WO2004066949A2 - Oligonucleotides anti-sens inhibant l'expression du hif-1 - Google Patents

Oligonucleotides anti-sens inhibant l'expression du hif-1 Download PDF

Info

Publication number
WO2004066949A2
WO2004066949A2 PCT/US2004/002344 US2004002344W WO2004066949A2 WO 2004066949 A2 WO2004066949 A2 WO 2004066949A2 US 2004002344 W US2004002344 W US 2004002344W WO 2004066949 A2 WO2004066949 A2 WO 2004066949A2
Authority
WO
WIPO (PCT)
Prior art keywords
hif
oligonucleotide
cells
oligonucleotides
compound
Prior art date
Application number
PCT/US2004/002344
Other languages
English (en)
Other versions
WO2004066949A8 (fr
WO2004066949A3 (fr
Inventor
Heejeong Yoon
Lingjun Mao
Young Bok Lee
Chang-Ho Ahn
Xiaoming Jiang
Original Assignee
Rexahn Corporation
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Rexahn Corporation filed Critical Rexahn Corporation
Priority to JP2006503098A priority Critical patent/JP4546454B2/ja
Priority to BR0407070-4A priority patent/BRPI0407070A/pt
Priority to AT04706034T priority patent/ATE441710T1/de
Priority to DE602004022921T priority patent/DE602004022921D1/de
Priority to CA2513398A priority patent/CA2513398C/fr
Priority to EP04706034A priority patent/EP1601325B1/fr
Priority to AU2004207576A priority patent/AU2004207576B8/en
Publication of WO2004066949A2 publication Critical patent/WO2004066949A2/fr
Publication of WO2004066949A8 publication Critical patent/WO2004066949A8/fr
Publication of WO2004066949A3 publication Critical patent/WO2004066949A3/fr

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • 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
    • C12N2310/10Type of nucleic acid
    • C12N2310/11Antisense
    • 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
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6876Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
    • C12Q1/6883Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material
    • C12Q1/6886Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material for cancer

Definitions

  • This invention relates to new antisense oligonucleotide compounds, RX- 0047 and RX-0149, that inhibit expression of a human protein, HIF-1 , and also induce cytotoxicity in several cancer cell lines.
  • HIF-1 is one of the major regulators of cancer cell growth. HIF renders cells capable of surviving hypoxia and stimulating endothelial growth and it is upregulated in a broad range of cancers (Zhong et al., Cancer Res. 1999 59: 5830-5835).
  • VHL is a tumor suppressor gene that is mutated in most sporadic clear-cell renal carcinoma and in VHL disease.
  • HIF-1 HIF-1 pathway mediated response that drives tumor angiogenesis.
  • United States Patent No., 6,222,018 issued to Semenza, April 24, 2001 relates to the nucleotide sequences encoding HIF-1. The specific oligonucleotides disclosed and claimed in the present invention were not disclosed in that patent.
  • FIG. 2 Western blot analysis of inhibition of HIF-1 protein expression by RX-0047 and RX-0149. Summary of the Invention
  • the present invention is directed to antisense oligonucleotides, which are targeted to a nucleic acid encoding HIF-1 (hypoxia-inducible factor 1 ), and which modulate the expression of HIF-1. Also provided is a method of inhibiting expression of HIF-1 in cells comprising contacting the cells with the oligonucleotide compounds and compositions of the invention.
  • An advantage of the presently described oligonucleotides is that, in addition to inhibiting expression of HIF-1 , they have a cytotoxic effect on several different cancer cell lines.
  • the advantages of the present invention can be obtained by contacting cells of various cancer cell lines with an antisense compound that is specifically hybridizable to a site on the HIF-1 gene having the following sequence: 5' ttggacactggtggctcatt 3' at site 2,772 of HIF-1 gene (Genebank # NM001530) (Seq. Id. No. 1 ). Particularly preferred is RX-0047, comprising 5' aatgagccaccagtgtccaa 3' (Seq. Id. No. 2).
  • HIF hyperoxia-inducible factor
  • HIF-1 ⁇ and HIF-1 ⁇ which is also known as ARNT (Aryl Receptor Nuclear Translocator).
  • ARNT Aryl Receptor Nuclear Translocator
  • ODD oxygen-dependent degradation
  • VHL von Hippel-Lindau
  • HIF-1 ⁇ is not hydroxylated because the hydroxylase, which requires dioxygen for activity, is inactive and thus HIF-1 ⁇ is not recognized by pVHL and accumulates in the cell. HIF-1 ⁇ then translocates to the nucleus and dimerizes with the constitutively present HIF-1 ⁇ subunit (Semenza, Genes & Development, 1985. 14: 1983-1991 ).
  • the dimer then binds to the hypoxia responsive element (HRE) in target genes, resulting in their transactivation of genes such as erythropoitin, VEGF (Forsythe et al., Mol. Cell. Biol., 1996. 16: 4604-4613), platelet-derived growth factor- ⁇ (PDGF- ⁇ ), glucose transporter (GLUT1 ) and nitrous oxide synthetase (Neckers, J. Natl. Cancer Ins., 1999. 91 : 106-107).
  • HRE hypoxia responsive element
  • HIF-1 ⁇ certain hormones and growth factors also lead to increased levels of HIF-1 ⁇ as well as mutations in certain oncogenes and tumor-suppressor genes, VHL for example, result in an increase in HIF-1 ⁇ level (Ivan and Kaelin, Current Opinion in Genetics & Development, 2001 11 : 27-34). It will be interesting to determine whether hydroxylation or alternative mechanisms are involved in this hypoxia-independent HIF activation.
  • Several current strategies for cancer therapies that exploit the hypoxic microenvironment and response are as follows; 1 ) hypoxia-dependent drugs or gene-therapy vectors, 2) inhibition of HIF stability, 3) inhibition of transactivation by HIF, and 4) VEGF inhibitors.
  • HIF transcription factor Given the pivotal role of the HIF transcription factor in the development of cancers, it would be desirable to inhibit its operation during oncogenesis. However, it would also be desirable, to the extent possible, to avoid interrupting the family's roles in other aspects of cellular metabolism.
  • One approach might be to identify the gene that encodes a likely transcription factor that expressed highly during hypoxia, and devise an antisense oligonucleotide that can be used to inhibit that gene's activity in the right context. The inventors have found that two antisense oligonucleotides both exhibit an enhanced ability to inhibit the production of protein by the HIF-1 gene, and further, induce cytotoxicity in a variety of cancer cell lines.
  • an antisense compound is a tool that can be used to introduce modifications into the nucleic acids found in living cells.
  • the term "antisense” refers to the notion that nucleic acids "encode” proteins. That is, the sequence of nucleotides found in a given nucleic acid determines, among other things, what protein will be produced. A “sense” sequence for a full gene will yield a normal protein in the usual amount, in response to a given stimulus. A “sense” oligonucleotide will hybridize with a normal gene sequence, and will not affect the amount of, or properties of, the protein. A "nonsense" sequence will not yield a product, or may yield a non-functional product.
  • antisense oligonucleotide will hybridize with a normal gene, but will yield a protein altered with respect to its structure, or amount. It has been found that antisense oligomers, that is antisense compounds that are relatively short, can be easily inserted into cells, where they alter gene function. Antisense compounds are commonly used as research reagents for the exploration of gene function because they are able to alter gene expression with extraordinar specificity, and may be used to elucidate the function of particular genes. Antisense compounds can be used, for example, to distinguish between functions of various members of a biological pathway.
  • Antisense oligonucleotides can be used to selectively block disease- causing genes, thereby inhibiting production of disease-associated proteins. Some antisense oligonucleotides have been safely and effectively administered to humans, and numerous clinical trials are presently underway. It is thus possible that oligonucleotides can be used to treat cells, tissues, and animals, especially humans.
  • the term "oligonucleotide” refers to an oligomer or polymer of ribonucleic acid (RNA) or deoxyribonucleic acid (DNA) or mimetics thereof.
  • This term includes oligonucleotides composed of naturally- occurring nucleobases, sugars and covalent internucleoside (backbone) linkages as well as oligonucleotides having non-naturally-occurring portions which function similarly.
  • modified or substituted oligonucleotides are often preferred over native forms because of desirable properties such as, for example, enhanced cellular uptake, enhanced affinity for a nucleic acid target and increased stability in the presence of nucleobases.
  • the present invention employs oligomeric nucleotide compounds, particularly antisense oligonucleotides, which are targeted to a portion of a nucleic acid encoding HIF-1 , and which modulate the expression of HIF-1.
  • the oligonucleotide compounds are designed to specifically hybridize with one or more nucleic acids encoding HIF-1.
  • One oligonucleotide, RX-0047 is targeted to a site on the HIF-1 gene having the following sequence: 5' ttggacactggtggctcatt 3' at site 2,772 of HIF-1 gene (Genebank # NM001530 ) (Seq. Id. No. 1).
  • the sequence for the backbone of RX-0047 is complementary to this site.
  • the other oligonucleotide, RX-0149 is targeted to a site in the coding region of the HIF-1 gene having the following sequence: 5' gacttggagatgttagctcc 3' at site 1 ,936 of HIF-1 gene (Genebank # NM001530) (Seq. Id. No. 3).
  • the sequence for the backbone of RX-0149 is complementary to this site.
  • the inventors have found that oligomers comprising either 5 or 10 nucleotides upstream and downstream from the sequence where the 20-mer of RX-0047 and RX-0149 were derived showed a measurable inhibition of HIF-1 mRNA expression.
  • this oligonucleotide is more sensitive to variability, and that while 18-mer of RX-0149 showed some inhibition of HIF-1 mRNA expression, further truncation from either end resulted in a substantial loss of inhibition of HIF-1 mRNA expression
  • the oligomers comprising either 5 or 10 nucleotides upstream and downstream from the sequence where the 20-mer of RX-0047 and RX-0149 were derived demonstrated an inhibition of proliferation of cancer cells.
  • the truncated versions of RX-0047 and RX-0149 which showed some inhibition of HIF-1 mRNA expression also showed an inhibition of cancer cell proliferation.
  • nucleic acid encoding HIF-1 encompasses
  • DNA encoding HIF-1 DNA encoding HIF-1 , RNA (including pre-mRNA) transcribed from such DNA, and also cDNA derived from such RNA.
  • the specific hybridization of an antisense oligomeric compound with its target nucleic acid interferes with the normal function of the nucleic acid.
  • the functions of DNA to be interfered with include replication and transcription.
  • the functions of RNA to be interfered with include all vital functions such as, for example, translocation of the RNA to the site of protein translation, translation of protein from the RNA, splicing of the RNA to yield one or more mRNA species, and catalytic activity which may be engaged in or facilitated by the RNA.
  • the overall effect of such interference with target nucleic acid function is modulation of the expression, or production of, a protein.
  • modulation means either an increase (stimulation) or a decrease (inhibition) in the expression of a gene.
  • the gene encoding HIF-1 is modulated so that expression of HIF-1 is inhibited.
  • to hybridize means to hydrogen bond, which may be via Watson-Crick, Hoogsteen or reversed Hoogsteen hydrogen bonding, between complementary nucleoside or nucleotide bases.
  • adenine and thymine are complementary nucleobases which pair through the formation of hydrogen bonds.
  • “Complementary,” as used herein, refers to the capacity for precise pairing between two nucleotides.
  • oligonucleotide and the DNA or RNA are considered to be complementary to each other at that position.
  • the oligonucleotide and the DNA or RNA are complementary to each other when a sufficient number of corresponding positions in each molecule are occupied by nucleotides which can hydrogen bond with each other.
  • “specifically hybridizable” and “complementary” are terms which are used to indicate a sufficient degree of complementarity or precise pairing such that stable and specific binding occurs between the oligonucleotide and the DNA or RNA target.
  • an antisense compound need not be 100% complementary to that of its target nucleic acid to be specifically hybridizable.
  • An antisense compound is specifically hybridizable when binding of the compound to the target DNA or RNA molecule interferes with the normal function of the target DNA or RNA to cause a loss of utility, and there is a sufficient degree of complementarity to avoid non-specific binding of the antisense compound 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, and in the case of in vitro assays, under conditions in which the assays are performed.
  • antisense oligonucleotides are a preferred form of antisense compound
  • the present invention comprehends other oligomeric antisense compounds, including but not limited to oligonucleotide mimetics such as are described below.
  • the antisense compounds in accordance with this invention preferably comprise from about 10 to about 30 nucleobases. Particularly preferred are antisense oligonucleotides comprising about 20 nucleobases (i.e. about 20 linked nucleosides).
  • a nucleoside is a base-sugar combination. The base portion of the nucleoside is normally a heterocyclic base. The two most common classes of such heterocyclic bases are the purines and the pyrimidines.
  • Nucleotides are nucleosides that further include a phosphate group covalently linked to the sugar portion of the nucleoside.
  • the phosphate group can be linked to either the 2', 3' or 5' hydroxyl moiety of the sugar.
  • the phosphate groups covalently link adjacent nucleosides to one another to form a linear polymeric compound.
  • the respective ends of this linear polymeric structure can be further joined to form a circular structure, however, open linear structures are generally preferred.
  • the phosphate groups are commonly referred to as forming the intemucleoside backbone of the oligonucleotide.
  • the normal linkage or backbone of RNA and DNA is a 3' to 5' phosphodiester linkage.
  • Specific examples of preferred antisense compounds useful in this invention include oligonucleotides containing modified backbones or non-natural intemucleoside linkages.
  • oligonucleotides having modified backbones include those that retain a phosphorus atom in the backbone and those that do not have a phosphorus atom in the backbone.
  • modified oligonucleotides that do not have a phosphorus atom in their intemucleoside backbone can also be considered to be oligonucleosides.
  • Preferred modified oligonucleotide backbones include, for example, phosphorothioates, chiral phosphorothioates, phosphorodithioates, phosphotriesters, aminoalkylphosphotriesters, methyl and other alkyl phosphonates including 3'-alkylene phosphonates and chiral phosphonates, phosphinates, phosphoramidates including 3'-amino phosphoramidate and aminoalkylphosphoramidates, thionophosphoramidates, thionoalkylphosphonates, thionoalkylphosphotriesters, and boranophosphates having normal 3'-5' linkages, 2'-5' linked analogs of these, and those having inverted polarity wherein the adjacent pairs of nucleoside units are linked 3'-5' to 5'-3' or 2'-5' to 5'-2'.
  • Various salts, mixed salts and free acid forms are also included.
  • Preferred modified oligonucleotide backbones that do not include a phosphorus atom therein have backbones that are formed by short chain alkyl or cycloalkyl intemucleoside linkages, mixed heteroatom and alkyl or cycloalkyl intemucleoside linkages, or one or more short chain heteroatomic or heterocyclic intemucleoside linkages.
  • morpholino linkages formed in part from the sugar portion of a nucleoside
  • siloxane backbones sulfide, sulfoxide and sulfone backbones
  • formacetyl and thioformacetyl backbones methylene formacetyl and thioformacetyl backbones
  • alkene containing backbones sulfamate backbones
  • sulfonate and sulfonamide backbones amide backbones; and others having mixed N, O, S and CH 2 component parts.
  • both the sugar and the intemucleoside linkage, i.e., the backbone, of the nucleotide units are replaced with novel groups.
  • the base units are maintained for hybridization with an appropriate nucleic acid target compound.
  • an oligomeric compound an oligonucleotide mimetic that has been shown to have excellent hybridization properties, is referred to as a peptide nucleic acid (PNA).
  • PNA peptide nucleic acid
  • the sugar-backbone of an oligonucleotide is replaced with an amide containing backbone, in particular an aminoethylglycine backbone.
  • nucleobases are retained and are bound directly or indirectly to aza nitrogen atoms of the amide portion of the backbone.
  • Most preferred embodiments of the invention are oligonucleotides with phosphorothioate backbones and oligonucleosides with heteroatom backbones, and in particular -CH 2 -NH-O-CH 2 -, -CH 2 -N(CH 3 )-O-CH 2 - [known as a methylene (methylimino) or MMI backbone], -CH2-O-N(CH 3 )-CH 2 -, -CH 2 -N(CH 3 )-N(CH 3 )-CH 2 - and -O-N(CH 3 )-CH 2 -CH 2 -[wherein the native phosphodiester backbone is represented as -O-P-O-CH 2 -].
  • oligonucleotides having morpholino backbone structures are also preferred.
  • Modified oligonucleotides may also contain one or more substituted sugar moieties.
  • Preferred oligonucleotides comprise one of the following at the 2' position: OH; F; O-, S-, or N-alkyl; O-, S-, or N-alkenyl; O-, S- or N-alkynyl; or O- alkyl-O-alkyl, wherein the alkyl, alkenyl and alkynyl may be substituted or unsubstituted C ⁇ to C ⁇ 0 alkyl or C 2 to C 1 0 alkenyl and alkynyl.
  • oligonucleotides comprise one of the following at the 2' position: C ⁇ to C ⁇ o lower alkyl, substituted lower alkyl, alkaryl, aralkyl, O-alkaryl or O-aralkyl, SH, SCH 3 , OCN, Cl, Br, CN, CF 3 , OCF 3 , SOCH 3 , SO 2 CH 3 , ONO 2 , NO 2 , N 3 , NH 2 , heterocycloalkyl, heterocycloalkaryl, aminoalkylamino, polyalkylamino, substituted silyl, an RNA cleaving group, a reporter group, an intercalator, a group for improving the pharmacokinetic properties of an oligonucleotide, or a group for improving the pharmacodynamic properties of an oligonucleotide, and other substituents having similar properties.
  • a preferred modification includes 2'- methoxyethoxy (2'-O-CH 2 CH 2 OCH 3 , also known as 2'-O-(2-methoxyethyl) or 2'- MOE) (Martin et al., Helv. Chim. Ada, 1995, 78, 486-504) i.e., an alkoxyalkoxy group.
  • a further preferred modification includes 2'-dimethylaminooxyethoxy, i.e., a O(CH 2 ) 2 ON(CH 3 ) 2 group, also known as 2'-DMAOE, as described in examples hereinbelow.
  • Oligonucleotides may also have sugar mimetics such as cyclobutyl moieties in place of the pentofuranosyl sugar. Oligonucleotides may also include nucleobase (often referred to in the art simply as "base") modifications or substitutions.
  • unmodified or “natural” nucleobases include the purine bases adenine (A) and guanine (G), and the pyrimidine bases thymine (T), cytosine (C) and uracil (U).
  • Modified nucleobases include other synthetic and natural nucleobases such as 5-methylcytosine (5-Me-C), 5-hydroxymethyl cytosine, xanthine, hypoxanthine, 2-aminoadenine, 6-methyl and other alkyl derivatives of adenine and guanine, 2- propyl and other alkyl derivatives of adenine and guanine, 2-thiouracil, 2- thiothymine and 2-thiocytosine, 5-halouracil and cytosine, 5-propynyl uracil and cytosine, 6-azo uracil, cytosine and thymine, 5-uracil (pseudouracil), 4-thiouracil, 8-halo, 8-amino, 8-thiol, 8-thioalkyl, 8-hydroxyl and other 8-substituted adenines and guanines, 5-halo particularly 5-bromo, 5-trifluoromethyl and other 5- substituted
  • nucleobases are particularly useful for increasing the binding affinity of the oligomeric compounds of the invention. These include 5-substituted pyrimidines, 6-azapyrimidines and N-2, N-6 and O-6 substituted purines, including 2-aminopropyladenine, 5-propynyluracil and 5- propynylcytosine. 5-methylcytosine substitutions have been shown to increase nucleic acid duplex stability by 0.6-1.2° C. and are presently preferred base substitutions, even more particularly when combined with 2'-O-methoxyethyl sugar modifications.
  • oligonucleotides of the invention involves chemically linking to the oligonucleotide one or more moieties or conjugates which enhance the activity, cellular distribution or cellular uptake of the oligonucleotide.
  • moieties include but are not limited to lipid moieties such as a cholesterol moiety, cholic acid, a thioether, e.g., hexyl-S-tritylthiol, a thiocholesterol, an aliphatic chain, e.g., dodecandiol or undecyl residues, a phospholipid, e.g., di- hexadecyl-rac-glycerol or tri ⁇ thyl-ammonium 1 ,2-di-0-hexadecyl-rac-glycero-3-H- phosphonate, a polyamine or a polyethylene glycol chain, or adamantane acetic acid, a palmity
  • the present invention also includes antisense compounds which are chimeric compounds.
  • "Chimeric” antisense compounds or “chimeras,” in the context of this invention, are antisense compounds, particularly oligonucleotides, which contain two or more chemically distinct regions, each made up of at least one monomer unit, i.e., a nucleotide in the case of an oligonucleotide compound.
  • oligonucleotides typically contain at least one region wherein the oligonucleotide is modified so as to confer upon the oligonucleotide increased resistance to nuclease degradation, increased cellular uptake, and/or increased binding affinity for the target nucleic acid.
  • An additional region of the oligonucleotide may serve as a substrate for enzymes capable of cleaving RNA:DNA or RNA:RNA hybrids.
  • RNase H is a cellular endonuclease which cleaves the RNA strand of an RNA: DNA duplex.
  • RNA target Activation of RNase H, therefore, results in cleavage of the RNA target, thereby greatly enhancing the efficiency of oligonucleotide inhibition of gene expression. Consequently, comparable results can often be obtained with shorter oligonucleotides when chimeric oligonucleotides are used, compared to phosphorothioate deoxyoligonucleotides hybridizing to the same target region.
  • Cleavage of the RNA target can be routinely detected by gel electrophoresis and, if necessary, associated nucleic acid hybridization techniques known in the art.
  • Chimeric antisense compounds of the invention may be formed as composite structures of two or more oligonucleotides, modified oligonucleotides, oligonucleosides and/or oligonucleotide mimetics as described above. Such compounds have also been referred to in the art as hybrids or gapmers.
  • the antisense compounds used in accordance with this invention may be conveniently and routinely made through the well-known technique of solid phase synthesis.
  • Equipment for such synthesis is sold by several vendors including, for example, Applied Biosystems (Foster City, CA). Any other means for such synthesis known in the art may additionally or alternatively be employed. It is well known to use similar techniques to prepare oligonucleotides such as the phosphorothioates and alkylated derivatives.
  • the antisense compounds of the invention are synthesized in vitro and do not include antisense compositions of biological origin, or genetic vector constructs designed to direct the in vivo synthesis of antisense molecules.
  • the compounds of the invention may also be admixed, encapsulated, conjugated or otherwise associated with other molecules, molecule structures or mixtures of ⁇ compounds, as for example, liposomes, receptor targeted molecules, oral, rectal, topical or other formulations, for assisting in uptake, distribution and/or absorption.
  • the antisense compounds of the invention encompass any pharmaceutically acceptable salts, esters, or salts of such esters, or any other compound which, upon administration to an animal including a human, is capable of providing (directly or indirectly) the biologically active metabolite or residue thereof. Accordingly, for example, the disclosure is also drawn to prodrugs and pharmaceutically acceptable salts of the compounds of the invention, pharmaceutically acceptable salts of such prodrugs, and other bioequivalents.
  • prodrug indicates a therapeutic agent that is prepared in an inactive form that is converted to an active form (i.e., drug) within the body or cells thereof by the action of endogenous enzymes or other chemicals and/or conditions.
  • prodrug versions of the oligonucleotides of the invention are prepared as SATE [(S-acetyl-2-thioethyl) phosphate] derivatives.
  • pharmaceutically acceptable salts refers to physiologically and pharmaceutically acceptable salts of the compounds of the invention: i.e., salts that retain the desired biological activity of the parent compound and do not impart undesired toxicological effects thereto.
  • salts formed with cations such as sodium, potassium, ammonium, magnesium, calcium, polyamines such as spermine and spermidine, etc.
  • acid addition salts formed with inorganic acids for example hydrochloric acid, hydrobromic acid, sulfuric acid, phosphoric acid, nitric acid and the like
  • salts formed with organic acids such as, for example, acetic acid, oxalic acid, tartaric acid, succinic acid, maleic acid, fumaric acid, gluconic acid, citric acid, malic acid, ascorbic acid, benzoic acid, tannic acid, palmitic acid, alginic acid, polyglutarnic acid, naphthalenesulfonic acid, methanesulfonic acid, p- toluenesulfonic acid, naphthalenedisulfonic acid, polygalacturonic acid
  • the compounds of the invention can be utilized in pharmaceutical compositions by adding an effective amount of an antisense compound to a suitable pharmaceutically acceptable diluent or carrier.
  • Use of the antisense compounds and methods of the invention may also be useful prophylactically, e.g., to prevent or delay infection, inflammation or tumor formation, for example.
  • the antisense compounds of the invention are useful for research and diagnostics, because these compounds hybridize to nucleic acids encoding HIF-1 , enabling sandwich and other assays to easily be constructed to exploit this fact.
  • Hybridization of the antisense oligonucleotides of the invention with a nucleic acid encoding HIF-1 can be detected by means known in the art. Such means may include conjugation of an enzyme to the oligonucleotide, radiolabelling of the oligonucleotide or any other suitable detection means. Kits using such detection means for detecting the level of HIF-1 in a sample may also be prepared.
  • the present invention also includes pharmaceutical compositions and formulations which include the antisense compounds of the invention.
  • the pharmaceutical compositions of the present invention may be administered in a number of ways depending upon whether local or systemic treatment is desired and upon the area to be treated. Administration may be topical (including ophthalmic and to mucous membranes including vaginal and rectal delivery), pulmonary, e.g., by inhalation or insufflation of powders or aerosols, including by nebulizer; intratracheal, intranasal, epidermal and transdermal), oral or parenteral. Parenteral administration includes intravenous, intraarterial, subcutaneous, intraperitoneal or intramuscular injection or infusion; or intracranial, e.g. , intrathecal or intraventricular administration. Oligonucleotides with at least one 2'- O-methoxyethyl modification are believed to be particularly useful for oral administration.
  • compositions and formulations for topical administration may include transdermal patches, ointments, lotions, creams, gels, drops, suppositories, sprays, liquids and powders.
  • Conventional pharmaceutical carriers, aqueous, powder or oily bases, thickeners and the like may be necessary or desirable
  • compositions and formulations for oral administration include powders or granules, suspensions or solutions in water or non-aqueous media, capsules, sachets or tablets. Thickeners, flavoring agents, diluents, emulsifiers, dispersing aids or binders may be desirable.
  • compositions and formulations for parenteral, intrathecal or intraventricular administration may include sterile aqueous solutions which may also contain buffers, diluents and other suitable additives such as, but not limited to, penetration enhancers, carrier compounds and other pharmaceutically acceptable carriers or excipients.
  • compositions of the present invention include, but are not limited to, solutions, emulsions, and liposome-containing formulations. These compositions may be generated from a variety of components that include, but are not limited to, preformed liquids, self-emulsifying solids and self-emulsifying semisolids.
  • Example 1- Growth of cancer cell lines Cancer cells used to determine the effect of oligonucleotide compounds were obtained from the following sources: Human OVCAR-3 (ovary), MCF-7 (breast, hormone-dependent), HeLa (cervix), PC3 (prostate), HepG2 (liver), and A549 (lung), HT-29 (colon), PANC-1 (pancreas), Caki-1 (kidney) from the American Type Culture Collection (ATCC) (Manassas, VA); U251 (brain), from Riken Cell Bank (Japan); MKN-45 (stomach) from the German Collection of Microorganisms and Cell Cultures (DSMZ) (Germany); UMRC2 (kidney) and Lox IMVI (melanoma) from the United States National Cancer Institute (Bethesda, MD).
  • ATCC American Type Culture Collection
  • VA Manassas, VA
  • U251 brain
  • Riken Cell Bank Japan
  • MKN-45
  • phosphorothioates were conducted the same manner as for the corresponding phosphodiester oligonucleotides except the standard oxidation bottle was replaced by 0.2 M 3H-1 ,2-benzodithiole-3-one 1 ,1 -dioxide in acetonitrile for the stepwise thiation of the phosphite linkages. After cleavage from the controlled pore glass column and deblocking in concentrated ammonium hydroxide, the oligonucleotide compound was heated in the presence of ammonium hydroxide at 55 °C overnight.
  • Solutions A and B were combined and incubated at room temperature for 15 minutes.
  • cells were washed once with 2 ml of serum free medium or PBS and 800 ⁇ l of serum free medium (Opti-MEM) were added to each well.
  • the combined solution A and B was added to each well and mixed gently. Subsequently cells were incubated for 3 hours at 37 °C, the medium was replaced with regular medium and incubated for the indicated time.
  • Opti-MEM serum free medium
  • LIPOFECTAMINE 2000 reagent was diluted (1 :50) in 25 ⁇ l OPTI-MEM for 5 minutes at room temperature.
  • the oligonucleotide and reagent were mixed and incubated at room temperature for 20 minutes to allow complex formation, and then the complex was added directly to the cells in their growth medium and gently mixed. Subsequently cells were incubated for 4 hours at 37°C, and then the medium was replaced with regular medium and incubated for the indicated time.
  • Example 4 Inhibition of HIF-1 ⁇ x mRNA expression by antisense oligonucleotides
  • the antisense oligonucleotides were then tested for their ability to down- regulate, or inhibit, the expression of mRNA encoding HIF-1 ⁇ .
  • the level of expression of HIF-1 ⁇ mRNA in cells transfected with the antisense oligonucleotides was measured by RT-PCR analysis. Samples were taken at 2 hours after transfection (change of media), RNA was isolated and subjected to RT-PCR analysis.
  • RNA-STAT kit TEL-TEST, Inc., Friendswood, TX
  • media were removed from the two 6-well plates and total 0.5 ml RNA-STAT solution was added and mixed by pipetting several times, and transferred to an eppendorf tube.
  • RNA precipitate was centrifuged at 14,000 rpm for 10 minutes. The resulting pellet was washed with 70 % ethanol, dried briefly and reconstituted with 20 ⁇ l water. RNA concentration was determined by spectrophotometer. RT reaction was carried out using M-MLV enzyme kit (Invitrogen).
  • RNA 5 ⁇ g of total RNA was used to synthesize cDNA in 20 ⁇ l RT reaction.
  • First-strand cDNA was synthesized by incubating total RNA, oligo dT (0.5 mg) and dNTP (0.5 mM) mixture at 65 °C for 5 minutes and by quick-chilling on ice.
  • First-strand buffer, 7.4 mM DTT and 1 ⁇ L M-MLV Reverse Transcriptase (200 units) was added to the above reaction mixture and incubated at 37 °C for 50 minutes and the enzyme inactivation was followed at 70 °C for 15 minutes.
  • HIF-1 cDNA synthesized by RT reaction was measured by PCR using Sapphire RCR mix (SuperBio Inc., Seoul, Korea) with appropriate primers.
  • beta-actin was used as an internal PCR control. Primers for beta-actin were 5' C CCATGC CATCCTGCGTCTG 3' (Seq . Id. No. 7) and 5' ACGGAGTACTTGCGCTCAG 3' (Seq. Id. No. 8). PCR products were analyzed on 1.5 % agarose gel by electrophoresis.
  • oligonucleotide A total of 124 oligonucleotides were initially screened, and the results from the preferred four are shown in Table 1 below. Each oligonucleotide was retested to confirm the down-regulation of mRNA expression level. Each reaction was performed in duplicate.
  • RX-0047, RX-0073, RX-0149 and RX-0158 are new sequences designed by the inventors. These were chosen as representatives from two regions found in the reference, the 3' UTR and ORF regions of the HIF-1 a gene, both exhibiting the highest % inhibition for that region according to the test used in that reference. All of the new sequences exhibited enhanced % inhibition over the reference, using the test described herein. However, it was found that % inhibition did not correlate with cytotoxicity, as discussed in Example 6. Subsequently, two oligonucleotides that exhibited both high % inhibition of HIF-1 mRNA expression and cytotoxicity in UMRC2 cells were selected for testing in 12 other cancer cell lines. A list of the nucleotides tested is shown in Table 2, below.
  • Fig. 1 shows down-regulation of HIF-1 mRNA level in 13 cancer cell lines (UMRC2, OVCAR-3, MKN-45, A549, PC3, U251 , Lox IMVI, HeLa, HepG2, HT-29, Caki-1 , PANC-1 and MCF-7) after transfection with 0.3 ⁇ M RX-0047 and RX- 0149.
  • High level down-regulation of HIF-1 was observed in UMRC2, PANC-1 , OVCAR-3, MCF-7, Lox IMVI, A549 and PC3 cell lines, moderate level down- regulation was observed in HT-29 and Caki-1 cells, and low level down-regulation was observed in HeLa, HepG2, MKN-45, and U251.
  • the level of down-regulation of HIF-1 mRNA expression by RX-0047 and RX-0149 was similar except RX-0047-treated cells showed little higher level of down-regulation than RX-0149-treated in a few cell lines.
  • Example 5 Western blot analysis of HIF-1 protein levels Various cancer cell lines were transfected as described above in Example
  • the pellet was resuspended in 0.3-0.5 ml of CE buffer, pH 7.6 (10 mM HEPES, 60 mM KCI, 1 mM EDTA, 1 mM DTT, 1 mM PMSF, 1x protease inhibitor cocktail and 0.1 mM NaVO 4 ) with 0.5 % NP40 and cells were allowed to swell on ice for 5 minutes.
  • the preparation was spun at 2000 rpm for 5 minutes.
  • the cytoplasmic extract was removed from the pellet and transferred to a new tube.
  • the nuclei were washed gently with 0.5 ml of CE buffer without NP40. The nuclei were centrifuged as above at 2,000 rpm for 5 minutes.
  • NE buffer pH 8.0 (20 mM Tris-HCI, 420 mM NaCI, 1.5 mM MgCI 2 , 0.2 mM EDTA, 1 mM PMSF, 25 % glycerol, 0.1 mM NaVO 4 , and 1x protease inhibitor cocktail) was added to nuclear pellet and vortexed to resuspend the pellet.
  • the extract was incubated on ice for 40 minutes with a periodic vortexing to resuspend the pellet and the CE and NE fractions were spun at maximum speed for 15 minutes to pellet any nuclei. The supernatant was transferred to a new tube (soluble nuclear fraction) and stored at -70°C.
  • BCA protein assay reagent (Pierce Biotechnology, Rockford, IL) was used to measure protein concentration. Crude cell extracts were used to determine HIF-1 protein expression by SDS-PAGE and subsequent Western analysis using an anti-HIF1 antibody (Transduction Labs, Lexington, KY). Anti- beta-actin antibody (Santa Cruz Biotechnology) was used as an internal control. Results are shown in Fig. 2. Both RX-0047 and RX-0149 demonstrated inhibition of HIF-1 protein expression, to a greater or lesser degree in all cell lines.
  • Example 6 Cell Cytotoxicity Test Human cancer cell lines were used to test cell cytotoxicity of experimental oligonucleotides. Sulforhodamine B (“SRB”) method [Skehan et al., J. National Cancer Institute, 82: 1107-1112 (1990)] was used to assess the cell survival after RX-oligonucleotide transfection.
  • SRB Sulforhodamine B
  • Cells were plated onto a 96-well plate and transfected with the oligonucleotides the next day. Following a 72-hour incubation period, the surviving cells were stained with sulforhodamine B and measured using a microplate reader. Briefly, 1 ,000-10,000 cells were plated onto each well in a 96-well plate and transfected with experimental oligomers using Lipofectamine 2000 reagent (Invitrogen). After 4 hour incubation, the transfection agent was removed and the fresh media were added to each well. After 72 hours incubation, media were removed.
  • TCA trichloroacetic acid
  • the experimental compounds which showed down-regulation of HIF-1 mRNA were used to test their effect on UMRC2 cell viability.
  • the following oligo compounds, RX-0047, RX-0073, RX-0149 and RX-0158 were used for cytotoxicity test.
  • RX-0047 and RX-0149 showed the most potent cell cytotoxic effect compared with the other oligonucleotides tested.
  • RX-0118 and RX- 0121 new oligonucleotides that had exhibited 74 and 45 % inhibition of mRNA respectively, did not exhibit as much cytotoxicity as RX-0047 and RX-0149. Therefore, the test for mRNA inhibition did not correlate with cytotoxicity.
  • RX- 0047 reduced cell viability in the following human cancer cell lines; PC3 (prostate), U251 (brain), HeLa (cervix), OVCAR-3 (ovary), Lox IMVI (melanoma), HepG2 (liver), MCF-7 (breast), UMRC2 (kidney), MKN-45 (stomach), PANC-1 (pancreas), HT-29 (colon), Caki-1 (kidney) and A549 (lung).
  • PC3 prostate
  • U251 brain
  • HeLa cervix
  • OVCAR-3 ovary
  • Lox IMVI melanoma
  • HepG2 liver
  • MCF-7 breast-7
  • UMRC2 kidney
  • MKN-45 tomach
  • PANC-1 pancreas
  • HT-29 colon
  • Caki-1 Kidney
  • A549 lung
  • 0.1 ⁇ M of RX-0047 caused more than 50 % cell death in all 13 cell lines tested. Similar results were obtained for RX-0149. Again, cytotoxicity of RX- 0149 was demonstrated in all cell lines, and it increased with concentration to varying degrees among the different cell lines.
  • 0.1 ⁇ M of RX-0149 in PC3, U251, HeLa, OVCAR-3, Lox IMVI, MCF-7, MKN-45, A549, Cak ⁇ -1 and UMRC2 caused more than 50 % of cell death. But more than 50 % of cells in PANC-1 , HT-29 and HepG2 survived at 0.1 ⁇ M.
  • the experimental oligonucleotides were screened for relative effective dosage. Thirteen different cancer cell lines were transfected with RX-0047 or RX-0149 at concentrations ranging from 0.01 ⁇ M to 1 ⁇ M and after 72 hours post-transfection, cells were stained with sulforhodamine B and the number of surviving cells were counted using Bio-Rad Microplate reader (Bio-Rad Laboratories). The IC50 value, or concentration of drug needed to kill half the cells, was calculated using the KaleidaGraph Software (Synergy Software, Reading, PA) program. The results are reported in Table 3, below.
  • RX-0149 the 18- and 16-mer versions of RX-0149 showed similar inhibition of HIF-1 mRNA expression as the 20-mer version of RX-0149. However, when the sequence was truncated to the 14-, and 10-mer versions of RX-0149, the inhibition became insignificant. This indicates that for RX-0047, the 18- and 16-mer versions also worked as efficiently as the 20-mer version. For RX-0149, the 20-mer full-length sequence is required to achieve the maximum inhibition of HIF-1 mRNA expression.
  • oligomers comprising either 5 or 10 nucleotides, both upstream and downstream from the sequence where the 20-mer of RX-0047 was derived, showed a measurable inhibition of HIF-1 mRNA expression.
  • Cytotoxicity was tested using the same oligomers comprising 5 or 10 nucleotides upstream and downstream from the sequence where 20-mer of RX- 0047 was derived in UMRC2 cell line. All 4 modified oligomers demonstrated cytotoxic effects comparable to the 20-mer of RX-0047, consistent with the RT- PCR data. The truncated versions of RX-0047 and RX-0149 described above also showed a similar inhibition pattern of cancer cell proliferation as observed in RT-PCR analysis.
  • Example 9 Ex Vivo Xenograft Study
  • RX-0047 in animal models, an ex vivo xenograft study of nude mice was conducted.
  • the A549 human lung cancer cell line was grown in a 4:1 mixture of Dulbecco's modified Eagle's medium and medium 199 supplemented with 10% cosmic calf serum (HyClone, Logan, UT). Cells were maintained at 37°C under 5% CO 2 .
  • a marker gene, luciferase was introduced into tumor cells. The following methods were utilized. Cells were infected with luciferase using a lentiviral vector containing the luciferase gene and a G418/neo selection marker. Cells were incubated for 24 hours in the presence of viral supernatant. Media was changed following the infection, and the G418 selection was initiated 3-4 days following the infection. Luciferase-positive cells were confirmed using a luminometer.
  • mice (Nu/Nu; Harlan Sprague Dawley, Inc., Indianapolis, IN) were maintained in pathogen-free conditions within the animal resources center (ARC) at University of Texas Southwestern Medical Center and treated according to ARC and IACUC guidelines.
  • ARC animal resources center
  • mice were ⁇ -irradiated and 1 x
  • mice 10 6 cells were introduced intravenously through the tail vein. Animals were imaged using luciferase-based bioluminescence imaging each week. Mice were terminated either based on negative results (after 3-4 months) or were imaged each week until the tumor burden exceeded 10% of the host animal's normal body weight (1-2 cm in diameter for an adult mouse) per ARC/IACUC guidelines.
  • Table 4 shows the measurement ofluciferase-base bioluminescence as an indicator of tumor growth in control and RX-0047-treated athymic nude mice sc- implanted with A549 human lung carcinoma xenografts.
  • RX-0047 is a potent anti-tumor agent in tumor xenograft model.

Abstract

L'invention concerne de nouveaux composés oligonucléotidiques anti-sens, RX-0047 et RX-0149, qui inhibent l'expression du HIF-1 et induisent également une cytotoxicité dans plusieurs lignées cellulaires cancéreuses.
PCT/US2004/002344 2003-01-28 2004-01-28 Oligonucleotides anti-sens inhibant l'expression du hif-1 WO2004066949A2 (fr)

Priority Applications (7)

Application Number Priority Date Filing Date Title
JP2006503098A JP4546454B2 (ja) 2003-01-28 2004-01-28 Hif−1発現を抑制するアンチセンスオリゴヌクレオチド
BR0407070-4A BRPI0407070A (pt) 2003-01-31 2004-01-28 Oligonucleotìdeos antisentido que inibem a expressão de hif-1
AT04706034T ATE441710T1 (de) 2003-01-31 2004-01-28 Antisense oligonukleotide, die die ausprägungsstufe von hif-1 hemmen
DE602004022921T DE602004022921D1 (de) 2003-01-31 2004-01-28 Antisense oligonukleotide, die die ausprägungsstufe von hif-1 hemmen
CA2513398A CA2513398C (fr) 2003-01-31 2004-01-28 Oligonucleotides anti-sens inhibant l'expression du hif-1
EP04706034A EP1601325B1 (fr) 2003-01-31 2004-01-28 Oligonucleotides anti-sens inhibant l'expression du hif-1
AU2004207576A AU2004207576B8 (en) 2003-01-28 2004-01-28 Antisense oligonucleotides that inhibit expression of HIF-1

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US76618503A 2003-01-28 2003-01-28
US10/766,185 2003-01-28
US44436703P 2003-01-31 2003-01-31
US60/444,367 2003-01-31

Publications (3)

Publication Number Publication Date
WO2004066949A2 true WO2004066949A2 (fr) 2004-08-12
WO2004066949A8 WO2004066949A8 (fr) 2004-11-11
WO2004066949A3 WO2004066949A3 (fr) 2005-12-01

Family

ID=32829853

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2004/002344 WO2004066949A2 (fr) 2003-01-28 2004-01-28 Oligonucleotides anti-sens inhibant l'expression du hif-1

Country Status (3)

Country Link
JP (1) JP4546454B2 (fr)
AU (1) AU2004207576B8 (fr)
WO (1) WO2004066949A2 (fr)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2013177415A1 (fr) * 2012-05-23 2013-11-28 The Ohio State University Compositions à nanoparticules lipidiques utilisées pour l'administration d'oligonucléotides antisens
KR101390966B1 (ko) 2010-12-30 2014-06-30 주식회사 삼양바이오팜 Hifla의 발현을 저해하는 siRNA 및 이를 포함하는 항암 조성물

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6222018B1 (en) 1995-06-06 2001-04-24 The Johns Hopkins University School Of Medicine Hypoxia inducible factor-1 and method of use

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU5717400A (en) * 1999-06-14 2001-01-02 Cancer Research Ventures Limited Cancer therapy
WO2002034291A2 (fr) * 2000-10-26 2002-05-02 The Brigham And Women's Hospital, Inc. Compositions et procedes servant a traiter des malignites hematologiques et la resistance a des medicaments multiples
WO2003085110A2 (fr) * 2002-04-05 2003-10-16 Santaris Pharma A/S Composes oligomeres servant a moduler l'expression du facteur hif-1$g(a)
US7144999B2 (en) * 2002-11-23 2006-12-05 Isis Pharmaceuticals, Inc. Modulation of hypoxia-inducible factor 1 alpha expression

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6222018B1 (en) 1995-06-06 2001-04-24 The Johns Hopkins University School Of Medicine Hypoxia inducible factor-1 and method of use

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
BLAGOSKLONNY, INTERNATIONAL J. ONCOL., vol. 19, 2001, pages 257 - 262
See also references of EP1601325A4
ZHONG ET AL., CANCER RES., vol. 59, 1999, pages 5830 - 5835

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR101390966B1 (ko) 2010-12-30 2014-06-30 주식회사 삼양바이오팜 Hifla의 발현을 저해하는 siRNA 및 이를 포함하는 항암 조성물
WO2013177415A1 (fr) * 2012-05-23 2013-11-28 The Ohio State University Compositions à nanoparticules lipidiques utilisées pour l'administration d'oligonucléotides antisens
US10307490B2 (en) 2012-05-23 2019-06-04 The Ohio State University Lipid nanoparticle compositions for antisense oligonucleotides delivery

Also Published As

Publication number Publication date
AU2004207576A1 (en) 2004-08-12
WO2004066949A8 (fr) 2004-11-11
JP2006520195A (ja) 2006-09-07
AU2004207576B8 (en) 2008-11-20
JP4546454B2 (ja) 2010-09-15
AU2004207576B2 (en) 2008-10-23
WO2004066949A3 (fr) 2005-12-01

Similar Documents

Publication Publication Date Title
JP4658936B2 (ja) 肺癌を治療するための組成物および方法
MXPA04009361A (es) Oligomeros de nucleobases iap anti-sentido y sus usos.
KR20110017005A (ko) Tgf-베타 수용체 유전자 발현 억제용 조성물 및 방법
KR20170107958A (ko) 트랜스페린 수용체 (TfR)에 대한 RNA 압타머
US7122527B2 (en) Use of antisense oligonucleotides to inhibit the expression of human Akt-1
US7205283B2 (en) Antisense oligonucleotides that inhibit expression of HIF-1
EP1966377A2 (fr) Modulation de l'expression d' eif4e-bp2
US20050153918A1 (en) Methods and compositions relating to hnRNP A1, A1B, A2, and B1 nucleic acid molecules
US20020068711A1 (en) Arrest of proliferation of highly glycolytic tumors
AU2004207576B2 (en) Antisense oligonucleotides that inhibit expression of HIF-1
Zvereva et al. Oligonucleotide inhibitors of telomerase: prospects for anticancer therapy and diagnostics
US20060089322A1 (en) Antisense oligonucleotides for identifying drug targets and enhancing cancer therapies
US20050191672A1 (en) Antisense oligonucleotides and RNA-interfering molecules targeting PAK4
KR100986465B1 (ko) Oip5 유전자의 신규한 용도
CA2534996A1 (fr) Utilisation du silencage sirna dans la prevention des metastases
WO2008106796A1 (fr) Induction d'apoptose et inhibition de prolifération cellulaire par modulation de l'activité carnitine palmitoyltransférase 1c

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BW BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE EG ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NA NI NO NZ OM PG PH PL PT RO RU SC SD SE SG SK SL SY TJ TM TN TR TT TZ UA UG US UZ VC VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): BW GH GM KE LS MW MZ SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IT LU MC NL PT RO SE SI SK TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
CFP Corrected version of a pamphlet front page
CR1 Correction of entry in section i

Free format text: IN PCT GAZETTE 33/2004 UNDER (30) DELETE "10/766,18, 28 JANUARY 2003 (28.01.2003), US"

WWE Wipo information: entry into national phase

Ref document number: 2513398

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 2004207576

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 1020057013673

Country of ref document: KR

WWE Wipo information: entry into national phase

Ref document number: 2006503098

Country of ref document: JP

WWE Wipo information: entry into national phase

Ref document number: 20048032331

Country of ref document: CN

WWE Wipo information: entry into national phase

Ref document number: 2004706034

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 2004207576

Country of ref document: AU

DPEN Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed from 20040101)
WWP Wipo information: published in national office

Ref document number: 1020057013673

Country of ref document: KR

WWP Wipo information: published in national office

Ref document number: 2004706034

Country of ref document: EP

ENP Entry into the national phase

Ref document number: PI0407070

Country of ref document: BR