WO1999009214A1 - Antisense oligonucleotide compositions and methods for the modulation of jnk proteins - Google Patents

Antisense oligonucleotide compositions and methods for the modulation of jnk proteins Download PDF

Info

Publication number
WO1999009214A1
WO1999009214A1 PCT/US1998/016488 US9816488W WO9909214A1 WO 1999009214 A1 WO1999009214 A1 WO 1999009214A1 US 9816488 W US9816488 W US 9816488W WO 9909214 A1 WO9909214 A1 WO 9909214A1
Authority
WO
WIPO (PCT)
Prior art keywords
oligonucleotide
oligonucleotides
jnkl
protein
expression
Prior art date
Application number
PCT/US1998/016488
Other languages
French (fr)
Inventor
Robert Mckay
Nicholas Dean
Brett P. Monia
Pamela Scott Nero
William A. Gaarde
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 EP98939287A priority Critical patent/EP1003916A4/en
Priority to CA002301139A priority patent/CA2301139A1/en
Priority to JP2000509875A priority patent/JP3527200B2/en
Priority to AU87750/98A priority patent/AU730916B2/en
Publication of WO1999009214A1 publication Critical patent/WO1999009214A1/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
    • C12N15/1137Non-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 enzymes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H21/00Compounds containing two or more mononucleotide units having separate phosphate or polyphosphate groups linked by saccharide radicals of nucleoside groups, e.g. nucleic acids
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6876Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
    • C12Q1/6883Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material
    • C12Q1/6886Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material for cancer
    • 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
    • C12N2310/32Chemical 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
    • C12N2310/33Chemical 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
    • C12N2310/35Nature of the modification
    • C12N2310/351Conjugate
    • C12N2310/3515Lipophilic moiety, e.g. cholesterol
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/158Expression markers

Definitions

  • the present invention provides compositions and methods for detecting and modulating levels of Jun N- terminal inases (JNK proteins) , enzymes which are encoded by JNK genes.
  • the invention relates to antisense oligonucleotides specifically hybridizable with nucleic acids encoding JNK proteins. It has been found that antisense oligonucleotides can modulate the expression of these and other JNK proteins, kinases which were initially discovered due to their ability to catalyze the phosphorylation of the c-Jun subunit of transcription factor AP-1 and thereby increase AP-1 activity.
  • transcription factor AP-1 has been implicated in abnormal cell proliferation, oncogenic transformation, and tumor formation, development and maintenance (Vogt, Chapter 15 In : The FOS and JUN Families of Transcription Factors,
  • JNK proteins are aberrantly expressed in some neoplasms and tumors with resultant increased AP-1 activity, and (2) even in abnormally proliferating cells in which a JNK gene is not aberrantly expressed, inhibition of JNK expression will result in decreased AP-1 activity and thus, inhibition of abnormal cell proliferation and tumor formation, development and maintenance.
  • the invention is thus directed to diagnostic methods for detecting, and therapeutic methods for inhibiting, the hyperproliferation of cells and the formation, development and maintenance of tumors. Furthermore, this invention is directed to treatment of conditions associated with abnormal expression of JNK genes.
  • This invention also relates to therapies, diagnostics, and research reagents for disease states or disorders which respond to modulation of the expression of JNK proteins. Inhibition of the 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
  • AP-1 alter gene expression in response to growth factors, cytokines, tumor promoters, carcinogens and increased expression of certain oncogenes (Rahmsdorf, Chapter 13, and Rapp et al . , Chapter 16 In : The FOS and JUN Families of
  • AP-1 denotes one member of a family of related heterodimeric transcription factor complexes found in eukaryotic cells or viruses ⁇ The FOS and JUN Families of Transcription Factors, Angel and Herrlich, Eds., CRC Press, Boca Raton, FL, 1994; Bohmann et al . , Science, 1987, 238, 1386; Angel et al . , Na ture, 1988, 332, 166) .
  • Two relatively well-characterized AP-1 subunits are c-Fos and c-Jun; these two proteins are products of the c- fos and c-j un proto-oncogenes, respectively.
  • Repression of the activity of either c-fos or c-j un, or 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.
  • Mitogen-activated protein kinases MAPKs
  • 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).
  • MAPKs also referred to as extracellular signal-regulated protein kinases, or ERKs
  • ERKs extracellular signal-regulated protein kinases
  • ERKs extracellular signal-regulated protein kinases
  • MAPKs are themselves activated by phosphorylation catalyzed by, e.g., receptor tyrosine kinases, G protein-coupled receptors, protein kinase C (PKC) , and the apparently MAPK-dedicated kinases MEK1 and MEK2.
  • PKC protein kinase C
  • MEK1 and MEK2 the apparently MAPK-dedicated kinases
  • 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 (Seger et al . , FASEB J. , 1995, 9, 726; Cano et al .
  • MAP kinase pathway In a typical MAP kinase pathway, it is thought that a first MAP kinase, called a MEKK, phosphorylates and thereby activates a second MAP kinase, called a MEK, which, in turn, phosphorylates and activates a MAPK/ERK or JNK/SAPK enzyme ("SAPK” is an abbreviation for stress- activated protein kinase) . Finally, the activated MAPK/ERK or JNK/SAPK enzyme itself phosphorylates and activates a transcription factor (such as, e . g. , AP-1) or other substrates (Cano et al . , Trends Biochem . Sci . , 1995, 20, 117) .
  • This canonical cascade can be simply represented as follows :
  • JNK1 Jun N-terminal kinase 1
  • JNK2 Jun N- terminal kinase 2
  • JNK protein shall mean a member of the JNK family of kinases, including but not limited to JNK1, JNK2 and JNK3, their isoforms (Gupta et al . , EMBO J. , 1996, 15, 2760) and other members of the JNK family of proteins whether they function as Jun N-terminal kinases per se (that is, phosphorylate Jun at a specific amino terminally located position) or not.
  • At least one human leukemia oncogene has been shown to enhance Jun N-terminal kinase function (Raitano et al . , Proc . Na tl . Acad . Sci . (USA) , 1995, 92, 11746).
  • Modulation of the expression of one or more JNK proteins is desirable in order to interfere with hyperproliferation of cells and with the transcription of genes stimulated by AP-1 and other JNK protein phosphorylation substrates.
  • Modulation of the expression of one or more other JNK proteins is also desirable in order to interfere with hyperproliferation of cells resulting from abnormalities in specific signal transduction pathways.
  • cellular hyperproliferation in an animal can have several outcomes. Internal processes may eliminate hyperproliferative cells 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 hyperproliferative 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., Garland Publishing, Inc., New York, 1983) .
  • antisense oligonucleotides it has surprisingly been discovered that several genes encoding enzymes required for metastasis are positively regulated by AP-1, which may itself be modulated by antisense oligonucleotides targeted to one or more JNK proteins. Consequently, the invention satisfies the long-felt need for improved compositions and methods for modulating the metastasis of malignant tumors.
  • oligonucleotides are provided which specifically hybridize with a nucleic acid encoding a JNK protein.
  • Certain oligonucleotides of the invention are designed to bind either directly to mRNA transcribed from, or to a selected DNA portion of, a JNK gene that encodes a JNK protein, thereby modulating the expression thereof and/or the phosphorylation of one or more substrates for the JNK protein.
  • Pharmaceutical compositions comprising the oligonucleotides of the invention, and various methods of using the oligonucleotides of the invention, including methods of modulating one or more metastatic events, are also herein provided.
  • 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 genes ⁇ JNK) encoding Jun N-terminal kinase (JNK proteins) are particularly amenable to this approach.
  • JNK protein refers to proteins actually known to phosphorylate the amino terminal (N- terminal) portion of the Jun subunit of AP-1, as well as those that have been tentatively identified as JNK proteins based on amino acid sequence but which may in fact additionally or alternatively bind and/or phosphorylate either other transcription factors (e.g., ATF2) or kinase substrates that are not known to be involved in transcription (Derijard et al . , Cell , 1994, 76, 1025; Kallunki et al . , Genes & Development , 1994, 8, 2996; Gupta et al . , EMBO J.
  • ATF2 kinase substrates
  • the present invention is directed to antisense oligonucleotides that modulate the JNK1, JNK2 and JNK3 proteins.
  • the present invention is directed to antisense oligonucleotides that modulate the JNK1, JNK2 and JNK3 proteins.
  • JNK1, JNK2 and JNK3 proteins As a consequence of the association between cellular proliferation and activation ⁇ via phosphorylation) of AP-1, other transcription factors and/or other proteins by JNK proteins, inhibition of the expression of one or more JNK proteins leads to inhibition of the activation of AP-1 and/or other factors involved in cellular proliferation, cell cycle progression or metastatic events, and, accordingly, results in modulation of these activities.
  • Such modulation is desirable for treating, alleviating or preventing 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 the expression of one JNK protein can be combined with modulation of one or more additional JNK proteins in order to achieve a requisite level of interference with AP-1-mediated transcription .
  • Methods of modulating the expression of JNK proteins comprising contacting animals with oligonucleotides specifically hybridizable with a nucleic acid encoding a JNK protein are herein provided. These methods are believed to be useful both therapeutically and diagnostically as a consequence of the association between kinase-mediated activation of AP-1 and cellular proliferation. These methods are also useful as tools, for example, in the detection and determination of the role of kinase-mediated activation of AP-1 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 JNK-mediated activation using the oligonucleotides of the invention.
  • Methods of treating conditions in which abnormal or excessive JNK-mediated cellular proliferation occurs 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.
  • the present invention employs oligonucleotides for use in antisense modulation of the function of DNA or messenger RNA (mRNA) encoding a protein the modulation of which is desired, and ultimately to regulate the amount of such a protein.
  • mRNA messenger RNA
  • Hybridization of an antisense oligonucleotide with its mRNA target 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.
  • modulation means either an increase (stimulation) or a decrease (inhibition) in the expression of the protein.
  • inhibition is the preferred form of modulation of gene expression.
  • 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 associated with hyperproliferative disorders.
  • the targeting process also includes determination of a site or sites 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 site or sites 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 5' untranslated 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”
  • the 3' untranslated 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'-UTR, tIR, ORF, tTR, 3'-UTR.
  • ORFs As is 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 , 1983, Garland Publishing Inc., New York, pp. 411-415). 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.
  • 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 vitro (Brigstock et al., Growth Factors, 1990, 4, 45; Gelbert et al., Somat. Cell. Mol. Genet., 1990, 16, 173; Gold and Stormo, in:
  • ransmid DNA Escherichia coli and Salmonella typhimurium: Cellular and Molecular Biology, Vol. 2, 1987, Neidhardt et al., eds., American Society for Microbiology, Washington, D.C., p. 1303,) .
  • 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) .
  • 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.
  • 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 JNK 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.
  • Oligonucleotides of the Invention The present invention employs oligonucleotides for use in antisense modulation of one or more JNK proteins.
  • 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.
  • 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.
  • the oligonucleotides in accordance with this invention preferably comprise from about 8 to about 30 nucleotides.
  • An unmodified (naturally occurring) nucleotide has three components: (1) a nitrogen- containing heterocyclic ase linked by one of its nitrogen atoms to (2) a 5-pentofuranosyl sugar and (3) a phospha te esterified to one of the 5' or 3' carbon atoms of the sugar.
  • the phosphate of a first nucleotide is also esterified to an adjacent sugar of a second, adjacent nucleotide via a 3 '-5' phosphate linkage.
  • the "backbone" of an unmodified oligonucleotide consists of (2) and (3), that is, sugars linked together by phosphodiester linkages between the 5' carbon of the sugar of a first nucleotide and the 3' carbon 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 Replica tion, W.H.
  • Oligonucleotides may comprise nucleotide sequences sufficient in identity and number to effect specific hybridization with a particular nucleic acid.
  • 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.
  • 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.
  • “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.
  • An oligonucleotide is specifically hybridizable to its target sequence due to the formation of base pairs between specific partner nucleobases in the interior of a nucleic acid duplex.
  • guanine (G) binds to cytosine (C)
  • adenine (A) binds to thymine (T) or uracil (U) .
  • nucleobase equivalents include 5- methylcytosine, 5-hydroxymethylcytosine (HMC) , glycosyl HMC and gentiobiosyl HMC (C equivalents) , and 5- hydroxymethyluracil (U equivalent) .
  • synthetic nucleobases which retain partner specificity include, for example, 7-deaza-Guanine, which retains partner specificity for C.
  • an oligonucleotide' s capacity to specifically hybridize with its target sequence will not be altered by any chemical modification to a nucleobase in the nucleotide sequence of the oligonucleotide which does not significantly effect its specificity for the partner nucleobase in the target oligonucleotide. 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 there is a sufficient degree of complementarity to avoid nonspecific 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.
  • Antisense oligonucleotides are commonly used as research reagents, diagnostic aids, and therapeutic agents.
  • 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.
  • Modified Linkages 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.
  • 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 O-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 No.
  • 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- hydroxymethylcytosine (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 Replica tion, W.H.
  • nucleobases found only infrequently or transiently in natural nucleic acids, e.g., hypoxanthine, 6-methyladenine, 5-methylcytosine, 5- hydroxymethylcytosine (HMC) ,
  • 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 x to C 10 lower alkyl, alkoxyalkoxy, substituted lower alkyl, alkaryl or aralkyl; Cl; Br; CN; CF 3 ; OCF 3 ; 0-, S-, or N- alkyl; 0-, S-, or N-alkenyl; SOCH 3 ; S0 2 CH 3 ; 0N0 2 ; N0 2 ; N 3 ; NH 2 ; heterocycloalkyl; heterocycloalkaryl; aminoalkylamino; polyalkylamino; substituted silyl; an
  • a preferred modification includes 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) .
  • 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 Conj uga tes (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, G., 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. U.S.A., 1989, 86:6553), cholic acid (Manoharan et al., Bioorg. Med. Chem.
  • 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.
  • 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.
  • Oligonucleotides comprising lipophilic moieties, and methods for preparing such oligonucleotides, are disclosed in U.S. Patents Nos. 5,138,045, 5,218,105 and 5,459,255.
  • 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) and those having substantially chirally pure (Sp or Rp) alkylphosphonate, phosphoamidate or phosphotriester linkages (Cook, U.S. Patents Nos. 5,212,295 and 5,521,302).
  • 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. Cleavage of the RNA target can be routinely detected by gel electrophoresis and, if necessary, associated nucleic acid hybridization techniques known in the art.
  • such “chimeras” may be “gap ers,” 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) .
  • nuclease activity e.g., 2 '-fluoro- or 2 ' -methoxyethoxy- substituted
  • 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 .
  • 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.
  • 5,506,351 drawn to processes for the preparation of 2'-0-alkyl guanosine and related compounds, including 2, 6-diaminopurine compounds;
  • U.S. Patent No. 5,587,469 drawn to oligonucleotides having N-2 substituted purines;
  • U.S. Patent No. 5,587,470 drawn to oligonucleotides having 3-deazapurines;
  • U.S. Patent Nos. 5,602,240, and 5,610,289 drawn to backbone modified oligonucleotide analogs; and
  • 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.
  • N-Benzoyl-2 ' -O-methoxyethyl-5 ' -O- dimethoxytrityl-5- ⁇ tethylcytidine-3 ' -amidite N-Benzoyl-2 ' - O-methoxyethyl-5 ' -O-dimethoxytrityl-5-methylcytidine (74 g, 0.10 M) was dissolved in CH 2 C1 2 (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 NaHC0 3 (lx 300 mL) and saturated NaCl (3x 300 mL) .
  • the aqueous washes were back- extracted with CH 2 C1 2 (300 mL) , and the extracts were combined, dried over MgS0 4 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 .
  • the compounds of the invention encompass any pharmaceutically acceptable salts, esters, or salts of such esters, or any other compound which, upon administration to an animal including a human, is capable of providing (directly or indirectly) the biologically active metabolite or residue thereof. Accordingly, for example, the disclosure is also drawn to "prodrugs” and “pharmaceutically acceptable salts” of the oligonucleotides of the invention, pharmaceutically acceptable salts of such prodrugs, and other bioequivalents.
  • 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.
  • 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.
  • compositions of the oligonucleotides of the invention refers to physiologically and pharmaceutically acceptable salts of the oligonucleotides of the invention: i.e., salts that retain the desired biological activity of the parent compound and do not impart undesired toxicological effects thereto.
  • Pharmaceutically acceptable base addition salts are formed with metals or amines, such as alkali and alkaline earth metals or organic amines. Examples of metals used as cations are sodium, potassium, magnesium, calcium, and the like.
  • Suitable amines are N,N ' -dibenzylethylenediamine, chloroprocaine, choline, diethanolamine, dicyclohexylamine, ethylenediamine, N-methylglucamine, and procaine (see, for example, Berge et al . , "Pharmaceutical Salts," J. of Pharma Sci . , 1977, 66:1) .
  • the base addition salts of said acidic compounds are prepared by contacting the free acid form with a sufficient amount of the desired base to produce the salt in the conventional manner.
  • the free acid form may be regenerated by contacting the salt form with an acid and isolating the free acid in the conventional manner.
  • a "pharmaceutical addition salt” includes a pharmaceutically acceptable salt of an acid form of one of the components of the compositions of the invention. These include organic or inorganic acid salts of the amines. Preferred acid salts are the hydrochlorides, acetates, salicylates, nitrates and phosphates.
  • Suitable pharmaceutically acceptable salts include basic salts of a variety of inorganic and organic acids, such as, for example, with inorganic acids, such as for example hydrochloric acid, hydrobromic acid, sulfuric acid or phosphoric acid; with organic carboxylic, sulfonic, sulfo or phospho acids or N-substituted sulfamic acids, for example acetic acid, propionic acid, glycolic acid, succinic acid, maleic acid, hydroxymaleic acid, methylmaleic acid, fumaric acid, malic acid, tartaric acid, lactic acid, oxalic acid, gluconic acid, glucaric acid, glucuronic acid, citric acid, benzoic acid, cinnamic acid, mandelic acid, salicylic acid, 4-aminosalicylic acid, 2-phenoxybenzoic acid, 2-acetoxybenzoic acid, embonic acid, nicotinic acid or isonicotin
  • Suitable pharmaceutically acceptable cations are well known to those skilled in the art and include alkaline, alkaline earth, ammonium and quaternary ammonium cations. Carbonates or hydrogen carbonates are also possible.
  • preferred examples of pharmaceutically acceptable salts include but are not limited to (a) salts formed with cations such as sodium, potassium, ammonium, magnesium, calcium, polyamines such as spermine and spermidine, etc.
  • salts formed with inorganic acids for example hydrochloric acid, hydrobromic acid, sulfuric acid, phosphoric acid, nitric acid and the like
  • salts formed with organic acids such as, for example, acetic acid, oxalic acid, tartaric acid, succinic acid, maleic acid, fumaric acid, gluconic acid, citric acid, malic acid, ascorbic acid, benzoic acid, tannic acid, palmitic acid, alginic acid, polyglutamic acid, naphthalenesulfonic acid, methanesulfonic acid, p-toluenesulfonic acid, naphthalenedisulfonic acid, polygalacturonic acid, and the like; and (d) salts formed from elemental anions such as chlorine, bromine, and iodine .
  • oligonucleotides of the present invention specifically hybridize to nucleic acids (e.g., mRNAs) encoding a JNK protein.
  • the oligonucleotides of the present invention can be utilized as therapeutic compounds, as diagnostic tools or research reagents that can be incorporated into kits, and in purifications and cellular product preparations, as well as other methodologies, which are appreciated by persons of ordinary skill in the art.
  • the oligonucleotides of the present invention can be used to detect the presence of JNK protein-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 , Cold Spring Harbor Laboratory Press, 1989, Volume 2, pg.
  • Radiolabeled oligonucleotides are then contacted with cell or tissue samples suspected of containing JNK protein message RNAs (and thus JNK 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 JNK 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 JNK protein 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 JNK protein 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. Other means of labeling oligonucleotides are known in the art (see, e . g. , Ruth, Chapter 6 In : Methods in Molecular Biology, Vol . 26: Protocols for Oligonucleotide Conj uga tes, Agrawal, ed. , Humana Press Inc., Totowa, NJ, 1994, pages 167-185) .
  • Kits for detecting the presence or absence of expression of a JNK protein may also be prepared.
  • Such kits include an oligonucleotide targeted to an appropriate gene, i.e., a gene encoding a JNK 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 JNK 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.
  • the oligonucleotides of the invention are also useful for the purification of specific Jun kinase proteins from cells that normally express a set of JNK proteins which are similar to each other in terms of their polypeptide sequences and biochemical properties.
  • the purification of a JNK1 protein from cells that expresses JNK1, JNK2 and JNK3 proteins can be enhanced by first treating such cells with oligonucleotides that inhibit the expression of JNK2 and JNK3 and/or with oligonucleotides that increase the expression of JNK1, because such treatments will increase the relative ratio of JNK1 relative to JNK2 and JNK3.
  • the yield of JNK1 from subsequent purification steps will be improved as the amount of the biochemically similar (and thus likely to contaminate) JNK2 and JNK3 proteins in extracts prepared from cells so treated will be diminished.
  • Biologically Active Oligonucleotides The invention is also drawn to the administration of oligonucleotides having biological activity to cultured cells, isolated tissues and organs and animals.
  • having biological activity it is meant that the oligonucleotide functions to modulate the expression of one or more genes in cultured cells, isolated tissues or organs and/or animals.
  • Such modulation can be achieved by an antisense oligonucleotide by a variety of mechanisms known in the art, including but not limited to transcriptional arrest; effects on RNA processing (capping, polyadenylation and splicing) and transportation; enhancement of cellular degradation of the target nucleic acid; and translational arrest (Crooke et al . , Exp. Opin . Ther. Pa tents, 1996, 6:855) .
  • compositions and methods of the invention can be used to study the function of one or more genes in the animal.
  • antisense oligonucleotides have been systemically administered to rats in order to study the role of the W-methyl-D-aspartate receptor in neuronal death, to mice in order to investigate the biological role of protein kinase C- , and to rats in order to examine the role of the neuropeptide Yl receptor in anxiety (Wahlestedt et al . , Na ture, 1993, 363:260; Dean et al . , Proc . Na tl . Acad. Sci . U. S . A.
  • antisense knockouts i.e., inhibition of a gene by systemic administration of antisense oligonucleotides
  • antisense oligonucleotides may represent the most accurate means for examining a specific member of the family (see, generally, Albert et al . , Trends Pharmacol . Sci . , 1994, 15:250).
  • compositions and methods of the invention also have therapeutic uses in an animal, including a human, having (i.e., suffering from), or known to be or suspected of being prone to having, a disease or disorder that is treatable in whole or in part with one or more nucleic acids.
  • 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 JNK 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 carrier such as, e.g., a diluent.
  • a suitable pharmaceutically acceptable carrier such as, e.g., a diluent.
  • 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.
  • 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 following U.S. patents demonstrate palliative, therapeutic and other methods utilizing antisense oligonucleotides.
  • U. S. Patent No. 5,135,917 provides antisense oligonucleotides that inhibit human interleukin-1 receptor expression.
  • U.S. Patent No. 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 No. 5,087,617 provides methods for treating cancer patients with antisense oligonucleotides.
  • Patent No. 5,166,195 provides oligonucleotide inhibitors of Human Immunodeficiency Virus (HIV).
  • U.S. Patent No. 5,004,810 provides oligomers capable of hybridizing to herpes simplex virus Vmw65 mRNA and inhibiting replication.
  • U.S. Patent No. 5,194,428 provides antisense oligonucleotides having antiviral activity against influenzavirus .
  • U.S. Patent No. 4,806,463 provides antisense oligonucleotides and methods using them to inhibit HTLV-III replication.
  • U.S. Patent No. 5,286,717 provides oligonucleotides having a complementary base sequence to a portion of an oncogene.
  • U.S. Patent No. 5,264,423 are directed to phosphorothioate oligonucleotide analogs used to prevent replication of foreign nucleic acids in cells.
  • U.S. Patent No. 4,689,320 is directed to antisense oligonucleotides as antiviral agents specific to cytomegalovirus (CMV) .
  • CMV cytomegalovirus
  • U.S. Patent No. 5,098,890 provides oligonucleotides complementary to at least a portion of the mRNA transcript of the human c-myb gene.
  • U.S. Patent No. 5,242,906 provides antisense oligonucleotides useful in the treatment of latent Epstein-Barr virus (EBV) infections.
  • EBV Epstein-Barr virus
  • the term "disease or disorder” (1) includes any abnormal condition of an organism or part, especially as a consequence of infection, inherent weakness, environmental stress, that impairs normal physiological functioning; (2) excludes pregnancy per se but not autoimmune and other diseases associated with pregnancy; and (3) includes cancers and tumors.
  • the term "known to be or suspected of being prone to having a disease or disorder” indicates that the subject animal has been determined to be, or is suspected of being, at increased risk, relative to the general population of such animals, of developing a particular disease or disorder as herein defined.
  • a subject animal "known to be or suspected of being prone to having a disease or disorder” could have a personal and/or family medical history that includes frequent occurrences of a particular disease or disorder.
  • a subject animal "known to be or suspected of being prone to having a disease or disorder" could have had such a susceptibility determined by genetic screening according to techniques known in the art (see, e . g. , U.S. Congress, Office of Technology Assessment, Chapter 5 In : Genetic Moni toring and Screening in the Workplace, OTA-BA-455, U.S. Government Printing Office, Washington, D.C., 1990, pages 75-99).
  • a disease or disorder that is treatable in whole or in part with one or more nucleic acids refers to a disease or disorder, as herein defined, (1) the management, modulation or treatment thereof, and/or (2) therapeutic, curative, palliative and/or prophylactic relief therefrom, can be provided via the administration of an antisense oligonucleotide .
  • Pharmaceutical Compositions The formulation of pharmaceutical compositions comprising the oligonucleotides of the invention, and their subsequent administration, are believed to be within the skill of those in the art. A.
  • a patient i.e., an animal, including a human, having or predisposed to a disease or disorder
  • a pharmaceutically acceptable carrier in doses ranging from 0.01 ⁇ g 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.
  • the term "treatment regimen” is meant to encompass therapeutic, palliative and prophylactic modalities.
  • the dosage of the nucleic acid 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 ablated.
  • 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.
  • 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.
  • An optimal dosing schedule is used to deliver a therapeutically effective amount of the oligonucleotide being administered via a particular mode of administration.
  • the term "therapeutically effective amount,” for the purposes of the invention refers to the amount of oligonucleotide-containing pharmaceutical composition which is effective to achieve an intended purpose without undesirable side effects (such as toxicity, irritation or allergic response) .
  • side effects such as toxicity, irritation or allergic response
  • human doses can be extrapolated from animal studies (Katocs et al .
  • the dosage required to provide an effective amount of a pharmaceutical composition will vary depending on the age, health, physical condition, weight, type and extent of the disease or disorder of the recipient, frequency of treatment, the nature of concurrent therapy (if any) and the nature and scope of the desired effect (s) (Nies et al . , Chapter 3 In : Goodman & Gilman ' s The Pharmacological Basis of Therapeutics, 9th Ed., Hardman et al .
  • the term "high risk individual” is meant to refer to an individual for whom it has been determined, via, e.g., individual or family history or genetic testing, has a significantly higher than normal probability of being susceptible to the onset or recurrence of a disease or disorder.
  • the individual can be prophylactically treated to prevent the onset or recurrence of the disease or disorder.
  • prophylactically effective amount is meant to refer to an amount of a pharmaceutical composition which produces an effect observed as the prevention of the onset or recurrence of a disease or disorder.
  • Prophylactically effective amounts of a pharmaceutical composition are typically determined by the effect they have compared to the effect observed when a second pharmaceutical composition lacking the active agent is administered to a similarly situated individual.
  • nucleic acid is administered in maintenance doses, ranging from 0.01 ⁇ g to 100 g per kg of body weight, once or more daily, to once every 20 years.
  • maintenance doses ranging from 0.01 ⁇ g to 100 g per kg of body weight, once or more daily, to once every 20 years.
  • preventative doses ranging from 0.01 ⁇ g to 100 g per kg of body weight, once or more daily, to once every 20 years.
  • an individual may be made less susceptible to an inflammatory condition that is expected to occur as a result of some medical treatment, e.g., graft versus host disease resulting from the transplantation of cells, tissue or an organ into the individual.
  • 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.
  • 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-
  • 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) .
  • a first antisense oligonucleotide targeted to a first JNK protein is used in combination with a second antisense oligonucleotide targeted to a second JNK protein in order to such JNK proteins to a more extensive degree than can be achieved when either oligonucleotide is used individually.
  • the first and second JNK proteins which are targeted by such oligonucleotides are identical, are different JNK proteins or are different isoforms of the same JNK protein.
  • compositions for the non-parenteral administration of oligonucleotides may include sterile aqueous solutions which may also contain buffers, diluents and other suitable additives.
  • Pharmaceutically acceptable organic or inorganic carrier substances suitable for non-parenteral administration which do not deleteriously react with oligonucleotides can be used.
  • Suitable pharmaceutically acceptable carriers include, but are not limited to, water, salt solutions, alcohol, polyethylene glycols, gelatin, lactose, amylose, magnesium stearate, talc, silicic acid, viscous paraffin, hydroxymethylcellulose, polyvinylpyrrolidone and the like.
  • compositions can be sterilized and, if desired, mixed with auxiliary agents, e.g., lubricants, preservatives, stabilizers, wetting agents, emulsifiers, salts for influencing osmotic pressure, buffers, colorings flavorings and/or aromatic substances and the like which do not deleteriously react with the oligonucleotide (s) of the pharmaceutical composition.
  • auxiliary agents e.g., lubricants, preservatives, stabilizers, wetting agents, emulsifiers, salts for influencing osmotic pressure, buffers, colorings flavorings and/or aromatic substances and the like which do not deleteriously react with the oligonucleotide (s) of the pharmaceutical composition.
  • Pharmaceutical compositions in the form of aqueous suspensions may contain substances which increase the viscosity of the suspension including, for example, sodium carboxymethylcellulose, sorbitol and/or dextran.
  • such suspensions may also contain stabilize
  • an oligonucleotide is administered via the rectal mode.
  • pharmaceutical compositions for rectal administration include foams, solutions (enemas) and suppositories. Rectal suppositories for adults are usually tapered at one or both ends and typically weigh about 2 g each, with infant rectal suppositories typically weighing about one-half as much, when the usual base, cocoa butter, is used (Block, Chapter 87 In : Remington ' ' s Pharmaceutical Sciences, 18th Ed., Gennaro, ed., Mack Publishing Co., Easton, PA, 1990) .
  • one or more oligonucleotides are administered via oral delivery.
  • Pharmaceutical compositions for oral administration include powders or granules, suspensions or solutions in water or non-aqueous media, capsules, sachets, troches, tablets or SECs (soft elastic capsules or "caplets") . Thickeners, flavoring agents, diluents, emulsifiers, dispersing aids, carrier substances or binders may be desirably added to such pharmaceutical compositions.
  • the use of such pharmaceutical compositions has the effect of delivering the oligonucleotide to the alimentary canal for exposure to the mucosa thereof.
  • the pharmaceutical composition can comprise material effective in protecting the oligonucleotide from pH extremes of the stomach, or in releasing the oligonucleotide over time, to optimize the delivery thereof to a particular mucosal site.
  • Enteric coatings for acid-resistant tablets, capsules and caplets are known in the art and typically include acetate phthalate, propylene glycol and sorbitan monoleate.
  • Various methods for producing pharmaceutical compositions for alimentary delivery are well known in the art. See, generally, Nairn, Chapter 83; Block, Chapter 87; Rudnic et al . , Chapter 89; Porter, Chapter 90; and Longer et al .
  • oligonucleotides of the invention can be incorporated in a known manner into customary pharmaceutical compositions, such as tablets, coated tablets, pills, granules, aerosols, syrups, emulsions, suspensions and solutions, using inert, non-toxic, pharmaceutically acceptable carriers (excipients) .
  • the therapeutically active compound should in each case be present here in a concentration of about 0.5% to about 95% by weight of the total mixture, i.e., in amounts which are sufficient to achieve the stated dosage range.
  • compositions are prepared, for example, by diluting the active compounds with pharmaceutically acceptable carriers, if appropriate using emulsifying agents and/or dispersing agents, and, for example, in the case where water is used as the diluent, organic solvents can be used as auxiliary solvents if appropriate.
  • Pharmaceutical compositions may be formulated in a conventional manner using additional pharmaceutically acceptable carriers as appropriate.
  • compositions may be prepared by conventional means with additional excipients such as binding agents (e.g., pregelatinised maize starch, polyvinylpyrrolidone or hydroxypropyl methylcellulose) ; fillers (e.g., lactose, microcrystalline cellulose or calcium hydrogen phosphate); lubricants (e.g., magnesium stearate, talc or silica); disintegrates (e.g., starch or sodium starch glycolate) ; or wetting agents (e.g., sodium lauryl sulfate). Tablets may be coated by methods well known in the art.
  • the preparations may also contain flavoring, coloring and/or sweetening agents as appropriate.
  • compositions which may conveniently be presented in unit dosage form, may be prepared according to conventional techniques well known in the pharmaceutical industry. Such techniques include the step of bringing into association the active ingredient (s) with the pharmaceutically acceptable carrier (s). In general the pharmaceutical compositions are prepared by uniformly and intimately bringing into association the active ingredient (s) with liquid excipients or finely divided solid excipients or both, and then, if necessary, shaping the product.
  • compositions of the present invention suitable for oral administration may be presented as discrete units such as capsules, cachets or tablets each containing predetermined amounts of the active ingredients; as powders or granules; as solutions or suspensions in an aqueous liquid or a non-aqueous liquid; or as oil-in-water emulsions or water-in-oil liquid emulsions.
  • a tablet may be made by compression or molding, optionally with one or more accessory ingredients.
  • Compressed tablets may be prepared by compressing in a suitable machine, the active ingredients in a free-flowing form such as a powder or granules, optionally mixed with a binder, lubricant, inert diluent, preservative, surface active or dispersing agent.
  • Molded tablets may be made by molding in a suitable machine a mixture of the powdered compound moistened with an inert liquid diluent.
  • the tablets may optionally be coated or scored and may be formulated so as to provide slow or controlled release of the active ingredients therein.
  • Pharmaceutical 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.
  • C. Penetration Enhancers Pharmaceutical compositions comprising the oligonucleotides of the present invention may also include penetration enhancers in order to enhance the alimentary delivery of the oligonucleotides.
  • Penetration enhancers may be classified as belonging to one of five broad categories, i.e., fatty acids, bile salts, chelating agents, surfactants and non-surfactants (Lee et al . , Cri tical Reviews in Therapeutic Drug Carrier Systems, 1991, 8:91-192; Muranishi, Cri tical Reviews in Therapeutic Drug Carrier Systems, 1990, 7:1) .
  • Fatty Acids Various fatty acids and their derivatives which act as penetration enhancers include, for example, oleic acid, lauric acid, capric acid, myristic acid, palmitic acid, stearic acid, linoleic acid, linolenic acid, dicaprate, tricaprate, recinleate, monoolein (a.k.a.
  • Bile Salts The physiological roles of bile include the facilitation of dispersion and absorption of lipids and fat-soluble vitamins (Brunton, Chapter 38 In : Goodman & Gilman ' s The Pharmacological Basis of Therapeutics, 9th Ed., Hardman et al . , eds., McGraw-Hill, New York, NY, 1996, pages 934-935) .
  • Various natural bile salts, and their synthetic derivatives act as penetration enhancers.
  • the term "bile salt” includes any of the naturally occurring components of bile as well as any of their synthetic derivatives.
  • Chelating agents have the added advantage of also serving as DNase inhibitors and include, but are not limited to, disodium ethylenediaminetetraacetate (EDTA) , citric acid, salicylates (e.g., sodium salicylate, 5-methoxysalicylate and homovanilate) , W-acyl derivatives of collagen, laureth- 9 and IV-amino acyl derivatives of beta-diketones (enamines) (Lee et al . , Cri tical Reviews in Therapeutic Drug Carrier Systems, 1991, page 92; Muranishi, Cri tical Reviews in Therapeutic Drug Carrier Systems, 1990, 7:1; Buur et al . , J. Control Rel . , 1990, 14:43).
  • EDTA disodium ethylenediaminetetraacetate
  • citric acid citric acid
  • salicylates e.g., sodium salicylate, 5-methoxysalicylate and homovanilate
  • Surfactants include, for example, sodium lauryl sulfate, polyoxyethylene-9-lauryl ether and polyoxyethylene-20-cetyl ether (Lee et al . , Cri tical Reviews in Therapeuti c Drug Carrier Systems, 1991, page 92) ; and perfluorochemical emulsions, such as FC-43 (Takahashi et al . , J. Pharm . Phama col . , 1988, 40:252). 5.
  • Non-surfactants include, for example, unsaturated cyclic ureas, 1-alkyl- and 1-alkenylazacyclo-alkanone derivatives (Lee et a l . , Cri tical Reviews in Therapeutic Drug Carrier Systems, 1991, page 92) ; and non-steroidal anti-inflammatory agents such as diclofenac sodium, indomethacin and phenylbutazone (Yamashita et al . , J. Pharm . Pharmacol . , 1987, 39:621).
  • carrier compound refers to a nucleic acid, or analog thereof, which is inert (i.e., does not possess biological activity per se) but is recognized as a nucleic acid by in vivo processes that reduce the bioavailability of a nucleic acid having biological activity by, for example, degrading the biologically active nucleic acid or promoting its removal from circulation.
  • carrier compound typically with an excess of the latter substance, can result in a substantial reduction of the amount of nucleic acid recovered in the liver, kidney or other extracirculatory reservoirs, presumably due to competition between the carrier compound and the nucleic acid for a common receptor.
  • the recovery of a partially phosphorothioated oligonucleotide in hepatic tissue is reduced when it is coadministered with polyinosinic acid, dextran sulfate, polycytidic acid or 4- acetamido-4 ' -isothiocyano-stilbene-2 , 2 ' -disulfonic acid (Miyao et al . , Antisense Res . Dev. , 1995, 5:115; Takakura et al . , Antisense & Nucl . Acid Drug Dev. , 1996, 6:177).
  • a "pharmaceutically acceptable carrier” is a pharmaceutically acceptable solvent, suspending agent or any other pharmacologically inert vehicle for delivering one or more nucleic acids to an animal.
  • the pharmaceutically acceptable carrier may be liquid or solid and is selected with the planned manner of administration in mind so as to provide for the desired bulk, consistency, etc. , when combined with a nucleic acid and the other components of a given pharmaceutical composition.
  • Typical pharmaceutically acceptable carriers include, but are not limited to, binding agents (e.g., pregelatinised maize starch, polyvinylpyrrolidone or hydroxypropyl methylcellulose, etc.); fillers (e.g., lactose and other sugars, microcrystalline cellulose, pectin, gelatin, calcium sulfate, ethyl cellulose, polyacrylates or calcium hydrogen phosphate, etc. ) ; lubricants (e.g., magnesium stearate, talc, silica, colloidal silicon dioxide, stearic acid, metallic stearates, hydrogenated vegetable oils, corn starch, polyethylene glycols, sodium benzoate, sodium acetate, etc.
  • binding agents e.g., pregelatinised maize starch, polyvinylpyrrolidone or hydroxypropyl methylcellulose, etc.
  • fillers e.g., lactose and other sugars, microcrystalline cellulose, pectin, gelatin
  • disintegrates e.g., starch, sodium starch glycolate, etc.
  • wetting agents e.g., sodium lauryl sulphate, etc.
  • Sustained release oral delivery systems and/or enteric coatings for orally administered dosage forms are described in U.S. Patents Nos. 4,704,295; 4,556,552; 4,309,406; and 4,309,404.
  • compositions of the present invention may additionally contain other adjunct components conventionally found in pharmaceutical compositions, at their art-established usage levels.
  • the compositions may contain additional compatible pharmaceutically-active materials such as, e.g., antipruritics, astringents, local anesthetics or anti-inflammatory agents, or may contain additional materials useful in physically formulating various dosage forms of the composition of present invention, such as dyes, flavoring agents, preservatives, antioxidants, opacifiers, thickening agents and stabilizers.
  • additional compatible pharmaceutically-active materials such as, e.g., antipruritics, astringents, local anesthetics or anti-inflammatory agents
  • additional materials useful in physically formulating various dosage forms of the composition of present invention such as dyes, flavoring agents, preservatives, antioxidants, opacifiers, thickening agents and stabilizers.
  • such materials when added, should not unduly interfere with the biological activities of the components of the compositions of the invention.
  • 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, 6, 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 . , 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.
  • lipids useful in liposome production include phosphatidyl compounds, such as phosphatidylglycerol, phosphatidylcholine, phosphatidylserine, sphingolipids, phosphatidylethanolamine, 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 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 comprising oligonucleotides intended for administration to an animal.
  • animal is meant to encompass humans as well as other mammals, as well as reptiles, amphibians, and birds.
  • parenteral delivery refers to the administration of an oligonucleotide of the invention to an animal in a manner other than through the digestive canal.
  • Means of preparing and administering parenteral pharmaceutical compositions are known in the art (see, e.g., Avis, Chapter 84 In : Remington ' s Pharmaceutical Sciences, 18th Ed., Gennaro, ed., Mack Publishing Co., Easton, PA, 1990, pages 1545- 1569) .
  • Parenteral means of delivery include, but are not limited to, the following illustrative examples.
  • 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, 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).
  • a biocompatible matrix such as a hydrogel material
  • 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. 5.
  • Intrathecal drug administration for the introduction of a 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.
  • 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).
  • 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) .
  • Epidermal and Transdermal Delivery in which pharmaceutical compositions containing drugs are applied topically, can be used to administer drugs to be absorbed by the local dermis or for further penetration and absorption by underlying tissues, respectively.
  • Means of preparing and administering medications topically are known in the art (see, e.g., Block, Chapter 87 In : Remington ' s Pharmaceutical Sciences, 18th Ed., Gennaro, ed. , Mack Publishing Co., Easton, PA, 1990, pages 1596-1609).
  • Vaginal Delivery provides local treatment and avoids first pass metabolism, degradation by digestive enzymes, and potential systemic side-effects. This mode of administration may be preferred for antisense oligonucleotides targeted to pathogenic organisms for which the vagina is the usual habitat, e.g., Trichomonas vaginalis . In another embodiment, antisense oligonucleotides to genes encoding sperm-specific antibodies can be delivered by this mode of administration in order to increase the probability of conception and subsequent pregnancy. Vaginal suppositories (Block, Chapter 87 In : Remington ' s Pharmaceutical Sciences, 18th Ed., Gennaro, ed. , Mack Publishing Co., Easton, PA, 1990, pages 1609-1614) or topical ointments can be used to effect this mode of delivery.
  • Intravesical Delivery provides local treatment and avoids first pass metabolism, degradation by digestive enzymes, and potential systemic side-effects.
  • the method requires urethral catheterization of the patient and a skilled staff. Nevertheless, this mode of administration may be preferred for antisense oligonucleotides targeted to pathogenic organisms, such as T. vaginalis, which may invade the urogenital tract.
  • Alimentary Delivery refers to the administration, directly or otherwise, to a portion of the alimentary canal of an animal.
  • the term “alimentary canal” refers to the tubular passage in an animal that functions in the digestion and absorption of food and the elimination of food residue, which runs from the mouth to the anus, and any and all of its portions or segments, e.g., the oral cavity, the esophagus, the stomach, the small and large intestines and the colon, as well as compound portions thereof such as, e.g., the gastro-intestinal tract.
  • the term “alimentary delivery” encompasses several routes of administration including, but not limited to, oral, rectal, endoscopic and sublingual/buccal administration. A common requirement for these modes of administration is absorption over some portion or all of the alimentary tract and a need for efficient mucosal penetration of the nucleic acid(s) so administered .
  • Delivery of a drug via the oral mucosa has several desirable features, including, in many instances, a more rapid rise in plasma concentration of the drug than via oral delivery (Harvey, Chapter 35 In : Remington ' s
  • Endoscopic Administration can be used for drug delivery directly to an interior portion of the alimentary tract.
  • endoscopic retrograde cystopancreatography ERCP
  • ERCP endoscopic retrograde cystopancreatography
  • Rectal Administration Drugs administered by the oral route can often be alternatively administered by the lower enteral route, i.e., through the anal portal into the rectum or lower intestine.
  • Rectal suppositories, retention enemas or rectal catheters can be used for this purpose and may be preferred when patient • compliance might otherwise be difficult to achieve (e.g., in pediatric and geriatric applications, or when the patient is vomiting or unconscious) . Rectal administration may result in more prompt and higher blood levels than the oral route, but the converse may be true as well (Harvey, Chapter 35 In : Remington ' s Pharmaceutical Sciences, 18th Ed., Gennaro, ed., Mack Publishing Co., Easton, PA, 1990, page 711) . Because about 50% of the drug that is absorbed from the rectum will bypass the liver, administration by this route significantly reduces the potential for first- pass metabolism (Benet et al . , Chapter 1 In : Goodman & Gilman ' s The Pharmacological Basis of Therapeutics, 9th Ed., Hardman et al . , eds., McGraw-Hill, New York, NY, 1996) .
  • Oral Administration The preferred method of administration is oral delivery, which is typically the most convenient route for access to the systemic circulation. Absorption from the alimentary canal is governed by factors that are generally applicable, e.g., surface area for absorption, blood flow to the site of absorption, the physical state of the drug and its concentration at the site of absorption (Benet et al . , Chapter 1 In : Goodman & Gilman ' s The Pharmacological Basis of Therapeutics, 9th Ed., Hardman et al . , eds., McGraw- Hill, New York, NY, 1996, pages 5-7) .
  • factors that are generally applicable e.g., surface area for absorption, blood flow to the site of absorption, the physical state of the drug and its concentration at the site of absorption (Benet et al . , Chapter 1 In : Goodman & Gilman ' s The Pharmacological Basis of Therapeutics, 9th Ed., Hardman et al . , eds., McG
  • a significant factor which may limit the oral bioavailability of a drug is the degree of "first pass effects.” For example, some substances have such a rapid hepatic uptake that only a fraction of the material absorbed enters the peripheral blood (Van Berge-Henegouwen et al . , Gastroenterology, 1977, 73:300) .
  • the compositions and methods of the invention circumvent, at least partially, such first pass effects by providing improved uptake of nucleic acids and thereby, e.g., causing the hepatic uptake system to become saturated and allowing a significant portion of the nucleic acid so administered to reach the peripheral circulation. Additionally or alternatively, the hepatic uptake system is saturated with one or more inactive carrier compounds prior to administration of the active nucleic acid.
  • 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-l, 1-dioxide in acetonitrile for the stepwise thiation of the phosphite linkages.
  • the 2 ' -fluoro-phosphorothioate oligonucleotides of the invention are 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.
  • 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 are prepared essentially as described in U.S. Patents Nos. 5,539,082 and 5,539,083, both of which (1) issued July 23, 1996, and (2) are assigned to the same assignee as the instant application.
  • Example 2 Assays for Oligonucleotide-Mediated Inhibition of JNK mRNA Expression in Human Tumor Cells
  • human lung carcinoma cell line A549 American Type Culture Collection, Rockville, MD No. ATCC CCL-185 cells or other cell lines as indicated in the Examples, were grown and treated with oligonucleotides or control solutions as detailed below. After harvesting, cellular extracts were prepared and examined for specific JNK mRNA levels or JNK protein levels (i.e., Northern or Western assays, respectively) .
  • the blots were stripped and reprobed with a 32 P-labeled glyceraldehyde 3-phosphate dehydrogenase (G3PDH) probe (Clontech Laboratories, Inc., Palo Alto, CA) in order to confirm equal loading of RNA and to allow the levels of JNK transcripts to be normalized with regard to the G3PDH signals.
  • G3PDH glyceraldehyde 3-phosphate dehydrogenase
  • DMEM media
  • DOPE dioleoyl phosphatidylethanolamine
  • the oligonucleotides were added from a 10 ⁇ M stock solution to a final concentration of 400 nM, and the two solutions were mixed by swirling the flasks.
  • As a control cells were treated with LIPOFECTINTM without oligonucleotide under the same conditions and for the same times as the oligonucleotide-treated samples. After 4 hours at 37°C, the medium was replaced with fresh DMEM containing 10% serum. The cells were allowed to recover for 18 hours.
  • the primary antibodies were detected by means well known in the art (see, e.g., Short Protocols in Molecular Biology, 2nd Ed., Ausubel et al . , eds., John Wiley & Sons, New York, 1992, pages 10-33 to 10-35; and Chapter 18 In : Molecular Cloning: A Labora tory Manual , 2nd Ed., Sambrook et al . , eds., pages 18.1-18.75 and 18.86-18.88) and quantitated using a PHOSPHORIMAGERTM essentially according to the manufacturer's instructions (Molecular Dynamics, Sunnyvale, CA) . Levels of JNK proteins can also be quantitated by measuring the level of their corresponding kinase activity.
  • Such kinase assays can be done in gels in si tu (Hibi et al . , Genes & Dev. , 1993, 7, 2135) or after immunoprecipitation from cellular extracts (Derijard et al . , Cell , 1994, 76, 1025).
  • Substrates and/or kits for such assays are commercially available from, for example, Upstate Biotechnology, Inc. (Lake Placid, NY) , New England Biolabs, Inc., (Beverly, MA) and Calbiochem-Novabiochem Biosciences, Inc., (La Jolla, CA) .
  • Example 3 Oligonucleotide-Mediated Inhibition of JNKl Expression
  • Table 1 lists the nucleotide sequences of a set of oligonucleotides designed to specifically hybridize to JNKl mRNAs and their corresponding ISIS and SEQ ID numbers. The nucleotide coordinates of the target gene, JNKl , and gene target regions are also included. The nucleotide co-ordinates are derived from GenBank accession No. L26318, locus name "HUMJNK1" (see also Figure 1(A) of Derijard et al . , Cell , 1994, 76, 1025) .
  • gene target regions are as follows: 5'-UTR, 5' untranslated region; tIR, translation initiation region; ORF, open reading frame; 3'-UTR, 3' untranslated region.
  • the nucleotides of the oligonucleotides whose sequences are presented in Table 1 are connected by phosphorothioate linkages and are unmodified at the 2' position (i.e., 2 ' -deoxy) . It should be noted that the oligonucleotide target co-ordinate positions and gene target regions may vary within RNAs encoding related isoforms of JNKl (see subsection G, below) .
  • ISIS Nos. 12548 SEQ ID NO: 17
  • 12551 SEQ ID NO: 20
  • ISIS 12548 hybridizes to bases 498-517 of GenBank accession No.
  • L27129, locus name "RATSAPKD, " and ISIS 12551 hybridizes to bases 803-822 of the same sequence.
  • These oligonucleotides are thus preferred embodiments of the invention for investigating the role of the p54 ⁇ protein kinase in rat in vi tro, i . e . , in cultured cells or tissues derived from whole animals, or in vivo.
  • JNKl-specific probes In initial screenings of a set of oligonucleotides derived from the JNKl sequence (Table 2) for biological activity, a cDNA clone of JNKl (Derijard et al . , Cell , 1994, 76, 1025) was radiolabeled and used as a JNKl-specific probe in Northern blots. Alternatively, however, one or more of the oligonucleotides of Table 1 is detectably labeled and used as a JNKl- specific probe.
  • Oligonucleotides are thus preferred embodiments of the invention for modulating JNKl expression.
  • Oligonucleotides showing levels of inhibition of from > about 80% to about 100% of JNKl mRNAs in this assay include ISIS Nos. 11982, 12539, 12548, 12554 and 12464 (SEQ ID NOS: 5, 16, 17 and 23, respectively) . These oligonucleotides are thus more preferred embodiments of the invention for modulating JNKl expression.
  • JNKl oligonucleotides The results for JNKl-specific oligonucleotides (Table 2) indicate that one of the most active phosphorothioate oligonucleotides for modulating JNKl expression is ISIS 12539 (SEQ ID NO: 16) . As detailed in Table 4, additional oligonucleotides based on this oligonucleotide were designed to confirm and extend the findings described above.
  • Oligonucleotides ISIS Nos. 14320 (SEQ ID NO: 27) and 14321 (SEQ ID NO: 28) are 2 ' -deoxy-phosphorothioate sense strand and scrambled controls for ISIS 12539 (SEQ ID NO: 16), respectively.
  • ISIS Nos. 15346 and 15347 are "gapmers” corresponding to ISIS 12539; both have 2 ' -methoxyethoxy "wings” (having phosphorothioate linkages in the case of ISIS 15346 and phosphodiester linkages in the case of ISIS 15347) and a central 2 ' -deoxy "gap" designed to support RNaseH activity on the target mRNA molecule.
  • ISIS Nos. 15348 to 15350 are "wingmers” corresponding to ISIS 12539 and have a 5' or 3' 2 ' -methoxyethoxy RNaseH- refractory "wing” and a 3' or 5' (respectively) 2 ' -deoxy "wing” designed to support RNaseH activity on the target JNKl mRNA.
  • P 0, phosphodiester linkage
  • MOE methoxyethoxy-.
  • Dose- and sequence-dependent response to JNKl oligonucleotides In order to demonstrate a dose-dependent response to ISIS 12539 (SEQ ID NO: 16), different concentrations (i.e., 50, 100, 200 and 400 nM) of ISIS 12539 were tested for their effect on JNKl mRNA levels in A549 cells (Table 6) . In addition, two control oligonucleotides (ISIS 14320, SEQ ID NO: 27, sense control, and ISIS 14321, SEQ ID NO: 28, scrambled control; see also Table 4) were also applied to A549 cells in order to demonstrate the specificity of ISIS 12539. The results (Table 6) demonstrate that the response of A549 cells to ISIS 12539 is dependent on dose in an approximately linear fashion. In contrast, neither of the control oligonucleotides effect any consistent response on JNKl mRNA levels.
  • oligonucleotides showing a level of mRNA % inhibition from > about 70% to about 100% include ISIS Nos. 12539 (phosphorothioate linkages), 15346 and 15347 (“gapmers"), and 15348 and 15351 (5' "wingmers") (SEQ ID NO: 16) .
  • Oligonucleotides showing levels of mRNA inhibition of from > about 90% to about 100% of JNKl mRNAs in this assay include ISIS Nos. 12539, 15345 AND 15346 (SEQ ID NO: 16) .
  • the oligonucleotides tested showed approximately parallel levels of JNKl protein inhibition; ISIS Nos. 12539, 15346-15348 and 15351 effected levels of protein inhibition > about 40%, and ISIS Nos. 12539, 15346 and 15347 effected levels of protein inhibition > . about
  • oligonucleotides showing a level of mRNA % inhibition from > about 70% to about 100% include ISIS Nos. 12539 (phosphorothioate linkages) , 15346 and 15347 (“gapmers"), and 15348 (5' "wingmers”) (SEQ ID NO: 16). Oligonucleotides showing levels of mRNA inhibition of from > about 90% to about 100% of JNKl mRNAs at this point in the assay include ISIS Nos. 12539 and 15346 (SEQ ID NO: 16) . Overall, the oligonucleotides tested showed higher levels of JNKl protein inhibition at this point in the assay.
  • JNKl- ⁇ l GenBank accession No. L26318, locus name "HUMJNKl"
  • JNKl- ⁇ l GeneBank accession No. U34822, locus name "HSU34822”
  • JNKl- ⁇ l GenBank accession No.
  • JNKl- ⁇ 2 GenBank accession No. U35005, locus name "HSU35005" .
  • the four isoforms of JNKl which probably arise from alternative mRNA splicing, may each interact with different transcription factors or sets of transcription factors
  • oligonucleotides of the invention are specific for certain members or sets of these isoforms of JNKl.
  • nucleotides (nt) 631-665 of JNKl/JNKl- ⁇ l (Genbank accession No. L26318) and nt 625-659 of JNKl- ⁇ 2 (Genbank accession No. U34822) have the sequence shown below as SEQ ID NO: 63
  • nt 631-665 of JNKl- ⁇ l GenBank accession No. U35004
  • nt 626-660 of JNKl- ⁇ 2 GenBank accession No. U35005
  • SEQ ID NO: 64 For purposes of illustration, SEQ ID NOS: 63 and 64 are shown aligned with each other (vertical marks, "I,” indicate bases that are identical in both sequences) :
  • antisense oligonucleotides derived from the reverse complement of SEQ ID NO: 63 can be used to modulate the expression of JNK1/JNK1- ⁇ l and JNKl- ⁇ 2 without significantly effecting the expression of JNKl- ⁇ l and JNKl- ⁇ 2.
  • antisense oligonucleotides derived from the reverse complement of SEQ ID NO: 64 can be selected and used to modulate the expression of JNKl- ⁇ l and JNKl- ⁇ 2 without significantly effecting the expression of JNKl/JNKl- ⁇ l and JNKl- ⁇ 2.
  • SEQ ID NO: 66 see below
  • an oligonucleotide having a sequence derived from SEQ ID NO: 65 but not to SEQ ID NO: 66 is specifically hybridizable to mRNAs encoding JNKl/JNKl- ⁇ l and JNKl- ⁇ 2 but not to those encoding JNKl- ⁇ l and JNKl- ⁇ 2:
  • antisense oligonucleotides derived from the reverse complement of SEQ ID NO: 67 are specifically hybridizable to mRNAs encoding, and may be selected and used to modulate the expression of, JNKl/JNKl- ⁇ l and JNKl- ⁇ 2 without significantly effecting the expression of JNKl- ⁇ l and JNKl- ⁇ 2.
  • antisense oligonucleotides derived from the reverse complement of SEQ ID NO: 68 are specifically hybridizable to mRNAs encoding, and may be selected and used to modulate the expression of, can be selected and used to modulate the expression of JNKl- ⁇ l and JNKl- ⁇ 2 without significantly effecting the expression of JNKl/JNKl- ⁇ l and JNKl- ⁇ 2 :
  • JNKl/JNKl- ⁇ l shares identity with JNKl- ⁇ l; similarly, JNKl- ⁇ 2 and JNKl- ⁇ 2 have identical carboxy terminal portions.
  • the substantial differences in the amino acid sequences of these isoforms (5 amino acids in JNKl/JNKl- ⁇ l and JNKl- ⁇ l are replaced with 48 amino acids in JNKl- ⁇ 2 and JNKl- ⁇ 2) result from a slight difference in nucleotide sequence that shifts the reading frame.
  • nt 1144-1175 of JNKl/JNKl- ⁇ l (Genbank accession No. L26318) and JNKl- ⁇ l (Genbank accession No. U35004) have the sequence shown below as SEQ ID NO: 71
  • nt 1138-1164 of JNKl- ⁇ 2 (GenBank accession No. U34822)
  • nt 1139-1165 of JNKl- ⁇ 2 (GenBank accession No. U35005) have the sequence shown below as SEQ ID NO: 72.
  • SEQ ID NOS: 71 and 72 are shown aligned with each other (dashes, "-,” indicate bases that are absent in the indicated sequence, and emboldened bases indicate the stop codon for the JNKl/JNKl- ⁇ l and JNKl- ⁇ l ORFs) :
  • antisense oligonucleotides derived from the reverse complement of SEQ ID NO: 71 are specifically hybridizable to mRNAs encoding, and may be selected and used to modulate the expression of, JNKl/JNKl- ⁇ l and JNKl- ⁇ l without significantly effecting the expression of JNK1- ⁇ 2 and JNKl- ⁇ 2.
  • antisense oligonucleotides derived from the reverse complement of SEQ ID NO: 72 are specifically hybridizable to mRNAs encoding, and may be selected and used to modulate the expression of, JNKl- ⁇ 2 and JNKl- ⁇ 2 without significantly effecting the expression of JNKl/JNKl- ⁇ l and JNKl- ⁇ l: 5 ' -GATCACTGCTGCACCTGTGCTAAAGGAGAGGG SEQ ID NO: 73
  • such isoform-specific oligonucleotides such as are described above are methoxyethoxy "gapmers” or “wingmers” in which the RNase H- sensitive "gap” or “wing” is positioned so as to overlap a region of nonidentity in the above antisense sequences, i.e., SEQ ID NOS: 65, 66, 69, 70, 73 and 74.
  • Example 4 Oligonucleotide-Mediated Inhibition of JNK2 Expression
  • Table 8 lists the nucleotide sequences of oligonucleotides designed to specifically hybridize to JNK2 mRNAs and the corresponding ISIS and SEQ ID numbers thereof.
  • the target gene nucleotide co-ordinates and gene target region are also included.
  • the nucleotide co-ordinates are derived from GenBank accession No. L31951, locus name "HUMJNK2" (see also Figure 1(A) of Sluss et al . , Mol . Cel . Biol . , 1994, 1 4 , 8376, and Kallunki et a l .
  • gene target regions are as follows: 5 ' -UTR, 5' untranslated region; tIR, translation initiation region; ORF, open reading frame; 3' -UTR, 3' untranslated region.
  • the nucleotides of the oligonucleotides whose sequences are presented in Table 8 are connected by phosphorothioate linkages and are unmodified at the 2' position (i.e., 2-deoxy) . It should be noted that the oligonucleotide target co-ordinate positions and gene target regions may vary within mRNAs encoding related isoforms of JNK2 (see subsection G, below) .
  • ISIS No. 12562 hybridizes to the ORF of mRNAs from Ra ttus norvegicus that encode a stress-activated protein kinase named "p54 ⁇ 2" (Kyriakis et al . , Na ture, 1994, 369, 156). Specifically, ISIS 12562 (SEQ ID NO: 33) hybridizes to bases 649-668 of GenBank accession No.
  • This oligonucleotide is thus a preferred embodiment of the invention for investigating the role of the p54 ⁇ 2 protein kinase in rat in vi tro, i . e . , in cultured cells or tissues derived from whole animals, or in vivo .
  • JNK2-specific probes In initial screenings of a set of oligonucleotides derived from the JNK2 sequence (Table 9) for biological activity, a cDNA clone of JNK2 (Kallunki et al . , Genes & Development , 1994, 8, 2996) was radiolabeled and used as a JNK2-specific probe in Northern blots. Alternatively, however, one or more of the oligonucleotides of Table 8 is detectably labeled and used as a JNK2-specific probe. C.
  • Oligonucleotides showing levels of inhibition of from > about 80% to about 100% of JNK2 mRNAs in this assay include ISIS Nos. 12558, 12560, 12565, 12567, 12568 and 12569 (SEQ ID NOS: 29, 31, 36, 38, 39 and 40, respectively) . These oligonucleotides are thus more preferred embodiments of the invention for modulating JNK2 expression.
  • JNK2 oligonucleotides The results for JNK2-specific oligonucleotides (Table 9) indicate that one of the most active phosphorothioate oligonucleotides for modulating JNK2 expression is ISIS 12560 (SEQ ID NO: 31) . As detailed in Table 11, additional oligonucleotides based on this oligonucleotide were designed to confirm and extend the findings described above. Oligonucleotides ISIS Nos. 14318 (SEQ ID NO: 42) and 14319 (SEQ ID NO: 43) are 2 ' -deoxy-phosphorothioate sense strand and scrambled controls for ISIS 12560 (SEQ ID NO: 31), respectively.
  • ISIS Nos. 15353 and 15354 are "gapmers" corresponding to ISIS 12560; both have 2 ' -methoxyethoxy "wings” (having phosphorothioate linkages in the case of ISIS 15353 and phosphodiester linkages in the case of ISIS 15354) and a central 2 ' -deoxy "gap” designed to support RNaseH activity on the target mRNA molecule.
  • 15355 to 15358 are "wingmers” corresponding to ISIS 12560 and have a 5' or 3 ' 2 ' -methoxyethoxy RNaseH- refractory "wing” and a 3' or 5 ' (respectively) 2-deoxy "wing” designed to support RNaseH activity on the target JNK2 mRNA.
  • Dose- and sequence-dependent response to JNK2 oligonucleotides In order to demonstrate a dose-dependent response to ISIS 12560 (SEQ ID NO: 31), different concentrations (i.e., 50, 100, 200 and 400 nM) of ISIS 12560 were tested for their effect on JNK2 mRNA levels in A549 cells (Table 13) . In addition, two control oligonucleotides (ISIS 14318, SEQ ID NO: 42, sense control, and ISIS 14319, SEQ ID NO: 43, scrambled control; see also Table 11) were also applied to A549 cells in order to demonstrate the specificity of ISIS 12560. The results (Table 12) demonstrate that the response of A549 cells to ISIS 12539 is dependent on dose in an approximately linear fashion. In contrast, neither of the control oligonucleotides effect any consistent response on JNK2 mRNA levels.
  • Western Assays In order to assess the effect of oligonucleotides targeted to JNK2 mRNAs on JNK2 protein levels, Western assays are performed essentially as described above in Examples 2 and 3.
  • a primary antibody that specifically binds to JNK2 is purchased from, for example, Santa Cruz Biotechnology, Inc., Santa Cruz, CA; Upstate Biotechnology, Inc., Lake Placid, NY; StressGen Biotechnologies, Inc., Victoria, BC, Canada; or Research Diagnostics, Inc., Flanders, NJ.
  • JNK2- ⁇ 2 (GenBank accession No. L31951, locus name "HUMJNK2”), which encodes a polypeptide having an amino acid sequence identical to that of JNK2, the additional isoforms include JNK2- ⁇ l (GenBank accession No. U34821, locus name "HSU34821”) , JNK2- ⁇ l (GenBank accession No. U35002, locus name "HSU35002”) and JNK2- ⁇ 2 (GenBank accession No. U35003, locus name "HSU35003”) .
  • the four isoforms of JNK2, which probably arise from alternative mRNA splicing, may each interact with different transcription factors or sets of transcription factors (Gupta et al .
  • the oligonucleotides of the invention are specific for certain members or sets of these isoforms of JNK2.
  • nucleotides (nt) 689-748 of JNK2/JNK2- ⁇ 2 GenBank accession No. L31951)
  • nt 675-734 of JNK2- ⁇ l GenBank accession No.
  • SEQ ID NO: 75 nt 653-712 of JNK2- ⁇ l (GenBank accession No. U35002) and nt 665-724 of JNK2- ⁇ 2 (GenBank accession No. U35003) have the sequence shown below as SEQ ID NO: 76.
  • SEQ ID NOS: 75 and 76 are shown aligned with each other (vertical marks, "
  • antisense oligonucleotides derived from the reverse complement of SEQ ID NO: 75 are specifically hybridizable to, and may be selected and used to modulate the expression of, JNK2/JNK2- ⁇ 2 and JNK2- ⁇ l without significantly effecting the expression of JNKl- ⁇ l and JNKl- ⁇ 2.
  • antisense oligonucleotides derived from the reverse complement of SEQ ID NO: 76 are specifically hybridizable to, and may be selected and used to modulate the expression of, JNK2- ⁇ l and JNK2- ⁇ 2 without significantly effecting the expression of JNK2/JNK2- ⁇ 2 and JNK2- ⁇ l.
  • SEQ ID NO: 78 an oligonucleotide having a sequence derived from SEQ ID NO: 77 but not from SEQ ID NO : 78 is specifically hybridi zable to , mRNAs encoding JNKl/JNKl- ⁇ l and JNKl- ⁇ 2 but not to those encoding JNK2- ⁇ l and JNK2- ⁇ 2 :
  • JNK2/JNK2- ⁇ 2 shares identity with JNKl- ⁇ 2; similarly, JNK2- ⁇ l and JNK2- ⁇ l have identical carboxy terminal portions.
  • the substantial differences in the amino acid sequences of these isoforms (5 amino acids in JNK2- ⁇ 2 and JNK2- ⁇ 2 are replaced with 47 amino acids in JNK2/JNK2- ⁇ 2 and JNK2- ⁇ 2) result from a slight difference in nucleotide sequence that shifts the reading frame.
  • nt 1164-1198 of JNK2- ⁇ l (GenBank accession No.
  • nt 1142-1176 of JNK2- ⁇ l (GenBank accession No. U35002) have the sequence shown below as SEQ ID NO: 79, whereas, in the ORFs of mRNAs encoding JNK2/JNK2- ⁇ 2 and JNK2- ⁇ 2, nt 1178-1207 of JNK2/JNK2- ⁇ 2 (GenBank accession No. L31951) and nt 1154-1183 of JNK2- ⁇ 2 (GenBank accession No. U35003) have the sequence shown below as SEQ ID NO: 80.
  • SEQ ID NOS: 79 and 80 are shown aligned with each other (dashes, "-,” indicate bases that are absent in the indicated sequence, and emboldened bases indicate the stop codon for the JNK2- ⁇ l and JNK2- ⁇ l ORFs) : 5 ' -GATCAGCCTTCAGCACAGATGCAGCAGTAAGTAGC SEQ ID NO: 79
  • antisense oligonucleotides derived from the reverse complement of SEQ ID NO: 79 are specifically hybridizable to, and may be selected and used to modulate the expression of, mRNAs encoding JNK2- ⁇ l and JNK2- ⁇ l without significantly effecting the expression of JNK2/JNK2- ⁇ 2 and JNK2- ⁇ 2.
  • antisense oligonucleotides derived from the reverse complement of SEQ ID NO: 80 are specifically hybridizable to, and may be selected and used to modulate the expression of, mRNAs encoding JNK2/JNK2- ⁇ 2 and JNK2- ⁇ 2 without significantly effecting the expression of JNK2- ⁇ l and JNK2- ⁇ l.
  • ISIS 12564 (SEQ ID NO: 35) corresponds to SEQ ID NO: 82 but not to SEQ ID NO: 81, and is thus specifically hybridizable to, and may be used to modulate the expression of, mRNAs encoding JNK2/JNK2- ⁇ 2 and JNK2- ⁇ 2 but not those encoding JNK2- ⁇ l and JNK2- ⁇ l:
  • such isoform-specific oligonucleotides such as are described above are methoxyethoxy "gapmers” or “wingmers” in which the RNase H- sensitive "gap” or “wing” is positioned so as to overlap a region of nonidentity in the above antisense sequences, i.e., SEQ ID NOS: 77, 78, 81 and 82.
  • JNK3 oligonucleotide sequences Table 14 lists the nucleotide sequences of oligonucleotides designed to specifically hybridize to JNK3 mRNAs and the corresponding ISIS and SEQ ID numbers thereof.
  • the target gene nucleotide co-ordinates and gene target region are also included.
  • the nucleotide co-ordinates are derived from GenBank accession No. U07620, locus name "HSU07620” see also Figure 4(A) of Mohit et al . , Neuron, 1994, 14, 67).
  • gene target regions are as follows: 5' -UTR, 5' untranslated region; tIR, translation initiation region; ORF, open reading frame; 3 ' -UTR, 3' untranslated region. It should be noted that the oligonucleotide target co-ordinate positions and gene target regions may vary within mRNAs encoding related isoforms of JNK3 (see subsection D, below) .
  • nucleotides of the oligonucleotides whose sequences are presented in Table 14 are connected by phosphorothioate linkages and are "gapmers.” Specifically, the six nucleotides of the 3' and 5' termini are 2'- methoxyethoxy- modified and are shown emboldened in Table 14, whereas the central eight nucleotides are unmodified at the 2' position (i.e., 2-deoxy) .
  • the full oligonucleotide sequences of ISIS Nos. 16692, 16693, 16703, 16704, 16705, 16707, and 16708 specifically hybridize to mRNAs from Ra ttus norvegicus that encode a stress- activated protein kinase named "p54 ⁇ " (Kyriakis et al . , Na ture, 1994, 369, 156; GenBank accession No.
  • the full oligonucleotide sequences of 16692, 16693, 16695, 16703, 16704, 16705, 16707 and 16708 (SEQ ID NOS: 46, 47, 49, 56, 57, 58, 60 and 61, respectively) specifically hybridize to mRNAs from Mus musculus that encode a mitogen activated protein (MAP) kinase stress activated protein named the "p459 3F12 SAP kinase” (Martin et al . , Brain Res . Mol . Brain Res . , 1996, 35, 47; GenBank accession No. L35236, locus name "MUSMAPK”) .
  • MAP mitogen activated protein
  • oligonucleotides are thus preferred embodiments of the invention for investigating the role of the p54 ⁇ and p459 3F12 SAP protein kinases in rat or mouse, respectively, in vi tro, i . e . , in cultured cells or tissues derived from whole animals or in vivo .
  • the target gene nucleotide co-ordinates and gene target regions for these oligonucleotides, as defined for these GenBank entries, are detailed in Table 15.
  • JNK3-specific probes In initial screenings of a set of oligonucleotides derived from the JNK3 sequence for biological activity, a cDNA clone of JNK3 (Derijard et al . , Cell , 1994, 76, 1025) was radiolabeled and used as a JNK3- specific probe in Northern blots. Alternatively, however, one or more of the oligonucleotides of Table 14 is detectably labeled and used as a JNK3-specific probe.
  • Emboldened residues are 2 ' -methoxyethoxy- modified .
  • JNK3- ⁇ l was initially cloned and named "p49 3F12 kinase" by Mohit et al . ⁇ Neuron, 1995, 14, 67) . Subsequently, two cDNAs encoding related isoforms of JNK3 were cloned and their nucleotide sequences determined (Gupta et al . , EMBO Journal , 1996, 15, 2760) . The isoforms are named JNK3- ⁇ l (GenBank accession No.
  • JNK3- ⁇ 2 GenBank accession No. U34819, locus name "HSU34819" herein.
  • the two isoforms of JNK3, which probably arise from alternative mRNA splicing, may each interact with different transcription factors or sets of transcription factors (Gupta et al . , EMBO Journal, 1996, 15, 2760).
  • certain oligonucleotides of the invention are specific for each of these isoforms of JNK3.
  • JNK3- ⁇ l and JNK- ⁇ 2 differ at their carboxyl terminal portions.
  • the substantial differences in the amino acid sequences of these isoforms (5 amino acids in JNK3- ⁇ l are replaced with 47 amino acids in JNK3- ⁇ 2) result from a slight difference in nucleotide sequence that shifts the reading frame.
  • nucleotides (nt) 1325-1362 of JNK3- ⁇ l (GenBank accession No. U34820) have the sequence shown below as SEQ ID NO: 83
  • nt 1301-1333 of JNK3- ⁇ 2 GenBank accession No.
  • SEQ ID NO: 84 have the sequence shown below as SEQ ID NO: 84.
  • SEQ ID NOS: 83 and 202 are shown aligned with each other (vertical marks, "I,” indicate bases that are identical in both sequences; dashes, "-,” indicate bases that are absent in the indicated sequence; and emboldened bases indicate the stop codon for the JNK3- ⁇ l ORF) : 5 ' -GGACAGCCTTCTCCTTCAGCACAGGTGCAGCAGTGAAC SEQ ID NO: 83
  • antisense oligonucleotides derived from the reverse complement of SEQ ID NO: 83 are specifically hybridizable to mRNAs encoding, and may be selected and used to modulate the expression of JNK3- ⁇ l without significantly effecting the expression of JNK3- ⁇ 2.
  • antisense oligonucleotides derived from the reverse complement of SEQ ID NO: 84 are specifically hybridizable to mRNAs encoding, and may be selected and used to modulate the expression of JNK3- ⁇ 2 without significantly effecting the expression of JNK3-0.1:
  • such isoform-specific oligonucleotides such as are described above are methoxyethoxy "gapmers” or “wingmers” in which the RNase H- sensitive "gap” or “wing” is positioned so as to overlap a region of nonidentity in the above antisense sequences, i.e., SEQ ID NOS: 85 and 86.
  • E. Activities of JNK3 oligonucleotides The JNK3- specific phosphorothioate, 2 ' -methoxyethoxy "gapmer” oligonucleotides (Table 14) were screened for their ability to affect JNK3 mRNA levels in SH-SY5Y cells (Biedler et al .
  • SH-SY5Y cells express a variety of mitogen-activated protein kinases (MAPKs; see, e.g., Cheng et al . , J. Biol . Chem . , 1998, 273, 14560).
  • MAPKs mitogen-activated protein kinases
  • Cells were grown in DMEM essentially as previously described (e.g., Singleton et al . , J. Biol . Chem . , 1996, 271, 31791; Jalava et al . , Cancer Res . , 1990, 50, 3422) and treated with oligonucleotides at a concentration of 200 nM as described in Example 2. Control cultures were treated with an aliquot of LIPOFECTINTM that contained no oligonucleotide .
  • Oligonucleotides showing levels of inhibition from > . about 45% to about 100% of JNK3 mRNA levels include ISIS Nos. 16692, 16693, 16694, 16695, 16696, 16697, 16702, 16703, 16704, 16705 and 16706 (SEQ ID NOS:46, 47, 48, 49, 50, 51, 55, 56, 57, 58 and 59, respectively) . These oligonucleotides are preferred embodiments of the invention for modulating JNK3 expression.
  • Oligonucleotides showing levels of inhibition of from > about 60% to about 100% of JNK3 mRNAs in this assay, wherein "about” indicates ⁇ 5%, include ISIS Nos. 16693, 16694, 16695, 16702, 16703, 16704 and 16705 (SEQ ID NOS:47, 48, 49, 55, 56, 57 and 58, respectively). These oligonucleotides are thus more preferred embodiments of the invention for modulating JNK3 expression.
  • Example 6 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).
  • MMPs matrix metalloproteinases
  • MMP-9 matrix metalloproteinase-9
  • NHEKs normal human epidermal keratinocytes
  • TPA (12-O-tetradecanoylphorbol 13-acetate)
  • ISIS 10582 an oligonucleotide targeted to c- jun, was evaluated for its ability to modulate MMP- 9 expression (see pending application Serial No. 08/837,201, filed April 14, 1997, attorney docket No. ISPH-0209. The results (Table 16) demonstrate that ISIS 10582 is able to completely inhibit the expression of MMP- 9 after induction with TPA.
  • Example 7 Treatment of Human Tumors in Mice with
  • Tumor size was measured and tumor volume was calculated on days 12, 19, 26 and 33 following tumor cell inoculation.
  • the antisense compounds of the invention are also tested for their ability to slow or eliminate the growth of xenografts resulting from, for example, human cervical epithelial carcinoma cells (HeLa cell line, ATCC No. ATCC CCL-2), human lung carcinoma cells (cell line A549, ATCC No. ATCC CCL-185), human adenocarcinoma cells (cell line SW480, ATCC No. ATCC CCL-228), human bladder carcinoma cells (cell line T24, ATCC No. HTB-4), human pancreatic - Ill
  • carcinoma cells cell line MIA PaCa, ATCC No. CRL-1420
  • human small cell carcinoma cells cell line NCI-H69, ATCC HTB-119 .
  • Xenografts resulting from these and other cell lines are established using essentially the same techniques as were used for the experiments using MDA-MB 231 cells.
  • oligonucleotides targeted to the genes encoding JNKl, JNK2 and JNK3 of Ra ttus norvegicus were prepared. These oligonucleotides are 2 ' -methoxyethoxy, phosphodiester / 2 '-hydroxyl, phosphorothioate / 2 ' -methoxyethoxy, phosphodiester "gapmers" in which every cytosine residue is 5-methylcytosine (m5c) . These antisense compounds were synthesized according to the methods of the disclosure. Certain of these oligonucleotides are additionally specifically hybridizable to JNK genes from other species as indicated herein.
  • oligonucleotides described in this Example were tested for their ability to modulate rat JNK mRNA levels essentially according to the methods described in the preceding Examples, with the exceptions that the cell line used was rat A10 aortic smooth muscle cells (ATCC No. ATCC CRL-1476) and the probes used were specific for rat JNKl, JNK2 or JNK3 (see infra ) .
  • A10 cells were grown and treated with oligonucleotides essentially as described by Cioffi et al . ⁇ Mol . Pharmacol . , 1997, 51 , 383) .
  • JNKl Table 19 describes the sequences and structures of a set of oligonucleotides, ISIS Nos. 21857 to 21870 (SEQ ID NOS: 111 to 124, respectively) that were designed to be specifically hybridizable to nucleic acids from Ra ttus norvegicus that encode a stress-activated protein kinase named "p54 ⁇ ” or "SAPKy” that is homologous to the human protein JNKl (Kyriakis et al . , Na ture, 1994, 369, 156; GenBank accession No. L27129, locus name "RATSAPKD”) .
  • emboldened residues are 2'- methoxyethoxy-residues (others are 2 '-deoxy-); "C” residues are 2 ' -methoxyethoxy-5-methyl-cytosines and “C” residues are 5-methyl-cytosines; "o” indicates a phosphodiester linkage; and "s” indicates a phosphorothioate linkage.
  • the target gene co-ordinates are from GenBank Accession No. L27129, locus name "RATSAPKD.” TABLE 19: Nucleotide Sequences of Rat JNKl Oligonucleotides
  • Oligonucleotides showing levels of inhibition from > . about 75% to about 100% of rat JNKl mRNA levels include ISIS Nos. 21857 to 21870 (SEQ ID NOS: 111 to 124, respectively). These oligonucleotides are preferred embodiments of the invention for modulating rat JNKl expression. Oligonucleotides showing levels of inhibition of from > . about 90% to about 100% of rat JNKl mRNAs in this assay include ISIS Nos. 21858, 21859, 21860, 21861, 21862, 21865,
  • Pan JNK oligonucleotides demonstrated a capacity to modulate both JNKl and JNK2.
  • Such oligonucleotides are referred to herein as "Pan JNK” antisense compounds because the term "Pan” is used in immunological literature to refer to an antibody that recognizes, e.g., all isoforms of a protein or subtypes of a cell type.
  • Pan JNK oligonucleotides are discussed in more detail infra .
  • ISIS 21859 (SEQ ID NO: 113) is complementary to bases 4 to 23 of cDNAs encoding human JNKl ⁇ l and JNKl ⁇ l (i.e., GenBank accession Nos. L26318 and U35004, respectively).
  • ISIS 21862 (SEQ ID NO:116) is complementary to bases 294 to 313 of the human JNKl ⁇ l and JNKl ⁇ l cDNAs (GenBank accession Nos.
  • ISIS 18257 is specifically hybridizable to nucleic acids encoding p54 ⁇ 2 (i.e., GenBank accession No. L27112, locus name "RATSAPKB”) .
  • emboldened residues are 2 ' -methoxyethoxy-residues (others are 2 '-deoxy-);
  • C residues are 2 ' -methoxyethoxy-5-methyl-cytosines and "C” residues are 5-methyl-cytosines; "o” indicates a phosphodiester linkage; and "s” indicates a phosphorothioate linkage.
  • the target gene co-ordinates are from GenBank Accession No. L27112, locus name "RATSAPKB.”
  • Oligonucleotides showing levels of inhibition from > . about 60% to about 100% of rat JNK2 mRNA levels include ISIS Nos. 18254, 18255, 18257, 18258, 18259, 18260 and 18264 (SEQ ID NOS:125, 126, 128, 129, 130, 131 and 135, respectively). These oligonucleotides are preferred embodiments of the invention for modulating rat JNK2 expression. Oligonucleotides showing levels of inhibition of from > .
  • rat JNKl mRNAs in this assay include ISIS Nos. 18254, 18255, 18258 and 18259 (SEQ ID NOS:125, 126, 129 and 130, respectively). These oligonucleotides are thus more preferred embodiments of the invention for modulating rat JNK2 expression.
  • ISIS 18259 SEQ ID NO: 130 was chosen for use in further studies ⁇ infra ) .
  • C. Dose Response A dose response study was conducted using oligonucleotides targeted to rat JNKl (ISIS 21859; SEQ ID NO: 113) and JNK2 (ISIS 18259; SEQ ID NO: 130) and Northern assays.
  • JNK3 Table 24 describes the sequences and structures of a set of oligonucleotides, ISIS Nos. 21899 to 21912 (SEQ ID NOS: 142 to 155, respectively) that were designed to be specifically hybridizable to nucleic acids from Ra ttus norvegicus that encode a stress-activated protein kinase named "p54 ⁇ " that is homologous to the human protein JNK3 (Kyriakis et al . , Na ture, 1994, 369, 156; GenBank accession No. L27128, locus name "RATSAPKC”) .
  • emboldened residues are 2 ' -methoxyethoxy-residues (others are 2 '-deoxy-); "C” residues are 2 ' -methoxyethoxy- 5-methyl-cytosines and “C” residues are 5-methyl-cytosines; "o” indicates a phosphodiester linkage; and "s” indicates a phosphorothioate linkage.
  • the target gene co-ordinates are from GenBank Accession No. L27128, locus name "RATSAPKC.”
  • the oligonucleotides are tested for their ability to modulate rat JNK3 mRNA levels essentially according to the methods described in the preceding Examples.
  • oligonucleotides described in Table 24 are also specifically hybridizable with JNK3-encoding nucleic acids from humans and Mus musculus (mouse) .
  • Table 25 sets out these relationships. These oligonucleotides are tested for their ability to modulate mRNA levels of the human JNK genes according to the methods described in Example 5.
  • GenBank accession No. U34819 locus name "HSU34819” (see also Gupta et al . , EMBO Journal , 1996, 15, 2760).
  • Pan JNK Oligonucleotides Certain of the oligonucleotides of the invention are capable of modulating two or more JNK proteins and are referred to herein as "Pan JNK" oligonucleotides.
  • ISIS Nos. Nos. 21861 and 21867 demonstrated a capacity to modulate both JNKl and JNK2 (Table 20) .
  • Such oligonucleotides are useful when the concomitant modulation of several JNK proteins is desired.
  • Human Pan JNK oligonucleotides are described in Table 26.
  • oligonucleotides are designed to be complementary to sequences that are identically conserved in (i.e., SEQ ID NOS:156, 158, 159, 160 and 161), or which occur with no more than a one-base mismatch (SEQ ID NO: 157), in nucleic acids encoding human JNKl ⁇ l, JNKl ⁇ 2, JNK2 ⁇ l and JNK2 ⁇ 2.
  • SEQ ID NOS:156, 158, 159, 160 and 161 or which occur with no more than a one-base mismatch (SEQ ID NO: 157), in nucleic acids encoding human JNKl ⁇ l, JNKl ⁇ 2, JNK2 ⁇ l and JNK2 ⁇ 2.
  • SEQ ID NO: 157 one-base mismatch
  • hypoxanthine inosine (I) is capable of hydrogen bonding with a variety of nucleobases and thus serves as a "universal" base for hybridization purposes.
  • an oligonucleotide having a sequence that is a derivative of SEQ ID NO: 157 having one inosine substitution (TAGGAIATTCTTTCATGATC, SEQ ID NO: 162) is predicted to bind to nucleic acids encoding human JNKl ⁇ l, JNKl ⁇ 2, JNK2 ⁇ l and JNK2 ⁇ 2 with no mismatched bases.
  • an oligonucleotide having a sequence that is a derivative of SEQ ID NO: 161 having one inosine substitution is predicted to bind with no mismatched bases to nucleic acids encoding human JNK3 ⁇ l and JNK3 ⁇ 2 in addition to JNKl ⁇ l, JNKl ⁇ 2, JNK2 ⁇ l and JNK2 ⁇ 2.
  • Such oligonucleotides are evaluated for their ability to modulate JNKl and JNK2 mRNA levels in A549 cells, and JNK3 mRNA levels in SH-SY5Y cells, using the methods and assays described in Examples 3, 4 and 5.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Organic Chemistry (AREA)
  • Genetics & Genomics (AREA)
  • Engineering & Computer Science (AREA)
  • Molecular Biology (AREA)
  • Biochemistry (AREA)
  • Biotechnology (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • General Health & Medical Sciences (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • General Engineering & Computer Science (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Biomedical Technology (AREA)
  • Microbiology (AREA)
  • Immunology (AREA)
  • Pathology (AREA)
  • Analytical Chemistry (AREA)
  • Physics & Mathematics (AREA)
  • Biophysics (AREA)
  • Virology (AREA)
  • Hospice & Palliative Care (AREA)
  • Plant Pathology (AREA)
  • Oncology (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Peptides Or Proteins (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)

Abstract

Compositions and methods for the treatment and diagnosis of diseases or disorders amenable to treatment through modulation of expression of a gene encoding a Jun N-terminal kinase (JNK protein) are provided. Oligonucleotide are herein provided which are specifically hybridizable with nucleic acids encoding JNK1, JNK2 and JNK3, as well as other JNK proteins and specific isoforms thereof. Methods of treating animals suffering from diseases or disorders amenable to therapeutic intervention by modulating the expression of one or more JNK proteins with such oligonucleotide are also provided. Methods for the treatment and diagnosis of diseases or disorders associated with aberrant expression of one or more JNK proteins are also provided. The invention is thus directed to compositions for modulating, diagnostic methods for detecting, and therapeutic methods for inhibiting, the hyperproliferation of cells and formation, development and maintenance of tumors.

Description

ANTISENSE OLIGONUCLEOTIDE COMPOSITIONS AND METHODS FOR THE MODULATION OF JNK PROTEINS
INTRODUCTION
This application is a continuation-in-part of U.S. Application Serial No. 08/910,629 filed August 13, 1997.
FIELD OF THE INVENTION
The present invention provides compositions and methods for detecting and modulating levels of Jun N- terminal inases (JNK proteins) , enzymes which are encoded by JNK genes. In particular, the invention relates to antisense oligonucleotides specifically hybridizable with nucleic acids encoding JNK proteins. It has been found that antisense oligonucleotides can modulate the expression of these and other JNK proteins, kinases which were initially discovered due to their ability to catalyze the phosphorylation of the c-Jun subunit of transcription factor AP-1 and thereby increase AP-1 activity. Other transcription factors, such as ATF-2, are similarly activated by JNK proteins, and a variety of other cellular effectors may serve as substrates for JNK proteins (Gupta et al . , Science, 1995, 267, 389). In any event, transcription factor AP-1 has been implicated in abnormal cell proliferation, oncogenic transformation, and tumor formation, development and maintenance (Vogt, Chapter 15 In : The FOS and JUN Families of Transcription Factors,
Angel and Herrlich, Eds., CRC Press, Boca Raton, FL, 1994). Accordingly, it is believed that (1) JNK proteins are aberrantly expressed in some neoplasms and tumors with resultant increased AP-1 activity, and (2) even in abnormally proliferating cells in which a JNK gene is not aberrantly expressed, inhibition of JNK expression will result in decreased AP-1 activity and thus, inhibition of abnormal cell proliferation and tumor formation, development and maintenance. The invention is thus directed to diagnostic methods for detecting, and therapeutic methods for inhibiting, the hyperproliferation of cells and the formation, development and maintenance of tumors. Furthermore, this invention is directed to treatment of conditions associated with abnormal expression of JNK genes. This invention also relates to therapies, diagnostics, and research reagents for disease states or disorders which respond to modulation of the expression of JNK proteins. Inhibition of the 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 (Rahmsdorf, Chapter 13, and Rapp et al . , Chapter 16 In : The FOS and JUN Families of
Transcription Factors, Angel and Herrlich, Eds., CRC Press, Boca Raton, FL, 1994). 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, Biochim . 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 { The FOS and JUN Families of Transcription Factors, Angel and Herrlich, Eds., CRC Press, Boca Raton, FL, 1994; Bohmann et al . , Science, 1987, 238, 1386; Angel et al . , Na ture, 1988, 332, 166) . Two relatively well-characterized AP-1 subunits are c-Fos and c-Jun; these two proteins are products of the c- fos and c-j un proto-oncogenes, respectively. Repression of the activity of either c-fos or c-j un, or 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 kinases (MAPKs), 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). MAPKs (also referred to as extracellular signal-regulated protein kinases, or ERKs) are themselves activated by phosphorylation catalyzed by, e.g., receptor tyrosine kinases, G protein-coupled receptors, protein kinase C (PKC) , and the apparently MAPK-dedicated kinases MEK1 and MEK2. 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 (Seger et al . , FASEB J. , 1995, 9, 726; Cano et al . , Trends Biochem . Sci . , 1995, 20, 117). In a typical MAP kinase pathway, it is thought that a first MAP kinase, called a MEKK, phosphorylates and thereby activates a second MAP kinase, called a MEK, which, in turn, phosphorylates and activates a MAPK/ERK or JNK/SAPK enzyme ("SAPK" is an abbreviation for stress- activated protein kinase) . Finally, the activated MAPK/ERK or JNK/SAPK enzyme itself phosphorylates and activates a transcription factor (such as, e . g. , AP-1) or other substrates (Cano et al . , Trends Biochem . Sci . , 1995, 20, 117) . This canonical cascade can be simply represented as follows :
MEKK > MEK > MAPK/ERK > transcription factor or JNK/SAPK or other substrate (s)
One of the signal transduction pathways involves the MAP kinases Jun N-terminal kinase 1 (JNK1) and Jun N- terminal kinase 2 (JNK2) which are responsible for the phosphorylation of specific sites (Serine 63 and Serine 73) on the amino terminal portion of 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 . , Na ture, 1991, 354, 494). Besides JNK1 and JNK2, other JNK family members have been described, including JNK3 (Gupta et al . , EMBO J. , 1996, 15, 2760), initially named p493F12 kinase (Mohit et al . , Neuron, 1994, 14, 67). The term "JNK protein" as used herein shall mean a member of the JNK family of kinases, including but not limited to JNK1, JNK2 and JNK3, their isoforms (Gupta et al . , EMBO J. , 1996, 15, 2760) and other members of the JNK family of proteins whether they function as Jun N-terminal kinases per se (that is, phosphorylate Jun at a specific amino terminally located position) or not.
At least one human leukemia oncogene has been shown to enhance Jun N-terminal kinase function (Raitano et al . , Proc . Na tl . Acad . Sci . (USA) , 1995, 92, 11746). Modulation of the expression of one or more JNK proteins is desirable in order to interfere with hyperproliferation of cells and with the transcription of genes stimulated by AP-1 and other JNK protein phosphorylation substrates. Modulation of the expression of one or more other JNK proteins is also desirable in order to interfere with hyperproliferation of cells resulting from abnormalities in specific signal transduction pathways. To date, there are no known therapeutic agents which effectively inhibit gene expression of one or more JNK proteins. Consequently, there remains a long-felt need for improved compositions and methods for modulating the expression of specific JNK proteins .
Moreover, cellular hyperproliferation in an animal can have several outcomes. Internal processes may eliminate hyperproliferative cells 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 hyperproliferative 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., Garland Publishing, Inc., New York, 1983) . Using antisense oligonucleotides, it has surprisingly been discovered that several genes encoding enzymes required for metastasis are positively regulated by AP-1, which may itself be modulated by antisense oligonucleotides targeted to one or more JNK proteins. Consequently, the invention satisfies the long-felt need for improved compositions and methods for modulating the metastasis of malignant tumors.
SUMMARY OF THE INVENTION
In accordance with the present invention, oligonucleotides are provided which specifically hybridize with a nucleic acid encoding a JNK protein. Certain oligonucleotides of the invention are designed to bind either directly to mRNA transcribed from, or to a selected DNA portion of, a JNK gene that encodes a JNK protein, thereby modulating the expression thereof and/or the phosphorylation of one or more substrates for the JNK protein. Pharmaceutical compositions comprising the oligonucleotides of the invention, and various methods of using the oligonucleotides of the invention, including methods of modulating one or more metastatic events, are also herein provided.
DETAILED DESCRIPTION OF THE INVENTION
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 genes { JNK) encoding Jun N-terminal kinase (JNK proteins) are particularly amenable to this approach. In the context of the invention, the terms "Jun N-terminal kinase" and "JNK protein" refer to proteins actually known to phosphorylate the amino terminal (N- terminal) portion of the Jun subunit of AP-1, as well as those that have been tentatively identified as JNK proteins based on amino acid sequence but which may in fact additionally or alternatively bind and/or phosphorylate either other transcription factors (e.g., ATF2) or kinase substrates that are not known to be involved in transcription (Derijard et al . , Cell , 1994, 76, 1025; Kallunki et al . , Genes & Development , 1994, 8, 2996; Gupta et al . , EMBO J. , 1996, 15, 2760). More specifically, the present invention is directed to antisense oligonucleotides that modulate the JNK1, JNK2 and JNK3 proteins. As a consequence of the association between cellular proliferation and activation { via phosphorylation) of AP-1, other transcription factors and/or other proteins by JNK proteins, inhibition of the expression of one or more JNK proteins leads to inhibition of the activation of AP-1 and/or other factors involved in cellular proliferation, cell cycle progression or metastatic events, and, accordingly, results in modulation of these activities. Such modulation is desirable for treating, alleviating or preventing 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 the expression of one JNK protein can be combined with modulation of one or more additional JNK proteins in order to achieve a requisite level of interference with AP-1-mediated transcription .
Methods of modulating the expression of JNK proteins comprising contacting animals with oligonucleotides specifically hybridizable with a nucleic acid encoding a JNK protein are herein provided. These methods are believed to be useful both therapeutically and diagnostically as a consequence of the association between kinase-mediated activation of AP-1 and cellular proliferation. These methods are also useful as tools, for example, in the detection and determination of the role of kinase-mediated activation of AP-1 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 JNK-mediated activation using the oligonucleotides of the invention. Methods of treating conditions in which abnormal or excessive JNK-mediated cellular proliferation occurs 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.
The present invention employs oligonucleotides for use in antisense modulation of the function of DNA or messenger RNA (mRNA) encoding a protein the modulation of which is desired, and ultimately to regulate the amount of such a protein. Hybridization of an antisense oligonucleotide with its mRNA target 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 protein, wherein "modulation" means either an increase (stimulation) or a decrease (inhibition) in the expression of the protein. In the context of the present invention, inhibition is the preferred form of modulation of gene expression.
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 associated with hyperproliferative disorders. The targeting process also includes determination of a site or sites 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 site or sites 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. Generally, there are five regions of a gene that may be targeted for antisense modulation: the 5' untranslated 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 region (hereinafter, the "3'-UTR") . 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'-UTR, tIR, ORF, tTR, 3'-UTR. As is 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 , 1983, Garland Publishing Inc., New York, pp. 411-415). 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 vitro (Brigstock et al., Growth Factors, 1990, 4, 45; Gelbert et al., Somat. Cell. Mol. Genet., 1990, 16, 173; Gold and Stormo, in:
Escherichia coli and Salmonella typhimurium: Cellular and Molecular Biology, Vol. 2, 1987, Neidhardt et al., eds., American Society for Microbiology, Washington, D.C., p. 1303,) . 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. Na tl . 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 JNK 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.
The remainder of the Detailed Description relates in more detail the (1) Oligonucleotides of the Invention and their (2) Bioequivalents, (3) Utility, (4) Pharmaceutical Compositions and (5) Means of Administration. 1. Oligonucleotides of the Invention: The present invention employs oligonucleotides for use in antisense modulation of one or more JNK proteins. 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. The oligonucleotides in accordance with this invention preferably comprise from about 8 to about 30 nucleotides. An unmodified (naturally occurring) nucleotide has three components: (1) a nitrogen- containing heterocyclic ase linked by one of its nitrogen atoms to (2) a 5-pentofuranosyl sugar and (3) a phospha te esterified to one of the 5' or 3' carbon atoms of the sugar. When incorporated into an oligonucleotide chain, the phosphate of a first nucleotide is also esterified to an adjacent sugar of a second, adjacent nucleotide via a 3 '-5' phosphate linkage. The "backbone" of an unmodified oligonucleotide consists of (2) and (3), that is, sugars linked together by phosphodiester linkages between the 5' carbon of the sugar of a first nucleotide and the 3' carbon 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 Replica tion, W.H. Freeman & Co., San Francisco, 1980, pages 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. 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." In the context of the 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. An oligonucleotide is specifically hybridizable to its target sequence due to the formation of base pairs between specific partner nucleobases in the interior of a nucleic acid duplex. Among the naturally occurring nucleobases, guanine (G) binds to cytosine (C) , and adenine (A) binds to thymine (T) or uracil (U) . In addition to the equivalency of U (RNA) and T (DNA) as partners for A, other naturally occurring nucleobase equivalents are known, including 5- methylcytosine, 5-hydroxymethylcytosine (HMC) , glycosyl HMC and gentiobiosyl HMC (C equivalents) , and 5- hydroxymethyluracil (U equivalent) . Furthermore, synthetic nucleobases which retain partner specificity are known in the art and include, for example, 7-deaza-Guanine, which retains partner specificity for C. Thus, an oligonucleotide' s capacity to specifically hybridize with its target sequence will not be altered by any chemical modification to a nucleobase in the nucleotide sequence of the oligonucleotide which does not significantly effect its specificity for the partner nucleobase in the target oligonucleotide. 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 there is a sufficient degree of complementarity to avoid nonspecific 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.
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.
A. Modified Linkages: 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 O-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 No. 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).
B. Modified Nucleobases: 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- hydroxymethylcytosine (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 Replica tion, W.H. Freeman & Co., San Francisco, 1980, pages 75-77; Gebeyehu, G. , et al . , Nucleic Acids Res . , 1987, 15, 4513) . C. Sugar Modifications: 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; Cx to C10 lower alkyl, alkoxyalkoxy, substituted lower alkyl, alkaryl or aralkyl; Cl; Br; CN; CF3; OCF3; 0-, S-, or N- alkyl; 0-, S-, or N-alkenyl; SOCH3; S02CH3; 0N02; 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 includes 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) . D. Other Modifications: 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 Conj uga tes (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, G., 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. U.S.A., 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, 253:327; 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 Nos. 5,138,045, 5,218,105 and 5,459,255.
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) and those having substantially chirally pure (Sp or Rp) alkylphosphonate, phosphoamidate or phosphotriester linkages (Cook, U.S. Patents Nos. 5,212,295 and 5,521,302).
E. Chimeric Oligonucleotides: 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 "gap ers," 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 .
F. Synthesis: 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.
1. Teachings regarding the synthesis of particular modified oligonucleotides may be found in 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 β- 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; U.S. Patents Nos. 5,587,361 and 5,599,797, drawn to oligonucleotides having phosphorothioate linkages of high chiral purity; U.S. Patent No. 5,506,351, drawn to processes for the preparation of 2'-0-alkyl guanosine and related compounds, including 2, 6-diaminopurine compounds; U.S. Patent No. 5,587,469, drawn to oligonucleotides having N-2 substituted purines; U.S. Patent No. 5,587,470, drawn to oligonucleotides having 3-deazapurines; U.S. Patents Nos. 5,223,168, issued June 29, 1993, and 5,608,046, both drawn to conjugated 4 ' -desmethyl nucleoside analogs; U.S. Patent Nos. 5,602,240, and 5,610,289, drawn to backbone modified oligonucleotide analogs; and U.S. patent application Serial No. 08/383,666, filed February 3, 1995, and U.S. Patent No. 5,459,255, drawn to, inter alia , methods of synthesizing 2 ' -fluoro-oligonucleotides . 2. 5-methyl-cytosine: In 2 ' -methoxyethoxy- modified oligonucleotides, 5-methyl-2 ' -methoxyethoxy- cytosine residues are used and are prepared as follows.
(a) 2,2'-Anhydro[l-(β-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 slightly 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.
(b) 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. (c) 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 NaCl. 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%) .
(d) 3 ' -O-Acetyl-2 ' -O-methoxyethyl-5 ' - O-dimethoxytrityl-5-methyluridine : 2 ' -O-Methoxyethyl-5 ' -0- 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 L of saturated NaCl. 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%) .
(e) 3 ' -O-Acetyl-2 ' -O-methoxyethyl-5 ' - 0-dimethoxytrityl-5-methyl-4-triazoleuridine: A first solution was prepared by dissolving 3 ' -O-acetyl-2 ' -0- 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 NaCl, dried over sodium sulfate and evaporated. The residue was triturated with EtOAc to give the title compound.
(f) 2 ' -O-Methoxyethyl-5 ' -O- dimethoxytrityl-5-methylcytidine: A solution of 3 ' -0- acetyl-2 ' -O-methoxyethyl-5 ' -0-dimethoxytrityl-5-methyl-4- triazoleuridine (103 g, 0.141 M) in dioxane (500 mL) and NH4OH (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 NaCl (200 mL) . The organics were dried over sodium sulfate and the solvent was evaporated to give 85 g (95%) of the title compound.
(g) N-Benzoyl-2 ' -O-methoxyethyl-5 ' -O- dimethoxytrityl-5-methylcytidine : 2 ' -O-Methoxyethyl-5 ' -0- 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 NaCl (2x 300 mL) , dried over MgSO, 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% EtNH as the eluting solvent. The pure product fractions were evaporated to give 90 g (90%) of the title compound.
(h) N-Benzoyl-2 ' -O-methoxyethyl-5 ' -O- dimethoxytrityl-5-πtethylcytidine-3 ' -amidite: N-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 NaCl (3x 300 mL) . The aqueous washes were back- extracted with CH2C12 (300 mL) , and the extracts were combined, dried over MgS04 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 .
2. Bioequivalents : The compounds of the invention encompass any pharmaceutically acceptable salts, esters, or salts of such esters, or any other compound which, upon administration to an animal including a human, is capable of providing (directly or indirectly) the biologically active metabolite or residue thereof. Accordingly, for example, the disclosure is also drawn to "prodrugs" and "pharmaceutically acceptable salts" of the oligonucleotides of the invention, pharmaceutically acceptable salts of such prodrugs, and other bioequivalents.
A. Oligonucleotide Prodrugs: 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.
B. Pharmaceutically Acceptable Salts: The term "pharmaceutically acceptable salts" refers to physiologically and pharmaceutically acceptable salts of the oligonucleotides of the invention: i.e., salts that retain the desired biological activity of the parent compound and do not impart undesired toxicological effects thereto. Pharmaceutically acceptable base addition salts are formed with metals or amines, such as alkali and alkaline earth metals or organic amines. Examples of metals used as cations are sodium, potassium, magnesium, calcium, and the like. Examples of suitable amines are N,N ' -dibenzylethylenediamine, chloroprocaine, choline, diethanolamine, dicyclohexylamine, ethylenediamine, N-methylglucamine, and procaine (see, for example, Berge et al . , "Pharmaceutical Salts," J. of Pharma Sci . , 1977, 66:1) . The base addition salts of said acidic compounds are prepared by contacting the free acid form with a sufficient amount of the desired base to produce the salt in the conventional manner. The free acid form may be regenerated by contacting the salt form with an acid and isolating the free acid in the conventional manner. The free acid forms differ from their respective salt forms somewhat in certain physical properties such as solubility in polar solvents, but otherwise the salts are equivalent to their respective free acid for purposes of the present invention. As used herein, a "pharmaceutical addition salt" includes a pharmaceutically acceptable salt of an acid form of one of the components of the compositions of the invention. These include organic or inorganic acid salts of the amines. Preferred acid salts are the hydrochlorides, acetates, salicylates, nitrates and phosphates. Other suitable pharmaceutically acceptable salts are well known to those skilled in the art and include basic salts of a variety of inorganic and organic acids, such as, for example, with inorganic acids, such as for example hydrochloric acid, hydrobromic acid, sulfuric acid or phosphoric acid; with organic carboxylic, sulfonic, sulfo or phospho acids or N-substituted sulfamic acids, for example acetic acid, propionic acid, glycolic acid, succinic acid, maleic acid, hydroxymaleic acid, methylmaleic acid, fumaric acid, malic acid, tartaric acid, lactic acid, oxalic acid, gluconic acid, glucaric acid, glucuronic acid, citric acid, benzoic acid, cinnamic acid, mandelic acid, salicylic acid, 4-aminosalicylic acid, 2-phenoxybenzoic acid, 2-acetoxybenzoic acid, embonic acid, nicotinic acid or isonicotinic acid; and with amino acids, such as the 20 alpha-amino acids involved in the synthesis of proteins in nature, for example glutamic acid or aspartic acid, and also with phenylacetic acid, methanesulfonic acid, ethanesulfonic acid,
2-hydroxyethanesulfonic acid, ethane-1, 2-disulfonic acid, benzenesulfonic acid, 4-methylbenzenesulfonic acid, naphthalene-2-sulfonic acid, naphthalene-1, 5-disulfonic acid, 2- or 3-phosphoglycerate, glucose-6-phosphate, N-cyclohexylsulfamic acid (with the formation of cyclamates) , or with other acid organic compounds, such as ascorbic acid. Pharmaceutically acceptable salts of compounds may also be prepared with a pharmaceutically acceptable cation. Suitable pharmaceutically acceptable cations are well known to those skilled in the art and include alkaline, alkaline earth, ammonium and quaternary ammonium cations. Carbonates or hydrogen carbonates are also possible. For oligonucleotides, preferred examples of pharmaceutically acceptable salts include but are not limited to (a) salts formed with cations such as sodium, potassium, ammonium, magnesium, calcium, polyamines such as spermine and spermidine, etc. ; (b) acid addition salts formed with inorganic acids, for example hydrochloric acid, hydrobromic acid, sulfuric acid, phosphoric acid, nitric acid and the like; (c) salts formed with organic acids such as, for example, acetic acid, oxalic acid, tartaric acid, succinic acid, maleic acid, fumaric acid, gluconic acid, citric acid, malic acid, ascorbic acid, benzoic acid, tannic acid, palmitic acid, alginic acid, polyglutamic acid, naphthalenesulfonic acid, methanesulfonic acid, p-toluenesulfonic acid, naphthalenedisulfonic acid, polygalacturonic acid, and the like; and (d) salts formed from elemental anions such as chlorine, bromine, and iodine .
3. Exemplary Utilities of the Invention : The oligonucleotides of the present invention specifically hybridize to nucleic acids (e.g., mRNAs) encoding a JNK protein. The oligonucleotides of the present invention can be utilized as therapeutic compounds, as diagnostic tools or research reagents that can be incorporated into kits, and in purifications and cellular product preparations, as well as other methodologies, which are appreciated by persons of ordinary skill in the art.
A. Assays and Diagnostic Applications: The oligonucleotides of the present invention can be used to detect the presence of JNK protein-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 , Cold Spring Harbor Laboratory Press, 1989, Volume 2, pg. 10.59.) Radiolabeled oligonucleotides are then contacted with cell or tissue samples suspected of containing JNK protein message RNAs (and thus JNK 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 JNK 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 JNK protein 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 JNK protein 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. Other means of labeling oligonucleotides are known in the art (see, e . g. , Ruth, Chapter 6 In : Methods in Molecular Biology, Vol . 26: Protocols for Oligonucleotide Conj uga tes, Agrawal, ed. , Humana Press Inc., Totowa, NJ, 1994, pages 167-185) .
Kits for detecting the presence or absence of expression of a JNK protein may also be prepared. Such kits include an oligonucleotide targeted to an appropriate gene, i.e., a gene encoding a JNK 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 JNK 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.
B. Protein Purifications: The oligonucleotides of the invention are also useful for the purification of specific Jun kinase proteins from cells that normally express a set of JNK proteins which are similar to each other in terms of their polypeptide sequences and biochemical properties. As an example, the purification of a JNK1 protein from cells that expresses JNK1, JNK2 and JNK3 proteins can be enhanced by first treating such cells with oligonucleotides that inhibit the expression of JNK2 and JNK3 and/or with oligonucleotides that increase the expression of JNK1, because such treatments will increase the relative ratio of JNK1 relative to JNK2 and JNK3. As a result, the yield of JNK1 from subsequent purification steps will be improved as the amount of the biochemically similar (and thus likely to contaminate) JNK2 and JNK3 proteins in extracts prepared from cells so treated will be diminished.
C . Biologically Active Oligonucleotides : The invention is also drawn to the administration of oligonucleotides having biological activity to cultured cells, isolated tissues and organs and animals. By "having biological activity," it is meant that the oligonucleotide functions to modulate the expression of one or more genes in cultured cells, isolated tissues or organs and/or animals. Such modulation can be achieved by an antisense oligonucleotide by a variety of mechanisms known in the art, including but not limited to transcriptional arrest; effects on RNA processing (capping, polyadenylation and splicing) and transportation; enhancement of cellular degradation of the target nucleic acid; and translational arrest (Crooke et al . , Exp. Opin . Ther. Pa tents, 1996, 6:855) .
In an animal other than a human, the compositions and methods of the invention can be used to study the function of one or more genes in the animal. For example, antisense oligonucleotides have been systemically administered to rats in order to study the role of the W-methyl-D-aspartate receptor in neuronal death, to mice in order to investigate the biological role of protein kinase C- , and to rats in order to examine the role of the neuropeptide Yl receptor in anxiety (Wahlestedt et al . , Na ture, 1993, 363:260; Dean et al . , Proc . Na tl . Acad. Sci . U. S . A. , 1994, 91:11762; and Wahlestedt et al . , Science, 1993, 259:528, respectively). In instances where complex families of related proteins are being investigated, "antisense knockouts" (i.e., inhibition of a gene by systemic administration of antisense oligonucleotides) may represent the most accurate means for examining a specific member of the family (see, generally, Albert et al . , Trends Pharmacol . Sci . , 1994, 15:250).
The compositions and methods of the invention also have therapeutic uses in an animal, including a human, having (i.e., suffering from), or known to be or suspected of being prone to having, a disease or disorder that is treatable in whole or in part with one or more nucleic acids. 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 JNK 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 carrier such as, e.g., a diluent. 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 following U.S. patents demonstrate palliative, therapeutic and other methods utilizing antisense oligonucleotides. U. S. Patent No. 5,135,917 provides antisense oligonucleotides that inhibit human interleukin-1 receptor expression. U.S. Patent No. 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 No. 5,087,617 provides methods for treating cancer patients with antisense oligonucleotides. U.S.
Patent No. 5,166,195 provides oligonucleotide inhibitors of Human Immunodeficiency Virus (HIV). U.S. Patent No. 5,004,810 provides oligomers capable of hybridizing to herpes simplex virus Vmw65 mRNA and inhibiting replication. U.S. Patent No. 5,194,428 provides antisense oligonucleotides having antiviral activity against influenzavirus . U.S. Patent No. 4,806,463 provides antisense oligonucleotides and methods using them to inhibit HTLV-III replication. U.S. Patent No. 5,286,717 provides oligonucleotides having a complementary base sequence to a portion of an oncogene. U.S. Patent No. 5,276,019 and U.S. Patent No. 5,264,423 are directed to phosphorothioate oligonucleotide analogs used to prevent replication of foreign nucleic acids in cells. U.S. Patent No. 4,689,320 is directed to antisense oligonucleotides as antiviral agents specific to cytomegalovirus (CMV) . U.S. Patent No. 5,098,890 provides oligonucleotides complementary to at least a portion of the mRNA transcript of the human c-myb gene. U.S. Patent No. 5,242,906 provides antisense oligonucleotides useful in the treatment of latent Epstein-Barr virus (EBV) infections.
As used herein, the term "disease or disorder" (1) includes any abnormal condition of an organism or part, especially as a consequence of infection, inherent weakness, environmental stress, that impairs normal physiological functioning; (2) excludes pregnancy per se but not autoimmune and other diseases associated with pregnancy; and (3) includes cancers and tumors. The term "known to be or suspected of being prone to having a disease or disorder" indicates that the subject animal has been determined to be, or is suspected of being, at increased risk, relative to the general population of such animals, of developing a particular disease or disorder as herein defined. For example, a subject animal "known to be or suspected of being prone to having a disease or disorder" could have a personal and/or family medical history that includes frequent occurrences of a particular disease or disorder. As another example, a subject animal "known to be or suspected of being prone to having a disease or disorder" could have had such a susceptibility determined by genetic screening according to techniques known in the art (see, e . g. , U.S. Congress, Office of Technology Assessment, Chapter 5 In : Genetic Moni toring and Screening in the Workplace, OTA-BA-455, U.S. Government Printing Office, Washington, D.C., 1990, pages 75-99). The term "a disease or disorder that is treatable in whole or in part with one or more nucleic acids" refers to a disease or disorder, as herein defined, (1) the management, modulation or treatment thereof, and/or (2) therapeutic, curative, palliative and/or prophylactic relief therefrom, can be provided via the administration of an antisense oligonucleotide . 4. Pharmaceutical Compositions: The formulation of pharmaceutical compositions comprising the oligonucleotides of the invention, and their subsequent administration, are believed to be within the skill of those in the art. A. Therapeutic Considerations: In general, for therapeutic applications, a patient (i.e., an animal, including a human, having or predisposed to a disease or disorder) is administered one or more oligonucleotides, in accordance with the invention in a pharmaceutically acceptable carrier in doses ranging from 0.01 μg 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. In the context of the invention, the term "treatment regimen" is meant to encompass therapeutic, palliative and prophylactic modalities. 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 nucleic acid 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 ablated.
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. An optimal dosing schedule is used to deliver a therapeutically effective amount of the oligonucleotide being administered via a particular mode of administration. The term "therapeutically effective amount," for the purposes of the invention, refers to the amount of oligonucleotide-containing pharmaceutical composition which is effective to achieve an intended purpose without undesirable side effects (such as toxicity, irritation or allergic response) . Although individual needs may vary, determination of optimal ranges for effective amounts of pharmaceutical compositions is within the skill of the art. Human doses can be extrapolated from animal studies (Katocs et al . , Chapter 27 In : Remington ' s Pharmaceutical Sciences, 18th Ed., Gennaro, ed. , Mack Publishing Co., Easton, PA, 1990) . Generally, the dosage required to provide an effective amount of a pharmaceutical composition, which can be adjusted by one skilled in the art, will vary depending on the age, health, physical condition, weight, type and extent of the disease or disorder of the recipient, frequency of treatment, the nature of concurrent therapy (if any) and the nature and scope of the desired effect (s) (Nies et al . , Chapter 3 In : Goodman & Gilman ' s The Pharmacological Basis of Therapeutics, 9th Ed., Hardman et al . , eds., McGraw-Hill, New York, NY, 1996). As used herein, the term "high risk individual" is meant to refer to an individual for whom it has been determined, via, e.g., individual or family history or genetic testing, has a significantly higher than normal probability of being susceptible to the onset or recurrence of a disease or disorder. As art of treatment regimen for a high risk individual, the individual can be prophylactically treated to prevent the onset or recurrence of the disease or disorder. The term "prophylactically effective amount" is meant to refer to an amount of a pharmaceutical composition which produces an effect observed as the prevention of the onset or recurrence of a disease or disorder. Prophylactically effective amounts of a pharmaceutical composition are typically determined by the effect they have compared to the effect observed when a second pharmaceutical composition lacking the active agent is administered to a similarly situated individual.
Following successful treatment, it may be desirable to have the patient undergo maintenance therapy to prevent the recurrence of the disease state, wherein the nucleic acid is administered in maintenance doses, ranging from 0.01 μg to 100 g per kg of body weight, once or more daily, to once every 20 years. For example, in the case of in individual known or suspected of being prone to an autoimmune or inflammatory condition, prophylactic effects may be achieved by administration of preventative doses, ranging from 0.01 μg to 100 g per kg of body weight, once or more daily, to once every 20 years. In like fashion, an individual may be made less susceptible to an inflammatory condition that is expected to occur as a result of some medical treatment, e.g., graft versus host disease resulting from the transplantation of cells, tissue or an organ into the individual. 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 a first JNK protein is used in combination with a second antisense oligonucleotide targeted to a second JNK protein in order to such JNK proteins to a more extensive degree than can be achieved when either oligonucleotide is used individually. In various embodiments of the invention, the first and second JNK proteins which are targeted by such oligonucleotides are identical, are different JNK proteins or are different isoforms of the same JNK protein.
B. Pharmaceutical Compositions: Pharmaceutical compositions for the non-parenteral administration of oligonucleotides may include sterile aqueous solutions which may also contain buffers, diluents and other suitable additives. Pharmaceutically acceptable organic or inorganic carrier substances suitable for non-parenteral administration which do not deleteriously react with oligonucleotides can be used. Suitable pharmaceutically acceptable carriers include, but are not limited to, water, salt solutions, alcohol, polyethylene glycols, gelatin, lactose, amylose, magnesium stearate, talc, silicic acid, viscous paraffin, hydroxymethylcellulose, polyvinylpyrrolidone and the like. The pharmaceutical compositions can be sterilized and, if desired, mixed with auxiliary agents, e.g., lubricants, preservatives, stabilizers, wetting agents, emulsifiers, salts for influencing osmotic pressure, buffers, colorings flavorings and/or aromatic substances and the like which do not deleteriously react with the oligonucleotide (s) of the pharmaceutical composition. Pharmaceutical compositions in the form of aqueous suspensions may contain substances which increase the viscosity of the suspension including, for example, sodium carboxymethylcellulose, sorbitol and/or dextran. Optionally, such suspensions may also contain stabilizers . In one embodiment of the invention, an oligonucleotide is administered via the rectal mode. In particular, pharmaceutical compositions for rectal administration include foams, solutions (enemas) and suppositories. Rectal suppositories for adults are usually tapered at one or both ends and typically weigh about 2 g each, with infant rectal suppositories typically weighing about one-half as much, when the usual base, cocoa butter, is used (Block, Chapter 87 In : Remington '' s Pharmaceutical Sciences, 18th Ed., Gennaro, ed., Mack Publishing Co., Easton, PA, 1990) .
In a preferred embodiment of the invention, one or more oligonucleotides are administered via oral delivery. Pharmaceutical compositions for oral administration include powders or granules, suspensions or solutions in water or non-aqueous media, capsules, sachets, troches, tablets or SECs (soft elastic capsules or "caplets") . Thickeners, flavoring agents, diluents, emulsifiers, dispersing aids, carrier substances or binders may be desirably added to such pharmaceutical compositions. The use of such pharmaceutical compositions has the effect of delivering the oligonucleotide to the alimentary canal for exposure to the mucosa thereof. Accordingly, the pharmaceutical composition can comprise material effective in protecting the oligonucleotide from pH extremes of the stomach, or in releasing the oligonucleotide over time, to optimize the delivery thereof to a particular mucosal site. Enteric coatings for acid-resistant tablets, capsules and caplets are known in the art and typically include acetate phthalate, propylene glycol and sorbitan monoleate. Various methods for producing pharmaceutical compositions for alimentary delivery are well known in the art. See, generally, Nairn, Chapter 83; Block, Chapter 87; Rudnic et al . , Chapter 89; Porter, Chapter 90; and Longer et al . , Chapter 91 In : Remington ' s Pharmaceutical Sciences, 18th Ed., Gennaro, ed. , Mack Publishing Co., Easton, PA, 1990. The oligonucleotides of the invention can be incorporated in a known manner into customary pharmaceutical compositions, such as tablets, coated tablets, pills, granules, aerosols, syrups, emulsions, suspensions and solutions, using inert, non-toxic, pharmaceutically acceptable carriers (excipients) . The therapeutically active compound should in each case be present here in a concentration of about 0.5% to about 95% by weight of the total mixture, i.e., in amounts which are sufficient to achieve the stated dosage range. The pharmaceutical compositions are prepared, for example, by diluting the active compounds with pharmaceutically acceptable carriers, if appropriate using emulsifying agents and/or dispersing agents, and, for example, in the case where water is used as the diluent, organic solvents can be used as auxiliary solvents if appropriate. Pharmaceutical compositions may be formulated in a conventional manner using additional pharmaceutically acceptable carriers as appropriate. Thus, the compositions may be prepared by conventional means with additional excipients such as binding agents (e.g., pregelatinised maize starch, polyvinylpyrrolidone or hydroxypropyl methylcellulose) ; fillers (e.g., lactose, microcrystalline cellulose or calcium hydrogen phosphate); lubricants (e.g., magnesium stearate, talc or silica); disintegrates (e.g., starch or sodium starch glycolate) ; or wetting agents (e.g., sodium lauryl sulfate). Tablets may be coated by methods well known in the art. The preparations may also contain flavoring, coloring and/or sweetening agents as appropriate. The pharmaceutical compositions, which may conveniently be presented in unit dosage form, may be prepared according to conventional techniques well known in the pharmaceutical industry. Such techniques include the step of bringing into association the active ingredient (s) with the pharmaceutically acceptable carrier (s). In general the pharmaceutical compositions are prepared by uniformly and intimately bringing into association the active ingredient (s) with liquid excipients or finely divided solid excipients or both, and then, if necessary, shaping the product.
Pharmaceutical compositions of the present invention suitable for oral administration may be presented as discrete units such as capsules, cachets or tablets each containing predetermined amounts of the active ingredients; as powders or granules; as solutions or suspensions in an aqueous liquid or a non-aqueous liquid; or as oil-in-water emulsions or water-in-oil liquid emulsions. A tablet may be made by compression or molding, optionally with one or more accessory ingredients. Compressed tablets may be prepared by compressing in a suitable machine, the active ingredients in a free-flowing form such as a powder or granules, optionally mixed with a binder, lubricant, inert diluent, preservative, surface active or dispersing agent. Molded tablets may be made by molding in a suitable machine a mixture of the powdered compound moistened with an inert liquid diluent. The tablets may optionally be coated or scored and may be formulated so as to provide slow or controlled release of the active ingredients therein. Pharmaceutical 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. C. Penetration Enhancers: Pharmaceutical compositions comprising the oligonucleotides of the present invention may also include penetration enhancers in order to enhance the alimentary delivery of the oligonucleotides. Penetration enhancers may be classified as belonging to one of five broad categories, i.e., fatty acids, bile salts, chelating agents, surfactants and non-surfactants (Lee et al . , Cri tical Reviews in Therapeutic Drug Carrier Systems, 1991, 8:91-192; Muranishi, Cri tical Reviews in Therapeutic Drug Carrier Systems, 1990, 7:1) .
1. Fatty Acids: Various fatty acids and their derivatives which act as penetration enhancers include, for example, oleic acid, lauric acid, capric acid, myristic acid, palmitic acid, stearic acid, linoleic acid, linolenic acid, dicaprate, tricaprate, recinleate, monoolein (a.k.a. 1-monooleoyl-rac-glycerol) , dilaurin, caprylic acid, arichidonic acid, glyceryl 1-monocaprate, 1- dodecylazacycloheptan-2-one, acylcarnitines, acylcholines, mono- and di-glycerides and physiologically acceptable salts thereof (i.e., oleate, laurate, caprate, myristate, palmitate, stearate, linoleate, etc. ) (Lee et al . , Cri tical Reviews in Therapeutic Drug Carrier Systems, 1991, page 92; Muranishi, Cri tical Reviews in Therapeutic Drug Carrier Systems, 1990, 7:1; El-Hariri et al . , J. Pharm . Pharmacol . , 1992, 44:651).
2. Bile Salts: The physiological roles of bile include the facilitation of dispersion and absorption of lipids and fat-soluble vitamins (Brunton, Chapter 38 In : Goodman & Gilman ' s The Pharmacological Basis of Therapeutics, 9th Ed., Hardman et al . , eds., McGraw-Hill, New York, NY, 1996, pages 934-935) . Various natural bile salts, and their synthetic derivatives, act as penetration enhancers. Thus, the term "bile salt" includes any of the naturally occurring components of bile as well as any of their synthetic derivatives.
3. Chelating Agents: Chelating agents have the added advantage of also serving as DNase inhibitors and include, but are not limited to, disodium ethylenediaminetetraacetate (EDTA) , citric acid, salicylates (e.g., sodium salicylate, 5-methoxysalicylate and homovanilate) , W-acyl derivatives of collagen, laureth- 9 and IV-amino acyl derivatives of beta-diketones (enamines) (Lee et al . , Cri tical Reviews in Therapeutic Drug Carrier Systems, 1991, page 92; Muranishi, Cri tical Reviews in Therapeutic Drug Carrier Systems, 1990, 7:1; Buur et al . , J. Control Rel . , 1990, 14:43).
4. Surfactants: Surfactants include, for example, sodium lauryl sulfate, polyoxyethylene-9-lauryl ether and polyoxyethylene-20-cetyl ether (Lee et al . , Cri tical Reviews in Therapeuti c Drug Carrier Systems, 1991, page 92) ; and perfluorochemical emulsions, such as FC-43 (Takahashi et al . , J. Pharm . Phama col . , 1988, 40:252). 5. Non-Surfactants: Non-surfactants include, for example, unsaturated cyclic ureas, 1-alkyl- and 1-alkenylazacyclo-alkanone derivatives (Lee et a l . , Cri tical Reviews in Therapeutic Drug Carrier Systems, 1991, page 92) ; and non-steroidal anti-inflammatory agents such as diclofenac sodium, indomethacin and phenylbutazone (Yamashita et al . , J. Pharm . Pharmacol . , 1987, 39:621).
D. Carrier Compounds: As used herein, "carrier compound" refers to a nucleic acid, or analog thereof, which is inert (i.e., does not possess biological activity per se) but is recognized as a nucleic acid by in vivo processes that reduce the bioavailability of a nucleic acid having biological activity by, for example, degrading the biologically active nucleic acid or promoting its removal from circulation. The coadministration of a nucleic acid and a carrier compound, typically with an excess of the latter substance, can result in a substantial reduction of the amount of nucleic acid recovered in the liver, kidney or other extracirculatory reservoirs, presumably due to competition between the carrier compound and the nucleic acid for a common receptor. For example, the recovery of a partially phosphorothioated oligonucleotide in hepatic tissue is reduced when it is coadministered with polyinosinic acid, dextran sulfate, polycytidic acid or 4- acetamido-4 ' -isothiocyano-stilbene-2 , 2 ' -disulfonic acid (Miyao et al . , Antisense Res . Dev. , 1995, 5:115; Takakura et al . , Antisense & Nucl . Acid Drug Dev. , 1996, 6:177). E. Pharmaceutically Acceptable Carriers: In contrast to a carrier compound, a "pharmaceutically acceptable carrier" (excipient) is a pharmaceutically acceptable solvent, suspending agent or any other pharmacologically inert vehicle for delivering one or more nucleic acids to an animal. The pharmaceutically acceptable carrier may be liquid or solid and is selected with the planned manner of administration in mind so as to provide for the desired bulk, consistency, etc. , when combined with a nucleic acid and the other components of a given pharmaceutical composition. Typical pharmaceutically acceptable carriers include, but are not limited to, binding agents (e.g., pregelatinised maize starch, polyvinylpyrrolidone or hydroxypropyl methylcellulose, etc.); fillers (e.g., lactose and other sugars, microcrystalline cellulose, pectin, gelatin, calcium sulfate, ethyl cellulose, polyacrylates or calcium hydrogen phosphate, etc. ) ; lubricants (e.g., magnesium stearate, talc, silica, colloidal silicon dioxide, stearic acid, metallic stearates, hydrogenated vegetable oils, corn starch, polyethylene glycols, sodium benzoate, sodium acetate, etc. ) ; disintegrates (e.g., starch, sodium starch glycolate, etc. ) ; or wetting agents (e.g., sodium lauryl sulphate, etc.). Sustained release oral delivery systems and/or enteric coatings for orally administered dosage forms are described in U.S. Patents Nos. 4,704,295; 4,556,552; 4,309,406; and 4,309,404.
F. Miscellaneous Additional Components: The compositions of the present invention may additionally contain other adjunct components conventionally found in pharmaceutical compositions, at their art-established usage levels. Thus, for example, the compositions may contain additional compatible pharmaceutically-active materials such as, e.g., antipruritics, astringents, local anesthetics or anti-inflammatory agents, or may contain additional materials useful in physically formulating various dosage forms of the composition of present invention, such as dyes, flavoring agents, preservatives, antioxidants, opacifiers, thickening agents and stabilizers. However, such materials, when added, should not unduly interfere with the biological activities of the components of the compositions of the invention.
G. Colloidal Dispersion Systems: 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, 6, 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, sphingolipids, phosphatidylethanolamine, 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. 5. Means of Administration: The present invention provides compositions comprising oligonucleotides intended for administration to an animal. For purposes of the invention, unless otherwise specified, the term "animal" is meant to encompass humans as well as other mammals, as well as reptiles, amphibians, and birds.
A. Parenteral Delivery: The term "parenteral delivery" refers to the administration of an oligonucleotide of the invention to an animal in a manner other than through the digestive canal. Means of preparing and administering parenteral pharmaceutical compositions are known in the art (see, e.g., Avis, Chapter 84 In : Remington ' s Pharmaceutical Sciences, 18th Ed., Gennaro, ed., Mack Publishing Co., Easton, PA, 1990, pages 1545- 1569) . Parenteral means of delivery include, but are not limited to, the following illustrative examples.
1. Intravitreal injection, for the direct delivery of drug to the vitreous humor of a mammalian eye, is described in U.S. Patent No. 5,591,720, the contents of which are hereby incorporated by reference. Means of preparing and administering ophthalmic preparations are known in the art (see, e.g., Mullins et al . , Chapter 86 In : Remington ' s Pharmaceutical Sciences, 18th Ed., Gennaro, ed., Mack Publishing Co., Easton, PA, 1990, pages 1581- 1595) .
2. Intravenous administration of antisense oligonucleotides to various non-human mammals has been described by Iversen (Chapter 26 In : Antisense Research and Applica tions, Crooke et al . , eds., CRC Press, Boca Raton, FL, 1993, pages 461-469) . Systemic delivery of oligonucleotides to non-human mammals via intraperitoneal means has also been described (Dean et al . , Proc . Na tl . Acad. Sci . (U. S . A . ) , 1994, 91 , 11766). 3. 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, 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).
4. 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. 5. Intrathecal drug administration, for the introduction of a 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) .
6. Epidermal and Transdermal Delivery, in which pharmaceutical compositions containing drugs are applied topically, can be used to administer drugs to be absorbed by the local dermis or for further penetration and absorption by underlying tissues, respectively. Means of preparing and administering medications topically are known in the art (see, e.g., Block, Chapter 87 In : Remington ' s Pharmaceutical Sciences, 18th Ed., Gennaro, ed. , Mack Publishing Co., Easton, PA, 1990, pages 1596-1609).
7. Vaginal Delivery provides local treatment and avoids first pass metabolism, degradation by digestive enzymes, and potential systemic side-effects. This mode of administration may be preferred for antisense oligonucleotides targeted to pathogenic organisms for which the vagina is the usual habitat, e.g., Trichomonas vaginalis . In another embodiment, antisense oligonucleotides to genes encoding sperm-specific antibodies can be delivered by this mode of administration in order to increase the probability of conception and subsequent pregnancy. Vaginal suppositories (Block, Chapter 87 In : Remington ' s Pharmaceutical Sciences, 18th Ed., Gennaro, ed. , Mack Publishing Co., Easton, PA, 1990, pages 1609-1614) or topical ointments can be used to effect this mode of delivery.
8. Intravesical Delivery provides local treatment and avoids first pass metabolism, degradation by digestive enzymes, and potential systemic side-effects. However, the method requires urethral catheterization of the patient and a skilled staff. Nevertheless, this mode of administration may be preferred for antisense oligonucleotides targeted to pathogenic organisms, such as T. vaginalis, which may invade the urogenital tract.
B. Alimentary Delivery: The term "alimentary delivery" refers to the administration, directly or otherwise, to a portion of the alimentary canal of an animal. The term "alimentary canal" refers to the tubular passage in an animal that functions in the digestion and absorption of food and the elimination of food residue, which runs from the mouth to the anus, and any and all of its portions or segments, e.g., the oral cavity, the esophagus, the stomach, the small and large intestines and the colon, as well as compound portions thereof such as, e.g., the gastro-intestinal tract. Thus, the term "alimentary delivery" encompasses several routes of administration including, but not limited to, oral, rectal, endoscopic and sublingual/buccal administration. A common requirement for these modes of administration is absorption over some portion or all of the alimentary tract and a need for efficient mucosal penetration of the nucleic acid(s) so administered . 1. Buccal/Sublingual Administration:
Delivery of a drug via the oral mucosa has several desirable features, including, in many instances, a more rapid rise in plasma concentration of the drug than via oral delivery (Harvey, Chapter 35 In : Remington ' s
Pharmaceutical Sciences, 18th Ed., Gennaro, ed. , Mack Publishing Co., Easton, PA, 1990, page 711). Furthermore, because venous drainage from the mouth is to the superior vena cava, this route also bypasses rapid first-pass metabolism by the liver. Both of these features contribute to the sublingual route being the mode of choice for nitroglycerin (Benet et al . , Chapter 1 In : Goodman & Gilman ' s The Pharmacological Basis of Therapeutics, 9th Ed., Hardman et al . , eds., McGraw-Hill, New York, NY, 1996, page 7 ) .
2. Endoscopic Administration: Endoscopy can be used for drug delivery directly to an interior portion of the alimentary tract. For example, endoscopic retrograde cystopancreatography (ERCP) takes advantage of extended gastroscopy and permits selective access to the biliary tract and the pancreatic duct (Hirahata et al . , Gan To Kagaku Ryoho, 1992, 19 (10 Suppl . ) ;1591) . However, the procedure is unpleasant for the patient, and requires a highly skilled staff. 3. Rectal Administration: Drugs administered by the oral route can often be alternatively administered by the lower enteral route, i.e., through the anal portal into the rectum or lower intestine. Rectal suppositories, retention enemas or rectal catheters can be used for this purpose and may be preferred when patient • compliance might otherwise be difficult to achieve (e.g., in pediatric and geriatric applications, or when the patient is vomiting or unconscious) . Rectal administration may result in more prompt and higher blood levels than the oral route, but the converse may be true as well (Harvey, Chapter 35 In : Remington ' s Pharmaceutical Sciences, 18th Ed., Gennaro, ed., Mack Publishing Co., Easton, PA, 1990, page 711) . Because about 50% of the drug that is absorbed from the rectum will bypass the liver, administration by this route significantly reduces the potential for first- pass metabolism (Benet et al . , Chapter 1 In : Goodman & Gilman ' s The Pharmacological Basis of Therapeutics, 9th Ed., Hardman et al . , eds., McGraw-Hill, New York, NY, 1996) .
4. Oral Administration: The preferred method of administration is oral delivery, which is typically the most convenient route for access to the systemic circulation. Absorption from the alimentary canal is governed by factors that are generally applicable, e.g., surface area for absorption, blood flow to the site of absorption, the physical state of the drug and its concentration at the site of absorption (Benet et al . , Chapter 1 In : Goodman & Gilman ' s The Pharmacological Basis of Therapeutics, 9th Ed., Hardman et al . , eds., McGraw- Hill, New York, NY, 1996, pages 5-7) . A significant factor which may limit the oral bioavailability of a drug is the degree of "first pass effects." For example, some substances have such a rapid hepatic uptake that only a fraction of the material absorbed enters the peripheral blood (Van Berge-Henegouwen et al . , Gastroenterology, 1977, 73:300) . The compositions and methods of the invention circumvent, at least partially, such first pass effects by providing improved uptake of nucleic acids and thereby, e.g., causing the hepatic uptake system to become saturated and allowing a significant portion of the nucleic acid so administered to reach the peripheral circulation. Additionally or alternatively, the hepatic uptake system is saturated with one or more inactive carrier compounds prior to administration of the active nucleic acid.
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 : Synthesis of Oligonucleotides
A. 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-l, 1-dioxide in acetonitrile for the stepwise thiation of the phosphite linkages.
The synthesis of 2 ' -O-methyl- (a.k.a. 2'-methoxy-) phosphorothioate oligonucleotides is according to the procedures set forth above substituting 2 ' -O-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 ' -O-propyl- (a.k.a 2'-propoxy-) phosphorothioate oligonucleotides are 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.
The 2 ' -fluoro-phosphorothioate oligonucleotides of the invention are 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. 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 are prepared essentially as described in U.S. Patents Nos. 5,539,082 and 5,539,083, both of which (1) issued July 23, 1996, and (2) are assigned to the same assignee as the instant application. B. Purification: After cleavage from the controlled pore glass column (Applied Biosystems) and deblocking in concentrated ammonium hydroxide at 55°C for 18 hours, the oligonucleotides were purified by precipitation twice out of 0.5 M NaCl with 2.5 volumes ethanol. Analytical gel electrophoresis was accomplished in 20% acrylamide, 8 M urea, 45 mM Tris-borate buffer, pH 7.0. Oligodeoxynucleotides and their phosphorothioate analogs were judged from electrophoresis to be greater than 80% full length material.
Example 2 : Assays for Oligonucleotide-Mediated Inhibition of JNK mRNA Expression in Human Tumor Cells In order to evaluate the activity of potential JNK- modulating oligonucleotides, human lung carcinoma cell line A549 (American Type Culture Collection, Rockville, MD No. ATCC CCL-185) cells or other cell lines as indicated in the Examples, were grown and treated with oligonucleotides or control solutions as detailed below. After harvesting, cellular extracts were prepared and examined for specific JNK mRNA levels or JNK protein levels (i.e., Northern or Western assays, respectively) . In all cases, "% expression" refers to the amount of JNK-specific signal in an oligonucleotide-treated cell relative to an untreated cell (or a cell treated with a control solution that lacks oligonucleotide) , and "% inhibition" is calculated as 100% - % Expression = % Inhibition.
Northern Assays: The mRNA expression of each JNK protein was determined by using a nucleic acid probe specifically hybridizable thereto. Nucleic acid probes specific for JNK1, JNK2 and JNK3 are described in Examples 3, 4 and 5, respectively. The probes were radiolabelled by means well known in the art (see, e.g., Short Protocols in Molecular Biology, 2nd Ed., Ausubel et al . , eds., John
Wiley & Sons, New York, 1992, pages 3-11 to 2-3-44 and 4-17 to 4-18; Ruth, Chapter 6 In : Methods in Molecular Biology, Vol . 26: Protocols for Oligonucleotide Conj uga tes, Agrawal, ed., Humana Press Inc., Totowa, NJ, 1994, pages 167-185; and Chapter 10 In : Molecular Cloning: A Labora tory Manual , 2nd Ed., Sambrook et al . , eds., pages 10.1-10.70). The blots were stripped and reprobed with a 32P-labeled glyceraldehyde 3-phosphate dehydrogenase (G3PDH) probe (Clontech Laboratories, Inc., Palo Alto, CA) in order to confirm equal loading of RNA and to allow the levels of JNK transcripts to be normalized with regard to the G3PDH signals.
A549 cells were grown in T-75 flasks until 80-90% confluent. 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., 1:1 (w/w) DOTMA/DOPE, Life Technologies, Gaithersburg, MD; DOTMA = N- [ 1- (2, 3-dioleyoxy) propyl] -N, N, N-trimethylammonium chloride; DOPE = dioleoyl phosphatidylethanolamine) . 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. As a control, cells were treated with LIPOFECTIN™ without oligonucleotide under the same conditions and for the same times as the oligonucleotide-treated samples. After 4 hours at 37°C, the medium was replaced with fresh DMEM containing 10% serum. The cells were allowed to recover for 18 hours. Total cellular RNA was then extracted in guanidinium, subject to gel electrophoresis and transferred to a filter according to techniques known in the art (see, e.g., Chapter 7 In : Molecular Cloning: A Labora tory Manual , 2nd Ed., Sambrook et al . , eds., pages 7.1-7.87, and Short Protocols in Molecular Biology, 2nd Ed., Ausubel et al . , eds., John Wiley & Sons, New York, 1992, pages 2-24 to 2-30 and 4-14 to 4-29) . Filters were typically hybridized overnight to a probe specific for the particular JNK gene of interest in hybridization buffer (25 mM KP04, pH 7.4; 5x SSC; 5x
Denhardt's solution, 100 μg/ml Salmon sperm DNA and 50% formamide) (Alahari et al . , Nucl . Acids Res . , 1993, 21 , 4079) . This was followed by two washes with lx SSC, 0.1%SDS and two washes with 0.25x SSC, 0.1% SDS. Hybridizing bands were visualized by exposure to X-OMAT AR film and quantitated using a PHOSPHORIMAGER™ essentially according to the manufacturer' s instructions (Molecular Dynamics, Sunnyvale, CA) .
Western Assays: A549 cells were grown and treated with oligonucleotides as described above. Cells were lysed, and protein extracts were electrophoresed (SDS-PAGE) and transferred to nitrocellulose filters by means known in the art (see, e.g., Chapter 18 In : Molecular Cloning: A Labora tory Manual , 2nd Ed., Sambrook et al . , eds., pages 18.34, 18.47-18.54 and 18.60-18.75)). The amount of each JNK protein was determined by using a primary antibody that specifically recognizes the appropriate JNK protein. The primary antibodies specific for each JNK protein are described in the appropriate Examples. The primary antibodies were detected by means well known in the art (see, e.g., Short Protocols in Molecular Biology, 2nd Ed., Ausubel et al . , eds., John Wiley & Sons, New York, 1992, pages 10-33 to 10-35; and Chapter 18 In : Molecular Cloning: A Labora tory Manual , 2nd Ed., Sambrook et al . , eds., pages 18.1-18.75 and 18.86-18.88) and quantitated using a PHOSPHORIMAGER™ essentially according to the manufacturer's instructions (Molecular Dynamics, Sunnyvale, CA) . Levels of JNK proteins can also be quantitated by measuring the level of their corresponding kinase activity. Such kinase assays can be done in gels in si tu (Hibi et al . , Genes & Dev. , 1993, 7, 2135) or after immunoprecipitation from cellular extracts (Derijard et al . , Cell , 1994, 76, 1025). Substrates and/or kits for such assays are commercially available from, for example, Upstate Biotechnology, Inc. (Lake Placid, NY) , New England Biolabs, Inc., (Beverly, MA) and Calbiochem-Novabiochem Biosciences, Inc., (La Jolla, CA) .
Example 3 : Oligonucleotide-Mediated Inhibition of JNKl Expression A. JNKl oligonucleotide sequences: Table 1 lists the nucleotide sequences of a set of oligonucleotides designed to specifically hybridize to JNKl mRNAs and their corresponding ISIS and SEQ ID numbers. The nucleotide coordinates of the target gene, JNKl , and gene target regions are also included. The nucleotide co-ordinates are derived from GenBank accession No. L26318, locus name "HUMJNK1" (see also Figure 1(A) of Derijard et al . , Cell , 1994, 76, 1025) . The abbreviations for gene target regions are as follows: 5'-UTR, 5' untranslated region; tIR, translation initiation region; ORF, open reading frame; 3'-UTR, 3' untranslated region. The nucleotides of the oligonucleotides whose sequences are presented in Table 1 are connected by phosphorothioate linkages and are unmodified at the 2' position (i.e., 2 ' -deoxy) . It should be noted that the oligonucleotide target co-ordinate positions and gene target regions may vary within RNAs encoding related isoforms of JNKl (see subsection G, below) .
In addition to hybridizing to human JNKl mRNAs, the full oligonucleotide sequences of ISIS Nos. 12548 (SEQ ID NO: 17) and 12551 (SEQ ID NO: 20) hybridize to the 5' ends of mRNAs from Ra ttus norvegicus that encode a stress- activated protein kinase named "p54γ" (Kyriakis et al . , Na ture, 1994, 369, 156). Specifically, ISIS 12548 (SEQ ID NO: 17) hybridizes to bases 498-517 of GenBank accession No. L27129, locus name "RATSAPKD, " and ISIS 12551 (SEQ ID NO: 20) hybridizes to bases 803-822 of the same sequence. These oligonucleotides are thus preferred embodiments of the invention for investigating the role of the p54γ protein kinase in rat in vi tro, i . e . , in cultured cells or tissues derived from whole animals, or in vivo.
B. JNKl-specific probes: In initial screenings of a set of oligonucleotides derived from the JNKl sequence (Table 2) for biological activity, a cDNA clone of JNKl (Derijard et al . , Cell , 1994, 76, 1025) was radiolabeled and used as a JNKl-specific probe in Northern blots. Alternatively, however, one or more of the oligonucleotides of Table 1 is detectably labeled and used as a JNKl- specific probe.
TABLE 1 Nucleotide Sequences of JNKl Oligonucleotides
Figure imgf000067_0001
Figure imgf000068_0001
Figure imgf000069_0001
C. Activities of JNKl oligonucleotides: The data from screening a set of JNKl-specific phosphorothioate oligonucleotides (Table 2) indicate the following results. Oligonucleotides showing activity in this assay, as reflected by levels of inhibition from > about 50% to about 100% of JNKl mRNA levels, include ISIS Nos. 11982, 11983, 12463, 12464, 12538, 12539, 12548, 12549, 12550, 12552, 12553, 12554, 12555, 12556 and 12557 (SEQ ID NOS: 5, 6, 13, 14, 15, 16, 17, 18, 19, 21, 22, 23, 24, 25 and 26, respectively) . These oligonucleotides are thus preferred embodiments of the invention for modulating JNKl expression. Oligonucleotides showing levels of inhibition of from > about 80% to about 100% of JNKl mRNAs in this assay, include ISIS Nos. 11982, 12539, 12548, 12554 and 12464 (SEQ ID NOS: 5, 16, 17 and 23, respectively) . These oligonucleotides are thus more preferred embodiments of the invention for modulating JNKl expression.
The time course of inhibition of JNKl mRNA expression by ISIS 12539 (SEQ ID NO: 16) is shown in Table 3. Following the 4 hour treatment with ISIS 12539, the level of inhibition of JNKl was greater than about 85% (t=0 h) , rose to about 95% inhibition at t=4h, and subsequently remained at greater than or equal to about 80% (t=12 and 48 h) or 60% (t=72 h) . TABLE 2 Activities of JNKl Oligonucleotides
Figure imgf000071_0001
Figure imgf000072_0001
TABLE 3
Time Course of Response to JNKl Antisense Oligonucleotides (ASOs)
Figure imgf000073_0001
D. Additional JNKl oligonucleotides: The results for JNKl-specific oligonucleotides (Table 2) indicate that one of the most active phosphorothioate oligonucleotides for modulating JNKl expression is ISIS 12539 (SEQ ID NO: 16) . As detailed in Table 4, additional oligonucleotides based on this oligonucleotide were designed to confirm and extend the findings described above.
Oligonucleotides ISIS Nos. 14320 (SEQ ID NO: 27) and 14321 (SEQ ID NO: 28) are 2 ' -deoxy-phosphorothioate sense strand and scrambled controls for ISIS 12539 (SEQ ID NO: 16), respectively. ISIS Nos. 15346 and 15347 are "gapmers" corresponding to ISIS 12539; both have 2 ' -methoxyethoxy "wings" (having phosphorothioate linkages in the case of ISIS 15346 and phosphodiester linkages in the case of ISIS 15347) and a central 2 ' -deoxy "gap" designed to support RNaseH activity on the target mRNA molecule. Similarly, ISIS Nos. 15348 to 15350 are "wingmers" corresponding to ISIS 12539 and have a 5' or 3' 2 ' -methoxyethoxy RNaseH- refractory "wing" and a 3' or 5' (respectively) 2 ' -deoxy "wing" designed to support RNaseH activity on the target JNKl mRNA.
The chemically modified derivatives of ISIS 12539 (SEQ ID NO: 16) were tested in the Northern assay described herein at concentrations of 100 and 400 nM, and the data (Table 5) indicate the following results. At 400 nM, relative to the 2 ' -unmodified oligonucleotide ISIS 12539, both "gapmers" (ISIS Nos. 15346 and 15347) effected inhibition of JNKl mRNA expression up to at least about 88% inhibition. Similarly, the four "wingmers" (ISIS Nos.
15348 to 15351) effected inhibition of JNKl expression of up to at least about 60 to 70% inhibition.
TABLE 4 Chemically Modified JNKl Oligonucleotides
Figure imgf000075_0001
*Emboldened residues, 2 ' -methoxyethoxy- residues (others are 2 '-deoxy-) including "C" residues, 5-methyl-cytosines; "°", phosphodiester linkage; "s", phosphorothioate linkage. "£" residues, 2 ' -deoxy 5-methylcytosine residues;
TABLE 5
Activity of Chemically Modified JNKl Antisense
Oligonucleotides
Figure imgf000076_0001
Figure imgf000077_0001
Abbreviations: P=0, phosphodiester linkage; P=S, phosphorothioate linkage; MOE, methoxyethoxy-.
Ξ . Dose- and sequence-dependent response to JNKl oligonucleotides: In order to demonstrate a dose-dependent response to ISIS 12539 (SEQ ID NO: 16), different concentrations (i.e., 50, 100, 200 and 400 nM) of ISIS 12539 were tested for their effect on JNKl mRNA levels in A549 cells (Table 6) . In addition, two control oligonucleotides (ISIS 14320, SEQ ID NO: 27, sense control, and ISIS 14321, SEQ ID NO: 28, scrambled control; see also Table 4) were also applied to A549 cells in order to demonstrate the specificity of ISIS 12539. The results (Table 6) demonstrate that the response of A549 cells to ISIS 12539 is dependent on dose in an approximately linear fashion. In contrast, neither of the control oligonucleotides effect any consistent response on JNKl mRNA levels.
F. Western Assays: In order to assess the effect of oligonucleotides targeted to JNKl mRNAs on JNKl protein levels, Western assays were performed essentially as described above in Example 2, with the following exception(s) and/or modification (s) . A primary antibody that specifically binds to JNKl (catalog No. sc-474-G) was purchased from Santa Cruz Biotechnology, Inc. (Santa Cruz, CA; other JNKl-specific antibodies are available from StressGen Biotechnologies, Inc., Victoria, BC, Canada; and Research Diagnostics, Inc., Flanders, NJ) . In this experiment, cells were grown and treated with oligonucleotide at 300 nM for the initial 20 hours and then at 200 nM for 4 hours. At t=48 h, aliquots were removed for Northern and Western analyses, and fresh media was added to the cells. Aliquots for analysis were also taken at t=72 h. The samples from t=48 h and t=72 h were analyzed using the Northern and Western assays described above.
TABLE 6
Dose-Dependent Responses to JNKl Antisense Oligonucleotides
Figure imgf000078_0001
The data (Table 7) indicate the following results. In this assay, at t=48 h, oligonucleotides showing a level of mRNA % inhibition from > about 70% to about 100% include ISIS Nos. 12539 (phosphorothioate linkages), 15346 and 15347 ("gapmers"), and 15348 and 15351 (5' "wingmers") (SEQ ID NO: 16) . Oligonucleotides showing levels of mRNA inhibition of from > about 90% to about 100% of JNKl mRNAs in this assay include ISIS Nos. 12539, 15345 AND 15346 (SEQ ID NO: 16) . The oligonucleotides tested showed approximately parallel levels of JNKl protein inhibition; ISIS Nos. 12539, 15346-15348 and 15351 effected levels of protein inhibition > about 40%, and ISIS Nos. 12539, 15346 and 15347 effected levels of protein inhibition >. about
J J"6. At t=72 h, oligonucleotides showing a level of mRNA % inhibition from > about 70% to about 100% include ISIS Nos. 12539 (phosphorothioate linkages) , 15346 and 15347 ("gapmers"), and 15348 (5' "wingmers") (SEQ ID NO: 16). Oligonucleotides showing levels of mRNA inhibition of from > about 90% to about 100% of JNKl mRNAs at this point in the assay include ISIS Nos. 12539 and 15346 (SEQ ID NO: 16) . Overall, the oligonucleotides tested showed higher levels of JNKl protein inhibition at this point in the assay. With the exception of the fully 2 ' -methoxyethoxy- modified ISIS 15345, all of the oligonucleotides in Table 7 effect > about 40% protein inhibition. ISIS Nos. 12539, 15346-15348 and 15351 effected levels of protein inhibition > about 60%, and ISIS Nos. 12539, 15346 and 15347 effected levels of protein inhibition >. about 70%. TABLE 7
Modulation of JNKl mRNA and JNKl Protein Levels by Modified JNKl Antisense Oligonucleotides
Figure imgf000080_0001
SEQ ID RNA RNA % Protein Protein %
ISIS # NO: % Control Inhibition % Control Inhibition
t = 72 h
s
Figure imgf000081_0002
Figure imgf000081_0001
G. Oligonucleotides specific for JNKl isoforms :
Subsequent to the initial descriptions of JNKl (Derijard et al . , Cell , 1994, 76, 1025), cDNAs encoding related isoforms of JNKl were cloned and their nucleotide sequences determined (Gupta et al . , EMBO Journal , 1996, 15, 2760). In addition to JNKl-αl (GenBank accession No. L26318, locus name "HUMJNKl"), which encodes a polypeptide having an amino acid sequence identical to that of JNKl, the additional isoforms include JNKl-α2 (GenBank accession No. U34822, locus name "HSU34822") , JNKl-βl (GenBank accession No. U35004, locus name "HSU35004") and JNKl-β2 (GenBank accession No. U35005, locus name "HSU35005") . The four isoforms of JNKl, which probably arise from alternative mRNA splicing, may each interact with different transcription factors or sets of transcription factors
(Gupta et al . , EMBO Journal , 1996, 15, 2760). As detailed below, the oligonucleotides of the invention are specific for certain members or sets of these isoforms of JNKl.
In the ORFs of mRNAs encoding JNKl/JNKl-αl and JNK1- α2, nucleotides (nt) 631-665 of JNKl/JNKl-αl (Genbank accession No. L26318) and nt 625-659 of JNKl-α2 (Genbank accession No. U34822) have the sequence shown below as SEQ ID NO: 63, whereas, in the ORFs of mRNAs encoding JNKl-βl and JNKl-β2, nt 631-665 of JNKl-βl (GenBank accession No. U35004) and nt 626-660 of JNKl-β2 (GenBank accession No. U35005) have the sequence shown below as SEQ ID NO: 64. For purposes of illustration, SEQ ID NOS: 63 and 64 are shown aligned with each other (vertical marks, "I," indicate bases that are identical in both sequences) :
5 ' -AACGTGGATTTATGGTCTGTGGGGTGCATTATGGG SEQ ID NO: 63 MM! II I Mill II II II I I II I I I II 5 ' -AACGTTGACATTTGGTCAGTTGGGTGCATCATGGG SEQ ID NO: 64 Due to this divergence between the a and b JNKl isoforms, antisense oligonucleotides derived from the reverse complement of SEQ ID NO: 63 (i.e., SEQ ID NO: 65, see below) can be used to modulate the expression of JNK1/JNK1- αl and JNKl-α2 without significantly effecting the expression of JNKl-βl and JNKl-β2. In like fashion, antisense oligonucleotides derived from the reverse complement of SEQ ID NO: 64 (i.e., SEQ ID NO: 66, see below) can be selected and used to modulate the expression of JNKl-βl and JNKl-β2 without significantly effecting the expression of JNKl/JNKl-αl and JNKl-α2. As an example, an oligonucleotide having a sequence derived from SEQ ID NO: 65 but not to SEQ ID NO: 66 is specifically hybridizable to mRNAs encoding JNKl/JNKl-αl and JNKl-α2 but not to those encoding JNKl-βl and JNKl-β2:
5 ' -CCCATAATGCACCCCACAGACCATAAATCCACGTT SEQ ID NO: 65
I I II I I I I I I I II II MM I I II II I I I 5 ' -CCCATGATGCACCCAACTGACCAAATGTCAACGTT SEQ ID NO: 66
As a further example, in the ORFs of mRNAs encoding JNKl/JNKl-αl and JNKl-α2, nt 668-711 of JNKl/JNKl-αl
(Genbank accession No. L26318) and nt 662-705 of JNKl-α2 (Genbank accession No. U34822) have the sequence shown below as SEQ ID NO: 67, whereas, in the ORFs of mRNAs encoding JNKl-βl and JNKl-β2, nt 668-711 of JNKl-βl (GenBank accession No. U35004) and nt 663-706 of JNKl-β2 (GenBank accession No. U35005) have the sequence shown below as SEQ ID NO: 68. For purposes of illustration, SEQ ID NOS: 67 and 68 are shown aligned with each other as follows :
5 ' -AAATGGTTTGCCACAAAATCCTCTTTCCAGGAAGGGACTATATT SEQ ID NO: 67 II I II I I I I I I II I I I II Mill
5 ' -AAATGATCAAAGGTGGTGTTTTGTTCCCAGGTACAGATCATATT SEQ ID NO: 68
Due to this divergence between the a and b JNKl isoforms, antisense oligonucleotides derived from the reverse complement of SEQ ID NO: 67 (i.e., SEQ ID NO: 69, see below) are specifically hybridizable to mRNAs encoding, and may be selected and used to modulate the expression of, JNKl/JNKl-αl and JNKl-α2 without significantly effecting the expression of JNKl-βl and JNKl-β2. In like fashion, antisense oligonucleotides derived from the reverse complement of SEQ ID NO: 68 (i.e., SEQ ID NO: 70, see below) are specifically hybridizable to mRNAs encoding, and may be selected and used to modulate the expression of, can be selected and used to modulate the expression of JNKl-βl and JNKl-β2 without significantly effecting the expression of JNKl/JNKl-αl and JNKl-α2 :
5 ' -AATATAGTCCCTTCCTGGAAAGAGGATTTTGTGGCAAACCATTT SEQ ID NO: 69
II II I II I II I II II I I I II II I 5 ' -AATATGATCTGTACCTGGGAACAAAACACCACCTTTGATCATTT SEQ ID NO: 70
In the case of the carboxyl terminal portion of the JNKl isoforms, JNKl/JNKl-αl shares identity with JNKl-βl; similarly, JNKl-α2 and JNKl-β2 have identical carboxy terminal portions. The substantial differences in the amino acid sequences of these isoforms (5 amino acids in JNKl/JNKl-αl and JNKl-βl are replaced with 48 amino acids in JNKl-α2 and JNKl-β2) result from a slight difference in nucleotide sequence that shifts the reading frame. Specifically, in the ORFs of mRNAs encoding JNKl/JNKl-αl and JNKl-βl, nt 1144-1175 of JNKl/JNKl-αl (Genbank accession No. L26318) and JNKl-βl (Genbank accession No. U35004) have the sequence shown below as SEQ ID NO: 71, whereas, in the ORFs of mRNAs encoding JNKl-α2 and JNKl-β2, nt 1138-1164 of JNKl-α2 (GenBank accession No. U34822) and nt 1139-1165 of JNKl-β2 (GenBank accession No. U35005) have the sequence shown below as SEQ ID NO: 72. For purposes of illustration, SEQ ID NOS: 71 and 72 are shown aligned with each other (dashes, "-," indicate bases that are absent in the indicated sequence, and emboldened bases indicate the stop codon for the JNKl/JNKl-αl and JNKl-βl ORFs) :
5 ' -CCCTCTCCTTTAGCACAGGTGCAGCAGTGATC SEQ ID NO: 71
II II I M I II II I II II I I II II I I I I 5 ' -CCCTCTCCTTTAG GTGCAGCAGTGATC SEQ ID NO: 72
Due to this divergence between the JNKl isoforms, antisense oligonucleotides derived from the reverse complement of SEQ ID NO: 71 (i.e., SEQ ID NO: 73, see below) are specifically hybridizable to mRNAs encoding, and may be selected and used to modulate the expression of, JNKl/JNKl-αl and JNKl- βl without significantly effecting the expression of JNK1- α2 and JNKl-β2. In like fashion, antisense oligonucleotides derived from the reverse complement of SEQ ID NO: 72 (i.e., SEQ ID NO: 74, see below) are specifically hybridizable to mRNAs encoding, and may be selected and used to modulate the expression of, JNKl-α2 and JNKl-β2 without significantly effecting the expression of JNKl/JNKl-αl and JNKl-βl: 5 ' -GATCACTGCTGCACCTGTGCTAAAGGAGAGGG SEQ ID NO: 73
II II II I II II II I II II II II II II I 5 ' -GATCACTGCTGCAC CTAAAGGAGAGGG SEQ ID NO: 74
In preferred embodiments, such isoform-specific oligonucleotides such as are described above are methoxyethoxy "gapmers" or "wingmers" in which the RNase H- sensitive "gap" or "wing" is positioned so as to overlap a region of nonidentity in the above antisense sequences, i.e., SEQ ID NOS: 65, 66, 69, 70, 73 and 74.
Example 4: Oligonucleotide-Mediated Inhibition of JNK2 Expression A. JNK2 oligonucleotide sequences: Table 8 lists the nucleotide sequences of oligonucleotides designed to specifically hybridize to JNK2 mRNAs and the corresponding ISIS and SEQ ID numbers thereof. The target gene nucleotide co-ordinates and gene target region are also included. The nucleotide co-ordinates are derived from GenBank accession No. L31951, locus name "HUMJNK2" (see also Figure 1(A) of Sluss et al . , Mol . Cel . Biol . , 1994, 1 4 , 8376, and Kallunki et a l . , Genes & Developmen t , 1994, 8, 2996) . The abbreviations for gene target regions are as follows: 5 ' -UTR, 5' untranslated region; tIR, translation initiation region; ORF, open reading frame; 3' -UTR, 3' untranslated region. The nucleotides of the oligonucleotides whose sequences are presented in Table 8 are connected by phosphorothioate linkages and are unmodified at the 2' position (i.e., 2-deoxy) . It should be noted that the oligonucleotide target co-ordinate positions and gene target regions may vary within mRNAs encoding related isoforms of JNK2 (see subsection G, below) . In addition to hybridizing to human JNK2 mRNAs, the full oligonucleotide sequence of ISIS No. 12562 (SEQ ID NO: 33) hybridizes to the ORF of mRNAs from Ra ttus norvegicus that encode a stress-activated protein kinase named "p54α2" (Kyriakis et al . , Na ture, 1994, 369, 156). Specifically, ISIS 12562 (SEQ ID NO: 33) hybridizes to bases 649-668 of GenBank accession No. L27112, locus name "RATSAPKB." This oligonucleotide is thus a preferred embodiment of the invention for investigating the role of the p54α2 protein kinase in rat in vi tro, i . e . , in cultured cells or tissues derived from whole animals, or in vivo .
B. JNK2-specific probes: In initial screenings of a set of oligonucleotides derived from the JNK2 sequence (Table 9) for biological activity, a cDNA clone of JNK2 (Kallunki et al . , Genes & Development , 1994, 8, 2996) was radiolabeled and used as a JNK2-specific probe in Northern blots. Alternatively, however, one or more of the oligonucleotides of Table 8 is detectably labeled and used as a JNK2-specific probe. C. Activities of JNK2 oligonucleotides: The data from screening a set of JNK2-specific phosphorothioate oligonucleotides (Table 9) indicate the following results. Oligonucleotides showing activity in this assay, as reflected by levels of inhibition from >. about 50% to about 100% of JNK2 mRNA levels, include ISIS Nos. 12558, 12559, 12560, 12563, 12564, 12565, 12566, 12567, 12568, 12569 and 12570 (SEQ ID NOS: 29, 30, 31, 34, 35, 36, 37, 38, 39, 40 and 41, respectively) . These oligonucleotides are thus preferred embodiments of the invention for modulating JNK2 expression. Oligonucleotides showing levels of inhibition of from > about 80% to about 100% of JNK2 mRNAs in this assay, include ISIS Nos. 12558, 12560, 12565, 12567, 12568 and 12569 (SEQ ID NOS: 29, 31, 36, 38, 39 and 40, respectively) . These oligonucleotides are thus more preferred embodiments of the invention for modulating JNK2 expression.
The time course of inhibition of JNK2 mRNA expression by ISIS 12560 (SEQ ID NO: 31) is shown in Table 10.
Following the 4 hour treatment with ISIS 12560, the level of inhibition of JNK2 was greater than or equal to about 80% for at least about 12 hours and greater than or equal to about 60% up to at least about t=48 h.
TABLE 8
Nucleotide Sequences of JNK2 Oligonucleotides
Figure imgf000089_0001
Figure imgf000090_0001
oo oo
TABLE 9
Activities of JNK2 Oligonucleotides
Figure imgf000091_0001
TABLE 10
Time Course of Response to JNK2 Antisense
Oligonucleotides (ASOs)
Figure imgf000092_0001
D. Additional JNK2 oligonucleotides: The results for JNK2-specific oligonucleotides (Table 9) indicate that one of the most active phosphorothioate oligonucleotides for modulating JNK2 expression is ISIS 12560 (SEQ ID NO: 31) . As detailed in Table 11, additional oligonucleotides based on this oligonucleotide were designed to confirm and extend the findings described above. Oligonucleotides ISIS Nos. 14318 (SEQ ID NO: 42) and 14319 (SEQ ID NO: 43) are 2 ' -deoxy-phosphorothioate sense strand and scrambled controls for ISIS 12560 (SEQ ID NO: 31), respectively. ISIS Nos. 15353 and 15354 are "gapmers" corresponding to ISIS 12560; both have 2 ' -methoxyethoxy "wings" (having phosphorothioate linkages in the case of ISIS 15353 and phosphodiester linkages in the case of ISIS 15354) and a central 2 ' -deoxy "gap" designed to support RNaseH activity on the target mRNA molecule. Similarly, ISIS Nos. 15355 to 15358 are "wingmers" corresponding to ISIS 12560 and have a 5' or 3 ' 2 ' -methoxyethoxy RNaseH- refractory "wing" and a 3' or 5 ' (respectively) 2-deoxy "wing" designed to support RNaseH activity on the target JNK2 mRNA. The chemically modified derivatives of ISIS 12560
(SEQ ID NO: 31) were tested in the Northern assay described herein at concentrations of 100 and 400 nM, and the data (Table 12) indicate the following results. At 400 nM, relative to the 2 ' -unmodified oligonucleotide ISIS 12560, both "gapmers" (ISIS Nos. 15353 and 15354) effected inhibition of JNK2 mRNA expression up to at least about 80% inhibition. Similarly, the four "wingmers" (ISIS Nos. 15355 to 15358) effected inhibition of JNK2 expression of up to at least about 70 to 90% inhibition. E. Dose- and sequence-dependent response to JNK2 oligonucleotides: In order to demonstrate a dose-dependent response to ISIS 12560 (SEQ ID NO: 31), different concentrations (i.e., 50, 100, 200 and 400 nM) of ISIS 12560 were tested for their effect on JNK2 mRNA levels in A549 cells (Table 13) . In addition, two control oligonucleotides (ISIS 14318, SEQ ID NO: 42, sense control, and ISIS 14319, SEQ ID NO: 43, scrambled control; see also Table 11) were also applied to A549 cells in order to demonstrate the specificity of ISIS 12560. The results (Table 12) demonstrate that the response of A549 cells to ISIS 12539 is dependent on dose in an approximately linear fashion. In contrast, neither of the control oligonucleotides effect any consistent response on JNK2 mRNA levels.
TABLE 11
Chemically Modified JNK2 Oligonucleotides
Figure imgf000094_0001
Figure imgf000095_0001
Emboldened residues, 2 ' -methoxyethoxy- residues (others are 2 '-deoxy-) including "C" residues, 5-methyl-cytosines ; "°", phosphodiester linkage; "s", phosphorothioate linkage. "£" residues, 2 ' -deoxy 5-methylcytosine residues;
TABLE 12 Activity of Chemically Modified JNK2 Antisense
Oligonucleotides
Figure imgf000095_0002
Figure imgf000096_0001
TABLE 13
Dose-Dependent Responses to JNK2 Antisense Oligonucleotides
Figure imgf000096_0002
Figure imgf000097_0001
F. Western Assays: In order to assess the effect of oligonucleotides targeted to JNK2 mRNAs on JNK2 protein levels, Western assays are performed essentially as described above in Examples 2 and 3. A primary antibody that specifically binds to JNK2 is purchased from, for example, Santa Cruz Biotechnology, Inc., Santa Cruz, CA; Upstate Biotechnology, Inc., Lake Placid, NY; StressGen Biotechnologies, Inc., Victoria, BC, Canada; or Research Diagnostics, Inc., Flanders, NJ.
G . Oligonucleotides specific for JNK2 isoforms : Subsequent to the initial descriptions of JNK2 (Sluss et al . , Mol . Cel . Biol . , 1994, 14 , 8376; Kallunki et al . , Genes & Development, 1994, 8, 2996; GenBank accession No. HSU09759, locus name "U09759"), cDNAs encoding related isoforms of JNK2 were cloned and their nucleotide sequences determined (Gupta et al . , EMBO Journal , 1996, 15, 2760).
In addition to JNK2-α2 (GenBank accession No. L31951, locus name "HUMJNK2"), which encodes a polypeptide having an amino acid sequence identical to that of JNK2, the additional isoforms include JNK2-αl (GenBank accession No. U34821, locus name "HSU34821") , JNK2-βl (GenBank accession No. U35002, locus name "HSU35002") and JNK2-β2 (GenBank accession No. U35003, locus name "HSU35003") . The four isoforms of JNK2, which probably arise from alternative mRNA splicing, may each interact with different transcription factors or sets of transcription factors (Gupta et al . , EMBO Journal , 1996, 15, 2760). As detailed below, the oligonucleotides of the invention are specific for certain members or sets of these isoforms of JNK2. In the ORFs of mRNAs encoding JNK2/JNK2-α2 and JNK2- αl, nucleotides (nt) 689-748 of JNK2/JNK2-α2 (GenBank accession No. L31951) and nt 675-734 of JNK2-αl (GenBank accession No. U34821) have the sequence shown below as SEQ ID NO: 75, whereas, in the ORFs of mRNAs encoding JNK2-βl and JNK2-β2, nt 653-712 of JNK2-βl (GenBank accession No. U35002) and nt 665-724 of JNK2-β2 (GenBank accession No. U35003) have the sequence shown below as SEQ ID NO: 76. For purposes of illustration, SEQ ID NOS: 75 and 76 are shown aligned with each other (vertical marks, "|," indicate bases that are identical in both sequences) :
SEQ ID NO: 75
5 ' -GTGGGTTGCATCATGGGAGAGCTGGTGAAAGGTTGTGTGATATTCCAAGGCACTGACCAT
I I II I I I I I I I I II I I I I I I I I II I I II I II I I I I I I
5 ' -GTCGGGTGCATCATGGCAGAAATGGTCCTCCATAAAGTCCTGTTCCCGGGAAGAGACTAT SEQ ID NO: 76
Due to this divergence between the a and b JNK2 isoforms, antisense oligonucleotides derived from the reverse complement of SEQ ID NO: 75 (i.e., SEQ ID NO: 77, see below) are specifically hybridizable to, and may be selected and used to modulate the expression of, JNK2/JNK2- α2 and JNK2-αl without significantly effecting the expression of JNKl-βl and JNKl-β2. In like fashion, antisense oligonucleotides derived from the reverse complement of SEQ ID NO: 76 (i.e., SEQ ID NO: 78, see below) are specifically hybridizable to, and may be selected and used to modulate the expression of, JNK2-βl and JNK2-β2 without significantly effecting the expression of JNK2/JNK2-α2 and JNK2-αl. As an example, an oligonucleotide having a sequence derived from SEQ ID NO: 77 but not from SEQ ID NO : 78 is specifically hybridi zable to , mRNAs encoding JNKl/JNKl-αl and JNKl-α2 but not to those encoding JNK2-βl and JNK2-β2 :
SEQ I D NO : 77 5 ' -ATGGTCAGTGCCTTGGAATATCACACAACCTTTCACCAGCTCTCCCATGATGCAACCCAC I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I I
5 ' -ATAGTCTCTTCCCGGGAACAGGACTTTATGGAGGACCATTTCTGCCATGATGCACCCGAC
SEQ I D NO : 78
In the case of the carboxyl terminal portion of the JNK2 isoforms, JNK2/JNK2-α2 shares identity with JNKl-β2; similarly, JNK2-αl and JNK2-βl have identical carboxy terminal portions. The substantial differences in the amino acid sequences of these isoforms (5 amino acids in JNK2-α2 and JNK2-β2 are replaced with 47 amino acids in JNK2/JNK2-α2 and JNK2-β2) result from a slight difference in nucleotide sequence that shifts the reading frame. Specifically, in the ORFs of mRNAs encoding JNK2-αl and JNKl-βl, nt 1164-1198 of JNK2-αl (GenBank accession No. U34821) and nt 1142-1176 of JNK2-βl (GenBank accession No. U35002) have the sequence shown below as SEQ ID NO: 79, whereas, in the ORFs of mRNAs encoding JNK2/JNK2-α2 and JNK2-β2, nt 1178-1207 of JNK2/JNK2-α2 (GenBank accession No. L31951) and nt 1154-1183 of JNK2-β2 (GenBank accession No. U35003) have the sequence shown below as SEQ ID NO: 80. For purposes of illustration, SEQ ID NOS: 79 and 80 are shown aligned with each other (dashes, "-," indicate bases that are absent in the indicated sequence, and emboldened bases indicate the stop codon for the JNK2-αl and JNK2-βl ORFs) : 5 ' -GATCAGCCTTCAGCACAGATGCAGCAGTAAGTAGC SEQ ID NO: 79
I II I II I I! II II I II II II I I II I II I I I 5 ' -GATCAGCCTTCAG ATGCAGCAGTAAGTAGC SEQ ID NO: 80
Due to this divergence between the JNK2 isoforms, antisense oligonucleotides derived from the reverse complement of SEQ ID NO: 79 (i.e., SEQ ID NO: 81, see below) are specifically hybridizable to, and may be selected and used to modulate the expression of, mRNAs encoding JNK2-αl and JNK2-βl without significantly effecting the expression of JNK2/JNK2-α2 and JNK2-β2. In like fashion, antisense oligonucleotides derived from the reverse complement of SEQ ID NO: 80 (i.e., SEQ ID NO: 82, see below) are specifically hybridizable to, and may be selected and used to modulate the expression of, mRNAs encoding JNK2/JNK2-α2 and JNK2-β2 without significantly effecting the expression of JNK2-αl and JNK2-βl. As an example, ISIS 12564 (SEQ ID NO: 35) corresponds to SEQ ID NO: 82 but not to SEQ ID NO: 81, and is thus specifically hybridizable to, and may be used to modulate the expression of, mRNAs encoding JNK2/JNK2-α2 and JNK2-β2 but not those encoding JNK2-αl and JNK2-αl:
5 ' -GCTACTTACTGCTGCATCTGTGCTGAAGGCTGATC SEQ ID NO: 81
M II II I II I II II II I I I M II II I I II 5 ' -GCTACTTACTGCTGCAT CTGAAGGCTGATC SEQ ID NO: 82
I II I II I II I M I II M I I I 5'-CTGCTGCAT CTGAAGGCTGA SEQ ID NO: 35
In preferred embodiments, such isoform-specific oligonucleotides such as are described above are methoxyethoxy "gapmers" or "wingmers" in which the RNase H- sensitive "gap" or "wing" is positioned so as to overlap a region of nonidentity in the above antisense sequences, i.e., SEQ ID NOS: 77, 78, 81 and 82.
Example 5 : Oligonucleotide-Mediated Inhibition of JNK3 Expression
A. JNK3 oligonucleotide sequences: Table 14 lists the nucleotide sequences of oligonucleotides designed to specifically hybridize to JNK3 mRNAs and the corresponding ISIS and SEQ ID numbers thereof. The target gene nucleotide co-ordinates and gene target region are also included. The nucleotide co-ordinates are derived from GenBank accession No. U07620, locus name "HSU07620" see also Figure 4(A) of Mohit et al . , Neuron, 1994, 14, 67). The abbreviations for gene target regions are as follows: 5' -UTR, 5' untranslated region; tIR, translation initiation region; ORF, open reading frame; 3 ' -UTR, 3' untranslated region. It should be noted that the oligonucleotide target co-ordinate positions and gene target regions may vary within mRNAs encoding related isoforms of JNK3 (see subsection D, below) .
The nucleotides of the oligonucleotides whose sequences are presented in Table 14 are connected by phosphorothioate linkages and are "gapmers." Specifically, the six nucleotides of the 3' and 5' termini are 2'- methoxyethoxy- modified and are shown emboldened in Table 14, whereas the central eight nucleotides are unmodified at the 2' position (i.e., 2-deoxy) .
In addition to hybridizing to human JNK3 mRNAs, the full oligonucleotide sequences of ISIS Nos. 16692, 16693, 16703, 16704, 16705, 16707, and 16708 (SEQ ID NOS: 46, 47, 56, 57, 58, 60 and 61, respectively) specifically hybridize to mRNAs from Ra ttus norvegicus that encode a stress- activated protein kinase named "p54β" (Kyriakis et al . , Na ture, 1994, 369, 156; GenBank accession No. L27128, locus name "RATSAPKC." Furthermore, the full oligonucleotide sequences of 16692, 16693, 16695, 16703, 16704, 16705, 16707 and 16708 (SEQ ID NOS: 46, 47, 49, 56, 57, 58, 60 and 61, respectively) specifically hybridize to mRNAs from Mus musculus that encode a mitogen activated protein (MAP) kinase stress activated protein named the "p4593F12 SAP kinase" (Martin et al . , Brain Res . Mol . Brain Res . , 1996, 35, 47; GenBank accession No. L35236, locus name "MUSMAPK") . These oligonucleotides are thus preferred embodiments of the invention for investigating the role of the p54β and p4593F12 SAP protein kinases in rat or mouse, respectively, in vi tro, i . e . , in cultured cells or tissues derived from whole animals or in vivo . The target gene nucleotide co-ordinates and gene target regions for these oligonucleotides, as defined for these GenBank entries, are detailed in Table 15.
B. JNK3-specific probes: In initial screenings of a set of oligonucleotides derived from the JNK3 sequence for biological activity, a cDNA clone of JNK3 (Derijard et al . , Cell , 1994, 76, 1025) was radiolabeled and used as a JNK3- specific probe in Northern blots. Alternatively, however, one or more of the oligonucleotides of Table 14 is detectably labeled and used as a JNK3-specific probe.
C. Western Assays: In order to assess the effect of oligonucleotides targeted to JNK3 mRNAs on JNK3 protein levels, Western assays are performed essentially as described above in Examples 2 through 4. A primary antibody that specifically binds to JNK3 is purchased from, for example, Upstate Biotechnology, Inc. (Lake Placid, NY) , StressGen Biotechnologies Corp. (Victoria, BC, Canada), or New England Biolabs, Inc. (Beverly, MA) .
TABLE 14 Nucleotide Sequences of JNK3 Oligonucleotides
Figure imgf000103_0001
Figure imgf000104_0001
Figure imgf000104_0002
Emboldened residues are 2 ' -methoxyethoxy- modified .
TABLE 15
Rat and Mouse Gene Target Locations of JNK3 Oligonucleotides
o
Figure imgf000105_0001
^o-ordinates from GenBank Accession No. L27128, locus name "RATSAPKC. 2Co-ordinates from GenBank Accession No. L35236, locus name "MUSMAPK.'
D . Oligonucleotides specific for JNK3 isoforms : Two isoforms of JNK3 have been described. JNK3-αl was initially cloned and named "p493F12 kinase" by Mohit et al . { Neuron, 1995, 14, 67) . Subsequently, two cDNAs encoding related isoforms of JNK3 were cloned and their nucleotide sequences determined (Gupta et al . , EMBO Journal , 1996, 15, 2760) . The isoforms are named JNK3-αl (GenBank accession No. U34820, locus name "HSU34820") and JNK3-α2 (GenBank accession No. U34819, locus name "HSU34819") herein. The two isoforms of JNK3, which probably arise from alternative mRNA splicing, may each interact with different transcription factors or sets of transcription factors (Gupta et al . , EMBO Journal, 1996, 15, 2760). As detailed below, certain oligonucleotides of the invention are specific for each of these isoforms of JNK3.
JNK3-αl and JNK-α2 differ at their carboxyl terminal portions. The substantial differences in the amino acid sequences of these isoforms (5 amino acids in JNK3-αl are replaced with 47 amino acids in JNK3-α2) result from a slight difference in nucleotide sequence that shifts the reading frame. Specifically, in the ORF of mRNAs encoding JNK3-αl, nucleotides (nt) 1325-1362 of JNK3-αl (GenBank accession No. U34820) have the sequence shown below as SEQ ID NO: 83, whereas, in the ORF of mRNAs encoding JNK3-α2, nt 1301-1333 of JNK3-α2 (GenBank accession No. U34819) have the sequence shown below as SEQ ID NO: 84. For purposes of illustration, SEQ ID NOS: 83 and 202 are shown aligned with each other (vertical marks, "I," indicate bases that are identical in both sequences; dashes, "-," indicate bases that are absent in the indicated sequence; and emboldened bases indicate the stop codon for the JNK3-αl ORF) : 5 ' -GGACAGCCTTCTCCTTCAGCACAGGTGCAGCAGTGAAC SEQ ID NO: 83
I II II I II II II I II II II I I II I I II II II I I
5 ' -GGACAGCCTTCTCCTTCAG GTGCAGCAGTGAAC SEQ ID NO: 84
Due to this divergence between the JNK3 isoforms, antisense oligonucleotides derived from the reverse complement of SEQ ID NO: 83 (i.e., SEQ ID NO: 85, see below) are specifically hybridizable to mRNAs encoding, and may be selected and used to modulate the expression of JNK3-αl without significantly effecting the expression of JNK3-α2. In like fashion, antisense oligonucleotides derived from the reverse complement of SEQ ID NO: 84 (i.e., SEQ ID NO: 86, see below) are specifically hybridizable to mRNAs encoding, and may be selected and used to modulate the expression of JNK3-α2 without significantly effecting the expression of JNK3-0.1:
5 ' -GTTCACTGCTGCACCTGTGCTGAAGGAGAAGGCTGTCC SEQ ID NO: 85
II II II II II II I I II II I I I II I I II I I II I I
5 ' -GTTCACTGCTGCAC CTGAAGGAGAAGGCTGTCC SEQ ID NO: 86
In preferred embodiments, such isoform-specific oligonucleotides such as are described above are methoxyethoxy "gapmers" or "wingmers" in which the RNase H- sensitive "gap" or "wing" is positioned so as to overlap a region of nonidentity in the above antisense sequences, i.e., SEQ ID NOS: 85 and 86. E. Activities of JNK3 oligonucleotides: The JNK3- specific phosphorothioate, 2 ' -methoxyethoxy "gapmer" oligonucleotides (Table 14) were screened for their ability to affect JNK3 mRNA levels in SH-SY5Y cells (Biedler et al . , Cancer Res . , 1973, 33, 2643). SH-SY5Y cells express a variety of mitogen-activated protein kinases (MAPKs; see, e.g., Cheng et al . , J. Biol . Chem . , 1998, 273, 14560). Cells were grown in DMEM essentially as previously described (e.g., Singleton et al . , J. Biol . Chem . , 1996, 271, 31791; Jalava et al . , Cancer Res . , 1990, 50, 3422) and treated with oligonucleotides at a concentration of 200 nM as described in Example 2. Control cultures were treated with an aliquot of LIPOFECTIN™ that contained no oligonucleotide .
The results are shown in Table 16. Oligonucleotides showing levels of inhibition from >. about 45% to about 100% of JNK3 mRNA levels, include ISIS Nos. 16692, 16693, 16694, 16695, 16696, 16697, 16702, 16703, 16704, 16705 and 16706 (SEQ ID NOS:46, 47, 48, 49, 50, 51, 55, 56, 57, 58 and 59, respectively) . These oligonucleotides are preferred embodiments of the invention for modulating JNK3 expression. Oligonucleotides showing levels of inhibition of from > about 60% to about 100% of JNK3 mRNAs in this assay, wherein "about" indicates ±5%, include ISIS Nos. 16693, 16694, 16695, 16702, 16703, 16704 and 16705 (SEQ ID NOS:47, 48, 49, 55, 56, 57 and 58, respectively). These oligonucleotides are thus more preferred embodiments of the invention for modulating JNK3 expression.
TABLE 16: Activities of JNK3 Oligonucleotides
Figure imgf000108_0001
Figure imgf000109_0001
1 Cells treated with LIPOFECTIN™ only (no oligonucleotide) .
2 N.D., not determined.
Example 6: 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 (12-O-tetradecanoylphorbol 13-acetate) . ISIS 10582, an oligonucleotide targeted to c- jun, was evaluated for its ability to modulate MMP- 9 expression (see pending application Serial No. 08/837,201, filed April 14, 1997, attorney docket No. ISPH-0209. The results (Table 16) demonstrate that ISIS 10582 is able to completely inhibit the expression of MMP- 9 after induction with TPA.
TABLE 17
Effect of c -jun Oligonucleotide on MMP-9 Expression
Figure imgf000111_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. Because JNK proteins activate AP-1 by phosphorylating the N- terminal portion of the Jun subunit thereof, modulation of one or more JNK proteins by the oligonucleotides of the present disclosure will also modulate the expression of MMP family members and limit the metastatic ability of cancer cells .
Example 7 : Treatment of Human Tumors in Mice with
Oligonucleotides Targeted to JNK Proteins Approximately 5 x 106 breast adenocarcinoma cells (cell line MDA-MB-231; American Type Culture Collection, Richmond, VA, No. ATCC HTB-26) were implanted subcutaneously in the right inner thigh of nude mice (n=6 for each of three sets of mice) . Oligonucleotides ISIS 15346 (JNKl, SEQ ID NO:16) and 15353 (JNK2, SEQ ID N0:31) were suspended in saline and administered once daily to two sets of mice on the first day the tumor volume was about 100 mm3. A saline-only (0.9% NaCl) solution was given to a third set of animals as a control. Oligonucleotides were given by intravenous injection at a dosage of 25 g/kg.
Tumor size was measured and tumor volume was calculated on days 12, 19, 26 and 33 following tumor cell inoculation.
The results are shown in Table 18. Both 15346 (JNKl, SEQ ID NO: 16) and 15353 (JNK2, SEQ ID NO: 31) inhibited tumor growth compared to the saline control. Specifically, on days 26 and 33, the MDA-MB-231 tumors in animals that had been treated with the oligonucleotides had smaller volumes than the tumors in saline-treated animals, indicating that the oligonucleotides inhibited the growth of the tumors.
The antisense compounds of the invention are also tested for their ability to slow or eliminate the growth of xenografts resulting from, for example, human cervical epithelial carcinoma cells (HeLa cell line, ATCC No. ATCC CCL-2), human lung carcinoma cells (cell line A549, ATCC No. ATCC CCL-185), human adenocarcinoma cells (cell line SW480, ATCC No. ATCC CCL-228), human bladder carcinoma cells (cell line T24, ATCC No. HTB-4), human pancreatic - Ill
carcinoma cells (cell line MIA PaCa, ATCC No. CRL-1420) and human small cell carcinoma cells (cell line NCI-H69, ATCC HTB-119) . Xenografts resulting from these and other cell lines are established using essentially the same techniques as were used for the experiments using MDA-MB 231 cells.
TABLE 18: Response of MDA-MB-231 Tumors in Mice to
Oligonucleotides Targeted to JNKl and JNK2
Figure imgf000113_0001
Example 8: Oligonucleotides Targeted to Genes Encoding Rat JNK Proteins
In order to study the role of JNK proteins in animal models, oligonucleotides targeted to the genes encoding JNKl, JNK2 and JNK3 of Ra ttus norvegicus were prepared. These oligonucleotides are 2 ' -methoxyethoxy, phosphodiester / 2 '-hydroxyl, phosphorothioate / 2 ' -methoxyethoxy, phosphodiester "gapmers" in which every cytosine residue is 5-methylcytosine (m5c) . These antisense compounds were synthesized according to the methods of the disclosure. Certain of these oligonucleotides are additionally specifically hybridizable to JNK genes from other species as indicated herein. The oligonucleotides described in this Example were tested for their ability to modulate rat JNK mRNA levels essentially according to the methods described in the preceding Examples, with the exceptions that the cell line used was rat A10 aortic smooth muscle cells (ATCC No. ATCC CRL-1476) and the probes used were specific for rat JNKl, JNK2 or JNK3 (see infra ) . A10 cells were grown and treated with oligonucleotides essentially as described by Cioffi et al . {Mol . Pharmacol . , 1997, 51 , 383) .
A. JNKl: Table 19 describes the sequences and structures of a set of oligonucleotides, ISIS Nos. 21857 to 21870 (SEQ ID NOS: 111 to 124, respectively) that were designed to be specifically hybridizable to nucleic acids from Ra ttus norvegicus that encode a stress-activated protein kinase named "p54γ" or "SAPKy" that is homologous to the human protein JNKl (Kyriakis et al . , Na ture, 1994, 369, 156; GenBank accession No. L27129, locus name "RATSAPKD") . In Table 19, emboldened residues are 2'- methoxyethoxy-residues (others are 2 '-deoxy-); "C" residues are 2 ' -methoxyethoxy-5-methyl-cytosines and "C" residues are 5-methyl-cytosines; "o" indicates a phosphodiester linkage; and "s" indicates a phosphorothioate linkage. The target gene co-ordinates are from GenBank Accession No. L27129, locus name "RATSAPKD." TABLE 19: Nucleotide Sequences of Rat JNKl Oligonucleotides
Figure imgf000115_0001
Figure imgf000116_0001
These antisense compounds were tested for their ability to modulate levels of p54γ (JNKl) and p54a (JNK2) mRNA in A10 cells via Northern assays. Due to the high degree of sequence identity between the human and rat genes, radiolabeled human JNKl (Example 3) and JNK2
(Example 4) cDNAs functioned as specific probes for the rat homologs .
The results are shown in Table 20. Oligonucleotides showing levels of inhibition from >. about 75% to about 100% of rat JNKl mRNA levels include ISIS Nos. 21857 to 21870 (SEQ ID NOS: 111 to 124, respectively). These oligonucleotides are preferred embodiments of the invention for modulating rat JNKl expression. Oligonucleotides showing levels of inhibition of from >. about 90% to about 100% of rat JNKl mRNAs in this assay include ISIS Nos. 21858, 21859, 21860, 21861, 21862, 21865,
21866 and 21867 (SEQ ID NOS.112, 113, 114, 115, 116, 119, 120 and 121, respectively) . These oligonucleotides are thus more preferred embodiments of the invention for modulating rat JNKl expression. ISIS 21859 (SEQ ID NO: 113) was chosen for use in further studies { infra ) .
Two of the oligonucleotides, ISIS Nos. 21861 and
21867 (SEQ ID NOS: 115 and 121, respectively) demonstrated a capacity to modulate both JNKl and JNK2. Such oligonucleotides are referred to herein as "Pan JNK" antisense compounds because the term "Pan" is used in immunological literature to refer to an antibody that recognizes, e.g., all isoforms of a protein or subtypes of a cell type. The Pan JNK oligonucleotides are discussed in more detail infra .
In addition to being specifically hybridizable to nucleic acids encoding rat JNKl, some of the oligonucleotides described in Table R-l are also specifically hybridizable with JNKl-encoding nucleic acids from other species. ISIS 21859 (SEQ ID NO: 113) is complementary to bases 4 to 23 of cDNAs encoding human JNKlαl and JNKlβl (i.e., GenBank accession Nos. L26318 and U35004, respectively). ISIS 21862 (SEQ ID NO:116) is complementary to bases 294 to 313 of the human JNKlαl and JNKlβl cDNAs (GenBank accession Nos. L26318 and U35004, respectively), bases 289 to 308 of the human JNKlβ2 cDNA (GenBank accession No. U35005) , and bases 288 to 307 of the human JNKlα2 cDNA (GenBank accession No. U34822) . Finally, ISIS 21865 is complementary to bases 654 to 673 of the human JNKlαl cDNA (GenBank accession No. L26318) and to bases 648 to 667 of the human JNKla2 cDNA (GenBank accession No. U34822) . These oligonucleotides are tested for their ability to modulate mRNA levels of human JNKl genes according to the methods described in Example 3.
TABLE 20: Activities of Oligonucleotides Targeted to Rat JNKl
Figure imgf000118_0001
Figure imgf000119_0001
1 Cells treated with LIPOFECTIN™ only (no oligonucleotide) . B. JNK2 : Table 21 describes the sequences and structures of a set of oligonucleotides, ISIS Nos. 18254 to 18267 (SEQ ID NOS: 125 to 138, respectively) that were designed to be specifically hybridizable to nucleic acids that encode a stress-activated protein kinase from Ra t tus norvegicus that encode a stress-activated protein kinase named "p54α" or "SAPKα" (Kyriakis et al . , Na ture, 1994, 369, 156) . The structures of three control oligonucleotides, ISIS Nos. 21914 to 21916 (SEQ ID NOS: 139 to 141, respectively) are also shown in the table. Two isoforms of p54α have been described: "p54αl" (GenBank accession No. L27112, locus name "RATSAPKA") and "p54α2" (GenBank accession No. L27111, locus name "RATSAPKB") . With the exception of ISIS 18257 (SEQ ID NO: 128), the oligonucleotides described in Table 21 are specifically hybridizable to nucleic acids encoding either p54αl or p54α2. ISIS 18257 is specifically hybridizable to nucleic acids encoding p54α2 (i.e., GenBank accession No. L27112, locus name "RATSAPKB") . In Table 21, emboldened residues are 2 ' -methoxyethoxy-residues (others are 2 '-deoxy-); "C" residues are 2 ' -methoxyethoxy-5-methyl-cytosines and "C" residues are 5-methyl-cytosines; "o" indicates a phosphodiester linkage; and "s" indicates a phosphorothioate linkage. The target gene co-ordinates are from GenBank Accession No. L27112, locus name "RATSAPKB."
TABLE 21: Nucleotide Sequences of Rat JNK2 Oligonucleotides
Figure imgf000121_0001
tsj
Figure imgf000122_0001
Figure imgf000122_0002
TABLE 22: Activities of Oligonucleotides Targeted to Rat JNK2
Figure imgf000123_0001
L Cells treated with LIPOFECTIN1™ only (no oligonucleotide)
These antisense compounds were tested for their ability to modulate levels of p54α (JNK2) mRNA in AlO cells using the radiolabeled human JNK2 cDNA as a probe as described supra . The results are shown in Table 22. Oligonucleotides showing levels of inhibition from >. about 60% to about 100% of rat JNK2 mRNA levels include ISIS Nos. 18254, 18255, 18257, 18258, 18259, 18260 and 18264 (SEQ ID NOS:125, 126, 128, 129, 130, 131 and 135, respectively). These oligonucleotides are preferred embodiments of the invention for modulating rat JNK2 expression. Oligonucleotides showing levels of inhibition of from >. about 80% to about 100% of rat JNKl mRNAs in this assay include ISIS Nos. 18254, 18255, 18258 and 18259 (SEQ ID NOS:125, 126, 129 and 130, respectively). These oligonucleotides are thus more preferred embodiments of the invention for modulating rat JNK2 expression. ISIS 18259 (SEQ ID NO: 130) was chosen for use in further studies { infra ) . C. Dose Response: A dose response study was conducted using oligonucleotides targeted to rat JNKl (ISIS 21859; SEQ ID NO: 113) and JNK2 (ISIS 18259; SEQ ID NO: 130) and Northern assays. The results (Table 23) demonstrate an increasing effect as the oligonucleotide concentration is raised and confirm that ISIS Nos. 21859 and 18259 (SEQ ID NOS: 113 and 130, respectively) specifically modulate levels of mRNA encoding JNKl and JNK2, respectively.
TABLE 23: Dose-Dependent Response to Rat JNK Antisense Oligonucleotides (ASOs)
Figure imgf000125_0001
D. JNK3 : Table 24 describes the sequences and structures of a set of oligonucleotides, ISIS Nos. 21899 to 21912 (SEQ ID NOS: 142 to 155, respectively) that were designed to be specifically hybridizable to nucleic acids from Ra ttus norvegicus that encode a stress-activated protein kinase named "p54β" that is homologous to the human protein JNK3 (Kyriakis et al . , Na ture, 1994, 369, 156; GenBank accession No. L27128, locus name "RATSAPKC") . In Table 24, emboldened residues are 2 ' -methoxyethoxy-residues (others are 2 '-deoxy-); "C" residues are 2 ' -methoxyethoxy- 5-methyl-cytosines and "C" residues are 5-methyl-cytosines; "o" indicates a phosphodiester linkage; and "s" indicates a phosphorothioate linkage. The target gene co-ordinates are from GenBank Accession No. L27128, locus name "RATSAPKC." The oligonucleotides are tested for their ability to modulate rat JNK3 mRNA levels essentially according to the methods described in the preceding Examples. In addition to being specifically hybridizable to nucleic acids encoding rat JNK3, some of the oligonucleotides described in Table 24 are also specifically hybridizable with JNK3-encoding nucleic acids from humans and Mus musculus (mouse) . Table 25 sets out these relationships. These oligonucleotides are tested for their ability to modulate mRNA levels of the human JNK genes according to the methods described in Example 5.
TABLE 24: Nucleotide Sequences of Rat JNK3 Oligonucleotides
Figure imgf000127_0001
Figure imgf000128_0001
I
TABLE 25 : Cross-Hybridizations of Rat JNK3 Oligonucleotides
Figure imgf000129_0001
λ GenBank accession No. U34820, locus name "HSU34820" (see also Mohit et al . , Neuron, 1995, 14, 67 and Gupta et al . , EMBO Journal , 1996, 15, 2760) .
2 GenBank accession No. U34819, locus name "HSU34819" (see also Gupta et al . , EMBO Journal , 1996, 15, 2760). 3 Also known as p4593F12 MAPK; GenBank accession No. L35236, locus name "MUSMAPK" (see also Martin et al . , Brain Res . Mol . Brain Res . , 1996, 35, 47).
E. Pan JNK Oligonucleotides: Certain of the oligonucleotides of the invention are capable of modulating two or more JNK proteins and are referred to herein as "Pan JNK" oligonucleotides. For example, ISIS Nos. Nos. 21861 and 21867 (SEQ ID NOS: 115 and 121, respectively) demonstrated a capacity to modulate both JNKl and JNK2 (Table 20) . Such oligonucleotides are useful when the concomitant modulation of several JNK proteins is desired. Human Pan JNK oligonucleotides are described in Table 26. These oligonucleotides are designed to be complementary to sequences that are identically conserved in (i.e., SEQ ID NOS:156, 158, 159, 160 and 161), or which occur with no more than a one-base mismatch (SEQ ID NO: 157), in nucleic acids encoding human JNKlαl, JNKlα2, JNK2αl and JNK2α2. The oligonucleotides described in Table 26 are evaluated for their ability to modulate JNKl and JNK2 mRNA levels in A549 cells using the methods and assays described in Examples 3 and 4.
In instances where such common sequences encompass one or more base differences between the JNK genes that it is desired to modulate, hypoxanthine (inosine) may be incorporated at the positions of the oligonucleotide corresponding to such base differences. ("Hypoxanthine" is the art-accepted term for the base that corresponds to the nucleoside inosine; however, the term "inosine" is used herein in accordance with U.S. and PCT rules regarding nucleotide sequences.) As is known in the art, inosine (I) is capable of hydrogen bonding with a variety of nucleobases and thus serves as a "universal" base for hybridization purposes. For example, an oligonucleotide having a sequence that is a derivative of SEQ ID NO: 157 having one inosine substitution (TAGGAIATTCTTTCATGATC, SEQ ID NO: 162) is predicted to bind to nucleic acids encoding human JNKlαl, JNKlα2, JNK2αl and JNK2α2 with no mismatched bases. As another example, an oligonucleotide having a sequence that is a derivative of SEQ ID NO: 161 having one inosine substitution (GGTTGCAITTTCTTCATGAA, SEQ ID NO: 163) is predicted to bind with no mismatched bases to nucleic acids encoding human JNK3αl and JNK3α2 in addition to JNKlαl, JNKlα2, JNK2αl and JNK2α2. Such oligonucleotides are evaluated for their ability to modulate JNKl and JNK2 mRNA levels in A549 cells, and JNK3 mRNA levels in SH-SY5Y cells, using the methods and assays described in Examples 3, 4 and 5.
TABLE 26: Human Pan JNK Oligonucleotides
Figure imgf000131_0001
Emboldened residues, 2 ' -methoxyethoxy- residues (others are 2'-deoxy-); all "C" residues are 5-methyl-cytosines; "°", phosphodiester linkage; "s", phosphorothioate linkage.

Claims

What is claimed is :
1. An oligonucleotide comprising from 8 to 30 nucleotides connected by covalent linkages, wherein said oligonucleotide has a sequence specifically hybridizable with a nucleic acid encoding a JNKl protein, wherein said oligonucleotide modulates the expression of said JNKl protein and wherein said oligonucleotide is not specifically hybridizable to the start codon of said nucleic acid encoding said JNKl protein.
2. An oligonucleotide comprising from 8 to 30 nucleotides connected by covalent linkages, wherein said oligonucleotide has a sequence specifically hybridizable to the start codon of a nucleic acid encoding a JNKl protein and wherein said oligonucleotide modulates the expression of said JNKl protein and wherein said sequence is SEQ ID NO:13.
3. An oligonucleotide comprising from 8 to 30 nucleotides connected by covalent linkages, wherein said oligonucleotide has a sequence specifically hybridizable with a nucleic acid encoding a JNK2 protein and wherein said oligonucleotide modulates the expression of said JNK2 protein.
4. An oligonucleotide comprising from 8 to 30 nucleotides connected by covalent linkages, wherein said oligonucleotide has a sequence specifically hybridizable with a nucleic acid encoding a JNK3 protein and wherein said oligonucleotide modulates the expression of said JNK3 protein.
5. The oligonucleotide of any one of claims 1 to 4, wherein at least one of said covalent linkages of said oligonucleotide is a modified covalent linkage.
6. The oligonucleotide of any one of claims 1 or 5, wherein at least one of said nucleotides has a modified nucleobase .
7. The oligonucleotide of claim 6 wherein said modified nucleobase is 5-methylcytosine .
8. The oligonucleotide of any one of claims 1 to 7, wherein at least one of said nucleotides has a modified sugar moiety.
9. The oligonucleotide of claim 8 having at least two non-contiguous nucleotides having modified sugar moieties .
10. The oligonucleotide of either of claims 8 or
9 wherein said modified sugar moiety is a 2 ' -modified sugar moiety.
11. The oligonucleotide of claim 10 wherein said 2 '-modified sugar moiety is a 2 ' -methoxyethoxy modified suga moiety.
12. The oligonucleotide of any one of claims 1 to 11 further comprising at least one lipophilic moiety which enhances the cellular uptake of said oligonucleotide.
13. The oligonucleotide of any one of claims 1 to 12 wherein said oligonucleotide has a sequence specifically hybridizable with two or more nucleic acids encoding different isoforms of a JNK protein and wherein said oligonucleotide modulates the expression of said two or more isoforms of said JNK protein.
14. The oligonucleotide of any one of claims 1 to 12 wherein said oligonucleotide has a sequence specifically hybridizable with a nucleic acid encoding a first isoform of a JNK protein, and said sequence of said oligonucleotide is not specifically hybridizable with a nucleic acid encoding a second isoform of said JNK protein, and wherein said oligonucleotide modulates the expression of said first isoform of said JNK protein but not that of said second isoform of said JNK protein.
15. The oligonucleotide of any one of claims 1 to 14 wherein said JNK protein is that of a mammal.
16. A method of modulating the expression of a JNK protein in cells or tissues comprising contacting said cells or tissues with the oligonucleotide of any one of claims 1 to 15.
17. 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 15.
18. A method of modulating, in cultured cells or the cells of an animal, the phosphorylation of a protein phosphorylated by a JNK protein, wherein said method comprises administering to said cells an effective amount of the oligonucleotide of any one of claims 1 to 15.
19. A method of modulating, in cultured cells or the cells of an animal, the expression of a cellular protein that promotes one or more metastatic events, wherein said method comprises administering to said cells an effective amount of the oligonucleotide of any one of claims 1 to 15.
20. A pharmaceutical composition comprising the oligonucleotide of any one of claims 1 to 15, or a bioequivalent thereof, and a pharmaceutically acceptable carrier .
21. The pharmaceutical composition of claim 20, further comprising one or more compounds from the list consisting of a stabilizing agent, a penetration enhancer, a carrier compound and a chemotherapeutic agent.
22. A pharmaceutical composition comprising a plurality of the oligonucleotides of any one of claims 1 to 15, or bioequivalents thereof, and a pharmaceutically acceptable carrier.
23. A method of treating an animal having, suspected of having or prone to having a hyperproliferative disease comprising administering to said animal a prophylactically or therapeutically effective amount of the pharmaceutical composition of any one of claims 20 to 22.
24. A method of inhibiting the growth of a tumor in an animal comprising administering to said animal an effective amount of the pharmaceutical composition of any one of claims 20 to 22.
PCT/US1998/016488 1997-08-13 1998-08-07 Antisense oligonucleotide compositions and methods for the modulation of jnk proteins WO1999009214A1 (en)

Priority Applications (4)

Application Number Priority Date Filing Date Title
EP98939287A EP1003916A4 (en) 1997-08-13 1998-08-07 Antisense oligonucleotide compositions and methods for the modulation of jnk proteins
CA002301139A CA2301139A1 (en) 1997-08-13 1998-08-07 Antisense oligonucleotide compositions and methods for the modulation of jnk proteins
JP2000509875A JP3527200B2 (en) 1997-08-13 1998-08-07 Antisense oligonucleotide composition and method of modulating JNK protein
AU87750/98A AU730916B2 (en) 1997-08-13 1998-08-07 Antisense oligonucleotide compositions and methods for the modulation of JNK proteins

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US08/910,629 US5877309A (en) 1997-08-13 1997-08-13 Antisense oligonucleotides against JNK
US08/910,629 1997-08-13

Publications (1)

Publication Number Publication Date
WO1999009214A1 true WO1999009214A1 (en) 1999-02-25

Family

ID=25429082

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1998/016488 WO1999009214A1 (en) 1997-08-13 1998-08-07 Antisense oligonucleotide compositions and methods for the modulation of jnk proteins

Country Status (6)

Country Link
US (2) US5877309A (en)
EP (1) EP1003916A4 (en)
JP (1) JP3527200B2 (en)
AU (1) AU730916B2 (en)
CA (1) CA2301139A1 (en)
WO (1) WO1999009214A1 (en)

Cited By (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2001034792A2 (en) * 1999-11-12 2001-05-17 The Government Of The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Compositions and methods for the diminution or elimination of various cancers
WO2002033064A1 (en) * 2000-10-17 2002-04-25 UNIVERSITé LAVAL Zipper protein kinase and uses thereof
US7422883B2 (en) 2000-12-22 2008-09-09 Inter-K Pty Limited Map kinase integrin-binding domain
WO2009024763A2 (en) * 2007-08-17 2009-02-26 The University Of York Combination therapy
WO2009044392A2 (en) 2007-10-03 2009-04-09 Quark Pharmaceuticals, Inc. Novel sirna structures
EP2371958A1 (en) 2006-10-25 2011-10-05 Quark Pharmaceuticals, Inc. Novel siRNAs and methods of use thereof
US8119594B1 (en) 1999-06-28 2012-02-21 Inter-K Pty Limited Method of modulating integrin mediated cellular activity and agents useful for same
US9765113B2 (en) 2003-10-17 2017-09-19 Interk Peptide Therapeutics Limited Methods and agents for the treatment of cancer

Families Citing this family (98)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6867039B2 (en) * 2001-05-25 2005-03-15 Isis Pharmaceuticals, Inc. Antisense modulation of dual specific phosphatase 5 expression
US7119184B2 (en) 1991-08-12 2006-10-10 Isis Pharmaceuticals, Inc. Oligonucleotides having A-DNA form and B-DNA form conformational geometry
US6369209B1 (en) 1999-05-03 2002-04-09 Isis Pharmaceuticals, Inc. Oligonucleotides having A-DNA form and B-DNA form conformational geometry
US20030045487A1 (en) * 2001-05-25 2003-03-06 Isis Pharmaceuticals Inc. Antisense modulation of phospholipase A2, group llA (synovial) expression
US6239116B1 (en) * 1994-07-15 2001-05-29 University Of Iowa Research Foundation Immunostimulatory nucleic acid molecules
US6207646B1 (en) 1994-07-15 2001-03-27 University Of Iowa Research Foundation Immunostimulatory nucleic acid molecules
US7935675B1 (en) * 1994-07-15 2011-05-03 University Of Iowa Research Foundation Immunostimulatory nucleic acid molecules
US20030026782A1 (en) * 1995-02-07 2003-02-06 Arthur M. Krieg Immunomodulatory oligonucleotides
US6294350B1 (en) * 1997-06-05 2001-09-25 Dalhousie University Methods for treating fibroproliferative diseases
AU731909B2 (en) 1997-07-01 2001-04-05 Isis Pharmaceuticals, Inc. Compositions and methods for the delivery of oligonucleotides via the alimentary canal
US20070149472A1 (en) * 1997-08-13 2007-06-28 Mckay Robert Antisense oligonucleotide compositions and methods for the modulation of jnk proteins
US6828151B2 (en) * 2001-12-04 2004-12-07 Isis Pharmaceuticals, Inc. Antisense modulation of hematopoietic cell protein tyrosine kinase expression
US5877309A (en) * 1997-08-13 1999-03-02 Isis Pharmaceuticals, Inc. Antisense oligonucleotides against JNK
US6943000B2 (en) * 1997-10-03 2005-09-13 University Of Massachusetts JNK3 modulators and methods of use
US6897297B1 (en) * 1997-12-03 2005-05-24 Curis, Inc. Hydrophobically-modified protein compositions and methods
US6897019B1 (en) * 1998-04-17 2005-05-24 Tufts College Methods for treating and preventing insulin resistance and related disorders
CA2329252A1 (en) * 1998-05-21 1999-11-25 Isis Pharmaceuticals Inc. Compositions and methods for topical delivery of oligonucleotides
AU745880B2 (en) * 1998-05-21 2002-04-11 Isis Pharmaceuticals, Inc. Compositions and methods for non-parenteral delivery of oligonucleotides
EP1733735B1 (en) 1998-05-22 2017-03-22 Ottawa Hospital Research Institute Methods and products for inducing mucosal immunity
US20030022854A1 (en) 1998-06-25 2003-01-30 Dow Steven W. Vaccines using nucleic acid-lipid complexes
US6693086B1 (en) * 1998-06-25 2004-02-17 National Jewish Medical And Research Center Systemic immune activation method using nucleic acid-lipid complexes
US20030144224A1 (en) * 2001-12-13 2003-07-31 Isis Pharmaceuticals Inc. Antisense modulation of B-cell associated protein expression
US6277967B1 (en) 1998-07-14 2001-08-21 Isis Pharmaceuticals, Inc. Carbohydrate or 2′-modified oligonucleotides having alternating internucleoside linkages
US20040077570A1 (en) * 2002-10-17 2004-04-22 Freier Susan M. Antisense modulation of fatty acid synthase expression
JP4854853B2 (en) 1998-11-12 2012-01-18 ライフ テクノロジーズ コーポレーション Transfection drug
US5985663A (en) * 1998-11-25 1999-11-16 Isis Pharmaceuticals Inc. Antisense inhibition of interleukin-15 expression
US6114517A (en) * 1998-12-10 2000-09-05 Isis Pharmaceuticals Inc. Methods of modulating tumor necrosis factor α-induced expression of cell adhesion molecules
JP2002535015A (en) * 1999-01-29 2002-10-22 エイブイアイ バイオファーマ, インコーポレイテッド Non-invasive method for detecting target RNA
EP1152009B2 (en) * 1999-02-12 2017-09-06 Daiichi Sankyo Company, Limited Novel nucleosides and oligonucleotide analogues
KR100820266B1 (en) 1999-02-26 2008-04-08 더 유니버시티 오브 브리티쉬 콜롬비아 TRPM-2 antisense therapy
US6140124A (en) * 1999-04-06 2000-10-31 Isis Pharmaceuticals Inc. Antisense modulation of P38 mitogen activated protein kinase expression
US20040171566A1 (en) * 1999-04-06 2004-09-02 Monia Brett P. Antisense modulation of p38 mitogen activated protein kinase expression
US8541548B2 (en) * 1999-06-07 2013-09-24 Arrowhead Madison Inc. Compounds and methods for reversible modification of biologically active molecules
US20080281041A1 (en) * 1999-06-07 2008-11-13 Rozema David B Reversibly Masked Polymers
US20040006031A1 (en) * 2002-07-02 2004-01-08 Isis Pharmaceuticals Inc. Antisense modulation of HMG-CoA reductase expression
JP4151751B2 (en) * 1999-07-22 2008-09-17 第一三共株式会社 New bicyclonucleoside analogues
US6787136B1 (en) * 1999-09-03 2004-09-07 The Brigham And Women's Hospital, Inc. Methods and compositions for treatment of inflammatory disease using cadherin-11 modulating agents
US6617442B1 (en) * 1999-09-30 2003-09-09 Isis Pharmaceuticals, Inc. Human Rnase H1 and oligonucleotide compositions thereof
US20030095953A1 (en) * 1999-11-12 2003-05-22 Myles C. Cabot Methods of reversing drug resistance in cancer cells
US6316259B1 (en) * 2000-01-21 2001-11-13 Isis Pharmaceuticals, Inc. Antisense inhibition of glycogen synthase kinase 3 alpha expression
US7569551B2 (en) 2000-02-25 2009-08-04 The University Of British Columbia Chemo- and radiation-sensitization of cancer by antisense TRPM-2 oligodeoxynucleotides
US20020045590A1 (en) * 2000-10-23 2002-04-18 Yuanxiang Tao Inhibition of interaction of PSD93 and PSD95 with nNOS and NMDA receptors
KR100917101B1 (en) * 2000-08-04 2009-09-15 도요 보세키 가부시키가이샤 Flexible metal laminate and production method thereof
US6346416B1 (en) * 2000-08-29 2002-02-12 Isis Pharmaceuticals, Inc. Antisense inhibition of HPK/GCK-like kinase expression
AU2002232387A1 (en) * 2000-10-27 2002-05-06 Invitrogen Corporation Method for introducing antisense oligonucleotides into eucaryotic cells
US20030125273A1 (en) * 2001-12-05 2003-07-03 Isis Pharmaceuticals Inc, Antisense modulation of MHC class II transactivator expression
US20030171310A1 (en) * 2001-02-23 2003-09-11 Isis Pharmaceuticals Inc. Antisense modulation of RECQL expression
US20030087855A1 (en) * 2001-09-13 2003-05-08 Isis Pharmaceuticals Inc. Antisense modulation of protein kinase R expression
US7232897B2 (en) * 2001-04-24 2007-06-19 Harvard University, President And Fellows Of Harvard College Compositions and methods for modulating NH2-terminal Jun Kinase activity
US20070042983A1 (en) * 2001-05-18 2007-02-22 Sirna Therapeutics, Inc. RNA interference mediated inhibition of gene expression using short interfering nucleic acid (siNA)
WO2003070969A2 (en) * 2002-02-20 2003-08-28 Sirna Therapeutics, Inc. RNA INTERFERENCE MEDIATED INHIBITION OF BCL2 GENE EXPRESSION USING SHORT INTERFERING NUCLEIC ACID (siNA)
US20050182006A1 (en) * 2001-05-18 2005-08-18 Sirna Therapeutics, Inc RNA interference mediated inhibition of protein kinase C alpha (PKC-alpha) gene expression using short interfering nucleic acid (siNA)
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)
US9994853B2 (en) 2001-05-18 2018-06-12 Sirna Therapeutics, Inc. Chemically modified multifunctional short interfering nucleic acid molecules that mediate RNA interference
US20080161256A1 (en) * 2001-05-18 2008-07-03 Sirna Therapeutics, Inc. RNA interference mediated inhibition of 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)
WO2003070983A1 (en) * 2002-02-20 2003-08-28 Sirna Therapeutics, Inc RNA INTERFERENCE MEDIATED INHIBITION OF PROTEIN KINASE C ALPHA (PKC-ALPHA) GENE EXPRESSION USING SHORT INTERFERING NUCLEIC ACID (siNA)
SG177000A1 (en) * 2001-08-17 2012-01-30 Coley Pharm Gmbh Combination motif immune stimulatory oligonucleotides with improved activity
US20030124196A1 (en) * 2001-08-22 2003-07-03 Susan Weinbach Pulsatile release compositions and methods for enhanced intestinal drug absorption
US20030083286A1 (en) * 2001-08-22 2003-05-01 Ching-Leou Teng Bioadhesive compositions and methods for enhanced intestinal drug absorption
US6750019B2 (en) * 2001-10-09 2004-06-15 Isis Pharmaceuticals, Inc. Antisense modulation of insulin-like growth factor binding protein 5 expression
DE60312656T2 (en) * 2002-01-31 2007-11-29 Daiichi Pharmaceutical Co., Ltd. MEDICAMENT FOR THE TREATMENT OF DISORDERS OF THE CIRCADIAN RHYTHM
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)
US20030224515A1 (en) * 2002-06-04 2003-12-04 Isis Pharmaceuticals Inc. Antisense modulation of sterol regulatory element-binding protein-1 expression
US20030211606A1 (en) * 2002-05-10 2003-11-13 Isis Pharmaceuticals Inc. Antisense modulation of DYRK4 expression
US20030224512A1 (en) * 2002-05-31 2003-12-04 Isis Pharmaceuticals Inc. Antisense modulation of beta-site APP-cleaving enzyme expression
US20030220273A1 (en) * 2002-05-15 2003-11-27 Isis Pharmaceuticals Inc. Antisense modulation of phosphodiesterase 4D expression
US20030228688A1 (en) * 2002-05-31 2003-12-11 Isis Pharmaceuticals Inc. Antisense modulation of isoprenylcysteine carboxyl methyltransferase expression
US20040096836A1 (en) * 2002-11-19 2004-05-20 Isis Pharmaceuticals Inc. Modulation of mitogen-activated protein kinase 13 expression
US7465714B2 (en) * 2002-06-20 2008-12-16 Mcgill University Oligonucleotide inhibitors of MBD2/DNA demethylase and uses thereof
US20050130924A1 (en) * 2002-06-26 2005-06-16 Monia Brett P. Antisense inhibition via RNAse H-independent reduction in mRNA
US7956043B2 (en) * 2002-12-11 2011-06-07 Coley Pharmaceutical Group, Inc. 5′ CpG nucleic acids and methods of use
CA2522680A1 (en) * 2003-04-15 2004-10-28 Xenon Pharmaceuticals Inc. Juvenile hemochromatosis gene (hfe2a), expression products and uses thereof
US7339051B2 (en) * 2003-04-28 2008-03-04 Isis Pharmaceuticals, Inc. Compositions and methods for the treatment of severe acute respiratory syndrome (SARS)
US20050013812A1 (en) * 2003-07-14 2005-01-20 Dow Steven W. Vaccines using pattern recognition receptor-ligand:lipid complexes
US7144907B2 (en) * 2003-09-03 2006-12-05 Array Biopharma Inc. Heterocyclic inhibitors of MEK and methods of use thereof
ES2662196T3 (en) 2003-09-09 2018-04-05 Geron Corporation Modified oligonucleotides for telomerase inhibition
US20050239733A1 (en) * 2003-10-31 2005-10-27 Coley Pharmaceutical Gmbh Sequence requirements for inhibitory oligonucleotides
US20060003953A1 (en) * 2003-12-04 2006-01-05 Bennett C F Compositions and their uses directed to bone growth modulators
EP2363480A3 (en) * 2004-01-20 2015-10-07 Isis Pharmaceuticals, Inc. Modulation of glucocorticoid receptor expression
TWI235440B (en) * 2004-03-31 2005-07-01 Advanced Semiconductor Eng Method for making leadless semiconductor package
US8710020B2 (en) * 2004-04-02 2014-04-29 The University Of British Columbia Clusterin antisense therapy for treatment of cancer
US10508277B2 (en) 2004-05-24 2019-12-17 Sirna Therapeutics, Inc. Chemically modified multifunctional short interfering nucleic acid molecules that mediate RNA interference
US20060223807A1 (en) * 2005-03-29 2006-10-05 University Of Massachusetts Medical School, A Massachusetts Corporation Therapeutic methods for type I diabetes
US20090117132A1 (en) * 2005-07-07 2009-05-07 Pfizer, Inc. Anti-Ctla-4 Antibody and Cpg-Motif-Containing Synthetic Oligodeoxynucleotide Combination Therapy for Cancer Treatment
US20090176725A1 (en) * 2005-08-17 2009-07-09 Sirna Therapeutics Inc. Chemically modified short interfering nucleic acid molecules that mediate rna interference
US8101585B2 (en) * 2006-08-04 2012-01-24 Isis Pharmaceuticals, Inc. Compositions and methods for the modulation of JNK proteins
CN102614528B (en) * 2006-08-18 2014-02-26 箭头研究公司 Polyconjugates for in vivo delivery of polynucleotides
WO2008094640A2 (en) * 2007-01-30 2008-08-07 Geron Corporation Compounds having anti-adhesive effects on cancer cells
NZ601737A (en) * 2010-02-24 2013-06-28 Arrowhead Res Corp Compositions for targeted delivery of sirna
US8877188B2 (en) 2010-05-04 2014-11-04 The Brigham And Women's Hospital, Inc. Detection and treatment of non-dermal fibrosis
WO2012058210A1 (en) 2010-10-29 2012-05-03 Merck Sharp & Dohme Corp. RNA INTERFERENCE MEDIATED INHIBITION OF GENE EXPRESSION USING SHORT INTERFERING NUCLEIC ACIDS (siNA)
CN103906838A (en) 2011-10-25 2014-07-02 Isis制药公司 Antisense modulation of GCCR expression
JOP20200257A1 (en) 2014-05-01 2017-06-16 Geron Corp Oligonucleotide Compositions and Methods of Making the Same
WO2016011203A1 (en) 2014-07-15 2016-01-21 Life Technologies Corporation Compositions with lipid aggregates and methods for efficient delivery of molecules to cells
US11097005B2 (en) 2014-12-15 2021-08-24 The Brigham And Women's Hospital, Inc. Use of cadherin-11 antagonists to treat metabolic disorders and/or increase insulin sensitivity

Family Cites Families (49)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4309406A (en) * 1979-07-10 1982-01-05 American Home Products Corporation Sustained release pharmaceutical compositions
US4309404A (en) * 1979-08-09 1982-01-05 American Home Products Corporation Sustained release pharmaceutical compositions
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
US4704295A (en) * 1983-09-19 1987-11-03 Colorcon, Inc. Enteric film-coating compositions
US4556552A (en) * 1983-09-19 1985-12-03 Colorcon, Inc. Enteric film-coating compositions
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
US5034506A (en) * 1985-03-15 1991-07-23 Anti-Gene Development Group Uncharged morpholino-based polymers having achiral intersubunit linkages
US5194428A (en) * 1986-05-23 1993-03-16 Worcester Foundation For Experimental Biology Inhibition of influenza virus replication by oligonucleotide phosphorothioates
US4806463A (en) * 1986-05-23 1989-02-21 Worcester Foundation For Experimental Biology Inhibition of HTLV-III by exogenous oligonucleotides
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
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
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
US5457191A (en) * 1990-01-11 1995-10-10 Isis Pharmaceuticals, Inc. 3-deazapurines
US5587470A (en) * 1990-01-11 1996-12-24 Isis Pharmaceuticals, Inc. 3-deazapurines
US5587361A (en) * 1991-10-15 1996-12-24 Isis Pharmaceuticals, Inc. Oligonucleotides having phosphorothioate linkages of high chiral purity
US5578718A (en) * 1990-01-11 1996-11-26 Isis Pharmaceuticals, Inc. Thiol-derivatized nucleosides
US5459255A (en) * 1990-01-11 1995-10-17 Isis Pharmaceuticals, Inc. N-2 substituted purines
US5506351A (en) * 1992-07-23 1996-04-09 Isis Pharmaceuticals Process for the preparation of 2'-O-alkyl guanosine and related compounds
US5212295A (en) * 1990-01-11 1993-05-18 Isis Pharmaceuticals Monomers for preparation of oligonucleotides having chiral phosphorus linkages
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
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
US5378825A (en) * 1990-07-27 1995-01-03 Isis Pharmaceuticals, Inc. Backbone modified oligonucleotide analogs
US5602240A (en) * 1990-07-27 1997-02-11 Ciba Geigy Ag. Backbone modified oligonucleotide analogs
US5138045A (en) * 1990-07-27 1992-08-11 Isis Pharmaceuticals Polyamine conjugated oligonucleotides
US5223618A (en) * 1990-08-13 1993-06-29 Isis Pharmaceuticals, Inc. 4'-desmethyl nucleoside analog compounds
US5608046A (en) * 1990-07-27 1997-03-04 Isis Pharmaceuticals, Inc. Conjugated 4'-desmethyl nucleoside analog compounds
US5610289A (en) * 1990-07-27 1997-03-11 Isis Pharmaceuticals, Inc. Backbone modified oligonucleotide analogues
US5218105A (en) * 1990-07-27 1993-06-08 Isis Pharmaceuticals Polyamine conjugated oligonucleotides
DK0544713T3 (en) 1990-08-16 1997-09-29 Isis Pharmaceuticals Inc Oligonucleutides for modulating the effects of cytomegalovirus infections
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
CA2082411A1 (en) 1991-06-28 1992-12-29 Robert D. Rosenberg Localized oligonucleotide therapy
US5599797A (en) * 1991-10-15 1997-02-04 Isis Pharmaceuticals, Inc. Oligonucleotides having phosphorothioate linkages of high chiral purity
FR2692265B1 (en) * 1992-05-25 1996-11-08 Centre Nat Rech Scient BIOLOGICALLY ACTIVE COMPOUNDS OF THE PHOSPHOTRIESTER TYPE.
DE69333698T2 (en) * 1992-07-20 2005-12-01 Isis Pharmaceutical, Inc., Carlsbad PSEUDO HALF NODES FORMING RNA BY HYBRIDIZING ANTISSESEOLIGON NUCLEOTIDES TO TARGETED RNA SECONDARY STRUCTURES
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
US5539083A (en) 1994-02-23 1996-07-23 Isis Pharmaceuticals, Inc. Peptide nucleic acid combinatorial libraries and improved methods of synthesis
US5554746A (en) * 1994-05-16 1996-09-10 Isis Pharmaceuticals, Inc. Lactam nucleic acids
US5744362A (en) * 1994-05-31 1998-04-28 Isis Pharmaceuticals, Inc. Antisense oligonucleotide modulation of raf gene expression
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
WO1998054203A1 (en) * 1997-05-28 1998-12-03 Mercola Daniel A Inhibition of stress activated protein kinase (sapk) pathway and sensitization of cells to cancer therapies
US5877309A (en) * 1997-08-13 1999-03-02 Isis Pharmaceuticals, Inc. Antisense oligonucleotides against JNK
US6943000B2 (en) * 1997-10-03 2005-09-13 University Of Massachusetts JNK3 modulators and methods of use

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
DERIJARD B., ET AL.: "JNK1: A PROTEIN KINASE STIMULATED BY UV LIGHT AND HA-RAS THAT BINDS AND PHOSPHORYLATES THE C-JUN ACTIVATION DOMAIN.", CELL, CELL PRESS, US, vol. 76., 25 March 1994 (1994-03-25), US, pages 1025 - 1037., XP002915274, ISSN: 0092-8674, DOI: 10.1016/0092-8674(94)90380-8 *
GUPTA S, ET AL.: "SELECTIVE INTERACTION OF JNK PROTEIN KINASE ISOFORMS WITH TRANSCRIPTION FACTORS", EMBO JOURNAL., OXFORD UNIVERSITY PRESS, SURREY., GB, vol. 15, no. 11, 27 June 1997 (1997-06-27), GB, pages 2760 - 2770, XP002915273, ISSN: 0261-4189 *
See also references of EP1003916A4 *
SEIMIYA H., ET AL.: "C-JUN NH2-TERMINAL KINASE-MEDIATED ACTIVATION OF INTERLEUKIN-1BETA CONVERTING ENZYME/CED-3-LIKE PROTEASE DURING ANTICANCER DRUG- INDUCED APOPTOSIS.", JOURNAL OF BIOLOGICAL CHEMISTRY, AMERICAN SOCIETY FOR BIOCHEMISTRY AND MOLECULAR BIOLOGY, US, vol. 272., no. 07., 14 February 1997 (1997-02-14), US, pages 4631 - 4636., XP002915272, ISSN: 0021-9258, DOI: 10.1074/jbc.272.37.23407 *

Cited By (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8618062B2 (en) 1999-06-26 2013-12-31 Inter-K Pty Limited Method of modulating integrin mediated cellular activity and agents useful for same
US8119594B1 (en) 1999-06-28 2012-02-21 Inter-K Pty Limited Method of modulating integrin mediated cellular activity and agents useful for same
WO2001034792A2 (en) * 1999-11-12 2001-05-17 The Government Of The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Compositions and methods for the diminution or elimination of various cancers
WO2001034792A3 (en) * 1999-11-12 2001-11-22 Us Health Compositions and methods for the diminution or elimination of various cancers
WO2002033064A1 (en) * 2000-10-17 2002-04-25 UNIVERSITé LAVAL Zipper protein kinase and uses thereof
US7422883B2 (en) 2000-12-22 2008-09-09 Inter-K Pty Limited Map kinase integrin-binding domain
US9765113B2 (en) 2003-10-17 2017-09-19 Interk Peptide Therapeutics Limited Methods and agents for the treatment of cancer
EP2371958A1 (en) 2006-10-25 2011-10-05 Quark Pharmaceuticals, Inc. Novel siRNAs and methods of use thereof
WO2009024763A2 (en) * 2007-08-17 2009-02-26 The University Of York Combination therapy
WO2009024763A3 (en) * 2007-08-17 2010-04-29 The University Of York Combination therapy
WO2009044392A2 (en) 2007-10-03 2009-04-09 Quark Pharmaceuticals, Inc. Novel sirna structures

Also Published As

Publication number Publication date
EP1003916A1 (en) 2000-05-31
US6221850B1 (en) 2001-04-24
US5877309A (en) 1999-03-02
JP3527200B2 (en) 2004-05-17
AU8775098A (en) 1999-03-08
AU730916B2 (en) 2001-03-22
CA2301139A1 (en) 1999-02-25
JP2001514905A (en) 2001-09-18
EP1003916A4 (en) 2002-01-09

Similar Documents

Publication Publication Date Title
US5877309A (en) Antisense oligonucleotides against JNK
US6133246A (en) Antisense oligonucleotide compositions and methods for the modulation of JNK proteins
US5985558A (en) Antisense oligonucleotide compositions and methods for the inibition of c-Jun and c-Fos
US8101585B2 (en) Compositions and methods for the modulation of JNK proteins
US20020082415A1 (en) Antisense oligonucleotide modulation of human serine/threonine protein phosphatase gene expression
US6111094A (en) Enhanced antisense modulation of ICAM-1
US6809193B2 (en) Antisense oligonucleotide compositions and methods for the modulation of JNK proteins
US20020151511A1 (en) Antisense oligonucleotide modulation of human MDM2 expression
JP2002531574A (en) Methods of modulating tumor necrosis factor α-induced expression of cell adhesion molecules
JP2003520241A (en) Antisense modulation of MEKK2 expression
JP2003505027A (en) Antisense modulation of PI3 kinase P110delta expression
JP2003505062A (en) Antisense modulation of PTEN expression
JP2003505063A (en) Antisense modulation of MEKK5 expression
JP2003508089A (en) Antisense modulation of PI3 kinase p110 beta expression
JP2003512079A (en) Antisense modulation of protein kinase C-theta expression
JP2003518928A (en) Antisense modulation of Akt-3 expression
JP2003527085A (en) Antisense modulation of PEPCK-mitochondrial expression
JP2003520041A (en) Antisense modulation of glycogen synthase kinase 3 beta expression
JP3590585B2 (en) Antisense regulation of LFA-3
JP2003505037A (en) Antisense modulation of telomere repeat binding factor 1 expression
JP2003520242A (en) Antisense modulation of glycogen synthase kinase 3 alpha expression
JP2003513640A (en) Antisense inhibition of PI3 kinase p55 gamma expression
US20070149472A1 (en) Antisense oligonucleotide compositions and methods for the modulation of jnk proteins
US20040029823A1 (en) Antisense oligonucleotide compositions and methods for the modulation of JNK proteins
US6607915B1 (en) Antisense inhibition of E2A-Pbx1 expression

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 HR 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 GW 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
ENP Entry into the national phase

Ref document number: 2301139

Country of ref document: CA

Ref country code: CA

Ref document number: 2301139

Kind code of ref document: A

Format of ref document f/p: F

NENP Non-entry into the national phase

Ref country code: KR

WWE Wipo information: entry into national phase

Ref document number: 1998939287

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 87750/98

Country of ref document: AU

WWP Wipo information: published in national office

Ref document number: 1998939287

Country of ref document: EP

REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

WWG Wipo information: grant in national office

Ref document number: 87750/98

Country of ref document: AU

WWW Wipo information: withdrawn in national office

Ref document number: 1998939287

Country of ref document: EP