells. In particular, the use of antisense oligonucleotides to the estrogen receptor, PSA, probasin, telomerase, prohibitin, src, ras, myc, blc-2, protein kinase- A, pla'sminogenctivator urokinase and methyl transferase genes is contemplated for the treatment of benign prostatic hypeφlasia or prostatic cancer.
Experimental Examples
The PC3-1435 permanent cell line of human prostatic cancer, obtained from the American Type Culture Collection, was grown in monolayer culture: The PC3-1435 cells are from an osseous metastasis and are androgen-insensitive. Cells were grown in Dulbecco's medium supplemented with 10 percent fetal calf semm, glutamate, pymvate, penicillin and streptomycin, in 25-150 cm flasks, incubated at 37°C in 6 percent CO2-air.
A number of AR and αFGF antisense oligonucleotides were tested for their inhibitory effect on prostatic cells. The base sequences of these oligonucleotides are disclosed as SEQ ID NO.: 5 through SEQ ID NO.: 8. SEQ ID NO.: 5 is antisense to positions 927-953 ofthe AR gene (SEQ ID NO.: 1). SEQ ID NO.: 6 is a self-stabilized or haiφin oligonucleotide. The first 21 bases are complementary to positions 916-936 ofthe AR gene. The remaining eight are identical to positions 920-927 ofthe gene, allowing formation of a 3' haiφin. SEQ ID NO.: 7 is another self-stabilized antisense oligonucleotide. The first 21 bases of this oligonucleotide are complementary to positions 927-947 ofthe AR gene. The remaining eight are identical to positions 931-938 ofthe gene, allowing for formation of a 3' haiφin. Finally, SEQ ID NO.: 8 is an antisense sequence corresponding to positions 611-635 of the αFGF gene.
Table 1 shows some ofthe antisense oligonucleotides tested. The numbers at the left of each sequence correspond to the sequence numbers in the sequence listing. Antisense oligonucleotides with unmodified or natural intemucleoside linkages (P=O) and oligonucleotides with all phosphorothioate synthetic linkages (P=S) were tested. In addition, modified oligonucleotides were tested in which just the terminal two phosphodiester linkages at each end had been replaced by phosphorothioate synthetic linkages (shown as a subscript S between nucleotides in Table 1) and/or in which small organic chemical groups (e.g., 2-hydroxy-3-amino- propyl, propylamine) were added to the 3' terminal phosphate or the penultimate 3' phosphate.
Growth ofthe PC3-1435 cell line in tissue culture monolayers was consistently inhibited by addition of phosphorothioate-modified oligodeoxynucleotides targeted against the AR or αFGF genes and incubation for 24-48 hours thereafter. As the concentration of modified oligonucleotides is decreased from the 10-20 μM level, most effective inhibition occurs with specific antisense oligodeoxynucleotides at the 2-5 μM level, as contrasted with mismatched oligodeoxynucleotides (see Tables 2 and 3).
While the effects on cell growth (i.e. cell numbers) are readily manifest, visual substage microscopy of wells revealed additional features ofthe inhibition events using AR antisense oligonucleotides against PC3-1435 cells. The first evidence of antisense inhibition is pture of the monolayer fabric. The stellate cells in a confluent culture lose contact with their neighbors, round up individually or in clumps, become pyknotic, and cease growing, as examined on successive days. There is an early loss of adhesiveness to the floor ofthe plastic wells. These changes are more severe (see Table 4) than those measured by 3H-thymidine incoφoration into DNA, in other words more drastic than the impairment of DNA synthesis. Each ofthe above-mentioned references and patents are incoφorated by reference.
TABLE 1 Antisense Oligonucleotides
Sequence Target
#5 5'CTG-CTG-CTG-TTG-CTG-AAG-GAG-TTG-CAT3' Androgen receptor,
P=S
#5 5' CTG -CTG -CTG -TTG -CTG -AAG -GAG -TTG -CAT3' Androgen receptor,
P=0
#5 5 ' CSTSG - CTG - CTG - TTG - CTG - AAG - GAG - TTG - CSAST3 ' Androgen receptor,
P=S termini #5 5'CTG-CTG-CTG-TTG-CTG-AAG-GAG-TTG-CAT3' Androgen receptor, modified with organic group
H3N-CH2CHCH20-P=0
OH OH
0 Androgen receptor,
1 modified with #5 5'CTG-CTG-CTG-TTG-CTG-AAG-GAG-TTG-CA-0-P-0-T3' organic group
CH,CH,CH,NH
#6 5'GGA-GTT-GCA-TGG-TGC-TGG-CCT-CAG-CAC-CA3' Androgen receptor
3' hairpin, P=S
#7 5'CTG-TTG-CTG-AAG-GAG-TTG-CAT-AAC-TCC-TT3' Androgen receptor
31 hairpin, P=S
#8 5'GGG-CTG-TGA-AGG-TGG-TGA-TTT-CCC-C3' αFGF, P=S #8 5 ' GGG- CTG- TGA-AGG-TGG-TGA- TTT - CCC- C3 αFGF , P=0
TABLE 2
3H-thymidine incorporation into DNA PC3-1435 human prostate cancer tissue culture
Genes Targeted Concentration (μM) CPMt % inhibition
Control (no oligo) — 38,000 0
Androgen receptor, (P = S) 20 15,000 60
5 20,000 48
Androgen receptor, (P = S)* 20 10,200 68
5 24,000 25
Mismatch (P = S) 20 20,000 47
5 27,000 30 t Averages of 3 separate wells
* 3' phosphate modified with -CH2CHOHCHNH3 +
TABLE 3
Degree of inhibition of DNA synthesis in PC3-1435 prostate cancer tissue cultures
Genes targeted Concentration ( μM) CPM t % inhibition
Control (no oligo) — 14,700 0 αFGF (P=S) 20 2,485 83
5 4,500 69
Mismatch 20 6,990 51
5 10,750 27
t Averages of 3 separate wells. TABLE 4
Moφhological Comparison of Treated and Control Cells
Concentration μM
Gene Target 20 10 5 2 αFGF (P=S) 4+ 4+ 1-1/2+ 1 + Androgen receptor (P=S) 3+ 3+ 1 + 1 +
Mismatch (P=S) 1-1/2+ 1/2+ 0 0
Observation 24 hours after oligonucleotide addition. Damage: 4+ devastating; 3+ severe; 2+ serious; 1+ visible; 1/2+ slight; 0 none
References
Agrawal (ed.) Meth. Mol. Biol.. Humana Press. Totowa, NJ (1993) Vol. 20.
Agrawal and Goodchild (1987) Tetrahedron Lett. 28:3539-3542. Agrawal et al. (1988) Proc. Natl. Acad. Sci. (USA1 85:7079-7083.
Agrawal et al.(1990) Proc. Natl. Acad. Sci. (USA1 87: 1401-1405.
Agrawal et al. (1992) Trends Biotechnol. 10:152-158.
Catalona (1994) N.E. J. Med. 331 :996-1004.
Forsgren et al. (19791 Cancer Res. 39:5155-5164. Gittes (19911 N.E. J. Med. 324: 236-245.
Harris et al. (1991) in Mol. and Cell. Biol. of Prostate Cancer. Karr et al (eds.) Plenum Press,
NY, pp. 315-330.
Klocker, et al. (1994) The Prostate 25:266-273. Lubahn et al. (19881 Mol. Endocrinol. 2(121:1265-1275.
Mansson, et al. (1989) Cancer Res. 49:2485-2494.
Metelev et al. (1994) Bioorg. Medicinal Chem. Lett. 4: 2929-2934.
Sainio et al. (1994) Cell. Mol. Neurobiol. 14(5):439-457.
Sheridan and Tew (19911 Cancer Surveys 11:239-254. Suzuki, et al. (19941 The Prostate 25:310-319.
Taplin et al. (19951 N.E.J. Med. 332(211:1393-1398.
Uhlmann et al. (1990) Chem. Rev. 90:534-583.
Wang et al. (1979) Invest. Urol. 17:159-163.
Wang et al. (1989) Mol. Cell. Biol. 9(6):2387-2395.
SEQUENCE LISTING
(1) GENERAL INFORMATION:
(i) APPLICANT: WORCESTER FOUNDATION FOR BIOMEDICAL RESEARCH, INC.
(ii) TITLE OF INVENTION: ANTISENSE OLIGONUCLEOTIDE CHEMOTHERAPY FOR BENIGN HYPERPLASIA OR CANCER OF THE PROSTATE
(iii) NUMBER OF SEQUENCES: 8
(iv) CORRESPONDENCE ADDRESS:
(A) ADDRESSEE: WOLF, GREENFIELD & SACKS, P.C. (B) STREET: 600 ATLANTIC AVENUE
(C) CITY: BOSTON
(D) STATE: MA
(E) COUNTRY: USA
(F) ZIP: 02210
(v) COMPUTER READABLE FORM:
(A) MEDIUM TYPE: Floppy disk
(B) COMPUTER: IBM PC compatible
(C) OPERATING SYSTEM: PC-DOS/MS-DOS (D) SOFTWARE: Patentin Release #1.0, Version #1.25
(vi) CURRENT APPLICATION DATA:
(A) APPLICATION NUMBER:
(B) FILING DATE: (C) CLASSIFICATION: (viii) ATTORNEY/AGENT INFORMATION:
(A) NAME: TWOMEY, MICHAEL J.
(B) REGISTRATION NUMBER: 38,349
(C) REFERENCE/DOCKET NUMBER: W0461/7035
(ix) TELECOMMUNICATION INFORMATION:
(A) TELEPHONE: 617-720-3500
(B) TELEFAX: 617-720-2441
(2) INFORMATION FOR SEQ ID NO:l
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 3569 base pairs (B) TYPE: nucleic acid
(C) STRANDEDNESS: double
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: cDNA
(iii) HYPOTHETICAL: NO
(iv) ANTI-SENSE: NO
(vi) ORIGINAL SOURCE:
(A) ORGANISM: HOMO SAPIENS
(ix) FEATURE:
(A) NAME/KEY: CDS (B) LOCATION: 363..3122
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 1 :
TAATAACTCA GTTCTTATTT GCACCTACTT CAGTGGACAC TGAATTTGGA AGGTGGAGGA 60 TTTTGTTTTT TTCTTTTAAG ATCTGGGCAT CTTTTGAATC TACCCTTCAA GTATTAAGAG 120
ACAGACTGTG AGCCTAGCAG GGCAGATCTT GTCCACCGTG TGTCTTCTTC TGCACGAGAC 180
TTTGAGGCTG TCAGAGCGCT TTTTGCGTGG TTGCTCCCGC AAGTTTCCTT CTCTGGAGCT 240
TCCCGCAGGT GGGCAGCTAG CTGCAGCGAC TACCGCATCA TCACAGCCTG TTGAACTCTT 300
CTGAGCAAGA GAAGGGGAGG CGGGGTAAGG GAAGTAGGTG GAAGATTCAG CCAAGCTCAA 360
GG ATG GAA GTG CAG TTA GGG CTG GGA AGG GTC TAC CCT CGG CCG CCG 407 Met Glu Val Gin Leu Gly Leu Gly Arg Val Tyr Pro Arg Pro Pro 1 5 10 15
TCC AAG ACC TAC CGA GGA GCT TTC CAG AAT CTG TTC CAG AGC GTG CGC 455 Ser Lys Thr Tyr Arg Gly Ala Phe Gin Asn Leu Phe Gin Ser Val Arg
20 25 30
GAA GTG ATC CAG AAC CCG GGC CCC AGG CAC CCA GAG GCC GCG AGC GCA 503 Glu Val lie Gin Asn Pro Gly Pro Arg His Pro Glu Ala Ala Ser Ala
35 40 45
GCA CCT CCC GGC GCC AGT TTG CTG CTG CTG CAG CAG CAG CAG CAG CAG 551 Ala Pro Pro Gly Ala Ser Leu Leu Leu Leu Gin Gin Gin Gin Gin Gin 50 55 60
CAG CAG CAG CAG CAG CAG CAG CAG CAG CAG CAG CAG CAG CAG CAA GAG 599 Gin Gin Gin Gin Gin Gin Gin Gin Gin Gin Gin Gin Gin Gin Gin Glu 65 70 75
ACT AGC CCC AGG CAG CAG CAG CAG CAG CAG GGT GAG GAT GGT TCT CCC 647 Thr Ser Pro Arg Gin Gin Gin Gin Gin Gin Gly Glu Asp Gly Ser Pro 80 85 90 95
CAA GCC CAT CGT AGA GGC CCC ACA GGC TAC CTG GTC CTG GAT GAG GAA 695 Gin Ala His Arg Arg Gly Pro Thr Gly Tyr Leu Val Leu Asp Glu Glu
100 105 110
CAG CAA CCT TCA CAG CCG CAG TCG GCC CTG GAG TGC CAC CCC GAG AGA 743 Gin Gin Pro Ser Gin Pro Gin Ser Ala Leu Glu Cys His Pro Glu Arg 115 120 125
GGT TGC GTC CCA GAG CCT GGA GCC GCC GTG GCC GCC AGC AAG GGG CTG 791 Gly Cys Val Pro Glu Pro Gly Ala Ala Val Ala Ala Ser Lys Gly Leu 130 135 140
CCG CAG CAG CTG CCA GCA CCT CCG GAC GAG GAT GAC TCA GCT GCC CCA 839 Pro Gin Gin Leu Pro Ala Pro Pro Asp Glu Asp Asp Ser Ala Ala Pro 145 150 155
TCC ACG TTG TCC CTG CTG GGC CCC ACT TTC CCC GGC TTA AGC AGC TGC 887 Ser Thr Leu Ser Leu Leu Gly Pro Thr Phe Pro Gly Leu Ser Ser Cys 160 165 170 175
TCC GCT GAC CTT AAA GAC ATC CTG AGC GAG GCC AGC ACC ATG CAA CTC 935 Ser Ala Asp Leu Lys Asp lie Leu Ser Glu Ala Ser Thr Met Gin Leu
180 185 190
CTT CAG CAA CAG CAG CAG GAA GCA GTA TCC GAA GGC AGC AGC AGC GGG 983 Leu Gin Gin Gin Gin Gin Glu Ala Val Ser Glu Gly Ser Ser Ser Gly 195 200 205 AGA GCG AGG GAG GCC TCG GGG GCT CCC ACT TCC TCC AAG GAC AAT TAC 1031
Arg Ala Arg Glu Ala Ser Gly Ala Pro Thr Ser Ser Lys Asp Asn Tyr 210 215 220
TTA GGG GGC ACT TCG ACC ATT TCT GAC AAC GCC AAG GAG TTG TGT AAG 1079
Leu Gly Gly Thr Ser Thr lie Ser Asp Asn Ala Lys Glu Leu Cys Lys 225 230 235
GCA GTG TCG GTG TCC ATG GGC CTG GGT GTG GAG GCG TTG GAG CAT CTG 1127 Ala Val Ser Val Ser Met Gly Leu Gly Val Glu Ala Leu Glu His Leu
240 245 250 255
AGT CCA GGG GAA CAG CTT CGG GGG GAT TGC ATG TAC GCC CCA CTT TTG 1175
Ser Pro Gly Glu Gin Leu Arg Gly Asp Cys Met Tyr Ala Pro Leu Leu 260 265 270
GGA GTT CCA CCC GCT GTG CGT CCC ACT CCT TGT GCC CCA TTG GCC GAA 1223
Gly Val Pro Pro Ala Val Arg Pro Thr Pro Cys Ala Pro Leu Ala Glu 275 280 285
TGC AAA GGT TCT CTG CTA GAC GAC AGC GCA GGC AAG AGC ACT GAA GAT 1271
Cys Lys Gly Ser Leu Leu Asp Asp Ser Ala Gly Lys Ser Thr Glu Asp 290 295 300
ACT GCT GAG TAT TCC CCT TTC AAG GGA GGT TAC ACC AAA GGG CTA GAA 1319
Thr Ala Glu Tyr Ser Pro Phe Lys Gly Gly Tyr Thr Lys Gly Leu Glu 305 310 315
GGC GAG AGC CTA GGC TGC TCT GGC AGC GCT GCA GCA GGG AGC TCC GGG 1367 Gly Glu Ser Leu Gly Cys Ser Gly Ser Ala Ala Ala Gly Ser Ser Gly
320 325 330 335
ACA CTT GAA CTG CCG TCT ACC CTG TCT CTC TAC AAG TCC GGA GCA CTG 1415
Thr Leu Glu Leu Pro Ser Thr Leu Ser Leu Tyr Lys Ser Gly Ala Leu 340 345 350 GAC GAG GCA GCT GCG TAC CAG AGT CGC GAC TAC TAC AAC TTT CCA CTG 1463 Asp Glu Ala Ala Ala Tyr Gin Ser Arg Asp Tyr Tyr Asn Phe Pro Leu 355 360 365
GCT CTG GCC GGA CCG CCG CCC CCT CCG CCG CCT CCC CAT CCC CAC GCT 1511 Ala Leu Ala Gly Pro Pro Pro Pro Pro Pro Pro Pro His Pro His Ala 370 375 380
CGC ATC AAG CTG GAG AAC CCG CTG GAC TAC GGC AGC GCC TGG GCG GCT 1559 Arg lie Lys Leu Glu Asn Pro Leu Asp Tyr Gly Ser Ala Trp Ala Ala 385 390 395
GCG GCG GCG CAG TGC CGC TAT GGG GAC CTG GCG AGC CTG CAT GGC GCG 1607 Ala Ala Ala Gin Cys Arg Tyr Gly Asp Leu Ala Ser Leu His Gly Ala 400 405 410 415
GGT GCA GCG GGA CCC GGT TCT GGG TCA CCC TCA GCC GCC GCT TCC TCA 1655 Gly Ala Ala Gly Pro Gly Ser Gly Ser Pro Ser Ala Ala Ala Ser Ser 420 425 430
TCC TGG CAC ACT CTC TTC ACA GCC GAA GAA GGC CAG TTG TAT GGA CCG 1703 Ser Trp His Thr Leu Phe Thr Ala Glu Glu Gly Gin Leu Tyr Gly Pro 435 440 445
TGT GGT GGT GGT GGG GGT GGT GGC GGC GGC GGC GGC GGC GGC GGC GGC 1751 Cys Gly Gly Gly Gly Gly Gly Gly Gly Gly Gly Gly Gly Gly Gly Gly 450 455 460
GGC GGC GGC GGC GGC GGC GGC GGC GGC GAG GCG GGA GCT GTA GCC CCC 1799 Gly Gly Gly Gly Gly Gly Gly Gly Gly Glu Ala Gly Ala Val Ala Pro 465 470 475
TAC GGC TAC ACT CGG CCC CCT CAG GGG CTG GCG GGC CAG GAA AGC GAC 1847 Tyr Gly Tyr Thr Arg Pro Pro Gin Gly Leu Ala Gly Gin Glu Ser Asp 480 485 490 495 TTC ACC GCA CCT GAT GTG TGG TAC CCT GGC GGC ATG GTG AGC AGA GTG 1895 Phe Thr Ala Pro Asp Val Trp Tyr Pro Gly Gly Met Val Ser Arg Val 500 505 510
CCC TAT CCC AGT CCC ACT TGT GTC AAA AGC GAA ATG GGC CCC TGG ATG 1943 Pro Tyr Pro Ser Pro Thr Cys Val Lys Ser Glu Met Gly Pro Trp Met 515 520 525
GAT AGC TAC TCC GGA CCT TAC GGG GAC ATG CGT TTG GAG ACT GCC AGG 1991 Asp Ser Tyr Ser Gly Pro Tyr Gly Asp Met Arg Leu Glu Thr Ala Arg 530 535 540
GAC CAT GTT TTG CCC ATT GAC TAT TAC TTT CCA CCC CAG AAG ACC TGC 2039 Asp His Val Leu Pro lie Asp Tyr Tyr Phe Pro Pro Gin Lys Thr Cys 545 550 555
CTG ATC TGT GGA GAT GAA GCT TCT GGG TGT CAC TAT GGA GCT CTC ACA 2087 Leu lie Cys Gly Asp Glu Ala Ser Gly Cys His Tyr Gly Ala Leu Thr 560 565 570 575
TGT GGA AGC TGC AAG GTC TTC TTC AAA AGA GCC GCT GAA GGG AAA CAG 2135 Cys Gly Ser Cys Lys Val Phe Phe Lys Arg Ala Ala Glu Gly Lys Gin 580 585 590
AAG TAC CTG TGC GCC AGC AGA AAT GAT TGC ACT ATT GAT AAA TTC CGA 2183 Lys Tyr Leu Cys Ala Ser Arg Asn Asp Cys Thr lie Asp Lys Phe Arg 595 600 605
AGG AAA AAT TGT CCA TCT TGT CGT CTT CGG AAA TGT TAT GAA GCA GGG 2231 Arg Lys Asn Cys Pro Ser Cys Arg Leu Arg Lys Cys Tyr Glu Ala Gly 610 615 620
ATG ACT CTG GGA GCC CGG AAG CTG AAG AAA CTT GGT AAT CTG AAA CTA 2279 Met Thr Leu Gly Ala Arg Lys Leu Lys Lys Leu Gly Asn Leu Lys Leu 625 630 635 CAG GAG GAA GGA GAG GCT TCC AGC ACC ACC AGC CCC ACT GAG GAG ACA 2327 Gin Glu Glu Gly Glu Ala Ser Ser Thr Thr Ser Pro Thr Glu Glu Thr 640 645 650 655
ACC CAG AAG CTG ACA GTG TCA CAC ATT GAA GGC TAT GAA TGT CAG CCC 2375 Thr Gin Lys Leu Thr Val Ser His He Glu Gly Tyr Glu Cys Gin Pro 660 665 670
ATC TTT CTG AAT GTC CTG GAA GCC ATT GAG CCA GGT GTA GTG TGT GCT 2423 He Phe Leu Asn Val Leu Glu Ala He Glu Pro Gly Val Val Cys Ala 675 680 685
GGA CAC GAC AAC AAC CAG CCC GAC TCC TTT GCA GCC TTG CTC TCT AGC 2471 Gly His Asp Asn Asn Gin Pro Asp Ser Phe Ala Ala Leu Leu Ser Ser 690 695 700
CTC AAT GAA CTG GGA GAG AGA CAG CTT GTA CAC GTG GTC AAG TGG GCC 2519
Leu Asn Glu Leu Gly Glu Arg Gin Leu Val His Val Val Lys Trp Ala
705 710 715
AAG GCC TTG CCT GGC TTC CGC AAC TTA CAC GTG GAC GAC CAG ATG GCT 2567
Lys Ala Leu Pro Gly Phe Arg Asn Leu His Val Asp Asp Gin Met Ala 720 725 730 735
GTC ATT CAG TAC TCC TGG ATG GGG CTC ATG GTG TTT GCC ATG GGC TGG 2615 Val He Gin Tyr Ser Trp Met Gly Leu Met Val Phe Ala Met Gly Trp 740 745 750
CGA TCC TTC ACC AAT GTC AAC TCC AGG ATG CTC TAC TTC GCC CCT GAT 2663 Arg Ser Phe Thr Asn Val Asn Ser Arg Met Leu Tyr Phe Ala Pro Asp 755 760 765
CTG GTT TTC AAT GAG TAC CGC ATG CAC AAG TCC CGG ATG TAC AGC CAG 2711 Leu Val Phe Asn Glu Tyr Arg Met His Lys Ser Arg Met Tyr Ser Gin 770 775 780 TGT GTC CGA ATG AGG CAC CTC TCT CAA GAG TTT GGA TGG CTC CAA ATC 2759 Cys Val Arg Met Arg His Leu Ser Gin Glu Phe Gly Trp Leu Gin He 785 790 795
ACC CCC CAG GAA TTC CTG TGC ATG AAA GCA CTG CTA CTC TTC AGC ATT 2807 Thr Pro Gin Glu Phe Leu Cys Met Lys Ala Leu Leu Leu Phe Ser He 800 805 810 815
ATT CCA GTG GAT GGG CTG AAA AAT CAA AAA TTC TTT GAT GAA CTT CGA 2855 He Pro Val Asp Gly Leu Lys Asn Gin Lys Phe Phe Asp Glu Leu Arg
820 825 830
ATG AAC TAC ATC AAG GAA CTC GAT CGT ATC ATT GCA TGC AAA AGA AAA 2903 Met Asn Tyr He Lys Glu Leu Asp Arg He He Ala Cys Lys Arg Lys 835 840 845
AAT CCC ACA TCC TGC TCA AGA CGC TTC TAC CAG CTC ACC AAG CTC CTG 2951 Asn Pro Thr Ser Cys Ser Arg Arg Phe Tyr Gin Leu Thr Lys Leu Leu 850 855 860
GAC TCC GTG CAG CCT ATT GCG AGA GAG CTG CAT CAG TTC ACT TTT GAC 2999 Asp Ser Val Gin Pro He Ala Arg Glu Leu His Gin Phe Thr Phe Asp 865 870 875
CTG CTA ATC AAG TCA CAC ATG GTG AGC GTG GAC TTT CCG GAA ATG ATG 3047 Leu Leu He Lys Ser His Met Val Ser Val Asp Phe Pro Glu Met Met 880 885 890 895
GCA GAG ATC ATC TCT GTG CAA GTG CCC AAG ATC CTT TCT GGG AAA GTC 3095 Ala Glu He He Ser Val Gin Val Pro Lys He Leu Ser Gly Lys Val
900 905 910
AAG CCC ATC TAT TTC CAC ACC CAG TGAAGCATTG GAAACCCTAT TTCCCCACCC 3149 Lys Pro He Tyr Phe His Thr Gin 915 920 CAGCTCATGC CCCCTTTCAG ATGTCTTCTG CCTGTTATAA CTCTGCACTA CTCCTCTGCA 3209
GTGCCTTGGG GAATTTCCTC TATTGATGTA CAGTCTGTCA TGAACATGTT CCTGAATTCT 3269
ATTTGCTGGG CTTTTTTTTT CTCTTTCTCT CCTTTCTTTT TCTTCTTCCC TCCCTATCTA 3329
ACCCTCCCAT GGCACCTTCA GACTTTGCTT CCCATTGTGG CTCCTATCTG TGTTTTGAAT 3389
GGTGTTGTAT GCCTTTAAAT CTGTGATGAT CCTCATATGG CCCAGTGTCA AGTTGTGCTT 3449
GTTTACAGCA CTACTCTGTG CCAGCCACAC AAACGTTTAC TTATCTTATG CCACGGGAAG 3509
TTTAGAGAGC TAAGATTATC TGGGGAAATC AAAACAAAAA ACAAGCAAAC AAAAAAAAAA 3569
(2) INFORMATION FOR SEQ ID NO:2 :
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 1082 base pairs (B) TYPE: nucleic acid
(C) STRANDEDNESS: double
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: DNA (genomic)
(iii) HYPOTHETICAL: NO
(iv) ANTI-SENSE: NO
(vi) ORIGINAL SOURCE:
(A) ORGANISM: HOMO SAPIENS
(ix) FEATURE:
(A) NAME/KEY: exon (B) LOCATION: 602..770 (D) OTHER INFORMATION: /note= "SEGMENT 1 OF 3."
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:2 :
AAGCTTCCCT TAACATACTA ACCCTTTACT TTCCCTGTTG TGTCCCTGAA AGGCCTCCTG 60
TGCCTTTGGC TGCAGGTCCC GAACGTCCAG GCCATCTGTG CTATCTGCTT CGCGGTACCT 120
CACCAACGCA ACGTGAGGGT GGAGGGCAGA ACCTTGGTCC TGGCCTCTCA GCTTTTGTGG 180
GTTTCAGCCA GACCCTAGGT GTTATTTTAG TGCAACTTTG GTGTTTAATT TGAGGATGTG 240
TGTGGACCAG AAGGAGGGAC CAAAACATGA TTCTTTTCCC CATGGTCAGA TGATTAAATT 300
TGAAGTTCTA AAAAATGCAG TTTGGTCCAA AGCTGTGTCC AATTGGGAAG AGAGAAAAAT 360
GCCCTGGAAA CCCCTCCCAG GCCTGGGACC ATCCTTCCTT AACCACCAGC CACCTCACAG 420
GCCCGCGGAC TGCGGGCATC ACCTGGGCAG GCTGTGCTTA CTCACTACCC GGGAACCCTG 480
TGCCCTGGAG CTGTCCTTCC TCTCTTCAAA GTGCATTTTG TGCCTTTGCT GGAAGAACCG 540
ACTACAGGTT TGTTCAATTT CTTACAGTCT TGAAAGCGCC ACAAGCAGCA GCTGCTGAGC 600
CATGGCTGAA GGGGAAATCA CCACCTTCAC AGCCCTGACC GAGAAGTTTA ATCTGCCTCC 660
AGGGAATTAC AAGAAGCCCA AACTCCTCTA CTGTAGCAAC GGGGGCCACT TCCTGAGGAT 720
CCTTCCGGAT GGCACAGTGG ATGGGACAAG GGACAGGAGC GACCAGCACA GTAAGCCCAT 780
CTCTATGGCA CCCCCCTTCC CTTTCTGACA TCTTCTGTAG TCAAGGTGGG AGGAAGGTGC 840
ACATTTAAGT ACAGGTACTT GCTTCTCCAA GGTTCTATTC AGGCATGACA CATTCAGAGG 900
TGGAGTCACA TAAATGCGTA AAATGTCTGG GAAATGAAAA TAGGGACTTG TGGGGGCCAC 960
CACTTACCCA AACGTGTCCT ATTTCAAGTT TTTTAAAGCA CTCTCTGCTG ACCCAACAGA 1020 ACGGGCTGCC GGTGCTCAAT TGCTGTATGT TTTCCCAGGT TTCTGTAACT AGTGAAAGAT 1080
CT 1082
(2) INFORMATION FOR SEQ ID NO:3 :
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 427 base pairs
(B) TYPE: nucleic acid (C) STRANDEDNESS: double
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: DNA (genomic)
(iii) HYPOTHETICAL: NO
(iv) ANTI-SENSE: NO
(vi) ORIGINAL SOURCE: (A) ORGANISM: HOMO SAPIENS
(ix) FEATURE:
(A) NAME/KEY: exon
(B) LOCATION: 186..289 (D) OTHER INFORMATION: /note= "SEGMENT 2 OF 3. UNKNOWN
NUMBER OF BP AFTER SEGMENT 1. "
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:3 :
CAGCTTTCTT TGGAAGGCAA AGAAAAAGGG ACTGTATTTC TATGTTTTGA TTAATCTGAG 60
GCTCATCCTG AGGGCTCCGT GAAATGAATG AGCAGAATTT TCCATGGCCA ACTGTCCTGG 120
CTGCCGGGTC CTATCGGCAA AAGCGTAGTG TTTATTTACT TTTGCTCGTG TTATTTTTAT 180 TCCAGTTCAG CTGCAGCTCA GTGCGGAAAG CGTGGGGGAG GTGTATATAA AGAGTACCGA 240
GACTGGCCAG TACTTGGCCA TGGACACCGA CGGGCTTTTA TACGGCTCAG TAAGTATGAA 300
GCTGACATGC TTCCAGACGT TGGCCAAGGT TTGAGGTTTC CAGAAATCTT GTTACATGGA 360
GTGAGGCAAA CTATAAAGCA ACAATTAGTC TCTGTTTGTT ATTTTTTCCA GAAGGATTCC 420
CACCCTC 427
(2) INFORMATION FOR SEQ ID NO:4:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 664 base pairs (B) TYPE: nucleic acid
(C) STRANDEDNESS: double
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: DNA (genomic)
(iii) HYPOTHETICAL: NO
(iv) ANTI-SENSE: NO
(vi) ORIGINAL SOURCE:
(A) ORGANISM: HOMO SAPIENS
(ix) FEATURE:
(A) NAME/KEY: exon (B) LOCATION: 304..498
(D) OTHER INFORMATION: /note= "SEGMENT 3 OF 3. UNKNOWN NUMBER OF BP AFTER SEGMENT 2. "
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:4 : TGAGGACTCT TAGAAGTGCT CTTATCAGTA GCATCTTAAT TACTTTACAA TGGATTTTAA 60
ATGGAAAGGA AGTTTACAAT AATAGCAAAT GCATATTGAC AGCTCTTTAG TGCCCGGTGC 120
TGTTCTAAGT CCTTATGACT ACCCTGTGAA ATAAGTTCCA CCATGACCCC AATTTTCCTG 180
AAAAGGAGAC TGAGGCATGG AGAGCTTTAG TATTTTGCCC AATGTCACAC AGCTAGTAAA 240
TGGGGACCCC CATGTGAAAC TACTCACTGA TTGTCCTACT CTCTTGTGGT TTTATCTTTT 300
TAGCAGACAC CAAATGAGGA ATGTTTGTTC CTGGAAAGGC TGGAGGAGAA CCATTACAAC 360
ACCTATATAT CCAAGAAGCA TGCAGAGAAG AATTGGTTTG TTGGCCTCAA GAAGAATGGG 420
AGCTGCAAAC GCGGTCCTCG GACTCACTAT GGCCAGAAAG CAATCTTGTT TCTCCCCCTG 480
CCAGTCTCTT CTGATTAAAG AGATCTGTTC TGGGTGTTGA CCACTCCAGA GAAGTTTCGA 540
GGGGTCCTCA CCTGGTTGAC CCAAAAATGT TCCCTTGACC ATTGGCTGCG CTAACCCCCA 600
GCCCACAGAG CCTGAATTTG TAAGCAACTT GCTTCTAAAT GCCCAGTTCA CTTCTTTGCA 660
GAGC 664
(2) INFORMATION FOR SEQ ID NO: 5:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 27 base pairs
(B) TYPE: nucleic acid (C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: cDNA
(iii) HYPOTHETICAL: NO (iv) ANTI-SENSE: YES
(vi) ORIGINAL SOURCE:
(A) ORGANISM: SYNTHETIC OLIGONUCLEOTIDE
(ix) FEATURE:
(A) NAME/KEY: misc_feature
(B) LOCATION: 1..27
(D) OTHER INFORMATION: /note= "ANTISENSE TO POSITIONS 927-953 OF SEQ ID NO. : 1."
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 5:
CTGCTGCTGT TGCTGAAGGA GTTGCAT 27
(2) INFORMATION FOR SEQ ID NO:6:
(i) SEQUENCE CHARACTERISTICS: (A) LENGTH: 29 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: cDNA
(iii) HYPOTHETICAL: NO
(iv) ANTI-SENSE: YES
(vi) ORIGINAL SOURCE:
(A) ORGANISM: SYNTHETIC OLIGONUCLEOTIDE
(ix) FEATURE: (A) NAME/KEY: misc feature ( B ) LOCATION : 1 . . 21
(D) OTHER INFORMATION: /note= "ANTISENSE TO POSITIONS 916-936 OF SEQ ID NO.: 1."
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:6 :
GGAGTTGCAT GGTGCTGGCC TCAGCACCA 29
(2) INFORMATION FOR SEQ ID NO:7:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 29 base pairs
(B) TYPE: nucleic acid (C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: CDNA
(iii) HYPOTHETICAL: NO
(iv) ANTI-SENSE: YES
(vi) ORIGINAL SOURCE: (A) ORGANISM: SYNTHETIC OLIGONUCLEOTIDE
(ix) FEATURE:
(A) NAME/KEY: misc_feature
(B) LOCATION: 1..21 (D) OTHER INFORMATION: /note= "ANTISENSE TO POSITIONS
927-947 OF SEQ ID NO. : 1."
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 7 : CTGTTGCTGA AGGAGTTGCA TAACTCCTT 29
(2) INFORMATION FOR SEQ ID NO:8:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 25 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: cDNA
(iii) HYPOTHETICAL: NO
(iv) ANTI-SENSE: YES
(vi) ORIGINAL SOURCE:
(A) ORGANISM: SYNTHETIC OLIGONUCLEOTIDE
(ix) FEATURE:
(A) NAME/KEY: misc_feature
(B) LOCATION: 1..25
(D) OTHER INFORMATION: /note= "ANTISENSE TO POSITIONS 611-635 OF SEQ ID NO. : 2."
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:8 :
GGGCTGTGAA GGTGGTGATT TCCCC 25

Claims

CLAIMS We claim:
1. A method for treating a patient diagnosed as having benign prostatic hypeφlasia or a prostatic cancer comprising administering to said patient a therapeutically effective amount of a composition comprising an antisense oligonucleotide which selectively hybridizes to a gene or mRNA sequence of said patient; wherein said antisense inhibits expression of said gene or mRNA sequence; and wherein said gene or mRNA sequence is selected from the group consisting of an AR and an αFGF gene or mRNA sequence.
2. A method as in claim 1 wherein said oligonucleotide is selected from the group consisting of (a) oligonucleotides comprising at least 10 consecutive bases from SEQ ID NO.: 1 ;
(b) oligonucleotides comprising at least 10 consecutive bases from a genomic sequence corresponding to SEQ ID NO.: 1 ; and
(c) oligonucleotides that hybridize to the complements of the oligonucleotides of (a) or (b) under physiological conditions.
3. A method as in claim 1 wherein said oligonucleotide is selected from the group consisting of
(a) oligonucleotides comprising at least 20 consecutive bases from SEQ ID NO.: 1 ;
(b) oligonucleotides comprising at least 10 consecutive bases from a genomic sequence corresponding to SEQ ID NO.: 1; and
(c) oligonucleotides that hybridize to the complements ofthe oligonucleotides of (a) or (b) under physiological conditions.
4. A method as in claim 1 wherein said oligonucleotide is selected from the group consisting of
(a) oligonucleotides comprising at least 10 consecutive bases from the group consisting of SEQ ID NO.: 2, SEQ ID NO.: 3 and SEQ ID NO.: 4; (b) oligonucleotides comprising at least 10 consecutive bases from the joined exons of
SEQ ID NO. : 2, SEQ ID NO.: 3 and SEQ ID NO.: 4; and
(c) oligonucleotides that hybridize to the complements ofthe oligonucleotides of (a) or (b) under physiological conditions.
5. A method as in claim 1 wherein said oligonucleotide is selected from the group consisting of
(a) oligonucleotides comprising at least 20 consecutive bases from the group consisting of SEQ ID NO.: 2, SEQ ID NO.: 3 and SEQ ID NO.: 4;
(b) oligonucleotides comprising at least 20 consecutive bases from the joined exons of SEQ ID NO. : 2, SEQ ID NO.: 3 and SEQ ID NO.: 4; and
(c) oligonucleotides that hybridize to the complements ofthe oligonucleotides of (a) or (b) under physiological conditions.
6. A method as in claim 1 wherein said oligonucleotide comprises a nucleotide sequence selected from the group consisting of SEQ ID NO.: 5, SEQ ID NO.: 6. SEQ ID NO.: 7, and SEQ ID NO.: 8.
7. A method as in claim 1 wherein said oligonucleotide is a modified oligonucleotide.
8. A method as in claim 7 wherein said oligonucleotide is a modified oligonucleotide including at least one synthetic intemucleoside linkage.
9. A method as in claim 8 wherein said synthetic intemucleoside linkage is selected from the group consisting of phosphorothioates, alkylphosphonates, phosphorodithioates, phosphate esters, alkylphosphonothioates, phosphoramidates, carbamates, carbonates, phosphate triesters, acetamidates, and carboxymethyl esters.
10. A method as in claim 7 wherein said oligonucleotide is a modified oligonucleotide having at least one low molecular weight organic group covalently bound to a phosphate group of said oligonucleotide.
11. A method as in claim 7 wherein said oligonucleotide is a modified oligonucleotide having at least one low molecular weight organic group covalently bound to a 2' position of a ribose of said oligonucleotide.
12. A method as in claim 7 wherein said oligonucleotide is a modified oligonucleotide having covalently attached thereto a compound selected from the group consisting of androgen, androgen derivatives, estrogen, estrogen derivatives, estramustine, emcyt and estracyt.
13. A method as in claim 1 wherein said oligonucleotide is administered intravenously at a dosage between 1.0 μg and 100 mg per kg body weight of said patient.
14. A method as in claim 1 wherein said patient has a prostatic cancer which is refractory to anti-androgen or estrogen hormonal therapy.
15. A pharmaceutical composition comprising a sterile pharmaceutically acceptable carrier; and a therapeutically effective amount of an isolated antisense oligonucleotide which selectively hybridizes to a gene or mRNA sequence of a patient; wherein said antisense inhibits expression of said gene or mRNA sequence; and wherein said gene or mRNA sequence is selected from the group consisting of an AR and an αFGF gene or mRNA sequence.
16. A composition as in claim 15 wherein said oligonucleotide is selected from the group consisting of (a) oligonucleotides comprising at least 10 consecutive bases from SEQ ID NO.: 1;
(b) oligonucleotides comprising at least 10 consecutive bases from the joined exons of SEQ ID NO.: l; and
(c) oligonucleotides that hybridize to the complements ofthe oligonucleotides of (a) or (b) under physiological conditions.
17. A composition as in claim 15 wherein said oligonucleotide is selected from the group consisting of
(a) oligonucleotides comprising at least 20 consecutive bases from SEQ ID NO.: 1 ; (b) oligonucleotides comprising at least 20 consecutive bases from the joined exons of
SEQ ID NO.: l ; and
(c) oligonucleotides that hybridize to the complements of the oligonucleotides of (a) or (b) under physiological conditions.
18. A composition as in claim 15 wherein said oligonucleotide is selected from the group consisting of
(a) oligonucleotides comprising at least 10 consecutive bases from SEQ ID NO.: 2;
(b) oligonucleotides comprising at least 10 consecutive bases from a genomic sequence corresponding to SEQ ID NO.: 2; and (c) oligonucleotides that hybridize to the complements of the oligonucleotides of (a) or
(b) under physiological conditions.
19. A composition as in claim 15 wherein said oligonucleotide is selected from the group consisting of (a) oligonucleotides comprising at least 20 consecutive bases from SEQ ID NO.: 2;
(b) oligonucleotides comprising at least 20 consecutive bases from a genomic sequence corresponding to SEQ ID NO.: 2; and
(c) oligonucleotides that hybridize to the complements ofthe oligonucleotides of (a) or (b) under physiological conditions.
20. A composition as in claim 15 wherein said oligonucleotide comprises a nucleotide sequence selected from the group consisting of SEQ ID NO.: 5, SEQ ID NO.: 6, SEQ ID NO.: 7. SEQ ID NO.: 8, and SEQ ID NO.: 9.
21. A composition as in claim 15 wherein said oligonucleotide is a modified oligonucleotide.
22. A composition as in claim 15 wherein said oligonucleotide is a modified oligonucleotide including at least one synthetic intemucleoside linkage.
23. A composition as in claim 22 wherein said synthetic intemucleoside linkage is selected from the group consisting of phosphorothioates, alkylphosphonates, phosphorodithioates, phosphate esters, alkylphosphonothioates, phosphoramidates, carbamates, carbonates, phosphate triesters, acetamidates, and carboxymethyl esters.
24. A composition as in claim 21 wherein said oligonucleotide is a modified oligonucleotide having at least one low molecular weight organic group covalently bound to a phosphate group of said oligonucleotide.
25. A composition as in claim 21 wherein said oligonucleotide is a modified oligonucleotide having at least one low molecular weight organic group covalently bound to a 2' position of a ribose of said oligonucleotide.
26. A composition as in claim 21 wherein said oligonucleotide is a modified oligonucleotide having covalently attached thereto a compound selected from the group consisting of androgen, androgen derivatives, estrogen, estrogen derivatives, estramustine. emcyt and estracyt.
27. A pharmaceutical kit comprising the pharmaceutical composition of claim 15 in a pharmaceutically acceptable carrier for intravenous administration.
28. A method for treating a patient diagnosed as having benign prostatic hyperplasia or a prostatic cancer comprising administering to said patient a therapeutically effective amount of a composition comprising an antisense oligonucleotide which selectively hybridizes to a gene or mRNA sequence of said patient; wherein said antisense inhibits expression of said gene or mRNA sequence; and wherein said antisense inhibits or represses prostatic cell growth.
29. A method as in claim 28 wherein said gene is selected from the group consisting of a PSA gene, a probasin gene, an αFGF gene, an androgen receptor gene, an estrogen receptor gene, a telomerase gene, a prohibitin gene, a src gene, a ras gene, a myc gene, a blc-2 gene, a protein kinase-A gene, a plasminogen activator urokinase gene and a methyl transferase gene.
PCT/US1996/015081 1995-09-20 1996-09-20 Antisense oligonucleotide chemotherapy for benign hyperplasia or cancer of the prostate WO1997011170A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
EP96935879A EP0851919A1 (en) 1995-09-20 1996-09-20 Antisense oligonucleotide chemotherapy for benign hyperplasia or cancer of the prostate
AU73662/96A AU7366296A (en) 1995-09-20 1996-09-20 Antisense oligonucleotide chemotherapy for benign hyperplasia or cancer of the prostate

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US401895P 1995-09-20 1995-09-20
US60/004,018 1995-09-20

Publications (1)

Publication Number Publication Date
WO1997011170A1 true WO1997011170A1 (en) 1997-03-27

Family

ID=21708727

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1996/015081 WO1997011170A1 (en) 1995-09-20 1996-09-20 Antisense oligonucleotide chemotherapy for benign hyperplasia or cancer of the prostate

Country Status (4)

Country Link
EP (1) EP0851919A1 (en)
AU (1) AU7366296A (en)
CA (1) CA2239976A1 (en)
WO (1) WO1997011170A1 (en)

Cited By (17)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2000027858A1 (en) * 1998-11-09 2000-05-18 Institute Of Radiation Medicine, Academy Of Military Medecine Science Antisense oligonucleotides which inhibit telomerase's activity and their uses
EP1071762A1 (en) 1998-03-20 2001-01-31 Benitec Australia Ltd. Control of gene expression
EP1518928A1 (en) * 2003-09-26 2005-03-30 Fundacion Pablo Cassara Antiandrogen oligonucleotides usable for the treatment of dermatological androgen-related disorders relating to androgen metabolism, their pharmaceutical compositions, their uses and treatment method
US7056704B2 (en) 2000-12-01 2006-06-06 Max-Planck-Gesellschaft Zur Foerderung Der Wissenschaften E.V. RNA interference mediating small RNA molecules
WO2007041497A2 (en) * 2005-09-30 2007-04-12 Cleveland Biolabs, Inc. Modulation of androgen receptor
US7737125B2 (en) 2007-11-26 2010-06-15 Enzon Pharamaceuticals, Inc. LNA antagonists targeting the androgen receptor
US8048670B2 (en) 1998-03-20 2011-11-01 Commonwealth Scientific And Industrial Research Organisation Synthetic genes and genetic constructs
WO2012065051A1 (en) 2010-11-12 2012-05-18 Enzon Pharmaceuticals, Inc. Compositions and methods for treating androgen receptor dependent disorders including cancers
US8183217B2 (en) 1999-08-13 2012-05-22 Commonwealth Scientific And Industrial Research Organisation Methods and means for obtaining modified phenotypes
US8394628B2 (en) 2000-03-30 2013-03-12 University Of Massachusetts RNA sequence-specific mediators of RNA interference
US8450290B2 (en) 2007-11-26 2013-05-28 Enzon Pharmaceuticals, Inc. Methods for treating androgen receptor dependent disorders including cancers
US8729036B2 (en) 2002-08-07 2014-05-20 University Of Massachusetts Compositions for RNA interference and methods of use thereof
US9051566B2 (en) 2001-01-31 2015-06-09 Alnylam Pharmaceuticals, Inc. Post-transcriptional gene silencing using expressed double stranded RNA
US9102939B2 (en) 1997-12-23 2015-08-11 The Carnegie Institution Of Washington Genetic inhibition by double-stranded RNA
US9175291B2 (en) 2012-10-11 2015-11-03 Isis Pharmaceuticals Inc. Modulation of androgen receptor expression
US20160281085A1 (en) * 2013-11-01 2016-09-29 Everon Biosciences, Inc. Molecular targets for selective eradication of senescent cells
EP3327144A1 (en) 2013-02-25 2018-05-30 Novartis AG Novel androgen receptor mutation

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1989009791A1 (en) * 1988-04-14 1989-10-19 University Of North Carolina At Chapel Hill Dna encoding androgen receptor protein
WO1994005268A1 (en) * 1992-09-04 1994-03-17 Baylor College Of Medicine Novel triplex forming oligonucleotides and methods for their use
WO1995011301A1 (en) * 1993-10-19 1995-04-27 The Regents Of The University Of Michigan P53-mediated apoptosis
WO1996003875A1 (en) * 1994-07-29 1996-02-15 Emory University Compositions for targeting materials to cells containing androgen receptors
US5556956A (en) * 1993-11-04 1996-09-17 Board Of Regents, The University Of Texas System Methods and compositions relating to the androgen receptor gene and uses thereof

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1989009791A1 (en) * 1988-04-14 1989-10-19 University Of North Carolina At Chapel Hill Dna encoding androgen receptor protein
WO1994005268A1 (en) * 1992-09-04 1994-03-17 Baylor College Of Medicine Novel triplex forming oligonucleotides and methods for their use
WO1995011301A1 (en) * 1993-10-19 1995-04-27 The Regents Of The University Of Michigan P53-mediated apoptosis
US5556956A (en) * 1993-11-04 1996-09-17 Board Of Regents, The University Of Texas System Methods and compositions relating to the androgen receptor gene and uses thereof
WO1996003875A1 (en) * 1994-07-29 1996-02-15 Emory University Compositions for targeting materials to cells containing androgen receptors

Non-Patent Citations (8)

* Cited by examiner, † Cited by third party
Title
87TH ANNUAL MEETING OF THE AMERICAN ASSOCIATION FOR CANCER RESEARCH, WASHINGTON, D.C., USA, APRIL 20-24, 1996. *
ACHBAROU, A. ET AL.: "Urokinase overproduction results in increased skeletal metastasis by prostate cancer cells in vivo.", CANCER RESEARCH, (1994 MAY 1) 54 (9) 2372-7., XP002025258 *
BOFFA, L. ET AL.: "Invasion of the CAG triplet repeats by a complementary peptide nucleic acid inhibits transcription of the androgen receptor and TATA-binding protein genes and correlates with refolding of an active nucleosome containing a unique AR gene sequence.", JOURNAL OF BIOLOGICAL CHEMISTRY, (1996 MAY 31) 271 (22) 13228-33., XP002025260 *
HEAD, M. ET AL.: "Penetration and stability of antisense oligonucleotides injected into the early embryonic chick eye", ANTISENSE RES.DEV. 5 ( FALL 1995); PAGE 239; ABSTRACT III12, XP002025259 *
INT.CONF.:'THERAPEUTIC OLIGONUCLEOTIDES FROM CELL TO MAN'; 4 TO 7 APRIL 1995; SEILLAC; FRANCE *
SHAIN, S. ET AL.: "Endogenous fibroblast growth factor - 1 or fibroblast growth factor -2 modulate prostate cancer cell proliferation.", CELL GROWTH AND DIFFERENTIATION, (1996 MAY) 7 (5) 573-86., XP000616505 *
SHERIDAN, V. & TEW, K.: "Mechanism based chemotherapy for prostate cancer", CANCER SURVEYS, vol. 11, 1991, pages 239 - 254, XP000616360 *
STEINER, M. ET AL.: "Gene therapy of advanced prostate cancer by in vivo transduction with prostate-targeted antisense c- myc RNA retroviruses.", PROCEEDINGS OF THE AMERICAN ASSOCIATION FOR CANCER RESEARCH ANNUAL MEETING 37 (0), March 1996 (1996-03-01), pages 344, XP002025261 *

Cited By (59)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9102939B2 (en) 1997-12-23 2015-08-11 The Carnegie Institution Of Washington Genetic inhibition by double-stranded RNA
US7754697B2 (en) 1998-03-20 2010-07-13 Commonwealth Scientific And Industrial Research Organisation Control of gene expression
US9029527B2 (en) 1998-03-20 2015-05-12 Commonwealth Scientific And Industrial Research Organisation Synthetic genes and genetic constructs
US8168774B2 (en) 1998-03-20 2012-05-01 Commonwealth Scientific And Industrial Research Organisation Control of gene expression
US9963698B2 (en) 1998-03-20 2018-05-08 Commonwealth Scientific And Industrial Research Organisation Control of gene expression
EP2302057A1 (en) 1998-03-20 2011-03-30 Commonwealth Scientific and Industrial Research Organization Control of gene expression
EP1071762A1 (en) 1998-03-20 2001-01-31 Benitec Australia Ltd. Control of gene expression
EP2302057B1 (en) * 1998-03-20 2019-02-20 Commonwealth Scientific and Industrial Research Organisation Control of gene expression
US8067383B2 (en) 1998-03-20 2011-11-29 Commonwealth Scientific And Industrial Research Organisation Synthetic genes and genetic constructs comprising same I
US8053419B2 (en) 1998-03-20 2011-11-08 Commonwealth Scientific And Industrial Research Organisation Synthetic genes and genetic constructs
US8048670B2 (en) 1998-03-20 2011-11-01 Commonwealth Scientific And Industrial Research Organisation Synthetic genes and genetic constructs
US8431547B2 (en) 1998-03-20 2013-04-30 Commonwealth Scientific And Industrial Research Organisation Synthetic genes and genetic constructs
WO2000027858A1 (en) * 1998-11-09 2000-05-18 Institute Of Radiation Medicine, Academy Of Military Medecine Science Antisense oligonucleotides which inhibit telomerase's activity and their uses
US9708621B2 (en) 1999-08-13 2017-07-18 Commonwealth Scientific And Industrial Research Organisation Methods and means for obtaining modified phenotypes
US8183217B2 (en) 1999-08-13 2012-05-22 Commonwealth Scientific And Industrial Research Organisation Methods and means for obtaining modified phenotypes
US10190127B2 (en) 1999-08-13 2019-01-29 Commonwealth Scientific And Industrial Research Organisation Methods and means for obtaining modified phenotypes
US8334374B2 (en) 1999-08-13 2012-12-18 Commonwealth Scientific And Industrial Research Organisation Methods and means for obtaining modified phenotypes
US8632997B2 (en) 2000-03-30 2014-01-21 University Of Massachusetts RNA sequence-specific mediators of RNA interference
US8742092B2 (en) 2000-03-30 2014-06-03 University Of Massachusetts RNA sequence-specific mediators of RNA interference
US9193753B2 (en) 2000-03-30 2015-11-24 University Of Massachusetts RNA sequence-specific mediators of RNA interference
US9012138B2 (en) 2000-03-30 2015-04-21 MAX-PLANCK-Gesellschaft zur Förderung der Wissenschaften e.V. RNA sequence-specific mediators of RNA interference
US9012621B2 (en) 2000-03-30 2015-04-21 MAX-PLANCK-Gesellschaft zur Förderung der Wissenschaften e.V. RNA sequence-specific mediators of RNA interference
US8394628B2 (en) 2000-03-30 2013-03-12 University Of Massachusetts RNA sequence-specific mediators of RNA interference
US8420391B2 (en) 2000-03-30 2013-04-16 University Of Massachusetts RNA sequence-specific mediators of RNA interference
US10472625B2 (en) 2000-03-30 2019-11-12 MAX-PLANCK-Gesellschaft zur Förderung der Wissenschaften e.V. RNA sequence-specific mediators of RNA interference
US8790922B2 (en) 2000-03-30 2014-07-29 MAX-PLANCK-Gesellschaft zur Förderung der Wissenschaften e.V. RNA sequence-specific mediators of RNA interference
US8552171B2 (en) 2000-03-30 2013-10-08 University Of Massachusetts RNA sequence-specific mediators of RNA interference
US8853384B2 (en) 2000-12-01 2014-10-07 MAX-PLANCK-Gesellschaft zur Förderung der Wissenschaften e.V. RNA interference mediating small RNA molecules
US8765930B2 (en) 2000-12-01 2014-07-01 MAX-PLANCK-Gesellschaft zur Förderung der Wissenschaften e.V. RNA interference mediating small RNA molecules
US7056704B2 (en) 2000-12-01 2006-06-06 Max-Planck-Gesellschaft Zur Foerderung Der Wissenschaften E.V. RNA interference mediating small RNA molecules
AU2002235744B8 (en) * 2000-12-01 2007-06-28 Europaisches Laboratorium Fur Molekularbiologie (Embl) RNA interference mediating small RNA molecules
US8895721B2 (en) 2000-12-01 2014-11-25 MAX-PLANCK-Gesellschaft zur Förderung der Wissenschaften e.V. RNA interference mediating small RNA molecules
US8778902B2 (en) 2000-12-01 2014-07-15 MAX-PLANCK-Gesellschaft zur Förderung der Wissenschaften e.V. RNA interference mediating small RNA molecules
US8445237B2 (en) 2000-12-01 2013-05-21 MAX-PLANCK-Gesellschaft zur Förderung der Wissenschaften e.V. RNA interference mediating small RNA molecules
US8895718B2 (en) 2000-12-01 2014-11-25 MAX-PLANCK-Gesellschaft zur Förderung der Wissenschaften e.V. RNA interference mediating small RNA molecules
US8329463B2 (en) 2000-12-01 2012-12-11 MAX-PLANCK-Gesellschaft zur Förderung der Wissenschaften e.V. RNA interference mediating small RNA molecules
US7078196B2 (en) 2000-12-01 2006-07-18 Max-Planck-Gesellschaft Zur Foerderung Der Wissenschaften, E.V. RNA interference mediating small RNA molecules
US8796016B2 (en) 2000-12-01 2014-08-05 MAX-PLANCK-Gesellschaft zur Förderung der Wissenschaften e.V. RNA interference mediating small RNA molecules
US8933044B2 (en) 2000-12-01 2015-01-13 MAX-PLANCK-Gesellschaft zur Förderung der Wissenschaften e.V. RNA interference mediating small RNA molecules
US8993745B2 (en) 2000-12-01 2015-03-31 MAX-PLANCK-Gesellschaft zur Förderung der Wissenschaften e.V. RNA interference mediating small RNA molecules
US8372968B2 (en) 2000-12-01 2013-02-12 MAX-PLANCK-Gesellschaft zur Förderung der Wissenschaften e.V. RNA interference mediating small RNA molecules
US8362231B2 (en) 2000-12-01 2013-01-29 Max-Planck-Gesellschaft zur Föderung der Wissenschaften E.V. RNA interference mediating small RNA molecules
US10633656B2 (en) 2000-12-01 2020-04-28 Max-Planck-Gesellschaft Zur Forderung Der Wissenschaften E.V. RNA interference mediating small RNA molecules
US9051566B2 (en) 2001-01-31 2015-06-09 Alnylam Pharmaceuticals, Inc. Post-transcriptional gene silencing using expressed double stranded RNA
US9611472B2 (en) 2002-08-07 2017-04-04 University Of Massachusetts Compositions for RNA interference and methods of use thereof
US8729036B2 (en) 2002-08-07 2014-05-20 University Of Massachusetts Compositions for RNA interference and methods of use thereof
EP1518928A1 (en) * 2003-09-26 2005-03-30 Fundacion Pablo Cassara Antiandrogen oligonucleotides usable for the treatment of dermatological androgen-related disorders relating to androgen metabolism, their pharmaceutical compositions, their uses and treatment method
WO2007041497A3 (en) * 2005-09-30 2011-05-26 Cleveland Biolabs, Inc. Modulation of androgen receptor
WO2007041497A2 (en) * 2005-09-30 2007-04-12 Cleveland Biolabs, Inc. Modulation of androgen receptor
US8450290B2 (en) 2007-11-26 2013-05-28 Enzon Pharmaceuticals, Inc. Methods for treating androgen receptor dependent disorders including cancers
US7737125B2 (en) 2007-11-26 2010-06-15 Enzon Pharamaceuticals, Inc. LNA antagonists targeting the androgen receptor
US7989429B2 (en) 2007-11-26 2011-08-02 Enzon Pharmaceuticals, Inc. LNA antagonists targeting the androgen receptor
WO2012065051A1 (en) 2010-11-12 2012-05-18 Enzon Pharmaceuticals, Inc. Compositions and methods for treating androgen receptor dependent disorders including cancers
US9175291B2 (en) 2012-10-11 2015-11-03 Isis Pharmaceuticals Inc. Modulation of androgen receptor expression
EP3327144A1 (en) 2013-02-25 2018-05-30 Novartis AG Novel androgen receptor mutation
US10011874B2 (en) 2013-02-25 2018-07-03 Novartis Ag Androgen receptor mutation
EP3696276A1 (en) 2013-02-25 2020-08-19 Novartis AG Novel androgen receptor mutation
US20160281085A1 (en) * 2013-11-01 2016-09-29 Everon Biosciences, Inc. Molecular targets for selective eradication of senescent cells
US9909125B2 (en) * 2013-11-01 2018-03-06 Everon Biosciences, Inc. Molecular targets for selective eradication of senescent cells

Also Published As

Publication number Publication date
CA2239976A1 (en) 1997-03-27
AU7366296A (en) 1997-04-09
EP0851919A1 (en) 1998-07-08

Similar Documents

Publication Publication Date Title
WO1997011170A1 (en) Antisense oligonucleotide chemotherapy for benign hyperplasia or cancer of the prostate
US5783683A (en) Antisense oligonucleotides which reduce expression of the FGFRI gene
US5652222A (en) Selective inhibition of leukemic cell proliferation by bcr-abl antisense oligonucleotides
FI112379B (en) A process for the preparation of therapeutically useful oligonucleotides
US20100216867A1 (en) Methods of treatment of a bcl-2 disorder using bcl-2 antisense oligomers
US5990299A (en) Control of CD44 gene expression for therapeutic use
CA2164288A1 (en) Method for treating kaposi's sarcoma with antisense oligonucleotides
US5866699A (en) Oligonucleotides with anti-MDR-1 gene activity
WO1991018625A1 (en) Ribozyme mediated reversal of transformation by cleavage of the hras oncogene rna
US20040082534A1 (en) Treatment of melanoma by reduction in clusterin levels
US20040147473A1 (en) Methods of treatment of a bcl-2 disorder using bcl-2 antisense oligomers
EP0668782B1 (en) Combination of antineoplastic agent and antisense oligonucleotides for treatment of cancer
Rubenstein et al. Antisense oligonucleotide intralesional therapy for human PC‐3 prostate tumors carried in athymic nude mice
CA2375467A1 (en) Antisense therapy for hormone-regulated tumors
JPH11512601A (en) Modified protein kinase A-specific oligonucleotides and uses thereof
CA2223109A1 (en) Use of antisense oligonucleotides to il-6 receptor mrna to inhibit cellular proliferation
US5872007A (en) CAPL-specific oligonucleotides and methods of inhibiting metastatic cancer
EP1071764B1 (en) Antisense oligonucleotides for the inhibition of integrin alphav-subunit expression
EP0856052A1 (en) Antisense oligonucleotide chemotherapy for benign hyperplasia or cancer of the prostate
WO1998013072A1 (en) Compositions for and methods of treating multiple drug resistance
US7855183B2 (en) Methods of treatment of a bcl-2 disorder using bcl-2 antisense oligomers
WO1998049287A2 (en) Antisense oligonucleotides specific for thymidylate synthase
US20030176384A1 (en) Antisense oligonucleotides for the inhibition of integrin alphav -subunit expression
AU2003271376B2 (en) Antisense Oligonucleotides for the Inhibition of Integrin alpha-Subunit Expression
WO1994007538A1 (en) ANTISENSE OLIGONUCLEOTIDES TO B-m^_y^_b^_ PROTO-ONCOGENE

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AU CA CN JP KP NZ US

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): AT BE CH DE DK ES FI FR GB GR IE IT LU MC NL PT SE

DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
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: 2239976

Country of ref document: CA

Ref country code: CA

Ref document number: 2239976

Kind code of ref document: A

Format of ref document f/p: F

WWE Wipo information: entry into national phase

Ref document number: 1996935879

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 1996935879

Country of ref document: EP

WWW Wipo information: withdrawn in national office

Ref document number: 1996935879

Country of ref document: EP