WO1995032987A1 - ANTISENSE OLIGONUCLEOTIDE MODULATION OF raf GENE EXPRESSION - Google Patents

ANTISENSE OLIGONUCLEOTIDE MODULATION OF raf GENE EXPRESSION Download PDF

Info

Publication number
WO1995032987A1
WO1995032987A1 PCT/US1995/007111 US9507111W WO9532987A1 WO 1995032987 A1 WO1995032987 A1 WO 1995032987A1 US 9507111 W US9507111 W US 9507111W WO 9532987 A1 WO9532987 A1 WO 9532987A1
Authority
WO
WIPO (PCT)
Prior art keywords
oligonucleotide
raf
seq
targeted
oligonucleotides
Prior art date
Application number
PCT/US1995/007111
Other languages
French (fr)
Inventor
Brett P. Monia
Russell T. Boggs
Original Assignee
Isis Pharmaceuticals, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority to DE69533697T priority Critical patent/DE69533697T2/en
Priority to AT95922217T priority patent/ATE280772T1/en
Priority to CA002191795A priority patent/CA2191795C/en
Priority to EP95922217A priority patent/EP0763052B1/en
Priority to JP50125796A priority patent/JP3231779B2/en
Priority to AU26982/95A priority patent/AU689267B2/en
Priority to KR1019960706806A priority patent/KR100211178B1/en
Application filed by Isis Pharmaceuticals, Inc. filed Critical Isis Pharmaceuticals, Inc.
Publication of WO1995032987A1 publication Critical patent/WO1995032987A1/en
Priority to US08/756,806 priority patent/US5952229A/en
Priority to NO965080A priority patent/NO965080L/en
Priority to FI964792A priority patent/FI964792A/en
Priority to US09/462,261 priority patent/US6391636B1/en
Priority to US09/143,214 priority patent/US6090626A/en
Priority to US09/506,073 priority patent/US6410518B1/en
Priority to US10/057,550 priority patent/US6806258B2/en
Priority to US10/173,225 priority patent/US20030119769A1/en
Priority to US11/364,481 priority patent/US20060142236A1/en
Priority to US13/185,406 priority patent/US20120149755A1/en
Priority to US13/678,432 priority patent/US20130079387A1/en
Priority to US13/956,213 priority patent/US20140187601A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H21/00Compounds containing two or more mononucleotide units having separate phosphate or polyphosphate groups linked by saccharide radicals of nucleoside groups, e.g. nucleic acids
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • 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
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • 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
    • A61P35/00Antineoplastic agents
    • A61P35/04Antineoplastic agents specific for metastasis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • C12N15/1135Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing against oncogenes or tumor suppressor genes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/31Chemical structure of the backbone
    • C12N2310/315Phosphorothioates
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/32Chemical structure of the sugar
    • C12N2310/3212'-O-R Modification
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/32Chemical structure of the sugar
    • C12N2310/3222'-R Modification
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/33Chemical structure of the base
    • 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
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/35Nature of the modification
    • C12N2310/351Conjugate
    • C12N2310/3517Marker; Tag

Definitions

  • This invention relates to compositions and methods for modulating expression of the raf gene, a naturally present cellular gene which has been implicated in abnormal cell proliferation and tumor formation. This invention is also directed to methods for inhibiting hyperproliferation of cells; these methods can be used diagnostically or therapeutically. Furthermore, this invention is directed to treatment of conditions associated with expression of the raf gene.
  • the raf gene family includes three highly conserved genes termed A- , B- and c-raf (also called raf-1) .
  • Raf genes encode protein kinases that are thought to play important regulatory roles in signal transduction processes that regulate cell proliferation. Expression of the c-raf protein is believed to play a role in abnormal cell proliferation since it has been reported that 60% of all lung carcinoma cell lines express unusually high levels of c-raf mRNA and protein. Rapp et al . , The Oncogene Handbook, E.P. Reddy, A.M Skalka and T. Curran, eds .
  • Oligonucleotides have been employed as therapeutic moieties in the treatment of disease states in animals and man.
  • workers in the field have now identified antisense, triplex and other oligonucleotide compositions which are capable of modulating expression of genes implicated in viral, fungal and metabolic diseases.
  • U. S. Patent 5,135,917 provides antisense oligonucleotides that inhibit human interleukin-1 receptor expression.
  • U.S. Patent 5,098,890 issued March 24, 1992 in the name of Gewirtz et al . , is directed to antisense oligonucleotides complementary to the c- myb oncogene and antisense oligonucleotide therapies for certain cancerous conditions.
  • U.S. Patent 5,087,617, issued February 11, 1992 provides methods for treating cancer patients with antisense oligonucleotides.
  • U.S. Patent 5,166,195 issued November 24, 1992 provides oligonucleotide inhibitors of HIV.
  • Patent 5,004,810 issued April 2, 1991, provides oligomers capable of hybridizing to herpes simplex virus Vmw65 mRNA and inhibiting replication.
  • U.S. Patent 5,194,428, issued March 16, 1993 provides antisense oligonucleotides having antiviral activity against influenzavirus .
  • U.S. Patent 4,806,463, issued February 21, 1989 provides antisense oligonucleotides and methods using them to inhibit HTLV-III replication.
  • U.S. Patent 5,286,717 (Cohen et al . ) , issued February 15, 1994, is directed to a mixed linkage oligonucleotide phosphorothioates complementary to an oncogene;
  • Patent 5,264,423 (Cohen et al . ) are directed to phosphorothioate oligonucleotide analogs used to prevent replication of foreign nucleic acids in cells.
  • Antisense oligonucleotides have been safely administered to humans and clinical trials of several antisense oligonucleotide drugs, targeted both to viral and cellular gene products, are presently underway.
  • the phosphorothioate oligonucleotide, ISIS 2922 has been shown to be effective against cvtomegalovirus retinitis in AIDS patients. BioWorld Today, April 29, 1994, p. 3. It is thus established that oligonucleotides can be useful therapeutic instrumentalities and can be configured to be useful in treatment regimes for treatment of cells and animal subjects, especially humans.
  • Antisense oligonucleotide inhibition of gene expression has proven to be a useful tool in understanding the roles of raf genes.
  • An antisense oligonucleotide complementary to the first six codons of human c-raf has been used to demonstrate that the mitogenic response of T cells to interleukin-2 (I -2) requires c-raf.
  • Cells treated with the oligonucleotide showed a near-total loss of c-raf protein and a substantial reduction in proliferative response to IL-2.
  • WO application 93/04170 An antisense oligodeoxyribonucleotide complementary to codons 1-6 of murine c-Raf has been used to abolish insulin stimulation of DNA synthesis in the rat hepatoma cell line H4IIE. Tornkvist et al . , J. Biol . Chem . 1994, 269, 13919- 13921.
  • WO Application 93/06248 discloses methods for identifying an individual at increased risk of developing cancer and for determining a prognosis and proper treatment of patients afflicted with cancer comprising amplifying a region of the c-raf gene and analyzing it for evidence of mutation.
  • Denner et al disclose antisense polynucleotides hybridizing to the gene for raf, and processes using them.
  • WO 94/15645 Oligonucleotides hybridizing to human and rat raf sequences are disclosed.
  • Iversen et al disclose heterotypic antisense oligonucleotides complementary to raf which are able to kill ras-activated cancer cells, and methods of killing raf- activated cancer cells. Numerous oligonucleotide sequences are disclosed, none of which are actually antisense oligonucleotide sequences.
  • the present invention provides oligonucleotides which are targeted to nucleic acids encoding human raf and are capable of inhibiting raf expression.
  • the present invention also provides chimeric oligonucleotides targeted to nucleic acids encoding human raf.
  • the oligonucleotides 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 inhibiting the expression of human raf, particularly the abnormal expression of raf. These methods are believed to be useful both therapeutically and diagnostically as a consequence of the association between raf expression and hyperproliferation. These methods are also useful as tools, for example for detecting and determining the role of raf expression in various cell functions and physiological processes and conditions and for diagnosing conditions associated with raf expression.
  • the present invention also comprises methods of inhibiting hyperproliferation of cells using oligonucleotides of the invention. These methods are believed to be useful, for example in diagnosing raf-associated cell hyperproliferation. Methods of treating abnormal proliferative conditions are also provided. These methods employ the oligonucleotides of the invention. These methods are believed to be useful both therapeutically and as clinical research and diagnostic tools.
  • ISIS 5132 ( Figure 1A) and a scrambled control oligonucleotide ISIS 10353 ( Figure IB) on growth of A549 lung tumor xenografts in nude mice.
  • ISIS 5132 decreased tumor size at all doses (0.006 mg/kg; 0.06 mg/kg; 0.6 . mg/kg; and 6.0 mg/kg) in a dose- dependent manner.
  • the scrambled raf oligonucleotide, ISIS 10353 had no effect at any dose ( Figure IB) .
  • raf genes are members of a gene family which encode related proteins termed A- , B- and c-raf. Raf genes code for highly conserved serine-threonine- specific protein kinases .
  • c-raf the most thoroughly characterized, is expressed in all organs and in all cell lines that have been examined.
  • A- and B-raf are expressed in urogenital and brain tissues, respectively.
  • c-raf protein kinase activity and subcellular distribution are regulated by mitogens via phosphorylation.
  • Various growth factors including epidermal growth factor, acidic fibroblast growth factor, platelet- derived growth factor, insulin, granulocyte-macrophage colony- stimulating factor, interleukin-2, interleukin-3 and erythropoietin, have been shown to induce phosphorylation of c- raf.
  • c-raf is believed to play a fundamental role in the normal cellular signal transduction pathway, coupling a multitude of growth factors to their net effect, cellular proliferation.
  • abnormal proliferative conditions are believed to be associated with raf expression and are, therefore, believed to be responsive to inhibition of raf expression. Abnormally high levels of expression of the raf protein are also implicated in transformation and abnormal cell proliferation. These abnormal proliferative conditions are also believed to be responsive to inhibition of raf expression. Examples of abnormal proliferative conditions are hyperproliferative disorders such as cancers, tumors, hyperplasias, pulmonary fibrosis, angiogenesis, psoriasis, atherosclerosis and smooth muscle cell proliferation in the blood vessels, such as stenosis or restenosis following angioplasty.
  • T-cell proliferation T-cell activation and growth
  • tissue graft rejection endotoxin shock
  • glomerular nephritis glomerular nephritis
  • raf gene expression may halt or reverse abnormal cell proliferation. This has been found even in when levels of raf expression are not abnormally high. There is a great desire to provide compositions of matter which can modulate the expression of the raf gene. It is greatly desired to provide methods of detection of the raf gene in cells, tissues and animals. It is also desired to provide methods of diagnosis and treatment of abnormal proliferative conditions associated with abnormal raf gene expression. In addition, kits and reagents for detection and study of the raf gene are desired. "Abnormal" raf gene expression is defined herein as abnormally high levels of expression of the raf protein, or any level of raf expression in an abnormal proliferative condition or state.
  • the present invention employs oligonucleotides targeted to nucleic acids encoding raf.
  • This relationship between an oligonucleotide and its complementary nucleic acid target to which it hybridizes is commonly referred to as "antisenseā€ .
  • "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 target is a nucleic acid encoding raf; in other words, the raf gene or mRNA expressed from the raf gene.
  • the targeting process also includes determination of a site or sites within the nucleic acid sequence for the oligonucleotide interaction to occur such that the desired effect- modulation of gene expression- will result. Once the target site or sites have been identified, oligonucleotides are chosen which are sufficiently complementary to the target, i.e., hybridize sufficiently well and with sufficient specificity, to give the desired modulation.
  • modulation means either inhibition or stimulation. Inhibition of raf gene expression is presently the preferred form of 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.
  • oligonucleotide and ā€œcomplementaryā€ 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. It is understood that an oligonucleotide need not be 100% complementary to its target nucleic acid sequence to be specifically hybridizable.
  • 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 vi tro assays, under conditions in which the assays are conducted.
  • oligonucleotides are provided which are targeted to mRNA encoding c-raf and A-raf.
  • mRNA includes not only the coding region which carries 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, intron regions and intron/exon or splice 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 coding ribonucleotides.
  • the oligonucleotide is targeted to a translation initiation site (AUG codon) or sequences in the 5' - or 3 ' -untranslated region of the human c- raf mRNA.
  • the functions of messenger RNA to be interfered with include all vital functions such as translocation of the RNA to the site for protein translation, actual translation of protein from the RNA, splicing or maturation of the RNA and possibly even independent catalytic activity which may be engaged in by the RNA.
  • the overall effect of such interference with the RNA function is to cause interference with raf protein expression.
  • the present invention provides oligonucleotides for modulation of raf gene expression. Such oligonucleotides are targeted to nucleic acids encoding raf. Oligonucleotides and methods for modulation of c-raf and A-raf are presently preferred; however, compositions and methods for modulating expression of other forms of raf are also believed to have utility and are comprehended by this invention.
  • modulation means either inhibition or stimulation. Inhibition of raf gene expression is presently the preferred form of modulation.
  • oligonucleotide refers to an oligomer or polymer of nucleotide or nucleoside monomers consisting of naturally occurring bases, sugars and intersugar (backbone) linkages.
  • oligonucleotide also includes oligomers comprising non-naturally occurring monomers, or portions thereof, which function similarly. Such modified or substituted oligonucleotides are often preferred over native forms because of properties such as, for example, enhanced cellular uptake and increased stability in the presence of nucleases.
  • oligonucleotides of this invention are chimeric oligonucleotides.
  • "Chimeric oligonucleotidesā€ or ā€œchimerasā€, 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 of modified nucleotides that confers one or more beneficial properties (such as, for example, increased nuclease resistance, increased uptake into cells, increased binding affinity for the RNA ' target) and a region that is a substrate for RNase H cleavage.
  • a chimeric oligonucleotide comprises at least one region modified to increase target binding affinity, and, usually, a region that acts as a substrate for RNAse H.
  • Affinity of an oligonucleotide for its target is routinely determined by measuring the Tm of an oligonucleotide/target pair, which is the temperature at which the oligonucleotide and target dissociate; dissociation is detected spectrophotometrically. The higher the Tm, the greater the affinity of the oligonucleotide for the target.
  • the region of the oligonucleotide which is modified to increase raf mRNA binding affinity comprises at least one nucleotide modified at the 2' position of the sugar, most preferably a 2'-0-alkyl or 2'-fluoro- modified nucleotide.
  • modifications are routinely incorporated into oligonucleotides and these oligonucleotides have been shown to have a higher Tm (i.e., higher target binding affinity) than 2' -deoxyoligonucleotides against a given target. The effect of such increased affinity is to greatly enhance antisense oligonucleotide inhibition of raf gene expression.
  • RNAse H is a cellular endonuclease that cleaves the RNA strand of RNA:DNA duplexes; activation of this enzyme therefore results in cleavage of the RNA target, and thus can greatly enhance the efficiency of antisense inhibition. Cleavage of the RNA target can be routinely demonstrated by gel electrophoresis .
  • the chimeric oligonucleotide is also modified to enhance nuclease resistance. Cells contain a variety of exo- and endo-nucleases which can degrade nucleic acids.
  • nucleotide and nucleoside modifications have been shown to make the oligonucleotide into which they are incorporated more resistant to nuclease digestion than the native oligodeoxynucleotide .
  • Nuclease resistance is routinely measured by incubating oligonucleotides with cellular extracts or isolated nuclease solutions and measuring the extent of intact oligonucleotide remaining over time, usually by gel electrophoresis .
  • Oligonucleotides which have been modified to enhance their nuclease resistance survive intact for a longer time than unmodified oligonucleotides.
  • a variety of oligonucleotide modifications have been demonstrated to enhance or confer nuclease resistance. Oligonucleotides which contain at least one phosphorothioate modification are presently more preferred. In some cases, oligonucleotide modifications which enhance target binding affinity are also, independently, able to enhance nuclease resistance.
  • oligonucleotides envisioned for this invention may contain phosphorothioates, phosphotriesters, methyl phosphonates, short chain alkyl or cycloalkyl intersugar linkages or short chain heteroatomic or heterocyclic intersugar ("backboneā€) linkages.
  • phosphorothioates and those with CH 2 -NH-0-CH 2 , CH 2 -N(CH 3 ) -O- CH 2 (known as the methylene (methylimino) or MMI backbone) , CH 2 - 0-N(CH 3 ) -CH 2 , CH 2 -N(CH 3 ) -N(CH 3 ) -CH 2 and 0-N(CH 3 ) -CH 2 -CH 2 backbones (where phosphodiester is 0-P-0-CH 2 ) .
  • oligonucleotides having morpholino backbone structures Summerton, J.E. and Weller, D.D., U.S. Patent No: 5,034,506.
  • the phosphodiester backbone of the oligonucleotide may be replaced with a polyamide backbone, the bases being bound directly or indirectly to the aza nitrogen atoms of the polyamide backbone.
  • oligonucleotides may contain alkyl and halogen-substituted sugar moieties comprising one of the following at the 2' position: OH, SH, SCH 3 , F, OCN, OCH 3 OCH 3 , OCH 3 0(CH 2 ) n CH 3 , 0(CH 2 ) n NH 2 or 0(CH 2 ) n CH 3 where n is from 1 to about 10; C- L to C 10 lower alkyl, substituted lower alkyl, alkaryl or aralkyl; Cl; Br; CN; CF 3 ; OCF 3 ; 0- , S-, or N-alkyl; O- , S-, or N-alkenyl; SOCH 3 ; S0 2 CH 3 ; 0N0 2 ; N0 2 ; N 3 ; NH 2 ; heterocycloalkyl; heterocycloalkaryl ; aminoalkylamino; polyalkylamino; substituted silyl
  • oligonucleotides in accordance with this invention preferably are from about 8 to about 50 nucleotides in length. In the context of this invention it is understood that this encompasses non-naturally occurring oligomers as hereinbefore described, having 8 to 50 monomers.
  • 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 also well known to use similar techniques to prepare other oligonucleotides such as the phosphorothioates and alkylated derivatives.
  • CPG controlled-pore glass
  • tissues or cells are contacted with oligonucleotides.
  • 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 vi tro or ex vivo, or to administer the oligonucleotide (s) to cells or tissues within an animal.
  • compositions of this 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) , oral, or parenteral, for example by intravenous drip, intravenous injection or subcutaneous, intraperitoneal or intramuscular injection.
  • Formulations for topical administration may include 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, flavorings, diluents, emulsifiers, dispersing aids or binders may be desirable.
  • Formulations for parenteral administration may include sterile aqueous solutions which may also contain buffers, diluents and other suitable additives.
  • cationic lipids may be included in the formulation to facilitate oligonucleotide uptake.
  • One such composition shown to facilitate uptake is Lipofectin (BRL, Bethesda MD) .
  • Dosing is dependent on severity and responsiveness of the condition to be treated, with course of treatment lasting from several days to several months or until a cure is effected or a diminution of disease state is achieved.
  • Optimal dosing schedules can be calculated from measurements of drug accumulation in the body. 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 calculated based on EC50's in in vi tro and in vivo animal studies. For example, given the molecular weight of compound (derived from oligonucleotide sequence and chemical structure) and an effective dose such as an IC50, for example (derived experimentally) , a dose in mg/kg is routinely calculated.
  • the present invention is also suitable for diagnosing abnormal proliferative states in tissue or other samples from patients suspected of having a hyperproliferative disease such as cancer, psoriasis or blood vessel restenosis or atherosclerosis.
  • a hyperproliferative disease such as cancer, psoriasis or blood vessel restenosis or atherosclerosis.
  • the ability of the oligonucleotides of the present invention to inhibit cell proliferation may be employed to diagnose such states.
  • 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.
  • the present invention can be used to distinguish raf-associated tumors from tumors having other etiologies, in order that an efficacious treatment regime can be designed.
  • 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.
  • oligonucleotides of the invention are also useful for detection and diagnosis of raf expression.
  • radiolabeled oligonucleotides can be prepared by 32 P labeling at the 5' end with polynucleotide kinase. Sambrook et al . , Molecular Cloning. A Labora tory Manual , Cold Spring Harbor Laboratory Press, 1989, Volume 2, p. 10.59. Radiolabeled oligonucleotides are then contacted with tissue or cell samples suspected of raf expression and the sample is washed to remove unbound oligonucleotide.
  • Radioactivity remaining in the sample indicates bound oligonucleotide (which in turn indicates the presence of raf) and can be quantitated using a scintillation counter or other routine means.
  • Radiolabeled oligo can also be used to perform autoradiography of tissues to determine the localization, distribution and quantitation of raf expression for research, diagnostic or therapeutic purposes. In such studies, tissue sections are treated with radiolabeled oligonucleotide and washed as described above, then exposed to photographic emulsion according to routine autoradiography procedures. The emulsion, when developed, yields an image of silver grains over the regions expressing raf. Quantitation of the silver grains permits raf expression to be detected.
  • Analogous assays for fluorescent detection of raf expression can be developed using oligonucleotides of the invention which are conjugated with fluorescein or other fluorescent tag instead of radiolabeling.
  • conjugations are routinely accomplished during solid phase synthesis using fluorescently labeled amidites or CPG (e.g., fluorescein- labeled amidites and CPG available from Glen Research, Sterling VA. See 1993 Catalog of Products for DNA Research, Glen Research, Sterling VA, p. 21) .
  • Oligonucleotide inhibition of c-raf expression The oligonucleotides shown in Table 1 were designed using the Genbank c-raf sequence HUMRAFR (Genbank listing X03484) , synthesized and tested for inhibition of c-raf mRNA expression in T24 bladder carcinoma cells using a Northern blot assay. All are oligodeoxynucleotides with phosphorothioate backbones.
  • oligonucleotides 5074, 5075, 5132, 8034, 7826, 7827 and 7828 showed at least 50% inhibition of c-raf mRNA and these oligonucleotides are therefore preferred.
  • Oligonucleotides 5132 and 7826 (SEQ ID NO: 8 and SEQ ID NO: 17) showed greater than about 90% inhibition and are more preferred.
  • oligonucleotides 6721, 7848, 7847 and 8094 decreased c-raf mRNA levels by greater than 50%. These oligonucleotides are also preferred. Of these, 7847 (SEQ ID NO: 22) showed greater than about 90% inhibition of c-raf mRNA and is more preferred.
  • ISIS 5132 Specificity of ISIS 5132 for raf mRNA was demonstrated by a Northern blot assay in which this oligonucleotide was tested for the ability to inhibit Ha-ras mRNA as well as c-raf mRNA in T24 cells.
  • Ha-ras is a cellular oncogene which is implicated in transformation and tumorigenesis .
  • ISIS 5132 was shown to abolish c-raf mRNA almost completely with no effect on Ha-ras mRNA levels.
  • Oligonucleotides from Table 2 having uniform 2'O-methyl modifications and a phosphorothioate backbone were tested for ability to inhibit c-raf protein expression in T24 cells as determined by Western blot assay. Oligonucleotides 8033, 7834 and 7835 showed the greatest inhibition and are preferred. Of these, 8033 and 7834 are more preferred.
  • Chimeric oligonucleotides having SEQ ID NO: 8 were prepared. These oligonucleotides had central "gap" regions of 6, 8, or 10 deoxynucleotides flanked by two regions of 2'-0- methyl modified nucleotides. Backbones were uniformly phosphorothioate. In Northern blot analysis, all three of these oligonucleotides (ISIS 6720, 6-deoxy gap; ISIS 6717, 8- deoxy gap; ISIS 6729, 10-deoxy gap) showed greater than 70% inhibition of c-raf mRNA expression in T24 cells. These oligonucleotides are preferred. The 8-deoxy gap compound (6717) showed greater than 90% inhibition and is more preferred.
  • Additional chimeric oligonucleotides were synthesized having one or more regions of 2 '-O-methyl modification and uniform phosphorothioate backbones. These are shown in Table 3. All are phosphorothioates; bold regions indicate 2'-0- methyl modified regions.
  • ISIS 7848, 7849, 7851, 7856, 7855, 7854, 7847, and 7853 gave better than 70% inhibition and are therefore preferred. Of these, 7851, 7855, 7847 and 7853 gave greater than 90% inhibition and are more preferred.
  • oligonucleotides 6720, 6717, 6729, 9720 and 9058 are preferred. Oligonucleotides 6717, 6729, 9720 and 9058 are more preferred.
  • the phosphorothioate oligonucleotide ISIS 5132 was shown to inhibit T24 bladder cancer cell proliferation.
  • Cells were treated with various concentrations of oligonucleotide in conjunction with lipofectin (cationic lipid which increases uptake of oligonucleotide) .
  • lipofectin cationic lipid which increases uptake of oligonucleotide
  • a dose-dependent inhibition of cell proliferation was demonstrated, as indicated in Table 6, in which "None" indicates untreated control (no oligonucleotide) and "Controlā€ indicates treatment with negative control oligonucleotide. Results are shown as percent inhibition compared to untreated control.
  • Subcutaneous human T24 bladder carcinoma xenografts in nude mice were established and treated with ISIS 5132 and an unrelated control phosphorothioate oligonucleotide administered intraperitoneally three times weekly at a dosage of 25 mg/kg.
  • ISIS 5132 inhibited tumor growth after eleven days by 35% compared to controls.
  • Oligonucleotide-treated tumors remained smaller than control tumors throughout the course of the study.
  • Subcutaneous human MDA-MB 231 breast carcinoma xenografts in nude mice were established and treated with ISIS 5132 and an unrelated control phosphorothioate oligonucleotide administered intravenously once per day at a dosage of 0.6 mg/kg or 6.0 mg/kg.
  • ISIS 5132 inhibited tumor growth after 27 days (end of study) by approximately 80% compared to controls.
  • ISIS 5132 was also effective when administered intraperitoneally to MDA-MB 231 xenografts in nude mice. Oligonucleotide was administered once per day at 0.6 mg/kg or 6.0 mg/kg. By day 27 (end of study) , tumor volume was inhibited by 57% (0.6 mg/kg dose) or 64% (6.0 mg/kg) compared to control .
  • ISIS 5132 decreased raf RNA levels after 27 days by 67% when given intravenously and 39% when given intraperitoneally (both at 6 mg/kg) .
  • ISIS 5132 Effect of ISIS 5132 on Colo 205 human colon carcinoma tumors
  • Subcutaneous human Colo 205 colon carcinoma xenografts in nude mice were established and treated with ISIS 5132 and an unrelated control phosphorothioate oligonucleotide administered intravenously once per day at a dosage of 6.0 mg/kg.
  • ISIS 5132 inhibited tumor growth after 25 days by over 40% compared to controls.
  • Subcutaneous human A549 lung adenocarcinoma xenografts were established in male Balb/c nude mice and treated with ISIS 5132 and a control oligonucleotide administered daily by intravenous injection at doses ranging from 0.006 to 6.0 mg/kg.
  • ISIS 5132 decreased tumor size at all doses in a dose-dependent manner, as shown in Figure 1A.
  • ISIS 6717 a 2' -O-methyl gapped oligonucleotide
  • ISIS 5132 for ability to inhibit A549 tumor xenograft growth.
  • doses of 0.006, 0.06, 0.6 and 6.0 mg/kg given intravenously the two oligonucleotides were virtually indistinguishable in their effects on tumor growth.
  • ISIS 6717 is therefore a preferred embodiment of this invention.
  • oligonucleotides targeted to portions of the A-raf mRNA and which inhibit A-raf expression will be useful for interfering with cell hyperproliferation.
  • Methods for inhibiting A-raf expression using such antisense oligonucleotides are, likewise, believed to be useful for interfering with cell hyperproliferation.
  • the phosphorothioate deoxyoligonucleotides shown in Table 7 were designed and synthesized using the Genbank A-raf sequence HUMARAFIR (Genbank listing X04790) .
  • Oligonucleotides ISIS 9061, ISIS 9069 and ISIS 10228 were evaluated by Northern blot analysis for their effects on A-raf mRNA levels in A549, T24 and NHDF cells. All three oligonucleotides decreased A-raf RNA levels in a dose-dependent manner in all three cell types, with inhibition of greater than 50% at a 500 nM dose in all cell types. The greatest inhibition (88%) was achieved with ISIS 9061 and 9069 in T24 cells. These three oligonucleotides (ISIS 9061, 9069 and 10228) are preferred, with ISIS 9069 and 9061 being more preferred.
  • tissue graft rejection is a condition which is likely to be ameliorated by interference with raf expression; but, clearly, this must be evaluated in animals rather than human transplant patients.
  • Another such example is restenosis.
  • Oligonucleotide sequences for inhibiting c-raf expression in rat and mouse cells were identified.
  • Rat and mouse c-raf genes have regions of high homology; a series of oligonucleotides which target both rat and mouse c-raf mRNA sequence were designed and synthesized, using information gained from evaluation of oligonucleotides targeted to human c- raf. These oligonucleotides were screened for activity in mouse bEND cells and rat A-10 cells using Northern blot assays. The oligonucleotides (all phosphorothioates) are shown in Table 8:
  • Oligonucleotides ISIS 11061 and 10707 were found to inhibit c- raf RNA levels by greater than 90% in mouse bEND cells at a dose of 400 nM. These two oligonucleotides inhibited raf RNA levels virtually entirely in rat A-10 cells at a concentration of 200 nM.
  • the IC50 for ISIS 10707 was found to be 170 nM in mouse bEND cells and 85 nM in rat A-10 cells.
  • the IC50 for ISIS 11061 was determined to be 85 nM in mouse bEND cells and 30 nM in rat A-10 cells. Effect of ISIS-11061 on endogenous c-raf mRNA expression in mice
  • mice were injected intraperitoneally with ISIS 11061 (50 mg/kg) or control oligonucleotide or saline control once daily for three days. Animals were sacrificed and organs were analyzed for c-raf mRNA expression by Northern blot analysis.
  • ISIS 11061 was found to decrease levels of c-raf mRNA in liver by approximately 70%. Control oligonucleotides had no effects on c-raf expression. The effect of ISIS 11061 was specific for c-raf; A-raf and G3PDH RNA levels were unaffected by oligonucleotide treatment.
  • Antisense oligonucleotide to c-raf increases survival in murine heart allograft model
  • oligonucleotide ISIS 11061 was tested for activity in a murine vascularized heterotopic heart transplant model.
  • Hearts from C57BI10 mice were transplanted into the abdominal cavity of C3H mice as primary vascularized grafts essentially as described by Isobe et al . , Circulation 1991, 84, 1246-1255.
  • Oligonucleotides were administered by continuous intravenous administration via a 7-day Alzet pump.
  • the mean allograft survival time for untreated mice was 7.83 ā‡ 0.75 days ( 7, 7, 8, 8, 8, 9 days) .
  • mice treated for 7 days with 20 mg/kg or 40 mg/kg ISIS 11061 all survived at least 11 days (11,11,12 days for 20 mg/kg dose and >11, >11, >11 days for the 40 mg/kg dose) .
  • oligonucleotides are administered either by intravenous injection, continuous intravenous administration via Alzet pump, or direct administration to the carotid artery in a pluronic gel matrix as described by Bennett et al .
  • rats are sacrificed, carotid arteries are examined by microscopy and effects of treatment on luminal cross-sections are determined.
  • Unmodified DNA oligonucleotides were synthesized on an automated DNA synthesizer (Applied Biosystems model 380B) using standard phosphoramidite chemistry with oxidation by iodine. ā‡ -cyanoethyldiisopropyl phosphoramidites were purchased from Applied Biosystems (Foster City, CA) . For phosphorothioate oligonucleotides, the standard oxidation bottle was replaced by a 0.2 M solution of H-l, 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.
  • 2 '-O-methyl phosphorothioate oligonucleotides were synthesized using 2'-O-methyl ā‡ - cyanoethyldiisopropyl-phosphoramidites (Chemgenes, Needham MA) and the standard cycle for unmodified oligonucleotides, except the wait step after pulse delivery of tetrazole and base was increased to 360 seconds.
  • the 3 '-base used to start the synthesis was a 2 ' -deoxyribonucleotide.
  • 2'-0-propyl oligonucleotides were prepared by a slight modification of this procedure.
  • 2'-fluoro phosphorothioate oligonucleotides were synthesized using 5' -dimethoxytrityl-3 ' -phosphoramidites and prepared as disclosed in U.S. patent application Serial No. 463,358, filed January 11, 1990, and 566,977, filed August 13, 1990, which are assigned to the same assignee as the instant application and which are incorporated by reference herein.
  • the 2'-fluoro oligonucleotides were prepared using phosphoramidite chemistry and a slight modification of the standard DNA synthesis protocol: deprotection was effected using methanolic ammonia at room temperature. After cleavage from the controlled pore glass column
  • oligonucleotides were purified by precipitation twice out of 0.5 M NaCl with 2.5 volumes ethanol. Analytical gel electrophoresis was accomplished in 20% acrylamide, 8 M urea, 45 mM Tris-borate buffer, pH 7.0. Oligodeoxynucleotides and their phosphorothioate analogs were judged from electrophoresis to be greater than 80% full length material .
  • the human urinary bladder cancer cell line T24 was obtained from the American Type Culture Collection (Rockville MD) .
  • Cells were grown in McCoy's 5A medium with L-glutamine (Gibco BRL, Gaithersburg MD) , supplemented with 10% heat-inactivated fetal calf serum and 50 U/ml each of penicillin and streptomycin.
  • Cells were seeded on 100 mm plates. When they reached 70% confluency, they were treated with oligonucleotide. Plates were washed with 10 ml prewarmed PBS and 5 ml of Opti-MEM reduced- serum medium containing 2.5 ā‡ l DOTMA. Oligonucleotide with lipofectin was then added to the desired concentration.
  • Kingston, R.E. in Current Protocols in Molecular Biology, (F.M. Ausubel, R. Brent, R.E. Kingston, D.D. Moore, J.A. Smith, J.G. Seidman and K. Strahl, eds . ) , John Wiley and Sons, NY.
  • RNA samples were electrophoresed through 1.2% agarose-formaldehyde gels and transferred to hybridization membranes by capillary diffusion over a 12-14 hour period.
  • the RNA was cross-linked to the membrane by exposure to UV light in a Stratalinker (Stratagene,
  • T24 cells were treated on day 0 for two hours with various concentrations of oligonucleotide and lipofectin (50 nM oligonucleotide in the presence of 2 ā‡ g/ml lipofectin; 100 nM oligonucleotide and 2 ā‡ g/ml lipofectin; 250 nM oligonucleotide and 6 ā‡ g/ml lipofectin or 500 nM oligonucleotide and 10 ā‡ g/ml lipofectin) .
  • cells were treated for a second time at desired oligonucleotide concentration for two hours.
  • cells were counted.
  • Oligonucleotides ISIS 5132 and an unrelated control phosphorothioate oligonucleotide suspended in saline) were administered three times weekly beginning on day
  • Oligonucleotides were given by intraperitoneal injection.
  • Oligonucleotide dosage was 25 mg/kg. Tumor size was measured and tumor volume was calculated on the eleventh, fifteenth and eighteenth treatment days.
  • Oligonucleotides ISIS 5132 and an unrelated control phosphorothioate oligonucleotide suspended in saline) were administered once daily beginning on day 10 after tumor cell inoculation. A saline-only control was also given. Oligonucleotides were given by intravenous injection at a dosage of 0.6 mg/kg or 6.0 mg/kg. Tumor size was measured and tumor volume was calculated on days 10, 13, 16, 20, 23 and 27 following tumor cell inoculation.
  • oligonucleotides were administered once daily beginning on day 10 after tumor cell inoculation. A saline-only control was also given. Oligonucleotides were given by intraperitoneal injection at a dosage of 0.6 mg/kg or 6.0 mg/kg. Tumor size was measured and tumor volume was calculated on days 10, 13, 16, 20, 23 and 27 following tumor cell inoculation.
  • Example 7 Effect of ISIS 5132 on Colo 205 Human Colon Carcinoma Tumor Xenografts in Nude Mice 5 x 10 6 Colo 205 cells were implanted subcutaneously in the right inner thigh of nude mice. Oligonucleotides (ISIS 5132 and an unrelated control phosphorothioate oligonucleotide suspended in saline) were administered once per day beginning on day 5 after tumor cell inoculation. A saline-only control was also given. Oligonucleotides were given by intravenous injection. Oligonucleotide dosage was 6 mg/kg. Tumor size was measured and tumor volume was calculated on days 5, 8, 11, 14,
  • Tumors arising from raf expression are diagnosed and distinguished from other tumors using this assay.
  • a biopsy sample of the tumor is treated, e.g. , with collagenase or trypsin or other standard methods, to dissociate the tumor mass.
  • 5 x 10 6 tumor cells are implanted subcutaneously in the inner thighs of two or more nude mice.
  • Antisense oligonucleotide e.g. , ISIS 5132
  • suspended in saline is administered to one or more mice by intraperitoneal injection three times weekly beginning on day 4 after tumor cell inoculation. Saline only is given to a control mouse.
  • Oligonucleotide dosage is 25 mg/kg. Tumor size is measured and tumor volume is calculated on the eleventh treatment day.
  • Tumor volume of the oligonucleotide-treated mice is compared to that of the control mouse.
  • the volume of raf- associated tumors in the treated mice are measurably smaller than tumors in the control mouse.
  • Tumors arising from causes other than raf expression are not expected to respond to the oligonucleotides targeted to raf and, therefore, the tumor volumes of oligonucleotide- treated and control mice are equivalent .
  • Example 9 Detection of raf expression
  • Oligonucleotides are radiolabeled after synthesis by 32 P labeling at the 5' end with polynucleotide kinase.
  • Radiolabeled oligonucleotides are contacted with tissue or cell samples suspected of raf expression, such as tumor biopsy samples or skin samples where psoriasis is suspected, under conditions in which specific hybridization can occur, and the sample is washed to remove unbound oligonucleotide. Radioactivity remaining in the sample indicates bound oligonucleotide and is quantitated using a scintillation counter or other routine means.
  • Radiolabeled oligonucleotides of the invention are also used in autoradiography. Tissue sections are treated with radiolabeled oligonucleotide and washed as described above, then exposed to photographic emulsion according to standard autoradiography procedures. The emulsion, when developed, yields an image of silver grains over the regions expressing raf . The extent of raf expression is determined by quantitation of the silver grains.
  • Analogous assays for fluorescent detection of raf expression use oligonucleotides of the invention which are labeled with fluorescein or other fluorescent tags .
  • Labeled DNA oligonucleotides are synthesized on an automated DNA synthesizer (Applied Biosystems model 380B) using standard phosphoramidite chemistry with oxidation by iodine.
  • ā‡ -cyanoethyldiisopropyl phosphoramidites are purchased from Applied Biosystems (Foster City, CA) .
  • Fluorescein-labeled amidites are purchased from Glen Research (Sterling VA) .
  • oligonucleotide and biological sample are carried out as described for radiolabeled oligonucleotides except that instead of a scintillation counter, a fluorimeter or fluorescence microscope is used to detect the fluorescence which indicates raf expression.
  • Example 10 A549 xenografts 5 x 10 6 A549 cells were implanted subcutaneously in the inner thigh of nude mice. Oligonucleotides (ISIS 5132 and a scrambled raf control phosphorothioate oligonucleotide, ISIS 10353) suspended in saline were administered once daily by intravenous injection at doses ranging from 0.006 to 6.0 mg/kg. Resulting tumors were measured on days 9, 12, 17 and 21 and tumor volumes were calculated.
  • Oligonucleotides ISIS 5132 and a scrambled raf control phosphorothioate oligonucleotide, ISIS 10353
  • Example 11 Effect of oligonucleotide on endogenous c-raf expression
  • mice were treated by intraperitoneal injection at an oligonucleotide dose of 50 mg/kg on days 1, 2 and 3. On day 4 animals were sacrificed and organs removed for c-raf mRNA assay by Northern blot analysis. Four groups of animals were employed: 1) no oligonucleotide treatment (saline) ; 2) negative control oligonucleotide ISIS 1082 (targeted to herpes simplex virus; 3) negative control oligonucleotide 4189 (targeted to mouse protein kinase C- ā‡ ; 4) ISIS 11061 targeted to rodent c-raf.
  • Hearts were transplanted into the abdominal cavity of rats or mice (of a different strain from the donor) as primary vascu ā‡ larized grafts essentially as described by Isobe et al . , Circulation 1991, 84, 1246-1255. Oligonucleotides were administered by continuous intravenous administration via a 7-day Alzet pump. Cardiac allograft survival was monitored by listening for the presence of a second heartbeat in the abdominal cavity.
  • Example 13 Proliferation assay using rat A-10 smooth muscle cells
  • A10 cells were plated into 96-well plates in Dulbecco's modified Eagle medium (DMEM) + 10% fetal calf serum and allowed to attach for 24 hours. Cells were made quiescent by the addition of DMEM + 0.2% dialyzed fetal calf serum for an additional 24 hours. During the last 4 hours of quiescence, cells were treated with ISIS 11061 + lipofectin (Gibco-BRL, Bethesda MD) in serum-free medium. Medium was then removed, replaced with fresh medium and the cells were stimulated with 10% fetal calf serum. The plates were the placed into the incubator and cell growth was evaluated by MTS conversion to formozan (Promega cell proliferation kit) at 24 and 48 hours after serum stimulation. A control oligonucleotide, ISIS 1082 (an unrelated oligonucleotide targeted to herpes simplex virus) , was also tested.
  • ISIS 1082 an unrelated oligonucleotide targeted to herpes simplex virus
  • Example 14 Rat carotid artery restenosis model
  • Intimal hyperplasia is induced by balloon catheter dilatation of the carotid artery of the rat. Rats are anesthetized and common carotid artery injury is induced by passage of a balloon embolectomy catheter distended with 20 ml of saline. Oligonucleotides are applied to the adventitial surface of the arterial wall in a pluronic gel solution. Oligonucleotides are dissolved in a 0.25% pluronic gel solution at 4Ā°C (F127, BASF Corp.) at the desired dose.
  • rats are treated by intravenous injection or continuous intravenous infusion (via Alzet pump) of oligonucleotide.

Abstract

Oligonucleotides are provided which are targeted to nucleic acids encoding human raf and capable of inhibiting raf expression. In preferred embodiments, the oligonucleotides are targeted to mRNA encoding human c-raf or human A-raf. The oligonucleotides may have chemical modifications at one or more positions and may be chimeric oligonucleotides. Methods of inhibiting the expression of human raf using oligonucleotides of the invention are also provided. The present invention further comprises methods of inhibiting hyperproliferation of cells and methods of treating abnormal proliferative conditions which employ oligonucleotides of the invention.

Description

ANTISENSE OLIGONUCLEOTIDE MODULATION OF raf GENE EXPRESSION
FIELD OF THE INVENTION
This invention relates to compositions and methods for modulating expression of the raf gene, a naturally present cellular gene which has been implicated in abnormal cell proliferation and tumor formation. This invention is also directed to methods for inhibiting hyperproliferation of cells; these methods can be used diagnostically or therapeutically. Furthermore, this invention is directed to treatment of conditions associated with expression of the raf gene.
BACKGROUND OF THE INVENTION
Alterations in the cellular genes which directly or indirectly control cell growth and differentiation are considered to be the main cause of cancer. The raf gene family includes three highly conserved genes termed A- , B- and c-raf (also called raf-1) . Raf genes encode protein kinases that are thought to play important regulatory roles in signal transduction processes that regulate cell proliferation. Expression of the c-raf protein is believed to play a role in abnormal cell proliferation since it has been reported that 60% of all lung carcinoma cell lines express unusually high levels of c-raf mRNA and protein. Rapp et al . , The Oncogene Handbook, E.P. Reddy, A.M Skalka and T. Curran, eds . , Elsevier Science Publishers, New York, 1988, pp. 213-253. Oligonucleotides have been employed as therapeutic moieties in the treatment of disease states in animals and man. For example, workers in the field have now identified antisense, triplex and other oligonucleotide compositions which are capable of modulating expression of genes implicated in viral, fungal and metabolic diseases.
As examples, U. S. Patent 5,135,917, issued August 4, 1992, provides antisense oligonucleotides that inhibit human interleukin-1 receptor expression. U.S. Patent 5,098,890, issued March 24, 1992 in the name of Gewirtz et al . , is directed to antisense oligonucleotides complementary to the c- myb oncogene and antisense oligonucleotide therapies for certain cancerous conditions. U.S. Patent 5,087,617, issued February 11, 1992, provides methods for treating cancer patients with antisense oligonucleotides. U.S. Patent 5,166,195 issued November 24, 1992, provides oligonucleotide inhibitors of HIV. U.S. Patent 5,004,810, issued April 2, 1991, provides oligomers capable of hybridizing to herpes simplex virus Vmw65 mRNA and inhibiting replication. U.S. Patent 5,194,428, issued March 16, 1993, provides antisense oligonucleotides having antiviral activity against influenzavirus . U.S. Patent 4,806,463, issued February 21, 1989, provides antisense oligonucleotides and methods using them to inhibit HTLV-III replication. U.S. Patent 5,286,717 (Cohen et al . ) , issued February 15, 1994, is directed to a mixed linkage oligonucleotide phosphorothioates complementary to an oncogene; U.S. Patent 5,276,019 and U.S. Patent 5,264,423 (Cohen et al . ) are directed to phosphorothioate oligonucleotide analogs used to prevent replication of foreign nucleic acids in cells. Antisense oligonucleotides have been safely administered to humans and clinical trials of several antisense oligonucleotide drugs, targeted both to viral and cellular gene products, are presently underway. The phosphorothioate oligonucleotide, ISIS 2922, has been shown to be effective against cvtomegalovirus retinitis in AIDS patients. BioWorld Today, April 29, 1994, p. 3. It is thus established that oligonucleotides can be useful therapeutic instrumentalities and can be configured to be useful in treatment regimes for treatment of cells and animal subjects, especially humans.
Antisense oligonucleotide inhibition of gene expression has proven to be a useful tool in understanding the roles of raf genes. An antisense oligonucleotide complementary to the first six codons of human c-raf has been used to demonstrate that the mitogenic response of T cells to interleukin-2 (I -2) requires c-raf. Cells treated with the oligonucleotide showed a near-total loss of c-raf protein and a substantial reduction in proliferative response to IL-2. Riedel et al . , Eur . J. Immunol . 1993, 23, 3146-3150. Rapp et al . have disclosed expression vectors containing a raf gene in an antisense orientation downstream of a promoter, and methods of inhibiting raf expression by expressing an antisense Raf gene or a mutated Raf gene in a cell. WO application 93/04170. An antisense oligodeoxyribonucleotide complementary to codons 1-6 of murine c-Raf has been used to abolish insulin stimulation of DNA synthesis in the rat hepatoma cell line H4IIE. Tornkvist et al . , J. Biol . Chem . 1994, 269, 13919- 13921. WO Application 93/06248 discloses methods for identifying an individual at increased risk of developing cancer and for determining a prognosis and proper treatment of patients afflicted with cancer comprising amplifying a region of the c-raf gene and analyzing it for evidence of mutation.
Denner et al . disclose antisense polynucleotides hybridizing to the gene for raf, and processes using them. WO 94/15645. Oligonucleotides hybridizing to human and rat raf sequences are disclosed. Iversen et al . disclose heterotypic antisense oligonucleotides complementary to raf which are able to kill ras-activated cancer cells, and methods of killing raf- activated cancer cells. Numerous oligonucleotide sequences are disclosed, none of which are actually antisense oligonucleotide sequences.
There remains a long-felt need for improved compositions and methods for inhibiting raf gene expression.
SUMMARY OF THE INVENTION
The present invention provides oligonucleotides which are targeted to nucleic acids encoding human raf and are capable of inhibiting raf expression. The present invention also provides chimeric oligonucleotides targeted to nucleic acids encoding human raf. The oligonucleotides 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 inhibiting the expression of human raf, particularly the abnormal expression of raf. These methods are believed to be useful both therapeutically and diagnostically as a consequence of the association between raf expression and hyperproliferation. These methods are also useful as tools, for example for detecting and determining the role of raf expression in various cell functions and physiological processes and conditions and for diagnosing conditions associated with raf expression.
The present invention also comprises methods of inhibiting hyperproliferation of cells using oligonucleotides of the invention. These methods are believed to be useful, for example in diagnosing raf-associated cell hyperproliferation. Methods of treating abnormal proliferative conditions are also provided. These methods employ the oligonucleotides of the invention. These methods are believed to be useful both therapeutically and as clinical research and diagnostic tools.
DESCRIPTION OF THE DRAWING Figure 1 is a line graph showing the effect of ISIS
5132 (Figure 1A) and a scrambled control oligonucleotide ISIS 10353 (Figure IB) on growth of A549 lung tumor xenografts in nude mice. ISIS 5132 decreased tumor size at all doses (0.006 mg/kg; 0.06 mg/kg; 0.6 . mg/kg; and 6.0 mg/kg) in a dose- dependent manner. The scrambled raf oligonucleotide, ISIS 10353, had no effect at any dose (Figure IB) .
DETAILED DESCRIPTION OF THE INVENTION
Malignant tumors develop through a series of stepwise, progressive changes that lead to the loss of growth control characteristic of cancer cells, i.e., continuous unregulated proliferation, the ability to invade surrounding tissues, and the ability to metastasize to different organ sites. Carefully controlled in vi tro studies have helped define the factors that characterize the growth of normal and neoplastic cells and have led to the identification of specific proteins that control cell growth and differentiation. The raf genes are members of a gene family which encode related proteins termed A- , B- and c-raf. Raf genes code for highly conserved serine-threonine- specific protein kinases . These enzymes are differentially expressed; c-raf, the most thoroughly characterized, is expressed in all organs and in all cell lines that have been examined. A- and B-raf are expressed in urogenital and brain tissues, respectively. c-raf protein kinase activity and subcellular distribution are regulated by mitogens via phosphorylation. Various growth factors, including epidermal growth factor, acidic fibroblast growth factor, platelet- derived growth factor, insulin, granulocyte-macrophage colony- stimulating factor, interleukin-2, interleukin-3 and erythropoietin, have been shown to induce phosphorylation of c- raf. Thus, c-raf is believed to play a fundamental role in the normal cellular signal transduction pathway, coupling a multitude of growth factors to their net effect, cellular proliferation.
Certain abnormal proliferative conditions are believed to be associated with raf expression and are, therefore, believed to be responsive to inhibition of raf expression. Abnormally high levels of expression of the raf protein are also implicated in transformation and abnormal cell proliferation. These abnormal proliferative conditions are also believed to be responsive to inhibition of raf expression. Examples of abnormal proliferative conditions are hyperproliferative disorders such as cancers, tumors, hyperplasias, pulmonary fibrosis, angiogenesis, psoriasis, atherosclerosis and smooth muscle cell proliferation in the blood vessels, such as stenosis or restenosis following angioplasty. The cellular signalling pathway of which raf is a part has also been implicated in inflammatory disorders characterized by T-cell proliferation (T-cell activation and growth) , such as tissue graft rejection, endotoxin shock, and glomerular nephritis, for example.
It has now been found that elimination or reduction of raf gene expression may halt or reverse abnormal cell proliferation. This has been found even in when levels of raf expression are not abnormally high. There is a great desire to provide compositions of matter which can modulate the expression of the raf gene. It is greatly desired to provide methods of detection of the raf gene in cells, tissues and animals. It is also desired to provide methods of diagnosis and treatment of abnormal proliferative conditions associated with abnormal raf gene expression. In addition, kits and reagents for detection and study of the raf gene are desired. "Abnormal" raf gene expression is defined herein as abnormally high levels of expression of the raf protein, or any level of raf expression in an abnormal proliferative condition or state.
The present invention employs oligonucleotides targeted to nucleic acids encoding raf. This relationship between 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 target is a nucleic acid encoding raf; in other words, the raf gene or mRNA expressed from the raf gene. The targeting process also includes determination of a site or sites within the nucleic acid sequence for the oligonucleotide interaction to occur such that the desired effect- modulation of gene expression- will result. Once the target site or sites have been identified, oligonucleotides are chosen which are sufficiently complementary to the target, i.e., hybridize sufficiently well and with sufficient specificity, to give the desired modulation. In the context of this invention "modulation" means either inhibition or stimulation. Inhibition of raf gene expression is presently the preferred form of modulation. This modulation can be measured in ways which are routine in the art, for example by Northern blot assay of mRNA expression or Western blot assay of protein expression as taught in the examples of the instant application. Effects on cell proliferation or tumor cell growth can also be measured, as taught in the examples of the instant application. "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. 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 vi tro assays, under conditions in which the assays are conducted. In preferred embodiments of this invention, oligonucleotides are provided which are targeted to mRNA encoding c-raf and A-raf. In accordance with this invention, persons of ordinary skill in the art will understand that mRNA includes not only the coding region which carries 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, intron regions and intron/exon or splice 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 coding ribonucleotides. In preferred embodiments, the oligonucleotide is targeted to a translation initiation site (AUG codon) or sequences in the 5' - or 3 ' -untranslated region of the human c- raf mRNA. The functions of messenger RNA to be interfered with include all vital functions such as translocation of the RNA to the site for protein translation, actual translation of protein from the RNA, splicing or maturation of the RNA and possibly even independent catalytic activity which may be engaged in by the RNA. The overall effect of such interference with the RNA function is to cause interference with raf protein expression.
The present invention provides oligonucleotides for modulation of raf gene expression. Such oligonucleotides are targeted to nucleic acids encoding raf. Oligonucleotides and methods for modulation of c-raf and A-raf are presently preferred; however, compositions and methods for modulating expression of other forms of raf are also believed to have utility and are comprehended by this invention. As hereinbefore defined, "modulation" means either inhibition or stimulation. Inhibition of raf gene expression is presently the preferred form of modulation.
In the context of this invention, the term "oligonucleotide" refers to an oligomer or polymer of nucleotide or nucleoside monomers consisting of naturally occurring bases, sugars and intersugar (backbone) linkages. The term "oligonucleotide" also includes oligomers comprising non-naturally occurring monomers, or portions thereof, which function similarly. Such modified or substituted oligonucleotides are often preferred over native forms because of properties such as, for example, enhanced cellular uptake and increased stability in the presence of nucleases.
Certain preferred oligonucleotides of this invention 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 of modified nucleotides that confers one or more beneficial properties (such as, for example, increased nuclease resistance, increased uptake into cells, increased binding affinity for the RNA' target) and a region that is a substrate for RNase H cleavage. In one preferred embodiment, a chimeric oligonucleotide comprises at least one region modified to increase target binding affinity, and, usually, a region that acts as a substrate for RNAse H. Affinity of an oligonucleotide for its target (in this case a nucleic acid encoding raf) is routinely determined by measuring the Tm of an oligonucleotide/target pair, which is the temperature at which the oligonucleotide and target dissociate; dissociation is detected spectrophotometrically. The higher the Tm, the greater the affinity of the oligonucleotide for the target. In a more preferred embodiment, the region of the oligonucleotide which is modified to increase raf mRNA binding affinity comprises at least one nucleotide modified at the 2' position of the sugar, most preferably a 2'-0-alkyl or 2'-fluoro- modified nucleotide. Such modifications are routinely incorporated into oligonucleotides and these oligonucleotides have been shown to have a higher Tm (i.e., higher target binding affinity) than 2' -deoxyoligonucleotides against a given target. The effect of such increased affinity is to greatly enhance antisense oligonucleotide inhibition of raf gene expression. RNAse H is a cellular endonuclease that cleaves the RNA strand of RNA:DNA duplexes; activation of this enzyme therefore results in cleavage of the RNA target, and thus can greatly enhance the efficiency of antisense inhibition. Cleavage of the RNA target can be routinely demonstrated by gel electrophoresis . In another preferred embodiment, the chimeric oligonucleotide is also modified to enhance nuclease resistance. Cells contain a variety of exo- and endo-nucleases which can degrade nucleic acids. A number of nucleotide and nucleoside modifications have been shown to make the oligonucleotide into which they are incorporated more resistant to nuclease digestion than the native oligodeoxynucleotide . Nuclease resistance is routinely measured by incubating oligonucleotides with cellular extracts or isolated nuclease solutions and measuring the extent of intact oligonucleotide remaining over time, usually by gel electrophoresis . Oligonucleotides which have been modified to enhance their nuclease resistance survive intact for a longer time than unmodified oligonucleotides. A variety of oligonucleotide modifications have been demonstrated to enhance or confer nuclease resistance. Oligonucleotides which contain at least one phosphorothioate modification are presently more preferred. In some cases, oligonucleotide modifications which enhance target binding affinity are also, independently, able to enhance nuclease resistance.
Specific examples of some preferred oligonucleotides envisioned for this invention may contain phosphorothioates, phosphotriesters, methyl phosphonates, short chain alkyl or cycloalkyl intersugar linkages or short chain heteroatomic or heterocyclic intersugar ("backbone") linkages. Most preferred are phosphorothioates and those with CH2-NH-0-CH2, CH2-N(CH3) -O- CH2 (known as the methylene (methylimino) or MMI backbone) , CH2- 0-N(CH3) -CH2, CH2-N(CH3) -N(CH3) -CH2 and 0-N(CH3) -CH2-CH2 backbones (where phosphodiester is 0-P-0-CH2) . Also preferred are oligonucleotides having morpholino backbone structures. Summerton, J.E. and Weller, D.D., U.S. Patent No: 5,034,506. In other preferred embodiments, such as the protein-nucleic acid or peptide-nucleic acid (PNA) backbone, the phosphodiester backbone of the oligonucleotide may be replaced with a polyamide backbone, the bases being bound directly or indirectly to the aza nitrogen atoms of the polyamide backbone. P.E. Nielsen, M. Egholm, R.H. Berg, O. Buchardt, Science 1991, 254, 1497. Other preferred oligonucleotides may contain alkyl and halogen-substituted sugar moieties comprising one of the following at the 2' position: OH, SH, SCH3, F, OCN, OCH3OCH3, OCH30(CH2)nCH3, 0(CH2)nNH2 or 0(CH2)nCH3 where n is from 1 to about 10; C-L to C10 lower alkyl, substituted lower alkyl, alkaryl or aralkyl; Cl; Br; CN; CF3; OCF3; 0- , S-, or N-alkyl; O- , S-, or N-alkenyl; SOCH3; S02CH3; 0N02; N02; N3; NH2; heterocycloalkyl; heterocycloalkaryl ; aminoalkylamino; polyalkylamino; substituted silyl; an RNA cleaving group; a cholesteryl group; a conjugate; 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. Oligonucleotides may also have sugar mimetics such as cyclobutyls in place of the pentofuranosyl group. Other preferred embodiments may include at least one modified base form or "universal base" such as inosine.
The oligonucleotides in accordance with this invention preferably are from about 8 to about 50 nucleotides in length. In the context of this invention it is understood that this encompasses non-naturally occurring oligomers as hereinbefore described, having 8 to 50 monomers.
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 also well known to use similar techniques to prepare other oligonucleotides such as the phosphorothioates and alkylated derivatives. 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 modified oligonucleotides such as cholesterol-modified oligonucleotides . It has now been found that certain oligonucleotides targeted to portions of the c-raf mRNA are particularly useful for inhibiting raf expression and for interfering with cell hyperproliferation. Methods for inhibiting c-raf expression using antisense oligonucleotides are, likewise, useful for interfering with cell hyperproliferation. In the methods of the invention, tissues or cells are contacted with oligonucleotides. 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 vi tro or ex vivo, or to administer the oligonucleotide (s) to cells or tissues within an animal.
For therapeutics, methods of inhibiting hyperproliferation of cells and methods of treating abnormal proliferative conditions are provided. The formulation of therapeutic compositions and their subsequent administration is believed to be within the skill in the art. In general, for therapeutics, a patient suspected of needing such therapy is given an oligonucleotide in accordance with the invention, commonly in a pharmaceutically acceptable carrier, in amounts and for periods which will vary depending upon the nature of the particular disease, its severity and the patient's overall condition. The pharmaceutical compositions of this 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) , oral, or parenteral, for example by intravenous drip, intravenous injection or subcutaneous, intraperitoneal or intramuscular injection.
Formulations for topical administration may include 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, flavorings, diluents, emulsifiers, dispersing aids or binders may be desirable.
Formulations for parenteral administration may include sterile aqueous solutions which may also contain buffers, diluents and other suitable additives. In addition to such pharmaceutical carriers, cationic lipids may be included in the formulation to facilitate oligonucleotide uptake. One such composition shown to facilitate uptake is Lipofectin (BRL, Bethesda MD) .
Dosing is dependent on severity and responsiveness of the condition to be treated, with course of treatment lasting from several days to several months or until a cure is effected or a diminution of disease state is achieved. Optimal dosing schedules can be calculated from measurements of drug accumulation in the body. 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 calculated based on EC50's in in vi tro and in vivo animal studies. For example, given the molecular weight of compound (derived from oligonucleotide sequence and chemical structure) and an effective dose such as an IC50, for example (derived experimentally) , a dose in mg/kg is routinely calculated.
The present invention is also suitable for diagnosing abnormal proliferative states in tissue or other samples from patients suspected of having a hyperproliferative disease such as cancer, psoriasis or blood vessel restenosis or atherosclerosis. The ability of the oligonucleotides of the present invention to inhibit cell proliferation may be employed to diagnose such states. 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. Similarly, the present invention can be used to distinguish raf-associated tumors from tumors having other etiologies, in order that an efficacious treatment regime can be designed.
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.
The oligonucleotides of the invention are also useful for detection and diagnosis of raf expression. For example, radiolabeled oligonucleotides can be prepared by 32P labeling at the 5' end with polynucleotide kinase. Sambrook et al . , Molecular Cloning. A Labora tory Manual , Cold Spring Harbor Laboratory Press, 1989, Volume 2, p. 10.59. Radiolabeled oligonucleotides are then contacted with tissue or cell samples suspected of raf expression and the sample is washed to remove unbound oligonucleotide. Radioactivity remaining in the sample indicates bound oligonucleotide (which in turn indicates the presence of raf) and can be quantitated using a scintillation counter or other routine means. Radiolabeled oligo can also be used to perform autoradiography of tissues to determine the localization, distribution and quantitation of raf expression for research, diagnostic or therapeutic purposes. In such studies, tissue sections are treated with radiolabeled oligonucleotide and washed as described above, then exposed to photographic emulsion according to routine autoradiography procedures. The emulsion, when developed, yields an image of silver grains over the regions expressing raf. Quantitation of the silver grains permits raf expression to be detected.
Analogous assays for fluorescent detection of raf expression can be developed using oligonucleotides of the invention which are conjugated with fluorescein or other fluorescent tag instead of radiolabeling. Such conjugations are routinely accomplished during solid phase synthesis using fluorescently labeled amidites or CPG (e.g., fluorescein- labeled amidites and CPG available from Glen Research, Sterling VA. See 1993 Catalog of Products for DNA Research, Glen Research, Sterling VA, p. 21) .
Each of these assay formats is known in the art. One of skill could easily adapt these known assays for detection of raf expression in accordance with the teachings of the invention providing a novel and useful means to detect raf expression.
Oligonucleotide inhibition of c-raf expression The oligonucleotides shown in Table 1 were designed using the Genbank c-raf sequence HUMRAFR (Genbank listing X03484) , synthesized and tested for inhibition of c-raf mRNA expression in T24 bladder carcinoma cells using a Northern blot assay. All are oligodeoxynucleotides with phosphorothioate backbones.
Table 1
Human c-raf Kinase Antisense Oligonucleotides
Isis # Sequence (5' -* 3') Site SEQ ID NO:
5000 TGAAGGTGAGCTGGAGCCAT Coding 1 5074 GCTCCATTGATGCAGCTTAA AUG 2
5075 CCCTGTATGTGCTCCATTGA AUG 3
5076 GGTGCAAAGTCAACTAGAAG STOP 4
5097 ATTCTTAAACCTGAGGGAGC 5'UTR 5
5098 GATGCAGCTTAAACAATTCT 5'UTR 6 5131 CAGCACTGCAAATGGCTTCC 3'UTR 7
5132 TCCCGCCTGTGACATGCATT 3'UTR 8 5133 GCCGAGTGCCTTGCCTGGAA 3'UTR 9
5148 AGAGATGCAGCTGGAGCCAT Coding 10 5149 AGGTGAAGGCCTGGAGCCAT Coding 11 6721 GTCTGGCGCTGCACCACTCT 3'UTR 12
6722 CTGATTTCCAAAATCCCATG 3'UTR 13 6731 CTGGGCTGTTTGGTGCCTTA 3'UTR 14
6723 TCAGGGCTGGACTGCCTGCT 3'UTR 15 7825 GGTGAGGGAGCGGGAGGCGG 5'UTR 16 7826 CGCTCCTCCTCCCCGCGGCG 5'UTR 17
7827 TTCGGCGGCAGCTTCTCGCC 5'UTR 18
7828 GCCGCCCCAACGTCCTGTCG 5'UTR 19
7848 TCCTCCTCCCCGCGGCGGGT 5'UTR 20
7849 CTCGCCCGCTCCTCCTCCCC 5'UTR 21 7847 CTGGCTTCTCCTCCTCCCCT 3'UTR 22
8034 CGGGAGGCGGTCACATTCGG 5'UTR 23
8094 TCTGGCGCTGCACCACTCTC 3'UTR 24
In a first round screen of oligonucleotides at concentrations of 100 nM or 200 nM, oligonucleotides 5074, 5075, 5132, 8034, 7826, 7827 and 7828 showed at least 50% inhibition of c-raf mRNA and these oligonucleotides are therefore preferred. Oligonucleotides 5132 and 7826 (SEQ ID NO: 8 and SEQ ID NO: 17) showed greater than about 90% inhibition and are more preferred. In additional assays, oligonucleotides 6721, 7848, 7847 and 8094 decreased c-raf mRNA levels by greater than 50%. These oligonucleotides are also preferred. Of these, 7847 (SEQ ID NO: 22) showed greater than about 90% inhibition of c-raf mRNA and is more preferred.
Specificity of ISIS 5132 for raf
Specificity of ISIS 5132 for raf mRNA was demonstrated by a Northern blot assay in which this oligonucleotide was tested for the ability to inhibit Ha-ras mRNA as well as c-raf mRNA in T24 cells. Ha-ras is a cellular oncogene which is implicated in transformation and tumorigenesis . ISIS 5132 was shown to abolish c-raf mRNA almost completely with no effect on Ha-ras mRNA levels.
2' -modified oligonucleotides
Certain of these oligonucleotides were synthesized with either phosphodiester (P=0) or phosphorothioate (P=S) backbones and were also uniformly substituted at the 2' position of the sugar with either a 2' -O-methyl, 2'-0-propyl, or 2'-fluoro group. Oligonucleotides are shown in Table 2.
Table 2
Uniformly 2' Sugar-modified c-raf Oligonucleotides
ISIS # Sequence lite Modif. SEQ ID NO
6712 TCCCGCCTGTGACATGCATT 3'UTR OMe/P=S 8 8033 CGGGAGGCGGTCACATTCGG 5'UTR OMe/P=S 23
7829 GGTGAGGGAGCGGGAGGCGG 5'UTR OMe/P=S 16
7830 CGCTCCTCCTCCCCGCGGCG 5'UTR OMe/P=S 17
7831 TTCGGCGGCAGCTTCTCGCC 5'UTR OMe/P=S 18
7832 GCCGCCCCAACGTCCTGTCG 5'UTR OMe/P=S 19 7833 ATTCTTAAACCTGAGGGAGC 5'UTR OMe/P=S 5
7834 GATGCAGCTTAAACAATTCT 5'UTR OMe/P=S 6
7835 GCTCCATTGATGCAGCTTAA AUG OMe/P=S 2
7836 CCCTGTATGTGCTCCATTGA AUG OMe/P=S 3
8035 CGGGAGGCGGTCACATTCGG 5'UTR OPr/P=0 23 7837 GGTGAGGGAGCGGGAGGCGG 5'UTR OPr/P=0 16
7838 CGCTCCTCCTCCCCGCGGCG 5'UTR OPr/P=0 17
7839 TTCGGCGGCAGCTTCTCGCC 5'UTR OPr/P=0 18 7840 GCCGCCCCAACGTCCTGTCG 5'UTR OPr/P=0 19 7841 ATTCTTAAACCTGAGGGAGC 5'UTR OPr/P=0 5 7842 GATGCAGCTTAAACAATTCT 5'UTR OPr/P=0 6
7843 GCTCCATTGATGCAGCTTAA AUG OPr/P=0 2 7844 CCCTGTATGTGCTCCATTGA AUG OPr/P=0 3 9355 CGGGAGGCGGTCACATTCGG 5'UTR 2'F/P=S 23
Oligonucleotides from Table 2 having uniform 2'O-methyl modifications and a phosphorothioate backbone were tested for ability to inhibit c-raf protein expression in T24 cells as determined by Western blot assay. Oligonucleotides 8033, 7834 and 7835 showed the greatest inhibition and are preferred. Of these, 8033 and 7834 are more preferred.
Chimeric oligonucleotides
Chimeric oligonucleotides having SEQ ID NO: 8 were prepared. These oligonucleotides had central "gap" regions of 6, 8, or 10 deoxynucleotides flanked by two regions of 2'-0- methyl modified nucleotides. Backbones were uniformly phosphorothioate. In Northern blot analysis, all three of these oligonucleotides (ISIS 6720, 6-deoxy gap; ISIS 6717, 8- deoxy gap; ISIS 6729, 10-deoxy gap) showed greater than 70% inhibition of c-raf mRNA expression in T24 cells. These oligonucleotides are preferred. The 8-deoxy gap compound (6717) showed greater than 90% inhibition and is more preferred. Additional chimeric oligonucleotides were synthesized having one or more regions of 2 '-O-methyl modification and uniform phosphorothioate backbones. These are shown in Table 3. All are phosphorothioates; bold regions indicate 2'-0- methyl modified regions.
Table 3 Chimeric 2' -O-methyl P=S c-raf oligonucleotides IsiĪµ # Sequence Target site SEQ ID NO:
7848 TCCTCCTCCCCGCGGCGGGT 5 ' UTR 20 7852 TCCTCCTCCCCGCGGCGGGT 5 ' UTR 20
7849 CTCGCCCGCTCCTCCTCCCC 5'UTR 21
7851 CTCGCCCGCTCCTCCTCCCC 5'UTR 21
7856 TTCTCGCCCGCTCCTCCTCC 5'UTR 25
7855 TTCTCGCCCGCTCCTCCTCC 5'UTR 25 7854 TTCTCCTCCTCCCCTGGCAG 3 ' UTR 26
7847 CTGGCTTCTCCTCCTCCCCT 3 ' UTR 22
7850 CTGGCTTCTCCTCCTCCCCT 3 ' UTR 22
7853 CCTGCTGGCTTCTCCTCCTC 3 ' UTR 27
When tested for their ability to inhibit c-raf mRNA by Northern blot analysis, ISIS 7848, 7849, 7851, 7856, 7855, 7854, 7847, and 7853 gave better than 70% inhibition and are therefore preferred. Of these, 7851, 7855, 7847 and 7853 gave greater than 90% inhibition and are more preferred.
Additional chimeric oligonucleotides with various 2' modifications were prepared and tested. These are shown in Table 4. All are phosphorothioates; bold regions indicate 2'- modified regions.
Table 4 Chimeric 2'-modified P=S c-raf oligonucleotides Isis # Sequence Target site Modif.SEQ ID
6720 TCCCGCCTGTGACATGCATT 3 'UTR 2 ' -O-Me 8
6717 TCCCGCCTGTGACATGCATT 3 'UTR 2 ' -O-Me 8
6729 TCCCGCCTGTGACATGCATT 3 'UTR 2 ' -O-Me 8
8097 TCTGGCGCTGCACCACTCTC 3 'UTR 2 ' -O-Me 24 9270 TCCCGCCTGTGACATGCATT 3 'UTR 2 ' -O-Pro 8
9058 TCCCGCCTGTGACATGCATT 3 'UTR 2 ' - F 8
9057 TCTGGCGCTGCACCACTCTC 3 'UTR 2 ' - F 24 Of these, oligonucleotides 6720, 6717, 6729, 9720 and 9058 are preferred. Oligonucleotides 6717, 6729, 9720 and 9058 are more preferred.
Two chimeric oligonucleotides with 2'-0-propyl sugar modifications and chimeric P=0/P=S backbones were also synthesized. These are shown in Table 5, in which i talic regions indicate regions which are both 2 '-modified and have phosphodiester backbones .
Table 5 Chimeric 2'-modified P=S/P=0 c-raf oligonucleotides
Isis # Sequence Target site Modif.SEQ ID
9271 TCCCGCCTGTGACATGCATT 3 'UTR 2' -O-Pro 8 8096 TCTGGCGCTGCACCACTCTC 3 'UTR 2' -O-Pro 24
Inhibition of cancer cell proliferation
The phosphorothioate oligonucleotide ISIS 5132 was shown to inhibit T24 bladder cancer cell proliferation. Cells were treated with various concentrations of oligonucleotide in conjunction with lipofectin (cationic lipid which increases uptake of oligonucleotide) . A dose-dependent inhibition of cell proliferation was demonstrated, as indicated in Table 6, in which "None" indicates untreated control (no oligonucleotide) and "Control" indicates treatment with negative control oligonucleotide. Results are shown as percent inhibition compared to untreated control.
Table 6 Inhibition of T24 Cell Proliferation by ISIS 5132
Oligo cone. None Control 5132
50 nM 0 +9% 23% 100 nM 0 +4% 24% 250 nM 0 10% 74% 500 nM 0 18% 82% Effect of ISIS 5132 on T24 human bladder carcinoma tumors
Subcutaneous human T24 bladder carcinoma xenografts in nude mice were established and treated with ISIS 5132 and an unrelated control phosphorothioate oligonucleotide administered intraperitoneally three times weekly at a dosage of 25 mg/kg. In this preliminary study, ISIS 5132 inhibited tumor growth after eleven days by 35% compared to controls. Oligonucleotide-treated tumors remained smaller than control tumors throughout the course of the study.
Effect of ISIS 5132 on MDA-MB 231 human breast carcinoma tumors
Subcutaneous human MDA-MB 231 breast carcinoma xenografts in nude mice were established and treated with ISIS 5132 and an unrelated control phosphorothioate oligonucleotide administered intravenously once per day at a dosage of 0.6 mg/kg or 6.0 mg/kg. ISIS 5132 inhibited tumor growth after 27 days (end of study) by approximately 80% compared to controls.
ISIS 5132 was also effective when administered intraperitoneally to MDA-MB 231 xenografts in nude mice. Oligonucleotide was administered once per day at 0.6 mg/kg or 6.0 mg/kg. By day 27 (end of study) , tumor volume was inhibited by 57% (0.6 mg/kg dose) or 64% (6.0 mg/kg) compared to control .
Effect of ISIS 5132 on c-raf RNA levels in MDA-MB231 tumors
RNA was isolated from MDA-MD231 tumor xenografts and Northern blots were performed to evaluate oligonucleotide effects on raf RNA levels. ISIS 5132 decreased raf RNA levels after 27 days by 67% when given intravenously and 39% when given intraperitoneally (both at 6 mg/kg) .
Effect of ISIS 5132 on Colo 205 human colon carcinoma tumors Subcutaneous human Colo 205 colon carcinoma xenografts in nude mice were established and treated with ISIS 5132 and an unrelated control phosphorothioate oligonucleotide administered intravenously once per day at a dosage of 6.0 mg/kg. In this study, ISIS 5132 inhibited tumor growth after 25 days by over 40% compared to controls.
Effect of ISIS 5132 on A549 human lung adenocarcinoma tumors
Subcutaneous human A549 lung adenocarcinoma xenografts were established in male Balb/c nude mice and treated with ISIS 5132 and a control oligonucleotide administered daily by intravenous injection at doses ranging from 0.006 to 6.0 mg/kg. ISIS 5132 decreased tumor size at all doses in a dose-dependent manner, as shown in Figure 1A. A scrambled raf oligonucleotide, ISIS 10353, had no effect at any dose (Figure IB) .
Effect of ISIS 5132 on c-raf RNA levels in A549 tumor cells
RNA was isolated from A549 tumor xenografts and Northern blots were performed to evaluate oligonucleotide effects on raf RNA levels. ISIS 5132 progressively decreased raf RNA levels beginning 8 hours after start of oligo treatment. When the experiment was terminated at day 13, RNA levels were still declining and had reached levels approximately 15% of control.
Effect of ISIS 6717, a 2' -O-methyl gapped oligonucleotide, on A549 lung xenograft tumors ISIS 6717, a 2'-O-methyl gapped oligonucleotide shown in Table 4, was compared to ISIS 5132 for ability to inhibit A549 tumor xenograft growth. At doses of 0.006, 0.06, 0.6 and 6.0 mg/kg given intravenously, the two oligonucleotides were virtually indistinguishable in their effects on tumor growth. ISIS 6717 is therefore a preferred embodiment of this invention.
Antisense oligonucleotides targeted to A-raf
It is believed that certain oligonucleotides targeted to portions of the A-raf mRNA and which inhibit A-raf expression will be useful for interfering with cell hyperproliferation. Methods for inhibiting A-raf expression using such antisense oligonucleotides are, likewise, believed to be useful for interfering with cell hyperproliferation.
The phosphorothioate deoxyoligonucleotides shown in Table 7 were designed and synthesized using the Genbank A-raf sequence HUMARAFIR (Genbank listing X04790) .
Table 7 Oligonucleotides Targeted to Human A-raf Isis # Sequence Site SEQ ID NO
9060 GTC AAG ATG GGC TGA GGT GG 5' UTR 28
9061 CCA TCC CGG ACA GTC ACC AC Coding 29
9062 ATG AGC TCC TCG CCA TCC AG Coding 30 9063 AAT GCT GGT GGA ACT TGT AG Coding 31 9064 CCG GTA CCC CAG GTT CTT CA Coding 32
9065 CTG GGC AGT CTG CCG GGC CA Coding 33
9066 CAC CTC AGC TGC CAT CCA CA Coding 34
9067 GAG ATT TTG CTG AGG TCC GG Coding 35
9068 GCA CTC CGC TCA ATC TTG GG Coding 36 9069 CTA AGG CAC AAG GCG GGC TG Stop 37
9070 ACG AAC ATT GAT TGG CTG GT 3' UTR 38
9071 GTA TCC CCA AAG CCA AGA GG 3' UTR 39
10228 CAT CAG GGC AGA GAC GAA CA 3' UTR 40
Oligonucleotides ISIS 9061, ISIS 9069 and ISIS 10228 were evaluated by Northern blot analysis for their effects on A-raf mRNA levels in A549, T24 and NHDF cells. All three oligonucleotides decreased A-raf RNA levels in a dose-dependent manner in all three cell types, with inhibition of greater than 50% at a 500 nM dose in all cell types. The greatest inhibition (88%) was achieved with ISIS 9061 and 9069 in T24 cells. These three oligonucleotides (ISIS 9061, 9069 and 10228) are preferred, with ISIS 9069 and 9061 being more preferred.
Identification of oligonucleotides targeted to rat and mouse c- raf
Many conditions which are believed to be mediated by raf kinase are not amenable to study in humans. For example, tissue graft rejection is a condition which is likely to be ameliorated by interference with raf expression; but, clearly, this must be evaluated in animals rather than human transplant patients. Another such example is restenosis. These conditions can be tested in animal models, however, such as the rat and mouse models used here. Oligonucleotide sequences for inhibiting c-raf expression in rat and mouse cells were identified. Rat and mouse c-raf genes have regions of high homology; a series of oligonucleotides which target both rat and mouse c-raf mRNA sequence were designed and synthesized, using information gained from evaluation of oligonucleotides targeted to human c- raf. These oligonucleotides were screened for activity in mouse bEND cells and rat A-10 cells using Northern blot assays. The oligonucleotides (all phosphorothioates) are shown in Table 8:
TABLE 8 Oligonucleotides targeted to mouse and rat c-raf
ISIS # Target Lte Sequence SEQ ID:
10705 Coding GGAACATCTGGAATTTGGTC 41 1 100770066 C Cooddiinnqg GATTCACTGTGACTTCGAAT 42
10707 3 'UTR GCTTCCATTTCCAGGGCAGG 43
10708 3 'UTR AAGAAGGCAATATGAAGTTA 44
10709 3 'UTR GTGGTGCCTGCTGACTCTTC 45
10710 3 'UTR CTGGTGGCCTAAGAACAGCT 46 1 100771111 A AUUGG GTATGTGCTCCATTGATGCA 47
10712 AUG TCCCTGTATGTGCTCCATTG 48
11060 5'UTR ATACTTATACCTGAGGGAGC 49
11061 5'UTR ATGCATTCTGCCCCCAAGGA 50
11062 3 'UTR GACTTGTATACCTCTGGAGC 51 1 111006633 3 3''UUTTRR ACTGGCACTGCACCACTGTC 52
11064 3 'UTR AAGTTCTGTAGTACCAAAGC 53
11065 3 'UTR CTCCTGGAAGACAGATTCAG 54
Oligonucleotides ISIS 11061 and 10707 were found to inhibit c- raf RNA levels by greater than 90% in mouse bEND cells at a dose of 400 nM. These two oligonucleotides inhibited raf RNA levels virtually entirely in rat A-10 cells at a concentration of 200 nM. The IC50 for ISIS 10707 was found to be 170 nM in mouse bEND cells and 85 nM in rat A-10 cells. The IC50 for ISIS 11061 was determined to be 85 nM in mouse bEND cells and 30 nM in rat A-10 cells. Effect of ISIS-11061 on endogenous c-raf mRNA expression in mice
Mice were injected intraperitoneally with ISIS 11061 (50 mg/kg) or control oligonucleotide or saline control once daily for three days. Animals were sacrificed and organs were analyzed for c-raf mRNA expression by Northern blot analysis.
ISIS 11061 was found to decrease levels of c-raf mRNA in liver by approximately 70%. Control oligonucleotides had no effects on c-raf expression. The effect of ISIS 11061 was specific for c-raf; A-raf and G3PDH RNA levels were unaffected by oligonucleotide treatment.
Antisense oligonucleotide to c-raf increases survival in murine heart allograft model
To determine the therapeutic effects of the c-raf antisense oligonucleotide ISIS 11061 in preventing allograft rejection, this oligonucleotide was tested for activity in a murine vascularized heterotopic heart transplant model. Hearts from C57BI10 mice were transplanted into the abdominal cavity of C3H mice as primary vascularized grafts essentially as described by Isobe et al . , Circulation 1991, 84, 1246-1255. Oligonucleotides were administered by continuous intravenous administration via a 7-day Alzet pump. The mean allograft survival time for untreated mice was 7.83 Ā± 0.75 days ( 7, 7, 8, 8, 8, 9 days) . Allografts in mice treated for 7 days with 20 mg/kg or 40 mg/kg ISIS 11061 all survived at least 11 days (11,11,12 days for 20 mg/kg dose and >11, >11, >11 days for the 40 mg/kg dose) .
In a pilot study conducted in rats, hearts from Lewis rats were transplanted into the abdominal cavity of ACI rats. Rats were dosed with ISIS 11061 at 20 mg/kg for 7 days via Alzet pump. The mean allograft survival time for untreated rats was 8.86+_ 0.69 days (8, 8, 9, 9, 9, 9, 10 days) . In rats treated with oligonucleotide, the allograft survival time was 15.3 +.1.15 days (14, 16, 16 days) .
Effects of antisense oligonucleotide targeted to c-raf on smooth muscle cell proliferation Smooth muscle cell proliferation is a cause of blood vessel stenosis, for example in atherosclerosis and restenosis after angioplasty. Experiments were performed to determine the effect of ISIS 11061 on proliferation of A-10 rat smooth muscle cells. Cells in culture were grown with and without ISIS 11061 (plus lipofectin) and cell proliferation was measured 24 and 48 hours after stimulation with fetal calf serum. ISIS 11061 (500 nM) was found to inhibit serum-stimulated cell growth in a dose- dependent manner with a maximal inhibition of 46% and 75% at 24 hours and 48 hours, respectively. An IC50 value of 200 nM was obtained for this compound. An unrelated control oligonucleotide had no effect at doses up to 500 nM.
Effects of antisense oligonucleotides targeted to c-raf on restenosis in rats A rat carotid artery injury model of angioplasty restenosis has been developed and has been used to evaluate the effects on restenosis of antisense oligonucleotides targeted to the c-myc oncogene. Bennett et al . , J". Clin . Invest . 1994, 93, 820-828. This model will be used to evaluate the effects of antisense oligonucleotides targeted to rat c-raf, particularly ISIS 11061, on restenosis. Following carotid artery injury with a balloon catheter, oligonucleotides are administered either by intravenous injection, continuous intravenous administration via Alzet pump, or direct administration to the carotid artery in a pluronic gel matrix as described by Bennett et al . After recovery, rats are sacrificed, carotid arteries are examined by microscopy and effects of treatment on luminal cross-sections are determined.
The invention is further illustrated by the following examples which are illustrations only and are not intended to limit the present invention to specific embodiments.
EXAMPLES
Example 1 Synthesis and Characterization of Oligonucleotides
Unmodified DNA oligonucleotides were synthesized on an automated DNA synthesizer (Applied Biosystems model 380B) using standard phosphoramidite chemistry with oxidation by iodine. Ī²-cyanoethyldiisopropyl phosphoramidites were purchased from Applied Biosystems (Foster City, CA) . For phosphorothioate oligonucleotides, the standard oxidation bottle was replaced by a 0.2 M solution of H-l, 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. 2 '-O-methyl phosphorothioate oligonucleotides were synthesized using 2'-O-methyl Ī²- cyanoethyldiisopropyl-phosphoramidites (Chemgenes, Needham MA) and the standard cycle for unmodified oligonucleotides, except the wait step after pulse delivery of tetrazole and base was increased to 360 seconds. The 3 '-base used to start the synthesis was a 2 ' -deoxyribonucleotide. 2'-0-propyl oligonucleotides were prepared by a slight modification of this procedure. 2'-fluoro phosphorothioate oligonucleotides were synthesized using 5' -dimethoxytrityl-3 ' -phosphoramidites and prepared as disclosed in U.S. patent application Serial No. 463,358, filed January 11, 1990, and 566,977, filed August 13, 1990, which are assigned to the same assignee as the instant application and which are incorporated by reference herein. The 2'-fluoro oligonucleotides were prepared using phosphoramidite chemistry and a slight modification of the standard DNA synthesis protocol: deprotection was effected using methanolic ammonia at room temperature. After cleavage from the controlled pore glass column
(Applied Biosystems) and deblocking in concentrated ammonium hydroxide at 55Ā°C for 18 hours, the oligonucleotides were purified by precipitation twice out of 0.5 M NaCl with 2.5 volumes ethanol. Analytical gel electrophoresis was accomplished in 20% acrylamide, 8 M urea, 45 mM Tris-borate buffer, pH 7.0. Oligodeoxynucleotides and their phosphorothioate analogs were judged from electrophoresis to be greater than 80% full length material .
Example 2 Northern blot analysis of inhibition of c-raf mRNA expression
The human urinary bladder cancer cell line T24 was obtained from the American Type Culture Collection (Rockville MD) . Cells were grown in McCoy's 5A medium with L-glutamine (Gibco BRL, Gaithersburg MD) , supplemented with 10% heat-inactivated fetal calf serum and 50 U/ml each of penicillin and streptomycin. Cells were seeded on 100 mm plates. When they reached 70% confluency, they were treated with oligonucleotide. Plates were washed with 10 ml prewarmed PBS and 5 ml of Opti-MEM reduced- serum medium containing 2.5 Ī¼l DOTMA. Oligonucleotide with lipofectin was then added to the desired concentration. After 4 hours of treatment, the medium was replaced with McCoy's medium. Cells were harvested 24 to 72 hours after oligonucleotide treatment and RNA was isolated using a standard CsCl purification method. Kingston, R.E., in Current Protocols in Molecular Biology, (F.M. Ausubel, R. Brent, R.E. Kingston, D.D. Moore, J.A. Smith, J.G. Seidman and K. Strahl, eds . ) , John Wiley and Sons, NY. Total RNA was isolated by centrifugation of cell lysates over a CsCl cushion. RNA samples were electrophoresed through 1.2% agarose-formaldehyde gels and transferred to hybridization membranes by capillary diffusion over a 12-14 hour period. The RNA was cross-linked to the membrane by exposure to UV light in a Stratalinker (Stratagene,
La Jolla, CA) and hybridized to random-primed 3 P-labeled c-raf cDNA probe (obtained from ATCC) or G3PDH probe as a control.
RNA was quantitated using a Phosphorimager (Molecular Dynamics,
Sunnyvale, CA) .
Example 3 Specific inhibition of c-raf kinase protein expression in T24 cells
T24 cells were treated with oligonucleotide (200 nM) and lipofectin at T=0 and T=24 hours. Protein extracts were prepared at T=48 hours, electrophoresed on acrylamide gels and analyzed by Western blot using polyclonal antibodies against c-raf (UBI,
Lake Placid, NY) or A-raf (Transduction Laboratories, Knoxville,
TN) . Radiolabeled secondary antibodies were used and raf protein was quantitated using a Phosphorimager (Molecular Dynamics, Sunnyvale CA) . Example 4 Antisense inhibition of cell proliferation
T24 cells were treated on day 0 for two hours with various concentrations of oligonucleotide and lipofectin (50 nM oligonucleotide in the presence of 2 Ī¼g/ml lipofectin; 100 nM oligonucleotide and 2Ī¼g/ml lipofectin; 250 nM oligonucleotide and 6 Ī¼g/ml lipofectin or 500 nM oligonucleotide and 10 Ī¼g/ml lipofectin) . On day 1, cells were treated for a second time at desired oligonucleotide concentration for two hours. On day 2, cells were counted.
Example 5 Effect of ISIS 5132 on T24 Human Bladder Carcinoma Tumor Xenografts in Nude Mice
5 x 10s T24 cells were implanted subcutaneously in the right inner thigh of nude mice. Oligonucleotides (ISIS 5132 and an unrelated control phosphorothioate oligonucleotide suspended in saline) were administered three times weekly beginning on day
4 after tumor cell inoculation. A saline-only control was also given. Oligonucleotides were given by intraperitoneal injection.
Oligonucleotide dosage was 25 mg/kg. Tumor size was measured and tumor volume was calculated on the eleventh, fifteenth and eighteenth treatment days.
Example 6 Effect of ISIS 5132 on MDA-MB 231 Human Breast Carcinoma Tumor Xenografts in Nude Mice
5 x 106 MDA-MB 231 cells were implanted subcutaneously in the right inner thigh of nude mice. Oligonucleotides (ISIS 5132 and an unrelated control phosphorothioate oligonucleotide suspended in saline) were administered once daily beginning on day 10 after tumor cell inoculation. A saline-only control was also given. Oligonucleotides were given by intravenous injection at a dosage of 0.6 mg/kg or 6.0 mg/kg. Tumor size was measured and tumor volume was calculated on days 10, 13, 16, 20, 23 and 27 following tumor cell inoculation.
For intraperitoneal oligonucleotide administration, oligonucleotides were administered once daily beginning on day 10 after tumor cell inoculation. A saline-only control was also given. Oligonucleotides were given by intraperitoneal injection at a dosage of 0.6 mg/kg or 6.0 mg/kg. Tumor size was measured and tumor volume was calculated on days 10, 13, 16, 20, 23 and 27 following tumor cell inoculation.
Example 7 Effect of ISIS 5132 on Colo 205 Human Colon Carcinoma Tumor Xenografts in Nude Mice 5 x 106 Colo 205 cells were implanted subcutaneously in the right inner thigh of nude mice. Oligonucleotides (ISIS 5132 and an unrelated control phosphorothioate oligonucleotide suspended in saline) were administered once per day beginning on day 5 after tumor cell inoculation. A saline-only control was also given. Oligonucleotides were given by intravenous injection. Oligonucleotide dosage was 6 mg/kg. Tumor size was measured and tumor volume was calculated on days 5, 8, 11, 14,
18, 22 and 25 after tumor inoculation.
Example 8 Diagnostic Assay for raf-associated Tumors Using Xenografts in Nude Mice
Tumors arising from raf expression are diagnosed and distinguished from other tumors using this assay. A biopsy sample of the tumor is treated, e.g. , with collagenase or trypsin or other standard methods, to dissociate the tumor mass. 5 x 106 tumor cells are implanted subcutaneously in the inner thighs of two or more nude mice. Antisense oligonucleotide (e.g. , ISIS 5132) suspended in saline is administered to one or more mice by intraperitoneal injection three times weekly beginning on day 4 after tumor cell inoculation. Saline only is given to a control mouse. Oligonucleotide dosage is 25 mg/kg. Tumor size is measured and tumor volume is calculated on the eleventh treatment day. Tumor volume of the oligonucleotide-treated mice is compared to that of the control mouse. The volume of raf- associated tumors in the treated mice are measurably smaller than tumors in the control mouse. Tumors arising from causes other than raf expression are not expected to respond to the oligonucleotides targeted to raf and, therefore, the tumor volumes of oligonucleotide- treated and control mice are equivalent . Example 9 Detection of raf expression
Oligonucleotides are radiolabeled after synthesis by 32P labeling at the 5' end with polynucleotide kinase. Sambrook et al . , Molecular Cloning. A Labora tory Manual , Cold Spring Harbor Laboratory Press, 1989, Volume 2, pg. 11.31-11.32. Radiolabeled oligonucleotides are contacted with tissue or cell samples suspected of raf expression, such as tumor biopsy samples or skin samples where psoriasis is suspected, under conditions in which specific hybridization can occur, and the sample is washed to remove unbound oligonucleotide. Radioactivity remaining in the sample indicates bound oligonucleotide and is quantitated using a scintillation counter or other routine means.
Radiolabeled oligonucleotides of the invention are also used in autoradiography. Tissue sections are treated with radiolabeled oligonucleotide and washed as described above, then exposed to photographic emulsion according to standard autoradiography procedures. The emulsion, when developed, yields an image of silver grains over the regions expressing raf . The extent of raf expression is determined by quantitation of the silver grains.
Analogous assays for fluorescent detection of raf expression use oligonucleotides of the invention which are labeled with fluorescein or other fluorescent tags . Labeled DNA oligonucleotides are synthesized on an automated DNA synthesizer (Applied Biosystems model 380B) using standard phosphoramidite chemistry with oxidation by iodine. Ī²-cyanoethyldiisopropyl phosphoramidites are purchased from Applied Biosystems (Foster City, CA) . Fluorescein-labeled amidites are purchased from Glen Research (Sterling VA) . Incubation of oligonucleotide and biological sample is carried out as described for radiolabeled oligonucleotides except that instead of a scintillation counter, a fluorimeter or fluorescence microscope is used to detect the fluorescence which indicates raf expression.
Example 10: A549 xenografts 5 x 106 A549 cells were implanted subcutaneously in the inner thigh of nude mice. Oligonucleotides (ISIS 5132 and a scrambled raf control phosphorothioate oligonucleotide, ISIS 10353) suspended in saline were administered once daily by intravenous injection at doses ranging from 0.006 to 6.0 mg/kg. Resulting tumors were measured on days 9, 12, 17 and 21 and tumor volumes were calculated.
Example 11: Effect of oligonucleotide on endogenous c-raf expression
Mice were treated by intraperitoneal injection at an oligonucleotide dose of 50 mg/kg on days 1, 2 and 3. On day 4 animals were sacrificed and organs removed for c-raf mRNA assay by Northern blot analysis. Four groups of animals were employed: 1) no oligonucleotide treatment (saline) ; 2) negative control oligonucleotide ISIS 1082 (targeted to herpes simplex virus; 3) negative control oligonucleotide 4189 (targeted to mouse protein kinase C-Ī±; 4) ISIS 11061 targeted to rodent c-raf.
Example 12: Cardiac allograft rejection model
Hearts were transplanted into the abdominal cavity of rats or mice (of a different strain from the donor) as primary vascuĀ¬ larized grafts essentially as described by Isobe et al . , Circulation 1991, 84, 1246-1255. Oligonucleotides were administered by continuous intravenous administration via a 7-day Alzet pump. Cardiac allograft survival was monitored by listening for the presence of a second heartbeat in the abdominal cavity.
Example 13: Proliferation assay using rat A-10 smooth muscle cells
A10 cells were plated into 96-well plates in Dulbecco's modified Eagle medium (DMEM) + 10% fetal calf serum and allowed to attach for 24 hours. Cells were made quiescent by the addition of DMEM + 0.2% dialyzed fetal calf serum for an additional 24 hours. During the last 4 hours of quiescence, cells were treated with ISIS 11061 + lipofectin (Gibco-BRL, Bethesda MD) in serum-free medium. Medium was then removed, replaced with fresh medium and the cells were stimulated with 10% fetal calf serum. The plates were the placed into the incubator and cell growth was evaluated by MTS conversion to formozan (Promega cell proliferation kit) at 24 and 48 hours after serum stimulation. A control oligonucleotide, ISIS 1082 (an unrelated oligonucleotide targeted to herpes simplex virus) , was also tested.
Example 14: Rat carotid artery restenosis model
This model has been described by Bennett et al . , J". Clin . Invest . 1994, 93, 820-828. Intimal hyperplasia is induced by balloon catheter dilatation of the carotid artery of the rat. Rats are anesthetized and common carotid artery injury is induced by passage of a balloon embolectomy catheter distended with 20 ml of saline. Oligonucleotides are applied to the adventitial surface of the arterial wall in a pluronic gel solution. Oligonucleotides are dissolved in a 0.25% pluronic gel solution at 4Ā°C (F127, BASF Corp.) at the desired dose. 100 Ī¼l of the gel solution is applied to the distal third of the common carotid artery immediately after injury. Control rats are treated similarly with gel containing control oligonucleotide or no oligonucleotide. The neck wounds are closed and the animals allowed to recover. 14 days later, rats are sacrificed, exsanguinated and the carotid arteries fixed in situ by perfusion with paraformaldehyde and glutaraldehyde, excised and processed for microscopy. Cross-sections of the arteries are calculated.
In an alternative to the pluronic gel administration procedure, rats are treated by intravenous injection or continuous intravenous infusion (via Alzet pump) of oligonucleotide.
SEQUENCE LISTING
(1) GENERAL INFORMATION:
(i) APPLICANT: Monia, Brett P. and Boggs, Russell T.
(ii) TITLE OF INVENTION: Antisense Oligonucleotide Modulati of raf Gene Expression
(iii) NUMBER OF SEQUENCES: 54
(iv) CORRESPONDENCE ADDRESS:
(A) ADDRESSEE: Law Offices of Jane Massey Licata
(B) STREET: 210 Lake Drive East, Suite 201
(C) CITY: Cherry Hill
(D) STATE: NJ
(E) COUNTRY: USA
(F) ZIP: 08002
(v) COMPUTER READABLE FORM:
(A) MEDIUM TYPE: DISKETTE, 3.5 INCH, 1.44 Mb STORAGE
(B) COMPUTER: IBM PS/2
(C) OPERATING SYSTEM: PC-DOS
(D) SOFTWARE: WORDPERFECT 5.1 (vi) CURRENT APPLICATION DATA:
(A) APPLICATION NUMBER: n/a
(B) FILING DATE: Herewith
(C) CLASSIFICATION: (vii) PRIOR APPLICATION DATA:
(A) APPLICATION NUMBER: 08/250,856
(B) FILING DATE: May 31, 1995 (viii) ATTORNEY/AGENT INFORMATION:
(A) NAME: Jane Massey Licata
(B) REGISTRATION NUMBER: 32,257 (C) REFERENCE/DOCKET NUMBER: ISPH-0135 (ix) TELECOMMUNICATION INFORMATION:
(A) TELEPHONE: (609) 779-2400
(B) TELEFAX: (609) 779-8488 (2) INFORMATION FOR SEQ ID NO: 1:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS : Single
(D) TOPOLOGY: Linear (iv) ANTI-SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 1: TGAAGGTGAG CTGGAGCCAT (20) (2) INFORMATION FOR SEQ ID NO: 2: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI-SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 2: GCTCCATTGA TGCAGCTTAA (20) (2) INFORMATION FOR SEQ ID NO: 3: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear ( iv ) ANTI - SENSE : Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 3: CCCTGTATGT GCTCCATTGA (20)
(2) INFORMATION FOR SEQ ID NO: 4: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI-SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 4: GGTGCAAAGT CAACTAGAAG (20) (2) INFORMATION FOR SEQ ID NO: 5: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI-SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 5: ATTCTTAAAC CTGAGGGAGC (20) (2) INFORMATION FOR SEQ ID NO: 6: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI-SENSE: Yes (xi) SEQUENCE DESCRIPTION: SEQ ID NO: 6: GATGCAGCTT AAACAATTCT (20) (2) INFORMATION FOR SEQ ID NO: 7: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI-SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 7: CAGCACTGCA AATGGCTTCC (20) (2) INFORMATION FOR SEQ ID NO: 8: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI-SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 8: TCCCGCCTGT GACATGCATT (20) (2) INFORMATION FOR SEQ ID NO: 9: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI-SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 9: GCCGAGTGCC TTGCCTGGAA (20) (2) INFORMATION FOR SEQ ID NO: 10: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI-SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 10: AGAGATGCAG CTGGAGCCAT (20) (2) INFORMATION FOR SEQ ID NO: 11: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI-SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 11: AGGTGAAGGC CTGGAGCCAT (20) (2) INFORMATION FOR SEQ ID NO: 12: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI-SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 12: GTCTGGCGCT GCACCACTCT (20) (2) INFORMATION FOR SEQ ID NO: 13: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI-SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 13: CTGATTTCCA AAATCCCATG (20) (2) INFORMATION FOR SEQ ID NO: 14: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI-SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 14: CTGGGCTGTT TGGTGCCTTA (20) (2) INFORMATION FOR SEQ ID NO: 15: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI-SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 15: TCAGGGCTGG ACTGCCTGCT (20) (2) INFORMATION FOR SEQ ID NO: 16: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI-SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 16: GGTGAGGGAG CGGGAGGCGG (20) (2) INFORMATION FOR SEQ ID NO: 17: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI-SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 17: CGCTCCTCCT CCCCGCGGCG (20) (2) INFORMATION FOR SEQ ID NO: 18: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI-SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 18: TTCGGCGGCA GCTTCTCGCC (20) (2) INFORMATION FOR SEQ ID NO: 19: (i) SEQUENCE CHARACTERISTICS: (A) LENGTH : 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI-SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 19: GCCGCCCCAA CGTCCTGTCG (20) (2) INFORMATION FOR SEQ ID NO: 20: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI-SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 20: TCCTCCTCCC CGCGGCGGGT (20) (2) INFORMATION FOR SEQ ID NO: 21: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI-SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 21: CTCGCCCGCT CCTCCTCCCC (20) (2) INFORMATION FOR SEQ ID NO: 22: (i) SEQUENCE CHARACTERISTICS: (A) LENGTH: 20 (B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI-SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 22: CTGGCTTCTC CTCCTCCCCT (20) (2) INFORMATION FOR SEQ ID NO: 23: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI-SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 23: CGGGAGGCGG TCACATTCGG (20) (2) INFORMATION FOR SEQ ID NO: 24: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI-SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 24: TCTGGCGCTG CACCACTCTC (20) (2) INFORMATION FOR SEQ ID NO: 25: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid (C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI-SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 25: TTCTCGCCCG CTCCTCCTCC (20) (2) INFORMATION FOR SEQ ID NO: 26: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI-SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 26: TTCTCCTCCT CCCCTGGCAG (20) (2) INFORMATION FOR SEQ ID NO: 27: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI-SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 27: CCTGCTGGCT TCTCCTCCTC (20) (2) INFORMATION FOR SEQ ID NO: 28: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single (D) TOPOLOGY: Linear (iv) ANTI-SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 28: GTCAAGATGG GCTGAGGTGG (20) (2) INFORMATION FOR SEQ ID NO: 29: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI-SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 29: CCATCCCGGA CAGTCACCAC (20) (2) INFORMATION FOR SEQ ID NO: 30: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI-SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 30: ATGAGCTCCT CGCCATCCAG (20) (2) INFORMATION FOR SEQ ID NO: 31: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear ( iv ) ANTI - SENSE : Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 31: AATGCTGGTG GAACTTGTAG (20) (2) INFORMATION FOR SEQ ID NO: 32: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI-SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 32: CCGGTACCCC AGGTTCTTCA (20) (2) INFORMATION FOR SEQ ID NO: 33: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI-SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 33: CTGGGCAGTC TGCCGGGCCA (20) (2) INFORMATION FOR SEQ ID NO: 34: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI-SENSE: Yes (xi) SEQUENCE DESCRIPTION: SEQ ID NO: 34: CACCTCAGCT GCCATCCACA (20) (2) INFORMATION FOR SEQ ID NO: 35: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI-SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 35: GAGATTTTGC TGAGGTCCGG (20) (2) INFORMATION FOR SEQ ID NO: 36: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI-SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 36: GCACTCCGCT CAATCTTGGG (20) (2) INFORMATION FOR SEQ ID NO: 37: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI-SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 37: CTAAGGCACA AGGCGGGCTG (20) (2) INFORMATION FOR SEQ ID NO: 38: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI-SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 38: ACGAACATTG ATTGGCTGGT (20) (2) INFORMATION FOR SEQ ID NO: 39: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI-SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 39: GTATCCCCAA AGCCAAGAGG (20) (2) INFORMATION FOR SEQ ID NO: 40: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI-SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 40: CATCAGGGCA GAGACGAACA (20) (2) INFORMATION FOR SEQ ID NO: 41: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI-SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 41: GGAACATCTG GAATTTGGTC (20) (2) INFORMATION FOR SEQ ID NO: 42: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI-SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:42 : GATTCACTGT GACTTCGAAT (20) (2) INFORMATION FOR SEQ ID NO: 43: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI-SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 43: GCTTCCATTT CCAGGGCAGG (20) (2) INFORMATION FOR SEQ ID NO: 44: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI-SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 44: AAGAAGGCAA TATGAAGTTA (20) (2) INFORMATION FOR SEQ ID NO: 45: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI-SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 45: GTGGTGCCTG CTGACTCTTC (20) (2) INFORMATION FOR SEQ ID NO: 46: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI-SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 46: CTGGTGGCCT AAGAACAGCT (20) (2) INFORMATION FOR SEQ ID NO: 47: (i) SEQUENCE CHARACTERISTICS: (A) LENGTH : 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI-SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 47: GTATGTGCTC CATTGATGCA (20) (2) INFORMATION FOR SEQ ID NO: 48: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI-SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 48: TCCCTGTATG TGCTCCATTG (20) (2) INFORMATION FOR SEQ ID NO: 49: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI-SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 49: ATACTTATAC CTGAGGGAGC (20) (2) INFORMATION FOR SEQ ID NO: 50: (i) SEQUENCE CHARACTERISTICS: (A) LENGTH: 20 (B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI-SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 50: ATGCATTCTG CCCCCAAGGA (20) (2) INFORMATION FOR SEQ ID NO: 51: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI-SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 51: GACTTGTATA CCTCTGGAGC (20) (2) INFORMATION FOR SEQ ID NO: 52: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI-SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 52: ACTGGCACTG CACCACTGTC (20) (2) INFORMATION FOR SEQ ID NO: 53: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid (C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI-SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 53: AAGTTCTGTA GTACCAAAGC (20) (2) INFORMATION FOR SEQ ID NO: 54: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20
(B) TYPE: Nucleic Acid
(C) STRANDEDNESS: Single
(D) TOPOLOGY: Linear (iv) ANTI-SENSE: Yes
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 54: CTCCTGGAAG ACAGATTCAG (20)

Claims

What Is Claimed Is:
I. An oligonucleotide 8 to 50 nucleotides in length which is targeted to mRNA encoding human raf and which is capable of inhibiting raf expression.
2. The oligonucleotide of claim 1 which is targeted to mRNA encoding human A-raf.
3. The oligonucleotide of claim 2 having SEQ ID NO: 29, 37 or 40.
4. The oligonucleotide of claim 1 which is targeted to mRNA encoding human c-raf.
5. The oligonucleotide of claim 4 which is targeted to a translation initiation site, 3' untranslated region or 5' untranslated region of mRNA encoding human c-raf.
6. The oligonucleotide of claim 1 which has at least one phosphorothioate linkage.
7. The oligonucleotide of claim 1 wherein at least one of the nucleotide units of the oligonucleotide is modified at the 2' position of the sugar moiety.
8. The oligonucleotide of claim 7 wherein said modification at the 2' position of the sugar moiety is a 2'-0- alkyl or a 2'-fluoro modification.
9. The oligonucleotide of claim 1 in a pharmaceutically acceptable carrier.
10. The oligonucleotide of claim 5 comprising SEQ ID NO: 2, 6, 8, 12, 17, 20, 21, 22, 23, 24, 25, 26 or 27.
II. An oligonucleotide having SEQ ID NO: 8.
12. The oligonucleotide of claim 11 comprising at least one 2'-O-methyl modification.
13. A chimeric oligonucleotide 8 to 50 nucleotides in length which is targeted to mRNA encoding human raf which contains a first region having at least one nucleotide which is modified to enhance target affinity and a second region which is a substrate for RNAse H, said chimeric oligonucleotide being capable of inhibiting raf expression.
14. The oligonucleotide of claim 13 wherein the nucleotide which is modified to enhance target affinity is modified at the
2' position of the sugar moiety.
15. The oligonucleotide of claim 14 wherein the modification at the 2' position of the sugar moiety is a 2'-0- alkyl or a 2'-fluoro modification.
16. The oligonucleotide of claim 13 wherein the region which is a substrate for RNAse H comprises at least one 2'- deoxynucleotide.
17. The oligonucleotide of claim 13 which has at least one phosphorothioate linkage.
18. The oligonucleotide of claim 13 which is targeted to a translation initiation site, 3' untranslated region or 5' untranslated region of mRNA encoding human c-raf.
19. The oligonucleotide of claim 13 in a pharmaceutically acceptable carrier.
20. The oligonucleotide of claim 17 comprising SEQ ID NO: 8, 21, 24, 25, 26 or 27.
21. A chimeric oligonucleotide having SEQ ID NO: 8 which contains a first region having at least one nucleotide which is modified to enhance target affinity and a second region which is a substrate for RNAse H, said chimeric oligonucleotide being capable of inhibiting raf expression.
22. A method of inhibiting the expression of human raf comprising contacting tissues or cells which express human raf with an oligonucleotide 8 to 50 nucleotides in length which is targeted to mRNA encoding human raf and which is capable of inhibiting raf expression.
23. The method of claim 22 wherein said oligonucleotide is targeted to mRNA encoding human A-raf.
24. The method of claim 22 wherein said oligonucleotide is targeted to mRNA encoding human c-raf.
25. The method of claim 24 wherein said oligonucleotide is targeted to a translation initiation site, 3' untranslated region or 5' untranslated region of mRNA encoding human c-raf.
26. The method of claim 25 wherein said oligonucleotide comprises SEQ ID NO: 2, 6, 8, 12, 17, 20, 21, 22, 23, 24, 25, 26 or 27.
27. The method of claim 22 wherein said expression of human raf is abnormal expression.
28. The method of claim 22 wherein said oligonucleotide is a chimeric oligonucleotide.
29. The method of claim 28 wherein said chimeric oligonucleotide comprises SEQ ID NO: 8, 21, 24, 25, 26 or 27.
30. The method of claim 22 wherein said oligonucleotide has at least one phosphorothioate linkage.
31. A method of inhibiting the expression of human raf comprising contacting tissues or cells which express human raf with an oligonucleotide having SEQ ID NO: 8.
32. A method of inhibiting hyperproliferation of cells comprising contacting hyperproliferating cells with an oligonucleotide 8 to 50 nucleotides in length which is targeted to mRNA encoding human raf and which is capable of inhibiting raf expression.
33. The method of claim 32 wherein said oligonucleotide is targeted to mRNA encoding human A-raf.
34. The method of claim 32 wherein said oligonucleotide is targeted to mRNA encoding human c-raf.
35. The method of claim 34 wherein said oligonucleotide is targeted to a translation initiation site, 3' untranslated region or 5' untranslated region of mRNA encoding human c-raf.
36. The method of claim 35 wherein said oligonucleotide comprises SEQ ID NO: 2, 6, 8, 12, 17, 20, 21, 22, 23, 24, 25, 26 or 27.
37. The method of claim 32 wherein said oligonucleotide is a chimeric oligonucleotide.
38. The method of claim 37 wherein said chimeric oligonucleotide comprises SEQ ID NO: 8, 21, 24, 25, 26 or 27.
39. The method of claim 32 wherein said oligonucleotide has at least one phosphorothioate linkage.
40. A method of inhibiting hyperproliferation of cells comprising contacting hyperproliferating cells with an oligonucleotide having SEQ ID NO: 8.
41. A method of treating an abnormal proliferative condition comprising contacting a subject, or cells, tissues or a bodily fluid of said subject, suspected of having an abnormal proliferative condition with an oligonucleotide 8 to 50 nucleotides in length which is targeted to mRNA encoding human raf and which is capable of inhibiting raf expression.
42. The method of claim 41 wherein the condition is a hyperproliferative disorder.
43. The method of claim 42 wherein the hyperproliferative disorder is cancer, restenosis, psoriasis or a disorder characterized by T-cell activation and growth.
44. The method of claim 41 wherein said oligonucleotide is targeted to mRNA encoding human A-raf.
45. The method of claim 41 wherein said oligonucleotide is targeted to mRNA encoding human c-raf.
46. The method of claim 45 wherein said oligonucleotide is targeted to the 3' untranslated region or the 5' untranslated region of mRNA encoding human c-raf.
47. The method of claim 46 wherein said oligonucleotide comprises SEQ ID NO: 2, 6, 8, 12, 17, 20, 21, 22, 23, 24, 25,
26 or 27.
48. The method of claim 41 wherein said oligonucleotide is a chimeric oligonucleotide.
49. The method of claim 48 wherein said chimeric oligonucleotide comprises SEQ ID NO: 8, 21, 24, 25, 26 or 27.
50. The method of claim 41 wherein said oligonucleotide has at least one phosphorothioate linkage.
51. A method of treating an abnormal proliferative condition comprising contacting a subject, or cells, tissues or a bodily fluid of said subject, suspected of having an abnormal proliferative condition with an oligonucleotide having SEQ ID NO: 8.
PCT/US1995/007111 1994-05-31 1995-05-31 ANTISENSE OLIGONUCLEOTIDE MODULATION OF raf GENE EXPRESSION WO1995032987A1 (en)

Priority Applications (19)

Application Number Priority Date Filing Date Title
DE69533697T DE69533697T2 (en) 1994-05-31 1995-05-31 ANTISENSE OLIGONUCLEOTIDE MODULATION OF Raf GENE EXPRESSION
AT95922217T ATE280772T1 (en) 1994-05-31 1995-05-31 ANTISENSE OLIGONUCLEOTIDE MODULATION OF RAF GENE EXPRESSION
CA002191795A CA2191795C (en) 1994-05-31 1995-05-31 Antisense oligonucleotide modulation of raf gene expression
EP95922217A EP0763052B1 (en) 1994-05-31 1995-05-31 ANTISENSE OLIGONUCLEOTIDE MODULATION OF raf GENE EXPRESSION
JP50125796A JP3231779B2 (en) 1994-05-31 1995-05-31 Antisense oligonucleotide regulation of raf gene expression
AU26982/95A AU689267B2 (en) 1994-05-31 1995-05-31 Antisense oligonucleotide modulation of RAF gene expression
KR1019960706806A KR100211178B1 (en) 1994-05-31 1995-05-31 Antisense oligonucleotide modulation of raf gene expression
US08/756,806 US5952229A (en) 1994-05-31 1996-11-26 Antisense oligonucleotide modulation of raf gene expression
NO965080A NO965080L (en) 1994-05-31 1996-11-28 Antigen oligonucleotide modulation of gene expression
FI964792A FI964792A (en) 1994-05-31 1996-11-29 altering raf gene expression with antisense oligonucleotides
US09/462,261 US6391636B1 (en) 1994-05-31 1998-07-06 Antisense oligonucleotide modulation of raf gene expression
US09/143,214 US6090626A (en) 1994-05-31 1998-08-28 Antisense oligonucleotide modulation of raf gene expression
US09/506,073 US6410518B1 (en) 1994-05-31 2000-02-18 Antisense oligonucleotide inhibition of raf gene expression
US10/057,550 US6806258B2 (en) 1994-05-31 2002-01-25 Antisense oligonucleotide modulation of raf gene expression
US10/173,225 US20030119769A1 (en) 1994-05-31 2002-06-14 Antisense oligonucleotide modulation of raf gene expression
US11/364,481 US20060142236A1 (en) 1994-05-31 2006-02-28 Antisense oligonucleotide modulation of raf gene expression
US13/185,406 US20120149755A1 (en) 1994-05-31 2011-07-18 Antisense oligonucleotide modulation of raf gene expression
US13/678,432 US20130079387A1 (en) 1994-05-31 2012-11-15 Antisense oligonucleotide modulation of raf gene expression
US13/956,213 US20140187601A1 (en) 1994-05-31 2013-07-31 Antisense oligonucleotide modulation of raf gene expression

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US08/250,856 US5563255A (en) 1994-05-31 1994-05-31 Antisense oligonucleotide modulation of raf gene expression
US08/250,856 1994-05-31

Related Parent Applications (2)

Application Number Title Priority Date Filing Date
US08/250,856 Continuation-In-Part US5563255A (en) 1994-05-31 1994-05-31 Antisense oligonucleotide modulation of raf gene expression
US08/250,856 Continuation US5563255A (en) 1994-05-31 1994-05-31 Antisense oligonucleotide modulation of raf gene expression

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US08/756,806 Continuation-In-Part US5952229A (en) 1994-05-31 1996-11-26 Antisense oligonucleotide modulation of raf gene expression
US08/756,806 Continuation US5952229A (en) 1994-05-31 1996-11-26 Antisense oligonucleotide modulation of raf gene expression

Publications (1)

Publication Number Publication Date
WO1995032987A1 true WO1995032987A1 (en) 1995-12-07

Family

ID=22949424

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1995/007111 WO1995032987A1 (en) 1994-05-31 1995-05-31 ANTISENSE OLIGONUCLEOTIDE MODULATION OF raf GENE EXPRESSION

Country Status (12)

Country Link
US (4) US5563255A (en)
EP (1) EP0763052B1 (en)
JP (2) JP3231779B2 (en)
AT (1) ATE280772T1 (en)
AU (1) AU689267B2 (en)
CA (1) CA2191795C (en)
DE (1) DE69533697T2 (en)
FI (1) FI964792A (en)
HU (1) HUT76675A (en)
IL (1) IL113804A (en)
NO (1) NO965080L (en)
WO (1) WO1995032987A1 (en)

Cited By (61)

* Cited by examiner, ā€  Cited by third party
Publication number Priority date Publication date Assignee Title
WO1996039415A1 (en) * 1995-06-05 1996-12-12 Isis Pharmaceuticals, Inc. ANTISENSE OLIGONUCLEOTIDE MODULATION OF raf GENE EXPRESSION
WO1997004787A1 (en) * 1995-08-01 1997-02-13 Novartis Ag Liposomal oligonucleotide compositions
WO1997010829A1 (en) * 1995-09-19 1997-03-27 Novartis Ag Pharmaceutical compositions containing an oligonucleotide and an anionic polysaccharide
US5744460A (en) * 1996-03-07 1998-04-28 Novartis Corporation Combination for treatment of proliferative diseases
WO1998043095A1 (en) * 1997-03-21 1998-10-01 Georgetown University Liposomes containing oligonucleotides
EP0906099A1 (en) * 1996-04-03 1999-04-07 Merck & Co., Inc. A method of treating cancer
US6027892A (en) * 1996-12-30 2000-02-22 Chang Esther H Compositions and methods for reducing radiation and drug resistance in cells
WO2000018781A1 (en) 1998-09-29 2000-04-06 Isis Pharmaceuticals, Inc. Antisense modulation of survivin expression
WO2000056342A2 (en) * 1999-03-19 2000-09-28 Assistance Publique-Hopitaux De Paris Use of stabilised oligonucleotides for preparing a medicine with antitumour activity
DE10020138A1 (en) * 2000-04-14 2001-10-31 Ulf R Rapp Nucleic acid coding for at least one partial sequence with a MEKK1 binding site
US6562960B1 (en) * 1996-03-05 2003-05-13 Isis Pharmaceuticals, Inc. Oligonucleotide analogues
US6803360B1 (en) 1996-12-30 2004-10-12 Georgetown University Compositions and methods for reducing radiation and drug resistance in cells
WO2006123182A2 (en) 2005-05-17 2006-11-23 Merck Sharp & Dohme Limited Cyclohexyl sulphones for treatment of cancer
WO2007093827A1 (en) 2006-02-15 2007-08-23 Istituto Di Ricerche Di Biologia Molecolare P. Angeletti Spa Thiophene and thiazole substituted trifluoroethanone derivatives as histone deacetylase (hdac) inhibitors
US7262173B2 (en) 1997-03-21 2007-08-28 Georgetown University Chemosensitizing with liposomes containing oligonucleotides
WO2008106692A1 (en) 2007-03-01 2008-09-04 Novartis Vaccines And Diagnostics, Inc. Pim kinase inhibitors and methods of their use
WO2008144062A1 (en) 2007-05-21 2008-11-27 Novartis Ag Csf-1r inhibitors, compositions, and methods of use
WO2009002495A1 (en) 2007-06-27 2008-12-31 Merck & Co., Inc. 4-carboxybenzylamino derivatives as histone deacetylase inhibitors
US7553854B2 (en) 2006-04-19 2009-06-30 Novartis Vaccines And Diagnostics, Inc. 6-O-substituted benzoxazole and benzothiazole compounds and methods of inhibiting CSF-1R signaling
WO2010114780A1 (en) 2009-04-01 2010-10-07 Merck Sharp & Dohme Corp. Inhibitors of akt activity
WO2011046771A1 (en) 2009-10-14 2011-04-21 Schering Corporation SUBSTITUTED PIPERIDINES THAT INCREASE p53 ACTIVITY AND THE USES THEREOF
EP2336120A1 (en) 2007-01-10 2011-06-22 Istituto di ricerche di Biologia Molecolare P. Angeletti S.R.L. Combinations containing amide substituted indazoles as poly(ADP-ribose)polymerase (PARP) inhibitors
WO2011115725A2 (en) 2010-03-16 2011-09-22 Dana-Farber Cancer Institute, Inc. Indazole compounds and their uses
WO2011163330A1 (en) 2010-06-24 2011-12-29 Merck Sharp & Dohme Corp. Novel heterocyclic compounds as erk inhibitors
WO2012018754A2 (en) 2010-08-02 2012-02-09 Merck Sharp & Dohme Corp. RNA INTERFERENCE MEDIATED INHIBITION OF CATENIN (CADHERIN-ASSOCIATED PROTEIN), BETA 1 (CTNNB1) GENE EXPRESSION USING SHORT INTERFERING NUCLEIC ACID (siNA)
WO2012024170A2 (en) 2010-08-17 2012-02-23 Merck Sharp & Dohme Corp. RNA INTERFERENCE MEDIATED INHIBITION OF HEPATITIS B VIRUS (HBV) GENE EXPRESSION USING SHORT INTERFERING NUCLEIC ACID (siNA)
WO2012027236A1 (en) 2010-08-23 2012-03-01 Schering Corporation NOVEL PYRAZOLO[1,5-a]PYRIMIDINE DERIVATIVES AS mTOR INHIBITORS
WO2012030685A2 (en) 2010-09-01 2012-03-08 Schering Corporation Indazole derivatives useful as erk inhibitors
WO2012036997A1 (en) 2010-09-16 2012-03-22 Schering Corporation Fused pyrazole derivatives as novel erk inhibitors
WO2012058210A1 (en) 2010-10-29 2012-05-03 Merck Sharp & Dohme Corp. RNA INTERFERENCE MEDIATED INHIBITION OF GENE EXPRESSION USING SHORT INTERFERING NUCLEIC ACIDS (siNA)
WO2012087772A1 (en) 2010-12-21 2012-06-28 Schering Corporation Indazole derivatives useful as erk inhibitors
WO2012145471A1 (en) 2011-04-21 2012-10-26 Merck Sharp & Dohme Corp. Insulin-like growth factor-1 receptor inhibitors
WO2013063214A1 (en) 2011-10-27 2013-05-02 Merck Sharp & Dohme Corp. Novel compounds that are erk inhibitors
WO2013165816A2 (en) 2012-05-02 2013-11-07 Merck Sharp & Dohme Corp. SHORT INTERFERING NUCLEIC ACID (siNA) COMPOSITIONS
EP2698157A1 (en) 2006-09-22 2014-02-19 Merck Sharp & Dohme Corp. Method of treatment using fatty acid synthesis inhibitors
WO2014052563A2 (en) 2012-09-28 2014-04-03 Merck Sharp & Dohme Corp. Novel compounds that are erk inhibitors
WO2014085216A1 (en) 2012-11-28 2014-06-05 Merck Sharp & Dohme Corp. Compositions and methods for treating cancer
WO2014100065A1 (en) 2012-12-20 2014-06-26 Merck Sharp & Dohme Corp. Substituted imidazopyridines as hdm2 inhibitors
US8765747B2 (en) 2009-06-12 2014-07-01 Dana-Farber Cancer Institute, Inc. Fused 2-aminothiazole compounds
WO2014120748A1 (en) 2013-01-30 2014-08-07 Merck Sharp & Dohme Corp. 2,6,7,8 substituted purines as hdm2 inhibitors
WO2015034925A1 (en) 2013-09-03 2015-03-12 Moderna Therapeutics, Inc. Circular polynucleotides
WO2016020864A1 (en) 2014-08-06 2016-02-11 Novartis Ag Protein kinase c inhibitors and methods of their use
US9758522B2 (en) 2012-10-19 2017-09-12 Dana-Farber Cancer Institute, Inc. Hydrophobically tagged small molecules as inducers of protein degradation
US9862688B2 (en) 2014-04-23 2018-01-09 Dana-Farber Cancer Institute, Inc. Hydrophobically tagged janus kinase inhibitors and uses thereof
WO2018058022A1 (en) 2016-09-26 2018-03-29 Merck Sharp & Dohme Corp. Anti-cd27 antibodies
US10000483B2 (en) 2012-10-19 2018-06-19 Dana-Farber Cancer Institute, Inc. Bone marrow on X chromosome kinase (BMX) inhibitors and uses thereof
US10017477B2 (en) 2014-04-23 2018-07-10 Dana-Farber Cancer Institute, Inc. Janus kinase inhibitors and uses thereof
WO2018190719A2 (en) 2017-04-13 2018-10-18 Aduro Biotech Holdings, Europe B.V. Anti-sirp alpha antibodies
US10112927B2 (en) 2012-10-18 2018-10-30 Dana-Farber Cancer Institute, Inc. Inhibitors of cyclin-dependent kinase 7 (CDK7)
US10144730B2 (en) 2011-11-17 2018-12-04 Dana-Farber Cancer Institute, Inc. Inhibitors of c-Jun-N-terminal kinase (JNK)
WO2019094311A1 (en) 2017-11-08 2019-05-16 Merck Sharp & Dohme Corp. Prmt5 inhibitors
WO2019152642A1 (en) 2018-02-01 2019-08-08 Merck Sharp & Dohme Corp. Anti-pd-1/lag3 bispecific antibodies
US10550121B2 (en) 2015-03-27 2020-02-04 Dana-Farber Cancer Institute, Inc. Inhibitors of cyclin-dependent kinases
WO2020033284A1 (en) 2018-08-07 2020-02-13 Merck Sharp & Dohme Corp. Prmt5 inhibitors
WO2020033282A1 (en) 2018-08-07 2020-02-13 Merck Sharp & Dohme Corp. Prmt5 inhibitors
US10702527B2 (en) 2015-06-12 2020-07-07 Dana-Farber Cancer Institute, Inc. Combination therapy of transcription inhibitors and kinase inhibitors
US10870651B2 (en) 2014-12-23 2020-12-22 Dana-Farber Cancer Institute, Inc. Inhibitors of cyclin-dependent kinase 7 (CDK7)
US11040957B2 (en) 2013-10-18 2021-06-22 Dana-Farber Cancer Institute, Inc. Heteroaromatic compounds useful for the treatment of proliferative diseases
US11096950B2 (en) 2006-11-01 2021-08-24 Barbara Brooke Jennings Compounds, methods, and treatments for abnormal signaling pathways for prenatal and postnatal development
US11142507B2 (en) 2015-09-09 2021-10-12 Dana-Farber Cancer Institute, Inc. Inhibitors of cyclin-dependent kinases
US11826365B2 (en) 2009-12-29 2023-11-28 Dana-Farber Cancer Institute, Inc. Type II raf kinase inhibitors

Families Citing this family (39)

* Cited by examiner, ā€  Cited by third party
Publication number Priority date Publication date Assignee Title
US6465188B1 (en) * 1990-06-11 2002-10-15 Gilead Sciences, Inc. Nucleic acid ligand complexes
KR100211178B1 (en) * 1994-05-31 1999-07-15 ķŒŒģƒ¬ ė¹„. ė¦°ė„¤ Antisense oligonucleotide modulation of raf gene expression
US6090626A (en) * 1994-05-31 2000-07-18 Isis Pharmaceuticals Inc. Antisense oligonucleotide modulation of raf gene expression
US6358932B1 (en) * 1994-05-31 2002-03-19 Isis Pharmaceticals, Inc. Antisense oligonucleotide inhibition of raf gene expression
US6391636B1 (en) 1994-05-31 2002-05-21 Isis Pharmaceuticals, Inc. Antisense oligonucleotide modulation of raf gene expression
US6410518B1 (en) * 1994-05-31 2002-06-25 Isis Pharmaceuticals, Inc. Antisense oligonucleotide inhibition of raf gene expression
US5981731A (en) * 1994-05-31 1999-11-09 Isis Pharmaceuticals, Inc. Antisense oligonucleotide modulation of B-raf gene expression
US6608035B1 (en) * 1994-10-25 2003-08-19 Hybridon, Inc. Method of down-regulating gene expression
US8071737B2 (en) * 1995-05-04 2011-12-06 Glead Sciences, Inc. Nucleic acid ligand complexes
US6699843B2 (en) * 1995-06-07 2004-03-02 Gilead Sciences, Inc. Method for treatment of tumors using nucleic acid ligands to PDGF
US6229002B1 (en) * 1995-06-07 2001-05-08 Nexstar Pharmaceuticlas, Inc. Platelet derived growth factor (PDGF) nucleic acid ligand complexes
US6127533A (en) 1997-02-14 2000-10-03 Isis Pharmaceuticals, Inc. 2'-O-aminooxy-modified oligonucleotides
US6172209B1 (en) 1997-02-14 2001-01-09 Isis Pharmaceuticals Inc. Aminooxy-modified oligonucleotides and methods for making same
US6576752B1 (en) 1997-02-14 2003-06-10 Isis Pharmaceuticals, Inc. Aminooxy functionalized oligomers
AU6578298A (en) 1997-03-21 1998-10-20 President And Fellows Of Harvard College Antisense inhibition of angiogenin expression
US20030036516A1 (en) * 1997-09-10 2003-02-20 Sudhir Agrawal Method for using oligonucleotides having modified cpg dinucleotides
US6492111B1 (en) 1998-11-25 2002-12-10 Isis Pharmaceuticals, Inc. In situ binary synthesis of biologically effective molecules
US6114517A (en) * 1998-12-10 2000-09-05 Isis Pharmaceuticals Inc. Methods of modulating tumor necrosis factor Ī±-induced expression of cell adhesion molecules
WO2000045167A2 (en) * 1999-01-29 2000-08-03 Avi Biopharma, Inc. Non-invasive method for detecting target rna
CA2850318A1 (en) * 1999-02-26 2000-08-31 The University Of British Columbia Trpm-2 antisense therapy
US6147200A (en) * 1999-08-19 2000-11-14 Isis Pharmaceuticals, Inc. 2'-O-acetamido modified monomers and oligomers
US6677445B1 (en) 1999-08-27 2004-01-13 Chiron Corporation Chimeric antisense oligonucleotides and cell transfecting formulations thereof
US7569551B2 (en) * 2000-02-25 2009-08-04 The University Of British Columbia Chemo- and radiation-sensitization of cancer by antisense TRPM-2 oligodeoxynucleotides
DK1295611T3 (en) * 2000-06-20 2010-10-04 Dainippon Sumitomo Pharma Co Oligonucleotide Transforming Compounds
US6867349B2 (en) * 2000-07-31 2005-03-15 Regents Of The University Of Minnesota Inhibition of gene expression using polynucleotide analogues
WO2002085308A2 (en) * 2001-04-24 2002-10-31 Epigenesis Pharmaceuticals, Inc. Antisense and anti-inflammatory based compositions to treat respiratory disorders
US8742091B2 (en) * 2001-06-20 2014-06-03 Dainippon Sumitomo Pharma Co., Ltd. Method of promoting nucleic acid transfer
EP1552002A4 (en) * 2002-06-18 2006-02-08 Archemix Corp Aptamer-toxin molecules and methods for using same
US20040249130A1 (en) * 2002-06-18 2004-12-09 Martin Stanton Aptamer-toxin molecules and methods for using same
US8853376B2 (en) 2002-11-21 2014-10-07 Archemix Llc Stabilized aptamers to platelet derived growth factor and their use as oncology therapeutics
ES2554660T3 (en) * 2002-11-25 2015-12-22 Masafumi Matsuo ENA nucleic acid drugs that modify splicing in mRNA precursors
ES2305808T3 (en) * 2003-05-20 2008-11-01 Bayer Healthcare Llc DIARILURES WITH INHIBITING ACTIVITY OF QUINASAS.
EP1696034A4 (en) * 2003-12-19 2006-12-27 Dainippon Sumitomo Pharma Co Novel method of nucleic acid transfer
US8710020B2 (en) * 2004-04-02 2014-04-29 The University Of British Columbia Clusterin antisense therapy for treatment of cancer
US8101350B1 (en) 2004-05-24 2012-01-24 Isis Pharmaceuticals, Inc. Modulation of exportin 5 expression
US20090062184A1 (en) * 2005-03-24 2009-03-05 Dainippon Sumitomo Pharma Co., Ltd. Fine particulate preparation comprising complex of nucleic acid molecule and collagen
WO2007014150A2 (en) * 2005-07-22 2007-02-01 Neopharm, Inc. Method of administering liposomes containing oligonucleotides
US20130028889A1 (en) 2010-02-04 2013-01-31 Ico Therapeutics Inc. Dosing regimens for treating and preventing ocular disorders using c-raf antisense
WO2011112516A1 (en) 2010-03-08 2011-09-15 Ico Therapeutics Inc. Treating and preventing hepatitis c virus infection using c-raf kinase antisense oligonucleotides

Citations (4)

* Cited by examiner, ā€  Cited by third party
Publication number Priority date Publication date Assignee Title
US5190931A (en) * 1983-10-20 1993-03-02 The Research Foundation Of State University Of New York Regulation of gene expression by employing translational inhibition of MRNA utilizing interfering complementary MRNA
WO1993004170A1 (en) * 1991-08-23 1993-03-04 The United States Of America Represented By The Secretary, Department Of Health And Human Services Raf protein kinase therapeutics
WO1994015645A1 (en) * 1992-12-31 1994-07-21 Texas Biotechnology Corporation Antisense molecules directed against genes of the raf oncogene family
WO1994023755A1 (en) * 1993-04-09 1994-10-27 Board Of Regents Of The University Of Nebraska Novel methods and compositions for the treatment of ras-activated cancer with heterotypic anti-raf antisense oligonucleotides

Family Cites Families (15)

* Cited by examiner, ā€  Cited by third party
Publication number Priority date Publication date Assignee Title
US5034506A (en) * 1985-03-15 1991-07-23 Anti-Gene Development Group Uncharged morpholino-based polymers having achiral intersubunit linkages
US4871838A (en) * 1985-07-23 1989-10-03 The Board Of Rijks Universiteit Leiden Probes and methods for detecting activated ras oncogenes
US5194428A (en) * 1986-05-23 1993-03-16 Worcester Foundation For Experimental Biology Inhibition of influenza virus replication by oligonucleotide phosphorothioates
US4806463A (en) * 1986-05-23 1989-02-21 Worcester Foundation For Experimental Biology Inhibition of HTLV-III by exogenous oligonucleotides
US5264423A (en) * 1987-03-25 1993-11-23 The United States Of America As Represented By The Department Of Health And Human Services Inhibitors for replication of retroviruses and for the expression of oncogene products
US5276019A (en) * 1987-03-25 1994-01-04 The United States Of America As Represented By The Department Of Health And Human Services Inhibitors for replication of retroviruses and for the expression of oncogene products
US5004810A (en) * 1988-09-30 1991-04-02 Schering Corporation Antiviral oligomers
US5098890A (en) * 1988-11-07 1992-03-24 Temple University-Of The Commonwealth System Of Higher Education Antisence oligonucleotides to c-myb proto-oncogene and uses thereof
US5087617A (en) * 1989-02-15 1992-02-11 Board Of Regents, The University Of Texas System Methods and compositions for treatment of cancer using oligonucleotides
US5166195A (en) * 1990-05-11 1992-11-24 Isis Pharmaceuticals, Inc. Antisense inhibitors of the human immunodeficiency virus phosphorothioate oligonucleotides
US5135917A (en) * 1990-07-12 1992-08-04 Nova Pharmaceutical Corporation Interleukin receptor expression inhibiting antisense oligonucleotides
US5541307A (en) * 1990-07-27 1996-07-30 Isis Pharmaceuticals, Inc. Backbone modified oligonucleotide analogs and solid phase synthesis thereof
WO1994008003A1 (en) * 1991-06-14 1994-04-14 Isis Pharmaceuticals, Inc. ANTISENSE OLIGONUCLEOTIDE INHIBITION OF THE ras GENE
EP0612353B1 (en) * 1991-09-16 1999-03-31 THE GOVERNMENT OF THE UNITED STATES OF AMERICA, as represented by THE SECRETARY, DEPARTMENT OF HEALTH AND HUMAN SERVICES Detection method for c-raf-1 genes
WO1996019240A1 (en) * 1994-12-21 1996-06-27 Novartis Ag Oligonucleotide-dendrimer conjugates

Patent Citations (4)

* Cited by examiner, ā€  Cited by third party
Publication number Priority date Publication date Assignee Title
US5190931A (en) * 1983-10-20 1993-03-02 The Research Foundation Of State University Of New York Regulation of gene expression by employing translational inhibition of MRNA utilizing interfering complementary MRNA
WO1993004170A1 (en) * 1991-08-23 1993-03-04 The United States Of America Represented By The Secretary, Department Of Health And Human Services Raf protein kinase therapeutics
WO1994015645A1 (en) * 1992-12-31 1994-07-21 Texas Biotechnology Corporation Antisense molecules directed against genes of the raf oncogene family
WO1994023755A1 (en) * 1993-04-09 1994-10-27 Board Of Regents Of The University Of Nebraska Novel methods and compositions for the treatment of ras-activated cancer with heterotypic anti-raf antisense oligonucleotides

Non-Patent Citations (8)

* Cited by examiner, ā€  Cited by third party
Title
CHEMICAL REVIEWS, Vol. 90, No. 4, issued June 1990, UHLMANN et al., "Antisense Oligonucleotides: A New Therapeutic Principle", pages 544-584. *
CRITICAL REVIEWS IN ONCOGENESIS, Vol. 3, Nos. 1 and 2, issued 1992, NECKERS et al., "Antisense Inhibition of Oncogene Expression", pages 175-230. *
CROOKE et al., "Antisense Research and Applications", Published 1993, by CRC PRESS (BOCA RATON), pages 7-35. *
EUROPEAN JOURNAL OF IMMUNOLOGY, Vol. 23, issued 1993, RIEDEL et al., "The Mitogenic Response of T Cells to Interleukin-2 Requires Raf-1", pages 3146-3150. *
JOURNAL OF BIOLOGICAL CHEMISTRY, Vol. 269, No. 19, issued 13 May 1994, TORNKVIST et al., "Inhibition of Raf-1 Kinase Expression Abolishes Insulin Stimulation of DNA Synthesis in H4IIE Hepatoma Cells", pages 13919-13921. *
NUCLEIC ACIDS RESEARCH, Vol. 14, No. 2, issued 1986, BONNER et al., "The Complete Coding Sequence of the Human Raf Oncogene and the Corresponding Sequence of the C-Raf-1 Gene", pages 1009-1015. *
NUCLEIC ACIDS RESEARCH, Vol. 15, No. 2, issued 1987, BECK et al., "The Complete Coding Sequence of the Human A-Raf-1 Oncogene and Transforming Activity of A Human A-Raf Carrying Retrovirus", pages 595-609. *
SCIENCE, Vol. 243, issued 10 March 1989, KASID et al., "Effect of Antisense C-Raf-1 on Tumorigenicity and Radiation Sensitivity of a Human Squamous Carcinoma", pages 1354-1356. *

Cited By (82)

* Cited by examiner, ā€  Cited by third party
Publication number Priority date Publication date Assignee Title
WO1996039415A1 (en) * 1995-06-05 1996-12-12 Isis Pharmaceuticals, Inc. ANTISENSE OLIGONUCLEOTIDE MODULATION OF raf GENE EXPRESSION
US6096720A (en) * 1995-08-01 2000-08-01 Novartis Ag Liposomal oligonucleotide compositions
WO1997004787A1 (en) * 1995-08-01 1997-02-13 Novartis Ag Liposomal oligonucleotide compositions
WO1997010829A1 (en) * 1995-09-19 1997-03-27 Novartis Ag Pharmaceutical compositions containing an oligonucleotide and an anionic polysaccharide
US6562960B1 (en) * 1996-03-05 2003-05-13 Isis Pharmaceuticals, Inc. Oligonucleotide analogues
US5744460A (en) * 1996-03-07 1998-04-28 Novartis Corporation Combination for treatment of proliferative diseases
EP0906099A1 (en) * 1996-04-03 1999-04-07 Merck & Co., Inc. A method of treating cancer
EP0906099A4 (en) * 1996-04-03 2001-02-07 Merck & Co Inc A method of treating cancer
US6027892A (en) * 1996-12-30 2000-02-22 Chang Esther H Compositions and methods for reducing radiation and drug resistance in cells
US6803360B1 (en) 1996-12-30 2004-10-12 Georgetown University Compositions and methods for reducing radiation and drug resistance in cells
US7262173B2 (en) 1997-03-21 2007-08-28 Georgetown University Chemosensitizing with liposomes containing oligonucleotides
US6126965A (en) * 1997-03-21 2000-10-03 Georgetown University School Of Medicine Liposomes containing oligonucleotides
AU749927B2 (en) * 1997-03-21 2002-07-04 Georgetown University Liposomes containing oligonucleotides
WO1998043095A1 (en) * 1997-03-21 1998-10-01 Georgetown University Liposomes containing oligonucleotides
WO2000018781A1 (en) 1998-09-29 2000-04-06 Isis Pharmaceuticals, Inc. Antisense modulation of survivin expression
WO2000056342A3 (en) * 1999-03-19 2001-07-26 Assist Publ Hopitaux De Paris Use of stabilised oligonucleotides for preparing a medicine with antitumour activity
WO2000056342A2 (en) * 1999-03-19 2000-09-28 Assistance Publique-Hopitaux De Paris Use of stabilised oligonucleotides for preparing a medicine with antitumour activity
DE10020138A1 (en) * 2000-04-14 2001-10-31 Ulf R Rapp Nucleic acid coding for at least one partial sequence with a MEKK1 binding site
WO2006123182A2 (en) 2005-05-17 2006-11-23 Merck Sharp & Dohme Limited Cyclohexyl sulphones for treatment of cancer
WO2007093827A1 (en) 2006-02-15 2007-08-23 Istituto Di Ricerche Di Biologia Molecolare P. Angeletti Spa Thiophene and thiazole substituted trifluoroethanone derivatives as histone deacetylase (hdac) inhibitors
US7553854B2 (en) 2006-04-19 2009-06-30 Novartis Vaccines And Diagnostics, Inc. 6-O-substituted benzoxazole and benzothiazole compounds and methods of inhibiting CSF-1R signaling
US8710048B2 (en) 2006-04-19 2014-04-29 Novartis Ag 6-O-substituted benzoxazole and benzothiazole compounds and methods of inhibiting CSF-1R signaling
US8173689B2 (en) 2006-04-19 2012-05-08 Novartis Ag 6-O-substituted benzoxazole and benzothiazole compounds and methods of inhibiting CSF-1R signaling
EP2698157A1 (en) 2006-09-22 2014-02-19 Merck Sharp & Dohme Corp. Method of treatment using fatty acid synthesis inhibitors
EP2946778A1 (en) 2006-09-22 2015-11-25 Merck Sharp & Dohme Corp. Method of treatment using fatty acid synthesis inhibitors
US11096950B2 (en) 2006-11-01 2021-08-24 Barbara Brooke Jennings Compounds, methods, and treatments for abnormal signaling pathways for prenatal and postnatal development
EP2336120A1 (en) 2007-01-10 2011-06-22 Istituto di ricerche di Biologia Molecolare P. Angeletti S.R.L. Combinations containing amide substituted indazoles as poly(ADP-ribose)polymerase (PARP) inhibitors
EP2805945A1 (en) 2007-01-10 2014-11-26 MSD Italia S.r.l. Amide substituted indazoles as poly(ADP-ribose)polymerase (PARP) inhibitors
WO2008106692A1 (en) 2007-03-01 2008-09-04 Novartis Vaccines And Diagnostics, Inc. Pim kinase inhibitors and methods of their use
WO2008144062A1 (en) 2007-05-21 2008-11-27 Novartis Ag Csf-1r inhibitors, compositions, and methods of use
EP3103791A1 (en) 2007-06-27 2016-12-14 Merck Sharp & Dohme Corp. 4-carboxybenzylamino derivatives as histone deacetylase inhibitors
WO2009002495A1 (en) 2007-06-27 2008-12-31 Merck & Co., Inc. 4-carboxybenzylamino derivatives as histone deacetylase inhibitors
WO2010114780A1 (en) 2009-04-01 2010-10-07 Merck Sharp & Dohme Corp. Inhibitors of akt activity
US8765747B2 (en) 2009-06-12 2014-07-01 Dana-Farber Cancer Institute, Inc. Fused 2-aminothiazole compounds
US9505784B2 (en) 2009-06-12 2016-11-29 Dana-Farber Cancer Institute, Inc. Fused 2-aminothiazole compounds
WO2011046771A1 (en) 2009-10-14 2011-04-21 Schering Corporation SUBSTITUTED PIPERIDINES THAT INCREASE p53 ACTIVITY AND THE USES THEREOF
US11826365B2 (en) 2009-12-29 2023-11-28 Dana-Farber Cancer Institute, Inc. Type II raf kinase inhibitors
US8987275B2 (en) 2010-03-16 2015-03-24 Dana-Farber Cancer Institute, Inc. Indazole compounds and their uses
WO2011115725A2 (en) 2010-03-16 2011-09-22 Dana-Farber Cancer Institute, Inc. Indazole compounds and their uses
WO2011163330A1 (en) 2010-06-24 2011-12-29 Merck Sharp & Dohme Corp. Novel heterocyclic compounds as erk inhibitors
WO2012018754A2 (en) 2010-08-02 2012-02-09 Merck Sharp & Dohme Corp. RNA INTERFERENCE MEDIATED INHIBITION OF CATENIN (CADHERIN-ASSOCIATED PROTEIN), BETA 1 (CTNNB1) GENE EXPRESSION USING SHORT INTERFERING NUCLEIC ACID (siNA)
EP3330377A1 (en) 2010-08-02 2018-06-06 Sirna Therapeutics, Inc. Rna interference mediated inhibition of catenin (cadherin-associated protein), beta 1 (ctnnb1) gene expression using short interfering nucleic acid (sina)
WO2012024170A2 (en) 2010-08-17 2012-02-23 Merck Sharp & Dohme Corp. RNA INTERFERENCE MEDIATED INHIBITION OF HEPATITIS B VIRUS (HBV) GENE EXPRESSION USING SHORT INTERFERING NUCLEIC ACID (siNA)
EP4079856A1 (en) 2010-08-17 2022-10-26 Sirna Therapeutics, Inc. Rna interference mediated inhibition of hepatitis b virus (hbv) gene expression using short interfering nucleic acid (sina)
WO2012027236A1 (en) 2010-08-23 2012-03-01 Schering Corporation NOVEL PYRAZOLO[1,5-a]PYRIMIDINE DERIVATIVES AS mTOR INHIBITORS
WO2012030685A2 (en) 2010-09-01 2012-03-08 Schering Corporation Indazole derivatives useful as erk inhibitors
WO2012036997A1 (en) 2010-09-16 2012-03-22 Schering Corporation Fused pyrazole derivatives as novel erk inhibitors
EP3327125A1 (en) 2010-10-29 2018-05-30 Sirna Therapeutics, Inc. Rna interference mediated inhibition of gene expression using short interfering nucleic acids (sina)
EP3766975A1 (en) 2010-10-29 2021-01-20 Sirna Therapeutics, Inc. Rna interference mediated inhibition of gene expression using short interfering nucleic acid (sina)
WO2012058210A1 (en) 2010-10-29 2012-05-03 Merck Sharp & Dohme Corp. RNA INTERFERENCE MEDIATED INHIBITION OF GENE EXPRESSION USING SHORT INTERFERING NUCLEIC ACIDS (siNA)
WO2012087772A1 (en) 2010-12-21 2012-06-28 Schering Corporation Indazole derivatives useful as erk inhibitors
WO2012145471A1 (en) 2011-04-21 2012-10-26 Merck Sharp & Dohme Corp. Insulin-like growth factor-1 receptor inhibitors
WO2013063214A1 (en) 2011-10-27 2013-05-02 Merck Sharp & Dohme Corp. Novel compounds that are erk inhibitors
US10144730B2 (en) 2011-11-17 2018-12-04 Dana-Farber Cancer Institute, Inc. Inhibitors of c-Jun-N-terminal kinase (JNK)
WO2013165816A2 (en) 2012-05-02 2013-11-07 Merck Sharp & Dohme Corp. SHORT INTERFERING NUCLEIC ACID (siNA) COMPOSITIONS
EP3919620A1 (en) 2012-05-02 2021-12-08 Sirna Therapeutics, Inc. Short interfering nucleic acid (sina) compositions
WO2014052563A2 (en) 2012-09-28 2014-04-03 Merck Sharp & Dohme Corp. Novel compounds that are erk inhibitors
US10112927B2 (en) 2012-10-18 2018-10-30 Dana-Farber Cancer Institute, Inc. Inhibitors of cyclin-dependent kinase 7 (CDK7)
US10787436B2 (en) 2012-10-18 2020-09-29 Dana-Farber Cancer Institute, Inc. Inhibitors of cyclin-dependent kinase 7 (CDK7)
US9758522B2 (en) 2012-10-19 2017-09-12 Dana-Farber Cancer Institute, Inc. Hydrophobically tagged small molecules as inducers of protein degradation
USRE48175E1 (en) 2012-10-19 2020-08-25 Dana-Farber Cancer Institute, Inc. Hydrophobically tagged small molecules as inducers of protein degradation
US10000483B2 (en) 2012-10-19 2018-06-19 Dana-Farber Cancer Institute, Inc. Bone marrow on X chromosome kinase (BMX) inhibitors and uses thereof
WO2014085216A1 (en) 2012-11-28 2014-06-05 Merck Sharp & Dohme Corp. Compositions and methods for treating cancer
WO2014100065A1 (en) 2012-12-20 2014-06-26 Merck Sharp & Dohme Corp. Substituted imidazopyridines as hdm2 inhibitors
WO2014120748A1 (en) 2013-01-30 2014-08-07 Merck Sharp & Dohme Corp. 2,6,7,8 substituted purines as hdm2 inhibitors
WO2015034925A1 (en) 2013-09-03 2015-03-12 Moderna Therapeutics, Inc. Circular polynucleotides
US11040957B2 (en) 2013-10-18 2021-06-22 Dana-Farber Cancer Institute, Inc. Heteroaromatic compounds useful for the treatment of proliferative diseases
US9862688B2 (en) 2014-04-23 2018-01-09 Dana-Farber Cancer Institute, Inc. Hydrophobically tagged janus kinase inhibitors and uses thereof
US10017477B2 (en) 2014-04-23 2018-07-10 Dana-Farber Cancer Institute, Inc. Janus kinase inhibitors and uses thereof
EP3514151A1 (en) 2014-08-06 2019-07-24 Novartis AG Protein kinase c inhibitors and methods of their use
WO2016020864A1 (en) 2014-08-06 2016-02-11 Novartis Ag Protein kinase c inhibitors and methods of their use
US10870651B2 (en) 2014-12-23 2020-12-22 Dana-Farber Cancer Institute, Inc. Inhibitors of cyclin-dependent kinase 7 (CDK7)
US10550121B2 (en) 2015-03-27 2020-02-04 Dana-Farber Cancer Institute, Inc. Inhibitors of cyclin-dependent kinases
US11325910B2 (en) 2015-03-27 2022-05-10 Dana-Farber Cancer Institute, Inc. Inhibitors of cyclin-dependent kinases
US10702527B2 (en) 2015-06-12 2020-07-07 Dana-Farber Cancer Institute, Inc. Combination therapy of transcription inhibitors and kinase inhibitors
US11142507B2 (en) 2015-09-09 2021-10-12 Dana-Farber Cancer Institute, Inc. Inhibitors of cyclin-dependent kinases
WO2018058022A1 (en) 2016-09-26 2018-03-29 Merck Sharp & Dohme Corp. Anti-cd27 antibodies
WO2018190719A2 (en) 2017-04-13 2018-10-18 Aduro Biotech Holdings, Europe B.V. Anti-sirp alpha antibodies
WO2019094311A1 (en) 2017-11-08 2019-05-16 Merck Sharp & Dohme Corp. Prmt5 inhibitors
WO2019152642A1 (en) 2018-02-01 2019-08-08 Merck Sharp & Dohme Corp. Anti-pd-1/lag3 bispecific antibodies
WO2020033282A1 (en) 2018-08-07 2020-02-13 Merck Sharp & Dohme Corp. Prmt5 inhibitors
WO2020033284A1 (en) 2018-08-07 2020-02-13 Merck Sharp & Dohme Corp. Prmt5 inhibitors

Also Published As

Publication number Publication date
IL113804A0 (en) 1995-08-31
FI964792A0 (en) 1996-11-29
JP2000152797A (en) 2000-06-06
CA2191795C (en) 2003-06-10
IL113804A (en) 2000-12-06
EP0763052A4 (en) 1998-10-14
EP0763052A1 (en) 1997-03-19
JPH09507502A (en) 1997-07-29
FI964792A (en) 1996-11-29
HU9603277D0 (en) 1997-01-28
US5563255A (en) 1996-10-08
EP0763052B1 (en) 2004-10-27
AU2698295A (en) 1995-12-21
US20140187601A1 (en) 2014-07-03
ATE280772T1 (en) 2004-11-15
DE69533697D1 (en) 2004-12-02
JP3121599B2 (en) 2001-01-09
NO965080D0 (en) 1996-11-28
AU689267B2 (en) 1998-03-26
NO965080L (en) 1996-11-28
US20120149755A1 (en) 2012-06-14
DE69533697T2 (en) 2006-02-09
US20130079387A1 (en) 2013-03-28
JP3231779B2 (en) 2001-11-26
HUT76675A (en) 1997-10-28
CA2191795A1 (en) 1995-12-07

Similar Documents

Publication Publication Date Title
AU689267B2 (en) Antisense oligonucleotide modulation of RAF gene expression
US5656612A (en) Antisense oligonucleotide modulation of raf gene expression
EP0863911B1 (en) ANTISENSE OLIGONUCLEOTIDE MODULATION OF raf GENE EXPRESSION
US6358932B1 (en) Antisense oligonucleotide inhibition of raf gene expression
US6391636B1 (en) Antisense oligonucleotide modulation of raf gene expression
US5948902A (en) Antisense oligonucleotides to human serine/threonine protein phosphatase genes
AU678628B2 (en) Oligonucleotide modulation of multidrug resistance-associated protein
US5981731A (en) Antisense oligonucleotide modulation of B-raf gene expression
US20020151511A1 (en) Antisense oligonucleotide modulation of human MDM2 expression
US6806258B2 (en) Antisense oligonucleotide modulation of raf gene expression
WO1994008003A1 (en) ANTISENSE OLIGONUCLEOTIDE INHIBITION OF THE ras GENE
US6090626A (en) Antisense oligonucleotide modulation of raf gene expression
US5952229A (en) Antisense oligonucleotide modulation of raf gene expression
EP0928419A1 (en) Compositions and methods for modulating type i interleukin-1 receptor expression
AU5295993A (en) Antisense oligonucleotide inhibition of the ras gene
AU714142B2 (en) Antisense oligonucleotide modulation of RAF gene expression
US20030119769A1 (en) Antisense oligonucleotide modulation of raf gene expression
US5814619A (en) Oligonucleotide inhibition of P120

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AM AU BB BG BR BY CA CN CZ EE FI GE HU IS JP KG KP KR KZ LK LR LT LV MD MG MN MX NO NZ PL PT RO RU SI SK TJ TT UA US UZ VN

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): KE MW SD SZ UG AT BE CH DE DK ES FR GB GR IE IT LU MC NL PT SE BF BJ CF CG CI CM GA GN ML MR NE SN TD TG

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

Ref document number: 08756806

Country of ref document: US

WWE Wipo information: entry into national phase

Ref document number: 2191795

Country of ref document: CA

Ref document number: 964792

Country of ref document: FI

WWE Wipo information: entry into national phase

Ref document number: 1019960706806

Country of ref document: KR

WWE Wipo information: entry into national phase

Ref document number: 1995922217

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 1995922217

Country of ref document: EP

WWG Wipo information: grant in national office

Ref document number: 1995922217

Country of ref document: EP