CA2474414A1 - Oligonucleotides comprising alternating segments and uses thereof - Google Patents

Oligonucleotides comprising alternating segments and uses thereof Download PDF

Info

Publication number
CA2474414A1
CA2474414A1 CA002474414A CA2474414A CA2474414A1 CA 2474414 A1 CA2474414 A1 CA 2474414A1 CA 002474414 A CA002474414 A CA 002474414A CA 2474414 A CA2474414 A CA 2474414A CA 2474414 A1 CA2474414 A1 CA 2474414A1
Authority
CA
Canada
Prior art keywords
oligonucleotide
target rna
preventing
rnase
group
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
CA002474414A
Other languages
French (fr)
Other versions
CA2474414C (en
Inventor
Michael A. Parniak
Masad J. Damha
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.)
McGill University
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 CA2474414A1 publication Critical patent/CA2474414A1/en
Application granted granted Critical
Publication of CA2474414C publication Critical patent/CA2474414C/en
Anticipated expiration legal-status Critical
Expired - Lifetime legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • 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
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • 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
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • 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
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/18Antivirals for RNA viruses for HIV
    • 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
    • A61P31/20Antivirals for DNA viruses
    • 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
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • 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
    • 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
    • C12N2310/32Chemical structure of the sugar
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/32Chemical structure of the sugar
    • C12N2310/3212'-O-R Modification
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/32Chemical structure of the sugar
    • C12N2310/3222'-R Modification
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/32Chemical structure of the sugar
    • C12N2310/323Chemical structure of the sugar modified ring structure
    • 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
    • C12N2310/34Spatial arrangement of the modifications
    • C12N2310/346Spatial arrangement of the modifications having a combination of backbone and sugar modifications

Abstract

The invention relates to oligonucleosides having alternating segments of sugar-modified nucleosides and 2~-deoxynucleosides, and uses thereof. The invention further relates to oligonucleotides having alternating segments of sugar-modified nucleotides and 2~-deoxynucleotides, and uses thereof. Such uses include the preparation of antisense oligonucleotides and their use for the prevention or depletion of function of a target nucleic acid of interest, such as an RNA, in a system. Accordingly, an oligonucleotide of the invention is useful for therapeutic, analytical and diagnostic methods and uses, as well as a component of compositions and commercial packages corresponding to such methods and uses.

Description

OLIGONUCLEOTIDES COMPRISING ALTERNATING SEGMENTS AND USES
THEREOF
FIELD OF THE INVENTION
The invention relates to oligonucleosides and oligonucleotides and uses thereof, and particularly relates to modified oligonucleosides and oligonucleotides and uses thereof.
BACKGROUND OF THE INVENTION
Oligonucleotides are utilized for a variety of biotechnological applications, based on their ability to confer specificity by virtue of their sequence composition.
Given, for example, their ability to be designed to target a protein-encoding molecule, such as RNA, a particular use of oligonucleotides is in antisense technology.
Anta.sense oligonucleota.des (AONs) Antisense oligonucleotides (AONs) have attracted considerable interest in the biotechnology sector, and have exceptional potential for use in therapeutic strategies against a range of human diseases. The formation of a duplex between the A0N and its complementary sequence on its target (usually messenger RNA [mRNA]) prevents the translation of such RNA, in part by "translation arrest" (via duplex formation between the AON and the target RNA, thus inhibiting/preventing complete translation by physically or sterically blocking the translational machinery) but more importantly by eliciting degradation of the targeted RNA
through the action of ribonuclease H (RNase H), a ubiquitous
2 and endogenous cellular enzyme that specifically degrades the RNA strand in the AON/RNA duplex.
Since the natural substrate of RNase H is a DNA/RNA
heteroduplex, DNA has been utilized for antisense technology.
However, as serum and intracellular nucleases rapidly degrade AONs with phosphodiester (PDE) linkages, AON consisting of PDE-DNA have had limited utility in such systems. DNA with phosphorothioate linkages (PS-DNA) can induce RNase H
degradation of the targeted RNA, and is resistant to degradation by ,serum and cellular nucleases, however, it forms weaker duplexes with the target RNA compared to PDE-DNA.
RNase H
RNase H selectively degrades the RNA strand of a DNA/RNA heteroduplex (Hausen, P. ; Stein, H. Eur. J. Biochem.
1970, 14, 279). Studies with eukaryotic cell extracts containing RNase H suggest that both prokaryotic and eukaryotic enzymes exhibit similar RNA-cleavage properties (Monia et al. J. Biol. Ch em. 1993, 268, 14514; Crooke et al.
Biochem J. 1995, 312, 599; Lima, W.F.; Crooke, S.T.
Biochemistry 1997, 36, 390). E. coli RNase H1 is thought to bind to the minor groove of the DNA/RNA double helix and to cleave the RNA by both endonuclease and processive 3'-to-5' exonuclease activities (Nakamura, H. et al. Proc. Natl.
Acad. ,Sci. US'A 1991, 8~, 11535; Fedoroff, O.Y. et al., J.
Mol. Biol. 1993, 233, 509). The efficiency of RNase H
degradation displays minimal sequence dependence and, as mentioned above, is quite sensitive to chemical changes in the antisense oligonucleotide.
There is therefore a need for an improved oligonucleotide, to address one or more of the limitations noted above.
3 SUMMARY OF THE INVENTION
According to a first aspect, the invention provides an oligonucleoside comprising alternating segments of sugar-modified nucleosides and 2'-deoxynucleosides, wherein the segments or units each independently comprise at least one sugar-modified nucleoside or 2'-deoxynucleoside, respectively. For example, the oligonucleoside comprises alternating first and second segments, wherein the first segment comprises at least one sugar-modified nucleoside, and wherein the second segment comprises at least one 2'-deoxynucleoside. In embodiments, the oligonucleoside comprises at least 2 of each of the first and second segments thereby comprising at least 4 alternating segments.
In an embodiment, the oligonucleoside comprises an internucleoside linkage comprising a phosphate, thereby being an oligonucleotide. In embodiments the sugar-modified nucleosides and/or 2'-deoxynucleosides comprise a phosphate, thereby being sugar-modified nucleotides and/or 2'-deoxynucleotides, a In an embodiment, the invention provides an oligonucleotide comprising alternating segments or units of arabinonucleotides and 2'-deoxynucleotides, wherein said segments or units each independently comprise at least one arabinonucleotide or 2'-deoxynucleotide, respectively. In an embodiment, the oligonucleotide comprises at least 2 arabinonucleotide segments and at least 2 2'-deoxynucleotide segments thereby having at least 4 of the alternating units.
In an embodiment, the sugar-modified oligonucleotide is capable of adopting a DNA-like conformation. In an embodiment, the sugar-modified nucleotide is selected from the group consisting of arabinonucleotides, alpha-L-locked
4 nucleic acids, cyclohexene nucleic acids, and ribonucleotides lacking an electronegative 2'-oxygen atom.
In an embodiment, the ribonucleotides lacking an electronegative 2'-oxygen atom are selected from the group consisting of 2'-alkyl-D-ribose and 2'-SCH3-D-ribose.
In an embodiment, the segments each independently comprise about 1 to about 6 arabinonucleotides or 2'-deoxynucleotides. In further embodiments, the segments each independently comprise about 2 to about 5 or about 3 to about 4 arabinonucleotides or 2'-deoxynucleotides. In a further embodiment, the segments each independently comprise about 3 arabinonucleotides or 2'-deoxynucleo'tides.
In an embodiment, the above-mentioned oligonucleotide has a structure selected from the group consisting of:
a) (Ax- Dy)n I
b ) ( Dy-Ax ) n I I
c) (Ax-Dy)m Ax-Dy-Ax III
d ) ( Dy-Ax ) i,i Dy -Ax-Dy IV
wherein each of m, x and y are each independently an integer greater than or equal to 1, n is an integer greater than or equal to 2, A is an sugar-modified nucleotide and D
is a 2'-deoxyribonucleotide.
In an embodiment, the above-mentioned sugar-modified nucleotide comprises a 2' substituent selected from the group consisting of fluorine, hydroxyl, amino, cyano, azido, -CH=CH2, -C=CH, alkyl, functionalized alkyl, alkoxy and functionalized alkoxy groups. In an embodiment, the alkyl group is a lower alkyl group. In an embodiment, the lower alkyl group is selected from the group consisting of methyl, ethyl and propyl groups. In an embodiment, the functionalized alkyl group is selected from the group consisting of methylamino, ethylamino and propylamino
5 groups. In an embodiment, the alkoxy group is selected from the group consisting of methoxy, ethoxy and propoxy groups.
In an embodiment, the functionalized alkoxy group is -0 (CH2) q-R, wherein q=2, 3 or 4 and -R is selected from the group consisting of -NH2, -OCH3, and -OCH2CH3 groups .
In an embodiment, the sugar-modified nucleotide is an arabinonucleotide. In a further emlaodiment, the 2' substituent is fluorine and the arabinonucleotide is a 2'-fluoroarabinonucleotide (2'F-ANA; also abbreviated "FANA").
In an embodiment, the above-mentioned oligonucleotide comprises one or more internucleotide linkages selected from the group consisting of:
a) phosphodiester;
b) phosphotriester;
c) phosphorothioate;
d) phosphorodithioate;
e) Rp-phosphorothioate;
f) Sp-phosphorothioate;
g) boranophosphate;
h ) methylene (methylimino ) ( 3' CH2-N ( CH3 ) -05' ) .;
i) 3'-thioformacetal (3'S-CH2-05') j) amide (3'CH2-C(0)NH-5');
k) methylphosphonate;
1) phosphoramidate (3' -OP (02) -N5' ) ; and m) any combination of (a) to (1).
In an embodiment, the above-mentioned oligonucleotide consists of about 30 or fewer nucleotides, in a further embodiment, about 8 to about 25 nucleotides, in yet a further embodiment, about 18 nucleotides.
6 In an embodiment, the above-mentioned oligonucleotide has structure I wherein x=1, y=1 and n=9, thereby having a structure:
A-D-A-D-A-D-A-D-A-D-A-D-A-D-A-D-A-D.
In an embodiment, the above-mentioned oligonucleotide has structure II wherein x=1, y=1 and n=9, thereby having a structure:
D-A-D-A-D-A-D-A-D-A-D-A-D-A-D-A-D-A.
In an embodiment, the above-mentioned oligonucleotide has structure III wherein x=2, y=2 and m=3, thereby having a structure:
A-A-D-D-A-A-D-D-A-A-D-D-A-A-D-D-A-A.
In an embodiment, the above-mentioned oligonucleotide ~0 has structure IV wherein x=2, y=2 and m=3, thereby having a structure:
D-D-A-A-D-D-A-A-D-D-A-A-D-D-A-A-D-D.
In an embodiment, the above-mentioned oligonucleotide has structure I wherein x=3, y=3 and n=3, thereby having a structure:
A-A-A-D-D-D-A-A-A-D-D-D-A-A-A-D-D-D.
In an embodiment, the above-mentioned oligonucleotide has structure II wherein x=3, y=3 and n=3, thereby having a structure:
7 D-D-D-A-A-A-D-D-D-A-A-A-D-D-D-A-A-A.
In an embodiment, the above-mentioned oligonucleotide has structure III wherein x=4, y=3 and m=1, thereby having a structure:
A-A-A-A-D-D-D-A-A-A-A-D-D-D-A-A-A-A.
In an embodiment, the above-mentioned oligonucleotide has said structure IV wherein x=4, y=3 and m=1, thereby having a structure:
D-D-D-D-A-A-A-D-D-D-D-A-A-A-D-D-D-D.
15~
In an embodiment, the above-mentioned oligonucleoside further comprises a third segment comprising a modified nucleoside, wherein said third segment is adjacent to (a) the 5' end of said alternating first and second segments, (b) the 3' end of said alternating first and second segments, or (c) both (a) and (b).
In an embodiment, the above-mentioned oligonucleotide further comprises a third segment comprising a modified nucleotide, wherein said third segment is adjacent to (a) the 5' end of said alternating first and second segments, (b) the 3' end of said alternating first and second segments, or (c) both (a) and (b). In an embodiment, the modified nucleotide is a modified ribonucleotide. In an embodiment, the modified ribonucleotide comprises a modification at its 2' position. In an embodiment, the 2' modification is selected from the group consisting of methoxy, methoxyethyl, fluoro and propylamino groups.
8 In an embodiment, the above-mentioned oligonucleotide is,antisense to a target RNA.
The invention further provides a method of preventing or decreasing translation, reverse transcription and/or replication of a target RNA in a system, said method comprising contacting said target RNA with the above-mentioned oligonucleotide. In an embodiment, the system is selected from the group consisting of a cell, tissue or subject. In an embodiment, the cell, tissue or subject is a mammalian cell, tissue or subject, in a further embodiment, a human cell, tissue or subject.
The invention further provides a method of inducing RNase H-mediated cleavage of a target RNA in a system, the method comprising contacting the target RNA with the above-mentioned oligonucleotide. In an embodiment, the RNase H-mediated cleavage is effected by RNase H activity associated with a reverse transcriptase of a virus. In an embodiment, the virus is a human pathogenic virus, in a further embodiment, the virus is selected from the group consisting, of HIV (e. g. HIV-1 and HIV-2) and hepadnaviruses (e. g.
hepatitis B virus). In an embodiment, the RNase H-mediated cleavage is effected by RNase H activity associated with an RNase H enzyme of prokaryotic or eukaryotic origin. In an embodiment, the euka.ryotic RNase H is a mammalian RNase H, in a further embodiment, a human RNase H (e.g. RNase H1 and RNase H2).
The invention further provides a method of preventing or decreasing translation, reverse transcription and/or replication of a target RNA in a system, and/or for detecting the presence of a target RNA in a system and/or validating . a gene target in a system, said method comprising:
9 a) contacting the target RNA with the above-mentioned oligonucleotide; and b) allowing RNase H cleavage of the target RNA.
The invention further provides a method of effecting a process selected from the group consisting of:
(a) inducing RNase H-mediated cleavage of a target RNA
in a system;
(b) preventing or decreasing translation of a target RNA in a system;
(c) preventing or decreasing reverse transcription of a target RNA in a system;
(d) preventing or decreasing replication of a target RNA in a system (e) detecting the presence of a target RNA in a system ( f ) validating a gene 'target corresponding to a target RNA in a system;
(g) preventing or treating a disease related to a target RNA in a system; and (h) any combination of (a) to (g);
said method comprising contacting said target RNA with the above-mentioned oligonucleotide.
The invention further provides a method of effecting a process selected from the group consisting of:
(a) inducing RNase H-mediated cleavage of a target RNA
in a system;
(b) preventing or decreasing translation of a target RNA in a system;
(c) preventing ar decreasing reverse transcription of a target RNA in a system;
(d) preventing or decreasing replication of a target RNA in a system (e) detecting the presence of a target RNA in a system (f) validating a gene target corresponding to a target RNA in a system;
(g) preventing or treating a disease related to a target RNA in a system; and 5 (h) any combination of (a) to (g);
said method comprising introducing the above-mentioned oligonucleotide into said system.
The invention further provides a use of the above mentioned oligonucleotide for a medical or research use. In
10 embodiments, the medical or research use is selected from the group consisting of:
(a) inducing RNase H-mediated cleavage of a target RNA
in a system;
(b) preventing or decreasing translation of a target RNA in a system;
(c) preventing or decreasing reverse transcription of a target RNA in a system;
(d) preventing or decreasing replication of a target RNA in a system (e) detecting the presence of a target RNA in a system (f) validating a gene target in a system;
(g) preventing or treating a disease related to a target RNA in a system; and (h) any combination of (a) to (g).
The invention further provides a use of the above-mentioned oligonucleotide for the preparation of a medicament. In an embodiment the medicament is for a use selected from the group consisting of:
(a) inducing RNase H-mediated cleavage of a target RNA
in a system;
(b) preventing or decreasing translation of a target RNA in a system;
11 (c) preventing or decreasing reverse transcription of a target RNA in a system;
(d) preventing or decreasing replication of a target RNA in a system;
(e) detecting the presence of a target RNA in a system;
(f) validating a gene target in a system;
(g) preventing or treating a disease related to a target RNA in a system; and (h) any combination of (a) to (g).
The invention further provides a composition comprising the alcove-mentioned oligonucleotide in admixture with a pharmaceutically acceptable carrier. In an embodiment, the composition is for a use selected from the group consisting of (a) inducing RNase H-mediated cleavage of a target RNA
in a system;
(b) preventing or decreasing translation of a' target RNA in a system;
(c) preventing or decreasing reverse transcription of a target RNA in a system;
(d) preventing or decreasing replication of a target RNA in a system;
(e) detecting the presence of a target RNA in a system;
(f) validating a gene target in a system;
(g) preventing or treating a disease related to a target RNA in a system; and (h) any combination of (a) to (g).
The invention further provides a commercial package comprising the above-mentioned oligonucleotide together with instructions for its use. In an embodiment the instructions are for a use selected from the group consisting of:
(a) inducing RNase H-mediated cleavage of a target RNA
in a system;
12 (b) preventing or decreasing translation of a target RNA in a system;
(c) preventing or decreasing reverse transcription of a target RNA in a system;
(d) preventing br decreasing replication of a target RNA in a system;
(e) detecting the presence of a target RNA in a system;
(f) validating a gene target in a system;
(g) preventing or treating a disease related to a target RNA in a system; and (h) any combination of (a) to (g).
BRIEF DESCRIPTION OF THE DRAWINGS
Figure 1. Structures of examples of certain nucleotide components utilized in antisense oligonucleotides (AONs).
Figure 2. Human RNase HII mediated cleavage of RNA duplexed with various antisense oligonucleotides according to certain embodiments of the invention. (A). Electrophoretic analysis of 32P-labeled target RNA degradation products. AON/5'-[3~P]-RNA duplexes were incubated with human RNase HII at room temperature, and aliquots were taken at 0, 5, 10, and 20 min, electrophoresed and reaction products visualized by autoradiography. (B). Residual full-length 5'-[32P]-target as a function of reaction time. Data were obtained by densitometric analysis of the autoradiogram shown in A.
Figure 3. Ability of the various AON listed in Table 1, according to certain embodiments of the invention, to elicit RNase H degradation of target RNA. AON/5' - [32P] -RNA duplexes were incubated with human RNase HII (black bars) or E. coli RNase HI (shaded bars) for 10 minutes at room temperature,
13 then the reaction mixtures were resolved by electrophoresis, visualized by autoradiography, and the loss of intact RNA
was quantified by densitometry. Values are normalized to those found for the all 2'F-ANA AON 2 as 100%.
DETAILED DESCRIPTION OF THE INVENTION
The invention relates to oligonucleosides comprising alternating segments ("altimers") of sugar-modified nucleosides and 2'-deoxynucleosides. In an embodiment, The oligonucleoside or nucleosides comprise a phosphate, thereby being oligonucleotide or nucleotides, respectively.
In an embodiment, such "altimers" comprise alternating segments of arabinonucleotide (ANA) such as 2'F-ANA (or FANA) and DNA. "Arabinonucleotide" as used herein refers to a nucleotide comprising an arabinofuranose sugar.
Results presented herein include studies of (1) oligonucleotide binding affinity to target RNA and (2) the ability of oligonucleotide to elicit RNase H cleavage of a target RNA. Both phosphodiester and phosphorothioate linked "altimers" were evaluated in the results described herein.
Accordingly, the invention relates to modified oligonucleotides which in embodiments are used to selectively prevent gene expression in a sequence-specific manner. In an embodiment, the invention relates to the selective inhibition of protein biosynthesis via an antisense strategy using short strands comprising alternating segments or units of sugar-modified nucleic acids (e. g. arabinonucleic acids [e. g. FANA]) and DNA. Each segment or unit may contain one or more nucleotides. In embodiments the invention relates to the use of modified oligonucleotides comprising alternating units of sugar-modified nucleic acids and DNA, to hybridize to
14 complementary (in an embodiment, exactly complementary) RNA
such as cellular messenger RNA, viral RNA, etc. In a further embodiment, the invention relates to the use of such modified oligonucleotides to hybridize to and induce cleavage of complementary RNA via RNase H
activation/induction of RNase H activity.
In an embodiment, the invention relates to antisense oligonucleotide (AON) chimeras constructed from sugar-modified nucleotides and 2'-deoxyribonucleotides, which in certain embodiments are modified, that are capable of forming a duplex with a target RNA sequence. In an embodiment, the resulting AON/RNA duplexes are substrates for RNase H, an enzyme that recognizes such a duplex and degrades the RNA target portion. RNase H-mediated cleavage of RNA targets is considered to be a major mechanism of action of antisense oligonucleotides.
The present inventidn relates to the unexpected and surprising discovery that antisense chimeras constructed from alternating units or segments of a sugar-modified nucleotides such as modified ANA (such as 2'-deoxy-2'-fluoro-(3-D-arabinonucleotides [FANA]), and 2'-deoxynucleotides (DNA), each unit containing one or more such residues, are superior at eliciting RNase H (e. g.
eukaryotic RNase H) activity in vitro compared to (a) the native DNA structure, and (b) uniformly modified FANS
oligomers. Similarly, the present invention shows that the RNase H competency of oligodeoxynucleotides (such as DNA) can be improved by inserting such sugar-modified nucleotides (e.g. arabinonucleotide [e.g. FANA]) residues within the oligonucleotide chain. Accordingly, oligonucleotides of the invention comprising alternating units or segments of (modified) sugar-modified nucleotide and deoxyribonucleotide, are useful as therapeutic agents and/or tools for the study and control of specific gene expression in cells and organisms, e.g. for a variety of medical and research uses. The oligonucleotides of the invention are also useful for diagnostic and detection methods to identify 5 the presence of~a particular nucleic acid, based on their ability to target the nucleic acid.
"Sugar-modified nucleoside" or "sugar-modified nucleotide" as used herein refers to a nucleoside or nucleotide, respectively, which has a different or modified 10 sugar structure as compared to the sugar moiety of a native deoxyribonucleoside or deoxyribonucleotide, respectively, or ribonucleoside or ribonucleotide, respectively. Such modifications include but are not limited to changes in conformation of the sugar ring, substitution or addition'of
15 different ring- structures, and the modification (substitution, deletion or addition) of any sugar ring substituents. In a further embodiment, such a sugar-modified nucleoside or nucleotide is capable of adopting a DNA-like conformation. A "DNA-like conformation" as used herein refers to the sugar structure of the nucleoside or nucleotide, and refers to a conformation which resembles the conformation of a native 2'-deoxyribonucleoside or 2'-deoxyribonucleotide residue, i.e. one whose sugar residue is capable of adopting a C2'-endo (south pucker) and/or 04'-endo (east pucker) conformation. As arabinonucleotides may adopt such a C2'-endo (south pucker) and/or 04'-endo (east pucker) conformation,. arabinonucleic acids and DNA exhibit similar conformational preferences (Venkateswarlu, D. et al.
J. elm. Chem. Soc. 1999, 121, 5609; Trempe, J-F. et al., J.
Am. Chem. Soc. 2001, 123, 4896; Denisov, A.Y. et al., Nucleic .Acids Res. 2001, 29, 4284), and thus in embodiments ANA and its derivatives (e. g. FANA), are a type of DNA-like nucleotide as defined herein. Other DNA-like nucleotides
16 include but are not limited to alpha-L-LNA (Petersen, M. et al., J. Am. Chem. Soc. 2001; 123; 7431) and cyclohexene nucleic acids Wang,1 J. et al., J. Am. Chem. Soc., 2000, 122,8595).
In embodiments, the internucleotide linkages of the oligonucleotides of the invention include but are not limited to phosphodiester, phosphotriester, phosphorothioate (5' 0-P (S) O-3' 0-, 5' S-P (0) 0-3' 0-, and 5' 0-P (O) 0-3' S-) , phosphorodithioate, Rp-phosphorothioate, Sp-phosphorothioate, boranophosphate, methylene(methylimino), amide (3'-CH2-CO-NH-5' and 3'-CH2-NH-CO-5'), methylphosphonate, 3'-thioformacetal, (3'S-CH2-05'), amide (3'CH2-C(O)NH-5'); phosphoramidate (e.g. 5'N-3'P) groups or any combinations thereof. The 2'-substituent, e.g. of the arabinose sugar in ANA residues, includes but is not limited to fluorine, hydroxyl, amino, cyano, azido, -CH=CH2, -C=CH, alkyl (e. g. lower alkyl [e. g. C1-C9 alkyl] e.g. methyl, ethyl, propyl, etc.), alkoxy ([e.g. lower alkoxy, e.g. C1-C9 alkoxy] e.g. methoxy, ethoxy, proproxy, etc.) and funotionali~ed alkyl (e. g. functionalized lower alkyl [e.g.2'-CF3]) and alkoxy groups (e. g. ethylamino, propylamino and butylamino groups), and alkoxyalkyl (e. g.
methoxyethyl, ethoxyethyl, etc.) groups. In an embodiment, the 2' substituent of the arabinose sugar is fluorine and the arabinonucleotide derivative is 2'F-ANA (or FANA). In addition to those described above, the arabinose sugar also includes the carbocyclic (4'-CH2) derivative (e. g., carbocyclic FANA). In embodiments, the sugar modified nucleotide comprises other backbones that elicit RNase H
activity (e. g., alpha-L-locked nucleic acids, cyclohexene nucleic acids), or by riboses lacking the electronegative 2°-oxygen atom (e. g., 2'-alkyl-D-ribose, 2'-SCH3-D-ribose).
17 Applicants demonstrate herein that mixed backbone AON
comprising alternating segments of a sugar-modified nucleotide (e.g. ANA [e.g. FANA]) and DNA ("altimers") are capable of eliciting RNase H (e. g. human RNase HII) degradation of target RNA. Certain "altimer" AON, namely those possessing alternating trinucleotide segments, are particularly better in this regard.
Therefore, an oligonucleotide of the invention comprises alternating segments or units of sugar-modified nucleotides (e.g. arabinonucleotide analogues [e.g., FANA]) and 2'-deoxyribonucleotides (DNA). In an embodiment, the oligonucleotide comprises at least 2 of each of sugar-modified nucleotide and 2'-deoxynucleotide segments, thereby having at least 4 alternating segments overall., Each alternating segment or unit may contain 1 or a plurality of nucleotides. In embodiments, the plurality of nucleotides may consist of 2, 3, 4, 5 or 6 nucleotides. The oligonucleotide may contain in embodiments an odd or even number of alternating ~ segments or units. The oligonucleotide may commence and/or terminate with a segment containing sugar-modified nucleotide residues or DNA
residues. Accordingly, in embodiments, the oligonucleotides of the invention may be represented as follows:
A1-D1-A2-D2-A3-D3 ... AZ-D
Where each of Al, Az, etc. represents a unit of one or more sugar-modified nucleotide residues and each of D1, D2, etc.
represents a unit of one or more DNA residues. The number of residues within each unit may be the same or variable from one unit to another. The oligonucleotide may have an odd or an even number of units. The oligonucleotide may start (i.e. at its 5' end) with either an ANA-containing
18 unit or a DNA-containing unit. The oligonucleotide may terminate (i.e. at its 3' end) with either an sugar-modified nucleotide-containing unit or a DNA-.containing unit. The total number of units may be as few as 4 (i.e. at least 2 of each type).
In embodiments, the ~~altimer" portion of an oligonucleoside or oligonucleotide of the invention may further comprise one or more modified nucleosides or nucleotides at (i.e. adjacent to) its 5' and/or 3' ends, including but not limited to modified ribonucleosides, or ribonucleotides, such as 2'-modified ribonucleosides or ribonucleotides, such as 2'-methoxy RNA (2'-O-Me-RNA) or 2'-methoxyethyl RNA (2'-MOE-RNA). Such a 2'-0-Me-RNA - altimer - 2'-Ome-RNA based oligonucleotide is capable~of eliciting RNase H activity of a suitable RNA target, as described in the Examples herein.
In embodiments, the overall lengtri of an oligonucleotide of the invention is about 30 or fewer nucleotide residues, in a further embodiment about 8 to about 25 nucleotide residues. In further embodiments, the length is about 9 to about 24, about 10 to about 23, about 11 to about 22, about 12 to about 21, about 13 to about 20, about 14 to about 19, about 15 to about 18, or about 16 to about 17 nucleotide residues. In an embodiment, the length of an oligonucleotide of the invention is 18 nucleotide residues.
In embodiments, DNA residues may contain any of the bases selected amongst adenine (A), cytosine (C), guanine (G) or thymine (T) or versions comprising modifications of the nucleotide base or backbone structures. In embodiments, ANA residues may contain any of the bases selected amongst adenine (A), inosine (I), 2,6-diaminopurine (2;6-DAP),
19 cytosine (C), 5-methylcytosine (5meC), guanine (G) or thymine (T) or uracil (U).
The AONs of this invention contain a sequence that is complementary (in certain embodiments partially complementary, and in other embodiments exactly complementary) to a "target RNA". "Hybridization" as used herein refers to hydrogen bonding between complementary nucleotides. The degree of complementarity between an AON
and its target sequence may be variable, and in embodiments the AON is exactly complementary to its target sequence as noted above. It is understood that it is not essential that an AON be exactly complementary to its target sequence to achieve sufficient specificity, i.e. to minimize non-specific binding of the oligonucleotide to non-target sequences under the particular binding conditions being used (e. g. in vivo physiological conditions or in vitro assay conditions). "Target RNA" refers to an RNA molecule of interest which is the target for hybridizing with/binding to an oligonucleotide of the invention to prevent or decrease for example the translation, reverse transcription and or replication of the RNA. In embodiments, such prevention and inhibition is via an induction of RNase H-mediated cleavage of the target RNA, and therefore in an embodiment, the invention provides a method of cleaving a target RNA, said method comprising contacting the RNA with an oligonucleotide of the invention. In embodiments, such cleavage may be further facilitated by' additionally providing conditions conducive to RNase H activity, such as buffer means (e. g. to ~ control pH and ionic strength),.
temperature control means, and any other components which may contribute to an induction in RNase H activity. In certain embodiments, RNase H activity is of an RNase H
enzyme or of a multifunctional enzyme possessing RNase H

activity (e. g., HIV reverse transcriptase). In certain embodiments, such RNase H ,activity includes, but is not limited to RNase H activity associated with the reverse transcriptases of human pathogenic viruses such. as HIV (e. g.
5 the retroviruses HIV-1 and HIV-2) and hepadnavirus, e.g.
hepatitis B virus. In further embodiments, such RNase H
activity includes, but is not limited to RNase H activity associated with an RNase H enzyme of prokaryotic or eukaryotic origin, in an embodiment, of mammalian origin, in 10 an embodiment, of human origin. In further embodiments, such RNase H activity includes, but is not limited to ,RNase H activity associated with RNase H1 and RNase H2 (sometimes referred to as RNase HII) of eukaryotic or prokaryotic origin. In an embodiment, such RNase H activity is 15 associated with human RNase H2.
In embodiments, the above-noted RNA includes messenger RNA, or viral genomic RNA, such that the oligonucleotide can specifically inhibit the biosynthesis of proteins encoded by the mRNA, or inhibit virus replication, respectively.
20 Partial modifications to the oligonucleotide directed to the 5' and/or 3'-terminus, or the phosphate backbone or sugar residues to enhance their antisense properties (e. g.
nuclease resistance) are within the scope of the invention.' As demonstrated in this invention (vida infra), these oligonucleotides meet one of the requirements for antisense therapeutics, i.e., they are capable of binding to target RNA forming an AON/RNA duplex, which in an embodiment is recognized and degraded by RNase H. Furthermore, as shown in the Examples below, the efficiency by which the "altimer"
oligonucleotides of the invention promote RNA cleavage is superior to that seen with AON containing only FANA and in some cases superior that seen with AON containing only DNA
residues. This holds true whether the internucleotide
21 linkages of the "altimer" are phosphodiester or phosphorothioate linkages.
Wherefore, the results presented herein establish that the "altimer"-comprising oligonucleosides or oligonucleotides of the invention can in embodiments be used as antisense agents, and should serve as therapeutics and/or valuable tools for studying and controlling gene expression in cells and organisms.
As such, in alternative embodiments, the invention provides antisense molecules that bind to, induce degradation of and/or inhibit the translation of (e.g. by inducing RNase H activity and/or by effectingr"translational arrest" or block.ixig) a target RNA (e. g. mRNA) . Examples of therapeutic antisense oligonucleotide applications, incorporated herein by reference, include: U.S. Pat. No.
'5,135,917, issued Aug. 4, 1992; U.S. Pat. No. 5,098,890, issued Mar. 24, 1992 U.S. Pat. No. 5,087,617, issued Feb.
~11, 1992; U.S. Pat. No. 5,166,195 issued Nov. 24, 1992; U.S.
Pat. No. 5,004,810, issued Apr. 2, 1991 U.S. Pat. No.
5,194,428, issued Mar. 16, 1993; U.S. Pat. No. 4,806,463, issued Feb. 21, 1989; U.S. Pat. No. 5,286,717 issued Feb.
15, 1994; U.S. Pat. 'No. 5,276,019 and U.S. Pat. No.
5,264,423; BioWorld Today, Apr. 29, 1994, p. 3.
. Preferably, in antisense molecules, there is a sufficient degree of complementarity to the target RNA to avoid non-specific binding of the antisense molecule to non target sequences under conditions in which specific binding is desired, such as under physiological conditions in the case of in vivo assays or therapeutic treatment or, in the case of in vitro assays, under conditions in which the assays are conducted. The target RNA for antisense binding may include not only the information to encode a protein, but also associated ribonucleotides, which for example form
22 the 5'-untranslated region, the 3'-untranslated region, the 5' cap region and intron/exon junction ribonucleotides. A
method of screening for antisense and ribozyme nucleic acids that may be used to provide such molecules as PLA2 inhibitors of the invention is disclosed in U.S. Patent No. 5,932,435.
Antisense molecules (oligonucleosides or oligonucleotides) of the invention may include those which contain intersugar backbone linkages such as phosphotriesters, methyl phosphonates, 3'-thioformacetal, amide, short chain alkyl or cycloalkyl intersugar linkages or short chain heteroatomic or heterocyclic intersugar linkages, phosphorothioates and those with CH2--NH--0--CH2, CH2--N(CH3)--0--CH2 (known as methylene(methylimino) or MMI
backbone ) , CHI--0--N ( CH3 ) --CH2, CH2--N ( CH3 ) --N ( CH3 ) --CH2 and O--N(CH3)--CH2 --CH2 backbones (where phosphodiester is 0-P(0)2--0--CH2). In alternative embodiments, antisense oligonucleotides may have a peptide nucleic acid (PNA, sometimes referred to as "protein" or "peptide" nucleic acid) backbone, in which the phosphodiester backbone of the oligonucleotide may be replaced with a polyamide backbone wherein nucleosidic bases are bound directly or indirectly to aza nitrogen atoms or methylene groups in the polyamide backbone (Nielsen et al., Science, 1991, 254, 1497 and U.S.
Pat. No. 5,539,082). The phosphodiester bonds may be substituted with structures that are chiral and i enantiomerically specific.
As noted above, oligonucleotides may also include species which include at least one modified nucleotide base.
Thus, purines and pyrimidines other than those normally found in nature may be used. As noted above, a nucleotide of the sugar-modified nucleotide segment (e. g. ANA segment) may comprise modifications on its pentofuranosyl portion.
Examples of such modifications are 2'-0-alkyl- and 2'-
23 halogen-substituted nucleotides. Some specific examples of modifications at the 2' position of sugar moieties which are useful in the present invention are OH, SH, SCH3, F, OCN, 0 (CH2) n NH2 or O (CH2) n CH3 where n is from 1 to about 10; Cl to Clo lower alkyl, substituted lower alkyl, alkaryl or aralkyl;
C1; Br; CN; CF3 ; OCF3 ; 0-, S-, or N-alkyl; 0-, S-, or N-alkenyl; ' SOCH3 ; S02 CH3; ONOZ ; N02 ; N3; NH2;
heterocycloalkyl; heterocycloalkaryl; aminoalkylamino;
polyalkylamino; substituted silyl; an RNA cleaving group; a reporter group; an intercalator; a group for improving the pharmacokinetic~properties of an oligonucleotide; or a group for improving the pharmacodynamic properties of an oligonucleotide and other substituents having similar properties. One or more pentofuranosyl groups of the nucleotide of the sugar-modified nucleotide segment may be replaced by another sugar, by a sugar mimic such as cyclobutyl or by another moiety which takes the place of the sugar.
"Nucleoside" refers to a base (e.g. a purine [e.g. A
and G] or pyrimidine [e. g. C, 5-methyl-C, T and U]) combined with a sugar (e.g. [deoxy]ribose, arabinose and derivatives). " Nucleotide" refers to a nucleoside having a phosphate group attached to its sugar moiety. In embodiments these structures may include various modifications, e.g. either in the base, sugar and/or phosphate moieties. "Modified nucleotide/nucleoside" as used herein refers to a nucleotide/nucleoside that differs from and thus excludes the defined native form.
"Oligonucleotide" as used herein refers to a sequence comprising a plurality of nucleotides joined together. An oligonucl~eotide may comprise modified structures in its backbone structure and/or in one or more of its component nucleotides. In embodiments, oligonucleotides of the
24 invention are about 1 to 200 bases in length, in further embodiments from about 5 to about 50 bases, from about' 8 to about 40 bases, and yet further embodiments, from about 12 to about 25 bases in length.
"Alkyl" refers to straight and branched chain saturated hydrocarbon groups (e. g. methyl, ethyl, propyl, butyl, isopropyl etc.). "Alkenyl" and "alkynyl" refer to hydrocarbon groups having at least one C-C double and one C-C triple bond, respectively. "Alkoxy" refers to an -0-alkyl structure. "Alkylamino" refers to -NH(alkyl) or -N(alkyl)2 structures. "Aryl" refers to substituted and unsubstituted aromatic cyclic structures (e. g. phenyl, naphthyl, anthracyl, phenanthryl, pyrenyl, and xylyl groups). "Hetero" refers to an atom other than C; including but not limited to N, 0, or S. In embodiments, the above-mentioned groups may be substituted.
Accordingly, in various embodiments, a modified oligonucleotide of the invention may be used therapeutically in formulations or medicaments to prevent or treat a disease characterized by the expression of a particular target RNA.
In certain embodiments, such a target nucleic acid is contained in or derived from an infectious agent and/or is required for the function and/or viability and/or replication/propagation of the infectious agent. In certain embodiments, such an infectious agent is a virus, in certain embodiments, a retrovirus, in a further embodiment, HIV. In further embodiments the expression of such a target nucleic acid is associated with the diseases including but not limited to inflammatory diseases, diabetes, cardiovascular disease (e.g. restinos.is), and cancer. The invention provides corresponding methods of medical treatment, in which a therapeutic dose of a modified oligonucleotide of the invention is administered in a pharmacologically acceptable formulation. Accordingly, the invention also provides therapeutic compositions comprising a modified oligonucleotide of the invention, and a pharmacologically acceptable excipient or carrier. The 5 therapeutic composition may be soluble in an aqueous solution at a physiologically acceptable pH.
In an embodiment, such compositions include an oligonucleotide of the invention in a therapeutically or prophylactically effective amount sufficient to treat or 10 prevent a disease characterized by the expression of a particular target nucleic acid, and a pharmaceutically acceptable carrier.
A "therapeutically effective amount" refers to an amount effective, at dosages and for periods of time 15 necessary, to achieve the desired therapeutic result, such as a decrease in or a prevention of the expression ,of a particular target nucleic acid. A therapeutically effective amount of a modified nucleic acid of the invention may vary according to factors such as the disease state, age, sex, 20 and weight of the individual, and the ability of the modified nucleic acid to elicit a desired response in the individual. Dosage regimens may be adjusted to provide the optimum therapeutic response. A therapeutically effective amount is also one in which any toxic or detrimental effects
25 of the compound are outweighed by the therapeutically beneficial effects. A "prophylactically effective amount"
refers to an amount effective, at dosages and for periods of time necessary, to achieve the desired prophylactic result, such as preventing or treating a disease characterized by the expression of a particular target nucleic acid. A ' prophylactically effective amount can be determined as described above for the therapeutically effective amount.
For any particular subject, specific dosage regimens may be
26 adjusted over time according to the individual need and the professional judgement of the person administering or supervising the administration of the compositions.
As used herein "pharmaceutically acceptable carrier" or "excipient" includes any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like that are physiologically compatible. In one embodiment, the carrier is suitable for parenteral administration. Alternatively, the carrier ~ can be suitable for intravenous, intraperitoneal, intramuscular, sublingual or oral administration. Pharmaceutically acceptable carriers include sterile aqueous solutions or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersion. The use of such media and agents for pharmaceutically active substances is well known in the art. Except insofar as any conventional media or agent is incompatible with the active compound, use thereof in the pharmaceutical compositions of the invention is contemplated. Supplementary active compounds can also be incorporated into the compositions.
Therapeutic compositions typically must be sterile and stable under the conditions of manufacture and storage. The composition can be formulated as a solution, microemulsion, liposome, or other ordered structure suitable to, high drug concentration. The carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, and liquid polyethylene glycol, and the like), and suitable mixtures thereof. The proper fluidity can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants. In many cases, it will be preferable to
27 include isotonic agents, for example, sugars, polyalcohols such as mannitol, sorbitol, or sodium chloride in the composition. Prolonged absorption of the injectable compositions can be brought about by including in the composition an agent which delays absorption, for example, monostearate salts and gelatin. Moreover, an oligonucleotide of the invention can be administered in a time release formulation, for example in a composition which includes a slow release polymer. The modified oligonucleotide can be prepared with carriers that will protect the modified oligonucleotide against rapid release, such as a controlled release formulation, including implants and mi,croencapsulated delivery systems. Biodegradable, biocompatible polymers can be used, such as ethylene vinyl acetate, polyanhydrides, polyglycolic acid, collagen, polyorthoesters, polylactic acid and polylactic, polyglycolic copolymers (PZG). Many methods for the preparation of such formulations are patented or generally known to those skilled in the art.
Sterile injectable solutions can be prepared by incorporating an active compound, such as an oligonucleotide of the invention, in the required amount in an appropriate solvent with one or a combination of ingredients enumerated above, as required, followed by filtered sterilisation.
Generally, dispersions are prepared by incorporating the active compound into a sterile vehicle which contains a basic dispersion medium and the required other ingredients from those enumerated above. In the case of sterile powders for the preparation of sterile injectable solutions, the preferred methods of preparation are vacuum drying and freeze-drying which yields a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof. In accordance with an
28 , alternative aspect of the invention, an oligonucleotide of the invention may be formulated with one or more additional compounds that enhance its. solubility.
Since the oligonucleotides of the invention are capable of inducing the RNase H-mediated cleavage of a target RNA, thus decreasing the production of the protein encoded by the target RNA, the modified oligonucleotides of the invention may be used in any system where the selective inactivation or inhibition of a particular target RNA is desirable. As noted above, examples of such uses include antisense therapeutics, in which expression of the target RNA is associated with illness or disease.
A further example of such a use is the selective depletion of a particular target gene product in a system to study the phenotypic effects) of such depletion on the system. Observations made via such depletion studies may thus allow the determination of the function of the target gene product. In certain embodiments, such uses include "target validation", in which the above-described strategy enables the confirmation as to whether a particular target nucleic acid is associated with a particular phenotype or activity, and thus allows "validation" of the target. The above noted system may be cell or cell-free; in vitro or in vivo; prokaryotic or eukaryotic.
The invention further provides commercial packages comprising an oligonucleotide of the invention. In an embodiment, the commercial package further comprises instructions for use of the oligonucleotide. In certain embodiments, such instructions for use include at least one of the following: use of the oligonucleotide for (a) decreasing the expression of a target RNA sequence; (b) inducing the RNase H cleavage of a target RNA sequence; (c) preventing or treating a disease characterized by the
29 expression of a particular RNA target; (d) preventing or decreasing reverse transcription of a target RNA in a system;~(e) preventing or decreasing replication of a target RNA in a system;(f) detecting the presence of a target RNA
in a system;(g) validating a gene target in a system; and (h) any combination of (a) to (g).
The invention further provides a use of an oligonucleotide of the invention, such as for (a) decreasing the expression of a target RNA sequence; (b) inducing the RNase H cleavage of a target RNA sequence; (e)~ preventing or treating a disease characterized by the expression of a particular RNA target; (d) preventing or decreasing reverse transcription of a target RNA in a system;(e) preventing or decreasing replication of a target RNA in a system;(f) detecting the presence of a target RNA in a system;(g) validating a gene target in a system; and (h) any combination of (a) to (g).
The invention further provides a use of an oligonucleotide of the invention for the preparation of a medicament, such as for (a) decreasing the expression of. a target RNA sequence; (b) inducing the RNase H cleavage of a target RNA sequence; (c) preventing or treating a disease characterized by the expression of a particular RNA target;
(.d) preventing or decreasing reverse transcription of a target RNA in a ~system;(e) preventing or decreasing replication of a target RNA in a system;(f) detecting the presence of a target RNA in a system;(g) validating a gene target in a system; and (h) any combination of (a) to (g).
Although various embodiments of the invention are disclosed herein, many adaptations arid modifications may be made within the scope of the invention in accordance with the common general knowledge of those skilled in this art.
Such. modifications include the substitution of known equivalents for any aspect of the invention in order to achieve the same result in 'substantially the same way.
Numeric ranges are inclusive of the numbers defining the range. In the claims, the word "comprising" is used as an 5 open-ended term, substantially equivalent to the phrase "including, but not limited to". The following examples are illustrative of various aspects of the invention, and do not limit the broad aspects of the invention as disclosed herein. , EXAMPhES
Example 1: Materials and methods Synthesis of AONs. 5'-Monomethoxytritylated 2'-deoxy-2'-fluoroarabinonucleoside 3'-O-cyanoethylphosphoramidite monomers were synthesized as previously described [Wilds, C.J. & Damha, M.J. Nucleic Acids Res. 2000, 2~, 3625;
Elzagheid, M.I. et al., In Current Protocols in Nucleic Acid Chemistry, Unit 1.7, Beaucage, S. L., Bergstrom, D. E., Gli, G. D., Eds., 2002]. Synthesis of oligonucleotides shown on Table 1 and 2 were synthesized on a 1-micromole scale using an Expedit a 8909 DNA-synthesizer. Long-chain alkylamine controlled-pore glass (LCAA-CPG) was used as the solid support. The synthesis cycle consisted of the following steps: (a) Detritylation of nucleoside/tide bound to CPG
(3o trichloroacetic acid/dichloromethane): 150 sec.; (b) coupling of 2'-F-arabinonucleoside (15 min) or 2'-deoxyribonucleoside 3'-phosphoramidite (2 min) monomers.
Concentration of monomers used were 50 mg/mL for araF-T, araF-C and DNA monomers, and 60 mg/mL for araA and araF-G
(acetonitrile as solvent); (c) acetylation using the standard capping step: 20 sec. The capping solution consisted of 1:1 (v/v) of "cap A" and "cap B" reagents.(Cap A: acetic anhydride/collidine/THF, 1:1:8; cap B: N-Methylimidazole/THF, 4:21); (d) extensive washing with acetonitrile (50 pulses); (e) 20-second iodine/water oxidation (in the case of phosphodiester linked oligomers) or 10-min sulfuration (in the case of PS-oligomers) with a fresh solution of 0.1 M 3-amino-1,2,4-dithiazoline-5-thione (ADTT) in pyridine/acetonitrile (1/1, v/v); (f) washing with acetonitrile: 20 pulses; (g) drying of the solid support by addition of the capping reagent (see step,c above): 5 sec;
(h) washing with acetonitrile (20 pulses).
Following chain assembly, oligonucleotides were cleaved from the solid support and deprotected as previously described [Wilds, C.J. & Damha, M.J. Nucleic Acids Res. 2000, 2~, 3625; Viazovkina, E. et al., In Current Protocols in Nucleic Acid Chemistry, Unit 4.15, Beaucage, S.L., Bergstrom, D. E., Gli, G. D., Eds., 2002]. The crude oligomers were purified by anion-exchange HPLC followed by desalting (SepPak cartridges). Yields: 50-100 AZSO units. Conditions for HPLC Purification: Column: Protein Pak DEAF-5PW (7.5mm ae 7.5cm, Waters), Solvents: Buffer A: H20; Buffer B: 1M
LiC104 (or 1M NaC104), Gradient: 1000 buffer A isocratic for 12 min, 100 o A - 15 o B , linear (over 5 min) , 15 o B - 55 0 B, linear (over 60 min); Flow rate was set at 1 ml/min, temperature was adjusted to 50°C. The detector was set at 260 nm for analytical and 290 nm for preparative chromatography. Under these conditions, the desired full-length oligomer eluted last. Oligonucleotides were characterized by gel electrophoresis and mass spectrometry.
Sequences of the oligonucleotides used are provided in Tables 1 and 2.

Tm measurements. AON and complementary target RNA
oligonucleotides were mixed in equimolar ratios in.140 mM
KCl, 1 mM MgCl2, and 5 mM Na2HP04 buffer, pH 7.2, to provide a total duplex concentration of ca. 5 ~,M. Samples were heated to 90°C for 15 min, then cooled slowly to room temperature. The AON/RNA duplex solution was then exposed to increasing temperature (0.5°C/measurement), and the UV
absorbance at 260 nm was determined after temperature equilibration. Tm values provided on Table 1 and 2 were calculated using the base-line method and have an uncertainty of ~ 0.5°C.
Purification of RNase H. E. coli RNase HI was purified as described previously (7). Human RNase HII was over-expressed and purified following published procedures (Wu, H. et al., J. Biol . Ch em. , 1999, 274, 28270 ) .
RNase H assay. RNase H assays were carried out at room temperature (~ 20 °C) (homopolymeric oligonucleotides shown in Table 1), or 37 °C (mixed-based oligonucleotides shown in Table 2). Homopolymeric nucleic acid duplex substrates were prepared by mixing the phosphodiester linked AON (2 pmol) with 5'-32P-labeled complementary target oligo-rAl$ RNA (0.5 pmol; SEQ ID N0: 21) in 10 ~l of 60 mM Tris-HCl (pH 7.8) containing 60 mM KC1 and 2.5 mM MgCl2, followed by heating at 90°C for 2 minutes and slow cooling to room temperature.
Duplex substrate solutions were allowed to stand at room temperature for at least 1 h prior to use. Reactions were initiated by the addition of RNase H (7 ng of enzyme in 2 ~L buffer) and aliquots were removed at various times and' quenched by the addition of an equal volume of 98o deionized formamide containing 10 mM EDTA, 1mg/mL bromophenol blue and 1 mg/mL xylene cyanol. After heating at 100°C for 5 min, reaction products were resolved by electrophoresis on 160 polyacrylamide sequencing gels containing 7 M urea, visualized by autoradiography, and product formation was quantified by densitometry.
AON/RNA hybrids of mixed base composition were prepared by mixing the phosphorothioate AON strand (see oligomers listed on Table 2) with the corresponding 5'-radiolabeled target RNA (AAG GGA UAC GAC AAG GAU AUA A [SEQ ID NO: 22]).
This RNA was 5'-end labeled with 32P using [y-32P]-ATP using T4 polynucleotide kinase. Twenty pmol (20 pmol) antisense oligonucleotides and 10 pmol 5'-32P-labeled RNA were mixed in a buffer (100 ~1 final) containing 60 mM Tris.HCl (pH
7.8), 60 mM KC1, 2.5 mM MgCl2, heated at. 90 °C for 5 minutes and slowly cooled to room temperature. To initiate reactions, human RNase H (5 ng in 2 ~1 buffer) was added to 8 ~l of the above substrate solution. After incubation at 37 °C, the reactions were terminated by adding an equal volume of denaturing loading buffer (98 o deionized formamide, 10 mM EDTA, 1 mg/mL bromophenol blue and 1 mg/mL xylene cyanol). The products were separated on a denaturing 16 0 gel (w/v) polyacrylamide/7 M urea gel in Tris-borate/EDTA
buffer at 2000 V for approximately 2 h. After electrophoresis, the gel was exposed to an X-ray film and the resulting autoradiograms were scanned and quantitated.
Luciferase assay. HeLa X1/5 cells (stably transfected with the luciferase gene and expressing a functional luciferase enzyme) were cultured in Dulbecco's modified Eagle's medium (DMEM) supplemented with 10o fetal bovine serum (FBS). Cells were seeded in 96-well plates at 2 x 104 cells/well.
Antisense experiments were carried out 24 h after seeding, by which time cells were- 80 o confluent. Lipofectin was used to deliver antisense oligonucleotides to the cells. Briefly, antisense oligonucleotides and Lipofectin were diluted with DMEM without serum to provide a 10~e final concentration of antisense and 50 ~.g/ml Lipofectin. Equal volumes of oligonucleotide and Lipofectin solutions were mixed in plastic tubes and incubated for 15 min at room temperature to allow complex formation. This complex was diluted 5-fold with DMEM containing 10o FBS, and then the cell culture medium was replaced with this mixture and cells incubated for 4 hours at 37°C. The antisense/Lipofectin mixture was removed from the cells and replaced with DMEM containing 100 FBS, and then the cells were incubated for an additional 16 hours at 37°C. After this additional 16 hours incubation, cellular luciferase activity was assessed using the luciferase assay system (Promega, Madison, WI, USA) according to the manufacturer's protocol.. Briefly, the culture medium was removed, the cells were washed with phosphate-buffered saline, and then the cells were lysed.
~0 Aliquots of the cell lysates were transferred to assay microplates, luciferin substrate solution was added, and the resulting luminescence was immediately measured using a SPECTRAmax GEMINI XS microplate spectrofluorometer (Molecular Devices, Sunnyvale, CA, USA) set at luminescence reading mode.
Example 2: Stability of altimer:RNA duplexes Applicants demonstrate herein that ANA/DNA (e. g. FANA/DNA) "altimer" oligonucleotides form duplexes with target RNA
(Tables 1 & 2), and that the melting temperature for these AON chimeras directly correlates with the FANA content.
Previous studies have shown that 2'-OMe RNA AON also bind to target RNA with a higher affinity than do the corresponding DNA AON. However, mixed backbone 2'-OMe RNA/DNA AON (SEQ ID
Nos: 8 - 10) showed only similar or lower thermal binding affinity for target RNA compared to the all DNA A0N (SEQ ID
5 N0: 1) .
Table 1. Altimer AONs and their duplex formation with target octadecariboadenylic acid (r-A18).
AON NO. or AON Tm Sequences SEQ ID NO: (C) 1 5' -TTT TTT TTT TTT TTT TTT-3' 40 2 5' -FFF FFF FFF FFF FFF FFF-3' 53 3 5' -FTF TFT FTF TFT FTF TFT-3' 45.5 4 5' -FFT TFF TTF FTT FFT TFF-3' 46 5 5' -FFF TTT FFF TTT FFF TTT-3' 47 6 5' -FFF FTT TFF FFT TTF FFF-3' 47 7 5' -FFF FFF TTT TTT FFF FFF-3' 48 8 5' -UTU TUT UTU TUT UTU TUT-3' 33 9 5' -UUU TTT UUU TTT UUU TTT-3' 42 10 5' -UUU UUU TTT TTT UUU UUU-3' 41 a T, F, and U refer, respectively, to the natural 2' deoxyribothymidine nucleotide, 2'-deoxy-2'-fluoro-D-arabinothymidine nucleotide, and 2'-0-methyl-D-uridine nucleotide. Tm is the melting temperature of the AON/RNA
duplex, which is defined as the temperature at which half the population (500) of molecules are duplexed (AON/RNA), and the remainder being single stranded (AON + RNA). Thus Tm values are an indication of the stability of the AON/RNA
duplex.

Example 3: Ability of AON of the invention to elicit RNase H degradation of target RNA
Studies with mixed backbone AON suggest that the ability of these AON to elicit RNase H degradation of the target RNA in vitro is predictive of the ability of these AON to inhibit intracellular gene expression (Monia, B.P. et al. J. Biol.
Chem. 1993, 268, 14514; Gutierrez, A.J. et al., Biochemistry 1997, 36, 743; Flanagan, W.M. et al. , Proc. Natl. Acad. Sci.
U.S.A 1999, 96, 3513). Applicants therefore evaluated duplexes of the various AON listed in Table 1 bound to complementary RNA as substrates for E. coli RNase HI and human RNase HII. Figure 2 shows that all FANA/DNA chimeras induced target RNA cleavage by human RNase HII. RNase H
cleavage efficiency increased as the size of the alternating DNA segments within the FANA background was increased.
Optimal activity was noted with SEQ ID N0: 5, which comprises alternating trinucleotide segments of FANA and DNA. The ability of this "altimer" AON to elicit human RNase HII degradation of target RNA was significantly better than that of the equivalent all-DNA SEQ ID N0: 1.
Furthermore, this characteristic of SEQ ID N0: 5 was improved relative to the FANA /DNA/FANA SEQ ID N0: 7 (Figure 3) .
Unlike "altimer" AON comprised of FANA and DNA, similar AON (SEQ ID NOs: 8 and 9) comprised of 2'-O-methyl RNA and DNA showed only poor ability to elicit RNase H degradation of target RNA (Figure 3).
Example 4: Effect of antisense oligonucleotides on luciferase expression.

Various oligonucleotides were prepared and characterized for binding to a luciferase-encoding target RNA, and assayed for their effect on luciferase expression, as described above. Results are presented in Table 2. With the exception of the "scrambled" and "mismatch" controls shown below, all oligonucleotides comprising FANA/DNA
alternating segments exhibited significant inhibition of luciferase activity. While such inhibition was greatest with an oligonucleotide of 3-nucleotide alternating segments (SEQ ID N0: 12), it was also observed in the cases where flanking 2'-methoxy RNA nucleotides were added to a FANA/DNA
alternating oligonucleotide (e. g. SEQ ID NOs: 15 and 16).
Oligonucleotides comprising FANA/DNA alternating segments were superior in this regard as compared to a pure DNA
oligonucleotide (SEQ ID N0: 11) or a 2'-methoxy RNA - DNA -2'-methoxy RNA "gapmer" oligonucleotide (SEQ ID N0: 20) which only exhibited very marginal levels of inhibition as compared to non-oligonucleotide controls.

Table 2: Table 2: Physical and Biological Properties of AON
Oligonucleotides SEQ AON Sequences Tm b Lucife-ID o0 krei rase NO: Activi-tYc (~) 11 Ata-tcc-ttg-tcg-tat-ccc 57 3.4 80 12 ATA-tcc-TTG-tcg-TAT-ccc 62 4.2 21 13 ATATCCTT-gtcgtatccc 61 2.9 60 14 TA get CCA ca CTA ga CC n.a. n.d. 102 (scrambled altimer control) 15 [ 2' oMe-AUAU ] -cc-TT-gt-CG-to [ 2' 6 6 3 . 5 7 oMe-UCCC ] 3 16 [ 2' oMe-AUAU ] -CCT-tgt-CG-ta- [ 2' 6 6 3 . 4 2 oMe-UCCC ] 3 17 [ 2' oMe-AUAU ] -CCTTG-tcgta- [ 2' 6 5 3 . 7 6 ome-UCCC ] 8 18 [ 2' oMe-AUAU ] -CCTTGTCGTA- [ 2' oMe-UCCC6 8 0 . 5 3 ] 3 19 [a' oMe-AUAA] -cct-tTt-cTt-A- [2'oMe-ACCC]n. n. 98 (4 by mismatch control) a. d.

20 [2'oMe-AUAU]-ccttgtcgta-[2'oMe-UCCC] 64 3.6 82 "Lower case letters, uivH; upper case noia le~~er5, rruvH;
Upper case letters in square brackets, 2'-OMe-RNA. All AONs are phosphorothioates (all PS linkages). bPseudo-first rate constants for RNase-HII mediated hydrolysis of target RNA
when duplexed to AON. °The column "luciferase activity (%)"
gives luciferase activity expressed as percent relative to luciferase activity in the absence of AON. Concentration of AON was 250 nM: N.a.=not applicable; n.d.= not determined.

SEQUENCE LISTING
<110> MCGILL UNIVERSITY ET AL.
<120> OLIGONUCLEOTIDES COMPRISING ALTERNATING SEGMENTS AND USES THEREOF
<130> 85827-63 <150> US 60/352,873 1 0 <151> 2002-02-01 <160> 22 <170> Patentln version 3.1 <210> 1 <211> 18 <212> DNA
<213> Artificial <220>
. <223> Oligonucleotide <400> 1 2 5 tttttttttt tttttttt. 1g <210> 2 <211> 18 ~ <212> DNA
<213> Artificial <220>
<223> ~0ligonucleotide <220>
<221> misc_feature ' <222> (1)...(18) <223> Residues 1 to 18 are 2'-deoxy-2'-fluoroarabinothymidine <400> 2 nnnnnnnnnn nnnnnnnn 1g <210> 3 <211> 18 <212> DNA
<213> Artificial <220>
<223> Oligonucleotide <220>
5 5 <221> misc_feature <222> (1)..(17) <223> Residues 1, 3, 5, 7, 9, 11, 13, 15, 17 are 2'-deoxy-2'-fluoroarabinothymidine <220>
<221> misc_feature <222> (1)..(17) <223> Residues 1, 3, 5, 7; 9, 11, 13, 15 and 17 are 2'-deoxy-2'-fluoroarabinothymidine <400> 3 ntntntntnt ntntntnt 1g <210> 4 <211> 18 <212> DNA
<213> Artificial ~
<220>
<223> Oligonucleotide <220>
<221> misc_feature <222> (1). (18) <223> Residues 1, 2, 5, 6, 9, 10, 13, 14, 17 and 18 are 2'-deoxy-2'-~ 5 fluoroarabinothymidine <400> 4 nnttnnttnn ttnnttnn 18 <210> 5 <211> 18 <212> DNA
<213> Artificial <220>
<223> 0ligonucleotide <220>
<221> misc_feature <222> (1). (15) <223> Residues 1-3, 7-9 and 13-15 are 2'-deoxy-2'-fluoroarabinothymidine <400> 5 nnntttnnnt ttnnnttt 18 <210> 6 <211> 18 <212> DNA
<213> Artificial <220>
<223> Oligonucleotide 3/~
<220>
<221> misc_featu,re <222> (1)..(18) <223> Residues 1-4, 8-11 and 15-18 are 2'-deoxy-2'-fluoroarabinothymidine <400> 6 nnnntttnnn ntttnnnn 18 <210> 7 <211> 18 <212> DNA
<213> Artificial <220>
<223> Oligonucleotide <220>
<221> misc_feature <222> (1). (18) <223> Residues 1-6 and 13-18 are 2'-deoxy-2'-fluoroarabinothymidine <400> 7 nnnnnntttt ttnnnnnn 1g <210> 8 <211> 18 <212> DNA
<213> Artificial <220>
<223> 0ligonucleotide <220>
<221> misc_feature 4 0 <222> (1)..(17) <223> Residues 1, 3, 5, 7, 9, 11, 13, 15 and 17 are 2'-0-methyl-D-uridine <400> 8 ututututut utututut 18 <210> 9 <211> 18 <212> DNA
<213> Artificial <220>
<223> 0ligonucleotide <220>
<221> misc_feature <222> (1). (15) <223> Residues 1-3, 7-9, and 13-15 are 2'-0-methyl-D-uridine, <400> 9 uuutttuuut ttuuuttt 1g <210> 10 <211> 18 <212> DNA
<213> Artificial <220>
<223> Oligonucleotide <220>
2 0 <221> misc_feature <222> (1) . (18) <223> Residues 1-6 and 13-18 are 2'-0-methyl-D-uridine <400> 10 uuuuuutttt ttuuuuuu 18 <210> 11 <211> 18 <212> DNA
<213> Artificial <220>
<223> Oligonucleotide <220>
<221> misc_feature <222> (1)..(18) '40 <223> Residues 1-18 are linked by phosphorothioate linkages <400> 11 atatccttgt cgtatccc 1g <210> 12 <211> 18 <212> DNA
<213> Artificial <220>
<223> Oligonucleotide <220>
<221> misc_feature <222> (1)..(15) <223> Residues 2, 7, 8, 13 and 15 are 2'-deoxy-2'-fluoroarabinothymidine; residues 1, 3 and 14 are 2'-deoxy-2'-fluoroarabinoadenosine; residue 9 is 2'-deoxy-2'-fluoroarabinoguanosine <220>
<221> misc_feature <222> (1)..(18) <223> Residues 1-18 are linked by phosphorothioate linkages <400> 12 nnntccnnnt cgnnnccc lg ' <210> 13 <211> 18 <212> DNA
<213> Artificial <220>
<223> Oligonucleotide <220>
2 0 <221> misc_feature <222> (1). (8) <223> Residues 1 and 3 are 2'-deoxy-2'-fluoroarabinothymidine; residues 2, 4, 7 and 8 are 2'-deoxy-2'-fluoroarabinoadenosine; residues 5 and 6 are 2'-deoxy-2'-fluoroarabinocytidine <220>
<221> misc_feature <222> (1). (18) <223> Residues 1-18 are linked by phosphorothioate linkages <400> 13 nnnnnnnngt cgtatccc 1g <210> 14 <211> 17 <2l2> DNA
<213> Artificial <220>
<223> Oligonucleotide <220>
<221> misc_feature 4 5 <222> (1)..(17) <223> Residues 2, 8 and 13 are 2'-deoxy-2'-fluoroarabinothymidine;
residues 3 and 5 are 2'-deoxy-2'-fluoroarabinoadenosine; residues 12 and 16-18 are 2'-deoxy-2'-fluoroarabinocytidine <220>
<221> misc_feature <222> (1)..(17) <223> Residues 1-17 are linked by phosphorothioate linkages <400> 14 nngctnnnca nnngann 17 <210> 15 <211> 18 <212> DNA
<213> Artificial <220>
<223> Oligonucleotide <220>
<221> misc_feature <222> (1)..(18) <223> Residues 1 and 3 are 2'-0-methyl-D-adenosine; residues 2, 4 and l5,are 2'-0-methyl-D-uridine; residues 16-18 are 2'-O-methyl-D-cytidine;
residues 7 and 8 are 2'-deoxy-2'-fluoroarabinothymidine; residue 11 is 2'-deoxy-2'-fluoroarabinocytidine; residue 12 is 2'-deoxy-2'-fluoroarabinoguanosine <220>
<221> misc_feature 2 0 <222> (1)..(18) <223> Residues 1-18 are linked by phosphorothioate linkages <400> 15 auauccnngt nntauccc 18 <210> 16 <211> 18 <212> DNA
3 0 <213> Artificial <220>
<223> Oligonucleotide <220>
<221> misc_feature <222> (1). (18) .
<223> Residues 1 and 3 are 2'-0-methyl-D-adenosine; residues 2, 4 and 15 are 2'-O-methyl-D-uridine; residues 16-18 are 2'-O-methyl-D-cytidine;
4 0 residue 7 is 2'-deoxy-2'-fluoroarabinothymidine; residues 5, 6 and 11 are 2'-deoxy-2'-fluoroarabinocytidine; residue 12 is 2'-deoxy-2'-fluoroarabinoguanosine <220>
<221> misc_feature <222> (1)..(18) <223> Residues 1-18 are linked by phosphorothioate linkages <400> 16 5 0 auaunnntgt nntauccc , 18 <210> 17 <211> 18 <212> DNA
<213> Artificial ~l8 <220>
<223> Oligonucleotide <220>
<221> misc_feature <222> (1). (18) <223> Residues 1 and 3 are 2'-0-methyl-D-adenosine;. residues 2, 4 and are 2'-0-methyl-D-uridine; residues 16-18 are 2'-O-methyl-D-cytidine;
residues 7 and 8 are 2'-deoxy-2'-fluoroarabinothymidine; residues 5 and 10 6 are 2'-deoxy-2'-fluoroarabinocytidine; residue 9 is 2'-deoxy-2'-fluoroarabinoguanosine <220>
<22l> misc_feature 15 <222> (1) . (18) <223> Residues 1-18 are linked by phosphorothioate linkages <400> 17 auaunnnnnt cgtauccc 1g 20.
<210> 18 <211> 18 <212> DNA
2 5 <213> Artificial <220>
<223> Oligonucleotide
30 <220>
<221> misc_feature <222> (1) . (18~) <223> Residues 1 and 3 are 2'-0-methyl-D-adenosine; residues 2, 4 and 15 are 2'-0-methyl-D-uridine; residues 16-18 are 2'-0-methyl-D-cytidine;
35 residues 7, 8, 10 and 13 are 2'-deoxy-2'-fluoroarabinothymidine; residue 14 is 2'-deoxy-2'-fluoroarabinoadenosine; residues~5, 6 and 11 are 2'-deoxy-2'-fluoroarabinocytidine; residues 9 and 12 are 2'-deoxy-2'-fluoroarabinoguanosine 40 <220>
<221> misc_feature <222> (1)..(18) <223> Residues 1-18 are linked by phosphorothioate linkages 45 <400> 18 auaunnnnnn nnnnuccc 1g <210> 19 50 <211> 18 <212> DNA
<213> Artificial <220>
55 <223> Oligonucleotide <220>
<221> misc_feature <222> (1). (18) <223> Residues 1, 3, 4 and 16 are 2'-0-methyl-D-adenosine; residue 2 is 2'-0-methyl-D-uridine; residues 17-19 are 2'-0-methyl-D-cytidine;
residues 9 and 12 are 2'-deoxy-2'-fluoroarabinothymidine; residue 14 is 2'-deoxy-2'-fluoroarabinoadenosine <220>
<221> misc_feature <222> (1). (18) <223> Residues 1-18 are linked by phosphorothioate linkages <400> 19 auaaccttnt cntnaccc 18 <210> 20 <211> 18 <212> DNA
<213> Artificial <220>
<223> Oligonucleotide <220>
i <221> misc_feature 2 5 <222> (1)..(18) <223> Residues 1 and 3 are 2'-0-methyl-D-adenosine; residues 2, 4 and 15 are 2'-0-methyl-D-uridine; residues 16-18 are 2'-0-methyl-D-cytidine <220>
3 0 <221> misc_feature <222> (1)..(18) <223> Residues 1-18 are linked by phosphorothioate linkages <400> 20 3 5 auauccttgt cgtauccc~ 1g <210> 21 <211> 18 4 0 < 212 > RNA
<213> Artificial <220>
<223> Target RNA oligonucleotide <400> 21 aaaaaaaaaa aaaaaaaa 18 <210> 22 <211> 22 <212> RNA
<213> Artificial <220>
<223> Target RNA oligonucleotide <400> 22 aagggauacg acaaggauau as 22

Claims (61)

1. An oligonucleoside comprising alternating first and second segments, wherein said first segment comprises at least one sugar-modified nucleoside, wherein said second segment comprises at least one 2'-deoxynucleoside, wherein said oligonucleoside comprises at least 2 of each of said first and second segments thereby comprising at least 4 alternating segments.
2. The oligonucleoside of claim 1, wherein (a) said oligonucleoside further comprises an internucleoside linkage comprising a phosphate, thereby being an oligonucleotide, (b)wherein said sugar-modified nucleoside comprises an attached phosphate thereby being a sugar-modified nucleotide, (c) wherein said 2'-deoxynucleoside comprises an attached phosphate thereby being a 2'-deoxynucleotide, or (d) any combination of (a) to (c).
3. The oligonucleotide of claim 2, wherein said sugar modified oligonucleotide is capable of adopting a DNA-like conformation.
4. The oligonucleotide of claim 2, wherein said sugar-modified nucleotide is selected from the group consisting of arabinonucleotides, alpha-L-locked nucleic acids, cyclohexene nucleic acids, and ribonucleotides lacking an electronegative 2'-oxygen atom.
5. The oligonucleotide of claim 3, wherein the ribonucleotides lacking an electronegative 2'-oxygen atom are selected from the group consisting of 2'-alkyl-D-ribose and 2'-SCH3-D-ribose.
6. The oligonucleotide of claim 2, wherein said segments each independently comprise about 1 to about 6 sugar-modified nucleotides or 2'-deoxynucleotides.
7. The oligonucleotide of claim 6, wherein said segments each independently comprise about 2 to about 5 sugar-modified nucleotides or 2'-deoxynucleotides.
8. The oligonucleotide of claim 7, wherein said segments each independently comprise about 3 to about 4 sugar-modified nucleotides or 2'-deoxynucleotides.
9. The oligonucleotide of claim 8, wherein said segments each independently comprise about 3 sugar-modified nucleotides or 2'-deoxynucleotides.
10. The oligonucleotide of claim 2, wherein said oligonucleotide has a structure selected from the group consisting of:
a) (A x-D y)n I
b) (D y-A x)n II
c) (A x-D y)m-A x-D y-A x III
d) (D y-A x)m-D y-A x-D y IV
wherein each of m, x and y are each independently an integer greater than or equal to 1, n is an integer greater than or equal to 2, A is a sugar-modified nucleotide and D
is a 2'-deoxyribonucleotide.
11. The oligonucleotide of claim 2, wherein said sugar-modified nucleotide comprises a 2' substituent selected from the group consisting of fluorine, hydroxyl, amino, cyano, azido, -CH=CH2, -C.ident.CH, alkyl, functionalized alkyl, alkoxy and functionalized alkoxy groups.
12. The oligonucleotide of claim 11, wherein said alkyl group is a lower alkyl group.
13. The oligonucleotide of claim 12, wherein said lower alkyl group is selected from the group consisting of methyl, ethyl and propyl groups.
14. The oligonucleotide of claim 11, wherein said functionalized alkyl group is selected from the group consisting of methylamino, ethylamino and propylamino groups.
15. The oligonucleotide of claim 11, wherein said alkoxy group is selected from the group consisting of methoxy, ethoxy and propoxy groups.
16. The oligonucleotide of claim 11, wherein said functionalized alkoxy group is -O(CH2)q-R, wherein q=2, 3 or 4 and -R is selected from the group consisting of -NH2, -OCH3, and -OCH2CH3 groups.
17. The oligonucleotide of claim 3, wherein the sugar-modified nucleotide is an arabinonucleotide.
18. The oligonucleotide of claim 17, wherein the arabinonucleotide is a 2'-fluoroarabinonucleotide (2'F-ANA).
19. The oligonucleoside of claim 1, wherein said oligonucleoside comprises one or more internucleoside linkages selected from the group consisting of:
a) phosphodiester;
b) phosphotriester;
c) phosphorothioate;
d) phosphorodithioate;
e) Rp-phosphorothioate;
f) Sp-phosphorothioate;
g) boranophosphate;
h) 3'-thioformacetal i) methylene(methylimino);
j) amide;
k) methylphosphonate;
l) phosphoramidate (5'P-N3'); and m) any combination of (a) to (l).
20. The oligonucleotide of claim 2, wherein said oligonucleotide consists of about 30 or fewer nucleotides.
21. The oligonucleotide of claim 20, wherein said oligonucleotide consists of about 8 to about 25 nucleotides.
22. The oligonucleotide of claim 21, wherein said oligonucleotide consists of about 18 nucleotides.
23. The oligonucleotide of claim 10, wherein said oligonucleotide has structure I wherein x=1, y=1 and n=9, thereby having a structure:

A-D-A-D-A-D-A-D-A-D-A-D-A-D-A-D-A-D.
24. The oligonucleotide of claim 10, wherein said oligonucleotide has structure II wherein x=1, y=1 and n=9, thereby having a structure:
D-A-D-A-D-A-D-A-D-A-D-A-D-A-D-A-D-A.
25. The oligonucleotide of claim 10, wherein said oligonucleotide has structure III wherein x=2, y=2 and m=3, thereby having a structure:
A-A-D-D-A-A-D-D-A-A-D-D-A-A-D-D-A-A.
26. The oligonucleotide of claim 10, wherein said oligonucleotide has structure IV wherein x=2, y=2 and m=3, thereby having a structure:
D-D-A-A-D-D-A-A-D-D-A-A-D-D-A-A-D-D.
27. The oligonucleotide of claim 10, wherein said oligonucleotide has structure I wherein x=3, y=3 and n=3, thereby having a structure:
A-A-A-D-D-D-A-A-A-D-D-D-A-A-A-D-D-D.
28. The oligonucleotide of claim 10, wherein said oligonucleotide has structure II wherein x=3, y=3 and n=3, thereby having a structure:
D-D-D-A-A-A-D-D-D-A-A-A-D-D-D-A-A-A.
29. The oligonucleotide of claim 10, wherein said oligonucleotide has said structure III wherein x=4, y=3 and m=1, thereby having a structure:
A-A-A-A-D-D-D-A-A-A-A-D-D-D-A-A-A-A.
30. The oligonucleotide of claim 10, wherein said oligonucleotide has said structure IV wherein x=4, y=3 and m=1, thereby having a structure:
D-D-D-D-A-A-A-D-D-D-D-A-A-A-D-D-D-D.
31. The oligonucleoside of claim 1, further comprising a third segment comprising a modified nucleoside, wherein said third segment is adjacent to (a) the 5' end of said alternating first and second segments, (b) the 3' end of said alternating first and second segments, or (c) both (a) and (b).
32. The oligonucleotide of claim 2, further comprising a third segment comprising a modified nucleotide, wherein said third segment is adjacent to (a) the 5' end of said alternating first and second segments, (b) the 3' end of said alternating first and second segments, or (c) both (a) and (b).
33. The oligonucleotide of claim 32, wherein said modified nucleotide is a modified ribonucleotide.
34. The oligonucleotide of claim 33, wherein said modified ribonucleotide.is comprises a modification at its 2' position.
35. The oligonucleotide of claim 33, wherein said 2' modification is selected from the group consisting of methoxy, methoxyethyl, fluoro and propylamino groups.
36. The oligonucleotide of claim 2, wherein said oligonucleotide is antisense to a target RNA.
37. A method of preventing or decreasing translation, reverse transcription and/or replication of a target RNA in a system, said method comprising contacting said target RNA, with the oligonucleotide of claim 36.
38. The method of claim 37, wherein said system is selected from the group consisting of a cell, tissue or subject.
39. The method of claim 38, wherein said cell, tissue or subject is a mammalian cell, tissue or subject.
40. The method of claim 38, wherein said cell, tissue or subject is a human cell, tissue or subject.
41. A method of inducing RNase H-mediated cleavage of a target RNA in a system, said method comprising contacting said target RNA with the oligonucleotide of claim 36.
42. The method of claim 41, wherein said RNase H-mediated cleavage is effected by RNase H activity associated with a reverse transcriptase of a virus.
43. The method of claim 42, wherein the virus is a human pathogenic virus.
44. The method of claim 43 wherein the human pathogenic virus is selected from the group consisting of HIV and hepadnaviruses.
45. The method of claim 44 wherein the HIV is selected from the group consisting of HIV-1 and HIV-2.
46. The method of claim 44 wherein the hepadnavirus is hepatitis B virus.
47. The method of claim 41, wherein said RNase H-mediated cleavage is effected by RNase H activity associated with an RNase H enzyme of prokaryotic or eukaryotic origin.
48. The method of claim 47, wherein the eukaryotic RNase H is a mammalian RNase H.
49. The method of claim 48, wherein the eukaryotic RNase H is a human RNase H.
50. The method of claim 49, wherein the human RNase H
is selected from the group consisting of RNase H1 and RNase H2.
51. A method of preventing or decreasing translation, reverse transcription and/or replication of a target RNA in a system, said method comprising:
a) contacting said target RNA with the oligonucleotide of claim 36; and b) allowing RNase H cleavage of said target RNA.
52. A method of effecting a process selected from the group consisting of:

(a) inducing RNase H-mediated cleavage of a target RNA
in a system;
(b) preventing or decreasing translation of a target RNA in a system;
(c) preventing or decreasing reverse transcription of a target RNA in a system;
(d) preventing or decreasing replication of a target RNA in a system (e) detecting the presence of a target RNA in a system (f) validating a gene target corresponding to a target RNA in a system;
(g) preventing or treating a disease related to a target RNA in a system; and (h) any combination of (a) to (g);
said method comprising contacting said target RNA with the oligonucleotide of claim 36.
53. A method of effecting a process selected from the group consisting of:
(a) inducing RNase H-mediated cleavage of a target RNA
in a system;
(b) preventing or decreasing translation of a target RNA in a system;
(c) preventing or decreasing reverse transcription of a target RNA in a system;
(d) preventing or decreasing replication of a target RNA in a system (e) detecting the presence of a target RNA in a system (f) validating a gene target corresponding to a target RNA in a system;
(g) preventing or treating a disease related to a target RNA in a system; and (h) any combination of (a) to (g);

said method comprising introducing the oligonucleotide of claim 36 into said system.
54. Use of the oligonucleotide according to claim 36 for a medical or research use.
55. The use of claim 54, wherein said medical or research use is selected from the group consisting of:
(a) inducing RNase H-mediated cleavage of a target RNA
in a system;
(b) preventing or decreasing translation of a target RNA in a system;
(c) preventing or decreasing reverse transcription of a target RNA in a system;
(d) preventing or decreasing replication of a target RNA in a system (e) detecting the presence of a target RNA in a system (f) validating a gene target in a system;
(g) preventing or treating a disease related to a target RNA in a system; and (h) any combination of (a) to (g).
56. Use of the oligonucleotide according to claim 36 for the preparation of a medicament.
57. The use of claim 56 wherein said medicament is for a use selected from the group consisting of:
(a) inducing RNase H-mediated cleavage of a target RNA
in a system;
(b) preventing or decreasing translation of a target RNA in a system;
(c) preventing or decreasing reverse transcription of a target RNA in a system;

(d) preventing or decreasing replication of a target RNA in a system;
(e) detecting the presence of a target RNA in a system;
(f) validating a gene target in a system;
(g) preventing or treating a disease related to a target RNA in a system; and (h) any combination of (a) to (g).
58. A composition comprising the oligonucleotide according to claim 36 in admixture with a pharmaceutically acceptable carrier.
59. The composition of claim 58, wherein said composition is for a use selected from the group consisting of:
(a) inducing RNase H-mediated cleavage of a target RNA
in a system;
(b) preventing or decreasing translation of a target RNA in a system;
(c) preventing or decreasing reverse transcription of a target RNA in a system;
(d) preventing or decreasing replication of a target RNA in a system;
(e) detecting the presence of a target RNA in a system;
(f) validating a gene target in a system;
(g) preventing or treating a disease related to a target RNA in a system; and (h) any combination of (a) to (g).
60. A commercial package comprising the oligonucleotide according to claim 36 together with instructions for its use.
61. The commercial package of claim 60, wherein said instructions are for a use selected from the group consisting of:
(a) inducing RNase H-mediated cleavage of a target RNA
in a system;
(b) preventing or decreasing translation of a target RNA in a system;
(c) preventing or decreasing reverse transcription of a target RNA in a system;
(d) preventing or decreasing replication of a target RNA in a system;
(e) detecting the presence of a target RNA in a system;
(f) validating a gene target in a system;
(g) preventing or treating a disease related to a target RNA in a system; and (h) any combination of (a) to (g).
CA2474414A 2002-02-01 2003-01-31 Oligonucleotides comprising alternating segments and uses thereof Expired - Lifetime CA2474414C (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US35287302P 2002-02-01 2002-02-01
US60/352,873 2002-02-01
PCT/CA2003/000129 WO2003064441A2 (en) 2002-02-01 2003-01-31 Oligonucleotides comprising alternating segments and uses thereof

Publications (2)

Publication Number Publication Date
CA2474414A1 true CA2474414A1 (en) 2003-08-07
CA2474414C CA2474414C (en) 2018-03-06

Family

ID=27663144

Family Applications (1)

Application Number Title Priority Date Filing Date
CA2474414A Expired - Lifetime CA2474414C (en) 2002-02-01 2003-01-31 Oligonucleotides comprising alternating segments and uses thereof

Country Status (13)

Country Link
US (3) US8278103B2 (en)
EP (1) EP1470144B1 (en)
JP (1) JP2005522997A (en)
AT (1) ATE416183T1 (en)
AU (1) AU2003202376B2 (en)
CA (1) CA2474414C (en)
DE (1) DE60325016D1 (en)
DK (1) DK1470144T3 (en)
ES (1) ES2318106T3 (en)
MX (1) MXPA04007403A (en)
NZ (1) NZ534396A (en)
PT (1) PT1470144E (en)
WO (1) WO2003064441A2 (en)

Families Citing this family (41)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5898031A (en) * 1996-06-06 1999-04-27 Isis Pharmaceuticals, Inc. Oligoribonucleotides for cleaving RNA
US7812149B2 (en) 1996-06-06 2010-10-12 Isis Pharmaceuticals, Inc. 2′-Fluoro substituted oligomeric compounds and compositions for use in gene modulations
US9096636B2 (en) * 1996-06-06 2015-08-04 Isis Pharmaceuticals, Inc. Chimeric oligomeric compounds and their use in gene modulation
US20050042647A1 (en) * 1996-06-06 2005-02-24 Baker Brenda F. Phosphorous-linked oligomeric compounds and their use in gene modulation
ATE346918T1 (en) * 1998-06-19 2006-12-15 Univ Mcgill ANTISENSE OLIGONUCLEOTIDES BASED ON BETA-ARABINOSE AND ITS ANALOGUES
US8258288B2 (en) * 2002-02-20 2012-09-04 Sirna Therapeutics, Inc. RNA interference mediated inhibition of respiratory syncytial virus (RSV) expression using short interfering nucleic acid (siNA)
US20090137510A1 (en) * 2002-02-20 2009-05-28 Sirna Therapeutics, Inc. RNA INTERFERENCE MEDIATED INHIBITION OF NF-KAPPA B/ REL-A GENE EXPRESSION USING SHORT INTERFERING NUCLEIC ACID (siNA)
EP1857547B2 (en) 2002-08-05 2020-12-02 Silence Therapeutics GmbH Further novel forms of interfering RNA molecules
AU2003273336A1 (en) * 2002-09-18 2004-04-08 Isis Pharmaceuticals, Inc. Efficient reduction of target rna's by single- and double-stranded oligomeric compounds
CA2504929C (en) * 2002-11-05 2014-07-22 Charles Allerson Compositions comprising alternating 2'-modified nucleosides for use in gene modulation
US9150605B2 (en) * 2002-11-05 2015-10-06 Isis Pharmaceuticals, Inc. Compositions comprising alternating 2′-modified nucleosides for use in gene modulation
US9150606B2 (en) * 2002-11-05 2015-10-06 Isis Pharmaceuticals, Inc. Compositions comprising alternating 2'-modified nucleosides for use in gene modulation
US8569474B2 (en) 2004-03-09 2013-10-29 Isis Pharmaceuticals, Inc. Double stranded constructs comprising one or more short strands hybridized to a longer strand
CA2569419A1 (en) * 2004-06-03 2005-12-22 Isis Pharmaceuticals, Inc. Double strand compositions comprising differentially modified strands for use in gene modulation
US8394947B2 (en) 2004-06-03 2013-03-12 Isis Pharmaceuticals, Inc. Positionally modified siRNA constructs
US7884086B2 (en) 2004-09-08 2011-02-08 Isis Pharmaceuticals, Inc. Conjugates for use in hepatocyte free uptake assays
JP5314283B2 (en) 2004-10-29 2013-10-16 トピジェン・ファーマシューティカルズ・インコーポレーテッド Antisense oligonucleotides for treating allergy and neoplastic cell proliferation
EP1937312B1 (en) * 2005-08-30 2016-06-29 Ionis Pharmaceuticals, Inc. Chimeric oligomeric compounds for modulation of splicing
EP2392645A1 (en) * 2005-10-14 2011-12-07 MUSC Foundation For Research Development Targeting PAX2 for the induction of DEFB1-mediated tumor immunity and cancer therapy
WO2007134451A1 (en) 2006-05-19 2007-11-29 Topigen Pharmaceuticals Inc. Oligonucleotides affecting expression of phosphodiesterases
CA2715289C (en) * 2008-02-11 2019-12-24 Rxi Pharmaceuticals Corporation Modified rnai polynucleotides and uses thereof
WO2009137912A1 (en) 2008-05-15 2009-11-19 Topigen Pharmaceuticals Inc. Oligonucleotides for treating inflammation and neoplastic cell proliferation
US8815818B2 (en) 2008-07-18 2014-08-26 Rxi Pharmaceuticals Corporation Phagocytic cell delivery of RNAI
CN108165548B (en) 2008-09-22 2022-10-14 菲奥医药公司 Reduced size self-delivering RNAi compounds
US9074211B2 (en) 2008-11-19 2015-07-07 Rxi Pharmaceuticals Corporation Inhibition of MAP4K4 through RNAI
WO2010078536A1 (en) 2009-01-05 2010-07-08 Rxi Pharmaceuticals Corporation Inhibition of pcsk9 through rnai
WO2010090762A1 (en) 2009-02-04 2010-08-12 Rxi Pharmaceuticals Corporation Rna duplexes with single stranded phosphorothioate nucleotide regions for additional functionality
KR20180044433A (en) 2010-03-24 2018-05-02 알엑스아이 파마슈티칼스 코포레이션 Rna interference in dermal and fibrotic indications
WO2011119852A1 (en) 2010-03-24 2011-09-29 Rxi Pharmaceuticals Corporation Reduced size self-delivering rnai compounds
EP2550001B1 (en) 2010-03-24 2019-05-22 Phio Pharmaceuticals Corp. Rna interference in ocular indications
EP4269584A3 (en) * 2011-08-11 2024-03-27 Ionis Pharmaceuticals, Inc. Selective antisense compounds and uses thereof
CN106061488B (en) 2013-12-02 2021-04-09 菲奥医药公司 Immunotherapy of cancer
EP3137119B1 (en) 2014-04-28 2020-07-01 Phio Pharmaceuticals Corp. Methods for treating cancer using a nucleic acid targeting mdm2
EP3188799B1 (en) 2014-09-05 2022-07-06 Phio Pharmaceuticals Corp. Methods for treating aging and skin disorders using nucleic acids targeting tyr or mmp1
WO2017007825A1 (en) 2015-07-06 2017-01-12 Rxi Pharmaceuticals Corporation Methods for treating neurological disorders using a synergistic small molecule and nucleic acids therapeutic approach
WO2017007813A1 (en) 2015-07-06 2017-01-12 Rxi Pharmaceuticals Corporation Nucleic acid molecules targeting superoxide dismutase 1 (sod1)
WO2017070151A1 (en) 2015-10-19 2017-04-27 Rxi Pharmaceuticals Corporation Reduced size self-delivering nucleic acid compounds targeting long non-coding rna
US20200030361A1 (en) * 2016-09-23 2020-01-30 City Of Hope Oligonucleotides containing 2'-deoxy-2'fluoro-beta-d-arabinose nucleic acid (2'-fana) for treatment and diagnosis of retroviral diseases
US20210040481A1 (en) * 2018-03-19 2021-02-11 The Trustees Of The University Of Pennsylvania 2'f-ana-let7 mediated utrophin upregulation for dmd therapy
US11254945B2 (en) * 2018-07-06 2022-02-22 The United States Of America, As Represented By The Secretary Of Agriculture Targeted control of pests and pathogens by plant delivery of 2'F-ANA-oligonucleotides
WO2020069044A1 (en) * 2018-09-26 2020-04-02 AUM LifeTech, Inc. 2' fana modified foxp3 antisense oligonucleotides and methods of use thereof

Family Cites Families (18)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4806463A (en) * 1986-05-23 1989-02-21 Worcester Foundation For Experimental Biology Inhibition of HTLV-III by exogenous oligonucleotides
US5194428A (en) * 1986-05-23 1993-03-16 Worcester Foundation For Experimental Biology Inhibition of influenza virus replication by oligonucleotide phosphorothioates
US5264423A (en) * 1987-03-25 1993-11-23 The United States Of America As Represented By The Department Of Health And Human Services Inhibitors for replication of retroviruses and for the expression of oncogene products
US5276019A (en) * 1987-03-25 1994-01-04 The United States Of America As Represented By The Department Of Health And Human Services Inhibitors for replication of retroviruses and for the expression of oncogene products
US5004810A (en) * 1988-09-30 1991-04-02 Schering Corporation Antiviral oligomers
US5098890A (en) * 1988-11-07 1992-03-24 Temple University-Of The Commonwealth System Of Higher Education Antisence oligonucleotides to c-myb proto-oncogene and uses thereof
US5087617A (en) * 1989-02-15 1992-02-11 Board Of Regents, The University Of Texas System Methods and compositions for treatment of cancer using oligonucleotides
US5955589A (en) * 1991-12-24 1999-09-21 Isis Pharmaceuticals Inc. Gapped 2' modified oligonucleotides
US5166195A (en) * 1990-05-11 1992-11-24 Isis Pharmaceuticals, Inc. Antisense inhibitors of the human immunodeficiency virus phosphorothioate oligonucleotides
US5135917A (en) * 1990-07-12 1992-08-04 Nova Pharmaceutical Corporation Interleukin receptor expression inhibiting antisense oligonucleotides
WO1994008003A1 (en) * 1991-06-14 1994-04-14 Isis Pharmaceuticals, Inc. ANTISENSE OLIGONUCLEOTIDE INHIBITION OF THE ras GENE
US5539082A (en) * 1993-04-26 1996-07-23 Nielsen; Peter E. Peptide nucleic acids
US5985558A (en) * 1997-04-14 1999-11-16 Isis Pharmaceuticals Inc. Antisense oligonucleotide compositions and methods for the inibition of c-Jun and c-Fos
AUPM516994A0 (en) * 1994-04-20 1994-05-12 Gene Shears Pty. Limited An in vivo gene expression system
US20010049436A1 (en) 1998-04-01 2001-12-06 Wen-Qiang Zhou Mixed-backbone oligonucleotides containing pops blocks to obtain reduced phosphorothioate content
ATE346918T1 (en) * 1998-06-19 2006-12-15 Univ Mcgill ANTISENSE OLIGONUCLEOTIDES BASED ON BETA-ARABINOSE AND ITS ANALOGUES
US6277967B1 (en) * 1998-07-14 2001-08-21 Isis Pharmaceuticals, Inc. Carbohydrate or 2′-modified oligonucleotides having alternating internucleoside linkages
WO2002020773A2 (en) 2000-09-06 2002-03-14 Mcgill University Chimeric antisense oligonucleotides of arabinofuranose analogues and deoxyribose nucleotides

Also Published As

Publication number Publication date
EP1470144A2 (en) 2004-10-27
NZ534396A (en) 2006-11-30
US9902953B2 (en) 2018-02-27
MXPA04007403A (en) 2005-07-01
ES2318106T3 (en) 2009-05-01
AU2003202376B2 (en) 2008-10-30
PT1470144E (en) 2009-02-10
WO2003064441A3 (en) 2003-10-02
DE60325016D1 (en) 2009-01-15
US20130203977A1 (en) 2013-08-08
WO2003064441A2 (en) 2003-08-07
DK1470144T3 (en) 2009-04-06
CA2474414C (en) 2018-03-06
ATE416183T1 (en) 2008-12-15
US8278103B2 (en) 2012-10-02
US20050142535A1 (en) 2005-06-30
US20150368643A1 (en) 2015-12-24
JP2005522997A (en) 2005-08-04
EP1470144B1 (en) 2008-12-03

Similar Documents

Publication Publication Date Title
CA2474414C (en) Oligonucleotides comprising alternating segments and uses thereof
AU2003202376A1 (en) Oligonucleotides comprising alternating segments and uses thereof
Shea et al. Synthesis, hybridization properties and antiviral activity of lipid-oligodeoxynucleotide conjugates
US6060456A (en) Chimeric oligonucleoside compounds
US6028188A (en) Synthetic oligomers having chirally pure phosphonate internucleosidyl linkages mixed with non-phosphonate internucleosidyl linkages
US6015886A (en) Oligonucleotide phosphate esters
US5986083A (en) Synthetic oligomers having phosphonate internucleosidyl linkages of undefined chirality mixed with non-phosphonate internucleosidyl linkages
US20090105467A1 (en) Antisense oligonucleotide constructs based on beta-arabinofuranose and its analogues
CA2223103A1 (en) Oligonucleotides having phosphorothioate linkages of high chiral purity
CA2465129A1 (en) Acyclic linker-containing oligonucleotides and uses thereof
AU689182B2 (en) Chimeric oligonucleoside compounds
US8178348B2 (en) Chimeric antisense oligonucleotides of arabinofuranose analogue and deoxyribose nucleotides
AU2001289448A1 (en) Chimeric antisense oligonucleotides of arabinofuranose analogues and deoxyribose nucleotides
CA2331333C (en) Antisense oligonucleotide constructs based on .beta.-arabinofuranose and its analogues

Legal Events

Date Code Title Description
EEER Examination request
MKEX Expiry

Effective date: 20230131