AU2012261570B2 - Compositions and methods for inhibiting expression of the PCSK9 gene - Google Patents

Compositions and methods for inhibiting expression of the PCSK9 gene Download PDF

Info

Publication number
AU2012261570B2
AU2012261570B2 AU2012261570A AU2012261570A AU2012261570B2 AU 2012261570 B2 AU2012261570 B2 AU 2012261570B2 AU 2012261570 A AU2012261570 A AU 2012261570A AU 2012261570 A AU2012261570 A AU 2012261570A AU 2012261570 B2 AU2012261570 B2 AU 2012261570B2
Authority
AU
Australia
Prior art keywords
dsrna
pcsk9
expression
cell
pcsk9 gene
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Ceased
Application number
AU2012261570A
Other versions
AU2012261570A1 (en
Inventor
Akin Akinc
Birgit Bramlage
Kevin Fitzgerald
Maria Frank-Kamenetsky
Victor E. Kotelianski
Pamela Tan
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Alnylam Pharmaceuticals Inc
Original Assignee
Alnylam Pharmaceuticals Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from AU2010241357A external-priority patent/AU2010241357B2/en
Application filed by Alnylam Pharmaceuticals Inc filed Critical Alnylam Pharmaceuticals Inc
Priority to AU2012261570A priority Critical patent/AU2012261570B2/en
Publication of AU2012261570A1 publication Critical patent/AU2012261570A1/en
Application granted granted Critical
Publication of AU2012261570B2 publication Critical patent/AU2012261570B2/en
Priority to AU2016203687A priority patent/AU2016203687A1/en
Ceased legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Abstract

The invention relates to a double-stranded ribonucleic acid (dsRNA) for inhibiting the expression of the PCSK9 gene (PCSK9 gene), comprising an antisense strand having a nucleotide sequence which is less that 30 nucleotides in length, generally 19-25 nucleotides in length, and which is substantially complementary to at least a part of the PCSK9 gene. The invention also relates to a pharmaceutical composition comprising the dsRNA together with a pharmaceutically acceptable carrier; methods for treating diseases caused by PCSK9 gene expression and the expression of the PCSK9 gene using the pharmaceutical composition; and

Description

AUSTRALIA Patents Act 1990 Alnylam Pharmaceuticals, Inc. COMPLETE SPECIFICATION STANDARD PATENT Invention Title: Compositions and methods for inhibiting expression of the PCSK9 gene The following statement is a full description of this invention including the best method of performing it known to us: 1 COMPOSITIONS AND METHODS FOR INHIBITING EXPRESSION OF THE PCSK9 GENE Cross-Reference To Related Applications This application is a divisional application under S 79B of the Patents Act 1990 of Australian 5 Patent Application No. 2010241357 which is a divisional application of Australian Patent 200724932, the Australian national stage filing of PCT/US/2007/068655 filed May 10, 2007, which claims priority to U.S. Provisional Application No. 60/799,458, filed May 11, 2006; U.S. Provisional Application No. 60/817,203, filed June 27, 2006; U.S. Provisional Application No. 60/840,089, filed August 25, 2006; U.S. Provisional Application No. 60/829,914, filed October 18, 2006; and U.S. 0 Provisional Application No. 60/901,134, filed February 13, 2007. The contents of all of these provisional applications are hereby incorporated by reference in their entirety. Field of the Invention This invention relates to double-stranded ribonucleic acid (dsRNA), and its use in mediating RNA interference to inhibit the expression of the PCSK9 gene and the use of the 5 dsRNA to treat pathological processes which can be mediated by down regulating PCSK9, such as hyperlipidemia. Background of the Invention Proprotein convertase subtilisin kexin 9 (PCSK9) is a member of the subtilisin serine protease family. The other eight mammalian subtilisin proteases, PCSK1-PCSK8 (also called 20 PC 1/3, PC2, furin, PC4, PC5/6, PACE4, PC7, and SIP/SKI-l) are proprotein convertases that process a wide variety of proteins in the secretory pathway and play roles in diverse biological processes (Bergeron, F. (2000) J. Mol. Endocrinol. 24, 1-22, Gensberg, K., (1998) Semin. Cell Dev. Biol. 9, 11-17, Seidah, N. G. (1999) Brain Res. 848, 45-62, Taylor, N. A., (2003) FASEB J. 17, 1215-1227, and Zhou, A., (1999) J. Biol. Chem. 274, 20745-20748). PCSK9 has been 25 proposed to play a role in cholesterol metabolism. PCSK9 mRNA expression is down-regulated 2 by dietary cholesterol feeding in mice (Maxwell, K. N., (2003) J. Lipid Res. 44, 2109-2119), up regulated by statins in HepG2 cells (Dubuc, G., (2004) Arterioscler. Thromb. Vasc. Biol. 24, 1454-1459), and up-regulated in sterol regulatory element binding protein (SREBP) transgenic mice (Horton, J. D., (2003) Proc. Natl. Acad. Sci. USA 100, 12027-12032), similar to the 5 cholesterol biosynthetic enzymes and the low-density lipoprotein receptor (LDLR). Furthermore, PCSK9 missense mutations have been found to be associated with a form of autosomal dominant hypercholesterolemia (Hchola3) (Abifadel, M., et al. (2003) Nat. Genet. 34, 154-156, Timms, K. M., (2004) Hum. Genet. 114, 349-353, Leren, T. P. (2004) Clin. Genet. 65, 419-422). PCSK9 may also play a role in determining LDL cholesterol levels in the general population, because 0 single-nucleotide polymorphisms (SNPs) have been associated with cholesterol levels in a Japanese population (Shioji, K., (2004) J. Hum. Genet. 49, 109-114). Autosomal dominant hypercholesterolemias (ADHs) are monogenic diseases in which patients exhibit elevated total and LDL cholesterol levels, tendon xanthomas, and premature atherosclerosis (Rader, D. J., (2003) J. Clin. Invest. 111, 1795-1803). The pathogenesis of ADHs 5 and a recessive form, autosomal recessive hypercholesterolemia (ARH) (Cohen, J. C., (2003) Curr. Opin. Lipidol. 14, 121-127), is due to defects in LDL uptake by the liver. ADH may be caused by LDLR mutations, which prevent LDL uptake, or by mutations in the protein on LDL, apolipoprotein B, which binds to the LDLR. ARH is caused by mutations in the ARH protein that are necessary for endocytosis of the LDLR-LDL complex via its interaction with clathrin. 0 Therefore, if PCSK9 mutations are causative in Hchola3 families, it seems likely that PCSK9 plays a role in receptor-mediated LDL uptake. Overexpression studies point to a role for PCSK9 in controlling LDLR levels and, hence, LDL uptake by the liver (Maxwell, K. N. (2004) Proc. Natl. Acad. Sci. USA 101, 7100-7105, Benjannet, S., et al. (2004) J. Biol. Chem. 279, 48865-48875, Park, S. W., (2004) J. Biol. Chem. 25 279, 50630-50638). Adenoviral-mediated overexpression of mouse or human PCSK9 for 3 or 4 days in mice results in elevated total and LDL cholesterol levels; this effect is not seen in LDLR knockout animals (Maxwell, K. N. (2004) Proc. Natl. Acad. Sci. USA 101, 7100-7105, Benjannet, S., et al. (2004) J. Biol. Chem. 279, 48865-48875, Park, S. W., (2004) J. Biol. Chem. 279, 50630-50638). In addition, PCSK9 overexpression results in a severe reduction in hepatic 3 LDLR protein, without affecting LDLR mRNA levels, SREBP protein levels, or SREBP protein nuclear to cytoplasmic ratio. These results indicate that PCSK9, either directly or indirectly, reduces LDLR protein levels by a posttranscriptional mechanism Loss of function mutations in PCSK9 have been designed in mouse models (Rashid et.al., 5 (2005) PNAS, 102, 5374-5379., and identified in human individuals Cohen et al., (2005), Nature Genetics., 37, 161-165. In both cases loss of PCSK9 function lead to lowering of total and LDLc cholesterol. In a retrospective outcome study over 15 years, loss of one copy of PCSK9 was shown to shift LDLc lower and to lead to an increased risk-benefit protection from developing cardiovascular heart disease (Cohen et.al., 2006 N. Engl. J. Med., 354., 1264-1272.). Clearly the 0 evidence to date indicates that lowering of PCSK9 levels will lower LDLc. Recently, double-stranded RNA molecules (dsRNA) have been shown to block gene expression in a highly conserved regulatory mechanism known as RNA interference (RNAi). WO 99/32619 (Fire et al.) discloses the use of a dsRNA of at least 25 nucleotides in length to inhibit the expression of genes in C. elegans. dsRNA has also been shown to degrade target 5 RNA in other organisms, including plants (see, e.g., WO 99/53050, Waterhouse et al.; and WO 99/61631, Heifetz et al.), Drosophila (see, e.g., Yang, D., et al., Curr. Biol. (2000) 10:1191 1200), and mammals (see WO 00/44895, Limmer; and DE 101 00 586.5, Kreutzer et al.). This natural mechanism has now become the focus for the development of a new class of pharmaceutical agents for treating disorders that are caused by the aberrant or unwanted 0 regulation of a gene. Despite significant advances in the field of RNAi and advances in the treatment of pathological processes which can be mediated by down regulating PCSK9 gene expression, there remains a need for agents that can inhibit PCSK9 gene expression and that can treat diseases associated with PCSK9 gene expression such as hyperlipidemia. 4 Summary of the Invention The invention provides a solution to the problem of treating diseases that can be modulated by down regulating the proprotein convertase subtilisin kexin 9 (PCSK9) by using double-stranded ribonucleic acid (dsRNA) to silence PCSK9 expression. 5 The invention provides double-stranded ribonucleic acid (dsRNA), as well as compositions and methods for inhibiting the expression of the PCSK9 gene in a cell or mammal using such dsRNA. The invention also provides compositions and methods for treating pathological conditions that can modulated by down regulating the expression of the PCSK9 gene, such as hyperlipidemia. The dsRNA of the invention comprises an RNA strand (the 0 antisense strand) having a region which is less than 30 nucleotides in length, generally 19-24 nucleotides in length, and is substantially complementary to at least part of an mRNA transcript of the PCSK9 gene. In one embodiment, the invention provides double-stranded ribonucleic acid (dsRNA) for inhibiting the expression of a human PCSK9 gene in a cell, wherein said dsRNA comprises 5 at least two sequences that are complementary to each other and wherein a sense strand comprises a first sequence according to SEQ ID NO:453 and an antisense strand comprises a second sequence according to SEQ ID NO:454 and wherein said dsRNA, upon contact with a cell expressing said PCSK9, inhibits expression of said PCSK9 gene by at least 40%. In one embodiment, the invention provides double-stranded ribonucleic acid (dsRNA) 20 molecules for inhibiting the expression of the PCSK9 gene. The dsRNA comprises at least two sequences that are complementary to each other. The dsRNA comprises a sense strand comprising a first sequence and an antisense strand comprising a second sequence. The antisense 5 strand comprises a nucleotide sequence which is substantially complementary to at least part of an mRNA encoding PCSK9, and the region of complementarity is less than 30 nucleotides in length, generally 19-24 nucleotides in length. The dsRNA, upon contacting with a cell expressing the PCSK9, inhibits the expression of the PCSK9 gene by at least 40%. For example, the dsRNA molecules of the invention can be comprised of a first sequence of the dsRNA that is selected from the group consisting of the sense sequences of Table 1 and Table 2 the second sequence is selected from the group consisting of the antisense sequences of Tables 1 and Table 2. The dsRNA molecules of the invention can be comprised of naturally occurring nucleotides or can be comprised of at least one modified nucleotide, such as a 2'-0 methyl modified nucleotide, a nucleotide comprising a 5'-phosphorothioate group, and a terminal nucleotide linked to a cholesteryl derivative. Alternatively, the modified nucleotide may be chosen from the group of: a 2'-deoxy-2'-fluoro modified nucleotide, a 2'-deoxy-modified 5A nucleotide, a locked nucleotide, an abasic nucleotide, 2'-amino-modified nucleotide, 2'-alkyl modified nucleotide, morpholino nucleotide, a phosphoramidate, and a non-natural base comprising nucleotide. Generally, such modified sequence will be based on a first sequence of said dsRNA selected from the group consisting of the sense sequences of Tables 1 and Table 2 5 and a second sequence selected from the group consisting of the antisense sequences of Tables 1, and Table 2. In another embodiment, the invention provides a cell comprising one of the dsRNAs of the invention. The cell is generally a mammalian cell, such as a human cell. In another embodiment, the invention provides a pharmaceutical composition for 0 inhibiting the expression of the PCSK9 gene in an organism, generally a human subject, comprising one or more of the dsRNA of the invention and a pharmaceutically acceptable carrier or delivery vehicle. In another embodiment, the invention provides a method for inhibiting the expression of the PCSK9 gene in a cell, comprising the following steps: 5 (a) introducing into the cell a double-stranded ribonucleic acid (dsRNA), wherein the dsRNA comprises at least two sequences that are complementary to each other. The dsRNA comprises a sense strand comprising a first sequence and an antisense strand comprising a second sequence. The antisense strand comprises a region of complementarity which is substantially complementary to at least a part of a 20 mRNA encoding PCSK9, and wherein the region of complementarity is less than 30 nucleotides in length, generally 19-24 nucleotides in length, and wherein the dsRNA, upon contact with a cell expressing the PCSK9, inhibits expression of the PCSK9 gene by at least 40%; and (b) maintaining the cell produced in step (a) for a time sufficient to obtain 25 degradation of the mRNA transcript of the PCSK9 gene, thereby inhibiting expression of the PCSK9 gene in the cell. 6 In another embodiment, the invention provides methods for treating, preventing or managing pathological processes which can be mediated by down regulating PCSK9 gene expression, e.g. hyperlipidemia, comprising administering to a patient in need of such treatment, prevention or management a therapeutically or prophylactically effective amount of one or more of the dsRNAs of the invention. In another embodiment, the invention provides vectors for inhibiting the expression of the PCSK9 gene in a cell, comprising a regulatory sequence operably linked to a nucleotide sequence that encodes at least one strand of one of the dsRNA of the invention. In another embodiment, the invention provides a cell comprising a vector for inhibiting the expression of the PCSK9 gene in a cell. The vector comprises a regulatory sequence operably linked to a nucleotide sequence that encodes at least one strand of one of the dsRNA of the invention. In another embodiment, the invention provides pharmaceutical composition for inhibiting the expression of the PCSK9 gene in an organism, comprising a dsRNA and a pharmaceutically acceptable carrier, wherein the dsRNA is as defined herein. In another embodiment, the invention provides a method for inhibiting the expression of the PCSK9 gene in a cell, the method comprising: (a) introducing into the cell a double-stranded ribonucleic acid (dsRNA), wherein the dsRNA is as defined herein; and (b) maintaining the cell produced in step (a) for a time sufficient to obtain degradation of the mRNA transcript of the PCSK9 gene, thereby inhibiting expression of the PCSK9 gene in the cell. In another embodiment, the invention provides a dsRNA for treating, preventing or managing pathological processes which can be mediated by down regulating PCSK9 gene expression, wherein the dsRNA is as defined herein. 7 In another embodiment, the invention provides a vector for inhibiting the expression of the PCSK9 gene in a cell, said vector comprising a regulatory sequence operably linked to a nucleotide sequence that encodes at least one strand of a dsRNA, wherein the dsRNA is as defined herein. In another embodiment, the invention provides an isolated cell comprising the dsRNA defined herein or the vector defined herein. In another embodiment, the invention provides a double-stranded ribonucleic acid (dsRNA) for reducing the expression level of a human PCSK9 gene in a cell, wherein the dsRNA is as defined herein. In another embodiment, the invention provides a pharmaceutical composition for reducing the expression level of the PCSK9 gene in an organism, comprising the dsRNA defined herein and a pharmaceutically acceptable carrier. In another embodiment, the invention provides a method of treating a PCSK9 associated disorder comprising administering to a patient in need of such treatment, a therapeutically effective amount of a dsRNA defined herein. In another embodiment, the invention provides use of the dsRNA defined herein in the manufacture of a medicament for treating a PCSK9 associated disorder. In another embodiment, the invention provides the dsRNA defined herein, or the pharmaceutical composition defined herein, or the method of for inhibiting the expression of the PCSK9 gene in a cell as described herein, or the vector as described herein, or the isolated cell as described herein, or the method of treating a PCSK9 associated disorder as described herein, or the use of the dsRNA as described herein, substantially as described herein with reference to the examples and/or figures. 7A Throughout this specification the word "comprise", or variations such as "comprises" or "comprising", will be understood to imply the inclusion of a stated element, integer or step, or group of elements, integers or steps, but not the exclusion of any other element, integer or step, or group of elements, integers or steps. Any discussion of documents, acts, materials, devices, articles or the like which has been included in the present specification is not to be taken as an admission that any or all of these matters form part of the prior art base or were common general knowledge in the field relevant to the present disclosure as it existed before the priority date of each claim of this application. Brief Description of the Figures Fig. 1 shows the structure of the ND-98 lipid. Fig. 2 shows the results of the in vivo screen of 16 mouse specific (AL-DP-9327 through AL-DP-9342) PCSK9 siRNAs directed against different ORF regions of PCSK9 mRNA (having the first nucleotide corresponding to the ORF position indicated on the graph) in C57/BL6 mice (5 animals/group). The ratio of PCSK9 mRNA to GAPDH mRNA in liver lysates was averaged over each treatment group and compared to a control group treated with PBS or a control group treated with an unrelated siRNA (blood coagulation factor VII). Fig. 3 shows the results of the in vivo screen of 16 human/mouse/rat crossreactive (AL DP-9311 through AL-DP-9326) PCSK9 siRNAs directed against different ORF regions of PCSK9 mRNA (having the first nucleotide corresponding to the ORF position indicated on the graph) in C57/BL6 mice (5 animals/group). The ratio of PCSK9 mRNA to GAPDH mRNA in liver lysates was averaged over each treatment group and compared to a control group treated with PBS or a control group treated with an unrelated siRNA (blood coagulation factor VII). 7B Silencing of PCSK9 mRNA resulted in lowering total serum cholesterol levels. The most efficacious in terms of knocking down PSCK9 message siRNAs showed the most pronounced cholesterol lowering effect (around 20-30%). Fig. 4 shows the results of the in vivo screen of 16 mouse specific (AL-DP-9327 through 5 AL-DP-9342) PCSK9 siRNAs in C57/BL6 mice (5 animals/group). Total serum cholesterol levels were averaged over each treatment group and compared to a control group treated with PBS or a control group treated with an unrelated siRNA (blood coagulation factor VII). Fig. 5 shows the results of the in vivo screen of 16 human/mouse/rat crossreactive (AL DP-9311 through AL-DP-9326) PCSK9 siRNAs in C57/BL6 mice (5 animals/group). Total 0 serum cholesterol levels were averaged over each treatment group and compared to a control group treated with PBS or a control group treated with an unrelated siRNA (blood coagulation factor VII). Fig. 6 shows a comparison of the in vitro and in vivo results for silencing PCSK9. Fig. 7A and Fig. 7B show in vitro results for silencing PCSK9 using monkey primary 5 hepatocytes. Fig. 8 shows in vivo activity of LNP-01 formulated siRNAs to pcsk-9. Fig. 9 shows in vivo activity of LNP-01 Formulated chemically modified 9314 and 10792 parent molecules at different times. Clearly modified versions of 10792 display in vivo silencing activity. 20 Detailed Description of the Invention The invention provides a solution to the problem of treating diseases that can be modulated by the down regulation of the PCSK9 gene, by using double-stranded ribonucleic acid (dsRNA) to silence the PCSK9 gene thus providing treatment for diseases such as hyperlipidemia. 8 The invention provides double-stranded ribonucleic acid (dsRNA), as well as compositions and methods for inhibiting the expression of the PCSK9 gene in a cell or mammal using the dsRNA. The invention also provides compositions and methods for treating pathological conditions and diseases that can be modulated by down regulating the expression of 5 the PCSK9 gene. dsRNA directs the sequence-specific degradation of mRNA through a process known as RNA interference (RNAi). The dsRNA of the invention comprises an RNA strand (the antisense strand) having a region which is less than 30 nucleotides in length, generally 19-24 nucleotides in length, and is substantially complementary to at least part of an mRNA transcript of the PCSK9 gene. The use 0 of these dsRNAs enables the targeted degradation of an mRNA that is involved in sodium transport. Using cell-based and animal assays, the present inventors have demonstrated that very low dosages of these dsRNA can specifically and efficiently mediate RNAi, resulting in significant inhibition of expression of the PCSK9 gene. Thus, the methods and compositions of the invention comprising these dsRNAs are useful for treating pathological processes which can 5 be mediated by down regulating PCSK9, such as in the treatment of hyperlipidemia. The following detailed description discloses how to make and use the dsRNA and compositions containing dsRNA to inhibit the expression of the target PCSK9 gene, as well as compositions and methods for treating diseases that can be modulated by down regulating the expression of PCSK9, such as hyperlipidemia. The pharmaceutical compositions of the 0 invention comprise a dsRNA having an antisense strand comprising a region of complementarity which is less than 30 nucleotides in length, generally 19-24 nucleotides in length, and is substantially complementary to at least part of an RNA transcript of the PCSK9 gene, together with a pharmaceutically acceptable carrier. Accordingly, certain aspects of the invention provide pharmaceutical compositions 25 comprising the dsRNA of the invention together with a pharmaceutically acceptable carrier, methods of using the compositions to inhibit expression of the PCSK9 gene, and methods of using the pharmaceutical compositions to treat diseases that can be modulated by down regulating the expression of PCSK9. 9 I. Definitions For convenience, the meaning of certain terms and phrases used in the specification, examples, and appended claims, are provided below. If there is an apparent discrepancy between the usage of a term in other parts of this specification and its definition provided in this section, 5 the definition in this section shall prevail. "G," "C," "A" and "U" each generally stand for a nucleotide that contains guanine, cytosine, adenine, and uracil as a base, respectively. However, it will be understood that the term "ribonucleotide" or "nucleotide" can also refer to a modified nucleotide, as further detailed below, or a surrogate replacement moiety. The skilled person is well aware that guanine, 0 cytosine, adenine, and uracil may be replaced by other moieties without substantially altering the base pairing properties of an oligonucleotide comprising a nucleotide bearing such replacement moiety. For example, without limitation, a nucleotide comprising inosine as its base may base pair with nucleotides containing adenine, cytosine, or uracil. Hence, nucleotides containing uracil, guanine, or adenine may be replaced in the nucleotide sequences of the invention by a 5 nucleotide containing, for example, inosine. Sequences comprising such replacement moieties are embodiments of the invention. As used herein, "PCSK9" refers to the proprotein convertase subtilisin kexin 9 gene or protein (also known as FH3, HCHOLA3, NARC-1, NARCI). mRNA sequences to PCSK9 are provided as human: NM_174936; mouse: NM_153565, and rat: NM_199253. 20 As used herein, "target sequence" refers to a contiguous portion of the nucleotide sequence of an mRNA molecule formed during the transcription of the PCSK9 gene, including mRNA that is a product of RNA processing of a primary transcription product. As used herein, the term "strand comprising a sequence" refers to an oligonucleotide comprising a chain of nucleotides that is described by the sequence referred to using the standard 25 nucleotide nomenclature. As used herein, and unless otherwise indicated, the term "complementary," when used to describe a first nucleotide sequence in relation to a second nucleotide sequence, refers to the 10 ability of an oligonucleotide or polynucleotide comprising the first nucleotide sequence to hybridize and form a duplex structure under certain conditions with an oligonucleotide or polynucleotide comprising the second nucleotide sequence, as will be understood by the skilled person. Such conditions can, for example, be stringent conditions, where stringent conditions 5 may include: 400 mM NaCl, 40 mM PIPES pH 6.4, 1 mM EDTA, 50 0 C or 70 0 C for 12-16 hours followed by washing. Other conditions, such as physiologically relevant conditions as may be encountered inside an organism, can apply. The skilled person will be able to determine the set of conditions most appropriate for a test of complementarity of two sequences in accordance with the ultimate application of the hybridized nucleotides. 0 This includes base-pairing of the oligonucleotide or polynucleotide comprising the first nucleotide sequence to the oligonucleotide or polynucleotide comprising the second nucleotide sequence over the entire length of the first and second nucleotide sequence. Such sequences can be referred to as "fully complementary" with respect to each other herein. However, where a first sequence is referred to as "substantially complementary" with respect to a second sequence 5 herein, the two sequences can be fully complementary, or they may form one or more, but generally not more than 4, 3 or 2 mismatched base pairs upon hybridization, while retaining the ability to hybridize under the conditions most relevant to their ultimate application. However, where two oligonucleotides are designed to form, upon hybridization, one or more single stranded overhangs, such overhangs shall not be regarded as mismatches with regard to the 0 determination of complementarity. For example, a dsRNA comprising one oligonucleotide 21 nucleotides in length and another oligonucleotide 23 nucleotides in length, wherein the longer oligonucleotide comprises a sequence of 21 nucleotides that is fully complementary to the shorter oligonucleotide, may yet be referred to as "fully complementary" for the purposes of the invention. 25 "Complementary" sequences, as used herein, may also include, or be formed entirely from, non-Watson-Crick base pairs and/or base pairs formed from non-natural and modified nucleotides, in as far as the above requirements with respect to their ability to hybridize are fulfilled. 11 The terms "complementary", "fully complementary" and "substantially complementary" herein may be used with respect to the base matching between the sense strand and the antisense strand of a dsRNA, or between the antisense strand of a dsRNA and a target sequence, as will be understood from the context of their use. 5 As used herein, a polynucleotide which is "substantially complementary to at least part of' a messenger RNA (mRNA) refers to a polynucleotide which is substantially complementary to a contiguous portion of the mRNA of interest (e.g., encoding PCSK9). For example, a polynucleotide is complementary to at least a part of a PCSK9 mRNA if the sequence is substantially complementary to a non-interrupted portion of a mRNA encoding PCSK9. 0 The term "double-stranded RNA" or "dsRNA", as used herein, refers to a complex of ribonucleic acid molecules, having a duplex structure comprising two anti-parallel and substantially complementary, as defined above, nucleic acid strands. The two strands forming the duplex structure may be different portions of one larger RNA molecule, or they may be separate RNA molecules. Where separate RNA molecules, such dsRNA are often referred to in 5 the literature as siRNA ("short interfering RNA"). Where the two strands are part of one larger molecule, and therefore are connected by an uninterrupted chain of nucleotides between the 3' end of one strand and the 5'end of the respective other strand forming the duplex structure, the connecting RNA chain is referred to as a "hairpin loop", "short hairpin RNA" or "shRNA". Where the two strands are connected covalently by means other than an uninterrupted chain of O nucleotides between the 3'-end of one strand and the 5'end of the respective other strand forming the duplex structure, the connecting structure is referred to as a "linker". The RNA strands may have the same or a different number of nucleotides. The maximum number of base pairs is the number of nucleotides in the shortest strand of the dsRNA minus any overhangs that are present in the duplex. In addition to the duplex structure, a dsRNA may comprise one or more 25 nucleotide overhangs. In addition, as used in this specification, "dsRNA" may include chemical modifications to ribonucleotides, including substantial modifications at multiple nucleotides and including all types of modifications disclosed herein or known in the art. Any such modifications, as used in an siRNA type molecule, are encompassed by "dsRNA" for the purposes of this specification and claims. 12 As used herein, a "nucleotide overhang" refers to the unpaired nucleotide or nucleotides that protrude from the duplex structure of a dsRNA when a 3'-end of one strand of the dsRNA extends beyond the 5'-end of the other strand, or vice versa. "Blunt" or "blunt end" means that there are no unpaired nucleotides at that end of the dsRNA, i.e., no nucleotide overhang. A 5 "blunt ended" dsRNA is a dsRNA that is double-stranded over its entire length, i.e., no nucleotide overhang at either end of the molecule. For clarity, chemical caps or non-nucleotide chemical moieties conjugated to the 3' end or 5' end of an siRNA are not considered in determining whether an siRNA has an overhang or is blunt ended. The term "antisense strand" refers to the strand of a dsRNA which includes a region that 0 is substantially complementary to a target sequence. As used herein, the term "region of complementarity" refers to the region on the antisense strand that is substantially complementary to a sequence, for example a target sequence, as defined herein. Where the region of complementarity is not fully complementary to the target sequence, the mismatches are most tolerated in the terminal regions and, if present, are generally in a terminal region or regions, e.g., 5 within 6, 5, 4, 3, or 2 nucleotides of the 5' and/or 3' terminus. The term "sense strand," as used herein, refers to the strand of a dsRNA that includes a region that is substantially complementary to a region of the antisense strand. "Introducing into a cell", when referring to a dsRNA, means facilitating uptake or absorption into the cell, as is understood by those skilled in the art. Absorption or uptake of ,.0 dsRNA can occur through unaided diffusive or active cellular processes, or by auxiliary agents or devices. The meaning of this term is not limited to cells in vitro; a dsRNA may also be "introduced into a cell", wherein the cell is part of a living organism. In such instance, introduction into the cell will include the delivery to the organism. For example, for in vivo delivery, dsRNA can be injected into a tissue site or administered systemically. In vitro 25 introduction into a cell includes methods known in the art such as electroporation and lipofection. The terms "silence" and "inhibit the expression of', in as far as they refer to the PCSK9 gene, herein refer to the at least partial suppression of the expression of the PCSK9 gene, as 13 manifested by a reduction of the amount of mRNA transcribed from the PCSK9 gene which may be isolated from a first cell or group of cells in which the PCSK9 gene is transcribed and which has or have been treated such that the expression of the PCSK9 gene is inhibited, as compared to a second cell or group of cells substantially identical to the first cell or group of cells but which 5 has or have not been so treated (control cells). The degree of inhibition is usually expressed in terms of (mRNA in control cells) - (mRNA in treated cells) 0100% (mRNA in control cells) Alternatively, the degree of inhibition may be given in terms of a reduction of a parameter that is functionally linked to PCSK9 gene transcription, e.g. the amount of protein encoded by the 0 PCSK9 gene which is secreted by a cell, or the number of cells displaying a certain phenotype, e.g apoptosis. In principle, PCSK9 gene silencing may be determined in any cell expressing the target, either constitutively or by genomic engineering, and by any appropriate assay. However, when a reference is needed in order to determine whether a given dsRNA inhibits the expression of the PCSK9 gene by a certain degree and therefore is encompassed by the instant invention, the 5 assay provided in the Examples below shall serve as such reference. For example, in certain instances, expression of the PCSK9 gene is suppressed by at least about 20%, 25%, 35%, or 50% by administration of the double-stranded oligonucleotide of the invention. In some embodiment, the PCSK9 gene is suppressed by at least about 60%, 70%, or 80% by administration of the double-stranded oligonucleotide of the invention. In some 20 embodiments, the PCSK9 gene is suppressed by at least about 85%, 90%, or 95% by administration of the double-stranded oligonucleotide of the invention. Tables 1, 2, provides a wide range of values for inhibition of expression obtained in an in vitro assay using various PCSK9 dsRNA molecules at various concentrations. As used herein in the context of PCSK9 expression, the terms "treat", "treatment", and the 25 like, refer to relief from or alleviation of pathological processes which can be mediated by down regulating PCSK9 gene. In the context of the present invention insofar as it relates to any of the 14 other conditions recited herein below (other than pathological processes which can be mediated by down regulating the PCSK9 gene), the terms "treat", "treatment", and the like mean to relieve or alleviate at least one symptom associated with such condition, or to slow or reverse the progression of such condition. For example, in the context of hyperlipidemia, treatment will 5 involve a decrease in serum lipid levels. As used herein, the phrases "therapeutically effective amount" and "prophylactically effective amount" refer to an amount that provides a therapeutic benefit in the treatment, prevention, or management of pathological processes which can be mediated by down regulating the PCSK9 gene on or an overt symptom of pathological processes which can be mediated by 0 down regulating the PCSK9 gene. The specific amount that is therapeutically effective can be readily determined by ordinary medical practitioner, and may vary depending on factors known in the art, such as, e.g. the type of pathological processes which can be mediated by down regulating the PCSK9 gene, the patient's history and age, the stage of pathological processes which can be mediated by down regulating PCSK9 gene expression, and the administration of 5 other anti-pathological processes which can be mediated by down regulating PCSK9 gene expression. As used herein, a "pharmaceutical composition" comprises a pharmacologically effective amount of a dsRNA and a pharmaceutically acceptable carrier. As used herein, "pharmacologically effective amount," "therapeutically effective amount" or simply "effective O amount" refers to that amount of an RNA effective to produce the intended pharmacological, therapeutic or preventive result. For example, if a given clinical treatment is considered effective when there is at least a 25% reduction in a measurable parameter associated with a disease or disorder, a therapeutically effective amount of a drug for the treatment of that disease or disorder is the amount necessary to effect at least a 25% reduction in that parameter. 25 The term "pharmaceutically acceptable carrier" refers to a carrier for administration of a therapeutic agent. Such carriers include, but are not limited to, saline, buffered saline, dextrose, water, glycerol, ethanol, and combinations thereof and are described in more detail below. The term specifically excludes cell culture medium. 15 As used herein, a "transformed cell" is a cell into which a vector has been introduced from which a dsRNA molecule may be expressed. II. Double-stranded ribonucleic acid (dsRNA) In one embodiment, the invention provides double-stranded ribonucleic acid (dsRNA) 5 molecules for inhibiting the expression of the PCSK9 gene in a cell or mammal, wherein the dsRNA comprises an antisense strand comprising a region of complementarity which is complementary to at least a part of an mRNA formed in the expression of the PCSK9 gene, and wherein the region of complementarity is less than 30 nucleotides in length, generally 19-24 nucleotides in length, and wherein said dsRNA, upon contact with a cell expressing said PCSK9 0 gene, inhibits the expression of said PCSK9 gene by at least 40%. The dsRNA comprises two RNA strands that are sufficiently complementary to hybridize to form a duplex structure. One strand of the dsRNA (the antisense strand) comprises a region of complementarity that is substantially complementary, and generally fully complementary, to a target sequence, derived from the sequence of an mRNA formed during the expression of the PCSK9 gene, the other 5 strand (the sense strand) comprises a region which is complementary to the antisense strand, such that the two strands hybridize and form a duplex structure when combined under suitable conditions. Generally, the duplex structure is between 15 and 30, more generally between 18 and 25, yet more generally between 19 and 24, and most generally between 19 and 21 base pairs in length. Similarly, the region of complementarity to the target sequence is between 15 and 30, 0 more generally between 18 and 25, yet more generally between 19 and 24, and most generally between 19 and 21 nucleotides in length. The dsRNA of the invention may further comprise one or more single-stranded nucleotide overhang(s). The dsRNA can be synthesized by standard methods known in the art as further discussed below, e.g., by use of an automated DNA synthesizer, such as are commercially available from, for example, Biosearch, Applied 25 Biosystems, Inc. In a preferred embodiment, the PCSK9 gene is the human PCSK9 gene. In specific embodiments, the antisense strand of the dsRNA comprises a strand selected from the sense sequences of Tables 1 and 2, and a second sequence selected from the group consisting of the antisense sequences of Tables 1 and 2. Alternative antisense agents that target elsewhere in 16 the target sequence provided in Tables 1 and 2, can readily be determined using the target sequence and the flanking PCSK9 sequence. In further embodiments, the dsRNA comprises at least one nucleotide sequence selected from the groups of sequences provided in Tables 1 and 2. In other embodiments, the dsRNA 5 comprises at least two sequences selected from this group, wherein one of the at least two sequences is complementary to another of the at least two sequences, and one of the at least two sequences is substantially complementary to a sequence of an mRNA generated in the expression of the PCSK9 gene. Generally, the dsRNA comprises two oligonucleotides, wherein one oligonucleotide is described as the sense strand in Tables 1 and 2 and the second oligonucleotide 0 is described as the antisense strand in Tables 1 and 2 The skilled person is well aware that dsRNAs comprising a duplex structure of between 20 and 23, but specifically 21, base pairs have been hailed as particularly effective in inducing RNA interference (Elbashir et al., EMBO 2001, 20:6877-6888). However, others have found that shorter or longer dsRNAs can be effective as well. In the embodiments described above, by 5 virtue of the nature of the oligonucleotide sequences provided in Tables 1 and 2, the dsRNAs of the invention can comprise at least one strand of a length of minimally 21nt. It can be reasonably expected that shorter dsRNAs comprising one of the sequences of Tables 1 and 2 minus only a few nucleotides on one or both ends may be similarly effective as compared to the dsRNAs described above. Hence, dsRNAs comprising a partial sequence of at least 15, 16, 17, 18, 19, 20, 0 or more contiguous nucleotides from one of the sequences of Tables 1 and 2, and differing in their ability to inhibit the expression of the PCSK9 gene in a FACS assay as described herein below by not more than 5, 10, 15, 20, 25, or 30 % inhibition from a dsRNA comprising the full sequence, are contemplated by the invention. Further dsRNAs that cleave within the target sequence provided in Tables 1 and 2 can readily be made using the PCSK9 sequence and the 25 target sequence provided. In addition, the RNAi agents provided in Tables 1 and 2 identify a site in the PCSK9 mRNA that is susceptible to RNAi based cleavage. As such the present invention further includes RNAi agents that target within the sequence targeted by one of the agents of the present invention. As used herein a second RNAi agent is said to target within the sequence of a first 17 RNAi agent if the second RNAi agent cleaves the message anywhere within the mRNA that is complementary to the antisense strand of the first RNAi agent. Such a second agent will generally consist of at least 15 contiguous nucleotides from one of the sequences provided in Tables 1 and 2 coupled to additional nucleotide sequences taken from the region contiguous to 5 the selected sequence in the PCSK9 gene. For example, the last 15 nucleotides of SEQ ID NO: 1 (minus the added AA sequences) combined with the next 6 nucleotides from the target PCSK9 gene produces a single strand agent of 21 nucleotides that is based on one of the sequences provided in Tables 1 and 2. The dsRNA of the invention can contain one or more mismatches to the target sequence. 0 In a preferred embodiment, the dsRNA of the invention contains no more than 3 mismatches. If the antisense strand of the dsRNA contains mismatches to a target sequence, it is preferable that the area of mismatch not be located in the center of the region of complementarity. If the antisense strand of the dsRNA contains mismatches to the target sequence, it is preferable that the mismatch be restricted to 5 nucleotides from either end, for example 5, 4, 3, 2, or 1 5 nucleotide from either the 5' or 3' end of the region of complementarity. For example, for a 23 nucleotide dsRNA strand which is complementary to a region of the PCSK9 gene, the dsRNA generally does not contain any mismatch within the central 13 nucleotides. The methods described within the invention can be used to determine whether a dsRNA containing a mismatch to a target sequence is effective in inhibiting the expression of the PCSK9 gene. Consideration of O the efficacy of dsRNAs with mismatches in inhibiting expression of the PCSK9 gene is important, especially if the particular region of complementarity in the PCSK9 gene is known to have polymorphic sequence variation within the population. In one embodiment, at least one end of the dsRNA has a single-stranded nucleotide overhang of 1 to 4, generally 1 or 2 nucleotides. dsRNAs having at least one nucleotide 25 overhang have unexpectedly superior inhibitory properties than their blunt-ended counterparts. Moreover, the present inventors have discovered that the presence of only one nucleotide overhang strengthens the interference activity of the dsRNA, without affecting its overall stability. dsRNA having only one overhang has proven particularly stable and effective in vivo, as well as in a variety of cells, cell culture mediums, blood, and serum. Generally, the single 18 stranded overhang is located at the 3'-terminal end of the antisense strand or, alternatively, at the 3'-terminal end of the sense strand. The dsRNA may also have a blunt end, generally located at the 5'-end of the antisense strand. Such dsRNAs have improved stability and inhibitory activity, thus allowing administration at low dosages, i.e., less than 5 mg/kg body weight of the recipient 5 per day. Generally, the antisense strand of the dsRNA has a nucleotide overhang at the 3'-end, and the 5'-end is blunt. In another embodiment, one or more of the nucleotides in the overhang is replaced with a nucleoside thiophosphate. In yet another embodiment, the dsRNA is chemically modified to enhance stability. The nucleic acids of the invention may be synthesized and/or modified by methods well established in 0 the art, such as those described in "Current protocols in nucleic acid chemistry", Beaucage, S.L. et al. (Edrs.), John Wiley & Sons, Inc., New York, NY, USA, which is hereby incorporated herein by reference. Chemical modifications may include, but are not limited to 2' modifications, modifications at other sites of the sugar or base of an oligonucleotide, introduction of non-natural bases into the olibonucleotide chain, covalent attachment to a ligand or chemical moiety, and 5 replacement of internucleotide phosphate linkages with alternate linkages such as thiophosphates. More than one such modification may be employed. Chemical linking of the two separate dsRNA strands may be achieved by any of a variety of well-known techniques, for example by introducing covalent, ionic or hydrogen bonds; hydrophobic interactions, van der Waals or stacking interactions; by means of metal-ion O coordination, or through use of purine analogues. Generally, the chemical groups that can be used to modify the dsRNA include, without limitation, methylene blue; bifunctional groups, generally bis-(2-chloroethyl)amine; N-acetyl-N'-(p-glyoxylbenzoyl)cystamine; 4-thiouracil; and psoralen. In one embodiment, the linker is a hexa-ethylene glycol linker. In this case, the dsRNA are produced by solid phase synthesis and the hexa-ethylene glycol linker is incorporated 25 according to standard methods (e.g., Williams, D.J., and K.B. Hall, Biochem. (1996) 35:14665 14670). In a particular embodiment, the 5'-end of the antisense strand and the 3'-end of the sense strand are chemically linked via a hexaethylene glycol linker. In another embodiment, at least one nucleotide of the dsRNA comprises a phosphorothioate or phosphorodithioate groups. The 19 chemical bond at the ends of the dsRNA is generally formed by triple-helix bonds. Tables 1 and 2 provides examples of modified RNAi agents of the invention. In yet another embodiment, the nucleotides at one or both of the two single strands may be modified to prevent or inhibit the degradation activities of cellular enzymes, such as, for 5 example, without limitation, certain nucleases. Techniques for inhibiting the degradation activity of cellular enzymes against nucleic acids are known in the art including, but not limited to, 2' amino modifications, 2'-amino sugar modifications, 2'-F sugar modifications, 2'-F modifications, 2'-alkyl sugar modifications, uncharged backbone modifications, morpholino modifications, 2'-O-methyl modifications, and phosphoramidate (see, e.g., Wagner, Nat. Med. 0 (1995) 1:1116-8). Thus, at least one 2'-hydroxyl group of the nucleotides on a dsRNA is replaced by a chemical group, generally by a 2'-amino or a 2'-methyl group. Also, at least one nucleotide may be modified to form a locked nucleotide. Such locked nucleotide contains a methylene bridge that connects the 2'-oxygen of ribose with the 4'-carbon of ribose. Oligonucleotides containing the locked nucleotide are described in Koshkin, A.A., et al., 5 Tetrahedron (1998), 54: 3607-3630) and Obika, S. et al., Tetrahedron Lett. (1998), 39: 5401 5404). Introduction of a locked nucleotide into an oligonucleotide improves the affinity for complementary sequences and increases the melting temperature by several degrees (Braasch, D.A. and D.R. Corey, Chem. Biol. (2001), 8:1-7). Conjugating a ligand to a dsRNA can enhance its cellular absorption as well as targeting 0 to a particular tissue or uptake by specific types of cells such as liver cells. In certain instances, a hydrophobic ligand is conjugated to the dsRNA to facilitate direct permeation of the cellular membrane and or uptake across the liver cells. Alternatively, the ligand conjugated to the dsRNA is a substrate for receptor-mediated endocytosis. These approaches have been used to facilitate cell permeation of antisense oligonucleotides as well as dsRNA agents. For example, 25 cholesterol has been conjugated to various antisense oligonucleotides resulting in compounds that are substantially more active compared to their non-conjugated analogs. See M. Manoharan Antisense & Nucleic Acid Drug Development 2002, 12, 103. Other lipophilic compounds that have been conjugated to oligonucleotides include 1-pyrene butyric acid, 1,3-bis-O (hexadecyl)glycerol, and menthol. One example of a ligand for receptor-mediated endocytosis is 20 folic acid. Folic acid enters the cell by folate-receptor-mediated endocytosis. dsRNA compounds bearing folic acid would be efficiently transported into the cell via the folate receptor-mediated endocytosis. Li and coworkers report that attachment of folic acid to the 3' terminus of an oligonucleotide resulted in an 8-fold increase in cellular uptake of the 5 oligonucleotide. Li, S.; Deshmukh, H. M.; Huang, L. Pharm. Res. 1998, 15, 1540. Other ligands that have been conjugated to oligonucleotides include polyethylene glycols, carbohydrate clusters, cross-linking agents, porphyrin conjugates, delivery peptides and lipids such as cholesterol. In certain instances, conjugation of a cationic ligand to oligonucleotides results in 0 improved resistance to nucleases. Representative examples of cationic ligands are propylammonium and dimethylpropylammonium. Interestingly, antisense oligonucleotides were reported to retain their high binding affinity to mRNA when the cationic ligand was dispersed throughout the oligonucleotide. See M. Manoharan Antisense & Nucleic Acid Drug Development 2002, 12, 103 and references therein. 5 The ligand-conjugated dsRNA of the invention may be synthesized by the use of a dsRNA that bears a pendant reactive functionality, such as that derived from the attachment of a linking molecule onto the dsRNA. This reactive oligonucleotide may be reacted directly with commercially-available ligands, ligands that are synthesized bearing any of a variety of protecting groups, or ligands that have a linking moiety attached thereto. The methods of the 0 invention facilitate the synthesis of ligand-conjugated dsRNA by the use of, in some preferred embodiments, nucleoside monomers that have been appropriately conjugated with ligands and that may further be attached to a solid-support material. Such ligand-nucleoside conjugates, optionally attached to a solid-support material, are prepared according to some preferred embodiments of the methods of the invention via reaction of a selected serum-binding ligand 25 with a linking moiety located on the 5' position of a nucleoside or oligonucleotide. In certain instances, an dsRNA bearing an aralkyl ligand attached to the 3'-terminus of the dsRNA is prepared by first covalently attaching a monomer building block to a controlled-pore-glass support via a long-chain aminoalkyl group. Then, nucleotides are bonded via standard solid phase synthesis techniques to the monomer building-block bound to the solid support. The 21 monomer building block may be a nucleoside or other organic compound that is compatible with solid-phase synthesis. The dsRNA used in the conjugates of the invention may be conveniently and routinely made through the well-known technique of solid-phase synthesis. Equipment for such synthesis 5 is sold by several vendors including, for example, Applied Biosystems (Foster City, CA). Any other means for such synthesis known in the art may additionally or alternatively be employed. It is also known to use similar techniques to prepare other oligonucleotides, such as the phosphorothioates and alkylated derivatives. Teachings regarding the synthesis of particular modified oligonucleotides may be found 0 in the following U.S. patents: U.S. Pat. Nos. 5,138,045 and 5,218,105, drawn to polyamine conjugated oligonucleotides; U.S. Pat. No. 5,212,295, drawn to monomers for the preparation of oligonucleotides having chiral phosphorus linkages; U.S. Pat. Nos. 5,378,825 and 5,541,307, drawn to oligonucleotides having modified backbones; U.S. Pat. No. 5,386,023, drawn to backbone-modified oligonucleotides and the preparation thereof through reductive coupling; U.S. 5 Pat. No. 5,457,191, drawn to modified nucleobases based on the 3-deazapurine ring system and methods of synthesis thereof; U.S. Pat. No. 5,459,255, drawn to modified nucleobases based on N-2 substituted purines; U.S. Pat. No. 5,521,302, drawn to processes for preparing oligonucleotides having chiral phosphorus linkages; U.S. Pat. No. 5,539,082, drawn to peptide nucleic acids; U.S. Pat. No. 5,554,746, drawn to oligonucleotides having P-lactam backbones; 0 U.S. Pat. No. 5,571,902, drawn to methods and materials for the synthesis of oligonucleotides; U.S. Pat. No. 5,578,718, drawn to nucleosides having alkylthio groups, wherein such groups may be used as linkers to other moieties attached at any of a variety of positions of the nucleoside; U.S. Pat. Nos. 5,587,361 and 5,599,797, drawn to oligonucleotides having phosphorothioate linkages of high chiral purity; U.S. Pat. No. 5,506,351, drawn to processes for the preparation of 25 2'-O-alkyl guanosine and related compounds, including 2,6-diaminopurine compounds; U.S. Pat. No. 5,587,469, drawn to oligonucleotides having N-2 substituted purines; U.S. Pat. No. 5,587,470, drawn to oligonucleotides having 3-deazapurines; U.S. Pat. No. 5,223,168, and U.S. Pat. No. 5,608,046, both drawn to conjugated 4'-desmethyl nucleoside analogs; U.S. Pat. Nos. 5,602,240, and 5,610,289, drawn to backbone-modified oligonucleotide analogs; U.S. Pat. Nos. 22 6,262,241, and 5,459,255, drawn to, inter alia, methods of synthesizing 2'-fluoro oligonucleotides. In the ligand-conjugated dsRNA and ligand-molecule bearing sequence-specific linked nucleosides of the invention, the oligonucleotides and oligonucleosides may be assembled on a 5 suitable DNA synthesizer utilizing standard nucleotide or nucleoside precursors, or nucleotide or nucleoside conjugate precursors that already bear the linking moiety, ligand-nucleotide or nucleoside-conjugate precursors that already bear the ligand molecule, or non-nucleoside ligand bearing building blocks. When using nucleotide-conjugate precursors that already bear a linking moiety, the 0 synthesis of the sequence-specific linked nucleosides is typically completed, and the ligand molecule is then reacted with the linking moiety to form the ligand-conjugated oligonucleotide. Oligonucleotide conjugates bearing a variety of molecules such as steroids, vitamins, lipids and reporter molecules, has previously been described (see Manoharan et al., PCT Application WO 93/07883). In a preferred embodiment, the oligonucleotides or linked nucleosides of the 5 invention are synthesized by an automated synthesizer using phosphoramidites derived from ligand-nucleoside conjugates in addition to the standard phosphoramidites and non-standard phosphoramidites that are commercially available and routinely used in oligonucleotide synthesis. The incorporation of a 2'-O-methyl, 2'-O-ethyl, 2'-O-propyl, 2'-O-allyl, 2'-O-aminoalkyl 20 or 2'-deoxy-2'-fluoro group in nucleosides of an oligonucleotide confers enhanced hybridization properties to the oligonucleotide. Further, oligonucleotides containing phosphorothioate backbones have enhanced nuclease stability. Thus, functionalized, linked nucleosides of the invention can be augmented to include either or both a phosphorothioate backbone or a 2'-0 methyl, 2'-O-ethyl, 2'-O-propyl, 2'-O-aminoalkyl, 2'-O-allyl or 2'-deoxy-2'-fluoro group. A 25 summary listing of some of the oligonucleotide modifications known in the art is found at, for example, PCT Publication WO 200370918. 23 In some embodiments, functionalized nucleoside sequences of the invention possessing an amino group at the 5'-terminus are prepared using a DNA synthesizer, and then reacted with an active ester derivative of a selected ligand. Active ester derivatives are well known to those skilled in the art. Representative active esters include N-hydrosuccinimide esters, 5 tetrafluorophenolic esters, pentafluorophenolic esters and pentachlorophenolic esters. The reaction of the amino group and the active ester produces an oligonucleotide in which the selected ligand is attached to the 5'-position through a linking group. The amino group at the 5' terminus can be prepared utilizing a 5'-Amino-Modifier C6 reagent. In one embodiment, ligand molecules may be conjugated to oligonucleotides at the 5'-position by the use of a ligand 0 nucleoside phosphoramidite wherein the ligand is linked to the 5'-hydroxy group directly or indirectly via a linker. Such ligand-nucleoside phosphoramidites are typically used at the end of an automated synthesis procedure to provide a ligand-conjugated oligonucleotide bearing the ligand at the 5'-terminus. Examples of modified internucleoside linkages or backbones include, for example, 5 phosphorothioates, chiral phosphorothioates, phosphorodithioates, phosphotriesters, aminoalkylphosphotriesters, methyl and other alkyl phosphonates including 3'-alkylene phosphonates and chiral phosphonates, phosphinates, phosphoramidates including 3'-amino phosphoramidate and aminoalkylphosphoramidates, thionophosphoramidates, thionoalkylphosphonates, thionoalkylphosphotriesters, and boranophosphates having normal Y-5' O linkages, 2'-5' linked analogs of these, and those having inverted polarity wherein the adjacent pairs of nucleoside units are linked 3'-5' to 5'-3' or 2'-5' to 5'-2'. Various salts, mixed salts and free-acid forms are also included. Representative United States Patents relating to the preparation of the above phosphorus atom-containing linkages include, but are not limited to, U.S. Pat. Nos. 3,687,808; 4,469,863; 25 4,476,301; 5,023,243; 5,177,196; 5,188,897; 5,264,423; 5,276,019; 5,278,302; 5,286,717; 5,321,131; 5,399,676; 5,405,939; 5,453,496; 5,455,233; 5,466,677; 5,476,925; 5,519,126; 5,536,821; 5,541,306; 5,550,111; 5,563,253; 5,571,799; 5,587,361; 5,625,050; and 5,697,248, each of which is herein incorporated by reference. 24 Examples of modified internucleoside linkages or backbones that do not include a phosphorus atom therein (i.e., oligonucleosides) have backbones that are formed by short chain alkyl or cycloalkyl intersugar linkages, mixed heteroatom and alkyl or cycloalkyl intersugar linkages, or one or more short chain heteroatomic or heterocyclic intersugar linkages. These 5 include those having morpholino linkages (formed in part from the sugar portion of a nucleoside); siloxane backbones; sulfide, sulfoxide and sulfone backbones; formacetyl and thioformacetyl backbones; methylene formacetyl and thioformacetyl backbones; alkene containing backbones; sulfamate backbones; methyleneimino and methylenehydrazino backbones; sulfonate and sulfonamide backbones; amide backbones; and others having mixed N, 0 0, S and CH 2 component parts. Representative United States patents relating to the preparation of the above oligonucleosides include, but are not limited to, U.S. Pat. Nos. 5,034,506; 5,166,315; 5,185,444; 5,214,134; 5,216,141; 5,235,033; 5,264,562; 5,264,564; 5,405,938; 5,434,257; 5,466,677; 5,470,967; 5,489,677; 5,541,307; 5,561,225; 5,596,086; 5,602,240; 5,610,289; 5,602,240; 5 5,608,046; 5,610,289; 5,618,704; 5,623,070; 5,663,312; 5,633,360; 5,677,437; and 5,677,439, each of which is herein incorporated by reference. In certain instances, the oligonucleotide may be modified by a non-ligand group. A number of non-ligand molecules have been conjugated to oligonucleotides in order to enhance the activity, cellular distribution or cellular uptake of the oligonucleotide, and procedures for O performing such conjugations are available in the scientific literature. Such non-ligand moieties have included lipid moieties, such as cholesterol (Letsinger et al., Proc. Natl. Acad. Sci. USA, 1989, 86:6553), cholic acid (Manoharan et al., Bioorg. Med. Chem. Lett., 1994, 4:1053), a thioether, e.g., hexyl-S-tritylthiol (Manoharan et al., Ann. N.Y. Acad. Sci., 1992, 660:306; Manoharan et al., Bioorg. Med. Chem. Let., 1993, 3:2765), a thiocholesterol (Oberhauser et al., 25 Nucl. Acids Res., 1992, 20:533), an aliphatic chain, e.g., dodecandiol or undecyl residues (Saison-Behmoaras et al., EMBO J., 1991, 10:111; Kabanov et al., FEBS Lett., 1990, 259:327; Svinarchuk et al., Biochimie, 1993, 75:49), a phospholipid, e.g., di-hexadecyl-rac-glycerol or triethylammonium 1,2-di-0-hexadecyl-rac-glycero-3-H-phosphonate (Manoharan et al., Tetrahedron Lett., 1995, 36:365 1; Shea et al., Nucl. Acids Res., 1990, 18:3777), a polyamine or a 25 polyethylene glycol chain (Manoharan et al., Nucleosides & Nucleotides, 1995, 14:969), or adamantane acetic acid (Manoharan et al., Tetrahedron Lett., 1995, 36:3651), a palmityl moiety (Mishra et al., Biochim. Biophys. Acta, 1995, 1264:229), or an octadecylamine or hexylamino carbonyl-oxycholesterol moiety (Crooke et al., J. Pharmacol. Exp. Ther., 1996, 277:923). 5 Representative United States patents that teach the preparation of such oligonucleotide conjugates have been listed above. Typical conjugation protocols involve the synthesis of oligonucleotides bearing an aminolinker at one or more positions of the sequence. The amino group is then reacted with the molecule being conjugated using appropriate coupling or activating reagents. The conjugation reaction may be performed either with the oligonucleotide 0 still bound to the solid support or following cleavage of the oligonucleotide in solution phase. Purification of the oligonucleotide conjugate by HPLC typically affords the pure conjugate. The use of a cholesterol conjugate is particularly preferred since such a moiety can increase targeting liver cells cells, a site of PCSK9 expression. Vector encoded RNAi agents 5 The dsRNA of the invention can also be expressed from recombinant viral vectors intracellularly in vivo. The recombinant viral vectors of the invention comprise sequences encoding the dsRNA of the invention and any suitable promoter for expressing the dsRNA sequences. Suitable promoters include, for example, the U6 or HI RNA pol III promoter sequences and the cytomegalovirus promoter. Selection of other suitable promoters is within the O skill in the art. The recombinant viral vectors of the invention can also comprise inducible or regulatable promoters for expression of the dsRNA in a particular tissue or in a particular intracellular environment. The use of recombinant viral vectors to deliver dsRNA of the invention to cells in vivo is discussed in more detail below. dsRNA of the invention can be expressed from a recombinant viral vector either as two 25 separate, complementary RNA molecules, or as a single RNA molecule with two complementary regions. 26 Any viral vector capable of accepting the coding sequences for the dsRNA molecule(s) to be expressed can be used, for example vectors derived from adenovirus (AV); adeno-associated virus (AAV); retroviruses (e.g, lentiviruses (LV), Rhabdoviruses, murine leukemia virus); herpes virus, and the like. The tropism of viral vectors can be modified by pseudotyping the vectors with 5 envelope proteins or other surface antigens from other viruses, or by substituting different viral capsid proteins, as appropriate. For example, lentiviral vectors of the invention can be pseudotyped with surface proteins from vesicular stomatitis virus (VSV), rabies, Ebola, Mokola, and the like. AAV vectors of the invention can be made to target different cells by engineering the vectors to express different 0 capsid protein serotypes. For example, an AAV vector expressing a serotype 2 capsid on a serotype 2 genome is called AAV 2/2. This serotype 2 capsid gene in the AAV 2/2 vector can be replaced by a serotype 5 capsid gene to produce an AAV 2/5 vector. Techniques for constructing AAV vectors which express different capsid protein serotypes are within the skill in the art; see, e.g., Rabinowitz J E et al. (2002), J Virol 76:791-801, the entire disclosure of which is herein 5 incorporated by reference. Selection of recombinant viral vectors suitable for use in the invention, methods for inserting nucleic acid sequences for expressing the dsRNA into the vector, and methods of delivering the viral vector to the cells of interest are within the skill in the art. See, for example, Dornburg R (1995), Gene Therap. 2: 301-3 10; Eglitis M A (1988), Biotechniques 6: 608-614; O Miller A D (1990), Hum Gene Therap. 1: 5-14; Anderson W F (1998), Nature 392: 25-30; and Rubinson D A et al., Nat. Genet. 33: 401-406, the entire disclosures of which are herein incorporated by reference. Preferred viral vectors are those derived from AV and AAV. In a particularly preferred embodiment, the dsRNA of the invention is expressed as two separate, complementary single 25 stranded RNA molecules from a recombinant AAV vector comprising, for example, either the U6 or HI RNA promoters, or the cytomegalovirus (CMV) promoter. 27 A suitable AV vector for expressing the dsRNA of the invention, a method for constructing the recombinant AV vector, and a method for delivering the vector into target cells, are described in Xia H et al. (2002), Nat. Biotech. 20: 1006-1010. Suitable AAV vectors for expressing the dsRNA of the invention, methods for 5 constructing the recombinant AV vector, and methods for delivering the vectors into target cells are described in Samulski R et al. (1987), J. Virol. 61: 3096-3101; Fisher K J et al. (1996), J. Virol, 70: 520-532; Samulski R et al. (1989), J. Virol. 63: 3822-3826; U.S. Pat. No. 5,252,479; U.S. Pat. No. 5,139,941; International Patent Application No. WO 94/13788; and International Patent Application No. WO 93/24641, the entire disclosures of which are herein incorporated by 0 reference. III. Pharmaceutical compositions comprising dsRNA In one embodiment, the invention provides pharmaceutical compositions comprising a dsRNA, as described herein, and a pharmaceutically acceptable carrier. The pharmaceutical composition comprising the dsRNA is useful for treating a disease or disorder associated with the 5 expression or activity of the PCSK9 gene, such as pathological processes which can be mediated by down regulating PCSK9 gene expression, sucxh as hyperlipidemia. Such pharmaceutical compositions are formulated based on the mode of delivery. One example is compositions that are formulated for delivery to the liver via parenteral delivery. The pharmaceutical compositions of the invention are administered in dosages sufficient 20 to inhibit expression of the PCSK9 gene. The present inventors have found that, because of their improved efficiency, compositions comprising the dsRNA of the invention can be administered at surprisingly low dosages. A dosage of 5 mg dsRNA per kilogram body weight of recipient per day is sufficient to inhibit or suppress expression of the PCSK9 gene and may be administered systemically to the patient. 25 In general, a suitable dose of dsRNA will be in the range of 0.01 to 5.0 milligrams per kilogram body weight of the recipient per day, generally in the range of 1 microgram to 1 mg per kilogram body weight per day. The pharmaceutical composition may be administered once daily, 28 or the dsRNA may be administered as two, three, or more sub-doses at appropriate intervals throughout the day or even using continuous infusion or delivery through a controlled release formulation. In that case, the dsRNA contained in each sub-dose must be correspondingly smaller in order to achieve the total daily dosage. The dosage unit can also be compounded for 5 delivery over several days, e.g., using a conventional sustained release formulation which provides sustained release of the dsRNA over a several day period. Sustained release formulations are well known in the art. The skilled artisan will appreciate that certain factors may influence the dosage and timing required to effectively treat a subject, including but not limited to the severity of the 0 disease or disorder, previous treatments, the general health and/or age of the subject, and other diseases present. Moreover, treatment of a subject with a therapeutically effective amount of a composition can include a single treatment or a series of treatments. Estimates of effective dosages and in vivo half-lives for the individual dsRNAs encompassed by the invention can be made using conventional methodologies or on the basis of in vivo testing using an appropriate 5 animal model, as described elsewhere herein. Advances in mouse genetics have generated a number of mouse models for the study of various human diseases, such as pathological processes which can be mediated by down regulating PCSK9 gene expression. Such models are used for in vivo testing of dsRNA, as well as for determining a therapeutically effective dose. 0O Any method can be used to administer a dsRNA of the present invention to a mammal. For example, administration can be direct; oral; or parenteral (e.g., by subcutaneous, intraventricular, intramuscular, or intraperitoneal injection, or by intravenous drip). Administration can be rapid (e.g., by injection), or can occur over a period of time (e.g., by slow infusion or administration of slow release formulations). 25 Typically, when treating a mammal with hyperlipidemia, the dsRNA molecules are administered systemically via parental means. For example, dsRNAs, conjugated or unconjugate or formulated with or without liposomes, can be administered intravenously to a patient. For such, a dsRNA molecule can be formulated into compositions such as sterile and non-sterile 29 aqueous solutions, non-aqueous solutions in common solvents such as alcohols, or solutions in liquid or solid oil bases. Such solutions also can contain buffers, diluents, and other suitable additives. For parenteral, intrathecal, or intraventricular administration, a dsRNA molecule can be formulated into compositions such as sterile aqueous solutions, which also can contain 5 buffers, diluents, and other suitable additives (e.g., penetration enhancers, carrier compounds, and other pharmaceutically acceptable carriers). In addition, dsRNA molecules can be administered to a mammal as biologic or abiologic means as described in, for example, U.S. Pat. No. 6,271,359. Abiologic delivery can be accomplished by a variety of methods including, without limitation, (1) loading liposomes with a 0 dsRNA acid molecule provided herein and (2) complexing a dsRNA molecule with lipids or liposomes to form nucleic acid-lipid or nucleic acid-liposome complexes. The liposome can be composed of cationic and neutral lipids commonly used to transfect cells in vitro. Cationic lipids can complex (e.g., charge-associate) with negatively charged nucleic acids to form liposomes. Examples of cationic liposomes include, without limitation, lipofectin, lipofectamine, 5 lipofectace, and DOTAP. Procedures for forming liposomes are well known in the art. Liposome compositions can be formed, for example, from phosphatidylcholine, dimyristoyl phosphatidylcholine, dipalmitoyl phosphatidylcholine, dimyristoyl phosphatidylglycerol, or dioleoyl phosphatidylethanolamine. Numerous lipophilic agents are commercially available, including Lipofectin.RTM. (Invitrogen/Life Technologies, Carlsbad, Calif.) and Effectene.TM. O (Qiagen, Valencia, Calif.). In addition, systemic delivery methods can be optimized using commercially available cationic lipids such as DDAB or DOTAP, each of which can be mixed with a neutral lipid such as DOPE or cholesterol. In some cases, liposomes such as those described by Templeton et al. (Nature Biotechnology, 15: 647-652 (1997)) can be used. In other embodiments, polycations such as polyethyleneimine can be used to achieve delivery in vivo and 25 ex vivo (Boletta et al., J. Am Soc. Nephrol. 7: 1728 (1996)). Additional information regarding the use of liposomes to deliver nucleic acids can be found in U.S. Pat. No. 6,271,359, PCT Publication WO 96/40964 and Morrissey, D. et al. 2005. Nat Biotechnol. 23(8):1002-7. Biologic delivery can be accomplished by a variety of methods including, without limitation, the use of viral vectors. For example, viral vectors (e.g., adenovirus and herpesvirus 30 vectors) can be used to deliver dsRNA molecules to liver cells. Standard molecular biology techniques can be used to introduce one or more of the dsRNAs provided herein into one of the many different viral vectors previously developed to deliver nucleic acid to cells. These resulting viral vectors can be used to deliver the one or more dsRNAs to cells by, for example, infection. 5 dsRNAs of the present invention can be formulated in a pharmaceutically acceptable carrier or diluent. A "pharmaceutically acceptable carrier" (also referred to herein as an "excipient") is a pharmaceutically acceptable solvent, suspending agent, or any other pharmacologically inert vehicle. Pharmaceutically acceptable carriers can be liquid or solid, and can be selected with the planned manner of administration in mind so as to provide for the 0 desired bulk, consistency, and other pertinent transport and chemical properties. Typical pharmaceutically acceptable carriers include, by way of example and not limitation: water; saline solution; binding agents (e.g., polyvinylpyrrolidone or hydroxypropyl methylcellulose); fillers (e.g., lactose and other sugars, gelatin, or calcium sulfate); lubricants (e.g., starch, polyethylene glycol, or sodium acetate); disintegrates (e.g., starch or sodium starch glycolate); and wetting 5 agents (e.g., sodium lauryl sulfate). In addition, dsRNA that target the PCSK9 gene can be formulated into compositions containing the dsRNA admixed, encapsulated, conjugated, or otherwise associated with other molecules, molecular structures, or mixtures of nucleic acids. For example, a composition containing one or more dsRNA agents that target the PCSK9 gene can contain other therapeutic O agents such as othr lipid lowering agents (e.g., statins). Methods for treating diseases that can be modulated by down regulating the expression of PCSK9 The methods and compositions described herein can be used to treat diseases and conditions that can be modulated by down regulating PCSK9 gene expression. For example, the 25 compositions described herein can be used to treat hyperlipidemia and other forms of lipid inbalance such as hypercholesterolemia, hypertriglyceridemia and the pathological conditions associated with thiese disorders such as heart and circulatory diseases. 31 Methods for inhibiting expression of the PCSK9 gene In yet another aspect, the invention provides a method for inhibiting the expression of the PCSK9 gene in a mammal. The method comprises administering a composition of the invention to the mammal such that expression of the target PCSK9 gene is silenced. Because of their high 5 specificity, the dsRNAs of the invention specifically target RNAs (primary or processed) of the target PCSK9 gene. Compositions and methods for inhibiting the expression of these PCSK9 genes using dsRNAs can be performed as described elsewhere herein. In one embodiment, the method comprises administering a composition comprising a dsRNA, wherein the dsRNA comprises a nucleotide sequence which is complementary to at least 0 a part of an RNA transcript of the PCSK9 gene of the mammal to be treated. When the organism to be treated is a mammal such as a human, the composition may be administered by any means known in the art including, but not limited to oral or parenteral routes, including intravenous, intramuscular, subcutaneous, transdermal, airway (aerosol) administration. In preferred embodiments, the compositions are administered by intravenous infusion or injection. 5 Unless otherwise defined, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. Although methods and materials similar or equivalent to those described herein can be used in the practice or testing of the invention, suitable methods and materials are described below. All publications, patent applications, patents, and other references mentioned herein are O incorporated by reference in their entirety. In case of conflict, the present specification, including definitions, will control. In addition, the materials, methods, and examples are illustrative only and not intended to be limiting. 32 EXAMPLES Gene Walking of the PCSK9 gene siRNA design was carried out to identify in two separate selections a) siRNAs targeting PCSK9 human and either mouse or rat mRNA and 5 b) all human reactive siRNAs with predicted specificity to the target gene PCSK9. mRNA sequences to human, mouse and rat PCSK9 were used: Human sequence NM_174936.2 was used as reference sequence during the complete siRNA selection procedure. 19 mer stretches conserved in human and mouse, and human and rat PCSK9 mRNA sequences were identified in the first step, resulting in the selection of siRNAs crossreactive to 0 human and mouse, and siRNAs crossreactive to human and rat targets SiRNAs specifically targeting human PCSK9 were identified in a second selection. All potential 19mer sequences of human PCSK9 were extracted and defined as candidate target sequences. Sequences cross-reactive to human, monkey, and those cross-reactive to mouse, rat, human and monkey are all listed in Tables 1 and 2. Chemically modified versions of those 5 sequences and their activity in both in vitro and in vivo assays are also listed in tables 1 and 2 and examples given in Figures 2-8. In order to rank candidate target sequences and their corresponding siRNAs and select appropriate ones, their predicted potential for interacting with irrelevant targets (off-target potential) was taken as a ranking parameter. siRNAs with low off-target potential were defined 20 as preferable and assumed to be more specific in vivo. For predicting siRNA-specific off-target potential, the following assumptions were made: 1) positions 2 to 9 (counting 5' to 3') of a strand (seed region) may contribute more to off-target potential than rest of sequence (non-seed and cleavage site region) 33 2) positions 10 and 11 (counting 5' to 3') of a strand (cleavage site region) may contribute more to off-target potential than non-seed region 3) positions 1 and 19 of each strand are not relevant for off-target interactions 4) an off-target score can be calculated for each gene and each strand, based on 5 complementarity of siRNA strand sequence to the gene's sequence and position of mismatches 5) number of predicted off-targets as well as highest off-target score must be considered for off-target potential 6) off-target scores are to be considered more relevant for off-target potential than numbers of off-targets 0 7) assuming potential abortion of sense strand activity by internal modifications introduced, only off-target potential of antisense strand will be relevant To identify potential off-target genes, 19mer candidate sequences were subjected to a homology search against publically available human mRNA sequences. The following off-target properties for each 19mer input sequence were extracted for each 5 off-target gene to calculate the off-target score: Number of mismatches in non-seed region Number of mismatches in seed region Number of mismatches in cleavage site region The off-target score was calculated for considering assumption 1 to 3 as follows: 20 Off-target score number of seed mismatches * 10 + number of cleavage site mismatches * 1.2 + number of non-seed mismatches * 1 34 The most relevant off-target gene for each siRNA corresponding to the input 19mer sequence was defined as the gene with the lowest off-target score. Accordingly, the lowest off target score was defined as the relevant off-target score for each siRNA. dsRNA synthesis 5 Source of reagents Where the source of a reagent is not specifically given herein, such reagent may be obtained from any supplier of reagents for molecular biology at a quality/purity standard for application in molecular biology. siRNA synthesis 0 Single-stranded RNAs were produced by solid phase synthesis on a scale of 1 pmole using an Expedite 8909 synthesizer (Applied Biosystems, Applera Deutschland GmbH, Darmstadt, Germany) and controlled pore glass (CPG, 500A, Proligo Biochemie GmbH, Hamburg, Germany) as solid support. RNA and RNA containing 2'-0-methyl nucleotides were generated by solid phase synthesis employing the corresponding phosphoramidites and 2'-0 5 methyl phosphoramidites, respectively (Proligo Biochemie GmbH, Hamburg, Germany). These building blocks were incorporated at selected sites within the sequence of the oligoribonucleotide chain using standard nucleoside phosphoramidite chemistry such as described in Current protocols in nucleic acid chemistry, Beaucage, S.L. et al. (Edrs.), John Wiley & Sons, Inc., New York, NY, USA. Phosphorothioate linkages were introduced by replacement of the iodine 20 oxidizer solution with a solution of the Beaucage reagent (Chruachem Ltd, Glasgow, UK) in acetonitrile (1%). Further ancillary reagents were obtained from Mallinckrodt Baker (Griesheim, Germany). Deprotection and purification of the crude oligoribonucleotides by anion exchange HPLC were carried out according to established procedures. Yields and concentrations were determined 25 by UV absorption of a solution of the respective RNA at a wavelength of 260 nm using a spectral photometer (DU 640B, Beckman Coulter GmbH, UnterschleiBheim, Germany). Double stranded RNA was generated by mixing an equimolar solution of complementary strands in annealing 35 buffer (20 mM sodium phosphate, pH 6.8; 100 mM sodium chloride), heated in a water bath at 85 - 90'C for 3 minutes and cooled to room temperature over a period of 3 - 4 hours. The annealed RNA solution was stored at -20 'C until use. For the synthesis of 3'-cholesterol-conjugated siRNAs (herein referred to as -Chol-3'), an 5 appropriately modified solid support was used for RNA synthesis. The modified solid support was prepared as follows: Diethyl-2-azabutane- 1,4-dicarboxylate AA 0 N H 6 AA A 4.7 M aqueous solution of sodium hydroxide (50 mL) was added into a stirred, ice 0 cooled solution of ethyl glycinate hydrochloride (32.19 g, 0.23 mole) in water (50 mL). Then, ethyl acrylate (23.1 g, 0.23 mole) was added and the mixture was stirred at room temperature until completion of the reaction was ascertained by TLC. After 19 h the solution was partitioned with dichloromethane (3 x 100 mL). The organic layer was dried with anhydrous sodium sulfate, filtered and evaporated. The residue was distilled to afford AA (28.8 g, 6 1%). 5 3- { Ethoxycarbonylmethyl-[6-(9H-fluoren-9-ylmethoxycarbonyl-amino)-hexanoyl] amino I -propionic acid ethyl ester AB 0 O N FmocHN 0 0 AB Fmoc-6-amino-hexanoic acid (9.12 g, 25.83 mmol) was dissolved in dichloromethane (50 20 mL) and cooled with ice. Diisopropylcarbodiimde (3.25 g, 3.99 mL, 25.83 mmol) was added to the solution at 0 0 C. It was then followed by the addition of Diethyl-azabutane-1,4-dicarboxylate 36 (5 g, 24.6 mmol) and dimethylamino pyridine (0.305 g, 2.5 mmol). The solution was brought to room temperature and stirred further for 6 h. Completion of the reaction was ascertained by TLC. The reaction mixture was concentrated under vacuum and ethyl acetate was added to precipitate diisopropyl urea. The suspension was filtered. The filtrate was washed with 5% aqueous 5 hydrochloric acid, 5% sodium bicarbonate and water. The combined organic layer was dried over sodium sulfate and concentrated to give the crude product which was purified by column chromatography (50 % EtOAC/Hexanes) to yield 11.87 g (88%) of AB. 3-[(6-Amino-hexanoyl)-ethoxycarbonylmethyl-amino]-propionic acid ethyl ester AC 0 N
H
2 N 0 0 AC 3- { Ethoxycarbonylmethyl-[6-(9H-fluoren-9-ylmethoxycarbonylamino)-hexanoyl] amino I -propionic acid ethyl ester AB (11.5 g, 21.3 mmol) was dissolved in 20% piperidine in dimethylformamide at 0 0 C. The solution was continued stirring for 1 h. The reaction mixture was concentrated under vacuum, water was added to the residue, and the product was extracted with 5 ethyl acetate. The crude product was purified by conversion into its hydrochloride salt. 3-({6-[17-(1,5-Dimethyl-hexyl)-10,13-dimethyl-2,3,4,7,8,9,10,11,12,13,14,15,16,17 tetradecahydro-1H-cyclopenta[a]phenanthren-3-yloxycarbonylamino] hexanoyl I ethoxycarbonylmethyl-amino)-propionic acid ethyl ester AD 0 O N O N O0 0 20 AD 37 The hydrochloride salt of 3-[(6-Amino-hexanoyl)-ethoxycarbonylmethyl-amino] propionic acid ethyl ester AC (4.7 g, 14.8 mmol) was taken up in dichloromethane. The suspension was cooled to 0 0 C on ice. To the suspension diisopropylethylamine (3.87 g, 5.2 mL, 30 mmol) was added. To the resulting solution cholesteryl chloroformate (6.675 g, 14.8 mmol) 5 was added. The reaction mixture was stirred overnight. The reaction mixture was diluted with dichloromethane and washed with 10% hydrochloric acid. The product was purified by flash chromatography (10.3 g, 92%). 1-{6-[17-(1,5-Dimethyl-hexyl)-10,13-dimethyl-2,3,4,7,8,9,10,11,12,13,14,15,16,17 tetradecahydro-1H-cyclopenta[a] phenanthren-3-yloxycarbonylamino]-hexanoyl}-4-oxo 0 pyrrolidine-3-carboxylic acid ethyl ester AE 0 0O N 00 AE Potassium t-butoxide (1.1 g, 9.8 mmol) was slurried in 30 mL of dry toluene. The mixture was cooled to 0 0 C on ice and 5 g (6.6 mmol) of diester AD was added slowly with stirring within 15 20 mins. The temperature was kept below 5'C during the addition. The stirring was continued for 30 mins at 0 0 C and 1 mL of glacial acetic acid was added, immediately followed by 4 g of NaH 2
PO
4
-H
2 0 in 40 mL of water The resultant mixture was extracted twice with 100 mL of dichloromethane each and the combined organic extracts were washed twice with 10 mL of phosphate buffer each, dried, and evaporated to dryness. The residue was dissolved in 60 mL of 20 toluene, cooled to 0 0 C and extracted with three 50 mL portions of cold pH 9.5 carbonate buffer. The aqueous extracts were adjusted to pH 3 with phosphoric acid, and extracted with five 40 mL portions of chloroform which were combined, dried and evaporated to dryness. The residue was 38 purified by column chromatography using 25% ethylacetate/hexane to afford 1.9 g of b-ketoester (39%). [6-(3-Hydroxy-4-hydroxymethyl-pyrrolidin-1-yl)-6-oxo-hexyl]-carbamic acid 17-(1,5 dimethyl-hexyl)-10,13-dimethyl-2,3,4,7,8,9,10,11,12,13,14,15,16,17-tetradecahydro-1H 5 cyclopenta[a]phenanthren-3-yl ester AF HO OH H OH N 0 Y 0 0 AF Methanol (2 mL) was added dropwise over a period of 1 h to a refluxing mixture of b ketoester AE (1.5 g, 2.2 mmol) and sodium borohydride (0.226 g, 6 mmol) in tetrahydrofuran (10 0 mL). Stirring was continued at reflux temperature for 1 h. After cooling to room temperature, 1 N HCl (12.5 mL) was added, the mixture was extracted with ethylacetate (3 x 40 mL). The combined ethylacetate layer was dried over anhydrous sodium sulfate and concentrated under vacuum to yield the product which was purified by column chromatography (10% MeOH/CHCl 3 ) (89%). 39 (6-{ 3-[Bis-(4-methoxy-phenyl)-phenyl-methoxymethyl]-4-hydroxy-pyrrolidin-1-yl 1-6 oxo-hexyl)-carbamic acid 17-(1,5-dimethyl-hexyl)-10,13-dimethyl 2,3,4,7,8,9,10,11,12,13,14,15,16,17-tetradecahydro-1H-cyclopenta[a]phenanthren-3-yl ester AG
OCH
3 HO N
OCH
3 0 5 AG Diol AF (1.25 gm 1.994 mmol) was dried by evaporating with pyridine (2 x 5 mL) in vacuo. Anhydrous pyridine (10 mL) and 4,4'-dimethoxytritylchloride (0.724 g, 2.13 mmol) were added with stirring. The reaction was carried out at room temperature overnight. The reaction was quenched by the addition of methanol. The reaction mixture was concentrated under vacuum 0 and to the residue dichloromethane (50 mL) was added. The organic layer was washed with IM aqueous sodium bicarbonate. The organic layer was dried over anhydrous sodium sulfate, filtered and concentrated. The residual pyridine was removed by evaporating with toluene. The crude product was purified by column chromatography (2% MeOH/Chloroform, Rf = 0.5 in 5% MeOH/CHCl 3 ) (1.75 g, 95%). 40 Succinic acid mono-(4- [bis-(4-methoxy-phenyl)-phenyl-methoxymethyl] -1- { 6- [17-(1,5 dimethyl-hexyl)-10,13-dimethyl 2,3,4,7,8,9,10,11,12,13,14,15,16,17-tetradecahydro-1H cyclopenta[a]phenanthren-3-yloxycarbonylamino]-hexanoyl}-pyrrolidin-3-yl) ester AH
H
3 CO HO 0 CH 2 0 0 N OCH 3 OHN O' 0 5 AH Compound AG (1.0 g, 1.05 mmol) was mixed with succinic anhydride (0.150 g, 1.5 mmol) and DMAP (0.073 g, 0.6 mmol) and dried in a vacuum at 40'C overnight. The mixture was dissolved in anhydrous dichloroethane (3 mL), triethylamine (0.318 g, 0.440 mL, 3.15 mmol) was added and the solution was stirred at room temperature under argon atmosphere for 0 16 h. It was then diluted with dichloromethane (40 mL) and washed with ice cold aqueous citric acid (5 wt%, 30 mL) and water (2 X 20 mL). The organic phase was dried over anhydrous sodium sulfate and concentrated to dryness. The residue was used as such for the next step. Cholesterol derivatised CPG AI
H
3 CO O HN 0 CH 2 0 0 H OCH 3 OHN O' 0 r 0 15 Al 41 Succinate AH (0.254 g, 0.242 mmol) was dissolved in a mixture of dichloromethane/acetonitrile (3:2, 3 mL). To that solution DMAP (0.0296 g, 0.242 mmol) in acetonitrile (1.25 mL), 2,2'-Dithio-bis(5-nitropyridine) (0.075 g, 0.242 mmol) in acetonitrile/dichloroethane (3:1, 1.25 mL) were added successively. To the resulting solution 5 triphenylphosphine (0.064 g, 0.242 mmol) in acetonitrile (0.6 ml) was added. The reaction mixture turned bright orange in color. The solution was agitated briefly using a wrist-action shaker (5 mins). Long chain alkyl amine-CPG (LCAA-CPG) (1.5 g, 61 mM) was added. The suspension was agitated for 2 h. The CPG was filtered through a sintered funnel and washed with acetonitrile, dichloromethane and ether successively. Unreacted amino groups were masked using 0 acetic anhydride/pyridine. The achieved loading of the CPG was measured by taking UV measurement (37 mM/g). The synthesis of siRNAs bearing a 5'-12-dodecanoic acid bisdecylamide group (herein referred to as "5'-C32-") or a 5'-cholesteryl derivative group (herein referred to as "5'-Chol-") was performed as described in WO 2004/065601, except that, for the cholesteryl derivative, the 5 oxidation step was performed using the Beaucage reagent in order to introduce a phosphorothioate linkage at the 5'-end of the nucleic acid oligomer. Nucleic acid sequences are represented below using standard nomenclature, and specifically the abbreviations of Table 1-2. Table 1-2: Abbreviations of nucleotide monomers used in nucleic acid sequence ,0 representation. It will be understood that these monomers, when present in an oligonucleotide, are mutually linked by 5'-3'-phosphodiester bonds. Abbreviation Nucleotide(s) A, a 2'-deoxy-adenosine-5'-phosphate, adenosine-5'-phosphate C, c 2'-deoxy-cytidine-5'-phosphate, cytidine-5'-phosphate G, g 2'-deoxy-guanosine-5'-phosphate, guanosine-5'-phosphate T, t 2'-deoxy-thymidine-5'-phosphate, thymidine-5'-phosphate U, u 2'-deoxy-uridine-5'-phosphate, uridine-5'-phosphate N, n any 2'-deoxy-nucleotide/nucleotide (G, A, C, or T, g, a, c or u) Am 2'-O-methyladenosine-5'-phosphate Cm 2'-O-methylcytidine-5'-phosphate 42 Abbreviation Nucleotide(s) Gm 2'-0-methylguanosine-5'-phosphate Tm 2'-O-methyl-thymidine-5'-phosphate Um 2'-O-methyluridine-5'-phosphate Af 2'-fluoro-2'-deoxy-adenosine-5'-phosphate Cf 2'-fluoro-2'-deoxy-cytidine-5'-phosphate Gf 2'-fluoro-2'-deoxy-guanosine-5'-phosphate Tf 2'-fluoro-2'-deoxy-thymidine-5'-phosphate Uf 2'-fluoro-2'-deoxy-uridine-5'-phosphate A, C, G, T, U, a underlined: nucleoside-5'-phosphorothioate c, g, t, u am, cm, gm, tm, underlined: 2-0-methyl-nucleoside-5'-phosphorothioate um capital letters represent 2'-deoxyribonucleotides (DNA), lower case letters represent ribonucleotides (RNA) PCSK9 siRNA screening in HuH7 , HepG2, Hela and Primary Monkey Hepatocytes Discovers Highly Active Sequences 5 HuH-7cells were obtained from JCRB Cell Bank (Japanese Collection of Research Bioresources) (Shinjuku, Japan, cat. No.: JCRB0403) Cells were cultured in Dulbecco's MEM (Biochrom AG, Berlin, Germany, cat. No. F0435) supplemented to contain 10% fetal calf serum (FCS) (Biochrom AG, Berlin, Germany, cat. No. S0115), Penicillin 100 U/ml, Streptomycin 100 pg/ml (Biochrom AG, Berlin, Germany, cat. No. A2213) and 2mM L-Glutamin (Biochrom AG, 10 Berlin, Germany, cat. No K0282) at 37 0 C in an atmosphere with 5% CO 2 in a humidified incubator (Heraeus HERAcell, Kendro Laboratory Products, Langenselbold, Germany). HepG2 and Hela cells were obtained from American Type Culture Collection (Rockville, MD, cat. No. HB-8065) and cultured in MEM (Gibco Invitrogen, Karlsruhe, Germany, cat. No. 21090-022) supplemented to contain 10% fetal calf serum (FCS) (Biochrom AG, Berlin, Germany, cat. No. 15 SO115), Penicillin 100 U/ml, Streptomycin 100 p g/ml (Biochrom AG, Berlin, Germany, cat. No. A2213), 1x Non Essential Amino Acids (Biochrom AG, Berlin, Germany, cat. No. K-0293), and 1mM Sodium Pyruvate (Biochrom AG, Berlin, Germany, cat. No. L-0473) at 37 0 C in an 43 atmosphere with 5% CO 2 in a humidified incubator (Heraeus HERAcell, Kendro Laboratory Products, Langenselbold, Germany). For transfection with siRNA, HuH7, HepG2, or Hela cells were seeded at a density of 2.0 x 104 cells/well in 96-well plates and transfected directly. Transfection of siRNA (30nM for 5 single dose screen) was carried out with lipofectamine 2000 (Invitrogen GmbH, Karlsruhe, Germany, cat. No. 11668-019) as described by the manufacturer. 24 hours after transfection HuH7 and HepG2 cells were lysed and PCSK9 mRNA levels were quantified with the Quantigene Explore Kit (Genosprectra, Dumbarton Circle Fremont, USA, cat. No. QG-000-02) according to the protocol. PCSK9 mRNA levels were normalized to 0 GAP-DH mRNA. For each siRNA eight individual datapoints were collected. siRNA duplexes unrelated to PCSK9 gene were used as control. The activity of a given PCSK9 specific siRNA duplex was expressed as percent PCSK9 mRNA concentration in treated cells relative to PCSK9 mRNA concentration in cells treated with the control siRNA duplex. Primary cynomolgus monkey hepatocytes (cryopreserved) were obtained from In vitro 5 Technologies, Inc. (Baltimore, Maryland, USA, cat No M00305) and cultured in InVitroGRO CP Medium (cat No Z99029) at 37'C in an atmosphere with 5% CO 2 in a humidified incubator. For transfection with siRNA, primary cynomolgus monkey cells were seeded on Collagen coated plates (Fisher Scientific, cat. No. 08-774-5) at a density of 3.5 x 104 cells/well in 96-well plates and transfected directly. Transfection of siRNA (eight 2-fold dilution series starting from 20 30nM ) in duplicates was carried out with lipofectamine 2000 (Invitrogen GmbH, Karlsruhe, Germany, cat. No. 11668-019) as described by the manufacturer. 16 hours after transfection medium was changed to fresh InVitroGRO CP Medium with Torpedo Antibiotic Mix (In vitro Technologies, Inc, cat. No Z99000) added. 24 hours after medium change primary cynomolgus monkey cells were lysed and PCSK9 25 mRNA levels were quantified with the Quantigene Explore Kit (Genosprectra, Dumbarton Circle Fremont, USA, cat. No. QG-000-02) according to the protocol. PCSK9 mRNA levels were 44 normalized to GAPDH mRNA. Normalized PCSK9/GAPDH ratios were then compared to PCSK9/GAPDH ratio of lipofectamine 2000 only control. Tables 1-2 (and Figure 6) summarize the results and provides examples of in vitro screens in different cell lines at different doses. Silencing of PCSK9 transcript was expressed as 5 percentage of remaining transcript at a given dose. Highly active sequences are those with less than 70% transcript remaining post treatment with a given siRNA at a dose less than or equal to 100nm. Very active sequences are those that have less than 60% of transcript remaining after treatment with a dose.less than or equal to lOOnM. Active sequences are those that have less than 85% transcript remaining after treatment with a high dose (100nM). Examples of active 0 siRNA's were also screened in vivo in mouse in lipidoid formulations as described below. Active sequences in vitro were also generally active in vivo (See figure Figure 6 example). In vivo Efficacy Screen of PCSK9 siRNAs Formulation Procedure 5 The lipidoid LNP-01-4HCl (MW 1487) (Figure 1), Cholesterol (Sigma-Aldrich), and PEG-Ceramide C16 (Avanti Polar Lipids) were used to prepare lipid-siRNA nanoparticles. Stock solutions of each in ethanol were prepared: LNP-01, 133 mg/mL; Cholesterol, 25 mg/mL, PEG Ceramide C16, 100 mg/mL. LNP-01, Cholesterol, and PEG-Ceramide C16 stock solutions were then combined in a 42:48:10 molar ratio. Combined lipid solution was mixed rapidly with 20 aqueous siRNA (in sodium acetate pH 5) such that the final ethanol concentration was 35-45% and the final sodium acetate concentration was 100-300 mM. Lipid-siRNA nanoparticles formed spontaneously upon mixing. Depending on the desired particle size distribution, the resultant nanoparticle mixture was in some cases extruded through a polycarbonate membrane (100 nm cut-off) using a thermobarrel extruder (Lipex Extruder, Northern Lipids, Inc). In other cases, the 25 extrusion step was omitted. Ethanol removal and simultaneous buffer exchange was accomplished by either dialysis or tangential flow filtration. Buffer was exchanged to phosphate buffered saline (PBS) pH 7.2. 45 Characterization of formulations Formulations prepared by either the standard or extrusion-free method are characterized in a similar manner. Formulations are first characterized by visual inspection. They should be 5 whitish translucent solutions free from aggregates or sediment. Particle size and particle size distribution of lipid-nanoparticles are measured by dynamic light scattering using a Malvern Zetasizer Nano ZS (Malvern, USA). Particles should be 20-300 nm, and ideally, 40-100 nm in size. The particle size distribution should be unimodal. The total siRNA concentration in the formulation, as well as the entrapped fraction, is estimated using a dye exclusion assay. A sample 0 of the formulated siRNA is incubated with the RNA-binding dye Ribogreen (Molecular Probes) in the presence or absence of a formulation disrupting surfactant, 0.5% Triton-X100. The total siRNA in the formulation is determined by the signal from the sample containing the surfactant, relative to a standard curve. The entrapped fraction is determined by subtracting the "free" siRNA content (as measured by the signal in the absence of surfactant) from the total siRNA 5 content. Percent entrapped siRNA is typically >85%. Bolus dosing Bolus dosing of formulated siRNAs in C57/BL6 mice (5/group, 8-10 weeks old, Charles River Laboratories, MA) was performed by tail vein injection using a 27G needle. SiRNAs were formulated in LNP-01 (and then dialyzed against PBS) at 0.5 mg/ml concentration allowing the 20 delivery of the 5mg/kg dose in 10 pl/g body weight. Mice were kept under an infrared lamp for approximately 3 min prior to dosing to ease injection. 48 hour post dosing mice were sacrificed by C0 2 -asphyxiation. 0.2 ml blood was collected by retro-orbital bleeding and the liver was harvested and frozen in liquid nitrogen. Serum and livers were stored at -80'C. 25 Frozen livers were grinded using 6850 Freezer/Mill Cryogenic Grinder (SPEX CentriPrep, Inc) and powders stored at -80'C until analysis. 46 PCSK9 mRNA levels were detected using the branched-DNA technology based kit from QuantiGene Reagent System (Genospectra) according to the protocol. 10-20mg of frozen liver powders was lysed in 600 ul of 0.16 ug/ml Proteinase K (Epicentre, #MPRK092) in Tissue and Cell Lysis Solution (Epicentre, #MTC096H) at 65 0 C for 3hours. Then 10 ul of the lysates were 5 added to 90ul of Lysis Working Reagent (1 volume of stock Lysis Mixture in two volumes of water) and incubated at 52 0 C overnight on Genospectra capture plates with probe sets specific to mouse PCSK9 and mouse GAPDH or cyclophilin B. Nucleic acid sequences for Capture Extender (CE), Label Extender (LE) and blocking (BL) probes were selected from the nucleic acid sequences of PCSK9, GAPDH and cyclophilin B with the help of the QuantiGene 0 ProbeDesigner Software 2.0 (Genospectra, Fremont, CA, USA, cat. No. QG-002-02). Chemo luminescence was read on a Victor2-Light (Perkin Elmer) as Relative light units. The ratio of PCSK9 mRNA to GAPDH or cyclophilin B mRNA in liver lysates was averaged over each treatment group and compared to a control group treated with PBS or a control group treated with an unrelated siRNA (blood coagulation factor VII). 5 Total serum cholesterol in mouse serum was measured using the StanBio Cholesterol LiquiColor kit (StanBio Laboratoriy, Boerne, Texas, USA) according to manufacturer's instructions. Measurements were taken on a Victor2 1420 Multilabel Counter (Perkin Elmer) at 495 nm. 0O Examples 32 PCSK9 siRNAs formulated in LNP-01 liposomes were tested in vivo in a mouse model. The experiment was performed at 5mg/kg siRNA dose and at least 10 PCSK9 siRNAs showed more than 40% PCSK9 mRNA knock down compared to a control group treated with PBS, while control group treated with an unrelated siRNA (blood coagulation factor VII) had no 25 effect (Figures 2-5). Silencing of PCSK9 transcript also coorelated with a lowering of cholesterol in these animals (Figures 4-5). In addition there was a strong coorelation between those molecules that were active in vitro and those active in vivo (Figure 6). Sequences containing different chemical modifications were also screened in vitro (Tables 1 and 2) and in 47 vivo. As an example, less modified sequences 9314 and 9318, and a more modified versions of that sequence 9314-(10792, 10793, and 10796); 9318-(10794, 10795, 10797) were tested both in vitro (In primary monkey hepatocytes) or in vivo (9314 and 10792) formulated in LNP-01. Figure 7 (also see Tables 1 and2) shows that the parent molecules 9314 and 9318 and the 5 modified versions are all active in vitro. Figure 8 as an example shows that both the parent 9314 and the more highly modified 10792 sequences are active in vivo displaying 50-60% silencing of endogenous PCSK9 in mice. Figure 9 furthur exemplifies that activity of other chemically modified versions of the parents 9314 and 10792. 0 dsRNA expression vectors In another aspect of the invention, PCSK9 specific dsRNA molecules that modulate PCSK9 gene expression activity are expressed from transcription units inserted into DNA or RNA vectors (see, e.g., Couture, A, et al., TIG. (1996), 12:5-10; Skillern, A., et al., International PCT Publication No. WO 00/22113, Conrad, International PCT Publication No. WO 00/22114, 5 and Conrad, US Pat. No. 6,054,299). These transgenes can be introduced as a linear construct, a circular plasmid, or a viral vector, which can be incorporated and inherited as a transgene integrated into the host genome. The transgene can also be constructed to permit it to be inherited as an extrachromosomal plasmid (Gassmann, et al., Proc. Nati. Acad. Sci. USA (1995) 92:1292). The individual strands of a dsRNA can be transcribed by promoters on two separate 20 expression vectors and co-transfected into a target cell. Alternatively each individual strand of the dsRNA can be transcribed by promoters both of which are located on the same expression plasmid. In a preferred embodiment, a dsRNA is expressed as an inverted repeat joined by a linker polynucleotide sequence such that the dsRNA has a stem and loop structure. The recombinant dsRNA expression vectors are generally DNA plasmids or viral vectors. 25 dsRNA expressing viral vectors can be constructed based on, but not limited to, adeno-associated virus (for a review, see Muzyczka, et al., Curr. Topics Micro. Immunol. (1992) 158:97-129)); adenovirus (see, for example, Berkner, et al., BioTechniques (1998) 6:616), Rosenfeld et al. 48 (1991, Science 252:431-434), and Rosenfeld et al. (1992), Cell 68:143-155)); or alphavirus as well as others known in the art. Retroviruses have been used to introduce a variety of genes into many different cell types, including epithelial cells, in vitro and/or in vivo (see, e.g., Eglitis, et al., Science (1985) 230:1395-1398; Danos and Mulligan, Proc. NatI. Acad. Sci. USA (1998) 5 85:6460-6464; Wilson et al., 1988, Proc. NatI. Acad. Sci. USA 85:3014-3018; Armentano et al., 1990, Proc. NatI. Acad. Sci. USA 87:61416145; Huber et al., 1991, Proc. NatI. Acad. Sci. USA 88:8039-8043; Ferry et al., 1991, Proc. NatI. Acad. Sci. USA 88:8377-8381; Chowdhury et al., 1991, Science 254:1802-1805; van Beusechem. et al., 1992, Proc. Nad. Acad. Sci. USA 89:7640 19 ; Kay et al., 1992, Human Gene Therapy 3:641-647; Dai et al., 1992, Proc. Natl.Acad. Sci. 0 USA 89:10892-10895; Hwu et al., 1993, J. Immunol. 150:4104-4115; U.S. Patent No. 4,868,116; U.S. Patent No. 4,980,286; PCT Application WO 89/07136; PCT Application WO 89/02468; PCT Application WO 89/05345; and PCT Application WO 92/07573). Recombinant retroviral vectors capable of transducing and expressing genes inserted into the genome of a cell can be produced by transfecting the recombinant retroviral genome into suitable packaging cell lines 5 such as PA317 and Psi-CRIP (Comette et al., 1991, Human Gene Therapy 2:5-10; Cone et al., 1984, Proc. Natl. Acad. Sci. USA 81:6349). Recombinant adenoviral vectors can be used to infect a wide variety of cells and tissues in susceptible hosts (e.g., rat, hamster, dog, and chimpanzee) (Hsu et al., 1992, J. Infectious Disease, 166:769), and also have the advantage of not requiring mitotically active cells for infection. 0O The promoter driving dsRNA expression in either a DNA plasmid or viral vector of the invention may be a eukaryotic RNA polymerase I (e.g. ribosomal RNA promoter), RNA polymerase II (e.g. CMV early promoter or actin promoter or U1 snRNA promoter) or generally RNA polymerase III promoter (e.g. U6 snRNA or 7SK RNA promoter) or a prokaryotic promoter, for example the T7 promoter, provided the expression plasmid also encodes T7 RNA 25 polymerase required for transcription from a T7 promoter. The promoter can also direct transgene expression to the pancreas (see, e.g. the insulin regulatory sequence for pancreas (Bucchini et al., 1986, Proc. Natl. Acad. Sci. USA 83:2511-2515)). In addition, expression of the transgene can be precisely regulated, for example, by using an inducible regulatory sequence and expression systems such as a regulatory sequence that is 49 sensitive to certain physiological regulators, e.g., circulating glucose levels, or hormones (Docherty et al., 1994, FASEB J. 8:20-24). Such inducible expression systems, suitable for the control of transgene expression in cells or in mammals include regulation by ecdysone, by estrogen, progesterone, tetracycline, chemical inducers of dimerization, and isopropyl-beta-D 1 5 thiogalactopyranoside (EPTG). A person skilled in the art would be able to choose the appropriate regulatory/promoter sequence based on the intended use of the dsRNA transgene. Generally, recombinant vectors capable of expressing dsRNA molecules are delivered as described below, and persist in target cells. Alternatively, viral vectors can be used that provide for transient expression of dsRNA molecules. Such vectors can be repeatedly administered as 0 necessary. Once expressed, the dsRNAs bind to target RNA and modulate its function or expression. Delivery of dsRNA expressing vectors can be systemic, such as by intravenous or intramuscular administration, by administration to target cells ex-planted from the patient followed by reintroduction into the patient, or by any other means that allows for introduction into a desired target cell. 5 dsRNA expression DNA plasmids are typically transfected into target cells as a complex with cationic lipid carriers (e.g. Oligofectamine) or non-cationic lipid-based carriers (e.g. Transit TKOTM ). Multiple lipid transfections for dsRNA-mediated knockdowns targeting different regions of a single PCSK9 gene or multiple PCSK9 genes over a period of a week or more are also contemplated by the invention. Successful introduction of the vectors of the invention into 0 host cells can be monitored using various known methods. For example, transient transfection. can be signaled with a reporter, such as a fluorescent marker, such as Green Fluorescent Protein (GFP). Stable transfection. of ex vivo cells can be ensured using markers that provide the transfected cell with resistance to specific environmental factors (e.g., antibiotics and drugs), such as hygromycin B resistance. 25 The PCSK9 specific dsRNA molecules can also be inserted into vectors and used as gene therapy vectors for human patients. Gene therapy vectors can be delivered to a subject by, for example, intravenous injection, local administration (see U.S. Patent 5,328,470) or by stereotactic injection (see e.g., Chen et al. (1994) Proc. Natl. Acad. Sci. USA 91:3054-3057). The pharmaceutical preparation of the gene therapy vector can include the gene therapy vector in an 50 acceptable diluent, or can comprise a slow release matrix in which the gene delivery vehicle is imbedded. Alternatively, where the complete gene delivery vector can be produced intact from recombinant cells, e.g., retroviral vectors, the pharmaceutical preparation can include one or more cells which produce the gene delivery system. 5 Those skilled in the art are familiar with methods and compositions in addition to those specifically set out in the instant disclosure which will allow them to practice this invention to the full scope of the claims hereinafter appended. It is also to be understood that any discussion of documents, acts, materials, devices, articles or the like which has been included in the present specification is not to be taken as an 0 admission that any or all of these matters form part of the prior art base or were common general knowledge in the field relevant to the present invention as it existed before the priority date of each claim of this application. Throughout this specification the word "comprise", or variations such as "comprises" or "comprising", will be understood to imply the inclusion of a stated element, integer or step, or 5 group of elements, integers or steps, but not the exclusion of any other element, integer or step, or group of elements, integers or steps. 51 C) U 4 fl e < Cu Cu ul u - o e uC a UT <l C- Ut UT OT <T '0 C- QT C UT f 8 a U < QU C -- < U0 n U O O O e uo eu e< eu e C- '0 u u0 5< C<QQOU e U < UO O U uU O U UUCU e OUCU)O E~C U - '0 U < QT - <t flU o Cl U '0 CC O l~ 0 C C t ' C ~ 0 U<< <U U U < cQ < U U~ < ~ U U ( -U< < U (Q( U( U U U <U eU D U < < U < U~ < ( U U < Q( mu U u u ~ < U U < e e u~ (5 e (H ( u~(~ ' O. U (O (Q U < U U < < U 9( U U U U u~( < e e U O U < (e a e < U (- < U U (a o e~( < < 0 U U U < U U U < 0 0 U < U u <o U U U < (O U (Q < U U U U <~ (0 ( <~ ( < - < fl U Q> < U l U < Q> - Q fl Q SU< (Q U O U U Q < (0 U < U~ (O ( U < U e U<e aee o < U H (8 H <~ U o < (O u o < < u (o U U u U u 8 m H cu u~ U e < e e e e~ < u u< eu 3 U (.O U Q < (O U U U < < < 0 < u~ (e U u (e u U e < U u~ o U (e u 8 u~< ~ ( ( U <~ c ~ ~ (~(52 C) o o o~ c- l l~ o - r c c 0 ~ h ~ - ' i ~ h - C c C T o o oo -' '0 '0 8 U 0 5< UOO 8 C<0U ) TC Cl cc c U c O T -U Cl Th Uf e U U~ (C U < U < jU( ~ c a U (O U (O U <. U < Q UUU ~U ~( 0 U Q( U O O < (0( < < -3 < U ( < 0 < U U ( ~( . -O (O O < 0 ( U U <~( ~ ~( < U O (Q U U <~ (0 <( ~(. 0 c0 U U < U - U U U U Q < (0 < O ~( ~ <( U U . U (Q < U U0 U 9 0 0 U U (. U U < < <- U~ U (O<( U U Q( U Q < (u ( <U U (O ( U U < O ( U U ~ ~ '0 U<UO 0 '0 '0 U'0 O < Q c cc (o (O U< U U < ~ (O O U U (Q ( U U U < - 0 U O < U (Q Q U U U (Q( U U UH U H < U (. U O U < U (Q O U U < U < < u U< U U (O U U U O (O (O ( U U U U U- (O U U~( U U < U O (.. U ( U ( u U O < 0 U U cO U O U < 0 U . U < <~ U U < ( 0 < U U O UU UO ~ U)~ U <~ U < U . ( U U O U U U < (0 < < 8 $ < 0U < (Q U < U ( U U O U U U ( U O < oUUU O< U U O U U < cU U U u U as <z ( a a U (.. U (. U < Ca (a (a (a a U U ~U ( U u~ < (~(*~ *~ 53 C) ~ 4 C] C] - C] C] ~Th fl C] C] cc '0 ~t fl ~t ~n ~ cc z~ ~ cc cc C] ~C) C] C] C] Q> ~C) CC - C] C] - C] C] C] C] Q> C] C] Q> Q> CTh fl 5< C. ~C) fl ~Th '0 C] CC fl ~i- CC fl ~ cc C] ,~ ,~t ,~ ,~ ,~ '0CC ,~t '0 C] CC ,a~ ,a~ ,~ cc ,~ '0 CC ~ C] ~t '0 CC ~ C] CC ~ C] ~t '0 CC ~ C] ~t '0 ~ ~ ~ H H H H H H H HHHHH H H H H H ~ (~ < (~ ~ ~ < ~ U(.~ (~ c~ C~ ~ < < C~ ~ ~ ~ U U ~ U U < U U 5(~ < (~ < U U U < U ~ < < (~ ~ ~ U (~ (~ U (~ (..~ ~ U < U < (~ (~ (~ ~ ~ U U U (~ < ~< < U -~U U < ~ < U U U < < U U U ~ (~ (~ U U ~ U (~ (. U (~ (~ U U < < (~ (~ (~ (~ ~ ~ U ~ < (~ (~ < < < U U (~ (~ U (~ U ~ ~ U U ~ c~ ci < < U U U ~ (~ ~ < < U U < < U U ~ U ~ U ~ < ~ (~ ~ U (~ (~ (~ (~ (~ ~ ~ U U (~ U < (~ c~ .- ~ < < (~ (~ U U U (~ (~ U U (~ (~ ~ ~ (~ (~ < ~ < < U U < < < U U U U ~ (~ U U < (~ U <U U < < < < < ~ (~ U U (~ (~ U < U fl C] Q> - ~ fl C] Q> - ~ '~ - ~ '~ - ~ C] C] C] H H H H H H H H H H H H H H H H H H H H H H H H H H H H H H H H H ~ < < U U U U '~ (~ H ~ ~ H ~ H ~ < < U (.~ (~ ~ U (~ (~ ~< (~ U U ~ ~ (~ c~ c~ < c~ < ~ < ~ < U ( U U ~ ~ U < ~ U (~ (~ U ~ U U U (.~ < U < (~ < C)U U ~ U U ~ U U U U (~ U U U U U < -'U < < ~ < U ~ U (~ < < U U ~ U U U U (~ (~ U (~ U (~ U U < < < (~ U U U < U (~ ~ U U U U U U U U U cc '0 '0 Q> ~t C] C] C] fl '0 C] '0 C] ~t fl C] CC C] C] C] C] ~Th fl '0 '0 '0 C] ~ CC CC - - C] C] C] '0 '0 Q> Q> C] CC Q> CC ~t '0 C] Q> CC CC CC CC 54 u )~ Cl C ulC T b ef ' l f ~ l C o m ' l l Cl ' - ' az Zo e l l a-- n 0 e oC '0C ~~C) - Ul Q '0 - Q> C C) o <e au < < -b < e UC C U C~ D (. ( ( U < 0 (< U < 2 .. (U U O U U <U 0 U U Q <~ (Q < (U (Q e U O < 0 < 0 U~ U O (e < < U U< < U U <U < O U < U U U O < < U( U~ U~ U O U < - O l Q - O flC QU - Q fl Cl Q - e fl C U~( U < U (QO < U~ U < U U U <U Q < U (Q U < < (0 H < H Q < ~ <U ( U (~ U <~ U UU SU (.. U < 0 U O U Q < ( ( < U SU U Q U Q (Q < (0 U i < 's < ( ( <U c < e < e e 2 < U~ Q c < cU < (0 < u U C). e o o u a < e e~ o e < '- e 3, ( < U U U~ < < (0 U U U < C)< (0 U < U U < (0 Q U (O < (0 < <u (O O e o o < a u < (e o < (e < u o _ < < a 3 U U U U U (. U < (0 8 < (0 U 8 < b U < U U Q U (O U < Q U < < < C)U Q < (0 < U U Q U (O U (O ( < < Q ( O (O < U Q (O < (0 < Q U Q U (O < < (0 ( (O e < < U < 5$ ( O ( ( < U U Q U Q U u O O ( < < U o < (0 < (0 ( U U U Q U u O O O O O U CC C l Cle- C T Th . 0 t Cl C l C 55 C) xl C f b cT -- C hC _ fl C > C - > f - T 8 Q t ' C- C U U a U l t <0 CC Q < Ut '0 C lQ 0 C C C C C CC 0 C - Cl Cl ClUCl Cl<Cl < < u o a e o a < o O < CC CC CC CC CC Q < U < H < < U H < -' H < < U < < U~ u < Q < < 0 U < U U < U u U Q < < Q U O O (Q < U U U U < a (o < U u e o u u u UUe < Q < ( ( U( o< < ( (O < U U O ~( ~( M e-C C C CC C CC C CCC C C C C C C C C - fl fl< '0 C- CC C CC CC CC CC CC CC CC CC CC CC CC CC Cl C- - - C- C- - C C- C C- C- C- C- C- C C C C C C- C 2 0 U Q OO Oe D < < 8 56 C u )u 0 =o t~c cr C zz d C 0 C C r f 0 C Cl C~C ~ C t ~ CC Cf2tCr2C u uCu0 u -a aO m 5< uuuuoa C" e u u C C CC o U < U u - U <- N N N -CO C 0 Cl t C C l t C C Cl C C Cl< Cl C Cl Cl C l C l C l C l l C l C l C l H H H H H a a a C cn 0 8 < a 0 a 0 e H~ < u < a 0 20 < 0 0 < 0 0u 0 0 U 0 < 0 m < u i 0 0 0 0 H H 0 0 C- < C<- < a u uon 0 on = e e o a U u C- - o u 2 - C 0 u < 0 0 < < 0 < <8~ < 0 0 < a a < o 0 < <<-<< < 0 0 0 0 0 C- C C C R e e one0eon 0 < < < < < e o- 8 0 0 0 < < < < < < < < 0 o C C.e )z0o o o o o o o o o on on7 0 C C c) 0 = uc 0 t 2 6Cr; 21 ZC e e u X Cc a o n - - C U U T Cl$ $< Ze) ~ C C C c) S U U _ t e C0lm a c a a e e a uCu C< a e e e e u oO mu 02 2 0' ' < ' < H 09 H H K<0 K0d 3 H 2 HO 2 0-S < 0 a o a 0 00<0< u<u 000U < a u0 a 0< < < u a U < < < < 0< uo a o0 a < zz o es < z < < <s -8ze0 0 0 0 $ $ 0 0 3 3 0 0 0 3 3 < < 0 ~n n ~n 0 U ~58 C) ~ 4 (4 ~ '0 ~ ~= - cc ~C) ~n '0 ~i- '0 ~n r- '0 CC ~i- CC t~- CC ~Th _ CC _ ~C) Cl OC OC fl CC ~t ~t Cl~ - ~Th ~fl 5< .0 cC ~C) C) ~Th - ~ ,~- ci cc ,~- '0 Cl ,~ .0 ,~n ,~ ,~n ,~m ,~ Cl CC ~ cC ~ Cl ~t .0 CC ~ Cl ~t .0 CC ~ Cl ~t .0 Cl Cl ~ - Cl Cl Cl Cl Cl ~ H H H H H H H H ~ H ~ H ~ H H _ H H H _ - - - H H H ~ - H H ~ H ~ H ~ H H H ~ H U H H ~ (~ (~ (~ (~ (~ ~ ~ < (~ ~ < - ~ C~ ~ U ~U (~ ~ C)U < (~ < < ~ ~ (~ (~ (~ (~ ~ < < ~ ~ (~ (~ U U U U ~< (~ (~ ~ ~ U < ~ ~ (~ (~ U U U U U c~ (~ ~ ~ (~ (~ U U ~ < < C)~ U U ~ (~ (~ U U U U ~ < < c~ (~ (~ ~ ~ < < c~ ~ ~ U U ~ (~ (~ ~ ~ < < U U ~ < < U U U U U U <U (~ (~ ~ ~ U U U U U U < < (~ (~ ~ ~ U U U U Cl Cl Cl Cl Cl Cl ~ H H H H H H H H H H H H H H H H H U < ~ (~ H ~ ~ H < H ~ ~ < H U H < < H ~ ~ ~ (~ H (~ H -~ H < ~ (~ H (~ ~ U ~ U < < U ~ < U U < < ~ C)~ U ~ U < U U ~ < _ ~ < U ~ < ~ ~ < U U < < U U (~ (~ ~ U < < U CJ (~ ~ < < < -< < < ~ (~ ~ < (~ U ~ U < U -~ ~ < < U U U (~ < < U U < < < < < (~ (~ ~ U U U ~ < U U ~ < ~ 5o~ ~ ~ U U < ~ < < (~ (~ (~ (~ U U U U ~ U U (~ (~ U U < < (~ (~ (~ (~ (~ (~ < < CC Q> - - fi fi ~Th CC CC Q> - - ~Th ~i ~i fi fi fi .0 .0 .0 .0 .0 .0 c- r- r- r- r- r- CC CC CC CC CC CC CC CC CC CC CC CC CC CC CC CC CC CC CC CC CC CC CC CC CC CC CC ~ -~ -~ C Cl ~Th ~- fin - - nfl .0.0 ~i ~i ~i ~i ~i ~i ~i ~i ~i fi fi .0 .0 .0 .0 .0 .0 .0 .0 .0 .0 ~--~ ~Z.0 CC CC CC CC CC CC CC CC CC CC CC CC CC CC CC CC CC CC CC CC CC 59 u )u U = a a -4 - m 0 Ojo oj oj bt b5 U U U U cl C U 1C C zZ r- 5- r0 r- r r- - -~S ~ C60 e om fl C l l e C C N Nu a2 0 0 0 2' 0 c< u H H < <U Su U U e 0 0u u~ CU U a a u u u 0 0 o e cU U U U u U e u U ~ o o~ U U U 0 0 0 0 0 U o o o L< < a 0 a a U U e0 0 U U U ~ ~ U U U U < < o tU U U U ~ U U U < < U 3 Uo a U U o e HY 2H < < u <0 0 < < & <D& <D< < <~ t. t ' -- - m C SN Cr N-- S H H u < a < 0 o < H U a- e U U 0u >0 u U 0 o 2 < 0 0 0 u ~o 0 0 0o a 0 < U o ~ U U o on on on o n0 0 0 n00s< < QJCd a C 0C C 0CCC C cJ CC C C C t C C C cC i CC <C 0 0C <Ooon on < < ~ <U6< C) ~ 4 '0 fl - Cl '0 ~ fl C) Cl fl ~ Z~ ~ Cl '0 ~C) ~C) ~S'~C~Th Q> Cl ~Th fl - - Cl '0 fl - ~QC ~- CC '0 fl _ _ _ ~Th _ CC ~Th CC fl Q> 5< C. '0 CC fl '0 Cl ~t '0 CC ~ Cl ~t '0 CC CC CC CC CC Cl '0 CC ~ Cl ~t '0 CC ~ -Cl H ~ H H H H H H H H H ~ H H H H H ~ E ~ H H H H H H H H ~ < ~ ~ C~ C~ C~ U - < c~ < c~ c~ (.~ < < < < (~ C~ (~ U C~ < C~ ~ < < ~ (~ (~ U U U < -~U c~ < < < C~ ~ (..~ (~ U ~ U ~ U U U U U U U ~ U U < < ~ ~ U ~ U ~ ~)< < U U ~ (.~ (.~ < < < (.~ U ~ ~ ~ U ~U U (~ (~ (..~ (..~ U U U U < (~ ~ ~ (~ (~ ~ U ~ (~ (~ .~ < < ~ ~ U U ~ U < U U ~ < < (~ (~ ~ ~ (~ (~ (~ ~ ~ (~ (~ U U (~ < U U U U c~ (~ (~ ~ ~ - ~ - ~ - ~ - ~ CC CC CC CC CC Q> H H H H H H H H H H H , H , H H H H H H H H H ~ H H U H H < H U H U U < ~' < U ~ U ~ ~ H U < ~ U U < ~ U U U U H ~ H "(s (~ ~ < U U < U (~ (~ U ~ (~ UC~~ < ~ ~ < '~ U U ~ < < U ~ < ~ ~ (~ U U ~< < (~ (~ U < < ~ < U U U ~ U cj ~ U c~ U ~ U < ~ < (~ C U ~ (~ (~ (~ U < < < < U (~ U (~ U U CC '0 '0 CC CC ~ '0 '0 CC CC ~ CC CC Q> - - , , , , , , , , , , , , , , , , , , , ~ CC CC ~ Cl Cl Cl Cl fl fl '0 '0 Cl Cl Q> - Cl ~Th CC CC CC CC CC CC CC Q> Cl Q> CC CC CC CC CC CC CC CC CC CC CC Q> 61 C) O - C]- c 5<o C.o e o o o 7 < Q UU QQ QQ < 0 0 < U C H e u u U a u~ < u < U (u < U U e U O U U U U U Q (U U U U U U~ U (O < < U 8 U U U U U < U (O ( < U < < -e U u U a u~ u U < <~ U u (a < u u < u u 8 U (.U U U < U U (C < U U < < U 8 U U U U U U < < U U U < < cQ O e U U~ U U < < U U < U < << e U U U < < U U C (O O U U o U UO U < < Q U (.. 0 < 0 Q < U < < <~U < -3Q < < Q U (Q U U (O ( < < Q (O U < U U U U E O O < Q U (O U U (O Q < (0 Q U (O U Q U Q < U U Q U (O U U (O Q (O ( Q U U < Q < Q s' H O < (Q U < ( U < Q < e o < (Q U s e e o < (o < (e o < U < o < U (o e e o ( U O < (Q < ( U < U < (g U < < U < < ( eU U~ O O < (Q < U U U < U O Q (8 ( e < < (e u < u . (. a a e~ (e u u o~ < (e a C). ( e ( (O ( U < U (O O (. U U < u <U( ( U O Q~U ~< < < (3 < < <~ (0 0 U < U (.. < (0 ( U CQ U < < 0 SU < (O( < < (0 ( (0 U (O Q ( U U U < Q (Q (O ( S< < (0 (0 ( < < (0 < U Q Q U U U (O U U - , l '0 ] CC > - C] 0 C] o - oj C] - -c - 62 C) ~ 4 C. Cl Cl fl '0 Cl '0 C ~ '0 fl Cl ~Th Cl fl ~ ~C. ~m - Cl - Cl CC Cl _ - CC - Cl Cl C) - _ ~n ~n Q> _ _ CC '0 - Q> '0 5< C. Cl C. ~C) - fl '0 Cl CC ,~t ,~fl ,~ '0 '0 Cl ,~t ,~ Cl Cl ,~Th ,~Th CC ,~ ,~ ,~ '0 '0 ~ ~ '0 CC ~ Cl ~t '0 CC ~ Cl ~t '0 CC ~ Cl ~t '0 CC ~ '0 '0 ~ ~ Cl Cl CC CC CC CC CC Q> ~t ~n ~n ~n - A H H H H H H H H H H H ~ H H H H H H v ~ ~ C~ C~ U U c~ (~ < (~ (~ C~ (~ ~ ~ U U (~ (~ (~ (~ ~ ~ U (~ (~ U (~ < U (~ < < U ~ (~ (~ (~ (~ < < U U (~ U U (~ (~ (~ (~ (~ U ~ ~ fl Cl Q> - ~ fl Cl Q> - ~ fl Cl Q> - ~ fl Cl Q> '0 '0 '0 Cl Cl Cl Cl Cl CC CC CC CC CC Q> ~t ~n ~n H H H H H H H H H ~ H H H H H H H H H H H H H U ~ U H ~ H ~ H ~ ~ H ~ ~ U U U ~ U ~ H (~ (~ H (~ (~ U U ~ U (~ U ~ < < < < U < ~ < ~ U U ~ ~ U U (~ < < < (~ ~ ~ (~ < U U ~ < (~ U ~ < < ~ < (~ -- -- - - - ~ ~ (~ < < (~ (~ < ~ U ~ (~ (~ U ~ (~ U (~ U r~ < ~ U C)~ U ~ (~ (~ (~ (~ U U ~ U - ~t CC CC Cl Cl ~ n fl '0 ~ Cl ~ - ~t fl Cl Cl Cl Cl Cl Cl CC CC Q> Q> ~Th ~Th fl '0 '0 ~ fl Cl Cl CC Cl ~t Cl ~Th '0 '0 Cl fl fl fl fl fl fl '0 '~ Cl Cl - - Cl Cl Cl C.~~Z'0 63 bc oo Cn oox 4 4 U _ '0 - Q < < O ccO '0Q C] UUUOO<< S <0 CC < <] t 0 cc U C Ot ' cc Q C] O 0 cc U C C) D u < u e O OO OO Q O < <U UU e $ < a U U U U Q < < < < Q (* Q Q (* (e (* e < < .0 ( U U O O < < 0( (0 e CO U U U < < 0 ~ < <(~ ( U U U U < < U~ u < u (Q (Q < <~ U U U U U U < < < < 0 (0 (Q ( H O H U U U - < s H H & H (- < x 2 U H Sxu < u u e H < m~ ( e < u o o H a H u u e u e u u U 0 < M U U ( < U 8 U U U < uU s U e OU < 0 U U & < u~ (e 2 - < < U u eu e U U oU oO e U (O cQ e e (O < U < Q u U (O U U U U U ( ( U b u < < u e o u co < < < < e (e (e ( ; U u eu e ( e ( u 3 < < u e U U ( 9 (e O O U U U O U 8 < < oJ~ e e (O ( U u U u U u (O ( U u O (O Q e (O ( f.la - - C] C] -l -l -] -] -C - - - - - - - - - -0 64 u )~ cc e ci c '0 c c 0 i'0 c -0 c e c e 0 c u c ate0 c U U UU UC U < U o o~ e (a o ~UUUU ~( 8 < U U U U U u ~cjc e< < 0 (Q < U ( U O < Q U~( ~~ a e o < < e < U < < < < e ~( 3 U < <~ U~ < < < < < < u~( ~~( <(. U < ~ ( U o U u U u ~( H H U H H H H H H H HHU u U < a < < U u u e - U < U~ Q U U e H e H 9 U H( 'e'' U < (0 < U (O Q , U u U , < o < U < O H U (O y SoU < U U (Q U u O U ( uQ< < U U U < u O U u U0 C)U < Q U U U d U N D U U e o < (. ( ee e U U u e U 0 U (. < (Q U U O ( Q U y O ( 0 < (0 U U Q ( 9< sO (O 2 e < u e. < < u u e a e u < (e e u e < (e e (e U U (O ( < < u Q (Q U 8 0 c U U U < (0 < U 8 Q (O < < < U U < < U << U 3 U u o ~ ( 5( U Q U U < U < <0 (6 < U ( ( e (e u u u u u U (e (e < u '0 t- cc Q> Ci~ T ' ' c c c c c c c c65n Oy~ C)) 35 '0 sl C- Q O= C - - AC C Cl 1 0 CC 10 Cl 10 10 CC 10 Cl 10 10 CC 1 '0 ' 0 ' 'Au' 0 0 ' 0 '0 ' 0 ' o lH ~ H ~ H~ H H H H H H H - H ~ ~ ~ H H LA LA H H J r ZtH Zt HS HS HS - ~ ~ ~ ~ ~ - - - - - ~,< <66 cLc u xu b L L Lc Lx LA) 6l 6~ ol - c ~c - , C ,0 ' , Cl C, , , ,- , c ,n .0,- ,- , -C - , C ,m , , n OOUU U U fl l fl 0 .0 0 '0 < e U U~ U C~ CD CJ e < U U e < Q (Q U < ( 8 (Q U U U U U D U U < < 8 < U U U U 2 2 < u < U U U e U U U U O O c U i < < 0 U < 0 e U U (Q Q (Q (Q (Q e < < U < (0 U U c c O O< < U c c < c c c ( )U (O O U U < <<< U Q ( 6 < < U U U U *U U U (O ( < < < < ( < < < < Q e < U U < U 0 < H < < U U U < < - U U H Q H ~HU ~ ' m < (D <U Q Q < 0u < 9 ~ U 0 < ') e < 0 UU<0 < 0 U U < U 0 o 0 < Q 0 0 < < 0 0 0 o < 0 U = U < 0 u 0 0 U 0< 0 U U 0 0 0 0 U U < <U0 0 0 0 0 U < 0 0< u U o < 0 e 0 o e a U y 27 U U U 0 < < o 0 0 U < 0 U 0 o U O OU0 o U < 0 0 0 < < u U U < U 0 0 SUU U U U 0 0 0< 0 0 0 0 U U < 0 0 O '0 '0a C CCe CC CC e < - '0 - - u Cl < h f UlO l '0 '0 U CU < Cl Qt et 67 C)u Go0 bel '0 C C > fl C l 0 C ol '0 ' t 0 l C 2 Cl C l - C T C Q l ' Cl C Cl eT 2T 2T 3T - l u Cu e 2eu ) a 5< <<<0 c a ' 0 0 0 '0 '0 '0 '0 '0 '0 '0 ' 0 '0C Cl Cl Cl Cl Cl C C t C< H H H< 39 <s H H C~ ( C < ee 2 e e - < < < a u u u U ~ UUrU U U U -S< (0 e- u U U U<e < c0 o ( 00 - U U U U u( < U e a U e U U u o U D U U u U cj U cj U U U < <~ < < U U U 2 _ U U (O U U < < l Cl Cl C CC CC C CC Q> <- a < Q < H o H s se H H H H m < H H ~Q < H _ H OU H 2( oooo o. o~ o~ o~ o o~ o o JC d2 e N N N N N N N ~ ~ ~ c68 c)u 0 = Cai o o C o o c~u C o ao- oO um 5<8 C C C l t C CC C Cl t C p C p Cl< 0 e < < < 9 0 2 0 H H H H 0 a 0 - 0 - 0 H H e e <<oa 0 a 2- 000 euuu ~ 0 & < - 0 0 0 u 0 on 0 0 0 0 e S < 0 0 a a o 0 on 0 a - 0 0e S0 - E onde - on e 0 0 0 e 0 a 8 e 0 <0 onu onouon 20 u a0 CO 0 0 0 _ < < - - 00 a 4 < a e - zz < zz < 0 e0 0 < < 0 x < < 0 0 cU 0 e u U < < S< 0 0 m 0 0 0 0 0g 0 0 0 0 0 0 S o 0 e e a a e a e a 0- e 0 0o o < < < on <eo< < < < < :? 0 HY<O 00 A CdeN N N N N N N N N N N N N N N N N N N N N 69 x C] m oC ml Ml C C C n - ~i n n - n 7i- n] 7l nT 7Th n ] 7 i T 0 C] ] 0 1C 0 C] U I0 Uc U 0t 10 U uu uu c CC CC CC CC CC oo . . . H H H H H H H H H H H H H H H L H H c70 u .. u u u oU < < u U (u( u ~ ( u u o 6 8 U (C < U < < <U U < ( U < ( < U < U < <~ U U < U < Q Q < 0 U ~ u ( a < e <~ u < < U < a U 0 U U U e ~ U (~~ ~ U < U U < Q U < < U 2 < C)C. U U~ ~ ~ U U U < U < (. U Q < Q U U (~(~ ~ ~ < U < U < U < ~ O Q U (Q U < ~u (Q (9 ( O U U U < U < < < U~ < U U (Q (. < (0 U(~ u ( Q < U < < U < < < U~ < < U O (Q U U < < U <~ u o o < e < u <~ <~ u e a (a 8 U U U < (0 U U < < U < <~ U (O U e e U u u (.u < u u U < U < u < u (~U ( < (~< < U U c~~70( z a e eC - '0 C Q C) Q - '0Q UC '0 < - - Q <l UC U < < - 0U U U UC U C t <0 C Q Cl < 0 UC < Cl t '0 C < UC$ 0 U~ < < C0 0 U U < < 0 < U < U O U U U U < U~ U < O U < < U < (0 -3 < U Q (Q U (Q (. U < < < <j U < < < E- O Q (Q U U < U Q U U~ U U < < U < < (0 0 < ( U U U U (Q < < 0 < < U < < (0 (0 < -e( u e ( < U u < u e < < u e u u mU o < U U < Q U < U U U < U Q Q s' c O u Q < (U U U L jQ U U < <. U. U Q (O U US U (e U U U < < Lj cQ U < < ( U U < U~ < < U (e < U U < <~ (U U U < < (2 ( U U (C < U U u U U < (. < U H ~ ~o < U U <U U H0 < c ~ U < 4 < U < ~U < U ( < U ~ 71 u )~ Clo~' l ' > ' l~~f 0~ ul~ h C > C t~ h - - - -- ~ h C T c ~C e o o o c. o e ' 0 '0QQ <C u Cl < U0 < 0 C ( U (2 CJ Q CQ < (O U U U < < Q < < (..0 < 8 < < < < (Q U U < (0 U U U < < < U U < e OU (O U < (Q U U < (0 U U U < U U < <~ U U e < U < < (0 U U < (..U < U < (.U U~ U < U U < U u < U U < 0 U U < < < U < U *N U U < <~ U U (.. U U < U < 0 < U U< U~ (O < ( U U U U~ U (Q U < U < U < U U U U~ U U U~ < U U < < < < U < U (O < (0 U U U (O U Q U (O < (Q (Q Q Q Q ( U Q U (O ( (O U Q < (0 U -< (0 ( U (O U Q O (O U Q (O ( Q (O U Q U e ( O ( U (O ( U Q O (Q U U (O ( Q U U < Q U s'< (Q Q Q U (O < (0 U Q O U Q (O ( Q (Q U S-U U U (O < U (O < (0 U Q (O ( U U (O ( Q U < (0 e U U (Q < (Q U U < < (U UQ < Q 2- ( U U U < (0 2 U ( < < U U < Q (Q U< (U U U < < U < U~ (O~ < ( (O U < < ( U ( my U < < U < < U < U. ( (Q < U o O U U U < < U < U < (2( U <0 (Q U U U < < (.. < (C Q Q < U < < < U < (C Q U < C (C U U < Q C)2 U U U U Q U < < < U < (. Q (. < Q U < -u < < < a a o < < < u e. e.. o e. u a o o < C)Q U < U Q < Q Q U < < < < (0 Q U (O U O U U u u (e a a u < a a u < < u < (e o < (e a a u -fl C- 2T C- 9t ; - Cl 4T ~t f0 C4 > - Cl l '0 C oc C- ' ' Th~t ~ t ~ ~ ~ ~ l l -- C- -- 72C 00 00 -0 xf l Cl-oT l -l ol -l l - -l =N =N =N = z y r- o, m w - o 0r- N : x =N = = = = 00 = N = N N = N = 0 0 , 0 ~C) --. tb 5<b . . .. . . . . L CC ~ - l > - - C ~h l t t f ' ' ~ 73C c) 0 = SC l l o o- =N; =N = N =CN = = l l l - l ol -l Oo -n.- 7 x C zUz OO e - uOom 0 D G ,*t C. *t C- *- * u 2 0 p H H H H a H H H H on H H on < 22 H H H < 0 0 0 0 0 0 C- a t t C. 2o o e oU Q, ,< <- -Q<Q C-= a U U U4 o e 0 0 0 a 2 0 0 '- < U '- Co w < < < u ' 0 0 < 0 0 e < w C< ,< 0< , < 0 < < < e s".o 0 o c)0 C- 0 00 5< 50 ~- 0 C.- < C. < U UU0U Q SU U U < u- < C- o u < u < 0 < < < 0 a ee0 0 a et a a < <t < 0 e 0 < o < 0 0 o). 0 U UM U LO C.- \ OOH - O UUs QJ>oe N - Nr N/ N SN - Nr N/ N SN - Nr N N SN - Nr N 74 C) ~ 4 CC fl ~ - Cl Q> Q> fl Q> CC ~ - Cl ~Th ~Th - fl fl Cl ~ ~Cl ~C) ~'' '0 CC C- ~Th _ _ ~Th C- '0 fl _ C-- ~t 5< C. - C. ~C) ~~C) ~ C-- Cl fl fl ~Th '0 C-- fl '0 C-- Q> ~ ~ ~ ~ Cl ~t '0 CC CC ~ Cl ~t '0 CC ~ Cl ~t '0 CC ~ Cl ~t '0 CC ~ ~- ~- CC CC CC CC CC Q> H H H H H H -H H H H H ~ H H H H H H H H H H H H H H ~ H H ~U (~ < < U U (~ (~ (~ (~ U (~ (~ (~ < (~ U < ~ U U - ~ ~n ~ '0 '~ C-- 1- 1- ~- ~- CC CC CC CC CC Q> H H H H H H H H H H H H H H H H H H H H ~ H H H H H H H H H H U ~ U U < U U U ~ U ~ U U ~ (~ U ~ ~ (~ H U H ~ H (~ U U U ~ U (~ ~ < ~ H ~ E < E < ~ < ~ ~ H ~ < ~ < U (~ ~ U ~ ~ < ~ ~ ~ (~ U ~ U ~ < 5 < (~ (~ U U ~ ~ U ~ < ~ 5< (~ (~ U (~ < U < U < (~ < c~ cj ~ < c~ ~ ~ < (~ (~ (~ U C)(.~ < ~ U < U < U < U ~ ~ U ~ (~ (~ < < (~ ~ (~ ~ < < U U U < U ~ U ~ ~ < < U ~ ~ (~ (~ U ~ (~ U ~ (..~ (~ U U ~ U ~ < ~ ~ U ~ ~ (~ (~ U ~ U U ~ < U U U ~ 5(~ < ~ ~ U 5 (~ (~ U ~ U U (~ (~ < < < Cl ~ fl - - Cl Cl ~Th ~Th fl ~ - Cl ~t '0 Q> fl fl Cl Cl Cl Cl ~t T ~t Q> C-- Cl Cl Cl Cl Cl Cl Cl Cl Cl Cl Cl Cl Cl Cl Cl Cl Cl Cl Cl ~Th CC CC Cl Cl C-- C-- C-- C-- C-- CC CC CC CC CC C-- fl fl '0 C-- Cl ~Th '0 CC - C-- C- CC CC CC Q> C.~~Z'0 Cl Cl Cl Cl Cl Cl Cl Cl Cl Cl Cl Cl Cl Cl Cl Cl Cl Cl Cl Cl Cl 75 .. c - oco o-occ 5< 33 -z 9 < U < '0 c Q C t U0 <C < <l <t 0 CC Q Cl < 0 CC Q 8 < U~ U (O < (Q < U Q (O U U ( Q < (Q ( U~ U 8- U < < U U (Q < < 0 0 (0 < 0 U U U 8 ( O U O Q < U < U Q U U O O U U e UQ (Q ( < Q U Q < 0 (0 ( < < Q Q cU U U< E < U U cu cu ( (O Q (O ( (O e O U U < Q u U (C (D ( < Q U (Q < Q Q U U (C e < (Q U < Q -U Q (O U Q U < U < Q U (O ( Q e (.. (O Q O E O U (O U U < (0 Q O (O ( U Q O < < -z-- - - - - H U H H H H H H H H H H H H H H H H H S< U U < 0 U O (Q U U u U U (Q (Q U < U< U < < (e U < (e a u u < U o e < u < u cUU < < < <~ U U U U U u < (Q (. < U o < U < (Q < < <~~ U o U o < U < U U < < U U < U U O (< Ca C < < u u < u~ e < u u ~ o cj( < < U 3, ( D < < U (Q U U U U < C U U ( < (0 0 e U U (O U U < U Q Q Q < (0 ( 3 < < (0 ( < < Q U U < U U U (Q ( U < M < (0 U U U < Q (Q U U Q G C G (G G u a e (e U a U o (O < u (e o < u a u u e SU U Q < (0 U < U Q U Q < (0 < U U U U U (Q (O ( (O Q < U (O < (0 ( (0 U < U U (O < U U U 5$ (O Q U (O < (0 (0 (0 Q U U < U Q < U U ~< u U e (~u < (e o < u u U (e ( < u u o < u '0e - o - - r- -n -0 - 76 C) o ol~ ' > C l Q C C l C l f C C l f bc- C T - ~ h f C C Cl fl ' ' 0 0 ' C l l C C CC C C C C -'O U~ U U Q < Q Q ( Q ( e <~ U < 8 U U U U ~ ( ,a e U U U o U U < Q U U U < Q U O (. (Q < 0 Q e Q U (Q U < Q U a e < e u a u u < e a < (e o a e u e e ae o e < (0 U O < <<< < <<< ( UOQ U U U U < <a < < < a E QO UOQ <0 e O < < < a U < 00 0Q <U <<H < H U H < U e <u < u < < uo m U u < <u e~ e C- C- 00 ,'Uueu<< H H C-H<( >< U U U <~ < U H HU H U U < cQ < < c~ < (< C U UU e < < U ~ U U < < < K U~ O U < . < U U U< < u < < (e u U u < u- - C C e 5c e < u e U < < o~ e U U U < a a< e U < U o < u < <~ C- < U -e a b (00 U cO U < U < < U U U (U < U < < U U (.O < U < U = 4-4C < e < e e < < u.. < < u u < u uC- Ce u u C- C C) e o < (e e < < (e e < U e e et< < e et< r(.u < u~( < < < < < < < <,< < < <a<U (a ( S l T et f '0 Q> o -C N N N N Nl Cl Ni Ni Ni Ni N N N N N N N 77 beb - ~~~~~7 't Cl SJ~ l t l - CC~h~C i Q mm m mm mm - - - 'o - 10 r- r . . . 278 o < ~C) H < H Q a O (O (O (O (O CO U~~~C < Q U U < -3 2 Q U U (O ( U U U < Q Q U Q U U U e N U < Q Q (O U < U < Q < . ( H( ( O (O ( < U U U < Q Q < U H U c e < U (e (e < o<o o o e~( ( e o < < U < U ( < ( U U U < ~U ( c U - C- U o < a o o ~ U U < U ~ U U - U e U U U U < U Q U U U <~ C) a o %a U U U U U < < Q < < < U~ z C- Q U Q ( O U Q U U U < Q U < U e- - - - - c - Cl o - Cloh~ l ' C > C 0 C 78 u )~ cc - ' co ~ c c ~ Q l C o = - - - r l C _ C i Q i C ~co H H a o o~ u Ua < u u u o < < u eu - U < Q U~ U U < 2 (2 U < U U( ( ( ( ( Q (Q < U < Q (.2 U < 0 0 8 U U O (U U < < Q U U e U a < u~ u < < < < U < U U U S U Q U <~ < < < 0 < Q < U SU U U <U Q < O < Q < o U< U < < Q < < U U < U < U Q U O U Q <~ ( (Q U U U < <~ U~ U (O ( < U < <~ ( 0< U < U U < 0 U (Q (~ U U U U Q U U Q O < U U~( U < U 30 H<< H C)< < < < (0 ( U e u u u e e u U( O U C (O Q < < U O U < U Q < C) ( Q < U U cU < U U (O ( U (O U U < a O O U < U (O O U U Q (O U (O U U u < (0 e u o u < u (. < (e < u 3 0 0 < < u~ < a p u (e < a < < u u a u U < U (O ( U~ Q Q U Q U U Q < U U UU U b~ (e (e ( u a (e u a < u o < u a e u < < Q (O < < (0 < U Q U U Q U Q U ( eQ 6 < < (0 U U (O < < < 0 (0 Q U Q < U 79 c) U = c <U a Co o O-~ -- m2 maa> c a> a> a> a a a> a> Cf tr Cr tf tr t/ t/mu CC 0 Cl t C CC 0 Cl t ,C CC 0 C t C Cl t C N CC C CC CC C a a> a a a> 0 0 H H <a m j j s H H H 0 < < < < H< <D 0 e <U zz<<o U e ooo -. e < < o- < o o on on o 0 o o on on on o o o C< < ~ < << ~ ~ ~~'80 '0 Z Z 0 O C Cl c Cc c e %Nol C Cl Cl(N on. - o m 5< C CC) <00 -< <C Cl C C C C Cu CuCl Cl C l Cl Cl C e C C C <, CC eN ,C l Cr CC SN , Cl Cr ,O C 0 C < u < C oC C <l t C CC C Cl t C C C C t C C C Cl4 el <l C l C; C; C; C; C t/m tf t/ t/ </ - H H < H < 0 < - < < H <r; 0 0 0 < < C- < C- < H H H i <si H H H < < <0 <0 <2- C- 2 ~< - 6 0 0 8 6 0 0 6 0 z e < e < 0 - a < < on 0 s < < co 0 0 0 0 0 < 0 0 U m S < < 0 S 2 0 0 0 D 0 0 0 tO 0 < 0 0 < < < < S S S 0 O 0 e 0 0 0 c0 < < 0 < 0 2 C. 0 0 0 C. C.i 0 0 < S 0 < < 0 < one a < 0 < < 2 < m cJ 0 0 g 0 0 5 2 5 < 8 < a zz < zz < u- < < < u U S S U < < < o < Z aZO S 0 0 < < b . 0 < 0 < 0 < 0 < 2 < < < a o <0 < u < < & < a < <e < & <H <H< < < < m HoU H<< e -oE D DoE < H s H HHEHC-S'C S8 S 555 a~zae SO81 C - S NC C N SN N 0 C - S N C <Uu u on-- - C2 2 0 C2l ~u C s -O m 25<9a 0u 2C CC82 C -c c~) = Ct C) F -~ .~ Vt C) ~ .fl C C) -c _ _ CC CC) CC *t CC) C) C) 0= nC tO cr _ CC) C) C C .rc CC ZC x~- CC) 0 on.- zon C zz C ZZC) ~ C -C m 5< 0 C 0 CC) 0= ~ AC) 6~t;A~ ~ < - c.-2 -c ~ C) C) Z -c ~ ______ _ CCC fl C) C ~ CC)' C ~ -n H H 0 C) ~ ~C)C) c-~ on~Z5 y~ CO $- c~) ~ o C) < 0 CO C Ct C C)C)~ -c .~ 7 -~ -z -~ 0 C -~tC 0 H H U H~ C) >0 *~~z -c ~ 0 "0 ~ 0 C C)~ C ~ U C Ctr C< ~- < < 0 t~-~c < < 0 -'U 5< (fl < CC)~ C) C -~ 0,C C ~ '0 C 2 < ~ N- cr CC ~ UC)5 CCC) '0 0z -zZc cn ~ C 83 u r_ Cl ~ 0 C t 0 Z Cl~ ~00 Z l t~ 0 C U -:: cu u u f~H H uu HHHHHHHH cUH U ~H~HHHHHHH= U= U=HH~ U U U U U U U uuuu u U U U U~ U U U U U U U U U U U U U U U uuuuu V5 0 .0 10 0 0 . Cl c. t 0 0 . Cl C. t-0 0 00 OF F N Z 00 C, Cl Cl NlC lC lC l C Cl NlC lC l lC l 84 U. COOu C uO 0u 4u. 4u CO u 4.l u 00 Z Cl 00 Z Cl~t 00 Z C ~t 00 Z C ~t~ 00 Z C ~t~ 0 V~ ~ ~ r~- r~- -~- -~- r-~- 00 c 00 00 0 O if) u u < CZO CO CO cOz CZ-'-'Z cz l~t 4c t t H H H H COIzczc CO if)CO CO C O CO W) CO< C Z WZ C ,W - C C if) ~ ~ ~ ~ ~ ~ ~ ~ ~ C CO CO CO c - - r - r 0 0 0 0 0 ~~r CO COCmZ C if) C0 0 0 0 0 ~ 0O lc - 0O C tV Cl ~ ~ ~ ~ t Cl: Cl: Cl: ClC l C lC lClC lC lC l Cl C lC lC l C 85 z z z u u ~* u o4 Z u 4u. -4 00 Z Cl- 0 l, 0 l,5~0 l t ~ 0 Cl C Cl Cl C c'c'~ '~c~ c ~ ~t~t ~ ~t V~V~ V~V 1( 41 .b41 H 4. 4. H U u U U u U U 4. UH uuuHu u u u U ,,U u cz cz c<<c c <~ CZ tkl cb cz cc 0 0 0 0 0 cz cz CZ 4. z U1. U U U co 00 00 00u 0 00 00 0 ulu U.UC9()C9ut 000000 oHHHHHHHH OOOOOOOZZZEEEEEEEHHHHHHHH uu~m) m~uu uuu uu u u rn W) C- C, rn If EC n I)C n W - C n W -C ~4-, 0 0 0 0 0 Cif) 0 u -0 rn 0f 0- 00 0N 0) N r )r 00 CC 0000 00 00 or r n n r r n n rn r n 0 0 0 0 0 00nrn r r n n r r n n r - - - - - - U U6 r- - - -- -00 00 t- - - -C 4. 4. 6. 4. ' z z rn ~ ~ ~~ if) r- C f) r , n C QD~ QD~ QD~ QD~ QD~ 000000 QD Q 4.Q Q CCCC 04 4. 4. 4. 4. 4. 4 t H < << << < 4 : . 4 . 4 . 4 tr C cu 0 4. 4. u 0 . 0 u )C u u u. 4. 4. u uH rn f) c - 0 C C N r ' tr ID r 00 CNC) r Ho 00 H00 0 0 - - - - - - - - - - - - - - - -0 H ~ H EC 87< If~ -~ I ~ -~ ~.) I Ln ~) -~ I 88

Claims (15)

1. A double-stranded ribonucleic acid (dsRNA) for inhibiting the expression of a human PCSK9 gene in a cell, wherein said dsRNA comprises at least two sequences that are complementary to each other and wherein a sense strand comprises a first sequence according to SEQ ID NO:453 and an antisense strand comprises a second sequence according to SEQ ID NO:454 and wherein said dsRNA, upon contact with a cell expressing said PCSK9, inhibits expression of said PCSK9 gene by at least 40%.
2. A pharmaceutical composition for inhibiting the expression of the PCSK9 gene in an organism, comprising a dsRNA and a pharmaceutically acceptable carrier, wherein the dsRNA is as defined in claim 1.
3. A method for inhibiting the expression of the PCSK9 gene in a cell, the method comprising: (a) introducing into the cell a double-stranded ribonucleic acid (dsRNA), wherein the dsRNA is as defined in claim 1; and (b) maintaining the cell produced in step (a) for a time sufficient to obtain degradation of the mRNA transcript of the PCSK9 gene, thereby inhibiting expression of the PCSK9 gene in the cell.
4. A dsRNA for treating, preventing or managing pathological processes which can be mediated by down regulating PCSK9 gene expression, wherein the dsRNA is as defined in claim 1.
5. A vector for inhibiting the expression of the PCSK9 gene in a cell, said vector comprising a regulatory sequence operably linked to a nucleotide sequence that encodes at least one strand of a dsRNA, wherein the dsRNA is as defined in claim 1.
6. An isolated cell comprising the dsRNA is as defined in claim 1 or the vector of claim 5.
7. A double-stranded ribonucleic acid (dsRNA) for reducing the expression level of a human PCSK9 gene in a cell, wherein the dsRNA is as defined in claim 1. 89
8. The dsRNA of claim 7, wherein said dsRNA, upon contact with a cell expressing said PCSK9 reduces the expression level of said PCSK9 gene.
9. The dsRNA of claim 8, wherein said contact is performed in vitro at 30 nM or less.
10. A pharmaceutical composition for reducing the expression level of the PCSK9 gene in an organism, comprising the dsRNA as defined in claim 1 and a pharmaceutically acceptable carrier.
11. A method of treating a PCSK9 associated disorder comprising administering to a patient in need of such treatment, a therapeutically effective amount of a dsRNA of any one of claims 1 or 7 to 9.
12. The method of claim 11, wherein said PCSK9 associated disorder is hyperlipidemia.
13. Use of the dsRNA of any one of claim 1 or 7 to 9 in the manufacture of a medicament for treating a PCSK9 associated disorder.
14. The use of claim 13, wherein said PCSK9 associated disorder is hyperlipidemia.
15. The dsRNA of any one of claim 1 or 7 to 9, or the pharmaceutical composition of claim 2 or claim 10, or the method of for inhibiting the expression of the PCSK9 gene in a cell of claim 3, or the vector of claim 5, or the isolated cell of claim 6, or the method of treating a PCSK9 associated disorder of claim 11 or claim 12, or the use of the dsRNA of claim 13 or claim 14, substantially as described herein with reference to the examples and/or figures. 90
AU2012261570A 2006-05-11 2012-12-07 Compositions and methods for inhibiting expression of the PCSK9 gene Ceased AU2012261570B2 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
AU2012261570A AU2012261570B2 (en) 2006-05-11 2012-12-07 Compositions and methods for inhibiting expression of the PCSK9 gene
AU2016203687A AU2016203687A1 (en) 2006-05-11 2016-06-02 Compositions and methods for inhibiting expression of the pcsk9 gene

Applications Claiming Priority (7)

Application Number Priority Date Filing Date Title
US60/799,458 2006-05-11
US60/817,203 2006-06-27
US60/840,089 2006-08-25
US60/829,914 2006-10-18
US60/901,134 2007-02-13
AU2010241357A AU2010241357B2 (en) 2006-05-11 2010-11-09 Compositions and methods for inhibiting expression of the PCSK9 gene
AU2012261570A AU2012261570B2 (en) 2006-05-11 2012-12-07 Compositions and methods for inhibiting expression of the PCSK9 gene

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
AU2010241357A Division AU2010241357B2 (en) 2006-05-11 2010-11-09 Compositions and methods for inhibiting expression of the PCSK9 gene

Related Child Applications (1)

Application Number Title Priority Date Filing Date
AU2016203687A Division AU2016203687A1 (en) 2006-05-11 2016-06-02 Compositions and methods for inhibiting expression of the pcsk9 gene

Publications (2)

Publication Number Publication Date
AU2012261570A1 AU2012261570A1 (en) 2013-01-10
AU2012261570B2 true AU2012261570B2 (en) 2016-05-19

Family

ID=47520519

Family Applications (1)

Application Number Title Priority Date Filing Date
AU2012261570A Ceased AU2012261570B2 (en) 2006-05-11 2012-12-07 Compositions and methods for inhibiting expression of the PCSK9 gene

Country Status (1)

Country Link
AU (1) AU2012261570B2 (en)

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2005014782A2 (en) * 2003-06-13 2005-02-17 Alnylam Europe Ag., Double-stranded ribonucleic acid with increased effectiveness in an organism

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2005014782A2 (en) * 2003-06-13 2005-02-17 Alnylam Europe Ag., Double-stranded ribonucleic acid with increased effectiveness in an organism

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
BENJANNET, S. et al., The Journal of Biological Chemistry, 2004, vol. 47, pages 48865-48875 *

Also Published As

Publication number Publication date
AU2012261570A1 (en) 2013-01-10

Similar Documents

Publication Publication Date Title
US20240093199A1 (en) Compositions and methods for inhibiting expression of the pcsk9 gene
AU2007299629C1 (en) Compositions and methods for inhibiting expression of the HAMP gene
EP2245039A2 (en) Optimized methods for delivery of dsrna targeting the pcsk9 gene
AU2012261570B2 (en) Compositions and methods for inhibiting expression of the PCSK9 gene
AU2016203687A1 (en) Compositions and methods for inhibiting expression of the pcsk9 gene

Legal Events

Date Code Title Description
FGA Letters patent sealed or granted (standard patent)
MK14 Patent ceased section 143(a) (annual fees not paid) or expired