WO1998046272A1 - Antisense oligonucleotide compositions and methods for the modulation of activating protein 1 - Google Patents

Antisense oligonucleotide compositions and methods for the modulation of activating protein 1 Download PDF

Info

Publication number
WO1998046272A1
WO1998046272A1 PCT/US1998/007386 US9807386W WO9846272A1 WO 1998046272 A1 WO1998046272 A1 WO 1998046272A1 US 9807386 W US9807386 W US 9807386W WO 9846272 A1 WO9846272 A1 WO 9846272A1
Authority
WO
WIPO (PCT)
Prior art keywords
seq
oligonucleotide
oligonucleotides
nucleic acid
strandedness
Prior art date
Application number
PCT/US1998/007386
Other languages
French (fr)
Inventor
Nicholas M. Dean
Robert Mckay
Loren Miraglia
Brenda Baker
Original Assignee
Isis Pharmaceuticals, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Isis Pharmaceuticals, Inc. filed Critical Isis Pharmaceuticals, Inc.
Priority to AU69688/98A priority Critical patent/AU6968898A/en
Priority to EP98915526A priority patent/EP0975371A4/en
Publication of WO1998046272A1 publication Critical patent/WO1998046272A1/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
    • C12N15/1135Non-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 oncogenes or tumor suppressor genes
    • 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
    • C12N2310/31Chemical structure of the backbone
    • C12N2310/318Chemical structure of the backbone where the PO2 is completely replaced, e.g. MMI or formacetal
    • C12N2310/3181Peptide nucleic acid, PNA
    • 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
    • C12N2310/32Chemical structure of the sugar
    • C12N2310/3212'-O-R Modification
    • 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
    • C12N2310/32Chemical structure of the sugar
    • C12N2310/3222'-R Modification
    • 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
    • C12N2310/33Chemical structure of the base
    • 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
    • C12N2310/33Chemical structure of the base
    • C12N2310/334Modified C
    • 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
    • C12N2310/33Chemical structure of the base
    • C12N2310/335Modified T or U

Definitions

  • the present invention provides compositions and methods for modulating levels of the c-fos and c-jun genes, which encode the c-Fos and c-Jun subunits of AP-1, respectively.
  • AP-1 or transcription factor activating protein 1
  • AP-1 is a heterogenous mixture of heterodimers of several related protein subunits including, in addition to c-Fos and c-Jun, FosB, Fra-1, Fra-2, c-Jun,
  • AP-1 has been implicated in abnormal cell proliferation and tumor formation, events that thus might be controlled by modulating the expression of c-fos and/or c-jun .
  • the invention is further directed to therapeutic, diagnostic, and research based reagents and methods for evaluating and treating disease states or disorders which result from and/or respond positively to modulation of one or more AP-1 subunits.
  • disease states and disorders include those involving the hyperproliferation of cells such as, e . g. , a tumor (neoplasm) or malignant cancer.
  • Inhibition of AP-1 -mediated hyperproliferation of cells, and corresponding prophylactic, palliative and therapeutic effects result from treatment with the oligonucleotides of the invention.
  • Transcription factors play a central role in the expression of specific genes upon stimulation by extracellular signals, thereby regulating a complex array of biological processes.
  • AP-1 activating protein-1
  • cytokines cytokines
  • tumor promoters tumor promoters
  • carcinogens cytokines
  • cytokines exert their function by binding to specific cell surface receptors.
  • Receptor occupancy triggers a signal transduction cascade to the nucleus.
  • transcription factors such as AP-1 execute long term responses to the extracellular factors by modulating gene expression.
  • Such changes in cellular gene expression lead to DNA synthesis, and eventually the formation of differentiated derivatives (Angel and Karin, Biochi . Biophys . Acta , 1991, 1072, 129) .
  • AP-1 denotes one member of a family of related heterodimeric transcription factor complexes found in eukaryotic cells or viruses.
  • AP-1 denotes one member of a family of related heterodimeric transcription factor complexes found in eukaryotic cells or viruses.
  • AP-1 specifically refers to the heterodimer formed of c-Fos and c-Jun (Angel and Herrlich, Chapter 1, and Schuermann, Chapter 2 in : The FOS and JUN Families of Proteins , Angel and Herrlich, eds., pp. 3-35, CRC Press, Boca Raton, FL, 1994; Bohmann et al . , Science, 1987, 238, 1386; Angel et al . ,
  • MAP Mitogen-activated protein
  • PKC protein kinase C
  • MEK1 and MEK2 The phosphorylation of proteins plays a key role in the transduction of extracellular signals into the cell.
  • Mitogen-activated protein (MAP) kinases enzymes which effect such phosphorylations are targets for the action of growth factors, hormones, and other agents involved in cellular metabolism, proliferation and differentiation (Cobb et al . , J. Biol . Chem . , 1995, 270, 14843).
  • MAP kinases are themselves activated by phosphorylation catalyzed by, e . g. , receptor tyrosine kinases, G protein-coupled receptors, protein kinase C (PKC) , and the apparently MAP kinase dedicated kinases MEK1 and MEK2.
  • MAP kinases include, but are not limited to, ERK1 , ERK2 , two isoforms of ERK3 , ERK4 (ERK stands for "extracellular signal -regulated protein kinase) , Jun N-terminal kinases/stress-activated protein kinases (JNKs/SAPKs) , p38/HOGl, p57 MAP kinases, MKK3 (MAP kinase kinase 3) and MKK4 (MAP kinase kinase 4, also known as SAPK/ERK kinase (SEK) or JNK kinase (JNKK) ) (Cobb et al .
  • ERK extracellular signal -regulated protein kinase
  • JNKs/SAPKs Jun N-terminal kinases/stress-activated protein kinases
  • MKK3 MAP kinase kinase 3
  • MAP kinases are involved in a variety of signal transduction pathways (sometimes overlapping and sometimes parallel) that function to convey extracellular stimuli to protooncogene products to modulate cellular proliferation and/or differentiation.
  • One of the signal transduction pathways involves the MAP kinases Jun kinase 1 and Jun kinase 2 which are responsible for the phosphorylation of specific sites (Serine 63 and Serine 73) on c-Jun. Phosphorylation of these sites potentiates the ability of AP-1 to activate transcription (Binetruy et al . , Na ture, 1991, 351 , 122; Smeal et al . , Nature, 1991, 354 , 494) . At least one human leukemia oncogene has been shown to enhance Jun N-terminal Kinase (JNK) function (Raitano et al . , Proc . Na tl . Acad .
  • JNK Jun N-terminal Kinase
  • Cellular hyperproliferation in an animal can have several outcomes. Hyperproliferating cells might be attacked and killed by the animal's immune system before a tumor can form. Tumors are abnormal growths resulting from the hyperproliferation of cells. Cells that proliferate to excess but stay put form benign tumors, which can typically be removed by local surgery. In contrast, malignant tumors or cancers comprise cells that are capable of undergoing metastasis, i . e .
  • oligonucleotides of the invention it has surprisingly been discovered that several genes encoding enzymes required for metastasis are positively regulated by AP-1. Accordingly, inhibition of expression of c-fos and/or c-jun serves as a means to modulate the metastasis of malignant tumors.
  • a method of modulating one or more metastatic events using the oligonucleotides of the invention is thus herein provided.
  • Liu et al . ⁇ Ann . Neurol . , 1994, 36, 566) describe the suppression of c-fos by intraventricular infusion of an antisense oligodeoxynucleotide targeted to c-fos mRNA.
  • Gillardon et al describe the topical application of c-fos antisense oligodeoxynucleotides to the rat spinal cord ( Eur . J. Neurosci . , 1994, 6, 880) ultraviolet (UV) -exposed rat eyes (Bri tish J. Ophthal . , 1995, 79 , 277) and UV- irradiated rat skin ( Carcinogenesis , 1995, 16, 1853) .
  • U.S. Patent No. 5,602,156 which issued February 11, 1997, to Kohn et al . , discloses non-oligonucleotide compositions and methods for inhibiting the expression of two metalloproteinases, MMP-1 and MMP-2.
  • International Publication Number WO 95/02051 published January 19, 1995, discloses antisense oligonucleotides targeted to the mRNA of c-fos and c-jun .
  • compositions and methods of the invention overcome these limitations. Further objectives of the invention are apparent from the present disclosure.
  • oligonucleotides are provided which specifically hybridize with nucleic acids encoding c-Fos or c-Jun.
  • Certain oligonucleotides of the invention are designed to bind either directly to mRNA transcribed from, or to a selected DNA portion of, the respective gene, thereby modulating the amount of protein translated from a c-fos or c-jun mRNA and/or the amount of mRNA transcribed from a c-fos or c-jun gene, respectively.
  • Such modulation can, in turn, effect the modulation of enzymes and cellular processes involved in the metastasis of malignant cells.
  • Oligonucleotides may comprise nucleotide sequences sufficient in identity and number to effect specific hybridization with a particular nucleic acid. Such oligonucleotides are commonly described as “antisense.” Antisense oligonucleotides are commonly used as research reagents, diagnostic aids, and therapeutic agents.
  • c-fos and c-jun genes encoding the c-Fos and c-Jun proteins, respectively, are particularly amenable to this approach.
  • modulation of the expression of c-fos and/or c-jun leads to modulation of AP-1, and, accordingly, modulation of cellular proliferation.
  • Such modulation is desirable for treating or modulating various hyperproliferative disorders or diseases, such as various cancers.
  • Such inhibition is further desirable for preventing or modulating the development of such diseases or disorders in an animal suspected of being, or known to be, prone to such diseases or disorders.
  • modulation of one subunit can be combined with modulation of the subunit of AP- 1 in order to achieve a requisite degree of effect upon AP-1- mediated transcription.
  • Methods of modulating the expression of c-Fos or c-Jun proteins comprising contacting animals with oligonucleotides specifically hybridizable with a c-fos or c-jun gene, respectively, are herein provided. These methods are believed to be useful both therapeutically and diagnostically as a consequence of the association between AP-1 expression and cellular proliferation. These methods are also useful as tools, for example, in the detection and determination of the role of AP-1 protein expression in various cell functions and physiological processes and conditions, and for the diagnosis of conditions associated with such expression and activation.
  • the present invention also comprises methods of inhibiting AP-1-mediated transcriptional activation using the oligonucleotides of the invention.
  • Methods of treating conditions in which abnormal or excessive AP-1-mediated transcriptional activation and cellular proliferation occur are also provided. These methods employ the oligonucleotides of the invention and are believed to be useful both therapeutically and as clinical research and diagnostic tools.
  • the oligonucleotides of the present invention may also be used for research purposes.
  • the specific hybridization exhibited by the oligonucleotides of the present invention may be used for assays, purifications, cellular product preparations and in other methodologies which may be appreciated by persons of ordinary skill in the art .
  • Methods comprising contacting animals with oligonucleotides specifically hybridizable with nucleic acids encoding c-Fos or c-Jun proteins are herein provided. Such methods can be used to modulate or detect the expression of c-fos or c-jun genes and are thus believed to be useful both therapeutically and diagnostically.
  • the methods disclosed herein are also useful, for example, as clinical research tools in the detection and determination of the role of AP-1-mediated gene expression in various immune system functions and physiological processes and conditions, and for the diagnosis of conditions associated with their expression.
  • the specific hybridization exhibited by the oligonucleotides of the present invention may be used for assays, purifications, cellular product preparations and in other methodologies which may be appreciated by persons of ordinary skill in the art.
  • the oligonucleotides of this invention specifically hybridize to nucleic acids encoding c-Fos or c- Jun, sandwich and other assays can easily be constructed to exploit this fact.
  • Detection of specific hybridization of an oligonucleotide of the invention with a nucleic acid encoding a c-Fos or c-Jun protein present in a sample can routinely be accomplished. Such detection may include detectably labeling an oligonucleotide of the invention by enzyme conjugation, radiolabeling or any other suitable detection system.
  • a number of assays may be formulated employing the present invention, which assays will commonly comprise contacting a tissue or cell sample with a detectably labeled oligonucleotide of the present invention under conditions selected to permit hybridization and measuring such hybridization by detection of the label, as is appreciated by those of ordinary skill in the art.
  • the present invention employs oligonucleotides for use in antisense inhibition of the function of RNA and DNA encoding a c-Fos protein or a c-Jun protein.
  • the present invention also employs oligonucleotides which are designed to be specifically hybridizable to DNA or messenger RNA (mRNA) encoding such proteins and ultimately modulating the amount of such proteins transcribed from their respective genes.
  • mRNA messenger RNA
  • Such hybridization with mRNA interferes with the normal role of mRNA and causes a modulation of its function in cells.
  • the functions of mRNA to be interfered with include all vital functions such as translocation of the RNA to the site for protein translation, actual translation of protein from the RNA, splicing of the RNA to yield one or more mRNA species, and possibly even independent catalytic activity which may be engaged in by the RNA.
  • the overall effect of such interference with mRNA function is modulation of the expression of a c-Fos protein or a c-Jun protein.
  • modulation means either an increase (stimulation) or a decrease (inhibition) in the expression of a gene. In the context of the present invention, inhibition is the preferred form of modulation of gene expression.
  • oligonucleotide refers to an oligomer or polymer of ribonucleic acid or deoxyribonucleic acid. This term includes oligonucleotides composed of naturally-occurring nucleobases, sugars and covalent intersugar (backbone) linkages as well as oligonucleotides having non-naturally- occurring portions which function similarly. Such modified or substituted oligonucleotides are often preferred over native forms because of desirable properties such as, for example, enhanced cellular uptake, enhanced binding to target and increased stability in the presence of nucleases .
  • An oligonucleotide is a polymer of a repeating unit generically known as a nucleotide.
  • An unmodified (naturally occurring) nucleotide has three components: (1) a nitrogenous base linked by one of its nitrogen atoms to (2) a 5 -carbon cyclic sugar and (3) a phospha te, esterified to carbon 5 of the sugar.
  • the phosphate of a first nucleotide is also esterified to carbon 3 of the sugar of a second, adjacent nucleotide.
  • the "backbone” of an unmodified oligonucleotide consists of (2) and (3), that is, sugars linked together by phosphodiester linkages between the carbon 5 (5') position of the sugar of a first nucleotide and the carbon 3 (3') position of a second, adjacent nucleotide.
  • a “nucleoside” is the combination of (1) a nucleobase and (2) a sugar in the absence of (3) a phosphate moiety (Kornberg, A., DNA Replication, pp. 4-7,
  • the backbone of an oligonucleotide positions a series of bases in a specific order; the written representation of this series of bases, which is conventionally written in 5 ' to 3 ' order, is known as a nucleotide sequence.
  • the oligonucleotides in accordance with this invention preferably comprise from about 8 to about 30 nucleotides. It is more preferred that such oligonucleotides comprise from about 15 to 25 nucleotides. Oligonucleotides may comprise nucleotide sequences sufficient in identity and number to effect specific hybridization with a particular nucleic acid.
  • Antisense oligonucleotides which specifically hybridize to a portion of the sense strand of a gene are commonly described as “antisense.”
  • Antisense oligonucleotides are commonly used as research reagents, diagnostic aids, and therapeutic agents.
  • antisense oligonucleotides which are able to inhibit gene expression with extraordinar specificity, are often used by those of ordinary skill to elucidate the function of particular genes, for example to distinguish between the functions of various members of a biological pathway. This specific inhibitory effect has, therefore, been harnessed by those skilled in the art for research uses .
  • the specificity and sensitivity of oligonucleotides is also harnessed by those of skill in the art for therapeutic uses. For example, the following U.S.
  • U. S. Patent 5,135,917 provides antisense oligonucleotides that inhibit human interleukin-1 receptor expression.
  • U.S. Patent 5,098,890 is directed to antisense oligonucleotides complementary to the c-myb oncogene and antisense oligonucleotide therapies for certain cancerous conditions.
  • U.S. Patent 5,087,617 provides methods for treating cancer patients with antisense oligonucleotides.
  • U.S. Patent 5,166,195 provides oligonucleotide inhibitors of Human Immunodeficiency Virus (HIV).
  • HAV Human Immunodeficiency Virus
  • Patent 5,004,810 provides oligomers capable of hybridizing to herpes simplex virus Vmw65 mRNA and inhibiting replication.
  • U.S. Patent 5,194,428 provides antisense oligonucleotides having antiviral activity against influenzavirus .
  • U.S. Patent 4,806,463 provides antisense oligonucleotides and methods using them to inhibit HTLV-III replication.
  • U.S. Patent 5,286,717 provides oligonucleotides having a complementary base sequence to a portion of an oncogene.
  • U.S. Patent 5,276,019 and U.S. Patent 5,264,423 are directed to phosphorothioate oligonucleotide analogs used to prevent replication of foreign nucleic acids in cells.
  • U.S. Patent 4,689,320 is directed to antisense oligonucleotides as antiviral agents specific to cytomegalovirus (CMV) .
  • CMV cytomegalovirus
  • U.S. Patent 5,098,890 provides oligonucleotides complementary to at least a portion of the mRNA transcript of the human c-myb gene.
  • U.S. Patent 5,242,906 provides antisense oligonucleotides useful in the treatment of latent Epstein- Barr virus (EBV) infections.
  • EBV Epstein- Barr virus
  • Targeting an oligonucleotide to the associated nucleic acid, in the context of this invention, is a multistep process. The process usually begins with the identification of a nucleic acid sequence whose function is to be modulated. This may be, for example, a cellular gene (or mRNA transcribed from the gene) whose expression is associated with a particular disorder or disease state, or a foreign nucleic acid from an infectious agent.
  • the target is a cellular gene ( c-fos or c- un) encoding a subunit of AP-1, for which modulation is desired in certain instances.
  • the targeting process also includes determination of a region (or regions) within this gene for the oligonucleotide interaction to occur such that the desired effect, either detection or modulation of expression of the protein, will result. Once the target region have been identified, oligonucleotides are chosen which are sufficiently complementary to the target, i . e . , hybridize sufficiently well and with sufficient specificity to give the desired effect.
  • the region of the 5' Cap a specialized structure that at least partially mediates ribosome binding; the 5' untranslated (noncoding) region (hereinafter, the "5' -UTR”); the translation initiation codon region (hereinafter, the "tIR”); the open reading frame
  • the ORF the translation termination codon region
  • the tTR translation termination codon region
  • the 3' untranslated (noncoding) region hereinafter, the "3' -UTR”
  • these regions are arranged in a typical messenger RNA molecule in the following order (left to right, 5' to 3') : 5' Cap, 5'- UTR, tIR, ORF, tTR, 3' -UTR, poly A tail.
  • ORFs contain one or more sequences, known as "introns,” which are excised from a transcript before it is translated; the expressed (unexcised) portions of the ORF are referred to as “exons” (Alberts et al . , Molecular Biology of the Cell , pp. 331-332 and 411-415, Garland Publishing Inc., New York, 1983) .
  • exons the sequences of the ORF.
  • many eukaryotic ORFs are a thousand nucleotides or more in length, it is often convenient to subdivide the ORF into, e . g. , the 5' ORF region, the central ORF region, and the 3' ORF region.
  • an ORF contains one or more sites that may be targeted due to some functional significance in vivo .
  • sites include intragenic stem-loop structures (see, e . g. , U.S. Patent No. 5,512,438) and, in unprocessed mRNA molecules, intron/exon splice sites.
  • one preferred intragenic site is the region encompassing the translation initiation codon of the open reading frame (ORF) of the gene.
  • the translation initiation codon is typically 5 ' -AUG (in transcribed mRNA molecules; 5'- ATG in the corresponding DNA molecule)
  • the translation initiation codon is also referred to as the "AUG codon,” the “start codon” or the "AUG start codon.”
  • a minority of genes have a translation initiation codon having the RNA sequence 5'-GUG, 5'-UUG or 5'-CUG, and 5'-AUA, 5 ' -ACG and 5'-CUG have been shown to function in vivo .
  • 5 ' -UUU functions as a translation initiation codon in vi tro (Brigstock et al .
  • translation initiation codon and “start codon” can encompass many codon sequences, even though the initiator amino acid in each instance is typically methionine (in eukaryotes) or formylmethionine (prokaryotes) . It is also known in the art that eukaryotic and prokaryotic genes may have two or more alternative start codons, any one of which may be preferentially utilized for translation initiation in a particular cell type or tissue, or under a particular set of conditions, in order to generate related polypeptides having different amino terminal sequences (Markussen et al . , Development , 1995, 121 , 3723; Gao et al . , Cancer Res .
  • start codon and “translation initiation codon” refer to the codon or codons that are used in vivo to initiate translation of an mRNA molecule transcribed from a gene encoding a c-Fos or c- Jun protein, regardless of the sequence (s) of such codons.
  • a translation termination codon (or "stop codon”) of a gene may have one of three sequences, i.e., 5'-UAA, 5'-UAG and 5'-UGA (the corresponding DNA sequences are 5'-TAA, 5' -TAG and 5'-TGA, respectively).
  • start codon region and “translation initiation region” refer to a portion of such an mRNA or gene that encompasses from about 25 to about 50 contiguous nucleotides in either direction (i.e., 5' or 3 ' ) from a translation initiation codon.
  • stop codon region and “translation termination region” refer to a portion of such an mRNA or gene that encompasses from about 25 to about 50 contiguous nucleotides in either direction (i.e., 5' or 3') from a translation termination codon.
  • translation termination region refers to a portion of such an mRNA or gene that encompasses from about 25 to about 50 contiguous nucleotides in either direction (i.e., 5' or 3') from a translation termination codon.
  • oligonucleotide includes oligonucleotides composed of naturally-occurring nucleobases, sugars and covalent intersugar (backbone) linkages as well as oligonucleotides having non-naturally-occurring portions which function similarly. Such modified or substituted oligonucleotides may be preferred over native forms because of desirable properties such as, for example, enhanced cellular uptake, enhanced binding to target and increased stability in the presence of nucleases .
  • modified oligonucleotides envisioned for this invention include those containing phosphorothioates , phosphotriesters , methyl phosphonates, short chain alkyl or cycloalkyl intersugar linkages or short chain heteroatomic or heterocyclic intersugar linkages.
  • oligonucleotides with phosphorothioates and those with CH 2 -NH-0-CH 2 , CH 2 - N(CH 3 )-0-CH 2 [known as a methylene (methylimino) or MMI backbone], CH 2 -0-N (CH 3 ) -CH 2 , CH 2 -N (CH 3 ) -N (CH 3 ) -CH 2 and 0-N (CH 3 ) - CH 2 -CH 2 backbones, wherein the native phosphodiester backbone is represented as 0-P-0-CH 2 ) .
  • oligonucleotides having morpholino backbone structures (Summerton and Weller, U.S. Patent 5,034,506).
  • oligonucleotides with NR-C (*) -CH 2 -CH 2 , CH 2 -NR- C(*)-CH 2 , CH 2 -CH 2 -NR-C(*) , C ( * ) -NR-CH 2 -CH 2 and CH 2 -C ( * ) -NR-CH 2 backbones, wherein "*" represents 0 or S (known as amide backbones; DeMesmaeker et al . , WO 92/20823, published November 26, 1992) .
  • the phosphodiester backbone of the oligonucleotide is replaced with a polyamide backbone, the nucleobases being bound directly or indirectly to the aza nitrogen atoms of the polyamide backbone (Nielsen et al . , Science, 1991, 254 , 1497; U.S. Patent No. 5,539,082).
  • the oligonucleotides of the invention may additionally or alternatively include nucleobase modifications or substitutions.
  • unmodified or “natural” nucleobases include adenine (A) , guanine (G) , thymine (T) , cytosine (C) and uracil (U) .
  • Modified nucleobases include nucleobases found only infrequently or transiently in natural nucleic acids, e . g.
  • hypoxanthine 6-methyladenine , 5- methylcytosine, 5-hydroxy ⁇ nethylcytosine (HMC) , glycosyl HMC and gentiobiosyl HMC, as well synthetic nucleobases, e.g., 2- aminoadenine, 2-thiouracil , 2-thiothymine , 5-bromouracil , 5- hydroxymethyluracil, 8-azaguanine, 7-deazaguanine , N 6 (6- aminohexyl) adenine and 2 , 6-diaminopurine (Kornberg, A., DNA Replication, pp. 75-77, W.H. Freeman & Co., San Francisco, 1980; Gebeyehu, G. , et al . , Nucleic Acids Res . , 1987, 15 , 4513) .
  • nucleobases e.g., 2- aminoadenine, 2-thiouracil , 2-thiothymine ,
  • oligonucleotides of the invention may additionally or alternatively comprise substitutions of the sugar portion of the individual nucleotides.
  • oligonucleotides may also have sugar mimetics such as cyclobutyls in place of the pentofuranosyl group.
  • modified oligonucleotides may contain one or more substituted sugar moieties comprising one of the following at the 2' position: OH, SH, SCH 3 , F, OCN, OCH 3 OCH 3 , OCH 3 0 (CH 2 ) n CH 3 , 0(CH 2 ) n NH 2 or 0(CH 2 ) n CH 3 where n is from 1 to about 10; C to C 10 lower alkyl, alkoxyalkoxy, substituted lower alkyl, alkaryl or aralkyl; Cl ; Br; CN; CF 3 ; OCF 3 ; 0-, S-, or N-alkyl; O- , S-, or N-alkenyl; SOCH 3 ; S0 2 CH 3 ; ON0 2 ; N0 2 ; N 3 ; NH 2 ; heterocycloalkyl ; heterocycloalkaryl ; aminoalkylamino; polyalkylamino; substituted silyl; an
  • a preferred modification, particularly for orally deliverable pharmaceutical compositions, is 2'- methoxyethoxy [2 ' -0-CH 2 CH 2 OCH 3 , also known as 2'-0-(2- methoxyethyl) ] (Martin et al . , Helv. Chim . Acta , 1995, 78 , 486).
  • Other preferred modifications include 2 ' -methoxy (2'- 0-CH 3 ) , 2'-propoxy (2 ' -OCH 2 CH 2 CH 3 ) and 2'-fluoro (2'-F).
  • oligonucleotide Similar modifications may also be made at other positions on the oligonucleotide, particularly the 3' position of the sugar on the 3' terminal nucleotide and the 5' position of 5' terminal nucleotide.
  • the 5' and 3' termini of an oligonucleotide may also be modified to serve as points of chemical conjugation of, e.g., lipophilic moieties (see immediately subsequent paragraph) , intercalating agents (Kuyavin et al . , WO 96/32496, published October 17, 1996; Nguyen et al . , U.S. Patent No. 4,835,263, issued May 30, 1989) or hydroxyalkyl groups (Helene et al . , WO 96/34008, published October 31, 1996) .
  • effector group is a chemical moiety that is capable of carrying out a particular chemical or biological function.
  • effector groups include, but are not limited to, an RNA cleaving group, a reporter group, an intercalator, a group for improving the pharmacokinetic properties of an oligonucleotide, or a group for improving the pharmacodynamic properties of an oligonucleotide and other substituents having similar properties.
  • a variety of chemical linkers may be used to conjugate an effector group to an oligonucleotide of the invention.
  • U.S. Patent No. 5,578,718 to Cook et al discloses methods of attaching an alkylthio linker, which may be further derivatized to include additional groups, to ribofuranosyl positions, nucleosidic base positions, or on internucleoside linkages. Additional methods of conjugating oligonucleotides to various effector groups are known in the art; see, e . g. , Protocols for
  • Oligonucleotide Conjugates (Methods in Molecular Biology, Volume 26) Agrawal , S., ed. , Humana Press, Totowa, NJ, 1994.
  • oligonucleotides of the invention involves chemically linking to the oligonucleotide one or more lipophilic moieties which enhance the cellular uptake of the oligonucleotide.
  • lipophilic moieties may be linked to an oligonucleotide at several different positions on the oligonucleotide. Some preferred positions include the 3' position of the sugar of the 3' terminal nucleotide, the 5' position of the sugar of the 5' terminal nucleotide, and the 2' position of the sugar of any nucleotide.
  • the N 6 position of a purine nucleobase may also be utilized to link a lipophilic moiety to an oligonucleotide of the invention (Gebeyehu et al . , Nucleic Acids Res . , 1987, 15 , 4513).
  • lipophilic moieties include but are not limited to a cholesteryl moiety (Letsinger et al . , Proc . Natl . Acad . Sci . USA, 1989, 86, 6553), cholic acid (Manoharan et al . , Bioorg . Med . Chem . Let . , 1994, 4, 1053), a thioether, e . g. , hexyl-S- tritylthiol (Manoharan et al . , Ann . N. Y. Acad . Sci . , 1992,
  • a phospholipid e.g., di-hexadecyl-rac-glycerol or triethylammonium 1,2-di-O- hexadecyl-rac-glycero-3 -H-phosphonate
  • a polyamine or a polyethylene glycol chain Manoharan et al . , Nucleosides & Nucleotides , 1995, 14 , 969
  • adamantane acetic acid Manoharan et al . ,
  • the oligonucleotides of the invention may additionally or alternatively be prepared to be delivered in a "prodrug" form.
  • prodrug indicates a therapeutic agent that is prepared in an inactive form that is converted to an active form ( i . e . , drug) within the body or cells thereof by the action of endogenous enzymes or other chemicals and/or conditions.
  • prodrug versions of the oligonucleotides of the invention are prepared as SATE [ (S- acetyl-2-thioethyl) phosphate] derivatives according to the methods disclosed in WO 93/24510 to Gosselin et al . , published December 9, 1993.
  • the present invention also includes oligonucleotides that are substantially chirally pure with regard to particular positions within the oligonucleotides.
  • substantially chirally pure oligonucleotides include, but are not limited to, those having phosphorothioate linkages that are at least 75% Sp or Rp (Cook et al . , U.S. Patent No. 5,587,361, issued December 24, 1996) and those having substantially chirally pure (Sp or Rp) alkylphosphonate, phosphoamidate or phosphotriester linkages (Cook, U.S. Patent No. 5,212,295, issued May 18, 1993; Cook, U.S. Patent No. 5,521,302, issued May 28, 1996) .
  • the present invention also includes oligonucleotides which are chimeric oligonucleotides.
  • "Chimeric" oligonucleotides or “chimeras,” in the context of this invention, are oligonucleotides which contain two or more chemically distinct regions, each made up of at least one nucleotide. These oligonucleotides typically contain at least one region wherein the oligonucleotide is modified so as to confer upon the oligonucleotide increased resistance to nuclease degradation, increased cellular uptake, and/or increased binding affinity for the target nucleic acid.
  • An additional region of the oligonucleotide may serve as a substrate for enzymes capable of cleaving RNA: DNA or RNA: RNA hybrids.
  • RNase H is a cellular endonuclease which cleaves the RNA strand of an RNA: DNA duplex. Activation of RNase H, therefore, results in cleavage of the RNA target, thereby greatly enhancing the efficiency of antisense inhibition of gene expression.
  • RNA target can be routinely detected by gel electrophoresis and, if necessary, associated nucleic acid hybridization techniques known in the art.
  • chimeras may be "gapmers,” i . e . , oligonucleotides in which a central portion (the "gap") of the oligonucleotide serves as a substrate for, e . g.
  • wings are modified in such a fashion so as to have greater affinity for the target RNA molecule but are unable to support nuclease activity (e.g., 2'-fluoro- or 2 ' -methoxyethoxy substituted).
  • Other chimeras include "wingmers,” that is, oligonucleotides in which the 5' portion of the oligonucleotide serves as a substrate for, e . g. , RNase H, whereas the 3' portion is modified in such a fashion so as to have greater affinity for the target RNA molecule but is unable to support nuclease activity ( e . g.
  • oligonucleotides used in accordance with this invention may be conveniently and routinely made through the well-known technique of solid phase synthesis. Equipment for such synthesis is sold by several vendors including, for example, Applied Biosystems (Foster City, CA) . Any other means for such synthesis known in the art may additionally or alternatively be employed. It is also known to use similar techniques to prepare other oligonucleotides such as the phosphorothioates and alkylated derivatives. Teachings regarding the synthesis of particular modified oligonucleotides are hereby incorporated by reference from the following U.S.
  • the oligonucleotides of the present invention can be utilized as therapeutic compounds, diagnostic tools and as research reagents and kits.
  • the term "therapeutic uses" is intended to encompass prophylactic, palliative and curative uses wherein the oligonucleotides of the invention are contacted with animal cells either in vivo or ex vivo .
  • a therapeutic use includes incorporating such cells into an animal after treatment with one or more oligonucleotides of the invention.
  • an animal suspected of having a disease or disorder which can be treated or prevented by modulating the expression or activity of a c-Fos or c-Jun protein is, for example, treated by administering oligonucleotides in accordance with this invention.
  • the oligonucleotides of the invention can be utilized in pharmaceutical compositions by adding an effective amount of an oligonucleotide to a suitable pharmaceutically acceptable diluent or carrier.
  • Workers in the field have identified antisense, triplex and other oligonucleotide compositions which are capable of modulating expression of genes implicated in viral, fungal and metabolic diseases.
  • Antisense oligonucleotides have been safely administered to humans and several clinical trials are presently underway. It is thus established that oligonucleotides can be useful therapeutic instrumentalities that can be configured to be useful in treatment regimes for treatment of cells, tissues and animals, especially humans.
  • the oligonucleotides of the present invention can be further used to detect the presence of c-fos - or c-jun- specific nucleic acids in a cell or tissue sample.
  • radiolabeled oligonucleotides can be prepared by 32 P labeling at the 5' end with polynucleotide kinase (Sambrook et al . , Molecular Cloning. A Labora tory Manual , Vol. 2, p. 10.59, Cold Spring Harbor Laboratory Press, 1989).
  • Radiolabeled oligonucleotides are then contacted with cell or tissue samples suspected of containing c-fos or c-jun message RNAs (and thus c-Fos or c-Jun proteins) , and the samples are washed to remove unbound oligonucleotide. Radioactivity remaining in the sample indicates the presence of bound oligonucleotide, which in turn indicates the presence of nucleic acids complementary to the oligonucleotide, and can be quantitated using a scintillation counter or other routine means. Expression of nucleic acids encoding these proteins is thus detected.
  • Radiolabeled oligonucleotides of the present invention can also be used to perform autoradiography of tissues to determine the localization, distribution and quantitation of c-Fos or c-Jun proteins for research, diagnostic or therapeutic purposes.
  • tissue sections are treated with radiolabeled oligonucleotide and washed as described above, then exposed to photographic emulsion according to routine autoradiography procedures.
  • the emulsion when developed, yields an image of silver grains over the regions expressing a c-Fos or c-Jun gene.
  • Quantitation of the silver grains permits detection of the expression of mRNA molecules encoding these proteins and permits targeting of oligonucleotides to these areas.
  • Analogous assays for fluorescent detection of expression of c-fos or c-jun nucleic acids can be developed using oligonucleotides of the present invention which are conjugated with fluorescein or other fluorescent tags instead of radiolabeling. Such conjugations are routinely accomplished during solid phase synthesis using fluorescently-labeled amidites or controlled pore glass (CPG) columns. Fluorescein-labeled amidites and CPG are available from, e . g. , Glen Research (Sterling, VA) .
  • kits for detecting the presence or absence of expression of a c-Fos and/or c-Jun protein may also be prepared.
  • kits include an oligonucleotide targeted to an appropriate gene, i.e., a gene encoding a c-Fos or c-Jun protein.
  • Hybridization of the oligonucleotides of the invention with a nucleic acid encoding a c-Fos or c-Jun protein can be detected by means known in the art . Such means may include conjugation of an enzyme to the oligonucleotide, radiolabelling of the oligonucleotide or any other suitable detection systems. Kits for detecting the presence or absence of a c-Fos or c-Jun protein may also be prepared.
  • hybridization means hydrogen bonding, which may be Watson-Crick, Hoogsteen or reversed Hoogsteen hydrogen bonding, between complementary nucleotides.
  • adenine and thymine are complementary nucleobases which pair through the formation of hydrogen bonds.
  • “Complementary,” as used herein, refers to the capacity for precise pairing between two nucleotides. For example, if a nucleotide at a certain position of an oligonucleotide is capable of hydrogen bonding with a nucleotide at the same position of a DNA or RNA molecule, then the oligonucleotide and the DNA or RNA are considered to be complementary to each other at that position.
  • oligonucleotide and the DNA or RNA are complementary to each other when a sufficient number of corresponding positions in each molecule are occupied by nucleotides which can hydrogen bond with each other.
  • “specifically hybridizable” and “complementary” are terms which are used to indicate a sufficient degree of complementarity or precise pairing such that stable and specific binding occurs between the oligonucleotide and the DNA or RNA target. It is understood in the art that an oligonucleotide need not be 100% complementary to its target DNA sequence to be specifically hybridizable.
  • An oligonucleotide is specifically hybridizable when binding of the oligonucleotide to the target DNA or RNA molecule interferes with the normal function of the target DNA or RNA to cause a decrease or loss of function, and there is a sufficient degree of complementarity to avoid non-specific binding of the oligonucleotide to non-target sequences under conditions in which specific binding is desired, i.e., under physiological conditions in the case of in vivo assays or therapeutic treatment, or in the case of in vi tro assays, under conditions in which the assays are performed.
  • oligonucleotide in accordance with the invention commonly in a pharmaceutically acceptable carrier, in doses ranging from 0.01 ug to 100 g per kg of body weight depending on the age of the patient and the severity of the disorder or disease state being treated.
  • the treatment regimen may last for a period of time which will vary depending upon the nature of the particular disease or disorder, its severity and the overall condition of the patient, and may extend from once daily to once every 20 years. Following treatment, the patient is monitored for changes in his/her condition and for alleviation of the symptoms of the disorder or disease state .
  • the dosage of the oligonucleotide may either be increased in the event the patient does not respond significantly to current dosage levels, or the dose may be decreased if an alleviation of the symptoms of the disorder or disease state is observed, or if the disorder or disease state has been abated.
  • treatment regimen is meant to encompass therapeutic, palliative and prophylactic modalities.
  • a patient may be treated with conventional chemotherapeutic agents, particularly those used for tumor and cancer treatment.
  • chemotherapeutic agents include but are not limited to daunorubicin, daunomycin, dactinomycin, doxorubicin, epirubicin, idarubicin, esorubicin, bleomycin, mafosfamide, ifosfamide, cytosine arabinoside, bis-chloroethylnitrosurea, busulfan, mitomycin C, actinomycin D, mithramycin, prednisone, hydroxyprogesterone, testosterone, tamoxifen, dacarbazine, procarbazine, hexamethylmelamine, pentamethylmelamine, mitoxantrone, amsacrine, chlorambucil , methylcyclohexylnitrosurea, nitrogen mustards, melphalan, cyclophosphamide, 6-mercaptopurine, 6-thioguanine, cytarabine (CA) , 5-azacytidine, 5-a
  • chemotherapeutic agents may be used individually (e . g . , 5-FU and oligonucleotide), sequentially (e . g. , 5-FU and oligonucleotide for a period of time followed by MTX and oligonucleotide) , or in combination with one or more other such chemotherapeutic agents ( e . g.
  • a first antisense oligonucleotide targeted to c-fos is used in combination with a second antisense oligonucleotide targeted to c-jun in order to modulate AP-1 molecules to a more extensive degree than can be achieved when either oligonucleotide used individually.
  • oligonucleotide is administered in maintenance doses, ranging from 0.01 ug to 100 g per kg of body weight, once or more daily, to once every 20 years.
  • prophylactic effects may be achieved by administration of preventative doses, ranging from 0.01 ug to 100 g per kg of body weight, once or more daily, to once every 20 years.
  • compositions of the present invention may be administered in a number of ways depending upon whether local or systemic treatment is desired and upon the area to be treated.
  • administration indicates the topical (including ophthalmic, vaginal, rectal, intranasal, transdermal) , oral or parenteral contacting of an oligonucleotide, or pharmaceutical composition comprising an oligonucleotide, with cells, tissues or organs of a mammal including a human.
  • Parenteral administration includes intravenous drip; subcutaneous, intraperitoneal, intravitreal or intramuscular injection; and intrathecal or intraventricular administration as herein described.
  • Formulations for topical administration may include transdermal patches, ointments, lotions, creams, gels, drops, suppositories, sprays, liquids and powders.
  • Conventional pharmaceutical carriers, nucleic acid carriers, aqueous, powder or oily bases, thickeners and the like may be necessary or desirable in certain instances.
  • Topical administration also includes the delivery of oligonucleotides into the epidermis of a mammal by electroporation (Zewert et al . , WO 96/39531, published December 12, 1996).
  • compositions for oral administration include powders or granules, suspensions or solutions in water or non-aqueous media, capsules, sachets or tablets. Thickeners, flavoring agents, diluents, emulsifiers, dispersing aids or binders may be desirable.
  • Intravitreal injection for the direct delivery of drug to the vitreous humor of a mammalian eye, is described in U.S. Patent No. 5,595,978, which issued January 21, 1997, and which is assigned to the same assignee as the instant application, the contents of which are hereby incorporated by reference .
  • Intraluminal drug administration for the direct delivery of drug to an isolated portion of a tubular organ or tissue (e.g., such as an artery, vein, ureter or urethra), may be desired for the treatment of patients with diseases or conditions afflicting the lumen of such organs or tissues.
  • a catheter or cannula is surgically introduced by appropriate means.
  • a cannula is inserted thereinto via the external carotid artery.
  • a composition comprising the oligonucleotides of the invention is infused through the cannula or catheter into the isolated segment .
  • the infusion cannula or catheter is removed and flow within the tubular organ or tissue is restored by removal of the ligatures which effected the isolation of a segment thereof (Morishita et al . , Proc . Na tl . Acad . Sci . (U. S . A . ) , 1993, 90 , 8474) .
  • Antisense oligonucleotides may also be combined with a biocompatible matrix or carrier, such as a hydrogel material, and applied directly to vascular tissue in vivo (Rosenberg et al . , U.S. Patent No. 5,593,974, issued January 14, 1997).
  • Intraventricular drug administration for the direct delivery of drug to the brain of a patient, may be desired for the treatment of patients with diseases or conditions afflicting the brain.
  • a silicon catheter is surgically introduced into a ventricle of the brain of a human patient, and is connected to a subcutaneous infusion pump (Medtronic Inc., Minneapolis, MN) that has been surgically implanted in the abdominal region (Zimm et al . , Cancer Research, 1984, 44 , 1698; Shaw, Cancer, 1993, 72 (11 Suppl . ) , 3416).
  • the pump is used to inject the oligonucleotides and allows precise dosage adjustments and variation in dosage schedules with the aid of an external programming device.
  • the reservoir capacity of the pump is 18-20 mL and infusion rates may range from 0.1 mL/h to 1 mL/h.
  • the pump reservoir may be refilled at 3-10 week intervals. Refilling of the pump is accomplished by percutaneous puncture of the self-sealing septum of the pump.
  • Intrathecal drug administration for the introduction of drug into the spinal column of a patient may be desired for the treatment of patients with diseases of the central nervous system.
  • a silicon catheter is surgically implanted into the L3-4 lumbar spinal interspace of a human patient, and is connected to a subcutaneous infusion pump which has been surgically implanted in the upper abdominal region (Luer and Hatton, The Annals of Pharmacotherapy , 1993, 27, 912; Ettinger et al . , 1978, Cancer, 41 , 1270, 1978; Yaida et al . , Regul . Pept . , 1995, 59 , 193).
  • the pump is used to inject the oligonucleotides and allows precise dosage adjustments and variations in dose schedules with the aid of an external programming device.
  • the reservoir capacity of the pump is 18-20 mL, and infusion rates may vary from 0.1 mL/h to 1 mL/h.
  • the pump reservoir may be refilled at 3-10 week intervals. Refilling of the pump is accomplished by a single percutaneous puncture to the self-sealing septum of the pump.
  • oligonucleotides within the central nervous system may be followed according to known methods (Whitesell et al . , Proc . Natl . Acad. Sci . (USA) , 1993, 90 , 4665).
  • the silicon catheter is configured to connect the subcutaneous infusion pump to, e.g., the hepatic artery, for delivery to the liver
  • Infusion pumps may also be used to effect systemic delivery of oligonucleotides
  • colloidal dispersion systems may be used as delivery vehicles to enhance the in vivo stability of the oligonucleotides and/or to target the oligonucleotides to a particular organ, tissue or cell type.
  • Colloidal dispersion systems include, but are not limited to, macromolecule complexes, nanocapsules , microspheres , beads and lipid-based systems including oil-in- water emulsions, micelles, mixed micelles and liposomes.
  • a preferred colloidal dispersion system is a plurality of liposomes, artificial membrane vesicles which may be used as cellular delivery vehicles for bioactive agents in vi tro and in vivo (Mannino et al . , Biotechniques , 1988, _, 682; Blume and Cevc, Biochem . et Biophys . Acta, 1990, 1029 , 91; Lappalainen et al . , Antiviral Res . , 1994, 23 , 119; Chonn and Cullis, Current Op . Biotech . , 1995, 6, 698).
  • RNA, DNA and intact virions can be encapsulated within the aqueous interior and delivered to brain cells in a biologically active form (Fraley et al . ,
  • the composition of the liposome is usually a combination of lipids, particularly phospholipids, in particular, high phase transition temperature phospholipids, usually in combination with one or more steroids, particularly cholesterol.
  • lipids useful in liposome production include phosphatidyl compounds, such as phosphatidylglycerol, phosphatidylcholine, phosphatidylserine, phosphatidylethanolamine, sphingolipids , cerebrosides and gangliosides .
  • diacyl phosphatidylglycerols where the lipid moiety contains from 14-18 carbon atoms, particularly from 16-18 carbon atoms, and is saturated (lacking double bonds within the 14- 18 carbon atom chain) .
  • Illustrative phospholipids include phosphatidylcholine, dipalmitoylphosphatidylcholine and distearoylphosphatidylcholine .
  • the targeting of colloidal dispersion systems can be either passive or active. Passive targeting utilizes the natural tendency of liposomes to distribute to cells of the reticuloendothelial system in organs that contain sinusoidal capillaries. Active targeting, by contrast, involves modification of the liposome by coupling thereto a specific ligand such as a viral protein coat (Morishita et al . , Proc. Na tl . Acad . Sci . (U. S . A .
  • lipid groups can be incorporated into the lipid bilayer of the liposome in order to maintain the targeting ligand in close association with the lipid bilayer.
  • Various linking groups can be used for joining the lipid chains to the targeting ligand.
  • the targeting ligand which binds a specific cell surface molecule found predominantly on cells to which delivery of the oligonucleotides of the invention is desired, may be, for example, (1) a hormone, growth factor or a suitable oligopeptide fragment thereof which is bound by a specific cellular receptor predominantly expressed by cells to which delivery is desired or (2) a polyclonal or monoclonal antibody, or a suitable fragment thereof (e.g., Fab; F(ab') 2 ) which specifically binds an antigenic epitope found predominantly on targeted cells.
  • Two or more bioactive agents e.g., an oligonucleotide and a conventional drug; two oligonucleotides
  • compositions for parenteral, intrathecal or intraventricular administration, or colloidal dispersion systems may include sterile aqueous solutions which may also contain buffers, diluents and other suitable additives. Dosing is dependent on severity and responsiveness of the disease state to be treated, with the course of treatment lasting from several days to several months, or until a cure is effected or a diminution of the disease state is achieved. Optimal dosing schedules can be calculated from measurements of drug accumulation in the body of the patient. Persons of ordinary skill can easily determine optimum dosages, dosing methodologies and repetition rates.
  • Optimum dosages may vary depending on the relative potency of individual oligonucleotides, and can generally be estimated based on EC 50 s found to be effective in in vi tro and in vivo animal models. In general, dosage is from 0.01 ⁇ g to 100 g per kg of body weight, and may be given once or more daily, weekly, monthly or yearly, or even once every 2 to 20 years.
  • Oligonucleotides were synthesized on an automated DNA synthesizer using standard phosphoramidite chemistry with oxidation using iodine.
  • ⁇ - Cyanoethyldiisopropyl phosphoramidites were purchased from Applied Biosystems (Foster City, CA) .
  • the standard oxidation bottle was replaced by a 0.2 M solution of 3H-1, 2-benzodithiole-3 -one-1, 1-dioxide in acetonitrile for the stepwise thiation of the phosphite linkages .
  • 2'-methoxy-) phosphorothioate oligonucleotides was according to the procedures set forth above substituting 2'-0-methyl ⁇ - cyanoethyldiisopropyl phosphoramidites (Chemgenes, Needham, MA) for standard phosphoramidites and increasing the wait cycle after the pulse delivery of tetrazole and base to 360 seconds .
  • the 2 ' -fluoro-phosphorothioate oligonucleotides of the invention were synthesized using 5 ' -dimethoxytrityl-3 ' - phosphoramidites and prepared as disclosed in U.S. patent application Serial No. 08/383,666, filed February 3, 1995, and U.S. Patent 5,459,255, which issued October 8, 1996, both of which are assigned to the same assignee as the instant application and which are incorporated by reference herein.
  • the 2 ' -fluoro-oligonucleotides were prepared using phosphoramidite chemistry and a slight modification of the standard DNA synthesis protocol (i.e., deprotection was effected using methanolic ammonia at room temperature) .
  • the 2 ' -methoxyethoxy oligonucleotides were synthesized essentially according to the methods of Martin et al . ( Helv. Chim . Acta , 1995, 78 , 486).
  • the 3' nucleotide of the 2 ' -methoxyethoxy oligonucleotides was a deoxynucleotide, and 2 ' -0-CH 2 CH 2 OCH 3 _cytosines were 5-methyl cytosines, which were synthesized according to the procedures described below.
  • PNA antisense analogs were prepared essentially as described in U.S. Patents Nos. 5,539,082 and 5,539,083, both of which (1) issued July 23, 1996, (2) are assigned to the same assignee as the instant application and (3) are incorporated by reference herein.
  • Oligonucleotides comprising 2 , 6-diaminopurine were prepared essentially as described in U.S. Patents No. 5,506,351 which issued April 9, 1996, is assigned to the same assignee as the instant application and which is incorporated by reference herein. Oligonucleotides comprising 2,6- diaminopurine can also be prepared by enzymatic means (Bailly et al . , Proc . Na tl . Acad . Sci . U. S . A . , 1996, 53:13623) .
  • oligonucleotides were purified by precipitation 2x from 0.5 M NaCI with 2.5 volumes of ethanol. Analytical gel electrophoresis was accomplished in 20% acrylamide, 8 M urea and 45 mM Tris-borate buffer (pH 7) .
  • the resulting syrup was poured into diethylether (2.5 L) , with stirring.
  • the product formed a gum.
  • the ether was decanted and the residue was dissolved in a minimum amount of methanol (ca. 400 mL) .
  • the solution was poured into fresh ether (2.5 L) to yield a stiff gum.
  • the ether was decanted and the gum was dried in a vacuum oven (60°C at 1 mm Hg for 24 h) to give a solid which was crushed to a light tan powder (57 g, 85% crude yield) .
  • the material was used- as is for further reactions.
  • 2' -O-Methoxyethyl-5 -methyluridine 2,2' -Anhydro-5- methyluridine (195 g, 0.81 M) , tris (2-methoxyethyl) borate (231 g, 0.98 M) and 2 -methoxyethanol (1.2 L) were added to a 2 L stainless steel pressure vessel and placed in a pre- heated oil bath at 160°C. After heating for 48 hours at 155- 160°C, the vessel was opened and the solution evaporated to dryness and triturated with MeOH (200 mL) . The residue was suspended in hot acetone (1 L) . The insoluble salts were filtered, washed with acetone (150 mL) and the filtrate evaporated.
  • 2' -O-Methoxyethyl-5' -O-dimethoxytrityl-5-methyluridine 2'- O-Methoxyethyl- 5 -methyluridine (160 g, 0.506 M) was co- evaporated with pyridine (250 mL) and the dried residue dissolved in pyridine (1.3 L) .
  • a first aliquot of dimethoxytrityl chloride (94.3 g, 0.278 M) was added and the mixture stirred at room temperature for one hour.
  • a second aliquot of dimethoxytrityl chloride (94.3 g, 0.278 M) was added and the reaction stirred for an additional one hour.
  • Methanol (170 mL) was then added to stop the reaction. HPLC showed the presence of approximately 70% product.
  • the solvent was evaporated and triturated with CH 3 CN (200 mL) .
  • 3' -O-Acetyl-2' -O-methoxyethyl-5 ' -O-dimethoxytrityl-5- methyluridine 2' -O-Methoxyethyl -5 ' -O-dimethoxytrityl-5- methyluridine (106 g, 0.167 M) , DMF/pyridine (750 mL of a 3:1 mixture prepared from 562 mL of DMF and 188 mL of pyridine) and acetic anhydride (24.38 mL, 0.258 M) were combined and stirred at room temperature for 24 hours. The reaction was monitored by tic by first quenching the tic sample with the addition of MeOH .
  • 3' -O-Acetyl-2' -O-methoxyethyl-5' -0-dimethoxytrityl-5-methyl- 4-triazoleuridine A first solution was prepared by dissolving 3 ' -O-acetyl-2 ' -O-methoxyethyl-5 ' -O- dimethoxytrityl- 5-methyluridine (96 g, 0.144 M) in CH 3 CN (700 mL) and set aside. Triethylamine (189 mL, 1.44 M) was added to a solution of triazole (90 g, 1.3 M) in CH 3 CN (1 L) , cooled to -5°C and stirred for 0.5 h using an overhead stirrer.
  • P0C1 3 was added dropwise, over a 30 minute period, to the stirred solution maintained at 0-10°C, and the resulting mixture stirred for an additional 2 hours.
  • the first solution was added dropwise, over a 45 minute period, to the later solution.
  • the resulting reaction mixture was stored overnight in a cold room. Salts were filtered from the reaction mixture and the solution was evaporated. The residue was dissolved in EtOAc (1 L) and the insoluble solids were removed by filtration. The filtrate was washed with lx 300 mL of NaHC0 3 and 2x 300 mL of saturated NaCI, dried over sodium sulfate and evaporated. The residue was triturated with EtOAc to give the title compound.
  • N 4 -Benzoyl-2 ' -O-methoxyethyl-5' -O-dimethoxytrityl-5- methylcytidine 2' -O-Methoxyethyl-5 ' -O-dimethoxytrityl-5- methylcytidine (85 g, 0.134 M) was dissolved in DMF (800 mL) and benzoic anhydride (37.2 g, 0.165 M) was added with stirring. After stirring for 3 hours, tic showed the reaction to be approximately 95% complete. The solvent was evaporated and the residue azeotroped with MeOH (200 mL) .
  • Table 1 shows the sequence and activity of each of the oligonucleotides evaluated for inhibition of c-jun mRNA expression
  • Table 2 shows the sequence and activity of the oligonucleotides evaluated for inhibition of c-fos mRNA expression. Oligonucleotide activities were evaluated as described infra in Example 2 et seq. For the nucleotide sequence of the human c-jun gene, see Hattori et al . , Proc. Natl. Acad. Sci. U.S.A., 1988, 85:9148, and Genbank accession No. J04111 ( "HUMJUNA” ) .
  • nucleotide sequence of the human c-fos gene is described by Van Straaten et al . , Proc. Natl. Acad. Sci. U.S.A. , 1983, 80:3183, and in Genbank accession No. K00650 ("HUMFOS") .
  • Control is TPA induction, at 1 hour, in A549 cells N.D., not determined.
  • Example 2 Screening for Oligonucleotides that Modulate mRNA Expression of the AP-1 Subunits c-fos and c-jun
  • A549 cells were grown in T-75 flasks until 80-90% confluent.
  • Cell line A549 is available from, inter alia , the American Type Culture Collection, Rockville, MD, as ATCC No.
  • the cells were washed twice with 10 mL of media (DMEM) , followed by the addition of 5 mL of DMEM containing 20 ⁇ g/mL of LIPOFECTINTM (i.e., DOTMA/DOPE (N-[l-(2,3- dioleyloxy) propyl] -N, N,N-triethylammonium chloride/dioleoylphosphatidyl ethanolamine) ) .
  • LIPOFECTINTM i.e., DOTMA/DOPE (N-[l-(2,3- dioleyloxy) propyl] -N, N,N-triethylammonium chloride/dioleoylphosphatidyl ethanolamine)
  • DOTMA/DOPE N-[l-(2,3- dioleyloxy) propyl] -N, N,N-triethylammonium chloride/dioleoylphosphatidyl
  • Probes for human c-jun and c-fos were PCR products prepared using primers based on the published sequences thereof (respectively, Hattori et al . , Proc . Na tl . Acad . Sci . U. S . A . , 1988, 85:9148, and Van Straaten et al . , Proc . Natl . Acad . Sci . U. S. A . , 1983, 80:3183).
  • ISIS 10580, 10581, 10582 and 10583 were most active (81%, 76%, 83% and 80% inhibition, respectively) .
  • Treatment of cells with these oligonucleotides reduced c-jun expression to 19%, 24%, 17% and 20% (81%, 76%, 83% and 80% inhibition) , respectively, of the level seen in control experiments (100% expression, 0% inhibition) .
  • These oligonucleotides effect significant inhibition of c-jun and are therefor preferred.
  • Basal levels of c-jun mRNA are typically about 30% of the control value; ISIS 10572, 10578 and 10579 reduce c-jun levels to near basal levels (39%, 37% and 41%, respectively) and are thus also preferred.
  • the oligonucleotides most effective in reducing c-fos mRNA expression are ISIS 10633, 10635, 10638 and 10639.
  • oligonucleotides Treatment of cells with these oligonucleotides reduced c-fos expression to 24%, 12%, 22% and 9% (76%, 88%, 78% and 91% inhibition) , respectively, of the level seen in control experiments (100% expression, 0% inhibition) ; basal levels of c-fos mRNA are typically about 3% of the control value. These oligonucleotides effect significant inhibition of c-fos and are therefor preferred.
  • Dose-response experiments were performed at different oligonucleotide concentrations to determine the potency (i.e., ability to decrease expression of the appropriate mRNA target) of the most active compounds identified in the initial screen (Tables 3 and 4) .
  • the decreases in target mRNA expression effected by ISIS 10582 ( c-jun) and ISIS 10639 ( c-fos) are dose-dependent, as shown in Tables 3 and 4, respectively .
  • oligonucleotide-mediated inhibition of c-fos and c-jun mRNA expression was further examined by determining the effects of scrambled, 6- or 7- base mismatch and sense control versions of the most active oligonucleotides, ISIS 10582 ( c-jun) and ISIS 10639 ( c-fos) .
  • ISIS 10582 exhibited potent and specific inhibition of c-jun mRNA expression, but ISIS 11562 (sense version of ISIS 10582; SEQ ID NO:29), ISIS 11564 (6 base pair mismatch version of ISIS 10639; SEQ ID NO: 31) and ISIS 11563 (scrambled version of ISIS 10639; SEQ ID NO:30) had no detectable effect on c-jun mRNA levels during TPA induction (the sequences of ISIS 11562-11564 are given in Table 1) .
  • ISIS 10639 exhibited potent and specific inhibition of c-fos mRNA expression, but ISIS 11184 (sense version of ISIS 10639; SEQ ID NO:32) , ISIS 11185 (7 base pair mismatch version of ISIS 10639; SEQ ID NO:33) and ISIS 11186 (scrambled version of ISIS 10639; SEQ ID NO: 34) had no detectable effect on c-fos mRNA levels during TPA induction (the sequences of ISIS
  • ISIS 10582 effects potent and specific modulation (i.e., inhibition) of c-jun mRNA levels and that ISIS 10639 effects potent and specific modulation of c-fos mRNA levels.
  • ISIS 10639 was administered daily, i.v., for three weeks.
  • the oligonucleotide dosage was 25 mg/kg.
  • Tumor size was recorded weekly, and the results are shown in Table 6.
  • a substantial reduction in tumor growth rate was obtained upon treatment with ISIS 10639.
  • saline-treated tumors were 0.56 + . 0.12 g by weight, while tumors treated with ISIS 10639 were 0.31 ⁇ 0.1 g by weight.
  • c-fos active oligonucleotide ISIS 10639 was examined as follows. A549 cells were treated with oligonucleotides as in Examples 2-3, except that induction of c-Fos was effected by treatment of cells with TPA (1 uM) for three hours. At this time, whole cell protein was extracted in SDS (sodium dodecyl sulfate) buffer. Samples of extracts were electrophoresed, transferred to nitrocellulose filters which were immunoblotted using a c-Fos-specific antibody (Santa Cruz AB, Santa Cruz, CA) .
  • SDS sodium dodecyl sulfate
  • oligonucleotides that modulate c-Fos or c-Jun are identified, derivative or modified oligonucleotides having the same sequence thereas are prepared.
  • the modified oligonucleotides described in Tables 8 and 9 were prepared.
  • the effect of the c-fos- targeted oligonucleotides on c-fos RNA levels were evaluated as described in Examples 2-3.
  • the results (Table 10) demonstrate that some enhancement of c-fos modulation can be achieved by the use modifications such as, e.g., 2'-fluoro (ISIS 11200) .
  • PNA analogs of the invention are introduced into appropriate cell lines by microinjection according to the method of Hanvey e t al . ( Science, 1992, 258:1481) . Intracellular delivery of PNA analogs is confirmed by the use of a fluorescently tagged PNA antisense analog conjugate such as, e.g., ISIS 14240. TABLE 8 Additional Oligonucleotides and PNA Antisense Analogs Targeted to Human AP-1 Subunits
  • Emboldened residues indicate 2,6-A residues, i . e . , those having 2 , 6-diaminopurine as a nucleobase.
  • ISIS 14663 is a 2'-deoxy- rather than a 2 ' -meLhoxyethoxy-oligonucleotide .
  • Example 7 Oligonucleotides to Mouse AP-1 Subunits Tables 11 and 12 show the sequences of oligonucleotides designed to modulate mouse c-j un and c-fos mRNA expression, respectively.
  • Tables 11 and 12 show the sequences of oligonucleotides designed to modulate mouse c-j un and c-fos mRNA expression, respectively.
  • nucleotide sequence of the mouse c-jun gene see Genbank accession No. J04115/MUSCJUN and Ryder et al . , Proc . Na tl . Acad . Sci . U. S . A . , 1988, 85:8464.
  • the nucleotide sequence of the mouse c-fos gene is described in Genbank accession No. J00370/MUSFOS and by Van Beveren et al .
  • Tables 13 and 14 show the sequences of oligonucleotides designed to modulate rat c-jun and c-fos mRNA expression, respectively.
  • nucleotide sequence of the rat c-jun gene see Genbank accession No.
  • rat c-fos gene The nucleotide sequence of the rat c-fos gene is described in Genbank accession No. X06769/RNCFOSR and Curran et al . , Oncogene, 1987, 2:79. Oligonucleotide activities were evaluated as described infra in Example 2 et seg. with the exception that rat A10 cells (available from, inter alia , the American Type Culture Collection, Rockville, MD, as ATCC No. CRL-1476) were used instead of human A549 cells. Due to the high degree of homology between human and rat c-jun and c-fos nucleotide sequences, probes derived from the human genes were used to detect the rat messages .
  • ISIS 12633 (SEQ ID NO:78), a-20-mer phosphorothioate oligonucleotide complementary to a portion of the 3' UTR of rat c-jun mRNA, was selected as an active modulator of c-jun for further studies.
  • Another preferred oligonucleotide targeted to rat AP-1 subunits is ISIS 12635 (SEQ ID NO:80).
  • Tables 15 and 16 show the sequences and chemical modifications of second generation oligonucleotides designed to modulate mouse c-jun and c-fos mRNA expression. The activities of these modified oligonucleotide are evaluated as described infra in Example 8.
  • RASM cultured rat aortic smooth muscle
  • PDGF platelet-derived growth factor
  • PDGF ng/ml
  • c-jun mRNA levels were markedly less in ISIS 12633 -treated cells as compared to untreated cells or cells treated with ISIS 12898.
  • the decrease in c-jun mRNA levels was associated with a significant decrease in the proportion of cells in the G2/M interface at 24 hours. This result provides evidence of the role of AP-1-mediated gene expression in cellular proliferation and indicate that cell cycle progression can be modulated by preventing expression of one or both of the genes which encode a subunit of AP-1.
  • Invasion refers to the ability of cancer cells to detach from a primary site of attachment and penetrate, e.g., an underlying basement membrane.
  • Metastasis indicates a sequence of events wherein (1) a cancer cell detaches from its extracellular matrices, (2) the detached cancer cell migrates to another portion of the patient's body, often via the circulatory system, and (3) attaches to a distal and inappropriate extracellular matrix, thereby created a focus from which a secondary tumor can arise.
  • Normal cells do not possess the ability to invade or metastasize and/or undergo apoptosis (programmed cell death) if such events occur (Ruoslahti, Sci . Amer . , 1996, 275, 72).
  • the matrix metalloproteinases (MMPs) are a family of enzymes which have the ability to degrade components of the extracellular matrix (Birkedal-Hansen, Current Op .
  • MMP- 9 matrix metalloproteinase-9
  • NHEKs normally express no detectable MMP- 9
  • MMP- 9 can be induced by a number of stimuli, including TPA.
  • ISIS 10582 an oligonucleotide targeted to c-jun, was evaluated for its ability to modulate MMP- 9 expression according to the protocols described in Examples 2-3 with the following exceptions: (1) NHEK cells were used instead of A549 cells, (2) the probe used, a PCR product prepared using the published sequence of the MMP-9 gene (Huhtala et al . , J. Biol . Chem . , 1991, 266:16485; Sato et al .
  • Example 13 Antagonism of Inducers Other than TPA by
  • TNF- ⁇ , IL-13 or TGF-3 were treated and evaluated as in Examples 2 et seg. with the exception that TNF- ⁇ , IL-13 or TGF-3 (each at 10 ng/ml and all from R&D Systems,
  • CAGTGGCCAT CAAACCCGTG 20

Abstract

Compositions and methods for the treatment and diagnosis of diseases or disorders amenable to treatment through modulation of Activating Protein 1 (AP-1) expression are provided. In accordance with various embodiments of the present invention, oligonucleotides are provided which are specifically hybridizable with c-fos or c-jun, the genes encoding c-Fos or c-Jun, respectively. In a preferred embodiment, a method of modulating the metastasis of malignant tumors via modulation of one or more of the AP-1 subunits is provided; this method can be effected using the oligonucleotides of the invention or any other agent which modulates AP-1 or AP-1-mediated transcription.

Description

ANTISENSE OLIGONUCLEOTIDE COMPOSITIONS AND METHODS FOR THE MODULATION OF ACTIVATING PROTEIN 1
FIELD OF THE INVENTION
The present invention provides compositions and methods for modulating levels of the c-fos and c-jun genes, which encode the c-Fos and c-Jun subunits of AP-1, respectively. In vivo, AP-1, or transcription factor activating protein 1, is a heterogenous mixture of heterodimers of several related protein subunits including, in addition to c-Fos and c-Jun, FosB, Fra-1, Fra-2, c-Jun,
JunB, JunD, etc. ( The FOS and JUN Families of Proteins , Angel and Herrlich, eds., CRC Press, Boca Raton, FL, 1994). AP-1 has been implicated in abnormal cell proliferation and tumor formation, events that thus might be controlled by modulating the expression of c-fos and/or c-jun . The invention is further directed to therapeutic, diagnostic, and research based reagents and methods for evaluating and treating disease states or disorders which result from and/or respond positively to modulation of one or more AP-1 subunits. Such disease states and disorders include those involving the hyperproliferation of cells such as, e . g. , a tumor (neoplasm) or malignant cancer. Inhibition of AP-1 -mediated hyperproliferation of cells, and corresponding prophylactic, palliative and therapeutic effects result from treatment with the oligonucleotides of the invention. BACKGROUND OF THE INVENTION
Transcription factors play a central role in the expression of specific genes upon stimulation by extracellular signals, thereby regulating a complex array of biological processes. Members of the family of transcription factors termed AP-1 (activating protein-1) alter gene expression in response to growth factors, cytokines, tumor promoters, carcinogens and increased expression of certain oncogenes. Growth factors and cytokines exert their function by binding to specific cell surface receptors. Receptor occupancy triggers a signal transduction cascade to the nucleus. In this cascade, transcription factors such as AP-1 execute long term responses to the extracellular factors by modulating gene expression. Such changes in cellular gene expression lead to DNA synthesis, and eventually the formation of differentiated derivatives (Angel and Karin, Biochi . Biophys . Acta , 1991, 1072, 129) .
In general terms, AP-1 denotes one member of a family of related heterodimeric transcription factor complexes found in eukaryotic cells or viruses. However, as used herein,
"AP-1" specifically refers to the heterodimer formed of c-Fos and c-Jun (Angel and Herrlich, Chapter 1, and Schuermann, Chapter 2 in : The FOS and JUN Families of Proteins , Angel and Herrlich, eds., pp. 3-35, CRC Press, Boca Raton, FL, 1994; Bohmann et al . , Science, 1987, 238, 1386; Angel et al . ,
Na ture , 1988, 332 , 166) . These two proteins are products of the c-fos and c-jun proto-oncogenes, respectively. Repression of expression of either c-fos or c-jun, ox of both proto-oncogenes, and the resultant inhibition of the formation of c-Fos and c-Jun proteins, is desirable for the inhibition of cell proliferation, tumor formation and tumor growth .
The phosphorylation of proteins plays a key role in the transduction of extracellular signals into the cell. Mitogen-activated protein (MAP) kinases, enzymes which effect such phosphorylations are targets for the action of growth factors, hormones, and other agents involved in cellular metabolism, proliferation and differentiation (Cobb et al . , J. Biol . Chem . , 1995, 270, 14843). MAP kinases are themselves activated by phosphorylation catalyzed by, e . g. , receptor tyrosine kinases, G protein-coupled receptors, protein kinase C (PKC) , and the apparently MAP kinase dedicated kinases MEK1 and MEK2. MAP kinases include, but are not limited to, ERK1 , ERK2 , two isoforms of ERK3 , ERK4 (ERK stands for "extracellular signal -regulated protein kinase) , Jun N-terminal kinases/stress-activated protein kinases (JNKs/SAPKs) , p38/HOGl, p57 MAP kinases, MKK3 (MAP kinase kinase 3) and MKK4 (MAP kinase kinase 4, also known as SAPK/ERK kinase (SEK) or JNK kinase (JNKK) ) (Cobb et al . , J". Biol . Chem . , 1995, 270 , 14843, and references cited therein). In general, MAP kinases are involved in a variety of signal transduction pathways (sometimes overlapping and sometimes parallel) that function to convey extracellular stimuli to protooncogene products to modulate cellular proliferation and/or differentiation.
One of the signal transduction pathways involves the MAP kinases Jun kinase 1 and Jun kinase 2 which are responsible for the phosphorylation of specific sites (Serine 63 and Serine 73) on c-Jun. Phosphorylation of these sites potentiates the ability of AP-1 to activate transcription (Binetruy et al . , Na ture, 1991, 351 , 122; Smeal et al . , Nature, 1991, 354 , 494) . At least one human leukemia oncogene has been shown to enhance Jun N-terminal Kinase (JNK) function (Raitano et al . , Proc . Na tl . Acad . Sci . (USA) , 1995, 92 , 11746), thus indirectly demonstrating a role for AP-1 in cellular hyperproliferation and tumorigenesis . Cellular hyperproliferation in an animal can have several outcomes. Hyperproliferating cells might be attacked and killed by the animal's immune system before a tumor can form. Tumors are abnormal growths resulting from the hyperproliferation of cells. Cells that proliferate to excess but stay put form benign tumors, which can typically be removed by local surgery. In contrast, malignant tumors or cancers comprise cells that are capable of undergoing metastasis, i . e . , a process by which hyperproliterative cells spread to, and secure themselves within, other parts of the body via the circulatory or lymphatic system (see, generally, Chapter 16 In : Molecular Biology of the Cell , Alberts et al . , eds., pp. 891-950, Garland Publishing, Inc., New York, 1983). Using the oligonucleotides of the invention, it has surprisingly been discovered that several genes encoding enzymes required for metastasis are positively regulated by AP-1. Accordingly, inhibition of expression of c-fos and/or c-jun serves as a means to modulate the metastasis of malignant tumors. A method of modulating one or more metastatic events using the oligonucleotides of the invention is thus herein provided.
RELEVANT ART Soprano et al . {Ann . N. Y. Acad. Sci . , 1992, 660 , 231) have used antisense oligodeoxynucleotides targeted to c-jun mRNA to study their ability to inhibit DNA synthesis and cell division.
Liu et al . {Ann . Neurol . , 1994, 36, 566) describe the suppression of c-fos by intraventricular infusion of an antisense oligodeoxynucleotide targeted to c-fos mRNA.
Chen et al . ( Cancer Lett . , 1994, 85 , 119) describe repression of c-jun expression by antisense oligodeoxynucleotides resulting in the inhibition of cell proliferation in E5a transformed cells.
Gillardon et al . describe the topical application of c-fos antisense oligodeoxynucleotides to the rat spinal cord ( Eur . J. Neurosci . , 1994, 6, 880) ultraviolet (UV) -exposed rat eyes (Bri tish J. Ophthal . , 1995, 79 , 277) and UV- irradiated rat skin ( Carcinogenesis , 1995, 16, 1853) .
U.S. Patent No. 5,602,156, which issued February 11, 1997, to Kohn et al . , discloses non-oligonucleotide compositions and methods for inhibiting the expression of two metalloproteinases, MMP-1 and MMP-2. International Publication Number WO 95/02051, published January 19, 1995, discloses antisense oligonucleotides targeted to the mRNA of c-fos and c-jun .
International Publication Number WO 95/03323, published February 2, 1995, discloses antisense nucleic acids which are complementary to the polynucleotide encoding a polypeptide which is capable of phosphorylating the c-jun N- terminal activation domain. Also provided are methods for treating a cell proliferative disorder associated with said polypeptide .
International Publication Number WO 95/03324, published February 2, 1995, describes a polypeptide which phosphorylates the c-jun N-terminal activation domain. This publication also discloses a polynucleotide sequence encoding the polypeptide.
To date, there are no known therapeutic agents which effectively inhibit gene expression of c-fos and/or c-jun . Furthermore, there are to date no known therapeutic agents that modulate the metastasis of malignant cells. The compositions and methods of the invention overcome these limitations. Further objectives of the invention are apparent from the present disclosure.
SUMMARY OF THE INVENTION
In accordance with the present invention, oligonucleotides are provided which specifically hybridize with nucleic acids encoding c-Fos or c-Jun. Certain oligonucleotides of the invention are designed to bind either directly to mRNA transcribed from, or to a selected DNA portion of, the respective gene, thereby modulating the amount of protein translated from a c-fos or c-jun mRNA and/or the amount of mRNA transcribed from a c-fos or c-jun gene, respectively. Such modulation can, in turn, effect the modulation of enzymes and cellular processes involved in the metastasis of malignant cells.
Oligonucleotides may comprise nucleotide sequences sufficient in identity and number to effect specific hybridization with a particular nucleic acid. Such oligonucleotides are commonly described as "antisense." Antisense oligonucleotides are commonly used as research reagents, diagnostic aids, and therapeutic agents.
It has been discovered that the c-fos and c-jun genes, encoding the c-Fos and c-Jun proteins, respectively, are particularly amenable to this approach. As a consequence of the association between cellular proliferation and AP-1 (the heterodimer of c-Fos and c-Jun) expression, modulation of the expression of c-fos and/or c-jun leads to modulation of AP-1, and, accordingly, modulation of cellular proliferation. Such modulation is desirable for treating or modulating various hyperproliferative disorders or diseases, such as various cancers. Such inhibition is further desirable for preventing or modulating the development of such diseases or disorders in an animal suspected of being, or known to be, prone to such diseases or disorders. If desired, modulation of one subunit can be combined with modulation of the subunit of AP- 1 in order to achieve a requisite degree of effect upon AP-1- mediated transcription.
Methods of modulating the expression of c-Fos or c-Jun proteins comprising contacting animals with oligonucleotides specifically hybridizable with a c-fos or c-jun gene, respectively, are herein provided. These methods are believed to be useful both therapeutically and diagnostically as a consequence of the association between AP-1 expression and cellular proliferation. These methods are also useful as tools, for example, in the detection and determination of the role of AP-1 protein expression in various cell functions and physiological processes and conditions, and for the diagnosis of conditions associated with such expression and activation. The present invention also comprises methods of inhibiting AP-1-mediated transcriptional activation using the oligonucleotides of the invention. Methods of treating conditions in which abnormal or excessive AP-1-mediated transcriptional activation and cellular proliferation occur are also provided. These methods employ the oligonucleotides of the invention and are believed to be useful both therapeutically and as clinical research and diagnostic tools. The oligonucleotides of the present invention may also be used for research purposes. Thus, the specific hybridization exhibited by the oligonucleotides of the present invention may be used for assays, purifications, cellular product preparations and in other methodologies which may be appreciated by persons of ordinary skill in the art .
Methods comprising contacting animals with oligonucleotides specifically hybridizable with nucleic acids encoding c-Fos or c-Jun proteins are herein provided. Such methods can be used to modulate or detect the expression of c-fos or c-jun genes and are thus believed to be useful both therapeutically and diagnostically.
The methods disclosed herein are also useful, for example, as clinical research tools in the detection and determination of the role of AP-1-mediated gene expression in various immune system functions and physiological processes and conditions, and for the diagnosis of conditions associated with their expression. The specific hybridization exhibited by the oligonucleotides of the present invention may be used for assays, purifications, cellular product preparations and in other methodologies which may be appreciated by persons of ordinary skill in the art. For example, because the oligonucleotides of this invention specifically hybridize to nucleic acids encoding c-Fos or c- Jun, sandwich and other assays can easily be constructed to exploit this fact. Detection of specific hybridization of an oligonucleotide of the invention with a nucleic acid encoding a c-Fos or c-Jun protein present in a sample can routinely be accomplished. Such detection may include detectably labeling an oligonucleotide of the invention by enzyme conjugation, radiolabeling or any other suitable detection system. A number of assays may be formulated employing the present invention, which assays will commonly comprise contacting a tissue or cell sample with a detectably labeled oligonucleotide of the present invention under conditions selected to permit hybridization and measuring such hybridization by detection of the label, as is appreciated by those of ordinary skill in the art.
DETAILED DESCRIPTION OF THE INVENTION
The present invention employs oligonucleotides for use in antisense inhibition of the function of RNA and DNA encoding a c-Fos protein or a c-Jun protein. The present invention also employs oligonucleotides which are designed to be specifically hybridizable to DNA or messenger RNA (mRNA) encoding such proteins and ultimately modulating the amount of such proteins transcribed from their respective genes.
Such hybridization with mRNA interferes with the normal role of mRNA and causes a modulation of its function in cells. The functions of mRNA to be interfered with include all vital functions such as translocation of the RNA to the site for protein translation, actual translation of protein from the RNA, splicing of the RNA to yield one or more mRNA species, and possibly even independent catalytic activity which may be engaged in by the RNA. The overall effect of such interference with mRNA function is modulation of the expression of a c-Fos protein or a c-Jun protein. In the context of this invention, "modulation" means either an increase (stimulation) or a decrease (inhibition) in the expression of a gene. In the context of the present invention, inhibition is the preferred form of modulation of gene expression.
In the context of this invention, the term "oligonucleotide" refers to an oligomer or polymer of ribonucleic acid or deoxyribonucleic acid. This term includes oligonucleotides composed of naturally-occurring nucleobases, sugars and covalent intersugar (backbone) linkages as well as oligonucleotides having non-naturally- occurring portions which function similarly. Such modified or substituted oligonucleotides are often preferred over native forms because of desirable properties such as, for example, enhanced cellular uptake, enhanced binding to target and increased stability in the presence of nucleases . An oligonucleotide is a polymer of a repeating unit generically known as a nucleotide. An unmodified (naturally occurring) nucleotide has three components: (1) a nitrogenous base linked by one of its nitrogen atoms to (2) a 5 -carbon cyclic sugar and (3) a phospha te, esterified to carbon 5 of the sugar. When incorporated into an oligonucleotide chain, the phosphate of a first nucleotide is also esterified to carbon 3 of the sugar of a second, adjacent nucleotide. The "backbone" of an unmodified oligonucleotide consists of (2) and (3), that is, sugars linked together by phosphodiester linkages between the carbon 5 (5') position of the sugar of a first nucleotide and the carbon 3 (3') position of a second, adjacent nucleotide. A "nucleoside" is the combination of (1) a nucleobase and (2) a sugar in the absence of (3) a phosphate moiety (Kornberg, A., DNA Replication, pp. 4-7,
W.H. Freeman & Co., San Francisco, 1980) . The backbone of an oligonucleotide positions a series of bases in a specific order; the written representation of this series of bases, which is conventionally written in 5 ' to 3 ' order, is known as a nucleotide sequence. The oligonucleotides in accordance with this invention preferably comprise from about 8 to about 30 nucleotides. It is more preferred that such oligonucleotides comprise from about 15 to 25 nucleotides. Oligonucleotides may comprise nucleotide sequences sufficient in identity and number to effect specific hybridization with a particular nucleic acid. Such oligonucleotides which specifically hybridize to a portion of the sense strand of a gene are commonly described as "antisense." Antisense oligonucleotides are commonly used as research reagents, diagnostic aids, and therapeutic agents. For example, antisense oligonucleotides, which are able to inhibit gene expression with exquisite specificity, are often used by those of ordinary skill to elucidate the function of particular genes, for example to distinguish between the functions of various members of a biological pathway. This specific inhibitory effect has, therefore, been harnessed by those skilled in the art for research uses . The specificity and sensitivity of oligonucleotides is also harnessed by those of skill in the art for therapeutic uses. For example, the following U.S. patents demonstrate palliative, therapeutic and other methods utilizing antisense oligonucleotides. U. S. Patent 5,135,917 provides antisense oligonucleotides that inhibit human interleukin-1 receptor expression. U.S. Patent 5,098,890 is directed to antisense oligonucleotides complementary to the c-myb oncogene and antisense oligonucleotide therapies for certain cancerous conditions. U.S. Patent 5,087,617 provides methods for treating cancer patients with antisense oligonucleotides. U.S. Patent 5,166,195 provides oligonucleotide inhibitors of Human Immunodeficiency Virus (HIV). U.S. Patent 5,004,810 provides oligomers capable of hybridizing to herpes simplex virus Vmw65 mRNA and inhibiting replication. U.S. Patent 5,194,428 provides antisense oligonucleotides having antiviral activity against influenzavirus . U.S. Patent 4,806,463 provides antisense oligonucleotides and methods using them to inhibit HTLV-III replication. U.S. Patent 5,286,717 provides oligonucleotides having a complementary base sequence to a portion of an oncogene. U.S. Patent 5,276,019 and U.S. Patent 5,264,423 are directed to phosphorothioate oligonucleotide analogs used to prevent replication of foreign nucleic acids in cells. U.S. Patent 4,689,320 is directed to antisense oligonucleotides as antiviral agents specific to cytomegalovirus (CMV) . U.S. Patent 5,098,890 provides oligonucleotides complementary to at least a portion of the mRNA transcript of the human c-myb gene. U.S. Patent 5,242,906 provides antisense oligonucleotides useful in the treatment of latent Epstein- Barr virus (EBV) infections.
It is preferred to target specific genes for antisense attack. "Targeting" an oligonucleotide to the associated nucleic acid, in the context of this invention, is a multistep process. The process usually begins with the identification of a nucleic acid sequence whose function is to be modulated. This may be, for example, a cellular gene (or mRNA transcribed from the gene) whose expression is associated with a particular disorder or disease state, or a foreign nucleic acid from an infectious agent. In the present invention, the target is a cellular gene ( c-fos or c- un) encoding a subunit of AP-1, for which modulation is desired in certain instances. The targeting process also includes determination of a region (or regions) within this gene for the oligonucleotide interaction to occur such that the desired effect, either detection or modulation of expression of the protein, will result. Once the target region have been identified, oligonucleotides are chosen which are sufficiently complementary to the target, i . e . , hybridize sufficiently well and with sufficient specificity to give the desired effect. There are many regions of a gene that may be targeted for antisense modulation: the region of the 5' Cap, a specialized structure that at least partially mediates ribosome binding; the 5' untranslated (noncoding) region (hereinafter, the "5' -UTR"); the translation initiation codon region (hereinafter, the "tIR"); the open reading frame
(hereinafter, the "ORF"); the translation termination codon region (hereinafter, the "tTR" ) ; and the 3' untranslated (noncoding) region (hereinafter, the "3' -UTR"), which has at its 3' end a "poly A" tail. As is known in the art, these regions are arranged in a typical messenger RNA molecule in the following order (left to right, 5' to 3') : 5' Cap, 5'- UTR, tIR, ORF, tTR, 3' -UTR, poly A tail. As is also known in the art, although some eukaryotic transcripts are directly translated, many ORFs contain one or more sequences, known as "introns," which are excised from a transcript before it is translated; the expressed (unexcised) portions of the ORF are referred to as "exons" (Alberts et al . , Molecular Biology of the Cell , pp. 331-332 and 411-415, Garland Publishing Inc., New York, 1983) . Furthermore, because many eukaryotic ORFs are a thousand nucleotides or more in length, it is often convenient to subdivide the ORF into, e . g. , the 5' ORF region, the central ORF region, and the 3' ORF region. In some instances, an ORF contains one or more sites that may be targeted due to some functional significance in vivo . Examples of the latter types of sites include intragenic stem-loop structures (see, e . g. , U.S. Patent No. 5,512,438) and, in unprocessed mRNA molecules, intron/exon splice sites. Within the context of the present invention, one preferred intragenic site is the region encompassing the translation initiation codon of the open reading frame (ORF) of the gene. Because, as is known in the art, the translation initiation codon is typically 5 ' -AUG (in transcribed mRNA molecules; 5'- ATG in the corresponding DNA molecule) , the translation initiation codon is also referred to as the "AUG codon," the "start codon" or the "AUG start codon." A minority of genes have a translation initiation codon having the RNA sequence 5'-GUG, 5'-UUG or 5'-CUG, and 5'-AUA, 5 ' -ACG and 5'-CUG have been shown to function in vivo . Furthermore, 5 ' -UUU functions as a translation initiation codon in vi tro (Brigstock et al . , Growth Factors, 1990, 4, 45; Gelbert et al . , Somat . Cell . Mol . Genet . , 1990, 16, 173; Gold and Stormo, Chapter 78 in : Escherichia coli and Salmonella typhimurium : Cellular and Molecular Biology, Vol. 2, p. 1303, Neidhardt et al . , eds., American Society for Microbiology, Washington, D.C., 1981 ) . Thus, the terms "translation initiation codon" and "start codon" can encompass many codon sequences, even though the initiator amino acid in each instance is typically methionine (in eukaryotes) or formylmethionine (prokaryotes) . It is also known in the art that eukaryotic and prokaryotic genes may have two or more alternative start codons, any one of which may be preferentially utilized for translation initiation in a particular cell type or tissue, or under a particular set of conditions, in order to generate related polypeptides having different amino terminal sequences (Markussen et al . , Development , 1995, 121 , 3723; Gao et al . , Cancer Res . , 1995, 55 , 743; McDermott et al . , Gene, 1992, 117, 193; Perri et al . , J. Biol . Chem . , 1991, 266, 12536; French et al . , J. Virol . , 1989, 63 , 3270; Pushpa-Rekha et al . , J. Biol . Chem . , 1995, 270, 26993; Monaco et al . , J. Biol . Chem . , 1994, 269 , 347; DeVirgilio et al . , Yeast , 1992, 8 , 1043; Kanagasundaram et al . , Biochim . Biophys . Acta , 1992, 1171 , 198; Olsen et al . , Mol . Endocrinol . , 1991, 5, 1246; Saul et al . , Appl . Environ . Microbiol . , 1990, 56, 3117; Yaoita et al . , Proc .
Natl . Acad . Sci . USA, 1990, 87, 7090; Rogers et al . , EMBO J. , 1990, 9 , 2273). In the context of the invention, "start codon" and "translation initiation codon" refer to the codon or codons that are used in vivo to initiate translation of an mRNA molecule transcribed from a gene encoding a c-Fos or c- Jun protein, regardless of the sequence (s) of such codons. It is also known in the art that a translation termination codon (or "stop codon") of a gene may have one of three sequences, i.e., 5'-UAA, 5'-UAG and 5'-UGA (the corresponding DNA sequences are 5'-TAA, 5' -TAG and 5'-TGA, respectively). The terms "start codon region" and "translation initiation region" refer to a portion of such an mRNA or gene that encompasses from about 25 to about 50 contiguous nucleotides in either direction (i.e., 5' or 3 ' ) from a translation initiation codon. Similarly, the terms "stop codon region" and "translation termination region" refer to a portion of such an mRNA or gene that encompasses from about 25 to about 50 contiguous nucleotides in either direction (i.e., 5' or 3') from a translation termination codon. In the context of this invention, the term
"oligonucleotide" includes oligonucleotides composed of naturally-occurring nucleobases, sugars and covalent intersugar (backbone) linkages as well as oligonucleotides having non-naturally-occurring portions which function similarly. Such modified or substituted oligonucleotides may be preferred over native forms because of desirable properties such as, for example, enhanced cellular uptake, enhanced binding to target and increased stability in the presence of nucleases . Specific examples of some preferred modified oligonucleotides envisioned for this invention include those containing phosphorothioates , phosphotriesters , methyl phosphonates, short chain alkyl or cycloalkyl intersugar linkages or short chain heteroatomic or heterocyclic intersugar linkages. Most preferred are oligonucleotides with phosphorothioates and those with CH2-NH-0-CH2, CH2- N(CH3)-0-CH2 [known as a methylene (methylimino) or MMI backbone], CH2-0-N (CH3) -CH2, CH2-N (CH3) -N (CH3) -CH2 and 0-N (CH3) - CH2-CH2 backbones, wherein the native phosphodiester backbone is represented as 0-P-0-CH2) . Also preferred are oligonucleotides having morpholino backbone structures (Summerton and Weller, U.S. Patent 5,034,506). Further preferred are oligonucleotides with NR-C (*) -CH2-CH2, CH2-NR- C(*)-CH2, CH2-CH2-NR-C(*) , C ( * ) -NR-CH2-CH2 and CH2-C ( * ) -NR-CH2 backbones, wherein "*" represents 0 or S (known as amide backbones; DeMesmaeker et al . , WO 92/20823, published November 26, 1992) . In other preferred embodiments, such as the peptide nucleic acid (PNA) backbone, the phosphodiester backbone of the oligonucleotide is replaced with a polyamide backbone, the nucleobases being bound directly or indirectly to the aza nitrogen atoms of the polyamide backbone (Nielsen et al . , Science, 1991, 254 , 1497; U.S. Patent No. 5,539,082). The oligonucleotides of the invention may additionally or alternatively include nucleobase modifications or substitutions. As used herein, "unmodified" or "natural" nucleobases include adenine (A) , guanine (G) , thymine (T) , cytosine (C) and uracil (U) . Modified nucleobases include nucleobases found only infrequently or transiently in natural nucleic acids, e . g. , hypoxanthine , 6-methyladenine , 5- methylcytosine, 5-hydroxyτnethylcytosine (HMC) , glycosyl HMC and gentiobiosyl HMC, as well synthetic nucleobases, e.g., 2- aminoadenine, 2-thiouracil , 2-thiothymine , 5-bromouracil , 5- hydroxymethyluracil, 8-azaguanine, 7-deazaguanine , N6(6- aminohexyl) adenine and 2 , 6-diaminopurine (Kornberg, A., DNA Replication, pp. 75-77, W.H. Freeman & Co., San Francisco, 1980; Gebeyehu, G. , et al . , Nucleic Acids Res . , 1987, 15 , 4513) .
The oligonucleotides of the invention may additionally or alternatively comprise substitutions of the sugar portion of the individual nucleotides. For example, oligonucleotides may also have sugar mimetics such as cyclobutyls in place of the pentofuranosyl group. Other preferred modified oligonucleotides may contain one or more substituted sugar moieties comprising one of the following at the 2' position: OH, SH, SCH3, F, OCN, OCH3OCH3, OCH30 (CH2) nCH3, 0(CH2)nNH2 or 0(CH2)nCH3 where n is from 1 to about 10; C to C10 lower alkyl, alkoxyalkoxy, substituted lower alkyl, alkaryl or aralkyl; Cl ; Br; CN; CF3 ; OCF3; 0-, S-, or N-alkyl; O- , S-, or N-alkenyl; SOCH3 ; S02CH3 ; ON02; N02; N3 ; NH2 ; heterocycloalkyl ; heterocycloalkaryl ; aminoalkylamino; polyalkylamino; substituted silyl; an RNA cleaving group; a reporter group; an intercalator ; a group for improving the pharmacokinetic properties of an oligonucleotide; or a group for improving the pharmacodynamic properties of an oligonucleotide and other substituents having similar properties. A preferred modification, particularly for orally deliverable pharmaceutical compositions, is 2'- methoxyethoxy [2 ' -0-CH2CH2OCH3, also known as 2'-0-(2- methoxyethyl) ] (Martin et al . , Helv. Chim . Acta , 1995, 78 , 486). Other preferred modifications include 2 ' -methoxy (2'- 0-CH3) , 2'-propoxy (2 ' -OCH2CH2CH3) and 2'-fluoro (2'-F).
Similar modifications may also be made at other positions on the oligonucleotide, particularly the 3' position of the sugar on the 3' terminal nucleotide and the 5' position of 5' terminal nucleotide. The 5' and 3' termini of an oligonucleotide may also be modified to serve as points of chemical conjugation of, e.g., lipophilic moieties (see immediately subsequent paragraph) , intercalating agents (Kuyavin et al . , WO 96/32496, published October 17, 1996; Nguyen et al . , U.S. Patent No. 4,835,263, issued May 30, 1989) or hydroxyalkyl groups (Helene et al . , WO 96/34008, published October 31, 1996) .
Other positions within an oligonucleotide of the invention can be used to chemically link thereto one or more effector groups to form an oligonucleotide conjugate. An "effector group" is a chemical moiety that is capable of carrying out a particular chemical or biological function. Examples of such effector groups include, but are not limited to, an RNA cleaving group, a reporter group, an intercalator, a group for improving the pharmacokinetic properties of an oligonucleotide, or a group for improving the pharmacodynamic properties of an oligonucleotide and other substituents having similar properties. A variety of chemical linkers may be used to conjugate an effector group to an oligonucleotide of the invention. As an example, U.S. Patent No. 5,578,718 to Cook et al . discloses methods of attaching an alkylthio linker, which may be further derivatized to include additional groups, to ribofuranosyl positions, nucleosidic base positions, or on internucleoside linkages. Additional methods of conjugating oligonucleotides to various effector groups are known in the art; see, e . g. , Protocols for
Oligonucleotide Conjugates (Methods in Molecular Biology, Volume 26) Agrawal , S., ed. , Humana Press, Totowa, NJ, 1994.
Another preferred additional or alternative modification of the oligonucleotides of the invention involves chemically linking to the oligonucleotide one or more lipophilic moieties which enhance the cellular uptake of the oligonucleotide. Such lipophilic moieties may be linked to an oligonucleotide at several different positions on the oligonucleotide. Some preferred positions include the 3' position of the sugar of the 3' terminal nucleotide, the 5' position of the sugar of the 5' terminal nucleotide, and the 2' position of the sugar of any nucleotide. The N6 position of a purine nucleobase may also be utilized to link a lipophilic moiety to an oligonucleotide of the invention (Gebeyehu et al . , Nucleic Acids Res . , 1987, 15 , 4513). Such lipophilic moieties include but are not limited to a cholesteryl moiety (Letsinger et al . , Proc . Natl . Acad . Sci . USA, 1989, 86, 6553), cholic acid (Manoharan et al . , Bioorg . Med . Chem . Let . , 1994, 4, 1053), a thioether, e . g. , hexyl-S- tritylthiol (Manoharan et al . , Ann . N. Y. Acad . Sci . , 1992,
660 , 306; Manoharan et al . , Bioorg. Med . Chem . Let . , 1993, 3 , 2765), a thiocholesterol (Oberhauser et al . , Nucl . Acids Res . , 1992, 20 , 533), an aliphatic chain, e . g. , dodecandiol or undecyl residues (Saison-Behmoaras et al . , EMBO J. , 1991, 10 , 111; Kabanov et al . , FEBS Lett . , 1990, 259 , 327; Svinarchuk et al . , Biochimie, 1993, 75 , 49), a phospholipid, e.g., di-hexadecyl-rac-glycerol or triethylammonium 1,2-di-O- hexadecyl-rac-glycero-3 -H-phosphonate (Manoharan et al . , Tetrahedron Lett . , 1995, 36, 3651; Shea et al . , Nucl . Acids Res . , 1990, 18 , 3777), a polyamine or a polyethylene glycol chain (Manoharan et al . , Nucleosides & Nucleotides , 1995, 14 , 969), or adamantane acetic acid (Manoharan et al . ,
Tetrahedron Lett . , 1995, 36, 3651), a palmityl moiety (Mishra et al . , Biochim . Biophys . Acta , 1995, 1264 , 229), or an octadecylamine or hexylamino-carbonyl-oxycholesterol moiety (Crooke et al . , J. Pharmacol . Exp . Ther. , 1996, 277, 923). Oligonucleotides comprising lipophilic moieties, and methods for preparing such oligonucleotides, are disclosed in U.S. Patents No. 5,138,045, No. 5,218,105 and No . 5,459,255, the contents of which are hereby incorporated by reference.
The oligonucleotides of the invention may additionally or alternatively be prepared to be delivered in a "prodrug" form. The term "prodrug" indicates a therapeutic agent that is prepared in an inactive form that is converted to an active form ( i . e . , drug) within the body or cells thereof by the action of endogenous enzymes or other chemicals and/or conditions. In particular, prodrug versions of the oligonucleotides of the invention are prepared as SATE [ (S- acetyl-2-thioethyl) phosphate] derivatives according to the methods disclosed in WO 93/24510 to Gosselin et al . , published December 9, 1993. The present invention also includes oligonucleotides that are substantially chirally pure with regard to particular positions within the oligonucleotides. Examples of substantially chirally pure oligonucleotides include, but are not limited to, those having phosphorothioate linkages that are at least 75% Sp or Rp (Cook et al . , U.S. Patent No. 5,587,361, issued December 24, 1996) and those having substantially chirally pure (Sp or Rp) alkylphosphonate, phosphoamidate or phosphotriester linkages (Cook, U.S. Patent No. 5,212,295, issued May 18, 1993; Cook, U.S. Patent No. 5,521,302, issued May 28, 1996) .
The present invention also includes oligonucleotides which are chimeric oligonucleotides. "Chimeric" oligonucleotides or "chimeras," in the context of this invention, are oligonucleotides which contain two or more chemically distinct regions, each made up of at least one nucleotide. These oligonucleotides typically contain at least one region wherein the oligonucleotide is modified so as to confer upon the oligonucleotide increased resistance to nuclease degradation, increased cellular uptake, and/or increased binding affinity for the target nucleic acid. An additional region of the oligonucleotide may serve as a substrate for enzymes capable of cleaving RNA: DNA or RNA: RNA hybrids. By way of example, RNase H is a cellular endonuclease which cleaves the RNA strand of an RNA: DNA duplex. Activation of RNase H, therefore, results in cleavage of the RNA target, thereby greatly enhancing the efficiency of antisense inhibition of gene expression.
Cleavage of the RNA target can be routinely detected by gel electrophoresis and, if necessary, associated nucleic acid hybridization techniques known in the art. By way of example, such "chimeras" may be "gapmers," i . e . , oligonucleotides in which a central portion (the "gap") of the oligonucleotide serves as a substrate for, e . g. , RNase H, and the 5' and 3' portions (the "wings") are modified in such a fashion so as to have greater affinity for the target RNA molecule but are unable to support nuclease activity (e.g., 2'-fluoro- or 2 ' -methoxyethoxy substituted). Other chimeras include "wingmers," that is, oligonucleotides in which the 5' portion of the oligonucleotide serves as a substrate for, e . g. , RNase H, whereas the 3' portion is modified in such a fashion so as to have greater affinity for the target RNA molecule but is unable to support nuclease activity ( e . g. , 2'-fluoro- or 2 ' -methoxyethoxy substituted), or vice-versa . The oligonucleotides used in accordance with this invention may be conveniently and routinely made through the well-known technique of solid phase synthesis. Equipment for such synthesis is sold by several vendors including, for example, Applied Biosystems (Foster City, CA) . Any other means for such synthesis known in the art may additionally or alternatively be employed. It is also known to use similar techniques to prepare other oligonucleotides such as the phosphorothioates and alkylated derivatives. Teachings regarding the synthesis of particular modified oligonucleotides are hereby incorporated by reference from the following U.S. patents or pending patent applications, each of which is commonly assigned with this application: U.S. Patents Nos. 5,138,045 and 5,218,105, drawn to polyamine conjugated oligonucleotides; U.S. Patent No. 5,212,295, drawn to monomers for the preparation of oligonucleotides having chiral phosphorus linkages; U.S. Patents Nos. 5,378,825 and 5,541,307, drawn to oligonucleotides having modified backbones; U.S. Patent No. 5,386,023, drawn to backbone modified oligonucleotides and the preparation thereof through reductive coupling; U.S. Patent No. 5,457,191, drawn to modified nucleobases based on the 3-deazapurine ring system and methods of synthesis thereof; U.S. Patent No. 5,459,255, drawn to modified nucleobases based on N-2 substituted purines; U.S. Patent No. 5,521,302, drawn to processes for preparing oligonucleotides having chiral phosphorus linkages; U.S. Patent No. 5,539,082, drawn to peptide nucleic acids; U.S. Patent No. 5,554,746, drawn to oligonucleotides having b-lactam backbones; U.S. Patent No. 5,571,902, drawn to methods and materials for the synthesis of oligonucleotides; U.S. Patent No. 5,578,718, drawn to nucleosides having alkylthio groups, wherein such groups may be used as linkers to other moieties, attached at any of a variety of positions of the nucleoside; and U.S. Patent No. 5,587,361, drawn to oligonucleotides having phosphorothioate linkages of high chiral purity.
The oligonucleotides of the present invention can be utilized as therapeutic compounds, diagnostic tools and as research reagents and kits. The term "therapeutic uses" is intended to encompass prophylactic, palliative and curative uses wherein the oligonucleotides of the invention are contacted with animal cells either in vivo or ex vivo . When contacted with animal cells ex vivo, a therapeutic use includes incorporating such cells into an animal after treatment with one or more oligonucleotides of the invention.
For therapeutic uses, an animal suspected of having a disease or disorder which can be treated or prevented by modulating the expression or activity of a c-Fos or c-Jun protein is, for example, treated by administering oligonucleotides in accordance with this invention. The oligonucleotides of the invention can be utilized in pharmaceutical compositions by adding an effective amount of an oligonucleotide to a suitable pharmaceutically acceptable diluent or carrier. Workers in the field have identified antisense, triplex and other oligonucleotide compositions which are capable of modulating expression of genes implicated in viral, fungal and metabolic diseases. Antisense oligonucleotides have been safely administered to humans and several clinical trials are presently underway. It is thus established that oligonucleotides can be useful therapeutic instrumentalities that can be configured to be useful in treatment regimes for treatment of cells, tissues and animals, especially humans.
The oligonucleotides of the present invention can be further used to detect the presence of c-fos - or c-jun- specific nucleic acids in a cell or tissue sample. For example, radiolabeled oligonucleotides can be prepared by 32P labeling at the 5' end with polynucleotide kinase (Sambrook et al . , Molecular Cloning. A Labora tory Manual , Vol. 2, p. 10.59, Cold Spring Harbor Laboratory Press, 1989). Radiolabeled oligonucleotides are then contacted with cell or tissue samples suspected of containing c-fos or c-jun message RNAs (and thus c-Fos or c-Jun proteins) , and the samples are washed to remove unbound oligonucleotide. Radioactivity remaining in the sample indicates the presence of bound oligonucleotide, which in turn indicates the presence of nucleic acids complementary to the oligonucleotide, and can be quantitated using a scintillation counter or other routine means. Expression of nucleic acids encoding these proteins is thus detected.
Radiolabeled oligonucleotides of the present invention can also be used to perform autoradiography of tissues to determine the localization, distribution and quantitation of c-Fos or c-Jun proteins for research, diagnostic or therapeutic purposes. In such studies, tissue sections are treated with radiolabeled oligonucleotide and washed as described above, then exposed to photographic emulsion according to routine autoradiography procedures. The emulsion, when developed, yields an image of silver grains over the regions expressing a c-Fos or c-Jun gene.
Quantitation of the silver grains permits detection of the expression of mRNA molecules encoding these proteins and permits targeting of oligonucleotides to these areas. Analogous assays for fluorescent detection of expression of c-fos or c-jun nucleic acids can be developed using oligonucleotides of the present invention which are conjugated with fluorescein or other fluorescent tags instead of radiolabeling. Such conjugations are routinely accomplished during solid phase synthesis using fluorescently-labeled amidites or controlled pore glass (CPG) columns. Fluorescein-labeled amidites and CPG are available from, e . g. , Glen Research (Sterling, VA) .
The present invention employs oligonucleotides targeted to nucleic acids encoding c-Fos or c-Jun proteins and oligonucleotides targeted to nucleic acids encoding such proteins. Kits for detecting the presence or absence of expression of a c-Fos and/or c-Jun protein may also be prepared. Such kits include an oligonucleotide targeted to an appropriate gene, i.e., a gene encoding a c-Fos or c-Jun protein. Appropriate kit and assay formats, such as, e . g. , "sandwich" assays, are known in the art and can easily be adapted for use with the oligonucleotides of the invention. Hybridization of the oligonucleotides of the invention with a nucleic acid encoding a c-Fos or c-Jun protein can be detected by means known in the art . Such means may include conjugation of an enzyme to the oligonucleotide, radiolabelling of the oligonucleotide or any other suitable detection systems. Kits for detecting the presence or absence of a c-Fos or c-Jun protein may also be prepared.
In the context of this invention, "hybridization" means hydrogen bonding, which may be Watson-Crick, Hoogsteen or reversed Hoogsteen hydrogen bonding, between complementary nucleotides. For example, adenine and thymine are complementary nucleobases which pair through the formation of hydrogen bonds. "Complementary," as used herein, refers to the capacity for precise pairing between two nucleotides. For example, if a nucleotide at a certain position of an oligonucleotide is capable of hydrogen bonding with a nucleotide at the same position of a DNA or RNA molecule, then the oligonucleotide and the DNA or RNA are considered to be complementary to each other at that position. The oligonucleotide and the DNA or RNA are complementary to each other when a sufficient number of corresponding positions in each molecule are occupied by nucleotides which can hydrogen bond with each other. Thus, "specifically hybridizable" and "complementary" are terms which are used to indicate a sufficient degree of complementarity or precise pairing such that stable and specific binding occurs between the oligonucleotide and the DNA or RNA target. It is understood in the art that an oligonucleotide need not be 100% complementary to its target DNA sequence to be specifically hybridizable. An oligonucleotide is specifically hybridizable when binding of the oligonucleotide to the target DNA or RNA molecule interferes with the normal function of the target DNA or RNA to cause a decrease or loss of function, and there is a sufficient degree of complementarity to avoid non-specific binding of the oligonucleotide to non-target sequences under conditions in which specific binding is desired, i.e., under physiological conditions in the case of in vivo assays or therapeutic treatment, or in the case of in vi tro assays, under conditions in which the assays are performed.
The formulation of therapeutic compositions and their subsequent administration is believed to be within the skill of those in the art. In general, for therapeutics, a patient in need of such therapy is administered an oligonucleotide in accordance with the invention, commonly in a pharmaceutically acceptable carrier, in doses ranging from 0.01 ug to 100 g per kg of body weight depending on the age of the patient and the severity of the disorder or disease state being treated. Further, the treatment regimen may last for a period of time which will vary depending upon the nature of the particular disease or disorder, its severity and the overall condition of the patient, and may extend from once daily to once every 20 years. Following treatment, the patient is monitored for changes in his/her condition and for alleviation of the symptoms of the disorder or disease state . The dosage of the oligonucleotide may either be increased in the event the patient does not respond significantly to current dosage levels, or the dose may be decreased if an alleviation of the symptoms of the disorder or disease state is observed, or if the disorder or disease state has been abated.
In some cases it may be more effective to treat a patient with an oligonucleotide of the invention in conjunction with other traditional therapeutic modalities in order to increase the efficacy of a treatment regimen. In the context of the invention, the term "treatment regimen" is meant to encompass therapeutic, palliative and prophylactic modalities. For example, a patient may be treated with conventional chemotherapeutic agents, particularly those used for tumor and cancer treatment. Examples of such chemotherapeutic agents include but are not limited to daunorubicin, daunomycin, dactinomycin, doxorubicin, epirubicin, idarubicin, esorubicin, bleomycin, mafosfamide, ifosfamide, cytosine arabinoside, bis-chloroethylnitrosurea, busulfan, mitomycin C, actinomycin D, mithramycin, prednisone, hydroxyprogesterone, testosterone, tamoxifen, dacarbazine, procarbazine, hexamethylmelamine, pentamethylmelamine, mitoxantrone, amsacrine, chlorambucil , methylcyclohexylnitrosurea, nitrogen mustards, melphalan, cyclophosphamide, 6-mercaptopurine, 6-thioguanine, cytarabine (CA) , 5-azacytidine, hydroxyurea, deoxycoformycin, 4- hydroxyperoxycyclophosphoramide, 5-fluorouracil (5-FU) , 5- fluorodeoxyuridine (5-FUdR) , methotrexate (MTX) , colchicine, vincristine, vinblastine, etoposide, trimetrexate, teniposide, cisplatin and diethylstilbestrol (DES) . See, generally, The Merck Manual of Diagnosis and Therapy, 15th Ed., pp. 1206-1228, Berkow et al . , eds., Rahay, N.J., 1987). When used with the compounds of the invention, such chemotherapeutic agents may be used individually ( e . g . , 5-FU and oligonucleotide), sequentially ( e . g. , 5-FU and oligonucleotide for a period of time followed by MTX and oligonucleotide) , or in combination with one or more other such chemotherapeutic agents ( e . g. , 5-FU, MTX and oligonucleotide, or 5-FU, radiotherapy and oligonucleotide) . In another preferred embodiment of the invention, a first antisense oligonucleotide targeted to c-fos is used in combination with a second antisense oligonucleotide targeted to c-jun in order to modulate AP-1 molecules to a more extensive degree than can be achieved when either oligonucleotide used individually.
Following successful treatment, it may be desirable to have the patient undergo maintenance therapy to prevent the recurrence of the disease state, wherein the oligonucleotide is administered in maintenance doses, ranging from 0.01 ug to 100 g per kg of body weight, once or more daily, to once every 20 years. In the case of in individual known or suspected of being prone to a neoplastic or malignant condition, prophylactic effects may be achieved by administration of preventative doses, ranging from 0.01 ug to 100 g per kg of body weight, once or more daily, to once every 20 years.
The pharmaceutical compositions of the present invention may be administered in a number of ways depending upon whether local or systemic treatment is desired and upon the area to be treated. Within the context of the invention, "administration" indicates the topical (including ophthalmic, vaginal, rectal, intranasal, transdermal) , oral or parenteral contacting of an oligonucleotide, or pharmaceutical composition comprising an oligonucleotide, with cells, tissues or organs of a mammal including a human. Parenteral administration includes intravenous drip; subcutaneous, intraperitoneal, intravitreal or intramuscular injection; and intrathecal or intraventricular administration as herein described.
Formulations for topical administration may include transdermal patches, ointments, lotions, creams, gels, drops, suppositories, sprays, liquids and powders. Conventional pharmaceutical carriers, nucleic acid carriers, aqueous, powder or oily bases, thickeners and the like may be necessary or desirable in certain instances. Topical administration also includes the delivery of oligonucleotides into the epidermis of a mammal by electroporation (Zewert et al . , WO 96/39531, published December 12, 1996).
Compositions for oral administration include powders or granules, suspensions or solutions in water or non-aqueous media, capsules, sachets or tablets. Thickeners, flavoring agents, diluents, emulsifiers, dispersing aids or binders may be desirable.
Intravitreal injection, for the direct delivery of drug to the vitreous humor of a mammalian eye, is described in U.S. Patent No. 5,595,978, which issued January 21, 1997, and which is assigned to the same assignee as the instant application, the contents of which are hereby incorporated by reference .
Intraluminal drug administration, for the direct delivery of drug to an isolated portion of a tubular organ or tissue (e.g., such as an artery, vein, ureter or urethra), may be desired for the treatment of patients with diseases or conditions afflicting the lumen of such organs or tissues. To effect this mode of oligonucleotide administration, a catheter or cannula is surgically introduced by appropriate means. For example, for treatment of the left common carotid artery, a cannula is inserted thereinto via the external carotid artery. After isolation of a portion of the tubular organ or tissue for which treatment is sought, a composition comprising the oligonucleotides of the invention is infused through the cannula or catheter into the isolated segment . After incubation for from about 1 to about 120 minutes, during which the oligonucleotide is taken up by cells of the interior lumen of the vessel, the infusion cannula or catheter is removed and flow within the tubular organ or tissue is restored by removal of the ligatures which effected the isolation of a segment thereof (Morishita et al . , Proc . Na tl . Acad . Sci . (U. S . A . ) , 1993, 90 , 8474) . Antisense oligonucleotides may also be combined with a biocompatible matrix or carrier, such as a hydrogel material, and applied directly to vascular tissue in vivo (Rosenberg et al . , U.S. Patent No. 5,593,974, issued January 14, 1997). Intraventricular drug administration, for the direct delivery of drug to the brain of a patient, may be desired for the treatment of patients with diseases or conditions afflicting the brain. To effect this mode of oligonucleotide administration, a silicon catheter is surgically introduced into a ventricle of the brain of a human patient, and is connected to a subcutaneous infusion pump (Medtronic Inc., Minneapolis, MN) that has been surgically implanted in the abdominal region (Zimm et al . , Cancer Research, 1984, 44 , 1698; Shaw, Cancer, 1993, 72 (11 Suppl . ) , 3416). The pump is used to inject the oligonucleotides and allows precise dosage adjustments and variation in dosage schedules with the aid of an external programming device. The reservoir capacity of the pump is 18-20 mL and infusion rates may range from 0.1 mL/h to 1 mL/h. Depending on the frequency of administration, ranging from daily to monthly, and the dose of drug to be administered, ranging from 0.01 μg to 100 g per kg of body weight, the pump reservoir may be refilled at 3-10 week intervals. Refilling of the pump is accomplished by percutaneous puncture of the self-sealing septum of the pump.
Intrathecal drug administration for the introduction of drug into the spinal column of a patient may be desired for the treatment of patients with diseases of the central nervous system. To effect this route of oligonucleotide administration, a silicon catheter is surgically implanted into the L3-4 lumbar spinal interspace of a human patient, and is connected to a subcutaneous infusion pump which has been surgically implanted in the upper abdominal region (Luer and Hatton, The Annals of Pharmacotherapy , 1993, 27, 912; Ettinger et al . , 1978, Cancer, 41 , 1270, 1978; Yaida et al . , Regul . Pept . , 1995, 59 , 193). The pump is used to inject the oligonucleotides and allows precise dosage adjustments and variations in dose schedules with the aid of an external programming device. The reservoir capacity of the pump is 18-20 mL, and infusion rates may vary from 0.1 mL/h to 1 mL/h. Depending on the frequency of drug administration, ranging from daily to monthly, and dosage of drug to be administered, ranging from 0.01 μg to 100 g per kg of body weight, the pump reservoir may be refilled at 3-10 week intervals. Refilling of the pump is accomplished by a single percutaneous puncture to the self-sealing septum of the pump. The distribution, stability and pharmacokinetics of oligonucleotides within the central nervous system may be followed according to known methods (Whitesell et al . , Proc . Natl . Acad. Sci . (USA) , 1993, 90 , 4665).
To effect delivery of oligonucleotides to areas other than the brain or spinal column via this method, the silicon catheter is configured to connect the subcutaneous infusion pump to, e.g., the hepatic artery, for delivery to the liver
(Kemeny et al . , Cancer, 1993, 71:1964). Infusion pumps may also be used to effect systemic delivery of oligonucleotides
(Ewel et al . , Cancer Research, 1992, 52:3005; Rubenstein et al . , J. Surg. Oncol . , 1996, 62:194).
Regardless of the method by which the oligonucleotides of the invention are introduced into a patient, colloidal dispersion systems may be used as delivery vehicles to enhance the in vivo stability of the oligonucleotides and/or to target the oligonucleotides to a particular organ, tissue or cell type. Colloidal dispersion systems include, but are not limited to, macromolecule complexes, nanocapsules , microspheres , beads and lipid-based systems including oil-in- water emulsions, micelles, mixed micelles and liposomes. A preferred colloidal dispersion system is a plurality of liposomes, artificial membrane vesicles which may be used as cellular delivery vehicles for bioactive agents in vi tro and in vivo (Mannino et al . , Biotechniques , 1988, _, 682; Blume and Cevc, Biochem . et Biophys . Acta, 1990, 1029 , 91; Lappalainen et al . , Antiviral Res . , 1994, 23 , 119; Chonn and Cullis, Current Op . Biotech . , 1995, 6, 698). It has been shown that large unilamellar vesicles (LUV) , which range in size from 0.2-0.4 μm, can encapsulate a substantial percentage of an aqueous buffer containing large macromolecules . RNA, DNA and intact virions can be encapsulated within the aqueous interior and delivered to brain cells in a biologically active form (Fraley et al . ,
Trends Biochem . Sci . , 1981, 6, 77) . The composition of the liposome is usually a combination of lipids, particularly phospholipids, in particular, high phase transition temperature phospholipids, usually in combination with one or more steroids, particularly cholesterol. Examples of lipids useful in liposome production include phosphatidyl compounds, such as phosphatidylglycerol, phosphatidylcholine, phosphatidylserine, phosphatidylethanolamine, sphingolipids , cerebrosides and gangliosides . Particularly useful are diacyl phosphatidylglycerols, where the lipid moiety contains from 14-18 carbon atoms, particularly from 16-18 carbon atoms, and is saturated (lacking double bonds within the 14- 18 carbon atom chain) . Illustrative phospholipids include phosphatidylcholine, dipalmitoylphosphatidylcholine and distearoylphosphatidylcholine .
The targeting of colloidal dispersion systems, including liposomes, can be either passive or active. Passive targeting utilizes the natural tendency of liposomes to distribute to cells of the reticuloendothelial system in organs that contain sinusoidal capillaries. Active targeting, by contrast, involves modification of the liposome by coupling thereto a specific ligand such as a viral protein coat (Morishita et al . , Proc. Na tl . Acad . Sci . (U. S . A . ) , 1993, 90 , 8474), monoclonal antibody (or a suitable binding portion thereof) , sugar, glycolipid or protein (or a suitable oligopeptide fragment thereof) , or by changing the composition and/or size of the liposome in order to achieve distribution to organs and cell types other than the naturally occurring sites of localization. The surface of the targeted colloidal dispersion system can be modified in a variety of ways. In the case of a liposomal targeted delivery system, lipid groups can be incorporated into the lipid bilayer of the liposome in order to maintain the targeting ligand in close association with the lipid bilayer. Various linking groups can be used for joining the lipid chains to the targeting ligand. The targeting ligand, which binds a specific cell surface molecule found predominantly on cells to which delivery of the oligonucleotides of the invention is desired, may be, for example, (1) a hormone, growth factor or a suitable oligopeptide fragment thereof which is bound by a specific cellular receptor predominantly expressed by cells to which delivery is desired or (2) a polyclonal or monoclonal antibody, or a suitable fragment thereof (e.g., Fab; F(ab')2) which specifically binds an antigenic epitope found predominantly on targeted cells. Two or more bioactive agents (e.g., an oligonucleotide and a conventional drug; two oligonucleotides) can be combined within, and delivered by, a single liposome. It is also possible to add agents to colloidal dispersion systems which enhance the intercellular stability and/or targeting of the contents thereof. Compositions for parenteral, intrathecal or intraventricular administration, or colloidal dispersion systems, may include sterile aqueous solutions which may also contain buffers, diluents and other suitable additives. Dosing is dependent on severity and responsiveness of the disease state to be treated, with the course of treatment lasting from several days to several months, or until a cure is effected or a diminution of the disease state is achieved. Optimal dosing schedules can be calculated from measurements of drug accumulation in the body of the patient. Persons of ordinary skill can easily determine optimum dosages, dosing methodologies and repetition rates. Optimum dosages may vary depending on the relative potency of individual oligonucleotides, and can generally be estimated based on EC50s found to be effective in in vi tro and in vivo animal models. In general, dosage is from 0.01 μg to 100 g per kg of body weight, and may be given once or more daily, weekly, monthly or yearly, or even once every 2 to 20 years.
The following examples illustrate the invention and are not intended to limit the same. Those skilled in the art will recognize, or be able to ascertain through routine experimentation, numerous equivalents to the specific substances and procedures described herein. Such equivalents are considered to be within the scope of the present invention.
EXAMPLES
Example 1: Chemical Synthesis and Nucleotide Sequence of Oligonucleotides
General Synthetic Techniques: Oligonucleotides were synthesized on an automated DNA synthesizer using standard phosphoramidite chemistry with oxidation using iodine. β- Cyanoethyldiisopropyl phosphoramidites were purchased from Applied Biosystems (Foster City, CA) . For phosphorothioate oligonucleotides, the standard oxidation bottle was replaced by a 0.2 M solution of 3H-1, 2-benzodithiole-3 -one-1, 1-dioxide in acetonitrile for the stepwise thiation of the phosphite linkages . The synthesis of 2'-0-methyl- (a.k.a. 2'-methoxy-) phosphorothioate oligonucleotides was according to the procedures set forth above substituting 2'-0-methyl β- cyanoethyldiisopropyl phosphoramidites (Chemgenes, Needham, MA) for standard phosphoramidites and increasing the wait cycle after the pulse delivery of tetrazole and base to 360 seconds .
Similarly, 2'-0-propyl- (a.k.a 2'-propoxy-) phosphorothioate oligonucleotides were prepared by slight modifications of this procedure and essentially according to procedures disclosed in U.S. patent application Serial No. 08/383,666, filed February 3, 1995, which is assigned to the same assignee as the instant application and which is incorporated by reference herein.
The 2 ' -fluoro-phosphorothioate oligonucleotides of the invention were synthesized using 5 ' -dimethoxytrityl-3 ' - phosphoramidites and prepared as disclosed in U.S. patent application Serial No. 08/383,666, filed February 3, 1995, and U.S. Patent 5,459,255, which issued October 8, 1996, both of which are assigned to the same assignee as the instant application and which are incorporated by reference herein. The 2 ' -fluoro-oligonucleotides were prepared using phosphoramidite chemistry and a slight modification of the standard DNA synthesis protocol (i.e., deprotection was effected using methanolic ammonia at room temperature) .
The 2 ' -methoxyethoxy oligonucleotides were synthesized essentially according to the methods of Martin et al . ( Helv. Chim . Acta , 1995, 78 , 486). For ease of synthesis, the 3' nucleotide of the 2 ' -methoxyethoxy oligonucleotides was a deoxynucleotide, and 2 ' -0-CH2CH2OCH3_cytosines were 5-methyl cytosines, which were synthesized according to the procedures described below.
PNA antisense analogs were prepared essentially as described in U.S. Patents Nos. 5,539,082 and 5,539,083, both of which (1) issued July 23, 1996, (2) are assigned to the same assignee as the instant application and (3) are incorporated by reference herein.
Oligonucleotides comprising 2 , 6-diaminopurine were prepared essentially as described in U.S. Patents No. 5,506,351 which issued April 9, 1996, is assigned to the same assignee as the instant application and which is incorporated by reference herein. Oligonucleotides comprising 2,6- diaminopurine can also be prepared by enzymatic means (Bailly et al . , Proc . Na tl . Acad . Sci . U. S . A . , 1996, 53:13623) .
After cleavage from the controlled pore glass (CPG) column (Applied Biosystems) and deblocking in concentrated ammonium hydroxide, at 55°C for 18 hours, the oligonucleotides were purified by precipitation 2x from 0.5 M NaCI with 2.5 volumes of ethanol. Analytical gel electrophoresis was accomplished in 20% acrylamide, 8 M urea and 45 mM Tris-borate buffer (pH 7) .
Synthesis of 5 -Methyl Cytosine Monomers:
2 , 2 ' -Anhydro [1- (jS-D-arabinofuranosyl) -5-methyluridine] : 5- Methyluridine (ribosylthymine, commercially available through Yamasa, Choshi, Japan) (72.0 g, 0.279 M) , diphenylcarbonate (90.0 g, 0.420 M) and sodium bicarbonate (2.0 g, 0.024 M) were added to DMF (300 mL) . The mixture was heated to reflux, with stirring, allowing the evolved carbon dioxide gas to be released in a controlled manner. After 1 hour, the sli-ghtly darkened solution was concentrated under reduced pressure. The resulting syrup was poured into diethylether (2.5 L) , with stirring. The product formed a gum. The ether was decanted and the residue was dissolved in a minimum amount of methanol (ca. 400 mL) . The solution was poured into fresh ether (2.5 L) to yield a stiff gum. The ether was decanted and the gum was dried in a vacuum oven (60°C at 1 mm Hg for 24 h) to give a solid which was crushed to a light tan powder (57 g, 85% crude yield) . The material was used- as is for further reactions.
2' -O-Methoxyethyl-5 -methyluridine: 2,2' -Anhydro-5- methyluridine (195 g, 0.81 M) , tris (2-methoxyethyl) borate (231 g, 0.98 M) and 2 -methoxyethanol (1.2 L) were added to a 2 L stainless steel pressure vessel and placed in a pre- heated oil bath at 160°C. After heating for 48 hours at 155- 160°C, the vessel was opened and the solution evaporated to dryness and triturated with MeOH (200 mL) . The residue was suspended in hot acetone (1 L) . The insoluble salts were filtered, washed with acetone (150 mL) and the filtrate evaporated. The residue (280 g) was dissolved in CH3CN (600 mL) and evaporated. A silica gel column (3 kg) was packed in CH2Cl2/acetone/MeOH (20:5:3) containing 0.5% Et3NH. The residue was dissolved in CH2C12 (250 mL) and adsorbed onto silica (150 g) prior to loading onto the column. The product was eluted with the packing solvent to give 160 g (63%) of product . 2' -O-Methoxyethyl-5' -O-dimethoxytrityl-5-methyluridine: 2'- O-Methoxyethyl- 5 -methyluridine (160 g, 0.506 M) was co- evaporated with pyridine (250 mL) and the dried residue dissolved in pyridine (1.3 L) . A first aliquot of dimethoxytrityl chloride (94.3 g, 0.278 M) was added and the mixture stirred at room temperature for one hour. A second aliquot of dimethoxytrityl chloride (94.3 g, 0.278 M) was added and the reaction stirred for an additional one hour. Methanol (170 mL) was then added to stop the reaction. HPLC showed the presence of approximately 70% product. The solvent was evaporated and triturated with CH3CN (200 mL) .
The residue was dissolved in CHC13 (1.5 L) and extracted with 2x 500 mL of saturated NaHC03 and 2x 500 mL of saturated NaCI. The organic phase was dried over Na2S04, filtered and evaporated. 275 g of residue was obtained. The residue was purified on a 3.5 kg silica gel column, packed and eluted with EtOAc/Hexane/Acetone (5:5:1) containing 0.5% Et3NH. The pure fractions were evaporated to give 164 g of product. Approximately 20 g additional was obtained from the impure fractions to give a total yield of 183 g (57%) . 3' -O-Acetyl-2' -O-methoxyethyl-5 ' -O-dimethoxytrityl-5- methyluridine: 2' -O-Methoxyethyl -5 ' -O-dimethoxytrityl-5- methyluridine (106 g, 0.167 M) , DMF/pyridine (750 mL of a 3:1 mixture prepared from 562 mL of DMF and 188 mL of pyridine) and acetic anhydride (24.38 mL, 0.258 M) were combined and stirred at room temperature for 24 hours. The reaction was monitored by tic by first quenching the tic sample with the addition of MeOH . Upon completion of the reaction, as judged by tic, MeOH (50 mL) was added and the mixture evaporated at 35°C. The residue was dissolved in CHC13 (800 mL) and extracted with 2x 200 mL of saturated sodium bicarbonate and 2x 200 mL of saturated NaCI. The water layers were back extracted with 200 mL of CHC13. The combined organics were dried with sodium sulfate and evaporated to give 122 g of residue (approximately 90% product) . The residue was purified on a 3.5 kg silica gel column and eluted using EtOAc/Hexane (4:1) . Pure product fractions were evaporated to yield 96 g (84%) .
3' -O-Acetyl-2' -O-methoxyethyl-5' -0-dimethoxytrityl-5-methyl- 4-triazoleuridine: A first solution was prepared by dissolving 3 ' -O-acetyl-2 ' -O-methoxyethyl-5 ' -O- dimethoxytrityl- 5-methyluridine (96 g, 0.144 M) in CH3CN (700 mL) and set aside. Triethylamine (189 mL, 1.44 M) was added to a solution of triazole (90 g, 1.3 M) in CH3CN (1 L) , cooled to -5°C and stirred for 0.5 h using an overhead stirrer. P0C13 was added dropwise, over a 30 minute period, to the stirred solution maintained at 0-10°C, and the resulting mixture stirred for an additional 2 hours. The first solution was added dropwise, over a 45 minute period, to the later solution. The resulting reaction mixture was stored overnight in a cold room. Salts were filtered from the reaction mixture and the solution was evaporated. The residue was dissolved in EtOAc (1 L) and the insoluble solids were removed by filtration. The filtrate was washed with lx 300 mL of NaHC03 and 2x 300 mL of saturated NaCI, dried over sodium sulfate and evaporated. The residue was triturated with EtOAc to give the title compound. 2' -O-Methoxyethyl-5' -O-dimethoxytrityl- 5-methylcytidine: A solution of 3 ' -O-acetyl-2' -O-methoxyethyl-5 ' -O-dimethoxy- trityl-5-methyl-4-triazoleuridine (103 g, 0.141 M) in dioxane (500 mL) and NH40H (30 mL) was stirred at room temperature for 2 hours. The dioxane solution was evaporated and the residue azeotroped with MeOH (2x 200 mL) . The residue was dissolved in MeOH (300 mL) and transferred to a 2 liter stainless steel pressure vessel. Methanol (400 mL) saturated with NH3 gas was added and the vessel heated to 100°C for 2 hours (thin layer chromatography, tic, showed complete conversion) . The vessel contents were evaporated to dryness and the residue was dissolved in EtOAc (500 mL) and washed once with saturated NaCI (200 mL) . The organics were dried over sodium sulfate and the solvent was evaporated to give 85 g (95%) of the title compound.
N4-Benzoyl-2 ' -O-methoxyethyl-5' -O-dimethoxytrityl-5- methylcytidine: 2' -O-Methoxyethyl-5 ' -O-dimethoxytrityl-5- methylcytidine (85 g, 0.134 M) was dissolved in DMF (800 mL) and benzoic anhydride (37.2 g, 0.165 M) was added with stirring. After stirring for 3 hours, tic showed the reaction to be approximately 95% complete. The solvent was evaporated and the residue azeotroped with MeOH (200 mL) . The residue was dissolved in CHC13 (700 mL) and extracted with saturated NaHC03 (2x 300 mL) and saturated NaCI (2x 300 mL) , dried over MgS04 and evaporated to give a residue (96 g) . The residue was chromatographed on a 1.5 kg silica column using EtOAc/Hexane (1:1) containing 0.5% Et3NH as the eluting solvent. The pure product fractions were evaporated to give 90 g (90%) of the title compound. N4-Benzoyl-2 ' -O-methoxyethyl-5' -O-dimethoxytrityl-5- methylcytidine-3 ' -amidite: N4-Benzoyl -2 ' -O-methoxyethyl-5 ' - O-dimethoxytrityl- 5 -methylcytidine (74 g, 0.10 M) was dissolved in CH2C12 (1 L) . Tetrazole diisopropylamine (7.1 g) and 2-cyanoethoxy-tetra (isopropyl) phosphite (40.5 mL, 0.123 M) were added with stirring, under a nitrogen atmosphere. The resulting mixture was stirred for 20 hours at room temperature (tic showed the reaction to be 95% complete) . The reaction mixture was extracted with saturated NaHC03 (lx 300 mL) and saturated NaCI (3x 300 mL) . The aqueous washes were back-extracted with CH2C12 (300 mL) , and the extracts were combined, dried over MgSO„ and concentrated. The residue obtained was chromatographed on a 1.5 kg silica column using EtOAc\Hexane (3:1) as the eluting solvent. The pure fractions were combined to give 90.6 g (87%) of the title compound. Nucleotide Sequences: Table 1 shows the sequence and activity of each of the oligonucleotides evaluated for inhibition of c-jun mRNA expression, and Table 2 shows the sequence and activity of the oligonucleotides evaluated for inhibition of c-fos mRNA expression. Oligonucleotide activities were evaluated as described infra in Example 2 et seq. For the nucleotide sequence of the human c-jun gene, see Hattori et al . , Proc. Natl. Acad. Sci. U.S.A., 1988, 85:9148, and Genbank accession No. J04111 ( "HUMJUNA" ) . The nucleotide sequence of the human c-fos gene is described by Van Straaten et al . , Proc. Natl. Acad. Sci. U.S.A. , 1983, 80:3183, and in Genbank accession No. K00650 ("HUMFOS") .
TABLE 1 Phosphorothioate Oligonucleotides Targeted to Human c -jun
Figure imgf000039_0001
Control is TPA induction, at 1 hour, in A549 cells N.D., not determined.
TABLE 2 Phosphorothioate Oligonucleotides Targeted to Human c -fos
Figure imgf000040_0001
Figure imgf000041_0001
) &
"Control is TPA induction, at 1 hour, in A549 cells.
Example 2 : Screening for Oligonucleotides that Modulate mRNA Expression of the AP-1 Subunits c-fos and c-jun
In order to evaluate the activity of potential c-fos and c-jun modulating oligonucleotides, A549 cells were grown in T-75 flasks until 80-90% confluent. (Cell line A549 is available from, inter alia , the American Type Culture Collection, Rockville, MD, as ATCC No. CCL-185.) At this time, the cells were washed twice with 10 mL of media (DMEM) , followed by the addition of 5 mL of DMEM containing 20 μg/mL of LIPOFECTIN™ (i.e., DOTMA/DOPE (N-[l-(2,3- dioleyloxy) propyl] -N, N,N-triethylammonium chloride/dioleoylphosphatidyl ethanolamine) ) . The oligonucleotides were added from a 10 μM stock solution to a final concentration of 400 nM, and the two solutions were mixed by swirling the flasks. After 4 hours at 37°C, the medium was replaced with DMEM containing 10% serum. At this point, 1 μM 12-O-tetradecanoylphorbol 13 -acetate (TPA) was added to induce expression of c-fos and c-jun . Cells were extracted in guanidinium one hour later, and the c-fos and c- jun mRNA expression was determined by Northern blotting.
Probes for human c-jun and c-fos were PCR products prepared using primers based on the published sequences thereof (respectively, Hattori et al . , Proc . Na tl . Acad . Sci . U. S . A . , 1988, 85:9148, and Van Straaten et al . , Proc . Natl . Acad . Sci . U. S. A . , 1983, 80:3183).
As described in Table 1, for inhibiting c-jun mRNA expression, ISIS 10580, 10581, 10582 and 10583 were most active (81%, 76%, 83% and 80% inhibition, respectively) . Treatment of cells with these oligonucleotides reduced c-jun expression to 19%, 24%, 17% and 20% (81%, 76%, 83% and 80% inhibition) , respectively, of the level seen in control experiments (100% expression, 0% inhibition) . These oligonucleotides effect significant inhibition of c-jun and are therefor preferred. Basal levels of c-jun mRNA are typically about 30% of the control value; ISIS 10572, 10578 and 10579 reduce c-jun levels to near basal levels (39%, 37% and 41%, respectively) and are thus also preferred. As described in Table 2, the oligonucleotides most effective in reducing c-fos mRNA expression are ISIS 10633, 10635, 10638 and 10639. Treatment of cells with these oligonucleotides reduced c-fos expression to 24%, 12%, 22% and 9% (76%, 88%, 78% and 91% inhibition) , respectively, of the level seen in control experiments (100% expression, 0% inhibition) ; basal levels of c-fos mRNA are typically about 3% of the control value. These oligonucleotides effect significant inhibition of c-fos and are therefor preferred.
Example 3 : Dose Response and Specificity of Oligonucleotides Targeted to AP-1 Subunits
Dose-response experiments were performed at different oligonucleotide concentrations to determine the potency (i.e., ability to decrease expression of the appropriate mRNA target) of the most active compounds identified in the initial screen (Tables 3 and 4) . The decreases in target mRNA expression effected by ISIS 10582 ( c-jun) and ISIS 10639 ( c-fos) are dose-dependent, as shown in Tables 3 and 4, respectively .
TABLE 3
Dose-Response to Oligonucleotides Targeted to c -j un
Figure imgf000043_0001
"Control is TPA induction, at 1 hour, in A549 cells TABLE 4 Dose-Response to Oligonucleotides Targeted to c -fos
Figure imgf000044_0001
'Control is TPA induction, at 1 hour, in A549 cells.
The specificity of the oligonucleotide-mediated inhibition of c-fos and c-jun mRNA expression was further examined by determining the effects of scrambled, 6- or 7- base mismatch and sense control versions of the most active oligonucleotides, ISIS 10582 ( c-jun) and ISIS 10639 ( c-fos) . As can be seen in Table 5, ISIS 10582 exhibited potent and specific inhibition of c-jun mRNA expression, but ISIS 11562 (sense version of ISIS 10582; SEQ ID NO:29), ISIS 11564 (6 base pair mismatch version of ISIS 10639; SEQ ID NO: 31) and ISIS 11563 (scrambled version of ISIS 10639; SEQ ID NO:30) had no detectable effect on c-jun mRNA levels during TPA induction (the sequences of ISIS 11562-11564 are given in Table 1) .
As can further be seen in Table 5, ISIS 10639 exhibited potent and specific inhibition of c-fos mRNA expression, but ISIS 11184 (sense version of ISIS 10639; SEQ ID NO:32) , ISIS 11185 (7 base pair mismatch version of ISIS 10639; SEQ ID NO:33) and ISIS 11186 (scrambled version of ISIS 10639; SEQ ID NO: 34) had no detectable effect on c-fos mRNA levels during TPA induction (the sequences of ISIS
11184-11186 are given in Table 2) . Finally, it can also be seen from Table 5 that neither active oligonucleotide has any detectable effect on mRNA levels of the other active oligonucleotide' s target. That is, ISIS 10639, targeted to c-fos , has no detectable effect on c-jun levels; similarly, ISIS 10582, targeted to c-jun, has no detectable effect on c- fos levels.
TABLE 5
Specificity of c -fos and c -jun Oligonucleotides
Figure imgf000045_0001
These results demonstrate that ISIS 10582 effects potent and specific modulation (i.e., inhibition) of c-jun mRNA levels and that ISIS 10639 effects potent and specific modulation of c-fos mRNA levels.
Example 4: Effect of Oligonucleotides Targeted to an AP-1 Subunit on Human Tumor Growth in Nude Mice
In order to evaluate the in vivo activity of c-fos oligonucleotides, 25 mg of tumor fragments of A549 tumors were implanted subcutaneously in nude mice (n = 6) . ISIS 10639 was administered daily, i.v., for three weeks. The oligonucleotide dosage was 25 mg/kg. Tumor size was recorded weekly, and the results are shown in Table 6. A substantial reduction in tumor growth rate was obtained upon treatment with ISIS 10639. By day 34, saline-treated tumors were 0.56 +. 0.12 g by weight, while tumors treated with ISIS 10639 were 0.31 ± 0.1 g by weight.
TABLE 6 Response of Transplanted Tumors in Mice to Oligonucleotides Targeted to c -jun
Figure imgf000046_0001
Example 5 : Effect of Oligonucleotides on Protein Levels of AP-1 Subunits
The ability of the c-fos active oligonucleotide ISIS 10639 to reduce expression of the c-Fos protein was examined as follows. A549 cells were treated with oligonucleotides as in Examples 2-3, except that induction of c-Fos was effected by treatment of cells with TPA (1 uM) for three hours. At this time, whole cell protein was extracted in SDS (sodium dodecyl sulfate) buffer. Samples of extracts were electrophoresed, transferred to nitrocellulose filters which were immunoblotted using a c-Fos-specific antibody (Santa Cruz AB, Santa Cruz, CA) . The results (Table 7) demonstrate that treatment of cells with the c-fos antisense oligonucleotide results in basal levels of c-Fos protein. TABLE 7 Effect of c -fos Oligonucleotides on c -Fos Protein Levels
Figure imgf000047_0001
Example 6: Modified Oligonucleotides and PNA Antisense Analogs to Human AP-1 Subunits
Once oligonucleotides that modulate c-Fos or c-Jun are identified, derivative or modified oligonucleotides having the same sequence thereas are prepared. In order to evaluate the effect of chemical modifications to oligonucleotides to c-fos and c-jun, the modified oligonucleotides described in Tables 8 and 9 were prepared. The effect of the c-fos- targeted oligonucleotides on c-fos RNA levels were evaluated as described in Examples 2-3. The results (Table 10) demonstrate that some enhancement of c-fos modulation can be achieved by the use modifications such as, e.g., 2'-fluoro (ISIS 11200) . Other modified oligonucleotides of the invention are evaluated in like fashion. In order to evaluate the effect of PNA antisense analogs, the PNA analogs of the invention are introduced into appropriate cell lines by microinjection according to the method of Hanvey e t al . ( Science, 1992, 258:1481) . Intracellular delivery of PNA analogs is confirmed by the use of a fluorescently tagged PNA antisense analog conjugate such as, e.g., ISIS 14240. TABLE 8 Additional Oligonucleotides and PNA Antisense Analogs Targeted to Human AP-1 Subunits
Figure imgf000048_0001
Figure imgf000049_0001
_» α>
Figure imgf000050_0001
SO
Figure imgf000051_0001
* Phosphorothioate linkages are indicated by " s " and "P=S" ; emboldened residues indicate the additional indicated modifications: 2'F = 2'-fluoro-; 2'propoxy = 2 ' -propoxy- ; 2'MO = 2'-methoxy-; 2'ME = 2 ' -methoxyethoxy- ; PNA = peptide (polyamide) nucleic acid backbone having a side chain corresponding to that of either glycine (N) or D-Lys (K) ; 5'FITC = 5 ' -fluorescein isothiocyanate .
TABLE 9
5-Methyl-Cytosine, Fully 2 ' -Methoxyethoxy- Oligonucleotides Targeted to the 3' -UTR of Human c -fos
O
Figure imgf000052_0001
Emboldened residues indicate 2,6-A residues, i . e . , those having 2 , 6-diaminopurine as a nucleobase.
**ISIS 14663 is a 2'-deoxy- rather than a 2 ' -meLhoxyethoxy-oligonucleotide .
TABLE 10 Effect of Modified c -fos Oligonucleotides on c -fos Expression
Figure imgf000053_0001
Example 7: Oligonucleotides to Mouse AP-1 Subunits Tables 11 and 12 show the sequences of oligonucleotides designed to modulate mouse c-j un and c-fos mRNA expression, respectively. For the nucleotide sequence of the mouse c-jun gene, see Genbank accession No. J04115/MUSCJUN and Ryder et al . , Proc . Na tl . Acad . Sci . U. S . A . , 1988, 85:8464. The nucleotide sequence of the mouse c-fos gene is described in Genbank accession No. J00370/MUSFOS and by Van Beveren et al . , Cell, 1983, 32:1241. Oligonucleotide activities are evaluated as described infra in Example 2 et seg. with the exception that mouse Swiss 3T3 cells (available from, inter alia , the American Type Culture Collection, Rockville, MD, as ATCC No. CCL-92) are used instead of human A549 cells. Due to the high degree of homology between human and murine c-jun and c-fos nucleotide sequences (Van Straaten et al . , Proc . Natl . Acad . Sci . U. S. A. , 1983, 80:3183), probes derived from the human genes were used to detect murine messages . TABLE 11 Phosphorothioate Oligonucleotides Targeted to Mouse c -jun
Figure imgf000054_0001
* Nucleotide co-ordinates from Genbank accession No. J04115/MUSCJUN.
TABLE 12 Phosphorothioate Oligonucleotides Targeted to Mouse c -fos
Ul LO
Figure imgf000055_0001
* Nucleotide co-ordinates from Genbank accession No. J00370/MUSFOS
Example 8: Oligonucleotides to Rat AP-1 Subunits
Tables 13 and 14 show the sequences of oligonucleotides designed to modulate rat c-jun and c-fos mRNA expression, respectively. For the nucleotide sequence of the rat c-jun gene, see Genbank accession No.
X17163/RSJUNAP1 and Saaki et al . , Cancer Res . , 1989, 45:5633. The nucleotide sequence of the rat c-fos gene is described in Genbank accession No. X06769/RNCFOSR and Curran et al . , Oncogene, 1987, 2:79. Oligonucleotide activities were evaluated as described infra in Example 2 et seg. with the exception that rat A10 cells (available from, inter alia , the American Type Culture Collection, Rockville, MD, as ATCC No. CRL-1476) were used instead of human A549 cells. Due to the high degree of homology between human and rat c-jun and c-fos nucleotide sequences, probes derived from the human genes were used to detect the rat messages .
ISIS 12633 (SEQ ID NO:78), a-20-mer phosphorothioate oligonucleotide complementary to a portion of the 3' UTR of rat c-jun mRNA, was selected as an active modulator of c-jun for further studies. Another preferred oligonucleotide targeted to rat AP-1 subunits is ISIS 12635 (SEQ ID NO:80).
Example 9: Modified Oligonucleotides to Rat AP-1 Subunits
Tables 15 and 16 show the sequences and chemical modifications of second generation oligonucleotides designed to modulate mouse c-jun and c-fos mRNA expression. The activities of these modified oligonucleotide are evaluated as described infra in Example 8.
TABLE 13 Phosphorothioate Oligonucleotides Targeted to Rat c -jun
Ui Ji
Figure imgf000057_0001
Figure imgf000058_0001
L
Figure imgf000058_0004
Nucleotide co-ordinates from Genbank accession No. X17163/RSJUNAP1.
Figure imgf000058_0002
Figure imgf000058_0003
TABLE 14 Phosphorothioate Oligonucleotides Targeted to Rat c-fos
Figure imgf000059_0001
Figure imgf000060_0001
Figure imgf000060_0002
Nucleotide co-ordinates from Genbank accession No. X06769/RNCFOSR (see also Curran LΠ oo et al . , Oncogene, 1987, 2:79).
TABLE 15 Additional Oligonucleotides Targeted to Rat c -jun
Figure imgf000061_0001
Figure imgf000061_0002
Figure imgf000062_0001
Figure imgf000062_0002
* Phosphorothioate linkages are indicated by " s ", whereas phosphodiester linkages are signified by " ° "; emboldened residues comprise the additional indicated modifications 2 'MO = 2'-methoxy-; 2 'ME = 2 ' -methoxyethoxy- .
TABLE 16 Additional Oligonucleotides Targeted to Rat c-fos
I
<T
Figure imgf000063_0001
σ.
Figure imgf000064_0001
* Phosphorothioate and phosphodiester linkages are indicated by " s " and " respectively, whereas " d " indicates a dideoxy (chain-terminating) residue; emboldened residues comprise the additional indicated modifications: 2 'MO, 2'-methoxy; 2'ME, 2 ' -methoxyethoxy- .
Example 11: Effect of Oligonucleotides Targeted to AP-1
Subunits on PDGF-Induced Proliferation of Rat Aortic Smooth Muscle Cells
In order to evaluate the effect of AP-1 modulation on cell cycle progression, the following study was performed. Cultured rat aortic smooth muscle (RASM) cells are stimulated to proliferate upon contact with platelet-derived growth factor (PDGF) . Primary RASM cells (passages 6-8) were synchronized by incubation for 48 hours in DMEM containing 0.1% FBS . The cells were treated for 4 hours with 200 nM ISIS 12633 (SEQ ID NO:78), a 20-mer phosphorothioate oligonucleotide complementary to a portion of the 3' UTR of rat c-jun mRNA, or ISIS 12898 (SEQ ID NO:92), a scrambled control of ISIS 12633. Cells were then contacted with PDGF (10 ng/ml) (R&D Systems, Minneapolis, MN) , and cell cycle progression was assessed by FACS analysis 24 hours later. At 2 and 6 hours after exposure to PDGF, c-jun mRNA levels were markedly less in ISIS 12633 -treated cells as compared to untreated cells or cells treated with ISIS 12898. The decrease in c-jun mRNA levels was associated with a significant decrease in the proportion of cells in the G2/M interface at 24 hours. This result provides evidence of the role of AP-1-mediated gene expression in cellular proliferation and indicate that cell cycle progression can be modulated by preventing expression of one or both of the genes which encode a subunit of AP-1.
Example 12: Effect of Oligonucleotides Targeted to AP-1
Subunits on Enzymes Involved in Metastasis
Patients having benign tumors, and primary malignant tumors that have been detected early in the course of their development, may often be successfully treated by the surgical removal of the benign or primary tumor. If unchecked, however, cells from malignant tumors are spread throughout a patient's body through the processes of invasion and metastasis. Invasion refers to the ability of cancer cells to detach from a primary site of attachment and penetrate, e.g., an underlying basement membrane. Metastasis indicates a sequence of events wherein (1) a cancer cell detaches from its extracellular matrices, (2) the detached cancer cell migrates to another portion of the patient's body, often via the circulatory system, and (3) attaches to a distal and inappropriate extracellular matrix, thereby created a focus from which a secondary tumor can arise. Normal cells do not possess the ability to invade or metastasize and/or undergo apoptosis (programmed cell death) if such events occur (Ruoslahti, Sci . Amer . , 1996, 275, 72). The matrix metalloproteinases (MMPs) are a family of enzymes which have the ability to degrade components of the extracellular matrix (Birkedal-Hansen, Current Op . Biol . , 1995, 7, 728) . Many members of the MMP family have been found to have elevated levels of activity in human tumors as well as other disease states (Stetler-Stevenson et al . , Annu . Rev. Cell Biol . , 1993, 9 , 541; Bernhard et al . , Proc . Natl . Acad . Sci . (U. S.A . ) , 1994, 91 , 4293). In particular, one member of this family, matrix metalloproteinase-9 (MMP- 9) , is often found to be expressed only in tumors and other diseased tissues (Himelstein et al . , Invasion & Metastasis , 1994, 14 , 246) . Several studies have shown that regulation of the MMP- 9 gene may be controlled by the AP-1 transcription factor (Kerr et al . , Science, 1988, 242, 1242; Kerr et al . , Cell , 1990, 61 , 267; Gum et al . , J. Biol . Chem. , 1996, 271, 10672; Hua et al . , Cancer Res . , 1996, 56, 5279) . In order to determine whether MMP- 9 expression can be influenced by AP-1 modulation, the following experiments were conducted on normal human epidermal keratinocytes (NHEKs) . Although NHEKs normally express no detectable MMP- 9, MMP- 9 can be induced by a number of stimuli, including TPA. ISIS 10582, an oligonucleotide targeted to c-jun, was evaluated for its ability to modulate MMP- 9 expression according to the protocols described in Examples 2-3 with the following exceptions: (1) NHEK cells were used instead of A549 cells, (2) the probe used, a PCR product prepared using the published sequence of the MMP-9 gene (Huhtala et al . , J. Biol . Chem . , 1991, 266:16485; Sato et al . , Oncogene , 1993, 8:395), is specific for MMP-9 rather than c-jun, and (3) the cells were harvested 24 hours after TPA treatment for 4 hours. The results (Table 17) demonstrate that ISIS 10582 is able to completely inhibit the expression of MMP- after induction with TPA.
TABLE 17 Effect of c-jun Oligonucleotide on MMP-9 Expression
Figure imgf000067_0001
These results demonstrate that c-Jun is required for TPA-mediated induction of MMP- 9, and indicate that oligonucleotides targeted to AP-1 subunits can inhibit the expression of MMP family members, thereby modulating the ability of cancer cells to invade other tissues and/or metastasize to other sites in a patient's body.
Example 13 : Antagonism of Inducers Other than TPA by
Oligonucleotides Targeted to AP-1 Subunits Inducing agents other than TPA function to raise AP-1 levels in vivo . In order to assess the ability of oligonucleotides targeted to AP-1 to antagonize the action of three such inducers, A549 cells were treated and evaluated as in Examples 2 et seg. with the exception that TNF-α, IL-13 or TGF-3 (each at 10 ng/ml and all from R&D Systems,
Minneapolis, MN) were used in place of TPA as inducers. The results (Table 18) demonstrate that ISIS 10582 (SEQ ID NO : 8 , targeted to human c-jun) effectively reduces stimulation of c-Jun by TNF-α or IL-13. in contrast, a scrambled control oligonucleotide, ISIS 11563 (SEQ ID NO:30), did not reverse the induction of c-Jun.
TABLE 18
Effect of Oligonucleotides Targeted to c-Jun on Induction by TNF-α, IL-1/3 or TGF-/S
Figure imgf000068_0001
SEQUENCE LISTING
(1) GENERAL INFORMATION:
(i) APPLICANT: Nicholas M. Dean; Robert A. McKay;
Loren J. Miraglia; Brenda F. Baker (ii) TITLE OF INVENTION: Antisense Oligonucleotide
Compositions and Methods for the Modulation of
Activating Protein 1 (iii) NUMBER OF SEQUENCES: 138 (iv) CORRESPONDENCE ADDRESS:
(A) ADDRESSEE: Law Offices of Jane Massey Licata
(B) STREET: 66 East Main Street
(C) CITY: Marlton
(D) STATE: NJ
(E) COUNTRY: USA
(F) ZIP: 08053
(v) COMPUTER READABLE FORM:
(A) MEDIUM TYPE: DISKETTE, 3.5 INCH, 1.44 MB STORAGE
(B) COMPUTER: IBM PS/2
(C) OPERATING SYSTEM: PC-DOS
(D) SOFTWARE: WORDPERFECT 5.1 (vi) CURRENT APPLICATION DATA:
(A) APPLICATION NUMBER: not assigned
(B) FILING DATE: herewith
(C) CLASSIFICATION: (vii) PRIOR APPLICATION DATA:
(A) APPLICATION NUMBER: 08/837 , 201
(B) FILING DATE: April 14, 1997 (viii) ATTORNEY/AGENT INFORMATION:
(A) NAME: Jane Massey Licata
(B) REGISTRATION NUMBER: 32,257
(C) REFERENCE/DOCKET NUMBER: ISPH-0305 (ix) TELECOMMUNICATION INFORMATION:
(A) TELEPHONE: (609) 779-2400
(B) TELEFAX: (609) 779-8488 (2) INFORMATION FOR SEQ ID NO : 1 :
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI -SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 1:
GCCACACTCA GTGCAACTCT 20
(2) INFORMATION FOR SEQ ID NO : 2: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI -SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO : 2:
CGCACCTCCA CTCCCGCCTC 20
(2) INFORMATION FOR SEQ ID NO: 3: (i) SEQUENCE CHARACTERISTICS: (A) LENGTH: 20 (B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI -SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 3:
ACCAGCCCGG GAGCCACAGG 20
(2) INFORMATION FOR SEQ ID NO: 4: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI -SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 4:
GCTGCGCCGC CGACGTGACG 20
(2) INFORMATION FOR SEQ ID NO : 5: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI -SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO : 5:
CGCCCCGCCG CCGCTGCTCA 20
(2) INFORMATION FOR SEQ ID NO: 6: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid (C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI -SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 6:
GTGTCTCGCC GGGCATCTCG 20
(2) INFORMATION FOR SEQ ID NO : 7: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI -SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO : 7:
CCCCCGACGG TCTCTCTTCA 20
(2) INFORMATION FOR SEQ ID NO: 8: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI -SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 8:
TCAGCCCCCG ACGGTCTCTC 20
(2) INFORMATION FOR SEQ ID NO : 9: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single (D) TOPOLOGY: Linear (iv) ANTI -SENSE: Yes (xi) SEQUENCE DESCRIPTION: SEQ ID NO: 9:
TGCCCCTCAG CCCCCGACGG 20
(2) INFORMATION FOR SEQ ID NO: 10: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI -SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 10:
TGCTCGCTGC AGATGCGGTT 20
(2) INFORMATION FOR SEQ ID NO: 11: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI -SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 11:
CGGTCACTGC TCGTTCGCTG 20
(2) INFORMATION FOR SEQ ID NO: 12: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single (D) TOPOLOGY: Linear (iv) ANTI -SENSE: Yes (xi) SEQUENCE DESCRIPTION: SEQ ID NO: 12:
CATCGTGGCG GTTAGGCAAA 20
(2) INFORMATION FOR SEQ ID NO: 13: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI -SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 13:
GAGAACATCA TCGTGGCGGT 20
(2) INFORMATION FOR SEQ ID NO: 14: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI -SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 14:
ACCGTGGGAA TGAAGTTGGC 20
(2) INFORMATION FOR SEQ ID NO: 15: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear ( iv) ANTI - SENSE : Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 15:
AGCTCCCTCC TCCGGTTGCG 20
(2) INFORMATION FOR SEQ ID NO: 16: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI -SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 16:
TTGCAGGCAG GTCGGTGAGC 20
(2) INFORMATION FOR SEQ ID NO: 17: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI -SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 17:
TGGCACGGAG CGGGCTGTCT 20
(2) INFORMATION FOR SEQ ID NO: 18: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI -SENSE: Yes (xi) SEQUENCE DESCRIPTION: SEQ ID NO: 18:
TGCTGCTGCC CTTGCGGTGG 20
(2) INFORMATION FOR SEQ ID NO : 19: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI -SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO : 19:
CCTCACAGGG CCAGCAGCGT 20
(2) INFORMATION FOR SEQ ID NO : 20: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI -SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 20:
GGTGCCGGCT GCCTCCCCTT 20
(2) INFORMATION FOR SEQ ID NO : 21: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI -SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 21: AAGTCCTTGA GGCCCACAGC 20
(2) INFORMATION FOR SEQ ID NO: 22: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI -SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 22:
CCCCTCCAGC AGCTACCCTT 20
(2) INFORMATION FOR SEQ ID NO: 23: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI -SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 23:
TCCCGTCCCC AGAAGCAGTA 20
(2) INFORMATION FOR SEQ ID NO: 24: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI -SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 24:
CGCGCCCGGC CTGAAAATTT 20 (2) INFORMATION FOR SEQ ID NO : 25: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI-SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 25:
CCTGCCTCGG CCTCCCAAAG 20
(2) INFORMATION FOR SEQ ID NO : 26: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI -SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 26:
CCCCCACTTC CGCCCACTAT 20
(2) INFORMATION FOR SEQ ID NO : 27: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI -SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 27:
TGGTGCCTGC GTGATACTCG 20
(2) INFORMATION FOR SEQ ID NO: 28: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI -SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 28:
CCCTCCCAGG CTCAAGTCAT 20
(2) INFORMATION FOR SEQ ID NO : 29: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI -SENSE: No
(xi) SEQUENCE DESCRIPTION: SEQ ID NO : 29:
GAGAGACCGT CGGGGGCTGA 20
(2) INFORMATION FOR SEQ ID NO: 30: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI -SENSE: No
(xi) SEQUENCE DESCRIPTION: SEQ ID NO : 30:
CACCTCCACG CGCTTCTGGC 20
(2) INFORMATION FOR SEQ ID NO : 31: (i) SEQUENCE CHARACTERISTICS: (A) LENGTH : 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI-SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 31:
TCGGCACCTG AAGGACTTTC 20
( 2 ) INFORMATION FOR SEQ ID NO: 32: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI -SENSE: No
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 32:
GCTGTGGGCC TCAAGGACTT 20
(2) INFORMATION FOR SEQ ID NO: 33: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI -SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 33:
ATGTGCTAGA TGCGCAAAGT 20
(2) INFORMATION FOR SEQ ID NO: 34: (i) SEQUENCE CHARACTERISTICS: (A) LENGTH: 20 (B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI -SENSE: No
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 34:
ACGTCCGATT CCGAGCGCAA 20
(2) INFORMATION FOR SEQ ID NO: 35: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI -SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO : 35:
CAGTGGCCAT CAAACCCGTG 20
(2) INFORMATION FOR SEQ ID NO : 36: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 16
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI -SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO : 36:
CACTCAGTGC AACTCT 16
(2) INFORMATION FOR SEQ ID NO : 37: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 16
(B) TYPE: Nucleic Acid (C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI -SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO : 37:
CCTCCACTCC CGCCTC 16
(2) INFORMATION FOR SEQ ID NO: 38: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI -SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 38:
CTCGCCCAAC TTCAGCCGCC 20
(2) INFORMATION FOR SEQ ID NO : 39: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI -SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 39: CCAGTCCCAG CAACAGCGGC 20
(2) INFORMATION FOR SEQ ID NO: 40: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single (D) TOPOLOGY: Linear (iv) ANTI -SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 40: GCAACAGCGC GCCGGGAAGC 20
(2) INFORMATION FOR SEQ ID NO : 41: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI -SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 41:
CCGGCGACGC CAGCTTGAGC 20
(2) INFORMATION FOR SEQ ID NO: 42: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI -SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 42:
GGCTGTGCCG CGGAGGTGAC 20
(2) INFORMATION FOR SEQ ID NO: 43: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI-SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 43:
CGCCCCACCG CCGCTGCTCA 20
(2) INFORMATION FOR SEQ ID NO : 44: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI -SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 44:
AGCCCGGCCG CGCCATAGGA 20
(2) INFORMATION FOR SEQ ID NO: 45: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI -SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 45:
CTGCACCGGG ATCTGTTGGG 20
(2) INFORMATION FOR SEQ ID NO : 46: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 23
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI -SENSE: Yes (xi) SEQUENCE DESCRIPTION: SEQ ID NO: 46:
GGCGGCGTCT CTCCCGGCAT CTC 23
(2) INFORMATION FOR SEQ ID NO : 47: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI -SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 47:
TGGAGGCGGC AATGCGGTTC 20
(2) INFORMATION FOR SEQ ID NO : 48: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI -SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 48:
CCCTGAGCAT GTTGGCCGTG 20
(2) INFORMATION FOR SEQ ID NO : 49: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI -SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO : 49: CAAAGCCAGG CGCGCCACGT 20
(2) INFORMATION FOR SEQ ID NO : 50: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI -SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 50:
TTGAGAGAGG CAGGCCAGGG 20
(2) INFORMATION FOR SEQ ID NO: 51: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI -SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 51:
TGGACTTGTG TGTTGCCGGG 20
(2) INFORMATION FOR SEQ ID NO: 52: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI -SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 52:
TCCATGGGTC CCTGCTTTGA 20 (2) INFORMATION FOR SEQ ID NO: 53: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI -SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 53:
TGGTCGCGCG CGGGCACAGC 20
(2) INFORMATION FOR SEQ ID NO: 54: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI -SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 54:
AGCTCCCTCC TCCGATTCCG 20
(2) INFORMATION FOR SEQ ID NO : 55: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI -SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 55:
GCTCTGTGAC CATGGGCCCC 20
(2) INFORMATION FOR SEQ ID NO: 56: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI -SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 56:
GAACCGCCGG CTCTATCCAG 20
(2) INFORMATION FOR SEQ ID NO: 57: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI -SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 57:
GCCCCTGCGA GTCACACCCC 20
(2) INFORMATION FOR SEQ ID NO : 58: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI-SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 58:
TAAGGCTGCT CTGACCGCGC 20
(2) INFORMATION FOR SEQ ID NO: 59: (i) SEQUENCE CHARACTERISTICS: (A) LENGTH : 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI -SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO : 59:
CGCCCGCAGC ACCCTCCTCC 20
(2) INFORMATION FOR SEQ ID NO: 60: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI -SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 60:
CAGGCGCTGC TCCGGAGTCT 20
(2) INFORMATION FOR SEQ ID NO : 61: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI -SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO : 61:
TCCCTTGAAT TCCGCAGCGC 20
(2) INFORMATION FOR SEQ ID NO: 62: (i) SEQUENCE CHARACTERISTICS: (A) LENGTH: 20 (B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI -SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 62:
AGCGGAGGTG AGCGAGGAGG 20
(2) INFORMATION FOR SEQ ID NO: 63: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI -SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 63:
CCCCAGCCCA CAAAGGTCCA 20
(2) INFORMATION FOR SEQ ID NO: 64: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI -SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 64:
TGCTCAAGGA CCCTGCGCCC 20
(2) INFORMATION FOR SEQ ID NO: 65: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid (C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI -SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 65:
GGGAAGCCAA GGTCATCGGG 20
(2) INFORMATION FOR SEQ ID NO : 66: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI-SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 66:
TGCTGCTGCC CTTTCGGTGG 20
(2) INFORMATION FOR SEQ ID NO: 67: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI -SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 67:
CTGGATGCCG GCTGCCTTGC 20
(2) INFORMATION FOR SEQ ID NO : 68: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single (D) TOPOLOGY: Linear (iv) ANTI-SENSE: Yes (xi) SEQUENCE DESCRIPTION: SEQ ID NO: 68:
CAGCTCGGGA AGTGGCACGT 20
(2) INFORMATION FOR SEQ ID NO : 69: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI -SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 69:
GGAACACGCT ATTGCCAGGA 20
(2) INFORMATION FOR SEQ ID NO: 70: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI -SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO : 70:
CGGCGGCGCA GACCAGTCGT 20
(2) INFORMATION FOR SEQ ID NO: 71: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear ( iv) ANTI - SENSE : Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 71:
GGCATCGTCG TAGAAGGTCG 20
(2) INFORMATION FOR SEQ ID NO: 72: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI -SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 72:
GGAGGTGCGG CTTCAGATTG 20
(2) INFORMATION FOR SEQ ID NO: 73: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI -SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 73:
CCCTCCTGCT CGTCGGTCAC 20
(2) INFORMATION FOR SEQ ID NO: 74: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI -SENSE: Yes (xi) SEQUENCE DESCRIPTION: SEQ ID NO: 74:
ACTGACTGGT TGTGCCGCGG 20
(2) INFORMATION FOR SEQ ID NO : 75: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI -SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 75:
CGCTGTAGCC GCCGCCGCCG 20
(2) INFORMATION FOR SEQ ID NO: 76: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI -SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 76:
CCTTGATCCG CTCCTGAGAC 20
(2) INFORMATION FOR SEQ ID NO : 77: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI -SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 77: GCCAGCTCGG AGTTTTGCGC 20
(2) INFORMATION FOR SEQ ID NO: 78: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI -SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 78:
TTTTCTTCCA CTGCCCCTCA 20
(2) INFORMATION FOR SEQ ID NO: 79: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI -SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO : 79:
CCCTTGGCTT CAGTACTCGG 20
(2) INFORMATION FOR SEQ ID NO: 80: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI -SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO : 80:
CTTCCCACTC CAGCACATTG 20 (2) INFORMATION FOR SEQ ID NO: 81: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI -SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 81:
GCACAGCCCG TTCGCAAAGC 20
(2) INFORMATION FOR SEQ ID NO : 82: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI -SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 82:
AATGCAGCAG AGAGGTTGGG 20
(2) INFORMATION FOR SEQ ID NO: 83: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI -SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 83:
GACGGGAGGG ACTACAGGCT 20
(2) INFORMATION FOR SEQ ID NO: 84: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI -SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 84:
TCTGGACTTG TGGGTTGCTG 20
(2) INFORMATION FOR SEQ ID NO: 85: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI -SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 85:
TAAACGATCA CAGCGCATGC 20
(2) INFORMATION FOR SEQ ID NO : 86: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI -SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO : 86:
GGGAGGACGA GCAGCCAGTG 20
(2) INFORMATION FOR SEQ ID NO: 87: (i) SEQUENCE CHARACTERISTICS: (A) LENGTH : 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI -SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO : 87:
CCCGGCCTTT TGACCGCCTC 20
(2) INFORMATION FOR SEQ ID NO : 88: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI -SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO : 88:
CCGCCTCCCC GGCCTTTTGA 20
(2) INFORMATION FOR SEQ ID NO : 89: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI -SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO : 89:
CCGTCGTGGT CCTCCGTGAC 20
(2) INFORMATION FOR SEQ ID NO : 90: (i) SEQUENCE CHARACTERISTICS: (A) LENGTH: 20 (B) TYPE: Nucleic Acid
(C)- STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI -SENSE: Yes (xi) SEQUENCE DESCRIPTION: SEQ ID NO: 90:
GTGACCGACG AGCAGGAGGG 20
(2) INFORMATION FOR SEQ ID NO: 91: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI -SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 91:
AAAAGAAGGT GACGGGGAGT 20
(2) INFORMATION FOR SEQ ID NO: 92: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI -SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 92:
TTCTCTTTTA GCCTCCCCCA 20
(2) INFORMATION FOR SEQ ID NO: 93: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid (C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI -SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 93:
TCCCCCATTC TCTTTTAGCC 20
(2) INFORMATION FOR SEQ ID NO: 94: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI -SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 94:
TTATCATCGA CAGCGCCACA 20
(2) INFORMATION FOR SEQ ID NO: 95: - (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI -SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 95:
TGAGGGGCAG TGGAAGAAAA 20
(2) INFORMATION FOR SEQ ID NO: 96: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single (D) TOPOLOGY: Linear (iv) ANTI -SENSE: Yes (xi) SEQUENCE DESCRIPTION: SEQ ID NO: 96:
GTTCTCGGCT CCGCCGGCTC 20
(2) INFORMATION FOR SEQ ID NO: 97: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI -SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 97:
CATCATGGTC GTGGTTTGGG 20
(2) INFORMATION FOR SEQ ID NO : 98: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI -SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 98:
TCCGCGTTGA AACCCGAGAA 20
(2) INFORMATION FOR SEQ ID NO: 99: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear ( iv ) ANTI - SENSE : Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO : 99:
TGGGCTGGTG GAGATGGCTG 20
(2) INFORMATION FOR SEQ ID NO : 100: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI -SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 100:
CGATGCTCTG CGCTCTGCCG 20
(2) INFORMATION FOR SEQ ID NO : 101: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI -SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 101:
TTCGGTGGGC AGCTGCGCAG 20
(2) INFORMATION FOR SEQ ID NO: 102: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI -SENSE: Yes (xi) SEQUENCE DESCRIPTION: SEQ ID NO: 102: CAGGGCTAGC AGTGTGGGCG 20
(2) INFORMATION FOR SEQ ID NO: 103: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI -SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 103:
CCAGCTCAGT CAGTGCCGGC 20
(2) INFORMATION FOR SEQ ID NO: 104: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI -SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 104:
TCTACGGGAA CCCCTCGAGG 20
(2) INFORMATION FOR SEQ ID NO : 105: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI -SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO : 105: CTCCATGCGG TTGCTTTTGA 20
(2) INFORMATION FOR SEQ ID NO: 106: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI -SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 106:
CAGGCCTGGC TCACATGCTA 20
(2) INFORMATION FOR SEQ ID NO : 107: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI -SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 107:
AGATGCCCTT GGGGAGCTCC 20
(2) INFORMATION FOR SEQ ID NO : 108: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI -SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 108:
TGACTATAGA CCGCCGCCGG 20 (2) INFORMATION FOR SEQ ID NO: 109: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI -SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 109:
CCGCCGGTGA CTATAGACCG 20
(2) INFORMATION FOR SEQ ID NO: 110: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI -SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 110:
TCAACCGGTA CGCCACGTGG 20
(2) INFORMATION FOR SEQ ID NO : 111: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI -SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 111: CCTCGAGGGG TTCCCGTAGA 20 (2) INFORMATION FOR SEQ ID NO: 112: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI -SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 112:
GTCCGGACCG AGTGTACGAT 20
(2) INFORMATION FOR SEQ ID NO: 113: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI -SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 113:
GCGCACCGTC ATTACGTCGA 20
(2) INFORMATION FOR SEQ ID NO: 114: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI -SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 114:
ACGTCGAGCG CACCGTCATT 20 (2) INFORMATION FOR SEQ ID NO: 115: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI -SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 115:
CACGCGTGCC TGACTTGGTA 20
(2) INFORMATION FOR SEQ ID NO: 116: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI -SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 116:
TAGCATGTGA GCCAGGCCTG 20
(2) INFORMATION FOR SEQ ID NO: 117: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI -SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 117:
TTCTCACCCA CCACGTACGT 20 (2) INFORMATION FOR SEQ ID NO: 118: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 16
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI-SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 118:
TGCGGGTGAG TGGTAG 16
(2) INFORMATION FOR SEQ ID NO : 119: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI -SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO : 119:
GCCGCGGGAC CAGCCCCAGC 20
(2) INFORMATION FOR SEQ ID NO: 120: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 16
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI -SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO : 120:
TGCGGGTGAG TGGTAG 16 (2) INFORMATION FOR SEQ ID NO : 121: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 16
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI -SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO : 121:
CGCTGCAGAT GCGGTT 16
(2) INFORMATION FOR SEQ ID NO: 122: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 16
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI -SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO : 122:
CCGCCGGCTC AGTCTT 16
(2) INFORMATION FOR SEQ ID NO: 123: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 16
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI -SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 123:
CATCGTGGCG GTTAGG 16 (2) INFORMATION FOR SEQ ID NO: 124: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 30
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI -SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 124:
CCATCTTAAT AAATAAATTA AAAACACAAT 30
(2) INFORMATION FOR SEQ ID NO : 125: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI -SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 125:
AAATAAATTA AAAACACAAT 20
(2) INFORMATION FOR SEQ ID NO: 126: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI -SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 126:
AATTAAAAAC ACAATAAAAC 20 (2) INFORMATION FOR SEQ ID NO : 127: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI -SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 127:
ATATAAATAT CTGAGAATCC 20
(2) INFORMATION FOR SEQ ID NO: 128: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI-SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 128:
ATCTGAGAAT CCATCTTAAT 20
(2) INFORMATION FOR SEQ ID NO : 129: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI -SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO : 129:
AAATATAAAT ATCTGAGAAT 20 (2) INFORMATION FOR SEQ ID NO : 130: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI -SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 130:
AAGACCTCAA GGTAGAAAAA 20
(2) INFORMATION FOR SEQ ID NO : 131: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 30
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI-SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO : 131: CCATCTTAAT AAATAAATTA AAAACACAAT 30
(2) INFORMATION FOR SEQ ID NO: 132: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI -SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 132: AAATAAATTA AAAACACAAT 20
(2) INFORMATION FOR SEQ ID NO: 133: - Ill -
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI -SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 133: AATTAAAAAC ACAATAAAAC 20
(2) INFORMATION FOR SEQ ID NO: 134: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI -SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 134: ATATAAATAT CTGAGAATCC 20
(2) INFORMATION FOR SEQ ID NO: 135: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI -SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO : 135: ATATAAATAT CTGAGAATCC 20
(2) INFORMATION FOR SEQ ID NO: 136: (i) SEQUENCE CHARACTERISTICS: (A) LENGTH : 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI -SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO : 136: ATCTGAGAAT CCATCTTAAT 20
(2) INFORMATION FOR SEQ ID NO : 137: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI -SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO : 137: AAATATAAAT ATCTGAGAAT 20
(2) INFORMATION FOR SEQ ID NO : 138: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI -SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 138: AAGACCTCAA GGTAGAAAAA 20

Claims

CLAIMS What is claimed is:
1. An oligonucleotide comprising 8 to 30 nucleotides connected by covalent linkages, wherein said antisense compound comprises a nucleobase sequence specifically hybridizable with a region of a nucleic acid encoding a c-Fos protein of a mammal and wherein said antisense compound modulates the expression of said c-Fos protein.
2. The oligonucleotide of claim 1 wherein said nucleic acid is an mRNA molecule encoding said c-Fos protein.
3. The oligonucleotide of claim 2 wherein said region is selected from the group consisting of the 5' -cap, the 5'- untranslated region, the start codon region, the open reading frame, the stop codon region and the 3 ' -untranslated region.
4. The oligonucleotide of claim 1 wherein said mammal is a human .
5. The oligonucleotide of claim 4, wherein said sequence is selected from the group consisting of SEQ ID NOS: 10, 11, 12, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 27, 120, 121, 122, 123, 124, 131, 132, 133, 134, 135, 136, 137 and 138.
6. The oligonucleotide of any of claims 1 to 5 wherein said antisense compound comprises one or more chemical modifications selected from the group consisting of one or more modified linkages, one or more modified nucleobases and one or more sugar modifications.
7. A pharmaceutical composition comprising the oligonucleotide of any one of claims 1 to 6 and a pharmaceutically acceptable carrier.
8. An oligonucleotide comprising 8 to 30 nucleotides connected by covalent linkages, wherein said antisense compound comprises a nucleobase sequence specifically hybridizable with a region of a nucleic acid encoding a c-Jun protein of a mammal and wherein said antisense compound modulates the expression of said c-Jun protein.
9. The oligonucleotide of claim 8 wherein said nucleic acid is an mRNA molecule encoding said c-Jun protein.
10. The oligonucleotide of claim 9 wherein said region is selected from the group consisting of the 5' -cap, the 5'- untranslated region, the start codon region, the open reading frame, the stop codon region and the 3 ' -untranslated region.
11. The oligonucleotide of claim 8 wherein said mammal is a human .
12. The oligonucleotide of claim 11, wherein said sequence is selected from the group consisting of SEQ ID NOS: 1, 3, 4, 5, 6, 7, 8, 9, 36, 37 and 118.
13. The oligonucleotide of any of claims 8 to 12 wherein said antisense compound comprises one or more chemical modifications selected from the group consisting of one or more modified linkages, one or more modified nucleobases and one or more sugar modifications.
14. A pharmaceutical composition comprising the oligonucleotide of any one of claims 8 to 13 and a pharmaceutically acceptable carrier.
15. The oligonucleotide of claim 1 or claim 8 wherein said mammal is a rodent .
16. A pharmaceutical composition comprising one or more oligonucleotides of any one of claims 1 to 6 , one or more oligonucleotides of any one of claims 8 to 13, and a pharmaceutically acceptable carrier.
17. The pharmaceutical composition of any one of claims 7, 14 or 16 further comprising a chemotherapeutic agent.
18. A method of modulating the expression of a c-Fos protein in cells or tissues comprising contacting said cells or tissues with an oligonucleotide of any one of claims 1 to 6.
19. A method of modulating the expression of a c-Jun protein in cells or tissues comprising contacting said cells or tissues with an oligonucleotide of any one of claims 8 to 13.
20. A method of inhibiting metastasis in an animal comprising administering to said animal a therapeutically effective amount of an oligonucleotide of any one of claims 1 to 6 and 8 to 13.
21. A method of inhibiting metastasis in an animal comprising administering to said animal a therapeutically effective amount of the pharmaceutical composition of any one of claims 7, 14 and 16.
22. A method of modulating cell cycle progression in cultured cells or the cells of an animal comprising administering to said cells an effective amount of the oligonucleotide of any one of claims 1 to 6 and 8 to 13.
23. A method of modulating cell cycle progression in the cells of an animal comprising administering to said animal an effective amount of the pharmaceutical composition of any one of claims 7, 14 and 16.
24. A method of treating an animal having, suspected of having or prone to having a hyperproliferative disease comprising administering to said animal a therapeutically effective amount of the pharmaceutical composition of any one of claims 7, 14 and 16.
PCT/US1998/007386 1997-04-14 1998-04-14 Antisense oligonucleotide compositions and methods for the modulation of activating protein 1 WO1998046272A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
AU69688/98A AU6968898A (en) 1997-04-14 1998-04-14 Antisense oligonucleotide compositions and methods for the modulation of activating protein
EP98915526A EP0975371A4 (en) 1997-04-14 1998-04-14 Antisense oligonucleotide compositions and methods for the modulation of activating protein 1

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US08/837,201 US5985558A (en) 1997-04-14 1997-04-14 Antisense oligonucleotide compositions and methods for the inibition of c-Jun and c-Fos
US08/837,201 1997-04-14

Publications (1)

Publication Number Publication Date
WO1998046272A1 true WO1998046272A1 (en) 1998-10-22

Family

ID=25273801

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1998/007386 WO1998046272A1 (en) 1997-04-14 1998-04-14 Antisense oligonucleotide compositions and methods for the modulation of activating protein 1

Country Status (4)

Country Link
US (4) US5985558A (en)
EP (1) EP0975371A4 (en)
AU (1) AU6968898A (en)
WO (1) WO1998046272A1 (en)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1140971A1 (en) * 1999-01-07 2001-10-10 Isis Pharmaceuticals, Inc. Antisense modulation of novel anti-apoptotic bcl-2-related proteins
WO2003072114A1 (en) 2002-02-27 2003-09-04 Unisearch Limited Vascular therapeutics
JP2004507263A (en) * 2000-09-02 2004-03-11 グリュネンタール・ゲゼルシャフト・ミト・ベシュレンクテル・ハフツング Antisense oligonucleotides to VR1

Families Citing this family (60)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7119184B2 (en) 1991-08-12 2006-10-10 Isis Pharmaceuticals, Inc. Oligonucleotides having A-DNA form and B-DNA form conformational geometry
US5985558A (en) * 1997-04-14 1999-11-16 Isis Pharmaceuticals Inc. Antisense oligonucleotide compositions and methods for the inibition of c-Jun and c-Fos
US20040005707A1 (en) * 2002-07-02 2004-01-08 Isis Pharmaceuticals Inc. Antisense modulation of integrin beta 5 expression
US6410518B1 (en) * 1994-05-31 2002-06-25 Isis Pharmaceuticals, Inc. Antisense oligonucleotide inhibition of raf gene expression
WO1999060167A1 (en) * 1998-05-21 1999-11-25 Isis Pharmaceuticals, Inc. Compositions and methods for topical delivery of oligonucleotides
US20030232438A1 (en) * 2002-06-17 2003-12-18 Isis Pharmaceuticals Inc. Antisense modulation of KOX 1 expression
US6277967B1 (en) 1998-07-14 2001-08-21 Isis Pharmaceuticals, Inc. Carbohydrate or 2′-modified oligonucleotides having alternating internucleoside linkages
US6077709A (en) 1998-09-29 2000-06-20 Isis Pharmaceuticals Inc. Antisense modulation of Survivin expression
AU772847B2 (en) * 1998-11-12 2004-05-06 Invitrogen Corporation Transfection reagents
CN1273478C (en) 1999-02-12 2006-09-06 三共株式会社 Novel nucleosides and oligonucleotide analogues
US20030166592A1 (en) * 1999-07-19 2003-09-04 Monia Brett P. Antisense modulation of liver glycogen phosphorylase expression
US20030125276A1 (en) * 2001-11-08 2003-07-03 Isis Pharmaceuticals Inc. Antisense modulation of thyroid hormone receptor interactor 6 expression
JP4151751B2 (en) * 1999-07-22 2008-09-17 第一三共株式会社 New bicyclonucleoside analogues
US7394850B1 (en) 1999-10-25 2008-07-01 Sedna Patent Services, Llc Method and apparatus for performing digital-to-digital video insertion
US6368856B1 (en) * 2000-09-14 2002-04-09 Isis Pharmaceuticals, Inc. Antisense inhibition of Phosphorylase kinase beta expression
US9994853B2 (en) 2001-05-18 2018-06-12 Sirna Therapeutics, Inc. Chemically modified multifunctional short interfering nucleic acid molecules that mediate RNA interference
US20060241075A1 (en) * 2001-05-18 2006-10-26 Sirna Therapeutics, Inc. RNA interference mediated inhibition of desmoglein gene expression using short interfering nucleic acid (siNA)
US20050079610A1 (en) * 2001-05-18 2005-04-14 Sirna Therapeutics, Inc. RNA interference mediated inhibition of Fos gene expression using short interfering nucleic acid (siNA)
US20050282188A1 (en) * 2001-05-18 2005-12-22 Sirna Therapeutics, Inc. RNA interference mediated inhibition of gene expression using short interfering nucleic acid (siNA)
US20070270579A1 (en) * 2001-05-18 2007-11-22 Sirna Therapeutics, Inc. RNA interference mediated inhibition of gene expression using short interfering nucleic acid (siNA)
US20070042983A1 (en) * 2001-05-18 2007-02-22 Sirna Therapeutics, Inc. RNA interference mediated inhibition of gene expression using short interfering nucleic acid (siNA)
US20080161256A1 (en) * 2001-05-18 2008-07-03 Sirna Therapeutics, Inc. RNA interference mediated inhibition of gene expression using short interfering nucleic acid (siNA)
US20030096772A1 (en) * 2001-07-30 2003-05-22 Crooke Rosanne M. Antisense modulation of acyl CoA cholesterol acyltransferase-2 expression
US20030096773A1 (en) * 2001-08-01 2003-05-22 Crooke Rosanne M. Antisense modulation of acyl coenzyme a cholesterol acyltransferase-1 expression
US7488313B2 (en) 2001-11-29 2009-02-10 Boston Scientific Scimed, Inc. Mechanical apparatus and method for dilating and delivering a therapeutic agent to a site of treatment
PT1470144E (en) * 2002-02-01 2009-02-10 Univ Mcgill Oligonucleotides comprising alternating segments and uses thereof
US7910724B2 (en) * 2002-02-20 2011-03-22 Sirna Therapeutics, Inc. RNA interference mediated inhibition of Fos gene expression using short interfering nucleic acid (siNA)
AU2003207708A1 (en) * 2002-02-20 2003-09-09 Sirna Therapeutics, Inc. Rna interference mediated inhibition of map kinase genes
US9657294B2 (en) 2002-02-20 2017-05-23 Sirna Therapeutics, Inc. RNA interference mediated inhibition of gene expression using chemically modified short interfering nucleic acid (siNA)
US9181551B2 (en) 2002-02-20 2015-11-10 Sirna Therapeutics, Inc. RNA interference mediated inhibition of gene expression using chemically modified short interfering nucleic acid (siNA)
US20030191075A1 (en) * 2002-02-22 2003-10-09 Cook Phillip Dan Method of using modified oligonucleotides for hepatic delivery
US20030232443A1 (en) * 2002-06-18 2003-12-18 Isis Pharmaceuticals Inc. Antisense modulation of centromere protein B expression
US20040014050A1 (en) * 2002-07-19 2004-01-22 Isis Pharmaceuticals Inc. Antisense modulation of EDG8 expression
US20030199463A1 (en) * 2002-04-23 2003-10-23 Silviu Itescu DNA enzyme to inhibit plasminogen activator inhibitor-1
US6828149B2 (en) * 2002-06-20 2004-12-07 Isis Pharmaceuticals, Inc. Antisense modulation of PPP3R1 expression
US7344833B2 (en) 2002-11-07 2008-03-18 Irm Llc Methods and compositions for modulating activator protein 1
US7399853B2 (en) * 2003-04-28 2008-07-15 Isis Pharmaceuticals Modulation of glucagon receptor expression
US20050053981A1 (en) 2003-09-09 2005-03-10 Swayze Eric E. Gapped oligomeric compounds having linked bicyclic sugar moieties at the termini
WO2005071082A1 (en) * 2004-01-22 2005-08-04 Good Will Okinawa Co.,Ltd Antisense oligonucleotide having anticancerous activity
US20050261233A1 (en) * 2004-04-21 2005-11-24 Sanjay Bhanot Modulation of glucose-6-phosphatase translocase expression
US10508277B2 (en) 2004-05-24 2019-12-17 Sirna Therapeutics, Inc. Chemically modified multifunctional short interfering nucleic acid molecules that mediate RNA interference
IL162276A0 (en) * 2004-06-01 2005-11-20 Hadasit Med Res Service Nucleic acid molecules as heparanase potent inhibitors, compositions and methods of use thereof
US7807816B2 (en) 2004-06-28 2010-10-05 University Of Western Australia Antisense oligonucleotides for inducing exon skipping and methods of use thereof
EP2397563A3 (en) * 2004-09-17 2012-07-18 Isis Pharmaceuticals, Inc. Enhanced antisense oligonucleotides
US20090176725A1 (en) * 2005-08-17 2009-07-09 Sirna Therapeutics Inc. Chemically modified short interfering nucleic acid molecules that mediate rna interference
CA2623772A1 (en) * 2005-09-19 2007-03-29 Johnson & Johnson Pharmaceutical Research & Development, L.L.C. Modulation of glucocorticoid receptor expression
AU2007356036A1 (en) * 2007-06-29 2009-01-08 Newsouth Innovations Pty Limited Treatment of rheumatoid arthritis
KR20230025924A (en) 2008-10-24 2023-02-23 사렙타 쎄러퓨틱스 인코퍼레이티드 Multiple exon skipping compositions for dmd
EP2499249B1 (en) 2009-11-12 2018-08-08 The University Of Western Australia Antisense molecules and methods for treating pathologies
US9260471B2 (en) 2010-10-29 2016-02-16 Sirna Therapeutics, Inc. RNA interference mediated inhibition of gene expression using short interfering nucleic acids (siNA)
EP2758533B1 (en) 2011-09-20 2018-04-11 Ionis Pharmaceuticals, Inc. Antisense modulation of gcgr expression
IN2014CN03749A (en) 2011-10-25 2015-09-25 Isis Pharmaceuticals Inc
JP6449231B2 (en) 2013-03-14 2019-01-09 サレプタ セラピューティクス, インコーポレイテッド Exon skipping composition for treating muscular dystrophy
EA201591792A1 (en) 2013-03-15 2016-02-29 Сарепта Терапьютикс, Инк. IMPROVED COMPOSITIONS FOR THE TREATMENT OF MUSCULAR DISTROPHIA
EP3169310A1 (en) 2014-07-15 2017-05-24 Life Technologies Corporation Compositions with lipid aggregates and methods for efficient delivery of molecules to cells
JP2019503394A (en) 2016-01-31 2019-02-07 ユニバーシティ・オブ・マサチューセッツUniversity Of Massachusetts Branched oligonucleotide
WO2018031933A2 (en) 2016-08-12 2018-02-15 University Of Massachusetts Conjugated oligonucleotides
US20190352638A1 (en) * 2016-11-17 2019-11-21 Children's Medical Center Corporation Therapeutic modulation of c-fos in the eye
EP3840759A4 (en) * 2018-08-23 2022-06-01 University Of Massachusetts O-methyl rich fully stabilized oligonucleotides
AU2022299169A1 (en) 2021-06-23 2024-02-08 Beth Israel Deaconess Medical Center, Inc. Optimized anti-flt1 oligonucleotide compounds for treatment of preeclampsia and other angiogenic disorders

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1995002051A2 (en) * 1993-07-10 1995-01-19 Biognostik Gesellschaft für Biomolekulare Diagnostik mbH A pharmaceutical composition comprising antisense-nucleic acid for prevention and/or treatment of neuronal injury, degeneration and cell death and for the treatment of neoplasms
US5585479A (en) * 1992-07-24 1996-12-17 The United States Of America As Represented By The Secretary Of The Navy Antisense oligonucleotides directed against human ELAM-I RNA

Family Cites Families (36)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
FR2540122B1 (en) * 1983-01-27 1985-11-29 Centre Nat Rech Scient NOVEL COMPOUNDS COMPRISING A SEQUENCE OF OLIGONUCLEOTIDE LINKED TO AN INTERCALATION AGENT, THEIR SYNTHESIS PROCESS AND THEIR APPLICATION
NZ209840A (en) * 1983-10-17 1988-11-29 Kaji Akira A method of inhibiting viral propagation by hybridising dna with the viral rna thus blocking its action
US4806463A (en) * 1986-05-23 1989-02-21 Worcester Foundation For Experimental Biology Inhibition of HTLV-III by exogenous oligonucleotides
US5194428A (en) * 1986-05-23 1993-03-16 Worcester Foundation For Experimental Biology Inhibition of influenza virus replication by oligonucleotide phosphorothioates
US5276019A (en) * 1987-03-25 1994-01-04 The United States Of America As Represented By The Department Of Health And Human Services Inhibitors for replication of retroviruses and for the expression of oncogene products
US5264423A (en) * 1987-03-25 1993-11-23 The United States Of America As Represented By The Department Of Health And Human Services Inhibitors for replication of retroviruses and for the expression of oncogene products
US5749847A (en) * 1988-01-21 1998-05-12 Massachusetts Institute Of Technology Delivery of nucleotides into organisms by electroporation
US5004810A (en) * 1988-09-30 1991-04-02 Schering Corporation Antiviral oligomers
US5098890A (en) * 1988-11-07 1992-03-24 Temple University-Of The Commonwealth System Of Higher Education Antisence oligonucleotides to c-myb proto-oncogene and uses thereof
US5087617A (en) * 1989-02-15 1992-02-11 Board Of Regents, The University Of Texas System Methods and compositions for treatment of cancer using oligonucleotides
US5587361A (en) * 1991-10-15 1996-12-24 Isis Pharmaceuticals, Inc. Oligonucleotides having phosphorothioate linkages of high chiral purity
US5457191A (en) * 1990-01-11 1995-10-10 Isis Pharmaceuticals, Inc. 3-deazapurines
US5578718A (en) * 1990-01-11 1996-11-26 Isis Pharmaceuticals, Inc. Thiol-derivatized nucleosides
US5212295A (en) * 1990-01-11 1993-05-18 Isis Pharmaceuticals Monomers for preparation of oligonucleotides having chiral phosphorus linkages
US5459255A (en) * 1990-01-11 1995-10-17 Isis Pharmaceuticals, Inc. N-2 substituted purines
US5166195A (en) * 1990-05-11 1992-11-24 Isis Pharmaceuticals, Inc. Antisense inhibitors of the human immunodeficiency virus phosphorothioate oligonucleotides
US5135917A (en) * 1990-07-12 1992-08-04 Nova Pharmaceutical Corporation Interleukin receptor expression inhibiting antisense oligonucleotides
US5378825A (en) * 1990-07-27 1995-01-03 Isis Pharmaceuticals, Inc. Backbone modified oligonucleotide analogs
US5218105A (en) * 1990-07-27 1993-06-08 Isis Pharmaceuticals Polyamine conjugated oligonucleotides
US5386023A (en) * 1990-07-27 1995-01-31 Isis Pharmaceuticals Backbone modified oligonucleotide analogs and preparation thereof through reductive coupling
US5541307A (en) * 1990-07-27 1996-07-30 Isis Pharmaceuticals, Inc. Backbone modified oligonucleotide analogs and solid phase synthesis thereof
US5138045A (en) * 1990-07-27 1992-08-11 Isis Pharmaceuticals Polyamine conjugated oligonucleotides
US5242906A (en) * 1991-04-22 1993-09-07 University Of North Carolina At Chapel Hill Antisense oligonucleotides against Epstein-Barr virus
US5539082A (en) * 1993-04-26 1996-07-23 Nielsen; Peter E. Peptide nucleic acids
EP0558697A1 (en) * 1991-06-28 1993-09-08 Massachusetts Institute Of Technology Localized oligonucleotide therapy
FR2692265B1 (en) * 1992-05-25 1996-11-08 Centre Nat Rech Scient BIOLOGICALLY ACTIVE COMPOUNDS OF THE PHOSPHOTRIESTER TYPE.
WO1994002637A1 (en) * 1992-07-20 1994-02-03 Isis Pharmaceuticals, Inc. Pseudo-half-knot rna formation by hybridization of antisense oligonucleotides to target rna's secondary structure
US5985558A (en) * 1997-04-14 1999-11-16 Isis Pharmaceuticals Inc. Antisense oligonucleotide compositions and methods for the inibition of c-Jun and c-Fos
US6514745B1 (en) * 1993-07-19 2003-02-04 The Regents Of The University Of California Oncoprotein protein kinase
US5571902A (en) * 1993-07-29 1996-11-05 Isis Pharmaceuticals, Inc. Synthesis of oligonucleotides
US5602156A (en) * 1993-09-17 1997-02-11 The United States Of America As Represented By The Department Of Health And Human Services Method for inhibiting metalloproteinase expression
WO1998003542A1 (en) * 1996-07-24 1998-01-29 Buchardt, Dorte Peptide nucleic acids having enhanced binding affinity, sequence specificity and solubility
US5554746A (en) * 1994-05-16 1996-09-10 Isis Pharmaceuticals, Inc. Lactam nucleic acids
US5801155A (en) * 1995-04-03 1998-09-01 Epoch Pharmaceuticals, Inc. Covalently linked oligonucleotide minor grove binder conjugates
FR2733500B1 (en) * 1995-04-28 1997-07-25 Centre Nat Rech Scient NOVEL ANTISENS DIRECTED AGAINST RAS, PREPARATION AND USES
EP0856579A1 (en) * 1997-01-31 1998-08-05 BIOGNOSTIK GESELLSCHAFT FÜR BIOMOLEKULARE DIAGNOSTIK mbH An antisense oligonucleotide preparation method

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5585479A (en) * 1992-07-24 1996-12-17 The United States Of America As Represented By The Secretary Of The Navy Antisense oligonucleotides directed against human ELAM-I RNA
WO1995002051A2 (en) * 1993-07-10 1995-01-19 Biognostik Gesellschaft für Biomolekulare Diagnostik mbH A pharmaceutical composition comprising antisense-nucleic acid for prevention and/or treatment of neuronal injury, degeneration and cell death and for the treatment of neoplasms

Non-Patent Citations (6)

* Cited by examiner, † Cited by third party
Title
BRANCH A. D.: "A GOOD ANTISENSE MOLECULE IS HARD TO FIND.", TRENDS IN BIOCHEMICAL SCIENCES., ELSEVIER, HAYWARDS., GB, vol. 23., 1 February 1998 (1998-02-01), GB, pages 45 - 50., XP002910770, ISSN: 0968-0004, DOI: 10.1016/S0968-0004(97)01155-9 *
CHEN S.-L., TSAO L.-T., TSAO Y.-P.: "ANTISENSE OLIGODEOXYNUCLEOTIDES TO C-JUN INHIBITS PROLIFERATION OF TRANSFORMED NIH 3T3 CELLS INDUCED BY E5A OF HPV-11.", CANCER LETTERS, NEW YORK, NY, US, vol. 85., 1 January 1994 (1994-01-01), US, pages 119 - 123., XP002910766, ISSN: 0304-3835, DOI: 10.1016/0304-3835(94)90247-X *
GEWIRTZ A. M., STEIN C. A., GLAZER P. M.: "FACILITATING OLIGONUCLEOTIDE DELIVERY: HELPING ANTISENSE DELIVER ON ITS PROMISE.", PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES, NATIONAL ACADEMY OF SCIENCES, US, vol. 93., 1 April 1996 (1996-04-01), US, pages 3161 - 3163., XP002910769, ISSN: 0027-8424, DOI: 10.1073/pnas.93.8.3161 *
ROJANASAKUL Y.: "ANTISENSE OLIGONUCLEOTIDE THERAPEUTICS: DRUG DELIVERY AND TARGETING", ADVANCED DRUG DELIVERY REVIEWS, ELSEVIER, AMSTERDAM, NL, vol. 18., 1 January 1996 (1996-01-01), AMSTERDAM, NL, pages 115 - 131., XP002910768, ISSN: 0169-409X, DOI: 10.1016/0169-409X(95)00020-8 *
See also references of EP0975371A4 *
SU Z.-Z., ET AL.: "C-FOS IS A POSITIVE REGULATOR OF CARCINOGEN ENHANCEMENT OF ADENOVIRUS TRANSFORMATION.", ONCOGENE, NATURE PUBLISHING GROUP, GB, vol. 10., 1 January 1995 (1995-01-01), GB, pages 2037 - 2049., XP002910767, ISSN: 0950-9232 *

Cited By (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1140971A1 (en) * 1999-01-07 2001-10-10 Isis Pharmaceuticals, Inc. Antisense modulation of novel anti-apoptotic bcl-2-related proteins
EP1140971A4 (en) * 1999-01-07 2002-05-15 Isis Pharmaceuticals Inc Antisense modulation of novel anti-apoptotic bcl-2-related proteins
EP1806353A1 (en) * 1999-01-07 2007-07-11 Isis Pharmaceuticals, Inc. Antisense modulation of novel anti-apoptotic BCL-2-related proteins
JP2004507263A (en) * 2000-09-02 2004-03-11 グリュネンタール・ゲゼルシャフト・ミト・ベシュレンクテル・ハフツング Antisense oligonucleotides to VR1
US7662948B2 (en) * 2000-09-02 2010-02-16 Gruenenthal Gmbh Antisense oligonucleotides against VR1
WO2003072114A1 (en) 2002-02-27 2003-09-04 Unisearch Limited Vascular therapeutics
EP1485109A1 (en) * 2002-02-27 2004-12-15 Unisearch Limited Vascular therapeutics
AU2003205434B2 (en) * 2002-02-27 2008-07-17 Unisearch Limited Vascular therapeutics
EP1485109A4 (en) * 2002-02-27 2009-07-29 Unisearch Ltd Vascular therapeutics
AU2008202976B2 (en) * 2002-02-27 2012-05-31 Newsouth Innovations Pty Limited Vascular therapeutics
US8242090B2 (en) 2002-02-27 2012-08-14 Newsouth Innovations Pty Limited Vascular therapeutics
US8686128B2 (en) 2002-02-27 2014-04-01 Levon Michael Khachigian Agent for targeting c-Jun mRNA

Also Published As

Publication number Publication date
EP0975371A1 (en) 2000-02-02
US20020039741A1 (en) 2002-04-04
EP0975371A4 (en) 2002-10-23
US6312900B1 (en) 2001-11-06
US20030194738A1 (en) 2003-10-16
US5985558A (en) 1999-11-16
AU6968898A (en) 1998-11-11

Similar Documents

Publication Publication Date Title
US5985558A (en) Antisense oligonucleotide compositions and methods for the inibition of c-Jun and c-Fos
US5877309A (en) Antisense oligonucleotides against JNK
US6133246A (en) Antisense oligonucleotide compositions and methods for the modulation of JNK proteins
US6238921B1 (en) Antisense oligonucleotide modulation of human mdm2 expression
US6391636B1 (en) Antisense oligonucleotide modulation of raf gene expression
AU732144B2 (en) Antisense oligonucleotide inhibition of RAS
US5968748A (en) Antisense oligonucleotide modulation of human HER-2 expression
US20020082415A1 (en) Antisense oligonucleotide modulation of human serine/threonine protein phosphatase gene expression
US6111094A (en) Enhanced antisense modulation of ICAM-1
US6001991A (en) Antisense oligonucleotide modulation of MDR P-glycoprotein gene expression
JP2002531574A (en) Methods of modulating tumor necrosis factor α-induced expression of cell adhesion molecules
US5981731A (en) Antisense oligonucleotide modulation of B-raf gene expression
JP2003505027A (en) Antisense modulation of PI3 kinase P110delta expression
JP2003520042A (en) Antisense modulation of inducible nitric oxide synthase expression
WO2003048315A2 (en) Antisense modulation of mdm2 expression
JP2003508089A (en) Antisense modulation of PI3 kinase p110 beta expression
JP2003512079A (en) Antisense modulation of protein kinase C-theta expression
WO1999063114A1 (en) Antisense oligonucleotide modulation of human thymidylate synthase expression
JP2003518928A (en) Antisense modulation of Akt-3 expression
US6784290B1 (en) Antisense oligonucleotide inhibition of ras
US6117847A (en) Oligonucleotides for enhanced modulation of protein kinase C expression
JP2003505037A (en) Antisense modulation of telomere repeat binding factor 1 expression
JP2003520242A (en) Antisense modulation of glycogen synthase kinase 3 alpha expression
US20070149472A1 (en) Antisense oligonucleotide compositions and methods for the modulation of jnk proteins
AU5188801A (en) Antisense oligonucleotide compositions and methods for the modulation of JNK proteins

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

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

AL Designated countries for regional patents

Kind code of ref document: A1

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

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
WWE Wipo information: entry into national phase

Ref document number: 1998915526

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 1998915526

Country of ref document: EP

REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

NENP Non-entry into the national phase

Ref country code: JP

Ref document number: 1998544162

Format of ref document f/p: F

NENP Non-entry into the national phase

Ref country code: CA

WWW Wipo information: withdrawn in national office

Ref document number: 1998915526

Country of ref document: EP