CA2480311A1 - Oligomeric compounds for the modulation of hif-1alpha expression - Google Patents

Oligomeric compounds for the modulation of hif-1alpha expression Download PDF

Info

Publication number
CA2480311A1
CA2480311A1 CA002480311A CA2480311A CA2480311A1 CA 2480311 A1 CA2480311 A1 CA 2480311A1 CA 002480311 A CA002480311 A CA 002480311A CA 2480311 A CA2480311 A CA 2480311A CA 2480311 A1 CA2480311 A1 CA 2480311A1
Authority
CA
Canada
Prior art keywords
compound
lna
hif
alpha
oligonucleotide
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Granted
Application number
CA002480311A
Other languages
French (fr)
Other versions
CA2480311C (en
Inventor
Anja Molhart Hog
Charlotte Albaek Thrue
Paul E. G. Kristjansen
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Roche Innovation Center Copenhagen AS
Original Assignee
Individual
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Individual filed Critical Individual
Publication of CA2480311A1 publication Critical patent/CA2480311A1/en
Application granted granted Critical
Publication of CA2480311C publication Critical patent/CA2480311C/en
Anticipated expiration legal-status Critical
Expired - Fee Related legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7088Compounds having three or more nucleosides or nucleotides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • 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
    • A61P3/00Drugs for disorders of the metabolism
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • 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
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • 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
    • 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
    • C07H21/04Compounds containing two or more mononucleotide units having separate phosphate or polyphosphate groups linked by saccharide radicals of nucleoside groups, e.g. nucleic acids with deoxyribosyl as saccharide radical
    • 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
    • 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
    • C12N2310/323Chemical structure of the sugar modified ring structure
    • C12N2310/3231Chemical structure of the sugar modified ring structure having an additional ring, e.g. LNA, ENA
    • 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
    • C12N2310/331Universal or degenerate base
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/33Chemical structure of the base
    • C12N2310/334Modified C
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/33Chemical structure of the base
    • C12N2310/334Modified 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
    • C12N2310/33Chemical structure of the base
    • C12N2310/335Modified T or U
    • 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 ===---===

Abstract

Oligonucleotides directed against the hypoxia-inducible factor-l.alpha.. (HI F- 1.alpha.)- gene are provided for modulating the expression of HIF-1.alpha.. The compositions comprise oligonucleotides, particularly antisense oligonucleotides, targeted to nucleic acids encoding the HIF-1.alpha.. Metho ds of using these compounds for modulation of HIF-1.alpha.. expression and for the treatment of diseases associated with the hypoxia-inducible factor- 1.alpha. are provided. Examples of diseases are cancer and pre-eclampsia. Th e oligonucleotides may be composed of deoxyribonucleosides, a nucleic acid analogue, or Locked Nucleic Acid (LNA) or a combination thereof.

Description

Oligomeric Compounds For The Modulation HIF-la Expression CROSS-REFERENCE TO RELATED APPLICATIONS
The present application is a continuation of U.S. Provisional application serial number 60/370,126, filed April 5, 2002 of which application is fully incorporated herein by reference.
FIELD OF THE INVENTION
The present invention provides compositions and methods for modulating the expression of HIF-la. In particular, this invention relates to oligomeric compounds and preferred such compounds are oligonucleotides, which are specifically hybridisable with nucleic acids encoding HIF-la. The oligonucleotide compounds have been shown to modulate the expression of HIF-la and pharmaceutical preparations thereof and their use as treatment of cancer diseases and pre-eclampsia are disclosed.
BACKGROUND OF THE INVENTION
Solid tumors must establish a blood supply and have enhanced glucose metabolism to grow beyond a few millimeters. How they sense hypoxia, and respond by activating hypoxia-inducible genes and secreting angiogeneic factors to establish a blood system is central to cancer biology. Many tumors contain hypoxic microenvironments, which have been associated with malignant progression, metastasis and resistance to radiotherapy and chemotherapy.
The discovery of hypoxia-inducible factor-1 (HIF-1) gave some insight into the regulation of hypoxia-inducible genes (LTS 5882914 and W09639426;
W09948916). HIF-1 is composed of two subunits HIF-1 a and HIF-1 [3 and it binds hypoxia-response elements (HREs) in enhancers of genes encoding angiogenic factors such as VEGF and glycolysis-related proteins such as glycolytic enzymes and glucose transporter l and 3 (GLU-l and 3). .
It has been demonstrated that engineered down-regulation of HIF-la by intratumoral gene transfer of an antisense HIF-1a plasmid leads to the down-regulation of VEGF, and decreased tumor microvessel density (WO 0076497, Sun X
et al, Gene Therapy (2001) 8, 638-645). The plasmid contained a 320-by cDNA
fragment encoding 5'-end of HIF-la (nucleotides 152-454; Genebank AF003698).
Furthermore, in the International Patent Application cited above a method was described based on that the expression vector should be used in conjunction with an ixmnunotherapeutic agent. However, a major weakness with the expression plasmid approach is that it will not be suitable as a therapeutic agent due to its size and the nuclease sensitivity of the expression product.
Besides the plasmid expressing a HIF-la fragment a few antisense oligonucleotides targeting HIF-1 a have been designed as research tools to study a specific biological mechanism or biological target. For example the antisense inhibition of HIF-la expression in hypoxic explants have been shown to inhibit expression of TGF/3 (Caniggia, L, et al J. of Clinical Investigation, March 2000, 105, 577-587). In this particular study, only one antisense oligonucleotide was synthesized, a phosphorothioate targeted against the sequence adjacent to the AUG
initiation codon of HIF-1 a mRNA. The sequences were HIF-1 a 5'-GCCGGCGCCCTCCAT-3' and the HIF-la down regulation was demonstrated at mRNA level. This oligo has been used to study the role of HIF-1 a in extravillous trophoblast outgrowth and invasion, and implicated at potential role of HIF-la in pre-eclampsia (Caniggia, I. et al Placenta (2000), 21, Supplement A, , Trophoblast Research 14, S25-S30).
Another study, using the same oligonucleotide sequence as above, showed that antisense inhibition of HIF-1 a resulted in loss of peroxisome proliferator-active receptors (PPARs) (Narravula, S. and Colgan S.P., J. of Immunology, 2001, 166, 7543-7548). The above mentioned oligo has also been used to show that nickel requires HIF-la to induce plasminogen activator inhibitior-1 (PAI-1) (Andrew, A.S.
Klei L.R., Barchowsky A, Am. J. Physiol. Lung Cell Mol. Physiol. 281, L607-L615, 2001).
_2_ A single antisense oligonucleotide has also been used to study the two splice variants of the hypoxia-inducible factor HIF-1a as potential dimerization partner of ARNT2 in neurons. The antisense oligonucleotide was the phosphorothioate-modification of the sequence: 5'-TCTTCTCGTTCTCGCC-3'. Treating cells with this oligonucleotide resulted in inhibition of [3H]thymidine incorporation, but did not have an effect on apoptosis in normoxic cells (Drutel et.al. (2000) Eur. J.
Neurosci. 12, 3701-3708).
Furthermore, a single antisense oligonucleotide for HIF-1 a have been showed to inhibit the increased gene expression of cardiac endothelin (ET)-l and it was hypothesized that HIF-la is involved in increased myocardial expression of the gene in heart failure (I~akinuma, Y. et al, Circulation, 2001; 103, 2387-2394). The antisense oligonucleotide had the following sequence:
CCTCCATGGCGAATCGGTGC.
Currently, there are no known therapeutic antisense agents, which effectively inhibit the synthesis of HIF-1 a and which can be used for the treatment of a disease.
Consequently, there is a need for agents capable of effectively inhibiting the HIF-la function to be used in the treatment of e.g. cancer and pre-eclampsia.
SUMMARY OF THE INVENTION
The present invention is directed to oligomeric compounds, particularly LNA
antisense oligonucleotides, which are targeted to a nucleic acid encoding HIF-la and which modulate the expression of the HIF-1a. Pharmaceutical and other compositions comprising the oligomeric compounds of the invention are also provided.
Further provided are methods of modulating the expression of HIF-1 a in cells or tissues comprising contacting said cells or tissues with one or more of the oligomeric compounds or compositions of the invention. Also disclosed are methods of treating an animal or a human, suspected of having or being prone to a disease or condition, associated with expression of HIF-la by administering a therapeutically or prophylactically effective amount of one or more of the oligomeric compounds or compositions of the invention. Further, methods of using oligomeric compounds for the inhibition of expression of HIF-1a and for treatment of diseases associated with these HIF-1 a are provided. Examples of such diseases are different types of cancer, particularly common cancers, as e.g. primary and metastatic breast, colorectal, prostate, pancreas, other GI-cancers, lung, cervical, ovarian, and brain tumors, as well as pre-eclampsia, inflammatory bowel disease and Alzheimers disease. Other examples axe cancer of the colon, liver, thyroid, kidney, testes, stomach, intestine, bowel, esophagus, spinal cord, sinuses, bladder or urinary tract.
BRIEF DESCRIPTION OF THE FIGURES
Figure 1 shows a Western blot of HIF-la protein. Cells were treated with the different oligos at 100 nM for 4 hours. The cells were allowed to grow for 18 hours before they were exposed to severe hypoxia for 6 hours.
Figure 2 shows a Western blot of HIF-la protein. U87 cells were treated with three of the oligos at 200 nM for 4 hours. The cells were exposed to severe hypoxia for 18 hours immediately after the treatment.
Figure 3 shows Western blots of HIF-la, VEGF Glutl and tubulin protein in U87 cells treated with oligo Cur0813. Cells were treated with oligo for 24 hours at 100 nM, 200 nM, 300 nM and 400 nM. The cells were exposed to severe hypoxia for 18 hours immediately after the treatment.
Figure 4 shows Western blots of HIF-la and tubulin protein in U87 cells treated with mismatch oligos (Cur0960 and Cur0961). Cells were treated with oligo for 24 hours at 100 nM, 200 nM, 300 nM and 400 nM. The cells were exposed to severe hypoxia for 18 hours immediately after the treatment.
Figure 5 shows Western blots of HIF-la, VEGF and tubulin protein in 15PC3 cells treated with oligo Cur813. Cells were treated with oligo for 16 hours at 125 nM, 25 nM, 5 nM and 1 nM. The cells were exposed to severe hypoxia for 6 hours immediately after the treatment.
Figure 6 shows Western blots of HlF-la and tubulin protein in 15PC3 cells treated with different oligos at 5 nM for 16 hours. The 'cells were exposed to severe hypoxia for 6 hours immediately after the treatment.
Figure 7 shows Western blots of HIF-la and tubulin protein in U373 cells treated with different oligos at 100 nM for 6 hours. The cells were exposed to severe hypoxia for 20 hours immediately after the treatment.

Figure 8 shows Western blots of HIF-1 a and tubulin protein in U373 cells treated with different oligos at 100 nM for 6 hours. The cells were exposed to severe hypoxia for 20 hours immediately after the treatment.
Figure 9 shows growth curves of U373 xenograft tumours treated with PBS or Cur813 at 5 mg/kg/day i.p. lx daily for 7 days. Bars represent standard errors.
Figure 10 shows human HIF-la sequence, using GenBank accession number NM 001530, incorporated herein as SEQ m NO:1.
DEFINITION
As used herein, the terms "target nucleic acid" encompass DNA encoding the hypoxia-inducible factor or encoding hypoxia-inducible factor-la (HIF-la), RNA
(including pre-mRNA and mRNA) transcribed from such DNA, and also cDNA
derived from such RNA.
As used herein, the term "gene" means the gene including exons, introns, non-coding 5'and 3'regions and regulatory elements and all currently known variants thereof and any further variants, which may be elucidated.
As used herein, the terms "oligomeric compound" refers to an oligonucleotide which can induce a desired therapeutic effect in humans through for example binding by hydrogen bonding to either a target gene "Chimeraplast" and "TFO", to the RNA transcripts) of the target gene "antisense inhibitors", "siRNA", "ribozymes" and oligozymes" or to the proteins) encoding by the target gene "aptamer", spiegeliner" or "decoy".
As used herein, the term "mRNA" means the presently known mRNA
transcripts) of a targeted gene, and any further transcripts, which may be identified.
As used herein, the term "modulation" means either an increase (stimulation) or a decrease (inhibition) in the expression of a gene. In the present invention, inhibition is the preferred form of modulation of gene expression and mRNA is a preferred target.
As used herein, the term "targeting" an antisense compound to a particular target nucleic acid means providing the antisense oligonucleotide to the cell, animal or human in such a way that the antisense compound are able to bind to and modulate the function of its intended target.

As used herein, "hybridisation" means hydrogen bonding, which may be Watson-Crick, Hoogsteen, reversed Hoogsteen hydrogen bonding, etc. between complementary nucleoside or nucleotide bases. Watson and Crick showed approximately fifty years ago that deoxyribo nucleic acid (DNA) is composed of two strands which are held together in a helical configuration by hydrogen bonds formed between opposing complementary nucleobases in the two strands. The four nucleobases, commonly found in DNA are guanine (G), adenine (A), thymine (T) and cytosine (C) of which the G nucleobase pairs with C, and the A nucleobase pairs with T. In RNA the nucleobase thymine is replaced by the nucleobase uracil (L>], which similarly to the T nucleobase pairs with A. The chemical groups in the nucleobases that participate in standard duplex formation constitute the Watson-Crick face.
Hoogsteen showed a couple of years later that the purine nucleobases (G and A) in addition to their Watson-Crick face have a Hoogsteen face that can be recognised from the outside of a duplex, and used to bind pyrimidine oligonucleotides via hydrogen bonding, thereby forming a triple helix structure.
In the context of the present invention "complementary" refers to the capacity for precise pairing between two nucleotides or nucleoside sequences with one another. For example, if a nucleotide at a certain position of an oligonucleotide is capable of hydrogen bonding with a nucleotide at the corresponding position of a DNA or RNA molecule, then the oligonucleotide and the DNA or RNA axe considered to be complementary to each other at that position. The DNA or RNA
and the oligonucleotide are considered complementary to each other when a sufficient number of nucleotides in the oligonucleotide can form hydrogen bonds with corresponding nucleotides in the target DNA or RNA to enable the formation of a stable complex. To be stable in vitro or in vivo the sequence of an antisense compound need not be 100% complementary to its target nucleic acid. The terms "complementary" and "specifically hybridisable" thus imply that the antisense compound binds sufficiently strongly and specifically to the target molecule to provide the desired interference with the normal function of the target whilst leaving the function of non-target mRNAs unaffected.
The term "Nucleic Acid Analogues" refers to a non-natural nucleic acid binding compound. Nucleic Acid Analogues are described in e.g. Freier &
Altmann (Nucl. Acid Res., 1997, 25, 4429-4443) and Uhlinann (Curr. Opinion in Drug &

Development (2000, 3(2): 293-213). Scheme 1 illustrates selected examples.
O O B O O $ O O $ O O B
O p o~ O O O F
O=P-S_ O_P-O- O-P-O- ~o, O
Phosphorthioate 2'-O-Methyl 2'-MOE 2'-Fluoro O $ ~ O $ $
O ~ $ O
/ O ~ ~O ~' ~ / ~ O
V
O O ~' ~,. N
O P-O- N~
H
2'-AP ~A CeNA PNA
0 0 $ O F $ O O $ O O $
O
/ o N
o P N ~ O=~ O_ O-P-O_ \ O P-O-Mo holino 2'-F-ANA OH 3'-Phosphoramidate 2'-(3-hydroxy)propyl O O $
O
O=P-BH3-Boranophosphates Scheme 1 The term "LNA" refers to an oligonucleotide containing one or more bicyclic nucleoside analogues also referred to as a LNA monomer. LNA monomers are described in WO 9914226 and subsequent applications, W00056746, W00056748, W00066604, W000125248, W00228875, W02002094250 and PCT/DI~02/00488.
One particular example of a thymidine LNA monomer is the (1S,3R, 4R, 7S)-7-hydroxy-1-hydroxyrnethyl-5-methyl-3-(thymin-lyl)-2,5-dioxa-bicyclo[2:2:1]heptane.
_7_ The term "oligonucleotide" refers, in the context of the present invention, to an oligomer (also called oligo) or nucleic acid polymer (e.g. ribonucleic acid (RNA) or deoxyribonucleic acid (DNA)) or nucleic acid analogue of those known in the art, preferably Locked Nucleic Acid (LNA), or a mixture thereof. This term includes oligonucleotides composed of naturally occurring nucleobases, sugars and internucleoside (backbone) linkages as well as oligonucleotides having non-naturally-occurring portions which function similarly or with specific improved functions. A
fully or partly modified or substituted oligonucleotides are often preferred over native forms because of several desirable properties of such oligonucleotides such as for instance, the ability to penetrate a cell membrane, good resistance to extra-and intracellular nucleases, high affinity and specificity for the nucleic acid target. The LNA analogue is particularly preferred exhibiting the above-mentioned properties.
By the term "unit" is understood a monomer.
The term "at least one" comprises the integers larger than or equal to 1, such as 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17 and so forth.
The term "thio-LNA" comprises a locked nucleotide in which at least one of X or Y in Scheme 2 is selected from S or -CHz-S-. Thio-LNA can be in both beta-D
and alpha-L-configuration.
The term "amino-LNA" comprises a locked nucleotide in which at least one of X or Y in Scheme 2 -N(H)-, N(R)-, CHZ-N(H)-, -CH2-N(R)- where R is selected from hydrogen and Cl_4-alkyl. Amino-LNA can be in both beta-D and alpha-L-configuration.
The term "oxy-LNA" comprises a locked nucleotide in which at least one of X
or Y in Scheme 2 represents O or -CHa-O-. Oxy-LNA can be in both beta-D and alpha-L-configuration.
The term "ena-LNA" comprises a locked nucleotide in which Y in Scheme 2 is -CH2-O-.
By the term "alpha-L-LNA" comprises a locked nucleotide represented as shown in Scheme 3.
By the term "LNA derivatives" comprises all locked nucleotide in Scheme 2 except beta-D-methylene LNA e.g. thio-LNA, amino-LNA, alpha-L-oxy-LNA and ena-LNA.
_g_ DETAILED DESCRIPTION OF THE INVENTION
The present invention employs oligomeric compounds, particularly antisense oligonucleotides, for use in modulating the function of nucleic acid molecules encoding HIF-la. The modulation is ultimately a change in the amount of HIF-la produced. In one embodiment this is accomplished by providing antisense compounds, which specifically hybridise with nucleic acids encoding HIF-la.
The modulation is preferably an inhibition of the expression of HIF-la, which leads to a decrease in the number of functional proteins produced. HIF-1 may be involved in angiogenesis as well as red blood cell proliferation, cellular proliferation, iron metabolism, glucose and energy metabolism, pH regulation, tissue invasion, apoptosis, multi-drug resistance, cellular stress response or matrix metabolism.
Antisense and other oligomeric compounds of the invention, which modulate expression of the target, are identified through experimentation or though rational design based on sequence information on the target and know-how on how best to design an oligomeric compound against a desired target. The sequences of these compounds are preferred embodiments of the invention. Likewise, the sequence motifs in the target to which these preferred oligomeric compounds are complementary (referred to as "hot spots") are preferred sites for targeting.
Preferred oligomeric compounds according to the invention are SEQ ID NO 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, 100, 101, 102, 103, 104, 105, 106, 107, 108, 109, 110, 111, 112, 113, 114 or 115 and their sequences are presented in table 1 and table 2. The oligomeric compounds according to the invention are potent modulators of target. This is showed experimentally both in vitro and in vivo. In vitro inhibition of target is shown in table 1 and Figure 1-8 using three different cancer cell lines. Figure 9 shows in vivo down regulation of target. Furthermore, the oligomeric compounds are shown to be potent inhibitors in much lower concentration than e.g. the standard condition for phosphorthioate antisense oligonucleotides. Figure 5 and 6 show inhibition of compounds of the invention down to 5 nM. Inhibition of HIF-1a by oligomeric compounds of the invention can also inhibit the expression of Vascular Endothelial Growth Factor (VEGF) known to be involved in angiogenesis and Glucose Transporter-1 (GLUT-1) known to be involved in glucose uptake as shown in fig.

and 5. Various designs of oligomeric compounds shown in table 2 targeted to two motifs were identified as potent inhibitors of the target as shown in fig. 1 and 7. A
genewalk was performed using oligomeric compounds from table 1, and the effect of the potent oligomeric compounds is shown in fig. 8. All the above-mentioned experimental observations show that the compounds according to the invention can constitute the active compound in a pharmaceutical composition.
Furthermore, the oligomeric compounds according to the invention may inhibit HIF-1cc under normoxia and hypoxia.
In one embodiment of the invention the oligomeric compounds are containing at least on unit of chemistry termed LNA (Locked Nucleic Acid).
LNA monomer typically refers to a bicyclic nucleoside analogue, as described in the International Patent Application WO 99/14226 and subsequent applications, W00056746, W00056748, W00066604, W000125248, W00228875, WO2002094250 and PCT/DK02/00488 all incorporated herein by reference.
Preferred LNA monomers structures are exemplified in Scheme 2 2o Y
Scheme2 X and Y are independently selected among the groups -O-, -S-, -N(H)-, N(R)-, -CH~-or -CH- (if part of a double bond), -CHa-O-, -CH2-S-, -CH2-N(H)-, -CHa-N(R)-, -CH2-CH2- or -CHa-CH- (if part of a double bond), -CH=CH-, where R is selected from hydrogen and Cl~-alkyl. The asymmetric groups may be found in either orientation.
In Scheme 2 the 4 chiral centers are shown in a fixed configuration. However, also comprised in this invention are compounds of the general Scheme 2 in which the chiral centers are found in different configurations. Thus, each chiral center in Scheme 2 can exist in either R or S configuration. The definition of R
(rectus) and S
(sinister) are described in the IUPAC 1974 Recommendations, Section E, Fundamental Stereochemistry: The rules can be found in Pure Appl. Chem. 45, 13-30, (1976) and in "Nomenclature of organic Chemistry" Pergamon, New York, 1979.
Z and Z* are independently selected among an internucleoside linkage, a terminal group or a protecting group.
The internucleoside linkage may be -O-P(O)2-O-, -O-P(O,S)-O-, -O-P(S)2-O-, -S-P(O)2-O-, -S-P(O,S)-O-, -S-P(S)2-O-, -O-P(O)2-S-, -O-P(O,S)-S-, -S-P(O)Z-S-, -O-PO(RH)-O-, O-PO(OCH3)-O-, -O-PO(NRH)-O-, -O-PO(OCH2CHZS-R)-O-, -O-PO(BH3)-O-, -O-PO(NHRH)-O-, -O-P(O)2-NRH-, -NRH-P(O)Z-O-, -NRH-CO-O-, -NRH-CO-NRH-, -O-CO-O-, -O-CO-NRH-, -NRH-CO-CH2-, -O-CH2-CO-NRH-, -O-CHZ-CH2-NRH-, -CO-NRH-CH2-, -CH2-NRH-CO-, -O-CHZ-CH2-S-, -S-CH2-CHZ_ O-, -S-CHZ-CH2-S-, -CHa-S02-CH2-, -CHa-CO-NRH-, -O-CHa-CH2-NRH-CO -, -CHZ-NCH3-O-CH2-, where RH is selected form hydrogen and C1_4-alkyl, The terminal groups are selected independently from hydrogen, azido, halogen, cyano, vitro, hydroxy, Prot-O-, Act-O-, mercapto, Prot-S-, Act-S-, Cl_s-alkylthio, amino, Prot-N(RH)-, Act-N(RH)-, mono- or di(C1_6-alkyl)amino, optionally substituted C1_6-alkoxy, optionally substituted Cl_6-alkyl, optionally substituted C2_6-alkenyl, optionally substituted Ca_6-alkenyloxy, optionally substituted C2_6-alkynyl, optionally substituted C~_6-alkynyloxy, monophosphate-or protected monophosphate, monothiophosphate-or protected monothiophosphate, diphosphate-or protected diphosphate, dithiophosphate - or protected dithiophosphate, triphosphate- or protected triphosphate, trithiophosphate - or protected trithiophosphate.
Examples of such protection groups on the phosphate residues are S-acetylthioethyl (SATE) or S-pivaloylthioethyl (t-butyl-SATE), DNA intercalators, photochemically active groups, thermochemically active groups, chelating groups, reporter groups, ligands, carboxy, sulphono, hydroxymethyl, Prot-O-CH2-, Act-O-CH2-, aminomethyl, Prot-N(RH)-CHa-Act-N(RH)-CHZ-, carboxymethyl, sulphonomethyl, where Prot is a protection group for -OH, -SH, and -NH(RH), respectively, Act is an activation group for -OH, -SH, and -NH(RH), respectively, and RH is selected from hydrogen and Cl_6-alkyl;
The protection groups of hydroxy substituents comprises substituted trityl, such as 4,4'-dimethoxytrityloxy (DMT), 4-monomethoxytrityloxy (MMT), and trityloxy, optionally substituted 9-(9-phenyl)xanthenyloxy (pixyl), optionally substituted methoxytetrahydropyranyloxy (mthp), silyloxy such as trimethylsilyloxy (TMS), triisopropylsilyloxy (TIPS), tert-butyldimethylsilyloxy (TBDMS), triethylsilyloxy, and phenyldimethylsilyloxy, tent-butylethers, acetals (including two hydroxy groups), acyloxy such as acetyl or halogen substituted acetyls, e.g.
chloroacetyloxy or fluoroacetyloxy, isobutyryloxy, pivaloyloxy, benzoyloxy and substituted benzoyls, methoxymethyloxy (MOM), benzyl ethers or substituted benzyl ethers such as 2,6-dichlorobenzyloxy (2,6-Cl2Bzl). Alternatively when Z or Z*
is hydroxyl they may be protected by attachment to a solid support optionally through a linker.
When Z or Z* is amino groups illustrative examples of the amino protection protections are fluorenylmethoxycarbonylamino (Fmoc), tent-butyloxycarbonylamino (BOC), trifluoroacetylamino, allyloxycarbonylamino (allot, AOC), Z benzyl-oxycarbonylamino ( Cbz), substituted benzyloxycarbonylaminos such as 2-chloro benzyloxycarbonylamino (2-C1Z), ~ monomethoxytritylamino (MMT), dimethoxytritylamino (DMT), phthaloylamino, and 9-(9-phenyl)xanthenylamino (pixyl).
In the embodiment above, Act designates an activation group for -OH, -SH, and -NH(RH). In a preferred embodiment such activators mediates couplings to other residues, monomers. After such successful couplings the act-group is converted to an internucleoside linkage. Such activation groups are, e.g., selected from optionally substituted O-phosphoramidite, optionally substituted O-phosphortriester, optionally substituted O-phosphordiester, optionally substituted H-phosphonate, and optionally substituted O-phosphonate.
In the present context, the term "phosphoramidite" means a group of the formula -P(OR")-N(Ry)Z, wherein R" designates an optionally substituted alkyl group, e.g. methyl, 2-cyanoethyl, or benzyl, and each of Ry designate optionally substituted alkyl groups, e.g. ethyl or isopropyl, or the group -N(RY)Z forms a morpholino group (-N(CHZCHa)20). R" preferably designates 2-cyanoethyl and the two RY are preferably identical and designate isopropyl. Thus, an especially relevant phosphoramidite is N,N-diisopropyl-O-(2-cyanoethyl)phosphoramidite.
B constitutes a natural or non-natural nucleobase and selected among adenine, cytosine, 5-methylcytosine, isocytosine, pseudoisocytosine, guanine, thymine, uracil, 5-bromouracil, 5-propynyluracil, 5-propyny-6-fluoroluracil, 5-methylthiazoleuracil, 6-aminopurine, 2-aminopurine, inosine, diaminopurine, 7-propyne-7-deazaadenine, 7-propyne-7-deazaguanine, 2-chloro-6-aminopurine.
Particularly preferred bicyclic structures are shown in Scheme 3 below:
*Z
Z
when at least one X=O
Scheme 3 Where X is -O-, -S-, -NH-, and N(RH), Z and Z* are independently selected among an internucleoside linkage, a terminal group or a protecting group.
The internucleotide linkage may be -O-P(O)2-O-, -O-P(O,S)-O-, -O-P(S)Z-O-, _S_P(O)a_O_~ _S_P(O~S)_O_~ _S_P(S)a_O_~ _O_P(O)a_S_~ _O_P(O~S)_S_~
_S_P(O)a_S_~
-O-PO(RH)-O-, O-PO(OCH3)-O-, -O-PO(NRH)-O-, -O-PO(OCH2CH2S-R)-O-, -O-PO(BH3)-O-, -O-PO(NHRH)-O-, -O-P(O)2-NRH-, -NRH-P(O)a-O-, -NRH-CO-O-, where RH is selected form hydrogen and C1~-alkyl.
The terminal groups are selected independently among from hydrogen, azido, halogen, cyano, vitro, hydroxy, Prot-O-, Act-O-, mercapto, Prot-S-, Act-S-, Cl_s-alkylthio, amino, Prot-N(RH)-, Act-N(RH)-, mono- or di(Cl_6-alkyl)amino, optionally substituted C1_6-alkoxy, optionally substituted C1_6-alkyl, optionally substituted monophosphate, monothiophosphate, diphosphate, dithiophosphate triphosphate, trithiophosphate, where Prot is a protection group for -OH, -SH, and -NH(RH), respectively, Act is an activation group for -OH, -SH, and -NH(RH), respectively, and RH is selected from hydrogen and C1_6-alkyl.
The protection groups of hydroxy substituents comprises substituted trityl, such as 4,4'-dimethoxytrityloxy (DMT), 4-monomethoxytrityloxy (MMT), optionally substituted 9-(9-phenyl)xanthenyloxy (pixyl), optionally substituted methoxytetra-hydropyranyloxy (mthp), silyloxy such as trimethylsilyloxy (TMS), triisopropylsilyloxy (TIPS), tent-butyldimethylsilyloxy (TBDMS), triethylsilyloxy, and phenyldimethylsilyloxy, text-butylethers, acetals (including two hydroxy groups), acyloxy such as acetyl Alternatively when Z or Z* is hydroxyl they may be protected by attachment to a solid support optionally through a linker.
When Z or Z* is amino groups illustrative examples of the amino protection protections are fluorenylmethoxycarbonylamino (Fmoc), tart-butyloxycarbonylamino (BOC), trifluoroacetylamino, allyloxycarbonylamino (alloc, AOC), monomethoxytritylamino (MMT), dimethoxytritylamino (DMT), phthaloylamino.
In the embodiment above, Act designates an activation group for -OH, -SH, and -NH(RH). In a preferred embodiment such activators mediates couplings to other residues, monomers. After such successful couplings the act-group is converted to an internucleoside linkage. Such activation groups are, e.g., selected from optionally substituted O-phosphoramidite, optionally substituted O-phosphortriester, optionally substituted O-phosphordiester, optionally substituted H-phosphonate, and optionally substituted O-phosphonate.
In the present context, the term "phosphoramidite" means a group of the formula -P(OR")-N(RY)2, wherein R" designates an optionally substituted alkyl group, e.g. methyl, 2-cyanoethyl, and each of Ry designate optionally substituted alkyl groups, R" preferably designates 2-cyanoethyl and the two Ry are preferably identical and designate isopropyl. Thus, an especially relevant phosphoramidite is N,N-diisopropyl-O-(2-cyanoethyl)phosphoramidite.
B constitutes a natural or non-natural nucleobase and selected among adenine, cytosine, 5-methylcytosine, isocytosine, pseudoisocytosine, guanine, thymine, uracil, 5-bromouracil, 5-propynyluracil, 6-aminopurine, 2-aminopurine, inosine, diaminopurine, 2-chloro-6-aminopurine.
Therapeutic principle A person skilled in the art will appreciate that oligomeric compounds containing LNA can be used to combat HIF-la linked diseases by many different principles, which thus falls within the spirit of the present invention.
For instance, LNA oligomeric compounds may be designed as antisense inhibitors, which are single stranded nucleic acids that prevent the production of a disease causing protein, by intervention at the mRNA level. Also, they may be designed as Ribozymes or Oligozymes which are antisense oligonucleotides which in addition to the target binding domains) comprise a catalytic activity that degrades the target mRNA (ribozymes) or comprise an external guide sequence (EGS) that recruit an endogenous enzyme (RNase P) which degrades the target mRNA (oligozymes) Equally well, the LNA oligomeric compounds may be designed as siRNA's which are small double stranded RNA molecules that are used by cells to silence specific endogenous or exogenous genes by an as yet poorly understood "antisense-like" mechanism.
LNA oligomeric compounds may also be designed as Aptamers (and a variation thereof, termed Spiegelmers) which are nucleic acids that through intra-molecular hydrogen bonding adopt three-dimensional structures that enable them to bind to and block their biological targets with high affinity and specificity.
Also, LNA
oligomeric compounds may be designed as Decoys, which are small double-stranded nucleic acids that prevent cellular transcription factors from transactivating their target genes by selectively blocking their DNA binding site.
Furthermore, LNA oligomeric compounds may be designed as Chimeraplasts, which are small single stranded nucleic acids that are able to specifically pair with and alter a target gene sequence. LNA containing oligomeric compounds exploiting this principle therefore may be particularly useful for treating HIF-1 a, linked diseases that are caused by a mutation in the HIF-1a, gene.
Finally, LNA oligomeric compounds may be designed as TFO's (triplex forming oligonucleotides), which are nucleic acids that bind to double stranded DNA
and prevent the production of a disease causing protein, by intervention at the RNA
transcription level.
Dictated in part by the therapeutic principle by which the oligonucleotide is intended to operate, the LNA oligomeric compounds in accordance with this invention preferably comprise from about 8 to about 60 nucleobases i.e. from about 8 to about 60 linked nucleosides. Particularly preferred compounds are antisense oligonucleotides comprising from about 12 to about 30 nucleobases and most preferably are ,antisense compounds comprising about 12-20 nucleobases.
Refernng to the above principles by which an LNA oligomeric compound can elicit its therapeutic action the target of the present invention may be the HIF-la gene, the mRNA or the protein. In the most preferred embodiment the LNA
oligomeric compounds is designed as an antisense inhibitor directed against the HIF-1 a pre-mRNA or HIF-1 a mRNA. The oligonucleotides may hybridize to any site along the HIF-la pre-mRNA or mRNA such as sites in the 5' untranslated leader, exons, introns and 3'untranslated tail.
In a preferred embodiment, the oligonucleotide hybridizes to a portion of the human HIF-1 a pre-mRNA or mRNA that comprises the translation-initiation site.
More preferably, the HIF-la oligonucleotide comprises a CAT sequence, which is complementary to the AUG initiation sequence of the HIF-1 a pre-mRNA or RNA.
In another embodiment, the HIF-1 a oligonucleotide hybridizes to a portion of the splice donor site of the human HIF-la pre-mRNA. In yet another embodiment, HIF-la oligonucleotide hybridizes to a portion of the splice acceptor site of the human HIF-1 a pre-mRNA. In another embodiment, the HIF-1 a oligonucleotide hybridizes to portions of the human HIF-la pre-mRNA or mRNA involved in polyadenylation, transport or degradation.
The skilled person will appreciate that preferred oligonucleotides are those that hybridize .to a portion of the HIF-la pre-mRNA or mRNA whose sequence does not commonly occur in transcripts from unrelated genes so as to maintain treatment specificity.
The oligomeric compound of the invention are designed to be sufficiently complementary to the target to provide the desired clinical response e.g. the oligomeric compound must bind with sufficient strength and specificity to its target to give the desired effect. In one embodiment, said compound modulating HIF-la is designed so as to also modulate other specific nucleic acids which do not encode HIF-1 a.
It is preferred that the oligomeric compound according to the invention is designed so that infra- and intermolecular oligonucleotide hybridisation is avoided.
In many cases the identification of an LNA oligomeric compound effective in modulating HIF-la activity in vivo or clinically is based on sequence information on the target gene. However, one of ordinary skill in the art will appreciate that such oligomeric compounds can also be identified by empirical testing. As such HIF-1 a oligomeric compounds having, for example, less sequence homology, greater or fewer modified nucleotides, or longer or shorter lengths, compared to those of the preferred embodiments, but which nevertheless demonstrate responses in clinical treatments, are also within the scope of the invention.
Antisense drugs In one embodiment of the invention the oligomeric compounds are suitable antisense drugs. The design of a potent and safe antisense drug requires the fine-tuning of diverse parameters such as affinity/specificity, stability in biological fluids, cellular uptake, mode of action, pharmacokinetic properties and toxicity.
Affinit~pecificity: LNA with an oxymethylene 2'-O, 4'-C linkage ((3-D-oxy-LNA), exhibits unprecedented binding properties towards DNA and RNA target sequences. Likewise LNA derivatives, such as amino-, thio- and a-L-oxy-LNA
display unprecedented affinities towards complementary RNA and DNA and in the case of thio-LNA the affinity towards RNA is even better than with the (3-D-oxy-LNA.
In addition to these remarkable hybridization properties, LNA monomers can be mixed and act cooperatively with DNA and RNA monomers, and with other nucleic acid analogues, such as 2'-O-alkyl modified RNA monomers. As such, the oligonucleotides of the present invention can be composed entirely of (3-D-oxy-LNA
monomers or it may be composed of (3-D-oxy-LNA in any combination with DNA, RNA or contemporary nucleic acid analogues which includes LNA derivatives such as for instance amino-, thio- and a-L-oxy-LNA . The unprecedented binding affinity of LNA towards DNA or RNA target sequences and its ability to mix freely with DNA, RNA and a range of contemporary nucleic acid analogues has a range of important consequences according to the invention for the development of effective and safe antisense compounds.
Firstly, in one embodiment of the invention it enables a considerable shortening of the usual length of an antisense oligo (from 20-25 mers to, e.g., 12-15 mers) without compromising the affinity required for pharmacological activity.
As the intrinsic specificity of an oligo is inversely correlated to its length, such a shortening will significantly increase the specificity of the antisense compound towards its RNA
target. One embodiment of the invention is to, due to the sequence of the human genome is available and the annotation of its genes rapidly progressing, identify the shortest possible, unique sequences in the target mRNA.
In another embodiment, the use of LNA to reduce the size of oligos significantly eases the process and prize of manufacture thus providing the basis for antisense therapy to become a commercially competitive treatment offer for a diversity of diseases.
In another embodiment, the unprecedented affinity of LNA can be used to substantially enhance the ability of an antisense oligo to hybridize to its target mRNA
in-vivo thus significantly reducing the time and effort required for identifying an active compound as compared to the situation with other chemistries.
In another embodiment, the unprecedented affinity of LNA is used to enhance the potency of antisense oligonucleotides thus enabling the development of compounds with more favorable therapeutic windows than those currently in clinical trials.
When designed as an antisense inhibitor, the oligonucleotides of the invention bind to the target nucleic acid and modulate the expression of its cognate protein.
Preferably, such modulation produces an inhibition of expression of at least 10% or 20% compared to the normal expression level, more preferably at least a 30%, 40%, 50%, 60%, 70%, 80%, or 90% inhibition compared to the normal expression level.
Typically, the LNA oligonucleotides of the invention will contain other residues than (3-D-oxy-LNA such as native DNA monomers, RNA monomers, N3 °-PS'phosphoroamidates, 2'-F, 2'-O-Me, 2'-O-methoxyethyl (MOE), 2'-O-(3-aminopropyl) (AP), hexitol nucleic acid (HNA), 2 °-F-arabino nucleic acid (2'-F-ANA) and D-cyclohexenyl nucleoside (CeNA). Also, the (3-D-oxy-LNA-modified oligonucleotide may also contain other LNA units in addition to or in place of an oxy-LNA group. In particular, preferred additional LNA units include thio-LNA or amino-LNA monomers in either the D-13 or L-oc configurations or combinations thereof or ena-LNA. In general, an LNA-modified oligonucleotide will contain at least about 5, 10, 15 or 20 percent LNA units, based on total nucleotides of the oligonucleotide, more typically at least about 20, 25, 30, 40, 50, 60, 70, 80 or 90 percent LNA units, based on total bases of the oligonucleotide.
Stability in biological fluids: One embodiment of the invention includes the S incorporation of LNA monomers into a standard DNA or RNA oligonucleotide to increase the stability of the resulting oligomeric compound in biological fluids e.g.
through the increase of resistance towards nucleases (endonucleases and exonucleases). The extent of stability will depend on the number of LNA
monomers used, their position in the oligonucleotide and the type of LNA monomer used.
Compared to DNA and phosphorothioates the following order of ability to stabilize an oligonucleotide against nucleolytic degradation can be established: DNA «
phosphorothioates ~ oxy-LNA < a-L-LNA < amino-LNA < thio-LNA.
Given the fact that LNA is compatible with standard DNA synthesis and mixes freely with many contemporary nucleic acid analogues nuclease resistance of LNA- oligomeric compounds can be further enhanced according to the invention by either incorporating other analogues that display increased nuclease stability or by exploiting nuclease-resistant internucleoside linkages e.g.
phosphoromonothioate, phosphorodithioate, and methylphosphonate linkages, etc.
Mode of action: Antisense compounds according to the invention may elicit their therapeutic action via a variety of mechanisms and may be able to combine several of these in the same compound. In one scenario, binding of the oligonucleotide to its target (pre-mRNA or mRNA) acts to prevent binding of other factors (proteins, other nucleic acids, etc.) needed for the proper function of the target i.e. operate by steric hindrance. For instance, the antisense oligonucleotide may bind to sequence motifs in either the pre-mRNA or mRNA that are important for recognition and binding of transacting factors involved in splicing, poly-adenylation, cellular transport, post-transcriptional modifications of nucleosides in the RNA, capping of the 5'-end, translation, etc. In the case of pre-mRNA splicing, the outcome of the interaction between the oligonucleotide and its target may be either suppression of expression of an undesired protein, generation of alternative spliced mRNA encoding a desired protein or both. In another embodiment, binding of the oligonucleotide to its target disables the translation process by creating a physical block to the ribosomal machinery, i.e. tranlational arrest.
In yet another embodiment, binding of the oligonucleotide to its target interferes with the RNAs ability to adopt secondary and higher order structures that are important for its proper function, i.e. structural interference. For instance, the oligonucleotide may interfere with the formation of stem-loop structures that play crucial roles in different functions, such as providing additional stability to the RNA
or adopting essential recognition motifs for different proteins.
In still another embodiment, binding of the oligonucleotide inactivates the target toward further cellular metabolic processes by recruiting cellular enzymes that degrades the mRNA. For instance, the oligonucleotide may comprise a segment of nucleosides that have the ability to recruit ribonuclease H (RNaseH) that degrades the RNA part of a DNA/RNA duplex. Likewise, the oligonucleotide may comprise a segment which recruits double stranded RNAses, such as for instance RNAseIII
or it may comprise an external guide sequence (EGS) that recruit an endogenous enzyme (RNase P) which degrades the target mRNA. Also, the oligonucleotide may comprise a sequence motif which exhibit RNAse catalytic activity or moieties may be attached to the oligonucleotides which when brought into proximity with the target by the hybridization event disables the target from further metabolic activities.
It has been shown that ~i-D-oxy-LNA does not support RNaseH activity.
However, this can be changed according to the invention by creating chimeric oligonucleotides composed of (3-D-oxy-LNA and DNA, called gapmers. A gapmer is based on a central stretch of 4-12 nt DNA or modified monomers recognizable and cleavable by the RNaseH (the gap) typically flanked by 1 to 6 residues of [3-D-oxy-LNA (the flanks). The flanks can also be constructed with LNA derivatives.
There are other chimeric constructs according to the invention that are able to act via an RNaseH mediated mechanism. A headmer is defined by a contiguous stretch of ~i-D-oxy-LNA or LNA derivatives at the 5'-end followed by a contiguous stretch of DNA
or modified monomers recognizable and cleavable by the RNaseH towards the 3'-end, and a tailiner is defined by a contiguous stretch of DNA or modified monomers recognizable and cleavable by the RNaseH at the 5'-end followed by a contiguous stretch of (3-D-oxy-LNA or LNA derivatives towards the 3'-end. Other chimeras according to the invention, called mixmers consisting of an alternate composition of DNA or modified monomers recognizable and cleavable by RNaseH and (3-D-oxy-LNA and/or LNA derivatives might also be able to mediate RNaseH binding and cleavage. Since a-L-LNA recruits RNaseH activity to a certain extent, smaller gaps of DNA or modified monomers recognizable and cleavable by the RNaseH for the gapmer construct might be required, and more flexibility in the mixmer construction might be introduced. Figure 1 shows an outline of different designs according to the invention.
Pharmacokinetic properties The clinical effectiveness of antisense oligonucleotides depends to a significant extent on their pharmacokinetics e.g. absorption, distribution, cellular uptake, metabolism and excretion. In turn these parameters are guided significantly by the underlying chemistry and the size and three-dimensional structure of the oligonucleotide.
As mentioned earlier LNA according to the invention is not a single, but several related chemistries, which although molecularly different all exhibit stunning affinity towards complementary DNA and RNA, Thus, the LNA family of chemistries are uniquely suited of development oligos according to the invention with tailored phaxmacokinetic properties exploiting either the high affinity of LNA to modulate the size of the active compounds or exploiting different LNA chemistries to modulate the exact molecular composition of the active compounds. In the latter case, the use of for instance amino-LNA rather than oxy-LNA will change the overall charge of the oligo and affect uptake and distribution behavior. Likewise the use of thio-LNA
instead of oxy-LNA will increase the lipophilicity of the oligonucleotide and thus influence its ability to pass through lipophilic barriers such as for instance the cell membrane.
Modulating the pharmacokinetic properties of an LNA oligonucleotide according to the invention may further be achieved through attachment of a variety of different moieties. For instance, the ability of oligonucleotides to pass the cell membrane may be enhanced by attaching for instance lipid moieties such as a cholesterol moiety, a thioether, an aliphatic chain, a phospholipid or a polyamine to the oligonucleotide. Likewise, uptake of LNA oligonucleotides into cells may be enhanced by conjugating moieties to the oligonucleotide that interacts with molecules in the membrane, which mediates transport into the cytoplasm.

Pharmacodynamic properties The pharmacodynamic properties can according to the invention be enhanced with groups that improve oligomer uptake, enhance biostability such as enhanced oligomer resistance to degradation, and/or increase the specificity and affinity of oligonucleotides hybridisation characteristics with target sequence e.g. a mRNA
sequence.
Toxicology There are basically two types of toxicity associated with antisense oligos:
sequence-dependant toxicity, involving the base sequence, and sequence-independent, class-related toxicity. With the exception of the issues related to immunostimulation by native CpG sequence motifs, the toxicities that have been the most prominent in the development of antisense oligonucleotides are independent of the sequence, e.g.
related to the chemistry of the oligonucleotide and dose, mode, frequency and duration of administration. The phosphorothioates class of oligonucleotides have been particularly well characterized and found to elicit a number of adverse effects such as complement activation, prolonged PTT (partial thromboplastin time), thrombocytopenia, hepatotoxicity (elevation of liver enzymes), cardiotoxicity, splenomegaly and hyperplasia of reticuloendothelial cells.
As mentioned earlier, the LNA family of chemistries provide unprecedented affinity, very high bio-stablity and the ability to modulate the exact molecular composition of the oligonucleotide. In one embodiment of the invention, LNA
containing compounds enables the development of oligonucleotides which combine high potency with little- if any- phosphorothioate linkages and which are therefore likely to display better efficacy and safety than contemporary antisense compounds.
Manufacture Oligo- and polynucleotides of the invention may be produced using the polymerisation techniques of nucleic acid chemistry well known to a person of ordinary skill in the art of organic chemistry. Generally, standard oligomerisation cycles of the phosphoramidite approach (S. L. Beaucage and R. P. Iyer, Tetrahedron, 1993, 49, 6123; S. L. Beaucage and R. P. Iyer, Tet~ahedYOh, 1992, 48, 2223) is used, but e.g. H-phosphonate chemistry, phosphortriester chemistry can also be used.
For some monomers of the invention longer coupling time, and/or repeated couplings with fresh reagents, and/or use of more concentrated coupling reagents were used.
The phosphoramidites employed coupled with satisfactory >98% step-wise coupling yields. Thiolation of the phosphate is performed by exchanging the normal, e.g. iodine/pyridine/H20, oxidation used for synthesis of phosphordiester oligomers with an oxidation using Beaucage's reagent (commercially available) other sulfurisation reagents are also comprised. The phosphorthioate LNA oligomers were efficiently synthesised with stepwise coupling yields >= 98 %.
The (3-D-amino-LNA, (3-D-thio-LNA oligonucleotides, a-L-LNA and (3-D-methylamino-LNA oligonucleotides were also efficiently synthesised with step-wise coupling yields >_ 98% using the phosphoramidite procedures.
Purification of LNA oligomeric compounds was done using disposable reversed phase purification cartridges and/or reversed phase HPLC and/or precipitation from ethanol or butanol. Gel electrophoresis, reversed phase HPLC, MALDI-MS, and ESI-MS was used to verify the purity of the synthesized oligonucleotides. Furthermore, solid support materials having immobilised thereto an optionally nucleobase protected and optionally 5'-OH protected LNA are especially interesting as material for the synthesis of LNA containing oligomeric compounds where an LNA monomer is included in at the 3' end. In this instance, the solid support material is preferable CPG, e.g. a readily (commercially) available CPG
material or polystyrene onto which a 3'-functionalised, optionally nucleobase protected and optionally 5'-OH protected LNA is linked using the conditions stated by the supplier for that particular material.
Indications The pharmaceutical composition according to the invention can be used for the treatment of many different diseases. For example HIF-1oc has been found to be overexpressed in various solid human tumours and their metastases, e.g.
tumours of the breast, colon, prostate, pancreas, brain, lung, ovary, gastro-intestinal system, head and neck, liver, bladder and cervix (thong, H. et al., Cancer Research 59, 5830-5835, 1999; Talks, I~.L. et al., American Journal of Pathology 157(2), 411-421, 2000) The methods of the invention is preferably employed for treatment or prophylaxis against diseases caused by cancer, particularly for treatment of cancer as may occur in tissue such as lung, breast, colon, prostate, pancreas, liver, brain, testes, stomach, intestine, bowel, spinal cord, sinuses, cervix, urinary tract or ovaries cancer.
Furthermore, the invention described herein encompasses a method of preventing or treating cancer comprising a therapeutically effective amount of a HIF-la modulating oligomeric compound, including but not limited to high doses of the oligomer, to a human in need of such therapy. The invention further encompasses the use of a short period of administration of a HIF-1 a modulating oligomeric compound.
Normal, non-cancerous cells divide at a frequency characteristic for the particular cell type. When a cell has been transformed into a cancerous state, uncontrolled cell proliferation and reduced cell death results, and therefore, promiscuous cell division or cell growth is a hallmark of a cancerous cell type. Examples of types of cancer, include, but are not limited to, non-Hodgkin's lymphoma, Hodgkin's lymphoma, leukemia (e.g., acute leukemia such as acute lymphocytic leukemia, acute myelocytic leukemia, chronic myeloid leukemia, chronic lymphocytic leukemia, multiple myeloma), colon carcinoma, rectal carcinoma, pancreatic cancer, breast cancer, ovarian cancer, prostate cancer, renal cell carcinoma, hepatoma, bile duct carcinoma, choriocarcinoma, cervical cancer, testicular cancer, lung carcinoma, bladder carcinoma, melanoma, head and neck cancer, brain cancer, cancers of unknown primary site, neoplasms, cancers of the peripheral nervous system, cancers of the central nervous system, tumors (e.g., fibrosarcoma, myxosarcoma, liposarcoma, chondrosarcoma, osteogenic sarcoma, chordoma, angiosarcoma, endotheliosarcoma, lymphangiosarcoma, lymphangioendotheliosarcoma, synovioma, mesothelioma, Ewing's tumor, leiomyosarcoma, rhabdomyosarcoma, squamous cell carcinoma, basal cell carcinoma, adenocarcinoma, sweat gland carcinoma, sebaceous gland carcinoma, papillary carcinoma, papillary adenocarcinomas, cystadenocarcinoma, medullary carcinoma, bronchogenic carcinoma, seminoma, embryonal carcinoma, Wilms' tumor, small cell lung ~ carcinoma, epithelial carcinoma, glioma, astrocytoma, medulloblastoma, craniopharyngioma, ependymoma, pinealoma, hemangioblastoma, acoustic neuroma, oligodendroglioma, meningioma, neuroblastoma, and retinoblastoma), heavy chain disease, metastases, or any disease or disorder characterized by uncontrolled or abnormal cell growth.
Pharmaceutical composition It should be understood that the invention also relates to a pharmaceutical composition, which comprises a least one antisense oligonucleotide construct of the invention as an active ingredient. It should be understood that the pharmaceutical composition according to the invention optionally comprises a pharmaceutical carrier, and that the pharmaceutical composition optionally comprises further antisense compounds, chemotherapeutic compounds, anti-inflammatory compounds, antiviral compounds and/or immuno-modulating compounds.
Salts The oligomeric compound comprised in this invention can be employed in a variety of pharmaceutically acceptable salts. As used herein, the term refers to salts that retain the desired biological activity of the herein identified compounds and exhibit miiumal undesired toxicological effects. Non-limiting examples of such salts can be formed with organic amino acid and base addition salts formed with metal cations such as zinc, calcium, bismuth, barium, magnesium, aluminum, copper, cobalt, nickel, cadmium, sodium, potassium, and the like, or with a cation formed from ammonia, N,N dibenzylethylene-diamine, D-glucosamine, tetraethylammonium, or ethylenediamine; or (c) combinations of (a) and (b); e.g., a zinc tannate salt or the like.
Prodrugs In one embodiment of the invention the oligomeric compound may be in the form of a pro-drug. Oligonucleotides are by virtue negatively charged ions.
Due to the lipophilic nature of cell membranes the cellular uptake of oligonucleotides are reduced compared to neutral or lipophilic equivalents. This polarity "hindrance" can be avoided by using the pro-drug approach (see e.g. Crooke, R. M. (1990 in Crooke, S. T. Antisense research arad Application. Springer-Verlag, Berlin, Germany, vol.
131, pp. 103-140). In this approach the oligonucleotides are prepared in a protected manner so that the oligo is neutral when it is administered. These protection groups are designed in such a way that so they can be removed then the oligo is taken up be the cells. Examples of such protection groups are S-acetylthioethyl (SATE) or S-pivaloylthioethyl (t-butyl-SATE). These protection groups are nuclease resistant and are selectively removed intracellulary.
Conjugates In one embodiment of the invention the oligomeric compound is linked to ligands/conjugates. It is way to increase the cellular uptake of antisense oligonucleotides. This conjugation can take place at the terminal positions 5'/3'-OH
but the ligands may also take place at the sugars and/or the bases. In particular, the growth factor to which the antisense oligonucleotide may be conjugated, may comprise transferrin or folate. Transferrin-polylysine-oligonucleotide complexes or folate-polylysine-oligonucleotide complexes may be prepared for uptake by cells expressing high levels of transferrin or folate receptor. Other examples of conjugates/lingands are cholesterol moieties, duplex intercalators such as acridine, poly-L-lysine, "end-capping" with one or more nuclease-resistant linkage groups such as phosphoromonothioate, and the like.
The preparation of transferrin complexes as carriers of oligonucleotide uptake into cells is described by Wagner et al ., Proc. Natl. Acad. Sci. USA 87, 3410-(1990). Cellular delivery of folate-macromolecule conjugates via folate receptor endocytosis, including delivery of an antisense oligonucleotide, is described by Low et al., U.S. Patent 5,108,921. Also see, Leamon et al., Proc. Natl. Acad. Sci.
88, 5572 (1991).
Formulations The invention also includes the formulation of one or more oligonucleotide compound as disclosed herein. Pharmaceutically acceptable binding agents and adjuvants may comprise part of the formulated drug. Capsules, tablets and pills etc.
may contain for example the following compounds: microcrystalline cellulose, gum or gelatin as binders; starch or lactose as excipients; stearates as lubricants; various sweetening or flavouring agents. For capsules the dosage unit may contain a liquid Garner like fatty oils. Likewise coatings of sugar or enteric agents may be part of the dosage unit. The oligonucleotide formulations may also be emulsions of the active pharmaceutical ingredients and a lipid forming a micellar emulsion.
An oligonucleotide of the invention may be mixed with any material that do not impair the desired action, or with material that supplement the desired action.
These could include other drugs including other nucleoside compounds.
For parenteral, subcutaneous, intradermal or topical administration the formulation may include a sterile diluent, buffers, regulators of tonicity and antibacterials. The active compound may be prepared with carriers that protect against degradation or immediate elimination from the body, including implants or microcapsules with controlled release properties. For intravenous administration the preferred carriers are physiological saline or phosphate buffered saline.
Preferably, an oligomeric compound is included in a unit formulation such as in a pharmaceutically acceptable carrier or diluent in an amount sufficient to deliver to a patient a therapeutically effective amount without causing serious side effects in the treated patient.
Administration 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 (a) oral (b) pulmonary, e.g., by inhalation or insufflation of powders or aerosols, including by nebulizer; intratracheal, intranasal, (c) topical including epidermal, transdermal, ophthalmic and to mucous membranes including vaginal and rectal delivery; or (d) parenteral including intravenous, intraarterial, subcutaneous, intraperitoneal or intramuscular injection or infusion; or intracranial, e.g., intrathecal or intraventricular, administration. In one embodiment the active oligo is administered IV, IP, orally, topically or as a bolus injection or administered directly in to the target organ.
Pharmaceutical compositions and formulations for topical administration may include transdermal patches, ointments, lotions, creams, gels, drops, sprays, suppositories, liquids and powders. Conventional pharmaceutical Garners, aqueous, powder or oily bases, thickeners and the like may be necessary or desirable.
Coated condoms, gloves and the like may also be useful. Preferred topical formulations include those in which the oligonucleotides of the invention are in admixture with a _27_ topical delivery agent such as lipids, liposomes, fatty acids, fatty acid esters, steroids, chelating agents and surfactants. Compositions and formulations for oral administration include but is not restricted to powders or granules, microparticulates, nanoparticulates, suspensions or solutions in water or non-aqueous media, capsules, gel capsules, sachets, tablets or minitablets. 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, earner compounds and other pharmaceutically acceptable carriers or excipients.
Delivery Pharmaceutical 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.
Delivery of drug to tumour tissue may be enhanced by carrier-mediated delivery including, but not limited to, cationic liposomes, cyclodextrins, porphyrin derivatives, branched chain dendrimers, polyethylenimine polymers, nanoparticles and microspheres (Dass CR. J Pharm Pharmacol 2002; 54(1):3-27).
The pharmaceutical formulations of the present invention, which may conveniently be presented in unit dosage form, may be prepared according to conventional techniques well knowxn in the pharmaceutical industry. Such techniques include the step of bringing into association the active ingredients with the pharmaceutical carner(s) or excipient(s). In general the formulations are prepared by uniformly and intimately bringing into association the active ingredients with liquid earners or finely divided solid carriers or both, and then, if necessary, shaping the product.
The compositions of the present invention may be formulated into any of many possible dosage forms such as, but not limited to, tablets, capsules, gel capsules, liquid syrups, soft gels and suppositories. The compositions of the present invention may also be formulated as suspensions in aqueous, non-aqueous or mixed media.
Aqueous suspensions may further contain substances which increase the viscosity of -2~-the suspension including, for example, sodium caxboxymethylcellulose, sorbitol and/or dextran. The suspension may also contain stabilizers.
Combination drug Oligonucleotides of the invention may be used to abolish the effects of HIF-1 a induction by acute hypoxia induced by androgen withdrawal therapy in prostate cancer.
Oligonucleotides of the invention may also be conjugated to active drug substances, for example, aspirin, ibuprofen, a sulfa drug, an antidiabetic, an antibacterial or an antibiotic.
LNA containing oligomeric compounds are useful for a number of therapeutic applications as indicated above. In general, therapeutic methods of the invention include administration of a therapeutically effective amount of an LNA-modified oligonucleotide to a mammal, particularly a human.
In a certain embodiment, the present invention provides pharmaceutical compositions containing (a) one or more antisense compounds and (b) one or more other chemotherapeutic agents which function by a non-antisense mechanism.
When used with the compounds of the invention, such chemotherapeutic agents may be used individually (e.g. mithramycin and oligonucleotide), sequentially (e.g.
mithramycin and oligonucleotide for a period of time followed by another agent and oligonucleotide), or in combination with one or more other such chemotherapeutic agents or in combination with radiotherapy. All chemotherapeutic agents known to a person skilled in the art are here incorporated as combination treatments with compound according to the invention.
Anti-inflammatory drugs, including but not limited to nonsteroidal anti-inflammatory drugs and corticosteroids, antiviral drugs, and immuno-modulating drugs may also be combined in compositions of the invention. Two or more combined compounds may be used together or sequentially.
In another embodiment, compositions of the invention may contain one or more antisense compounds, particularly oligonucleotides, targeted to a first nucleic acid and one or more additional antisense compounds targeted to a second nucleic acid target. Two or more combined compounds may be used together or sequentially.

Dosage Dosing is dependent on severity and responsiveness of the disease state to be treated, and the course of treatment lasting from several days to several months, or until a cure is effected or a diminution of the disease state is achieved.
Optimal dosing S schedules can be calculated from measurements of drug accumulation in the body of the patient.
Optimum dosages may vary depending on the relative potency of individual oligonucleotides. Generally it can be estimated based on ECSOs found to be effective in in vitro and in vivo animal models. In general, dosage is from 0.01 ~g to 1 g per kg of body weight, and may be given once or more daily, weekly, monthly or yearly, or even once every 2 to 10 years or by continuous infusion for hours up to several months. The repetition rates for dosing can be estimated based on measured residence times and concentrations of the drug in bodily fluids or tissues. Following successful treatment, it may be desirable to have the patient undergo maintenance therapy to prevent the recurrence of the disease state.
Uses The LNA containing oligomeric compounds of the present invention can be utilized for as research reagents for diagnostics, therapeutics and prophylaxis. In research, the antisense oligonucleotides may be used to specifically inhibit the synthesis of HIF-la genes in cells and experimental animals thereby facilitating functional analysis of the target or an appraisal of its usefulness as a target for therapeutic intervention. In diagnostics the antisense oligonucleotides may be used to detect and quantitate HIF-la expression in cell and tissues by Northern blotting, in-situ hybridisation or similar techniques. For therapeutics, an animal or a human, suspected of having a disease or disorder, which can be treated by modulating the expression of HIF-1 a is treated by administering antisense compounds in accordance with this invention. Further provided are methods of treating an animal particular mouse and rat and treating a human, suspected of having or being prone to a disease or condition, associated with expression of HIF-la by administering a therapeutically or prophylactically effective amount of one or more of the antisense compounds or compositions of the invention.

Methods The methods of the invention is preferably employed for treatment or prophylaxis against diseases caused by a disease. One embodiment of the invention involves a method of inhibiting the expression of HIF-la, in cells or tissues comprising contacting said cells or tissues with the compound of the invention so that expression of HIF-la is inhibited. Furthermore, another embodiment is a method of modulating expression of a gene involved in a disease comprising contacting the gene or RNA from the gene with an oligomeric compound wherein said compound has a sequence comprising at least an 8 nucleobase portion of SEQ ID NO: 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, 100, 101, 102, 103, 104, 105, 106, 107, 108, 109, 110, 111, 112, 113, 114 or whereby gene expression is modulated. These compounds may comprise one or more LNA units. The compound may hybridizes with messenger RNA of the gene to inhibit expression thereof. Another embodiment is a method of treating a mammal suffering from or susceptible from a cancer disease, comprising, administering to the mammal an therapeutically effective amount of an oligonucleotide targeted to HIF-la that comprises one or more LNA units. The described methods may target a common cancers, as e.g. primary and metastatic breast, colorectal, prostate, pancreas, other GI-cancers, lung, cervical, ovarian, brain, head and neck, cervix, colon, liver, thyroid, kidney, testes, stomach, intestine, bowel, esophagus, spinal cord, sinuses, bladder or urinary tract tumors, as well as pre-eclampsia, inflammatory bowel disease and Alzheimers disease. The method may also modulate angiogenesis as well as red blood cell proliferation, cellular proliferation, iron metabolism, glucose and energy metabolism, pH regulation, tissue invasion, apoptosis, multi-drug resistance, cellular stress response or matrix metabolism comprising contacting a cell with the antisense compound of claim the invention so that the cell is modulated.
All documents mentioned herein are incorporated herein by reference.

EXAMPLES
The present invention has been described with specificity in accordance with certain of its preferred embodiments. Therefore, the following examples serve only to illustrate the invention and are not intended to limit the same.
Example 1 Monomer synthesis Preparation of the monomers shown in Scheme 2 in which Y and X are -O-and Z and Z* are protected -O- is described in great detail in the reference, Koshkin et al, J. OYg. Chem., 2001, 66, 8504-8512; Sorensen et al., J. Am. Chem. Soc., 2002, 124 (10), 2164-2176; Pedersen et al., Synthesis, 2002, 6, 802-809 and references found therein, where the protection groups of Z and Z* are respectively oxy-N,N-diisopropyl-O-(2-cyanoethyl)phosphoramidite and dimethoxytrityloxy. The preparation of monomers of the Scheme 2 in which X is -O- and Y is -S- and -N(CH3)- is described in Rosenbohm, et al. Org. Biomol. Chem., 2003,1, 655 -663.
Example 2 LNA oligonucleotide synthesis All oligonucleotide syntheses are carned out in 1 pmol scale on a MOSS
Expedite instrument platform. The synthesis procedures are essentially carried out as described in the instrument manual. The LNA monomers used were synthesised according Koshin A.A. et al J. Org. Chem., 2001, 66, 8504-8512.
Preparation of the LNA succinyl hemiester 5'-O-Dmt-3'-hydroxy-LNA monomer (500 mg), succinic anhidride (1.2 eq.) and DMAP (1.2 eq.) were dissolved in DCM (35 mL). The reaction was stirred at room temperature overnight. After extractions with NaH2P04 0.1 M pH 5.5 (2x) and brine (lx), the organic layer was further dried with anhydrous Na2S04 filtered and evaporated. The hemiester derivative was obtained in 95 % yield and was used without any further purification.

Preparation of the LNA-CPG resin The above prepared hemiester derivative (90 ~mol) was dissolved in a minimum amount of DMF, DIEA and pyBOP (90 ~,mol) were added and mixed together for 1 min. This pre-activated mixture was combined with LCAA-CPG (500 1~, 80-120 mesh size, 300 mg) in a manual synthesizer and stirred. After 1.5 h at room temperature, the support was filtered off and washed with DMF, DCM and MeOH.
After drying the loading was determined, and resulted to be 57 ~,mol/g.
Phosphorothioate cycles 5'-O-Dmt (A(bz), C(bz), G(ibu), and T) linked to CPG (controlled pore glass) were deprotected using a solution of 3 % trichloroacetic acid (v/v) in dichloromethane. The resin is washed with acetonitrile. Coupling of phosphoramidites (A(bz), G(ibu), 5-methyl-C(bz)) or T -(3-cyanoethylphosphoramidite) is performed by using a solution of 0.08 M of the 5 °-O-Dmt-protected amidite in acetonitrile and activation is done by using DCI (4,5 - dicyanoimidazole) in acetonitrile (0.25 M).
Coupling is carried out in 2 minutes. Thiolation is carried out by using Beaucage reagent (0.05 M in acetonitrile) and is allowed to react for 3 minutes. The support is thoroughly washed with acetonitrile and the subsequent capping capping is carried out by using a solution of and acetic anhydride in THF (CAP A) and N-methylimidazole /pyridine/THF (1:1:8) (CAP B) to cap unreacted 5'- hydroxyl groups. The capping step is then repeated and the cycle is concluded by acetonitrile washing.
LNA cycles 5'-O-Dmt (locA(bz), locC(bz), locG(ibu) or loci) linked to CPG (controlled pore glass) is deprotected by using the same procedure as above. Coupling is performed by using 5'-O-Dmt (locA(bz), locC(bz), locG(ibu) or locT)-[3-cyanoethylphosphoramidite (0.1 M in acetonitrile) and activation is done by DCI
(0.25 M in acetonitrile). Coupling is prolonged to 7 minutes. Capping is done by using acetic anhydride in THF (CAP A) and a solution of N-methylimidazole/pyridine/THF (1:1:8) (CAP B) for 30 sec. The phosphite triester is oxidized to the more stable phosphate triester by using a solution of I2 and pyridine in THF for 30 sec. The support is washed with acetonitrile and the capping step is repeated. The cycle is concluded by thorough acetonitrile wash. Abbreviations:
Dmt:
Dimethoxytrityl and DCI: 4,5-Dicyanoimidazole.
Oligonucleotide Cleavage and Deprotection The oligomers are cleaved from the support and the (3-cyanoethyl protecting group removed by treating the support with 35 % NH40H 1 h at room temperature.
The support is filtered off and the base protecting groups are removed by raising the temperature to 65 °C for 4 hours. The oligosolution is then evaporated to dryness.
Oligonucleotide Purification The oligos are either purified by (reversed-phase) RP-HPLC or (anion exchange) AIE.
RP-HPLC:
Column: VYDACTM cat. No. 218TP1010 (vydac) Flow rate: 3 mL/min Buffer: A 0.1 M ammonium acetate pH 7.6 B acetonitrile Gradient:

Time 0 10 18 22 23 28 B % 0 5 30 100 100 0 IE:

Column: ResourceTM 15Q (amersham pharmacia biotech) Flow rate: 1.2 mL/min Buffer: A 0.1 M NaOH

B 0.1 M NaOH, 2,0 M NaCI

Gradient:

Time 0 1 27 28 32 33 B % 0 25 55 100 100 0 Abbreviations Dmt: Dimethoxytrityl DCI: 4,5-Dicyanoimidazole DMAP: 4-Dimethylaminopyridine DCM: Dichloromethane DMF: Dimethylformamide THF: Tetrahydrofurane DIEA: N,N diisopropylethylamine PyBOP: Benzotria.zole-1-yl-oxy-tris-pyrrolidino-phosphonium hexafluorophosphate Bz: Benzoyl Ibu: Isobutyryl Beaucage: 3H-1,2-Benzodithiole-3-one-1,1-dioxide A(bz), C(bz), G(ibu) or T: LNA-monomers (LNA-locked nucleic acid) Example 3. Cell Culture Antisense compounds and their effect on target nucleic acid expression can be tested in any of a variety of cell types provided that the target nucleic acid or protein is present at measurable levels. This can be routinely determined using, for example, RT-PCR or Northern blot or Western blot analysis. The following cell types are provided for illustrative purposes, but other cell types can be routinely used, provided that the target is expressed in the cell type chosen.
Cell lines were cultured in the appropriate medium as described below and maintained at 37°C at 95-98% humidity and 5% COa. Cells were routinely passaged 2-3 times weekly.
U87-MG: The human glioblastoma cell line U87-MG was cultured in Modified Eagle Medium (MEM) with Earle's salts and 10% Fetal Calf Serum (FCS) U373: The human glioblastoma cell line U373 was cultured in Modified Eagle Medium (MEM) with Earle's salts and 10% Fetal Calf Serum (FCS) 15PC3: The human prostate cancer cell line 15PC3 was kindly donated by Dr. F.
Baas, Neurozintuigen Laboratory, AMC, The Netherlands and was cultured in DMEM (Sigma) + 10% fetal bovine serum (FBS) + Glutamax I + gentamicin Anaerobic cell culture: To monitor changes in HIF expression under hypoxic conditions, cells were cultured under anaerobic conditions at an OZ level of 0,1-1,5%
in an incubation bag (Merck) with Anaerocult (Merck) added to chemically bind OZ.
Anaerobic conditions were obtained within 1-2 hours. Cells were subjected to anoxia for 6 or 18 hours.
Example 4 Treatment with antisense oligonucleotide The cells (described above) were treated with oligonucleotide using the cationic liposome formulation LipofectAMINE 2000 (Gibco) as transfection vehicle.
Cells were seeded in 100 mm x 20 mm cell culture petri dishes (Corning) or 6-well plates (NUNC) and treated when 90% confluent. Oligo concentrations used ranged from 1 nM to 400 nM final concentration. Formulation of oligo-lipid complexes was carried out according to the manufacturers instructions using serum-free MEM
and a final lipid concentration of 5 ~g/ml in 6 ml total volume. Cells were incubated at 37°C for 4 or 24 hours and treatment was stopped by removal of oligo-containing culture medium. Cells were washed and serum-containing MEM was added. After oligo treatment cells were either allowed to recover for 18 hours before they were subjected to anoxia for 6 hours or directly subjected to anoxia for 18 hours.
Example 5 Extraction of Total RNA
Total RNA was isolated either using RNeasy mini kit (e.g. Qiagen cat. no.
74104) or using the Trizol reagent (e.g. Life technologies cat. no. 15596).
For RNA
isolation from cell lines, RNeasy is the preferred method and for tissue samples Trizol is the preferred method. Total RNA was isolated from cell lines using the RNeasy mini kit (Qiagen) according to the protocol provided by the manufacturer.
Tissue samples were homogenised using an Ultra Turrax T8 homogeniser (e.g. IKA
Analysen technik) and total RNA was isolated using the Trizol reagent protocol provided by the manufacturer.
Example 6 First strand cDNA synthesis First strand synthesis was performed using OmniScript Reverse Transcriptase kit (cat# 205113, Qiagen) according to the manufacturers instructions.
For each sample 0.5 ~.g total RNA was adjusted to 12 ~,1 each with RNase free and mixed with 2 ~,1 poly (dT)la-i8 (2.5 ~,g/ml) (Life Technologies, GibcoBRL, Roskilde, DK), 2 ~,1 dNTP mix (5 mM each dNTP), 2 ~,l lOx Buffer RT, 1 ~,1 RNAguardTMRnase INHIBITOR (33.3U/ml), (cat# 27-0816-O1, Amersham Pharmacia Biotech, Horsholm, DK) and 1 ~,1 OmniScript Reverse Transcriptase (4 U/~1) followed by incubation at 37°C for 60 minutes and heat inactivation of the enzyme at 93°C for 5 minutes.

Example 7 Antisense modulation of HIF-la expression analysis Antisense modulation of HIF-la expression can be assayed in a variety of ways known in the art. For example, HIF-la mRNA levels can be quantitated by, e.g., Northern blot analysis, competitive polymerase chain reaction (PCR), or real-time PCR. Real-time quantitative PCR is presently preferred. RNA analysis can be performed on total cellular RNA or mRNA.
Methods of RNA isolation and RNA analysis such as Northern blot analysis is routine in the art and is taught in, for example, Current Protocols in Molecular Biology, John Wiley and Sons.
Real-time quantitative (PCR) can be conveniently accomplished using the commercially available iQ Multi-Color Real Time PCR Detection System, available from BioRAD.
Real-time Quantitative PCR Analysis of Ha-ras mRNA Levels Quantitation of mRNA levels was determined by real-time quantitative PCR using the iQ Multi-Color Real Time PCR Detection System (BioRAD) according to the manufacturers instructions.
Real-time Quantitative PCR is a technique well known in the art and is taught in for example Heid et al. Real time quantitative PCR, Genome Research (1996), 6:
986-994.
Platinum Quantitative PCR SuperMix UDG 2x PCR master mix was obtained from Invitrogen cat# 11730. Primers and TaqMan~ probes were obtained from MWG-Biotech AG, Ebersberg, Germany Probes and primers to human HIF-1 oc were designed to hybridise to a human Ha-ras sequence, using published sequence information (GenBank accession number NM 001530, incorporated herein as SEQ m NO:1).
For human HIF-1 a the PCR primers were:
forward primer: 5' CTCATCCAAGAAGCCCTAACGTGTT 3' (final concentration in the assay; 0.9 qM)(SEQ ID NO: 116) reverse primer: 5' GCTTTCTCTGAGCATTCTGCAAAGC 3' (final concentration in the assay; 0.9 ~,M)(SEQ m NO: 117) and the PCR probe was: 5' FAM-CCTCAGGAACTGTAGTTCTTTGACTCAAAGCGACA -TAMR.A 3' (final concentration in the assay; 0.1 ~,M)(SEQ ID NO: 118) Glyceraldehyde-3-phosphate dehydrogenase (GAPDH) mRNA quantity was used as an endogenous control for normalizing any variance in sample preparation.
The sample content of human GAPDH mRNA was quantified using the human GAPDH ABI Prism Pre-Developed TaqMan Assay Reagent (Applied Biosystems cat.
no. 4310884E) according to the manufacturers instructions.
For quantification of mouse GAPDH mRNA the following primers and probes were designed: Sense primer 5'aaggctgtgggcaaggtcatc 3' (0.3 ~M final concentration), antisense primer 5' gtcagatccacgacggacacatt (0.6 ~.M final concentration), TaqMan probe 5' FAM-gaagctcactggcatggcatggccttccgtgttc-TAMRA
3' (0.2 ~M final concentration).
Real time PCR
The cDNA from the first strand synthesis performed as described in example 8 was diluted 2-20 times, and analyzed by real time quantitative PCR. The primers and probe were mixed with 2 x Platinum Quantitative PCR SuperMix UDG (cat. #
11730, Invitrogen) and added to 3.3 ~1 cDNA to a final volume of 25 p,l. Each sample was analysed in triplicates. Assaying 2 fold dilutions of a cDNA that had been prepared on material purified from a cell line expressing the RNA of interest generated standard curves for the assays. Sterile H20 was used instead of cDNA for the no template control. PCR program: 50° C for 2 minutes, 95° C for 10 minutes followed by 40 cycles of 95° C, 15 seconds, 60° C, 1 minutes.
Relative quantities of target mRNA sequence were determined from the calculated Threshold cycle using the iCycler iQ Real-time Detection System software.
Example 8 Western blot analysis of HIF-la Protein Levels Protein levels of HIF-1 a can be quantitated in a variety of ways well known in the art, such as immunoprecipitation, Western blot analysis (immunoblotting), ELISA, RIA (Radio Immuno Assay) or fluorescence-activated cell sorting (FACS).
Antibodies directed to HIF-1 a can be identified and obtained from a variety of sources, such as Upstate Biotechnologies (Lake Placid, USA), Novus Biologicals (Littleton, Colorado), Santa Cruz Biotechnology (Santa Cruz, California) or can be prepared via conventional antibody generation methods.
To measure the effect of treatment with antisense oligonucleotides against HIF-la, protein levels of HIF1-a in treated and untreated cells were determined using Western blotting.
After treatment with oligonucleotide as described above, cell were harvested by scraping with a rubber policeman in ice-cold phosphate-buffered saline (PBS) containing 0,37 mg/ml of the protease inhibitor phenyl methyl sulfonyl fluoride (PMSF) The .harvested cells were washed in 1 ml. PBS containing PMSF as described above and cell pellets were kept frozen at -80°C.
For protein extraction, frozen cell pellets were dissolved in 3 volumes of ice-cold lysis buffer (50 mM Tris, pH 7,5, 150 mM NaCl, 1% Non-idet P 40 (NP-40), 0,1 % SDS, 1 % (w/v) natrium-deoxycholat, 1 mM dithiothreitol (DTT), Complete protein inhibitor cocktail (Boehringer Mannheim)). The samples were sonicated times a 5-10 seconds in a Vibra Cell 50 sonicator (Sonics & Materials Inc.).
The lysate was stored at -80°C until further use.
Protein concentration of the protein lysate was determined using the BCA
Protein Assay Kit (Pierce) as described by the manufacturer.
SDS gel electrophoresis:
Protein samples prepared as described above were thawed on ice and denatured at 70°C for 10 min.
Samples were loaded on 1,0 mm 10% NuPage Bis-Tris gel (NOVEX) and gels were run in running buffer, either NuPage MES SDS Running Buffer or NuPage MOPS SDS Running Buffer (both NOVEX) depending on desired separation of proteins in an Xcell II Mini-cell electrophoresis module (NOVEX).
In the inner chamber of the electrophoresis module NuPage Antioxidant (NOVEX) was added to the running buffer at a final concentration of 2,5 ~,1/ml. For size reference, SeeBlue Plus2 Prestained Standard (Invitrogen) was loaded on the gel.
The electrophoresis was run at 160 V for 2 hours.

Semi- blotting After electrophoresis, the separated proteins were transferred to a polyvinyliden difluoride (PVDF) membrane by semi-dry blotting. The gel was equilibrated in NuPage Transfer Buffer (NOVEX) until blotted. The blotting procedure was carried out in a Trans-blot SD Semi-Dry transfer cell (BioRAD) according to the manufacturers instructions. The membrane was stored at 4°C until further use.
Immunodetection:
To detect the desired protein, the membrane was incubated with either polyclonal or monoclonal antibodies against the protein. The membrane was blocked in blocking buffer (2,5% skim milk powder and 5 % BSA dissolved in TS-buffer (150 mM NaCl, 10 mM Tris.base pH 7,4)) for 1 hour with agitation. The membrane was then washed 2 x 15 min. in TS-buffer at room temperature and incubated over night with primary antibody in TS-Tween20-buffer with 0,1% NaN3 at 4 C. The following primary monoclonal antibodies and concentrations/dilutions were used: Mouse-anti-Glutl (from T. Ploug, The Panum Institute, Copenhagen) 1:20, mouse-anti-HIF-la (H72320, Transduction Laboratories) 1 ~,g/ml, mouse-anti-a-tubulin (T-9026, Sigma) 1:10.000. After incubation with the primary antibody the membrane was washed in TS-Tween20-buffer for 15 minutes followed by 2 additional washes of 5 minutes each with agitation at room temperature. Subsequently the membrane was incubated with a 1:5000 dilution of the secondary antibody, peroxidase conjugated polyclonal goat-anti-mouse-immunoglobulins (P0447, DAKO A/S) for 1 hour at room temperature.
The membrane was then washed in TS-Tween20-buffer for 15 minutes followed by 3 additional of washes 5 minutes each with agitation at room temperature. After the last wash the membrane was incubated with ECL+ Plus (Amersham), for 5 minutes followed by an immediate scanning with a STORM 840 (Molecular Dynamics Inc.).
The membrane was stripped in stripping-buffer (100mM 2-mercapto-ethanol, 2 SDS, 62,5 mM Tris-base) pH 6,7 by incubation with low agitation for 30 minutes at 50 C. After wash in TS-Tween20-buffer 2 x 10 minutes at room temperature, the membrane was dried and sealed in a plastic bag and stored at 4 C. The Protein expression levels were quantified relative to expression of a housekeeping protein using Image Quant version 5,0 software (Molecular Dynamics Inc).

Example 9 Antisense Inhibition of Human HIF-la Expression by oligonucleotides In accordance with the present invention, a series of oligonucleotides were designed to target different regions of the human HIF-la RNA, using published sequences (GenBank accession number NM 001530, incorporated herein as SEQ m NO: 1 and Figure 7). The oligonucleotides 16 nucleotides in length are shown in Table 1 and are having a SEQ m NO. The oligonucleotides are designed so they are to be particularly potent as antisense oligonucleotides, particularly when synthesised using artificial nucleotides such as LNA or phosphorothioates etc. "Target site"
indicates the first nucleotide number on the particular target sequence to which the oligonucleotide binds. The compounds were analysed for their effects on HIF-la protein levels by Western blot analysis as described in other examples herein.
Table 1 Inhibition of human HIF-la protein levels antisense oligonucleotides SEQ ID.NO. % ?arget'Oligo Sepuence O(igo Design lGureon Inhibition"site ' S'-3' nci: 100 N~ .5'-3' oii o ~

SEQ ID NO 92 234 GCGATGTCTTCACGGC GSCSGSAStSgStScStstScSaSCSGSGSC

SEQ ID NO 94 2256 TGGTGAGGCTGTCCGA TSGSGSTSgSasgsgscstsgstsCsCsGsA
SEQ ID NO 96 153 AGGCAAGTCCAGAGGT ASGSGSCSaSaSgStscScSaSgSASGSGST
SEQ ID NO 93 2680 TTCTGCTGCCTTGTAT TSTSCSTSg5c5tsgScScS~tSGSTSAST

SEQ ID NO 93 2783 TTTAGGTAGTGAGCCA TSTSTSASgSgstSasgstsgSasGsCsCsA

SEQ ID NO. % TargefOligo-Sequence Ol~go Design, /Gureon Inhibifionsite 5'-3' 5'-3' no. 100 Nm oligo SEQ ID NO ~ 2369 TTTAGTTCTTCCTCAG

SEQ ID NO 87 479 TCACCAGCATCCAGAA TSCSASCScSaSgScSaStScScSASGSASA

SEQ ID NO 84 917 ATCAGCACCAAGCAGG ASTSCSASgScSaScScSaSasgsCsAsGSG

SEQ ID NO 95 1177 TGGCAAGCATCCTGTA TSGSGSCSaSaSgScSaStscScSTSGSTsA

SEQ ID NO 93 1505 TCTGTGTCGTTGCTGC TSCsTsGstsgs~scsgsts~sgsCsTsGsC

SEQ ID NO 86 2223 TGGTGATGATGTGGCA TSGSGSTSgSaslsgSastsgstSGsGsCSA

SEQ ID NO 79 2477 TCGTCTGGCTGCTGTA TSCSGSTScStsgSgscstsgscsTsGsTSA

SEQ ID NO 85 2553 TTGCTCCATTCCATTC TSTSGSCStscscSastStScscsAsTsTsC

SEQ ID NO. % TargetOligo Sequence Oligo pesign ' Inhibitionsite 5'-3' ' S'-3' ICureon 100 Nm no. oligo SEQ ID NO 97 3091 GTTACTGCCTTCTTAC GSTSTSAScStSgScScStSt5cST5TSASC

Cur2652 SEQ ID NO 90 293 CCGGCGCCCTCCATGG CSCSGSGScSgScScScStScScSASTSGSG

Cur2653 The sequences that demonstrated at least 20% inhibition of HIF-la expression in this experiment are preferred (see also figure 1-9). The target sites to which these preferred sequences are complementary are herein referred to as "hot spots"
and are therefore preferred sites for targeting by compounds of the present invention.
Example 10 Antisense Inhibition of HIF-la by phosphorothioate, LNA containing oligonucleotides or chimeric oligonucleotides having at least one LNA segments and at least one phosphorothioate segment In accordance with the present invention, a second series of antisense oligonucleotides were also synthesized (table 2). These series of compounds are full-modified phosphorothioate oligonucleotide, full modified LNA oligonucleotide or a chimeric oligonucleotides 16 nucleotides in length targeting two different sites. The chimeric oligonucleotides are a "gapmer" (GM), "headmer" (WMS) or "tailiner"
(WM3) composed of a region consisting of phosphoroptioates (P S) which is flanked on one or both sides) with a LNA segment. These segments are composed of oxy-LNA nucleotides. Some the oligonucleotides also had a fluorescent colour (FAM) incorporated. Mismatch oligonucleotides were also designed (lVlllil). All cytosines in oxy-LNA are methylated at the CS position of the nucleobases. The compounds were analysed for their effect on HIF-la protein levels by western blotting as described in other examples herein. "Target site" indicates the first nucleotide number on the particular target sequence to which the oligonucleotide binds.
Table 2 Inhibition of human HIF-la protein levels by phosphorothioate oligonucleotides, LNA containing oligonucleotides or chimeric oligonucleotides having one or two LNA segments) and one phosphorothioate segment (backbone linkage is P=O unless other indicated. s; P=S linkage, small letters;
deoxynucleic acid, capital letters; oxy-LNA; ).
Name SEQ NO % inhibitionTarget Sequence & Design 100 nM oligoSite & 5' - 3' Desi n Cur0805SEQ ID 24 234 FM GCGATGTCTTCACGGC

Cur0806SEQ ID 21 234 PS gScSgSaStsgstscstStscsascsgsgsc Cur0807SEQ ID 234 GM GCGAStSgStScStStScSaSCGGC

Cur0808SEQ ID 234 FAM FAM-GCGAStSgStScStStScSaSCGGC

Cur0809SEQ ID 34 234 WM5 GCGATGTCststscsascsgsgsc Cur0810SEQ ID 54 234 WM3 gScSgSaStSgstScSTTCACGGC

Cur0811SEQ ID 24 2256 FM TGGTGAGGCTGTCCGA

Cur0812SEQ ID 2256 PS tsgsgstsg5asgsgscstsgstscscsgsa Cur0813SEQ ID 86 2256 GM TGGTSgSaSgs95cstSgStsCCGA

Cur0814SEQ ID 2256 FAM FAM-TGGTSgSaSgsgScstSgStsCCGA

Cur0815SEQ ID 2256 WM5 TGGTGAGGscstsgstscscsgsa Cur0816SEQ ID 37 2256 WM3 ts9SgSt5gsaSgSgsCTGTCCGA

Cur0959SEQ ID 234 MM1 GCGAtScStScStStScSaSGGGC

Cur0960SEQ ID ~ 234 MM2 GCGTtSgStScSaStScSaSCGGC

Cur0961SEQ ID 2256 MM1 TGGTgSaSgScScStS95tSCGGA

Cur0962SEQ ID 2256 MM2 TGCTgSaSgsgSgStSgstSCCGA

Cur2627SEQ ID 94 2256 GM TSGSGSTSgSaSgSgscstsgstSCsCSGSA

Cur2628SEQ ID 95 2256 GM TSGSGStSgSaSgSgScstSgstSCsCsGsA

Cur2629SEQ ID 96 2256 GM TSGSGStSgSaSgsgScStSgstsCsCsGsa Cur2630SEQ ID 95 2256 GM TSGSGSTSgSaSgSgscStSgstSCSCSGsa Cur2631SEQ ID 95 2256 GM TSGSGSTSGSAsgsgscstsgstscscsgsa Cur2632SEQ ID 94 2256 GM TGGtSgSasgsgScstSgstsCCGA

Cur2633SEQ ID 94 2256 GM TGGtSgSaSgsgScstSgstsCCGa Cur2634SEQ ID 91 2256 GM TGGTgSaSgsgScStsgStSCCGa Cur2635SEQ ID 94 2256 WM5 TGGTGAgsgscstsgstscscsgsa Cur2412SEQ ID 2256 GM TGGTSgSasgsgsmcstsgstsCCGA

As. shown in Table 2 and Figures 1-6, most of SEQ ID NOs 89-104 demonstrated at least 20% inhibition of HIF-la expression in this experiment and are therefore preferred. The target sites to which these preferred sequences are complementary are herein referred to as "hot spots" and are therefore preferred sites for targeting by compounds of the present invention.
Example 11 IfZ vivo efficacy oligomeric compounds targeting HIF-la The effect of oligonucleotide treatment on growth of tumour xenografts on nude mice can be measured using different tumour cell lines. Examples of such cell lines are human tumour cell lines U87 (glioblastoma), U373 (glioblastoma), (prostate cancer) and CPH 54A (small cell lung carcinoma) and marine tumour cell line B 16 (melanoma).
Treatment of subcutaneous tumour xeno~rafts on nude mice using LNA-containing oligos.
Tumour cells were implanted subcutaneously and then serially passaged by three consecutive transplantations. Tumour fragments of 1 mm were implanted subcutaneously with a trocar needle in NMRI nude mice. Alternatively, cancer cells typically 106 cells suspended in 300 ~l matrigel (BD Bioscience), were subcutaneously injected into the flanks of NMRI nude mice.
Mice were treated by infra-peritoneal or subcutaneous injection of oligonucleotide at various doses, maximum dose 5 mg/kg/day or by administration of up to 5 mg/kg/day for up to 28 days using ALZET osmotic pumps implanted subcutaneously. Individual treatment of the mice started when tumour volume reached 50 mm3. Treatment with PBS was initiated when mean tumour volume of control group reached 50 mm3. The experiment was terminated when tumours of any group reached maximum allowed sizes. The tumour sizes of all mice were measured daily by caliper measurements. The effect of treatment was measured as tumour size and tumour growth rate. Oligonucleotide treated mice were sacrificed 24 hours after the last oligonucleotide injection.

At the end of treatment period mice were anaesthetised and the tumours were excised and immediately frozen in liquid nitrogen for target analysis.
Results: Mice bearing U373 xenograft tumours were treated with Cur813 (SEQ m NO 97) 5 mg/kg/day, i.p. x 1 daily for 7 days or PBS 100~,1/10g/day, i.p. x 1 daily for 7 days. Five mice were treated in each group. Tumour evaluation was carried out as outlined above. Tumour growth curves are shown in fig. 9.
Comparison of tumour size (t-test) Day P-value 4 0.0477 5 0.0156 6 0.0354 7 0.0461 Kaplan Meier analysis:
Terminal event: Tumour size 150 mm3:
Group No. Censored Events Median survival, days Cur813 5 1 4 7 Logrank test for equality of survival distributions: P = 0.0138 Treatment of intracranial tumor xeno~rafts on nude mice using LNA-containing oligos.
Tumour cells axe implanted intracranially on NMRI nude mice and oligonucleotide treatment is initiated 1 week after implantation. Mice are treated by infra-peritonal or subcutaneous injection of oligonucleotide at various doses, maximum dose 2 mg/kg or PBS. The number of treatments will depend on the tumour growth rate in the control group. The experiment will be terminated when tumours of any group reach maximum allowed sizes or until death ensues in any group. The effect of treatment will be measured as time until chronic neurological impairment.
Oligonucleotide treated mice will be killed 24 hours after the last oligonucleotide inj ection.

Example 12 Ih vivo analysis: Inhibition of HIF-la protein level in human tumour cells grown ih vivo systemic treatment with antisense oligonucleotides The tumours were homogenised in lysis buffer (20 mM Tris-Cl [pH 7.5]; 2%
Triton X-100; 11100 vol. Protease Inhibitor Cocktail Set III (Calbiochem);
1/100 vol.
Protease Inhibitor Cocktail Set II (Calbiochem)) at 4°C with the use of a motor-driven tissue homogeniser. 500 ~1 lysis buffer is applied per 100 mg tumour tissue.
Tumour lysates were centrifuged at 13.000 g for 5 min at 4°C to remove tissue debris. Protein concentrations of the tumour extracts were determined using the BCA Protein Assay Reagent Kit (Pierce, Rockford). Western blot analysis of target protein expression was carried out as described in example 8.
The present invention has been described with specificity in accordance with certain of its preferred embodiments. Therefore, the following examples serve only to illustrate the invention and are not intended to limit the same.

Claims (36)

1. ~A compound of 8 to 50 nucleobases in length which modulates HIF-1.alpha.
and wherein said compound has a sequence comprising at least an 8-nucleobase portion of SEQ ID NO: 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, 100, 101, 102, 103, 104, 105, 106, 107, 108, 109, 110, 111, 112, 113, 114 or 115.
2. ~A compound of 8 to 50 nucleobases in length targeted to a nucleic acid molecule encoding HIF-1.alpha., wherein said compound specifically hybridises with a nucleic acid encoding HIF-1.alpha. and inhibits the expression of HIF-1.alpha.
and wherein said compound has a sequence comprising at least an 8 nucleobase portion of SEQ ID
NO:
2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, 100, 101, 102, 103, 104, 105, 106, 107, 108, 109, 110, 111, 112, 113, 114 or 115.
3. ~The compound according to any one of claims 1-2, which is an antisense oligonucleotide.
4. ~The compound according to any one of claims 1-3, wherein at least one nucleotide comprises at least one Nucleic Acid Analogue.
5. ~The compound according to any one of claims 1-4, wherein at least one nucleotide comprises at least one Locked Nucleic Acid (LNA) unit.
6. ~The compound according to claims 1-5, wherein the LNA unit has the structure of the general formula in Scheme 2.
7. ~The compound according to claims 1 through 6, wherein at least one nucleotide comprises a LNA unit according any of the formulas in Scheme 3:
Where X is independently selected from -O-, -S-, -NH-, and N(R H), Z and Z* are independently selected from the group consisting of an internucleoside linkage, a terminal group or a protecting group, B constitutes a natural or non-natural nucleobase.
8. ~The compound according to claims 4 through 7, wherein the internucleoside linkage may be selected from the group consisting of -O-P(O)2-O-, -O-P(O,S)-O-, -O-P(S)2-O-, -S-P(O)2-O-, -S-P(O,S)-O-, -S-P(S)2-O-, -O-P(O)2-S-, -O-P(O,S)-S-, -S-P(O)2-S-, -O-PO(R H)-O-, O-PO(OCH3)-O-, -O-PO(NR H)-O-, -O-PO(OCH2CH2S-R)-O-, -O-PO(BH3)-O-, -O-PO(NHR H)-O-, -O-P(O)2-NR H-, -NR H-P(O)2-O-, -NR H-CO-O-, where R H is selected form hydrogen and C1-4-alkyl.
9. ~The compound of claim 8, wherein the modified internucleoside linkage is a phosphorothioate linkage.
10. ~The compound according to claims 4 through 9, wherein the nucleobases is a modified nucleobases selected from the group consisting of 5-methylcytosine, isocytosine, pseudoisocytosine, 5-bromouracil, 5-propynyluracil, 6-aminopurine, 2-aminopurine, inosine, diaminopurine, 2-chloro-6-aminopurine.
11. The compound according to any one of claims 4-10, wherein the LNA unit is oxy-LNA, thio-LNA, amino-LNA in either the D-.beta. or L-.alpha.
configurations or combinations thereof.
12. The compound of any one of claims 1-11, wherein the antisense oligonucleotide is a chimeric oligonucleotide.
13. The compound of claim 12, wherein the antisense oligonucleotide is a gapmer, headmer or tailmer.
14. The compound according to any one of claims 1-13, wherein the antisense oligonucleotide is a 13, 14, 15, 16, 17, 18, 19, 20 or 21-mer.
15. The compound according to any one of claims 1-14, wherein the number of LNA units are 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20 or 21.
16. The compound according to any one of claims 1-15, wherein at least four nucleotides are LNAs.
17. A compound of 8 to 50 nucleobases in length targeted to a nucleic acid molecule encoding H1F-1.alpha., wherein said compound specifically hybridises with a nucleic acid encoding HIF-1.alpha. and inhibits the expression of HIF-1.alpha.
and wherein said compound has a sequence comprising at least an 8-nucleobase portion of SEQ ID
NO:

2, 3, 12, 22, 23, 53, 54, 55, 56, 59, 60, 61, 89, 90, 93, 94, 95, 97, 100, 105, 106, 107, 108, 109, 110, 111, 112, 114 or 115.
18. A composition comprising the compound of claims 1 through 17 and a pharmaceutically acceptable carrier or diluent.
19. A pharmaceutical composition comprising the compound of any one of claims 1-18, which further comprises a pharmaceutically acceptable carrier.
20. A pharmaceutical composition comprising the compound of any one of claims 1-18, which is employed in a pharmaceutically acceptable salt.
21. A pharmaceutical composition comprising the compound of any one of claims 1-18, which further comprises a conjugate or formulation.
22. A pharmaceutical composition comprising the compound of any one of claims 1-18, which constitutes a pro-drug.
23. A pharmaceutical composition comprising the compound of any one of claims 1-18, which further comprises other oligomeric compounds, chemotherapeutic compounds, antiinflamatory compounds and/or antiviral compounds.
24. A method of inhibiting the expression of HIF-1.alpha., in cells or tissues comprising contacting said cells or tissues with the compound according to any one of claims 1-23 so that expression of HIF-1.alpha. is inhibited.
25. A method of modulating expression of a gene involved in a disease comprising contacting the gene or RNA from the gene with an oligomeric compound wherein said compound has a sequence comprising at least an 8 nucleobase portion of SEQ ID NO: 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, 100, 101, 102, 103, 104, 105, 106, 107, 108, 109, 110, 111, 112, 113, 114 or 115 whereby gene expression is modulated.
26. A method according to claim 25, wherein the compounds comprise one or more LNA units.
27. The method of claim 25 or 26, wherein the compound hybridizes with messenger RNA of the gene to inhibit expression thereof.
28. A method of treating a mammal suffering from or susceptible from an cancer disease, comprising:
administering to the mammal an therapeutically effective amount of an oligonucleotide targeted to HIF-1.alpha. that comprises one or more LNA units.
29. The method according to any one of claims 25-28 wherein the disease is a common cancer, as e.g. primary and metastatic breast, colorectal, prostate, pancreas, other GI-cancers, lung, cervical, ovarian, brain, head and neck, cervix, colon, liver, thyroid, kidney, testes, stomach, intestine, bowel, esophagus, spinal cord, sinuses, bladder or urinary tract tumors, as well as pre-eclampsia, inflammatory bowel disease and Alzheimers disease.
30. A method of modulating angiogenesis as well as red blood cell proliferation, cellular proliferation, iron metabolism, glucose and energy metabolism, pH
regulation, tissue invasion, apoptosis, multi-drug resistance, cellular stress response or matrix metabolism comprising contacting a cell with the antisense compound of claim 1-18 so that the cell is modulated.
31. A method of inhibiting the proliferation of cells comprising contacting cells in vitro with an effective amount of the antisense compound of claim 1-18, so that proliferation of the cells is inhibited.
32. The method of claim 31 wherein said cells are cancer cells.
33. A method of modulating apoptosis in a cell comprising contacting a cell with the antisense compound of claim 1-18 so that apoptosis is modulated.
34. The method of claim 33 wherein said modulation of apoptosis is sensitization to an apoptotic stimulus.
35. The method of claim 33 or 34 wherein said apoptotic stimulus is a cytotoxic chemotherapeutic agent.
36. The method of claim 35 further comprising contacting said cells with a chemotherapeutic agent.
CA2480311A 2002-04-05 2003-04-04 Oligomeric compounds for the modulation of hif-1alpha expression Expired - Fee Related CA2480311C (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US37012602P 2002-04-05 2002-04-05
US60/370,126 2002-04-05
PCT/IB2003/001758 WO2003085110A2 (en) 2002-04-05 2003-04-04 Oligomeric compounds for the modulation hif-1alpha expression

Publications (2)

Publication Number Publication Date
CA2480311A1 true CA2480311A1 (en) 2003-10-16
CA2480311C CA2480311C (en) 2015-01-27

Family

ID=28792033

Family Applications (1)

Application Number Title Priority Date Filing Date
CA2480311A Expired - Fee Related CA2480311C (en) 2002-04-05 2003-04-04 Oligomeric compounds for the modulation of hif-1alpha expression

Country Status (12)

Country Link
US (5) US7737264B2 (en)
EP (2) EP2264172B1 (en)
JP (2) JP4338527B2 (en)
AU (1) AU2003225495B2 (en)
CA (1) CA2480311C (en)
CY (1) CY1119614T1 (en)
DK (1) DK2264172T3 (en)
ES (1) ES2649817T3 (en)
HU (1) HUE037352T2 (en)
PT (1) PT2264172T (en)
SI (1) SI2264172T1 (en)
WO (1) WO2003085110A2 (en)

Families Citing this family (119)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
SI2264172T1 (en) 2002-04-05 2017-12-29 Roche Innovation Center Copenhagen A/S Oligomeric compounds for the modulation of hif-1alpha expression
WO2003087368A2 (en) * 2002-04-18 2003-10-23 Lynkeus Bio Tech Gmbh Means and methods for the specific modulation of target genes in the cns and the eye and methods for their identification
US7148342B2 (en) 2002-07-24 2006-12-12 The Trustees Of The University Of Pennyslvania Compositions and methods for sirna inhibition of angiogenesis
WO2004042024A2 (en) 2002-11-01 2004-05-21 The Trustees Of The University Of Pennsylvania COMPOSITIONS AND METHODS FOR siRNA INHIBITION OF HIF-1 ALPHA
AU2003281969B2 (en) 2002-11-18 2011-01-27 Roche Innovation Center Copenhagen A/S Amino-LNA, thio-LNA and alpha-L-oxy-LN
US7144999B2 (en) 2002-11-23 2006-12-05 Isis Pharmaceuticals, Inc. Modulation of hypoxia-inducible factor 1 alpha expression
WO2004053104A2 (en) 2002-12-11 2004-06-24 Coley Pharmaceutical Group, Inc. 5’ cpg nucleic acids and methods of use
JP4546454B2 (en) * 2003-01-28 2010-09-15 レクサーン・コーポレイション Antisense oligonucleotide that suppresses HIF-1 expression
CA2513398C (en) * 2003-01-31 2013-09-10 Rexahn Corporation Antisense oligonucleotides that inhibit expression of hif-1
US20040241717A1 (en) * 2003-02-10 2004-12-02 Santaris Pharma A/S Oligomeric compounds for the modulation of thioredoxin expression
WO2005013901A2 (en) 2003-07-31 2005-02-17 Isis Pharmaceuticals, Inc. Oligomeric compounds and compositions for use in modulation of small non-coding rnas
US20050053981A1 (en) * 2003-09-09 2005-03-10 Swayze Eric E. Gapped oligomeric compounds having linked bicyclic sugar moieties at the termini
US7480382B2 (en) * 2003-09-30 2009-01-20 Microsoft Corporation Image file container
US8188254B2 (en) 2003-10-30 2012-05-29 Coley Pharmaceutical Gmbh C-class oligonucleotide analogs with enhanced immunostimulatory potency
CA2551100A1 (en) * 2003-12-23 2005-07-14 The Trustees Of The University Of Pennsylvania Compositions and methods for combined therapy of disease
US7563885B1 (en) * 2004-05-24 2009-07-21 Isis Pharmaceuticals, Inc. Modulation of Tudor-SN expression
US7618947B2 (en) * 2004-08-25 2009-11-17 Isis Pharmaceuticals, Inc. Modulation of HIF-1 beta expression
ES2543207T3 (en) 2004-09-01 2015-08-17 Dynavax Technologies Corporation Methods and compositions for the inhibition of innate immune responses and autoimmunity
JP2008513513A (en) * 2004-09-21 2008-05-01 アネシヴァ, インコーポレイテッド Delivery of polynucleotides
ZA200704253B (en) * 2004-11-09 2008-09-25 Santaris Pharma As Potent LNA oligonucleotides for the inhibition of HIF-1A expression
US9447138B2 (en) 2004-11-09 2016-09-20 Roche Innovation Center Copenhagen A/S Potent LNA oligonucleotides for the inhibition of HIF-1a expression
EA014097B1 (en) 2004-11-09 2010-08-30 Сантарис Фарма А/С Potent lna oligonucleotides for modulating of hif-1a expression and use thereof
JP2008543328A (en) * 2005-06-20 2008-12-04 アヴァリス・アクチエボラーグ Constructs that stabilize hybridization
WO2007031081A2 (en) * 2005-09-15 2007-03-22 Santaris Pharma A/S RNA ANTAGONIST COMPOUNDS FOR THE INHIBITION OF APO-Bl00 EXPRESSION
KR20080065617A (en) * 2005-09-19 2008-07-14 존슨 앤드 존슨 파머슈티컬 리서치 앤드 디벨로프먼트 엘엘씨 Modulation of glucocorticoid receptor expression
JP2007119396A (en) * 2005-10-28 2007-05-17 Hosokawa Funtai Gijutsu Kenkyusho:Kk Pharmaceutical formulation for transpulmonary administration containing nanoparticle having sealed nucleic acid compound
WO2007073149A1 (en) * 2005-12-22 2007-06-28 Keygene N.V. Alternative nucleotides for improved targeted nucleotide exchange
WO2007087113A2 (en) 2005-12-28 2007-08-02 The Scripps Research Institute Natural antisense and non-coding rna transcripts as drug targets
TW200731980A (en) 2005-12-29 2007-09-01 Alcon Mfg Ltd RNAi-mediated inhibition of HIF1A for treatment of ocular angiogenesis
AU2007258117B2 (en) * 2006-05-05 2013-05-30 Isis Pharmaceuticals, Inc. Compounds and methods for modulating gene expression
WO2007133758A1 (en) * 2006-05-15 2007-11-22 Physical Pharmaceutica, Llc Composition and improved method for preparation of small particles
DK2029746T3 (en) * 2006-06-12 2012-10-08 Exegenics Inc D B A Opko Health Inc Compositions and Methods for siRNA Inhibition of Angiogenesis
US7872118B2 (en) * 2006-09-08 2011-01-18 Opko Ophthalmics, Llc siRNA and methods of manufacture
EP2076597A2 (en) * 2006-10-09 2009-07-08 Santaris Pharma A/S Rna antagonist compounds for the modulation of pcsk9
EP2455471A3 (en) * 2006-11-27 2012-09-12 Isis Pharmaceuticals, Inc. Methods for treating hypercholesterolemia
US8093222B2 (en) 2006-11-27 2012-01-10 Isis Pharmaceuticals, Inc. Methods for treating hypercholesterolemia
WO2008070616A2 (en) * 2006-12-01 2008-06-12 University Of Utah Research Foundation METHODS AND COMPOSITIONS RELATED TO HIF-1α
DK2149605T3 (en) * 2007-03-22 2013-09-30 Santaris Pharma As Short RNA antagonist compounds to modulate the desired mRNA
EP2149605B1 (en) * 2007-03-22 2013-07-03 Santaris Pharma A/S Short RNA antagonist compounds for the modulation of target mRNA
WO2009039173A2 (en) 2007-09-19 2009-03-26 Applied Biosystems Inc. SiRNA SEQUENCE-INDEPENDENT MODIFICATION FORMATS FOR REDUCING OFF-TARGET PHENOTYPIC EFFECTS IN RNAi, AND STABILIZED FORMS THEREOF
JP2010539961A (en) * 2007-10-04 2010-12-24 サンタリス ファーマ アー/エス Short RNA antagonist compounds for modulation of HIF1α
EP2209896B1 (en) 2007-10-26 2017-03-01 Dynavax Technologies Corporation Methods and compositions for inhibition of immune responses and autoimmunity
CA2705714A1 (en) * 2007-11-26 2009-06-04 Santaris Pharma A/S Lna antagonists targeting the androgen receptor
ES2727549T3 (en) * 2008-10-03 2019-10-17 Curna Inc Treatment of diseases related to apolipoprotein a1 by inhibition of the natural antisense transcript to apolipoprotein a1
US20110230420A1 (en) * 2008-11-17 2011-09-22 Enzon Pharmaceuticals, Inc. Releasable conjugates for nucleic acids delivery systems
MX2011005910A (en) * 2008-12-04 2011-06-17 Opko Curna Llc Treatment of erythropoietin (epo) related diseases by inhibition of natural antisense transcript to epo.
CA2745832A1 (en) 2008-12-04 2010-06-10 Opko Ophthalmics, Llc Compositions and methods for selective inhibition of pro-angiogenic vegf isoforms
US8927511B2 (en) * 2008-12-04 2015-01-06 Curna, Inc. Treatment of vascular endothelial growth factor (VEGF) related diseases by inhibition of natural antisense transcript to VEGF
KR101840618B1 (en) 2008-12-04 2018-03-20 큐알엔에이, 인크. Treatment of tumor suppressor gene related diseases by inhibition of natural antisense transcript to the gene
CN102387817B (en) 2009-02-12 2018-01-30 库尔纳公司 By suppressing to treat the related diseases of BDNF for the natural antisense transcript of neurotrophic factor derived from brain (BDNF)
EP2408919B1 (en) 2009-03-16 2017-10-18 CuRNA, Inc. Treatment of nuclear factor (erythroid-derived 2)-like 2 (nrf2) related diseases by inhibition of natural antisense transcript to nrf2
CA2755404C (en) 2009-03-17 2020-03-24 Joseph Collard Treatment of delta-like 1 homolog (dlk1) related diseases by inhibition of natural antisense transcript to dlk1
EP2427553A4 (en) 2009-05-06 2012-11-07 Opko Curna Llc Treatment of lipid transport and metabolism gene related diseases by inhibition of natural antisense transcript to a lipid transport and metabolism gene
CN102803492B (en) 2009-05-06 2016-06-29 库尔纳公司 TTP relevant disease is treated for the natural antisense transcript of triple four proline (TTP) by suppression
KR101742334B1 (en) 2009-05-08 2017-06-01 큐알엔에이, 인크. Treatment of dystrophin family related diseases by inhibition of natural antisense transcript to dmd family
DK2432881T3 (en) 2009-05-18 2018-02-26 Curna Inc TREATMENT OF REPROGRAMMING FACTOR-RELATED DISEASES BY INHIBITING NATURAL ANTISENSE TRANSCRIPTS TO A REPROGRAMMING FACTOR
US8895527B2 (en) 2009-05-22 2014-11-25 Curna, Inc. Treatment of transcription factor E3 (TFE3) and insulin receptor substrate 2(IRS2) related diseases by inhibition of natural antisense transcript to TFE3
WO2010138806A2 (en) 2009-05-28 2010-12-02 Curna, Inc. Treatment of antiviral gene related diseases by inhibition of natural antisense transcript to an antiviral gene
KR101801404B1 (en) 2009-06-16 2017-12-20 큐알엔에이, 인크. Treatment of collagen gene related diseases by inhibition of natural antisense transcript to a collagen gene
US8951981B2 (en) 2009-06-16 2015-02-10 Curna, Inc. Treatment of paraoxonase 1 (PON1) related diseases by inhibition of natural antisense transcript to PON1
KR101807323B1 (en) 2009-06-24 2017-12-08 큐알엔에이, 인크. Ttreatment of tumor necrosis factor receptor 2 (tnfr2) related diseases by inhibition of natural antisense transcript to tnfr2
CA2765815A1 (en) 2009-06-26 2010-12-29 Opko Curna, Llc Treatment of down syndrome gene related diseases by inhibition of natural antisense transcript to a down syndrome gene
US8563528B2 (en) 2009-07-21 2013-10-22 Santaris Pharma A/S Antisense oligomers targeting PCSK9
US20120252869A1 (en) 2009-07-24 2012-10-04 Opko Curna, Llc Treatment of sirtuin (sirt) related diseases by inhibition of natural antisense transcript to a sirtuin (sirt)
ES2585360T3 (en) 2009-08-05 2016-10-05 Curna, Inc. Treatment of diseases related to an insulin gene (INS) by inhibition of natural antisense transcription in an insulin gene (INS)
CN104313027B (en) 2009-08-11 2018-11-20 库尔纳公司 By inhibiting the natural antisense transcript of adiponectin (ADIPOQ) to treat adiponectin (ADIPOQ) related disease
KR101805213B1 (en) 2009-08-21 2017-12-06 큐알엔에이, 인크. Treatment of 'c terminus of hsp70-interacting protein' (chip) related diseases by inhibition of natural antisense transcript to chip
JP5964232B2 (en) 2009-08-25 2016-08-03 カッパーアールエヌエー,インコーポレイテッド Treatment of IQGAP-related diseases by inhibition of natural antisense transcripts against 'IQ motif-containing GTPase-activating protein' (IQGAP)
CA2775111C (en) 2009-09-25 2019-12-31 Opko Curna, Llc Treatment of filaggrin (flg) related diseases by modulation of flg expression and activity
CA2782366A1 (en) 2009-12-16 2011-07-14 Opko Curna, Llc Treatment of membrane bound transcription factor peptidase, site 1 (mbtps1) related diseases by inhibition of natural antisense transcript to mbtps1
EP2515947B1 (en) 2009-12-23 2021-10-06 CuRNA, Inc. Treatment of uncoupling protein 2 (ucp2) related diseases by inhibition of natural antisense transcript to ucp2
CA2782373C (en) 2009-12-23 2019-03-26 Opko Curna, Llc Treatment of hepatocyte growth factor (hgf) related diseases by inhibition of natural antisense transcript to hgf
EP2519633B1 (en) 2009-12-29 2017-10-25 CuRNA, Inc. Treatment of nuclear respiratory factor 1 (nrf1) related diseases by inhibition of natural antisense transcript to nrf1
EP2519634B1 (en) 2009-12-29 2016-06-01 CuRNA, Inc. TREATMENT OF TUMOR PROTEIN 63 (p63) RELATED DISEASES BY INHIBITION OF NATURAL ANTISENSE TRANSCRIPT TO p63
DK2519632T3 (en) 2009-12-31 2018-07-23 Curna Inc TREATMENT OF INSULIN RECEPTOR SUBSTRATE 2- (IRS2) RELATED DISEASES BY INHIBITION OF NATURAL ANTISENSE TRANSCRIPTION TO IRS2 AND TRANSCRIPTION FACTOR E3 (TFE3)
JP5886757B2 (en) 2010-01-04 2016-03-16 カッパーアールエヌエー,インコーポレイテッド Treatment of interferon regulatory factor 8 (IRF8) related diseases by inhibition of natural antisense transcripts against interferon regulatory factor 8 (IRF8)
US8912157B2 (en) 2010-01-06 2014-12-16 Curna, Inc. Treatment of pancreatic developmental gene related diseases by inhibition of natural antisense transcript to a pancreatic developmental gene
US9200277B2 (en) 2010-01-11 2015-12-01 Curna, Inc. Treatment of sex hormone binding globulin (SHBG) related diseases by inhibition of natural antisense transcript to SHBG
DK2529015T3 (en) 2010-01-25 2018-02-26 Curna Inc TREATMENT OF RNASE H1-RELATED DISEASES BY INHIBITION OF NATURAL ANTISENSE TRANSCRIPT TO RNASE H1
JP5976548B2 (en) 2010-02-22 2016-08-23 カッパーアールエヌエー,インコーポレイテッド Treatment of pyrroline-5-carboxylate reductase 1 (PYCR1) related diseases by inhibition of natural antisense transcripts against PYCR1
CA2795145C (en) 2010-04-02 2019-01-22 Curna, Inc. Treatment of colony-stimulating factor 3 (csf3) related diseases by inhibition of natural antisense transcript to csf3
KR101900962B1 (en) 2010-04-09 2018-09-20 큐알엔에이, 인크. Treatment of fibroblast growth factor 21 (fgf21) related diseases by inhibition of natural antisense transcript to fgf21
KR101936011B1 (en) 2010-05-03 2019-01-07 큐알엔에이, 인크. Treatment of sirtuin (sirt) related diseases by inhibition of natural antisense transcript to a sirtuin (sirt)
TWI531370B (en) 2010-05-14 2016-05-01 可娜公司 Treatment of par4 related diseases by inhibition of natural antisense transcript to par4
KR101902197B1 (en) 2010-05-26 2018-10-01 큐알엔에이, 인크. Treatment of methionine sulfoxide reductase a (msra) related diseases by inhibition of natural antisense transcript to msra
WO2011150005A2 (en) 2010-05-26 2011-12-01 Opko Curna Llc Treatment of atonal homolog 1 (atoh1) related diseases by inhibition of natural antisense transcript to atoh1
KR101810593B1 (en) 2010-06-16 2017-12-22 다이나박스 테크놀로지 코퍼레이션 Methods of treatment using tlr7 and/or tlr9 inhibitors
WO2011163499A2 (en) 2010-06-23 2011-12-29 Opko Curna, Llc Treatment of sodium channel, voltage-gated, alpha subunit (scna) related diseases by inhibition of natural antisense transcript to scna
JP5998131B2 (en) 2010-07-14 2016-09-28 カッパーアールエヌエー,インコーポレイテッド DISCSLARGEHOMOLOG (DLG) Treatment of DLG-related diseases by inhibition of natural antisense transcripts on DLG1
KR101886457B1 (en) 2010-10-06 2018-08-07 큐알엔에이, 인크. Treatment of sialidase 4 (neu4) related diseases by inhibition of natural antisense transcript to neu4
JP6049623B2 (en) 2010-10-22 2016-12-21 カッパーアールエヌエー,インコーポレイテッド Treatment of IDUA-related diseases by inhibition of natural antisense transcripts to α-L-iduronidase (IDUA)
US10000752B2 (en) 2010-11-18 2018-06-19 Curna, Inc. Antagonat compositions and methods of use
CA2818824A1 (en) 2010-11-23 2012-05-31 Joseph Collard Treatment of nanog related diseases by inhibition of natural antisense transcript to nanog
WO2012082765A2 (en) 2010-12-16 2012-06-21 The United State Of America. As Represented By The Secretary Department Of Health And Human Services Methods for decreasing body weight and treating diabetes
RU2620980C2 (en) 2011-06-09 2017-05-30 Курна, Инк. Treatment of diseases associated with frataxin (fxn), by inhibiting natural antisense fxn transcript
DK2742136T3 (en) 2011-08-11 2017-11-20 Ionis Pharmaceuticals Inc GAPMER COMPOUNDS INCLUDING 5 'MODIFIED DEOXYRIBONUCLEOSIDES IN GAP AND APPLICATIONS THEREOF
EA029151B1 (en) 2011-09-06 2018-02-28 Курна, Инк. TREATMENT OF DISEASES RELATED TO ALPHA SUBUNITS OF VOLTAGE-GATED SODIUM CHANNELS (SCNxA) WITH SMALL MOLECULES
US9580708B2 (en) 2011-09-14 2017-02-28 Rana Therapeutics, Inc. Multimeric oligonucleotides compounds
US8901098B2 (en) 2011-10-25 2014-12-02 Isis Pharmaceuticals, Inc. Antisense modulation of GCCR expression
WO2013138374A2 (en) 2012-03-15 2013-09-19 Curna, Inc. Treatment of brain derived neurotrophic factor (bdnf) related diseases by inhibition of natural antisense transcript to bdnf
US20150133529A1 (en) * 2012-05-16 2015-05-14 Rana Therapeutics, Inc. Compositions and methods for modulating bdnf expression
CA2873766A1 (en) 2012-05-16 2013-11-21 Rana Therapeutics Inc. Compositions and methods for modulating atp2a2 expression
US10837014B2 (en) 2012-05-16 2020-11-17 Translate Bio Ma, Inc. Compositions and methods for modulating SMN gene family expression
JP2015518710A (en) 2012-05-16 2015-07-06 ラナ セラピューティクス インコーポレイテッド Compositions and methods for regulating hemoglobin gene family expression
EA201492123A1 (en) 2012-05-16 2015-10-30 Рана Терапьютикс, Инк. COMPOSITIONS AND METHODS FOR MODULATING THE EXPRESSION OF THE SMN GENES FAMILY
WO2013173608A1 (en) 2012-05-16 2013-11-21 Rana Therapeutics, Inc. Compositions and methods for modulating mecp2 expression
KR20160074368A (en) 2012-05-16 2016-06-28 라나 테라퓨틱스, 인크. Compositions and methods for modulating utrn expression
AU2013315225B2 (en) 2012-09-14 2018-11-08 Translate Bio Ma, Inc. Multimeric oligonucleotide compounds
CN105378083B (en) 2013-03-27 2018-09-21 伊萨纳治疗有限公司 The TGF-β oligonucleotides of modification
WO2014201306A1 (en) * 2013-06-12 2014-12-18 Oncoimmunin, Inc. Systemic in vivo delivery of oligonucleotides
MY183049A (en) 2013-06-27 2021-02-09 Roche Innovation Ct Copenhagen As Antisense oligomers and conjugates targeting pcsk9
CA2921148C (en) * 2013-08-12 2020-10-20 Norges Veterinaerhogskole Detection of organophosphate resistance in crustaceans
WO2015023975A1 (en) * 2013-08-16 2015-02-19 Rana Therapeutics, Inc. Compositions and methods for modulating rna
JP6429264B2 (en) * 2013-11-12 2018-11-28 学校法人東京理科大学 Boranophosphate compounds and nucleic acid oligomers
JP6689279B2 (en) * 2014-12-16 2020-05-20 ロシュ イノベーション センター コペンハーゲン エーエス Chiral toxicity screening method
US10758558B2 (en) 2015-02-13 2020-09-01 Translate Bio Ma, Inc. Hybrid oligonucleotides and uses thereof
MA43072A (en) 2015-07-22 2018-05-30 Wave Life Sciences Ltd COMPOSITIONS OF OLIGONUCLEOTIDES AND RELATED PROCESSES
CN110248956B (en) * 2016-10-11 2023-02-21 奥利通公司 HIF-1 alpha antisense oligonucleotides
WO2018160772A1 (en) 2017-02-28 2018-09-07 The United State Of America, As Represented By The Secretary, Department Of Health & Human Services Method of treating obesity, insulin resistance, non-alcoholic fatty liver disease including non-alcoholic steatohepatitis

Family Cites Families (47)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
FR2601676B1 (en) 1986-07-17 1988-09-23 Rhone Poulenc Sante PROCESS FOR THE PREPARATION OF TAXOL AND DESACETYL-10 TAXOL
US5108921A (en) 1989-04-03 1992-04-28 Purdue Research Foundation Method for enhanced transmembrane transport of exogenous molecules
US5278324A (en) 1990-08-28 1994-01-11 Virginia Tech Intellectual Properties, Inc. Water soluble derivatives of taxol
MX9102128A (en) 1990-11-23 1992-07-08 Rhone Poulenc Rorer Sa DERIVATIVES OF TAXANE, PROCEDURE FOR ITS PREPARATION AND PHARMACEUTICAL COMPOSITION THAT CONTAINS THEM
US6030954A (en) 1991-09-05 2000-02-29 University Of Connecticut Targeted delivery of poly- or oligonucleotides to cells
US5250683A (en) 1991-09-23 1993-10-05 Florida State University Certain substituted taxanes and pharmaceutical compositions containing them
US5227400A (en) 1991-09-23 1993-07-13 Florida State University Furyl and thienyl substituted taxanes and pharmaceutical compositions containing them
US5272171A (en) 1992-02-13 1993-12-21 Bristol-Myers Squibb Company Phosphonooxy and carbonate derivatives of taxol
FR2688518B1 (en) 1992-03-13 1994-05-06 Rhone Poulenc Rorer Sa PROCESS FOR THE PREPARATION OF TAXANE DERIVATIVES.
US5248796A (en) 1992-06-18 1993-09-28 Bristol-Myers Squibb Company Taxol derivatives
US5254580A (en) 1993-01-19 1993-10-19 Bristol-Myers Squibb Company 7,8-cyclopropataxanes
US5672659A (en) 1993-01-06 1997-09-30 Kinerton Limited Ionic molecular conjugates of biodegradable polyesters and bioactive polypeptides
US5801154A (en) * 1993-10-18 1998-09-01 Isis Pharmaceuticals, Inc. Antisense oligonucleotide modulation of multidrug resistance-associated protein
US5595760A (en) 1994-09-02 1997-01-21 Delab Sustained release of peptides from pharmaceutical compositions
US5882914A (en) 1995-06-06 1999-03-16 The Johns Hopkins University School Of Medicine Nucleic acids encoding the hypoxia inducible factor-1
US6770748B2 (en) * 1997-03-07 2004-08-03 Takeshi Imanishi Bicyclonucleoside and oligonucleotide analogue
US5994076A (en) * 1997-05-21 1999-11-30 Clontech Laboratories, Inc. Methods of assaying differential expression
EP2341057A3 (en) 1997-09-12 2011-11-23 Exiqon A/S Oligonucleotide Analogues
US6238921B1 (en) * 1998-03-26 2001-05-29 Isis Pharmaceuticals, Inc. Antisense oligonucleotide modulation of human mdm2 expression
WO1999048916A2 (en) 1998-03-27 1999-09-30 The Board Of Trustees Of The Leland Stanford Jr. University Hypoxia-inducible human genes, proteins, and uses thereof
AU5493899A (en) 1998-08-26 2000-03-21 Salem-Teikyo University Nucleic acid and polypeptide p10 of a borna disease virus (bdv) and their use for diagnostic and immunization purposes
US6821724B1 (en) * 1998-09-17 2004-11-23 Affymetrix, Inc. Methods of genetic analysis using nucleic acid arrays
EP1162948B1 (en) 1999-03-17 2005-11-23 Sujoy Kumar Guha An improved reversible contraceptive for male and female
ATE465168T1 (en) 1999-03-18 2010-05-15 Exiqon As XYLO-LNA ANALOGUE
WO2000056746A2 (en) 1999-03-24 2000-09-28 Exiqon A/S Improved synthesis of [2.2.1]bicyclo nucleosides
WO2000064262A1 (en) * 1999-04-26 2000-11-02 The University Of North Carolina At Chapel Hill Antisense human fucosyltransferase sequences and methods of use thereof
EP1178999B1 (en) 1999-05-04 2007-03-14 Santaris Pharma A/S L-ribo-lna analogues
WO2000076497A1 (en) 1999-06-14 2000-12-21 Cancer Research Ventures Limited Cancer therapy
CA2376284A1 (en) * 1999-06-21 2000-12-28 Murdoch Childrens Research Institute A method for the prophylaxis and/or treatment of medical disorders
US6168950B1 (en) * 1999-07-23 2001-01-02 Isis Pharmaceuticals, Inc. Antisense modulation of MEKK1 expression
WO2001019505A2 (en) 1999-09-17 2001-03-22 Millipore Corporation Patterned porous structures
AU7406700A (en) 1999-10-04 2001-05-10 Exiqon A/S Design of high affinity rnase h recruiting oligonucleotide
US6706505B1 (en) * 2000-03-08 2004-03-16 Amgen Inc Human E3α ubiquitin ligase family
PL366039A1 (en) * 2000-09-02 2005-01-24 Grunenthal Gmbh Antisense oligonucleotides against vr1
EP1334109B1 (en) 2000-10-04 2006-05-10 Santaris Pharma A/S Improved synthesis of purine locked nucleic acid analogues
US7105656B2 (en) * 2000-10-26 2006-09-12 The Brigham And Women's Hospital, Inc. Compositions and methods for treating hematologic malignancies and multiple drug resistance
WO2002094250A2 (en) 2001-05-18 2002-11-28 Cureon A/S Therapeutic uses of lna-modified oligonucleotides in infectious diseases
JP4130955B2 (en) * 2001-06-05 2008-08-13 真寛 平岡 Polypeptide that destabilizes proteins in cells under aerobic conditions and DNA encoding the same
US7153954B2 (en) * 2001-07-12 2006-12-26 Santaris Pharma A/S Method for preparation of LNA phosphoramidites
ATE287413T1 (en) 2001-07-12 2005-02-15 Santaris Pharma As METHOD FOR PRODUCING THE LNA PHOSPHORAMIDITE
SI2264172T1 (en) 2002-04-05 2017-12-29 Roche Innovation Center Copenhagen A/S Oligomeric compounds for the modulation of hif-1alpha expression
ES2290448T3 (en) 2002-05-08 2008-02-16 Santaris Pharma A/S SYSTEM OF BLOCKED NUCLEIC ACID DERIVATIVES.
WO2004042024A2 (en) 2002-11-01 2004-05-21 The Trustees Of The University Of Pennsylvania COMPOSITIONS AND METHODS FOR siRNA INHIBITION OF HIF-1 ALPHA
AU2003281969B2 (en) 2002-11-18 2011-01-27 Roche Innovation Center Copenhagen A/S Amino-LNA, thio-LNA and alpha-L-oxy-LN
US7144999B2 (en) 2002-11-23 2006-12-05 Isis Pharmaceuticals, Inc. Modulation of hypoxia-inducible factor 1 alpha expression
ES2329131T3 (en) 2003-02-10 2009-11-23 Santaris Pharma A/S OLIGOMERIC COMPOUNDS TO MODULATE SURVIVINE EXPRESSION.
JP2006518197A (en) 2003-02-10 2006-08-10 サンタリス・ファルマ・アクティーゼルスカブ Oligomeric compounds for modification of lath expression

Also Published As

Publication number Publication date
JP2005529589A (en) 2005-10-06
US7737264B2 (en) 2010-06-15
US8357670B2 (en) 2013-01-22
US20100093839A1 (en) 2010-04-15
JP4338527B2 (en) 2009-10-07
EP2264172B1 (en) 2017-09-27
US7846911B2 (en) 2010-12-07
AU2003225495B2 (en) 2009-01-15
JP2009189372A (en) 2009-08-27
US20130116309A1 (en) 2013-05-09
HUE037352T2 (en) 2018-08-28
ES2649817T3 (en) 2018-01-15
CA2480311C (en) 2015-01-27
CY1119614T1 (en) 2018-04-04
WO2003085110A3 (en) 2004-05-21
WO2003085110A2 (en) 2003-10-16
EP1501930A2 (en) 2005-02-02
DK2264172T3 (en) 2017-11-27
SI2264172T1 (en) 2017-12-29
EP2264172A1 (en) 2010-12-22
US20040096848A1 (en) 2004-05-20
US20120270924A1 (en) 2012-10-25
AU2003225495A1 (en) 2003-10-20
PT2264172T (en) 2017-12-06
US8785617B2 (en) 2014-07-22
US20110224285A1 (en) 2011-09-15
US8207140B2 (en) 2012-06-26

Similar Documents

Publication Publication Date Title
EP2264172B1 (en) Oligomeric compounds for the modulation of hif-1alpha expression
US7741309B2 (en) Oligomeric compounds for the modulation of survivin expression
EP1592793B1 (en) Oligomeric compounds for the modulation of survivin expression
EP1706489B1 (en) Oligomeric compounds for the modulation of bcl-2
US20080188432A1 (en) Oligomeric compounds for the modulation ras expression
WO2007031091A2 (en) Rna antagonist compounds for the modulation of p21 ras expression
WO2004069990A2 (en) Oligomeric compounds for the modulation of thioredoxin expression

Legal Events

Date Code Title Description
EEER Examination request
MKLA Lapsed

Effective date: 20200831