US20040192628A1 - Antisense modulation of focal adhesion kinase expression - Google Patents

Antisense modulation of focal adhesion kinase expression Download PDF

Info

Publication number
US20040192628A1
US20040192628A1 US10/646,569 US64656903A US2004192628A1 US 20040192628 A1 US20040192628 A1 US 20040192628A1 US 64656903 A US64656903 A US 64656903A US 2004192628 A1 US2004192628 A1 US 2004192628A1
Authority
US
United States
Prior art keywords
antisense compound
antisense
leu
glu
focal adhesion
Prior art date
Legal status (The legal status 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 status listed.)
Abandoned
Application number
US10/646,569
Inventor
Brett Monia
William Gaarde
Pamela Nero
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Individual
Original Assignee
Individual
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
Priority to US09/966,451 priority Critical patent/US6692959B2/en
Priority to PCT/US2002/030574 priority patent/WO2003028636A2/en
Priority to EP02776002A priority patent/EP1436308A4/en
Priority to US10/630,399 priority patent/US20040019009A1/en
Application filed by Individual filed Critical Individual
Priority to US10/646,569 priority patent/US20040192628A1/en
Publication of US20040192628A1 publication Critical patent/US20040192628A1/en
Priority to US11/117,013 priority patent/US20050267063A1/en
Priority to US11/505,758 priority patent/US20070049545A1/en
Abandoned legal-status Critical Current

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
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/31Chemical structure of the backbone
    • C12N2310/315Phosphorothioates
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/32Chemical 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
    • C12N2310/33415-Methylcytosine
    • 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
    • C12N2310/34Spatial arrangement of the modifications
    • C12N2310/341Gapmers, i.e. of the type ===---===
    • 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
    • C12N2310/34Spatial arrangement of the modifications
    • C12N2310/346Spatial arrangement of the modifications having a combination of backbone and sugar modifications

Definitions

  • This invention relates to compositions and methods for modulating expression of the human focal adhesion kinase (FAK) gene, which encodes a signaling protein involved in growth factor response and cell migration and is implicated in disease.
  • FAK focal adhesion kinase
  • This invention is also directed to methods for inhibiting FAK-mediated signal transduction; these methods can be used diagnostically or therapeutically.
  • this invention is directed to treatment of conditions associated with expression of the human FAK gene.
  • FAM focal adhesion kinase
  • ECM cell substratum-extracellular matrix
  • FAK In adherent cells, FAK is often associated with integrins at focal adhesions (Schaller, M. D., et al., Proc. Natl. Acad. Sci. USA , 1992, 89, 5192-5196). Numerous other signaling proteins, including other protein tyrosine kinases are associated with FAK at these regions. Phosphorylation of FAK results in activation of the mitogen-activated protein kinase pathway. In addition, FAK regulates activation of phosphatidylinositol 3′-kinase which may serve to prevent apoptosis. FAK has also been shown to be required for internalization of bacteria mediated by invasin (Alrutz, M. A. and Isberg, R. R., Proc. Natl. Acad. Sci. USA , 1998, 95, 13658-13663).
  • Normal cells typically require anchorage to the extracellular matrix in order to grow. When these cells are removed from the extracellular matrix, they undergo apoptosis. Transformed cells, on the other hand, can grow under anchorage-independent conditions, providing them a growth advantage and the ability to be removed from their normal cellular environment.
  • FAK's role in cell migration has led to the speculation that it may be relevant in other diseases such as embryonic development disfunctions and angiogenic disorders (Kornberg, L. J., Head Neck , 1998, 20, 634-639).
  • Antisense oligonucleotides have been used in several studies. Tanaka, S. et al. ( J. Cell. Biochem ., 1995, 58, 424-435) disclose two antisense phosphorothioate oligonucleotides targeted to the start site of mouse FAK. Xu, L.-H., et al. ( Cell Growth Diff ., 1996, 7, 413-418) disclose two antisense phosphorothioate oligonucleotides targeted within the coding region of human FAK. They also show that FAK antisense treatment could induce apoptosis in tumor cells. Sonoda, Y., et al. ( Biochem. Biophys. Res.
  • the present invention provides antisense compounds which are targeted to nucleic acids encoding focal adhesion kinase expression (FAK) and are capable of modulating FAK mediated signaling.
  • the present invention also provides chimeric oligonucleotides targeted to nucleic acids encoding human FAK.
  • the antisense compounds of the invention are believed to be useful both diagnostically and therapeutically, and are believed to be particularly useful in the methods of the present invention.
  • the present invention also comprises methods of modulating FAK mediated signaling, in cells and tissues, using the antisense compounds of the invention.
  • Methods of inhibiting FAK expression are provided; these methods are believed to be useful both therapeutically and diagnostically. These methods are also useful as tools, for example, for detecting and determining the role of FAK in various cell functions and physiological processes and conditions and for diagnosing conditions associated with expression of FAK.
  • the present invention also comprises methods for diagnosing and treating cancers, including those of the colon, breast and mouth. These methods are believed to be useful, for example, in diagnosing FAK-associated disease progression. These methods employ the antisense compounds of the invention. These methods are believed to be useful both therapeutically, including prophylactically, and as clinical research and diagnostic tools.
  • FAK plays important roles in integrin-mediated signal transduction. Overexpression of FAK is associated with tumor progression and metastatic potential. As such, this protein represents an attractive target for treatment of such diseases. In particular, modulation of the expression of FAK may be useful for the treatment of diseases such as colon cancer, breast cancer and cancer of the mouth.
  • the present invention employs antisense compounds, particularly oligonucleotides, for use in modulating the function of nucleic acid molecules encoding FAK, ultimately modulating the amount of FAK produced. This is accomplished by providing oligonucleotides which specifically hybridize with nucleic acids, preferably mRNA, encoding FAK.
  • an antisense compound such as an oligonucleotide and its complementary nucleic acid target, to which it hybridizes, is commonly referred to as “antisense”.
  • “Targeting” an oligonucleotide to a chosen nucleic acid target is a multistep process. The process usually begins with identifying a nucleic acid sequence whose function is to be modulated. This may be, as examples, a cellular gene (or mRNA made from the gene) whose expression is associated with a particular disease state, or a foreign nucleic acid from an infectious agent.
  • the targets are nucleic acids encoding FAK; in other words, a gene encoding FAK, or mRNA expressed from the FAK gene. mRNA which encodes FAK is presently the preferred target.
  • the targeting process also includes determination of a site or sites within the nucleic acid sequence for the antisense interaction to occur such that modulation of gene expression will result.
  • messenger RNA includes not only the information to encode a protein using the three letter genetic code, but also associated ribonucleotides which form a region known to such persons as the 5′-untranslated region, the 3′-untranslated region, the 5′ cap region and intron/exon junction ribonucleotides.
  • oligonucleotides may be formulated in accordance with this invention which are targeted wholly or in part to these associated ribonucleotides as well as to the informational ribonucleotides.
  • the oligonucleotide may therefore be specifically hybridizable with a transcription initiation site region, a translation initiation codon region, a 5′ cap region, an intron/exon junction, coding sequences, a translation termination codon region or sequences in the 5′- or 3′-untranslated region.
  • 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.
  • 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.
  • 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 FAK, 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).
  • start codon region refers to a portion of such an mRNA or gene that encompasses from about 25 to about 50 contiguous nucleotides in either direction (i.e., 5′ or 3′)from a translation initiation codon. This region is a preferred target region.
  • stop codon region refers to a portion of such an mRNA or gene that encompasses from about 25 to about 50 contiguous nucleotides in either direction (i.e., 5′ or 3′) from a translation termination codon. This region is a preferred target region.
  • Other preferred target regions include the 5′ untranslated region (5′UTR), known in the art to refer to the portion of an mRNA in the 5′ direction from the translation initiation codon, and thus including nucleotides between the 5′ cap site and the translation initiation codon of an mRNA or corresponding nucleotides on the gene and the 3′ untranslated region (3′UTR), known in the art to refer to the portion of an mRNA in the 3′ direction from the translation termination codon, and thus including nucleotides between the translation termination codon and 3′ end of an mRNA or corresponding nucleotides on the gene.
  • 5′UTR 5′ untranslated region
  • 3′UTR 3′ untranslated region
  • the 5′ cap of an mRNA comprises an N7-methylated guanosine residue joined to the 5′-most residue of the mRNA via a 5′-5′ triphosphate linkage.
  • the 5′ cap region of an mRNA is considered to include the 5′ cap structure itself as well as the first 50 nucleotides adjacent to the cap.
  • the 5′ cap region may also be a preferred target region.
  • mRNA splice sites i.e., exon-exon or intron-exon junctions
  • introns may also be preferred target regions, and are particularly useful in situations where aberrant splicing is implicated in disease, or where an overproduction of a particular mRNA splice product is implicated in disease.
  • Aberrant fusion junctions due to rearrangements or deletions are also preferred targets.
  • Targeting particular exons in alternatively spliced mRNAs may also be preferred. It has also been found that introns can also be effective, and therefore preferred, target regions for antisense compounds targeted, for example, to DNA or pre-mRNA.
  • oligonucleotides are chosen which are sufficiently complementary to the target, i.e., hybridize sufficiently well and with sufficient specificity, to give the desired modulation.
  • Hybridization in the context of this invention, means hydrogen bonding, also known as Watson-Crick base pairing, between complementary bases, usually on opposite nucleic acid strands or two regions of a nucleic acid strand.
  • Guanine and cytosine are examples of complementary bases which are known to form three hydrogen bonds between them.
  • Adenine and thymine are examples of complementary bases which form two hydrogen bonds between them.
  • “Specifically hybridizable” and “complementary” are terms which are used to indicate a sufficient degree of complementarity such that stable and specific binding occurs between the DNA or RNA target and the oligonucleotide.
  • an oligonucleotide need not be 100% complementary to its target nucleic acid sequence to be specifically hybridizable.
  • An oligonucleotide is specifically hybridizable when binding of the oligonucleotide to the target interferes with the normal function of the target molecule to cause a loss of utility, and there is a sufficient degree of complementarity to avoid non-specific binding of the oligonucleotide to non-target sequences under conditions in which specific binding is desired, i.e., under physiological conditions in the case of in vivo assays or therapeutic treatment or, in the case of in vitro assays, under conditions in which the assays are conducted.
  • Hybridization of antisense oligonucleotides with mRNA interferes with one or more of the normal functions of mRNA.
  • the functions of mRNA to be interfered with include all vital functions such as, for example, translocation of the RNA to the site of protein translation, translation of protein from the RNA, splicing of the RNA to yield one or more mRNA species, and catalytic activity which may be engaged in by the RNA. Binding of specific protein(s) to the RNA may also be interfered with by antisense oligonucleotide hybridization to the RNA.
  • modulation means either inhibition or stimulation; i.e., either a decrease or increase in expression.
  • This modulation can be measured in ways which are routine in the art, for example by Northern blot assay of mRNA expression, or reverse transcriptase PCR, as taught in the examples of the instant application or by Western blot or ELISA assay of protein expression, or by an immunoprecipitation assay of protein expression. Effects on cell proliferation or tumor cell growth can also be measured, as taught in the examples of the instant application. Inhibition is presently preferred.
  • the oligonucleotides of this invention can be used in diagnostics, therapeutics, prophylaxis, and as research reagents and in kits. Since the oligonucleotides of this invention hybridize to nucleic acids encoding FAK, sandwich, calorimetric and other assays can easily be constructed to exploit this fact. Provision of means for detecting hybridization of oligonucleotide with the FAK genes or mRNA can routinely be accomplished. Such provision may include enzyme conjugation, radiolabelling or any other suitable detection systems. Kits for detecting the presence or absence of FAK may also be prepared.
  • the present invention is also suitable for diagnosing abnormal inflammatory states or certain cancers in tissue or other samples from patients suspected of having an autoimmune or inflammatory disease such as hepatitis or cancers such as those of the colon, liver or lung, and lymphomas.
  • a number of assays may be formulated employing the present invention, which assays will commonly comprise contacting a tissue sample with an oligonucleotide of the invention under conditions selected to permit detection and, usually, quantitation of such inhibition.
  • to “contact” tissues or cells with an oligonucleotide or oligonucleotides means to add the oligonucleotide(s), usually in a liquid carrier, to a cell suspension or tissue sample, either in vitro or ex vivo, or to administer the oligonucleotide(s) to cells or tissues within an animal.
  • the oligonucleotides of this invention may also be used for research purposes.
  • the specific hybridization exhibited by the oligonucleotides 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.
  • 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.
  • the antisense compounds in accordance with this invention preferably comprise from about 5 to about 50 nucleobases.
  • Particularly preferred are antisense oligonucleotides comprising from about 8 to about 30 nucleobases (i.e. from about 8 to about 30 linked nucleosides).
  • a nucleoside is a base-sugar combination.
  • the base portion of the nucleoside is normally a heterocyclic base.
  • the two most common classes of such heterocyclic bases are the purines and the pyrimidines.
  • Nucleotides are nucleosides that further include a phosphate group covalently linked to the sugar portion of the nucleoside.
  • the phosphate groups covalently link adjacent nucleosides to one another to form a linear polymeric compound. In turn the respective ends of this linear polymeric structure can be further joined to form a circular structure, however, open linear structures are generally preferred.
  • oligonucleotides containing modified backbones or non-natural internucleoside linkages include those that retain a phosphorus atom in the backbone and those that do not have a phosphorus atom in the backbone.
  • modified oligonucleotides that do not have a phosphorus atom in their internucleoside backbone can also be considered to be oligonucleosides.
  • Various salts, mixed salts and free acid forms are also included.
  • Representative United States patents that teach the preparation of the above phosphorus-containing linkages include, but are not limited to, U.S. Pat. Nos. 3,687,808; 4,469,863; 4,476,301; 5,023,243; 5,177,196; 5,188,897; 5,264,423; 5,276,019; 5,278,302; 5,286,717; 5,321,131; 5,399,676; 5,405,939; 5,453,496; 5,455,233; 5,466,677; 5,476,925; 5,519,126; 5,536,821; 5,541,306; 5,550,111; 5,563,253; 5,571,799; 5,587,361; and 5,625,050.
  • Preferred modified oligonucleotide backbones that do not include a phosphorus atom therein have backbones that are formed by short chain alkyl or cycloalkyl internucleoside linkages, mixed heteroatom and alkyl or cycloalkyl internucleoside linkages, or one or more short chain heteroatomic or heterocyclic internucleoside linkages.
  • morpholino linkages formed in part from the sugar portion of a nucleoside
  • siloxane backbones sulfide, sulfoxide and sulfone backbones
  • formacetyl and thioformacetyl backbones methylene formacetyl and thioformacetyl backbones
  • alkene containing backbones sulfamate backbones
  • sulfonate and sulfonamide backbones amide backbones; and others having mixed N, O, S and CH 2 component parts.
  • Representative United States patents that teach the preparation of the above oligonucleosides include, but are not limited to, U.S. Pat. Nos. 5,034,506; 5,166,315; 5,185,444; 5,214,134; 5,216,141; 5,235,033; 5,264,562; 5,264,564; 5,405,938; 5,434,257; 5,466,677; 5,470,967; 5,489,677; 5,541,307; 5,561,225; 5,596,086; 5,602,240; 5,610,289; 5,602,240; 5,608,046; 5,610,289; 5,618,704; 5,623,070; 5,663,312; 5,633,360; 5,677,437; and 5,677,439.
  • both the sugar and the internucleoside linkage, i.e., the backbone, of the nucleotide units are replaced with novel groups.
  • the base units are maintained for hybridization with an appropriate nucleic acid target compound.
  • an oligonucleotide mimetic that has been shown to have excellent hybridization properties, is referred to as a peptide nucleic acid (PNA).
  • PNA peptide nucleic acid
  • the sugar-backbone of an oligonucleotide is replaced with an amide containing backbone, in particular an aminoethylglycine backbone.
  • nucleobases are retained and are bound directly or indirectly to aza nitrogen atoms of the amide portion of the backbone.
  • Representative United States patents that teach the preparation of PNA compounds include, but are not limited to, U.S. Pat. Nos. 5,539,082; 5,714,331; and 5,719,262. Further teaching of PNA compounds can be found in Nielsen et al. ( Science , 1991, 254, 1497-1500).
  • Most preferred embodiments of the invention are oligonucleotides with phosphorothioate backbones and oligonucleosides with heteroatom backbones, and in particular —CH 2 —NH—O—CH 2 —, —CH 2 —N(CH 3 )—O—CH 2 — [known as a methylene (methylimino) or MMI backbone], —CH 2 —O—N(CH 3 ) —CH 2 —, —CH 2 —N(CH 3 )—N(CH 3 )—CH 2 — and —O—N(CH 3 )—CH 2 —CH 2 — [wherein the native phosphodiester backbone is represented as —O—P—O—CH 2 —] of the above referenced U.S.
  • Modified oligonucleotides may also contain one or more substituted sugar moieties.
  • Preferred oligonucleotides comprise one of the following at the 2′ position: OH; F; O—, S—, or N-alkyl, O-alkyl-O-alkyl, O—, S—, or N-alkenyl, or O—, S— or N-alkynyl, wherein the alkyl, alkenyl and alkynyl may be substituted or unsubstituted C 1 to C 10 alkyl or C 2 to C 10 alkenyl and alkynyl.
  • a preferred modification includes 2′-methoxyethoxy (2′-O—CH 2 CH 2 OCH 3 , also known as 2′-O-(2-methoxyethyl) or 2′-MOE) (Martin et al., Helv. Chim. Acta 1995, 78, 486-504) i.e., an alkoxyalkoxy group.
  • a further preferred modification includes 2′-dimethylaminooxyethoxy, i.e., a O(CH 2 ) 2 ON(CH 3 ) 2 group, also known as 2′-DMAOE, and 2′-dimethylamino-ethoxyethoxy (2′-DMAEOE), i.e., 2′-O—CH 2 —O—CH 2 —N(CH 2 ) 2 .
  • patents that teach the preparation of such modified sugars structures include, but are not limited to, U.S. Pat. Nos. 4,981,957; 5,118,800; 5,319,080; 5,359,044; 5,393,878; 5,446,137; 5,466,786; 5,514,785; 5,519,134; 5,567,811; 5,576,427; 5,591,722; 5,597,909; 5,610,300; 5,627,0531 5,639,873; 5,646,265; 5,658,873; 5,670,633; and 5,700,920.
  • Oligonucleotides may also include nucleobase (often referred to in the art simply as “base”) modifications or substitutions.
  • nucleobases include the purine bases adenine (A) and guanine (G), and the pyrimidine bases thymine (T), cytosine (C) and uracil (U).
  • Modified nucleobases include other synthetic and natural nucleobases such as 5-methylcytosine (5-me-C or m5c), 5-hydroxymethyl cytosine, xanthine, hypoxanthine, 2-aminoadenine, 6-methyl and other alkyl derivatives of adenine and guanine, 2-propyl and other alkyl derivatives of adenine and guanine, 2-thiouracil, 2-thiothymine and 2-thiocytosine, 5-halouracil and cytosine, 5-propynyl uracil and cytosine, 6-azo uracil, cytosine and thymine, 5-uracil (pseudouracil), 4-thiouracil, 8-halo, 8-amino, 8-thiol, 8-thioalkyl, 8-hydroxyl and other 8-substituted adenines and guanines, 5-halo particularly 5-bromo, 5-trifluoromethyl and other
  • nucleobases include those disclosed in U.S. Pat. No. 3,687,808, those disclosed in the Concise Encyclopedia Of Polymer Science And Engineering 1990, pages 858-859, Kroschwitz, J. I., ed. John Wiley & Sons, those disclosed by Englisch et al. ( Angewandte Chemie, International Edition 1991, 30, 613-722), and those disclosed by Sanghvi, Y. S., Chapter 15 , Antisense Research and Applications 1993, pages 289-302, Crooke, S. T. and Lebleu, B., ed., CRC Press. Certain of these nucleobases are particularly useful for increasing the binding affinity of the oligomeric compounds of the invention.
  • 5-substituted pyrimidines include 5-substituted pyrimidines, 6-azapyrimidines and N-2, N-6 and O-6 substituted purines, including 2-aminopropyladenine, 5-propynyluracil and 5-propynylcytosine.
  • 5-methylcytosine substitutions have been shown to increase nucleic acid duplex stability by 0.6-1.2° C. (Sanghvi, Y. S., Crooke, S. T. and Lebleu, B., eds., Antisense Research and Applications 1993, CRC Press, Boca Raton, pages 276-278) and are presently preferred base substitutions, even more particularly when combined with 2′-O-methoxyethyl sugar modifications.
  • oligonucleotides of the invention involves chemically linking to the oligonucleotide one or more moieties or conjugates which enhance the activity, cellular distribution or cellular uptake of the oligonucleotide.
  • moieties include but are not limited to lipid moieties such as a cholesterol moiety (Letsinger et al., Proc. Natl. Acad. Sci. USA 1989, 86, 6553-6556), cholic acid (Manoharan et al., Bioorg. Med. Chem. Lett .
  • a thioether e.g., hexyl-S-tritylthiol (Manoharan et al., Ann. N.Y. Acad. Sci . 1992, 660, 306-309; Manoharan et al., Bioorg. Med. Chem. Let . 1993, 3, 2765-2770), a thiocholesterol (Oberhauser et al., Nucl. Acids Res . 1992, 20, 533-538), 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, 3651-3654; Shea et al., Nucl. Acids Res .
  • 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.
  • This RNAse H-mediated cleavage of the RNA target is distinct from the use of ribozymes to cleave nucleic acids. Ribozymes are not comprehended by the present invention.
  • Examples of chimeric oligonucleotides include but are not limited to “gapmers,” in which three distinct regions are present, normally with a central region flanked by two regions which are chemically equivalent to each other but distinct from the gap.
  • a preferred example of a gapmer is an oligonucleotide in which a central portion (the “gap”) of the oligonucleotide serves as a substrate for RNase H and is preferably composed of 2′-deoxynucleotides, while the flanking portions (the 5′ and 3′ “wings”) are modified to have greater affinity for the target RNA molecule but are unable to support nuclease activity (e.g., fluoro- or 2′-O-methoxyethyl-substituted).
  • Chimeric oligonucleotides are not limited to those with modifications on the sugar, but may also include oligonucleosides or oligonucleotides with modified backbones, e.g., with regions of phosphorothioate (P ⁇ S) and phosphodiester (P ⁇ O) backbone linkages or with regions of MMI and P ⁇ S backbone linkages.
  • modified backbones e.g., with regions of phosphorothioate (P ⁇ S) and phosphodiester (P ⁇ O) backbone linkages or with regions of MMI and P ⁇ S backbone linkages.
  • Other chimeras include “wingmers,” also known in the art as “hemimers,” that is, oligonucleotides with two distinct regions.
  • the 5′ portion of the oligonucleotide serves as a substrate for RNase H and is preferably composed of 2′-deoxynucleotides, 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′-O-methoxyethyl-substituted), or vice-versa.
  • the oligonucleotides of the present invention contain a 2′-O-methoxyethyl (2′-O—CH 2 CH 2 OCH 3 ) modification on the sugar moiety of at least one nucleotide.
  • nucleotide subunits of the oligonucleotides of the invention may bear a 2′-O-methoxyethyl (—O—CH 2 CH 2 OCH 3 ) modification.
  • Oligonucleotides comprising a plurality of nucleotide subunits having a 2′-O-methoxyethyl modification can have such a modification on any of the nucleotide subunits within the oligonucleotide, and may be chimeric oligonucleotides.
  • oligonucleotides containing other modifications which enhance antisense efficacy, potency or target affinity are also preferred. Chimeric oligonucleotides comprising one or more such modifications are presently preferred.
  • 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 Applied Biosystems. Any other means for such synthesis may also be employed; the actual synthesis of the oligonucleotides is well within the talents of the routineer. It is well known to use similar techniques to prepare oligonucleotides such as the phosphorothioates and 2′-alkoxy or 2′-alkoxyalkoxy derivatives, including 2′-O-methoxyethyl oligonucleotides (Martin, P., Helv. Chim. Acta 1995, 78, 486-504).
  • CPG controlled-pore glass
  • the antisense compounds of the present invention include bioequivalent compounds, including pharmaceutically acceptable salts and prodrugs. This is intended to 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 pharmaceutically acceptable salts of the nucleic acids of the invention and prodrugs of such nucleic acids.
  • Pharmaceutically acceptable salts are physiologically and pharmaceutically acceptable salts of the nucleic acids of the invention: i.e., salts that retain the desired biological activity of the parent compound and do not impart undesired toxicological effects thereto (see, for example, Berge et al., “Pharmaceutical Salts,” J. of Pharma Sci . 1977, 66, 1-19).
  • 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, polygalactu
  • the oligonucleotides of the invention may additionally or alternatively be prepared to be delivered in a Aprodrug form.
  • Aprodrug 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 Dec. 9, 1993.
  • oligonucleotides are administered in accordance with this invention.
  • Oligonucleotide compounds of the invention may be formulated in a pharmaceutical composition, which may include pharmaceutically acceptable carriers, thickeners, diluents, buffers, preservatives, surface active agents, neutral or cationic lipids, lipid complexes, liposomes, penetration enhancers, carrier compounds and other pharmaceutically acceptable carriers or excipients and the like in addition to the oligonucleotide.
  • a pharmaceutical composition which may include pharmaceutically acceptable carriers, thickeners, diluents, buffers, preservatives, surface active agents, neutral or cationic lipids, lipid complexes, liposomes, penetration enhancers, carrier compounds and other pharmaceutically acceptable carriers or excipients and the like in addition to the oligonucleotide.
  • Such compositions and formulations are comprehended by the present invention.
  • compositions comprising the oligonucleotides of the present invention may 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., Critical Reviews in Therapeutic Drug Carrier Systems 1991, 8, 91-192; Muranishi, Critical Reviews in Therapeutic Drug Carrier Systems 1990, 7, 1-33).
  • One or more penetration enhancers from one or more of these broad categories may be included.
  • 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 salt includes any of the naturally occurring components of bile as well as any of their synthetic derivatives.
  • Complex formulations comprising one or more penetration enhancers may be used.
  • bile salts may be used in combination with fatty acids to make complex formulations.
  • Chelating agents include, but are not limited to, disodium ethylenediaminetetraacetate (EDTA), citric acid, salicylates (e.g., sodium salicylate, 5-methoxysalicylate and homovanilate), N-acyl derivatives of collagen, laureth-9 and N-amino acyl derivatives of beta-diketones (enamines) [Lee et al., Critical Reviews in Therapeutic Drug Carrier Systems 1991, page 92; Muranishi, Critical Reviews in Therapeutic Drug Carrier Systems 1990, 7, 1-33; Buur et al., J. Control Rel . 1990, 14, 43-51). Chelating agents have the added advantage of also serving as DNase inhibitors.
  • Surfactants include, for example, sodium lauryl sulfate, polyoxyethylene-9-lauryl ether and polyoxyethylene-20-cetyl ether (Lee et al., Critical Reviews in Therapeutic Drug Carrier Systems 1991, page 92); and perfluorochemical emulsions, such as FC-43 (Takahashi et al., J. Pharm. Phamacol . 1988, 40, 252-257).
  • Non-surfactants include, for example, unsaturated cyclic ureas, 1-alkyl- and 1-alkenylazacyclo-alkanone derivatives (Lee et al., Critical 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, 621-626).
  • 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.
  • 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., pregelatinized 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. Pat. Nos
  • 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, liposomes and lipid:oligonucleotide complexes of uncharacterized structure.
  • a preferred colloidal dispersion system is a plurality of liposomes.
  • Liposomes are microscopic spheres having an aqueous core surrounded by one or more outer layers made up of lipids arranged in a bilayer configuration (see, generally, Chonn et al., Current Op. Biotech . 1995, 6, 698-708).
  • compositions of the present invention may be administered in a number of ways depending upon whether local or systemic treatment is desired and upon the area to be treated. Administration may be topical (including ophthalmic, vaginal, rectal, intranasal, epidermal, and transdermal), oral or parenteral. Parenteral administration includes intravenous drip, subcutaneous, intraperitoneal or intramuscular injection, pulmonary administration, e.g., by inhalation or insufflation, or intracranial, e.g., intrathecal or intraventricular, administration. Oligonucleotides with at least one 2′-O-methoxyethyl modification are believed to be particularly useful for oral administration.
  • Formulations for topical administration may include transdermal patches, ointments, lotions, creams, gels, drops, suppositories, sprays, liquids and powders.
  • Conventional pharmaceutical carriers, aqueous, powder or oily bases, thickeners and the like may be necessary or desirable.
  • Coated condoms, gloves and the like may also be useful.
  • compositions for oral administration include powders or granules, suspensions or solutions in water or non-aqueous media, capsules, sachets or tablets. Thickeners, flavoring agents, diluents, emulsifiers, dispersing aids or binders may be desirable.
  • compositions for parenteral administration may include sterile aqueous solutions which may also contain buffers, diluents and other suitable additives. 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.
  • 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, hydroxyure
  • 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).
  • 5-FU and oligonucleotide e.g., 5-FU and oligonucleotide
  • sequentially e.g., 5-FU and oligonucleotide for a period of time followed by MTX and oligonucleotide
  • one or more other such chemotherapeutic agents e.g., 5-FU, MTX and oligonucleotide, or 5-FU, radiotherapy and oligonucleotide.
  • compositions and their subsequent administration is believed to be within the skill of those in the art. 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 vitro and in 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. Persons of ordinary skill in the art can easily estimate repetition rates for dosing based on measured residence times and concentrations of the drug in bodily fluids or tissues. Following successful treatment, it may be desirable to have the patient undergo maintenance therapy to prevent the recurrence of the disease state, wherein the oligonucleotide is administered in maintenance doses, ranging from 0.01 ⁇ g to 100 g per kg of body weight, once or more daily, to once every 20 years.
  • Unmodified oligodeoxynucleotides are synthesized on an automated DNA synthesizer (Applied Biosystems model 380B) using standard phosphoramidite chemistry with oxidation by iodine. ⁇ -cyanoethyldiisopropyl-phosphoramidites are purchased from Applied Biosystems (Foster City, Calif.). For phosphorothioate oligonucleotides, the standard oxidation bottle was replaced by a 0.2 M solution of 3 H-1,2-benzodithiole-3-one 1,1-dioxide in acetonitrile for the stepwise thiation of the phosphite linkages.
  • Cytosines may be 5-methyl cytosines. (5-methyl deoxycytidine phosphoramidites available from Glen Research, Sterling, Va. or Amersham Pharmacia Biotech, Piscataway, N.J.)
  • 2′-methoxy oligonucleotides are synthesized using 2′-methoxy ⁇ -cyanoethyldiisopropyl-phosphoramidites (Chemgenes, Needham, Mass.) and the standard cycle for unmodified oligonucleotides, except the wait step after pulse delivery of tetrazole and base is increased to 360 seconds.
  • Other 2′-alkoxy oligonucleotides are synthesized by a modification of this method, using appropriate 2′-modified amidites such as those available from Glen Research, Inc., Sterling, Va.
  • 2′-fluoro oligonucleotides are synthesized as described in Kawasaki et al. ( J. Med. Chem. 1993, 36, 831-841). Briefly, the protected nucleoside N 6 -benzoyl-2′-deoxy-2′-fluoroadenosine is synthesized utilizing commercially available 9- ⁇ -D-arabinofuranosyladenine as starting material and by modifying literature procedures whereby the 2′-a-fluoro atom is introduced by a S N 2-displacement of a 2′- ⁇ -O-trifyl group.
  • N 6 -benzoyl-9- ⁇ -D-arabinofuranosyladenine is selectively protected in moderate yield as the 3′,5′-ditetrahydropyranyl (THP) intermediate.
  • THP 3′,5′-ditetrahydropyranyl
  • Deprotection of the THP and N 6 -benzoyl groups is accomplished using standard methodologies and standard methods are used to obtain the 5′-dimethoxytrityl-(DMT) and 5′-DMT-3′-phosphoramidite intermediates.
  • TPDS tetraisopropyldisiloxanyl
  • 9- ⁇ -D-arabinofuranosylguanine as starting material
  • conversion to the intermediate diisobutyryl-arabinofuranosylguanosine deprotection of the TPDS group is followed by protection of the hydroxyl group with THP to give diisobutyryl di-THP protected arabinofuranosylguanine.
  • Selective O-deacylation and triflation is followed by treatment of the crude product with fluoride, then deprotection of the THP groups. Standard methodologies are used to obtain the 5′-DMT- and 5′-DMT-3′-phosphoramidites.
  • Synthesis of 2′-deoxy-2′-fluorouridine is accomplished by the modification of a known procedure in which 2,2′-anhydro-1- ⁇ -D-arabinofuranosyluracil is treated with 70% hydrogen fluoride-pyridine. Standard procedures are used to obtain the 5′-DMT and 5′-DMT-3′ phosphoramidites. 2′-deoxy-2′-fluorocytidine is synthesized via amination of 2′-deoxy-2′-fluorouridine, followed by selective protection to give N 4 -benzoyl-2′-deoxy-2′-fluorocytidine. Standard procedures are used to obtain the 5′-DMT and 5′-DMT-3′phosphoramidites.
  • 2′-(2-methoxyethyl)-modified amidites were synthesized according to Martin, P. ( Helv. Chim. Acta 1995, 78, 486-506).
  • the last nucleotide may be a deoxynucleotide.
  • 2′-O—CH 2 CH 2 OCH 3- cytosines may be 5-methyl cytosines.
  • 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.
  • a first solution was prepared by dissolving 3′-O-acetyl-2′-O-methoxyethyl-5′-O-dimethoxytrityl-5-methyluridine (96 g, 0.144 M) in CH 3 CN (700 mL) and set aside. Triethylamine (189 mL, 1.44 M) was added to a solution of triazole (90 g, 1.3 M) in CH 3 CN (1 L), cooled to ⁇ 5?C. and stirred for 0.5 h using an overhead stirrer. POCl 3 was added dropwise, over a 30 minute period, to the stirred solution maintained at 0-10° C., and the resulting mixture stirred for an additional 2 hours.
  • the first solution was added dropwise, over a 45 minute period, to the later solution.
  • the resulting reaction mixture was stored overnight in a cold room. Salts were filtered from the reaction mixture and the solution was evaporated. The residue was dissolved in EtOAc (1 L) and the insoluble solids were removed by filtration. The filtrate was washed with 1 ⁇ 300 mL of NaHCO 3 and 2 ⁇ 300 mL of saturated NaCl, dried over sodium sulfate and evaporated. The residue was triturated with EtOAc to give the title compound.
  • N 4 -Benzoyl-2′-O-methoxyethyl-5′-O-dimethoxytrityl-5-methylcytidine (74 g, 0.10 M) was dissolved in CH 2 Cl 2 (1 L).
  • Tetrazole diisopropylamine (7.1 g) and 2-cyanoethoxytetra(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 (tlc showed the reaction to be 95% complete).
  • the reaction mixture was extracted with saturated NaHCO 3 (1 ⁇ 300 mL) and saturated NaCl (3 ⁇ 300 mL).
  • 5-methyl-2′-deoxycytidine (5-me-C) containing oligonucleotides were synthesized according to published methods (Sanghvi et al., Nucl. Acids Res . 1993, 21, 3197-3203) using commercially available phosphoramidites (Glen Research, Sterling Va. or ChemGenes, Needham Mass.).
  • 2′-(Dimethylaminooxyethoxy) nucleoside amidites [also known in the art as 2′-O-(dimethylaminooxyethyl) nucleoside amidites] are prepared as described in the following paragraphs.
  • Adenosine, cytidine and guanosine nucleoside amidites are prepared similarly to the thymidine (5-methyluridine) except the exocyclic amines are protected with a benzoyl moiety in the case of adenosine and cytidine and with isobutyryl in the case of guanosine.
  • O 2 -2′-anhydro-5-methyluridine (Pro. Bio. Sint., Varese, Italy, 100.0 g, 0.416 mmol), dimethylaminopyridine (0.66 g, 0.013 eq, 0.0054 mmol) were dissolved in dry pyridine (500 ml) at ambient temperature under an argon atmosphere and with mechanical stirring tert-Butyldiphenylchlorosilane (125.8 g, 119.0 mL, 1.1 eq, 0.458 mmol) was added in one portion. The reaction was stirred for 16 h at ambient temperature. TLC (Rf 0.22, ethyl acetate) indicated a complete reaction.
  • reaction vessel was cooled to ambient and opened.
  • TLC Rf 0.67 for desired product and Rf 0.82 for ara-T side product, ethyl acetate
  • the reaction was stopped, concentrated under reduced pressure (10 to 1 mm Hg) in a warm water bath (40-100° C.) with the more extreme conditions used to remove the ethylene glycol.
  • the remaining solution can be partitioned between ethyl acetate and water.
  • the product will be in the organic phase.
  • the residue was purified by column chromatography (2 kg silica gel, ethyl acetate-hexanes gradient 1:1 to 4:1).
  • reaction vessel was removed from the ice bath and stirred at room temperature for 2 hr, the reaction monitored-by TLC (5% MeOH in CH 2 Cl 2 ).
  • Aqueous NaHCO 3 solution (5%, 10 mL) was added and extracted with ethyl acetate (2 ⁇ 20 mL). Ethyl acetate phase was dried over anhydrous Na 2 SO 4 , evaporated to dryness.
  • Residue was dissolved in a solution of 1M PPTS in MeOH (30.6 mL). Formaldehyde (20% w/w, 30 mL, 3.37 mmol) was added and the reaction mixture was stirred at room temperature for 10 minutes. Reaction mixture cooled to 10° C.
  • Triethylamine trihydrofluoride (3.91 mL, 24.0 mmol) was dissolved in dry THF and triethylamine (1.67 mL, 12 mmol, dry, kept over KOH). This mixture of triethylamine-2HF was then added to 5′-O-tert-butyldiphenylsilyl-2′-O-[N,N-dimethylaminooxyethyl]-5-methyluridine (1.40 g, 2.4 mmol) and stirred at room temperature for 24 hrs. Reaction was monitored by TLC (5% MeOH in CH 2 Cl 2 ). Solvent was removed under vacuum and the residue placed on a flash column and eluted with 10% MeOH in CH 2 Cl 2 to get 2′-O-(dimethylaminooxyethyl)-5-methyluridine (766 mg, 92.5%).
  • reaction mixture was stirred at ambient temperature for 4 hrs under inert atmosphere. The progress of the reaction was monitored by TLC (hexane:ethyl acetate 1:1). The solvent was evaporated, then the residue was dissolved in ethyl acetate (70 mL) and washed with 5% aqueous NaHCO 3 (40 mL). Ethyl acetate layer was dried over anhydrous Na 2 SO 4 and concentrated.
  • Residue obtained was chromatographed (ethyl acetate as eluent) to get 5′-O-DMT-2′-O-(2-N,N-dimethylaminooxyethyl)-5-methyluridine-3′-[(2-cyanoethyl)-N,N-diisopropylphosphoramidite] as a foam (1.04 g, 74.9%).
  • Oligonucleotides having methylene(methylimino) (MMI) backbones are synthesized according to U.S. Pat. No. 5,378,825, which is coassigned to the assignee of the present invention and is incorporated herein in its entirety.
  • various nucleoside dimers containing MMI linkages are synthesized and incorporated into oligonucleotides.
  • Other nitrogen-containing backbones are synthesized according to WO 92/20823 which is also coassigned to the assignee of the present invention and incorporated herein in its entirety.
  • Oligonucleotides having amide backbones are synthesized according to De Mesmaeker et al. ( Acc. Chem. Res . 1995, 28, 366-374).
  • the amide moiety is readily accessible by simple and well-known synthetic methods and is compatible with the conditions required for solid phase synthesis of oligonucleotides.
  • Oligonucleotides with morpholino backbones are synthesized according to U.S. Pat. No. 5,034,506 (Summerton and Weller).
  • PNA Peptide-nucleic acid
  • oligonucleotides are purified by precipitation twice out of 0.5 M NaCl with 2.5 volumes ethanol. Synthesized oligonucleotides were analyzed by polyacrylamide gel electrophoresis on denaturing gels or capillary gel electrophoresis and judged to be at least 85% full length material. The relative amounts of phosphorothioate and phosphodiester linkages obtained in synthesis were periodically checked by 31 P nuclear magnetic resonance spectroscopy, and for some studies oligonucleotides were purified by HPLC, as described by Chiang et al. ( J. Biol. Chem . 1991, 266, 18162). Results obtained with HPLC-purified material were similar to those obtained with non-HPLC purified material.
  • oligonucleotides are synthesized in 96 well plate format via solid phase P(III) phosphoramidite chemistry on an automated synthesizer capable of assembling 96 sequences simultaneously in a standard 96 well format.
  • Phosphodiester internucleotide linkages are afforded by oxidation with aqueous iodine.
  • Phosphorothioate internucleotide linkages are generated by sulfurization utilizing 3,H-1,2 benzodithilole-3-one 1,1 dioxide (Beaucage Reagent) in anhydrous acetonitrile.
  • Standard base-protected beta-cyanoethyl-di-isopropyl phosphoramidites are purchased from commercial vendors (e.g. PE-Applied Biosystems, Foster City, Calif., or Pharmacia, Piscataway, N.J.). Non-standard nucleosides are synthesized as per published methods. They are utilized as base protected beta-cyanoethyldiisopropyl phosphoramidites.
  • Oligonucleotides were cleaved from support and deprotected with concentrated NH 4 OH at elevated temperature (55-60° C.) for 12-16 hours and the released product then dried in vacuo. The dried product was then re-suspended in sterile water to afford a master plate from which all analytical and test plate samples are then diluted utilizing robotic pipettors.
  • Antisense oligonucleotides were designed to target human FAK.
  • Target sequence data are from the focal adhesion kinase (FAK) cDNA sequence published by Whitney, G.S., et al. ( DNA Cell Biol ., 1993, 12, 823-830); Genbank accession number L13616, provided herein as SEQ ID NO: 1.
  • FAK focal adhesion kinase
  • oligonucleotides were synthesized as chimeric oligonucleotides (“gapmers”), 20 nucleotides in length, composed of a central “gap” region consisting of ten 2′-deoxynucleotides, which is flanked on both sides (5′ and 3′ directions) by five-nucleotide “wings.”
  • the wings are composed of 2′-methoxyethyl (2′-MOE) nucleotides.
  • the internucleoside (backbone) linkages are phosphorothioate (P ⁇ S) throughout the oligonucleotide. All 2′-MOE cytosines were 5-methyl-cytosines.
  • oligonucleotide sequences are shown in Table 1.
  • An identical set of sequences were prepared as fully phosphorothioated oligodeoxynucleotides. These are shown in Table 2.
  • An additional set of oligonucleotides were synthesized as chimeric oligonucleotides (“gapmers”), 15 nucleotides in length, composed of a central “gap” region consisting of five 2′-deoxynucleotides, which is flanked on both sides (5′ and 3′ directions) by five-nucleotide “wings.”
  • the wings are composed of 2′-methoxyethyl (2′-MOE) nucleotides.
  • the internucleoside (backbone) linkages are phosphorothioate (P ⁇ S) throughout the oligonucleotide.
  • All 2′-MOE cytosines were 5-methyl-cytosines.
  • Table 3 An identical set of sequences were prepared as fully phosphorothioated oligodeoxynucleotides. These are shown in Table 4.
  • Human A549 lung carcinoma cells (American Type Culture Collection, Manassas, Va.) were grown in DMEM supplemented with 10% fetal bovine serum (FBS), non-essential amino acids for MEM, sodium pyruvate (1 mM), penicillin (50 U/ml) and streptomycin (50 ⁇ g/ml). All cell culture reagents were obtained from Life Technologies (Rockville, Md.).
  • the cells were washed once with OPTIMEMTM (Life Technologies, Rockville, Md.), then transfected with 400 nM oligonucleotide and 12 mg/ml LIPOFECTIN® (Life Technologies, Rockville, Md.), a 1:1 (w/w) liposome formulation of the cationic lipid N-[1-(2,3-dioleyloxy)propyl]-n,n,n-trimethylammonium chloride (DOTMA), and dioleoyl phosphotidylethanolamine (DOPE) in membrane filtered water.
  • DOTMA dioleoyl phosphotidylethanolamine
  • DOPE dioleoyl phosphotidylethanolamine
  • RNA was isolated using an ATLASTM Pure RNA isolation kit (Clontech, Palo Alto, Calif.). RNA was then separated on a 1.2% agarose-formaldehyde gel, transferred to Hybond-N+membrane (Amersham Pharmacia Biotech, Arlington Heights, Ill.), a positively charged nylon membrane. Immobilized RNA was cross-linked by exposure to UV light. Membranes were probed with either FAK or glyceraldehyde 3-phosphate dehydrogenase (G3PDH) probes. The probes were labeled by random primer using the PRIME-A-GENE 7 Labeling System, Promega, Madison, Wis.) and hybridized to the membranes. mRNA signals were quantitated by a PhosphoImager (Molecular Dynamics, Sunnyvale, Calif.).
  • oligonucleotide [0123] Several of the more active oligonucleotides were chosen for a dose response study. A549 cells were grown, treated and processed as described in Example 2, except the concentration of oligonucleotide was varied.
  • a dose response experiment on protein levels was done with two oligonucleotides.
  • A549 cells were grown and treated as described in Example 2 except the concentration was varied as shown in Table 3.
  • the LIPOFECTIN® to oligonucleotide ratio was maintained at 3 mg/ml LIPOFECTIN® per 100 nM oligonucleotide.
  • FAK protein levels were determined 48 hours after antisense treatment in whole cell lysates by anti-FAK blotting.
  • HNTG buffer 50 mM HEPES, pH 7.4, 150 mM NaCl, 0.1% Triton X-100, 10% glycerol
  • proteins were transferred to polyvinylidene fluoride membranes (Millipore) and incubated with a 1:1000 dilution of polyclonal antibody for 2 hr at room temperature. Bound primary antibody was visualized by enhanced chemiluminescent detection.
  • Results are shown in Table 8. TABLE 8 Dose Response of A549 cells to FAK Phosphorothioate Oligonucleotides SEQ ID ASO Gene % protein % protein ISIS # NO: Target Dose Expression Inhibition control — — — 100% — 15409 12 coding 25 nM 60% 40% ′′ ′′ ′′ 100 nM 57% 43% ′′ ′′ ′′ 200 nM 23% 77% 15421 18 3′-UTR 25 nM 73% 27% ′′ ′′ ′′ 100 nM 34% 66% ′′ ′′ ′′ 200 nM 24% 76%
  • Integrin-regulated focal adhesion kinase is an important component of epidermal (EGF) and platelet-drived (PDGF) growth factor-induced motility of primary fibroblasts, smooth muscle, and adenocarcinoma cells.
  • EGF epidermal
  • PDGF platelet-drived growth factor-induced motility of primary fibroblasts, smooth muscle, and adenocarcinoma cells.
  • a modified Boyden chamber (Millipore, Bedford, Mass.) assay was used (Sieg, D. J., et al., J. Cell Sci ., 1999, 112, 2677-2691).
  • Both membrane sides were coated with rat tail collagen (5 ?g/ml in PBS, Boehringer Mannheim) for 2 hr at 37° C., washed with PBS, and the chambers were placed into 24 well dishes containing migration media (0.5 ml DMEM containing 0.5% BSA) with or without human recombinant PDGF-BB, EGF, or basic-FGF (Calbiochem, San Diego, Calif.) at the indicated concentrations.
  • migration media 0.5 ml DMEM containing 0.5% BSA
  • human recombinant PDGF-BB, EGF, or basic-FGF Calbiochem, San Diego, Calif.
  • Serum-starved A549 cells (1 ⁇ 10 5 cells in 0.3 ml migration media) were added to the upper chamber and after 3 hr at 37° C., the cells on the membrane upper surface were removed by a cotton tip applicator, the migratory cells on the lower membrane surface were fixed, stained (0.1% crystal violet, 0.1 M borate pH 9.0, 2% EtOH), and the dye eluted for absorbance measurements at 600 nM.
  • Individual experiments represent the average from three individual chambers. Background levels of cell migration (less than 5% of total) in the absence of chemotaxis stimuli (0.5% BSA only) were subtracted from all points.
  • ISIS 17636 (SEQ ID NO. 43) is a five base mismatch control oligonucleotide for ISIS 15421 (SEQ ID NO. 18). TABLE 9 Effect of FAK Antisense Phosphorothioate Oligonucleotides on EGF-Stimulated Cell Migration SEQ ID ASO Gene EGF ISIS # NO: Target (ng/ml) A 600 control — — 2.5 0.74 15421 18 3′-UTR ′′ 0.26 17636 43 control ′′ 0.90 control — — 5.0 0.89 15421 18 3′-UTR ′′ 0.25 17636 43 control ′′ 0.77
  • FAK antisense oligonucleotides were tested in an in vitro invasion assay using an ⁇ 1 mm MATRIGEL® (Becton Dickinson, Franklin Lakes, N.J.) basement membrane barrier (Albini, A., Pathol. Oncol. Res ., 1998, 4, 230-241). Migration chambers were coated with the indicated concentration of MATRIGEL®, dried under laminar flow and then rehydrated with cold serum free DMEM for 90 min on an orbital shaker. A549 cells were grown and transfected as described in Example 2. Cells (1 ⁇ 10 5 ) were then placed onto the MATRIGEL® coated membrane and allowed to invade through the MATRIGEL® towards a 10% FBS chemoattractant for the indicated times. Cells that invaded through the MATRIGEL® were visualized by crystal violet staining as detailed in the migration assay. The amount of MATRIGEL® was varied in the assay to show that invasion was being measured and that the migration was not serum-induced.
  • FAK antisense oligonucleotides were tested in a rabbit model of retinal neovascularization (Kimura, H., et al., Invest. Opthalmol. Vis. Sci ., 1995, 36, 2110-2119). In this model, growth factors are encapsulated and injected beneath the retina.
  • C8161 and BL human melanoma cell lines were treated with ISIS 15421 (SEQ. ID. NO 18) or a control oligonucleotide, ISIS 29848, a 20-mer random oligonucleotide (NNNNNNNNNNNNNNNNNNNNNNNN, wherein each N is a mixture of A, C, G and T; herein incorporated as SEQ ID NO: 44) using the lipofectin protocol described herein.
  • Oligonucleotides were transfected for four hours at 300 nM in lipofectin reagent and 5-FU (200 ⁇ g/mL; SIGMA) was added after the incubation for 20 hours. Cell viability was determined by the MTT assay.
  • mice transplanted with human cancer cells or cell line tumors Another model used to investigate the efficacy of antisense oligonucleotides on tumor growth involves the use of mice transplanted with human cancer cells or cell line tumors.
  • human C8161 melanoma tumor xenografts were transplanted onto the side of nude mice with sutures or surgical staples.
  • Mice were treated with ISIS 15421 (SEQ ID NO. 18) or the control ISIS 29848 (SEQ ID No. 44) over a 28 day treatment course.
  • mice were sacrificed and tumor volumes measured. Tumor volumes in the antisense treated mice were significantly smaller than tumor volumes in control-treated mice with no observation of toxicity to the mice. Additionally, one third of the control-treated mice had grossly evident intraperitoneal metastases, while none of the antisense-treated mice displayed such metastases.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Organic Chemistry (AREA)
  • Genetics & Genomics (AREA)
  • Chemical & Material Sciences (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biomedical Technology (AREA)
  • General Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Molecular Biology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Medicinal Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Public Health (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • General Engineering & Computer Science (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Biotechnology (AREA)
  • Biophysics (AREA)
  • Physics & Mathematics (AREA)
  • Plant Pathology (AREA)
  • Microbiology (AREA)
  • Virology (AREA)
  • Biochemistry (AREA)
  • Cardiology (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Ophthalmology & Optometry (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

Compounds, compositions and methods are provided for inhibiting FAK mediated signaling. The compositions comprise antisense compounds targeted to nucleic acids encoding FAK. Methods of using these antisense compounds for inhibition of FAK expression and for treatment of diseases, particularly cancers, associated with overexpression or constitutive activation of FAK are provided.

Description

  • This application is a continuation of U.S. Application Ser. No. 09/757,100 filed Jan. 9, 2001, which is a continuation-in-part of the PCT Application No. PCT/US00/18999 filed Jul. 13, 2000 which corresponds to U.S. Application Ser. No. 09/377,310 filed Aug. 19, 1999 now issued U.S. Pat. No. 6,133,031.[0001]
  • FIELD OF THE INVENTION
  • This invention relates to compositions and methods for modulating expression of the human focal adhesion kinase (FAK) gene, which encodes a signaling protein involved in growth factor response and cell migration and is implicated in disease. This invention is also directed to methods for inhibiting FAK-mediated signal transduction; these methods can be used diagnostically or therapeutically. Furthermore, this invention is directed to treatment of conditions associated with expression of the human FAK gene. [0002]
  • BACKGROUND OF THE INVENTION
  • Cell migration is fundamental to a variety of biological processes and can be induced by both integrin receptor-mediated signals (haptotaxis migration) and/or soluble growth factor-mediated signals (chemotaxis migration). Integrin receptor engagement activates focal adhesion kinase (FAK, also pp125FAK), a non-receptor protein-tyrosine kinase localized to cell substratum-extracellular matrix (ECM) contact sites that function as part of a cytoskeletal-associated network of signaling proteins (Schlaepfer, D. D., et al., [0003] Prog. Biophys. Mol. Biol., 1999, 71, 435-478). In adherent cells, FAK is often associated with integrins at focal adhesions (Schaller, M. D., et al., Proc. Natl. Acad. Sci. USA, 1992, 89, 5192-5196). Numerous other signaling proteins, including other protein tyrosine kinases are associated with FAK at these regions. Phosphorylation of FAK results in activation of the mitogen-activated protein kinase pathway. In addition, FAK regulates activation of phosphatidylinositol 3′-kinase which may serve to prevent apoptosis. FAK has also been shown to be required for internalization of bacteria mediated by invasin (Alrutz, M. A. and Isberg, R. R., Proc. Natl. Acad. Sci. USA, 1998, 95, 13658-13663).
  • Normal cells typically require anchorage to the extracellular matrix in order to grow. When these cells are removed from the extracellular matrix, they undergo apoptosis. Transformed cells, on the other hand, can grow under anchorage-independent conditions, providing them a growth advantage and the ability to be removed from their normal cellular environment. [0004]
  • Overexpression of FAK is involved in cancer progression. High levels of FAK correlates with invasiveness and metastatic potential in colon tumors (Weiner, T. M., et al., [0005] Lancet, 1993, 342, 1024-1025), breast tumors (Owens, L. V., et al., Cancer Res., 1995, 55, 2752-2755), and oral cancers (Kornberg, L. J., Head Neck, 1998, 20, 634-639).
  • FAK's role in cell migration has led to the speculation that it may be relevant in other diseases such as embryonic development disfunctions and angiogenic disorders (Kornberg, L. J., [0006] Head Neck, 1998, 20, 634-639).
  • There is a lack of specific inhibitors of FAK. Antisense approaches have been a means by which the function of FAK has been investigated. Lou, J. et al. ([0007] J. Orthopaedic Res., 1997, 15, 911-918) used an adenoviral based vector to express antisense FAK RNA to show that FAK is involved in wound healing in tendons. Another antisense FAK expression vector containing 400 bp of complementary sequence was used to study the interaction of type I collagen and ?2?1 integrin (Takeuchi, Y., et al., J. Biol. Chem., 1997, 272, 29309-29316).
  • Antisense oligonucleotides have been used in several studies. Tanaka, S. et al. ([0008] J. Cell. Biochem., 1995, 58, 424-435) disclose two antisense phosphorothioate oligonucleotides targeted to the start site of mouse FAK. Xu, L.-H., et al. (Cell Growth Diff., 1996, 7, 413-418) disclose two antisense phosphorothioate oligonucleotides targeted within the coding region of human FAK. They also show that FAK antisense treatment could induce apoptosis in tumor cells. Sonoda, Y., et al. (Biochem. Biophys. Res. Comm., 1997, 241, 769-774) also demonstrated a role for FAK in apoptosis using antisense phosphorothioate oligonucleotides targeted to the start site and within the coding region of human FAK. Shibata, K., et al. (Cancer Res., 1998, 58, 900-903) disclose antisense phosphorothioate oligonucleotides targeted to the start site and coding region of human FAK. Narase, K., et al. (Oncogene, 1998, 17, 455-463) disclose an antisense phosphorothioate oligonucleotide targeted to the start site of human FAK.
  • There remains a long-felt need for improved compositions and methods for inhibiting FAK gene expression. [0009]
  • SUMMARY OF THE INVENTION
  • The present invention provides antisense compounds which are targeted to nucleic acids encoding focal adhesion kinase expression (FAK) and are capable of modulating FAK mediated signaling. The present invention also provides chimeric oligonucleotides targeted to nucleic acids encoding human FAK. The antisense compounds of the invention are believed to be useful both diagnostically and therapeutically, and are believed to be particularly useful in the methods of the present invention. [0010]
  • The present invention also comprises methods of modulating FAK mediated signaling, in cells and tissues, using the antisense compounds of the invention. Methods of inhibiting FAK expression are provided; these methods are believed to be useful both therapeutically and diagnostically. These methods are also useful as tools, for example, for detecting and determining the role of FAK in various cell functions and physiological processes and conditions and for diagnosing conditions associated with expression of FAK. [0011]
  • The present invention also comprises methods for diagnosing and treating cancers, including those of the colon, breast and mouth. These methods are believed to be useful, for example, in diagnosing FAK-associated disease progression. These methods employ the antisense compounds of the invention. These methods are believed to be useful both therapeutically, including prophylactically, and as clinical research and diagnostic tools.[0012]
  • DETAILED DESCRIPTION OF THE INVENTION
  • FAK plays important roles in integrin-mediated signal transduction. Overexpression of FAK is associated with tumor progression and metastatic potential. As such, this protein represents an attractive target for treatment of such diseases. In particular, modulation of the expression of FAK may be useful for the treatment of diseases such as colon cancer, breast cancer and cancer of the mouth. [0013]
  • The present invention employs antisense compounds, particularly oligonucleotides, for use in modulating the function of nucleic acid molecules encoding FAK, ultimately modulating the amount of FAK produced. This is accomplished by providing oligonucleotides which specifically hybridize with nucleic acids, preferably mRNA, encoding FAK. [0014]
  • This relationship between an antisense compound such as an oligonucleotide and its complementary nucleic acid target, to which it hybridizes, is commonly referred to as “antisense”. “Targeting” an oligonucleotide to a chosen nucleic acid target, in the context of this invention, is a multistep process. The process usually begins with identifying a nucleic acid sequence whose function is to be modulated. This may be, as examples, a cellular gene (or mRNA made from the gene) whose expression is associated with a particular disease state, or a foreign nucleic acid from an infectious agent. In the present invention, the targets are nucleic acids encoding FAK; in other words, a gene encoding FAK, or mRNA expressed from the FAK gene. mRNA which encodes FAK is presently the preferred target. The targeting process also includes determination of a site or sites within the nucleic acid sequence for the antisense interaction to occur such that modulation of gene expression will result. [0015]
  • In accordance with this invention, persons of ordinary skill in the art will understand that messenger RNA includes not only the information to encode a protein using the three letter genetic code, but also associated ribonucleotides which form a region known to such persons as the 5′-untranslated region, the 3′-untranslated region, the 5′ cap region and intron/exon junction ribonucleotides. Thus, oligonucleotides may be formulated in accordance with this invention which are targeted wholly or in part to these associated ribonucleotides as well as to the informational ribonucleotides. The oligonucleotide may therefore be specifically hybridizable with a transcription initiation site region, a translation initiation codon region, a 5′ cap region, an intron/exon junction, coding sequences, a translation termination codon region or sequences in the 5′- or 3′-untranslated region. Since, 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. 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 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 FAK, 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,” “AUG region” and “translation initiation codon 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. This region is a preferred target region. Similarly, the terms “stop codon region” and “translation termination codon 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. This region is a preferred target region. The open reading frame (ORF) or “coding region,” which is known in the art to refer to the region between the translation initiation codon and the translation termination codon, is also a region which may be targeted effectively. Other preferred target regions include the 5′ untranslated region (5′UTR), known in the art to refer to the portion of an mRNA in the 5′ direction from the translation initiation codon, and thus including nucleotides between the 5′ cap site and the translation initiation codon of an mRNA or corresponding nucleotides on the gene and the 3′ untranslated region (3′UTR), known in the art to refer to the portion of an mRNA in the 3′ direction from the translation termination codon, and thus including nucleotides between the translation termination codon and 3′ end of an mRNA or corresponding nucleotides on the gene. The 5′ cap of an mRNA comprises an N7-methylated guanosine residue joined to the 5′-most residue of the mRNA via a 5′-5′ triphosphate linkage. The 5′ cap region of an mRNA is considered to include the 5′ cap structure itself as well as the first 50 nucleotides adjacent to the cap. The 5′ cap region may also be a preferred target region. [0016]
  • Although some eukaryotic mRNA transcripts are directly translated, many contain one or more regions, known as “introns”, which are excised from a pre-mRNA transcript to yield one or more mature mRNA. The remaining (and therefore translated) regions are known as “exons” and are spliced together to form a continuous mRNA sequence. mRNA splice sites, i.e., exon-exon or intron-exon junctions, may also be preferred target regions, and are particularly useful in situations where aberrant splicing is implicated in disease, or where an overproduction of a particular mRNA splice product is implicated in disease. Aberrant fusion junctions due to rearrangements or deletions are also preferred targets. Targeting particular exons in alternatively spliced mRNAs may also be preferred. It has also been found that introns can also be effective, and therefore preferred, target regions for antisense compounds targeted, for example, to DNA or pre-mRNA. [0017]
  • 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 modulation. [0018]
  • “Hybridization”, in the context of this invention, means hydrogen bonding, also known as Watson-Crick base pairing, between complementary bases, usually on opposite nucleic acid strands or two regions of a nucleic acid strand. Guanine and cytosine are examples of complementary bases which are known to form three hydrogen bonds between them. Adenine and thymine are examples of complementary bases which form two hydrogen bonds between them. [0019]
  • “Specifically hybridizable” and “complementary” are terms which are used to indicate a sufficient degree of complementarity such that stable and specific binding occurs between the DNA or RNA target and the oligonucleotide. [0020]
  • It is understood that an oligonucleotide need not be 100% complementary to its target nucleic acid sequence to be specifically hybridizable. An oligonucleotide is specifically hybridizable when binding of the oligonucleotide to the target interferes with the normal function of the target molecule to cause a loss of utility, and there is a sufficient degree of complementarity to avoid non-specific binding of the oligonucleotide to non-target sequences under conditions in which specific binding is desired, i.e., under physiological conditions in the case of in vivo assays or therapeutic treatment or, in the case of in vitro assays, under conditions in which the assays are conducted. [0021]
  • Hybridization of antisense oligonucleotides with mRNA interferes with one or more of the normal functions of mRNA. The functions of mRNA to be interfered with include all vital functions such as, for example, translocation of the RNA to the site of protein translation, translation of protein from the RNA, splicing of the RNA to yield one or more mRNA species, and catalytic activity which may be engaged in by the RNA. Binding of specific protein(s) to the RNA may also be interfered with by antisense oligonucleotide hybridization to the RNA. [0022]
  • The overall effect of interference with mRNA function is modulation of expression of FAK. In the context of this invention “modulation” means either inhibition or stimulation; i.e., either a decrease or increase in expression. This modulation can be measured in ways which are routine in the art, for example by Northern blot assay of mRNA expression, or reverse transcriptase PCR, as taught in the examples of the instant application or by Western blot or ELISA assay of protein expression, or by an immunoprecipitation assay of protein expression. Effects on cell proliferation or tumor cell growth can also be measured, as taught in the examples of the instant application. Inhibition is presently preferred. [0023]
  • The oligonucleotides of this invention can be used in diagnostics, therapeutics, prophylaxis, and as research reagents and in kits. Since the oligonucleotides of this invention hybridize to nucleic acids encoding FAK, sandwich, calorimetric and other assays can easily be constructed to exploit this fact. Provision of means for detecting hybridization of oligonucleotide with the FAK genes or mRNA can routinely be accomplished. Such provision may include enzyme conjugation, radiolabelling or any other suitable detection systems. Kits for detecting the presence or absence of FAK may also be prepared. [0024]
  • The present invention is also suitable for diagnosing abnormal inflammatory states or certain cancers in tissue or other samples from patients suspected of having an autoimmune or inflammatory disease such as hepatitis or cancers such as those of the colon, liver or lung, and lymphomas. A number of assays may be formulated employing the present invention, which assays will commonly comprise contacting a tissue sample with an oligonucleotide of the invention under conditions selected to permit detection and, usually, quantitation of such inhibition. In the context of this invention, to “contact” tissues or cells with an oligonucleotide or oligonucleotides means to add the oligonucleotide(s), usually in a liquid carrier, to a cell suspension or tissue sample, either in vitro or ex vivo, or to administer the oligonucleotide(s) to cells or tissues within an animal. [0025]
  • The oligonucleotides of this invention may also be used for research purposes. Thus, the specific hybridization exhibited by the oligonucleotides 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. [0026]
  • 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. [0027]
  • The antisense compounds in accordance with this invention preferably comprise from about 5 to about 50 nucleobases. Particularly preferred are antisense oligonucleotides comprising from about 8 to about 30 nucleobases (i.e. from about 8 to about 30 linked nucleosides). As is known in the art, a nucleoside is a base-sugar combination. The base portion of the nucleoside is normally a heterocyclic base. The two most common classes of such heterocyclic bases are the purines and the pyrimidines. Nucleotides are nucleosides that further include a phosphate group covalently linked to the sugar portion of the nucleoside. For those nucleosides that include a pentofuranosyl sugar, the phosphate group can be linked to either the 2=, 3= or 5=hydroxyl moiety of the sugar. In forming oligonucleotides, the phosphate groups covalently link adjacent nucleosides to one another to form a linear polymeric compound. In turn the respective ends of this linear polymeric structure can be further joined to form a circular structure, however, open linear structures are generally preferred. Within the oligonucleotide structure, the phosphate groups are commonly referred to as forming the internucleoside backbone of the oligonucleotide. The normal linkage or backbone of RNA and DNA is a 3= to 5=phosphodiester linkage. [0028]
  • Specific examples of preferred antisense compounds useful in this invention include oligonucleotides containing modified backbones or non-natural internucleoside linkages. As defined in this specification, oligonucleotides having modified backbones include those that retain a phosphorus atom in the backbone and those that do not have a phosphorus atom in the backbone. For the purposes of this specification, and as sometimes referenced in the art, modified oligonucleotides that do not have a phosphorus atom in their internucleoside backbone can also be considered to be oligonucleosides. [0029]
  • Preferred modified oligonucleotide backbones include, for example, phosphorothioates, chiral phosphorothioates, phosphorodithioates, phosphotriesters, aminoalkylphosphotriesters, methyl and other alkyl phosphonates including 3=-alkylene phosphonates and chiral phosphonates, phosphinates, phosphoramidates including 3=-amino phosphoramidate and aminoalkylphosphoramidates, thionophosphoramidates, thionoalkylphosphonates, thionoalkylphosphotriesters, and boranophosphates having normal 3=-5=linkages, 2=-5=linked analogs of these, and those having inverted polarity wherein the adjacent pairs of nucleoside units are linked 3=-5= to 5=-3= or 2=-5= to 5=-2=. Various salts, mixed salts and free acid forms are also included. [0030]
  • Representative United States patents that teach the preparation of the above phosphorus-containing linkages include, but are not limited to, U.S. Pat. Nos. 3,687,808; 4,469,863; 4,476,301; 5,023,243; 5,177,196; 5,188,897; 5,264,423; 5,276,019; 5,278,302; 5,286,717; 5,321,131; 5,399,676; 5,405,939; 5,453,496; 5,455,233; 5,466,677; 5,476,925; 5,519,126; 5,536,821; 5,541,306; 5,550,111; 5,563,253; 5,571,799; 5,587,361; and 5,625,050. [0031]
  • Preferred modified oligonucleotide backbones that do not include a phosphorus atom therein have backbones that are formed by short chain alkyl or cycloalkyl internucleoside linkages, mixed heteroatom and alkyl or cycloalkyl internucleoside linkages, or one or more short chain heteroatomic or heterocyclic internucleoside linkages. These include those having morpholino linkages (formed in part from the sugar portion of a nucleoside); siloxane backbones; sulfide, sulfoxide and sulfone backbones; formacetyl and thioformacetyl backbones; methylene formacetyl and thioformacetyl backbones; alkene containing backbones; sulfamate backbones; methyleneimino and methylenehydrazino backbones; sulfonate and sulfonamide backbones; amide backbones; and others having mixed N, O, S and CH[0032] 2 component parts.
  • Representative United States patents that teach the preparation of the above oligonucleosides include, but are not limited to, U.S. Pat. Nos. 5,034,506; 5,166,315; 5,185,444; 5,214,134; 5,216,141; 5,235,033; 5,264,562; 5,264,564; 5,405,938; 5,434,257; 5,466,677; 5,470,967; 5,489,677; 5,541,307; 5,561,225; 5,596,086; 5,602,240; 5,610,289; 5,602,240; 5,608,046; 5,610,289; 5,618,704; 5,623,070; 5,663,312; 5,633,360; 5,677,437; and 5,677,439. [0033]
  • In other preferred oligonucleotide mimetics, both the sugar and the internucleoside linkage, i.e., the backbone, of the nucleotide units are replaced with novel groups. The base units are maintained for hybridization with an appropriate nucleic acid target compound. One such oligomeric compound, an oligonucleotide mimetic that has been shown to have excellent hybridization properties, is referred to as a peptide nucleic acid (PNA). In PNA compounds, the sugar-backbone of an oligonucleotide is replaced with an amide containing backbone, in particular an aminoethylglycine backbone. The nucleobases are retained and are bound directly or indirectly to aza nitrogen atoms of the amide portion of the backbone. Representative United States patents that teach the preparation of PNA compounds include, but are not limited to, U.S. Pat. Nos. 5,539,082; 5,714,331; and 5,719,262. Further teaching of PNA compounds can be found in Nielsen et al. ([0034] Science, 1991, 254, 1497-1500).
  • Most preferred embodiments of the invention are oligonucleotides with phosphorothioate backbones and oligonucleosides with heteroatom backbones, and in particular —CH[0035] 2—NH—O—CH2—, —CH2—N(CH3)—O—CH2— [known as a methylene (methylimino) or MMI backbone], —CH2—O—N(CH3) —CH2—, —CH2—N(CH3)—N(CH3)—CH2— and —O—N(CH3)—CH2—CH2— [wherein the native phosphodiester backbone is represented as —O—P—O—CH2—] of the above referenced U.S. Pat. No. 5,489,677, and the amide backbones of the above referenced U.S. Pat. No. 5,602,240. Also preferred are oligonucleotides having morpholino backbone structures of the above-referenced U.S. Pat. No. 5,034,506.
  • Modified oligonucleotides may also contain one or more substituted sugar moieties. Preferred oligonucleotides comprise one of the following at the 2′ position: OH; F; O—, S—, or N-alkyl, O-alkyl-O-alkyl, O—, S—, or N-alkenyl, or O—, S— or N-alkynyl, wherein the alkyl, alkenyl and alkynyl may be substituted or unsubstituted C[0036] 1 to C10 alkyl or C2 to C10 alkenyl and alkynyl. Particularly preferred are O[(CH2)nO]mCH3, O(CH2)nOCH3, O(CH2)2ON(CH3)2, O(CH2)nNH2, O(CH2)nCH3, O(CH2)nONH2, and O(CH2)nON[(CH2)nCH3)]2, where n and m are from 1 to about 10. Other preferred oligonucleotides comprise one of the following at the 2=position: C1 to C10 lower alkyl, substituted lower alkyl, alkaryl, aralkyl, O-alkaryl or O-aralkyl, SH, SCH3, OCN, Cl, Br, CN, CF3, OCF3, SOCH3, SO2CH3, ONO2, NO2, 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′-O—CH2CH2OCH3, also known as 2′-O-(2-methoxyethyl) or 2′-MOE) (Martin et al., Helv. Chim. Acta 1995, 78, 486-504) i.e., an alkoxyalkoxy group. A further preferred modification includes 2′-dimethylaminooxyethoxy, i.e., a O(CH2)2ON(CH3)2 group, also known as 2′-DMAOE, and 2′-dimethylamino-ethoxyethoxy (2′-DMAEOE), i.e., 2′-O—CH2—O—CH2—N(CH2)2.
  • Other preferred modifications include 2′-methoxy (2′-O—CH[0037] 3), 2′-aminopropoxy (2′-OCH2CH2CH2NH2) and 2′-fluoro (2′-F). Similar modifications may also be made at other positions on the oligonucleotide, particularly the 3′ position of the sugar on the 3′ terminal nucleotide or in 2=-5=linked oligonucleotides and the 5′ position of 5′ terminal nucleotide. Oligonucleotides may also have sugar mimetics such as cyclobutyl moieties in place of the pentofuranosyl sugar. Representative U.S. patents that teach the preparation of such modified sugars structures include, but are not limited to, U.S. Pat. Nos. 4,981,957; 5,118,800; 5,319,080; 5,359,044; 5,393,878; 5,446,137; 5,466,786; 5,514,785; 5,519,134; 5,567,811; 5,576,427; 5,591,722; 5,597,909; 5,610,300; 5,627,0531 5,639,873; 5,646,265; 5,658,873; 5,670,633; and 5,700,920.
  • Oligonucleotides may also include nucleobase (often referred to in the art simply as “base”) modifications or substitutions. As used herein, “unmodified” or “natural” nucleobases include the purine bases adenine (A) and guanine (G), and the pyrimidine bases thymine (T), cytosine (C) and uracil (U). Modified nucleobases include other synthetic and natural nucleobases such as 5-methylcytosine (5-me-C or m5c), 5-hydroxymethyl cytosine, xanthine, hypoxanthine, 2-aminoadenine, 6-methyl and other alkyl derivatives of adenine and guanine, 2-propyl and other alkyl derivatives of adenine and guanine, 2-thiouracil, 2-thiothymine and 2-thiocytosine, 5-halouracil and cytosine, 5-propynyl uracil and cytosine, 6-azo uracil, cytosine and thymine, 5-uracil (pseudouracil), 4-thiouracil, 8-halo, 8-amino, 8-thiol, 8-thioalkyl, 8-hydroxyl and other 8-substituted adenines and guanines, 5-halo particularly 5-bromo, 5-trifluoromethyl and other 5-substituted uracils and cytosines, 7-methylguanine and 7-methyladenine, 8-azaguanine and 8-azaadenine, 7-deazaguanine and 7-deazaadenine and 3-deazaguanine and 3-deazaadenine. Further nucleobases include those disclosed in U.S. Pat. No. 3,687,808, those disclosed in the [0038] Concise Encyclopedia Of Polymer Science And Engineering 1990, pages 858-859, Kroschwitz, J. I., ed. John Wiley & Sons, those disclosed by Englisch et al. (Angewandte Chemie, International Edition 1991, 30, 613-722), and those disclosed by Sanghvi, Y. S., Chapter 15, Antisense Research and Applications 1993, pages 289-302, Crooke, S. T. and Lebleu, B., ed., CRC Press. Certain of these nucleobases are particularly useful for increasing the binding affinity of the oligomeric compounds of the invention. These include 5-substituted pyrimidines, 6-azapyrimidines and N-2, N-6 and O-6 substituted purines, including 2-aminopropyladenine, 5-propynyluracil and 5-propynylcytosine. 5-methylcytosine substitutions have been shown to increase nucleic acid duplex stability by 0.6-1.2° C. (Sanghvi, Y. S., Crooke, S. T. and Lebleu, B., eds., Antisense Research and Applications 1993, CRC Press, Boca Raton, pages 276-278) and are presently preferred base substitutions, even more particularly when combined with 2′-O-methoxyethyl sugar modifications.
  • Representative U.S. patents that teach the preparation of certain of the above noted modified nucleobases as well as other modified nucleobases include, but are not limited to, the above noted U.S. Pat. No. 3,687,808, as well as U.S. Pat. Nos. 4,845,205; 5,130,302; 5,134,066; 5,175,273; 5,367,066; 5,432,272; 5,457,187; 5,459,255; 5,484,908; 5,502,177; 5,525,711; 5,552,540; 5,587,469; 5,594,121, 5,596,091; 5,614,617; and 5,681,941. [0039]
  • Another modification of the oligonucleotides of the invention involves chemically linking to the oligonucleotide one or more moieties or conjugates which enhance the activity, cellular distribution or cellular uptake of the oligonucleotide. Such moieties include but are not limited to lipid moieties such as a cholesterol moiety (Letsinger et al., [0040] Proc. Natl. Acad. Sci. USA 1989, 86, 6553-6556), cholic acid (Manoharan et al., Bioorg. Med. Chem. Lett. 1994, 4, 1053-1059), a thioether, e.g., hexyl-S-tritylthiol (Manoharan et al., Ann. N.Y. Acad. Sci. 1992, 660, 306-309; Manoharan et al., Bioorg. Med. Chem. Let. 1993, 3, 2765-2770), a thiocholesterol (Oberhauser et al., Nucl. Acids Res. 1992, 20, 533-538), an aliphatic chain, e.g., dodecandiol or undecyl residues (Saison-Behmoaras et al., EMBO J. 1991, 10, 1111-1118; Kabanov et al., FEBS Lett. 1990, 259, 327-330; Svinarchuk et al., Biochimie 1993, 75, 49-54), 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, 3651-3654; Shea et al., Nucl. Acids Res. 1990, 18, 3777-3783), a polyamine or a polyethylene glycol chain (Manoharan et al., Nucleosides & Nucleotides 1995, 14, 969-973), or adamantane acetic acid (Manoharan et al., Tetrahedron Lett. 1995, 36, 3651-3654), a palmityl moiety (Mishra et al., Biochim. Biophys. Acta 1995, 1264, 229-237), or an octadecylamine or hexylamino-carbonyl-oxycholesterol moiety (Crooke et al., J. Pharmacol. Exp. Ther. 1996, 277, 923-937).
  • Representative U.S. patents that teach the preparation of such oligonucleotide conjugates include, but are not limited to, U.S. Pat. Nos. 4,828,979; 4,948,882; 5,218,105; 5,525,465; 5,541,313; 5,545,730; 5,552,538; 5,578,717; 5,580,731; 5,580,731; 5,591,584; 5,109,124; 5,118,802; 5,138,045; 5,414,077; 5,486,603; 5,512,439; 5,578,718; 5,608,046; 4,587,044; 4,605,735; 4,667,025; 4,762,779; 4,789,737; 4,824,941; 4,835,263; 4,876,335; 4,904,582; 4,958,013; 5,082,830; 5,112,963; 5,214,136; 5,082,830; 5,112,963; 5,214,136; 5,245,022; 5,254,469; 5,258,506; 5,262,536; 5,272,250; 5,292,873; 5,317,098; 5,371,241; 5,391,723; 5,416,203; 5,451,463; 5,510,475; 5,512,667; 5,514,785; 5,565,552; 5,567,810; 5,574,142; 5,585,481; 5,587,371; 5,595,726; 5,597,696; 5,599,923; 5,599,928 and 5,688,941. [0041]
  • 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. This RNAse H-mediated cleavage of the RNA target is distinct from the use of ribozymes to cleave nucleic acids. Ribozymes are not comprehended by the present invention. [0042]
  • Examples of chimeric oligonucleotides include but are not limited to “gapmers,” in which three distinct regions are present, normally with a central region flanked by two regions which are chemically equivalent to each other but distinct from the gap. A preferred example of a gapmer is an oligonucleotide in which a central portion (the “gap”) of the oligonucleotide serves as a substrate for RNase H and is preferably composed of 2′-deoxynucleotides, while the flanking portions (the 5′ and 3′ “wings”) are modified to have greater affinity for the target RNA molecule but are unable to support nuclease activity (e.g., fluoro- or 2′-O-methoxyethyl-substituted). Chimeric oligonucleotides are not limited to those with modifications on the sugar, but may also include oligonucleosides or oligonucleotides with modified backbones, e.g., with regions of phosphorothioate (P═S) and phosphodiester (P═O) backbone linkages or with regions of MMI and P═S backbone linkages. Other chimeras include “wingmers,” also known in the art as “hemimers,” that is, oligonucleotides with two distinct regions. In a preferred example of a wingmer, the 5′ portion of the oligonucleotide serves as a substrate for RNase H and is preferably composed of 2′-deoxynucleotides, 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′-O-methoxyethyl-substituted), or vice-versa. In one embodiment, the oligonucleotides of the present invention contain a 2′-O-methoxyethyl (2′-O—CH[0043] 2CH2OCH3) modification on the sugar moiety of at least one nucleotide. This modification has been shown to increase both affinity of the oligonucleotide for its target and nuclease resistance of the oligonucleotide. According to the invention, one, a plurality, or all of the nucleotide subunits of the oligonucleotides of the invention may bear a 2′-O-methoxyethyl (—O—CH2CH2OCH3) modification. Oligonucleotides comprising a plurality of nucleotide subunits having a 2′-O-methoxyethyl modification can have such a modification on any of the nucleotide subunits within the oligonucleotide, and may be chimeric oligonucleotides. Aside from or in addition to 2′-O-methoxyethyl modifications, oligonucleotides containing other modifications which enhance antisense efficacy, potency or target affinity are also preferred. Chimeric oligonucleotides comprising one or more such modifications are presently preferred.
  • 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 Applied Biosystems. Any other means for such synthesis may also be employed; the actual synthesis of the oligonucleotides is well within the talents of the routineer. It is well known to use similar techniques to prepare oligonucleotides such as the phosphorothioates and 2′-alkoxy or 2′-alkoxyalkoxy derivatives, including 2′-O-methoxyethyl oligonucleotides (Martin, P., [0044] Helv. Chim. Acta 1995, 78, 486-504). It is also well known to use similar techniques and commercially available modified amidites and controlled-pore glass (CPG) products such as biotin, fluorescein, acridine or psoralen-modified amidites and/or CPG (available from Glen Research, Sterling, Va.) to synthesize fluorescently labeled, biotinylated or other conjugated oligonucleotides.
  • The antisense compounds of the present invention include bioequivalent compounds, including pharmaceutically acceptable salts and prodrugs. This is intended to 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 pharmaceutically acceptable salts of the nucleic acids of the invention and prodrugs of such nucleic acids. Pharmaceutically acceptable salts are physiologically and pharmaceutically acceptable salts of the nucleic acids of the invention: i.e., salts that retain the desired biological activity of the parent compound and do not impart undesired toxicological effects thereto (see, for example, Berge et al., “Pharmaceutical Salts,” [0045] J. of Pharma Sci. 1977, 66, 1-19).
  • For oligonucleotides, 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. [0046]
  • The oligonucleotides of the invention may additionally or alternatively be prepared to be delivered in a Aprodrug form. The term Aprodrug 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 Dec. 9, 1993. [0047]
  • For therapeutic or prophylactic treatment, oligonucleotides are administered in accordance with this invention. Oligonucleotide compounds of the invention may be formulated in a pharmaceutical composition, which may include pharmaceutically acceptable carriers, thickeners, diluents, buffers, preservatives, surface active agents, neutral or cationic lipids, lipid complexes, liposomes, penetration enhancers, carrier compounds and other pharmaceutically acceptable carriers or excipients and the like in addition to the oligonucleotide. Such compositions and formulations are comprehended by the present invention. [0048]
  • Pharmaceutical compositions comprising the oligonucleotides of the present invention may 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., [0049] Critical Reviews in Therapeutic Drug Carrier Systems 1991, 8, 91-192; Muranishi, Critical Reviews in Therapeutic Drug Carrier Systems 1990, 7, 1-33). One or more penetration enhancers from one or more of these broad categories may be included.
  • 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, arachidonic 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., [0050] Critical Reviews in Therapeutic Drug Carrier Systems 1991, page 92; Muranishi, Critical Reviews in Therapeutic Drug Carrier Systems 1990, 7, 1; El-Hariri et al., J. Pharm. Pharmacol. 1992 44, 651-654).
  • The physiological roles of bile include the facilitation of dispersion and absorption of lipids and fat-soluble vitamins (Brunton, Chapter 38 In: [0051] Goodman & Gilman's The Pharmacological Basis of Therapeutics, 9th Ed., Hardman et al., eds., McGraw-Hill, New York, N.Y., 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.
  • Complex formulations comprising one or more penetration enhancers may be used. For example, bile salts may be used in combination with fatty acids to make complex formulations. [0052]
  • Chelating agents include, but are not limited to, disodium ethylenediaminetetraacetate (EDTA), citric acid, salicylates (e.g., sodium salicylate, 5-methoxysalicylate and homovanilate), N-acyl derivatives of collagen, laureth-9 and N-amino acyl derivatives of beta-diketones (enamines) [Lee et al., [0053] Critical Reviews in Therapeutic Drug Carrier Systems 1991, page 92; Muranishi, Critical Reviews in Therapeutic Drug Carrier Systems 1990, 7, 1-33; Buur et al., J. Control Rel. 1990, 14, 43-51). Chelating agents have the added advantage of also serving as DNase inhibitors.
  • Surfactants include, for example, sodium lauryl sulfate, polyoxyethylene-9-lauryl ether and polyoxyethylene-20-cetyl ether (Lee et al., [0054] Critical Reviews in Therapeutic Drug Carrier Systems 1991, page 92); and perfluorochemical emulsions, such as FC-43 (Takahashi et al., J. Pharm. Phamacol. 1988, 40, 252-257).
  • Non-surfactants include, for example, unsaturated cyclic ureas, 1-alkyl- and 1-alkenylazacyclo-alkanone derivatives (Lee et al., [0055] Critical 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, 621-626).
  • 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. [0056]
  • 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., pregelatinized 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. Pat. Nos. 4,704,295; 4,556,552; 4,309,406; and 4,309,404. [0057]
  • 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. [0058]
  • 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, liposomes and lipid:oligonucleotide complexes of uncharacterized structure. A preferred colloidal dispersion system is a plurality of liposomes. Liposomes are microscopic spheres having an aqueous core surrounded by one or more outer layers made up of lipids arranged in a bilayer configuration (see, generally, Chonn et al., [0059] Current Op. Biotech. 1995, 6, 698-708).
  • The pharmaceutical compositions of the present invention may be administered in a number of ways depending upon whether local or systemic treatment is desired and upon the area to be treated. Administration may be topical (including ophthalmic, vaginal, rectal, intranasal, epidermal, and transdermal), oral or parenteral. Parenteral administration includes intravenous drip, subcutaneous, intraperitoneal or intramuscular injection, pulmonary administration, e.g., by inhalation or insufflation, or intracranial, e.g., intrathecal or intraventricular, administration. Oligonucleotides with at least one 2′-O-methoxyethyl modification are believed to be particularly useful for oral administration. [0060]
  • Formulations for topical administration may include transdermal patches, ointments, lotions, creams, gels, drops, suppositories, sprays, liquids and powders. Conventional pharmaceutical carriers, aqueous, powder or oily bases, thickeners and the like may be necessary or desirable. Coated condoms, gloves and the like may also be useful. [0061]
  • Compositions for oral administration include powders or granules, suspensions or solutions in water or non-aqueous media, capsules, sachets or tablets. Thickeners, flavoring agents, diluents, emulsifiers, dispersing aids or binders may be desirable. [0062]
  • Compositions for parenteral administration may include sterile aqueous solutions which may also contain buffers, diluents and other suitable additives. 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, taxol, vincristine, vinblastine, etoposide, trimetrexate, teniposide, cisplatin and diethylstilbestrol (DES). See, generally, [0063] The Merck Manual of Diagnosis and Therapy, 15th Ed. 1987, pp. 1206-1228; Berkow et al., eds., Rahway, N.J. 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).
  • The formulation of therapeutic compositions and their subsequent administration is believed to be within the skill of those in the art. 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[0064] 50s found to be effective in vitro and in 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. Persons of ordinary skill in the art can easily estimate repetition rates for dosing based on measured residence times and concentrations of the drug in bodily fluids or tissues. Following successful treatment, it may be desirable to have the patient undergo maintenance therapy to prevent the recurrence of the disease state, wherein the oligonucleotide is administered in maintenance doses, ranging from 0.01 μg to 100 g per kg of body weight, once or more daily, to once every 20 years.
  • The following examples illustrate the present invention and are not intended to limit the same. [0065]
  • EXAMPLES Example 1 Synthesis of Oligonucleotides
  • Unmodified oligodeoxynucleotides are synthesized on an automated DNA synthesizer (Applied Biosystems model 380B) using standard phosphoramidite chemistry with oxidation by iodine. β-cyanoethyldiisopropyl-phosphoramidites are purchased from Applied Biosystems (Foster City, Calif.). For phosphorothioate oligonucleotides, the standard oxidation bottle was replaced by a 0.2 M solution of [0066] 3H-1,2-benzodithiole-3-one 1,1-dioxide in acetonitrile for the stepwise thiation of the phosphite linkages. The thiation cycle wait step was increased to 68 seconds and was followed by the capping step. Cytosines may be 5-methyl cytosines. (5-methyl deoxycytidine phosphoramidites available from Glen Research, Sterling, Va. or Amersham Pharmacia Biotech, Piscataway, N.J.)
  • 2′-methoxy oligonucleotides are synthesized using 2′-methoxy β-cyanoethyldiisopropyl-phosphoramidites (Chemgenes, Needham, Mass.) and the standard cycle for unmodified oligonucleotides, except the wait step after pulse delivery of tetrazole and base is increased to 360 seconds. Other 2′-alkoxy oligonucleotides are synthesized by a modification of this method, using appropriate 2′-modified amidites such as those available from Glen Research, Inc., Sterling, Va. [0067]
  • 2′-fluoro oligonucleotides are synthesized as described in Kawasaki et al. ([0068] J. Med. Chem. 1993, 36, 831-841). Briefly, the protected nucleoside N6-benzoyl-2′-deoxy-2′-fluoroadenosine is synthesized utilizing commercially available 9-β-D-arabinofuranosyladenine as starting material and by modifying literature procedures whereby the 2′-a-fluoro atom is introduced by a SN2-displacement of a 2′-β-O-trifyl group. Thus N6-benzoyl-9-β-D-arabinofuranosyladenine is selectively protected in moderate yield as the 3′,5′-ditetrahydropyranyl (THP) intermediate. Deprotection of the THP and N6-benzoyl groups is accomplished using standard methodologies and standard methods are used to obtain the 5′-dimethoxytrityl-(DMT) and 5′-DMT-3′-phosphoramidite intermediates.
  • The synthesis of 2′-deoxy-2′-fluoroguanosine is accomplished using tetraisopropyldisiloxanyl (TPDS) protected 9-β-D-arabinofuranosylguanine as starting material, and conversion to the intermediate diisobutyryl-arabinofuranosylguanosine. Deprotection of the TPDS group is followed by protection of the hydroxyl group with THP to give diisobutyryl di-THP protected arabinofuranosylguanine. Selective O-deacylation and triflation is followed by treatment of the crude product with fluoride, then deprotection of the THP groups. Standard methodologies are used to obtain the 5′-DMT- and 5′-DMT-3′-phosphoramidites. [0069]
  • Synthesis of 2′-deoxy-2′-fluorouridine is accomplished by the modification of a known procedure in which 2,2′-anhydro-1-β-D-arabinofuranosyluracil is treated with 70% hydrogen fluoride-pyridine. Standard procedures are used to obtain the 5′-DMT and 5′-DMT-3′ phosphoramidites. 2′-deoxy-2′-fluorocytidine is synthesized via amination of 2′-deoxy-2′-fluorouridine, followed by selective protection to give N[0070] 4-benzoyl-2′-deoxy-2′-fluorocytidine. Standard procedures are used to obtain the 5′-DMT and 5′-DMT-3′phosphoramidites.
  • 2′-(2-methoxyethyl)-modified amidites were synthesized according to Martin, P. ([0071] Helv. Chim. Acta 1995, 78, 486-506). For ease of synthesis, the last nucleotide may be a deoxynucleotide. 2′-O—CH2CH2OCH3-cytosines may be 5-methyl cytosines.
  • Synthesis of 5-Methyl Cytosine Monomers
  • 2,2′-Anhydro [1-(β-D-arabinofuranosyl)-5-methyluridine]: [0072]
  • 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. [0073]
  • 2′-O-Methoxyethyl-5-methyluridine: [0074]
  • 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 CH[0075] 3CN (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 CH2Cl2 (250 mL) and adsorbed onto silica (150 g) prior to loading onto the column. The product was eluted with the packing solvent to give 160 g (63%) of product.
  • 2′-O-Methoxyethyl-5′-O-dimethoxytrityl-5-methyluridine: [0076]
  • 2′-O-Methoxyethyl-5-methyluridine (160 g, 0.506 M). was co-evaporated with pyridine (250 mL) and the dried residue dissolved in pyridine (1.3 L). A first aliquot of dimethoxytrityl chloride (94.3 g, 0.278 M) was added and the mixture stirred at room temperature for one hour. A second aliquot of dimethoxytrityl chloride (94.3 g, 0.278 M) was added and the reaction stirred for an additional one hour. Methanol (170 mL) was then added to stop the reaction. HPLC showed the presence of approximately 70% product. The solvent was evaporated and triturated with CH[0077] 3CN (200 mL). The residue was dissolved in CHCl3 (1.5 L) and extracted with 2×500 mL of saturated NaHCO3 and 2×500 mL of saturated NaCl. The organic phase was dried over Na2SO4, filtered and evaporated. 275 g of residue was obtained. The residue was purified on a 3.5 kg silica gel column, packed and eluted with EtOAc/Hexane/Acetone (5:5:1) containing 0.5% Et3NH. The pure fractions were evaporated to give 164 g of product. Approximately 20 g additional was obtained from the impure fractions to give a total yield of 183 g (57%).
  • 3′-O-Acetyl-2′-O-methoxyethyl-5′-O-dimethoxytrityl-5-methyluridine: [0078]
  • 2′-O-Methoxyethyl-5′-O-dimethoxytrityl-5-methyl-uridine (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 tlc by first quenching the tlc sample with the addition of MeOH. Upon completion of the reaction, as judged by tlc, MeOH (50 mL) was added and the mixture evaporated at 35° C. The residue was dissolved in CHCl[0079] 3 (800 mL) and extracted with 2×200 mL of saturated sodium bicarbonate and 2×200 mL of saturated NaCl. The water layers were back extracted with 200 mL of CHCl3. The combined organics were dried with sodium sulfate and evaporated to give 122 g of residue (approx. 90% product). The residue was purified on a 3.5 kg silica gel column and eluted using EtOAc/Hexane(4:1). Pure product fractions were evaporated to yield 96 g (84%).
  • 3′-O-Acetyl-2′-O-methoxyethyl-5′-O-dimethoxytrityl-5-methyl-4-triazoleuridine: [0080]
  • A first solution was prepared by dissolving 3′-O-acetyl-2′-O-methoxyethyl-5′-O-dimethoxytrityl-5-methyluridine (96 g, 0.144 M) in CH[0081] 3CN (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. POCl3 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 1×300 mL of NaHCO3 and 2×300 mL of saturated NaCl, dried over sodium sulfate and evaporated. The residue was triturated with EtOAc to give the title compound.
  • 2′-O-Methoxyethyl-5′-O-dimethoxytrityl-5-methylcytidine: [0082]
  • A solution of 3′-O-acetyl-2′-O-methoxyethyl-5′-O-dimethoxytrityl-5-methyl-4-triazoleuridine (103 g, 0.141 M) in dioxane (500 mL) and NH[0083] 4OH (30 mL) was stirred at room temperature for 2 hours. The dioxane solution was evaporated and the residue azeotroped with MeOH (2×200 mL). The residue was dissolved in MeOH (300 mL) and transferred to a 2 liter stainless steel pressure vessel. MeOH (400 mL) saturated with NH3 gas was added and the vessel heated to 100° C. for 2 hours (tlc 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.
  • N[0084] 4-Benzoyl-2′-O-methoxyethyl-5′-O-dimethoxytrityl-5-methyl-cytidine:
  • 2′-O-Methoxyethyl-5′-O-dimethoxytrityl-5-methyl-cytidine (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, tlc 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 CHCl[0085] 3 (700 mL) and extracted with saturated NaHCO3 (2×300 mL) and saturated NaCl (2×300 mL), dried over MgSO4 and evaporated to give a residue (96 g). The residue was chromatographed on a 1.5 kg silica column using EtOAc/Hexane (1:1) containing 0.5% Et3NH as the eluting solvent. The pure product fractions were evaporated to give 90 g (90%) of the title compound.
  • N[0086] 4-Benzoyl-2′-O-methoxyethyl-5′-O-dimethoxytrityl-5-methylcytidine-3′-amidite:
  • N[0087] 4-Benzoyl-2′-O-methoxyethyl-5′-O-dimethoxytrityl-5-methylcytidine (74 g, 0.10 M) was dissolved in CH2Cl2 (1 L). Tetrazole diisopropylamine (7.1 g) and 2-cyanoethoxytetra(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 (tlc showed the reaction to be 95% complete). The reaction mixture was extracted with saturated NaHCO3 (1×300 mL) and saturated NaCl (3×300 mL). The aqueous washes were back-extracted with CH2Cl2 (300 mL), and the extracts were combined, dried over MgSO4 and concentrated. The residue obtained was chromatographed on a 1.5 kg silica column using EtOAcHexane (3:1) as the eluting solvent. The pure fractions were combined to give 90.6 g (87%) of the title compound.
  • 5-methyl-2′-deoxycytidine (5-me-C) containing oligonucleotides were synthesized according to published methods (Sanghvi et al., [0088] Nucl. Acids Res. 1993, 21, 3197-3203) using commercially available phosphoramidites (Glen Research, Sterling Va. or ChemGenes, Needham Mass.).
  • 2=-O-(dimethylaminooxyethyl) nucleoside amidites
  • 2′-(Dimethylaminooxyethoxy) nucleoside amidites [also known in the art as 2′-O-(dimethylaminooxyethyl) nucleoside amidites] are prepared as described in the following paragraphs. Adenosine, cytidine and guanosine nucleoside amidites are prepared similarly to the thymidine (5-methyluridine) except the exocyclic amines are protected with a benzoyl moiety in the case of adenosine and cytidine and with isobutyryl in the case of guanosine. [0089]
  • 5′-O-tert-Butyldiphenylsilyl-O[0090] 2-2′-anhydro-5-methyluridine
  • O[0091] 2-2′-anhydro-5-methyluridine (Pro. Bio. Sint., Varese, Italy, 100.0 g, 0.416 mmol), dimethylaminopyridine (0.66 g, 0.013 eq, 0.0054 mmol) were dissolved in dry pyridine (500 ml) at ambient temperature under an argon atmosphere and with mechanical stirring tert-Butyldiphenylchlorosilane (125.8 g, 119.0 mL, 1.1 eq, 0.458 mmol) was added in one portion. The reaction was stirred for 16 h at ambient temperature. TLC (Rf 0.22, ethyl acetate) indicated a complete reaction. The solution was concentrated under reduced pressure to a thick oil. This was partitioned between dichloromethane (1 L) and saturated sodium bicarbonate (2×1 L) and brine (1 L). The organic layer was dried over sodium sulfate and concentrated under reduced pressure to a thick oil. The oil was dissolved in a 1:1 mixture of ethyl acetate and ethyl ether (600 mL) and the solution was cooled to −10° C. The resulting crystalline product was collected by filtration, washed with ethyl ether (3×200 mL) and dried (40° C., 1 mm Hg, 24 h) to 149 g (74.8%) of white solid. TLC and NMR were consistent with pure product.
  • 5′-O-tert-Butyldiphenylsilyl-2′-O-(2-hydroxyethyl)-5-methyluridine [0092]
  • In a 2 L stainless steel, unstirred pressure reactor was added borane in tetrahydrofuran (1.0 M, 2.0 eq, 622 mL). In the fume hood and with manual stirring, ethylene glycol (350 mL, excess) was added cautiously at first until the evolution of hydrogen gas subsided. 5′-O-tert-Butyldiphenylsilyl-O[0093] 2-2′-anhydro-5-methyluridine (149 g, 0.311 mol) and sodium bicarbonate (0.074 g, 0.003 eq) were added with manual stirring. The reactor was sealed and heated in an oil bath until an internal temperature of 160° C. was reached and then maintained for 16 h (pressure<100 psig). The reaction vessel was cooled to ambient and opened. TLC (Rf 0.67 for desired product and Rf 0.82 for ara-T side product, ethyl acetate) indicated about 70% conversion to the product. In order to avoid additional side product formation, the reaction was stopped, concentrated under reduced pressure (10 to 1 mm Hg) in a warm water bath (40-100° C.) with the more extreme conditions used to remove the ethylene glycol. [Alternatively, once the low boiling solvent is gone, the remaining solution can be partitioned between ethyl acetate and water. The product will be in the organic phase.] The residue was purified by column chromatography (2 kg silica gel, ethyl acetate-hexanes gradient 1:1 to 4:1). The appropriate fractions were combined, stripped and dried to product as a white crisp foam (84 g, 50%), contaminated starting material (17.4 g) and pure reusable starting material 20 g. The yield based on starting material less pure recovered starting material was 58%. TLC and NMR were consistent with 99% pure product.
  • 2′-O-([2-phthalimidoxy)ethyl]-5′-t-butyldiphenylsilyl-5-methyluridine [0094]
  • 5′-O-tert-Butyldiphenylsilyl-2′-O-(2-hydroxyethyl)-5-methyluridine (20 g, 36.98 mmol) was mixed with triphenylphosphine (11.63 g, 44.36 mmol) and N-hydroxyphthalimide (7.24 g, 44.36 mmol). It was then dried over P[0095] 2O5 under high vacuum for two days at 40° C. The reaction mixture was flushed with argon and dry THF (369.8 mL, Aldrich, sure seal bottle) was added to get a clear solution. Diethyl-azodicarboxylate (6.98 mL, 44.36 mmol) was added dropwise to the reaction mixture. The rate of addition is maintained such that resulting deep red coloration is just discharged before adding the next drop. After the addition was complete, the reaction was stirred for 4 hrs. By that time TLC showed the completion of the reaction (ethylacetate:hexane, 60:40). The solvent was evaporated in vacuum. Residue obtained was placed on a flash column and eluted with ethyl acetate:hexane (60:40), to get 2′-O-([2-phthalimidoxy)ethyl]-5′-t-butyldiphenylsilyl-5-methyluridine as white foam (21.819, 86%).
  • 5′-O-tert-butyldiphenylsilyl-2′-O-[(2-formadoximinooxy)ethyl]-5-methyluridine [0096]
  • 2′-O-([2-phthalimidoxy)ethyl]-5′-t-butyldiphenylsilyl-5-methyluridine (3.1 g, 4.5 mmol) was dissolved in dry CH[0097] 2Cl2 (4.5 mL) and methylhydrazine (300 mL, 4.64 mmol) was added dropwise at −10° C. to 0° C. After 1 hr the mixture was filtered, the filtrate was washed with ice cold CH2Cl2 and the combined organic phase was washed with water, brine and dried over anhydrous Na2SO4. The solution was concentrated to get 2′-O-(aminooxyethyl) thymidine, which was then dissolved in MeOH (67.5 mL). To this formaldehyde (20% aqueous solution, w/w, 1.1 eg.) was added and the mixture for 1 hr. Solvent was removed under vacuum; residue chromatographed to get 5′-O-tert-butyldiphenylsilyl-2′-O-[(2-formadoximinooxy) ethyl]-5-methyluridine as white foam (1.95, 78%).
  • 5′-O-tert-Butyldiphenylsilyl-2 ′-O-[N,N-dimethylaminooxyethyl]-5-methyluridine [0098]
  • 5′-O-tert-butyldiphenylsilyl-2′-O-[(2-formadoximinooxy)ethyl]-5-methyluridine (1.77 g, 3.12 mmol) was dissolved in a solution of 1M pyridinium p-toluenesulfonate (PPTS) in dry MeOH (30.6 mL). Sodium cyanoborohydride (0.39 g, 6.13 mmol) was added to this solution at 10° C. under inert atmosphere. The reaction mixture was stirred for 10 minutes at 10° C. After that the reaction vessel was removed from the ice bath and stirred at room temperature for 2 hr, the reaction monitored-by TLC (5% MeOH in CH[0099] 2Cl2). Aqueous NaHCO3 solution (5%, 10 mL) was added and extracted with ethyl acetate (2×20 mL). Ethyl acetate phase was dried over anhydrous Na2SO4, evaporated to dryness. Residue was dissolved in a solution of 1M PPTS in MeOH (30.6 mL). Formaldehyde (20% w/w, 30 mL, 3.37 mmol) was added and the reaction mixture was stirred at room temperature for 10 minutes. Reaction mixture cooled to 10° C. in an ice bath, sodium cyanoborohydride (0.39 g, 6.13 mmol) was added and reaction mixture stirred at 10° C. for 10 minutes. After 10 minutes, the reaction mixture was removed from the ice bath and stirred at room temperature for 2 hrs. To the reaction mixture 5% NaHCO3 (25 mL) solution was added and extracted with ethyl acetate (2×25 mL). Ethyl acetate layer was dried over anhydrous Na2SO4 and evaporated to dryness . The residue obtained was purified by flash column chromatography and eluted with 5% MeOH in CH2Cl2 to get 5′-O-tert-butyldiphenylsilyl-2′-O-[N,N-dimethylaminooxyethyl]-5-methyluridine as a white foam (14.6 g, 80%).
  • 2′-O-(dimethylaminooxyethyl)-5-methyluridine [0100]
  • Triethylamine trihydrofluoride (3.91 mL, 24.0 mmol) was dissolved in dry THF and triethylamine (1.67 mL, 12 mmol, dry, kept over KOH). This mixture of triethylamine-2HF was then added to 5′-O-tert-butyldiphenylsilyl-2′-O-[N,N-dimethylaminooxyethyl]-5-methyluridine (1.40 g, 2.4 mmol) and stirred at room temperature for 24 hrs. Reaction was monitored by TLC (5% MeOH in CH[0101] 2Cl2). Solvent was removed under vacuum and the residue placed on a flash column and eluted with 10% MeOH in CH2Cl2 to get 2′-O-(dimethylaminooxyethyl)-5-methyluridine (766 mg, 92.5%).
  • 5′-O-DMT-2′-O-(dimethylaminooxyethyl)-5-methyluridine [0102]
  • 2′-O-(dimethylaminooxyethyl)-5-methyluridine (750 mg, 2.17 mmol) was dried over P[0103] 2O5 under high vacuum overnight at 40° C. It was then co-evaporated with anhydrous pyridine (20 mL). The residue obtained was dissolved in pyridine (11 mL) under argon atmosphere. 4-dimethylaminopyridine (26.5 mg, 2.60 mmol), 4,4′-dimethoxytrityl chloride (880 mg, 2.60 mmol) was added to the mixture and the reaction mixture was stirred at room temperature until all of the starting material disappeared. Pyridine was removed under vacuum and the residue chromatographed and eluted with 10% MeOH in CH2Cl2 (containing a few drops of pyridine) to get 5′-O-DMT-2′-O-(dimethylamino-oxyethyl)-5-methyluridine (1.13 g, 80%).
  • 5′-O-DMT-2′-O-(2-N,N-dimethylaminooxyethyl)-5-methyluridine-3′-[(2-cyanoethyl)-N,N-diisopropylphosphoramidite][0104]
  • 5′-O-DMT-2′-O-(dimethylaminooxyethyl)-5-methyluridine (1.08 g, 1.67 mmol) was co-evaporated with toluene (20 mL). To the residue N,N-diisopropylamine tetrazonide (0.29 g, 1.67 mmol) was added and dried over P[0105] 2O5 under high vacuum overnight at 40° C. Then the reaction mixture was dissolved in anhydrous acetonitrile (8.4 mL) and 2-cyanoethyl-N,N,N1,N1-tetraisopropylphosphoramidite (2.12 mL, 6.08 mmol) was added. The reaction mixture was stirred at ambient temperature for 4 hrs under inert atmosphere. The progress of the reaction was monitored by TLC (hexane:ethyl acetate 1:1). The solvent was evaporated, then the residue was dissolved in ethyl acetate (70 mL) and washed with 5% aqueous NaHCO3 (40 mL). Ethyl acetate layer was dried over anhydrous Na2SO4 and concentrated. Residue obtained was chromatographed (ethyl acetate as eluent) to get 5′-O-DMT-2′-O-(2-N,N-dimethylaminooxyethyl)-5-methyluridine-3′-[(2-cyanoethyl)-N,N-diisopropylphosphoramidite] as a foam (1.04 g, 74.9%).
  • Oligonucleotides having methylene(methylimino) (MMI) backbones are synthesized according to U.S. Pat. No. 5,378,825, which is coassigned to the assignee of the present invention and is incorporated herein in its entirety. For ease of synthesis, various nucleoside dimers containing MMI linkages are synthesized and incorporated into oligonucleotides. Other nitrogen-containing backbones are synthesized according to WO 92/20823 which is also coassigned to the assignee of the present invention and incorporated herein in its entirety. [0106]
  • Oligonucleotides having amide backbones are synthesized according to De Mesmaeker et al. ([0107] Acc. Chem. Res. 1995, 28, 366-374). The amide moiety is readily accessible by simple and well-known synthetic methods and is compatible with the conditions required for solid phase synthesis of oligonucleotides.
  • Oligonucleotides with morpholino backbones are synthesized according to U.S. Pat. No. 5,034,506 (Summerton and Weller). [0108]
  • Peptide-nucleic acid (PNA) oligomers are synthesized according to P. E. Nielsen et al. ([0109] Science 1991, 254, 1497-1500).
  • After cleavage from the controlled pore glass column (Applied Biosystems) and deblocking in concentrated ammonium hydroxide at 55° C. for 18 hours, the oligonucleotides are purified by precipitation twice out of 0.5 M NaCl with 2.5 volumes ethanol. Synthesized oligonucleotides were analyzed by polyacrylamide gel electrophoresis on denaturing gels or capillary gel electrophoresis and judged to be at least 85% full length material. The relative amounts of phosphorothioate and phosphodiester linkages obtained in synthesis were periodically checked by [0110] 31P nuclear magnetic resonance spectroscopy, and for some studies oligonucleotides were purified by HPLC, as described by Chiang et al. (J. Biol. Chem. 1991, 266, 18162). Results obtained with HPLC-purified material were similar to those obtained with non-HPLC purified material.
  • Alternatively, oligonucleotides are synthesized in 96 well plate format via solid phase P(III) phosphoramidite chemistry on an automated synthesizer capable of assembling 96 sequences simultaneously in a standard 96 well format. Phosphodiester internucleotide linkages are afforded by oxidation with aqueous iodine. Phosphorothioate internucleotide linkages are generated by sulfurization utilizing 3,H-1,2 benzodithilole-3-one 1,1 dioxide (Beaucage Reagent) in anhydrous acetonitrile. Standard base-protected beta-cyanoethyl-di-isopropyl phosphoramidites are purchased from commercial vendors (e.g. PE-Applied Biosystems, Foster City, Calif., or Pharmacia, Piscataway, N.J.). Non-standard nucleosides are synthesized as per published methods. They are utilized as base protected beta-cyanoethyldiisopropyl phosphoramidites. [0111]
  • Oligonucleotides were cleaved from support and deprotected with concentrated NH[0112] 4OH at elevated temperature (55-60° C.) for 12-16 hours and the released product then dried in vacuo. The dried product was then re-suspended in sterile water to afford a master plate from which all analytical and test plate samples are then diluted utilizing robotic pipettors.
  • Example 2 Human FAK Oligonucleotide Sequences
  • Antisense oligonucleotides were designed to target human FAK. Target sequence data are from the focal adhesion kinase (FAK) cDNA sequence published by Whitney, G.S., et al. ([0113] DNA Cell Biol., 1993, 12, 823-830); Genbank accession number L13616, provided herein as SEQ ID NO: 1. One set of oligonucleotides were synthesized as chimeric oligonucleotides (“gapmers”), 20 nucleotides in length, composed of a central “gap” region consisting of ten 2′-deoxynucleotides, which is flanked on both sides (5′ and 3′ directions) by five-nucleotide “wings.” The wings are composed of 2′-methoxyethyl (2′-MOE) nucleotides. The internucleoside (backbone) linkages are phosphorothioate (P═S) throughout the oligonucleotide. All 2′-MOE cytosines were 5-methyl-cytosines. These oligonucleotide sequences are shown in Table 1. An identical set of sequences were prepared as fully phosphorothioated oligodeoxynucleotides. These are shown in Table 2. An additional set of oligonucleotides were synthesized as chimeric oligonucleotides (“gapmers”), 15 nucleotides in length, composed of a central “gap” region consisting of five 2′-deoxynucleotides, which is flanked on both sides (5′ and 3′ directions) by five-nucleotide “wings.” The wings are composed of 2′-methoxyethyl (2′-MOE) nucleotides. The internucleoside (backbone) linkages are phosphorothioate (P═S) throughout the oligonucleotide. All 2′-MOE cytosines were 5-methyl-cytosines. These oligonucleotide sequences are shown in Table 3. An identical set of sequences were prepared as fully phosphorothioated oligodeoxynucleotides. These are shown in Table 4.
  • Human A549 lung carcinoma cells (American Type Culture Collection, Manassas, Va.) were grown in DMEM supplemented with 10% fetal bovine serum (FBS), non-essential amino acids for MEM, sodium pyruvate (1 mM), penicillin (50 U/ml) and streptomycin (50 μg/ml). All cell culture reagents were obtained from Life Technologies (Rockville, Md.). [0114]
  • The cells were washed once with OPTIMEM™ (Life Technologies, Rockville, Md.), then transfected with 400 nM oligonucleotide and 12 mg/ml LIPOFECTIN® (Life Technologies, Rockville, Md.), a 1:1 (w/w) liposome formulation of the cationic lipid N-[1-(2,3-dioleyloxy)propyl]-n,n,n-trimethylammonium chloride (DOTMA), and dioleoyl phosphotidylethanolamine (DOPE) in membrane filtered water. The cells were incubated with oligonucleotide for four hours, after which the media was replaced with fresh media and the cells incubated for another 20 hours. [0115]
  • Total cellular RNA was isolated using an ATLAS™ Pure RNA isolation kit (Clontech, Palo Alto, Calif.). RNA was then separated on a 1.2% agarose-formaldehyde gel, transferred to Hybond-N+membrane (Amersham Pharmacia Biotech, Arlington Heights, Ill.), a positively charged nylon membrane. Immobilized RNA was cross-linked by exposure to UV light. Membranes were probed with either FAK or glyceraldehyde 3-phosphate dehydrogenase (G3PDH) probes. The probes were labeled by random primer using the PRIME-A-GENE[0116] 7 Labeling System, Promega, Madison, Wis.) and hybridized to the membranes. mRNA signals were quantitated by a PhosphoImager (Molecular Dynamics, Sunnyvale, Calif.).
  • Results of an initial screen of the FAK antisense oligonucleotides are shown in Tables 5 (20 mers) and 6 (15 mers). Oligonucleotides 15392 (SEQ ID NO. 3), 15394 (SEQ ID NO. 4), 15397 (SEQ ID NO. 6), 15399 (SEQ ID NO. 7), 15401 (SEQ ID NO. 8), 15403.(SEQ ID NO. 9), 15405 (SEQ ID NO. 10), 15407 (SEQ ID NO. 11), 15409 (SEQ ID NO. 12), 15413 (SEQ ID NO. 14), 15415 (SEQ ID NO. 15), 15458 (SEQ ID NO. 16), 15460 (SEQ ID NO. 17), 15421 (SEQ ID NO. 18), 15425 (SEQ ID NO. 20), 15393 (SEQ ID NO. 23), 15406 (SEQ ID NO. 30), 15408 (SEQ ID NO. 31) and 15412 (SEQ ID NO. 33) resulted in about 50% or greater inhibition of FAK mRNA expression in this assay. Oligonucleotides 15401 (SEQ ID NO. 8), 15403 (SEQ ID NO. 9), 15409 (SEQ ID NO. 12), 15413 (SEQ ID NO. 14), 15415 (SEQ ID NO. 15), and 15421 (SEQ ID NO. 18) resulted in about 80% or greater inhibition of FAK mRNA expression. [0117]
    TABLE 1
    Nucleotide Sequences of Human FAK Chimeric
    (deoxy gapped) 20 mer Phosphorothioate
    Oligonucleotides
    SEQ TARGET GENE GENE
    ISIS NUCLEOTIDE SEQUENCE1 ID NUCLEOTIDE TARGET
    NO. (5′−>3′) NO: CO-ORDINATES2 REGION
    15392 CCGCGGGCTCACAGTGGTCG 3 0001-0020 5′-UTR
    15394 GGCGCCGTGAAGCGAAGGCA 4 0078-0097 5′-UTR
    15395 CAGTTCTGCTCGGACCGCGG 5 0101-0120 5′-UTR
    15397 GAAACTGCAGAAGGCACTGA 6 0150-0169 5′-UTR
    15399 TTCTCCCTTCCGTTATTCTT 7 0183-0202 5′-UTR
    15401 CTAGATGCTAGGTATCTGTC 8 0206-0225 5′-UTR
    15403 TTTTGCTAGATGCTAGGTAT 9 0211-0230 5′-UTR
    15405 GGTAAGCAGCTGCCATTATT 10 0229-0248 start
    15407 AGTACCCAGGTGAGTCTTAG 11 0285-0304 coding
    15409 CCTGACATCAGTAGCATCTC 12 0408-0427 coding
    15411 GTTGGCTTATCTTCAGTAAA 13 0641-0660 coding
    15413 GGTTAGGGATGGTGCCGTCA 14 1218-1237 coding
    15415 TGTTGGTTTCCAATCGGACC 15 2789-2808 coding
    15417 CTAGGGGAGGCTCAGTGTGG 16 3383-3402 stop
    15419 ATTCCTCGCTGCTGGTGGAA 17 3444-3463 3′-UTR
    15421 TTTCAACCAGATGGTCATTC 18 3510-3529 3′-UTR
    15423 TTCTGAATATCATGATTGAA 19 3590-3609 3′-UTR
    15425 CATGATGCTTAAAAGCTTAC 20 3658-3677 3′-UTR
    15427 AATGTGAACATAAATTGTTC 21 3680-3699 3′-UTR
    15429 AAGGTAGTTTAGGAATTAAG 22 3738-3757 3′-UTR
  • [0118]
    TABLE 2
    Nucleotide Sequences of Human FAK 20 mer
    Phosphorothioate Oligonucleotides
    SEQ TARGET GENE GENE
    ISIS NUCLEOTIDE SEQUENCE1 ID NUCLEOTIDE TARGET
    NO. (5′−>3′) NO: CO-ORDINATES2 REGION
    15432 CCGCGGGCTCACAGTGGTCG 3 0001-0020 5′-UTR
    15434 GGCGCCGTGAAGCGAAGGCA 4 0078-0097 5′-UTR
    15436 CAGTTCTGCTCGGACCGCGG 5 0101-0120 5′-UTR
    15438 GAAACTGCAGAAGGCACTGA 6 0150-0169 5′-UTR
    15440 TTCTCCCTTCCGTTATTCTT 7 0183-0202 5′-UTR
    15442 CTAGATGCTAGGTATCTGTC 8 0206-0225 5′-UTR
    15444 TTTTGCTAGATGCTAGGTAT 9 0211-0230 5′-UTR
    15446 GGTAAGCAGCTGCCATTATT 10 0229-0248 start
    15448 AGTACCCAGGTGAGTCTTAG 11 0285-0304 coding
    15450 CCTGACATCAGTAGCATCTC 12 0408-0427 coding
    15452 GTTGGCTTATCTTCAGTAAA 13 0641-0660 coding
    15454 GGTTAGGGATGGTGCCGTCA 14 1218-1237 coding
    15456 TGTTGGTTTCCAATCGGACC 15 2789-2808 coding
    15458 CTAGGGGAGGCTCAGTGTGG 16 3383-3402 stop
    15460 ATTCCTCGCTGCTGGTGGAA 17 3444-3463 3′-UTR
    15462 TTTCAACCAGATGGTCATTC 18 3510-3529 3′-UTR
    15464 TTCTGAATATCATGATTGAA 19 3590-3609 3′-UTR
    15466 CATGATGCTTAAAAGCTTAC 20 3658-3677 3′-UTR
    15468 AATGTGAACATAAATTGTTC 21 3680-3699 3′-UTR
    15470 AAGGTAGTTTAGGAATTAAG 22 3738-3757 3′-UTR
  • [0119]
    TABLE 3
    Nucleotide Sequences of Human FAK Chimeric
    (deoxy gapped) 15 mer Phosphorothioate
    Oligonucleotides
    SEQ TARGET GENE GENE
    ISIS NUCLEOTIDE SEQUENCE1 ID NUCLEOTIDE TARGET
    NO. (5′−>3′) NO: CO-ORDINATES2 REGION
    15393 GCGGGCTCACAGTGG 23 0004-0018 5′-UTR
    15431 CGCCGTGAAGCGAAG 24 0081-0095 5′-UTR
    15396 GTTCTGCTCGGACCG 25 0104-0118 5′-UTR
    15398 AACTGCAGAAGGCAC 26 0153-0167 5′-UTR
    15400 CTCCCTTCCGTTATT 27 0186-0200 5′-UTR
    15402 AGATGCTAGGTATCT 28 0209-0223 5′-UTR
    15404 TTGCTAGATGCTAGG 29 0214-0228 5′-UTR
    15406 TAAGCAGCTGCCATT 30 0232-0246 start
    15408 TACCCAGGTGAGTCT 31 0288-0302 coding
    15410 TGACATCAGTAGCAT 32 0411-0425 coding
    15412 TGGCTTATCTTCAGT 33 0644-0658 coding
    15414 TTAGGGATGGTGCCG 34 1221-1235 coding
    15416 TTGGTTTCCAATCGG 35 2792-2806 coding
    15418 AGGGGAGGCTCAGTG 36 3386-3400 stop
    15420 TCCTCGCTGCTGGTG 37 3447-3461 3′-UTR
    15422 TCAACCAGATGGTCA 38 3513-3527 3′-UTR
    15424 CTGAATATCATGATT 39 3593-3607 3′-UTR
    15426 TGATGCTTAAAAGCT 40 3661-3675 3′-UTR
    15428 TGTGAACATAAATTG 41 3683-3697 3′-UTR
    15430 GGTAGTTTAGGAATT 42 3741-3755 3′-UTR
  • [0120]
    TABLE 4
    Nucleotide Sequences of Human FAK 15 mer
    Phosphorothioate Oligonucleotides
    SEQ TARGET GENE GENE
    ISIS NUCLEOTIDE SEQUENCE1 ID NUCLEOTIDE TARGET
    NO. (5′−>3′) NO: CO-ORDINATES2 REGION
    15433 GCGGGCTCACAGTGG 23 0004-0018 5′-UTR
    15435 CGCCGTGAAGCGAAG 24 0081-0095 5′-UTR
    15437 GTTCTGCTCGGACCG 25 0104-0118 5′-UTR
    15439 AACTGCAGAAGGCAC 26 0153-0167 5′-UTR
    15441 CTCCCTTCCGTTATT 27 0186-0200 5′-UTR
    15443 AGATGCTAGGTATCT 28 0209-0223 5′-UTR
    15445 TTGCTAGATGCTAGG 29 0214-0228 5′-UTR
    15447 TAAGCAGCTGCCATT 30 0232-0246 start
    15449 TACCCAGGTGAGTCT 31 0288-0302 coding
    15451 TGACATCAGTAGCAT 32 0411-0425 coding
    15453 TGGCTTATCTTCAGT 33 0644-0658 coding
    15455 TTAGGGATGGTGCCG 34 1221-1235 coding
    15457 TTGGTTTCCAATCGG 35 2792-2806 coding
    15459 AGGGGAGGCTCAGTG 36 3386-3400 stop
    15461 TCCTCGCTGCTGGTG 37 3447-3461 3′-UTR
    15463 TCAACCAGATGGTCA 38 3513-3527 3′-UTR
    15465 CTGAATATCATGATT 39 3593-3607 3′-UTR
    15467 TGATGCTTAAAAGCT 40 3661-3675 3′-UTR
    15469 TGTGAACATAAATTG 41 3683-3697 3′-UTR
    15471 GGTAGTTTAGGAATT 42 3741-3755 3′-UTR
  • [0121]
    TABLE 5
    Inhibition of Human Fak mRNA expression in A549
    Cells by FAK 20 mer Antisense Oligonucleotides
    SEQ GENE
    ISIS ID TARGET % mRNA % mRNA
    No: NO: REGION EXPRESSION INHIBITION
    control 100%  0%
    15392  3 5′-UTR  29% 71%
    15432  3 5′-UTR 108%
    15394  4 5′-UTR  30% 70%
    15434  4 5′-UTR 147%
    15395  5 5′-UTR  57% 43%
    15436  5 5′-UTR  88% 12%
    15397  6 5′-UTR  31% 69%
    15438  6 5′-UTR  64% 36%
    15399  7 5′-UTR  48% 52%
    15440  7 5′-UTR  92%  8%
    15401  8 5′-UTR  17% 83%
    15442  8 5′-UTR  63% 37%
    15403  9 5′-UTR  17% 83%
    15444  9 5′-UTR 111%
    15405 10 start  46% 54%
    15446 10 start 145%
    15407 11 coding  36% 64%
    15448 11 coding  90% 10%
    15409 12 coding  13% 87%
    15450 12 coding 149%
    15411 13 coding  70% 30%
    15452 13 coding 129%
    15413 14 coding  22% 78%
    15454 14 coding  82% 18%
    15415 15 coding  20% 80%
    15456 15 coding  88% 12%
    15417 16 stop  56% 44%
    15458 16 stop  39% 61%
    15419 17 3′-UTR  55% 45%
    15460 17 3′-UTR  42% 58%
    15421 18 3′-UTR  20% 80%
    15462 18 3′-UTR  60% 40%
    15423 19 3′-UTR  55% 45%
    15464 19 3′-UTR  97%  3%
    15425 20 3′UTR  51% 49%
    15466 20 3′-UTR  74% 26%
    15427 21 3′-UTR  67% 33%
    15468 21 3′-UTR 131%
    15429 22 3′-UTR  57% 43%
    15470 22 3′-UTR  71% 29%
  • [0122]
    TABLE 6
    Inhibition of Human Fak mRNA expression in A549
    Cells by FAK 15 mer antisense oligonucleotides
    SEQ GENE
    ISIS ID TARGET % mRNA % mRNA
    No: NO: REGION EXPRESSION INHIBITION
    control 100%  0%
    15393 23 5′-UTR  40% 60%
    15433 23 5′-UTR 160%
    15431 24 5′-UTR  59% 41%
    15435 24 5′-UTR 121%
    15396 25 5′-UTR  76% 24%
    15437 25 5′-UTR 123%
    15398 26 5′-UTR  72% 28%
    15439 26 5′-UTR  64% 36%
    15400 27 5′-UTR  79% 21%
    15441 27 5′-UTR  66% 34%
    15402 28 5′-UTR  69% 31%
    15443 28 5′-UTR  99%  1%
    15404 29 5′-UTR  70% 30%
    15445 29 5′-UTR 151%
    15406 30 start  32% 68%
    15447 30 start  69% 31%
    15408 31 coding  35% 65%
    15449 31 coding  89% 11%
    15410 32 coding  67% 33%
    15451 32 coding 142%
    15412 33 coding  43% 57%
    15453 33 coding 115%
    15414 34 coding  64% 36%
    15455 34 coding  59% 41%
    15416 35 coding  69% 31%
    15457 35 coding 121%
    15418 36 stop 140%
    15459 36 stop  72% 28%
    15420 37 3′-UTR 158%
    15461 37 3′-UTR  62% 38%
    15422 38 3′-UTR 153%
    15463 38 3′-UTR  91%  9%
    15424 39 3′-UTR 207%
    15465 39 3′-UTR  88% 12%
    15426 40 3′-UTR 171%
    15467 40 3′-UTR 105%
    15428 41 3′-UTR  95%  5%
    15469 41 3′-UTR  96%  4%
    15430 42 3′-UTR 137%
    15471 42 3′-UTR 131%
  • Example 3 Dose Response of Antisense Phosphorothioate Oligonucleotide Effects on FAK Levels in A549 Cells
  • Several of the more active oligonucleotides were chosen for a dose response study. A549 cells were grown, treated and processed as described in Example 2, except the concentration of oligonucleotide was varied. [0123]
  • Results are shown in Table 7. Many oligonucleotides showed IC[0124] 50s of 50 nM or less and maximal inhibition seen was 95%.
    TABLE 7
    Dose Response of A549 cells to FAK
    Phosphorothioate Oligonucleotides
    SEQ ID ASO Gene % mRNA % mRNA
    ISIS # NO: Target Dose Expression Inhibition
    control 100.0%
    15932  3 5′-UTR 50 nM 80.3% 19.7%
    200 nM 41.6% 58.4%
    400 nM 28.3% 71.7%
    15393 23 5′-UTR 50 nM 116.6%
    200 nM 87.8% 12.2%
    400 nM 60.7% 39.3%
    15401  8 5′UTR 50 nM 31.9% 68.1%
    200 nM 26.8% 73.2%
    400 nM 20.4% 79.6%
    15403  9 5′-UTR 50 nM 82.7% 17.3%
    200 nM 27.8% 72.2%
    400 nM 18.6% 81.4%
    15406 30 start 50 nM 51.6% 48.4%
    200 nM 40.5% 59.5%
    400 nM 39.3% 60.7%
    15408 31 coding 50 nM 47.7% 52.3%
    200 nM 67.8% 32.2%
    400 nM 53.2% 46.8%
    15409 12 coding 50 nM 30.1% 69.9%
    200 nM 29.7% 70.3%
    400 nM 18.9% 81.1%
    15413 14 coding 50 nM 45.6% 54.4%
    200 nM 21.6% 78.4%
    400 nM 20.6% 79.4%
    15415 15 coding 50 nM 46.9% 53.1%
    200 nM 18.0% 82.0%
    400 nM 8.0% 92.0%
    15421 18 3′-UTR 50 nM 25.0% 75.0%
    200 nM 14.8% 85.2%
    400 nM 5.0% 95.0%
  • A dose response experiment on protein levels was done with two oligonucleotides. A549 cells were grown and treated as described in Example 2 except the concentration was varied as shown in Table 3. The LIPOFECTIN® to oligonucleotide ratio was maintained at 3 mg/ml LIPOFECTIN® per 100 nM oligonucleotide. FAK protein levels were determined 48 hours after antisense treatment in whole cell lysates by anti-FAK blotting. Cells on 10 cm plates were lysed with 0.5 ml modified RIPA lysis buffer, diluted with 0.5 ml HNTG buffer (50 mM HEPES, pH 7.4, 150 mM NaCl, 0.1% Triton X-100, 10% glycerol), incubated with agarose beads, and cleared by centrifugation. Immunoprecipitations with a polyclonal FAK antibody (Salk Institute of Biological Studies, La Jolla, Calif.; additional FAK antibodies available from Upstate Biotechnology Incorporated, Lake Placid, N.Y.) were performed for 4 hr at 4° C., collected on protein A (Repligen, Cambridge, Mass.) or protein G-plus (Calbiochem) agarose beads, and the precipitated protein complexes were washed at 4° C. in Triton only lysis buffer (modified RIPA without sodium deoxycholate and SDS) followed by washing in HNTG buffer prior to direct analysis by SDS-PAGE. For immunoblotting, proteins were transferred to polyvinylidene fluoride membranes (Millipore) and incubated with a 1:1000 dilution of polyclonal antibody for 2 hr at room temperature. Bound primary antibody was visualized by enhanced chemiluminescent detection. [0125]
  • Results are shown in Table 8. [0126]
    TABLE 8
    Dose Response of A549 cells to FAK
    Phosphorothioate Oligonucleotides
    SEQ ID ASO Gene % protein % protein
    ISIS # NO: Target Dose Expression Inhibition
    control 100% 
    15409 12 coding 25 nM 60% 40%
    100 nM 57% 43%
    200 nM 23% 77%
    15421 18 3′-UTR 25 nM 73% 27%
    100 nM 34% 66%
    200 nM 24% 76%
  • Example 4 Effect of FAK Antisense Phosphorothioate Oligonucleotides on Growth Factor Stimulated Migration and Invasion
  • Integrin-regulated focal adhesion kinase (FAK) is an important component of epidermal (EGF) and platelet-drived (PDGF) growth factor-induced motility of primary fibroblasts, smooth muscle, and adenocarcinoma cells. To measure the effect of FAK antisense oligonucleotides on cell migration, a modified Boyden chamber (Millipore, Bedford, Mass.) assay was used (Sieg, D. J., et al., [0127] J. Cell Sci., 1999, 112, 2677-2691). Both membrane sides were coated with rat tail collagen (5 ?g/ml in PBS, Boehringer Mannheim) for 2 hr at 37° C., washed with PBS, and the chambers were placed into 24 well dishes containing migration media (0.5 ml DMEM containing 0.5% BSA) with or without human recombinant PDGF-BB, EGF, or basic-FGF (Calbiochem, San Diego, Calif.) at the indicated concentrations. Serum-starved A549 cells (1×105 cells in 0.3 ml migration media) were added to the upper chamber and after 3 hr at 37° C., the cells on the membrane upper surface were removed by a cotton tip applicator, the migratory cells on the lower membrane surface were fixed, stained (0.1% crystal violet, 0.1 M borate pH 9.0, 2% EtOH), and the dye eluted for absorbance measurements at 600 nM. Individual experiments represent the average from three individual chambers. Background levels of cell migration (less than 5% of total) in the absence of chemotaxis stimuli (0.5% BSA only) were subtracted from all points.
  • Results are shown in Table 9. ISIS 17636 (SEQ ID NO. 43) is a five base mismatch control oligonucleotide for ISIS 15421 (SEQ ID NO. 18). [0128]
    TABLE 9
    Effect of FAK Antisense Phosphorothioate
    Oligonucleotides on EGF-Stimulated Cell Migration
    SEQ ID ASO Gene EGF
    ISIS # NO: Target (ng/ml) A600
    control 2.5 0.74
    15421 18 3′-UTR 0.26
    17636 43 control 0.90
    control 5.0 0.89
    15421 18 3′-UTR 0.25
    17636 43 control 0.77
  • FAK antisense oligonucleotides were tested in an in vitro invasion assay using an ˜1 mm MATRIGEL® (Becton Dickinson, Franklin Lakes, N.J.) basement membrane barrier (Albini, A., [0129] Pathol. Oncol. Res., 1998, 4, 230-241). Migration chambers were coated with the indicated concentration of MATRIGEL®, dried under laminar flow and then rehydrated with cold serum free DMEM for 90 min on an orbital shaker. A549 cells were grown and transfected as described in Example 2. Cells (1×105) were then placed onto the MATRIGEL® coated membrane and allowed to invade through the MATRIGEL® towards a 10% FBS chemoattractant for the indicated times. Cells that invaded through the MATRIGEL® were visualized by crystal violet staining as detailed in the migration assay. The amount of MATRIGEL® was varied in the assay to show that invasion was being measured and that the migration was not serum-induced.
  • Results are shown in Table 10. [0130]
    TABLE 10
    Effect of FAK Antisense Phosphorothioate
    Oligonucleotides on Tumor Cell Invasion
    SEQ ID ASO Gene MATRIGELR Migration
    ISIS # NO: Target (μg/chamber) (A600)
    control  0 8.3
    15421 18 3′-UTR 2.8
    17636 43 control 9.9
    control 15 4.5
    15421 18 3′-UTR 2.0
    17636 43 control 4.3
    control 26 1.6
    15421 18 3′-UTR 0.7
    17636 43 control 1.3
  • Example 5 FAK Antisense Oligonucleotides in a Retinal Neovascularization Model
  • FAK antisense oligonucleotides were tested in a rabbit model of retinal neovascularization (Kimura, H., et al., [0131] Invest. Opthalmol. Vis. Sci., 1995, 36, 2110-2119). In this model, growth factors are encapsulated and injected beneath the retina.
  • Eight male Dutch Belt rabbits and one male Black Satin/New Zealand White Cross rabbit were used in this study. ISIS 15409 (SEQ ID NO. 12) was administered intravitreally by injection, once prior to surgical implantation of the polymeric pellets and once during pellet implantation. Retinal neovascularization was monitored by indirect opthalmolscopy and documented by fundus photography. Retinal neovascularization was graded on a scale from 1 to 5, with one being normal and five showing retinal hemorrhaging and/or detachment. In animals injected with saline and the growth factor containing pellets, evidence of retinal neovascularization could be detected in the first week and retinal hemorrhaging began by the end of the third week. Animals receiving the antisense FAK oligonucleotide showed no evidence of retinal neovascularization over a four week period. [0132]
  • Example 6 Effect of FAK Antisense Phosphorothioate Oligonucleotide (ISIS 15421) Alone and in Combination with 5-Flurouracil on the Viability of Melanoma Cell Lines
  • Inhibition of FAK in tumor cell lines causes cell rounding, loss of adhesion, and apoptosis which suggests a role for these inhibitors in the treatment of metastatic conditions. In these studies, an antisense inhibitor of FAK was tested alone and in combination with the chemotherapeutic agent, 5-FU for its effects on melanoma cell line viability. [0133]
  • C8161 and BL human melanoma cell lines were treated with ISIS 15421 (SEQ. ID. NO 18) or a control oligonucleotide, ISIS 29848, a 20-mer random oligonucleotide (NNNNNNNNNNNNNNNNNNNN, wherein each N is a mixture of A, C, G and T; herein incorporated as SEQ ID NO: 44) using the lipofectin protocol described herein. Oligonucleotides were transfected for four hours at 300 nM in lipofectin reagent and 5-FU (200 μg/mL; SIGMA) was added after the incubation for 20 hours. Cell viability was determined by the MTT assay. Loss of adhesion and apoptosis were determined by cell counting and the TUNEL assay, respectively. FAK expression was assayed by Western blot, probing with the anti-FAK clone 4.47 antibody (Upstate Biotechnology, Lake Placid, N.Y.). [0134]
  • In The BL melanoma cell line, treatment with ISIS 15421 resulted in a 23% reduction in cell viability compared to control (p<0.0001). Addition of 5-FU to the antisense treated cells resulted in a significant further reduction in cell viability (69%; p<0.0001) compared to treatment with ISIS 15421 or 5-FU alone (4.4% reduction; p=0.15) or the control oligonucleotide, ISIS 29848. Similar results were seen with the C8161 cell line. [0135]
  • In both cell lines, reduction in cell viability was accompanied by a proportional loss of cell adhesion and an increase in apoptosis. Western blots showed that treatment with ISIS 15421 resulted in a decrease of FAK protein expression. FAK protein levels were decreased in BL melanoma cells upon treatment with 5-FU alone and were undetectable upon treatment with the combination of ISIS 15421 and 5-FU. These studies suggest that ISIS 15421, in combination with the chemotherapeutic agent 5-FU, may be a useful in the treatment of melanoma. [0136]
  • Example 7 Effect of FAK Antisense Phosphorothioate Oligonucleotide (ISIS 15421) on Human Melanoma Xenograft Tumor Growth in Mice
  • Another model used to investigate the efficacy of antisense oligonucleotides on tumor growth involves the use of mice transplanted with human cancer cells or cell line tumors. In these experiments human C8161 melanoma tumor xenografts were transplanted onto the side of nude mice with sutures or surgical staples. Mice were treated with ISIS 15421 (SEQ ID NO. 18) or the control ISIS 29848 (SEQ ID No. 44) over a 28 day treatment course. [0137]
  • At the end of the timecourse, mice were sacrificed and tumor volumes measured. Tumor volumes in the antisense treated mice were significantly smaller than tumor volumes in control-treated mice with no observation of toxicity to the mice. Additionally, one third of the control-treated mice had grossly evident intraperitoneal metastases, while none of the antisense-treated mice displayed such metastases. These studies suggest that antisense oligonucleotides represent potential chemotherapeutic agents in the treatment of melanoma and the prevention of tumor metastasis. [0138]
  • 0
    SEQUENCE LISTING
    <160> NUMBER OF SEQ ID NOS: 44
    <210> SEQ ID NO 1
    <211> LENGTH: 3791
    <212> TYPE: DNA
    <213> ORGANISM: Homo sapiens
    <220> FEATURE:
    <221> NAME/KEY: CDS
    <222> LOCATION: (233)..(3391)
    <300> PUBLICATION INFORMATION:
    <303> JOURNAL: DNA
    <304> VOLUME: 12
    <305> ISSUE: 9
    <306> PAGES: 823-830
    <307> DATE: 1993-11
    <308> DATABASE ACCESSION NUMBER: L13616/Genbank
    <309> DATABASE ENTRY DATE: 1995-01-02
    <400> SEQUENCE: 1
    cgaccactgt gagcccgcgg cgtgaggcgt cgggaggaag cgcggctgct gtcgcccagc 60
    gccgccccgt cgtcgtctgc cttcgcttca cggcgccgag ccgcggtccg agcagaactg 120
    gggctccctt gcatcttcca gttacaaatt cagtgccttc tgcagtttcc ccagagctcc 180
    tcaagaataa cggaagggag aatatgacag atacctagca tctagcaaaa ta atg gca 238
    Met Ala
    1
    gct gct tac ctt gac ccc aac ttg aat cac aca cca aat tcg agt act 286
    Ala Ala Tyr Leu Asp Pro Asn Leu Asn His Thr Pro Asn Ser Ser Thr
    5 10 15
    aag act cac ctg ggt act ggt atg gaa cgt tct cct ggt gca atg gag 334
    Lys Thr His Leu Gly Thr Gly Met Glu Arg Ser Pro Gly Ala Met Glu
    20 25 30
    cga gta tta aag gtc ttt cat tat ttt gaa agc aat agt gag cca acc 382
    Arg Val Leu Lys Val Phe His Tyr Phe Glu Ser Asn Ser Glu Pro Thr
    35 40 45 50
    acc tgg gcc agt att atc agg cat gga gat gct act gat gtc agg ggc 430
    Thr Trp Ala Ser Ile Ile Arg His Gly Asp Ala Thr Asp Val Arg Gly
    55 60 65
    atc att cag aag ata gtg gac agt cac aaa gta aag cat gtg gcc tgc 478
    Ile Ile Gln Lys Ile Val Asp Ser His Lys Val Lys His Val Ala Cys
    70 75 80
    tat gga ttc cgc ctc agt cac ctg cgg tca gag gag gtt cac tgg ctt 526
    Tyr Gly Phe Arg Leu Ser His Leu Arg Ser Glu Glu Val His Trp Leu
    85 90 95
    cac gtg gat atg ggc gtc tcc agt gtg agg gag aag tat gag ctt gct 574
    His Val Asp Met Gly Val Ser Ser Val Arg Glu Lys Tyr Glu Leu Ala
    100 105 110
    cac cca cca gag gag tgg aaa tat gaa ttg aga att cgt tat ttg cca 622
    His Pro Pro Glu Glu Trp Lys Tyr Glu Leu Arg Ile Arg Tyr Leu Pro
    115 120 125 130
    aaa gga ttt cta aac cag ttt act gaa gat aag cca act ttg aat ttc 670
    Lys Gly Phe Leu Asn Gln Phe Thr Glu Asp Lys Pro Thr Leu Asn Phe
    135 140 145
    ttc tat caa cag gtg aag agc gat tat atg tta gag ata gct gat caa 718
    Phe Tyr Gln Gln Val Lys Ser Asp Tyr Met Leu Glu Ile Ala Asp Gln
    150 155 160
    gtg gac cag gaa att gct ttg aag ttg ggt tgt cta gaa ata cgg cga 766
    Val Asp Gln Glu Ile Ala Leu Lys Leu Gly Cys Leu Glu Ile Arg Arg
    165 170 175
    tca tac tgg gag atg cgg ggc aat gca cta gaa aag aag tct aac tat 814
    Ser Tyr Trp Glu Met Arg Gly Asn Ala Leu Glu Lys Lys Ser Asn Tyr
    180 185 190
    gaa gta tta gaa aaa gat gtt ggt tta aag cga ttt ttt cct aag agt 862
    Glu Val Leu Glu Lys Asp Val Gly Leu Lys Arg Phe Phe Pro Lys Ser
    195 200 205 210
    tta ctg gat tct gtc aag gcc aaa aca cta aga aaa ctg atc caa caa 910
    Leu Leu Asp Ser Val Lys Ala Lys Thr Leu Arg Lys Leu Ile Gln Gln
    215 220 225
    aca ttt aga caa ttt gcc aac ctt aat aga gaa gaa agt att ctg aaa 958
    Thr Phe Arg Gln Phe Ala Asn Leu Asn Arg Glu Glu Ser Ile Leu Lys
    230 235 240
    ttc ttt gag atc ctg tct cca gtc tac aga ttt gat aag gaa tgc ttc 1006
    Phe Phe Glu Ile Leu Ser Pro Val Tyr Arg Phe Asp Lys Glu Cys Phe
    245 250 255
    aag tgt gct ctt ggt tca agc tgg att att tca gtg gaa ctg gca atc 1054
    Lys Cys Ala Leu Gly Ser Ser Trp Ile Ile Ser Val Glu Leu Ala Ile
    260 265 270
    ggc cca gaa gaa gga atc agt tac cta acg gac aag ggc tgc aat ccc 1102
    Gly Pro Glu Glu Gly Ile Ser Tyr Leu Thr Asp Lys Gly Cys Asn Pro
    275 280 285 290
    aca cat ctt gct gac ttc act caa gtg caa acc att cag tat tca aac 1150
    Thr His Leu Ala Asp Phe Thr Gln Val Gln Thr Ile Gln Tyr Ser Asn
    295 300 305
    agt gaa gac aag gac aga aaa gga atg cta caa cta aaa ata gca ggt 1198
    Ser Glu Asp Lys Asp Arg Lys Gly Met Leu Gln Leu Lys Ile Ala Gly
    310 315 320
    gca ccc gag cct ctg aca gtg acg gca cca tcc cta acc att gcg gag 1246
    Ala Pro Glu Pro Leu Thr Val Thr Ala Pro Ser Leu Thr Ile Ala Glu
    325 330 335
    aat atg gct gac cta ata gat ggg tac tgc cgg ctg gtg aat gga acc 1294
    Asn Met Ala Asp Leu Ile Asp Gly Tyr Cys Arg Leu Val Asn Gly Thr
    340 345 350
    tcg cag tca ttt atc atc aga cct cag aaa gaa ggt gaa cgg gct ttg 1342
    Ser Gln Ser Phe Ile Ile Arg Pro Gln Lys Glu Gly Glu Arg Ala Leu
    355 360 365 370
    cca tca ata cca aag ttg gcc aac agc gaa aag caa ggc atg cgg aca 1390
    Pro Ser Ile Pro Lys Leu Ala Asn Ser Glu Lys Gln Gly Met Arg Thr
    375 380 385
    cac gcc gtc tct gtg tca gaa aca gat gat tat gct gag att ata gat 1438
    His Ala Val Ser Val Ser Glu Thr Asp Asp Tyr Ala Glu Ile Ile Asp
    390 395 400
    gaa gaa gat act tac acc atg ccc tca acc agg gat tat gag att caa 1486
    Glu Glu Asp Thr Tyr Thr Met Pro Ser Thr Arg Asp Tyr Glu Ile Gln
    405 410 415
    aga gaa aga ata gaa ctt gga cga tgt att gga gaa ggc caa ttt gga 1534
    Arg Glu Arg Ile Glu Leu Gly Arg Cys Ile Gly Glu Gly Gln Phe Gly
    420 425 430
    gat gta cat caa ggc att tat atg agt cca gag aat cca gct ttg gcg 1582
    Asp Val His Gln Gly Ile Tyr Met Ser Pro Glu Asn Pro Ala Leu Ala
    435 440 445 450
    gtt gca att aaa aca tgt aaa aac tgt act tcg gac agc gtg aga gag 1630
    Val Ala Ile Lys Thr Cys Lys Asn Cys Thr Ser Asp Ser Val Arg Glu
    455 460 465
    aaa ttt ctt caa gaa gcc tta aca atg cgt cag ttt gac cat cct cat 1678
    Lys Phe Leu Gln Glu Ala Leu Thr Met Arg Gln Phe Asp His Pro His
    470 475 480
    att gtg aag ctg att gga gtc atc aca gag aat cct gtc tgg ata atc 1726
    Ile Val Lys Leu Ile Gly Val Ile Thr Glu Asn Pro Val Trp Ile Ile
    485 490 495
    atg gag ctg tgc aca ctt gga gag ctg agg tca ttt ttg caa gta agg 1774
    Met Glu Leu Cys Thr Leu Gly Glu Leu Arg Ser Phe Leu Gln Val Arg
    500 505 510
    aaa tac agt ttg gat cta gca tct ttg atc ctg tat gcc tat cag ctt 1822
    Lys Tyr Ser Leu Asp Leu Ala Ser Leu Ile Leu Tyr Ala Tyr Gln Leu
    515 520 525 530
    agt aca gct ctt gca tat cta gag agc aaa aga ttt gta cac agg gac 1870
    Ser Thr Ala Leu Ala Tyr Leu Glu Ser Lys Arg Phe Val His Arg Asp
    535 540 545
    att gct gct cgg aat gtt ctg gtg tcc tca aat gat tgt gta aaa tta 1918
    Ile Ala Ala Arg Asn Val Leu Val Ser Ser Asn Asp Cys Val Lys Leu
    550 555 560
    gga gac ttt gga tta tcc cga tat atg gaa gat agt act tac tac aaa 1966
    Gly Asp Phe Gly Leu Ser Arg Tyr Met Glu Asp Ser Thr Tyr Tyr Lys
    565 570 575
    gct tcc aaa gga aaa ttg cct att aaa tgg atg gct cca gag tca atc 2014
    Ala Ser Lys Gly Lys Leu Pro Ile Lys Trp Met Ala Pro Glu Ser Ile
    580 585 590
    aat ttt cga cgt ttt acc tca gct agt gac gta tgg atg ttt ggt gtg 2062
    Asn Phe Arg Arg Phe Thr Ser Ala Ser Asp Val Trp Met Phe Gly Val
    595 600 605 610
    tgt atg tgg gag ata ctg atg cat ggt gtg aag cct ttt caa gga gtg 2110
    Cys Met Trp Glu Ile Leu Met His Gly Val Lys Pro Phe Gln Gly Val
    615 620 625
    aag aac aat gat gta atc ggt cga att gaa aat ggg gaa aga tta cca 2158
    Lys Asn Asn Asp Val Ile Gly Arg Ile Glu Asn Gly Glu Arg Leu Pro
    630 635 640
    atg cct cca aat tgt cct cct acc ctc tac agc ctt atg acg aaa tgc 2206
    Met Pro Pro Asn Cys Pro Pro Thr Leu Tyr Ser Leu Met Thr Lys Cys
    645 650 655
    tgg gcc tat gac ccc agc agg cgg ccc agg ttt act gaa ctt aaa gct 2254
    Trp Ala Tyr Asp Pro Ser Arg Arg Pro Arg Phe Thr Glu Leu Lys Ala
    660 665 670
    cag ctc agc aca atc ctg gag gaa gag aag gct cag caa gaa gag cgc 2302
    Gln Leu Ser Thr Ile Leu Glu Glu Glu Lys Ala Gln Gln Glu Glu Arg
    675 680 685 690
    atg agg atg gag tcc aga aga cag gcc aca gtg tcc tgg gac tcc gga 2350
    Met Arg Met Glu Ser Arg Arg Gln Ala Thr Val Ser Trp Asp Ser Gly
    695 700 705
    ggg tct gat gaa gca ccg ccc aag ccc agc aga ccg ggt tat ccc agt 2398
    Gly Ser Asp Glu Ala Pro Pro Lys Pro Ser Arg Pro Gly Tyr Pro Ser
    710 715 720
    ccg agg tcc agc gaa gga ttt tat ccc agc cca cag cac atg gta caa 2446
    Pro Arg Ser Ser Glu Gly Phe Tyr Pro Ser Pro Gln His Met Val Gln
    725 730 735
    acc aat cat tac cag gtt tct ggc tac cct ggt tca cat gga atc aca 2494
    Thr Asn His Tyr Gln Val Ser Gly Tyr Pro Gly Ser His Gly Ile Thr
    740 745 750
    gcc atg gct ggc agc atc tat cca ggt cag gca tct ctt ttg gac caa 2542
    Ala Met Ala Gly Ser Ile Tyr Pro Gly Gln Ala Ser Leu Leu Asp Gln
    755 760 765 770
    aca gat tca tgg aat cat aga cct cag gag ata gca atg tgg cag ccc 2590
    Thr Asp Ser Trp Asn His Arg Pro Gln Glu Ile Ala Met Trp Gln Pro
    775 780 785
    aat gtg gag gac tct aca gta ttg gac ctg cga ggg att ggg caa gtg 2638
    Asn Val Glu Asp Ser Thr Val Leu Asp Leu Arg Gly Ile Gly Gln Val
    790 795 800
    ttg cca acc cat ctg atg gaa gag cgt cta atc cga cag caa cag gaa 2686
    Leu Pro Thr His Leu Met Glu Glu Arg Leu Ile Arg Gln Gln Gln Glu
    805 810 815
    atg gaa gaa gat cag cgc tgg ctg gaa aaa gag gaa aga ttt ctg aaa 2734
    Met Glu Glu Asp Gln Arg Trp Leu Glu Lys Glu Glu Arg Phe Leu Lys
    820 825 830
    cct gat gtg aga ctc tct cga ggc agt att gac agg gag gat gga agt 2782
    Pro Asp Val Arg Leu Ser Arg Gly Ser Ile Asp Arg Glu Asp Gly Ser
    835 840 845 850
    ctt cag ggt ccg att gga aac caa cat ata tat cag cct gtg ggt aaa 2830
    Leu Gln Gly Pro Ile Gly Asn Gln His Ile Tyr Gln Pro Val Gly Lys
    855 860 865
    cca gat cct gca gct cca cca aag aaa ccg cct cgc cct gga gct ccc 2878
    Pro Asp Pro Ala Ala Pro Pro Lys Lys Pro Pro Arg Pro Gly Ala Pro
    870 875 880
    ggt cat ctg gga agc ctt gcc agc ctc agc agc cct gct gac agc tac 2926
    Gly His Leu Gly Ser Leu Ala Ser Leu Ser Ser Pro Ala Asp Ser Tyr
    885 890 895
    aac gag ggt gtc aag ctt cag ccc cag gaa atc agc ccc cct cct act 2974
    Asn Glu Gly Val Lys Leu Gln Pro Gln Glu Ile Ser Pro Pro Pro Thr
    900 905 910
    gcc aac ctg gac cgg tcg aat gat aag gtg tac gag aat gtg acg ggc 3022
    Ala Asn Leu Asp Arg Ser Asn Asp Lys Val Tyr Glu Asn Val Thr Gly
    915 920 925 930
    ctg gtg aaa gct gtc atc gag atg tcc agt aaa atc cag cca gcc cca 3070
    Leu Val Lys Ala Val Ile Glu Met Ser Ser Lys Ile Gln Pro Ala Pro
    935 940 945
    cca gag gag tat gtc cct atg gtg aag gaa gtc ggc ttg gcc ctg agg 3118
    Pro Glu Glu Tyr Val Pro Met Val Lys Glu Val Gly Leu Ala Leu Arg
    950 955 960
    aca tta ttg gcc act gtg gat gag acc att ccc ctc cta cca gcc agc 3166
    Thr Leu Leu Ala Thr Val Asp Glu Thr Ile Pro Leu Leu Pro Ala Ser
    965 970 975
    acc cac cga gag att gag atg gca cag aag cta ttg aac tct gac ctg 3214
    Thr His Arg Glu Ile Glu Met Ala Gln Lys Leu Leu Asn Ser Asp Leu
    980 985 990
    ggt gag ctc atc aac aag atg aaa ctg gcc cag cag tat gtc atg acc 3262
    Gly Glu Leu Ile Asn Lys Met Lys Leu Ala Gln Gln Tyr Val Met Thr
    995 1000 1005 1010
    agc ctc cag caa gag tac aaa aag caa atg ctg act gct gct cac gcc 3310
    Ser Leu Gln Gln Glu Tyr Lys Lys Gln Met Leu Thr Ala Ala His Ala
    1015 1020 1025
    ctg gct gtg gat gcc aaa aac tta ctc gat gtc att gac caa gca aga 3358
    Leu Ala Val Asp Ala Lys Asn Leu Leu Asp Val Ile Asp Gln Ala Arg
    1030 1035 1040
    ctg aaa atg ctt ggg cag acg aga cca cac tga gcctccccta ggagcacgtc 3411
    Leu Lys Met Leu Gly Gln Thr Arg Pro His
    1045 1050
    ttgctaccct cttttgaaga tgttctctag ccttccacca gcagcgagga attaaccctg 3471
    tgtcctcagt cgccagcact tacagctcca acttttttga atgaccatct ggttgaaaaa 3531
    tctttctcat ataagtttaa ccacactttg atttgggttc attttttgtt ttgttttttt 3591
    caatcatgat attcagaaaa atccaggatc caaaatgtgg cgtttttcta agaatgaaaa 3651
    ttatatgtaa gcttttaagc atcatgaaga acaatttatg ttcacattaa gatacgttct 3711
    aaagggggat ggccaagggg tgacatctta attcctaaac taccttagct gcatagtgga 3771
    agaggagagc tagaagcaaa 3791
    <210> SEQ ID NO 2
    <211> LENGTH: 1052
    <212> TYPE: PRT
    <213> ORGANISM: Homo sapiens
    <400> SEQUENCE: 2
    Met Ala Ala Ala Tyr Leu Asp Pro Asn Leu Asn His Thr Pro Asn Ser
    1 5 10 15
    Ser Thr Lys Thr His Leu Gly Thr Gly Met Glu Arg Ser Pro Gly Ala
    20 25 30
    Met Glu Arg Val Leu Lys Val Phe His Tyr Phe Glu Ser Asn Ser Glu
    35 40 45
    Pro Thr Thr Trp Ala Ser Ile Ile Arg His Gly Asp Ala Thr Asp Val
    50 55 60
    Arg Gly Ile Ile Gln Lys Ile Val Asp Ser His Lys Val Lys His Val
    65 70 75 80
    Ala Cys Tyr Gly Phe Arg Leu Ser His Leu Arg Ser Glu Glu Val His
    85 90 95
    Trp Leu His Val Asp Met Gly Val Ser Ser Val Arg Glu Lys Tyr Glu
    100 105 110
    Leu Ala His Pro Pro Glu Glu Trp Lys Tyr Glu Leu Arg Ile Arg Tyr
    115 120 125
    Leu Pro Lys Gly Phe Leu Asn Gln Phe Thr Glu Asp Lys Pro Thr Leu
    130 135 140
    Asn Phe Phe Tyr Gln Gln Val Lys Ser Asp Tyr Met Leu Glu Ile Ala
    145 150 155 160
    Asp Gln Val Asp Gln Glu Ile Ala Leu Lys Leu Gly Cys Leu Glu Ile
    165 170 175
    Arg Arg Ser Tyr Trp Glu Met Arg Gly Asn Ala Leu Glu Lys Lys Ser
    180 185 190
    Asn Tyr Glu Val Leu Glu Lys Asp Val Gly Leu Lys Arg Phe Phe Pro
    195 200 205
    Lys Ser Leu Leu Asp Ser Val Lys Ala Lys Thr Leu Arg Lys Leu Ile
    210 215 220
    Gln Gln Thr Phe Arg Gln Phe Ala Asn Leu Asn Arg Glu Glu Ser Ile
    225 230 235 240
    Leu Lys Phe Phe Glu Ile Leu Ser Pro Val Tyr Arg Phe Asp Lys Glu
    245 250 255
    Cys Phe Lys Cys Ala Leu Gly Ser Ser Trp Ile Ile Ser Val Glu Leu
    260 265 270
    Ala Ile Gly Pro Glu Glu Gly Ile Ser Tyr Leu Thr Asp Lys Gly Cys
    275 280 285
    Asn Pro Thr His Leu Ala Asp Phe Thr Gln Val Gln Thr Ile Gln Tyr
    290 295 300
    Ser Asn Ser Glu Asp Lys Asp Arg Lys Gly Met Leu Gln Leu Lys Ile
    305 310 315 320
    Ala Gly Ala Pro Glu Pro Leu Thr Val Thr Ala Pro Ser Leu Thr Ile
    325 330 335
    Ala Glu Asn Met Ala Asp Leu Ile Asp Gly Tyr Cys Arg Leu Val Asn
    340 345 350
    Gly Thr Ser Gln Ser Phe Ile Ile Arg Pro Gln Lys Glu Gly Glu Arg
    355 360 365
    Ala Leu Pro Ser Ile Pro Lys Leu Ala Asn Ser Glu Lys Gln Gly Met
    370 375 380
    Arg Thr His Ala Val Ser Val Ser Glu Thr Asp Asp Tyr Ala Glu Ile
    385 390 395 400
    Ile Asp Glu Glu Asp Thr Tyr Thr Met Pro Ser Thr Arg Asp Tyr Glu
    405 410 415
    Ile Gln Arg Glu Arg Ile Glu Leu Gly Arg Cys Ile Gly Glu Gly Gln
    420 425 430
    Phe Gly Asp Val His Gln Gly Ile Tyr Met Ser Pro Glu Asn Pro Ala
    435 440 445
    Leu Ala Val Ala Ile Lys Thr Cys Lys Asn Cys Thr Ser Asp Ser Val
    450 455 460
    Arg Glu Lys Phe Leu Gln Glu Ala Leu Thr Met Arg Gln Phe Asp His
    465 470 475 480
    Pro His Ile Val Lys Leu Ile Gly Val Ile Thr Glu Asn Pro Val Trp
    485 490 495
    Ile Ile Met Glu Leu Cys Thr Leu Gly Glu Leu Arg Ser Phe Leu Gln
    500 505 510
    Val Arg Lys Tyr Ser Leu Asp Leu Ala Ser Leu Ile Leu Tyr Ala Tyr
    515 520 525
    Gln Leu Ser Thr Ala Leu Ala Tyr Leu Glu Ser Lys Arg Phe Val His
    530 535 540
    Arg Asp Ile Ala Ala Arg Asn Val Leu Val Ser Ser Asn Asp Cys Val
    545 550 555 560
    Lys Leu Gly Asp Phe Gly Leu Ser Arg Tyr Met Glu Asp Ser Thr Tyr
    565 570 575
    Tyr Lys Ala Ser Lys Gly Lys Leu Pro Ile Lys Trp Met Ala Pro Glu
    580 585 590
    Ser Ile Asn Phe Arg Arg Phe Thr Ser Ala Ser Asp Val Trp Met Phe
    595 600 605
    Gly Val Cys Met Trp Glu Ile Leu Met His Gly Val Lys Pro Phe Gln
    610 615 620
    Gly Val Lys Asn Asn Asp Val Ile Gly Arg Ile Glu Asn Gly Glu Arg
    625 630 635 640
    Leu Pro Met Pro Pro Asn Cys Pro Pro Thr Leu Tyr Ser Leu Met Thr
    645 650 655
    Lys Cys Trp Ala Tyr Asp Pro Ser Arg Arg Pro Arg Phe Thr Glu Leu
    660 665 670
    Lys Ala Gln Leu Ser Thr Ile Leu Glu Glu Glu Lys Ala Gln Gln Glu
    675 680 685
    Glu Arg Met Arg Met Glu Ser Arg Arg Gln Ala Thr Val Ser Trp Asp
    690 695 700
    Ser Gly Gly Ser Asp Glu Ala Pro Pro Lys Pro Ser Arg Pro Gly Tyr
    705 710 715 720
    Pro Ser Pro Arg Ser Ser Glu Gly Phe Tyr Pro Ser Pro Gln His Met
    725 730 735
    Val Gln Thr Asn His Tyr Gln Val Ser Gly Tyr Pro Gly Ser His Gly
    740 745 750
    Ile Thr Ala Met Ala Gly Ser Ile Tyr Pro Gly Gln Ala Ser Leu Leu
    755 760 765
    Asp Gln Thr Asp Ser Trp Asn His Arg Pro Gln Glu Ile Ala Met Trp
    770 775 780
    Gln Pro Asn Val Glu Asp Ser Thr Val Leu Asp Leu Arg Gly Ile Gly
    785 790 795 800
    Gln Val Leu Pro Thr His Leu Met Glu Glu Arg Leu Ile Arg Gln Gln
    805 810 815
    Gln Glu Met Glu Glu Asp Gln Arg Trp Leu Glu Lys Glu Glu Arg Phe
    820 825 830
    Leu Lys Pro Asp Val Arg Leu Ser Arg Gly Ser Ile Asp Arg Glu Asp
    835 840 845
    Gly Ser Leu Gln Gly Pro Ile Gly Asn Gln His Ile Tyr Gln Pro Val
    850 855 860
    Gly Lys Pro Asp Pro Ala Ala Pro Pro Lys Lys Pro Pro Arg Pro Gly
    865 870 875 880
    Ala Pro Gly His Leu Gly Ser Leu Ala Ser Leu Ser Ser Pro Ala Asp
    885 890 895
    Ser Tyr Asn Glu Gly Val Lys Leu Gln Pro Gln Glu Ile Ser Pro Pro
    900 905 910
    Pro Thr Ala Asn Leu Asp Arg Ser Asn Asp Lys Val Tyr Glu Asn Val
    915 920 925
    Thr Gly Leu Val Lys Ala Val Ile Glu Met Ser Ser Lys Ile Gln Pro
    930 935 940
    Ala Pro Pro Glu Glu Tyr Val Pro Met Val Lys Glu Val Gly Leu Ala
    945 950 955 960
    Leu Arg Thr Leu Leu Ala Thr Val Asp Glu Thr Ile Pro Leu Leu Pro
    965 970 975
    Ala Ser Thr His Arg Glu Ile Glu Met Ala Gln Lys Leu Leu Asn Ser
    980 985 990
    Asp Leu Gly Glu Leu Ile Asn Lys Met Lys Leu Ala Gln Gln Tyr Val
    995 1000 1005
    Met Thr Ser Leu Gln Gln Glu Tyr Lys Lys Gln Met Leu Thr Ala Ala
    1010 1015 1020
    His Ala Leu Ala Val Asp Ala Lys Asn Leu Leu Asp Val Ile Asp Gln
    1025 1030 1035 1040
    Ala Arg Leu Lys Met Leu Gly Gln Thr Arg Pro His
    1045 1050
    <210> SEQ ID NO 3
    <211> LENGTH: 20
    <212> TYPE: DNA
    <213> ORGANISM: Artificial Sequence
    <220> FEATURE:
    <223> OTHER INFORMATION: antisense sequence
    <400> SEQUENCE: 3
    ccgcgggctc acagtggtcg 20
    <210> SEQ ID NO 4
    <211> LENGTH: 20
    <212> TYPE: DNA
    <213> ORGANISM: Artificial Sequence
    <220> FEATURE:
    <223> OTHER INFORMATION: antisense sequence
    <400> SEQUENCE: 4
    ggcgccgtga agcgaaggca 20
    <210> SEQ ID NO 5
    <211> LENGTH: 20
    <212> TYPE: DNA
    <213> ORGANISM: Artificial Sequence
    <220> FEATURE:
    <223> OTHER INFORMATION: antisense sequence
    <400> SEQUENCE: 5
    cagttctgct cggaccgcgg 20
    <210> SEQ ID NO 6
    <211> LENGTH: 20
    <212> TYPE: DNA
    <213> ORGANISM: Artificial Sequence
    <220> FEATURE:
    <223> OTHER INFORMATION: antisense sequence
    <400> SEQUENCE: 6
    gaaactgcag aaggcactga 20
    <210> SEQ ID NO 7
    <211> LENGTH: 20
    <212> TYPE: DNA
    <213> ORGANISM: Artificial Sequence
    <220> FEATURE:
    <223> OTHER INFORMATION: antisense sequence
    <400> SEQUENCE: 7
    ttctcccttc cgttattctt 20
    <210> SEQ ID NO 8
    <211> LENGTH: 20
    <212> TYPE: DNA
    <213> ORGANISM: Artificial Sequence
    <220> FEATURE:
    <223> OTHER INFORMATION: antisense sequence
    <400> SEQUENCE: 8
    ctagatgcta ggtatctgtc 20
    <210> SEQ ID NO 9
    <211> LENGTH: 20
    <212> TYPE: DNA
    <213> ORGANISM: Artificial Sequence
    <220> FEATURE:
    <223> OTHER INFORMATION: antisense sequence
    <400> SEQUENCE: 9
    ttttgctaga tgctaggtat 20
    <210> SEQ ID NO 10
    <211> LENGTH: 20
    <212> TYPE: DNA
    <213> ORGANISM: Artificial Sequence
    <220> FEATURE:
    <223> OTHER INFORMATION: antisense sequence
    <400> SEQUENCE: 10
    ggtaagcagc tgccattatt 20
    <210> SEQ ID NO 11
    <211> LENGTH: 20
    <212> TYPE: DNA
    <213> ORGANISM: Artificial Sequence
    <220> FEATURE:
    <223> OTHER INFORMATION: antisense sequence
    <400> SEQUENCE: 11
    agtacccagg tgagtcttag 20
    <210> SEQ ID NO 12
    <211> LENGTH: 20
    <212> TYPE: DNA
    <213> ORGANISM: Artificial Sequence
    <220> FEATURE:
    <223> OTHER INFORMATION: antisense sequence
    <400> SEQUENCE: 12
    cctgacatca gtagcatctc 20
    <210> SEQ ID NO 13
    <211> LENGTH: 20
    <212> TYPE: DNA
    <213> ORGANISM: Artificial Sequence
    <220> FEATURE:
    <223> OTHER INFORMATION: antisense sequence
    <400> SEQUENCE: 13
    gttggcttat cttcagtaaa 20
    <210> SEQ ID NO 14
    <211> LENGTH: 20
    <212> TYPE: DNA
    <213> ORGANISM: Artificial Sequence
    <220> FEATURE:
    <223> OTHER INFORMATION: antisense sequence
    <400> SEQUENCE: 14
    ggttagggat ggtgccgtca 20
    <210> SEQ ID NO 15
    <211> LENGTH: 20
    <212> TYPE: DNA
    <213> ORGANISM: Artificial Sequence
    <220> FEATURE:
    <223> OTHER INFORMATION: antisense sequence
    <400> SEQUENCE: 15
    tgttggtttc caatcggacc 20
    <210> SEQ ID NO 16
    <211> LENGTH: 20
    <212> TYPE: DNA
    <213> ORGANISM: Artificial Sequence
    <220> FEATURE:
    <223> OTHER INFORMATION: antisense sequence
    <400> SEQUENCE: 16
    ctaggggagg ctcagtgtgg 20
    <210> SEQ ID NO 17
    <211> LENGTH: 20
    <212> TYPE: DNA
    <213> ORGANISM: Artificial Sequence
    <220> FEATURE:
    <223> OTHER INFORMATION: antisense sequence
    <400> SEQUENCE: 17
    attcctcgct gctggtggaa 20
    <210> SEQ ID NO 18
    <211> LENGTH: 20
    <212> TYPE: DNA
    <213> ORGANISM: Artificial Sequence
    <220> FEATURE:
    <223> OTHER INFORMATION: antisense sequence
    <400> SEQUENCE: 18
    tttcaaccag atggtcattc 20
    <210> SEQ ID NO 19
    <211> LENGTH: 20
    <212> TYPE: DNA
    <213> ORGANISM: Artificial Sequence
    <220> FEATURE:
    <223> OTHER INFORMATION: antisense sequence
    <400> SEQUENCE: 19
    ttctgaatat catgattgaa 20
    <210> SEQ ID NO 20
    <211> LENGTH: 20
    <212> TYPE: DNA
    <213> ORGANISM: Artificial Sequence
    <220> FEATURE:
    <223> OTHER INFORMATION: antisense sequence
    <400> SEQUENCE: 20
    catgatgctt aaaagcttac 20
    <210> SEQ ID NO 21
    <211> LENGTH: 20
    <212> TYPE: DNA
    <213> ORGANISM: Artificial Sequence
    <220> FEATURE:
    <223> OTHER INFORMATION: antisense sequence
    <400> SEQUENCE: 21
    aatgtgaaca taaattgttc 20
    <210> SEQ ID NO 22
    <211> LENGTH: 20
    <212> TYPE: DNA
    <213> ORGANISM: Artificial Sequence
    <220> FEATURE:
    <223> OTHER INFORMATION: antisense sequence
    <400> SEQUENCE: 22
    aaggtagttt aggaattaag 20
    <210> SEQ ID NO 23
    <211> LENGTH: 15
    <212> TYPE: DNA
    <213> ORGANISM: Artificial Sequence
    <220> FEATURE:
    <223> OTHER INFORMATION: antisense sequence
    <400> SEQUENCE: 23
    gcgggctcac agtgg 15
    <210> SEQ ID NO 24
    <211> LENGTH: 15
    <212> TYPE: DNA
    <213> ORGANISM: Artificial Sequence
    <220> FEATURE:
    <223> OTHER INFORMATION: antisense sequence
    <400> SEQUENCE: 24
    cgccgtgaag cgaag 15
    <210> SEQ ID NO 25
    <211> LENGTH: 15
    <212> TYPE: DNA
    <213> ORGANISM: Artificial Sequence
    <220> FEATURE:
    <223> OTHER INFORMATION: antisense sequence
    <400> SEQUENCE: 25
    gttctgctcg gaccg 15
    <210> SEQ ID NO 26
    <211> LENGTH: 15
    <212> TYPE: DNA
    <213> ORGANISM: Artificial Sequence
    <220> FEATURE:
    <223> OTHER INFORMATION: antisense sequence
    <400> SEQUENCE: 26
    aactgcagaa ggcac 15
    <210> SEQ ID NO 27
    <211> LENGTH: 15
    <212> TYPE: DNA
    <213> ORGANISM: Artificial Sequence
    <220> FEATURE:
    <223> OTHER INFORMATION: antisense sequence
    <400> SEQUENCE: 27
    ctcccttccg ttatt 15
    <210> SEQ ID NO 28
    <211> LENGTH: 15
    <212> TYPE: DNA
    <213> ORGANISM: Artificial Sequence
    <220> FEATURE:
    <223> OTHER INFORMATION: antisense sequence
    <400> SEQUENCE: 28
    agatgctagg tatct 15
    <210> SEQ ID NO 29
    <211> LENGTH: 15
    <212> TYPE: DNA
    <213> ORGANISM: Artificial Sequence
    <220> FEATURE:
    <223> OTHER INFORMATION: antisense sequence
    <400> SEQUENCE: 29
    ttgctagatg ctagg 15
    <210> SEQ ID NO 30
    <211> LENGTH: 15
    <212> TYPE: DNA
    <213> ORGANISM: Artificial Sequence
    <220> FEATURE:
    <223> OTHER INFORMATION: antisense sequence
    <400> SEQUENCE: 30
    taagcagctg ccatt 15
    <210> SEQ ID NO 31
    <211> LENGTH: 15
    <212> TYPE: DNA
    <213> ORGANISM: Artificial Sequence
    <220> FEATURE:
    <223> OTHER INFORMATION: antisense sequence
    <400> SEQUENCE: 31
    tacccaggtg agtct 15
    <210> SEQ ID NO 32
    <211> LENGTH: 15
    <212> TYPE: DNA
    <213> ORGANISM: Artificial Sequence
    <220> FEATURE:
    <223> OTHER INFORMATION: antisense sequence
    <400> SEQUENCE: 32
    tgacatcagt agcat 15
    <210> SEQ ID NO 33
    <211> LENGTH: 15
    <212> TYPE: DNA
    <213> ORGANISM: Artificial Sequence
    <220> FEATURE:
    <223> OTHER INFORMATION: antisense sequence
    <400> SEQUENCE: 33
    tggcttatct tcagt 15
    <210> SEQ ID NO 34
    <211> LENGTH: 15
    <212> TYPE: DNA
    <213> ORGANISM: Artificial Sequence
    <220> FEATURE:
    <223> OTHER INFORMATION: antisense sequence
    <400> SEQUENCE: 34
    ttagggatgg tgccg 15
    <210> SEQ ID NO 35
    <211> LENGTH: 15
    <212> TYPE: DNA
    <213> ORGANISM: Artificial Sequence
    <220> FEATURE:
    <223> OTHER INFORMATION: antisense sequence
    <400> SEQUENCE: 35
    ttggtttcca atcgg 15
    <210> SEQ ID NO 36
    <211> LENGTH: 15
    <212> TYPE: DNA
    <213> ORGANISM: Artificial Sequence
    <220> FEATURE:
    <223> OTHER INFORMATION: antisense sequence
    <400> SEQUENCE: 36
    aggggaggct cagtg 15
    <210> SEQ ID NO 37
    <211> LENGTH: 15
    <212> TYPE: DNA
    <213> ORGANISM: Artificial Sequence
    <220> FEATURE:
    <223> OTHER INFORMATION: antisense sequence
    <400> SEQUENCE: 37
    tcctcgctgc tggtg 15
    <210> SEQ ID NO 38
    <211> LENGTH: 15
    <212> TYPE: DNA
    <213> ORGANISM: Artificial Sequence
    <220> FEATURE:
    <223> OTHER INFORMATION: antisense sequence
    <400> SEQUENCE: 38
    tcaaccagat ggtca 15
    <210> SEQ ID NO 39
    <211> LENGTH: 15
    <212> TYPE: DNA
    <213> ORGANISM: Artificial Sequence
    <220> FEATURE:
    <223> OTHER INFORMATION: antisense sequence
    <400> SEQUENCE: 39
    ctgaatatca tgatt 15
    <210> SEQ ID NO 40
    <211> LENGTH: 15
    <212> TYPE: DNA
    <213> ORGANISM: Artificial Sequence
    <220> FEATURE:
    <223> OTHER INFORMATION: antisense sequence
    <400> SEQUENCE: 40
    tgatgcttaa aagct 15
    <210> SEQ ID NO 41
    <211> LENGTH: 15
    <212> TYPE: DNA
    <213> ORGANISM: Artificial Sequence
    <220> FEATURE:
    <223> OTHER INFORMATION: antisense sequence
    <400> SEQUENCE: 41
    tgtgaacata aattg 15
    <210> SEQ ID NO 42
    <211> LENGTH: 15
    <212> TYPE: DNA
    <213> ORGANISM: Artificial Sequence
    <220> FEATURE:
    <223> OTHER INFORMATION: antisense sequence
    <400> SEQUENCE: 42
    ggtagtttag gaatt 15
    <210> SEQ ID NO 43
    <211> LENGTH: 20
    <212> TYPE: DNA
    <213> ORGANISM: Artificial Sequence
    <220> FEATURE:
    <223> OTHER INFORMATION: control sequence
    <400> SEQUENCE: 43
    ttttaatcat attgttattc 20
    <210> SEQ ID NO 44
    <211> LENGTH: 20
    <212> TYPE: DNA
    <213> ORGANISM: Artificial Sequence
    <220> FEATURE:
    <223> OTHER INFORMATION: control sequence
    <220> FEATURE:
    <221> NAME/KEY: unsure
    <222> LOCATION: (1)..(20)
    <400> SEQUENCE: 44
    nnnnnnnnnn nnnnnnnnnn 20

Claims (44)

What is claimed is:
1. An antisense compound 8 to 30 nucleobases in length targeted to the 5′-untranslated region, translational termination region or 3′ untranslated region of a nucleic acid molecule encoding focal adhesion kinase, wherein said antisense compound inhibits the expression of said focal adhesion kinase.
2. The antisense compound of claim 1 which is an antisense oligonucleotide.
3. The antisense compound of claim 2 wherein the antisense oligonucleotide has a sequence comprising SEQ ID NO: 3, 4, 6, 7, 8, 9, 16, 17, 18, 20 or 23.
4. The antisense compound of claim 2 wherein the antisense oligonucleotide comprises at least one modified internucleoside linkage.
5. The antisense compound of claim 4 wherein the modified internucleoside linkage is a phosphorothioate linkage.
6. The antisense compound of claim 2 wherein the antisense oligonucleotide comprises at least one modified sugar moiety.
7. The antisense compound of claim 6 wherein the modified sugar moiety is a 2′-O-methoxyethyl moiety.
8. The antisense compound of claim 2 wherein the antisense oligonucleotide comprises at least one modified nucleobase.
9. The antisense compound of claim 8 wherein the modified nucleobase is a 5-methyl cytosine.
10. The antisense compound of claim 2 wherein the antisense oligonucleotide is a chimeric oligonucleotide.
11. A pharmaceutical composition comprising the antisense compound of claim 1 and a pharmaceutically acceptable carrier or diluent.
12. The pharmaceutical composition of claim 11 further comprising a colloidal dispersion system.
13. The pharmaceutical composition of claim 11 wherein the antisense compound is an antisense oligonucleotide.
14. The pharmaceutical composition of claim 11 further comprising a chemotherapeutic agent.
15. The pharmaceutical composition of claim 14 wherein the chemotherapeutic agent is 5-fluorouracil.
16. 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 claim 14.
17. A method of inhibiting the expression of focal adhesion kinase in cells or tissues comprising contacting said cells or tissue with the antisense compound of claim 1 so that expression of focal adhesion kinase is inhibited.
18. An antisense compound up to 30 nucleobases in length targeted to the coding region, or start site of a nucleic acid molecule encoding focal adhesion kinase, wherein said antisense compound inhibits the expression of said focal adhesion kinase and has a sequence comprising at least an 8 nucleobasic portion of SEQ ID NO: 10, 11, 12, 14, 15, 30, 31 or 33.
19. The antisense compound of claim 18 which is an antisense oligonucleotide.
20. The antisense compound of claim 19 wherein the antisense oligonucleotide comprises at least one modified internucleoside linkage.
21. The antisense compound of claim 20 wherein the modified internucleoside linkage is a phosphorothioate linkage.
22. The antisense compound of claim 19 wherein the antisense oligonucleotide comprises at least one modified sugar moiety.
23. The antisense compound of claim 22 wherein the modified sugar moiety is a 2′-O-methoxyethyl moiety.
24. The antisense compound of claim 19 wherein the antisense oligonucleotide comprises at least one modified nucleobase.
25. The antisense compound of claim 24 wherein the modified nucleobase is a 5-methyl cytosine.
26. The antisense compound of claim 19 wherein the antisense oligonucleotide is a chimeric oligonucleotide.
27. A pharmaceutical composition comprising the antisense compound of claim 18 and a pharmaceutically acceptable carrier or diluent.
28. The pharmaceutical composition of claim 27 further comprising a colloidal dispersion system.
29. The pharmaceutical composition of claim 27 wherein the antisense compound is an antisense oligonucleotide.
30. The pharmaceutical composition of claim 27 further comprising a chemotherapeutic agent.
31. The pharmaceutical composition of claim 30 wherein the chemotherapeutic agent is 5-fluorouracil.
32. 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 claim 30.
33. A method of inhibiting the expression of focal adhesion kinase in cells or tissues comprising contacting said cells or tissue with the antisense compound of claim 18 so that expression of focal adhesion kinase is inhibited.
34. A method of treating an animal having a disease or condition associated with focal adhesion kinase comprising administering to said animal a therapeutically or prophylactically effective amount of an antisense compound 8 to 30 nucleobases in length targeted to a nucleic acid molecule encoding human focal adhesion kinase wherein said antisense compound inhibits the expression of human focal adhesion kinase.
35. The method of claim 34 wherein the disease or condition is cancer.
36. The method of claim 35 wherein said cancer is of the breast, colon, mouth or skin.
37. The method of claim 34 wherein said disease or condition is an angiogenic disorder.
38. The method of claim 37 wherein said angiogenic disorder is retinal neovascularization.
39. A method of preventing migration of cells associated with expression of focal adhesion kinase comprising administering to said cells a therapeutically or prophylactically effective amount of an antisense compound 8 to 30 nucleobases in length targeted to a nucleic acid molecule encoding human focal adhesion kinase wherein said antisense compound inhibits the expression of human focal adhesion kinase.
40. A method of preventing neovascularization associated with expression of focal adhesion kinase in an animal comprising administering to said animal a therapeutically or prophylactically effective amount of an antisense compound 8 to 30 nucleobases in length targeted to a nucleic acid molecule encoding human focal adhesion kinase wherein said antisense compound inhibits the expression of human focal adhesion kinase.
41. A method of treating an animal having a disease or condition associated with focal adhesion kinase comprising administering to said animal a therapeutically or prophylactically effective amount of an antisense compound 8 to 30 nucleobases in length targeted to a nucleic acid molecule encoding human focal adhesion kinase in combination with a therapeutically or prophylactically effective amount of a chemotherapeutic agent.
42. The method of claim 41 wherein the chemotherapeutic agent is 5-fluorouracil.
43. The method of claim 41 wherein the disease or condition is cancer.
44. The method of claim 43 wherein said cancer is melanoma.
US10/646,569 1998-11-25 2003-08-22 Antisense modulation of focal adhesion kinase expression Abandoned US20040192628A1 (en)

Priority Applications (7)

Application Number Priority Date Filing Date Title
US09/966,451 US6692959B2 (en) 1998-11-25 2001-09-28 Antisense modulation of IL-1 receptor-associated kinase-4 expression
PCT/US2002/030574 WO2003028636A2 (en) 1998-11-25 2002-09-26 Antisense modulation of il-1 receptor-associated kinase-4 expression
EP02776002A EP1436308A4 (en) 1998-11-25 2002-09-26 Antisense modulation of il-1 receptor-associated kinase-4 expression
US10/630,399 US20040019009A1 (en) 1998-11-25 2003-07-30 Antisense modulation of IL-1 receptor-associated kinase-4 expression
US10/646,569 US20040192628A1 (en) 1999-08-19 2003-08-22 Antisense modulation of focal adhesion kinase expression
US11/117,013 US20050267063A1 (en) 1998-11-25 2005-04-27 Antisense modulation of p70 S6 kinase expression
US11/505,758 US20070049545A1 (en) 1998-11-25 2006-08-17 Antisense modulation of fibroblast growth factor receptor 3 expression

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US09/377,310 US6133031A (en) 1999-08-19 1999-08-19 Antisense inhibition of focal adhesion kinase expression
PCT/US2000/018999 WO2001014402A1 (en) 1999-08-19 2000-07-13 Antisense modulation of focal adhesion kinase expression
US09/757,100 US20010034329A1 (en) 1999-08-19 2001-01-09 Antisense modulation of focal adhesion kinase expression
US10/646,569 US20040192628A1 (en) 1999-08-19 2003-08-22 Antisense modulation of focal adhesion kinase expression

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US09/757,100 Continuation US20010034329A1 (en) 1998-11-25 2001-01-09 Antisense modulation of focal adhesion kinase expression

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US11/117,013 Continuation-In-Part US20050267063A1 (en) 1998-11-25 2005-04-27 Antisense modulation of p70 S6 kinase expression

Publications (1)

Publication Number Publication Date
US20040192628A1 true US20040192628A1 (en) 2004-09-30

Family

ID=23488586

Family Applications (3)

Application Number Title Priority Date Filing Date
US09/377,310 Expired - Lifetime US6133031A (en) 1998-11-25 1999-08-19 Antisense inhibition of focal adhesion kinase expression
US09/757,100 Abandoned US20010034329A1 (en) 1998-11-25 2001-01-09 Antisense modulation of focal adhesion kinase expression
US10/646,569 Abandoned US20040192628A1 (en) 1998-11-25 2003-08-22 Antisense modulation of focal adhesion kinase expression

Family Applications Before (2)

Application Number Title Priority Date Filing Date
US09/377,310 Expired - Lifetime US6133031A (en) 1998-11-25 1999-08-19 Antisense inhibition of focal adhesion kinase expression
US09/757,100 Abandoned US20010034329A1 (en) 1998-11-25 2001-01-09 Antisense modulation of focal adhesion kinase expression

Country Status (5)

Country Link
US (3) US6133031A (en)
EP (1) EP1208110A4 (en)
JP (1) JP2003507051A (en)
AU (1) AU6091200A (en)
WO (1) WO2001014402A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2016061263A1 (en) * 2014-10-14 2016-04-21 Ionis Pharmaceuticals, Inc. Antisense compounds and uses thereof

Families Citing this family (165)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1998029138A2 (en) 1996-12-27 1998-07-09 Instituto De Investigaciones Bioquimicas Fundacion Campomar Compositions and methods for tumour therapy
CA2294988C (en) 1997-07-01 2015-11-24 Isis Pharmaceuticals Inc. Compositions and methods for the delivery of oligonucleotides via the alimentary canal
US20030165888A1 (en) * 2001-07-18 2003-09-04 Brown Bob D. Oligonucleotide probes and primers comprising universal bases for diagnostic purposes
US6518017B1 (en) * 1997-10-02 2003-02-11 Oasis Biosciences Incorporated Combinatorial antisense library
WO1999060012A1 (en) * 1998-05-21 1999-11-25 Isis Pharmaceuticals, Inc. Compositions and methods for non-parenteral delivery of oligonucleotides
CA2365984A1 (en) * 1999-04-08 2000-10-19 Oasis Biosciences, Inc. Antisense oligonucleotides comprising universal and/or degenerate bases
EP1191097A1 (en) * 2000-09-21 2002-03-27 Leids Universitair Medisch Centrum Induction of exon skipping in eukaryotic cells
EP1546372A4 (en) * 2002-09-19 2006-08-23 Pfizer Prod Inc Inducible focal adhesion kinase cell assay
AU2003225410A1 (en) 2003-03-21 2004-10-11 Academisch Ziekenhuis Leiden Modulation of exon recognition in pre-mrna by interfering with the secondary rna structure
US7244829B2 (en) * 2004-11-08 2007-07-17 Gem Pharmaceuticals, Llc Compositions and processes for preparing 13-deoxy-anthracyclines
WO2006113679A2 (en) * 2005-04-15 2006-10-26 Board Of Regents, The University Of Texas System Delivery of sirna by neutral lipid compositions
JP2008538500A (en) * 2005-04-22 2008-10-30 アカデミス ツィーケンホイス ライデン Regulation of exon recognition in pre-mRNA by interference with SR protein binding and RNA secondary structure
DK2559690T3 (en) 2005-05-10 2016-04-25 Incyte Holdings Corp Modulators of indoleamine 2,3-dioxygenase and methods of use thereof
CA2621261C (en) 2005-09-22 2014-05-20 Incyte Corporation Azepine inhibitors of janus kinases
PT3184526T (en) 2005-12-13 2018-12-19 Incyte Holdings Corp Pyrrolo[2,3-d]pyrimidine derivatives as janus kinase inhibitor
WO2007123391A1 (en) * 2006-04-20 2007-11-01 Academisch Ziekenhuis Leiden Therapeutic intervention in a genetic disease in an individual by modifying expression of an aberrantly expressed gene.
EP1857548A1 (en) * 2006-05-19 2007-11-21 Academisch Ziekenhuis Leiden Means and method for inducing exon-skipping
PT2049664E (en) 2006-08-11 2012-01-03 Prosensa Technologies Bv Single stranded oligonucleotides complementary to repetitive elements for treating dna repeat instability associated genetic disorders
EP2121692B1 (en) 2006-12-22 2013-04-10 Incyte Corporation Substituted heterocycles as janus kinase inhibitors
CL2008001709A1 (en) 2007-06-13 2008-11-03 Incyte Corp Compounds derived from pyrrolo [2,3-b] pyrimidine, jak kinase modulators; pharmaceutical composition; and use in the treatment of diseases such as cancer, psoriasis, rheumatoid arthritis, among others.
US20080312259A1 (en) 2007-06-13 2008-12-18 Incyte Corporation SALTS OF THE JANUS KINASE INHIBITOR (R)-3-(4-(7H-PYRROLO[2,3-d]PYRIMIDIN-4-YL)-1H-PYRAZOL-1-YL)-3-CYCLOPENTYLPROPANENITRILE
WO2009008725A2 (en) * 2007-07-12 2009-01-15 Prosensa Technologies B.V. Molecules for targeting compounds to various selected organs, tissues or tumor cells
CA2693048C (en) * 2007-07-12 2016-10-18 Prosensa Technologies B.V. Molecules for targeting compounds to various selected organs or tissues
CA2704049A1 (en) 2007-10-26 2009-04-30 Academisch Ziekenhuis Leiden Means and methods for counteracting muscle disorders
USRE48468E1 (en) 2007-10-26 2021-03-16 Biomarin Technologies B.V. Means and methods for counteracting muscle disorders
EA020777B1 (en) 2007-11-16 2015-01-30 Инсайт Корпорейшн 4-pyrazolyl-n-arylpyrimidin-2-amines, 4-pyrazolyl-n-pyrazolylpyrimidin-2-amines and 4-pyrazolyl-n-pyridylpyrimidin-2-amines as janus kinase inhibitors
NZ587178A (en) * 2008-02-08 2011-11-25 Prosensa Holding Bv Methods and means for treating dna repeat instability associated genetic disorders
TWI444382B (en) 2008-03-11 2014-07-11 Incyte Corp Azetidine and cyclobutane derivatives as jak inhibitors
JP2011518836A (en) 2008-04-24 2011-06-30 インサイト・コーポレイション Macrocycles and their use as kinase inhibitors
EP2119783A1 (en) 2008-05-14 2009-11-18 Prosensa Technologies B.V. Method for efficient exon (44) skipping in Duchenne Muscular Dystrophy and associated means
BRPI0915692B8 (en) 2008-07-08 2021-05-25 Incyte Corp compounds derived from 1,2,5-oxadiazoles, their solid form, their composition, as well as their uses
CL2009001884A1 (en) 2008-10-02 2010-05-14 Incyte Holdings Corp Use of 3-cyclopentyl-3- [4- (7h-pyrrolo [2,3-d] pyrimidin-4-yl) -1h-pyrazol-1-yl) propanonitrile, janus kinase inhibitor, and use of a composition that understands it for the treatment of dry eye.
WO2010085597A1 (en) 2009-01-23 2010-07-29 Incyte Corporation Macrocyclic compounds and their use as kinase inhibitors
AU2010239779A1 (en) 2009-04-24 2011-11-17 Prosensa Technologies B.V. Oligonucleotide comprising an inosine for treating DMD
CN106967070A (en) 2009-05-22 2017-07-21 因塞特控股公司 It is used as the compound of JAK inhibitor
ES2487542T3 (en) 2009-05-22 2014-08-21 Incyte Corporation N- (hetero) aryl-pyrrolidine derivatives of pyrazol-4-yl-pyrrolo [2,3-d] pyrimidines and pyrrol-3-yl-pyrrolo [2,3-d] pyrimidines as Janus kinase inhibitors
KR101763656B1 (en) 2009-06-29 2017-08-01 인사이트 홀딩스 코포레이션 Pyrimidinones as pi3k inhibitors
AR078012A1 (en) 2009-09-01 2011-10-05 Incyte Corp HETEROCICLIC DERIVATIVES OF PIRAZOL-4-IL-PIRROLO (2,3-D) PYRIMIDINS AS INHIBITORS OF THE QUANASA JANUS
BR112012008267B1 (en) 2009-10-09 2022-10-04 Incyte Holdings Corporation 3-(4-(7H-PYROLO[2,3-D]PYRIMIDIN-4-IL)-1H-PYRAZOL-1-IL)-3-CYCLOPENTYLPROPANONITRILE 3-(4-(7H-PYROXYL, KETO, AND GLUCURONIDE) DERIVATIVES
TW201130842A (en) 2009-12-18 2011-09-16 Incyte Corp Substituted fused aryl and heteroaryl derivatives as PI3K inhibitors
WO2011075643A1 (en) 2009-12-18 2011-06-23 Incyte Corporation Substituted heteroaryl fused derivatives as pi3k inhibitors
JP6141018B2 (en) 2009-12-24 2017-06-07 バイオマリン テクノロジーズ ベー.フェー. Molecules for treating inflammatory disorders
CA2790070C (en) 2010-02-18 2018-03-06 Incyte Corporation Cyclobutane and methylcyclobutane derivatives as janus kinase inhibitors
TWI766281B (en) 2010-03-10 2022-06-01 美商英塞特控股公司 Piperidin-4-yl azetidine derivatives as jak1 inhibitors
JP5816678B2 (en) 2010-04-14 2015-11-18 インサイト・コーポレイションIncyte Corporation Condensed derivatives as PI3Kδ inhibitors
PE20130216A1 (en) 2010-05-21 2013-02-27 Incyte Corp TOPICAL FORMULATION FOR A JAK INHIBITOR
WO2011163195A1 (en) 2010-06-21 2011-12-29 Incyte Corporation Fused pyrrole derivatives as pi3k inhibitors
JP5917544B2 (en) 2010-11-19 2016-05-18 インサイト・ホールディングス・コーポレイションIncyte Holdings Corporation Heterocyclic substituted pyrrolopyridines and pyrrolopyrimidines as JAK inhibitors
AR083933A1 (en) 2010-11-19 2013-04-10 Incyte Corp PIRROLOPIRIDINE AND PIRROLOPIRIMIDINE DERIVATIVES REPLACED WITH CYCLOBUTILO AS JAK INHIBITORS
JP5961187B2 (en) 2010-12-20 2016-08-02 インサイト・ホールディングス・コーポレイションIncyte Holdings Corporation N- (1- (substituted phenyl) ethyl) -9H-purin-6-amine as a PI3K inhibitor
US9993480B2 (en) 2011-02-18 2018-06-12 Novartis Pharma Ag mTOR/JAK inhibitor combination therapy
WO2012125629A1 (en) 2011-03-14 2012-09-20 Incyte Corporation Substituted diamino-pyrimidine and diamino-pyridine derivatives as pi3k inhibitors
US9126948B2 (en) 2011-03-25 2015-09-08 Incyte Holdings Corporation Pyrimidine-4,6-diamine derivatives as PI3K inhibitors
EA201490042A1 (en) 2011-06-20 2014-10-30 Инсайт Корпорейшн AZETIDINYL-PHENYL-, PYRIDYL- OR PYRAZYNYLKARBOXAMID DERIVATIVES AS INHIBITORS
WO2013023119A1 (en) 2011-08-10 2013-02-14 Novartis Pharma Ag JAK P13K/mTOR COMBINATION THERAPY
TW201313721A (en) 2011-08-18 2013-04-01 Incyte Corp Cyclohexyl azetidine derivatives as JAK inhibitors
WO2013033569A1 (en) 2011-09-02 2013-03-07 Incyte Corporation Heterocyclylamines as pi3k inhibitors
UA111854C2 (en) 2011-09-07 2016-06-24 Інсайт Холдінгс Корпорейшн METHODS AND INTERMEDIATE COMPOUNDS FOR JAK INHIBITORS
CA2862628C (en) 2012-01-27 2021-08-24 Prosensa Technologies B.V. Rna modulating oligonucleotides with improved characteristics for the treatment of duchenne and becker muscular dystrophy
AR090548A1 (en) 2012-04-02 2014-11-19 Incyte Corp BICYCLIC AZAHETEROCICLOBENCILAMINS AS PI3K INHIBITORS
AR091079A1 (en) 2012-05-18 2014-12-30 Incyte Corp DERIVATIVES OF PIRROLOPIRIMIDINA AND PIRROLOPIRIDINA REPLACED WITH PIPERIDINILCICLOBUTILO AS JAK INHIBITORS
PT3495367T (en) 2012-06-13 2020-11-12 Incyte Holdings Corp Substituted tricyclic compounds as fgfr inhibitors
CN107936039A (en) 2012-11-01 2018-04-20 因赛特公司 Tricyclic condensed thiophene derivant as JAK inhibitor
SG11201503695XA (en) 2012-11-15 2015-06-29 Incyte Corp Sustained-release dosage forms of ruxolitinib
TWI687220B (en) 2013-03-01 2020-03-11 美商英塞特控股公司 Use of pyrazolopyrimidine derivatives for the treatment of pi3kδ related disorders
HUE057262T2 (en) 2013-03-06 2022-04-28 Incyte Holdings Corp Processes and intermediates for making a jak inhibitor
TWI715901B (en) 2013-04-19 2021-01-11 美商英塞特控股公司 Bicyclic heterocycles as fgfr inhibitors
PL3786162T3 (en) 2013-05-17 2024-04-08 Incyte Holdings Corporation Bipyrazole derivatives as jak inhibitors
US9655854B2 (en) 2013-08-07 2017-05-23 Incyte Corporation Sustained release dosage forms for a JAK1 inhibitor
US20150065447A1 (en) 2013-08-20 2015-03-05 Incyte Corporation Survival benefit in patients with solid tumors with elevated c-reactive protein levels
PE20161388A1 (en) 2014-02-28 2016-12-28 Incyte Corp JAK1 INHIBITORS FOR THE TREATMENT OF MYELODYSPLASIC SYNDROMES
CN106413716B (en) 2014-04-08 2020-03-27 因赛特公司 Treatment of B cell malignancies by combination of JAK and PI3K inhibitors
MA39987A (en) 2014-04-30 2017-03-08 Incyte Corp Processes of preparing a jak1 inhibitor and new forms thereto
US9498467B2 (en) 2014-05-30 2016-11-22 Incyte Corporation Treatment of chronic neutrophilic leukemia (CNL) and atypical chronic myeloid leukemia (aCML) by inhibitors of JAK1
WO2015191677A1 (en) 2014-06-11 2015-12-17 Incyte Corporation Bicyclic heteroarylaminoalkyl phenyl derivatives as pi3k inhibitors
US9586949B2 (en) 2015-02-09 2017-03-07 Incyte Corporation Aza-heteroaryl compounds as PI3K-gamma inhibitors
MA41551A (en) 2015-02-20 2017-12-26 Incyte Corp BICYCLIC HETEROCYCLES USED AS FGFR4 INHIBITORS
CN113004278B (en) 2015-02-20 2023-07-21 因赛特控股公司 Bicyclic heterocycles as FGFR inhibitors
NZ734993A (en) 2015-02-27 2024-03-22 Incyte Holdings Corp Salts of pi3k inhibitor and processes for their preparation
US9732097B2 (en) 2015-05-11 2017-08-15 Incyte Corporation Process for the synthesis of a phosphoinositide 3-kinase inhibitor
WO2016183062A1 (en) 2015-05-11 2016-11-17 Incyte Corporation Salts of (s)-7-(1-(9h-purin-6-ylamino)ethyl)-6-(3-fluorophenyl)-3-methyl-5h-thiazolo[3,2-a]pyrimidin-5-one
US9988401B2 (en) 2015-05-11 2018-06-05 Incyte Corporation Crystalline forms of a PI3K inhibitor
AR106595A1 (en) 2015-11-06 2018-01-31 Incyte Corp HETEROCYCLIC COMPOUNDS AS PI3K-g INHIBITORS
US20170190689A1 (en) 2016-01-05 2017-07-06 Incyte Corporation Pyridine and pyridimine compounds as pi3k-gamma inhibitors
US10138248B2 (en) 2016-06-24 2018-11-27 Incyte Corporation Substituted imidazo[2,1-f][1,2,4]triazines, substituted imidazo[1,2-a]pyridines, substituted imidazo[1,2-b]pyridazines and substituted imidazo[1,2-a]pyrazines as PI3K-γ inhibitors
AR111960A1 (en) 2017-05-26 2019-09-04 Incyte Corp CRYSTALLINE FORMS OF A FGFR INHIBITOR AND PROCESSES FOR ITS PREPARATION
US11725056B2 (en) 2017-10-03 2023-08-15 Cedars-Sinai Medical Center Methods for targeting the immune checkpoint PD1 pathway for treating pulmonary fibrosis
AU2018350980B2 (en) 2017-10-18 2023-02-23 Incyte Corporation Condensed imidazole derivatives substituted by tertiary hydroxy groups as pi3k-y gamma inhibitors
AR113922A1 (en) 2017-12-08 2020-07-01 Incyte Corp LOW DOSE COMBINATION THERAPY FOR THE TREATMENT OF MYELOPROLIFERATIVE NEOPLASMS
US11306079B2 (en) 2017-12-21 2022-04-19 Incyte Corporation 3-(5-amino-pyrazin-2-yl)-benzenesulfonamide derivatives and related compounds as PI3K-gamma kinase inhibitors
CN112105608B (en) 2018-01-30 2023-07-14 因赛特公司 Process for the preparation of (1- (3-fluoro-2- (trifluoromethyl) isonicotinyl) piperidin-4-one
EP3752146A1 (en) 2018-02-16 2020-12-23 Incyte Corporation Jak1 pathway inhibitors for the treatment of cytokine-related disorders
CN112384515A (en) 2018-02-27 2021-02-19 因赛特公司 Imidazopyrimidines and triazolopyrimidines as A2A/A2B inhibitors
HUE057970T2 (en) 2018-03-08 2022-06-28 Incyte Corp Aminopyrazine diol compounds as pi3k-y inhibitors
AU2019245420A1 (en) 2018-03-30 2020-11-12 Incyte Corporation Treatment of hidradenitis suppurativa using JAK inhibitors
US11220510B2 (en) 2018-04-09 2022-01-11 Incyte Corporation Pyrrole tricyclic compounds as A2A / A2B inhibitors
WO2019213544A2 (en) 2018-05-04 2019-11-07 Incyte Corporation Solid forms of an fgfr inhibitor and processes for preparing the same
CR20200591A (en) 2018-05-04 2021-03-31 Incyte Corp Salts of an fgfr inhibitor
US11168089B2 (en) 2018-05-18 2021-11-09 Incyte Corporation Fused pyrimidine derivatives as A2A / A2B inhibitors
JP2021525706A (en) 2018-05-25 2021-09-27 インサイト・コーポレイションIncyte Corporation Tricyclic heterocyclic compound as a STING activator
US11046658B2 (en) 2018-07-02 2021-06-29 Incyte Corporation Aminopyrazine derivatives as PI3K-γ inhibitors
GEP20237548B (en) 2018-07-05 2023-10-10 Incyte Corp Fused pyrazine derivatives as a2a /a2b inhibitors
WO2020028565A1 (en) 2018-07-31 2020-02-06 Incyte Corporation Tricyclic heteraryl compounds as sting activators
US10875872B2 (en) 2018-07-31 2020-12-29 Incyte Corporation Heteroaryl amide compounds as sting activators
LT3847175T (en) 2018-09-05 2024-03-25 Incyte Corporation Crystalline forms of a phosphoinositide 3-kinase (pi3k) inhibitor
US11066404B2 (en) 2018-10-11 2021-07-20 Incyte Corporation Dihydropyrido[2,3-d]pyrimidinone compounds as CDK2 inhibitors
WO2020102198A1 (en) 2018-11-13 2020-05-22 Incyte Corporation Heterocyclic derivatives as pi3k inhibitors
US11161838B2 (en) 2018-11-13 2021-11-02 Incyte Corporation Heterocyclic derivatives as PI3K inhibitors
US11396502B2 (en) 2018-11-13 2022-07-26 Incyte Corporation Substituted heterocyclic derivatives as PI3K inhibitors
US11596692B1 (en) 2018-11-21 2023-03-07 Incyte Corporation PD-L1/STING conjugates and methods of use
US20220056049A1 (en) 2019-01-07 2022-02-24 Incyte Corporation Heteroaryl amide compounds as sting activators
TWI829857B (en) 2019-01-29 2024-01-21 美商英塞特公司 Pyrazolopyridines and triazolopyridines as a2a / a2b inhibitors
US11384083B2 (en) 2019-02-15 2022-07-12 Incyte Corporation Substituted spiro[cyclopropane-1,5′-pyrrolo[2,3-d]pyrimidin]-6′(7′h)-ones as CDK2 inhibitors
US20200316064A1 (en) 2019-02-15 2020-10-08 Incyte Corporation Cyclin-dependent kinase 2 biomarkers and uses thereof
WO2020180959A1 (en) 2019-03-05 2020-09-10 Incyte Corporation Pyrazolyl pyrimidinylamine compounds as cdk2 inhibitors
WO2020185532A1 (en) 2019-03-08 2020-09-17 Incyte Corporation Methods of treating cancer with an fgfr inhibitor
WO2020205560A1 (en) 2019-03-29 2020-10-08 Incyte Corporation Sulfonylamide compounds as cdk2 inhibitors
US11447494B2 (en) 2019-05-01 2022-09-20 Incyte Corporation Tricyclic amine compounds as CDK2 inhibitors
WO2020223469A1 (en) 2019-05-01 2020-11-05 Incyte Corporation N-(1-(methylsulfonyl)piperidin-4-yl)-4,5-di hydro-1h-imidazo[4,5-h]quinazolin-8-amine derivatives and related compounds as cyclin-dependent kinase 2 (cdk2) inhibitors for treating cancer
US20200405627A1 (en) 2019-06-10 2020-12-31 Incyte Corporation Topical treatment of vitiligo by a jak inhibitor
WO2021007269A1 (en) 2019-07-09 2021-01-14 Incyte Corporation Bicyclic heterocycles as fgfr inhibitors
EP4007602A1 (en) 2019-08-01 2022-06-08 Incyte Corporation A dosing regimen for an ido inhibitor
AR119765A1 (en) 2019-08-14 2022-01-12 Incyte Corp IMIDAZOLIL PYRIMIDINILAMINE COMPOUNDS AS CDK2 INHIBITORS
MX2022002219A (en) 2019-08-26 2022-06-14 Incyte Corp Triazolopyrimidines as a2a / a2b inhibitors.
MX2022004390A (en) 2019-10-11 2022-08-08 Incyte Corp Bicyclic amines as cdk2 inhibitors.
TW202128685A (en) 2019-10-14 2021-08-01 美商英塞特公司 Bicyclic heterocycles as fgfr inhibitors
JP2023506118A (en) 2019-10-16 2023-02-15 インサイト・コーポレイション Use of JAK1 inhibitors for the treatment of cutaneous lupus erythematosus and lichen planus (LP)
US11566028B2 (en) 2019-10-16 2023-01-31 Incyte Corporation Bicyclic heterocycles as FGFR inhibitors
US11992490B2 (en) 2019-10-16 2024-05-28 Incyte Corporation Use of JAK1 inhibitors for the treatment of cutaneous lupus erythematosus and Lichen planus (LP)
CA3162010A1 (en) 2019-12-04 2021-06-10 Incyte Corporation Derivatives of an fgfr inhibitor
EP4069696A1 (en) 2019-12-04 2022-10-12 Incyte Corporation Tricyclic heterocycles as fgfr inhibitors
US12012409B2 (en) 2020-01-15 2024-06-18 Incyte Corporation Bicyclic heterocycles as FGFR inhibitors
WO2021178779A1 (en) 2020-03-06 2021-09-10 Incyte Corporation Combination therapy comprising axl/mer and pd-1/pd-l1 inhibitors
CR20220584A (en) 2020-04-16 2023-02-15 Incyte Corp Fused tricyclic kras inhibitors
US11739102B2 (en) 2020-05-13 2023-08-29 Incyte Corporation Fused pyrimidine compounds as KRAS inhibitors
PE20231102A1 (en) 2020-06-02 2023-07-19 Incyte Corp PROCESSES FOR PREPARING A JAK1 INHIBITOR
US11833155B2 (en) 2020-06-03 2023-12-05 Incyte Corporation Combination therapy for treatment of myeloproliferative neoplasms
WO2022047093A1 (en) 2020-08-28 2022-03-03 Incyte Corporation Vinyl imidazole compounds as inhibitors of kras
CN116261447A (en) 2020-09-16 2023-06-13 因赛特公司 Topical treatment of vitiligo
US11767320B2 (en) 2020-10-02 2023-09-26 Incyte Corporation Bicyclic dione compounds as inhibitors of KRAS
MX2023006542A (en) 2020-12-08 2023-08-25 Incyte Corp Jak1 pathway inhibitors for the treatment of vitiligo.
WO2022155941A1 (en) 2021-01-25 2022-07-28 Qilu Regor Therapeutics Inc. Cdk2 inhibitors
WO2022206888A1 (en) 2021-03-31 2022-10-06 Qilu Regor Therapeutics Inc. Cdk2 inhibitors and use thereof
JP2024513575A (en) 2021-04-12 2024-03-26 インサイト・コーポレイション Combination therapy including FGFR inhibitor and Nectin-4 targeting agent
AR126101A1 (en) 2021-06-09 2023-09-13 Incyte Corp TRICYCLIC HETEROCYCLES AS FGFR INHIBITORS
CA3220274A1 (en) 2021-06-09 2022-12-15 Incyte Corporation Tricyclic heterocycles as fgfr inhibitors
US11981671B2 (en) 2021-06-21 2024-05-14 Incyte Corporation Bicyclic pyrazolyl amines as CDK2 inhibitors
KR20240032915A (en) 2021-07-07 2024-03-12 인사이트 코포레이션 Tricyclic compounds as inhibitors of KRAS
US20230114765A1 (en) 2021-07-14 2023-04-13 Incyte Corporation Tricyclic compounds as inhibitors of kras
US20230174555A1 (en) 2021-08-31 2023-06-08 Incyte Corporation Naphthyridine compounds as inhibitors of kras
WO2023049697A1 (en) 2021-09-21 2023-03-30 Incyte Corporation Hetero-tricyclic compounds as inhibitors of kras
CA3234375A1 (en) 2021-10-01 2023-04-06 Incyte Corporation Pyrazoloquinoline kras inhibitors
WO2023064857A1 (en) 2021-10-14 2023-04-20 Incyte Corporation Quinoline compounds as inhibitors of kras
AU2022389961A1 (en) 2021-11-22 2024-06-06 Incyte Corporation Combination therapy comprising an fgfr inhibitor and a kras inhibitor
TW202329937A (en) 2021-12-03 2023-08-01 美商英塞特公司 Bicyclic amine cdk12 inhibitors
US11976073B2 (en) 2021-12-10 2024-05-07 Incyte Corporation Bicyclic amines as CDK2 inhibitors
WO2023107705A1 (en) 2021-12-10 2023-06-15 Incyte Corporation Bicyclic amines as cdk12 inhibitors
US20230192722A1 (en) 2021-12-22 2023-06-22 Incyte Corporation Salts and solid forms of an fgfr inhibitor and processes of preparing thereof
WO2023168686A1 (en) 2022-03-11 2023-09-14 Qilu Regor Therapeutics Inc. Substituted cyclopentanes as cdk2 inhibitors
WO2023116884A1 (en) 2021-12-24 2023-06-29 Qilu Regor Therapeutics Inc. Cdk2 inhibitors and use thereof
US20230279004A1 (en) 2022-03-07 2023-09-07 Incyte Corporation Solid forms, salts, and processes of preparation of a cdk2 inhibitor
US20230399331A1 (en) 2022-06-14 2023-12-14 Incyte Corporation Solid forms of jak inhibitor and process of preparing the same
WO2023250430A1 (en) 2022-06-22 2023-12-28 Incyte Corporation Bicyclic amine cdk12 inhibitors
WO2024015731A1 (en) 2022-07-11 2024-01-18 Incyte Corporation Fused tricyclic compounds as inhibitors of kras g12v mutants
US20240058343A1 (en) 2022-08-05 2024-02-22 Incyte Corporation Treatment of urticaria using jak inhibitors

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6015893A (en) * 1994-07-18 2000-01-18 The University Of North Carolina At Chapel Hill Oligonucleoside compounds and methods for inhibiting tumor growth, invasion and metastasis

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6015893A (en) * 1994-07-18 2000-01-18 The University Of North Carolina At Chapel Hill Oligonucleoside compounds and methods for inhibiting tumor growth, invasion and metastasis

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2016061263A1 (en) * 2014-10-14 2016-04-21 Ionis Pharmaceuticals, Inc. Antisense compounds and uses thereof

Also Published As

Publication number Publication date
US20010034329A1 (en) 2001-10-25
AU6091200A (en) 2001-03-19
US6133031A (en) 2000-10-17
WO2001014402A8 (en) 2001-06-28
EP1208110A4 (en) 2004-12-22
EP1208110A1 (en) 2002-05-29
JP2003507051A (en) 2003-02-25
WO2001014402A1 (en) 2001-03-01

Similar Documents

Publication Publication Date Title
US20040192628A1 (en) Antisense modulation of focal adhesion kinase expression
US6159694A (en) Antisense modulation of stat3 expression
US6613567B1 (en) Antisense inhibition of Her-2 expression
US6410323B1 (en) Antisense modulation of human Rho family gene expression
US6656688B1 (en) Antisense modulation of NF-kappa-B p65 subunit expression
US6838283B2 (en) Antisense modulation of survivin expression
US6077709A (en) Antisense modulation of Survivin expression
US6180403B1 (en) Antisense inhibition of tumor necrosis factor alpha converting enzyme (TACE) expression
EP1137658A1 (en) METHODS OF MODULATING TUMOR NECROSIS FACTOR $g(a)-INDUCED EXPRESSION OF CELL ADHESION MOLECULES
US6204055B1 (en) Antisense inhibition of Fas mediated signaling
US5959097A (en) Antisense modulation of MEK2 expression
US6200807B1 (en) Antisense inhibition of SHP-2 expression
US20030092654A1 (en) Antisense modulation of Inhibitor-kappa B Kinase-alpha expression
US6087489A (en) Antisense oligonucleotide modulation of human thymidylate synthase expression
US6440738B1 (en) Antisense modulation of casein kinase 2-beta expression
US6235723B1 (en) Antisense oligonucleotide modulation of human protein kinase C-δ expression
US6168950B1 (en) Antisense modulation of MEKK1 expression
US6077672A (en) Antisense modulation of TRADD expression
US20020004490A1 (en) Antisense modulation of Fas mediated signaling
US6300320B1 (en) Modulation of c-jun using inhibitors of protein kinase C
US6607915B1 (en) Antisense inhibition of E2A-Pbx1 expression
US6632667B1 (en) Modulation of L-selectin shedding via inhibition of tumor necrosis factor-α converting enzyme (TACE)
US6309882B1 (en) Antisense inhibition of replication protein a p70 subunit
US20030050270A1 (en) Antisense modulation of Inhibitor-kappa B Kinase-beta expression
US6368856B1 (en) Antisense inhibition of Phosphorylase kinase beta expression

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION