WO2007134161A2 - 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
WO2007134161A2
WO2007134161A2 PCT/US2007/068655 US2007068655W WO2007134161A2 WO 2007134161 A2 WO2007134161 A2 WO 2007134161A2 US 2007068655 W US2007068655 W US 2007068655W WO 2007134161 A2 WO2007134161 A2 WO 2007134161A2
Authority
WO
WIPO (PCT)
Prior art keywords
dsrna
sequence
pcsk9
gene
expression
Prior art date
Application number
PCT/US2007/068655
Other languages
French (fr)
Other versions
WO2007134161A8 (en
WO2007134161A3 (en
Inventor
Pamela Tan
Birgit Bramlage
Maria Frank-Kamenetsky
Kevin Fitzgerald
Akin Akinc
Victor E. Kotelianski
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 to PL19168142T priority Critical patent/PL3578656T3/en
Priority to AU2007249329A priority patent/AU2007249329C1/en
Priority to EA200870528A priority patent/EA015676B1/en
Priority to KR1020107013424A priority patent/KR101320916B1/en
Priority to CA2651839A priority patent/CA2651839C/en
Priority to EP17162101.4A priority patent/EP3249052B1/en
Priority to CN2007800248541A priority patent/CN101484588B/en
Application filed by Alnylam Pharmaceuticals, Inc. filed Critical Alnylam Pharmaceuticals, Inc.
Priority to EP21150171.3A priority patent/EP3872179A1/en
Priority to NZ572666A priority patent/NZ572666A/en
Priority to EP19168142.8A priority patent/EP3578656B1/en
Priority to EP07762085A priority patent/EP2021507A4/en
Priority to JP2009510173A priority patent/JP5570806B2/en
Publication of WO2007134161A2 publication Critical patent/WO2007134161A2/en
Publication of WO2007134161A3 publication Critical patent/WO2007134161A3/en
Priority to IL195181A priority patent/IL195181A/en
Publication of WO2007134161A8 publication Critical patent/WO2007134161A8/en
Priority to AU2010241357A priority patent/AU2010241357B2/en
Priority to IL220102A priority patent/IL220102A/en
Priority to IL250678A priority patent/IL250678A0/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • C12N15/1137Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing against enzymes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7088Compounds having three or more nucleosides or nucleotides
    • A61K31/713Double-stranded nucleic acids or oligonucleotides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/06Antihyperlipidemics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/10Type of nucleic acid
    • C12N2310/11Antisense
    • C12N2310/111Antisense spanning the whole gene, or a large part of it
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/10Type of nucleic acid
    • C12N2310/14Type of nucleic acid interfering N.A.
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/31Chemical structure of the backbone
    • C12N2310/315Phosphorothioates
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/32Chemical structure of the sugar
    • C12N2310/3212'-O-R Modification
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/32Chemical structure of the sugar
    • C12N2310/3222'-R Modification
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/33Chemical structure of the base
    • C12N2310/332Abasic residue
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/35Nature of the modification
    • C12N2310/351Conjugate
    • C12N2310/3515Lipophilic moiety, e.g. cholesterol

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

COMPOSITIONS AND METBOBS FOR IMfIEITING EXPRESSION
OF THE PCSK9 GENE
£g>g^BefegIPe 1^o ^ebted AppIIcitfoiis
This application claims priority to U.S. Provisional Application No. 60/709,458, filed May I I , 2006; U.S. Provisional Application No. 60/8] 7,203, filed June 27, 2006; U.S. Provisions! Application No. 60/840,089, filed August 25, 2006; U.S. Provisional Application No, 60/829,914, filed October ! 8, 2006; and U.S. 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.
This invention relates ι.o double- stranded ribonucleic acid (dsRNA). and its use m mediating R.NA interference to inhibit the expression of the PCSKV gene and the use of the dsRNA to treat pathological processes which can "be mediated by down regulating PCSK9, such as hyperlipidemia.
Background of She iiiveisikm
Proproieiπ coπvertase sublilisi.0 kexhi 9 (PCSK9) is a member of the subtslisiπ serine protease faπniy, The other sight mammaϋan sυbuiisin proteases, PCSK i -PCSKS (also called PCl/3, PC2, ibria, PC4, PC5/6. PACE4, PC7, and S1 P/SKM ) are proproteirj converiases ihat process a wide variety of proteins m the secretory pathway and play roles in diverse biological processes (Bergeron, F. (2000) ,/. MoL EmiochnoL 24, 1-22, GensKerg, K., ( Ϊ99B i Sendn, CeIi Dev. Bioi % 1 1-ϊ 7, Seidah, N. G. ( 1999) Brain Res. 848, 45-62, Taylor, N. A., (2003) FASEBJ, 17, 1215-1227, and Zhou, A., (1999) J. BhL Chem, 274, 20745-20748). FCSK9 has been proposed io play a rale in cholesterol metabolism. PC1SK 9 mRNA expression is down-regulated by dietary cholesterol feeding m mice (Maxwell, K. N., {2003} / Lund Res. 44, 2109-21 19), up- regulated by statins in HepG2 cells (Dubuc, G.« (2004) Arteήoscier. Thromb. Vase. Biol. 24, 1454- 1459), and up-regulated iu sterol regulator/ element binding protein (SREBP) transgenic mice (Morton, J. D., (2003) Proc. Natl Acad, Sd. USA 190, 12027- 12032), similar to the cholesterol biosyntheiic enzymes and the low-density lipoprotein receptor (LDLR). Furthermore, PCSR9 raissense mutations have been found to be associated with a form of autosomal dominant h>perchcsiesterolemia (Hchoiaj) (Abi&del, M, ei uL (2003) NaL Genei. 34, 154-LS6, Timms, K. M, (2(104) Hum. Genet 114, 349-353, Lerεn, T. P. (2004) C/iw. GeneL 65, 419-4221, PCSK9 may also play a role in determining LDL cholesterol levels in the genera! population, because sirjgle-nuclaoiids polymorphisms (SNPs) have been associated with cholesterol levels in a Japanese population {Shioji, &., 12004) J. Hum. Genei. 4% 10*5- 1 14).
Autosomal dominant, hypercholesterolemias (ADMs) are monogenic diseases in which patients exhibit elsvaled total and LDL cholesterol levels, tendon xanthomas, and premature atherosclerosis {Rader, D. ]., (2003) ./: CHn. Invest, Hl, 1795-I803K The pathogenesis of AD Ms and a recessive form, autosomal recessive hypercholesterolemia (ARH) (Cohen, J. C, (2003) Curr. Opin. Lipkioi. .14, 121-127), is due to defects in LDL uptake by the Hver, ΛDIf may be caused by LDLR mutations, which prevent .LDL uptake, or by mutations in the protein on U)L, apoiipoprotdn 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 claihπn, Therefore, if PCSK9 mutations are causative in Hchoϊa3 families, n seems ii.kt?!y that PCSK9 plays a role in receptor-mediated LDL uptake.
Overexpression studies point to a role for PCSKS in controiihig LDLR levels ami hence,
LDL aptfike by the liver {Maxwell K, N. (2004) /Vm\ Nail. Acad. Sa. USA 101, 7 U)O- 7105, Serφmnet, S,, & a!. (2004) J. BwL Chew. 21% 4S865-48S75, Park, S. W., 0004} ./ BioL Chem. 279, 50630-5063Sji. Adenovirai-mediated overexpres&ion of αiouae or hunum PCSK9 for 3 or 4 days m mice results \n elevaieα $oια! and LDL choϊe&terϋi levels; this effect is not seen in LDLR knockout animals (Maxwell K. R (2004) Proc. Nail. Acad, ScL USA 161, 71 Q0--71O5,
Benjannet, S., et a!, (2004) ,/ Biol. Chem. 279, 48865-48875, Park, S. W., (2004) ./ Biol. Chem. 279, 50630-50638}- fa addition, PCSK9 overexpression results m a severe reciuciion in hepatic LDLR. protein, without affecting LDLR raRNA levels, SR.EBP protein levels, or SREBP protein nuclear to cytoplasmic ratio. These results indicate that PCSKv, cither direcϊiy or indirectly, reduces LDLR protein levels by a posttranseπpuonai mechanism
!..oss of function mutations in PCSK.9 have been designed in mouse models (Rashki eta!., (2005) PM4S, 102, 5374-S379., and identified in human individuals Cohen εt a!., (2005), Nature Genetics,, 37, 161 -165, In both cases loss of PCSK.9 function lead to lowering of tola! 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 \o an increased risk-benefit protection from developing cardiovascular heart disease (Cohen et.al., 2006 N. Engl. J. Med, 354, 1264-1272.). Clearly the evidence to date indicates that lowering of PCSK9 levels will lower LDLc.
Recently, double-stranded RMA molecules {dsRNA) !u;ve beers sliowrs to block gene expression in a highly conserved regulatory mechanism known as RNA interference (RNAi), WO 99/32619 {Firs et al.) discloses the use of a dsRNA cf at least 25 nucleotides in length Co iαhibit the expression of genes in C elegαns. dxRNA has also been shown to degrade target RNA in other organisms, including plams (see, e.g., WO 99/53050, Waterhouse et a].; and WO 99/61631 , Heil«tz et aJ.), DrøiOj?ή/7β {see, e.g., Yang, D., et a!., Curr. BioL (2000) 10: 1 191- 120O)5 and mammals (see WO 00/44895, Liππner; 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 phiirmaceutϊcαi agents tor treating disorders that are caused by the aberrant or unwanted 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 agems that can inhibit PCSK9 gene expression and that can treat diseases associated with PCSK9 gene expression such as Iiypcdipidemia,
The irjveπtion provides a solution to the problem of treating diseases that can be modulated by down regulating the proprotem converl&se subtilisin kexiπ 9 (FCSK9) by using double-stranded ribonucleic acid (dsRNA) to silence PCSK9 expression. The invention provides double-stranded ribonucleic acid (dsRNA), as well as compositions and methods for inhibiting the expression of the PCSK.9 gene w a cell or mamma! using such dsRNA. The invention also provides compositions and methods for treating pathological conditions that can modulated by down regulating the expression of the PCSIC9 gene, such as hyperhpklerrha. The dsRNA of the invention comprises an RNA strand (the anlisense 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 m-RNA transcript of uie PCSK9 gene.
In one embodiment, the mventi on provides double-stranded ribonucleic ycid fdsRNA) molecules for inhibiting the expression of the PCSK9 gene. The dsRNA comprises ai least two sequences that are complementary to each other. The slsRJNA comprises a sense strand comprising a first sequence and an antise-use strand comprising a second sequence. The antiseme strand comprises a nucleotide sequence which is substantially complementary to at least part, of an KiRN A encoding PCSK9, and the region of complementarity is less than 30 nucleotides in length, generally ! S -24 nucleotides to 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 oft.be sense sequences of Table 1 &nά 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 he comprised of naturally occurring nucleotides or cars be comprised of at least one modified nucleotide, such as a .2'-O- methyl modified nucleotide, a nucleotide comprising a S'-phαsphorothioate group, and a terminal nucleotide linked to a ehoiesteryl derivative. Aitemaiively, the modified nucleotide may he chosen from the group of: a 2!-cieoxy-2!~fsικ>rø modified nucleotide, a 2;-deoxy-modified nucleotide, a locked nucleotide, an abasic nucleotide, 2'-ami.no-modified nucleotide, 2'-alkyl- modified nucleotide, morpholmo nucleotide, a phosptsoramidate. 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 I and Table 2 and a second sequence selected from the group consisting of the antisense sequences of Tables I, and Table 2,
Sm another embodiment, the invention, provides a eel! comprising one of the dsRNAs of the invention. The ceil is- ' ό &evnerailv a mammalian ceil, such as a human cell.
In another embodiment, the invention provides a pharmaceutical composition for inhibiting the expression of the PCSK9 gene in an organism,, generally a human subject, comprising one or more of the dsRNΛ 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 POSK.9 gene m a cell, comprising the following steps;
(a) introducing into the eel; a double- stranded ribonucleic acid (dsRN A), wherein the dsRN A 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 amiseαse strand comprises a region of complementarity which Ls substantially complementary to at least a part of a raRNA encoding PCSK9, and wherein the region of complementarity is leas than 30 nucleotides in length, generally 19-24 nucleotides in length, and wherein the dsRNA, upon contact with a cell expressing the PCSKO, inhibits expression of the PCSK9 gene by at least 40%; and
(fo) maintaining the cell produced in step (a) for a time sufficient to obtain degradation of the niRNA transcript of the PCSK9 gene, thereby mhibήing expression of the PCSK9 gene in the ceil
b another embodiment, the invention provides methods for treating preventing or managing pathological processes which can be mediated by down regulating FCSK9 gene expression, e.g. hyperiipidemia, comprising administering to a patient in need of such treatment, prevention or management a therapeutically or prophylacticaily effective amount of one or more of the ds'RNAs 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 io a nucleotide sequence that encodes at føast one strand of one of the dsRNA of the invention.
In another embodiment, the invention provides a cell comprising a vector for inhibiting 5 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.
B iief Descr jptstm of the Figures
Fig. I shows lhe structure of the ND-9S lipid.
1.0 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 diflereot ORf regions of PCSK9 mRNA (having ihe ilrst nucleotide coiresponding to the ORF position indicated on the graph) in C57/BL6 mice (5 animals/group). The ratio of PCSK9 mRNA to CsAPDH mRNA in liver lysates was averaged over each treatment group and compared to a eorstroi group treated with !5BS or a control group
15 treated with an unrelated siRNA (blood coagulation .(actor VO).
Fig. 3 shows the results of the in vivo screen of 16 humasv'rooase/rat crossreactive (AL- DP-931 ! through ΛL-DP-9326) PC^SK9 SiRNAs directed against different DRF regions of PCSK9 niRNA (having the first nucleotide coπxsspondύig to the ORF position indicated on the graph) in C57/BL6 mice (5
Figure imgf000008_0001
1"he ratio of PCSK9 mRNA to GAPDH niRN A in 20 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 VU),
Silencin 1Sg of PCSK9 mRNA resulted in lowering total serum cholesterol levels.
The most efficacious in terms of knocking down PSCX9 message siRNAs showed the most pronounced cholesterol lowering effect (around 20-30%).
25 Fig. 4 shows the results of the in vivo screen of 16 mouse specific {AL-DP-9327 through
AL-DP-9342) PCSK9 siRNAs in C57/BL6 mice (5 aniraais/group). Total serum cholesterol levels were averaged over each treatment group and compared to a control group treated with PBS or a conirol group treated with an unrelated siRRA (bkxxi coagulation factor VII).
Fsg. 5 shows the results of the in vivo screen of } 6 hurrs&n/motis&'rat crosss'caetive (AL- DP-931 1 through AL-OP-9326) PCSK.9 si RNAs in C57/BL6 mice (5 animais/groυp}. Total ^erum cholesterol levels were averaged over each treatment group and compared to a control group treated with PBS or a control gtoop treated with an unrelated si RNA (blood coagulation factor VO).
Fig, 6 shows a comparison of the in vitro and m vivo results for silencing PCSK9.
Fig, 7A and fig, 7B show /« vitro results for silencing PCSK.9 using monkey pmrs&ry hepatocytεs.
Fig, 8 shows in vivo sciivity of LNP-OI foniiuiiUed siRNAs to pcsk-9.
Fig. 9 shows in vivo activity of LNP-OI Fonmdated chemically modified 9314 and 10792 parent molecules at different times, Oeariy modified versions of 10792 display if? vivo silencing activity.
Detatied Descnptioiϊ of Use I'avcjϊtloa
The iϊvvaπtioϊi provides a solution to the problem of treating diseases thai can be modulated by the down regulation of the PCSK9 gene, by using doubie-straπdαl ribonucleic acid (dsRNA) to silence the PCSK9 gene thus providing treatment for diseases such as hyperlipidemia.
The invention provides double-stranded ribonucleic acid (UsRNA), as well as compositions and methods lor inhibiting the expression of the PCSK9 gene in a cell or mamma: 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 the PCSK.9 gene. dsRNA directs the sequence- specs Hc degradation of πiRN.A through a process knowrs as RNA mterfersncε (RNAi). 'The dsRNA of the invention comprises an RNA sirand (the antisense strand) having a region which is less than 30 nucleotides in length, generally 19-24 nueleotiάes in length, and is substantially complementary to at least part of an .O)RNA transcript of the PCSK9 gene. The use of these dsRNAs enables the targeted degradation of an, mRNA that is involved in sodium 5 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 P€SK.9 gerse. Thus, the methods and compositions of the invention comprising these dsRNAs are useful for treating pathological processes which can be mediated by down regulating PCSK.9, such as in the treatment oOiyperhpkknna,
IO The following detailed description discloses how to make and use the dsRNA and compositions containing dsRNA to inhibit tbe 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 invention comprise a dsRNA having an antisense strand comprising a region of complementarity
I S 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 PCSK5) gene, together with a pharmaceutically acceptable carrier.
Accordingly, certain aspects of the invention provide pharmaceutical compositions comprising the dsRNA of the invention together with a pharmaceutically seceptahle carrier, 20 methods of using the compositions to inhibit expression of the FCSK5) gene, and methods of using the pharmaceutical compositions Io ireat diseases that can he modulated by down remdathiE the ex -ip'ression of P€SK.9.
L Defmitlpns
For convenience, the meaning of certain terms and phrases used in the specification, 25 examples, and appended claims, are provided helow. 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, the definition in this section shaii prevail "G," ''1C " "A" and. !!ϋ" each generally stand for a nucleotide that contains guanine, cytokine, adenine, and uracil as a bass, respectively. However, it will be understood that s.he term "ribonucleotide" or
Figure imgf000011_0001
can also refer to a modified nucleotide, as further detailed below, or a surrogate replacement moiety. The skilled person is well aware that guanine. eytosme, 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 irsαsine as its base may base parr with nucleotides containing adenine, eytosinε, or uracil. Hence, nucleotides containing uracil, guanine, or adenine may be replaced in the nucleotide sequences of the invention by a nucleotide containing, for example, inosme. Sequences comprising such replacement moieties are embodiments of the invention.
As used herein, "PCSK9" refers to the proproteiπ eonvertase subtiϊism kexin 9 gene or protein (also knowsi as FH3» HCHGLA3, NARC-I , NARC I )- mRNA sequences to PCSK.9 are provided as human; NM J 74936; mouse: NM 153565. and rat: NM^ 199253.
As used herein, NVt&rget sequence'1 refers to a contiguous portion of the nucleotide sequence of as mRNA molecule formed durum the transcnmiou of die PCSK9 uene. uicludins?. mRNA that is a product of RNA processing of a primary transcription product.
As used herein, the term "strand comprising a sequence" refers to aα oligonucleotide comprising a chain of nucleotides that is described by the sequence referred k> using the standard nucleotide nomenclature.
Aa used herein, and unless otherwise indicated, the term "complementary," when used to describe a first nucleotide sequence in rehnion to a second nucleotide sequence, refers to the ability of an oligonucleotide or polynucleotide comprising the first nucleotide sequence to hybridize and fomi 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 oiay include: 400 mM NaCl, 40 IBM PIPES pϊ-3 ό A i rnM EIJ[ A, 5QX: or 7CfC for 12-16 hours followed by washing. Other conditions, sucli as physiologically relevant conditions as
Figure imgf000011_0002
be encountered inside an organism, can apply. The skilled person will he 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.
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 ears be referred to as "fully complementary" with respect to each olher herein. However, where a first sequence is referred to as "substantially complementary" with respect to a second sequence herein, the two sequences can he 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 \o their ultimate application. However, where two oligonucleotides are designed to form, upon hybridization, one or more single stranded overhangs, such overhangs shall not he regarded as mismatches whli regard to the determination of corapiementaruy. for example, a dsRNA comprising one oligonucleotide 21 nucleotides in length and another oligonucleotide 23 nucleotides in length, wherein the longer oligonucleotide comprises a aequen.ee of 2! nucleotides that is MIy complementary to the shorter oligonucleotide, may yet be referred to as "fully complementary" for fee purposes of the invention,
"Complementary0 sequences, as used herein, may also include, or he formed entirely from, fion-Watsoii-€rick base pairs and/or base pairs termed from non-natural and modi 0 eel nucleotides, in as far as the above requirements with respect to their ability to bybridixe are fulfilled.
'The terms "complementary"',, "fully complementary" and "substantially complementary'* herein may he used with respect to the base matching between the sense strand and the and sense strand of a ds&KlA, or between the anύsensc strand of a dsRNA and a target sequence, as will be understood from the context ol their use.
As used herein, a polynucleotide which is "substantially complementary to at least part of a messenger RNA (mRNAj refers to a polynucleotide which is substantially complementary to a contiguous portion of the raRNA of interest (e.g., encoding PCSK9). For example, a polynucleotide is complementary to at least a part of a PCSK9 raRNA if the sequence is substantially complementary to a ruπi-iπterrupted portion of a mRN A encoding PCSK9.
The terπi "double-stranded RNA" or ~"d$RN,V\ as used herein, refers to a complex of ribonucleic acid molecules, having Ά duplex structure comprising two aaii-parallei and substantially complementary, as deimed 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 cisRNA art oiten referred to in me literature as si RN A ("short interfering RNA"). Where the two strands are part of one larger molecule, wiά therefore are connected by an uninterrupted chain of nucleotides between the 3'- eud of one strand and the S'end of the respective other strand forming the duplex structure, the connecting RNA chain is referred to as a "hairpin loop", "short, haiφin RNA" or "shRKLAΛ Where the two strands are connected covalently by means other than 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 structure is referred to as a >s!inker*\ The RNA strands may have the same or a different number of nucleotides. The maximum number of base pairs is the number of nucleotides hi the shortest strand of the cisRNA minus any overhangs that are present in the duplex. In addition to the duplex structure, a dsRN. A may comprise one or more nucleotide overhangs, hi addition, as used in this specification, "dsRN A" 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 irs an siRNΛ type molecule, are encompassed by 5idsRNΛvl for the purposes of this specification and claims.
As used herein, a "nucleotide overhang" refers to die impaired nucleotide or nucleotides that protrude from She duplex structure of a dsRNA when a 3'-end of one strand of the dsRNA extends beyond the 5'-εnd of ihe other straiui. or vice versa, "Blunt" or "blunt end" means that there are BO unpaired nucleotides at ihat end of the dsRNA, i.e.. no nucleotide overhang, A "blunt ended" dsRNA is a dsRNA that is double- stranded over its entire length, i.e., no nucleotide overhung at either end of the molecule. For clarity, chemical caps or non-rsucieotide
I i chemical moieties conjugated to the 3' end or 5' end of an si RNA are not considered in determining whether an si'RNA has an overhang or is blunt ended.
The term "antisense strand" refers to the strand of a dsRNA which includes a region that is substantially complementary to a target sequence. As used herein, ihe term, "region of cxnnplemeufanLy" refers to the region on Ihe antisense strand thai is substantially complementary to a sequence, tor example a target sequence, as defined herein. Where the region of complementarity is not fully complementary to the target sequence, the mismaSe&es are most tolerated in ihe terminal regions and, if present, are generally in a ierrnimd region or regions,, e.g., within 6, 5, 4, 3, or 2 nucleotides of the 5' and/or 3' terminus.
The term "sense .strand,"' as used herein, refers io the strand of a dsRNA thai includes a region that is substantially complementary to a region of ihe anϋseπse strand.
"Introducing into a ceil", when referring to a dsRNA, mearss facilitating uptake or absorption into ihe ceil as is understood by those skilled in the an. Absorption or uptake of dsRNA can occur through unaided diffusive or active cellular processes, or by auxiliary agents or devices. The meaning of this ierrn Is not limited to cells in vtiro; a oLsRMA. 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 ihe organism. For example, for /?? vivo delivery, dsRNA can be injected into a tissue site or administered systemicaliy. In vitro introduction into a cell includes methods known in the art such as eleetropαration and lipøfeciiem.
The terms "silence" and 'inhibit the expression of, in as far as they refer to the PCSK^ gene, heroin refer to the at least partial suppression of the expression of ihe FCSK9 gene, as manifested by a reduction of the amount of mRNA transcribed from the PCSKf) gene wbich may be isolated from a first cell or group of cells in whkh the PCSK-) gens ss transcribed and which has or have been treated such thai the expression of ihe FC1SKv gene is inhibited, as compared to a second ceil or group of cells substantially identical to the first cell or group of cells but which has or have not been so treated (control cells). The degree of inhibition is usually expressed in terms of (πiRNA in control cells) ~ (rnRN A in treated cells?
• 100% imRNA in control cells)
Alternatively, the degree of inhibition may be given m terms of a reduction of a parameter thai is functionally linked to PCSK9 gene transcription, e.g. the amount of protein encoded by the PCSfs.9 gene which is secreted by a cell, or the number of cell's displaying a certain phenotype, e.g apoptosis. In. principle, PCSK9 gene silencing may be determined in any ceil expressing the target, either eoostitutiveiy 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 hisi&αt invention, the assay provided in the Examples below shall serve as such reference.
For example, in certain instances, expression of the PCSK 9 gene is suppressed by st least about 20%, 25%, 35%, or 50% by administration ofihε double-stranded oligonucleotide of the invention, fa some embodiment., ihe PCSK.9 gene is suppressed by at least about 60%. 70%, or 80% by administration of the double-stranded oligonucleotide of the invention. In some embodiments, the PCSK.9 gene is suppressed by at least about 85%, 90%, or 95% by administration of ihe double-stranded oligonucleotide of the invention. Tables ls 2, provides a wide range of values for inhibition of expression obtained in an it! vitro assay using various PCSK9 dsRNA molecules at various concentrations.
As used herein w the context of PC1SK*) expression, the terms "treat", "treatment", and the like, κS&- 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 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 aϊ least one symptom associated with such condition, or to slow or reverse the progression of sucli condition, for example, in the context of liyperiipidemia. treatment will involve a decrease in serum lipid levels,
As oscd herein, the phrases "therapeutically effective amount" and "prophyiaciicaliy effective amovmt" refer io an amount that provides a therapeutic benefit in the treatment, prevention, or management of pathological processes which can be mediated by down regulating the PCSKO gent; on or an overt symptom of pathological processes which can be mediated by 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 patho logical 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 other anti-pathological processes which can be mediated by down regulating PCSK9 gene expression.
As used herein, a "pharmaceutical com position" comprises a pharmacologically effective amoiπu of a dsRNA and a pharmaceutically acceptable carrier. As used herein, "pharmacologically effective amount,"' "therapeutically effective amount" or simply "effective 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'HJ reduction in a measurable parameter associated, with a disease or disorder, a therapeutically effective amount of a drag for the treatment of thai disease or disorder is ihe amount necessary to effect at least a 25% reduction in that parameter.
The term "pharmaceutically acceptable earner" refers to a carrier for aάπruύstration 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 teπtt specifically excludes cell culture medium.
As used herein, a '"transformed cell" is a cell into which a vector has been, introduced from which, a dsRNA molecule may be expressed.
In one embodiment, the invention provides double-stranded ribonucleic acid (dsRNA) molecules tor inhibiting tiie expression of the FCSK9 gene in a cell or mammal wherein the dsRNΛ comprises an antssense strand comprising a region of complementarity which is complementary Io at least a part of an niRNA formed in the expression of the PCSK9 gene, and wherein the region of complementarity is less than 30 nucleotides m length, generally 19-24 nucleotides irs length, arsd wherein said dsRNA, upon contact with a cell exnressma; said PCSK9 gene, inhibits the expression, of said P€SK.9 gene by at least 40%. The dsRNΛ comprises two RNA strands thai are sufficiently complementary to hybridise to form a duplex structure.. One strand of the dsRNA {the anti sense strand) comprises a region of complementarity that is substantially complementary, and generally fully complementary, to a large! sequence, derived from the sequence of an mRNA formed during the expression of the PCSK.9 gene, the other strand (the sense strand) comprises a region which is complementary to the aπtisense 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 1.8 and 25, yet more generally between ! 9 and 24, and most generally between ! 9 and 21 base pairs in length. Similarly, the region of complementarity to the target sequence is between ! 5 and 3ϋ, more generally between I S and 25, yet snore 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 dsRNΛ 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 Biosystems, Inc. In a preferred embodiment the PCSKO gene is the human PCSO 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 irom the group consisting of the ami sense sequences of Tables 1 and 2. Alternative ^nusense agents that target elsewhere in the target sequence provided in Tables 1 uτιά 2, can readily he 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. Lu other embodiments, the dsRNA comprises at least t^o sequences selected from this group, wherein one of the at least t^vo sequences is complementary to another of the at least two sequences, nx\ά one of the at least two sequences is substantially complementary to a sequence of an uiR.NA generated irs 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 is described as the anliserise strand in Tables 1 and 2
The skilled person is wcli aware that dsRNAs comprising a duplex structure of between 20 and 23, biu specifically 21 , base pairs have been hailed as particularly effective in inducing S RNA interference (Eibashir et ah, EMBO 2(.HlI , 20;6877-68§8). However, others have found Uiat shorter or longer dsRNAs can be effective as well. In the embodiments described above, by virtue of the nature of the oligonucleotide sequences provided in Tables 1 and 2, .he dsRNAs of the invention can comprise at least, one strand of a length of minimally 2 int.. Ii can be reasonably expected thai shorter dsRNAs comprising one of the sequences of Tables 1 and 2 minus only a
I O few nucleotides cm one or both ends may he similarly eSleetive as compared Lo ihe dsRNAs described above. Hεnce, dsRNAs comprising a partial sequence of a*, least ! 5, l<x 17, I S, 19, 20, or more contiguous nucleotides from one of the sequences of Tables 1 and 2, and dulering in their ability to inhibit the expression of Hie PCSK.9 gene in a FACS assay as described herein beiow by not more than 5, 10, 15, 20, 25 , or 30 % inhibition from a dsRNA comprising the full
\ 5 sequence, are contemplated by die invention. Further dsRNAs that cleave within the target sequence provided in. Tables 1 and 2 can readily be made using ihe PCSK9 sequence and the target sequence provided.
In addition., ihe RNAi agents provided m 'fables 1 and 2 identify a site m the PCSK9 mRNA that is susceptible to RNAi based cleavage. As such the present invention further 0 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 RNAi agent if the second RNAi agent cleaves the message anywhere within the niRNA that is complementary to the tmtiseπse strand of the first RNAi agent. Such tx second agent will generally consist of at least 15 contiguous nucleotides lfom one of the sequences provided in
25 Tables I and 2 eotφled to additional nucleotide sequences taken from the region contiguous to the selected sequence in the PCSK9 gene. For example, the last 15 nucleotides of SEQ ID NQ:! (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 OTOvided in Tables 1 and 2. The dsRNA of the invention can contain one or more mismatches to the iarget sequence. Irs u preferred embodiment, the dsRN A of the invention contains no snore than 3 mismatches. If the arstisense strand of the ύsRNA contains mismatches to a target, sequence, it is preferable that fee area of mismatch not be located in the center of the region of complementarity. If the arsdseose sirand of the dsRNA contains mismatches to the target sequence, u is preferable that the rsmmatch be restricted to 5 nucleotides .from either end. for example 5. 4, 3. 2, or 1 nucleotide from either the 5 ' or 3 s eml of the region of complementarity. For example, for a 23 nucleotide dsRKA. strand which is complementary to a region of the PCSK9 gene, the άsRNA 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 die expression of the PCSK9 gene. Consideration of the efficacy of dsilNAs 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 dsRN A has a single-stranded nucleotide overhang of 1 to 4, generally 1 or 2 nucleotides, dsRNAs having at least one nucleotide overhang have unexpectedly superior inhibitory properties ihan their bUwtf-ended counterparts. Moreover, the present inventors have discovered thai the presence of only one nucleotide overhang strengthens the interference activity of ihe dsRNA, wnhoui affecting its overall slahihly. dsRNA having orJy one overhang has proven particularly stable and effective in vivo, as well as m a variety of cells, cell culture mediums, blood, and scrum. Generally, the single- siranded overhang is located at the 3f-terxnin_ιl end of ihe andsease strand or, alternatively, at the .V-iermina! end of the sense strand. The dsRNA may also have a blunt end, generally located at the 5 '-end of the ami sense strand. Such tlsRNAs have improved stability and inhibitory activity, thus allowing administration at low dosages, Le,, less than 5 nig/kg body weight of the recipient per day. Generally, tns antisense strand of the dsRNA has a nucleotide overhang at the 3 "-end, and the 5 '-end is biunt. In another embodiment, one or more of the nucleotides in the overhang is replaced with a nucleoside thiophosphate. hi yei another embodiment, the dsRNA is chemically modified to enhance siabiϋiy. The nucleic acids of the invention may be synthesized and'Or modified by methods well established in the art, such as lhose described in ''Cu item protocols in nucleic acid chemistry", Beauoage, S,L. el al. fEάrs.}, John Wiley & Sons, Inc., New York, NY, USA, which is hereby incorporated herein by reference. Chemica! modifications may include, but are not limited to 2! modifications, modifications at other sites of the sugar or base of <m oligonucleotide, introduction of non-natural bases into the oligonucleotide chain, covaient attachment to a ligand or chemical moiety, and replacement of intemncleotick phosphate Linkages with alternate linkages such as tm'αphαsphates. More than one such modification maybe 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 eovajent, ionic or hydrogen bonds; hydrophobic interactions, van der Waals or stacking interactions; by means of raetai-ion coordination, or through use of purine analogues, Generally; the chemical groups that car? he used to modify the dsRNA include, without iimitaiiors, methylene blue; bifunctiona) groups. generally his-(2-chk)roethyl)arnme; N-acetyl-N4p-gIyαxyihen?x>yl)cyst&mine; 4~thiσuraeif; and psoralen, In one embodiment, the linker is a hexa-ethyieπe glycol linker, hi this cass, the dsRNA are produced by solid phase synthesis and the hexa-ethyiene glycol linker is incorporated according to standard methods (e.g., Williams, DX, and K.S. HaH, Biochem. ( 1996) 35: 14665- 14670). Jn a particular embodiment, the 5'-eπd of the anti&ense strand and the 3!~end of the sense strand are chemically linked via a hexaeOiyleπe glycol linker. In another embodiment at least one nucleotide of t.be dsRNA comprises a pbosphorothioate or phosphorodithioate groups. The chemical bond at the ends of the dsRNA is generally formed by triple-helix bonds. Tables 1 tmύ 2 provides examples of oκx1ified RNAi aaenta of the inversion.
In yet another embodiinent, lhe 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 example, without limitation, certain nucleases. Techniques for inhibiting the degradation activity of cellular enzymes against nucleic acids am known m the ar! including, hut nol limited to, 2'- amino mαtiifications, 2 '-amino sugar modifications, 2"-P sugar modifications, 2%-F modifications, 2'-alkyl sugar modifications, uncharged backbone modifications, mαipholmo modifications, 2'--O-rnethy1 modifications, and phøsphøramidate (see, e.g., Wagner, NaL Med, (1995) 1 :1 116-8). Thus, at least one 2'4rydroxyl group of the nucleotides on a dsRNA is replaced by a chemical group, generally by a I'-amiπo or a 2*-methyi group. Also, at least one rmdeotide may be modified to form a locked nucleotide. Sue!) locked nucleotide contains a methylene bridge thai connects the 2 '-oxygen of ribnse with the ^-carbon of riboβε. Oligonucleotides containing the locked nucleotide are described in Kosbkin, A. A., et aL Tetrahedron ( 1998), 54: 3607-3630} and Obika, S, et a!.. Tetrahedron LvU. ( 199S), 39; 5401 - 5404), Introduction of a locked nucleotide into an oligonucleotide improves live affinity for complementary sequences and increases the melting temperature, by several degrees (Braasch, D,A. and D. R, Corey, Chem. Bioi. (2001 }, 8:1-7).
Conjugating a ligand to a dsRNA can enhance its cellular absorption as well as targeting io 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 Hver cells. Alternatively, the ligand conjugated to the άsRNA is a substrate for receptor-mediated eπdαcytosis. These approaches have been used to facilitate eel! permeation of&ntisεnse oligonucleotides as well as dsRNA agents. For example, cholesterol has been conjugated to various amisai$e oligonucleotides resulting in compounds thai are substantially more active compared to their non-conjugated analogs. See M. MaBoharai? Λπiisense & Nuckic Λad Drug Development 2002, /2, 103, Other lipophilic compounds thai have been conjugated to oligonucleotides include l.-pyτene butyric acid, 1 ,3-bb-O-
(hexade'csi)glyeeroi, and menthol One example of a ligand for receptor-mediated endocvtosis is folic acid. Folk acid enters the cell by folέue-recepior-niediated endαeylαsis. ds.RNA com pounds bearing folic acid would be efficiently transported into the cell via the folate- receptor-mediated ersdocyiosis. Li and coworkers report that attachment of folic acJd to the T- terminus of an oligonucleotide resulted in an S~fb;d Increase in cellular uptake of the oligonucleotide. IJ, S-; Dεshmukh, H. M.; Huang, L Pharm. Res. 1998, /5, 1540. Other Ijgands that have been conjugated to oligonucleotides include polyethylene glycols, carbohydrate clusters, cross-iiαking agents, porphyrin conjugates, delivery peptides and hpids such as cholesterol. In certain instances, conjugation of a eatiome ligand to oligonucleotides results in improved resistance to nucleases. Representative examples or'eationsc hgaiids arc propylammoninrπ ami dimeihylpropyhimnioniitm. Interestingly, amiseπse oligonucleotides were reported to retain their high binding affinity to mRNA when the cationic iigand was dispersed t throughoiH the oligonucleotide. See M. Manoharan Antisense ά Nucleic Acid Drug Development 2902, 12, 103 »ncl references therein.
The Hgand-conjugated άsRNA of the invention may be synthesized by the use of a dsRNΛ that bears a pendant reactive functionality, such as that derived irom the attachment of a linking molecule onto the dsRNA. This reactive oligonucleotide may be reacted directly with commercially-available ϊigands. ligands thai are synthesized bearing any of a variety of protecting groups, or ligands that have a linking moiety attached thereto.. The methods of the invention facilitate the synthesis of ϋgand-conjugatεd 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 scrum-binding Iigand with a Unking moiety located on the 5' position of a nucleoside or oligonucleotide. In certain instances, an dsRNA bearing an aralkyi Iigand attached to the 3 '-terminus of the dsRNA is prepared by first eovalently attaching a monomer building block to a controlied-pore-glass support via a long-chain arai.noalkyi group. Then, nucleotides are honcied via standard solid- phase synthesis techniques to the monomer buiiding-biock hound to the solid support. The monomer building block may he a nucleoside or other organic compound that is compatible with so iid-p base syπth asi s .
'The dsRNA used in the conjugates of the invention rnaybs conveniently and routinely made through the weli-knowπ technique of solid-phase synthesis. Equipment for such synthesis is sold by several vendors including, for example. Applied Biαsystεms (Foster City, CA). Any other means for such synthesis known in the art may additionally or alternatively be employed. Ii is also known to use similar techniques to prepare other oligonucleotides, such as the pbosphorothioates and alkylated derivatives. Teachings regarding the synthesis of particular modified oligonucleotides may be found in the following U.S. patents: U.S. Pat. Nos. 5,138,045 and 5,218,105, drawn to polyamiπe conjugated oligonucleotides: U.S. Pat. No, 5.212.295, drams Lo monomers for the preparation of oligonucleotides having chiral phosph.on.5s linkages; LJ. S- Pat Nos. 5.37S.S25 ami 5,541 ,307, drawvj Io oligonucleotides having modi fled backbones; U S. Pat. No. 5386,023, drawn io backlxme-mαdiiled oligonucleotides and the preparation thereof through reductive coupling; U.S. Pat. No. 5,457,191, drawn to modified nuclεobases based on the 3-deazapurine nag system and .methods of synthesis thereof; U.S. Pat No. 5,459,255, drawn to modified nueleobases based on N-2 substituted purines; U. S, Fat. No. 5,521,302. drawn to processes for preparing oligonucleotides having ehir&l phosphorus linkages; U. S Fat. No. 5,539s0S2. drawn to peptide nucleic acids; U.S. Pat. No. 5,554,746. drawn to oligonucleotides having pMactam backbones; U.S. Pat. No. 5.571 ,902» drawn io methods ami materials for the synthesis of oligonucleotides; U.S. Pat. No, 5,578,718» drawn to nucleosides having aikylthio 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.SS7.361 and 5.599,797, drawn to oligonucleotides having phosphorothioate lin&ages of high chiral parity; U.S. Pat, "No. 5,506351 , drawn to processes for the preparation of 2?-(>--alkyi gii&nosme and related compounds, including 2.ό~diatT!inopu.nπe compounds; U.S. Pat. No. 5,58?,4C>9, drawn to oligonucleotides havmg N-2 substituted purines; U.S. Pat, No. 5.587,470, drawn to oligonucleotides having 3 -de&xap urines; U, S Pat. No. 5,223,.K(S, and IKS. PaL No. 5,608,046, both drawn to conjugated 4'~desm ethyl nucleoside analogs: U.S. Pat. Nαs.
5.602.240, and 5,610,289, drawn to backbone-modified oligonucleotide analogs; U.S. Pat, Nos,
6.262.241, mύ 5,459,255, drawn to, inter aiia, methods of synthesizing 2!-flιsorø- oli se»o^ nycieotides.
In lbs iigand-coπjugated dsRNA and ligand-molecuk bearing Bsqnence-ψ«cific linked nucleosides of ihe invention, the oligonucleotides and oligonudeosides may be assembled on a suitable DNA synthesizer uiilizing standard nucleotide or nucleoside precursors, or nucleotide or nucleoside conjugate precursors that already bear (he unking moiety, figand-πudeoiide or nueleoskie-eorsrαgais precursors that already bear the ligand molecule, or non-mseieoskfe ligand- bearine binldina blocks. When using nudeoiide-coπjugate precursor that already bear a linking moiety, the synthesis of the sequence-specific; linked nucleosides is typically completed, and the hgand molecule is then reacted with the linking moiety to form the ligand-corrjugsted oligonucleotide. Oligonucleotide conjugates bearing a variety of molecules such as steroids, Vitamins, lipids and reporter molecules, has previously been described (see Maπoharan et ai, PCT Application W() 93/07883). In a preferred embodiment ihe oligonucleotides or linked nucleosides of the invention are synthesized by an automated synthesizer using phosphofainidites derived from lig&nd-rmcleoside conjugates in addition to the standard pbosphoramidites and non-standard phϋspiior&msciues that are commercially available and routinely used in oligonucleotide synthesis.
The incorporation of a 2 -0-roethyL 2!-O-etiryL 2!-()~propyL 2'-OaIIyI, 2;-Q-armnoalky! or 2'-deoxy-2'-fluoro group in nucleosides of an oligonucleotide confers enhanced hybridization properties to the oligonucleotide. Further, oligonucleotides containing phospborothioaie backbones have enhanced nuclease stability. Thus, functionanzed, linked .nucleosides of the invention can be augmented to include either or both a phosphorolhio&ie backbone or a 2'-G- meihvl 2"-0-ethy!, 2'-O-nropyi, 2!-O-amirjoalkyl, 2'-OaIIy! or 2!-deoxy-2!~fϊuoro group. A stsnmiary lisϋng of some of the oligonucleotide modifications known in the art is found at for exaniple, PCT Publication WO 200370918.
In some embodiments, functionali^ed nucleoside sequences of the invention possessing an amino group at the .S'-tennmus are prepared using a DNA synthesizer, and then reacted with an active ester derivative of a selected ligand. Active ester derivatives are weii known to those skilled in the art. Representative active esters include N-hydrosucdnimide esters, tetrafluoropheπolic esters, pentatluoroplienolic esters and peritachiorophenolic esters. The reaction of the amino group and the active ester produces an oligonucleotide In which ihe selected ligand is attached to the 5!-ρosit.ion through a linking group. Th« aπiino group at the 5*- terminus can be prepared utilizing a S'-A.mino~lvlodi1ler CO reagent. In one embodiment, Hgand molecules may be conjugated to oligonucleotides nt. the 5'-ρosition by the use of a Hgand- iuicleoside phosphorasnidite wherein the ligand is linked to the S'-hydroxy gronp directly or indirectly via a linker. Such ligand-πucleoside phosphoramidites are typically used at the end of
11 an automated synthesis procedure to provide a iigaπd-eoπjugated oligonucleotide bearing the Hgand at the S!~termious,
Examples of modified iruernucisoskie linkages or backbones include, for example, phosphoroihioat.es, chiral phosphorothioates, phosphoroditJhioaies, phosphoiriesters, aminoaikylphospbotriesters, methyl and other alkyl phosphorates including S'-aikyiene phosphonates and dikai phosphorsates, phosphinates, phosphoramidates including 3'-aimno phosp'horamkiaie and aminoalkyipααsphoraraidates, thionopbosphoramidates, ihioiioalk ylphosphorsales, .hiαnoaikylphosphotri esters, and boranophosphates having normal 3'~5! linkages, 2'-S' linked analogs of these, and those having invested 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 arid iree-aeid forms axe also included.
Representative United States Patents relating to the preparation of the above phosphorus- atom-coniaiiiing linkages inciude, but are not limited to, U.S. Pat. Nos, 3,687,808; 4,469.863; 4,4/6301; 5,023,243; 5,177J%; 5.188,897; 5,264,423; 5,276,019; 5,278,302; 5,286,7 ).?; 5,321 J 31 ; 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; 5,550,1 1 1; 5,563,253; 5,571 ,799; 5,58736 k 5,625,050; jiπd 5,6^7,248, each of which is herein, incorporated by reference.
Examples of modified internuejeoside linkages or backbones that do not include a phosphorus atora therein (i.e., oligonucieosides) have backbones that are farmed by short chain alkyl or eyeløalkyl intersugar linkages, πiixed hetcroatoπi and alky! or cyeloalkyl mters^gar Hakages, or osie or more short chain heteroalorrsie or heterocyclic intεrsugar linkages. 'These include those liaving τπorpholiπo linkages (formed in part from tile sugar portion of a nticieoside); siloxaαe backbones; sulfide, sulfoxide and sαlfone backbones; foroiacetyl and tliiofomiacety! backbones; methylene forrøacetyt and ihioiormricetyl backbones; alkcne containing backbones; siufamate backbones; ineiliy1eneir.ni.no and πiethyleαehydrazino backbones; sulfonate and sulfonamide backbones; amide backbones; and others having mixed N, O5 S and CH2 component parts. Representative United Slates patents relating to the preparation of the above oUgonucleosJdcs 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; 5,6/7,437; and 5,677,439, each Oi" wluch is herein incorporated by reference.
In certain instances, the oligonucleotide maybe modified by a noii-Hgand group. A number of non-ligarsd molecules have been conjugated to oligonucleotides m order to enhance the activity, cellular distribution or cellular uptake of the oligonucleotide, and procedures for
H) performing such conjugations are available in the scientific literature. Such nors-Ugand moieties have included lipid moieties, such as cholesterol (Letsioger et aL Proc, Natl, Acad, Sci, USA, 1989, $6:0553}.. cholic acid (Manoharan et aL Bioorg. Med. Clieπi. Lett., 5994, 4:1053}.. a thioether, e.g> ; Iiexyl-S-trUyithso} (Manαharan et al, Ann. RY. Acad, Sci., 1992, 660:306; Manohararj et aL, Bborg. Mεd. C hem. Let,, 1993, 3:2765), a thiochoksterol (Oberhauser et a!,,
1 S NucL Acids Res., 1992, 20:533), an aliphatic chain, e.g., dodεcandiol or undecyl residues
(Saison-Behmoaras et aL, EMBO 1, 1991, 10:1 11 ; Kabanov et a!., FEBS Lett,, 1990, 259:327; Sviπarchuk et al, Biochimie, 1993, ?5:49X a phospholipid, e,g,, di-hexadecyl-rac-glycerol or iriethylammonium KI-di-Ohexadecyl-rac-glycero-S-H-ph.osphon.ate (Manoharau et al.: Tetrahedron Lett,, 1995. 36:3651 ; Shea et al, NucL Acids Res.f 1990, ; S;37??κ a polyaπiine or a 0 polyethylene glycol chain {Manoharaυ ei aL, Nucleosides & Nucleotides, 1995, i 4:969k or ad&πiantaπe acetic acid CManoharan Oi aL, Tetrahedron Lett, 1995, 36:3651), a paimityl moiety (Kiishra et aL, Biochirrs. Biophys. AcU^ 1995, 1264:229), or an octadecykπiine or hexylamino- carbouyl-oxvcholesteroi nioiety (Crooke εt a!., ,L Pharmacol. Exp. Then, 1996, 277:923), Representative United Slates patents that teach the preparation of such oligonucleotide
25 conjugates have heeα listed above. Typical conjugation protocols involve (he synthesis of oligonucleotides bearing an aπn'nolmker at one or more positions of the sequence. Hie amino gϊx>op is then reached with the molecule being conjugated using appropriate coupling or activating reagems. The conjugation reaction may he performed either with the oligonucleotide siii! bound to ϊhε solid support or following cleavage of the oligonucleotide in solution phase. Purification of the oligonucleotide conj ugaie 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 irons recombinant viral vectors intraceUuiarly in vivo. The recombinant viral vectors of the invention comprise sequences encoding the dsRNA of lfse invention and any suitable promoter for expressing the dsRNA sequences. Suitable promoters include, far example, the Ij6 or H 1 RNA pol IH promoter sequences and the cytomegalovirus promoter. Selection of other suitable promoters is within the I O ski Il in the art The recombinant viral vectors of the invention can also comprise inducible or regolatable 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 IH discussed in more detail below.
dsR-NA of the inventsors can be expressed from a recombinant viral vector either as two 1 S separate, complementary RNA molecules, or as a. single RNA molecule with two complementary regions.
Any viral vector capable of accepting the coding sequences for the dsRNA moi.eeidefs) to be expressed can be used, tor example vectors derived from adenovirus ( AV); aderso-associated virus (AAV); retroviruses (e.g.. Ienti viruses ^LVl, Rhahdovimses, .murine leukemia virus); herpes 20 virus, and the like. The ϊropisjn of viral vectors can be modified by pseudotyping lhe vectors with envelope proteins or other surface antigens from other viruses, or by substituting different viral eapsiti proteins, as appropriate.
For example, lenϋviral vectors of the invention can be pseuαotyped with surface proteins from vesicular stomatitis virus (YSY 5, rabies. Ebola, Mokola, and Uw like, AAV vectors of the 25 invention can be made to target different ceils by engineering die vectors to express different capskl protein serotypes. For example, an AAY vector expressing a serotype 2 capaid on a
7 S serotype 2 genome is called AAV 2/2. This serotype 2 capsid gene is 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., RabinowUz j E at al (2002), I Virol 76:791 -8(Ji, the antim disclosure of which is herein 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 m the art. See, for example. Dornbiirg R (1995), Gεne Therap. 2: 301 -310; Eglitis M A (1988}. Biolechniqueκ 6: 608-614; Miller A D (1990), Hum Gene Therap. 1 : 5-14; Anderson W F ( 1998), Nature 392: 25-30; and Rubisison D A εt. aL, Nat, Genet. 33; 40!. -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- stranded RNA molecules from a recombinant AAV vector comprising, for example, either the IJ6 or H i RNA promoters, or the cytomegalovirus (CMV) promoter.
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, ar e described in Xia B et al. (2002), Nat. Biotech. 20: 1006-1010.
Suitable AAV vectors lor expressing the dsRN A of the invention, methods for constructing the recombinant AV vector, and methods tor delivering the vectors into target cells arc described in Sarnaiski R et al. S 1987), j. Viroi. 6! : 3096-3101 ; Fisher K J ct aL U 996), j, ViroL "0: 520-532; Sarmilski R el ai. ( 1989), J, ViroL 63: 3822-3826; U.S. Pat. No, 5,252,479; U.S. Pat. No. 5J 39,941 ; international Patent Application No. WO 94/13788; and international Patent Application No. WO 93/24641, the entire disclosures of which are herem incorporated by reference.
r? fi 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 pharmaceutics! composition comprising the dsRNA is useful for treating a disease or disorder associated with the expression or activity of the PCSK9 gene, such as pathological processes which can be mediated by down regulating PCSK9 gene expression, sucxh as hypεrlipidrøύa. Such pharmaceutical compositions are formulated based on the mode of delivery. One example is compositions that are formulated tor delivery Lo the liver via parenteral delivery.
The pharmaceutical compositions of the invention arc administered irs dosages sufficient 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 nig dsRNA per kilogram body weight of recipient par clay is siύϊicieni to inhibit or suppress expression of the FCSKS1 gene and may he administered sysiernically to the patient.
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 ! microgram to 1 mg per kilogram body weight per day. The pharmaceutical composition may be administered on.ee daily, or the dsRN A 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, ϊxi that case, the dsRNA contained in each sαh-dose mast be correspondingly smaller m order to achieve the total daily dosage. The dosage unit can also be compounded for 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 thai certain lactors may influence the dosage and timing required to effectively treat a subject including but not limited So the severity of the disea.se 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
\> 7 composition can include a single treatment or a series of treatments. Estimates of effective dosages and in vivo half-livss for the individual dsRNAs encompassed by the invention can be made using conventional methodologies or on the basis Q Ϊ in vivo testing using an appropriate animal mode), 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 hi vivo testing of dsRNΛ, as well as for determining a therapeutically effective dose.
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 injecϋon), or can occur over a period of time (e.g., by slow Infusion or administration of slow release formulations).
Typically, when treating a mammal with hyperlipldemia, the dsRNA .molecules are administered systemically via parental means. For example, dxRNAs, conjugated or unconjugate or lbnmdated with or without liposomes, can be administered intravenously Io a patient. For such, a άsRNA molecule can be formulated into compositions such as sterile and non-sterile aqueous solutions, non-aqueous solutions in common solvents such as alcohols, or solutions ifs 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 sierile aqueous solutions, which also can contain bulϊers. diluents, and other suitable additives (e.g., penetration enhancers, carrier compounds, and other pharmaceutically acceptable earners).
in addition, dsRNA molecules can be administered to a mammal as biologic or abiologic means as described in, for example, US. PaL No. 6,271 ,359. Abiofogic delivery can be accomplished by a variety of methods including, without limitation, (I) loading liposomes with a dsRNA acid molecule provided herein and (2) eonjplexing a dsRNA molecule with lipids or liposomes Io form nucleic acid- lipid or nucleic acld-iiposome complexes. The liposome can be composed of cationie and neutral Upkis commonly used to traπsfect cells />.' vitro. Catsonic lipids can complex (e.g., charge-associatei with negatively charged nucleic acids to form liposomes. Examples of catώπic liposomes include, without limitation, lipolectin, lipofεctamine, hpofeetaee, and DOTAP. Procedures for forming liposomes are well known in the art. Liposome compositions can be formed-, for example, from phosphatidylcholine, dimyristαyl phosphatidylcholine, dipalmiioy) phosphatidylcholine, dirπyristoyl phosphatiάylglycerol or eUokoyi phosphatldylethanolaπuπe. Numerous lipophilic agents arc commercial iy available, including Lipαfecun.RTM. (Invitrogen/tife Technologies, Carlsbad, CaIiD and Efreeterie.TM. (Qiagen, Valencia, Calif), In addition, systemic delivery methods can be optimized using commercially available cation ic lipids such as DDAB or DOTAP5 each of which can be mixed with a neutral lipid such as DOPE or cholesterol, fa some cases, liposomes such as those described by Tempteton. et at {'Nature Biotechnology, 15: 647-652 (1997)) can be used. In other embodiments, polycatiorss such as poiyethyleneimine can be used to achieve delivery m vivo and ex vivo (Boletta et a!.. J. Airs Soc, Nephrol. 7: 1728 ( 19%)}. Additional information regarding the use of liposomes to deliver nucleic acids can he (bund in U.S. Pat. No. 6.271.359, PCT Publication WO 96/40964 and Morrissey, D. et al. 2005. Nat Bioiechnol. 23(8): 1002-7.
Biologic delivery can be accomplished by a variety of methods including, without lii>ύtaϋoτi, the use of viral vectors. For example, viral vectors (e.g.. adenovirus and herpesvirus vectors) can be used to deliver dsENA molecules io liver cells. Standard molecular biology techniques can be used to introduce one or more of the dsRNAs provided herein into one of the maxsy different viral vectors previously developed io deliver nucleic acid to cells. These resulting viral vectors ear; be used to deliver she one or more dsRN As to cells by, for example, infection,
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 rnind so as to provide for the 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 irydroxypropyl niethyicelJuiose); tilt ere (eg,, lactose and other sugars, geialin, or calcium sulfate); lubricants (e.g., starch, polyethylene glycol, or sodiu.ni acetate); disintegrates le.g., search or sodium starch glycolate); and wetting agents (e.g., sodium iaury] 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 snore dsRNA agents that target the PCSK9 gene can contain other therapeutic agents such as othr lipid lowering agents (e.g., statins).
PCSK9
The methods and compositions described herein can he used to treat diseases and conditions that can be modulated by down regulating PCSK9 gene expression. Far example, the compositions described herein can be used to treat hyperhpidemia and other forms σfiipid inhafance such as hypercholesterolemia, hypertriglyceridemia mά the pathological conditions associated with tiuese disorders such as heart and circulatory diseases.
Methods for inhibiring expression of the PCSK9 gene
In yet another aspect, the invention provides a method for inhibiting the expression of the PCSK*) gene in a mammal. The method comprises administering a composition of the invention to the mammal such that expression of the target P€SK.9 gene is silenced. Because of their high specificity, the dsRNAs of the invention specifically target RNAs (primary or processed) of the target PCSKc) gens. 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 dsRN A, wherein the dsRNA comprises a nucleotide sequence which is complementary to at least a pan of an RNA transcript of the PCSK.9 gene of the mammal to be treated, When the orgarsisrrs to be treated is a mammal such as a human, the composition may be administered by arty 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 miusioo or injection.
Uniess otherwise defined, ail technical and scientific teπns 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, AU publications, patent applications, patents, and other references mentioned herein are incorporated by reference in their entirety. ID case of conflict, the present specification, including definitions, will control, In addition, the materials, methods, and examples are illustrative only and not intended to b& limning
EXAMPLES
Gene Walking of the PCSK9 gene
siRNA design was carried out to identify m two separate selections
a) siRNAs targeting PCSK9 human and either mouse or rat niRN A and
5 b) all human reactive siRNAs with predicted specificity to the target gene FOSK.9.
roRNA sequences to human, mouse and rat. PCSK9 were used; Human sequence NM J.74936.2 was used as reference sequence during the complete siRNA selection procedure,
19 m er stretches conserved in humars and mouse, axsd human and rat PCSO JΠRNA sequences were identified i.n the first step, resulting in the selection of siRNAs crossrεactjvε to 0 human and mouse, and siRNAs crossreactive to human and rat targets
SiRNAs specifically targeting human PCSK9 were identified in a second selection. AH potential ! 9mer sequences ofhuman PCSK9 were extracted and defined as candidate target sequences. Sequences cross-reactive to human, monkey, and those cross-reactive to mouse, rai, human and monkey are all listed in Tables 1 and 2. Chemically modified versions of those IS sequences and their activity m both ?>? vitro tmά 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 0 as preferable and assumed to be more specific in vivo.
For predicting siRN A-spedfic off-target potential the following assumptions were made;
1 ) positions 2 to c> (counting S' to 3") of a strand (seed regions may contribute more to off-target potential than rest of sequence (αoτi-seed imd cleavage site region)
10 2} posi.tio.ns IO and .1 1 (counting 5! to 3') of a strand (cleavage site region) may contribute more to off-target potential than non-seed region
3} positions i and 19 of each strand are not relevant for off-target interactions
4) an off-target score cars bo calculated for each gene and each strand, based on 5 complementarity of siRNA strand sequence to the gene's sequence arid position o C" mismatches
5} number of predicted off-targets as well as highest off-target score must be considered for oil-target potential
6) off-target scores are to be considered more relevant tor off-target potential than numbers of off-targets
I O ?) assuming potential abortion of sense strand activity by interna! modifications introduced, only off-target potential of antise.ose 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 caes IS off-target gene to calculate the off-rarget score:
Number σf 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 Uo 3 as follows:
20 Off-target score :::: number of seed mismatches 5MO
' mxiϊiber of cleavage Site mismatches * ! .2
÷ number of non-seed mismatches * 1 The most relevant off-target gene for each siR'NA corresponding to the input 19nier sequence was defined as the gene with the lowest off-target score. Accordingly, the lowest off- target score was defmed as the relevant off -target score for each si UNA.
Source of reagents
Where the source of a reagent Ls not specifically given herein, such reagent maybe obtained from any supplier of reagents tor molecular biology at a qualhy/pusity standard for application in molecular biology.
sJEISA. synthesis
Single-stranded RNAs were produced by solid phase synthesis on a scale of ! μmole using an Expedite 8909 synthesizer (Applied Biosysvenis, Appbra Deutsdiland €irnb-Bu Darmstadt, Germany) and controlled pore glass (CPG, 500A, ProHgo Bbcheniie GmbH, Hamburg. Cjerrπasy) as solid support. RNA and RNA containing 2 -ϋ~methyi nucleotides were generated by solid phase synthesis employing the corresponding phosphoraroidiies ami 2 -0- methyl phosphorarnidit.es, respectively (Proligo Bioehemie GmbH., Hamburg, Germany). These building blocks were incorporated at selected sites within the sequence of the oligoribonuciεoπde chain using standard .nucleoside phαsphoramidite chemistry such as described in Current protocols in nucleic acid chemistry, Beaucage, SJ.,, et a!. CEdrs.k John Wiley & Sons, inc., New York, NY, USA. Phosphorothioate linkages ware introduced by replacement of the iodine oxidizer solution with a solution of the Beaucage reagent {Chruachern Ltd, Glasgow, IiK) in aeetonitri Ie ( 1 %). Further ancillary reagents were obtained from MalHsickrodt Baker (Grieshcim, Germany),
Deprotecπon and purification of the catde oligortbαnucieotides by anion exchange HPLC wesre carried out according to established procedures. Yields and concentration?; were determined by IiV absorption of a solution of die respective RNA at a wavelength of 260 am using a spectral photometer (DU 640B, Beekman Coulter GmbH, ϋnterschleiBheϊm, Germany). Double stranded
RNA was generated by mixing an cquimoJar solution of complementary strands in annealing buffer (20 raM sodium phosphate, pH 6,8; 100 m'M. sodium chloride), heated in a water bath at 85 - 90cC for 3 rmrsuies 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-conjUgaied siRNΛs (herein referred to aβ -ChOkV), an appropriately modified solid support was used for RNA synthesis. The modified solid support was prepared as follows:
Diεlhvl-2-azabutane-h4-diearboχγIate AA
Figure imgf000037_0001
A 4.7 M aqueous solution of sodium hydroxide (50 mi) was added into a stirred, ice- 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 TIX, After I θ h the solution was partitioned with clicbSoroniethϋnε (3 x 100 snL). The organic layer was dried with anhydrous sotliisra stsifate, filtered and evaporated. The residue was distilled Io aftbrd AA (28,8 g, 61%).
3- (EllK>xycarbonylmeihyl-[6-(<)H-fluoren-9-ylniethoxycarbony!-aniino}--bexai.ioy!}-' amino ^ -propionic acid ethyl ester AB
Figure imgf000037_0002
AS
Fnioc-6-amino-hexanoic acid (9,12 g, 25,83 nimol} was dissolved in dichloromcthaπe (50 mL) and cooled with ice. Diisopropylcarbodiimde (3,25 g, 3,9s) niL. 25. S3 mmol) was added to the soUuion at if 'C. h was fhssi followed hv the addition of Dieihyl-azabotane-! ,4-dicarboxylate (5 g. 24.ό rørooO and dimethylamioα pyridine (0,305 g, 2,5 mmol}. The solution was brought to room temperature and stirred foπher for 6 h. Completion of the reaction was ascertained by TLC. The reaction mixture was concentrated under vacuum and ethyl acetate was added io precipitate diisopropyi 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 siU fate and concentrated to give the crude product which was purified by column chromatography (50 % EtQAOHexanes) to yield 1 1 ,8? » (SS%) of AB.
3~[{6-Amino-hexaiioy1}~cthoxycarhonylmethyl-affiiπoj-prαpioαic acid ethyl ester AC:
Figure imgf000038_0001
J O
3- ^Ethoxycarboiiylπieth>1-[ό~{yH-πuoren-^-ylmethoxycarbony(arnino)-hcxaπoy[]- amino I -propionic acid ethyl ester AB (1 1.5 g, 21.3 mmol) was dissolved in 20% piperidine \n dmiethylfoπnamide at ϋ"JC, The solution was continued stirring lor ! h. The reaction mixture was concentrated ursder vacuum, water was added to the residue, and the product was extracted with ethyl acetate. The crude product was purified by conversion into its hydrochloride salt.
34^(>-[17^ 1 , 5-Dimethyl-bexy1)-10J 3-dimethyi-2>'?I8,9α0!.l 1 ,12,n,I4,15,l 6.1 7- tetrsdecahydro-] ϊ-l-cyclopenta[a]phenaπtiiren-3~yioxycarbonylamino]- hsxanoγ]H-tlioxycarbonyimet!ryi-amino}-propioπie acid ethyl ester Al>
Figure imgf000038_0002
20 AB The hydrochloride salt of 3~[(6-Λmmo-htsxanoyl}~ethoxycarboτiyimethyl-asrimo'|- propioπie acid eihyi ester AC (4.7 g, 14.8 srmiol) was taken up in dichlαrømethaae. The suspension, was cooled to 0°€ on ice. To the suspension diisopropylsthyiamine (3.8? g, 5,2 raL, 30 mnioi) was added, To fhc resulting solution cholesteryϊ chlotoibrrrsate {6.675 g. 14.8 mmol) was added. The reaction mixture was stirred overnight. The reaction mixture was diluted with dichlorornethane and washed with 10% hydrochloric acid. The product was purified by flash chromatography ( 10.3 g, 92%).
ϊ
Figure imgf000039_0001
i 0.ϊ3-<IJmethyl-2,3Λ7,S.f)J0,l L] 2,! 3j4,I5α6.17- ietradecahydro~lH~cydopentafa| phenaBthrcii-?-y!oxycarbQnylami«o|-hexaHay! |~4-oxo- ρyrroHdiαe-3-carboxylic acid ethyl ester AE
Figure imgf000039_0002
AE
Potassium t-butox.ide { 1. i g, (>.S nuτrøl) was slurried in 30 raL of dry toluene. The mixture was cooled io 0''C- on ice and 5 g (6,6 mmol} ot'diester AD was added slowly with stirring within 20 rains. The temperature was kepi below S0C during the addition. The stirring was con tinned tor 30 rains ai {}"C and 1 ml, of glacial acetk acid was added, irjimediateiy followed by 4 g of NaI-IsPO4-I-LO ITS 40 ml.- of water The restutaui mixture was extracted twice with 100 ml, of diciiiorornethane each and the combined organic exiraets were washed twice with 10 mL of phosphate buffer each, dried, and evaporated to dryness. The residue was dissolved in 60 nil, of toluene, cooled to (PC and extracted with three 50 IYΪL portions of cold pH 9.5 carbonate butler. The aqueous extracts were adjusted to pϊ-i 3 with phosphoric acki, and extracted with five 40 mL portions of chloroform which were combined, dried and evaporated to dryness. The residue was purified by column chromatography using 25% eihyiaeetate/hexane to afford 1.9 g of b-keioester (39%).
[(>-(3-^Iv^roxy-4-hydroxymsihyl-pyττolidit1-i-yl}~0~oxo-hexy1]■■cαrbail)ic■ acid 17-0 ,5- dimethyl-hexylVlOJ 3-dimtfhyi-23,4J,S,9,l OJ 1 J2J 3..R] 5 J 6J 7-ιεtradβcahydrα-I H- cyclopeπta[ajphenaϊithreπ-3-yl ester AF
Figure imgf000040_0001
Methanol (2 m.t) was added dropwis€ over a period of i h to a refluxing mixture of b- ketoester AE (L5 g, 2.2 πmiol) and sαdhtrs horobydride (0.226 g, (> mmol) in tetrabydro&ran OO TΠL). Stirring Λvas continued at reflux temperature for 1 h. After cooling to room temperature, 1 N HQ (12.5 nil.) was added, the mixture was extracted with ethylacetatε (3 x 4ϋ mL). The combined sthylaceiate layer was dried aver anhydrous sodiαπi sulfate and concentrated under vacuum to yield the product whicb was purified by column chromatography (10% MeOH/CHCϊϊ) (89%).
{6- i3-[Bis-{4-n>ethoxy~phenyl)-phenyl-me{hoxymethyl]-4-h>jdroxy~pyrroHdin-] ~yl|-~6~ oxo-hexyu-carb&rruc acid !?■■( U5-difnethy1-hexyl)~10.13-dimethy1~
2:3 A J SS, I OJ 1 , 12Λ 3, 14 J 5,16 J 7-tεiradecahydro ■ I H-cycloρema|a jpheπanthren-3-yl ester AG
Figure imgf000041_0001
AG
Di o ! AF (1 ,25 gm 1,994 m«io!) vvas dried by evaporating with pyridine (2 x 5 jtiL) m ixifuch Anhydrous pyridine (H) πiL) and
Figure imgf000041_0002
13 rnmo!) were added with stirrmg. The reaction w;is carried out at room teπvperaiure overnight. Trie reaction was quetiched by the addition of methanol. The reaction mixture was concentrated under vacuum and to the residue dichloroπiethane {50 xnh) 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 puπOed by column chromatography (2% MeOR'Chioroform. Rf ::: 0,5 in. 5% MeOH/CHCiO s l.75 z* 95%),
Succinic acid moπo-{4-[bis~(4-rnetlioxy-pheτiyi}-pheπyl~metboxyιiiε1i>yrhl -K>-[l*'-{l,5 \_ dimeihyl-hexyj}-iϋj3-climetliy! 2,3,4J5S^JO511,12 J 3,14,15 J όJ 7-tetradecahydrø-lH cyclopcϊita[a]pb&naathren-3-yiox>'<;arboπylaτnij"5.o]-hexaπoyl |-pyτro1Idm~3-yl} ester AM
Figure imgf000042_0001
AH
Compound AG { 1 ,0 g, I .05 mmol) was mixed with succinic anhydride (0, 1 SO g, 1 ,5 mmα!) and DMAP (0.073 g, 0.6 mmol) and dried m a vacuuπi at 40°C ovemiglu. The mixture was dissolved in anhydrous dichloroethaiie (3 oil.), triethylanime (0.3 ! S g, 0.440 ml.., 3, LS mmol) was added ami ihe solution was stirred at room iemperature under argon atmosphere for 16 h. Il was then diluted with dichtoromothaae (40 ml) and washed wύh ice cold aqueous citric acid (5 wt%, 30 mL) and water (2 X 20 nit). The organic phase was dried over anhydrous sodsuπi sulfate and cojκeπtra(ed to dryness. The residue was used as such for s.he next step.
Cholesterol derivatised CPC r .
Figure imgf000042_0002
AI Succinate AH (0,254 g, 0.242 rnmol) was dissolved m a mixture of dichlorome-ihane/acfiJomtrHe s 3:2, 3 mi). To thai solution DMAP ((10296 g, 0.242 rranøt) in accioiύfxile (1.25 ml), 2,2"-Dithio-bIs(5-αitrop>τidiϊie} (0,075 g, 0.242 mmo!) in aceiotύtriie/diehloroethaαe (3: 1 , 1 ,25 ml.) were added successively. To the resulting solution iφheuyiphαsphrnε {0.064 g, 0,242 mrnol) in acetoiiitrile (0.6 nil} was added. The reaction mixture turned bright orange in color. 'The solution was agitated briefly using a wrist-acϋon shaker (S πiins}. Long chain aikyl amme--€PG (LCAA-CPG) i 1.5 g, (;1 -is Kf) was added, 'The suspension was agslated for 2 h. The CPG was iϋtered ihmugh a sintered tunnel and washed with ac≤tonitrile, dichbromeihane and ether successively. Unreaαeα amino groups were masked using acetic anhydride/pyridϊne. The achieved loading of the CPG was measured by taking UV measurement (37 mM/g).
The synthesis of si RNAa bearing a SM 2-dodecaπoie acid bisdecylamide- group (herein reierred to as "5!-C32-"} or a S'-ehαlesteτyl derivative group (herein referred to as "5!-Chol-"} v/as pcrfoππed as described ITΪ WO 2004/065601, except that, tor ihe ehoksiery! derivative, the oxidation step was performed using the Baaocage reagent in order IQ introduce a phosphorothioate linkage at the 5 '-end of the nucleic acid oligomer.
Nucleic acid sequences are represented beiow using standard nomenclature, and specifically the abbreviations of Table 1-2.
Table 1-2: Abbreviations of aucleotide ino»ORiers used m nucleic acid sequence representation. It will bs? understood that these monomers, whe» present m mi oligonucleotide, are aiatuaHy linked by S'-3'-ρhosphodiester bonds.
Figure imgf000043_0001
A 1
Figure imgf000044_0001
PCSK9 siRlMA screening in HuH7 , HepG2, Mela and Primary Monkey Hepafeocytes Discovers BHglϊiy Active S«q«e«ces
HuH-ZGeHs wefe obiained from JCRB CsH Bank {Japanese Collection of Research
Bioresources) (Shinjuku, lapas, cat. No.; JCRB041B) Ceils wes'e cultured in Dulbecco's MEM (Biochroπi AG, Berlin, Germany, cat No. P 0435) suppkmiemed to contain 10% feiaf calf serum (PCS) (Biochrorn AG, Berlin, Germany, cat. No. SOl 15), Penicillin 100 U/røi, Streptomycin 100 μg/m! CBioehrom ACK Berlin, Germany, cat. No. A2213) and 2mM L-Glmmm (Biochrom AG, Berlin, Germatiy, cat. No K02S2) at 370C in an atmosphere with 5% CCh m a humidifietf incubator (fieraeus HERAcclL fCendro Laboratory Produ.«s, !.,απgeriselboid, Germany). Hcp(}2 and HeIa cells were obtained from American Type Culture Collection (Rockviϋe, MD, cai. No, HB-SOo'5) and cultured in MEM ((5sbco Invitrogers, Karlsruhe, Germany, cat. No. 21090-022} supplemenled iα contain 10% fetal calf serum (FCS) (Biochrom AG, Berlin, Germany, cai. No, SOU S), Penicillin 100 U/ml> Streptomycin K)O μg/ml (Biochrom AG, Berlin, Germany, cat. No. A2213), I x Non Essential Amino Acids (Biochrom AG, Berlin. Germany, cat. No, K-0293), and 1Λ' ϊhi Sodium Pyruvate (Biochrom AG, Berlin, Germany, cat. No. 1,-0473 j at 3?:''C in an atmosphere with 5% CO.; m a humidified incubator {{-fcraeus jHERAceiL Kemiro Laboratory? Products. Lsngenseϊbokl Germany).
For transfeetiors with si RNA, HuH7, HepG2, or HeIa cells were seeded at a density of 2.0 x KT ceils/well in 96-svdI plates ami transfeeted directly. Transfeciion of siRNA (3OnM. for single dose screen) was carried oui with hpofeαamme 2iK)0 (ϊnvitrogen GmbH, Karlsruhe, Germany, cat. No. 11668-019} as described by the manimcturer.
24 hours after traπsieelbn HuH 7 and HepG2 ceils were jysed and PCSK9 mRNA levels were quantified with the Quantigeπe Explore Kit (Gsniαspreetra, Dumbarton Circle Fremotn, USA, cat No. QG-000-02) according to the protocol. PCSK9 mRNA levels were normalised io GAP-DH mRNA. For each siRNA eight individual datapoints were coHeeied, siRNA duplexes unrelated to PCSK9 gene were used as control. The activity of a given PCSK9 specific siRNA duplex was expressed us percent PCSK9 mRNA conceτuration iu treated ceils relative to F*ζ-SK9 mRNA concentration in cells «-eatεd with f.he control siRNA duplex.
Frimsr/ cyiiomoigus monkey hepatocytes (cryopreseπ'ed) were obtained from In vitro Technologies, Inc. (Baltimore, Maryland, USA, cat No M0030S) and cultured io InVitroGRO CP Medioin {cat No Z99029) at 3?°C in an atmosphere with 5% CO> in a iiumkϋfkd incubator.
For transection with siRNA, primaiy ey.nomolgys iBOiikey ceils were seeded on Collagen coated plates (Fisher Scientific, cat. Mo. OS-774-5) a^ a density of 3.5 x Uf ceHs/weH in 96-weH plates and trasisleeted directly. Transfection of siRNA (eight 2-lbld dikuion series starting (Vo«5 3OrM ) in duplicates was carried out with iipofectamine 2000 (ϊπvitvogcπ GmbH, Karismhe, Geπiiany, cat. No. 11668-019} as described by the manufacturer.
16 hours afler transfeottoD niediimi was changed to fresh InVϋroGRG C? Medium wkh 'Torpedo Antibiotic Mix (In vitro Technologies, ϊnc, cat. No Z99000) added.
24 hours after medium change primary cyoomolgus jrrønk'ey cells were Iyaed and PCSKS) mRNA levels were quantified with the Quaniigerse Eixpiore KiI (Geπosprectra, Dumbarton Circle Fremont, USA7 cat. No. Q€ -000-02) according to the protocol PCSK9 mRNA levels were normalized to GAPDH niRNA. Normalized PC'S KWGAFDH ratios were then compared to PCSK.9/GAPDH ratio of iipα fee Uu nine 2000 only control.
Tables 1-2 (and Figure (>) summarize the results and provides examples of m vitro screens in different ceil lines at different doses. Silencing of POSK.9 transenpi was expressed as percentage of remaining transcript at a given dose. Highly active sequences are those with less than 70% transcript remaining post treatment with a given siRN A at a dose less than or equal to } Oϋnrπ. Very active secμicnces are those thai have less than hϋ% of transcript remaining after treatment wiih a doseJess than or eqaa! Lo 1 DOnM Active sequences are those that have less man &5% transcript remaining after treatment wύ h a high dose { 10OnM ). Examples of active siRRVs were also screened in viw in mouse in lipidoid formulations as deseπbed below. Active sequences in vitro were also generally active m viva (See figure Figure 6 example}.
In ήvo Effleaey Sereeo of PC SK.9 siRNAs
Forrøuiadon Procedure
15 The iipidoid
Figure imgf000046_0001
PEG-Ceramide CIh (Avanϋ Polar Lipids) were used to prepare lipid-siRNA naτκiparticks Stock solutions of each in ethanol were prepared: LNP-Ol , \ 33 nig/mi.; Cholesterol, 25 rng/mL, PEG- Ceramide CIt\ !00 mg/'niL. LNP-OL Cholesteroi, and PEG-Ceramide Cl 6 stock solutions were then combined hi a 42:48:10 molar ratio. Combined lipid solution was rnixeα rapidly with
2.0 aqueous si RNA (in sodium ateintc pH 5} such that the final ethanol concentration was 35-45% and the final soάiurrs acciate concentration was 100-300 rsM, Upsci-siRNA naBoparticIes foππeϋ spontaneously upon mixing. Depending on the desired particle size distribution, the rosuliam ntmopanicle mixture was in sonic cases exirtidcd through a polycarbonate membrane (KKi nm cnt-ofi) using a iherniobarre! extruder (Lipex Extruder, Northern Lipids, Iπe). In other cases, the
25 extrusion step was omitted, Ethsnol removal and simnUaneous buffer excbas^ge was accomplished by either dialysis or tangential flow Duration. Bu Her was exchanged to phosphate buffered saline (PBS) pH 7,2. Qsaracterkatloss of ioruralsUons
Formulations prepared by cither the standard or extrusion-free method are characterized m a similar marker Formulations are first characterized by visual inspection. They should be S whitish translucent sohuions free from aggregates or sediment. Particle size and particle size distribution of hpid-rsanopsnides are measured by dynamic light scattering using a Maiveπi Zetasαer Nana ZS (Marvem, USA), Panicles should be 20-300 nmu and ideally, 40-100 nai in size. The particle size distribution should be unimodal. The total siRNA concentration in the .Cbπniύatωn, as well as ibe entrapped fraction, is estimated using a dye exclusion assay, A sample 0 of the formulated si RNA is incubated w-th the RNA~binding dye Ribogreen (Molecular Probes) in the presence or absence of a formulation disrupting surfactant, 0.5% Tr? ton- XI 00. The total siRNA ui the formulation is determined by die signal from the sample containing the surfactant, relative io a standard curve. The entrapped fraction is determined by subtracting the "free" siRNA content (as measured by the signal m the absence of surfactant) from the total siRNA 5 content. Percent entrapped si RNA i s typically >B 5 %.
Boles dosmg
BoI as dosing of formulated si RNAs in C57/BL6 mice f 5/group, 8~ 10 weeks old, Charles River Laboratories, IvIA) was pεrfbvrøed by iail vem iryecdon using a 27 Q needie. SiRNAs were formula led in LNP-O) (and then dialyzed against PBS) aL 0.5 mg/nii concentration allowing the 0 delivery of ihe 5mg/kg dose in 10 μl/g body weight. Mice were kept under an iπfraxed lamp for approximately 3 mhi prior to dosing to ease injection,
48 hour posi dosing mice were sacrificed by COj-aBphyxsutior!, 0.2 m\ blood was eoileαed by retro-orbital bleed ing and the lives- was harvested and frozen in liquid nitrogers. Serum and livers were stored at - SO0C".
5 Frozen livers were gnnded using 6850 Freezer/Mill Cryogenic Grinder (SPEX
CentriPrep, inc) and powders stored at -SO0C until analysis. POSK.9 mRNA levels were defected using the branched-DNΛ technology based kit from Quantiϋene Reagent System (Gesiospectra) according to the protocol. 10-2ϋrag of frozen liver powders was iysed m 600 u) of 0.16 αg/rnl Proteinase K (Epicentre, #MFRK092) in Tissue and Cell Lysis Solution (Epicentre. #MΪC0%.H} at 6ST for 3 hours. Then 10 ul of the iysates were added to 90ul of Lysis Working Reagent (1 volume of stock Lysis Mixture in two volumes of water) and incubated at 52"C overnight on Genospectra capture plates with probe sets specific to mouse PCSKi) and mouse GAFDH or cyclophϋin B. Nucleic acid sequences lor Capture Extender (CE), Label Extender (LE) and blocking (BL) probes were selected from the nucleic acid sequences of PCSK9. GAPDH and cyclophilϊn B with the help of the QuaπtiGene ProbeDesigπer Software 2.0 (Genospectra, Fremont, CA, USA, cat. No. QG-002-02). Chemo luminescence was read on a V*ctor2-Light (Perkin. Elmer) as Relative light units. The ratio of PCSK1) m RN A 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 grøiφ treated with an unrelated siRNA (hiood coagulation factor VII)1
Total seru.rn cholesterol in mouse serum was measured using the SumBki Cholesterol LiquiColor kit (StanBio Laboratoriy, Boeme, Texas, USA) according to manufacturer's instructions. Measurements were taken on a Victor2 1420 Multi label Counter {Perkin Elmer) at
495 nm.
Exao;i|?les
32 PCSK.9 siRNAs formulated in LNP-01 liposomes were tested in vivo m a mouse model. The experiment was performed at 3mg/kg siRNA dose and at least 10 PGSK9 siRNAs showed more than. 40% PCSK9 mRNA knock down compared to a control group treated with FBS, while control group treated with an unrelated siRNA (blood coagulation factor VlI) had no eii'ect (Figures 2-5), Silencing of PCSK9 transcript also eoorelated with a lowering of cholesterol in these animals (Figures 4-5). In addition ihers? was a strong coorslatson between those molecules that were active in vitro mid those active in vivo (Figure 6). Sequences containing different chemical modsfieaiions were also screened in vitro (Tables I and 2) and m vivo. As an example, less modified sequences 9314 and 9318, and a more modified versions of that sequence 9314-(10792^ 10793, and 10796); 93 IS-(10794, 10795. 10797} were tested both in vitro (in primary monkey hepaiocytes) or in vivo (9314 and 10792} formulated is* LNP-O! > Figure 7 (also see Tables 1 and2) shows that the parent molecules 9314 aαd 93 IS and the modified versions arc all active in vitro. Figure 8 as an example shows that both the parent 9314 and the more highly modified 1.0792 sequences are active m vivo displaying 50-60% silencing of endogenous PCSK9 m -mice. Figure 9 furthur exemplifies; that activity of other chemically modified versions of the parents 9314 and 10792.
dsRNA expression vectors
In another aspect of the invention. PCS&9 specific dsRNA molecules thai modulate PCSK9 gene expression activity are expressed from transc.ript.ion units inserted into DMA or RNA vectors (see, e.g.. Couture, A, et al., TIG. (19%), 12:5-10; Skiiiern, A., ei si,. International PCT Publication No. WO 00/221 13, Conrad, international PCT Publication No. WO 00/221 R and Conrad, US Pat. No. 6.054,299). These transgenes can be introduced as a linear construct, a circular p lasπi id, or tx viral vector, which can be incorporated and inherited as a tnmsgerse miegrated irsio the host genome. The trsosgeue crø a^BO bs constructed to permit it to be inherited as an extracbromosomal plasmid (Gassmann, et al.,
Figure imgf000049_0001
NutL Acad, Sd. USA (S V95.I 92: 1292 K
The individual strands of a dsRNA can be transcribed by promoters on two separate expression vectors and εo-tπmsfbcted into a target ceil. Alternatively each individual strand of ihe dsRKA cars bs transcribed by promoters both of which are located on the same expression plasπiid. In a preferred embodiment, a dsRNA is expressed as an inverted repeat joined by a linker polynucleotide sequence such thai the dsRNA has a stem and loop structure.
The recombinant dsRNA expression vectors are generally DNA plasm ids or viral vectors. dsRNA expressing viral vectors can be constructed based on, but ml limited to, adeno- associated virus (for a review, see Mazyczka, et ai.. Cum Topics Micro. Immunol (1992} !S8:97-I29)); adenovirus (see, for example, Berkner, et al., BioTecbriiqoes (1998) 6:616), koserrfsfd et a!. { 1991 , Science 252:431 -434), and Rosenfεld et a!, (1992), Ceil 68:143-155}); or slphavirus 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 celis. in vnro and/or m vivo (see., e.g., Egiitis, ei al.. Science ( 1983) 238; 1395~!39S; Danos and Mulligan, Prσc, NaU. Acad Sd USA (1998) 85:6460-6464; Wilson et aL, 1988, Proc. Natl. Acad. Sci. CiSA $5:3014-3018; Aπneπtano et al.,
1990, Piχ>c. NaIi Acad. SeL USA 87;6f 416145; Ruber et ah, mL Free, NaU. Acad. Sci. USA 88:8039-8043; Perry et ai., 1991 , Proc. NaJl. Acad Sci. USA $8:8377-8381 ; Ohβwdhuiy et aL
1991, Science 254: 1802-1805; van Bcuβecbem. el HL 1992, Proc. Nad. Acad. Sci. USA 89:7640- 19 ; Kay et al., 1992, Human Gene Therapy 3:641-047; Dai ef al, 1992, Proc. Natl. Acad. Set USA 89: 10892-10895; Hwu ei al., 1993, 1. Immunol 150:4104-411 5; US. Patent No. 4,868,116; U.S. Patent No. 4,980,286; PCX Application WO 89/07136; PCT Application WO S9/0246S; PCT Application WO 89/05345; and PCT Application WO 92/07573). Recombinant retroviral vectors capable of transducing and expressing genes inserted into ths genome of a cell can be produced by transfecϋng the recombinant retroviral genome into suitable packaging eel! lines such as P A3! 7 and Psi-CRIF (Cometie et aL, 1991 , Hum&n 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 mitoticairy active ceils for infection.
The promoter driving dsRNA expression in either a DNA pUismid or viral vector of the invention may be a eαkaryodc RNA polymerase I (e.g. rihosoraa! RKA promoter), RNA polymerase 0 (e.g. CMV early promoter or actin promoter or Ul snRN A 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 pl&smid also encodes T7 RNA polymerase required for transcription from a T? promoter. The promoter can also direct iransgene expression to the pancreas (see, e.g. the insulin regulatory sequence for pancreas (Bucchini et aL, 1986, Proc. Natl Acad, Sci. USA 83:251 1 -2515}}.
In addition, expression of the transgεrse can be precisely regulated, for example, by using ars inducible regulatory sequence and expression systems such as a reguhnory sequence that is sensitive to certain physiologies! regulators, e.g., circulating glucose levels, or hormones (Docherty el &L 1994, FASEB J, 8:20-24). Such inducible expression systems, suitable for the control of transgenε expression in cells or in. mammals include regulation by ecdysone, by estrogen, progesterone, tetracycline, chemical inducers of dimerizatiors. and isopropyl-beta-Dl - ihiogaiactopyranoside (H-PTCiJ). A person skilled in the art would be able to choose die 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 beknv, and persist, in target cdls. Alternatively, viral vectors can be used that provide for transient expression of dsSlNA molecules. Such vectors can be repeatedly administered as necessary. Once expressed, the dsRNΛs bind to target RNA and modulate its function or e&pression, Delivery of dsRNA expressing vectors cars be systemic such as by intravenous or intramuscular administration, by adnivnisrration to large! cells ex-planted from the patient followed by reratroductkni into the patient, or by any other means that allows for introduction into ά - desired target cell,
dsRMA expression DNA plasnrids are typically trail steeled imo target cells as a complex with cationic lipid carriers (eg, Oiigofεetamiαe} or non-cationie lipid -based carriers (e.g. Transit- TKO v:), Multiple lipid transiections for dsRNA-mediated knockdowns iargeiing different regions of a single PCSK9 gene or multiple P€SK.9 genes ov«r a period of a week or more are also contemplated by the invention. Successful introduction of the vectors of the invention mio host cells can be monitored using various known methods, For example, transient irαπsfectioπ. can be signaled with a reporter, such as a fluorescent marker, such as Green Fluorescent Protein S GFP). Stable traπsfection. of ex vivo cells can be ensured using markers {bat provide the transfected cell with resistance to specific environmental factors {e.g., antibiotics and drugs), such as hygromycirs B resistance.
The PCSK9 specific- dsENA 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 5328,470) or by stereotactic injection, (see e.g., Chen et ai. ( 1994) Proc. Natl Acad. Sci. USA 91:3054-3057). The pharmaceutical preparation of Die gene therapy vector can include rhe gene therapy vector in an acceptable dikie.nL or can comprise a slow release matrix hi 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 mstarn disclosure which will allow them Io practice Qi is invention to the full scope of the claims hereinafter appended.
IO
15
}<}
Figure imgf000053_0001
Figure imgf000054_0001
Figure imgf000055_0001
Figure imgf000056_0001
54
Figure imgf000057_0001
55
Figure imgf000058_0001

Figure imgf000059_0001
Figure imgf000060_0001
SS
Figure imgf000061_0001
Figure imgf000062_0001
60
Figure imgf000063_0001
(>l
Figure imgf000064_0001
62
Figure imgf000065_0001
Figure imgf000066_0001
Figure imgf000067_0001
Figure imgf000068_0001
66
Figure imgf000069_0001
Figure imgf000070_0001
Figure imgf000071_0001
Figure imgf000072_0001
/IJ
Figure imgf000073_0001
71
Figure imgf000074_0001
Figure imgf000075_0001
Figure imgf000076_0001
Figure imgf000077_0001
Figure imgf000078_0001
Figure imgf000079_0001
Figure imgf000080_0001

Figure imgf000081_0001
Figure imgf000082_0001
Si?
Figure imgf000083_0001
Figure imgf000084_0001
Tabic
1H
Figure imgf000085_0001
Figure imgf000086_0001
Figure imgf000087_0001
Figure imgf000088_0001
86 connected by 3M>~5'-<9phosphorothiodiester groups; unless denoted by prefix "p-", oligonucleotides are devoid of a S '-phosphate group on the 5'-most nucleotide; ail oligonucleotides beat 3* -OH on the 3 '-most nucleotide

Claims

A double-stranded ribonucleic acid (dsRNAj for inhibiting the expression of a human PCSK^ gene in a cell wherein said dsRNA comprises at least two sequences that are covnple?ne.mary to each other and wherein a sense strand comprises a Orsi sequence and an ami sense strand comprises a second sequence comprising a region of complementarity which is substantially complementary to ai least a pan of a rnRNA encoding PCSK9, and wherein said region of complementarity is less than 30 nucleotides in length and wherein said dsRNA, upon contact with a ceil expressing said PCSK9, inhibits expression of said PCS K9 gene.
The (JsRNA of claim L wherein said first sequence is selected from the group consisting of Tables i and 2 and said second sequence is selected from the group consisting of Tables 1 and 2.
, The dsRNA of claim 1 , wherein said dsRNA comprises at least one rrsoenf sed niϊc!eofκ!c.
, The dsRNA of claim 2, wherein said dsRNA comprises at least one modi ied nucleotide.
, The dsRN'A of claim 3, wherein said modified nucleotide is chosen &om he uroiw of; 2'-O-meϋiyl modified nucleotide, a nucleotide comprising a 5!-ρhosρhoro" moaie group, and a terminal nucleotide linked to a cholustεryi derivative or dodecanoic acid bisdeeylamidε group.
The dsRNA of claim 3, wherein said modified nucleotide is chosen from the group of; a 2t--deoxy-2<-fhιøro modified nucleotide, a 21~deo.xy~nκxlilied nucleotide, & locked nucleotide, an abaaic nucleotide, 2'-amino-modified nucleotide, ^'-alkyi-modifted nucleotide, morpholino nucleotide, a
Figure imgf000090_0001
and a non-naturai base comprising nucleotide.
7. The dsJINA of claim 3, wherein said first sequence is selected from (he group consisting of Tables 1 ami 2 and said second sequence is selected from the group consisting of Tables 1 and 2
S. The dsRNA of claim 6, wherein said first sequence is selected from the group consisting of Tables I and 2, and said second sequence is selected from the group consisting of Tables 1 and 2.
9, A eel! comprising the dsRNA of claim 1 .
10, A pharmaceutical composition for inhibiting the expression of the PCSK9 gene in an organism, composing a dsRNA and a pharmaceutically acceptable carrier, wherein the dsRNA comprises at least, two sequences that are complementary to each other and wherein a sense strand comprises a first sequence and an an fi sense strand comprises a second sequence comprising a region of complementarity which is substantially complementary to at least a part of a mRNA encoding PCSK9, and wherein said region of complementarity is less ih&n 30 nucleotides in length and wherein said dsfi'NA, upon contact wnh a cell expressing said P€SK.9, inhibits expression of said PCSK.9 gene.
1 1 , The pharmaceutical composition of claim 10, wherein said first sequence of said dsRNA is selectee! from the group consisting of Tables 1 mά 2, and said second sequence of said dsRNA is selected from the group consisting of Tables 1 and 2,
12, The pharmaceutical composition of claim 10, wherein said first sequence of said dsRNA is selected from the group consisting of Tables 1 and 2 and said second sequence of said dsRNA is selected Ik) at tire group consisting of Tables 1 and 2.
13, A method for inhibiting the expression of the FCSK9 gene m a cell, die method
"ompπsmg;
(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 and wherein a sense strand comprises a first sequence and an antisense strand comprises a second sequence comprising a region of conrpiementarity which is snbstanlisily complementary to at least a part of a mϊlNA encoding PCSK.9, and wherein said region of complementarity is less than 30 nucleotides in length and wherein said dsRNA, upon contact with a cell expressing said PCSKO, inhibits expression of said PCSK9 gens; and
(b) maintaining the eel! produced in step in) lor a lime sufficient to obtain degradation of the rnRNA transcript of the PCSK 9 gene, thereby inhibiting expression of the POSX.9 gene in the cell.
14, A method of treating, preventing or managing pathological processes which can be mediated by down regulating PCSK.9 gene expression comprising administering io a patient in need of such treatment, prevention or management a therapeutically or prαpnyiactieaily effective amount of a dsRNA, wherein the dsRNA comprises at least two sequences that are complementary io each other and wherein a. sense strand comprises a tlrst sequence and sn antisense strand comprises a second sequence comprising a region of complementarity winch is substantially complementary to at least a part of a rnRNA encoding PCSK.9, and wherein said region of complementarity is less than 30 nucleotides in length and wherein said dsRNA, upon contact with a cell expressing said PCSK9, inhibits expression of said PCSK.9 gene.
\ S. A vector tor inhibiting the expression of the PCSK9 gene in a cell, said vector comprising a regulatory sequence operabϊy linked to a nucleotide sequence thai encodes at least one strand of a άsRNA. wherein one of the strands of sa.id άsRNA is substantially complementary to at least a pan of a niRNA encoding PCSK9 and wherein said dsRNA is less than 30 base pairs in length am! wherein said dsRNA, upon contact with a coll expressing said PCSK9\ inhibits the expression of said PCSK9 gene.
16. A cell comprising the Yector of claim 15.
17. A doubk-stxanded ribonucleic acid fdsRNΛ) for reducing the expression level of a human PCSK9 gens in a cell, wherein said dsRNA comprises at bast two sequences that are complementary to each other and wherein a sense strand comprises a first sequence and an amisense strand comprises a second sequence comprising a region of complementarity which is substantially complementary to at least a par* of a πiRNA encoding PCSK.9, and wherein said dsRNA, upon contact with a cell expressing said PCSK9, reduces the expression level of said PCSK^ gene.
. The tlsR'NA ordain? 17, wherein said contact reduces the expression level of said PCSKS
, The dsllNA of claim 17, wherein said contact Is performed in vitro al 30 oM or less.
, A pharmaceutical composition for reducing the expression level of the PCSK9 gene in an organise comprising the dsRNA of claim i 7 arsd a pharmaceutically acceptable CEsitier,
, A method of treating a PCSK9 associated disorder comprising administering to a patient m need of such treatment, a therapeutically effective amount of a dsRNA of claim 17.
, A method of treating a PCS K9~associated disorder
Figure imgf000093_0001
administering to a patient m natά of such treatment, a therapeutically effective amount of a dsRI^A of claim 1 7.
PCT/US2007/068655 2006-05-11 2007-05-10 Compositions and methods for inhibiting expression of the pcsk9 gene WO2007134161A2 (en)

Priority Applications (16)

Application Number Priority Date Filing Date Title
NZ572666A NZ572666A (en) 2006-05-11 2007-05-10 Compositions comprising double stranded rna and methods for inhibiting expression of the pcsk9 gene
EA200870528A EA015676B1 (en) 2006-05-11 2007-05-10 Compositions and methods for inhibiting expression of the pcsk9 gene
KR1020107013424A KR101320916B1 (en) 2006-05-11 2007-05-10 Compositions and methods for inhibiting expression of the PCSK9 gene
CA2651839A CA2651839C (en) 2006-05-11 2007-05-10 Compositions and methods for inhibiting expression of the pcsk9 gene
EP17162101.4A EP3249052B1 (en) 2006-05-11 2007-05-10 Compositions and methods for inhibiting expression of the pcsk9 gene
CN2007800248541A CN101484588B (en) 2006-05-11 2007-05-10 Compositions and methods for inhibiting expression of the pcsk9 gene
EP19168142.8A EP3578656B1 (en) 2006-05-11 2007-05-10 Compositions and methods for inhibiting expression of the pcsk9 gene
EP21150171.3A EP3872179A1 (en) 2006-05-11 2007-05-10 Compositions and methods for inhibiting expression of the pcsk9 gene
AU2007249329A AU2007249329C1 (en) 2006-05-11 2007-05-10 Compositions and methods for inhibiting expression of the PCSK9 gene
PL19168142T PL3578656T3 (en) 2006-05-11 2007-05-10 Compositions and methods for inhibiting expression of the pcsk9 gene
EP07762085A EP2021507A4 (en) 2006-05-11 2007-05-10 Compositions and methods for inhibiting expression of the pcsk9 gene
JP2009510173A JP5570806B2 (en) 2006-05-11 2007-05-10 Compositions and methods for inhibiting the expression of the PCSK9 gene
IL195181A IL195181A (en) 2006-05-11 2008-11-09 Compositions for inhibiting the expression of the pcsk9 gene
AU2010241357A AU2010241357B2 (en) 2006-05-11 2010-11-09 Compositions and methods for inhibiting expression of the PCSK9 gene
IL220102A IL220102A (en) 2006-05-11 2012-05-31 Compositions and methods for inhibiting expression of the pcsk9 gene
IL250678A IL250678A0 (en) 2006-05-11 2017-02-20 Compositions and methods for inhibiting expression of the pcsk9 gene

Applications Claiming Priority (10)

Application Number Priority Date Filing Date Title
US79945806P 2006-05-11 2006-05-11
US60/799,458 2006-05-11
US81720306P 2006-06-27 2006-06-27
US60/817,203 2006-06-27
US84008906P 2006-08-25 2006-08-25
US60/840,089 2006-08-25
US82991406P 2006-10-18 2006-10-18
US60/829,914 2006-10-18
US90113407P 2007-02-13 2007-02-13
US60/901,134 2007-02-13

Publications (3)

Publication Number Publication Date
WO2007134161A2 true WO2007134161A2 (en) 2007-11-22
WO2007134161A3 WO2007134161A3 (en) 2008-02-21
WO2007134161A8 WO2007134161A8 (en) 2009-05-07

Family

ID=38694697

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2007/068655 WO2007134161A2 (en) 2006-05-11 2007-05-10 Compositions and methods for inhibiting expression of the pcsk9 gene

Country Status (15)

Country Link
US (9) US7605251B2 (en)
EP (8) EP2584048B1 (en)
JP (4) JP5570806B2 (en)
KR (2) KR101320916B1 (en)
CN (2) CN103614375A (en)
AU (2) AU2007249329C1 (en)
CA (2) CA2915441A1 (en)
EA (2) EA015676B1 (en)
ES (2) ES2392478T3 (en)
HK (1) HK1183910A1 (en)
IL (3) IL195181A (en)
NZ (2) NZ572666A (en)
PL (2) PL3578656T3 (en)
SG (1) SG171676A1 (en)
WO (1) WO2007134161A2 (en)

Cited By (39)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009134487A3 (en) * 2008-01-31 2010-02-04 Alnylam Pharmaceuticals, Inc. Optimized methods for delivery of dsrna targeting the pcsk9 gene
JP2010510807A (en) * 2006-11-27 2010-04-08 アイシス ファーマシューティカルズ, インコーポレーテッド Methods for treating hypercholesterolemia
EP2268316A2 (en) * 2008-03-20 2011-01-05 Quark Pharmaceuticals, Inc. NOVEL siRNA COMPOUNDS FOR INHIBITING RTP801
US8188233B2 (en) 2008-02-07 2012-05-29 Merck Sharp & Dohme Corp. 1B20 PCSK9 antagonists
US8188234B2 (en) 2008-02-07 2012-05-29 Merck Sharp & Dohme Corp. 1D05 PCSK9 antagonists
US8222222B2 (en) 2006-05-11 2012-07-17 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of the PCSK9 gene
US8273869B2 (en) 2009-06-15 2012-09-25 Alnylam Pharmaceuticals, Inc. Lipid formulated dsRNA targeting the PCSK9 gene
WO2012145729A2 (en) 2011-04-20 2012-10-26 Smith Holdings, Llc Methods and compositions for modulating gene expression using components that self assemble in cells and produce rnai activity
US8344114B2 (en) 2006-11-07 2013-01-01 Merck Sharp & Dohme Corp. Antagonists of PCSK9
WO2014182661A2 (en) 2013-05-06 2014-11-13 Alnylam Pharmaceuticals, Inc Dosages and methods for delivering lipid formulated nucleic acid molecules
US9051567B2 (en) 2009-06-15 2015-06-09 Tekmira Pharmaceuticals Corporation Methods for increasing efficacy of lipid formulated siRNA
US9187746B2 (en) 2009-09-22 2015-11-17 Alnylam Pharmaceuticals, Inc. Dual targeting siRNA agents
US9200276B2 (en) 2009-06-01 2015-12-01 Halo-Bio Rnai Therapeutics, Inc. Polynucleotides for multivalent RNA interference, compositions and methods of use thereof
EP3252068A2 (en) 2009-10-12 2017-12-06 Larry J. Smith Methods and compositions for modulating gene expression using oligonucleotide based drugs administered in vivo or in vitro
US10494621B2 (en) 2015-06-18 2019-12-03 The Broad Institute, Inc. Crispr enzyme mutations reducing off-target effects
US10550372B2 (en) 2013-12-12 2020-02-04 The Broad Institute, Inc. Systems, methods and compositions for sequence manipulation with optimized functional CRISPR-Cas systems
US10577630B2 (en) 2013-06-17 2020-03-03 The Broad Institute, Inc. Delivery and use of the CRISPR-Cas systems, vectors and compositions for hepatic targeting and therapy
US10696986B2 (en) 2014-12-12 2020-06-30 The Board Institute, Inc. Protected guide RNAS (PGRNAS)
US10711285B2 (en) 2013-06-17 2020-07-14 The Broad Institute, Inc. Optimized CRISPR-Cas double nickase systems, methods and compositions for sequence manipulation
US10731157B2 (en) 2015-08-24 2020-08-04 Halo-Bio Rnai Therapeutics, Inc. Polynucleotide nanoparticles for the modulation of gene expression and uses thereof
US10781444B2 (en) 2013-06-17 2020-09-22 The Broad Institute, Inc. Functional genomics using CRISPR-Cas systems, compositions, methods, screens and applications thereof
US10851357B2 (en) 2013-12-12 2020-12-01 The Broad Institute, Inc. Compositions and methods of use of CRISPR-Cas systems in nucleotide repeat disorders
EP3677679A4 (en) * 2017-12-26 2020-12-02 Guangzhou Ribobio Co., Ltd. Sirna molecule inhibiting pcsk9 gene expression and application thereof
US10930367B2 (en) 2012-12-12 2021-02-23 The Broad Institute, Inc. Methods, models, systems, and apparatus for identifying target sequences for Cas enzymes or CRISPR-Cas systems for target sequences and conveying results thereof
US10946108B2 (en) 2013-06-17 2021-03-16 The Broad Institute, Inc. Delivery, use and therapeutic applications of the CRISPR-Cas systems and compositions for targeting disorders and diseases using viral components
US11008588B2 (en) 2013-06-17 2021-05-18 The Broad Institute, Inc. Delivery, engineering and optimization of tandem guide systems, methods and compositions for sequence manipulation
US11041173B2 (en) 2012-12-12 2021-06-22 The Broad Institute, Inc. Delivery, engineering and optimization of systems, methods and compositions for sequence manipulation and therapeutic applications
US11045488B2 (en) 2014-12-26 2021-06-29 Nitto Denko Corporation RNA interference agents for GST-π gene modulation
US11155795B2 (en) 2013-12-12 2021-10-26 The Broad Institute, Inc. CRISPR-Cas systems, crystal structure and uses thereof
EP4035659A1 (en) 2016-11-29 2022-08-03 PureTech LYT, Inc. Exosomes for delivery of therapeutic agents
US11407985B2 (en) 2013-12-12 2022-08-09 The Broad Institute, Inc. Delivery, use and therapeutic applications of the CRISPR-Cas systems and compositions for genome editing
US11492620B2 (en) 2017-12-01 2022-11-08 Suzhou Ribo Life Science Co., Ltd. Double-stranded oligonucleotide, composition and conjugate comprising double-stranded oligonucleotide, preparation method thereof and use thereof
US11578312B2 (en) 2015-06-18 2023-02-14 The Broad Institute Inc. Engineering and optimization of systems, methods, enzymes and guide scaffolds of CAS9 orthologs and variants for sequence manipulation
WO2023017004A1 (en) * 2021-08-09 2023-02-16 Cargene Therapeutics Pte. Ltd. Inhibitory nucleic acids for pcsk9
USRE49431E1 (en) 2014-12-26 2023-02-28 Nitto Denko Corporation RNA interference agents for GST-PI gene modulation
US11633482B2 (en) 2017-12-29 2023-04-25 Suzhou Ribo Life Science Co., Ltd. Conjugates and preparation and use thereof
US11660347B2 (en) 2017-12-01 2023-05-30 Suzhou Ribo Life Science Co., Ltd. Nucleic acid, composition and conjugate containing same, preparation method, and use thereof
US11896674B2 (en) 2018-09-30 2024-02-13 Suzhou Ribo Life Science Co., Ltd. SiRNA conjugate, preparation method therefor and use thereof
US11918600B2 (en) 2018-08-21 2024-03-05 Suzhou Ribo Life Science Co., Ltd. Nucleic acid, pharmaceutical composition and conjugate containing nucleic acid, and use thereof

Families Citing this family (118)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20070173473A1 (en) * 2001-05-18 2007-07-26 Sirna Therapeutics, Inc. RNA interference mediated inhibition of proprotein convertase subtilisin Kexin 9 (PCSK9) gene expression using short interfering nucleic acid (siNA)
EP3037105B1 (en) * 2003-06-27 2021-03-17 Amgen Fremont Inc. Antibodies directed to the deletion mutants of epidermal growth factor receptor and uses thereof
CN101437933B (en) 2005-12-28 2013-11-06 斯克里普斯研究所 Natural antisense and non-coding RNA transcripts as drug targets
EP2548438B1 (en) * 2006-11-08 2015-08-05 Veritas Bio, LLC In vivo delivery of double stranded RNA to a target cell
US8093222B2 (en) 2006-11-27 2012-01-10 Isis Pharmaceuticals, Inc. Methods for treating hypercholesterolemia
US7910722B2 (en) 2007-07-05 2011-03-22 Florida State University Research Foundation RNAi therapeutic for treatment of hepatitis C infection
JOP20080381B1 (en) 2007-08-23 2023-03-28 Amgen Inc Antigen Binding Proteins to Proprotein Convertase subtillisin Kexin type 9 (pcsk9)
US10131904B2 (en) 2008-02-11 2018-11-20 Rxi Pharmaceuticals Corporation Modified RNAi polynucleotides and uses thereof
JP2012502991A (en) 2008-09-22 2012-02-02 アールエックスアイ ファーマシューティカルズ コーポレーション RNA interference in dermal applications
JP6128732B2 (en) 2008-10-03 2017-05-17 クルナ・インコーポレーテッド Treatment of apolipoprotein-A1 related diseases by suppression of natural antisense transcripts against apolipoprotein-A1
ES2637063T3 (en) 2008-12-04 2017-10-10 Curna, Inc. Treatment of diseases related to tumor suppressor genes by inhibiting the natural antisense transcript to the gene
WO2010065792A2 (en) 2008-12-04 2010-06-10 Curna, Inc. Treatment of erythropoietin (epo) related diseases by inhibition of natural antisense transcript to epo
JP5971948B2 (en) 2008-12-04 2016-08-17 クルナ・インコーポレーテッド Treatment of vascular endothelial growth factor (VEGF) -related diseases by suppression of natural antisense transcripts against VEGF
US9493774B2 (en) 2009-01-05 2016-11-15 Rxi Pharmaceuticals Corporation Inhibition of PCSK9 through RNAi
WO2010090762A1 (en) 2009-02-04 2010-08-12 Rxi Pharmaceuticals Corporation Rna duplexes with single stranded phosphorothioate nucleotide regions for additional functionality
PL2396038T3 (en) 2009-02-12 2016-05-31 Curna Inc Treatment of brain derived neurotrophic factor (bdnf) related diseases by inhibition of natural antisense transcript to bdnf
US9464287B2 (en) 2009-03-16 2016-10-11 Curna, Inc. Treatment of nuclear factor (erythroid-derived 2)-like 2 (NRF2) related diseases by inhibition of natural antisense transcript to NRF2
CA2755404C (en) 2009-03-17 2020-03-24 Joseph Collard Treatment of delta-like 1 homolog (dlk1) related diseases by inhibition of natural antisense transcript to dlk1
CN106237345A (en) 2009-05-06 2016-12-21 库尔纳公司 By suppression therapy lipid transfer and the metabolic gene relevant disease of the natural antisense transcript for lipid transfer and metabolic gene
ES2609655T3 (en) 2009-05-06 2017-04-21 Curna, Inc. Treatment of diseases related to tristetraproline (TTP) by inhibition of natural antisense transcript for TTP
ES2618572T3 (en) 2009-05-08 2017-06-21 Curna, Inc. Treatment of diseases related to the dystrophin family by inhibiting a natural antisense transcript for the dmd family
JP5922017B2 (en) 2009-05-18 2016-05-24 クルナ・インコーポレーテッド Treatment of reprogramming factor-related diseases by suppression of natural antisense transcripts against the reprogramming factor
WO2010135695A2 (en) 2009-05-22 2010-11-25 Curna, Inc. TREATMENT OF TRANSCRIPTION FACTOR E3 (TFE3) and INSULIN RECEPTOR SUBSTRATE 2 (IRS2) RELATED DISEASES BY INHIBITION OF NATURAL ANTISENSE TRANSCRIPT TO TFE3
EP2435571B1 (en) 2009-05-28 2016-12-14 CuRNA, Inc. Treatment of antiviral gene related diseases by inhibition of natural antisense transcript to an antiviral gene
ES2629339T3 (en) 2009-06-16 2017-08-08 Curna, Inc. Treatment of diseases related to paraoxonase 1 (pon1) by inhibition of natural antisense transcript to pon1
KR101801404B1 (en) 2009-06-16 2017-12-20 큐알엔에이, 인크. Treatment of collagen gene related diseases by inhibition of natural antisense transcript to a collagen gene
CA2765889A1 (en) 2009-06-24 2010-12-29 Opko Curna, Llc Treatment of tumor necrosis factor receptor 2 (tnfr2) related diseases by inhibition of natural antisense transcript to tnfr2
KR101807324B1 (en) 2009-06-26 2017-12-08 큐알엔에이, 인크. Treatment of down syndrome gene related diseases by inhibition of natural antisense transcript to a down syndrome gene
US8283333B2 (en) 2009-07-01 2012-10-09 Protiva Biotherapeutics, Inc. Lipid formulations for nucleic acid delivery
US8563528B2 (en) 2009-07-21 2013-10-22 Santaris Pharma A/S Antisense oligomers targeting PCSK9
CN102712925B (en) 2009-07-24 2017-10-27 库尔纳公司 It is diseases related that SIRTUIN (SIRT) is treated by suppressing SIRTUIN (SIRT) natural antisense transcript
CA2769665A1 (en) 2009-08-05 2011-02-10 Opko Curna, Llc Treatment of insulin gene (ins) related diseases by inhibition of natural antisense transcript to an insulin gene (ins)
EP2464731B1 (en) 2009-08-11 2016-10-05 CuRNA, Inc. Treatment of adiponectin (adipoq) related diseases by inhibition of natural antisense transcript to an adiponectin (adipoq)
WO2011022606A2 (en) 2009-08-21 2011-02-24 Curna, Inc. Treatment of 'c terminus of hsp70-interacting protein' (chip) related diseases by inhibition of natural antisense transcript to chip
KR101892760B1 (en) 2009-08-25 2018-08-28 큐알엔에이, 인크. Treatment of 'iq motif containing gtpase activating protein' (iqgap) related diseases by inhibition of natural antisense transcript to iqgap
WO2011028938A1 (en) * 2009-09-02 2011-03-10 Alnylam Pharmaceuticals, Inc. Methods for lowering serum cholestrol in a subject using inhibition of pcsk9
CN102791861B (en) 2009-09-25 2018-08-07 库尔纳公司 FLG relevant diseases are treated by adjusting expression and the activity of Filaggrin (FLG)
AR079336A1 (en) * 2009-12-11 2012-01-18 Irm Llc ANTAGONISTS OF THE PRO-PROTEIN CONVERTASE-SUBTILISINE / TYPE 9 QUEXINE (PCSK9)
ES2661813T3 (en) 2009-12-16 2018-04-04 Curna, Inc. Treatment of diseases related to membrane transcription factor peptidase, site 1 (mbtps1) by inhibition of the natural antisense transcript to the mbtps1 gene
US20110152343A1 (en) * 2009-12-22 2011-06-23 Functional Genetics, Inc. Protease inhibitors and broad-spectrum antiviral
EP2515947B1 (en) 2009-12-23 2021-10-06 CuRNA, Inc. Treatment of uncoupling protein 2 (ucp2) related diseases by inhibition of natural antisense transcript to ucp2
WO2011079261A2 (en) 2009-12-23 2011-06-30 Curna, Inc. Treatment of hepatocyte growth factor (hgf) related diseases by inhibition of natural antisense transcript to hgf
RU2615450C2 (en) 2009-12-29 2017-04-04 Курна, Инк. Treating diseases associated with nuclear respiratory factor 1 (nrf1) by inhibition of natural antisense transcript to nrf1
JP5982288B2 (en) 2009-12-29 2016-08-31 カッパーアールエヌエー,インコーポレイテッド Treatment of tumor protein 63-related diseases by inhibition of natural antisense transcripts against tumor protein 63 (p63)
EP2519632B1 (en) 2009-12-31 2018-04-11 CuRNA, Inc. Treatment of insulin receptor substrate 2 (irs2) related diseases by inhibition of natural antisense transcript to irs2 and transcription factor e3 (tfe3)
DK2521784T3 (en) 2010-01-04 2018-03-12 Curna Inc TREATMENT OF INTERFERON REGULATORY FACTOR 8- (IRF8) RELATED DISEASES BY INHIBITION OF NATURAL ANTISENCE TRANSCRIPT TO IRF8
JP5963680B2 (en) 2010-01-06 2016-08-03 カッパーアールエヌエー,インコーポレイテッド Treatment of pancreatic developmental gene diseases by inhibition of natural antisense transcripts against pancreatic developmental genes
WO2011085347A2 (en) 2010-01-11 2011-07-14 Opko Curna, Llc Treatment of sex hormone binding globulin (shbg) related diseases by inhibition of natural antisense transcript to shbg
ES2671877T3 (en) 2010-01-25 2018-06-11 Curna, Inc. Treatment of diseases related to RNASA (H1) by inhibition of the natural antisense transcript to RNASA H1
CN102844435B (en) 2010-02-22 2017-05-10 库尔纳公司 Treatment of pyrroline-5-carboxylate reductase 1 (pycr1) related diseases by inhibition of natural antisense transcript to pycr1
RU2612884C2 (en) 2010-04-02 2017-03-13 Курна, Инк. Treatment of diseases associated with colonystimulating factor 3 (csf3) by inhibition of natural antisense transcript to csf3
EP2556160A4 (en) 2010-04-09 2013-08-21 Curna Inc Treatment of fibroblast growth factor 21 (fgf21) related diseases by inhibition of natural antisense transcript to fgf21
WO2011139387A1 (en) 2010-05-03 2011-11-10 Opko Curna, Llc Treatment of sirtuin (sirt) related diseases by inhibition of natural antisense transcript to a sirtuin (sirt)
TWI586356B (en) 2010-05-14 2017-06-11 可娜公司 Treatment of par4 related diseases by inhibition of natural antisense transcript to par4
EP2576784B1 (en) 2010-05-26 2017-11-15 CuRNA, Inc. Treatment of methionine sulfoxide reductase a (msra) related diseases by inhibition of natural antisense transcript to msra
KR20180053419A (en) 2010-05-26 2018-05-21 큐알엔에이, 인크. Treatment of atonal homolog 1 (atoh1) related diseases by inhibition of natural antisense transcript to atoh1
WO2011163499A2 (en) 2010-06-23 2011-12-29 Opko Curna, Llc Treatment of sodium channel, voltage-gated, alpha subunit (scna) related diseases by inhibition of natural antisense transcript to scna
WO2012009402A2 (en) 2010-07-14 2012-01-19 Opko Curna Llc Treatment of discs large homolog (dlg) related diseases by inhibition of natural antisense transcript to dlg
CA2813901C (en) 2010-10-06 2019-11-12 Curna, Inc. Treatment of sialidase 4 (neu4) related diseases by inhibition of natural antisense transcript to neu4
EP2630241B1 (en) 2010-10-22 2018-10-17 CuRNA, Inc. Treatment of alpha-l-iduronidase (idua) related diseases by inhibition of natural antisense transcript to idua
EP2633046A4 (en) * 2010-10-29 2015-05-06 Alnylam Pharmaceuticals Inc Compositions and methods for inhibition of pcsk9 genes
US10000752B2 (en) 2010-11-18 2018-06-19 Curna, Inc. Antagonat compositions and methods of use
ES2657590T3 (en) 2010-11-23 2018-03-06 Curna, Inc. Treatment of nanog related diseases by inhibiting the natural antisense transcript to nanog
RU2721279C2 (en) 2011-01-28 2020-05-18 Санофи Байотекнолоджи Human antibodies to pcsk9 for use in methods of treating specific groups of individuals
RU2620980C2 (en) 2011-06-09 2017-05-30 Курна, Инк. Treatment of diseases associated with frataxin (fxn), by inhibiting natural antisense fxn transcript
AR087305A1 (en) 2011-07-28 2014-03-12 Regeneron Pharma STABILIZED FORMULATIONS CONTAINING ANTI-PCSK9 ANTIBODIES, PREPARATION METHOD AND KIT
US10583128B2 (en) 2011-09-06 2020-03-10 Curna, Inc. Treatment of diseases related to alpha subunits of sodium channels, voltage-gated (SCNxA) with small molecules
PL2570135T3 (en) * 2011-09-13 2016-07-29 Affiris Ag PCSK9 Vaccine
KR102239887B1 (en) 2012-02-24 2021-04-13 아뷰터스 바이오파마 코포레이션 Trialkyl cationic lipids and methods of use thereof
US20150031750A1 (en) 2012-03-15 2015-01-29 The Scripps Research Institute Treatment of brain derived neurotrophic factor (bdnf) related diseases by inhibition of natural antisense transcript to bdnf
US9255154B2 (en) 2012-05-08 2016-02-09 Alderbio Holdings, Llc Anti-PCSK9 antibodies and use thereof
EP3336187A1 (en) * 2012-12-05 2018-06-20 Alnylam Pharmaceuticals, Inc. Pcsk9 irna compositions and methods of use thereof
EP2810955A1 (en) 2013-06-07 2014-12-10 Sanofi Methods for inhibiting atherosclerosis by administering an inhibitor of PCSK9
CA2914721A1 (en) 2013-06-07 2014-12-11 Regeneron Pharmaceuticals, Inc. Methods for inhibiting atherosclerosis by administering an inhibitor of pcsk9
EP2862877A1 (en) 2013-10-18 2015-04-22 Sanofi Methods for inhibiting atherosclerosis by administering an inhibitor of PCSK9
WO2014204728A1 (en) * 2013-06-17 2014-12-24 The Broad Institute Inc. Delivery, engineering and optimization of systems, methods and compositions for targeting and modeling diseases and disorders of post mitotic cells
EA201592215A1 (en) 2013-06-27 2016-05-31 Рош Инновейшен Сентер Копенгаген А/С ANTISMINAL OLIGOMERS AND THEIR CONJUGATES AIMED AT PROPROTEIN CONVERTASY SUBTILYSINE / KEXIN TYPE 9 (PCSK9)
CA2932479A1 (en) * 2013-12-12 2015-06-18 The Rockefeller University Delivery, use and therapeutic applications of the crispr-cas systems and compositions for hbv and viral diseases and disorders
US8980273B1 (en) 2014-07-15 2015-03-17 Kymab Limited Method of treating atopic dermatitis or asthma using antibody to IL4RA
US9034332B1 (en) 2014-07-15 2015-05-19 Kymab Limited Precision medicine by targeting rare human PCSK9 variants for cholesterol treatment
US9051378B1 (en) 2014-07-15 2015-06-09 Kymab Limited Targeting rare human PCSK9 variants for cholesterol treatment
US9023359B1 (en) 2014-07-15 2015-05-05 Kymab Limited Targeting rare human PCSK9 variants for cholesterol treatment
US9045545B1 (en) 2014-07-15 2015-06-02 Kymab Limited Precision medicine by targeting PD-L1 variants for treatment of cancer
US8883157B1 (en) 2013-12-17 2014-11-11 Kymab Limited Targeting rare human PCSK9 variants for cholesterol treatment
US9017678B1 (en) 2014-07-15 2015-04-28 Kymab Limited Method of treating rheumatoid arthritis using antibody to IL6R
US8986694B1 (en) 2014-07-15 2015-03-24 Kymab Limited Targeting human nav1.7 variants for treatment of pain
US9045548B1 (en) 2014-07-15 2015-06-02 Kymab Limited Precision Medicine by targeting rare human PCSK9 variants for cholesterol treatment
US8992927B1 (en) 2014-07-15 2015-03-31 Kymab Limited Targeting human NAV1.7 variants for treatment of pain
US9914769B2 (en) 2014-07-15 2018-03-13 Kymab Limited Precision medicine for cholesterol treatment
US9067998B1 (en) 2014-07-15 2015-06-30 Kymab Limited Targeting PD-1 variants for treatment of cancer
US8986691B1 (en) 2014-07-15 2015-03-24 Kymab Limited Method of treating atopic dermatitis or asthma using antibody to IL4RA
US8945560B1 (en) 2014-07-15 2015-02-03 Kymab Limited Method of treating rheumatoid arthritis using antibody to IL6R
US10934542B2 (en) * 2013-12-27 2021-03-02 Bonac Corporation Artificial match-type miRNA for controlling gene expression and use therefor
GB201403775D0 (en) 2014-03-04 2014-04-16 Kymab Ltd Antibodies, uses & methods
US9139648B1 (en) 2014-07-15 2015-09-22 Kymab Limited Precision medicine by targeting human NAV1.9 variants for treatment of pain
US9150660B1 (en) 2014-07-15 2015-10-06 Kymab Limited Precision Medicine by targeting human NAV1.8 variants for treatment of pain
AU2015289613B2 (en) 2014-07-16 2021-07-01 Regeneron Pharmaceuticals, Inc. Methods for treating patients with heterozygous familial hypercholesterolemia (heFH)
JOP20200115A1 (en) * 2014-10-10 2017-06-16 Alnylam Pharmaceuticals Inc Compositions And Methods For Inhibition Of HAO1 (Hydroxyacid Oxidase 1 (Glycolate Oxidase)) Gene Expression
ES2789049T3 (en) * 2014-12-26 2020-10-23 Nitto Denko Corp RNA Interference Agents for GST-pi Gene Modulation
US20180002702A1 (en) 2014-12-26 2018-01-04 Nitto Denko Corporation Methods and compositions for treating malignant tumors associated with kras mutation
US10792299B2 (en) 2014-12-26 2020-10-06 Nitto Denko Corporation Methods and compositions for treating malignant tumors associated with kras mutation
JP2018523684A (en) 2015-08-18 2018-08-23 リジェネロン・ファーマシューティカルズ・インコーポレイテッドRegeneron Pharmaceuticals, Inc. Anti-PCSK9 inhibitory antibody for treating hyperlipidemic patients undergoing lipoprotein apheresis
AU2016310494B2 (en) 2015-08-25 2022-06-09 Alnylam Pharmaceuticals, Inc. Methods and compositions for treating a proprotein convertase subtilisin kexin (PCSK9) gene-associated disorder
JP2018531037A (en) 2015-10-19 2018-10-25 アールエックスアイ ファーマシューティカルズ コーポレーション Reduced size self-delivering nucleic acid compounds targeting long non-coding RNAs
KR20190077411A (en) * 2016-10-18 2019-07-03 더 메디슨스 컴퍼니 Preventing Cardiovascular Events Through Protein Reduction Protein Conjugate Enzymes (PCSK9)
WO2018083248A1 (en) 2016-11-03 2018-05-11 Kymab Limited Antibodies, combinations comprising antibodies, biomarkers, uses & methods
JOP20190112A1 (en) 2016-11-14 2019-05-14 Amgen Inc Combined therapies for atherosclerosis, including atherosclerotic cardiovascular disease
CN108265052B (en) * 2016-12-30 2021-05-28 苏州瑞博生物技术股份有限公司 Small interfering nucleic acid, pharmaceutical composition and application thereof
JP2019508379A (en) * 2017-02-16 2019-03-28 日東電工株式会社 Therapeutic method and therapeutic composition for malignant tumor
JOP20190215A1 (en) * 2017-03-24 2019-09-19 Ionis Pharmaceuticals Inc Modulators of pcsk9 expression
CN109957565B (en) * 2017-12-26 2023-04-07 广州市锐博生物科技有限公司 Modified siRNA molecule and application thereof
CN109957567B (en) * 2017-12-26 2022-09-23 阿格纳生物制药有限公司 siRNA molecule for inhibiting PCSK9 gene expression and application thereof
JP7357002B2 (en) * 2018-04-18 2023-10-05 ディセルナ ファーマシューティカルズ インコーポレイテッド Oligonucleotides targeting PCSK9 for treating hypercholesterolemia and related conditions
CN111378656B (en) * 2018-12-28 2022-07-26 苏州瑞博生物技术股份有限公司 Nucleic acid for inhibiting Ebola virus, pharmaceutical composition containing nucleic acid and application of pharmaceutical composition
JP2023519140A (en) * 2020-03-16 2023-05-10 アルゴノート アールエヌエー リミテッド PCSK9 antagonist
WO2022089486A1 (en) * 2020-10-28 2022-05-05 江苏柯菲平医药股份有限公司 Sirna for inhibiting pcsk9 gene expression and modifier thereof and use thereof
CN113980966A (en) * 2021-09-29 2022-01-28 阿格纳生物制药有限公司 Nucleic acid sequence for inhibiting PCSK9 target gene expression and application thereof
CN117384907B (en) * 2023-12-11 2024-03-29 上海鼎新基因科技有限公司 siRNA molecule for inhibiting PCSK9 expression and application thereof

Family Cites Families (133)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3687808A (en) 1969-08-14 1972-08-29 Univ Leland Stanford Junior Synthetic polynucleotides
US4469863A (en) 1980-11-12 1984-09-04 Ts O Paul O P Nonionic nucleic acid alkyl and aryl phosphonates and processes for manufacture and use thereof
US5023243A (en) 1981-10-23 1991-06-11 Molecular Biosystems, Inc. Oligonucleotide therapeutic agent and method of making same
US4476301A (en) 1982-04-29 1984-10-09 Centre National De La Recherche Scientifique Oligonucleotides, a process for preparing the same and their application as mediators of the action of interferon
US5550111A (en) 1984-07-11 1996-08-27 Temple University-Of The Commonwealth System Of Higher Education Dual action 2',5'-oligoadenylate antiviral derivatives and uses thereof
US5034506A (en) 1985-03-15 1991-07-23 Anti-Gene Development Group Uncharged morpholino-based polymers having achiral intersubunit linkages
US5166315A (en) 1989-12-20 1992-11-24 Anti-Gene Development Group Sequence-specific binding polymers for duplex nucleic acids
US5235033A (en) 1985-03-15 1993-08-10 Anti-Gene Development Group Alpha-morpholino ribonucleoside derivatives and polymers thereof
US5185444A (en) 1985-03-15 1993-02-09 Anti-Gene Deveopment Group Uncharged morpolino-based polymers having phosphorous containing chiral intersubunit linkages
US5405938A (en) 1989-12-20 1995-04-11 Anti-Gene Development Group Sequence-specific binding polymers for duplex nucleic acids
AU6131086A (en) 1985-07-05 1987-01-30 Whitehead Institute For Biomedical Research Epithelial cells expressing foreign genetic material
US4980286A (en) 1985-07-05 1990-12-25 Whitehead Institute For Biomedical Research In vivo introduction and expression of foreign genetic material in epithelial cells
US5139941A (en) 1985-10-31 1992-08-18 University Of Florida Research Foundation, Inc. AAV transduction vectors
US5276019A (en) 1987-03-25 1994-01-04 The United States Of America As Represented By The Department Of Health And Human Services Inhibitors for replication of retroviruses and for the expression of oncogene products
US5264423A (en) 1987-03-25 1993-11-23 The United States Of America As Represented By The Department Of Health And Human Services Inhibitors for replication of retroviruses and for the expression of oncogene products
EP0633318A1 (en) 1987-09-11 1995-01-11 Whitehead Institute For Biomedical Research Transduced fibroblasts and uses therefor
US4924624A (en) 1987-10-22 1990-05-15 Temple University-Of The Commonwealth System Of Higher Education 2,',5'-phosphorothioate oligoadenylates and plant antiviral uses thereof
US5188897A (en) 1987-10-22 1993-02-23 Temple University Of The Commonwealth System Of Higher Education Encapsulated 2',5'-phosphorothioate oligoadenylates
ATE110108T1 (en) 1987-12-11 1994-09-15 Whitehead Biomedical Inst GENETIC MODIFICATION OF ENDOTHELIAL CELLS.
ATE152169T1 (en) 1988-02-05 1997-05-15 Whitehead Biomedical Inst MODIFIED HEPATOCYTES AND THEIR APPLICATION
EP0406309A4 (en) 1988-03-25 1992-08-19 The University Of Virginia Alumni Patents Foundation Oligonucleotide n-alkylphosphoramidates
US5278302A (en) 1988-05-26 1994-01-11 University Patents, Inc. Polynucleotide phosphorodithioates
US5216141A (en) 1988-06-06 1993-06-01 Benner Steven A Oligonucleotide analogs containing sulfur linkages
US5328470A (en) 1989-03-31 1994-07-12 The Regents Of The University Of Michigan Treatment of diseases by site-specific instillation of cells or site-specific transformation of cells and kits therefor
US5399676A (en) 1989-10-23 1995-03-21 Gilead Sciences Oligonucleotides with inverted polarity
US5264564A (en) 1989-10-24 1993-11-23 Gilead Sciences Oligonucleotide analogs with novel linkages
US5264562A (en) 1989-10-24 1993-11-23 Gilead Sciences, Inc. Oligonucleotide analogs with novel linkages
US5177198A (en) 1989-11-30 1993-01-05 University Of N.C. At Chapel Hill Process for preparing oligoribonucleoside and oligodeoxyribonucleoside boranophosphates
US5223168A (en) 1989-12-12 1993-06-29 Gary Holt Surface cleaner and treatment
US5212295A (en) 1990-01-11 1993-05-18 Isis Pharmaceuticals Monomers for preparation of oligonucleotides having chiral phosphorus linkages
US5506351A (en) 1992-07-23 1996-04-09 Isis Pharmaceuticals Process for the preparation of 2'-O-alkyl guanosine and related compounds
US5587361A (en) 1991-10-15 1996-12-24 Isis Pharmaceuticals, Inc. Oligonucleotides having phosphorothioate linkages of high chiral purity
US5459255A (en) 1990-01-11 1995-10-17 Isis Pharmaceuticals, Inc. N-2 substituted purines
US5587470A (en) 1990-01-11 1996-12-24 Isis Pharmaceuticals, Inc. 3-deazapurines
US5457191A (en) 1990-01-11 1995-10-10 Isis Pharmaceuticals, Inc. 3-deazapurines
US5578718A (en) 1990-01-11 1996-11-26 Isis Pharmaceuticals, Inc. Thiol-derivatized nucleosides
US5321131A (en) 1990-03-08 1994-06-14 Hybridon, Inc. Site-specific functionalization of oligodeoxynucleotides for non-radioactive labelling
US5470967A (en) 1990-04-10 1995-11-28 The Dupont Merck Pharmaceutical Company Oligonucleotide analogs with sulfamate linkages
US5677437A (en) 1990-07-27 1997-10-14 Isis Pharmaceuticals, Inc. Heteroatomic oligonucleoside linkages
US5610289A (en) 1990-07-27 1997-03-11 Isis Pharmaceuticals, Inc. Backbone modified oligonucleotide analogues
US5618704A (en) 1990-07-27 1997-04-08 Isis Pharmacueticals, Inc. Backbone-modified oligonucleotide analogs and preparation thereof through radical coupling
US5602240A (en) 1990-07-27 1997-02-11 Ciba Geigy Ag. Backbone modified oligonucleotide analogs
US5541307A (en) 1990-07-27 1996-07-30 Isis Pharmaceuticals, Inc. Backbone modified oligonucleotide analogs and solid phase synthesis thereof
US5608046A (en) 1990-07-27 1997-03-04 Isis Pharmaceuticals, Inc. Conjugated 4'-desmethyl nucleoside analog compounds
US5623070A (en) 1990-07-27 1997-04-22 Isis Pharmaceuticals, Inc. Heteroatomic oligonucleoside linkages
US5138045A (en) 1990-07-27 1992-08-11 Isis Pharmaceuticals Polyamine conjugated oligonucleotides
US5386023A (en) 1990-07-27 1995-01-31 Isis Pharmaceuticals Backbone modified oligonucleotide analogs and preparation thereof through reductive coupling
US5218105A (en) 1990-07-27 1993-06-08 Isis Pharmaceuticals Polyamine conjugated oligonucleotides
US5489677A (en) 1990-07-27 1996-02-06 Isis Pharmaceuticals, Inc. Oligonucleoside linkages containing adjacent oxygen and nitrogen atoms
US5378825A (en) 1990-07-27 1995-01-03 Isis Pharmaceuticals, Inc. Backbone modified oligonucleotide analogs
WO1992002534A2 (en) 1990-08-03 1992-02-20 Sterling Drug, Inc. Compounds and methods for inhibiting gene expression
US6262241B1 (en) 1990-08-13 2001-07-17 Isis Pharmaceuticals, Inc. Compound for detecting and modulating RNA activity and gene expression
US5177196A (en) 1990-08-16 1993-01-05 Microprobe Corporation Oligo (α-arabinofuranosyl nucleotides) and α-arabinofuranosyl precursors thereof
US5214134A (en) 1990-09-12 1993-05-25 Sterling Winthrop Inc. Process of linking nucleosides with a siloxane bridge
US5561225A (en) 1990-09-19 1996-10-01 Southern Research Institute Polynucleotide analogs containing sulfonate and sulfonamide internucleoside linkages
WO1992005186A1 (en) 1990-09-20 1992-04-02 Gilead Sciences Modified internucleoside linkages
EP0568537B1 (en) 1990-10-31 1998-02-04 Somatix Therapy Corporation Genetic modification of endothelial cells
US5539082A (en) 1993-04-26 1996-07-23 Nielsen; Peter E. Peptide nucleic acids
EP0745833A3 (en) 1991-07-25 1998-09-23 The Whitaker Corporation Liquid level sensor
US5571799A (en) 1991-08-12 1996-11-05 Basco, Ltd. (2'-5') oligoadenylate analogues useful as inhibitors of host-v5.-graft response
US5599797A (en) 1991-10-15 1997-02-04 Isis Pharmaceuticals, Inc. Oligonucleotides having phosphorothioate linkages of high chiral purity
ATE239484T1 (en) 1991-10-24 2003-05-15 Isis Pharmaceuticals Inc DERIVATIZED OLIGONUCLEOTIDES WITH IMPROVED ABSORPTION CAPACITY
US5252479A (en) 1991-11-08 1993-10-12 Research Corporation Technologies, Inc. Safe vector for gene therapy
US5633360A (en) 1992-04-14 1997-05-27 Gilead Sciences, Inc. Oligonucleotide analogs capable of passive cell membrane permeation
US5434257A (en) 1992-06-01 1995-07-18 Gilead Sciences, Inc. Binding compentent oligomers containing unsaturated 3',5' and 2',5' linkages
US5587308A (en) 1992-06-02 1996-12-24 The United States Of America As Represented By The Department Of Health & Human Services Modified adeno-associated virus vector capable of expression from a novel promoter
US5478745A (en) 1992-12-04 1995-12-26 University Of Pittsburgh Recombinant viral vector system
US5476925A (en) 1993-02-01 1995-12-19 Northwestern University Oligodeoxyribonucleotides including 3'-aminonucleoside-phosphoramidate linkages and terminal 3'-amino groups
GB9304618D0 (en) 1993-03-06 1993-04-21 Ciba Geigy Ag Chemical compounds
EP0691977B1 (en) 1993-03-31 1997-11-26 Sanofi Oligonucleotides with amide linkages replacing phosphodiester linkages
US5571902A (en) 1993-07-29 1996-11-05 Isis Pharmaceuticals, Inc. Synthesis of oligonucleotides
US5625050A (en) 1994-03-31 1997-04-29 Amgen Inc. Modified oligonucleotides and intermediates useful in nucleic acid therapeutics
US6054299A (en) 1994-04-29 2000-04-25 Conrad; Charles A. Stem-loop cloning vector and method
US5554746A (en) 1994-05-16 1996-09-10 Isis Pharmaceuticals, Inc. Lactam nucleic acids
JP4335310B2 (en) 1995-06-07 2009-09-30 ザ ユニバーシティ オブ ブリティッシュ コロンビア Lipid-nucleic acid particles prepared through hydrophobic lipid-nucleic acid complex intermediates and use for gene transfer
US6506559B1 (en) 1997-12-23 2003-01-14 Carnegie Institute Of Washington Genetic inhibition by double-stranded RNA
EP1068311B2 (en) 1998-04-08 2020-12-09 Commonwealth Scientific and Industrial Research Organisation Methods and means for obtaining modified phenotypes
AR020078A1 (en) 1998-05-26 2002-04-10 Syngenta Participations Ag METHOD FOR CHANGING THE EXPRESSION OF AN OBJECTIVE GENE IN A PLANT CELL
BR9914773A (en) 1998-10-09 2002-02-05 Ingene Inc Set of generic elements, method for the production of single-stranded DNA, mrna transcription, nucleic acid construction, ssdna transcription, vector, vector system, host cell, set for the production of a single-stranded nucleic acid sequence, method for in vivo or in vitro production of a single-stranded nucleic acid sequence, transcription of single-stranded cdna, inhibitory nucleic acid, heteroduplex molecule, and pharmaceutical composition
AU6430599A (en) 1998-10-09 2000-05-01 Cytogenix, Inc. Enzymatic synthesis of ssdna
DE19956568A1 (en) 1999-01-30 2000-08-17 Roland Kreutzer Method and medicament for inhibiting the expression of a given gene
US6271359B1 (en) 1999-04-14 2001-08-07 Musc Foundation For Research Development Tissue-specific and pathogen-specific toxic agents and ribozymes
DE10100586C1 (en) 2001-01-09 2002-04-11 Ribopharma Ag Inhibiting gene expression in cells, useful for e.g. treating tumors, by introducing double-stranded complementary oligoRNA having unpaired terminal bases
AU2001247951A1 (en) * 2000-02-07 2001-08-14 Roche Diagnostics Corporation Novel cationic amphiphiles
WO2003070918A2 (en) 2002-02-20 2003-08-28 Ribozyme Pharmaceuticals, Incorporated Rna interference by modified short interfering nucleic acid
US20070026394A1 (en) * 2000-02-11 2007-02-01 Lawrence Blatt Modulation of gene expression associated with inflammation proliferation and neurite outgrowth using nucleic acid based technologies
CZ308053B6 (en) * 2000-12-01 2019-11-27 Max Planck Gesellschaft Isolated double-stranded RNA molecule, process for producing it and its use
US20070173473A1 (en) * 2001-05-18 2007-07-26 Sirna Therapeutics, Inc. RNA interference mediated inhibition of proprotein convertase subtilisin Kexin 9 (PCSK9) gene expression using short interfering nucleic acid (siNA)
US20080249040A1 (en) * 2001-05-18 2008-10-09 Sirna Therapeutics, Inc. RNA interference mediated inhibition of sterol regulatory element binding protein 1 (SREBP1) gene expression using short interfering nucleic acid (siNA)
US20030170891A1 (en) * 2001-06-06 2003-09-11 Mcswiggen James A. RNA interference mediated inhibition of epidermal growth factor receptor gene expression using short interfering nucleic acid (siNA)
US20040009216A1 (en) * 2002-04-05 2004-01-15 Rodrigueza Wendi V. Compositions and methods for dosing liposomes of certain sizes to treat or prevent disease
US7956176B2 (en) * 2002-09-05 2011-06-07 Sirna Therapeutics, Inc. RNA interference mediated inhibition of gene expression using chemically modified short interfering nucleic acid (siNA)
JP2006507841A (en) * 2002-11-14 2006-03-09 ダーマコン, インコーポレイテッド Functional and ultrafunctional siRNA
DE10302421A1 (en) 2003-01-21 2004-07-29 Ribopharma Ag New double-stranded interfering RNA, useful for inhibiting hepatitis C virus, has one strand linked to a lipophilic group to improve activity and eliminate the need for transfection auxiliaries
AU2004220556B2 (en) 2003-03-07 2009-05-07 Alnylam Pharmaceuticals, Inc. Therapeutic compositions
EP1608735A4 (en) 2003-04-03 2008-11-05 Alnylam Pharmaceuticals Irna conjugates
EP1471152A1 (en) * 2003-04-25 2004-10-27 Institut National De La Sante Et De La Recherche Medicale (Inserm) Mutations in the human PCSK9 gene associated to hypercholesterolemia
DK3604537T3 (en) * 2003-06-13 2022-02-28 Alnylam Europe Ag Double-stranded ribonucleic acid with increased efficiency in an organism
AU2004257373B2 (en) 2003-07-16 2011-03-24 Arbutus Biopharma Corporation Lipid encapsulated interfering RNA
CA2542835A1 (en) * 2003-10-23 2005-05-19 Sirna Therapeutics, Inc. Rna interference mediated inhibition of gene expression using short interfering nucleic acid (sina)
US20080253960A1 (en) * 2004-04-01 2008-10-16 The Trustees Of The University Of Pennsylvania Center For Technology Transfer Lipoprotein-Based Nanoplatforms
JP4796062B2 (en) * 2004-06-07 2011-10-19 プロチバ バイオセラピューティクス インコーポレイティッド Lipid-encapsulating interfering RNA
EP1781593B1 (en) 2004-06-07 2011-12-14 Protiva Biotherapeutics Inc. Cationic lipids and methods of use
CA2597724A1 (en) * 2005-02-14 2007-08-02 Sirna Therapeutics, Inc. Cationic lipids and formulated molecular compositions containing them
PL1866414T3 (en) * 2005-03-31 2012-10-31 Calando Pharmaceuticals Inc Inhibitors of ribonucleotide reductase subunit 2 and uses thereof
US7915230B2 (en) 2005-05-17 2011-03-29 Molecular Transfer, Inc. Reagents for transfection of eukaryotic cells
CN101267805A (en) 2005-07-27 2008-09-17 普洛体维生物治疗公司 Systems and methods for manufacturing liposomes
EP2395012B8 (en) 2005-11-02 2018-06-06 Arbutus Biopharma Corporation Modified siRNA molecules and uses thereof
US20070218122A1 (en) 2005-11-18 2007-09-20 Protiva Biotherapeutics, Inc. siRNA silencing of influenza virus gene expression
KR101547579B1 (en) * 2006-03-31 2015-08-27 알닐람 파마슈티칼스 인코포레이티드 DsRNA for inhibiting expression of Eg5 gene
WO2007134161A2 (en) * 2006-05-11 2007-11-22 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of the pcsk9 gene
US8598333B2 (en) * 2006-05-26 2013-12-03 Alnylam Pharmaceuticals, Inc. SiRNA silencing of genes expressed in cancer
JP2010503382A (en) * 2006-07-17 2010-02-04 サーナ・セラピューティクス・インコーポレイテッド RNA interference-mediated inhibition of proprotein convertase subtilisin kexin 9 (PCSK9) gene expression using small interfering nucleic acids (siNA)
ES2611924T3 (en) 2006-10-03 2017-05-11 Arbutus Biopharma Corporation Formulations containing lipids
CA2670563A1 (en) 2006-11-27 2008-06-05 Isis Pharmaceuticals, Inc. Methods for treating hypercholesterolemia
WO2008109369A2 (en) 2007-03-02 2008-09-12 Mdrna, Inc. Nucleic acid compounds for inhibiting tnf gene expression and uses thereof
JOP20080381B1 (en) * 2007-08-23 2023-03-28 Amgen Inc Antigen Binding Proteins to Proprotein Convertase subtillisin Kexin type 9 (pcsk9)
CA2710713C (en) * 2007-12-27 2017-09-19 Protiva Biotherapeutics, Inc. Silencing of polo-like kinase expression using interfering rna
EP2224912B1 (en) 2008-01-02 2016-05-11 TEKMIRA Pharmaceuticals Corporation Improved compositions and methods for the delivery of nucleic acids
AU2009241591A1 (en) * 2008-01-31 2009-11-05 Alnylam Pharmaceuticals, Inc. Optimized methods for delivery of DSRNA targeting the PCSK9 gene
JP2011518117A (en) 2008-03-05 2011-06-23 アルナイラム ファーマシューティカルズ, インコーポレイテッド Compositions and methods for inhibiting expression of Eg5 and VEGF genes
US20110065644A1 (en) 2008-03-09 2011-03-17 Intradigm Corporation Compositions comprising human pcsk9 and apolipoprotein b sirna and methods of use
NZ588583A (en) * 2008-04-15 2012-08-31 Protiva Biotherapeutics Inc Novel lipid formulations for nucleic acid delivery
EA029762B1 (en) * 2008-10-20 2018-05-31 Элнилэм Фармасьютикалз, Инк. Compositions and methods for inhibiting expression of transthyretin
KR20220150995A (en) 2008-11-10 2022-11-11 알닐람 파마슈티칼스 인코포레이티드 Novel lipids and compositions for the delivery of therapeutics
WO2010068816A1 (en) 2008-12-10 2010-06-17 Alnylam Pharmaceuticals, Inc. Gnaq targeted dsrna compositions and methods for inhibiting expression
US8311190B2 (en) * 2008-12-23 2012-11-13 International Business Machines Corporation Performing human client verification over a voice interface
CA2751342C (en) 2009-01-29 2019-05-07 Alnylam Pharmaceuticals, Inc. Lipid formulations comprising cationic lipid and a targeting lipid comprising n-acetyl galactosamine for delivery of nucleic acid
SG10201402054UA (en) 2009-05-05 2014-09-26 Muthiah Manoharan Lipid compositions
CN104873464B (en) 2009-06-10 2018-06-22 阿布特斯生物制药公司 Improved lipid formulations
WO2010147992A1 (en) 2009-06-15 2010-12-23 Alnylam Pharmaceuticals, Inc. Methods for increasing efficacy of lipid formulated sirna
CN104651408A (en) 2009-06-15 2015-05-27 阿尔尼拉姆医药品有限公司 Lipid formulated siRNA targeted to PCSK9 gene
US8563528B2 (en) 2009-07-21 2013-10-22 Santaris Pharma A/S Antisense oligomers targeting PCSK9

Non-Patent Citations (8)

* Cited by examiner, † Cited by third party
Title
BASAK A.: 'Inhibitors of proprotein convertases' JOURNAL OF MOLECULAR MEDICINE vol. 83, 2005, pages 844 - 855, XP019320304 *
DATABASE GENBANK [Online] XP008099660 Database accession no. (NM_174936) *
DOWNWARD J.: 'Science, medicine, and the future RNA interference' BRITISH MEDICAL JOURNAL vol. 328, 2004, pages 1245 - 1248, XP003002362 *
ELBASHIR ET AL.: 'Functional anatomy of siRNAs for mediating efficient RNAi in Drosophila melanogaster embryo lysate' THE EMBO JOURNAL vol. 20, 2001, pages 6877 - 6888, XP002225998 *
GRAHAM M.J. ET AL: 'Antisense inhibition of proportein convertase subtilisin/kexin type 9 reduces serum LDL in hyperlipidemic mice' JOURNAL OF LIPID RESEARCH vol. 48, 2007, pages 763 - 767, XP002478035 *
LU ET AL.: 'Delivering siRNA in vivo for functional genomics and novel therapeutics' RNA INTERFERENCE TECHNOLOGY (CAMBRIDGE) 2005, pages 303 - 317, XP008070935 *
SAMARSKY ET AL.: 'RNAi in drug development' RNA INTERFERENCE TECHNOLOGY (CAMBRIDGE) 2005, pages 384 - 395, XP008070657 *
See also references of EP2021507A2 *

Cited By (59)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10501742B2 (en) 2006-05-11 2019-12-10 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of the PCSK9 gene
US9260718B2 (en) 2006-05-11 2016-02-16 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of the PCSK9 gene
US9822365B2 (en) 2006-05-11 2017-11-21 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of the PCSK9 gene
US8809292B2 (en) 2006-05-11 2014-08-19 Alnylam Pharmaceuticals, Inc Compositions and methods for inhibiting expression of the PCSK9 gene
US8222222B2 (en) 2006-05-11 2012-07-17 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of the PCSK9 gene
US8344114B2 (en) 2006-11-07 2013-01-01 Merck Sharp & Dohme Corp. Antagonists of PCSK9
JP2010510807A (en) * 2006-11-27 2010-04-08 アイシス ファーマシューティカルズ, インコーポレーテッド Methods for treating hypercholesterolemia
JP2011511004A (en) * 2008-01-31 2011-04-07 アルナイラム ファーマシューティカルズ インコーポレイテッド Optimized method for delivering dsRNA targeting the PCSK9 gene
WO2009134487A3 (en) * 2008-01-31 2010-02-04 Alnylam Pharmaceuticals, Inc. Optimized methods for delivery of dsrna targeting the pcsk9 gene
US8697070B2 (en) 2008-02-07 2014-04-15 Merck Sharp & Dohme Corp. 1D05 PCSK9 antagonists
US8188234B2 (en) 2008-02-07 2012-05-29 Merck Sharp & Dohme Corp. 1D05 PCSK9 antagonists
US8188233B2 (en) 2008-02-07 2012-05-29 Merck Sharp & Dohme Corp. 1B20 PCSK9 antagonists
US8957194B2 (en) 2008-02-07 2015-02-17 Merck Sharpe & Dohme Corp. 1B20 PCSK9 antagonists
EP2268316A4 (en) * 2008-03-20 2011-05-25 Quark Pharmaceuticals Inc NOVEL siRNA COMPOUNDS FOR INHIBITING RTP801
EP2268316A2 (en) * 2008-03-20 2011-01-05 Quark Pharmaceuticals, Inc. NOVEL siRNA COMPOUNDS FOR INHIBITING RTP801
US9957505B2 (en) 2009-06-01 2018-05-01 Halo-Bio Rnai Therapeutics, Inc. Polynucleotides for multivalent RNA interference, compositions and methods of use thereof
US9200276B2 (en) 2009-06-01 2015-12-01 Halo-Bio Rnai Therapeutics, Inc. Polynucleotides for multivalent RNA interference, compositions and methods of use thereof
JP2016074685A (en) * 2009-06-15 2016-05-12 アルナイラム ファーマシューティカルズ, インコーポレイテッドAlnylam Pharmaceuticals, Inc. Lipid formulated dsrna targeting pcsk9 gene
US9051567B2 (en) 2009-06-15 2015-06-09 Tekmira Pharmaceuticals Corporation Methods for increasing efficacy of lipid formulated siRNA
CN104651408A (en) * 2009-06-15 2015-05-27 阿尔尼拉姆医药品有限公司 Lipid formulated siRNA targeted to PCSK9 gene
JP2012530143A (en) * 2009-06-15 2012-11-29 アルナイラム ファーマシューティカルズ, インコーポレイテッド DSRNA formulated with lipids targeting the PCSK9 gene
US10053689B2 (en) 2009-06-15 2018-08-21 Arbutus Biopharma Corporation Methods for increasing efficacy of lipid formulated siRNA
US8273869B2 (en) 2009-06-15 2012-09-25 Alnylam Pharmaceuticals, Inc. Lipid formulated dsRNA targeting the PCSK9 gene
US9187746B2 (en) 2009-09-22 2015-11-17 Alnylam Pharmaceuticals, Inc. Dual targeting siRNA agents
EP3252068A2 (en) 2009-10-12 2017-12-06 Larry J. Smith Methods and compositions for modulating gene expression using oligonucleotide based drugs administered in vivo or in vitro
EP4089169A1 (en) 2009-10-12 2022-11-16 Larry J. Smith Methods and compositions for modulating gene expression using oligonucleotide based drugs administered in vivo or in vitro
WO2012145729A2 (en) 2011-04-20 2012-10-26 Smith Holdings, Llc Methods and compositions for modulating gene expression using components that self assemble in cells and produce rnai activity
US10930367B2 (en) 2012-12-12 2021-02-23 The Broad Institute, Inc. Methods, models, systems, and apparatus for identifying target sequences for Cas enzymes or CRISPR-Cas systems for target sequences and conveying results thereof
US11041173B2 (en) 2012-12-12 2021-06-22 The Broad Institute, Inc. Delivery, engineering and optimization of systems, methods and compositions for sequence manipulation and therapeutic applications
WO2014182661A2 (en) 2013-05-06 2014-11-13 Alnylam Pharmaceuticals, Inc Dosages and methods for delivering lipid formulated nucleic acid molecules
US10577630B2 (en) 2013-06-17 2020-03-03 The Broad Institute, Inc. Delivery and use of the CRISPR-Cas systems, vectors and compositions for hepatic targeting and therapy
US11597949B2 (en) 2013-06-17 2023-03-07 The Broad Institute, Inc. Optimized CRISPR-Cas double nickase systems, methods and compositions for sequence manipulation
US10711285B2 (en) 2013-06-17 2020-07-14 The Broad Institute, Inc. Optimized CRISPR-Cas double nickase systems, methods and compositions for sequence manipulation
US10781444B2 (en) 2013-06-17 2020-09-22 The Broad Institute, Inc. Functional genomics using CRISPR-Cas systems, compositions, methods, screens and applications thereof
US11008588B2 (en) 2013-06-17 2021-05-18 The Broad Institute, Inc. Delivery, engineering and optimization of tandem guide systems, methods and compositions for sequence manipulation
US10946108B2 (en) 2013-06-17 2021-03-16 The Broad Institute, Inc. Delivery, use and therapeutic applications of the CRISPR-Cas systems and compositions for targeting disorders and diseases using viral components
US11407985B2 (en) 2013-12-12 2022-08-09 The Broad Institute, Inc. Delivery, use and therapeutic applications of the CRISPR-Cas systems and compositions for genome editing
US10851357B2 (en) 2013-12-12 2020-12-01 The Broad Institute, Inc. Compositions and methods of use of CRISPR-Cas systems in nucleotide repeat disorders
US10550372B2 (en) 2013-12-12 2020-02-04 The Broad Institute, Inc. Systems, methods and compositions for sequence manipulation with optimized functional CRISPR-Cas systems
US11597919B2 (en) 2013-12-12 2023-03-07 The Broad Institute Inc. Systems, methods and compositions for sequence manipulation with optimized functional CRISPR-Cas systems
US11591581B2 (en) 2013-12-12 2023-02-28 The Broad Institute, Inc. Compositions and methods of use of CRISPR-Cas systems in nucleotide repeat disorders
US11155795B2 (en) 2013-12-12 2021-10-26 The Broad Institute, Inc. CRISPR-Cas systems, crystal structure and uses thereof
US10696986B2 (en) 2014-12-12 2020-06-30 The Board Institute, Inc. Protected guide RNAS (PGRNAS)
US11624078B2 (en) 2014-12-12 2023-04-11 The Broad Institute, Inc. Protected guide RNAS (pgRNAS)
USRE49431E1 (en) 2014-12-26 2023-02-28 Nitto Denko Corporation RNA interference agents for GST-PI gene modulation
US11045488B2 (en) 2014-12-26 2021-06-29 Nitto Denko Corporation RNA interference agents for GST-π gene modulation
US10876100B2 (en) 2015-06-18 2020-12-29 The Broad Institute, Inc. Crispr enzyme mutations reducing off-target effects
US10494621B2 (en) 2015-06-18 2019-12-03 The Broad Institute, Inc. Crispr enzyme mutations reducing off-target effects
US11578312B2 (en) 2015-06-18 2023-02-14 The Broad Institute Inc. Engineering and optimization of systems, methods, enzymes and guide scaffolds of CAS9 orthologs and variants for sequence manipulation
US10731157B2 (en) 2015-08-24 2020-08-04 Halo-Bio Rnai Therapeutics, Inc. Polynucleotide nanoparticles for the modulation of gene expression and uses thereof
EP4035659A1 (en) 2016-11-29 2022-08-03 PureTech LYT, Inc. Exosomes for delivery of therapeutic agents
US11492620B2 (en) 2017-12-01 2022-11-08 Suzhou Ribo Life Science Co., Ltd. Double-stranded oligonucleotide, composition and conjugate comprising double-stranded oligonucleotide, preparation method thereof and use thereof
US11660347B2 (en) 2017-12-01 2023-05-30 Suzhou Ribo Life Science Co., Ltd. Nucleic acid, composition and conjugate containing same, preparation method, and use thereof
AU2017445584B2 (en) * 2017-12-26 2021-08-12 Argorna Pharmaceuticals Limited SiRNA molecule inhibiting the expression of the PCSK9 gene and use thereof
EP3677679A4 (en) * 2017-12-26 2020-12-02 Guangzhou Ribobio Co., Ltd. Sirna molecule inhibiting pcsk9 gene expression and application thereof
US11633482B2 (en) 2017-12-29 2023-04-25 Suzhou Ribo Life Science Co., Ltd. Conjugates and preparation and use thereof
US11918600B2 (en) 2018-08-21 2024-03-05 Suzhou Ribo Life Science Co., Ltd. Nucleic acid, pharmaceutical composition and conjugate containing nucleic acid, and use thereof
US11896674B2 (en) 2018-09-30 2024-02-13 Suzhou Ribo Life Science Co., Ltd. SiRNA conjugate, preparation method therefor and use thereof
WO2023017004A1 (en) * 2021-08-09 2023-02-16 Cargene Therapeutics Pte. Ltd. Inhibitory nucleic acids for pcsk9

Also Published As

Publication number Publication date
US7605251B2 (en) 2009-10-20
AU2007249329B2 (en) 2010-10-14
US20130331430A1 (en) 2013-12-12
EP3578656A1 (en) 2019-12-11
EP2021507A2 (en) 2009-02-11
KR101320916B1 (en) 2013-10-23
US20200318119A1 (en) 2020-10-08
JP2010246544A (en) 2010-11-04
US10501742B2 (en) 2019-12-10
EP2584047B1 (en) 2014-11-19
EA200870528A1 (en) 2009-06-30
EP3578656B1 (en) 2021-01-06
EP2021507A4 (en) 2009-10-28
WO2007134161A8 (en) 2009-05-07
ES2392478T3 (en) 2012-12-11
NZ587616A (en) 2012-03-30
CN103614375A (en) 2014-03-05
IL220102A0 (en) 2012-07-31
EP2835429A1 (en) 2015-02-11
US20140350075A1 (en) 2014-11-27
US20210403917A1 (en) 2021-12-30
JP6034316B2 (en) 2016-11-30
CA2915441A1 (en) 2007-11-22
JP5570806B2 (en) 2014-08-13
US20160348117A1 (en) 2016-12-01
AU2010241357A1 (en) 2010-12-02
US9260718B2 (en) 2016-02-16
EA015676B1 (en) 2011-10-31
ES2874149T3 (en) 2021-11-04
IL220102A (en) 2017-03-30
PL3578656T3 (en) 2021-08-02
AU2007249329C1 (en) 2011-03-24
EP2194128A1 (en) 2010-06-09
KR20090016021A (en) 2009-02-12
KR20100085186A (en) 2010-07-28
EP2584047A1 (en) 2013-04-24
EP2584048A1 (en) 2013-04-24
CA2651839A1 (en) 2007-11-22
EP3872179A1 (en) 2021-09-01
HK1183910A1 (en) 2014-01-10
JP2009536827A (en) 2009-10-22
AU2007249329A1 (en) 2007-11-22
US9822365B2 (en) 2017-11-21
US8809292B2 (en) 2014-08-19
NZ572666A (en) 2010-11-26
JP2014079258A (en) 2014-05-08
JP2016027830A (en) 2016-02-25
US20240093199A1 (en) 2024-03-21
IL195181A (en) 2013-02-28
EA201100907A1 (en) 2012-03-30
WO2007134161A3 (en) 2008-02-21
AU2010241357B2 (en) 2012-09-13
CN101484588B (en) 2013-11-06
EP3249052B1 (en) 2019-04-10
CA2651839C (en) 2016-02-09
CN101484588A (en) 2009-07-15
EP2584048B1 (en) 2014-07-23
EP2835429B1 (en) 2017-12-13
US20110230542A1 (en) 2011-09-22
KR101036126B1 (en) 2011-05-23
IL250678A0 (en) 2017-04-30
IL195181A0 (en) 2009-08-03
EP3249052A1 (en) 2017-11-29
EP2194128B1 (en) 2012-08-01
US20080113930A1 (en) 2008-05-15
US20180216115A1 (en) 2018-08-02
US8222222B2 (en) 2012-07-17
PL2194128T3 (en) 2012-12-31
EA020840B1 (en) 2015-02-27
SG171676A1 (en) 2011-06-29

Similar Documents

Publication Publication Date Title
EP2021507A2 (en) Compositions and methods for inhibiting expression of the pcsk9 gene
CN108064154B (en) Compositions and methods for inhibiting expression of the HAO1 (glycolate oxidase 1) gene
KR102031027B1 (en) Compositions and methods for inhibiting expression of transthyretin
CN107287202B (en) Angiopoietin-like 3(ANGPTL3) iRNA compositions and methods of use thereof
JP5704741B2 (en) Compositions and methods for suppression of Eg5 gene expression
JP5723378B2 (en) Lipid formulation composition and method for inhibiting transthyretin (TTR)
JP2019103501A (en) PCSK9 iRNA COMPOSITIONS AND METHODS OF USE THEREOF
JP5431940B2 (en) RNAi regulation of SCAP and its therapeutic use
CN112111492A (en) Compositions and methods for inhibiting expression of apolipoprotein C-III (APOC3) gene
KR20080106554A (en) Dsrna compositions and methods for treating hpv infection
AU2016203687A1 (en) Compositions and methods for inhibiting expression of the pcsk9 gene

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 200780024854.1

Country of ref document: CN

121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 07762085

Country of ref document: EP

Kind code of ref document: A2

WWE Wipo information: entry into national phase

Ref document number: 572666

Country of ref document: NZ

Ref document number: 2651839

Country of ref document: CA

Ref document number: 2007249329

Country of ref document: AU

Ref document number: 2009510173

Country of ref document: JP

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 6166/CHENP/2008

Country of ref document: IN

ENP Entry into the national phase

Ref document number: 2007249329

Country of ref document: AU

Date of ref document: 20070514

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 2007762085

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 1020087030164

Country of ref document: KR

Ref document number: 200870528

Country of ref document: EA

WWE Wipo information: entry into national phase

Ref document number: 1020107013424

Country of ref document: KR

WWE Wipo information: entry into national phase

Ref document number: 220102

Country of ref document: IL

Ref document number: 220103

Country of ref document: IL

WWE Wipo information: entry into national phase

Ref document number: 250678

Country of ref document: IL