RNA interference mediated inhibition of hepatitis C virus (HCV) expression using short interfering nucleic acid (siNA)

Information

  • Patent Application
  • 20050209180
  • Publication Number
    20050209180
  • Date Filed
    September 15, 2004
    20 years ago
  • Date Published
    September 22, 2005
    19 years ago
Abstract
This invention relates to compounds, compositions, and methods useful for modulating HCV gene expression using short interfering nucleic acid (siNA) molecules. This invention also relates to compounds, compositions, and methods useful for modulating the expression and activity of other genes involved in pathways of HCV gene expression and/or activity by RNA interference (RNAi) using small nucleic acid molecules. In particular, the instant invention features small nucleic acid molecules, such as short interfering nucleic acid (siNA), short interfering RNA (siRNA), double-stranded RNA (dsRNA), micro-RNA (miRNA), and short hairpin RNA (shRNA) molecules and methods used to modulate the expression of HCV genes.
Description
FIELD OF THE INVENTION

The present invention relates to compounds, compositions, and methods for the study, diagnosis, and treatment of traits, diseases and conditions that respond to the modulation of hepatitis C virus (HCV) gene expression and/or activity. The present invention is also directed to compounds, compositions, and methods relating to traits, diseases and conditions that respond to the modulation of expression and/or activity of genes involved in hepatitis C virus (HCV) gene expression pathways or other cellular processes that mediate the maintenance or development of such traits, diseases and conditions. Specifically, the invention relates to small nucleic acid molecules, such as short interfering nucleic acid (siNA), short interfering RNA (siRNA), double-stranded RNA (dsRNA), micro-RNA (miRNA), and short hairpin RNA (shRNA) molecules capable of mediating RNA interference (RNAi) against hepatitis C virus (HCV) gene expression. Such small nucleic acid molecules are useful, for example, in providing compositions to prevent, inhibit, or reduce HCV infection, liver failure, hepatocellular carcinoma, cirrhosis, and/or other disease states associated with HCV infection in a subject or organism.


BACKGROUND OF THE INVENTION

The following is a discussion of relevant art pertaining to RNAi. The discussion is provided only for understanding of the invention that follows. The summary is not an admission that any of the work described below is prior art to the claimed invention.


RNA interference refers to the process of sequence-specific post-transcriptional gene silencing in animals mediated by short interfering RNAs (siRNAs) (Zamore et al., 2000, Cell, 101, 25-33; Fire et al., 1998, Nature, 391, 806; Hamilton et al., 1999, Science, 286, 950-951; Lin et al., 1999, Nature, 402, 128-129; Sharp, 1999, Genes & Dev., 13:139-141; and Strauss, 1999, Science, 286, 886). The corresponding process in plants (Heifetz et al., International PCT Publication No. WO 99/61631) is commonly referred to as post-transcriptional gene silencing or RNA silencing and is also referred to as quelling in fungi. The process of post-transcriptional gene silencing is thought to be an evolutionarily-conserved cellular defense mechanism used to prevent the expression of foreign genes and is commonly shared by diverse flora and phyla (Fire et al., 1999, Trends Genet., 15, 358). Such protection from foreign gene expression may have evolved in response to the production of double-stranded RNAs (dsRNAs) derived from viral infection or from the random integration of transposon elements into a host genome via a cellular response that specifically destroys homologous single-stranded RNA or viral genomic RNA. The presence of dsRNA in cells triggers the RNAi response through a mechanism that has yet to be fully characterized. This mechanism appears to be different from other known mechanisms involving double stranded RNA-specific ribonucleases, such as the interferon response that results from dsRNA-mediated activation of protein kinase PKR and 2′,5′-oligoadenylate synthetase resulting in non-specific cleavage of mRNA by ribonuclease L (see for example U.S. Pat. Nos. 6,107,094; 5,898,031; Clemens et al., 1997, J. Interferon & Cytokine Res., 17, 503-524; Adah et al., 2001, Curr. Med. Chem., 8, 1189).


The presence of long dsRNAs in cells stimulates the activity of a ribonuclease III enzyme referred to as dicer (Bass, 2000, Cell, 101, 235; Zamore et al., 2000, Cell, 101, 25-33; Hammond et al., 2000, Nature, 404, 293). Dicer is involved in the processing of the dsRNA into short pieces of dsRNA known as short interfering RNAs (siRNAs) (Zamore et al., 2000, Cell, 101, 25-33; Bass, 2000, Cell, 101, 235; Berstein et al., 2001, Nature, 409, 363). Short interfering RNAs derived from dicer activity are typically about 21 to about 23 nucleotides in length and comprise about 19 base pair duplexes (Zamore et al., 2000, Cell, 101, 25-33; Elbashir et al., 2001, Genes Dev., 15, 188). Dicer has also been implicated in the excision of 21- and 22-nucleotide small temporal RNAs (stRNAs) from precursor RNA of conserved structure that are implicated in translational control (Hutvagner et al., 2001, Science, 293, 834). The RNAi response also features an endonuclease complex, commonly referred to as an RNA-induced silencing complex (RISC), which mediates cleavage of single-stranded RNA having sequence complementary to the antisense strand of the siRNA duplex. Cleavage of the target RNA takes place in the middle of the region complementary to the antisense strand of the siRNA duplex (Elbashir et al., 2001, Genes Dev., 15, 188).


RNAi has been studied in a variety of systems. Fire et al., 1998, Nature, 391, 806, were the first to observe RNAi in C. elegans. Bahramian and Zarbl, 1999, Molecular and Cellular Biology, 19, 274-283 and Wianny and Goetz, 1999, Nature Cell Biol., 2, 70, describe RNAi mediated by dsRNA in mammalian systems. Hammond et al., 2000, Nature, 404, 293, describe RNAi in Drosophila cells transfected with dsRNA. Elbashir et al., 2001, Nature, 411, 494 and Tuschl et al., International PCT Publication No. WO 01/75164, describe RNAi induced by introduction of duplexes of synthetic 21-nucleotide RNAs in cultured mammalian cells including human embryonic kidney and HeLa cells. Recent work in Drosophila embryonic lysates (Elbashir et al., 2001, EMBO J, 20, 6877 and Tuschl et al., International PCT Publication No. WO 01/75164) has revealed certain requirements for siRNA length, structure, chemical composition, and sequence that are essential to mediate efficient RNAi activity. These studies have shown that 21-nucleotide siRNA duplexes are most active when containing 3′-terminal dinucleotide overhangs. Furthermore, complete substitution of one or both siRNA strands with 2′-deoxy (2′-H) or 2′-O-methyl nucleotides abolishes RNAi activity, whereas substitution of the 3′-terminal siRNA overhang nucleotides with 2′-deoxy nucleotides (2′-H) was shown to be tolerated. Single mismatch sequences in the center of the siRNA duplex were also shown to abolish RNAi activity. In addition, these studies also indicate that the position of the cleavage site in the target RNA is defined by the 5′-end of the siRNA guide sequence rather than the 3′-end of the guide sequence (Elbashir et al., 2001, EMBO J., 20, 6877). Other studies have indicated that a 5′-phosphate on the target-complementary strand of a siRNA duplex is required for siRNA activity and that ATP is utilized to maintain the 5′-phosphate moiety on the siRNA (Nykanen et al., 2001, Cell, 107, 309).


Studies have shown that replacing the 3′-terminal nucleotide overhanging segments of a 21-mer siRNA duplex having two-nucleotide 3′-overhangs with deoxyribonucleotides does not have an adverse effect on RNAi activity. Replacing up to four nucleotides on each end of the siRNA with deoxyribonucleotides has been reported to be well tolerated, whereas complete substitution with deoxyribonucleotides results in no RNAi activity (Elbashir et al., 2001, EMBO J., 20, 6877 and Tuschl et al., International PCT Publication No. WO 01/75164). In addition, Elbashir et al., supra, also report that substitution of siRNA with 2′-O-methyl nucleotides completely abolishes RNAi activity. Li et al., International PCT Publication No. WO 00/44914, and Beach et al., International PCT Publication No. WO 01/68836 preliminarily suggest that siRNA may include modifications to either the phosphate-sugar backbone or the nucleoside to include at least one of a nitrogen or sulfur heteroatom, however, neither application postulates to what extent such modifications would be tolerated in siRNA molecules, nor provides any further guidance or examples of such modified siRNA. Kreutzer et al., Canadian Patent Application No. 2,359,180, also describe certain chemical modifications for use in dsRNA constructs in order to counteract activation of double-stranded RNA-dependent protein kinase PKR, specifically 2′-amino or 2′-O-methyl nucleotides, and nucleotides containing a 2′-O or 4′-C methylene bridge. However, Kreutzer et al. similarly fails to provide examples or guidance as to what extent these modifications would be tolerated in dsRNA molecules.


Parrish et al., 2000, Molecular Cell, 6, 1077-1087, tested certain chemical modifications targeting the unc-22 gene in C. elegans using long (>25 nt) siRNA transcripts. The authors describe the introduction of thiophosphate residues into these siRNA transcripts by incorporating thiophosphate nucleotide analogs with T7 and T3 RNA polymerase and observed that RNAs with two phosphorothioate modified bases also had substantial decreases in effectiveness as RNAi. Further, Parrish et al. reported that phosphorothioate modification of more than two residues greatly destabilized the RNAs in vitro such that interference activities could not be assayed. Id. at 1081. The authors also tested certain modifications at the 2′-position of the nucleotide sugar in the long siRNA transcripts and found that substituting deoxynucleotides for ribonucleotides produced a substantial decrease in interference activity, especially in the case of Uridine to Thymidine and/or Cytidine to deoxy-Cytidine substitutions. Id. In addition, the authors tested certain base modifications, including substituting, in sense and antisense strands of the siRNA, 4-thiouracil, 5-bromouracil, 5-iodouracil, and 3-(aminoallyl)uracil for uracil, and inosine for guanosine. Whereas 4-thiouracil and 5-bromouracil substitution appeared to be tolerated, Parrish reported that inosine produced a substantial decrease in interference activity when incorporated in either strand. Parrish also reported that incorporation of 5-iodouracil and 3-(aminoallyl)uracil in the antisense strand resulted in a substantial decrease in RNAi activity as well.


The use of longer dsRNA has been described. For example, Beach et al., International PCT Publication No. WO 01/68836, describes specific methods for attenuating gene expression using endogenously-derived dsRNA. Tuschl et al., International PCT Publication No. WO 01/75164, describe a Drosophila in vitro RNAi system and the use of specific siRNA molecules for certain functional genomic and certain therapeutic applications; although Tuschl, 2001, Chem. Biochem., 2, 239-245, doubts that RNAi can be used to cure genetic diseases or viral infection due to the danger of activating interferon response. Li et al., International PCT Publication No. WO 00/44914, describe the use of specific long (141 bp488 bp) enzymatically synthesized or vector expressed dsRNAs for attenuating the expression of certain target genes. Zernicka-Goetz et al., International PCT Publication No. WO 01/36646, describe certain methods for inhibiting the expression of particular genes in mammalian cells using certain long (550 bp-714 bp), enzymatically synthesized or vector expressed dsRNA molecules. Fire et al., International PCT Publication No. WO 99/32619, describe particular methods for introducing certain long dsRNA molecules into cells for use in inhibiting gene expression in nematodes. Plaetinck et al., International PCT Publication No. WO 00/01846, describe certain methods for identifying specific genes responsible for conferring a particular phenotype in a cell using specific long dsRNA molecules. Mello et al., International PCT Publication No. WO 01/29058, describe the identification of specific genes involved in dsRNA-mediated RNAi. Pachuck et al., International PCT Publication No. WO 00/63364, describe certain long (at least 200 nucleotide) dsRNA constructs. Deschamps Depaillette et al., International PCT Publication No. WO 99/07409, describe specific compositions consisting of particular dsRNA molecules combined with certain anti-viral agents. Waterhouse et al., International PCT Publication No. 99/53050 and 1998, PNAS, 95, 13959-13964, describe certain methods for decreasing the phenotypic expression of a nucleic acid in plant cells using certain dsRNAs. Driscoll et al., International PCT Publication No. WO 01/49844, describe specific DNA expression constructs for use in facilitating gene silencing in targeted organisms.


Others have reported on various RNAi and gene-silencing systems. For example, Parrish et al., 2000, Molecular Cell, 6, 1077-1087, describe specific chemically-modified dsRNA constructs targeting the unc-22 gene of C. elegans. Grossniklaus, International PCT Publication No. WO 01/38551, describes certain methods for regulating polycomb gene expression in plants using certain dsRNAs. Churikov et al., International PCT Publication No. WO 01/42443, describe certain methods for modifying genetic characteristics of an organism using certain dsRNAs. Cogoni et al,, International PCT Publication No. WO 01/53475, describe certain methods for isolating a Neurospora silencing gene and uses thereof. Reed et al., International PCT Publication No. WO 01/68836, describe certain methods for gene silencing in plants. Honer et al., International PCT Publication No. WO 01/70944, describe certain methods of drug screening using transgenic nematodes as Parkinson's Disease models using certain dsRNAs. Deak et al., International PCT Publication No. WO 01/72774, describe certain Drosophila-derived gene products that may be related to RNAi in Drosophila. Arndt et al., International PCT Publication No. WO 01/92513 describe certain methods for mediating gene suppression by using factors that enhance RNAi. Tuschl et al., International PCT Publication No. WO 02/44321, describe certain synthetic siRNA constructs. Pachuk et al., International PCT Publication No. WO 00/63364, and Satishchandran et al., International PCT Publication No. WO 01/04313, describe certain methods and compositions for inhibiting the function of certain polynucleotide sequences using certain long (over 250 bp), vector expressed dsRNAs. Echeverri et al., International PCT Publication No. WO 02/38805, describe certain C. elegans genes identified via RNAi. Kreutzer et al., International PCT Publications Nos. WO 02/055692, WO 02/055693, and EP 1144623 B1 describes certain methods for inhibiting gene expression using dsRNA. Graham et al., International PCT Publications Nos. WO 99/49029 and WO 01/70949, and AU 4037501 describe certain vector expressed siRNA molecules. Fire et al., U.S. Pat. No. 6,506,559, describe certain methods for inhibiting gene expression in vitro using certain long dsRNA (299 bp-1033 bp) constructs that mediate RNAi. Martinez et al., 2002, Cell, 110, 563-574, describe certain single stranded siRNA constructs, including certain 5′-phosphorylated single stranded siRNAs that mediate RNA interference in Hela cells. Harborth et al., 2003, Antisense & Nucleic Acid Drug Development, 13, 83-105, describe certain chemically and structurally modified siRNA molecules. Chiu and Rana, 2003, RNA, 9, 1034-1048, describe certain chemically and structurally modified siRNA molecules. Woolf et al., International PCT Publication Nos. WO 03/064626 and WO 03/064625 describe certain chemically modified dsRNA constructs.


McCaffrey et al., 2002, Nature, 418, 38-39, describes the use of certain siRNA constructs targeting a chimeric HCV NS5B protein/luciferase transcript in mice.


Randall et al., 2003, PNAS USA, 100, 235-240, describe certain siRNA constructs targeting HCV RNA in Huh7 hepatoma cell lines.


SUMMARY OF THE INVENTION

This invention relates to compounds, compositions, and methods useful for modulating the expression of genes, such as those genes associated with the development or maintenance of HCV infection, liver failure, hepatocellular carcinoma, cirrhosis, and/or other disease states associated with HCV infection, by RNA interference (RNAi) using short interfering nucleic acid (siNA) molecules. This invention also relates to compounds, compositions, and methods useful for modulating the expression and activity of other genes involved in pathways of HCV gene expression and/or activity by RNA interference (RNAi) using small nucleic acid molecules. In particular, the instant invention features small nucleic acid molecules, such as short interfering nucleic acid (siNA), short interfering RNA (siRNA), double-stranded RNA (dsRNA), micro-RNA (miRNA), and short hairpin RNA (shRNA) molecules and methods used to modulate the expression of HCV genes and/or other genes (e.g., cellular or host genes) involved in pathways of HCV gene expression and/or infection.


A siNA of the invention can be unmodified or chemically-modified. A siNA of the instant invention can be chemically synthesized, expressed from a vector or enzymatically synthesized. The instant invention also features various chemically-modified synthetic short interfering nucleic acid (siNA) molecules capable of modulating HCV gene expression or activity in cells by RNA interference (RNAi). The use of chemically-modified siNA improves various properties of native siNA molecules through increased resistance to nuclease degradation in vivo and/or through improved cellular uptake. Further, contrary to earlier published studies, siNA having multiple chemical modifications retains its RNAi activity. The siNA molecules of the instant invention provide useful reagents and methods for a variety of therapeutic, veterinary, diagnostic, target validation, genomic discovery, genetic engineering, and pharmacogenomic applications.


In one embodiment, the invention features one or more siNA molecules and methods that independently or in combination modulate the expression of gene(s) encoding HCV and/or cellular proteins associated with the maintenance or development of HCV infection, liver failure, hepatocellular carcinoma, and cirrhosis, such as genes encoding sequences comprising those sequences referred to by GenBank Accession Nos. shown in Table I, referred to herein generally as HCV. The description below of the various aspects and embodiments of the invention is provided with reference to exemplary hepatitis C virus (HCV) genes, generally referred to herein as HCV. However, such reference is meant to be exemplary only and the various aspects and embodiments of the invention are also directed to other genes that express alternate HCV genes, such as mutant HCV genes, splice variants of HCV genes, and genes encoding different strains of HCV, as well as as cellular targets for HCV, such as those described herein. The various aspects and embodiments are also directed to other genes involved in HCV pathways, including genes that encode cellular proteins involved in the maintenance and/or development of HCV infection, liver failure, hepatocellular carcinoma, and cirrhosis or other genes that express other proteins associated with HCV infection, such as cellular proteins that are utilized in the HCV life-cycle. Such additional genes can be analyzed for target sites using the methods described herein for HCV. Thus, the inhibition and the effects of such inhibition of the other genes can be performed as described herein. In other words, the term “HCV” as it is defined herein below and recited in the described embodiments, is meant to encompass genes associated with the development and/or maintenance of HCV infection, such as genes which encode HCV polypeptides, including polypeptides of different strains of HCV, mutant HCV genes, and splice variants of HCV genes, as well as cellular genes involved in HCV pathways of gene expression, replication, and/or HCV activity. Also, the term “HCV” as it is defined herein below and recited in the described embodiments, is meant to encompass HCV viral gene products and cellular gene products involved in HCV infection, such as those described herein. Thus, each of the embodiments described herein with reference to the term “HCV” are applicable to all of the virus, cellular and viral protein, peptide, polypeptide, and/or polynucleotide molecules covered by the term “HCV”, as that term is defined herein.


In one embodiment, the invention features siNA molecules having RNAi specificity for the HCV minus strand, for example, Genbank Accession No. HPCK1S1, Hepatitis C virus (strain HCV-1b, clone HCV-K1-S1), complete genome; Genbank Accession No. D50483, 9410 nt.


In one embodiment, the invention features one or more siNA molecules and methods that independently or in combination modulate the expression of genes representing cellular targets for HCV infection, such as cellular receptors, cell surface molecules, cellular enzymes, cellular transcription factors, and/or cytokines, second messengers, and cellular accessory molecules including, but not limited to, La antigen (see for example Costa-Mattioli et al., 2004, Mol Cell Biol., 24, 6861-70, e.g., Genbank Accession No. NM003142); FAS (e.g., Genbank Accession No. NM000043) or FAS ligand (e.g., Genbank Accession No. NM000639); interferon regulatory factors (IRFs; e.g., Genbank Accession No. AF082503.1); cellular PKR protein kinase (e.g., Genbank Accession No. XM002661.7); human eukaryotic initiation factors 2B (elF2Bgamma; e.g., Genbank Accession No. AF256223, and/or elF2gamma; e.g., Genbank Accession No. NM006874.1); human DEAD Box protein (DDX3; e.g., Genbank Accession No. XM018021.2); and cellular proteins that bind to the poly(U) tract of the HCV 3′-UTR, such as polypyrimidine tract-binding protein (e.g., Genbank Accession Nos. NM031991.1 and XM042972.3). Such cellular targets are also referred to herein generally as HCV targets, and specifically as “host target” or “host targets”.


Due to the high sequence variability of the HCV genome, selection of siNA molecules for broad therapeutic applications likely involve the conserved regions of the HCV genome. In one embodiment, the present invention relates to siNA molecules that target the conserved regions of the HCV genome. Examples of conserved regions of the HCV genome include, but are not limited to, the 5′-Non Coding Region (NCR, also referred to as the 5′-untranslated region, UTR), the 5′-end of the core protein coding region, and the 3′-NCR. HCV genomic RNA contains an internal ribosome entry site (IRES) in the 5′-NCR which mediates translation independently of a 5′-cap structure (Wang et al., 1993, J. Virol., 67, 3338-44). The full-length sequence of the HCV RNA genome is heterologous among clinically isolated subtypes, of which there are at least fifteen (Simmonds, 1995, Hepatology, 21, 570-583), however, the 5′-NCR sequence of HCV is highly conserved across all known subtypes, most likely to preserve the shared IRES mechanism (Okamoto et al., 1991, J. General Virol., 72, 2697-2704). Therefore, a siNA molecule can be designed to target the different isolates of HCV by targeting a conserved region, such as the 5′ NCR sequence. siNA molecules designed to target conserved regions of various HCV isolates enable efficient inhibition of HCV replication in diverse patient populations and ensure the effectiveness of the siNA molecules against HCV quasi species which evolve due to mutations in the non-conserved regions of the HCV genome. As described, a single siNA molecule can be targeted against all isolates of HCV by designing the siNA molecule to interact with conserved nucleotide sequences of HCV (e.g., sequences that are expected to be present in the RNA of various HCV isolates).


In one embodiment, the invention features a double-stranded short interfering nucleic acid (siNA) molecule that down-regulates expression of a HCV gene or that directs cleavage of a HCV RNA, wherein said siNA molecule comprises about 15 to about 28 base pairs.


In one embodiment, the invention features a double stranded short interfering nucleic acid (siNA) molecule that directs cleavage of a HCV RNA via RNA interference (RNAi), wherein the double stranded siNA molecule comprises a first and a second strand, each strand of the siNA molecule is about 18 to about 28 nucleotides in length, the first strand of the siNA molecule comprises nucleotide sequence having sufficient complementarity to the HCV RNA for the siNA molecule to direct cleavage of the HCV RNA via RNA interference, and the second strand of said siNA molecule comprises nucleotide sequence that is complementary to the first strand.


In one embodiment, the invention features a double stranded short interfering nucleic acid (siNA) molecule that directs cleavage of a HCV RNA via RNA interference (RNAi), wherein the double stranded siNA molecule comprises a first and a second strand, each strand of the siNA molecule is about 18 to about 23 nucleotides in length, the first strand of the siNA molecule comprises nucleotide sequence having sufficient complementarity to the HCV RNA for the siNA molecule to direct cleavage of the HCV RNA via RNA interference, and the second strand of said siNA molecule comprises nucleotide sequence that is complementary to the first strand.


In one embodiment, the invention features a chemically synthesized double stranded short interfering nucleic acid (siNA) molecule that directs cleavage of a HCV RNA via RNA interference (RNAi), wherein each strand of the siNA molecule is about 18 to about 28 nucleotides in length; and one strand of the siNA molecule comprises nucleotide sequence having sufficient complementarity to the HCV RNA for the siNA molecule to direct cleavage of the HCV RNA via RNA interference.


In one embodiment, the invention features a chemically synthesized double stranded short interfering nucleic acid (siNA) molecule that directs cleavage of a HCV RNA via RNA interference (RNAi), wherein each strand of the siNA molecule is about 18 to about 23 nucleotides in length; and one strand of the siNA molecule comprises nucleotide sequence having sufficient complementarity to the HCV RNA for the siNA molecule to direct cleavage of the HCV RNA via RNA interference.


In one embodiment, the invention features a siNA molecule that down-regulates expression of a HCV gene or that directs cleavage of a HCV RNA, for example, wherein the HCV gene or RNA comprises HCV encoding sequence. In one embodiment, the invention features a siNA molecule that down-regulates expression of a HCV gene or that directs cleavage of a HCV RNA, for example, wherein the HCV gene of RNA comprises HCV non-coding sequence or regulatory elements involved in HCV gene expression.


In one embodiment, a siNA of the invention is used to inhibit the expression of HCV genes or a HCV gene family (e.g., different HCV strains), wherein the genes or gene family sequences share sequence homology. Such homologous sequences can be identified as is known in the art, for example using sequence alignments. siNA molecules can be designed to target such homologous sequences, for example using perfectly complementary sequences or by incorporating non-canonical base pairs, for example mismatches and/or wobble base pairs, that can provide additional target sequences. In instances where mismatches are identified, non-canonical base pairs (for example, mismatches and/or wobble bases) can be used to generate siNA molecules that target more than one gene sequence. In a non-limiting example, non-canonical base pairs such as UU and CC base pairs are used to generate siNA molecules that are capable of targeting sequences for differing HCV targets that share sequence homology. As such, one advantage of using siNAs of the invention is that a single siNA can be designed to include nucleic acid sequence that is complementary to the nucleotide sequence that is conserved between the homologous genes. In this approach, a single siNA can be used to inhibit expression of more than one gene instead of using more than one siNA molecule to target the different genes.


In one embodiment, the invention features a siNA molecule having RNAi activity against HCV RNA, wherein the siNA molecule comprises a sequence complementary to any RNA having HCV encoding sequence, such as those sequences having GenBank Accession Nos. shown in Table I. In another embodiment, the invention features a siNA molecule having RNAi activity against HCV RNA, wherein the siNA molecule comprises a sequence complementary to an RNA having variant HCV encoding sequence, for example other mutant HCV genes not shown in Table I but known in the art to be associated with, for example, the maintenance and/or development of HCV infection, liver failure, hepatocellular carcinoma, or cirrhosis. Chemical modifications as shown in Tables III and IV or otherwise described herein can be applied to any siNA construct of the invention. In another embodiment, a siNA molecule of the invention includes a nucleotide sequence that can interact with nucleotide sequence of a HCV gene and thereby mediate silencing of HCV gene expression, for example, wherein the siNA mediates regulation of HCV gene expression by cellular processes that modulate the transcription or translation of the HCV gene and prevent expression of the HCV gene.


In one embodiment, siNA molecules of the invention are used to down regulate or inhibit the expression of proteins arising from haplotype polymorphisms (e.g., cellular genes involved in HCV infection or replication) that are associated with a trait, disease or condition. Analysis of genes, or protein or RNA levels can be used to identify subjects with such polymorphisms or those subjects who are at risk of developing traits, conditions, or diseases described herein (see for example Silvestri et al., 2003, Int J Cancer., 104, 310-7). These subjects are amenable to treatment, for example, treatment with siNA molecules of the invention and any other composition useful in treating diseases related to HCV gene expression. As such, analysis of HCV protein or RNA levels can be used to determine treatment type and the course of therapy in treating a subject. Monitoring of HCV protein or RNA levels can be used to predict treatment outcome and to determine the efficacy of compounds and compositions that modulate the level and/or activity of certain HCV proteins associated with a trait, condition, or disease.


In one embodiment of the invention a siNA molecule comprises an antisense strand comprising a nucleotide sequence that is complementary to a nucleotide sequence or a portion thereof encoding a HCV protein. The siNA further comprises a sense strand, wherein said sense strand comprises a nucleotide sequence of a HCV gene or a portion thereof.


In another embodiment, a siNA molecule comprises an antisense region comprising a nucleotide sequence that is complementary to a nucleotide sequence encoding a HCV protein or a portion thereof. The siNA molecule further comprises a sense region, wherein said sense region comprises a nucleotide sequence of a HCV gene or a portion thereof.


In another embodiment, the invention features a siNA molecule comprising a nucleotide sequence in the antisense region of the siNA molecule that is complementary to a nucleotide sequence or portion of sequence of a HCV gene. In another embodiment, the invention features a siNA molecule comprising a region, for example, the antisense region of the siNA construct, complementary to a sequence comprising a HCV gene sequence or a portion thereof.


In one embodiment, the antisense region of HCV siNA constructs comprises a sequence complementary to sequence having any of SEQ ID NOs. 1-696 or 1393-1466. In one embodiment, the antisense region of HCV constructs comprises sequence having any of antisense SEQ ID NOs. in Tables II and III and FIGS. 4 and 5. In another embodiment, the sense region of HCV constructs comprises sequence having any of sense SEQ ID NOs. in Tables II and III and FIGS. 4 and 5.


In one embodiment, a siNA molecule of the invention comprises any of SEQ ID NOs. 1-2027. The sequences shown in SEQ ID NOs: 1-2027 are not limiting. A siNA molecule of the invention can comprise any contiguous HCV sequence (e.g., about 15 to about 25 or more, or about 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, or 25 or more contiguous HCV nucleotides).


In yet another embodiment, the invention features a siNA molecule comprising a sequence, for example, the antisense sequence of the siNA construct, complementary to a sequence or portion of sequence comprising sequence represented by GenBank Accession Nos. shown in Table I. Chemical modifications in Tables III and IV and described herein can be applied to any siNA construct of the invention.


In one embodiment of the invention a siNA molecule comprises an antisense strand having about 15 to about 30 (e.g., about 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30) nucleotides, wherein the antisense strand is complementary to a RNA sequence or a portion thereof encoding HCV or a HCV protein, and wherein said siNA further comprises a sense strand having about 15 to about 30 (e.g., about 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30) nucleotides, and wherein said sense strand and said antisense strand are distinct nucleotide sequences where at least about 15 nucleotides in each strand are complementary to the other strand.


In another embodiment of the invention a siNA molecule of the invention comprises an antisense region having about 15 to about 30 (e.g., about 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30) nucleotides, wherein the antisense region is complementary to a RNA sequence encoding HCV or a HCV protein, and wherein said siNA further comprises a sense region having about 15 to about 30 (e.g., about 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30) nucleotides, wherein said sense region and said antisense region are comprised in a linear molecule where the sense region comprises at least about 15 nucleotides that are complementary to the antisense region.


In one embodiment, a siNA molecule of the invention has RNAi activity that modulates expression of RNA encoded by a HCV gene. Because HCV genes can share some degree of sequence homology with each other, siNA molecules can be designed to target a class of HCV genes (e.g., a class of different HCV strains) or alternately specific HCV genes (e.g., escape mutants, resistant strains, or other polymorphic variants) by selecting sequences that are either shared amongst different HCV targets or alternatively that are unique for a specific HCV target. Therefore, in one embodiment, the siNA molecule can be designed to target conserved regions of HCV RNA sequences having homology among several HCV gene variants so as to target a class of HCV genes with one siNA molecule. Accordingly, in one embodiment, the siNA molecule of the invention modulates the expression of one or more HCV stains in a subject or organism. In another embodiment, the siNA molecule can be designed to target a sequence that is unique to a specific HCV RNA sequence (e.g., a single HCV strain or HCV single nucleotide polymorphism (SNP)) due to the high degree of specificity that the siNA molecule requires to mediate RNAi activity.


In one embodiment, nucleic acid molecules of the invention that act as mediators of the RNA interference gene silencing response are double-stranded nucleic acid molecules. In another embodiment, the siNA molecules of the invention consist of duplex nucleic acid molecules containing about 15 to about 30 base pairs between oligonucleotides comprising about 15 to about 30 (e.g., about 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30) nucleotides. In yet another embodiment, siNA molecules of the invention comprise duplex nucleic acid molecules with overhanging ends of about 1 to about 3 (e.g., about 1, 2, or 3) nucleotides, for example, about 21-nucleotide duplexes with about 19 base pairs and 3′-terminal mononucleotide, dinucleotide, or trinucleotide overhangs. In yet another embodiment, siNA molecules of the invention comprise duplex nucleic acid molecules with blunt ends, where both ends are blunt, or alternatively, where one of the ends is blunt.


In one embodiment, the invention features one or more chemically-modified siNA constructs having specificity for HCV expressing nucleic acid molecules, such as RNA encoding a HCV protein. In one embodiment, the invention features a RNA based siNA molecule (e.g., a siNA comprising 2′-OH nucleotides) having specificity for HCV expressing nucleic acid molecules that includes one or more chemical modifications described herein. Non-limiting examples of such chemical modifications include without limitation phosphorothioate internucleotide linkages, 2′-deoxyribonucleotides, 2′-O-methyl ribonucleotides, 2′-deoxy-2′-fluoro ribonucleotides, “universal base” nucleotides, “acyclic” nucleotides, 5-C-methyl nucleotides, and terminal glyceryl and/or inverted deoxy abasic residue incorporation. These chemical modifications, when used in various siNA constructs, (e.g., RNA based siNA constructs), are shown to preserve RNAi activity in cells while at the same time, dramatically increasing the serum stability of these compounds. Furthermore, contrary to the data published by Parrish et al., supra, applicant demonstrates that multiple (greater than one) phosphorothioate substitutions are well-tolerated and confer substantial increases in serum stability for modified siNA constructs.


In one embodiment, a siNA molecule of the invention comprises modified nucleotides while maintaining the ability to mediate RNAi. The modified nucleotides can be used to improve in vitro or in vivo characteristics such as stability, activity, and/or bioavailability. For example, a siNA molecule of the invention can comprise modified nucleotides as a percentage of the total number of nucleotides present in the siNA molecule. As such, a siNA molecule of the invention can generally comprise about 5% to about 100% modified nucleotides (e.g., about 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% or 100% modified nucleotides). The actual percentage of modified nucleotides present in a given siNA molecule will depend on the total number of nucleotides present in the siNA. If the siNA molecule is single stranded, the percent modification can be based upon the total number of nucleotides present in the single stranded siNA molecules. Likewise, if the siNA molecule is double stranded, the percent modification can be based upon the total number of nucleotides present in the sense strand, antisense strand, or both the sense and antisense strands.


One aspect of the invention features a double-stranded short interfering nucleic acid (siNA) molecule that down-regulates expression of a HCV gene or that directs cleavage of a HCV RNA. In one embodiment, the double stranded siNA molecule comprises one or more chemical modifications and each strand of the double-stranded siNA is about 21 nucleotides long. In one embodiment, the double-stranded siNA molecule does not contain any ribonucleotides. In another embodiment, the double-stranded siNA molecule comprises one or more ribonucleotides. In one embodiment, each strand of the double-stranded siNA molecule independently comprises about 15 to about 30 (e.g., about 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30) nucleotides, wherein each strand comprises about 15 to about 30 (e.g., about 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30) nucleotides that are complementary to the nucleotides of the other strand. In one embodiment, one of the strands of the double-stranded siNA molecule comprises a nucleotide sequence that is complementary to a nucleotide sequence or a portion thereof of the HCV gene, and the second strand of the double-stranded siNA molecule comprises a nucleotide sequence substantially similar to the nucleotide sequence of the HCV gene or a portion thereof.


In another embodiment, the invention features a double-stranded short interfering nucleic acid (siNA) molecule that down-regulates expression of a HCV gene or that directs cleavage of a HCV RNA, comprising an antisense region, wherein the antisense region comprises a nucleotide sequence that is complementary to a nucleotide sequence of the HCV gene or a portion thereof, and a sense region, wherein the sense region comprises a nucleotide sequence substantially similar to the nucleotide sequence of the HCV gene or a portion thereof. In one embodiment, the antisense region and the sense region independently comprise about 15 to about 30 (e.g. about 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30) nucleotides, wherein the antisense region comprises about 15 to about 30 (e.g. about 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30) nucleotides that are complementary to nucleotides of the sense region.


In another embodiment, the invention features a double-stranded short interfering nucleic acid (siNA) molecule that down-regulates expression of a HCV gene or that directs cleavage of a HCV RNA, comprising a sense region and an antisense region, wherein the antisense region comprises a nucleotide sequence that is complementary to a nucleotide sequence of RNA encoded by the HCV gene or a portion thereof and the sense region comprises a nucleotide sequence that is complementary to the antisense region.


In one embodiment, a siNA molecule of the invention comprises blunt ends, i.e., ends that do not include any overhanging nucleotides. For example, a siNA molecule comprising modifications described herein (e.g., comprising nucleotides having Formulae I-VII or siNA constructs comprising “Stab 00”-“Stab 32” (Table IV) or any combination thereof (see Table IV)) and/or any length described herein can comprise blunt ends or ends with no overhanging nucleotides.


In one embodiment, any siNA molecule of the invention can comprise one or more blunt ends, i.e. where a blunt end does not have any overhanging nucleotides. In one embodiment, the blunt ended siNA molecule has a number of base pairs equal to the number of nucleotides present in each strand of the siNA molecule. In another embodiment, the siNA molecule comprises one blunt end, for example wherein the 5′-end of the antisense strand and the 3′-end of the sense strand do not have any overhanging nucleotides. In another example, the siNA molecule comprises one blunt end, for example wherein the 3′-end of the antisense strand and the 5′-end of the sense strand do not have any overhanging nucleotides. In another example, a siNA molecule comprises two blunt ends, for example wherein the 3′-end of the antisense strand and the 5′-end of the sense strand as well as the 5′-end of the antisense strand and 3′-end of the sense strand do not have any overhanging nucleotides. A blunt ended siNA molecule can comprise, for example, from about 15 to about 30 nucleotides (e.g., about 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30 nucleotides). Other nucleotides present in a blunt ended siNA molecule can comprise, for example, mismatches, bulges, loops, or wobble base pairs to modulate the activity of the siNA molecule to mediate RNA interference.


By “blunt ends” is meant symmetric termini or termini of a double stranded siNA molecule having no overhanging nucleotides. The two strands of a double stranded siNA molecule align with each other without over-hanging nucleotides at the termini. For example, a blunt ended siNA construct comprises terminal nucleotides that are complementary between the sense and antisense regions of the siNA molecule.


In one embodiment, the invention features a double-stranded short interfering nucleic acid (siNA) molecule that down-regulates expression of a HCV gene or that directs cleavage of a HCV RNA, wherein the siNA molecule is assembled from two separate oligonucleotide fragments wherein one fragment comprises the sense region and the second fragment comprises the antisense region of the siNA molecule. The sense region can be connected to the antisense region via a linker molecule, such as a polynucleotide linker or a non-nucleotide linker.


In one embodiment, the invention features double-stranded short interfering nucleic acid (siNA) molecule that down-regulates expression of a HCV gene or that directs cleavage of a HCV RNA, wherein the siNA molecule comprises about 15 to about 30 (e.g. about 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30) base pairs, and wherein each strand of the siNA molecule comprises one or more chemical modifications. In another embodiment, one of the strands of the double-stranded siNA molecule comprises a nucleotide sequence that is complementary to a nucleotide sequence of a HCV gene or a portion thereof, and the second strand of the double-stranded siNA molecule comprises a nucleotide sequence substantially similar to the nucleotide sequence or a portion thereof of the HCV gene. In another embodiment, one of the strands of the double-stranded siNA molecule comprises a nucleotide sequence that is complementary to a nucleotide sequence of a HCV gene or portion thereof, and the second strand of the double-stranded siNA molecule comprises a nucleotide sequence substantially similar to the nucleotide sequence or portion thereof of the HCV gene. In another embodiment, each strand of the siNA molecule comprises about 15 to about 30 (e.g. about 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30) nucleotides, and each strand comprises at least about 15 to about 30 (e.g. about 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30) nucleotides that are complementary to the nucleotides of the other strand. The HCV gene can comprise, for example, sequences referred to in Table I.


In one embodiment, a siNA molecule of the invention comprises no ribonucleotides. In another embodiment, a siNA molecule of the invention comprises ribonucleotides.


In one embodiment, a siNA molecule of the invention comprises an antisense region comprising a nucleotide sequence that is complementary to a nucleotide sequence of a HCV gene or a portion thereof, and the siNA further comprises a sense region comprising a nucleotide sequence substantially similar to the nucleotide sequence of the HCV gene or a portion thereof. In another embodiment, the antisense region and the sense region each comprise about 15 to about 30 (e.g. about 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30) nucleotides and the antisense region comprises at least about 15 to about 30 (e.g. about 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30) nucleotides that are complementary to nucleotides of the sense region. The HCV gene can comprise, for example, sequences referred to in Table I. In another embodiment, the siNA is a double stranded nucleic acid molecule, where each of the two strands of the siNA molecule independently comprise about 15 to about 40 (e.g. about 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 23, 33, 34, 35, 36, 37, 38, 39, or 40) nucleotides, and where one of the strands of the siNA molecule comprises at least about 15 (e.g. about 15, 16, 17, 18, 19, 20, 21, 22, 23, 24 or 25 or more) nucleotides that are complementary to the nucleic acid sequence of the HCV gene or a portion thereof.


In one embodiment, a siNA molecule of the invention comprises a sense region and an antisense region, wherein the antisense region comprises a nucleotide sequence that is complementary to a nucleotide sequence of RNA encoded by a HCV gene, or a portion thereof, and the sense region comprises a nucleotide sequence that is complementary to the antisense region. In one embodiment, the siNA molecule is assembled from two separate oligonucleotide fragments, wherein one fragment comprises the sense region and the second fragment comprises the antisense region of the siNA molecule. In another embodiment, the sense region is connected to the antisense region via a linker molecule. In another embodiment, the sense region is connected to the antisense region via a linker molecule, such as a nucleotide or non-nucleotide linker. The HCV gene can comprise, for example, sequences referred in to Table I.


In one embodiment, the invention features a double-stranded short interfering nucleic acid (siNA) molecule that down-regulates expression of a HCV gene or that directs cleavage of a HCV RNA, comprising a sense region and an antisense region, wherein the antisense region comprises a nucleotide sequence that is complementary to a nucleotide sequence of RNA encoded by the HCV gene or a portion thereof and the sense region comprises a nucleotide sequence that is complementary to the antisense region, and wherein the siNA molecule has one or more modified pyrimidine and/or purine nucleotides. In one embodiment, the pyrimidine nucleotides in the sense region are 2′-O-methylpyrimidine nucleotides or 2′-deoxy-2′-fluoro pyrimidine nucleotides and the purine nucleotides present in the sense region are 2′-deoxy purine nucleotides. In another embodiment, the pyrimidine nucleotides in the sense region are 2′-deoxy-2′-fluoro pyrimidine nucleotides and the purine nucleotides present in the sense region are 2′-O-methyl purine nucleotides. In another embodiment, the pyrimidine nucleotides in the sense region are 2′-deoxy-2′-fluoro pyrimidine nucleotides and the purine nucleotides present in the sense region are 2′-deoxy purine nucleotides. In one embodiment, the pyrimidine nucleotides in the antisense region are 2′-deoxy-2′-fluoro pyrimidine nucleotides and the purine nucleotides present in the antisense region are 2′-O-methyl or 2′-deoxy purine nucleotides. In another embodiment of any of the above-described siNA molecules, any nucleotides present in a non-complementary region of the sense strand (e.g. overhang region) are 2′-deoxy nucleotides.


In one embodiment, the invention features a double-stranded short interfering nucleic acid (siNA) molecule that down-regulates expression of a HCV gene or that directs cleavage of a HCV RNA, wherein the siNA molecule is assembled from two separate oligonucleotide fragments wherein one fragment comprises the sense region and the second fragment comprises the antisense region of the siNA molecule, and wherein the fragment comprising the sense region includes a terminal cap moiety at the 5′-end, the 3′-end, or both of the 5′ and 3′ ends of the fragment. In one embodiment, the terminal cap moiety is an inverted deoxy abasic moiety or glyceryl moiety. In one embodiment, each of the two fragments of the siNA molecule independently comprise about 15 to about 30 (e.g. about 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30) nucleotides. In another embodiment, each of the two fragments of the siNA molecule independently comprise about 15 to about 40 (e.g. about 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 23, 33, 34, 35, 36, 37, 38, 39, or 40) nucleotides. In a non-limiting example, each of the two fragments of the siNA molecule comprise about 21 nucleotides.


In one embodiment, the invention features a siNA molecule comprising at least one modified nucleotide, wherein the modified nucleotide is a 2′-deoxy-2′-fluoro nucleotide. The siNA can be, for example, about 15 to about 40 nucleotides in length. In one embodiment, all pyrimidine nucleotides present in the siNA are 2′-deoxy-2′-fluoro pyrimidine nucleotides. In one embodiment, the modified nucleotides in the siNA include at least one 2′-deoxy-2′-fluoro cytidine or 2′-deoxy-2′-fluoro uridine nucleotide. In another embodiment, the modified nucleotides in the siNA include at least one 2′-fluoro cytidine and at least one 2′-deoxy-2′-fluoro uridine nucleotides. In one embodiment, all uridine nucleotides present in the siNA are 2′-deoxy-2′-fluoro uridine nucleotides. In one embodiment, all cytidine nucleotides present in the siNA are 2′-deoxy-2′-fluoro cytidine nucleotides. In one embodiment, all adenosine nucleotides present in the siNA are 2′-deoxy-2′-fluoro adenosine nucleotides. In one embodiment, all guanosine nucleotides present in the siNA are 2′-deoxy-2′-fluoro guanosine nucleotides. The siNA can further comprise at least one modified internucleotidic linkage, such as phosphorothioate linkage. In one embodiment, the 2′-deoxy-2′-fluoronucleotides are present at specifically selected locations in the siNA that are sensitive to cleavage by ribonucleases, such as locations having pyrimidine nucleotides.


In one embodiment, the invention features a method of increasing the stability of a siNA molecule against cleavage by ribonucleases comprising introducing at least one modified nucleotide into the siNA molecule, wherein the modified nucleotide is a 2′-deoxy-2′-fluoro nucleotide. In one embodiment, all pyrimidine nucleotides present in the siNA are 2′-deoxy-2′-fluoro pyrimidine nucleotides. In one embodiment, the modified nucleotides in the siNA include at least one 2′-deoxy-2′-fluoro cytidine or 2′-deoxy-2′-fluoro uridine nucleotide. In another embodiment, the modified nucleotides in the siNA include at least one 2′-fluoro cytidine and at least one 2′-deoxy-2′-fluoro uridine nucleotides. In one embodiment, all uridine nucleotides present in the siNA are 2′-deoxy-2′-fluoro uridine nucleotides. In one embodiment, all cytidine nucleotides present in the siNA are 2′-deoxy-2′-fluoro cytidine nucleotides. In one embodiment, all adenosine nucleotides present in the siNA are 2′-deoxy-2′-fluoro adenosine nucleotides. In one embodiment, all guanosine nucleotides present in the siNA are 2′-deoxy-2′-fluoro guanosine nucleotides. The siNA can further comprise at least one modified internucleotidic linkage, such as phosphorothioate linkage. In one embodiment, the 2′-deoxy-2′-fluoronucleotides are present at specifically selected locations in the siNA that are sensitive to cleavage by ribonucleases, such as locations having pyrimidine nucleotides.


In one embodiment, the invention features a double-stranded short interfering nucleic acid (siNA) molecule that down-regulates expression of a HCV gene or that directs cleavage of a HCV RNA, comprising a sense region and an antisense region, wherein the antisense region comprises a nucleotide sequence that is complementary to a nucleotide sequence of RNA encoded by the HCV gene or a portion thereof and the sense region comprises a nucleotide sequence that is complementary to the antisense region, and wherein the purine nucleotides present in the antisense region comprise 2′-deoxy-purine nucleotides. In an alternative embodiment, the purine nucleotides present in the antisense region comprise 2′-O-methyl purine nucleotides. In either of the above embodiments, the antisense region can comprise a phosphorothioate internucleotide linkage at the 3′ end of the antisense region. Alternatively, in either of the above embodiments, the antisense region can comprise a glyceryl modification at the 3′ end of the antisense region. In another embodiment of any of the above-described siNA molecules, any nucleotides present in a non-complementary region of the antisense strand (e.g. overhang region) are 2′-deoxy nucleotides.


In one embodiment, the antisense region of a siNA molecule of the invention comprises sequence complementary to a portion of an endogenous transcript having sequence unique to a particular HCV disease related allele in a subject or organism, such as sequence comprising a single nucleotide polymorphism (SNP) associated with the disease specific allele. As such, the antisense region of a siNA molecule of the invention can comprise sequence complementary to sequences that are unique to a particular allele to provide specificity in mediating selective RNAi against the disease, condition, or trait related allele.


In one embodiment, the invention features a double-stranded short interfering nucleic acid (siNA) molecule that down-regulates expression of a HCV gene or that directs cleavage of a HCV RNA, wherein the siNA molecule is assembled from two separate oligonucleotide fragments wherein one fragment comprises the sense region and the second fragment comprises the antisense region of the siNA molecule. In another embodiment, the siNA molecule is a double stranded nucleic acid molecule, where each strand is about 21 nucleotides long and where about 19 nucleotides of each fragment of the siNA molecule are base-paired to the complementary nucleotides of the other fragment of the siNA molecule, wherein at least two 3′ terminal nucleotides of each fragment of the siNA molecule are not base-paired to the nucleotides of the other fragment of the siNA molecule. In another embodiment, the siNA molecule is a double stranded nucleic acid molecule, where each strand is about 19 nucleotide long and where the nucleotides of each fragment of the siNA molecule are base-paired to the complementary nucleotides of the other fragment of the siNA molecule to form at least about 15 (e.g., 15, 16, 17, 18, or 19) base pairs, wherein one or both ends of the siNA molecule are blunt ends. In one embodiment, each of the two 3′ terminal nucleotides of each fragment of the siNA molecule is a 2′-deoxy-pyrimidine nucleotide, such as a 2′-deoxy-thymidine. In another embodiment, all nucleotides of each fragment of the siNA molecule are base-paired to the complementary nucleotides of the other fragment of the siNA molecule. In another embodiment, the siNA molecule is a double stranded nucleic acid molecule of about 19 to about 25 base pairs having a sense region and an antisense region, where about 19 nucleotides of the antisense region are base-paired to the nucleotide sequence or a portion thereof of the RNA encoded by the HCV gene. In another embodiment, about 21 nucleotides of the antisense region are base-paired to the nucleotide sequence or a portion thereof of the RNA encoded by the HCV gene. In any of the above embodiments, the 5′-end of the fragment comprising said antisense region can optionally include a phosphate group.


In one embodiment, the invention features a double-stranded short interfering nucleic acid (siNA) molecule that inhibits the expression of a HCV RNA sequence (e.g., wherein said target RNA sequence is encoded by a HCV gene involved in the HCV pathway), wherein the siNA molecule does not contain any ribonucleotides and wherein each strand of the double-stranded siNA molecule is about 15 to about 30 nucleotides. In one embodiment, the siNA molecule is 21 nucleotides in length. Examples of non-ribonucleotide containing siNA constructs are combinations of stabilization chemistries shown in Table IV in any combination of Sense/Antisense chemistries, such as Stab 7/8, Stab 7/11, Stab 8/8, Stab 18/8, Stab 18/11, Stab 12/13, Stab 7/13, Stab 18/13, Stab 7/19, Stab 8/19, Stab 18/19, Stab 7/20, Stab 8/20, Stab 18/20, Stab 7/32, Stab 8/32, or Stab 18/32 (e.g., any siNA having Stab 7, 8, 11, 12, 13, 14, 15, 17, 18, 19, 20, or 32 sense or antisense strands or any combination thereof).


In one embodiment, the invention features a chemically synthesized double stranded RNA molecule that directs cleavage of a HCV RNA via RNA interference, wherein each strand of said RNA molecule is about 15 to about 30 nucleotides in length; one strand of the RNA molecule comprises nucleotide sequence having sufficient complementarity to the HCV RNA for the RNA molecule to direct cleavage of the HCV RNA via RNA interference; and wherein at least one strand of the RNA molecule optionally comprises one or more chemically modified nucleotides described herein, such as without limitation deoxynucleotides, 2′-O-methyl nucleotides, 2′-deoxy-2′-fluoro nucleotides, 2′-O-methoxyethyl nucleotides etc.


In one embodiment, the invention features a medicament comprising a siNA molecule of the invention.


In one embodiment, the invention features an active ingredient comprising a siNA molecule of the invention.


In one embodiment, the invention features the use of a double-stranded short interfering nucleic acid (siNA) molecule to inhibit, down-regulate, or reduce expression of a HCV gene, wherein the siNA molecule comprises one or more chemical modifications and each strand of the double-stranded siNA is independently about 15 to about 30 or more (e.g., about 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29 or 30 or more) nucleotides long. In one embodiment, the siNA molecule of the invention is a double stranded nucleic acid molecule comprising one or more chemical modifications, where each of the two fragments of the siNA molecule independently comprise about 15 to about 40 (e.g. about 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 23, 33, 34, 35, 36, 37, 38, 39, or 40) nucleotides and where one of the strands comprises at least 15 nucleotides that are complementary to nucleotide sequence of HCV encoding RNA or a portion thereof. In a non-limiting example, each of the two fragments of the siNA molecule comprise about 21 nucleotides. In another embodiment, the siNA molecule is a double stranded nucleic acid molecule comprising one or more chemical modifications, where each strand is about 21 nucleotide long and where about 19 nucleotides of each fragment of the siNA molecule are base-paired to the complementary nucleotides of the other fragment of the siNA molecule, wherein at least two 3′ terminal nucleotides of each fragment of the siNA molecule are not base-paired to the nucleotides of the other fragment of the siNA molecule. In another embodiment, the siNA molecule is a double stranded nucleic acid molecule comprising one or more chemical modifications, where each strand is about 19 nucleotide long and where the nucleotides of each fragment of the siNA molecule are base-paired to the complementary nucleotides of the other fragment of the siNA molecule to form at least about 15 (e.g., 15, 16, 17, 18, or 19) base pairs, wherein one or both ends of the siNA molecule are blunt ends. In one embodiment, each of the two 3′ terminal nucleotides of each fragment of the siNA molecule is a 2′-deoxy-pyrimidine nucleotide, such as a 2′-deoxy-thymidine. In another embodiment, all nucleotides of each fragment of the siNA molecule are base-paired to the complementary nucleotides of the other fragment of the siNA molecule. In another embodiment, the siNA molecule is a double stranded nucleic acid molecule of about 19 to about 25 base pairs having a sense region and an antisense region and comprising one or more chemical modifications, where about 19 nucleotides of the antisense region are base-paired to the nucleotide sequence or a portion thereof of the RNA encoded by the HCV gene. In another embodiment, about 21 nucleotides of the antisense region are base-paired to the nucleotide sequence or a portion thereof of the RNA encoded by the HCV gene. In any of the above embodiments, the 5′-end of the fragment comprising said antisense region can optionally include a phosphate group.


In one embodiment, the invention features the use of a double-stranded short interfering nucleic acid (siNA) molecule that inhibits, down-regulates, or reduces expression of a HCV gene, wherein one of the strands of the double-stranded siNA molecule is an antisense strand which comprises nucleotide sequence that is complementary to nucleotide sequence of HCV RNA or a portion thereof, the other strand is a sense strand which comprises nucleotide sequence that is complementary to a nucleotide sequence of the antisense strand and wherein a majority of the pyrimidine nucleotides present in the double-stranded siNA molecule comprises a sugar modification.


In one embodiment, the invention features a double-stranded short interfering nucleic acid (siNA) molecule that inhibits, down-regulates, or reduces expression of a HCV gene, wherein one of the strands of the double-stranded siNA molecule is an antisense strand which comprises nucleotide sequence that is complementary to nucleotide sequence of HCV RNA or a portion thereof, wherein the other strand is a sense strand which comprises nucleotide sequence that is complementary to a nucleotide sequence of the antisense strand and wherein a majority of the pyrimidine nucleotides present in the double-stranded siNA molecule comprises a sugar modification.


In one embodiment, the invention features a double-stranded short interfering nucleic acid (siNA) molecule that inhibits, down-regulates, or reduces expression of a HCV gene, wherein one of the strands of the double-stranded siNA molecule is an antisense strand which comprises nucleotide sequence that is complementary to nucleotide sequence of HCV RNA that encodes a protein or portion thereof, the other strand is a sense strand which comprises nucleotide sequence that is complementary to a nucleotide sequence of the antisense strand and wherein a majority of the pyrimidine nucleotides present in the double-stranded siNA molecule comprises a sugar modification. In one embodiment, each strand of the siNA molecule comprises about 15 to about 30 or more (e.g., about 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30 or more) nucleotides, wherein each strand comprises at least about 15 nucleotides that are complementary to the nucleotides of the other strand. In one embodiment, the siNA molecule is assembled from two oligonucleotide fragments, wherein one fragment comprises the nucleotide sequence of the antisense strand of the siNA molecule and a second fragment comprises nucleotide sequence of the sense region of the siNA molecule. In one embodiment, the sense strand is connected to the antisense strand via a linker molecule, such as a polynucleotide linker or a non-nucleotide linker. In a further embodiment, the pyrimidine nucleotides present in the sense strand are 2′-deoxy-2′fluoro pyrimidine nucleotides and the purine nucleotides present in the sense region are 2′-deoxy purine nucleotides. In another embodiment, the pyrimidine nucleotides present in the sense strand are 2′-deoxy-2′fluoro pyrimidine nucleotides and the purine nucleotides present in the sense region are 2′-O-methyl purine nucleotides. In still another embodiment, the pyrimidine nucleotides present in the antisense strand are 2′-deoxy-2′-fluoro pyrimidine nucleotides and any purine nucleotides present in the antisense strand are 2′-deoxy purine nucleotides. In another embodiment, the antisense strand comprises one or more 2′-deoxy-2′-fluoro pyrimidine nucleotides and one or more 2′-O-methyl purine nucleotides. In another embodiment, the pyrimidine nucleotides present in the antisense strand are 2′-deoxy-2′-fluoro pyrimidine nucleotides and any purine nucleotides present in the antisense strand are 2′-O-methyl purine nucleotides. In a further embodiment the sense strand comprises a 3′-end and a 5′-end, wherein a terminal cap moiety (e.g., an inverted deoxy abasic moiety or inverted deoxy nucleotide moiety such as inverted thymidine) is present at the 5′-end, the 3′-end, or both of the 5′ and 3′ ends of the sense strand. In another embodiment, the antisense strand comprises a phosphorothioate internucleotide linkage at the 3′ end of the antisense strand. In another embodiment, the antisense strand comprises a glyceryl modification at the 3′ end. In another embodiment, the 5′-end of the antisense strand optionally includes a phosphate group.


In any of the above-described embodiments of a double-stranded short interfering nucleic acid (siNA) molecule that inhibits expression of a HCV gene, wherein a majority of the pyrimidine nucleotides present in the double-stranded siNA molecule comprises a sugar modification, each of the two strands of the siNA molecule can comprise about 15 to about 30 or more (e.g., about 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30 or more) nucleotides. In one embodiment, about 15 to about 30 or more (e.g., about 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30 or more) nucleotides of each strand of the siNA molecule are base-paired to the complementary nucleotides of the other strand of the siNA molecule. In another embodiment, about 15 to about 30 or more (e.g., about 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30 or more) nucleotides of each strand of the siNA molecule are base-paired to the complementary nucleotides of the other strand of the siNA molecule, wherein at least two 3′ terminal nucleotides of each strand of the siNA molecule are not base-paired to the nucleotides of the other strand of the siNA molecule. In another embodiment, each of the two 3′ terminal nucleotides of each fragment of the siNA molecule is a 2′-deoxy-pyrimidine, such as 2′-deoxy-thymidine. In one embodiment, each strand of the siNA molecule is base-paired to the complementary nucleotides of the other strand of the siNA molecule. In one embodiment, about 15 to about 30 (e.g., about 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30) nucleotides of the antisense strand are base-paired to the nucleotide sequence of the HCV RNA or a portion thereof. In one embodiment, about 18 to about 25 (e.g., about 18, 19, 20, 21, 22, 23, 24, or 25) nucleotides of the antisense strand are base-paired to the nucleotide sequence of the HCV RNA or a portion thereof.


In one embodiment, the invention features a double-stranded short interfering nucleic acid (siNA) molecule that inhibits expression of a HCV gene, wherein one of the strands of the double-stranded siNA molecule is an antisense strand which comprises nucleotide sequence that is complementary to nucleotide sequence of HCV RNA or a portion thereof, the other strand is a sense strand which comprises nucleotide sequence that is complementary to a nucleotide sequence of the antisense strand and wherein a majority of the pyrimidine nucleotides present in the double-stranded siNA molecule comprises a sugar modification, and wherein the 5′-end of the antisense strand optionally includes a phosphate group.


In one embodiment, the invention features a double-stranded short interfering nucleic acid (siNA) molecule that inhibits expression of a HCV gene, wherein one of the strands of the double-stranded siNA molecule is an antisense strand which comprises nucleotide sequence that is complementary to nucleotide sequence of HCV RNA or a portion thereof, the other strand is a sense strand which comprises nucleotide sequence that is complementary to a nucleotide sequence of the antisense strand and wherein a majority of the pyrimidine nucleotides present in the double-stranded siNA molecule comprises a sugar modification, and wherein the nucleotide sequence or a portion thereof of the antisense strand is complementary to a nucleotide sequence of the untranslated region or a portion thereof of the HCV RNA.


In one embodiment, the invention features a double-stranded short interfering nucleic acid (siNA) molecule that inhibits expression of a HCV gene, wherein one of the strands of the double-stranded siNA molecule is an antisense strand which comprises nucleotide sequence that is complementary to nucleotide sequence of HCV RNA or a portion thereof, wherein the other strand is a sense strand which comprises nucleotide sequence that is complementary to a nucleotide sequence of the antisense strand, wherein a majority of the pyrimidine nucleotides present in the double-stranded siNA molecule comprises a sugar modification, and wherein the nucleotide sequence of the antisense strand is complementary to a nucleotide sequence of the HCV RNA or a portion thereof that is present in the HCV RNA.


In one embodiment, the invention features a composition comprising a siNA molecule of the invention in a pharmaceutically acceptable carrier or diluent.


In a non-limiting example, the introduction of chemically-modified nucleotides into nucleic acid molecules provides a powerful tool in overcoming potential limitations of in vivo stability and bioavailability inherent to native RNA molecules that are delivered exogenously. For example, the use of chemically-modified nucleic acid molecules can enable a lower dose of a particular nucleic acid molecule for a given therapeutic effect since chemically-modified nucleic acid molecules tend to have a longer half-life in serum. Furthermore, certain chemical modifications can improve the bioavailability of nucleic acid molecules by targeting particular cells or tissues and/or improving cellular uptake of the nucleic acid molecule. Therefore, even if the activity of a chemically-modified nucleic acid molecule is reduced as compared to a native nucleic acid molecule, for example, when compared to an all-RNA nucleic acid molecule, the overall activity of the modified nucleic acid molecule can be greater than that of the native molecule due to improved stability and/or delivery of the molecule. Unlike native unmodified siNA, chemically-modified siNA can also minimize the possibility of activating interferon activity in humans.


In any of the embodiments of siNA molecules described herein, the antisense region of a siNA molecule of the invention can comprise a phosphorothioate internucleotide linkage at the 3′-end of said antisense region. In any of the embodiments of siNA molecules described herein, the antisense region can comprise about one to about five phosphorothioate internucleotide linkages at the 5′-end of said antisense region. In any of the embodiments of siNA molecules described herein, the 3′-terminal nucleotide overhangs of a siNA molecule of the invention can comprise ribonucleotides or deoxyribonucleotides that are chemically-modified at a nucleic acid sugar, base, or backbone. In any of the embodiments of siNA molecules described herein, the 3′-terminal nucleotide overhangs can comprise one or more universal base ribonucleotides. In any of the embodiments of siNA molecules described herein, the 3′-terminal nucleotide overhangs can comprise one or more acyclic nucleotides.


One embodiment of the invention provides an expression vector comprising a nucleic acid sequence encoding at least one siNA molecule of the invention in a manner that allows expression of the nucleic acid molecule. Another embodiment of the invention provides a mammalian cell comprising such an expression vector. The mammalian cell can be a human cell. The siNA molecule of the expression vector can comprise a sense region and an antisense region. The antisense region can comprise sequence complementary to a RNA or DNA sequence encoding HCV and the sense region can comprise sequence complementary to the antisense region. The siNA molecule can comprise two distinct strands having complementary sense and antisense regions. The siNA molecule can comprise a single strand having complementary sense and antisense regions.


In one embodiment, the invention features a chemically-modified short interfering nucleic acid (siNA) molecule capable of mediating RNA interference (RNAi) against HCV inside a cell or reconstituted in vitro system, wherein the chemical modification comprises one or more (e.g., about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or more) nucleotides comprising a backbone modified internucleotide linkage having Formula I:
embedded image

    • wherein each R1 and R2 is independently any nucleotide, non-nucleotide, or polynucleotide which can be naturally-occurring or chemically-modified, each X and Y is independently O, S, N, alkyl, or substituted alkyl, each Z and W is independently O, S, N, alkyl, substituted alkyl, O-alkyl, S-alkyl, alkaryl, aralkyl, or acetyl and wherein W, X, Y, and Z are optionally not all O. In another embodiment, a backbone modification of the invention comprises a phosphonoacetate and/or thiophosphonoacetate internucleotide linkage (see for example Sheehan et al., 2003, Nucleic Acids Research, 31, 4109-4118).


The chemically-modified internucleotide linkages having Formula I, for example, wherein any Z, W, X, and/or Y independently comprises a sulphur atom, can be present in one or both oligonucleotide strands of the siNA duplex, for example, in the sense strand, the antisense strand, or both strands. The siNA molecules of the invention can comprise one or more (e.g., about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or more) chemically-modified internucleotide linkages having Formula I at the 3′-end, the 5′-end, or both of the 3′ and 5′-ends of the sense strand, the antisense strand, or both strands. For example, an exemplary siNA molecule of the invention can comprise about 1 to about 5 or more (e.g., about 1, 2, 3, 4, 5, or more) chemically-modified internucleotide linkages having Formula I at the 5′-end of the sense strand, the antisense strand, or both strands. In another non-limiting example, an exemplary siNA molecule of the invention can comprise one or more (e.g., about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or more) pyrimidine nucleotides with chemically-modified internucleotide linkages having Formula I in the sense strand, the antisense strand, or both strands. In yet another non-limiting example, an exemplary siNA molecule of the invention can comprise one or more (e.g., about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or more) purine nucleotides with chemically-modified internucleotide linkages having Formula I in the sense strand, the antisense strand, or both strands. In another embodiment, a siNA molecule of the invention having internucleotide linkage(s) of Formula I also comprises a chemically-modified nucleotide or non-nucleotide having any of Formulae I-VII.


In one embodiment, the invention features a chemically-modified short interfering nucleic acid (siNA) molecule capable of mediating RNA interference (RNAi) against HCV inside a cell or reconstituted in vitro system, wherein the chemical modification comprises one or more (e.g., about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or more) nucleotides or non-nucleotides having Formula II:
embedded image

wherein each R3, R4, R5, R6, R7, R8, R10, R11 and R12 is independently H, OH, alkyl, substituted alkyl, alkaryl or aralkyl, F, Cl, Br, CN, CF3, OCF3, OCN, O-alkyl, S-alkyl, N-alkyl, O-alkenyl, S-alkenyl, N-alkenyl, SO-alkyl, alkyl-OSH, alkyl-OH, O-alkyl-OH, O-alkyl-SH, S-alkyl-OH, S-alkyl-SH, alkyl-S-alkyl, alkyl-O-alkyl, ONO2, NO2, N3, NH2, aminoalkyl, aminoacid, aminoacyl, ONH2, O-aminoalkyl, O-aminoacid, O-aminoacyl, heterocycloalkyl, heterocycloalkaryl, aminoalkylamino, polyalklylamino, substituted silyl, or group having Formula I or II; R9 is O, S, CH2, S═O, CHF, or CF2, and B is a nucleosidic base such as adenine, guanine, uracil, cytosine, thymine, 2-aminoadenosine, 5-methylcytosine, 2,6-diaminopurine, or any other non-naturally occurring base that can be complementary or non-complementary to target RNA or a non-nucleosidic base such as phenyl, naphthyl, 3-nitropyrrole, 5-nitroindole, nebularine, pyridone, pyridinone, or any other non-naturally occurring universal base that can be complementary or non-complementary to target RNA.


The chemically-modified nucleotide or non-nucleotide of Formula II can be present in one or both oligonucleotide strands of the siNA duplex, for example in the sense strand, the antisense strand, or both strands. The siNA molecules of the invention can comprise one or more chemically-modified nucleotides or non-nucleotides of Formula II at the 3′-end, the 5′-end, or both of the 3′ and 5′-ends of the sense strand, the antisense strand, or both strands. For example, an exemplary siNA molecule of the invention can comprise about 1 to about 5 or more (e.g., about 1, 2, 3, 4, 5, or more) chemically-modified nucleotides or non-nucleotides of Formula II at the 5′-end of the sense strand, the antisense strand, or both strands. In anther non-limiting example, an exemplary siNA molecule of the invention can comprise about 1 to about 5 or more (e.g., about 1, 2, 3, 4, 5, or more) chemically-modified nucleotides or non-nucleotides of Formula II at the 3′-end of the sense strand, the antisense strand, or both strands.


In one embodiment, the invention features a chemically-modified short interfering nucleic acid (siNA) molecule capable of mediating RNA interference (RNAi) against HCV inside a cell or reconstituted in vitro system, wherein the chemical modification comprises one or more (e.g., about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or more) nucleotides or non-nucleotides having Formula III:
embedded image

wherein each R3, R4, R5, R6, R7, R8, R10, R11 and R12 is independently H, OH, alkyl, substituted alkyl, alkaryl or aralkyl, F, Cl, Br, CN, CF3, OCF3, OCN, O-alkyl, S-alkyl, N-alkyl, O-alkenyl, S-alkenyl, N-alkenyl, SO-alkyl, alkyl-OSH, alkyl-OH, O-alkyl-OH, O-alkyl-SH, S-alkyl-OH, S-alkyl-SH, alkyl-S-alkyl, alkyl-O-alkyl, ONO2, NO2, N3, NH2, aminoalkyl, aminoacid, aminoacyl, ONH2, O-aminoalkyl, O-aminoacid, O-aminoacyl, heterocycloalkyl, heterocycloalkaryl, aminoalkylamino, polyalklylamino, substituted silyl, or group having Formula I or II; R9 is O, S, CH2, S═O, CHF, or CF2, and B is a nucleosidic base such as adenine, guanine, uracil, cytosine, thymine, 2-aminoadenosine, 5-methylcytosine, 2,6-diaminopurine, or any other non-naturally occurring base that can be employed to be complementary or non-complementary to target RNA or a non-nucleosidic base such as phenyl, naphthyl, 3-nitropyrrole, 5-nitroindole, nebularine, pyridone, pyridinone, or any other non-naturally occurring universal base that can be complementary or non-complementary to target RNA.


The chemically-modified nucleotide or non-nucleotide of Formula III can be present in one or both oligonucleotide strands of the siNA duplex, for example, in the sense strand, the antisense strand, or both strands. The siNA molecules of the invention can comprise one or more chemically-modified nucleotides or non-nucleotides of Formula III at the 3′-end, the 5′-end, or both of the 3′ and 5′-ends of the sense strand, the antisense strand, or both strands. For example, an exemplary siNA molecule of the invention can comprise about 1 to about 5 or more (e.g., about 1, 2, 3, 4, 5, or more) chemically-modified nucleotide(s) or non-nucleotide(s) of Formula III at the 5′-end of the sense strand, the antisense strand, or both strands. In anther non-limiting example, an exemplary siNA molecule of the invention can comprise about 1 to about 5 or more (e.g., about 1, 2, 3, 4, 5, or more) chemically-modified nucleotide or non-nucleotide of Formula III at the 3′-end of the sense strand, the antisense strand, or both strands.


In another embodiment, a siNA molecule of the invention comprises a nucleotide having Formula II or III, wherein the nucleotide having Formula II or III is in an inverted configuration. For example, the nucleotide having Formula II or III is connected to the siNA construct in a 3′-3′,3′-2′,2′-3′, or 5′-5′ configuration, such as at the 3′-end, the 5′-end, or both of the 3′ and 5′-ends of one or both siNA strands.


In one embodiment, the invention features a chemically-modified short interfering nucleic acid (siNA) molecule capable of mediating RNA interference (RNAi) against HCV inside a cell or reconstituted in vitro system, wherein the chemical modification comprises a 5′-terminal phosphate group having Formula IV:
embedded image

wherein each X and Y is independently O, S, N, alkyl, substituted alkyl, or alkylhalo; wherein each Z and W is independently O, S, N, alkyl, substituted alkyl, O-alkyl, S-alkyl, alkaryl, aralkyl, alkylhalo, or acetyl; and wherein W, X, Y and Z are not all 0.


In one embodiment, the invention features a siNA molecule having a 5′-terminal phosphate group having Formula IV on the target-complementary strand, for example, a strand complementary to a target RNA, wherein the siNA molecule comprises an all RNA siNA molecule. In another embodiment, the invention features a siNA molecule having a 5′-terminal phosphate group having Formula IV on the target-complementary strand wherein the siNA molecule also comprises about 1 to about 3 (e.g., about 1, 2, or 3) nucleotide 3′-terminal nucleotide overhangs having about 1 to about 4 (e.g., about 1, 2, 3, or 4) deoxyribonucleotides on the 3′-end of one or both strands. In another embodiment, a 5′-terminal phosphate group having Formula IV is present on the target-complementary strand of a siNA molecule of the invention, for example a siNA molecule having chemical modifications having any of Formulae I-VII.


In one embodiment, the invention features a chemically-modified short interfering nucleic acid (siNA) molecule capable of mediating RNA interference (RNAi) against HCV inside a cell or reconstituted in vitro system, wherein the chemical modification comprises one or more phosphorothioate internucleotide linkages. For example, in a non-limiting example, the invention features a chemically-modified short interfering nucleic acid (siNA) having about 1, 2, 3, 4, 5, 6, 7, 8 or more phosphorothioate internucleotide linkages in one siNA strand. In yet another embodiment, the invention features a chemically-modified short interfering nucleic acid (siNA) individually having about 1, 2, 3, 4, 5, 6, 7, 8 or more phosphorothioate internucleotide linkages in both siNA strands. The phosphorothioate internucleotide linkages can be present in one or both oligonucleotide strands of the siNA duplex, for example in the sense strand, the antisense strand, or both strands. The siNA molecules of the invention can comprise one or more phosphorothioate internucleotide linkages at the 3′-end, the 5′-end, or both of the 3′- and 5′-ends of the sense strand, the antisense strand, or both strands. For example, an exemplary siNA molecule of the invention can comprise about 1 to about 5 or more (e.g., about 1, 2, 3, 4, 5, or more) consecutive phosphorothioate internucleotide linkages at the 5′-end of the sense strand, the antisense strand, or both strands. In another non-limiting example, an exemplary siNA molecule of the invention can comprise one or more (e.g., about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or more) pyrimidine phosphorothioate internucleotide linkages in the sense strand, the antisense strand, or both strands. In yet another non-limiting example, an exemplary siNA molecule of the invention can comprise one or more (e.g., about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or more) purine phosphorothioate internucleotide linkages in the sense strand, the antisense strand, or both strands.


In one embodiment, the invention features a siNA molecule, wherein the sense strand comprises one or more, for example, about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or more phosphorothioate internucleotide linkages, and/or one or more (e.g., about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 or more) 2′-deoxy, 2′-O-methyl, 2′-deoxy-2′-fluoro, and/or about one or more (e.g., about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 or more) universal base modified nucleotides, and optionally a terminal cap molecule at the 3′-end, the 5′-end, or both of the 3′- and 5′-ends of the sense strand; and wherein the antisense strand comprises about 1 to about 10 or more, specifically about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or more phosphorothioate internucleotide linkages, and/or one or more (e.g., about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 or more) 2′-deoxy, 2′-O-methyl, 2′-deoxy-2′-fluoro, and/or one or more (e.g., about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 or more) universal base modified nucleotides, and optionally a terminal cap molecule at the 3′-end, the 5′-end, or both of the 3′- and 5′-ends of the antisense strand. In another embodiment, one or more, for example about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or more, pyrimidine nucleotides of the sense and/or antisense siNA strand are chemically-modified with 2′-deoxy, 2′-O-methyl and/or 2′-deoxy-2′-fluoro nucleotides, with or without one or more, for example about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or more, phosphorothioate internucleotide linkages and/or a terminal cap molecule at the 3′-end, the 5′-end, or both of the 3′- and 5′-ends, being present in the same or different strand.


In another embodiment, the invention features a siNA molecule, wherein the sense strand comprises about 1 to about 5, specifically about 1, 2, 3, 4, or 5 phosphorothioate internucleotide linkages, and/or one or more (e.g., about 1, 2, 3, 4, 5, or more) 2′-deoxy, 2′-O-methyl, 2′-deoxy-2′-fluoro, and/or one or more (e.g., about 1, 2, 3, 4, 5, or more) universal base modified nucleotides, and optionally a terminal cap molecule at the 3-end, the 5′-end, or both of the 3′- and 5′-ends of the sense strand; and wherein the antisense strand comprises about 1 to about 5 or more, specifically about 1, 2, 3, 4, 5, or more phosphorothioate internucleotide linkages, and/or one or more (e.g., about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 or more) 2′-deoxy, 2′-O-methyl, 2′-deoxy-2′-fluoro, and/or one or more (e.g., about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 or more) universal base modified nucleotides, and optionally a terminal cap molecule at the 3′-end, the 5′-end, or both of the 3′- and 5′-ends of the antisense strand. In another embodiment, one or more, for example about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or more, pyrimidine nucleotides of the sense and/or antisense siNA strand are chemically-modified with 2′-deoxy, 2′-O-methyl and/or 2′-deoxy-2′-fluoro nucleotides, with or without about 1 to about 5 or more, for example about 1, 2, 3, 4, 5, or more phosphorothioate internucleotide linkages and/or a terminal cap molecule at the 3′-end, the 5′-end, or both of the 3′- and 5′-ends, being present in the same or different strand.


In one embodiment, the invention features a siNA molecule, wherein the antisense strand comprises one or more, for example, about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or more phosphorothioate internucleotide linkages, and/or about one or more (e.g., about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 or more) 2′-deoxy, 2′-O-methyl, 2′-deoxy-2′-fluoro, and/or one or more (e.g., about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 or more) universal base modified nucleotides, and optionally a terminal cap molecule at the 3′-end, the 5′-end, or both of the 3′- and 5′-ends of the sense strand; and wherein the antisense strand comprises about 1 to about 10 or more, specifically about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 or more phosphorothioate internucleotide linkages, and/or one or more (e.g., about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 or more) 2′-deoxy, 2′-O-methyl, 2′-deoxy-2′-fluoro, and/or one or more (e.g., about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 or more) universal base modified nucleotides, and optionally a terminal cap molecule at the 3′-end, the 5′-end, or both of the 3′- and 5′-ends of the antisense strand. In another embodiment, one or more, for example about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 or more pyrimidine nucleotides of the sense and/or antisense siNA strand are chemically-modified with 2′-deoxy, 2′-O-methyl and/or 2′-deoxy-2′-fluoro nucleotides, with or without one or more, for example, about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 or more phosphorothioate internucleotide linkages and/or a terminal cap molecule at the 3′-end, the 5′-end, or both of the 3′ and 5′-ends, being present in the same or different strand.


In another embodiment, the invention features a siNA molecule, wherein the antisense strand comprises about 1 to about 5 or more, specifically about 1, 2, 3, 4, 5 or more phosphorothioate internucleotide linkages, and/or one or more (e.g., about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 or more) 2′-deoxy, 2′-O-methyl, 2′-deoxy-2′-fluoro, and/or one or more (e.g., about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 or more) universal base modified nucleotides, and optionally a terminal cap molecule at the 3′-end, the 5′-end, or both of the 3′- and 5′-ends of the sense strand; and wherein the antisense strand comprises about 1 to about 5 or more, specifically about 1, 2, 3, 4, 5 or more phosphorothioate internucleotide linkages, and/or one or more (e.g., about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 or more) 2′-deoxy, 2′-O-methyl, 2′-deoxy-2′-fluoro, and/or one or more (e.g., about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 or more) universal base modified nucleotides, and optionally a terminal cap molecule at the 3′-end, the 5′-end, or both of the 3′- and 5′-ends of the antisense strand. In another embodiment, one or more, for example about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 or more pyrimidine nucleotides of the sense and/or antisense siNA strand are chemically-modified with 2′-deoxy, 2′-O-methyl and/or 2′-deoxy-2′-fluoro nucleotides, with or without about 1 to about 5, for example about 1, 2, 3, 4, 5 or more phosphorothioate internucleotide linkages and/or a terminal cap molecule at the 3′-end, the 5′-end, or both of the 3′- and 5′-ends, being present in the same or different strand.


In one embodiment, the invention features a chemically-modified short interfering nucleic acid (siNA) molecule having about 1 to about 5 or more (specifically about 1, 2, 3, 4, 5 or more) phosphorothioate internucleotide linkages in each strand of the siNA molecule.


In another embodiment, the invention features a siNA molecule comprising 2′-5′ internucleotide linkages. The 2′-5′ internucleotide linkage(s) can be at the 3′-end, the 5′-end, or both of the 3′- and 5′-ends of one or both siNA sequence strands. In addition, the 2′-5′ internucleotide linkage(s) can be present at various other positions within one or both siNA sequence strands, for example, about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or more including every internucleotide linkage of a pyrimidine nucleotide in one or both strands of the siNA molecule can comprise a 2′-5′ internucleotide linkage, or about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or more including every internucleotide linkage of a purine nucleotide in one or both strands of the siNA molecule can comprise a 2′-5′ internucleotide linkage.


In another embodiment, a chemically-modified siNA molecule of the invention comprises a duplex having two strands, one or both of which can be chemically-modified, wherein each strand is independently about 15 to about 30 (e.g., about 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30) nucleotides in length, wherein the duplex has about 15 to about 30 (e.g., about 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30) base pairs, and wherein the chemical modification comprises a structure having any of Formulae I-VII. For example, an exemplary chemically-modified siNA molecule of the invention comprises a duplex having two strands, one or both of which can be chemically-modified with a chemical modification having any of Formulae I-VII or any combination thereof, wherein each strand consists of about 21 nucleotides, each having a 2-nucleotide 3′-terminal nucleotide overhang, and wherein the duplex has about 19 base pairs. In another embodiment, a siNA molecule of the invention comprises a single stranded hairpin structure, wherein the siNA is about 36 to about 70 (e.g., about 36, 40, 45, 50, 55, 60, 65, or 70) nucleotides in length having about 15 to about 30 (e.g., about 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30) base pairs, and wherein the siNA can include a chemical modification comprising a structure having any of Formulae I-VII or any combination thereof. For example, an exemplary chemically-modified siNA molecule of the invention comprises a linear oligonucleotide having about 42 to about 50 (e.g., about 42, 43, 44, 45, 46, 47, 48, 49, or 50) nucleotides that is chemically-modified with a chemical modification having any of Formulae I-VII or any combination thereof, wherein the linear oligonucleotide forms a hairpin structure having about 19 to about 21 (e.g., 19, 20, or 21) base pairs and a 2-nucleotide 3′-terminal nucleotide overhang. In another embodiment, a linear hairpin siNA molecule of the invention contains a stem loop motif, wherein the loop portion of the siNA molecule is biodegradable. For example, a linear hairpin siNA molecule of the invention is designed such that degradation of the loop portion of the siNA molecule in vivo can generate a double-stranded siNA molecule with 3′-terminal overhangs, such as 3′-terminal nucleotide overhangs comprising about 2 nucleotides.


In another embodiment, a siNA molecule of the invention comprises a hairpin structure, wherein the siNA is about 25 to about 50 (e.g., about 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, or 50) nucleotides in length having about 3 to about 25 (e.g., about 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, or 25) base pairs, and wherein the siNA can include one or more chemical modifications comprising a structure having any of Formulae I-VII or any combination thereof. For example, an exemplary chemically-modified siNA molecule of the invention comprises a linear oligonucleotide having about 25 to about 35 (e.g., about 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, or 35) nucleotides that is chemically-modified with one or more chemical modifications having any of Formulae I-VII or any combination thereof, wherein the linear oligonucleotide forms a hairpin structure having about 3 to about 25 (e.g., about 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, or 25) base pairs and a 5′-terminal phosphate group that can be chemically modified as described herein (for example a 5′-terminal phosphate group having Formula IV). In another embodiment, a linear hairpin siNA molecule of the invention contains a stem loop motif, wherein the loop portion of the siNA molecule is biodegradable. In one embodiment, a linear hairpin siNA molecule of the invention comprises a loop portion comprising a non-nucleotide linker.


In another embodiment, a siNA molecule of the invention comprises an asymmetric hairpin structure, wherein the siNA is about 25 to about 50 (e.g., about 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, or 50) nucleotides in length having about 3 to about 25 (e.g., about 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, or 25) base pairs, and wherein the siNA can include one or more chemical modifications comprising a structure having any of Formulae I-VII or any combination thereof. For example, an exemplary chemically-modified siNA molecule of the invention comprises a linear oligonucleotide having about 25 to about 35 (e.g., about 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, or 35) nucleotides that is chemically-modified with one or more chemical modifications having any of Formulae I-VII or any combination thereof, wherein the linear oligonucleotide forms an asymmetric hairpin structure having about 3 to about 25 (e.g., about 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, or 25) base pairs and a 5′-terminal phosphate group that can be chemically modified as described herein (for example a 5′-terminal phosphate group having Formula IV). In one embodiment, an asymmetric hairpin siNA molecule of the invention contains a stem loop motif, wherein the loop portion of the siNA molecule is biodegradable. In another embodiment, an asymmetric hairpin siNA molecule of the invention comprises a loop portion comprising a non-nucleotide linker.


In another embodiment, a siNA molecule of the invention comprises an asymmetric double stranded structure having separate polynucleotide strands comprising sense and antisense regions, wherein the antisense region is about 15 to about 30 (e.g., about 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30) nucleotides in length, wherein the sense region is about 3 to about 25 (e.g., about 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, or 25) nucleotides in length, wherein the sense region and the antisense region have at least 3 complementary nucleotides, and wherein the siNA can include one or more chemical modifications comprising a structure having any of Formulae I-VII or any combination thereof. For example, an exemplary chemically-modified siNA molecule of the invention comprises an asymmetric double stranded structure having separate polynucleotide strands comprising sense and antisense regions, wherein the antisense region is about 18 to about 23 (e.g., about 18, 19, 20, 21, 22, or 23) nucleotides in length and wherein the sense region is about 3 to about 15 (e.g., about 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, or 15) nucleotides in length, wherein the sense region the antisense region have at least 3 complementary nucleotides, and wherein the siNA can include one or more chemical modifications comprising a structure having any of Formulae I-VII or any combination thereof. In another embodiment, the asymmetric double stranded siNA molecule can also have a 5′-terminal phosphate group that can be chemically modified as described herein (for example a 5′-terminal phosphate group having Formula IV).


In another embodiment, a siNA molecule of the invention comprises a circular nucleic acid molecule, wherein the siNA is about 38 to about 70 (e.g., about 38, 40, 45, 50, 55, 60, 65, or 70) nucleotides in length having about 15 to about 30 (e.g., about 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30) base pairs, and wherein the siNA can include a chemical modification, which comprises a structure having any of Formulae I-VII or any combination thereof. For example, an exemplary chemically-modified siNA molecule of the invention comprises a circular oligonucleotide having about 42 to about 50 (e.g., about 42, 43, 44, 45, 46, 47, 48, 49, or 50) nucleotides that is chemically-modified with a chemical modification having any of Formulae I-VII or any combination thereof, wherein the circular oligonucleotide forms a dumbbell shaped structure having about 19 base pairs and 2 loops.


In another embodiment, a circular siNA molecule of the invention contains two loop motifs, wherein one or both loop portions of the siNA molecule is biodegradable. For example, a circular siNA molecule of the invention is designed such that degradation of the loop portions of the siNA molecule in vivo can generate a double-stranded siNA molecule with 3′-terminal overhangs, such as 3′-terminal nucleotide overhangs comprising about 2 nucleotides.


In one embodiment, a siNA molecule of the invention comprises at least one (e.g., about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or more) abasic moiety, for example a compound having Formula V:
embedded image

wherein each R3, R4, R5, R6, R7, R8, R10, R11, R12, and R13 is independently H, OH, alkyl, substituted alkyl, alkaryl or aralkyl, F, Cl, Br, CN, CF3, OCF3, OCN, O-alkyl, S-alkyl, N-alkyl, O-alkenyl, S-alkenyl, N-alkenyl, SO-alkyl, alkyl-OSH, alkyl-OH, O-alkyl-OH, O-alkyl-SH, S-alkyl-OH, S-alkyl-SH, alkyl-S-alkyl, alkyl-O-alkyl, ONO2, NO2, N3, NH2, aminoalkyl, aminoacid, aminoacyl, ONH2, O-aminoalkyl, O-aminoacid, O-aminoacyl, heterocycloalkyl, heterocycloalkaryl, aminoalkylamino, polyalklylamino, substituted silyl, or group having Formula I or II; R9 is O, S, CH2, S═O, CHF, or CF2.


In one embodiment, a siNA molecule of the invention comprises at least one (e.g., about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or more) inverted abasic moiety, for example a compound having Formula VI:
embedded image

wherein each R3, R4, R5, R6, R7, R8, R10, R11, R12, and R13 is independently H, OH, alkyl, substituted alkyl, alkaryl or aralkyl, F, Cl, Br, CN, CF3, OCF3, OCN, O-alkyl, S-alkyl, N-alkyl, O-alkenyl, S-alkenyl, N-alkenyl, SO-alkyl, alkyl-OSH, alkyl-OH, O-alkyl-OH, O-alkyl-SH, S-alkyl-OH, S-alkyl-SH, alkyl-S-alkyl, alkyl-O-alkyl, ONO2, NO2, N3, NH2, aminoalkyl, aminoacid, aminoacyl, ONH2, O-aminoalkyl, O-aminoacid, O-aminoacyl, heterocycloalkyl, heterocycloalkaryl, aminoalkylamino, polyalklylamino, substituted silyl, or group having Formula I or II; R9 is O, S, CH2, S═O, CHF, or CF2, and either R2, R3, R8 or R1 3 serve as points of attachment to the siNA molecule of the invention.


In another embodiment, a siNA molecule of the invention comprises at least one (e.g., about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or more) substituted polyalkyl moieties, for example a compound having Formula VII:
embedded image

wherein each n is independently an integer from 1 to 12, each R1, R2 and R3 is independently H, OH, alkyl, substituted alkyl, alkaryl or aralkyl, F, Cl, Br, CN, CF3, OCF3, OCN, O-alkyl, S-alkyl, N-alkyl, O-alkenyl, S-alkenyl, N-alkenyl, SO-alkyl, alkyl-OSH, alkyl-OH, O-alkyl-OH, O-alkyl-SH, S-alkyl-OH, S-alkyl-SH, alkyl-S-alkyl, alkyl-O-alkyl, ONO2, NO2, N3, NH2, aminoalkyl, aminoacid, aminoacyl, ONH2, O-aminoalkyl, O-aminoacid, O-aminoacyl, heterocycloalkyl, heterocycloalkaryl, aminoalkylamino, polyalklylamino, substituted silyl, or a group having Formula I, and R1, R2 or R3 serves as points of attachment to the siNA molecule of the invention.


In another embodiment, the invention features a compound having Formula VII, wherein R1 and R2 are hydroxyl (OH) groups, n=1, and R3 comprises 0 and is the point of attachment to the 3′-end, the 5′-end, or both of the 3′ and 5′-ends of one or both strands of a double-stranded siNA molecule of the invention or to a single-stranded siNA molecule of the invention. This modification is referred to herein as “glyceryl” (for example modification 6 in FIG. 10).


In another embodiment, a chemically modified nucleoside or non-nucleoside (e.g. a moiety having any of Formula V, VI or VII) of the invention is at the 3′-end, the 5′-end, or both of the 3′ and 5′-ends of a siNA molecule of the invention. For example, chemically modified nucleoside or non-nucleoside (e.g., a moiety having Formula V, VI or VII) can be present at the 3′-end, the 5′-end, or both of the 3′ and 5′-ends of the antisense strand, the sense strand, or both antisense and sense strands of the siNA molecule. In one embodiment, the chemically modified nucleoside or non-nucleoside (e.g., a moiety having Formula V, VI or VII) is present at the 5′-end and 3′-end of the sense strand and the 3′-end of the antisense strand of a double stranded siNA molecule of the invention. In one embodiment, the chemically modified nucleoside or non-nucleoside (e.g., a moiety having Formula V, VI or VII) is present at the terminal position of the 5′-end and 3′-end of the sense strand and the 3′-end of the antisense strand of a double stranded siNA molecule of the invention. In one embodiment, the chemically modified nucleoside or non-nucleoside (e.g., a moiety having Formula V, VI or VII) is present at the two terminal positions of the 5′-end and 3′-end of the sense strand and the 3′-end of the antisense strand of a double stranded siNA molecule of the invention. In one embodiment, the chemically modified nucleoside or non-nucleoside (e.g., a moiety having Formula V, VI or VII) is present at the penultimate position of the 5′-end and 3′-end of the sense strand and the 3′-end of the antisense strand of a double stranded siNA molecule of the invention. In addition, a moiety having Formula VII can be present at the 3′-end or the 5′-end of a hairpin siNA molecule as described herein.


In another embodiment, a siNA molecule of the invention comprises an abasic residue having Formula V or VI, wherein the abasic residue having Formula VI or VI is connected to the siNA construct in a 3′-3′,3′-2′,2′-3′, or 5′-5′ configuration, such as at the 3′-end, the 5′-end, or both of the 3′ and 5′-ends of one or both siNA strands.


In one embodiment, a siNA molecule of the invention comprises one or more (e.g., about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or more) locked nucleic acid (LNA) nucleotides, for example, at the 5′-end, the 3′-end, both of the 5′ and 3′-ends, or any combination thereof, of the siNA molecule.


In another embodiment, a siNA molecule of the invention comprises one or more (e.g., about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or more) acyclic nucleotides, for example, at the 5′-end, the 3′-end, both of the S′ and 3′-ends, or any combination thereof, of the siNA molecule.


In one embodiment, the invention features a chemically-modified short interfering nucleic acid (siNA) molecule of the invention comprising a sense region, wherein any (e.g., one or more or all) pyrimidine nucleotides present in the sense region are 2′-deoxy-2′-fluoro pyrimidine nucleotides (e.g., wherein all pyrimidine nucleotides are 2′-deoxy-2′-fluoro pyrimidine nucleotides or alternately a plurality of pyrimidine nucleotides are 2′-deoxy-2′-fluoro pyrimidine nucleotides), and wherein any (e.g., one or more or all) purine nucleotides present in the sense region are 2′-deoxy purine nucleotides (e.g., wherein all purine nucleotides are 2′-deoxy purine nucleotides or alternately a plurality of purine nucleotides are 2′-deoxy purine nucleotides).


In one embodiment, the invention features a chemically-modified short interfering nucleic acid (siNA) molecule of the invention comprising a sense region, wherein any (e.g., one or more or all) pyrimidine nucleotides present in the sense region are 2′-deoxy-2′-fluoro pyrimidine nucleotides (e.g., wherein all pyrimidine nucleotides are 2′-deoxy-2′-fluoro pyrimidine nucleotides or alternately a plurality of pyrimidine nucleotides are 2′-deoxy-2′-fluoro pyrimidine nucleotides), and wherein any (e.g., one or more or all) purine nucleotides present in the sense region are 2′-deoxy purine nucleotides (e.g., wherein all purine nucleotides are 2′-deoxy purine nucleotides or alternately a plurality of purine nucleotides are 2′-deoxy purine nucleotides), wherein any nucleotides comprising a 3′-terminal nucleotide overhang that are present in said sense region are 2′-deoxy nucleotides.


In one embodiment, the invention features a chemically-modified short interfering nucleic acid (siNA) molecule of the invention comprising a sense region, wherein any (e.g., one or more or all) pyrimidine nucleotides present in the sense region are 2′-deoxy-2′-fluoro pyrimidine nucleotides (e.g., wherein all pyrimidine nucleotides are 2′-deoxy-2′-fluoro pyrimidine nucleotides or alternately a plurality of pyrimidine nucleotides are 2′-deoxy-2′-fluoro pyrimidine nucleotides), and wherein any (e.g., one or more or all) purine nucleotides present in the sense region are 2′-O-methyl purine nucleotides (e.g., wherein all purine nucleotides are 2′-O-methyl purine nucleotides or alternately a plurality of purine nucleotides are 2′-O-methyl purine nucleotides).


In one embodiment, the invention features a chemically-modified short interfering nucleic acid (siNA) molecule of the invention comprising a sense region, wherein any (e.g., one or more or all) pyrimidine nucleotides present in the sense region are 2′-deoxy-2′-fluoro pyrimidine nucleotides (e.g., wherein all pyrimidine nucleotides are 2′-deoxy-2′-fluoro pyrimidine nucleotides or alternately a plurality of pyrimidine nucleotides are 2′-deoxy-2′-fluoro pyrimidine nucleotides), wherein any (e.g., one or more or all) purine nucleotides present in the sense region are 2′-O-methyl purine nucleotides (e.g., wherein all purine nucleotides are 2′-O-methyl purine nucleotides or alternately a plurality of purine nucleotides are 2′-O-methyl purine nucleotides), and wherein any nucleotides comprising a 3′-terminal nucleotide overhang that are present in said sense region are 2′-deoxy nucleotides.


In one embodiment, the invention features a chemically-modified short interfering nucleic acid (siNA) molecule of the invention comprising an antisense region, wherein any (e.g., one or more or all) pyrimidine nucleotides present in the antisense region are 2′-deoxy-2′-fluoro pyrimidine nucleotides (e.g., wherein all pyrimidine nucleotides are 2′-deoxy-2′-fluoro pyrimidine nucleotides or alternately a plurality of pyrimidine nucleotides are 2′-deoxy-2′-fluoro pyrimidine nucleotides), and wherein any (e.g., one or more or all) purine nucleotides present in the antisense region are 2′-O-methyl purine nucleotides (e.g., wherein all purine nucleotides are 2′-O-methyl purine nucleotides or alternately a plurality of purine nucleotides are 2′-O-methyl purine nucleotides).


In one embodiment, the invention features a chemically-modified short interfering nucleic acid (siNA) molecule of the invention comprising an antisense region, wherein any (e.g., one or more or all) pyrimidine nucleotides present in the antisense region are 2′-deoxy-2′-fluoro pyrimidine nucleotides (e.g., wherein all pyrimidine nucleotides are 2′-deoxy-2′-fluoro pyrimidine nucleotides or alternately a plurality of pyrimidine nucleotides are 2′-deoxy-2′-fluoro pyrimidine nucleotides), wherein any (e.g., one or more or all) purine nucleotides present in the antisense region are 2′-O-methyl purine nucleotides (e.g., wherein all purine nucleotides are 2′-O-methyl purine nucleotides or alternately a plurality of purine nucleotides are 2′-O-methyl purine nucleotides), and wherein any nucleotides comprising a 3′-terminal nucleotide overhang that are present in said antisense region are 2′-deoxy nucleotides.


In one embodiment, the invention features a chemically-modified short interfering nucleic acid (siNA) molecule of the invention comprising an antisense region, wherein any (e.g., one or more or all) pyrimidine nucleotides present in the antisense region are 2′-deoxy-2′-fluoro pyrimidine nucleotides (e.g., wherein all pyrimidine nucleotides are 2′-deoxy-2′-fluoro pyrimidine nucleotides or alternately a plurality of pyrimidine nucleotides are 2′-deoxy-2′-fluoro pyrimidine nucleotides), and wherein any (e.g., one or more or all) purine nucleotides present in the antisense region are 2′-deoxy purine nucleotides (e.g., wherein all purine nucleotides are 2′-deoxy purine nucleotides or alternately a plurality of purine nucleotides are 2′-deoxy purine nucleotides).


In one embodiment, the invention features a chemically-modified short interfering nucleic acid (siNA) molecule of the invention comprising an antisense region, wherein any (e.g., one or more or all) pyrimidine nucleotides present in the antisense region are 2′-deoxy-2′-fluoro pyrimidine nucleotides (e.g., wherein all pyrimidine nucleotides are 2′-deoxy-2′-fluoro pyrimidine nucleotides or alternately a plurality of pyrimidine nucleotides are 2′-deoxy-2′-fluoro pyrimidine nucleotides), and wherein any (e.g., one or more or all) purine nucleotides present in the antisense region are 2′-O-methyl purine nucleotides (e.g., wherein all purine nucleotides are 2′-O-methyl purine nucleotides or alternately a plurality of purine nucleotides are 2′-O-methyl purine nucleotides).


In one embodiment, the invention features a chemically-modified short interfering nucleic acid (siNA) molecule of the invention capable of mediating RNA interference (RNAi) against HCV inside a cell or reconstituted in vitro system comprising a sense region, wherein one or more pyrimidine nucleotides present in the sense region are 2′-deoxy-2′-fluoro pyrimidine nucleotides (e.g., wherein all pyrimidine nucleotides are 2′-deoxy-2′-fluoro pyrimidine nucleotides or alternately a plurality of pyrimidine nucleotides are 2′-deoxy-2′-fluoro pyrimidine nucleotides), and one or more purine nucleotides present in the sense region are 2′-deoxy purine nucleotides (e.g., wherein all purine nucleotides are 2′-deoxy purine nucleotides or alternately a plurality of purine nucleotides are 2′-deoxy purine nucleotides), and an antisense region, wherein one or more pyrimidine nucleotides present in the antisense region are 2′-deoxy-2′-fluoro pyrimidine nucleotides (e.g., wherein all pyrimidine nucleotides are 2′-deoxy-2′-fluoro pyrimidine nucleotides or alternately a plurality of pyrimidine nucleotides are 2′-deoxy-2′-fluoro pyrimidine nucleotides), and one or more purine nucleotides present in the antisense region are 2′-O-methyl purine nucleotides (e.g., wherein all purine nucleotides are 2′-O-methyl purine nucleotides or alternately a plurality of purine nucleotides are 2′-O-methyl purine nucleotides). The sense region and/or the antisense region can have a terminal cap modification, such as any modification described herein or shown in FIG. 10, that is optionally present at the 3′-end, the 5′-end, or both of the 3′ and 5′-ends of the sense and/or antisense sequence. The sense and/or antisense region can optionally further comprise a 3′-terminal nucleotide overhang having about 1 to about 4 (e.g., about 1, 2, 3, or 4) 2′-deoxynucleotides. The overhang nucleotides can further comprise one or more (e.g., about 1, 2, 3, 4 or more) phosphorothioate, phosphonoacetate, and/or thiophosphonoacetate internucleotide linkages. Non-limiting examples of these chemically-modified siNAs are shown in FIGS. 4 and 5 and Tables III and IV herein. In any of these described embodiments, the purine nucleotides present in the sense region are alternatively 2′-O-methyl purine nucleotides (e.g., wherein all purine nucleotides are 2′-O-methyl purine nucleotides or alternately a plurality of purine nucleotides are 2′-O-methyl purine nucleotides) and one or more purine nucleotides present in the antisense region are 2′-O-methyl purine nucleotides (e.g., wherein all purine nucleotides are 2′-O-methyl purine nucleotides or alternately a plurality of purine nucleotides are 2′-O-methyl purine nucleotides). Also, in any of these embodiments, one or more purine nucleotides present in the sense region are alternatively purine ribonucleotides (e.g., wherein all purine nucleotides are purine ribonucleotides or alternately a plurality of purine nucleotides are purine ribonucleotides) and any purine nucleotides present in the antisense region are 2′-O-methyl purine nucleotides (e.g., wherein all purine nucleotides are 2′-O-methyl purine nucleotides or alternately a plurality of purine nucleotides are 2′-O-methyl purine nucleotides). Additionally, in any of these embodiments, one or more purine nucleotides present in the sense region and/or present in the antisense region are alternatively selected from the group consisting of 2′-deoxy nucleotides, locked nucleic acid (LNA) nucleotides, 2′-methoxyethyl nucleotides, 4′-thionucleotides, and 2′-O-methyl nucleotides (e.g., wherein all purine nucleotides are selected from the group consisting of 2′-deoxy nucleotides, locked nucleic acid (LNA) nucleotides, 2′-methoxyethyl nucleotides, 4′-thionucleotides, and 2′-O-methyl nucleotides or alternately a plurality of purine nucleotides are selected from the group consisting of 2′-deoxy nucleotides, locked nucleic acid (LNA) nucleotides, 2′-methoxyethyl nucleotides, 4′-thionucleotides, and 2′-O-methyl nucleotides).


In another embodiment, any modified nucleotides present in the siNA molecules of the invention, preferably in the antisense strand of the siNA molecules of the invention, but also optionally in the sense and/or both antisense and sense strands, comprise modified nucleotides having properties or characteristics similar to naturally occurring ribonucleotides. For example, the invention features siNA molecules including modified nucleotides having a Northern conformation (e.g., Northern pseudorotation cycle, see for example Saenger, Principles of Nucleic Acid Structure, Springer-Verlag ed., 1984). As such, chemically modified nucleotides present in the siNA molecules of the invention, preferably in the antisense strand of the siNA molecules of the invention, but also optionally in the sense and/or both antisense and sense strands, are resistant to nuclease degradation while at the same time maintaining the capacity to mediate RNAi. Non-limiting examples of nucleotides having a northern configuration include locked nucleic acid (LNA) nucleotides (e.g., 2′-O, 4′-C-methylene-(D-ribofuranosyl) nucleotides); 2′-methoxyethoxy (MOE) nucleotides; 2′-methyl-thio-ethyl, 2′-deoxy-2′-fluoro nucleotides, 2′-deoxy-2′-chloro nucleotides, 2′-azido nucleotides, and 2′-O-methyl nucleotides.


In one embodiment, the sense strand of a double stranded siNA molecule of the invention comprises a terminal cap moiety, (see for example FIG. 10) such as an inverted deoxyabaisc moiety, at the 3′-end, 5′-end, or both 3′ and 5′-ends of the sense strand.


In one embodiment, the invention features a chemically-modified short interfering nucleic acid molecule (siNA) capable of mediating RNA interference (RNAi) against HCV inside a cell or reconstituted in vitro system, wherein the chemical modification comprises a conjugate covalently attached to the chemically-modified siNA molecule. Non-limiting examples of conjugates contemplated by the invention include conjugates and ligands described in Vargeese et al., U.S. Ser. No. 10/427,160, filed Apr. 30, 2003, incorporated by reference herein in its entirety, including the drawings. In another embodiment, the conjugate is covalently attached to the chemically-modified siNA molecule via a biodegradable linker. In one embodiment, the conjugate molecule is attached at the 3′-end of either the sense strand, the antisense strand, or both strands of the chemically-modified siNA molecule. In another embodiment, the conjugate molecule is attached at the 5′-end of either the sense strand, the antisense strand, or both strands of the chemically-modified siNA molecule. In yet another embodiment, the conjugate molecule is attached both the 3′-end and 5′-end of either the sense strand, the antisense strand, or both strands of the chemically-modified siNA molecule, or any combination thereof. In one embodiment, a conjugate molecule of the invention comprises a molecule that facilitates delivery of a chemically-modified siNA molecule into a biological system, such as a cell. In another embodiment, the conjugate molecule attached to the chemically-modified siNA molecule is a polyethylene glycol, human serum albumin, or a ligand for a cellular receptor that can mediate cellular uptake. Examples of specific conjugate molecules contemplated by the instant invention that can be attached to chemically-modified siNA molecules are described in Vargeese et al., U.S. Ser. No. 10/201,394, filed Jul. 22, 2002 incorporated by reference herein. The type of conjugates used and the extent of conjugation of siNA molecules of the invention can be evaluated for improved pharmacokinetic profiles, bioavailability, and/or stability of siNA constructs while at the same time maintaining the ability of the siNA to mediate RNAi activity. As such, one skilled in the art can screen siNA constructs that are modified with various conjugates to determine whether the siNA conjugate complex possesses improved properties while maintaining the ability to mediate RNAi, for example in animal models as are generally known in the art.


In one embodiment, the invention features a short interfering nucleic acid (siNA) molecule of the invention, wherein the siNA further comprises a nucleotide, non-nucleotide, or mixed nucleotide/non-nucleotide linker that joins the sense region of the siNA to the antisense region of the siNA. In one embodiment, a nucleotide linker of the invention can be a linker of ≧2 nucleotides in length, for example about 3, 4, 5, 6, 7, 8, 9, or 10 nucleotides in length. In another embodiment, the nucleotide linker can be a nucleic acid aptamer. By “aptamer” or “nucleic acid aptamer” as used herein is meant a nucleic acid molecule that binds specifically to a target molecule wherein the nucleic acid molecule has sequence that comprises a sequence recognized by the target molecule in its natural setting. Alternately, an aptamer can be a nucleic acid molecule that binds to a target molecule where the target molecule does not naturally bind to a nucleic acid. The target molecule can be any molecule of interest. For example, the aptamer can be used to bind to a ligand-binding domain of a protein, thereby preventing interaction of the naturally occurring ligand with the protein. This is a non-limiting example and those in the art will recognize that other embodiments can be readily generated using techniques generally known in the art. (See, for example, Gold et al., 1995, Annu. Rev. Biochem., 64, 763; Brody and Gold, 2000, J. Biotechnol., 74, 5; Sun, 2000, Curr. Opin. Mol. Ther., 2, 100; Kusser, 2000, J. Biotechnol., 74, 27; Hermann and Patel, 2000, Science, 287, 820; and Jayasena, 1999, Clinical Chemistry, 45, 1628.)


In yet another embodiment, a non-nucleotide linker of the invention comprises abasic nucleotide, polyether, polyamine, polyamide, peptide, carbohydrate, lipid, polyhydrocarbon, or other polymeric compounds (e.g. polyethylene glycols such as those having between 2 and 100 ethylene glycol units). Specific examples include those described by Seela and Kaiser, Nucleic Acids Res. 1990, 18:6353 and Nucleic Acids Res. 1987, 15:3113; Cload and Schepartz, J. Am. Chem. Soc. 1991, 113:6324; Richardson and Schepartz, J. Am. Chem. Soc. 1991, 113:5109; Ma et al., Nucleic Acids Res. 1993, 21:2585 and Biochemistry 1993, 32:1751; Durand et al., Nucleic Acids Res. 1990, 18:6353; McCurdy et al., Nucleosides & Nucleotides 1991, 10:287; Jschke et al., Tetrahedron Lett. 1993, 34:301; Ono et al., Biochemistry 1991, 30:9914; Arnold et al., International Publication No. WO 89/02439; Usman et al., International Publication No. WO 95/06731; Dudycz et al., International Publication No. WO 95/11910 and Ferentz and Verdine, J. Am. Chem. Soc. 1991, 113:4000, all hereby incorporated by reference herein. A “non-nucleotide” further means any group or compound that can be incorporated into a nucleic acid chain in the place of one or more nucleotide units, including either sugar and/or phosphate substitutions, and allows the remaining bases to exhibit their enzymatic activity. The group or compound can be abasic in that it does not contain a commonly recognized nucleotide base, such as adenosine, guanine, cytosine, uracil or thymine, for example at the C1 position of the sugar.


In one embodiment, the invention features a short interfering nucleic acid (siNA) molecule capable of mediating RNA interference (RNAi) inside a cell or reconstituted in vitro system, wherein one or both strands of the siNA molecule that are assembled from two separate oligonucleotides do not comprise any ribonucleotides. For example, a siNA molecule can be assembled from a single oligonculeotide where the sense and antisense regions of the siNA comprise separate oligonucleotides that do not have any ribonucleotides (e.g., nucleotides having a 2′-OH group) present in the oligonucleotides. In another example, a siNA molecule can be assembled from a single oligonculeotide where the sense and antisense regions of the siNA are linked or circularized by a nucleotide or non-nucleotide linker as described herein, wherein the oligonucleotide does not have any ribonucleotides (e.g., nucleotides having a 2′-OH group) present in the oligonucleotide. Applicant has surprisingly found that the presense of ribonucleotides (e.g., nucleotides having a 2′-hydroxyl group) within the siNA molecule is not required or essential to support RNAi activity. As such, in one embodiment, all positions within the siNA can include chemically modified nucleotides and/or non-nucleotides such as nucleotides and or non-nucleotides having Formula I, II, III, IV, V, VI, or VII or any combination thereof to the extent that the ability of the siNA molecule to support RNAi activity in a cell is maintained.


In one embodiment, a siNA molecule of the invention is a single stranded siNA molecule that mediates RNAi activity in a cell or reconstituted in vitro system comprising a single stranded polynucleotide having complementarity to a target nucleic acid sequence. In another embodiment, the single stranded siNA molecule of the invention comprises a 5′-terminal phosphate group. In another embodiment, the single stranded siNA molecule of the invention comprises a 5′-terminal phosphate group and a 3′-terminal phosphate group (e.g., a 2′,3′-cyclic phosphate). In another embodiment, the single stranded siNA molecule of the invention comprises about 15 to about 30 (e.g., about 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30) nucleotides. In yet another embodiment, the single stranded siNA molecule of the invention comprises one or more chemically modified nucleotides or non-nucleotides described herein. For example, all the positions within the siNA molecule can include chemically-modified nucleotides such as nucleotides having any of Formulae I-VII, or any combination thereof to the extent that the ability of the siNA molecule to support RNAi activity in a cell is maintained.


In one embodiment, a siNA molecule of the invention is a single stranded siNA molecule that mediates RNAi activity in a cell or reconstituted in vitro system comprising a single stranded polynucleotide having complementarity to a target nucleic acid sequence, wherein one or more pyrimidine nucleotides present in the siNA are 2′-deoxy-2′-fluoro pyrimidine nucleotides (e.g., wherein all pyrimidine nucleotides are 2′-deoxy-2′-fluoro pyrimidine nucleotides or alternately a plurality of pyrimidine nucleotides are 2′-deoxy-2′-fluoro pyrimidine nucleotides), and wherein any purine nucleotides present in the antisense region are 2′-O-methyl purine nucleotides (e.g., wherein all purine nucleotides are 2′-O-methyl purine nucleotides or alternately a plurality of purine nucleotides are 2′-O-methyl purine nucleotides), and a terminal cap modification, such as any modification described herein or shown in FIG. 10, that is optionally present at the 3′-end, the 5′-end, or both of the 3′ and 5′-ends of the antisense sequence. The siNA optionally further comprises about 1 to about 4 or more (e.g., about 1, 2, 3, 4 or more) terminal 2′-deoxynucleotides at the 3′-end of the siNA molecule, wherein the terminal nucleotides can further comprise one or more (e.g., 1, 2, 3, 4 or more) phosphorothioate, phosphonoacetate, and/or thiophosphonoacetate internucleotide linkages, and wherein the siNA optionally further comprises a terminal phosphate group, such as a 5′-terminal phosphate group. In any of these embodiments, any purine nucleotides present in the antisense region are alternatively 2′-deoxy purine nucleotides (e.g., wherein all purine nucleotides are 2′-deoxy purine nucleotides or alternately a plurality of purine nucleotides are 2′-deoxy purine nucleotides). Also, in any of these embodiments, any purine nucleotides present in the siNA (i.e., purine nucleotides present in the sense and/or antisense region) can alternatively be locked nucleic acid (LNA) nucleotides (e.g., wherein all purine nucleotides are LNA nucleotides or alternately a plurality of purine nucleotides are LNA nucleotides). Also, in any of these embodiments, any purine nucleotides present in the siNA are alternatively 2′-methoxyethyl purine nucleotides (e.g., wherein all purine nucleotides are 2′-methoxyethyl purine nucleotides or alternately a plurality of purine nucleotides are 2′-methoxyethyl purine nucleotides). In another embodiment, any modified nucleotides present in the single stranded siNA molecules of the invention comprise modified nucleotides having properties or characteristics similar to naturally occurring ribonucleotides. For example, the invention features siNA molecules including modified nucleotides having a Northern conformation (e.g., Northern pseudorotation cycle, see for example Saenger, Principles of Nucleic Acid Structure, Springer-Verlag ed., 1984). As such, chemically modified nucleotides present in the single stranded siNA molecules of the invention are preferably resistant to nuclease degradation while at the same time maintaining the capacity to mediate RNAi.


In one embodiment, a siNA molecule of the invention comprises chemically modified nucleotides or non-nucleotides (e.g., having any of Formulae I-VII, such as 2′-deoxy, 2′-deoxy-2′-fluoro, or 2′-O-methyl nucleotides) at alternating positions within one or more strands or regions of the siNA molecule. For example, such chemical modifications can be introduced at every other position of a RNA based siNA molecule, starting at either the first or second nucleotide from the 3′-end or 5′-end of the siNA. In a non-limiting example, a double stranded siNA molecule of the invention in which each strand of the siNA is 21 nucleotides in length is featured wherein positions 1, 3, 5, 7, 9, 11, 13, 15, 17, 19 and 21 of each strand are chemically modified (e.g., with compounds having any of Formulae 1-VII, such as such as 2′-deoxy, 2′-deoxy-2′-fluoro, or 2′-O-methyl nucleotides). In another non-limiting example, a double stranded siNA molecule of the invention in which each strand of the siNA is 21 nucleotides in length is featured wherein positions 2, 4, 6, 8, 10, 12, 14, 16, 18, and 20 of each strand are chemically modified (e.g., with compounds having any of Formulae 1-VII, such as such as 2′-deoxy, 2′-deoxy-2′-fluoro, or 2′-O-methyl nucleotides). Such siNA molecules can further comprise terminal cap moieties and/or backbone modifications as described herein.


In one embodiment, the invention features a method for modulating the expression of a HCV gene within a cell comprising: (a) synthesizing a siNA molecule of the invention, which can be chemically-modified, wherein one of the siNA strands comprises a sequence complementary to RNA of the HCV gene; and (b) introducing the siNA molecule into a cell under conditions suitable to modulate (e.g., inhibit) the expression of the HCV gene in the cell.


In one embodiment, the invention features a method for modulating the expression of a HCV gene within a cell comprising: (a) synthesizing a siNA molecule of the invention, which can be chemically-modified, wherein one of the siNA strands comprises a sequence complementary to RNA of the HCV gene and wherein the sense strand sequence of the siNA comprises a sequence identical or substantially similar to the sequence of the target RNA; and (b) introducing the siNA molecule into a cell under conditions suitable to modulate (e.g., inhibit) the expression of the HCV gene in the cell.


In another embodiment, the invention features a method for modulating the expression of more than one HCV gene within a cell comprising: (a) synthesizing siNA molecules of the invention, which can be chemically-modified, wherein one of the siNA strands comprises a sequence complementary to RNA of the HCV genes; and (b) introducing the siNA molecules into a cell under conditions suitable to modulate (e.g., inhibit) the expression of the HCV genes in the cell.


In another embodiment, the invention features a method for modulating the expression of two or more HCV genes within a cell comprising: (a) synthesizing one or more siNA molecules of the invention, which can be chemically-modified, wherein the siNA strands comprise sequences complementary to RNA of the HCV genes and wherein the sense strand sequences of the siNAs comprise sequences identical or substantially similar to the sequences of the target RNAs; and (b) introducing the siNA molecules into a cell under conditions suitable to modulate (e.g., inhibit) the expression of the HCV genes in the cell.


In another embodiment, the invention features a method for modulating the expression of more than one HCV gene within a cell comprising: (a) synthesizing a siNA molecule of the invention, which can be chemically-modified, wherein one of the siNA strands comprises a sequence complementary to RNA of the HCV gene and wherein the sense strand sequence of the siNA comprises a sequence identical or substantially similar to the sequences of the target RNAs; and (b) introducing the siNA molecule into a cell under conditions suitable to modulate (e.g., inhibit) the expression of the HCV genes in the cell.


In one embodiment, siNA molecules of the invention are used as reagents in ex vivo applications. For example, siNA reagents are introduced into tissue or cells that are transplanted into a subject for therapeutic effect. The cells and/or tissue can be derived from an organism or subject that later receives the explant, or can be derived from another organism or subject prior to transplantation. The siNA molecules can be used to modulate the expression of one or more genes in the cells or tissue, such that the cells or tissue obtain a desired phenotype or are able to perform a function when transplanted in vivo. In one embodiment, certain target cells from a patient are extracted. These extracted cells are contacted with siNAs targeting a specific nucleotide sequence within the cells under conditions suitable for uptake of the siNAs by these cells (e.g. using delivery reagents such as cationic lipids, liposomes and the like or using techniques such as electroporation to facilitate the delivery of siNAs into cells). The cells are then reintroduced back into the same patient or other patients. In one embodiment, the invention features a method of modulating the expression of a HCV gene in a tissue explant comprising: (a) synthesizing a siNA molecule of the invention, which can be chemically-modified, wherein one of the siNA strands comprises a sequence complementary to RNA of the HCV gene; and (b) introducing the siNA molecule into a cell of the tissue explant derived from a particular organism under conditions suitable to modulate (e.g., inhibit) the expression of the HCV gene in the tissue explant. In another embodiment, the method further comprises introducing the tissue explant back into the organism the tissue was derived from or into another organism under conditions suitable to modulate (e.g., inhibit) the expression of the HCV gene in that organism.


In one embodiment, the invention features a method of modulating the expression of a HCV gene in a tissue explant comprising: (a) synthesizing a siNA molecule of the invention, which can be chemically-modified, wherein one of the siNA strands comprises a sequence complementary to RNA of the HCV gene and wherein the sense strand sequence of the siNA comprises a sequence identical or substantially similar to the sequence of the target RNA; and (b) introducing the siNA molecule into a cell of the tissue explant derived from a particular organism under conditions suitable to modulate (e.g., inhibit) the expression of the HCV gene in the tissue explant. In another embodiment, the method further comprises introducing the tissue explant back into the organism the tissue was derived from or into another organism under conditions suitable to modulate (e.g., inhibit) the expression of the HCV gene in that organism.


In another embodiment, the invention features a method of modulating the expression of more than one HCV gene in a tissue explant comprising: (a) synthesizing siNA molecules of the invention, which can be chemically-modified, wherein one of the siNA strands comprises a sequence complementary to RNA of the HCV genes; and (b) introducing the siNA molecules into a cell of the tissue explant derived from a particular organism under conditions suitable to modulate (e.g., inhibit) the expression of the HCV genes in the tissue explant. In another embodiment, the method further comprises introducing the tissue explant back into the organism the tissue was derived from or into another organism under conditions suitable to modulate (e.g., inhibit) the expression of the HCV genes in that organism.


In one embodiment, the invention features a method of modulating the expression of a HCV gene in a subject or organism comprising: (a) synthesizing a siNA molecule of the invention, which can be chemically-modified, wherein one of the siNA strands comprises a sequence complementary to RNA of the HCV gene; and (b) introducing the siNA molecule into the subject or organism under conditions suitable to modulate (e.g., inhibit) the expression of the HCV gene in the subject or organism. The level of HCV protein or RNA can be determined using various methods well-known in the art.


In another embodiment, the invention features a method of modulating the expression of more than one HCV gene in a subject or organism comprising: (a) synthesizing siNA molecules of the invention, which can be chemically-modified, wherein one of the siNA strands comprises a sequence complementary to RNA of the HCV genes; and (b) introducing the siNA molecules into the subject or organism under conditions suitable to modulate (e.g., inhibit) the expression of the HCV genes in the subject or organism. The level of HCV protein or RNA can be determined as is known in the art.


In one embodiment, the invention features a method for modulating the expression of a HCV gene within a cell comprising: (a) synthesizing a siNA molecule of the invention, which can be chemically-modified, wherein the siNA comprises a single stranded sequence having complementarity to RNA of the HCV gene; and (b) introducing the siNA molecule into a cell under conditions suitable to modulate (e.g., inhibit) the expression of the HCV gene in the cell.


In another embodiment, the invention features a method for modulating the expression of more than one HCV gene within a cell comprising: (a) synthesizing siNA molecules of the invention, which can be chemically-modified, wherein the siNA comprises a single stranded sequence having complementarity to RNA of the HCV gene; and (b) contacting the cell in vitro or in vivo with the siNA molecule under conditions suitable to modulate (e.g., inhibit) the expression of the HCV genes in the cell.


In one embodiment, the invention features a method of modulating the expression of a HCV gene in a tissue explant (e.g., a liver transplant) comprising: (a) synthesizing a siNA molecule of the invention, which can be chemically-modified, wherein the siNA comprises a single stranded sequence having complementarity to RNA of the HCV gene; and (b) contacting a cell of the tissue explant derived from a particular subject or organism with the siNA molecule under conditions suitable to modulate (e.g., inhibit) the expression of the HCV gene in the tissue explant. In another embodiment, the method further comprises introducing the tissue explant back into the subject or organism the tissue was derived from or into another subject or organism under conditions suitable to modulate (e.g., inhibit) the expression of the HCV gene in that subject or organism.


In another embodiment, the invention features a method of modulating the expression of more than one HCV gene in a tissue explant (e.g., a liver transplant) comprising: (a) synthesizing siNA molecules of the invention, which can be chemically-modified, wherein the siNA comprises a single stranded sequence having complementarity to RNA of the HCV gene; and (b) introducing the siNA molecules into a cell of the tissue explant derived from a particular subject or organism under conditions suitable to modulate (e.g., inhibit) the expression of the HCV genes in the tissue explant. In another embodiment, the method further comprises introducing the tissue explant back into the subject or organism the tissue was derived from or into another subject or organism under conditions suitable to modulate (e.g., inhibit) the expression of the HCV genes in that subject or organism.


In one embodiment, the invention features a method of modulating the expression of a HCV gene in a subject or organism comprising: (a) synthesizing a siNA molecule of the invention, which can be chemically-modified, wherein the siNA comprises a single stranded sequence having complementarity to RNA of the HCV gene; and (b) introducing the siNA molecule into the subject or organism under conditions suitable to modulate (e.g., inhibit) the expression of the HCV gene in the subject or organism.


In another embodiment, the invention features a method of modulating the expression of more than one HCV gene in a subject or organism comprising: (a) synthesizing siNA molecules of the invention, which can be chemically-modified, wherein the siNA comprises a single stranded sequence having complementarity to RNA of the HCV gene; and (b) introducing the siNA molecules into the subject or organism under conditions suitable to modulate (e.g., inhibit) the expression of the HCV genes in the subject or organism.


In one embodiment, the invention features a method of modulating the expression of a HCV gene in a subject or organism comprising contacting the subject or organism with a siNA molecule of the invention under conditions suitable to modulate (e.g., inhibit) the expression of the HCV gene in the subject or organism.


In one embodiment, the invention features a method for treating or preventing HCV infection in a subject or organism comprising contacting the subject or organism with a siNA molecule of the invention under conditions suitable to modulate (e.g., inhibit) the expression of an inhibitor of HCV gene expression in the subject or organism.


In one embodiment, the invention features a method for treating or preventing liver failure in a subject or organism comprising contacting the subject or organism with a siNA molecule of the invention under conditions suitable to modulate (e.g., inhibit) the expression of an inhibitor of HCV gene expression in the subject or organism.


In one embodiment, the invention features a method for treating or preventing hepatocellular carcinoma in a subject or organism comprising contacting the subject or organism with a siNA molecule of the invention under conditions suitable to modulate (e.g., inhibit) the expression of an inhibitor of HCV gene expression in the subject or organism.


In one embodiment, the invention features a method for treating or preventing cirrhosis in a subject or organism comprising contacting the subject or organism with a siNA molecule of the invention under conditions suitable to modulate (e.g., inhibit) the expression of an inhibitor of HCV gene expression in the subject or organism.


In another embodiment, the invention features a method of modulating the expression of more than one HCV gene in a subject or organism comprising contacting the subject or organism with one or more siNA molecules of the invention under conditions suitable to modulate (e.g., inhibit) the expression of the HCV genes in the subject or organism.


The siNA molecules of the invention can be designed to down regulate or inhibit target (e.g., HCV) gene expression through RNAi targeting of a variety of RNA molecules. In one embodiment, the siNA molecules of the invention are used to target various RNAs corresponding to a target gene. Non-limiting examples of such RNAs include messenger RNA (mRNA), alternate RNA splice variants of target gene(s), post-transcriptionally modified RNA of target gene(s), pre-mRNA of target gene(s), and/or RNA templates. If alternate splicing produces a family of transcripts that are distinguished by usage of appropriate exons, the instant invention can be used to inhibit gene expression through the appropriate exons to specifically inhibit or to distinguish among the functions of gene family members. For example, a protein that contains an alternatively spliced transmembrane domain can be expressed in both membrane bound and secreted forms. Use of the invention to target the exon containing the transmembrane domain can be used to determine the functional consequences of pharmaceutical targeting of membrane bound as opposed to the secreted form of the protein. Non-limiting examples of applications of the invention relating to targeting these RNA molecules include therapeutic pharmaceutical applications, pharmaceutical discovery applications, molecular diagnostic and gene function applications, and gene mapping, for example using single nucleotide polymorphism mapping with siNA molecules of the invention. Such applications can be implemented using known gene sequences or from partial sequences available from an expressed sequence tag (EST).


In another embodiment, the siNA molecules of the invention are used to target conserved sequences corresponding to a gene family or gene families such as HCV family genes (e.g., all known HCV strains, groups of related HCV strains, or groups of divergent HCV strains). As such, siNA molecules targeting multiple HCV targets can provide increased therapeutic effect. In addition, siNA can be used to characterize pathways of gene function in a variety of applications. For example, the present invention can be used to inhibit the activity of target gene(s) in a pathway to determine the function of uncharacterized gene(s) in gene function analysis, mRNA function analysis, or translational analysis. The invention can be used to determine potential target gene pathways involved in various diseases and conditions toward pharmaceutical development. The invention can be used to understand pathways of gene expression involved in, for example proliferative diseases, disorders and conditions.


In one embodiment, siNA molecule(s) and/or methods of the invention are used to down regulate the expression of gene(s) that encode RNA referred to by Genbank Accession, for example, HCV genes encoding RNA sequence(s) referred to herein by Genbank Accession number, for example, Genbank Accession Nos. shown in Table I.


In one embodiment, the invention features a method comprising: (a) generating a library of siNA constructs having a predetermined complexity; and (b) assaying the siNA constructs of (a) above, under conditions suitable to determine RNAi target sites within the target RNA sequence. In one embodiment, the siNA molecules of (a) have strands of a fixed length, for example, about 23 nucleotides in length. In another embodiment, the siNA molecules of (a) are of differing length, for example having strands of about 15 to about 30 (e.g., about 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30) nucleotides in length. In one embodiment, the assay can comprise a reconstituted in vitro siNA assay as described herein. In another embodiment, the assay can comprise a cell culture system in which target RNA is expressed. In another embodiment, fragments of target RNA are analyzed for detectable levels of cleavage, for example by gel electrophoresis, northern blot analysis, or RNAse protection assays, to determine the most suitable target site(s) within the target RNA sequence. The target RNA sequence can be obtained as is known in the art, for example, by cloning and/or transcription for in vitro systems, and by cellular expression in in vivo systems.


In one embodiment, the invention features a method comprising: (a) generating a randomized library of siNA constructs having a predetermined complexity, such as of 4N, where N represents the number of base paired nucleotides in each of the siNA construct strands (eg. for a siNA construct having 21 nucleotide sense and antisense strands with 19 base pairs, the complexity would be 419); and (b) assaying the siNA constructs of (a) above, under conditions suitable to determine RNAi target sites within the target HCV RNA sequence. In another embodiment, the siNA molecules of (a) have strands of a fixed length, for example about 23 nucleotides in length. In yet another embodiment, the siNA molecules of (a) are of differing length, for example having strands of about 15 to about 30 (e.g., about 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30) nucleotides in length. In one embodiment, the assay can comprise a reconstituted in vitro siNA assay as described in Example 6 herein. In another embodiment, the assay can comprise a cell culture system in which target RNA is expressed. In another embodiment, fragments of HCV RNA are analyzed for detectable levels of cleavage, for example, by gel electrophoresis, northern blot analysis, or RNAse protection assays, to determine the most suitable target site(s) within the target HCV RNA sequence. The target HCV RNA sequence can be obtained as is known in the art, for example, by cloning and/or transcription for in vitro systems, and by cellular expression in in vivo systems.


In another embodiment, the invention features a method comprising: (a) analyzing the sequence of a RNA target encoded by a target gene; (b) synthesizing one or more sets of siNA molecules having sequence complementary to one or more regions of the RNA of (a); and (c) assaying the siNA molecules of (b) under conditions suitable to determine RNAi targets within the target RNA sequence. In one embodiment, the siNA molecules of (b) have strands of a fixed length, for example about 23 nucleotides in length. In another embodiment, the siNA molecules of (b) are of differing length, for example having strands of about 15 to about 30 (e.g., about 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30) nucleotides in length. In one embodiment, the assay can comprise a reconstituted in vitro siNA assay as described herein. In another embodiment, the assay can comprise a cell culture system in which target RNA is expressed. Fragments of target RNA are analyzed for detectable levels of cleavage, for example by gel electrophoresis, northern blot analysis, or RNAse protection assays, to determine the most suitable target site(s) within the target RNA sequence. The target RNA sequence can be obtained as is known in the art, for example, by cloning and/or transcription for in vitro systems, and by expression in in vivo systems.


By “target site” is meant a sequence within a target RNA that is “targeted” for cleavage mediated by a siNA construct which contains sequences within its antisense region that are complementary to the target sequence.


By “detectable level of cleavage” is meant cleavage of target RNA (and formation of cleaved product RNAs) to an extent sufficient to discern cleavage products above the background of RNAs produced by random degradation of the target RNA. Production of cleavage products from 1-5% of the target RNA is sufficient to detect above the background for most methods of detection.


In one embodiment, the invention features a composition comprising a siNA molecule of the invention, which can be chemically-modified, in a pharmaceutically acceptable carrier or diluent. In another embodiment, the invention features a pharmaceutical composition comprising siNA molecules of the invention, which can be chemically-modified, targeting one or more genes in a pharmaceutically acceptable carrier or diluent. In another embodiment, the invention features a method for diagnosing a disease or condition in a subject comprising administering to the subject a composition of the invention under conditions suitable for the diagnosis of the disease or condition in the subject. In another embodiment, the invention features a method for treating or preventing a disease or condition in a subject, comprising administering to the subject a composition of the invention under conditions suitable for the treatment or prevention of the disease or condition in the subject, alone or in conjunction with one or more other therapeutic compounds. In yet another embodiment, the invention features a method for inhibiting, reducing or preventing HCV infection, liver failure, hepatocellular carcinoma, cirrhosis in a subject or organism comprising administering to the subject a composition of the invention under conditions suitable for inhibiting, reducing or preventing HCV infection, liver failure, hepatocellular carcinoma, cirrhosis in the subject or organism.


In another embodiment, the invention features a method for validating a HCV gene target, comprising: (a) synthesizing a siNA molecule of the invention, which can be chemically-modified, wherein one of the siNA strands includes a sequence complementary to RNA of a HCV target gene; (b) introducing the siNA molecule into a cell, tissue, subject, or organism under conditions suitable for modulating expression of the HCV target gene in the cell, tissue, subject, or organism; and (c) determining the function of the gene by assaying for any phenotypic change in the cell, tissue, subject, or organism.


In another embodiment, the invention features a method for validating a HCV target comprising: (a) synthesizing a siNA molecule of the invention, which can be chemically-modified, wherein one of the siNA strands includes a sequence complementary to RNA of a HCV target gene; (b) introducing the siNA molecule into a biological system under conditions suitable for modulating expression of the HCV target gene in the biological system; and (c) determining the function of the gene by assaying for any phenotypic change in the biological system.


By “biological system” is meant, material, in a purified or unpurified form, from biological sources, including but not limited to human or animal, wherein the system comprises the components required for RNAi activity. The term “biological system” includes, for example, a cell, tissue, subject, or organism, or extract thereof. The term biological system also includes reconstituted RNAi systems that can be used in an in vitro setting.


By “phenotypic change” is meant any detectable change to a cell that occurs in response to contact or treatment with a nucleic acid molecule of the invention (e.g., siNA). Such detectable changes include, but are not limited to, changes in shape, size, proliferation, motility, protein expression or RNA expression or other physical or chemical changes as can be assayed by methods known in the art. The detectable change can also include expression of reporter genes/molecules such as Green Florescent Protein (GFP) or various tags that are used to identify an expressed protein or any other cellular component that can be assayed.


In one embodiment, the invention features a kit containing a siNA molecule of the invention, which can be chemically-modified, that can be used to modulate the expression of a HCV target gene in a biological system, including, for example, in a cell, tissue, subject, or organism. In another embodiment, the invention features a kit containing more than one siNA molecule of the invention, which can be chemically-modified, that can be used to modulate the expression of more than one HCV target gene in a biological system, including, for example, in a cell, tissue, subject, or organism.


In one embodiment, the invention features a cell containing one or more siNA molecules of the invention, which can be chemically-modified. In another embodiment, the cell containing a siNA molecule of the invention is a mammalian cell. In yet another embodiment, the cell containing a siNA molecule of the invention is a human cell.


In one embodiment, the synthesis of a siNA molecule of the invention, which can be chemically-modified, comprises: (a) synthesis of two complementary strands of the siNA molecule; (b) annealing the two complementary strands together under conditions suitable to obtain a double-stranded siNA molecule. In another embodiment, synthesis of the two complementary strands of the siNA molecule is by solid phase oligonucleotide synthesis. In yet another embodiment, synthesis of the two complementary strands of the siNA molecule is by solid phase tandem oligonucleotide synthesis.


In one embodiment, the invention features a method for synthesizing a siNA duplex molecule comprising: (a) synthesizing a first oligonucleotide sequence strand of the siNA molecule, wherein the first oligonucleotide sequence strand comprises a cleavable linker molecule that can be used as a scaffold for the synthesis of the second oligonucleotide sequence strand of the siNA; (b) synthesizing the second oligonucleotide sequence strand of siNA on the scaffold of the first oligonucleotide sequence strand, wherein the second oligonucleotide sequence strand further comprises a chemical moiety than can be used to purify the siNA duplex; (c) cleaving the linker molecule of (a) under conditions suitable for the two siNA oligonucleotide strands to hybridize and form a stable duplex; and (d) purifying the siNA duplex utilizing the chemical moiety of the second oligonucleotide sequence strand. In one embodiment, cleavage of the linker molecule in (c) above takes place during deprotection of the oligonucleotide, for example, under hydrolysis conditions using an alkylamine base such as methylamine. In one embodiment, the method of synthesis comprises solid phase synthesis on a solid support such as controlled pore glass (CPG) or polystyrene, wherein the first sequence of (a) is synthesized on a cleavable linker, such as a succinyl linker, using the solid support as a scaffold. The cleavable linker in (a) used as a scaffold for synthesizing the second strand can comprise similar reactivity as the solid support derivatized linker, such that cleavage of the solid support derivatized linker and the cleavable linker of (a) takes place concomitantly. In another embodiment, the chemical moiety of (b) that can be used to isolate the attached oligonucleotide sequence comprises a trityl group, for example a dimethoxytrityl group, which can be employed in a trityl-on synthesis strategy as described herein. In yet another embodiment, the chemical moiety, such as a dimethoxytrityl group, is removed during purification, for example, using acidic conditions.


In a further embodiment, the method for siNA synthesis is a solution phase synthesis or hybrid phase synthesis wherein both strands of the siNA duplex are synthesized in tandem using a cleavable linker attached to the first sequence which acts a scaffold for synthesis of the second sequence. Cleavage of the linker under conditions suitable for hybridization of the separate siNA sequence strands results in formation of the double-stranded siNA molecule.


In another embodiment, the invention features a method for synthesizing a siNA duplex molecule comprising: (a) synthesizing one oligonucleotide sequence strand of the siNA molecule, wherein the sequence comprises a cleavable linker molecule that can be used as a scaffold for the synthesis of another oligonucleotide sequence; (b) synthesizing a second oligonucleotide sequence having complementarity to the first sequence strand on the scaffold of (a), wherein the second sequence comprises the other strand of the double-stranded siNA molecule and wherein the second sequence further comprises a chemical moiety than can be used to isolate the attached oligonucleotide sequence; (c) purifying the product of (b) utilizing the chemical moiety of the second oligonucleotide sequence strand under conditions suitable for isolating the full-length sequence comprising both siNA oligonucleotide strands connected by the cleavable linker and under conditions suitable for the two siNA oligonucleotide strands to hybridize and form a stable duplex. In one embodiment, cleavage of the linker molecule in (c) above takes place during deprotection of the oligonucleotide, for example, under hydrolysis conditions. In another embodiment, cleavage of the linker molecule in (c) above takes place after deprotection of the oligonucleotide. In another embodiment, the method of synthesis comprises solid phase synthesis on a solid support such as controlled pore glass (CPG) or polystyrene, wherein the first sequence of (a) is synthesized on a cleavable linker, such as a succinyl linker, using the solid support as a scaffold. The cleavable linker in (a) used as a scaffold for synthesizing the second strand can comprise similar reactivity or differing reactivity as the solid support derivatized linker, such that cleavage of the solid support derivatized linker and the cleavable linker of (a) takes place either concomitantly or sequentially. In one embodiment, the chemical moiety of (b) that can be used to isolate the attached oligonucleotide sequence comprises a trityl group, for example a dimethoxytrityl group.


In another embodiment, the invention features a method for making a double-stranded siNA molecule in a single synthetic process comprising: (a) synthesizing an oligonucleotide having a first and a second sequence, wherein the first sequence is complementary to the second sequence, and the first oligonucleotide sequence is linked to the second sequence via a cleavable linker, and wherein a terminal 5′-protecting group, for example, a 5′-O-dimethoxytrityl group (5′-O-DMT) remains on the oligonucleotide having the second sequence; (b) deprotecting the oligonucleotide whereby the deprotection results in the cleavage of the linker joining the two oligonucleotide sequences; and (c) purifying the product of (b) under conditions suitable for isolating the double-stranded siNA molecule, for example using a trityl-on synthesis strategy as described herein.


In another embodiment, the method of synthesis of siNA molecules of the invention comprises the teachings of Scaringe et al., U.S. Pat. Nos. 5,889,136; 6,008,400; and 6,111,086, incorporated by reference herein in their entirety.


In one embodiment, the invention features siNA constructs that mediate RNAi against HCV, wherein the siNA construct comprises one or more chemical modifications, for example, one or more chemical modifications having any of Formulae I-VII or any combination thereof that increases the nuclease resistance of the siNA construct.


In another embodiment, the invention features a method for generating siNA molecules with increased nuclease resistance comprising (a) introducing nucleotides having any of Formula I-VII or any combination thereof into a siNA molecule, and (b) assaying the siNA molecule of step (a) under conditions suitable for isolating siNA molecules having increased nuclease resistance.


In another embodiment, the invention features a method for generating siNA molecules with improved toxicologic profiles (e.g., have attenuated or no immunstimulatory properties) comprising (a) introducing nucleotides having any of Formula I-VII (e.g., siNA motifs referred to in Table IV) or any combination thereof into a siNA molecule, and (b) assaying the siNA molecule of step (a) under conditions suitable for isolating siNA molecules having improved toxicologic profiles.


In another embodiment, the invention features a method for generating siNA molecules that do not stimulate an interferon response (e.g., no interferon response or attenuated interferon response) in a cell, subject, or organism, comprising (a) introducing nucleotides having any of Formula I-VII (e.g., siNA motifs referred to in Table IV) or any combination thereof into a siNA molecule, and (b) assaying the siNA molecule of step (a) under conditions suitable for isolating siNA molecules that do not stimulate an interferon response.


By “improved toxicologic profile”, is meant that the chemically modified siNA construct exhibits decreased toxicity in a cell, subject, or organism compared to an unmodified siNA or siNA molecule having fewer modifications or modifications that are less effective in imparting improved toxicology. In a non-limiting example, siNA molecules with improved toxicologic profiles are associated with a decreased or attenuated immunostimulatory response in a cell, subject, or organism compared to an unmodified siNA or siNA molecule having fewer modifications or modifications that are less effective in imparting improved toxicology. In one embodiment, a siNA molecule with an improved toxicological profile comprises no ribonucleotides. In one embodiment, a siNA molecule with an improved toxicological profile comprises less than 5 ribonucleotides (e.g., 1, 2, 3, or 4 ribonucleotides). In one embodiment, a siNA molecule with an improved toxicological profile comprises Stab 7, Stab 8, Stab 11, Stab 12, Stab 13, Stab 16, Stab 17, Stab 18, Stab 19, Stab 20, Stab 23, Stab 24, Stab 25, Stab 26, Stab 27, Stab 28, Stab 29, Stab 30, Stab 31, Stab 32 or any combination thereof (see Table IV). In one embodiment, the level of immunostimulatory response associated with a given siNA molecule can be measured as is known in the art, for example by determining the level of PKR/interferon response, proliferation, B-cell activation, and/or cytokine production in assays to quantitate the immunostimulatory response of particular siNA molecules (see, for example, Leifer et al., 2003, J. Immunother. 26, 313-9; and U.S. Pat. No. 5,968,909, incorporated in its entirety by reference).


In one embodiment, the invention features siNA constructs that mediate RNAi against HCV, wherein the siNA construct comprises one or more chemical modifications described herein that modulates the binding affinity between the sense and antisense strands of the siNA construct.


In another embodiment, the invention features a method for generating siNA molecules with increased binding affinity between the sense and antisense strands of the siNA molecule comprising (a) introducing nucleotides having any of Formula I-VII or any combination thereof into a siNA molecule, and (b) assaying the siNA molecule of step (a) under conditions suitable for isolating siNA molecules having increased binding affinity between the sense and antisense strands of the siNA molecule.


In one embodiment, the invention features siNA constructs that mediate RNAi against HCV, wherein the siNA construct comprises one or more chemical modifications described herein that modulates the binding affinity between the antisense strand of the siNA construct and a complementary target RNA sequence within a cell.


In one embodiment, the invention features siNA constructs that mediate RNAi against HCV, wherein the siNA construct comprises one or more chemical modifications described herein that modulates the binding affinity between the antisense strand of the siNA construct and a complementary target DNA sequence within a cell.


In another embodiment, the invention features a method for generating siNA molecules with increased binding affinity between the antisense strand of the siNA molecule and a complementary target RNA sequence comprising (a) introducing nucleotides having any of Formula I-VII or any combination thereof into a siNA molecule, and (b) assaying the siNA molecule of step (a) under conditions suitable for isolating siNA molecules having increased binding affinity between the antisense strand of the siNA molecule and a complementary target RNA sequence.


In another embodiment, the invention features a method for generating siNA molecules with increased binding affinity between the antisense strand of the siNA molecule and a complementary target DNA sequence comprising (a) introducing nucleotides having any of Formula I-VII or any combination thereof into a siNA molecule, and (b) assaying the siNA molecule of step (a) under conditions suitable for isolating siNA molecules having increased binding affinity between the antisense strand of the siNA molecule and a complementary target DNA sequence.


In one embodiment, the invention features siNA constructs that mediate RNAi against HCV, wherein the siNA construct comprises one or more chemical modifications described herein that modulate the polymerase activity of a cellular polymerase capable of generating additional endogenous siNA molecules having sequence homology to the chemically-modified siNA construct.


In another embodiment, the invention features a method for generating siNA molecules capable of mediating increased polymerase activity of a cellular polymerase capable of generating additional endogenous siNA molecules having sequence homology to a chemically-modified siNA molecule comprising (a) introducing nucleotides having any of Formula I-VII or any combination thereof into a siNA molecule, and (b) assaying the siNA molecule of step (a) under conditions suitable for isolating siNA molecules capable of mediating increased polymerase activity of a cellular polymerase capable of generating additional endogenous siNA molecules having sequence homology to the chemically-modified siNA molecule.


In one embodiment, the invention features chemically-modified siNA constructs that mediate RNAi against HCV in a cell, wherein the chemical modifications do not significantly effect the interaction of siNA with a target RNA molecule, DNA molecule and/or proteins or other factors that are essential for RNAi in a manner that would decrease the efficacy of RNAi mediated by such siNA constructs.


In another embodiment, the invention features a method for generating siNA molecules with improved RNAi activity against HCV comprising (a) introducing nucleotides having any of Formula I-VII or any combination thereof into a siNA molecule, and (b) assaying the siNA molecule of step (a) under conditions suitable for isolating siNA molecules having improved RNAi activity.


In yet another embodiment, the invention features a method for generating siNA molecules with improved RNAi activity against HCV target RNA comprising (a) introducing nucleotides having any of Formula I-VII or any combination thereof into a siNA molecule, and (b) assaying the siNA molecule of step (a) under conditions suitable for isolating siNA molecules having improved RNAi activity against the target RNA.


In yet another embodiment, the invention features a method for generating siNA molecules with improved RNAi activity against HCV target DNA comprising (a) introducing nucleotides having any of Formula I-VII or any combination thereof into a siNA molecule, and (b) assaying the siNA molecule of step (a) under conditions suitable for isolating siNA molecules having improved RNAi activity against the target DNA.


In one embodiment, the invention features siNA constructs that mediate RNAi against HCV, wherein the siNA construct comprises one or more chemical modifications described herein that modulates the cellular uptake of the siNA construct.


In another embodiment, the invention features a method for generating siNA molecules against HCV with improved cellular uptake comprising (a) introducing nucleotides having any of Formula I-VII or any combination thereof into a siNA molecule, and (b) assaying the siNA molecule of step (a) under conditions suitable for isolating siNA molecules having improved cellular uptake.


In one embodiment, the invention features siNA constructs that mediate RNAi against HCV, wherein the siNA construct comprises one or more chemical modifications described herein that increases the bioavailability of the siNA construct, for example, by attaching polymeric conjugates such as polyethyleneglycol or equivalent conjugates that improve the pharmacokinetics of the siNA construct, or by attaching conjugates that target specific tissue types or cell types in vivo. Non-limiting examples of such conjugates are described in Vargeese et al., U.S. Ser. No. 10/201,394 incorporated by reference herein.


In one embodiment, the invention features a method for generating siNA molecules of the invention with improved bioavailability comprising (a) introducing a conjugate into the structure of a siNA molecule, and (b) assaying the siNA molecule of step (a) under conditions suitable for isolating siNA molecules having improved bioavailability. Such conjugates can include ligands for cellular receptors, such as peptides derived from naturally occurring protein ligands; protein localization sequences, including cellular ZIP code sequences; antibodies; nucleic acid aptamers; vitamins and other co-factors, such as folate and N-acetylgalactosamine; polymers, such as polyethyleneglycol (PEG); phospholipids; cholesterol; polyamines, such as spermine or spermidine; and others.


In one embodiment, the invention features a double stranded short interfering nucleic acid (siNA) molecule that comprises a first nucleotide sequence complementary to a target RNA sequence or a portion thereof, and a second sequence having complementarity to said first sequence, wherein said second sequence is chemically modified in a manner that it can no longer act as a guide sequence for efficiently mediating RNA interference and/or be recognized by cellular proteins that facilitate RNAi.


In one embodiment, the invention features a double stranded short interfering nucleic acid (siNA) molecule that comprises a first nucleotide sequence complementary to a target RNA sequence or a portion thereof, and a second sequence having complementarity to said first sequence, wherein the second sequence is designed or modified in a manner that prevents its entry into the RNAi pathway as a guide sequence or as a sequence that is complementary to a target nucleic acid (e.g., RNA) sequence. Such design or modifications are expected to enhance the activity of siNA and/or improve the specificity of siNA molecules of the invention. These modifications are also expected to minimize any off-target effects and/or associated toxicity.


In one embodiment, the invention features a double stranded short interfering nucleic acid (siNA) molecule that comprises a first nucleotide sequence complementary to a target RNA sequence or a portion thereof, and a second sequence having complementarity to said first sequence, wherein said second sequence is incapable of acting as a guide sequence for mediating RNA interference.


In one embodiment, the invention features a double stranded short interfering nucleic acid (siNA) molecule that comprises a first nucleotide sequence complementary to a target RNA sequence or a portion thereof, and a second sequence having complementarity to said first sequence, wherein said second sequence does not have a terminal 5′-hydroxyl (5′-OH) or 5′-phosphate group.


In one embodiment, the invention features a double stranded short interfering nucleic acid (siNA) molecule that comprises a first nucleotide sequence complementary to a target RNA sequence or a portion thereof, and a second sequence having complementarity to said first sequence, wherein said second sequence comprises a terminal cap moiety at the 5′-end of said second sequence. In one embodiment, the terminal cap moiety comprises an inverted abasic, inverted deoxy abasic, inverted nucleotide moiety, a group shown in FIG. 10, an alkyl or cycloalkyl group, a heterocycle, or any other group that prevents RNAi activity in which the second sequence serves as a guide sequence or template for RNAi.


In one embodiment, the invention features a double stranded short interfering nucleic acid (siNA) molecule that comprises a first nucleotide sequence complementary to a target RNA sequence or a portion thereof, and a second sequence having complementarity to said first sequence, wherein said second sequence comprises a terminal cap moiety at the 5′-end and 3′-end of said second sequence. In one embodiment, each terminal cap moiety individually comprises an inverted abasic, inverted deoxy abasic, inverted nucleotide moiety, a group shown in FIG. 10, an alkyl or cycloalkyl group, a heterocycle, or any other group that prevents RNAi activity in which the second sequence serves as a guide sequence or template for RNAi.


In one embodiment, the invention features a method for generating siNA molecules of the invention with improved specificity for down regulating or inhibiting the expression of a target nucleic acid (e.g., a DNA or RNA such as a gene or its corresponding RNA), comprising (a) introducing one or more chemical modifications into the structure of a siNA molecule, and (b) assaying the siNA molecule of step (a) under conditions suitable for isolating siNA molecules having improved specificity. In another embodiment, the chemical modification used to improve specificity comprises terminal cap modifications at the 5′-end, 3′-end, or both 5′ and 3′-ends of the siNA molecule. The terminal cap modifications can comprise, for example, structures shown in FIG. 10 (e.g. inverted deoxyabasic moieties) or any other chemical modification that renders a portion of the siNA molecule (e.g. the sense strand) incapable of mediating RNA interference against an off target nucleic acid sequence. In a non-limiting example, a siNA molecule is designed such that only the antisense sequence of the siNA molecule can serve as a guide sequence for RISC mediated degradation of a corresponding target RNA sequence. This can be accomplished by rendering the sense sequence of the siNA inactive by introducing chemical modifications to the sense strand that preclude recognition of the sense strand as a guide sequence by RNAi machinery. In one embodiment, such chemical modifications comprise any chemical group at the 5′-end of the sense strand of the siNA, or any other group that serves to render the sense strand inactive as a guide sequence for mediating RNA interference. These modifications, for example, can result in a molecule where the 5′-end of the sense strand no longer has a free 5′-hydroxyl (5′-OH) or a free 5′-phosphate group (e.g., phosphate, diphosphate, triphosphate, cyclic phosphate etc.). Non-limiting examples of such siNA constructs are described herein, such as “Stab 9/10”, “Stab 7/8”, “Stab 7/19”, “Stab 17/22”, “Stab 23/24”, “Stab 24/25”, and “Stab 24/26” (e.g., any siNA having Stab 7, 9, 17, 23, or 24 sense strands) chemistries and variants thereof (see Table 1) wherein the 5′-end and 3′-end of the sense strand of the siNA do not comprise a hydroxyl group or phosphate group.


In one embodiment, the invention features a method for generating siNA molecules of the invention with improved specificity for down regulating or inhibiting the expression of a target nucleic acid (e.g., a DNA or RNA such as a gene or its corresponding RNA), comprising introducing one or more chemical modifications into the structure of a siNA molecule that prevent a strand or portion of the siNA molecule from acting as a template or guide sequence for RNAi activity. In one embodiment, the inactive strand or sense region of the siNA molecule is the sense strand or sense region of the siNA molecule, i.e. the strand or region of the siNA that does not have complementarity to the target nucleic acid sequence. In one embodiment, such chemical modifications comprise any chemical group at the 5′-end of the sense strand or region of the siNA that does not comprise a 5′-hydroxyl (5′-OH) or 5′-phosphate group, or any other group that serves to render the sense strand or sense region inactive as a guide sequence for mediating RNA interference. Non-limiting examples of such siNA constructs are described herein, such as “Stab 9/10”, “Stab 7/8”, “Stab 7/19”, “Stab 17/22”, “Stab 23/24”, “Stab 24/25”, and “Stab 24/26” (e.g., any siNA having Stab 7, 9, 17, 23, or 24 sense strands) chemistries and variants thereof (see Table IV) wherein the 5′-end and 3′-end of the sense strand of the siNA do not comprise a hydroxyl group or phosphate group.


In one embodiment, the invention features a method for screening siNA molecules that are active in mediating RNA interference against a target nucleic acid sequence comprising (a) generating a plurality of unmodified siNA molecules, (b) screening the siNA molecules of step (a) under conditions suitable for isolating siNA molecules that are active in mediating RNA interference against the target nucleic acid sequence, and (c) introducing chemical modifications (e.g. chemical modifications as described herein or as otherwise known in the art) into the active siNA molecules of (b). In one embodiment, the method further comprises re-screening the chemically modified siNA molecules of step (c) under conditions suitable for isolating chemically modified siNA molecules that are active in mediating RNA interference against the target nucleic acid sequence.


In one embodiment, the invention features a method for screening chemically modified siNA molecules that are active in mediating RNA interference against a target nucleic acid sequence comprising (a) generating a plurality of chemically modified siNA molecules (e.g. siNA molecules as described herein or as otherwise known in the art), and (b) screening the siNA molecules of step (a) under conditions suitable for isolating chemically modified siNA molecules that are active in mediating RNA interference against the target nucleic acid sequence.


The term “ligand” refers to any compound or molecule, such as a drug, peptide, hormone, or neurotransmitter, that is capable of interacting with another compound, such as a receptor, either directly or indirectly. The receptor that interacts with a ligand can be present on the surface of a cell or can alternately be an intercellular receptor. Interaction of the ligand with the receptor can result in a biochemical reaction, or can simply be a physical interaction or association.


In another embodiment, the invention features a method for generating siNA molecules of the invention with improved bioavailability comprising (a) introducing an excipient formulation to a siNA molecule, and (b) assaying the siNA molecule of step (a) under conditions suitable for isolating siNA molecules having improved bioavailability. Such excipients include polymers such as cyclodextrins, lipids, cationic lipids, polyamines, phospholipids, nanoparticles, receptors, ligands, and others.


In another embodiment, the invention features a method for generating siNA molecules of the invention with improved bioavailability comprising (a) introducing nucleotides having any of Formulae I-VII or any combination thereof into a siNA molecule, and (b) assaying the siNA molecule of step (a) under conditions suitable for isolating siNA molecules having improved bioavailability.


In another embodiment, polyethylene glycol (PEG) can be covalently attached to siNA compounds of the present invention. The attached PEG can be any molecular weight, preferably from about 100 to about 50,000 daltons (Da).


The present invention can be used alone or as a component of a kit having at least one of the reagents necessary to carry out the in vitro or in vivo introduction of RNA to test samples and/or subjects. For example, preferred components of the kit include a siNA molecule of the invention and a vehicle that promotes introduction of the siNA into cells of interest as described herein (e.g., using lipids and other methods of transfection known in the art, see for example Beigelman et al, U.S. Pat. No. 6,395,713). The kit can be used for target validation, such as in determining gene function and/or activity, or in drug optimization, and in drug discovery (see for example Usman et al., U.S. Ser. No. 60/402,996). Such a kit can also include instructions to allow a user of the kit to practice the invention.


The term “short interfering nucleic acid”, “siNA”, “short interfering RNA”, “siRNA”, “short interfering nucleic acid molecule”, “short interfering oligonucleotide molecule”, or “chemically-modified short interfering nucleic acid molecule” as used herein refers to any nucleic acid molecule capable of inhibiting or down regulating gene expression or viral replication, for example by mediating RNA interference “RNAi” or gene silencing in a sequence-specific manner; see for example Zamore et al., 2000, Cell, 101, 25-33; Bass, 2001, Nature, 411, 428429; Elbashir et al., 2001, Nature, 411, 494-498; and Kreutzer et al., International PCT Publication No. WO 00/44895; Zemicka-Goetz et al., International PCT Publication No. WO 01/36646; Fire, International PCT Publication No. WO 99/32619; Plaetinck et al., International PCT Publication No. WO 00/01846; Mello and Fire, International PCT Publication No. WO 01/29058; Deschamps-Depaillette, International PCT Publication No. WO 99/07409; and Li et al., International PCT Publication No. WO 00/44914; Allshire, 2002, Science, 297, 1818-1819; Volpe et al., 2002, Science, 297, 1833-1837; Jenuwein, 2002, Science, 297, 2215-2218; and Hall et al., 2002, Science, 297, 2232-2237; Hutvagner and Zamore, 2002, Science, 297, 2056-60; McManus et al., 2002, RNA, 8, 842-850; Reinhart et al., 2002, Gene & Dev., 16, 1616-1626; and Reinhart & Bartel, 2002, Science, 297, 1831). Non limiting examples of siNA molecules of the invention are shown in FIGS. 4-6, and Tables II and III herein. For example the siNA can be a double-stranded polynucleotide molecule comprising self-complementary sense and antisense regions, wherein the antisense region comprises nucleotide sequence that is complementary to nucleotide sequence in a target nucleic acid molecule or a portion thereof and the sense region having nucleotide sequence corresponding to the target nucleic acid sequence or a portion thereof. The siNA can be assembled from two separate oligonucleotides, where one strand is the sense strand and the other is the antisense strand, wherein the antisense and sense strands are self-complementary (i.e. each strand comprises nucleotide sequence that is complementary to nucleotide sequence in the other strand; such as where the antisense strand and sense strand form a duplex or double stranded structure, for example wherein the double stranded region is about 15 to about 30, e.g., about 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29 or 30 base pairs; the antisense strand comprises nucleotide sequence that is complementary to nucleotide sequence in a target nucleic acid molecule or a portion thereof and the sense strand comprises nucleotide sequence corresponding to the target nucleic acid sequence or a portion thereof (e.g., about 15 to about 25 or more nucleotides of the siNA molecule are complementary to the target nucleic acid or a portion thereof). Alternatively, the siNA is assembled from a single oligonucleotide, where the self-complementary sense and antisense regions of the siNA are linked by means of a nucleic acid based or non-nucleic acid-based linker(s). The siNA can be a polynucleotide with a duplex, asymmetric duplex, hairpin or asymmetric hairpin secondary structure, having self-complementary sense and antisense regions, wherein the antisense region comprises nucleotide sequence that is complementary to nucleotide sequence in a separate target nucleic acid molecule or a portion thereof and the sense region having nucleotide sequence corresponding to the target nucleic acid sequence or a portion thereof. The siNA can be a circular single-stranded polynucleotide having two or more loop structures and a stem comprising self-complementary sense and antisense regions, wherein the antisense region comprises nucleotide sequence that is complementary to nucleotide sequence in a target nucleic acid molecule or a portion thereof and the sense region having nucleotide sequence corresponding to the target nucleic acid sequence or a portion thereof, and wherein the circular polynucleotide can be processed either in vivo or in vitro to generate an active siNA molecule capable of mediating RNAi. The siNA can also comprise a single stranded polynucleotide having nucleotide sequence complementary to nucleotide sequence in a target nucleic acid molecule or a portion thereof (for example, where such siNA molecule does not require the presence within the siNA molecule of nucleotide sequence corresponding to the target nucleic acid sequence or a portion thereof), wherein the single stranded polynucleotide can further comprise a terminal phosphate group, such as a 5′-phosphate (see for example Martinez et al., 2002, Cell., 110, 563-574 and Schwarz et al., 2002, Molecular Cell, 10, 537-568), or 5′,3′-diphosphate. In certain embodiments, the siNA molecule of the invention comprises separate sense and antisense sequences or regions, wherein the sense and antisense regions are covalently linked by nucleotide or non-nucleotide linkers molecules as is known in the art, or are alternately non-covalently linked by ionic interactions, hydrogen bonding, van der waals interactions, hydrophobic interactions, and/or stacking interactions. In certain embodiments, the siNA molecules of the invention comprise nucleotide sequence that is complementary to nucleotide sequence of a target gene. In another embodiment, the siNA molecule of the invention interacts with nucleotide sequence of a target gene in a manner that causes inhibition of expression of the target gene. As used herein, siNA molecules need not be limited to those molecules containing only RNA, but further encompasses chemically-modified nucleotides and non-nucleotides. In certain embodiments, the short interfering nucleic acid molecules of the invention lack 2′-hydroxy (2′-OH) containing nucleotides. Applicant describes in certain embodiments short interfering nucleic acids that do not require the presence of nucleotides having a 2′-hydroxy group for mediating RNAi and as such, short interfering nucleic acid molecules of the invention optionally do not include any ribonucleotides (e.g., nucleotides having a 2′-OH group). Such siNA molecules that do not require the presence of ribonucleotides within the siNA molecule to support RNAi can however have an attached linker or linkers or other attached or associated groups, moieties, or chains containing one or more nucleotides with 2′-OH groups. Optionally, siNA molecules can comprise ribonucleotides at about 5, 10, 20, 30, 40, or 50% of the nucleotide positions. The modified short interfering nucleic acid molecules of the invention can also be referred to as short interfering modified oligonucleotides “siMON.” As used herein, the term siNA is meant to be equivalent to other terms used to describe nucleic acid molecules that are capable of mediating sequence specific RNAI, for example short interfering RNA (siRNA), double-stranded RNA (dsRNA), micro-RNA (miRNA), short hairpin RNA (shRNA), short interfering oligonucleotide, short interfering nucleic acid, short interfering modified oligonucleotide, chemically-modified siRNA, post-transcriptional gene silencing RNA (ptgsRNA), and others. In addition, as used herein, the term RNAi is meant to be equivalent to other terms used to describe sequence specific RNA interference, such as post transcriptional gene silencing, translational inhibition, or epigenetics. For example, siNA molecules of the invention can be used to epigenetically silence genes at both the post-transcriptional level or the pre-transcriptional level. In a non-limiting example, epigenetic regulation of gene expression by siNA molecules of the invention can result from siNA mediated modification of chromatin structure or methylation pattern to alter gene expression (see, for example, Verdel et al., 2004, Science, 303, 672-676; Pal-Bhadra et al., 2004, Science, 303, 669-672; Allshire, 2002, Science, 297, 1818-1819; Volpe et al., 2002, Science, 297, 1833-1837; Jenuwein, 2002, Science, 297, 2215-2218; and Hall et al., 2002, Science, 297, 2232-2237).


In one embodiment, a siNA molecule of the invention is a duplex forming oligonucleotide “DFO”, (see for example FIGS. 14-15 and Vaish et al., U.S. Ser. No. 10/727,780 filed Dec. 3, 2003 and International PCT Application No. US04/16390, filed May 24, 2004).


In one embodiment, a siNA molecule of the invention is a multifunctional siNA, (see for example FIGS. 16-21 and Jadhav et al., U.S. Ser. No. 60/543,480 filed Feb. 10, 2004 and International PCT Application No. US04/16390, filed May 24, 2004). In one embodiment, the multifunctional siNA of the invention can comprise sequence targeting, for example, two or more regions of HCV RNA (see for example target sequences in Tables II and III). In one embodiment, the multifunctional siNA of the invention can comprise sequence targeting HCV RNA and one or more cellular targets involved in the HCV lifecyle, such as cellular receptors, cell surface molecules, cellular enzymes, cellular transcription factors, and/or cytokines, second messengers, and cellular accessory molecules including, but not limited to, La antigen (see for example Costa-Mattioli et al., 2004, Mol Cell Biol., 24, 6861-70, e.g., Genbank Accession No. NM003142) (e.g., interferon regulatory factors (IRFs; e.g., Genbank Accession No. AF082503.1); cellular PKR protein kinase (e.g., Genbank Accession No. XM002661.7); human eukaryotic initiation factors 2B (elF2Bgamma; e.g., Genbank Accession No. AF256223, and/or elF2gamma; e.g., Genbank Accession No. NM006874.1); human DEAD Box protein (DDX3; e.g., Genbank Accession No. XM018021.2); and cellular proteins that bind to the poly(U) tract of the HCV 3′-UTR, such as polypyrimidine tract-binding protein (e.g., Genbank Accession Nos. NM031991.1 and XM042972.3).


By “asymmetric hairpin” as used herein is meant a linear siNA molecule comprising an antisense region, a loop portion that can comprise nucleotides or non-nucleotides, and a sense region that comprises fewer nucleotides than the antisense region to the extent that the sense region has enough complementary nucleotides to base pair with the antisense region and form a duplex with loop. For example, an asymmetric hairpin siNA molecule of the invention can comprise an antisense region having length sufficient to mediate RNAi in a cell or in vitro system (e.g. about 15 to about 30, or about 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30 nucleotides) and a loop region comprising about 4 to about 12 (e.g., about 4, 5, 6, 7, 8, 9, 10, 11, or 12) nucleotides, and a sense region having about 3 to about 25 (e.g., about 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, or 25) nucleotides that are complementary to the antisense region. The asymmetric hairpin siNA molecule can also comprise a 5′-terminal phosphate group that can be chemically modified. The loop portion of the asymmetric hairpin siNA molecule can comprise nucleotides, non-nucleotides, linker molecules, or conjugate molecules as described herein.


By “asymmetric duplex” as used herein is meant a siNA molecule having two separate strands comprising a sense region and an antisense region, wherein the sense region comprises fewer nucleotides than the antisense region to the extent that the sense region has enough complementary nucleotides to base pair with the antisense region and form a duplex. For example, an asymmetric duplex siNA molecule of the invention can comprise an antisense region having length sufficient to mediate RNAi in a cell or in vitro system (e.g. about 15 to about 30, or about 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30 nucleotides) and a sense region having about 3 to about 25 (e.g., about 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, or 25) nucleotides that are complementary to the antisense region.


By “modulate” is meant that the expression of the gene, or level of RNA molecule or equivalent RNA molecules encoding one or more proteins or protein subunits, or activity of one or more proteins or protein subunits is up regulated or down regulated, such that expression, level, or activity is greater than or less than that observed in the absence of the modulator. For example, the term “modulate” can mean “inhibit,” but the use of the word “modulate” is not limited to this definition.


By “inhibit”, “down-regulate”, or “reduce”, it is meant that the expression of the gene, or level of RNA molecules or equivalent RNA molecules encoding one or more proteins or protein subunits, or activity of one or more proteins or protein subunits, is reduced below that observed in the absence of the nucleic acid molecules (e.g., siNA) of the invention. In one embodiment, inhibition, down-regulation or reduction with an siNA molecule is below that level observed in the presence of an inactive or attenuated molecule. In another embodiment, inhibition, down-regulation, or reduction with siNA molecules is below that level observed in the presence of, for example, an siNA molecule with scrambled sequence or with mismatches. In another embodiment, inhibition, down-regulation, or reduction of gene expression with a nucleic acid molecule of the instant invention is greater in the presence of the nucleic acid molecule than in its absence. In one embodiment, inhibition, down regulation, or reduction of gene expression is associated with post transcriptional silencing, such as RNAi mediated cleavage of a target nucleic acid molecule (e.g. RNA) or inhibition of translation. In one embodiment, inhibition, down regulation, or reduction of gene expression is associated with pretranscriptional silencing.


By “gene”, or “target gene”, is meant a nucleic acid that encodes an RNA, for example, nucleic acid sequences including, but not limited to, structural genes encoding a polypeptide. A gene or target gene can also encode a functional RNA (fRNA) or non-coding RNA (ncRNA), such as small temporal RNA (stRNA), micro RNA (miRNA), small nuclear RNA (snRNA), short interfering RNA (siRNA), small nucleolar RNA (snRNA), ribosomal RNA (rRNA), transfer RNA (tRNA) and precursor RNAs thereof. Such non-coding RNAs can serve as target nucleic acid molecules for siNA mediated RNA interference in modulating the activity of fRNA or ncRNA involved in functional or regulatory cellular processes. Abberant FRNA or ncRNA activity leading to disease can therefore be modulated by siNA molecules of the invention. siNA molecules targeting fRNA and ncRNA can also be used to manipulate or alter the genotype or phenotype of a subject, organism or cell, by intervening in cellular processes such as genetic imprinting, transcription, translation, or nucleic acid processing (e.g., transamination, methylation etc.). The target gene can be a gene derived from a cell, an endogenous gene, a transgene, or exogenous genes such as genes of a pathogen, for example a virus, which is present in the cell after infection thereof. The cell containing the target gene can be derived from or contained in any organism, for example a plant, animal, protozoan, virus, bacterium, or fungus. Non-limiting examples of plants include monocots, dicots, or gymnosperms. Non-limiting examples of animals include vertebrates or invertebrates. Non-limiting examples of fungi include molds or yeasts. For a review, see for example Snyder and Gerstein, 2003, Science, 300, 258-260.


By “non-canonical base pair” is meant any non-Watson Crick base pair, such as mismatches and/or wobble base pairs, including flipped mismatches, single hydrogen bond mismatches, trans-type mismatches, triple base interactions, and quadruple base interactions. Non-limiting examples of such non-canonical base pairs include, but are not limited to, AC reverse Hoogsteen, AC wobble, AU reverse Hoogsteen, GU wobble, AA N7 amino, CC 2-carbonyl-amino(H1)-N-3-amino(H2), GA sheared, UC 4-carbonyl-amino, UU imino-carbonyl, AC reverse wobble, AU Hoogsteen, AU reverse Watson Crick, CG reverse Watson Crick, GC N3-amino-amino N3, AA N1-amino symmetric, AA N7-amino symmetric, GA N7-N1 amino-carbonyl, GA+carbonyl-amino N7-N1, GG N1-carbonyl symmetric, GG N3-amino symmetric, CC carbonyl-amino symmetric, CC N3-amino symmetric, UU 2-carbonyl-imino symmetric, UU 4-carbonyl-imino symmetric, AA amino-N3, AA N1-amino, AC amino 2-carbonyl, AC N3-amino, AC N7-amino, AU amino-4-carbonyl, AU N1-imino, AU N3-imino, AU N7-imino, CC carbonyl-amino, GA amino-N1, GA amino-N7, GA carbonyl-amino, GA N3-amino, GC amino-N3, GC carbonyl-amino, GC N3-amino, GC N7-amino, GG amino-N7, GG carbonyl-imino, GG N7-amino, GU amino-2-carbonyl, GU carbonyl-imino, GU imino-2-carbonyl, GU N7-imino, psiU imino-2-carbonyl, UC 4-carbonyl-amino, UC imino-carbonyl, UU imino4-carbonyl, AC C2-H-N3, GA carbonyl-C2-H, UU imino-4-carbonyl 2 carbonyl-C5-H, AC amino(A) N3(C)-carbonyl, GC imino amino-carbonyl, Gpsi imino-2-carbonyl amino-2-carbonyl, and GU imino amino-2-carbonyl base pairs.


By “HCV” as used herein is meant, any hepatitis C virus or HCV protein, peptide, or polypeptide having HCV activity, such as encoded by HCV Genbank Accession Nos. shown in Table I. The term HCV also refers to nucleic acid sequences encoding any HCV protein, peptide, or polypeptide having HCV activity. The term “HCV” is also meant to include other HCV encoding sequence, such as other HCV isoforms, mutant HCV genes, splice variants of HCV genes, and HCV gene polymorphisms. In one embodiment, the term HCV as used herein refers to cellular or host proteins or polynucleotides encoding such proteins or that are otherwise involved in HCV infection and/or replication.


By “homologous sequence” is meant, a nucleotide sequence that is shared by one or more polynucleotide sequences, such as genes, gene transcripts and/or non-coding polynucleotides. For example, a homologous sequence can be a nucleotide sequence that is shared by two or more genes encoding related but different proteins, such as different members of a gene family, different protein epitopes, different protein isoforms or completely divergent genes, such as a cytokine and its corresponding receptors. A homologous sequence can be a nucleotide sequence that is shared by two or more non-coding polynucleotides, such as noncoding DNA or RNA, regulatory sequences, introns, and sites of transcriptional control or regulation. Homologous sequences can also include conserved sequence regions shared by more than one polynucleotide sequence. Homology does not need to be perfect homology (e.g., 100%), as partially homologous sequences are also contemplated by the instant invention (e.g., 99%, 98%, 97%, 96%, 95%, 94%, 93%, 92%, 91%, 90%, 89%, 88%, 87%, 86%, 85%, 84%, 83%, 82%, 81%, 80% etc.).


By “conserved sequence region” is meant, a nucleotide sequence of one or more regions in a polynucleotide does not vary significantly between generations or from one biological system, subject, or organism to another biological system, subject, or organism. The polynucleotide can include both coding and non-coding DNA and RNA.


By “sense region” is meant a nucleotide sequence of a siNA molecule having complementarity to an antisense region of the siNA molecule. In addition, the sense region of a siNA molecule can comprise a nucleic acid sequence having homology with a target nucleic acid sequence.


By “antisense region” is meant a nucleotide sequence of a siNA molecule having complementarity to a target nucleic acid sequence. In addition, the antisense region of a siNA molecule can optionally comprise a nucleic acid sequence having complementarity to a sense region of the siNA molecule.


By “target nucleic acid” is meant any nucleic acid sequence whose expression or activity is to be modulated. The target nucleic acid can be DNA or RNA.


By “complementarity” is meant that a nucleic acid can form hydrogen bond(s) with another nucleic acid sequence by either traditional Watson-Crick or other non-traditional types. In reference to the nucleic molecules of the present invention, the binding free energy for a nucleic acid molecule with its complementary sequence is sufficient to allow the relevant function of the nucleic acid to proceed, e.g., RNAi activity. Determination of binding free energies for nucleic acid molecules is well known in the art (see, e.g., Turner et al., 1987, CSH Symp. Quant. Biol. LII pp. 123-133; Frier et al., 1986, Proc. Nat. Acad. Sci. USA 83:9373-9377; Turner et al., 1987, J. Am. Chem. Soc. 109:3783-3785). A percent complementarity indicates the percentage of contiguous residues in a nucleic acid molecule that can form hydrogen bonds (e.g., Watson-Crick base pairing) with a second nucleic acid sequence (e.g., 5, 6, 7, 8, 9, or 10 nucleotides out of a total of 10 nucleotides in the first oligonucleotide being based paired to a second nucleic acid sequence having 10 nucleotides represents 50%, 60%, 70%, 80%, 90%, and 100% complementary respectively). “Perfectly complementary” means that all the contiguous residues of a nucleic acid sequence will hydrogen bond with the same number of contiguous residues in a second nucleic acid sequence. In one embodiment, a siNA molecule of the invention comprises about 15 to about 30 or more (e.g., about 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30 or more) nucleotides that are complementary to one or more target nucleic acid molecules or a portion thereof.


In one embodiment, siNA molecules of the invention that down regulate or reduce HCV gene expression are used for treating, preventing or reducing HCV infection, liver failure, hepatocellular carcinoma, or cirrhosis in a subject or organism.


In one embodiment of the present invention, each sequence of a siNA molecule of the invention is independently about 15 to about 30 nucleotides in length, in specific embodiments about 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30 nucleotides in length. In another embodiment, the siNA duplexes of the invention independently comprise about 15 to about 30 base pairs (e.g., about 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30). In another embodiment, one or more strands of the siNA molecule of the invention independently comprises about 15 to about 30 nucleotides (e.g., about 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30) that are complementary to a target nucleic acid molecule. In yet another embodiment, siNA molecules of the invention comprising hairpin or circular structures are about 35 to about 55 (e.g., about 35, 40, 45, 50 or 55) nucleotides in length, or about 38 to about 44 (e.g., about 38, 39, 40, 41, 42, 43, or 44) nucleotides in length and comprising about 15 to about 25 (e.g., about 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, or 25) base pairs. Exemplary siNA molecules of the invention are shown in Table II. Exemplary synthetic siNA molecules of the invention are shown in Table III and/or FIGS. 4-5.


As used herein “cell” is used in its usual biological sense, and does not refer to an entire multicellular organism, e.g., specifically does not refer to a human. The cell can be present in an organism, e.g., birds, plants and mammals such as humans, cows, sheep, apes, monkeys, swine, dogs, and cats. The cell can be prokaryotic (e.g., bacterial cell) or eukaryotic (e.g., mammalian or plant cell). The cell can be of somatic or germ line origin, totipotent or pluripotent, dividing or non-dividing. The cell can also be derived from or can comprise a gamete or embryo, a stem cell, or a fully differentiated cell.


The siNA molecules of the invention are added directly, or can be complexed with cationic lipids, packaged within liposomes, or otherwise delivered to target cells or tissues. The nucleic acid or nucleic acid complexes can be locally administered to relevant tissues ex vivo, or in vivo through direct dermal application, transdermal application, or injection, with or without their incorporation in biopolymers. In particular embodiments, the nucleic acid molecules of the invention comprise sequences shown in Tables II-III and/or FIGS. 4-5. Examples of such nucleic acid molecules consist essentially of sequences defined in these tables and figures. Furthermore, the chemically modified constructs described in Table IV can be applied to any siNA sequence of the invention.


In another aspect, the invention provides mammalian cells containing one or more siNA molecules of this invention. The one or more siNA molecules can independently be targeted to the same or different sites.


By “RNA” is meant a molecule comprising at least one ribonucleotide residue. By “ribonucleotide” is meant a nucleotide with a hydroxyl group at the 2′ position of a β-D-ribofuranose moiety. The terms include double-stranded RNA, single-stranded RNA, isolated RNA such as partially purified RNA, essentially pure RNA, synthetic RNA, recombinantly produced RNA, as well as altered RNA that differs from naturally occurring RNA by the addition, deletion, substitution and/or alteration of one or more nucleotides. Such alterations can include addition of non-nucleotide material, such as to the end(s) of the siNA or internally, for example at one or more nucleotides of the RNA. Nucleotides in the RNA molecules of the instant invention can also comprise non-standard nucleotides, such as non-naturally occurring nucleotides or chemically synthesized nucleotides or deoxynucleotides. These altered RNAs can be referred to as analogs or analogs of naturally-occurring RNA.


By “subject” is meant an organism, which is a donor or recipient of explanted cells or the cells themselves. “Subject” also refers to an organism to which the nucleic acid molecules of the invention can be administered. A subject can be a mammal or mammalian cells, including a human or human cells.


The term “phosphorothioate” as used herein refers to an internucleotide linkage having Formula I, wherein Z and/or W comprise a sulfur atom. Hence, the term phosphorothioate refers to both phosphorothioate and phosphorodithioate internucleotide linkages.


The term “phosphonoacetate” as used herein refers to an internucleotide linkage having Formula I, wherein Z and/or W comprise an acetyl or protected acetyl group.


The term “thiophosphonoacetate” as used herein refers to an internucleotide linkage having Formula I, wherein Z comprises an acetyl or protected acetyl group and W comprises a sulfur atom or alternately W comprises an acetyl or protected acetyl group and Z comprises a sulfur atom.


The term “universal base” as used herein refers to nucleotide base analogs that form base pairs with each of the natural DNA/RNA bases with little discrimination between them. Non-limiting examples of universal bases include C-phenyl, C-naphthyl and other aromatic derivatives, inosine, azole carboxamides, and nitroazole derivatives such as 3-nitropyrrole, 4-nitroindole, 5-nitroindole, and 6-nitroindole as known in the art (see for example Loakes, 2001, Nucleic Acids Research, 29, 2437-2447).


The term “acyclic nucleotide” as used herein refers to any nucleotide having an acyclic ribose sugar, for example where any of the ribose carbons (C1, C2, C3, C4, or C5), are independently or in combination absent from the nucleotide.


The nucleic acid molecules of the instant invention, individually, or in combination or in conjunction with other drugs, can be used to treat, inhibit, reduce, or prevent HCV infection, liver failure, hepatocellular carcinoma, and/or cirrhosis in a subject or organism. For example, the siNA molecules can be administered to a subject or can be administered to other appropriate cells evident to those skilled in the art, individually or in combination with one or more drugs under conditions suitable for the treatment.


In a further embodiment, the siNA molecules can be used in combination with other known treatments to treat, inhibit, reduce, or prevent HCV infection, liver failure, hepatocellular carcinoma, and/or cirrhosis in a subject or organism. For example, the described molecules could be used in combination with one or more known compounds, treatments, or procedures to treat, inhibit, reduce, or prevent HCV infection, liver failure, hepatocellular carcinoma, and/or cirrhosis in a subject or organism as are known in the art.


In one embodiment, the invention features an expression vector comprising a nucleic acid sequence encoding at least one siNA molecule of the invention, in a manner which allows expression of the siNA molecule. For example, the vector can contain sequence(s) encoding both strands of a siNA molecule comprising a duplex. The vector can also contain sequence(s) encoding a single nucleic acid molecule that is self-complementary and thus forms a siNA molecule. Non-limiting examples of such expression vectors are described in Paul et al., 2002, Nature Biotechnology, 19, 505; Miyagishi and Taira, 2002, Nature Biotechnology, 19, 497; Lee et al., 2002, Nature Biotechnology, 19, 500; and Novina et al., 2002, Nature Medicine, advance online publication doi: 10.1038/nm725.


In another embodiment, the invention features a mammalian cell, for example, a human cell, including an expression vector of the invention.


In yet another embodiment, the expression vector of the invention comprises a sequence for a siNA molecule having complementarity to a RNA molecule referred to by a Genbank Accession numbers, for example Genbank Accession Nos. shown in Table I.


In one embodiment, an expression vector of the invention comprises a nucleic acid sequence encoding two or more siNA molecules, which can be the same or different.


In another aspect of the invention, siNA molecules that interact with target RNA molecules and down-regulate gene encoding target RNA molecules (for example target RNA molecules referred to by Genbank Accession numbers herein) are expressed from transcription units inserted into DNA or RNA vectors. The recombinant vectors can be DNA plasmids or viral vectors. siNA expressing viral vectors can be constructed based on, but not limited to, adeno-associated virus, retrovirus, adenovirus, or alphavirus. The recombinant vectors capable of expressing the siNA molecules can be delivered as described herein, and persist in target cells. Alternatively, viral vectors can be used that provide for transient expression of siNA molecules. Such vectors can be repeatedly administered as necessary. Once expressed, the siNA molecules bind and down-regulate gene function or expression via RNA interference (RNAi). Delivery of siNA expressing vectors can be systemic, such as by intravenous or intramuscular administration, by administration to target cells ex-planted from a subject followed by reintroduction into the subject, or by any other means that would allow for introduction into the desired target cell.


By “vectors” is meant any nucleic acid- and/or viral-based technique used to deliver a desired nucleic acid.


Other features and advantages of the invention will be apparent from the following description of the preferred embodiments thereof, and from the claims.




BRIEF DESCRIPTION OF THE DRAWINGS


FIG. 1 shows a non-limiting example of a scheme for the synthesis of siNA molecules. The complementary siNA sequence strands, strand 1 and strand 2, are synthesized in tandem and are connected by a cleavable linkage, such as a nucleotide succinate or abasic succinate, which can be the same or different from the cleavable linker used for solid phase synthesis on a solid support. The synthesis can be either solid phase or solution phase, in the example shown, the synthesis is a solid phase synthesis. The synthesis is performed such that a protecting group, such as a dimethoxytrityl group, remains intact on the terminal nucleotide of the tandem oligonucleotide. Upon cleavage and deprotection of the oligonucleotide, the two siNA strands spontaneously hybridize to form a siNA duplex, which allows the purification of the duplex by utilizing the properties of the terminal protecting group, for example by applying a trityl on purification method wherein only duplexes/oligonucleotides with the terminal protecting group are isolated.



FIG. 2 shows a MALDI-TOF mass spectrum of a purified siNA duplex synthesized by a method of the invention. The two peaks shown correspond to the predicted mass of the separate siNA sequence strands. This result demonstrates that the siNA duplex generated from tandem synthesis can be purified as a single entity using a simple trityl-on purification methodology.



FIG. 3 shows a non-limiting proposed mechanistic representation of target RNA degradation involved in RNAi. Double-stranded RNA (dsRNA), which is generated by RNA-dependent RNA polymerase (RdRP) from foreign single-stranded RNA, for example viral, transposon, or other exogenous RNA, activates the DICER enzyme that in turn generates siNA duplexes. Alternately, synthetic or expressed siNA can be introduced directly into a cell by appropriate means. An active siNA complex forms which recognizes a target RNA, resulting in degradation of the target RNA by the RISC endonuclease complex or in the synthesis of additional RNA by RNA-dependent RNA polymerase (RdRP), which can activate DICER and result in additional siNA molecules, thereby amplifying the RNAi response.



FIG. 4A-F shows non-limiting examples of chemically-modified siNA constructs of the present invention. In the figure, N stands for any nucleotide (adenosine, guanosine, cytosine, uridine, or optionally thymidine, for example thymidine can be substituted in the overhanging regions designated by parenthesis (N N). Various modifications are shown for the sense and antisense strands of the siNA constructs.



FIG. 4A: The sense strand comprises 21 nucleotides wherein the two terminal 3′-nucleotides are optionally base paired and wherein all nucleotides present are ribonucleotides except for (N N) nucleotides, which can comprise ribonucleotides, deoxynucleotides, universal bases, or other chemical modifications described herein. The antisense strand comprises 21 nucleotides, optionally having a 3′-terminal glyceryl moiety wherein the two terminal 3′-nucleotides are optionally complementary to the target RNA sequence, and wherein all nucleotides present are ribonucleotides except for (N N) nucleotides, which can comprise ribonucleotides, deoxynucleotides, universal bases, or other chemical modifications described herein. A modified internucleotide linkage, such as a phosphorothioate, phosphorodithioate or other modified internucleotide linkage as described herein, shown as “s”, optionally connects the (N N) nucleotides in the antisense strand.



FIG. 4B: The sense strand comprises 21 nucleotides wherein the two terminal 3′-nucleotides are optionally base paired and wherein all pyrimidine nucleotides that may be present are 2′deoxy-2′-fluoro modified nucleotides and all purine nucleotides that may be present are 2′-O-methyl modified nucleotides except for (N N) nucleotides, which can comprise ribonucleotides, deoxynucleotides, universal bases, or other chemical modifications described herein. The antisense strand comprises 21 nucleotides, optionally having a 3′-terminal glyceryl moiety and wherein the two terminal 3′-nucleotides are optionally complementary to the target RNA sequence, and wherein all pyrimidine nucleotides that may be present are 2′-deoxy-2′-fluoro modified nucleotides and all purine nucleotides that may be present are 2′-O-methyl modified nucleotides except for (N N) nucleotides, which can comprise ribonucleotides, deoxynucleotides, universal bases, or other chemical modifications described herein. A modified internucleotide linkage, such as a phosphorothioate, phosphorodithioate or other modified internucleotide linkage as described herein, shown as “s”, optionally connects the (N N) nucleotides in the sense and antisense strand.



FIG. 4C: The sense strand comprises 21 nucleotides having 5′- and 3′-terminal cap moieties wherein the two terminal 3′-nucleotides are optionally base paired and wherein all pyrimidine nucleotides that may be present are 2′-O-methyl or 2′-deoxy-2′-fluoro modified nucleotides except for (N N) nucleotides, which can comprise ribonucleotides, deoxynucleotides, universal bases, or other chemical modifications described herein. The antisense strand comprises 21 nucleotides, optionally having a 3′-terminal glyceryl moiety and wherein the two terminal 3′-nucleotides are optionally complementary to the target RNA sequence, and wherein all pyrimidine nucleotides that may be present are 2′-deoxy-2′-fluoro modified nucleotides except for (N N) nucleotides, which can comprise ribonucleotides, deoxynucleotides, universal bases, or other chemical modifications described herein. A modified internucleotide linkage, such as a phosphorothioate, phosphorodithioate or other modified internucleotide linkage as described herein, shown as “s”, optionally connects the (N N) nucleotides in the antisense strand.



FIG. 4D: The sense strand comprises 21 nucleotides having 5′- and 3′-terminal cap moieties wherein the two terminal 3′-nucleotides are optionally base paired and wherein all pyrimidine nucleotides that may be present are 2′-deoxy-2′-fluoro modified nucleotides except for (N N) nucleotides, which can comprise ribonucleotides, deoxynucleotides, universal bases, or other chemical modifications described herein and wherein and all purine nucleotides that may be present are 2′-deoxy nucleotides. The antisense strand comprises 21 nucleotides, optionally having a 3′-terminal glyceryl moiety and wherein the two terminal 3′-nucleotides are optionally complementary to the target RNA sequence, wherein all pyrimidine nucleotides that may be present are 2′-deoxy-2′-fluoro modified nucleotides and all purine nucleotides that may be present are 2′-O-methyl modified nucleotides except for (N N) nucleotides, which can comprise ribonucleotides, deoxynucleotides, universal bases, or other chemical modifications described herein. A modified internucleotide linkage, such as a phosphorothioate, phosphorodithioate or other modified internucleotide linkage as described herein, shown as “s”, optionally connects the (N N) nucleotides in the antisense strand.



FIG. 4E: The sense strand comprises 21 nucleotides having 5′- and 3′-terminal cap moieties wherein the two terminal 3′-nucleotides are optionally base paired and wherein all pyrimidine nucleotides that may be present are 2′-deoxy-2′-fluoro modified nucleotides except for (N N) nucleotides, which can comprise ribonucleotides, deoxynucleotides, universal bases, or other chemical modifications described herein. The antisense strand comprises 21 nucleotides, optionally having a 3′-terminal glyceryl moiety and wherein the two terminal 3′-nucleotides are optionally complementary to the target RNA sequence, and wherein all pyrimidine nucleotides that may be present are 2′-deoxy-2′-fluoro modified nucleotides and all purine nucleotides that may be present are 2′-O-methyl modified nucleotides except for (N N) nucleotides, which can comprise ribonucleotides, deoxynucleotides, universal bases, or other chemical modifications described herein. A modified internucleotide linkage, such as a phosphorothioate, phosphorodithioate or other modified internucleotide linkage as described herein, shown as “s”, optionally connects the (N N) nucleotides in the antisense strand.



FIG. 4F: The sense strand comprises 21 nucleotides having 5′- and 3′-terminal cap moieties wherein the two terminal 3′-nucleotides are optionally base paired and wherein all pyrimidine nucleotides that may be present are 2′-deoxy-2′-fluoro modified nucleotides except for (N N) nucleotides, which can comprise ribonucleotides, deoxynucleotides, universal bases, or other chemical modifications described herein and wherein and all purine nucleotides that may be present are 2′-deoxy nucleotides. The antisense strand comprises 21 nucleotides, optionally having a 3′-terminal glyceryl moiety and wherein the two terminal 3′-nucleotides are optionally complementary to the target RNA sequence, and having one 3′-terminal phosphorothioate internucleotide linkage and wherein all pyrimidine nucleotides that may be present are 2′-deoxy-2′-fluoro modified nucleotides and all purine nucleotides that may be present are 2′-deoxy nucleotides except for (N N) nucleotides, which can comprise ribonucleotides, deoxynucleotides, universal bases, or other chemical modifications described herein. A modified internucleotide linkage, such as a phosphorothioate, phosphorodithioate or other modified internucleotide linkage as described herein, shown as “s”, optionally connects the (N N) nucleotides in the antisense strand. The antisense strand of constructs A-F comprise sequence complementary to any target nucleic acid sequence of the invention. Furthermore, when a glyceryl moiety (L) is present at the 3′-end of the antisense strand for any construct shown in FIG. 4A-F, the modified internucleotide linkage is optional.



FIG. 5A-F shows non-limiting examples of specific chemically-modified siNA sequences of the invention. A-F applies the chemical modifications described in FIG. 4A-F to a HCV siNA sequence. Such chemical modifications can be applied to any HCV sequence and/or cellular target sequence.



FIG. 6 shows non-limiting examples of different siNA constructs of the invention. The examples shown (constructs 1, 2, and 3) have 19 representative base pairs; however, different embodiments of the invention include any number of base pairs described herein. Bracketed regions represent nucleotide overhangs, for example, comprising about 1, 2, 3, or 4 nucleotides in length, preferably about 2 nucleotides. Constructs 1 and 2 can be used independently for RNAi activity. Construct 2 can comprise a polynucleotide or non-nucleotide linker, which can optionally be designed as a biodegradable linker. In one embodiment, the loop structure shown in construct 2 can comprise a biodegradable linker that results in the formation of construct 1 in vivo and/or in vitro. In another example, construct 3 can be used to generate construct 2 under the same principle wherein a linker is used to generate the active siNA construct 2 in vivo and/or in vitro, which can optionally utilize another biodegradable linker to generate the active siNA construct 1 in vivo and/or in vitro. As such, the stability and/or activity of the siNA constructs can be modulated based on the design of the siNA construct for use in vivo or in vitro and/or in vitro.



FIG. 7A-C is a diagrammatic representation of a scheme utilized in generating an expression cassette to generate siNA hairpin constructs.



FIG. 7A: A DNA oligomer is synthesized with a 5′-restriction site (R1) sequence followed by a region having sequence identical (sense region of siNA) to a predetermined HCV target sequence, wherein the sense region comprises, for example, about 19, 20, 21, or 22 nucleotides (N) in length, which is followed by a loop sequence of defined sequence (X), comprising, for example, about 3 to about 10 nucleotides.



FIG. 7B: The synthetic construct is then extended by DNA polymerase to generate a hairpin structure having self-complementary sequence that will result in a siNA transcript having specificity for a HCV target sequence and having self-complementary sense and antisense regions.



FIG. 7C: The construct is heated (for example to about 95° C.) to linearize the sequence, thus allowing extension of a complementary second DNA strand using a primer to the 3′-restriction sequence of the first strand. The double-stranded DNA is then inserted into an appropriate vector for expression in cells. The construct can be designed such that a 3′-terminal nucleotide overhang results from the transcription, for example, by engineering restriction sites and/or utilizing a poly-U termination region as described in Paul et al., 2002, Nature Biotechnology, 29, 505-508.



FIG. 8A-C is a diagrammatic representation of a scheme utilized in generating an expression cassette to generate double-stranded siNA constructs.



FIG. 8A: A DNA oligomer is synthesized with a 5′-restriction (R1) site sequence followed by a region having sequence identical (sense region of siNA) to a predetermined HCV target sequence, wherein the sense region comprises, for example, about 19, 20, 21, or 22 nucleotides (N) in length, and which is followed by a 3′-restriction site (R2) which is adjacent to a loop sequence of defined sequence (X).



FIG. 8B: The synthetic construct is then extended by DNA polymerase to generate a hairpin structure having self-complementary sequence.



FIG. 8C: The construct is processed by restriction enzymes specific to R1 and R2 to generate a double-stranded DNA which is then inserted into an appropriate vector for expression in cells. The transcription cassette is designed such that a U6 promoter region flanks each side of the dsDNA which generates the separate sense and antisense strands of the siNA. Poly T termination sequences can be added to the constructs to generate U overhangs in the resulting transcript.



FIG. 9A-E is a diagrammatic representation of a method used to determine target sites for siNA mediated RNAi within a particular target nucleic acid sequence, such as messenger RNA.



FIG. 9A: A pool of siNA oligonucleotides are synthesized wherein the antisense region of the siNA constructs has complementarity to target sites across the target nucleic acid sequence, and wherein the sense region comprises sequence complementary to the antisense region of the siNA.


FIGS. 9B&C: (FIG. 9B) The sequences are pooled and are inserted into vectors such that (FIG. 9C) transfection of a vector into cells results in the expression of the siNA.



FIG. 9D: Cells are sorted based on phenotypic change that is associated with modulation of the target nucleic acid sequence.



FIG. 9E: The siNA is isolated from the sorted cells and is sequenced to identify efficacious target sites within the target nucleic acid sequence.



FIG. 10 shows non-limiting examples of different stabilization chemistries (1-10) that can be used, for example, to stabilize the 3′-end of siNA sequences of the invention, including (1) [3-3′]-inverted deoxyribose; (2) deoxyribonucleotide; (3) [5′-3′]-3′-deoxyribonucleotide; (4) [5′-3′]-ribonucleotide; (5) [5′-3′]-3′-O-methyl ribonucleotide; (6) 3′-glyceryl; (7) [3′-5′]-3′-deoxyribonucleotide; (8) [3′-3′]-deoxyribonucleotide; (9) [5′-2′]-deoxyribonucleotide; and (10) [5-3′]-dideoxyribonucleotide. In addition to modified and unmodified backbone chemistries indicated in the figure, these chemistries can be combined with different backbone modifications as described herein, for example, backbone modifications having Formula I. In addition, the 2′-deoxy nucleotide shown 5′ to the terminal modifications shown can be another modified or unmodified nucleotide or non-nucleotide described herein, for example modifications having any of Formulae I-VII or any combination thereof.



FIG. 11 shows a non-limiting example of a strategy used to identify chemically modified siNA constructs of the invention that are nuclease resistance while preserving the ability to mediate RNAi activity. Chemical modifications are introduced into the siNA construct based on educated design parameters (e.g. introducing 2′-mofications, base modifications, backbone modifications, terminal cap modifications etc). The modified construct in tested in an appropriate system (e.g. human serum for nuclease resistance, shown, or an animal model for PK/delivery parameters). In parallel, the siNA construct is tested for RNAi activity, for example in a cell culture system such as a luciferase reporter assay). Lead siNA constructs are then identified which possess a particular characteristic while maintaining RNAi activity, and can be further modified and assayed once again. This same approach can be used to identify siNA-conjugate molecules with improved pharmacokinetic profiles, delivery, and RNAi activity.



FIG. 12 shows non-limiting examples of phosphorylated siNA molecules of the invention, including linear and duplex constructs and asymmetric derivatives thereof.



FIG. 13 shows non-limiting examples of chemically modified terminal phosphate groups of the invention.



FIG. 14A shows a non-limiting example of methodology used to design self complementary DFO constructs utilizing palindrome and/or repeat nucleic acid sequences that are identified in a target nucleic acid sequence. (i) A palindrome or repeat sequence is identified in a nucleic acid target sequence. (ii) A sequence is designed that is complementary to the target nucleic acid sequence and the palindrome sequence. (iii) An inverse repeat sequence of the non-palindrome/repeat portion of the complementary sequence is appended to the 3′-end of the complementary sequence to generate a self complementary DFO molecule comprising sequence complementary to the nucleic acid target. (iv) The DFO molecule can self-assemble to form a double stranded oligonucleotide. FIG. 14B shows a non-limiting representative example of a duplex forming oligonucleotide sequence. FIG. 14C shows a non-limiting example of the self assembly schematic of a representative duplex forming oligonucleotide sequence. FIG. 14D shows a non-limiting example of the self assembly schematic of a representative duplex forming oligonucleotide sequence followed by interaction with a target nucleic acid sequence resulting in modulation of gene expression.



FIG. 15 shows a non-limiting example of the design of self complementary DFO constructs utilizing palindrome and/or repeat nucleic acid sequences that are incorporated into the DFO constructs that have sequence complementary to any target nucleic acid sequence of interest. Incorporation of these palindrome/repeat sequences allow the design of DFO constructs that form duplexes in which each strand is capable of mediating modulation of target gene expression, for example by RNAi. First, the target sequence is identified. A complementary sequence is then generated in which nucleotide or non-nucleotide modifications (shown as X or Y) are introduced into the complementary sequence that generate an artificial palindrome (shown as XYXYXY in the Figure). An inverse repeat of the non-palindrome/repeat complementary sequence is appended to the 3′-end of the complementary sequence to generate a self complementary DFO comprising sequence complementary to the nucleic acid target. The DFO can self-assemble to form a double stranded oligonucleotide.



FIG. 16 shows non-limiting examples of multifunctional siNA molecules of the invention comprising two separate polynucleotide sequences that are each capable of mediating RNAi directed cleavage of differing target nucleic acid sequences. FIG. 16A shows a non-limiting example of a multifunctional siNA molecule having a first region that is complementary to a first target nucleic acid sequence (complementary region 1) and a second region that is complementary to a second target nucleic acid sequence (complementary region 2), wherein the first and second complementary regions are situated at the 3′-ends of each polynucleotide sequence in the multifunctional siNA. The dashed portions of each polynucleotide sequence of the multifunctional siNA construct have complementarity with regard to corresponding portions of the siNA duplex, but do not have complementarity to the target nucleic acid sequences. FIG. 16B shows a non-limiting example of a multifunctional siNA molecule having a first region that is complementary to a first target nucleic acid sequence (complementary region 1) and a second region that is complementary to a second target nucleic acid sequence (complementary region 2), wherein the first and second complementary regions are situated at the 5′-ends of each polynucleotide sequence in the multifunctional siNA. The dashed portions of each polynucleotide sequence of the multifunctional siNA construct have complementarity with regard to corresponding portions of the siNA duplex, but do not have complementarity to the target nucleic acid sequences.



FIG. 17 shows non-limiting examples of multifunctional siNA molecules of the invention comprising a single polynucleotide sequence comprising distinct regions that are each capable of mediating RNAi directed cleavage of differing target nucleic acid sequences. FIG. 17A shows a non-limiting example of a multifunctional siNA molecule having a first region that is complementary to a first target nucleic acid sequence (complementary region 1) and a second region that is complementary to a second target nucleic acid sequence (complementary region 2), wherein the second complementary region is situated at the 3′-end of the polynucleotide sequence in the multifunctional siNA. The dashed portions of each polynucleotide sequence of the multifunctional siNA construct have complementarity with regard to corresponding portions of the siNA duplex, but do not have complementarity to the target nucleic acid sequences. FIG. 17B shows a non-limiting example of a multifunctional siNA molecule having a first region that is complementary to a first target nucleic acid sequence (complementary region 1) and a second region that is complementary to a second target nucleic acid sequence (complementary region 2), wherein the first complementary region is situated at the 5′-end of the polynucleotide sequence in the multifunctional siNA. The dashed portions of each polynucleotide sequence of the multifunctional siNA construct have complementarity with regard to corresponding portions of the siNA duplex, but do not have complementarity to the target nucleic acid sequences. In one embodiment, these multifunctional siNA constructs are processed in vivo or in vitro to generate multifunctional siNA constructs as shown in FIG. 16.



FIG. 18 shows non-limiting examples of multifunctional siNA molecules of the invention comprising two separate polynucleotide sequences that are each capable of mediating RNAi directed cleavage of differing target nucleic acid sequences and wherein the multifunctional siNA construct further comprises a self complementary, palindrome, or repeat region, thus enabling shorter bifuctional siNA constructs that can mediate RNA interference against differing target nucleic acid sequences. FIG. 18A shows a non-limiting example of a multifunctional siNA molecule having a first region that is complementary to a first target nucleic acid sequence (complementary region 1) and a second region that is complementary to a second target nucleic acid sequence (complementary region 2), wherein the first and second complementary regions are situated at the 3′-ends of each polynucleotide sequence in the multifunctional siNA, and wherein the first and second complementary regions further comprise a self complementary, palindrome, or repeat region. The dashed portions of each polynucleotide sequence of the multifunctional siNA construct have complementarity with regard to corresponding portions of the siNA duplex, but do not have complementarity to the target nucleic acid sequences. FIG. 18B shows a non-limiting example of a multifunctional siNA molecule having a first region that is complementary to a first target nucleic acid sequence (complementary region 1) and a second region that is complementary to a second target nucleic acid sequence (complementary region 2), wherein the first and second complementary regions are situated at the 5′-ends of each polynucleotide sequence in the multifunctional siNA, and wherein the first and second complementary regions further comprise a self complementary, palindrome, or repeat region. The dashed portions of each polynucleotide sequence of the multifunctional siNA construct have complementarity with regard to corresponding portions of the siNA duplex, but do not have complementarity to the target nucleic acid sequences.



FIG. 19 shows non-limiting examples of multifunctional siNA molecules of the invention comprising a single polynucleotide sequence comprising distinct regions that are each capable of mediating RNAi directed cleavage of differing target nucleic acid sequences and wherein the multifunctional siNA construct further comprises a self complementary, palindrome, or repeat region, thus enabling shorter bifuctional siNA constructs that can mediate RNA interference against differing target nucleic acid sequences. FIG. 19A shows a non-limiting example of a multifunctional siNA molecule having a first region that is complementary to a first target nucleic acid sequence (complementary region 1) and a second region that is complementary to a second target nucleic acid sequence (complementary region 2), wherein the second complementary region is situated at the 3′-end of the polynucleotide sequence in the multifunctional siNA, and wherein the first and second complementary regions further comprise a self complementary, palindrome, or repeat region. The dashed portions of each polynucleotide sequence of the multifunctional siNA construct have complementarity with regard to corresponding portions of the siNA duplex, but do not have complementarity to the target nucleic acid sequences. FIG. 19B shows a non-limiting example of a multifunctional siNA molecule having a first region that is complementary to a first target nucleic acid sequence (complementary region 1) and a second region that is complementary to a second target nucleic acid sequence (complementary region 2), wherein the first complementary region is situated at the 5′-end of the polynucleotide sequence in the multifunctional siNA, and wherein the first and second complementary regions further comprise a self complementary, palindrome, or repeat region. The dashed portions of each polynucleotide sequence of the multifunctional siNA construct have complementarity with regard to corresponding portions of the siNA duplex, but do not have complementarity to the target nucleic acid sequences. In one embodiment, these multifunctional siNA constructs are processed in vivo or in vitro to generate multifunctional siNA constructs as shown in FIG. 18.



FIG. 20 shows a non-limiting example of how multifunctional siNA molecules of the invention can target two separate target nucleic acid molecules, such as separate RNA molecules encoding differing proteins, for example, a cytokine and its corresponding receptor, differing viral strains, a virus and a cellular protein involved in viral infection or replication, or differing proteins involved in a common or divergent biologic pathway that is implicated in the maintenance of progression of disease. Each strand of the multifunctional siNA construct comprises a region having complementarity to separate target nucleic acid molecules. The multifunctional siNA molecule is designed such that each strand of the siNA can be utilized by the RISC complex to initiate RNA interference mediated cleavage of its corresponding target. These design parameters can include destabilization of each end of the siNA construct (see for example Schwarz et al., 2003, Cell, 115, 199-208). Such destabilization can be accomplished for example by using guanosine-cytidine base pairs, alternate base pairs (e.g., wobbles), or destabilizing chemically modified nucleotides at terminal nucleotide positions as is known in the art.



FIG. 21 shows a non-limiting example of how multifunctional siNA molecules of the invention can target two separate target nucleic acid sequences within the same target nucleic acid molecule, such as alternate coding regions of a RNA, coding and non-coding regions of a RNA, or alternate splice variant regions of a RNA. Each strand of the multifunctional siNA construct comprises a region having complementarity to the separate regions of the target nucleic acid molecule. The multifunctional siNA molecule is designed such that each strand of the siNA can be utilized by the RISC complex to initiate RNA interference mediated cleavage of its corresponding target region. These design parameters can include destabilization of each end of the siNA construct (see for example Schwarz et al., 2003, Cell, 115, 199-208). Such destabilization can be accomplished for example by using guanosine-cytidine base pairs, alternate base pairs (e.g., wobbles), or destabilizing chemically modified nucleotides at terminal nucleotide positions as is known in the art.



FIG. 22 shows non-limiting examples of inhibition of viral replication of a HCV/poliovirus chimera by siNA constructs targeted to HCV chimera (29579/29586; 29578/29585) compared to control (29593/29600).



FIG. 23 shows a non-limiting example of a dose response study demonstrating the inhibition of viral replication of a HCV/poliovirus chimera by siNA construct (29579/29586) at various concentrations (1 nM, 5 nM, 10 nM, and 25 nM) compared to control (29593/29600).



FIG. 24 shows a non-limiting example demonstrating the inhibition of viral replication of a HCV/poliovirus chimera by a chemically modified siRNA construct (30051/30053) compared to control construct (30052/30054).



FIG. 25 shows a non-limiting example demonstrating the inhibition of viral replication of a HCV/poliovirus chimera by a chemically modified siRNA construct (30055/30057) compared to control construct (30056/30058).



FIG. 26 shows a non-limiting example of several chemically modified siRNA constructs targeting viral replication of an HCV/poliovirus chimera at 10 nM treatment in comparison to a lipid control and an inverse siNA control construct 29593/29600.



FIG. 27 shows a non-limiting example of several chemically modified siRNA constructs targeting viral replication of a HCV/poliovirus chimera at 25 nM treatment in comparison to a lipid control and an inverse siNA control construct 29593/29600.



FIG. 28 shows a non-limiting example of several chemically modified siRNA constructs targeting viral replication of a Huh7 HCV replicon system at 25 nM treatment in comparison to untreated cells (“cells”), cells transfected with lipofectamine (“LFA2K”) and inverse siNA control constructs (“Inv”).



FIG. 29 shows a non-limiting example of a dose response study using chemically modified siNA molecules (Stab 4/5, see Table IV) targeting HCV RNA sites 291, 300, and 303 in a Huh7 HCV replicon system at 5, 10, 25, and 100 nM treatment in comparison to untreated cells (“cells”), cells transfected with lipofectamine (“LFA”) and inverse siNA control constructs (“Inv”).



FIG. 30 shows a non-limiting example of several chemically modified siNA constructs (Stab 7/8, see Table IV) targeting viral replication in a Huh7 HCV replicon system at 25 nM treatment in comparison to untreated cells (“cells”), cells transfected with lipofectamine (“Lipid”) and inverse siNA control constructs (“Inv”).



FIG. 31 shows a non-limiting example of a dose response study using chemically modified siNA molecules (Stab 7/8, see Table IV) targeting HCV site 327 in a Huh7 HCV replicon system at 5, 10, 25, 50, and 100 nM treatment in comparison to inverse siNA control constructs.



FIG. 32 shows the results of a study in which siNA/interferon combination treatments were assayed using 0-100 nM siNA in a HCV Subgenomic Replicon system in Huh7 cells compared to interferon alone.



FIG. 33 shows the results of a dose response study in which multifunctional siNAs targeting sites 304 and 327 (MF 36447/34588/38310) and sites 282 and 304 (MF 34588/36445/38311) of HCV RNA were evaluated with an irrelevant multifunctional siNA control (MF Control) and pools of individual siNA constructs targeting sites 304 (34583/34588) and 327 (34585/32201), and sites 282 (34581/34586) and 304 (34583/34588), along with untreated cells and a transfection control (LFA) from 0.1 to 10 nM. Compound numbers for the siNA constructs are shown in Table III. As shown in the figure, the multifunctional siNA constructs show equivalent activity to the corresponding pooled siNA constructs.



FIG. 34 shows the results of a dose response study in which chemically stabilized multifunctional siNAs targeting sites 282 and 304 (MF 38314/38294/38300) of HCV RNA was evaluated with individual siNA constructs targeting sites 282 (33139/38294) and 304 (33149/38300) and a pool of the individual siNA constructs targeting sites 282 (33139/38294) and 304 (33149/38300), along with untreated cells and a transfection control (LFA) from 0.1 to 25 nM. Compound numbers for the siNA constructs are shown in Table III. As shown in the figure, the multifunctional siNA constructs show equivalent activity to the corresponding individual and pooled siNA constructs.



FIG. 35 shows the results of a dose response study in which chemically stabilized multifunctional siNAs targeting sites 282 and 304 (MF 38314/38297/38300) of HCV RNA was evaluated with individual siNA constructs targeting site 282 (33139/38297) and a pool of individual siNA constructs targeting sites 282 (33139/38297) and 304 (33149/38300), along with untreated cells and a transfection control (LFA) from 0.1 to 25 nM. Compound numbers for the siNA constructs are shown in Table III. As shown in the figure, the multifunctional siNA constructs show equivalent activity to the corresponding individual and pooled siNA constructs.



FIG. 36 shows the results of a dose response study in which chemically stabilized multifunctional siNAs targeting sites 327 and 304 (MF 38312/37791/38300) of HCV RNA was evaluated with individual siNA constructs targeting site 327 (31703/37791) and a pool of individual siNA constructs targeting sites 327 (31703/37791) and 304 (33149/38300), along with untreated cells and a transfection control (LFA) from 0.1 to 25 nM. Compound numbers for the siNA constructs are shown in Table III. As shown in the figure, the multifunctional siNA constructs show equivalent activity to the corresponding individual and pooled siNA constructs.



FIG. 37 shows the results of a dose response study in which chemically stabilized multifunctional siNAs targeting sites 327 and 304 (MF 38312/38302/38300) of HCV RNA was evaluated with individual siNA constructs targeting site 327 (31703/38302) and a pool of individual siNA constructs targeting sites 327 (31703/38302) and 304 (33149/38300), along with untreated cells and a transfection control (LFA) from 0.1 to 25 nM. Compound numbers for the siNA constructs are shown in Table III. As shown in the figure, the multifunctional siNA constructs show equivalent activity to the corresponding individual and pooled siNA constructs.



FIG. 38 shows the results of a dose response study in which chemically stabilized multifunctional siNAs targeting sites 327 and 282 (MF 38313/38302/38297) of HCV RNA was evaluated a pool of individual siNA constructs targeting sites 327 (31703/37791) and 282 (33139/38294), and another pool of individual siNA constructs targeting sites 327 (31703/38302) and 282 (33139/38397) along with untreated cells and a transfection control (LFA) from 0.1 to 25 nM. Compound numbers for the siNA constructs are shown in Table III. As shown in the figure, the multifunctional siNA constructs show equivalent activity to the corresponding pooled siNA constructs.



FIG. 39 shows the results of a dose response study in which chemically stabilized multifunctional siNAs targeting sites 282 and 304 (MF 38314/38297/38300) of HCV RNA was evaluated a pool of individual siNA constructs targeting sites 282 (33139/38294) and 304 (33149/38300), and another pool of individual siNA constructs targeting sites 282 (33139/38297) and 304 (33149/38300) along with untreated cells and a transfection control (LFA) from 0.1 to 25 nM. Compound numbers for the siNA constructs are shown in Table III. As shown in the figure, the multifunctional siNA constructs show equivalent activity to the corresponding pooled siNA constructs.



FIG. 40 shows the results of a dose response study in which chemically stabilized multifunctional siNAs targeting sites 327 and 304 (MF 38312/38302/38300) of HCV RNA was evaluated a pool of individual siNA constructs targeting sites 327 (31703/37791) and 304 (33149/38300), and another pool of individual siNA constructs targeting sites 327 (31703/38302) and 304 (33149/38300) along with untreated cells and a transfection control (LFA) from 0.1 to 25 nM. Compound numbers for the siNA constructs are shown in Table III. As shown in the figure, the multifunctional siNA constructs show equivalent activity to the corresponding pooled siNA constructs.



FIG. 41 shows the results of a dose response study in which chemically stabilized multifunctional siNAs targeting sites 282 and 327 (MF 38313/38297/38302) of HCV RNA was evaluated a pool of individual siNA constructs targeting sites 282 (33139/38294) and 327 (31703/37791), and another pool of individual siNA constructs targeting sites 282 (33139/38297) and 327 (31703/38302) along with untreated cells and a transfection control (LFA) from 0.1 to 25 nM. Compound numbers for the siNA constructs are shown in Table III. As shown in the figure, the multifunctional siNA constructs show equivalent activity to the corresponding pooled siNA constructs.



FIGS. 42A-42H shows non-limiting examples of tethered multiifunctional siNA constructs of the invention. In the examples shown, a linker (e.g., nucleotide or non-nucleotide linker) connects two siNA regions (e.g., two sense, two antisense, or alternately a sense and an antisense region together. Separate sense (or sense and antisense) sequences corresponding to a first target sequence and second target sequence are hybridized to their corresponding sense and/or antisense sequences in the multifunctional siNA. In addition, various conjugates, ligands, aptamers, polymers or reporter molecules can be attached to the linker region for selective or improved delivery and/or pharmacokinetic properties.



FIG. 43 shows a non-limiting example of various dendrimer based multifunctional siNA designs.



FIG. 44 shows a non-limiting example of various supramolecular multifunctional siNA designs.



FIG. 45 shows a non-limiting example of a dicer enabled multifunctional siNA design using a 30 nucleotide precursor siNA construct. A 30 base pair duplex is cleaved by Dicer into 22 and 8 base pair products from either end (8 b.p. fragments not shown). For ease of presentation the overhangs generated by dicer are not shown—but can be compensated for. Three targeting sequences are shown. The required sequence identity overlapped is indicated by grey boxes. The N's of the parent 30 b.p. siNA are suggested sites of 2′-OH positions to enable Dicer cleavage if this is tested in stabilized chemistries. Note that processing of a 30mer duplex by Dicer RNase III does not give a precise 22+8 cleavage, but rather produces a series of closely related products (with 22+8 being the primary site). Therefore, processing by Dicer will yield a series of active siNAs.



FIG. 46 shows a non-limiting example of a dicer enabled multifunctional siNA design using a 40 nucleotide precursor siNA construct. A 40 base pair duplex is cleaved by Dicer into 20 base pair products from either end. For ease of presentation the overhangs generated by dicer are not shown—but can be compensated for. Four targeting sequences are shown. The required sequence identity overlapped is indicated by grey boxes. This design format can be extended to larger RNAs. If chemically stabilized siNAs are bound by Dicer, then strategically located ribonucleotide linkages can enable designer cleavage products that permit our more extensive repertoire of multiifunctional designs. For example cleavage products not limited to the Dicer standard of approximately 22-nucleotides can allow multifunctional siNA constructs with a target sequence identity overlap ranging from, for example, about 3 to about 15 nucleotides.



FIG. 47 shows a non-limiting example of inhibition of HBV RNA by dicer enabled multifunctional siNA constructs targeting HBV site 263. When the first 17 nucleotides of a siNA antisense strand (e.g., 21 nucleotide strands in a duplex with 3′-TT overhangs) are complementary to a target RNA, robust silencing was observed at 25 nM. 80% silencing was observed with only 16 nucleotide complementarity in the same format.



FIG. 48 shows a non-limiting example of additional multifunctional siNA construct designs of the invention. In one example, a conjugate, ligand, aptamer, label, or other moiety is attached to a region of the multifunctional siNA to enable improved delivery or pharmacokinetic profiling.



FIG. 49 shows a non-limiting example of additional multifunctional siNA construct designs of the invention. In one example, a conjugate, ligand, aptamer, label, or other moiety is attached to a region of the multifunctional siNA to enable improved delivery or pharmacokinetic profiling.




DETAILED DESCRIPTION OF THE INVENTION

Mechanism of Action of Nucleic Acid Molecules of the Invention


The discussion that follows discusses the proposed mechanism of RNA interference mediated by short interfering RNA as is presently known, and is not meant to be limiting and is not an admission of prior art. Applicant demonstrates herein that chemically-modified short interfering nucleic acids possess similar or improved capacity to mediate RNAi as do siRNA molecules and are expected to possess improved stability and activity in vivo; therefore, this discussion is not meant to be limiting only to siRNA and can be applied to siNA as a whole. By “improved capacity to mediate RNAi” or “improved RNAi activity” is meant to include RNAi activity measured in vitro and/or in vivo where the RNAi activity is a reflection of both the ability of the siNA to mediate RNAi and the stability of the siNAs of the invention. In this invention, the product of these activities can be increased in vitro and/or in vivo compared to an all RNA siRNA or a siNA containing a plurality of ribonucleotides. In some cases, the activity or stability of the siNA molecule can be decreased (i.e., less than ten-fold), but the overall activity of the siNA molecule is enhanced in vitro and/or in vivo.


RNA interference refers to the process of sequence specific post-transcriptional gene silencing in animals mediated by short interfering RNAs (siRNAs) (Fire et al., 1998, Nature, 391, 806). The corresponding process in plants is commonly referred to as post-transcriptional gene silencing or RNA silencing and is also referred to as quelling in fungi. The process of post-transcriptional gene silencing is thought to be an evolutionarily-conserved cellular defense mechanism used to prevent the expression of foreign genes which is commonly shared by diverse flora and phyla (Fire et al., 1999, Trends Genet., 15, 358). Such protection from foreign gene expression may have evolved in response to the production of double-stranded RNAs (dsRNAs) derived from viral infection or the random integration of transposon elements into a host genome via a cellular response that specifically destroys homologous single-stranded RNA or viral genomic RNA. The presence of dsRNA in cells triggers the RNAi response though a mechanism that has yet to be fully characterized. This mechanism appears to be different from the interferon response that results from dsRNA-mediated activation of protein kinase PKR and 2′,5′-oligoadenylate synthetase resulting in non-specific cleavage of mRNA by ribonuclease L.


The presence of long dsRNAs in cells stimulates the activity of a ribonuclease III enzyme referred to as Dicer. Dicer is involved in the processing of the dsRNA into short pieces of dsRNA known as short interfering RNAs (siRNAs) (Berstein et al., 2001, Nature, 409, 363). Short interfering RNAs derived from Dicer activity are typically about 21 to about 23 nucleotides in length and comprise about 19 base pair duplexes. Dicer has also been implicated in the excision of 21- and 22-nucleotide small temporal RNAs (stRNAs) from precursor RNA of conserved structure that are implicated in translational control (Hutvagner et al., 2001, Science, 293, 834). The RNAi response also features an endonuclease complex containing a siRNA, commonly referred to as an RNA-induced silencing complex (RISC), which mediates cleavage of single-stranded RNA having sequence homologous to the siRNA. Cleavage of the target RNA takes place in the middle of the region complementary to the guide sequence of the siRNA duplex (Elbashir et al., 2001, Genes Dev., 15, 188). In addition, RNA interference can also involve small RNA (e.g., micro-RNA or miRNA) mediated gene silencing, presumably though cellular mechanisms that regulate chromatin structure and thereby prevent transcription of target gene sequences (see for example Allshire, 2002, Science, 297, 1818-1819; Volpe et al., 2002, Science, 297, 1833-1837; Jenuwein, 2002, Science, 297, 2215-2218; and Hall et al., 2002, Science, 297, 2232-2237). As such, siNA molecules of the invention can be used to mediate gene silencing via interaction with RNA transcripts or alternately by interaction with particular gene sequences, wherein such interaction results in gene silencing either at the transcriptional level or post-transcriptional level.


RNAi has been studied in a variety of systems. Fire et al., 1998, Nature, 391, 806, were the first to observe RNAi in C. elegans. Wianny and Goetz, 1999, Nature Cell Biol., 2, 70, describe RNAi mediated by dsRNA in mouse embryos. Hammond et al., 2000, Nature, 404, 293, describe RNAi in Drosophila cells transfected with dsRNA. Elbashir et al., 2001, Nature, 411, 494, describe RNAi induced by introduction of duplexes of synthetic 21-nucleotide RNAs in cultured mammalian cells including human embryonic kidney and HeLa cells. Recent work in Drosophila embryonic lysates has revealed certain requirements for siRNA length, structure, chemical composition, and sequence that are essential to mediate efficient RNAi activity. These studies have shown that 21 nucleotide siRNA duplexes are most active when containing two 2-nucleotide 3′-terminal nucleotide overhangs. Furthermore, substitution of one or both siRNA strands with 2′-deoxy or 2′-O-methyl nucleotides abolishes RNAi activity, whereas substitution of 3′-terminal siRNA nucleotides with deoxy nucleotides was shown to be tolerated. Mismatch sequences in the center of the siRNA duplex were also shown to abolish RNAi activity. In addition, these studies also indicate that the position of the cleavage site in the target RNA is defined by the 5′-end of the siRNA guide sequence rather than the 3′-end (Elbashir et al., 2001, EMBO J., 20, 6877). Other studies have indicated that a 5′-phosphate on the target-complementary strand of a siRNA duplex is required for siRNA activity and that ATP is utilized to maintain the 5′-phosphate moiety on the siRNA (Nykanen et al., 2001, Cell, 107, 309); however, siRNA molecules lacking a 5′-phosphate are active when introduced exogenously, suggesting that 5′-phosphorylation of siRNA constructs may occur in vivo.


Duplex Forming Oligonucleotides (DFO) of the Invention


In one embodiment, the invention features siNA molecules comprising duplex forming oligonucleotides (DFO) that can self-assemble into double stranded oligonucleotides. The duplex forming oligonucleotides of the invention can be chemically synthesized or expressed from transcription units and/or vectors. The DFO molecules of the instant invention provide useful reagents and methods for a variety of therapeutic, diagnostic, agricultural, veterinary, target validation, genomic discovery, genetic engineering and pharmacogenomic applications.


Applicant demonstrates herein that certain oligonucleotides, refered to herein for convenience but not limitation as duplex forming oligonucleotides or DFO molecules, are potent mediators of sequence specific regulation of gene expression. The oligonucleotides of the invention are distinct from other nucleic acid sequences known in the art (e.g., siRNA, miRNA, stRNA, shRNA, antisense oligonucleotides etc.) in that they represent a class of linear polynucleotide sequences that are designed to self-assemble into double stranded oligonucleotides, where each strand in the double stranded oligonucleotides comprises a nucleotide sequence that is complementary to a target nucleic acid molecule. Nucleic acid molecules of the invention can thus self assemble into functional duplexes in which each strand of the duplex comprises the same polynucleotide sequence and each strand comprises a nucleotide sequence that is complementary to a target nucleic acid molecule.


Generally, double stranded oligonucleotides are formed by the assembly of two distinct oligonucleotide sequences where the oligonucleotide sequence of one strand is complementary to the oligonucleotide sequence of the second strand; such double stranded oligonucleotides are assembled from two separate oligonucleotides, or from a single molecule that folds on itself to form a double stranded structure, often referred to in the field as hairpin stem-loop structure (e.g., shRNA or short hairpin RNA). These double stranded oligonucleotides known in the art all have a common feature in that each strand of the duplex has a distict nucleotide sequence.


Distinct from the double stranded nucleic acid molecules known in the art, the applicants have developed a novel, potentially cost effective and simplified method of forming a double stranded nucleic acid molecule starting from a single stranded or linear oligonucleotide. The two strands of the double stranded oligonucleotide formed according to the instant invention have the same nucleotide sequence and are not covalently linked to each other. Such double-stranded oligonucleotides molecules can be readily linked post-synthetically by methods and reagents known in the art and are within the scope of the invention. In one embodiment, the single stranded oligonucleotide of the invention (the duplex forming oligonucleotide) that forms a double stranded oligonucleotide comprises a first region and a second region, where the second region includes a nucleotide sequence that is an inverted repeat of the nucleotide sequence in the first region, or a portion thereof, such that the single stranded oligonucleotide self assembles to form a duplex oligonucleotide in which the nucleotide sequence of one strand of the duplex is the same as the nucleotide sequence of the second strand. Non-limiting examples of such duplex forming oligonucleotides are illustrated in FIGS. 14 and 15. These duplex forming oligonucleotides (DFOs) can optionally include certain palindrome or repeat sequences where such palindrome or repeat sequences are present in between the first region and the second region of the DFO.


In one embodiment, the invention features a duplex forming oligonucleotide (DFO) molecule, wherein the DFO comprises a duplex forming self complementary nucleic acid sequence that has nucleotide sequence complementary to a HCV target nucleic acid sequence. The DFO molecule can comprise a single self complementary sequence or a duplex resulting from assembly of such self complementary sequences.


In one embodiment, a duplex forming oligonucleotide (DFO) of the invention comprises a first region and a second region, wherein the second region comprises a nucleotide sequence comprising an inverted repeat of nucleotide sequence of the first region such that the DFO molecule can assemble into a double stranded oligonucleotide. Such double stranded oligonucleotides can act as a short interfering nucleic acid (siNA) to modulate gene expression. Each strand of the double stranded oligonucleotide duplex formed by DFO molecules of the invention can comprise a nucleotide sequence region that is complementary to the same nucleotide sequence in a target nucleic acid molecule (e.g., target HCV RNA).


In one embodiment, the invention features a single stranded DFO that can assemble into a double stranded oligonucleotide. The applicant has surprisingly found that a single stranded oligonucleotide with nucleotide regions of self complementarity can readily assemble into duplex oligonucleotide constructs. Such DFOs can assemble into duplexes that can inhibit gene expression in a sequence specific manner. The DFO moleucles of the invention comprise a first region with nucleotide sequence that is complementary to the nucleotide sequence of a second region and where the sequence of the first region is complementary to a target nucleic acid (e.g., RNA). The DFO can form a double stranded oligonucleotide wherein a portion of each strand of the double stranded oligonucleotide comprises a sequence complementary to a target nucleic acid sequence.


In one embodiment, the invention features a double stranded oligonucleotide, wherein the two strands of the double stranded oligonucleotide are not covalently linked to each other, and wherein each strand of the double stranded oligonucleotide comprises a nucleotide sequence that is complementary to the same nucleotide sequence in a target nucleic acid molecule or a portion thereof (e.g., HCV RNA target). In another embodiment, the two strands of the double stranded oligonucleotide share an identical nucleotide sequence of at least about 15, preferably at least about 16, 17, 18, 19, 20, or 21 nucleotides.


In one embodiment, a DFO molecule of the invention comprises a structure having Formula DFO-I:

5′-p-XZX′-3′

wherein Z comprises a palindromic or repeat nucleic acid sequence optionally with one or more modified nucleotides (e.g., nucleotide with a modified base, such as 2-amino purine, 2-amino-1,6-dihydro purine or a universal base), for example of length about 2 to about 24 nucleotides in even numbers (e.g., about 2, 4, 6, 8, 10, 12, 14, 16, 18, 20, or 22 or 24 nucleotides), X represents a nucleic acid sequence, for example of length between about 1 to about 21 nucleotides (e.g., about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, or 21 nucleotides), X′ comprises a nucleic acid sequence, for example of length about 1 and about 21 nucleotides (e.g., about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20 or 21 nucleotides) having nucleotide sequence complementarity to sequence X or a portion thereof, p comprises a terminal phosphate group that can be present or absent, and wherein sequence X and Z, either independently or together, comprise nucleotide sequence that is complementary to a target nucleic acid sequence or a portion thereof and is of length sufficient to interact (e.g., base pair) with the target nucleic acid sequence or a portion thereof (e.g., HCV RNA target). For example, X independently can comprise a sequence from about 12 to about 21 or more (e.g., about 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, or more) nucleotides in length that is complementary to nucleotide sequence in a target HCV RNA or a portion thereof. In another non-limiting example, the length of the nucleotide sequence of X and Z together, when X is present, that is complementary to the target RNA or a portion thereof (e.g., HCV RNA target) is from about 12 to about 21 or more nucleotides (e.g., about 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, or more). In yet another non-limiting example, when X is absent, the length of the nucleotide sequence of Z that is complementary to the target HCV RNA or a portion thereof is from about 12 to about 24 or more nucleotides (e.g., about 12, 14, 16, 18, 20, 22, 24, or more). In one embodiment X, Z and X′ are independently oligonucleotides, where X and/or Z comprises a nucleotide sequence of length sufficient to interact (e.g., base pair) with a nucleotide sequence in the target RNA or a portion thereof (e.g., HCV RNA target). In one embodiment, the lengths of oligonucleotides X and X′ are identical. In another embodiment, the lengths of oligonucleotides X and X′ are not identical. In another embodiment, the lengths of oligonucleotides X and Z, or Z and X′, or X, Z and X′ are either identical or different.


When a sequence is described in this specification as being of “sufficient” length to interact (i.e., base pair) with another sequence, it is meant that the the length is such that the number of bonds (e.g., hydrogen bonds) formed between the two sequences is enough to enable the two sequence to form a duplex under the conditions of interest. Such conditions can be in vitro (e.g., for diagnostic or assay purposes) or in vivo (e.g., for therapeutic purposes). It is a simple and routine matter to determine such lengths.


In one embodiment, the invention features a double stranded oligonucleotide construct having Formula DFO-I(a):

5′-p-X Z X′-3′  3′-X′ Z X-p-5′


wherein Z comprises a palindromic or repeat nucleic acid sequence or palindromic or repeat-like nucleic acid sequence with one or more modified nucleotides (e.g., nucleotides with a modified base, such as 2-amino purine, 2-amino-1,6-dihydro purine or a universal base), for example of length about 2 to about 24 nucleotides in even numbers (e.g., about 2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22 or 24 nucleotides), X represents a nucleic acid sequence, for example of length about 1 to about 21 nucleotides (e.g., about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, or 21 nucleotides), X′ comprises a nucleic acid sequence, for example of length about 1 to about 21 nucleotides (e.g., about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20 or 21 nucleotides) having nucleotide sequence complementarity to sequence X or a portion thereof, p comprises a terminal phosphate group that can be present or absent, and wherein each X and Z independently comprises a nucleotide sequence that is complementary to a target nucleic acid sequence or a portion thereof (e.g., HCV RNA target) and is of length sufficient to interact with the target nucleic acid sequence of a portion thereof (e.g., HCV RNA target). For example, sequence X independently can comprise a sequence from about 12 to about 21 or more nucleotides (e.g., about 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, or more) in length that is complementary to a nucleotide sequence in a target RNA or a portion thereof (e.g., HCV RNA target). In another non-limiting example, the length of the nucleotide sequence of X and Z together (when X is present) that is complementary to the target HCV RNA or a portion thereof is from about 12 to about 21 or more nucleotides (e.g., about 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, or more). In yet another non-limiting example, when X is absent, the length of the nucleotide sequence of Z that is complementary to the target HCV RNA or a portion thereof is from about 12 to about 24 or more nucleotides (e.g., about 12, 14, 16, 18, 20, 22, 24 or more). In one embodiment X, Z and X′ are independently oligonucleotides, where X and/or Z comprises a nucleotide sequence of length sufficient to interact (e.g., base pair) with nucleotide sequence in the target RNA or a portion thereof (e.g., HCV RNA target). In one embodiment, the lengths of oligonucleotides X and X′ are identical. In another embodiment, the lengths of oligonucleotides X and X′ are not identical. In another embodiment, the lengths of oligonucleotides X and Z or Z and X′ or X, Z and X′ are either identical or different. In one embodiment, the double stranded oligonucleotide construct of Formula I(a) includes one or more, specifically 1, 2, 3 or 4, mismatches, to the extent such mismatches do not significantly diminish the ability of the double stranded oligonucleotide to inhibit target gene expression.


In one embodiment, a DFO molecule of the invention comprises structure having Formula DFO-II:

5′-p-XX′-3′

wherein each X and X′ are independently oligonucleotides of length about 12 nucleotides to about 21 nucleotides, wherein X comprises, for example, a nucleic acid sequence of length about 12 to about 21 nucleotides (e.g., about 12, 13, 14, 15, 16, 17, 18, 19, 20 or 21 nucleotides), X′ comprises a nucleic acid sequence, for example of length about 12 to about 21 nucleotides (e.g., about 12, 13, 14, 15, 16, 17, 18, 19, 20, or 21 nucleotides) having nucleotide sequence complementarity to sequence X or a portion thereof, p comprises a terminal phosphate group that can be present or absent, and wherein X comprises a nucleotide sequence that is complementary to a target nucleic acid sequence (e.g., HCV RNA) or a portion thereof and is of length sufficient to interact (e.g., base pair) with the target nucleic acid sequence of a portion thereof. In one embodiment, the length of oligonucleotides X and X′ are identical. In another embodiment the length of oligonucleotides X and X′ are not identical. In one embodiment, length of the oligonucleotides X and X′ are sufficint to form a relatively stable double stranded oligonucleotide.


In one embodiment, the invention features a double stranded oligonucleotide construct having Formula DFO-II(a):

5′-p-X X′-3′  3′-X′ X-p-5′


wherein each X and X′ are independently oligonucleotides of length about 12 nucleotides to about 21 nucleotides, wherein X comprises a nucleic acid sequence, for example of length about 12 to about 21 nucleotides (e.g., about 12, 13, 14, 15, 16, 17, 18, 19, 20 or 21 nucleotides), X′ comprises a nucleic acid sequence, for example of length about 12 to about 21 nucleotides (e.g., about 12, 13, 14, 15, 16, 17, 18, 19, 20 or 21 nucleotides) having nucleotide sequence complementarity to sequence X or a portion thereof, p comprises a terminal phosphate group that can be present or absent, and wherein X comprises nucleotide sequence that is complementary to a target nucleic acid sequence or a portion thereof (e.g., HCV RNA target) and is of length sufficient to interact (e.g., base pair) with the target nucleic acid sequence (e.g., HCV RNA) or a portion thereof. In one embodiment, the lengths of oligonucleotides X and X′ are identical. In another embodiment, the lengths of oligonucleotides X and X′ are not identical. In one embodiment, the lengths of the oligonucleotides X and X′ are sufficint to form a relatively stable double stranded oligonucleotide. In one embodiment, the double stranded oligonucleotide construct of Formula 11(a) includes one or more, specifically 1, 2, 3 or 4, mismatches, to the extent such mismatches do not significantly diminish the ability of the double stranded oligonucleotide to inhibit target gene expression.


In one embodiment, the invention features a DFO molecule having Formula DFO-I(b):

5′-p-Z-3′

where Z comprises a palindromic or repeat nucleic acid sequence optionally including one or more non-standard or modified nucleotides (e.g., nucleotide with a modified base, such as 2-amino purine or a universal base) that can facilitate base-pairing with other nucleotides. Z can be, for example, of length sufficient to interact (e.g., base pair) with nucleotide sequence of a target nucleic acid (e.g., HCV RNA) molecule, preferably of length of at least 12 nucleotides, specifically about 12 to about 24 nucleotides (e.g., about 12, 14, 16, 18, 20, 22 or 24 nucleotides). p represents a terminal phosphate group that can be present or absent.


In one embodiment, a DFO molecule having any of Formula DFO-I, DFO-I(a), DFO-I(b), DFO-II(a) or DFO-II can comprise chemical modifications as described herein without limitation, such as, for example, nucleotides having any of Formulae I-VII, stabilization chemistries as described in Table IV, or any other combination of modified nucleotides and non-nucleotides as described in the various embodiments herein.


In one embodiment, the palidrome or repeat sequence or modified nucleotide (e.g., nucleotide with a modified base, such as 2-amino purine or a universal base) in Z of DFO constructs having Formula DFO-I, DFO-I(a) and DFO-I(b), comprises chemically modified nucleotides that are able to interact with a portion of the target nucleic acid sequence (e.g., modified base analogs that can form Watson Crick base pairs or non-Watson Crick base pairs).


In one embodiment, a DFO molecule of the invention, for example a DFO having Formula DFO-I or DFO-II, comprises about 15 to about 40 nucleotides (e.g., about 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, or 40 nucleotides). In one embodiment, a DFO molecule of the invention comprises one or more chemical modifications. In a non-limiting example, the introduction of chemically modified nucleotides and/or non-nucleotides into nucleic acid molecules of the invention provides a powerful tool in overcoming potential limitations of in vivo stability and bioavailability inherent to unmodified RNA molecules that are delivered exogenously. For example, the use of chemically modified nucleic acid molecules can enable a lower dose of a particular nucleic acid molecule for a given therapeutic effect since chemically modified nucleic acid molecules tend to have a longer half-life in serum or in cells or tissues. Furthermore, certain chemical modifications can improve the bioavailability and/or potency of nucleic acid molecules by not only enhancing half-life but also facilitating the targeting of nucleic acid molecules to particular organs, cells or tissues and/or improving cellular uptake of the nucleic acid molecules. Therefore, even if the activity of a chemically modified nucleic acid molecule is reduced in vitro as compared to a native/unmodified nucleic acid molecule, for example when compared to an unmodified RNA molecule, the overall activity of the modified nucleic acid molecule can be greater than the native or unmodified nucleic acid molecule due to improved stability, potency, duration of effect, bioavailability and/or delivery of the molecule.


Multifunctional or Multi-Targeted siNA Molecules of the Invention In one embodiment, the invention features siNA molecules comprising multifunctional short interfering nucleic acid (multifunctional siNA) molecules that modulate the expression of one or more genes in a biologic system, such as a cell, tissue, or organism. The multifunctional short interfering nucleic acid (multifunctional siNA) molecules of the invention can target more than one region of the HCV or cellular/host target nucleic acid sequence or can target sequences of more than one distinct target nucleic acid molecules (e.g., HCV RNA or cellular/host RNA targets). The multifunctional siNA molecules of the invention can be chemically synthesized or expressed from transcription units and/or vectors. The multifunctional siNA molecules of the instant invention provide useful reagents and methods for a variety of human applications, therapeutic, diagnostic, agricultural, veterinary, target validation, genomic discovery, genetic engineering and pharmacogenomic applications.


Applicant demonstrates herein that certain oligonucleotides, refered to herein for convenience but not limitation as multifunctional short interfering nucleic acid or multifunctional siNA molecules, are potent mediators of sequence specific regulation of gene expression. The multifunctional siNA molecules of the invention are distinct from other nucleic acid sequences known in the art (e.g., siRNA, miRNA, stRNA, shRNA, antisense oligonucleotides, etc.) in that they represent a class of polynucleotide molecules that are designed such that each strand in the multifunctional siNA construct comprises a nucleotide sequence that is complementary to a distinct nucleic acid sequence in one or more target nucleic acid molecules. A single multifunctional siNA molecule (generally a double-stranded molecule) of the invention can thus target more than one (e.g., 2, 3, 4, 5, or more) differing target nucleic acid target molecules. Nucleic acid molecules of the invention can also target more than one (e.g., 2, 3, 4, 5, or more) region of the same target nucleic acid sequence. As such multifunctional siNA molecules of the invention are useful in down regulating or inhibiting the expression of one or more target nucleic acid molecules. For example, a multifunctional siNA molecule of the invention can target nucleic acid molecules encoding a virus or viral proteins and corresponding cellular proteins required for viral infection and/or replication, or differing strains of a particular virus (e.g., HCV). By reducing or inhibiting expression of more than one target nucleic acid molecule with one multifunctional siNA construct, multifunctional siNA molecules of the invention represent a class of potent therapeutic agents that can provide simultaneous inhibition of multiple targets within a disease or pathogen related pathway. Such simultaneous inhibition can provide synergistic therapeutic treatment strategies without the need for separate preclinical and clinical development efforts or complex regulatory approval process.


Use of multifunctional siNA molecules that target more then one region of a target nucleic acid molecule (e.g., messenger RNA or HCV RNA) is expected to provide potent inhibition of gene expression. For example, a single multifunctional siNA construct of the invention can target both conserved and variable regions of a target nucleic acid molecule (e.g., HCV RNA), thereby allowing down regulation or inhibition of different strain variants or a virus, or splice variants encoded by a single host gene, or allowing for targeting of both coding and non-coding regions of the host target nucleic acid molecule.


Generally, double stranded oligonucleotides are formed by the assembly of two distinct oligonucleotides where the oligonucleotide sequence of one strand is complementary to the oligonucleotide sequence of the second strand; such double stranded oligonucleotides are generally assembled from two separate oligonucleotides (e.g., siRNA). Alternately, a duplex can be formed from a single molecule that folds on itself (e.g., shRNA or short hairpin RNA). These double stranded oligonucleotides are known in the art to mediate RNA interference and all have a common feature wherein only one nucleotide sequence region (guide sequence or the antisense sequence) has complementarity to a target nucleic acid sequence (e.g., HCV or host RNA) and the other strand (sense sequence) comprises nucleotide sequence that is homologous to the target nucleic acid sequence. Generally, the antisense sequence is retained in the active RISC complex and guides the RISC to the target nucleotide sequence by means of complementary base-pairing of the antisense sequence with the target seqeunce for mediating sequence-specific RNA interference. It is known in the art that in some cell culture systems, certain types of unmodified siRNAs can exhibit “off target” effects. It is hypothesized that this off-target effect involves the participation of the sense sequence instead of the antisense sequence of the siRNA in the RISC complex (see for example Schwarz et al., 2003, Cell, 115, 199-208). In this instance the sense sequence is believed to direct the RISC complex to a sequence (off-target sequence) that is distinct from the intended target sequence, resulting in the inhibition of the off-target sequence. In these double stranded nucleic acid molecules, each strand is complementary to a distinct target nucleic acid sequence. However, the off-targets that are affected by these dsRNAs are not entirely predictable and are non-specific.


Distinct from the double stranded nucleic acid molecules known in the art, the applicants have developed a novel, potentially cost effective and simplified method of down regulating or inhibiting the expression of more than one target nucleic acid sequence using a single multifunctional siNA construct. The multifunctional siNA molecules of the invention are designed to be double-stranded or partially double stranded, such that a portion of each strand or region of the multifunctional siNA is complementary to a target nucleic acid sequence of choice. As such, the multifunctional siNA molecules of the invention are not limited to targeting sequences that are complementary to each other, but rather to any two differing target nucleic acid sequences. Multifunctional siNA molecules of the invention are designed such that each strand or region of the multifunctional siNA molecule, that is complementary to a given target nucleic acid sequence, is of suitable length (e.g., from about 16 to about 28 nucleotides in length, preferably from about 18 to about 28 nucleotides in length) for mediating RNA interference against the target nucleic acid sequence. The complementarity between the target nucleic acid sequence and a strand or region of the multifunctional siNA must be sufficient (at least about 8 base pairs) for cleavage of the target nucleic acid sequence by RNA interference multifunctional siNA of the invention is expected to minimize off-target effects seen with certain siRNA sequences, such as those described in (Schwarz et al., supra).


It has been reported that dsRNAs of length between 29 base pairs and 36 base pairs (Tuschl et al., International PCT Publication No. WO 02/44321) do not mediate RNAi. One reason these dsRNAs are inactive may be the lack of turnover or dissociation of the strand that interacts with the target RNA sequence, such that the RISC complex is not able to efficiently interact with multiple copies of the target RNA resulting in a significant decrease in the potency and efficiency of the RNAi process. Applicant has surprisingly found that the multifunctional siNAs of the invention can overcome this hurdle and are capable of enhancing the efficiency and potency of RNAi process. As such, in certain embodiments of the invention, multifunctional siNAs of length between about 29 to about 36 base pairs can be designed such that, a portion of each strand of the multifunctional siNA molecule comprises a nucleotide sequence region that is complementary to a target nucleic acid of length sufficient to mediate RNAi efficiently (e.g., about 15 to about 23 base pairs) and a nucleotide sequence region that is not complementary to the target nucleic acid. By having both complementary and non-complementary portions in each strand of the multifunctional siNA, the multifunctional siNA can mediate RNA interference against a target nucleic acid sequence without being prohibitive to turnover or dissociation (e.g., where the length of each strand is too long to mediate RNAi against the respective target nucleic acid sequence). Furthermore, design of multifunctional siNA molecules of the invention with internal overlapping regions allows the multifunctional siNA molecules to be of favorable (decreased) size for mediating RNA interference and of size that is well suited for use as a therapeutic agent (e.g., wherein each strand is independently from about 18 to about 28 nucleotides in length). Non-limiting examples are lillustrated in the enclosed FIGS. 16-21 and 42.


In one embodiment, a multifunctional siNA molecule of the invention comprises a first region and a second region, where the first region of the multifunctional siNA comprises a nucleotide sequence complementary to a nucleic acid sequence of a first target nucleic acid molecule, and the second region of the multifunctional siNA comprises nucleic acid sequence complementary to a nucleic acid sequence of a second target nucleic acid molecule. In one embodiment, a multifunctional siNA molecule of the invention comprises a first region and a second region, where the first region of the multifunctional siNA comprises nucleotide sequence complementary to a nucleic acid sequence of the first region of a target nucleic acid molecule, and the second region of the multifunctional siNA comprises nucleotide sequence complementary to a nucleic acid sequence of a second region of a the target nucleic acid molecule. In another embodiment, the first region and second region of the multifunctional siNA can comprise separate nucleic acid sequences that share some degree of complementarity (e.g., from about 1 to about 10 complementary nucleotides). In certain embodiments, multifunctional siNA constructs comprising separate nucleic acid seqeunces can be readily linked post-synthetically by methods and reagents known in the art and such linked constructs are within the scope of the invention. Alternately, the first region and second region of the multifunctional siNA can comprise a single nucleic acid sequence having some degree of self complementarity, such as in a hairpin or stem-loop structure. Non-limiting examples of such double stranded and hairpin multifunctional short interfering nucleic acids are illustrated in FIGS. 16 and 17 respectively. These multifunctional short interfering nucleic acids (multifunctional siNAs) can optionally include certain overlapping nucleotide sequence where such overlapping nucleotide sequence is present in between the first region and the second region of the multifunctional siNA (see for example FIGS. 18 and 19).


In one embodiment, the invention features a multifunctional short interfering nucleic acid (multifunctional siNA) molecule, wherein each strand of the the multifunctional siNA independently comprises a first region of nucleic acid sequence that is complementary to a distinct target nucleic acid sequence and the second region of nucleotide sequence that is not complementary to the target sequence. The target nucleic acid sequence of each strand is in the same target nucleic acid molecule or different target nucleic acid molecules.


In another embodiment, the multifunctional siNA comprises two strands, where: (a) the first strand comprises a region having sequence complementarity to a target nucleic acid sequence (complementary region 1) and a region having no sequence complementarity to the target nucleotide sequence (non-complementary region 1); (b) the second strand of the multifunction siNA comprises a region having sequence complementarity to a target nucleic acid sequence that is distinct from the target nucleotide sequence complementary to the first strand nucleotide sequence (complementary region 2), and a region having no sequence complementarity to the target nucleotide sequence of complementary region 2 (non-complementary region 2); (c) the complementary region 1 of the first strand comprises a nucleotide sequence that is complementary to a nucleotide sequence in the non-complementary region 2 of the second strand and the complementary region 2 of the second strand comprises a nucleotide sequence that is complementary to a nucleotide sequence in the non-complementary region 1 of the first strand. The target nucleic acid sequence of complementary region 1 and complementary region 2 is in the same target nucleic acid molecule or different target nucleic acid molecules.


In another embodiment, the multifunctional siNA comprises two strands, where: (a) the first strand comprises a region having sequence complementarity to a target nucleic acid sequence derived from a gene (e.g., HCV or host gene) (complementary region 1) and a region having no sequence complementarity to the target nucleotide sequence of complementary region 1 (non-complementary region 1); (b) the second strand of the multifunction siNA comprises a region having sequence complementarity to a target nucleic acid sequence derived from a gene that is distinct from the gene of complementary region 1 (complementary region 2), and a region having no sequence complementarity to the target nucleotide sequence of complementary region 2 (non-complementary region 2); (c) the complementary region 1 of the first strand comprises a nucleotide sequence that is complementary to a nucleotide sequence in the non-complementary region 2 of the second strand and the complementary region 2 of the second strand comprises a nucleotide sequence that is complementary to a nucleotide sequence in the non-complementary region 1 of the first strand.


In another embodiment, the multifunctional siNA comprises two strands, where: (a) the first strand comprises a region having sequence complementarity to a target nucleic acid sequence derived from a gene (e.g., HCV or host gene) (complementary region 1) and a region having no sequence complementarity to the target nucleotide sequence of complementary region 1 (non-complementary region 1); (b) the second strand of the multifunction siNA comprises a region having sequence complementarity to a target nucleic acid sequence distinct from the target nucleic acid sequence of complementary region 1(complementary region 2), provided, however, that the target nucleic acid sequence for complementary region 1 and target nucleic acid sequence for complementary region 2 are both derived from the same gene, and a region having no sequence complementarity to the target nucleotide sequence of complementary region 2 (non-complementary region 2); (c) the complementary region 1 of the first strand comprises a nucleotide sequence that is complementary to a nucleotide sequence in the non-complementary region 2 of the second strand and the complementary region 2 of the second strand comprises a nucleotide sequence that is complementary to nucleotide sequence in the non-complementary region 1 of the first strand.


In one embodiment, the invention features a multifunctional short interfering nucleic acid (multifunctional siNA) molecule, wherein the multifunctional siNA comprises two complementary nucleic acid sequences in which the first sequence comprises a first region having nucleotide sequence complementary to nucleotide sequence within a target nucleic acid molecule, and in which the second seqeunce comprises a first region having nucleotide sequence complementary to a distinct nucleotide sequence within the same target nucleic acid molecule. Preferably, the first region of the first sequence is also complementary to the nucleotide sequence of the second region of the second sequence, and where the first region of the second sequence is complementary to the nucleotide sequence of the second region of the first sequence,


In one embodiment, the invention features a multifunctional short interfering nucleic acid (multifunctional siNA) molecule, wherein the multifunctional siNA comprises two complementary nucleic acid sequences in which the first sequence comprises a first region having a nucleotide sequence complementary to a nucleotide sequence within a first target nucleic acid molecule, and in which the second seqeunce comprises a first region having a nucleotide sequence complementary to a distinct nucleotide sequence within a second target nucleic acid molecule. Preferably, the first region of the first sequence is also complementary to the nucleotide sequence of the second region of the second sequence, and where the first region of the second sequence is complementary to the nucleotide sequence of the second region of the first sequence,


In one embodiment, the invention features a multifunctional siNA molecule comprising a first region and a second region, where the first region comprises a nucleic acid sequence having about 18 to about 28 nucleotides complementary to a nucleic acid sequence within a first target nucleic acid molecule, and the second region comprises nucleotide sequence having about 18 to about 28 nucleotides complementary to a distinct nucleic acid sequence within a second target nucleic acid molecule.


In one embodiment, the invention features a multifunctional siNA molecule comprising a first region and a second region, where the first region comprises nucleic acid sequence having about 18 to about 28 nucleotides complementary to a nucleic acid sequence within a target nucleic acid molecule, and the second region comprises nucleotide sequence having about 18 to about 28 nucleotides complementary to a distinct nucleic acid sequence within the same target nucleic acid molecule.


In one embodiment, the invention features a double stranded multifunctional short interfering nucleic acid (multifunctional siNA) molecule, wherein one strand of the multifunctional siNA comprises a first region having nucleotide sequence complementary to a first target nucleic acid sequence, and the second strand comprises a first region having a nucleotide sequence complementary to a second target nucleic acid sequence. The first and second target nucleic acid sequences can be present in separate target nucleic acid molecules or can be different regions within the same target nucleic acid molecule. As such, multifunctional siNA molecules of the invention can be used to target the expression of different genes, splice variants of the same gene, both mutant and conserved regions of one or more gene transcripts, or both coding and non-coding sequences of the same or differeing genes or gene transcripts.


In one embodiment, a target nucleic acid molecule of the invention encodes a single protein. In another embodiment, a target nucleic acid molecule encodes more than one protein (e.g., 1, 2, 3, 4, 5 or more proteins). As such, a multifunctional siNA construct of the invention can be used to down regulate or inhibit the expression of several proteins. For example, a multifunctional siNA molecule comprising a region in one strand having nucleotide sequence complementarity to a first target nucleic acid sequence derived from a viral genome (e.g., HCV) and the second strand comprising a region with nucleotide sequence complementarity to a second target nucleic acid sequence present in target nucleic acid molecules derived from genes encoding two proteins (e.g., two differing host proteins involved in the HCV life-cycle) can be used to down regulate, inhibit, or shut down a particular biologic pathway by targeting, for example, a viral RNA (e.g., HCV RNA) and one or more host RNAs that are involved in viral infection or the viral life-cycle (e.g., La antigen or interferon regulatory factors).


In another non-limiting example, a multifunctional siNA molecule comprising a region in one strand having a nucleotide sequence complementarity to a first target nucleic acid sequence derived from a target nucleic acid molecule encoding a virus or a viral protein (e.g., HIV) and the second strand comprising a region having a nucleotide sequence complementarity to a second target nucleic acid sequence present in target nucleic acid molecule encoding a cellular protein (e.g., a receptor for the virus, such as CCR5 receptor for HIV) can be used to down regulate, inhibit, or shut down the viral replication and infection by targeting the virus and cellular proteins necessary for viral infection or replication.


In another nonlimiting example, a multifunctional siNA molecule comprising a region in one strand having a nucleotide sequence complementarity to a first target nucleic acid sequence (e.g., conserved sequence) present in a target nucleic acid molecule such as a viral genome (e.g., HCV RNA) and the second strand comprising a region having a nucleotide sequence complementarity to a second target nucleic acid sequence (e.g., conserved sequence) present in target nucleic acid molecule derived from a gene encoding a viral protein (e.g., HCV proteins) to down regulate, inhibit, or shut down the viral replication and infection by targeting the viral genome and viral encoded proteins necessary for viral infection or replication.


In one embodiment the invention takes advantage of conserved nucleotide sequences present in different strains, isotypes or forms of a virus and genes encoded by these different strains, isotypes and forms of the virus (e.g., HCV). By designing multifunctional siNAs in a manner where one strand includes a sequence that is complementary to target nucleic acid sequence conserved among various strains, isotypes or forms of a virus and the other strand includes sequence that is complementary to target nucleic acid sequence conserved in a protein encoded by the virus, it is possible to selectively and effectively inhibit viral replication or infection using a single multifunctional siNA.


In one embodiment, a multifunctional short interfering nucleic acid (multifunctional siNA) of the invention comprises a first region and a second region, wherein the first region comprises nucleotide sequence complementary to a HCV viral RNA of a first viral strain and the second region comprises nucleotide sequence complementary to a HCV viral RNA of a second viral strain. In one embodiment, the first and second regions can comprise nucleotide sequence complementary to shared or conserved RNA sequences of differing viral strains or classes or viral strains.


In one embodiment, a multifunctional short interfering nucleic acid (multifunctional siNA) of the invention comprises a first region and a second region, wherein the first region comprises a nucleotide sequence complementary to a HCV viral RNA encoding one or more HCV viruses (e.g., one or more strains of HCV) and the second region comprises a nucleotide sequence complementary to a viral RNA encoding one or more interferon agonist proteins. In one embodiment, the first region can comprise a nucleotide sequence complementary to shared or conserved RNA sequences of differing HCV viral strains or classes of HCV viral strains. Non-limiting example of interferon agonist proteins include any protein that is capable of inhibition or suppressing RNA silencing (e.g., RNA binding proteins such as E3L or NS1 or equivalents thereof, see for example Li et al., 2004, PNAS, 101, 1350-1355)


In one embodiment, a multifunctional short interfering nucleic acid (multifunctional siNA) of the invention comprises a first region and a second region, wherein the first region comprises nucleotide sequence complementary to a HCV viral RNA and the second region comprises nucleotide sequence complementary to a cellular RNA that is involved in HCV viral infection and/or replication. Non-limiting examples of cellular RNAs involved in viral infection and/or replication include cellular receptors, cell surface molecules, cellular enzymes, cellular transcription factors, and/or cytokines, second messengers, and cellular accessory molecules including, but not limited to, La antigen, FAS, interferon agonsit proteins (e.g., E3L or NS1 or equivalents thereof, see for example Li et al., 2004, PNAS, 101, 1350-1355), interferon regulatory factors (IRFs); cellular PKR protein kinase (PKR); human eukaryotic initiation factors 2B (elF2B gamma and/or elF2gamma); human DEAD Box protein (DDX3); and cellular proteins that bind to the poly(U) tract of the HCV 3′-UTR, such as polypyrimidine tract-binding protein.


In one embodiment, a double stranded multifunctional siNA molecule of the invention comprises a structure having Formula MF-I:

5′-p-X Z X′-3′  3′-Y′ Z Y-p-5′


wherein each 5′-p-XZX′-3′ and 5′-p-YZY′-3′ are independently an oligonucleotide of length about 20 nucleotides to about 300 nucleotides, preferably about 20 to about 200 nucleotides, about 20 to about 100 nucleotides, about 20 to about 40 nucleotides, about 20 to about 40 nucleotides, about 24 to about 38 nucleotides, or about 26 to about 38 nucleotides; XZ comprises a nucleic acid sequence that is complementary to a first HCV target nucleic acid sequence; YZ is an oligonucleotide comprising nucleic acid sequence that is complementary to a second HCV target nucleic acid sequence; Z comprises nucleotide sequence of length about 1 to about 24 nucleotides (e.g., about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, or 24 nucleotides) that is self complementary; X comprises nucleotide sequence of length about 1 to about 100 nucleotides, preferably about 1 to about 21 nucleotides (e.g., about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, or 21 nucleotides) that is complementary to nucleotide sequence present in region Y′; Y comprises nucleotide sequence of length about 1 to about 100 nucleotides, preferably about 1 to about 21 nucleotides (e.g., about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20 or 21 nucleotides) that is complementary to nucleotide sequence present in region X′; each p comprises a terminal phosphate group that is independently present or absent; each XZ and YZ is independently of length sufficient to stably interact (i.e., base pair) with the first and second target nucleic acid sequence, respectively, or a portion thereof. For example, each sequence X and Y can independently comprise sequence from about 12 to about 21 or more nucleotides in length (e.g., about 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, or more) that is complementary to a target nucleotide sequence in different target nucleic acid molecules, such as target RNAs or a portion thereof. In another non-limiting example, the length of the nucleotide sequence of X and Z together that is complementary to the first HCV target nucleic acid sequence or a portion thereof is from about 12 to about 21 or more nucleotides (e.g., about 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, or more). In another non-limiting example, the length of the nucleotide sequence of Y and Z together, that is complementary to the second HCV target nucleic acid sequence or a portion thereof is from about 12 to about 21 or more nucleotides (e.g., about 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, or more). In one embodiment, the first HCV target nucleic acid sequence and the second HCV target nucleic acid sequence are present in the same target nucleic acid molecule (e.g., HCV RNA or host RNA). In another embodiment, the first HCV target nucleic acid sequence and the second HCV target nucleic acid sequence are present in different target nucleic acid molecules (e.g., HCV RNA and host RNA). In one embodiment, Z comprises a palindrome or a repeat sequence. In one embodiment, the lengths of oligonucleotides X and X′ are identical. In another embodiment, the lengths of oligonucleotides X and X′ are not identical. In one embodiment, the lengths of oligonucleotides Y and Y′ are identical. In another embodiment, the lengths of oligonucleotides Y and Y′ are not identical. In one embodiment, the double stranded oligonucleotide construct of Formula I(a) includes one or more, specifically 1, 2, 3 or 4, mismatches, to the extent such mismatches do not significantly diminish the ability of the double stranded oligonucleotide to inhibit target gene expression.


In one embodiment, a multifunctional siNA molecule of the invention comprises a structure having Formula MF-II:

5′-p-X X′-3′-   3′-Y′ Y-p-5′


wherein each 5′-p-XX′-3′ and 5′-p-YY′-3′ are independently an oligonucleotide of length about 20 nucleotides to about 300 nucleotides, preferably about 20 to about 200 nucleotides, about 20 to about 100 nucleotides, about 20 to about 40 nucleotides, about 20 to about 40 nucleotides, about 24 to about 38 nucleotides, or about 26 to about 38 nucleotides; X comprises a nucleic acid sequence that is complementary to a first target nucleic acid sequence; Y is an oligonucleotide comprising nucleic acid sequence that is complementary to a second target nucleic acid sequence; X comprises a nucleotide sequence of length about 1 to about 100 nucleotides, preferably about 1 to about 21 nucleotides (e.g., about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, or 21 nucleotides) that is complementary to nucleotide sequence present in region Y′; Y comprises nucleotide sequence of length about 1 to about 100 nucleotides, preferably about 1 to about 21 nucleotides (e.g., about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20 or 21 nucleotides) that is complementary to nucleotide sequence present in region X′; each p comprises a terminal phosphate group that is independently present or absent; each X and Y independently is of length sufficient to stably interact (i.e., base pair) with the first and second target nucleic acid sequence, respectively, or a portion thereof. For example, each sequence X and Y can independently comprise sequence from about 12 to about 21 or more nucleotides in length (e.g., about 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, or more) that is complementary to a target nucleotide sequence in different target nucleic acid molecules, such as HCV target RNAs or a portion thereof. In one embodiment, the first HCV target nucleic acid sequence and the second HCV target nucleic acid sequence are present in the same target nucleic acid molecule (e.g., HCV RNA or host RNA). In another embodiment, the first HCV target nucleic acid sequence and the second HCV target nucleic acid sequence are present in different target nucleic acid molecules (e.g., HCV RNA and host RNA). In one embodiment, Z comprises a palindrome or a repeat sequence. In one embodiment, the lengths of oligonucleotides X and X′ are identical. In another embodiment, the lengths of oligonucleotides X and X′ are not identical. In one embodiment, the lengths of oligonucleotides Y and Y′ are identical. In another embodiment, the lengths of oligonucleotides Y and Y′ are not identical. In one embodiment, the double stranded oligonucleotide construct of Formula I(a) includes one or more, specifically 1, 2, 3 or 4, mismatches, to the extent such mismatches do not significantly diminish the ability of the double stranded oligonucleotide to inhibit target gene expression.


In one embodiment, a multifunctional siNA molecule of the invention comprises a structure having Formula MF-III:

X   X′Y′-W-Y


wherein each X, X′, Y, and Y′ is independently an oligonucleotide of length about 15 nucleotides to about 50 nucleotides, preferably about 18 to about 40 nucleotides, or about 19 to about 23 nucleotides; X comprises nucleotide sequence that is complementary to nucleotide sequence present in region Y′; X′ comprises nucleotide sequence that is complementary to nucleotide sequence present in region Y; each X and X′ is independently of length sufficient to stably interact (i.e., base pair) with a first and a second HCV target nucleic acid sequence, respectively, or a portion thereof; W represents a nucleotide or non-nucleotide linker that connects sequences Y′ and Y; and the multifunctional siNA directs cleavage of the first and second HCV target sequence via RNA interference. In one embodiment, the first HCV target nucleic acid sequence and the second HCV target nucleic acid sequence are present in the same target nucleic acid molecule (e.g., HCV RNA or host RNA). In another embodiment, the first HCV target nucleic acid sequence and the second HCV target nucleic acid sequence are present in different target nucleic acid molecules (e.g., HCV RNA and host RNA). In one embodiment, region W connects the 3′-end of sequence Y′ with the 3′-end of sequence Y. In one embodiment, region W connects the 3′-end of sequence Y′ with the 5′-end of sequence Y. In one embodiment, region W connects the 5′-end of sequence Y′ with the 5′-end of sequence Y. In one embodiment, region W connects the 5′-end of sequence Y′ with the 3′-end of sequence Y. In one embodiment, a terminal phosphate group is present at the 5′-end of sequence X. In one embodiment, a terminal phosphate group is present at the 5′-end of sequence X′. In one embodiment, a terminal phosphate group is present at the 5′-end of sequence Y. In one embodiment, a terminal phosphate group is present at the 5′-end of sequence Y′. In one embodiment, W connects sequences Y and Y′ via a biodegradable linker. In one embodiment, W further comprises a conjugate, lable, aptamer, ligand, lipid, or polymer.


In one embodiment, a multifunctional siNA molecule of the invention comprises a structure having Formula MF-IV:

X   X′Y′-X-Y


wherein each X, X′, Y, and Y′ is independently an oligonucleotide of length about 15 nucleotides to about 50 nucleotides, preferably about 18 to about 40 nucleotides, or about 19 to about 23 nucleotides; X comprises nucleotide sequence that is complementary to nucleotide sequence present in region Y′; X′ comprises nucleotide sequence that is complementary to nucleotide sequence present in region Y; each Y and Y′ is independently of length sufficient to stably interact (i.e., base pair) with a first and a second HCV target nucleic acid sequence, respectively, or a portion thereof; W represents a nucleotide or non-nucleotide linker that connects sequences Y′ and Y; and the multifunctional siNA directs cleavage of the first and second HCV target sequence via RNA interference. In one embodiment, the first HCV target nucleic acid sequence and the second HCV target nucleic acid sequence are present in the same target nucleic acid molecule (e.g., HCV RNA or host RNA). In another embodiment, the first HCV target nucleic acid sequence and the second HCV target nucleic acid sequence are present in different target nucleic acid molecules (e.g., HCV RNA and host RNA). In one embodiment, region W connects the 3′-end of sequence Y′ with the 3′-end of sequence Y. In one embodiment, region W connects the 3′-end of sequence Y′ with the 5′-end of sequence Y. In one embodiment, region W connects the 5′-end of sequence Y′ with the 5′-end of sequence Y. In one embodiment, region W connects the 5′-end of sequence Y′ with the 3′-end of sequence Y. In one embodiment, a terminal phosphate group is present at the 5′-end of sequence X. In one embodiment, a terminal phosphate group is present at the 5′-end of sequence X′. In one embodiment, a terminal phosphate group is present at the 5′-end of sequence Y. In one embodiment, a terminal phosphate group is present at the 5′-end of sequence Y′. In one embodiment, W connects sequences Y and Y′ via a biodegradable linker. In one embodiment, W further comprises a conjugate, lable, aptamer, ligand, lipid, or polymer.


In one embodiment, a multifunctional siNA molecule of the invention comprises a structure having Formula MF-V:

X   X′Y′-W-Y


wherein each X, X′, Y, and Y′ is independently an oligonucleotide of length about 15 nucleotides to about 50 nucleotides, preferably about 18 to about 40 nucleotides, or about 19 to about 23 nucleotides; X comprises nucleotide sequence that is complementary to nucleotide sequence present in region Y′; X′ comprises nucleotide sequence that is complementary to nucleotide sequence present in region Y; each X, X′, Y, or Y′ is independently of length sufficient to stably interact (i.e., base pair) with a first, second, third, or fourth HCV target nucleic acid sequence, respectively, or a portion thereof; W represents a nucleotide or non-nucleotide linker that connects sequences Y′ and Y; and the multifunctional siNA directs cleavage of the first, second, third, and/or fourth target sequence via RNA interference. In one embodiment, the first, second, third and fourth HCV target nucleic acid sequence are all present in the same target nucleic acid molecule (e.g., HCV RNA or host RNA). In another embodiment, the first, second, third and fourth HCV target nucleic acid sequence are independently present in different target nucleic acid molecules (e.g., HCV RNA and host RNA). In one embodiment, region W connects the 3′-end of sequence Y′ with the 3′-end of sequence Y. In one embodiment, region W connects the 3′-end of sequence Y′ with the 5′-end of sequence Y. In one embodiment, region W connects the 5′-end of sequence Y′ with the 5′-end of sequence Y. In one embodiment, region W connects the 5′-end of sequence Y′ with the 3′-end of sequence Y. In one embodiment, a terminal phosphate group is present at the 5′-end of sequence X. In one embodiment, a terminal phosphate group is present at the 5′-end of sequence X′. In one embodiment, a terminal phosphate group is present at the 5′-end of sequence Y. In one embodiment, a terminal phosphate group is present at the 5′-end of sequence Y′. In one embodiment, W connects sequences Y and Y′ via a biodegradable linker. In one embodiment, W further comprises a conjugate, lable, aptamer, ligand, lipid, or polymer.


In one embodiment, regions X and Y of multifunctional siNA molecule of the invention (e.g., having any of Formula MF-1-MF-V), are complementary to different target nucleic acid sequences that are portions of the same target nucleic acid molecule. In one embodiment, such target nucleic acid sequences are at different locations within the coding region of a RNA transcript. In one embodiment, such target nucleic acid sequences comprise coding and non-coding regions of the same RNA transcript. In one embodiment, such target nucleic acid sequences comprise regions of alternately spliced transcripts or precursors of such alternately spliced transcripts.


In one embodiment, a multifunctional siNA molecule having any of Formula MF-I-MF-V can comprise chemical modifications as described herein without limitation, such as, for example, nucleotides having any of Formulae I-VII described herein, stabilization chemistries as described in Table IV, or any other combination of modified nucleotides and non-nucleotides as described in the various embodiments herein.


In one embodiment, the palidrome or repeat sequence or modified nucleotide (e.g., nucleotide with a modified base, such as 2-amino purine or a universal base) in Z of multifunctional siNA constructs having Formula MF-I or MF-II comprises chemically modified nucleotides that are able to interact with a portion of the target nucleic acid sequence (e.g., modified base analogs that can form Watson Crick base pairs or non-Watson Crick base pairs).


In one embodiment, a multifunctional siNA molecule of the invention, for example each strand of a multifunctional siNA having MF-I-MF-V, independently comprises about 15 to about 40 nucleotides (e.g., about 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, or 40 nucleotides). In one embodiment, a multifunctional siNA molecule of the invention comprises one or more chemical modifications. In a non-limiting example, the introduction of chemically modified nucleotides and/or non-nucleotides into nucleic acid molecules of the invention provides a powerful tool in overcoming potential limitations of in vivo stability and bioavailability inherent to unmodified RNA molecules that are delivered exogenously. For example, the use of chemically modified nucleic acid molecules can enable a lower dose of a particular nucleic acid molecule for a given therapeutic effect since chemically modified nucleic acid molecules tend to have a longer half-life in serum or in cells or tissues. Furthermore, certain chemical modifications can improve the bioavailability and/or potency of nucleic acid molecules by not only enhancing half-life but also facilitating the targeting of nucleic acid molecules to particular organs, cells or tissues and/or improving cellular uptake of the nucleic acid molecules. Therefore, even if the activity of a chemically modified nucleic acid molecule is reduced in vitro as compared to a native/unmodified nucleic acid molecule, for example when compared to an unmodified RNA molecule, the overall activity of the modified nucleic acid molecule can be greater than the native or unmodified nucleic acid molecule due to improved stability, potency, duration of effect, bioavailability and/or delivery of the molecule.


In another embodiment, the invention features multifunctional siNAs, wherein the multifunctional siNAs are assembled from two separate double-stranded siNAs, with one of the ends of each sense strand is tethered to the end of the sense strand of the other siNA molecule, such that the two antisense siNA strands are annealed to their corresponding sense strand that are tethered to each other at one end (see FIG. 42). The tethers or linkers can be nucleotide-based linkers or non-nucleotide based linkers as generally known in the art and as described herein.


In one embodiment, the invention features a multifunctional siNA, wherein the multifunctional siNA is assembled from two separate double-stranded siNAs, with the 5′-end of one sense strand of the siNA is tethered to the 5′-end of the sense strand of the other siNA molecule, such that the 5′-ends of the two antisense siNA strands, annealed to their corresponding sense strand that are tethered to each other at one end, point away (in the opposite direction) from each other (see FIG. 42(A)). The tethers or linkers can be nucleotide-based linkers or non-nucleotide based linkers as generally known in the art and as described herein.


In one embodiment, the invention features a multifunctional siNA, wherein the multifunctional siNA is assembled from two separate double-stranded siNAs, with the 3′-end of one sense strand of the siNA is tethered to the 3′-end of the sense strand of the other siNA molecule, such that the 5′-ends of the two antisense siNA strands, annealed to their corresponding sense strand that are tethered to each other at one end, face each other (see FIG. 42(B)). The tethers or linkers can be nucleotide-based linkers or non-nucleotide based linkers as generally known in the art and as described herein.


In one embodiment, the invention features a multifunctional siNA, wherein the multifunctional siNA is assembled from two separate double-stranded siNAs, with the 5′-end of one sense strand of the siNA is tethered to the 3′-end of the sense strand of the other siNA molecule, such that the 5′-end of the one of the antisense siNA strands annealed to their corresponding sense strand that are tethered to each other at one end, faces the 3′-end of the other antisense strand (see FIG. 42 (C-D)). The tethers or linkers can be nucleotide-based linkers or non-nucleotide based linkers as generally known in the art and as described herein.


In one embodiment, the invention features a multifunctional siNA, wherein the multifunctional siNA is assembled from two separate double-stranded siNAs, with the 5′-end of one antisense strand of the siNA is tethered to the 3′-end of the antisense strand of the other siNA molecule, such that the 5′-end of the one of the sense siNA strands annealed to their corresponding antisense sense strand that are tethered to each other at one end, faces the 3′-end of the other sense strand (see FIG. 42 (G-H)). In one embodiment, the linkage between the 5′-end of the first antisense strand and the 3′-end of the second antisense strand is designed in such a way as to be readily cleavable (e.g., biodegradable linker) such that the 5′end of each antisense strand of the multifunctional siNA has a free 5′-end suitable to mediate RNA interefence-based cleavage of the target RNA. The tethers or linkers can be nucleotide-based linkers or non-nucleotide based linkers as generally known in the art and as described herein.


In one embodiment, the invention features a multifunctional siNA, wherein the multifunctional siNA is assembled from two separate double-stranded siNAs, with the 5′-end of one antisense strand of the siNA is tethered to the 5′-end of the antisense strand of the other siNA molecule, such that the 3′-end of the one of the sense siNA strands annealed to their corresponding antisense sense strand that are tethered to each other at one end, faces the 3′-end of the other sense strand (see FIG. 42(E)). In one embodiment, the linkage between the 5′-end of the first antisense strand and the 5′-end of the second antisense strand is designed in such a way as to be readily cleavable (e.g., biodegradable linker) such that the 5′end of each antisense strand of the multifunctional siNA has a free 5′-end suitable to mediate RNA interefence-based cleavage of the target RNA. The tethers or linkers can be nucleotide-based linkers or non-nucleotide based linkers as generally known in the art and as described herein.


In one embodiment, the invention features a multifunctional siNA, wherein the multifunctional siNA is assembled from two separate double-stranded siNAs, with the 3′-end of one antisense strand of the siNA is tethered to the 3′-end of the antisense strand of the other siNA molecule, such that the 5′-end of the one of the sense siNA strands annealed to their corresponding antisense sense strand that are tethered to each other at one end, faces the 3′-end of the other sense strand (see FIG. 42(F)). In one embodiment, the linkage between the 5′-end of the first antisense strand and the 5′-end of the second antisense strand is designed in such a way as to be readily cleavable (e.g., biodegradable linker) such that the 5′end of each antisense strand of the multifunctional siNA has a free 5′-end suitable to mediate RNA interefence-based cleavage of the target RNA. The tethers or linkers can be nucleotide-based linkers or non-nucleotide based linkers as generally known in the art and as described herein.


In any of the above embodiments, a first target nucleic acid sequence or second target nucleic acid sequence can independently comprise HCV RNA or a portion thereof or a polynucleotide coding or non-coding sequence of cellular or host target that is invoved in HCV infection or replication, or disease processes associated with HCV infection such as such as cellular receptors, cell surface molecules, cellular enzymes, cellular transcription factors, and/or cytokines, second messengers, and cellular accessory molecules including, but not limited to, La antigen (see for example Costa-Mattioli et al., 2004, Mol Cell Biol., 24, 6861-70, e.g., Genbank Accession No. NM003142); FAS (e.g., Genbank Accession No. NM000043) or FAS ligand (e.g., Genbank Accession No. NM000639); interferon regulatory factors (IRFs; e.g., Genbank Accession No. AF082503.1); cellular PKR protein kinase (e.g., Genbank Accession No. XM002661.7); human eukaryotic initiation factors 2B (elF2Bgamma; e.g., Genbank Accession No. AF256223, and/or eIF2gamma; e.g., Genbank Accession No. NM006874.1); human DEAD Box protein (DDX3; e.g., Genbank Accession No. XM018021.2); and cellular proteins that bind to the poly(U) tract of the HCV 3′-UTR, such as polypyrimidine tract-binding protein (e.g., Genbank Accession Nos. NM031991.1 and XM042972.3). In one embodiment, the first HCV target nucleic acid sequence is a HCV RNA or a portion thereof and the second HCV target nucleic acid sequence is a HCV RNA of a portion thereof. In one embodiment, the first HCV target nucleic acid sequence is a HCV RNA or a portion thereof and the second HCV target nucleic acid sequence is a host RNA or a portion thereof. In one embodiment, the first HCV target nucleic acid sequence is a host RNA or a portion thereof and the second HCV target nucleic acid sequence is a host RNA or a portion thereof. In one embodiment, the first HCV target nucleic acid sequence is a host RNA or a portion thereof and the second HCV target nucleic acid sequence is a HCV RNA or a portion thereof.


Synthesis of Nucleic Acid Molecules


Synthesis of nucleic acids greater than 100 nucleotides in length is difficult using automated methods, and the therapeutic cost of such molecules is prohibitive. In this invention, small nucleic acid motifs (“small” refers to nucleic acid motifs no more than 100 nucleotides in length, preferably no more than 80 nucleotides in length, and most preferably no more than 50 nucleotides in length; e.g., individual siNA oligonucleotide sequences or siNA sequences synthesized in tandem) are preferably used for exogenous delivery. The simple structure of these molecules increases the ability of the nucleic acid to invade targeted regions of protein and/or RNA structure. Exemplary molecules of the instant invention are chemically synthesized, and others can similarly be synthesized.


Oligonucleotides (e.g., certain modified oligonucleotides or portions of oligonucleotides lacking ribonucleotides) are synthesized using protocols known in the art, for example as described in Caruthers et al., 1992, Methods in Enzymology 211, 3-19, Thompson et al., International PCT Publication No. WO 99/54459, Wincott et al., 1995, Nucleic Acids Res. 23, 2677-2684, Wincott et al., 1997, Methods Mol. Bio., 74, 59, Brennan et al., 1998, Biotechnol Bioeng., 61, 33-45, and Brennan, U.S. Pat. No. 6,001,311. All of these references are incorporated herein by reference. The synthesis of oligonucleotides makes use of common nucleic acid protecting and coupling groups, such as dimethoxytrityl at the 5′-end, and phosphoramidites at the 3′-end. In a non-limiting example, small scale syntheses are conducted on a 394 Applied Biosystems, Inc. synthesizer using a 0.2 mmol scale protocol with a 2.5 min coupling step for 2′-O-methylated nucleotides and a 45 second coupling step for 2′-deoxy nucleotides or 2′-deoxy-2′-fluoro nucleotides. Table V outlines the amounts and the contact times of the reagents used in the synthesis cycle. Alternatively, syntheses at the 0.2 μmol scale can be performed on a 96-well plate synthesizer, such as the instrument produced by Protogene (Palo Alto, Calif.) with minimal modification to the cycle. A 33-fold excess (60 μL of 0.11 M=6.6 μmol) of 2′-O-methyl phosphoramidite and a 105-fold excess of S-ethyl tetrazole (60 μL of 0.25 M=15 μmol) can be used in each coupling cycle of 2′-O-methyl residues relative to polymer-bound 5′-hydroxyl. A 22-fold excess (40 μL of 0.11 M=4.4 μmol) of deoxy phosphoramidite and a 70-fold excess of S-ethyl tetrazole (40 μL of 0.25 M=10 μmol) can be used in each coupling cycle of deoxy residues relative to polymer-bound 5′-hydroxyl. Average coupling yields on the 394 Applied Biosystems, Inc. synthesizer, determined by calorimetric quantitation of the trityl fractions, are typically 97.5-99%. Other oligonucleotide synthesis reagents for the 394 Applied Biosystems, Inc. synthesizer include the following: detritylation solution is 3% TCA in methylene chloride (ABI); capping is performed with 16% N-methyl imidazole in THF (ABI) and 10% acetic anhydride/10% 2,6-lutidine in THF (ABI); and oxidation solution is 16.9 mM 12, 49 mM pyridine, 9% water in THF (PerSeptive Biosystems, Inc.). Burdick & Jackson Synthesis Grade acetonitrile is used directly from the reagent bottle. S-Ethyltetrazole solution (0.25 M in acetonitrile) is made up from the solid obtained from American International Chemical, Inc. Alternately, for the introduction of phosphorothioate linkages, Beaucage reagent (3H-1,2-Benzodithiol-3-one 1,1-diokide, 0.05 M in acetonitrile) is used.


Deprotection of the DNA-based oligonucleotides is performed as follows: the polymer-bound trityl-on oligoribonucleotide is transferred to a 4 mL glass screw top vial and suspended in a solution of 40% aqueous methylamine (1 mL) at 65° C. for 10 minutes. After cooling to −20° C., the supernatant is removed from the polymer support. The support is washed three times with 1.0 mL of EtOH:MeCN:H2O/3:1:1, vortexed and the supernatant is then added to the first supernatant. The combined supernatants, containing the oligoribonucleotide, are dried to a white powder.


The method of synthesis used for RNA including certain siNA molecules of the invention follows the procedure as described in Usman et al., 1987, J. Am. Chem. Soc., 109, 7845; Scaringe et al., 1990, Nucleic Acids Res., 18, 5433; and Wincott et al., 1995, Nucleic Acids Res. 23, 2677-2684 Wincott et al., 1997, Methods Mol. Bio., 74, 59, and makes use of common nucleic acid protecting and coupling groups, such as dimethoxytrityl at the 5′-end, and phosphoramidites at the 3′-end. In a non-limiting example, small scale syntheses are conducted on a 394 Applied Biosystems, Inc. synthesizer using a 0.2 μmol scale protocol with a 7.5 min coupling step for alkylsilyl protected nucleotides and a 2.5 min coupling step for 2′-O-methylated nucleotides. Table V outlines the amounts and the contact times of the reagents used in the synthesis cycle. Alternatively, syntheses at the 0.2 μmol scale can be done on a 96-well plate synthesizer, such as the instrument produced by Protogene (Palo Alto, Calif.) with minimal modification to the cycle. A 33-fold excess (60 μL of 0.11 M=6.6 μmol) of 2′-O-methyl phosphoramidite and a 75-fold excess of S-ethyl tetrazole (60 μL of 0.25 M=15 μmol) can be used in each coupling cycle of 2′-O-methyl residues relative to polymer-bound 5′-hydroxyl. A 66-fold excess (120 μL of 0.11 M=13.2 μmol) of alkylsilyl (ribo) protected phosphoramidite and a 150-fold excess of S-ethyl tetrazole (120 μL of 0.25 M=30 μmol) can be used in each coupling cycle of ribo residues relative to polymer-bound 5′-hydroxyl. Average coupling yields on the 394 Applied Biosystems, Inc. synthesizer, determined by colorimetric quantitation of the trityl fractions, are typically 97.5-99%. Other oligonucleotide synthesis reagents for the 394 Applied Biosystems, Inc. synthesizer include the following: detritylation solution is 3% TCA in methylene chloride (ABI); capping is performed with 16% N-methyl imidazole in THF (ABI) and 10% acetic anhydride/10% 2,6-lutidine in THF (ABI); oxidation solution is 16.9 mM I2, 49 mM pyridine, 9% water in THF (PerSeptive Biosystems, Inc.). Burdick & Jackson Synthesis Grade acetonitrile is used directly from the reagent bottle. S-Ethyltetrazole solution (0.25 M in acetonitrile) is made up from the solid obtained from American International Chemical, Inc. Alternately, for the introduction of phosphorothioate linkages, Beaucage reagent (3H-1,2-Benzodithiol-3-one 1,1-dioxide 0.05 M in acetonitrile) is used.


Deprotection of the RNA is performed using either a two-pot or one-pot protocol. For the two-pot protocol, the polymer-bound trityl-on oligoribonucleotide is transferred to a 4 mL glass screw top vial and suspended in a solution of 40% aq. methylamine (1 mL) at 65° C. for 10 min. After cooling to −20° C., the supernatant is removed from the polymer support. The support is washed three times with 1.0 mL of EtOH:MeCN:H2O/3:1:1, vortexed and the supernatant is then added to the first supernatant. The combined supernatants, containing the oligoribonucleotide, are dried to a white powder. The base deprotected oligoribonucleotide is resuspended in anhydrous TEA/HF/NMP solution (300 μL of a solution of 1.5 mL N-methylpyrrolidinone, 750 μL TEA and 1 mL TEA-3HF to provide a 1.4 M HF concentration) and heated to 65° C. After 1.5 h, the oligomer is quenched with 1.5 M NH4HCO3.


Alternatively, for the one-pot protocol, the polymer-bound trityl-on oligoribonucleotide is transferred to a 4 mL glass screw top vial and suspended in a solution of 33% ethanolic methylamine/DMSO: 1/1 (0.8 mL) at 65° C. for 15 minutes. The vial is brought to room temperature TEA-3HF (0.1 mL) is added and the vial is heated at 65° C. for 15 minutes. The sample is cooled at −20° C. and then quenched with 1.5 M NH4HCO3.


For purification of the trityl-on oligomers, the quenched NH4HCO3 solution is loaded onto a C-18 containing cartridge that had been prewashed with acetonitrile followed by 50 mM TEAA. After washing the loaded cartridge with water, the RNA is detritylated with 0.5% TFA for 13 minutes. The cartridge is then washed again with water, salt exchanged with 1 M NaCl and washed with water again. The oligonucleotide is then eluted with 30% acetonitrile.


The average stepwise coupling yields are typically >98% (Wincott et al., 1995 Nucleic Acids Res. 23, 2677-2684). Those of ordinary skill in the art will recognize that the scale of synthesis can be adapted to be larger or smaller than the example described above including but not limited to 96-well format.


Alternatively, the nucleic acid molecules of the present invention can be synthesized separately and joined together post-synthetically, for example, by ligation (Moore et al., 1992, Science 256, 9923; Draper et al., International PCT publication No. WO 93/23569; Shabarova et al., 1991, Nucleic Acids Research 19, 4247; Bellon et al., 1997, Nucleosides & Nucleotides, 16, 951; Bellon et al., 1997, Bioconjugate Chem. 8, 204), or by hybridization following synthesis and/or deprotection.


The siNA molecules of the invention can also be synthesized via a tandem synthesis methodology as described in Example 1 herein, wherein both siNA strands are synthesized as a single contiguous oligonucleotide fragment or strand separated by a cleavable linker which is subsequently cleaved to provide separate siNA fragments or strands that hybridize and permit purification of the siNA duplex. The linker can be a polynucleotide linker or a non-nucleotide linker. The tandem synthesis of siNA as described herein can be readily adapted to both multiwell/multiplate synthesis platforms such as 96 well or similarly larger multi-well platforms. The tandem synthesis of siNA as described herein can also be readily adapted to large scale synthesis platforms employing batch reactors, synthesis columns and the like.


A siNA molecule can also be assembled from two distinct nucleic acid strands or fragments wherein one fragment includes the sense region and the second fragment includes the antisense region of the RNA molecule.


The nucleic acid molecules of the present invention can be modified extensively to enhance stability by modification with nuclease resistant groups, for example, 2′-amino, 2′-C-allyl, 2′-fluoro, 2′-O-methyl, 2′-H (for a review see Usman and Cedergren, 1992, TIBS 17, 34; Usman et al., 1994, Nucleic Acids Symp. Ser. 31, 163). siNA constructs can be purified by gel electrophoresis using general methods or can be purified by high pressure liquid chromatography (HPLC; see Wincott et al., supra, the totality of which is hereby incorporated herein by reference) and re-suspended in water.


In another aspect of the invention, siNA molecules of the invention are expressed from transcription units inserted into DNA or RNA vectors. The recombinant vectors can be DNA plasmids or viral vectors. siNA expressing viral vectors can be constructed based on, but not limited to, adeno-associated virus, retrovirus, adenovirus, or alphavirus. The recombinant vectors capable of expressing the siNA molecules can be delivered as described herein, and persist in target cells. Alternatively, viral vectors can be used that provide for transient expression of siNA molecules.


Optimizing Activity of the Nucleic Acid Molecule of the Invention.


Chemically synthesizing nucleic acid molecules with modifications (base, sugar and/or phosphate) can prevent their degradation by serum ribonucleases, which can increase their potency (see e.g., Eckstein et al., International Publication No. WO 92/07065; Perrault et al., 1990 Nature 344, 565; Pieken et al., 1991, Science 253, 314; Usman and Cedergren, 1992, Trends in Biochem. Sci. 17, 334; Usman et al., International Publication No. WO 93/15187; and Rossi et al., International Publication No. WO 91/03162; Sproat, U.S. Pat. No. 5,334,711; Gold et al., U.S. Pat. No. 6,300,074; and Burgin et al., supra; all of which are incorporated by reference herein). All of the above references describe various chemical modifications that can be made to the base, phosphate and/or sugar moieties of the nucleic acid molecules described herein. Modifications that enhance their efficacy in cells, and removal of bases from nucleic acid molecules to shorten oligonucleotide synthesis times and reduce chemical requirements are desired.


There are several examples in the art describing sugar, base and phosphate modifications that can be introduced into nucleic acid molecules with significant enhancement in their nuclease stability and efficacy. For example, oligonucleotides are modified to enhance stability and/or enhance biological activity by modification with nuclease resistant groups, for example, 2′-amino, 2′-C-allyl, 2′-fluoro, 2′-O-methyl, 2′-O-allyl, 2′-H, nucleotide base modifications (for a review see Usman and Cedergren, 1992, TIBS. 17, 34; Usman et al., 1994, Nucleic Acids Symp. Ser. 31, 163; Burgin et al., 1996, Biochemistry, 35, 14090). Sugar modification of nucleic acid molecules have been extensively described in the art (see Eckstein et al., International Publication PCT No. WO 92/07065; Perrault et al. Nature, 1990, 344, 565-568; Pieken et al. Science, 1991, 253, 314-317; Usman and Cedergren, Trends in Biochem. Sci., 1992, 17, 334-339; Usman et al. International Publication PCT No. WO 93/15187; Sproat, U.S. Pat. No. 5,334,711 and Beigelman et al., 1995, J. Biol. Chem., 270, 25702; Beigelman et al., International PCT publication No. WO 97/26270; Beigelman et al., U.S. Pat. No. 5,716,824; Usman et al., U.S. Pat. No. 5,627,053; Woolf et al., International PCT Publication No. WO 98/13526; Thompson et al., U.S. Ser. No. 60/082,404 which was filed on Apr. 20, 1998; Karpeisky et al., 1998, Tetrahedron Lett., 39, 1131; Earnshaw and Gait, 1998, Biopolymers (Nucleic Acid Sciences), 48, 39-55; Verma and Eckstein, 1998, Annu. Rev. Biochem., 67, 99-134; and Burlina et al., 1997, Bioorg. Med. Chem., 5, 1999-2010; all of the references are hereby incorporated in their totality by reference herein). Such publications describe general methods and strategies to determine the location of incorporation of sugar, base and/or phosphate modifications and the like into nucleic acid molecules without modulating catalysis, and are incorporated by reference herein. In view of such teachings, similar modifications can be used as described herein to modify the siNA nucleic acid molecules of the instant invention so long as the ability of siNA to promote RNAi is cells is not significantly inhibited.


While chemical modification of oligonucleotide internucleotide linkages with phosphorothioate, phosphorodithioate, and/or 5′-methylphosphonate linkages improves stability, excessive modifications can cause some toxicity or decreased activity. Therefore, when designing nucleic acid molecules, the amount of these internucleotide linkages should be minimized. The reduction in the concentration of these linkages should lower toxicity, resulting in increased efficacy and higher specificity of these molecules.


Short interfering nucleic acid (siNA) molecules having chemical modifications that maintain or enhance activity are provided. Such a nucleic acid is also generally more resistant to nucleases than an unmodified nucleic acid. Accordingly, the in vitro and/or in vivo activity should not be significantly lowered. In cases in which modulation is the goal, therapeutic nucleic acid molecules delivered exogenously should optimally be stable within cells until translation of the target RNA has been modulated long enough to reduce the levels of the undesirable protein. This period of time varies between hours to days depending upon the disease state. Improvements in the chemical synthesis of RNA and DNA (Wincott et al., 1995, Nucleic Acids Res. 23, 2677; Caruthers et al., 1992, Methods in Enzymology 211, 3-19 (incorporated by reference herein)) have expanded the ability to modify nucleic acid molecules by introducing nucleotide modifications to enhance their nuclease stability, as described above.


In one embodiment, nucleic acid molecules of the invention include one or more (e.g., about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or more) G-clamp nucleotides. A G-clamp nucleotide is a modified cytosine analog wherein the modifications confer the ability to hydrogen bond both Watson-Crick and Hoogsteen faces of a complementary guanine within a duplex, see for example Lin and Matteucci, 1998, J. Am. Chem. Soc., 120, 8531-8532. A single G-clamp analog substitution within an oligonucleotide can result in substantially enhanced helical thermal stability and mismatch discrimination when hybridized to complementary oligonucleotides. The inclusion of such nucleotides in nucleic acid molecules of the invention results in both enhanced affinity and specificity to nucleic acid targets, complementary sequences, or template strands. In another embodiment, nucleic acid molecules of the invention include one or more (e.g., about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or more) LNA “locked nucleic acid” nucleotides such as a 2′,4′-C methylene bicyclo nucleotide (see for example Wengel et al., International PCT Publication No. WO 00/66604 and WO 99/14226).


In another embodiment, the invention features conjugates and/or complexes of siNA molecules of the invention. Such conjugates and/or complexes can be used to facilitate delivery of siNA molecules into a biological system, such as a cell. The conjugates and complexes provided by the instant invention can impart therapeutic activity by transferring therapeutic compounds across cellular membranes, altering the pharmacokinetics, and/or modulating the localization of nucleic acid molecules of the invention. The present invention encompasses the design and synthesis of novel conjugates and complexes for the delivery of molecules, including, but not limited to, small molecules, lipids, cholesterol, phospholipids, nucleosides, nucleotides, nucleic acids, antibodies, toxins, negatively charged polymers and other polymers, for example proteins, peptides, hormones, carbohydrates, polyethylene glycols, or polyamines, across cellular membranes. In general, the transporters described are designed to be used either individually or as part of a multi-component system, with or without degradable linkers. These compounds are expected to improve delivery and/or localization of nucleic acid molecules of the invention into a number of cell types originating from different tissues, in the presence or absence of serum (see Sullenger and Cech, U.S. Pat. No. 5,854,038). Conjugates of the molecules described herein can be attached to biologically active molecules via linkers that are biodegradable, such as biodegradable nucleic acid linker molecules.


The term “biodegradable linker” as used herein, refers to a nucleic acid or non-nucleic acid linker molecule that is designed as a biodegradable linker to connect one molecule to another molecule, for example, a biologically active molecule to a siNA molecule of the invention or the sense and antisense strands of a siNA molecule of the invention. The biodegradable linker is designed such that its stability can be modulated for a particular purpose, such as delivery to a particular tissue or cell type. The stability of a nucleic acid-based biodegradable linker molecule can be modulated by using various chemistries, for example combinations of ribonucleotides, deoxyribonucleotides, and chemically-modified nucleotides, such as 2′-O-methyl, 2′-fluoro, 2′-amino, 2′-O-amino, 2′-C-allyl, 2′-O-allyl, and other 2′-modified or base modified nucleotides. The biodegradable nucleic acid linker molecule can be a dimer, trimer, tetramer or longer nucleic acid molecule, for example, an oligonucleotide of about 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 nucleotides in length, or can comprise a single nucleotide with a phosphorus-based linkage, for example, a phosphoramidate or phosphodiester linkage. The biodegradable nucleic acid linker molecule can also comprise nucleic acid backbone, nucleic acid sugar, or nucleic acid base modifications.


The term “biodegradable” as used herein, refers to degradation in a biological system, for example, enzymatic degradation or chemical degradation.


The term “biologically active molecule” as used herein refers to compounds or molecules that are capable of eliciting or modifying a biological response in a system. Non-limiting examples of biologically active siNA molecules either alone or in combination with other molecules contemplated by the instant invention include therapeutically active molecules such as antibodies, cholesterol, hormones, antivirals, peptides, proteins, chemotherapeutics, small molecules, vitamins, co-factors, nucleosides, nucleotides, oligonucleotides, enzymatic nucleic acids, antisense nucleic acids, triplex forming oligonucleotides, 2,5-A chimeras, siNA, dsRNA, allozymes, aptamers, decoys and analogs thereof. Biologically active molecules of the invention also include molecules capable of modulating the pharmacokinetics and/or pharmacodynamics of other biologically active molecules, for example, lipids and polymers such as polyamines, polyamides, polyethylene glycol and other polyethers.


The term “phospholipid” as used herein, refers to a hydrophobic molecule comprising at least one phosphorus group. For example, a phospholipid can comprise a phosphorus-containing group and saturated or unsaturated alkyl group, optionally substituted with OH, COOH, oxo, amine, or substituted or unsubstituted aryl groups.


Therapeutic nucleic acid molecules (e.g., siNA molecules) delivered exogenously optimally are stable within cells until reverse transcription of the RNA has been modulated long enough to reduce the levels of the RNA transcript. The nucleic acid molecules are resistant to nucleases in order to function as effective intracellular therapeutic agents. Improvements in the chemical synthesis of nucleic acid molecules described in the instant invention and in the art have expanded the ability to modify nucleic acid molecules by introducing nucleotide modifications to enhance their nuclease stability as described above.


In yet another embodiment, siNA molecules having chemical modifications that maintain or enhance enzymatic activity of proteins involved in RNAi are provided. Such nucleic acids are also generally more resistant to nucleases than unmodified nucleic acids. Thus, in vitro and/or in vivo the activity should not be significantly lowered.


Use of the nucleic acid-based molecules of the invention will lead to better treatments by affording the possibility of combination therapies (e.g., multiple siNA molecules targeted to different genes; nucleic acid molecules coupled with known small molecule modulators; or intermittent treatment with combinations of molecules, including different motifs and/or other chemical or biological molecules). The treatment of subjects with siNA molecules can also include combinations of different types of nucleic acid molecules, such as enzymatic nucleic acid molecules (ribozymes), allozymes, antisense, 2,5-A oligoadenylate, decoys, and aptamers.


In another aspect a siNA molecule of the invention comprises one or more 5′ and/or a 3′-cap structure, for example, on only the sense siNA strand, the antisense siNA strand, or both siNA strands.


By “cap structure” is meant chemical modifications, which have been incorporated at either terminus of the oligonucleotide (see, for example, Adamic et al., U.S. Pat. No. 5,998,203, incorporated by reference herein). These terminal modifications protect the nucleic acid molecule from exonuclease degradation, and may help in delivery and/or localization within a cell. The cap may be present at the 5′-terminus (5′-cap) or at the 3′-terminal (3′-cap) or may be present on both termini. In non-limiting examples, the 5′-cap includes, but is not limited to, glyceryl, inverted deoxy abasic residue (moiety); 4′,5′-methylene nucleotide; 1-(beta-D-erythrofuranosyl) nucleotide, 4′-thio nucleotide; carbocyclic nucleotide; 1,5-anhydrohexitol nucleotide; L-nucleotides; alpha-nucleotides; modified base nucleotide; phosphorodithioate linkage; threo-pentofuranosyl nucleotide; acyclic 3′,4′-seco nucleotide; acyclic 3,4-dihydroxybutyl nucleotide; acyclic 3,5-dihydroxypentyl nucleotide, 3′-3′-inverted nucleotide moiety; 3′-3′-inverted abasic moiety; 3′-2′-inverted nucleotide moiety; 3′-2′-inverted abasic moiety; 1,4-butanediol phosphate; 3′-phosphoramidate; hexylphosphate; aminohexyl phosphate; 3′-phosphate; 3′-phosphorothioate; phosphorodithioate; or bridging or non-bridging methylphosphonate moiety. Non-limiting examples of cap moieties are shown in FIG. 10.


Non-limiting examples of the 3′-cap include, but are not limited to, glyceryl, inverted deoxy abasic residue (moiety), 4′,5′-methylene nucleotide; 1-(beta-D-erythrofuranosyl) nucleotide; 4′-thio nucleotide, carbocyclic nucleotide; 5′-amino-alkyl phosphate; 1,3-diamino-2-propyl phosphate; 3-aminopropyl phosphate; 6-aminohexyl phosphate; 1,2-aminododecyl phosphate; hydroxypropyl phosphate; 1,5-anhydrohexitol nucleotide; L-nucleotide; alpha-nucleotide; modified base nucleotide; phosphorodithioate; threo-pentofuranosyl nucleotide; acyclic 3′,4′-seco nucleotide; 3,4-dihydroxybutyl nucleotide; 3,5-dihydroxypentyl nucleotide, 5′-5′-inverted nucleotide moiety; 5′-5′-inverted abasic moiety; 5′-phosphoramidate; 5′-phosphorothioate; 1,4-butanediol phosphate; 5′-amino; bridging and/or non-bridging 5′-phosphoramidate, phosphorothioate and/or phosphorodithioate, bridging or non bridging methylphosphonate and 5′-mercapto moieties (for more details see Beaucage and Iyer, 1993, Tetrahedron 49, 1925; incorporated by reference herein).


By the term “non-nucleotide” is meant any group or compound which can be incorporated into a nucleic acid chain in the place of one or more nucleotide units, including either sugar and/or phosphate substitutions, and allows the remaining bases to exhibit their enzymatic activity. The group or compound is abasic in that it does not contain a commonly recognized nucleotide base, such as adenosine, guanine, cytosine, uracil or thymine and therefore lacks a base at the 1′-position.


An “alkyl” group refers to a saturated aliphatic hydrocarbon, including straight-chain, branched-chain, and cyclic alkyl groups. Preferably, the alkyl group has 1 to 12 carbons. More preferably, it is a lower alkyl of from 1 to 7 carbons, more preferably 1 to 4 carbons. The alkyl group can be substituted or unsubstituted. When substituted the substituted group(s) is preferably, hydroxyl, cyano, alkoxy, ═O, ═S, NO2 or N(CH3)2, amino, or SH. The term also includes alkenyl groups that are unsaturated hydrocarbon groups containing at least one carbon-carbon double bond, including straight-chain, branched-chain, and cyclic groups. Preferably, the alkenyl group has 1 to 12 carbons. More preferably, it is a lower alkenyl of from 1 to 7 carbons, more preferably 1 to 4 carbons. The alkenyl group may be substituted or unsubstituted. When substituted the substituted group(s) is preferably, hydroxyl, cyano, alkoxy, ═O, ═S, NO2, halogen, N(CH3)2, amino, or SH. The term “alkyl” also includes alkynyl groups that have an unsaturated hydrocarbon group containing at least one carbon-carbon triple bond, including straight-chain, branched-chain, and cyclic groups. Preferably, the alkynyl group has 1 to 12 carbons. More preferably, it is a lower alkynyl of from 1 to 7 carbons, more preferably 1 to 4 carbons. The alkynyl group may be substituted or unsubstituted. When substituted the substituted group(s) is preferably, hydroxyl, cyano, alkoxy, ═O, ═S, NO2 or N(CH3)2, amino or SH.


Such alkyl groups can also include aryl, alkylaryl, carbocyclic aryl, heterocyclic aryl, amide and ester groups. An “aryl” group refers to an aromatic group that has at least one ring having a conjugated pi electron system and includes carbocyclic aryl, heterocyclic aryl and biaryl groups, all of which may be optionally substituted. The preferred substituent(s) of aryl groups are halogen, trihalomethyl, hydroxyl, SH, OH, cyano, alkoxy, alkyl, alkenyl, alkynyl, and amino groups. An “alkylaryl” group refers to an alkyl group (as described above) covalently joined to an aryl group (as described above). Carbocyclic aryl groups are groups wherein the ring atoms on the aromatic ring are all carbon atoms. The carbon atoms are optionally substituted. Heterocyclic aryl groups are groups having from 1 to 3 heteroatoms as ring atoms in the aromatic ring and the remainder of the ring atoms are carbon atoms. Suitable heteroatoms include oxygen, sulfur, and nitrogen, and include furanyl, thienyl, pyridyl, pyrrolyl, N-lower alkyl pyrrolo, pyrimidyl, pyrazinyl, imidazolyl and the like, all optionally substituted. An “amide” refers to an —C(O)—NH—R, where R is either alkyl, aryl, alkylaryl or hydrogen. An “ester” refers to an —C(O)—OR′, where R is either alkyl, aryl, alkylaryl or hydrogen.


By “nucleotide” as used herein is as recognized in the art to include natural bases (standard), and modified bases well known in the art. Such bases are generally located at the 1′ position of a nucleotide sugar moiety. Nucleotides generally comprise a base, sugar and a phosphate group. The nucleotides can be unmodified or modified at the sugar, phosphate and/or base moiety, (also referred to interchangeably as nucleotide analogs, modified nucleotides, non-natural nucleotides, non-standard nucleotides and other; see, for example, Usman and McSwiggen, supra; Eckstein et al., International PCT Publication No. WO 92/07065; Usman et al., International PCT Publication No. WO 93/15187; Uhlman & Peyman, supra, all are hereby incorporated by reference herein). There are several examples of modified nucleic acid bases known in the art as summarized by Limbach et al., 1994, Nucleic Acids Res. 22, 2183. Some of the non-limiting examples of base modifications that can be introduced into nucleic acid molecules include, inosine, purine, pyridin4-one, pyridin-2-one, phenyl, pseudouracil, 2, 4, 6-trimethoxy benzene, 3-methyl uracil, dihydrouridine, naphthyl, aminophenyl, 5-alkylcytidines (e.g., 5-methylcytidine), 5-alkyluridines (e.g., ribothymidine), 5-halouridine (e.g., 5-bromouridine) or 6-azapyrimidines or 6-alkylpyrimidines (e.g. 6-methyluridine), propyne, and others (Burgin et al., 1996, Biochemistry, 35, 14090; Uhlman & Peyman, supra). By “modified bases” in this aspect is meant nucleotide bases other than adenine, guanine, cytosine and uracil at 1′ position or their equivalents.


In one embodiment, the invention features modified siNA molecules, with phosphate backbone modifications comprising one or more phosphorothioate, phosphorodithioate, methylphosphonate, phosphotriester, morpholino, amidate carbamate, carboxymethyl, acetamidate, polyamide, sulfonate, sulfonamide, sulfamate, formacetal, thioformacetal, and/or alkylsilyl, substitutions. For a review of oligonucleotide backbone modifications, see Hunziker and Leumann, 1995, Nucleic Acid Analogues: Synthesis and Properties, in Modern Synthetic Methods, VCH, 331417, and Mesmaeker et al., 1994, Novel Backbone Replacements for Oligonucleotides, in Carbohydrate Modifications in Antisense Research, ACS, 24-39.


By “abasic” is meant sugar moieties lacking a base or having other chemical groups in place of a base at the 1′ position, see for example Adamic et al., U.S. Pat. No. 5,998,203.


By “unmodified nucleoside” is meant one of the bases adenine, cytosine, guanine, thymine, or uracil joined to the 1′ carbon of β-D-ribo-furanose.


By “modified nucleoside” is meant any nucleotide base which contains a modification in the chemical structure of an unmodified nucleotide base, sugar and/or phosphate. Non-limiting examples of modified nucleotides are shown by Formulae I-VII and/or other modifications described herein.


In connection with 2′-modified nucleotides as described for the present invention, by “amino” is meant 2′-NH2 or 2′-O—NH2, which can be modified or unmodified. Such modified groups are described, for example, in Eckstein et al., U.S. Pat. No. 5,672,695 and Matulic-Adamic et al., U.S. Pat. No. 6,248,878, which are both incorporated by reference in their entireties.


Various modifications to nucleic acid siNA structure can be made to enhance the utility of these molecules. Such modifications will enhance shelf-life, half-life in vitro, stability, and ease of introduction of such oligonucleotides to the target site, e.g., to enhance penetration of cellular membranes, and confer the ability to recognize and bind to targeted cells.


Administration of Nucleic Acid Molecules


A siNA molecule of the invention can be adapted for use to treat, prevent, inhibit, or reduce HCV infection, liver failure, hepatocellular carcinoma, cirrhosis and/or any other trait, disease or condition that is related to or will respond to the levels of HCV in a cell or tissue, alone or in combination with other therapies. In one embodiment, the siNA molecules of the invention and formulations or compositions thereof are administered to the liver as is generally known in the art (see for example Wen et al., 2004, World J Gastroenterol., 10, 244-9; Murao et al., 2002, Pharm Res., 19, 1808-14; Liu et al., 2003, Gene Ther., 10, 180-7; Hong et al., 2003, J Pharm Pharmacol., 54, 51-8; Herrmann et al., 2004, Arch Virol., 149, 1611-7; and Matsuno et al., 2003, Gene Ther., 10, 1559-66).


For example, a siNA molecule can comprise a delivery vehicle, including liposomes, for administration to a subject, carriers and diluents and their salts, and/or can be present in pharmaceutically acceptable formulations. Methods for the delivery of nucleic acid molecules are described in Akhtar et al., 1992, Trends Cell Bio., 2, 139; Delivery Strategies for Antisense Oligonucleotide Therapeutics, ed. Akhtar, 1995, Maurer et al., 1999, Mol. Membr. Biol., 16, 129-140; Hofland and Huang, 1999, Handb. Exp. Pharmacol., 137, 165-192; and Lee et al., 2000, ACS Symp. Ser., 752, 184-192, all of which are incorporated herein by reference. Beigelman et al., U.S. Pat. No. 6,395,713 and Sullivan et al., PCT WO 94/02595 further describe the general methods for delivery of nucleic acid molecules. These protocols can be utilized for the delivery of virtually any nucleic acid molecule. Nucleic acid molecules can be administered to cells by a variety of methods known to those of skill in the art, including, but not restricted to, encapsulation in liposomes, by iontophoresis, or by incorporation into other vehicles, such as biodegradable polymers, hydrogels, cyclodextrins (see for example Gonzalez et al., 1999, Bioconjugate Chem., 10, 1068-1074; Wang et al., International PCT publication Nos. WO 03/47518 and WO 03/46185), poly(lactic-co-glycolic)acid (PLGA) and PLCA microspheres (see for example U.S. Pat. No. 6,447,796 and US Patent Application Publication No. U.S. 2002130430), biodegradable nanocapsules, and bioadhesive microspheres, or by proteinaceous vectors (O'Hare and Normand, International PCT Publication No. WO 00/53722). In another embodiment, the nucleic acid molecules of the invention can also be formulated or complexed with polyethyleneimine and derivatives thereof, such as polyethyleneimine-polyethyleneglycol-N-acetylgalactosamine (PEI-PEG-GAL) or polyethyleneimine-polyethyleneglycol-tri-N-acetylgalactosamine (PEI-PEG-triGAL) derivatives. In one embodiment, the nucleic acid molecules of the invention are formulated as described in U.S. Patent Application Publication No. 20030077829, incorporated by reference herein in its entirety.


In one embodiment, a siNA molecule of the invention is complexed with membrane disruptive agents such as those described in U.S. Patent Application Publication No. 20010007666, incorporated by reference herein in its entirety including the drawings. In another embodiment, the membrane disruptive agent or agents and the siNA molecule are also complexed with a cationic lipid or helper lipid molecule, such as those lipids described in U.S. Pat. No. 6,235,310, incorporated by reference herein in its entirety including the drawings.


In one embodiment, a siNA molecule of the invention is complexed with delivery systems as described in U.S. Patent Application Publication No. 2003077829 and International PCT Publication Nos. WO 00/03683 and WO 02/087541, all incorporated by reference herein in their entirety including the drawings.


In one embodiment, the siNA molecules of the invention and formulations or compositions thereof are administered directly or topically (e.g., locally) to the dermis or follicles as is generally known in the art (see for example Brand, 2001, Curr. Opin. Mol. Ther., 3, 244-8; Regnier et al., 1998, J Drug Target, 5, 275-89; Kanikkannan, 2002, BioDrugs, 16, 33947; Wraight et al., 2001, Pharmacol. Ther., 90, 89-104; Preat and Dujardin, 2001, STP PharmaSciences, 11, 57-68; and Vogt et al., 2003, Hautarzt. 54, 692-8).


In one embodiment, delivery systems of the invention include, for example, aqueous and nonaqueous gels, creams, multiple emulsions, microemulsions, liposomes, ointments, aqueous and nonaqueous solutions, lotions, aerosols, hydrocarbon bases and powders, and can contain excipients such as solubilizers, permeation enhancers (e.g., fatty acids, fatty acid esters, fatty alcohols and amino acids), and hydrophilic polymers (e.g., polycarbophil and polyvinylpyrolidone). In one embodiment, the pharmaceutically acceptable carrier is a liposome or a transdermal enhancer. Examples of liposomes which can be used in this invention include the following: (1) CellFectin, 1:1.5 (M/M) liposome formulation of the cationic lipid N,NI,NII,NIII-tetramethyl-N,NI,NII,NIII-tetrapalmit-y-spermine and dioleoyl phosphatidylethanolamine (DOPE) (GIBCO BRL); (2) Cytofectin GSV, 2:1 (M/M) liposome formulation of a cationic lipid and DOPE (Glen Research); (3) DOTAP (N-[1-(2,3-dioleoyloxy)-N,N,N-tri-methyl-ammoniummethylsulfate) (Boehringer Manheim); and (4) Lipofectamine, 3:1 (M/M) liposome formulation of the polycationic lipid DOSPA and the neutral lipid DOPE (GIBCO BRL).


In one embodiment, delivery systems of the invention include patches, tablets, suppositories, pessaries, gels and creams, and can contain excipients such as solubilizers and enhancers (e.g., propylene glycol, bile salts and amino acids), and other vehicles (e.g., polyethylene glycol, fatty acid esters and derivatives, and hydrophilic polymers such as hydroxypropylmethylcellulose and hyaluronic acid).


In one embodiment, transdermal delivery systems of the invention include patches, tablets, suppositories, pessaries, gels and creams, and can contain excipients such as solubilizers and enhancers (e.g., propylene glycol, bile salts and amino acids), and other vehicles (e.g., polyethylene glycol, fatty acid esters and derivatives, and hydrophilic polymers such as hydroxypropylmethylcellulose and hyaluronic acid).


In one embodiment, siNA molecules of the invention are formulated or complexed with polyethylenimine (e.g., linear or branched PEI) and/or polyethylenimine derivatives, including for example grafted PEIs such as galactose PEI, cholesterol PEI, antibody derivatized PEI, and polyethylene glycol PEI (PEG-PEI) derivatives thereof (see for example Ogris et al., 2001, AAPA PhannSci, 3, 1-11; Furgeson et al., 2003, Bioconjugate Chem., 14, 840-847; Kunath et al., 2002, Phramaceutical Research, 19, 810-817; Choi et al., 2001, Bull. Korean Chem. Soc., 22, 46-52; Bettinger et al., 1999, Bioconjugate Chem., 10, 558-561; Peterson et al., 2002, Bioconjugate Chem., 13, 845-854; Erbacher et al., 1999, Journal of Gene Medicine Preprint, 1, 1-18; Godbey et al., 1999., PNAS USA, 96, 5177-5181; Godbey et al., 1999, Journal of Controlled Release, 60, 149-160; Diebold et al., 1999, Journal of Biological Chemistry, 274, 19087-19094; Thomas and Klibanov, 2002, PNAS USA, 99, 14640-14645; and Sagara, U.S. Pat. No. 6,586,524, incorporated by reference herein.


In one embodiment, a siNA molecule of the invention comprises a bioconjugate, for example a nucleic acid conjugate as described in Vargeese et al., U.S. Ser. No. 10/427,160, filed Apr. 30, 2003; U.S. Pat. No. 6,528,631; U.S. Pat. No. 6,335,434; U.S. Pat. No. 6,235,886; U.S. Pat. No. 6,153,737; U.S. Pat. No. 5,214,136; U.S. Pat. No. 5,138,045, all incorporated by reference herein.


Thus, the invention features a pharmaceutical composition comprising one or more nucleic acid(s) of the invention in an acceptable carrier, such as a stabilizer, buffer, and the like. The polynucleotides of the invention can be administered (e.g., RNA, DNA or protein) and introduced to a subject by any standard means, with or without stabilizers, buffers, and the like, to form a pharmaceutical composition. When it is desired to use a liposome delivery mechanism, standard protocols for formation of liposomes can be followed. The compositions of the present invention can also be formulated and used as creams, gels, sprays, oils and other suitable compositions for topical, dermal, or transdermal administration as is known in the art.


The present invention also includes pharmaceutically acceptable formulations of the compounds described. These formulations include salts of the above compounds, e.g., acid addition salts, for example, salts of hydrochloric, hydrobromic, acetic acid, and benzene sulfonic acid.


A pharmacological composition or formulation refers to a composition or formulation in a form suitable for administration, e.g., systemic or local administration, into a cell or subject, including for example a human. Suitable forms, in part, depend upon the use or the route of entry, for example oral, transdermal, or by injection. Such forms should not prevent the composition or formulation from reaching a target cell (i.e., a cell to which the negatively charged nucleic acid is desirable for delivery). For example, pharmacological compositions injected into the blood stream should be soluble. Other factors are known in the art, and include considerations such as toxicity and forms that prevent the composition or formulation from exerting its effect.


In one embodiment, siNA molecules of the invention are administered to a subject by systemic administration in a pharmaceutically acceptable composition or formulation. By “systemic administration” is meant in vivo systemic absorption or accumulation of drugs in the blood stream followed by distribution throughout the entire body. Administration routes that lead to systemic absorption include, without limitation: intravenous, subcutaneous, intraperitoneal, inhalation, oral, intrapulmonary and intramuscular. Each of these administration routes exposes the siNA molecules of the invention to an accessible diseased tissue. The rate of entry of a drug into the circulation has been shown to be a function of molecular weight or size. The use of a liposome or other drug carrier comprising the compounds of the instant invention can potentially localize the drug, for example, in certain tissue types, such as the tissues of the reticular endothelial system (RES). A liposome formulation that can facilitate the association of drug with the surface of cells, such as, lymphocytes and macrophages is also useful. This approach can provide enhanced delivery of the drug to target cells by taking advantage of the specificity of macrophage and lymphocyte immune recognition of abnormal cells.


By “pharmaceutically acceptable formulation” or “pharmaceutically acceptable composition” is meant, a composition or formulation that allows for the effective distribution of the nucleic acid molecules of the instant invention in the physical location most suitable for their desired activity. Non-limiting examples of agents suitable for formulation with the nucleic acid molecules of the instant invention include: P-glycoprotein inhibitors (such as Pluronic P85),; biodegradable polymers, such as poly (DL-lactide-coglycolide) microspheres for sustained release delivery (Emerich, DF et al, 1999, Cell Transplant, 8, 47-58); and loaded nanoparticles, such as those made of polybutylcyanoacrylate. Other non-limiting examples of delivery strategies for the nucleic acid molecules of the instant invention include material described in Boado et al., 1998, J. Pharm. Sci., 87, 1308-1315; Tyler et al., 1999, FEBS Lett., 421, 280-284; Pardridge et al., 1995, PNAS USA., 92, 5592-5596; Boado, 1995, Adv. Drug Delivery Rev., 15, 73-107; Aldrian-Herrada et al., 1998, Nucleic Acids Res., 26, 4910-4916; and Tyler et al., 1999, PNAS USA., 96, 7053-7058.


The invention also features the use of a composition comprising surface-modified liposomes containing poly (ethylene glycol) lipids (PEG-modified, or long-circulating liposomes or stealth liposomes) and nucleic acid molecules of the invention. These formulations offer a method for increasing the accumulation of drugs (e.g., siNA) in target tissues. This class of drug carriers resists opsonization and elimination by the mononuclear phagocytic system (MPS or RES), thereby enabling longer blood circulation times and enhanced tissue exposure for the encapsulated drug (Lasic et al. Chem. Rev. 1995, 95, 2601-2627; Ishiwata et al., Chem. Pharm. BulL 1995, 43, 1005-1011). Such liposomes have been shown to accumulate selectively in tumors, presumably by extravasation and capture in the neovascularized target tissues (Lasic et al., Science 1995, 267, 1275-1276; Oku et al., 1995, Biochim. Biophys. Acta, 1238, 86-90). The long-circulating liposomes enhance the pharmacokinetics and pharmacodynamics of DNA and RNA, particularly compared to conventional cationic liposomes which are known to accumulate in tissues of the MPS (Liu et al., J. Biol. Chem. 1995, 42, 24864-24870; Choi et al., International PCT Publication No. WO 96/10391; Ansell et al., International PCT Publication No. WO 96/10390; Holland et al., International PCT Publication No. WO 96/10392). Long-circulating liposomes are also likely to protect drugs from nuclease degradation to a greater extent compared to cationic liposomes, based on their ability to avoid accumulation in metabolically aggressive MPS tissues such as the liver and spleen.


The present invention also includes compositions prepared for storage or administration that include a pharmaceutically effective amount of the desired compounds in a pharmaceutically acceptable carrier or diluent. Acceptable carriers or diluents for therapeutic use are well known in the pharmaceutical art, and are described, for example, in Remington's Pharmaceutical Sciences, Mack Publishing Co. (A. R. Gennaro edit. 1985), hereby incorporated by reference herein. For example, preservatives, stabilizers, dyes and flavoring agents can be provided. These include sodium benzoate, sorbic acid and esters of p-hydroxybenzoic acid. In addition, antioxidants and suspending agents can be used.


A pharmaceutically effective dose is that dose required to prevent, inhibit the occurrence, or treat (alleviate a symptom to some extent, preferably all of the symptoms) of a disease state. The pharmaceutically effective dose depends on the type of disease, the composition used, the route of administration, the type of mammal being treated, the physical characteristics of the specific mammal under consideration, concurrent medication, and other factors that those skilled in the medical arts will recognize. Generally, an amount between 0.1 mg/kg and 100 mg/kg body weight/day of active ingredients is administered dependent upon potency of the negatively charged polymer.


The nucleic acid molecules of the invention and formulations thereof can be administered orally, topically, parenterally, by inhalation or spray, or rectally in dosage unit formulations containing conventional non-toxic pharmaceutically acceptable carriers, adjuvants and/or vehicles. The term parenteral as used herein includes percutaneous, subcutaneous, intravascular (e.g., intravenous), intramuscular, or intrathecal injection or infusion techniques and the like. In addition, there is provided a pharmaceutical formulation comprising a nucleic acid molecule of the invention and a pharmaceutically acceptable carrier. One or more nucleic acid molecules of the invention can be present in association with one or more non-toxic pharmaceutically acceptable carriers and/or diluents and/or adjuvants, and if desired other active ingredients. The pharmaceutical compositions containing nucleic acid molecules of the invention can be in a form suitable for oral use, for example, as tablets, troches, lozenges, aqueous or oily suspensions, dispersible powders or granules, emulsion, hard or soft capsules, or syrups or elixirs.


Compositions intended for oral use can be prepared according to any method known to the art for the manufacture of pharmaceutical compositions and such compositions can contain one or more such sweetening agents, flavoring agents, coloring agents or preservative agents in order to provide pharmaceutically elegant and palatable preparations. Tablets contain the active ingredient in admixture with non-toxic pharmaceutically acceptable excipients that are suitable for the manufacture of tablets. These excipients can be, for example, inert diluents; such as calcium carbonate, sodium carbonate, lactose, calcium phosphate or sodium phosphate; granulating and disintegrating agents, for example, corn starch, or alginic acid; binding agents, for example starch, gelatin or acacia; and lubricating agents, for example magnesium stearate, stearic acid or talc. The tablets can be uncoated or they can be coated by known techniques. In some cases such coatings can be prepared by known techniques to delay disintegration and absorption in the gastrointestinal tract and thereby provide a sustained action over a longer period. For example, a time delay material such as glyceryl monosterate or glyceryl distearate can be employed.


Formulations for oral use can also be presented as hard gelatin capsules wherein the active ingredient is mixed with an inert solid diluent, for example, calcium carbonate, calcium phosphate or kaolin, or as soft gelatin capsules wherein the active ingredient is mixed with water or an oil medium, for example peanut oil, liquid paraffin or olive oil.


Aqueous suspensions contain the active materials in a mixture with excipients suitable for the manufacture of aqueous suspensions. Such excipients are suspending agents, for example sodium carboxymethylcellulose, methylcellulose, hydropropyl-methylcellulose, sodium alginate, polyvinylpyrrolidone, gum tragacanth and gum acacia; dispersing or wetting agents can be a naturally-occurring phosphatide, for example, lecithin, or condensation products of an alkylene oxide with fatty acids, for example polyoxyethylene stearate, or condensation products of ethylene oxide with long chain aliphatic alcohols, for example heptadecaethyleneoxycetanol, or condensation products of ethylene oxide with partial esters derived from fatty acids and a hexitol such as polyoxyethylene sorbitol monooleate, or condensation products of ethylene oxide with partial esters derived from fatty acids and hexitol anhydrides, for example polyethylene sorbitan monooleate. The aqueous suspensions can also contain one or more preservatives, for example ethyl, or n-propyl p-hydroxybenzoate, one or more coloring agents, one or more flavoring agents, and one or more sweetening agents, such as sucrose or saccharin.


Oily suspensions can be formulated by suspending the active ingredients in a vegetable oil, for example arachis oil, olive oil, sesame oil or coconut oil, or in a mineral oil such as liquid paraffin. The oily suspensions can contain a thickening agent, for example beeswax, hard paraffin or cetyl alcohol. Sweetening agents and flavoring agents can be added to provide palatable oral preparations. These compositions can be preserved by the addition of an anti-oxidant such as ascorbic acid.


Dispersible powders and granules suitable for preparation of an aqueous suspension by the addition of water provide the active ingredient in admixture with a dispersing or wetting agent, suspending agent and one or more preservatives. Suitable dispersing or wetting agents or suspending agents are exemplified by those already mentioned above. Additional excipients, for example sweetening, flavoring and coloring agents, can also be present.


Pharmaceutical compositions of the invention can also be in the form of oil-in-water emulsions. The oily phase can be a vegetable oil or a mineral oil or mixtures of these. Suitable emulsifying agents can be naturally-occurring gums, for example gum acacia or gum tragacanth, naturally-occurring phosphatides, for example soy bean, lecithin, and esters or partial esters derived from fatty acids and hexitol, anhydrides, for example sorbitan monooleate, and condensation products of the said partial esters with ethylene oxide, for example polyoxyethylene sorbitan monooleate. The emulsions can also contain sweetening and flavoring agents.


Syrups and elixirs can be formulated with sweetening agents, for example glycerol, propylene glycol, sorbitol, glucose or sucrose. Such formulations can also contain a demulcent, a preservative and flavoring and coloring agents. The pharmaceutical compositions can be in the form of a sterile injectable aqueous or oleaginous suspension. This suspension can be formulated according to the known art using those suitable dispersing or wetting agents and suspending agents that have been mentioned above. The sterile injectable preparation can also be a sterile injectable solution or suspension in a non-toxic parentally acceptable diluent or solvent, for example as a solution in 1,3-butanediol. Among the acceptable vehicles and solvents that can be employed are water, Ringer's solution and isotonic sodium chloride solution. In addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium. For this purpose, any bland fixed oil can be employed including synthetic mono- or diglycerides. In addition, fatty acids such as oleic acid find use in the preparation of injectables.


The nucleic acid molecules of the invention can also be administered in the form of suppositories, e.g., for rectal administration of the drug. These compositions can be prepared by mixing the drug with a suitable non-irritating excipient that is solid at ordinary temperatures but liquid at the rectal temperature and will therefore melt in the rectum to release the drug. Such materials include cocoa butter and polyethylene glycols.


Nucleic acid molecules of the invention can be administered parenterally in a sterile medium. The drug, depending on the vehicle and concentration used, can either be suspended or dissolved in the vehicle. Advantageously, adjuvants such as local anesthetics, preservatives and buffering agents can be dissolved in the vehicle.


Dosage levels of the order of from about 0.1 mg to about 140 mg per kilogram of body weight per day are useful in the treatment of the above-indicated conditions (about 0.5 mg to about 7 g per subject per day). The amount of active ingredient that can be combined with the carrier materials to produce a single dosage form varies depending upon the host treated and the particular mode of administration. Dosage unit forms generally contain between from about 1 mg to about 500 mg of an active ingredient.


It is understood that the specific dose level for any particular subject depends upon a variety of factors including the activity of the specific compound employed, the age, body weight, general health, sex, diet, time of administration, route of administration, and rate of excretion, drug combination and the severity of the particular disease undergoing therapy.


For administration to non-human animals, the composition can also be added to the animal feed or drinking water. It can be convenient to formulate the animal feed and drinking water compositions so that the animal takes in a therapeutically appropriate quantity of the composition along with its diet. It can also be convenient to present the composition as a premix for addition to the feed or drinking water.


The nucleic acid molecules of the present invention can also be administered to a subject in combination with other therapeutic compounds to increase the overall therapeutic effect. The use of multiple compounds to treat an indication can increase the beneficial effects while reducing the presence of side effects.


In one embodiment, the invention comprises compositions suitable for administering nucleic acid molecules of the invention to specific cell types. For example, the asialoglycoprotein receptor (ASGPr) (Wu and Wu, 1987, J. Biol. Chem. 262, 4429-4432) is unique to hepatocytes and binds branched galactose-terminal glycoproteins, such as asialoorosomucoid (ASOR). In another example, the folate receptor is overexpressed in many cancer cells. Binding of such glycoproteins, synthetic glycoconjugates, or folates to the receptor takes place with an affinity that strongly depends on the degree of branching of the oligosaccharide chain, for example, triatennary structures are bound with greater affinity than biatenarry or monoatennary chains (Baenziger and Fiete, 1980, Cell, 22, 611-620; Connolly et al., 1982, J. Biol. Chem., 257, 939-945). Lee and Lee, 1987, Glycoconjugate J., 4, 317-328, obtained this high specificity through the use of N-acetyl-D-galactosamine as the carbohydrate moiety, which has higher affinity for the receptor, compared to galactose. This “clustering effect” has also been described for the binding and uptake of mannosyl-terminating glycoproteins or glycoconjugates (Ponpipom et al., 1981, J. Med. Chem., 24, 1388-1395). The use of galactose, galactosamine, or folate based conjugates to transport exogenous compounds across cell membranes can provide a targeted delivery approach to, for example, the treatment of liver disease, cancers of the liver, or other cancers. The use of bioconjugates can also provide a reduction in the required dose of therapeutic compounds required for treatment. Furthermore, therapeutic bioavailability, pharmacodynamics, and pharmacokinetic parameters can be modulated through the use of nucleic acid bioconjugates of the invention. Non-limiting examples of such bioconjugates are described in Vargeese et al., U.S. Ser. No. 10/201,394, filed Aug. 13, 2001; and Matulic-Adamic et al., U.S. Ser. No. 60/362,016, filed Mar. 6, 2002.


Alternatively, certain siNA molecules of the instant invention can be expressed within cells from eukaryotic promoters (e.g., Izant and Weintraub, 1985, Science, 229, 345; McGarry and Lindquist, 1986, Proc. Natl. Acad. Sci., USA 83, 399; Scanlon et al., 1991, Proc. Natl. Acad. Sci. USA, 88, 10591-5; Kashani-Sabet et al., 1992, Antisense Res. Dev., 2, 3-15; Dropulic et al., 1992, J. Virol., 66, 1432-41; Weerasinghe et al., 1991, J. Virol., 65, 5531-4; Ojwang et al., 1992, Proc. Natl. Acad. Sci. USA, 89, 10802-6; Chen et al., 1992, Nucleic Acids Res., 20, 4581-9; Sarver et al., 1990 Science, 247, 1222-1225; Thompson et al., 1995, Nucleic Acids Res., 23, 2259; Good et al., 1997, Gene Therapy, 4, 45. Those skilled in the art realize that any nucleic acid can be expressed in eukaryotic cells from the appropriate DNA/RNA vector. The activity of such nucleic acids can be augmented by their release from the primary transcript by a enzymatic nucleic acid (Draper et al., PCT WO 93/23569, and Sullivan et al., PCT WO 94/02595; Ohkawa et al., 1992, Nucleic Acids Symp. Ser., 27, 15-6; Taira et al., 1991, Nucleic Acids Res., 19, 5125-30; Ventura et al., 1993, Nucleic Acids Res., 21, 3249-55; Chowrira et al., 1994, J. Biol. Chem., 269, 25856.


In another aspect of the invention, RNA molecules of the present invention can be expressed from transcription units (see for example Couture et al., 1996, TIG., 12, 510) inserted into DNA or RNA vectors. The recombinant vectors can be DNA plasmids or viral vectors. siNA expressing viral vectors can be constructed based on, but not limited to, adeno-associated virus, retrovirus, adenovirus, or alphavirus. In another embodiment, pol III based constructs are used to express nucleic acid molecules of the invention (see for example Thompson, U.S. Pats. Nos. 5,902,880 and 6,146,886). The recombinant vectors capable of expressing the siNA molecules can be delivered as described above, and persist in target cells. Alternatively, viral vectors can be used that provide for transient expression of nucleic acid molecules. Such vectors can be repeatedly administered as necessary. Once expressed, the siNA molecule interacts with the target mRNA and generates an RNAi response. Delivery of siNA molecule expressing vectors can be systemic, such as by intravenous or intra-muscular administration, by administration to target cells ex-planted from a subject followed by reintroduction into the subject, or by any other means that would allow for introduction into the desired target cell (for a review see Couture et al., 1996, TIG., 12, 510).


In one aspect the invention features an expression vector comprising a nucleic acid sequence encoding at least one siNA molecule of the instant invention. The expression vector can encode one or both strands of a siNA duplex, or a single self-complementary strand that self hybridizes into a siNA duplex. The nucleic acid sequences encoding the siNA molecules of the instant invention can be operably linked in a manner that allows expression of the siNA molecule (see for example Paul et al., 2002, Nature Biotechnology, 19, 505; Miyagishi and Taira, 2002, Nature Biotechnology, 19, 497; Lee et al., 2002, Nature Biotechnology, 19, 500; and Novina et al., 2002, Nature Medicine, advance online publication doi: 10.1038/nm725).


In another aspect, the invention features an expression vector comprising: a) a transcription initiation region (e.g., eukaryotic pol I, II or III initiation region); b) a transcription termination region (e.g., eukaryotic pol I, II or III termination region); and c) a nucleic acid sequence encoding at least one of the siNA molecules of the instant invention, wherein said sequence is operably linked to said initiation region and said termination region in a manner that allows expression and/or delivery of the siNA molecule. The vector can optionally include an open reading frame (ORF) for a protein operably linked on the 5′ side or the 3′-side of the sequence encoding the siNA of the invention; and/or an intron (intervening sequences).


Transcription of the siNA molecule sequences can be driven from a promoter for eukaryotic RNA polymerase I (pol I), RNA polymerase II (pol II), or RNA polymerase III (pol 111). Transcripts from pol II or pol III promoters are expressed at high levels in all cells; the levels of a given pol II promoter in a given cell type depends on the nature of the gene regulatory sequences (enhancers, silencers, etc.) present nearby. Prokaryotic RNA polymerase promoters are also used, providing that the prokaryotic RNA polymerase enzyme is expressed in the appropriate cells (Elroy-Stein and Moss, 1990, Proc. Natl. Acad. Sci. USA, 87, 6743-7; Gao and Huang 1993, Nucleic Acids Res., 21, 2867-72; Lieber et al., 1993, Methods Enzymol., 217, 47-66; Zhou et al., 1990, Mol. Cell. Biol., 10, 4529-37). Several investigators have demonstrated that nucleic acid molecules expressed from such promoters can function in mammalian cells (e.g. Kashani-Sabet et al., 1992, Antisense Res. Dev., 2, 3-15; Ojwang et al., 1992, Proc. Natl. Acad. Sci. USA, 89, 10802-6; Chen et al., 1992, Nucleic Acids Res., 20, 4581-9; Yu et al., 1993, Proc. Natl. Acad. Sci. USA, 90, 6340-4; L'Huillier et al., 1992, EMBO J., 11, 4411-8; Lisziewicz et al., 1993, Proc. Natl. Acad. Sci. U S. A, 90, 80004; Thompson et al., 1995, Nucleic Acids Res., 23, 2259; Sullenger & Cech, 1993, Science, 262, 1566). More specifically, transcription units such as the ones derived from genes encoding U6 small nuclear (snRNA), transfer RNA (tRNA) and adenovirus VA RNA are useful in generating high concentrations of desired RNA molecules such as siNA in cells (Thompson et al., supra; Couture and Stinchcomb, 1996, supra; Noonberg et al., 1994, Nucleic Acid Res., 22, 2830; Noonberg et al., U.S. Pat. No. 5,624,803; Good et al., 1997, Gene Ther., 4, 45; Beigelman et al., International PCT Publication No. WO 96/18736. The above siNA transcription units can be incorporated into a variety of vectors for introduction into mammalian cells, including but not restricted to, plasmid DNA vectors, viral DNA vectors (such as adenovirus or adeno-associated virus vectors), or viral RNA vectors (such as retroviral or alphavirus vectors) (for a review see Couture and Stinchcomb, 1996, supra).


In another aspect the invention features an expression vector comprising a nucleic acid sequence encoding at least one of the siNA molecules of the invention in a manner that allows expression of that siNA molecule. The expression vector comprises in one embodiment; a) a transcription initiation region; b) a transcription termination region; and c) a nucleic acid sequence encoding at least one strand of the siNA molecule, wherein the sequence is operably linked to the initiation region and the termination region in a manner that allows expression and/or delivery of the siNA molecule.


In another embodiment the expression vector comprises: a) a transcription initiation region; b) a transcription termination region; c) an open reading frame; and d) a nucleic acid sequence encoding at least one strand of a siNA molecule, wherein the sequence is operably linked to the 3′-end of the open reading frame and wherein the sequence is operably linked to the initiation region, the open reading frame and the termination region in a manner that allows expression and/or delivery of the siNA molecule. In yet another embodiment, the expression vector comprises: a) a transcription initiation region; b) a transcription termination region; c) an intron; and d) a nucleic acid sequence encoding at least one siNA molecule, wherein the sequence is operably linked to the initiation region, the intron and the termination region in a manner which allows expression and/or delivery of the nucleic acid molecule.


In another embodiment, the expression vector comprises: a) a transcription initiation region; b) a transcription termination region; c) an intron; d) an open reading frame; and e) a nucleic acid sequence encoding at least one strand of a siNA molecule, wherein the sequence is operably linked to the 3′-end of the open reading frame and wherein the sequence is operably linked to the initiation region, the intron, the open reading frame and the termination region in a manner which allows expression and/or delivery of the siNA molecule.


HCV Biology and Biochemistry


In 1989, the Hepatitis C Virus (HCV) was determined to be an RNA virus and was identified as the causative agent of most non-A non-B viral Hepatitis (Choo et al., 1989, Science, 244, 359-362). Unlike retroviruses such as HIV, HCV does not go though a DNA replication phase and no integrated forms of the viral genome into the host chromosome have been detected (Houghton et al., 1991, Hepatology, 14, 381-388). Rather, replication of the coding (plus) strand is mediated by the production of a replicative (minus) strand leading to the generation of several copies of plus strand HCV RNA. The genome consists of a single, large, open-reading frame that is translated into a polyprotein (Kato et al., 1991, FEBS Letters, 280: 325-328). This polyprotein subsequently undergoes post-translational cleavage, producing several viral proteins (Leinbach et al., 1994, Virology, 204:163-169).


Examination of the 9.5-kilobase genome of HCV has demonstrated that the viral nucleic acid can mutate at a high rate (Smith et al., 1997 Mol. Evol. 45, 238-246). This rate of mutation has led to the evolution of several distinct genotypes of HCV that share approximately 70% sequence identity (Simmonds et al., 1994, J. Gen. Virol. 75, 1053-1061). It is important to note that these sequences are evolutionarily quite distant. For example, the genetic identity between humans and primates such as the chimpanzee is approximately 98%. In addition, it has been demonstrated that an HCV infection in an individual patient is composed of several distinct and evolving quasispecies that have 98% identity at the RNA level. Thus, the HCV genome is hypervariable and continuously changing. Although the HCV genome is hypervariable, there are 3 regions of the genome that are highly conserved. These conserved sequences occur in the 5′ and 3′ non-coding regions as well as the 5′-end of the core protein coding region and are thought to be vital for HCV RNA replication as well as translation of the HCV polyprotein. Thus, therapeutic agents that target these conserved HCV genomic regions may have a significant impact over a wide range of HCV genotypes. Moreover, it is unlikely that drug resistance will occur with enzymatic nucleic acids specific to conserved regions of the HCV genome. In contrast, therapeutic modalities that target inhibition of enzymes such as the viral proteases or helicase are likely to result in the selection for drug resistant strains since the RNA for these viral encoded enzymes is located in the hypervariable portion of the HCV genome.


After initial exposure to HCV, a patient experiences a transient rise in liver enzymes, which indicates that inflammatory processes are occurring (Alter et al, IN: Seeff L B, Lewis J H, eds. Current Perspectives in Hepatology. New York: Plenum Medical Book Co; 1989:83-89). This elevation in liver enzymes occurs at least 4 weeks after the initial exposure and may last for up to two months (Farci et al., 1991, New England Journal of Medicine. 325, 98-104). Prior to the rise in liver enzymes, it is possible to detect HCV RNA in the patient's serum using RT-PCR analysis (Takahashi et al., 1993, American Journal of Gastroenterology. 88, 240-243). This stage of the disease is called the acute stage and usually goes undetected since 75% of patients with acute viral hepatitis from HCV infection are asymptomatic. The remaining 25% of these patients develop jaundice or other symptoms of hepatitis.


Although acute HCV infection is a benign disease, as many as 80% of acute HCV patients progress to chronic liver disease as evidenced by persistent elevation of serum alanine aminotransferase (ALT) levels and by continual presence of circulating HCV RNA (Sherlock, 1992, Lancet, 339, 802). The natural progression of chronic HCV infection over a 10 to 20 year period leads to cirrhosis in 20 to 50% of patients (Davis et al., 1993, Infectious Agents and Disease, 2, 150, 154) and progression of HCV infection to hepatocellular carcinoma has been well documented (Liang et al., 1993, Hepatology. 18, 1326-1333; Tong et al., 1994, Western Journal of Medicine, 160, 133-138). There have been no studies that have determined sub-populations that are most likely to progress to cirrhosis and/or hepatocellular carcinoma, thus all patients have equal risk of progression.


It is important to note that the survival for patients diagnosed with hepatocellular carcinoma is only 0.9 to 12.8 months from initial diagnosis (Takahashi et al., 1993, American Journal of Gastroenterology. 88, 240-243). Treatment of hepatocellular carcinoma with chemotherapeutic agents has not proven effective and only 10% of patients will benefit from surgery due to extensive tumor invasion of the liver (Trinchet et al., 1994, Presse Medicine. 23, 831-833). Given the aggressive nature of primary hepatocellular carcinoma, the only viable treatment alternative to surgery is liver transplantation (Pichlmayr et al., 1994, Hepatology. 20, 33S-40S).


Upon progression to cirrhosis, patients with chronic HCV infection present with clinical features, which are common to clinical cirrhosis regardless of the initial cause (D'Amico et al., 1986, Digestive Diseases and Sciences. 31, 468-475). These clinical features may include: bleeding esophageal varices, ascites, jaundice, and encephalopathy (Zakim D, Boyer T D. Hepatology a textbook of liver disease. Second Edition Volume 1. 1990 W. B. Saunders Company. Philadelphia). In the early stages of cirrhosis, patients are classified as compensated, the stage at which the patient's liver is still able to detoxify metabolites in the blood-stream although liver tissue damage has occurred. In addition, most patients with compensated liver disease are asymptomatic and the minority with symptoms report only minor symptoms, such as dyspepsia and weakness. In the later stages of cirrhosis, patients are classified as decompensated, the stage at which the ability of the liver to detoxify metabolites in the bloodstream is diminished. It is at the decompensated stage that the clinical features described above present.


In 1986, D'Amico et al. described the clinical manifestations and survival rates in 1155 patients with both alcoholic and viral associated cirrhosis (D'Amico supra). Of the 1155 patients, 435 (37%) had compensated disease although 70% were asymptomatic at the beginning of the study. The remaining 720 patients (63%) had decompensated liver disease with 78% presenting with a history of ascites, 31% with jaundice, 17% had bleeding and 16% had encephalopathy. Hepatocellular carcinoma was observed in six (0.5%) patients with compensated disease and in 30 (2.6%) patients with decompensated disease.


Over the course of six years, the patients with compensated cirrhosis developed clinical features of decompensated disease at a rate of 10% per year. In most cases, ascites was the first presentation of decompensation. In addition, hepatocellular carcinoma developed in 59 patients who initially presented with compensated disease by the end of the six-year study.


With respect to survival, the D'Amico study indicated that the five-year survival rate for all patients in the study was only 40%. The six-year survival rate for the patients who initially had compensated cirrhosis was 54% while the six-year survival rate for patients who initially presented with decompensated disease was only 21%. There were no significant differences in the survival rates between the patients who had alcoholic cirrhosis and the patients with viral related cirrhosis. The major causes of death for the patients in the D'Amico study were liver failure in 49%; hepatocellular carcinoma in 22%; and bleeding in 13% (D'Amico supra).


Chronic Hepatitis C is a slowly progressing inflammatory disease of the liver, mediated by a virus (HCV) that can lead to cirrhosis, liver failure and/or hepatocellular carcinoma over a period of 10 to 20 years. In the US, it is estimated that infection with HCV accounts for 50,000 new cases of acute hepatitis in the United States each year (NIH Consensus Development Conference Statement on Management of Hepatitis C Mar. 1997). The prevalence of HCV in the United States is estimated at 1.8% and the CDC places the number of chronically infected Americans at approximately 4.5 million people. The CDC also estimates that up to 10,000 deaths per year are caused by chronic HCV infection.


Numerous well controlled clinical trials using interferon (IFN-alpha) in the treatment of chronic HCV infection have demonstrated that treatment three times a week results in lowering of serum ALT values in approximately 50% (40%-70%) of patients by the end of 6 months of therapy (Davis et al., 1989, New England Journal of Medicine, 321, 1501-1506; Marcellin et al., 1991, Hepatology, 13, 393-397; Tong et al., 1997, Hepatology, 26, 747-754; Tong et al., 1997, Hepatology, 26, 1640-1645). However, following cessation of interferon treatment, approximately 50% of the responding patients relapsed, resulting in a “durable” response rate as assessed by normalization of serum ALT concentrations of approximately 20-25%.


Direct measurement of HCV RNA is possible through use of either the branched-DNA or Reverse Transcriptase Polymerase Chain Reaction (RT-PCR) analysis. In general, RT-PCR methodology is more sensitive and leads to a more accurate assessment of the clinical course (Tong el aL, supra). Studies that have examined six months of type 1 interferon therapy using changes in HCV RNA values as a clinical endpoint have demonstrated that up to 35% of patients have a loss of HCV RNA by the end of therapy (Marcellin et al., supra). However, as with the ALT endpoint, about 50% of the patients relapse within six months following cessation of therapy, resulting in a durable virologic response of only 12% (Marcellin et al., supra). Studies that have examined 48 weeks of therapy have demonstrated that the sustained virological response is up to 25% (NIH consensus statement: 1997). Thus, standard of care for treatment of chronic HCV infection with type 1 interferon is now 48 weeks of therapy using changes in HCV RNA concentrations as the primary assessment of efficacy (Hooffnagle et al., 1997, New England Journal of Medicine, 336, 347-356).


Side effects resulting from treatment with type 1 interferons can be divided into four general categories, which include: (1) Influenza-like symptoms; (2) Neuropsychiatric; (3) Laboratory abnormalities; and (4) Miscellaneous (Dusheiko et al., 1994, Journal of Viral Hepatitis, 1, 3-5). Examples of influenza-like symptoms include fatigue, fever, myalgia, malaise, appetite loss, tachycardia, rigors, headache, and arthralgias. The influenza-like symptoms are usually short-lived and tend to abate after the first four weeks of dosing (Dushieko et al., supra). Neuropsychiatric side effects include irritability, apathy, mood changes, insomnia, cognitive changes, and depression. The most important of these neuropsychiatric side effects is depression and patients who have a history of depression should not be given type 1 interferon. Laboratory abnormalities include reduction in myeloid cells, including granulocytes, platelets and to a lesser extent red blood cells. These changes in blood cell counts rarely lead to any significant clinical sequellae (Dushieko et al., supra). In addition, increases in triglyceride concentrations and elevations in serum alanine and aspartate aminotransferase concentration have been observed. Finally, thyroid abnormalities have been reported. These thyroid abnormalities are usually reversible after cessation of interferon therapy and can be controlled with appropriate medication while on therapy. Miscellaneous side effects include nausea, diarrhea, abdominal and back pain, pruritus, alopecia, and rhinorrhea. In general, most side effects will abate after 4 to 8 weeks of therapy (Dushieko et al., supra).


The use of small interfering nucleic acid molecules targeting HCV genes and cellular/host gene targets associated with the HIV life cycle therefore provides a class of novel therapeutic agents that can be used in the treatment and diagnosis of HCV infection, liver failure, hepatocellular carcinoma, cirrhosis or any other disease or condition that responds to modulation (e.g., inhibition) of HCV genes in a subject or organism.


EXAMPLES

The following are non-limiting examples showing the selection, isolation, synthesis and activity of nucleic acids of the instant invention.


Example 1
Tandem Synthesis of siNA Constructs

Exemplary siNA molecules of the invention are synthesized in tandem using a cleavable linker, for example, a succinyl-based linker. Tandem synthesis as described herein is followed by a one-step purification process that provides RNAi molecules in high yield. This approach is highly amenable to siNA synthesis in support of high throughput RNAi screening, and can be readily adapted to multi-column or multi-well synthesis platforms.


After completing a tandem synthesis of a siNA oligo and its complement in which the 5′-terminal dimethoxytrityl (5′-O-DMT) group remains intact (trityl on synthesis), the oligonucleotides are deprotected as described above. Following deprotection, the siNA sequence strands are allowed to spontaneously hybridize. This hybridization yields a duplex in which one strand has retained the 5′-O-DMT group while the complementary strand comprises a terminal 5′-hydroxyl. The newly formed duplex behaves as a single molecule during routine solid-phase extraction purification (Trityl-On purification) even though only one molecule has a dimethoxytrityl group. Because the strands form a stable duplex, this dimethoxytrityl group (or an equivalent group, such as other trityl groups or other hydrophobic moieties) is all that is required to purify the pair of oligos, for example, by using a C 18 cartridge.


Standard phosphoramidite synthesis chemistry is used up to the point of introducing a tandem linker, such as an inverted deoxy abasic succinate or glyceryl succinate linker (see FIG. 1) or an equivalent cleavable linker. A non-limiting example of linker coupling conditions that can be used includes a hindered base such as diisopropylethylamine (DIPA) and/or DMAP in the presence of an activator reagent such as Bromotripyrrolidinophosphoniumhexaflurorophosphate (PyBrOP). After the linker is coupled, standard synthesis chemistry is utilized to complete synthesis of the second sequence leaving the terminal the 5′-O-DMT intact. Following synthesis, the resulting oligonucleotide is deprotected according to the procedures described herein and quenched with a suitable buffer, for example with 50 mM NaOAc or 1.5M NH4H2CO3.


Purification of the siNA duplex can be readily accomplished using solid phase extraction, for example, using a Waters C18 SepPak 1 g cartridge conditioned with 1 column volume (CV) of acetonitrile, 2 CV H2O, and 2 CV 50 mM NaOAc. The sample is loaded and then washed with 1 CV H2O or 50 mM NaOAc. Failure sequences are eluted with 1 CV 14% ACN (Aqueous with 50 mM NaOAc and 50 mM NaCl). The column is then washed, for example with 1 CV H2O followed by on-column detritylation, for example by passing 1 CV of 1% aqueous trifluoroacetic acid (TFA) over the column, then adding a second CV of 1% aqueous TFA to the column and allowing to stand for approximately 10 minutes. The remaining TFA solution is removed and the column washed with H2O followed by 1 CV 1 M NaCl and additional H2O. The siNA duplex product is then eluted, for example, using 1 CV 20% aqueous CAN.



FIG. 2 provides an example of MALDI-TOF mass spectrometry analysis of a purified siNA construct in which each peak corresponds to the calculated mass of an individual siNA strand of the siNA duplex. The same purified siNA provides three peaks when analyzed by capillary gel electrophoresis (CGE), one peak presumably corresponding to the duplex siNA, and two peaks presumably corresponding to the separate siNA sequence strands. Ion exchange HPLC analysis of the same siNA contract only shows a single peak. Testing of the purified siNA construct using a luciferase reporter assay described below demonstrated the same RNAi activity compared to siNA constructs generated from separately synthesized oligonucleotide sequence strands.


Example 2
Identification of Potential siNA Target Sites in any RNA Sequence

The sequence of an RNA target of interest, such as a viral or human mRNA transcript, is screened for target sites, for example by using a computer folding algorithm. In a non-limiting example, the sequence of a gene or RNA gene transcript derived from a database, such as Genbank, is used to generate siNA targets having complementarity to the target. Such sequences can be obtained from a database, or can be determined experimentally as known in the art. Target sites that are known, for example, those target sites determined to be effective target sites based on studies with other nucleic acid molecules, for example ribozymes or antisense, or those targets known to be associated with a disease or condition such as those sites containing mutations or deletions, can be used to design siNA molecules targeting those sites. Various parameters can be used to determine which sites are the most suitable target sites within the target RNA sequence. These parameters include but are not limited to secondary or tertiary RNA structure, the nucleotide base composition of the target sequence, the degree of homology between various regions of the target sequence, or the relative position of the target sequence within the RNA transcript. Based on these determinations, any number of target sites within the RNA transcript can be chosen to screen siNA molecules for efficacy, for example by using in vitro RNA cleavage assays, cell culture, or animal models. In a non-limiting example, anywhere from 1 to 1000 target sites are chosen within the transcript based on the size of the siNA construct to be used. High throughput screening assays can be developed for screening siNA molecules using methods known in the art, such as with multi-well or multi-plate assays to determine efficient reduction in target gene expression.


Example 3
Selection of siNA Molecule Target Sites in a RNA

The following non-limiting steps can be used to carry out the selection of siNAs targeting a given gene sequence or transcript.


1. The target sequence is parsed in silico into a list of all fragments or subsequences of a particular length, for example 23 nucleotide fragments, contained within the target sequence. This step is typically carried out using a custom Perl script, but commercial sequence analysis programs such as Oligo, MacVector, or the GCG Wisconsin Package can be employed as well.


2. In some instances the siNAs correspond to more than one target sequence; such would be the case for example in targeting different transcripts of the same gene, targeting different transcripts of more than one gene, or for targeting both the human gene and an animal homolog. In this case, a subsequence list of a particular length is generated for each of the targets, and then the lists are compared to find matching sequences in each list. The subsequences are then ranked according to the number of target sequences that contain the given subsequence; the goal is to find subsequences that are present in most or all of the target sequences. Alternately, the ranking can identify subsequences that are unique to a target sequence, such as a mutant target sequence. Such an approach would enable the use of siNA to target specifically the mutant sequence and not effect the expression of the normal sequence.


3. In some instances the siNA subsequences are absent in one or more sequences while present in the desired target sequence; such would be the case if the siNA targets a gene with a paralogous family member that is to remain untargeted. As in case 2 above, a subsequence list of a particular length is generated for each of the targets, and then the lists are compared to find sequences that are present in the target gene but are absent in the untargeted paralog.


4. The ranked siNA subsequences can be further analyzed and ranked according to GC content. A preference can be given to sites containing 30-70% GC, with a further preference to sites containing 40-60% GC.


5. The ranked siNA subsequences can be further analyzed and ranked according to self-folding and internal hairpins. Weaker internal folds are preferred; strong hairpin structures are to be avoided.


6. The ranked siNA subsequences can be further analyzed and ranked according to whether they have runs of GGG or CCC in the sequence. GGG (or even more Gs) in either strand can make oligonucleotide synthesis problematic and can potentially interfere with RNAi activity, so it is avoided whenever better sequences are available. CCC is searched in the target strand because that will place GGG in the antisense strand.


7. The ranked siNA subsequences can be further analyzed and ranked according to whether they have the dinucleotide UU (uridine dinucleotide) on the 3′-end of the sequence, and/or AA on the 5′-end of the sequence (to yield 3′ UU on the antisense sequence). These sequences allow one to design siNA molecules with terminal TT thymidine dinucleotides.


8. Four or five target sites are chosen from the ranked list of subsequences as described above. For example, in subsequences having 23 nucleotides, the right 21 nucleotides of each chosen 23-mer subsequence are then designed and synthesized for the upper (sense) strand of the siNA duplex, while the reverse complement of the left 21 nucleotides of each chosen 23-mer subsequence are then designed and synthesized for the lower (antisense) strand of the siNA duplex (see Tables II and III). If terminal TT residues are desired for the sequence (as described in paragraph 7), then the two 3′ terminal nucleotides of both the sense and antisense strands are replaced by TT prior to synthesizing the oligos.


9. The siNA molecules are screened in an in vitro, cell culture or animal model system to identify the most active siNA molecule or the most preferred target site within the target RNA sequence.


10. Other design considerations can be used when selecting target nucleic acid sequences, see, for example, Reynolds et al., 2004, Nature Biotechnology Advanced Online Publication, 1 Feb. 2004, doi:10.1038/nbt936 and Ui-Tei et al., 2004, Nucleic Acids Research, 32, doi:10.1093/nar/gkh247.


In an alternate approach, a pool of siNA constructs specific to a HCV target sequence is used to screen for target sites in cells expressing HCV RNA, such as human hepatoma (Huh7) cells (see for example Randall et al., 2003, PNAS USA, 100, 235-240). The general strategy used in this approach is shown in FIG. 9. A non-limiting example of such is a pool comprising sequences having any of SEQ ID NOS 1-2027. Cells expressing HCV are transfected with the pool of siNA constructs and cells that demonstrate a phenotype associated with HCV inhibition are sorted. The pool of siNA constructs can be expressed from transcription cassettes inserted into appropriate vectors (see for example FIG. 7 and FIG. 8). The siNA from cells demonstrating a positive phenotypic change (e.g., decreased HCV mRNA levels or decreased HCV protein expression), are sequenced to determine the most suitable target site(s) within the target HCV RNA sequence.


Example 4
HCV Targeted siNA Design

siNA target sites were chosen by analyzing sequences of the HCV RNA target and optionally prioritizing the target sites on the basis of folding (structure of any given sequence analyzed to determine siNA accessibility to the target), by using a library of siNA molecules as described in Example 3, or alternately by using an in vitro siNA system as described in Example 6 herein. siNA molecules were designed that could bind each target and are optionally individually analyzed by computer folding to assess whether the siNA molecule can interact with the target sequence. Varying the length of the siNA molecules can be chosen to optimize activity. Generally, a sufficient number of complementary nucleotide bases are chosen to bind to, or otherwise interact with, the target RNA, but the degree of complementarity can be modulated to accommodate siNA duplexes or varying length or base composition. By using such methodologies, siNA molecules can be designed to target sites within any known RNA sequence, for example those RNA sequences corresponding to the any gene transcript.


Chemically modified siNA constructs are designed to provide nuclease stability for systemic administration in vivo and/or improved pharmacokinetic, localization, and delivery properties while preserving the ability to mediate RNAi activity. Chemical modifications as described herein are introduced synthetically using synthetic methods described herein and those generally known in the art. The synthetic siNA constructs are then assayed for nuclease stability in serum and/or cellular/tissue extracts (e.g. liver extracts). The synthetic siNA constructs are also tested in parallel for RNAi activity using an appropriate assay, such as a luciferase reporter assay as described herein or another suitable assay that can quantity RNAi activity. Synthetic siNA constructs that possess both nuclease stability and RNAi activity can be further modified and re-evaluated in stability and activity assays. The chemical modifications of the stabilized active siNA constructs can then be applied to any siNA sequence targeting any chosen RNA and used, for example, in target screening assays to pick lead siNA compounds for therapeutic development (see for example FIG. 11).


Example 5
Chemical Synthesis and Purification of siNA

siNA molecules can be designed to interact with various sites in the RNA message, for example, target sequences within the RNA sequences described herein. The sequence of one strand of the siNA molecule(s) is complementary to the target site sequences described above. The siNA molecules can be chemically synthesized using methods described herein. Inactive siNA molecules that are used as control sequences can be synthesized by scrambling the sequence of the siNA molecules such that it is not complementary to the target sequence. Generally, siNA constructs can by synthesized using solid phase oligonucleotide synthesis methods as described herein (see for example Usman et al., U.S. Pat. Nos. 5,804,683; 5,831,071; 5,998,203; 6,117,657; 6,353,098; 6,362,323; 6,437,117; 6,469,158; Scaringe et al., U.S. Pat. Nos. 6,111,086; 6,008,400; 6,111,086 all incorporated by reference herein in their entirety).


In a non-limiting example, RNA oligonucleotides are synthesized in a stepwise fashion using the phosphoramidite chemistry as is known in the art. Standard phosphoramidite chemistry involves the use of nucleosides comprising any of 5′-O-dimethoxytrityl, 2′-O-tert-butyldimethylsilyl, 3′-O-2-Cyanoethyl N,N-diisopropylphos-phoroamidite groups, and exocyclic amine protecting groups (e.g. N6-benzoyl adenosine, N4 acetyl cytidine, and N2-isobutyryl guanosine). Alternately, 2′-O-Silyl Ethers can be used in conjunction with acid-labile 2′-O-orthoester protecting groups in the synthesis of RNA as described by Scaringe supra. Differing 2′ chemistries can require different protecting groups, for example 2′-deoxy-2′-amino nucleosides can utilize N-phthaloyl protection as described by Usman et al., U.S. Pat. No. 5,631,360, incorporated by reference herein in its entirety).


During solid phase synthesis, each nucleotide is added sequentially (3′- to 5′-direction) to the solid support-bound oligonucleotide. The first nucleoside at the 3′-end of the chain is covalently attached to a solid support (e.g., controlled pore glass or polystyrene) using various linkers. The nucleotide precursor, a ribonucleoside phosphoramidite, and activator are combined resulting in the coupling of the second nucleoside phosphoramidite onto the 5′-end of the first nucleoside. The support is then washed and any unreacted 5′-hydroxyl groups are capped with a capping reagent such as acetic anhydride to yield inactive 5′-acetyl moieties. The trivalent phosphorus linkage is then oxidized to a more stable phosphate linkage. At the end of the nucleotide addition cycle, the 5′-O-protecting group is cleaved under suitable conditions (e.g., acidic conditions for trityl-based groups and Fluoride for silyl-based groups). The cycle is repeated for each subsequent nucleotide.


Modification of synthesis conditions can be used to optimize coupling efficiency, for example by using differing coupling times, differing reagent/phosphoramidite concentrations, differing contact times, differing solid supports and solid support linker chemistries depending on the particular chemical composition of the siNA to be synthesized. Deprotection and purification of the siNA can be performed as is generally described in Usman et al., U.S. Pat. No. 5,831,071, U.S. Pat. No. 6,353,098, U.S. Pat. No. 6,437,117, and Bellon et al., U.S. Pat. No. 6,054,576, U.S. Pat. No. 6,162,909, U.S. Pat. No. 6,303,773, or Scaringe supra, incorporated by reference herein in their entireties. Additionally, deprotection conditions can be modified to provide the best possible yield and purity of siNA constructs. For example, applicant has observed that oligonucleotides comprising 2′-deoxy-2′-fluoro nucleotides can degrade under inappropriate deprotection conditions. Such oligonucleotides are deprotected using aqueous methylamine at about 35° C. for 30 minutes. If the 2′-deoxy-2′-fluoro containing oligonucleotide also comprises ribonucleotides, after deprotection with aqueous methylamine at about 35° C. for 30 minutes, TEA-HF is added and the reaction maintained at about 65° C. for an additional 15 minutes.


Example 6
RNAi In Vitro Assay to Assess siNA Activity

An in vitro assay that recapitulates RNAi in a cell-free system is used to evaluate siNA constructs targeting HCV RNA targets. The assay comprises the system described by Tuschl et al., 1999, Genes and Development, 13, 3191-3197 and Zamore et al., 2000, Cell, 101, 25-33 adapted for use with HCV target RNA. A Drosophila extract derived from syncytial blastoderm is used to reconstitute RNAi activity in vitro. Target RNA is generated via in vitro transcription from an appropriate HCV expressing plasmid using T7 RNA polymerase or via chemical synthesis as described herein. Sense and antisense siNA strands (for example 20 uM each) are annealed by incubation in buffer (such as 100 mM potassium acetate, 30 mM HEPES-KOH, pH 7.4, 2 mM magnesium acetate) for 1 minute at 90° C. followed by 1 hour at 37° C., then diluted in lysis buffer (for example 100 mM potassium acetate, 30 mM HEPES-KOH at pH 7.4, 2 mM magnesium acetate). Annealing can be monitored by gel electrophoresis on an agarose gel in TBE buffer and stained with ethidium bromide. The Drosophila lysate is prepared using zero to two-hour-old embryos from Oregon R flies collected on yeasted molasses agar that are dechorionated and lysed. The lysate is centrifuged and the supernatant isolated. The assay comprises a reaction mixture containing 50% lysate [vol/vol], RNA (10-50 pM final concentration), and 10% [vol/vol] lysis buffer containing siNA (10 nM final concentration). The reaction mixture also contains 10 mM creatine phosphate, 10 ug/ml creatine phosphokinase, 100 um GTP, 100 uM UTP, 100 uM CTP, 500 uM ATP, 5 mM DTT, 0.1 U/uL RNasin (Promega), and 100 uM of each amino acid. The final concentration of potassium acetate is adjusted to 100 mM. The reactions are pre-assembled on ice and preincubated at 25° C. for 10 minutes before adding RNA, then incubated at 25° C. for an additional 60 minutes. Reactions are quenched with 4 volumes of 1.25×Passive Lysis Buffer (Promega). Target RNA cleavage is assayed by RT-PCR analysis or other methods known in the art and are compared to control reactions in which siNA is omitted from the reaction.


Alternately, internally-labeled target RNA for the assay is prepared by in vitro transcription in the presence of [alpha-32P] CTP, passed over a G50 Sephadex column by spin chromatography and used as target RNA without further purification. Optionally, target RNA is 5′-32P-end labeled using T4 polynucleotide kinase enzyme. Assays are performed as described above and target RNA and the specific RNA cleavage products generated by RNAi are visualized on an autoradiograph of a gel. The percentage of cleavage is determined by PHOSPHOR IMAGER® (autoradiography) quantitation of bands representing intact control RNA or RNA from control reactions without siNA and the cleavage products generated by the assay.


In one embodiment, this assay is used to determine target sites in the HCV RNA target for siNA mediated RNAi cleavage, wherein a plurality of siNA constructs are screened for RNAi mediated cleavage of the HCV RNA target, for example, by analyzing the assay reaction by electrophoresis of labeled target RNA, or by northern blotting, as well as by other methodology well known in the art.


Example 7
Nucleic Acid Inhibition of HCV Target RNA

siNA molecules targeted to the human HCV RNA are designed and synthesized as described above. These nucleic acid molecules can be tested for cleavage activity in vivo, for example, using the following procedure. The target sequences and the nucleotide location within the HCV RNA are given in Tables II and III.


Two formats are used to test the efficacy of siNAs targeting HCV. First, the reagents are tested in cell culture using, for example, human hepatoma (Huh7) cells, to determine the extent of RNA and protein inhibition. siNA reagents (e.g.; see Tables II and III) are selected against the HCV target as described herein. RNA inhibition is measured after delivery of these reagents by a suitable transfection agent to, for example, cultured epidermal keratinocytes. Relative amounts of target RNA are measured versus actin using real-time PCR monitoring of amplification (eg., ABI 7700 TAQMAN®). A comparison is made to a mixture of oligonucleotide sequences made to unrelated targets or to a randomized siNA control with the same overall length and chemistry, but randomly substituted at each position. Primary and secondary lead reagents are chosen for the target and optimization performed. After an optimal transfection agent concentration is chosen, a RNA time-course of inhibition is performed with the lead siNA molecule. In addition, a cell-plating format can be used to determine RNA inhibition.


In addition, a cell-plating format can be used to determine RNA inhibition. A non-limiting example of a multiple target screen to assay siNA mediated inhibition of HCV RNA is shown in FIG. 28. siNA constructs (Table III) were transfected at 25 nM into Huh7 cells and HCV RNA quantitated compared to untreated cells (“cells” column in the figure) and cells transfected with lipofectamine (“LFA2K” column in the figure). As shown in FIG. 28, several siNA constructs show significant inhibition of HCV RNA expression in the Huh7 replicon system. This system is described in Rice et al., U.S. Pat. No. 5,874,565 and U.S. Pat. No. 6,127,116, both incorporated by reference herein.


Delivery of siNA to Cells


Huh7b cells stably transfected with the HCV subgenomic replicon Clone A or Ava.5 are seeded, for example, at 8.5×103 cells per well of a 96-well platein DMEM(Gibco) the day before transfection. siNA (final concentration, for example 25 nM) and cationic lipid Lipofectamine2000 (e.g., final concentration0.5 μl/well) are complexed in Optimem (Gibco) at 37° C. for 20 minutes inpolypropelyne microtubes. Following vortexing, the complexed siNA is added to each well and incubated for 24-72 hours.


TAQMAN® (Real-Time PCR Monitoring of Amplification) and Lightcycler Quantification of mRNA


Total RNA is prepared from cells following siNA delivery, for example, using Ambion Rnaqueous 4-PCR purification kit for large scale extractions, or Ambion Rnaqueous-96 purification kit for 96-well assays. For Taqman analysis, dual-labeled probes are synthesized with, for example, the reporter dyes FAM or VIC covalently linked at the 5′-end and the quencher dye TAMARA conjugated to the 3′-end. One-step RT-PCR amplifications are performed on, for example, an ABI PRISM 7700 Sequence detector using 50 uL reactions consisting of 10 uL total RNA, 100 nM forward primer, 100 mM reverse primer, 100 nM probe, 1×TaqMan PCR reaction buffer (PE-Applied Biosystems), 5.5 mM MgCl2, 100 uM each dATP, dCTP, dGTP and dTTP, 0.2U RNase Inhibitor (Promega), 0.025U AmpliTaq Gold (PE-Applied Biosystems) and 0.2U M-MLV Reverse Transcriptase (Promega). The thermal cycling conditions can consist of 30 minutes at 48° C., 10 minutes at 95° C., followed by 40 cycles of 15 seconds at 95° C. and 1 minute at 60° C. Quantitation of target mRNA level is determined relative to standards generated from serially diluted total cellular RNA (300, 100, 30, 10 ng/rxn) and normalizing to, for example, 36B4 mRNA in either parallel or same tube TaqMan reactions. For HCV Replicon mRNA quantitation, PCR primers and probe specific for the neomycin gene were used:

neo-forward primer,(SEQ ID NO: 2032)5′-CCGGCTACCTGCCCATTC-3′;neo-reverse primer,(SEQ ID NO: 2033)5′-CCAGATCATCCTGATCGACAAG-3′;neo-probe,(SEQ ID NO: 2034)5′FAM-ACATCGCATCGAGCGAGCACGTAC-TAMARA3′;


For normalization, 36B4 PCR primers and probe were used:

36B4-forward primer,(SEQ ID NO: 2035)5′-TCTATCATCAACGGGTACAAACGA-3′;36B4 reverse primer,(SEQ ID NO: 2036)5′-CTTTTCAGCAAGTGGGAAGGTG-3′;36B4 probe,(SEQ ID NO: 2037)5′VIC-CCTGGCCTfGTCTGTGGAGACGGATTA-TAMARA3′;


Western Blotting


Nuclear extracts can be prepared using a standard micro preparation technique (see for example Andrews and Faller, 1991, Nucleic Acids Research, 19, 2499). Protein extracts from supernatants are prepared, for example using TCA precipitation. An equal volume of 20% TCA is added to the cell supernatant, incubated on ice for 1 hour and pelleted by centrifugation for 5 minutes. Pellets are washed in acetone, dried and resuspended in water. Cellular protein extracts are run on a 10% Bis-Tris NuPage (nuclear extracts) or 4-12% Tris-Glycine (supernatant extracts) polyacrylamide gel and transferred onto nitro-cellulose membranes. Non-specific binding can be blocked by incubation, for example, with 5% non-fat milk for 1 hour followed by primary antibody for 16 hour at 4° C. Following washes, the secondary antibody is applied, for example (1:10,000 dilution) for 1 hour at room temperature and the signal detected with SuperSignal reagent (Pierce).


Example 8
Models Useful to Evaluate the Down-Regulation of HCV Gene Expression

Cell Culture


Although there have been reports of replication of HCV in cell culture (see below), these systems are difficult to reproduce and have proven unreliable. Therefore, as was the case for development of other anti-HCV therapeutics, such as interferon and ribavirin, after demonstration of safety in animal studies applicant can proceed directly into a clinical feasibility study.


Several recent reports have documented in vitro growth of HCV in human cell lines (Mizutani et al., Biochem Biophys Res Commun 1996 227(3):822-826; Tagawa et al., Journal of Gasteroenterology and Hepatology 1995 10(5):523-527; Cribier et al., Journal of General Virology 76(10):2485-2491; Seipp et al., Journal of General Virology 1997 78(10)2467-2478; lacovacci et al., Research Virology 1997 148(2):147-151; Iocavacci et al., Hepatology 1997 26(5) 1328-1337; Ito et al., Journal of General Virology 1996 77(5):1043-1054; Nakajima et al., Journal of Virology 1996 70(5):3325-3329; Mizutani et al., Journal of Virology 1996 70(10):7219-7223; Valli et al., Res Virol 1995 146(4): 285-288; Kato et al., Biochem Biophys Res Comm 1995 206(3):863-869). Replication of HCV has been reported in both T and B cell lines, as well as cell lines derived from human hepatocytes. Detection of low level replication was documented using either RT-PCR based assays or the b-DNA assay. It is important to note that the most recent publications regarding HCV cell cultures document replication for up to 6-months. However, the level of HCV replication observed in these cell lines has not been robust enough for screening of antiviral compounds.


In addition to cell lines that can be infected with HCV, several groups have reported the successful transformation of cell lines with cDNA clones of full-length or partial HCV genomes (Harada et al., Journal of General Virology, 1995, 76(5)1215-1221; Haramatsu et al., Journal of Viral Hepatitis 1997 4S(1):61-67; Dash et al., American Journal of Pathology 1997 151(2):363-373; Mizuno et al., Gasteroenterology 1995 109(6):1933-40; Yoo et al., Journal Of Virology 1995 69(1):32-38).


The recent development of subgenomic HCV RNA replicons capable of successfully replicating in the human hepatoma cell line, Huh7, represents a significant advance toward a dependable cell culture model. These replicons contain the neomycin gene upstream of the HCV nonstructural genes allowing for the selection of replicative RNAs in Huh7 cells. Initially, RNA replication was detected at a low frequency (Lohmann et al. Science 1999 285: 110-113) but the identification of replicons with cell-adaptive mutations in the NS5A region has improved the efficiency of replication 10,000-fold (Blight et al. Science 2000 290:1972-1975). Steps in the HCV life cycle, such as translation, protein processing, and RNA replication are recapitulated in the subgenomic replicon systems, but early events (viral attachment and uncoating) and viral assembly is absent. Inclusion of the structural genes of HCV within the replicons results in the production of HCV core and envelope proteins, but virus assembly does not occur (Pietschmann et al. Journal of Virology 2002 76: 4008-4021). Such replicon systems have been used to study siRNA mediated inhibition of HCV RNA, see for example, Randall et al., 2003, PNAS USA, 100, 235-240.


In several cell culture systems, cationic lipids have been shown to enhance the bioavailability of oligonucleotides to cells in culture (Bennet, et al., 1992, Mol. Pharmacology, 41, 1023-1033). In one embodiment, siNA molecules of the invention are complexed with cationic lipids for cell culture experiments. siNA and cationic lipid mixtures are prepared in serum-free DMEM immediately prior to addition to the cells. DMEM plus additives are warmed to room temperature (about 20-25° C.) and cationic lipid is added to the final desired concentration and the solution is vortexed briefly. siNA molecules are added to the final desired concentration and the solution is again vortexed briefly and incubated for 10 minutes at room temperature. In dose response experiments, the RNA/lipid complex is serially diluted into DMEM following the 10 minute incubation.


Animal Models


Evaluating the efficacy of anti-HCV agents in animal models is an important prerequisite to human clinical trials. The best characterized animal system for HCV infection is the chimpanzee. Moreover, the chronic hepatitis that results from HCV infection in chimpanzees and humans is very similar. Although clinically relevant, the chimpanzee model suffers from several practical impediments that make use of this model difficult. These include high cost, long incubation requirements and lack of sufficient quantities of animals. Due to these factors, a number of groups have attempted to develop rodent models of chronic hepatitis C infection. While direct infection has not been possible, several groups have reported on the stable transfection of either portions or entire HCV genomes into rodents (Yamamoto et al., Hepatology 1995 22(3): 847-855; Galun et al., Journal of Infectious Disease 1995 172(1):25-30; Koike et al., Journal of general Virology 1995 76(12)3031-3038; Pasquinelli et al., Hepatology 1997 25(3): 719-727; Hayashi et al., Princess Takamatsu Symp 1995 25:1430149; Mariya et al., Journal of General Virology 1997 78(7) 1527-1531; Takehara et al., Hepatology 1995 21(3):746-751; Kawamura et al., Hepatology 1997 25(4): 1014-1021). In addition, transplantation of HCV infected human liver into immunocompromised mice results in prolonged detection of HCV RNA in the animal's blood.


A method for expressing hepatitis C virus in an in vivo animal model has been developed (Vierling, International PCT Publication No. WO 99/16307). Viable, HCV infected human hepatocytes are transplanted into a liver parenchyma of a scid/scid mouse host. The scid/scid mouse host is then maintained in a viable state, whereby viable, morphologically intact human hepatocytes persist in the donor tissue and hepatitis C virus is replicated in the persisting human hepatocytes. This model provides an effective means for the study of HCV inhibition by enzymatic nucleic acids in vivo.


As such, these models can be used in evaluating the efficacy of siNA molecules of the invention in inhibiting HCV expression. These models and others can similarly be used to evaluate the safety and efficacy of siNA molecules of the invention in a pre-clinical setting.


Example 9
RNAi Mediated Inhibition of HCV Expression

siNA constructs (Table III) are tested for efficacy in reducing HCV RNA expression in, for example, Huh7 cells. Cells are plated approximately 24 hours before transfection in 96-well plates at 5,000-7,500 cells/well, 100 μl/well, such that at the time of transfection cells are 70-90% confluent. For transfection, annealed siNAs are mixed with the transfection reagent (Lipofectamine 2000, Invitrogen) in a volume of 50 μl/well and incubated for 20 minutes at room temperature. The siNA transfection mixtures are added to cells to give a final siNA concentration of 25 nM in a volume of 150 μl. Each siNA transfection mixture is added to 3 wells for triplicate siNA treatments. Cells are incubated at 37° for 24 hours in the continued presence of the siNA transfection mixture. At 24 hours, RNA is prepared from each well of treated cells. The supernatants with the transfection mixtures are first removed and discarded, then the cells are lysed and RNA prepared from each well. Target gene expression following treatment is evaluated by RT-PCR for the target gene and for a control gene (36B4, an RNA polymerase subunit) for normalization. The triplicate data is averaged and the standard deviations determined for each treatment. Normalized data are graphed and the percent reduction of target mRNA by active siNAs in comparison to their respective inverted control siNAs is determined.


Example 10
siNA Inhibition of a Chimeric HCV/Poliovirus in HeLa Cells

Inhibition of a chimeric HCV/Poliovirus was investigated using 21 nucleotide siNA duplexes in HeLa cells. Seven siNA were designed that target three regions in the highly conserved 5′ untranslated region (UTR) of HCV RNA. The siNAs were screened in two cell culture systems dependent upon the 5′-UTR of HCV; one requires translation of an HCV/luciferase gene, while the other involves replication of a chimeric HCV/poliovirus (PV) (see Blatt et al., U.S. Ser. No. 09/740,332, filed Dec. 18, 2000, incorporated by reference herein). Transfection for the HCV/PV system was performed in HeLa cells (grown in DMEM supplemented with sodium pyruvate and 100 mM HEPES with 5% FBS) using either cationic lipid NC168 or LFA2K, with an siNA concentration of 10 nM or 25 nM. HeLa cells were innoculated with HCV/PV virus at an moi=0.01 pfu/cell for 30 minutes in serum-free media. The innoculum was removed and 80 μL media was added, with 20 μL of transfection complex added to each well. The cells and supernatants were frozen at 20-24 hours post transfection. Each plate underwent 3 freeze-thaw cycles and the supernatant was collected. The supernatant was titered on HeLa cells for 3 days, then stained and counted. The results shown in FIGS. 24-27 are reported as pfu/ml×105.


Two siNAs (29579/29586 and 29578/29585, see Table III) targeting the same region (shifted by one nucleotide) are active in both systems (see FIG. 22). For example, a >85% reduction in HCVPV replication was observed in siNA-treated cells compared to an inverse siNA control 29593/29600 (FIG. 22) with an IC50=˜2.5 nM (FIG. 23). To develop nuclease-resistant siNA for in vivo applications, siNAs can be modified to contain stabilizing chemical modifications. Such modifications include phosphorothioate linkages (P═S), 2′-Omethyl nucleotides, 2′-fluoro (F) nucleotides, 2′-deoxy nucleotides, universal base nucleotides, 5′ and/or 3′ end modifications and a variety of other nucleotide and non-nucleotide modifications, such as those described herein, in one or both siNA strands. Using this systematic approach, active siNA molecules have been identified that are substantially more resistant to nucleases. Several of these constructs were tested in the HCV/poliovirus chimera system, demonstrating significant reduction in viral replication (see FIGS. 24-27). siNA constructs shown in FIGS. 24-27 are referred to by Compound numbers that are cross referenced and identified in Table III. siNA activity is compared to relevant controls (untreated cells, scrambled/inactive control sequences, or transfection controls). FIG. 24 shows the inhibition of HCV RNA in the HCV/poliovirus chimera system using chemically modified siNA construct 30051/30053, which construct has inverted deoxy abasic nucleotides at the 3′ and 5′ ends, several phosphorothioate linkages, and 5-nitroindole nucleotides. FIG. 25 shows the inhibition of HCV RNA in the HCV/poliovirus chimera system using chemically modified siNA construct 30055/30057, which construct has inverted deoxy abasic nucleotides at the 3′ and 5′ ends, several phosphorothioate linkages, and 5-nitroindole nucleotides. FIGS. 26 and 27 show the inhibition of HCV RNA in the HCV/poliovirus chimera system using unmodified siNA construct (29586/29579) and chemically modified siNA constructs 30417/30419, 30417/30420, 30418/30419, and combinations thereof at 10 nM and 25 nM siNA, respectively. As shown in FIGS. 24-27, siNA constructs of the invention provide potent inhibition of HCV RNA in the HCV/poliovirus chimera system. As such, siNA constructs, inlcuding chemically modified, nuclease resistant siNA molecules, represent an important class of therapeutic agents for treating chronic HCV infection.


Example 11
siNA Inhibition of HCV RNA Expression in a HCV Replicon System

A HCV replicon system was used to test the efficacy of siNAs targeting HCV RNA. The reagents are tested in cell culture using Huh7 cells (see for example Randall et al., 2003, PNAS USA, 100, 235-240) to determine the extent of RNA and protein inhibition. siNA were selected against the HCV target as described herein. RNA inhibition was measured after delivery of these reagents by a suitable transfection agent to Huh7 cells. Relative amounts of target RNA are measured versus actin using real-time PCR monitoring of amplification (eg., ABI 7700 Taqman®). A comparison is made to a mixture of oligonucleotide sequences designed to target unrelated targets or to a randomized siNA control with the same overall length and chemistry, but with randomly substituted nucleotides at each position. Primary and secondary lead reagents were chosen for the target and optimization performed. After an optimal transfection agent concentration is chosen, a RNA time-course of inhibition is performed with the lead siNA molecule. In addition, a cell-plating format can be used to determine RNA inhibition. A non-limiting example of a multiple target screen to assay siNA mediated inhibition of HCV RNA is shown in FIG. 28. siNA reagents (Table III) were transfected at 25 nM into Huh7 cells and HCV RNA quantitated compared to untreated cells (“cells” column in the figure),cells transfected with lipofectamine (“LFA2K” column in the figure) and matched chemistry inverted controls (“Inv”). As shown in the Figure, several siNA constructs show significant inhibition of HCV RNA expression in the Huh7 replicon system. Chemically modified siNA constructs were then screened as described above, with a non-limiting example of a Stab 7/8 (see Table IV) chemisty siNA construct screen shown in FIG. 30. A follow up dose response study using chemically modified siNA constructs (Stab 4/5, see Table 1) at concentrations of 5 nM, 10 nM, 25 nM and 100 nM compared to matched chemistry inverted controls is shown in FIG. 29, whereas a dose response study for Stab 7/8 constructs at concentrations of 5 nM, 10 nM, 25 nM, 50 nM and 100 nM compared to matched chemistry inverted controls is shown in FIG. 31.


Example 12
Effect of Interferon/siNA Combination Treatment on Replication of HCV Subgenomic Replicon in Huh7 Cells

To investigate combination use of RNAi and interferon in the inhibition of HCV replication, siNA and interferon combination treatments were assayed in the HCV Subgenomic Replicon in Huh7 cells. Huh7 cells containing the HCV subgenomic replicon Clone A were plated in 96-well plates at a density of 9,600 cells per well and incubated overnight at 37° C. The cells were then treated with interferon alone, siNAs or inverted sequence controls alone, or with interferon in combination with siNAs or inverted controls. A sub-optimal dose of interferon was used in order to observe possible potentiation of the interferon anti-viral activity in the presence of the HCV-targeted siNA. The cells were transfected with HCV targeted siNAs (31703/31707) or inverted sequence controls (31711/31715) at 5, 10, 25, 50, or 100 nM using 0.35 ul/well of Lipofectamine 2000 in media alone, or media to which was added 1.7 Units/ml of Infergen (Amgen). The cells were then incubated at 37° C. for 48 or 72 hours, at which time total RNA was isolated using an Invitek 96-well RNA isolation kit. To quantitate the levels of RNA from the HCV replicon, real-time RT-PCR was performed using probes and primers to the neomycin resistance region of the replicon. Results are shown in FIG. 32. Levels of the replicon RNA were normalized to the levels of cellular GAPDH mRNA. These data demonstrate potentiation of the effect of combination siNA/interferon treatment compared to interferon alone.


Example 13
Multifunctional siNA Inhibition of HCV RNA Expression in a HCV Replicon System

To investigate the use of multifunctional siNA constructs that target different sites within HCV RNA via RNAi, multifunctional siNA treatments were assayed in the HCV Subgenomic Replicon in Huh7 cells. The multifunctional siNA constructs that target either sites 304 and 327 or sites 282 and 304 of HCV RNA were compared to pools of siNA molecules that individually target site 304 and site 327 or site 282 and site 304 of HCV RNA. Huh7 cells containing the HCV subgenomic replicon Clone A were plated in 96-well plates at a density of 9,600 cells per well and incubated overnight at 37° C. The cells were then treated with the individual siNAs, pooled siNAs, multifunctional siNAs, irrelevant multifunctional siNA controls, and a transfection control (LFA only). The cells were then incubated at 37° C. for 48 or 72 hours, at which time total RNA was isolated using an Invitek 96-well RNA isolation kit. To quantitate the levels of RNA from the HCV replicon, real-time RT-PCR was performed using probes and primers to the neomycin resistance region of the replicon. Dose response results for unmodified multifunctional siNA constructs assayed at 0.1, 1.0 and 10 nM are shown in FIG. 33, whereas results for modified multifunctional siNA constructs are shown in FIGS. 34-41. Levels of the replicon RNA were normalized to the levels of cellular GAPDH mRNA. These data demonstrate that the multifunctional siNA constructs are equally effective in inhibition of HCV RNA expression by targeting multiple sites as are pools of individual siNA constructs that target each site.


Example 14
Multifunctional siNA Design

Once target sites have been identified for multifunctional siNA constructs, each strand of the siNA is designed with a complementary region of length, for example, between about 18 and about 28 nucleotides, that is complementary to a different target nucleic acid sequence. Each complementary region is designed with an adjacent flanking region of about 4 to about 22 nucleotides that is not complementary to the target sequence, but which comprises complementarity to the complementary region of the other sequence (see for example FIG. 16). Hairpin constructs can likewise be designed (see for example FIG. 17). Identification of complementary, palindrome or repeat sequences that are shared between the different target nucleic acid sequences can be used to shorten the overall length of the multifunctional siNA constructs (see for example FIGS. 18 and 19).


In a non-limiting example, a multifunctional siNA is designed to target two separate nucleic acid sequences. The goal is to combine two different siNAs together in one siNA that is active against two different targets. The siNAs are joined in a way that the 5′ of each strand starts with the “antisense” sequence of one of two siRNAs as shown in italics below.

SEQ ID NO: 20383′ TTAGAAACCAGACGUAAGUGU GGUACGACCUGACGACCGU 5′SEQ ID NO: 20395′ UCUUUGGUCUGCAUUCACAC CAUGCUGGACUGCUGGCATT3′


RISC is expected to incorporate either of the two strands from the 5′ end. This would lead to two types of active RISC populations carrying either strand. The 5′ 19 nt of each strand will act as guide sequence for degradation of separate target sequences.


In another example, the size of multifunctional siNA molecules is reduced by either finding overlaps or truncating the individual siNA length. The exemplary excercise described below indicates that for any given first target sequence, a shared complementary sequence in a second target sequence is likely to be found.


The number of spontaneous matches of short polynucleotide sequences (e.g., less than 14 nucleotides) that are expected to occur between two longer sequences generated independent of one another was investigated. A simulation using the uniform random generator SAS V8.1 utilized a 4,000 character string that was generated as a random repeating occurrence of the letters {ACGU}. This sequence was then broken into the nearly 4000 overlapping sets formed by taking S1 as the characters from positions (1,2 . . . n), S2 from positions (2,3 . . . , n+1) completely through the sequence to the last set, S 4000-n+1 from position (4000-n+1, . . . , 4000). This process was then repeated for a second 4000 character string. Occurrence of same sets (of size n) were then checked for sequence identity between the two strings by a sorting and match-merging routine. This procedure was repeated for sets of 9-11 characters. Results were an average of 55 matching sequences of length n=9 characters (range 39 to 72); 13 common sets (range 6 to 18) for size n=10, and 4 matches on average (range 0 to 6) for sets of 11 characters. The choice of 4000 for the original string length is approximately the length of the coding region of both target nucleic acid molecules. This simple simulation suggests that any two long coding regions formed independent of one-another will share common short sequences that can be used to shorten the length of multifunctional siNA constructs. In this example, common sequences of size 9 occurred by chance alone in >1% frequency.


In another example, the length of a multifunctional siNA construct is reduced by determining whether fewer base pairs of sequence homology to each target sequence can be tolerated for effective RNAi activity. If so, the overall length of multifunctional siNA can be reduced as shown below. In the following hypothetical example, 4 nucleotides (bold) are reduced from each 19 nucleotide siNA ′1′and siNA ′2′ constructs. The resulting multifunctional siNA is 30 base pairs long.


Additional Multifuctional siNA Designs


Three categories of additional multifunctional siNA designs are presented that allow a single siNA molecule to silence multiple targets. The first method utilizes linkers to join siNAs (or multiunctional siNAs) in a direct manner. This can allow the most potent siNAs to be joined without creating a long, continuous stretch of RNA that has potential to trigger an interferon response. The second method is a dendrimeric extension of the overlapping or the linked multifunctional design; or alternatively the organization of siNA in a supramolecular format. The third method uses helix lengths greater than 30 base pairs. Processing of these siNAs by Dicer will reveal new, active 5′ antisense ends. Therefore, the long siNAs can target the sites defined by the original 5′ ends and those defined by the new ends that are created by Dicer processing. When used in combination with traditional multifunctional siNAs (where the sense and antisense strands each define a target) the approach can be used for example to target 4 or more sites.


I. Tethered Bifunctional siNAs


The basic idea is a novel approach to the design of multifunctional siNAs in which two antisense siNA strands are annealed to a single sense strand. The sense strand oligonucleotide contains a linker (e.g., non-nulcoetide linker as described herein) and two segments that anneal to the antisense siNA strands (see FIG. 42). The linkers can also optionally comprise nucleotide-based linkers. Several potential advantages and variations to this approach include, but are not limited to:


1. The two antisense siNAs are independent. Therefore, the choice of target sites is not constrained by a requirement for sequence conservation between two sites. Any two highly active siNAs can be combined to form a multifunctional siNA.


2. When used in combination with target sites having homology, siNAs that target a sequence present in two genes (e.g., different HCV strains), the design can be used to target more than two sites. A single multifunctional siNA can be for example, used to target RNA of two different HCV RNAs (using one antisense strand of the multifunctional siNA targeting of conserved sequence between to the two RNAs) and a host RNA (using the second antisense strand of the multifunctional siNA targeting host RNA (e.g., La antigen or FAS) This approach allows targeting of more than one HCV strain and one or more host RNAs using a single multifunctional siNA.


3. Multifunctional siNAs that use both the sense and antisense strands to target a gene can also be incorporated into a tethered multifuctional design. This leaves open the possibility of targeting 6 4 or more sites with a single complex.


4. It can be possible to anneal more than two antisense strand siNAs to a single tethered sense strand.


5. The design avoids long continuous stretches of dsRNA. Therefore, it is less likely to initiate an interferon response.


6. The linker (or modifications attached to it, such as conjugates described herein) can improve the pharmacokinetic properties of the complex or improve its incorporation into liposomes. Modifications introduced to the linker should not impact siNA activity to the same extent that they would if directly attached to the siNA (see for example FIGS. 48 and 49).


7. The sense strand can extend beyond the annealed antisense strands to provide additional sites for the attachment of conjugates.


8. The polarity of the complex can be switched such that both of the antisense 3′ ends are adjacent to the linker and the 5′ ends are distal to the linker or combination thereof.


Dendrimer and Supramolecular siNAs


In the dendrimer siNA approach, the synthesis of siNA is initiated by first synthesizing the dendrimer template followed by attaching various functional siNAs. Various constructs are depicted in FIG. 43. The number of functional siNAs that can be attached is only limited by the dimensions of the dendrimer used.


Supramolecular Approach to Multifunctional siNA


The supramolecular format simplifies the challenges of dendrimer synthesis. In this format, the siNA strands are synthesized by standard RNA chemistry, followed by annealing of various complementary strands. The individual strand synthesis contains an antisense sense sequence of one siNA at the 5′-end followed by a nucleic acid or synthetic linker, such as hexaethyleneglyol, which in turn is followed by sense strand of another siNA in 5′ to 3′ direction. Thus, the synthesis of siNA strands can be carried out in a standard 3′ to 5′ direction. Representative examples of trifunctional and tetrafunctional siNAs are depicted in FIG. 44. Based on a similar principle, higher functionality siNA constucts can be designed as long as efficient annealing of various strands is achieved.


Dicer enabled multifunctional siNA


Using bioinformatic analysis of multiple targets, stretches of identical sequences shared between differeing target sequences can be identified ranging from about two to about fourteen nucleotides in length. These identical regions can be designed into extended siNA helixes (e.g., >30 base pairs) such that the processing by Dicer reveals a secondary functional 5′-antisense site (see for example FIG. 45). For example, when the first 17 nucleotides of a siNA antisense strand (e.g., 21 nucleotide strands in a duplex with 3′-TT overhangs) are complementary to a target RNA, robust silencing was observed at 25 nM. 80% silencing was observed with only 16 nucleotide complementarity in the same format (see FIG. 47).


Incorporation of this property into the designs of siNAs of about 30 to 40 or more base pairs results in additional multifunctional siNA constructs. The example in FIG. 45 illustrates how a 30 base-pair duplex can target three distinct sequences after processing by Dicer-RNaseIII; these sequences can be on the same mRNA or separate RNAs, such as viral and host factor messages, or multiple points along a given pathway (e.g., inflammatory cascades). Furthermore, a 40 base-pair duplex can combine a bifunctional design in tandem, to provide a single duplex targeting four target sequences. An even more extensive approach can include use of homologous sequences (e.g. VEGFR-1/VEGFR-2) to enable five or six targets silenced for one multifunctional duplex. The example in FIG. 45 demonstrates how this can be achieved. A 30 base pair duplex is cleaved by Dicer into 22 and 8 base pair products from either end (8 b.p. fragments not shown). For ease of presentation the overhangs generated by dicer are not shown—but can be compensated for. Three targeting sequences are shown. The required sequence identity overlapped is indicated by grey boxes. The N's of the parent 30 b.p. siNA are suggested sites of 2′-OH positions to enable Dicer cleavage if this is tested in stabilized chemistries. Note that processing of a 30mer duplex by Dicer RNase III does not give a precise 22+8 cleavage, but rather produces a series of closely related products (with 22+8 being the primary site). Therefore, processing by Dicer will yield a series of active siNAs. Another non-limiting example is shown in FIG. 46. A 40 base pair duplex is cleaved by Dicer into 20 base pair products from either end. For ease of presentation the overhangs generated by dicer are not shown—but can be compensated for. Four targeting sequences are shown in four colors, blue, light-blue and red and orange. The required sequence identity overlapped is indicated by grey boxes. This design format can be extended to larger RNAs. If chemically stabilized siNAs are bound by Dicer, then strategically located ribonucleotide linkages can enable designer cleavage products that permit our more extensive repertoire of multiifunctional designs. For example cleavage products not limited to the Dicer standard of approximately 22-nucleotides can allow multifunctional siNA constructs with a target sequence identity overlap ranging from, for example, about 3 to about 15 nucleotides.


Another important aspect of this approach is its ability to restrict escape mutants. Processing to reveal an internal target site can ensure that escape mutations complementary to the eight nucleotides at the antisense 5′ end will not reduce siNA effectiveness. If about 17 nucleotidest of complementarity are required for RISC-mediated target cleavage, this will restrict, for example 8/17 or 47% of potential escape mutants.


Example 15
Indications

The present body of knowledge in HCV research indicates the need for methods to assay HCV activity and for compounds that can regulate HCV expression for research, diagnostic, and therapeutic use. As described herein, the nucleic acid molecules of the present invention can be used in assays to diagnose disease state related of HCV levels. In addition, the nucleic acid molecules can be used to treat disease state related to HCV levels.


Particular degenerative and disease states that can be associated with HCV expression modulation include, but are not limited to, HCV infection, liver failure, hepatocellular carcinoma, cirrhosis, and/or other disease states associated with HCV infection.


Example 16
Interferons

Interferons represent a non-limiting example of a class of compounds that can be used in conjuction with the siNA molecules of the invention for treating the diseases and/or conditions described herein. Type I interferons (IFN) are a class of natural cytokines that includes a family of greater than 25 IFN-α (Pesta, 1986, Methods Enzymol. 119, 3-14) as well as IFN-β, and IFN-co. Although evolutionarily derived from the same gene (Diaz et al., 1994, Genomics 22, 540-552), there are many differences in the primary sequence of these molecules, implying an evolutionary divergence in biologic activity. All type I IFN share a common pattern of biologic effects that begin with binding of the IFN to the cell surface receptor (Pfeffer & Strulovici, 1992, Transmembrane secondary messengers for IFN-α/β. In: Interferon. Principles and Medical Applications., S. Baron, D. H. Coopenhaver, F. Dianzani, W. R. Fleischmann Jr., T. K. Hughes Jr., G. R. Kimpel, D. W. Niesel, G. J. Stanton, and S. K. Tyring, eds. 151-160). Binding is followed by activation of tyrosine kinases, including the Janus tyrosine kinases and the STAT proteins, which leads to the production of several IFN-stimulated gene products (Johnson et al., 1994, Sci. Am. 270, 68-75). The IFN-stimulated gene products are responsible for the pleotropic biologic effects of type I IFN, including antiviral, antiproliferative, and immunomodulatory effects, cytokine induction, and HLA class I and class II regulation (Pestka et al., 1987, Annu. Rev. Biochem 56, 727). Examples of IFN-stimulated gene products include 2-5-oligoadenylate synthetase (2-5 OAS), β2-microglobulin, neopterin, p68 kinases, and the Mx protein (Chebath & Revel, 1992, The 2-5 A system: 2-5 A synthetase, isospecies and functions. In: Interferon. Principles and Medical Applications, S. Baron, D. H. Coopenhaver, F. Dianzani, W. R. Jr. Fleischmann, T. K. Jr Hughes, G. R. Kimpel, D. W. Niesel, G. J. Stanton, and S. K. Tyring, eds., pp. 225-236; Samuel, 1992, The RNA-dependent P1/eIF-2α protein kinase. In: Interferon. Principles and Medical Applications. S. Baron, D. H. Coopenhaver, F. Dianzani, W. R. Fleischmann Jr., T. K. Hughes Jr., G. R. Kimpel, D. W. Niesel, G. H. Stanton, and S. K. Tyring, eds. 237-250; Horisberger, 1992, MX protein: function and Mechanism of Action. In: Interferon. Principles and Medical Applications. S. Baron, D. H. Coopenhaver, F. Dianzani, W. R. Fleischmann Jr., T. K. Hughes Jr., G. R. Kimpel, D. W. Niesel, G. H. Stanton, and S. K. Tyring, eds. 215-224). Although all type I IFN have similar biologic effects, not all the activities are shared by each type I IFN, and in many cases, the extent of activity varies quite substantially for each IFN subtype (Fish et al, 1989, J Interferon Res. 9, 97-114; Ozes et al., 1992, J Interferon Res. 12, 55-59). More specifically, investigations into the properties of different subtypes of IFN-α and molecular hybrids of IFN-α have shown differences in pharmacologic properties (Rubinstein, 1987, J Interferon Res. 7, 545-551). These pharmacologic differences can arise from as few as three amino acid residue changes (Lee et al., 1982, Cancer Res. 42, 1312-1316).


Eighty-five to 166 amino acids are conserved in the known IFN-α subtypes. Excluding the IFN-α pseudogenes, there are approximately 25 known distinct IFN-α subtypes. Pairwise comparisons of these nonallelic subtypes show primary sequence differences ranging from 2% to 23%. In addition to the naturally occurring IFNs, a non-natural recombinant type I interferon known as consensus interferon (CIFN) has been synthesized as a therapeutic compound (Tong et al., 1997, Hepatology 26, 747-754).


Interferon is currently in use for at least 12 different indications, including infectious and autoimmune diseases and cancer (Borden, 1992, N. Engl. J. Med. 326, 1491-1492). For autoimmune diseases, IFN has been utilized for treatment of rheumatoid arthritis, multiple sclerosis, and Crohn's disease. For treatment of cancer, IFN has been used alone or in combination with a number of different compounds. Specific types of cancers for which IFN has been used include squamous cell carcinomas, melanomas, hypemephromas, hemangiomas, hairy cell leukemia, and Kaposi's sarcoma. In the treatment of infectious diseases, IFNs increase the phagocytic activity of macrophages and cytotoxicity of lymphocytes and inhibits the propagation of cellular pathogens. Specific indications for which IFN has been used as treatment include hepatitis B, human papillomavirus types 6 and 11 (i.e. genital warts) (Leventhal et al., 1991, N Engl J Med 325, 613-617), chronic granulomatous disease, and hepatitis C virus.


Numerous well controlled clinical trials using IFN-alpha in the treatment of chronic HCV infection have demonstrated that treatment three times a week results in lowering of serum ALT values in approximately 50% (range 40% to 70%) of patients by the end of 6 months of therapy (Davis et al., 1989, N. Engl. J. Med. 321, 1501-1506; Marcellin et al., 1991, Hepatology 13, 393-397; Tong et al., 1997, Hepatology 26, 747-754; Tong et al., Hepatology 26, 1640-1645). However, following cessation of interferon treatment, approximately 50% of the responding patients relapsed, resulting in a “durable” response rate as assessed by normalization of serum ALT concentrations of approximately 20 to 25%. In addition, studies that have examined six months of type 1 interferon therapy using changes in HCV RNA values as a clinical endpoint have demonstrated that up to 35% of patients will have a loss of HCV RNA by the end of therapy (Tong et al., 1997, supra). However, as with the ALT endpoint, about 50% of the patients relapse six months following cessation of therapy resulting in a durable virologic response of only 12% (23). Studies that have examined 48 weeks of therapy have demonstrated that the sustained virological response is up to 25%.


Pegylated interferons, i.e., interferons conjugated with polyethylene glycol (PEG), have demonstrated improved characteristics over interferon. Advantages incurred by PEG conjugation can include an improved pharmacokinetic profile compared to interferons lacking PEG, thus imparting more convenient dosing regimes, improved tolerance, and improved antiviral efficacy. Such improvements have been demonstrated in clinical studies of both polyethylene glycol interferon alfa-2a (PEGASYS, Roche) and polyethylene glycol interferon alfa-2b (VIRAFERON PEG, PEG-INTRON, Enzon/Schering Plough).


siNA molecules in combination with interferons and polyethylene glycol interferons have the potential to improve the effectiveness of treatment of HCV or any of the other indications discussed above. siNA molecules targeting RNAs associated with HCV infection can be used individually or in combination with other therapies such as interferons and polyethylene glycol interferons and to achieve enhanced efficacy.


Example 17
Diagnostic Uses

The siNA molecules of the invention can be used in a variety of diagnostic applications, such as in the identification of molecular targets (e.g., RNA) in a variety of applications, for example, in clinical, industrial, environmental, agricultural and/or research settings. Such diagnostic use of siNA molecules involves utilizing reconstituted RNAi systems, for example, using cellular lysates or partially purified cellular lysates. siNA molecules of this invention can be used as diagnostic tools to examine genetic drift and mutations within diseased cells or to detect the presence of endogenous or exogenous, for example viral, RNA in a cell. The close relationship between siNA activity and the structure of the target RNA allows the detection of mutations in any region of the molecule, which alters the base-pairing and three-dimensional structure of the target RNA. By using multiple siNA molecules described in this invention, one can map nucleotide changes, which are important to RNA structure and function in vitro, as well as in cells and tissues. Cleavage of target RNAs with siNA molecules can be used to inhibit gene expression and define the role of specified gene products in the progression of disease or infection. In this manner, other genetic targets can be defined as important mediators of the disease. These experiments will lead to better treatment of the disease progression by affording the possibility of combination therapies (e.g., multiple siNA molecules targeted to different genes, siNA molecules coupled with known small molecule inhibitors, or intermittent treatment with combinations siNA molecules and/or other chemical or biological molecules). Other in vitro uses of siNA molecules of this invention are well known in the art, and include detection of the presence of mRNAs associated with a disease, infection, or related condition. Such RNA is detected by determining the presence of a cleavage product after treatment with a siNA using standard methodologies, for example, fluorescence resonance emission transfer (FRET).


In a specific example, siNA molecules that cleave only wild-type or mutant forms of the target RNA are used for the assay. The first siNA molecules (i.e., those that cleave only wild-type forms of target RNA) are used to identify wild-type RNA present in the sample and the second siNA molecules (i.e., those that cleave only mutant forms of target RNA) are used to identify mutant RNA in the sample. As reaction controls, synthetic substrates of both wild-type and mutant RNA are cleaved by both siNA molecules to demonstrate the relative siNA efficiencies in the reactions and the absence of cleavage of the “non-targeted” RNA species. The cleavage products from the synthetic substrates also serve to generate size markers for the analysis of wild-type and mutant RNAs in the sample population. Thus, each analysis requires two siNA molecules, two substrates and one unknown sample, which is combined into six reactions. The presence of cleavage products is determined using an RNase protection assay so that full-length and cleavage fragments of each RNA can be analyzed in one lane of a polyacrylamide gel. It is not absolutely required to quantify the results to gain insight into the expression of mutant RNAs and putative risk of the desired phenotypic changes in target cells. The expression of mRNA whose protein product is implicated in the development of the phenotype (i.e., disease related or infection related) is adequate to establish risk. If probes of comparable specific activity are used for both transcripts, then a qualitative comparison of RNA levels is adequate and decreases the cost of the initial diagnosis. Higher mutant form to wild-type ratios are correlated with higher risk whether RNA levels are compared qualitatively or quantitatively.


All patents and publications mentioned in the specification are indicative of the levels of skill of those skilled in the art to which the invention pertains. All references cited in this disclosure are incorporated by reference to the same extent as if each reference had been incorporated by reference in its entirety individually.


One skilled in the art would readily appreciate that the present invention is well adapted to carry out the objects and obtain the ends and advantages mentioned, as well as those inherent therein. The methods and compositions described herein as presently representative of preferred embodiments are exemplary and are not intended as limitations on the scope of the invention. Changes therein and other uses will occur to those skilled in the art, which are encompassed within the spirit of the invention, are defined by the scope of the claims.


It will be readily apparent to one skilled in the art that varying substitutions and modifications can be made to the invention disclosed herein without departing from the scope and spirit of the invention. Thus, such additional embodiments are within the scope of the present invention and the following claims. The present invention teaches one skilled in the art to test various combinations and/or substitutions of chemical modifications described herein toward generating nucleic acid constructs with improved activity for mediating RNAi activity. Such improved activity can comprise improved stability, improved bioavailability, and/or improved activation of cellular responses mediating RNAi. Therefore, the specific embodiments described herein are not limiting and one skilled in the art can readily appreciate that specific combinations of the modifications described herein can be tested without undue experimentation toward identifying siNA molecules with improved RNAi activity.


The invention illustratively described herein suitably can be practiced in the absence of any element or elements, limitation or limitations that are not specifically disclosed herein. Thus, for example, in each instance herein any of the terms “comprising”, “consisting essentially of”, and “consisting of” may be replaced with either of the other two terms. The terms and expressions which have been employed are used as terms of description and not of limitation, and there is no intention that in the use of such terms and expressions of excluding any equivalents of the features shown and described or portions thereof, but it is recognized that various modifications are possible within the scope of the invention claimed. Thus, it should be understood that although the present invention has been specifically disclosed by preferred embodiments, optional features, modification and variation of the concepts herein disclosed may be resorted to by those skilled in the art, and that such modifications and variations are considered to be within the scope of this invention as defined by the description and the appended claims.


In addition, where features or aspects of the invention are described in terms of Markush groups or other grouping of alternatives, those skilled in the art will recognize that the invention is also thereby described in terms of any individual member or subgroup of members of the Markush group or other group.

TABLE IHCV Accession NumbersSeq NameAcc#LOCUSgi|329763|gb|M84754.1|HPCGENANTIM84754.1HPCGENANTIgi|567059|gb|U16362.1|HCU16362U16362.1HCU16362gi|5918956|gb|AF165059.1|AF165059AF165059.1AF165059gi|385583|gb|S62220.1|S62220S62220.1S62220gi|6010587|gb|AF177040.1|AF177040AF177040.1AF177040gi|5748510|emb|AJ238800.1|AJ238800.1HCJ238800HCJ238800gi|7650221|gb|AF207752.1|AF207752AF207752.1AF207752gi|11559454|dbj|AB049094.1|AB049094.1AB049094AB049094gi|3550760|dbj|D84263.1|D84263D84263.1D84263gi|221610|dbj|D90208.1|HPCJCGD90208.1HPCJCGgi|558520|dbj|D28917.1|HPCK3AD28917.1HPCK3Agi|2176577|dbj|E08461.1|E08461E08461.1E08461gi|6707285|gb|AF169005.1|AF169005AF169005.1AF169005gi|12309923|emb|AX057094.1|AX057094.1AX057094AX057094gi|6010585|gb|AF177039.1|AF177039AF177039.1AF177039gi|7329202|gb|AF238482.1|AF238482AF238482.1AF238482gi|11559464|dbj|AB049099.1|AB049099.1AB049099AB049099gi|5918932|gb|AF165047.1|AF165047AF165047.1AF165047gi|5918946|gb|AF165054.1|AF165054AF165054.1AF165054gi|7650233|gb|AF207758.1|AF207758AF207758.1AF207758gi|19568932|gb|AF483269.1|AF483269.1gi|7650247|gb|AF207765.1|AF207765AF207765.1AF207765gi|12309919|emb|AX057086.1|AX057086.1AX057086AX057086gi|5708597|dbj|E10839.1|E10839E10839.1E10839gi|2327074|gb|AF011753.1|AF011753AF011753.1AF011753gi|12310062|emb|AX057317.1|AX057317.1AX057317AX057317gi|221606|dbj|D10750.1|HPCJ491D10750.1HPCJ491gi|2174448|dbj|E06261.1|E06261E06261.1E06261gi|3098640|gb|AF054251.1|AF054251AF054251.1AF054251gi|18027684|gb|AF313916.1|AF313916AF313916.1AF313916gi|329873|gb|M62321.1|HPCPLYPREM62321.1HPCPLYPREgi|464177|dbj|D14853.1|HPCCGSD14853.1HPCCGSgi|15422182|gb|AY051292.1|AY051292.1gi|676877|dbj|D49374.1|HPCFGD49374.1HPCFGgi|1030706|dbj|D50480.1|HPCK1R1D50480.1HPCK1R1gi|7650223|gb|AF207753.1|AF207753AF207753.1AF207753gi|7650237|gb|AF207760.1|AF207760AF207760.1AF207760gi|11559444|dbj|AB049089.1|AB049089.1AB049089AB049089gi|3550762|dbj|D84264.1|D84264D84264.1D84264gi|12831192|gb|AF333324.1|AF333324AF333324.1AF333324gi|13122265|dbj|AB047641.1|AB04764 1.1AB047641AB047641gi|7329204|gb|AF238483.1|AF238483AF238483.1AF238483gi|11559468|dbj|AB049101.1|AB049101.1AB049101AB049101gi|5918934|gb|AF165048.1|AF165048AF165048.1AF165048gi|5918948|gb|AF165055.1|AF165055AF165055.1AF165055gi|7650235|gb|AF207759.1|AF207759AF207759.1AF207759gi|7650249|gb|AF207766.1|AF207766AF207766.1AF207766gi|9843676|emb|AJ278830.1|AJ278830.1HEC278830HEC278830gi|11559450|dbj|AB049092.1|AB049092.1AB049092AB049092gi|2943783|dbj|D89815.1 D89815D89815.1D89815gi|9626438|ref|NC_001433.1|NC_001433.1gi|12310134|emb|AX057395.1|AX057395.1AX057395AX057395gi|11559460|dbj|AB049097.1|AB049097.1AB049097AB049097gi|12309922|emb|AX057092.1|AX057092.1AX057092AX057092gi|2174644|dbj|E06457.1|E06457E06457.1E06457gi|2176559|dbj|E08443.1|E08443E08443.1E08443gi|5918960|gb|AF165061.1|AF165061AF165061.1AF165061gi|2326454|emb|Y12083.1|HCV12083Y12083.1HCV12083gi|5918938|gb|AF165050.1|AF165050AF165050.1AF165050gi|7650225|gb|AF207754.1|AF207754AF207754.1AF207754gi|7650261|gb|AF207772.1|AF207772AF207772.1AF207772gi|1030704|dbj|D50485.1|HPCK1S2D50485.1HPCK1S2gi|3550758|dbj|D84262.1|D84262D84262.1D84262gi|7650239|gb|AF207761.1|AF207761AF207761.1AF207761gi|3550764|dbj|D84265.1|D84265D84265.1D84265gi|7329206|gb|AF238484.1|AF238484AF238484.1AF238484gi|2176516|dbj|E08399.1|E08399E08399.1E08399gi|5918936|gb|AF165049.1|AF165049AF165049.1AF165049gi|11559446|dbj|AB049090.1|AB049090.1AB049090AB049090gi|5441837|emb|AJ242653.1|AJ242653.1SSE242653SSE242653gi|3098641|gb|AF054252.1|AF054252.1AF054252AF054252gi|4753720|emb|AJ132997.1|AJ132997.1HCV132997HCV132997gi|5420376|emb|AJ238799.1|AJ238799.1HCJ238799HCJ238799gi|11559440|dbj|AB049087.1|AB049087.1AB049087AB049087gi|15529110|gb|AY045702.1|AY045702.1gi|560788|dbj|D30613.1|HPCPPD30613.1HPCPPgi|11225869|emb|AX036253.1|AX036253.1AX036253AX036253gi|11559456|dbj|AB049095.1|AB049095.1AB049095AB049095gi|329770|gb|M58335.1|HPCHUMRM58335.1HPCHUMRgi|6707279|gb|AF169002.1|AF169002AF169002.1AF169002gi|221586|dbj|D10749.1|HPCHCJ1D10749.1HPCHCJ1gi|2171981|dbj|E03766.1|E03766E03766.1E03766gi|6010579|gb|AF177036.1|AF177036AF177036.1AF177036gi|1030703|dbj|D50484.1|HPCK1S3D50484.1HPCK1S3gi|3098650|gb|AF054257.1|AF054257AF054257.1AF054257gi|5821154|dbj|AB016785.1|AB016785AB016785.1AB016785gi|5918962|gb|AF165062.1|AF165062AF165062.1AF165062gi|7650227|gb|AF207755.1|AF207755AF207755.1AF207755gi|7650263|gb|AF207773.1|AF207773AF207773.1AF207773gi|1183030|dbj|D63822.1|HPCJK046E2D63822.1HPCJK046E2gi|13122271|dbj|AB047644.1|AB047644.1AB047644AB047644gi|2443428|gb|U89019.1|HCU89019U89019.1HCU89019gi|2462303|emb|Y13184.1|HCV1480Y13184.1HCV1480gi|7329208|gb|AF238485.1|AF238485AF238485.1AF238485gi|1160327|dbj|D14484.1|HPCJRNAD14484.1HPCJRNAgi|12309921|emb|AX057090.1|AX057090.1AX057090AX057090gi|3098643|gb|AF054253.1|AF054253AF054253.1AF054253gi|21397075|gb|AF511948.1|AF511948.1gi|1030701|dbj|D50482.1|HPCK1R3D50482.1HPCK1R3gi|1030702|dbj|D50483.1|HPCK1S1D50483.1HPCK1S1gi|3098632|gb|AF054247.1|AF054247AF054247.1AF054247gi|59478|emb|X61596.1|HCVJK1GX61596.1HCVJK1Ggi|3098652|gb|AF054258.1|AF054258AF054258.1AF054258gi|5918950|gb|AF165056.1|AF165056AF165056.1AF165056gi|7650251|gb|AF207767.1|AF207767AF207767.1AF207767gi|5918964|gb|AF165063.1|AF165063AF165063.1AF165063gi|5918928|gb|AF165045.1|AF165045AF165045.1AF165045gi|5532421|gb|AF139594.1|AF139594AF139594.1AF139594gi|13122267|dbj|AB047642.1|AB047642.1AB047642AB047642gi|5441831|emb|AJ242651.1|AJ242651.1SSE242651SSE242651gi|7650265|gb|AF207774.1|AF207774AF207774.1AF207774gi|7650229|gb|AF207756.1|AF207756AF207756.1AF207756gi|1183032|dbj|D63821.1|HPCJK049E1D63821.1HPCJK049E1gi|2175714|dbj|E07579.1|E07579E07579.1E07579gi|1212741|dbj|D45172.1|HPCHCPOD45172.1HPCHCPOgi|5708511|dbj|E05027.1|E05027E05027.1E05027gi|1483141|dbj|D50409.1|D50409D50409.1D50409gi|13122261|dbj|AB047639.1|AB047639.1AB047639AB047639gi|6521008|dbj|AB031663.1|AB031663AB031663.1AB031663gi|633201|emb|X76918.1|HCVCENS1X76918.1HCVCENS1gi|329737|gb|M67463.1|HPCCGAAM67463.1HPCCGAAgi|11559452|dbj|AB049093.1|AB049093.1AB049093AB049093gi|13619567|emb|AX100563.1|AX100563.1AX100563AX100563gi|221604|dbj|D13558.1|HPCJ483D13558.1HPCJ483gi|11225872|emb|AX036256.1|AX036256.1AX036256AX036256gi|1749761|dbj|D89872.1|D89872D89872.1D89872gi|5918940|gb|AF165051.1|AF165051AF165051.1AF165051gi|4753718|emb|AJ132996.1|AJ132996.1HCV132996HCV132996gi|7650241|gb|AF207762.1|AF207762AF207762.1AF207762gi|3098645|gb|AF054254.1|AF054254AF054254.1AF054254gi|9930556|gb|AF290978.1|AF290978AF290978.1AF290978gi|11559462|dbj|AB049098.1|AB049098.1AB049098AB049098gi|2764397|emb|AJ000009.1|AJ000009.1HCVPOLYPHCVPOLYPgi|221608|dbj|D10988.1|HPCJ8GD10988.1HPCJ8Ggi|3098634|gb|AF054248.1|AF054248AF054248.1AF054248gi|221650|dbj|D00944.1|HPCPOLPD00944.1HPCPOLPgi|306286|gb|M96362.1|HPCUNKCDSM96362.1HPCUNKCDSgi|3098654|gb|AF054259.1|AF054259AF054259.1AF054259gi|5918952|gb|AF165057.1|AF165057AF165057.1AF165057gi|7650253|gb|AF207768.1|AF207768AF207768.1AF207768gi|5918966|gb|AF165064.1|AF165064AF165064.1AF165064gi|15487693|gb|AF356827.1|AF356827AF356827.1AF356827gi|5738246|gb|AF176573.1|AF176573AF176573.1AF176573gi|11559448|dbj|AB049091.1|AB049091.1AB049091AB049091gi|21397077|gb|AF511950.1|AF511950.1gi|3098638|gb|AF054250.1|AF054250AF054250.1AF054250gi|6707281|gb|AF169003.1|AF169003AF169003.1AF169003gi|329739|gb|L02836.1|HPCCGENOML02836.1HPCCGENOMgi|6010581|gb|AF177037.1|AF177037AF177037.1AF177037gi|11559442|dbj|AB049088.1|AB049088.1AB049088AB049088gi|21397076|gb|AF511949.1|AF511949.1gi|1030705|dbj|D50481.1|HPCK1R2D50481.1HPCK1R2gi|2176384|dbj|E08264.1|E08264E08264.1E08264gi|3660725|gb|AF064490.1|AF064490AF064490.1AF064490gi|2252489|emb|Y11604.1|Y11604.1HCV4APOLYHCV4APOLYgi|5918942|gb|AF165052.1|AF165052AF165052.1AF165052gi|2895898|gb|AF046866.1|AF046866AF046866.1AF046866gi|7650243|gb|AF207763.1|AF207763AF207763.1AF207763gi|11559458|dbj|AB049096.1|AB049096.1AB049096AB049096gi|3122263|dbj|AB047640.1|AB047640AB047640.1AB047640gi|5708574|dbj|E08263.1|E08263E08263.1E08263gi|7650257|gb|AF207770.1|AF207770AF207770.1AF207770gi|3098647|gb|AF054255.1|AF054255AF054255.1AF054255gi|11559466|dbj|AB049100.1|AB049100.1AB049100AB049100gi|1181831|gb|U45476.1|HCU45476U45476.1HCU45476gi|2327070|gb|AF011751.1|AF011751AF011751.1AF011751gi|3098636|gb|AF054249.1|AF054249AF054249.1AF054249gi|7329210|gb|AF238486.1|AF238486AF238486.1AF238486gi|221612|dbj|D11168.1|HPCJTAD11168.1HPCJTAgi|960359|dbj|D63857.1|HPVHCVND63857.1HPVHCVNgi|13122273|dbj|AB047645.1|AB047645.1AB047645AB047645gi|5918954|gb|AF165058.1|AF165058AF165058.1AF165058gi|7650255|gb|AF207769.1|AF207769AF207769.1AF207769gi|437107|gb|U01214.1|HCU01214U01214.1HCU01214gi|471116|dbj|D10934.1|HPCRNAD10934.1HPCRNAgi|13026028|dbj|E66593.1 |E66593E66593.1E66593gi|2316097|gb|AF009606.1|AF009606AF009606.1AF009606gi|6707283|gb|AF169004.1|AF169004AF169004.1AF169004gi|514395|dbj|D17763.1|HPCEGSD17763.1HPCEGSgi|9757541|dbj|AB030907.1|AB030907AB030907.1AB030907gi|7329200|gb|AF238481.1|AF238481AF238481.1AF238481gi|6010583|gb|AF177038.1|AF177038AF177038.1AF177038gi|2172621|dbj|E04420.1|E04420E04420.1E04420gi|8926244|gb|AF271632.1|AF271632AF271632.1AF271632gi|5918930|gb|AF165046.1|AF165046AF165046.1AF165046gi|7650231|gb|AF207757.1|AF207757AF207757.1AF207757gi|5918944|gb|AF165053.1|AF165053AF165053.1AF165053gi|7650245|gb|AF207764.1|AF207764AF207764.1AF207764gi|12309920|emb|AX057088.1|AX057088.1AX057088AX057088gi|5918958|gb|AF165060.1|AF165060AF165060.1AF165060gi|7650259|gb|AF207771.1|AF207771AF207771.1AF207771gi|7341102|gb|AF208024.1|AF208024AF208024.1AF208024gi|3098649|gb|AF054256.1|AF054256AF054256.1AF054256gi|1944375|dbj|D85516.1|D85516D85516.1085516gi|2327072|gb|AF011752.1|AF011752AF011752.1AF011752gi|221614|dbj|D11355.1|HPCJTBD11355.1HPCJTBgi|13122269|dbj|AB047643.1|AB047643.1AB047643AB047643









TABLE II










HCV siNA AND TARGET SEQUENCES














Seq

Seq

Seq



Sequence
ID
Upper seq
ID
Lower seq
ID
















GCCCCGGGAGGUCUCGUAG
1
GCCCCGGGAGGUCUCGUAG
1
CUACGAGACCUCCCGGGGC
697






UGUGGUACUGCCUGAUAGG
2
UGUGGUACUGCCUGAUAGG
2
CCUAUCAGGCAGUACCACA
698





UUGUGGUACUGCCUGAUAG
3
UUGUGGUACUGCCUGAUAG
3
CUAUCAGGCAGUACCACAA
699





CCCCGGGAGGUCUCGUAGA
4
CCCCGGGAGGUCUCGUAGA
4
UCUACGAGACCUCCCGGGG
700





GUGGUACUGCCUGAUAGGG
5
GUGGUACUGCCUGAUAGGG
5
CCCUAUCAGGCAGUACCAC
701





CUGCCUGAUAGGGUGCUUG
6
CUGCCUGAUAGGGUGCUUG
6
CAAGCACCCUAUCAGGCAG
702





CCUUGUGGUACUGCCUGAU
7
CCUUGUGGUACUGCCUGAU
7
AUCAGGCAGUACCACAAGG
703





GCGAAAGGCCUUGUGGUAC
8
GCGAAAGGCCUUGUGGUAC
8
GUACCACAAGGCCUUUCGC
704





UACUGCCUGAUAGGGUGCU
9
UACUGCCUGAUAGGGUGCU
9
AGCACCCUAUCAGGCAGUA
705





GGUACUGCCUGAUAGGGUG
10
GGUACUGCCUGAUAGGGUG
10
CACCCUAUCAGGCAGUACC
706





AAAGGCCUUGUGGUACUGC
11
AAAGGCCUUGUGGUACUGC
11
GCAGUACCACAAGGCCUUU
707





AAGGCCUUGUGGUACUGCC
12
AAGGCCUUGUGGUACUGCC
12
GGCAGUACCACAAGGCCUU
708





CUUGUGGUACUGCCUGAUA
13
CUUGUGGUACUGCCUGAUA
13
UAUCAGGCAGUACCACAAG
709





AGGCCUUGUGGUACUGCCU
14
AGGCCUUGUGGUACUGCCU
14
AGGCAGUACCACAAGGCCU
710





GUACUGCCUGAUAGGGUGC
15
GUACUGCCUGAUAGGGUGC
15
GCACCCUAUCAGGCAGUAC
711





ACUGCCUGAUAGGGUGCUU
16
ACUGCCUGAUAGGGUGCUU
16
AAGCACCCUAUCAGGCAGU
712





CUUGCGAGUGCCCCGGGAG
17
CUUGCGAGUGCCCCGGGAG
17
CUCCCGGGGCACUCGCAAG
713





CUGAUAGGGUGCUUGCGAG
18
CUGAUAGGGUGCUUGCGAG
18
CUCGCAAGCACCCUAUCAG
714





UUGCGAGUGCCCCGGGAGG
19
UUGCGAGUGCCCCGGGAGG
19
CCUCCCGGGGCACUCGCAA
715





CCUGAUAGGGUGCUUGCGA
20
CCUGAUAGGGUGCUUGCGA
20
UCGCAAGCACCCUAUCAGG
716





GGCCUUGUGGUACUGCCUG
21
GGCCUUGUGGUACUGCCUG
21
CAGGCAGUACCACAAGGCC
717





GCUUGCGAGUGCCCCGGGA
22
GCUUGCGAGUGCCCCGGGA
22
UCCCGGGGCACUCGCAAGC
718





UGCCUGAUAGGGUGCUUGC
23
UGCCUGAUAGGGUGCUUGC
23
GCAAGCACCCUAUCAGGCA
719





GAAAGGCCUUGUGGUACUG
24
GAAAGGCCUUGUGGUACUG
24
CAGUACCACAAGGCCUUUC
720





GCCUGAUAGGGUGCUUGCG
25
GCCUGAUAGGGUGCUUGCG
25
CGCAAGCACCCUAUCAGGC
721





CGAAAGGCCUUGUGGUACU
26
CGAAAGGCCUUGUGGUACU
26
AGUACCACAAGGCCUUUCG
722





GCCUUGUGGUACUGCCUGA
27
GCCUUGUGGUACUGCCUGA
27
UCAGGCAGUACCACAAGGC
723





GAGUGCCCCGGGAGGUCUC
28
GAGUGCCCCGGGAGGUCUC
28
GAGACCUCCCGGGGCACUC
724





CCCGGGAGGUCUCGUAGAC
29
CCCGGGAGGUCUCGUAGAC
29
GUCUACGAGACCUCCCGGG
725





UGCGAGUGCCCCGGGAGGU
30
UGCGAGUGCCCCGGGAGGU
30
ACCUCCCGGGGCACUCGCA
726





UGGUACUGCCUGAUAGGGU
31
UGGUACUGCCUGAUAGGGU
31
ACCCUAUCAGGCAGUACCA
727





CCGGUGAGUACACCGGAAU
32
CCGGUGAGUACACCGGAAU
32
AUUCCGGUGUACUCACCGG
728





GCGAGUGCCCCGGGAGGUC
33
GCGAGUGCCCCGGGAGGUC
33
GACCUCCCGGGGCACUCGC
729





CGAGUGCCCCGGGAGGUCU
34
CGAGUGCCCCGGGAGGUCU
34
AGACCUCCCGGGGCACUCG
730





UGCCCCGGGAGGUCUCGUA
35
UGCCCCGGGAGGUCUCGUA
35
UACGAGACCUCCCGGGGCA
731





GUGCCCCGGGAGGUCUCGU
36
GUGCCCCGGGAGGUCUCGU
36
ACGAGACCUCCCGGGGCAC
732





AGUGCCCCGGGAGGUCUCG
37
AGUGCCCCGGGAGGUCUCG
37
CGAGACCUCCCGGGGCACU
733





CCGGGAGGUCUCGUAGACC
38
CCGGGAGGUCUCGUAGACC
38
GGUCUACGAGACCUCCCGG
734





UGAUAGGGUGCUUGCGAGU
39
UGAUAGGGUGCUUGCGAGU
39
ACUCGCAAGCACCCUAUCA
735





GUGCUUGCGAGUGCCCCGG
40
GUGCUUGCGAGUGCCCCGG
40
CCGGGGCACUCGCAAGCAC
736





AUAGGGUGCUUGCGAGUGC
41
AUAGGGUGCUUGCGAGUGC
41
GCACUCGCAAGCACCCUAU
737





GGGUGCUUGCGAGUGCCCC
42
GGGUGCUUGCGAGUGCCCC
42
GGGGCACUCGCAAGCACCC
738





CGGGAGGUCUCGUAGACCG
43
CGGGAGGUCUCGUAGACCG
43
CGGUCUACGAGACCUCCCG
739





GGGAGGUCUCGUAGACCGU
44
GGGAGGUCUCGUAGACCGU
44
ACGGUCUACGAGACCUCCC
740





GAUAGGGUGCUUGCGAGUG
45
GAUAGGGUGCUUGCGAGUG
45
CACUCGCAAGCACCCUAUC
741





GGAGGUCUCGUAGACCGUG
46
GGAGGUCUCGUAGACCGUG
46
CACGGUCUACGAGACCUCC
742





AGGGUGCUUGCGAGUGCCC
47
AGGGUGCUUGCGAGUGCCC
47
GGGCACUCGCAAGCACCCU
743





UGCUUGCGAGUGCCCCGGG
48
UGCUUGCGAGUGCCCCGGG
48
CCCGGGGCACUCGCAAGCA
744





GGUGCUUGCGAGUGCCCCG
49
GGUGCUUGCGAGUGCCCCG
49
CGGGGCACUCGCAAGCACC
745





UAGGGUGCUUGCGAGUGCC
50
UAGGGUGCUUGCGAGUGCC
50
GGCACUCGCAAGCACCCUA
746





AGGUCUCGUAGACCGUGCA
51
AGGUCUCGUAGACCGUGCA
51
UGCACGGUCUACGAGACCU
747





GAGGUCUCGUAGACCGUGC
52
GAGGUCUCGUAGACCGUGC
52
GCACGGUCUACGAGACCUC
748





GGAACCGGUGAGUACACCG
53
GGAACCGGUGAGUACACCG
53
CGGUGUACUCACCGGUUCC
749





CGGAACCGGUGAGUACACC
54
CGGAACCGGUGAGUACACC
54
GGUGUACUCACCGGUUCCG
750





CGGUGAGUACACCGGAAUU
55
CGGUGAGUACACCGGAAUU
55
AAUUCCGGUGUACUCACCG
751





GCGGAACCGGUGAGUACAC
56
GCGGAACCGGUGAGUACAC
56
GUGUACUCACCGGUUCCGC
752





AACCGGUGAGUACACCGGA
57
AACCGGUGAGUACACCGGA
57
UCCGGUGUACUCACCGGUU
753





ACCGGUGAGUACACCGGAA
58
ACCGGUGAGUACACCGGAA
58
UUCCGGUGUACUCACCGGU
754





CUGCGGAACCGGUGAGUAC
59
CUGCGGAACCGGUGAGUAC
59
GUACUCACCGGUUCCGCAG
755





GUCUGCGGAACCGGUGAGU
60
GUCUGCGGAACCGGUGAGU
60
ACUCACCGGUUCCGCAGAC
756





GAACCGGUGAGUACACCGG
61
GAACCGGUGAGUACACCGG
61
CCGGUGUACUCACCGGUUC
757





UGCGGAACCGGUGAGUACA
62
UGCGGAACCGGUGAGUACA
62
UGUACUCACCGGUUCCGCA
758





UCUGCGGAACCGGUGAGUA
63
UCUGCGGAACCGGUGAGUA
63
UACUCACCGGUUCCGCAGA
759





GGGAGAGCCAUAGUGGUCU
64
GGGAGAGCCAUAGUGGUCU
64
AGACCACUAUGGCUCUCCC
760





GUGGUCUGCGGAACCGGUG
65
GUGGUCUGCGGAACCGGUG
65
CACCGGUUCCGCAGACCAC
761





GGUCUGCGGAACCGGUGAG
66
GGUCUGCGGAACCGGUGAG
66
CUCACCGGUUCCGCAGACC
762





CGGGAGAGCCAUAGUGGUC
67
CGGGAGAGCCAUAGUGGUC
67
GACCACUAUGGCUCUCCCG
763





CCGGGAGAGCCAUAGUGGU
68
CCGGGAGAGCCAUAGUGGU
68
ACCACUAUGGCUCUCCCGG
764





UGGUCUGCGGAACCGGUGA
69
UGGUCUGCGGAACCGGUGA
69
UCACCGGUUCCGCAGACCA
765





GUGAGUACACCGGAAUUGC
70
GUGAGUACACCGGAAUUGC
70
GCAAUUCCGGUGUACUCAC
766





UGAGUACACCGGAAUUGCC
71
UGAGUACACCGGAAUUGCC
71
GGCAAUUCCGGUGUACUCA
767





GGUGAGUACACCGGAAUUG
72
GGUGAGUACACCGGAAUUG
72
CAAUUCCGGUGUACUCACC
768





GAGCCAUAGUGGUCUGCGG
73
GAGCCAUAGUGGUCUGCGG
73
CCGCAGACCACUAUGGCUC
769





AGAGCCAUAGUGGUCUGCG
74
AGAGCCAUAGUGGUCUGCG
74
CGCAGACCACUAUGGCUCU
770





UAGUGGUCUGCGGAACCGG
75
UAGUGGUCUGCGGAACCGG
75
CCGGUUCCGCAGACCACUA
771





AUAGUGGUCUGCGGAACCG
76
AUAGUGGUCUGCGGAACCG
76
CGGUUCCGCAGACCACUAU
772





GAGAGCCAUAGUGGUCUGC
77
GAGAGCCAUAGUGGUCUGC
77
GCAGACCACUAUGGCUCUC
773





GCCAUAGUGGUCUGCGGAA
78
GCCAUAGUGGUCUGCGGAA
78
UUCCGCAGACCACUAUGGC
774





AGUGGUCUGCGGAACCGGU
79
AGUGGUCUGCGGAACCGGU
79
ACCGGUUCCGCAGACCACU
775





CAUAGUGGUCUGCGGAACC
80
CAUAGUGGUCUGCGGAACC
80
GGUUCCGCAGACCACUAUG
776





AGCCAUAGUGGUCUGCGGA
81
AGCCAUAGUGGUCUGCGGA
81
UCCGCAGACCACUAUGGCU
777





CCAUAGUGGUCUGCGGAAC
82
CCAUAGUGGUCUGCGGAAC
82
GUUCCGCAGACCACUAUGG
778





CCCCUCCCGGGAGAGCCAU
83
CCCCUCCCGGGAGAGCCAU
83
AUGGCUCUCCCGGGAGGGG
779





GGAGAGCCAUAGUGGUCUG
84
GGAGAGCCAUAGUGGUCUG
84
CAGACCACUAUGGCUCUCC
780





CCCGGGAGAGCCAUAGUGG
85
CCCGGGAGAGCCAUAGUGG
85
CCACUAUGGCUCUCCCGGG
781





CCCCCUCCCGGGAGAGCCA
86
CCCCCUCCCGGGAGAGCCA
86
UGGCUCUCCCGGGAGGGGG
782





UCCCGGGAGAGCCAUAGUG
87
UCCCGGGAGAGCCAUAGUG
87
CACUAUGGCUCUCCCGGGA
783





CCCCCCUCCCGGGAGAGCC
88
CCCCCCUCCCGGGAGAGCC
88
GGCUCUCCCGGGAGGGGGG
784





CCCUCCCGGGAGAGCCAUA
89
CCCUCCCGGGAGAGCCAUA
89
UAUGGCUCUCCCGGGAGGG
785





CCUCCCGGGAGAGCCAUAG
90
CCUCCCGGGAGAGCCAUAG
90
CUAUGGCUCUCCCGGGAGG
786





CUCCCGGGAGAGCCAUAGU
91
CUCCCGGGAGAGCCAUAGU
91
ACUAUGGCUCUCCCGGGAG
787





UGUUGCCGCGCAGGGGCCC
92
UGUUGCCGCGCAGGGGCCC
92
GGGCCCCUGCGCGGCAACA
788





CCCCCCCUCCCGGGAGAGC
93
CCCCCCCUCCCGGGAGAGC
93
GCUCUCCCGGGAGGGGGGG
789





CAUGGCGUUAGUAUGAGUG
94
CAUGGCGUUAGUAUGAGUG
94
CACUCAUACUAACGCCAUG
790





UAGCCAUGGCGUUAGUAUG
95
UAGCCAUGGCGUUAGUAUG
95
CAUACUAACGCCAUGGCUA
791





AGCCAUGGCGUUAGUAUGA
96
AGCCAUGGCGUUAGUAUGA
96
UCAUACUAACGCCAUGGCU
792





CCAUGGCGUUAGUAUGAGU
97
CCAUGGCGUUAGUAUGAGU
97
ACUCAUACUAACGCCAUGG
793





AUGGCGUUAGUAUGAGUGU
98
AUGGCGUUAGUAUGAGUGU
98
ACACUCAUACUAACGCCAU
794





AAGCGUCUAGCCAUGGCGU
99
AAGCGUCUAGCCAUGGCGU
99
ACGCCAUGGCUAGACGCUU
795





GUCUAGCCAUGGCGUUAGU
100
GUCUAGCCAUGGCGUUAGU
100
ACUAACGCCAUGGCUAGAC
796





AAAGCGUCUAGCCAUGGCG
101
AAAGCGUCUAGCCAUGGCG
101
CGCCAUGGCUAGACGCUUU
797





GCGUCUAGCCAUGGCGUUA
102
GCGUCUAGCCAUGGCGUUA
102
UAACGCCAUGGCUAGACGC
798





GCCAUGGCGUUAGUAUGAG
103
GCCAUGGCGUUAGUAUGAG
103
CUCAUACUAACGCCAUGGC
799





AGCGUCUAGCCAUGGCGUU
104
AGCGUCUAGCCAUGGCGUU
104
AACGCCAUGGCUAGACGCU
800





CGUCUAGCCAUGGCGUUAG
105
CGUCUAGCCAUGGCGUUAG
105
CUAACGCCAUGGCUAGACG
801





UCUAGCCAUGGCGUUAGUA
106
UCUAGCCAUGGCGUUAGUA
106
UACUAACGCCAUGGCUAGA
802





GAAAGCGUCUAGCCAUGGC
107
GAAAGCGUCUAGCCAUGGC
107
GCCAUGGCUAGACGCUUUC
803





CUAGCCAUGGCGUUAGUAU
108
CUAGCCAUGGCGUUAGUAU
108
AUACUAACGCCAUGGCUAG
804





CACUCCCCUGUGAGGAACU
109
CACUCCCCUGUGAGGAACU
109
AGUUCCUCACAGGGGAGUG
805





ACCUCAAAGAAAAACCAAA
110
ACCUCAAAGAAAAACCAAA
110
UUUGGUUUUUCUUUGAGGU
806





CGCAGAAAGCGUCUAGCCA
111
CGCAGAAAGCGUCUAGCCA
111
UGGCUAGACGCUUUCUGCG
807





GGGUAAGGUCAUCGAUACC
112
GGGUAAGGUCAUCGAUACC
112
GGUAUCGAUGACCUUACCC
808





CAGAAAGCGUCUAGCCAUG
113
CAGAAAGCGUCUAGCCAUG
113
CAUGGCUAGACGCUUUCUG
809





AAACCUCAAAGAAAAACCA
114
AAACCUCAAAGAAAAACCA
114
UGGUUUUUCUUUGAGGUUU
810





GCAGAAAGCGUCUAGCCAU
115
GCAGAAAGCGUCUAGCCAU
115
AUGGCUAGACGCUUUCUGC
811





AGAAAGCGUCUAGCCAUGG
116
AGAAAGCGUCUAGCCAUGG
116
CCAUGGCUAGACGCUUUCU
812





ACGCAGAAAGCGUCUAGCC
117
ACGCAGAAAGCGUCUAGCC
117
GGCUAGACGCUUUCUGCGU
813





AACCUCAAAGAAAAACCAA
118
AACCUCAAAGAAAAACCAA
118
UUGGUUUUUCUUUGAGGUU
814





UGGGUAAGGUCAUCGAUAC
119
UGGGUAAGGUCAUCGAUAC
119
GUAUCGAUGACCUUACCCA
815





GUAAGGUCAUCGAUACCCU
120
GUAAGGUCAUCGAUACCCU
120
AGGGUAUCGAUGACCUUAC
816





UUCACGCAGAAAGCGUCUA
121
UUCACGCAGAAAGCGUCUA
121
UAGACGCUUUCUGCGUGAA
817





GGUAAGGUCAUCGAUACCC
122
GGUAAGGUCAUCGAUACCC
122
GGGUAUCGAUGACCUUACC
818





AUCACUCCCCUGUGAGGAA
123
AUCACUCCCCUGUGAGGAA
123
UUCCUCACAGGGGAGUGAU
819





UCACUCCCCUGUGAGGAAC
124
UCACUCCCCUGUGAGGAAC
124
GUUCCUCACAGGGGAGUGA
820





UGUCUUCACGCAGAAAGCG
125
UGUCUUCACGCAGAAAGCG
125
CGCUUUCUGCGUGAAGACA
821





UCACGCAGAAAGCGUCUAG
126
UCACGCAGAAAGCGUCUAG
126
CUAGACGCUUUCUGCGUGA
822





CACGCAGAAAGCGUCUAGC
127
CACGCAGAAAGCGUCUAGC
127
GCUAGACGCUUUCUGCGUG
823





GACCGGGUCCUUUCUUGGA
128
GACCGGGUCCUUUCUUGGA
128
UCCAAGAAAGGACCCGGUC
824





GAGGAACUACUGUCUUCAC
129
GAGGAACUACUGUCUUCAC
129
GUGAAGACAGUAGUUCCUC
825





CUGUGAGGAACUACUGUCU
130
CUGUGAGGAACUACUGUCU
130
AGACAGUAGUUCCUCACAG
826





GGAACUACUGUCUUCACGC
131
GGAACUACUGUCUUCACGC
131
GCGUGAAGACAGUAGUUCC
827





ACUCCCCUGUGAGGAACUA
132
ACUCCCCUGUGAGGAACUA
132
UAGUUCCUCACAGGGGAGU
828





GUCUUCACGCAGAAAGCGU
133
GUCUUCACGCAGAAAGCGU
133
ACGCUUUCUGCGUGAAGAC
829





AGGAACUACUGUCUUCACG
134
AGGAACUACUGUCUUCACG
134
CGUGAAGACAGUAGUUCCU
830





CCUGUGAGGAACUACUGUC
135
CCUGUGAGGAACUACUGUC
135
GACAGUAGUUCCUCACAGG
831





UGUGAGGAACUACUGUCUU
136
UGUGAGGAACUACUGUCUU
136
AAGACAGUAGUUCCUCACA
832





UCUUCACGCAGAAAGCGUC
137
UCUUCACGCAGAAAGCGUC
137
GACGCUUUCUGCGUGAAGA
833





GAACUACUGUCUUCACGCA
138
GAACUACUGUCUUCACGCA
138
UGCGUGAAGACAGUAGUUC
834





CCCUGUGAGGAACUACUGU
139
CCCUGUGAGGAACUACUGU
139
ACAGUAGUUCCUCACAGGG
835





CUUCACGCAGAAAGCGUCU
140
CUUCACGCAGAAAGCGUCU
140
AGACGCUUUCUGCGUGAAG
836





UGAGGAACUACUGUCUUCA
141
UGAGGAACUACUGUCUUCA
141
UGAAGACAGUAGUUCCUCA
837





UGGCGUUAGUAUGAGUGUC
142
UGGCGUUAGUAUGAGUGUC
142
GACACUCAUACUAACGCCA
838





CCCCUGUGAGGAACUACUG
143
CCCCUGUGAGGAACUACUG
143
CAGUAGUUCCUCACAGGGG
839





GUGAGGAACUACUGUCUUC
144
GUGAGGAACUACUGUCUUC
144
GAAGACAGUAGUUCCUCAC
840





GGCGUUAGUAUGAGUGUCG
145
GGCGUUAGUAUGAGUGUCG
145
CGACACUCAUACUAACGCC
841





GCCGAGUAGUGUUGGGUCG
146
GCCGAGUAGUGUUGGGUCG
146
CGACCCAACACUACUCGGC
842





ACUGUCUUCACGCAGAAAG
147
ACUGUCUUCACGCAGAAAG
147
CUUUCUGCGUGAAGACAGU
843





UGGGUCGCGAAAGGCCUUG
148
UGGGUCGCGAAAGGCCUUG
148
CAAGGCCUUUCGCGACCCA
844





CUACUGUCUUCACGCAGAA
149
CUACUGUCUUCACGCAGAA
149
UUCUGCGUGAAGACAGUAG
845





CGAGUAGUGUUGGGUCGCG
150
CGAGUAGUGUUGGGUCGCG
150
CGCGACCCAACACUACUCG
846





GUAGUGUUGGGUCGCGAAA
151
GUAGUGUUGGGUCGCGAAA
151
UUUCGCGACCCAACACUAC
847





UAAACCUCAAAGAAAAACC
152
UAAACCUCAAAGAAAAACC
152
GGUUUUUCUUUGAGGUUUA
848





CCGAGUAGUGUUGGGUCGC
153
CCGAGUAGUGUUGGGUCGC
153
GCGACCCAACACUACUCGG
849





AGCCGAGUAGUGUUGGGUC
154
AGCCGAGUAGUGUUGGGUC
154
GACCCAACACUACUCGGCU
850





GUCGCGAAAGGCCUUGUGG
155
GUCGCGAAAGGCCUUGUGG
155
CCACAAGGCCUUUCGCGAC
851





UAGUGUUGGGUCGCGAAAG
156
UAGUGUUGGGUCGCGAAAG
156
CUUUCGCGACCCAACACUA
852





CUAGCCGAGUAGUGUUGGG
157
CUAGCCGAGUAGUGUUGGG
157
CCCAACACUACUCGGCUAG
853





GAGUAGUGUUGGGUCGCGA
158
GAGUAGUGUUGGGUCGCGA
158
UCGCGACCCAACACUACUC
854





UCGCGAAAGGCCUUGUGGU
159
UCGCGAAAGGCCUUGUGGU
159
ACCACAAGGCCUUUCGCGA
855





GCGUUAGUAUGAGUGUCGU
160
GCGUUAGUAUGAGUGUCGU
160
ACGACACUCAUACUAACGC
856





UAGCCGAGUAGUGUUGGGU
161
UAGCCGAGUAGUGUUGGGU
161
ACCCAACACUACUCGGCUA
857





AACUACUGUCUUCACGCAG
162
AACUACUGUCUUCACGCAG
162
CUGCGUGAAGACAGUAGUU
858





CGCGAAAGGCCUUGUGGUA
163
CGCGAAAGGCCUUGUGGUA
163
UACCACAAGGCCUUUCGCG
859





AGUGUUGGGUCGCGAAAGG
164
AGUGUUGGGUCGCGAAAGG
164
CCUUUCGCGACCCAACACU
860





GUUGGGUCGCGAAAGGCCU
165
GUUGGGUCGCGAAAGGCCU
165
AGGCCUUUCGCGACCCAAC
861





AGUAGUGUUGGGUCGCGAA
166
AGUAGUGUUGGGUCGCGAA
166
UUCGCGACCCAACACUACU
862





UUGGGUCGCGAAAGGCCUU
167
UUGGGUCGCGAAAGGCCUU
167
AAGGCCUUUCGCGACCCAA
863





UCCCCUGUGAGGAACUACU
168
UCCCCUGUGAGGAACUACU
168
AGUAGUUCCUCACAGGGGA
864





UACUGUCUUCACGCAGAAA
169
UACUGUCUUCACGCAGAAA
169
UUUCUGCGUGAAGACAGUA
865





GUGUUGGGUCGCGAAAGGC
170
GUGUUGGGUCGCGAAAGGC
170
GCCUUUCGCGACCCAACAC
866





ACUACUGUCUUCACGCAGA
171
ACUACUGUCUUCACGCAGA
171
UCUGCGUGAAGACAGUAGU
867





CUGUCUUCACGCAGAAAGC
172
CUGUCUUCACGCAGAAAGC
172
GCUUUCUGCGUGAAGACAG
868





GGGUCGCGAAAGGCCUUGU
173
GGGUCGCGAAAGGCCUUGU
173
ACAAGGCCUUUCGCGACCC
869





CCUAAACCUCAAAGAAAAA
174
CCUAAACCUCAAAGAAAAA
174
UUUUUCUUUGAGGUUUAGG
870





GGUCGCGAAAGGCCUUGUG
175
GGUCGCGAAAGGCCUUGUG
175
CACAAGGCCUUUCGCGACC
871





CUAAACCUCAAAGAAAAAC
176
CUAAACCUCAAAGAAAAAC
176
GUUUUUCUUUGAGGUUUAG
872





UGUUGGGUCGCGAAAGGCC
177
UGUUGGGUCGCGAAAGGCC
177
GGCCUUUCGCGACCCAACA
873





CUCCCCUGUGAGGAACUAC
178
CUCCCCUGUGAGGAACUAC
178
GUAGUUCCUCACAGGGGAG
874





UCCUAAACCUCAAAGAAAA
179
UCCUAAACCUCAAAGAAAA
179
UUUUCUUUGAGGUUUAGGA
875





ACCGGGUCCUUUCUUGGAU
180
ACCGGGUCCUUUCUUGGAU
180
AUCCAAGAAAGGACCCGGU
876





AAUCCUAAACCUCAAAGAA
181
AAUCCUAAACCUCAAAGAA
181
UUCUUUGAGGUUUAGGAUU
877





UCAAUGCCUGGAGAUUUGG
182
UCAAUGCCUGGAGAUUUGG
182
CCAAAUCUCCAGGCAUUGA
878





AUGCCUGGAGAUUUGGGCG
183
AUGCCUGGAGAUUUGGGCG
183
CGCCCAAAUCUCCAGGCAU
879





AAUGCCUGGAGAUUUGGGC
184
AAUGCCUGGAGAUUUGGGC
184
GCCCAAAUCUCCAGGCAUU
880





CCGACCUCAUGGGGUACAU
185
CCGACCUCAUGGGGUACAU
185
AUGUACCCCAUGAGGUCGG
881





GCUCAAUGCCUGGAGAUUU
186
GCUCAAUGCCUGGAGAUUU
186
AAAUCUCCAGGCAUUGAGC
882





CUCAAUGCCUGGAGAUUUG
187
CUCAAUGCCUGGAGAUUUG
187
CAAAUCUCCAGGCAUUGAG
883





GCUAGCCGAGUAGUGUUGG
188
GCUAGCCGAGUAGUGUUGG
188
CCAACACUACUCGGCUAGC
884





CGCUCAAUGCCUGGAGAUU
189
CGCUCAAUGCCUGGAGAUU
189
AAUCUCCAGGCAUUGAGCG
885





CAAUGCCUGGAGAUUUGGG
190
CAAUGCCUGGAGAUUUGGG
190
CCCAAAUCUCCAGGCAUUG
886





GCCGACCUCAUGGGGUACA
191
GCCGACCUCAUGGGGUACA
191
UGUACCCCAUGAGGUCGGC
887





AUCCUAAACCUCAAAGAAA
192
AUCCUAAACCUCAAAGAAA
192
UUUCUUUGAGGUUUAGGAU
888





AGAUUUGGGCGUGCCCCCG
193
AGAUUUGGGCGUGCCCCCG
193
CGGGGGCACGCCCAAAUCU
889





CCCGCUCAAUGCCUGGAGA
194
CCCGCUCAAUGCCUGGAGA
194
UCUCCAGGCAUUGAGCGGG
890





GAGAUUUGGGCGUGCCCCC
195
GAGAUUUGGGCGUGCCCCC
195
GGGGGCACGCCCAAAUCUC
891





GGAGAUUUGGGCGUGCCCC
196
GGAGAUUUGGGCGUGCCCC
196
GGGGCACGCCCAAAUCUCC
892





GAUUUGGGCGUGCCCCCGC
197
GAUUUGGGCGUGCCCCCGC
197
GCGGGGGCACGCCCAAAUC
893





CCGCUCAAUGCCUGGAGAU
198
CCGCUCAAUGCCUGGAGAU
198
AUCUCCAGGCAUUGAGCGG
894





AGUACACCGGAAUUGCCAG
199
AGUACACCGGAAUUGCCAG
199
CUGGCAAUUCCGGUGUACU
895





UACACCGGAAUUGCCAGGA
200
UACACCGGAAUUGCCAGGA
200
UCCUGGCAAUUCCGGUGUA
896





GAGUACACCGGAAUUGCCA
201
GAGUACACCGGAAUUGCCA
201
UGGCAAUUCCGGUGUACUC
897





GUACACCGGAAUUGCCAGG
202
GUACACCGGAAUUGCCAGG
202
CCUGGCAAUUCCGGUGUAC
898





UUGCCGCGCAGGGGCCCCA
203
UUGCCGCGCAGGGGCCCCA
203
UGGGGCCCCUGCGCGGCAA
899





CUGGAGAUUUGGGCGUGCC
204
CUGGAGAUUUGGGCGUGCC
204
GGCACGCCCAAAUCUCCAG
900





GUUGCCGCGCAGGGGCCCC
205
GUUGCCGCGCAGGGGCCCC
205
GGGGCCCCUGCGCGGCAAC
901





GCCUGGAGAUUUGGGCGUG
206
GCCUGGAGAUUUGGGCGUG
206
CACGCCCAAAUCUCCAGGC
902





UGGAGAUUUGGGCGUGGCC
207
UGGAGAUUUGGGCGUGCCC
207
GGGCACGCCCAAAUCUCCA
903





CCUGGAGAUUUGGGCGUGC
208
CCUGGAGAUUUGGGCGUGC
208
GCACGCCCAAAUCUCCAGG
904





UGCUAGCCGAGUAGUGUUG
209
UGCUAGCCGAGUAGUGUUG
209
CAACACUACUCGGCUAGCA
905





UGCCUGGAGAUUUGGGCGU
210
UGCCUGGAGAUUUGGGCGU
210
ACGCCCAAAUCUCCAGGCA
906





CUGCUAGCCGAGUAGUGUU
211
CUGCUAGCCGAGUAGUGUU
211
AACACUACUCGGCUAGCAG
907





ACUGCUAGCCGAGUAGUGU
212
ACUGCUAGCCGAGUAGUGU
212
ACACUACUCGGCUAGCAGU
908





GACUGCUAGCCGAGUAGUG
213
GACUGCUAGCCGAGUAGUG
213
CACUACUCGGCUAGCAGUC
909





AGACUGCUAGCCGAGUAGU
214
AGACUGCUAGCCGAGUAGU
214
ACUACUCGGCUAGCAGUCU
910





ACCCGCUCAAUGCCUGGAG
215
ACCCGCUCAAUGCCUGGAG
215
CUCCAGGCAUUGAGCGGGU
911





AACCCGCUCAAUGCCUGGA
216
AACCCGCUCAAUGCCUGGA
216
UCCAGGCAUUGAGCGGGUU
912





UGCCGCGCAGGGGCCCCAG
217
UGCCGCGCAGGGGCCCCAG
217
CUGGGGCCCCUGCGCGGCA
913





AGGGGCCCCAGGUUGGGUG
218
AGGGGCCCCAGGUUGGGUG
218
CACCCAACCUGGGGCCCCU
914





GGGCCCCAGGUUGGGUGUG
219
GGGCCCCAGGUUGGGUGUG
219
CACACCCAACCUGGGGCCC
915





CAGGGGCCCCAGGUUGGGU
220
CAGGGGCCCCAGGUUGGGU
220
ACCCAACCUGGGGCCCCUG
916





GGCCCCAGGUUGGGUGUGC
221
GGCCCCAGGUUGGGUGUGC
221
GCACACCCAACCUGGGGCC
917





CGCAGGGGCCCCAGGUUGG
222
CGCAGGGGCCCCAGGUUGG
222
CCAACCUGGGGCCCCUGCG
918





UGGGCAGGAUGGCUCCUGU
223
UGGGCAGGAUGGCUCCUGU
223
ACAGGAGCCAUCCUGCCCA
919





GCCCCAGGUUGGGUGUGCG
224
GCCCCAGGUUGGGUGUGCG
224
CGCACACCCAACCUGGGGC
920





GCAGGGGCCCCAGGUUGGG
225
GCAGGGGCCCCAGGUUGGG
225
CCCAACCUGGGGCCCCUGC
921





GGGCAGGAUGGCUCCUGUC
226
GGGCAGGAUGGCUCCUGUC
226
GACAGGAGCCAUCCUGCCC
922





GGGGCCCCAGGUUGGGUGU
227
GGGGCCCCAGGUUGGGUGU
227
ACACCCAACCUGGGGCCCC
923





GCCGCGCAGGGGCCCCAGG
228
GCCGCGCAGGGGCCCCAGG
228
CCUGGGGCCCCUGCGCGGC
924





GCGCAGGGGCCCCAGGUUG
229
GCGCAGGGGCCCCAGGUUG
229
CAACCUGGGGCCCCUGCGC
925





CGCGCAGGGGCCCCAGGUU
230
CGCGCAGGGGCCCCAGGUU
230
AACCUGGGGCCCCUGCGCG
926





CCGCGCAGGGGCCCCAGGU
231
CCGCGCAGGGGCCCCAGGU
231
ACCUGGGGCCCCUGCGCGG
927





AGGACGACCGGGUCCUUUC
232
AGGACGACCGGGUCCUUUC
232
GAAAGGACCCGGUCGUCCU
928





CAGGACGACCGGGUCCUUU
233
CAGGACGACCGGGUCCUUU
233
AAAGGACCCGGUCGUCCUG
929





UGCCAGGACGACCGGGUCC
234
UGCCAGGACGACCGGGUCC
234
GGACCCGGUCGUCCUGGCA
930





AUUGCCAGGACGACCGGGU
235
AUUGCCAGGACGACCGGGU
235
ACCCGGUCGUCCUGGCAAU
931





AAUUGCCAGGACGACCGGG
236
AAUUGCCAGGACGACCGGG
236
CCCGGUCGUCCUGGCAAUU
932





UUGCCAGGACGACCGGGUC
237
UUGCCAGGACGACCGGGUC
237
GACCCGGUCGUCCUGGCAA
933





CCAGGACGACCGGGUCCUU
238
CCAGGACGACCGGGUCCUU
238
AAGGACCCGGUCGUCCUGG
934





GCCAGGACGACCGGGUCCU
239
GCCAGGACGACCGGGUCCU
239
AGGACCCGGUCGUCCUGGC
935





GAAUUGCCAGGACGACCGG
240
GAAUUGCCAGGACGACCGG
240
CCGGUCGUCCUGGCAAUUC
936





ACGACCGGGUCCUUUCUUG
241
ACGACCGGGUCCUUUCUUG
241
CAAGAAAGGACCCGGUCGU
937





GACGACCGGGUCCUUUCUU
242
GACGACCGGGUCCUUUCUU
242
AAGAAAGGACCCGGUCGUC
938





CGACCGGGUCCUUUCUUGG
243
CGACCGGGUCCUUUCUUGG
243
CCAAGAAAGGACCCGGUCG
939





GGACGACCGGGUCCUUUCU
244
GGACGACCGGGUCCUUUCU
244
AGAAAGGACCCGGUCGUCC
940





CCGGAAUUGCCAGGACGAC
245
CCGGAAUUGCCAGGACGAC
245
GUCGUCCUGGCAAUUCCGG
941





ACACCGGAAUUGCCAGGAC
246
ACACCGGAAUUGCCAGGAC
246
GUCCUGGCAAUUCCGGUGU
942





ACCGGAAUUGCCAGGACGA
247
ACCGGAAUUGCCAGGACGA
247
UCGUCCUGGCAAUUCCGGU
943





CGGAAUUGCCAGGACGACC
248
CGGAAUUGCCAGGACGACC
248
GGUCGUCCUGGCAAUUCCG
944





GGAAUUGCCAGGACGACCG
249
GGAAUUGCCAGGACGACCG
249
CGGUCGUCCUGGCAAUUCC
945





CACCGGAAUUGCCAGGACG
250
CACCGGAAUUGCCAGGACG
250
CGUCCUGGCAAUUCCGGUG
946





CCCCAGGUUGGGUGUGCGC
251
CCCCAGGUUGGGUGUGCGC
251
GCGCACACCCAACCUGGGG
947





GAUCGUUGGUGGAGUUUAC
252
GAUCGUUGGUGGAGUUUAC
252
GUAAACUCCACCAACGAUC
948





CAGAUCGUUGGUGGAGUUU
253
CAGAUCGUUGGUGGAGUUU
253
AAACUCCACCAACGAUCUG
949





AGAUCGUUGGUGGAGUUUA
254
AGAUCGUUGGUGGAGUUUA
254
UAAACUCCACCAACGAUCU
950





CCCAGGUUGGGUGUGCGCG
255
CCCAGGUUGGGUGUGCGCG
255
CGCGCACACCCAACCUGGG
951





CCAGGUUGGGUGUGCGCGC
256
CCAGGUUGGGUGUGCGCGC
256
GCGCGCACACCCAACCUGG
952





AGGUUGGGUGUGCGCGCGA
257
AGGUUGGGUGUGCGCGCGA
257
UCGCGCGCACACCCAACCU
953





CAGGUUGGGUGUGCGCGCG
258
CAGGUUGGGUGUGCGCGCG
258
CGCGCGCACACCCAACCUG
954





GGUUGGGUGUGCGCGCGAC
259
GGUUGGGUGUGCGCGCGAC
259
GUCGCGCGCACACCCAACC
955





GAAAAACCAAACGUAACAC
260
GAAAAACCAAACGUAACAC
260
GUGUUACGUUUGGUUUUUC
956





AGAAAAACCAAACGUAACA
261
AGAAAAACCAAACGUAACA
261
UGUUACGUUUGGUUUUUCU
957





AACCAAACGUAACACCAAC
262
AACCAAACGUAACACCAAC
262
GUUGGUGUUACGUUUGGUU
958





AAAGAAAAACCAAACGUAA
263
AAAGAAAAACCAAACGUAA
263
UUACGUUUGGUUUUUCUUU
959





AAAAACCAAACGUAACACC
264
AAAAACCAAACGUAACACC
264
GGUGUUACGUUUGGUUUUU
960





AAGAAAAACCAAACGUAAC
265
AAGAAAAACCAAACGUAAC
265
GUUACGUUUGGUUUUUCUU
961





CAAAGAAAAACCAAACGUA
266
CAAAGAAAAACCAAACGUA
266
UACGUUUGGUUUUUCUUUG
962





ACCCCCGGCGUAGGUCGCG
267
ACCCCCGGCGUAGGUCGCG
267
CGCGACCUACGCCGGGGGU
963





GACCCCCGGCGUAGGUCGC
268
GACCCCCGGCGUAGGUCGC
268
GCGACCUACGCCGGGGGUC
964





CGUUAGUAUGAGUGUCGUG
269
CGUUAGUAUGAGUGUCGUG
269
CACGACACUCAUACUAACG
965





GUUAGUAUGAGUGUCGUGC
270
GUUAGUAUGAGUGUCGUGC
270
GCACGACACUCAUACUAAC
966





UUAGUAUGAGUGUCGUGCA
271
UUAGUAUGAGUGUCGUGCA
271
UGCACGACACUCAUACUAA
967





CCAAACGUAACACCAACCG
272
CCAAACGUAACACCAACCG
272
CGGUUGGUGUUACGUUUGG
968





ACCAAACGUAACACCAACC
273
ACCAAACGUAACACCAACC
273
GGUUGGUGUUACGUUUGGU
969





UUGGGCGUGCCCCCGCGAG
274
UUGGGCGUGCCCCCGCGAG
274
CUCGCGGGGGCACGCCCAA
970





AUUUGGGCGUGCCCCCGCG
275
AUUUGGGCGUGCCCCCGCG
275
CGCGGGGGCACGCCCAAAU
971





UUUGGGCGUGCCCCCGCGA
276
UUUGGGCGUGCCCCCGCGA
276
UCGCGGGGGCACGCCCAAA
972





AAACCAAACGUAACACCAA
277
AAACCAAACGUAACACCAA
277
UUGGUGUUACGUUUGGUUU
973





UGGGCGUGCCCCCGCGAGA
278
UGGGCGUGCCCCCGCGAGA
278
UCUCGCGGGGGCACGCCCA
974





GUCAGAUCGUUGGUGGAGU
279
GUCAGAUCGUUGGUGGAGU
279
ACUCCACCAACGAUCUGAC
975





GUGUCGUGCAGCCUCCAGG
280
GUGUCGUGCAGCCUCCAGG
280
CCUGGAGGCUGCACGACAC
976





GGUCAGAUCGUUGGUGGAG
281
GGUCAGAUCGUUGGUGGAG
281
CUCCACCAACGAUCUGACC
977





AGUGUCGUGCAGCCUCCAG
282
AGUGUCGUGCAGCCUCCAG
282
CUGGAGGCUGCACGACACU
978





GAGUGUCGUGCAGCCUCCA
283
GAGUGUCGUGCAGCCUCCA
283
UGGAGGCUGCACGACACUC
979





UCGUAGACCGUGCACCAUG
284
UCGUAGACCGUGCACCAUG
284
CAUGGUGCACGGUCUACGA
980





GACCGUGCACCAUGAGCAC
285
GACCGUGCACCAUGAGCAC
285
GUGCUCAUGGUGCACGGUC
981





AGUAUGAGUGUCGUGCAGC
286
AGUAUGAGUGUCGUGCAGC
286
GCUGCACGACACUCAUACU
982





UAGUAUGAGUGUCGUGCAG
287
UAGUAUGAGUGUCGUGCAG
287
CUGCACGACACUCAUACUA
983





UCAGAUCGUUGGUGGAGUU
288
UCAGAUCGUUGGUGGAGUU
288
AACUCCACCAACGAUCUGA
984





AGACCGUGCACCAUGAGCA
289
AGACCGUGCACCAUGAGCA
289
UGCUCAUGGUGCACGGUCU
985





AAAACCAAACGUAACACCA
290
AAAACCAAACGUAACACCA
290
UGGUGUUACGUUUGGUUUU
986





GUAGACCGUGCACCAUGAG
291
GUAGACCGUGCACCAUGAG
291
CUCAUGGUGCACGGUCUAC
987





CUCGUAGACCGUGCACCAU
292
CUCGUAGACCGUGCACCAU
292
AUGGUGCACGGUCUACGAG
988





CGUAGACCGUGCACCAUGA
293
CGUAGACCGUGCACCAUGA
293
UCAUGGUGCACGGUCUACG
989





CCUGGGCUCAGCCCGGGUA
294
CCUGGGCUCAGCCCGGGUA
294
UACCCGGGCUGAGCCCAGG
990





UAGACCGUGCACCAUGAGC
295
UAGACCGUGCACCAUGAGC
295
GCUCAUGGUGCACGGUCUA
991





GGUCUCGUAGACCGUGCAC
296
GGUCUCGUAGACCGUGCAC
296
GUGCACGGUCUACGAGACC
992





UCUCGUAGACCGUGCACCA
297
UCUCGUAGACCGUGCACCA
297
UGGUGCACGGUCUACGAGA
993





GUCUCGUAGACCGUGCACC
298
GUCUCGUAGACCGUGCACC
298
GGUGCACGGUCUACGAGAC
994





UUGGGUAAGGUCAUCGAUA
299
UUGGGUAAGGUCAUCGAUA
299
UAUCGAUGACCUUACCCAA
995





UCGCCGACCUCAUGGGGUA
300
UCGCCGACCUCAUGGGGUA
300
UACCCCAUGAGGUCGGCGA
996





CCUCAAAGAAAAACCAAAC
301
CCUCAAAGAAAAACCAAAC
301
GUUUGGUUUUUCUUUGAGG
997





GGGCGUGCCCCCGCGAGAC
302
GGGCGUGCCCCCGCGAGAC
302
GUCUCGCGGGGGCACGCCC
998





GGAUGAACCGGCUGAUAGC
303
GGAUGAACCGGCUGAUAGC
303
GCUAUCAGCCGGUUCAUCC
999





UGGAUGAACCGGCUGAUAG
304
UGGAUGAACCGGCUGAUAG
304
CUAUCAGCCGGUUCAUCCA
1000





CUCAAAGAAAAACCAAACG
305
CUCAAAGAAAAACCAAACG
305
CGUUUGGUUUUUCUUUGAG
1001





AGGAAGACUUCCGAGCGGU
306
AGGAAGACUUCCGAGCGGU
306
ACCGCUCGGAAGUCUUCCU
1002





UCAAAGAAAAACCAAACGU
307
UCAAAGAAAAACCAAACGU
307
ACGUUUGGUUUUUCUUUGA
1003





GGAAGACUUCCGAGCGGUC
308
GGAAGACUUCCGAGCGGUC
308
GACCGCUCGGAAGUCUUCC
1004





CGCCGACCUCAUGGGGUAC
309
CGCCGACCUCAUGGGGUAC
309
GUACCCCAUGAGGUCGGCG
1005





CUUCCGAGCGGUCGCAACC
310
CUUCCGAGCGGUCGCAACC
310
GGUUGCGACCGCUCGGAAG
1006





GGCGUGCCCCCGCGAGACU
311
GGCGUGCCCCCGCGAGACU
311
AGUCUCGCGGGGGCACGCC
1007





UAUGAGUGUCGUGCAGCCU
312
UAUGAGUGUCGUGCAGCCU
312
AGGCUGCACGACACUCAUA
1008





UGCCCCCGCGAGACUGCUA
313
UGCCCCCGCGAGACUGCUA
313
UAGCAGUCUCGCGGGGGCA
1009





CGAGACUGCUAGCCGAGUA
314
CGAGACUGCUAGCCGAGUA
314
UACUCGGCUAGCAGUCUCG
1010





UGAGUGUCGUGCAGCCUCC
315
UGAGUGUCGUGCAGCCUCC
315
GGAGGCUGCACGACACUCA
1011





GCCCCCGCGAGACUGCUAG
316
GCCCCCGCGAGACUGCUAG
316
CUAGCAGUCUCGCGGGGGC
1012





GAGACUGCUAGCCGAGUAG
317
GAGACUGCUAGCCGAGUAG
317
CUACUCGGCUAGCAGUCUC
1013





CCCCCGCGAGACUGCUAGC
318
CCCCCGCGAGACUGCUAGC
318
GCUAGCAGUCUCGCGGGGG
1014





CGCGAGACUGCUAGCCGAG
319
CGCGAGACUGCUAGCCGAG
319
CUCGGCUAGCAGUCUCGCG
1015





GUAUGAGUGUCGUGCAGCC
320
GUAUGAGUGUCGUGCAGCC
320
GGCUGCACGACACUCAUAC
1016





AUGAGUGUCGUGCAGCCUC
321
AUGAGUGUCGUGCAGCCUC
321
GAGGCUGCACGACACUCAU
1017





GCGAGACUGCUAGCCGAGU
322
GCGAGACUGCUAGCCGAGU
322
ACUCGGCUAGCAGUCUCGC
1018





CCCCGCGAGACUGCUAGCC
323
CCCCGCGAGACUGCUAGCC
323
GGCUAGCAGUCUCGCGGGG
1019





CCGCGAGACUGCUAGCCGA
324
CCGCGAGACUGCUAGCCGA
324
UCGGCUAGCAGUCUCGCGG
1020





CCCGCGAGACUGCUAGCCG
325
CCCGCGAGACUGCUAGCCG
325
CGGCUAGCAGUCUCGCGGG
1021





GCGUGCCCCCGCGAGACUG
326
GCGUGCCCCCGCGAGACUG
326
CAGUCUCGCGGGGGCACGC
1022





GACCCCCCCUCCCGGGAGA
327
GACCCCCCCUCCCGGGAGA
327
UCUCCCGGGAGGGGGGGUC
1023





CGGGUCCUUUCUUGGAUCA
328
CGGGUCCUUUCUUGGAUCA
328
UGAUCCAAGAAAGGACCCG
1024





GUGCCCCCGCGAGACUGCU
329
GUGCCCCCGCGAGACUGCU
329
AGCAGUCUCGCGGGGGCAC
1025





CGUGCCCCCGCGAGACUGC
330
CGUGCCCCCGCGAGACUGC
330
GCAGUCUCGCGGGGGCACG
1026





UUCGCCGACCUCAUGGGGU
331
UUCGCCGACCUCAUGGGGU
331
ACCCCAUGAGGUCGGCGAA
1027





CGCCCACAGGACGUCAAGU
332
CGCCCACAGGACGUCAAGU
332
ACUUGACGUCCUGUGGGCG
1028





GCCCACAGGACGUCAAGUU
333
GCCCACAGGACGUCAAGUU
333
AACUUGACGUCCUGUGGGC
1029





ACCCCCCCUCCCGGGAGAG
334
ACCCCCCCUCCCGGGAGAG
334
CUCUCCCGGGAGGGGGGGU
1030





GGACCCCCCCUCCCGGGAG
335
GGACCCCCCCUCCCGGGAG
335
CUCCCGGGAGGGGGGGUCC
1031





CCGGGUCCUUUCUUGGAUC
336
CCGGGUCCUUUCUUGGAUC
336
GAUCCAAGAAAGGACCCGG
1032





CAGGACCCCCCCUCCCGGG
337
CAGGACCCCCCCUCCCGGG
337
CCCGGGAGGGGGGGUCCUG
1033





AGGACGUCAAGUUCCCGGG
338
AGGACGUCAAGUUCCCGGG
338
CCCGGGAACUUGACGUCCU
1034





AGGACCCCCCCUCCCGGGA
339
AGGACCCCCCCUCCCGGGA
339
UCCCGGGAGGGGGGGUCCU
1035





CCACAGGACGUCAAGUUCC
340
CCACAGGACGUCAAGUUCC
340
GGAACUUGACGUCCUGUGG
1036





CAGGACGUCAAGUUCCCGG
341
CAGGACGUCAAGUUCCCGG
341
CCGGGAACUUGACGUCCUG
1037





ACAGGACGUCAAGUUCCCG
342
ACAGGACGUCAAGUUCCCG
342
CGGGAACUUGACGUCCUGU
1038





CACAGGACGUCAAGUUCCC
343
CACAGGACGUCAAGUUCCC
343
GGGAACUUGACGUCCUGUG
1039





CAGUGGAUGAACCGGCUGA
344
CAGUGGAUGAACCGGCUGA
344
UCAGCCGGUUCAUCCACUG
1040





GGGCUCAGCCCGGGUACCC
345
GGGCUCAGCCCGGGUACCC
345
GGGUACCCGGGCUGAGCCC
1041





CCGAGCGGUCGCAACCUCG
346
CCGAGCGGUCGCAACCUCG
346
CGAGGUUGCGACCGCUCGG
1042





CUGGGCUCAGCCCGGGUAC
347
CUGGGCUCAGCCCGGGUAC
347
GUACCCGGGCUGAGCCCAG
1043





AGUGGAUGAACCGGCUGAU
348
AGUGGAUGAACCGGCUGAU
348
AUCAGCCGGUUCAUCCACU
1044





UCCGAGCGGUCGCAACCUC
349
UCCGAGCGGUCGCAACCUC
349
GAGGUUGCGACCGCUCGGA
1045





UGGGCUCAGCCCGGGUACC
350
UGGGCUCAGCCCGGGUACC
350
GGUACCCGGGCUGAGCCCA
1046





GGUACCCUUGGCCCCUCUA
351
GGUACCCUUGGCCCCUCUA
351
UAGAGGGGCCAAGGGUACC
1047





UUCCGAGCGGUCGCAACCU
352
UUCCGAGCGGUCGCAACCU
352
AGGUUGCGACCGCUCGGAA
1048





GGGUACCCUUGGCCCCUCU
353
GGGUACCCUUGGCCCCUCU
353
AGAGGGGCCAAGGGUACCC
1049





GGGUCCUUUCUUGGAUCAA
354
GGGUCCUUUCUUGGAUCAA
354
UUGAUCCAAGAAAGGACCC
1050





CCCACAGGACGUCAAGUUC
355
CCCACAGGACGUCAAGUUC
355
GAACUUGACGUCCUGUGGG
1051





GGUUGCUCUUUCUCUAUCU
356
GGUUGCUCUUUCUCUAUCU
356
AGAUAGAGAAAGAGCAACC
1052





GUGGGCAGGAUGGCUCCUG
357
GUGGGCAGGAUGGCUCCUG
357
CAGGAGCCAUCCUGCCCAC
1053





GGUGGGCAGGAUGGCUCCU
358
GGUGGGCAGGAUGGCUCCU
358
AGGAGCCAUCCUGCCCACC
1054





GUUGCUCUUUCUCUAUCUU
359
GUUGCUCUUUCUCUAUCUU
359
AAGAUAGAGAAAGAGCAAC
1055





GUGGAUGAACCGGCUGAUA
360
GUGGAUGAACCGGCUGAUA
360
UAUCAGCCGGUUCAUCCAC
1056





CCAGGACCCCCCCUCCCGG
361
CCAGGACCCCCCCUCCCGG
361
CCGGGAGGGGGGGUCCUGG
1057





GGGUGGGCAGGAUGGCUCC
362
GGGUGGGCAGGAUGGCUCC
362
GGAGCCAUCCUGCCCACCC
1058





CUUCACGGAGGCUAUGACU
363
CUUCACGGAGGCUAUGACU
363
AGUCAUAGCCUCCGUGAAG
1059





ACCGCCGCCCACAGGACGU
364
ACCGCCGCCCACAGGACGU
364
ACGUCCUGUGGGCGGCGGU
1060





UCCAGGACCCCCCCUCCCG
365
UCCAGGACCCCCCCUCCCG
365
CGGGAGGGGGGGUCCUGGA
1061





AUAUGAUGAUGAACUGGUC
366
AUAUGAUGAUGAACUGGUC
366
GACCAGUUCAUCAUCAUAU
1062





UUCACGGAGGCUAUGACUA
367
UUCACGGAGGCUAUGACUA
367
UAGUCAUAGCCUCCGUGAA
1063





UCACGGAGGCUAUGACUAG
368
UCACGGAGGCUAUGACUAG
368
CUAGUCAUAGCCUCCGUGA
1064





AUGAACCGGCUGAUAGCGU
369
AUGAACCGGCUGAUAGCGU
369
ACGCUAUCAGCCGGUUCAU
1065





GGGAUAUGAUGAUGAACUG
370
GGGAUAUGAUGAUGAACUG
370
CAGUUCAUCAUCAUAUCCC
1066





UGCAGUGGAUGAACCGGCU
371
UGCAGUGGAUGAACCGGCU
371
AGCCGGUUCAUCCACUGCA
1067





GUGCAGUGGAUGAACCGGC
372
GUGCAGUGGAUGAACCGGC
372
GCCGGUUCAUCCACUGCAC
1068





UGAACCGGCUGAUAGCGUU
373
UGAACCGGCUGAUAGCGUU
373
AACGCUAUCAGCCGGUUCA
1069





GGAUAUGAUGAUGAACUGG
374
GGAUAUGAUGAUGAACUGG
374
CCAGUUCAUCAUCAUAUCC
1070





GCUCUUUCUCUAUCUUCCU
375
GCUCUUUCUCUAUCUUCCU
375
AGGAAGAUAGAGAAAGAGC
1071





GGGGGCGACACUCCACCAU
376
GGGGGCGACACUCCACCAU
376
AUGGUGGAGUGUCGCCCCC
1072





GAUGAACCGGCUGAUAGCG
377
GAUGAACCGGCUGAUAGCG
377
CGCUAUCAGCCGGUUCAUC
1073





GAUAUGAUGAUGAACUGGU
378
GAUAUGAUGAUGAACUGGU
378
ACCAGUUCAUCAUCAUAUC
1074





UGGGAUAUGAUGAUGAACU
379
UGGGAUAUGAUGAUGAACU
379
AGUUCAUCAUCAUAUCCCA
1075





UUGCUCUUUCUCUAUCUUC
380
UUGCUCUUUCUCUAUCUUC
380
GAAGAUAGAGAAAGAGCAA
1076





UGGGGGCGACACUCCACCA
381
UGGGGGCGACACUCCACCA
381
UGGUGGAGUGUCGCCCCCA
1077





UGCUCUUUCUCUAUCUUCC
382
UGCUCUUUCUCUAUCUUCC
382
GGAAGAUAGAGAAAGAGCA
1078





GGUCCUUUCUUGGAUCAAC
383
GGUCCUUUCUUGGAUCAAC
383
GUUGAUCCAAGAAAGGACC
1079





AAGACUUCCGAGCGGUCGC
384
AAGACUUCCGAGCGGUCGC
384
GCGACCGCUCGGAAGUCUU
1080





AGCCCGGGUACCCUUGGCC
385
AGCCCGGGUACCCUUGGCC
385
GGCCAAGGGUACCCGGGCU
1081





UUUCUUGGAUCAACCCGCU
386
UUUCUUGGAUCAACCCGCU
386
AGCGGGUUGAUCCAAGAAA
1082





CAGCCCGGGUACCCUUGGC
387
CAGCCCGGGUACCCUUGGC
387
GCCAAGGGUACCCGGGCUG
1083





AGACUUCCGAGCGGUCGCA
388
AGACUUCCGAGCGGUCGCA
388
UGCGACCGCUCGGAAGUCU
1084





UUCUUGGAUCAACCCGCUC
389
UUCUUGGAUCAACCCGCUC
389
GAGCGGGUUGAUCCAAGAA
1085





CCCGGGUACCCUUGGCCCC
390
CCCGGGUACCCUUGGCCCC
390
GGGGCCAAGGGUACCCGGG
1086





GUCCUUUCUUGGAUCAACC
391
GUCCUUUCUUGGAUCAACC
391
GGUUGAUCCAAGAAAGGAC
1087





CUUUCUUGGAUCAACCCGC
392
CUUUCUUGGAUCAACCCGC
392
GCGGGUUGAUCCAAGAAAG
1088





CCUUUCUUGGAUCAACCCG
393
CCUUUCUUGGAUCAACCCG
393
CGGGUUGAUCCAAGAAAGG
1089





UCCUUUCUUGGAUCAACCC
394
UCCUUUCUUGGAUCAACCC
394
GGGUUGAUCCAAGAAAGGA
1090





AAGUUCCCGGGCGGUGGUC
395
AAGUUCCCGGGCGGUGGUC
395
GACCACCGCCCGGGAACUU
1091





GCAGUGGAUGAACCGGCUG
396
GCAGUGGAUGAACCGGCUG
396
CAGCCGGUUCAUCCACUGC
1092





CCGGGUACCCUUGGCCCCU
397
CCGGGUACCCUUGGCCCCU
397
AGGGGCCAAGGGUACCCGG
1093





AGUUCCCGGGCGGUGGUCA
398
AGUUCCCGGGCGGUGGUCA
398
UGACCACCGCCCGGGAACU
1094





CUUGGAUCAACCCGCUCAA
399
CUUGGAUCAACCCGCUCAA
399
UUGAGCGGGUUGAUCCAAG
1095





GGAUCAACCCGCUCAAUGC
400
GGAUCAACCCGCUCAAUGC
400
GCAUUGAGCGGGUUGAUCC
1096





ACUUCCGAGCGGUCGCAAC
401
ACUUCCGAGCGGUCGCAAC
401
GUUGCGACCGCUCGGAAGU
1097





UCUUGGAUCAACCCGCUCA
402
UCUUGGAUCAACCCGCUCA
402
UGAGCGGGUUGAUCCAAGA
1098





UUGGAUCAACCCGCUCAAU
403
UUGGAUCAACCCGCUCAAU
403
AUUGAGCGGGUUGAUCCAA
1099





AACCGCCGCCCACAGGACG
404
AACCGCCGCCCACAGGACG
404
CGUCCUGUGGGCGGCGGUU
1100





GCGUGAACUAUGCAACAGG
405
GCGUGAACUAUGCAACAGG
405
CCUGUUGCAUAGUUCACGC
1101





AUCAACCCGCUCAAUGCCU
406
AUCAACCCGCUCAAUGCCU
406
AGGCAUUGAGCGGGUUGAU
1102





GAUCAACCCGCUCAAUGCC
407
GAUCAACCCGCUCAAUGCC
407
GGCAUUGAGCGGGUUGAUC
1103





CAACCCGCUCAAUGCCUGG
408
CAACCCGCUCAAUGCCUGG
408
CCAGGCAUUGAGCGGGUUG
1104





GCUUCGCCGACCUCAUGGG
409
GCUUCGCCGACCUCAUGGG
409
CCCAUGAGGUCGGCGAAGC
1105





GACUUCCGAGCGGUCGCAA
410
GACUUCCGAGCGGUCGCAA
410
UUGCGACCGCUCGGAAGUC
1106





UCAACCCGCUCAAUGCCUG
411
UCAACCCGCUCAAUGCCUG
411
CAGGCAUUGAGCGGGUUGA
1107





GGCUUCGCCGACCUCAUGG
412
GGCUUCGCCGACCUCAUGG
412
CCAUGAGGUCGGCGAAGCC
1108





UGGAUCAACCCGCUCAAUG
413
UGGAUCAACCCGCUCAAUG
413
CAUUGAGCGGGUUGAUCCA
1109





CGGGCGGUGGUCAGAUCGU
414
CGGGCGGUGGUCAGAUCGU
414
ACGAUCUGACCACCGCCCG
1110





CUUGGCCCCUCUAUGGCAA
415
CUUGGCCCCUCUAUGGCAA
415
UUGCCAUAGAGGGGCCAAG
1111





CCGGGCGGUGGUCAGAUCG
416
CCGGGCGGUGGUCAGAUCG
416
CGAUCUGACCACCGCCCGG
1112





UGGGGUGGGCAGGAUGGCU
417
UGGGGUGGGCAGGAUGGCU
417
AGCCAUCCUGCCCACCCCA
1113





GGAGUUUACCUGUUGCCGC
418
GGAGUUUACCUGUUGCCGC
418
GCGGCAACAGGUAAACUCC
1114





CCUUGGCCCCUCUAUGGCA
419
CCUUGGCCCCUCUAUGGCA
419
UGCCAUAGAGGGGCCAAGG
1115





GUGGAGUUUACCUGUUGCC
420
GUGGAGUUUACCUGUUGCC
420
GGCAACAGGUAAACUCCAC
1116





GGUGGAGUUUACCUGUUGC
421
GGUGGAGUUUACCUGUUGC
421
GCAACAGGUAAACUCCACC
1117





UUCCCGGGCGGUGGUCAGA
422
UUCCCGGGCGGUGGUCAGA
422
UCUGACCACCGCCCGGGAA
1118





UGAACUAUGCAACAGGGAA
423
UGAACUAUGCAACAGGGAA
423
UUCCCUGUUGCAUAGUUCA
1119





AGUUUACCUGUUGCCGCGC
424
AGUUUACCUGUUGCCGCGC
424
GCGCGGCAACAGGUAAACU
1120





GUGAACUAUGCAACAGGGA
425
GUGAACUAUGCAACAGGGA
425
UCCCUGUUGCAUAGUUCAC
1121





UUACCUGUUGCCGCGCAGG
426
UUACCUGUUGCCGCGCAGG
426
CCUGCGCGGCAACAGGUAA
1122





UCCCGGGCGGUGGUCAGAU
427
UCCCGGGCGGUGGUCAGAU
427
AUCUGACCACCGCCCGGGA
1123





GUUCCCGGGCGGUGGUCAG
428
GUUCCCGGGCGGUGGUCAG
428
CUGACCACCGCCCGGGAAC
1124





GCCCGGGUACCCUUGGCCC
429
GCCCGGGUACCCUUGGCCC
429
GGGCCAAGGGUACCCGGGC
1125





AAGGAGAUGAAGGCGAAGG
430
AAGGAGAUGAAGGCGAAGG
430
CCUUCGCCUUCAUCUCCUU
1126





AGGAGAUGAAGGCGAAGGC
431
AGGAGAUGAAGGCGAAGGC
431
GCCUUCGCCUUCAUCUCCU
1127





GUUUACCUGUUGCCGCGCA
432
GUUUACCUGUUGCCGCGCA
432
UGCGCGGCAACAGGUAAAC
1128





CUGUUGCCGCGCAGGGGCC
433
CUGUUGCCGCGCAGGGGCC
433
GGCCCCUGCGCGGCAACAG
1129





AACACCAACCGCCGCCCAC
434
AACACCAACCGCCGCCCAC
434
GUGGGCGGCGGUUGGUGUU
1130





GAGUUUACCUGUUGCCGCG
435
GAGUUUACCUGUUGCCGCG
435
CGCGGCAACAGGUAAACUC
1131





UUUACCUGUUGCCGCGCAG
436
UUUACCUGUUGCCGCGCAG
436
CUGCGCGGCAACAGGUAAA
1132





GGGGUGGGCAGGAUGGCUC
437
GGGGUGGGCAGGAUGGCUC
437
GAGCCAUCCUGCCCACCCC
1133





GAAGACUUCCGAGCGGUCG
438
GAAGACUUCCGAGCGGUCG
438
CGACCGCUCGGAAGUCUUC
1134





ACCUGUUGCCGCGCAGGGG
439
ACCUGUUGCCGCGCAGGGG
439
CCCCUGCGCGGCAACAGGU
1135





UACCUGUUGCCGCGCAGGG
440
UACCUGUUGCCGCGCAGGG
440
CCCUGCGCGGCAACAGGUA
1136





UACCUCUUCAACUGGGCAG
441
UACCUCUUCAACUGGGCAG
441
CUGCCCAGUUGAAGAGGUA
1137





CGUGAACUAUGCAACAGGG
442
CGUGAACUAUGCAACAGGG
442
CCCUGUUGCAUAGUUCACG
1138





ACACCAACCGCCGCCCACA
443
ACACCAACCGCCGCCCACA
443
UGUGGGCGGCGGUUGGUGU
1139





CCCGGGCGGUGGUCAGAUC
444
CCCGGGCGGUGGUCAGAUC
444
GAUCUGACCACCGCCCGGG
1140





ACCUCUUCAACUGGGCAGU
445
ACCUCUUCAACUGGGCAGU
445
ACUGCCCAGUUGAAGAGGU
1141





CUUCGCCGACCUCAUGGGG
446
CUUCGCCGACCUCAUGGGG
446
CCCCAUGAGGUCGGCGAAG
1142





CCUGUUGCCGCGCAGGGGC
447
CCUGUUGCCGCGCAGGGGC
447
GCCCCUGCGCGGCAACAGG
1143





CCAACCGCCGCCCACAGGA
448
CCAACCGCCGCCCACAGGA
448
UCCUGUGGGCGGCGGUUGG
1144





ACCAACCGCCGCCCACAGG
449
ACCAACCGCCGCCCACAGG
449
CCUGUGGGCGGCGGUUGGU
1145





UGGAGUUUACCUGUUGCCG
450
UGGAGUUUACCUGUUGCCG
450
CGGCAACAGGUAAACUCCA
1146





CACCAACCGCCGCCCACAG
451
CACCAACCGCCGCCCACAG
451
CUGUGGGCGGCGGUUGGUG
1147





CAAACGUAACACCAACCGC
452
CAAACGUAACACCAACCGC
452
GCGGUUGGUGUUACGUUUG
1148





CAAGCGGAGACGGCUGGAG
453
CAAGCGGAGACGGCUGGAG
453
CUCCAGCCGUCUCCGCUUG
1149





ACGGAGGCUAUGACUAGGU
454
ACGGAGGCUAUGACUAGGU
454
ACCUAGUCAUAGCCUCCGU
1150





UAACACCAACCGCCGCCCA
455
UAACACCAACCGCCGCCCA
455
UGGGCGGCGGUUGGUGUUA
1151





AUCGUUGGUGGAGUUUACC
456
AUCGUUGGUGGAGUUUACC
456
GGUAAACUCCACCAACGAU
1152





GGGAGACAUAUAUCACAGC
457
GGGAGACAUAUAUCACAGC
457
GCUGUGAUAUAUGUCUCCC
1153





AACCUCGUGGAAGGCGACA
458
AACCUCGUGGAAGGCGACA
458
UGUCGCCUUCCACGAGGUU
1154





GGGGGAGACAUAUAUCACA
459
GGGGGAGACAUAUAUCACA
459
UGUGAUAUAUGUCUCCCCC
1155





AACGUAACACCAACCGCCG
460
AACGUAACACCAACCGCCG
460
CGGCGGUUGGUGUUACGUU
1156





AAACGUAACACCAACCGCC
461
AAACGUAACACCAACCGCC
461
GGCGGUUGGUGUUACGUUU
1157





GGGGAGACAUAUAUCACAG
462
GGGGAGACAUAUAUCACAG
462
CUGUGAUAUAUGUCUCCCC
1158





GAGAUGAAGGCGAAGGCGU
463
GAGAUGAAGGCGAAGGCGU
463
ACGCCUUCGCCUUCAUCUC
1159





AAGCGGAGACGGCUGGAGC
464
AAGCGGAGACGGCUGGAGC
464
GCUCCAGCCGUCUCCGCUU
1160





GUACCCUUGGCCCCUCUAU
465
GUACCCUUGGCCCCUCUAU
465
AUAGAGGGGCCAAGGGUAC
1161





CCUCCAGGACCCCCCCUCC
466
CCUCCAGGACCCCCCCUCC
466
GGAGGGGGGGUCCUGGAGG
1162





CUCCAGGACCCCCCCUCCC
467
CUCCAGGACCCCCCCUCCC
467
GGGAGGGGGGGUCCUGGAG
1163





UACCCUUGGCCCCUCUAUG
468
UACCCUUGGCCCCUCUAUG
468
CAUAGAGGGGCCAAGGGUA
1164





CAACCUCGUGGAAGGCGAC
469
CAACCUCGUGGAAGGCGAC
469
GUCGCCUUCCACGAGGUUG
1165





CGGAGGCUAUGACUAGGUA
470
CGGAGGCUAUGACUAGGUA
470
UACCUAGUCAUAGCCUCCG
1166





GGAGAUGAAGGCGAAGGCG
471
GGAGAUGAAGGCGAAGGCG
471
CGCCUUCGCCUUCAUCUCC
1167





AGAUGAAGGCGAAGGCGUC
472
AGAUGAAGGCGAAGGCGUC
472
GACGCCUUCGCCUUCAUCU
1168





GUAACACCAACCGCCGCCC
473
GUAACACCAACCGCCGCCC
473
GGGCGGCGGUUGGUGUUAC
1169





CGUAACACCAACCGCCGCC
474
CGUAACACCAACCGCCGCC
474
GGCGGCGGUUGGUGUUACG
1170





ACGUAACACCAACCGCCGC
475
ACGUAACACCAACCGCCGC
475
GCGGCGGUUGGUGUUACGU
1171





CACGGAGGCUAUGACUAGG
476
CACGGAGGCUAUGACUAGG
476
CCUAGUCAUAGCCUCCGUG
1172





GUUGGUGGAGUUUACCUGU
477
GUUGGUGGAGUUUACCUGU
477
ACAGGUAAACUCCACCAAC
1173





CGUUGGUGGAGUUUACCUG
478
CGUUGGUGGAGUUUACCUG
478
CAGGUAAACUCCACCAACG
1174





ACCCUUGGCCCCUCUAUGG
479
ACCCUUGGCCCCUCUAUGG
479
CCAUAGAGGGGCCAAGGGU
1175





UUGGUGGAGUUUACCUGUU
480
UUGGUGGAGUUUACCUGUU
480
AACAGGUAAACUCCACCAA
1176





UGGUGGAGUUUACCUGUUG
481
UGGUGGAGUUUACCUGUUG
481
CAACAGGUAAACUCCACCA
1177





UCGUUGGUGGAGUUUACCU
482
UCGUUGGUGGAGUUUACCU
482
AGGUAAACUCCACCAACGA
1178





CGGGUACCCUUGGCCCCUC
483
CGGGUACCCUUGGCCCCUC
483
GAGGGGCCAAGGGUACCCG
1179





GGCUCAGCCCGGGUACCCU
484
GGCUCAGCCCGGGUACCCU
484
AGGGUACCCGGGCUGAGCC
1180





GAUCACUCCCCUGUGAGGA
485
GAUCACUCCCCUGUGAGGA
485
UCCUCACAGGGGAGUGAUC
1181





GGUGGUCAGAUCGUUGGUG
486
GGUGGUCAGAUCGUUGGUG
486
CACCAACGAUCUGACCACC
1182





GAUGAAGGCGAAGGCGUCC
487
GAUGAAGGCGAAGGCGUCC
487
GGACGCCUUCGCCUUCAUC
1183





AGGAUGGCUCCUGUCACCC
488
AGGAUGGCUCCUGUCACCC
488
GGGUGACAGGAGCCAUCCU
1184





CUCAGCCCGGGUACCCUUG
489
CUCAGCCCGGGUACCCUUG
489
CAAGGGUACCCGGGCUGAG
1185





UCAGCCCGGGUACCCUUGG
490
UCAGCCCGGGUACCCUUGG
490
CCAAGGGUACCCGGGCUGA
1186





AUGAAGGCGAAGGCGUCCA
491
AUGAAGGCGAAGGCGUCCA
491
UGGACGCCUUCGCCUUCAU
1187





CGGGGGAGACAUAUAUCAC
492
CGGGGGAGACAUAUAUCAC
492
GUGAUAUAUGUCUCCCCCG
1188





CAGGAUGGCUCCUGUCACC
493
CAGGAUGGCUCCUGUCACC
493
GGUGACAGGAGCCAUCCUG
1189





UGAAGGCGAAGGCGUCCAC
494
UGAAGGCGAAGGCGUCCAC
494
GUGGACGCCUUCGCCUUCA
1190





UGGUCAGAUCGUUGGUGGA
495
UGGUCAGAUCGUUGGUGGA
495
UCCACCAACGAUCUGACCA
1191





GCUCAGCCCGGGUACCCUU
496
GCUCAGCCCGGGUACCCUU
496
AAGGGUACCCGGGCUGAGC
1192





GUGGUCAGAUCGUUGGUGG
497
GUGGUCAGAUCGUUGGUGG
497
CCACCAACGAUCUGACCAC
1193





CAGCCUCCAGGACCCCCCC
498
CAGCCUCCAGGACCCCCCC
498
GGGGGGGUCCUGGAGGCUG
1194





GGCGGUGGUCAGAUCGUUG
499
GGCGGUGGUCAGAUCGUUG
499
CAACGAUCUGACCACCGCC
1195





GCCUCCAGGACCCCCCCUC
500
GCCUCCAGGACCCCCCCUC
500
GAGGGGGGGUCCUGGAGGC
1196





AACCGGCUGAUAGCGUUCG
501
AACCGGCUGAUAGCGUUCG
501
CGAACGCUAUCAGCCGGUU
1197





AGCCUCCAGGACCCCCCCU
502
AGCCUCCAGGACCCCCCCU
502
AGGGGGGGUCCUGGAGGCU
1198





CGGCUUCGCCGACCUCAUG
503
CGGCUUCGCCGACCUCAUG
503
CAUGAGGUCGGCGAAGCCG
1199





GCGGAGACGGCUGGAGCGC
504
GCGGAGACGGCUGGAGCGC
504
GCGCUCCAGCCGUCUCCGC
1200





UCAUGGGGUACAUUCCGCU
505
UCAUGGGGUACAUUCCGCU
505
AGCGGAAUGUACCCCAUGA
1201





GAACCGGCUGAUAGCGUUC
506
GAACCGGCUGAUAGCGUUC
506
GAACGCUAUCAGCCGGUUC
1202





GCGGUGGUCAGAUCGUUGG
507
GCGGUGGUCAGAUCGUUGG
507
CCAACGAUCUGACCACCGC
1203





GGCAGGAUGGCUCCUGUCA
508
GGCAGGAUGGCUCCUGUCA
508
UGACAGGAGCCAUCCUGCC
1204





GCAGGAUGGCUCCUGUCAC
509
GCAGGAUGGCUCCUGUCAC
509
GUGACAGGAGCCAUCCUGC
1205





AUUUGGGUAAGGUCAUCGA
510
AUUUGGGUAAGGUCAUCGA
510
UCGAUGACCUUACCCAAAU
1206





ACCGGCUGAUAGCGUUCGC
511
ACCGGCUGAUAGCGUUCGC
511
GCGAACGCUAUCAGCCGGU
1207





CGGAGACGGCUGGAGCGCG
512
CGGAGACGGCUGGAGCGCG
512
CGCGCUCCAGCCGUCUCCG
1208





GCGGCUUCGCCGACCUCAU
513
GCGGCUUCGCCGACCUCAU
513
AUGAGGUCGGCGAAGCCGC
1209





AAUUUGGGUAAGGUCAUCG
514
AAUUUGGGUAAGGUCAUCG
514
CGAUGACCUUACCCAAAUU
1210





GGGCGGUGGUCAGAUCGUU
515
GGGCGGUGGUCAGAUCGUU
515
AACGAUCUGACCACCGCCC
1211





CAACCGCCGCCCACAGGAC
516
CAACCGCCGCCCACAGGAC
516
GUCCUGUGGGCGGCGGUUG
1212





UGCGGCUUCGCCGACCUCA
517
UGCGGCUUCGCCGACCUCA
517
UGAGGUCGGCGAAGCCGCA
1213





CGGUGGUCAGAUCGUUGGU
518
CGGUGGUCAGAUCGUUGGU
518
ACCAACGAUCUGACCACCG
1214





UUGGGUGUGCGCGCGACUA
519
UUGGGUGUGCGCGCGACUA
519
UAGUCGCGCGCACACCCAA
1215





GUGUGCGCGCGACUAGGAA
520
GUGUGCGCGCGACUAGGAA
520
UUCCUAGUCGCGCGCACAC
1216





GAUGGCUCCUGUCACCCCG
521
GAUGGCUCCUGUCACCCCG
521
CGGGGUGACAGGAGCCAUC
1217





GGAUGGCUCCUGUCACCCC
522
GGAUGGCUCCUGUCACCCC
522
GGGGUGACAGGAGCCAUCC
1218





UGUGCGCGCGACUAGGAAG
523
UGUGCGCGCGACUAGGAAG
523
CUUCCUAGUCGCGCGCACA
1219





UGGGUGUGCGCGCGACUAG
524
UGGGUGUGCGCGCGACUAG
524
CUAGUCGCGCGCACACCCA
1220





GGUGUGCGCGCGACUAGGA
525
GGUGUGCGCGCGACUAGGA
525
UCCUAGUCGCGCGCACACC
1221





GGGUGUGCGCGCGACUAGG
526
GGGUGUGCGCGCGACUAGG
526
CCUAGUCGCGCGCACACCC
1222





CCCCGGCGUAGGUCGCGUA
527
CCCCGGCGUAGGUCGCGUA
527
UACGCGACCUACGCCGGGG
1223





GAAGGCGACAACCUAUCCC
528
GAAGGCGACAACCUAUCCC
528
GGGAUAGGUUGUCGCCUUC
1224





CCCGGCGUAGGUCGCGUAA
529
CCCGGCGUAGGUCGCGUAA
529
UUACGCGACCUACGCCGGG
1225





AGCGGAGACGGCUGGAGCG
530
AGCGGAGACGGCUGGAGCG
530
CGCUCCAGCCGUCUCCGCU
1226





CCCCCGGCGUAGGUCGCGU
531
CCCCCGGCGUAGGUCGCGU
531
ACGCGACCUACGCCGGGGG
1227





AGGCGAAGGCGUCCACAGU
532
AGGCGAAGGCGUCCACAGU
532
ACUGUGGACGCCUUCGCCU
1228





AAGGCGAAGGCGUCCACAG
533
AAGGCGAAGGCGUCCACAG
533
CUGUGGACGCCUUCGCCUU
1229





GUUGGGUGUGCGCGCGACU
534
GUUGGGUGUGCGCGCGACU
534
AGUCGCGCGCACACCCAAC
1230





CUCAUGGGGUACAUUCCGC
535
CUCAUGGGGUACAUUCCGC
535
GCGGAAUGUACCCCAUGAG
1231





GGAAGGCGACAACCUAUCC
536
GGAAGGCGACAACCUAUCC
536
GGAUAGGUUGUCGCCUUCC
1232





GCAAGUUCCUUGCCGACGG
537
GCAAGUUCCUUGCCGACGG
537
CCGUCGGCAAGGAACUUGC
1233





UGCAGCCUCCAGGACCCCC
538
UGCAGCCUCCAGGACCCCC
538
GGGGGUCCUGGAGGCUGCA
1234





GGACUGCACGAUGCUCGUG
539
GGACUGCACGAUGCUCGUG
539
CACGAGCAUCGUGCAGUCC
1235





GAAGGCGAAGGCGUCCACA
540
GAAGGCGAAGGCGUCCACA
540
UGUGGACGCCUUCGCCUUC
1236





GCAACCUCGUGGAAGGCGA
541
GCAACCUCGUGGAAGGCGA
541
UCGCCUUCCACGAGGUUGC
1237





GACGCGGGCUGUGCUUGGU
542
GACGCGGGCUGUGCUUGGU
542
ACCAAGCACAGCCCGCGUC
1238





ACGCGGGCUGUGCUUGGUA
543
ACGCGGGCUGUGCUUGGUA
543
UACCAAGCACAGCCCGCGU
1239





GUGCAGCCUCCAGGACCCC
544
GUGCAGCCUCCAGGACCCC
544
GGGGUCCUGGAGGCUGCAC
1240





GCAGCCUCCAGGACCCCCC
545
GCAGCCUCCAGGACCCCCC
545
GGGGGGUCCUGGAGGCUGC
1241





CGCAACCUCGUGGAAGGCG
546
CGCAACCUCGUGGAAGGCG
546
CGCCUUCCACGAGGUUGCG
1242





UGUCGUGCAGCCUCCAGGA
547
UGUCGUGCAGCCUCCAGGA
547
UCCUGGAGGCUGCACGACA
1243





AUGGCUUGGGAUAUGAUGA
548
AUGGCUUGGGAUAUGAUGA
548
UCAUCAUAUCCCAAGCCAU
1244





CUUGGGAUAUGAUGAUGAA
549
CUUGGGAUAUGAUGAUGAA
549
UUCAUCAUCAUAUCCCAAG
1245





CCCUUGGCCCCUCUAUGGC
550
CCCUUGGCCCCUCUAUGGC
550
GCCAUAGAGGGGCCAAGGG
1246





UGGCUUGGGAUAUGAUGAU
551
UGGCUUGGGAUAUGAUGAU
551
AUCAUCAUAUCCCAAGCCA
1247





CUGUGCAGUGGAUGAACCG
552
CUGUGCAGUGGAUGAACCG
552
CGGUUCAUCCACUGCACAG
1248





AUGACGCGGGCUGUGCUUG
553
AUGACGCGGGCUGUGCUUG
553
CAAGCACAGCCCGCGUCAU
1249





GCUUGGGAUAUGAUGAUGA
554
GCUUGGGAUAUGAUGAUGA
554
UCAUCAUCAUAUCCCAAGC
1250





UAUGACGCGGGCUGUGCUU
555
UAUGACGCGGGCUGUGCUU
555
AAGCACAGCCCGCGUCAUA
1251





UGACGCGGGCUGUGCUUGG
556
UGACGCGGGCUGUGCUUGG
556
CCAAGCACAGCCCGCGUCA
1252





GGCUUGGGAUAUGAUGAUG
557
GGCUUGGGAUAUGAUGAUG
557
CAUCAUCAUAUCCCAAGCC
1253





UGUGCAGUGGAUGAACCGG
558
UGUGCAGUGGAUGAACCGG
558
CCGGUUCAUCCACUGCACA
1254





GCUGUGCAGUGGAUGAACC
559
GCUGUGCAGUGGAUGAACC
559
GGUUCAUCCACUGCACAGC
1255





CUCUUCAACUGGGCAGUAA
560
CUCUUCAACUGGGCAGUAA
560
UUACUGCCCAGUUGAAGAG
1256





CCUCGUGGAAGGCGACAAC
561
CCUCGUGGAAGGCGACAAC
561
GUUGUCGCCUUCCACGAGG
1257





UGUGUCACCCAGACAGUCG
562
UGUGUCACCCAGACAGUCG
562
CGACUGUCUGGGUGACACA
1258





GGCGUGAACUAUGCAACAG
563
GGCGUGAACUAUGCAACAG
563
CUGUUGCAUAGUUCACGCC
1259





CGGCGUGAACUAUGCAACA
564
CGGCGUGAACUAUGCAACA
564
UGUUGCAUAGUUCACGCCG
1260





GUGUCACCCAGACAGUCGA
565
GUGUCACCCAGACAGUCGA
565
UCGACUGUCUGGGUGACAC
1261





CCUCUUCAACUGGGCAGUA
566
CCUCUUCAACUGGGCAGUA
566
UACUGCCCAGUUGAAGAGG
1262





CGUGGAAGGCGACAACCUA
567
CGUGGAAGGCGACAACCUA
567
UAGGUUGUCGCCUUCCACG
1263





UCGUGGAAGGCGACAACCU
568
UCGUGGAAGGCGACAACCU
568
AGGUUGUCGCCUUCCACGA
1264





CGGCCUAGUUGGGGCCCCA
569
CGGCCUAGUUGGGGCCCCA
569
UGGGGCCCCAACUAGGCCG
1265





CGACUAGGAAGACUUCCGA
570
CGACUAGGAAGACUUCCGA
570
UCGGAAGUCUUCCUAGUCG
1266





UUUGGGUAAGGUCAUCGAU
571
UUUGGGUAAGGUCAUCGAU
571
AUCGAUGACCUUACCCAAA
1267





GUGGAAGGCGACAACCUAU
572
GUGGAAGGCGACAACCUAU
572
AUAGGUUGUCGCCUUCCAC
1268





ACCUCGUGGAAGGCGACAA
573
ACCUCGUGGAAGGCGACAA
573
UUGUCGCCUUCCACGAGGU
1269





GCGACUAGGAAGACUUCCG
574
GCGACUAGGAAGACUUCCG
574
CGGAAGUCUUCCUAGUCGC
1270





GUCGUGCAGCCUCCAGGAC
575
GUCGUGCAGCCUCCAGGAC
575
GUCCUGGAGGCUGCACGAC
1271





UAGGAAGACUUCCGAGCGG
576
UAGGAAGACUUCCGAGCGG
576
CCGCUCGGAAGUCUUCCUA
1272





ACGGCGUGAACUAUGCAAC
577
ACGGCGUGAACUAUGCAAC
577
GUUGCAUAGUUCACGCCGU
1273





CUCGUGGAAGGCGACAACC
578
CUCGUGGAAGGCGACAACC
578
GGUUGUCGCCUUCCACGAG
1274





GGUCGCAACCUCGUGGAAG
579
GGUCGCAACCUCGUGGAAG
579
CUUCCACGAGGUUGCGACC
1275





CGGUCGCAACCUCGUGGAA
580
CGGUCGCAACCUCGUGGAA
580
UUCCACGAGGUUGCGACCG
1276





GCGCGCGACUAGGAAGACU
581
GCGCGCGACUAGGAAGACU
581
AGUCUUCCUAGUCGCGCGC
1277





GACGGCGUGAACUAUGCAA
582
GACGGCGUGAACUAUGCAA
582
UUGCAUAGUUCACGCCGUC
1278





UAGAUCACUCCCCUGUGAG
583
UAGAUCACUCCCCUGUGAG
583
CUCACAGGGGAGUGAUCUA
1279





AGCGGUCGCAACCUCGUGG
584
AGCGGUCGCAACCUCGUGG
584
CCACGAGGUUGCGACCGCU
1280





UGGAAGGCGACAACCUAUC
585
UGGAAGGCGACAACCUAUC
585
GAUAGGUUGUCGCCUUCCA
1281





CGCGCGACUAGGAAGACUU
586
CGCGCGACUAGGAAGACUU
586
AAGUCUUCCUAGUCGCGCG
1282





CUAGGAAGACUUCCGAGCG
587
CUAGGAAGACUUCCGAGCG
587
CGCUCGGAAGUCUUCCUAG
1283





GUGCGCGCGACUAGGAAGA
588
GUGCGCGCGACUAGGAAGA
588
UCUUCCUAGUCGCGCGCAC
1284





AGAUCACUCCCCUGUGAGG
589
AGAUCACUCCCCUGUGAGG
589
CCUCACAGGGGAGUGAUCU
1285





UGCGCGCGACUAGGAAGAC
590
UGCGCGCGACUAGGAAGAC
590
GUCUUCCUAGUCGCGCGCA
1286





AUAGAUCACUCCCCUGUGA
591
AUAGAUCACUCCCCUGUGA
591
UCACAGGGGAGUGAUCUAU
1287





GAGCGGUCGCAACCUCGUG
592
GAGCGGUCGCAACCUCGUG
592
CACGAGGUUGCGACCGCUC
1288





CACGAACGACUGCUCCAAC
593
CACGAACGACUGCUCCAAC
593
GUUGGAGCAGUCGUUCGUG
1289





GGCAAGUUCCUUGCCGACG
594
GGCAAGUUCCUUGCCGACG
594
CGUCGGCAAGGAACUUGCC
1290





UCGUGCAGCCUCCAGGACC
595
UCGUGCAGCCUCCAGGACC
595
GGUCCUGGAGGCUGCACGA
1291





GUCACGAACGACUGCUCCA
596
GUCACGAACGACUGCUCCA
596
UGGAGCAGUCGUUCGUGAC
1292





GCGGUCGCAACCUCGUGGA
597
GCGGUCGCAACCUCGUGGA
597
UCCACGAGGUUGCGACCGC
1293





GCGCGACUAGGAAGACUUC
598
GCGCGACUAGGAAGACUUC
598
GAAGUCUUCCUAGUCGCGC
1294





GCUAUGACGCGGGCUGUGC
599
GCUAUGACGCGGGCUGUGC
599
GCACAGCCCGCGUCAUAGC
1295





UCACGAACGACUGCUCCAA
600
UCACGAACGACUGCUCCAA
600
UUGGAGCAGUCGUUCGUGA
1296





UCGCAACCUCGUGGAAGGC
601
UCGCAACCUCGUGGAAGGC
601
GCCUUCCACGAGGUUGCGA
1297





CGUGCAGCCUCCAGGACCC
602
CGUGCAGCCUCCAGGACCC
602
GGGUCCUGGAGGCUGCACG
1298





GUCGCAACCUCGUGGAAGG
603
GUCGCAACCUCGUGGAAGG
603
CCUUCCACGAGGUUGCGAC
1299





ACUAGGAAGACUUCCGAGC
604
ACUAGGAAGACUUCCGAGC
604
GCUCGGAAGUCUUCCUAGU
1300





CGCGACUAGGAAGACUUCC
605
CGCGACUAGGAAGACUUCC
605
GGAAGUCUUCCUAGUCGCG
1301





UGGGCGAAGCACAUGUGGA
606
UGGGCGAAGCACAUGUGGA
606
UCCACAUGUGCUUCGCCCA
1302





CCUUGCCUACUAUUCCAUG
607
CCUUGCCUACUAUUCCAUG
607
CAUGGAAUAGUAGGCAAGG
1303





GCCUCAGGAAACUUGGGGU
608
GCCUCAGGAAACUUGGGGU
608
ACCCCAAGUUUCCUGAGGC
1304





UGCUAUGACGCGGGCUGUG
609
UGCUAUGACGCGGGCUGUG
609
CACAGCCCGCGUCAUAGCA
1305





UCGUGCUCGCCACCGCUAC
610
UCGUGCUCGCCACCGCUAC
610
GUAGCGGUGGCGAGCACGA
1306





UGCCUCAGGAAACUUGGGG
611
UGCCUCAGGAAACUUGGGG
611
CCCCAAGUUUCCUGAGGCA
1307





UGUCUCGUGCCCGACCCCG
612
UGUCUCGUGCCCGACCCCG
612
CGGGGUCGGGCACGAGACA
1308





UGUGGCGGCAGGAGAUGGG
613
UGUGGCGGCAGGAGAUGGG
613
CCCAUCUCCUGCCGCCACA
1309





GUCGUGCUCGCCACCGCUA
614
GUCGUGCUCGCCACCGCUA
614
UAGCGGUGGCGAGCACGAC
1310





GAUUUCCACUACGUGACGG
615
GAUUUCCACUACGUGACGG
615
CCGUCACGUAGUGGAAAUC
1311





GGGCCUUGCCUACUAUUCC
616
GGGCCUUGCCUACUAUUCC
616
GGAAUAGUAGGCAAGGCCC
1312





GCCUUGCCUACUAUUCCAU
617
GCCUUGCCUACUAUUCCAU
617
AUGGAAUAGUAGGCAAGGC
1313





GACUAGGAAGACUUCCGAG
618
GACUAGGAAGACUUCCGAG
618
CUCGGAAGUCUUCCUAGUC
1314





GCGGGGGAGACAUAUAUCA
619
GCGGGGGAGACAUAUAUCA
619
UGAUAUAUGUCUCCCCCGC
1315





CGAGCGGUCGCAACCUCGU
620
CGAGCGGUCGCAACCUCGU
620
ACGAGGUUGCGACCGCUCG
1316





GGCCUUGCCUACUAUUCCA
621
GGCCUUGCCUACUAUUCCA
621
UGGAAUAGUAGGCAAGGCC
1317





AUUUCCACUACGUGACGGG
622
AUUUCCACUACGUGACGGG
622
CCCGUCACGUAGUGGAAAU
1318





GGACGUCAAGUUCCCGGGC
623
GGACGUCAAGUUCCCGGGC
623
GCCCGGGAACUUGACGUCC
1319





GAGUGCUAUGACGCGGGCU
624
GAGUGCUAUGACGCGGGCU
624
AGCCCGCGUCAUAGCACUC
1320





GACGUCAAGUUCCCGGGCG
625
GACGUCAAGUUCCCGGGCG
625
CGCCCGGGAACUUGACGUC
1321





UCAGCGACGGGUCUUGGUC
626
UCAGCGACGGGUCUUGGUC
626
GACCAAGACCCGUCGCUGA
1322





UCAAGUUCCCGGGCGGUGG
627
UCAAGUUCCCGGGCGGUGG
627
CCACCGCCCGGGAACUUGA
1323





UCAAGGAGAUGAAGGCGAA
628
UCAAGGAGAUGAAGGCGAA
628
UUCGCCUUCAUCUCCUUGA
1324





CCUAUCCCCAAGGCUCGCC
629
CCUAUCCCCAAGGCUCGCC
629
GGCGAGCCUUGGGGAUAGG
1325





CUUGACCUACCUCAGAUCA
630
CUUGACCUACCUCAGAUCA
630
UGAUCUGAGGUAGGUCAAG
1326





UUUCCACUACGUGACGGGC
631
UUUCCACUACGUGACGGGC
631
GCCCGUCACGUAGUGGAAA
1327





AGUGCUAUGACGCGGGCUG
632
AGUGCUAUGACGCGGGCUG
632
CAGCCCGCGUCAUAGCACU
1328





ACGUCAAGUUCCCGGGCGG
633
ACGUCAAGUUCCCGGGCGG
633
CCGCCCGGGAACUUGACGU
1329





UCUGGAGACAUCGGGCCAG
634
UCUGGAGACAUCGGGCCAG
634
CUGGCCCGAUGUCUCCAGA
1330





GGGCGAAGCACAUGUGGAA
635
GGGCGAAGCACAUGUGGAA
635
UUCCACAUGUGCUUCGCCC
1331





UUGACCUACCUCAGAUCAU
636
UUGACCUACCUCAGAUCAU
636
AUGAUCUGAGGUAGGUCAA
1332





CCAAGCGGAGACGGCUGGA
637
CCAAGCGGAGACGGCUGGA
637
UCCAGCCGUCUCCGCUUGG
1333





ACCAAGCGGAGACGGCUGG
638
ACCAAGCGGAGACGGCUGG
638
CCAGCCGUCUCCGCUUGGU
1334





GGGUGGCUUCAUGCCUCAG
639
GGGUGGCUUCAUGCCUCAG
639
CUGAGGCAUGAAGCCACCC
1335





GUCAAGUUCCCGGGCGGUG
640
GUCAAGUUCCCGGGCGGUG
640
CACCGCCCGGGAACUUGAC
1336





CUCAAGGAGAUGAAGGCGA
641
CUCAAGGAGAUGAAGGCGA
641
UCGCCUUCAUCUCCUUGAG
1337





GACCAAGCGGAGACGGCUG
642
GACCAAGCGGAGACGGCUG
642
CAGCCGUCUCCGCUUGGUC
1338





UCCAGGUCGGGCUCAACCA
643
UCCAGGUCGGGCUCAACCA
643
UGGUUGAGCCCGACCUGGA
1339





CUCUUUCUCUAUCUUCCUC
644
CUCUUUCUCUAUCUUCCUC
644
GAGGAAGAUAGAGAAAGAG
1340





GUCUGGAGACAUCGGGCCA
645
GUCUGGAGACAUCGGGCCA
645
UGGCCCGAUGUCUCCAGAC
1341





GUUGUGACUUGGCCCCCGA
646
GUUGUGACUUGGCCCCCGA
646
UCGGGGGCCAAGUCACAAC
1342





AGACCUGGCUCCAGUCCAA
647
AGACCUGGCUCCAGUCCAA
647
UUGGACUGGAGCCAGGUCU
1343





CUUGCCUACUAUUCCAUGG
648
CUUGCCUACUAUUCCAUGG
648
CCAUGGAAUAGUAGGCAAG
1344





CCCGGUUGCUCUUUCUCUA
649
CCCGGUUGCUCUUUCUCUA
649
UAGAGAAAGAGCAACCGGG
1345





CUUUCUCUAUCUUCCUCUU
650
CUUUCUCUAUCUUCCUCUU
650
AAGAGGAAGAUAGAGAAAG
1346





AGGGUGGCUUCAUGCCUCA
651
AGGGUGGCUUCAUGCCUCA
651
UGAGGCAUGAAGCCACCCU
1347





AAGACCUGGCUCCAGUCCA
652
AAGACCUGGCUCCAGUCCA
652
UGGACUGGAGCCAGGUCUU
1348





CCGGUUGCUCUUUCUCUAU
653
CCGGUUGCUCUUUCUCUAU
653
AUAGAGAAAGAGCAACCGG
1349





CGGUUGCUCUUUCUCUAUC
654
CGGUUGCUCUUUCUCUAUC
654
GAUAGAGAAAGAGCAACCG
1350





UGGGGGAUUUCCACUACGU
655
UGGGGGAUUUCCACUACGU
655
ACGUAGUGGAAAUCCCCCA
1351





AUGUCACGAACGACUGCUC
656
AUGUCACGAACGACUGCUC
656
GAGCAGUCGUUCGUGACAU
1352





GGCCUAGUUGGGGCCCCAC
657
GGCCUAGUUGGGGCCCCAC
657
GUGGGGCCCCAACUAGGCC
1353





UGGACCAAGCGGAGACGGC
658
UGGACCAAGCGGAGACGGC
658
GCCGUCUCCGCUUGGUCCA
1354





UUCCAGGUCGGGCUCAACC
659
UUCCAGGUCGGGCUCAACC
659
GGUUGAGCCCGACCUGGAA
1355





AGCGGGUCGAGUUCCUGGU
660
AGCGGGUCGAGUUCCUGGU
660
ACCAGGAACUCGACCCGCU
1356





CAAGGAGAUGAAGGCGAAG
661
CAAGGAGAUGAAGGCGAAG
661
CUUCGCCUUCAUCUCCUUG
1357





CAUGUCACGAACGACUGCU
662
CAUGUCACGAACGACUGCU
662
AGCAGUCGUUCGUGACAUG
1358





CAGCGGGUCGAGUUCCUGG
663
CAGCGGGUCGAGUUCCUGG
663
CCAGGAACUCGACCCGCUG
1359





UUCCACUACGUGACGGGCA
664
UUCCACUACGUGACGGGCA
664
UGCCCGUCACGUAGUGGAA
1360





UAGGGUGGCUUCAUGCCUC
665
UAGGGUGGCUUCAUGCCUC
665
GAGGCAUGAAGCCACCCUA
1361





UCCAGGACUGCACGAUGCU
666
UCCAGGACUGCACGAUGCU
666
AGCAUCGUGCAGUCCUGGA
1362





UCCACUACGUGACGGGCAU
667
UCCACUACGUGACGGGCAU
667
AUGCCCGUCACGUAGUGGA
1363





AAUAGGGUGGCUUCAUGCC
668
AAUAGGGUGGCUUCAUGCC
668
GGCAUGAAGCCACCCUAUU
1364





GUCUUCACGGAGGCUAUGA
669
GUCUUCACGGAGGCUAUGA
669
UCAUAGCCUCCGUGAAGAC
1365





AUAGGGUGGCUUCAUGCCU
670
AUAGGGUGGCUUCAUGCCU
670
AGGCAUGAAGCCACCCUAU
1366





UCUUCACGGAGGCUAUGAC
671
UCUUCACGGAGGCUAUGAC
671
GUCAUAGCCUCCGUGAAGA
1367





AUGCCUCAGGAAACUUGGG
672
AUGCCUCAGGAAACUUGGG
672
CCCAAGUUUCCUGAGGCAU
1368





ACCGGGACGUGCUCAAGGA
673
ACCGGGACGUGCUCAAGGA
673
UCCUUGAGCACGUCCCGGU
1369





GGGGCUGUGCAGUGGAUGA
674
GGGGCUGUGCAGUGGAUGA
674
UCAUCCACUGCACAGCCCC
1370





AAGCUCCAGGACUGCACGA
675
AAGCUCCAGGACUGCACGA
675
UCGUGCAGUCCUGGAGCUU
1371





GCUCCAGGACUGCACGAUG
676
GCUCCAGGACUGCACGAUG
676
CAUCGUGCAGUCCUGGAGC
1372





UACCGGGACGUGCUCAAGG
677
UACCGGGACGUGCUCAAGG
677
CCUUGAGCACGUCCCGGUA
1373





GGGCUGUGCAGUGGAUGAA
678
GGGCUGUGCAGUGGAUGAA
678
UUCAUCCACUGCACAGCCC
1374





CGUCAAGUUCCCGGGCGGU
679
CGUCAAGUUCCCGGGCGGU
679
ACCGCCCGGGAACUUGACG
1375





UCAAUAGGGUGGCUUCAUG
680
UCAAUAGGGUGGCUUCAUG
680
CAUGAAGCCACCCUAUUGA
1376





AGUCUUCACGGAGGCUAUG
681
AGUCUUCACGGAGGCUAUG
681
CAUAGCCUCCGUGAAGACU
1377





GGACCAAGCGGAGACGGCU
682
GGACCAAGCGGAGACGGCU
682
AGCCGUCUCCGCUUGGUCC
1378





GGCUCCAGUCCAAGCUCCU
683
GGCUCCAGUCCAAGCUCCU
683
AGGAGCUUGGACUGGAGCC
1379





GGCUGUGCAGUGGAUGAAC
684
GGCUGUGCAGUGGAUGAAC
684
GUUCAUCCACUGCACAGCC
1380





CUCCAGGACUGCACGAUGC
685
CUCCAGGACUGCACGAUGC
685
GCAUCGUGCAGUCCUGGAG
1381





GAGUCUUCACGGAGGCUAU
686
GAGUCUUCACGGAGGCUAU
686
AUAGCCUCCGUGAAGACUC
1382





UGGCUCCAGUCCAAGCUCC
687
UGGCUCCAGUCCAAGCUCC
687
GGAGCUUGGACUGGAGCCA
1383





GGGGAUUUCCACUACGUGA
688
GGGGAUUUCCACUACGUGA
688
UCACGUAGUGGAAAUCCCC
1384





CAUGCCUCAGGAAACUUGG
689
CAUGCCUCAGGAAACUUGG
689
CCAAGUUUCCUGAGGCAUG
1385





AUCAAUAGGGUGGCUUCAU
690
AUCAAUAGGGUGGCUUCAU
690
AUGAAGCCACCCUAUUGAU
1386





GCGGGCCUUGCCUACUAUU
691
GCGGGCCUUGCCUACUAUU
691
AAUAGUAGGCAAGGCCCGC
1387





CCGGGACGUGCUCAAGGAG
692
CCGGGACGUGCUCAAGGAG
692
CUCCUUGAGCACGUCCCGG
1388





CCAUGGUGGGGAACUGGGC
693
CCAUGGUGGGGAACUGGGC
693
GCCCAGUUCCCCACCAUGG
1389





CAAUAGGGUGGCUUCAUGC
694
CAAUAGGGUGGCUUCAUGC
694
GCAUGAAGCCACCCUAUUG
1390





AGCUCCAGGACUGCACGAU
695
AGCUCCAGGACUGCACGAU
695
AUCGUGCAGUCCUGGAGCU
1391





CGGGCCUUGCCUACUAUUC
696
CGGGCCUUGCCUACUAUUC
696
GAAUAGUAGGCAAGGCCCG
1392







The 3′-ends of the Upper sequence and the Lower sequence of the siNA construct can include an overhang sequence, for example about 1, 2, 3, or 4 nucleotides in length, preferably 2 nucleotides in length, wherein the overhanging sequence of the lower sequence is optionally complementary to a portion of the target sequence. The upper and lower sequences in the Table further can further comprise a chemical modification having Formulae I-VII, such as exemplary siNA constructs



# shown in FIGS. 4 and 5, or having modifications described in Table IV or any combination thereof.













TABLE III










HCV Synthetic Modified siNA Constructs














Tar-









get

Seq



Seq


Pos
Target
ID
Cmpd#
Aliases
Sequence
ID

















183
GGUCCUUUCUUGGAUCAACCCGC
1393
25237
HCV IRES Loop IIIb
B GGUCCUUUCUUGGAUCAACCC B
1467







(Heptazyme site) as






siNA str1 (sense)





183
GGUCCUUUCUUGGAUCAACCCGC
1393
25238
HCV IRES Loop IIIb
B GGGUUGAUCCAAGAAAGGACC B
1468






(Heptazyme site) as






siNA str2 (antisense)





183
GGUCCUUUCUUGGAUCAACCCGC
1393
25251
HCV IRES Loop IIIb
B CCCAACUAGGUUCUUUCCUGG B
1469






(Heptazyme site) as






siNA str1 (sense)






Inverted Control





183
GGUCCUUUCUUGGAUCAACCCGC
1393
25252
HCV IRES Loop IIIb
B CCAGGAAAGAACCUAGUUGGG B
1470






(Heptazyme site) as






siNA str1 (sense)






Inverted Control






Compliment





183
GGUCCUUUCUUGGAUCAACCCGC
1393
25814
HCV IRES Loop IIIb
GGUCCUUUCUUGGAUCAACCCUU
1471






(Heptazyme site) as






siNA str1 (sense) +






2U overhang





183
GGUCCUUUCUUGGAUCAACCCGC
1393
25815
HCV IRES Loop IIIb
GGGUUGAUCCAAGAAAGGACCUU
1472






(Heptazyme site) as






siNA str2






(antisense) + 2U






overhang





183
GGUCCUUUCUUGGAUCAACCCGC
1393
25834
HCV IRES Loop IIIb
BGGUCCUUUCUUGGAUCAACCCUUB
1473






(Heptazyme site) as






siNA str1 (sense) +






2U overhang





183
GGUCCUUUCUUGGAUCAACCCGC
1393
25835
HCV IRES Loop IIIb
BGGGUUGAUCCAAGAAAGGACCUUB
1474






(Heptazyme site) as






siNA str2






(antisense) + 2U






overhang





325
UGCCCCGGGAGGUCUCGUAGACC
1394
28415
HCVa:325U21 sense TT
CCCCGGGAGGUCUCGUAGATT
1475






siNA





162
CGGAACCGGUGAGUACACC
54
28416
HCVa:162U21 sense TT
CGGAACCGGUGAGUACACCTT
1476






siNA





324
GCCCCGGGAGGUCUCGUAG
1
28417
HCVa:324U21 sense TT
GCCCCGGGAGGUCUCGUAGTT
1477






siNA





163
GGAACCGGUGAGUACACCG
53
28418
HCVa:163U21 sense TT
GGAACCGGUGAGUACACCGTT
1478






siNA





294
GUGGUACUGCCUGAUAGGG
5
28419
HCVa:294U21 sense TT
GUGGUACUGCCUGAUAGGGTT
1479






siNA





293
UGUGGUACUGCCUGAUAGG
2
28420
HCVa:293U21 sense TT
UGUGGUACUGCCUGAUAGGTT
1480






siNA





292
UUGUGGUACUGCCUGAUAG
3
28421
HCVa:292U21 sense TT
UUGUGGUACUGCCUGAUAGTT
1481






siNA





325
UGCCCCGGGAGGUCUCGUAGACC
1394
28422
HCVa:343L21 antisense
UCUACGAGACCUCCCGGGGTT
1482






TT siNA (325C)





162
CGGAACCGGUGAGUACACC
54
28423
HCVa:180L21 antisense
GGUGUACUCACCGGUUCCGTT
1483






TT siNA (162C)





324
GCCCCGGGAGGUCUCGUAG
1
28424
HCVa:342L21 antisense
CUACGAGACCUCCCGGGGCTT
1484






TT siNA (324C)





163
GGAACCGGUGAGUACACCG
53
28425
HCVa:181L21 antisense
CGGUGUACUCACCGGUUCCTT
1485






TT siNA (163C)





294
GUGGUACUGCCUGAUAGGG
5
28426
HCVa:312L21 antisense
CCCUAUCAGGCAGUACCACTT
1486






TT siNA (294C)





293
UGUGGUACUGCCUGAUAGG
2
28427
HCVa:311L21 antisense
CCUAUCAGGCAGUACCACATT
1487






TT siNA (293C)





292
UUGUGGUACUGCCUGAUAG
3
28428
HCVa:310L21 antisense
CUAUCAGGCAGUACCACAATT
1488






TT siNA (292C)





325
UGCCCCGGGAGGUCUCGUAGACC
1394
28429
HCVa:325U21 sense TT
TTAGAUGCUCUGGAGGGCCCC
1489






siNA inv





162
CGGAACCGGUGAGUACACC
54
28430
HCVa:162U21 sense TT
TTCCACAUGAGUGGCCAAGGC
1490






siNA inv





324
GCCCCGGGAGGUCUCGUAG
1
28431
HCVa:324U21 sense TT
TTGAUGCUCUGGAGGGCCCCG
1491






siNA inv





163
GGAACCGGUGAGUACACCG
53
28432
HCVa:163U21 sense TT
TTGCCACAUGAGUGGCCAAGG
1492






siNA inv





294
GUGGUACUGCCUGAUAGGG
5
28433
HCVa:294U21 sense TT
TTGGGAUAGUCCGUCAUGGUG
1493






siNA inv





293
UGUGGUACUGCCUGAUAGG
2
28434
HCVa:293U21 sense TT
TTGGAUAGUCCGUCAUGGUGU
1494






siNA inv





292
UUGUGGUACUGCCUGAUAG
3
28435
HCVa:292U21 sense TT
TTGAUAGUCCGUCAUGGUGUU
1495






siNA inv





325
UGCCCCGGGAGGUCUCGUAGACC
1394
28436
HCVa:343L21 antisense
TTGGGGCCCUCCAGAGCAUCU
1496






TT siNA (325C) inv





162
CGGAACCGGUGAGUACACC
54
28437
HCVa:180L21 antisense
TTGCCUUGGCCACUCAUGUGG
1497






TT siNA (162C) inv





324
GCCCCGGGAGGUCUCGUAG
1
28438
HCVa:342L21 antisense
TTCGGGGCCCUCCAGAGCAUC
1498






TT siNA (324C) inv





163
GGAACCGGUGAGUACACCG
53
28439
HCVa:181L21 antisense
TTCCUUGGCCACUCAUGUGGC
1499






TT siNA (163C) inv





294
GUGGUACUGCCUGAUAGGG
5
28440
HCVa:312L21 antisense
TTCACCAUGACGGACUAUCCC
1500






TT siNA (294C) inv





293
UGUGGUACUGCCUGAUAGG
2
28441
HCVa:311L21 antisense
TTACACCAUGACGGACUAUCC
1501






TT siNA (293C) inv





292
UUGUGGUACUGCCUGAUAG
3
28442
HCVa:310L21 antisense
TTAACACCAUGACGGACUAUC
1502






TT siNA (292C) inv





162
UGCGGAACCGGUGAGUACACCGG
1395
29573
HCVa:162U21 sense
CGGAACCGGUGAGUACACCGG
1503






siNA





163
GCGGAACCGGUGAGUACACCGGA
1396
29574
HCVa:163U21 sense
GGAACCGGUGAGUACACCGGA
1504






siNA





292
CCUUGUGGUACUGCCUGAUAGGG
1397
29575
HCVa:292U21 sense
UUGUGGUACUGCCUGAUAGGG
1505






siNA





293
CUUGUGGUACUGCCUGAUAGGGU
1398
29576
HCVa:293U21 sense
UGUGGUACUGCCUGAUAGGGU
1508






siNA





294
UUGUGGUACUGCCUGAUAGGGUG
1399
29577
HCVa:294U21 sense
GUGGUACUGCCUGAUAGGGUG
1507






siNA





324
GUGCCCCGGGAGGUCUCGUAGAC
1400
29578
HCVa:324U21 sense
GCCCCGGGAGGUCUCGUAGAC
1508






siNA





325
UGCCCCGGGAGGUCUCGUAGACC
1394
29579
HCVa:325U21 sense
CCCCGGGAGGUCUCGUAGACC
1509






siNA





162
UGCGGAACCGGUGAGUACACCGG
1395
29580
HCVa:180L21 antisense
GGUGUACUCACCGGUUCCGCA
1510






siNA (162C)





163
GCGGAACCGGUGAGUACACCGGA
1396
29581
HCVa:181L21 antisense
CGGUGUACUCACCGGUUCCGC
1511






siNA (163C)





292
CCUUGUGGUACUGCCUGAUAGGG
1397
29582
HCVa:310L21 antisense
CUAUCAGGCAGUACCACAAGG
1512






siNA (292C)





293
CUUGUGGUACUGCCUGAUAGGGU
1398
29583
HCVa:311L21 antisense
CCUAUCAGGCAGUACCACAAG
1513






siNA (293C)





294
UUGUGGUACUGCCUGAUAGGGUG
1399
29584
HCVa:312L21 antisense
CCCUAUCAGGCAGUACCACAA
1514






siNA (294C)





324
GUGCCCCGGGAGGUCUCGUAGAC
1400
29585
HCVa:342L21 antisense
CUACGAGACCUCCCGGGGCAC
1515






siNA (324C)





325
UGCCCCGGGAGGUCUCGUAGACC
1394
29586
HCVa:343L21 antisense
UCUACGAGACCUCCCGGGGCA
1516






siNA (325C)





162
UGCGGAACCGGUGAGUACACCGG
1395
29587
HCVa:162U21 sense
GGCCACAUGAGUGGCCAAGGC
1517






siNA inv





163
GCGGAACCGGUGAGUACACCGGA
1396
29588
HCVa:163U21 sense
AGGCCACAUGAGUGGCCAAGG
1518






siNA inv





292
CCUUGUGGUACUGCCUGAUAGGG
1397
29589
HCVa:292U21 sense
GGGAUAGUCCGUCAUGGUGUU
1519






siNA inv





293
CUUGUGGUACUGCCUGAUAGGGU
1398
29590
HCVa:293U21 sense
UGGGAUAGUCCGUCAUGGUGU
1520






siNA inv





294
UUGUGGUACUGCCUGAUAGGGUG
1399
29591
HCVa:294U21 sense
GUGGGAUAGUCCGUCAUGGUG
1521






siNA inv





324
GUGCCCCGGGAGGUCUCGUAGAC
1400
29592
HCVa:324U21 sense
CAGAUGCUCUGGAGGGCCCCG
1522






siNA inv





325
UGCCCCGGGAGGUCUCGUAGACC
1394
29593
HCVa:325U21 sense
CCAGAUGCUCUGGAGGGCCCC
1523






siNA inv





162
UGCGGAACCGGUGAGUACACCGG
1395
29594
HCVa:180L21 antisense
ACGCCUUGGCCACUCAUGUGG
1524






siNA (162C) inv





163
GCGGAACCGGUGAGUACACCGGA
1396
29595
HCVa:181L21 antisense
CGCCUUGGCCACUCAUGUGGC
1525






siNA (163C) inv





292
CCUUGUGGUACUGCCUGAUAGGG
1397
29596
HCVa:310L21 antisense
GGAACACCAUGACGGACUAUC
1526






siNA (292C) inv





293
CUUGUGGUACUGCCUGAUAGGGU
1398
29597
HCVa:311L21 antisense
GAACACCAUGACGGACUAUCC
1527






siNA (293C) inv





294
UUGUGGUACUGCCUGAUAGGGUG
1399
29598
HCVa:312L21 antisense
AACACCAUGACGGACUAUCCC
1528






siNA (294C) inv





324
GUGCCCCGGGAGGUCUCGUAGAC
1400
29599
HCVa:342L21 antisense
CACGGGGCCCUCCAGAGCAUC
1529






siNA (324C) inv





325
UGCCCCGGGAGGUCUCGUAGACC
1394
29600
HCVa:343L21 antisense
ACGGGGCCCUCCAGAGCAUCU
1530






siNA (325C) inv





325
UGCCCCGGGAGGUCUCGUAGACC
1394
30051
HCVa:325U21 sense
BCsCsCsCsGsGGAGGUCUCGUAGAXXB
1531






siNA 5 5′ P = S + 3′






univ. base 2 + 5′/3′






invAba





325
UGCCCCGGGAGGUCUCGUAGACC
1394
30052
HCVa:325U21 sense
BAsGsAsUsGsCUCUGGAGGGCCCCXXB
1532






siNA inv 5 5′ P = S +






3′ univ. base 2 +






5′/3′ invAba





325
UGCCCCGGGAGGUCUCGUAGACC
1394
30053
HCVa:343L21 antisense
UsCsUsAsCsGAGACCUCCCGGGGXXB
1533






siNA (325C) 5 5′






P = S + 3′ univ. base






2 + 3′ invAba





325
UGCCCCGGGAGGUCUCGUAGACC
1394
30054
HCVa:343L21 antisense
GsGsGsGsCsCCUCCAGAGCAUCUXXB
1534






siNA (325C) inv 5 5′






P = S + 3′ univ. base






2 + 3′ invAba





325
UGCCCCGGGAGGUCUCGUAGACC
1394
30055
HCVa:325U21 sense
BCsCsCsCsGGGAGGUsCsUsCsGUsAGAXXB
1535






siNA all Y P = S + 3′






univ. base 2 + 5′/3′






invAba





325
UGCCCCGGGAGGUCUCGUAGACC
1394
30056
HCVa:325U21 sense
BAGAUsGCsUsCsUsGGAGGGCsCsCsCsXXB
1536






siNA inv all Y






P = S + 3′ univ. base






2 + 5′/3′ invAba





325
UGCCCCGGGAGGUCUCGUAGACC
1394
30057
HCVa:343L21 antisense
UsCsUsACsGAGACsCsUsCsCsCsGGGGXXB
1537






siNA (325C) all Y






P = S + 3′ univ. base






2 + 3′ invAba





325
UGCCCCGGGAGGUCUCGUAGACC
1394
30058
HCVa:343L21 antisense
GGGGCsCsCsUsCsCsAGAGCsAUsCsUsXXB
1538






siNA (325C) inv all Y






P = S + 3′ univ. base






2 + 3′ invAba





325
UGCCCCGGGAGGUCUCGUAGACC
1394
30059
HCVa:325U21 sense
BcscscscsGGGAGGucucGuAsGsAsXXB
1539






siNA 4/3 P = S ends +






all Y − 2′F + 3′






univ. base 2 + 5′/3′






invAba





325
UGCCCCGGGAGGUCUCGUAGACC
1394
30060
HCVa:325U21 sense
BAsGsAsusGcucuGGAGGGccscscsXXB
1540






siNA inv 4/3 P = S






ends + all Y − 2′F +






3′ univ. base 2 +






5′/3′ invAba





325
UGCCCCGGGAGGUCUCGUAGACC
1394
30170
HCVa:325U21 sense
B ccccGGGAGGucucGuAGAXX B
1541






siNA all Y − 2′F +






3′ univ. base 2 +






5′/3′ invAba





325
UGCCCCGGGAGGUCUCGUAGACC
1394
30171
HCVa:325U21 sense
B AGAuGcucuGGAGGGccccXX B 1542






siNA inv all Y −






2′F + 3′ univ. base






2 + 5′/3′ invAba





325
UGCCCCGGGAGGUCUCGUAGACC
1394
30172
HCVa:343L21 antisense
B UsCsUsACsGAGACsCsUsCsCsCsGGGGXX B
1543






siNA (325C) all Y






P = S + 3′ univ. base






2 + 5′/3′ invAba





325
UGCCCCGGGAGGUCUCGUAGACC
1394
30173
HCVa:343L19 antisense
ucuAcGAGAccucccGGGG
1544






siNA (325C) all Y −






2′F








325
UGCCCCGGGAGGUCUCGUAGACC
1394
30175
HCVa:343L21 antisense
ucuAcGAGAccucccGGGGXX
1545






siNA (325C) all






Y − 2′F + 3′ univ.






Base 2





325
UGCCCCGGGAGGUCUCGUAGACC
1394
30176
HCVa:343L21 antisense
GGGGcccuccAGAGcAucuXX
1546






siNA (325C) inv all






Y − 2′F + 3′ univ.






Base 2





325
UGCCCCGGGAGGUCUCGUAGACC
1394
30177
HCVa:343L21 antisense
B ucuAcGAGAccucccGGGGXX B
1547






siNA (325C) all






Y − 2′F + 3′ univ.






Base 2 + 5′/3′ iB





325
UGCCCCGGGAGGUCUCGUAGACC
1394
30178
HCVa:325U21 sense
CsCsCsCsGGGAGGUsCsUsCsGUsAGAXX B
1548






siNA all Y P = S + 3′






univ. base 2 + 3′






invAba





325
UGCCCCGGGAGGUCUCGUAGACC
1394
30417
HCVa:325U21 sense
CCCCGGGAGGUCUCGUAGACC B
1549






siNA w/iB





325
UGCCCCGGGAGGUCUCGUAGACC
1394
30418
HCVa:325U21 sense
B CCCCGGGAGGUCUCGUAGACC B
1550






siNA w/iB





325
UGCCCCGGGAGGUCUCGUAGACC
1394
30419
HCVa:343L21 antisense
UCUACGAGACCUCCCGGGGCA B
1551






siNA (325C) w/iB





325
UGCCCCGGGAGGUCUCGUAGACC
1394
30420
HCVa:343L21 antisense
B UCUACGAGACCUCCCGGGGCA B
1552






siNA (325C) w/iB





325
UGCCCCGGGAGGUCUCGUAGACC
1394
30561
HCVa:325U21 sense
B ccccGGGAGGucucGuAGATT B
1553






siNA Y − 2′Ome






(stab06) + 5′/3′






invAba





325
UGCCCCGGGAGGUCUCGUAGACC
1394
30562
HCVa:343L21 antisense
ucuAcGAGAccucccGGGGTsT
1554






siNA (325C) Y − 2′F,






R − 2′Ome + TsT





153
AUAGUGGUCUGCGGAACCGGUGA
1401
30649
HCVa:153U21 sense
B AGuGGucuGcGGAAccGGuTT B
1555






siNA stab07





159
GUCUGCGGAACCGGUGAGUACAC
1402
30650
HCVa:159U21 sense
B cuGcGGAAccGGuGAGuAcTT B
1556






siNA stab07





291
GCCUUGUGGUACUGCCUGAUAGG
1403
30651
HCVa:291U21 sense
B cuuGuGGuAcuGccuGAuATT B
1557






siNA stab07





295
UGUGGUACUGCCUGAUAGGGUGC
1404
30652
HCVa:295U21 sense
B uGGuAcuGccuGAuAGGGuTT B
1558






siNA stab07





296
GUGGUACUGCCUGAUAGGGUGCU
1405
30653
HCVa:296U21 sense
B GGuAcuGccuGAuAGGGuGTT B
1559






siNA stab07





297
UGGUACUGCCUGAUAGGGUGCUU
1406
30654
HCVa:297U21 sense
B GuAcuGccuGAuAGGGuGcTT B
1560






siNA stab07





298
GGUACUGCCUGAUAGGGUGCUUG
1407
30655
HCVa:298U21 sense
B uAcuGccuGAuAGGGuGcuTT B
1561






siNA stab07





300
UACUGCCUGAUAGGGUGCUUGCG
1408
30656
HCVa:300U21 sense
B cuGccuGAuAGGGuGcuuGTT B
1562






siNA stab07





301
ACUGCCUGAUAGGGUGCUUGCGA
1409
30657
HCVa:301U21 sense
B uGccuGAuAGGGuGcuuGcTT B
1563






siNA stab07





303
UGCCUGAUAGGGUGCUUGCGAGU
1410
30658
HCVa:303U21 sense
B ccuGAuAGGGuGcuuGcGATT B
1564






siNA stab07





306
CUGAUAGGGUGCUUGCGAGUGCC
1411
30659
HCVa:306U21 sense
B GAuAGGGuGcuuGcGAGuGTT B
1565






siNA stab07





324
GUGCCCCGGGAGGUCUCGUAGAC
1400
30660
HCVa:324U21 sense
B GccccGGGAGGucucGuAGTT B
1566






siNA stab07





153
AUAGUGGUCUGCGGAACCGGUGA
1401
30661
HCVa:171L21 antisense

AccGGuuccGcAGAccAcuTsT

1567






siNA (153C) stab08





159
GUCUGCGGAACCGGUGAGUACAC
1402
30662
HCVa:177L21 antisense

GuAcucAccGGuuccGcAGTsT

1568






siNA (159C) stab08





291
GCCUUGUGGUACUGCCUGAUAGG
1403
30663
HCVa:309L21 antisense
uAucAGGcAGuAccAcAAGTsT
1569






siNA (291C) stab08





295
UGUGGUACUGCCUGAUAGGGUGC
1404
30664
HCVa:313L21 antisense

AcccuAucAGGcAGuAccATsT

1570






siNA (295C) stab08





296
GUGGUACUGCCUGAUAGGGUGCU
1405
30665
HCVa:314L21 antisense
cAcccuAucAGGcAGuAccTsT
1571






siNA (296C) stab08





297
UGGUACUGCCUGAUAGGGUGCUU
1406
30666
HCVa:315L21 antisense

GcAcccuAucAGGcAGuAcTsT

1572






siNA (297C) stab08





298
GGUACUGCCUGAUAGGGUGCUUG
1407
30667
HCVa:316L21 antisense

AGcAcccuAucAGGcAGuATsT

1573






siNA (298C) stab08





300
UACUGCCUGAUAGGGUGCUUGCG
1408
30668
HCVa:318L21 antisense
cAAGcAcccuAucAGGcAGTsT
1574






siNA (300C) stab08





301
ACUGCCUGAUAGGGUGCUUGCGA
1409
30669
HCVa:319L21 antisense

GcAAGcAcccuAucAGGcATsT

1575






siNA (301C) stab08





303
UGCCUGAUAGGGUGCUUGCGAGU
1410
30670
HCVa:321L21 antisense
ucGcAAGcAcccuAucAGGTsT
1576






siNA (303C) stab08





306
CUGAUAGGGUGCUUGCGAGUGCC
1411
30671
HCVa:324L21 antisense
cAcucGcAAGcAcccuAucTsT
1577






siNA (306C) stab08





324
GUGCCCCGGGAGGUCUCGUAGAC
1400
30672
HCVa:342L21 antisense
cuAcGAGAccucccGGGGcTsT
1578






siNA (324C) stab08





153
AUAGUGGUCUGCGGAACCGGUGA
1401
30673
HCVa:153U21 sense
B uGGccAAGGcGucuGGuGATT B
1579






siNA stab07 inv





159
GUCUGCGGAACCGGUGAGUACAC
1402
30674
HCVa:159U21 sense
B cAuGAGuGGccAAGGcGucTT B
1580






siNA stab07 inv





291
GCCUUGUGGUACUGCCUGAUAGG
1403
30675
HCVa:291U21 sense
B AuAGuccGucAuGGuGuucTT B
1581






siNA stab07 inv





295
UGUGGUACUGCCUGAUAGGGUGC
1404
30676
HCVa:295U21 sense
B uGGGAuAGuccGucAuGGuTT B
1582






siNA stab07 inv





296
GUGGUACUGCCUGAUAGGGUGCU
1405
30677
HCVa:296U21 sense
B GuGGGAuAGuccGucAuGGTT B
1583






siNA stab07 inv





297
UGGUACUGCCUGAUAGGGUGCUU
1406
30678
HCVa:297U21 sense
B cGuGGGAuAGuccGucAuGTT B
1584






siNA stab07 inv





298
GGUACUGCCUGAUAGGGUGCUUG
1407
30679
HCVa:298U21 sense
B ucGuGGGAuAGuccGucAuTT B
1585






siNA stab07 inv





300
UACUGCCUGAUAGGGUGCUUGCG
1408
30680
HCVa:300U21 sense
B GuucGuGGGAuAGuccGucTT B
1586






siNA stab07 inv





301
ACUGCCUGAUAGGGUGCUUGCGA
1409
30681
HCVa:301U21 sense
B cGuucGuGGGAuAGuccGuTT B
1587






siNA stab07 inv





303
UGCCUGAUAGGGUGCUUGCGAGU
1410
30682
HCVa:303U21 sense
B AGcGuucGuGGGAuAGuccTT B
1588






siNA stab07 inv





306
CUGAUAGGGUGCUUGCGAGUGCC
1411
30683
HCVa:306U21 sense
B GuGAGcGuucGuGGGAuAGTT B
1589






siNA stab07 inv





324
GUGCCCCGGGAGGUCUCGUAGAC
1400
30684
HCVa:324U21 sense
B GAuGcucuGGAGGGccccGTT B
1590






siNA stab07 inv





153
AUAGUGGUCUGCGGAACCGGUGA
1401
30685
HCVa:171L21 antisense
ucAccAGAcGccuuGGccATsT
1591






siNA (153C) stab08






inv





159
GUCUGCGGAACCGGUGAGUACAC
1402
30686
HCVa:177L21 antisense

GacGccuuGGccAcucAuGTsT

1592






siNA (159C) stab08






inv





291
GCCUUGUGGUACUGCCUGAUAGG
1403
30687
HCVa:309L21 antisense

GAAcAccAuGAcGGAcuAuTsT

1593






siNA (291C) stab08






inv





295
UGUGGUACUGCCUGAUAGGGUGC
1404
30688
HCVa:313L21 antisense

AccAuGAcGGAcuAucccATsT

1594






siNA (295C) stab08






inv





296
GUGGUACUGCCUGAUAGGGUGCU
1405
30689
HCVa:314L21 antisense
ccAuGAcGGAcuAucccAcTsT
1595






siNA (296C) stab08






inv





297
UGGUACUGCCUGAUAGGGUGCUU
1406
30690
HCVa:315L21 antisense
cAuGAcGGAcuAucccAcGTsT
1596






siNA (297C) stab08






inv





298
GGUACUGCCUGAUAGGGUGCUUG
1407
30691
HCVa:316L21 antisense

AuGAcGGAcuAucccAcGATsT

1597






siNA (298C) stab08






inv





300
UACUGCCUGAUAGGGUGCUUGCG
1408
30692
HCVa:318L21 antisense

GAcGGAcuAucccAcGAAcTsT

1598






siNA (300C) stab08






inv





301
ACUGCCUGAUAGGGUGCUUGCGA
1409
30693
HCVa:319L21 antisense

AcGGAcuAucccAcGAAcGTsT

1599






siNA (301C) stab08






inv





303
UGCCUGAUAGGGUGCUUGCGAGU
1410
30694
HCVa:321L21 antisense

GGAcuAucccAcGAAcGcuTsT

1600






siNA (303C) stab08






inv





306
CUGAUAGGGUGCUUGCGAGUGCC
1411
30695
HCVa:324L21 antisense
cuAucccAcGAAcGcucAcTsT
1601






siNA (306C) stab08






inv





324
GUGCCCCGGGAGGUCUCGUAGAC
1400
30696
HCVa:342L21 antisense
cGGGGcccuccAGAGcAucTsT
1602






siNA (324C) stab08






inv





325
UGCCCCGGGAGGUCUCGUAGACC
1394
31340
HCVa:325U21 sense
B ccccGGGAGGucucGuAGATT B
1603






siNA stab04





325
UGCCCCGGGAGGUCUCGUAGACC
1394
31341
HCVa:325U21 sense
B AGAuGcucuGGAGGGccccTT B
1604






siNA inv stab04





325
UGCCCCGGGAGGUCUCGUAGACC
1394
31342
HCVa:343L21 antisense
ucuAcGAGAccucccGGGGTsT
1605






siNA (325C) stab05





325
UGCCCCGGGAGGUCUCGUAGACC
1394
31343
HCVa:343L21 antisense
GGGGcccuccAGAGcAucuTsT
1606






siNA (325C) inv






stab05





325
UGCCCCGGGAGGUCUCGUAGACC
1394
31344
HCVa:325U21 sense
B ccccGGGAGGucucGuAGATT B
1607






siNA stab07





325
UGCCCCGGGAGGUCUCGUAGACC
1394
31345
HCVa:325U21 sense
B AGAuGcucuGGAGGGccccTT B
1608






siNA inv stab07





325
UGCCCCGGGAGGUCUCGUAGACC
1394
31346
HCVa:343L21 antisense

GGGGcccuccAGAGcAucuTsT

1609






siNA (325C) inv






stab08





325
UGCCCCGGGAGGUCUCGUAGACC
1394
31347
HCVa:343L21 antisense
ucuAcGAGAccucccGGGGTsT
1610






siNA (325C) stab11





325
UGCCCCGGGAGGUCUCGUAGACC
1394
31348
HCVa:343L21 antisense

GGGGcccuccAGAGcAucuTsT

1611






siNA (325C) inv






stab11





153
AUAGUGGUCUGCGGAACCGGUGA
1401
31453
HCVa:153U21 sense
B AGuGGucuGcGGAAccGGuTT B
1612






siNA stab04





159
GUCUGCGGAACCGGUGAGUACAC
1402
31454
HCVa:159U21 sense
B cuGcGGAAccGGuGAGuAcTT B
1613






siNA stab04





287
AAAGGCCUUGUGGUACUGCCUGA
1412
31455
HCVa:287U21 sense
B AGGccuuGuGGuAcuGccuTT B
1614






siNA stab04





291
GCCUUGUGGUACUGCCUGAUAGG
1403
31456
HCVa:291U21 sense
B cuuGuGGuAcuGccuGAuATT B
1615






siNA stab04





295
UGUGGUACUGCCUGAUAGGGUGC
1404
31457
HCVa:295U21 sense
B uGGuAcuGccuGAuAGGGuTT B
1616






siNA stab04





296
GUGGUACUGCCUGAUAGGGUGCU
1405
31458
HCVa:296U21 sense
B GGuAcuGccuGAuAGGGuGTT B
1617






siNA stab04





297
UGGUACUGCCUGAUAGGGUGCUU
1406
31459
HCVa:297U21 sense
B GuAcuGccuGAuAGGGuGcTT B
1618






siNA stab04





298
GGUACUGCCUGAUAGGGUGCUUG
1407
31460
HCVa:298U21 sense
B uAcuGccuGAuAGGGuGcuTT B
1619






siNA stab04





300
UACUGCCUGAUAGGGUGCUUGCG
1408
31461
HCVa:300U21 sense
B cuGccuGAuAGGGuGcuuGTT B
1620






siNA stab04





301
ACUGCCUGAUAGGGUGCUUGCGA
1409
31462
HCVa:301U21 sense
B uGccuGAuAGGGuGcuuGcTT B
1621






siNA stab04





303
UGCCUGAUAGGGUGCUUGCGAGU
1410
31463
HCVa:303U21 sense
B ccuGAuAGGGuGcuuGcGATT B
1622






siNA stab04





306
CUGAUAGGGUGCUUGCGAGUGCC
1411
31464
HCVa:306U21 sense
B GAuAGGGuGcuuGcGAGuGTT B
1623






siNA stab04





153
AUAGUGGUCUGCGGAACCGGUGA
1401
31465
HCVa:171L21 antisense
AccGGuuccGcAGAccAcuTsT
1624






siNA (153C) stab05





159
GUCUGCGGAACCGGUGAGUACAC
1402
31466
HCVa:177L21 antisense
GuAcucAccGGuuccGcAGTsT
1625






siNA (159C) stab05





287
AAAGGCCUUGUGGUACUGCCUGA
1412
31467
HCVa:305L21 antisense
AGGcAGuAccAcAAGGccuTsT
1626






siNA (287C) stab05





291
GCCUUGUGGUACUGCCUGAUAGG
1403
31468
HCVa:309L21 antisense
uAucAGGcAGuAccAcAAGTsT
1627






siNA (291C) stab05





295
UGUGGUACUGCCUGAUAGGGUGC
1404
31469
HCVa:313L21 antisense
AcccuAucAGGcAGuAccATsT
1628






siNA (295C) stab05





296
GUGGUACUGCCUGAUAGGGUGCU
1405
31470
HCVa:314L21 antisense
cAcccuAucAGGcAGuAccTsT
1629






siNA (296C) stab05





297
UGGUACUGCCUGAUAGGGUGCUU
1406
31471
HCVa:315L21 antisense
GcAcccuAucAGGcAGuAcTsT
1630






siNA (297C) stab05





298
GGUACUGCCUGAUAGGGUGCUUG
1407
31472
HCVa:316L21 antisense
AGcACccuAucAGGcAGuATsT
1631






siNA (298C) stab05





300
UACUGCCUGAUAGGGUGCUUGCG
1408
31473
HCVa:318L21 antisense
cAAGcAcccuAucAGGcAGTsT
1632






siNA (300C) stab05





301
ACUGCCUGAUAGGGUGCUUGCGA
1409
31474
HCVa:319L21 antisense
GcAAGcAcccuAucAGGcATsT
1633






siNA (301C) stab05





303
UGCCUGAUAGGGUGCUUGCGAGU
1410
31475
HCVa:321L21 antisense
ucGcAAGcAcccuAucAGGTsT
1634






siNA (303C) stab05





306
CUGAUAGGGUGCUUGCGAGUGCC
1411
31476
HCVa:324L21 antisense
cAcucGcAAGcAcccuAucTsT
1635






siNA (306C) stab05





153
AUAGUGGUCUGCGGAACCGGUGA
1401
31477
HCVa:153U21 sense
B uGGccAAGGcGucuGGuGATT B
1636






siNA inv stab04





159
GUCUGCGGAACCGGUGAGUACAC
1402
31478
HCVa:159U21 sense
B cAuGAGuGGccAAGGcGucTT B
1637






siNA inv stab04





287
AAAGGCCUUGUGGUACUGCCUGA
1412
31479
HCVa:287U21 sense
B uccGucAuGGuGuuccGGATT B
1638






siNA inv stab04





291
GCCUUGUGGUACUGCCUGAUAGG
1403
31480
HCVa:291U21 sense
B AuAGuccGucAuGGuGuucTT B
1639






siNA inv stab04





295
UGUGGUACUGCCUGAUAGGGUGC
1404
31481
HCVa:295U21 sense
B uGGGAuAGuccGucAuGGuTT B
1640






siNA inv stab04





296
GUGGUACUGCCUGAUAGGGUGCU
1405
31482
HCVa:296U21 sense
B GuGGGAuAGuccGucAuGGTT B
1641






siNA inv stab04





297
UGGUACUGCCUGAUAGGGUGCUU
1406
31483
HCVa:297U21 sense
B cGuGGGAuAGuccGucAuGTT B
1642






siNA inv stab04





298
GGUACUGCCUGAUAGGGUGCUUG
1407
31484
HCVa:298U21 sense
B ucGuGGGAuAGuccGucAuTT B
1643






siNA inv stab04





300
UACUGCCUGAUAGGGUGCUUGCG
1408
31485
HCVa:300U21 sense
B GuucGuGGGAuAGuCcGucTT B
1644






siNA inv stab04





301
ACUGCCUGAUAGGGUGCUUGCGA
1409
31486
HCVa:301U21 sense
B cGuucGuGGGAuAGuccGuTT B
1645






siNA inv stab04





303
UGCCUGAUAGGGUGCUUGCGAGU
1410
31487
HCVa:303U21 sense
B AGcGuucGuGGGAuAGuccTT B
1646






siNA inv stab04





306
CUGAUAGGGUGCUUGCGAGUGCC
1411
31488
HCVa:306U21 sense
B GuGAGcGuucGuGGGAuAGTT B
1647






siNA inv stab04





153
AUAGUGGUCUGCGGAACCGGUGA
1401
31489
HCVa:171L21 antisense
ucAccAGAcGccuuGGccATsT
1648






siNA (153C) inv






stab05





159
GUCUGCGGAACCGGUGAGUACAC
1402
31490
HCVa:177L21 antisense
GacGccuuGGccAcucAuGTsT
1649






siNA (159C) inv






stab05





287
AAAGGCCUUGUGGUACUGCCUGA
1412
31491
HCVa:305L21 antisense
uccGGAAcAccAuGAcGGATsT
1650






siNA (287C) inv






stab05





291
GCCUUGUGGUACUGCCUGAUAGG
1403
31492
HCVa:309L21 antisense
GAAcAccAuGAcGGAcuAuTsT
1651






siNA (291C) inv






stab05





295
UGUGGUACUGCCUGAUAGGGUGC
1404
31493
HCVa:313L21 antisense
AccAuGAcGGAcuAucccATsT
1652






siNA (295C) inv






stab05





296
GUGGUACUGCCUGAUAGGGUGCU
1405
31494
HCVa:314L21 antisense
ccAuGAcGGAcuAucccAcTsT
1653






siNA (296C) inv






stab05





297
UGGUACUGCCUGAUAGGGUGCUU
1406
31495
HCVa:315L21 antisense
cAuGAcGGAcuAucccAcGTsT
1654






siNA (297C) inv






stab05





298
GGUACUGCCUGAUAGGGUGCUUG
1407
31496
HCVa:316L21 antisense
AuGAcGGAcuAucccAcGATsT
1655






siNA (298C) inv






stab05





300
UACUGCCUGAUAGGGUGCUUGCG
1408
31497
HCVa:318L21 antisense
GAcGGAcuAucccAcGAAcTsT
1656






siNA (300C) inv






stab05





301
ACUGCCUGAUAGGGUGCUUGCGA
1409
31498
HCVa:319L21 antisense
AcGGAcuAucccAcGAAcGTsT
1657






siNA (301C) inv






stab05





303
UGCCUGAUAGGGUGCUUGCGAGU
1410
31499
HCVa:321L21 antisense
GGAcuAucccAcGAAcGcuTsT
1658






siNA (303C) inv






stab05





306
CUGAUAGGGUGCUUGCGAGUGCC
1411
31500
HCVa:324L21 antisense
cuAucccAcGAAcGcucAcTsT
1659






siNA (306C) inv






stab05





190
GGGUCCUUUCUUGGAUCAACCCG
1413
31659
HCVb:190U21 sense
B GuccuuucuuGGAucAAccTT B
1660






siNA stab04





191
GGUCCUUUCUUGGAUCAACCCGC
1393
31660
HCVb:191U21 sense
B uccuuucuuGGAucAAcccTT B
1661






siNA stab04





189
CGGGUCCUUUCUUGGAUCAACCC
1414
31661
HCVb:189U21 sense
B GGuccuuucuuGGAucAAcTT B
1662






siNA stab04





186
GACCGGGUCCUUUCUUGGAUCAA
1415
31662
HCVb:186U21 sense
B ccGGGuccuuucuuGGAucTT B
1663






siNA stab04





190
GGGUCCUUUCUUGGAUCAACCCG
1413
31663
HCVb:208L21 antisense
GGuuGAuccAAGAAAGGAcTsT
1664






siNA (190C) stab05





191
GGUCCUUUCUUGGAUCAACCCGC
1393
31664
HCVb:209L21 antisense
GGGuuGAuccAAGAAAGGATsT
1665






siNA (191C) stab05





189
CGGGUCCUUUCUUGGAUCAACCC
1414
31665
HCVb:207L21 antisense
GuuGAuccAAGAAAGGAccTsT
1666






siNA (189C) stab05





186
GACCGGGUCCUUUCUUGGAUCAA
1415
31666
HCVb:204L21 antisense
GAuccAAGAAAGGAcccGGTsT
1667






siNA (186C) stab05





190
GGGUCCUUUCUUGGAUCAACCCG
1413
31667
HCVb:190U21 sense
B ccAAcuAGGuucuuuccuGTT B
1668






siNA inv stab04





191
GGUCCUUUCUUGGAUCAACCCGC
1393
31668
HCVb:191U21 sense
B cccAAcuAGGuucuuuccuTT B
1669






siNA inv stab04





189
CGGGUCCUUUCUUGGAUCAACCC
1414
31669
HCVb:189U21 sense
B cAAcuAGGuucuuuccuGGTT B
1670






siNA inv stab04





186
GACCGGGUCCUUUCUUGGAUCAA
1415
31670
HCVb:186U21 sense
B cuAGGuucuuuccuGGGccTT B
1671






siNA inv stab04





190
GGGUCCUUUCUUGGAUCAACCCG
1413
31671
HCVb:208L21 antisense
cAGGAAAGAAccuAGuuGGTsT
1672






siNA (190C) inv stab05





191
GGUCCUUUCUUGGAUCAACCCGC
1393
31672
HCVb:209L21 antisense
AGGAAAGAAccuAGuuGGGTsT
1673






siNA (191C) inv stab05





189
CGGGUCCUUUCUUGGAUCAACCC
1414
31673
HCVb:207L21 antisense
ccAGGAAAGAAccuAGuuGTsT
1674






siNA (189C) inv stab05





186
GACCGGGUCCUUUCUUGGAUCAA
1415
31674
HCVb:204L21 antisense
GGcccAGGAAAGAAccuAGTsT
1675






siNA (186C) inv stab05





326
GCCCCGGGAGGUCUCGUAGACCG
1416
31702
HCVa:326U21 sense
B cccGGGAGGucucGuAGAcTT B
1676






siNA stab07





327
CCCCGGGAGGUCUCGUAGACCGU
1417
31703
HCVa:327U21 sense
B ccGGGAGGucucGuAGAccTT B
1677






siNA stab07





328
CCCGGGAGGUCUCGUAGACCGUG
1418
31704
HCVa:328U21 sense
B cGGGAGGucucGuAGAccGTT B
1678






siNA stab07





329
CCGGGAGGUCUCGUAGACCGUGC
1419
31705
HCVa:329U21 sense
B GGGAGGucucGuAGAccGuTT B
1679






siNA stab07





326
GCCCCGGGAGGUCUCGUAGACCG
1416
31706
HCVa:344L21 antisense

GucuAcGAGAccucccGGGTsT

1680






siNA (326C) stab08





327
CCCCGGGAGGUCUCGUAGACCGU
1417
31707
HCVa:345L21 antisense

GGucuAcGAGAccucccGGTsT

1681






siNA (327C) stab08





328
CCCGGGAGGUCUCGUAGACCGUG
1418
31708
HCVa:346L21 antisense
cGGucuAcGAGAccucccGTsT
1682






siNA (328C) stab08





329
CCGGGAGGUCUCGUAGACCGUGC
1419
31709
HCVa:347L21 antisense

AcGGucuAcGAGAccucccTsT

1683






siNA (329C) stab08





326
GCCCCGGGAGGUCUCGUAGACCG
1416
31710
HCVa:326U21 sense
B cAGAuGcucuGGAGGGcccTT B
1684






siNA inv stab07





327
CCCCGGGAGGUCUCGUAGACCGU
1417
31711
HCVa:327U21 sense
B ccAGAuGcucuGGAGGGccTT B
1685






siNA inv stab07





328
CCCGGGAGGUCUCGUAGACCGUG
1418
31712
HCVa:328U21 sense
B GccAGAuGcucuGGAGGGcTT B
1686






siNA inv stab07





329
CCGGGAGGUCUCGUAGACCGUGC
1419
31713
HCVa:329U21 sense
B uGccAGAuGcucuGGAGGGTT B
1687






siNA inv stab07





326
GCCCCGGGAGGUCUCGUAGACCG
1416
31714
HCVa:344L21 antisense

GGGcccuccAGAGcAucuGTsT

1688






siNA (326C) inv






stab08





327
CCCCGGGAGGUCUCGUAGACCGU
1417
31715
HCVa:345L21 antisense

GGcccuccAGAGcAucuGGTsT

1689






siNA (327C) inv






stab08





328
CCCGGGAGGUCUCGUAGACCGUG
1418
31716
HCVa:346L21 antisense

GcccuccAGAGcAucuGGcTsT

1690






siNA (328C) inv






stab08





329
CCGGGAGGUCUCGUAGACCGUGC
1419
31717
HCVa:347L21 antisense
cccuccAGAGcAucuGGcATsT
1691






siNA (329C) inv






stab08





291
GCCUUGUGGUACUGCCUGAUAGG
1403
31762
HCVa:291U21 sense
cuuGuGGuAcuGccuGAuATsT
1692






siNA stab08





295
UGUGGUACUGCCUGAUAGGGUGC
1404
31763
HCVa:295U21 sense
uGGuAcuGccuGAuAGGGuTsT
1693






siNA stab08





325
UGCCCCGGGAGGUCUCGUAGACC
1394
31764
HCVa:325U21 sense
ccccGGGAGGucucGuAGATsT
1694






siNA stab08





291
GCCUUGUGGUACUGCCUGAUAGG
1403
31765
HCVa:291U21 sense

AuAGuccGucAuGGuGuucTsT

1695






siNA inv stab08





295
UGUGGUACUGCCUGAUAGGGUGC
1404
31766
HCVa:295U21 sense
uGGGAuAGuccGucAuGGuTsT
1696






siNA inv stab08





325
UGCCCCGGGAGGUCUCGUAGACC
1394
31767
HCVa:325U21 sense

AGAuGcucuGGAGGGccccTsT

1697






siNA inv stab08





327
CCCCGGGAGGUCUCGUAGACCGU
1417
31928
HCVa:327U21 sense
ccGGGAGGucucGuAGAccTsT
1698






siNA stab08





327
CCCCGGGAGGUCUCGUAGACCGU
1417
31929
HCVa:327U21 sense
ccAGAuGcucuGGAGGGccTsT
1699






siNA inv stab08





328
CCCGGGAGGUCUCGUAGACCGUG
1418
31930
HCVa:328U21 sense
cGGGAGGucucGuAGAccGTsT
1700






siNA stab08





328
CCCGGGAGGUCUCGUAGACCGUG
1418
31931
HCVa:328U21 sense

GccAGAuGcucuGGAGGGcTsT

1701






siNA inv stab08





327
CCCCGGGAGGUCUCGUAGACCGU
1417
32007
HCVa:327U21 sense
B ccGGGAGGucucGuAGAccTsT
1702






siNA stab08 + 5′






abasic





327
CCCCGGGAGGUCUCGUAGACCGU
1417
32008
HCVa:327U21 sense
ccGGGAGGucucGuAGAccTsT B
1703






siNA stab08 + 3′






abasic





327
CCCCGGGAGGUCUCGUAGACCGU
1417
32009
HCVa:327U21 sense
B ccGGGAGGucucGuAGAccTsT B
1704






siNA stab08 + 5′ & 3′






abasic





327
CCCCGGGAGGUCUCGUAGACCGU
1417
32174
HCVa:327 siNA
UCUCGUAGACCUU
1705






3′-classl 10 bp
GGUCUACGAGACCUCCCGGTT





327
CCCCGGGAGGUCUCGUAGACCGU
1417
32175
HCVa:327 siNA
UCGUAGACCUU
1706






3′-classl 8 bp
GGUCUACGAGACCUCCCGGTT





327
CCCCGGGAGGUCUCGUAGACCGU
1417
32176
HCVa:327 siNA
GUAGACCUU GGUCUACGAGACCUCCCGGTT
1707






3′-classl 6 bp





327
CCCCGGGAGGUCUCGUAGACCGU
1417
32177
HCVa:327 siNA
AGACCUU GGUCUACGAGACCUCCCGGTT
1708






3′-classl 4 bp





327
CCCCGGGAGGUCUCGUAGACCGU
1417
32178
HCVa:327 siNA
GGUCUACGAGACCUCCCGGUU
1709






5′-classl 10 bp
CCGGGAGGUCU





327
CCCCGGGAGGUCUCGUAGACCGU
1417
32179
HCVa:327 siNA
GGUCUACGAGACCUCCCGGUU
1710






5′-classl 8 bp
CCGGGAGGU





327
CCCCGGGAGGUCUCGUAGACCGU
1417
32180
HCVa:327 siNA
GGUCUACGAGACCUCCCGGUU CCGGGAG
1711






5′-classl 6 bp





327
CCCCGGGAGGUCUCGUAGACCGU
1417
32181
HCVa:327 siNA
GGUCUACGAGACCUCCCGGUU CCGGG
1712






5′-classl 4 bp





327
CCCCGGGAGGUCUCGUAGACCGU
1417
32182
HCVa:327 siNA
CUCGUAGACC GAAA
1713






3′-gaaa 10 bp
GGUCUACGAGACCUCCCGGTT





327
CCCCGGGAGGUCUCGUAGACCGU
1417
32183
HCVa:327 siNA
CGUAGACC GAAA
1714






3′-gaaa 8 bp
GGUCUACGAGACCUCCCGGTT





327
CCCCGGGAGGUCUCGUAGACCGU
1417
32184
HCVa:327 siNA
UAGACC GAAA
1715






3′-gaaa 6 bp
GGUCUACGAGACCUCCCGGTT





327
CCCCGGGAGGUCUCGUAGACCGU
1417
32185
HCVa:327 siNA
GACC GAAA GGUCUACGAGACCUCCCGGTT
1716






3′-gaaa 4 bp





327
CCCCGGGAGGUCUCGUAGACCGU
1417
32186
HCVa:327 siNA
GGUCUACGAGACCUCCCGGUU GAAA
1717






5′-gaaa 10 bp
CCGGGAGGUC





327
CCCCGGGAGGUCUCGUAGACCGU
1417
32187
HCVa:327 siNA
GGUCUACGAGACCUCCCGGUU GAAA
1718






5′-gaaa 8 bp
CCGGGAGG





327
CCCCGGGAGGUCUCGUAGACCGU
1417
32188
HCVa:327 siNA
GGUCUACGAGACCUCCCGGUU GAAA
1719






5′-gaaa 6 bp
CCGGGA





327
CCCCGGGAGGUCUCGUAGACCGU
1417
32189
HCVa:327 siNA
GGUCUACGAGACCUCCCGGUU GAAA CCGG
1720






5′-gaaa 4 bp





327
CCCCGGGAGGUCUCGUAGACCGU
1417
32190
HCVa:327 siNA
CGUAGACCUU UUUGUGUAG
1721






3′-uuuguguag 10 bp
GGUCUACGAGACCUCCCGGTT





327
CCCCGGGAGGUCUCGUAGACCGU
1417
32191
HCVa:327 siNA
UAGACCUU UUUGUGUAG
1722






3′-uuuguguag 8 bp
GGUCUACGAGACCUCCCGGTT





327
CCCCGGGAGGUCUCGUAGACCGU
1417
32192
HCVa:327 siNA
GACCUU UUUGUGUAG
1723






3′-uuuguguag 6 bp
GGUCUACGAGACCUCCCGGTT





327
CCCCGGGAGGUCUCGUAGACCGU
1417
32193
HCVa:327 siNA
CCUU UUUGUGUAG
1724






3′-uuuguguag 4 bp
GGUCUACGAGACCUCCCGGTT





327
CCCCGGGAGGUCUCGUAGACCGU
1417
32194
HCVa:327 siNA
GGUCUACGAGACCUCCCGGUU UUUGUGUAG
1725






5′-uuuguguag 10 bp
CCGGGAGGUC





327
CCCCGGGAGGUCUCGUAGACCGU
1417
32195
HCVa:327 siNA
GGUCUACGAGACCUCCCGGUU UUUGUGUAG
1726






5′-uuuguguag 8 bp
CCGGGAGG





327
CCCCGGGAGGUCUCGUAGACCGU
1417
32196
HCVa:327 siNA
GGUCUACGAGACCUCCCGGUU UUUGUGUAG
1727






5′-uuuguguag 6 bp
CCGGGA





327
CCCCGGGAGGUCUCGUAGACCGU
1417
32197
HCVa:327 siNA
GGUCUACGAGACCUCCCGGUU UUUGUGUAG
1728






5′-uuuguguag 4 bp
CCGG





327
CCCCGGGAGGUCUCGUAGACCGU
1417
32198
HCVa:345L21 antisense
GGucuAcGAGAccucccGGTsT
1729






(327C) stab05 siNA





327
CCCCGGGAGGUCUCGUAGACCGU
1417
32199
HCVa:345L21 antisense
pGGucuAcGAGAccucccGGTsT
1730






(327C) stab05 5′p






siNA





327
CCCCGGGAGGUCUCGUAGACCGU
1417
32200
HCVa:345L21 antisense
sGGucuAcGAGAccucccGGTsT
1731






(327C) stab05 5′ps






siNA





327
CCCCGGGAGGUCUCGUAGACCGU
1417
32201
HCVa:345L21 antisense
GGUCUACGAGACCUCCCGGTT
1732






(327C) stab00 siNA





327
CCCCGGGAGGUCUCGUAGACCGU
1417
32202
HCVa:345L21 antisense
pGGUCUACGAGACCUCCCGGTT
1733






(327C) v1 5′p siNA





327
CCCCGGGAGGUCUCGUAGACCGU
1417
32203
HCVa:345L21 antisense
sGGUCUACGAGACCUCCCGGTT
1734






(327C) v1 5′ps siNA





327
CCCCGGGAGGUCUCGUAGACCGU
1417
32204
HCVa:345L21 antisense
pGGUCUACGAGACCUCCCGGGGTT
1735






(327C) v2 5′p siNA





327
CCCCGGGAGGUCUCGUAGACCGU
1417
32205
HCVa:345L21 antisense
pGGUCUACGAGACCUCCCGG UCUCGUA u B
1736






(327C) v3 5′p siNA





327
CCCCGGGAGGUCUCGUAGACCGU
1417
32206
HCVa:345L21 antisense
pGGUCUACGAGACCUCCCGG AGGUCUCGUA
1737






(327C) v4 5′p siNA
uu B





327
CCCCGGGAGGUCUCGUAGACCGU
1417
32207
HCVa:345L21 antisense
pGGUCUACGAGACCUCCCGGTT UCUCGUA u
1738






(327C) v5 5′p siNA
B





327
CCCCGGGAGGUCUCGUAGACCGU
1417
32208
HCVa:345L21 antisense
pGGUCUACGAGACCUCCCGGTT
1739






(327C) v6 5′p siNA
AGGUCUCGUA u B





327
CCCCGGGAGGUCUCGUAGACCGU
1417
32501
HCVa:327U21 sense
B ccGGGAGGucucGuAGAccTT B
1740






siNA stab04





325
UGCCCCGGGAGGUCUCGUAGACC
1394
32502
HCVa:325U21 sense
B CCCCGGGAGGUCUCGUAGATT B
1741






siNA stab09





326
GCCCCGGGAGGUCUCGUAGACCG
1416
32503
HCVa:326U21 sense
B CCCGGGAGGUCUCGUAGACTT B
1742






siNA stab09





327
CCCCGGGAGGUCUCGUAGACCGU
1417
32504
HCVa:327U21 sense
B CCGGGAGGUCUCGUAGACCTT B
1743






siNA stab09





328
CCCGGGAGGUCUCGUAGACCGUG
1418
32505
HCVa:328U21 sense
B CGGGAGGUCUCGUAGACCGTT B
1744






siNA stab09





329
CCGGGAGGUCUCGUAGACCGUGC
1419
32506
HCVa:329U21 sense
B GGGAGGUCUCGUAGACCGUTT B
1745






siNA stab09





325
UGCCCCGGGAGGUCUCGUAGACC
1394
32507
HCVa:343L21 antisense
UCUACGAGACCUCCCGGGGTsT
1746






siNA (325C) stab10





326
GCCCCGGGAGGUCUCGUAGACCG
1416
32508
HCVa:344L21 antisense
GUCUACGAGACCUCCCGGGTsT
1747






siNA (326C) stab10





327
CCCCGGGAGGUCUCGUAGACCGU
1417
32509
HCVa:345L21 antisense
GGUCUACGAGACCUCCCGGTsT
1748






siNA (327C) stab10





328
CCCGGGAGGUCUCGUAGACCGUG
1418
32510
HCVa:346L21 antisense
CGGUCUACGAGACCUCCCGTsT
1749






siNA (328C) stab10





329
CCGGGAGGUCUCGUAGACCGUGC
1419
32511
HCVa:347L21 antisense
ACGGUCUACGAGACCUCCCTsT
1750






siNA (329C) stab10





327
CCCCGGGAGGUCUCGUAGACCGU
1417
32512
HCVa:327U21 sense
B ccAGAuGcucuGGAGGGccTT B
1751






siNA inv stab04





327
CCCCGGGAGGUCUCGUAGACCGU
1417
32513
HCVa:345L21 antisense
GGcccuccAGAGcAucuGGTsT
1752






siNA (327C) inv






stab05





325
UGCCCCGGGAGGUCUCGUAGACC
1394
32514
HCVa:325U21 sense
B AGAUGCUCUGGAGGGCCCCTT B
1753






siNA inv stab09





326
GCCCCGGGAGGUCUCGUAGACCG
1416
32515
HCVa:326U21 sense
B CAGAUGCUCUGGAGGGCCCTT B
1754






siNA inv stab09





327
CCCCGGGAGGUCUCGUAGACCGU
1417
32516
HCVa:327U21 sense
B CCAGAUGCUCUGGAGGGCCTT B
1755






siNA inv stab09





328
CCCGGGAGGUCUCGUAGACCGUG
1418
32517
HCVa:328U21 sense
B GCCAGAUGCUCUGGAGGGCTT B
1756






siNA inv stab09





329
CCGGGAGGUCUCGUAGACCGUGC
1419
32518
HCVa:329U21 sense
B UGCCAGAUGCUCUGGAGGGTT B
1757






siNA inv stab09





325
UGCCCCGGGAGGUCUCGUAGACC
1394
32519
HCVa:343L21 antisense
GGGGCCCUCCAGAGCAUCUTsT
1758






siNA (325C) inv






stab10





326
GCCCCGGGAGGUCUCGUAGACCG
1416
32520
HCVa:344L21 antisense
GGGCCCUCCAGAGCAUCUGTsT
1759






siNA (326C) inv






stab10





327
CCCCGGGAGGUCUCGUAGACCGU
1417
32521
HCVa:345L21 antisense
GGCCCUCCAGAGCAUCUGGTsT
1760






siNA (327C) inv






stab10





328
CCCGGGAGGUCUCGUAGACCGUG
1418
32522
HCVa:346L21 antisense
GCCCUCCAGAGCAUCUGGCTsT
1761






siNA (328C) inv






stab10





329
CCGGGAGGUCUCGUAGACCGUGC
1419
32523
HCVa:347L21 antisense
CCCUCCAGAGCAUCUGGCATsT
1762






siNA (329C) inv






stab10





295
UGUGGUACUGCCUGAUAGGGUGC
1404
32714
HCVa:313L21 antisense
pACCCUAUCAGGCAGUACCA
1763






siNA (295C) v1 5′p
GUACUGCCUGAU B






palindrome





295
UGUGGUACUGCCUGAUAGGGUGC
1404
32715
HCVa:313L21 antisense
pACCCUAUCAGGCAGUACC
1764






siNA (295C) v2 5′p
GGUACUGCCUGAU B






palindrome





327
CCCCGGGAGGUCUCGUAGACCGU
1417
32716
HCVa 5′p-345L21
pGGUCUACGAGACCUCCCGG
1765






antisense (327C) v5
AGGUCUCGUAGA B






5′p palindrome siNA





327
CCCCGGGAGGUCUCGUAGACCGU
1417
32717
HCVa 5′p-345L21
pGGUCUACGAGACCUCC GGAGGUCUCGUA B
1766






antisense (327C) v6






5′p palindrome siNA





291
GCCUUGUGGUACUGCCUGAUAGG
1403
32796
HCVa:309L21 antisense
uAucAgGcaguaccAcaAgTsT
1767






siNA (291C) stab08






mod pair to #30651





295
UGUGGUACUGCCUGAUAGGGUGC
1404
32797
HCVa:313L21 antisense
acccuaucaggcaguAccaTsT
1768






siNA (295C) stab08






mod pair to #30652





303
UGCCUGAUAGGGUGCUUGCGAGU
1410
32798
HCVa:321L21 antisense
ucgcaaGcacccuAucaggTsT
1769






siNA (303C) stab08






mod pair to #30658





306
CUGAUAGGGUGCUUGCGAGUGCC
1411
32799
HCVa:324L21 antisense
cacucgcAagcacccuaucTsT
1770






siNA (306C) stab08






mod A pair to #30659





306
CUGAUAGGGUGCUUGCGAGUGCC
1411
32800
HCVa:324L21 antisense
cAcucgcAagcacccuaucTsT
1771






siNA (306C) stab08






mod B pair to #30659





140
UCCCGGGAGAGCCAUAGUGGUCU
1420
33125
HCVa:140U21 sense
B ccGGGAGAGccAuAGuGGuTT B
1772






siNA stab07





141
CCCGGGAGAGCCAUAGUGGUCUG
1421
33126
HCVa:141U21 sense
B cGGGAGAGccAuAGuGGucTT B
1773






siNA stab07





142
CCGGGAGAGCCAUAGUGGUCUGC
1422
33127
HCVa:142U21 sense
B GGGAGAGccAuAGuGGucuTT B
1774






siNA stab07





154
UAGUGGUCUGCGGAACCGGUGAG
1423
33128
HCVa:154U21 sense
B GuGGucuGcGGAAccGGuGTT B
1775






siNA stab07





155
AGUGGUCUGCGGAACCGGUGAGU
1424
33129
HCVa:155U21 sense
B uGGucuGcGGAAccGGuGATT B
1776






siNA stab07





156
GUGGUCUGCGGAACCGGUGAGUA
1425
33130
HCVa:156U21 sense
B GGucuGcGGAAccGGuGAGTT B
1777






siNA stab07





157
UGGUCUGCGGAACCGGUGAGUAC
1426
33131
HCVa:157U21 sense
B GucuGcGGAAccGGuGAGuTT B
1778






siNA stab07





158
GGUCUGCGGAACCGGUGAGUACA
1427
33132
HCVa:158U21 sense
B ucuGcGGAAccGGuGAGuATT B
1779






siNA stab07





160
UCUGCGGAACCGGUGAGUACACC
1428
33133
HCVa:160U21 sense
B uGcGGAAccGGuGAGuAcATT B
1780






siNA stab07





161
CUGCGGAACCGGUGAGUACACCG
1429
33134
HCVa:161U21 sense
B GcGGAAccGGuGAGuAcAcTT B
1781






siNA stab07





164
CGGAACCGGUGAGUACACCGGAA
1430
33135
HCVa:164U21 sense
B GAAccGGuGAGuAcAccGGTT B
1782






siNA stab07





165
GGAACCGGUGAGUACACCGGAAU
1431
33136
HCVa:165U21 sense
B AAccGGuGAGuAcAccGGATT B
1783






siNA stab07





166
GAACCGGUGAGUACACCGGAAUU
1432
33137
HCVa:166U21 sense
B AccGGuGAGuAcAccGGAATT B
1784






siNA stab07





167
AACCGGUGAGUACACCGGAAUUG
1433
33138
HCVa:167U21 sense
B ccGGuGAGuAcAccGGAAuTT B
1785






siNA stab07





282
UCGCGAAAGGCCUUGUGGUACUG
1434
33139
HCVa:282U21 sense
B GcGAAAGGccuuGuGGuAcTT B
1786






siNA stab07





283
CGCGAAAGGCCUUGUGGUACUGC
1435
33140
HCVa:283U21 sense
B cGAAAGGccuuGuGGuAcuTT B
1787






siNA stab07





284
GCGAAAGGCCUUGUGGUACUGCC
1436
33141
HCVa:284U21 sense
B GAAAGGccuuGuGGuAcuGTT B
1788






siNA stab07





285
CGAAAGGCCUUGUGGUACUGCCU
1437
33142
HCVa:285U21 sense
B AAAGGccuuGuGGuAcuGcTT B
1789






siNA stab07





286
GAAAGGCCUUGUGGUACUGCCUG
1438
33143
HCVa:286U21 sense
B AAGGccuuGuGGuAcuGccTT B
1790






siNA stab07





288
AAGGCCUUGUGGUACUGCCUGAU
1439
33144
HCVa:288U21 sense
B GGccuuGuGGuAcuGccuGTT B
1791






siNA stab07





289
AGGCCUUGUGGUACUGCCUGAUA
1440
33145
HCVa:289U21 sense
B GccuuGuGGuAcuGccuGATT B
1792






siNA stab07





290
GGCCUUGUGGUACUGCCUGAUAG
1441
33146
HCVa:290U21 sense
B ccuuGuGGuAcuGccuGAuTT B
1793






siNA stab07





299
GUACUGCCUGAUAGGGUGCUUGC
1442
33147
HCVa:299U21 sense
B AcuGccuGAuAGGGuGcuuTT B
1794






siNA stab07





302
CUGCCUGAUAGGGUGCUUGCGAG
1443
33148
HCVa:302U21 sense
B GccuGAuAGGGuGcuuGcGTT B
1795






siNA stab07





304
GCCUGAUAGGGUGCUUGCGAGUG
1444
33149
HCVa:304U21 sense
B cuGAuAGGGuGcuuGcGAGTT B
1796






siNA stab07





305
CCUGAUAGGGUGCUUGCGAGUGC
1445
33150
HCVa:305U21 sense
B uGAuAGGGuGcuuGcGAGuTT B
1797






siNA stab07





307
UGAUAGGGUGCUUGCGAGUGCCC
1446
33151
HCVa:307U21 sense
B AuAGGGuGcuuGcGAGuGcTT B
1798






siNA stab07





308
GAUAGGGUGCUUGCGAGUGCCCC
1447
33152
HCVa:308U21 sense
B uAGGGuGcuuGcGAGuGccTT B
1799






siNA stab07





310
UAGGGUGCUUGCGAGUGCCCCGG
1448
33153
HCVa:310U21 sense
B GGGuGcuuGcGAGuGccccTT B
1800






siNA stab07





311
AGGGUGCUUGCGAGUGCCCCGGG
1449
33154
HCVa:311U21 sense
B GGuGcuuGcGAGuGccccGTT B
1801






siNA stab07





314
GUGCUUGCGAGUGCCCCGGGAGG
1450
33155
HCVa:314U21 sense
B GcuuGcGAGuGccccGGGATT B
1802






siNA stab07





315
UGCUUGCGAGUGCCCCGGGAGGU
1451
33156
HCVa:315U21 sense
B cuuGcGAGuGccccGGGAGTT B
1803






siNA stab07





316
GCUUGCGAGUGCCCCGGGAGGUC
1452
33157
HCVa:316U21 sense
B uuGcGAGuGccccGGGAGGTT B
1804






siNA stab07





317
CUUGCGAGUGCCCCGGGAGGUCU
1453
33158
HCVa:317U21 sense
B uGcGAGuGccccGGGAGGuTT B
1805






siNA stab07





318
UUGCGAGUGCCCCGGGAGGUCUC
1454
33159
HCVa:318U21 sense
B GcGAGuGccccGGGAGGucTT B
1806






siNA stab07





319
UGCGAGUGCCCCGGGAGGUCUCG
1455
33160
HCVa:319U21 sense
B cGAGuGccccGGGAGGucuTT B
1807






siNA stab07





320
GCGAGUGCCCCGGGAGGUCUCGU
1456
33161
HCVa:320U21 sense
B GAGuGccccGGGAGGucucTT B
1808






siNA stab07





322
GAGUGCCCCGGGAGGUCUCGUAG
1457
33162
HCVa:322U21 sense
B GuGccccGGGAGGucucGuTT B
1809






siNA stab07





323
AGUGCCCCGGGAGGUCUCGUAGA
1458
33163
HCVa:323U21 sense
B uGccccGGGAGGucucGuATT B
1810






siNA stab07





330
CGGGAGGUCUCGUAGACCGUGCA
1459
33164
HCVa:330U21 sense
B GGAGGucucGuAGAccGuGTT B
1811






siNA stab07





140
UCCCGGGAGAGCCAUAGUGGUCU
1420
33165
HCVa:158L21 antisense

AccAcuAuGGcucucccGGTsT

1812






siNA (140C) stab08





141
CCCGGGAGAGCCAUAGUGGUCUG
1421
33166
HCVa:159L21 antisense

GaccAcuAuGGcucucccGTsT

1813






siNA (141C) stab08





142
CCGGGAGAGCCAUAGUGGUCUGC
1422
33167
HCVa:160L21 antisense

AGAccAcuAuGGcucucccTsT

1814






siNA (142C) stab08





154
UAGUGGUCUGCGGAACCGGUGAG
1423
33168
HCVa:172L21 antisense
cAccGGuuccGcAGAccAcTsT
1815






siNA (154C) stab08





155
AGUGGUCUGCGGAACCGGUGAGU
1424
33169
HCVa:173L21 antisense
ucAccGGuuccGcAGAccATsT
1816






siNA (155C) stab08





156
GUGGUCUGCGGAACCGGUGAGUA
1425
33170
HCVa:174L21 antisense
cucAccGGuuccGcAGAccTsT
1817






siNA (156C) stab08





157
UGGUCUGCGGAACCGGUGAGUAC
1426
33171
HCVa:175L21 antisense

AcucAccGGuuccGcAGAcTsT

1818






siNA (157C) stab08





158
GGUCUGCGGAACCGGUGAGUACA
1427
33172
HCVa:176L21 antisense
uAcucAccGGuuccGcAGATsT
1819






siNA (158C) stab08





160
UCUGCGGAACCGGUGAGUACACC
1428
33173
HCVa:178L21 antisense
uGuAcucAccGGuuccGcATsT
1820






siNA (160C) stab08





161
CUGCGGAACCGGUGAGUACACCG
1429
33174
HCVa:179L21 antisense

GuGuAcucAccGGuuccGcTsT

1821






siNA (161C) stab08





164
CGGAACCGGUGAGUACACCGGAA
1430
33175
HCVa:182L21 antisense
ccGGuGuAcucAccGGuucTsT
1822






siNA (164C) stab08





165
GGAACCGGUGAGUACACCGGAAU
1431
33176
HCVa:183L21 antisense
uccGGuGuAcucAccGGuuTsT
1823






siNA (165C) stab08





166
GAACCGGUGAGUACACCGGAAUU
1432
33177
HCVa:184L21 antisense
uuccGGuGuAcucAccGGuTsT
1824






siNA (166C) stab08





167
AACCGGUGAGUACACCGGAAUUG
1433
33178
HCVa:185L21 antisense

AuuccGGuGuAcucAccGGTsT

1825






siNA (167C) stab08





282
UCGCGAAAGGCCUUGUGGUACUG
1434
33179
HCVa:300L21 antisense

GuAccAcAAGGccuuucGcTsT

1826






siNA (282C) stab08





283
CGCGAAAGGCCUUGUGGUACUGC
1435
33180
HCVa:301L21 antisense

AGuAccAcAAGGccuuucGTsT

1827






siNA (283C) stab08





284
GCGAAAGGCCUUGUGGUACUGCC
1436
33181
HCVa:302L21 antisense
cAGuAccAcAAGGccuuucTsT
1828






siNA (284C) stab08





285
CGAAAGGCCUUGUGGUACUGCCU
1437
33182
HCVa:303L21 antisense

GcAGuAccAcAAGGccuuuTsT

1829






siNA (285C) stab08





286
GAAAGGCCUUGUGGUACUGCCUG
1438
33183
HCVa:304L21 antisense

GGcAGuAccAcAAGGccuuTsT

1830






siNA (286C) stab08





288
AAGGCCUUGUGGUACUGCCUGAU
1439
33184
HCVa:306L21 antisense
cAGGcAGuAccAcAAGGccTsT
1831






siNA (288C) stab08





289
AGGCCUUGUGGUACUGCCUGAUA
1440
33185
HCVa:307L21 antisense
ucAGGcAGuAccAcAAGGcTsT
1832






siNA (289C) stab08





290
GGCCUUGUGGUACUGCCUGAUAG
1441
33186
HCVa:308L21 antisense

AucAGGcAGuAccAcAAGGTsT

1833






siNA (290C) stab08





299
GUACUGCCUGAUAGGGUGCUUGC
1442
33187
HCVa:317L21 antisense

AAGcAcccuAucAGGcAGuTsT

1834






siNA (299C) stab08





302
CUGCCUGAUAGGGUGCUUGCGAG
1443
33188
HCVa:320L21 antisense
cGcAAGcAcccuAucAGGcTsT
1835






siNA (302C) stab08





304
GCCUGAUAGGGUGCUUGCGAGUG
1444
33189
HCVa:322L21 antisense
cucGcAAGcAcccuAucAGTsT
1836






siNA (304C) stab08





305
CCUGAUAGGGUGCUUGCGAGUGC
1445
33190
HCVa:323L21 antisense

AcucGcAAGcAcccuAucATsT

1837






siNA (305C) stab08





307
UGAUAGGGUGCUUGCGAGUGCCC
1446
33191
HCVa:325L21 antisense

GcAcucGcAAGcAcccuAuTsT

1838






siNA (307C) stab08





308
GAUAGGGUGCUUGCGAGUGCCCC
1447
33192
HCVa:326L21 antisense

GGcAcucGcAAGcAcccuATsT

1839






siNA (308C) stab08





310
UAGGGUGCUUGCGAGUGCCCCGG
1448
33193
HCVa:328L21 antisense

GGGGcAcucGcAAGcAcccTsT

1840






siNA (310C) stab08





311
AGGGUGCUUGCGAGUGCCCCGGG
1449
33194
HCVa:329L21 antisense
cGGGGcAcucGcAAGcAccTsT
1841






siNA (311C) stab08





314
GUGCUUGCGAGUGCCCCGGGAGG
1450
33195
HCVa:332L21 antisense
ucccGGGGcAcucGcAAGcTsT
1842






siNA (314C) stab08





315
UGCUUGCGAGUGCCCCGGGAGGU
1451
33196
HCVa:333L21 antisense
cucccGGGGcAcucGcAAGTsT
1843






siNA (315C) stab08





316
GCUUGCGAGUGCCCCGGGAGGUC
1452
33197
HCVa:334L21 antisense
ccucccGGGGcAcucGcAATsT
1844






siNA (316C) stab08





317
CUUGCGAGUGCCCCGGGAGGUCU
1453
33198
HCVa:335L21 antisense

AccucccGGGGcAcucGcATsT

1845






siNA (317C) stab08





318
UUGCGAGUGCCCCGGGAGGUCUC
1454
33199
HCVa:336L21 antisense

GAccucccGGGGcAcucGcTsT

1846






siNA (318C) stab08





319
UGCGAGUGCCCCGGGAGGUCUCG
1455
33200
HCVa:337L21 antisense

AGAccucccGGGGcAcucGTsT

1847






siNA (319C) stab08





320
GCGAGUGCCCCGGGAGGUCUCGU
1456
33201
HCVa:338L21 antisense

GAGAccucccGGGGcAcucTsT

1848






siNA (320C) stab08





322
GAGUGCCCCGGGAGGUCUCGUAG
1457
33202
HCVa:340L21 antisense

AcGAGAccucccGGGGcAcTsT

1849






siNA (322C) stab08





323
AGUGCCCCGGGAGGUCUCGUAGA
1458
33203
HCVa:341L21 antisense
uAcGAGAccucccGGGGcATsT
1850






siNA (323C) stab08





330
CGGGAGGUCUCGUAGACCGUGCA
1459
33204
HCVa:348L21 antisense
cAcGGucuAcGAGAccuccTsT
1851






siNA (330C) stab08





303
UGCCUGAUAGGGUGCUUGCGAGU
1410
33329
HCVa:321L21 antisense
pucGcAAGcAcccuAucAGGTsT
1852






siNA (303C) stab08 +






5′ P





303
UGCCUGAUAGGGUGCUUGCGAGU
1410
33330
HCVa:321L21 antisense
pucGcAAGcAcccuAucAGGTsT
1853






siNA (303C) stab05 +






5′ P





295
UGUGGUACUGCCUGAUAGGGUGC
1404
33331
HCVa:313L21 antisense
pAcccuAucAGGcAGuAccATsT
1854






siNA (295C) stab05 +






5′ P





295
UGUGGUACUGCCUGAUAGGGUGC
1404
33332
HCVa:313L21 antisense
pAcccuAucAGGcAGuAccATsT
1855






siNA (295C) stab08 +






5′ P





306
CUGAUAGGGUGCUUGCGAGUGCC
1411
33333
HCVa:324L21 antisense
pcAcucGcAAGcAcccuAucTsT
1856






siNA (306C) stab08 +






5′ P





327
CCCCGGGAGGUCUCGUAGACCGU
1417
33334
HCVa:345L21 antisense
pGGucuAcGAGAccucccGGTsT
1857






siNA (327C) stab08 +






5′ P





303
UGCCUGAUAGGGUGCUUGCGAGU
1410
33346
HCVa:321L21 antisense
L ucGcAAGcAcccuAucAGGTsT
1858






siNA (303C) stab08 +






5′ aminoL





303
UGCCUGAUAGGGUGCUUGCGAGU
1410
33347
HCVa:321L21 antisense
L ucGcAAGcAcccuAucAGGTsT
1859






siNA (303C) stab05 +






5′ aminoL





295
UGUGGUACUGCCUGAUAGGGUGC
1404
33348
HCVa:313L21 antisense
L AcccuAucAGGcAGuAccATsT
1860






siNA (295C) stab05 +






5′ aminoL





295
UGUGGUACUGCCUGAUAGGGUGC
1404
33349
HCVa:313L21 antisense
L AcccuAucAGGcAGuAccATsT
1861






siNA (295C) stab08 +






5′ aminoL





306
CUGAUAGGGUGCUUGCGAGUGCC
1411
33350
HCVa:324L21 antisense
L cAcucGcAAGcAcccuAucTsT
1862






siNA (306C) stab08 +






5′ aminoL





327
CCCCGGGAGGUCUCGUAGACCGU
1417
33351
HCVa:345L21 antisense
L GGucuAcGAGAccucccGGTsT
1863






siNA (327C) stab08 +






5′ aminoL





327
CCCCGGGAGGUCUCGUAGACCGU
1417
34024
HCVa:327U21 sense
B ccGAGAGGucGcGuAGuccTT B
1864






siNA inact1 stab07





327
CCCCGGGAGGUCUCGUAGACCGU
1417
34025
HCVa:327U21 sense
B ccGAGAGGucGcGucGAucTT B
1865






siNA inact2 stab07





327
CCCCGGGAGGUCUCGUAGACCGU
1417
34026
HCVa:327U21 sense
B ccGGuAGGucccGuGGAcATT B
1866






siNA inact3 stab07





327
CCCCGGGAGGUCUCGUAGACCGU
1417
34027
HCVa:345L21 antisense

GGAcuAcGcGAccucucGGTsT

1867






siNA (327C) inact1






stab08





327
CCCCGGGAGGUCUCGUAGACCGU
1417
34028
HCVa:345L21 antisense

GAucGAcGcGAccucucGGTsT

1868






siNA (327C) inact2






stab08





327
CCCCGGGAGGUCUCGUAGACCGU
1417
34029
HCVa:345L21 antisense
uGuccAcGGGAccuAccGGTsT
1869






siNA (327C) inact3






stab08





282
UCGCGAAAGGCCUUGUGGUACUG
1434
34030
HCVa:282U21 sense
B GcuAAAGGcGuuGuGGcAcTT B
1870






siNA inact1 stab07





282
UCGCGAAAGGCCUUGUGGUACUG
1434
34031
HCVa:282U21 sense
B GcGuAAGGcccuGuGGuAATT B
1871






siNA inact2 stab07





282
UCGCGAAAGGCCUUGUGGUACUG
1434
34032
HCVa:282U21 sense
B GAGAAAcGccuGGuGGuucTT B
1872






siNA inact3 stab07





283
CGCGAAAGGCCUUGUGGUACUGC
1435
34033
HCVa:283U21 sense
B cGuAAGGcAuuGuGGcAcuTT B
1873






siNA inact1 stab07





283
CGCGAAAGGCCUUGUGGUACUGC
1435
34034
HCVa:283U21 sense
B cGAGAGGcAuuGuGcuAcuTT B
1874






siNA inact2 stab07





283
CGCGAAAGGCCUUGUGGUACUGC
1435
34035
HCVa:283U21 sense
B ccAAAGGucuuGAGGuGcuTT B
1875






siNA inact3 stab07





304
GCCUGAUAGGGUGCUUGCGAGUG
1444
34036
HCVa:304U21 sense
B cGGAuAGGcuGcuuGuGAGTT B
1876






siNA inact1 stab07





304
GCCUGAUAGGGUGCUUGCGAGUG
1444
34037
HCVa:304U21 sense
B cuGcuAGGGuAcuuGGGAGTT B
1877






siNA inact2 stab07





304
GCCUGAUAGGGUGCUUGCGAGUG
1444
34038
HCVa:304U21 sense
B ccGAuAuGGuGAuuGcGGGTT B
1878






siNA inact3 stab07





307
UGAUAGGGUGCUUGCGAGUGCCC
1446
34039
HCVa:307U21 sense
B AuuGGGuGcuGGcGAGuAcTT B
1879






siNA inact1 stab07





307
UGAUAGGGUGCUUGCGAGUGCCC
1446
34040
HCVa:307U21 sense
B AuAuGGuGccuGcGAGuGGTT B
1880






siNA inact2 stab07





307
UGAUAGGGUGCUUGCGAGUGCCC
1446
34041
HCVa:307U21 sense
B AGAGGGuAcuuGcGcGuGuTT B
1881






siNA inact3 stab07





282
UCGCGAAAGGCCUUGUGGUACUG
1434
34042
HCVa:300L21 antisense

GuGccAcAAcGccuuuAGcTsT

1882






siNA (282C) inact1






stab08





282
UCGCGAAAGGCCUUGUGGUACUG
1434
34043
HCVa:300L21 antisense
uuAccAcAGGGccuuAcGcTsT
1883






siNA (282C) inact2






stab08





282
UCGCGAAAGGCCUUGUGGUACUG
1434
34044
HCVa:300L21 antisense

GAAccAccAGGcGuuucucTsT

1884






siNA (282C) inact3






stab08





283
CGCGAAAGGCCUUGUGGUACUGC
1435
34045
HCVa:301L21 antisense

AguGccAcAAuGccuuAcGTsT

1885






siNA (283C) inact1






stab08





283
CGCGAAAGGCCUUGUGGUACUGC
1435
34046
HCVa:301L21 antisense

AguAGcAcAAuGccucucGTsT

1886






siNA (283C) inact2






stab08





283
CGCGAAAGGCCUUGUGGUACUGC
1435
34047
HCVa:301L21 antisense

AGcAccucAAGAccuuuGGTsT

1887






siNA (283C) inact3






stab08





304
GCCUGAUAGGGUGCUUGCGAGUG
1444
34048
HCVa:322L21 antisense
cucAcAAGcAGccuAuccGTsT
1888






siNA (304C) inact1






stab08





304
GCCUGAUAGGGUGCUUGCGAGUG
1444
34049
HCVa:322L21 antisense
cucccAAGuAcccuAGcAGTsT
1889






siNA (304C) inact2






stab08





304
GCCUGAUAGGGUGCUUGCGAGUG
1444
34050
HCVa:322L21 antisense
cccGcAAucAccAuAucGGTsT
1890






siNA (304C) inact3






stab08





307
UGAUAGGGUGCUUGCGAGUGCCC
1446
34051
HCVa:325L21 antisense

GuAcucGccAGcAcccAAuTsT

1891






siNA (307C) inact1






stab08





307
UGAUAGGGUGCUUGCGAGUGCCC
1446
34052
HCVa:325L21 antisense
ccAcucGcAGGcAccAuAuTsT
1892






siNA (307C) inact2






stab08





307
UGAUAGGGUGCUUGCGAGUGCCC
1446
34053
HCVa:325L21 antisense

AcAcGcGcAAGuAcccucuTsT

1893






siNA (307C) inact3






stab08





282
UCGCGAAAGGCCUUGUGGUACUG
1434
34054
HCVa:282U21 sense
B cAuGGuGuuccGGAAAGcGTT B
1894






siNA inv stab07





283
CGCGAAAGGCCUUGUGGUACUGC
1435
34055
HCVa:283U21 sense
B ucAuGGuGuuccGGAAAGcTT B
1895






siNA inv stab07





304
GCCUGAUAGGGUGCUUGCGAGUG
1444
34056
HCVa:304U21 sense
B GAGcGuucGuGGGAuAGucTT B
1896






siNA inv stab07





307
UGAUAGGGUGCUUGCGAGUGCCC
1446
34057
HCVa:307U21 sense
B cGuGAGcGuucGuGGGAuATT B
1897






siNA inv stab07





282
UCGCGAAAGGCCUUGUGGUACUG
1434
34058
HCVa:300L21 antisense
cGcuuuccGGAAcAccAuGTsT
1898






siNA (282C) inv






stab08





283
CGCGAAAGGCCUUGUGGUACUGC
1435
34059
HCVa:301L21 antisense

GcuuuccGGAAcAccAuGATsT

1899






siNA (283C) inv






stab08





304
GCCUGAUAGGGUGCUUGCGAGUG
1444
34060
HCVa:322L21 antisense

GAcuAucccAcGAAcGcucTsT

1900






siNA (304C) inv






stab08





307
UGAUAGGGUGCUUGCGAGUGCCC
1446
34061
HCVa:325L21 antisense
uAucccAcGAAcGcucAcGTsT
1901






siNA (307C) inv






stab08





82
UAGCCAUGGCGUUAGUAUGAGUG
1460
34128
HCVb:100L18 (82C) 5′p
pUCAUACUAACGCCAUGGC GUUAGUAUGAB
1902






palindrome siNA





82
UAGCCAUGGCGUUAGUAUGAGUG
1460
34129
HCVb:100L17 (82C) 5′p
pCAUACUAACGCCAUGGC GUUAGUAUGB
1903






palindrome siNA





82
UAGCCAUGGCGUUAGUAUGAGUG
1460
34130
HCVb:100L16 (82C) 5′p
pAUACUAACGCCAUGGC GUUAGUAUB
1904






palindrome siNA





82
UAGCCAUGGCGUUAGUAUGAGUG
1460
34131
HCVb:100L15 (82C) 5′p
pUACUAACGCCAUGGC GUUAGUAB
1905






palindrome siNA





126
CCCUCCCGGGAGAGCCAUAGUGG
1461
34132
HCVb:144L19 (126C)
pACUAUGGCUCUCCCGGGAG AGCCAUAGUB
1906






5′p palindrome siNA





126
CCCUCCCGGGAGAGCCAUAGUGG
1461
34133
HCVb:144L18 (126C)
pCUAUGGCUCUCCCGGGAG AGCCAUAGB
1907






5′p palindrome siNA





126
CCCUCCCGGGAGAGCCAUAGUGG
1461
34134
HCVb:144L17 (126C)
pUAUGGCUCUCCCGGGAG AGCCAUAB
1908






5′p palindrome siNA





126
CCCUCCCGGGAGAGCCAUAGUGG
1461
34135
HCVb:144L16 (126C)
pAUGGCUCUCCCGGGAG AGCCAUB
1909






5′p palindrome siNA





126
CCCUCCCGGGAGAGCCAUAGUGG
1461
34136
HCVb:144L15 (126C)
pUGGCUCUCCCGGGAG AGCCAB
1910






5′p palindrome siNA





155
GAACCGGUGAGUACACCGGAAUU
1432
34137
HCVb:171L17 (155C)
pCCGGUGUACUCACCGGU GAGUACACCGGB
1911






5′p palindrome siNA





155
GAACCGGUGAGUACACCGGAAUU
1432
34138
HCVb:170L16 (155C)
pCGGUGUACUCACCGGU GAGUACACCGB
1912






5′p palindrome siNA





155
GAACCGGUGAGUACACCGGAAUU
1432
34139
HCVb:169L15 (155C)
pGGUGUACUCACCGGU GAGUACACCB
1913






5′p palindrome siNA





315
GCCCCGGGAGGUCUCGUAGACCG
1416
34140
HCVb:331L17 (315C)
pCUACGAGACCUCCCGGG AGGUCUCGUAGB
1914






5′p palindrome siNA





315
GCCCCGGGAGGUCUCGUAGACCG
1416
34141
HCVb:330L16 (315C)
pUACGAGACCUCCCGGG AGGUCUCGUAB
1915






5′p palindrome siNA





315
GCCCCGGGAGGUCUCGUAGACCG
1416
34142
HCVb:329L15 (315C)
pACGAGACCUCCCGGG AGGUCUCGUB
1916






5′p palindrome siNA





327
CCCCGGGAGGUCUCGUAGACCGU
1417
34494
HCVa:345L21 antisense
GGucuAcGAGAccucccGGTT B
1917






siNA (327C) stab19





327
CCCCGGGAGGUCUCGUAGACCGU
1417
34495
HCVa:345L21 antisense
GGcccuccAGAGcAucuGGTT B
1918






siNA (327C) inv






stab19





304
GCCUGAUAGGGUGCUUGCGAGUG
1444
34496
HCVa:322L21 antisense
cucGcAAGcAcccuAucAGTT B
1919






(304C) stab19





304
GCCUGAUAGGGUGCUUGCGAGUG
1444
34499
HCVa:322L21 antisense
GAcuAucccAcGAAcGcucTT B
1920






siNA (304C) inv






stab19





282
UCGCGAAAGGCCUUGUGGUACUG
1434
34581
HCVa:282U21 sense
GCGAAAGGCCUUGUGGUACTT
1921






siNA stab00





283
CGCGAAAGGCCUUGUGGUACUGC
1435
34582
HCVa:283U21 sense
CGAAAGGCCUUGUGGUACUTT
1922






siNA stab00





304
GCCUGAUAGGGUGCUUGCGAGUG
1444
34583
HCVa:304U21 sense
CUGAUAGGGUGCUUGCGAGTT
1923






siNA stab00





307
UGAUAGGGUGCUUGCGAGUGCCC
1446
34584
HCVa:307U21 sense
AUAGGGUGCUUGCGAGUGCTT
1924






siNA stab00





327
CCCCGGGAGGUCUCGUAGACCGU
1417
34585
HCVa:327U21 sense
CCGGGAGGUCUCGUAGACCTT
1925






siNA stab00





282
UCGCGAAAGGCCUUGUGGUACUG
1434
34586
HCVa:300L21 antisense
GUACCACAAGGCCUUUCGCTT
1926






siNA (282C) stab00





283
CGCGAAAGGCCUUGUGGUACUGC
1435
34587
HCVa:301L21 antisense
AGUACCACAAGGCCUUUCGTT
1927






siNA (283C) stab00





304
GCCUGAUAGGGUGCUUGCGAGUG
1444
34588
HCVa:322L21 antisense
CUCGCAAGCACCCUAUCAGTT
1928






siNA (304C) stab00





307
UGAUAGGGUGCUUGCGAGUGCCC
1446
34589
HCVa:325L21 antisense
GCACUCGCAAGCACCCUAUTT
1929






siNA (307C) stab00





282
UCGCGAAAGGCCUUGUGGUACUG
1434
34590
HCVa:282U21 sense
CAUGGUGUUCCGGAAAGCGTT
1930






siNA inv stab00





283
CGCGAAAGGCCUUGUGGUACUGC
1435
34591
HCVa:283U21 sense
UCAUGGUGUUCCGGAAAGCTT
1931






siNA inv stab00





304
GCCUGAUAGGGUGCUUGCGAGUG
1444
34592
HCVa:304U21 sense
GAGCGUUCGUGGGAUAGUCTT
1932






siNA inv stab00





307
UGAUAGGGUGCUUGCGAGUGCCC
1446
34593
HCVa:307U21 sense
CGUGAGCGUUCGUGGGAUATT
1933






siNA inv stab00





327
CCCCGGGAGGUCUCGUAGACCGU
1417
34594
HCVa:327U21 sense
CCAGAUGCUCUGGAGGGCCTT
1934






siNA inv stab00





282
UCGCGAAAGGCCUUGUGGUACUG
1434
34595
HCVa:300L21 antisense
CGCUUUCCGGAACACCAUGTT
1935






siNA (282C) inv






stab00





283
CGCGAAAGGCCUUGUGGUACUGC
1435
34596
HCVa:301L21 antisense
GCUUUCCGGAACACCAUGATT
1936






siNA (283C) inv






stab00





304
GCCUGAUAGGGUGCUUGCGAGUG
1444
34597
HCVa:322L21 antisense
GACUAUCCCACGAACGCUCTT
1937






siNA (304C) inv stab00





307
UGAUAGGGUGCUUGCGAGUGCCC
1446
34598
HCVa:325L21 antisense
UAUCCCACGAACGCUCACGTT
1938






siNA (307C) inv stab00





327
CCCCGGGAGGUCUCGUAGACCGU
1417
34599
HCVa:345L21 antisense
GGCCCUCCAGAGCAUCUGGTT
1939






siNA (327C) inv stab00





327
CCCCGGGAGGUCUCGUAGACCGU
1417
35173
HCVa:327U21 sense
B ccGGGAGGucucGUAGACCTT B
1940






siNA stab07 N1





327
CCCCGGGAGGUCUCGUAGACCGU
1417
35174
HCVa:345L21 antisense

GGUCUAcGAGAccucccGGTsT

1941






siNA (327C) stab08 N1





327
CCCCGGGAGGUCUCGUAGACCGU
1417
35175
HCVa:345L21 antisense

GGUcuAcGAGAccucccGGTsT

1942






siNA (327C) stab25





327
CCCCGGGAGGUCUCGUAGACCGU
1417
35176
HCVa:345L21 antisense

GGucuAcGAGAccucccGGTsT

1943






siNA (327C) stab08 N3





327
CCCCGGGAGGUCUCGUAGACCGU
1417
35177
HCVa:345L21 antisense

GGucuAcGAGAccucccGGTsT

1944






siNA (327C) stab24





304
GCCUGAUAGGGUGCUUGCGAGUG
1444
35178
HCVa:304U21 sense
B cuGAuAGGGuGcuUGCGAGTT B
1945






siNA stab01 N1





304
GCCUGAUAGGGUGCUUGCGAGUG
1444
35179
HCVa:322L21 antisense
CUCGCAAGcAcccuAucAGTsT
1946






siNA (304C) stab08 N1





304
GCCUGAUAGGGUGCUUGCGAGUG
1444
35180
HCVa:322L21 antisense
CUCGcAAGcAcccuAucAGTsT
1947






siNA (304C) stab25





304
GCCUGAUAGGGUGCUUGCGAGUG
1444
35181
HCVa:322L21 antisense
CUcGcAAGcAcccuAucAGTsT
1948






siNA (304C) stab08 N3





304
GCCUGAUAGGGUGCUUGCGAGUG
1444
35182
HCVa:322L21 antisense
CucGcAAGcAcccuAucAGTsT
1949






siNA (304C) stab24





327
CCCCGGGAGGUCUCGUAGACCGU
1417
35225
HCVa:327 siNA stab0/0
GGUCUACGAGACCUCCCGG
1950






Pal01
CCGGGAGGUCUCGUAGACC





327
CCCCGGGAGGUCUCGUAGACCGU
1417
35226
HCVa:327 siNA stab0/0
GGUCUACGAGACCUCCCGG
1951






Pal02
CCGGGAGGUCUCGUAGACCTT





327
CCCCGGGAGGUCUCGUAGACCGU
1417
35227
HCVa:327 siNA stab0/0
GGUCUACGAGACCUCCCG
1952






Pal03
CGGGAGGUCUCGUAGACC





327
CCCCGGGAGGUCUCGUAGACCGU
1417
35228
HCVa:327 siNA stab0/0
GGUCUACGAGACCUCCCG
1953






Pal04
CGGGAGGUCUCGUAGACCTT





327
CCCCGGGAGGUCUCGUAGACCGU
1417
35229
HCVa:327 siNA stab0/0
GGUCUACGAGACCUCCC
1954






Pal05
GGGAGGUCUCGUAGACC





327
CCCCGGGAGGUCUCGUAGACCGU
1417
35230
HCVa:327 siNA stab0/0
GGUCUACGAGACCUCCC
1955






Pal06
GGGAGGUCUCGUAGACCTT





327
CCCCGGGAGGUCUCGUAGACCGU
1417
35231
HCVa:327 siNA stab0/0
GGUCUACGAGACCUCC
1956






Pal07
GGAGGUCUCGUAGACC





327
CCCCGGGAGGUCUCGUAGACCGU
1417
35232
HCVa:327 siNA stab0/0
GGUCUACGAGACCUCC
1957






Pal08
GGAGGUCUCGUAGACCTT





327
CCCCGGGAGGUCUCGUAGACCGU
1417
35235
HCVa:327 siNA stab0/0
GUCUACGAGACCUCCCGG
1958






Pal11
GAGGUCUCGUAGAC





327
CCCCGGGAGGUCUCGUAGACCGU
1417
35236
HCVa:327 siNA stab0/0
GUCUACGAGACCUCCCGG
1959






Pal12
GAGGUCUCGUAGACTT





327
CCCCGGGAGGUCUCGUAGACCGU
1417
35237
HCVa:327 siNA stab0/0
UCUACGAGACCUCCCGG GAGGUCUCGUAGA
1960






Pal13





327
CCCCGGGAGGUCUCGUAGACCGU
1417
35238
HCVa:327 siNA stab0/0
UCUACGAGACCUCCCGG
1961






Pal14
GAGGUCUCGUAGATT





327
CCCCGGGAGGUCUCGUAGACCGU
1417
35239
HCVa:327 siNA stab0/0
CUACGAGACCUCCCGG GAGGUCUCGUAG
1962






Pal15





327
CCCCGGGAGGUCUCGUAGACCGU
1417
35240
HCVa:327 siNA stab0/0
CUACGAGACCUCCCGG GAGGUCUCGUAGTT
1963






Pal16





327
CCCCGGGAGGUCUCGUAGACCGU
1417
35241
HCVa:327 siNA stab0/0
GGUCUACGAGACCUCCAGG UCUCGUAGACC
1964






Pal17





327
CCCCGGGAGGUCUCGUAGACCGU
1417
35242
HCVa:327 siNA stab0/0
GGUCUACGAGACCUCCAGG
1965






Pal18
UCUCGUAGACCTT





327
CCCCGGGAGGUCUCGUAGACCGU
1417
35243
HCVa:327 siNA stab0/0
GGUCUACGAGACCUCGAGG UCUCGUAGACC
1966






Pal19





327
CCCCGGGAGGUCUCGUAGACCGU
1417
35244
HCVa:327 siNA stab0/0
GGUCUACGAGACCUCGAGG
1967






Pal20
UCUCGUAGACCTT





327
CCCCGGGAGGUCUCGUAGACCGU
1417
35245
HCVa:327 siNA stab0/0
GGUCUACGAGACCUGCAGG UCUCGUAGACC
1968






Pal21





327
CCCCGGGAGGUCUCGUAGACCGU
1417
35246
HCVa:327 siNA stab0/0
GGUCUACGAGACCUGCAGG
1969






Pal22
UCUCGUAGACCTT





304
GCCUGAUAGGGUGCUUGCGAGUG
1444
35247
HCVa:304 siNA stab0/0
GACUAUCCCACGAACGCUC
1970






Pal01
GAGCGUUCGUGGGAUAGUCTT





304
GCCUGAUAGGGUGCUUGCGAGUG
1444
35248
HCVa:304 siNA stab0/0
GACUAUCCCACGAACGCUC
1971






Pal02
GAGCGUUCGUGGGAUAGUC





304
GCCUGAUAGGGUGCUUGCGAGUG
1444
35249
HCVa:304 siNA stab0/0
GACUAUCCCACGAACGCGU
1972






Pal03
UCGUGGGAUAGUCTT





304
GCCUGAUAGGGUGCUUGCGAGUG
1444
35250
HCVa:304 siNA stab0/0
GACUAUCCCACGAACGCGU
1973






Pal04
UCGUGGGAUAGUC





304
GCCUGAUAGGGUGCUUGCGAGUG
1444
35251
HCVa:304 siNA stab0/0
GACUAUCCCACGAACGUUC
1974






Pal05
GUGGGAUAGUCTT





304
GCCUGAUAGGGUGCUUGCGAGUG
1444
35252
HCVa:304 siNA stab0/0
GACUAUCCCACGAACGUUC GUGGGAUAGUC
1975






Pal06





304
GCCUGAUAGGGUGCUUGCGAGUG
1444
35253
HCVa:304 siNA stab0/0
ACUAUCCCACGAACGUUC GUGGGAUAGUTT
1976






Pal07





304
GCCUGAUAGGGUGCUUGCGAGUG
1444
35254
HCVa:304 siNA stab0/0
ACUAUCCCACGAACGUUC GUGGGA
1977






Pal08





327
CCCCGGGAGGUCUCGUAGACCGU
1462
36414
HCVa bf-L-21 siNA
CCGGGAGGUCUCGUAGACCTT L
1978



UCGCGAAAGGCCUUGUGGUACUG


stab00 [HCVa:327U21
GCGAAAGGCCUUGUGGUACTT






sense o18S






HCVa:282U21 sense]





327
CCCCGGGAGGUCUCGUAGACCGU
1463
36415
HCVa bf-L-22 siNA
CCGGGAGGUCUCGUAGACCTT L
1979



UGAUAGGGUGCUUGCGAGUGCCC


stab00 [HCVa:327U21
AUAGGGUGCUUGCGAGUGCTT






sense o18S






HCVa:307U21 sense]





307
UGAUAGGGUGCUUGCGAGUGCCC
1464
36430
HCVa bf-L-20 siNA
AUAGGGUGCUUGCGAGUGCTT L
1980



UCGCGAAAGGCCUUGUGGUACUG


stab00 [HCVa:307U21
GCGAAAGGCCUUGUGGUACTT






sense o18S






HCVa:282U21 sense]





307
UGAUAGGGUGCUUGCGAGUGCCC
1446
36438
HCVa:307U21 sense
AUAGGGUGCUUGCGAGUGCTT
1924






siNA stab00





307
UGAUAGGGUGCUUGCGAGUGCCC
1446
36446
HCVa:325L21 antisense
GCACUCGCAAGCACCCUAUTT
1929






siNA (307C) stab00





327
CCCCGGGAGGUCUCGUAGACCGU
1417
36447
HCVa:345L21 antisense
GGUCUACGAGACCUCCCGGTT
1732






siNA (327C) stab00





304
GCCUGAUAGGGUGCUUGCGAGUG
1444
36727
HCVa:304U21 sense
B CUGAUAGGGUGCUUGCGAGTT B
1981






siNA stab09





304
GCCUGAUAGGGUGCUUGCGAGUG
1444
36728
HCVa:322L21 antisense
CUCGCAAGCACCCUAUCAGTsT
1982






siNA (304C) stab10





304
GCCUGAUAGGGUGCUUGCGAGUG
1444
37010
HCVa:304U21 sense
B cuGAuAGGGuGcuuGcGAGTT B
1983






siNA stab04





304
GCCUGAUAGGGUGCUUGCGAGUG
1444
37011
HCVa:322L21 antisense
cucGcAAGcAcccuAucAGTsT
1984






siNA (304C) stab05





307
CCCCGGGAGGUCUCGUAGACCGU
1463
37781
HCVa bf-L-22 siNA
B ccGGGAGGucucGuAGAccTT L
1985



UGAUAGGGUGCUUGCGAGUGCCC


stab07 [HCVa:327U21
AuAGGGuGcuuGcGAGuGcTT B






sense o18S






HCVa:307U21 sense]





307
UGAUAGGGUGCUUGCGAGUGCCC
1446
37790
HCVa:325L21 antisense

GCAcucGcAAGcAcccuAuTT

1986






siNA (307C) stab26





327
CCCCGGGAGGUCUCGUAGACCGU
1417
37791
HCVa:345L21 antisense

GGUcuAcGAGAccucccGGTT

1987






siNA (327C) stab26





282
UCGCGAAAGGCCUUGUGGUACUG
1434
38279
HCVa:300L21 antisense

GUAccAcAAGGccuuucGcTsT

1988






siNA (282C) stab25





283
CGCGAAAGGCCUUGUGGUACUGC
1435
38280
HCVa:301L21 antisense

AGUAccAcAAGGccuuucGTsT

1989






siNA (283C) stab25





307
UGAUAGGGUGCUUGCGAGUGCCC
1446
38281
HCVa:325L21 antisense

GCAcucGcAAGcAcccuAuTsT

1990






siNA (307C) stab25





304
GCCUGAUAGGGUGCUUGCGAGUG
1444
38283
HCVa:322L21 antisense
CUCGcAAGcAcccuAucAGTT
1991






siNA (304C) stab26





304
GCCUGAUAGGGUGCUUGCGAGUG
1444
38284
HCVa:322L21 antisense
CUCGcAAGcAcccuAucAGTTB
1992






siNA (304C) stab27





282
UCGCGAAAGGCCUUGUGGUACUG
1434
38293
HCVa:300L21 antisense

GuAccAcAAGGccuuucGcTT B

1993






siNA (282C) stab19





282
UCGCGAAAGGCCUUGUGGUACUG
1434
38294
HCVa:300L21 antisense

GUAccAcAAGGccuuucGcTT

1994






siNA (282C) stab26





282
UCGCGAAAGGCCUUGUGGUACUG
1434
38295
HCVa:300L21 antisense

GUAccAcAAGGccuuucGcTT B

1995






siNA (282C) stab27





282
UCGCGAAAGGCCUUGUGGUACUG
1434
38296
HCVa:300L21 antisense

GuAccAcAAGGccuuucGcTsT

1996






siNA (282C) stab29





282
UCGCGAAAGGCCUUGUGGUACUG
1434
38297
HCVa:300L21 antisense

GuAccAcAAGGccuuucGcTT

1997






siNA (282C) stab30





282
UCGCGAAAGGCCUUGUGGUACUG
1434
38298
HCVa:300L21 antisense

GuAccAcAAGGccuuucGcTT B

1998






siNA (282C) stab31





282
UCGCGAAAGGCCUUGUGGUACUG
1434
38299
HCVa:300L21 antisense

GuAccAcAAGGccuuucGcTT

1999






siNA (282C) stab32





304
GCCUGAUAGGGUGCUUGCGAGUG
1444
38300
HCVa:322L21 antisense
cucGcAAGcAcccuAucAGTT
2000






siNA (304C) stab32





327
CCCCGGGAGGUCUCGUAGACCGU
1417
38301
HCVa:345L21 antisense

GGUcuAcGAGAccucccGGTT B

2001






siNA (327C) stab21





327
CCCCGGGAGGUCUCGUAGACCGU
1417
38302
HCVa:345L21 antisense

GGucuAcGAGAccucccGGTT

2002






siNA (327C) stab30





327
CCCCGGGAGGUCUCGUAGACCGU
1417
38303
HCVa:345L21 antisense

GGucuAcGAGAccucccGGTT B

2003






siNA (327C) stab31





327
CCCCGGGAGGUCUCGUAGACCGU
1417
38304
HCVa:345L21 antisense

GGucuAcGAGAccucccGGTT

2004






siNA (327C) stab32





304
CCCCGGGAGGUCUCGUAGACCGU
1465
38310
HCV bf-L-23 siNA
CCGGGAGGUCUCGUAGACCTT L
2005



GCCUGAUAGGGUGCUUGCGAGUG


stab00 [HCV:327U21
CUGAUAGGGUGCUUGCGAGTT






sense o18S






HCV:304U21 sense]





282
GCCUGAUAGGGUGCUUGCGAGUG
1466
38311
HCV bf-L-24 siNA
CUGAUAGGGUGCUUGCGAGTT L
2006



UCGCGAAAGGCCUUGUGGUACUG


stab00 [HCV:304U21
GCGAAAGGCCUUGUGGUACTT






sense o18S






HCV:282U21 sense]





304
CCCCGGGAGGUCUCGUAGACCGU
1465
38312
HCV bf-L-23 siNA
B ccGGGAGGucucGuAGAccTT L
2007



GCCUGAUAGGGUGCUUGCGAGUG


stab07 [HCV:327U21
cuGAuAGGGuGcuuGcGAGTT B






sense o18S






HCV:304U21 sense]





282
CCCCGGGAGGUCUCGUAGACCGU
1462
38313
HCV bf-L-21 siNA
B ccGGGAGGucucGuAGAccTT L
2008



UCGCGAAAGGCCUUGUGGUACUG


stab07 [HCVa:327U21

GcGAAAGGccuuGuGGuAcTT B







sense o18S






HCVa:282U21 sense]





282
GCCUGAUAGGGUGCUUGCGAGUG
1466
38314
HCV bf-L-24 siNA
B cuGAuAGGGuGcuuGcGAGTT L
2009



UCGCGAAAGGCCUUGUGGUACUG


stab07 [HCV:304U21

GcGAAAGGccuuGuGGuAcTT B







sense o18S






HCV:282U21 sense]







Uppercase = ribonucleotide





u,c = 2′-deoxy-2′-fluoro U,C





T = thymidine





B = inverted deoxy abasic





s = phosphorothioate linkage






A = deoxy Adenosine






G = deoxy Guanosine






G = 2′-O-methyl Guanosine






A = 2′-O-methyl Adenosine





L = hegS = hexethelyne glycol spacer; spacer-18 (Glen Research 10-1918-xx)





p = terminal phosphate














TABLE IV










Non-limiting examples of Stabilization Chemistries


for chemically modified siNA constructs












Chemistry
pyrimidine
Purine
cap
p = S
Strand





“Stab 00”
Ribo
Ribo
TT at 3′-

S/AS





ends


“Stab 1”
Ribo
Ribo

5 at 5′-end
S/AS






1 at 3′-end


“Stab 2”
Ribo
Ribo

All linkages
Usually







AS


“Stab 3”
2′-fluoro
Ribo

4 at 5′-end
Usually






4 at 3′-end
S


“Stab 4”
2′-fluoro
Ribo
5′ and 3′-

Usually





ends

S


“Stab 5”
2′-fluoro
Ribo

1 at 3′-end
Usually







AS


“Stab 6”
2′-O-Methyl
Ribo
5′ and 3′-

Usually





ends

S


“Stab 7”
2′-fluoro
2′-deoxy
5′ and 3′-

Usually





ends

S


“Stab 8”
2′-fluoro
2′-O-

1 at 3′-end
S/AS




Methyl


“Stab 9”
Ribo
Ribo
5′ and 3′-

Usually





ends

S


“Stab 10”
Ribo
Ribo

1 at 3′-end
Usually







AS


“Stab 11”
2′-fluoro
2′-deoxy

1 at 3′-end
Usually







AS


“Stab 12”
2′-fluoro
LNA
5′ and 3′-

Usually





ends

S


“Stab 13”
2′-fluoro
LNA

1 at 3′-end
Usually







AS


“Stab 14”
2′-fluoro
2′-deoxy

2 at 5′-end
Usually






1 at 3′-end
AS


“Stab 15”
2′-deoxy
2′-deoxy

2 at 5′-end






1 at 3′-end
AS


“Stab 16”
Ribo
2′-O-
5′ and 3′-

Usually




Methyl
ends

S


“Stab 17”
2′-O-Methyl
2′-O-
5′ and 3′-

Usually




Methyl
ends

S


“Stab 18”
2′-fluoro
2′-O-
5′ and 3′-

Usually




Methyl
ends

S


“Stab 19”
2′-fluoro
2′-O-
3′-end

S/AS




Methyl


“Stab 20”
2′-fluoro
2′-deoxy
3′-end

Usually







AS


“Stab 21”
2′-fluoro
Ribo
3′-end

Usually







AS


“Stab 22”
Ribo
Ribo
3′-end

Usually







AS


“Stab 23”
2′-fluoro*
2′-deoxy*
5′ and 3′-

Usually





ends

S


“Stab 24”
2′-fluoro*
2′-O-

1 at 3′-end
S/AS




Methyl*


“Stab 25”
2′-fluoro*
2′-O-

1 at 3′-end
S/AS




Methyl*


“Stab 26”
2′-fluoro*
2′-O-


S/AS




Methyl*


“Stab 27”
2′-fluoro*
2′-O-
3′-end

S/AS




Methyl*


“Stab 28”
2′-fluoro*
2′-O-
3′-end

S/AS




Methyl*


“Stab 29”
2′-fluoro*
2′-O-

1 at 3′-end
S/AS




Methyl*


“Stab 30”
2′-fluoro*
2′-O-


S/AS




Methyl*


“Stab 31”
2′-fluoro*
2′-O-
3′-end

S/AS




Methyl*


“Stab 32”
2′-fluoro
2′-O-


S/AS




Methyl







CAP = any terminal cap, see for example FIG. 10.





All Stab 00-32 chemistries can comprise 3′-terminal thymidine (TT) residues





All Stab 00-32 chemistries typically comprise about 21 nucleotides, but can vary as described herein.





S = sense strand





AS = antisense strand





*Stab 23 has a single ribonucleotide adjacent to 3′-CAP





*Stab 24 and Stab 28 have a single ribonucleotide at 5′-terminus





*Stab 25, Stab 26, and Stab 27 have three ribonucleotides at 5′-terminus





*Stab 29, Stab 30, and Stab 31, any purine at first three nucleotide positions from 5′-terminus are ribonucleotides





p = phosphorothioate linkage



















TABLE V














Wait Time*



Reagent
Equivalents
Amount
Wait Time* DNA
2′-O-methyl
Wait Time* RNA










A. 2.5 μmol Synthesis Cycle ABI 394 Instrument
















Phosphoramidites
6.5
163
μL
45
sec
2.5
min
7.5
min


S-Ethyl Tetrazole
23.8
238
μL
45
sec
2.5
min
7.5
min


Acetic Anhydride
100
233
μL
5
sec
5
sec
5
sec


N-Methyl
186
233
μL
5
sec
5
sec
5
sec


Imidazole


TCA
176
2.3
mL
21
sec
21
sec
21
sec


Iodine
11.2
1.7
mL
45
sec
45
sec
45
sec


Beaucage
12.9
645
μL
100
sec
300
sec
300
sec













Acetonitrile
NA
6.67
mL
NA
NA
NA







B. 0.2 μmol Synthesis Cycle ABI 394 Instrument
















Phosphoramidites
15
31
μL
45
sec
233
sec
465
sec


S-Ethyl Tetrazole
38.7
31
μL
45
sec
233
min
465
sec


Acetic Anhydride
655
124
μL
5
sec
5
sec
5
sec


N-Methyl
1245
124
μL
5
sec
5
sec
5
sec


Imidazole


TCA
700
732
μL
10
sec
10
sec
10
sec


Iodine
20.6
244
μL
15
sec
15
sec
15
sec


Beaucage
7.7
232
μL
100
sec
300
sec
300
sec













Acetonitrile
NA
2.64
mL
NA
NA
NA










C. 0.2 μmol Synthesis Cycle 96 well Instrument













Equivalents:







DNA/2′-O-
Amount: DNA/
Wait Time*
Wait Time*
Wait Time*


Reagent
methyl/Ribo
2′-O-methyl/Ribo
DNA
2′-O-methyl
Ribo



















Phosphoramidites
22/33/66
40/60/120
μL
60
sec
180
sec
360
sec


S-Ethyl Tetrazole
70/105/210
40/60/120
μL
60
sec
180
min
360
sec


Acetic Anhydride
265/265/265
50/50/50
μL
10
sec
10
sec
10
sec


N-Methyl
502/502/502
50/50/50
μL
10
sec
10
sec
10
sec


Imidazole


TCA
238/475/475
250/500/500
μL
15
sec
15
sec
15
sec


Iodine
6.8/6.8/6.8
80/80/80
μL
30
sec
30
sec
30
sec


Beaucage
34/51/51
80/120/120

100
sec
200
sec
200
sec













Acetonitrile
NA
1150/1150/1150
μL
NA
NA
NA







Wait time does not include contact time during delivery.





Tandem synthesis utilizes double coupling of linker molecule







Claims
  • 1. A multifunctional siNA molecule comprising a structure having Formula MF-III:
  • 2. The multifunctional siNA molecule of claim 1, wherein W connects the 3′-end of sequence Y′ with the 3′-end of sequence Y.
  • 3. The multifunctional siNA molecule of claim 1, wherein W connects the 3′-end of sequence Y′ with the 5′-end of sequence Y.
  • 4. The multifunctional siNA molecule of claim 1, wherein W connects the 5′-end of sequence Y′ with the 5′-end of sequence Y.
  • 5. The multifunctional siNA molecule of claim 1, wherein W connects the 5′-end of sequence Y′ with the 3′-end of sequence Y.
  • 6. The multifunctional siNA molecule of claim 1, wherein a terminal phosphate group is present at the 5′-end of any of sequence X, X′, Y, or Y′.
  • 7. The multifunctional siNA molecule of claim 1, wherein W connects sequences Y and Y′ via a biodegradable linker.
  • 8. The multifunctional siNA molecule of claim 1, wherein W further comprises a conjugate, label, aptamer, ligand, lipid, or polymer.
  • 9. The multifunctional siNA molecule of claim 1, wherein any of sequence X, X′, Y, or Y′ comprises a 3′-terminal cap moiety.
  • 10. The multifunctional siNA molecule of claim 9, wherein said terminal cap moiety is an inverted deoxyabasic moiety.
  • 11. The multifunctional siNA molecule of claim 10, wherein said terminal cap moiety is an inverted deoxynucleotide moiety.
  • 12. The multifunctional siNA molecule of claim 10, wherein said terminal cap moiety is a dinucleotide moiety.
  • 13. The multifunctional siNA molecule of claim 12, wherein said dinucleotide is dithymidine (TT).
  • 14. The multifunctional siNA molecule of claim 1, wherein said siNA molecule comprises no ribonucleotides.
  • 15. The multifunctional siNA molecule of claim 1, wherein said siNA molecule comprises one or more ribonucleotides.
  • 16. The multifunctional siNA molecule of claim 1, wherein any purine nucleotide in said siNA is a 2′-O-methyl purine nucleotide.
  • 17. The multifunctional siNA molecule of claim 1, wherein any purine nucleotide in said siNA is a 2′-deoxy purine nucleotide.
  • 18. The multifunctional siNA molecule of claim 1, wherein any pyrimidine nucleotide in said siNA is a 2′-deoxy-2′-fluoro pyrimidine nucleotide.
  • 19. The multifunctional siNA molecule of claim 1, wherein each X, X′, Y, and Y′ independently comprises about 19 to about 23 nucleotides.
  • 20. The multifunctional siNA molecule of claim 1, wherein said first and second HCV target sequence each is a HCV RNA sequence.
  • 21. The multifunctional siNA molecule of claim 1, wherein said first HCV target sequence is a HCV RNA sequence, and said second HCV target sequence is a cellular target RNA sequence that is required for HCV infection or replication.
  • 22. The multifunctional siNA molecule of claim 1, wherein said first HCV target sequence is a cellular target RNA sequence that is required for HCV infection or replication, and said second HCV target sequence is a HCV RNA sequence.
  • 23. The multifunctional siNA molecule of claim 1, wherein said first and second HCV target sequences are each a cellular target RNA sequence that is required for HCV infection or replication.
  • 24. The multifunctional siNA molecule of claim 21 or claim 22, wherein said cellular target RNA sequence is selected from a La antigen, FAS, FAS ligand, interferon regulatory factor, cellular PKR protein, elF2Bgamma, elF2gamma, human DEAD Box protein (DDX3), and polypyrimidine tract-binding protein.
  • 25. A pharmaceutical composition comprising the multifunctional siNA molecule of claim 1 and an acceptable carrier or diluent.
Parent Case Info

This invention is a continuation-in-part of U.S. patent application Ser. No. 10/667,271, filed Sep. 16, 2003, which is a continuation-in-part of International Patent Application No. PCT/US03/05043, filed Feb. 20, 2003, which is a continuation-in-part of McSwiggen PCT/US02/09187, filed Mar. 26, 2002 and claims the benefit of McSwiggen U.S. Ser. No. 60/401,104, filed Aug. 5, 2002. This application is also a continuation-in-part of International Patent Application No. PCT/US04/16390, filed May 24, 2004, which is a continuation-in-part of U.S. patent application Ser. No. 10/826,966, filed Apr. 16, 2004, which is continuation-in-part of U.S. patent application Ser. No. 10/757,803, filed Jan. 14, 2004, which is a continuation-in-part of U.S. patent application Ser. No. 10/720,448, filed Nov. 24, 2003, which is a continuation-in-part of U.S. patent application Ser. No. 10/693,059, filed Oct. 23, 2003, which is a continuation-in-part of U.S. patent application Ser. No. 10/444,853, filed May 23, 2003, which is a continuation-in-part of International Patent Application No. PCT/US03/05346, filed Feb. 20, 2003, and a continuation-in-part of International Patent Application No. PCT/US03/05028, filed Feb. 20, 2003, both of which claim the benefit of U.S. Provisional Application No. 60/358,580 filed Feb. 20, 2002, U.S. Provisional Application No. 60/363,124 filed Mar. 11, 2002, U.S. Provisional Application No. 60/386,782 filed Jun. 6, 2002, U.S. Provisional Application No. 60/406,784 filed Aug. 29, 2002, U.S. Provisional Application No. 60/408,378 filed Sep. 5, 2002, U.S. Provisional Application No. 60/409,293 filed Sep. 9, 2002, and U.S. Provisional Application No. 60/440,129 filed Jan. 15, 2003. This application is also a continuation-in-part of International Patent Application No. PCT/US04/13456, filed Apr. 30, 2004, which is a continuation-in-part of U.S. patent application Ser. No. 10/780,447, filed Feb. 13, 2004, which is a continuation-in-part of U.S. patent application Ser. No. 10/427,160, filed Apr. 30, 2003, which is a continuation-in-part of International Patent Application No. PCT/US02/15876 filed May 17, 2002, which claims the benefit of U.S. Provisional Application No. 60/292,217, filed May 18, 2001, U.S. Provisional Application No. 60/362,016, filed Mar. 6, 2002, U.S. Provisional Application No. 60/306,883, filed Jul. 20, 2001, and U.S. Provisional Application No. 60/311,865, filed Aug. 13, 2001. This application is also a continuation-in-part of U.S. patent application Ser. No. 10/727,780 filed Dec. 3, 2003. This application also claims the benefit of U.S. Provisional Application No. 60/543,480, filed Feb. 10, 2004. The instant application claims the benefit of all the listed applications, which are hereby incorporated by reference herein in their entireties, including the drawings.

Provisional Applications (20)
Number Date Country
60401104 Aug 2002 US
60358580 Feb 2002 US
60358580 Feb 2002 US
60363124 Mar 2002 US
60363124 Mar 2002 US
60386782 Jun 2002 US
60386782 Jun 2002 US
60406784 Aug 2002 US
60406784 Aug 2002 US
60408378 Sep 2002 US
60408378 Sep 2002 US
60409293 Sep 2002 US
60409293 Sep 2002 US
60440129 Jan 2003 US
60440129 Jan 2003 US
60292217 May 2001 US
60362016 Mar 2002 US
60306883 Jul 2001 US
60311865 Aug 2001 US
60543480 Feb 2004 US
Continuation in Parts (16)
Number Date Country
Parent 10667271 Sep 2003 US
Child 10942560 Sep 2004 US
Parent PCT/US03/05043 Feb 2003 US
Child 10667271 Sep 2003 US
Parent PCT/US02/09187 Mar 2002 US
Child 10667271 Sep 2003 US
Parent PCT/US04/16390 May 2004 US
Child 10942560 Sep 2004 US
Parent 10826966 Apr 2004 US
Child PCT/US04/16390 May 2004 US
Parent 10757803 Jan 2004 US
Child 10826966 Apr 2004 US
Parent 10720448 Nov 2003 US
Child 10757803 Jan 2004 US
Parent 10693059 Oct 2003 US
Child 10720448 Nov 2003 US
Parent 10444853 May 2003 US
Child 10693059 Oct 2003 US
Parent PCT/US03/05346 Feb 2003 US
Child 10444853 May 2003 US
Parent PCT/US03/05028 Feb 2003 US
Child 10444853 May 2003 US
Parent PCT/US04/13456 Apr 2004 US
Child 10942560 Sep 2004 US
Parent 10780447 Feb 2004 US
Child PCT/US04/13456 Apr 2004 US
Parent 10427160 Apr 2003 US
Child 10780447 Feb 2004 US
Parent PCT/US02/15876 May 2002 US
Child 10427160 Apr 2003 US
Parent 10727780 Dec 2003 US
Child 10942560 Sep 2004 US