WO2016011123A1 - Organic compositions to treat apoc3-related diseases - Google Patents

Organic compositions to treat apoc3-related diseases Download PDF

Info

Publication number
WO2016011123A1
WO2016011123A1 PCT/US2015/040517 US2015040517W WO2016011123A1 WO 2016011123 A1 WO2016011123 A1 WO 2016011123A1 US 2015040517 W US2015040517 W US 2015040517W WO 2016011123 A1 WO2016011123 A1 WO 2016011123A1
Authority
WO
WIPO (PCT)
Prior art keywords
strand
apoc3
rnai agent
sequence
modified
Prior art date
Application number
PCT/US2015/040517
Other languages
French (fr)
Inventor
Jan Weiler
William CHUTKOW
Jeremy Lee BARYZA
Andrew KRUEGER
Junping Zhao
Original Assignee
Arrowhead Research Corporation
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Arrowhead Research Corporation filed Critical Arrowhead Research Corporation
Priority to EP20178547.4A priority Critical patent/EP3736334A1/en
Priority to EP15822399.0A priority patent/EP3169784B1/en
Publication of WO2016011123A1 publication Critical patent/WO2016011123A1/en
Priority to US15/403,919 priority patent/US10240153B2/en
Priority to US16/269,282 priority patent/US11066667B2/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
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H21/00Compounds containing two or more mononucleotide units having separate phosphate or polyphosphate groups linked by saccharide radicals of nucleoside groups, e.g. nucleic acids
    • C07H21/02Compounds containing two or more mononucleotide units having separate phosphate or polyphosphate groups linked by saccharide radicals of nucleoside groups, e.g. nucleic acids with ribosyl as saccharide radical
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • 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
    • C12N2310/32Chemical 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
    • C12N2310/34Spatial arrangement of the modifications
    • C12N2310/341Gapmers, i.e. of the type ===---===
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/34Spatial arrangement of the modifications
    • C12N2310/344Position-specific modifications, e.g. on every purine, at the 3'-end
    • 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
    • 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
    • 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
    • C12N2320/00Applications; Uses
    • C12N2320/30Special therapeutic applications
    • C12N2320/31Combination therapy

Definitions

  • the present disclosure pertains to RNAi agents to Apolipoprotein III (APOC3).
  • the disclosure pertains to an APOC3 RNAi agent comprising a first and a second strand, wherein the sequence of the first and/or second strand is that of any sequence disclosed herein.
  • the disclosure pertains to APOC3 RNAi agents which comprise any sequence or an 18-nt portion of any APOC3 RNAi agent sequence disclosed herein (e.g., nt 1 -18 or nt 2-19 of any sequence disclosed herein). In various embodiments, these APOC3 RNAi agents are blunt-ended.
  • the disclosure pertains to APOC3 RNAi agents which comprise any sequence of at least 14 contiguous nt of any APOC3 RNAi agent sequence disclosed herein (e.g., nt 1-14, 2-15, 3-16, etc., of any sequence disclosed herein).
  • compositions comprising an APOC3 RNAi agent having any of various formats.
  • the APOC3 RNAi agent has the 18-mer format.
  • These APOC3 RNAi agents comprise a sense and an anti- sense strand, each strand being an 18-mer and the strands together forming a blunt-ended duplex, wherein the 3' end of at least one strand terminates in a phosphate or modified internucleoside linker and further comprises, in 5' to 3' order: a spacer; a second phosphate or modified internucleoside linker; and a 3' end cap.
  • the 3' end of both the sense and anti-sense strand terminate in a phosphate or modified internucleoside linker and further comprise, in 5' to 3' order: a spacer; a second phosphate or modified internucleoside linker; and a 3' end cap.
  • the disclosure pertains to a RNAi agent that comprises a first and a second strand, wherein the first and second strand are both 18-mers, and the first and second strand together form a blunt-ended duplex, and wherein the 3' end of the first strand terminates in a phosphate or modified internucleoside linker and further comprises, in 5' to 3' order: a spacer, a phosphate or modified
  • the disclosure pertains to a RNAi agent that comprises a first and a second strand, wherein the first and second strands are both 18-mers, and the first and second strand together form a blunt-ended duplex, and wherein the 3' end of both the first and second strand terminate in a phosphate or modified internucleoside linker and both further comprise a 3' end cap.
  • the APOC3 RNAi agent has a 19-mer format; in this format, both the first and second strand are 19-mers, wherein the 3' terminus of one or both strands further comprises a 3' terminal dinucleotide, spacer, and/or a 3' end cap.
  • one or both strands are longer than 19-mers (e.g., a 20-, 21-, 22-, 23-, 24-, 25-, 26-, 27-, 28-, 29-, or 30-mer), wherein one or both ends of the RNAi comprises an overhang, 3' terminal dinucleotide, spacer, and/or 3' end cap.
  • the APOC3 RNAi agent has an internal spacer format; e.g., one or more subunits [sugar+base] of the first or second strand is/are replaced by a spacer.
  • the 3' end of one or both strands further comprises another spacer and a 3' end cap.
  • the APOC3 RNAi agent has a shortened sense strand format; e.g., the sense strand is shortened (e.g., to a 18-, 17-, 16-, 15- or 14-mer).
  • the disclosure also pertains to any RNAi agent of any sequence, to any target which has a shortened sense strand format. In this format, the sense strand is shortened (to, for example, a 14-, 15-, 16-, or 17-mer), and the anti-sense strand is an 18-mer or longer.
  • the first strand is the antisense strand and the second strand is the sense strand. In other embodiments, the first strand is the sense strand and the second strand is the antisense strand.
  • the two strands can have the same or different spacers, phosphates or modified internucleoside linkers, and/or 3' end caps.
  • the strands can be ribonucleotides, or, optionally, one or more nucleotide can be modified or substituted.
  • At least one nucleotide comprises a modified internucleoside linker.
  • the RNAi agent can be modified on one or both 5' end.
  • the sense strand can comprise a 5' end cap which reduces the amount of the RNA interference mediated by this strand.
  • the RNAi agent is attached to a ligand. The disclosure also relates to processes for making such compositions, and methods and uses of such compositions, e.g., to mediate RNA interference.
  • Lipoproteins are globular, micelle-like particles that consist of a non-polar core of acylglycerols and cholesteryl esters surrounded by an amphiphilic coating of protein, phospholipid and cholesterol. Lipoproteins have been classified into five broad categories on the basis of their functional and physical properties: chylomicrons, which transport dietary lipids from intestine to tissues; very low density lipoproteins (VLDL); intermediate density lipoproteins (IDL); low density lipoproteins (LDL); all of which transport triacylglycerols and cholesterol from the liver to tissues; and high density lipoproteins (HDL), which transport endogenous cholesterol from tissues to the liver.
  • VLDL very low density lipoproteins
  • IDL intermediate density lipoproteins
  • LDL low density lipoproteins
  • HDL high density lipoproteins
  • Lipoprotein particles undergo continuous metabolic processing and have variable properties and compositions. Lipoprotein densities increase without decreasing particle diameter because the density of their outer coatings is less than that of the inner core.
  • the protein components of lipoproteins are known as apolipoproteins. At least nine apolipoproteins are distributed in significant amounts among the various human lipoproteins.
  • Apolipoprotein C-lll is a constituent of HDL and of triglyceride-rich lipoproteins and has a role in hypertriglyceridemia, a risk factor for coronary artery disease.
  • Apolipoprotein C- III slows this clearance of triglyceride-rich lipoproteins by inhibiting lipolysis, both through inhibition of lipoprotein lipase and by interfering with lipoprotein binding to the cell-surface glycosaminoglycan matrix (Shachter, Curr. Opin. Lipidol., 2001 , 12, 297-304).
  • apolipoprotein C-lll also called APOC3, APOC-III, APO Clll, and APO C-lll
  • the gene encoding human apolipoprotein C-lll was cloned in 1984 by three research groups (Levy-Wilson et al., DNA, 1984, 3, 359-364; Protter et al., DNA, 1984, 3, 449-456; Sharpe et al., Nucleic Acids Res., 1984, 12, 3917-3932).
  • the coding sequence is interrupted by three introns (Protter et al., DNA, 1984, 3, 449-456).
  • the human apolipoprotein C-lll gene is located approximately 2.6 kB to the 3' direction of the apolipoprotein A-1 gene and these two genes are convergently transcribed (Karathanasis, Proc. Natl. Acad. Sci. U.S.A., 1985, 82, 6374-6378). Also cloned was a variant of human apolipoprotein C-lll with a Thr74 to Ala74 mutation from a patient with unusually high level of serum apolipoprotein C-lll.
  • the Ala74 mutant therefore resulted in increased levels of serum apolipoprotein C-lll lacking the carbohydrate moiety (Maeda et al., J. Lipid Res., 1987, 28, 1405-1409).
  • the Sstl site distinguishes the S1 and S2 alleles and the S2 allele has been associated with elevated plasma triglyceride levels (Dammerman et al., Proc. Natl. Acad. Sci. U.S.A., 1993, 90, 4562-4566).
  • the apolipoprotein C-lll promoter is downregulated by insulin and this polymorphic site abolishes the insulin regulation.
  • the potential overexpression of apolipoprotein C-lll resulting from the loss of insulin regulation may be a contributing factor to the development of hypertriglyceridemia associated with the S2 allele (Li et al., J. Clin. Invest., 1995, 96, 2601-2605).
  • the T(-455) to C polymorphism has been associated with an increased risk of coronary artery disease (Olivieri et al., J. Lipid Res., 2002, 43, 1450-1457).
  • a response element for the nuclear orphan receptor rev-erb alpha has been located at positions -23/-18 in the apolipoprotein C-lll promoter region and rev-erb alpha decreases apolipoprotein C-lll promoter activity (Raspe et al., J. Lipid Res., 2002, 43, 2172- 2179).
  • the apolipoprotein C-lll promoter region -86 to -74 is recognized by two nuclear factors Clllbl and CIIIB2 (Ogami et al., J. Biol. Chem., 1991 , 266, 9640-9646).
  • Apolipoprotein C-lll expression is also upregulated by retinoids acting via the retinoid X receptor, and alterations in retinoid X receptor abundance affects apolipoprotein C-lll transcription (Vu-Dac et al., J. Clin. Invest., 1998, 102, 625-632).
  • Specificity protein 1 (Sp1 ) and hepatocyte nuclear factor-4 (HNF-4) have been shown to work synergistically to transactivate the apolipoprotein C-lll promoter via the HNF-4 binding site (Kardassis et al., Biochemistry, 2002, 41 , 1217-1228).
  • HNF-4 also works in conjunction with SMAD3-SMAD4 to transactivate the apolipoprotein C-lll promoter (Kardassis et al., J. Biol. Chem., 2000, 275, 41405-41414).
  • Transgenic and knockout mice have further defined the role of apolipoprotein C-lll in lipolysis. Overexpression of apolipoprotein C-lll in transgenic mice leads to
  • statin class of hypolipidemic drugs also lower triglyceride levels via an unknown mechanism, which results in increases in lipoprotein lipase mRNA and a decrease in plasma levels of apolipoprotein C-lll (Schoonjans et al., FEBS Lett., 1999, 452, 160-164). Consequently, there remains a long felt need for additional agents capable of effectively inhibiting apolipoprotein C-lll function.
  • RNAi agents to APOC3, for inhibition of APOC3, which are useful in treatment of an APOC3-related disease, such as obesity and metabolic-related disorders such as hyperlipidemia or hypertriglyceridemia (e.g., Type V Hypertriglyceridemia).
  • an APOC3-related disease such as obesity and metabolic-related disorders such as hyperlipidemia or hypertriglyceridemia (e.g., Type V Hypertriglyceridemia).
  • the present disclosure also encompasses a method for inhibiting and/or decreasing the expression and/or activity of APOC3 in a human subject.
  • the subject has an APOC3-related disease (a pathological state mediated at least in part by APOC3 over-expression or hyper-activity), and the method comprises the step of administering to the subject a therapeutically effective amount of a RNAi agent to APOC3.
  • the APOC3-related disease is selected from:
  • hypertriglyceridemia e.g., Type V Hypertriglyceridemia
  • abnormal lipid metabolism abnormal cholesterol metabolism
  • atherosclerosis hyperlipidemia
  • diabetes including Type 2 diabetes, obesity, cardiovascular disease, and coronary artery disease, among other disorders relating to abnormal metabolism or otherwise.
  • the present disclosure pertains to a RNAi agent to APOC3, comprising two strands, wherein the sequence of the first and/or second strand is, comprises, comprises 18 contiguous nt of, or comprises a sequence of 18 contiguous nt with 0-3 mismatches from, comprises 14 contiguous nt of, or comprises a sequence of 14 contiguous nt with 0-3 mismatches from: the sequence of any RNAi agent disclosed herein.
  • the disclosure pertains to an APOC3 RNAi agent comprising a first and second strand, wherein the sequence of the first and/or second strand is, comprises, or comprises 14 contiguous nt of any sequence of any APOC3 RNAi agent disclosed herein, except that one or both strands is nicked and/or one or more nt of the sequence disclosed herein has been replaced by a spacer (e.g., the first comprises the sequence of nt 1 -3 and 5- 14, nt 1 -5 and 7-18, or of 1 -4 and 6-18 of a sequence disclosed herein).
  • a spacer e.g., the first comprises the sequence of nt 1 -3 and 5- 14, nt 1 -5 and 7-18, or of 1 -4 and 6-18 of a sequence disclosed herein.
  • the RNAi agent to APOC3 comprises a first strand and a second strand, wherein the sequence of the first strand is, comprises, comprises 14 contiguous nt of, or comprises 14 contiguous nt of (with 1-3 mismatches from) any sequence of any APOC3 RNAi agent listed herein.
  • the present disclosure provides specific RNAi agents for inhibition of APOC3, and methods that are useful in reducing APOC3 levels in a subject, e.g., a mammal, such as a human.
  • the present disclosure specifically provides double-stranded RNAi agents comprising at least 14, 15, 16, 17, 18, or 19 or more contiguous nucleotides of APOC3.
  • the present disclosure provides agents comprising sequences of 14 or more contiguous nucleotides differing by 0, 1 , 2 or 3 from those of any of the RNAi agents provided, e.g., in any table herein, or otherwise disclosed herein.
  • RNAi agents particularly can in one aspect comprise less than 30 nucleotides per strand, e.g., such as 17- 23 nucleotides, 15-19, 18-22, and/or 19-21 nucleotides, and/or such as those provided, and modified and unmodified variants thereof (e.g., wherein the sense and/or anti-sense or first and/or second strand are modified or unmodified).
  • the present disclosure also provides RNAi agents to APOC3 comprising a sense strand and an anti-sense strand, wherein the sense and/or the anti-sense strand comprise sequences of 19 or more contiguous nucleotides differing by 0, 1 , 2 or 3 from those of the RNAi agents provided, and modified or unmodified variants thereof.
  • the present disclosure also provides RNAi agents to APOC3 having a shorted sense strand format. In this format, the sense strand is shortened (to, for example, a 14-, 15-, 16-, or 17-mer), and the anti-sense strand is an 18-mer or longer.
  • the disclosure also pertains to any RNAi agent of any sequence, to any target which has a shortened sense strand format.
  • the sense strand is shortened (to, for example, a 14-, 15-, 16-, or 17-mer), and the anti-sense strand is an 18-mer or longer.
  • the sense and anti-sense strand can be contiguous, or physically connected, e.g., by covalently bonds, a loop or linker.
  • the sense and/or anti- sense strand is discontinuous. Either strand can be nicked.
  • one or more nucleobase [sugar+base]) is replaced by a spacer.
  • the sense strand can be shortened (e.g., to a 14-, 15-, 16-, 17- or 18-mer), for example, in the shortened sense strand format.
  • the double-stranded RNAi agents can have 0, 1 or 2 blunt ends, and/or overhangs of 1 , 2, 3 or 4 nucleotides (i.e., 1 to 4 nt) from one or both 3' and/or 5' ends.
  • the double-stranded RNAi agents can also optionally comprise one or two 3' caps and/or one or more modified nucleotides. Modified variants of sequences as provided herein include those that are otherwise identical but contain substitutions of a naturally-occurring nucleotide for a corresponding modified nucleotide.
  • RNAi agent can either contain only nucleotides, e.g., naturally- occurring ribonucleotide subunits, or optionally one or more modifications to the sugar, phosphate or base of one or more of the substitute nucleotide subunits, whether they comprise ribonucleotide subunits or deoxyribonucleotide subunits.
  • modified variants of the disclosed RNAi agents have a thymidine (as RNA, or, preferably, DNA) replacing a uridine, or have an inosine base.
  • the modified variants of the disclosed RNAi agents can have a nick in the passenger strand, mismatches between the guide and passenger strand, DNA replacing the RNA of a portion of both the guide and passenger strand (e.g., the seed region of nt 2-7 counting from the 5' end of the anti-sense strand), and/or a shortened passenger (sense) strand (e.g., 14, 15, 16, 17 or 18 nt).
  • a functional guide strand is identified, modifications and variants of the RNAi agent can be readily made.
  • any two or more modifications which are not mutually exclusive can be combined (e.g., the combination of base modifications with shortened passenger strand; or nicked passenger strand and base modifications; or DNA replacing part or all of the seed region and base modifications in the remaining RNA; etc.).
  • modified variants of the disclosed RNAi agents include RNAi agents with the same sequence (e.g., the same sequence of bases) as any RNAi agent disclosed in any of the tables herein or otherwise disclosed herein. It is also noted that various authors have shown that once a successful RNAi agent is designed, generally adding a few nt to one or both strands does not impair RNA activity. Lengthening one or both strands, e.g, to 24 or 25 nt, does not impair RNA interference activity.
  • RNAi agent comprising a first strand and a second strand, wherein the sequence of the first and/or second strand comprises the sequence of any APOC3 RNAi agent disclosed herein, further comprising 1 -5 nt.
  • modified variants of the disclosed RNAi agents include RNAi agents with the same sequence (e.g., the same sequence of bases) as any RNAi agent disclosed in any of the tables herein or otherwise disclosed herein, but with one or more modifications to one or more of the sugar or phosphate of one or more of the nucleotide subunits.
  • the modifications improve efficacy, stability (e.g., against nucleases in, for example, blood serum or intestinal fluid), and/or reduce immunogenicity of the RNAi agent.
  • One aspect of the present disclosure relates to a double-stranded oligonucleotide comprising at least one non-natural nucleobase.
  • the non-natural nucleobase is difluorotolyl, nitroindolyl, nitropyrrolyl, or nitroimidazolyl. In a particular aspect, the non-natural nucleobase is difluorotolyl. In certain aspects, only one of the two oligonucleotide strands contains a non-natural nucleobase. In certain aspects, both of the oligonucleotide strands contain a non-natural nucleobase.
  • compositions comprising an APOC3 RNAi agent having the 18-mer format.
  • APOC3 RNAi agents comprise a sense and an anti-sense strand, each strand being an 18-mer and the strands together forming a blunt-ended duplex, wherein the 3' end of at least one strand terminates in a phosphate or modified internucleoside linker and further comprises, in 5' to 3' order: a spacer; a second phosphate or modified internucleoside linker; and a 3' end cap.
  • the 3' end of both the sense and anti-sense strand terminate in a phosphate or modified internucleoside linker and further comprise, in 5' to 3' order: a spacer; a second phosphate or modified internucleoside linker; and a 3' end cap.
  • the disclosure pertains to an APOC3 RNAi agent that comprises a first and a second strand, wherein the first and second strand are both 18-mers, and the first and second strand together form a blunt-ended duplex, and wherein the 3' end of the first strand terminates in a phosphate or modified internucleoside linker and further comprises, in 5' to 3' order: a spacer, a phosphate or modified internucleoside linker, and a 3' end cap; and wherein the 3' end of the second strand terminates in a phosphate or modified internucleoside linker and further comprises a 3' end cap.
  • the disclosure pertains to an APOC3 RNAi agent that comprises a first and a second strand, wherein the first and second strands are both 18-mers, and the first and second strand together form a blunt-ended duplex, and wherein the 3' end of both the first and second strand terminate in a phosphate or modified internucleoside linker and both further comprise a 3' end cap.
  • the first strand is the antisense strand and the second strand is the sense strand. In other embodiments, the first strand is the sense strand and the second strand is the antisense strand.
  • the two strands can have the same or different spacers, phosphates or modified internucleoside linkers, and/or 3' end caps.
  • the strands can be ribonucleotides, or, optionally, one or more nucleotide can be modified or substituted.
  • at least one nucleotide comprises a modified internucleoside linker.
  • the RNAi agent can be modified on one or both 5' end.
  • the sense strand can comprise a 5' end cap which reduces the amount of the RNA interference mediated by this strand.
  • the RNAi agent is attached to a ligand.
  • the disclosure also relates to processes for making such compositions, and methods and uses of such compositions, e.g., to mediate RNA interference.
  • the disclosure pertains to any RNAi agent of any sequence which has the 18-mer format or any format disclosed herein (e.g., internal spacer format or shortened sense strand format).
  • the disclosure pertains to an APOC3 RNAi agent comprising a sense strand and an anti-sense strand, wherein each strand is an 18-mer, wherein one or more nucleoside subunit ([sugar+base]) of the 18-mer anti-sense strand is replaced by a spacer, and wherein the 3' terminus of one or both strands further comprises: a 3' end cap, or a spacer and a 3' end cap.
  • the nucleobase subunit ([sugar+base]) replaced by a spacer is at position 1 , 3, 5, 6, 7, 15, 16, 17, or 18 (counting 5' to 3'). In various embodiments, the nucleobase subunit ([sugar+base]) replaced by a spacer is at position 5, 6, or 17 (counting 5' to 3').
  • the spacer can be sugar, alkyl, cycloakyl, ribitol or other type of abasic nucleotide, 2'-deoxy-ribitol, diribitol, 2'-methoxyethoxy-ribitol (ribitol with 2'-MOE), C3, C4, C5, C6, or 4-methoxybutane-1 ,3-diol (5300). More than one different type of spacer can be incorporated into the same composition.
  • the disclosure pertains to an RNAi agent to APOC3 having a shortened sense strand format.
  • the sense strand is shortened to a 14-, 15-, 16-, or 17-mer, and the anti-sense strand is a 18-mer or longer.
  • the sense strand can comprise, in 5' to 3' order, a 14-mer which terminates at the 3' end with a phosphate or modified internucleoside linker, a spacer, a second phosphate or modified internucleoside linker, and a 3' end cap.
  • the sense strand can comprise, in 5' to 3' order, a 15-mer which terminates at the 3' end with a phosphate or modified internucleoside linker, a spacer, a second phosphate or modified internucleoside linker, and a 3' end cap. In some embodiments, the sense strand can comprise, in 5' to 3' order, a 16-mer which terminates at the 3' end with a phosphate or modified internucleoside linker, a spacer, a second phosphate or modified internucleoside linker, and a 3' end cap.
  • the sense strand can comprise, in 5' to 3' order, a 17-mer which terminates at the 3' end with a phosphate or modified internucleoside linker, a spacer, a second phosphate or modified internucleoside linker, and a 3' end cap.
  • the 14-, 15-, 16-, or 17-mer can comprise RNA, wherein one or more RNA subunits (nucleotides) has been modified or substituted.
  • the last 2 nucleotides on the 3' end of the anti-sense and sense strand are 2'-MOE (a 2'-MOE clamp). In various embodiments, with or without the 2'-MOE clamp, one or more
  • nucleotides can be modified with a 2'-MOE, 2'-OMe, 2'-F, or substituted with DNA, a peptide nucleic acid (PNA), locked nucleic acid (LNA), morpholino nucleotide, threose nucleic acid (TNA), glycol nucleic acid (GNA), arabinose nucleic acid (ANA), 2 ' -fluoroarabinose nucleic acid (FANA), cyclohexene nucleic acid (CeNA), anhydrohexitol nucleic acid (HNA), unlocked nucleic acid (UNA).
  • PNA peptide nucleic acid
  • LNA locked nucleic acid
  • TAA threose nucleic acid
  • GAA glycol nucleic acid
  • ANA arabinose nucleic acid
  • FANA 2 ' -fluoroarabinose nucleic acid
  • CeNA cyclohexene nucleic acid
  • HNA un
  • the spacer can be a sugar, alkyl, cycloakyi, ribitol or other type of abasic nucleotide, 2'-deoxy-ribitol, diribitol, 2'-methoxyethoxy-ribitol (ribitol with 2'-MOE), C3, C4, C5, C6, or 4-methoxybutane-1 ,3-diol (5300).
  • the 3' end cap can be selected from any of various 3' end caps described herein or known in the art.
  • the 3' end cap can be a PAZ ligand (e.g., X058 or X2).
  • the shortened sense strand format is suitable for use with APOC3 RNAi agents, as disclosed herein, or with any RNAi agent of any sequence directed to any gene target.
  • the RNAi agent(s) can optionally be attached to a ligand selected to improve one or more characteristic, such as, e.g., stability, distribution and/or cellular uptake of the agent, e.g., cholesterol or a derivative thereof.
  • the RNAi agent(s) can be isolated or be part of a pharmaceutical composition used for the methods described herein.
  • the pharmaceutical composition can be formulated for delivery to specific tissues (e.g., those afflicted with an APOC3-related disease) or formulated for parenteral administration.
  • the pharmaceutical composition can optionally comprise two or more RNAi agents, each one directed to the same, overlapping or a different segment of the APOC3 mRNA.
  • the pharmaceutical composition can further comprise or be used in conjunction with any known treatment for any APOC3-related disease.
  • Second agent or treatment The method also optionally further comprises the step of administering a second agent or treatment.
  • this second agent is another RNAi agent to APOC3.
  • the second agent or treatment is iron administration, chelation therapy, phlebotomy, erythropoiesis stimulating agent (ESA) (e.g., Epoetin alfa or darbepoetin alfa), anti-APOC3 antibody, hemodialysis, or hyperbaric oxygen.
  • ESA erythropoiesis stimulating agent
  • the second agent or treatment is directed to another target, which is also hyper-active, mutated and/or over-expressed in the pathological state.
  • the present disclosure further provides methods for reducing the level of APOC3 mRNA in a cell, particularly in the case of a disease characterized by over- expression or hyper-activity of APOC3.
  • Cells comprising an alteration such as a mutation, over-expression and/or hyperactivity of APOC3 are termed "APOC3-defective" cells.
  • Such methods comprise the step of administering one or more of the RNAi agents of the present disclosure to an APOC3-defective cell, as further described below.
  • the present methods utilize the cellular mechanisms involved in RNA interference to selectively degrade the target RNA in a cell and are comprised of the step of contacting a cell with one of the RNAi agents of the present disclosure.
  • the present disclosure also encompasses a method of treating a human subject having a pathological state mediated at least in part by APOC3 over-expression or hyperactivity, the method comprising the step of administering to the subject a therapeutically effective amount of a RNAi agent APOC3. Additional methods involve preventing, treating, modulating and/or ameliorating a pathological state wherein disease progression requires APOC3, although APOC3 is not amplified or over-expressed. Such methods comprise the step of administering one of the RNAi agents of the present disclosure to a subject, as further described below.
  • Such methods can be performed directly on a cell or can be performed on a mammalian subject by administering to a subject one of the RNAi agents/pharmaceutical compositions of the present disclosure.
  • Reduction of target APOC3 mRNA in a cell results in a reduction in the amount of encoded APOC3 protein produced. In an organism, this can result in restoration of balance in a pathway involving APOC3, and/or prevention of APOC3 accumulation, and/or a reduction in APOC3 activity and/or expression, and/or prevention of APOC3-mediated activation of other genes, and/or amelioration, treatment and/or prevention of an APOC3-related disease.
  • a reduction in APOC3 expression, level or activity can limit disease growth.
  • compositions of the present disclosure can be used in any appropriate dosage and/or formulation described herein or known in the art, as well as with any suitable route of administration described herein or known in the art.
  • any combination of modifications, 5' end caps, and/or 3' end caps can be used with any RNAi agent sequence disclosed herein.
  • Any RNAi agent disclosed herein (with any combination of modifications or endcaps or without either modifications or endcaps) can be combined with any other RNAi agent or other treatment composition or method disclosed herein.
  • FIG. 1 Various anti-ApoC3 RNAi agents.
  • FIG. 2 Various anti-ApoC3 RNAi agents.
  • FIG. 3B Various anti -ApoC3 RNAi agents.
  • FIG. 3B Various anti -ApoC3 RNAi agents.
  • FIG. 4A Various anti -ApoC3 RNAi agents.
  • FIG. 4B Various anti -ApoC3 RNAi agents.
  • FIG. 5A Various anti -ApoC3 RNAi agents.
  • FIG. 5B Various anti -ApoC3 RNAi agents.
  • FIG. 6 Various anti-ApoC3 RNAi agents.
  • FIG. 8 Structure representing X058 PAZ ligand.
  • FIG. 9 Structure representing X003.
  • RNAi agents to APOC3, for targeting and inhibition of APOC3, which are useful in treatment of APOC3-related diseases (e.g., diseases associated with mutations in and/or altered expression, level and/or activity of APOC3, diseases requiring APOC3, diseases affected by a factor whose expression, over- expression, or hyper-activity is directly or indirectly affected by APOC3, and/or diseases treatable by modulating the expression, level and/or activity of APOC3).
  • APOC3-related diseases include those listed herein or known in the art.
  • the present disclosure also provides methods of treating a human subject having a pathological state mediated at least in part by APOC3 over-expression or hyper-activity, or requiring APOC3, the method comprising the step of administering to the subject a therapeutically effective amount of a RNAi agent to APOC3.
  • an RNAi agent comprising an antisense strand of an RNAi agent described herein.
  • an aspect of the present disclosure relates to a composition comprising an RNAi agent comprising an antisense strand, wherein the antisense strand is, comprises, comprises at least 14 contiguous nucleotides (nt) or, or comprises at least 14 contiguous nucleotides differing by 0, 1 , 2, or 3 nucleotides from the antisense strand of an RNAi agent to APOC3 selected from any sequence provided herein (e.g., in any of the tables herein).
  • the disclosure pertains to an APOC3 RNAi agent comprising a first and second strand, wherein the sequence of the first and/or second strand is, comprises, or comprises 14 contiguous nt of any sequence of any APOC3 RNAi agent disclosed herein, wherein one or both strands is nicked and/or one or more nt of the sequence disclosed herein has been replaced by a spacer (e.g., the first comprises the sequence of nt 1 -5 and 7-18, or of 1 -4 and 6-18 of a sequence disclosed herein).
  • the present disclosure relates to a composition
  • a composition comprising an RNAi agent comprising a first strand and a second strand, wherein the first strand comprises at least 14 contiguous nucleotides differing by 0, 1 , 2, or 3 nucleotides from the first strand of an RNAi agent to APOC3 from any sequence provided herein.
  • the present disclosure relates to a composition
  • a composition comprising an RNAi agent comprising a sense strand and an antisense strand, wherein the sense strand comprises at least 14 contiguous nucleotides differing by 0, 1 , 2, or 3 nucleotides from the sense strand and the antisense strand comprises at least 14 contiguous nucleotides differing by 0, 1 , 2, or 3 nucleotides from the antisense strand of an RNAi agent to APOC3 listed immediately above.
  • one or both strands of the RNAi agent is an 18-mer.
  • both strands are 18-mers and together they form a blunt-ended duplex.
  • the present disclosure relates to a composition comprising an RNAi agent comprising a first strand and a second strand, wherein the sequence of the first strand comprises the sequence of the first strand of any sequence provided herein.
  • the present disclosure relates to a composition comprising an RNAi agent comprising a first strand and a second strand, wherein the sequence of the first strand is the sequence of the first strand of any sequence provided herein.
  • the first and second strands are the anti- sense and sense strand, respectively, of any RNAi agent disclosed herein.
  • the first and second strands are the sense and anti-sense strand, respectively, of any RNAi agent disclosed herein.
  • compositions comprising an APOC3 RNAi agent having a novel format (the "18-mer format").
  • APOC3 RNAi agents comprise a sense and an anti-sense strand, each strand being an 18-mer and the strands together forming a blunt-ended duplex, wherein the 3' end of at least one strand terminates in a phosphate or modified internudeoside linker and further comprises, in 5' to 3' order: a spacer; a second phosphate or modified internudeoside linker; and a 3' end cap.
  • the 3' end of both the sense and anti-sense strand terminate in a phosphate or modified internucleoside linker and further comprise, in 5' to 3' order: a spacer; a second phosphate or modified internucleoside linker; and a 3' end cap.
  • the disclosure pertains to an APOC3 RNAi agent that comprises a first and a second strand, wherein the first and second strand are both 18-mers, and the first and second strand together form a blunt-ended duplex, and wherein the 3' end of the first strand terminates in a phosphate or modified internucleoside linker and further comprises, in 5' to 3' order: a spacer, a phosphate or modified internucleoside linker, and a 3' end cap; and wherein the 3' end of the second strand terminates in a phosphate or modified internucleoside linker and further comprises a 3' end cap.
  • the disclosure pertains to an APOC3 RNAi agent that comprises a first and a second strand, wherein the first and second strands are both 18-mers, and the first and second strand together form a blunt-ended duplex, and wherein the 3' end of both the first and second strand terminate in a phosphate or modified internucleoside linker and both further comprise a 3' end cap.
  • the APOC3 RNAi agent has a 19-mer format; in this format, both the first and second strand are 19-mers, wherein the 3' terminus of one or both strands further comprises a 3' terminal dinucleotide, spacer, and/or a 3' end cap.
  • one or both strands are longer than 19-mers (e.g., a 20-, 21 -, 22-, 23-, 24-, 25-, 26-, 27-, 28-, 29-, or 30-mer), wherein one or both ends of the RNAi comprises an overhang, 3' terminal dinucleotide, spacer, and/or 3' end cap.
  • the APOC3 RNAi agent has an internal spacer format; e.g., one or more subunits [sugar+base] of the first or second strand is/are replaced by a spacer.
  • the 3' end of one or both strands further comprises another spacer and a 3' end cap.
  • the APOC3 RNAi agent has a shortened sense strand format; e.g., the sense strand is shortened (e.g., to a 18-, 17-, 16-, 15- or 14-mer).
  • the first strand is the antisense strand and the second strand is the sense strand.
  • the first strand is the sense strand and the second strand is the antisense strand.
  • the two strands can have the same or different spacers, phosphates or modified internucleoside linkers, and/or 3' end caps.
  • the strands can be ribonucleotides, or, optionally, one or more nucleotide can be modified or substituted.
  • at least one nucleotide comprises a modified internucleoside linker.
  • the RNAi agent can be modified on one or both 5' end.
  • the sense strand can comprise a 5' end cap which reduces the amount of the RNA interference mediated by this strand.
  • the RNAi agent is attached to a ligand.
  • compositions comprising an APOC3 RNAi agent conjugated to GalNAc.
  • N-Acetylgalactosamine also known as GalNAc
  • GalNAc is an amino sugar derivative of galactose.
  • GalNAc is necessary for intercellular communication, and is concentrated in sensory nerve structures. It is typically the first monosaccharide that connects serine or threonine in particular forms of protein O-glycosylation. In humans it is the terminal carbohydrate forming the antigen of blood group A.
  • duplexes include the unmodified (e.g., "generic") and example modified variants listed in the tables herein and otherwise disclosed herein; additional sequences and data for these RNAi agents are presented in the subsequent Tables.
  • other modified variants can be made using the nucleotide sequences provided.
  • underline nucleotide cyno/human mismatch
  • n (lower case) 2'-0-Methyl nucleotide
  • Nm 2 -MOE nucleotide
  • Nf 2'-fluoro nucleotide
  • dN DNA
  • X1 abasic site with ribitol spacer
  • X2 abasic site with 1 ,6-hexane- diol spacer
  • X058 3' end cap
  • One or both strands of the 18-mer format can comprise, in various embodiments, in 5' to 3' order, a 18-mer, a spacer, and a 3' end cap.
  • the 18-mer can be modified or unmodified.
  • the modifications include the two nt on the 3' end being 2'-MOE (a "2-MOE clamp").
  • nucleotides can be modified with a 2'-MOE, 2'-OMe, 2'-F, or substituted with DNA, a peptide nucleic acid (PNA), locked nucleic acid (LNA), morpholino nucleotide, threose nucleic acid (TNA), glycol nucleic acid (GNA), arabinose nucleic acid (ANA), 2 - fluoroarabinose nucleic acid (FANA), cyclohexene nucleic acid (CeNA), anhydrohexitol nucleic acid (HNA), unlocked nucleic acid (UNA).
  • PNA peptide nucleic acid
  • LNA locked nucleic acid
  • TAA threose nucleic acid
  • GAA glycol nucleic acid
  • ANA arabinose nucleic acid
  • FANA 2 - fluoroarabinose nucleic acid
  • CeNA cyclohexene nucleic acid
  • HNA unlocked nucleic
  • the spacer can be a sugar, alkyl, cycloakyl, ribitol or other type of abasic nucleotide, 2'-deoxy-ribitol, diribitol, 2'- methoxyethoxy-ribitol (ribitol with 2 -MOE), C3, C4, C5, C6, or 4-methoxybutane-1 ,3-diol (5300).
  • the 3' end cap can be selected from any of various 3' end caps described herein or known in the art.
  • At least one nucleotide of the RNAi agent is modified.
  • said at least one modified nucleotide is selected from among 2' alkoxyribonucleotide, 2' alkoxyalkoxy ribonucleotide, or 2'-fluoro ribonucleotide.
  • said at least one modified nucleotide is selected from 2'-OMe, 2'-MOE and 2'-H.
  • the nucleotide subunit is chemically modified at the 2' position of the sugar.
  • the 2' chemical modification is selected from a halo, a C1 -10 alkyl, a C1-10 alkoxy, a halo, and the like.
  • the 2' chemical modification is a C1 -10 alkoxy selected from -OCH 3 (i.e., "OMe”), -OCH 2 CH 3 (i.e., "OEt") or -CH 2 OCH 2 CH 3 (i.e., methoxyethyl or "MOE”); or is a halo selected from F.
  • one of more nucleotides is DNA or is replaced by a peptide nucleic acid (PNA), locked nucleic acid (LNA), morpholino nucleotide, threose nucleic acid (TNA), glycol nucleic acid (GNA), arabinose nucleic acid (ANA), 2 - fluoroarabinose nucleic acid (FANA), cyclohexene nucleic acid (CeNA), anhydrohexitol nucleic acid (HNA), and/or unlocked nucleic acid (UNA); and/or at least one nucleotide comprises a modified internucleoside linker (e.g., wherein at least one phosphate of a nucleotide is replaced by a modified internucleoside linker), wherein the modified internucleoside linker is selected from phosphorothioate, phosphorodithioate,
  • the first two base-pairing nucleotides on the 3' end of the first and/or second strand are modified.
  • the first two base-pairing nucleotides on the 3' end of the first and/or second strand are 2'-MOE.
  • lower-case letters e.g., c, u
  • upper case letters e.g., C, U, A, G
  • example modified versions of each of the sequences are shown.
  • present disclosure also contemplates and encompasses unmodified versions of these sequences and other versions which comprise additional or alternative
  • the modified and unmodified variants can optionally further comprise the sequence "TT", "dTdT”, “dTsdT” or "UU” as a single-stranded overhang at the 3' end, also termed herein a terminal dinudeotide or 3' terminal dinudeotide.
  • dT is 2'-deoxy-thymidine-5'-phosphate
  • sdT is 2'-deoxy Thymidine 5'-phosphorothioate.
  • terminal dinudeotide "UU” is UU or 2'-OMe-U 2'-OMe-U, and the terminal TT and the terminal UU can be in the inverted/reverse orientation.
  • the terminal dithymidine (e.g., UU) is not part of the APOC3 target sequence, but is a modified variant of the dithymidine dinudeotide commonly placed as an overhang to protect the ends of siRNAs from nucleases (see, for example, Elbashir et al. 2001 Nature 41 1 : 494-498; Elbashir et al. 2001 EMBO J. 20: 6877-6888; and Kraynack et al. 2006 RNA 12:163-176).
  • a terminal dinudeotide is known from these references to enhance nuclease resistance but not contribute to target recognition.
  • the present disclosure also encompasses any modified or any unmodified variant disclosed herein, wherein the modified variant comprises a terminal TT, dTdT, sdT, dTsdT, sdTsdT, sdTdT, or the like which may be in either the inverted/reverse orientation or in the same 5' to 3' orientation as the APOC3 specific sequence in the duplex.
  • the APOC3 portion of a RNAi agent sequence and the like indicate the portion of the sequence of a RNAi agent which is derived from APOC3 (thus "the APOC3 portion of a RNAi agent sequence” does not include, for example, a terminal dTdT, TT, UU, U (2'-OMe) dT, U (2 - OMe) U (2'-OMe), T(2'-OMe) T (2'-OMe), T(2'-OMe) dT, or the like, but does include the portion of the RNAi agent that corresponds to or is complementary to a portion of the RNAi agent that corresponds to or is complementary to a portion of the RNAi agent.
  • the composition comprises a RNAi agent comprising a first and a second strand, wherein the sequence of the first strand and the sequence of the second strand are the sequences of the first and second strand, respectively, of any RNAi agent provided herein, further comprising a 3' terminal dinucleotide (a single-stranded overhang comprising 2 nt at the 3' end).
  • the composition comprises a RNAi agent comprising a first and a second strand, wherein the sequence of the first strand and the sequence of the second strand are the sequences of the first and second strand, respectively, of any RNAi agent provided herein, further comprising a 3' terminal dinucleotide selected from TT, UU, U (2'-OMe) dT, U (2'-OMe) U (2'-OMe), T(2'- OMe) T (2'-OMe), T(2'-OMe) dT, dTdT, sdT, dTsdT, sdTsdT, and sdTdT.
  • TT 3' terminal dinucleotide selected from TT, UU, U (2'-OMe) dT, U (2'-OMe) U (2'-OMe), T(2'- OMe) T (2'-OMe), T(2'-OMe) dT, dTdT, sd
  • the composition comprises a RNAi agent comprising a first and a second strand, wherein the sequence of the first strand and the sequence of the second strand are the sequences of the first and second strand, respectively, of any RNAi agent provided herein, further comprising a 3' terminal UU dinucleotide.
  • a 3' end cap can be used instead of or in addition to a terminal dinucleotide to stabilize the end from nuclease degradation provided that the 3' end cap is able to both stablize the RNAi agent (e.g., against nucleases) and not interfere excessively with siRNA activity.
  • the present disclosure also encompasses any modified or any unmodified variant disclosed herein, wherein the modified variant further comprises a terminal 3' end cap.
  • RNAi agent comprising an antisense strand of an RNAi agent described herein.
  • the present disclosure relates to a composition
  • a composition comprising an RNAi agent comprising an antisense strand, wherein the antisense strand comprises at least 14 contiguous nucleotides differing by 0, 1 , 2, or 3 nucleotides from the antisense strand of an RNAi agent to APOC3 selected from those antisense strands in the specific duplexes provided herein.
  • the composition further comprises a second RNAi agent to APOC3.
  • the second RNAi agent is physically separate from the first, or the two are physically connected (e.g., covalently linked or otherwise conjugated).
  • the composition comprises a RNAi agent to APOC3 comprising a first and a second strand, wherein the sequence of the first strand and the sequence of the second strand are the sequences of the first and second strand, respectively, of any RNAi agent provided herein.
  • the composition comprises a RNAi agent comprising a first and a second strand, wherein the sequence of the first strand and the sequence of the second strand are the sequences of the first and second strand, respectively, of any RNAi agent provided herein, further comprising an additional about 6 to 20 nucleotides on one or both strands (e.g., about 6, 7, 8, 9, 10, 1 1 , 12, 13, 14, 15, 16, 17, 18, 19 or 20 nt).
  • the composition comprises a RNAi agent comprising a first and a second strand, wherein the sequence of the first strand and the sequence of the second strand are the sequences of the first and second strand, respectively, of any RNAi agent provided herein, further comprising a 3' terminal dinucleotide.
  • the composition comprises a RNAi agent comprising a first and a second strand, wherein the sequence of the first strand and the sequence of the second strand are the sequences of the first and second strand, respectively, of any RNAi agent provided herein, further comprising a 3' terminal dinucleotide selected from TT, UU, U (2 - OMe) dT, U (2'-OMe) U (2'-OMe), T(2'-OMe) T (2'-OMe), T(2'-OMe) dT, dTdT, sdT, dTsdT, sdTsdT, and sdTdT.
  • TT 3' terminal dinucleotide selected from TT, UU, U (2 - OMe) dT, U (2'-OMe) U (2'-OMe), T(2'-OMe) T (2'-OMe), T(2'-OMe) dT, dTdT, sd
  • the composition comprises a RNAi agent comprising a first and a second strand, wherein the sequence of the first strand and the sequence of the second strand are the sequences of the first and second strand, respectively, of any RNAi agent provided herein, further comprising a 3' terminal UU dinucleotide.
  • the first and second strands are the sense and anti-sense strands listed herein, respectively.
  • the first and second strands are the anti-sense and sense strands listed herein, respectively.
  • the sense strand is about 30 or fewer nucleotides (nt) in length.
  • the antisense strand is about 30 or fewer nucleotides in length.
  • the antisense strand forms a duplex region with a sense strand, wherein the duplex region is about 15 to 30 nucleotide pairs in length.
  • the antisense strand is about 15 to about 30 nucleotides in length, including 18, about 18, and about 19 to about 23 nucleotides in length. In one aspect, the antisense strand has at least the length selected from about 15 nucleotides, about 16 nucleotides, about 17 nucleotides, 18 nucleotides, about 18 nucleotides, about 19 nucleotides, about 20 nucleotides, about 21 nucleotides, about 22 nucleotides, about 23 nucleotides, about 24 nucleotides, about 25 nucleotides, about 26 nucleotides, about 27 nucleotides, about 28 nucleotides, about 29 nucleotides and 30 nucleotides. In various aspects, the anti-sense strand is a 17-, 18-, 19-, 20-, 21 -, 22-, 23-, 24-, 25-, 26-, 27-, 28-, 29- or 30-mer.
  • the sense strand can be shorter than the anti-sense strand.
  • the sense strand is a 14-, 15-, 16-, 17-, 18-, 19-, 20-, 21 -, 22-, 23-, 24-, 25-, 26-, 27-, 28-, 29- or 30-mer.
  • the RNAi agent comprises a modification that causes the RNAi agent to have increased stability in a biological sample or environment (e.g., cytoplasm, interstitial fluid, blood serum, or lung or intestinal lavage).
  • a biological sample or environment e.g., cytoplasm, interstitial fluid, blood serum, or lung or intestinal lavage.
  • the RNAi agent comprises at least one sugar backbone modification (e.g., phosphorothioate linkage) or at least one 2'-modified nucleotide.
  • the RNAi agent comprises: at least one 5'-uridine-adenine-3' (5'-ua- 3') dinucleotide, wherein the uridine is a 2'-modified nucleotide; at least one 5'-uridine- guanine-3' (5'-ug-3') dinucleotide, wherein the 5'-uridine is a 2'-modified nucleotide; at least one 5'-cytidine-adenine-3' (5'-ca-3') dinucleotide, wherein the 5'-cytidine is a 2'-modified nucleotide; or at least one 5'-uridine-uridine-3' (5'-uu-3') dinucleotide, wherein the 5'-uridine is a 2'-modified nucleotide.
  • dinucleotide motifs are particularly prone to serum nuclease degradation (e.g. RNase A).
  • Chemical modification at the 2'-position of the first pyrimidine nucleotide in the motif prevents or slows down such cleavage.
  • This modification recipe is also known under the term 'endo light'.
  • the RNAi agent comprises a 2'-modification selected from the group consisting of: 2'-deoxy, 2'-deoxy-2'-fluoro, 2'-0-methyl (2'-OMe or 2'OMe), 2 -0- methoxyethyl (2 -O-MOE), 2'-0-aminopropyl (2 -O-AP), 2'-0-dimethylaminoethyl (2 -0-
  • DMAOE 2'-0-dimethylaminopropyl (2'-0-DMAP), 2'-0-dimethylaminoethyloxyethyl (2'-0-
  • DMAEOE 2'-0-N-methylacetamido (2'-0-NMA).
  • all pyrimidines uridine and cytidine are 2' O-methyl-modified nucleosides.
  • the RNAi agent comprises at least one blunt end.
  • the RNAi agent comprises an overhang having 1 nt to 4 nt unpaired.
  • the RNAi agent comprises an overhang at the 3'-end of the antisense strand of the RNAi agent.
  • the RNAi agent is ligated to one or more diagnostic compound, reporter group, cross-linking agent, nuclease-resistance conferring moiety, natural or unusual nucleobase, lipophilic molecule, cholesterol, lipid, lectin, steroid, uvaol, hecigenin, diosgenin, terpene, triterpene, sarsasapogenin, Friedelin, epifriedelanol-derivatized lithocholic acid, vitamin, carbohydrate, dextran, pullulan, chitin, chitosan, synthetic carbohydrate, oligo lactate 15-mer, natural polymer, low- or medium-molecular weight polymer, inulin, cyclodextrin, hyaluronic acid, protein, protein-binding agent, integrin- targeting molecule, polycationic, peptide, polyamine, peptide mimic, and/or transferrin.
  • the RNAi agent is capable of inhibiting expression of APOC3 by at least about 50% in Huh-7 cells in nude mice.
  • the RNAi agent is capable of inhibiting expression of APOC3 by at least about 70% at a concentration of 10 nM in Huh-7 cells in vitro.
  • the RNAi agent is capable of inhibiting expression of APOC3 by at least about 75% at a concentration of 10 nM in Huh-7 cells in vitro. [0090] In one aspect, the RNAi agent is capable of inhibiting expression of APOC3 by at least about 80% at a concentration of 10 nM in Huh-7 cells in vitro.
  • the RNAi agent is capable of inhibiting expression of APOC3 by at least about 90% at a concentration of 10 nM in Huh-7 cells in vitro.
  • the RNAi agent is capable of inhibiting expression of APOC3 by at least about 95% at a concentration of 10 nM in Huh-7 cells in vitro.
  • the RNAi agent is capable of inhibiting expression of APOC3 by at least about 99% at a concentration of 10 nM in Huh-7 cells in vitro.
  • the RNAi has an EC50 of no more than about 0.1 nM in Huh-7 cells in vitro. EC50 is effective concentration to reduce gene expression by 50%.
  • the RNAi has an EC50 of no more than about 0.01 nM in Huh-7 cells in vitro.
  • the RNAi has an EC50 of no more than about 0.001 nM in Huh-7 cells in vitro.
  • RNAi agent comprising a sense and antisense strand of an RNAi described herein.
  • the present disclosure relates to a composition
  • a composition comprising a RNAi agent comprising a first strand and a second strand, wherein the sequence of the first strand and/or second strand comprise at least 14 contiguous nucleotides, differing by 0, 1 , 2, or 3 nucleotides from the sequence of the first and/or second strand of a RNAi agent to APOC3 selected from the specific duplexes provided herein.
  • the present disclosure relates to a composition
  • a composition comprising a RNAi agent comprising a first strand and a second strand, wherein the sequence of the first strand and/or second strand comprise the sequence of the first and/or second strand, respectively, of a RNAi agent to APOC3 selected from the specific duplexes provided herein.
  • the present disclosure relates to a composition
  • a composition comprising a RNAi agent comprising a first strand and a second strand, wherein the sequence of the first strand and/or second strand are the sequence of the first and/or second strand, respectively, of a RNAi agent to APOC3 selected from the specific duplexes provided herein.
  • the present disclosure relates to a composition
  • a composition comprising a RNAi agent comprising a first strand and a second strand, wherein the sequence of the first strand and/or second strand are the sequence of the first and/or second strand, respectively, of a RNAi agent to APOC3 selected from the specific duplexes provided herein, wherein the sequence of the first and/or second strand further comprise a terminal dinucleotide.
  • the present disclosure relates to a composition
  • a composition comprising a RNAi agent comprising a first strand and a second strand, wherein the sequence of the first strand and/or second strand are the sequence of the first and/or second strand, respectively, of a RNAi agent to APOC3 selected from the specific duplexes provided herein, wherein the sequence of the first and/or second strand further comprise a terminal UU dinucleotide.
  • the present disclosure relates to a composition
  • a composition comprising an RNAi agent comprising a sense strand and an antisense strand, wherein the sense strand and antisense strand comprise at least 14 contiguous nucleotides differing by 0, 1 , 2, or 3 nucleotides, from the sense and antisense strand, respectively, of an RNAi agent to APOC3 selected from the specific duplexes provided herein.
  • the composition comprises a second RNAi agent to APOC3.
  • the second RNAi agent is physically separate from the first, or the two are physically connected (e.g., chemically linked or otherwise conjugated).
  • the first and second RNAi agents are combined within the same composition (e.g., both in the same lipid nanoparticle).
  • the antisense strand is about 30 or fewer nucleotides in length.
  • the sense strand and the antisense strand form a duplex region about 15 to about 30 nucleotide pairs in length.
  • the antisense strand is about 15 to about 36 nt in length including about 18 to about 23 nt in length, and including about 19 to about 21 nt in length and about 19 to about 23 nt in length.
  • the antisense strand has at least the length selected from about 15 nt, about 16 nt, about 17 nt, 18, about 18 nt, about 19 nt, about 20 nt, about 21 nt, about 22 nt, about 23 nt, about 24 nt, about 25 nt, about 26 nt, about 27 nt, about 28 nt, about 29 nt and about 30 nt.
  • the anti-sense strand is a 17-, 18-, 19-, 20-, 21-, 22-, 23-, 24-, 25-, 26-, 27-, 28-, 29- or 30-mer.
  • the sense strand can be shorter than the anti-sense strand.
  • the sense strand is a 14-, 15-, 16-, 17-, 18-, 19-, 20-, 21 -, 22-, 23-, 24-, 25-, 26-, 27-, 28-, 29- or 30-mer.
  • the RNAi agent comprises a modification that causes the RNAi agent to have increased stability in a biological sample or environment.
  • the RNAi agent comprises a modified sugar backbone such as, e.g., a phosphorothioate linkage, or comprises a 2'-modified nucleotide.
  • the RNAi agent comprises: at least one 5'-uridine-adenine-3' (5'-ua-3') dinucleotide, wherein the uridine is a 2'-modified nucleotide; at least one 5'-uridine- guanine-3' (5'-ug-3') dinucleotide, wherein the 5'-uridine is a 2'-modified nucleotide; at least one 5'-cytidine-adenine-3' (5'-ca-3') dinucleotide, wherein the 5'-cytidine is a 2'-modified nucleotide; or at least one 5'-uridine-uridine-3' (5'-uu-3') dinucleotide,
  • the RNAi agent comprises a 2'-modification selected from the group consisting of: 2'-deoxy, 2'-deoxy-2'-fluoro, 2'-0-methyl, 2'-0-methoxyethyl (2 -O-MOE), 2'-0-aminopropyl (2 -O-AP), 2'-0-dimethylaminoethyl (2 -O-DMAOE), 2 -0- dimethylaminopropyl (2 -O-DMAP), 2'-0-dimethylaminoethyloxyethyl (2'-0-DMAEOE), and 2'-0-N-methylacetamido (2'-0-NMA).
  • all pyrimidines are 2' O-methyl-modified nucleosides.
  • the RNAi agent comprises at least one blunt end.
  • the RNAi agent comprises an overhang having 1 to 4 nt unpaired.
  • the RNAi agent comprises an overhang at the 3'-end of the antisense strand of the RNAi agent.
  • the RNAi agent is ligated to one or more diagnostic compound, reporter group, cross-linking agent, nuclease-resistance conferring moiety, natural or unusual nucleobase, lipophilic molecule, cholesterol, lipid, lectin, steroid, uvaol, hecigenin, diosgenin, terpene, triterpene, sarsasapogenin, Friedelin, epifriedelanol-derivatized lithocholic acid, vitamin, carbohydrate, dextran, pullulan, chitin, chitosan, synthetic carbohydrate, oligo lactate 15-mer, natural polymer, low- or medium-molecular weight polymer, inulin, cyclodextrin, hyaluronic acid, protein, protein-binding agent, integrin- targeting molecule, polycationic, peptide, polyamine, peptide mimic, and/or transferrin.
  • the present disclosure relates to a method of treating an APOC3-related disease in an individual, comprising the step of administering to the individual a therapeutically effective amount of a composition comprising an RNAi agent comprising an antisense strand, wherein the antisense strand comprises at least 14 contiguous nucleotides differing by 0, 1 , 2, or 3 nucleotides from the antisense strand of an RNAi agent to APOC3 selected from those specific duplexes provided above and as listed in any of the tables herein.
  • the RNAi agent to APOC3 comprises an antisense strand duplexed with a sense strand, wherein the sense and antisense strands are selected from one or more of the sequences provided in any of the tables herein. [00120] Various particular specific aspects of this aspect are described below. Any aspects disclosed herein that are not mutually exclusive can be combined.
  • the present disclosure relates to such a method, wherein the composition comprising a RNAi agent further comprises a sense strand, wherein the sense strand comprises at least 14 contiguous nucleotides differing by 0, 1 , 2, or 3 nucleotides from the sense strand of a RNAi agent to APOC3 selected from the specific duplexes provided herein and as listed, e.g., in any Table herein.
  • the RNAi agent comprises at least an anti-sense strand, and/or comprises a sense and an anti-sense strand, wherein the sequence of the sense and/or anti-sense strand is the sequence of the sense and/or the anti-sense strand of a RNAi agent to APOC3 selected from those specific duplex provided herein and as listed, e.g., in Table 1 , wherein the composition further comprises a pharmaceutically effective formulation.
  • the RNAi agent comprises at least an anti-sense strand, and/or comprises a sense and an anti-sense strand, wherein the sequence of the sense and/or anti-sense strand comprises the sequence of the sense and/or the anti-sense strand of a RNAi agent to APOC3 selected from those specific duplex provided herein and as listed, e.g., in any of the tables herein, wherein the composition further comprises a pharmaceutically effective formulation.
  • the APOC3-related disease is selected from hypertriglyceridemia (e.g., Type V Hypertriglyceridemia), abnormal lipid metabolism, abnormal cholesterol metabolism, atherosclerosis, hyperlipidemia, diabetes, including Type 2 diabetes, obesity, cardiovascular disease, and coronary artery disease, among other disorders relating to abnormal metabolism or otherwise.
  • hypertriglyceridemia e.g., Type V Hypertriglyceridemia
  • abnormal lipid metabolism e.g., abnormal cholesterol metabolism, atherosclerosis
  • hyperlipidemia e.g., diabetes, including Type 2 diabetes, obesity, cardiovascular disease, and coronary artery disease, among other disorders relating to abnormal metabolism or otherwise.
  • the method further comprises the step of administering an additional treatment.
  • the additional treatment is a method (or procedure). In one aspect, the additional treatment is a therapeutically effective dose of a composition.
  • the additional treatment and the RNAi agent can be administered in any order, or can be administered simultaneously.
  • the method further comprises the step of administering an additional treatment for an APOC3-related disease.
  • the method further comprises the step of administering an additional treatment.
  • a RNAi agent to APOC3 can be used in conjunction with any additional treatment disclosed herein, as appropriate for the disease, optionally, in further conjunction with one or more additional RNAi agents to APOC3.
  • references to any additional treatment are meant to also include the pharmaceutically acceptable salts of any of the active substances. If active substances comprised by components (a) and/or (b) have, for example, at least one basic center, they can form acid addition salts. Corresponding acid addition salts can also be formed having, if desired, an additionally present basic center. Active substances having an acid group, e.g., COOH, can form salts with bases.
  • the active substances comprised in components (a) and/or (b) or a pharmaceutically acceptable salts thereof may also be used in form of a hydrate or include other solvents used for crystallization.
  • the composition comprises a second RNAi agent to APOC3.
  • the second RNAi agent is physically distinct from the first, or the two are physically connected (e.g., linked or conjugated).
  • the first and second RNAi agents are combined within the same composition (e.g., both in the same lipid nanoparticle).
  • the present disclosure relates to a method of inhibiting the expression of APOC3 in an individual, comprising the step of administering to the individual a therapeutically effective amount of a composition comprising an RNAi agent of the disclosure.
  • the RNAi comprises a sense strand and an antisense strand, wherein the antisense strand comprises at least 14 contiguous nucleotides differing by 0, 1 , 2, or 3 nucleotides from the antisense strand of an RNAi agent to APOC3 selected from those specific duplexes provided above and as listed in any of the tables herein.
  • the RNAi agent comprises at least an anti-sense strand, and/or comprises a sense and an anti-sense strand, wherein the sequence of the sense and/or anti-sense strand is the sequence of the sense and/or the anti-sense strand of a RNAi agent to APOC3 selected from those specific duplex provided herein and as listed, e.g., in any of the tables herein, wherein the composition is in a pharmaceutically effective formulation.
  • the RNAi agent comprises at least an anti-sense strand, and/or comprises a sense and an anti-sense strand, wherein the sequence of the sense and/or anti-sense strand comprises the sequence of the sense and/or the anti-sense strand of a RNAi agent to APOC3 selected from those specific duplex provided herein and as listed, e.g., in any of the tables herein, wherein the composition is in a pharmaceutically effective formulation.
  • the individual is afflicted with or susceptible to an APOC3-related disease.
  • the APOC3-related disease is selected from hypertriglyceridemia (e.g., Type V Hypertriglyceridemia), abnormal lipid metabolism, abnormal cholesterol metabolism, atherosclerosis, hyperlipidemia, diabetes, including Type 2 diabetes, obesity, cardiovascular disease, and coronary artery disease, among other disorders relating to abnormal metabolism or otherwise.
  • hypertriglyceridemia e.g., Type V Hypertriglyceridemia
  • abnormal lipid metabolism e.g., abnormal cholesterol metabolism, atherosclerosis
  • hyperlipidemia e.g., diabetes, including Type 2 diabetes, obesity, cardiovascular disease, and coronary artery disease, among other disorders relating to abnormal metabolism or otherwise.
  • the method further comprises the step of administering an additional treatment.
  • the additional treatment and the RNAi agent can be administered in any order or can be administered simultaneously.
  • the method further comprises the step of administering an additional treatment for an ApoC3 condition such as hypertriglyceridemia (e.g., Type V).
  • an ApoC3 condition such as hypertriglyceridemia (e.g., Type V
  • Hypertriglyceridemia abnormal lipid metabolism, abnormal cholesterol metabolism, atherosclerosis, hyperlipidemia, diabetes, including Type 2 diabetes, obesity, cardiovascular disease, and coronary artery disease, among other disorders relating to abnormal metabolism or otherwise.
  • the composition comprises a second RNAi agent to APOC3.
  • the second RNAi agent is physically separate from the first, or the two are physically connected (e.g., covalently linked or otherwise conjugated).
  • the first and second RNAi agents are combined within the same composition (e.g., both in the same lipid nanoparticle).
  • the method further comprises the step of administering an additional RNAi agent which comprises at least 14 contiguous nucleotides differing by 0, 1 , 2, or 3 nucleotides from the antisense strand of a RNAi agent to APOC3 selected from the specific duplexes provided herein and as listed, e.g., in any Table herein.
  • an additional RNAi agent which comprises at least 14 contiguous nucleotides differing by 0, 1 , 2, or 3 nucleotides from the antisense strand of a RNAi agent to APOC3 selected from the specific duplexes provided herein and as listed, e.g., in any Table herein.
  • the present disclosure relates to a composition comprising a RNAi agent of the present disclosure.
  • the RNAi agent comprises at least an anti-sense strand, and/or comprises a sense and an anti-sense strand, wherein the anti-sense strand comprises at least 14 contiguous nucleotides differing by 0, 1 , 2, or 3 nucleotides from the anti-sense strand of a RNAi agent to APOC3 selected from those specific duplex provided herein and as listed, e.g., in any of the tables herein, wherein the composition is in a pharmaceutically effective formulation.
  • the RNAi agent comprises at least an anti-sense strand, and/or comprises a sense and an anti-sense strand, wherein the sequence of the sense and/or anti- sense strand is the sequence of the sense and/or the anti-sense strand of a RNAi agent to APOC3 selected from those specific duplex provided herein and as listed, e.g., in any of the tables herein, wherein the composition is in a pharmaceutically effective formulation.
  • the RNAi agent comprises at least an anti-sense strand, and/or comprises a sense and an anti-sense strand, wherein the sequence of the sense and/or anti- sense strand comprises the sequence of the sense and/or the anti-sense strand of a RNAi agent to APOC3 selected from those specific duplex provided herein and as listed, e.g., in any of the tables herein, wherein the composition is in a pharmaceutically effective formulation.
  • the present disclosure pertains to the use of a RNAi agent in the manufacture of a medicament for treatment of an APOC3-related disease, wherein the RNAi agent comprises a sense strand and an antisense strand, wherein the antisense strand comprises at least 14 contiguous nucleotides differing by 0, 1 , 2, or 3 nucleotides from the antisense strand of a RNAi agent to APOC3 selected from those specific duplex provided herein and as listed, e.g., in any Table herein.
  • RNAi agent to APOC3 Various specific aspects of a RNAi agent to APOC3 are disclosed herein.
  • the present disclosure encompasses the example modified variants provided in any of the tables herein, and the corresponding unmodified sequences and other modified variants.
  • Specific aspects of the present disclosure include RNAi agents which comprise sequences differing by 0, 1 , 2, or 3 nt (nucleotides) or bp [basepair(s)] (e.g., with 0, 1 , 2 or 3 mismatches) from any of the RNAi agents listed in any of the tables herein, and modified and unmodified variants thereof.
  • a mismatch is defined herein as a difference between the base sequence (e.g., A instead of G) or length when two sequences are maximally aligned and compared.
  • an "unmodified variant” is a variant in which the base sequence is identical, but none of the bases are modified; this includes, for example, a sequence identical to the corresponding portion of the wild-type APOC3 mRNA or gene.
  • a "modified variant” contains one or more modifications (or one or more fewer or different modifications) to a nucleotide, sugar, phosphate or backbone, and/or addition of one or more moieties; but without a change, substitution, addition, or deletion to the base sequence.
  • a particular sequence and its modified or unmodified variants have 0 mismatches among them.
  • the present disclosure comprises a RNAi agent comprising a sense and an anti-sense strand, wherein the sense and/or anti-sense strand comprises at least 14 contiguous nucleotides differing by 0, 1 , 2, or 3 nt from the sequence of the sense and/or anti-sense strand of: any of the RNAi agents listed in any of the tables herein, and modified and unmodified variants thereof.
  • the RNAi agent comprises a sense strand comprising at least 14 contiguous nucleotides differing by 0, 1 , 2, or 3 nt from the sense strand of any of the RNAi agents listed in any of the tables herein, and modified and unmodified variants thereof.
  • the disclosure pertains to a composition according to any of the above aspects, for use in a method of treating an APOC3-related disease in an individual, the method comprising the step of administering to the individual a therapeutically effective amount of a composition according to any of the claims.
  • the disclosure pertains to the composition according to any of the above aspects, for use in a method of inhibiting the expression of APOC3 in an individual, the method comprising the step of administering to the individual a therapeutically effective amount of a composition according to any of the above aspects.
  • One aspect of the disclosure is the use of a composition according to any of the above aspects, in the manufacture of a medicament for treatment of an APOC3-related disease.
  • the APOC3-related disease is selected from any disease listed herein.
  • the disclosure pertains to the composition of any of the above aspects, for use in the treatment of an APOC3-related disease.
  • the APOC3-related disease is selected from any disease listed herein.
  • the disclosure relates to a method of inhibiting the expression of APOC3 in an cell, comprising the step of introducing into the cell a composition comprising an RNAi agent comprising an antisense strand, wherein the antisense strand comprises at least 14 contiguous nucleotides differing by 0, 1 , 2, or 3 nucleotides from the antisense strand of an RNAi agent to APOC3 selected from the APOC3 siRNAs disclosed herein.
  • the disclosure relates to a method of inhibiting the expression of APOC3 in an cell, comprising the step of introducing into the cell a composition comprising an RNAi agent comprising a sense strand and an antisense strand, wherein the antisense strand comprises at least 14 contiguous nucleotides differing by 0, 1 , 2, or 3 nucleotides from the antisense strand, and the sense strand comprises at least 14 contiguous nucleotides differing by 0, 1 , 2, or 3 nucleotides from the sense strand of an RNAi agent to APOC3 selected from the APOC3 siRNAs disclosed herein.
  • siRNAs to APOC3 can be used to treat APOC3-related diseases.
  • An "APOC3- related disease” is any disease associated with APOC3 and/or a mutation and/or an over- expression of a wild-type and/or mutant APOC3, and/or diseases wherein disease progression is enhanced by or prognosis worsened by the presence of APOC3 and/or a mutation and/or an over-expression of wild-type and/or mutant APOC3.
  • Non-limiting examples of APOC3-related diseases include: hypertriglyceridemia (e.g., Type V
  • Hypertriglyceridemia abnormal lipid metabolism, abnormal cholesterol metabolism, atherosclerosis, hyperlipidemia, diabetes, including Type 2 diabetes, obesity, cardiovascular disease, and coronary artery disease, among other disorders relating to abnormal metabolism or otherwise.
  • the present disclosure pertains to the use of APOC3 RNAi agents in inhibiting and/or reducing the level and/or activity of APOC3, for treatment of APOC3-related diseases, particularly those diseases in which the level and/or activity of APOC3 is excessive.
  • diseases include hypertriglyceridemia (e.g., Type V
  • Hypertriglyceridemia abnormal lipid metabolism, abnormal cholesterol metabolism, atherosclerosis, hyperlipidemia, diabetes, including Type 2 diabetes, obesity, cardiovascular disease, and coronary artery disease, among other disorders relating to abnormal metabolism or otherwise.
  • the present disclosure encompasses APOC3 RNAi agents and the uses thereof for APOC3-related diseases.
  • the present disclosure pertains to the use of APOC3 RNAi agents, which inhibit and/or decrease the level and/or activity of APOC3, to treat APOC3- related diseases.
  • APOC3 RNAi agents which inhibit and/or decrease the level and/or activity of APOC3, to treat APOC3- related diseases.
  • inhibiting or decreasing the level and/or activity of APOC3 have been shown to be effective in treatment of APOC3 related diseases.
  • APOC3 RNAi agent for use in treating various APOC3-related diseases
  • the APOC3 RNAi agent of the present disclosure comprises a sequence disclosed herein and is administered to a patient in need thereof (e.g., a patient suffering from an APOC3-related disease disclosed herein or known in the literature). In one aspect, the APOC3 RNAi agent of the present disclosure is administered to a patient in need thereof, along with one or more additional pharmaceutical agent appropriate for that disease.
  • a patient suffering from an APOC3-related disease can be administered a pharmacologically effective amount of one or more APOC3 RNAi agent along with a pharmacologically effective amount of one or more of any APOC3-related disease treatment listed herein, and/or any other APOC3-related disease treatment known in the art.
  • a patient suffering from an APOC3-related disease can be administered one or more RNAi agent to APOC3 and one or more additional APOC3-related disease treatment.
  • This additional treatment can be selected from the list of any disease treatment listed herein, and/or any anti- APOC3-related disease treatment known in the art.
  • the APOC3 RNAi agents of the instant disclosure can be administered along with (as part of the same therapeutic treatment regimen, prior to, simultaneously with, or after) one or more additional therapeutics to treat an APOC3-related disease.
  • additional therapeutics can be selected from: iron administration, chelation therapy, phlebotomy, erythropoiesis stimulating agent (ESA) (e.g., Epoetin alfa or darbepoetin alfa), anti-APOC3 antibody, hemodialysis, and hyperbaric oxygen.
  • ESA erythropoiesis stimulating agent
  • the patient can also be administered more than one RNAi agent to APOC3.
  • RNAi agent(s) and additional disease treatment(s) can be administered in any order, simultaneously or sequentially, or in multiple doses over time.
  • Administration of the RNAi agent and the additional treatment can be, for example, simultaneous, concurrent, separate or sequential.
  • Simultaneous administration may, e.g., take place in the form of one fixed combination with two or more active ingredients, or by simultaneously administering two or more active ingredients that are formulated independently.
  • Sequential use (administration) preferably means administration of one (or more) components of a combination at one time point, other components at a different time point, that is, in a chronically staggered manner, preferably such that the combination shows more efficiency than the single compounds administered independently (especially showing synergism).
  • Separate use (administration) preferably means administration of the components of the combination independently of each other at different time points, preferably meaning that the components (a) and (b) are administered such that no overlap of measurable blood levels of both compounds are present in an overlapping manner (at the same time).
  • combinations of two or more of sequential, separate and simultaneous administration are possible, preferably such that the combination component-drugs show a joint therapeutic effect that exceeds the effect found when the combination component-drugs are used independently at time intervals so large that no mutual effect on their therapeutic efficiency can be found, a synergistic effect being especially preferred.
  • delay of progression means administration of the combination to patients being in a pre-stage or in an early phase, of the first manifestation or a relapse of the disease to be treated, in which patients, e.g., a pre-form of the
  • corresponding disease is diagnosed or which patients are in a condition, e.g., during a medical treatment or a condition resulting from an accident, under which it is likely that a corresponding disease will develop.
  • “Jointly therapeutically active” or “joint therapeutic effect” means that the compounds may be given separately (in a chronically staggered manner, especially a sequence-specific manner) in such time intervals that they preferably, in the warm-blooded animal, especially human, to be treated, still show a (preferably synergistic) interaction (joint therapeutic effect). Whether this is the case, can inter alia be determined by following the blood levels, showing that both compounds are present in the blood of the human to be treated at least during certain time intervals.
  • Any method known in the art can be used to identify efficacious APOC3 RNAi agents useful for treating APOC3-related diseases.
  • a few non-limiting examples of such methods are disclosed herein. Any one or more of the following, in addition to other methods known in the art, can be used.
  • APOC3 RNAi agents are tested for efficacy and tolerance to modifications and modification from 19-mer to 18-mer format.
  • RNAi candidates can be tested by target knockdown in Huh7 cells and primary hepatocytes. This can be tested using any method known in the art, including but not limited to RNAi Max.
  • the RNAi candidate demonstrates at least about 80% mRNA knockdown at 48 hours.
  • the mechanism can optionally be confirmed to be RNAi (RNA interference) by identifying specific cleavage products using RACE.
  • Cross-reactivity with cyno monkeys can be assessed in primary cyno hepatocytes at 48 hours.
  • the IC50 can be within five-fold of human siRNA activity.
  • Off-target effects of the candidate APOC3 RNAi agent can be assed. For example, cell growth in non-ApoC3-expressing cells can be tested. Preferably, there is no effect relative to, for example, YFP siRNA.
  • Immune stimulation by the candidate RNAi agent can be tested using whole blood assay. Preferably, there would be no detectable activation of cytokine expression.
  • Toxicity can also be assessed.
  • MTD maximum tolerated dosage
  • a Mini-tox study can be performed for 2 weeks.
  • a conjugate can be used.
  • Efficacy in animal models can be assessed.
  • a conjugate can be used to deliver mouse ApoC3 siRNA to liver and effectively knockdown the target.
  • the mRNA knockdown will be at least about 80% at 96 hours.
  • PK pharmacokinetics
  • bio-distribution of formulated RNAi agents can be tested, for example, in rodents, using, for example, LNP (lipid nanoparticle)-formulated RNAi agents.
  • LNP lipid nanoparticle
  • the RNAi targets the liver.
  • APOC3 sequences are identified, including various lead sequences. Serum stability highlights the 5' terminal U as a fragile site.
  • the 3' overhang is optimized, e.g., PAZ ligands are screened.
  • Passenger strand optimization is done. For example, this was done using the assymetrical design.
  • RNAi agents to APOC3 for use in treating APOC3-related diseases. These agents have various structural properties, including having an 18-mer sequence.
  • the agent is modified, including but not limited to, modifications of one or more sugar or phosphate, and with, in 3' order, a linker and a 3' end cap.
  • the 5' and/or 3' end of the sense strand can be conjugated to other components which increase targeting to and/or uptake by the liver.
  • compositions comprising the RNAi agent to
  • Hepicidin and, for example, an excipient or pharmaceutically acceptable carrier.
  • the APOC3 mRNA is unusually short, thus leaving only a small room for siRNA design, and very small room for pan-specific siRNAs (those which target both humans and one or more test animals).
  • compositions comprising an APOC3 RNAi agent.
  • These comprise a first strand and a second strand, wherein the sequence of the first and/or second strand is, comprises, comprises 15 contiguous nt of, or comprises 15 contiguous nt of (with 1-3 mismatches from) any sequence of any APOC3 RNAi agent listed herein.
  • the disclosure pertains to an APOC3 RNAi agent comprising a first and second strand, wherein the sequence of the first and/or second strand is, comprises, or comprises 15 contiguous nt of any sequence of any APOC3 RNAi agent disclosed herein, except that one or both strands is nicked and/or one or more nt of the sequence disclosed herein has been replaced by a spacer (e.g., the first comprises the sequence of nt 1 -5 and 7-18, or of 1 -4 and 6-18 of a sequence disclosed herein).
  • APOC3 RNAi agents can be of any format described herein or known in the art.
  • RNAi agents of the various formats can comprise strands of various lengths, one or more spacer, modified internucleoside linker, and 3' end cap.
  • RNAi agents of the 18-mer format for example, comprise a sense and an anti-sense strand, each strand being an 18-mer and the strands together forming a blunt-ended duplex, wherein the 3' end of at least one strand terminates in a phosphate or modified internucleoside linker and further comprises, in 5' to 3' order: a spacer; a second phosphate or modified
  • the 3' end of both the sense and anti-sense strand further comprise, in 5' to 3' order: a spacer; a second phosphate or modified internucleoside linker; and a 3' end cap.
  • the two strands can have the same or different spacers, phosphate or modified internucleoside linker, and/or 3' end caps.
  • one or more nt can be modified and/or substituted.
  • Various formats, spacers, modified internucleoside linkers and 3' end caps are described below.
  • the present disclosure contemplates a variety of different formats for APOC3 RNAi agents, including various formats using any APOC3 RNAi agent sequence disclosed herein.
  • a particular novel format designated the 18-mer format, has been developed. This and any other format can be used for an APOC3 RNAi agent.
  • siRNAs naturally generated in a cell by Dicer typically comprise two 21-nt RNA strands, which form a 19-bp duplex region and two dinucleotide overhangs. This is the so- called “canonical” siRNA structure. See, for example, Elbashir et al. 2001 Nature 41 1 : 494- 498; Elbashir et al. 2001 EMBO J. 20: 6877. This is the so-called “canonical” structure or "21-mer” format of siRNAs.
  • RNA 12:163-176 The two dinucleotide overhangs do not contribute to target specificity. Elbashir et al. 2001 Nature 41 1 : 494-498; Elbashir et al. 2001 EMBO J. 20: 6877-6888; and Kraynack et al. 2006 RNA 12:163-176. They do, however, help protect the ends of the siRNA from nuclease degradation and sometimes improve activity. However, these di-nucleotides can optionally be replaced by 3' end caps. Ideal 3' end caps allow RNA interference activity, while increasing stability, e.g., against nucleases such as those in blood serum or intestinal fluid. U.S. Pat. No. 8,097,716; and 8,084,600.
  • Novel 3' end caps are disclosed herein; these can be used in place of or in addition to a 3' end cap in APOC3 RNAi agents. It is noted that, although some 3' end caps are designated "PAZ ligands", this disclosure is not bound by any particular theory.
  • the canonical siRNA structure comprises two strands, each a 19- mer with a 2 nt overhang. Strands of shorter length, e.g., 18-mers, are less commonly produced by a cell.
  • RNAi agents Disclosed here is a 18-mer format shown to be efficacious in producing functional RNAi agents to a variety of different targets, with a variety of different sequences.
  • compositions comprising an APOC3 RNAi agent having a novel format (the "18-mer format").
  • These RNAi agents comprise a sense and an anti-sense strand, each strand being an 18-mer and the strands together forming a blunt- ended duplex, wherein the 3' end of at least one strand further comprises, in 5' to 3' order: a spacer; a phosphate or modified internucleoside linker; and a 3' end cap.
  • each strand terminates at the 5' end with a hydroxyl, optionally linked to a 5' end cap or a ligand, and terminates at the 3' end with a phosphate or modified
  • the 3' end of both the sense and anti-sense strand further comprise, in 5' to 3' order: a spacer; a phosphate or modified internucleoside linker; and a 3' end cap.
  • the two strands can have the same or different spacers, phosphate or modified internucleoside linker, and/or 3' end caps.
  • the strands can be ribonucleotides, or, optionally, one or more nucleotide can be modified or substituted.
  • At least one nucleotide comprises a modified internucleoside linker.
  • the RNAi agent can be modified on one or both 5' end.
  • the sense strand can comprise a 5' end cap which reduces the amount of the RNA interference mediated by this strand.
  • the RNAi agent is attached to a ligand. The disclosure also relates to processes for making such compositions, and methods and uses of such compositions, e.g., to mediate RNA interference.
  • the 3' end of both the sense and anti-sense strand terminate in a phosphate or modified internucleoside linker and further comprise in 5' to 3' order: a spacer, a phosphate or modified internucleoside linker, and a 3' end cap.
  • the spacer is a sugar, alkyl, cycloakyl, ribitol or other type of abasic nucleotide, 2'-deoxy-ribitol, diribitol, 2'-methoxyethoxy-ribitol (ribitol with 2'-MOE),
  • the spacers on the sense and anti-sense strands can be the same or different.
  • the modified internucleoside linker is selected from phosphorothioate, phosphorodithioate, phosphoramidate, boranophosphonoate, an amide
  • RNAi agent of any format, e.g., any APOC3 RNAi agent disclosed herein of any format disclosed herein.
  • APOC3 RNAi agents contemplates a variety of different formats for APOC3 RNAi agents, including various formats using any APOC3 RNAi agent sequence disclosed herein.
  • a particular novel format, designated the internal spacer, has been developed.
  • nucleoside subunits ([sugar+base]) of one or both strands has been replaced by a spacer.
  • the nucleobase subunit ([sugar+base]) replaced by a spacer is at position 1 , 3, 5, 6, 7, 15, 16, 17, or 18 (counting 5' to 3').
  • the nucleobase subunit ([sugar+base]) replaced by a spacer is at position 5, 6, or 17 (counting 5' to 3').
  • the spacer can be sugar, alkyl, cycloakyl, ribitol or other type of abasic nucleotide, 2'-deoxy-ribitol, diribitol, 2'- methoxyethoxy-ribitol (ribitol with 2 -MOE), C3, C4, C5, C6, or 4-methoxybutane-1 ,3-diol (5300), or any other spacer described herein or known in the art. More than one different type of spacer can be incorporated into the same composition.
  • the internal spacer format can be combined with other formats.
  • the internal spacer format can be combined with the 19-mer format.
  • the internal spacer format can also be combined, for example, with the 18-mer format.
  • the APOC3 RNAi agent can comprise a first and a second strand, wherein the both strands are 18-mers, and wherein the first strand is the anti-sense strand and a nucleoside subunit ([consisting of a sugar and base]) at any of positions 1 to 18 is replaced by a spacer.
  • a nucleoside subunit [consisting of a sugar and base]) at any of positions 1 to 18 is replaced by a spacer.
  • One or more nucleoside subunits can be replaced.
  • one or more nucleoside subunits on the first and/or second strand can be replaced by a spacer.
  • Non-limiting examples of APOC3 RNAi agents having an internal spacer format are described herein.
  • RNAi agent of any sequence targeting any target, which has the internal spacer format.
  • the anti-sense strands of two different duplexes are identical except that, at one position, the nucleobase unit (i.e., sugar + base) has been replaced by a spacer (e.g., a spacer described herein), the sequences are considered to have one mismatch (e.g., to differ by 1 nt).
  • a first sequence comprising at least 14 contiguous nt differing by 0, 1 , 2 or 3 nt from a second sequence can have any combination of 0, 1 , 2 or 3 internal spacer, substitution, and/or other position wherein the base at one sequence does not match the corresponding base at the other sequence.
  • RNAi AGENT FORMAT SHORTENED SENSE STRAND
  • the APOC3 RNAi agent comprises an anti-sense and a sense strand, wherein the sense strand has been shortened.
  • the sense strand is a 14-, 15-, 16-, or 17-mer.
  • the shortened sense strand format can be combined with any RNAi agent format known. This includes but is not limited to the 18-mer format.
  • an APOC3 RNAi agent can comprise an anti-sense strand which is an 18-mer (or longer), and a sense strand which is a 14-, 15-, 16-, or 17-mer.
  • the anti- sense strand can be an 18-, 19-, 20, 21 -, 22-, 23-, 24-, 25-, 26-, 27-, 28-, 29-, or 30-mer or longer.
  • the 3' terminus of the anti-sense and/or sense strand can optionally terminate a phosphate or modified internucleoside linker and further comprise: (a) a 3' end cap; or (b) a spacer, a second phosphate or modified internucleoside linker, and a 3' end cap (in 5' to 3' order).
  • the spacer, modified internucleoside linker and 3' end cap can be any of those elements detailed herein or known in the art.
  • a RNAi agent of the shortened sense strand format can comprise RNA subunits (ribonucleotides) wherein one or more ribonucleotides is modified and/or substituted by any modification or substitution described herein or known in the art.
  • the disclosure also pertains to a RNAi agent of any sequence, targeting any gene target, which has a shortened sense strand format.
  • APOC3 RNAi agents can pertain to sequences disclosed herein, and be of any of several formats described herein or known in the art.
  • Components of these various RNAi agents include different 3' end caps, spacers, and modified internucleoside linkers. Components can be mixed and matched.
  • each useful for producing an APOC3 RNAi agent comprise a 3' end cap.
  • One or both strands of an APOC3 RNAi agent can comprise, at the 3' terminus: a 3' end cap; or a spacer, a phosphate or internucleoside linker, and a 3' end cap.
  • Any of the various 3' end caps disclosed herein or known in the art can be used to prepare an APOC3 RNAi agent.
  • the 3' end cap is selected from those represented by formula 1 a or 1 b, disclosed in Tables 5A, 5B, 5C, 5D, 5E, or otherwise described herein.
  • the 3' end caps on the sense and anti-sense strands can be the same or different.
  • the 3' end cap encompasses a compound of formula la:
  • X is a 3' end of molecule comprising: a strand of an APOC3 RNAi agent, wherein the 3' end of the strand terminates in a phosphate or modified
  • internucleoside linker and optionally further comprises, in 5' to 3' order: a spacer and a second phosphate or modified internucleoside linker;
  • Y is selected from CH and N;
  • n is selected from 0 and 1 ;
  • p is selected from 1 , 2 and 3;
  • R 3 is selected from hydrogen, 2-(hydroxy-methyl)-benzyl, 3-(hydroxy-methyl)- benzyl and succinate; or is attached to a solid support; wherein the (CH 2 )m-0-R 3 moiety is attached to the phenyl ring at position 3 or 4;
  • R 4 is hydrogen
  • R 5 is hydrogen; or R 4 and R 5 , together with the phenyl rings to which R 4 and R 5 are attached, form 6H-benzo[c]chromene.
  • the 3' end cap encompasses a compound selected from Table 5A.
  • X is a 3' end of a molecule comprising: a strand of an APOC3 RNAi agent, wherein the 3' end of the strand terminates in a phosphate or modified
  • internucleoside linker and optionally further comprises, in 5' to 3' order: a spacer and a second phosphate or modified internucleoside linker.
  • the 3' end cap encompasses a compound of formula lb:
  • X is a 3' end of a molecule comprising: a strand of an APOC3 RNAi agent, wherein the 3' end of the strand terminates in a phosphate or modified
  • internucleoside linker and optionally further comprises, in 5' to 3' order: a spacer and a second phosphate or modified internucleoside linker;
  • q is selected from 0, 1 and 2;
  • R 6 is selected from phenyl which is unsubstituted or substituted with benzoxy
  • R 7 is selected from hydrogen and hydroxy-ethyl, wherein if R 7 is hydroxy-ethyl, the hydroxyl can be optionally functionalized as succinate or attached to a solid support;
  • R 8 is selected from hydrogen and methoxy
  • Yi is selected from CH and N;
  • Y 2 is selected from N and CR 9 ; wherein R 9 is selected from hydrogen and methyl.
  • the 3' end cap encompasses a compound selected from Table 5B.
  • X is a 3' end of a molecule comprising: a strand of an APOC3 RNAi agent, wherein the 3' end of the strand terminates in a phosphate or modified internucleoside linker and optionally further comprises, in 5' to 3' order: a spacer and a second phosphate or modified internucleoside linker.
  • the 3' end cap encompasses a compound selected from Table 5C.
  • X is a 3' end of a molecule comprising: a strand of an APOC3 RNAi agent, wherein the 3' end of the strand terminates in a phosphate or modified
  • internucleoside linker and optionally further comprises, in 5' to 3' order: a spacer and a second phosphate or modified internucleoside linker, and
  • the 3' end cap encompasses a compound selected from Table 5D.
  • X is a 3' end of a molecule comprising: a strand of an APOC3 RNAi agent, wherein the 3' end of the strand terminates in a phosphate or modified
  • internucleoside linker and optionally further comprises, in 5' to 3' order: a spacer and a second phosphate or modified internucleoside linker.
  • the 3' end cap encompasses a compound selected from Table 5E.
  • X is a 3' end of a molecule comprising: a strand of an APOC3 RNAi agent, wherein the 3' end of the strand terminates in a phosphate or modified
  • internucleoside linker and optionally further comprises, in 5' to 3' order: a spacer and a second phosphate or modified internucleoside linker.
  • Suitable 3' end caps including but not limited to:C3, C6, C12, Triethylene glycol, Cyclohexyl (or Cyclohex), Phenyl, Biphenyl, Adamantane and Lithocholic acid (or Lithochol), are described herein and/or in U.S. Pat. Nos. 8,097,716; 8,084,600; 8,344, 128; 8,404,831 ; and 8,404,832.
  • the 3' end cap is a ribitol.
  • the RNAi agent comprises an 18-mer strand further comprising at the 3' terminus, in 5' to 3' order, a spacer (e.g., a sugar or alkyl, cycloakyi, ribitol or other type of abasic nucleotide, C3, C4, C5, C6, etc.), a phosphate or modified internucleoside linker, and a 3' end cap (e.g., a second ribitol or other type of abasic nucleotide).
  • a spacer e.g., a sugar or alkyl, cycloakyi, ribitol or other type of abasic nucleotide, C3, C4, C5, C6, etc.
  • a phosphate or modified internucleoside linker e.g., a second ribitol or other type of abasic nucleotide.
  • the 3' end cap is a diribitol.
  • the RNAi agent comprises an 18-mer strand further comprising at the 3' terminus, in 5' to 3' order, a spacer (e.g., a ribitol, C3, C4, C5, C6, etc.), a phosphate or modified internucleoside linker, and a 3' end cap (e.g., a diribitol).
  • compositions comprising an APOC3 RNAi agent.
  • APOC3 RNAi agents can be of any format. These include the 18-mer format, the 19-mer format, the internal spacer format, the shortened sense strand format, or any other format or length of RNAi agent known in the art.
  • RNAi agents of the various formats can comprise strands of various lengths, one or more spacer, modified
  • RNAi agents of the 18-mer format comprise a sense and an anti-sense strand, each strand being an 18-mer and the strands together forming a blunt-ended duplex, wherein the 3' end of at least one strand terminates in a phosphate or modified internucleoside linker and further comprises, in 5' to 3' order: a spacer; a second phosphate or modified internucleoside linker; and a 3' end cap.
  • the 3' end of both the sense and anti-sense strand further comprise, in 5' to 3' order: a spacer; a second phosphate or modified internucleoside linker; and a 3' end cap.
  • the two strands can have the same or different spacers, phosphate or modified
  • internucleoside linker and/or 3' end caps.
  • one or more nt can be modified and/or substituted.
  • spacers, modified internucleoside linkers and 3' end caps are described below.
  • any of various spacers can be used in combination with strands of any sequence, with or without substitutions and/or modifications of nt, with phosphates or modified nucleoside spacers, and with any 3' end cap, in any combination without limitation.
  • a spacer is a chemical moiety intended or used to create or maintain a space (e.g., a proper or functional spacing) between two other chemical moieties; e.g., between two phosphates or modified internucleoside linkers.
  • the spacer is a ribitol, diribitol, 2'-deoxyribitol, or 2'-methoxyethoxy ribitol (ribitol with 2'-MOE) or an equivalent abasic nucleotide known to one skilled in the art, or a lower alkyl or alkoxy group such as a C3, C4, C5 or C6, or 4-methoxybutane-1 ,3-diol.
  • the spacer is ribitol.
  • the RNAi agent comprises, in 5' to 3' order: an 18-mer strand (comprising a 3' terminal phosphate or a modified internucleoside linker); a spacer which is ribitol; a phosphate or a modified internucleoside linker; and a 3' end cap (e.g., any 3' end cap described herein or otherwise known in the art).
  • the RNAi agent comprises, in 5' to 3' order: an 18-mer strand comprising a 3' terminal phosphate; a spacer which is ribitol; a phosphate or a modified internucleoside linker; and a 3' end cap.
  • the ribitol spacer is designated as N027 (C027, etc.).
  • RNAi agent comprises, in 5' to 3' order: an 18-mer strand, wherein the 3' end of the 18-mer strand terminates in a phosphate and further comprises, in 5' to 3' order: a spacer which is ribitol, a phosphate, and a 3' end cap which is X058.
  • This structure can be on any RNAi strand of any sequence or target.
  • any 3' end cap disclosed herein can be used.
  • ribitol with C6 cap wherein the last nucleotide of the 18- mer strand is and the 3' end of the 18-mer strand terminates in a phosphate and further comprises, in 5' to 3' order: a spacer which is ribitol, a phosphate, and a 3' end cap which is C6.
  • the RNAi agent comprises, in 5' to 3' order: an 18-mer strand terminating in a 3' phosphate, a ribitol spacer, a phosphate, and a C6 3' end cap.
  • the RNAi agent comprises, in 5' to 3' order: an 18-mer strand terminating in a 3' phosphate, a ribitol spacer, a phosphate, and a BP 3' end cap. In another embodiment, the RNAi agent comprises, in 5' to 3' order: an 18-mer strand terminating in a 3' phosphate, a ribitol spacer, a phosphate, and a C10 3' end cap.
  • the 3' end cap is a ribitol.
  • the RNAi agent comprises an 18-mer strand, wherein the 3' end of the 18-mer strand terminates in a phosphate or modified internucleoside linker and further comprises, in 5' to 3' order: a spacer which is ribitol, a second phosphate or modified internucleoside linker, and a 3' end cap which is a second ribitol.
  • the 3' end of the 18-mer strand terminates in a phosphate and further comprises, in 5' to 3' order: a spacer which is ribitol, a second phosphate, and a 3' end cap which is a second ribitol.
  • RNAi agent comprising, in 5' to 3' order, an 18-mer strand terminating in a 3' terminal phosphate or a modified internucleoside linker, a ribitol spacer, a phosphate or a modified internucleoside linker, and a 3' end cap
  • any RNAi agent of sequence or target including but not limited to a double-stranded RNA, wherein optionally one or more phosphates are replaced by a modified internucleoside linker, optionally one or more nucleotides are modified, and optionally one or more RNA nucleotides are replaced by DNA, PNA, LNA, morpholino, TNA, GNA, FANA, ANA, HNA, CeNA, and/or UNA.
  • the spacer is Diribitol.
  • the RNAi agent comprises, in 5' to 3' order: an 18-mer strand wherein the 3' end of the strand terminates in a phosphate or modified internucleoside linker and further comprises in 5' to 3' order: a spacer (wherein the spacer comprises in 5' to 3' order: a first ribitol; a phosphate or a modified internucleoside linker; a second ribitol; and a phosphate or a modified internucleoside linker); and a 3' end cap.
  • the RNAi agent comprises, in 5' to 3' order: an 18-mer strand comprising a 3' terminal phosphate; a first ribitol; a phosphate; a second ribitol; a phosphate or a modified internucleoside linker; and a 3' end cap.
  • the RNAi agent comprises, in 5' to 3' order: an 18-mer strand comprising a 3' terminal phosphate; a first ribitol spacer; a phosphate; a second ribitol spacer; a phosphate or a modified internucleoside linker; and a 3' end cap.
  • the RNAi agent comprises, in 5' to 3' order: an 18-mer strand terminating in a 3' terminal phosphate or a modified internucleoside linker, a first ribitol spacer, a phosphate or a modified internucleoside linker, a second ribitol spacer, a phosphate or a modified internucleoside linker, and a 3' end cap which is a ribitol; this structure is designated a triribitol.
  • RNAi agent comprising, in 5' to 3' order, an 18-mer strand terminating in a 3' terminal phosphate or a modified internucleoside linker, a first ribitol spacer, a phosphate or a modified internucleoside linker, a second ribitol spacer, a phosphate or a modified internucleoside linker, and a 3' end cap
  • any RNAi agent of sequence or target including but not limited to a double-stranded RNA, wherein optionally one or more phosphates are replaced by a modified internucleoside linker, optionally one or more nucleotides are modified, and optionally one or more RNA
  • nucleotides are replaced by DNA, PNA, LNA, morpholino, TNA, GNA, ANA, HNA, FANA, CeNA, and/or UNA.
  • the spacer is 2'-methoxyethoxy ribitol or other type of abasic nucleotide.
  • the RNAi agent comprises a strand, wherein the 3' end of the strand terminates in a phosphate or modified internucleoside linker and further comprises, in 5' to 3' order: a spacer which is 2'-methoxyethoxy ribitol, a second phosphate or modified internucleoside linker, and a 3' end (e.g., any 3' end cap described herein or known in the art).
  • the RNAi agent comprises, in 5' to 3' order: a strand comprising a 3' terminal phosphate or a modified internucleoside linker; a spacer which is 2'- methoxyethoxy ribitol; a phosphate or a modified internucleoside linker; and a 3' end cap (e.g., any 3' end cap described herein or known in the art).
  • the RNAi agent comprises, in 5' to 3' order: a strand comprising a 3' terminal phosphate; a spacer which is 2'-methoxyethoxy ribitol; a phosphate or a modified internucleoside linker; and a 3' end cap.
  • the RNAi agent comprises, in 5' to 3' order: an 18-mer strand, wherein the 3' end of the 18-mer strand terminates in a phosphate and further comprises, in
  • a spacer which is 2'-methoxyethoxy ribitol, a phosphate, and a 3' end cap which is X058.
  • This structure can be on any RNAi strand of any sequence or target.
  • any 3' end cap disclosed herein can be used in place of X058.
  • a related structure is 2'-methoxyethoxy ribitol with X058, wherein the last nucleotide of the 18-mer strand is a 2'-MOE), and the 3' end of the 18-mer strand terminates in a phosphate and further comprises, in 5' to 3' order: a spacer which is 2'-methoxyethoxy ribitol, a second phosphate, and a 3' end cap which is X058.
  • Another embodiment is 2'-methoxyethoxy ribitol with C6 cap, wherein the last nucleotide of the 18-mer strand is a 2'-MOE), and the 3' end of the 18-mer strand terminates in a phosphate and further comprises, in 5' to 3' order: a spacer which is 2'-methoxyethoxy ribitol, a phosphate, and a 3' end cap which is C6.
  • the RNAi agent comprises, in 5' to 3' order: an 18-mer strand terminating in a 3' phosphate, a 2'-methoxyethoxy ribitol spacer, a phosphate, and a C6 3' end cap.
  • the RNAi agent comprises, in 5' to 3' order: an 18-mer strand terminating in a 3' phosphate, a 2'-methoxyethoxy ribitol spacer, a phosphate, and a BP 3' end cap.
  • the RNAi agent comprises, in 5' to 3' order: an 18-mer strand terminating in a 3' phosphate, a 2'-methoxyethoxy ribitol spacer, a phosphate, and a C10 3' end cap.
  • the RNAi agent comprises a strand, wherein the 3' end of the strand terminates in a phosphate and further comprises, in 5' to 3' order: a spacer which is 2'-methoxyethoxy ribitol, a phosphate, and a 3' end cap which is X058.
  • the 3' end cap is a 2'-methoxyethoxy ribitol.
  • the RNAi agent comprises an 18-mer strand, wherein the 3' end of the 18-mer strand terminates in a phosphate or modified internucleoside linker and further comprises, in 5' to 3' order: a spacer which is 2'-methoxyethoxy ribitol, a second phosphate or modified internucleoside linker, and a 3' end cap which is a second 2'-methoxyethoxy ribitol.
  • the 3' end of the 18-mer strand terminates in a phosphate and further comprises, in 5' to 3' order: a spacer which is 2'-methoxyethoxy ribitol, a second phosphate, and a 3' end cap which is a second 2'-methoxyethoxy ribitol.
  • the structure comprises an RNAi agent comprising, in 5' to 3' order, a strand terminating in a 3' terminal phosphate or a modified internucleoside linker, a 2'-methoxyethoxy ribitol spacer, a phosphate or a modified internucleoside linker, and a 3' end cap (e.g., any 3' end cap known in the art) can be used on any RNAi agent of any length, sequence or target, including but not limited to a double-stranded RNA, wherein optionally one or more phosphates are replaced by a modified internucleoside linker, optionally one or more nucleotides are modified, and optionally one or more RNA
  • nucleotides are replaced by DNA, PNA, LNA, morpholino, TNA, GNA, ANA, HNA, CeNA, FAN A, and/or UNA.
  • 2' -deoxyribitol spacer In some embodiments the spacer is 2'-deoxyribitol.
  • the RNAi agent comprises, in 5' to 3' order: an 18-mer strand terminating in a 3' phosphate or a modified internucleoside linker, a spacer which is 2'- deoxyribitol (2'-deoxyrib), a phosphate or a modified internucleoside linker, and a 3' end cap.
  • the RNAi agent comprises, in 5' to 3' order: an 18-mer strand terminating in a 3' phosphate, a spacer which is 2'-deoxyribitol (2'-deoxyrib), a phosphate or a modified internucleoside linker, an ' end cap.
  • a 3' terminal phosphate 2'-deoxyribitol (2'-deoxyrib)
  • the RNAi agent comprises, in 5' to 3' order: an 18-mer strand terminating in a 3' phosphate, a 2'-deoxyribitol spacer, a phosphate, and a C12 3' end cap.
  • RNAi agent comprising, in 5' to 3' order, an 18-mer strand terminating in a 3' terminal phosphate or a modified internucleoside linker, a 2'- deoxyribitol spacer, a phosphate or a modified internucleoside linker, and a 3' end cap
  • any RNAi agent of any sequence or target including but not limited to a double- stranded RNA, wherein optionally one or more phosphates are replaced by a modified internucleoside linker, optionally one or more nucleotides are modified, and optionally one or more RNA nucleotides are replaced by DNA, PNA, LNA, morpholino, TNA, GNA, ANA, HNA, CeNA, FANA, and/or UNA.
  • C3 spacer In various embodiments, the spacer is C3.
  • the RNAi agent comprises an 18-mer strand, wherein the 3' end of the 18-mer strand terminates in a 3' phosphate or a modified internucleoside linker, and further comprises a spacer which is C3, a phosphate or a modified internucleoside linker, and a 3' end cap.
  • the RNAi agent comprises two 18-mer strands, wherein the 3' end of each 18-mer strand terminates in a 3' phosphate or a modified internucleoside linker, and further comprises a spacer, a second phosphate or a modified internucleoside linker, and a 3' end cap, wherein the spacer in one or both strands is C3.
  • the C3 spacer has the chemical form -(CH 2 ) 3 -.
  • RNAi agent comprises, in 5' to 3' order: an 18-mer strand terminating in a 3' phosphate, a C3 spacer, a phosphate, and a 3' end cap which is X058.
  • This structure can be on any RNAi strand of any sequence or target.
  • any 3' end cap disclosed herein or known in the art can be used in place of X058, and any modified internucleoside linker can be used in place of phosphate.
  • RNAi agent comprising an 18-mer strand, wherein the 18-mer strand terminates in a phosphate and further comprises in 5' to 3' order: a C3 spacer, a phosphate and a 3' end cap which is C6.
  • C3pC6 overhang This structure can be on any RNAi strand of any sequence or target.
  • any 3' end cap disclosed herein or known in the art can be used in place of C6, and any modified internucleoside linker can be used in place of phosphate.
  • RNAi agent comprising, in 5' to 3' order, an 18-mer strand terminating in a 3' terminal phosphate or a modified internucleoside linker, a C3 spacer, a phosphate or a modified internucleoside linker, and a 3' end cap
  • any RNAi agent of any sequence or target including but not limited to a double-stranded RNA, wherein optionally one or more phosphates are replaced by a modified internucleoside linker, optionally one or more nucleotides are modified, and optionally one or more RNA nucleotides are replaced by DNA, PNA, LNA, morpholino, TNA, GNA, ANA, HNA, CeNA, FAN A, and/or UNA.
  • the spacer is C4 or C5 or C6.
  • the APOC3 RNAi agent comprises an 18-mer strand, wherein the 3' end of the 18-mer strand terminates in a 3' phosphate or a modified internucleoside linker, and further comprises a spacer which is C4 or C5 or C6, a phosphate or a modified internucleoside linker, and a 3' end cap.
  • the RNAi agent comprises two 18-mer strands, wherein the 3' end of each 18-mer strand terminates in a 3' phosphate or a modified internucleoside linker, and further comprises a spacer, a second phosphate or a modified internucleoside linker, and a 3' end cap, wherein the spacer in one or both strands is C4 or C5 or C6.
  • the C4 spacer has the chemical formula -(CH 2 )4-
  • the C5 spacer has the chemical formula -(CH 2 )5-
  • the C6 spacer has the chemical formula -(CH 2 ) 6 -.
  • the structure comprising an RNAi agent comprises in 5' to 3' order, an 18-mer strand terminating in a 3' terminal phosphate or a modified internucleoside linker, a C4 or C5 or C6 spacer, a phosphate or a modified internucleoside linker, and a 3' end cap can be used on any RNAi agent of any sequence or target, including but not limited to a double-stranded RNA, wherein optionally one or more phosphates are replaced by a modified internucleoside linker, optionally one or more nucleotides are modified, and optionally one or more RNA nucleotides are replaced by DNA, PNA, LNA, morpholino, TNA, GNA, ANA, HNA, CeNA, FANA, and/or UNA.
  • C3 [-(CH 2 ) 3 -], "C4" [-(CH 2 ) 4 -], and "C5" [-(CH 2 ) 5 -] are generally used herein to designate spacers
  • similar terms C3, C4, C5 "linkers”
  • C3 is used to designate a C3 3' end cap (see, e.g, U.S. Pat. No. 8,097,716), a C3 spacer, and a C3 linker.
  • the C6 spacer should also be differentiated from the C6 3' end cap.
  • 4-methoxybutane-1 ,3-diol (5300) spacer.
  • the spacer is 4-methoxybutane-1 ,3-diol.
  • 4-methoxybutane-1 ,3-diol is also designated 5300, A5300, C5300, G5300, and UG5300.
  • the APOC3 RNAi agent comprises, in 5' to 3' order: an 18-mer strand, wherein the 3' end terminates in a 3' phosphate (a 3' terminal phosphate) or a modified internucleoside linker and further comprises: a spacer which is 4-methoxybutane- 1 ,3-diol, a phosphate or a modified internucleoside linker, and a 3' end cap.
  • the RNAi agent comprises, in 5' to 3' order: an 18-mer strand terminating in a 3' phosphate, a spacer which is 4-methoxybutane-1 ,3-diol, a phosphate or a modified internucleoside linker, and a 3' end cap.
  • the RNAi agent comprises, in 5' to 3' order: an 18-mer strand terminating in a 3' phosphate, a 4-methoxybutane-1 ,3-diol spacer, a phosphate, and a 3' end cap which is X058.
  • This structure can be on any RNAi strand of any sequence or target.
  • any 3' end cap disclosed herein or known in the art can be used in place of X058, and any modified internucleoside linker can be used in place of phosphate.
  • the RNAi agent comprises, in 5' to 3' order, an 18-mer strand terminating in a 3' terminal phosphate or a modified internucleoside linker, a 4- methoxybutane-1 ,3-diol spacer, a phosphate or a modified internucleoside linker, and a 3' end cap can be used on any RNAi agent of any sequence or target, including but not limited to a double-stranded RNA, wherein optionally one or more phosphates are replaced by a modified internucleoside linker, optionally one or more nucleotides are modified, and optionally one or more RNA nucleotides are replaced by DNA, PNA, LNA, morpholino, TNA, GNA, ANA, HNA, CeNA, FANA, and/or UNA.
  • the modified internucleoside linker is: phosphorothioate, phosphorodithioate, phosphoramidate, boranophosphonoate, an amide linker, or a compound of formula (I).
  • the disclosure relates to compositions comprising an APOC3 RNAi agent having a novel format.
  • RNAi agents comprise a sense and an anti-sense strand, each strand being an 18-mer and the strands together forming a blunt-ended duplex, wherein the 3' end of at least one strand terminates in a phosphate or modified internucleoside linker and further comprises, in 5' to 3' order: a spacer; a second phosphate or modified
  • the 3' end of both the sense and anti-sense strand further comprise, in 5' to 3' order: a spacer; a second phosphate or modified internucleoside linker; and a 3' end cap.
  • the two strands can have the same or different spacers, phosphate or modified internucleoside linker, and/or 3' end caps.
  • one or more nt can be modified and/or substituted.
  • spacers are described below.
  • Various phosphates or modified internucleoside linkers can be used in combination with strands of any sequence, with or without substitutions and/or modifications of nt, with any spacers, and with any 3' end cap, in any combination without limitation.
  • the modified internucleoside linker is interposed between the spacer and the 3' end cap.
  • one or more of the phosphates of one or both strands of the RNAi agent are replaced.
  • one or more nucleotide of one or both strands has a modified internucleoside linker.
  • the 3' terminal phosphate is replaced.
  • one or more nucleotide of one or both strands has a modified internucleoside linker, and/or a modified internucleoside linker is interposed between the spacer and the 3' end cap.
  • the present disclosure encompasses a RNAi agent comprising a sense and an anti-sense strand, each strand being an 18-mer and the strands together forming a blunt-ended duplex, wherein the 3' end of at least one strand terminates in a phosphate or modified internucleoside linker and further comprises, in 5' to 3' order: a spacer; a second phosphate or modified internucleoside linker; and a 3' end cap, wherein the 3' end cap is selected from the 3' end caps listed in any Table herein or otherwise disclosed herein, and wherein at least one nucleotide has a modified internucleoside linker a modified internucleoside linker (e.g., wherein at least one phosphate of a nucleotide is replaced by a modified internucleoside linker), where the modified internucleoside linker is:
  • R 3 is selected from O “ , S “ , NH 2 , BH 3 , CH 3 , Ci_ 6 alkyl, C 6- io aryl, Ci -6 alkoxy and C 6 -io aryl-oxy, wherein Ci -6 alkyl and C 6- io aryl are unsubstituted or optionally independently substituted with 1 to 3 groups independently selected from halo, hydroxyl and NH 2 ; and R 4 is selected from O, S, NH, or CH 2 .
  • the present disclosure encompasses a RNAi agent comprising a sense and an anti-sense strand, each strand being an 18-mer and the strands together forming a blunt-ended duplex, wherein the 3' end of at least one strand terminates in a phosphate or modified internucleoside linker and further comprises, in 5' to 3' order: a spacer; a second phosphate or modified internucleoside linker; and a 3' end cap, wherein the 3' end cap is selected from the 3' end caps listed in any Table herein or otherwise disclosed herein, and wherein at least the 3' terminal nucleotide on one or both strands has a modified internucleoside linker (e.g., wherein the phosphate of the 3' nucleotide on one or both strands is replaced by a modified internucleoside linker), wherein the modified internucleoside linker is phosphorothioate, phosphorodithio
  • the 3' end cap is linked via a terminal phosphate group (i.e., a phosphate group at the 3' end of a RNAi agent strand).
  • a terminal phosphate group i.e., a phosphate group at the 3' end of a RNAi agent strand.
  • Such compounds are shown in, for example, Table 5A-E.
  • a 3' end cap can be bound to a phosphate or modified internucleoside linker, which is bound to a spacer, which is bound to a phosphate or modified internucleoside linker bound to the 3' carbon at the 3' end of at least one RNAi agent strand.
  • compounds of table 5A-E have a terminal phosphorothioate group bound to the 3' carbon at the 3' end of at least one RNAi agent strand.
  • the phosphate group is replaced by a phosphorothioate.
  • the composition comprises a RNAi agent comprising a strand, wherein the 3' end of the strand terminates in a phosphorothioate and further comprises a compound of Table 5A-E (or any other 3' end cap described herein or known in the art).
  • the phosphate group of various 3' end caps listed herein as C3, C6, C12, Triethylene glycol, Cyclohexyl, Phenyl, Biphenyl, Adamantane, Lithocholic acid can be replaced by phosphorothioate.
  • the phosphate group in the C3 3' end cap is replaced by phosphorothioate.
  • the phosphate group in the C6 3' end cap is replaced by phosphorothioate.
  • the phosphate group in the C10 3' end cap is replaced by phosphorothioate.
  • the phosphate group in the biphenyl (BP) 3' end cap is replaced by phosphorothioate.
  • the APOC3 RNAi agent comprises a 18-mer strand further comprising at the 3' terminus a 3' end cap (but no spacer, or phosphate or internucleoside linker).
  • the RNAi agent comprises an 18-mer strand further comprising at the 3' terminus a 3' end cap (e.g., BP or C6).
  • the APOC3 RNAi agent comprises an 18-mer strand further comprising at the 3' terminus a spacer (but no phosphate or internucleoside linker, or 3' end cap).
  • the RNAi agent comprises an 18-mer strand further comprising at the 3' terminus a spacer (e.g., ribitol).
  • one or both strands can comprise ribonucleotide subunits, or one or more nucleotide can optionally be modified or substituted.
  • the RNAi agent can either contain only naturally-occurring ribonucleotide subunits, or one or more modifications to the sugar, phosphate or base of one or more of nucleotide subunits. In one embodiment, the modifications improve efficacy, stability and/or reduce immunogenicity of the RNAi agent.
  • an APOC3 RNAi agent comprising at least one non-natural nucleobase.
  • the non-natural nucleobase is difluorotolyl, nitroindolyl, nitropyrrolyl, or nitroimidazolyl.
  • the non- natural nucleobase is difluorotolyl.
  • only one of the two strands contains a non-natural nucleobase.
  • both of the strands contain a non-natural nucleobase.
  • the first two base-pairing nucleotides on the 3' end of the sense and/or anti-sense strand are modified. In one embodiment, the first two base-pairing nucleotides on the 3' end of the sense and/or anti-sense strand are 2'-MOE (a 2' MOE clamp).
  • the 3' terminal phosphate of the sense and/or anti-sense strands is replaced by a modified internucleoside linker.
  • one of more nucleotides is substituted with DNA, a peptide nucleic acid (PNA), locked nucleic acid (LNA), morpholino nucleotide, threose nucleic acid (TNA), glycol nucleic acid (GNA), arabinose nucleic acid (ANA), 2 - fluoroarabinose nucleic acid (FANA), cyclohexene nucleic acid (CeNA), anhydrohexitol nucleic acid (HNA), or unlocked nucleic acid (UNA).
  • PNA peptide nucleic acid
  • LNA locked nucleic acid
  • TAA threose nucleic acid
  • GAA glycol nucleic acid
  • ANA arabinose nucleic acid
  • FANA 2 - fluoroarabinose nucleic acid
  • CeNA cyclohexene nucleic acid
  • HNA unlocked nucleic acid
  • UNA unlocked nucleic acid
  • RNA with DNA or a nucleotide of a different backbone, or PNA, LNA, Morpholino, TNA, GNA, ANA, HNA, CeNA, FANA, and/or UNA can be considered a "modification".
  • At least one nucleotide comprises a modified
  • internucleoside linker wherein the modified internucleoside linker is selected from phosphorothioate, phosphorodithioate, phosphoramidate, boranophosphonoate, an amide linker, and a compound of formula (I).
  • the 3' terminal phosphate of the sense and/or anti-sense strands is replaced by a modified internucleoside linker.
  • the RNAi agent can be modified on one or both 5' end.
  • the sense strand can comprise a 5' end cap which reduces the amount of the RNA interference mediated by this strand.
  • the sense strand comprises a 5' end cap selected: a nucleotide lacking a 5' phosphate or 5'-OH; a nucleotide lacking a 5' phosphate or a 5'-OH and also comprising a 2-OMe or 2'-MOE modification; 5'-deoxy-2'-0-methyl modification; 5'- OME-dT; ddT; and 5'-OTr-dT.
  • the RNAi agent is optionally attached to a ligand.
  • the ligand can be selected to improve one or more characteristic, such as, e.g., stability, distribution and/or cellular uptake of the agent, e.g., cholesterol or a derivative thereof.
  • the RNAi agent can be isolated or be part of a
  • the pharmaceutical composition can be a lipid
  • the pharmaceutical composition can be a lipid
  • compositions can further comprise or be used in conjunction with any known treatment for any target gene-related disease.
  • the present disclosure further provides methods for reducing the level of target gene mRNA in a cell, particularly in the case of a disease characterized by over-expression or hyper-activity of the target gene product.
  • the present disclosure also encompasses a method of treating a human subject having a pathological state mediated at least in part by target gene expression, the method comprising the step of administering to the subject a therapeutically effective amount of a RNAi agent target gene.
  • Such methods comprise the step of administering one of the RNAi agents of the present disclosure to a subject.
  • Reduction of target gene mRNA in a cell results in a reduction in the amount of encoded target gene protein produced.
  • the invention provides an RNAi agent with any one or more of the above properties for use as a medicament.
  • RNAi agent with any one or more of the above properties for use as a medicament.
  • the methods and compositions of the present disclosure e.g., the methods and target gene RNAi agent compositions, can be used with any dosage and/or formulation described herein, as well as with any route of administration described herein.
  • the RNAi agent can be combined with one or more additional RNAi agents in the same formulation.
  • the one or more additional RNAi agents can have the same or different sequences, targets, spacers, 3' end caps, nucleotide replacements modifications, and/or ligands, etc.
  • the one or more additional RNAi agents can have a sense and an anti-sense strand wherein each is an 18- mer and together form a blunt-ended duplex.
  • the one or more additional RNAi agent can target the same or different sequence and/or the same or different target gene.
  • RNAi agents can be administered separately or co-administered.
  • the multiple RNAi agents can be administered in the same delivery vehicle, the same type of delivery vehicle, or in different delivery vehicles.
  • any of the various 3' end caps e.g., ligands or PAZ ligands
  • ligands or PAZ ligands can be used with the APOC3 RNAi agents described herein.
  • a 3' end cap is a non-nucleotidic chemical moiety bound to the 3' end of a molecule comprising a RNAi agent, e.g., the 3' terminus (or 3' end) of (a) a molecule comprising a strand, wherein the 3' end of the strand terminates in a phosphate or modified
  • RNA interference mediated by the molecule comprises, in 5' to 3' order: a strand (wherein the 3' end of the strand terminates in a phosphate or modified internucleoside linker), a spacer, and a second phosphate or modified internucleoside linker.
  • the 3' end cap performs at least one of the following functions: allowing RNA interference mediated by the molecule, protecting the molecule from degradation or reducing the amount or rate of degradation of the molecule (e.g., by nucleases), reducing the off-target effects of the sense strand, or increasing the activity, duration or efficacy of RNA interference mediated by the molecule.
  • nucleotide comprises three components: a phosphate, a pentose (e.g., a ribose or deoxyribose) and a nucleobase, and a 3' end cap does not comprise all three components.
  • 3' end caps include, but are not limited to compounds designated "PAZ ligands". These ligands can provide increased potency and/or higher siRNA turnover.
  • Human APOC3 RNAi agents were constructed, with the guide strand, with or without (+ or -) the MOE clamp (wherein the first two nt on the 3' end of each strand are 2'- MOE), with or without (+ or -) the ribitol spacer, and with each of the 17 listed PAZ ligands,
  • the passenger strands were constructed with or without (+ or -) the MOE clamp, with or without (+ or -) the ribitol spacer, and with C6 as the 3' end cap.
  • RNAi agents are tested in a 3 point dose response.
  • RNAi agents are introduced into Huh7 cells (10,000) using Optifect.
  • RNAi agent concentrations 30, 15, 10 and 5 nM.
  • qRT-PCR is performed and duplicate, and the gene product is normalized to GAPDH mRNA.
  • siRNAs Once efficacious siRNA sequences are identified, these siRNAs can be optimized by performing various modifications and testing those modifications to find those which improve efficacy, duration and/or stability and/or reduce off-target effects, or otherwise improve the performance of the siRNA.
  • the efficacious human APOC3 RNAi agents can be modified by replacing a RNA with 2'-OMe-RNA, DNA, 2 -F-RNA, 2'-MOE-RNA, and/or addition of a spacer (e.g., ribitol), and/or additional of a 3' end cap (X058, C6 or other 3' end cap).
  • a spacer e.g., ribitol
  • X058, C6 or other 3' end cap e.g., C6 or other 3' end cap
  • RNAi Agents can also be modified by replacement of RNA with DNA.
  • nucleotides in the seed region can be replaced by DNA.
  • various references define the exact limit of the seed region differently; some describe it as nt 4-8, 2-8 or 2-7, counting from the 5' end of the anti-sense strand.
  • the seed region refers to nt 2-7.
  • RNAi names sometimes have suffices identifying a base as DNA (e.g., "pos4_DNA” indicates that the nucleotide at position 4 is DNA; "pos8_DNA” indicates that that at position 8 is DNA, etc.).
  • Off-target effects can be measured, for example, as EC50 in nM in HeLa cells.
  • the data show that DNA is thus well-tolerated, e.g., in the seed region and can reduce the off-target phenotype.
  • other positions e.g., position 2
  • Modified variants of APOC3 RNAi agents include conjugate formats for 18-mers and 19-mers.
  • Modified variants of APOC3 RNAi agents include those with modifications of 5' and/or 3' end of the passenger strand to components which increase targeting to and uptake by the liver; and/or modification of the 5' end of the anti-sense strand to stabilize it; and/or modification of the 3' end of the anti-sense strand, e.g., by conjugation with a 3' end cap, to increase stability and/or potency.
  • Any pharmaceutical carrier known in the art can be used to deliver APOC3 siRNAs.
  • Criteria for an effective delivery vehicle include effective target knockdown in the liver at a reasonable dosage and duration, and acceptable pharmacokinetics and toxicity.
  • Delivery vehicles include, for example, lipid nanoparticles (LNPs), non-LNP delivery, peptide conjugation, and lipoprotein particles (LLPs).
  • LNPs lipid nanoparticles
  • LLPs lipoprotein particles
  • Particles can be, for example 10-100 nM in size, and can, for example, self-assemble in aqueous media.
  • Lipid nanoparticle Lipid nanoparticle
  • APOC3 siRNAs can be delivered using a lipid nanoparticle (LNP).
  • LNP lipid nanoparticle
  • Such an LNP can comprise, for example, a cationic lipid, cholesterol, a neutral lipid, and a shielding lipid.
  • APOC3 18-mer RNAi agents were able to deduce gene expression by over 70% compared to PBS control at 48 hours.
  • Other studies demonstrated the efficacy of APOC3 siRNAs in vivo using a variety of LNPs.
  • RNAi agent is a composition comprising at least an antisense nucleic acid sequence
  • an "antisense" nucleic acid comprises a nucleotide sequence complementary to a "sense" nucleic acid encoding the target protein (e.g., complementary to the coding strand of a double-stranded DNA, complementary to an mRNA or complementary to the coding strand of a target gene or nucleic acid).
  • RNAi agents include, as non-limiting examples, siRNAs (small interfering RNAs), dsRNAs (double stranded RNAs), shRNAs (short hairpin RNAs) and miRNAs (micro RNAs).
  • siRNAs small interfering RNAs
  • dsRNAs double stranded RNAs
  • shRNAs short hairpin RNAs
  • miRNAs miRNAs
  • a canonical siRNA comprises two strands, each a 21 -mer, forming a 19-bp double-stranded region and two 3' overhangs of 2 nt each.
  • other structures or formats of RNAi agents are known to be active, including blunt-ended 19-mers, or, as disclosed herein, blunt- ended 18-mers, or siRNAs with a shortened sense strand.
  • RNAi agents can comprise RNA subunits (ribonucleotides); however, one or more ribonucleotides can be modified and/or substituted. Modifications and substitutions include, as additional non-limiting examples, locked nucleic acid (LNA), Morpholino, threose nucleic acid (TNA), or glycol nucleic acid (GNA), arabinose nucleic acid (ANA), 2 ' -fluoroarabinose nucleic acid (FANA), cyclohexene nucleic acid (CeNA), anhydrohexitol nucleic acid (HNA), peptide nucleic acid (PNA), or UNA (unlocked nucleic acid), 2'-MOE, 2'-OMe, 2'-F, 2'-alkyl, or any other modification or substitution described herein or known in the art.
  • LNA locked nucleic acid
  • TAA threose nucleic acid
  • GAA glycol nucleic acid
  • RNAi agents also include molecules in which one or more strands are a mixture of RNA, DNA, LNA, morpholino, TNA, GNA, ANA, HNA, CeNA, FANA, UNA, and/or FANA, etc.
  • one or both strands of an RNAi agent could be, for example, RNA, except that one or more RNA nucleotides is replaced by DNA, LNA, morpholino, TNA, GNA, UNA, and/or FANA, etc.
  • one or both strands of the RNAi agent can be nicked, and both strands can be the same length, or one (e.g., the passenger strand), can be shorter than the other.
  • the present disclosure pertains to any RNAi agent comprising a RNA sequence disclosed herein and/or a RNA sequence corresponding to any DNA sequence disclosed herein (e.g., wherein the DNA nucleotides are replaced by the corresponding RNA nucleotide, for example, with T in DNA replaced by U in RNA, and with ribose instead of deoxyribose in the sugar-phosphate backbone).
  • the RNAi agent(s) of the present disclosure target (e.g., bind to, anneal to, hybridize, etc.) the APOC3 mRNA.
  • the use of the RNAi agent specific to APOC3 results in a decrease of APOC3 activity, level and/or expression, e.g., a "knock-down" (KD) or "knockout" of the target gene or target sequence.
  • KD knock-down
  • administration of a RNAi agent to APOC3 knocks down the APOC3 target enough to provide a more normal or therapeutic level of APOC3 activity or expression.
  • the patient or individual may have a disease state characterized by excessively high levels of APOC3 and the RNAi agent can restore a normal level.
  • the levels of APOC3 throughout the body are modulated such that APOC3 levels in one area (e.g., areas afflicted by an APOC3-related disease) are lower, while areas of the body not afflicted by the disease are closer to normal APOC3 levels.
  • the RNAi agent can be delivered locally so that levels of APOC3 outside the diseased areas can be maintained as close to normal as possible.
  • the level of APOC3 in the body can be modulated such that it is low enough to improve the disease state, but not so low that organ pathology occurs.
  • the RNAi comprises a single strand.
  • This single-stranded RNAi agent oligonucleotide or polynucleotide can comprise the sense or antisense strand, as described by Sioud 2005 J. Mol. Biol. 348:1079-1090, and references therein.
  • the disclosure encompasses RNAi agents with a single strand comprising either the sense or antisense strand of an RNAi agent described herein.
  • RNAi agents comprising a single strand, wherein the single strand comprises the sequences of both the antisense and sense strands of any RNAi agent disclosed herein, e.g., wherein the strands are contiguous, connected by a loop or otherwise linked.
  • RNAi agents include those with a hairpin between the sense and anti-sense sequences (e.g., shRNA).
  • one or both strands contain one or more nicks, i.e., a break or missing bond in the phosphate backbone, such that at least one nucleotide subunit is not covalently linked to the adjacent nucleotide subunit in any given sequence.
  • the passenger strand is nicked (see, for example, WO 2007/107162).
  • one or both strands contain one or more gaps, e.g., wherein at least one entire nucleotide subunit is absent from the disclosed sequence. Where a sense or antisense sequence contains a gap, that strand is envisioned to comprise two separate oligonucleotides.
  • Particularly useful siRNAs include those which can bind specifically to those regions of the APOC3 mRNA that have one or more of the following qualities: binding in the coding segment of APOC3; binding at or near the junction of the 5' untranslated region and the start of the coding segment; binding at or near the translational start site of the mRNA; binding at, across or near junctions of exons and introns; little or no binding to the mRNAs or transcripts of other genes (little or no "off-target effects"); binding to the APOC3 mRNA in or near a region or regions that is not double-stranded or a stem region, e.g., those in a loop or single-stranded portion; eliciting little or no immunogenicity; binding in a segment of the APOC3 mRNA sequence which is conserved among various animal species (including human, mouse, rat, cyno, etc.), as the presence of a conserved sequence facilitates testing using various laboratory animals; binding
  • double-stranded RNA refers to a RNAi agent comprising a first and a second strand; e.g., a composition that includes an RNA molecule or complex of molecules having a hybridized duplex region (i.e., a region where the nucleotide bases from the first strand and the second strand are paired) that comprises two anti-parallel and substantially complementary nucleic acid strands, which will be referred to as having "sense” and “antisense” orientations with respect to a target RNA.
  • a hybridized duplex region i.e., a region where the nucleotide bases from the first strand and the second strand are paired
  • the antisense strand with respect to the mRNA target, is also called the "guide” strand, and the sense strand is also called the “passenger” or “anti-guide” strand.
  • the passenger strand can include at least one or more of the following: one or more extra nucleotides (e.g., a bulge or 1 nt loop) compared to the other strand, and/or a nick, a gap, a mismatch, etc., compared to the other strand.
  • the RNAi agent comprises a first strand and a second strand.
  • first strand refers to the sense strand and the second strand refers to the anti-sense strand as listed in any Table herein.
  • first strand refers to the anti-sense strand
  • second strand refers to the sense strand as listed in any Table herein.
  • the duplex region can be of any length that permits specific degradation of a desired target RNA through a RISC pathway, but will typically range from 9 to 36 base pairs ("bp") in length, e.g., 15-30 bp in length.
  • the duplex can be any length in this range, for example, 9, 10, 1 1 , 12, 13, 14, 15, 16, 17, 18, 19, 20, 21 , 22, 23, 24, 25, 26, 27, 28, 29, 30, 31 , 32, 33, 34, 35, or 36 bp and any sub-range therebetween, including, but not limited to 15-30 bp, 15-26 bp, 15-23 bp, 15-22 bp, 15-21 bp, 15-20 bp, 15-19 bp, 15-18 bp, 15-17 bp, 18-30 bp, 18-26 bp, 18-23 bp, 18-22 bp, 18-21 bp, 18-20 bp, 19-30 bp, 19-26 bp, 19-23 bp, 19-22 bp, 19-21 bp, 19-20 bp, 19 bp, 20-30 bp, 20- 26 bp, 20-25 bp, 20-24 bp, 20-23 bp, 20-22
  • the dsRNAs generated in the cell by processing with Dicer and similar enzymes are generally in the range of about 19 to about 22 bp in length, though the strands of artificial dsRNAs can be shorter or longer.
  • siRNAs wherein one or both strands are as short as 16 or 15 nt still demonstrate RNA interference activity.
  • One strand of the duplex region of a dsRNA comprises a sequence that is substantially complementary to a region of a target RNA.
  • the two strands forming the duplex structure can be from a single RNA molecule having at least one self-complementary duplex region, or can be formed from two or more separate RNA molecules that hybridize to form the duplex.
  • the molecule can have a duplex region separated by a single-stranded chain of nucleotides (herein referred to as a "hairpin loop", e.g., such as found in an shRNA construct) between the 3'-end of one strand and the 5'-end of the respective other strand forming the duplex structure.
  • the hairpin loop can comprise at least one unpaired nucleotide; in some aspects the hairpin loop can comprise at least 3, at least 4, at least 5, at least 6, at least 7, at least 8, at least 9, at least 10, at least 20, at least 23 or more unpaired nucleotides.
  • dsRNA substantially complementary strands of a dsRNA
  • those molecules need not, but can, be covalently connected.
  • the construct is generally referred to herein and in the art as a "shRNA".
  • the connecting structure is referred to as a "linker.”
  • RNAi Agents to APOC3 comprising mismatches from the disclosed
  • RNAi agent to APOC3
  • RNAi agents which comprise sequences differing by 0, 1 , 2, or 3 nt (nucleotides) or bp [basepair(s)] (e.g., with 0, 1 , 2 or 3 mismatches) from any of the RNAi agents listed in any of the tables herein, and modified and unmodified variants thereof.
  • a mismatch is defined herein as a difference between the base sequence or length when two sequences are maximally aligned and compared.
  • a mismatch is defined as a position wherein the base of one sequence does not match the base of the other sequence.
  • a mismatch is counted, for example, if a position in one sequence has a particular base (e.g., A), and the corresponding position on the other sequence has a different base (e.g., G).
  • Substitution of A for example, with T, C, G or U would constitute a mismatch.
  • substitution of G with T, A, C or U would also constitute a mismatch.
  • C with T, G, A or U would also constitute a mismatch.
  • RNAi agent can thus comprise one or more DNA bases, e.g., T.
  • DNA can be used in place of RNA (e.g., in the seed region), to form a DNA-RNA hybrid. See, for example, Yamato et al. 201 1 cancer Gene Ther.
  • a mismatch is also counted, e.g., if a position in one sequence has a base (e.g., A), and the corresponding position on the other sequence has no base (e.g., that position is a spacer [e.g., in the internal spacer format described herein] or an abasic nucleotide, which comprises a phosphate-sugar backbone but no base).
  • a base e.g., A
  • no base e.g., that position is a spacer [e.g., in the internal spacer format described herein] or an abasic nucleotide, which comprises a phosphate-sugar backbone but no base.
  • the sequences are considered to have one mismatch (e.g., to differ by 1 nt).
  • a single-stranded nick in either sequence (or in the sense or anti-sense strand) is not counted as mismatch.
  • no mismatch would be counted if one sequence comprises the sequence AG, but the other sequence comprises the sequence AG with a single- stranded nick between the A and the G.
  • a nucleotide modification in the sugar or phosphate is also not considered a mismatch.
  • one sequence comprises a C
  • the other sequence comprises a modified C (e.g., 2'-modification) at the same position, no mismatch would be counted.
  • RNAi agent no mismatches are counted if modifications are made to the sugar, phosphate, or backbone of the RNAi agent without modifying the base.
  • a strand having a particular sequence of as an RNA would have zero mismatches from another strand having the same sequence as a PNA; or morpholino; or LNA; or TNA; or GNA; or UNA; or FANA; or a mix or chimera of RNA and DNA, TNA, GNA, UNA, FANA, morpholino, LNA, and/or PNA, etc.
  • RNAi agents in the Tables include sequences which comprise modifications. It is noted that dTdT (2'-deoxy-thymidine-5'- phosphate and 2'-deoxy-thymidine-5'-phosphate), or in some cases, TT or UU, can be added as a terminal dinucleotide cap or extension to one or both 3'-ends, but this cap or extension is not included in the calculation of the total number of mismatches and is not considered part of the target sequence. This is because the terminal dinucleotide protects the ends from nuclease degradation but does not contribute to target specificity (Elbashir et al. 2001 Nature 41 1 : 494-498; Elbashir et al. 2001 EMBO J. 20: 6877-6888; and Kraynack et al. 2006 RNA 12:163-176).
  • a modified variant can have one or more modifications from the corresponding unmodified sequence.
  • lowercase “c” represents 2'-0- methylcytidine-5'-phosphate
  • lowercase “u” represents 2'-0-methyluridine-5'-phosphate.
  • Uppercase “A”, “C”, “G” and “U” represent the un-modified adenosine-5'-phosphate, cytidine- 5'-phosphate, guanosine-5'-phosphate, and uridine-5'-phosphate, respectively.
  • the substitution, for example, of modified c for unmodified C does not count as a mismatch in numbering the 0, 1 , 2, or 3 mismatches between sequences.
  • the present disclosure comprises a RNAi agent comprising a anti-sense strand comprising at least 15 to 19 contiguous nucleotides differing by 0, 1 , 2, or 3 nt from the sequence of the anti-sense strand of: any of the RNAi agents listed in any of the tables herein, and modified and unmodified variants thereof.
  • the present disclosure pertains to "modified and unmodified variants" of the disclosed sequences.
  • An "unmodified variant" of a particular sequence is the corresponding portion of APOC3 without any modifications.
  • Example modified sequences are disclosed herein
  • the "unmodified variants" of the sequences of the Tables have the identical sequence, without base modifications or terminal dTdT.
  • a given sequence and an “unmodified variant” of it differ by 0 nt (and have no mismatches).
  • a "modified variant" of a particular sequence comprises one or more (or one or more fewer) modifications to the backbone, sugar, phosphate or base, and/or addition of a terminal dinucleotide (e.g., TT, dTdT, TsT or UU), but do not have any base substitutions (e.g., G for C, or A for G); thus a given sequence and a modified variant thereof differ by 0 nt (and have no mismatches).
  • a given sequence as a RNA and the same sequence as a PNA are modified variants of each other and differ by 0 nt (and have no mismatches).
  • LNA locked nucleic acid
  • TNA threose nucleic acid
  • GNA glycol nucleic acid
  • UNA unlocked nucleic acid
  • FANA or ANA or HNA or CeNA FANA or ANA or HNA or CeNA, etc.
  • one or both strands could be, for example, RNA except that one or more nucleotides is replaced by DNA, LNA, morpholino, TNA, GNA, ANA, HNA, CeNA, UNA, and/or FANA, etc.
  • the RNAi agent comprises a sense strand comprising at least 14 contiguous nucleotides differing by 0, 1 , 2, or 3 nt from the sense strand of any of the RNAi agents listed in the Tables and modified and unmodified variants thereof.
  • RNAi agents to APOC3 of the present disclosure can be used in RNA interference.
  • RNAi Agents [00399] Modifications of RNAi Agents.
  • the present disclosure encompasses both unmodified and example modified RNAi agents, such as those disclosed in the Tables.
  • RNAi agent e.g., a modified variant
  • the disclosure encompasses a RNAi agent with a substitution of a single nucleotide at a given position with a modified version of the same nucleotide.
  • a nucleotide (A, G, C or U) can be replaced by the corresponding 5-fluorouracil, 5-bromouracil, 5-chlorouracil, 5-iodouracil, hypoxanthine, xantine, 4-acetylcytosine, 5- (carboxyhydroxylmethyl) uracil, 5-carboxymethylaminomethyl-2-thiouridine, 5- carboxymethylaminomethyluracil, dihydrouracil, beta-D-galactosylqueosine, inosine, N6- isopentenyladenine, 1 -methylguanine, 1 -methylinosine, 2,2-dimethylguanine, 2- methyladenine, 2-methylguanine, 3-methylcytosine, 5-methylcytosine, N6-aden
  • Additional modified variants include the addition of any other moiety (e.g., a radiolabel or other tag or conjugate) to the RNAi agent; provided that the base sequence is identical, the addition of other moieties produces a "modified variant" (with no mismatches).
  • any other moiety e.g., a radiolabel or other tag or conjugate
  • RNAi agents to APOC3 of the present disclosure can be used in RNA interference.
  • RNA interference is a post-transcriptional, targeted gene-silencing technique that uses double-stranded RNA (dsRNA) to degrade messenger RNA (mRNA) containing the same sequence as the dsRNA.
  • dsRNA double-stranded RNA
  • mRNA messenger RNA
  • RNAi RNA interference
  • Dicer ribonuclease III
  • siRNAs small interfering RNAs
  • Dicer are typically about 21 to 23 nucleotides long and comprise about 19 base pair duplexes (though artificial siRNAs or RNAi agents can be shorter or longer, and/or blunt-ended, and/or comprises one or more endcaps).
  • RNA-induced silencing complex RISC
  • an RNA interference agent includes a single-stranded RNA that interacts with a target RNA sequence to direct the cleavage of the target RNA.
  • a target RNA sequence to direct the cleavage of the target RNA.
  • Dicer Type III endonuclease known as Dicer. Sharp et al. 2001 Genes Dev. 15:485.
  • Dicer a ribonuclease- lll-like enzyme, processes the dsRNA into 19-23 base pair short interfering RNAs with characteristic two base 3' overhangs. Bernstein, et al. 2001 Nature 409:363.
  • RNA-induced silencing complex RISC
  • one or more helicases unwind the siRNA duplex, enabling one of the now unpaired siRNA strands to act as a "guide" strand to guide target recognition.
  • Nykanen et al. 2001 Cell 107:309.
  • one or more endonucleases within the RISC cleaves the target to induce silencing.
  • the present disclosure relates to a single-stranded RNA that promotes the formation of a RISC complex to effect silencing of the target gene.
  • RNAi agent can be selected by any process known in the art or conceivable by one of ordinary skill in the art.
  • the selection criteria can include one or more of the following steps: initial analysis of the APOC3 gene sequence and design of RNAi agents; this design can take into consideration sequence similarity across species (human, cynomolgus, mouse, etc.) and dissimilarity to other (non-APOC3) genes; screening of RNAi agents in vitro (e.g., at 10 nM in RKO cells); determination of EC50 in RKO cells; determination of viability of cells treated with RNAi agents, including insensitive cells which do not require APOC3 for survival, or sensitive cells, which do require APOC3 for survival; testing with human PBMC (peripheral blood mononuclear cells), e.g., to test levels of TNF- alpha to estimate immunogenicity, wherein immunostimulatory sequences are less desired; testing in human whole blood assay, wherein fresh human blood is treated
  • PBMC peripheral blood
  • pharmacodynamic (PD) marker for example, other factors whose expression is affected by APOC3, wherein APOC3 knockdown leads to a dose-dependent reduction of abundance of those components; and optimization of specific modifications of the RNAi agents.
  • RNAi agents dsRNA molecules (RNAi agents) described herein are thus useful in RNA interference of APOC3.
  • RNAi agents Sense strand, Antisense Strand and (Optional) Overhangs.
  • the RNAi agents comprise a first strand and a second strand, e.g., a sense strand and an antisense strand (or an antisense and a sense strand), optionally, either or both both ends of either or both strand can comprise unpaired nucleotides (referred to herein as "overhangs").
  • antisense strand refers to the strand of a RNAi agent which includes a region that is substantially complementary to a target sequence.
  • region of complementarity refers to the region on the antisense strand that is substantially complementary to a sequence, for example a target sequence, as defined herein. Where the region of complementarity is not fully complementary to the target sequence, the
  • mismatches may be in the internal or terminal regions of the molecule. Generally, the most tolerated mismatches are in the terminal regions, e.g., within 5, 4, 3, or 2 nucleotides of the 5' and/or 3' terminus.
  • sense strand refers to the strand of a RNAi agent that includes a region that is substantially complementary to a region of the antisense strand as that term is defined herein.
  • sequence of a gene may vary from individual to individual, especially at wobble positions within the coding segment, or in the untranslated region; individuals may also differ from each other in coding sequence, resulting in additional differences in mRNA.
  • the sequence of the sense and antisense strands of the RNAi agent can thus be designed to correspond to that of an individual patient, if and where needed.
  • RNAi agents can also be modified in sequence to reduce immunogenicity, binding to undesired mRNAs (e.g., "off- target effects") or to increase stability in the blood.
  • RNAi agents independent of chemical modification of the bases or 5' or 3' or other end-caps of the RNAi agents.
  • RNAi agents can also have overhangs of 0, 1 , or 2 overhangs; in the case of a 0 nt overhang, they are blunt-ended. A RNAi agent can thus have 0, 1 or 2 blunt ends. In a "blunt-ended RNAi agent" both strands terminate in a base-pair; thus a blunt-ended molecule lacks either 3' or 5' single-stranded nucleotide overhangs.
  • RNAi agents can comprise overhang(s), blunt end(s), and/or 5' and 3' endcap(s).
  • the term "overhang” or “nucleotide overhang” refer to at least one unpaired nucleotide that protrudes from the end of at least one of the two strands of the duplex structure of a RNAi agent. For example, when a 3'-end of one strand of a dsRNA extends beyond the 5'-end of the other strand, or vice versa, this forms a nucleotidic overhang, e.g., the unpaired nucleotide(s) form the overhang.
  • a dsRNA can comprise an overhang of at least one nucleotide; alternatively the overhang can comprise at least two nucleotides, at least three nucleotides, at least four nucleotides, at least five nucleotides or more.
  • An overhang can comprise or consist of a nucleotide/nucleoside analog, including a deoxynucleotide/nucleoside. The overhang(s) may be on the sense strand, the antisense strand or any combination thereof.
  • the nucleotide(s) of an overhang can be present on the 5' end, 3' end or both ends of either an antisense or sense strand of a dsRNA.
  • the RNAi agent can also optionally comprise a cap.
  • the term "Cap” and the like include a chemical moiety attached to the end of a double-stranded nucleotide duplex, but is used herein to exclude a chemical moiety that is a nucleotide or nucleoside.
  • a "3' Cap” is attached at the 3' end of a nucleotide or oligonucleotide and protects the molecule from degradation, e.g., from nucleases, such as those in blood serum or intestinal fluid.
  • a non- nucleotidic 3' cap is not a nucleotide and can replace a TT or UU dinucleotide at the end of a blunt-ended RNAi agent.
  • non-nucleotidic 3' end caps are as disclosed in, for example, WO 2005/021749 and WO 2007/128477; and U.S. Pat. No. 8,097,716; U.S. Pat. No. 8,084,600; and U.S. Pat. No. 8,344,128.
  • a "5' cap” is attached at the 5' end of a nucleotide or oligonucleotide. A cap should not interfere (or unduly interfere) with RNAi activity.
  • RNAi agent specific to APOC3 comprising an antisense strand (which may be contiguous or connected via a linker or loop) in a RNAi agent.
  • an RNAi agent comprises an antisense strand and a sense strand which together comprise a double-stranded or complementary region. In one aspect, it can also optionally comprise one or two overhangs and/or one or two caps.
  • the RNAi agent is used to induce RNA interference of the target gene, APOC3.
  • RNAi agents of the present disclosure target (e.g., specifically bind to, anneal to, etc.) the mRNA encoding APOC3.
  • the use of the RNAi agent specific to APOC3 results in a decrease of APOC3 activity, level and/or expression.
  • administration of a RNAi agent to APOC3 knocks down the APOC3 gene enough to restore a normal level of APOC3 activity or expression.
  • the first or second strand of the RNAi comprises a sequence complementary to that of the target nucleic acid, APOC3.
  • strand comprising a sequence refers to an
  • oligonucleotide comprising a chain of nucleotides that is described by the sequence referred to using the standard nucleotide nomenclature.
  • target sequence or "target gene” refer to a contiguous portion of the nucleotide sequence of an mRNA molecule formed during the transcription of a gene, e.g., an APOC3 gene, including mRNA that is a product of RNA processing of a primary transcription product.
  • the target portion of the sequence will be at least long enough to serve as a substrate for RNAi-directed cleavage at or near that portion.
  • the target sequence will generally be from 9-36 nucleotides (“nt”) in length, e.g., 15-30 nt in length, including all sub-ranges therebetween.
  • the target sequence can be from 15-30 nt, 15-26 nt, 15-23 nt, 15-22 nt, 15-21 nt, 15-20 nt, 15-19 nt, 15-18 nt, 15-17 nt, 18-30 nt, 18-26 nt, 18-23 nt, 18-22 nt, 18-21 nt, 18-20 nt, 19-30 nt, 19-26 nt, 19-23 nt, 19-22 nt, 19-21 nt, 19-20 nt, 19 nt, 20-30 nt, 20-26 nt, 20-25 nt, 20-24 nt, 20-23 nt, 20-22 nt, 20-21 nt, 20 nt, 21-30 nt, 21 -26 nt, 21-25 nt, 21 -24 nt, 21-23 nt, or 21 -22 nt, 21 nt, 22 nt, or 23 nt.
  • the term “complementary” refers to the ability of an oligonucleotide or polynucleotide comprising a first nucleotide sequence to hybridize and form a duplex structure under certain conditions with an oligonucleotide or polynucleotide comprising a second nucleotide sequence.
  • Such conditions can, for example, be stringent, e.g., 400 mM NaCI, 40 mM PIPES pH 6.4, 1 mM EDTA, 50°C or 70°C for 12-16 hours followed by washing.
  • Other conditions such as physiologically relevant conditions as may be encountered inside an organism, can apply. The skilled person will be able to determine the set of conditions most appropriate for a test of complementarity of two sequences in accordance with the ultimate application of the hybridized nucleotides.
  • RNAi agent e.g., within a dsRNA as described herein
  • oligonucleotides or polynucleotides comprising a first nucleotide sequence to an oligonucleotide or polynucleotide comprising a second nucleotide sequence over the entire length of one or both nucleotide sequences.
  • sequences can be referred to as "fully complementary” with respect to each other herein.
  • first sequence is referred to as “substantially complementary” with respect to a second sequence herein
  • the two sequences can be fully complementary, or they may form one or more, but generally not more than 5, 4, 2 or 2 mismatched base pairs upon hybridization for a duplex up to 30 base pairs, while retaining the ability to hybridize under the conditions most relevant to their ultimate application, e.g., inhibition of gene expression via a RISC pathway.
  • a dsRNA comprising one oligonucleotide 21 nucleotides in length and another oligonucleotide 23 nucleotides in length, wherein the longer oligonucleotide comprises a sequence of 21 nucleotides that is fully complementary to the shorter oligonucleotide, may yet be referred to as "fully complementary" for the purposes described herein.
  • overhang describes an unpaired nucleotide at the 3' or 5' end of a double-stranded nucleotide duplex, as described above. In one aspect, the overhang is 1 to 4 nt long and is on the 3' end.
  • “Complementary" sequences may also include, or be formed entirely from, non-Watson-Crick base pairs and/or base pairs formed from non-natural and modified nucleotides, in as far as the above requirements with respect to their ability to hybridize are fulfilled.
  • Such non-Watson-Crick base pairs includes, but are not limited to, Wobble or Hoogstein base pairing.
  • the terms “complementary,” “fully complementary” and “substantially complementary” herein may furthermore be used with respect to the base matching between the sense strand and the antisense strand of a dsRNA, or between the antisense strand of a RNAi agent and a target sequence, as will be understood from the context of their use.
  • a polynucleotide that is "substantially complementary to at least part of a messenger RNA (mRNA) refers to a polynucleotide that is substantially complementary to a contiguous portion of the mRNA of interest (e.g., an mRNA encoding APOC3).
  • mRNA messenger RNA
  • a polynucleotide is complementary to at least a part of an APOC3 mRNA if the sequence is substantially complementary to a non-interrupted portion of an mRNA encoding APOC3.
  • the RNAi agent of the present disclosure is complimentary or substantially complimentary to a target sequence in the target APOC3 and is double-stranded, comprising a sense and an antisense strand (which can be contiguous, linked via a loop, or otherwise joined), where the double-stranded region an be 9 to 36 bp long (particularly for example, 19-22 bp or 19-23 bp long), and can furthermore optionally comprise a 3' or 5' overhang, and the RNAi agent can furthermore comprise a 3' cap.
  • RNAi agent mediates RNA interference, down-regulating or inhibiting or reducing the level, expression and/or activity of APOC3, and/or establishing or re-establishing an approximately normal level of APOC3 and/or APOC3 activity, or other biological function related to APOC3.
  • the RNAi agent of the present disclosure is complimentary or substantially complimentary to a target sequence in the target APOC3 and is double-stranded, comprising a sense and an antisense strand (which can be contiguous, linked via a loop, or otherwise joined), where the double-stranded region an be 9 to 36 bp long (particularly for example, 19-22 bp or 19-23 bp long), and can furthermore optionally comprise a 3' or 5' overhang, and the RNAi agent can furthermore comprise a 3' cap.
  • the RNAi agent mediates RNA interference, down-regulating or inhibiting the level, expression and/or activity of APOC3, and/or establishing or re-establishing an approximately normal level of APOC3 activity or expression.
  • double-stranded RNA refers to an RNAi agent comprising a first and a second strand; e.g., a composition that includes an RNA molecule or complex of molecules having a hybridized duplex region that comprises two anti-parallel and substantially complementary nucleic acid strands, which will be referred to as having "sense” and “antisense” orientations with respect to a target RNA.
  • the antisense strand, with respect to the mRNA target is also called the “guide” strand, and the sense strand is also called the “passenger” strand.
  • the "first" strand can be the guide or antisense strand
  • the "second” strand can be the passenger or sense strand
  • the "first” strand can be the passenger or sense strand
  • the "second” strand can be the guide or antisense.
  • the passenger strand can include at least one or more of the following: one or more extra nucleotides (e.g., a bulge or 1 nt loop) compared to the other strand, a nick, a gap, etc., compared to the other strand.
  • the first strand is the sense strand and the second strand is the anti-sense strand.
  • the first strand is the anti-sense strand
  • the second strand is the sense strand.
  • the duplex region can be of any length that permits loading into the RISC complex and subsequent specific degradation of a desired target RNA through a RISC pathway, but will typically range from 9 to 36 base pairs ("bp") in length, e.g., 15-30 base pairs in length.
  • the duplex can be any length in this range, for example, 9, 10, 1 1 , 12, 13, 14, 15, 16, 17, 18, 19, 20, 21 , 22, 23, 24, 25, 26, 27, 28, 29, 30, 31 , 32, 33, 34, 35, or 36 bp and any sub-range therebetween, including, but not limited to 15-30 base pairs, 15-26 bp, 15-23 bp, 15-22 bp, 15-21 bp, 15-20 bp, 15-19 bp, 15- 18 bp, 15-17 bp, 18-30 bp, 18-26 bp, 18-23 bp, 18-22 bp, 18-21 bp, 18-20 bp, 19-30 bp, 19- 26 bp, 19-23 bp, 19-22 bp, 19-21 bp, 19-20 bp, 19 bp, 20-30 bp, 20-26 bp, 20-25 bp, 20-24 bp, 20-23 bp, 20-22 bp and any sub-range therebetween, including,
  • the dsRNAs generated in the cell by processing with Dicer and similar enzymes are generally in the range of about 19 to about 22 base pairs in length, although artificial RNAi agents can be synethesized or made by any method known in the art.
  • One strand of the duplex region of a dsRNA comprises a sequence that is substantially complementary to a region of a target RNA.
  • the two strands forming the duplex structure can be from a single RNA molecule having at least one self-complementary duplex region, or can be formed from two or more separate RNA molecules that hybridize to form the duplex.
  • the molecule can have a duplex region separated by a single stranded chain of nucleotides (herein referred to as a "hairpin loop", e.g., such as found in an shRNA construct) between the 3'- end of one strand and the 5'-end of the respective other strand forming the duplex structure.
  • the hairpin loop can comprise at least one unpaired nucleotide; in some aspects the hairpin loop can comprise at least 3, at least 4, at least 5, at least 6, at least 7, at least 8, at least 9, at least 10, at least 20, at least 23 or more unpaired nucleotides.
  • RNA molecules where the two substantially complementary strands of a dsRNA are comprised by separate RNA molecules, those molecules need not, but can be covalently connected.
  • the construct is generally referred to herein and in the art as a "shRNA".
  • the connecting structure is referred to as a "linker.”
  • siRNA is also used herein to refer to a dsRNA as described above.
  • RNAi agents for targeting APOC3 include those which bind to an APOC3 sequence provided herein and which work to reduce APOC3 through a RNAi mechanism.
  • Example RNAi agents (e.g., siRNAs) to APOC3 are provided herein.
  • RNAi agents of the present disclosure silence, inhibit the expression of, down- regulate the expression of, and/or suppress the expression of APOC3, such that an approximately normal level of APOC3 activity or expression is restored.
  • RNAi agents of the present disclosure it is acceptable to use to establish a level of APOC3 expression, activity and/or level which is below the normal level, or above the normal level, depending on the therapeutic outcome that is desired.
  • any method known in the art can be use to measure changes in APOC3 activity, level and/or expression induced by an APOC3 siRNA. Measurements can be performed at multiple timepoints, prior to, during and after administration of the siRNA, to determine the effect of the siRNA.
  • the degree of inhibition is usually expressed in terms of
  • the degree of inhibition may be given in terms of a reduction of a parameter that is functionally linked to APOC3 gene expression, e.g., the amount of protein encoded by an APOC3 gene, alteration in expression of a protein whose expression is dependent on APOC3, etc.
  • APOC3 gene silencing may be determined in any cell expressing APOC3, either constitutively or by genomic engineering, and by any appropriate assay.
  • the assays provided in the Examples below shall serve as such reference.
  • expression of APOC3 is suppressed by at least about 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, or 50% by administration of a RNAi agent featured in the present disclosure.
  • APOC3 is suppressed by at least about 60%, 70%, or 80% by administration of a RNAi agent featured in the present disclosure.
  • APOC3js suppressed by at least about 85%, 90%, or 95% or more by administration of a RNAi agent, as described herein.
  • the degree of APOC3 suppression is determined by loss of full length APOC3 mRNA in a treated cell compared to an untreated cell.
  • the degree of APOC3 suppression is determined with a phenotypic assay that monitors loss of proliferative activity and/or cell death. Other aspects are as provided in the Examples.
  • RNAi agent can be selected by any process known in the art or conceivable by one of ordinary skill in the art.
  • the selection criteria can include one or more of the following steps: initial analysis of the APOC3 gene sequence and design of RNAi agents; this design can take into consideration sequence similarity across species (human, cynomolgus, mouse, etc.) and dissimilarity to other (non-APOC3) genes; screening of RNAi agents in vitro (e.g., at 10 nM and 1 nM in Huh-7 cells); selection of RNAi agents with high knock-down at 10 nM and 1 nM in Huh-7 cells; determination of EC50 in Huh-7 cells; confirmation of EC50 in a RCC cell line (Huh-7 cells); analysis of a lack of effect on cell growth relative to a control siRNA; reduction in Huh-7 cells of expression of a HRE-luc (luciferase) reporter gene and not control UB6-luc (luciferase)
  • APOC3 target gene modulation analysis e.g., using a pharmacodynamic (PD) marker, for example, EGLN3, SLC2A1 and VEGF, wherein APOC3 knockdown leads to a dose- dependent alteration of EGLN3, SLC2A1 and VEGF expression in cells; and optimization of specific modifications of the RNAi agents.
  • PD pharmacodynamic
  • other cell lines can be used in place of those listed above to identify RNAi agents capable of lowering APOC3 levels or decrease symptoms of an APOC3-related disease.
  • a symptom of an APOC3-related disease is meant a statistically significant decrease in such level.
  • the decrease can be, for example, at least 10%, at least 20%, at least 30%, at least 40% or more. If, for a particular disease, or for an individual suffering from a particular disease, the levels or expression of APOC3 are elevated, treatment with an APOC3 RNAi agent of the present disclosure can particularly reduce the level or expression of APOC3 to a level considered in the literature as within the range of normal for an individual without such disorder, or to a level that reduces or ameliorates symptoms of a disease.
  • the level or expression of APOC3 can be measured by evaluation of mRNA (e.g., via Northern blots or PCR), or protein (e.g., Western blots).
  • mRNA e.g., via Northern blots or PCR
  • protein e.g., Western blots
  • the effect of a RNAi agent on APOC3 expression can be determined by measuring APOC3 gene transcription rates (e.g., via Northern blots; or reverse transcriptase polymerase chain reaction or real-time polymerase chain reaction).
  • down-regulates refers to any statistically significant decrease in a biological activity and/or expression of APOC3, including full blocking of the activity (i.e., complete inhibition) and/or expression.
  • down-regulation can refer to a decrease of at least about 10, 20, 30, 40, 50, 60, 70, 80, 90 or 100 % in APOC3 level, activity and/or expression.
  • the term “inhibit” or “inhibiting” APOC3 refers to any statistically significant decrease in biological level, activity and/or expression of APOC3, including full blocking of the activity and/or expression.
  • “inhibition” can refer to a decrease of at least about 10, 20, 30, 40, 50, 60, 70, 80, 90 or 100 % in APOC3 level, activity and/or expression.
  • the term “inhibit” similarly refers to a significant decrease in level, activity and/or expression, while referring to any other biological agent or composition.
  • level it is meant that the APOC3 RNAi agent can alter the level of APOC3, e.g., the level of APOC3 mRNA or the level of APOC3 protein, or the level of activity of APOC3.
  • Some diseases include any APOC3-related disease disclosed herein or known in the literature. Particularly in one aspect, in the case of a disease characterized by over- expression and/or hyper-activity of APOC3, administration of a RNAi agent to APOC3 reduces the level, expression and/or activity of APOC3. Thus, in various aspects, administration of a RNAi agent to APOC3 particularly establishes or re-establishes a normal or approximately normal level of APOC3 activity, expression and/or level.
  • normal or “approximately normal” in terms of level, expression and/or activity, is meant at least: about 50%, about 60%, about 70%, about 80%, about 90%, and/or about 100%; and/or no more than: about 100%, about 120%, about 130%, about 140%, or about 150% of the level, expression and/or activity of APOC3 in a healthy cell, tissue, or organ. This can be measured using, for example, lung or kidney homogenates, as described in Gambling et al. 2 Kidney Intl. 65: 1774-1781.
  • administration of the appropriate amount of the appropriate APOC3 RNAi agent restores APOC3 level, activity and/or expression to about 50% to about 150%, more particularly about 60% to about 140%, more particularly to about 70% to about 130%, more particularly to about 80% to about 120%, more particularly to about 90% to about 1 10%, and most particularly to about 100% of that of a healthy cell, tissue or organ.
  • Administration of an APOC3 RNAi to a patient with an APOC3-related disease thus particularly restores the level, activity, and/or expression of APOC3 and the level of Na + reabsorption to an approximately normal level, as determined by direct measurements of APOC3 mRNA or protein levels, or indirect determinations.
  • the preferred target amount of APOC3 level, expression and/or activity after APOC3 RNAi agent administration can be calculated to take into account any other perturbations in an APOC3-related pathway. For example, if another factor in an APOC3- related pathway is either over- or under-expressed, APOC3 level, expression or activity may be modulated to attain a more normal state.
  • RNAi agents of the present disclosure it is acceptable to use to establish a level of APOC3 expression, activity and/or level which is below the normal level, or above the normal level.
  • RNAi agents or compositions comprising an antisense nucleic acid to down-modulate the expression of a particular protein in a cell is well known in the art.
  • a RNAi agent comprises a sequence complementary to, and is capable of hydrogen bonding to, the coding strand of another nucleic acid (e.g., an mRNA).
  • the RNAi agents of the present disclosure encompass any RNAi agents which target (e.g., are complementary, capable of hybridizing or hydrogen bonding to, etc.) any sequence presented, e.g., in any of the Tables.
  • RNAi agent may have
  • RNAi agents internally, or at one or both ends.
  • the modifications at the ends can help stabilize the RNAi agent, protecting it from degradation by nucleases in the blood.
  • the RNAi agents may optionally be directed to regions of the APOC3 mRNA known or predicted to be near or at splice sites of the gene.
  • RNAi agent can be constructed using chemical synthesis and enzymatic ligation reactions using procedures known in the art.
  • RNAi agent can be chemically synthesized using naturally-occurring nucleotides or variously modified nucleotides designed to decrease off-target effects, and/or increase the biological stability of the molecules or to increase the physical stability of the duplex formed between the antisense and sense nucleic acids, e.g., phosphorothioate derivatives and acridine substituted nucleotides can be used.
  • G,” “C,” “A,” “T” and “U” each generally stand for a nucleotide that contains guanine, cytosine, adenine, thymidine and uracil as a base, respectively.
  • ribonucleotide can also refer to a modified nucleotide or a surrogate replacement moiety.
  • guanine, cytosine, adenine, and uracil may be replaced by other moieties without substantially altering the base pairing properties of an oligonucleotide comprising a nucleotide bearing such replacement moiety.
  • a nucleotide comprising inosine as its base may base pair with nucleotides containing adenine, cytosine, or uracil.
  • nucleotides containing uracil, guanine, or adenine may be replaced in the nucleotide sequences of dsRNA featured in the present disclosure by a nucleotide containing, for example, inosine.
  • adenine and cytosine anywhere in the oligonucleotide can be replaced with guanine and uracil, respectively to form Wobble base pairing with the target mRNA. Sequences containing such replacement moieties are suitable for the compositions and methods featured in the present disclosure.
  • RNA molecule or "ribonucleic acid molecule” encompasses not only RNA molecules as expressed or found in nature (i.e., are naturally occurring), but also non-naturally occurring analogs and derivatives of RNA comprising one or more ribonucleotide/ribonucleoside analogs or derivatives as described herein or as known in the art.
  • the RNA can be modified in the nucleobase structure or in the ribose-phosphate backbone structure, e.g., as described herein below.
  • the molecules comprising ribonucleoside analogs or derivatives can retain the ability to form a duplex.
  • either or both strand of an RNAi agent can comprise at least one modified ribonucleoside, including but not limited to a 2'-0-methyl modified nucleotide, a nucleoside comprising a 5' phosphorothioate group, a terminal nucleoside linked to a cholesteryl derivative or dodecanoic acid bisdecylamide group, a locked nucleoside, an abasic nucleoside, a 2'-deoxy-2'-fluoro modified nucleoside, a 2'-amino- modified nucleoside, 2'-alkyl-modified nucleoside, morpholino nucleoside, an unlocked ribonucleotide (e.g., an acyclic nucleotide monomer, as described in WO 2008/147824), a phosphoramidate or a non-natural base comprising nucleoside, or any combination thereof.
  • a 2'-0-methyl modified nucleotide
  • an RNA molecule can comprise at least two modified ribonucleosides, at least 3, at least 4, at least 5, at least 6, at least 7, at least 8, at least 9, at least 10, at least 15, at least 20 or more, up to the entire length of the dsRNA molecule.
  • the modifications need not be the same for each of such a plurality of modified ribonucleosides in an RNA molecule.
  • modified RNAs contemplated for use in methods and compositions described herein are peptide nucleic acids (PNAs) that have the ability to form the required duplex structure and that permit or mediate the specific degradation of a target RNA via a RISC pathway.
  • PNAs peptide nucleic acids
  • the RNAi agent can comprise one or two strands which are a RNA, or a mixture of RNA, DNA, LNA, morpholino, TNA, GNA, ANA, HNA, CeNA, UNA, and/or FANA, etc.
  • one or both strands could be, for example, RNA except that one or more nucleotides is replaced by DNA, LNA, morpholino, TNA, GNA, ANA, HNA, CeNA, FANA, and/or UNA, etc.
  • modified nucleotides which can be used to generate the RNAi agent include 5-fluorouracil, 5-bromouracil, 5-chlorouracil, 5-iodouracil, hypoxanthine, xantine, 4- acetylcytosine, 5-(carboxyhydroxylmethyl) uracil, 5-carboxymethylaminomethyl-2-thiouridine, 5-carboxymethylaminomethyluracil, dihydrouracil, beta-D-galactosylqueosine, inosine, N6- isopentenyladenine, 1 -methylguanine, 1 -methylinosine, 2,2-dimethylguanine, 2- methyladenine, 2-methylguanine, 3-methylcytosine, 5-methylcytosine, N6-adenine, 7- methylguanine, 5-methylaminomethyluracil, 5-methoxyaminomethyl-2-thiouracil, beta-D- mannosylqueosine
  • a "modified variant" of a sequence disclosed herein includes any variant comprising the same sequence, but with a modification in the base, sugar, phosphate or backbone (but not a base substitution, e.g., A for G, or C for U).
  • a modified variant can comprise any modified nucleotide described above (e.g., 5-fluorouracil, 5-bromouracil, 5-chlorouracil, 5-iodouracil, hypoxanthine, xantine, 4-acetylcytosine, 5- (carboxyhydroxylmethyl) uracil, etc.).
  • a base is replaced by a corresponding modified base (e.g., A for modified A)
  • these modified nucleotides do not constitute a mismatch or base difference.
  • a given sequence with a U at a particular position and a modified variant comprising a 5-fluorouracil, 5-bromouracil, 5-chlorouracil, or 5-iodouracil at the same sequence would differ by 0 nt (or have no mismatches); however, a given sequence with a C at a particular position and a different sequence with a 5-fluorouracil (wherein the two sequences are otherwise identical) would differ by 1 nt (1 mismatch).
  • siRNA may include modifications to either the phosphate-sugar backbone or the nucleoside to include at least one of a nitrogen or sulfur heteroatom.
  • Kreutzer et al. Canadian Patent Application No. 2,359, 180 also describe certain chemical modifications for use in dsRNA constructs in order to counteract activation of double-stranded RNA- dependent protein kinase PKR, specifically 2'-amino or 2'-0-methyl nucleotides, and nucleotides containing a 2'-0 or 4'-C methylene bridge.
  • Additional 3'-terminal nucleotide overhangs include dT (deoxythimidine), 2'-0,4'-C-ethylene thymidine (eT), and 2- hydroxyethyl phosphate (hp). 4-thiouracil and 5-bromouracil substitutions can also be made. Parrish et al. 2000 Molecular Cell 6: 1077-1087. [00456] Those skilled in the art will appreciate that it is possible to synthesize and modify the siRNA as desired, using any conventional method known in the art (see Henschel et al. 2004 DEQOR: a web-based tool for the design and quality control of siRNAs. Nucleic Acids Research 32 (Web Server Issue): W1 13-W120).
  • RNAi agent is a shRNA
  • regulatory sequences for example, constitutive or inducible promoters, tissue-specific promoters or functional fragments thereof, etc.
  • nucleic acid molecules there are several examples in the art describing sugar, base, phosphate and backbone modifications that can be introduced into nucleic acid molecules with significant enhancement in their nuclease stability and efficacy.
  • oligonucleotides are modified to enhance stability and/or enhance biological activity by modification with nuclease resistant groups, for example, 2'-amino, 2'-C-allyl, 2'-flouro, 2'-0-methyl, 2'-0-allyl, 2'-H, nucleotide base modifications (for a review see Usman and Cedergren 1992 TIBS. 17: 34; Usman et al. 1994 Nucleic Acids Symp. Ser. 31 : 163; Burgin et al. 1996 Biochemistry 35: 14090).
  • Sugar modification of nucleic acid molecules are extensively described in the art.
  • the RNAi agent comprises a 2'-modification selected from the group consisting of: 2'-deoxy, 2'-deoxy-2'-fluoro, 2'-0-methyl, 2'-0-methoxyethyl (2'-0-MOE), 2'-0-aminopropyl (2 -O-AP), 2'-0-dimethylaminoethyl (2 -O-DMAOE), 2 -0- dimethylaminopropyl (2 -O-DMAP), 2'-0-dimethylaminoethyloxyethyl (2'-0-DMAEOE), and 2'-0-N-methylacetamido (2 -O-NMA).
  • 2'-deoxy, 2'-deoxy-2'-fluoro 2'-0-methyl, 2'-0-methoxyethyl (2'-0-MOE), 2'-0-aminopropyl (2 -O-AP), 2'-0-dimethylaminoethyl (2 -O-DMAOE),
  • RNAi agents Additional modifications and conjugations of RNAi agents have been described. Soutschek et al. 2004 Nature 432: 173-178 presented conjugation of cholesterol to the 3'- end of the sense strand of a siRNA molecule by means of a pyrrolidine linker, thereby generating a covalent and irreversible conjugate. Chemical modifications (including conjugation with other molecules) of RNAi agents may also be made to improve the in vivo pharmacokinetic retention time and efficiency.
  • the RNAi agent to APOC3 comprises at least one 5'-uridine- adenine-3' (5'-ua-3') dinucleotide, wherein the uridine is a 2'-modified nucleotide; at least one 5'-uridine-guanine-3' (5'-ug-3') dinucleotide, wherein the 5'-uridine is a 2'-modified nucleotide; at least one 5'-cytidine-adenine-3' (5'-ca-3') dinucleotide, wherein the 5'-cytidine is a 2'-modified nucleotide; and/or at least one 5'-uridine-uridine-3' (5'-uu-3') dinucleotide, wherein the 5'-uridine is a 2'-modified nucleotide.
  • the RNAi agent can comprise a non-natural nucleobase, wherein the non-natural nucleobase is difluorotolyl, nitroindolyl, nitropyrrolyl, or nitroimidazolyl.
  • the non-natural nucleobase is difluorotolyl.
  • only one of the two oligonucleotide strands contains a non-natural nucleobase.
  • both of the oligonucleotide strands contain a non-natural nucleobase.
  • the RNAi comprises a gap or contains mismatch comprising an abasic nucleotide.
  • the RNAi agent has a single-stranded nick (e.g., a break or missing bond in the backbone).
  • a single-stranded nick can be in either the sense or anti-sense strand, or both.
  • This nick can be, for example, in the sense strand, producing a small internally segmented interfering RNA, or sisiRNA, which may have less off-target effects than the corresponding RNAi agent without a nick. See, for example, WO 2007/107162 to Wengels and Kjems.
  • the antisense nucleic acid or RNAi agent can also have an alternative backbone such as locked nucleic acids (LNA), Morpholinos, peptidic nucleic acids (PNA), threose nucleic acid (TNA), or glycol nucleic acid (GNA), arabinose nucleic acid (ANA), 2 - fluoroarabinose nucleic acid (FANA), cyclohexene nucleic acid (CeNA), anhydrohexitol nucleic acid (HNA), or unlocked nucleic acid (UNA), and/or it can be labeled (e.g., radiolabeled or otherwise tagged).
  • FANA are described in Dowler et al. 2006 Nucl. Acids Res. 34: 1669-1675.
  • One or both strands can comprise an alternative backbone.
  • the RNAi agent employed by the methods of the present disclosure can include an a-anomeric nucleic acid molecule.
  • An a-anomeric nucleic acid molecule forms specific double-stranded hybrids with complementary RNA in which, contrary to the usual ⁇ -units, the strands run parallel to each other. Gaultier et al. 1987 Nucleic Acids. Res. 15: 6625-6641 .
  • the antisense nucleic acid molecule can also comprise a 2'-o-methylribonucleotide (Inoue et al. 1987 Nucleic Acids Res. 15: 6131-6148) or a chimeric RNA-DNA analogue (Inoue et al. 1987 FEBS Lett. 215: 327-330).
  • RNAi agent can be double-stranded DNA, while another portion is double- stranded RNA, forming a DNA-RNA chimera (See, for example, Yamato et al. 201 1. Cancer Gene Ther. 18: 587-597). Mismatches between the guide and passenger stand can also be introduced, though some positions may be better suited than others (See, for example, U.S. Patent App. No. 2009/0209626 to Khvorova).
  • the passenger strand can also be shortened, to as short as 15 or 16 nt, while the guide strand remains 19 nt or longer (See, for example, Sun et al. 2008 Nature Biotech.
  • RNA 14 26: 1379-1382; and Chu and Rana 2008 RNA 14: 1714- 1719).
  • This can increase incorporation of the guide strand into the RNA-induced Silence Complex (RISC), and decrease incorporation of the passenger strand, than reducing off- target effects.
  • the passenger strand may be more amenable to modification (e.g., single-stranded nicking, nucleotide modifications, and shortening) than the guide strand.
  • a “pharmaceutical composition” comprises a pharmaceutically effective amount of one or more APOC3 RNAi agent, a pharmaceutically acceptable carrier, and, optionally, an additional disease treatment which works synergistically with the RNAi agent.
  • pharmaceutically effective amount refers to that amount of a RNAi agent effective to produce the intended pharmacological, therapeutic or preventive result. For example, if a given clinical treatment is considered effective where there is at least a 10% reduction in a measurable parameter associated with a disease or disorder, a therapeutically effective amount of a drug for the treatment of that disease or disorder is the amount necessary to effect at least a 10% reduction in that parameter.
  • a therapeutically effective amount of a RNAi agent targeting APOC3 can reduce APOC3 protein levels by at least 10%.
  • a given clinical treatment is considered effective where there is at least a 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90 or 95% reduction in a measurable parameter associated with a disease or disorder, and the therapeutically effective amount of a drug for the treatment of that disease or disorder is the amount necessary to effect at least a 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90 or 95% reduction, respectively, in that parameter.
  • RNAi agents refers to a carrier for administration of a therapeutic agent.
  • Such carriers include, but are not limited to, lipid nanoparticles, saline, buffered saline, dextrose, water, glycerol, ethanol, and combinations thereof.
  • the term specifically excludes cell culture medium. Any appropriate phamaceutical carrier known in the art can be used in conjunction with the RNAi agents disclosed herein.
  • composition Comprising a RNAi Agent to APOC3
  • RNAi Agent to APOC3
  • Additional components of a pharmaceutical composition comprising a RNAi Agent to APOC3 are contemplated to aid in delivery, stability, efficacy, or reduction of
  • Liposomes have been used previously for drug delivery (e.g., delivery of a chemotherapeutic).
  • Liposomes e.g., cationic liposomes
  • a process of making liposomes is also described in WO04/002453A1.
  • neutral lipids have been incorporated into cationic liposomes (e.g., Farhood et al. 1995), as well as PEGylated lipids.
  • Cationic liposomes have been used to deliver RNAi agent to various cell types (Sioud and Sorensen 2003; U.S. Patent Application 2004/0204377; Duxbury et al., 2004; Donze and Picard, 2002).
  • SNALP refers to a stable nucleic acid-lipid particle.
  • a SNALP represents a vesicle of lipids coating a reduced aqueous interior comprising a nucleic acid such as an RNAi or a plasmid from which an RNAi is transcribed.
  • SNALPs are described, e.g., in U.S. Patent Application Publication Nos. 20060240093, 20070135372, and in International Application No. WO 2009082817.
  • RNAi agent delivery A variety of molecules have been used for cell-specific RNAi agent delivery. See, for example, WO/201 1/076807.
  • the nucleic acid-condensing property of protamine has been combined with specific antibodies to deliver siRNAs. Song et al. 2005 Nat Biotech. 23: 709-717.
  • the self-assembly PEGylated polycation polyethylenimine (PEI) has also been used to condense and protect siRNAs. Schiffelers et al. 2004 Nucl. Acids Res. 32: el49, 141 -1 10.
  • RNAi agents of the present disclosure can be delivered via, for example, Lipid nanoparticles (LNP); neutral liposomes (NL); polymer nanoparticles; double-stranded RNA binding motifs (dsRBMs); or via modification of the RNAi agent (e.g., covalent attachment to the dsRNA) or by any method known in the art for delivery of a RNAi agent comprising nucleic acids.
  • LNP Lipid nanoparticles
  • NL neutral liposomes
  • dsRBMs double-stranded RNA binding motifs
  • modification of the RNAi agent e.g., covalent attachment to the dsRNA
  • Lipid nanoparticles are self-assembling cationic lipid based systems. These can comprise, for example, a neutral lipid (the liposome base); a cationic lipid (for siRNA loading); cholesterol (for stabilizing the liposomes); and PEG-lipid (for stabilizing the formulation, charge shielding and extended circulation in the bloodstream).
  • the cationic lipid can comprise, for example, a headgroup, a linker, a tail and a cholesterol tail.
  • the LNP can have, for example, good delivery to the diseased area, extended circulation in the blood, small particles (e.g., less than 100 nm), and stability in the microenvironment of the diseased area (which may have low pH and/or be hypoxic).
  • Neutral liposomes are non-cationic lipid based particles.
  • Polymer nanoparticles are self-assembling polymer-based particles.
  • Double-stranded RNA binding motifs are self-assembling RNA binding proteins, which will need modifications.
  • RNAi agent compositions in a Lipid nanoparticles comprising a neutral lipid; a cationic lipid; cholesterol; and PEG-lipid
  • Lipid nanoparticles are self-assembling cationic lipid based systems. These can comprise, for example, a neutral lipid (the liposome base); a cationic lipid (for siRNA loading); cholesterol (for stabilizing the liposomes); and PEG-lipid (for stabilizing the formulation, charge shielding and extended circulation in the bloodstream).
  • a neutral lipid the liposome base
  • a cationic lipid for siRNA loading
  • cholesterol for stabilizing the liposomes
  • PEG-lipid for stabilizing the formulation, charge shielding and extended circulation in the bloodstream.
  • a neutral lipid is, for example, the liposome base.
  • a cationic lipid is, for example, for siRNA loading.
  • Cholesterol is, for example, for stabilizing the liposomes.
  • PEG-lipid is, for example, for stabilizing the formulation, charge shielding and extended circulation in the bloodstream.
  • the RNAi agent to APOC3 is packaged as a monotherapy into a delivery vehicle, or may be further ligated to one or more diagnostic compound, reporter group, cross-linking agent, nuclease-resistance conferring moiety, natural or unusual nucleobase, lipophilic molecule, cholesterol, lipid, lectin, steroid, uvaol, hecigenin, diosgenin, terpene, triterpene, sarsasapogenin, Friedelin, epifriedelanol-derivatized lithocholic acid, vitamin, carbohydrate, dextran, pullulan, chitin, chitosan, synthetic carbohydrate, oligo lactate 15-mer, natural polymer, low- or medium-molecular weight polymer, inulin, cyclodextrin, hyaluronic acid, protein, protein-binding agent, integrin-targeting molecule, polycationic, peptide
  • RNAi agents of the present disclosure can be prepared in a pharmaceutical composition comprising various components appropriate for the particular method of administration of the RNAi agent.
  • the present disclosure is a composition comprising one or more APOC3 RNAi agents.
  • the disclosure encompasses a composition comprising any one or more of the following:
  • a RNAi agent comprising a first and a second strand, wherein the sequence of the first strand is the sequence of SEQ ID NO: 1 , or modified or unmodified variants thereof.
  • a RNAi agent comprising a first and a second strand, wherein the sequence of the first strand is the sequence of SEQ ID NO: 2, or modified or unmodified variants thereof.
  • a RNAi agent comprising a first and a second strand, wherein the sequence of the first strand is the sequence of SEQ ID NO: 41 , or modified or unmodified variants thereof.
  • a RNAi agent comprising a first and a second strand, wherein the sequence of the first strand is the sequence of SEQ ID NO: 48, or modified or unmodified variants thereof.
  • a RNAi agent comprising a first and a second strand, wherein the sequence of the first strand is the sequence of SEQ ID NO: 56, or modified or unmodified variants thereof.
  • a RNAi agent comprising a first and a second strand, wherein the sequence of the first strand is the sequence of SEQ ID NO: 71 , or modified or unmodified variants thereof.
  • a RNAi agent comprising a first and a second strand, wherein the sequence of the first strand is the sequence of SEQ ID NO: 102, or modified or unmodified variants thereof.
  • the present disclosure is a composition comprising one or more APOC3 RNAi agents.
  • composition comprising any one or more of the following:
  • a RNAi agent comprising a first and a second strand, wherein the sequence of the first strand is the sequence of SEQ ID NO: 1 , and the sequence of the second strand is the sequence of SEQ ID NO: 45, or modified or unmodified variants thereof.
  • a RNAi agent comprising a first and a second strand, wherein the sequence of the first strand is the sequence of SEQ ID NO: 2, and the sequence of the second strand is the sequence of SEQ ID NO: 100, or modified or unmodified variants thereof.
  • a RNAi agent comprising a first and a second strand, wherein the sequence of the first strand is the sequence of SEQ ID NO: 41 , and the sequence of the second strand is the sequence of SEQ ID NO: 9, or modified or unmodified variants thereof.
  • a RNAi agent comprising a first and a second strand, wherein the sequence of the first strand is the sequence of SEQ ID NO: 48, and the sequence of the second strand is the sequence of SEQ ID NO: 39, or modified or unmodified variants thereof.
  • a RNAi agent comprising a first and a second strand, wherein the sequence of the first strand is the sequence of SEQ ID NO: 56, and the sequence of the second strand is the sequence of SEQ ID NO: 23, or modified or unmodified variants thereof.
  • a RNAi agent comprising a first and a second strand, wherein the sequence of the first strand is the sequence of SEQ ID NO: 102, and the sequence of the second strand is the sequence of SEQ ID NO: 23, or modified or unmodified variants thereof.
  • the present disclosure is a composition comprising one or more APOC3 RNAi agents.
  • composition comprising any one or more of the following:
  • a RNAi agent comprising a first and a second strand, wherein the sequence of the first strand comprises the sequence of SEQ ID NO: 41 , wherein the length of the first and second strand are each no more than about 30 nt, or modified or unmodified variants thereof.
  • a RNAi agent comprising a first and a second strand, wherein the sequence of the first strand comprises the sequence of SEQ ID NO: 71 , wherein the length of the first and second strand are each no more than about 30 nt, or modified or unmodified variants thereof.
  • a RNAi agent comprising a first and a second strand, wherein the sequence of the first strand comprises the sequence of SEQ ID NO: 56, wherein the length of the first and second strand are each no more than about 30 nt, or modified or unmodified variants thereof.
  • a RNAi agent comprising a first and a second strand, wherein the sequence of the first strand comprises the sequence of SEQ ID NO: 102, wherein the length of the first and second strand are each no more than about 30 nt, or modified or unmodified variants thereof.
  • a RNAi agent comprising a first and a second strand, wherein the sequence of the first strand comprises the sequence of SEQ ID NO: 1 , wherein the length of the first and second strand are each no more than about 30 nt, or modified or unmodified variants thereof.
  • a RNAi agent comprising a first and a second strand, wherein the sequence of the first strand comprises the sequence of SEQ ID NO: 2, wherein the length of the first and second strand are each no more than about 30 nt, or modified or unmodified variants thereof.
  • the disclosure comprises a RNAi agent comprising a first and a second strand, wherein the sequence of the first strand comprises at least 18 contiguous nucleotides of the sequence of any one or more RNAi agent disclosed herein, or modified or unmodified variants thereof.
  • composition comprising any one or more of the following:
  • a RNAi agent comprising a first and a second strand, wherein the sequence of the first strand comprises at least 18 contiguous nucleotides of the sequence of SEQ ID NO: 41 , or modified or unmodified variants thereof.
  • a RNAi agent comprising a first and a second strand, wherein the sequence of the first strand comprises at least 18 contiguous nucleotides of the sequence of SEQ ID NO: 48, or modified or unmodified variants thereof.
  • a RNAi agent comprising a first and a second strand, wherein the sequence of the first strand comprises at least 18 contiguous nucleotides of the sequence of SEQ ID NO: 56, or modified or unmodified variants thereof.
  • a RNAi agent comprising a first and a second strand, wherein the sequence of the first strand comprises at least 18 contiguous nucleotides of the sequence of SEQ ID NO: 102, or modified or unmodified variants thereof.
  • a RNAi agent comprising a first and a second strand, wherein the sequence of the first strand comprises at least 18 contiguous nucleotides of the sequence of SEQ ID NO: 1 , or modified or unmodified variants thereof.
  • a RNAi agent comprising a first and a second strand, wherein the sequence of the first strand comprises at least 18 contiguous nucleotides of the sequence of SEQ ID NO: 2, or modified or unmodified variants thereof.
  • the disclosure comprises a RNAi agent comprising a first and a second strand, wherein the sequence of the first strand comprises at least 18 contiguous nucleotides differing by 0, 1 , 2, or 3 nt from a first strand, or modified or unmodified variants thereof.
  • composition comprising any one or more of the following:
  • a RNAi agent comprising a first and a second strand, wherein the sequence of the first strand comprises at least 18 contiguous nucleotides differing by 0, 1 , 2, or 3 nt from the sequence of SEQ ID NO: 41 , or modified or unmodified variants thereof.
  • a RNAi agent comprising a first and a second strand, wherein the sequence of the first strand comprises at least 18 contiguous nucleotides differing by 0, 1 , 2, or 3 nt from the sequence of SEQ ID NO: 48, or modified or unmodified variants thereof.
  • a RNAi agent comprising a first and a second strand, wherein the sequence of the first strand comprises at least 18 contiguous nucleotides differing by 0, 1 , 2, or 3 nt from the sequence of SEQ ID NO: 56, or modified or unmodified variants thereof.
  • a RNAi agent comprising a first and a second strand, wherein the sequence of the first strand comprises at least 18 contiguous nucleotides differing by 0, 1 , 2, or 3 nt from the sequence of SEQ ID NO: 102, or modified or unmodified variants thereof.
  • a RNAi agent comprising a first and a second strand, wherein the sequence of the first strand comprises at least 18 contiguous nucleotides differing by 0, 1 , 2, or 3 nt from the sequence of SEQ ID NO: 1 , or modified or unmodified variants thereof.
  • a RNAi agent comprising a first and a second strand, wherein the sequence of the first strand comprises at least 18 contiguous nucleotides differing by 0, 1 , 2, or 3 nt from the sequence of SEQ ID NO: 2, or modified or unmodified variants thereof.
  • the disclosure comprises a RNAi agent comprising a first and a second strand, wherein the sequence of the first strand comprises at least 14 contiguous nucleotides of the sequence of any one or more RNAi agent disclosed herein, or modified or unmodified variants thereof.
  • composition comprising any one or more of the following:
  • a RNAi agent comprising a first and a second strand, wherein the sequence of the first strand comprises at least 14 contiguous nucleotides of the sequence of SEQ ID NO: 41 , or modified or unmodified variants thereof.
  • a RNAi agent comprising a first and a second strand, wherein the sequence of the first strand comprises at least 14 contiguous nucleotides of the sequence of SEQ ID NO: 48, or modified or unmodified variants thereof.
  • a RNAi agent comprising a first and a second strand, wherein the sequence of the first strand comprises at least 14 contiguous nucleotides of the sequence of SEQ ID NO: 56, or modified or unmodified variants thereof.
  • a RNAi agent comprising a first and a second strand, wherein the sequence of the first strand comprises at least 14 contiguous nucleotides of the sequence of SEQ ID NO: 102, or modified or unmodified variants thereof.
  • a RNAi agent comprising a first and a second strand, wherein the sequence of the first strand comprises at least 14 contiguous nucleotides of the sequence of SEQ ID NO: 1 , or modified or unmodified variants thereof.
  • a RNAi agent comprising a first and a second strand, wherein the sequence of the first strand comprises at least 14 contiguous nucleotides of the sequence of SEQ ID NO: 2, or modified or unmodified variants thereof.
  • the disclosure comprises a RNAi agent comprising a first and a second strand, wherein the sequence of the first strand comprises at least 14 contiguous nucleotides differing by 0, 1 , 2, or 3 nt from a first strand, or modified or unmodified variants thereof.
  • composition comprising any one or more of the following:
  • a RNAi agent comprising a first and a second strand, wherein the sequence of the first strand comprises at least 14 contiguous nucleotides differing by 0, 1 , 2, or 3 nt from the sequence of SEQ ID NO: 41 , or modified or unmodified variants thereof.
  • a RNAi agent comprising a first and a second strand, wherein the sequence of the first strand comprises at least 14 contiguous nucleotides differing by 0, 1 , 2, or 3 nt from the sequence of SEQ ID NO: 48, or modified or unmodified variants thereof.
  • a RNAi agent comprising a first and a second strand, wherein the sequence of the first strand comprises at least 14 contiguous nucleotides differing by 0, 1 , 2, or 3 nt from the sequence of SEQ ID NO: 56, or modified or unmodified variants thereof.
  • a RNAi agent comprising a first and a second strand, wherein the sequence of the first strand comprises at least 14 contiguous nucleotides differing by 0, 1 , 2, or 3 nt from the sequence of SEQ ID NO: 102, or modified or unmodified variants thereof.
  • a RNAi agent comprising a first and a second strand, wherein the sequence of the first strand comprises at least 14 contiguous nucleotides differing by 0, 1 , 2, or 3 nt from the sequence of SEQ ID NO: 1 , or modified or unmodified variants thereof.
  • a RNAi agent comprising a first and a second strand, wherein the sequence of the first strand comprises at least 14 contiguous nucleotides differing by 0, 1 , 2, or 3 nt from the sequence of SEQ ID NO: 2, or modified or unmodified variants thereof.
  • the present disclosure is a composition comprising one or more APOC3 RNAi agents.
  • the disclosure encompasses a composition comprising any one or more of the following: [00559] A RNAi agent comprising a first and a second strand, wherein the sequence of the first strand is the sequence of SEQ ID NO: 41 , and/or the sequence of the second strand is the sequence of SEQ ID NO: 9, or modified or unmodified variants thereof.
  • a RNAi agent comprising a first and a second strand, wherein the sequence of the first strand is the sequence of SEQ ID NO: 48, and/or the sequence of the second strand is the sequence of SEQ ID NO: 39, or modified or unmodified variants thereof.
  • a RNAi agent comprising a first and a second strand, wherein the sequence of the first strand is the sequence of SEQ ID NO: 56, and/or the sequence of the second strand is the sequence of SEQ ID NO: 23, or modified or unmodified variants thereof.
  • a RNAi agent comprising a first and a second strand, wherein the sequence of the first strand is the sequence of SEQ ID NO: 102, and/or the sequence of the second strand is the sequence of SEQ ID NO: 81 , or modified or unmodified variants thereof.
  • a RNAi agent comprising a first and a second strand, wherein the sequence of the first strand is the sequence of SEQ ID NO: 1 , and/or the sequence of the second strand is the sequence of SEQ ID NO: 45, or modified or unmodified variants thereof.
  • a RNAi agent comprising a first and a second strand, wherein the sequence of the first strand is the sequence of SEQ ID NO: 2, and/or the sequence of the second strand is the sequence of SEQ ID NO: 100, or modified or unmodified variants thereof.
  • the present disclosure is a composition comprising one or more APOC3 RNAi agents.
  • composition comprising any one or more of the following:
  • a RNAi agent comprising a first and a second strand, wherein the sequence of the first strand comprises the sequence of SEQ ID NO: 41 , and/or the sequence of the second strand comprises the sequence of SEQ ID NO: 9, wherein the length of the first and second strand are each no more than about 30 nt, or modified or unmodified variants thereof.
  • a RNAi agent comprising a first and a second strand, wherein the sequence of the first strand comprises the sequence of SEQ ID NO: 48, and/or the sequence of the second strand comprises the sequence of SEQ ID NO: 39, wherein the length of the first and second strand are each no more than about 30 nt, or modified or unmodified variants thereof.
  • a RNAi agent comprising a first and a second strand, wherein the sequence of the first strand comprises the sequence of SEQ ID NO: 56, and/or the sequence of the second strand comprises the sequence of SEQ ID NO: 23, wherein the length of the first and second strand are each no more than about 30 nt, or modified or unmodified variants thereof.
  • a RNAi agent comprising a first and a second strand, wherein the sequence of the first strand comprises the sequence of SEQ ID NO: 102, and/or the sequence of the second strand comprises the sequence of SEQ ID NO: 81 , wherein the length of the first and second strand are each no more than about 30 nt, or modified or unmodified variants thereof.
  • a RNAi agent comprising a first and a second strand, wherein the sequence of the first strand comprises the sequence of SEQ ID NO: 1 , and/or the sequence of the second strand comprises the sequence of SEQ ID NO: 45, wherein the length of the first and second strand are each no more than about 30 nt, or modified or unmodified variants thereof.
  • a RNAi agent comprising a first and a second strand, wherein the sequence of the first strand comprises the sequence of SEQ ID NO: 2, and/or the sequence of the second strand comprises the sequence of SEQ ID NO: 100, wherein the length of the first and second strand are each no more than about 30 nt, or modified or unmodified variants thereof.
  • the disclosure comprises a RNAi agent comprising a first and a second strand, wherein the sequence of the first strand comprises at least 14 contiguous nucleotides of a first strand, and/or the sequence of the second strand comprises at least 14 contiguous nucleotides of the second strand of any any one or more RNAi agent disclosed herein.
  • composition comprising any one or more of the following:
  • a RNAi agent comprising a first and a second strand, wherein the sequence of the first strand comprises at least 14 contiguous nucleotides of the sequence of SEQ ID NO: 41 , and/or the sequence of the second strand comprises at least 14 contiguous nucleotides of the sequence of SEQ ID NO: 9, wherein the length of the first and second strand are each no more than about 30 nt, or modified or unmodified variants thereof.
  • a RNAi agent comprising a first and a second strand, wherein the sequence of the first strand comprises at least 14 contiguous nucleotides of the sequence of SEQ ID NO: 48, and/or the sequence of the second strand comprises at least 14 contiguous nucleotides of the sequence of SEQ ID NO: 39, wherein the length of the first and second strand are each no more than about 30 nt, or modified or unmodified variants thereof.
  • a RNAi agent comprising a first and a second strand, wherein the sequence of the first strand comprises at least 14 contiguous nucleotides of the sequence of SEQ ID NO: 56, and/or the sequence of the second strand comprises at least 14 contiguous nucleotides of the sequence of SEQ ID NO: 23, wherein the length of the first and second strand are each no more than about 30 nt, or modified or unmodified variants thereof.
  • a RNAi agent comprising a first and a second strand, wherein the sequence of the first strand comprises at least 14 contiguous nucleotides of the sequence of SEQ ID NO: 102, and/or the sequence of the second strand comprises at least 14 contiguous nucleotides of the sequence of SEQ ID NO: 81 , wherein the length of the first and second strand are each no more than about 30 nt, or modified or unmodified variants thereof.
  • a RNAi agent comprising a first and a second strand, wherein the sequence of the first strand comprises at least 14 contiguous nucleotides of the sequence of SEQ ID NO: 1 , and/or the sequence of the second strand comprises at least 14 contiguous nucleotides of the sequence of SEQ ID NO: 45, wherein the length of the first and second strand are each no more than about 30 nt, or modified or unmodified variants thereof.
  • a RNAi agent comprising a first and a second strand, wherein the sequence of the first strand comprises at least 14 contiguous nucleotides of the sequence of SEQ ID NO: 2, and/or the sequence of the second strand comprises at least 14 contiguous nucleotides of the sequence of SEQ ID NO: 100, wherein the length of the first and second strand are each no more than about 30 nt, or modified or unmodified variants thereof.
  • the disclosure comprises a RNAi agent comprising a first and a second strand, wherein the sequence of the first strand comprises at least 14 contiguous nucleotides differing by 0, 1 , 2, or 3 nt from a first strand, and/or the sequence of the second strand comprises at least 14 contiguous nucleotides differing by 0, 1 , 2, or 3 nt from the second strand of any any one or more RNAi agent disclosed herein.
  • composition comprising any one or more of the following:
  • a RNAi agent comprising a first and a second strand, wherein the sequence of the first strand comprises at least 14 contiguous nucleotides differing by 0, 1 , 2, or 3 nt from the sequence of SEQ ID NO: 41 , and/or the sequence of the second strand comprises at least 14 contiguous nucleotides differing by 0, 1 , 2, or 3 nt from the sequence of SEQ ID NO: 9, wherein the length of the first and second strand are each no more than about 30 nt, or modified or unmodified variants thereof.
  • a RNAi agent comprising a first and a second strand, wherein the sequence of the first strand comprises at least 14 contiguous nucleotides differing by 0, 1 , 2, or 3 nt from the sequence of SEQ ID NO: 48, and/or the sequence of the second strand comprises at least 14 contiguous nucleotides differing by 0, 1 , 2, or 3 nt from the sequence of SEQ ID NO: 39, wherein the length of the first and second strand are each no more than about 30 nt, or modified or unmodified variants thereof.
  • a RNAi agent comprising a first and a second strand, wherein the sequence of the first strand comprises at least 14 contiguous nucleotides differing by 0, 1 , 2, or 3 nt from the sequence of SEQ ID NO: 56, and/or the sequence of the second strand comprises at least 14 contiguous nucleotides differing by 0, 1 , 2, or 3 nt from the sequence of SEQ ID NO: 23, wherein the length of the first and second strand are each no more than about 30 nt, or modified or unmodified variants thereof.
  • a RNAi agent comprising a first and a second strand, wherein the sequence of the first strand comprises at least 14 contiguous nucleotides differing by 0, 1 , 2, or 3 nt from the sequence of SEQ ID NO: 102, and/or the sequence of the second strand comprises at least 14 contiguous nucleotides differing by 0, 1 , 2, or 3 nt from the sequence of SEQ ID NO: 81 , wherein the length of the first and second strand are each no more than about 30 nt, or modified or unmodified variants thereof.
  • a RNAi agent comprising a first and a second strand, wherein the sequence of the first strand comprises at least 14 contiguous nucleotides differing by 0, 1 , 2, or 3 nt from the sequence of SEQ ID NO: 1 , and/or the sequence of the second strand comprises at least 14 contiguous nucleotides differing by 0, 1 , 2, or 3 nt from the sequence of SEQ ID NO: 45, wherein the length of the first and second strand are each no more than about 30 nt, or modified or unmodified variants thereof.
  • a RNAi agent comprising a first and a second strand, wherein the sequence of the first strand comprises at least 14 contiguous nucleotides differing by 0, 1 , 2, or 3 nt from the sequence of SEQ ID NO: 2, and/or the sequence of the second strand comprises at least 14 contiguous nucleotides differing by 0, 1 , 2, or 3 nt from the sequence of SEQ ID NO: 100, wherein the length of the first and second strand are each no more than about 30 nt, or modified or unmodified variants thereof.
  • the disclosure comprises a RNAi agent comprising a sense and an antisense strand, wherein the antisense strand comprises at least 14 contiguous nucleotides differing by 0, 1 , 2, or 3 nt from the antisense strand of any RNAi agent disclosed herein, or modified or unmodified variants thereof, wherein the antisense strand optionally further comprises 0, 1 , 2, 3, 4, 5, 6, 7, 8, 9, or 10 or more nt (or any range thereof, e.g., 0-1 , 1-2, 1 -3, 1-4 nt, etc.).
  • the disclosure comprises a RNAi agent comprising a sense and an antisense strand, wherein the antisense strand comprises at least 14 contiguous nucleotides differing by 0, 1 , 2, or 3 nt from any of: SEQ ID NOs in Table 2, or modified or unmodified variants thereof, wherein the antisense strand optionally further comprises 0, 1 , 2, 3, 4, 5, 6, 7, 8, 9, or 10 or more nt (or any range thereof, e.g., 0-1 , 1 -2, 1 - 3, 1-4 nt, etc.).
  • the disclosure comprises any one or more RNAi agent listed herein. [00596] Additional particular specific aspects
  • compositions comprising 1 , 2, 3, 4, or more of these RNAi agents.
  • Another aspect is a composition comprising any single RNAi agent, along with any other RNAi agents which overlap it.
  • Another aspect comprises two, three, four or more APOC3 RNAi agents which do not overlap and thus target different parts of the RNA molecule. When two or more RNAi agents are used, they can be administered simultaneously or sequentially.
  • RNAi agent comprises a sense strand comprising at least 14 contiguous nucleotides (identical in sequence) to the sense strand of any of the listed RNAi agents, and an antisense strand comprising at least 14 contiguous nucleotides (identical in sequence) to the antisense strand of the same RNAi agent.
  • the composition comprises one, two, three, four, or more such RNAi agents.
  • the composition comprises an RNAi agent which comprises an antisense strand comprising at least 14 contiguous nucleotides differing by 0, 1 , 2 or 3 mismatches from the antisense strand of a RNAi agent described herein.
  • the composition comprises an RNAi agent which comprises an antisense strand comprising at least 14 contiguous nucleotides differing by 0, 1 , 2 or 3 mismatches from the antisense strand of a RNAi agent described herein.
  • the composition comprises an RNAi agent which comprises a sense strand comprising at least 14 contiguous nucleotides differing by 0, 1 , 2, or 3 mismatches from the sense strand of one of the listed RNAi agents, and an antisense strand comprising at least 14 contiguous nucleotides differing by 0, 1 , 2 or 3 mismatches from the antisense strand of the same RNAi agent.
  • a mismatch is defined herein as a difference between the base sequence or length when two sequences are maximally aligned and compared.
  • a mismatch is counted if a difference exists between the base at a particular location in one sequence and the base at the corresponding position in another sequence (e.g., between the sequence of a given RNAi agent and an RNAi agent listed herein).
  • a mismatch is counted, for example, if a position in one sequence has a particular base (e.g., A), and the corresponding position on the other sequence has a different base (e.g., G, C or U).
  • a mismatch is also counted, e.g., if a position in one sequence has a base (e.g., A), and the corresponding position on the other sequence has no base (e.g., that position is an abasic nucleotide which comprises a phosphate-sugar backbone but no base).
  • a single- stranded nick in either sequence (or in the sense or antisense strand) is not counted as mismatch.
  • no mismatch would be counted if one sequence comprises the sequence A-G, but the other sequence comprises the sequence A-G with a single-stranded nick between the A and the G.
  • a base modification is also not considered a mismatch.
  • one sequence comprises a C
  • the other sequence comprises a modified C (e.g., with a 2'-modification) at the same position
  • no mismatch would be counted.
  • modifications of a nucleotide other than replacement or alteration of the base would not constitute a mismatch.
  • no mismatch would occur between a nucleotide which is A, and a nucleotide which is A with a 5' modification and/or a 2'-modification.
  • base replacement is that it would not be able to base-pair with the corresponding base on the opposite strand.
  • terminal overhangs such as "UU” or “dTdT” are not counted when counting the number of mismatches; the terminal "UU” and “dTdT” overhangs are also not included when calculating "15 contiguous nucleotides.”
  • a mismatch is defined as a position wherein the base of one sequence does not match the base of the other sequence.
  • the composition comprises 1 , 2, 3, 4, or more such RNAi agents.
  • the composition comprises an RNAi agent which comprises a sense strand comprising at least 14 contiguous nucleotides differing by 0, 1 , 2 or 3 mismatches from the sense strand of one of the listed RNAi agents, and an antisense strand comprising at least 14 contiguous nucleotides differing by 0, 1 , 2 or 3 mismatches from the antisense strand of the same RNAi agent
  • RNAi agents e.g., comprising different modifications, caps, etc.
  • Tables An unmodified and an example modified variant of various RNAi agents are provided.
  • the disclosure thus encompasses groups of overlapping modified and/or unmodified RNAi agents. More aspects are provided herein, and are included in the scope of each RNAi agents of the disclosure.
  • compositions intended for oral use can be prepared according to any method known to the art for the manufacture of pharmaceutical compositions and such compositions can contain one or more such sweetening agents, flavoring agents, coloring agents or preservative agents in order to provide pharmaceutically elegant and palatable preparations.
  • Tablets contain the active ingredient in admixture with non-toxic pharmaceutically acceptable excipients that are suitable for the manufacture of tablets.
  • excipients can be, for example, inert diluents; such as calcium carbonate, sodium carbonate, lactose, calcium phosphate or sodium phosphate; granulating and disintegrating agents, for example, corn starch, or alginic acid; binding agents, for example starch, gelatin or acacia; and lubricating agents, for example magnesium stearate, stearic acid or talc.
  • inert diluents such as calcium carbonate, sodium carbonate, lactose, calcium phosphate or sodium phosphate
  • granulating and disintegrating agents for example, corn starch, or alginic acid
  • binding agents for example starch, gelatin or acacia
  • lubricating agents for example magnesium stearate, stearic acid or talc.
  • the tablets can be uncoated or they can be coated by known techniques.
  • Formulations for oral use can also be presented as hard gelatin capsules wherein the active ingredient is mixed with an inert solid diluent, for example, calcium carbonate, calcium phosphate or kaolin, or as soft gelatin capsules wherein the active ingredient is mixed with water or an oil medium, for example peanut oil, liquid paraffin or olive oil.
  • Aqueous suspensions contain the active materials in a mixture with excipients suitable for the manufacture of aqueous suspensions.
  • compositions of the invention include all standard techniques for administering substances directly to the stomach or gut, most importantly by patient controlled swallowing of the dosage form, but also by other mechanical and assisted means of such delivery.
  • Dosage levels of the order of from about 0.1 mg to about 140 mg per kilogram of body weight per day are useful in the treatment of the above- indicated conditions (about 0.5 mg to about 7 g per subject per day).
  • the amount of active ingredient that can be combined with the carrier materials to produce a single dosage form varies depending upon the host treated and the particular mode of administration.
  • Dosage unit forms generally contain between from about 1 mg to about 500 mg of an active ingredient. It is understood that the specific dose level for any particular subject depends upon a variety of factors including the activity of the specific compound employed, the age, body weight, general health, sex, diet, time of administration, route of administration, and rate of excretion, drug combination and the severity of the particular disease undergoing therapy.
  • Therapeutic effect of the therapeutic agents of the invention may be enhanced by combination with other agents.
  • other agents will include agents known for use in treating similar diseases, such as angiogenic disorders.
  • RNAi agents of the invention and formulations thereof can be administered orally, topically, parenterally, by inhalation or spray, or rectally in dosage unit formulations containing conventional non-toxic pharmaceutically acceptable carriers, adjuvants and/or vehicles.
  • parenteral as used herein includes percutaneous, subcutaneous, intravascular (e.g., intravenous), intramuscular, intraperitoneal, or intrathecal injection, or infusion techniques and the like. Where two or more different RNAi agents are
  • each may be administered separately or co-administered.
  • the method and/or site of adminstration may be the same or different, e.g., both RNAi agents may be administered intraveneously or subcutaneously, or a first RNAi agent may be administered intraveneously with a second Rai agent
  • the disclosure encompasses a composition or
  • composition comprising a RNAi agent, wherein one or both strands comprises a 3' end cap, the composition further comprising a helper lipid, a neutral lipid, and/or a stealth lipid.
  • the present disclosure relates to a method of treating a target gene-related disease in an individual, comprising the step of
  • the present disclosure relates to a method of inhibiting the expression of target gene in an individual, comprising the step of administering to the individual a therapeutically effective amount of a composition comprising a RNAi agent of the present disclosure.
  • the composition further comprises a
  • the method further comprises the administration of an additional treatment.
  • the additional treatment is a therapeutically effective amount of a composition.
  • the additional treatment is a method (or procedure).
  • the additional treatment and the RNAi agent can be administered in any order, or can be administered simultaneously.
  • the method further comprises the step of administering an additional treatment for the disease.
  • the method further comprises the step of administering an additional treatment or therapy selected from the list of an additional antagonist to a target gene-related disease.
  • the composition comprises a second RNAi agent to target gene.
  • the second RNAi agent is physically separate from the first, or the two are physically connected (e.g., covalently linked or otherwise conjugated).
  • the disclosure pertains to a composition according to any of the embodiments described herein, for use in a method of treating a target gene-related disease in an individual, the method comprising the step of administering to the individual a therapeutically effective amount of a composition according to any of the claims.
  • One embodiment of the disclosure is the use of a composition according to any of these embodiments, in the manufacture of a medicament for treatment of an target gene-related disease.
  • the disclosure pertains to the composition of any of the above embodiments, for use in the treatment of an target gene-related disease.
  • RNAi agents wherein one or both strands comprises a 3' end cap are described in the Examples below, which do not limit the scope of the present disclosure as described in the claims.
  • Example 1 siRNA Lipid Formulations.
  • the siRNAs of the present invention can be encapsulated in lipid nanoparticles as follows.
  • the siRNA lipid nanoparticles were formed by mixing solutions of lipids dissolved in ethanol with siRNA dissolved in a citrate buffer as described generally above.
  • Mixing chambers were used having passages with inner diameters of 0.5, 1 .0, or 2.0 mm.
  • the processing chambers had lengths of from 50 mm to 1000 mm.
  • the dilution chambers had passages with inner diameters equivalent to or of at least to 2 times that of the mixing chamber about 0.5 or 1 .0 or 2.0 or 4.0 mm.
  • the lipid solution contained a cationic lipid, a helper lipid (cholesterol), a neutral lipid (DSPC) and a stealth lipid.
  • cholesterol helper lipid
  • DSPC neutral lipid
  • the concentrations of total lipids were either 16.7 mg/mL or 25 mg/mL.
  • the total lipid to siRNA ratio for these experiments was about 18.3:1.
  • the concentration of siRNA solutions were 0.225, 0.3, 0.3375, or 0.45 mg/mL in a sodium citrate: sodium chloride buffer with pH 5.
  • the concentration of NaCI was 50 mM, 66 mM, 75 mM, or 100 mM.
  • the flow rates and linear velocities were varied as described below.
  • siRNA used for these experiments had sequences and SEQ ID NOs. as shown in Table below.
  • lipid mixture Preparation of lipid mixture in ethanol.
  • siRNA encapsulated at a cationic lipid amine to siRNA phosphate (N:P) molar ratio of 4.5:1 .
  • Lipids cationic lipid, DSPC, cholesterol and lipidated PEG
  • the molar ratios of lipids are 45:9:44:2, respectively.
  • the mixture is sonicated briefly, then gently agitated for 5 minutes and then maintained at 37°C until use.
  • siRNA in citrate buffer Preparation of siRNA in citrate buffer.
  • the buffer for the siRNA streams is 100 mM sodium chloride and 25 mM sodium citrate at a pH of 5.
  • the pH of the citrate buffer is first confirmed to be pH 5.00. If it is not, the pH is adjusted before proceeding.
  • Enough siRNA in water for the encapsulation is thawed from -80°C storage.
  • the siRNA is added to citrate buffer solution and the concentration of the dissolved siRNA is measured by optical density at 260 nm in a UV spectrophotometer.
  • the final concentration of the siRNA is adjusted to 0.45 mg/ml in 150 ml of citrate buffer in a sterile PETG bottle and is held at room
  • the tubing is fluorinated ethylene propylene (FEP) tubing with 1.55 mm inner diameter.
  • Syringes (a), (b1 ) and (b2) are installed on syringe pump A.
  • Tubing leading from the center input of the cross opposite the lipid input is attached to a T junction with a 1 mm inner diameter.
  • Tubing leading from a Luer fitting on syringe (c) containing water alone is attached to the T junction to enable in- line dilution of siRNA lipid nanoparticles, and syringe (c) is installed on syringe pump B.
  • Have the output line from the T junction positioned over a sterile PETG bottle for collection of the diluted siRNA lipid nanoparticles.
  • the syringe pumps are set to the appropriate syringe manufacturer and size and a flow rate of 40 ml per minute. Start both pumps simultaneously, and start collecting material after approximately 0.5 seconds. Approximately 90 ml of encapsulated siRNA lipid nanoparticles will be collected and which contains 25% ethanol by volume, 0.23 mg/mL of the siRNA, 4.2 mg/mL of the lipids, and 50 mM NaCI. After a 60 min. post mixing incubation period, particle sizes are determined using a Malvern Zetasizer.
  • a second 140 mL syringe contains water.
  • the flow rate is set to 25 ml per minute.
  • the siRNA stream and the water stream enter the T junction at 180 degrees from each other.
  • the diluted siRNA lipid nanoparticle suspension is collected into a sterile PETG bottle.
  • the final volume will be approximately 280 ml, with an ethanol concentration of 12.5%.
  • siRNA lipid nanoparticles are filtered by heating approximately 10ml of the suspension in a glass vial which is placed in a aluminum block heater preheated to 50 °C for 10min. The vial is then removed and the solution is removed with a syringe and filtered through a 0.22 ⁇ PES syringe filter directly into a sterile vial. This final product is stored at 4 °C.
  • Percent encapsulation determination SYBR GOLD. To determine the efficiency of the siRNA formulation into lipid nanoparticles, the percent encapsulation of the siRNA can be determined by measuring sybr gold fluorescence. When bound to siRNA, sybr gold fluoresces. The intensity of sybr gold fluorescence is proportional to the amount of siRNA.
  • siRNA stock at approximately 0.9 mg/mL is prepared in PBS.
  • concentration of siRNA stock is verified by UV measurement.
  • the siRNA stock is diluted with PBS to 8 ⁇ g/mL. Serial dilution is done to prepare 4, 2, 1 , 0.5 and 0.25 ⁇ g/mL siRNA solutions. 6 ⁇ g/mL of siRNA is prepared by mixing equal volumes of 8 ⁇ g/mL and 4 ⁇ g/mL solutions.
  • a solution of 0.02% sybr gold in PBS is prepared (e.g., 3 ⁇ _ of sybr gold in 15ml_ of PBS) (solution 2).
  • solution 2 a solution of 0.02% sybr gold in PBS
  • ⁇ _ of solution 1 is mixed with 190 ⁇ _ of solution 2 to provide sample mixture 1.
  • sybr gold will bind only to the nonencapsulated (i.e. free) siRNA. It will not have access to the siRNA encapsulated in the liposome.
  • a solution of 0.02% sybr gold and 0.2% triton-x in PBS is prepared (e.g., 3 ⁇ _ of sybr gold in 15 mL of 0.2% triton in PBS) (solution 3).
  • solution 3 a solution of 0.02% sybr gold and 0.2% triton-x in PBS
  • ⁇ _ of solution 1 is mixed with 190 ⁇ _ of solution 3 to provide sample mixture 2.
  • triton-x disrupts the liposomes and exposes previously encapsulated siRNA to sybr gold binding.
  • sybr gold will bind to the non-encapsulated (i.e. free) siRNA and to all newly exposed siRNA.
  • siRNA+newly exposed siRNA total siRNA [00680]
  • the standard solutions described above (10 ⁇ _ each) are mixed with either 190 ⁇ _ solution 2 to provide standard mixtures 1 or 190 ⁇ _ solution 3 to provide standard mixtures 2.
  • the fluorescence intensity values obtained from standard mixtures 1 are used to create the calibration curve for free siRNA.
  • the fluorescence intensity of a sample 1 mixture is then plugged into the equation provided by the calibration curve for free siRNA.
  • the found concentration of the sample is then multiplied by the dilution magnitude to obtain the free siRNA in the lipid nanoparticle formulation.
  • the fluorescence intensity values obtained from standard mixtures 2 are used to create the calibration curve for total siRNA.
  • the fluorescence intensity of a sample 2 mixture is then plugged into the equation provided by the calibration curve for total siRNA.
  • the found concentration of the sample is then multiplied by the dilution magnitude to obtain the total siRNA in the lipid nanoparticle formulation.
  • the encapsulated siRNA is calculated by the formula:[(total siRNA - free siRNA) / (total siRNA)] x 100%.
  • siRNA stock at approximately 0.9 mg/mL is prepared in PBS.
  • 10 ⁇ _ of TRITON X-100 are added to a 200 ⁇ _ aliquot of stock.
  • the concentration of siRNA stock is verified by UV measurement.
  • Serial dilutions are done to prepare standards at 1/2, 1/4, 1/8, 1/16 and 1/32 concentration of the siRNA stock.
  • 10 ⁇ _ of TRITON X-100 are added to 200 ⁇ _ of each standard. The concentration of each standard is verified by UV measurement.
  • lipid nanoparticle formula In a HPLC vial, 25 ⁇ _ of lipid nanoparticle formula are added to 185 ⁇ _ of 1 X PBS (10X PBS (FISHER, BP399) diluted with deionized water to1 X). The dispersion is gently vortexed until homogeneous (dispersion 1 ). In another HPLC vial, 25 ⁇ _ of lipid nanoparticle formula are added to 185 ⁇ _ of 20% TRITON-X. The dispersion is gently vortexed until clear and homogeneous (dispersion 2).
  • the peak at -17 minutes represents the siRNA. From dispersion 1 , the peak at -10 minutes represents the lipid nanoparticle containing encapsulated siRNA and the peak at -17 minutes represents the nonencapsulated (i.e. free) siRNA.
  • TRITON-X disrupts the lipid nanoparticle enabling previously encapsulated siRNA to elute together with already free siRNA at 17 minutes.
  • the peak at -10 minutes disappears, and only one peak in the chromatogram remains, i.e. the peak at -17 minutes, representing both non-encapsulated (i.e. free) siRNA and newly free siRNA .
  • Free siRNA + newly free siRNA total siRNA.
  • the peak area values obtained from siRNA standards are used to create the siRNA concentration calibration curve.
  • the integrated area of the peak at -17 minutes in dipersion 1 is plugged into the equation provided by calibration curve for free siRNA to obtain the concentration of the free siRNA in the dispersion.
  • the found concentration of the sample is then multiplied by the dilution magnitude to obtain the free siRNA in the lipid nanoparticle formulation.
  • Encapsulated siRNA is calculated by formula: [(total siRNA - free siRNA) / (total siRNA)] x 100%.
  • siRNA lipid nanoparticles are analyzed for size and polydispersity using a Zetasizer Nano ZS from Malvern Instruments.
  • siRNA at an encapsulated siRNA concentration of > 1 mg/ml, dilute 5 ⁇ of sample with 1 15 ⁇ of 1x PBS. Add to a small volume disposable micro-cuvette. Insert the cuvette into the Zetasizer Nano ZS.
  • Process Example 2 Effect of dilution.
  • two aqueous nucleic acids streams having concentrations of 0.45 mg/mL of siRNA, 100 mM NaCI, and 25 mM sodium citrate at a pH of 5 were introduced from opposing directions into a cross-shaped mixing chamber having passages with inner diameters of 0.5 mm with flow rates of 40 mL/min each.
  • a lipid stream was introduced into the cross-shaped mixing chamber from a direction orthogonal to the two nucleic acid streams at a flow rate of 40 mL/min.
  • the lipid stream was made up of 45% cationic lipid A1 , 44% cholesterol, 9% DSPC, and 2% PEG-lipid B1 in ethanol. The total concentration of lipids was 16.7 mg/mL.
  • the joined stream from the cross-shaped mixing chamber was subjected to a dilution step with water using a T-shaped chamber (1.0 mm inner diameter).
  • the water was introduced into the T-shaped chamber at a rate of 40 mL/min.
  • the resultant solution was collected and the encapsulated nucleic acid nanoparticles analyzed for size and uniformity with the results shown in entry 1 in Table 8.
  • the solution obtained from entry 1 included 25% ethanol by volume, 0.23 mg/mL of the siRNA, 4.2 mg/mL of the lipids, and 50 mM NaCl.ln a separate experiment using the same concentrations, flow rates, and initial cross-shaped mixing chamber, the joined streams from the mixing chamber were diluted in a volume of water equivalent to the dilution volume used in entry 1 . These results are shown in entry 2 in Table 8.
  • the concentrations of the collected solution were the same as in entry 1 .
  • Process Example 3 Effect of flow rates, velocities, and solution concentrations.
  • a single nucleic acid stream and a single lipid stream were mixed at various flow rates/velocities and concentrations.
  • the nucleic acid and lipid streams included the same constituents as described above in Process Example 2. No dilution step was used in these examples since the initial siRNA concentrations were adjusted to obtain a final solution concentration the same as entry 1 in Table 8.
  • the concentration of lipids in ethanol in these experiments was either 16.7 mg/mL (1x) or 25 mg/mL (1 .5x).
  • Table 9 The results are shown in Table 9 below with the Z-Avg, #-Avg, and PDI determined 60 minutes post-mixing.
  • Process Example 4 Effect of flow rates and velocities.
  • a single nucleic acid stream and a single lipid stream were mixed from opposing directions at various flow rates/linear velocities and subsequently diluted with water at a 1 mm inner diameter dilution tee.
  • the siRNA, cationic lipid A1 , PEG lipid B1 , cholesterol, and DSPC were used in Process Example 4 in amounts as described above.
  • the total concentration of lipids in the ethanol stream was 16.7 mg/mL.
  • the concentration of siRNA in the nucleic acid stream was 0.9 mg/mL.
  • Table 10 show that increasing linear velocity decreases the particle size and the PDI.
  • Process Example 5 Effect of orientation of T-shaped mixing chamber.
  • Table 1 1 shows the results from two experiments using an alternate mixing chamber (0.5 mm inner diameter). In entry 1 are shown the results obtained where the siRNA enters from the branch and in entry 2 are shown the results obtained where the lipids enter from the branch.
  • the siRNA and lipids were the same as those described above in Process Example 2.
  • the flow rate for the siRNA streams was 120 mL/min and the flow rate of the lipid streams was 40 mL/min.
  • the siRNA concentration was 0.3 mg/mL in a buffer solution containing 66 mM NaCI.
  • the concentration of the lipids was 16.7 mg/mL.
  • the Z- Avg, #-Avg, and PDI were determined 60 minutes post-mixing.
  • Process Example 6 Effect of mixing chamber configurations.
  • a series of experiments were conducted using mixing chambers having various configurations of nucleic acid and lipid streams with the total number of streams ranging from 3 to 6.
  • the mixing chamber had a 1 mm inner diameter chamber and the flow rates were adjusted to maintain the combined flow of 240 mL/min for the siRNA streams and 80 mL/min for the lipid streams.
  • the concentration of siRNA in these experiments was 0.3 mg/mL and the concentration of lipids was 16.7 mg/mL.
  • the linear velocity of the combined siRNA streams was 5.1 meters/second.
  • the linear velocity of the combined lipid streams was 1.7 meters/second.
  • Table 1 1 indicates the angle between lipid or siRNA streams. For example, in the case of three lipid streams and three siRNA streams, each lipid stream is separated from the next lipid stream by either 60 degrees (i.e. ,3 adjacent lipid streams) or 120 degrees (i.e., lipid streams separated by 120 degrees with intervening siRNA streams also separated by 120 degrees).
  • siRNA was used with cationic lipid A4, DSPC, cholesterol, and PEG lipid B1 , in a ratio of 45:9:44:2.
  • the results shown in Table 1 1 indicate that substantially the same results are obtained for the various mixing configurations where the total flow rates/velocities remained constant.

Abstract

The present disclosure relates to compositions and methods for treating APOC3-related diseases such as: hypertriglyceridemia (e.g., Type V Hypertriglyceridemia), abnormal lipid metabolism, abnormal cholesterol metabolism, atherosclerosis, hyperlipidemia, diabetes, including Type 2 diabetes, obesity, cardiovascular disease, and coronary artery disease, among other disorders relating to abnormal metabolism or otherwise, using a therapeutically effective amount of a RNAi agent to APOC3.

Description

ORGANIC COMPOSITIONS TO TREAT APOC3-RELATED DISEASES
FIELD OF THE INVENTION
[001] The present disclosure pertains to RNAi agents to Apolipoprotein III (APOC3).
[002] In various embodiments, the disclosure pertains to an APOC3 RNAi agent comprising a first and a second strand, wherein the sequence of the first and/or second strand is that of any sequence disclosed herein.
[003] In various embodiments, the disclosure pertains to APOC3 RNAi agents which comprise any sequence or an 18-nt portion of any APOC3 RNAi agent sequence disclosed herein (e.g., nt 1 -18 or nt 2-19 of any sequence disclosed herein). In various embodiments, these APOC3 RNAi agents are blunt-ended.
[004] In various embodiments, the disclosure pertains to APOC3 RNAi agents which comprise any sequence of at least 14 contiguous nt of any APOC3 RNAi agent sequence disclosed herein (e.g., nt 1-14, 2-15, 3-16, etc., of any sequence disclosed herein).
[005] In various embodiments, the disclosure relates to compositions comprising an APOC3 RNAi agent having any of various formats. In one embodiment, the APOC3 RNAi agent has the 18-mer format. These APOC3 RNAi agents comprise a sense and an anti- sense strand, each strand being an 18-mer and the strands together forming a blunt-ended duplex, wherein the 3' end of at least one strand terminates in a phosphate or modified internucleoside linker and further comprises, in 5' to 3' order: a spacer; a second phosphate or modified internucleoside linker; and a 3' end cap. In some embodiments, the 3' end of both the sense and anti-sense strand terminate in a phosphate or modified internucleoside linker and further comprise, in 5' to 3' order: a spacer; a second phosphate or modified internucleoside linker; and a 3' end cap. In some embodiments, the disclosure pertains to a RNAi agent that comprises a first and a second strand, wherein the first and second strand are both 18-mers, and the first and second strand together form a blunt-ended duplex, and wherein the 3' end of the first strand terminates in a phosphate or modified internucleoside linker and further comprises, in 5' to 3' order: a spacer, a phosphate or modified
internucleoside linker, and a 3' end cap; and wherein the 3' end of the second strand terminates in a phosphate or modified internucleoside linker and further comprises a 3' end cap. In some embodiments, the disclosure pertains to a RNAi agent that comprises a first and a second strand, wherein the first and second strands are both 18-mers, and the first and second strand together form a blunt-ended duplex, and wherein the 3' end of both the first and second strand terminate in a phosphate or modified internucleoside linker and both further comprise a 3' end cap. In some embodiments the APOC3 RNAi agent has a 19-mer format; in this format, both the first and second strand are 19-mers, wherein the 3' terminus of one or both strands further comprises a 3' terminal dinucleotide, spacer, and/or a 3' end cap. In some embodiments, one or both strands are longer than 19-mers (e.g., a 20-, 21-, 22-, 23-, 24-, 25-, 26-, 27-, 28-, 29-, or 30-mer), wherein one or both ends of the RNAi comprises an overhang, 3' terminal dinucleotide, spacer, and/or 3' end cap. In some embodiments, the APOC3 RNAi agent has an internal spacer format; e.g., one or more subunits [sugar+base] of the first or second strand is/are replaced by a spacer.
[006] Optionally, the 3' end of one or both strands further comprises another spacer and a 3' end cap. In various embodiments, the APOC3 RNAi agent has a shortened sense strand format; e.g., the sense strand is shortened (e.g., to a 18-, 17-, 16-, 15- or 14-mer). The disclosure also pertains to any RNAi agent of any sequence, to any target which has a shortened sense strand format. In this format, the sense strand is shortened (to, for example, a 14-, 15-, 16-, or 17-mer), and the anti-sense strand is an 18-mer or longer. In some embodiments, the first strand is the antisense strand and the second strand is the sense strand. In other embodiments, the first strand is the sense strand and the second strand is the antisense strand. The two strands can have the same or different spacers, phosphates or modified internucleoside linkers, and/or 3' end caps. The strands can be ribonucleotides, or, optionally, one or more nucleotide can be modified or substituted.
Optionally, at least one nucleotide comprises a modified internucleoside linker. Optionally, the RNAi agent can be modified on one or both 5' end. Optionally, the sense strand can comprise a 5' end cap which reduces the amount of the RNA interference mediated by this strand. Optionally, the RNAi agent is attached to a ligand. The disclosure also relates to processes for making such compositions, and methods and uses of such compositions, e.g., to mediate RNA interference.
BACKGROUND OF THE INVENTION
[007] Lipoproteins are globular, micelle-like particles that consist of a non-polar core of acylglycerols and cholesteryl esters surrounded by an amphiphilic coating of protein, phospholipid and cholesterol. Lipoproteins have been classified into five broad categories on the basis of their functional and physical properties: chylomicrons, which transport dietary lipids from intestine to tissues; very low density lipoproteins (VLDL); intermediate density lipoproteins (IDL); low density lipoproteins (LDL); all of which transport triacylglycerols and cholesterol from the liver to tissues; and high density lipoproteins (HDL), which transport endogenous cholesterol from tissues to the liver.
[008] Lipoprotein particles undergo continuous metabolic processing and have variable properties and compositions. Lipoprotein densities increase without decreasing particle diameter because the density of their outer coatings is less than that of the inner core. The protein components of lipoproteins are known as apolipoproteins. At least nine apolipoproteins are distributed in significant amounts among the various human lipoproteins.
[009] Apolipoprotein C-lll is a constituent of HDL and of triglyceride-rich lipoproteins and has a role in hypertriglyceridemia, a risk factor for coronary artery disease. Apolipoprotein C- III slows this clearance of triglyceride-rich lipoproteins by inhibiting lipolysis, both through inhibition of lipoprotein lipase and by interfering with lipoprotein binding to the cell-surface glycosaminoglycan matrix (Shachter, Curr. Opin. Lipidol., 2001 , 12, 297-304).
[0010] The gene encoding human apolipoprotein C-lll (also called APOC3, APOC-III, APO Clll, and APO C-lll) was cloned in 1984 by three research groups (Levy-Wilson et al., DNA, 1984, 3, 359-364; Protter et al., DNA, 1984, 3, 449-456; Sharpe et al., Nucleic Acids Res., 1984, 12, 3917-3932). The coding sequence is interrupted by three introns (Protter et al., DNA, 1984, 3, 449-456). The human apolipoprotein C-lll gene is located approximately 2.6 kB to the 3' direction of the apolipoprotein A-1 gene and these two genes are convergently transcribed (Karathanasis, Proc. Natl. Acad. Sci. U.S.A., 1985, 82, 6374-6378). Also cloned was a variant of human apolipoprotein C-lll with a Thr74 to Ala74 mutation from a patient with unusually high level of serum apolipoprotein C-lll. As the Thr74 is O-glycosylated, the Ala74 mutant therefore resulted in increased levels of serum apolipoprotein C-lll lacking the carbohydrate moiety (Maeda et al., J. Lipid Res., 1987, 28, 1405-1409).
[0011] Five polymorphisms have been identified in the promoter region of the gene: C(-641 ) to A, G(-630) to A, T(-625) to deletion, C(-482) to T and T(-455) to C). All of these polymorphisms are in linkage disequilibrium with the Sstl polymorphism in the 3'
untranslated region. The Sstl site distinguishes the S1 and S2 alleles and the S2 allele has been associated with elevated plasma triglyceride levels (Dammerman et al., Proc. Natl. Acad. Sci. U.S.A., 1993, 90, 4562-4566). The apolipoprotein C-lll promoter is downregulated by insulin and this polymorphic site abolishes the insulin regulation. Thus the potential overexpression of apolipoprotein C-lll resulting from the loss of insulin regulation may be a contributing factor to the development of hypertriglyceridemia associated with the S2 allele (Li et al., J. Clin. Invest., 1995, 96, 2601-2605). The T(-455) to C polymorphism has been associated with an increased risk of coronary artery disease (Olivieri et al., J. Lipid Res., 2002, 43, 1450-1457).
[0012] In addition to insulin, other regulators of apolipoprotein C-lll gene expression have been identified. A response element for the nuclear orphan receptor rev-erb alpha has been located at positions -23/-18 in the apolipoprotein C-lll promoter region and rev-erb alpha decreases apolipoprotein C-lll promoter activity (Raspe et al., J. Lipid Res., 2002, 43, 2172- 2179). The apolipoprotein C-lll promoter region -86 to -74 is recognized by two nuclear factors Clllbl and CIIIB2 (Ogami et al., J. Biol. Chem., 1991 , 266, 9640-9646).
Apolipoprotein C-lll expression is also upregulated by retinoids acting via the retinoid X receptor, and alterations in retinoid X receptor abundance affects apolipoprotein C-lll transcription (Vu-Dac et al., J. Clin. Invest., 1998, 102, 625-632). Specificity protein 1 (Sp1 ) and hepatocyte nuclear factor-4 (HNF-4) have been shown to work synergistically to transactivate the apolipoprotein C-lll promoter via the HNF-4 binding site (Kardassis et al., Biochemistry, 2002, 41 , 1217-1228). HNF-4 also works in conjunction with SMAD3-SMAD4 to transactivate the apolipoprotein C-lll promoter (Kardassis et al., J. Biol. Chem., 2000, 275, 41405-41414).
[0013] Transgenic and knockout mice have further defined the role of apolipoprotein C-lll in lipolysis. Overexpression of apolipoprotein C-lll in transgenic mice leads to
hypertriglyceridemia and impaired clearance of VLDL-triglycerides (de Silva et al., J. Biol. Chem., 1994, 269, 2324-2335; Ito et al., Science, 1990, 249, 790-793). Knockout mice with a total absence of the apolipoprotein C-lll protein exhibited significantly reduced plasma cholesterol and triglyceride levels compared with wild-type mice and were protected from postprandial hypertriglyceridemia (Maeda et al., J. Biol. Chem., 1994, 269, 23610-23616).
[0014] Currently, there are no known therapeutic agents that affect the function of apolipoprotein C-lll. The hypolipidemic effect of the fibrate class of drugs has been postulated to occur via a mechanism where peroxisome proliferator activated receptor (PPAR) mediates the displacement of HNF-4 from the apolipoprotein C-lll promoter, resulting in transcriptional suppression of apolipoprotein C-lll (Hertz et al., J. Biol. Chem., 1995, 270, 13470-13475). The statin class of hypolipidemic drugs also lower triglyceride levels via an unknown mechanism, which results in increases in lipoprotein lipase mRNA and a decrease in plasma levels of apolipoprotein C-lll (Schoonjans et al., FEBS Lett., 1999, 452, 160-164). Consequently, there remains a long felt need for additional agents capable of effectively inhibiting apolipoprotein C-lll function.
BRIEF SUMMARY OF THE INVENTION
[0015] The present disclosure encompasses RNAi agents to APOC3, for inhibition of APOC3, and which are useful in treatment of an APOC3-related disease, such as obesity and metabolic-related disorders such as hyperlipidemia or hypertriglyceridemia (e.g., Type V Hypertriglyceridemia).
[0016] The present disclosure also encompasses a method for inhibiting and/or decreasing the expression and/or activity of APOC3 in a human subject. In various embodiments, the subject has an APOC3-related disease (a pathological state mediated at least in part by APOC3 over-expression or hyper-activity), and the method comprises the step of administering to the subject a therapeutically effective amount of a RNAi agent to APOC3. In various embodiments, the APOC3-related disease is selected from:
hypertriglyceridemia (e.g., Type V Hypertriglyceridemia), abnormal lipid metabolism, abnormal cholesterol metabolism, atherosclerosis, hyperlipidemia, diabetes, including Type 2 diabetes, obesity, cardiovascular disease, and coronary artery disease, among other disorders relating to abnormal metabolism or otherwise.
[0017] In various embodiments, the present disclosure pertains to a RNAi agent to APOC3, comprising two strands, wherein the sequence of the first and/or second strand is, comprises, comprises 18 contiguous nt of, or comprises a sequence of 18 contiguous nt with 0-3 mismatches from, comprises 14 contiguous nt of, or comprises a sequence of 14 contiguous nt with 0-3 mismatches from: the sequence of any RNAi agent disclosed herein. In some embodiments, the disclosure pertains to an APOC3 RNAi agent comprising a first and second strand, wherein the sequence of the first and/or second strand is, comprises, or comprises 14 contiguous nt of any sequence of any APOC3 RNAi agent disclosed herein, except that one or both strands is nicked and/or one or more nt of the sequence disclosed herein has been replaced by a spacer (e.g., the first comprises the sequence of nt 1 -3 and 5- 14, nt 1 -5 and 7-18, or of 1 -4 and 6-18 of a sequence disclosed herein).
[0018] In some embodiments, the RNAi agent to APOC3 comprises a first strand and a second strand, wherein the sequence of the first strand is, comprises, comprises 14 contiguous nt of, or comprises 14 contiguous nt of (with 1-3 mismatches from) any sequence of any APOC3 RNAi agent listed herein.
[0019] The present disclosure provides specific RNAi agents for inhibition of APOC3, and methods that are useful in reducing APOC3 levels in a subject, e.g., a mammal, such as a human. The present disclosure specifically provides double-stranded RNAi agents comprising at least 14, 15, 16, 17, 18, or 19 or more contiguous nucleotides of APOC3. In particular, the present disclosure provides agents comprising sequences of 14 or more contiguous nucleotides differing by 0, 1 , 2 or 3 from those of any of the RNAi agents provided, e.g., in any table herein, or otherwise disclosed herein. The RNAi agents particularly can in one aspect comprise less than 30 nucleotides per strand, e.g., such as 17- 23 nucleotides, 15-19, 18-22, and/or 19-21 nucleotides, and/or such as those provided, and modified and unmodified variants thereof (e.g., wherein the sense and/or anti-sense or first and/or second strand are modified or unmodified). The present disclosure also provides RNAi agents to APOC3 comprising a sense strand and an anti-sense strand, wherein the sense and/or the anti-sense strand comprise sequences of 19 or more contiguous nucleotides differing by 0, 1 , 2 or 3 from those of the RNAi agents provided, and modified or unmodified variants thereof. The present disclosure also provides RNAi agents to APOC3 having a shorted sense strand format. In this format, the sense strand is shortened (to, for example, a 14-, 15-, 16-, or 17-mer), and the anti-sense strand is an 18-mer or longer.
[0020] The disclosure also pertains to any RNAi agent of any sequence, to any target which has a shortened sense strand format. In this format, the sense strand is shortened (to, for example, a 14-, 15-, 16-, or 17-mer), and the anti-sense strand is an 18-mer or longer.
[0021] The sense and anti-sense strand can be contiguous, or physically connected, e.g., by covalently bonds, a loop or linker. In some embodiments, the sense and/or anti- sense strand is discontinuous. Either strand can be nicked. In some embodiments, one or more nucleobase ([sugar+base]) is replaced by a spacer. In some embodiments, the sense strand can be shortened (e.g., to a 14-, 15-, 16-, 17- or 18-mer), for example, in the shortened sense strand format.
[0022] The double-stranded RNAi agents can have 0, 1 or 2 blunt ends, and/or overhangs of 1 , 2, 3 or 4 nucleotides (i.e., 1 to 4 nt) from one or both 3' and/or 5' ends. The double-stranded RNAi agents can also optionally comprise one or two 3' caps and/or one or more modified nucleotides. Modified variants of sequences as provided herein include those that are otherwise identical but contain substitutions of a naturally-occurring nucleotide for a corresponding modified nucleotide.
[0023] Furthermore, the RNAi agent can either contain only nucleotides, e.g., naturally- occurring ribonucleotide subunits, or optionally one or more modifications to the sugar, phosphate or base of one or more of the substitute nucleotide subunits, whether they comprise ribonucleotide subunits or deoxyribonucleotide subunits. In one aspect, modified variants of the disclosed RNAi agents have a thymidine (as RNA, or, preferably, DNA) replacing a uridine, or have an inosine base. In some aspects, the modified variants of the disclosed RNAi agents can have a nick in the passenger strand, mismatches between the guide and passenger strand, DNA replacing the RNA of a portion of both the guide and passenger strand (e.g., the seed region of nt 2-7 counting from the 5' end of the anti-sense strand), and/or a shortened passenger (sense) strand (e.g., 14, 15, 16, 17 or 18 nt). Once a functional guide strand is identified, modifications and variants of the RNAi agent can be readily made. Any two or more modifications which are not mutually exclusive can be combined (e.g., the combination of base modifications with shortened passenger strand; or nicked passenger strand and base modifications; or DNA replacing part or all of the seed region and base modifications in the remaining RNA; etc.).
[0024] In one aspect, modified variants of the disclosed RNAi agents include RNAi agents with the same sequence (e.g., the same sequence of bases) as any RNAi agent disclosed in any of the tables herein or otherwise disclosed herein. It is also noted that various authors have shown that once a successful RNAi agent is designed, generally adding a few nt to one or both strands does not impair RNA activity. Lengthening one or both strands, e.g, to 24 or 25 nt, does not impair RNA interference activity. In addition, we show here that the sense strand can be shortened to a 14-, 15-, 16- or 17-mer (while the anti-sense strand is at least an 18-mer), and RNA interference activity is maintained. Thus, this disclosure encompasses RNAi agent comprising a first strand and a second strand, wherein the sequence of the first and/or second strand comprises the sequence of any APOC3 RNAi agent disclosed herein, further comprising 1 -5 nt.
[0025] In one aspect, modified variants of the disclosed RNAi agents include RNAi agents with the same sequence (e.g., the same sequence of bases) as any RNAi agent disclosed in any of the tables herein or otherwise disclosed herein, but with one or more modifications to one or more of the sugar or phosphate of one or more of the nucleotide subunits. In one aspect, the modifications improve efficacy, stability (e.g., against nucleases in, for example, blood serum or intestinal fluid), and/or reduce immunogenicity of the RNAi agent. One aspect of the present disclosure relates to a double-stranded oligonucleotide comprising at least one non-natural nucleobase. In certain aspects, the non-natural nucleobase is difluorotolyl, nitroindolyl, nitropyrrolyl, or nitroimidazolyl. In a particular aspect, the non-natural nucleobase is difluorotolyl. In certain aspects, only one of the two oligonucleotide strands contains a non-natural nucleobase. In certain aspects, both of the oligonucleotide strands contain a non-natural nucleobase.
[0026] In various embodiments, the disclosure relates to compositions comprising an APOC3 RNAi agent having the 18-mer format. These APOC3 RNAi agents comprise a sense and an anti-sense strand, each strand being an 18-mer and the strands together forming a blunt-ended duplex, wherein the 3' end of at least one strand terminates in a phosphate or modified internucleoside linker and further comprises, in 5' to 3' order: a spacer; a second phosphate or modified internucleoside linker; and a 3' end cap. In some embodiments, the 3' end of both the sense and anti-sense strand terminate in a phosphate or modified internucleoside linker and further comprise, in 5' to 3' order: a spacer; a second phosphate or modified internucleoside linker; and a 3' end cap. In some embodiments, the disclosure pertains to an APOC3 RNAi agent that comprises a first and a second strand, wherein the first and second strand are both 18-mers, and the first and second strand together form a blunt-ended duplex, and wherein the 3' end of the first strand terminates in a phosphate or modified internucleoside linker and further comprises, in 5' to 3' order: a spacer, a phosphate or modified internucleoside linker, and a 3' end cap; and wherein the 3' end of the second strand terminates in a phosphate or modified internucleoside linker and further comprises a 3' end cap. In some embodiments, the disclosure pertains to an APOC3 RNAi agent that comprises a first and a second strand, wherein the first and second strands are both 18-mers, and the first and second strand together form a blunt-ended duplex, and wherein the 3' end of both the first and second strand terminate in a phosphate or modified internucleoside linker and both further comprise a 3' end cap. In some embodiments, the first strand is the antisense strand and the second strand is the sense strand. In other embodiments, the first strand is the sense strand and the second strand is the antisense strand. The two strands can have the same or different spacers, phosphates or modified internucleoside linkers, and/or 3' end caps. The strands can be ribonucleotides, or, optionally, one or more nucleotide can be modified or substituted. Optionally, at least one nucleotide comprises a modified internucleoside linker. Optionally, the RNAi agent can be modified on one or both 5' end. Optionally, the sense strand can comprise a 5' end cap which reduces the amount of the RNA interference mediated by this strand. Optionally, the RNAi agent is attached to a ligand. The disclosure also relates to processes for making such compositions, and methods and uses of such compositions, e.g., to mediate RNA interference. In some embodiments, the disclosure pertains to any RNAi agent of any sequence which has the 18-mer format or any format disclosed herein (e.g., internal spacer format or shortened sense strand format). In some embodiments, the disclosure pertains to an APOC3 RNAi agent comprising a sense strand and an anti-sense strand, wherein each strand is an 18-mer, wherein one or more nucleoside subunit ([sugar+base]) of the 18-mer anti-sense strand is replaced by a spacer, and wherein the 3' terminus of one or both strands further comprises: a 3' end cap, or a spacer and a 3' end cap. In various
embodiments, the nucleobase subunit ([sugar+base]) replaced by a spacer is at position 1 , 3, 5, 6, 7, 15, 16, 17, or 18 (counting 5' to 3'). In various embodiments, the nucleobase subunit ([sugar+base]) replaced by a spacer is at position 5, 6, or 17 (counting 5' to 3'). In various embodiments, the spacer can be sugar, alkyl, cycloakyl, ribitol or other type of abasic nucleotide, 2'-deoxy-ribitol, diribitol, 2'-methoxyethoxy-ribitol (ribitol with 2'-MOE), C3, C4, C5, C6, or 4-methoxybutane-1 ,3-diol (5300). More than one different type of spacer can be incorporated into the same composition.
[0027] In some embodiments, the disclosure pertains to an RNAi agent to APOC3 having a shortened sense strand format. In this format, the sense strand is shortened to a 14-, 15-, 16-, or 17-mer, and the anti-sense strand is a 18-mer or longer. In some embodiments, the sense strand can comprise, in 5' to 3' order, a 14-mer which terminates at the 3' end with a phosphate or modified internucleoside linker, a spacer, a second phosphate or modified internucleoside linker, and a 3' end cap. In some embodiments, the sense strand can comprise, in 5' to 3' order, a 15-mer which terminates at the 3' end with a phosphate or modified internucleoside linker, a spacer, a second phosphate or modified internucleoside linker, and a 3' end cap. In some embodiments, the sense strand can comprise, in 5' to 3' order, a 16-mer which terminates at the 3' end with a phosphate or modified internucleoside linker, a spacer, a second phosphate or modified internucleoside linker, and a 3' end cap. In some embodiments, the sense strand can comprise, in 5' to 3' order, a 17-mer which terminates at the 3' end with a phosphate or modified internucleoside linker, a spacer, a second phosphate or modified internucleoside linker, and a 3' end cap. In various embodiments, the 14-, 15-, 16-, or 17-mer can comprise RNA, wherein one or more RNA subunits (nucleotides) has been modified or substituted. In some embodiments, the last 2 nucleotides on the 3' end of the anti-sense and sense strand are 2'-MOE (a 2'-MOE clamp). In various embodiments, with or without the 2'-MOE clamp, one or more
nucleotides can be modified with a 2'-MOE, 2'-OMe, 2'-F, or substituted with DNA, a peptide nucleic acid (PNA), locked nucleic acid (LNA), morpholino nucleotide, threose nucleic acid (TNA), glycol nucleic acid (GNA), arabinose nucleic acid (ANA), 2'-fluoroarabinose nucleic acid (FANA), cyclohexene nucleic acid (CeNA), anhydrohexitol nucleic acid (HNA), unlocked nucleic acid (UNA). The spacer can be a sugar, alkyl, cycloakyi, ribitol or other type of abasic nucleotide, 2'-deoxy-ribitol, diribitol, 2'-methoxyethoxy-ribitol (ribitol with 2'-MOE), C3, C4, C5, C6, or 4-methoxybutane-1 ,3-diol (5300). The 3' end cap can be selected from any of various 3' end caps described herein or known in the art. In various embodiments, the 3' end cap can be a PAZ ligand (e.g., X058 or X2). The shortened sense strand format is suitable for use with APOC3 RNAi agents, as disclosed herein, or with any RNAi agent of any sequence directed to any gene target.
[0028] The RNAi agent(s) can optionally be attached to a ligand selected to improve one or more characteristic, such as, e.g., stability, distribution and/or cellular uptake of the agent, e.g., cholesterol or a derivative thereof. The RNAi agent(s) can be isolated or be part of a pharmaceutical composition used for the methods described herein. Particularly, the pharmaceutical composition can be formulated for delivery to specific tissues (e.g., those afflicted with an APOC3-related disease) or formulated for parenteral administration. The pharmaceutical composition can optionally comprise two or more RNAi agents, each one directed to the same, overlapping or a different segment of the APOC3 mRNA. Optionally, the pharmaceutical composition can further comprise or be used in conjunction with any known treatment for any APOC3-related disease.
[0029] Second agent or treatment. The method also optionally further comprises the step of administering a second agent or treatment. In some aspects, this second agent is another RNAi agent to APOC3. In various embodiments, the second agent or treatment is iron administration, chelation therapy, phlebotomy, erythropoiesis stimulating agent (ESA) (e.g., Epoetin alfa or darbepoetin alfa), anti-APOC3 antibody, hemodialysis, or hyperbaric oxygen. In other aspects, the second agent or treatment is directed to another target, which is also hyper-active, mutated and/or over-expressed in the pathological state.
[0030] Methods. The present disclosure further provides methods for reducing the level of APOC3 mRNA in a cell, particularly in the case of a disease characterized by over- expression or hyper-activity of APOC3. Cells comprising an alteration such as a mutation, over-expression and/or hyperactivity of APOC3 are termed "APOC3-defective" cells. Such methods comprise the step of administering one or more of the RNAi agents of the present disclosure to an APOC3-defective cell, as further described below. The present methods utilize the cellular mechanisms involved in RNA interference to selectively degrade the target RNA in a cell and are comprised of the step of contacting a cell with one of the RNAi agents of the present disclosure.
[0031] The present disclosure also encompasses a method of treating a human subject having a pathological state mediated at least in part by APOC3 over-expression or hyperactivity, the method comprising the step of administering to the subject a therapeutically effective amount of a RNAi agent APOC3. Additional methods involve preventing, treating, modulating and/or ameliorating a pathological state wherein disease progression requires APOC3, although APOC3 is not amplified or over-expressed. Such methods comprise the step of administering one of the RNAi agents of the present disclosure to a subject, as further described below. Such methods can be performed directly on a cell or can be performed on a mammalian subject by administering to a subject one of the RNAi agents/pharmaceutical compositions of the present disclosure. Reduction of target APOC3 mRNA in a cell results in a reduction in the amount of encoded APOC3 protein produced. In an organism, this can result in restoration of balance in a pathway involving APOC3, and/or prevention of APOC3 accumulation, and/or a reduction in APOC3 activity and/or expression, and/or prevention of APOC3-mediated activation of other genes, and/or amelioration, treatment and/or prevention of an APOC3-related disease. In one aspect, a reduction in APOC3 expression, level or activity can limit disease growth.
[0032] The methods and compositions of the present disclosure, e.g., the methods and APOC3 RNAi agent compositions, can be used in any appropriate dosage and/or formulation described herein or known in the art, as well as with any suitable route of administration described herein or known in the art.
[0033] The details of one or more aspects of the present disclosure are set forth in the accompanying drawings and the description below. Elements of the various aspects (e.g., sequences, modifications, formats [18-mer format, 19-mer format, internal spacer format, shortened sense strand format, etc.], substitutions, spacers, modified internucleoside linkers, endcaps, combinations of RNAi agents, delivery vehicles, combination therapy involving an APOC3 RNAi agent and another agent, etc.) disclosed herein or known in the art which are not mutually exclusive can be combined with each other, provided that the agent or agents are still capable of mediating RNA interference. For example, any RNAi agent sequence disclosed herein can be combined with any set of modifications or endcaps disclosed herein. Similarly, any combination of modifications, 5' end caps, and/or 3' end caps can be used with any RNAi agent sequence disclosed herein. Any RNAi agent disclosed herein (with any combination of modifications or endcaps or without either modifications or endcaps) can be combined with any other RNAi agent or other treatment composition or method disclosed herein. [0034] Other features, objects, and advantages of the present disclosure will be apparent from this description, the drawings, and from the claims.
BRIEF DESCRIPTION OF THE FIGURES
[0035 FIG. 1. Various anti-ApoC3 RNAi agents.
[0036 FIG. 2. Various anti-ApoC3 RNAi agents.
[0037 FIG. 3B. Various anti -ApoC3 RNAi agents.
[0038 FIG. 3B. Various anti -ApoC3 RNAi agents.
[0039 FIG. 4A. Various anti -ApoC3 RNAi agents.
[0040 FIG. 4B. Various anti -ApoC3 RNAi agents.
[0041 FIG. 5A. Various anti -ApoC3 RNAi agents.
[0042 FIG. 5B. Various anti -ApoC3 RNAi agents.
[0043 FIG. 6. Various anti-ApoC3 RNAi agents.
[0044 FIG. 7. Structure representing X1053.
[0045 FIG. 8. Structure representing X058 PAZ ligand.
[0046 FIG. 9. Structure representing X003.
[0047 For each of Figures 1-6, p = phosphate, lower case letter = 2'-0-Methyl nucleotide, Nf = 2'-fluoro nucleotide, dN = deoxynucleotide =, Nm = 2 -MOE nucleotide phosphorothioate, X = abasic ribose, and X1053, X003, X1082, X058, and PAZ = end modifications.
DETAILED DESCRIPTION OF THE INVENTION
[0048] The present disclosure encompasses RNAi agents to APOC3, for targeting and inhibition of APOC3, which are useful in treatment of APOC3-related diseases (e.g., diseases associated with mutations in and/or altered expression, level and/or activity of APOC3, diseases requiring APOC3, diseases affected by a factor whose expression, over- expression, or hyper-activity is directly or indirectly affected by APOC3, and/or diseases treatable by modulating the expression, level and/or activity of APOC3). Such APOC3- related diseases include those listed herein or known in the art. The present disclosure also provides methods of treating a human subject having a pathological state mediated at least in part by APOC3 over-expression or hyper-activity, or requiring APOC3, the method comprising the step of administering to the subject a therapeutically effective amount of a RNAi agent to APOC3.
[0049] Various aspects of the disclosure include the following:
[0050] An RNAi agent comprising an antisense strand of an RNAi agent described herein. [0051] In one aspect, an aspect of the present disclosure relates to a composition comprising an RNAi agent comprising an antisense strand, wherein the antisense strand is, comprises, comprises at least 14 contiguous nucleotides (nt) or, or comprises at least 14 contiguous nucleotides differing by 0, 1 , 2, or 3 nucleotides from the antisense strand of an RNAi agent to APOC3 selected from any sequence provided herein (e.g., in any of the tables herein). In some embodiments, the disclosure pertains to an APOC3 RNAi agent comprising a first and second strand, wherein the sequence of the first and/or second strand is, comprises, or comprises 14 contiguous nt of any sequence of any APOC3 RNAi agent disclosed herein, wherein one or both strands is nicked and/or one or more nt of the sequence disclosed herein has been replaced by a spacer (e.g., the first comprises the sequence of nt 1 -5 and 7-18, or of 1 -4 and 6-18 of a sequence disclosed herein). In another aspect, the present disclosure relates to a composition comprising an RNAi agent comprising a first strand and a second strand, wherein the first strand comprises at least 14 contiguous nucleotides differing by 0, 1 , 2, or 3 nucleotides from the first strand of an RNAi agent to APOC3 from any sequence provided herein. In another aspect, the present disclosure relates to a composition comprising an RNAi agent comprising a sense strand and an antisense strand, wherein the sense strand comprises at least 14 contiguous nucleotides differing by 0, 1 , 2, or 3 nucleotides from the sense strand and the antisense strand comprises at least 14 contiguous nucleotides differing by 0, 1 , 2, or 3 nucleotides from the antisense strand of an RNAi agent to APOC3 listed immediately above. In some embodiments, one or both strands of the RNAi agent is an 18-mer. In one embodiment, both strands are 18-mers and together they form a blunt-ended duplex. In another aspect, the present disclosure relates to a composition comprising an RNAi agent comprising a first strand and a second strand, wherein the sequence of the first strand comprises the sequence of the first strand of any sequence provided herein. In another aspect, the present disclosure relates to a composition comprising an RNAi agent comprising a first strand and a second strand, wherein the sequence of the first strand is the sequence of the first strand of any sequence provided herein. In various aspects, the first and second strands are the anti- sense and sense strand, respectively, of any RNAi agent disclosed herein. In various aspects, the first and second strands are the sense and anti-sense strand, respectively, of any RNAi agent disclosed herein.
[0052] In various embodiments, the disclosure relates to compositions comprising an APOC3 RNAi agent having a novel format (the "18-mer format"). These APOC3 RNAi agents comprise a sense and an anti-sense strand, each strand being an 18-mer and the strands together forming a blunt-ended duplex, wherein the 3' end of at least one strand terminates in a phosphate or modified internudeoside linker and further comprises, in 5' to 3' order: a spacer; a second phosphate or modified internudeoside linker; and a 3' end cap. In some embodiments, the 3' end of both the sense and anti-sense strand terminate in a phosphate or modified internucleoside linker and further comprise, in 5' to 3' order: a spacer; a second phosphate or modified internucleoside linker; and a 3' end cap.
[0053] In some embodiments, the disclosure pertains to an APOC3 RNAi agent that comprises a first and a second strand, wherein the first and second strand are both 18-mers, and the first and second strand together form a blunt-ended duplex, and wherein the 3' end of the first strand terminates in a phosphate or modified internucleoside linker and further comprises, in 5' to 3' order: a spacer, a phosphate or modified internucleoside linker, and a 3' end cap; and wherein the 3' end of the second strand terminates in a phosphate or modified internucleoside linker and further comprises a 3' end cap. In some embodiments, the disclosure pertains to an APOC3 RNAi agent that comprises a first and a second strand, wherein the first and second strands are both 18-mers, and the first and second strand together form a blunt-ended duplex, and wherein the 3' end of both the first and second strand terminate in a phosphate or modified internucleoside linker and both further comprise a 3' end cap.
[0054] In some embodiments the APOC3 RNAi agent has a 19-mer format; in this format, both the first and second strand are 19-mers, wherein the 3' terminus of one or both strands further comprises a 3' terminal dinucleotide, spacer, and/or a 3' end cap. In some embodiments, one or both strands are longer than 19-mers (e.g., a 20-, 21 -, 22-, 23-, 24-, 25-, 26-, 27-, 28-, 29-, or 30-mer), wherein one or both ends of the RNAi comprises an overhang, 3' terminal dinucleotide, spacer, and/or 3' end cap. In some embodiments, the APOC3 RNAi agent has an internal spacer format; e.g., one or more subunits [sugar+base] of the first or second strand is/are replaced by a spacer. Optionally, the 3' end of one or both strands further comprises another spacer and a 3' end cap. In various embodiments, the APOC3 RNAi agent has a shortened sense strand format; e.g., the sense strand is shortened (e.g., to a 18-, 17-, 16-, 15- or 14-mer). In some embodiments, the first strand is the antisense strand and the second strand is the sense strand. In other embodiments, the first strand is the sense strand and the second strand is the antisense strand. The two strands can have the same or different spacers, phosphates or modified internucleoside linkers, and/or 3' end caps. The strands can be ribonucleotides, or, optionally, one or more nucleotide can be modified or substituted. Optionally, at least one nucleotide comprises a modified internucleoside linker. Optionally, the RNAi agent can be modified on one or both 5' end. Optionally, the sense strand can comprise a 5' end cap which reduces the amount of the RNA interference mediated by this strand. Optionally, the RNAi agent is attached to a ligand. The disclosure also relates to processes for making such compositions, and methods and uses of such compositions, e.g., to mediate RNA interference. [0055] In various embodiments, the disclosure relates to compositions comprising an APOC3 RNAi agent conjugated to GalNAc. N-Acetylgalactosamine (also known as GalNAc)(IUPAC name 2-(Acetylamino)-2-deoxy-D-galactose), is an amino sugar derivative of galactose. GalNAc is necessary for intercellular communication, and is concentrated in sensory nerve structures. It is typically the first monosaccharide that connects serine or threonine in particular forms of protein O-glycosylation. In humans it is the terminal carbohydrate forming the antigen of blood group A.
[0056] Particular duplexes include the unmodified (e.g., "generic") and example modified variants listed in the tables herein and otherwise disclosed herein; additional sequences and data for these RNAi agents are presented in the subsequent Tables. In addition to the described example modifications, other modified variants can be made using the nucleotide sequences provided.
[0057] TABLE 1. ANTI-APOC3 siRNA SEQUENCES
Figure imgf000015_0001
boxed sequence = seed region
underline nucleotide = cyno/human mismatch
bold = GU wobble human— > mouse. In vivo data confirms active for mouse
TABLE 2. Knockdown of APOC3 mRNA in HUH7 cells with 30 nM of the described RNAi agents. siRNA SEQUENCE 1 HEAVY-STEM CHEMISTRIES RELATED, INCLUDING RIBITOL WALK AND 1 1-25MERS. The following are used in Table 2 to represent different chemistries and components of the sequences: N=RNA, n (lower case) = 2'-0-Methyl nucleotide, Nm = 2 -MOE nucleotide, X1 = abasic site with ribitol spacer, X2 = abasic site with 1 ,6-hexane-diol spacer.
Figure imgf000016_0001
Figure imgf000017_0001
Figure imgf000018_0001
Figure imgf000019_0001
Figure imgf000020_0001
TABLE 3. Knockdown of APOC3 mRNA in HUH7 cells with 15 nM of the described RNAi agents. PROPERTIES OF VARIOUS MODIFIED VARIANTS OF HUMAN APOC3 RNAI AGENTS DERIVED FROM SEQUENCE HS_APOC3_457. The following are used in Table 3 to represent different chemistries and components of the sequences: N=RNA, p
=phosphate, n (lower case) =2'-0-Methyl nucleotide, Nm = 2 -MOE nucleotide, Nf = 2'-fluoro nucleotide, dN = DNA, X1 = abasic site with ribitol spacer, X2 = abasic site with 1 ,6-hexane- diol spacer, X058 = 3' end cap
Figure imgf000020_0002
Figure imgf000021_0001
uGfCmUmX2 135 a-3'cap 3.9±1.2 UfaAfaAfgGfgAfcAfgUfa UfuCfuCAG AGC AC UG Af gAf a Ufa Cf u Gf u Cf cCf u Uf u Uf a
98
UGCUX2 135
9.1±5.6 UfaAfaAfgGfgAfcAfgUfa UfuCfuCfaGf a Gf cAf cUf g Af gAf a Uf a Cf u Gf u Cf cCf u Uf u Uf
98
uGfcUfX2 135 a
6.3±1 UfaAfaAfgGfgAfcAfgUfa UfuCfuCAG AGC AC UG Af gAf a Ufa Cf u Gf u Cf cCf u Uf u Uf a
98
UGCUX2 135 -3'cap
7.2±2.5 UfaAfaAfgGfgAfcAfgUfa UfuCfuCfAG AGC AC Ug Af gAf a Ufa Cf u Gf u Cf cCf u Uf u Uf a
98
UGCUX2 135 -3'cap
6±1.8 UfaAfaAfgGfgAfcAfgUfa UfuCfuCfaG AGC AC Uf gAf gAf a Ufa Cf u Gf u Cf cCf u Uf u Uf
98
UGCUX2 135 a-3'cap
15.2±1.5 UfaAfaAfgGfgAfcAfgUfa UfuCfuCfaGf AGC Ac Uf gAf gAf a Ufa Cf u Gf u Cf cCf u Uf u Uf a
98
UGCUX2 135 -3'cap
9±2.9 UfaAfaAfgGfgAfcAfgUfa UfuCfuCfaGf AGC Af c Uf g Af g Af a Ufa Cf u Gf u Cf cCf u Uf u Uf
98
uGCUX2 135 a-3'cap
7.5±2.7 UfaAfaAfgGfgAfcAfgUfa UfuCfuCfaGf AGcAfcUfgAfgAfa UfaCfuGfuCfcCfu Ufu Uf
98
uGfCUX2 135 a-3'cap
10.6±2.1 UfaAfaAfgGfgAfcAfgUfa UfuCfuCfaGf AGf c Af c Uf g Af gAf a Uf a Cf u Gf u Cf cCf u Uf u Uf
98
uGfcUX2 135 a-3'cap
7.1±0.4 UfaAfaAfgGfgAfcAfgUfa UfuCfuCfaGf a Gf cAf cUf g Af gAf a Uf a Cf u Gf u Cf cCf u Uf u Uf
98
uGfcUfX2 135 a-3'cap
9±1.9 GGACAGUAU UCUCAGUGCUAAAU CAAU UUAGCACUGAGAAUACUGUCC-
85
d UdGX2 148 3'cap
88.9±16.4 85 GGAcAGuAuucucAGuGcuAAAuuGX2 CAAuuuAGCACuGAGAAuACuGuCC
148
84.9±6.9 85 GGAcAGuAuucucAGuGcuAAAuuGX2 CAAuuuAGCACuGAGAAuACuGuCC-3'cap
148
15.9±6.3 GGACAGUAU UCUCAGUGCUAAAU U CAAU UUAGCACUGAGAAUACUGUCC
85
GX2 148
7.9±4 GGACAGUAU UCUCAGUGCUAAAU U CAAU UUAGCACUGAGAAUACUGUCC-
85
GX2 148 3'cap
14.9±3.7 GfgAfcAfgUfa UfuCfuCfaGfuGfcUfaAf cAf a Uf u Uf aGf c Af c Uf gAf gAf a Ufa Cf u Gf u C
86
a UfUmGmAXlX2 149 mCmAXlX2
12.6±3.1 GfgAfcAfgUfa UfuCfuCfaGfuGfcUfaAf cAf a Uf u Uf aGf c Af c Uf gAf gAf a Ufa Cf u Gf u C
86
a UfUmGmAXlX2 149 mCmAXl-3'cap
17.1±3.9 GfgAfcAfgUfa UfuCfuCfaGfuGfcUfaA cAfa Ufu UAGfcAfcUfgAfgAfa UfaCfuGfuCf
85
AUfuGfX2 148 c-3'cap
11.8±2.1 GfgAfcAfgUfa UfuCfuCfaGfuGfcUfaAf cAfa UUUAGfcAfcUfgAfgAfaUfaCfuGfuCfc
85
a UfuGfX2 148 -3'cap
10.5±0.7 Gf gAf cAf gUf a Uf u Cf u Cf a Gf u Gf cU AA cAfa UUUAGfcAfcUfgAfgAfaUfaCfuGfuCfc
85
AUfuGfX2 148 -3'cap
6.2±0.7 GfgAfcAfgUfa UfuCfuCfaGfuGfcUfaA CAAU U Ua Gf cAf c Uf g Af g Af a Ufa Cf u Gf u Cf c-
85
AU UGX2 148 3'cap
16±2.9 GfgAfcAfgUfa UfuCfuCfaGfuGfcUfaAf cAf a Uf u Uf aGf c Af c Uf gAf gAf a Ufa Cf u Gf u Cf
85
a UfUmGmX2 148 c-3'cap
128.6±24.3 Gf gAf cAf gUf a Uf u Cf u Cf a Gf u Gf cU AA CAAU UUAGfcAfcUfgAfgAfa UfaCfuGfuCfc
85
AU UGX2 148
35.8±12.6 GfgAfcAfgUfa UfuCfuCfaGfuGfcUfaAf cAf a Uf u Uf aGf c Af c Uf gAf gAf a Ufa Cf u Gf u Cf
85
a UfuGfX2 148 c
6±2.4 Gf gAf cAf gUf a Uf u Cf u Cf a Gf u Gf cU AA CAAU UUAGfcAfcUfgAfgAfa UfaCfuGfuCfc
85
AU UGX2 148 -3'cap
Figure imgf000023_0001
X2 132 3'cap
Figure imgf000024_0001
X1X2 132 3'cap
[0058] One or both strands of the 18-mer format can comprise, in various embodiments, in 5' to 3' order, a 18-mer, a spacer, and a 3' end cap. The 18-mer can be modified or unmodified. In various embodiments, the modifications include the two nt on the 3' end being 2'-MOE (a "2-MOE clamp"). In various embodiments, with or without the 2'-MOE clamp, various nucleotides can be modified with a 2'-MOE, 2'-OMe, 2'-F, or substituted with DNA, a peptide nucleic acid (PNA), locked nucleic acid (LNA), morpholino nucleotide, threose nucleic acid (TNA), glycol nucleic acid (GNA), arabinose nucleic acid (ANA), 2 - fluoroarabinose nucleic acid (FANA), cyclohexene nucleic acid (CeNA), anhydrohexitol nucleic acid (HNA), unlocked nucleic acid (UNA). The spacer can be a sugar, alkyl, cycloakyl, ribitol or other type of abasic nucleotide, 2'-deoxy-ribitol, diribitol, 2'- methoxyethoxy-ribitol (ribitol with 2 -MOE), C3, C4, C5, C6, or 4-methoxybutane-1 ,3-diol (5300). The 3' end cap can be selected from any of various 3' end caps described herein or known in the art.
[0059] Various non-limiting examples of modified forms of sequences are shown in Figs. 1-5.
[0060] In one embodiment, at least one nucleotide of the RNAi agent is modified.
[0061] In one embodiment, said at least one modified nucleotide is selected from among 2' alkoxyribonucleotide, 2' alkoxyalkoxy ribonucleotide, or 2'-fluoro ribonucleotide. In another embodiment, said at least one modified nucleotide is selected from 2'-OMe, 2'-MOE and 2'-H. In various aspects, the nucleotide subunit is chemically modified at the 2' position of the sugar. In one aspect, the 2' chemical modification is selected from a halo, a C1 -10 alkyl, a C1-10 alkoxy, a halo, and the like. In specific aspects, the 2' chemical modification is a C1 -10 alkoxy selected from -OCH3 (i.e., "OMe"), -OCH2CH3 (i.e., "OEt") or -CH2OCH2CH3 (i.e., methoxyethyl or "MOE"); or is a halo selected from F.
[0062] In various embodiments, one of more nucleotides is DNA or is replaced by a peptide nucleic acid (PNA), locked nucleic acid (LNA), morpholino nucleotide, threose nucleic acid (TNA), glycol nucleic acid (GNA), arabinose nucleic acid (ANA), 2 - fluoroarabinose nucleic acid (FANA), cyclohexene nucleic acid (CeNA), anhydrohexitol nucleic acid (HNA), and/or unlocked nucleic acid (UNA); and/or at least one nucleotide comprises a modified internucleoside linker (e.g., wherein at least one phosphate of a nucleotide is replaced by a modified internucleoside linker), wherein the modified internucleoside linker is selected from phosphorothioate, phosphorodithioate,
phosphoramidate, boranophosphonoate, an amide linker, and a compound of formula (I) (as described elsewhere herein).
[0063] In one embodiment, the first two base-pairing nucleotides on the 3' end of the first and/or second strand are modified.
[0064] In one embodiment, the first two base-pairing nucleotides on the 3' end of the first and/or second strand are 2'-MOE.
[0065] In various sequences disclosed herein, lower-case letters (e.g., c, u) indicate modified nucleotides while upper case letters (e.g., C, U, A, G) indicate unmodified nucleotides. In this Table, example modified versions of each of the sequences are shown. However, the present disclosure also contemplates and encompasses unmodified versions of these sequences and other versions which comprise additional or alternative
modifications.
[0066] In various sequences disclosed herein, the modified and unmodified variants can optionally further comprise the sequence "TT", "dTdT", "dTsdT" or "UU" as a single-stranded overhang at the 3' end, also termed herein a terminal dinudeotide or 3' terminal dinudeotide. dT is 2'-deoxy-thymidine-5'-phosphate and sdT is 2'-deoxy Thymidine 5'-phosphorothioate. In the disclosed sequences, terminal dinudeotide "UU" is UU or 2'-OMe-U 2'-OMe-U, and the terminal TT and the terminal UU can be in the inverted/reverse orientation. The terminal dithymidine (e.g., UU) is not part of the APOC3 target sequence, but is a modified variant of the dithymidine dinudeotide commonly placed as an overhang to protect the ends of siRNAs from nucleases (see, for example, Elbashir et al. 2001 Nature 41 1 : 494-498; Elbashir et al. 2001 EMBO J. 20: 6877-6888; and Kraynack et al. 2006 RNA 12:163-176). A terminal dinudeotide is known from these references to enhance nuclease resistance but not contribute to target recognition. Thus, the present disclosure also encompasses any modified or any unmodified variant disclosed herein, wherein the modified variant comprises a terminal TT, dTdT, sdT, dTsdT, sdTsdT, sdTdT, or the like which may be in either the inverted/reverse orientation or in the same 5' to 3' orientation as the APOC3 specific sequence in the duplex. In addition, terminology used herein referring to "the APOC3 portion of a RNAi agent sequence" and the like indicate the portion of the sequence of a RNAi agent which is derived from APOC3 (thus "the APOC3 portion of a RNAi agent sequence" does not include, for example, a terminal dTdT, TT, UU, U (2'-OMe) dT, U (2 - OMe) U (2'-OMe), T(2'-OMe) T (2'-OMe), T(2'-OMe) dT, or the like, but does include the portion of the RNAi agent that corresponds to or is complementary to a portion of the
APOC3 gene sequence or mRNA sequence. In one aspect, the composition comprises a RNAi agent comprising a first and a second strand, wherein the sequence of the first strand and the sequence of the second strand are the sequences of the first and second strand, respectively, of any RNAi agent provided herein, further comprising a 3' terminal dinucleotide (a single-stranded overhang comprising 2 nt at the 3' end). In one aspect, the composition comprises a RNAi agent comprising a first and a second strand, wherein the sequence of the first strand and the sequence of the second strand are the sequences of the first and second strand, respectively, of any RNAi agent provided herein, further comprising a 3' terminal dinucleotide selected from TT, UU, U (2'-OMe) dT, U (2'-OMe) U (2'-OMe), T(2'- OMe) T (2'-OMe), T(2'-OMe) dT, dTdT, sdT, dTsdT, sdTsdT, and sdTdT. In one aspect, the composition comprises a RNAi agent comprising a first and a second strand, wherein the sequence of the first strand and the sequence of the second strand are the sequences of the first and second strand, respectively, of any RNAi agent provided herein, further comprising a 3' terminal UU dinucleotide.
[0067] On any modified or unmodified variant, a 3' end cap, as is known in the art, can be used instead of or in addition to a terminal dinucleotide to stabilize the end from nuclease degradation provided that the 3' end cap is able to both stablize the RNAi agent (e.g., against nucleases) and not interfere excessively with siRNA activity. Thus, the present disclosure also encompasses any modified or any unmodified variant disclosed herein, wherein the modified variant further comprises a terminal 3' end cap.
[0068] An RNAi agent comprising an antisense strand of an RNAi agent described herein.
[0069] In one particular specific aspect, the present disclosure relates to a composition comprising an RNAi agent comprising an antisense strand, wherein the antisense strand comprises at least 14 contiguous nucleotides differing by 0, 1 , 2, or 3 nucleotides from the antisense strand of an RNAi agent to APOC3 selected from those antisense strands in the specific duplexes provided herein.
[0070] Various particular specific aspects of this aspect are described below.
[0071] In one aspect, the composition further comprises a second RNAi agent to APOC3.
In various aspects, the second RNAi agent is physically separate from the first, or the two are physically connected (e.g., covalently linked or otherwise conjugated).
[0072] In one aspect, the composition comprises a RNAi agent to APOC3 comprising a first and a second strand, wherein the sequence of the first strand and the sequence of the second strand are the sequences of the first and second strand, respectively, of any RNAi agent provided herein. In one aspect, the composition comprises a RNAi agent comprising a first and a second strand, wherein the sequence of the first strand and the sequence of the second strand are the sequences of the first and second strand, respectively, of any RNAi agent provided herein, further comprising an additional about 6 to 20 nucleotides on one or both strands (e.g., about 6, 7, 8, 9, 10, 1 1 , 12, 13, 14, 15, 16, 17, 18, 19 or 20 nt). In one aspect, the composition comprises a RNAi agent comprising a first and a second strand, wherein the sequence of the first strand and the sequence of the second strand are the sequences of the first and second strand, respectively, of any RNAi agent provided herein, further comprising a 3' terminal dinucleotide.
[0073] In one aspect, the composition comprises a RNAi agent comprising a first and a second strand, wherein the sequence of the first strand and the sequence of the second strand are the sequences of the first and second strand, respectively, of any RNAi agent provided herein, further comprising a 3' terminal dinucleotide selected from TT, UU, U (2 - OMe) dT, U (2'-OMe) U (2'-OMe), T(2'-OMe) T (2'-OMe), T(2'-OMe) dT, dTdT, sdT, dTsdT, sdTsdT, and sdTdT. In one aspect, the composition comprises a RNAi agent comprising a first and a second strand, wherein the sequence of the first strand and the sequence of the second strand are the sequences of the first and second strand, respectively, of any RNAi agent provided herein, further comprising a 3' terminal UU dinucleotide. In various aspects, the first and second strands are the sense and anti-sense strands listed herein, respectively. In various aspects, the first and second strands are the anti-sense and sense strands listed herein, respectively.
[0074] In one aspect, the sense strand is about 30 or fewer nucleotides (nt) in length.
[0075] In one aspect, the antisense strand is about 30 or fewer nucleotides in length.
[0076] In one aspect, the antisense strand forms a duplex region with a sense strand, wherein the duplex region is about 15 to 30 nucleotide pairs in length.
[0077] In one aspect, the antisense strand is about 15 to about 30 nucleotides in length, including 18, about 18, and about 19 to about 23 nucleotides in length. In one aspect, the antisense strand has at least the length selected from about 15 nucleotides, about 16 nucleotides, about 17 nucleotides, 18 nucleotides, about 18 nucleotides, about 19 nucleotides, about 20 nucleotides, about 21 nucleotides, about 22 nucleotides, about 23 nucleotides, about 24 nucleotides, about 25 nucleotides, about 26 nucleotides, about 27 nucleotides, about 28 nucleotides, about 29 nucleotides and 30 nucleotides. In various aspects, the anti-sense strand is a 17-, 18-, 19-, 20-, 21 -, 22-, 23-, 24-, 25-, 26-, 27-, 28-, 29- or 30-mer.
[0078] In various aspects, the sense strand can be shorter than the anti-sense strand. In various aspects, the sense strand is a 14-, 15-, 16-, 17-, 18-, 19-, 20-, 21 -, 22-, 23-, 24-, 25-, 26-, 27-, 28-, 29- or 30-mer.
[0079] In one aspect, the RNAi agent comprises a modification that causes the RNAi agent to have increased stability in a biological sample or environment (e.g., cytoplasm, interstitial fluid, blood serum, or lung or intestinal lavage). [0080] In one aspect, the RNAi agent comprises at least one sugar backbone modification (e.g., phosphorothioate linkage) or at least one 2'-modified nucleotide.
[0081] In one aspect, the RNAi agent comprises: at least one 5'-uridine-adenine-3' (5'-ua- 3') dinucleotide, wherein the uridine is a 2'-modified nucleotide; at least one 5'-uridine- guanine-3' (5'-ug-3') dinucleotide, wherein the 5'-uridine is a 2'-modified nucleotide; at least one 5'-cytidine-adenine-3' (5'-ca-3') dinucleotide, wherein the 5'-cytidine is a 2'-modified nucleotide; or at least one 5'-uridine-uridine-3' (5'-uu-3') dinucleotide, wherein the 5'-uridine is a 2'-modified nucleotide. These dinucleotide motifs are particularly prone to serum nuclease degradation (e.g. RNase A). Chemical modification at the 2'-position of the first pyrimidine nucleotide in the motif prevents or slows down such cleavage. This modification recipe is also known under the term 'endo light'.
[0082] In one aspect, the RNAi agent comprises a 2'-modification selected from the group consisting of: 2'-deoxy, 2'-deoxy-2'-fluoro, 2'-0-methyl (2'-OMe or 2'OMe), 2 -0- methoxyethyl (2 -O-MOE), 2'-0-aminopropyl (2 -O-AP), 2'-0-dimethylaminoethyl (2 -0-
DMAOE), 2'-0-dimethylaminopropyl (2'-0-DMAP), 2'-0-dimethylaminoethyloxyethyl (2'-0-
DMAEOE), and 2'-0-N-methylacetamido (2'-0-NMA). In one aspect, all pyrimidines (uridine and cytidine) are 2' O-methyl-modified nucleosides.
[0083] In one aspect, the RNAi agent comprises at least one blunt end.
[0084] In one aspect, the RNAi agent comprises an overhang having 1 nt to 4 nt unpaired.
[0085] In one aspect, the RNAi agent comprises an overhang at the 3'-end of the antisense strand of the RNAi agent.
[0086] In one aspect, the RNAi agent is ligated to one or more diagnostic compound, reporter group, cross-linking agent, nuclease-resistance conferring moiety, natural or unusual nucleobase, lipophilic molecule, cholesterol, lipid, lectin, steroid, uvaol, hecigenin, diosgenin, terpene, triterpene, sarsasapogenin, Friedelin, epifriedelanol-derivatized lithocholic acid, vitamin, carbohydrate, dextran, pullulan, chitin, chitosan, synthetic carbohydrate, oligo lactate 15-mer, natural polymer, low- or medium-molecular weight polymer, inulin, cyclodextrin, hyaluronic acid, protein, protein-binding agent, integrin- targeting molecule, polycationic, peptide, polyamine, peptide mimic, and/or transferrin.
[0087] In one aspect, the RNAi agent is capable of inhibiting expression of APOC3 by at least about 50% in Huh-7 cells in nude mice.
[0088] In one aspect, the RNAi agent is capable of inhibiting expression of APOC3 by at least about 70% at a concentration of 10 nM in Huh-7 cells in vitro.
[0089] In one aspect, the RNAi agent is capable of inhibiting expression of APOC3 by at least about 75% at a concentration of 10 nM in Huh-7 cells in vitro. [0090] In one aspect, the RNAi agent is capable of inhibiting expression of APOC3 by at least about 80% at a concentration of 10 nM in Huh-7 cells in vitro.
[0091] In one aspect, the RNAi agent is capable of inhibiting expression of APOC3 by at least about 90% at a concentration of 10 nM in Huh-7 cells in vitro.
[0092] In one aspect, the RNAi agent is capable of inhibiting expression of APOC3 by at least about 95% at a concentration of 10 nM in Huh-7 cells in vitro.
[0093] In one aspect, the RNAi agent is capable of inhibiting expression of APOC3 by at least about 99% at a concentration of 10 nM in Huh-7 cells in vitro.
[0094] In one aspect, the RNAi has an EC50 of no more than about 0.1 nM in Huh-7 cells in vitro. EC50 is effective concentration to reduce gene expression by 50%.
[0095] In one aspect, the RNAi has an EC50 of no more than about 0.01 nM in Huh-7 cells in vitro.
[0096] In one aspect, the RNAi has an EC50 of no more than about 0.001 nM in Huh-7 cells in vitro.
[0097] An RNAi agent comprising a sense and antisense strand of an RNAi described herein.
[0098] In one particular specific aspect, the present disclosure relates to a composition comprising a RNAi agent comprising a first strand and a second strand, wherein the sequence of the first strand and/or second strand comprise at least 14 contiguous nucleotides, differing by 0, 1 , 2, or 3 nucleotides from the sequence of the first and/or second strand of a RNAi agent to APOC3 selected from the specific duplexes provided herein.
[0099] In one particular specific aspect, the present disclosure relates to a composition comprising a RNAi agent comprising a first strand and a second strand, wherein the sequence of the first strand and/or second strand comprise the sequence of the first and/or second strand, respectively, of a RNAi agent to APOC3 selected from the specific duplexes provided herein.
[00100] In one particular specific aspect, the present disclosure relates to a composition comprising a RNAi agent comprising a first strand and a second strand, wherein the sequence of the first strand and/or second strand are the sequence of the first and/or second strand, respectively, of a RNAi agent to APOC3 selected from the specific duplexes provided herein.
[00101] In one particular specific aspect, the present disclosure relates to a composition comprising a RNAi agent comprising a first strand and a second strand, wherein the sequence of the first strand and/or second strand are the sequence of the first and/or second strand, respectively, of a RNAi agent to APOC3 selected from the specific duplexes provided herein, wherein the sequence of the first and/or second strand further comprise a terminal dinucleotide.
[00102] In one particular specific aspect, the present disclosure relates to a composition comprising a RNAi agent comprising a first strand and a second strand, wherein the sequence of the first strand and/or second strand are the sequence of the first and/or second strand, respectively, of a RNAi agent to APOC3 selected from the specific duplexes provided herein, wherein the sequence of the first and/or second strand further comprise a terminal UU dinucleotide.
[00103] In one particular specific aspect, the present disclosure relates to a composition comprising an RNAi agent comprising a sense strand and an antisense strand, wherein the sense strand and antisense strand comprise at least 14 contiguous nucleotides differing by 0, 1 , 2, or 3 nucleotides, from the sense and antisense strand, respectively, of an RNAi agent to APOC3 selected from the specific duplexes provided herein.
[00104] Various particular specific aspects of this aspect are described below.
[00105] In one aspect, the composition comprises a second RNAi agent to APOC3. In various aspects, the second RNAi agent is physically separate from the first, or the two are physically connected (e.g., chemically linked or otherwise conjugated). In some aspects, the first and second RNAi agents are combined within the same composition (e.g., both in the same lipid nanoparticle).
[00106] In one aspect, the antisense strand is about 30 or fewer nucleotides in length.
[00107] In one aspect, the sense strand and the antisense strand form a duplex region about 15 to about 30 nucleotide pairs in length.
[00108] In one aspect, the antisense strand is about 15 to about 36 nt in length including about 18 to about 23 nt in length, and including about 19 to about 21 nt in length and about 19 to about 23 nt in length. In one aspect, the antisense strand has at least the length selected from about 15 nt, about 16 nt, about 17 nt, 18, about 18 nt, about 19 nt, about 20 nt, about 21 nt, about 22 nt, about 23 nt, about 24 nt, about 25 nt, about 26 nt, about 27 nt, about 28 nt, about 29 nt and about 30 nt. In various aspects, the anti-sense strand is a 17-, 18-, 19-, 20-, 21-, 22-, 23-, 24-, 25-, 26-, 27-, 28-, 29- or 30-mer.
[00109] In various aspects, the sense strand can be shorter than the anti-sense strand. In various aspects, the sense strand is a 14-, 15-, 16-, 17-, 18-, 19-, 20-, 21 -, 22-, 23-, 24-, 25-, 26-, 27-, 28-, 29- or 30-mer.
[00110] In one aspect, the RNAi agent comprises a modification that causes the RNAi agent to have increased stability in a biological sample or environment.
[00111] In one aspect, the RNAi agent comprises a modified sugar backbone such as, e.g., a phosphorothioate linkage, or comprises a 2'-modified nucleotide. [00112] In one aspect, the RNAi agent comprises: at least one 5'-uridine-adenine-3' (5'-ua-3') dinucleotide, wherein the uridine is a 2'-modified nucleotide; at least one 5'-uridine- guanine-3' (5'-ug-3') dinucleotide, wherein the 5'-uridine is a 2'-modified nucleotide; at least one 5'-cytidine-adenine-3' (5'-ca-3') dinucleotide, wherein the 5'-cytidine is a 2'-modified nucleotide; or at least one 5'-uridine-uridine-3' (5'-uu-3') dinucleotide, wherein the 5'-uridine is a 2'-modified nucleotide.
[00113] In one aspect, the RNAi agent comprises a 2'-modification selected from the group consisting of: 2'-deoxy, 2'-deoxy-2'-fluoro, 2'-0-methyl, 2'-0-methoxyethyl (2 -O-MOE), 2'-0-aminopropyl (2 -O-AP), 2'-0-dimethylaminoethyl (2 -O-DMAOE), 2 -0- dimethylaminopropyl (2 -O-DMAP), 2'-0-dimethylaminoethyloxyethyl (2'-0-DMAEOE), and 2'-0-N-methylacetamido (2'-0-NMA). In one aspect, all pyrimidines (uridine and cytidine) are 2' O-methyl-modified nucleosides.
[00114] In one aspect, the RNAi agent comprises at least one blunt end.
[00115] In one aspect, the RNAi agent comprises an overhang having 1 to 4 nt unpaired.
[00116] In one aspect, the RNAi agent comprises an overhang at the 3'-end of the antisense strand of the RNAi agent.
[00117] In one aspect, the RNAi agent is ligated to one or more diagnostic compound, reporter group, cross-linking agent, nuclease-resistance conferring moiety, natural or unusual nucleobase, lipophilic molecule, cholesterol, lipid, lectin, steroid, uvaol, hecigenin, diosgenin, terpene, triterpene, sarsasapogenin, Friedelin, epifriedelanol-derivatized lithocholic acid, vitamin, carbohydrate, dextran, pullulan, chitin, chitosan, synthetic carbohydrate, oligo lactate 15-mer, natural polymer, low- or medium-molecular weight polymer, inulin, cyclodextrin, hyaluronic acid, protein, protein-binding agent, integrin- targeting molecule, polycationic, peptide, polyamine, peptide mimic, and/or transferrin.
[00118] A method of treatment using a composition comprising a RNAi agent described herein.
[00119] In one particular specific aspect, the present disclosure relates to a method of treating an APOC3-related disease in an individual, comprising the step of administering to the individual a therapeutically effective amount of a composition comprising an RNAi agent comprising an antisense strand, wherein the antisense strand comprises at least 14 contiguous nucleotides differing by 0, 1 , 2, or 3 nucleotides from the antisense strand of an RNAi agent to APOC3 selected from those specific duplexes provided above and as listed in any of the tables herein. In one aspect, the RNAi agent to APOC3 comprises an antisense strand duplexed with a sense strand, wherein the sense and antisense strands are selected from one or more of the sequences provided in any of the tables herein. [00120] Various particular specific aspects of this aspect are described below. Any aspects disclosed herein that are not mutually exclusive can be combined.
[00121] In one aspect, the present disclosure relates to such a method, wherein the composition comprising a RNAi agent further comprises a sense strand, wherein the sense strand comprises at least 14 contiguous nucleotides differing by 0, 1 , 2, or 3 nucleotides from the sense strand of a RNAi agent to APOC3 selected from the specific duplexes provided herein and as listed, e.g., in any Table herein.
[00122] In one aspect of the method, the RNAi agent comprises at least an anti-sense strand, and/or comprises a sense and an anti-sense strand, wherein the sequence of the sense and/or anti-sense strand is the sequence of the sense and/or the anti-sense strand of a RNAi agent to APOC3 selected from those specific duplex provided herein and as listed, e.g., in Table 1 , wherein the composition further comprises a pharmaceutically effective formulation.
[00123] In one aspect of the method, the RNAi agent comprises at least an anti-sense strand, and/or comprises a sense and an anti-sense strand, wherein the sequence of the sense and/or anti-sense strand comprises the sequence of the sense and/or the anti-sense strand of a RNAi agent to APOC3 selected from those specific duplex provided herein and as listed, e.g., in any of the tables herein, wherein the composition further comprises a pharmaceutically effective formulation.
[00124] In one aspect, the APOC3-related disease is selected from hypertriglyceridemia (e.g., Type V Hypertriglyceridemia), abnormal lipid metabolism, abnormal cholesterol metabolism, atherosclerosis, hyperlipidemia, diabetes, including Type 2 diabetes, obesity, cardiovascular disease, and coronary artery disease, among other disorders relating to abnormal metabolism or otherwise.
[00125] In one aspect, the method further comprises the step of administering an additional treatment.
[00126] In one aspect, the additional treatment is a method (or procedure). In one aspect, the additional treatment is a therapeutically effective dose of a composition.
[00127] In one aspect, the additional treatment and the RNAi agent can be administered in any order, or can be administered simultaneously.
[00128] In one aspect, the method further comprises the step of administering an additional treatment for an APOC3-related disease.
[00129] In one aspect, the method further comprises the step of administering an additional treatment. A RNAi agent to APOC3 can be used in conjunction with any additional treatment disclosed herein, as appropriate for the disease, optionally, in further conjunction with one or more additional RNAi agents to APOC3. [00130] It will be understood that references to any additional treatment are meant to also include the pharmaceutically acceptable salts of any of the active substances. If active substances comprised by components (a) and/or (b) have, for example, at least one basic center, they can form acid addition salts. Corresponding acid addition salts can also be formed having, if desired, an additionally present basic center. Active substances having an acid group, e.g., COOH, can form salts with bases. The active substances comprised in components (a) and/or (b) or a pharmaceutically acceptable salts thereof may also be used in form of a hydrate or include other solvents used for crystallization.
[00131] In one aspect, the composition comprises a second RNAi agent to APOC3. In various aspects, the second RNAi agent is physically distinct from the first, or the two are physically connected (e.g., linked or conjugated). In some aspects, the first and second RNAi agents are combined within the same composition (e.g., both in the same lipid nanoparticle).
[00132] A method of inhibiting the expression of APOC3, using an RNAi comprising an RNAi agent described herein.
[00133] In one particular specific aspect, the present disclosure relates to a method of inhibiting the expression of APOC3 in an individual, comprising the step of administering to the individual a therapeutically effective amount of a composition comprising an RNAi agent of the disclosure. In one aspect, the RNAi comprises a sense strand and an antisense strand, wherein the antisense strand comprises at least 14 contiguous nucleotides differing by 0, 1 , 2, or 3 nucleotides from the antisense strand of an RNAi agent to APOC3 selected from those specific duplexes provided above and as listed in any of the tables herein.
[00134] In one aspect of the method, the RNAi agent comprises at least an anti-sense strand, and/or comprises a sense and an anti-sense strand, wherein the sequence of the sense and/or anti-sense strand is the sequence of the sense and/or the anti-sense strand of a RNAi agent to APOC3 selected from those specific duplex provided herein and as listed, e.g., in any of the tables herein, wherein the composition is in a pharmaceutically effective formulation.
[00135] In one aspect of the method, the RNAi agent comprises at least an anti-sense strand, and/or comprises a sense and an anti-sense strand, wherein the sequence of the sense and/or anti-sense strand comprises the sequence of the sense and/or the anti-sense strand of a RNAi agent to APOC3 selected from those specific duplex provided herein and as listed, e.g., in any of the tables herein, wherein the composition is in a pharmaceutically effective formulation.
[00136] Various particular specific aspects of this aspect are described below. [00137] In one aspect, the individual is afflicted with or susceptible to an APOC3-related disease.
[00138] In one aspect, the APOC3-related disease is selected from hypertriglyceridemia (e.g., Type V Hypertriglyceridemia), abnormal lipid metabolism, abnormal cholesterol metabolism, atherosclerosis, hyperlipidemia, diabetes, including Type 2 diabetes, obesity, cardiovascular disease, and coronary artery disease, among other disorders relating to abnormal metabolism or otherwise.
[00139] In one aspect, the method further comprises the step of administering an additional treatment.
[00140] In one aspect, the additional treatment and the RNAi agent can be administered in any order or can be administered simultaneously.
[00141] In one aspect, the method further comprises the step of administering an additional treatment for an ApoC3 condition such as hypertriglyceridemia (e.g., Type V
Hypertriglyceridemia), abnormal lipid metabolism, abnormal cholesterol metabolism, atherosclerosis, hyperlipidemia, diabetes, including Type 2 diabetes, obesity, cardiovascular disease, and coronary artery disease, among other disorders relating to abnormal metabolism or otherwise.
[00142] In one aspect, the composition comprises a second RNAi agent to APOC3. In various aspects, the second RNAi agent is physically separate from the first, or the two are physically connected (e.g., covalently linked or otherwise conjugated). In some aspects, the first and second RNAi agents are combined within the same composition (e.g., both in the same lipid nanoparticle).
[00143] In one aspect, the method further comprises the step of administering an additional RNAi agent which comprises at least 14 contiguous nucleotides differing by 0, 1 , 2, or 3 nucleotides from the antisense strand of a RNAi agent to APOC3 selected from the specific duplexes provided herein and as listed, e.g., in any Table herein.
[00144] Pharmaceutical compositions of a RNAi agent to APOC3
[00145] In one particular specific aspect, the present disclosure relates to a composition comprising a RNAi agent of the present disclosure. In one aspect, the RNAi agent comprises at least an anti-sense strand, and/or comprises a sense and an anti-sense strand, wherein the anti-sense strand comprises at least 14 contiguous nucleotides differing by 0, 1 , 2, or 3 nucleotides from the anti-sense strand of a RNAi agent to APOC3 selected from those specific duplex provided herein and as listed, e.g., in any of the tables herein, wherein the composition is in a pharmaceutically effective formulation.
[00146] In one aspect, the RNAi agent comprises at least an anti-sense strand, and/or comprises a sense and an anti-sense strand, wherein the sequence of the sense and/or anti- sense strand is the sequence of the sense and/or the anti-sense strand of a RNAi agent to APOC3 selected from those specific duplex provided herein and as listed, e.g., in any of the tables herein, wherein the composition is in a pharmaceutically effective formulation.
[00147] In one aspect, the RNAi agent comprises at least an anti-sense strand, and/or comprises a sense and an anti-sense strand, wherein the sequence of the sense and/or anti- sense strand comprises the sequence of the sense and/or the anti-sense strand of a RNAi agent to APOC3 selected from those specific duplex provided herein and as listed, e.g., in any of the tables herein, wherein the composition is in a pharmaceutically effective formulation.
[00148] In one aspect, the present disclosure pertains to the use of a RNAi agent in the manufacture of a medicament for treatment of an APOC3-related disease, wherein the RNAi agent comprises a sense strand and an antisense strand, wherein the antisense strand comprises at least 14 contiguous nucleotides differing by 0, 1 , 2, or 3 nucleotides from the antisense strand of a RNAi agent to APOC3 selected from those specific duplex provided herein and as listed, e.g., in any Table herein.
[00149] Specific Aspects of RNAi Agents to APOC3 comprising mismatches from the disclosed sequences
[00150] Various specific aspects of a RNAi agent to APOC3 are disclosed herein. The present disclosure encompasses the example modified variants provided in any of the tables herein, and the corresponding unmodified sequences and other modified variants. Specific aspects of the present disclosure include RNAi agents which comprise sequences differing by 0, 1 , 2, or 3 nt (nucleotides) or bp [basepair(s)] (e.g., with 0, 1 , 2 or 3 mismatches) from any of the RNAi agents listed in any of the tables herein, and modified and unmodified variants thereof. As described in additional detail below, a mismatch is defined herein as a difference between the base sequence (e.g., A instead of G) or length when two sequences are maximally aligned and compared. In addition, as described in more detail below, an "unmodified variant" is a variant in which the base sequence is identical, but none of the bases are modified; this includes, for example, a sequence identical to the corresponding portion of the wild-type APOC3 mRNA or gene. A "modified variant" contains one or more modifications (or one or more fewer or different modifications) to a nucleotide, sugar, phosphate or backbone, and/or addition of one or more moieties; but without a change, substitution, addition, or deletion to the base sequence. A particular sequence and its modified or unmodified variants have 0 mismatches among them.
[00151] In one particular aspect, the present disclosure comprises a RNAi agent comprising a sense and an anti-sense strand, wherein the sense and/or anti-sense strand comprises at least 14 contiguous nucleotides differing by 0, 1 , 2, or 3 nt from the sequence of the sense and/or anti-sense strand of: any of the RNAi agents listed in any of the tables herein, and modified and unmodified variants thereof.
[00152] In another particular aspect, the RNAi agent comprises a sense strand comprising at least 14 contiguous nucleotides differing by 0, 1 , 2, or 3 nt from the sense strand of any of the RNAi agents listed in any of the tables herein, and modified and unmodified variants thereof.
[00153] Other aspects
[00154] Various particular specific aspects of this disclosure are described below. Any aspects disclosed herein that are not mutually exclusive can be combined.
[00155] In one aspect, the disclosure pertains to a composition according to any of the above aspects, for use in a method of treating an APOC3-related disease in an individual, the method comprising the step of administering to the individual a therapeutically effective amount of a composition according to any of the claims.
[00156] Various particular specific aspects of this aspect are described below.
[00157] In one aspect, the disclosure pertains to the composition according to any of the above aspects, for use in a method of inhibiting the expression of APOC3 in an individual, the method comprising the step of administering to the individual a therapeutically effective amount of a composition according to any of the above aspects.
[00158] One aspect of the disclosure is the use of a composition according to any of the above aspects, in the manufacture of a medicament for treatment of an APOC3-related disease.
[00159] In one aspect, the APOC3-related disease is selected from any disease listed herein.
[00160] In one aspect, the disclosure pertains to the composition of any of the above aspects, for use in the treatment of an APOC3-related disease.
[00161] In one aspect, the APOC3-related disease is selected from any disease listed herein.
[00162] In one aspect, the disclosure relates to a method of inhibiting the expression of APOC3 in an cell, comprising the step of introducing into the cell a composition comprising an RNAi agent comprising an antisense strand, wherein the antisense strand comprises at least 14 contiguous nucleotides differing by 0, 1 , 2, or 3 nucleotides from the antisense strand of an RNAi agent to APOC3 selected from the APOC3 siRNAs disclosed herein.
[00163] In one aspect, the disclosure relates to a method of inhibiting the expression of APOC3 in an cell, comprising the step of introducing into the cell a composition comprising an RNAi agent comprising a sense strand and an antisense strand, wherein the antisense strand comprises at least 14 contiguous nucleotides differing by 0, 1 , 2, or 3 nucleotides from the antisense strand, and the sense strand comprises at least 14 contiguous nucleotides differing by 0, 1 , 2, or 3 nucleotides from the sense strand of an RNAi agent to APOC3 selected from the APOC3 siRNAs disclosed herein.
[00164] Definitions
[00165] For convenience, the meaning of certain terms and phrases used in the
specification, examples, and appended claims, are provided below. If there is an apparent discrepancy between the usage of a term in other parts of this specification and its definition provided in this section, the definition in this section shall prevail.
[00166] APOC3-related diseases
[00167] siRNAs to APOC3 can be used to treat APOC3-related diseases. An "APOC3- related disease" is any disease associated with APOC3 and/or a mutation and/or an over- expression of a wild-type and/or mutant APOC3, and/or diseases wherein disease progression is enhanced by or prognosis worsened by the presence of APOC3 and/or a mutation and/or an over-expression of wild-type and/or mutant APOC3. Non-limiting examples of APOC3-related diseases include: hypertriglyceridemia (e.g., Type V
Hypertriglyceridemia), abnormal lipid metabolism, abnormal cholesterol metabolism, atherosclerosis, hyperlipidemia, diabetes, including Type 2 diabetes, obesity, cardiovascular disease, and coronary artery disease, among other disorders relating to abnormal metabolism or otherwise.
[00168] In various aspects, the present disclosure pertains to the use of APOC3 RNAi agents in inhibiting and/or reducing the level and/or activity of APOC3, for treatment of APOC3-related diseases, particularly those diseases in which the level and/or activity of APOC3 is excessive. Such diseases include hypertriglyceridemia (e.g., Type V
Hypertriglyceridemia), abnormal lipid metabolism, abnormal cholesterol metabolism, atherosclerosis, hyperlipidemia, diabetes, including Type 2 diabetes, obesity, cardiovascular disease, and coronary artery disease, among other disorders relating to abnormal metabolism or otherwise.
[00169] Thus, the present disclosure encompasses APOC3 RNAi agents and the uses thereof for APOC3-related diseases.
[00170] USE OF APOC3 RNAi AGENTS TO TREAT APOC3-RELATED DISEASES
[00171] In various aspects, the present disclosure pertains to the use of APOC3 RNAi agents, which inhibit and/or decrease the level and/or activity of APOC3, to treat APOC3- related diseases. [00172] In earlier studies, inhibiting or decreasing the level and/or activity of APOC3 have been shown to be effective in treatment of APOC3 related diseases.
APOC3 RNAi agent for use in treating various APOC3-related diseases
[00173] In one aspect, the APOC3 RNAi agent of the present disclosure comprises a sequence disclosed herein and is administered to a patient in need thereof (e.g., a patient suffering from an APOC3-related disease disclosed herein or known in the literature). In one aspect, the APOC3 RNAi agent of the present disclosure is administered to a patient in need thereof, along with one or more additional pharmaceutical agent appropriate for that disease. For example, a patient suffering from an APOC3-related disease can be administered a pharmacologically effective amount of one or more APOC3 RNAi agent along with a pharmacologically effective amount of one or more of any APOC3-related disease treatment listed herein, and/or any other APOC3-related disease treatment known in the art.
[00174] A patient suffering from an APOC3-related disease can be administered one or more RNAi agent to APOC3 and one or more additional APOC3-related disease treatment. This additional treatment can be selected from the list of any disease treatment listed herein, and/or any anti- APOC3-related disease treatment known in the art.
[00175] The APOC3 RNAi agents of the instant disclosure can be administered along with (as part of the same therapeutic treatment regimen, prior to, simultaneously with, or after) one or more additional therapeutics to treat an APOC3-related disease. These one or more additional therapeutics can be selected from: iron administration, chelation therapy, phlebotomy, erythropoiesis stimulating agent (ESA) (e.g., Epoetin alfa or darbepoetin alfa), anti-APOC3 antibody, hemodialysis, and hyperbaric oxygen.
[00176] The patient can also be administered more than one RNAi agent to APOC3.
[00177] In the case of APOC3-related diseases, the RNAi agent(s) and additional disease treatment(s) can be administered in any order, simultaneously or sequentially, or in multiple doses over time. Administration of the RNAi agent and the additional treatment can be, for example, simultaneous, concurrent, separate or sequential.
[00178] Simultaneous administration may, e.g., take place in the form of one fixed combination with two or more active ingredients, or by simultaneously administering two or more active ingredients that are formulated independently. Sequential use (administration) preferably means administration of one (or more) components of a combination at one time point, other components at a different time point, that is, in a chronically staggered manner, preferably such that the combination shows more efficiency than the single compounds administered independently (especially showing synergism). Separate use (administration) preferably means administration of the components of the combination independently of each other at different time points, preferably meaning that the components (a) and (b) are administered such that no overlap of measurable blood levels of both compounds are present in an overlapping manner (at the same time).
[00179] Also combinations of two or more of sequential, separate and simultaneous administration are possible, preferably such that the combination component-drugs show a joint therapeutic effect that exceeds the effect found when the combination component-drugs are used independently at time intervals so large that no mutual effect on their therapeutic efficiency can be found, a synergistic effect being especially preferred.
[00180] The term "delay of progression" as used herein means administration of the combination to patients being in a pre-stage or in an early phase, of the first manifestation or a relapse of the disease to be treated, in which patients, e.g., a pre-form of the
corresponding disease is diagnosed or which patients are in a condition, e.g., during a medical treatment or a condition resulting from an accident, under which it is likely that a corresponding disease will develop.
[00181] "Jointly therapeutically active" or "joint therapeutic effect" means that the compounds may be given separately (in a chronically staggered manner, especially a sequence-specific manner) in such time intervals that they preferably, in the warm-blooded animal, especially human, to be treated, still show a (preferably synergistic) interaction (joint therapeutic effect). Whether this is the case, can inter alia be determined by following the blood levels, showing that both compounds are present in the blood of the human to be treated at least during certain time intervals.
[00182] Methods of identifying APOC3 RNAi agents
[00183] Any method known in the art can be used to identify efficacious APOC3 RNAi agents useful for treating APOC3-related diseases. A few non-limiting examples of such methods are disclosed herein. Any one or more of the following, in addition to other methods known in the art, can be used.
[00184] For example: In various steps, APOC3 RNAi agents are tested for efficacy and tolerance to modifications and modification from 19-mer to 18-mer format.
[00185] For example: APOC3 RNAi candidates can be tested by target knockdown in Huh7 cells and primary hepatocytes. This can be tested using any method known in the art, including but not limited to RNAi Max. Preferably, the RNAi candidate demonstrates at least about 80% mRNA knockdown at 48 hours. The mechanism can optionally be confirmed to be RNAi (RNA interference) by identifying specific cleavage products using RACE.
[00186] Cross-reactivity with cyno monkeys can be assessed in primary cyno hepatocytes at 48 hours. Optionally, the IC50 can be within five-fold of human siRNA activity. [00187] Off-target effects of the candidate APOC3 RNAi agent can be assed. For example, cell growth in non-ApoC3-expressing cells can be tested. Preferably, there is no effect relative to, for example, YFP siRNA.
[00188] Immune stimulation by the candidate RNAi agent can be tested using whole blood assay. Preferably, there would be no detectable activation of cytokine expression.
[00189] Toxicity can also be assessed. MTD (maximum tolerated dosage) can be assessed in mice. A Mini-tox study can be performed for 2 weeks. Optionally, a conjugate can be used.
[00190] Efficacy in animal models can be assessed. For example, a conjugate can be used to deliver mouse ApoC3 siRNA to liver and effectively knockdown the target. Preferably, the mRNA knockdown will be at least about 80% at 96 hours.
[00191] Efficacy in cyno monkeys can also be tested.
[00192] PK (pharmacokinetics) and bio-distribution of formulated RNAi agents can be tested, for example, in rodents, using, for example, LNP (lipid nanoparticle)-formulated RNAi agents. Preferably, the RNAi targets the liver.
[00193] These examples are meant to be non-limiting examples of methods that can be used to test candidate or potential APOC3 RNAi agents.
[00194] These and other methods known in the art and/or known or available to one of ordinary skill can be used to test candidate APOC3 RNAi agents.
[00195] ApoC3 RNAi agent candidate chemistry optimization
[00196] In particular, several steps can be performed for optimizing the chemistry of the lead(s) APOC3 RNAi agent(s):
[00197] APOC3 sequences are identified, including various lead sequences. Serum stability highlights the 5' terminal U as a fragile site.
[00198] Tolerance for chemical modification is assessed. Many APOC3 sequences appear to be the tolerant for chemical modification. Indirect evidence shows that 5' dT vs 5'U does stabilize the 5' end of the guide strand. No "more modified" format did improve potency.
[00199] The 3' overhang is optimized, e.g., PAZ ligands are screened.
[00200] Passenger strand optimization is done. For example, this was done using the assymetrical design.
[00201] Internal modifications for stabilization are assessed. For example, chemical stabilization at the cleavage sites is tested.
[00202] These and other tests available or known to one of ordinary skill in the art can be used to evaluate and modify the chemistry of a candidate APOC3 RNAi agent.
[00203] APOC3 RNAi Agent candidate selection [00204] The instant disclosure pertains to RNAi agents to APOC3 for use in treating APOC3-related diseases. These agents have various structural properties, including having an 18-mer sequence. Optionally, the agent is modified, including but not limited to, modifications of one or more sugar or phosphate, and with, in 3' order, a linker and a 3' end cap. The 5' and/or 3' end of the sense strand can be conjugated to other components which increase targeting to and/or uptake by the liver.
[00205] The disclosure also pertains to compositions comprising the RNAi agent to
Hepicidin and, for example, an excipient or pharmaceutically acceptable carrier.
[00206] The APOC3 mRNA is unusually short, thus leaving only a small room for siRNA design, and very small room for pan-specific siRNAs (those which target both humans and one or more test animals).
[00207] In this work, using the human sequence, 419 possible 19-mer APOC3 RNAi agents were identified.
[00208] A subset of these were selected and tested. These sequences are shown in Table 1 (as DNA 19-mers) and Table 2 (as RNA 19-mers).
[00209] VARIOUS APOC3 RNAi AGENTS
[00210] As noted above, the disclosure thus relates to compositions comprising an APOC3 RNAi agent. These comprise a first strand and a second strand, wherein the sequence of the first and/or second strand is, comprises, comprises 15 contiguous nt of, or comprises 15 contiguous nt of (with 1-3 mismatches from) any sequence of any APOC3 RNAi agent listed herein. In some embodiments, the disclosure pertains to an APOC3 RNAi agent comprising a first and second strand, wherein the sequence of the first and/or second strand is, comprises, or comprises 15 contiguous nt of any sequence of any APOC3 RNAi agent disclosed herein, except that one or both strands is nicked and/or one or more nt of the sequence disclosed herein has been replaced by a spacer (e.g., the first comprises the sequence of nt 1 -5 and 7-18, or of 1 -4 and 6-18 of a sequence disclosed herein). These APOC3 RNAi agents can be of any format described herein or known in the art. These include the 18-mer format, the 19-mer format, the internal spacer format, the shortened sense strand format, a format comprising a nicked strand, or any other format or length of RNAi agent known in the art. RNAi agents of the various formats can comprise strands of various lengths, one or more spacer, modified internucleoside linker, and 3' end cap. RNAi agents of the 18-mer format, for example, comprise a sense and an anti-sense strand, each strand being an 18-mer and the strands together forming a blunt-ended duplex, wherein the 3' end of at least one strand terminates in a phosphate or modified internucleoside linker and further comprises, in 5' to 3' order: a spacer; a second phosphate or modified
internucleoside linker; and a 3' end cap. In some embodiments, the 3' end of both the sense and anti-sense strand further comprise, in 5' to 3' order: a spacer; a second phosphate or modified internucleoside linker; and a 3' end cap. The two strands can have the same or different spacers, phosphate or modified internucleoside linker, and/or 3' end caps. In various embodiments, one or more nt can be modified and/or substituted. Various formats, spacers, modified internucleoside linkers and 3' end caps are described below.
[00211] VARIOUS FORMATS OF RNAi AGENTS: 18-mer format
[00212] The present disclosure contemplates a variety of different formats for APOC3 RNAi agents, including various formats using any APOC3 RNAi agent sequence disclosed herein.
[00213] A particular novel format, designated the 18-mer format, has been developed. This and any other format can be used for an APOC3 RNAi agent.
[00214] siRNAs naturally generated in a cell by Dicer typically comprise two 21-nt RNA strands, which form a 19-bp duplex region and two dinucleotide overhangs. This is the so- called "canonical" siRNA structure. See, for example, Elbashir et al. 2001 Nature 41 1 : 494- 498; Elbashir et al. 2001 EMBO J. 20: 6877. This is the so-called "canonical" structure or "21-mer" format of siRNAs.
[00215] The two dinucleotide overhangs do not contribute to target specificity. Elbashir et al. 2001 Nature 41 1 : 494-498; Elbashir et al. 2001 EMBO J. 20: 6877-6888; and Kraynack et al. 2006 RNA 12:163-176. They do, however, help protect the ends of the siRNA from nuclease degradation and sometimes improve activity. However, these di-nucleotides can optionally be replaced by 3' end caps. Ideal 3' end caps allow RNA interference activity, while increasing stability, e.g., against nucleases such as those in blood serum or intestinal fluid. U.S. Pat. No. 8,097,716; and 8,084,600. Novel 3' end caps (some described as "PAZ ligands") are disclosed herein; these can be used in place of or in addition to a 3' end cap in APOC3 RNAi agents. It is noted that, although some 3' end caps are designated "PAZ ligands", this disclosure is not bound by any particular theory.
[00216] As noted above, the canonical siRNA structure comprises two strands, each a 19- mer with a 2 nt overhang. Strands of shorter length, e.g., 18-mers, are less commonly produced by a cell.
[00217] While a few 18-mer RNAi agents have shown to have efficacy, many do not.
Disclosed here is a 18-mer format shown to be efficacious in producing functional RNAi agents to a variety of different targets, with a variety of different sequences.
[00218] The disclosure thus relates to compositions comprising an APOC3 RNAi agent having a novel format (the "18-mer format"). These RNAi agents comprise a sense and an anti-sense strand, each strand being an 18-mer and the strands together forming a blunt- ended duplex, wherein the 3' end of at least one strand further comprises, in 5' to 3' order: a spacer; a phosphate or modified internucleoside linker; and a 3' end cap. In some embodiments, each strand terminates at the 5' end with a hydroxyl, optionally linked to a 5' end cap or a ligand, and terminates at the 3' end with a phosphate or modified
internucleoside linker. In some embodiments, the 3' end of both the sense and anti-sense strand further comprise, in 5' to 3' order: a spacer; a phosphate or modified internucleoside linker; and a 3' end cap. The two strands can have the same or different spacers, phosphate or modified internucleoside linker, and/or 3' end caps. The strands can be ribonucleotides, or, optionally, one or more nucleotide can be modified or substituted.
Optionally, at least one nucleotide comprises a modified internucleoside linker. Optionally, the RNAi agent can be modified on one or both 5' end. Optionally, the sense strand can comprise a 5' end cap which reduces the amount of the RNA interference mediated by this strand. Optionally, the RNAi agent is attached to a ligand. The disclosure also relates to processes for making such compositions, and methods and uses of such compositions, e.g., to mediate RNA interference.
[00219] In one embodiment, the 3' end of both the sense and anti-sense strand terminate in a phosphate or modified internucleoside linker and further comprise in 5' to 3' order: a spacer, a phosphate or modified internucleoside linker, and a 3' end cap.
[00220] In various embodiments, the spacer is a sugar, alkyl, cycloakyl, ribitol or other type of abasic nucleotide, 2'-deoxy-ribitol, diribitol, 2'-methoxyethoxy-ribitol (ribitol with 2'-MOE),
C3, C4, C5, C6, or 4-methoxybutane-1 ,3-diol (5300). In various embodiments, the spacers on the sense and anti-sense strands can be the same or different.
[00221] In various embodiments, the modified internucleoside linker is selected from phosphorothioate, phosphorodithioate, phosphoramidate, boranophosphonoate, an amide
O
3- p= G
i
R4
linker, and a compound of formula (I): -— - (I), where R3 is selected from O", S", NH2, BH3, CH3, C1-6 alkyl, C6-io aryl, C1-6 alkoxy and C6-io aryl-oxy, wherein C1-6 alkyl and C6-io aryl are unsubstituted or optionally independently substituted with 1 to 3 groups independently selected from halo, hydroxyl and NH2; and R4 is selected from O, S, NH, or CH2. The internucleoside linker of formula (I) can be used in any RNAi agent of any format, e.g., any APOC3 RNAi agent disclosed herein of any format disclosed herein.
[00222] RNAi AGENT FORMAT: Internal spacer
[00223] As noted above, the present disclosure contemplates a variety of different formats for APOC3 RNAi agents, including various formats using any APOC3 RNAi agent sequence disclosed herein. [00224] A particular novel format, designated the internal spacer, has been developed.
[00225] In this format, one or more nucleoside subunits ([sugar+base]) of one or both strands has been replaced by a spacer. In various embodiments, the nucleobase subunit ([sugar+base]) replaced by a spacer is at position 1 , 3, 5, 6, 7, 15, 16, 17, or 18 (counting 5' to 3'). In various embodiments, the nucleobase subunit ([sugar+base]) replaced by a spacer is at position 5, 6, or 17 (counting 5' to 3'). In various embodiments, the spacer can be sugar, alkyl, cycloakyl, ribitol or other type of abasic nucleotide, 2'-deoxy-ribitol, diribitol, 2'- methoxyethoxy-ribitol (ribitol with 2 -MOE), C3, C4, C5, C6, or 4-methoxybutane-1 ,3-diol (5300), or any other spacer described herein or known in the art. More than one different type of spacer can be incorporated into the same composition.
[00226] The internal spacer format can be combined with other formats. For example, the internal spacer format can be combined with the 19-mer format. This produces a RNAi agent comprising a first strand and a second strand, wherein the first and second strand each comprise a 19-mer (optionally further comprising a 3' end and/or 3' terminal dinucleotide), wherein one or more nucleoside subunits of the first and/or second strand are replaced by a spacer.
[00227] The internal spacer format can also be combined, for example, with the 18-mer format. For example, the APOC3 RNAi agent can comprise a first and a second strand, wherein the both strands are 18-mers, and wherein the first strand is the anti-sense strand and a nucleoside subunit ([consisting of a sugar and base]) at any of positions 1 to 18 is replaced by a spacer. One or more nucleoside subunits can be replaced. Alternatively, one or more nucleoside subunits on the first and/or second strand can be replaced by a spacer.
[00228] Non-limiting examples of APOC3 RNAi agents having an internal spacer format are described herein.
[00229] This disclosure also pertains to any RNAi agent of any sequence, targeting any target, which has the internal spacer format. For example, if the anti-sense strands of two different duplexes are identical except that, at one position, the nucleobase unit (i.e., sugar + base) has been replaced by a spacer (e.g., a spacer described herein), the sequences are considered to have one mismatch (e.g., to differ by 1 nt). Thus, a first sequence comprising at least 14 contiguous nt differing by 0, 1 , 2 or 3 nt from a second sequence can have any combination of 0, 1 , 2 or 3 internal spacer, substitution, and/or other position wherein the base at one sequence does not match the corresponding base at the other sequence.
[00230] This disclosure also notes that, for the purposes of counting mismatches between sequences, the replacement of a nt with a spacer would count as a mismatch.
[00231] RNAi AGENT FORMAT: SHORTENED SENSE STRAND [00232] As noted above, the present disclosure contemplates a variety of different formats for APOC3 RNAi agents, including various formats using any APOC3 RNAi agent sequence disclosed herein.
[00233] A particular novel format, designated the shortened sense strand, has been developed.
[00234] In various embodiments, the APOC3 RNAi agent comprises an anti-sense and a sense strand, wherein the sense strand has been shortened. In various embodiments, the sense strand is a 14-, 15-, 16-, or 17-mer.
[00235] The shortened sense strand format can be combined with any RNAi agent format known. This includes but is not limited to the 18-mer format.
[00236] For example, an APOC3 RNAi agent can comprise an anti-sense strand which is an 18-mer (or longer), and a sense strand which is a 14-, 15-, 16-, or 17-mer. The anti- sense strand can be an 18-, 19-, 20, 21 -, 22-, 23-, 24-, 25-, 26-, 27-, 28-, 29-, or 30-mer or longer. The 3' terminus of the anti-sense and/or sense strand can optionally terminate a phosphate or modified internucleoside linker and further comprise: (a) a 3' end cap; or (b) a spacer, a second phosphate or modified internucleoside linker, and a 3' end cap (in 5' to 3' order). The spacer, modified internucleoside linker and 3' end cap can be any of those elements detailed herein or known in the art. A RNAi agent of the shortened sense strand format can comprise RNA subunits (ribonucleotides) wherein one or more ribonucleotides is modified and/or substituted by any modification or substitution described herein or known in the art.
[00237] The efficacy of various APOC3 RNAi agents having a shortened sense strand format is shown herein.
[00238] The disclosure also pertains to a RNAi agent of any sequence, targeting any gene target, which has a shortened sense strand format.
[00239] COMPONENTS OF APOC3 RNAi AGENTS
[00240] As noted above, APOC3 RNAi agents can pertain to sequences disclosed herein, and be of any of several formats described herein or known in the art.
[00241] Components of these various RNAi agents include different 3' end caps, spacers, and modified internucleoside linkers. Components can be mixed and matched.
[00242] These components are described in additional detail below.
[00243] 3' END CAPS
[00244] Various formats, each useful for producing an APOC3 RNAi agent comprise a 3' end cap. One or both strands of an APOC3 RNAi agent can comprise, at the 3' terminus: a 3' end cap; or a spacer, a phosphate or internucleoside linker, and a 3' end cap. [00245] Any of the various 3' end caps disclosed herein or known in the art can be used to prepare an APOC3 RNAi agent. In various embodiments, the 3' end cap is selected from those represented by formula 1 a or 1 b, disclosed in Tables 5A, 5B, 5C, 5D, 5E, or otherwise described herein. In various embodiments, the 3' end caps on the sense and anti-sense strands can be the same or different.
[00246] In one embodiment, the 3' end cap encompasses a compound of formula la:
Figure imgf000046_0001
la in which:
X is a 3' end of molecule comprising: a strand of an APOC3 RNAi agent, wherein the 3' end of the strand terminates in a phosphate or modified
internucleoside linker and optionally further comprises, in 5' to 3' order: a spacer and a second phosphate or modified internucleoside linker;
Y is selected from CH and N;
m is selected from 0 and 1 ;
p is selected from 1 , 2 and 3;
R3 is selected from hydrogen, 2-(hydroxy-methyl)-benzyl, 3-(hydroxy-methyl)- benzyl and succinate; or is attached to a solid support; wherein the (CH2)m-0-R3 moiety is attached to the phenyl ring at position 3 or 4;
R4 is hydrogen;
R5 is hydrogen; or R4 and R5, together with the phenyl rings to which R4 and R5 are attached, form 6H-benzo[c]chromene.
[00247] In various embodiments, the 3' end cap encompasses a compound selected from Table 5A.
TABLE 5A.
Figure imgf000047_0001

Figure imgf000048_0001
in which:
X is a 3' end of a molecule comprising: a strand of an APOC3 RNAi agent, wherein the 3' end of the strand terminates in a phosphate or modified
internucleoside linker and optionally further comprises, in 5' to 3' order: a spacer and a second phosphate or modified internucleoside linker. In one embodiment, the 3' end cap encompasses a compound of formula lb:
Figure imgf000048_0002
lb in which:
X is a 3' end of a molecule comprising: a strand of an APOC3 RNAi agent, wherein the 3' end of the strand terminates in a phosphate or modified
internucleoside linker and optionally further comprises, in 5' to 3' order: a spacer and a second phosphate or modified internucleoside linker;
q is selected from 0, 1 and 2;
R6 is selected from phenyl which is unsubstituted or substituted with benzoxy; R7 is selected from hydrogen and hydroxy-ethyl, wherein if R7 is hydroxy-ethyl, the hydroxyl can be optionally functionalized as succinate or attached to a solid support;
R8 is selected from hydrogen and methoxy;
Yi is selected from CH and N; and
Y2 is selected from N and CR9; wherein R9 is selected from hydrogen and methyl.
[00249] In various embodiments, the 3' end cap encompasses a compound selected from Table 5B.
TABLE 5B.
Figure imgf000049_0001
Figure imgf000050_0001

Figure imgf000051_0001
Figure imgf000051_0002
in which:
X is a 3' end of a molecule comprising: a strand of an APOC3 RNAi agent, wherein the 3' end of the strand terminates in a phosphate or modified internucleoside linker and optionally further comprises, in 5' to 3' order: a spacer and a second phosphate or modified internucleoside linker.
[00250] In various embodiments, the 3' end cap encompasses a compound selected from Table 5C.
TABLE 5C.
Figure imgf000052_0001
in which:
X is a 3' end of a molecule comprising: a strand of an APOC3 RNAi agent, wherein the 3' end of the strand terminates in a phosphate or modified
internucleoside linker and optionally further comprises, in 5' to 3' order: a spacer and a second phosphate or modified internucleoside linker, and
q is selected from 1 and 2. [00251] In various embodiments, the 3' end cap encompasses a compound selected from Table 5D.
TABLE 5D.
Figure imgf000053_0001
Figure imgf000054_0001
53
Figure imgf000055_0001
54
Figure imgf000056_0001
Figure imgf000056_0002
Figure imgf000057_0001
Figure imgf000058_0001
Figure imgf000059_0001
Figure imgf000060_0002
in which:
X is a 3' end of a molecule comprising: a strand of an APOC3 RNAi agent, wherein the 3' end of the strand terminates in a phosphate or modified
internucleoside linker and optionally further comprises, in 5' to 3' order: a spacer and a second phosphate or modified internucleoside linker.
[00252] In various embodiments, the 3' end cap encompasses a compound selected from Table 5E.
TABLE 5E.
Figure imgf000060_0001
X is a 3' end of a molecule comprising: a strand of an APOC3 RNAi agent, wherein the 3' end of the strand terminates in a phosphate or modified
internucleoside linker and optionally further comprises, in 5' to 3' order: a spacer and a second phosphate or modified internucleoside linker.
[00253] Suitable 3' end caps, including but not limited to:C3, C6, C12, Triethylene glycol, Cyclohexyl (or Cyclohex), Phenyl, Biphenyl, Adamantane and Lithocholic acid (or Lithochol), are described herein and/or in U.S. Pat. Nos. 8,097,716; 8,084,600; 8,344, 128; 8,404,831 ; and 8,404,832.
[00254] In some embodiments, the 3' end cap is a ribitol. Thus, in some embodiments, the RNAi agent comprises an 18-mer strand further comprising at the 3' terminus, in 5' to 3' order, a spacer (e.g., a sugar or alkyl, cycloakyi, ribitol or other type of abasic nucleotide, C3, C4, C5, C6, etc.), a phosphate or modified internucleoside linker, and a 3' end cap (e.g., a second ribitol or other type of abasic nucleotide).
[00255] In some embodiments, the 3' end cap is a diribitol. Thus: In some embodiments, the RNAi agent comprises an 18-mer strand further comprising at the 3' terminus, in 5' to 3' order, a spacer (e.g., a ribitol, C3, C4, C5, C6, etc.), a phosphate or modified internucleoside linker, and a 3' end cap (e.g., a diribitol).
ADDITIONAL ASPECTS OF AN APOC3 RNAi AGENT
[00256] As noted above, the disclosure thus relates to compositions comprising an APOC3 RNAi agent. These APOC3 RNAi agents can be of any format. These include the 18-mer format, the 19-mer format, the internal spacer format, the shortened sense strand format, or any other format or length of RNAi agent known in the art. RNAi agents of the various formats can comprise strands of various lengths, one or more spacer, modified
internucleoside linker, and 3' end cap. RNAi agents of the 18-mer format, for example, comprise a sense and an anti-sense strand, each strand being an 18-mer and the strands together forming a blunt-ended duplex, wherein the 3' end of at least one strand terminates in a phosphate or modified internucleoside linker and further comprises, in 5' to 3' order: a spacer; a second phosphate or modified internucleoside linker; and a 3' end cap. In some embodiments, the 3' end of both the sense and anti-sense strand further comprise, in 5' to 3' order: a spacer; a second phosphate or modified internucleoside linker; and a 3' end cap. The two strands can have the same or different spacers, phosphate or modified
internucleoside linker, and/or 3' end caps. In various embodiments, one or more nt can be modified and/or substituted. Various spacers, modified internucleoside linkers and 3' end caps are described below.
SPACERS: RIBITOL, DIRIBITOL, 2'-DEOXYRIBITOL, 2'-METHOXYETHOXY RIBITOL, C3, C4, C5, C6, OR 4-METHOXYBUTANE-1 ,3-DIOL (5300)
[00257] In the present disclosure, in an APOC3 RNAi agent, any of various spacers can be used in combination with strands of any sequence, with or without substitutions and/or modifications of nt, with phosphates or modified nucleoside spacers, and with any 3' end cap, in any combination without limitation. [00258] A spacer is a chemical moiety intended or used to create or maintain a space (e.g., a proper or functional spacing) between two other chemical moieties; e.g., between two phosphates or modified internucleoside linkers. In various embodiments, the spacer is a ribitol, diribitol, 2'-deoxyribitol, or 2'-methoxyethoxy ribitol (ribitol with 2'-MOE) or an equivalent abasic nucleotide known to one skilled in the art, or a lower alkyl or alkoxy group such as a C3, C4, C5 or C6, or 4-methoxybutane-1 ,3-diol. Various embodiments are described in more detail below.
[00259] Ribitol spacer.
[00260] In some embodiments, the spacer is ribitol.
[00261] In one embodiment, the RNAi agent comprises, in 5' to 3' order: an 18-mer strand (comprising a 3' terminal phosphate or a modified internucleoside linker); a spacer which is ribitol; a phosphate or a modified internucleoside linker; and a 3' end cap (e.g., any 3' end cap described herein or otherwise known in the art). In one embodiment, the RNAi agent comprises, in 5' to 3' order: an 18-mer strand comprising a 3' terminal phosphate; a spacer which is ribitol; a phosphate or a modified internucleoside linker; and a 3' end cap.
[00262] The structure of a 3' terminal phosphate and ribitol spacer is shown here:
Figure imgf000062_0001
[00263] In some documents, the ribitol spacer is designated as N027 (C027, etc.).
[00264] One embodiment the RNAi agent comprises, in 5' to 3' order: an 18-mer strand, wherein the 3' end of the 18-mer strand terminates in a phosphate and further comprises, in 5' to 3' order: a spacer which is ribitol, a phosphate, and a 3' end cap which is X058. This structure can be on any RNAi strand of any sequence or target. In addition, any 3' end cap disclosed herein can be used.
[00265] In one embodiment, ribitol with X058, wherein the last nucleotide of the 18-mer strand is a 2'-MOE and the 3' end of the 18-mer strand terminates in a phosphate and further comprises, in 5' to 3' order: a spacer which is ribitol, a second phosphate, and a 3' end cap which is X058.
[00266] In another embodiment, ribitol with C6 cap, wherein the last nucleotide of the 18- mer strand is and the 3' end of the 18-mer strand terminates in a phosphate and further comprises, in 5' to 3' order: a spacer which is ribitol, a phosphate, and a 3' end cap which is C6. In another embodiment, the RNAi agent comprises, in 5' to 3' order: an 18-mer strand terminating in a 3' phosphate, a ribitol spacer, a phosphate, and a C6 3' end cap. In another embodiment, the RNAi agent comprises, in 5' to 3' order: an 18-mer strand terminating in a 3' phosphate, a ribitol spacer, a phosphate, and a BP 3' end cap. In another embodiment, the RNAi agent comprises, in 5' to 3' order: an 18-mer strand terminating in a 3' phosphate, a ribitol spacer, a phosphate, and a C10 3' end cap.
[00267] In some embodiments, the 3' end cap is a ribitol. Thus, the RNAi agent comprises an 18-mer strand, wherein the 3' end of the 18-mer strand terminates in a phosphate or modified internucleoside linker and further comprises, in 5' to 3' order: a spacer which is ribitol, a second phosphate or modified internucleoside linker, and a 3' end cap which is a second ribitol. In one embodiment, the 3' end of the 18-mer strand terminates in a phosphate and further comprises, in 5' to 3' order: a spacer which is ribitol, a second phosphate, and a 3' end cap which is a second ribitol.
[00268] The structure comprising an RNAi agent comprising, in 5' to 3' order, an 18-mer strand terminating in a 3' terminal phosphate or a modified internucleoside linker, a ribitol spacer, a phosphate or a modified internucleoside linker, and a 3' end cap can be used on any RNAi agent of sequence or target, including but not limited to a double-stranded RNA, wherein optionally one or more phosphates are replaced by a modified internucleoside linker, optionally one or more nucleotides are modified, and optionally one or more RNA nucleotides are replaced by DNA, PNA, LNA, morpholino, TNA, GNA, FANA, ANA, HNA, CeNA, and/or UNA.
[00269] Diribitol spacer.
[00270] In some embodiments the spacer is Diribitol.
[00271] In one embodiment, the RNAi agent comprises, in 5' to 3' order: an 18-mer strand wherein the 3' end of the strand terminates in a phosphate or modified internucleoside linker and further comprises in 5' to 3' order: a spacer (wherein the spacer comprises in 5' to 3' order: a first ribitol; a phosphate or a modified internucleoside linker; a second ribitol; and a phosphate or a modified internucleoside linker); and a 3' end cap.
[00272] Thus: In one embodiment, the RNAi agent comprises, in 5' to 3' order: an 18-mer strand comprising a 3' terminal phosphate; a first ribitol; a phosphate; a second ribitol; a phosphate or a modified internucleoside linker; and a 3' end cap.
[00273] This structure of a 3' terminal phosphate, a first ribitol, a phosphate, and a second ribitol is shown here:
Figure imgf000064_0001
[00274] In one embodiment, the RNAi agent comprises, in 5' to 3' order: an 18-mer strand comprising a 3' terminal phosphate; a first ribitol spacer; a phosphate; a second ribitol spacer; a phosphate or a modified internucleoside linker; and a 3' end cap.
[00275] In one embodiment, the RNAi agent comprises, in 5' to 3' order: an 18-mer strand terminating in a 3' terminal phosphate or a modified internucleoside linker, a first ribitol spacer, a phosphate or a modified internucleoside linker, a second ribitol spacer, a phosphate or a modified internucleoside linker, and a 3' end cap which is a ribitol; this structure is designated a triribitol.
[00276] The structure comprising an RNAi agent comprising, in 5' to 3' order, an 18-mer strand terminating in a 3' terminal phosphate or a modified internucleoside linker, a first ribitol spacer, a phosphate or a modified internucleoside linker, a second ribitol spacer, a phosphate or a modified internucleoside linker, and a 3' end cap can be used on any RNAi agent of sequence or target, including but not limited to a double-stranded RNA, wherein optionally one or more phosphates are replaced by a modified internucleoside linker, optionally one or more nucleotides are modified, and optionally one or more RNA
nucleotides are replaced by DNA, PNA, LNA, morpholino, TNA, GNA, ANA, HNA, FANA, CeNA, and/or UNA.
[00277] 2'-methoxyethoxy ribitol spacer.
[00278] In some embodiments, the spacer is 2'-methoxyethoxy ribitol or other type of abasic nucleotide.
[00279] In one embodiment, the RNAi agent comprises a strand, wherein the 3' end of the strand terminates in a phosphate or modified internucleoside linker and further comprises, in 5' to 3' order: a spacer which is 2'-methoxyethoxy ribitol, a second phosphate or modified internucleoside linker, and a 3' end (e.g., any 3' end cap described herein or known in the art). In other words: In one embodiment, the RNAi agent comprises, in 5' to 3' order: a strand comprising a 3' terminal phosphate or a modified internucleoside linker; a spacer which is 2'- methoxyethoxy ribitol; a phosphate or a modified internucleoside linker; and a 3' end cap (e.g., any 3' end cap described herein or known in the art). Thus: In one embodiment, the RNAi agent comprises, in 5' to 3' order: a strand comprising a 3' terminal phosphate; a spacer which is 2'-methoxyethoxy ribitol; a phosphate or a modified internucleoside linker; and a 3' end cap.
[00280] The structure of the 3' terminal phosphate and 2'-methoxyethoxy ribitol spacer is
shown here
Figure imgf000065_0001
[00281] In one embodiment, the RNAi agent comprises, in 5' to 3' order: an 18-mer strand, wherein the 3' end of the 18-mer strand terminates in a phosphate and further comprises, in
5' to 3' order: a spacer which is 2'-methoxyethoxy ribitol, a phosphate, and a 3' end cap which is X058. This structure can be on any RNAi strand of any sequence or target. In addition, any 3' end cap disclosed herein can be used in place of X058.
[00282] A related structure is 2'-methoxyethoxy ribitol with X058, wherein the last nucleotide of the 18-mer strand is a 2'-MOE), and the 3' end of the 18-mer strand terminates in a phosphate and further comprises, in 5' to 3' order: a spacer which is 2'-methoxyethoxy ribitol, a second phosphate, and a 3' end cap which is X058.
[00283] Another embodiment is 2'-methoxyethoxy ribitol with C6 cap, wherein the last nucleotide of the 18-mer strand is a 2'-MOE), and the 3' end of the 18-mer strand terminates in a phosphate and further comprises, in 5' to 3' order: a spacer which is 2'-methoxyethoxy ribitol, a phosphate, and a 3' end cap which is C6.
[00284] In one embodiment, the RNAi agent comprises, in 5' to 3' order: an 18-mer strand terminating in a 3' phosphate, a 2'-methoxyethoxy ribitol spacer, a phosphate, and a C6 3' end cap.
[00285] In one embodiment, the RNAi agent comprises, in 5' to 3' order: an 18-mer strand terminating in a 3' phosphate, a 2'-methoxyethoxy ribitol spacer, a phosphate, and a BP 3' end cap.
[00286] In one embodiment, the RNAi agent comprises, in 5' to 3' order: an 18-mer strand terminating in a 3' phosphate, a 2'-methoxyethoxy ribitol spacer, a phosphate, and a C10 3' end cap. [00287] In another embodiment, the RNAi agent comprises a strand, wherein the 3' end of the strand terminates in a phosphate and further comprises, in 5' to 3' order: a spacer which is 2'-methoxyethoxy ribitol, a phosphate, and a 3' end cap which is X058.
[00288] In some embodiments, the 3' end cap is a 2'-methoxyethoxy ribitol. Thus, the RNAi agent comprises an 18-mer strand, wherein the 3' end of the 18-mer strand terminates in a phosphate or modified internucleoside linker and further comprises, in 5' to 3' order: a spacer which is 2'-methoxyethoxy ribitol, a second phosphate or modified internucleoside linker, and a 3' end cap which is a second 2'-methoxyethoxy ribitol. In one embodiment, the 3' end of the 18-mer strand terminates in a phosphate and further comprises, in 5' to 3' order: a spacer which is 2'-methoxyethoxy ribitol, a second phosphate, and a 3' end cap which is a second 2'-methoxyethoxy ribitol.
[00289] In various embodiments, the structure comprises an RNAi agent comprising, in 5' to 3' order, a strand terminating in a 3' terminal phosphate or a modified internucleoside linker, a 2'-methoxyethoxy ribitol spacer, a phosphate or a modified internucleoside linker, and a 3' end cap (e.g., any 3' end cap known in the art) can be used on any RNAi agent of any length, sequence or target, including but not limited to a double-stranded RNA, wherein optionally one or more phosphates are replaced by a modified internucleoside linker, optionally one or more nucleotides are modified, and optionally one or more RNA
nucleotides are replaced by DNA, PNA, LNA, morpholino, TNA, GNA, ANA, HNA, CeNA, FAN A, and/or UNA.
[00290] 2' -deoxyribitol spacer. In some embodiments the spacer is 2'-deoxyribitol.
[00291] In one embodiment, the RNAi agent comprises, in 5' to 3' order: an 18-mer strand terminating in a 3' phosphate or a modified internucleoside linker, a spacer which is 2'- deoxyribitol (2'-deoxyrib), a phosphate or a modified internucleoside linker, and a 3' end cap.
[00292] In one embodiment, the RNAi agent comprises, in 5' to 3' order: an 18-mer strand terminating in a 3' phosphate, a spacer which is 2'-deoxyribitol (2'-deoxyrib), a phosphate or a modified internucleoside linker, an ' end cap. The structure of a 3' terminal phosphate
and a 2'-deoxyribitol is shown here
Figure imgf000066_0001
: 2'-deoxyribitol (2'-deoxyrib).
[00293] In one embodiment, the RNAi agent comprises, in 5' to 3' order: an 18-mer strand terminating in a 3' phosphate, a 2'-deoxyribitol spacer, a phosphate, and a C12 3' end cap. [00294] The structure comprising an RNAi agent comprising, in 5' to 3' order, an 18-mer strand terminating in a 3' terminal phosphate or a modified internucleoside linker, a 2'- deoxyribitol spacer, a phosphate or a modified internucleoside linker, and a 3' end cap can be used on any RNAi agent of any sequence or target, including but not limited to a double- stranded RNA, wherein optionally one or more phosphates are replaced by a modified internucleoside linker, optionally one or more nucleotides are modified, and optionally one or more RNA nucleotides are replaced by DNA, PNA, LNA, morpholino, TNA, GNA, ANA, HNA, CeNA, FANA, and/or UNA.
[00295] C3 spacer. In various embodiments, the spacer is C3.
[00296] In one embodiment, the RNAi agent comprises an 18-mer strand, wherein the 3' end of the 18-mer strand terminates in a 3' phosphate or a modified internucleoside linker, and further comprises a spacer which is C3, a phosphate or a modified internucleoside linker, and a 3' end cap.
[00297] In one embodiment, the RNAi agent comprises two 18-mer strands, wherein the 3' end of each 18-mer strand terminates in a 3' phosphate or a modified internucleoside linker, and further comprises a spacer, a second phosphate or a modified internucleoside linker, and a 3' end cap, wherein the spacer in one or both strands is C3.
[00298] The C3 spacer has the chemical form -(CH2)3-. The structure of a 3' terminal
phosphate and a C3 spacer is shown here:
Figure imgf000067_0001
[00299] One embodiment the RNAi agent comprises, in 5' to 3' order: an 18-mer strand terminating in a 3' phosphate, a C3 spacer, a phosphate, and a 3' end cap which is X058. This structure can be on any RNAi strand of any sequence or target. In addition, any 3' end cap disclosed herein or known in the art can be used in place of X058, and any modified internucleoside linker can be used in place of phosphate.
[00300] In one embodiment a portion of a RNAi agent comprising an 18-mer strand, wherein the 18-mer strand terminates in a phosphate and further comprises in 5' to 3' order: a C3 spacer, a phosphate and a 3' end cap which is C6. This is designated "C3pC6 overhang". This structure can be on any RNAi strand of any sequence or target. In addition, any 3' end cap disclosed herein or known in the art can be used in place of C6, and any modified internucleoside linker can be used in place of phosphate. [00301] The structure comprising an RNAi agent comprising, in 5' to 3' order, an 18-mer strand terminating in a 3' terminal phosphate or a modified internucleoside linker, a C3 spacer, a phosphate or a modified internucleoside linker, and a 3' end cap can be used on any RNAi agent of any sequence or target, including but not limited to a double-stranded RNA, wherein optionally one or more phosphates are replaced by a modified internucleoside linker, optionally one or more nucleotides are modified, and optionally one or more RNA nucleotides are replaced by DNA, PNA, LNA, morpholino, TNA, GNA, ANA, HNA, CeNA, FAN A, and/or UNA.
[00302] In various embodiments, the spacer is C4 or C5 or C6.
[00303] In one embodiment, the APOC3 RNAi agent comprises an 18-mer strand, wherein the 3' end of the 18-mer strand terminates in a 3' phosphate or a modified internucleoside linker, and further comprises a spacer which is C4 or C5 or C6, a phosphate or a modified internucleoside linker, and a 3' end cap.
[00304] In one embodiment, the RNAi agent comprises two 18-mer strands, wherein the 3' end of each 18-mer strand terminates in a 3' phosphate or a modified internucleoside linker, and further comprises a spacer, a second phosphate or a modified internucleoside linker, and a 3' end cap, wherein the spacer in one or both strands is C4 or C5 or C6.
[00305] The C3 to C6 spacers can be defined as: C3 = 1 ,3-propane-diol, C4 = 1 ,4-butane- diol, C5 = 1 ,5-pentane-diol, or C6 = 1 ,6-hexane-diol
[00306] In some contexts: the C4 spacer has the chemical formula -(CH2)4-, the C5 spacer has the chemical formula -(CH2)5-, and the C6 spacer has the chemical formula -(CH2)6-.
[00307] In one embodiment, the structure comprising an RNAi agent comprises in 5' to 3' order, an 18-mer strand terminating in a 3' terminal phosphate or a modified internucleoside linker, a C4 or C5 or C6 spacer, a phosphate or a modified internucleoside linker, and a 3' end cap can be used on any RNAi agent of any sequence or target, including but not limited to a double-stranded RNA, wherein optionally one or more phosphates are replaced by a modified internucleoside linker, optionally one or more nucleotides are modified, and optionally one or more RNA nucleotides are replaced by DNA, PNA, LNA, morpholino, TNA, GNA, ANA, HNA, CeNA, FANA, and/or UNA.
[00308] As a note of clarification, this disclosure notes that the terms "C3" [-(CH2)3-], "C4" [-(CH2)4-], and "C5" [-(CH2)5-] are generally used herein to designate spacers, similar terms (C3, C4, C5 "linkers") are also used to designate a portion of a 3' end cap. It is also noted that the term "C3" is used to designate a C3 3' end cap (see, e.g, U.S. Pat. No. 8,097,716), a C3 spacer, and a C3 linker. The C6 spacer should also be differentiated from the C6 3' end cap. [00309] 4-methoxybutane-1 ,3-diol (5300) spacer. In various embodiments, the spacer is 4-methoxybutane-1 ,3-diol. 4-methoxybutane-1 ,3-diol is also designated 5300, A5300, C5300, G5300, and UG5300.
[00310] In one embodiment, the APOC3 RNAi agent comprises, in 5' to 3' order: an 18-mer strand, wherein the 3' end terminates in a 3' phosphate (a 3' terminal phosphate) or a modified internucleoside linker and further comprises: a spacer which is 4-methoxybutane- 1 ,3-diol, a phosphate or a modified internucleoside linker, and a 3' end cap.
[00311] The structure of a 3' terminal phosphate and a 4-methoxybutane-1 ,3-diol spacer is
Figure imgf000069_0001
shown here:
[00312] In one embodiment, the RNAi agent comprises, in 5' to 3' order: an 18-mer strand terminating in a 3' phosphate, a spacer which is 4-methoxybutane-1 ,3-diol, a phosphate or a modified internucleoside linker, and a 3' end cap.
[00313] In one embodiment the RNAi agent comprises, in 5' to 3' order: an 18-mer strand terminating in a 3' phosphate, a 4-methoxybutane-1 ,3-diol spacer, a phosphate, and a 3' end cap which is X058. This structure can be on any RNAi strand of any sequence or target. In addition, any 3' end cap disclosed herein or known in the art can be used in place of X058, and any modified internucleoside linker can be used in place of phosphate.
[00314] In one embodiment the RNAi agent comprises, in 5' to 3' order, an 18-mer strand terminating in a 3' terminal phosphate or a modified internucleoside linker, a 4- methoxybutane-1 ,3-diol spacer, a phosphate or a modified internucleoside linker, and a 3' end cap can be used on any RNAi agent of any sequence or target, including but not limited to a double-stranded RNA, wherein optionally one or more phosphates are replaced by a modified internucleoside linker, optionally one or more nucleotides are modified, and optionally one or more RNA nucleotides are replaced by DNA, PNA, LNA, morpholino, TNA, GNA, ANA, HNA, CeNA, FANA, and/or UNA.
[00315] Phosphate or modified internucleoside linker. In various embodiments, the modified internucleoside linker is: phosphorothioate, phosphorodithioate, phosphoramidate, boranophosphonoate, an amide linker, or a compound of formula (I). [00316] The disclosure relates to compositions comprising an APOC3 RNAi agent having a novel format. These RNAi agents comprise a sense and an anti-sense strand, each strand being an 18-mer and the strands together forming a blunt-ended duplex, wherein the 3' end of at least one strand terminates in a phosphate or modified internucleoside linker and further comprises, in 5' to 3' order: a spacer; a second phosphate or modified
internucleoside linker; and a 3' end cap. In some embodiments, the 3' end of both the sense and anti-sense strand further comprise, in 5' to 3' order: a spacer; a second phosphate or modified internucleoside linker; and a 3' end cap. The two strands can have the same or different spacers, phosphate or modified internucleoside linker, and/or 3' end caps. In various embodiments, one or more nt can be modified and/or substituted. Various spacers are described below. Various phosphates or modified internucleoside linkers can be used in combination with strands of any sequence, with or without substitutions and/or modifications of nt, with any spacers, and with any 3' end cap, in any combination without limitation.
[00317] In some embodiments, the modified internucleoside linker is interposed between the spacer and the 3' end cap.
[00318] In various embodiments, one or more of the phosphates of one or both strands of the RNAi agent are replaced. Thus: In various embodiments, one or more nucleotide of one or both strands has a modified internucleoside linker. In some embodiments, the 3' terminal phosphate is replaced. In some embodiments, one or more nucleotide of one or both strands has a modified internucleoside linker, and/or a modified internucleoside linker is interposed between the spacer and the 3' end cap.
[00319] In one embodiment, the present disclosure encompasses a RNAi agent comprising a sense and an anti-sense strand, each strand being an 18-mer and the strands together forming a blunt-ended duplex, wherein the 3' end of at least one strand terminates in a phosphate or modified internucleoside linker and further comprises, in 5' to 3' order: a spacer; a second phosphate or modified internucleoside linker; and a 3' end cap, wherein the 3' end cap is selected from the 3' end caps listed in any Table herein or otherwise disclosed herein, and wherein at least one nucleotide has a modified internucleoside linker a modified internucleoside linker (e.g., wherein at least one phosphate of a nucleotide is replaced by a modified internucleoside linker), where the modified internucleoside linker is:
Figure imgf000070_0001
boranophosphonoate, an amide linker, or a compound of formula (I): (I), where
Figure imgf000071_0001
R3 is selected from O", S", NH2, BH3, CH3, Ci_6 alkyl, C6-io aryl, Ci-6 alkoxy and C6-io aryl-oxy, wherein Ci-6 alkyl and C6-io aryl are unsubstituted or optionally independently substituted with 1 to 3 groups independently selected from halo, hydroxyl and NH2; and R4 is selected from O, S, NH, or CH2.
[00320] In one embodiment, the present disclosure encompasses a RNAi agent comprising a sense and an anti-sense strand, each strand being an 18-mer and the strands together forming a blunt-ended duplex, wherein the 3' end of at least one strand terminates in a phosphate or modified internucleoside linker and further comprises, in 5' to 3' order: a spacer; a second phosphate or modified internucleoside linker; and a 3' end cap, wherein the 3' end cap is selected from the 3' end caps listed in any Table herein or otherwise disclosed herein, and wherein at least the 3' terminal nucleotide on one or both strands has a modified internucleoside linker (e.g., wherein the phosphate of the 3' nucleotide on one or both strands is replaced by a modified internucleoside linker), wherein the modified internucleoside linker is phosphorothioate, phosphorodithioate, phosphoramidate, boranophosphonoate, an amide linker, or a compound of formula (I).
[00321] In various embodiments, the 3' end cap is linked via a terminal phosphate group (i.e., a phosphate group at the 3' end of a RNAi agent strand). Such compounds are shown in, for example, Table 5A-E. Alternatively, in 3' to 5' order, a 3' end cap can be bound to a phosphate or modified internucleoside linker, which is bound to a spacer, which is bound to a phosphate or modified internucleoside linker bound to the 3' carbon at the 3' end of at least one RNAi agent strand.
[00322] In one embodiment, compounds of table 5A-E have a terminal phosphorothioate group bound to the 3' carbon at the 3' end of at least one RNAi agent strand. Thus, in various embodiments, in the 3' end caps listed in Table 5A-E, the phosphate group is replaced by a phosphorothioate. In other words, the composition comprises a RNAi agent comprising a strand, wherein the 3' end of the strand terminates in a phosphorothioate and further comprises a compound of Table 5A-E (or any other 3' end cap described herein or known in the art). In additional embodiments, the phosphate group of various 3' end caps listed herein as C3, C6, C12, Triethylene glycol, Cyclohexyl, Phenyl, Biphenyl, Adamantane, Lithocholic acid can be replaced by phosphorothioate. In one embodiment, the phosphate group in the C3 3' end cap is replaced by phosphorothioate. In another embodiment, the phosphate group in the C6 3' end cap is replaced by phosphorothioate. In another embodiment, the phosphate group in the C10 3' end cap is replaced by phosphorothioate. In another embodiment, the phosphate group in the biphenyl (BP) 3' end cap is replaced by phosphorothioate.
[00323] Various additional embodiments of APOC3 RNAi agents in the 18-mer format
[00324] In some embodiments, the APOC3 RNAi agent comprises a 18-mer strand further comprising at the 3' terminus a 3' end cap (but no spacer, or phosphate or internucleoside linker). Thus: In some embodiments, the RNAi agent comprises an 18-mer strand further comprising at the 3' terminus a 3' end cap (e.g., BP or C6).
[00325] In some embodiments, the APOC3 RNAi agent comprises an 18-mer strand further comprising at the 3' terminus a spacer (but no phosphate or internucleoside linker, or 3' end cap). Thus: In some embodiments, the RNAi agent comprises an 18-mer strand further comprising at the 3' terminus a spacer (e.g., ribitol).
[00326] In various embodiments, one or both strands can comprise ribonucleotide subunits, or one or more nucleotide can optionally be modified or substituted. Thus, in various embodiments, the RNAi agent can either contain only naturally-occurring ribonucleotide subunits, or one or more modifications to the sugar, phosphate or base of one or more of nucleotide subunits. In one embodiment, the modifications improve efficacy, stability and/or reduce immunogenicity of the RNAi agent.
[00327] One aspect of the present disclosure relates to an APOC3 RNAi agent comprising at least one non-natural nucleobase. In certain embodiments, the non-natural nucleobase is difluorotolyl, nitroindolyl, nitropyrrolyl, or nitroimidazolyl. In a particular embodiment, the non- natural nucleobase is difluorotolyl. In certain embodiments, only one of the two strands contains a non-natural nucleobase. In certain embodiments, both of the strands contain a non-natural nucleobase.
[00328] In one embodiment, the first two base-pairing nucleotides on the 3' end of the sense and/or anti-sense strand are modified. In one embodiment, the first two base-pairing nucleotides on the 3' end of the sense and/or anti-sense strand are 2'-MOE (a 2' MOE clamp).
[00329] In one embodiment, the 3' terminal phosphate of the sense and/or anti-sense strands is replaced by a modified internucleoside linker.
[00330] In various embodiments, one of more nucleotides is substituted with DNA, a peptide nucleic acid (PNA), locked nucleic acid (LNA), morpholino nucleotide, threose nucleic acid (TNA), glycol nucleic acid (GNA), arabinose nucleic acid (ANA), 2 - fluoroarabinose nucleic acid (FANA), cyclohexene nucleic acid (CeNA), anhydrohexitol nucleic acid (HNA), or unlocked nucleic acid (UNA). In some embodiments, the replacement or substitution of RNA with DNA, or a nucleotide of a different backbone, or PNA, LNA, Morpholino, TNA, GNA, ANA, HNA, CeNA, FANA, and/or UNA can be considered a "modification".
[00331] In various embodiments, at least one nucleotide comprises a modified
internucleoside linker, wherein the modified internucleoside linker is selected from phosphorothioate, phosphorodithioate, phosphoramidate, boranophosphonoate, an amide linker, and a compound of formula (I).
[00332] In various embodiments, optionally the 3' terminal phosphate of the sense and/or anti-sense strands is replaced by a modified internucleoside linker.
[00333] In various embodiments, the RNAi agent can be modified on one or both 5' end. In various embodiments, the sense strand can comprise a 5' end cap which reduces the amount of the RNA interference mediated by this strand.
[00334] In various embodiments, the sense strand comprises a 5' end cap selected: a nucleotide lacking a 5' phosphate or 5'-OH; a nucleotide lacking a 5' phosphate or a 5'-OH and also comprising a 2-OMe or 2'-MOE modification; 5'-deoxy-2'-0-methyl modification; 5'- OME-dT; ddT; and 5'-OTr-dT.
[00335] In various embodiments, the RNAi agent is optionally attached to a ligand. The ligand can be selected to improve one or more characteristic, such as, e.g., stability, distribution and/or cellular uptake of the agent, e.g., cholesterol or a derivative thereof.
[00336] In various embodiments, the RNAi agent can be isolated or be part of a
pharmaceutical composition used for the methods described herein.
[00337] In various embodiments, the pharmaceutical composition can be a lipid
nanoparticle.
[00338] In various embodiments, the pharmaceutical composition can be a lipid
nanoparticle Optionally, the pharmaceutical compositions can further comprise or be used in conjunction with any known treatment for any target gene-related disease.
[00339] The present disclosure further provides methods for reducing the level of target gene mRNA in a cell, particularly in the case of a disease characterized by over-expression or hyper-activity of the target gene product. The present disclosure also encompasses a method of treating a human subject having a pathological state mediated at least in part by target gene expression, the method comprising the step of administering to the subject a therapeutically effective amount of a RNAi agent target gene. Such methods comprise the step of administering one of the RNAi agents of the present disclosure to a subject.
Reduction of target gene mRNA in a cell results in a reduction in the amount of encoded target gene protein produced.
[00340] In another embodiment, the invention provides an RNAi agent with any one or more of the above properties for use as a medicament. [00341] The methods and compositions of the present disclosure, e.g., the methods and target gene RNAi agent compositions, can be used with any dosage and/or formulation described herein, as well as with any route of administration described herein.
[00342] In various embodiments, the RNAi agent can be combined with one or more additional RNAi agents in the same formulation. The one or more additional RNAi agents can have the same or different sequences, targets, spacers, 3' end caps, nucleotide replacements modifications, and/or ligands, etc. In various embodiments, the one or more additional RNAi agents can have a sense and an anti-sense strand wherein each is an 18- mer and together form a blunt-ended duplex. The one or more additional RNAi agent can target the same or different sequence and/or the same or different target gene.
[00343] Thus: Multiple RNAi agents can be administered separately or co-administered. The multiple RNAi agents can be administered in the same delivery vehicle, the same type of delivery vehicle, or in different delivery vehicles.
[00344] Various additional embodiments are described below.
[00345] Any of the various 3' end caps (e.g., ligands or PAZ ligands) can be used with the APOC3 RNAi agents described herein.
[00346] A 3' end cap is a non-nucleotidic chemical moiety bound to the 3' end of a molecule comprising a RNAi agent, e.g., the 3' terminus (or 3' end) of (a) a molecule comprising a strand, wherein the 3' end of the strand terminates in a phosphate or modified
internucleoside linker; or (b) a molecule comprising, in 5' to 3' order: a strand (wherein the 3' end of the strand terminates in a phosphate or modified internucleoside linker), a spacer, and a second phosphate or modified internucleoside linker. The 3' end cap performs at least one of the following functions: allowing RNA interference mediated by the molecule, protecting the molecule from degradation or reducing the amount or rate of degradation of the molecule (e.g., by nucleases), reducing the off-target effects of the sense strand, or increasing the activity, duration or efficacy of RNA interference mediated by the molecule. By describing a 3' end cap as "non-nucleotidic", it is meant that a nucleotide comprises three components: a phosphate, a pentose (e.g., a ribose or deoxyribose) and a nucleobase, and a 3' end cap does not comprise all three components.
[00347] 3' end caps include, but are not limited to compounds designated "PAZ ligands". These ligands can provide increased potency and/or higher siRNA turnover.
[00348] Various PAZ ligands and 3' end caps have been tested with various APOC3 RNAi agents. It is noted that some documents also refer to C6, C8, C10, C12, BP and other 3' end caps as PAZ ligands.
[00349] Human APOC3 RNAi agents were constructed, with the guide strand, with or without (+ or -) the MOE clamp (wherein the first two nt on the 3' end of each strand are 2'- MOE), with or without (+ or -) the ribitol spacer, and with each of the 17 listed PAZ ligands,
C6 and BP as the 3' end caps. The passenger strands were constructed with or without (+ or -) the MOE clamp, with or without (+ or -) the ribitol spacer, and with C6 as the 3' end cap.
[00350] The RNAi agents are tested in a 3 point dose response.
[00351] RNAi agents are introduced into Huh7 cells (10,000) using Optifect.
[00352] Three RNAi agent concentrations are used: 30, 15, 10 and 5 nM.
[00353] A group of siRNA agents are tested.
[00354] 7x96-well plates are transfected in duplicate (with randomized distribution of siRNAs across the plates).
[00355] qRT-PCR is performed and duplicate, and the gene product is normalized to GAPDH mRNA.
[00356] Plate-to-plate normalization is also performed.
[00357] Modification of Human APOC3 RNAi agents is also described herein.
[00358] Once efficacious siRNA sequences are identified, these siRNAs can be optimized by performing various modifications and testing those modifications to find those which improve efficacy, duration and/or stability and/or reduce off-target effects, or otherwise improve the performance of the siRNA.
[00359] The efficacious human APOC3 RNAi agents can be modified by replacing a RNA with 2'-OMe-RNA, DNA, 2 -F-RNA, 2'-MOE-RNA, and/or addition of a spacer (e.g., ribitol), and/or additional of a 3' end cap (X058, C6 or other 3' end cap).
[00360] Human ApoC3 RNAi agents were thus modified in numerous ways and tested for activity.
[00361] Human APOC3 RNAi Agents can also be modified by replacement of RNA with DNA. In particular, nucleotides in the seed region (positions 2-7) can be replaced by DNA. It is noted that various references define the exact limit of the seed region differently; some describe it as nt 4-8, 2-8 or 2-7, counting from the 5' end of the anti-sense strand. In this disclosure, the seed region refers to nt 2-7. RNAi names sometimes have suffices identifying a base as DNA (e.g., "pos4_DNA" indicates that the nucleotide at position 4 is DNA; "pos8_DNA" indicates that that at position 8 is DNA, etc.). These modifications reduce off-target effects by weakening the binding affinity of the guide strand seed region. Off- target effects can be measured, for example, as EC50 in nM in HeLa cells. The data show that DNA is thus well-tolerated, e.g., in the seed region and can reduce the off-target phenotype. In some cases, other positions (e.g., position 2) can be replaced by DNA, or more than one nucleotide can be replaced by DNA (e.g., positions 2 and 4).
[00362] Modifications can be made to increase the serum stability of the molecules.
[00363] In various experiments, off-target effects are measured in HeLa cells (1000 cells, using HiPerFect, 166 hours post-transfection); and potency is measured (e.g., 12 dose- response, time course, in Huh7 cells, using 8000 cells, using OptiFect, at 24, 48, 72, 120 and 166 hours). Potency reporter: human/cyno (6 dose-response): COS1 (5000 cells), Fugene/Oligofectamine, 72 hours; antisense reporter human, sense reporter human, and antisense reporter cyno.
[00364] Modified variants of APOC3 RNAi agents include conjugate formats for 18-mers and 19-mers.
[00365] Modified variants of APOC3 RNAi agents include those with modifications of 5' and/or 3' end of the passenger strand to components which increase targeting to and uptake by the liver; and/or modification of the 5' end of the anti-sense strand to stabilize it; and/or modification of the 3' end of the anti-sense strand, e.g., by conjugation with a 3' end cap, to increase stability and/or potency.
[00366] DELIVERY
[00367] Any pharmaceutical carrier known in the art can be used to deliver APOC3 siRNAs.
[00368] Criteria for an effective delivery vehicle include effective target knockdown in the liver at a reasonable dosage and duration, and acceptable pharmacokinetics and toxicity.
[00369] Delivery vehicles include, for example, lipid nanoparticles (LNPs), non-LNP delivery, peptide conjugation, and lipoprotein particles (LLPs). Particles can be, for example 10-100 nM in size, and can, for example, self-assemble in aqueous media.
[00370] Lipid nanoparticle (LNP)
[00371] For example, APOC3 siRNAs can be delivered using a lipid nanoparticle (LNP). Such an LNP can comprise, for example, a cationic lipid, cholesterol, a neutral lipid, and a shielding lipid.
[00372] Several in vivo studies were done using APOC3 18-mer RNAi agents at 1 mg/kg administered to mice using LNPs. In one study, APOC3 RNAi agents were able to deduce gene expression by over 70% compared to PBS control at 48 hours. Other studies demonstrated the efficacy of APOC3 siRNAs in vivo using a variety of LNPs.
[00373] The use of one LNP and an 18-mer APOC3 RNAi yielded a 60% KD in mice 48 and 96 hrs post IV injection.
[00374] Additional Definitions
[00375] For convenience, the meaning of certain terms and phrases used in the
specification, examples, and appended claims, are provided below. If there is an apparent discrepancy between the usage of a term in other parts of this specification and its definition provided in this section, the definition in this section shall prevail. [00376] As used throughout this disclosure, articles such as "a" and "an" refer to one or more than one (at least one) of the grammatical object of the article.
RNAi agent
[00377] In one aspect, the present disclosure pertains to an APOC3 RNAi agent. An RNAi agent is a composition comprising at least an antisense nucleic acid sequence
complementary to a target nucleic acid (or portion thereof) and capable of mediating RNA interference against the target, or pertains to a recombinant expression vector encoding an shRNA or composition comprising the antisense nucleic acid that can function as an RNAi as defined below; an RNAi agent generally, but not always, also comprises a sense strand at least partially complementary to the anti-sense strand. As used herein, an "antisense" nucleic acid comprises a nucleotide sequence complementary to a "sense" nucleic acid encoding the target protein (e.g., complementary to the coding strand of a double-stranded DNA, complementary to an mRNA or complementary to the coding strand of a target gene or nucleic acid).
[00378] RNAi agents include, as non-limiting examples, siRNAs (small interfering RNAs), dsRNAs (double stranded RNAs), shRNAs (short hairpin RNAs) and miRNAs (micro RNAs). A canonical siRNA comprises two strands, each a 21 -mer, forming a 19-bp double-stranded region and two 3' overhangs of 2 nt each. However, other structures or formats of RNAi agents are known to be active, including blunt-ended 19-mers, or, as disclosed herein, blunt- ended 18-mers, or siRNAs with a shortened sense strand. RNAi agents can comprise RNA subunits (ribonucleotides); however, one or more ribonucleotides can be modified and/or substituted. Modifications and substitutions include, as additional non-limiting examples, locked nucleic acid (LNA), Morpholino, threose nucleic acid (TNA), or glycol nucleic acid (GNA), arabinose nucleic acid (ANA), 2'-fluoroarabinose nucleic acid (FANA), cyclohexene nucleic acid (CeNA), anhydrohexitol nucleic acid (HNA), peptide nucleic acid (PNA), or UNA (unlocked nucleic acid), 2'-MOE, 2'-OMe, 2'-F, 2'-alkyl, or any other modification or substitution described herein or known in the art. RNAi agents also include molecules in which one or more strands are a mixture of RNA, DNA, LNA, morpholino, TNA, GNA, ANA, HNA, CeNA, FANA, UNA, and/or FANA, etc. As a non-limiting example, one or both strands of an RNAi agent could be, for example, RNA, except that one or more RNA nucleotides is replaced by DNA, LNA, morpholino, TNA, GNA, UNA, and/or FANA, etc. In various aspects, one or both strands of the RNAi agent can be nicked, and both strands can be the same length, or one (e.g., the passenger strand), can be shorter than the other.
[00379] In various aspects, the present disclosure pertains to any RNAi agent comprising a RNA sequence disclosed herein and/or a RNA sequence corresponding to any DNA sequence disclosed herein (e.g., wherein the DNA nucleotides are replaced by the corresponding RNA nucleotide, for example, with T in DNA replaced by U in RNA, and with ribose instead of deoxyribose in the sugar-phosphate backbone).
[00380] The RNAi agent(s) of the present disclosure target (e.g., bind to, anneal to, hybridize, etc.) the APOC3 mRNA. The use of the RNAi agent specific to APOC3 results in a decrease of APOC3 activity, level and/or expression, e.g., a "knock-down" (KD) or "knockout" of the target gene or target sequence. In one aspect, in the case of a disease state characterized by over-expression or hyper-activity of APOC3, administration of a RNAi agent to APOC3 knocks down the APOC3 target enough to provide a more normal or therapeutic level of APOC3 activity or expression. Thus, a minimal expression of APOC3 in normal tissues can be beneficial. In various aspects of the disclosure, the patient or individual may have a disease state characterized by excessively high levels of APOC3 and the RNAi agent can restore a normal level. In one aspect of the disclosure, the levels of APOC3 throughout the body are modulated such that APOC3 levels in one area (e.g., areas afflicted by an APOC3-related disease) are lower, while areas of the body not afflicted by the disease are closer to normal APOC3 levels. In one aspect of the disclosure, the RNAi agent can be delivered locally so that levels of APOC3 outside the diseased areas can be maintained as close to normal as possible. In another aspect, the level of APOC3 in the body can be modulated such that it is low enough to improve the disease state, but not so low that organ pathology occurs.
[00381] In one aspect, the RNAi comprises a single strand. This single-stranded RNAi agent oligonucleotide or polynucleotide can comprise the sense or antisense strand, as described by Sioud 2005 J. Mol. Biol. 348:1079-1090, and references therein. Thus the disclosure encompasses RNAi agents with a single strand comprising either the sense or antisense strand of an RNAi agent described herein. The disclosure also encompasses RNAi agents comprising a single strand, wherein the single strand comprises the sequences of both the antisense and sense strands of any RNAi agent disclosed herein, e.g., wherein the strands are contiguous, connected by a loop or otherwise linked. Examples of such molecules include those with a hairpin between the sense and anti-sense sequences (e.g., shRNA).
[00382] In various aspects, one or both strands contain one or more nicks, i.e., a break or missing bond in the phosphate backbone, such that at least one nucleotide subunit is not covalently linked to the adjacent nucleotide subunit in any given sequence. In some aspects, the passenger strand is nicked (see, for example, WO 2007/107162). In various aspects, one or both strands contain one or more gaps, e.g., wherein at least one entire nucleotide subunit is absent from the disclosed sequence. Where a sense or antisense sequence contains a gap, that strand is envisioned to comprise two separate oligonucleotides. [00383] Particularly useful siRNAs include those which can bind specifically to those regions of the APOC3 mRNA that have one or more of the following qualities: binding in the coding segment of APOC3; binding at or near the junction of the 5' untranslated region and the start of the coding segment; binding at or near the translational start site of the mRNA; binding at, across or near junctions of exons and introns; little or no binding to the mRNAs or transcripts of other genes (little or no "off-target effects"); binding to the APOC3 mRNA in or near a region or regions that is not double-stranded or a stem region, e.g., those in a loop or single-stranded portion; eliciting little or no immunogenicity; binding in a segment of the APOC3 mRNA sequence which is conserved among various animal species (including human, mouse, rat, cyno, etc.), as the presence of a conserved sequence facilitates testing using various laboratory animals; binding to double-stranded region(s) of the mRNA; binding to an AT-rich region (e.g., at least about 50, 51 , 52, 53, 54, 55, 56, 57, 58, 59, or 60% AT- rich); and/or lacking particular sequences known or suspected to decrease siRNA activity, e.g., the presence of a GG sequence at the 5' end, which may decrease separation of the double-stranded portion of the siRNA. In one aspect, the RNAi agent specific to APOC3 can be a double-stranded RNA having any one or more of these qualities.
[00384] The term "double-stranded RNA" or "dsRNA," as used herein, refers to a RNAi agent comprising a first and a second strand; e.g., a composition that includes an RNA molecule or complex of molecules having a hybridized duplex region (i.e., a region where the nucleotide bases from the first strand and the second strand are paired) that comprises two anti-parallel and substantially complementary nucleic acid strands, which will be referred to as having "sense" and "antisense" orientations with respect to a target RNA. The antisense strand, with respect to the mRNA target, is also called the "guide" strand, and the sense strand is also called the "passenger" or "anti-guide" strand. The passenger strand can include at least one or more of the following: one or more extra nucleotides (e.g., a bulge or 1 nt loop) compared to the other strand, and/or a nick, a gap, a mismatch, etc., compared to the other strand. In various aspects, the RNAi agent comprises a first strand and a second strand. In various aspects, and as used herein and as is clear by context, terminology referring to the first strand refers to the sense strand and the second strand refers to the anti-sense strand as listed in any Table herein. In other aspects, and as used herein and as is clear by context, the first strand refers to the anti-sense strand, and the second strand refers to the sense strand as listed in any Table herein.
[00385] The duplex region can be of any length that permits specific degradation of a desired target RNA through a RISC pathway, but will typically range from 9 to 36 base pairs ("bp") in length, e.g., 15-30 bp in length. Considering a duplex between 9 and 36 bp, the duplex can be any length in this range, for example, 9, 10, 1 1 , 12, 13, 14, 15, 16, 17, 18, 19, 20, 21 , 22, 23, 24, 25, 26, 27, 28, 29, 30, 31 , 32, 33, 34, 35, or 36 bp and any sub-range therebetween, including, but not limited to 15-30 bp, 15-26 bp, 15-23 bp, 15-22 bp, 15-21 bp, 15-20 bp, 15-19 bp, 15-18 bp, 15-17 bp, 18-30 bp, 18-26 bp, 18-23 bp, 18-22 bp, 18-21 bp, 18-20 bp, 19-30 bp, 19-26 bp, 19-23 bp, 19-22 bp, 19-21 bp, 19-20 bp, 19 bp, 20-30 bp, 20- 26 bp, 20-25 bp, 20-24 bp, 20-23 bp, 20-22 bp, 20-21 bp, 20 basepairs, 21-30 bp, 21-26 bp, 21 -25 bp, 21-24 bp, 21 -23 bp, 21-22 bp, 21 bp, 22 bp, or 23 bp. The dsRNAs generated in the cell by processing with Dicer and similar enzymes are generally in the range of about 19 to about 22 bp in length, though the strands of artificial dsRNAs can be shorter or longer. siRNAs wherein one or both strands are as short as 16 or 15 nt still demonstrate RNA interference activity. Chu and Rana 2008 RNA 14: 1714-1719. One strand of the duplex region of a dsRNA comprises a sequence that is substantially complementary to a region of a target RNA. The two strands forming the duplex structure can be from a single RNA molecule having at least one self-complementary duplex region, or can be formed from two or more separate RNA molecules that hybridize to form the duplex. Where the duplex region is formed from two self-complementary regions of a single molecule, the molecule can have a duplex region separated by a single-stranded chain of nucleotides (herein referred to as a "hairpin loop", e.g., such as found in an shRNA construct) between the 3'-end of one strand and the 5'-end of the respective other strand forming the duplex structure. The hairpin loop can comprise at least one unpaired nucleotide; in some aspects the hairpin loop can comprise at least 3, at least 4, at least 5, at least 6, at least 7, at least 8, at least 9, at least 10, at least 20, at least 23 or more unpaired nucleotides. Where the two substantially complementary strands of a dsRNA are comprised by separate RNA molecules, those molecules need not, but can, be covalently connected. Where the two strands are connected covalently by a hairpin loop, the construct is generally referred to herein and in the art as a "shRNA". Where the two strands are connected covalently by means other than a hairpin loop, the connecting structure is referred to as a "linker."
[00386] RNAi Agents to APOC3 comprising mismatches from the disclosed
sequences. Various specific aspects of a RNAi agent to APOC3 are disclosed herein;
example sequences are provided in the Tables. Specific aspects of the present disclosure include RNAi agents which comprise sequences differing by 0, 1 , 2, or 3 nt (nucleotides) or bp [basepair(s)] (e.g., with 0, 1 , 2 or 3 mismatches) from any of the RNAi agents listed in any of the tables herein, and modified and unmodified variants thereof.
[00387] A mismatch is defined herein as a difference between the base sequence or length when two sequences are maximally aligned and compared. A mismatch is defined as a position wherein the base of one sequence does not match the base of the other sequence. Thus, a mismatch is counted, for example, if a position in one sequence has a particular base (e.g., A), and the corresponding position on the other sequence has a different base (e.g., G). Substitution of A, for example, with T, C, G or U would constitute a mismatch. Substitution of G with T, A, C or U would also constitute a mismatch. Substitution of C with T, G, A or U would also constitute a mismatch. Substitution of U with A, C or G would constitute a mismatch. Note, however, that on a given strand, a U can be replaced by T (either as RNA or, preferably, DNA, e.g., 2'-deoxy-thymidine); the replacement of a U with a T is not a mismatch as used herein, as either U or T can pair with A on the opposite strand. The RNAi agent can thus comprise one or more DNA bases, e.g., T. In some cases, in a portion or portions of the RNAi agent, DNA can be used in place of RNA (e.g., in the seed region), to form a DNA-RNA hybrid. See, for example, Yamato et al. 201 1 cancer Gene Ther. 18: 587-597. No mismatch is counted between a DNA portion(s) of the RNAi agent and the corresponding target mRNA if basepairing occurs (e.g., between A, G, C, or T in the DNA portion, and the corresponding U, C, G, or A, respectively in the mRNA).
[00388] A mismatch is also counted, e.g., if a position in one sequence has a base (e.g., A), and the corresponding position on the other sequence has no base (e.g., that position is a spacer [e.g., in the internal spacer format described herein] or an abasic nucleotide, which comprises a phosphate-sugar backbone but no base). For example, if the anti-sense strands of two different duplexes are identical except that, at one position, the nucleobase unit (i.e., sugar + base) has been replaced by a spacer (e.g., a spacer described herein), the sequences are considered to have one mismatch (e.g., to differ by 1 nt). A single-stranded nick in either sequence (or in the sense or anti-sense strand) is not counted as mismatch. Thus, as a non-limiting example, no mismatch would be counted if one sequence comprises the sequence AG, but the other sequence comprises the sequence AG with a single- stranded nick between the A and the G. A nucleotide modification in the sugar or phosphate is also not considered a mismatch. Thus, if one sequence comprises a C, and the other sequence comprises a modified C (e.g., 2'-modification) at the same position, no mismatch would be counted.
[00389] Thus, no mismatches are counted if modifications are made to the sugar, phosphate, or backbone of the RNAi agent without modifying the base. Thus, a strand having a particular sequence of as an RNA would have zero mismatches from another strand having the same sequence as a PNA; or morpholino; or LNA; or TNA; or GNA; or UNA; or FANA; or a mix or chimera of RNA and DNA, TNA, GNA, UNA, FANA, morpholino, LNA, and/or PNA, etc.
[00390] It is also noted that the sequences of the RNAi agents in the Tables include sequences which comprise modifications. It is noted that dTdT (2'-deoxy-thymidine-5'- phosphate and 2'-deoxy-thymidine-5'-phosphate), or in some cases, TT or UU, can be added as a terminal dinucleotide cap or extension to one or both 3'-ends, but this cap or extension is not included in the calculation of the total number of mismatches and is not considered part of the target sequence. This is because the terminal dinucleotide protects the ends from nuclease degradation but does not contribute to target specificity (Elbashir et al. 2001 Nature 41 1 : 494-498; Elbashir et al. 2001 EMBO J. 20: 6877-6888; and Kraynack et al. 2006 RNA 12:163-176).
[00391] In addition, a modified variant can have one or more modifications from the corresponding unmodified sequence. In this case, lowercase "c" represents 2'-0- methylcytidine-5'-phosphate, and lowercase "u" represents 2'-0-methyluridine-5'-phosphate. Uppercase "A", "C", "G" and "U" represent the un-modified adenosine-5'-phosphate, cytidine- 5'-phosphate, guanosine-5'-phosphate, and uridine-5'-phosphate, respectively. The substitution, for example, of modified c for unmodified C does not count as a mismatch in numbering the 0, 1 , 2, or 3 mismatches between sequences. This nomenclature is used for all sequences in any of the tables herein. Thus, an equal number of mismatches would be calculated (a) between a test sequence and that of another RNAi agent, and (b) between the same test sequence and the corresponding unmodified sequence from the APOC3 gene, and (c) between a modified sequence and a differently modified sequence which have the same base sequence.
[00392] In one particular aspect, the present disclosure comprises a RNAi agent comprising a anti-sense strand comprising at least 15 to 19 contiguous nucleotides differing by 0, 1 , 2, or 3 nt from the sequence of the anti-sense strand of: any of the RNAi agents listed in any of the tables herein, and modified and unmodified variants thereof.
[00393] The present disclosure pertains to "modified and unmodified variants" of the disclosed sequences.
[00394] An "unmodified variant" of a particular sequence is the corresponding portion of APOC3 without any modifications. Example modified sequences are disclosed herein The "unmodified variants" of the sequences of the Tables have the identical sequence, without base modifications or terminal dTdT. A given sequence and an "unmodified variant" of it differ by 0 nt (and have no mismatches).
[00395] A "modified variant" of a particular sequence comprises one or more (or one or more fewer) modifications to the backbone, sugar, phosphate or base, and/or addition of a terminal dinucleotide (e.g., TT, dTdT, TsT or UU), but do not have any base substitutions (e.g., G for C, or A for G); thus a given sequence and a modified variant thereof differ by 0 nt (and have no mismatches). As another example, a given sequence as a RNA and the same sequence as a PNA are modified variants of each other and differ by 0 nt (and have no mismatches). Similarly, the same sequence (with no base substitutions) as a locked nucleic acid (LNA), Morpholino, threose nucleic acid (TNA), or glycol nucleic acid (GNA) or unlocked nucleic acid (UNA) or FANA or ANA or HNA or CeNA, etc. would be a modified variant which has 0 mismatches. In addition, the same sequence could be used in strands which are a mixture of RNA, DNA, LNA, morpholino, TNA, GNA, ANA, HNA, CeNA, UNA, and/or FANA, etc. As a non-limiting example, one or both strands could be, for example, RNA except that one or more nucleotides is replaced by DNA, LNA, morpholino, TNA, GNA, ANA, HNA, CeNA, UNA, and/or FANA, etc.
[00396] As detailed below, substituting a single nucleotide at a given position with a modified version of the same nucleotide would produce a modified variant (with 0
mismatches).
[00397] In another particular aspect, the RNAi agent comprises a sense strand comprising at least 14 contiguous nucleotides differing by 0, 1 , 2, or 3 nt from the sense strand of any of the RNAi agents listed in the Tables and modified and unmodified variants thereof.
[00398] RNAi agents to APOC3 of the present disclosure can be used in RNA interference.
[00399] Modifications of RNAi Agents. The present disclosure encompasses both unmodified and example modified RNAi agents, such as those disclosed in the Tables.
[00400] The present disclosure further encompasses any other modification of a disclosed RNAi agent (e.g., a modified variant).
[00401] For example, the disclosure encompasses a RNAi agent with a substitution of a single nucleotide at a given position with a modified version of the same nucleotide. Thus a nucleotide (A, G, C or U) can be replaced by the corresponding 5-fluorouracil, 5-bromouracil, 5-chlorouracil, 5-iodouracil, hypoxanthine, xantine, 4-acetylcytosine, 5- (carboxyhydroxylmethyl) uracil, 5-carboxymethylaminomethyl-2-thiouridine, 5- carboxymethylaminomethyluracil, dihydrouracil, beta-D-galactosylqueosine, inosine, N6- isopentenyladenine, 1 -methylguanine, 1 -methylinosine, 2,2-dimethylguanine, 2- methyladenine, 2-methylguanine, 3-methylcytosine, 5-methylcytosine, N6-adenine, 7- methylguanine, 5-methylaminomethyluracil, 5-methoxyaminomethyl-2-thiouracil, beta-D- mannosylqueosine, 5'-methoxycarboxymethyluracil, 5-methoxyuracil, 2-methylthio-N6- isopentenyladenine, uracil-5-oxyacetic acid (v), wybutoxosine, pseudouracil, queosine, 2- thiocytosine, 5-methyl-2-thiouracil, 2-thiouracil, 4-thiouracil, 5-methyluracil, uracil-5- oxyacetic acid methylester, uracil-5-oxyacetic acid (v), 5-methyl-2-thiouracil, 3-(3-amino-3-N- 2-carboxypropyl) uracil, (acp3) w, or 2,6-diaminopurine.
[00402] Additional modified variants include the addition of any other moiety (e.g., a radiolabel or other tag or conjugate) to the RNAi agent; provided that the base sequence is identical, the addition of other moieties produces a "modified variant" (with no mismatches).
[00403] Various sets of modifications can be used. These include the following formats, which are used in various screens disclosed herein.
[00404] In addition to these modifications and patterns (e.g., formats) for modifications, other modifications or sets of modifications of the sequences provided can be generated using common knowledge of nucleic acid modification. These various aspects of the RNAi agents to APOC3 of the present disclosure can be used in RNA interference.
[00405] RNA Interference. RNA interference (RNAi) is a post-transcriptional, targeted gene-silencing technique that uses double-stranded RNA (dsRNA) to degrade messenger RNA (mRNA) containing the same sequence as the dsRNA. The process of RNAi occurs when ribonuclease III (Dicer) cleaves the longer dsRNA into shorter fragments called siRNAs. siRNAs (small interfering RNAs) produced by Dicer are typically about 21 to 23 nucleotides long and comprise about 19 base pair duplexes (though artificial siRNAs or RNAi agents can be shorter or longer, and/or blunt-ended, and/or comprises one or more endcaps). The smaller RNA segments then mediate the degradation of the target mRNA. Dicer has also been implicated in the excision of 21 - and 22-nucleotide small temporal RNAs (stRNAs) from precursor RNA of conserved structure that are implicated in translational control. Hutvagner et al. 2001 Science 293: 834. The RNAi response also features an endonuclease complex, commonly referred to as an RNA-induced silencing complex (RISC), which mediates cleavage of single-stranded mRNA complementary to the anti-sense strand of the RNAi agent. Cleavage of the target RNA takes place in the middle of the region complementary to the anti-sense strand of the siRNA duplex.
[00406] In one aspect, an RNA interference agent includes a single-stranded RNA that interacts with a target RNA sequence to direct the cleavage of the target RNA. Without wishing to be bound by theory, the present disclosure contemplates a long double-stranded RNA introduced into plants and invertebrate cells is broken down into siRNA by a Type III endonuclease known as Dicer. Sharp et al. 2001 Genes Dev. 15:485. Dicer, a ribonuclease- lll-like enzyme, processes the dsRNA into 19-23 base pair short interfering RNAs with characteristic two base 3' overhangs. Bernstein, et al. 2001 Nature 409:363. The siRNAs are then incorporated into an RNA-induced silencing complex (RISC) where one or more helicases unwind the siRNA duplex, enabling one of the now unpaired siRNA strands to act as a "guide" strand to guide target recognition. Nykanen, et al. 2001 Cell 107:309. Upon binding of the antisense guide strand to the appropriate target mRNA, one or more endonucleases within the RISC cleaves the target to induce silencing. Elbashir, et al. 2001 Genes Dev. 15:188. Thus, in one aspect the present disclosure relates to a single-stranded RNA that promotes the formation of a RISC complex to effect silencing of the target gene.
[00407] A suitable RNAi agent can be selected by any process known in the art or conceivable by one of ordinary skill in the art. For example, the selection criteria can include one or more of the following steps: initial analysis of the APOC3 gene sequence and design of RNAi agents; this design can take into consideration sequence similarity across species (human, cynomolgus, mouse, etc.) and dissimilarity to other (non-APOC3) genes; screening of RNAi agents in vitro (e.g., at 10 nM in RKO cells); determination of EC50 in RKO cells; determination of viability of cells treated with RNAi agents, including insensitive cells which do not require APOC3 for survival, or sensitive cells, which do require APOC3 for survival; testing with human PBMC (peripheral blood mononuclear cells), e.g., to test levels of TNF- alpha to estimate immunogenicity, wherein immunostimulatory sequences are less desired; testing in human whole blood assay, wherein fresh human blood is treated with an RNAi agent and cytokine/chemokine levels are determined [e.g., TNF-alpha (tumor necrosis factor-alpha) and/or MCP1 (monocyte chemotactic protein 1 )], wherein Immunostimulatory sequences are less desired; determination of gene knockdown in vivo using subcutaneous tumors in test animals; APOC3 target gene modulation analysis, e.g., using a
pharmacodynamic (PD) marker, for example, other factors whose expression is affected by APOC3, wherein APOC3 knockdown leads to a dose-dependent reduction of abundance of those components; and optimization of specific modifications of the RNAi agents.
[00408] The dsRNA molecules (RNAi agents) described herein are thus useful in RNA interference of APOC3.
[00409] Features of a RNAi Agent: Sense strand, Antisense Strand and (Optional) Overhangs. In various aspects, the RNAi agents comprise a first strand and a second strand, e.g., a sense strand and an antisense strand (or an antisense and a sense strand), optionally, either or both both ends of either or both strand can comprise unpaired nucleotides (referred to herein as "overhangs").
[00410] The term "antisense strand" refers to the strand of a RNAi agent which includes a region that is substantially complementary to a target sequence. As used herein, the term "region of complementarity" refers to the region on the antisense strand that is substantially complementary to a sequence, for example a target sequence, as defined herein. Where the region of complementarity is not fully complementary to the target sequence, the
mismatches may be in the internal or terminal regions of the molecule. Generally, the most tolerated mismatches are in the terminal regions, e.g., within 5, 4, 3, or 2 nucleotides of the 5' and/or 3' terminus.
[00411] The term "sense strand," as used herein, refers to the strand of a RNAi agent that includes a region that is substantially complementary to a region of the antisense strand as that term is defined herein.
[00412] The sequence of a gene may vary from individual to individual, especially at wobble positions within the coding segment, or in the untranslated region; individuals may also differ from each other in coding sequence, resulting in additional differences in mRNA. The sequence of the sense and antisense strands of the RNAi agent can thus be designed to correspond to that of an individual patient, if and where needed. RNAi agents can also be modified in sequence to reduce immunogenicity, binding to undesired mRNAs (e.g., "off- target effects") or to increase stability in the blood. These sequence variants are
independent of chemical modification of the bases or 5' or 3' or other end-caps of the RNAi agents.
[00413] The RNAi agents can also have overhangs of 0, 1 , or 2 overhangs; in the case of a 0 nt overhang, they are blunt-ended. A RNAi agent can thus have 0, 1 or 2 blunt ends. In a "blunt-ended RNAi agent" both strands terminate in a base-pair; thus a blunt-ended molecule lacks either 3' or 5' single-stranded nucleotide overhangs.
[00414] The RNAi agents can comprise overhang(s), blunt end(s), and/or 5' and 3' endcap(s).
[00415] As used herein, the term "overhang" or "nucleotide overhang" refer to at least one unpaired nucleotide that protrudes from the end of at least one of the two strands of the duplex structure of a RNAi agent. For example, when a 3'-end of one strand of a dsRNA extends beyond the 5'-end of the other strand, or vice versa, this forms a nucleotidic overhang, e.g., the unpaired nucleotide(s) form the overhang. A dsRNA can comprise an overhang of at least one nucleotide; alternatively the overhang can comprise at least two nucleotides, at least three nucleotides, at least four nucleotides, at least five nucleotides or more. An overhang can comprise or consist of a nucleotide/nucleoside analog, including a deoxynucleotide/nucleoside. The overhang(s) may be on the sense strand, the antisense strand or any combination thereof. Furthermore, the nucleotide(s) of an overhang can be present on the 5' end, 3' end or both ends of either an antisense or sense strand of a dsRNA.The RNAi agent can also optionally comprise a cap. The term "Cap" and the like include a chemical moiety attached to the end of a double-stranded nucleotide duplex, but is used herein to exclude a chemical moiety that is a nucleotide or nucleoside. A "3' Cap" is attached at the 3' end of a nucleotide or oligonucleotide and protects the molecule from degradation, e.g., from nucleases, such as those in blood serum or intestinal fluid. A non- nucleotidic 3' cap is not a nucleotide and can replace a TT or UU dinucleotide at the end of a blunt-ended RNAi agent. In one aspect, non-nucleotidic 3' end caps are as disclosed in, for example, WO 2005/021749 and WO 2007/128477; and U.S. Pat. No. 8,097,716; U.S. Pat. No. 8,084,600; and U.S. Pat. No. 8,344,128. A "5' cap" is attached at the 5' end of a nucleotide or oligonucleotide. A cap should not interfere (or unduly interfere) with RNAi activity.
[00416] The present disclosure thus contemplates a RNAi agent specific to APOC3 comprising an antisense strand (which may be contiguous or connected via a linker or loop) in a RNAi agent. In a more specific aspect, an RNAi agent comprises an antisense strand and a sense strand which together comprise a double-stranded or complementary region. In one aspect, it can also optionally comprise one or two overhangs and/or one or two caps. The RNAi agent is used to induce RNA interference of the target gene, APOC3.
[00417] Target and complementary sequences
[00418] The RNAi agents of the present disclosure target (e.g., specifically bind to, anneal to, etc.) the mRNA encoding APOC3. The use of the RNAi agent specific to APOC3 results in a decrease of APOC3 activity, level and/or expression. Particularly in one aspect, in the case of a disease state characterized by over-expression or hyper-activity of APOC3, administration of a RNAi agent to APOC3 knocks down the APOC3 gene enough to restore a normal level of APOC3 activity or expression.
[00419] In one aspect, the first or second strand of the RNAi comprises a sequence complementary to that of the target nucleic acid, APOC3.
[00420] As used herein, the term "strand comprising a sequence" refers to an
oligonucleotide comprising a chain of nucleotides that is described by the sequence referred to using the standard nucleotide nomenclature.
[00421] As used herein, "target sequence" or "target gene" refer to a contiguous portion of the nucleotide sequence of an mRNA molecule formed during the transcription of a gene, e.g., an APOC3 gene, including mRNA that is a product of RNA processing of a primary transcription product. The target portion of the sequence will be at least long enough to serve as a substrate for RNAi-directed cleavage at or near that portion. For example, the target sequence will generally be from 9-36 nucleotides ("nt") in length, e.g., 15-30 nt in length, including all sub-ranges therebetween. As non-limiting examples, the target sequence can be from 15-30 nt, 15-26 nt, 15-23 nt, 15-22 nt, 15-21 nt, 15-20 nt, 15-19 nt, 15-18 nt, 15-17 nt, 18-30 nt, 18-26 nt, 18-23 nt, 18-22 nt, 18-21 nt, 18-20 nt, 19-30 nt, 19-26 nt, 19-23 nt, 19-22 nt, 19-21 nt, 19-20 nt, 19 nt, 20-30 nt, 20-26 nt, 20-25 nt, 20-24 nt, 20-23 nt, 20-22 nt, 20-21 nt, 20 nt, 21-30 nt, 21 -26 nt, 21-25 nt, 21 -24 nt, 21-23 nt, or 21 -22 nt, 21 nt, 22 nt, or 23 nt. The sense and antisense strands of the RNAi comprise a sequence complementary to that of the target nucleic acid, APOC3.
[00422] As used herein, and unless otherwise indicated, the term "complementary" refers to the ability of an oligonucleotide or polynucleotide comprising a first nucleotide sequence to hybridize and form a duplex structure under certain conditions with an oligonucleotide or polynucleotide comprising a second nucleotide sequence. Such conditions can, for example, be stringent, e.g., 400 mM NaCI, 40 mM PIPES pH 6.4, 1 mM EDTA, 50°C or 70°C for 12-16 hours followed by washing. Other conditions, such as physiologically relevant conditions as may be encountered inside an organism, can apply. The skilled person will be able to determine the set of conditions most appropriate for a test of complementarity of two sequences in accordance with the ultimate application of the hybridized nucleotides.
[00423] Complementary sequences within a RNAi agent, e.g., within a dsRNA as described herein, include base-paired oligonucleotides or polynucleotides comprising a first nucleotide sequence to an oligonucleotide or polynucleotide comprising a second nucleotide sequence over the entire length of one or both nucleotide sequences. Such sequences can be referred to as "fully complementary" with respect to each other herein. However, where a first sequence is referred to as "substantially complementary" with respect to a second sequence herein, the two sequences can be fully complementary, or they may form one or more, but generally not more than 5, 4, 2 or 2 mismatched base pairs upon hybridization for a duplex up to 30 base pairs, while retaining the ability to hybridize under the conditions most relevant to their ultimate application, e.g., inhibition of gene expression via a RISC pathway.
However, where two oligonucleotides are designed to form, upon hybridization, one or more single-stranded overhangs, such overhangs shall not be regarded as mismatches with regard to the determination of complementarity. For example, a dsRNA comprising one oligonucleotide 21 nucleotides in length and another oligonucleotide 23 nucleotides in length, wherein the longer oligonucleotide comprises a sequence of 21 nucleotides that is fully complementary to the shorter oligonucleotide, may yet be referred to as "fully complementary" for the purposes described herein. The term "overhang" describes an unpaired nucleotide at the 3' or 5' end of a double-stranded nucleotide duplex, as described above. In one aspect, the overhang is 1 to 4 nt long and is on the 3' end.
[00424] "Complementary" sequences, as used herein, may also include, or be formed entirely from, non-Watson-Crick base pairs and/or base pairs formed from non-natural and modified nucleotides, in as far as the above requirements with respect to their ability to hybridize are fulfilled. Such non-Watson-Crick base pairs includes, but are not limited to, Wobble or Hoogstein base pairing. The terms "complementary," "fully complementary" and "substantially complementary" herein may furthermore be used with respect to the base matching between the sense strand and the antisense strand of a dsRNA, or between the antisense strand of a RNAi agent and a target sequence, as will be understood from the context of their use. As used herein, a polynucleotide that is "substantially complementary to at least part of a messenger RNA (mRNA) refers to a polynucleotide that is substantially complementary to a contiguous portion of the mRNA of interest (e.g., an mRNA encoding APOC3). For example, a polynucleotide is complementary to at least a part of an APOC3 mRNA if the sequence is substantially complementary to a non-interrupted portion of an mRNA encoding APOC3.
[00425] Thus, the RNAi agent of the present disclosure is complimentary or substantially complimentary to a target sequence in the target APOC3 and is double-stranded, comprising a sense and an antisense strand (which can be contiguous, linked via a loop, or otherwise joined), where the double-stranded region an be 9 to 36 bp long (particularly for example, 19-22 bp or 19-23 bp long), and can furthermore optionally comprise a 3' or 5' overhang, and the RNAi agent can furthermore comprise a 3' cap. The RNAi agent mediates RNA interference, down-regulating or inhibiting or reducing the level, expression and/or activity of APOC3, and/or establishing or re-establishing an approximately normal level of APOC3 and/or APOC3 activity, or other biological function related to APOC3.
[00426] Thus, the RNAi agent of the present disclosure is complimentary or substantially complimentary to a target sequence in the target APOC3 and is double-stranded, comprising a sense and an antisense strand (which can be contiguous, linked via a loop, or otherwise joined), where the double-stranded region an be 9 to 36 bp long (particularly for example, 19-22 bp or 19-23 bp long), and can furthermore optionally comprise a 3' or 5' overhang, and the RNAi agent can furthermore comprise a 3' cap. The RNAi agent mediates RNA interference, down-regulating or inhibiting the level, expression and/or activity of APOC3, and/or establishing or re-establishing an approximately normal level of APOC3 activity or expression.
[00427] The term "double-stranded RNA" or "dsRNA," as used herein, refers to an RNAi agent comprising a first and a second strand; e.g., a composition that includes an RNA molecule or complex of molecules having a hybridized duplex region that comprises two anti-parallel and substantially complementary nucleic acid strands, which will be referred to as having "sense" and "antisense" orientations with respect to a target RNA. The antisense strand, with respect to the mRNA target, is also called the "guide" strand, and the sense strand is also called the "passenger" strand. As used herein, depending on the context, the "first" strand can be the guide or antisense strand, and the "second" strand can be the passenger or sense strand. Also as used herein, again depending on the context, the "first" strand can be the passenger or sense strand, and the "second" strand can be the guide or antisense. The passenger strand can include at least one or more of the following: one or more extra nucleotides (e.g., a bulge or 1 nt loop) compared to the other strand, a nick, a gap, etc., compared to the other strand. In various aspects, the first strand is the sense strand and the second strand is the anti-sense strand. In other aspects, the first strand is the anti-sense strand, and the second strand is the sense strand.
[00428] The duplex region can be of any length that permits loading into the RISC complex and subsequent specific degradation of a desired target RNA through a RISC pathway, but will typically range from 9 to 36 base pairs ("bp") in length, e.g., 15-30 base pairs in length. Considering a duplex between 9 and 36 base pairs, the duplex can be any length in this range, for example, 9, 10, 1 1 , 12, 13, 14, 15, 16, 17, 18, 19, 20, 21 , 22, 23, 24, 25, 26, 27, 28, 29, 30, 31 , 32, 33, 34, 35, or 36 bp and any sub-range therebetween, including, but not limited to 15-30 base pairs, 15-26 bp, 15-23 bp, 15-22 bp, 15-21 bp, 15-20 bp, 15-19 bp, 15- 18 bp, 15-17 bp, 18-30 bp, 18-26 bp, 18-23 bp, 18-22 bp, 18-21 bp, 18-20 bp, 19-30 bp, 19- 26 bp, 19-23 bp, 19-22 bp, 19-21 bp, 19-20 bp, 19 bp, 20-30 bp, 20-26 bp, 20-25 bp, 20-24 bp, 20-23 bp, 20-22 bp, 20-21 bp, 20 bp, 21-30 bp, 21-26 bp, 21 -25 bp, 21 -24 bp, 21-23 bp, 21 -22 bp, 21 bp, 22 bp, or 23 bp.
[00429] The dsRNAs generated in the cell by processing with Dicer and similar enzymes are generally in the range of about 19 to about 22 base pairs in length, although artificial RNAi agents can be synethesized or made by any method known in the art. One strand of the duplex region of a dsRNA comprises a sequence that is substantially complementary to a region of a target RNA. The two strands forming the duplex structure can be from a single RNA molecule having at least one self-complementary duplex region, or can be formed from two or more separate RNA molecules that hybridize to form the duplex. Where the duplex region is formed from two self-complementary regions of a single molecule, the molecule can have a duplex region separated by a single stranded chain of nucleotides (herein referred to as a "hairpin loop", e.g., such as found in an shRNA construct) between the 3'- end of one strand and the 5'-end of the respective other strand forming the duplex structure. The hairpin loop can comprise at least one unpaired nucleotide; in some aspects the hairpin loop can comprise at least 3, at least 4, at least 5, at least 6, at least 7, at least 8, at least 9, at least 10, at least 20, at least 23 or more unpaired nucleotides. Where the two substantially complementary strands of a dsRNA are comprised by separate RNA molecules, those molecules need not, but can be covalently connected. Where the two strands are connected covalently by a hairpin loop, the construct is generally referred to herein and in the art as a "shRNA". Where the two strands are connected covalently by means other than a hairpin loop, the connecting structure is referred to as a "linker." The term "siRNA" is also used herein to refer to a dsRNA as described above.
[00430] RNAi Agents Lowering or Normalizing APOC3 Level, Expression And/Or Activity
[00431] RNAi agents for targeting APOC3 include those which bind to an APOC3 sequence provided herein and which work to reduce APOC3 through a RNAi mechanism. Example RNAi agents (e.g., siRNAs) to APOC3 are provided herein.
[00432] Any method known in the art can be use to measure changes in APOC3 activity, level, and/or expression induced by an APOC3 RNAi agent. Measurements can be performed at multiple timepoints, prior to, during and after administration of the RNAi agent, to determine the effect of the RNAi agent. [00433] The RNAi agents of the present disclosure silence, inhibit the expression of, down- regulate the expression of, and/or suppress the expression of APOC3, such that an approximately normal level of APOC3 activity or expression is restored.
[00434] In addition, in various aspects, depending on the disease condition and biological context, it is acceptable to use the RNAi agents of the present disclosure to establish a level of APOC3 expression, activity and/or level which is below the normal level, or above the normal level, depending on the therapeutic outcome that is desired.
[00435] Any method known in the art can be use to measure changes in APOC3 activity, level and/or expression induced by an APOC3 siRNA. Measurements can be performed at multiple timepoints, prior to, during and after administration of the siRNA, to determine the effect of the siRNA.
[00436] The terms "silence," "inhibit the expression of," "down-regulate the expression of," "suppress the expression of," and the like, in so far as they refer to an APOC3 gene, herein refer to the at least partial suppression of the expression of an APOC3 gene, as manifested by a reduction of the amount of APOC3 mRNA which may be isolated from or detected in a first cell or group of cells in which an APOC3 gene is transcribed and which has or have been treated such that the expression of an APOC3 gene is inhibited, as compared to a second cell or group of cells substantially identical to the first cell or group of cells but which has or have not been so treated (control cells). The degree of inhibition is usually expressed in terms of
(mRNA in control cells) - (mRNA in treated cells) ^ ^
(mRNA in control cells) Equation 1
[00437] Alternatively, the degree of inhibition may be given in terms of a reduction of a parameter that is functionally linked to APOC3 gene expression, e.g., the amount of protein encoded by an APOC3 gene, alteration in expression of a protein whose expression is dependent on APOC3, etc. In principle, APOC3 gene silencing may be determined in any cell expressing APOC3, either constitutively or by genomic engineering, and by any appropriate assay. However, when a reference or control is needed in order to determine whether a given RNAi agent inhibits the expression of APOC3 by a certain degree and therefore is encompassed by the instant disclosure, the assays provided in the Examples below shall serve as such reference.
[00438] For example, in certain instances, expression of APOC3 is suppressed by at least about 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, or 50% by administration of a RNAi agent featured in the present disclosure. In some aspects, APOC3 is suppressed by at least about 60%, 70%, or 80% by administration of a RNAi agent featured in the present disclosure. In some aspects, APOC3js suppressed by at least about 85%, 90%, or 95% or more by administration of a RNAi agent, as described herein. In one aspect, the degree of APOC3 suppression is determined by loss of full length APOC3 mRNA in a treated cell compared to an untreated cell. In one aspect, the degree of APOC3 suppression is determined with a phenotypic assay that monitors loss of proliferative activity and/or cell death. Other aspects are as provided in the Examples.
[00439] A suitable RNAi agent can be selected by any process known in the art or conceivable by one of ordinary skill in the art. For example, the selection criteria can include one or more of the following steps: initial analysis of the APOC3 gene sequence and design of RNAi agents; this design can take into consideration sequence similarity across species (human, cynomolgus, mouse, etc.) and dissimilarity to other (non-APOC3) genes; screening of RNAi agents in vitro (e.g., at 10 nM and 1 nM in Huh-7 cells); selection of RNAi agents with high knock-down at 10 nM and 1 nM in Huh-7 cells; determination of EC50 in Huh-7 cells; confirmation of EC50 in a RCC cell line (Huh-7 cells); analysis of a lack of effect on cell growth relative to a control siRNA; reduction in Huh-7 cells of expression of a HRE-luc (luciferase) reporter gene and not control UB6-luc (luciferase) reporter gene; Western blots to measure Hif-1 , Hif-2, and ARNT levels; testing with human PBMC (peripheral blood mononuclear cells), e.g., to test levels of TNF-alpha to estimate immunogenicity, wherein immunostimulatory sequences are less desired; testing in human whole blood assay, wherein fresh human blood is treated with an RNAi agent and cytokine/chemokine levels are determined [e.g., TNF-alpha (tumor necrosis factor-alpha) and/or MCP1 (monocyte chemotactic protein 1 )], wherein immunostimulatory sequences are less desired;
determination of gene knockdown in vivo using subcutaneous tumors in test animals;
APOC3 target gene modulation analysis, e.g., using a pharmacodynamic (PD) marker, for example, EGLN3, SLC2A1 and VEGF, wherein APOC3 knockdown leads to a dose- dependent alteration of EGLN3, SLC2A1 and VEGF expression in cells; and optimization of specific modifications of the RNAi agents. As appropriate, other cell lines can be used in place of those listed above to identify RNAi agents capable of lowering APOC3 levels or decrease symptoms of an APOC3-related disease.
[00440] By "lower" in the context of APOC3 or a symptom of an APOC3-related disease is meant a statistically significant decrease in such level. The decrease can be, for example, at least 10%, at least 20%, at least 30%, at least 40% or more. If, for a particular disease, or for an individual suffering from a particular disease, the levels or expression of APOC3 are elevated, treatment with an APOC3 RNAi agent of the present disclosure can particularly reduce the level or expression of APOC3 to a level considered in the literature as within the range of normal for an individual without such disorder, or to a level that reduces or ameliorates symptoms of a disease. The level or expression of APOC3 can be measured by evaluation of mRNA (e.g., via Northern blots or PCR), or protein (e.g., Western blots). The effect of a RNAi agent on APOC3 expression can be determined by measuring APOC3 gene transcription rates (e.g., via Northern blots; or reverse transcriptase polymerase chain reaction or real-time polymerase chain reaction).
[00441] As used herein, "down-regulates" refers to any statistically significant decrease in a biological activity and/or expression of APOC3, including full blocking of the activity (i.e., complete inhibition) and/or expression. For example, "down-regulation" can refer to a decrease of at least about 10, 20, 30, 40, 50, 60, 70, 80, 90 or 100 % in APOC3 level, activity and/or expression.
[00442] As used herein, the term "inhibit" or "inhibiting" APOC3 refers to any statistically significant decrease in biological level, activity and/or expression of APOC3, including full blocking of the activity and/or expression. For example, "inhibition" can refer to a decrease of at least about 10, 20, 30, 40, 50, 60, 70, 80, 90 or 100 % in APOC3 level, activity and/or expression. As used herein, the term "inhibit" similarly refers to a significant decrease in level, activity and/or expression, while referring to any other biological agent or composition.
[00443] By "level", it is meant that the APOC3 RNAi agent can alter the level of APOC3, e.g., the level of APOC3 mRNA or the level of APOC3 protein, or the level of activity of APOC3.
[00444] Some diseases include any APOC3-related disease disclosed herein or known in the literature. Particularly in one aspect, in the case of a disease characterized by over- expression and/or hyper-activity of APOC3, administration of a RNAi agent to APOC3 reduces the level, expression and/or activity of APOC3. Thus, in various aspects, administration of a RNAi agent to APOC3 particularly establishes or re-establishes a normal or approximately normal level of APOC3 activity, expression and/or level.
[00445] By "normal" or "approximately normal" in terms of level, expression and/or activity, is meant at least: about 50%, about 60%, about 70%, about 80%, about 90%, and/or about 100%; and/or no more than: about 100%, about 120%, about 130%, about 140%, or about 150% of the level, expression and/or activity of APOC3 in a healthy cell, tissue, or organ. This can be measured using, for example, lung or kidney homogenates, as described in Gambling et al. 2 Kidney Intl. 65: 1774-1781. Particularly in one aspect, administration of the appropriate amount of the appropriate APOC3 RNAi agent restores APOC3 level, activity and/or expression to about 50% to about 150%, more particularly about 60% to about 140%, more particularly to about 70% to about 130%, more particularly to about 80% to about 120%, more particularly to about 90% to about 1 10%, and most particularly to about 100% of that of a healthy cell, tissue or organ. Administration of an APOC3 RNAi to a patient with an APOC3-related disease thus particularly restores the level, activity, and/or expression of APOC3 and the level of Na+ reabsorption to an approximately normal level, as determined by direct measurements of APOC3 mRNA or protein levels, or indirect determinations. In addition, the preferred target amount of APOC3 level, expression and/or activity after APOC3 RNAi agent administration can be calculated to take into account any other perturbations in an APOC3-related pathway. For example, if another factor in an APOC3- related pathway is either over- or under-expressed, APOC3 level, expression or activity may be modulated to attain a more normal state.
[00446] In addition, in various aspects, depending on the disease condition and biological context, it is acceptable to use the RNAi agents of the present disclosure to establish a level of APOC3 expression, activity and/or level which is below the normal level, or above the normal level.
[00447] Types of RNAi Agents and Modification Thereof
[00448] The use of RNAi agents or compositions comprising an antisense nucleic acid to down-modulate the expression of a particular protein in a cell is well known in the art. A RNAi agent comprises a sequence complementary to, and is capable of hydrogen bonding to, the coding strand of another nucleic acid (e.g., an mRNA). Thus, in various aspects, the RNAi agents of the present disclosure encompass any RNAi agents which target (e.g., are complementary, capable of hybridizing or hydrogen bonding to, etc.) any sequence presented, e.g., in any of the Tables.
[00449] Once a functional guide strand has been identified, many variations to the guide and/or passenger strand can be made. For example, the RNAi agent may have
modifications internally, or at one or both ends. The modifications at the ends can help stabilize the RNAi agent, protecting it from degradation by nucleases in the blood. The RNAi agents may optionally be directed to regions of the APOC3 mRNA known or predicted to be near or at splice sites of the gene.
[00450] A RNAi agent can be constructed using chemical synthesis and enzymatic ligation reactions using procedures known in the art. For example, RNAi agent can be chemically synthesized using naturally-occurring nucleotides or variously modified nucleotides designed to decrease off-target effects, and/or increase the biological stability of the molecules or to increase the physical stability of the duplex formed between the antisense and sense nucleic acids, e.g., phosphorothioate derivatives and acridine substituted nucleotides can be used.
[00451] "G," "C," "A," "T" and "U" each generally stand for a nucleotide that contains guanine, cytosine, adenine, thymidine and uracil as a base, respectively. However, the terms "ribonucleotide", "deoxynucleotide", or "nucleotide" can also refer to a modified nucleotide or a surrogate replacement moiety. The skilled person is well aware that guanine, cytosine, adenine, and uracil may be replaced by other moieties without substantially altering the base pairing properties of an oligonucleotide comprising a nucleotide bearing such replacement moiety. For example, without limitation, a nucleotide comprising inosine as its base may base pair with nucleotides containing adenine, cytosine, or uracil. Hence, nucleotides containing uracil, guanine, or adenine may be replaced in the nucleotide sequences of dsRNA featured in the present disclosure by a nucleotide containing, for example, inosine. In another example, adenine and cytosine anywhere in the oligonucleotide can be replaced with guanine and uracil, respectively to form Wobble base pairing with the target mRNA. Sequences containing such replacement moieties are suitable for the compositions and methods featured in the present disclosure.
[00452] The skilled artisan will recognize that the term "RNA molecule" or "ribonucleic acid molecule" encompasses not only RNA molecules as expressed or found in nature (i.e., are naturally occurring), but also non-naturally occurring analogs and derivatives of RNA comprising one or more ribonucleotide/ribonucleoside analogs or derivatives as described herein or as known in the art. The RNA can be modified in the nucleobase structure or in the ribose-phosphate backbone structure, e.g., as described herein below. However, the molecules comprising ribonucleoside analogs or derivatives can retain the ability to form a duplex. As non-limiting examples, either or both strand of an RNAi agent can comprise at least one modified ribonucleoside, including but not limited to a 2'-0-methyl modified nucleotide, a nucleoside comprising a 5' phosphorothioate group, a terminal nucleoside linked to a cholesteryl derivative or dodecanoic acid bisdecylamide group, a locked nucleoside, an abasic nucleoside, a 2'-deoxy-2'-fluoro modified nucleoside, a 2'-amino- modified nucleoside, 2'-alkyl-modified nucleoside, morpholino nucleoside, an unlocked ribonucleotide (e.g., an acyclic nucleotide monomer, as described in WO 2008/147824), a phosphoramidate or a non-natural base comprising nucleoside, or any combination thereof. Alternatively, an RNA molecule can comprise at least two modified ribonucleosides, at least 3, at least 4, at least 5, at least 6, at least 7, at least 8, at least 9, at least 10, at least 15, at least 20 or more, up to the entire length of the dsRNA molecule. The modifications need not be the same for each of such a plurality of modified ribonucleosides in an RNA molecule. In one aspect, modified RNAs contemplated for use in methods and compositions described herein are peptide nucleic acids (PNAs) that have the ability to form the required duplex structure and that permit or mediate the specific degradation of a target RNA via a RISC pathway. In addition, the RNAi agent can comprise one or two strands which are a RNA, or a mixture of RNA, DNA, LNA, morpholino, TNA, GNA, ANA, HNA, CeNA, UNA, and/or FANA, etc. As a non-limiting example, one or both strands could be, for example, RNA except that one or more nucleotides is replaced by DNA, LNA, morpholino, TNA, GNA, ANA, HNA, CeNA, FANA, and/or UNA, etc.
[00453] Examples of modified nucleotides which can be used to generate the RNAi agent include 5-fluorouracil, 5-bromouracil, 5-chlorouracil, 5-iodouracil, hypoxanthine, xantine, 4- acetylcytosine, 5-(carboxyhydroxylmethyl) uracil, 5-carboxymethylaminomethyl-2-thiouridine, 5-carboxymethylaminomethyluracil, dihydrouracil, beta-D-galactosylqueosine, inosine, N6- isopentenyladenine, 1 -methylguanine, 1 -methylinosine, 2,2-dimethylguanine, 2- methyladenine, 2-methylguanine, 3-methylcytosine, 5-methylcytosine, N6-adenine, 7- methylguanine, 5-methylaminomethyluracil, 5-methoxyaminomethyl-2-thiouracil, beta-D- mannosylqueosine, 5'-methoxycarboxymethyluracil, 5-methoxyuracil, 2-methylthio-N6- isopentenyladenine, uracil-5-oxyacetic acid (v), wybutoxosine, pseudouracil, queosine, 2- thiocytosine, 5-methyl-2-thiouracil, 2-thiouracil, 4-thiouracil, 5-methyluracil, uracil-5- oxyacetic acid methylester, uracil-5-oxyacetic acid (v), 5-methyl-2-thiouracil, 3-(3-amino-3-N- 2-carboxypropyl) uracil, (acp3)w, and 2,6-diaminopurin (Usman et al. 1992 TIBS 17:34; Usman et al. 1994 Nucl. Acids Symp. Ser. 31 : 163; Burgin et al. 1996 Biochem. 35: 14090).
[00454] A "modified variant" of a sequence disclosed herein includes any variant comprising the same sequence, but with a modification in the base, sugar, phosphate or backbone (but not a base substitution, e.g., A for G, or C for U). Thus, a modified variant can comprise any modified nucleotide described above (e.g., 5-fluorouracil, 5-bromouracil, 5-chlorouracil, 5-iodouracil, hypoxanthine, xantine, 4-acetylcytosine, 5- (carboxyhydroxylmethyl) uracil, etc.). When a base is replaced by a corresponding modified base (e.g., A for modified A), these modified nucleotides do not constitute a mismatch or base difference. Thus a given sequence with a U at a particular position and a modified variant comprising a 5-fluorouracil, 5-bromouracil, 5-chlorouracil, or 5-iodouracil at the same sequence would differ by 0 nt (or have no mismatches); however, a given sequence with a C at a particular position and a different sequence with a 5-fluorouracil (wherein the two sequences are otherwise identical) would differ by 1 nt (1 mismatch).
[00455] Replacing the 3'-terminal nucleotide overhanging segments of a 21 -mer siRNA duplex having two-nucleotide 3'-overhangs with deoxyribonucleotides does not have an adverse effect on RNAi activity. Replacing up to four nucleotides on each end of the siRNA with deoxyribonucleotides has been well tolerated, whereas complete substitution with deoxyribonucleotides results in no RNAi activity. International PCT Publication No. WO 00/44914, and Beach et al. International PCT Publication No. WO 01/68836 preliminarily suggest that siRNA may include modifications to either the phosphate-sugar backbone or the nucleoside to include at least one of a nitrogen or sulfur heteroatom. Kreutzer et al. Canadian Patent Application No. 2,359, 180, also describe certain chemical modifications for use in dsRNA constructs in order to counteract activation of double-stranded RNA- dependent protein kinase PKR, specifically 2'-amino or 2'-0-methyl nucleotides, and nucleotides containing a 2'-0 or 4'-C methylene bridge. Additional 3'-terminal nucleotide overhangs include dT (deoxythimidine), 2'-0,4'-C-ethylene thymidine (eT), and 2- hydroxyethyl phosphate (hp). 4-thiouracil and 5-bromouracil substitutions can also be made. Parrish et al. 2000 Molecular Cell 6: 1077-1087. [00456] Those skilled in the art will appreciate that it is possible to synthesize and modify the siRNA as desired, using any conventional method known in the art (see Henschel et al. 2004 DEQOR: a web-based tool for the design and quality control of siRNAs. Nucleic Acids Research 32 (Web Server Issue): W1 13-W120). In addition, if the RNAi agent is a shRNA, it will be apparent to those skilled in the art that there are a variety of regulatory sequences (for example, constitutive or inducible promoters, tissue- specific promoters or functional fragments thereof, etc.) which are useful for shRNA expression construct/vector.
[00457] There are several examples in the art describing sugar, base, phosphate and backbone modifications that can be introduced into nucleic acid molecules with significant enhancement in their nuclease stability and efficacy. For example, oligonucleotides are modified to enhance stability and/or enhance biological activity by modification with nuclease resistant groups, for example, 2'-amino, 2'-C-allyl, 2'-flouro, 2'-0-methyl, 2'-0-allyl, 2'-H, nucleotide base modifications (for a review see Usman and Cedergren 1992 TIBS. 17: 34; Usman et al. 1994 Nucleic Acids Symp. Ser. 31 : 163; Burgin et al. 1996 Biochemistry 35: 14090). Sugar modification of nucleic acid molecules are extensively described in the art.
[00458] In various aspects, the RNAi agent comprises a 2'-modification selected from the group consisting of: 2'-deoxy, 2'-deoxy-2'-fluoro, 2'-0-methyl, 2'-0-methoxyethyl (2'-0-MOE), 2'-0-aminopropyl (2 -O-AP), 2'-0-dimethylaminoethyl (2 -O-DMAOE), 2 -0- dimethylaminopropyl (2 -O-DMAP), 2'-0-dimethylaminoethyloxyethyl (2'-0-DMAEOE), and 2'-0-N-methylacetamido (2 -O-NMA).
[00459] Additional modifications and conjugations of RNAi agents have been described. Soutschek et al. 2004 Nature 432: 173-178 presented conjugation of cholesterol to the 3'- end of the sense strand of a siRNA molecule by means of a pyrrolidine linker, thereby generating a covalent and irreversible conjugate. Chemical modifications (including conjugation with other molecules) of RNAi agents may also be made to improve the in vivo pharmacokinetic retention time and efficiency.
[00460] In various aspects, the RNAi agent to APOC3 comprises at least one 5'-uridine- adenine-3' (5'-ua-3') dinucleotide, wherein the uridine is a 2'-modified nucleotide; at least one 5'-uridine-guanine-3' (5'-ug-3') dinucleotide, wherein the 5'-uridine is a 2'-modified nucleotide; at least one 5'-cytidine-adenine-3' (5'-ca-3') dinucleotide, wherein the 5'-cytidine is a 2'-modified nucleotide; and/or at least one 5'-uridine-uridine-3' (5'-uu-3') dinucleotide, wherein the 5'-uridine is a 2'-modified nucleotide. In certain aspects, the RNAi agent can comprise a non-natural nucleobase, wherein the non-natural nucleobase is difluorotolyl, nitroindolyl, nitropyrrolyl, or nitroimidazolyl. In a particular aspect, the non-natural nucleobase is difluorotolyl. In certain aspects, only one of the two oligonucleotide strands contains a non-natural nucleobase. In certain aspects, both of the oligonucleotide strands contain a non-natural nucleobase. [00461] In another aspect, the RNAi comprises a gap or contains mismatch comprising an abasic nucleotide.
[00462] In another aspect, the RNAi agent has a single-stranded nick (e.g., a break or missing bond in the backbone). In various aspects, a single-stranded nick can be in either the sense or anti-sense strand, or both.
[00463] This nick can be, for example, in the sense strand, producing a small internally segmented interfering RNA, or sisiRNA, which may have less off-target effects than the corresponding RNAi agent without a nick. See, for example, WO 2007/107162 to Wengels and Kjems.
[00464] The antisense nucleic acid or RNAi agent can also have an alternative backbone such as locked nucleic acids (LNA), Morpholinos, peptidic nucleic acids (PNA), threose nucleic acid (TNA), or glycol nucleic acid (GNA), arabinose nucleic acid (ANA), 2 - fluoroarabinose nucleic acid (FANA), cyclohexene nucleic acid (CeNA), anhydrohexitol nucleic acid (HNA), or unlocked nucleic acid (UNA), and/or it can be labeled (e.g., radiolabeled or otherwise tagged). FANA are described in Dowler et al. 2006 Nucl. Acids Res. 34: 1669-1675.
[00465] One or both strands can comprise an alternative backbone.
[00466] In yet another aspect, the RNAi agent employed by the methods of the present disclosure can include an a-anomeric nucleic acid molecule. An a-anomeric nucleic acid molecule forms specific double-stranded hybrids with complementary RNA in which, contrary to the usual β-units, the strands run parallel to each other. Gaultier et al. 1987 Nucleic Acids. Res. 15: 6625-6641 .
[00467] The antisense nucleic acid molecule can also comprise a 2'-o-methylribonucleotide (Inoue et al. 1987 Nucleic Acids Res. 15: 6131-6148) or a chimeric RNA-DNA analogue (Inoue et al. 1987 FEBS Lett. 215: 327-330).
[00468] Other modifications and/or other changes can be made to the RNAi agent. A portion of the RNAi agent can be double-stranded DNA, while another portion is double- stranded RNA, forming a DNA-RNA chimera (See, for example, Yamato et al. 201 1. Cancer Gene Ther. 18: 587-597). Mismatches between the guide and passenger stand can also be introduced, though some positions may be better suited than others (See, for example, U.S. Patent App. No. 2009/0209626 to Khvorova). The passenger strand can also be shortened, to as short as 15 or 16 nt, while the guide strand remains 19 nt or longer (See, for example, Sun et al. 2008 Nature Biotech. 26: 1379-1382; and Chu and Rana 2008 RNA 14: 1714- 1719). This can increase incorporation of the guide strand into the RNA-induced Silence Complex (RISC), and decrease incorporation of the passenger strand, than reducing off- target effects. In some cases, the passenger strand may be more amenable to modification (e.g., single-stranded nicking, nucleotide modifications, and shortening) than the guide strand.
[00469] These and many other modifications can be made once a functional guide strand is identified.
[00470] Pharmaceutical Compositions of RNAi Agents
[00471] As used here, a "pharmaceutical composition" comprises a pharmaceutically effective amount of one or more APOC3 RNAi agent, a pharmaceutically acceptable carrier, and, optionally, an additional disease treatment which works synergistically with the RNAi agent. As used herein, "pharmacologically effective amount," "therapeutically effective amount" or simply "effective amount" refers to that amount of a RNAi agent effective to produce the intended pharmacological, therapeutic or preventive result. For example, if a given clinical treatment is considered effective where there is at least a 10% reduction in a measurable parameter associated with a disease or disorder, a therapeutically effective amount of a drug for the treatment of that disease or disorder is the amount necessary to effect at least a 10% reduction in that parameter. In this aspect, a therapeutically effective amount of a RNAi agent targeting APOC3 can reduce APOC3 protein levels by at least 10%. In additional aspects, a given clinical treatment is considered effective where there is at least a 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90 or 95% reduction in a measurable parameter associated with a disease or disorder, and the therapeutically effective amount of a drug for the treatment of that disease or disorder is the amount necessary to effect at least a 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90 or 95% reduction, respectively, in that parameter.
[00472] The term "pharmaceutically acceptable carrier" refers to a carrier for administration of a therapeutic agent. Such carriers include, but are not limited to, lipid nanoparticles, saline, buffered saline, dextrose, water, glycerol, ethanol, and combinations thereof. The term specifically excludes cell culture medium. Any appropriate phamaceutical carrier known in the art can be used in conjunction with the RNAi agents disclosed herein.
[00473] Pharmaceutical Composition Comprising a RNAi Agent to APOC3
[00474] Additional components of a pharmaceutical composition comprising a RNAi Agent to APOC3 are contemplated to aid in delivery, stability, efficacy, or reduction of
immunogenicity.
[00475] Liposomes have been used previously for drug delivery (e.g., delivery of a chemotherapeutic). Liposomes (e.g., cationic liposomes) are described in PCT publications WO02/100435A1 , WO03/015757A1 , WO04029213A2; and WO/201 1/076807; U.S. Pat. Nos. 5,962,016; 5,030,453; and 6,680,068; and U.S. Patent Application 2004/0208921 . A process of making liposomes is also described in WO04/002453A1. Furthermore, neutral lipids have been incorporated into cationic liposomes (e.g., Farhood et al. 1995), as well as PEGylated lipids.
[00476] Cationic liposomes have been used to deliver RNAi agent to various cell types (Sioud and Sorensen 2003; U.S. Patent Application 2004/0204377; Duxbury et al., 2004; Donze and Picard, 2002).
[00477] Use of neutral liposomes disclosed in Miller et al. 1998, and U.S. Patent Application 2003/0012812.
[00478] As used herein, the term "SNALP" refers to a stable nucleic acid-lipid particle. A SNALP represents a vesicle of lipids coating a reduced aqueous interior comprising a nucleic acid such as an RNAi or a plasmid from which an RNAi is transcribed. SNALPs are described, e.g., in U.S. Patent Application Publication Nos. 20060240093, 20070135372, and in International Application No. WO 2009082817.
[00479] Chemical transfection using lipid-based, amine-based and polymer-based techniques is disclosed in products from Ambion Inc., Austin, Tex.; and Novagen, EMD Biosciences, Inc, an Affiliate of Merck KGaA, Darmstadt, Germany); Ovcharenko D (2003) "Efficient delivery of siRNAs to human primary cells." Ambion TechNotes 10 (5): 15-16). Additionally, Song et al. (Nat Med. published online (Feb I 0, 2003) doi: 10.1038/nm828) and others [Caplen et al. 2001 Proc. Natl. Acad. Sci. (USA), 98: 9742-9747; and McCaffrey et al. Nature 414: 34-39] disclose that liver cells can be efficiently transfected by injection of the siRNA into a mammal's circulatory system.
[00480] A variety of molecules have been used for cell-specific RNAi agent delivery. See, for example, WO/201 1/076807. For example, the nucleic acid-condensing property of protamine has been combined with specific antibodies to deliver siRNAs. Song et al. 2005 Nat Biotech. 23: 709-717. The self-assembly PEGylated polycation polyethylenimine (PEI) has also been used to condense and protect siRNAs. Schiffelers et al. 2004 Nucl. Acids Res. 32: el49, 141 -1 10.
[00481] The RNAi agents of the present disclosure can be delivered via, for example, Lipid nanoparticles (LNP); neutral liposomes (NL); polymer nanoparticles; double-stranded RNA binding motifs (dsRBMs); or via modification of the RNAi agent (e.g., covalent attachment to the dsRNA) or by any method known in the art for delivery of a RNAi agent comprising nucleic acids.
[00482] Lipid nanoparticles (LNP) are self-assembling cationic lipid based systems. These can comprise, for example, a neutral lipid (the liposome base); a cationic lipid (for siRNA loading); cholesterol (for stabilizing the liposomes); and PEG-lipid (for stabilizing the formulation, charge shielding and extended circulation in the bloodstream). [00483] The cationic lipid can comprise, for example, a headgroup, a linker, a tail and a cholesterol tail. The LNP can have, for example, good delivery to the diseased area, extended circulation in the blood, small particles (e.g., less than 100 nm), and stability in the microenvironment of the diseased area (which may have low pH and/or be hypoxic).
[00484] Neutral liposomes (NL) are non-cationic lipid based particles.
[00485] Polymer nanoparticles are self-assembling polymer-based particles.
[00486] Double-stranded RNA binding motifs (dsRBMs) are self-assembling RNA binding proteins, which will need modifications.
[00487] APOC3 RNAi agent compositions in a Lipid nanoparticles (LNP) comprising a neutral lipid; a cationic lipid; cholesterol; and PEG-lipid
[00488] Lipid nanoparticles (LNP) are self-assembling cationic lipid based systems. These can comprise, for example, a neutral lipid (the liposome base); a cationic lipid (for siRNA loading); cholesterol (for stabilizing the liposomes); and PEG-lipid (for stabilizing the formulation, charge shielding and extended circulation in the bloodstream).
A neutral lipid is, for example, the liposome base. A cationic lipid is, for example, for siRNA loading. Cholesterol is, for example, for stabilizing the liposomes. PEG-lipid is, for example, for stabilizing the formulation, charge shielding and extended circulation in the bloodstream.
[00489] Additional pharmaceutical compositions
[00490] In various aspects, the RNAi agent to APOC3 is packaged as a monotherapy into a delivery vehicle, or may be further ligated to one or more diagnostic compound, reporter group, cross-linking agent, nuclease-resistance conferring moiety, natural or unusual nucleobase, lipophilic molecule, cholesterol, lipid, lectin, steroid, uvaol, hecigenin, diosgenin, terpene, triterpene, sarsasapogenin, Friedelin, epifriedelanol-derivatized lithocholic acid, vitamin, carbohydrate, dextran, pullulan, chitin, chitosan, synthetic carbohydrate, oligo lactate 15-mer, natural polymer, low- or medium-molecular weight polymer, inulin, cyclodextrin, hyaluronic acid, protein, protein-binding agent, integrin-targeting molecule, polycationic, peptide, polyamine, peptide mimic, and/or transferrin.
[00491] The RNAi agents of the present disclosure can be prepared in a pharmaceutical composition comprising various components appropriate for the particular method of administration of the RNAi agent.
[00492] Particular specific aspects
[00493] In a particular specific aspect, the present disclosure is a composition comprising one or more APOC3 RNAi agents. [00494] In various aspects, the disclosure encompasses a composition comprising any one or more of the following:
[00495] A RNAi agent comprising a first and a second strand, wherein the sequence of the first strand is the sequence of SEQ ID NO: 1 , or modified or unmodified variants thereof.
[00496] A RNAi agent comprising a first and a second strand, wherein the sequence of the first strand is the sequence of SEQ ID NO: 2, or modified or unmodified variants thereof.
[00497] A RNAi agent comprising a first and a second strand, wherein the sequence of the first strand is the sequence of SEQ ID NO: 41 , or modified or unmodified variants thereof.
[00498] A RNAi agent comprising a first and a second strand, wherein the sequence of the first strand is the sequence of SEQ ID NO: 48, or modified or unmodified variants thereof.
[00499] A RNAi agent comprising a first and a second strand, wherein the sequence of the first strand is the sequence of SEQ ID NO: 56, or modified or unmodified variants thereof.
[00500] A RNAi agent comprising a first and a second strand, wherein the sequence of the first strand is the sequence of SEQ ID NO: 71 , or modified or unmodified variants thereof.
[00501] A RNAi agent comprising a first and a second strand, wherein the sequence of the first strand is the sequence of SEQ ID NO: 102, or modified or unmodified variants thereof.
[00502] Particular specific aspects
[00503] In a particular specific aspect, the present disclosure is a composition comprising one or more APOC3 RNAi agents.
[00504] In various aspects, the disclosure encompasses a composition comprising any one or more of the following:
[00505] A RNAi agent comprising a first and a second strand, wherein the sequence of the first strand is the sequence of SEQ ID NO: 1 , and the sequence of the second strand is the sequence of SEQ ID NO: 45, or modified or unmodified variants thereof.
[00506] A RNAi agent comprising a first and a second strand, wherein the sequence of the first strand is the sequence of SEQ ID NO: 2, and the sequence of the second strand is the sequence of SEQ ID NO: 100, or modified or unmodified variants thereof.
[00507] A RNAi agent comprising a first and a second strand, wherein the sequence of the first strand is the sequence of SEQ ID NO: 41 , and the sequence of the second strand is the sequence of SEQ ID NO: 9, or modified or unmodified variants thereof.
[00508] A RNAi agent comprising a first and a second strand, wherein the sequence of the first strand is the sequence of SEQ ID NO: 48, and the sequence of the second strand is the sequence of SEQ ID NO: 39, or modified or unmodified variants thereof.
[00509] A RNAi agent comprising a first and a second strand, wherein the sequence of the first strand is the sequence of SEQ ID NO: 56, and the sequence of the second strand is the sequence of SEQ ID NO: 23, or modified or unmodified variants thereof. [00510] A RNAi agent comprising a first and a second strand, wherein the sequence of the first strand is the sequence of SEQ ID NO: 102, and the sequence of the second strand is the sequence of SEQ ID NO: 23, or modified or unmodified variants thereof.
[00511] Additional particular specific aspects
[00512] In a particular specific aspect, the present disclosure is a composition comprising one or more APOC3 RNAi agents.
[00513] In various aspects, the disclosure encompasses a composition comprising any one or more of the following:
[00514] A RNAi agent comprising a first and a second strand, wherein the sequence of the first strand comprises the sequence of SEQ ID NO: 41 , wherein the length of the first and second strand are each no more than about 30 nt, or modified or unmodified variants thereof.
[00515] A RNAi agent comprising a first and a second strand, wherein the sequence of the first strand comprises the sequence of SEQ ID NO: 71 , wherein the length of the first and second strand are each no more than about 30 nt, or modified or unmodified variants thereof.
[00516] A RNAi agent comprising a first and a second strand, wherein the sequence of the first strand comprises the sequence of SEQ ID NO: 56, wherein the length of the first and second strand are each no more than about 30 nt, or modified or unmodified variants thereof.
[00517] A RNAi agent comprising a first and a second strand, wherein the sequence of the first strand comprises the sequence of SEQ ID NO: 102, wherein the length of the first and second strand are each no more than about 30 nt, or modified or unmodified variants thereof.
[00518] A RNAi agent comprising a first and a second strand, wherein the sequence of the first strand comprises the sequence of SEQ ID NO: 1 , wherein the length of the first and second strand are each no more than about 30 nt, or modified or unmodified variants thereof.
[00519] A RNAi agent comprising a first and a second strand, wherein the sequence of the first strand comprises the sequence of SEQ ID NO: 2, wherein the length of the first and second strand are each no more than about 30 nt, or modified or unmodified variants thereof.
[00520] Additional particular specific aspects
[00521] In various aspects, the disclosure comprises a RNAi agent comprising a first and a second strand, wherein the sequence of the first strand comprises at least 18 contiguous nucleotides of the sequence of any one or more RNAi agent disclosed herein, or modified or unmodified variants thereof.
[00522] In various aspects, the disclosure encompasses a composition comprising any one or more of the following:
[00523] A RNAi agent comprising a first and a second strand, wherein the sequence of the first strand comprises at least 18 contiguous nucleotides of the sequence of SEQ ID NO: 41 , or modified or unmodified variants thereof.
[00524] A RNAi agent comprising a first and a second strand, wherein the sequence of the first strand comprises at least 18 contiguous nucleotides of the sequence of SEQ ID NO: 48, or modified or unmodified variants thereof.
[00525] A RNAi agent comprising a first and a second strand, wherein the sequence of the first strand comprises at least 18 contiguous nucleotides of the sequence of SEQ ID NO: 56, or modified or unmodified variants thereof.
[00526] A RNAi agent comprising a first and a second strand, wherein the sequence of the first strand comprises at least 18 contiguous nucleotides of the sequence of SEQ ID NO: 102, or modified or unmodified variants thereof.
[00527] A RNAi agent comprising a first and a second strand, wherein the sequence of the first strand comprises at least 18 contiguous nucleotides of the sequence of SEQ ID NO: 1 , or modified or unmodified variants thereof.
[00528] A RNAi agent comprising a first and a second strand, wherein the sequence of the first strand comprises at least 18 contiguous nucleotides of the sequence of SEQ ID NO: 2, or modified or unmodified variants thereof.
[00529] Additional particular specific aspects
[00530] In various aspects, the disclosure comprises a RNAi agent comprising a first and a second strand, wherein the sequence of the first strand comprises at least 18 contiguous nucleotides differing by 0, 1 , 2, or 3 nt from a first strand, or modified or unmodified variants thereof.
[00531] In various aspects, the disclosure encompasses a composition comprising any one or more of the following:
[00532] A RNAi agent comprising a first and a second strand, wherein the sequence of the first strand comprises at least 18 contiguous nucleotides differing by 0, 1 , 2, or 3 nt from the sequence of SEQ ID NO: 41 , or modified or unmodified variants thereof.
[00533] A RNAi agent comprising a first and a second strand, wherein the sequence of the first strand comprises at least 18 contiguous nucleotides differing by 0, 1 , 2, or 3 nt from the sequence of SEQ ID NO: 48, or modified or unmodified variants thereof. [00534] A RNAi agent comprising a first and a second strand, wherein the sequence of the first strand comprises at least 18 contiguous nucleotides differing by 0, 1 , 2, or 3 nt from the sequence of SEQ ID NO: 56, or modified or unmodified variants thereof.
[00535] A RNAi agent comprising a first and a second strand, wherein the sequence of the first strand comprises at least 18 contiguous nucleotides differing by 0, 1 , 2, or 3 nt from the sequence of SEQ ID NO: 102, or modified or unmodified variants thereof.
[00536] A RNAi agent comprising a first and a second strand, wherein the sequence of the first strand comprises at least 18 contiguous nucleotides differing by 0, 1 , 2, or 3 nt from the sequence of SEQ ID NO: 1 , or modified or unmodified variants thereof.
[00537] A RNAi agent comprising a first and a second strand, wherein the sequence of the first strand comprises at least 18 contiguous nucleotides differing by 0, 1 , 2, or 3 nt from the sequence of SEQ ID NO: 2, or modified or unmodified variants thereof.
[00538] Additional particular specific aspects
[00539] In various aspects, the disclosure comprises a RNAi agent comprising a first and a second strand, wherein the sequence of the first strand comprises at least 14 contiguous nucleotides of the sequence of any one or more RNAi agent disclosed herein, or modified or unmodified variants thereof.
[00540] In various aspects, the disclosure encompasses a composition comprising any one or more of the following:
[00541] A RNAi agent comprising a first and a second strand, wherein the sequence of the first strand comprises at least 14 contiguous nucleotides of the sequence of SEQ ID NO: 41 , or modified or unmodified variants thereof.
[00542] A RNAi agent comprising a first and a second strand, wherein the sequence of the first strand comprises at least 14 contiguous nucleotides of the sequence of SEQ ID NO: 48, or modified or unmodified variants thereof.
[00543] A RNAi agent comprising a first and a second strand, wherein the sequence of the first strand comprises at least 14 contiguous nucleotides of the sequence of SEQ ID NO: 56, or modified or unmodified variants thereof.
[00544] A RNAi agent comprising a first and a second strand, wherein the sequence of the first strand comprises at least 14 contiguous nucleotides of the sequence of SEQ ID NO: 102, or modified or unmodified variants thereof.
[00545] A RNAi agent comprising a first and a second strand, wherein the sequence of the first strand comprises at least 14 contiguous nucleotides of the sequence of SEQ ID NO: 1 , or modified or unmodified variants thereof. [00546] A RNAi agent comprising a first and a second strand, wherein the sequence of the first strand comprises at least 14 contiguous nucleotides of the sequence of SEQ ID NO: 2, or modified or unmodified variants thereof.
[00547] Additional particular specific aspects
[00548] In various aspects, the disclosure comprises a RNAi agent comprising a first and a second strand, wherein the sequence of the first strand comprises at least 14 contiguous nucleotides differing by 0, 1 , 2, or 3 nt from a first strand, or modified or unmodified variants thereof.
[00549] In various aspects, the disclosure encompasses a composition comprising any one or more of the following:
[00550] A RNAi agent comprising a first and a second strand, wherein the sequence of the first strand comprises at least 14 contiguous nucleotides differing by 0, 1 , 2, or 3 nt from the sequence of SEQ ID NO: 41 , or modified or unmodified variants thereof.
[00551] A RNAi agent comprising a first and a second strand, wherein the sequence of the first strand comprises at least 14 contiguous nucleotides differing by 0, 1 , 2, or 3 nt from the sequence of SEQ ID NO: 48, or modified or unmodified variants thereof.
[00552] A RNAi agent comprising a first and a second strand, wherein the sequence of the first strand comprises at least 14 contiguous nucleotides differing by 0, 1 , 2, or 3 nt from the sequence of SEQ ID NO: 56, or modified or unmodified variants thereof.
[00553] A RNAi agent comprising a first and a second strand, wherein the sequence of the first strand comprises at least 14 contiguous nucleotides differing by 0, 1 , 2, or 3 nt from the sequence of SEQ ID NO: 102, or modified or unmodified variants thereof.
[00554] A RNAi agent comprising a first and a second strand, wherein the sequence of the first strand comprises at least 14 contiguous nucleotides differing by 0, 1 , 2, or 3 nt from the sequence of SEQ ID NO: 1 , or modified or unmodified variants thereof.
[00555] A RNAi agent comprising a first and a second strand, wherein the sequence of the first strand comprises at least 14 contiguous nucleotides differing by 0, 1 , 2, or 3 nt from the sequence of SEQ ID NO: 2, or modified or unmodified variants thereof.
[00556] Additional Particular specific aspects
[00557] In a particular specific aspect, the present disclosure is a composition comprising one or more APOC3 RNAi agents.
[00558] In various aspects, the disclosure encompasses a composition comprising any one or more of the following: [00559] A RNAi agent comprising a first and a second strand, wherein the sequence of the first strand is the sequence of SEQ ID NO: 41 , and/or the sequence of the second strand is the sequence of SEQ ID NO: 9, or modified or unmodified variants thereof.
[00560] A RNAi agent comprising a first and a second strand, wherein the sequence of the first strand is the sequence of SEQ ID NO: 48, and/or the sequence of the second strand is the sequence of SEQ ID NO: 39, or modified or unmodified variants thereof.
[00561] A RNAi agent comprising a first and a second strand, wherein the sequence of the first strand is the sequence of SEQ ID NO: 56, and/or the sequence of the second strand is the sequence of SEQ ID NO: 23, or modified or unmodified variants thereof.
[00562] A RNAi agent comprising a first and a second strand, wherein the sequence of the first strand is the sequence of SEQ ID NO: 102, and/or the sequence of the second strand is the sequence of SEQ ID NO: 81 , or modified or unmodified variants thereof.
[00563] A RNAi agent comprising a first and a second strand, wherein the sequence of the first strand is the sequence of SEQ ID NO: 1 , and/or the sequence of the second strand is the sequence of SEQ ID NO: 45, or modified or unmodified variants thereof.
[00564] A RNAi agent comprising a first and a second strand, wherein the sequence of the first strand is the sequence of SEQ ID NO: 2, and/or the sequence of the second strand is the sequence of SEQ ID NO: 100, or modified or unmodified variants thereof.
[00565] Additional Particular specific aspects
[00566] In a particular specific aspect, the present disclosure is a composition comprising one or more APOC3 RNAi agents.
[00567] In various aspects, the disclosure encompasses a composition comprising any one or more of the following:
[00568] A RNAi agent comprising a first and a second strand, wherein the sequence of the first strand comprises the sequence of SEQ ID NO: 41 , and/or the sequence of the second strand comprises the sequence of SEQ ID NO: 9, wherein the length of the first and second strand are each no more than about 30 nt, or modified or unmodified variants thereof.
[00569] A RNAi agent comprising a first and a second strand, wherein the sequence of the first strand comprises the sequence of SEQ ID NO: 48, and/or the sequence of the second strand comprises the sequence of SEQ ID NO: 39, wherein the length of the first and second strand are each no more than about 30 nt, or modified or unmodified variants thereof.
[00570] A RNAi agent comprising a first and a second strand, wherein the sequence of the first strand comprises the sequence of SEQ ID NO: 56, and/or the sequence of the second strand comprises the sequence of SEQ ID NO: 23, wherein the length of the first and second strand are each no more than about 30 nt, or modified or unmodified variants thereof. [00571] A RNAi agent comprising a first and a second strand, wherein the sequence of the first strand comprises the sequence of SEQ ID NO: 102, and/or the sequence of the second strand comprises the sequence of SEQ ID NO: 81 , wherein the length of the first and second strand are each no more than about 30 nt, or modified or unmodified variants thereof.
[00572] A RNAi agent comprising a first and a second strand, wherein the sequence of the first strand comprises the sequence of SEQ ID NO: 1 , and/or the sequence of the second strand comprises the sequence of SEQ ID NO: 45, wherein the length of the first and second strand are each no more than about 30 nt, or modified or unmodified variants thereof.
[00573] A RNAi agent comprising a first and a second strand, wherein the sequence of the first strand comprises the sequence of SEQ ID NO: 2, and/or the sequence of the second strand comprises the sequence of SEQ ID NO: 100, wherein the length of the first and second strand are each no more than about 30 nt, or modified or unmodified variants thereof.
[00574] Additional particular specific aspects
[00575] In various aspects, the disclosure comprises a RNAi agent comprising a first and a second strand, wherein the sequence of the first strand comprises at least 14 contiguous nucleotides of a first strand, and/or the sequence of the second strand comprises at least 14 contiguous nucleotides of the second strand of any any one or more RNAi agent disclosed herein.
[00576] In various aspects, the disclosure encompasses a composition comprising any one or more of the following:
[00577] A RNAi agent comprising a first and a second strand, wherein the sequence of the first strand comprises at least 14 contiguous nucleotides of the sequence of SEQ ID NO: 41 , and/or the sequence of the second strand comprises at least 14 contiguous nucleotides of the sequence of SEQ ID NO: 9, wherein the length of the first and second strand are each no more than about 30 nt, or modified or unmodified variants thereof.
[00578] A RNAi agent comprising a first and a second strand, wherein the sequence of the first strand comprises at least 14 contiguous nucleotides of the sequence of SEQ ID NO: 48, and/or the sequence of the second strand comprises at least 14 contiguous nucleotides of the sequence of SEQ ID NO: 39, wherein the length of the first and second strand are each no more than about 30 nt, or modified or unmodified variants thereof.
[00579] A RNAi agent comprising a first and a second strand, wherein the sequence of the first strand comprises at least 14 contiguous nucleotides of the sequence of SEQ ID NO: 56, and/or the sequence of the second strand comprises at least 14 contiguous nucleotides of the sequence of SEQ ID NO: 23, wherein the length of the first and second strand are each no more than about 30 nt, or modified or unmodified variants thereof. [00580] A RNAi agent comprising a first and a second strand, wherein the sequence of the first strand comprises at least 14 contiguous nucleotides of the sequence of SEQ ID NO: 102, and/or the sequence of the second strand comprises at least 14 contiguous nucleotides of the sequence of SEQ ID NO: 81 , wherein the length of the first and second strand are each no more than about 30 nt, or modified or unmodified variants thereof.
[00581] A RNAi agent comprising a first and a second strand, wherein the sequence of the first strand comprises at least 14 contiguous nucleotides of the sequence of SEQ ID NO: 1 , and/or the sequence of the second strand comprises at least 14 contiguous nucleotides of the sequence of SEQ ID NO: 45, wherein the length of the first and second strand are each no more than about 30 nt, or modified or unmodified variants thereof.
[00582] A RNAi agent comprising a first and a second strand, wherein the sequence of the first strand comprises at least 14 contiguous nucleotides of the sequence of SEQ ID NO: 2, and/or the sequence of the second strand comprises at least 14 contiguous nucleotides of the sequence of SEQ ID NO: 100, wherein the length of the first and second strand are each no more than about 30 nt, or modified or unmodified variants thereof.
[00583] Additional particular specific aspects
[00584] In various aspects, the disclosure comprises a RNAi agent comprising a first and a second strand, wherein the sequence of the first strand comprises at least 14 contiguous nucleotides differing by 0, 1 , 2, or 3 nt from a first strand, and/or the sequence of the second strand comprises at least 14 contiguous nucleotides differing by 0, 1 , 2, or 3 nt from the second strand of any any one or more RNAi agent disclosed herein.
[00585] In various aspects, the disclosure encompasses a composition comprising any one or more of the following:
[00586] A RNAi agent comprising a first and a second strand, wherein the sequence of the first strand comprises at least 14 contiguous nucleotides differing by 0, 1 , 2, or 3 nt from the sequence of SEQ ID NO: 41 , and/or the sequence of the second strand comprises at least 14 contiguous nucleotides differing by 0, 1 , 2, or 3 nt from the sequence of SEQ ID NO: 9, wherein the length of the first and second strand are each no more than about 30 nt, or modified or unmodified variants thereof.
[00587] A RNAi agent comprising a first and a second strand, wherein the sequence of the first strand comprises at least 14 contiguous nucleotides differing by 0, 1 , 2, or 3 nt from the sequence of SEQ ID NO: 48, and/or the sequence of the second strand comprises at least 14 contiguous nucleotides differing by 0, 1 , 2, or 3 nt from the sequence of SEQ ID NO: 39, wherein the length of the first and second strand are each no more than about 30 nt, or modified or unmodified variants thereof. [00588] A RNAi agent comprising a first and a second strand, wherein the sequence of the first strand comprises at least 14 contiguous nucleotides differing by 0, 1 , 2, or 3 nt from the sequence of SEQ ID NO: 56, and/or the sequence of the second strand comprises at least 14 contiguous nucleotides differing by 0, 1 , 2, or 3 nt from the sequence of SEQ ID NO: 23, wherein the length of the first and second strand are each no more than about 30 nt, or modified or unmodified variants thereof.
[00589] A RNAi agent comprising a first and a second strand, wherein the sequence of the first strand comprises at least 14 contiguous nucleotides differing by 0, 1 , 2, or 3 nt from the sequence of SEQ ID NO: 102, and/or the sequence of the second strand comprises at least 14 contiguous nucleotides differing by 0, 1 , 2, or 3 nt from the sequence of SEQ ID NO: 81 , wherein the length of the first and second strand are each no more than about 30 nt, or modified or unmodified variants thereof.
[00590] A RNAi agent comprising a first and a second strand, wherein the sequence of the first strand comprises at least 14 contiguous nucleotides differing by 0, 1 , 2, or 3 nt from the sequence of SEQ ID NO: 1 , and/or the sequence of the second strand comprises at least 14 contiguous nucleotides differing by 0, 1 , 2, or 3 nt from the sequence of SEQ ID NO: 45, wherein the length of the first and second strand are each no more than about 30 nt, or modified or unmodified variants thereof.
[00591] A RNAi agent comprising a first and a second strand, wherein the sequence of the first strand comprises at least 14 contiguous nucleotides differing by 0, 1 , 2, or 3 nt from the sequence of SEQ ID NO: 2, and/or the sequence of the second strand comprises at least 14 contiguous nucleotides differing by 0, 1 , 2, or 3 nt from the sequence of SEQ ID NO: 100, wherein the length of the first and second strand are each no more than about 30 nt, or modified or unmodified variants thereof.
[00592] Additional particular aspects
[00593] In various aspects, the disclosure comprises a RNAi agent comprising a sense and an antisense strand, wherein the antisense strand comprises at least 14 contiguous nucleotides differing by 0, 1 , 2, or 3 nt from the antisense strand of any RNAi agent disclosed herein, or modified or unmodified variants thereof, wherein the antisense strand optionally further comprises 0, 1 , 2, 3, 4, 5, 6, 7, 8, 9, or 10 or more nt (or any range thereof, e.g., 0-1 , 1-2, 1 -3, 1-4 nt, etc.).
[00594] Thus, in various aspects, the disclosure comprises a RNAi agent comprising a sense and an antisense strand, wherein the antisense strand comprises at least 14 contiguous nucleotides differing by 0, 1 , 2, or 3 nt from any of: SEQ ID NOs in Table 2, or modified or unmodified variants thereof, wherein the antisense strand optionally further comprises 0, 1 , 2, 3, 4, 5, 6, 7, 8, 9, or 10 or more nt (or any range thereof, e.g., 0-1 , 1 -2, 1 - 3, 1-4 nt, etc.).
[00595] In one aspect, the disclosure comprises any one or more RNAi agent listed herein. [00596] Additional particular specific aspects
[00597] Other particular specific aspects include compositions comprising 1 , 2, 3, 4, or more of these RNAi agents. Another aspect is a composition comprising any single RNAi agent, along with any other RNAi agents which overlap it. Another aspect comprises two, three, four or more APOC3 RNAi agents which do not overlap and thus target different parts of the RNA molecule. When two or more RNAi agents are used, they can be administered simultaneously or sequentially.
[00598] Another particular specific aspect comprises an RNAi agent, wherein the RNAi agent comprises a sense strand comprising at least 14 contiguous nucleotides (identical in sequence) to the sense strand of any of the listed RNAi agents, and an antisense strand comprising at least 14 contiguous nucleotides (identical in sequence) to the antisense strand of the same RNAi agent. In another aspect, the composition comprises one, two, three, four, or more such RNAi agents.
[00599] In one aspect, the composition comprises an RNAi agent which comprises an antisense strand comprising at least 14 contiguous nucleotides differing by 0, 1 , 2 or 3 mismatches from the antisense strand of a RNAi agent described herein.
[00600] In one aspect, the composition comprises an RNAi agent which comprises an antisense strand comprising at least 14 contiguous nucleotides differing by 0, 1 , 2 or 3 mismatches from the antisense strand of a RNAi agent described herein.
[00601] In another aspect, the composition comprises an RNAi agent which comprises a sense strand comprising at least 14 contiguous nucleotides differing by 0, 1 , 2, or 3 mismatches from the sense strand of one of the listed RNAi agents, and an antisense strand comprising at least 14 contiguous nucleotides differing by 0, 1 , 2 or 3 mismatches from the antisense strand of the same RNAi agent.
[00602] A "mismatch" is defined herein as a difference between the base sequence or length when two sequences are maximally aligned and compared. As a non-limiting example, a mismatch is counted if a difference exists between the base at a particular location in one sequence and the base at the corresponding position in another sequence (e.g., between the sequence of a given RNAi agent and an RNAi agent listed herein). Thus, a mismatch is counted, for example, if a position in one sequence has a particular base (e.g., A), and the corresponding position on the other sequence has a different base (e.g., G, C or U). A mismatch is also counted, e.g., if a position in one sequence has a base (e.g., A), and the corresponding position on the other sequence has no base (e.g., that position is an abasic nucleotide which comprises a phosphate-sugar backbone but no base). A single- stranded nick in either sequence (or in the sense or antisense strand) is not counted as mismatch. Thus, as a non-limiting example, no mismatch would be counted if one sequence comprises the sequence A-G, but the other sequence comprises the sequence A-G with a single-stranded nick between the A and the G. A base modification is also not considered a mismatch. If one sequence comprises a C, and the other sequence comprises a modified C (e.g., with a 2'-modification) at the same position, no mismatch would be counted. Thus, modifications of a nucleotide other than replacement or alteration of the base would not constitute a mismatch. For example, no mismatch would occur between a nucleotide which is A, and a nucleotide which is A with a 5' modification and/or a 2'-modification. The key feature of a mismatch (base replacement) is that it would not be able to base-pair with the corresponding base on the opposite strand. In addition, terminal overhangs such as "UU" or "dTdT" are not counted when counting the number of mismatches; the terminal "UU" and "dTdT" overhangs are also not included when calculating "15 contiguous nucleotides."
[00603] In these aspects, a mismatch is defined as a position wherein the base of one sequence does not match the base of the other sequence.
[00604] In another aspect, the composition comprises 1 , 2, 3, 4, or more such RNAi agents.
[00605] In another aspect, the composition comprises an RNAi agent which comprises a sense strand comprising at least 14 contiguous nucleotides differing by 0, 1 , 2 or 3 mismatches from the sense strand of one of the listed RNAi agents, and an antisense strand comprising at least 14 contiguous nucleotides differing by 0, 1 , 2 or 3 mismatches from the antisense strand of the same RNAi agent
[00606] Variants of RNAi agents (e.g., comprising different modifications, caps, etc.) are disclosed herein, e.g., in the Tables. An unmodified and an example modified variant of various RNAi agents are provided. The disclosure thus encompasses groups of overlapping modified and/or unmodified RNAi agents. More aspects are provided herein, and are included in the scope of each RNAi agents of the disclosure.
[00607] Other modifications known to one skilled in the art are contemplated as being encompassed within the invention. Exemplary modifications include, but are not limited to, the presence of gaps or mismatches between the base pairs in the sense and antisense strands, the presence of nicks or breaks in the internucleoside linkages in the sense strand, and the like.
PHARMACEUTICAL COMPOSITIONS [00609] Compositions intended for oral use can be prepared according to any method known to the art for the manufacture of pharmaceutical compositions and such compositions can contain one or more such sweetening agents, flavoring agents, coloring agents or preservative agents in order to provide pharmaceutically elegant and palatable preparations. Tablets contain the active ingredient in admixture with non-toxic pharmaceutically acceptable excipients that are suitable for the manufacture of tablets. These excipients can be, for example, inert diluents; such as calcium carbonate, sodium carbonate, lactose, calcium phosphate or sodium phosphate; granulating and disintegrating agents, for example, corn starch, or alginic acid; binding agents, for example starch, gelatin or acacia; and lubricating agents, for example magnesium stearate, stearic acid or talc. The tablets can be uncoated or they can be coated by known techniques. Formulations for oral use can also be presented as hard gelatin capsules wherein the active ingredient is mixed with an inert solid diluent, for example, calcium carbonate, calcium phosphate or kaolin, or as soft gelatin capsules wherein the active ingredient is mixed with water or an oil medium, for example peanut oil, liquid paraffin or olive oil. Aqueous suspensions contain the active materials in a mixture with excipients suitable for the manufacture of aqueous suspensions.
[00610] Oral administration of the compositions of the invention include all standard techniques for administering substances directly to the stomach or gut, most importantly by patient controlled swallowing of the dosage form, but also by other mechanical and assisted means of such delivery.
[00611] Dosage levels of the order of from about 0.1 mg to about 140 mg per kilogram of body weight per day are useful in the treatment of the above- indicated conditions (about 0.5 mg to about 7 g per subject per day). The amount of active ingredient that can be combined with the carrier materials to produce a single dosage form varies depending upon the host treated and the particular mode of administration. Dosage unit forms generally contain between from about 1 mg to about 500 mg of an active ingredient. It is understood that the specific dose level for any particular subject depends upon a variety of factors including the activity of the specific compound employed, the age, body weight, general health, sex, diet, time of administration, route of administration, and rate of excretion, drug combination and the severity of the particular disease undergoing therapy.
[00612] Therapeutic effect of the therapeutic agents of the invention may be enhanced by combination with other agents. Typically such other agents will include agents known for use in treating similar diseases, such as angiogenic disorders.
[00613] The RNAi agents of the invention and formulations thereof can be administered orally, topically, parenterally, by inhalation or spray, or rectally in dosage unit formulations containing conventional non-toxic pharmaceutically acceptable carriers, adjuvants and/or vehicles. The term parenteral as used herein includes percutaneous, subcutaneous, intravascular (e.g., intravenous), intramuscular, intraperitoneal, or intrathecal injection, or infusion techniques and the like. Where two or more different RNAi agents are
administered, each may be administered separately or co-administered. Where each is adminitered separately, the method and/or site of adminstration may be the same or different, e.g., both RNAi agents may be administered intraveneously or subcutaneously, or a first RNAi agent may be administered intraveneously with a second Rai agent
administered subcutaneously, etc.
[00614] In various embodiments, the disclosure encompasses a composition or
pharmaceutical composition comprising a RNAi agent, wherein one or both strands comprises a 3' end cap, the composition further comprising a helper lipid, a neutral lipid, and/or a stealth lipid.
[00615] In one particular specific embodiment, the present disclosure relates to a method of treating a target gene-related disease in an individual, comprising the step of
administering to the individual a therapeutically effective amount of a composition comprising a RNAi agent comprising a first strand and a second strand, wherein the first and/or second strand comprise a 3' end cap selected from the 3' end caps listed in Table 5A-E. In one particular specific embodiment, the present disclosure relates to a method of inhibiting the expression of target gene in an individual, comprising the step of administering to the individual a therapeutically effective amount of a composition comprising a RNAi agent of the present disclosure.
[00616] In one embodiment of the method, the composition further comprises a
pharmaceutically effective formulation.
[00617] Various particular specific embodiments of these embodiments are described below.
[00618] In one embodiment, the method further comprises the administration of an additional treatment. In one embodiment, the additional treatment is a therapeutically effective amount of a composition.
[00619] In one embodiment, the additional treatment is a method (or procedure).
[00620] In one embodiment, the additional treatment and the RNAi agent can be administered in any order, or can be administered simultaneously.
[00621] In one embodiment, the method further comprises the step of administering an additional treatment for the disease.
[00622] In one embodiment, the method further comprises the step of administering an additional treatment or therapy selected from the list of an additional antagonist to a target gene-related disease. [00623] In one embodiment, the composition comprises a second RNAi agent to target gene. In various embodiments, the second RNAi agent is physically separate from the first, or the two are physically connected (e.g., covalently linked or otherwise conjugated).
[00624] Other embodiments
[00625] Various particular specific embodiments of this disclosure are described below.
[00626] In one embodiment, the disclosure pertains to a composition according to any of the embodiments described herein, for use in a method of treating a target gene-related disease in an individual, the method comprising the step of administering to the individual a therapeutically effective amount of a composition according to any of the claims.
[00627] One embodiment of the disclosure is the use of a composition according to any of these embodiments, in the manufacture of a medicament for treatment of an target gene- related disease.
[00628] In one embodiment, the disclosure pertains to the composition of any of the above embodiments, for use in the treatment of an target gene-related disease.
[00629] Additional Definitions
[00630] Unless defined otherwise, the technical and scientific terms used herein have the same meaning as that usually understood by a specialist familiar with the field to which the present disclosure belongs.
[00631] Unless indicated otherwise, all methods, steps, techniques and manipulations that are not specifically described in detail can be performed and have been performed in a manner known per se, as will be clear to the skilled person. Reference is for example again made to the standard handbooks and the general background art mentioned herein and to the further references cited therein.
[00632] Claims to the present disclosure are non-limiting and are provided below.
[00633] Although particular embodiments and claims have been disclosed herein in detail, this has been done by way of example for purposes of illustration only, and is not intended to be limiting with respect to the scope of the appended claims, or the scope of subject matter of claims of any corresponding future application. In particular, it is contemplated by the inventors that various substitutions, alterations, and modifications may be made to the present disclosure without departing from the spirit and scope of the present disclosure as defined by the claims. The choice of nucleic acid starting material, clone of interest, or library type is believed to be a matter of routine for a person of ordinary skill in the art with knowledge of the embodiments described herein or known in the art. Other aspects, advantages, and modifications considered to be within the scope of the following claims. Redrafting of claim scope in later-filed corresponding applications may be due to limitations by the patent laws of various countries and should not be interpreted as giving up subject matter of the claims.
[00634] Various additional formulations and obvious variants of the described 3' end caps can be devised by those of ordinary skill in the art. Non-limiting example RNAi agents wherein one or both strands comprises a 3' end cap are described in the Examples below, which do not limit the scope of the present disclosure as described in the claims.
EXAMPLES
[00635] Unless defined otherwise, the technical and scientific terms used herein have the same meaning as that usually understood by a specialist familiar with the field to which the disclosure belongs.
[00636] Unless indicated otherwise, all methods, steps, techniques and manipulations that are not specifically described in detail can be performed and have been performed in a manner known per se, as will be clear to the skilled person. Reference is for example again made to the standard handbooks and the general background art mentioned herein and to the further references cited therein. Unless indicated otherwise, each of the references cited herein is incorporated in its entirety by reference.
[00637] Claims to the invention are non-limiting and are provided below.
[00638] Although particular aspects and claims have been disclosed herein in detail, this has been done by way of example for purposes of illustration only, and is not intended to be limiting with respect to the scope of the appended claims, or the scope of subject matter of claims of any corresponding future application. In particular, it is contemplated by the inventors that various substitutions, alterations, and modifications may be made to the disclosure without departing from the spirit and scope of the disclosure as defined by the claims. The choice of nucleic acid starting material, clone of interest, or library type is believed to be a matter of routine for a person of ordinary skill in the art with knowledge of the aspects described herein. Other aspects, advantages, and modifications considered to be within the scope of the following claims. Those skilled in the art will recognize or be able to ascertain, using no more than routine experimentation, many equivalents of the specific aspects of the invention described herein. Such equivalents are intended to be
encompassed by the following claims. Redrafting of claim scope in later filed corresponding applications may be due to limitations by the patent laws of various countries and should not be interpreted as giving up subject matter of the claims.
[00663] Example 1: siRNA Lipid Formulations. The siRNAs of the present invention can be encapsulated in lipid nanoparticles as follows. The siRNA lipid nanoparticles were formed by mixing solutions of lipids dissolved in ethanol with siRNA dissolved in a citrate buffer as described generally above. Mixing chambers were used having passages with inner diameters of 0.5, 1 .0, or 2.0 mm. The processing chambers had lengths of from 50 mm to 1000 mm. The dilution chambers had passages with inner diameters equivalent to or of at least to 2 times that of the mixing chamber about 0.5 or 1 .0 or 2.0 or 4.0 mm. The lipid solution contained a cationic lipid, a helper lipid (cholesterol), a neutral lipid (DSPC) and a stealth lipid.
[00664] The concentrations of total lipids were either 16.7 mg/mL or 25 mg/mL. The total lipid to siRNA ratio for these experiments was about 18.3:1. The concentration of siRNA solutions were 0.225, 0.3, 0.3375, or 0.45 mg/mL in a sodium citrate: sodium chloride buffer with pH 5. The concentration of NaCI was 50 mM, 66 mM, 75 mM, or 100 mM. The flow rates and linear velocities were varied as described below.
[00665] For siRNA encapsulation experiments, the cationic lipids and stealth lipids (i.e., PEG lipid) used are shown in the Table below.
Table 4
Figure imgf000117_0001
[00666] The siRNA used for these experiments had sequences and SEQ ID NOs. as shown in Table below.
Table 5
Figure imgf000117_0002
Figure imgf000118_0002
[00667] Process Example 1 Encapsulation of siRNA
[00668] Preparation of lipid mixture in ethanol. The following is an example of siRNA encapsulated at a cationic lipid amine to siRNA phosphate (N:P) molar ratio of 4.5:1 . Lipids (cationic lipid, DSPC, cholesterol and lipidated PEG) in the amounts shown in Table 6 are dissolved in 150 ml. of ethanol. The molar ratios of lipids are 45:9:44:2, respectively. The mixture is sonicated briefly, then gently agitated for 5 minutes and then maintained at 37°C until use.
Table 6 Lipid mixture components
Figure imgf000118_0001
[00669] Preparation of siRNA in citrate buffer. The buffer for the siRNA streams is 100 mM sodium chloride and 25 mM sodium citrate at a pH of 5. The pH of the citrate buffer is first confirmed to be pH 5.00. If it is not, the pH is adjusted before proceeding. Enough siRNA in water for the encapsulation is thawed from -80°C storage. The siRNA is added to citrate buffer solution and the concentration of the dissolved siRNA is measured by optical density at 260 nm in a UV spectrophotometer. The final concentration of the siRNA is adjusted to 0.45 mg/ml in 150 ml of citrate buffer in a sterile PETG bottle and is held at room
temperature until use.
[00670] Encapsulation of siRNA in lipid nanoparticles. Sterile syringes are loaded with an equal volume (25 ml) of lipids in ethanol (syringe (a)), siRNA in citrate buffer (syringes (b1 ) and (b2)), and water alone (syringe (c)). Tubing leading from Luer fittings on the syringe (a) containing 16.7 mg/mL lipids is attached to the center input of a cross junction with a 0.5 mm inner diameter. Tubing leading from Luer fittings on syringes (b1 ) and (b2) containing siRNA at 0.45 mg/ml are attached to the side inputs of the cross junction. The tubing is fluorinated ethylene propylene (FEP) tubing with 1.55 mm inner diameter. Syringes (a), (b1 ) and (b2) are installed on syringe pump A. Tubing leading from the center input of the cross opposite the lipid input is attached to a T junction with a 1 mm inner diameter. Tubing leading from a Luer fitting on syringe (c) containing water alone is attached to the T junction to enable in- line dilution of siRNA lipid nanoparticles, and syringe (c) is installed on syringe pump B. Have the output line from the T junction positioned over a sterile PETG bottle for collection of the diluted siRNA lipid nanoparticles. It is important to make sure that all fittings are tight on the syringes. The syringe pumps are set to the appropriate syringe manufacturer and size and a flow rate of 40 ml per minute. Start both pumps simultaneously, and start collecting material after approximately 0.5 seconds. Approximately 90 ml of encapsulated siRNA lipid nanoparticles will be collected and which contains 25% ethanol by volume, 0.23 mg/mL of the siRNA, 4.2 mg/mL of the lipids, and 50 mM NaCI. After a 60 min. post mixing incubation period, particle sizes are determined using a Malvern Zetasizer.
[00671] Dilution of siRNA lipid nanoparticles. A 1 .0 mm inner diameter T junction is set up for further dilution of the siRNA lipid nanoparticle suspension. This dilution step is run with two syringes on one syringe pump. One 140 mL syringe contains the siRNA lipid
nanoparticles and a second 140 mL syringe contains water. The flow rate is set to 25 ml per minute. The siRNA stream and the water stream enter the T junction at 180 degrees from each other. The diluted siRNA lipid nanoparticle suspension is collected into a sterile PETG bottle. The final volume will be approximately 280 ml, with an ethanol concentration of 12.5%.
[00672] Dialysis and concentration of siRNA lipid nanoparticles by tangential flow filtration. For every 50 mg of siRNA in the encapsulation run, use a Vivaflow 50 cartridge. For a 100 mg siRNA encapsulation run, use 2 Vivaflow 50 cartridges attached in series. The regenerated cellulose cartridges is first rinsed to remove any storage solution from the manufacturer. This is done by filling an empty TFF reservoir with 500 ml of Dl water and recirculate with a peristaltic pump at a flow rate of 1 15ml/min. The permeate line should not be restricted and the rinsing process is complete when the entire 500ml of water is flushed through the membrane.
[00673] Load the siRNA lipid nanoparticle suspension into the Minimate TFF reservoir. Concentrate the mixture while maintaining an overall pressure of 20-25 psi. The filtrate should elute at approximately 4 ml per minute throughout the concentration step. This rate is achieved by restricting the permeate line with a pinch valve until the proper flow rate is achieved. Concentrate until the liquid level in the reservoir is at the 15 ml graduation.
Diafilter the concentrated siRNA lipid nanoparticle suspension against 225 ml of pyrogen- free, nuclease-free 1x PBS. Increase the flow rate to 80ml/min. After diafiltration, resume concentration of the material to the holdup volume of the TFF system. Collect the siRNA lipid nanoparticle suspension from the reservoir. It is possible to rinse the TFF system with additional 2 ml of 1x PBS and to collect this wash that contains diluted siRNA lipid nanoparticle suspension, but this wash should be collected separately from the concentrated siRNA lipid nanoparticle suspension. Store materials at 4°C until analysis.
[00674] Sterile filtration step. The siRNA lipid nanoparticles are filtered by heating approximately 10ml of the suspension in a glass vial which is placed in a aluminum block heater preheated to 50 °C for 10min. The vial is then removed and the solution is removed with a syringe and filtered through a 0.22 μηη PES syringe filter directly into a sterile vial. This final product is stored at 4 °C.
[00675] Percent encapsulation determination (SYBR GOLD). To determine the efficiency of the siRNA formulation into lipid nanoparticles, the percent encapsulation of the siRNA can be determined by measuring sybr gold fluorescence. When bound to siRNA, sybr gold fluoresces. The intensity of sybr gold fluorescence is proportional to the amount of siRNA.
[00676] A standard solution of siRNA stock at approximately 0.9 mg/mL is prepared in PBS. The concentration of siRNA stock is verified by UV measurement. The siRNA stock is diluted with PBS to 8 μg/mL. Serial dilution is done to prepare 4, 2, 1 , 0.5 and 0.25μg/mL siRNA solutions. 6μg/mL of siRNA is prepared by mixing equal volumes of 8μg/mL and 4μg/mL solutions.
[00677] To prepare test samples, 10 μΙ_ of siRNA lipid nanoparticle suspension are diluted with 990 μΙ_ of PBS (this is now solution A). Note: This first dilution step applies for the formulations with expected siRNA concentration of -3.6 mg/mL or less. If the concentration is higher than ~3.6mg/ml_, the dilution should be greater. 40uL of solution A is diluted with 160 μΙ_ of PBS (this is now solution 1 ).
[00678] For the measurement of free siRNA in a formulation, a solution of 0.02% sybr gold in PBS is prepared (e.g., 3μΙ_ of sybr gold in 15ml_ of PBS) (solution 2). In a 96-well black, clear-bottom plate 10 μΙ_ of solution 1 is mixed with 190 μΙ_ of solution 2 to provide sample mixture 1. In this mix, sybr gold will bind only to the nonencapsulated (i.e. free) siRNA. It will not have access to the siRNA encapsulated in the liposome.
[00679] For the measurement of total siRNA in a formulation, a solution of 0.02% sybr gold and 0.2% triton-x in PBS is prepared (e.g., 3 μΙ_ of sybr gold in 15 mL of 0.2% triton in PBS) (solution 3). In a 96-well black, clear-bottom plate 10 μΙ_ of solution 1 is mixed with 190 μΙ_ of solution 3 to provide sample mixture 2. In this mix, triton-x disrupts the liposomes and exposes previously encapsulated siRNA to sybr gold binding. Hence, sybr gold will bind to the non-encapsulated (i.e. free) siRNA and to all newly exposed siRNA. The free
siRNA+newly exposed siRNA = total siRNA [00680] The standard solutions described above (10 μΙ_ each) are mixed with either 190 μΙ_ solution 2 to provide standard mixtures 1 or 190 μΙ_ solution 3 to provide standard mixtures 2.
[00681] The fluorescence of all mixes is measured on the SpectraMax M5
spectrophotometer using software SoftMax pro 5.2 and the following parameters:
Figure imgf000121_0001
[00682] The fluorescence intensity values obtained from standard mixtures 1 are used to create the calibration curve for free siRNA. The fluorescence intensity of a sample 1 mixture is then plugged into the equation provided by the calibration curve for free siRNA. The found concentration of the sample is then multiplied by the dilution magnitude to obtain the free siRNA in the lipid nanoparticle formulation.
[00683] The fluorescence intensity values obtained from standard mixtures 2 are used to create the calibration curve for total siRNA. The fluorescence intensity of a sample 2 mixture is then plugged into the equation provided by the calibration curve for total siRNA. The found concentration of the sample is then multiplied by the dilution magnitude to obtain the total siRNA in the lipid nanoparticle formulation.
[00684] The encapsulated siRNA is calculated by the formula:[(total siRNA - free siRNA) / (total siRNA)] x 100%.
[00685] Percent encapsulation determination using Size Exclusion Chromatography (SEC). Because the size of free siRNA (5 nm) is different than the size of a liposome (50- 200nm), they will elute at different times in the size exclusion column. Free siRNA elutes after the liposomes. Retention time for siRNA is -17 minutes whereas the retention time for liposomes is ~10 minutes. Detection of eluted siRNA is carried out via UV detector with absorption wavelength set at 260 nm.
[00686] siRNA stock at approximately 0.9 mg/mL is prepared in PBS. To a 200 μΙ_ aliquot of stock, 10μΙ_ of TRITON X-100 are added. The concentration of siRNA stock is verified by UV measurement. Serial dilutions are done to prepare standards at 1/2, 1/4, 1/8, 1/16 and 1/32 concentration of the siRNA stock. 10 μΙ_ of TRITON X-100 are added to 200 μΙ_ of each standard. The concentration of each standard is verified by UV measurement.
[00687] In a HPLC vial, 25μΙ_ of lipid nanoparticle formula are added to 185 μΙ_ of 1 X PBS (10X PBS (FISHER, BP399) diluted with deionized water to1 X). The dispersion is gently vortexed until homogeneous (dispersion 1 ). In another HPLC vial, 25μΙ_ of lipid nanoparticle formula are added to 185 μΙ_ of 20% TRITON-X. The dispersion is gently vortexed until clear and homogeneous (dispersion 2).
[00688] The size exclusion chromatography is performed on an AGILENT 1200 HPLC using EMPOWER PRO software. The parameters are:
Figure imgf000122_0001
[00689] 20 L of standards and dispersions are injected onto size exclusion column, mobile phase 1 x PBS with pH adjusted to 7.7 flowing at 1 mL/min for 30 minutes. The eluted material is detected by UV detector with 260 nm absorption wavelength.
[00690] From the siRNA standards, the peak at -17 minutes represents the siRNA. From dispersion 1 , the peak at -10 minutes represents the lipid nanoparticle containing encapsulated siRNA and the peak at -17 minutes represents the nonencapsulated (i.e. free) siRNA.
[00691] In dispersion 2, TRITON-X disrupts the lipid nanoparticle enabling previously encapsulated siRNA to elute together with already free siRNA at 17 minutes. The peak at -10 minutes disappears, and only one peak in the chromatogram remains, i.e. the peak at -17 minutes, representing both non-encapsulated (i.e. free) siRNA and newly free siRNA . Free siRNA + newly free siRNA = total siRNA.
[00692] The peak area values obtained from siRNA standards are used to create the siRNA concentration calibration curve. The integrated area of the peak at -17 minutes in dipersion 1 is plugged into the equation provided by calibration curve for free siRNA to obtain the concentration of the free siRNA in the dispersion. The found concentration of the sample is then multiplied by the dilution magnitude to obtain the free siRNA in the lipid nanoparticle formulation.
[00693] The integrated area of the peak at -17 minutes in dispersion 2 is plugged into the equation provided by calibration curve for free siRNA to obtain the concentration of the total siRNA in the dispersion. The found concentration of sample is then multiplied by the dilution magnitude to obtain the total siRNA in the lipid nanoparticle formulation. [00694] Encapsulated siRNA is calculated by formula: [(total siRNA - free siRNA) / (total siRNA)] x 100%.
[00695] Particle analytics. The siRNA lipid nanoparticles are analyzed for size and polydispersity using a Zetasizer Nano ZS from Malvern Instruments. For formulated siRNA at an encapsulated siRNA concentration of > 1 mg/ml, dilute 5 μΙ of sample with 1 15 μΙ of 1x PBS. Add to a small volume disposable micro-cuvette. Insert the cuvette into the Zetasizer Nano ZS. For the machine settings, set material to be polystyrene latex, dispersant to be water, and cell to be ZEN040. Measure the sample at 25°C with no wait time. Record Z-Ave (set as diameter, in nanometer units) and polydispersity index (PDI).
[00696] In Table 7 are shown the results obtained for siRNA and lipid nanoparticle combinations using the procedures described above. Encapsulation percentages were determined using the SEC method.
Table 7 siRNA Li id Nano article Enca sulation Results
Figure imgf000123_0001
[00697] Process Example 2: Effect of dilution. Using the system substantially as described herein, two aqueous nucleic acids streams having concentrations of 0.45 mg/mL of siRNA, 100 mM NaCI, and 25 mM sodium citrate at a pH of 5 were introduced from opposing directions into a cross-shaped mixing chamber having passages with inner diameters of 0.5 mm with flow rates of 40 mL/min each. Simultaneously, a lipid stream was introduced into the cross-shaped mixing chamber from a direction orthogonal to the two nucleic acid streams at a flow rate of 40 mL/min. The lipid stream was made up of 45% cationic lipid A1 , 44% cholesterol, 9% DSPC, and 2% PEG-lipid B1 in ethanol. The total concentration of lipids was 16.7 mg/mL. In one run, the joined stream from the cross-shaped mixing chamber was subjected to a dilution step with water using a T-shaped chamber (1.0 mm inner diameter).
[00698] The water was introduced into the T-shaped chamber at a rate of 40 mL/min. The resultant solution was collected and the encapsulated nucleic acid nanoparticles analyzed for size and uniformity with the results shown in entry 1 in Table 8. The solution obtained from entry 1 included 25% ethanol by volume, 0.23 mg/mL of the siRNA, 4.2 mg/mL of the lipids, and 50 mM NaCl.ln a separate experiment using the same concentrations, flow rates, and initial cross-shaped mixing chamber, the joined streams from the mixing chamber were diluted in a volume of water equivalent to the dilution volume used in entry 1 . These results are shown in entry 2 in Table 8. The concentrations of the collected solution were the same as in entry 1 . For comparison, another experiment was conducted under the same process conditions but without any dilution step. These results are shown in entry 3 in Table 6. Without any dilution step, the collected solution in entry 3 included 33% ethanol, 0.3 mg/mL siRNA, 5.6 mg/mL lipids and 66 mM NaCI. The Z-Avg, #-Avg, and PDI for entries 1 and 2 were less than entry 3, which lacked the dilution step. The Z-Avg, #-Avg, and PDI in Table were determined 60 minutes post-mixing.
Figure imgf000124_0001
[00699] Uniformly-sized lipid nanoparticles were produced. Replacing the cross-shaped mixing chamber with a T-shaped mixing chamber (0.5 mm inner diameter) while increasing the concentration of siRNA to 0.9 mg/mL at a flow rate of 40 mg/mL (same total amount of siRNA in one-half the volume) produced the less uniformly-sized lipid nanoparticles.
[00700] Process Example 3 Effect of flow rates, velocities, and solution concentrations. In a series of experiments using a T-shaped mixing chamber and having an inner diameter of 0.5 mm, a single nucleic acid stream and a single lipid stream were mixed at various flow rates/velocities and concentrations. The nucleic acid and lipid streams included the same constituents as described above in Process Example 2. No dilution step was used in these examples since the initial siRNA concentrations were adjusted to obtain a final solution concentration the same as entry 1 in Table 8. The concentration of lipids in ethanol in these experiments was either 16.7 mg/mL (1x) or 25 mg/mL (1 .5x). The results are shown in Table 9 below with the Z-Avg, #-Avg, and PDI determined 60 minutes post-mixing.
Figure imgf000124_0002
Figure imgf000125_0002
[00701] Process Example 4: Effect of flow rates and velocities. In a series of experiments using a T-shaped mixing chamber, a single nucleic acid stream and a single lipid stream were mixed from opposing directions at various flow rates/linear velocities and subsequently diluted with water at a 1 mm inner diameter dilution tee. The siRNA, cationic lipid A1 , PEG lipid B1 , cholesterol, and DSPC were used in Process Example 4 in amounts as described above. The total concentration of lipids in the ethanol stream was 16.7 mg/mL. The concentration of siRNA in the nucleic acid stream was 0.9 mg/mL. The results in Table 10 show that increasing linear velocity decreases the particle size and the PDI.
Figure imgf000125_0001
[00702] Process Example 5: Effect of orientation of T-shaped mixing chamber. Table 1 1 shows the results from two experiments using an alternate mixing chamber (0.5 mm inner diameter). In entry 1 are shown the results obtained where the siRNA enters from the branch and in entry 2 are shown the results obtained where the lipids enter from the branch. For both experiments, the siRNA and lipids were the same as those described above in Process Example 2. The flow rate for the siRNA streams was 120 mL/min and the flow rate of the lipid streams was 40 mL/min. The siRNA concentration was 0.3 mg/mL in a buffer solution containing 66 mM NaCI. The concentration of the lipids was 16.7 mg/mL. The Z- Avg, #-Avg, and PDI were determined 60 minutes post-mixing.
Table 1 1
Figure imgf000126_0001
[00703] Process Example 6: Effect of mixing chamber configurations. A series of experiments were conducted using mixing chambers having various configurations of nucleic acid and lipid streams with the total number of streams ranging from 3 to 6. In each case the mixing chamber had a 1 mm inner diameter chamber and the flow rates were adjusted to maintain the combined flow of 240 mL/min for the siRNA streams and 80 mL/min for the lipid streams. The concentration of siRNA in these experiments was 0.3 mg/mL and the concentration of lipids was 16.7 mg/mL. The linear velocity of the combined siRNA streams was 5.1 meters/second. The linear velocity of the combined lipid streams was 1.7 meters/second. Where the number of streams allows for more than one arrangement of streams, Table 1 1 indicates the angle between lipid or siRNA streams. For example, in the case of three lipid streams and three siRNA streams, each lipid stream is separated from the next lipid stream by either 60 degrees (i.e. ,3 adjacent lipid streams) or 120 degrees (i.e., lipid streams separated by 120 degrees with intervening siRNA streams also separated by 120 degrees). For the experimental results summarized in Table 1 1 , siRNA was used with cationic lipid A4, DSPC, cholesterol, and PEG lipid B1 , in a ratio of 45:9:44:2. The results shown in Table 1 1 indicate that substantially the same results are obtained for the various mixing configurations where the total flow rates/velocities remained constant.
Figure imgf000126_0002
Example 2: Conjugation of a fatty acid to Apoc 3-siRNA
Figure imgf000127_0001
Figure imgf000127_0002
Figure imgf000128_0001

Figure imgf000129_0001
[00704] Preparation of 24b: To a solution of 24a (1.244g, 4.19mmol) in 25ml DCM, TEA (0.58ml, 4.19mmol) was added, followed by N3-pentanoic acid (500mg, 3.49mmol). EDC (804mg, 4.19mmol) was added at last. The reaction stirred at room temperature for 4hr. The reaction was extracted between brine and DCM. Combined all organics, dried, concentrated and purified over Si02 gel with 60% ethylacetate/heptane to afford 1 .20g compound 24b (89% yield) . 1H NMR (CHLOROFORM-d, 400NHz) d: 5.88-6.37 (m, 1 H), 4.60 (d, J=4.8 Hz, 2H), 3.77 (s, 3H), 3.33 (t, J=6.7 Hz, 2H), 3.13 (d, J=6.5 Hz, 2H), 2.30 (t, J=7.2 Hz, 2H), 1.81- 1.95 (m, 1 H), 1 .63-1.81 (m, 5H), 1.48-1.57 (m, 2H), 1.46 (s, 9H), 1 .36 (d, J=6.8 Hz, 2H)
[00705] Preparation of 24c: To a solution of 24b (860mg, 2.23mmol) in 12ml THF, 1 N NaOH (5.58ml, 5.58mmol) was added. The reaction stirred at room temperature for 0.5hr. The reaction was diluted with brine and 20ml DCM was added. The pH of aqueous layer was adjusted to pH ~5 with 1 N HCI, then extracted with DCM. Combined all organics, dried, concentrated and and the crude solid was redissolved into 6ml DCM. 0.6ml TFA was added and the reaction stirred at room temperature for 2hr. The reaction was concentrated and afforded the crude 24c (400mg, 54%), which was used directly without further purification. Under LC-MS method I, the product showed a major peak at 0.43min. with a mass of 272.5 (M+H+).
[00706] Preparation of 24e: To a solution of 24c (400mg, 1 .04mmol) in 10ml DCM, TEA (0.434ml, 3.1 1 mmol) was added, followed by the addition of 24d (538mg, 1 .35mmol). The reaction stirred at room temperature for 3hr. The reaction was extracted between H20 and DCM. Combined all organics, dried, concentrated and purified over Si02 gel with 8%
MeOH/DCM to afford 475mg of 24e (82% yield). 1 H NMR (CHLOROFORM-d, 400MHz) d: 6.74-6.98 (m, 1 H), 5.95-6.13 (m, 1 H), 4.55 (dd, J=7.2, 2.4 Hz, 2H), 3.32 (t, J=6.7 Hz, 4H), 3.1 1 (d, J=5.8 Hz, 2H), 2.30-2.37 (m, 2H), 2.20-2.26 (m, 2H), 1 .90 (br. s., 2H), 1 .71-1.80 (m, 2H), 1 .59-1.71 (m, 4H), 1.51-1 .59 (m, 2H), 1.35-1 .51 (m, 13H), 1.20-1 .35 (m, 13H)
[00707] Preparation of 24f
Figure imgf000130_0001
[00708] Preparation of intermediate 24f2: To a solution of 24f1 (1.0g, 4.97mmol) in MeOH (40ml), TEA (1 .04ml, 7.45mmol) was added, followed by di t-butyl dicarbonate (2.17g, 9.94mmol). The reaction was heated at 60oC for 1 .5hr. The reaction was concentrated and purified over Si02 column with 5% MeOH/DCM to afford 1 .2g compound 24f2 (80% yield). 1H NMR (CHLOROFORM-d, 400MHz) d: 4.51 (br. s, 1 H), 3.00-3.22 (m, 2H), 2.37 (t, J=7.4 Hz, 2H), 1.59-1 .71 (m, 2H), 1 .46 (s, 1 1 H), 1.29 (br. s., 12H).
[00709] Preparation of intermediate 24f3: To a solution of 24f2 (1.2g, 3.98mmol) in DCM (30ml), N-hydroxyl scuccinimide (0.60g, 5.18mmol) was added, followed by EDC (1.0g, 5.18mmol). The solution was stirred at room temperature for overnight. The reaction was conentrated, directly loaded onto Si02 column and purified with 40% ethylacetate/heptane to afford 1.46g compound 24f3 (92% yield). 1 H NMR (CHLOROFORM-d, 400MHz) d: 4.49 (br. s, 1 H), 3.04-3.19 (m, 2H), 2.86 (d, J=4.5 Hz, 4H), 2.62 (t, J=7.4 Hz, 2H), 1 .70-1.82 (m, 2H), 1.37-1 .53 (m, 13H), 1 .30 (br. s., 10H)
[00710] Preparation of intermediate 24f: To a solution of GalNAc3-NH2 (300mg, 0.16mmol) in DCM (1.5ml), TEA (0.1 1 ml, 0.79mmol) was added, followed by the addition of 24f3
(188mg, 0.47mmol). The reaction stirred at room temperature for overnight. Then
trifluoroacetic acid (1 .0ml) was added. After 2hrs, LC-MS showed the disappearance of the intermediate. The reaction was concentrated and purified on open access HPLC under acidic condition with ELSD as a detection. The HPLC fractions containing the product were collected and the solvent was evaporated to afford 220mg compound 24f (67% yield). LC- MS showed that partial product lost one acetyl group. HPLC conditions for purification: column: Sunfire 30x100mm 5um column; organic solvent: ACN w/ 7.5% TFA; aqueous solvent: H20 w/ 7.5% TFA; flow rate: 75ml/min. Gradient: 15-40% H20/AcCN; Time: 9.5min. detection: ELSD (Evaporative Light Scattering Detector) as detection. Under LC-MS method I, the product showed a peak at 0.83min. with a mass of 989.9 (M/2+H+).
[00711] Preparation of 24g: To a solution of 24e (172mg, 0.31 mmol) in 3ml DCM, 24f
(250mg, 0.12mmol) was added, followed by TEA (0.069ml, 0.50mmol). EDC (95mg, 0.5mmol) was added at last. The reaction stirred at room temperature for ovenright. The reaction was concentrated and purified over Si02 column with 5% MeOH/DCM to afford 230mg 24g (74% yield). Under LC-MS method I, the product showed a peak 1.30min. with a mass of 1258.2 (M/2+H+).
[00712] Preparation of 24h: To a solution of 24g (230mg, 0.091 mmol) in 1 ml THF, 0.457ml 4N HCI (in dioxane, 1 .83mmol) was added. The reaction stirred at room temperature for 1 hr. The reaction was concentrated and purified over HPLC to afford 50mg 24h (23% yield), which lost one acetyl group on the sugar. HPLC conditions for purification: column: Sunfire 30x100mm 5um column; organic solvent: ACN w/ 7.5% TFA; aqueous solvent: H20 w/ 7.5% TFA; flow rate: 75ml/min. Gradient: 15-40% H20/AcCN; Time: 9.5min. detection: ELSD (Evaporative Light Scattering Detector) as detection. Under LC-MS method I, the product showed a peak at 0.95min. with a mass of 1 187.1 (M/2+ H+).
[00713] Preparation of 24i: To a solution of 2,2, 13, 13-tetramethyltetradecanedioic acid (40mg, 0.127mmol) in 2ml DCM, N-OH succinimide (9.81 mg, 0.085mmol) was added, followed by the addition of EDC (16.34mg, 0.085mmol). The reaction was stirred at room temperature for overnight. The reaction was concentrated and purified over HPLC to afford 20mg 24i (38% yield). HPLC conditions for purification: column: Sunfire 30x100mm 5um column; organic solvent: ACN w/ 7.5% TFA; aqueous solvent: H20 w/ 7.5% TFA; flow rate: 75ml/min. Gradient: 45-70% H20/AcCN; Time: 9.5min. detection: ELSD (Evaporative Light Scattering Detector) as detection. Under LC-MS method I, the product showed a peak at 1.55 min. with a mass of 434.3 (M+Na+).
[00714] Preparation of 24j: To a solution of 24h (20mg, 8.05μηιοΙ) in 0.5ml DCM, TEA (4.5 I, 32μηιοΙ) was added, followed by the addition of 24i (6.62mg, 16μηιοΙ) and DMAP (3.93mg, 32μηιοΙ). The reaction stirred at room temperature for overnight. Then 0.5ml 2N MeNH2/MeOH was added for the deprotection. The reaction stirred at room temperature for another overnight. The reaction was concentrated and acetone was added to precipitate the product and remove excess reagents and lipids to afforded 12mg 24j (64% yield). Under LC- MS method I, the product showed a peak at 0.69min. with a mass of 1 166.9 (M/2+ H+).
[00715] Preparation of 24K
Figure imgf000132_0001
[00716] Step 1 :
[00717] Sequence of APOCIII siRNA (Synthesized using conventional methods known in the art):
[00718] Antisense strand: AGCACuGAGAAuACuGuc-rib-X058. (SEQ ID NO: 14). wherein:
A is DNA, uppercase letters indicate non-modified nucleotide, lowercase letters indicate a 2'- OMe modified nucleotide, italics letters indicate a 2'-MOE modified nucleotide, rib is ribitol, and X058 is a non-nucleotidic linker of Formula:
Figure imgf000132_0002
[00719] Sense Strand: GAcAGuAuucucAGuGcu-rib-C60H. (SEQ ID NO:71 ). [00720] APOCIII siRNA was reacted with with 2-Dimethoxytrityloxymethyl-6- fluorenylmethoxycarbonylamino-hexane-1 -succinoyl-long chain alkylamino-CPG (Glen Research Catalog No 20-2957) to generate product 24k1.
[00721] Step 2: APOCIII siRNA (24k1 : 401 μΙ_, 1 1.2mM in H20, 4.49 μΓηοΙ) was mixed with 401 I DMF to get a clear solution. Then TEA (180 μΙ, 0.25M in DMF, 45 μιηοΙ) was added, followed by BCN-NHS ((1 R,8S,9s)-Bicyclo[6.1 .0]non-4-yn-9-ylmethyl N-succinimidyl carbonate) (9.16mg, 31 μηηοΙ). The reaction stirred at room temperature for 1 hr. The reaction was diluted with H20 to 10ml and extracted with ethylacetate 3times. The aqueous layer was seperated and concentrated to ~5ml and purified over PD-10 desalting column (GE healthcare).
[00722] Preparation of 24: 24j (5.8mg, 2.5μηιοΙ) was added into 105μΙ compound 24k (Apoc 3 siRNA 9.5mM in H20, 0.993μηιοΙ). After 1 hr, the reaction became viscous. So another 60μΙ H20 was added and the reaction stirred at room temperature for overnight. The reaction was diluted with H20 and purified over HPLC to afford 5mg conjugate 24 (57% yield). HPLC conditions for purification: Column: Xselect Prep phenylhexyl 5um OBD 19x50mm; organic solvent: AcCN modified with 100mM TEA.HOAc; aqueous solvent: H20 modified with 100mM TEA.HOAc; Gradient: 5-50% AcCN/H20; Time: 10min. Under LC-MS method H, the product showed a peak at 5.96min. with the desired mass of 8896 after deconvolution.
Reference Example 3: APOC3 siRNA conjugated with GalNAc
Figure imgf000134_0001
[00723] Reference Example 3 was prepared according to procedure of Example 24 (replacing 24j with GalNac3-N3 (below).
Figure imgf000134_0002
[00725] Preparation of intermediate 2: Compound 1 (2.06 g, 1.07 mmol) was dissiolved in 20 ml ethanol, TFA (82ul, 1 .07mmol) was added, followed by 10% Pd/C (0.1 14g, 0.1 1 mmol). The reaction was treated under H2 balloon for 6 hours. The reaction was filtered, washed with ethanol and concentrated to get white solid, which was used directly for next step.
Under LC-MS method I, the product showed a peak at 0.81 min. with a mass of 898.5 (M/2+H+).
[00726] Preparation of intermediate 3: Compound 2 (4.848 g, 0.479 mmol) was dissiolved in 10 ml anhdrous DMF, 2,5-dioxopyrrolidin-1-yl-4 azidobutanoate (0.325 g, 1 .436 mmol) was added, followed by the addition of DIPEA (0.418 ml, 2.394mmol). The reaction was to react overnight at room temperature. The reaction was concentrated with no heating, directly loaded onto a pre-equilibrated Si02 column and purified with 0-20% methanol/DCM step gradient to afford 2.383g compound 3 (49.25% yield).
[00727] Under LC-MS method I, the product showed a peak at 1.27min. with a mass of 953.7 (M/2+H+).
[00728] Preparation of intermediate 4: Mixed compound 3 (1.33g, 0.70mmol) with MeNH2 (17.45ml, 2.0M in Methanol, 34.9mmol). The reaction stirred at room temperature for 2hr. LC-MS only showed the product peak. The reaction was concentrated. Then the solid redissolved into ethanol and was precipitated with acetone to afford 1.0g compound 4 (94% yield). Under LC-MS method I, the product showed a peak at 0.53min. with a mass of 764.5 (M/2+H+).

Claims

We claim:
1. A composition comprising an RNAi agent comprising a sense strand and an antisense strand, wherein the antisense strand comprises at least 14 contiguous nucleotides differing by 0, 1 , 2, or 3 nucleotides from the antisense strand of an RNAi agent to APOC3 provided in any of Tables 2 and 3 herein, wherein the sense and/or the antisense strand is less than about 49 nucleotides in length.
2. The composition of claim 1 , wherein 3' end of the sense and/or antisense strand terminates in a phosphate or modified internucleoside linker and optionally further comprises a 3' end cap.
3. The composition of claim 1 , wherein the sense strand and the antisense strand form a duplex region about 15 to about 30 nucleotide pairs in length, or about 18 to about 23 nucleotides in length.
4. The composition of claim 1 , wherein one or more nucleoside of the sense or antisense strand has been replaced by a spacer.
5. The composition of claim 1 , wherein the sense strand is a 14-, 15-, 16- or 17-mer.
6. The composition of claim 1 , wherein the RNAi agent comprises a modification that causes the RNAi agent to have increased stability in a biological sample or environment.
7. The composition of claim 1 , wherein the RNAi agent comprises a modified sugar, a modified phosphorothioate linkage, or a 2'-modified nucleotide.
8. The composition of claim 1 , wherein the 3' end of the sense and/or antisense strand terminates in a phosphate or modified internucleoside linker and further comprises, in 5' to 3' order: a spacer, a second phosphate or modified internucleoside linker, and a 3' end cap.
9. The composition of claim 8, wherein the spacer is selected from a sugar, alkyl, cycloalkyl, ribitol, 2'-deoxy-ribitol, diribitol, 2'-methoxyethoxy-ribitol, C3, C4, C5, C6, or 4- methoxybutane-1 ,3-diol.
10. The composition of claim 8, the modified internucleoside linker is selected from phosphorothioate, phosphorodithioate, phosphoramidate, boranophosphonoate, an amide linker, and a compound of formula (I):
Figure imgf000137_0001
(I), where R3 is selected from O", S", NH2, BH3, CH3, C1-6 alkyl, C6-io aryl, Ci-6 alkoxy and C6-io aryl-oxy, wherein Ci_6 alkyl and C6-io aryl are unsubstituted or optionally independently substituted with 1 to 3 groups independently selected from halo, hydroxyl and NH2; and R4 is selected from O, S, NH, and CH2.
11. The composition of claim 1 , wherein the RNAi agent is ligated to one or more diagnostic compound, reporter group, cross-linking agent, nuclease-resistance conferring moiety, natural or unusual nucleobase, lipophilic molecule, cholesterol, lipid, lectin, steroid, uvaol, hecigenin, diosgenin, terpene, triterpene, sarsasapogenin, Friedelin, epifriedelanol- derivatized lithocholic acid, vitamin, carbohydrate, dextran, pullulan, chitin, chitosan, synthetic carbohydrate, oligo lactate 15-mer, natural polymer, low- or medium-molecular weight polymer, inulin, cyclodextrin, hyaluronic acid, protein, protein-binding agent, integrin- targeting molecule, polycationic, peptide, polyamine, peptide mimic, and/or transferrin.
12. A method of treating an APOC3-related disease in an individual, comprising the step of administering to the individual a therapeutically effective amount of a composition according to claim 1.
13. The method of claim 12, wherein the APOC3-related disease is Type V
Hypertriglyceridemia.
14. The method of claim 12, wherein the APOC3-related disease is hypertriglyceridemia (e.g., Type V Hypertriglyceridemia), abnormal lipid metabolism, abnormal cholesterol metabolism, atherosclerosis, hyperlipidemia, diabetes, including Type 2 diabetes, obesity, cardiovascular disease, and coronary artery disease, among other disorders relating to abnormal metabolism or otherwise.
15. The method of claim 12, wherein the method further comprises the step of administering an additional treatment.
16. The method of claim 1 , wherein the composition comprises a second RNAi agent to APOC3.
17. The composition of claim 1 , for use in the treatment of an APOC3-related disease.
PCT/US2015/040517 2014-07-16 2015-07-15 Organic compositions to treat apoc3-related diseases WO2016011123A1 (en)

Priority Applications (4)

Application Number Priority Date Filing Date Title
EP20178547.4A EP3736334A1 (en) 2014-07-16 2015-07-15 Rnai compositions to treat apoc3-related diseases
EP15822399.0A EP3169784B1 (en) 2014-07-16 2015-07-15 Rnai compositions to treat apoc3-related diseases
US15/403,919 US10240153B2 (en) 2014-07-16 2017-01-11 Organic compositions to treat APOC3-related diseases
US16/269,282 US11066667B2 (en) 2014-07-16 2019-02-06 Organic compositions to treat APOC3-related diseases

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201462025164P 2014-07-16 2014-07-16
US62/025,164 2014-07-16

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US15/403,919 Continuation US10240153B2 (en) 2014-07-16 2017-01-11 Organic compositions to treat APOC3-related diseases

Publications (1)

Publication Number Publication Date
WO2016011123A1 true WO2016011123A1 (en) 2016-01-21

Family

ID=55079014

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2015/040517 WO2016011123A1 (en) 2014-07-16 2015-07-15 Organic compositions to treat apoc3-related diseases

Country Status (3)

Country Link
US (2) US10240153B2 (en)
EP (2) EP3736334A1 (en)
WO (1) WO2016011123A1 (en)

Cited By (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN108239644A (en) * 2016-12-23 2018-07-03 苏州瑞博生物技术有限公司 A kind of small RNA and medical composition and its use
US20200080088A1 (en) * 2014-11-17 2020-03-12 Alnylam Pharmaceuticals, Inc. APOLIPOPROTEIN C3 (APOC3) iRNA COMPOSITIONS AND METHODS OF USE THEREOF
US10597657B2 (en) 2017-09-11 2020-03-24 Arrowhead Pharmaceuticals, Inc. RNAi agents and compositions for inhibiting expression of apolipoprotein C-III (APOC3)
JP2021503929A (en) * 2017-12-01 2021-02-15 スーチョウ リボ ライフ サイエンス カンパニー、リミテッドSuzhou Ribo Life Science Co., Ltd. Nucleic acid, compositions and complexes containing the nucleic acid, and preparation methods and uses
CN112423794A (en) * 2018-12-28 2021-02-26 苏州瑞博生物技术股份有限公司 Nucleic acid, composition containing nucleic acid, conjugate, preparation method and application
CN112423795A (en) * 2018-12-28 2021-02-26 苏州瑞博生物技术股份有限公司 Nucleic acid, composition containing nucleic acid, conjugate, preparation method and application
US11162103B2 (en) 2020-02-18 2021-11-02 Alnylam Pharmaceuticals, Inc. Apolipoprotein C3 (APOC3) iRNA compositions and methods of use thereof
JP2022501059A (en) * 2018-09-30 2022-01-06 スーチョウ リボ ライフ サイエンス カンパニー、リミテッドSuzhou Ribo Life Science Co., Ltd. siRNA complex and its preparation method and use
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
US11633482B2 (en) * 2017-12-29 2023-04-25 Suzhou Ribo Life Science Co., Ltd. Conjugates and preparation and use thereof
WO2023102469A2 (en) 2021-12-01 2023-06-08 Dicerna Pharmaceuticals, Inc. Compositions and methods for modulating apoc3 expression
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 (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JOP20200228A1 (en) * 2015-12-21 2017-06-16 Novartis Ag Compositions and methods for decreasing tau expression
IL300869A (en) * 2016-09-02 2023-04-01 Arrowhead Pharmaceuticals Inc Targeting ligands, compositions comprising same and uses thereof
AU2022309416A1 (en) * 2021-07-16 2024-02-01 Suzhou Ribo Life Science Co., Ltd. Nucleic acid, composition and conjugate containing same, and preparation method and use

Citations (27)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5030453A (en) 1983-03-24 1991-07-09 The Liposome Company, Inc. Stable plurilamellar vesicles
US5962016A (en) 1997-01-31 1999-10-05 Depotech Corporation Multivesicular liposomes utilizing neutral lipids to modify in vivo release
WO2000044914A1 (en) 1999-01-28 2000-08-03 Medical College Of Georgia Research Institute, Inc. Composition and method for in vivo and in vitro attenuation of gene expression using double stranded rna
CA2359180A1 (en) 1999-01-30 2000-08-03 Roland Kreutzer Method and medicament for inhibiting the expression of a given gene
WO2001068836A2 (en) 2000-03-16 2001-09-20 Genetica, Inc. Methods and compositions for rna interference
WO2002100435A1 (en) 2001-06-11 2002-12-19 Centre Hospitalier Universitaire De Montreal Compositions and methods for enhancing nucleic acid transfer into cells
US20030012812A1 (en) 1996-10-04 2003-01-16 Mar Tormo Inhibition of bcl-2 protein expression by liposomal antisense oligodeoxynucleotides
WO2003015757A1 (en) 2001-08-16 2003-02-27 The Trustees Of The University Of Pennsylvania Synthesis and use of reagents for improved dna lipofection and/or slow release prodrug and drug therapies
WO2004002453A1 (en) 2002-06-28 2004-01-08 Protiva Biotherapeutics Ltd. Method and apparatus for producing liposomes
US6680068B2 (en) 2000-07-06 2004-01-20 The General Hospital Corporation Drug delivery formulations and targeting
WO2004029213A2 (en) 2002-09-28 2004-04-08 Massachusetts Institute Of Technology Compositions and methods for delivery of short interfering rna and short hairpin rna
US20040204377A1 (en) 2002-11-26 2004-10-14 University Of Massachusetts Delivery of siRNAs
US20040208921A1 (en) 2003-01-14 2004-10-21 Ho Rodney J. Y. Lipid-drug formulations and methods for targeted delivery of lipid-drug complexes to lymphoid tissues
WO2005021749A1 (en) 2003-08-28 2005-03-10 Novartis Ag Interfering rna duplex having blunt-ends and 3’-modifications
US20060240093A1 (en) 2003-07-16 2006-10-26 Protiva Biotherapeutics, Inc. Lipid encapsulated interfering rna
US20070135372A1 (en) 2005-11-02 2007-06-14 Protiva Biotherapeutics, Inc. Modified siRNA molecules and uses thereof
WO2007107162A2 (en) 2006-03-23 2007-09-27 Santaris Pharma A/S Small internally segmented interfering rna
WO2007128477A2 (en) 2006-05-04 2007-11-15 Novartis Ag SHORT INTERFERING RIBONUCLEIC ACID (siRNA) FOR ORAL ADMINISTRATION
WO2008147824A2 (en) 2007-05-22 2008-12-04 Mdrna, Inc. Hydroxymethyl substituted rna oligonucleotides and rna complexes
WO2009082817A1 (en) 2007-12-27 2009-07-09 Protiva Biotherapeutics, Inc. Silencing of polo-like kinase expression using interfering rna
US20090209626A1 (en) 2008-02-11 2009-08-20 Dharmacon, Inc. Duplex Oligonucleotides with Enhanced Functionality in Gene Regulation
WO2010083615A1 (en) 2009-01-26 2010-07-29 Protiva Biotherapeutics, Inc. Compositions and methods for silencing apolipoprotein c-iii expression
WO2011076807A2 (en) 2009-12-23 2011-06-30 Novartis Ag Lipids, lipid compositions, and methods of using them
WO2012177947A2 (en) 2011-06-21 2012-12-27 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibition of expression of apolipoprotein c-iii (apoc3) genes
WO2013003520A1 (en) * 2011-06-30 2013-01-03 Arrowhead Research Corporation Compositions and methods for inhibiting gene expression of hepatitis b virus
WO2013165816A2 (en) 2012-05-02 2013-11-07 Merck Sharp & Dohme Corp. SHORT INTERFERING NUCLEIC ACID (siNA) COMPOSITIONS
WO2015051366A2 (en) * 2013-10-04 2015-04-09 Novartis Ag Novel formats for organic compounds for use in rna interference

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7615374B2 (en) 2007-09-25 2009-11-10 Wisconsin Alumni Research Foundation Generation of clonal mesenchymal progenitors and mesenchymal stem cell lines under serum-free conditions

Patent Citations (33)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5030453A (en) 1983-03-24 1991-07-09 The Liposome Company, Inc. Stable plurilamellar vesicles
US20030012812A1 (en) 1996-10-04 2003-01-16 Mar Tormo Inhibition of bcl-2 protein expression by liposomal antisense oligodeoxynucleotides
US5962016A (en) 1997-01-31 1999-10-05 Depotech Corporation Multivesicular liposomes utilizing neutral lipids to modify in vivo release
WO2000044914A1 (en) 1999-01-28 2000-08-03 Medical College Of Georgia Research Institute, Inc. Composition and method for in vivo and in vitro attenuation of gene expression using double stranded rna
CA2359180A1 (en) 1999-01-30 2000-08-03 Roland Kreutzer Method and medicament for inhibiting the expression of a given gene
WO2001068836A2 (en) 2000-03-16 2001-09-20 Genetica, Inc. Methods and compositions for rna interference
US6680068B2 (en) 2000-07-06 2004-01-20 The General Hospital Corporation Drug delivery formulations and targeting
WO2002100435A1 (en) 2001-06-11 2002-12-19 Centre Hospitalier Universitaire De Montreal Compositions and methods for enhancing nucleic acid transfer into cells
WO2003015757A1 (en) 2001-08-16 2003-02-27 The Trustees Of The University Of Pennsylvania Synthesis and use of reagents for improved dna lipofection and/or slow release prodrug and drug therapies
WO2004002453A1 (en) 2002-06-28 2004-01-08 Protiva Biotherapeutics Ltd. Method and apparatus for producing liposomes
WO2004029213A2 (en) 2002-09-28 2004-04-08 Massachusetts Institute Of Technology Compositions and methods for delivery of short interfering rna and short hairpin rna
US20040204377A1 (en) 2002-11-26 2004-10-14 University Of Massachusetts Delivery of siRNAs
US20040208921A1 (en) 2003-01-14 2004-10-21 Ho Rodney J. Y. Lipid-drug formulations and methods for targeted delivery of lipid-drug complexes to lymphoid tissues
US20060240093A1 (en) 2003-07-16 2006-10-26 Protiva Biotherapeutics, Inc. Lipid encapsulated interfering rna
US8097716B2 (en) 2003-08-28 2012-01-17 Novartis Ag Interfering RNA duplex having blunt-ends and 3′-modifications
WO2005021749A1 (en) 2003-08-28 2005-03-10 Novartis Ag Interfering rna duplex having blunt-ends and 3’-modifications
US20070135372A1 (en) 2005-11-02 2007-06-14 Protiva Biotherapeutics, Inc. Modified siRNA molecules and uses thereof
WO2007107162A2 (en) 2006-03-23 2007-09-27 Santaris Pharma A/S Small internally segmented interfering rna
WO2007128477A2 (en) 2006-05-04 2007-11-15 Novartis Ag SHORT INTERFERING RIBONUCLEIC ACID (siRNA) FOR ORAL ADMINISTRATION
US8404832B2 (en) 2006-05-04 2013-03-26 Novartis Ag Short interfering ribonucleic acid (siRNA) for oral administration
US8404831B2 (en) 2006-05-04 2013-03-26 Novartis Ag Short interfering ribonucleic acid (siRNA) for oral administration
US8344128B2 (en) 2006-05-04 2013-01-01 Novartis Ag Short interfering ribonucleic acid (siRNA) for oral administration
US8084600B2 (en) 2006-05-04 2011-12-27 Novartis Ag Short interfering ribonucleic acid (siRNA) with improved pharmacological properties
WO2008147824A2 (en) 2007-05-22 2008-12-04 Mdrna, Inc. Hydroxymethyl substituted rna oligonucleotides and rna complexes
WO2009082817A1 (en) 2007-12-27 2009-07-09 Protiva Biotherapeutics, Inc. Silencing of polo-like kinase expression using interfering rna
US20090209626A1 (en) 2008-02-11 2009-08-20 Dharmacon, Inc. Duplex Oligonucleotides with Enhanced Functionality in Gene Regulation
WO2010083615A1 (en) 2009-01-26 2010-07-29 Protiva Biotherapeutics, Inc. Compositions and methods for silencing apolipoprotein c-iii expression
WO2011076807A2 (en) 2009-12-23 2011-06-30 Novartis Ag Lipids, lipid compositions, and methods of using them
WO2012177947A2 (en) 2011-06-21 2012-12-27 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibition of expression of apolipoprotein c-iii (apoc3) genes
US20140113957A1 (en) * 2011-06-21 2014-04-24 Brian Bettencourt Compositions and Methods for Inhibition of Expression of Apolipoprotein C-III (APOC3) Genes
WO2013003520A1 (en) * 2011-06-30 2013-01-03 Arrowhead Research Corporation Compositions and methods for inhibiting gene expression of hepatitis b virus
WO2013165816A2 (en) 2012-05-02 2013-11-07 Merck Sharp & Dohme Corp. SHORT INTERFERING NUCLEIC ACID (siNA) COMPOSITIONS
WO2015051366A2 (en) * 2013-10-04 2015-04-09 Novartis Ag Novel formats for organic compounds for use in rna interference

Non-Patent Citations (45)

* Cited by examiner, † Cited by third party
Title
BERNSTEIN ET AL., NATURE, vol. 409, 2001, pages 363 - 498
BURGIN ET AL., BIOCHEM., vol. 35, 1996, pages 14090
BURGIN ET AL., BIOCHEMISTRY, vol. 35, 1996, pages 14090
CAPLEN ET AL., PROC. NATL. ACAD. SCI. (USA, vol. 98, 2001, pages 9742 - 9747
CHURANA, RNA, vol. 14, 2008, pages 1714 - 1719
DAMMERMAN ET AL., PROC. NATL. ACAD. SCI. U.S.A., vol. 90, 1993, pages 4562 - 4566
DE SILVA ET AL., J. BIOL. CHEM., vol. 269, 1994, pages 23610 - 23616
DOWLER ET AL., NUCL. ACIDS RES., vol. 34, 2006, pages 1669 - 1675
ELBASHIR ET AL., EMBO J., vol. 20, 2001, pages 6877 - 6888
ELBASHIR ET AL., GENES DEV., vol. 15, 2001, pages 188
GAMBLING ET AL., 2 KIDNEY INTL., vol. 65, pages 1774 - 1781
GAULTIER ET AL., NUCLEIC ACIDS. RES., vol. 15, 1987, pages 6625 - 6641
HENSCHEL ET AL.: "DEQOR: a web-based tool for the design and quality control of siRNAs", NUCLEIC ACIDS RESEARCH, vol. 32, 2004, pages W113 - W120, XP001206600, doi:10.1093/nar/gnh112
HERTZ ET AL., J. BIOL. CHEM., vol. 270, 1995, pages 13470 - 13475
HUTVAGNER ET AL., SCIENCE, vol. 293, 2001, pages 834
INOUE ET AL., FEBS LETT., vol. 215, 1987, pages 327 - 330
INOUE ET AL., NUCLEIC ACIDS RES., vol. 15, 1987, pages 6131 - 6148
ITO ET AL., SCIENCE, vol. 249, 1990, pages 790 - 793
KARATHANASIS, PROC. NATL. ACAD. SCI. U.S.A., vol. 82, 1985, pages 6374 - 6378
KARDASSIS ET AL., BIOCHEMISTRY, vol. 41, 2002, pages 1217 - 1228
KARDASSIS ET AL., J. BIOL. CHEM., vol. 275, 2000, pages 41405 - 41414
KRAYNACK ET AL., RNA, vol. 12, 2006, pages 163 - 176
LEVY-WILSON ET AL., DNA, vol. 3, 1984, pages 449 - 456
LI ET AL., J. CLIN. INVEST., vol. 96, 1995, pages 2601 - 2605
MAEDA ET AL., J. LIPID RES., vol. 28, 1987, pages 1405 - 1409
MCCAFFREY ET AL., NATURE, vol. 414, pages 34 - 39
NYKANEN ET AL., CELL, vol. 107, 2001, pages 309
OGAMI ET AL., J. BIOL. CHEM., vol. 266, 1991, pages 9640 - 9646
OLIVIERI ET AL., J. LIPID RES., vol. 43, 2002, pages 2172 - 2179
OVCHARENKO D: "Efficient delivery of siRNAs to human primary cells", AMBION TECHNOTES, vol. 10, no. 5, 2003, pages 15 - 16
PARRISH ET AL., MOLECULAR CELL, vol. 6, 2000, pages 1077 - 1087
SCHIFFELERS ET AL., NUCL. ACIDS RES., vol. 32, 2004, pages 141 - 1 10
SCHOONJANS ET AL., FEBS LETT., vol. 452, 1999, pages 160 - 164
SHACHTER, CURR. OPIN. LIPIDOL., vol. 12, 2001, pages 297 - 304
SHARPE ET AL., NUCLEIC ACIDS RES., vol. 12, 1984, pages 3917 - 3932
SIOUD, J. MOL. BIOL., vol. 348, 2005, pages 1079 - 1090
SONG ET AL., NAT BIOTECH., vol. 23, 2005, pages 709 - 717
SONG ET AL., NAT MED., 10 February 2003 (2003-02-10)
SOUTSCHEK ET AL., NATURE, vol. 432, 2004, pages 173 - 178
SUN ET AL., NATURE BIOTECH., vol. 26, 2008, pages 1379 - 1382
USMAN ET AL., NUCL. ACIDS SYMP. SER., vol. 31, 1994, pages 163
USMAN ET AL., NUCLEIC ACIDS SYMP. SER., vol. 31, 1994, pages 163
USMANCEDERGREN, TIBS, vol. 17, 1992, pages 34
VU-DAC ET AL., J. CLIN. INVEST., vol. 102, 1998, pages 625 - 632
YAMATO ET AL., CANCER GENE THER., vol. 18, 2011, pages 587 - 597

Cited By (24)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11034958B2 (en) 2014-11-17 2021-06-15 Alnylam Pharmaceuticals, Inc. Apolipoprotein C3 (APOC3) iRNA compositions and methods of use thereof
US20200080088A1 (en) * 2014-11-17 2020-03-12 Alnylam Pharmaceuticals, Inc. APOLIPOPROTEIN C3 (APOC3) iRNA COMPOSITIONS AND METHODS OF USE THEREOF
US11408001B1 (en) 2014-11-17 2022-08-09 Alnylam Pharmaceuticals, Inc. Apolipoprotein C3 (APOC3) iRNA compositions and methods of use thereof
US11142766B2 (en) 2014-11-17 2021-10-12 Alnylam Pharmaceuticals, Inc. Apolipoprotein C3 (APOC3) iRNA compositions and methods of use thereof
EP4035659A1 (en) 2016-11-29 2022-08-03 PureTech LYT, Inc. Exosomes for delivery of therapeutic agents
CN108239644A (en) * 2016-12-23 2018-07-03 苏州瑞博生物技术有限公司 A kind of small RNA and medical composition and its use
CN108239644B (en) * 2016-12-23 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
US11214801B2 (en) 2017-09-11 2022-01-04 Arrowhead Pharmaceuticals, Inc. RNAi agents and compositions for inhibiting expression of apolipoprotein C-III (APOC3)
US10597657B2 (en) 2017-09-11 2020-03-24 Arrowhead Pharmaceuticals, Inc. RNAi agents and compositions for inhibiting expression of apolipoprotein C-III (APOC3)
EP3681516A4 (en) * 2017-09-11 2021-09-29 Arrowhead Pharmaceuticals, Inc. Rnai agents and compositions for inhibiting expression of apolipoprotein c-iii (apoc3)
JP7365052B2 (en) 2017-12-01 2023-10-19 スーチョウ リボ ライフ サイエンス カンパニー、リミテッド Nucleic acids, compositions and complexes containing the nucleic acids, and preparation methods and uses
EP3719127A4 (en) * 2017-12-01 2021-10-20 Suzhou Ribo Life Science Co., Ltd. Nucleic acid, composition and conjugate containing same, preparation method, and use
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
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
JP2021503929A (en) * 2017-12-01 2021-02-15 スーチョウ リボ ライフ サイエンス カンパニー、リミテッドSuzhou Ribo Life Science Co., Ltd. Nucleic acid, compositions and complexes containing the nucleic acid, and preparation methods and uses
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
JP2022501059A (en) * 2018-09-30 2022-01-06 スーチョウ リボ ライフ サイエンス カンパニー、リミテッドSuzhou Ribo Life Science Co., Ltd. siRNA complex and its preparation method and use
JP7376952B2 (en) 2018-09-30 2023-11-09 スーチョウ リボ ライフ サイエンス カンパニー、リミテッド siRNA complex and its preparation method and use
US11896674B2 (en) 2018-09-30 2024-02-13 Suzhou Ribo Life Science Co., Ltd. SiRNA conjugate, preparation method therefor and use thereof
CN112423794A (en) * 2018-12-28 2021-02-26 苏州瑞博生物技术股份有限公司 Nucleic acid, composition containing nucleic acid, conjugate, preparation method and application
CN112423795A (en) * 2018-12-28 2021-02-26 苏州瑞博生物技术股份有限公司 Nucleic acid, composition containing nucleic acid, conjugate, preparation method and application
US11162103B2 (en) 2020-02-18 2021-11-02 Alnylam Pharmaceuticals, Inc. Apolipoprotein C3 (APOC3) iRNA compositions and methods of use thereof
WO2023102469A2 (en) 2021-12-01 2023-06-08 Dicerna Pharmaceuticals, Inc. Compositions and methods for modulating apoc3 expression

Also Published As

Publication number Publication date
US20190233817A1 (en) 2019-08-01
EP3169784A1 (en) 2017-05-24
US11066667B2 (en) 2021-07-20
EP3169784A4 (en) 2018-03-21
EP3736334A1 (en) 2020-11-11
US10240153B2 (en) 2019-03-26
EP3169784B1 (en) 2020-06-10
US20170137814A1 (en) 2017-05-18

Similar Documents

Publication Publication Date Title
US11066667B2 (en) Organic compositions to treat APOC3-related diseases
US11261444B2 (en) Organic compositions to treat EPAS1-related diseases
US9376683B2 (en) Organic compositions to treat beta-ENaC-related diseases
EP3272868A1 (en) Organic compositions to treat kras-related diseases
AU2011244335A1 (en) Organic compositions to treat Beta-ENaC-related diseases
JP2022528487A (en) Oligonucleotide-based regulation of C9orf72
CN113811311A (en) Oligonucleotides for tissue-specific APOE modulation
CA2860676A1 (en) Organic compositions to treat beta-catenin-related diseases
CN115552006A (en) Oligonucleotides for MAPT modulation
CN115777021A (en) Oligonucleotides for SNCA modulation
WO2015051135A2 (en) Organic compositions to treat hepcidin-related diseases
AU2021240326A1 (en) Organic compositions to treat Beta-ENac-related diseases
CN117897482A (en) Compositions and methods for treating cancer

Legal Events

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

Ref document number: 15822399

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

REEP Request for entry into the european phase

Ref document number: 2015822399

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2015822399

Country of ref document: EP