[go: up one dir, main page]

WO2024227059A2 - Short interfering nucleic acid (sina) molecules targeting angptl 3 and angptl 8 and methods of using the same - Google Patents

Short interfering nucleic acid (sina) molecules targeting angptl 3 and angptl 8 and methods of using the same Download PDF

Info

Publication number
WO2024227059A2
WO2024227059A2 PCT/US2024/026619 US2024026619W WO2024227059A2 WO 2024227059 A2 WO2024227059 A2 WO 2024227059A2 US 2024026619 W US2024026619 W US 2024026619W WO 2024227059 A2 WO2024227059 A2 WO 2024227059A2
Authority
WO
WIPO (PCT)
Prior art keywords
nucleotide
sina
nucleotide sequence
nucleotides
antisense strand
Prior art date
Application number
PCT/US2024/026619
Other languages
French (fr)
Other versions
WO2024227059A3 (en
Inventor
Leonid Beigelman
Vivek Kumar Rajwanshi
David Bernard Smith
Jin Hong
Xuan LUONG
Saul MARTINEZ MONTERO
Antitsa Dimitrova Stoycheva
Aneerban BHATTACHARYA
Original Assignee
Aligos Therapeutics, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Aligos Therapeutics, Inc. filed Critical Aligos Therapeutics, Inc.
Publication of WO2024227059A2 publication Critical patent/WO2024227059A2/en
Publication of WO2024227059A3 publication Critical patent/WO2024227059A3/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • C12N15/1136Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing against growth factors, growth regulators, cytokines, lymphokines or hormones
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/10Type of nucleic acid
    • C12N2310/14Type of nucleic acid interfering N.A.
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/31Chemical structure of the backbone
    • C12N2310/315Phosphorothioates
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/32Chemical structure of the sugar
    • C12N2310/3212'-O-R Modification
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/32Chemical structure of the sugar
    • C12N2310/3222'-R Modification
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/34Spatial arrangement of the modifications
    • C12N2310/343Spatial arrangement of the modifications having patterns, e.g. ==--==--==--

Definitions

  • Short Interfering Nucleic Acid (siNA) Molecules Targeting ANGPTL 3 and ANGPTL 8 and Methods of Using the Same TECHNICAL FIELD The present disclosure is in the field of pharmaceutical compounds and preparations and method of their use in the treatment of disease. Described are short interfering nucleic acid (siNA) molecules comprising modified nucleotides that target ANGPTL3 or ANGPTL8, compositions containing the same, and uses thereof for treating or preventing diseases. In particular, the present disclosure is in the field of siNA molecules targeting ANGPTL3 and ANGPTL8 for treatment of cardiovascular disease and nonalcoholic fatty liver, among other diseases.
  • siNA Short Interfering Nucleic Acid
  • Triglycerides are the main form of lipid that provides and stores energy in the human body.
  • Lipoprotein lipase (LPL) breaks down triglycerides in lipoproteins, such as chylomicrons (CM) and very low-density lipoproteins (VLDL), into two free fatty acids and one monoacylglycerol molecule.
  • ANGPTL3 also known as Angiopoietin-like protein 3, is a protein exclusively and constitutively expressed in the liver.
  • ANGPTL8 also known as lipasin, is expressed in the liver and adipose tissues after fed conditions.
  • ANGPTL3 as well as ANGTL4, are inhibitors of LPL and they work in coordination with the cofactor ANGPTL8.
  • the inhibition of LPL subsequently increases plasma triglyceride (TG), LDL cholesterol (LDL-c), Total Cholesterol (TC), and HDL-cholesterol (HDL-c).
  • ANGPTL3 can form a complex with the co-factor ANGPTL8 (ANGPTL3/8).
  • the ANGPTL3/8 complex is a much stronger inhibitor of LPL in oxidative tissues such as heart and muscle (i.e., >100 fold) than ANGPTL3 alone, and ANGPTL8 is not an inhibitor of LPL.
  • ANGPTL3/8 complex Reducing the ANGPTL3/8 complex by applying a combination of ANGPTL3 siNA and ANGPTL8 siNA treatments could achieve greater TG, TC, and LDL-c reduction than ANGPTL3 siNA treatment alone.
  • ANGPTL3 also inhibits endothelial lipase (EL) which results in elevation of HDL-c.
  • EL endothelial lipase
  • ANGPTL8 cofactor is not required in inhibition of EL by ANGPTL3.
  • ANGPTL3 siNA treatment alone has been shown to reduce “good” cholesterol HDL-c which can be a problem.
  • ANGPTL3 siNA By optimizing the ANGPTL3 siNA vs ANGPTL8 siNA ratio, is possible to achieve lower HDL-c reduction than with ANGPTL3 siNA mono-therapy, or no HDL-c reduction, or even slight elevation of HDL-c in circulation.
  • Hypertriglyceridemia is associated with increased risk of atherosclerotic cardiovascular diseases and with fatty acid liver disease. Treatments that target the production and clearance of triglyceride-rich lipoproteins (TRLs) mitigate these complications.
  • TRLs triglyceride-rich lipoproteins
  • ANGPTL3 inhibition also lowers LDL particle concentration and may therefore have an overall advantage as a lipid-regulating agent.
  • RNA interference is a biological response to double-stranded RNA that mediates resistance to both endogenous parasitic and exogenous pathogenic nucleic acids, and regulates the expression of protein-coding genes.
  • the short interfering nucleic acids such as siRNA, have been developed for RNAi therapy to treat a variety of diseases.
  • RNAi therapy has been proposed for the treatment of metabolic diseases, neurodegenerative diseases, cancer, and pathogenic infections.
  • the present disclosure provides siNA molecules useful for targeting and inhibiting ANGPTL3 and ANGPTL8 in optimized combination ratios, which subsequently increases LPL activity in oxidative tissues, such as, heart and muscles resulting in decreased TG, TC, and LDL-c in circulation.
  • the combination therapy can maintain or increase HDL-c by maintaining a high level of free ANGPTL3 and subsequence inhibition of EL.
  • the dual targeting siNA approach reduces dosing frequency and results in better patient compliance.
  • the present disclosure provides a short interfering nucleic acid (siNA) molecule comprising: (a) a sense strand comprising a first nucleotide sequence that is at least about 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% identical to an RNA corresponding to a target gene, wherein the first nucleotide sequence: is 15 to 30 nucleotides in length; and comprises 15 or more modified nucleotides independently selected from a 2’-O-methyl nucleotide and a 2’-fluoro nucleotide, wherein at least one modified nucleotide is a 2’-O- methyl nucleotide and the nucleotide at position 3, 5, 7, 8, 9, 10, 11, 12, 14, 17, and/or 19 from the 5’ end of the first nucleotide sequence is a 2’-fluoro nucleotide or wherein at least one modified nucleotide is a 2’-O-methyl nucleot
  • the present disclosure provides a short interfering nucleic acid (siNA) molecule comprising: (a) a sense strand comprising a first nucleotide sequence that is at least about 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% identical to an RNA corresponding to a target gene, wherein the first nucleotide sequence: is 15 to 30 nucleotides in length; and comprises 15 or more modified nucleotides independently selected from a 2’-O-methyl nucleotide and a 2’-fluoro nucleotide, wherein at least one modified nucleotide is a 2’-O- methyl nucleotide and the nucleotide at position 3, 5, 7, 8, 9, 10, 11, 12, 14, 17, and/or 19 from the 5’ end of the first nucleotide sequence is a 2’-fluoro nucleotide or wherein at least one modified nucleotide is a 2’-O-methyl nucleot
  • the present disclosure provides double-stranded short interfering nucleic acid (ds-siNA) molecules comprising: (a) a sense strand comprising a first nucleotide sequence, wherein the first nucleotide sequence is 15 to 30 nucleotides in length and comprises a nucleotide sequence as set forth in in any one of SEQ ID NOs: 1-316, and an antisense strand comprising a second nucleotide sequence, wherein the second nucleotide sequence is 15 to 30 nucleotides in length and comprises a nucleotide sequence that is a substantial reverse complement of the first nucleotide sequence; or (b) a sense strand comprising a first nucleotide sequence, wherein the first nucleotide sequence is 15 to 30 nucleotides in length and comprises a nucleotide sequence that is a substantial reverse complement of a second nucleotide sequence; and an antisense strand comprising the second nucleotide sequence
  • the present disclosure provides double-stranded short interfering nucleic acid (ds-siNA) molecules comprising: (a) a sense strand comprising a first nucleotide sequence, wherein the first nucleotide sequence is 15 to 30 nucleotides in length and comprises a nucleotide sequence as set forth in in any one of SEQ ID NOs: 633-878, and an antisense strand comprising a second nucleotide sequence, wherein the second nucleotide sequence is 15 to 30 nucleotides in length and comprises a nucleotide sequence that is a substantial reverse complement of the first nucleotide sequence; or (b) a sense strand comprising a first nucleotide sequence, wherein the first nucleotide sequence is 15 to 30 nucleotides in length and comprises a nucleotide sequence that is a substantial reverse complement of a second nucleotide sequence; and an antisense strand comprising the second nucleot
  • the present disclosure provides double-stranded short interfering nucleic acid (ds-siNA) molecules comprising: (a) a sense strand comprising a nucleotide sequence as set forth in any one of SEQ ID NOs: 1-316, and (b) an antisense strand.
  • ds-siNA double-stranded short interfering nucleic acid
  • ds-siNA double-stranded short interfering nucleic acid
  • the present disclosure provides double-stranded short interfering nucleic acid (ds-siNA) molecules comprising (a) an antisense strand comprising a nucleotide sequence as set forth in any one of SEQ ID NOs: 317-632 (b) a sense strand.
  • the present disclosure provides double-stranded short interfering nucleic acid (ds-siNA) molecules comprising (a) an antisense strand comprising a nucleotide sequence as set forth in any one of SEQ ID NOs: 879-1125 (b) a sense strand.
  • the sense strand comprises SEQ ID NOs: 633, 634, 642, 653, 654, 674, 687, 688, 690-694, 708-713, 756-757, 765, 776, 777, 797, 810, 811, 813-817, or 831-836 and the antisense strand comprises SEQ ID NOs: 879, 880, 888, 899, 900, 920, 933, 934, 936-940, 954-959, 1003-1004, 1012, 1023, 1024, 1044, 1057, 1058, 1060-1064, or 1078-1083, respectively.
  • the sense strand comprises SEQ ID NOs: 633, 654, 688, 690, 708, 713, 756, 777, 811, 813, 831, or 836
  • the antisense strand comprises SEQ ID NOs: 879, 900, 934, 936, 954, 959, 1003, 1024, 1058, 1060, 1078, or 1083, respectively.
  • the sense and/or antisense strand further comprises a TT sequence at 3’ end.
  • at least one end of the ds-siNA is a blunt end.
  • At least one end of the ds-siNA comprises an overhang, wherein the overhang comprises at least one nucleotide, optionally wherein the at least one nucleotide comprises a 2’-O-methyl nucleotide, a deoxy nucleotide, or a uracil analogue or derivative.
  • both ends of the ds-siNA comprise an overhang, wherein the overhang comprises at least one nucleotide, optionally wherein the at least one nucleotide comprises a 2’-O-methyl nucleotide, a deoxy nucleotide, or a uracil analogue or derivative.
  • the sense strand and/or the antisense strand independently comprise 1 or more mesyl phosphoramidate internucleoside linkages, or 1 or more phosphorothioate internucleoside linkages. In some embodiments of any of the foregoing aspects or embodiments, the sense strand comprises at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15 or more phosphorothioate internucleoside linkages.
  • the antisense strand further comprises at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15 or more phosphorothioate internucleoside linkages.
  • At least one phosphorothioate internucleoside linkage in the antisense strand is between the nucleotides at positions 1 and 2 from the 5’ end of the antisense strand sequence; (ii) at least one phosphorothioate internucleoside linkage in the antisense strand is between the nucleotides at positions 2 and 3 from the 5’ end of the antisense strand sequence; (iii) at least one phosphorothioate internucleoside linkage in the antisense strand is between the nucleotides at positions 1 and 2 from the 3’ end of the antisense strand sequence; and/or (iv) at least one phosphorothioate internucleoside linkage is between the nucleotides at positions 2 and 3 from the 3’ end of the antisense strand sequence.
  • the sense strand comprises at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15 or more mesyl phosphoramidate internucleoside linkages.
  • at least one mesyl phosphoramidate internucleoside linkage in the sense strand is between the nucleotides at positions 1 and 2 from the 5’ end of the sense strand sequence;
  • at least one mesyl phosphoramidate internucleoside linkage is between the nucleotides at positions 2 and 3 from the 5’ end of the sense strand sequence.
  • the antisense strand further comprises at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15 or more mesyl phosphoramidate internucleoside linkages.
  • at least one mesyl phosphoramidate internucleoside linkage in the antisense strand is between the nucleotides at positions 1 and 2 from the 5’ end of the antisense strand sequence;
  • at least one mesyl phosphoramidate internucleoside linkage in the antisense strand is between the nucleotides at positions 2 and 3 from the 5’ end of the antisense strand sequence;
  • at least one mesyl phosphoramidate internucleoside linkage in the antisense strand is between the nucleotides at positions 1 and 2 from the 3’ end of the antisense strand sequence; and/or (iv) at least one mesyl phosphoramidate
  • the antisense strand comprises one or more modified nucleotides. In some embodiments of any of the foregoing aspects or embodiments, the sense strand comprises one or more modified nucleotides. In some embodiments of any of the foregoing aspects or embodiments, the sense strand and the antisense strand each independently comprise one or more modified nucleotides. In some embodiments, the modified nucleotides are independently selected from 2’-O-methyl nucleotides and 2’-fluoro nucleotides. Other modified nucleotides that may be included in the sense strand, antisense strand, or both are disclosed herein.
  • the antisense strand, sense strand, first nucleotide sequence, and/or second nucleotide sequence comprises at least one modified nucleotide selected from: , wherein Rx is a nucleobase, aryl, heteroaryl, or H, (mun34), wherein Ry is a nucleobase,
  • the ds-siNA further comprises a phosphorylation blocker, a 5’-stabilized end cap, or a combination thereof.
  • the phosphorylation blocker is attached to the 5’ end of the sense strand, and optionally, wherein the phosphorylation blocker is attached to the 5’ end of the sense strand via one or more of a phosphodiester linkage, a phosphorothioate linkage, a mesyl phosphoramidate linkage, or a phosphorodithioate linkage.
  • the 5’-stabilized end cap is a 5’ vinyl phosphonate (e.g., vm).
  • the 5’- stabilized end cap is attached to the 5’ end of the antisense strand, and optionally, wherein the 5’-stabilized end cap is attached to the 5’ end of the antisense strand one or more of a phosphodiester linkage, a phosphorothioate linkage, a mesyl phosphoramidate linkage, or a phosphorodithioate linkage.
  • the 5’-stabilzied end cap comprises a structure of: wherein B is a nucleobase, aryl, heteroaryl, or H; wherein represents a phosphodiester linkage, a phosphorothioate linkage, or a mesyl phosphoramidate linkage.
  • the ds-siNA further comprises a galactosamine.
  • the galactosamine is an N-acetylgalactosamine comprising a structure of:
  • the sense and/or anti-sense strand of the ds-siNA is conjugated to GalNAc, Folate, Cholesterol, or Palmitic Acid.
  • the ds-siNA reduces ANGPTL8 or ANGPTL3 expression by more than about 10 %, more than about 20% more than about 30%, more than about 40%, more than about 50%, more than about 60%, more than about 70%, more than about 80%, more than about 90%, or more than about 95%.
  • the ds-siNA reduces ANGPTL8 or ANGPTL3 expression by 100%. In some embodiments of any of the foregoing aspects or embodiments, the ds-siNA reduces ANGPTL8 or ANGPTL3 expression or activity with a EC50 value of 50 pM or less, of 40 pM or less, of 30 pM or less, of 20 pM or less, 15 pM or less, 10 pM or less, 5 pM or less, 2 pM or less, or 1 pM or less.
  • the ds-siNA has a CC50 value of more than 1 ⁇ M. In some embodiments of any of the foregoing aspects or embodiments, the ds-siNA comprises RNA nucleotides. In another aspect, the present disclosure provides ds-siNA, comprising a sense strand and an antisense strand, wherein the antisense strand comprises at its 5’ end one of the following (vm, wherein B is a nucleobase), (vm(5mim)), (vm(56mido)), and (vm(56amim)U, wherein R is H or Bz).
  • the present disclosure provides pharmaceutical compositions comprising at least one ds-siNA disclosed herein and a pharmaceutically acceptable carrier or diluent.
  • the present disclosure provides pharmaceutical compositions comprising two or more ds-siNA disclosed herein.
  • the combination of two or more ds-siNA disclosed herein will generally comprise at least one ds-siNA that reduces expression of ANGPTL3 and at least one ds-siNA that reduces expression of ANGPTL8.
  • the present disclosure provides pharmaceutical compositions comprising at least a first siNA that reduces ANGPTL3 expression and a second siNA that reduces ANGPTL8 expression and a pharmaceutically acceptable carrier.
  • the first siNA that reduces ANGPTL3 expression comprises a nucleic acid sequence disclosed in Table 3
  • the second siNA that reduces ANGPTL8 expression comprises a nucleic acid sequence disclosed in Table 1.
  • the first siNA that reduces ANGPTL3 expression is a ds-siNA disclosed in Table 4
  • the second siNA that reduces ANGPTL8 expression is a ds-siNA disclosed in Table 2.
  • a weight ratio of the first siNA and the second siNA is selected from 1:10, 1:9, 1:8, 1:7, 1:6, 1:5, 1:4, 1:3, 1:2, 1:1, 2:1, 3:1, 4:1, 5:1, 6:1, 7:1, 8:1, 9:1, or 10:1.
  • a molar ration of the first siNA and the second siNA is selected from 1:10, 1:9, 1:8, 1:7, 1:6, 1:5, 1:4, 1:3, 1:2, 1:1, 2:1, 3:1, 4:1, 5:1, 6:1, 7:1, 8:1, 9:1, or 10:1.
  • the first siNA and the second siNA are connected by a linker, which may be a nucleotide-based linker or a non-nucleotide-based linker.
  • the pharmaceutical compositions disclosed herein may be formulated for parenteral, ocular, nasal, transdermal, pulmonary, or topical administration or comprises a liposome that encapsulates the ds-siNA.
  • the present disclosure also provides uses of the ds-siNA disclosed herein in the manufacture of a medicament for treating cardiovascular disease or nonalcoholic fatty liver disease.
  • the present disclosure also provides uses of two or more ds-siNA disclosed herein in the manufacture of a medicament for treating cardiovascular disease or nonalcoholic fatty liver disease.
  • the present disclosure also provides ds-siNA disclosed herein or the pharmaceutical compositions disclosed herein for use in treating cardiovascular disease or nonalcoholic fatty liver disease.
  • the present disclosure also provides two or more ds-siNA disclosed herein for use in treating cardiovascular disease or nonalcoholic fatty liver disease.
  • the present disclosure provides methods of treating a disease in a subject in need thereof, comprising administering to the subject a ds-siNA disclosed herein or a pharmaceutical composition disclosed herein, wherein the disease is cardiovascular disease or nonalcoholic fatty liver disease.
  • the present disclosure provides methods of treating a disease in a subject in need thereof, comprising administering to the subject two or more ds-siNA disclosed herein, wherein the disease is cardiovascular disease or nonalcoholic fatty liver disease.
  • the present disclosure provides methods of treating a disease in a subject in need thereof, comprising administering to the subject a first pharmaceutical composition comprising at least a first ds-siNA that reduces ANGPTL3 expression and a second pharmaceutical composition comprising at least a second ds-siNA that reduces ANGPTL8 expression, wherein the disease is cardiovascular disease or nonalcoholic fatty liver disease.
  • the first siNA that reduces ANGPTL3 expression comprises a nucleic acid sequence disclosed in Table 3
  • the second siNA that reduces ANGPTL8 expression comprises a nucleic acid sequence disclosed in Table 1.
  • the first siNA that reduces ANGPTL3 expression is a ds-siNA disclosed in Table 4, and the second siNA that reduces ANGPTL8 expression is a ds-siNA disclosed in Table 2.
  • the cardiovascular disease is Hypertriglyceridemia (HTG) or Familial Hypercholesterolemia (FH).
  • HMG Hypertriglyceridemia
  • FH Familial Hypercholesterolemia
  • the subject is a mammal, and optionally wherein the mammal is a human or a non-human primate.
  • the ds-siNA or pharmaceutical composition(s) are administered intravenously, subcutaneously, or via inhalation.
  • the present disclosure provides methods of reducing ANGPTL8 and/or ANGPTL3 expression or activity in a tissue, an organ, or a cell of a subject in need thereof, wherein the method comprises delivering to the subject one or more of the ds-siNA disclosed herein or the pharmaceutical compositions disclosed herein.
  • the present disclosure provides modified nucleosides selected from (vm, wherein B is a nucleobase), (vm(5mim)), (vm(56mido)), and (vm(56amim)U, wherein R is H or Bz).
  • FIG. 1A-1F depicts examples of siNA molecules that target ANGPTL3.
  • FIG. 2A-2B provides the effects of exemplary siNAs after treatment of COS-7 cells with a dose response curve.
  • FIG. 2A shows the percentage of viable COS-7 cells after treatment.
  • FIG. 2B shows percentage inhibition of cynomologus monkey ANGPTL3 after treatment with 6 exemplary siNAs as determined by luciferase assay.
  • FIG. 1A-1F depicts examples of siNA molecules that target ANGPTL3.
  • FIG. 2A-2B provides the effects of exemplary siNAs after treatment of COS-7 cells with a dose response curve.
  • FIG. 2A shows the percentage of viable COS-7 cells after treatment.
  • FIG. 2B shows percentage inhibition of cynomologus monkey ANGPTL3 after treatment with 6 exemplary siNAs as determined by luciferase assay.
  • FIG. 3 depicts the percentage change in expression of ANGPTL3 protein from serum samples in groups treated with the ANGPTL3 siNAs relative to the expression levels from serum samples in the group treated with the negative vehicle control in the AAV-hANGPTL3 mice. Serum samples were taken at Day 28 post-dose.
  • FIG. 4 depicts the percentage change in expression of ANGPTL3 protein from serum samples in groups treated with the ANGPTL3 siNAs relative to the expression levels from serum samples in a group treated with the negative vehicle control in the hANGPTL3 knock- in mice. Serum samples were taken at Day 28 post-dose.
  • FIG. 4 depicts the percentage change in expression of ANGPTL3 protein from serum samples in groups treated with the ANGPTL3 siNAs relative to the expression levels from serum samples in a group treated with the negative vehicle control in the hANGPTL3 knock- in mice. Serum samples were taken at Day 28 post-dose.
  • FIG. 5 depicts the percentage change in expression of ANGPTL3 RNA of the liver samples in groups treated with ANGPTL3 siNAs relative to the expression levels of the same RNA in the livers of the group treated with the negative vehicle control in the hANGPTL3 knock-in mice. Liver samples were collected at Day 28 post-dose.
  • FIG. 6 depicts the results of the plasmid based on- and off-target luciferase assay for siNAs having destabilized seed regions.
  • FIG. 7 depicts the percentage change in expression of ANGPTL8 RNA in the livers of the groups treated with ANGPTL8 siNAs relative to the expression levels of the same RNA in the livers of the group treated with the negative vehicle control in the AAV-hANGPTL8 mice.
  • FIGS. 8A, 8B, 8C, 8D, and 8E depict percent reduction in various lipids following single or combination hANGPTL3 siNA and hANGPTL8 siNA treatment in the human ANGPTL3/8 double knock-in mouse study.
  • FIG. 8A shows the percent reduction in total cholesterol levels Day 7 post-dose.
  • FIG. 8B shows the percent reduction in total cholesterol levels Day 14 post-dose.
  • FIG. 8C shows the percent reduction in total cholesterol levels Day 21 post-dose.
  • FIG. 8D shows the percent reduction in total cholesterol levels Day 28 post- dose.
  • FIG. 8E shows the percent reduction in triglyceride levels Day 14 post-dose.
  • ANGPTL3 (along with ANGPTL4) work in coordination with cofactor ANGPTL8 to regulate lipid and triglyceride metabolism.
  • ANGPTL3 is exclusively and constitutively expressed in the liver, and its inhibition lowers LDL particle concentration.
  • ANGPTL8 is expressed in the liver and adipose tissues and is induced under the fed conditions. Inhibiting or lowering expression of either or both of ANGPTL3 and ANGPTL8 could treat cardiovascular disease, nonalcoholic fatty liver disease (NAFLD), metabolic dysfunction-associated steatohepatitis (MASH), and other diseases associated with high triglyceride levels.
  • NAFLD nonalcoholic fatty liver disease
  • MASH metabolic dysfunction-associated steatohepatitis
  • ANGPTL3-targeting siNA and ANGPTL8-targeting siNA that can be used together to treatments as disclosed in more detail herein.
  • Definitions It is to be understood that methods are not limited to the particular embodiments described, and as such may vary. It is also to be understood that the terminology used herein is for the purpose of describing particular embodiments only and is not intended to be limiting. The scope of the present technology will be limited only by the appended claims. Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. Although any methods and materials similar or equivalent to those described herein can also be used in the practice or testing of the present invention, representative illustrative methods and materials are now described.
  • compositions and methods include the recited elements, but not excluding others.
  • Consisting essentially of when used to define compositions and methods, shall mean excluding other elements of any essential significance to the composition or method.
  • Consisting of shall mean excluding more than trace elements of other ingredients for claimed compositions and substantial method steps. Embodiments defined by each of these transition terms are within the scope of this disclosure. Accordingly, it is intended that the methods and compositions can include additional steps and components (comprising) or alternatively including steps and compositions of no significance (consisting essentially of) or alternatively, intending only the stated method steps or compositions (consisting of).
  • “about” means plus or minus 10% as well as the specified number. For example, “about 10” should be understood as both “10” and “9-11.”
  • “optional” or “optionally” means that the subsequently described event or circumstance may or may not occur, and that the description includes instances where said event or circumstance occurs and instances where it does not.
  • the terms “individual,” “subject,” and “patient” are used interchangeably herein, and refer to any individual mammal, e.g., bovine, canine, feline, equine, simian, porcine, camelid, bat, or human, being treated according to the disclosed methods or uses. In preferred embodiments, the subject is a human.
  • the phrases “effective amount,” “therapeutically effective amount,” and “therapeutic level” mean the siNA dosage or concentration in a subject that provides the specific pharmacological effect for which the siNA is administered in a subject in need of such treatment, i.e. to treat or prevent a cardiovascular disease, nonalcoholic fatty liver disease, etc. It is emphasized that a therapeutically effective amount or therapeutic level of an siNA will not always be effective in treating the diseases described herein, even though such dosage is deemed to be a therapeutically effective amount by those of skill in the art. For convenience only, exemplary dosages, drug delivery amounts, therapeutically effective amounts, and therapeutic levels are provided below. Those skilled in the art can adjust such amounts in accordance with standard practices as needed to treat a specific subject and/or condition.
  • the therapeutically effective amount may vary based on the route of administration and dosage form, the age and weight of the subject, and/or the subject’s condition, including the type and severity of the disease or disorder.
  • ANGPTL3 and “angiopoietin like 3” refer to nucleic acid sequences encoding a ANGPTL3 protein, peptide, or polypeptide.
  • An example of an ANGPTL3 transcript is Genbank Accession No. NM_014495.4, which is incorporated by reference in its entirety.
  • ANGPTL3 may also include other ANGPTL3 encoding sequences, such as ANGPTL3 isoforms, mutant ANGPTL3 genes, splice variants of ANGPTL3 genes, and ANGPTL3 gene polymorphisms.
  • ANGPTL3 may also refer to a nucleic acid sequence that encodes the polypeptide gene product of a ANGPTL3 gene/transcript, e.g., a ANGPTL3 protein, peptide, or polypeptide according to for example Genbank Accession No. NM_014495.4 of which is incorporated by reference in its entirety.
  • ANGPTL8 refers to nucleic acid sequences encoding a ANGPTL8 protein, peptide, or polypeptide.
  • An example of an ANGPTL8 transcript is Genbank Accession No. NM_018687.7, which is incorporated by reference in its entirety.
  • the term “ANGPTL8” may also include other ANGPTL8 encoding sequences, such as ANGPTL8 isoforms, mutant ANGPTL8 genes, splice variants of ANGPTL8 genes, and ANGPTL8 gene polymorphisms.
  • ANGPTL8 may also refer to a nucleic acid sequence that encodes the polypeptide gene product of a ANGPTL8 gene/transcript, e.g., a ANGPTL8 protein, peptide, or polypeptide according to for example Genbank Accession No. NM_018687.7 of which is incorporated by reference in its entirety.
  • treatment or “treating” as used herein with reference to reducing or eliminating the disease and/or improving or ameliorating one or more symptoms of the disease.
  • prevent or “preventing” as used herein with reference to a disease refer to precluding a disease from developing in a subject at risk of developing cardiovascular disease and nonalcoholic fatty liver disease.
  • the term “pharmaceutical composition” refers to the combination of at least one active agent with a carrier, inert or active, making the composition especially suitable for diagnostic or therapeutic use in vivo or ex vivo.
  • pharmaceutically acceptable carrier refers to any of the standard pharmaceutical carriers, such as a phosphate buffered saline solution, water, emulsions (e.g., such as an oil/water or water/oil emulsions), and various types of wetting agents.
  • the compositions also can include stabilizers and preservatives. For examples of carriers, stabilizers and adjuvants, see, for example, Martin, Remington’s Pharmaceutical Sciences, 15 th Ed., Mack Publ.
  • parenteral administration and “administered parenterally” as used herein means modes of administration other than enteral and topical administration, usually by injection, and includes, without limitation, intravenous, intramuscular, intraarterial, intrathecal, intracapsular, intraorbital, intracardiac, intradermal, intraperitoneal, transtracheal, subcutaneous, subcuticular, intraarticular, subcapsular, subarachnoid, intraspinal and intrasternal injection and infusion.
  • nucleobase refers to a nitrogen-containing biological compound that forms a nucleoside. Examples of nucleobases include, but are not limited to, thymine, uracil, adenine, cytosine, guanine, and an analogue or derivative thereof.
  • RNA sequence that replaces all the U residues of an RNA sequence with T residues is “identical” to the RNA sequence, and vice versa. Accordingly, a sequence that is “identical to an RNA corresponding to” a DNA sequence constitutes the DNA sequence with all T replaced by U.
  • the presence of modified nucleotides or 2’-deoxynucleotides in a sequence does not make a sequence not “identical to an RNA” but rather a modified RNA.
  • modified nucleotide includes any nucleic acid or nucleic acid analogue residue that contains a modification or substitution in the chemical structure of an unmodified nucleotide base, sugar (including, but not limited to, 2’-substitution), or phosphate (including, but not limited to, alternate internucleotide linkers, such as phosphorothioates or the substitution of bridging oxygens in phosphate linkers with bridging sulfurs), or a combination thereof.
  • modified nucleotides are shown herein.
  • compositions are described as having, including, or comprising specific components, or where processes and methods are described as having, including, or comprising specific steps, it is contemplated that, additionally, there are compositions of the present disclosure that consist essentially of, or consist of, the recited components, and that there are processes and methods according to the present disclosure that consist essentially of, or consist of, the recited processing steps.
  • compositions specifying a percentage are by weight unless otherwise specified.
  • ANGPTL3, ANGPTL8, and Associated Diseases The siNA molecules and compositions described herein may be administered to a subject to treat a disease.
  • ANGPTL3 is an inhibitor of LPL and functions in combination with the cofactor ANGPTL8.
  • ANGPTL3 is exclusively and constitutively expressed in the liver, where ANGPTL8 is expressed in the adipose tissue and liver and is induced after fed conditions.
  • LPL breaks down fat in the form triglyceride in lipoproteins such as chylomicrons (CM) and very low-density lipoproteins (VLDL), into two free fatty acids and one monoacylglycerol molecule.
  • CM chylomicrons
  • VLDL very low-density lipoproteins
  • a disease or disorder associated with ANGPTL3 refers to a disease or disorder known in the art to be associated with altered LPL levels, activity, or both.
  • a disease or disorder associated with ANGPTL3, ANGPTL8 or ANGPTL3 and ANGPTL8 may include cardiovascular disease (CVD) and nonalcoholic fatty liver disease (NAFLD), such as metabolic dysfunction-associated steatohepatitis (MASH).
  • CVD cardiovascular disease
  • NAFLD nonalcoholic fatty liver disease
  • MASH metabolic dysfunction-associated steatohepatitis
  • Non-limiting examples of cardiovascular disease include hypertriglyceridemia (HTG) and Familial Hypercholesterolemia (FM).
  • a reporter system is used to assess the effectiveness of the siNA molecules of the siNA molecules of the present disclosure in modulating, reducing or inhibiting ANGPTL3 and ANGPTL8.
  • the reporter system is a luciferase assay. The expression of luciferase can be measured by methods well known in the art.
  • siNA targeting of a ANGPTL3 and ANGPTL8 target sequence is assessed by measuring ANGPTL3 and ANGPTL8 RNA or protein levels.
  • ANGPTL3 and ANGPTL8 RNA levels can be assessed by art-recognized methods such as RT-PCR, Northern blot, expression array, etc., and protein levels can be measured by immunoblotting, immunofluorescence, or other antibody-based methods.
  • the potency of the disclosed siNA molecules is determined by using a reporter assay.
  • the ds-siNA of the present disclosure reduces the level or activity of ANGPTL3 by more than about 10 %, more than about 20% more than about 30%, more than about 40%, more than about 50%, more than about 60%, more than about 70%, more than about 80%, more than about 90%, or more than about 95%.
  • the ds-siNA of the present disclosure reduces the level or activity of ANGPTL3 by 100%. In some embodiments, the ds-siNA of the present disclosure reduces the level or activity ANGPTL8 by more than about 10 %, more than about 20% more than about 30%, more than about 40%, more than about 50%, more than about 60%, more than about 70%, more than about 80%, more than about 90%, or more than about 95%. In some embodiments, the ds-siNA of the present disclosure reduces the level or activity of ANGPTL8 by 100%.
  • the level or activity of ANGPTL3 and ANGPTL8 can be determined by directly measuring ANGPTL3 and ANGPTL8 expression with for example RT-PCR followed by quantitative PCR.
  • the ds-siNA of the present disclosure reduces the expression level of ANGPTL3 and ANGPTL8 by more than about 10 %, more than about 20% more than about 30%, more than about 40%, more than about 50%, more than about 60%, more than about 70%, more than about 80%, more than about 90%, or more than about 95%.
  • the ds-siNA of the present disclosure reduces the expression level of ANGPTL3 and ANGPTL8 by 100%.
  • the ds- siNA of the present disclosure reduces the expression level of a ANGPTL3 and/or ANGPTL8 target genes by more than about 10 %, more than about 20% more than about 30%, more than about 40%, more than about 50%, more than about 60%, more than about 70%, more than about 80%, more than about 90%, or more than about 95%.
  • the ds- siNA of the present disclosure reduces the expression of a ANGPTL3 and/or ANGPTL8 target genes by 100%.
  • the ANGPTL3 target gene comprises NG_028169.1:5005-13798.
  • the ANGPTL8 target gene comprises NC_000019.10:11239619-11241943.
  • the EC50 (half maximum effective dosage) value of the ds-siNA molecules can be determined.
  • the ds- siNA has EC 50 value of 50 pM or less, of 40 pM or less, of 30 pM or less, of 20 pM or less, 15 pM or less, 10 pM or less, 5 pM or less, 2 pM or less, or 1 pM or less.
  • the CC50 i.e. the concentration that is required for reducing cell viability by 50%
  • the ds-siNA has a CC50 value of more than about 0.5 ⁇ M, more than about 1 ⁇ M, more than about 1.5 ⁇ M, more than about 2 ⁇ M, more than about 2.5 ⁇ M, more than about 3 ⁇ M, more than about 4 ⁇ M, or more than about 5 ⁇ M.
  • the effectiveness of the ds-siNA can be determined by measuring the expression level of ANGPTL3 and ANGPTL8, and/or a target gene of ANGPTL3 and ANGPTL8. In some embodiments, the effectiveness of the ds-siNA can be determined by measuring the expression level of ANGPTL3. In some embodiments, the effectiveness of the ds-siNA can be determined by measuring the expression level of ANGPTL8.
  • the pharmacodynamics of a GalNAc conjugated ANGPTL3 siNA, GalNAc conjugated ANGPTL8 siNA or combination regimen comprising GalNAc conjugated ANGPTL3 siNA and GalNAc conjugated ANGPTL8 siNA as disclosed here in can be determined by in vivo mice studies infected with recombinant viruses AAV-human ANGPTL3 or AAV-human ANGPTL8 or both. Serial and terminal blood collections can be tested for target protein knock down through human ANGPTL3 ELISA, or human ANGPTL8 ELISA or both.
  • Mouse livers can be tested for human NA knock down (human ANGPTL3 qPCR, or human ANMGPTL8 qPCR or both) or human target protein knockdown.
  • in vivo efficacy studies can be determined using knock in mice containing human ANGPTL3, human ANGPTL8 or both human ANGPTL3 and ANGPTL8.
  • Mice can be fed with high fat diet and treated with GalNAc conjugated ANGPTL3 siNA, GalNAc conjugated ANGPTL8 siNA or combination regimen comprising GalNAc conjugated ANGPTL3 siNA and GalNAc conjugated ANGPTL8 siNA as disclosed herein.
  • Serial and terminal blood collections can be tested for target protein knock down using ANGPTL3 ELISA, or human ANGPTL8 ELISA or both.
  • Blood LDL-c, TG, HDL-c and total cholesterol can also be measured as efficacy end points. Liver TG and other liver lipid measurements can also be applied.
  • the GalNAc conjugated ANGPTL3 siNA, GalNAc conjugated ANGPTL8 siNA or combination regimen comprising GalNAC conjugated ANGPTL3 siNA and GalNAc conjugated ANGPTL8 siNA as disclosed herein can also be tested in monkey dyslipidemia model and monitor blood LDL-c, total Cholesterol, HDL-c, TG and liver fat.
  • any of the siNA molecules disclosed herein may further comprise one or more linkers independently selected from a phosphodiester (PO) linker, phosphorothioate (PS) linker, phosphorodithioate linker, mesyl phosphoramidate (Ms), and PS-mimic linker.
  • the PS-mimic linker is a sulfur linker.
  • the linkers are internucleoside linkers.
  • the linkers may connect a nucleotide of the siNA molecule to at least one phosphorylation blocker, conjugated moiety, or 5’- stabilized end cap.
  • the linkers connect a conjugated moiety to a phosphorylation blocker or 5’-stabilized end cap.
  • the present disclosure provides empirical evidence showing that reducing expression of ANGPTL8 alone with ds-siNA, such as the ds-siNA disclosed here, can reduce cholesterol and triglyceride levels in an animal.
  • combinations of ds-siNA targeting ANGPTL3 and ds-siNA targeting ANGPTL8 can be used in combination to provide further therapeutic benefits.
  • Short interfering nucleic acid (siNA) molecules As indicated above, the present disclosure provides siNA molecules comprising modified nucleotides.
  • siNA molecules described herein may be double-stranded siNA (ds-siNA) molecules.
  • siNA molecules and “ds-siNA molecules” may be used interchangeably.
  • the ds-siNA molecules comprise a sense strand and an antisense strand.
  • Tables 1-4 details sequences of the present disclosure useful for sense and antisense strands, disclosed in SEQ ID NOs: 1-632 (ANGPTL8) and 633-1125 (ANGPTL3).
  • the double-stranded short interfering nucleic acid (ds-siNA) molecule comprises (a) a sense strand comprising a first nucleotide sequence, wherein the first nucleotide sequence is 15 to 30 nucleotides in length and comprises or consists of a nucleotide sequence as set forth in SEQ ID NOs: 1-316; and (b) an antisense strand comprising a second nucleotide sequence, wherein the second nucleotide sequence is 15 to 30 nucleotides in length and comprises or consists of a nucleotide sequence that is the reverse complement of the first nucleotide sequence or has a nucleic acid sequence with sufficient complementarity (i.e., not necessarily 100%) to hybridize to the first nucleotide sequence.
  • the first nucleotide sequence comprises or consists of a nucleotide sequence as set forth in SEQ ID NOs: 1-316; and the second nucleotide sequence comprises or consists of a nucleotide sequence according to SEQ ID NOs: 317-632.
  • the double-stranded short interfering nucleic acid (ds-siNA) molecule comprises (a) a sense strand comprising a first nucleotide sequence, wherein the first nucleotide sequence is 15 to 30 nucleotides in length and comprises or consists of a nucleotide sequence as set forth in SEQ ID NOs: 633-878; and (b) an antisense strand comprising a second nucleotide sequence, wherein the second nucleotide sequence is 15 to 30 nucleotides in length and comprises or consists of a nucleotide sequence that is the reverse complement of the first nucleotide sequence or has a nucleic acid sequence with sufficient complementarity (i.e., not necessarily 100%) to hybridize to the first nucleotide sequence.
  • the first nucleotide sequence comprises or consists of a nucleotide sequence as set forth in SEQ ID NOs: 633-878; and the second nucleotide sequence comprises or consists of a nucleotide sequence according to SEQ ID NOs: 879-1125.
  • the double-stranded short interfering nucleic acid (ds-siNA) molecule comprises (a) a sense strand comprising a first nucleotide sequence, wherein the first nucleotide sequence is 15 to 30 nucleotides in length and comprises a nucleotide sequence with 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 99%, or 100% similarity to SEQ ID NOs: 1-316; and (b) an antisense strand comprising a second nucleotide sequence, wherein the second nucleotide sequence is 15 to 30 nucleotides in length and comprises a nucleotide sequence that is the reverse complement of the first nucleotide sequence.
  • the first nucleotide sequence comprises a nucleotide sequence with 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 99%, or 100% similarity to SEQ ID NOs:1-316; and the second nucleotide sequence comprises a nucleotide sequence with 75%, 80%, 85%, 90%, 91 %, 92%, 93%, 94%, 95%, 99%, or 100% similarity to SEQ ID NOs: 317-632.
  • the double-stranded short interfering nucleic acid (ds-siNA) molecule comprises (a) a sense strand comprising a first nucleotide sequence, wherein the first nucleotide sequence is 15 to 30 nucleotides in length and comprises a nucleotide sequence with 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 99%, or 100% similarity to SEQ ID NOs: 633-878; and (b) an antisense strand comprising a second nucleotide sequence, wherein the second nucleotide sequence is 15 to 30 nucleotides in length and comprises a nucleotide sequence that is the reverse complement of the first nucleotide sequence.
  • the first nucleotide sequence comprises a nucleotide sequence with 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 99%, or 100% similarity to SEQ ID NOs:633-878; and the second nucleotide sequence comprises a nucleotide sequence with 75%, 80%, 85%, 90%, 91 %, 92%, 93%, 94%, 95%, 99%, or 100% similarity to SEQ ID NOs: 879-1125.
  • the double-stranded short interfering nucleic acid (ds-siNA) molecule comprises (a) a sense strand comprising a first nucleotide sequence, wherein the first nucleotide sequence is 15 to 30 nucleotides in length and comprises a nucleotide sequence that differs by 1, 2, 3, 4, or 5 nucleotides from any one of SEQ ID NOs:1-316; and (b) an antisense strand comprising a second nucleotide sequence, wherein the second nucleotide sequence is 15 to 30 nucleotides in length and comprises a nucleotide sequence that is the reverse complement of the first nucleotide sequence.
  • the second nucleotide sequence comprises a nucleotide sequence that differs by 1, 2, 3, 4, or 5 nucleotides from any one of SEQ ID NOs:317-632.
  • the double-stranded short interfering nucleic acid (ds-siNA) molecule comprises (a) a sense strand comprising a first nucleotide sequence, wherein the first nucleotide sequence is 15 to 30 nucleotides in length and comprises a nucleotide sequence that differs by 1, 2, 3, 4, or 5 nucleotides from any one of SEQ ID NOs:633-878; and (b) an antisense strand comprising a second nucleotide sequence, wherein the second nucleotide sequence is 15 to 30 nucleotides in length and comprises a nucleotide sequence that is the reverse complement of the first nucleotide sequence.
  • the second nucleotide sequence comprises a nucleotide sequence that differs by 1, 2, 3, 4, or 5 nucleotides from any one of SEQ ID NOs:879-1125.
  • the present disclosure provides a double-stranded short interfering nucleic acid (ds-siNA) molecule comprising (a) a sense strand comprising a nucleotide sequence identical to SEQ ID NOs: 1-316, and (b) an antisense strand.
  • ds-siNA double-stranded short interfering nucleic acid
  • the present disclosure provides a double-stranded short interfering nucleic acid (ds-siNA) molecule comprising (a) an antisense strand comprising a nucleotide sequence identical to SEQ ID NOs: 317-632; and (b) a sense strand.
  • the present disclosure provides a double-stranded short interfering nucleic acid (ds-siNA) molecule comprising (a) a sense strand comprising a nucleotide sequence identical to SEQ ID NOs: 633-878, and (b) an antisense strand.
  • the present disclosure provides a double-stranded short interfering nucleic acid (ds-siNA) molecule comprising (a) an antisense strand comprising a nucleotide sequence identical to SEQ ID NOs: 879-1125; and (b) a sense strand.
  • ds-siNA double-stranded short interfering nucleic acid
  • the present disclosure relates to a double-stranded short interfering nucleic acid (ds-siNA) molecule comprising: (a) a sense strand comprising a first nucleotide sequence, wherein the first nucleotide sequence is 15 to 30 nucleotides in length and comprises a nucleotide sequence as set forth in SEQ ID NOs: 1-316, and an antisense strand comprising a second nucleotide sequence, wherein the second nucleotide sequence is 15 to 30 nucleotides in length and comprises a nucleotide sequence that is a substantial reverse complement of the first nucleotide sequence; or (b) a sense strand comprising a first nucleotide sequence, wherein the first nucleotide sequence is 15 to 30 nucleotides in length and comprises a nucleotide sequence that is a substantial reverse complement of a second nucleotide sequence; and an antisense strand comprising the second nucleot
  • the present disclosure relates to a double-stranded short interfering nucleic acid (ds-siNA) molecule comprising: (a) a sense strand comprising a first nucleotide sequence, wherein the first nucleotide sequence is 15 to 30 nucleotides in length and comprises a nucleotide sequence as set forth in SEQ ID NOs: 633-878, and an antisense strand comprising a second nucleotide sequence, wherein the second nucleotide sequence is 15 to 30 nucleotides in length and comprises a nucleotide sequence that is a substantial reverse complement of the first nucleotide sequence; or (b) a sense strand comprising a first nucleotide sequence, wherein the first nucleotide sequence is 15 to 30 nucleotides in length and comprises a nucleotide sequence that is a substantial reverse complement of a second nucleotide sequence; and an antisense strand comprising the second nucle
  • the present disclosure relates to a double-stranded short interfering nucleic acid (ds-siNA) molecule comprising (a) a sense strand comprising a nucleotide sequence identical to SEQ ID NOs: 1-316, and (b) an antisense strand.
  • the present disclosure relates to a double-stranded short interfering nucleic acid (ds-siNA) molecule comprising (a) a sense strand comprising a nucleotide sequence identical to SEQ ID NOs: 633-878, and (b) an antisense strand.
  • the present disclosure relates to a double-stranded short interfering nucleic acid (ds-siNA) molecule comprising (a) an antisense strand comprising a nucleotide sequence identical to SEQ ID NOs: 317-632, and (b) a sense strand.
  • the present disclosure relates to a double-stranded short interfering nucleic acid (ds-siNA) molecule comprising (a) an antisense strand comprising a nucleotide sequence identical to SEQ ID NOs: 879-1125, and (b) a sense strand.
  • the present disclosure provides a ds-siNA comprising a sense strand comprising any one of SEQ ID NOs: 633, 634, 643, 653, 654, 674, 687, 688, 690-693, 694, 708-713, 756-757, 756, 776, 777, 797, 810, 811, 813-817, or 831-836; and an antisense strand comprising any one of SEQ ID NOs: 879, 880, 888, 899, 900, 920, 933, 934, 936-940, 954-959, 1003-1004, 1012, 1023, 1024, 1044, 1057-1064, or 1078-1083, respectively.
  • the present disclosure provides a ds-siNA comprising a sense strand comprising any one of SEQ ID NOs: 633, 654, 688, 690, 708, 713, 756, 777, 811, 813, 831, or 836, and an antisense strand comprising any one of SEQ ID NOs: 879, 900, 934, 936, 954, 959, 1003, 1024, 1058, 1060, 1078, or 1083, respectively.
  • the disclosed siNA molecules may comprise (a) at least one phosphorylation blocker, conjugated moiety, or 5’-stabilized end cap; and (b) a short interfering nucleic acid (siNA).
  • the phosphorylation blocker is a phosphorylation blocker disclosed herein.
  • the 5’-stabilized end cap is a 5’-stabilized end cap disclosed herein.
  • the siNA may comprise any of the first nucleotide, second nucleotide, sense strand, or antisense strand sequences disclosed herein.
  • the siNA may comprise 5 to 100, 5 to 90, 10 to 100, 10 to 90, 10 to 80, 10 to 70, 10 to 60, 10 to 50, 10 to 30, 10 to 25, 15 to 100, 15 to 90, 15 to 80, 15 to 70, 15 to 60, 15 to 50, 15 to 30, or 15 to 25 nucleotides.
  • the siNA may comprise at least 5, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, or 40 nucleotides.
  • the siNA may comprise less than or equal to 50, 45, 40, 39, 38, 37, 36, 35, 34, 33, 32, 31, 30, 29, 28, 27, 26, 25, 24, 23, 22, 21, 20, or 19 nucleotides.
  • the nucleotides may be modified nucleotides.
  • the siNA may be single stranded.
  • the siNA may be double stranded.
  • the siNA may comprise (a) a sense strand comprising 15 to 30, 15 to 25, 15 to 24, 15 to 23, 15 to 22, 15 to 21, 17 to 30, 17 to 25, 17 to 24, 17 to 23, 17 to 22, 17 to 21, 18 to 30, 18 to 25, 18 to 24, 18 to 23, 18 to 22, 18 to 21, 19 to 30, 19 to 25, 19 to 24, 19 to 23, 19 to 22, 19 to 21, 20 to 25, 20 to 24, 20 to 23, 21 to 25, 21 to 24, or 21 to 23 nucleotides; and (b) an antisense strand comprising 15 to 30, 15 to 25, 15 to 24, 15 to 23, 15 to 22, 15 to 21, 17 to 30, 17 to 25, 17 to 24, 17 to 23, 17 to 22, 17 to 21, 18 to 30, 18 to 25, 18 to 24, 18 to 23, 18 to 22, 18 to 21, 19 to 30, 19 to 25, 19 to 24, 19 to 23, 19 to 22, 19 to 21, 20 to 25, 20 to 24, 20 to 23, 21 to 25, 21 to 24, or 21 to 23 nucleotides.
  • the siNA may comprise (a) a sense strand comprising about 15, 16, 17, 18, 19, 20, 21, 22, or 23 nucleotides; and (b) an antisense strand comprising about 15, 16, 17, 18, 19, 20, 21, 22, or 23 nucleotides.
  • the siNA may comprise (a) a sense strand comprising about 19 nucleotides; and (b) an antisense strand comprising about 21 nucleotides.
  • the siNA may comprise (a) a sense strand comprising about 21 nucleotides; and (b) an antisense strand comprising about 23 nucleotides.
  • any of the siNA molecules disclosed herein may further comprise one or more linkers independently selected from a phosphodiester (PO) linker, phosphorothioate (PS) linker, phosphorodithioate linker, mesyl phosphoramidate (Ms), and PS-mimic linker.
  • the PS-mimic linker is a 3’ or 5’ sulfur linker.
  • the linkers are internucleoside linkers.
  • the linkers may connect a nucleotide of the siNA molecule to at least one phosphorylation blocker, conjugated moiety, or 5’-stabilized end cap.
  • the linkers connect a conjugated moiety to a phosphorylation blocker or 5’-stabilized end cap.
  • at least one end of the ds-siNA comprises an overhang, wherein the overhang comprises at least one nucleotide, preferably wherein the at least one nucleotide comprises a 2’-O-methyl nucleotide, a deoxy nucleotide, uracil, or a uracil analogue or derivative.
  • both ends of the ds-siNA comprise an overhang, wherein the overhang comprises at least one nucleotide, preferably wherein the at least one nucleotide comprises a 2’-O-methyl nucleotide, a deoxy nucleotide, uracil, or a uracil analogue or derivative.
  • a ds-siNA molecule may have the following formula: 5’-An 1 Bn 2 An 3 Bn 4 An 5 Bn 6 An 7 Bn 8 An 9 -3’ 3’-Cq 1 Aq 2 Bq 3 A q 4 Bq 5 Aq 6 Bq 7 Aq 8 Bq 9 Aq 10 Bq 11 Aq 12 -5’ wherein: the top strand is a sense strand comprising a first nucleotide sequence that is at least about 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, or 100% identical to an RNA corresponding to a target gene, wherein the first nucleotide sequence comprises 15 to 30 nucleotides; the bottom strand is an antisense strand comprising a second nucleotide sequence that is at least about 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, or
  • the ds-siNA may further comprise a conjugated moiety.
  • the conjugated moiety may comprise any of the galactosamines disclosed herein.
  • the ds-siNA may further comprise (i) phosphorothioate internucleoside linkages between the nucleotides at positions 1 and 2 and positions 2 and 3 from the 5’ end of the sense strand; and (ii) phosphorothioate internucleoside linkages between any nucleotide position, for example at positions 1 and 2; positions 2 and 3; positions 3 and 4 etc.
  • the ds-siNA may further comprise a 5’-stabilizing end cap.
  • the 5’-stabilizing end cap may be a 2’-OMe-vinyl phosphonate (vm).
  • the 5’- stabilizing end cap may feature a modified base (e.g., vm(5mim), and vm(56mido)), as disclosed herein.
  • the 5’-stabilizing end cap may be attached to the 5’ end of the antisense strand.
  • the 2’-O-methyl nucleotide at position 1 from the 5’ end of the sense strand is further modified to contain a 5’ stabilizing end cap.
  • the 2’-O-methyl nucleotide at position 1 from the 5’ end of the antisense strand is further modified to contain a 5’ stabilizing end cap.
  • the 2’-O-methyl nucleotide at position 1 from the 5’ end of the sense strand is further modified to contain a phosphorylation blocker. In some embodiments, the 2’-O-methyl nucleotide at position 1 from the 3’ end of the sense strand is further modified to contain a phosphorylation blocker. In some embodiments, the 2’-O-methyl nucleotide at position 1 from the 5’ end of the antisense strand is further modified to contain a phosphorylation blocker. In some embodiments, the 2’-O-methyl nucleotide at position 1 from the 3’ end of the antisense strand is further modified to contain a phosphorylation blocker.
  • An exemplary ds-siNA molecule may have the following formula: 5’-A2-4 B1A1-3 B2-3 A2-10 B0-1A0-4B0-1 A0-2-3’ 3’-C2A0-2B0-1A0-3B0-1A0-5B0-1A2-7 B1A2-11 B1A1-5’ wherein: the top strand is a sense strand comprising a first nucleotide sequence that is at least about 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, or 100% identical to an RNA corresponding to a target gene, wherein the first nucleotide sequence comprises 15 to 30 nucleotides; the bottom strand is an antisense strand comprising a second nucleotide sequence that is at least about 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, or 100% complementary to the RNA corresponding to the target gene
  • the ds-siNA may further comprise a conjugated moiety.
  • the conjugated moiety may comprise any of the galactosamines disclosed herein.
  • the ds-siNA may further comprise (i) phosphorothioate internucleoside linkages between the nucleotides at positions 1 and 2 and positions 2 and 3 from the 5’ end of the sense strand; and (ii) phosphorothioate internucleoside linkages between the nucleotides at positions 1 and 2; positions 2 and 3; positions 19 and 20; and positions 20 and 21 from the 5’ end of the antisense strand.
  • the ds- siNA may further comprise a 5’-stabilizing end cap.
  • the 5’-stabilizing end cap may include a vinyl phosphonate.
  • the vinyl phosphonate may be a deuterated vinyl phosphonate.
  • the deuterated vinyl phosphonate may be a mono-deuterated vinyl phosphonate.
  • the deuterated vinyl phosphonate may be a mono- or di-deuterated vinyl phosphonate.
  • the 5’ end of the antisense strand may comprise a wherein B is a nucleobase or derivative thereof), a a (vm(56mido)), or a (vm(56amim)U, wherein R is H or Bz).
  • the 5’-stabilizing end cap may be attached to the 5’ end of the antisense strand.
  • the 5’-stabilizing end cap may be attached to the 3’ end of the antisense strand.
  • the 5’-stabilizing end cap may be attached to the 5’ end of the sense strand.
  • the 5’-stabilizing end cap may be attached to the 3’ end of the sense strand.
  • the 2’-O-methyl nucleotide at position 1 from the 5’ end of the sense strand is further modified to contain a 5’ stabilizing end cap.
  • the 2’-O-methyl nucleotide at position 1 from the 5’ end of the antisense strand is further modified to contain a 5’ stabilizing end cap.
  • the 2’-O-methyl nucleotide at position 1 from the 5’ end of the sense strand is further modified to contain a phosphorylation blocker.
  • the 2’-O-methyl nucleotide at position 1 from the 3’ end of the sense strand is further modified to contain a phosphorylation blocker.
  • the 2’- O-methyl nucleotide at position 1 from the 5’ end of the antisense strand is further modified to contain a phosphorylation blocker.
  • the 2’-O-methyl nucleotide at position 1 from the 3’ end of the antisense strand is further modified to contain a phosphorylation blocker.
  • the ds-siNA may optionally further comprise (iii) a conjugated moiety, wherein the conjugated moiety (e.g GalNAc4 is attached to the 3’ end or the 5’ end of the sense strand or the antisense strand.
  • the ds-siNA may comprise a 2 nucleotide overhang at positions 22 and 23 from the 5’ end of the antisense strand.
  • the ds- siNA may further comprise 1, 2, 3, 4, 5, 6 or more phosphorothioate (ps) internucleoside linkages or mesyl phosphoramidate internucleoside linkage (Ms). At least one phosphorothioate internucleoside linkage or mesyl phosphoramidate internucleoside linkage (Ms) may be between the nucleotides at positions 1 and 2 or positions 2 and 3 from the 5’ end of the sense strand.
  • At least one phosphorothioate internucleoside linkage or mesyl phosphoramidate internucleoside linkage may be between the nucleotides at positions 1 and 2 or positions 2 and 3 from the 5’ end of the antisense strand. At least one phosphorothioate internucleoside linkage or mesyl phosphoramidate internucleoside linkage (Ms) may be between the nucleotides at positions 19 and 20, positions 20 and 21, positions 21 and 22, or positions 22 and 23 from the 5’ end of the antisense strand or any combination thereof. As shown in FIGs.
  • nucleotides in the sense strand may be 2’-fluoro nucleotides.
  • about 4 nucleotides (e.g., 1, 2, 3, 4, 5, or 6) in the antisense strand may be 2’-fluoro nucleotides.
  • 15-17 nucleotides in the sense strand may be 2’-O-methyl nucleotides.
  • 16-21 nucleotides in the antisense strand may be 2’-O-methyl nucleotides.
  • the 2’-O-methyl nucleotide at position 1 from the 5’ end of the sense strand can be further modified to contain a 5’ stabilizing end cap. In some embodiments, the 2’-O-methyl nucleotide at position 1 from the 5’ end of the antisense strand can be further modified to contain a 5’ stabilizing end cap. In some embodiments, the 2’-O- methyl nucleotide at position 1 from the 5’ end of the sense strand can be further modified to contain a phosphorylation blocker. In some embodiments, the 2’-O-methyl nucleotide at position 1 from the 3’ end of the sense strand can be further modified to contain a phosphorylation blocker.
  • the 2’-O-methyl nucleotide at position 1 from the 5’ end of the antisense strand can be further modified to contain a phosphorylation blocker. In some embodiments, the 2’-O-methyl nucleotide at position 1 from the 3’ end of the antisense strand can be further modified to contain a phosphorylation blocker.
  • the 2’-O-methyl nucleotide at position 1 from the 5’ end of the sense strand is a d2vd3 nucleotide, a d2vd3U nucleotide, a d2vd3A nucleotide, an omeco- d3 nucleotide, an omeco-d3U nucleotide, an omeco-d3A nucleotide, a 4h nucleotide, a 4hU nucleotide, a 4hA nucleotide, a v-mun nucleotide, a c2o-4h nucleotide, an omeco-munb nucleotide, a d2vm nucleotide, or a d2vmA nucleotide.
  • the 2’-O- methyl nucleotide at position 1 from the 5’ end of the antisense strand is a d2vd3 nucleotide, a d2vd3U nucleotide, a d2vd3A nucleotide, an omeco-d3 nucleotide, an omeco-d3U nucleotide, an omeco-d3A nucleotide, a 4h nucleotide, a 4hU nucleotide, a 4hA nucleotide, a v-mun nucleotide, a c2o-4h nucleotide, an omeco-munb nucleotide, a d2vm nucleotide, or a d2vmA nucleotide.
  • the 2’-O-methyl nucleotide at position 1 from the 3’ end of the sense strand is a d2vd3 nucleotide, a d2vd3U nucleotide, a d2vd3A nucleotide, an omeco-d3 nucleotide, an omeco-d3U nucleotide, an omeco-d3A nucleotide, a 4h nucleotide, a 4hU nucleotide, a 4hA nucleotide, a v-mun nucleotide, a c2o-4h nucleotide, an omeco-munb nucleotide, a d2vm nucleotide, or a d2vmA nucleotide.
  • the 2’-O-methyl nucleotide at position 1 from the 3’ end of the antisense strand is a d2vd3 nucleotide, a d2vd3U nucleotide, d2vd3A nucleotide, an omeco-d3 nucleotide, an omeco- d3U nucleotide, an omeco-d3A nucleotide, a 4h nucleotide, a 4hU nucleotide, a v-mun nucleotide, a c2o-4h nucleotide, an omeco-munb nucleotide, a d2vm nucleotide, or a d2vmA nucleotide.
  • At least 1, 2, 3, 4 or more 2’-fluoro nucleotides on the sense strand or antisense strand is a 2’-fluoro nucleotide mimic. In some embodiments, at least 1, 2, 3, 4 or more 2’-fluoro nucleotides on the sense strand or antisense strand is a fB, fN, f(4nh)Q, f4P, f2P, or fX nucleotide. In some embodiments, at least 1, 2, 3, 4 or more 2’- O-methyl nucleotide on the sense or antisense strand is a 2’-O-methyl nucleotide mimic.
  • one or more nucleotides in the sense strand and/or the antisense strand may be a 3’,4’ seco modified nucleotide in which the bond between the 3’ and 4’ positions of the furanose ring is broken (e.g., mun34).
  • the antisense strand, sense strand, first nucleotide sequence, and/or second nucleotide sequence comprises at least one modified nucleotide comprising (a) wherein Rx is a nucleobase, aryl, heteroaryl, or H, (b) O (mun34), wherein R y is a nucleobase, (c) (F4P), or (d)
  • the second nucleotide sequence (or antisense strand) comprises a modified nucleotide comprising at least
  • the sense strand of the siNA, the antisense strand or the siNA, or both comprise at least 1, at least 2, at least 3, at least 4, or at least 5 nucleotide(s) comprising a structure independently selected from:
  • nucleotide analog(s) comprising a structure independently selected from: (gans ⁇ ) and (ganr ⁇ ), wherein B is a nucleobase, an aryl, heteroaryl, or H; wherein represents a phosphodiester linkage, a phosphorothioate linkage, or H; and wherein * represent chiral center (e.g., R or S isomer).
  • the sense strand, the antisense strand, or both each independently comprise 1 or more phosphorothioate internucleoside linkages.
  • the siNA further comprises a phosphorylation blocker.
  • the sense strand of the siNA, the antisense strand of the siNA, or both each independently comprise at least 1, at least 2, at least 3, at least 4, or at least 5 or more of wherein Rx is a nucleobase, aryl, heteroaryl, or H, (mun34) wherein R y is a nucleobase, (tn) wherein R y is a nucleobase, or combinations thereof.
  • the siNA further comprises a galactosamine.
  • GalNAc N-acetylgalactosamine
  • the galactosamine is N-acetylgalactosamine (GalNAc) of Formula (VII): , wherein R z is OH or SH; and each n is independently 1 or 2.
  • A. siNA sense strand Any of the siNA molecules described herein may comprise a sense strand.
  • the sense strand may comprise a first nucleotide sequence.
  • the first nucleotide sequence may be 15 to 30, 15 to 25, 15 to 23, 17 to 23, 19 to 23, or 19 to 21 nucleotides in length. In some embodiments, the first nucleotide sequence is 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30 nucleotides in length.
  • the first nucleotide sequence is at least 19 nucleotides in length. In some embodiments, the first nucleotide sequence is at least 21 nucleotides in length. In some embodiments, the sense strand is the same length as the first nucleotide sequence. In some embodiments, the sense strand is longer than the first nucleotide sequence. In some embodiments, the sense strand may further comprise 1, 2, 3, 4, or 5 or more nucleotides than the first nucleotide sequence. In some embodiments, the sense strand may further comprise a deoxyribonucleic acid (DNA). In some embodiments, the DNA is thymine (T). In some embodiments, the sense strand may further comprise a TT sequence.
  • DNA is thymine (T).
  • the sense strand may further comprise one or more modified nucleotides that are adjacent to the first nucleotide sequence.
  • the one or more modified nucleotides are independently selected from any of the modified nucleotides disclosed herein (e.g., 2’-fluoro nucleotide, 2’-O-methyl nucleotide, 2’-fluoro nucleotide mimic, 2’-O-methyl nucleotide mimic, or a nucleotide comprising a modified nucleobase).
  • the first nucleotide sequence comprises 15, 16, 17, 18, 19, 20, 21, 22, 23, or more modified nucleotides independently selected from a 2’-O-methyl nucleotide and a 2′-fluoro nucleotide.
  • 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95% or 100% of the nucleotides in the first nucleotide sequence are modified nucleotides independently selected from a 2’-O-methyl nucleotide and a 2’-fluoro nucleotide.
  • 100% of the nucleotides in the first nucleotide sequence are modified nucleotides independently selected from a 2’-O-methyl nucleotide and a 2’- fluoro nucleotide.
  • the 2’-O-methyl nucleotide is a 2’-O-methyl nucleotide mimic.
  • the 2’-fluoro nucleotide is a 2’-fluoro nucleotide mimic.
  • nucleotides of the first nucleotide sequence are 2’-O-methyl nucleotides. In some embodiments, between about 2 to 20 modified nucleotides of the first nucleotide sequence are 2’-O-methyl nucleotides.
  • between about 5 to 25 modified nucleotides of the first nucleotide sequence are 2’-O-methyl nucleotides. In some embodiments, between about 10 to 25 modified nucleotides of the first nucleotide sequence are 2’-O-methyl nucleotides. In some embodiments, between about 12 to 25 modified nucleotides of the first nucleotide sequence are 2’-O-methyl nucleotides. In some embodiments, at least about 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, or 22 modified nucleotides of the first nucleotide sequence are 2’-O- methyl nucleotides.
  • At least about 12 modified nucleotides of the first nucleotide sequence are 2’-O-methyl nucleotides. In some embodiments, at least about 13 modified nucleotides of the first nucleotide sequence are 2’-O-methyl nucleotides. In some embodiments, at least about 14 modified nucleotides of the first nucleotide sequence are 2’- O-methyl nucleotides. In some embodiments, at least about 15 modified nucleotides of the first nucleotide sequence are 2’-O-methyl nucleotides. In some embodiments, at least about 16 modified nucleotides of the first nucleotide sequence are 2’-O-methyl nucleotides.
  • At least about 17 modified nucleotides of the first nucleotide sequence are 2’-O-methyl nucleotides. In some embodiments, at least about 18 modified nucleotides of the first nucleotide sequence are 2’-O-methyl nucleotides. In some embodiments, at least about 19 modified nucleotides of the first nucleotide sequence are 2’-O-methyl nucleotides. In some embodiments, less than or equal to 25, 24, 23, 22, 21, 20, 19, 18, 17, 16, 15, 14, 13, 12, 11, 10, 9, 8, 7, 6, 5, 4, 3, or 2 modified nucleotides of the first nucleotide sequence are 2’- O-methyl nucleotides.
  • less than or equal to 21 modified nucleotides of the first nucleotide sequence are 2’-O-methyl nucleotides. In some embodiments, less than or equal to 20 modified nucleotides of the first nucleotide sequence are 2’-O-methyl nucleotides. In some embodiments, less than or equal to 19 modified nucleotides of the first nucleotide sequence are 2’-O-methyl nucleotides. In some embodiments, less than or equal to 18 modified nucleotides of the first nucleotide sequence are 2’-O-methyl nucleotides.
  • less than or equal to 17 modified nucleotides of the first nucleotide sequence are 2’-O-methyl nucleotides. In some embodiments, less than or equal to 16 modified nucleotides of the first nucleotide sequence are 2’-O-methyl nucleotides. In some embodiments, less than or equal to 15 modified nucleotides of the first nucleotide sequence are 2’-O-methyl nucleotides. In some embodiments, less than or equal to 14 modified nucleotides of the first nucleotide sequence are 2’-O-methyl nucleotides.
  • modified nucleotides of the first nucleotide sequence are 2’-O-methyl nucleotides. In some embodiments, at least one modified nucleotide of the first nucleotide sequence is a 2’-O-methyl pyrimidine. In some embodiments, at least 5, 6, 7, 8, 9, or 10 modified nucleotides of the first nucleotide sequence are 2’-O-methyl pyrimidines. In some embodiments, at least one modified nucleotide of the first nucleotide sequence is a 2’-O-methyl purine. In some embodiments, at least 5, 6, 7, 8, 9, or 10 modified nucleotides of the first nucleotide sequence are 2’-O-methyl purines.
  • the 2’-O- methyl nucleotide is a 2’-O-methyl nucleotide mimic. In some embodiments, between 2 to 15 modified nucleotides of the first nucleotide sequence are 2’-fluoro nucleotides. In some embodiments, between 2 to 10 modified nucleotides of the first nucleotide sequence are 2’-fluoro nucleotides. In some embodiments, between 2 to 6 modified nucleotides of the first nucleotide sequence are 2’-fluoro nucleotides. In some embodiments, 1 to 6, 1 to 5, 1 to 4, or 1 to 3 modified nucleotides of the first nucleotide sequence are 2’-fluoro nucleotides.
  • At least 1, 2, 3, 4, 5, or 6 modified nucleotides of the first nucleotide sequence are 2’-fluoro nucleotides. In some embodiments, at least 1 modified nucleotide of the first nucleotide sequence is a 2’- fluoro nucleotide. In some embodiments, at least 2 modified nucleotides of the first nucleotide sequence are 2’-fluoro nucleotides. In some embodiments, at least 3 modified nucleotides of the first nucleotide sequence are 2’-fluoro nucleotides. In some embodiments, at least 4 modified nucleotides of the first nucleotide sequence are 2’-fluoro nucleotides.
  • At least 5 modified nucleotides of the first nucleotide sequence are 2’- fluoro nucleotides. In some embodiments, at least 6 modified nucleotides of the first nucleotide sequence are 2’-fluoro nucleotides. In some embodiments, 10, 9, 8, 7, 6, 5, 4, 3 or fewer modified nucleotides of the first nucleotide sequence are 2’-fluoro nucleotides. In some embodiments, 10 or fewer modified nucleotides of the first nucleotide sequence are 2’-fluoro nucleotides. In some embodiments, 7 or fewer modified nucleotides of the first nucleotide sequence are 2’-fluoro nucleotides.
  • 6 or fewer modified nucleotides of the first nucleotide sequence are 2’-fluoro nucleotides. In some embodiments, 5 or fewer modified nucleotides of the first nucleotide sequence are 2’-fluoro nucleotides. In some embodiments, 4 or fewer modified nucleotides of the first nucleotide sequence are 2’-fluoro nucleotides. In some embodiments, 3 or fewer modified nucleotides of the first nucleotide sequence are 2’-fluoro nucleotides. In some embodiments, 2 or fewer modified nucleotides of the first nucleotide sequence are 2’-fluoro nucleotides.
  • At least one modified nucleotide of the first nucleotide sequence is a 2’-fluoro pyrimidine. In some embodiments, 1, 2, 3, 4, 5, or 6 modified nucleotides of the first nucleotide sequence are 2’- fluoro pyrimidines. In some embodiments, at least one modified nucleotide of the first nucleotide sequence is a 2’-fluoro purine. In some embodiments, 1, 2, 3, 4, 5, or 6 modified nucleotides of the first nucleotide sequence are 2’-fluoro purines. In some embodiments, the 2’-fluoro nucleotide is a 2’-fluoro nucleotide mimic.
  • the nucleotide at position 3, 5, 7, 8, 9, 10, 11, 12, 14, 17, and/or 19 from the 5’ end of the first nucleotide sequence is a 2’-fluoro nucleotide. In some embodiments, at least two nucleotides at positions 3, 5, 7, 8, 9, 10, 11, 12, 14, 17, and/or 19 from the 5’ end of the first nucleotide sequence are 2’-fluoro nucleotides. In some embodiments, at least three nucleotides at positions 3, 5, 7, 8, 9, 10, 11, 12, 14, 17, and/or 19 from the 5’ end of the first nucleotide sequence are 2’-fluoro nucleotides.
  • At least four nucleotides at positions 3, 5, 7, 8, 9, 10, 11, 12, 14, 17, and/or 19 from the 5’ end of the first nucleotide sequence are 2’-fluoro nucleotides. In some embodiments, at least five nucleotides at positions 3, 5, 7, 8, 9, 10, 11, 12, 14, 17, and/or 19 from the 5’ end of the first nucleotide sequence are 2’-fluoro nucleotides. In some embodiments, the nucleotides at positions 3, 5, 7, 8, 9, 10, 11, 12, 14, 17, and/or 19 from the 5’ end of the first nucleotide sequence are 2’-fluoro nucleotides.
  • the nucleotide at position 3 from the 5’ end of the first nucleotide sequence is a 2’-fluoro nucleotide. In some embodiments, the nucleotide at position 7 from the 5’ end of the first nucleotide sequence is a 2’-fluoro nucleotide. In some embodiments, the nucleotide at position 8 from the 5’ end of the first nucleotide sequence is a 2’-fluoro nucleotide. In some embodiments, the nucleotide at position 9 from the 5’ end of the first nucleotide sequence is a 2’-fluoro nucleotide.
  • the nucleotide at position 12 from the 5’ end of the first nucleotide sequence is a 2’-fluoro nucleotide. In some embodiments, the nucleotide at position 17 from the 5’ end of the first nucleotide sequence is a 2’-fluoro nucleotide. In some embodiments, the 2’-fluoro nucleotide is a 2’-fluoro nucleotide mimic. In some embodiments, at least 1, 2, 3, 4, 5, 6, or 7 nucleotides at position 3, 5, 7, 8, 9, 10, 11, 12, 14, 17, and/or 19 from the 5’ end of the first nucleotide sequence is a 2’-fluoro nucleotide.
  • the nucleotide at positions 3, 5, 7, 8, 9, 10, 11, 12, 14, 17, and/or 19 from the 5’ end of the first nucleotide sequence is a 2’-fluoro nucleotide. In some embodiments, at least two nucleotides at positions 3, 5, 7, 8, 9, 10, 11, 12, 14, 17, and/or 19 from the 5’ end of the first nucleotide sequence are 2’-fluoro nucleotides. In some embodiments, at least three nucleotides at positions 3, 5, 7, 8, 9, 10, 11, 12, 14, 17, and/or 19 from the 5’ end of the first nucleotide sequence are 2’-fluoro nucleotides.
  • the nucleotides at positions 3, 5, 7, 8, 9, 10, 11, 12, 14, 17, and/or 19 from the 5’ end of the first nucleotide sequence are 2’-fluoro nucleotides. In some embodiments, the nucleotide at position 3 from the 5’ end of the first nucleotide sequence is a 2’-fluoro nucleotide. In some embodiments, the nucleotide at position 5 from the 5’ end of the first nucleotide sequence is a 2’-fluoro nucleotide. In some embodiments, the nucleotide at position 7 from the 5’ end of the first nucleotide sequence is a 2’-fluoro nucleotide.
  • the nucleotide at position 8 from the 5’ end of the first nucleotide sequence is a 2’-fluoro nucleotide. In some embodiments, the nucleotide at position 9 from the 5’ end of the first nucleotide sequence is a 2’-fluoro nucleotide. In some embodiments, the nucleotide at position 10 from the 5’ end of the first nucleotide sequence is a 2’-fluoro nucleotide. In some embodiments, the nucleotide at position 11 from the 5’ end of the first nucleotide sequence is a 2’-fluoro nucleotide.
  • the nucleotide at position 12 from the 5’ end of the first nucleotide sequence is a 2’-fluoro nucleotide. In some embodiments, the nucleotide at position 14 from the 5’ end of the first nucleotide sequence is a 2’-fluoro nucleotide. In some embodiments, the nucleotide at position 17 from the 5’ end of the first nucleotide sequence is a 2’-fluoro nucleotide. In some embodiments, the nucleotide at position 19 from the 5’ end of the first nucleotide sequence is a 2’-fluoro nucleotide.
  • the nucleotide at position 3, 7, 8, 9, 12, and/or 17 from the 5’ end of the first nucleotide sequence is a 2’-fluoro nucleotide. In some embodiments, the nucleotide at position 3, 7, 8, and/or 17 from the 5’ end of the first nucleotide sequence is a 2’-fluoro nucleotide. In some embodiments, the nucleotide at position 3, 7, 8, 9, 12, and/or 17 from the 5’ end of the first nucleotide sequence is a 2’-fluoro nucleotide.
  • the nucleotide at position 5, 7, 8, and/or 9 from the 5’ end of the first nucleotide sequence is a 2’- fluoro nucleotide. In some embodiments, the nucleotide at position 5, 9, 10, 11, 12, and/or 19 from the 5’ end of the first nucleotide sequence is a 2’-fluoro nucleotide. In some embodiments, the 2’-fluoro nucleotide is a 2’-fluoro nucleotide mimic.
  • the 2’-fluoro or 2’-O-methyl nucleotide mimic is a nucleotide mimic of Formula (V): , wherein Rx is independently a nucleobase, aryl, heteroaryl, or H, Q 1 and Q 2 are independently S or O, R 5 is independently –OCD 3 , –F, or – OCH 3 , and R 6 and R 7 are independently H, D, or CD 3 .
  • the nucleobase is selected from thymine, cytosine, guanine, adenine, uracil, and an analogue or derivative thereof.
  • the 2’-fluoro or 2’-O-methyl nucleotide mimic is a nucleotide mimic of Formula (16) – Formula (20): wherein Rx is independently a nucleobase, aryl, heteroaryl, or H and R 2 is F or –OCH3.
  • the nucleobase is selected from thymine, cytosine, guanine, adenine, uracil, and an analogue or derivative thereof.
  • the sense strand may comprise 1, 2, 3, 4, or 5 or more modified nucleotide(s) selected from: wherein Rx is a nucleobase, aryl, heteroaryl, or H, (mun34), wherein Ry is a nucleobase, ( wherein * represent chiral center), and (wherein * represent chiral center), wherein B is a nucleobase, aryl, heteroaryl, or H, and wherein or represents a phosphodiester linkage, a phosphorothioate linkage, a mesyl phosphoramidate linkage, or H.
  • the nucleobase is selected from thymine, cytosine, guanine, adenine, uracil, and an analogue or derivative thereof.
  • the first nucleotide sequence comprises, consists of, or consists essentially of ribonucleic acids (RNAs).
  • the first nucleotide sequence comprises, consists of, or consists essentially of modified RNAs.
  • the modified RNAs are selected from a 2’-O-methyl RNA and 2’-fluoro RNA.
  • 15, 16, 17, 18, 19, 20, 21, 22, or 23 modified nucleotides of the first nucleotide sequence are independently selected from 2’-O-methyl RNA and 2’-fluoro RNA.
  • the sense strand may further comprise one or more internucleoside linkages independently selected from a phosphodiester (PO) internucleoside linkage, phosphorothioate (PS) internucleoside linkage, mesyl phosphoramidate internucleoside linkage (Ms), phosphorodithioate internucleoside linkage, and PS-mimic internucleoside linkage.
  • PO phosphodiester
  • PS phosphorothioate
  • Ms mesyl phosphoramidate internucleoside linkage
  • PS-mimic internucleoside linkage PS-mimic internucleoside linkage
  • the PS-mimic internucleoside linkage is a sulfo internucleoside linkage.
  • the sense strand may further comprise at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, or 15 or more phosphorothioate internucleoside linkages.
  • the sense strand comprises 20, 19, 18, 17, 16, 15, 14, 13, 12, 11, 10, 9, 8, 7, 6, 5, 4, or 3 or fewer phosphorothioate internucleoside linkages.
  • the sense strand comprises 2 to 10, 2 to 8, 2 to 6, 1 to 5, 1 to 4, 1 to 3, or 1 to 2 phosphorothioate internucleoside linkages.
  • the sense strand comprises 1 to 2 phosphorothioate internucleoside linkages. In some embodiments, the sense strand comprises 2 to 4 phosphorothioate internucleoside linkages. In some embodiments, at least one phosphorothioate internucleoside linkage is between the nucleotides at positions 1 and 2 from the 5’ end of the first nucleotide sequence. In some embodiments, at least one phosphorothioate internucleoside linkage is between the nucleotides at positions 2 and 3 from the 5’ end of the first nucleotide sequence.
  • the sense strand comprises two phosphorothioate internucleoside linkages between the nucleotides at positions 1 to 3 from the 5’ end of the first nucleotide sequence.
  • the sense strand may further comprise at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, or 15 or more mesyl phosphoramidate internucleoside linkages.
  • the sense strand comprises 20, 19, 18, 17, 16, 15, 14, 13, 12, 11, 10, 9, 8, 7, 6, 5, 4, or 3 or fewer mesyl phosphoramidate internucleoside linkages.
  • the sense strand comprises 2 to 10, 2 to 8, 2 to 6, 1 to 5, 1 to 4, 1 to 3, or 1 to 2 mesyl phosphoramidate internucleoside linkages. In some embodiments, the sense strand comprises 1 to 2 mesyl phosphoramidate internucleoside linkages. In some embodiments, the sense strand comprises 2 to 4 mesyl phosphoramidate internucleoside linkages. In some embodiments, the sense strand may comprise any of the modified nucleotides disclosed in the sub-section titled “Modified Nucleotides” below.
  • the sense strand may comprise a 5’-stabilized end cap, and the 5’-stabilized end cap may be selected from those disclosed in the sub-section titled “5’-Stabilized End Cap” below.
  • siNA antisense strand Any of the siNA molecules described herein may comprise an antisense strand.
  • the antisense strand may comprise a second nucleotide sequence.
  • the second nucleotide sequence may be 15 to 30, 15 to 25, 15 to 23, 17 to 23, 19 to 23, or 19 to 21 nucleotides in length. In some embodiments, the second nucleotide sequence is 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30 nucleotides in length.
  • the second nucleotide sequence is at least 19 nucleotides in length. In some embodiments, the second nucleotide sequence is at least 21 nucleotides in length. In some embodiments, the antisense strand is the same length as the second nucleotide sequence. In some embodiments, the antisense strand is longer than the second nucleotide sequence. In some embodiments, the antisense strand may further comprise 1, 2, 3, 4, or 5 or more nucleotides than the second nucleotide sequence. In some embodiments, the antisense strand is the same length as the sense strand. In some embodiments, the antisense strand is longer than the sense strand.
  • the antisense strand may further comprise 1, 2, 3, 4, or 5 or more nucleotides than the sense strand.
  • the antisense strand may further comprise a deoxyribonucleic acid (DNA).
  • the DNA is thymine (T).
  • the antisense strand may further comprise a TT sequence.
  • the antisense strand may further comprise one or more modified nucleotides that are adjacent to the second nucleotide sequence.
  • the one or more modified nucleotides are independently selected from any of the modified nucleotides disclosed herein (e.g., 2’-fluoro nucleotide, 2’- O-methyl nucleotide, 2’-fluoro nucleotide mimic, 2’-O-methyl nucleotide mimic, or a nucleotide comprising a modified nucleobase).
  • the second nucleotide sequence comprises 15, 16, 17, 18, 19, 20, 21, 22, 23, or more modified nucleotides independently selected from a 2’-O-methyl nucleotide and a 2’-fluoro nucleotide.
  • 70%, 75%, 80%, 85%, 90%, 95% or 100% of the nucleotides in the second nucleotide sequence are modified nucleotides independently selected from a 2’-O-methyl nucleotide and a 2’-fluoro nucleotide. In some embodiments, 100% of the nucleotides in the second nucleotide sequence are modified nucleotides independently selected from a 2’-O-methyl nucleotide and a 2’-fluoro nucleotide.
  • nucleotides of the second nucleotide sequence are 2’-O-methyl nucleotides. In some embodiments, between about 2 to 20 modified nucleotides of the second nucleotide sequence are 2’-O-methyl nucleotides.
  • between about 5 to 25 modified nucleotides of the second nucleotide sequence are 2’-O-methyl nucleotides. In some embodiments, between about 10 to 25 modified nucleotides of the second nucleotide sequence are 2’-O-methyl nucleotides. In some embodiments, between about 12 to 25 modified nucleotides of the second nucleotide sequence are 2’-O-methyl nucleotides. In some embodiments, at least 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, or 22 modified nucleotides of the second nucleotide sequence are 2’-O- methyl nucleotides.
  • At least about 12 modified nucleotides of the second nucleotide sequence are 2’-O-methyl nucleotides. In some embodiments, at least about 13 modified nucleotides of the second nucleotide sequence are 2’-O-methyl nucleotides. In some embodiments, at least about 14 modified nucleotides of the second nucleotide sequence are 2’-O-methyl nucleotides. In some embodiments, at least about 15 modified nucleotides of the second nucleotide sequence are 2’-O-methyl nucleotides. In some embodiments, at least about 16 modified nucleotides of the second nucleotide sequence are 2’-O-methyl nucleotides.
  • At least about 17 modified nucleotides of the second nucleotide sequence are 2’-O-methyl nucleotides. In some embodiments, at least about 18 modified nucleotides of the second nucleotide sequence are 2’-O-methyl nucleotides. In some embodiments, at least about 19 modified nucleotides of the second nucleotide sequence are 2’-O-methyl nucleotides. In some embodiments, less than or equal to 25, 24, 23, 22, 21, 20, 19, 18, 17, 16, 15, 14, 13, 12, 11, 10, 9, 8, 7, 6, 5, 4, 3, or 2 modified nucleotides of the second nucleotide sequence are 2’-O-methyl nucleotides.
  • less than or equal to 21 modified nucleotides of the second nucleotide sequence are 2’-O-methyl nucleotides. In some embodiments, less than or equal to 20 modified nucleotides of the second nucleotide sequence are 2’-O-methyl nucleotides. In some embodiments, less than or equal to 19 modified nucleotides of the second nucleotide sequence are 2’-O-methyl nucleotides. In some embodiments, less than or equal to 18 modified nucleotides of the second nucleotide sequence are 2’-O-methyl nucleotides.
  • less than or equal to 17 modified nucleotides of the second nucleotide sequence are 2’-O-methyl nucleotides. In some embodiments, less than or equal to 16 modified nucleotides of the second nucleotide sequence are 2’-O-methyl nucleotides. In some embodiments, less than or equal to 15 modified nucleotides of the second nucleotide sequence are 2’-O-methyl nucleotides. In some embodiments, less than or equal to 14 modified nucleotides of the second nucleotide sequence are 2’-O-methyl nucleotides.
  • modified nucleotides of the second nucleotide sequence are 2’-O-methyl nucleotides. In some embodiments, at least one modified nucleotide of the second nucleotide sequence is a 2’-O-methyl pyrimidine. In some embodiments, at least 5, 6, 7, 8, 9, or 10 modified nucleotides of the second nucleotide sequence are 2’-O-methyl pyrimidines. In some embodiments, at least one modified nucleotide of the second nucleotide sequence is a 2’-O-methyl purine.
  • At least 5, 6, 7, 8, 9, or 10 modified nucleotides of the second nucleotide sequence are 2’-O- methyl purines.
  • the 2’-O-methyl nucleotide is a 2’-O-methyl nucleotide mimic.
  • between 2 to 15 modified nucleotides of the second nucleotide sequence are 2’-fluoro nucleotides.
  • between 2 to 10 modified nucleotides of the second nucleotide sequence are 2’-fluoro nucleotides.
  • between 2 to 6 modified nucleotides of the second nucleotide sequence are 2’- fluoro nucleotides.
  • 1 to 6, 1 to 5, 1 to 4, or 1 to 3 modified nucleotides of the second nucleotide sequence are 2’-fluoro nucleotides. In some embodiments, at least 1, 2, 3, 4, 5, or 6 modified nucleotides of the second nucleotide sequence are 2’-fluoro nucleotides. In some embodiments, at least 1 modified nucleotide of the second nucleotide sequence is a 2’-fluoro nucleotide. In some embodiments, at least 2 modified nucleotides of the second nucleotide sequence are 2’-fluoro nucleotides.
  • At least 3 modified nucleotides of the second nucleotide sequence are 2’-fluoro nucleotides. In some embodiments, at least 4 modified nucleotides of the second nucleotide sequence are 2’-fluoro nucleotides. In some embodiments, at least 5 modified nucleotides of the second nucleotide sequence are 2’-fluoro nucleotides. In some embodiments, 10, 9, 8, 7, 6, 5, 4, 3 or fewer modified nucleotides of the second nucleotide sequence are 2’-fluoro nucleotides. In some embodiments, 10 or fewer modified nucleotides of the second nucleotide sequence are 2’- fluoro nucleotides.
  • 7 or fewer modified nucleotides of the second nucleotide sequence are 2’-fluoro nucleotides. In some embodiments, 6 or fewer modified nucleotides of the second nucleotide sequence are 2’-fluoro nucleotides. In some embodiments, 5 or fewer modified nucleotides of the second nucleotide sequence are 2’- fluoro nucleotides. In some embodiments, 4 or fewer modified nucleotides of the second nucleotide sequence are 2’-fluoro nucleotides. In some embodiments, 3 or fewer modified nucleotides of the second nucleotide sequence are 2’-fluoro nucleotides.
  • 2 or fewer modified nucleotides of the second nucleotide sequence are 2’- fluoro nucleotides.
  • at least one modified nucleotide of the second nucleotide sequence is a 2’-fluoro pyrimidine.
  • 1, 2, 3, 4, 5, or 6 modified nucleotides of the second nucleotide sequence are 2’-fluoro pyrimidines.
  • at least one modified nucleotide of the second nucleotide sequence is a 2’- fluoro purine.
  • 1, 2, 3, 4, 5, or 6 modified nucleotides of the second nucleotide sequence are 2’-fluoro purines.
  • the 2’-fluoro nucleotide is a 2’-fluoro nucleotide mimic. In some embodiments, the 2’-fluoro nucleotide or 2’-O-methyl nucleotide is a 2’- fluoro or 2’-O-methyl nucleotide mimic.
  • the 2’-fluoro or 2’-O-methyl nucleotide mimic is a nucleotide mimic of Formula (V): , wherein Rx is independently a nucleobase, aryl, heteroaryl, or H, Q 1 and Q 2 are independently S or O, R 5 is independently –OCD 3 , –F, or –OCH 3 , and R 6 and R 7 are independently H, D, or CD 3 .
  • the nucleobase is selected from thymine, cytosine, guanine, adenine, uracil, and an analogue or derivative thereof.
  • the 2’-fluoro or 2’-O-methyl nucleotide mimic is a nucleotide mimic of Formula (16) – Formula (20): wherein R x is a nucleobase, aryl, heteroaryl, or H and R 2 is independently F or -OCH 3 .
  • the nucleobase is selected from thymine, cytosine, guanine, adenine, uracil, and an analogue or derivative thereof.
  • the antisense strand may comprise 1, 2, 3, 4, or 5 or more modified nucleotide(s) selected from: , wherein Rx is a nucleobase, aryl,
  • the nucleobase is selected from thymine, cytosine, guanine, adenine, uracil, and an analogue or derivative thereof.
  • At least 1, 2, 3, 4, 5, 6, 7, 8, or 9 nucleotides at position 2, 5, 6, 8, 10, 14, 16, 17, and/or 18 from the 5’ end of the second nucleotide sequence is a 2’-fluoro nucleotide.
  • the nucleotide at position 2, 5, 6, 8, 10, 14, 16, 17, and/or 18 from the 5’ end of the second nucleotide sequence is a 2’-fluoro nucleotide.
  • at least two nucleotides at positions 2, 5, 6, 8, 10, 14, 16, 17, and/or 18 from the 5’ end of the second nucleotide sequence are 2’-fluoro nucleotides.
  • At least three nucleotides at positions 2, 5, 6, 8, 10, 14, 16, 17, and/or 18 from the 5’ end of the second nucleotide sequence are 2’-fluoro nucleotides. In some embodiments, at least four nucleotides at positions 2, 5, 6, 8, 10, 14, 16, 17, and/or 18 from the 5’ end of the second nucleotide sequence are 2’-fluoro nucleotides. In some embodiments, at least five nucleotides at positions 2, 5, 6, 8, 10, 14, 16, 17, and/or 18 from the 5’ end of the second nucleotide sequence are 2’-fluoro nucleotides.
  • the nucleotides at positions 2 and/or 14 from the 5’ end of the second nucleotide sequence are 2’-fluoro nucleotides. In some embodiments, the nucleotides at positions 2, 6, and/or 16 from the 5’ end of the second nucleotide sequence are 2’-fluoro nucleotides. In some embodiments, the nucleotides at positions 2, 6, 14, and/or 16 from the 5’ end of the second nucleotide sequence are 2’-fluoro nucleotides. In some embodiments, the nucleotides at positions 2, 6, 10, 14, and/or 18 from the 5’ end of the second nucleotide sequence are 2’-fluoro nucleotides.
  • the nucleotides at positions 2, 5, 8, 14, and/or 17 from the 5’ end of the second nucleotide sequence are 2’-fluoro nucleotides. In some embodiments, the nucleotide at position 2 from the 5’ end of the second nucleotide sequence is a 2’-fluoro nucleotide. In some embodiments, the nucleotide at position 5 from the 5’ end of the second nucleotide sequence is a 2’-fluoro nucleotide. In some embodiments, the nucleotide at position 6 from the 5’ end of the second nucleotide sequence is a 2’-fluoro nucleotide.
  • the nucleotide at position 8 from the 5’ end of the second nucleotide sequence is a 2’-fluoro nucleotide. In some embodiments, the nucleotide at position 10 from the 5’ end of the second nucleotide sequence is a 2’-fluoro nucleotide. In some embodiments, the nucleotide at position 14 from the 5’ end of the second nucleotide sequence is a 2’-fluoro nucleotide. In some embodiments, the nucleotide at position 16 from the 5’ end of the second nucleotide sequence is a 2’-fluoro nucleotide.
  • the nucleotide at position 17 from the 5’ end of the second nucleotide sequence is a 2’-fluoro nucleotide. In some embodiments, the nucleotide at position 18 from the 5’ end of the second nucleotide sequence is a 2’-fluoro nucleotide. In some embodiments, the 2’-fluoro nucleotide is a 2’-fluoro nucleotide mimic.
  • the nucleotides in the second nucleotide sequence are arranged in an alternating 1:3 modification pattern, wherein 1 nucleotide is a 2’-fluoro nucleotide and 3 nucleotides are 2’-O-methyl nucleotides, and wherein the alternating 1:3 modification pattern occurs at least 2 times. In some embodiments, the alternating 1:3 modification pattern occurs 2-5 times. In some embodiments, at least two of the alternating 1:3 modification pattern occur consecutively. In some embodiments, at least two of the alternating 1:3 modification pattern occurs nonconsecutively.
  • At least 1, 2, 3, 4, or 5 alternating 1:3 modification pattern begins at nucleotide position 2, 6, 10, 14, and/or 18 from the 5’ end of the antisense strand. In some embodiments, at least one alternating 1:3 modification pattern begins at nucleotide position 2 from the 5’ end of the antisense strand. In some embodiments, wherein at least one alternating 1:3 modification pattern begins at nucleotide position 6 from the 5’ end of the antisense strand. In some embodiments, at least one alternating 1:3 modification pattern begins at nucleotide position 10 from the 5’ end of the antisense strand.
  • At least one alternating 1:3 modification pattern begins at nucleotide position 14 from the 5’ end of the antisense strand. In some embodiments, at least one alternating 1:3 modification pattern begins at nucleotide position 18 from the 5’ end of the antisense strand. In some embodiments, the 2’-fluoro nucleotide is a 2’-fluoro nucleotide mimic.
  • the nucleotides in the second nucleotide sequence are arranged in an alternating 1:2 modification pattern, wherein 1 nucleotide is a 2’-fluoro nucleotide and 2 nucleotides are 2’-O-methyl nucleotides, and wherein the alternating 1:2 modification pattern occurs at least 2 times.
  • the alternating 1:2 modification pattern occurs 2-5 times.
  • at least two of the alternating 1:2 modification pattern occurs consecutively.
  • at least two of the alternating 1:2 modification pattern occurs nonconsecutively.
  • at least 1, 2, 3, 4, or 5 alternating 1:2 modification pattern begins at nucleotide position 2, 5, 8, 14, and/or 17 from the 5’ end of the antisense strand.
  • At least one alternating 1:2 modification pattern begins at nucleotide position 2 from the 5’ end of the antisense strand. In some embodiments, at least one alternating 1:2 modification pattern begins at nucleotide position 5 from the 5’ end of the antisense strand. In some embodiments, at least one alternating 1:2 modification pattern begins at nucleotide position 8 from the 5’ end of the antisense strand. In some embodiments, at least one alternating 1:2 modification pattern begins at nucleotide position 14 from the 5’ end of the antisense strand. In some embodiments, at least one alternating 1:2 modification pattern begins at nucleotide position 17 from the 5’ end of the antisense strand.
  • the 2’-fluoro nucleotide is a 2’-fluoro nucleotide mimic.
  • the second nucleotide sequence comprises, consists of, or consists essentially of ribonucleic acids (RNAs).
  • the second nucleotide sequence comprises, consists of, or consists essentially of modified RNAs.
  • the modified RNAs are selected from a 2’-O-methyl RNA and 2’-fluoro RNA.
  • 15, 16, 17, 18, 19, 20, 21, 22, or 23 modified nucleotides of the second nucleotide sequence are independently selected from 2’-O-methyl RNA and 2’-fluoro RNA.
  • the 2’-fluoro nucleotide is a 2’-fluoro nucleotide mimic.
  • the sense strand may further comprise one or more internucleoside linkages independently selected from a phosphodiester (PO) internucleoside linkage, phosphorothioate (PS) internucleoside linkage, phosphorodithioate internucleoside linkage, and PS-mimic internucleoside linkage.
  • the PS-mimic internucleoside linkage is a 3’- or 5’-sulfur linkage.
  • the antisense strand may further comprise at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, or 15 or more phosphorothioate internucleoside linkages. In some embodiments, the antisense strand comprises 20, 19, 18, 17, 16, 15, 14, 13, 12, 11, 10, 9, 8, 7, 6, 5, 4, or 3 or fewer phosphorothioate internucleoside linkages. In some embodiments, the antisense strand comprises 2 to 10, 2 to 8, 2 to 6, 1 to 5, 1 to 4, 1 to 3, or 1 to 2 phosphorothioate internucleoside linkages.
  • the antisense strand comprises 2 to 10, 2 to 8, 2 to 6, 1 to 5, 1 to 4, 1 to 3, or 1 to 2 phosphorothioate internucleoside linkages. In some embodiments, the antisense strand comprises 2 to 8 phosphorothioate internucleoside linkages. In some embodiments, the antisense strand comprises 3 to 8 phosphorothioate internucleoside linkages. In some embodiments, the antisense strand comprises 4 to 8 phosphorothioate internucleoside linkages. In some embodiments, at least one phosphorothioate internucleoside linkage is between the nucleotides at positions 1 and 2 from the 5’ end of the second nucleotide sequence.
  • At least one phosphorothioate internucleoside linkage is between the nucleotides at positions 2 and 3 from the 5’ end of the second nucleotide sequence. In some embodiments, at least one phosphorothioate internucleoside linkage is between the nucleotides at positions 1 and 2 from the 3’ end of the second nucleotide sequence. In some embodiments, at least one phosphorothioate internucleoside linkage is between the nucleotides at positions 2 and 3 from the 3’ end of the second nucleotide sequence.
  • the antisense strand comprises two phosphorothioate internucleoside linkages between the nucleotides at positions 1 to 3 from the 5’ end of the first nucleotide sequence. In some embodiments, the antisense strand comprises two phosphorothioate internucleoside linkages between the nucleotides at positions 1 to 3 from the 3’ end of the first nucleotide sequence.
  • the antisense strand comprises (a) two phosphorothioate internucleoside linkages between the nucleotides at positions 1 to 3 from the 5’ end of the first nucleotide sequence; and (b) two phosphorothioate internucleoside linkages between the nucleotides at positions 1 to 3 from the 3’ end of the first nucleotide sequence.
  • the antisense strand may further comprise at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, or 15 or more mesyl phosphoramidate internucleoside linkages.
  • the antisense strand comprises 20, 19, 18, 17, 16, 15, 14, 13, 12, 11, 10, 9, 8, 7, 6, 5, 4, or 3 or fewer mesyl phosphoramidate internucleoside linkages. In some embodiments, the antisense strand comprises 2 to 10, 2 to 8, 2 to 6, 1 to 5, 1 to 4, 1 to 3, or 1 to 2 mesyl phosphoramidate internucleoside linkages. In some embodiments, the antisense strand comprises 2 to 10, 2 to 8, 2 to 6, 1 to 5, 1 to 4, 1 to 3, or 1 to 2 mesyl phosphoramidate internucleoside linkages. In some embodiments, the antisense strand comprises 2 to 8 mesyl phosphoramidate internucleoside linkages.
  • the antisense strand comprises 3 to 8 mesyl phosphoramidate internucleoside linkages. In some embodiments, the antisense strand comprises 4 to 8 mesyl phosphoramidate internucleoside linkages. In some embodiments, at least one end of the ds-siNA is a blunt end. In some embodiments, at least one end of the ds-siNA comprises an overhang, wherein the overhang comprises at least one nucleotide. In some embodiments, both ends of the ds-siNA comprise an overhang, wherein the overhang comprises at least one nucleotide.
  • the overhang comprises 1 to 5 nucleotides, 1 to 4 nucleotides, 1 to 3 nucleotides, or 1 to 2 nucleotides. In some embodiments, the overhang consists of 1 to 2 nucleotides.
  • the antisense strand may comprise any of the modified nucleotides disclosed in the sub-section titled “Modified Nucleotides” below. In some embodiments, the antisense strand may comprise a 5’-stabilized end cap, and the 5’-stabilized end cap may be selected from those disclosed in the sub-section titled “5’-Stabilized End Cap” below. Modified Nucleotides
  • the siNA molecules disclosed herein comprise one or more modified nucleotides.
  • the sense strands disclosed herein comprise one or more modified nucleotides.
  • any of the first nucleotide sequences disclosed herein comprise one or more modified nucleotides.
  • the antisense strands disclosed herein comprise one or more modified nucleotides.
  • any of the second nucleotide sequences disclosed herein comprise one or more modified nucleotides.
  • the one or more modified nucleotides is adjacent to the first nucleotide sequence. In some embodiments, at least one modified nucleotide is adjacent to the 5’ end of the first nucleotide sequence.
  • At least one modified nucleotide is adjacent to the 3’ end of the first nucleotide sequence. In some embodiments, at least one modified nucleotide is adjacent to the 5’ end of the first nucleotide sequence and at least one modified nucleotide is adjacent to the 3’ end of the first nucleotide sequence. In some embodiments, the one or more modified nucleotides is adjacent to the second nucleotide sequence. In some embodiments, at least one modified nucleotide is adjacent to the 5’ end of the second nucleotide sequence. In some embodiments, at least one modified nucleotide is adjacent to the 3’ end of the second nucleotide sequence.
  • At least one modified nucleotide is adjacent to the 5’ end of the second nucleotide sequence and at least one modified nucleotide is adjacent to the 3’ end of the second nucleotide sequence.
  • a 2’-O-methyl nucleotide in any of sense strands or first nucleotide sequences disclosed herein is replaced with a modified nucleotide.
  • a 2’-O-methyl nucleotide in any of antisense strands or second nucleotide sequences disclosed herein is replaced with a modified nucleotide.
  • any of the siNA molecules, siNAs, sense strands, first nucleotide sequences, antisense strands, and second nucleotide sequences disclosed herein comprise 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30 or more modified nucleotides.
  • a modified nucleotide is selected from the group consisting of 2’-fluoro nucleotide, 2’-O-methyl nucleotide, 2’-fluoro nucleotide mimic, 2’-O-methyl nucleotide mimic, a locked nucleic acid, an unlocked nucleic acid, and a nucleotide comprising a modified nucleobase.
  • the unlocked nucleic acid is a 2’,3’-unlocked nucleic acid.
  • the unlocked nucleic acid is a 3’,4’- unlocked nucleic acid (e.g., mun34) in which the furanose ring lacks a bond between the 3’ and 4; carbons.
  • the siNA of the present disclosure may comprise one or more modified nucleotide(s) selected from: , wherein Rx is a nucleobase, aryl, heteroaryl, or H, (mun34), wherein R y is a nucleobase, chiral center), and (wherein * represent chiral center), wherein B is a nucleobase, aryl, heteroaryl, or H, and wherein or represents a phosphodiester linkage, a phosphorothioate linkage, a mesyl phosphoramidate linkage, or H.
  • both the sense strand and the antisense strand may each independently comprise 1, 2, 3, 4, or 5 or more of the foregoing modified nucleotides.
  • one or more (e.g., 1, 2, 3, 4, 5, or more) of the foregoing modified nucleotides may independently be present in the sense strand, first nucleotide sequence, antisense strand, and/or second nucleotide sequence of any of the siNA disclosed herein.
  • the nucleobase is selected from thymine, cytosine, guanine, adenine, uracil, and an analogue or derivative thereof.
  • any of the siNAs disclosed herein may comprise other modified nucleotides, such as 2’-fluoro or 2’-O-methyl nucleotide mimics.
  • the disclosed siNA may comprise at least 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 or more 2’-fluoro or 2’-O- methyl nucleotide mimics.
  • any of the sense strands disclosed herein comprise at least 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 or more 2’-fluoro or 2’-O-methyl nucleotide mimics.
  • any of the first nucleotide sequences disclosed herein comprise at least 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 or more 2’-fluoro or 2’-O-methyl nucleotide mimics.
  • any of the antisense strands disclosed herein comprise at least 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 or more 2’-fluoro or 2’-O-methyl nucleotide mimics.
  • any of the second nucleotide sequences disclosed herein comprise at least 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 or more 2’-fluoro or 2’-O-methyl nucleotide mimics.
  • the 2’-fluoro or 2’-O-methyl nucleotide mimic is a nucleotide mimic of Formula (16) – Formula (20): wherein Rx is a nucleobase, aryl, heteroaryl, or H and R 2 is independently F or -OCH3.
  • the nucleobase is selected from thymine, cytosine, guanine, adenine, uracil, and an analogue or derivative thereof.
  • the siNA molecules disclosed herein comprise at least one 2’- fluoro nucleotide, at least one 2’-O-methyl nucleotide, and at least one 2’-fluoro or 2’-O- methyl nucleotide mimic. In some embodiments, the at least one 2’-fluoro or 2’-O-methyl nucleotide mimic is adjacent to the first nucleotide sequence.
  • the at least one 2’-fluoro or 2’-O-methyl nucleotide mimic is adjacent to the 5’ end of first nucleotide sequence. In some embodiments, the at least one 2’-fluoro or 2’-O-methyl nucleotide mimic is adjacent to the 3’ end of first nucleotide sequence. In some embodiments, the at least one 2’-fluoro or 2’-O-methyl nucleotide mimic is adjacent to the second nucleotide sequence. In some embodiments, the at least one 2’-fluoro or 2’-O-methyl nucleotide mimic is adjacent to the 5’ end of second nucleotide sequence.
  • the at least one 2’-fluoro or 2’-O-methyl nucleotide mimic is adjacent to the 3’ end of second nucleotide sequence.
  • the first nucleotide sequence does not comprise a 2’-fluoro nucleotide mimic. In some embodiments, the first nucleotide sequence does not comprise a 2’-O-methyl nucleotide mimic.
  • the second nucleotide sequence does not comprise a 2’-fluoro nucleotide mimic. In some embodiments, the second nucleotide sequence does not comprise a 2’-O-methyl nucleotide mimic. Conjugated Moiety Further disclosed herein are siNA molecules comprising a conjugated moiety.
  • the conjugated moiety is selected from galactosamine, peptides, proteins, sterols, lipids, phospholipids, biotin, phenoxazines, active drug substance, cholesterols, phenanthridine, anthraquinone, acridine, fluoresceins, rhodamines, coumarins, and dyes.
  • the conjugated moiety is attached to the 3’ end of the sense strand or first nucleotide sequence. In some embodiments, the conjugated moiety is attached to the 3’ end of the sense strand or first nucleotide sequence via 1, 2, 3, 4, or 5 or more linkers.
  • the conjugated moiety is attached to the 5’ end of the sense strand or first nucleotide sequence. In some embodiments, the conjugated moiety is attached to the 5’ end of the sense strand or first nucleotide sequence via 1, 2, 3, 4, or 5 or more linkers. In some embodiments, the conjugated moiety is attached to the 3’ end of the antisense strand or second nucleotide sequence. In some embodiments, the conjugated moiety is attached to the 3’ end of the antisense strand or second nucleotide sequence via 1, 2, 3, 4, or 5 or more linkers. In some embodiments, the conjugated moiety is attached to the 5’ end of the antisense strand or second nucleotide sequence.
  • the conjugated moiety is attached to the 5’ end of the antisense strand or second nucleotide sequence via 1, 2, 3, 4, or 5 or more linkers.
  • the one or more linkers are independently selected from the group consisting of a phosphodiester linker, phosphorothioate linker, phosphorodithioate linker, and mesyl phosphoramidate linker.
  • the conjugated moiety is galactosamine.
  • any of the siNAs disclosed herein are attached to a conjugated moiety that is galactosamine.
  • the galactosamine is N-acetylgalactosamine (GalNAc).
  • any of the siNA molecules disclosed herein comprise GalNAc.
  • the first protecting group is acetyl.
  • the second protecting group is trimethoxytrityl (TMT).
  • the activated group is a phosphoramidite group.
  • the phosphoramidite group is a cyanoethoxy N,N- diisopropylphosphoramidite group.
  • the linker is a C 6 -NH 2 group.
  • A is a short interfering nucleic acid (siNA) or siNA molecule.
  • m is 3.
  • R is H, Z is H, and n is 1.
  • R is H, Z is H, and n is 2.
  • the GalNAc is Formula (VII): wherein R z is OH or SH; and each n is independently 1 or 2.
  • the targeting ligand may be a GalNAc targeting ligand may comprise 1, 2, 3, 4, 5 or 6 GalNAc units.
  • the GalNAc may be GalNAc amidite (i.e., compound 40-9, see Example 22), GalNAc 4 CPG GalNAc phophoramidite, or GalNAc4-ps-GalNAc4-ps- GalNAc4. These GalNAc moieties are shown below:
  • GalNAc3, GalNAc4, GalNAc5 and GalNAc6 may be conjugated to an siNA disclosed herein during synthesis with 12, or 3 moieties. Further GalNAc moieties, such as GalNAc1 and GalNAc2, can be used to form 5’ and 3’-GalNAc using post synthesis conjugation.
  • the galactosamine is attached to the 3’ end of the sense strand or first nucleotide sequence. In some embodiments, the galactosamine is attached to the 3’ end of the sense strand or first nucleotide sequence via 1, 2, 3, 4, or 5 or more linkers. In some embodiments, the galactosamine is attached to the 5’ end of the sense strand or first nucleotide sequence. In some embodiments, the galactosamine is attached to the 5’ end of the sense strand or first nucleotide sequence via 1, 2, 3, 4, or 5 or more linkers. In some embodiments, the galactosamine is attached to the 3’ end of the antisense strand or second nucleotide sequence.
  • the galactosamine is attached to the 3’ end of the antisense strand or second nucleotide sequence via 1, 2, 3, 4, or 5 or more linkers. In some embodiments, the galactosamine is attached to the 5’ end of the antisense strand or second nucleotide sequence. In some embodiments, the galactosamine is attached to the 5’ end of the antisense strand or second nucleotide sequence via 1, 2, 3, 4, or 5 or more linkers.
  • the one or more linkers are independently selected from the group consisting of a phosphodiester (p or po) linker, phosphorothioate (ps) linker, mesyl phosphoramidate linker (Ms), phosphoramidite (HEG) linker, triethylene glycol (TEG) linker, and/or phosphorodithioate linker.
  • the one or more linkers are independently selected from the group consisting of p-(PS)2, (PS)2-p-TEG-p, (PS)2-p-HEG-p, and (PS)2-p- (HEG-p)2.
  • the conjugated moiety is a lipid moiety.
  • any of the siNAs disclosed herein are attached to a conjugated moiety that is a lipid moiety.
  • lipid moieties include, but are not limited to, a cholesterol moiety, a thioether, e.g., hexyl-S-tritylthiol, a thiocholesterol, an aliphatic chain, e.g., dodecandiol or undecyl residues a phospholipid, e.g., di-hexadecyl-rac-glycerol or triethylammonium 1-di-O- hexadecyl-rac-glycero-S-H-phosphonate, a polyamine or a polyethylene glycol chain, adamantane acetic acid, a palmityl moiety, or an octadecylamine or hexylamino-carbonyl- oxycholesterol moiety.
  • the conjugated moiety is an active drug substance.
  • any of the siNAs disclosed herein are attached to a conjugated moiety that is an active drug substance.
  • active drug substances include, but are not limited to, aspirin, warfarin, phenylbutazone, ibuprofen, suprofen, fenbufen, ketoprofen, (5)-(+)- pranoprofen, carprofen, dansylsarcosine, 2,3,5-triiodobenzoic acid, flufenamic acid, folinic acid, a benzothiadiazide, chlorothiazide, a diazepine, indomethicin, a barbiturate, a cephalosporin, a sulfa drug, an antidiabetic, an antibacterial or an antibiotic.
  • any of the disclosed siNA molecules may comprise a phosphorylation blocker.
  • a 2’-O-methyl nucleotide in any of sense strands or first nucleotide sequences disclosed herein is replaced with a nucleotide containing a phosphorylation blocker.
  • a 2’-O-methyl nucleotide in any of antisense strands or second nucleotide sequences disclosed herein is replaced with a nucleotide containing a phosphorylation blocker.
  • a 2’-O-methyl nucleotide in any of sense strands or first nucleotide sequences disclosed herein is further modified to contain a phosphorylation blocker.
  • a 2’-O-methyl nucleotide in any of antisense strands or second nucleotide sequences disclosed herein is further modified to contain a phosphorylation blocker.
  • any of the siNA molecules disclosed herein comprise a phosphorylation blocker of Formula (IV): , wherein Ry 4 is a nucleobase, R is – O-R 30 or –NR 31 R 32 , R 30 is C 1 -C 8 substituted or unsubstituted alkyl; and R 31 and R 32 together with the nitrogen to which they are attached form a substituted or unsubstituted heterocyclic ring.
  • the nucleobase is selected from thymine, cytosine, guanine, adenine, uracil, and an analogue or derivative thereof.
  • any of the siNA molecules disclosed herein comprise a phosphorylation blocker of Formula (IV): Formula (IV), wherein R y is a nucleobase, and R 4 is –OCH3 or –N(CH2CH2)2O.
  • the nucleobase is selected from thymine, cytosine, guanine, adenine, uracil, and an analogue or derivative thereof.
  • a siNA molecule comprises (a) a phosphorylation blocker of Formula (IV): wherein Ry is a nucleobase, R 4 is –O-R 30 or –NR 31 R 32 , R 30 is C1-C8 substituted or unsubstituted alkyl; and R 31 and R 32 together with the nitrogen to which they are attached form a substituted or unsubstituted heterocyclic ring; and (b) a short interfering nucleic acid (siNA), wherein the phosphorylation blocker is conjugated to the siNA.
  • a phosphorylation blocker of Formula (IV) wherein Ry is a nucleobase, R 4 is –O-R 30 or –NR 31 R 32 , R 30 is C1-C8 substituted or unsubstituted alkyl; and R 31 and R 32 together with the nitrogen to which they are attached form a substituted or unsubstituted heterocyclic ring; and (b) a short interfering nucleic
  • a siNA molecule comprises (a) a phosphorylation blocker of Formula (IV): Formula (IV), wherein R y is a nucleobase, and R 4 is –OCH 3 or –N(CH2CH2)2O; and (b) a short interfering nucleic acid (siNA), wherein the phosphorylation blocker is conjugated to the siNA.
  • the phosphorylation blocker is attached to the 3’ end of the sense strand or first nucleotide sequence.
  • the phosphorylation blocker is attached to the 3’ end of the sense strand or first nucleotide sequence via 1, 2, 3, 4, or 5 or more linkers. In some embodiments, the phosphorylation blocker is attached to the 5’ end of the sense strand or first nucleotide sequence. In some embodiments, the phosphorylation blocker is attached to the 5’ end of the sense strand or first nucleotide sequence via 1, 2, 3, 4, or 5 or more linkers. In some embodiments, the phosphorylation blocker is attached to the 3’ end of the antisense strand or second nucleotide sequence.
  • the phosphorylation blocker is attached to the 3’ end of the antisense strand or second nucleotide sequence via 1, 2, 3, 4, or 5 or more linkers. In some embodiments, the phosphorylation blocker is attached to the 5’ end of the antisense strand or second nucleotide sequence. In some embodiments, the phosphorylation blocker is attached to the 5’ end of the antisense strand or second nucleotide sequence via 1, 2, 3, 4, or 5 or more linkers. In some embodiments, the one or more linkers are independently selected from the group consisting of a phosphodiester linker, phosphorothioate linker, mesyl phosphoramidate linker and phosphorodithioate linker.
  • siNA molecules comprising a 5’-stabilized end cap.
  • the terms “5’-stabilized end cap” and “5’ end cap” are used interchangeably.
  • a 2’-O-methyl nucleotide in any of sense strands or first nucleotide sequences disclosed herein is replaced with a nucleotide containing a 5’-stabilized end cap.
  • a 2’-O-methyl nucleotide in any of antisense strands or second nucleotide sequences disclosed herein is replaced with a nucleotide containing a 5’-stabilized end cap.
  • a 2’-O-methyl nucleotide in any of sense strands or first nucleotide sequences disclosed herein is further modified to contain a 5’-stabilized end cap.
  • a 2’-O-methyl nucleotide in any of antisense strands or second nucleotide sequences disclosed herein is further modified to contain a 5’-stabilized end cap.
  • the 5’-stabilized end cap is a 5’ phosphate mimic.
  • the 5’-stabilized end cap is a modified 5’ phosphate mimic.
  • the modified 5’ phosphate is a chemically modified 5’ phosphate.
  • the 5’-stabilized end cap is a 5’-vinyl phosphonate. In some embodiments, the 5’-vinyl phosphonate is a 5’-(E)-vinyl phosphonate or 5’-(Z)-vinyl phosphonate. In some embodiments, the 5’-vinyl phosphonate is a deuterated vinyl phosphonate. In some embodiments, the deuterated vinyl phosphonate is a mono-deuterated vinyl phosphonate. In some embodiments, the deuterated vinyl phosphonate is a di-deuterated vinyl phosphonate. In some embodiments, the 5’-stabilized end cap is a phosphate mimic.
  • the 5’-stabilized end cap is not a phosphate mimic, per se, but rather includes a novel modified base.
  • examples of such 5’-stabilized end caps include, but are not limited to (vm, wherein B is a nucleobase or derivative thereof), a a (vm(56amim)U, wherein R is H or Bz).
  • any of the siNA molecules disclosed herein may comprise a 5’ vinyl phosphonate moiety comprising a structure of: wherein B is a nucleobase, aryl, heteroaryl, or H; wherein represents a phosphodiester linkage, a phosphorothioate linkage, or a mesyl phosphoramidate linkage.
  • the disclosed siNA may comprise a nucleotide phosphate mimic selected from:
  • the nucleobase is selected from thymine, cytosine, guanine, adenine, uracil, and an analogue or derivative thereof.
  • the nucleobase is uracil.
  • the nucleobase is adenine.
  • the nucleobase is guanine.
  • the nucleobase is cytosine.
  • the nucleobase is thymine.
  • uridine can be a particularly suitable nucleobase.
  • one of these nucleotide phosphate mimics are located at the 5’ end of the antisense strand; however, these nucleotide phosphate mimics may also be incorporated at the 5’ end of the sense strand, the 3’ end of the antisense strand, or the 3’ end of the sense strand.
  • the siNA molecules disclosed herein may comprise in the sense strand, the antisense strand, or both a 5’-stabilized end cap of Formula (Ia): , wherein R is H, a nucleobas 26 x e, aryl, or heteroaryl; R is O alkenylene)-Z and R 20 is H; or R 26 and R 20 together form a 3- to 7-membered carbocyclic ring substituted with –(CR 21 R 22 ) n -Z or –(C 2 -C 6 alkenylene)-Z; n is 1, 2, 3, or 4; Z is –ONR 23 R 24 , – OP(O)OH(CH2)mCO2R 23 , –OP(S)OH(CH2)mCO2R 23 , –P(O)(OH)2, -P(O)(OH)(OCH3), - P(O)(OH)(OCD3), –SO2(CH2)mP(O)(OH
  • R 1 is an aryl.
  • the aryl is a phenyl ring.
  • R x is a nucleobase, aryl, heteroaryl, or H
  • R 1 is an aryl.
  • the aryl is a phenyl ring.
  • the siNA molecules disclosed herein may comprise a 5’- stabilized end cap selected from the group consisting of Formula (1) to Formula (16), Formula (9X) to Formula (12X), Formula (16X), Formula (9Y) to Formula (12Y), Formula (16Y), Formula (21) to Formula (36), Formula 36X, Formula (41) to (56), Formula (49X) to (52X), Formula (49Y) to (52Y), Formula 56X, Formula 56Y, Formula (61) and Formula (62):
  • any of the siNA molecules disclosed herein comprise a 5’- stabilized end cap selected from the group consisting of Formula (50), Formula (50X), Formula (50Y), Formula (56), Formula (56X), Formula (56Y), Formula (61), and Formula (62):
  • any of the siNA molecules disclosed herein comprise a 5’- stabilized end cap selected from the group consisting of Formula (71) to Formula (86), Formula (79X) to Formula (82X), Formula (79Y) to (82Y), Formula 86X, Formula 86X’, Formula 86Y, and Formula 86Y’: wherein R x is a nucleobase, aryl, heteroaryl, or H.
  • any of the siNA molecules disclosed herein comprise a 5’- stabilized end cap selected from the group consisting of Formula (78), Formula (79), Formula (79X), Formula (79Y), Formula (86), Formula (86X), and Formula (86X’): , wherein Rx is a nucleobase, aryl, heteroaryl, or H.
  • any of the siNA molecules disclosed herein comprise a 5’- stabilized end cap selected from the group consisting of Formulas (1A)-(15A), Formulas (1A- 1)-(7A-1), Formulas (1A-2)-(7A-2), Formulas (1A-3)-(7A-3), Formulas (1A-4)-(7A-4), Formulas (9B)-(12B), Formulas (9AX)-(12AX), Formulas (9AY)-(12AY), Formulas (9BX)- (12BX), and Formulas (9BY)-(12BY):
  • any of the siNA molecules disclosed herein comprise a 5’- stabilized end cap selected from the group consisting of Formulas (21A)-(35A), Formulas (29B)-(32B), Formulas (29AX)-(32AX), Formulas (29AY)-(32AY), Formulas (29BX)- (32BX), and Formulas (29BY)-(32BY):
  • any of the siNA molecules disclosed herein comprise a 5’- stabilized end cap selected from the group consisting of Formulas (71A)-(86A), Formulas (79XA)-(82XA), Formulas (79YA)-(82YA); Formula (86XA), Formula (86X’A), Formula (86Y), and Formula (86Y’):
  • any of the siNA molecules disclosed herein comprise a 5’- stabilized end cap selected from the group consisting of Formula (78A), Formula (79A), Formula (79XA), Formula (79YA), Formula (86A), Formula (86XA), and Formula (86X’A):
  • any of the siNA molecules disclosed herein may comprise a 5’stabilized end cap with the structure: wherein R y is a nucleobase. In some embodiments, any of the siNA molecules disclosed herein may comprise a 5’stabilized end cap with the structure: wherein R y is a nucleobase and R 15 is H or CH 3 . In some embodiments, the 5’-stabilized end cap is attached to the 5’ end of the antisense strand. In some embodiments, the 5’-stabilized end cap is attached to the 5’ end of the antisense strand via 1, 2, 3, 4, or 5 or more linkers.
  • the one or more linkers are independently selected from the group consisting of a phosphodiester (p or po) linker, phosphorothioate (ps) linker, mesyl phosphoramidate (Ms) linker, phosphoramidite (HEG) linker, triethylene glycol (TEG) linker, and/or phosphorodithioate linker.
  • the one or more linkers are independently selected from the group consisting of p-(PS)2, (PS)2-p-TEG-p, (PS)2-p-HEG-p, and (PS)2-p-(HEG-p)2.
  • compositions comprising any of the siNA molecules, sense strands, antisense strands, first nucleotide sequences, or second nucleotide sequences described herein.
  • the disclosed siNA and compositions thereof can be used in the treatment of various diseases and conditions associated with ANGPTL3 and/or ANGPTL8.
  • Internucleoside Linkages In some embodiments, any of the siNAs, sense strands, first nucleotide sequences, antisense strands, and/or second nucleotide sequences disclosed herein comprise 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23 or more internucleoside linkages.
  • 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 or more internucleoside linkages are independently selected from the group consisting of a phosphodiester (p or po), phosphorothioate (ps), mesyl phosphoramidate (Ms), or phosphorodithioate.
  • any of the siNAs, sense strands, first nucleotide sequences, antisense strands, and/or second nucleotide sequences disclosed herein further comprise 1, 2, 3, 4 or more linkages or other linkers that attach a conjugated moiety, phosphorylation blocker, and/or 5’ end cap to the siNA, sense strand, first nucleotide sequence, antisense strand, and/or second nucleotide sequences.
  • the 1, 2, 3, 4 or more linkers are independently selected from the group consisting of a phosphodiester (p or po) linker, phosphorothioate (ps) linker, mesyl phosphoramidate (Ms), phosphoramidite (HEG) linker, triethylene glycol (TEG) linker, and/or phosphorodithioate linker.
  • the one or more linkers are independently selected from the group consisting of p-(PS)2, (PS)2-p-TEG-p, (PS)2-p-HEG-p, and (PS)2-p-(HEG-p)2.
  • siNA Targeting ANGPTL3 or ANGPTL8 The tables below provide examples of siNA that target ANGPTL3 or ANGPTL8. Table 1 - ANGPTL8 siNA SS and AS Unmodified Sequences (5’ to 3’)
  • compositions comprising siNAs of the present disclosure.
  • a pharmaceutical composition comprising one or more siNA of the present disclosure, and a pharmaceutically acceptable diluent or carrier.
  • the disclosed siNA can be formulated in a pharmaceutical composition to individually target or inhibit or reduce expression of ANGPTL3 or ANGPTL8, or the disclosed siNA can be formulated in a pharmaceutical composition to simultaneously target or inhibit or reduce expression of both ANGPTL3 and ANGPTL8.
  • a pharmaceutical composition may comprise at least one siNA comprising a sense strand comprising any one of SEQ ID NOs: 1-316 and an antisense strand comprising any one of SEQ ID NOs: 317-632.
  • a pharmaceutical composition may comprise at least one siNA comprising a sense strand comprising any one of SEQ ID NOs: 633-878 and an antisense strand comprising any one of SEQ ID NOs: 879-1125.
  • a pharmaceutical composition may comprise at least one siNA comprising a sense strand comprising any one of SEQ ID NOs: 1-316 and an antisense strand comprising any one of SEQ ID NOs: 317-632 and at least one siNA comprising a sense strand comprising any one of SEQ ID NOs: 633-878 and an antisense strand comprising any one of SEQ ID NOs: 879-1125.
  • two separate pharmaceutical compositions may be administered to a subject concurrently or sequentially, wherein one pharmaceutical composition comprises at least one siNA comprising a sense strand comprising any one of SEQ ID NOs: 1-316 and an antisense strand comprising any one of SEQ ID NOs: 317-632 and the other pharmaceutical composition comprises at least one siNA comprising a sense strand comprising any one of SEQ ID NOs: 633-878 and an antisense strand comprising any one of SEQ ID NOs: 879-1125.
  • compositions comprising any of the siNA molecules, sense strands, antisense strands, first nucleotide sequences, or second nucleotide sequences described herein.
  • the compositions may comprise 1, 2, 3, or more siNA molecules described herein.
  • the composition comprises a double-stranded short interfering nucleic acid (ds-siNA) molecule comprises (a) a sense strand comprising a first nucleotide sequence, wherein the first nucleotide sequence is 15 to 30 nucleotides in length and comprises a nucleotide sequence as set forth in SEQ ID NOs: 1-316; and (b) an antisense strand comprising a second nucleotide sequence, wherein the second nucleotide sequence is 15 to 30 nucleotides in length and comprises a nucleotide sequence that is the reverse complement of the first nucleotide sequence.
  • ds-siNA double-stranded short interfering nucleic acid
  • the first nucleotide sequence comprises a nucleotide sequence as set forth in SEQ ID NOs:1-316; and (b) an antisense strand that is the reverse complement of the first nucleotide sequence, wherein the antisense strand comprises a second nucleotide sequence, wherein the second nucleotide sequence is 15 to 30 nucleotides in length and comprises a nucleotide sequence as set forth in SEQ ID NOs: 317-632.
  • the pharmaceutical compositions comprising any of the siNA molecules, sense strands, antisense strands, first nucleotide sequences, or second nucleotide sequences described herein.
  • compositions may comprise 1, 2, 3, or more siNA molecules described herein.
  • the composition comprises a double-stranded short interfering nucleic acid (ds-siNA) molecule comprises (a) a sense strand comprising a first nucleotide sequence, wherein the first nucleotide sequence is 15 to 30 nucleotides in length and comprises a nucleotide sequence as set forth in SEQ ID NOs: 633-878; and (b) an antisense strand comprising a second nucleotide sequence, wherein the second nucleotide sequence is 15 to 30 nucleotides in length and comprises a nucleotide sequence that is the reverse complement of the first nucleotide sequence.
  • ds-siNA double-stranded short interfering nucleic acid
  • the first nucleotide sequence comprises a nucleotide sequence as set forth in SEQ ID NOs: 633-878; and (b) an antisense strand that is the reverse complement of the first nucleotide sequence, wherein the antisense strand comprises a second nucleotide sequence, wherein the second nucleotide sequence is 15 to 30 nucleotides in length and comprises a nucleotide sequence as set forth in SEQ ID NOs: 879-1125.
  • the composition comprises a double-stranded short interfering nucleic acid (ds-siNA) molecule comprising (a) a sense strand comprising a nucleotide sequence identical to SEQ ID NOs:1-316, and (b) an antisense strand.
  • ds-siNA double-stranded short interfering nucleic acid
  • the present disclosure provides a double-stranded short interfering nucleic acid (ds-siNA) molecule comprising (a) an antisense strand comprising a nucleotide sequence identical to SEQ ID NOs: 317-632; and (b) a sense strand.
  • the composition comprises a double-stranded short interfering nucleic acid (ds-siNA) molecule comprising (a) a sense strand comprising a nucleotide sequence identical to SEQ ID NOs:633-878, and (b) an antisense strand.
  • ds-siNA double-stranded short interfering nucleic acid
  • the present disclosure provides a double-stranded short interfering nucleic acid (ds-siNA) molecule comprising (a) an antisense strand comprising a nucleotide sequence identical to SEQ ID NOs: 879-1125; and (b) a sense strand.
  • the composition comprises a double-stranded short interfering nucleic acid (ds-siNA) molecule comprising (a) a sense strand as set forth in is SEQ ID NOs: 633, 634, 642, 653, 654, 674, 687, 688, 690-694, 708-713, 756-757, 765, 776, 777, 797, 810, 811, 813-817, or 831-836, and (b) an antisense strand complementary to the sense strand as set forth in SEQ ID NOs: 879, 880, 888, 899, 900, 920, 933, 934, 936-940, 954-959, 1003- 1004, 1012, 1023, 1024, 1044, 1057, 1058, 1060-1064, or 1078-1083, respectively.
  • ds-siNA double-stranded short interfering nucleic acid
  • the composition comprises a double-stranded short interfering nucleic acid (ds- siNA) molecule comprising (a) a sense strand according to SEQ ID NOs: 644, 654, 688, 690, 708, 713, 756, 777, 811, 813, 831, or 836, and (b) an antisense strand complementary to the sense strand according to SEQ ID NOs: 879, 900, 934, 936, 954, 959, 1003, 1024, 1058, 1060, 1078, or 1083, respectively.
  • the pharmaceutical compositions may comprise (a) a phosphorylation blocker; and (b) an oligomer.
  • the phosphorylation blocker is any of the phosphorylation blockers disclosed herein.
  • the oligomer is any of the oligomers disclosed herein.
  • the oligomer comprises any of the sense strands, antisense strands, first nucleotide sequences, or second nucleotide sequences described herein.
  • the oligomer comprises any of the sense strands, antisense strands, first nucleotide sequences, or second nucleotide sequences described herein.
  • the oligomer comprises one or more modified nucleotides.
  • the one or more modified nucleotides are independently selected from a 2’-fluoro nucleotide and a 2’-O-methyl nucleotide.
  • the 2’-fluoro nucleotide or the 2’-O-methyl nucleotide is independently selected from any of the 2’-fluoro or 2’-O-methyl nucleotide mimics disclosed herein.
  • the oligomer comprises a nucleotide sequence comprising any of the modification patterns disclosed herein.
  • the pharmaceutical composition comprises (a) a 5’-stabilized end cap; and (b) an oligomer.
  • the 5’-stabilized end cap is any of the 5- stabilized end caps disclosed herein.
  • the oligomer is any of the oligomers disclosed herein.
  • the oligomer comprises any of the sense strands, antisense strands, first nucleotide sequences, or second nucleotide sequences described herein.
  • the oligomer comprises one or more modified nucleotides.
  • the one or more modified nucleotides are independently selected from a 2’-fluoro nucleotide and a 2’-O-methyl nucleotide.
  • the 2’-fluoro nucleotide or the 2’-O-methyl nucleotide is independently selected from any of the 2’-fluoro or 2’-O-methyl nucleotide mimics disclosed herein.
  • the oligomer comprises a nucleotide sequence comprising any of the modification patterns disclosed herein.
  • the pharmaceutical composition comprises (a) at least one phosphorylation blocker or 5’-stabilized end cap; and (b) an oligomer.
  • the phosphorylation blocker is any of the phosphorylation blockers disclosed herein.
  • the conjugated moiety is any of the galactosamines disclosed herein.
  • the 5’-stabilized end cap is any of the 5-stabilized end caps disclosed herein.
  • the oligomer is any of the oligomers disclosed herein.
  • the oligomer comprises any of the sense strands, antisense strands, first nucleotide sequences, or second nucleotide sequences described herein.
  • the oligomer comprises one or more modified nucleotides.
  • the one or more modified nucleotides are independently selected from a 2’- fluoro nucleotide and a 2’-O-methyl nucleotide.
  • the 2’-fluoro nucleotide or the 2’-O-methyl nucleotide is independently selected from any of the 2’-fluoro or 2’-O-methyl nucleotide mimics disclosed herein.
  • the oligomer comprises a nucleotide sequence comprising any of the modification patterns disclosed herein.
  • the pharmaceutical composition containing the siNA of the present disclosure is formulated for systemic administration via parenteral delivery.
  • Parenteral administration includes intravenous, intra-arterial, subcutaneous, intraperitoneal or intramuscular injection or infusion; also subdermal administration, e.g., via an implanted device.
  • the pharmaceutical composition containing the siNA of the present disclosure is formulated for subcutaneous (SC) or intravenous (IV) delivery.
  • Formulations for parenteral administration may include sterile aqueous solutions, which may also contain buffers, diluents and other pharmaceutically acceptable additives as understood by the skilled artisan.
  • the total concentration of solutes may be controlled to render the preparation isotonic.
  • the pharmaceutical composition comprises two or more siNAs, the siNAs may be present in varying amounts.
  • the weight ratio of first siNA to second siNA is 1:10 to 10:1, e.g., 1:10, 1:9, 1:8, 1:7, 1:6, 1:5, 1:4, 1:3, 1:2, 1:1, 2:1, 3:1, 4:1, 5:1, 6:1, 7:1, 8:1, 9:1, or 10:1.
  • the molar ratio of first siNA to second siNA is 1:10 to 10:1, e.g., 1:10 to 10:1, e.g., 1:10, 1:9, 1:8, 1:7, 1:6, 1:5, 1:4, 1:3, 1:2, 1:1, 2:1, 3:1, 4:1, 5:1, 6:1, 7:1, 8:1, 9:1, or 10:1.
  • the pharmaceutical composition comprises an amount of one or more of the siNA molecules described herein formulated with one or more pharmaceutically acceptable carriers (additives) and/or diluents.
  • the pharmaceutical compositions may be specially formulated for administration in solid or liquid form, including those adapted for the following: (1) parenteral administration, for example, by subcutaneous, intramuscular, intravenous or epidural injection as, for example, a sterile solution or suspension, or sustained-release formulation; (2) topical application, for example, as a cream, ointment, or a controlled-release patch or spray applied to the skin; (3) intravaginally or intrarectally, for example, as a pessary, cream or foam; (4) sublingually; (5) ocularly; (6) transdermally; or (7) nasally.
  • wetting agents such as sodium lauryl sulfate and magnesium stearate, as well as coloring agents, release agents, coating agents, sweetening, flavoring and perfuming agents, preservatives and antioxidants can also be present in the compositions.
  • antioxidants examples include: (1) water soluble antioxidants, such as ascorbic acid, cysteine hydrochloride, sodium bisulfate, sodium metabisulfite, sodium sulfite and the like; (2) oil-soluble antioxidants, such as ascorbyl palmitate, butylated hydroxyanisole (BHA), butylated hydroxytoluene (BHT), lecithin, propyl gallate, alpha-tocopherol, and the like; and (3) metal chelating agents, such as citric acid, ethylenediamine tetraacetic acid (EDTA), sorbitol, tartaric acid, phosphoric acid, and the like.
  • water soluble antioxidants such as ascorbic acid, cysteine hydrochloride, sodium bisulfate, sodium metabisulfite, sodium sulfite and the like
  • oil-soluble antioxidants such as ascorbyl palmitate, butylated hydroxyanisole (BHA), butylated hydroxytoluene (BHT), le
  • Formulations of the present disclosure include those suitable for nasal, topical (including buccal and sublingual), rectal, vaginal and/or parenteral administration.
  • the formulations may conveniently be presented in unit dosage form and may be prepared by any methods well known in the art of pharmacy.
  • the amount of active ingredient which can be combined with a carrier material to produce a single dosage form will vary depending upon the host being treated, the particular mode of administration.
  • the amount of active ingredient which can be combined with a carrier material to produce a single dosage form will generally be that amount of the compound (e.g., siNA molecule) which produces a therapeutic effect.
  • a formulation of the present disclosure comprises an excipient selected from the group consisting of cyclodextrins, celluloses, liposomes, micelle forming agents, e.g., bile acids, and polymeric carriers, e.g., polyesters and polyanhydrides; and a compound (e.g., siNA molecule) of the present disclosure.
  • Methods of preparing these formulations or compositions include the step of bringing into association a compound (e.g., siNA molecule) of the present disclosure with the carrier and, optionally, one or more accessory ingredients.
  • a compound e.g., siNA molecule
  • the formulations are prepared by uniformly and intimately bringing into association a compound (e.g., siNA molecule) of the present disclosure with liquid carriers, or finely divided solid carriers, or both, and then, if necessary, shaping the product.
  • Formulations of the disclosure suitable for a suspension in an aqueous or non-aqueous liquid, or as an oil-in-water or water-in-oil liquid emulsion, or as an elixir or syrup, each containing a predetermined amount of a compound (e.g., siNA molecule) of the present disclosure as an active ingredient.
  • a compound (e.g., siNA molecule) of the present disclosure may also be administered as a bolus, electuary, or paste.
  • the active ingredient may be mixed with one or more pharmaceutically-acceptable carriers, such as sodium citrate or dicalcium phosphate, and/or any of the following: (1) fillers or extenders, such as starches, lactose, sucrose, glucose, mannitol, and/or silicic acid; (2) binders, such as, for example, carboxymethylcellulose, alginates, gelatin, polyvinyl pyrrolidone, sucrose and/or acacia; (3) humectants, such as glycerol; (4) disintegrating agents, such as agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates, and sodium carbonate; (5) solution retarding agents, such as paraffin; (6) absorption accelerators, such as quaternary ammonium compounds and surfactants, such as poloxamer and sodium lauryl sulfate; (7) wetting agents, such as, for example, cetyl alcohol, g
  • the disclosed dosage forms may be sterilized by, for example, filtration through a bacteria-retaining filter, or by incorporating sterilizing agents in the form of sterile solid compositions which can be dissolved in sterile water, or some other sterile injectable medium immediately before use. These compositions may also optionally contain opacifying agents and may be of a composition that they release the active ingredient(s) only, or preferentially, in a certain portion of the gastrointestinal tract, optionally, in a delayed manner. Examples of embedding compositions which can be used include polymeric substances and waxes. The active ingredient can also be in micro-encapsulated form, if appropriate, with one or more of the above-described excipients.
  • Liquid dosage forms of the compounds (e.g., siNA molecules) of the disclosure include pharmaceutically acceptable emulsions, microemulsions, solutions, suspensions, syrups and elixirs.
  • the liquid dosage forms may contain inert diluents commonly used in the art, such as, for example, water or other solvents, solubilizing agents and emulsifiers, such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1,3-butylene glycol, oils (I particular, cottonseed, groundnut, corn, germ, olive, castor and sesame oils), glycerol, tetrahydrofuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, and mixtures thereof.
  • inert diluents commonly used in the art, such as, for example, water or other solvents
  • compositions can also include adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, coloring, perfuming and preservative agents.
  • Suspensions in addition to the active compounds (e.g., siNA molecules), may contain suspending agents as, for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum metahydroxide, bentonite, agar-agar and tragacanth, and mixtures thereof.
  • Formulations of the pharmaceutical compositions of the disclosure for rectal or vaginal administration may be presented as a suppository, which may be prepared by mixing one or more compounds (e.g., siNA molecules) of the disclosure with one or more suitable nonirritating excipients or carriers comprising, for example, cocoa butter, polyethylene glycol, a suppository wax or a salicylate, and which is solid at room temperature, but liquid at body temperature and, therefore, will melt in the rectum or vaginal cavity and release the active compound (e.g., siNA molecule).
  • a suppository which may be prepared by mixing one or more compounds (e.g., siNA molecules) of the disclosure with one or more suitable nonirritating excipients or carriers comprising, for example, cocoa butter, polyethylene glycol, a suppository wax or a salicylate, and which is solid at room temperature, but liquid at body temperature and, therefore, will melt in the rectum or vaginal cavity and release the active compound (e.
  • Formulations of the present disclosure which are suitable for vaginal administration also include pessaries, tampons, creams, gels, pastes, foams or spray formulations containing such carriers as are known in the art to be appropriate.
  • Dosage forms for the topical or transdermal administration of a compound (e.g., siNA molecule) of this disclosure include powders, sprays, ointments, pastes, creams, lotions, gels, solutions, patches and inhalants.
  • the active compound e.g., siNA molecule
  • the ointments, pastes, creams and gels may contain, in addition to an active compound (e.g., siNA molecule) of this disclosure, excipients, such as animal and vegetable fats, oils, waxes, paraffins, starch, tragacanth, cellulose derivatives, polyethylene glycols, silicones, bentonites, silicic acid, talc and zinc oxide, or mixtures thereof.
  • Powders and sprays can contain, in addition to a compound (e.g., siNA molecule) of this disclosure, excipients such as lactose, talc, silicic acid, aluminum hydroxide, calcium silicates and polyamide powder, or mixtures of these substances.
  • Sprays can additionally contain customary propellants, such as chlorofluorohydrocarbons and volatile unsubstituted hydrocarbons, such as butane and propane.
  • Transdermal patches have the added advantage of providing controlled delivery of a compound (e.g., siNA molecule) of the present disclosure to the body.
  • dosage forms can be made by dissolving or dispersing the compound (e.g., siNA molecule) in the proper medium.
  • Absorption enhancers can also be used to increase the flux of the compound (e.g., siNA molecule) across the skin. The rate of such flux can be controlled by either providing a rate controlling membrane or dispersing the compound (e.g., siNA molecule) in a polymer matrix or gel.
  • compositions of this disclosure suitable for parenteral administration comprise one or more compounds (e.g., siNA molecules) of the disclosure in combination with one or more pharmaceutically-acceptable sterile isotonic aqueous or nonaqueous solutions, dispersions, suspensions or emulsions, or sterile powders which may be reconstituted into sterile injectable solutions or dispersions just prior to use, which may contain sugars, alcohols, antioxidants, buffers, bacteriostats, solutes which render the formulation isotonic with the blood of the intended recipient or suspending or thickening agents.
  • compounds e.g., siNA molecules
  • aqueous and nonaqueous carriers examples include water, ethanol, polyols (such as glycerol, propylene glycol, polyethylene glycol, and the like), and suitable mixtures thereof, vegetable oils, such as olive oil, and injectable organic esters, such as ethyl oleate.
  • polyols such as glycerol, propylene glycol, polyethylene glycol, and the like
  • vegetable oils such as olive oil
  • injectable organic esters such as ethyl oleate.
  • Proper fluidity can be maintained, for example, by the use of coating materials, such as lecithin, by the maintenance of the required particle size in the case of dispersions, and by the use of surfactants.
  • These compositions may also contain adjuvants such as preservatives, wetting agents, emulsifying agents and dispersing agents.
  • Prevention of the action of microorganisms upon the subject compounds may be ensured by the inclusion of various antibacterial and antifungal agents, for example, paraben, chlorobutanol, phenol sorbic acid, and the like. It may also be desirable to include isotonic agents, such as sugars, sodium chloride, and the like into the compositions. In addition, prolonged absorption of the injectable pharmaceutical form may be brought about by the inclusion of agents which delay absorption such as aluminum monostearate and gelatin. In some cases, in order to prolong the effect of a drug, it is desirable to slow the absorption of the drug from subcutaneous or intramuscular injection. This may be accomplished by the use of a liquid suspension of crystalline or amorphous material having poor water solubility.
  • the rate of absorption of the drug then depends upon its rate of dissolution which, in turn, may depend upon crystal size and crystalline form.
  • delayed absorption of a parenterally-administered drug form is accomplished by dissolving or suspending the drug in an oil vehicle.
  • injectable depot forms are made by forming microencapsule matrices of the subject compounds (e.g., siNA molecules) in biodegradable polymers such as polylactide- polyglycolide. Depending on the ratio of drug to polymer, and the nature of the particular polymer employed, the rate of drug release can be controlled. Examples of other biodegradable polymers include poly(orthoesters) and poly(anhydrides).
  • the present disclosure provides a pharmaceutical composition comprising the siNAs disclosed herein conjugated to a ligand targeting receptors expressed in hepatocytes to treat Cardiovascular Diseases (CVD) and MASH
  • the ligands that could be conjugated to the siNAs include but are not limited to GalNAc, Cholesterol, Palmitic Acid etc.
  • the pharmaceutical composition comprises a liposome that encapsulates the siNAs disclosed herein to form a lipid nanoparticle (LNP) targeting hepatocellular carcinoma (HCC) or other secondary cancers metastasized to the liver.
  • LNP lipid nanoparticle
  • HCC hepatocellular carcinoma
  • the present disclosure provides a pharmaceutical composition
  • a pharmaceutical composition comprising the siNAs disclosed herein conjugated to a ligand targeting receptors expressed in HCC.
  • the ligands that could be conjugated to the siNAs include but are not limited to GalNAc, Folate, Cholesterol, Palmitic Acid etc.
  • the compounds (e.g., siNA molecules) of the present disclosure are administered as pharmaceuticals, to humans and animals, they can be given per se or as a pharmaceutical composition containing, for example, 0.1 to 99% (more preferably, 10 to 30%) of active ingredient in combination with a pharmaceutically acceptable carrier.
  • the pharmaceutical acceptable carriers is a liposome that encapsulates the ds- siNA molecules.
  • siNA molecules of the present disclosure may be used to treat or prevent a disease in a subject in need thereof.
  • a method of treating or preventing a disease in a subject in need thereof comprises administering to the subject any of the siNA molecules disclosed herein.
  • a method of treating or preventing a disease in a subject in need thereof comprises administering to the subject any of the compositions disclosed herein.
  • the subject is a mammal.
  • the subject is a human.
  • the subject is a non-human primate.
  • the subject is a cat.
  • the subject is a camel.
  • the subject may be at least 40 years old, at least 45 years old, at least 50 years old, at least 55 years old, at least 60 years old, at least 65 years old, at least 70 years old, at least 75 years old, or at least 80 years old or older.
  • the subject is a pediatric subject (i.e., less than 18 years old).
  • the preparations (e.g., siNA molecules or pharmaceutical compositions thereof) of the present disclosure may be given parenterally, topically, or rectally or administered in the form of an inhalant. They are, of course, given in forms suitable for each administration route.
  • the siNA molecules are given in the form of nanoparticle where a liposome encapsulates the siNA molecules.
  • these compounds may be administered to humans and other animals for therapy or as a prophylactic by any suitable route of administration, including nasally (as by, for example, a spray), rectally, intravaginally, parenterally, intracisternally and topically, as by powders, ointments or drops, including buccally and sublingually.
  • the compounds or compositions are inhaled, as by, for example, an inhaler, a nebulizer, or in an aerosolized form.
  • the compounds (e.g., siNA molecules) of the present disclosure which may be used in a suitable hydrated form, and/or the pharmaceutical compositions of the present disclosure, are formulated into pharmaceutically-acceptable dosage forms by conventional methods known to those of skill in the art.
  • the present disclosure provides methods of treating or preventing a disease in a subject in need thereof, comprising administering the subject the ds- siNA molecules disclosed herein to reduce ANGPTL8 and/or ANGPTL3 activity or level.
  • the method of treating or preventing the disease comprises administering the subject the pharmaceutical composition comprising the ds-siNA molecules disclosed herein to reduce ANGPTL8 and/or ANGPTL3 activity or level.
  • the disease is a ANGPTL8 and/or ANGPTL3 associated disease or disorder.
  • the ANGPTL8 and/or ANGPTL3 associated disease or disorder comprises cardiovascular disease or nonalcoholic fatty liver disease (NAFLD), such as metabolic dysfunction-associated steatohepatitis (MASH).
  • the cardiovascular disease is hypertriglyceridemia (HTG) or familial hypercholesterolemia (FH).
  • compositions of this disclosure may be varied so as to obtain an amount of the active ingredient which is effective to achieve the desired therapeutic response for a particular patient, composition, and mode of administration, without being toxic to the patient.
  • active ingredients e.g., siNA molecules
  • the selected dosage level will depend upon a variety of factors including the activity of the particular compound (e.g., siNA molecule) of the present disclosure employed, or the ester, salt or amide thereof, the route of administration, the time of administration, the rate of excretion or metabolism of the particular compound being employed, the rate and extent of absorption, the duration of the treatment, other drugs, compounds and/or materials used in combination with the particular compound employed, the age, sex, weight, condition, general health and prior medical history of the patient being treated, and like factors well known in the medical arts.
  • a physician or veterinarian having ordinary skill in the art can readily determine and prescribe the effective amount of the pharmaceutical composition required.
  • a suitable daily dose of a compound (e.g., siNA molecule) of the disclosure is the amount of the compound that is the lowest dose effective to produce a therapeutic effect.
  • Such an effective dose generally depends upon the factors described above.
  • the compounds are administered at about 0.01 mg/kg to about 200 mg/kg, more preferably at about 0.1 mg/kg to about 100 mg/kg, even more preferably at about 0.5 mg/kg to about 50 mg/kg.
  • the compound is administered at a dose equal to or greater than 0.01, 0.02, 0.03, 0.04, 0.05, 0.06, 0.07, 0.08, 0.09, 0.10, 0.11, 0.12, 0.13, 0.14, 0.15, 0.16, 0.17, 0.18, 0.19, 0.20, 0.21, 0.22, 0.23, 0.24, 0.25, 0.26, 0.27, 0.28, 0.29, 0.30, 0.35, 0.40, 0.45, 0.50, 0.55, 0.60, 0.65, 0.7, 0.75, 0.8, 0.85, 0.9, 0.95, or 1 mg/kg.
  • the compound is administered at a dose equal to or less than 200, 190, 180, 170, 160, 150, 140, 130, 120, 110, 100, 95, 90, 85, 80, 75, 70, 65, 60, 55, 50, 45, 40, 35, 30, 25, 20, or 15 mg/kg.
  • the total daily dose of the compound is equal to or greater than 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 105, 110, 115, 120, 125, 130, 135, 140, 145, 150, 155, 160, 165, 170, 175, 180, 185, 190, 195, or 100 mg.
  • the effective amount may be less than when the compound is used alone.
  • the effective daily dose of the active compound e.g., siNA molecule
  • the effective daily dose of the active compound may be administered as two, three, four, five, six or more sub-doses administered separately at appropriate intervals throughout the day, optionally, in unit dosage forms. Preferred dosing is one administration per day.
  • the compound is administered at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, or 21 times a week.
  • the compound is administered at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, or 21 times a month.
  • the compound is administered once every 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, or 21 days. In some embodiments, the compound is administered once every 1, 2, 3, 4, 5, 6, 7, or 8 weeks.
  • the disclosed siNAs and pharmaceutical compositions comprising the disclosed siNAs are combined with approved or late stage development drugs in cardiovascular disease.
  • Non-limiting examples include statins including atorvastatin, fluvastatin, lovastatin, pitavastatin, pravastatin, rosuvastatin and simvastatin; PCSK9 inhibitors including alirocumab, evolocumab, and inclisiran; as well as cholesterol uptake inhibitor Ezetimibe.
  • the disclosed siNAs and pharmaceutical compositions comprising the disclosed siNAs are combined with approved or late stage development drugs in nonalcoholic fatty liver disease.
  • approved or late stage development drugs include Resmetirom, ALG-055009, VK2809, efruxifermin, Saroglitazar, Obeticholic acid, Cenicriviroc, Aramchol, Dapagliflozin, Semaglutide, Belapectin, MSDC0602K, Lanifibranor and Tesamorelin.
  • the pharmaceutical composition comprises at least a first siNA that reduces ANGPTL3 expression and a second siNA that reduces ANGPTL8 expression as disclosed herein and a pharmaceutically acceptable carrier.
  • the a weight ratio of the first siNA and the second siNA is selected from 1:10, 1:9, 1:8, 1:7, 1:6, 1:5, 1:4, 1:3, 1:2, 1:1, 2:1, 3:1, 4:1, 5:1, 6:1, 7:1, 8:1, 9:1, or 10:1.
  • a molar ration of the first siNA and the second ds-siNA is selected from 1:10, 1:9, 1:8, 1:7, 1:6, 1:5, 1:4, 1:3, 1:2, 1:1, 2:1, 3:1, 4:1, 5:1, 6:1, 7:1, 8:1, 9:1, or 10:1.
  • the first siNA and the second siNA are physically linked by a linker.
  • Non-limiting examples of linkers are nucleotide based linkers or non-nucleotide based linkers.
  • the nucleotide based linker is between about 1 to about 15 nucleotides in length.
  • the nucleotide based linker is 1 or more, 2 or more, 3 or more, 4 or more, 5 or more, 6 or more, 7 or more, 8 or more, 9 or more, 10 or more, 11 or more, 12 or more, 13 or more, 14 or more, or 15 or more nucleotides in length.
  • the linker is at least one or more one N-acetylgalactosamine (e.g., a GalNAc) as disclosed herein.
  • the siNAs of the disclosure optionally can be used in combination with modulators of other genes and/or gene products associated with the maintenance or development of diseases or disorders associated with aberrant ANGPTL8 and/or ANGPTL3 expression.
  • EXAMPLES These examples are provided for illustrative purposes only and not to limit the scope of the claims provided herein.
  • the dsNA identifiers refer to the modified sequences provided in Tables 2 and 4.
  • Comparative control siNAs for the examples below have the sequences set forth in Table 5.
  • Example 1 Evaluation of ANGPTLA3 siNAs using a three-point luciferase reporter assay
  • Renilla luciferase was used as the primary reporter gene with the ANGPTL3 gene (NM_014495.4) cloned downstream of its translational stop codon.
  • a second reporter gene, firefly luciferase was expressed and used as a negative transfection control.
  • the plasmid was first transfected using Lipofectamine 3000 (Invitrogen, L3000001) into COS-7 cells (ATCC, CRL-1651) seeded into 96-well microplates.
  • Example 2 Evaluation of ANGPTL3 siNAs using dose-response assays Select candidates were defined in Example 1 as showing ⁇ 65% inhibition at 1 nM and ⁇ 85% inhibition at 10 nM. Dose-response assays were conducted, with serial concentrations of siNAs starting at 10 nM (1:5 dilutions) for each select candidate siNA using the Dual- Glo® Luciferase Assay System for ANGPTL3 as described in Example 1. Dose-response curves were fitted by nonlinear regression with variable slope, and EC 50 values and maximum percentage inhibition were calculated (Table 7).
  • the Huh-7 cells were processed with the TaqMan Fast Advanced Cells-to-Ct Kit (Invitrogen, A35378), according to the manufacturer’s protocol.
  • the cell lysates were used for reverse transcription.
  • Gene expression was measured using TaqMan Fast Advanced Master Mix (Applied Biosystems, 4444964) and the following TaqMan Gene Expression assays (Applied Biosystems, 4331182): ACTB (Hs01060665_g1) and ANGPTL3 (Hs00205581_m1).
  • ACTB served as the endogenous control gene.
  • RT-qPCR reactions were run on the QuantStudioTM 6 Pro Real- Time PCR System (Applied Biosystems).
  • hepatoma-derived Hep3B cells (ATCC, HB-8064) were transfected with serially diluted siNA starting at 10 nM (1:8 dilutions) and Opti-MEMTM using Lipofectamine RNAiMAX (Invitrogen, 13778100).
  • a negative mock transfection control which consisted of transfecting 1 ⁇ phosphate-buffered saline, was included.
  • the Hep3B cells were processed with the TaqMan Fast Advanced Cells-to-Ct Kit (Invitrogen, A35378), according to the manufacturer’s protocol. The cell lysates were used for reverse transcription.
  • the human ANGPTL3 siNAs dsNA-240, dsNA-241, dsNA-242, dsNA-243, dsNA- 244, dsNA-245, dsNA-250, dsNA-251, dsNA-252, dsNA-253, dsNA-254, dsNA-255, dsNA- 260, dsNA-262, dsNA-263, dsNA-264, dsNA-265, dsNA-266, dsNA-267, dsNA-268, dsNA- 269, dsNA-274, dsNA-275, dsNA-276, dsNA-280 and dsNA-281 showed better in vitro potency than the comparative control siNA, while dsNA-257, dsNA-258, dsNA-270, dsNA- 271, dsNA-272, dsNA-273, dsNA-277, dsNA
  • Example 3 Toxicity assay and cross-species conservation for ANGPTL3 siNAs The candidates were further narrowed to six select siNAs, dsNA-159, dsNA-180, dsNA-214, dsNA-216, dsNA-234, and dsNA-239.
  • CellTiter-Glo ® Luminescent Cell Viability Assays were performed in siNA-treated COS-7 cells under conditions as described in Example 1. Assays were performed according to the manufacturer’s protocol and luminescence was measured on an EnVision plate reader. The luminescence from siNA- treated wells were then normalized to luminescence of mock-treated wells and percentage viability was calculated. (FIG. 2A).
  • hepatoma-derived Hep3B cells (ATCC, HB-8064) were transfected with siNA at 10.0, 1.0, or 0.1 nM and Opti-MEMTM using Lipofectamine RNAiMAX (Invitrogen, 13778100).
  • a negative mock transfection control which consisted of transfecting 1 ⁇ phosphate-buffered saline, was included.
  • the Hep3B cells were processed with the TaqMan Fast Advanced Cells-to-Ct Kit (Invitrogen, A35378), according to the manufacturer’s protocol. The cell lysates were used for reverse transcription.
  • Example 5 Evaluation of ANGPTL8 siNAs using a dose-response RT-qPCT assay.
  • hepatoma-derived Hep3B cells ATCC, HB-8064
  • Opti-MEMTM Lipofectamine RNAiMAX (Invitrogen, 13778100).
  • a negative mock transfection control which consisted of transfecting 1 ⁇ phosphate-buffered saline, was included.
  • the Hep3B cells were processed with the TaqMan Fast Advanced Cells-to-Ct Kit (Invitrogen, A35378), according to the manufacturer’s protocol.
  • the cell lysates were used for reverse transcription.
  • Gene expression was measured using TaqMan Fast Advanced Master Mix (Applied Biosystems, 4444964) and the following TaqMan Gene Expression assays (Applied Biosystems, 4331182): ACTB (Hs01060665_g1) and ANGPTL8 (Hs00218820_m1).
  • ACTB served as the endogenous control gene.
  • RT-qPCR reactions were run on the QuantStudioTM 6 Pro Real-Time PCR System (Applied Biosystems).
  • the RQ of gene expression was calculated via the 2 ⁇ Ct (RQ) method. Results are presented as expression relative to the expression levels of negative mock control samples. Dose-response curves were fitted by nonlinear regression with variable slope, EC50 values, and maximum percentage inhibition calculated (Table 11). Most of the tested siNAs exhibited similar or enhanced potency and maximum percentage of ANGPTL8 RNA inhibition as the comparative control siNA dsNA-284. Table 11 – Results of Dose-Response RT-qPCR Assay for ANGPTL8 in Hep3B Cells Example 6. AAV-hANGPTL3 Mouse Study Design To evaluate certain ANGPTL3 siNA agents, an AAV-ANGPTL3 mouse model was used.
  • mice were injected with an adeno-associated virus (AAV) expressing human ANGPTL3.
  • AAV adeno-associated virus
  • mice were subcutaneously administered a single dose (5 mg/kg) of siNA test article.
  • the animals were sacrificed; the right, lateral liver lobe of each animal was collected for RT- qPCR and blood of each animal was collected for ELISA.
  • RNA was extracted using the Rneasy Mini Kit (Qiagen, 74106), according to the manufacturer’s protocol.
  • RNA quantity and quality was analyzed with a NanoDropTM Lite Spectrophotometer (Thermo Scientific), and cDNA was synthesized using the SuperScript IV VILO Master Mix (Invitrogen, 11756500), according to the manufacturer’s protocol.
  • Gene expression was measured using TaqMan Fast Advanced Master Mix (Applied Biosystems, 4444964), and the following TaqMan Gene Expression assays (Applied Biosystems, 4331182): Actb (Mm00607939_s1), ANGPTL3 (Hs00205581_m1), B2m (Mm00437762_m1), and Hmbs (Mm01143545_m1). Actb, B2m, and Hmbs served as the endogenous control genes.
  • dsNA-240, dsNA-241, dsNA-243 and dsNA-244 showed better human ANGPTL3 RNA and protein knockdown than comparative control dsNA-283.
  • AAV adeno-associated virus
  • mice were subcutaneously administered a single dose (0.3, 1.5, or 5.0 mg/kg) of an siNA (dsNA-283; dsNA-244) or a single dose of 5 mg/kg (dsNA-247; dsNA-248; dsNA-249). Twenty-eight days after administration, the animals were sacrificed; the right, lateral liver lobe of each animal was collected for RT- qPCR and blood of each animal was collected for ELISA. (Table 13). Table 13 – Example 7 Design Summary Table
  • Results are presented as percent change in expression relative to the expression levels of negative vehicle control samples.
  • serum was separated from blood samples.
  • Levels human ANGPTL3 protein in sera were quantified using ELISA (R&D Systems, DANL30) according to the manufacturer’s protocol.
  • Results are presented as percent change in expression relative to the expression levels of negative vehicle control samples (FIG. 3)
  • the RQ of gene expression was calculated via the 2 ⁇ Ct (RQ) method. Results are presented as percent change in expression relative to the expression levels of negative vehicle control samples. Knockdown of ANGPTL3 RNA in the liver samples is shown in FIG. 5. Serum was separated from blood samples. Human ANGPTL3 protein levels in the serum were quantified using ELISA (Abcam, ab254510) (FIG. 4). Triglycerides and total cholesterol levels in the serum were also quantified using a Roche Cobas 6000 c501 Chemistry Analyzer. Absolute protein and lipid concentrations are reported (Table 16; FIG. 4). Table 16. Maximum Percent Serum Lipid Reduction vs. Vehicle-Only Negative Control During 28-Day Treatment Period
  • liver ANGPTL3 RNA knockdown experiment showed that dsNA- 240, dsNA-244, dsNA-245 and dsNA-247 showed greater target RNA knockdown than comparative control dsNA-283, while dsNA-243 and dsNA-246 showed less target RNA knockdown than the comparative control siNA.
  • results of the serum ANGPTL3 protein knockdown exhibited a similar trend with dsNA-240, dsNA-244 and dsNA-247 showing a higher percentage of target protein knockdown than comparative control dsNA-283, while dsNA-243, dsNA-245 and dsNA-246 showed similar target protein knockdown as the comparative control.
  • RNA-Seq Off-Target Analysis To assess the off-target profile, 10 nM siNA was transfected into PHH and after 24 hours, total RNA was isolated. On target RNA knockdown (human ANGPTL3) in total RNA was confirmed by RT-PCR and standard RNA-Seq was carried out by Genewiz/Azenta.
  • dsNA-240 has good potency and knockdown capability, it does not have ideal off target profile as it has 45 DE genes identified in the RNA-Seq analysis.
  • the seed region of dsNA-240 would be an ideal candidate for testing chemical modifications to reduce off targets while preserving the potency.
  • Example 10 Modifying siNA candidate dsNA-240 by applying Aligos chemistries to destabilize the seed region.
  • a seed destabilizing strategy was applied. This strategy involves incorporation of various nucleotide modifications, including UNA, seco, 3-OH, 3-OCP, or xylo-F chemistries, into key positions in the antisense seed region, which include positions 6 and 7 from the 5’ end of the antisense strand.
  • modified siNAs generated via this strategy were tested, all candidates maintained in vitro potency (Table 18). Additional seed destabilized siNAs were generated in a second subsequent round (Table 19), This second round of testing further confirmed that seed destabilization does not significantly affect compound potency.
  • CM complete sequence match
  • SM seed region
  • Seed region of siNA is referring to nucleotide #2 to #8 of the antisense strand numbering from 5’ end.
  • siNA seed region was referring to nucleotide #2 to #8 of the antisense strand numbering from 5’ end.
  • psiCheck 4 tandem repeats of siNA seed region in psiCheck.
  • siNA under investigation with serial dilutions were transfected into Cos7 cells along with constant concentration of either on-target plasmid or off-target plasmid.
  • Cos-7 cells containing on- or off- target plasmid, treated with siNA were incubated for 72 hours.
  • Renilla luciferase assay was carried out according to Promega’s protocol. Prism software was used to plot on-target (CM) and off-target (SM) dose response curves.
  • AAV adeno-associated virus
  • Results are presented as percent change in expression relative to the expression levels of negative vehicle control samples (FIG. 7).
  • the results of the experiment showed that ANGPTL8 siNA dsNA-149, dsNA-150, dsNA-151, dsNA-152, dsNA-153 provided greater human ANGPTL8 RNA knockdown than comparative control siNA dsNA-285 (FIG. 7).
  • Example 13 hANGPTL3 siNA and hANGPTL8 siNA Combination in human ANGPTL3/8 Double Knock-In Mouse Study To evaluate certain ANGPTL3 and ANGPTL8 siNA agents, a knock-in mouse model that expresses both human ANGPTL3 and ANGPTL8 (KI-ANGPTL3/8) was used.
  • mice were subcutaneously administered a single dose (1 or 5 mg/kg) of one siNA or a combination of two siNAs.
  • combination of ANGPTL3 and ANGPTL8 siNA was achieved by co-formulation of two siNAs in a single injection in this example.
  • Blood was collected at various timepoints throughout the study for lipid quantification. Twenty-eight days after administration, the animals were sacrificed; the right, lateral liver lobe of each animal was collected for RT-qPCR and blood of each animal was collected for ELISA and lipid quantification.(Tables 20 and 21) Table 20 – Treatment Groups for Example 13.
  • RT-qPCR reactions were run on the QuantStudioTM 6 Pro Real-Time PCR System (Applied Biosystems). The RQ of gene expression was calculated via the 2 ⁇ Ct (RQ) method. Results are presented as percent change in expression relative to the expression levels of negative vehicle control samples. Serum was separated from blood samples. Human ANGPTL3 protein levels in the serum were quantified using ELISA (R&D Systems, DANL30). Results are presented as percent change in expression of each treatment group at day 28 relative to each respective group’s expression level pre-dose. Triglycerides and total cholesterol levels in the serum were also quantified using a Roche Cobas 6000 c501 Chemistry Analyzer. Absolute lipid concentrations are reported.
  • FIG. 8 The results of the serum tests for total cholesterol and triglycerides are shown in FIG. 8.
  • Some combinations of the ANGPTL3 and ANGPTL8 siNAs exhibited a greater reduction in total cholesterol levels as compared to the comparative control siNA and single siNA treatments on Day 7 post-dose (FIG. 8A).
  • the combination of dsNA-244 and dsNA-152 showed a reduction in total cholesterol levels of 32%, while dsNA-244 alone only exhibited 19% reduction and dsNA-152 alone exhibited 29% reduction. This combination’s reduction was also significantly more than the comparative control dsNA-283 alone, which exhibited a reduction of only 25%.
  • the combination treatment of dsNA- 241 and dsNA-152 had a reduction of total cholesterol levels of 40%, which is a greater percent reduction as compared to dsNA-241 alone (31%) and ds-NA-152 alone (37%).
  • the combination of dsNA-244 and ds-152 also had a higher reduction of total cholesterol (40%) than either dsNA-244 (37%) or dsNA-152 alone.
  • knockdown of both ANGPTL3 and ANGPTL8 by dsNA-244 and dsNA-150 respectively showed a 49% reduction in total cholesterol, as compared to dsNA-244 alone or to dsNA-150 alone (39%).
  • dsNA-244 and dsNA-150 result in a likewise greater reduction in total cholesterol (39%) than either dsNA-244 or dsNA-150 alone. All of these combinations had a significantly greater reduction in total cholesterol levels than the comparative control dsNA-283 (21%).
  • the increase in the ability of combinations of siNAs to reduce total cholesterol levels was also seen at the final timepoint of Day 28 post-dose (FIG. 8D).
  • the combination of dsNA-241 and dsNA-150 reduced total cholesterol levels by 53% at Day 28, whereas dsNA- 241 alone and dsNA-150 alone both showed 41%.
  • dsNA-244 and dsNA-150 together also had an increased reduction in total cholesterol levels (45%) as compared to dsNA-244 alone (43%) and ds-150 alone. Both combinations showed significantly greater reductions in total cholesterol than comparative control dsNA-283 alone (-32%).
  • a similar effect is seen for the combination siNA treatments when serum triglyceride levels were analyzed at Day 14 post-dose (FIG. 8E).
  • the combination of dsNA-241 and dsNA-152 resulted in a 48% reduction in serum triglyceride levels as compared to dsNA-241 alone (40%) or dsNA-152 alone (19%).
  • Example 15 The Report of Production Process for intermediate 1b Preparation of (1b): To a solution of 1a (100.0 g, 787.4 mmol) in HCONH2 (800 mL) was stirred at 150°C for 8 hour. LC-MS and TLC show SM was completely consumed. The mixture was cooling to 0 ⁇ 5°C, and was added water (400 mL), the solid was collected by filtration.
  • Example 16 Production of Example 16 Monomer. Preparation of (2): To a solution of 1 (100.0 g, 387.3 mmol) in ACN (1.0 L). Then the mixture was added Ceric ammonium nitrate (106.2 g, 193.6 mmol) and I 2 (59.0 g, 232.4 mmol). The mixture was stirred at r.t for 3 h, the solid was collected by filtration. The filter cake was rinsed with ACN (2 x 500 mL), drying by rotary evaporator to give crude 2 (120.0 g) as a white solid without further purified and used directly for the next step. ESI-LCMS: m/z 384.9 [M+H] + .
  • Example 17 Production of Example 17 Monomer.

Landscapes

  • Health & Medical Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Biomedical Technology (AREA)
  • General Engineering & Computer Science (AREA)
  • Wood Science & Technology (AREA)
  • Organic Chemistry (AREA)
  • Chemical & Material Sciences (AREA)
  • Biotechnology (AREA)
  • Molecular Biology (AREA)
  • Zoology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Endocrinology (AREA)
  • Microbiology (AREA)
  • Plant Pathology (AREA)
  • Biophysics (AREA)
  • Biochemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Physics & Mathematics (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicinal Preparation (AREA)

Abstract

The present disclosure is in the field of pharmaceutical compounds and preparations and method of their use in the treatment of disease. Described are short interfering nucleic acid (siNA) molecules comprising modified nucleotides that target ANGPTL3 and ANGPTL8 as well as optimized co-formulation ratio or linker strategy in combining the ANGPTL3 siNA and ANGPTL8 siNA, compositions containing the same, and uses thereof for treating or preventing diseases. In particular, the present disclosure is in the field of siNA molecules that target ANGPTL3 and ANGPTL8 for treatment of cardiovascular disease and nonalcoholic fatty liver.

Description

Short Interfering Nucleic Acid (siNA) Molecules Targeting ANGPTL 3 and ANGPTL 8 and Methods of Using the Same TECHNICAL FIELD The present disclosure is in the field of pharmaceutical compounds and preparations and method of their use in the treatment of disease. Described are short interfering nucleic acid (siNA) molecules comprising modified nucleotides that target ANGPTL3 or ANGPTL8, compositions containing the same, and uses thereof for treating or preventing diseases. In particular, the present disclosure is in the field of siNA molecules targeting ANGPTL3 and ANGPTL8 for treatment of cardiovascular disease and nonalcoholic fatty liver, among other diseases. BACKGROUND The following discussion is merely provided to aid the reader in understanding the disclosure and is not admitted to describe or constitute prior art thereto. Triglycerides are the main form of lipid that provides and stores energy in the human body. Lipoprotein lipase (LPL) breaks down triglycerides in lipoproteins, such as chylomicrons (CM) and very low-density lipoproteins (VLDL), into two free fatty acids and one monoacylglycerol molecule. ANGPTL3, also known as Angiopoietin-like protein 3, is a protein exclusively and constitutively expressed in the liver. ANGPTL8, also known as lipasin, is expressed in the liver and adipose tissues after fed conditions. ANGPTL3, as well as ANGTL4, are inhibitors of LPL and they work in coordination with the cofactor ANGPTL8. The inhibition of LPL subsequently increases plasma triglyceride (TG), LDL cholesterol (LDL-c), Total Cholesterol (TC), and HDL-cholesterol (HDL-c). ANGPTL3 can form a complex with the co-factor ANGPTL8 (ANGPTL3/8). The ANGPTL3/8 complex is a much stronger inhibitor of LPL in oxidative tissues such as heart and muscle (i.e., >100 fold) than ANGPTL3 alone, and ANGPTL8 is not an inhibitor of LPL. Reducing the ANGPTL3/8 complex by applying a combination of ANGPTL3 siNA and ANGPTL8 siNA treatments could achieve greater TG, TC, and LDL-c reduction than ANGPTL3 siNA treatment alone. ANGPTL3 also inhibits endothelial lipase (EL) which results in elevation of HDL-c. ANGPTL8 cofactor is not required in inhibition of EL by ANGPTL3. ANGPTL3 siNA treatment alone has been shown to reduce “good” cholesterol HDL-c which can be a problem. Applying a combination of ANGPTL3 and ANGPTL8 siNA at optimum ratio could maximally reduce the abundance of the ANGPTL3/8 complex in heart and muscles, resulting in maximum reduction of LPL inhibition and maximum TG, TC, and LDL-c reduction in circulation, while leaving sufficient free ANGPTL3 to inhibit EL. Compared to ANGPTL3 siNA monotherapy, the amount of ANGPTL3 siNA dosed in combo therapy (ANGPTL3 siNA + ANGPTL8 siNA) is going to be lower, resulting in greater inhibition of EL by free ANGPTL3 and a better HDL-c profile. By optimizing the ANGPTL3 siNA vs ANGPTL8 siNA ratio, is possible to achieve lower HDL-c reduction than with ANGPTL3 siNA mono-therapy, or no HDL-c reduction, or even slight elevation of HDL-c in circulation. Hypertriglyceridemia is associated with increased risk of atherosclerotic cardiovascular diseases and with fatty acid liver disease. Treatments that target the production and clearance of triglyceride-rich lipoproteins (TRLs) mitigate these complications. ANGPTL3 inhibition also lowers LDL particle concentration and may therefore have an overall advantage as a lipid-regulating agent. ANGPTL3 controls LPL activity and has previously been treated as a target for inhibition by siNA, antisense oligonucleotide (ASO) and monoclonal antibodies. But the dual inhibition of ANGPTL3 and ANGPTL8 by siNA or ASO and the effects of dual inhibition on LPL and EL activities have not been previously explored. RNA interference (RNAi) is a biological response to double-stranded RNA that mediates resistance to both endogenous parasitic and exogenous pathogenic nucleic acids, and regulates the expression of protein-coding genes. The short interfering nucleic acids (siNA), such as siRNA, have been developed for RNAi therapy to treat a variety of diseases. For instance, RNAi therapy has been proposed for the treatment of metabolic diseases, neurodegenerative diseases, cancer, and pathogenic infections. SUMMARY The present disclosure provides siNA molecules useful for targeting and inhibiting ANGPTL3 and ANGPTL8 in optimized combination ratios, which subsequently increases LPL activity in oxidative tissues, such as, heart and muscles resulting in decreased TG, TC, and LDL-c in circulation. The combination therapy can maintain or increase HDL-c by maintaining a high level of free ANGPTL3 and subsequence inhibition of EL. Compared with bi-specific monoclonal antibodies that target the ANGPTL3/8 complex, the dual targeting siNA approach reduces dosing frequency and results in better patient compliance. In one aspect, the present disclosure provides a short interfering nucleic acid (siNA) molecule comprising: (a) a sense strand comprising a first nucleotide sequence that is at least about 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% identical to an RNA corresponding to a target gene, wherein the first nucleotide sequence: is 15 to 30 nucleotides in length; and comprises 15 or more modified nucleotides independently selected from a 2’-O-methyl nucleotide and a 2’-fluoro nucleotide, wherein at least one modified nucleotide is a 2’-O- methyl nucleotide and the nucleotide at position 3, 5, 7, 8, 9, 10, 11, 12, 14, 17, and/or 19 from the 5’ end of the first nucleotide sequence is a 2’-fluoro nucleotide or wherein at least one modified nucleotide is a 2’-O-methyl nucleotide and at least one modified nucleotide is a 2’-fluoro nucleotide; and an antisense strand comprising a second nucleotide sequence that is at least about 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% complementary to the RNA corresponding to the target gene, wherein the second nucleotide sequence: is 15 to 30 nucleotides in length; and comprises 15 or more modified nucleotides independently selected from a 2’-O-methyl nucleotide and a 2’-fluoro nucleotide, wherein at least one modified nucleotide is a 2’-O- methyl nucleotide and at least one modified nucleotide is a 2’-fluoro nucleotide; or (b) a sense strand comprising a first nucleotide sequence that is at least about 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% identical to an RNA corresponding to a target gene, wherein the first nucleotide sequence: (i) is 15 to 30 nucleotides in length; and (ii) comprises 15 or more modified nucleotides independently selected from a 2’-O- methyl nucleotide and a 2’-fluoro nucleotide, wherein at least one modified nucleotide is a 2’-O-methyl nucleotide and at least one modified nucleotide is a 2’-fluoro nucleotide; and an antisense strand comprising a second nucleotide sequence that is at least about 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% complementary to the RNA corresponding to the target gene, wherein the second nucleotide sequence: (iii) is 15 to 30 nucleotides in length; and (iv) comprises 15 or more modified nucleotides independently selected from a 2’-O-methyl nucleotide and a 2’-fluoro nucleotide, wherein at least one modified nucleotide is a 2’-O-methyl nucleotide and the nucleotide at position 2, 5, 6, 8, 10, 14, 16, 17, and/or 18 from the 5’ end of the second nucleotide sequence is a 2’- fluoro nucleotide; wherein the target gene is a gene encoding ANGPTL8 or a gene that controls expression of ANGPTL8. In one aspect, the present disclosure provides a short interfering nucleic acid (siNA) molecule comprising: (a) a sense strand comprising a first nucleotide sequence that is at least about 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% identical to an RNA corresponding to a target gene, wherein the first nucleotide sequence: is 15 to 30 nucleotides in length; and comprises 15 or more modified nucleotides independently selected from a 2’-O-methyl nucleotide and a 2’-fluoro nucleotide, wherein at least one modified nucleotide is a 2’-O- methyl nucleotide and the nucleotide at position 3, 5, 7, 8, 9, 10, 11, 12, 14, 17, and/or 19 from the 5’ end of the first nucleotide sequence is a 2’-fluoro nucleotide or wherein at least one modified nucleotide is a 2’-O-methyl nucleotide and at least one modified nucleotide is a 2’-fluoro nucleotide; and an antisense strand comprising a second nucleotide sequence that is at least about 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% complementary to the RNA corresponding to the target gene, wherein the second nucleotide sequence: is 15 to 30 nucleotides in length; and comprises 15 or more modified nucleotides independently selected from a 2’-O-methyl nucleotide and a 2’-fluoro nucleotide, wherein at least one modified nucleotide is a 2’-O- methyl nucleotide and at least one modified nucleotide is a 2’-fluoro nucleotide; or (b) a sense strand comprising a first nucleotide sequence that is at least about 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% identical to an RNA corresponding to a target gene, wherein the first nucleotide sequence: (i) is 15 to 30 nucleotides in length; and (ii) comprises 15 or more modified nucleotides independently selected from a 2’-O- methyl nucleotide and a 2’-fluoro nucleotide, wherein at least one modified nucleotide is a 2’-O-methyl nucleotide and at least one modified nucleotide is a 2’-fluoro nucleotide; and an antisense strand comprising a second nucleotide sequence that is at least about 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% complementary to the RNA corresponding to the target gene, wherein the second nucleotide sequence: (iii) is 15 to 30 nucleotides in length; and (iv) comprises 15 or more modified nucleotides independently selected from a 2’-O-methyl nucleotide and a 2’-fluoro nucleotide, wherein at least one modified nucleotide is a 2’-O-methyl nucleotide and the nucleotide at position 2, 5, 6, 8, 10, 14, 16, 17, and/or 18 from the 5’ end of the second nucleotide sequence is a 2’- fluoro nucleotide; wherein the target gene is a gene encoding ANGPTL3 or a gene that controls expression of ANGPTL3. In another aspect, the present disclosure provides double-stranded short interfering nucleic acid (ds-siNA) molecules comprising: (a) a sense strand comprising a first nucleotide sequence, wherein the first nucleotide sequence is 15 to 30 nucleotides in length and comprises a nucleotide sequence as set forth in in any one of SEQ ID NOs: 1-316, and an antisense strand comprising a second nucleotide sequence, wherein the second nucleotide sequence is 15 to 30 nucleotides in length and comprises a nucleotide sequence that is a substantial reverse complement of the first nucleotide sequence; or (b) a sense strand comprising a first nucleotide sequence, wherein the first nucleotide sequence is 15 to 30 nucleotides in length and comprises a nucleotide sequence that is a substantial reverse complement of a second nucleotide sequence; and an antisense strand comprising the second nucleotide sequence, wherein the second nucleotide sequence is 15 to 30 nucleotides in length and comprises a nucleotide sequence as set forth in in any one of SEQ ID NOs: 317-632. In another aspect, the present disclosure provides double-stranded short interfering nucleic acid (ds-siNA) molecules comprising: (a) a sense strand comprising a first nucleotide sequence, wherein the first nucleotide sequence is 15 to 30 nucleotides in length and comprises a nucleotide sequence as set forth in in any one of SEQ ID NOs: 633-878, and an antisense strand comprising a second nucleotide sequence, wherein the second nucleotide sequence is 15 to 30 nucleotides in length and comprises a nucleotide sequence that is a substantial reverse complement of the first nucleotide sequence; or (b) a sense strand comprising a first nucleotide sequence, wherein the first nucleotide sequence is 15 to 30 nucleotides in length and comprises a nucleotide sequence that is a substantial reverse complement of a second nucleotide sequence; and an antisense strand comprising the second nucleotide sequence, wherein the second nucleotide sequence is 15 to 30 nucleotides in length and comprises a nucleotide sequence as set forth in any one of SEQ ID NOs: 879-1125. In another aspect, the present disclosure provides double-stranded short interfering nucleic acid (ds-siNA) molecules comprising: (a) a sense strand comprising a nucleotide sequence as set forth in any one of SEQ ID NOs: 1-316, and (b) an antisense strand. In another aspect, the present disclosure provides double-stranded short interfering nucleic acid (ds-siNA) molecules comprising (a) a sense strand comprising a nucleotide sequence as set forth in any one of SEQ ID NOs: 633-878, and (b) an antisense strand. In another aspect, the present disclosure provides double-stranded short interfering nucleic acid (ds-siNA) molecules comprising (a) an antisense strand comprising a nucleotide sequence as set forth in any one of SEQ ID NOs: 317-632 (b) a sense strand. In another aspect, the present disclosure provides double-stranded short interfering nucleic acid (ds-siNA) molecules comprising (a) an antisense strand comprising a nucleotide sequence as set forth in any one of SEQ ID NOs: 879-1125 (b) a sense strand. In some embodiments of any of the foregoing aspects, the sense strand comprises SEQ ID NOs: 633, 634, 642, 653, 654, 674, 687, 688, 690-694, 708-713, 756-757, 765, 776, 777, 797, 810, 811, 813-817, or 831-836 and the antisense strand comprises SEQ ID NOs: 879, 880, 888, 899, 900, 920, 933, 934, 936-940, 954-959, 1003-1004, 1012, 1023, 1024, 1044, 1057, 1058, 1060-1064, or 1078-1083, respectively. In some embodiments, the sense strand comprises SEQ ID NOs: 633, 654, 688, 690, 708, 713, 756, 777, 811, 813, 831, or 836, and the antisense strand comprises SEQ ID NOs: 879, 900, 934, 936, 954, 959, 1003, 1024, 1058, 1060, 1078, or 1083, respectively. In some embodiments of any of the foregoing aspects, the sense and/or antisense strand further comprises a TT sequence at 3’ end. In some embodiments of any of the foregoing aspects, at least one end of the ds-siNA is a blunt end. In some embodiments of any of the foregoing aspects, at least one end of the ds-siNA comprises an overhang, wherein the overhang comprises at least one nucleotide, optionally wherein the at least one nucleotide comprises a 2’-O-methyl nucleotide, a deoxy nucleotide, or a uracil analogue or derivative. In some embodiments of any of the foregoing aspects, both ends of the ds-siNA comprise an overhang, wherein the overhang comprises at least one nucleotide, optionally wherein the at least one nucleotide comprises a 2’-O-methyl nucleotide, a deoxy nucleotide, or a uracil analogue or derivative. In some embodiments of any of the foregoing aspects or embodiments, the sense strand and/or the antisense strand independently comprise 1 or more mesyl phosphoramidate internucleoside linkages, or 1 or more phosphorothioate internucleoside linkages. In some embodiments of any of the foregoing aspects or embodiments, the sense strand comprises at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15 or more phosphorothioate internucleoside linkages. In some embodiments, (i) at least one phosphorothioate internucleoside linkage in the sense strand is between the nucleotides at positions 1 and 2 from the 5’ end of the sense strand sequence; (ii) at least one phosphorothioate internucleoside linkage is between the nucleotides at positions 2 and 3 from the 5’ end of the sense strand sequence. In some embodiments of any of the foregoing aspects or embodiments, the antisense strand further comprises at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15 or more phosphorothioate internucleoside linkages. In some embodiments: (i) at least one phosphorothioate internucleoside linkage in the antisense strand is between the nucleotides at positions 1 and 2 from the 5’ end of the antisense strand sequence; (ii) at least one phosphorothioate internucleoside linkage in the antisense strand is between the nucleotides at positions 2 and 3 from the 5’ end of the antisense strand sequence; (iii) at least one phosphorothioate internucleoside linkage in the antisense strand is between the nucleotides at positions 1 and 2 from the 3’ end of the antisense strand sequence; and/or (iv) at least one phosphorothioate internucleoside linkage is between the nucleotides at positions 2 and 3 from the 3’ end of the antisense strand sequence. In some embodiments of any of the foregoing aspects or embodiments, the sense strand comprises at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15 or more mesyl phosphoramidate internucleoside linkages. In some embodiments: (i) at least one mesyl phosphoramidate internucleoside linkage in the sense strand is between the nucleotides at positions 1 and 2 from the 5’ end of the sense strand sequence; (ii) at least one mesyl phosphoramidate internucleoside linkage is between the nucleotides at positions 2 and 3 from the 5’ end of the sense strand sequence. In some embodiments of any of the foregoing aspects or embodiments, the antisense strand further comprises at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15 or more mesyl phosphoramidate internucleoside linkages. In some embodiments: (i) at least one mesyl phosphoramidate internucleoside linkage in the antisense strand is between the nucleotides at positions 1 and 2 from the 5’ end of the antisense strand sequence; (ii) at least one mesyl phosphoramidate internucleoside linkage in the antisense strand is between the nucleotides at positions 2 and 3 from the 5’ end of the antisense strand sequence; (iii) at least one mesyl phosphoramidate internucleoside linkage in the antisense strand is between the nucleotides at positions 1 and 2 from the 3’ end of the antisense strand sequence; and/or (iv) at least one mesyl phosphoramidate internucleoside linkage is between the nucleotides at positions 2 and 3 from the 3’ end of the antisense strand sequence. In some embodiments of any of the foregoing aspects or embodiments, the antisense strand comprises one or more modified nucleotides. In some embodiments of any of the foregoing aspects or embodiments, the sense strand comprises one or more modified nucleotides. In some embodiments of any of the foregoing aspects or embodiments, the sense strand and the antisense strand each independently comprise one or more modified nucleotides. In some embodiments, the modified nucleotides are independently selected from 2’-O-methyl nucleotides and 2’-fluoro nucleotides. Other modified nucleotides that may be included in the sense strand, antisense strand, or both are disclosed herein. In some embodiments of any of the foregoing aspects or embodiments, the antisense strand, sense strand, first nucleotide sequence, and/or second nucleotide sequence comprises at least one modified nucleotide selected from:
Figure imgf000011_0001
, wherein Rx is a nucleobase, aryl, heteroaryl, or H,
Figure imgf000011_0002
(mun34), wherein Ry is a nucleobase,
Figure imgf000011_0003
Figure imgf000011_0004
Figure imgf000012_0001
Figure imgf000013_0001
(wherein * represent chiral center),
Figure imgf000013_0002
(vm(5mim))
Figure imgf000013_0003
(vm(56mido)), and
Figure imgf000013_0004
(vm(56amim)U, wherein R is H or Bz), wherein B is a nucleobase, aryl, heteroaryl, or H, and wherein or represents a phosphodiester linkage, a phosphorothioate linkage, a mesyl phosphoramidate linkage, or H. In some embodiments of any of the foregoing aspects or embodiments, the ds-siNA further comprises a phosphorylation blocker, a 5’-stabilized end cap, or a combination thereof. In some embotidments, the phosphorylation blocker is attached to the 5’ end of the sense strand, and optionally, wherein the phosphorylation blocker is attached to the 5’ end of the sense strand via one or more of a phosphodiester linkage, a phosphorothioate linkage, a mesyl phosphoramidate linkage, or a phosphorodithioate linkage. In some embodiments, the 5’-stabilized end cap is a 5’ vinyl phosphonate (e.g., vm). In some embodiments, the 5’- stabilized end cap is attached to the 5’ end of the antisense strand, and optionally, wherein the 5’-stabilized end cap is attached to the 5’ end of the antisense strand one or more of a phosphodiester linkage, a phosphorothioate linkage, a mesyl phosphoramidate linkage, or a phosphorodithioate linkage. In some embodiments, the 5’-stabilzied end cap comprises a structure of:
Figure imgf000014_0001
wherein B is a nucleobase, aryl, heteroaryl, or H; wherein represents a phosphodiester
Figure imgf000014_0002
linkage, a phosphorothioate linkage, or a mesyl phosphoramidate linkage. In some embodiments of any of the foregoing aspects or embodiments, the ds-siNA further comprises a galactosamine. In some embodiments, the galactosamine is an N- acetylgalactosamine comprising a structure of: , wherein
Figure imgf000014_0003
m is 1, 2, 3, 4, or 5; each n is independently 1 or 2; p is 0 or 1; each R is independently H; each Y is independently selected from –O-P(=O)(SH)–, –O-P(=O)(O)–, –O-P(=O)(OH)–, and -O-P(S)S-; Z is H or a second protecting group; either L is a linker or L and Y in combination are a linker; and A is H, OH, a third protecting group, an activated group, or an oligonucleotide. In some embodiments, the galactosamine is an N-acetylgalactosamine comprising a structure of:
Figure imgf000015_0001
, wherein Rz is OH or SH; and each n is independently 1 or 2. In some embodiments of any of the foregoing aspects or embodiments, the sense and/or anti-sense strand of the ds-siNA is conjugated to GalNAc, Folate, Cholesterol, or Palmitic Acid. In some embodiments of any of the foregoing aspects or embodiments, the ds-siNA reduces ANGPTL8 or ANGPTL3 expression by more than about 10 %, more than about 20% more than about 30%, more than about 40%, more than about 50%, more than about 60%, more than about 70%, more than about 80%, more than about 90%, or more than about 95%. In some embodiments of any of the foregoing aspects or embodiments, the ds-siNA reduces ANGPTL8 or ANGPTL3 expression by 100%. In some embodiments of any of the foregoing aspects or embodiments, the ds-siNA reduces ANGPTL8 or ANGPTL3 expression or activity with a EC50 value of 50 pM or less, of 40 pM or less, of 30 pM or less, of 20 pM or less, 15 pM or less, 10 pM or less, 5 pM or less, 2 pM or less, or 1 pM or less. In some embodiments of any of the foregoing aspects or embodiments, the ds-siNA has a CC50 value of more than 1 µM. In some embodiments of any of the foregoing aspects or embodiments, the ds-siNA comprises RNA nucleotides. In another aspect, the present disclosure provides ds-siNA, comprising a sense strand and an antisense strand, wherein the antisense strand comprises at its 5’ end one of the following
Figure imgf000016_0001
(vm, wherein B is a nucleobase),
Figure imgf000016_0002
(vm(5mim)),
Figure imgf000016_0003
(vm(56mido)), and
Figure imgf000016_0004
(vm(56amim)U, wherein R is H or Bz). In another aspect, the present disclosure provides pharmaceutical compositions comprising at least one ds-siNA disclosed herein and a pharmaceutically acceptable carrier or diluent. In another aspect, the present disclosure provides pharmaceutical compositions comprising two or more ds-siNA disclosed herein. For the purposes of pharmaceutical compositions that combine the disclosed ds-siNA, the combination of two or more ds-siNA disclosed herein will generally comprise at least one ds-siNA that reduces expression of ANGPTL3 and at least one ds-siNA that reduces expression of ANGPTL8. In another aspect, the present disclosure provides pharmaceutical compositions comprising at least a first siNA that reduces ANGPTL3 expression and a second siNA that reduces ANGPTL8 expression and a pharmaceutically acceptable carrier. In some embodiments, the first siNA that reduces ANGPTL3 expression comprises a nucleic acid sequence disclosed in Table 3, and the second siNA that reduces ANGPTL8 expression comprises a nucleic acid sequence disclosed in Table 1. In some embodiments, the first siNA that reduces ANGPTL3 expression is a ds-siNA disclosed in Table 4, and the second siNA that reduces ANGPTL8 expression is a ds-siNA disclosed in Table 2. In some embodiments, a weight ratio of the first siNA and the second siNA is selected from 1:10, 1:9, 1:8, 1:7, 1:6, 1:5, 1:4, 1:3, 1:2, 1:1, 2:1, 3:1, 4:1, 5:1, 6:1, 7:1, 8:1, 9:1, or 10:1. In some embodiments, a molar ration of the first siNA and the second siNA is selected from 1:10, 1:9, 1:8, 1:7, 1:6, 1:5, 1:4, 1:3, 1:2, 1:1, 2:1, 3:1, 4:1, 5:1, 6:1, 7:1, 8:1, 9:1, or 10:1. In some embodiments, the first siNA and the second siNA are connected by a linker, which may be a nucleotide-based linker or a non-nucleotide-based linker. The pharmaceutical compositions disclosed herein may be formulated for parenteral, ocular, nasal, transdermal, pulmonary, or topical administration or comprises a liposome that encapsulates the ds-siNA. The present disclosure also provides uses of the ds-siNA disclosed herein in the manufacture of a medicament for treating cardiovascular disease or nonalcoholic fatty liver disease. The present disclosure also provides uses of two or more ds-siNA disclosed herein in the manufacture of a medicament for treating cardiovascular disease or nonalcoholic fatty liver disease. The present disclosure also provides ds-siNA disclosed herein or the pharmaceutical compositions disclosed herein for use in treating cardiovascular disease or nonalcoholic fatty liver disease. The present disclosure also provides two or more ds-siNA disclosed herein for use in treating cardiovascular disease or nonalcoholic fatty liver disease. In another aspect the present disclosure provides methods of treating a disease in a subject in need thereof, comprising administering to the subject a ds-siNA disclosed herein or a pharmaceutical composition disclosed herein, wherein the disease is cardiovascular disease or nonalcoholic fatty liver disease. In another aspect the present disclosure provides methods of treating a disease in a subject in need thereof, comprising administering to the subject two or more ds-siNA disclosed herein, wherein the disease is cardiovascular disease or nonalcoholic fatty liver disease. In another aspect the present disclosure provides methods of treating a disease in a subject in need thereof, comprising administering to the subject a first pharmaceutical composition comprising at least a first ds-siNA that reduces ANGPTL3 expression and a second pharmaceutical composition comprising at least a second ds-siNA that reduces ANGPTL8 expression, wherein the disease is cardiovascular disease or nonalcoholic fatty liver disease. In some embodiments, the first siNA that reduces ANGPTL3 expression comprises a nucleic acid sequence disclosed in Table 3, and the second siNA that reduces ANGPTL8 expression comprises a nucleic acid sequence disclosed in Table 1. In some embodiments, the first siNA that reduces ANGPTL3 expression is a ds-siNA disclosed in Table 4, and the second siNA that reduces ANGPTL8 expression is a ds-siNA disclosed in Table 2. In some embodiments of the disclosed methods, the cardiovascular disease is Hypertriglyceridemia (HTG) or Familial Hypercholesterolemia (FH). In some embodiments of the disclosed methods, wherein the subject is a mammal, and optionally wherein the mammal is a human or a non-human primate. In some embodiments of the disclosed methods, wherein the ds-siNA or pharmaceutical composition(s) are administered intravenously, subcutaneously, or via inhalation. In some embodiments of the disclosed methods, wherein the subject has been treated with one or more additional therapeutic agents. In another aspect the present disclosure provides methods of reducing ANGPTL8 and/or ANGPTL3 expression or activity in a tissue, an organ, or a cell of a subject in need thereof, wherein the method comprises delivering to the subject one or more of the ds-siNA disclosed herein or the pharmaceutical compositions disclosed herein. In another aspect the present disclosure provides modified nucleosides selected from
Figure imgf000019_0001
(vm, wherein B is a nucleobase),
Figure imgf000019_0002
(vm(5mim)),
Figure imgf000019_0003
(vm(56mido)), and
Figure imgf000019_0004
(vm(56amim)U, wherein R is H or Bz). It should be appreciated that all combinations of the foregoing concepts and additional concepts discussed in greater detail below are provided as being part of the inventive subject matter disclosed herein and may be employed in any combination to achieve the benefits described herein. BRIEF DESCRIPTION OF THE DRAWINGS FIG. 1A-1F depicts examples of siNA molecules that target ANGPTL3. FIG. 2A-2B provides the effects of exemplary siNAs after treatment of COS-7 cells with a dose response curve. FIG. 2A shows the percentage of viable COS-7 cells after treatment. FIG. 2B shows percentage inhibition of cynomologus monkey ANGPTL3 after treatment with 6 exemplary siNAs as determined by luciferase assay. FIG. 3 depicts the percentage change in expression of ANGPTL3 protein from serum samples in groups treated with the ANGPTL3 siNAs relative to the expression levels from serum samples in the group treated with the negative vehicle control in the AAV-hANGPTL3 mice. Serum samples were taken at Day 28 post-dose. FIG. 4 depicts the percentage change in expression of ANGPTL3 protein from serum samples in groups treated with the ANGPTL3 siNAs relative to the expression levels from serum samples in a group treated with the negative vehicle control in the hANGPTL3 knock- in mice. Serum samples were taken at Day 28 post-dose. FIG. 5 depicts the percentage change in expression of ANGPTL3 RNA of the liver samples in groups treated with ANGPTL3 siNAs relative to the expression levels of the same RNA in the livers of the group treated with the negative vehicle control in the hANGPTL3 knock-in mice. Liver samples were collected at Day 28 post-dose. FIG. 6 depicts the results of the plasmid based on- and off-target luciferase assay for siNAs having destabilized seed regions. FIG. 7 depicts the percentage change in expression of ANGPTL8 RNA in the livers of the groups treated with ANGPTL8 siNAs relative to the expression levels of the same RNA in the livers of the group treated with the negative vehicle control in the AAV-hANGPTL8 mice. Livers samples were collected at Day 14 post-dose. FIGS. 8A, 8B, 8C, 8D, and 8E depict percent reduction in various lipids following single or combination hANGPTL3 siNA and hANGPTL8 siNA treatment in the human ANGPTL3/8 double knock-in mouse study. FIG. 8A shows the percent reduction in total cholesterol levels Day 7 post-dose. FIG. 8B shows the percent reduction in total cholesterol levels Day 14 post-dose. FIG. 8C shows the percent reduction in total cholesterol levels Day 21 post-dose. FIG. 8D shows the percent reduction in total cholesterol levels Day 28 post- dose. FIG. 8E shows the percent reduction in triglyceride levels Day 14 post-dose. DETAILED DESCRIPTION The present disclosure provides siNA that target and decrease expression of ANGPTL3 or ANGPTL8. ANGPTL3 (along with ANGPTL4) work in coordination with cofactor ANGPTL8 to regulate lipid and triglyceride metabolism. ANGPTL3 is exclusively and constitutively expressed in the liver, and its inhibition lowers LDL particle concentration. ANGPTL8 is expressed in the liver and adipose tissues and is induced under the fed conditions. Inhibiting or lowering expression of either or both of ANGPTL3 and ANGPTL8 could treat cardiovascular disease, nonalcoholic fatty liver disease (NAFLD), metabolic dysfunction-associated steatohepatitis (MASH), and other diseases associated with high triglyceride levels. The present disclosure provides combinations of ANGPTL3-targeting siNA and ANGPTL8-targeting siNA that can be used together to treatments as disclosed in more detail herein. Definitions It is to be understood that methods are not limited to the particular embodiments described, and as such may vary. It is also to be understood that the terminology used herein is for the purpose of describing particular embodiments only and is not intended to be limiting. The scope of the present technology will be limited only by the appended claims. Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. Although any methods and materials similar or equivalent to those described herein can also be used in the practice or testing of the present invention, representative illustrative methods and materials are now described. Where a range of values is provided, it is understood that each intervening value, to the tenth of the unit of the lower limit unless the context clearly dictates otherwise, between the upper and lower limit of that range and any other stated or intervening value in that stated range, is encompassed within the invention. The upper and lower limits of these smaller ranges may independently be included in the smaller ranges and are also encompassed within the invention, subject to any specifically excluded limit in the stated range. Where the stated range includes one or both of the limits, ranges excluding either or both of those included limits are also included in the invention. As used in the specification and claims, the singular form “a,” “an” and “the” include singular and plural references unless the context clearly dictates otherwise. As used herein, the term “comprising” is intended to mean that the compositions and methods include the recited elements, but not excluding others. “Consisting essentially of” when used to define compositions and methods, shall mean excluding other elements of any essential significance to the composition or method. “Consisting of” shall mean excluding more than trace elements of other ingredients for claimed compositions and substantial method steps. Embodiments defined by each of these transition terms are within the scope of this disclosure. Accordingly, it is intended that the methods and compositions can include additional steps and components (comprising) or alternatively including steps and compositions of no significance (consisting essentially of) or alternatively, intending only the stated method steps or compositions (consisting of). As used herein, “about” means plus or minus 10% as well as the specified number. For example, “about 10” should be understood as both “10” and “9-11.” As used herein, “optional” or “optionally” means that the subsequently described event or circumstance may or may not occur, and that the description includes instances where said event or circumstance occurs and instances where it does not. The terms “individual,” “subject,” and “patient” are used interchangeably herein, and refer to any individual mammal, e.g., bovine, canine, feline, equine, simian, porcine, camelid, bat, or human, being treated according to the disclosed methods or uses. In preferred embodiments, the subject is a human. As used herein, the phrases “effective amount,” “therapeutically effective amount,” and “therapeutic level” mean the siNA dosage or concentration in a subject that provides the specific pharmacological effect for which the siNA is administered in a subject in need of such treatment, i.e. to treat or prevent a cardiovascular disease, nonalcoholic fatty liver disease, etc. It is emphasized that a therapeutically effective amount or therapeutic level of an siNA will not always be effective in treating the diseases described herein, even though such dosage is deemed to be a therapeutically effective amount by those of skill in the art. For convenience only, exemplary dosages, drug delivery amounts, therapeutically effective amounts, and therapeutic levels are provided below. Those skilled in the art can adjust such amounts in accordance with standard practices as needed to treat a specific subject and/or condition. The therapeutically effective amount may vary based on the route of administration and dosage form, the age and weight of the subject, and/or the subject’s condition, including the type and severity of the disease or disorder. The terms “ANGPTL3” and “angiopoietin like 3” refer to nucleic acid sequences encoding a ANGPTL3 protein, peptide, or polypeptide. An example of an ANGPTL3 transcript is Genbank Accession No. NM_014495.4, which is incorporated by reference in its entirety. The term “ANGPTL3” may also include other ANGPTL3 encoding sequences, such as ANGPTL3 isoforms, mutant ANGPTL3 genes, splice variants of ANGPTL3 genes, and ANGPTL3 gene polymorphisms. The term “ANGPTL3” may also refer to a nucleic acid sequence that encodes the polypeptide gene product of a ANGPTL3 gene/transcript, e.g., a ANGPTL3 protein, peptide, or polypeptide according to for example Genbank Accession No. NM_014495.4 of which is incorporated by reference in its entirety. The terms “ANGPTL8,” “angiopoietin like 8,” and “lipasin” refer to nucleic acid sequences encoding a ANGPTL8 protein, peptide, or polypeptide. An example of an ANGPTL8 transcript is Genbank Accession No. NM_018687.7, which is incorporated by reference in its entirety. The term “ANGPTL8” may also include other ANGPTL8 encoding sequences, such as ANGPTL8 isoforms, mutant ANGPTL8 genes, splice variants of ANGPTL8 genes, and ANGPTL8 gene polymorphisms. The term “ANGPTL8” may also refer to a nucleic acid sequence that encodes the polypeptide gene product of a ANGPTL8 gene/transcript, e.g., a ANGPTL8 protein, peptide, or polypeptide according to for example Genbank Accession No. NM_018687.7 of which is incorporated by reference in its entirety. The terms “treatment” or “treating” as used herein with reference to reducing or eliminating the disease and/or improving or ameliorating one or more symptoms of the disease. The terms “prevent” or “preventing” as used herein with reference to a disease refer to precluding a disease from developing in a subject at risk of developing cardiovascular disease and nonalcoholic fatty liver disease. As used herein, the term “pharmaceutical composition” refers to the combination of at least one active agent with a carrier, inert or active, making the composition especially suitable for diagnostic or therapeutic use in vivo or ex vivo. As used herein, the term “pharmaceutically acceptable carrier” refers to any of the standard pharmaceutical carriers, such as a phosphate buffered saline solution, water, emulsions (e.g., such as an oil/water or water/oil emulsions), and various types of wetting agents. The compositions also can include stabilizers and preservatives. For examples of carriers, stabilizers and adjuvants, see, for example, Martin, Remington’s Pharmaceutical Sciences, 15th Ed., Mack Publ. Co., Easton, PA [1975]. The phrases “parenteral administration” and “administered parenterally” as used herein means modes of administration other than enteral and topical administration, usually by injection, and includes, without limitation, intravenous, intramuscular, intraarterial, intrathecal, intracapsular, intraorbital, intracardiac, intradermal, intraperitoneal, transtracheal, subcutaneous, subcuticular, intraarticular, subcapsular, subarachnoid, intraspinal and intrasternal injection and infusion. The phrases “systemic administration,” “administered systemically,” “peripheral administration” and “administered peripherally” as used herein mean the administration of a compound, drug or other material other than directly into the central nervous system, such that it enters the patient’s system and, thus, is subject to metabolism and other like processes, for example, subcutaneous administration. As used herein, the term “nucleobase” refers to a nitrogen-containing biological compound that forms a nucleoside. Examples of nucleobases include, but are not limited to, thymine, uracil, adenine, cytosine, guanine, and an analogue or derivative thereof. For the purposes of the present disclosure, a DNA sequence that replaces all the U residues of an RNA sequence with T residues is “identical” to the RNA sequence, and vice versa. Accordingly, a sequence that is “identical to an RNA corresponding to” a DNA sequence constitutes the DNA sequence with all T replaced by U. The presence of modified nucleotides or 2’-deoxynucleotides in a sequence does not make a sequence not “identical to an RNA” but rather a modified RNA. As used herein, “modified nucleotide” includes any nucleic acid or nucleic acid analogue residue that contains a modification or substitution in the chemical structure of an unmodified nucleotide base, sugar (including, but not limited to, 2’-substitution), or phosphate (including, but not limited to, alternate internucleotide linkers, such as phosphorothioates or the substitution of bridging oxygens in phosphate linkers with bridging sulfurs), or a combination thereof. Non-limiting examples of modified nucleotides are shown herein. Throughout the description, where compositions are described as having, including, or comprising specific components, or where processes and methods are described as having, including, or comprising specific steps, it is contemplated that, additionally, there are compositions of the present disclosure that consist essentially of, or consist of, the recited components, and that there are processes and methods according to the present disclosure that consist essentially of, or consist of, the recited processing steps. As a general matter, compositions specifying a percentage are by weight unless otherwise specified. Further, if a variable is not accompanied by a definition, then the previous definition of the variable controls. ANGPTL3, ANGPTL8, and Associated Diseases The siNA molecules and compositions described herein may be administered to a subject to treat a disease. Further disclosed herein are uses of any of the siNA molecules or compositions disclosed herein in the manufacture of a medicament for treating a disease. The disease being treated may be a disease associated with ANGPTL3 expression, ANGPTL8 expression, or both. ANGPTL3 is an inhibitor of LPL and functions in combination with the cofactor ANGPTL8. ANGPTL3 is exclusively and constitutively expressed in the liver, where ANGPTL8 is expressed in the adipose tissue and liver and is induced after fed conditions. LPL breaks down fat in the form triglyceride in lipoproteins such as chylomicrons (CM) and very low-density lipoproteins (VLDL), into two free fatty acids and one monoacylglycerol molecule. As used herein, “a disease or disorder associated with ANGPTL3,” “a disease or disorder associated with ANGPTL8,” and “a disease or disorder associated with ANGPTL3 and ANGPTL8” refers to a disease or disorder known in the art to be associated with altered LPL levels, activity, or both. A disease or disorder associated with ANGPTL3, ANGPTL8 or ANGPTL3 and ANGPTL8 may include cardiovascular disease (CVD) and nonalcoholic fatty liver disease (NAFLD), such as metabolic dysfunction-associated steatohepatitis (MASH). Non-limiting examples of cardiovascular disease include hypertriglyceridemia (HTG) and Familial Hypercholesterolemia (FM). Assessment of siNA molecules targeting ANGPTL3 and ANGPTL8 In some embodiments, a reporter system is used to assess the effectiveness of the siNA molecules of the siNA molecules of the present disclosure in modulating, reducing or inhibiting ANGPTL3 and ANGPTL8. In some embodiments, the reporter system is a luciferase assay. The expression of luciferase can be measured by methods well known in the art. In other embodiments, siNA targeting of a ANGPTL3 and ANGPTL8 target sequence is assessed by measuring ANGPTL3 and ANGPTL8 RNA or protein levels. In such embodiments, ANGPTL3 and ANGPTL8 RNA levels can be assessed by art-recognized methods such as RT-PCR, Northern blot, expression array, etc., and protein levels can be measured by immunoblotting, immunofluorescence, or other antibody-based methods. In some embodiments, the potency of the disclosed siNA molecules is determined by using a reporter assay. In some embodiments, the ds-siNA of the present disclosure reduces the level or activity of ANGPTL3 by more than about 10 %, more than about 20% more than about 30%, more than about 40%, more than about 50%, more than about 60%, more than about 70%, more than about 80%, more than about 90%, or more than about 95%. In some embodiments, the ds-siNA of the present disclosure reduces the level or activity of ANGPTL3 by 100%. In some embodiments, the ds-siNA of the present disclosure reduces the level or activity ANGPTL8 by more than about 10 %, more than about 20% more than about 30%, more than about 40%, more than about 50%, more than about 60%, more than about 70%, more than about 80%, more than about 90%, or more than about 95%. In some embodiments, the ds-siNA of the present disclosure reduces the level or activity of ANGPTL8 by 100%. In some embodiments, the level or activity of ANGPTL3 and ANGPTL8 can be determined by directly measuring ANGPTL3 and ANGPTL8 expression with for example RT-PCR followed by quantitative PCR. In some embodiments, the ds-siNA of the present disclosure reduces the expression level of ANGPTL3 and ANGPTL8 by more than about 10 %, more than about 20% more than about 30%, more than about 40%, more than about 50%, more than about 60%, more than about 70%, more than about 80%, more than about 90%, or more than about 95%. In some embodiments, the ds-siNA of the present disclosure reduces the expression level of ANGPTL3 and ANGPTL8 by 100%. In some embodiments, the ds- siNA of the present disclosure reduces the expression level of a ANGPTL3 and/or ANGPTL8 target genes by more than about 10 %, more than about 20% more than about 30%, more than about 40%, more than about 50%, more than about 60%, more than about 70%, more than about 80%, more than about 90%, or more than about 95%. In some embodiments, the ds- siNA of the present disclosure reduces the expression of a ANGPTL3 and/or ANGPTL8 target genes by 100%. In some embodiments, the ANGPTL3 target gene comprises NG_028169.1:5005-13798. In some embodiments, the ANGPTL8 target gene comprises NC_000019.10:11239619-11241943. By measuring percentage inhibition of ANGPTL3 and ANGPTL8 as determined by a reporter system after increasing amounts of ds-siNA, the EC50 (half maximum effective dosage) value of the ds-siNA molecules can be determined. In some embodiments, the ds- siNA has EC50 value of 50 pM or less, of 40 pM or less, of 30 pM or less, of 20 pM or less, 15 pM or less, 10 pM or less, 5 pM or less, 2 pM or less, or 1 pM or less. By measuring cell viability upon inhibition of ANGPTL3 and ANGPTL8, the CC50 (i.e. the concentration that is required for reducing cell viability by 50%) of the ds-siNA can be determined. In some embodiments, the ds-siNA has a CC50 value of more than about 0.5 µM, more than about 1 µM, more than about 1.5 µM, more than about 2 µM, more than about 2.5 µM, more than about 3 µM, more than about 4 µM, or more than about 5 µM. In some embodiments, the effectiveness of the ds-siNA can be determined by measuring the expression level of ANGPTL3 and ANGPTL8, and/or a target gene of ANGPTL3 and ANGPTL8. In some embodiments, the effectiveness of the ds-siNA can be determined by measuring the expression level of ANGPTL3. In some embodiments, the effectiveness of the ds-siNA can be determined by measuring the expression level of ANGPTL8. In some embodiments, the pharmacodynamics of a GalNAc conjugated ANGPTL3 siNA, GalNAc conjugated ANGPTL8 siNA or combination regimen comprising GalNAc conjugated ANGPTL3 siNA and GalNAc conjugated ANGPTL8 siNA as disclosed here in can be determined by in vivo mice studies infected with recombinant viruses AAV-human ANGPTL3 or AAV-human ANGPTL8 or both. Serial and terminal blood collections can be tested for target protein knock down through human ANGPTL3 ELISA, or human ANGPTL8 ELISA or both. Mouse livers can be tested for human NA knock down (human ANGPTL3 qPCR, or human ANMGPTL8 qPCR or both) or human target protein knockdown. In some embodiments, in vivo efficacy studies can be determined using knock in mice containing human ANGPTL3, human ANGPTL8 or both human ANGPTL3 and ANGPTL8. Mice can be fed with high fat diet and treated with GalNAc conjugated ANGPTL3 siNA, GalNAc conjugated ANGPTL8 siNA or combination regimen comprising GalNAc conjugated ANGPTL3 siNA and GalNAc conjugated ANGPTL8 siNA as disclosed herein. Serial and terminal blood collections can be tested for target protein knock down using ANGPTL3 ELISA, or human ANGPTL8 ELISA or both. Blood LDL-c, TG, HDL-c and total cholesterol can also be measured as efficacy end points. Liver TG and other liver lipid measurements can also be applied. In some embodiments, the GalNAc conjugated ANGPTL3 siNA, GalNAc conjugated ANGPTL8 siNA or combination regimen comprising GalNAC conjugated ANGPTL3 siNA and GalNAc conjugated ANGPTL8 siNA as disclosed herein can also be tested in monkey dyslipidemia model and monitor blood LDL-c, total Cholesterol, HDL-c, TG and liver fat. Any of the siNA molecules disclosed herein may further comprise one or more linkers independently selected from a phosphodiester (PO) linker, phosphorothioate (PS) linker, phosphorodithioate linker, mesyl phosphoramidate (Ms), and PS-mimic linker. In some embodiments, the PS-mimic linker is a sulfur linker. In some embodiments, the linkers are internucleoside linkers. Alternatively or additionally, the linkers may connect a nucleotide of the siNA molecule to at least one phosphorylation blocker, conjugated moiety, or 5’- stabilized end cap. In some embodiments, the linkers connect a conjugated moiety to a phosphorylation blocker or 5’-stabilized end cap. As disclosed in more detail here, the present disclosure provides empirical evidence showing that reducing expression of ANGPTL8 alone with ds-siNA, such as the ds-siNA disclosed here, can reduce cholesterol and triglyceride levels in an animal. As such, combinations of ds-siNA targeting ANGPTL3 and ds-siNA targeting ANGPTL8 can be used in combination to provide further therapeutic benefits. Short interfering nucleic acid (siNA) molecules As indicated above, the present disclosure provides siNA molecules comprising modified nucleotides. Any of the siNA molecules described herein may be double-stranded siNA (ds-siNA) molecules. The terms “siNA molecules” and “ds-siNA molecules” may be used interchangeably. In some embodiments, the ds-siNA molecules comprise a sense strand and an antisense strand. Tables 1-4 details sequences of the present disclosure useful for sense and antisense strands, disclosed in SEQ ID NOs: 1-632 (ANGPTL8) and 633-1125 (ANGPTL3). In some embodiments, the double-stranded short interfering nucleic acid (ds-siNA) molecule comprises (a) a sense strand comprising a first nucleotide sequence, wherein the first nucleotide sequence is 15 to 30 nucleotides in length and comprises or consists of a nucleotide sequence as set forth in SEQ ID NOs: 1-316; and (b) an antisense strand comprising a second nucleotide sequence, wherein the second nucleotide sequence is 15 to 30 nucleotides in length and comprises or consists of a nucleotide sequence that is the reverse complement of the first nucleotide sequence or has a nucleic acid sequence with sufficient complementarity (i.e., not necessarily 100%) to hybridize to the first nucleotide sequence. In some embodiments, the first nucleotide sequence comprises or consists of a nucleotide sequence as set forth in SEQ ID NOs: 1-316; and the second nucleotide sequence comprises or consists of a nucleotide sequence according to SEQ ID NOs: 317-632. In some embodiments, the double-stranded short interfering nucleic acid (ds-siNA) molecule comprises (a) a sense strand comprising a first nucleotide sequence, wherein the first nucleotide sequence is 15 to 30 nucleotides in length and comprises or consists of a nucleotide sequence as set forth in SEQ ID NOs: 633-878; and (b) an antisense strand comprising a second nucleotide sequence, wherein the second nucleotide sequence is 15 to 30 nucleotides in length and comprises or consists of a nucleotide sequence that is the reverse complement of the first nucleotide sequence or has a nucleic acid sequence with sufficient complementarity (i.e., not necessarily 100%) to hybridize to the first nucleotide sequence. In some embodiments, the first nucleotide sequence comprises or consists of a nucleotide sequence as set forth in SEQ ID NOs: 633-878; and the second nucleotide sequence comprises or consists of a nucleotide sequence according to SEQ ID NOs: 879-1125. In some embodiments, the double-stranded short interfering nucleic acid (ds-siNA) molecule comprises (a) a sense strand comprising a first nucleotide sequence, wherein the first nucleotide sequence is 15 to 30 nucleotides in length and comprises a nucleotide sequence with 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 99%, or 100% similarity to SEQ ID NOs: 1-316; and (b) an antisense strand comprising a second nucleotide sequence, wherein the second nucleotide sequence is 15 to 30 nucleotides in length and comprises a nucleotide sequence that is the reverse complement of the first nucleotide sequence. In some embodiments, the first nucleotide sequence comprises a nucleotide sequence with 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 99%, or 100% similarity to SEQ ID NOs:1-316; and the second nucleotide sequence comprises a nucleotide sequence with 75%, 80%, 85%, 90%, 91 %, 92%, 93%, 94%, 95%, 99%, or 100% similarity to SEQ ID NOs: 317-632. In some embodiments, the double-stranded short interfering nucleic acid (ds-siNA) molecule comprises (a) a sense strand comprising a first nucleotide sequence, wherein the first nucleotide sequence is 15 to 30 nucleotides in length and comprises a nucleotide sequence with 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 99%, or 100% similarity to SEQ ID NOs: 633-878; and (b) an antisense strand comprising a second nucleotide sequence, wherein the second nucleotide sequence is 15 to 30 nucleotides in length and comprises a nucleotide sequence that is the reverse complement of the first nucleotide sequence. In some embodiments, the first nucleotide sequence comprises a nucleotide sequence with 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 99%, or 100% similarity to SEQ ID NOs:633-878; and the second nucleotide sequence comprises a nucleotide sequence with 75%, 80%, 85%, 90%, 91 %, 92%, 93%, 94%, 95%, 99%, or 100% similarity to SEQ ID NOs: 879-1125. In some embodiments, the double-stranded short interfering nucleic acid (ds-siNA) molecule comprises (a) a sense strand comprising a first nucleotide sequence, wherein the first nucleotide sequence is 15 to 30 nucleotides in length and comprises a nucleotide sequence that differs by 1, 2, 3, 4, or 5 nucleotides from any one of SEQ ID NOs:1-316; and (b) an antisense strand comprising a second nucleotide sequence, wherein the second nucleotide sequence is 15 to 30 nucleotides in length and comprises a nucleotide sequence that is the reverse complement of the first nucleotide sequence. In some embodiments, the second nucleotide sequence comprises a nucleotide sequence that differs by 1, 2, 3, 4, or 5 nucleotides from any one of SEQ ID NOs:317-632. In some embodiments, the double-stranded short interfering nucleic acid (ds-siNA) molecule comprises (a) a sense strand comprising a first nucleotide sequence, wherein the first nucleotide sequence is 15 to 30 nucleotides in length and comprises a nucleotide sequence that differs by 1, 2, 3, 4, or 5 nucleotides from any one of SEQ ID NOs:633-878; and (b) an antisense strand comprising a second nucleotide sequence, wherein the second nucleotide sequence is 15 to 30 nucleotides in length and comprises a nucleotide sequence that is the reverse complement of the first nucleotide sequence. In some embodiments, the second nucleotide sequence comprises a nucleotide sequence that differs by 1, 2, 3, 4, or 5 nucleotides from any one of SEQ ID NOs:879-1125. In some embodiments, the present disclosure provides a double-stranded short interfering nucleic acid (ds-siNA) molecule comprising (a) a sense strand comprising a nucleotide sequence identical to SEQ ID NOs: 1-316, and (b) an antisense strand. In some embodiments, the present disclosure provides a double-stranded short interfering nucleic acid (ds-siNA) molecule comprising (a) an antisense strand comprising a nucleotide sequence identical to SEQ ID NOs: 317-632; and (b) a sense strand. In some embodiments, the present disclosure provides a double-stranded short interfering nucleic acid (ds-siNA) molecule comprising (a) a sense strand comprising a nucleotide sequence identical to SEQ ID NOs: 633-878, and (b) an antisense strand. In some embodiments, the present disclosure provides a double-stranded short interfering nucleic acid (ds-siNA) molecule comprising (a) an antisense strand comprising a nucleotide sequence identical to SEQ ID NOs: 879-1125; and (b) a sense strand. In some embodiments, the present disclosure relates to a double-stranded short interfering nucleic acid (ds-siNA) molecule comprising: (a) a sense strand comprising a first nucleotide sequence, wherein the first nucleotide sequence is 15 to 30 nucleotides in length and comprises a nucleotide sequence as set forth in SEQ ID NOs: 1-316, and an antisense strand comprising a second nucleotide sequence, wherein the second nucleotide sequence is 15 to 30 nucleotides in length and comprises a nucleotide sequence that is a substantial reverse complement of the first nucleotide sequence; or (b) a sense strand comprising a first nucleotide sequence, wherein the first nucleotide sequence is 15 to 30 nucleotides in length and comprises a nucleotide sequence that is a substantial reverse complement of a second nucleotide sequence; and an antisense strand comprising the second nucleotide sequence, wherein the second nucleotide sequence is 15 to 30 nucleotides in length and comprises a nucleotide sequence as set forth in SEQ ID NOs: 317-632. In some embodiments, the present disclosure relates to a double-stranded short interfering nucleic acid (ds-siNA) molecule comprising: (a) a sense strand comprising a first nucleotide sequence, wherein the first nucleotide sequence is 15 to 30 nucleotides in length and comprises a nucleotide sequence as set forth in SEQ ID NOs: 633-878, and an antisense strand comprising a second nucleotide sequence, wherein the second nucleotide sequence is 15 to 30 nucleotides in length and comprises a nucleotide sequence that is a substantial reverse complement of the first nucleotide sequence; or (b) a sense strand comprising a first nucleotide sequence, wherein the first nucleotide sequence is 15 to 30 nucleotides in length and comprises a nucleotide sequence that is a substantial reverse complement of a second nucleotide sequence; and an antisense strand comprising the second nucleotide sequence, wherein the second nucleotide sequence is 15 to 30 nucleotides in length and comprises a nucleotide sequence as set forth in SEQ ID NOs: 879-1125. In some embodiments, the present disclosure relates to a double-stranded short interfering nucleic acid (ds-siNA) molecule comprising (a) a sense strand comprising a nucleotide sequence identical to SEQ ID NOs: 1-316, and (b) an antisense strand. In some embodiments, the present disclosure relates to a double-stranded short interfering nucleic acid (ds-siNA) molecule comprising (a) a sense strand comprising a nucleotide sequence identical to SEQ ID NOs: 633-878, and (b) an antisense strand. In some embodiments, the present disclosure relates to a double-stranded short interfering nucleic acid (ds-siNA) molecule comprising (a) an antisense strand comprising a nucleotide sequence identical to SEQ ID NOs: 317-632, and (b) a sense strand. In some embodiments, the present disclosure relates to a double-stranded short interfering nucleic acid (ds-siNA) molecule comprising (a) an antisense strand comprising a nucleotide sequence identical to SEQ ID NOs: 879-1125, and (b) a sense strand. In some embodiments, the present disclosure provides a ds-siNA comprising a sense strand comprising any one of SEQ ID NOs: 633, 634, 643, 653, 654, 674, 687, 688, 690-693, 694, 708-713, 756-757, 756, 776, 777, 797, 810, 811, 813-817, or 831-836; and an antisense strand comprising any one of SEQ ID NOs: 879, 880, 888, 899, 900, 920, 933, 934, 936-940, 954-959, 1003-1004, 1012, 1023, 1024, 1044, 1057-1064, or 1078-1083, respectively. In some embodiments, the present disclosure provides a ds-siNA comprising a sense strand comprising any one of SEQ ID NOs: 633, 654, 688, 690, 708, 713, 756, 777, 811, 813, 831, or 836, and an antisense strand comprising any one of SEQ ID NOs: 879, 900, 934, 936, 954, 959, 1003, 1024, 1058, 1060, 1078, or 1083, respectively. The disclosed siNA molecules may comprise (a) at least one phosphorylation blocker, conjugated moiety, or 5’-stabilized end cap; and (b) a short interfering nucleic acid (siNA). In some embodiments, the phosphorylation blocker is a phosphorylation blocker disclosed herein. In some embodiments, the 5’-stabilized end cap is a 5’-stabilized end cap disclosed herein. The siNA may comprise any of the first nucleotide, second nucleotide, sense strand, or antisense strand sequences disclosed herein. The siNA may comprise 5 to 100, 5 to 90, 10 to 100, 10 to 90, 10 to 80, 10 to 70, 10 to 60, 10 to 50, 10 to 30, 10 to 25, 15 to 100, 15 to 90, 15 to 80, 15 to 70, 15 to 60, 15 to 50, 15 to 30, or 15 to 25 nucleotides. The siNA may comprise at least 5, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, or 40 nucleotides. The siNA may comprise less than or equal to 50, 45, 40, 39, 38, 37, 36, 35, 34, 33, 32, 31, 30, 29, 28, 27, 26, 25, 24, 23, 22, 21, 20, or 19 nucleotides. The nucleotides may be modified nucleotides. The siNA may be single stranded. The siNA may be double stranded. The siNA may comprise (a) a sense strand comprising 15 to 30, 15 to 25, 15 to 24, 15 to 23, 15 to 22, 15 to 21, 17 to 30, 17 to 25, 17 to 24, 17 to 23, 17 to 22, 17 to 21, 18 to 30, 18 to 25, 18 to 24, 18 to 23, 18 to 22, 18 to 21, 19 to 30, 19 to 25, 19 to 24, 19 to 23, 19 to 22, 19 to 21, 20 to 25, 20 to 24, 20 to 23, 21 to 25, 21 to 24, or 21 to 23 nucleotides; and (b) an antisense strand comprising 15 to 30, 15 to 25, 15 to 24, 15 to 23, 15 to 22, 15 to 21, 17 to 30, 17 to 25, 17 to 24, 17 to 23, 17 to 22, 17 to 21, 18 to 30, 18 to 25, 18 to 24, 18 to 23, 18 to 22, 18 to 21, 19 to 30, 19 to 25, 19 to 24, 19 to 23, 19 to 22, 19 to 21, 20 to 25, 20 to 24, 20 to 23, 21 to 25, 21 to 24, or 21 to 23 nucleotides. The siNA may comprise (a) a sense strand comprising about 15, 16, 17, 18, 19, 20, 21, 22, or 23 nucleotides; and (b) an antisense strand comprising about 15, 16, 17, 18, 19, 20, 21, 22, or 23 nucleotides. The siNA may comprise (a) a sense strand comprising about 19 nucleotides; and (b) an antisense strand comprising about 21 nucleotides. The siNA may comprise (a) a sense strand comprising about 21 nucleotides; and (b) an antisense strand comprising about 23 nucleotides. Any of the siNA molecules disclosed herein may further comprise one or more linkers independently selected from a phosphodiester (PO) linker, phosphorothioate (PS) linker, phosphorodithioate linker, mesyl phosphoramidate (Ms), and PS-mimic linker. In some embodiments, the PS-mimic linker is a 3’ or 5’ sulfur linker. In some embodiments, the linkers are internucleoside linkers. Alternatively or additionally, the linkers may connect a nucleotide of the siNA molecule to at least one phosphorylation blocker, conjugated moiety, or 5’-stabilized end cap. In some embodiments, the linkers connect a conjugated moiety to a phosphorylation blocker or 5’-stabilized end cap. In some embodiments, at least one end of the ds-siNA comprises an overhang, wherein the overhang comprises at least one nucleotide, preferably wherein the at least one nucleotide comprises a 2’-O-methyl nucleotide, a deoxy nucleotide, uracil, or a uracil analogue or derivative. In some embodiments, both ends of the ds-siNA comprise an overhang, wherein the overhang comprises at least one nucleotide, preferably wherein the at least one nucleotide comprises a 2’-O-methyl nucleotide, a deoxy nucleotide, uracil, or a uracil analogue or derivative. For the purposes of the present disclosure, a ds-siNA molecule may have the following formula: 5’-An1Bn2An3Bn4An5Bn6An7Bn8An9-3’ 3’-Cq1Aq2Bq3A q4Bq5Aq6Bq7Aq8Bq9Aq10Bq11Aq12-5’ wherein: the top strand is a sense strand comprising a first nucleotide sequence that is at least about 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, or 100% identical to an RNA corresponding to a target gene, wherein the first nucleotide sequence comprises 15 to 30 nucleotides; the bottom strand is an antisense strand comprising a second nucleotide sequence that is at least about 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, or 100% complementary to the RNA corresponding to the target gene, wherein the second nucleotide sequence comprises 15 to 30 nucleotides; each A is independently a 2’-O-methyl nucleotide or a nucleotide comprising a 5’ stabilized end cap or phosphorylation blocker; B is a 2’-fluoro nucleotide; C represents overhanging nucleotides and is a 2’-O-methyl nucleotide, a deoxy nucleotide, or uracil; n1 = 1-6 nucleotides in length; each n2, n6, n8, q3, q5, q7, q9, q11, and q12 is independently 0-1 nucleotides in length; each n3 and n4 is independently 1-3 nucleotides in length; n5 is 1-10 nucleotides in length; n7 is 0-4 nucleotides in length; each n9, q1, and q2 is independently 0-2 nucleotides in length; q4 is 0-3 nucleotides in length; q6 is 0-5 nucleotides in length; q8 is 2-7 nucleotides in length; and q10 is 2-11 nucleotides in length. The ds-siNA may further comprise a conjugated moiety. The conjugated moiety may comprise any of the galactosamines disclosed herein. The ds-siNA may further comprise (i) phosphorothioate internucleoside linkages between the nucleotides at positions 1 and 2 and positions 2 and 3 from the 5’ end of the sense strand; and (ii) phosphorothioate internucleoside linkages between any nucleotide position, for example at positions 1 and 2; positions 2 and 3; positions 3 and 4 etc. The ds-siNA may further comprise a 5’-stabilizing end cap. The 5’-stabilizing end cap may be a 2’-OMe-vinyl phosphonate (vm). The 5’- stabilizing end cap may feature a modified base (e.g., vm(5mim), and vm(56mido)), as disclosed herein. The 5’-stabilizing end cap may be attached to the 5’ end of the antisense strand. In some embodiments, the 2’-O-methyl nucleotide at position 1 from the 5’ end of the sense strand is further modified to contain a 5’ stabilizing end cap. In some embodiments, the 2’-O-methyl nucleotide at position 1 from the 5’ end of the antisense strand is further modified to contain a 5’ stabilizing end cap. In some embodiments, the 2’-O-methyl nucleotide at position 1 from the 5’ end of the sense strand is further modified to contain a phosphorylation blocker. In some embodiments, the 2’-O-methyl nucleotide at position 1 from the 3’ end of the sense strand is further modified to contain a phosphorylation blocker. In some embodiments, the 2’-O-methyl nucleotide at position 1 from the 5’ end of the antisense strand is further modified to contain a phosphorylation blocker. In some embodiments, the 2’-O-methyl nucleotide at position 1 from the 3’ end of the antisense strand is further modified to contain a phosphorylation blocker. An exemplary ds-siNA molecule may have the following formula: 5’-A2-4 B1A1-3 B2-3 A2-10 B0-1A0-4B0-1 A0-2-3’ 3’-C2A0-2B0-1A0-3B0-1A0-5B0-1A2-7 B1A2-11 B1A1-5’ wherein: the top strand is a sense strand comprising a first nucleotide sequence that is at least about 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, or 100% identical to an RNA corresponding to a target gene, wherein the first nucleotide sequence comprises 15 to 30 nucleotides; the bottom strand is an antisense strand comprising a second nucleotide sequence that is at least about 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, or 100% complementary to the RNA corresponding to the target gene, wherein the second nucleotide sequence comprises 15 to 30 nucleotides; each A is independently a 2’-O-methyl nucleotide or a nucleotide comprising a 5’ stabilized end cap or phosphorylation blocker; B is a 2’-fluoro nucleotide; C represents overhanging nucleotides and is a 2’-O-methyl nucleotide, a deoxy nucleotide, or uracil (or uracil containing nucleotide). The ds-siNA may further comprise a conjugated moiety. The conjugated moiety may comprise any of the galactosamines disclosed herein. The ds-siNA may further comprise (i) phosphorothioate internucleoside linkages between the nucleotides at positions 1 and 2 and positions 2 and 3 from the 5’ end of the sense strand; and (ii) phosphorothioate internucleoside linkages between the nucleotides at positions 1 and 2; positions 2 and 3; positions 19 and 20; and positions 20 and 21 from the 5’ end of the antisense strand. The ds- siNA may further comprise a 5’-stabilizing end cap. The 5’-stabilizing end cap may include a vinyl phosphonate. The vinyl phosphonate may be a deuterated vinyl phosphonate. The deuterated vinyl phosphonate may be a mono-deuterated vinyl phosphonate. The deuterated vinyl phosphonate may be a mono- or di-deuterated vinyl phosphonate. In some embodiments, the 5’ end of the antisense strand may comprise a
Figure imgf000038_0001
wherein B is a nucleobase or derivative thereof), a
Figure imgf000038_0002
a
Figure imgf000038_0003
(vm(56mido)), or a
Figure imgf000038_0004
(vm(56amim)U, wherein R is H or Bz). The 5’-stabilizing end cap may be attached to the 5’ end of the antisense strand. The 5’-stabilizing end cap may be attached to the 3’ end of the antisense strand. The 5’-stabilizing end cap may be attached to the 5’ end of the sense strand. The 5’-stabilizing end cap may be attached to the 3’ end of the sense strand. In some embodiments, the 2’-O-methyl nucleotide at position 1 from the 5’ end of the sense strand is further modified to contain a 5’ stabilizing end cap. In some embodiments, the 2’-O-methyl nucleotide at position 1 from the 5’ end of the antisense strand is further modified to contain a 5’ stabilizing end cap. In some embodiments, the 2’-O-methyl nucleotide at position 1 from the 5’ end of the sense strand is further modified to contain a phosphorylation blocker. In some embodiments, the 2’-O-methyl nucleotide at position 1 from the 3’ end of the sense strand is further modified to contain a phosphorylation blocker. In some embodiments, the 2’- O-methyl nucleotide at position 1 from the 5’ end of the antisense strand is further modified to contain a phosphorylation blocker. In some embodiments, the 2’-O-methyl nucleotide at position 1 from the 3’ end of the antisense strand is further modified to contain a phosphorylation blocker. The exemplary ds-siNA shown in FIG. 1A FIG. 1F comprise (i) a sense strand comprising 19-25 nucleotides (e.g., 21 nucleotides); and (ii) an antisense strand comprising 21-25 nucleotides (e.g., 23 nucleotides). The ds-siNA may optionally further comprise (iii) a conjugated moiety, wherein the conjugated moiety (e.g GalNAc4 is attached to the 3’ end or the 5’ end of the sense strand or the antisense strand. The ds-siNA may comprise a 2 nucleotide overhang at positions 22 and 23 from the 5’ end of the antisense strand. The ds- siNA may further comprise 1, 2, 3, 4, 5, 6 or more phosphorothioate (ps) internucleoside linkages or mesyl phosphoramidate internucleoside linkage (Ms). At least one phosphorothioate internucleoside linkage or mesyl phosphoramidate internucleoside linkage (Ms) may be between the nucleotides at positions 1 and 2 or positions 2 and 3 from the 5’ end of the sense strand. At least one phosphorothioate internucleoside linkage or mesyl phosphoramidate internucleoside linkage (Ms) may be between the nucleotides at positions 1 and 2 or positions 2 and 3 from the 5’ end of the antisense strand. At least one phosphorothioate internucleoside linkage or mesyl phosphoramidate internucleoside linkage (Ms) may be between the nucleotides at positions 19 and 20, positions 20 and 21, positions 21 and 22, or positions 22 and 23 from the 5’ end of the antisense strand or any combination thereof. As shown in FIGs. 1A -1F, about 4 nucleotides (e.g., 1, 2, 3, 4, 5, or 6) in the sense strand may be 2’-fluoro nucleotides. As shown in FIGs. 1A -1F, about 4 nucleotides (e.g., 1, 2, 3, 4, 5, or 6) in the antisense strand may be 2’-fluoro nucleotides. As shown in FIGs. 1A - 1F, 15-17 nucleotides in the sense strand may be 2’-O-methyl nucleotides. As shown in FIGs. 1A -1F 16-21 nucleotides in the antisense strand may be 2’-O-methyl nucleotides. In some embodiments, the 2’-O-methyl nucleotide at position 1 from the 5’ end of the sense strand can be further modified to contain a 5’ stabilizing end cap. In some embodiments, the 2’-O-methyl nucleotide at position 1 from the 5’ end of the antisense strand can be further modified to contain a 5’ stabilizing end cap. In some embodiments, the 2’-O- methyl nucleotide at position 1 from the 5’ end of the sense strand can be further modified to contain a phosphorylation blocker. In some embodiments, the 2’-O-methyl nucleotide at position 1 from the 3’ end of the sense strand can be further modified to contain a phosphorylation blocker. In some embodiments, the 2’-O-methyl nucleotide at position 1 from the 5’ end of the antisense strand can be further modified to contain a phosphorylation blocker. In some embodiments, the 2’-O-methyl nucleotide at position 1 from the 3’ end of the antisense strand can be further modified to contain a phosphorylation blocker. In some embodiments, the 2’-O-methyl nucleotide at position 1 from the 5’ end of the sense strand is a d2vd3 nucleotide, a d2vd3U nucleotide, a d2vd3A nucleotide, an omeco- d3 nucleotide, an omeco-d3U nucleotide, an omeco-d3A nucleotide, a 4h nucleotide, a 4hU nucleotide, a 4hA nucleotide, a v-mun nucleotide, a c2o-4h nucleotide, an omeco-munb nucleotide, a d2vm nucleotide, or a d2vmA nucleotide. In some embodiments, the 2’-O- methyl nucleotide at position 1 from the 5’ end of the antisense strand is a d2vd3 nucleotide, a d2vd3U nucleotide, a d2vd3A nucleotide, an omeco-d3 nucleotide, an omeco-d3U nucleotide, an omeco-d3A nucleotide, a 4h nucleotide, a 4hU nucleotide, a 4hA nucleotide, a v-mun nucleotide, a c2o-4h nucleotide, an omeco-munb nucleotide, a d2vm nucleotide, or a d2vmA nucleotide. In some embodiments, the 2’-O-methyl nucleotide at position 1 from the 3’ end of the sense strand is a d2vd3 nucleotide, a d2vd3U nucleotide, a d2vd3A nucleotide, an omeco-d3 nucleotide, an omeco-d3U nucleotide, an omeco-d3A nucleotide, a 4h nucleotide, a 4hU nucleotide, a 4hA nucleotide, a v-mun nucleotide, a c2o-4h nucleotide, an omeco-munb nucleotide, a d2vm nucleotide, or a d2vmA nucleotide. In some embodiments, the 2’-O-methyl nucleotide at position 1 from the 3’ end of the antisense strand is a d2vd3 nucleotide, a d2vd3U nucleotide, d2vd3A nucleotide, an omeco-d3 nucleotide, an omeco- d3U nucleotide, an omeco-d3A nucleotide, a 4h nucleotide, a 4hU nucleotide, a v-mun nucleotide, a c2o-4h nucleotide, an omeco-munb nucleotide, a d2vm nucleotide, or a d2vmA nucleotide. In some embodiments, at least 1, 2, 3, 4 or more 2’-fluoro nucleotides on the sense strand or antisense strand is a 2’-fluoro nucleotide mimic. In some embodiments, at least 1, 2, 3, 4 or more 2’-fluoro nucleotides on the sense strand or antisense strand is a fB, fN, f(4nh)Q, f4P, f2P, or fX nucleotide. In some embodiments, at least 1, 2, 3, 4 or more 2’- O-methyl nucleotide on the sense or antisense strand is a 2’-O-methyl nucleotide mimic. In some embodiments, one or more nucleotides in the sense strand and/or the antisense strand may be a 3’,4’ seco modified nucleotide in which the bond between the 3’ and 4’ positions of the furanose ring is broken (e.g., mun34). In some embodiments, the antisense strand, sense strand, first nucleotide sequence, and/or second nucleotide sequence comprises at least one modified nucleotide comprising (a) wherein Rx is a nucleobase, aryl, heteroaryl, or H, (b)
Figure imgf000041_0001
Figure imgf000041_0002
O (mun34), wherein Ry is a nucleobase, (c)
Figure imgf000041_0003
(F4P), or (d)
Figure imgf000041_0004
In some embodiments, the second nucleotide sequence (or antisense strand) comprises a modified nucleotide comprising at least
Figure imgf000041_0005
Figure imgf000041_0006
In some embodiments, the sense strand of the siNA, the antisense strand or the siNA, or both comprise at least 1, at least 2, at least 3, at least 4, or at least 5 nucleotide(s) comprising a structure independently selected from:
Figure imgf000041_0007
-
Figure imgf000042_0001
least 2, at least 3, at least 4, or at least 5 nucleotide analog(s) comprising a structure independently selected from: (gans^) and
Figure imgf000042_0003
(ganr^), wherein B is a nucleobase, an aryl,
Figure imgf000042_0002
heteroaryl, or H; wherein represents a phosphodiester linkage, a phosphorothioate linkage, or H; and wherein * represent chiral center (e.g., R or S isomer). In some embodiments, the sense strand, the antisense strand, or both each independently comprise 1 or more phosphorothioate internucleoside linkages. In some embodiments, the siNA further comprises a phosphorylation blocker. In some embodiments, the sense strand of the siNA, the antisense strand of the siNA, or both each independently comprise at least 1, at least 2, at least 3, at least 4, or at least 5 or more of
Figure imgf000043_0001
wherein Rx is a nucleobase, aryl, heteroaryl, or H,
Figure imgf000043_0002
(mun34) wherein Ry is a nucleobase,
Figure imgf000043_0003
(tn) wherein Ry is
Figure imgf000043_0004
a nucleobase, or combinations thereof. In some embodiments, the siNA further comprises a galactosamine. In some embodiments, the galactosamine is N-acetylgalactosamine (GalNAc) of Formula (VI):
Figure imgf000043_0005
wherein m is 1, 2, 3, 4, or 5; each n is independently 1 or 2; p is 0 or 1; each R is independently H; each Y is independently selected from –O-P(=O)(SH)–, –O-P(=O)(O)–, –O-P(=O)(OH)–, and -O-P(S)S-; Z is H or a second protecting group; either L is a linker or L and Y in combination are a linker; and A is H, OH, a third protecting group, an activated group, or an oligonucleotide. In some embodiments, the galactosamine is N-acetylgalactosamine (GalNAc) of Formula (VII):
Figure imgf000044_0001
, wherein Rz is OH or SH; and each n is independently 1 or 2.   A. siNA sense strand Any of the siNA molecules described herein may comprise a sense strand. The sense strand may comprise a first nucleotide sequence. The first nucleotide sequence may be 15 to 30, 15 to 25, 15 to 23, 17 to 23, 19 to 23, or 19 to 21 nucleotides in length. In some embodiments, the first nucleotide sequence is 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30 nucleotides in length. In some embodiments, the first nucleotide sequence is at least 19 nucleotides in length. In some embodiments, the first nucleotide sequence is at least 21 nucleotides in length. In some embodiments, the sense strand is the same length as the first nucleotide sequence. In some embodiments, the sense strand is longer than the first nucleotide sequence. In some embodiments, the sense strand may further comprise 1, 2, 3, 4, or 5 or more nucleotides than the first nucleotide sequence. In some embodiments, the sense strand may further comprise a deoxyribonucleic acid (DNA). In some embodiments, the DNA is thymine (T). In some embodiments, the sense strand may further comprise a TT sequence. In some embodiments, the sense strand may further comprise one or more modified nucleotides that are adjacent to the first nucleotide sequence. In some embodiments, the one or more modified nucleotides are independently selected from any of the modified nucleotides disclosed herein (e.g., 2’-fluoro nucleotide, 2’-O-methyl nucleotide, 2’-fluoro nucleotide mimic, 2’-O-methyl nucleotide mimic, or a nucleotide comprising a modified nucleobase). In some embodiments, the first nucleotide sequence comprises 15, 16, 17, 18, 19, 20, 21, 22, 23, or more modified nucleotides independently selected from a 2’-O-methyl nucleotide and a 2′-fluoro nucleotide. In some embodiments, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95% or 100% of the nucleotides in the first nucleotide sequence are modified nucleotides independently selected from a 2’-O-methyl nucleotide and a 2’-fluoro nucleotide. In some embodiments, 100% of the nucleotides in the first nucleotide sequence are modified nucleotides independently selected from a 2’-O-methyl nucleotide and a 2’- fluoro nucleotide. In some embodiments, the 2’-O-methyl nucleotide is a 2’-O-methyl nucleotide mimic. In some embodiments, the 2’-fluoro nucleotide is a 2’-fluoro nucleotide mimic. In some embodiments, between about 15 to 30, 15 to 25, 15 to 24, 15 to 23, 15 to 22, 15 to 21, 17 to 30, 17 to 25, 17 to 24, 17 to 23, 17 to 22, 17 to 21, 18 to 30, 18 to 25, 18 to 24, 18 to 23, 18 to 22, 18 to 21, 19 to 30, 19 to 25, 19 to 24, 19 to 23, 19 to 22, 19 to 21, 20 to 25, 20 to 24, 20 to 23, 21 to 25, 21 to 24, or 21 to 23 modified nucleotides of the first nucleotide sequence are 2’-O-methyl nucleotides. In some embodiments, between about 2 to 20 modified nucleotides of the first nucleotide sequence are 2’-O-methyl nucleotides. In some embodiments, between about 5 to 25 modified nucleotides of the first nucleotide sequence are 2’-O-methyl nucleotides. In some embodiments, between about 10 to 25 modified nucleotides of the first nucleotide sequence are 2’-O-methyl nucleotides. In some embodiments, between about 12 to 25 modified nucleotides of the first nucleotide sequence are 2’-O-methyl nucleotides. In some embodiments, at least about 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, or 22 modified nucleotides of the first nucleotide sequence are 2’-O- methyl nucleotides. In some embodiments, at least about 12 modified nucleotides of the first nucleotide sequence are 2’-O-methyl nucleotides. In some embodiments, at least about 13 modified nucleotides of the first nucleotide sequence are 2’-O-methyl nucleotides. In some embodiments, at least about 14 modified nucleotides of the first nucleotide sequence are 2’- O-methyl nucleotides. In some embodiments, at least about 15 modified nucleotides of the first nucleotide sequence are 2’-O-methyl nucleotides. In some embodiments, at least about 16 modified nucleotides of the first nucleotide sequence are 2’-O-methyl nucleotides. In some embodiments, at least about 17 modified nucleotides of the first nucleotide sequence are 2’-O-methyl nucleotides. In some embodiments, at least about 18 modified nucleotides of the first nucleotide sequence are 2’-O-methyl nucleotides. In some embodiments, at least about 19 modified nucleotides of the first nucleotide sequence are 2’-O-methyl nucleotides. In some embodiments, less than or equal to 25, 24, 23, 22, 21, 20, 19, 18, 17, 16, 15, 14, 13, 12, 11, 10, 9, 8, 7, 6, 5, 4, 3, or 2 modified nucleotides of the first nucleotide sequence are 2’- O-methyl nucleotides. In some embodiments, less than or equal to 21 modified nucleotides of the first nucleotide sequence are 2’-O-methyl nucleotides. In some embodiments, less than or equal to 20 modified nucleotides of the first nucleotide sequence are 2’-O-methyl nucleotides. In some embodiments, less than or equal to 19 modified nucleotides of the first nucleotide sequence are 2’-O-methyl nucleotides. In some embodiments, less than or equal to 18 modified nucleotides of the first nucleotide sequence are 2’-O-methyl nucleotides. In some embodiments, less than or equal to 17 modified nucleotides of the first nucleotide sequence are 2’-O-methyl nucleotides. In some embodiments, less than or equal to 16 modified nucleotides of the first nucleotide sequence are 2’-O-methyl nucleotides. In some embodiments, less than or equal to 15 modified nucleotides of the first nucleotide sequence are 2’-O-methyl nucleotides. In some embodiments, less than or equal to 14 modified nucleotides of the first nucleotide sequence are 2’-O-methyl nucleotides. In some embodiments, less than or equal to 13 modified nucleotides of the first nucleotide sequence are 2’-O-methyl nucleotides. In some embodiments, at least one modified nucleotide of the first nucleotide sequence is a 2’-O-methyl pyrimidine. In some embodiments, at least 5, 6, 7, 8, 9, or 10 modified nucleotides of the first nucleotide sequence are 2’-O-methyl pyrimidines. In some embodiments, at least one modified nucleotide of the first nucleotide sequence is a 2’-O-methyl purine. In some embodiments, at least 5, 6, 7, 8, 9, or 10 modified nucleotides of the first nucleotide sequence are 2’-O-methyl purines. In some embodiments, the 2’-O- methyl nucleotide is a 2’-O-methyl nucleotide mimic. In some embodiments, between 2 to 15 modified nucleotides of the first nucleotide sequence are 2’-fluoro nucleotides. In some embodiments, between 2 to 10 modified nucleotides of the first nucleotide sequence are 2’-fluoro nucleotides. In some embodiments, between 2 to 6 modified nucleotides of the first nucleotide sequence are 2’-fluoro nucleotides. In some embodiments, 1 to 6, 1 to 5, 1 to 4, or 1 to 3 modified nucleotides of the first nucleotide sequence are 2’-fluoro nucleotides. In some embodiments, at least 1, 2, 3, 4, 5, or 6 modified nucleotides of the first nucleotide sequence are 2’-fluoro nucleotides. In some embodiments, at least 1 modified nucleotide of the first nucleotide sequence is a 2’- fluoro nucleotide. In some embodiments, at least 2 modified nucleotides of the first nucleotide sequence are 2’-fluoro nucleotides. In some embodiments, at least 3 modified nucleotides of the first nucleotide sequence are 2’-fluoro nucleotides. In some embodiments, at least 4 modified nucleotides of the first nucleotide sequence are 2’-fluoro nucleotides. In some embodiments, at least 5 modified nucleotides of the first nucleotide sequence are 2’- fluoro nucleotides. In some embodiments, at least 6 modified nucleotides of the first nucleotide sequence are 2’-fluoro nucleotides. In some embodiments, 10, 9, 8, 7, 6, 5, 4, 3 or fewer modified nucleotides of the first nucleotide sequence are 2’-fluoro nucleotides. In some embodiments, 10 or fewer modified nucleotides of the first nucleotide sequence are 2’-fluoro nucleotides. In some embodiments, 7 or fewer modified nucleotides of the first nucleotide sequence are 2’-fluoro nucleotides. In some embodiments, 6 or fewer modified nucleotides of the first nucleotide sequence are 2’-fluoro nucleotides. In some embodiments, 5 or fewer modified nucleotides of the first nucleotide sequence are 2’-fluoro nucleotides. In some embodiments, 4 or fewer modified nucleotides of the first nucleotide sequence are 2’-fluoro nucleotides. In some embodiments, 3 or fewer modified nucleotides of the first nucleotide sequence are 2’-fluoro nucleotides. In some embodiments, 2 or fewer modified nucleotides of the first nucleotide sequence are 2’-fluoro nucleotides. In some embodiments, at least one modified nucleotide of the first nucleotide sequence is a 2’-fluoro pyrimidine. In some embodiments, 1, 2, 3, 4, 5, or 6 modified nucleotides of the first nucleotide sequence are 2’- fluoro pyrimidines. In some embodiments, at least one modified nucleotide of the first nucleotide sequence is a 2’-fluoro purine. In some embodiments, 1, 2, 3, 4, 5, or 6 modified nucleotides of the first nucleotide sequence are 2’-fluoro purines. In some embodiments, the 2’-fluoro nucleotide is a 2’-fluoro nucleotide mimic. In some embodiments, the nucleotide at position 3, 5, 7, 8, 9, 10, 11, 12, 14, 17, and/or 19 from the 5’ end of the first nucleotide sequence is a 2’-fluoro nucleotide. In some embodiments, at least two nucleotides at positions 3, 5, 7, 8, 9, 10, 11, 12, 14, 17, and/or 19 from the 5’ end of the first nucleotide sequence are 2’-fluoro nucleotides. In some embodiments, at least three nucleotides at positions 3, 5, 7, 8, 9, 10, 11, 12, 14, 17, and/or 19 from the 5’ end of the first nucleotide sequence are 2’-fluoro nucleotides. In some embodiments, at least four nucleotides at positions 3, 5, 7, 8, 9, 10, 11, 12, 14, 17, and/or 19 from the 5’ end of the first nucleotide sequence are 2’-fluoro nucleotides. In some embodiments, at least five nucleotides at positions 3, 5, 7, 8, 9, 10, 11, 12, 14, 17, and/or 19 from the 5’ end of the first nucleotide sequence are 2’-fluoro nucleotides. In some embodiments, the nucleotides at positions 3, 5, 7, 8, 9, 10, 11, 12, 14, 17, and/or 19 from the 5’ end of the first nucleotide sequence are 2’-fluoro nucleotides. In some embodiments, the nucleotide at position 3 from the 5’ end of the first nucleotide sequence is a 2’-fluoro nucleotide. In some embodiments, the nucleotide at position 7 from the 5’ end of the first nucleotide sequence is a 2’-fluoro nucleotide. In some embodiments, the nucleotide at position 8 from the 5’ end of the first nucleotide sequence is a 2’-fluoro nucleotide. In some embodiments, the nucleotide at position 9 from the 5’ end of the first nucleotide sequence is a 2’-fluoro nucleotide. In some embodiments, the nucleotide at position 12 from the 5’ end of the first nucleotide sequence is a 2’-fluoro nucleotide. In some embodiments, the nucleotide at position 17 from the 5’ end of the first nucleotide sequence is a 2’-fluoro nucleotide. In some embodiments, the 2’-fluoro nucleotide is a 2’-fluoro nucleotide mimic. In some embodiments, at least 1, 2, 3, 4, 5, 6, or 7 nucleotides at position 3, 5, 7, 8, 9, 10, 11, 12, 14, 17, and/or 19 from the 5’ end of the first nucleotide sequence is a 2’-fluoro nucleotide. In some embodiments, the nucleotide at positions 3, 5, 7, 8, 9, 10, 11, 12, 14, 17, and/or 19 from the 5’ end of the first nucleotide sequence is a 2’-fluoro nucleotide. In some embodiments, at least two nucleotides at positions 3, 5, 7, 8, 9, 10, 11, 12, 14, 17, and/or 19 from the 5’ end of the first nucleotide sequence are 2’-fluoro nucleotides. In some embodiments, at least three nucleotides at positions 3, 5, 7, 8, 9, 10, 11, 12, 14, 17, and/or 19 from the 5’ end of the first nucleotide sequence are 2’-fluoro nucleotides. In some embodiments, the nucleotides at positions 3, 5, 7, 8, 9, 10, 11, 12, 14, 17, and/or 19 from the 5’ end of the first nucleotide sequence are 2’-fluoro nucleotides. In some embodiments, the nucleotide at position 3 from the 5’ end of the first nucleotide sequence is a 2’-fluoro nucleotide. In some embodiments, the nucleotide at position 5 from the 5’ end of the first nucleotide sequence is a 2’-fluoro nucleotide. In some embodiments, the nucleotide at position 7 from the 5’ end of the first nucleotide sequence is a 2’-fluoro nucleotide. In some embodiments, the nucleotide at position 8 from the 5’ end of the first nucleotide sequence is a 2’-fluoro nucleotide. In some embodiments, the nucleotide at position 9 from the 5’ end of the first nucleotide sequence is a 2’-fluoro nucleotide. In some embodiments, the nucleotide at position 10 from the 5’ end of the first nucleotide sequence is a 2’-fluoro nucleotide. In some embodiments, the nucleotide at position 11 from the 5’ end of the first nucleotide sequence is a 2’-fluoro nucleotide. In some embodiments, the nucleotide at position 12 from the 5’ end of the first nucleotide sequence is a 2’-fluoro nucleotide. In some embodiments, the nucleotide at position 14 from the 5’ end of the first nucleotide sequence is a 2’-fluoro nucleotide. In some embodiments, the nucleotide at position 17 from the 5’ end of the first nucleotide sequence is a 2’-fluoro nucleotide. In some embodiments, the nucleotide at position 19 from the 5’ end of the first nucleotide sequence is a 2’-fluoro nucleotide. In some embodiments, the nucleotide at position 3, 7, 8, 9, 12, and/or 17 from the 5’ end of the first nucleotide sequence is a 2’-fluoro nucleotide. In some embodiments, the nucleotide at position 3, 7, 8, and/or 17 from the 5’ end of the first nucleotide sequence is a 2’-fluoro nucleotide. In some embodiments, the nucleotide at position 3, 7, 8, 9, 12, and/or 17 from the 5’ end of the first nucleotide sequence is a 2’-fluoro nucleotide. In some embodiments, the nucleotide at position 5, 7, 8, and/or 9 from the 5’ end of the first nucleotide sequence is a 2’- fluoro nucleotide. In some embodiments, the nucleotide at position 5, 9, 10, 11, 12, and/or 19 from the 5’ end of the first nucleotide sequence is a 2’-fluoro nucleotide. In some embodiments, the 2’-fluoro nucleotide is a 2’-fluoro nucleotide mimic. In some embodiments, the 2’-fluoro or 2’-O-methyl nucleotide mimic is a nucleotide mimic of Formula (V): , wherein Rx is independently a nucleobase, aryl,
Figure imgf000049_0001
heteroaryl, or H, Q1 and Q2 are independently S or O, R5 is independently –OCD3 , –F, or – OCH3, and R6 and R7 are independently H, D, or CD3. In some embodiments, the nucleobase is selected from thymine, cytosine, guanine, adenine, uracil, and an analogue or derivative thereof. In some embodiments, the 2’-fluoro or 2’-O-methyl nucleotide mimic is a nucleotide mimic of Formula (16) – Formula (20):
Figure imgf000050_0001
wherein Rx is independently a nucleobase, aryl, heteroaryl, or H and R2 is F or –OCH3. In some embodiments, the nucleobase is selected from thymine, cytosine, guanine, adenine, uracil, and an analogue or derivative thereof. In some embodiments, the sense strand may comprise 1, 2, 3, 4, or 5 or more modified nucleotide(s) selected from: wherein Rx is a nucleobase, aryl,
Figure imgf000050_0002
heteroaryl, or H,
Figure imgf000050_0003
(mun34), wherein Ry is a nucleobase,
Figure imgf000050_0004
Figure imgf000051_0001
( wherein * represent chiral center), and
Figure imgf000052_0002
(wherein * represent chiral center), wherein B is a nucleobase, aryl, heteroaryl, or H, and wherein
Figure imgf000052_0001
Figure imgf000052_0003
or represents a phosphodiester linkage, a phosphorothioate linkage, a mesyl phosphoramidate linkage, or H. In some embodiments, the nucleobase is selected from thymine, cytosine, guanine, adenine, uracil, and an analogue or derivative thereof. In some embodiments, the first nucleotide sequence comprises, consists of, or consists essentially of ribonucleic acids (RNAs). In some embodiments, the first nucleotide sequence comprises, consists of, or consists essentially of modified RNAs. In some embodiments, the modified RNAs are selected from a 2’-O-methyl RNA and 2’-fluoro RNA. In some embodiments, 15, 16, 17, 18, 19, 20, 21, 22, or 23 modified nucleotides of the first nucleotide sequence are independently selected from 2’-O-methyl RNA and 2’-fluoro RNA. In some embodiments, the sense strand may further comprise one or more internucleoside linkages independently selected from a phosphodiester (PO) internucleoside linkage, phosphorothioate (PS) internucleoside linkage, mesyl phosphoramidate internucleoside linkage (Ms), phosphorodithioate internucleoside linkage, and PS-mimic internucleoside linkage. In some embodiments, the PS-mimic internucleoside linkage is a sulfo internucleoside linkage. In some embodiments, the sense strand may further comprise at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, or 15 or more phosphorothioate internucleoside linkages. In some embodiments, the sense strand comprises 20, 19, 18, 17, 16, 15, 14, 13, 12, 11, 10, 9, 8, 7, 6, 5, 4, or 3 or fewer phosphorothioate internucleoside linkages. In some embodiments, the sense strand comprises 2 to 10, 2 to 8, 2 to 6, 1 to 5, 1 to 4, 1 to 3, or 1 to 2 phosphorothioate internucleoside linkages. In some embodiments, the sense strand comprises 1 to 2 phosphorothioate internucleoside linkages. In some embodiments, the sense strand comprises 2 to 4 phosphorothioate internucleoside linkages. In some embodiments, at least one phosphorothioate internucleoside linkage is between the nucleotides at positions 1 and 2 from the 5’ end of the first nucleotide sequence. In some embodiments, at least one phosphorothioate internucleoside linkage is between the nucleotides at positions 2 and 3 from the 5’ end of the first nucleotide sequence. In some embodiments, the sense strand comprises two phosphorothioate internucleoside linkages between the nucleotides at positions 1 to 3 from the 5’ end of the first nucleotide sequence. In some embodiments, the sense strand may further comprise at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, or 15 or more mesyl phosphoramidate internucleoside linkages. In some embodiments, the sense strand comprises 20, 19, 18, 17, 16, 15, 14, 13, 12, 11, 10, 9, 8, 7, 6, 5, 4, or 3 or fewer mesyl phosphoramidate internucleoside linkages. In some embodiments, the sense strand comprises 2 to 10, 2 to 8, 2 to 6, 1 to 5, 1 to 4, 1 to 3, or 1 to 2 mesyl phosphoramidate internucleoside linkages. In some embodiments, the sense strand comprises 1 to 2 mesyl phosphoramidate internucleoside linkages. In some embodiments, the sense strand comprises 2 to 4 mesyl phosphoramidate internucleoside linkages. In some embodiments, the sense strand may comprise any of the modified nucleotides disclosed in the sub-section titled “Modified Nucleotides” below. In some embodiments, the sense strand may comprise a 5’-stabilized end cap, and the 5’-stabilized end cap may be selected from those disclosed in the sub-section titled “5’-Stabilized End Cap” below. B. siNA antisense strand Any of the siNA molecules described herein may comprise an antisense strand. The antisense strand may comprise a second nucleotide sequence. The second nucleotide sequence may be 15 to 30, 15 to 25, 15 to 23, 17 to 23, 19 to 23, or 19 to 21 nucleotides in length. In some embodiments, the second nucleotide sequence is 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30 nucleotides in length. In some embodiments, the second nucleotide sequence is at least 19 nucleotides in length. In some embodiments, the second nucleotide sequence is at least 21 nucleotides in length. In some embodiments, the antisense strand is the same length as the second nucleotide sequence. In some embodiments, the antisense strand is longer than the second nucleotide sequence. In some embodiments, the antisense strand may further comprise 1, 2, 3, 4, or 5 or more nucleotides than the second nucleotide sequence. In some embodiments, the antisense strand is the same length as the sense strand. In some embodiments, the antisense strand is longer than the sense strand. In some embodiments, the antisense strand may further comprise 1, 2, 3, 4, or 5 or more nucleotides than the sense strand. In some embodiments, the antisense strand may further comprise a deoxyribonucleic acid (DNA). In some embodiments, the DNA is thymine (T). In some embodiments, the antisense strand may further comprise a TT sequence. In some embodiments, the antisense strand may further comprise one or more modified nucleotides that are adjacent to the second nucleotide sequence. In some embodiments, the one or more modified nucleotides are independently selected from any of the modified nucleotides disclosed herein (e.g., 2’-fluoro nucleotide, 2’- O-methyl nucleotide, 2’-fluoro nucleotide mimic, 2’-O-methyl nucleotide mimic, or a nucleotide comprising a modified nucleobase). In some embodiments, the second nucleotide sequence comprises 15, 16, 17, 18, 19, 20, 21, 22, 23, or more modified nucleotides independently selected from a 2’-O-methyl nucleotide and a 2’-fluoro nucleotide. In some embodiments, 70%, 75%, 80%, 85%, 90%, 95% or 100% of the nucleotides in the second nucleotide sequence are modified nucleotides independently selected from a 2’-O-methyl nucleotide and a 2’-fluoro nucleotide. In some embodiments, 100% of the nucleotides in the second nucleotide sequence are modified nucleotides independently selected from a 2’-O-methyl nucleotide and a 2’-fluoro nucleotide. In some embodiments, between about 15 to 30, 15 to 25, 15 to 24, 15 to 23, 15 to 22, 15 to 21, 17 to 30, 17 to 25, 17 to 24, 17 to 23, 17 to 22, 17 to 21, 18 to 30, 18 to 25, 18 to 24, 18 to 23, 18 to 22, 18 to 21, 19 to 30, 19 to 25, 19 to 24, 19 to 23, 19 to 22, 19 to 21, 20 to 25, 20 to 24, 20 to 23, 21 to 25, 21 to 24, or 21 to 23 modified nucleotides of the second nucleotide sequence are 2’-O-methyl nucleotides. In some embodiments, between about 2 to 20 modified nucleotides of the second nucleotide sequence are 2’-O-methyl nucleotides. In some embodiments, between about 5 to 25 modified nucleotides of the second nucleotide sequence are 2’-O-methyl nucleotides. In some embodiments, between about 10 to 25 modified nucleotides of the second nucleotide sequence are 2’-O-methyl nucleotides. In some embodiments, between about 12 to 25 modified nucleotides of the second nucleotide sequence are 2’-O-methyl nucleotides. In some embodiments, at least 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, or 22 modified nucleotides of the second nucleotide sequence are 2’-O- methyl nucleotides. In some embodiments, at least about 12 modified nucleotides of the second nucleotide sequence are 2’-O-methyl nucleotides. In some embodiments, at least about 13 modified nucleotides of the second nucleotide sequence are 2’-O-methyl nucleotides. In some embodiments, at least about 14 modified nucleotides of the second nucleotide sequence are 2’-O-methyl nucleotides. In some embodiments, at least about 15 modified nucleotides of the second nucleotide sequence are 2’-O-methyl nucleotides. In some embodiments, at least about 16 modified nucleotides of the second nucleotide sequence are 2’-O-methyl nucleotides. In some embodiments, at least about 17 modified nucleotides of the second nucleotide sequence are 2’-O-methyl nucleotides. In some embodiments, at least about 18 modified nucleotides of the second nucleotide sequence are 2’-O-methyl nucleotides. In some embodiments, at least about 19 modified nucleotides of the second nucleotide sequence are 2’-O-methyl nucleotides. In some embodiments, less than or equal to 25, 24, 23, 22, 21, 20, 19, 18, 17, 16, 15, 14, 13, 12, 11, 10, 9, 8, 7, 6, 5, 4, 3, or 2 modified nucleotides of the second nucleotide sequence are 2’-O-methyl nucleotides. In some embodiments, less than or equal to 21 modified nucleotides of the second nucleotide sequence are 2’-O-methyl nucleotides. In some embodiments, less than or equal to 20 modified nucleotides of the second nucleotide sequence are 2’-O-methyl nucleotides. In some embodiments, less than or equal to 19 modified nucleotides of the second nucleotide sequence are 2’-O-methyl nucleotides. In some embodiments, less than or equal to 18 modified nucleotides of the second nucleotide sequence are 2’-O-methyl nucleotides. In some embodiments, less than or equal to 17 modified nucleotides of the second nucleotide sequence are 2’-O-methyl nucleotides. In some embodiments, less than or equal to 16 modified nucleotides of the second nucleotide sequence are 2’-O-methyl nucleotides. In some embodiments, less than or equal to 15 modified nucleotides of the second nucleotide sequence are 2’-O-methyl nucleotides. In some embodiments, less than or equal to 14 modified nucleotides of the second nucleotide sequence are 2’-O-methyl nucleotides. In some embodiments, less than or equal to 13 modified nucleotides of the second nucleotide sequence are 2’-O-methyl nucleotides. In some embodiments, at least one modified nucleotide of the second nucleotide sequence is a 2’-O-methyl pyrimidine. In some embodiments, at least 5, 6, 7, 8, 9, or 10 modified nucleotides of the second nucleotide sequence are 2’-O-methyl pyrimidines. In some embodiments, at least one modified nucleotide of the second nucleotide sequence is a 2’-O-methyl purine. In some embodiments, at least 5, 6, 7, 8, 9, or 10 modified nucleotides of the second nucleotide sequence are 2’-O- methyl purines. In some embodiments, the 2’-O-methyl nucleotide is a 2’-O-methyl nucleotide mimic. In some embodiments, between 2 to 15 modified nucleotides of the second nucleotide sequence are 2’-fluoro nucleotides. In some embodiments, between 2 to 10 modified nucleotides of the second nucleotide sequence are 2’-fluoro nucleotides. In some embodiments, between 2 to 6 modified nucleotides of the second nucleotide sequence are 2’- fluoro nucleotides. In some embodiments, 1 to 6, 1 to 5, 1 to 4, or 1 to 3 modified nucleotides of the second nucleotide sequence are 2’-fluoro nucleotides. In some embodiments, at least 1, 2, 3, 4, 5, or 6 modified nucleotides of the second nucleotide sequence are 2’-fluoro nucleotides. In some embodiments, at least 1 modified nucleotide of the second nucleotide sequence is a 2’-fluoro nucleotide. In some embodiments, at least 2 modified nucleotides of the second nucleotide sequence are 2’-fluoro nucleotides. In some embodiments, at least 3 modified nucleotides of the second nucleotide sequence are 2’-fluoro nucleotides. In some embodiments, at least 4 modified nucleotides of the second nucleotide sequence are 2’-fluoro nucleotides. In some embodiments, at least 5 modified nucleotides of the second nucleotide sequence are 2’-fluoro nucleotides. In some embodiments, 10, 9, 8, 7, 6, 5, 4, 3 or fewer modified nucleotides of the second nucleotide sequence are 2’-fluoro nucleotides. In some embodiments, 10 or fewer modified nucleotides of the second nucleotide sequence are 2’- fluoro nucleotides. In some embodiments, 7 or fewer modified nucleotides of the second nucleotide sequence are 2’-fluoro nucleotides. In some embodiments, 6 or fewer modified nucleotides of the second nucleotide sequence are 2’-fluoro nucleotides. In some embodiments, 5 or fewer modified nucleotides of the second nucleotide sequence are 2’- fluoro nucleotides. In some embodiments, 4 or fewer modified nucleotides of the second nucleotide sequence are 2’-fluoro nucleotides. In some embodiments, 3 or fewer modified nucleotides of the second nucleotide sequence are 2’-fluoro nucleotides. In some embodiments, 2 or fewer modified nucleotides of the second nucleotide sequence are 2’- fluoro nucleotides. In some embodiments, at least one modified nucleotide of the second nucleotide sequence is a 2’-fluoro pyrimidine. In some embodiments, 1, 2, 3, 4, 5, or 6 modified nucleotides of the second nucleotide sequence are 2’-fluoro pyrimidines. In some embodiments, at least one modified nucleotide of the second nucleotide sequence is a 2’- fluoro purine. In some embodiments, 1, 2, 3, 4, 5, or 6 modified nucleotides of the second nucleotide sequence are 2’-fluoro purines. In some embodiments, the 2’-fluoro nucleotide is a 2’-fluoro nucleotide mimic. In some embodiments, the 2’-fluoro nucleotide or 2’-O-methyl nucleotide is a 2’- fluoro or 2’-O-methyl nucleotide mimic. In some embodiments, the 2’-fluoro or 2’-O-methyl nucleotide mimic is a nucleotide mimic of Formula (V):
Figure imgf000057_0001
, wherein Rx is independently a nucleobase, aryl, heteroaryl, or H, Q1 and Q2 are independently S or O, R5 is independently –OCD3 , –F, or –OCH3, and R6 and R7 are independently H, D, or CD3. In some embodiments, the nucleobase is selected from thymine, cytosine, guanine, adenine, uracil, and an analogue or derivative thereof. In some embodiments, the 2’-fluoro or 2’-O-methyl nucleotide mimic is a nucleotide mimic of Formula (16) – Formula (20):
Figure imgf000057_0002
wherein Rx is a nucleobase, aryl, heteroaryl, or H and R2 is independently F or -OCH3. In some embodiments, the nucleobase is selected from thymine, cytosine, guanine, adenine, uracil, and an analogue or derivative thereof. In some embodiments, the antisense strand may comprise 1, 2, 3, 4, or 5 or more modified nucleotide(s) selected from: , wherein Rx is a nucleobase, aryl,
Figure imgf000057_0003
Figure imgf000058_0001
Figure imgf000059_0001
( wherein * represent chiral center), and
Figure imgf000059_0002
(wherein * represent chiral center), wherein B is a nucleobase, aryl, heteroaryl, or H, and wherein represents a
Figure imgf000059_0003
phosphodiester linkage, a phosphorothioate linkage, a mesyl phosphoramidate linkage, or H. In some embodiments, the nucleobase is selected from thymine, cytosine, guanine, adenine, uracil, and an analogue or derivative thereof. In some embodiments, at least 1, 2, 3, 4, 5, 6, 7, 8, or 9 nucleotides at position 2, 5, 6, 8, 10, 14, 16, 17, and/or 18 from the 5’ end of the second nucleotide sequence is a 2’-fluoro nucleotide. In some embodiments, the nucleotide at position 2, 5, 6, 8, 10, 14, 16, 17, and/or 18 from the 5’ end of the second nucleotide sequence is a 2’-fluoro nucleotide. In some embodiments, at least two nucleotides at positions 2, 5, 6, 8, 10, 14, 16, 17, and/or 18 from the 5’ end of the second nucleotide sequence are 2’-fluoro nucleotides. In some embodiments, at least three nucleotides at positions 2, 5, 6, 8, 10, 14, 16, 17, and/or 18 from the 5’ end of the second nucleotide sequence are 2’-fluoro nucleotides. In some embodiments, at least four nucleotides at positions 2, 5, 6, 8, 10, 14, 16, 17, and/or 18 from the 5’ end of the second nucleotide sequence are 2’-fluoro nucleotides. In some embodiments, at least five nucleotides at positions 2, 5, 6, 8, 10, 14, 16, 17, and/or 18 from the 5’ end of the second nucleotide sequence are 2’-fluoro nucleotides. In some embodiments, the nucleotides at positions 2 and/or 14 from the 5’ end of the second nucleotide sequence are 2’-fluoro nucleotides. In some embodiments, the nucleotides at positions 2, 6, and/or 16 from the 5’ end of the second nucleotide sequence are 2’-fluoro nucleotides. In some embodiments, the nucleotides at positions 2, 6, 14, and/or 16 from the 5’ end of the second nucleotide sequence are 2’-fluoro nucleotides. In some embodiments, the nucleotides at positions 2, 6, 10, 14, and/or 18 from the 5’ end of the second nucleotide sequence are 2’-fluoro nucleotides. In some embodiments, the nucleotides at positions 2, 5, 8, 14, and/or 17 from the 5’ end of the second nucleotide sequence are 2’-fluoro nucleotides. In some embodiments, the nucleotide at position 2 from the 5’ end of the second nucleotide sequence is a 2’-fluoro nucleotide. In some embodiments, the nucleotide at position 5 from the 5’ end of the second nucleotide sequence is a 2’-fluoro nucleotide. In some embodiments, the nucleotide at position 6 from the 5’ end of the second nucleotide sequence is a 2’-fluoro nucleotide. In some embodiments, the nucleotide at position 8 from the 5’ end of the second nucleotide sequence is a 2’-fluoro nucleotide. In some embodiments, the nucleotide at position 10 from the 5’ end of the second nucleotide sequence is a 2’-fluoro nucleotide. In some embodiments, the nucleotide at position 14 from the 5’ end of the second nucleotide sequence is a 2’-fluoro nucleotide. In some embodiments, the nucleotide at position 16 from the 5’ end of the second nucleotide sequence is a 2’-fluoro nucleotide. In some embodiments, the nucleotide at position 17 from the 5’ end of the second nucleotide sequence is a 2’-fluoro nucleotide. In some embodiments, the nucleotide at position 18 from the 5’ end of the second nucleotide sequence is a 2’-fluoro nucleotide. In some embodiments, the 2’-fluoro nucleotide is a 2’-fluoro nucleotide mimic. In some embodiments, the nucleotides in the second nucleotide sequence are arranged in an alternating 1:3 modification pattern, wherein 1 nucleotide is a 2’-fluoro nucleotide and 3 nucleotides are 2’-O-methyl nucleotides, and wherein the alternating 1:3 modification pattern occurs at least 2 times. In some embodiments, the alternating 1:3 modification pattern occurs 2-5 times. In some embodiments, at least two of the alternating 1:3 modification pattern occur consecutively. In some embodiments, at least two of the alternating 1:3 modification pattern occurs nonconsecutively. In some embodiments, at least 1, 2, 3, 4, or 5 alternating 1:3 modification pattern begins at nucleotide position 2, 6, 10, 14, and/or 18 from the 5’ end of the antisense strand. In some embodiments, at least one alternating 1:3 modification pattern begins at nucleotide position 2 from the 5’ end of the antisense strand. In some embodiments, wherein at least one alternating 1:3 modification pattern begins at nucleotide position 6 from the 5’ end of the antisense strand. In some embodiments, at least one alternating 1:3 modification pattern begins at nucleotide position 10 from the 5’ end of the antisense strand. In some embodiments, at least one alternating 1:3 modification pattern begins at nucleotide position 14 from the 5’ end of the antisense strand. In some embodiments, at least one alternating 1:3 modification pattern begins at nucleotide position 18 from the 5’ end of the antisense strand. In some embodiments, the 2’-fluoro nucleotide is a 2’-fluoro nucleotide mimic. In some embodiments, the nucleotides in the second nucleotide sequence are arranged in an alternating 1:2 modification pattern, wherein 1 nucleotide is a 2’-fluoro nucleotide and 2 nucleotides are 2’-O-methyl nucleotides, and wherein the alternating 1:2 modification pattern occurs at least 2 times. In some embodiments, the alternating 1:2 modification pattern occurs 2-5 times. In some embodiments, at least two of the alternating 1:2 modification pattern occurs consecutively. In some embodiments, at least two of the alternating 1:2 modification pattern occurs nonconsecutively. In some embodiments, at least 1, 2, 3, 4, or 5 alternating 1:2 modification pattern begins at nucleotide position 2, 5, 8, 14, and/or 17 from the 5’ end of the antisense strand. In some embodiments, at least one alternating 1:2 modification pattern begins at nucleotide position 2 from the 5’ end of the antisense strand. In some embodiments, at least one alternating 1:2 modification pattern begins at nucleotide position 5 from the 5’ end of the antisense strand. In some embodiments, at least one alternating 1:2 modification pattern begins at nucleotide position 8 from the 5’ end of the antisense strand. In some embodiments, at least one alternating 1:2 modification pattern begins at nucleotide position 14 from the 5’ end of the antisense strand. In some embodiments, at least one alternating 1:2 modification pattern begins at nucleotide position 17 from the 5’ end of the antisense strand. In some embodiments, the 2’-fluoro nucleotide is a 2’-fluoro nucleotide mimic. In some embodiments, the second nucleotide sequence comprises, consists of, or consists essentially of ribonucleic acids (RNAs). In some embodiments, the second nucleotide sequence comprises, consists of, or consists essentially of modified RNAs. In some embodiments, the modified RNAs are selected from a 2’-O-methyl RNA and 2’-fluoro RNA. In some embodiments, 15, 16, 17, 18, 19, 20, 21, 22, or 23 modified nucleotides of the second nucleotide sequence are independently selected from 2’-O-methyl RNA and 2’-fluoro RNA. In some embodiments, the 2’-fluoro nucleotide is a 2’-fluoro nucleotide mimic. In some embodiments, the sense strand may further comprise one or more internucleoside linkages independently selected from a phosphodiester (PO) internucleoside linkage, phosphorothioate (PS) internucleoside linkage, phosphorodithioate internucleoside linkage, and PS-mimic internucleoside linkage. In some embodiments, the PS-mimic internucleoside linkage is a 3’- or 5’-sulfur linkage. In some embodiments, the antisense strand may further comprise at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, or 15 or more phosphorothioate internucleoside linkages. In some embodiments, the antisense strand comprises 20, 19, 18, 17, 16, 15, 14, 13, 12, 11, 10, 9, 8, 7, 6, 5, 4, or 3 or fewer phosphorothioate internucleoside linkages. In some embodiments, the antisense strand comprises 2 to 10, 2 to 8, 2 to 6, 1 to 5, 1 to 4, 1 to 3, or 1 to 2 phosphorothioate internucleoside linkages. In some embodiments, the antisense strand comprises 2 to 10, 2 to 8, 2 to 6, 1 to 5, 1 to 4, 1 to 3, or 1 to 2 phosphorothioate internucleoside linkages. In some embodiments, the antisense strand comprises 2 to 8 phosphorothioate internucleoside linkages. In some embodiments, the antisense strand comprises 3 to 8 phosphorothioate internucleoside linkages. In some embodiments, the antisense strand comprises 4 to 8 phosphorothioate internucleoside linkages. In some embodiments, at least one phosphorothioate internucleoside linkage is between the nucleotides at positions 1 and 2 from the 5’ end of the second nucleotide sequence. In some embodiments, at least one phosphorothioate internucleoside linkage is between the nucleotides at positions 2 and 3 from the 5’ end of the second nucleotide sequence. In some embodiments, at least one phosphorothioate internucleoside linkage is between the nucleotides at positions 1 and 2 from the 3’ end of the second nucleotide sequence. In some embodiments, at least one phosphorothioate internucleoside linkage is between the nucleotides at positions 2 and 3 from the 3’ end of the second nucleotide sequence. In some embodiments, the antisense strand comprises two phosphorothioate internucleoside linkages between the nucleotides at positions 1 to 3 from the 5’ end of the first nucleotide sequence. In some embodiments, the antisense strand comprises two phosphorothioate internucleoside linkages between the nucleotides at positions 1 to 3 from the 3’ end of the first nucleotide sequence. In some embodiments, the antisense strand comprises (a) two phosphorothioate internucleoside linkages between the nucleotides at positions 1 to 3 from the 5’ end of the first nucleotide sequence; and (b) two phosphorothioate internucleoside linkages between the nucleotides at positions 1 to 3 from the 3’ end of the first nucleotide sequence. In some embodiments, the antisense strand may further comprise at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, or 15 or more mesyl phosphoramidate internucleoside linkages. In some embodiments, the antisense strand comprises 20, 19, 18, 17, 16, 15, 14, 13, 12, 11, 10, 9, 8, 7, 6, 5, 4, or 3 or fewer mesyl phosphoramidate internucleoside linkages. In some embodiments, the antisense strand comprises 2 to 10, 2 to 8, 2 to 6, 1 to 5, 1 to 4, 1 to 3, or 1 to 2 mesyl phosphoramidate internucleoside linkages. In some embodiments, the antisense strand comprises 2 to 10, 2 to 8, 2 to 6, 1 to 5, 1 to 4, 1 to 3, or 1 to 2 mesyl phosphoramidate internucleoside linkages. In some embodiments, the antisense strand comprises 2 to 8 mesyl phosphoramidate internucleoside linkages. In some embodiments, the antisense strand comprises 3 to 8 mesyl phosphoramidate internucleoside linkages. In some embodiments, the antisense strand comprises 4 to 8 mesyl phosphoramidate internucleoside linkages. In some embodiments, at least one end of the ds-siNA is a blunt end. In some embodiments, at least one end of the ds-siNA comprises an overhang, wherein the overhang comprises at least one nucleotide. In some embodiments, both ends of the ds-siNA comprise an overhang, wherein the overhang comprises at least one nucleotide. In some embodiments, the overhang comprises 1 to 5 nucleotides, 1 to 4 nucleotides, 1 to 3 nucleotides, or 1 to 2 nucleotides. In some embodiments, the overhang consists of 1 to 2 nucleotides. In some embodiments, the antisense strand may comprise any of the modified nucleotides disclosed in the sub-section titled “Modified Nucleotides” below. In some embodiments, the antisense strand may comprise a 5’-stabilized end cap, and the 5’-stabilized end cap may be selected from those disclosed in the sub-section titled “5’-Stabilized End Cap” below. Modified Nucleotides The siNA molecules disclosed herein comprise one or more modified nucleotides. In some embodiments, the sense strands disclosed herein comprise one or more modified nucleotides. In some embodiments, any of the first nucleotide sequences disclosed herein comprise one or more modified nucleotides. In some embodiments, the antisense strands disclosed herein comprise one or more modified nucleotides. In some embodiments, any of the second nucleotide sequences disclosed herein comprise one or more modified nucleotides. In some embodiments, the one or more modified nucleotides is adjacent to the first nucleotide sequence. In some embodiments, at least one modified nucleotide is adjacent to the 5’ end of the first nucleotide sequence. In some embodiments, at least one modified nucleotide is adjacent to the 3’ end of the first nucleotide sequence. In some embodiments, at least one modified nucleotide is adjacent to the 5’ end of the first nucleotide sequence and at least one modified nucleotide is adjacent to the 3’ end of the first nucleotide sequence. In some embodiments, the one or more modified nucleotides is adjacent to the second nucleotide sequence. In some embodiments, at least one modified nucleotide is adjacent to the 5’ end of the second nucleotide sequence. In some embodiments, at least one modified nucleotide is adjacent to the 3’ end of the second nucleotide sequence. In some embodiments, at least one modified nucleotide is adjacent to the 5’ end of the second nucleotide sequence and at least one modified nucleotide is adjacent to the 3’ end of the second nucleotide sequence. In some embodiments, a 2’-O-methyl nucleotide in any of sense strands or first nucleotide sequences disclosed herein is replaced with a modified nucleotide. In some embodiments, a 2’-O-methyl nucleotide in any of antisense strands or second nucleotide sequences disclosed herein is replaced with a modified nucleotide. In some embodiments, any of the siNA molecules, siNAs, sense strands, first nucleotide sequences, antisense strands, and second nucleotide sequences disclosed herein comprise 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30 or more modified nucleotides. In some embodiments, 1%, 2%, 3%, 4%, 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 70%, 75%, 80%, 85%, 86%, 87%, 88%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% of the nucleotides in the siNA molecule, siNA, sense strand, first nucleotide sequence, antisense strand, or second nucleotide sequence are modified nucleotides. In some embodiments, a modified nucleotide is selected from the group consisting of 2’-fluoro nucleotide, 2’-O-methyl nucleotide, 2’-fluoro nucleotide mimic, 2’-O-methyl nucleotide mimic, a locked nucleic acid, an unlocked nucleic acid, and a nucleotide comprising a modified nucleobase. In some embodiments, the unlocked nucleic acid is a 2’,3’-unlocked nucleic acid. In some embodiments, the unlocked nucleic acid is a 3’,4’- unlocked nucleic acid (e.g., mun34) in which the furanose ring lacks a bond between the 3’ and 4; carbons. For example, the siNA of the present disclosure may comprise one or more modified nucleotide(s) selected from:
Figure imgf000065_0001
, wherein Rx is a nucleobase, aryl, heteroaryl, or H,
Figure imgf000065_0002
(mun34), wherein Ry is a nucleobase,
Figure imgf000065_0003
Figure imgf000065_0004
Figure imgf000066_0001
chiral center), and
Figure imgf000067_0001
(wherein * represent chiral center), wherein B is a nucleobase, aryl, heteroaryl, or H, and wherein or represents a phosphodiester linkage, a phosphorothioate linkage, a mesyl phosphoramidate linkage, or H. In some embodiments, both the sense strand and the antisense strand may each independently comprise 1, 2, 3, 4, or 5 or more of the foregoing modified nucleotides. In some embodiments, one or more (e.g., 1, 2, 3, 4, 5, or more) of the foregoing modified nucleotides may independently be present in the sense strand, first nucleotide sequence, antisense strand, and/or second nucleotide sequence of any of the siNA disclosed herein. In some embodiments, the nucleobase is selected from thymine, cytosine, guanine, adenine, uracil, and an analogue or derivative thereof. Additionally or alternatively, any of the siNAs disclosed herein may comprise other modified nucleotides, such as 2’-fluoro or 2’-O-methyl nucleotide mimics. For example, the disclosed siNA may comprise at least 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 or more 2’-fluoro or 2’-O- methyl nucleotide mimics. In some embodiments, any of the sense strands disclosed herein comprise at least 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 or more 2’-fluoro or 2’-O-methyl nucleotide mimics. In some embodiments, any of the first nucleotide sequences disclosed herein comprise at least 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 or more 2’-fluoro or 2’-O-methyl nucleotide mimics. In some embodiments, any of the antisense strands disclosed herein comprise at least 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 or more 2’-fluoro or 2’-O-methyl nucleotide mimics. In some embodiments, any of the second nucleotide sequences disclosed herein comprise at least 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 or more 2’-fluoro or 2’-O-methyl nucleotide mimics. In some embodiments, the 2’-fluoro or 2’-O-methyl nucleotide mimic is a nucleotide mimic of Formula (16) – Formula (20):
Figure imgf000067_0002
wherein Rx is a nucleobase, aryl, heteroaryl, or H and R2 is independently F or -OCH3. In some embodiments, the nucleobase is selected from thymine, cytosine, guanine, adenine, uracil, and an analogue or derivative thereof. In some embodiments, the siNA molecules disclosed herein comprise at least one 2’- fluoro nucleotide, at least one 2’-O-methyl nucleotide, and at least one 2’-fluoro or 2’-O- methyl nucleotide mimic. In some embodiments, the at least one 2’-fluoro or 2’-O-methyl nucleotide mimic is adjacent to the first nucleotide sequence. In some embodiments, the at least one 2’-fluoro or 2’-O-methyl nucleotide mimic is adjacent to the 5’ end of first nucleotide sequence. In some embodiments, the at least one 2’-fluoro or 2’-O-methyl nucleotide mimic is adjacent to the 3’ end of first nucleotide sequence. In some embodiments, the at least one 2’-fluoro or 2’-O-methyl nucleotide mimic is adjacent to the second nucleotide sequence. In some embodiments, the at least one 2’-fluoro or 2’-O-methyl nucleotide mimic is adjacent to the 5’ end of second nucleotide sequence. In some embodiments, the at least one 2’-fluoro or 2’-O-methyl nucleotide mimic is adjacent to the 3’ end of second nucleotide sequence. In some embodiments, the first nucleotide sequence does not comprise a 2’-fluoro nucleotide mimic. In some embodiments, the first nucleotide sequence does not comprise a 2’-O-methyl nucleotide mimic. In some embodiments, the second nucleotide sequence does not comprise a 2’-fluoro nucleotide mimic. In some embodiments, the second nucleotide sequence does not comprise a 2’-O-methyl nucleotide mimic. Conjugated Moiety Further disclosed herein are siNA molecules comprising a conjugated moiety. In some embodiments, the conjugated moiety is selected from galactosamine, peptides, proteins, sterols, lipids, phospholipids, biotin, phenoxazines, active drug substance, cholesterols, phenanthridine, anthraquinone, acridine, fluoresceins, rhodamines, coumarins, and dyes. In some embodiments, the conjugated moiety is attached to the 3’ end of the sense strand or first nucleotide sequence. In some embodiments, the conjugated moiety is attached to the 3’ end of the sense strand or first nucleotide sequence via 1, 2, 3, 4, or 5 or more linkers. In some embodiments, the conjugated moiety is attached to the 5’ end of the sense strand or first nucleotide sequence. In some embodiments, the conjugated moiety is attached to the 5’ end of the sense strand or first nucleotide sequence via 1, 2, 3, 4, or 5 or more linkers. In some embodiments, the conjugated moiety is attached to the 3’ end of the antisense strand or second nucleotide sequence. In some embodiments, the conjugated moiety is attached to the 3’ end of the antisense strand or second nucleotide sequence via 1, 2, 3, 4, or 5 or more linkers. In some embodiments, the conjugated moiety is attached to the 5’ end of the antisense strand or second nucleotide sequence. In some embodiments, the conjugated moiety is attached to the 5’ end of the antisense strand or second nucleotide sequence via 1, 2, 3, 4, or 5 or more linkers. In some embodiments, the one or more linkers are independently selected from the group consisting of a phosphodiester linker, phosphorothioate linker, phosphorodithioate linker, and mesyl phosphoramidate linker. In some embodiments, the conjugated moiety is galactosamine. In some embodiments, any of the siNAs disclosed herein are attached to a conjugated moiety that is galactosamine. In some embodiments, the galactosamine is N-acetylgalactosamine (GalNAc). In some embodiments, any of the siNA molecules disclosed herein comprise GalNAc. In some embodiments, the GalNAc is of Formula (VI):
Figure imgf000069_0001
wherein m is 1, 2, 3, 4, or 5; each n is independently 1 or 2; p is 0 or 1; each R is independently H or a first protecting group; each Y is independently selected from –O- P(=O)(SH) –, –O-P(=O)(O) –, –O-P(=O)(OH) –, –O-P(S)S–, and –O–; Z is H or a second protecting group; either L is a linker or L and Y in combination are a linker; and A is H, OH, a third protecting group, an activated group, or an oligonucleotide. In some embodiments, the first protecting group is acetyl. In some embodiments, the second protecting group is trimethoxytrityl (TMT). In some embodiments, the activated group is a phosphoramidite group. In some embodiments, the phosphoramidite group is a cyanoethoxy N,N- diisopropylphosphoramidite group. In some embodiments, the linker is a C6-NH2 group. In some embodiments, A is a short interfering nucleic acid (siNA) or siNA molecule. In some embodiments, m is 3. In some embodiments, R is H, Z is H, and n is 1. In some embodiments, R is H, Z is H, and n is 2. In some embodiments, the GalNAc is Formula (VII):
Figure imgf000070_0001
wherein Rz is OH or SH; and each n is independently 1 or 2. In some embodiments, the targeting ligand may be a GalNAc targeting ligand may comprise 1, 2, 3, 4, 5 or 6 GalNAc units. In some embodiments, the targeting ligand may be a GalNAc selected from GalNAc2, GalNAc3, GalNAc4 (the GalNAc of Formula VII, wherein n=1 and Rz=OH), GalNAc5, and GalNAc6. In some embodiments, the GalNAc may be GalNAc amidite (i.e., compound 40-9, see Example 22), GalNAc 4 CPG GalNAc phophoramidite, or GalNAc4-ps-GalNAc4-ps- GalNAc4. These GalNAc moieties are shown below:
Figure imgf000070_0002
Figure imgf000071_0001
GalNAc3, GalNAc4, GalNAc5 and GalNAc6 may be conjugated to an siNA disclosed herein during synthesis with 12, or 3 moieties. Further GalNAc moieties, such as GalNAc1 and GalNAc2, can be used to form 5’ and 3’-GalNAc using post synthesis conjugation. GalNAc Phosphoramidites
Figure imgf000071_0002
Figure imgf000072_0001
In some embodiments, the galactosamine is attached to the 3’ end of the sense strand or first nucleotide sequence. In some embodiments, the galactosamine is attached to the 3’ end of the sense strand or first nucleotide sequence via 1, 2, 3, 4, or 5 or more linkers. In some embodiments, the galactosamine is attached to the 5’ end of the sense strand or first nucleotide sequence. In some embodiments, the galactosamine is attached to the 5’ end of the sense strand or first nucleotide sequence via 1, 2, 3, 4, or 5 or more linkers. In some embodiments, the galactosamine is attached to the 3’ end of the antisense strand or second nucleotide sequence. In some embodiments, the galactosamine is attached to the 3’ end of the antisense strand or second nucleotide sequence via 1, 2, 3, 4, or 5 or more linkers. In some embodiments, the galactosamine is attached to the 5’ end of the antisense strand or second nucleotide sequence. In some embodiments, the galactosamine is attached to the 5’ end of the antisense strand or second nucleotide sequence via 1, 2, 3, 4, or 5 or more linkers. In some embodiments, the one or more linkers are independently selected from the group consisting of a phosphodiester (p or po) linker, phosphorothioate (ps) linker, mesyl phosphoramidate linker (Ms), phosphoramidite (HEG) linker, triethylene glycol (TEG) linker, and/or phosphorodithioate linker. In some embodiments, the one or more linkers are independently selected from the group consisting of p-(PS)2, (PS)2-p-TEG-p, (PS)2-p-HEG-p, and (PS)2-p- (HEG-p)2. In some embodiments, the conjugated moiety is a lipid moiety. In some embodiments, any of the siNAs disclosed herein are attached to a conjugated moiety that is a lipid moiety. Examples of lipid moieties include, but are not limited to, a cholesterol moiety, a thioether, e.g., hexyl-S-tritylthiol, a thiocholesterol, an aliphatic chain, e.g., dodecandiol or undecyl residues a phospholipid, e.g., di-hexadecyl-rac-glycerol or triethylammonium 1-di-O- hexadecyl-rac-glycero-S-H-phosphonate, a polyamine or a polyethylene glycol chain, adamantane acetic acid, a palmityl moiety, or an octadecylamine or hexylamino-carbonyl- oxycholesterol moiety. In some embodiments, the conjugated moiety is an active drug substance. In some embodiments, any of the siNAs disclosed herein are attached to a conjugated moiety that is an active drug substance. Examples of active drug substances include, but are not limited to, aspirin, warfarin, phenylbutazone, ibuprofen, suprofen, fenbufen, ketoprofen, (5)-(+)- pranoprofen, carprofen, dansylsarcosine, 2,3,5-triiodobenzoic acid, flufenamic acid, folinic acid, a benzothiadiazide, chlorothiazide, a diazepine, indomethicin, a barbiturate, a cephalosporin, a sulfa drug, an antidiabetic, an antibacterial or an antibiotic. Phosphorylation blocker Any of the disclosed siNA molecules may comprise a phosphorylation blocker. In some embodiments, a 2’-O-methyl nucleotide in any of sense strands or first nucleotide sequences disclosed herein is replaced with a nucleotide containing a phosphorylation blocker. In some embodiments, a 2’-O-methyl nucleotide in any of antisense strands or second nucleotide sequences disclosed herein is replaced with a nucleotide containing a phosphorylation blocker. In some embodiments, a 2’-O-methyl nucleotide in any of sense strands or first nucleotide sequences disclosed herein is further modified to contain a phosphorylation blocker. In some embodiments, a 2’-O-methyl nucleotide in any of antisense strands or second nucleotide sequences disclosed herein is further modified to contain a phosphorylation blocker. In some embodiments, any of the siNA molecules disclosed herein comprise a phosphorylation blocker of Formula (IV): , wherein Ry 4
Figure imgf000074_0001
is a nucleobase, R is – O-R30 or –NR31R32, R30 is C1-C8 substituted or unsubstituted alkyl; and R31 and R32 together with the nitrogen to which they are attached form a substituted or unsubstituted heterocyclic ring. In some embodiments, the nucleobase is selected from thymine, cytosine, guanine, adenine, uracil, and an analogue or derivative thereof. In some embodiments, any of the siNA molecules disclosed herein comprise a phosphorylation blocker of Formula (IV):
Figure imgf000074_0002
Formula (IV), wherein Ry is a nucleobase, and R4 is –OCH3 or –N(CH2CH2)2O. In some embodiments, the nucleobase is selected from thymine, cytosine, guanine, adenine, uracil, and an analogue or derivative thereof. In some embodiments, a siNA molecule comprises (a) a phosphorylation blocker of Formula (IV): wherein Ry
Figure imgf000075_0001
is a nucleobase, R4 is –O-R30 or –NR31R32, R30 is C1-C8 substituted or unsubstituted alkyl; and R31 and R32 together with the nitrogen to which they are attached form a substituted or unsubstituted heterocyclic ring; and (b) a short interfering nucleic acid (siNA), wherein the phosphorylation blocker is conjugated to the siNA. In some embodiments, the nucleobase is selected from thymine, cytosine, guanine, adenine, uracil, and an analogue or derivative thereof. In some embodiments, a siNA molecule comprises (a) a phosphorylation blocker of Formula (IV): Formula (IV), wherein Ry is a nucleobase, and R4 is –OCH3 or
Figure imgf000075_0002
–N(CH2CH2)2O; and (b) a short interfering nucleic acid (siNA), wherein the phosphorylation blocker is conjugated to the siNA. In some embodiments, the phosphorylation blocker is attached to the 3’ end of the sense strand or first nucleotide sequence. In some embodiments, the phosphorylation blocker is attached to the 3’ end of the sense strand or first nucleotide sequence via 1, 2, 3, 4, or 5 or more linkers. In some embodiments, the phosphorylation blocker is attached to the 5’ end of the sense strand or first nucleotide sequence. In some embodiments, the phosphorylation blocker is attached to the 5’ end of the sense strand or first nucleotide sequence via 1, 2, 3, 4, or 5 or more linkers. In some embodiments, the phosphorylation blocker is attached to the 3’ end of the antisense strand or second nucleotide sequence. In some embodiments, the phosphorylation blocker is attached to the 3’ end of the antisense strand or second nucleotide sequence via 1, 2, 3, 4, or 5 or more linkers. In some embodiments, the phosphorylation blocker is attached to the 5’ end of the antisense strand or second nucleotide sequence. In some embodiments, the phosphorylation blocker is attached to the 5’ end of the antisense strand or second nucleotide sequence via 1, 2, 3, 4, or 5 or more linkers. In some embodiments, the one or more linkers are independently selected from the group consisting of a phosphodiester linker, phosphorothioate linker, mesyl phosphoramidate linker and phosphorodithioate linker. 5’-Stabilized End Cap Further disclosed herein are siNA molecules comprising a 5’-stabilized end cap. As used herein the terms “5’-stabilized end cap” and “5’ end cap” are used interchangeably. In some embodiments, a 2’-O-methyl nucleotide in any of sense strands or first nucleotide sequences disclosed herein is replaced with a nucleotide containing a 5’-stabilized end cap. In some embodiments, a 2’-O-methyl nucleotide in any of antisense strands or second nucleotide sequences disclosed herein is replaced with a nucleotide containing a 5’-stabilized end cap. In some embodiments, a 2’-O-methyl nucleotide in any of sense strands or first nucleotide sequences disclosed herein is further modified to contain a 5’-stabilized end cap. In some embodiments, a 2’-O-methyl nucleotide in any of antisense strands or second nucleotide sequences disclosed herein is further modified to contain a 5’-stabilized end cap. In some embodiments, the 5’-stabilized end cap is a 5’ phosphate mimic. In some embodiments, the 5’-stabilized end cap is a modified 5’ phosphate mimic. In some embodiments, the modified 5’ phosphate is a chemically modified 5’ phosphate. In some embodiments, the 5’-stabilized end cap is a 5’-vinyl phosphonate. In some embodiments, the 5’-vinyl phosphonate is a 5’-(E)-vinyl phosphonate or 5’-(Z)-vinyl phosphonate. In some embodiments, the 5’-vinyl phosphonate is a deuterated vinyl phosphonate. In some embodiments, the deuterated vinyl phosphonate is a mono-deuterated vinyl phosphonate. In some embodiments, the deuterated vinyl phosphonate is a di-deuterated vinyl phosphonate. In some embodiments, the 5’-stabilized end cap is a phosphate mimic. Examples of phosphate mimics are disclosed in Parmar et al., J Med Chem, 201861(3):734-744, International Publication Nos. WO2018/045317 and WO2018/044350, and U.S. Patent No. 10,087,210, each of which is incorporated by reference in its entirety. In some embodiments, the 5’-stabilized end cap is not a phosphate mimic, per se, but rather includes a novel modified base. Examples of such 5’-stabilized end caps include, but are not limited to
Figure imgf000077_0001
(vm, wherein B is a nucleobase or derivative thereof), a
Figure imgf000077_0002
a
Figure imgf000077_0003
(vm(56amim)U, wherein R is H or Bz). In some embodiments, any of the siNA molecules disclosed herein may comprise a 5’ vinyl phosphonate moiety comprising a structure of:
Figure imgf000077_0004
wherein B is a nucleobase, aryl, heteroaryl, or H; wherein
Figure imgf000077_0005
represents a phosphodiester linkage, a phosphorothioate linkage, or a mesyl phosphoramidate linkage. In some embodiments, the disclosed siNA may comprise a nucleotide phosphate mimic selected from:
Figure imgf000078_0001
munb*enantiomer2); wherein Ry is a nucleobase and R15 is H or CH3. In some embodiments, the nucleobase is selected from thymine, cytosine, guanine, adenine, uracil, and an analogue or derivative thereof. In some embodiments, the nucleobase is uracil. In some embodiments, the nucleobase is adenine. In some embodiments, the nucleobase is guanine. In some embodiments, the nucleobase is cytosine. In some embodiments, the nucleobase is thymine. For the purposes of the omeco-d3 nucleotide, 4h nucleotide, v-mun nucleotide, c2o-4h nucleotide, omeco-munb nucleotide, and d2vm nucleotide, uridine can be a particularly suitable nucleobase. In some embodiments, one of these nucleotide phosphate mimics (e.g., omeco-d3 nucleotide, 4h nucleotide, v-mun nucleotide, c2o-4h nucleotide, omeco-munb nucleotide, or d2vm nucleotide) are located at the 5’ end of the antisense strand; however, these nucleotide phosphate mimics may also be incorporated at the 5’ end of the sense strand, the 3’ end of the antisense strand, or the 3’ end of the sense strand. Additionally or alternatively, the siNA molecules disclosed herein may comprise in the sense strand, the antisense strand, or both a 5’-stabilized end cap of Formula (Ia): , wherein R is H, a nucleobas 26
Figure imgf000079_0001
x e, aryl, or heteroaryl; R is
Figure imgf000079_0002
O
Figure imgf000079_0003
alkenylene)-Z and R20 is H; or R26 and R20 together form a 3- to 7-membered carbocyclic ring substituted with –(CR21R22)n-Z or –(C2-C6 alkenylene)-Z; n is 1, 2, 3, or 4; Z is –ONR23R24, – OP(O)OH(CH2)mCO2R23, –OP(S)OH(CH2)mCO2R23, –P(O)(OH)2, -P(O)(OH)(OCH3), - P(O)(OH)(OCD3), –SO2(CH2)mP(O)(OH)2, –SO2NR23R25, –NR23R24, –NR23SO2R24; either R21 and R22 are independently hydrogen or C1-C6 alkyl, or R21 and R22 together form an oxo group; R23 is hydrogen or C1-C6 alkyl; R24 is –SO2R25 or –C(O)R25; or R23 and R24 together with the nitrogen to which they are attached form a substituted or unsubstituted heterocyclic ring; R25 is C1-C6 alkyl; and m is 1, 2, 3, or 4. In some embodiments, R1 is an aryl. In some embodiments, the aryl is a phenyl ring. Additionally or alternatively, the siNA molecules disclosed herein may comprise in the sense strand, the antisense strand, or both a 5’-stabilized end cap of Formula (Ib):
Figure imgf000079_0004
Figure imgf000080_0001
–CH=CD-Z, –CD=CH-Z, –CD=CD-Z, –(CR21R22)n-Z, or –(C2-C6 alkenylene)-Z and R20 is H; or R26 and R20 together form a 3- to 7-membered carbocyclic ring substituted with –(CR21R22)n-Z or –(C2-C6 alkenylene)-Z; n is 1, 2, 3, or 4; Z is –ONR23R24, – OP(O)OH(CH2)mCO2R23, –OP(S)OH(CH2)mCO2R23, –P(O)(OH)2, -P(O)(OH)(OCH3), - P(O)(OH)(OCD3), –SO2(CH2)mP(O)(OH)2, –SO2NR23R25, –NR23R24, –NR23SO2R24; either R21 and R22 are independently hydrogen or C1-C6 alkyl, or R21 and R22 together form an oxo group; R23 is hydrogen or C1-C6 alkyl; R24 is –SO2R25 or –C(O)R25; or R23 and R24 together with the nitrogen to which they are attached form a substituted or unsubstituted heterocyclic ring; R25 is C1-C6 alkyl; and m is 1, 2, 3, or 4. In some embodiments, R1 is an aryl. In some embodiments, the aryl is a phenyl ring. Additionally or alternatively, the siNA molecules disclosed herein may comprise in the sense strand, the antisense strand, or both a 5’-stabilized end cap of Formula (Ic): , wherein Rx is a nucleobase, aryl, heteroaryl, or H,
Figure imgf000080_0002
Figure imgf000080_0003
–CH=CD-Z, –CD=CH 21 22
Figure imgf000080_0004
-Z, –CD=CD-Z, –(CR R )n-Z, or – (C2-C6 alkenylene)-Z and R20 is hydrogen; or R26 and R20 together form a 3- to 7-membered carbocyclic ring substituted with –(CR21R22)n-Z or –(C2-C6 alkenylene)-Z; n is 1, 2, 3, or 4; Z is –ONR23R24, –OP(O)OH(CH2)mCO2R23, –OP(S)OH(CH2)mCO2R23, –P(O)(OH)2, - P(O)(OH)(OCH3), -P(O)(OH)(OCD3), –SO2(CH2)mP(O)(OH)2, –SO2NR23R25, –NR23R24, or – NR23SO2R24; R21 and R22 either are independently hydrogen or C1-C6 alkyl, or R21 and R22 together form an oxo group; R23 is hydrogen or C1-C6 alkyl; R24 is –SO2R25 or –C(O)R25; or R23 and R24 together with the nitrogen to which they are attached form a substituted or unsubstituted heterocyclic ring; R25 is C1-C6 alkyl; and m is 1, 2, 3, or 4. In some embodiments, R1 is an aryl. In some embodiments, the aryl is a phenyl. Additionally or alternatively, the siNA molecules disclosed herein may comprise in the sense strand, the antisense strand, or both a 5’-stabilized end cap of Formula (IIa): wherein R is a nucleobase, aryl, h 26
Figure imgf000081_0001
x eteroaryl, or H, R is
Figure imgf000081_0002
Figure imgf000081_0003
–CH SO NH 9
Figure imgf000081_0004
2 2 CH3, or
Figure imgf000081_0005
R is –SO2CH3 or –COCH3,
Figure imgf000081_0006
is a double or single bond, R10 = –CH2PO3H or –NHCH3, R11 is –CH2– or –CO–, and R12 is H and R13 is CH3 or R12 and R13 together form –CH2CH2CH2–. In some embodiments, R1 is an aryl. In some embodiments, the aryl is a phenyl ring. Additionally or alternatively, the siNA molecules disclosed herein may comprise in the sense strand, the antisense strand, or both a 5’-stabilized end cap of Formula (IIb): , wherein R is a nucleobase, aryl, heteroaryl, or H 26
Figure imgf000081_0007
x , R is
Figure imgf000081_0008
,
Figure imgf000081_0009
Figure imgf000081_0010
–CH2SO2NHCH3, or
Figure imgf000081_0011
R9 is –SO2CH3 or –COCH3,
Figure imgf000081_0012
is a double or single bond, R10 = –CH2PO3H or –NHCH3, R11 is –CH2– or –CO–, and R12 is H and R13 is CH3 or R12 and R13 together form –CH2CH2CH2–. In some embodiments, R1 is an aryl. In some embodiments, the aryl is a phenyl ring. Additionally or alternatively, the siNA molecules disclosed herein may comprise in the sense strand, the antisense strand, or both a 5’-stabilized end cap of Formula (III): wherein Rx is a nucleobase, aryl, heteroaryl, or H, L is –CH2–, –CH=CH–,
Figure imgf000082_0001
–CO–, or –CH2CH2–, and A is –ONHCOCH3, –ONHSO2CH3, –PO3H, –OP(SOH)CH2CO2H, –SO2CH2PO3H, –SO2NHCH3, –NHSO2CH3, or –N(SO2CH2CH2CH2). In some embodiments, R1 is an aryl. In some embodiments, the aryl is a phenyl ring. Additionally or alternatively, the siNA molecules disclosed herein may comprise a 5’- stabilized end cap selected from the group consisting of Formula (1) to Formula (16), Formula (9X) to Formula (12X), Formula (16X), Formula (9Y) to Formula (12Y), Formula (16Y), Formula (21) to Formula (36), Formula 36X, Formula (41) to (56), Formula (49X) to (52X), Formula (49Y) to (52Y), Formula 56X, Formula 56Y, Formula (61) and Formula (62):
Figure imgf000082_0002
Figure imgf000083_0001
Figure imgf000084_0001
Figure imgf000085_0001
Figure imgf000086_0001
Figure imgf000086_0002
, wherein Rx is a nucleobase, aryl, heteroaryl, or H. In some embodiments, any of the siNA molecules disclosed herein comprise a 5’- stabilized end cap selected from the group consisting of Formula (50), Formula (50X), Formula (50Y), Formula (56), Formula (56X), Formula (56Y), Formula (61), and Formula (62):
Figure imgf000086_0003
Figure imgf000087_0001
Figure imgf000087_0002
wherein Rx is a nucleobase, aryl, heteroaryl, or H. In some embodiments, any of the siNA molecules disclosed herein comprise a 5’- stabilized end cap selected from the group consisting of Formula (71) to Formula (86), Formula (79X) to Formula (82X), Formula (79Y) to (82Y), Formula 86X, Formula 86X’, Formula 86Y, and Formula 86Y’:
Figure imgf000087_0003
Figure imgf000088_0001
Figure imgf000089_0001
wherein Rx is a nucleobase, aryl, heteroaryl, or H. In some embodiments, any of the siNA molecules disclosed herein comprise a 5’- stabilized end cap selected from the group consisting of Formula (78), Formula (79), Formula (79X), Formula (79Y), Formula (86), Formula (86X), and Formula (86X’):
Figure imgf000089_0002
Figure imgf000089_0003
, wherein Rx is a nucleobase, aryl, heteroaryl, or H. In some embodiments, any of the siNA molecules disclosed herein comprise a 5’- stabilized end cap selected from the group consisting of Formulas (1A)-(15A), Formulas (1A- 1)-(7A-1), Formulas (1A-2)-(7A-2), Formulas (1A-3)-(7A-3), Formulas (1A-4)-(7A-4), Formulas (9B)-(12B), Formulas (9AX)-(12AX), Formulas (9AY)-(12AY), Formulas (9BX)- (12BX), and Formulas (9BY)-(12BY):
Figure imgf000090_0001
Figure imgf000091_0001
Figure imgf000092_0001
Figure imgf000093_0001
In some embodiments, any of the siNA molecules disclosed herein comprise a 5’- stabilized end cap selected from the group consisting of Formulas (21A)-(35A), Formulas (29B)-(32B), Formulas (29AX)-(32AX), Formulas (29AY)-(32AY), Formulas (29BX)- (32BX), and Formulas (29BY)-(32BY):
Figure imgf000094_0001
Figure imgf000095_0001
Figure imgf000096_0001
In some embodiments, any of the siNA molecules disclosed herein comprise a 5’- stabilized end cap selected from the group consisting of Formulas (71A)-(86A), Formulas (79XA)-(82XA), Formulas (79YA)-(82YA); Formula (86XA), Formula (86X’A), Formula (86Y), and Formula (86Y’):
Figure imgf000096_0002
Figure imgf000097_0001
Figure imgf000098_0001
In some embodiments, any of the siNA molecules disclosed herein comprise a 5’- stabilized end cap selected from the group consisting of Formula (78A), Formula (79A), Formula (79XA), Formula (79YA), Formula (86A), Formula (86XA), and Formula (86X’A):
Figure imgf000098_0002
Figure imgf000099_0001
In some embodiments, any of the siNA molecules disclosed herein may comprise a 5’stabilized end cap with the structure:
Figure imgf000099_0002
wherein Ry is a nucleobase. In some embodiments, any of the siNA molecules disclosed herein may comprise a 5’stabilized end cap with the structure:
Figure imgf000099_0003
wherein Ry is a nucleobase and R15 is H or CH3. In some embodiments, the 5’-stabilized end cap is attached to the 5’ end of the antisense strand. In some embodiments, the 5’-stabilized end cap is attached to the 5’ end of the antisense strand via 1, 2, 3, 4, or 5 or more linkers. In some embodiments, the one or more linkers are independently selected from the group consisting of a phosphodiester (p or po) linker, phosphorothioate (ps) linker, mesyl phosphoramidate (Ms) linker, phosphoramidite (HEG) linker, triethylene glycol (TEG) linker, and/or phosphorodithioate linker. In some embodiments, the one or more linkers are independently selected from the group consisting of p-(PS)2, (PS)2-p-TEG-p, (PS)2-p-HEG-p, and (PS)2-p-(HEG-p)2. As indicated above, the present disclosure provides compositions comprising any of the siNA molecules, sense strands, antisense strands, first nucleotide sequences, or second nucleotide sequences described herein. The disclosed siNA and compositions thereof can be used in the treatment of various diseases and conditions associated with ANGPTL3 and/or ANGPTL8. Internucleoside Linkages In some embodiments, any of the siNAs, sense strands, first nucleotide sequences, antisense strands, and/or second nucleotide sequences disclosed herein comprise 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23 or more internucleoside linkages. In some embodiments, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 or more internucleoside linkages are independently selected from the group consisting of a phosphodiester (p or po), phosphorothioate (ps), mesyl phosphoramidate (Ms), or phosphorodithioate. In some embodiments, any of the siNAs, sense strands, first nucleotide sequences, antisense strands, and/or second nucleotide sequences disclosed herein further comprise 1, 2, 3, 4 or more linkages or other linkers that attach a conjugated moiety, phosphorylation blocker, and/or 5’ end cap to the siNA, sense strand, first nucleotide sequence, antisense strand, and/or second nucleotide sequences. In some embodiments, the 1, 2, 3, 4 or more linkers are independently selected from the group consisting of a phosphodiester (p or po) linker, phosphorothioate (ps) linker, mesyl phosphoramidate (Ms), phosphoramidite (HEG) linker, triethylene glycol (TEG) linker, and/or phosphorodithioate linker. In some embodiments, the one or more linkers are independently selected from the group consisting of p-(PS)2, (PS)2-p-TEG-p, (PS)2-p-HEG-p, and (PS)2-p-(HEG-p)2. Examples of siNA Targeting ANGPTL3 or ANGPTL8 The tables below provide examples of siNA that target ANGPTL3 or ANGPTL8. Table 1 - ANGPTL8 siNA SS and AS Unmodified Sequences (5’ to 3’)
Figure imgf000100_0001
Figure imgf000101_0001
Figure imgf000102_0001
Figure imgf000103_0001
Figure imgf000104_0001
Figure imgf000105_0001
Table 2 - ANGPTL8 siNA SS and AS Sequences with Nucleotide Modifications
Figure imgf000105_0002
Figure imgf000106_0001
Figure imgf000107_0001
Figure imgf000108_0001
Figure imgf000109_0001
Figure imgf000110_0001
Figure imgf000111_0001
Table 3 – ANGPTL3 siNA SS and AS Unmodified Sequences (5’ to 3’)
Figure imgf000111_0002
Figure imgf000112_0001
Figure imgf000113_0001
Figure imgf000114_0001
Figure imgf000115_0001
Table 4 – ANGPTL3 siNA SS and AS Sequences with Modified Nucleotides
Figure imgf000115_0002
Figure imgf000116_0001
Figure imgf000117_0001
Figure imgf000118_0001
Figure imgf000119_0001
Figure imgf000120_0001
Figure imgf000121_0001
Pharmaceutical Compositions The present disclosure also encompasses pharmaceutical compositions comprising siNAs of the present disclosure. In some embodiments is a pharmaceutical composition comprising one or more siNA of the present disclosure, and a pharmaceutically acceptable diluent or carrier. The disclosed siNA can be formulated in a pharmaceutical composition to individually target or inhibit or reduce expression of ANGPTL3 or ANGPTL8, or the disclosed siNA can be formulated in a pharmaceutical composition to simultaneously target or inhibit or reduce expression of both ANGPTL3 and ANGPTL8. Accordingly, a pharmaceutical composition may comprise at least one siNA comprising a sense strand comprising any one of SEQ ID NOs: 1-316 and an antisense strand comprising any one of SEQ ID NOs: 317-632. A pharmaceutical composition may comprise at least one siNA comprising a sense strand comprising any one of SEQ ID NOs: 633-878 and an antisense strand comprising any one of SEQ ID NOs: 879-1125. In instances in which the goal is to simultaneously target or inhibit or reduce expression of both ANGPTL3 and ANGPTL8, a pharmaceutical composition may comprise at least one siNA comprising a sense strand comprising any one of SEQ ID NOs: 1-316 and an antisense strand comprising any one of SEQ ID NOs: 317-632 and at least one siNA comprising a sense strand comprising any one of SEQ ID NOs: 633-878 and an antisense strand comprising any one of SEQ ID NOs: 879-1125. Alternatively, in instances in which the goal is to simultaneously target or inhibit or reduce expression of both ANGPTL3 and ANGPTL8, two separate pharmaceutical compositions may be administered to a subject concurrently or sequentially, wherein one pharmaceutical composition comprises at least one siNA comprising a sense strand comprising any one of SEQ ID NOs: 1-316 and an antisense strand comprising any one of SEQ ID NOs: 317-632 and the other pharmaceutical composition comprises at least one siNA comprising a sense strand comprising any one of SEQ ID NOs: 633-878 and an antisense strand comprising any one of SEQ ID NOs: 879-1125. In some embodiments, the pharmaceutical compositions comprising any of the siNA molecules, sense strands, antisense strands, first nucleotide sequences, or second nucleotide sequences described herein. The compositions may comprise 1, 2, 3, or more siNA molecules described herein. In some embodiments, the composition comprises a double-stranded short interfering nucleic acid (ds-siNA) molecule comprises (a) a sense strand comprising a first nucleotide sequence, wherein the first nucleotide sequence is 15 to 30 nucleotides in length and comprises a nucleotide sequence as set forth in SEQ ID NOs: 1-316; and (b) an antisense strand comprising a second nucleotide sequence, wherein the second nucleotide sequence is 15 to 30 nucleotides in length and comprises a nucleotide sequence that is the reverse complement of the first nucleotide sequence. In some embodiments, the first nucleotide sequence comprises a nucleotide sequence as set forth in SEQ ID NOs:1-316; and (b) an antisense strand that is the reverse complement of the first nucleotide sequence, wherein the antisense strand comprises a second nucleotide sequence, wherein the second nucleotide sequence is 15 to 30 nucleotides in length and comprises a nucleotide sequence as set forth in SEQ ID NOs: 317-632. In some embodiments, the pharmaceutical compositions comprising any of the siNA molecules, sense strands, antisense strands, first nucleotide sequences, or second nucleotide sequences described herein. The compositions may comprise 1, 2, 3, or more siNA molecules described herein. In some embodiments, the composition comprises a double-stranded short interfering nucleic acid (ds-siNA) molecule comprises (a) a sense strand comprising a first nucleotide sequence, wherein the first nucleotide sequence is 15 to 30 nucleotides in length and comprises a nucleotide sequence as set forth in SEQ ID NOs: 633-878; and (b) an antisense strand comprising a second nucleotide sequence, wherein the second nucleotide sequence is 15 to 30 nucleotides in length and comprises a nucleotide sequence that is the reverse complement of the first nucleotide sequence. In some embodiments, the first nucleotide sequence comprises a nucleotide sequence as set forth in SEQ ID NOs: 633-878; and (b) an antisense strand that is the reverse complement of the first nucleotide sequence, wherein the antisense strand comprises a second nucleotide sequence, wherein the second nucleotide sequence is 15 to 30 nucleotides in length and comprises a nucleotide sequence as set forth in SEQ ID NOs: 879-1125. In some embodiments, the composition comprises a double-stranded short interfering nucleic acid (ds-siNA) molecule comprising (a) a sense strand comprising a nucleotide sequence identical to SEQ ID NOs:1-316, and (b) an antisense strand. In some embodiments, the present disclosure provides a double-stranded short interfering nucleic acid (ds-siNA) molecule comprising (a) an antisense strand comprising a nucleotide sequence identical to SEQ ID NOs: 317-632; and (b) a sense strand. In some embodiments, the composition comprises a double-stranded short interfering nucleic acid (ds-siNA) molecule comprising (a) a sense strand comprising a nucleotide sequence identical to SEQ ID NOs:633-878, and (b) an antisense strand. In some embodiments, the present disclosure provides a double-stranded short interfering nucleic acid (ds-siNA) molecule comprising (a) an antisense strand comprising a nucleotide sequence identical to SEQ ID NOs: 879-1125; and (b) a sense strand. In some embodiments, the composition comprises a double-stranded short interfering nucleic acid (ds-siNA) molecule comprising (a) a sense strand as set forth in is SEQ ID NOs: 633, 634, 642, 653, 654, 674, 687, 688, 690-694, 708-713, 756-757, 765, 776, 777, 797, 810, 811, 813-817, or 831-836, and (b) an antisense strand complementary to the sense strand as set forth in SEQ ID NOs: 879, 880, 888, 899, 900, 920, 933, 934, 936-940, 954-959, 1003- 1004, 1012, 1023, 1024, 1044, 1057, 1058, 1060-1064, or 1078-1083, respectively. In some embodiments, the composition comprises a double-stranded short interfering nucleic acid (ds- siNA) molecule comprising (a) a sense strand according to SEQ ID NOs: 644, 654, 688, 690, 708, 713, 756, 777, 811, 813, 831, or 836, and (b) an antisense strand complementary to the sense strand according to SEQ ID NOs: 879, 900, 934, 936, 954, 959, 1003, 1024, 1058, 1060, 1078, or 1083, respectively. Alternatively or additionally, the pharmaceutical compositions may comprise (a) a phosphorylation blocker; and (b) an oligomer. In some embodiments, the phosphorylation blocker is any of the phosphorylation blockers disclosed herein. In some embodiments, the oligomer is any of the oligomers disclosed herein. In some embodiments, the oligomer comprises any of the sense strands, antisense strands, first nucleotide sequences, or second nucleotide sequences described herein. In some embodiments, the oligomer comprises any of the sense strands, antisense strands, first nucleotide sequences, or second nucleotide sequences described herein. In some embodiments, the oligomer comprises one or more modified nucleotides. In some embodiments, the one or more modified nucleotides are independently selected from a 2’-fluoro nucleotide and a 2’-O-methyl nucleotide. In some embodiments, the 2’-fluoro nucleotide or the 2’-O-methyl nucleotide is independently selected from any of the 2’-fluoro or 2’-O-methyl nucleotide mimics disclosed herein. In some embodiments, the oligomer comprises a nucleotide sequence comprising any of the modification patterns disclosed herein. In some embodiments, the pharmaceutical composition comprises (a) a 5’-stabilized end cap; and (b) an oligomer. In some embodiments, the 5’-stabilized end cap is any of the 5- stabilized end caps disclosed herein. In some embodiments, the oligomer is any of the oligomers disclosed herein. In some embodiments, the oligomer comprises any of the sense strands, antisense strands, first nucleotide sequences, or second nucleotide sequences described herein. In some embodiments, the oligomer comprises one or more modified nucleotides. In some embodiments, the one or more modified nucleotides are independently selected from a 2’-fluoro nucleotide and a 2’-O-methyl nucleotide. In some embodiments, the 2’-fluoro nucleotide or the 2’-O-methyl nucleotide is independently selected from any of the 2’-fluoro or 2’-O-methyl nucleotide mimics disclosed herein. In some embodiments, the oligomer comprises a nucleotide sequence comprising any of the modification patterns disclosed herein. In some embodiments, the pharmaceutical composition comprises (a) at least one phosphorylation blocker or 5’-stabilized end cap; and (b) an oligomer. In some embodiments, the phosphorylation blocker is any of the phosphorylation blockers disclosed herein. In some embodiments, the conjugated moiety is any of the galactosamines disclosed herein. In some embodiments, the 5’-stabilized end cap is any of the 5-stabilized end caps disclosed herein. In some embodiments, the oligomer is any of the oligomers disclosed herein. In some embodiments, the oligomer comprises any of the sense strands, antisense strands, first nucleotide sequences, or second nucleotide sequences described herein. In some embodiments, the oligomer comprises one or more modified nucleotides. In some embodiments, the one or more modified nucleotides are independently selected from a 2’- fluoro nucleotide and a 2’-O-methyl nucleotide. In some embodiments, the 2’-fluoro nucleotide or the 2’-O-methyl nucleotide is independently selected from any of the 2’-fluoro or 2’-O-methyl nucleotide mimics disclosed herein. In some embodiments, the oligomer comprises a nucleotide sequence comprising any of the modification patterns disclosed herein. In some embodiments, the pharmaceutical composition containing the siNA of the present disclosure is formulated for systemic administration via parenteral delivery. Parenteral administration includes intravenous, intra-arterial, subcutaneous, intraperitoneal or intramuscular injection or infusion; also subdermal administration, e.g., via an implanted device. In a preferred embodiment, the pharmaceutical composition containing the siNA of the present disclosure is formulated for subcutaneous (SC) or intravenous (IV) delivery. Formulations for parenteral administration may include sterile aqueous solutions, which may also contain buffers, diluents and other pharmaceutically acceptable additives as understood by the skilled artisan. For intravenous use, the total concentration of solutes may be controlled to render the preparation isotonic. When the pharmaceutical composition comprises two or more siNAs, the siNAs may be present in varying amounts. For example, in some embodiments, the weight ratio of first siNA to second siNA is 1:10 to 10:1, e.g., 1:10, 1:9, 1:8, 1:7, 1:6, 1:5, 1:4, 1:3, 1:2, 1:1, 2:1, 3:1, 4:1, 5:1, 6:1, 7:1, 8:1, 9:1, or 10:1. In some embodiments, the molar ratio of first siNA to second siNA is 1:10 to 10:1, e.g., 1:10 to 10:1, e.g., 1:10, 1:9, 1:8, 1:7, 1:6, 1:5, 1:4, 1:3, 1:2, 1:1, 2:1, 3:1, 4:1, 5:1, 6:1, 7:1, 8:1, 9:1, or 10:1. In some embodiments, the pharmaceutical composition comprises an amount of one or more of the siNA molecules described herein formulated with one or more pharmaceutically acceptable carriers (additives) and/or diluents. The pharmaceutical compositions may be specially formulated for administration in solid or liquid form, including those adapted for the following: (1) parenteral administration, for example, by subcutaneous, intramuscular, intravenous or epidural injection as, for example, a sterile solution or suspension, or sustained-release formulation; (2) topical application, for example, as a cream, ointment, or a controlled-release patch or spray applied to the skin; (3) intravaginally or intrarectally, for example, as a pessary, cream or foam; (4) sublingually; (5) ocularly; (6) transdermally; or (7) nasally. Wetting agents, emulsifiers and lubricants, such as sodium lauryl sulfate and magnesium stearate, as well as coloring agents, release agents, coating agents, sweetening, flavoring and perfuming agents, preservatives and antioxidants can also be present in the compositions. Examples of pharmaceutically-acceptable antioxidants include: (1) water soluble antioxidants, such as ascorbic acid, cysteine hydrochloride, sodium bisulfate, sodium metabisulfite, sodium sulfite and the like; (2) oil-soluble antioxidants, such as ascorbyl palmitate, butylated hydroxyanisole (BHA), butylated hydroxytoluene (BHT), lecithin, propyl gallate, alpha-tocopherol, and the like; and (3) metal chelating agents, such as citric acid, ethylenediamine tetraacetic acid (EDTA), sorbitol, tartaric acid, phosphoric acid, and the like. Formulations of the present disclosure include those suitable for nasal, topical (including buccal and sublingual), rectal, vaginal and/or parenteral administration. The formulations may conveniently be presented in unit dosage form and may be prepared by any methods well known in the art of pharmacy. The amount of active ingredient which can be combined with a carrier material to produce a single dosage form will vary depending upon the host being treated, the particular mode of administration. The amount of active ingredient which can be combined with a carrier material to produce a single dosage form will generally be that amount of the compound (e.g., siNA molecule) which produces a therapeutic effect. Generally, out of one hundred percent, this amount will range from about 0.1 percent to about ninety-nine percent of active ingredient, preferably from about 5 percent to about 70 percent, most preferably from about 10 percent to about 30 percent. In some embodiments, a formulation of the present disclosure comprises an excipient selected from the group consisting of cyclodextrins, celluloses, liposomes, micelle forming agents, e.g., bile acids, and polymeric carriers, e.g., polyesters and polyanhydrides; and a compound (e.g., siNA molecule) of the present disclosure. Methods of preparing these formulations or compositions include the step of bringing into association a compound (e.g., siNA molecule) of the present disclosure with the carrier and, optionally, one or more accessory ingredients. In general, the formulations are prepared by uniformly and intimately bringing into association a compound (e.g., siNA molecule) of the present disclosure with liquid carriers, or finely divided solid carriers, or both, and then, if necessary, shaping the product. Formulations of the disclosure suitable for a suspension in an aqueous or non-aqueous liquid, or as an oil-in-water or water-in-oil liquid emulsion, or as an elixir or syrup, each containing a predetermined amount of a compound (e.g., siNA molecule) of the present disclosure as an active ingredient. A compound (e.g., siNA molecule) of the present disclosure may also be administered as a bolus, electuary, or paste. In dosage forms of the disclosure, the active ingredient may be mixed with one or more pharmaceutically-acceptable carriers, such as sodium citrate or dicalcium phosphate, and/or any of the following: (1) fillers or extenders, such as starches, lactose, sucrose, glucose, mannitol, and/or silicic acid; (2) binders, such as, for example, carboxymethylcellulose, alginates, gelatin, polyvinyl pyrrolidone, sucrose and/or acacia; (3) humectants, such as glycerol; (4) disintegrating agents, such as agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates, and sodium carbonate; (5) solution retarding agents, such as paraffin; (6) absorption accelerators, such as quaternary ammonium compounds and surfactants, such as poloxamer and sodium lauryl sulfate; (7) wetting agents, such as, for example, cetyl alcohol, glycerol monostearate, and non-ionic surfactants; (8) absorbents, such as kaolin and bentonite clay; (9) lubricants, such as talc, calcium stearate, magnesium stearate, solid polyethylene glycols, sodium lauryl sulfate, zinc stearate, sodium stearate, stearic acid, and mixtures thereof; (10) coloring agents; and (11) controlled release agents such as crospovidone or ethyl cellulose. The disclosed dosage forms may be sterilized by, for example, filtration through a bacteria-retaining filter, or by incorporating sterilizing agents in the form of sterile solid compositions which can be dissolved in sterile water, or some other sterile injectable medium immediately before use. These compositions may also optionally contain opacifying agents and may be of a composition that they release the active ingredient(s) only, or preferentially, in a certain portion of the gastrointestinal tract, optionally, in a delayed manner. Examples of embedding compositions which can be used include polymeric substances and waxes. The active ingredient can also be in micro-encapsulated form, if appropriate, with one or more of the above-described excipients. Liquid dosage forms of the compounds (e.g., siNA molecules) of the disclosure include pharmaceutically acceptable emulsions, microemulsions, solutions, suspensions, syrups and elixirs. In addition to the active ingredient, the liquid dosage forms may contain inert diluents commonly used in the art, such as, for example, water or other solvents, solubilizing agents and emulsifiers, such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1,3-butylene glycol, oils (I particular, cottonseed, groundnut, corn, germ, olive, castor and sesame oils), glycerol, tetrahydrofuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, and mixtures thereof. Besides inert diluents, the compositions can also include adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, coloring, perfuming and preservative agents. Suspensions, in addition to the active compounds (e.g., siNA molecules), may contain suspending agents as, for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum metahydroxide, bentonite, agar-agar and tragacanth, and mixtures thereof. Formulations of the pharmaceutical compositions of the disclosure for rectal or vaginal administration may be presented as a suppository, which may be prepared by mixing one or more compounds (e.g., siNA molecules) of the disclosure with one or more suitable nonirritating excipients or carriers comprising, for example, cocoa butter, polyethylene glycol, a suppository wax or a salicylate, and which is solid at room temperature, but liquid at body temperature and, therefore, will melt in the rectum or vaginal cavity and release the active compound (e.g., siNA molecule). Formulations of the present disclosure which are suitable for vaginal administration also include pessaries, tampons, creams, gels, pastes, foams or spray formulations containing such carriers as are known in the art to be appropriate. Dosage forms for the topical or transdermal administration of a compound (e.g., siNA molecule) of this disclosure include powders, sprays, ointments, pastes, creams, lotions, gels, solutions, patches and inhalants. The active compound (e.g., siNA molecule) may be mixed under sterile conditions with a pharmaceutically acceptable carrier, and with any preservatives, buffers, or propellants which may be required. The ointments, pastes, creams and gels may contain, in addition to an active compound (e.g., siNA molecule) of this disclosure, excipients, such as animal and vegetable fats, oils, waxes, paraffins, starch, tragacanth, cellulose derivatives, polyethylene glycols, silicones, bentonites, silicic acid, talc and zinc oxide, or mixtures thereof. Powders and sprays can contain, in addition to a compound (e.g., siNA molecule) of this disclosure, excipients such as lactose, talc, silicic acid, aluminum hydroxide, calcium silicates and polyamide powder, or mixtures of these substances. Sprays can additionally contain customary propellants, such as chlorofluorohydrocarbons and volatile unsubstituted hydrocarbons, such as butane and propane. Transdermal patches have the added advantage of providing controlled delivery of a compound (e.g., siNA molecule) of the present disclosure to the body. Such dosage forms can be made by dissolving or dispersing the compound (e.g., siNA molecule) in the proper medium. Absorption enhancers can also be used to increase the flux of the compound (e.g., siNA molecule) across the skin. The rate of such flux can be controlled by either providing a rate controlling membrane or dispersing the compound (e.g., siNA molecule) in a polymer matrix or gel. Ophthalmic formulations, eye ointments, powders, solutions and the like, are also contemplated as being within the scope of this invention. Pharmaceutical compositions of this disclosure suitable for parenteral administration comprise one or more compounds (e.g., siNA molecules) of the disclosure in combination with one or more pharmaceutically-acceptable sterile isotonic aqueous or nonaqueous solutions, dispersions, suspensions or emulsions, or sterile powders which may be reconstituted into sterile injectable solutions or dispersions just prior to use, which may contain sugars, alcohols, antioxidants, buffers, bacteriostats, solutes which render the formulation isotonic with the blood of the intended recipient or suspending or thickening agents. Examples of suitable aqueous and nonaqueous carriers which may be employed in the pharmaceutical compositions of the disclosure include water, ethanol, polyols (such as glycerol, propylene glycol, polyethylene glycol, and the like), and suitable mixtures thereof, vegetable oils, such as olive oil, and injectable organic esters, such as ethyl oleate. Proper fluidity can be maintained, for example, by the use of coating materials, such as lecithin, by the maintenance of the required particle size in the case of dispersions, and by the use of surfactants. These compositions may also contain adjuvants such as preservatives, wetting agents, emulsifying agents and dispersing agents. Prevention of the action of microorganisms upon the subject compounds may be ensured by the inclusion of various antibacterial and antifungal agents, for example, paraben, chlorobutanol, phenol sorbic acid, and the like. It may also be desirable to include isotonic agents, such as sugars, sodium chloride, and the like into the compositions. In addition, prolonged absorption of the injectable pharmaceutical form may be brought about by the inclusion of agents which delay absorption such as aluminum monostearate and gelatin. In some cases, in order to prolong the effect of a drug, it is desirable to slow the absorption of the drug from subcutaneous or intramuscular injection. This may be accomplished by the use of a liquid suspension of crystalline or amorphous material having poor water solubility. The rate of absorption of the drug then depends upon its rate of dissolution which, in turn, may depend upon crystal size and crystalline form. Alternatively, delayed absorption of a parenterally-administered drug form is accomplished by dissolving or suspending the drug in an oil vehicle. Injectable depot forms are made by forming microencapsule matrices of the subject compounds (e.g., siNA molecules) in biodegradable polymers such as polylactide- polyglycolide. Depending on the ratio of drug to polymer, and the nature of the particular polymer employed, the rate of drug release can be controlled. Examples of other biodegradable polymers include poly(orthoesters) and poly(anhydrides). Depot injectable formulations are also prepared by entrapping the drug in liposomes or microemulsions which are compatible with body tissue. In some embodiments, the present disclosure provides a pharmaceutical composition comprising the siNAs disclosed herein conjugated to a ligand targeting receptors expressed in hepatocytes to treat Cardiovascular Diseases (CVD) and MASH The ligands that could be conjugated to the siNAs include but are not limited to GalNAc, Cholesterol, Palmitic Acid etc. In some embodiments, the pharmaceutical composition comprises a liposome that encapsulates the siNAs disclosed herein to form a lipid nanoparticle (LNP) targeting hepatocellular carcinoma (HCC) or other secondary cancers metastasized to the liver. In some embodiments, the present disclosure provides a pharmaceutical composition comprising the siNAs disclosed herein conjugated to a ligand targeting receptors expressed in HCC. The ligands that could be conjugated to the siNAs include but are not limited to GalNAc, Folate, Cholesterol, Palmitic Acid etc. When the compounds (e.g., siNA molecules) of the present disclosure are administered as pharmaceuticals, to humans and animals, they can be given per se or as a pharmaceutical composition containing, for example, 0.1 to 99% (more preferably, 10 to 30%) of active ingredient in combination with a pharmaceutically acceptable carrier. In some embodiments, the pharmaceutical acceptable carriers is a liposome that encapsulates the ds- siNA molecules. Methods of Treatment and Administration The siNA molecules of the present disclosure may be used to treat or prevent a disease in a subject in need thereof. In some embodiments, a method of treating or preventing a disease in a subject in need thereof comprises administering to the subject any of the siNA molecules disclosed herein. In some embodiments, a method of treating or preventing a disease in a subject in need thereof comprises administering to the subject any of the compositions disclosed herein. In some embodiments, the subject is a mammal. In some embodiments, the subject is a human. In some embodiments, the subject is a non-human primate. In some embodiments, the subject is a cat. In some embodiments, the subject is a camel. In preferred embodiments in which the subject is a human, the subject may be at least 40 years old, at least 45 years old, at least 50 years old, at least 55 years old, at least 60 years old, at least 65 years old, at least 70 years old, at least 75 years old, or at least 80 years old or older. In some embodiments, the subject is a pediatric subject (i.e., less than 18 years old). The preparations (e.g., siNA molecules or pharmaceutical compositions thereof) of the present disclosure may be given parenterally, topically, or rectally or administered in the form of an inhalant. They are, of course, given in forms suitable for each administration route. For example, they are administered in tablets or capsule form, administration by injection, infusion, or inhalation; topical by lotion or ointment; rectal by suppositories. Injection, infusion, or inhalation are preferred. In some embodiments, the siNA molecules are given in the form of nanoparticle where a liposome encapsulates the siNA molecules. These compounds may be administered to humans and other animals for therapy or as a prophylactic by any suitable route of administration, including nasally (as by, for example, a spray), rectally, intravaginally, parenterally, intracisternally and topically, as by powders, ointments or drops, including buccally and sublingually. In some embodiments, the compounds or compositions are inhaled, as by, for example, an inhaler, a nebulizer, or in an aerosolized form. Regardless of the route of administration selected, the compounds (e.g., siNA molecules) of the present disclosure, which may be used in a suitable hydrated form, and/or the pharmaceutical compositions of the present disclosure, are formulated into pharmaceutically-acceptable dosage forms by conventional methods known to those of skill in the art. In some embodiments, the present disclosure provides methods of treating or preventing a disease in a subject in need thereof, comprising administering the subject the ds- siNA molecules disclosed herein to reduce ANGPTL8 and/or ANGPTL3 activity or level. In some embodiments, the method of treating or preventing the disease comprises administering the subject the pharmaceutical composition comprising the ds-siNA molecules disclosed herein to reduce ANGPTL8 and/or ANGPTL3 activity or level. In some embodiments, the disease is a ANGPTL8 and/or ANGPTL3 associated disease or disorder. In some embodiments, the ANGPTL8 and/or ANGPTL3 associated disease or disorder comprises cardiovascular disease or nonalcoholic fatty liver disease (NAFLD), such as metabolic dysfunction-associated steatohepatitis (MASH). In some embodiments, the cardiovascular disease is hypertriglyceridemia (HTG) or familial hypercholesterolemia (FH). Actual dosage levels of the active ingredients (e.g., siNA molecules) in the pharmaceutical compositions of this disclosure may be varied so as to obtain an amount of the active ingredient which is effective to achieve the desired therapeutic response for a particular patient, composition, and mode of administration, without being toxic to the patient. The selected dosage level will depend upon a variety of factors including the activity of the particular compound (e.g., siNA molecule) of the present disclosure employed, or the ester, salt or amide thereof, the route of administration, the time of administration, the rate of excretion or metabolism of the particular compound being employed, the rate and extent of absorption, the duration of the treatment, other drugs, compounds and/or materials used in combination with the particular compound employed, the age, sex, weight, condition, general health and prior medical history of the patient being treated, and like factors well known in the medical arts. A physician or veterinarian having ordinary skill in the art can readily determine and prescribe the effective amount of the pharmaceutical composition required. For example, the physician or veterinarian could start doses of the compounds (e.g., siNA molecules) of the disclosure employed in the pharmaceutical composition at levels lower than that required in order to achieve the desired therapeutic effect and gradually increase the dosage until the desired effect is achieved. In general, a suitable daily dose of a compound (e.g., siNA molecule) of the disclosure is the amount of the compound that is the lowest dose effective to produce a therapeutic effect. Such an effective dose generally depends upon the factors described above. Preferably, the compounds are administered at about 0.01 mg/kg to about 200 mg/kg, more preferably at about 0.1 mg/kg to about 100 mg/kg, even more preferably at about 0.5 mg/kg to about 50 mg/kg. In some embodiments, the compound is administered at a dose equal to or greater than 0.01, 0.02, 0.03, 0.04, 0.05, 0.06, 0.07, 0.08, 0.09, 0.10, 0.11, 0.12, 0.13, 0.14, 0.15, 0.16, 0.17, 0.18, 0.19, 0.20, 0.21, 0.22, 0.23, 0.24, 0.25, 0.26, 0.27, 0.28, 0.29, 0.30, 0.35, 0.40, 0.45, 0.50, 0.55, 0.60, 0.65, 0.7, 0.75, 0.8, 0.85, 0.9, 0.95, or 1 mg/kg. In some embodiments, the compound is administered at a dose equal to or less than 200, 190, 180, 170, 160, 150, 140, 130, 120, 110, 100, 95, 90, 85, 80, 75, 70, 65, 60, 55, 50, 45, 40, 35, 30, 25, 20, or 15 mg/kg. In some embodiments, the total daily dose of the compound is equal to or greater than 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 105, 110, 115, 120, 125, 130, 135, 140, 145, 150, 155, 160, 165, 170, 175, 180, 185, 190, 195, or 100 mg. When the compounds (e.g., siNA molecules) described herein are co-administered with another, the effective amount may be less than when the compound is used alone. If desired, the effective daily dose of the active compound (e.g., siNA molecule) may be administered as two, three, four, five, six or more sub-doses administered separately at appropriate intervals throughout the day, optionally, in unit dosage forms. Preferred dosing is one administration per day. In some embodiments, the compound is administered at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, or 21 times a week. In some embodiments, the compound is administered at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, or 21 times a month. In some embodiments, the compound is administered once every 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, or 21 days. In some embodiments, the compound is administered once every 1, 2, 3, 4, 5, 6, 7, or 8 weeks. In some embodiments, the disclosed siNAs and pharmaceutical compositions comprising the disclosed siNAs are combined with approved or late stage development drugs in cardiovascular disease. Non-limiting examples include statins including atorvastatin, fluvastatin, lovastatin, pitavastatin, pravastatin, rosuvastatin and simvastatin; PCSK9 inhibitors including alirocumab, evolocumab, and inclisiran; as well as cholesterol uptake inhibitor Ezetimibe. In some embodiments, the disclosed siNAs and pharmaceutical compositions comprising the disclosed siNAs are combined with approved or late stage development drugs in nonalcoholic fatty liver disease. Non-limiting examples include Resmetirom, ALG-055009, VK2809, efruxifermin, Saroglitazar, Obeticholic acid, Cenicriviroc, Aramchol, Dapagliflozin, Semaglutide, Belapectin, MSDC0602K, Lanifibranor and Tesamorelin. In some embodiments, the pharmaceutical composition comprises at least a first siNA that reduces ANGPTL3 expression and a second siNA that reduces ANGPTL8 expression as disclosed herein and a pharmaceutically acceptable carrier. In some embodiments, the a weight ratio of the first siNA and the second siNA is selected from 1:10, 1:9, 1:8, 1:7, 1:6, 1:5, 1:4, 1:3, 1:2, 1:1, 2:1, 3:1, 4:1, 5:1, 6:1, 7:1, 8:1, 9:1, or 10:1. In some embodiments, a molar ration of the first siNA and the second ds-siNA is selected from 1:10, 1:9, 1:8, 1:7, 1:6, 1:5, 1:4, 1:3, 1:2, 1:1, 2:1, 3:1, 4:1, 5:1, 6:1, 7:1, 8:1, 9:1, or 10:1. In some embodiments, the first siNA and the second siNA are physically linked by a linker. Non-limiting examples of linkers are nucleotide based linkers or non-nucleotide based linkers. In some embodiments, the nucleotide based linker is between about 1 to about 15 nucleotides in length. In some embodiments, the nucleotide based linker is 1 or more, 2 or more, 3 or more, 4 or more, 5 or more, 6 or more, 7 or more, 8 or more, 9 or more, 10 or more, 11 or more, 12 or more, 13 or more, 14 or more, or 15 or more nucleotides in length. In some embodiments, the linker is at least one or more one N-acetylgalactosamine (e.g., a GalNAc) as disclosed herein. The siNAs of the disclosure optionally can be used in combination with modulators of other genes and/or gene products associated with the maintenance or development of diseases or disorders associated with aberrant ANGPTL8 and/or ANGPTL3 expression. EXAMPLES These examples are provided for illustrative purposes only and not to limit the scope of the claims provided herein. For the purposes of these examples, the dsNA identifiers refer to the modified sequences provided in Tables 2 and 4. Comparative control siNAs for the examples below have the sequences set forth in Table 5. Table 5 – Comparative Control siNAs Sequences
Figure imgf000137_0001
Figure imgf000138_0001
Example 1. Evaluation of ANGPTLA3 siNAs using a three-point luciferase reporter assay In the psiCHECK™-2 vector, Renilla luciferase was used as the primary reporter gene with the ANGPTL3 gene (NM_014495.4) cloned downstream of its translational stop codon. A second reporter gene, firefly luciferase, was expressed and used as a negative transfection control. The plasmid was first transfected using Lipofectamine 3000 (Invitrogen, L3000001) into COS-7 cells (ATCC, CRL-1651) seeded into 96-well microplates. The cells were then transfected with 10, 1, or 0.1 nM siNAs using Lipofectamine RNAiMAX (Invitrogen, 13778100). A mock, no-drug negative control, which consisted of transfecting 1× phosphate- buffered saline was included. Parental siNA dsNA-240 was used as the positive comparative control. After 72 hours of siNA treatment, the Dual-Glo® Luciferase Assay System (Promega, E2940) was used according to the manufacturer’s protocol to quantify firefly and Renilla luciferase activity. All luminescence was measured on an EnVision plate reader (Perkin Elmer). The firefly:Renilla luminescence ratio was calculated for each well. The ratios from siNA-treated wells were then normalized to ratios of the mock-treated wells and percentage inhibition was calculated (Table 6). Table 6 - Results of Three-Point Luciferase Reporter Assay for ANGPTL3 in COS-7 Cells
Figure imgf000139_0001
Figure imgf000140_0001
Figure imgf000141_0001
Example 2. Evaluation of ANGPTL3 siNAs using dose-response assays Select candidates were defined in Example 1 as showing ≥65% inhibition at 1 nM and ≥85% inhibition at 10 nM. Dose-response assays were conducted, with serial concentrations of siNAs starting at 10 nM (1:5 dilutions) for each select candidate siNA using the Dual- Glo® Luciferase Assay System for ANGPTL3 as described in Example 1. Dose-response curves were fitted by nonlinear regression with variable slope, and EC50 values and maximum percentage inhibition were calculated (Table 7). Table 7 – Results of Dose-Response Luciferase Reporter Assay for ANGPTL3 in COS-7 Cells
Figure imgf000142_0001
For the RT-qPCR assay, hepatoma-derived Huh-7 cells (JCRB Cell Bank, JCRB0403) were transfected with serially diluted siNA starting at 10 nM (1:5 dilutions) and Opti- MEM™ using Lipofectamine RNAiMAX (Invitrogen, 13778100). A negative mock transfection control, which consisted of transfecting 1× phosphate-buffered saline, was included. After about 48 hours of siNA treatment, the Huh-7 cells were processed with the TaqMan Fast Advanced Cells-to-Ct Kit (Invitrogen, A35378), according to the manufacturer’s protocol. The cell lysates were used for reverse transcription. Gene expression was measured using TaqMan Fast Advanced Master Mix (Applied Biosystems, 4444964) and the following TaqMan Gene Expression assays (Applied Biosystems, 4331182): ACTB (Hs01060665_g1) and ANGPTL3 (Hs00205581_m1). ACTB served as the endogenous control gene. RT-qPCR reactions were run on the QuantStudio™ 6 Pro Real- Time PCR System (Applied Biosystems). The RQ of gene expression was calculated via the 2−ΔΔCt (RQ) method. Results are presented as expression relative to the expression levels of negative mock control samples. Dose-response curves were fitted by nonlinear regression with variable slope, EC50 values, and maximum percentage inhibition calculated (Table 8). Table 8 – Results of Dose-Response RT-qPCR Assay for ANGPTL3 in Huh-7 Cells
Figure imgf000143_0001
Figure imgf000144_0001
For the RT-qPCR assay, hepatoma-derived Hep3B cells (ATCC, HB-8064) were transfected with serially diluted siNA starting at 10 nM (1:8 dilutions) and Opti-MEM™ using Lipofectamine RNAiMAX (Invitrogen, 13778100). A negative mock transfection control, which consisted of transfecting 1× phosphate-buffered saline, was included. After about 48 hours of siNA treatment, the Hep3B cells were processed with the TaqMan Fast Advanced Cells-to-Ct Kit (Invitrogen, A35378), according to the manufacturer’s protocol. The cell lysates were used for reverse transcription. Gene expression was measured using TaqMan Fast Advanced Master Mix (Applied Biosystems, 4444964) and the following TaqMan Gene Expression assays (Applied Biosystems, 4331182): ACTB (Hs01060665_g1) and ANGPTL3 (Hs00205581_m1). ACTB served as the endogenous control gene. RT-qPCR reactions were run on the QuantStudio™ 6 Pro Real-Time PCR System (Applied Biosystems). The RQ of gene expression was calculated via the 2−ΔΔCt (RQ) method. Results are presented as expression relative to the expression levels of negative mock control samples. Dose-response curves were fitted by nonlinear regression with variable slope, EC50 values, and maximum percentage inhibition calculated (Table 9). Table 9 – Results of Dose-Response RT-qPCR Assay for ANGPTL3 in Hep3B Cells
Figure imgf000145_0001
Figure imgf000146_0001
The human ANGPTL3 siNAs dsNA-240, dsNA-241, dsNA-242, dsNA-243, dsNA- 244, dsNA-245, dsNA-250, dsNA-251, dsNA-252, dsNA-253, dsNA-254, dsNA-255, dsNA- 260, dsNA-262, dsNA-263, dsNA-264, dsNA-265, dsNA-266, dsNA-267, dsNA-268, dsNA- 269, dsNA-274, dsNA-275, dsNA-276, dsNA-280 and dsNA-281 showed better in vitro potency than the comparative control siNA, while dsNA-257, dsNA-258, dsNA-270, dsNA- 271, dsNA-272, dsNA-273, dsNA-277, dsNA-278 and dsNA-279 showed similar potency to the comparative control siNA. Example 3. Toxicity assay and cross-species conservation for ANGPTL3 siNAs The candidates were further narrowed to six select siNAs, dsNA-159, dsNA-180, dsNA-214, dsNA-216, dsNA-234, and dsNA-239. CellTiter-Glo® Luminescent Cell Viability Assays were performed in siNA-treated COS-7 cells under conditions as described in Example 1. Assays were performed according to the manufacturer’s protocol and luminescence was measured on an EnVision plate reader. The luminescence from siNA- treated wells were then normalized to luminescence of mock-treated wells and percentage viability was calculated. (FIG. 2A). No siNAs exhibited cytotoxic effects and all had a CC50 of >10 nM. All six candidates were also assessed for ANGPTL3 inhibition against the cynomolgus monkey ANGPTL3 using a psiCHECK™-2 system similarly described in Example 1, but with the Macaca fascicularis gene (XM_005543185.3) inserted. The activity of the candidates was highly conserved between species (FIG. 2B). Example 4. Evaluation of ANGPTLA8 siNAs using a three-point RT-qPCR reporter assay. For the RT-qPCR assay, hepatoma-derived Hep3B cells (ATCC, HB-8064) were transfected with siNA at 10.0, 1.0, or 0.1 nM and Opti-MEM™ using Lipofectamine RNAiMAX (Invitrogen, 13778100). A negative mock transfection control, which consisted of transfecting 1× phosphate-buffered saline, was included. After about 48 hours of siNA treatment, the Hep3B cells were processed with the TaqMan Fast Advanced Cells-to-Ct Kit (Invitrogen, A35378), according to the manufacturer’s protocol. The cell lysates were used for reverse transcription. Gene expression was measured using TaqMan Fast Advanced Master Mix (Applied Biosystems, 4444964) and the following TaqMan Gene Expression assays (Applied Biosystems, 4331182): ACTB (Hs01060665_g1) and ANGPTL8 (Hs00218820_m1). ACTB served as the endogenous control gene. RT-qPCR reactions were run on the QuantStudio™ 6 Pro Real-Time PCR System (Applied Biosystems). The RQ of gene expression was calculated via the 2−ΔΔCt (RQ) method. Results are presented as expression relative to the expression levels of negative mock control samples. Percentage inhibition at each dose was calculated (Table 10). Table 10 - Results of Three-Point RT-qPCR Assay for ANGPTL8 in Hep3B Cells.
Figure imgf000147_0001
Figure imgf000148_0001
Figure imgf000149_0001
Figure imgf000150_0001
Figure imgf000151_0001
Example 5. Evaluation of ANGPTL8 siNAs using a dose-response RT-qPCT assay. For the RT-qPCR assay, hepatoma-derived Hep3B cells (ATCC, HB-8064) were transfected with serially diluted siNA starting at 10 nM (1:8 dilutions) and Opti-MEM™ using Lipofectamine RNAiMAX (Invitrogen, 13778100). A negative mock transfection control, which consisted of transfecting 1× phosphate-buffered saline, was included. After about 48 hours of siNA treatment, the Hep3B cells were processed with the TaqMan Fast Advanced Cells-to-Ct Kit (Invitrogen, A35378), according to the manufacturer’s protocol. The cell lysates were used for reverse transcription. Gene expression was measured using TaqMan Fast Advanced Master Mix (Applied Biosystems, 4444964) and the following TaqMan Gene Expression assays (Applied Biosystems, 4331182): ACTB (Hs01060665_g1) and ANGPTL8 (Hs00218820_m1). ACTB served as the endogenous control gene. RT-qPCR reactions were run on the QuantStudio™ 6 Pro Real-Time PCR System (Applied Biosystems). The RQ of gene expression was calculated via the 2−ΔΔCt (RQ) method. Results are presented as expression relative to the expression levels of negative mock control samples. Dose-response curves were fitted by nonlinear regression with variable slope, EC50 values, and maximum percentage inhibition calculated (Table 11). Most of the tested siNAs exhibited similar or enhanced potency and maximum percentage of ANGPTL8 RNA inhibition as the comparative control siNA dsNA-284. Table 11 – Results of Dose-Response RT-qPCR Assay for ANGPTL8 in Hep3B Cells
Figure imgf000152_0001
Figure imgf000153_0001
Example 6. AAV-hANGPTL3 Mouse Study Design To evaluate certain ANGPTL3 siNA agents, an AAV-ANGPTL3 mouse model was used. Mice (n=4-5/group) were injected with an adeno-associated virus (AAV) expressing human ANGPTL3. Fourteen days after AAV injection, mice were subcutaneously administered a single dose (5 mg/kg) of siNA test article. Fourteen days after administration, the animals were sacrificed; the right, lateral liver lobe of each animal was collected for RT- qPCR and blood of each animal was collected for ELISA. For RT-qPCR, RNA was extracted using the Rneasy Mini Kit (Qiagen, 74106), according to the manufacturer’s protocol. RNA quantity and quality was analyzed with a NanoDrop™ Lite Spectrophotometer (Thermo Scientific), and cDNA was synthesized using the SuperScript IV VILO Master Mix (Invitrogen, 11756500), according to the manufacturer’s protocol. Gene expression was measured using TaqMan Fast Advanced Master Mix (Applied Biosystems, 4444964), and the following TaqMan Gene Expression assays (Applied Biosystems, 4331182): Actb (Mm00607939_s1), ANGPTL3 (Hs00205581_m1), B2m (Mm00437762_m1), and Hmbs (Mm01143545_m1). Actb, B2m, and Hmbs served as the endogenous control genes. RT-qPCR reactions were run on the QuantStudio™ 6 Pro Real-Time PCR System (Applied Biosystems). The RQ of gene expression was calculated via the 2−ΔΔCt (RQ) method. Results are presented as percent change in expression relative to the expression levels of negative vehicle control samples. For ELISA, serum was separated from blood samples. Levels human ANGPTL3 protein in sera were quantified using ELISA (R&D Systems, DANL30) according to the manufacturer’s protocol. Results are presented as percent change in expression relative to the expression levels of negative vehicle control samples (Table 12). Note, the body weights of mice in all test article groups were normal and showed no toxicity from treatment. Table 12 – Day 28 Results of AAV-hANGPTL3 Mouse Study
Figure imgf000154_0001
Based on these results, dsNA-240, dsNA-241, dsNA-243 and dsNA-244 showed better human ANGPTL3 RNA and protein knockdown than comparative control dsNA-283. Example 7. AAV-hANGPTL3 Study Design. To evaluate certain ANGPTL3 siNA agents, an AAV-ANGPTL3 mouse model was used. Mice (n=4/group) were injected with an adeno-associated virus (AAV) expressing human ANGPTL3. Seven days after AAV injection, mice were subcutaneously administered a single dose (0.3, 1.5, or 5.0 mg/kg) of an siNA (dsNA-283; dsNA-244) or a single dose of 5 mg/kg (dsNA-247; dsNA-248; dsNA-249). Twenty-eight days after administration, the animals were sacrificed; the right, lateral liver lobe of each animal was collected for RT- qPCR and blood of each animal was collected for ELISA. (Table 13). Table 13 – Example 7 Design Summary Table
Figure imgf000154_0003
Figure imgf000154_0002
Figure imgf000155_0001
For RT-qPCR, RNA was extracted using the Rneasy Mini Kit (Qiagen, 74106), according to the manufacturer’s protocol. RNA quantity and quality was analyzed with a NanoDrop™ Lite Spectrophotometer (Thermo Scientific), and cDNA was synthesized using the SuperScript IV VILO Master Mix (Invitrogen, 11756500), according to the manufacturer’s protocol. Gene expression was measured using TaqMan Fast Advanced Master Mix (Applied Biosystems, 4444964), and the following TaqMan Gene Expression assays (Applied Biosystems, 4331182): Actb (Mm00607939_s1), ANGPTL3 (Hs00205581_m1), Angptl3 (Mm00803820_m1), B2m (Mm00437762_m1), and Hmbs (Mm01143545_m1). Actb, B2m, and Hmbs served as the endogenous control genes. RT- qPCR reactions were run on the QuantStudio™ 6 Pro Real-Time PCR System (Applied Biosystems). The RQ of gene expression was calculated via the 2−ΔΔCt (RQ) method. Results are presented as percent change in expression relative to the expression levels of negative vehicle control samples. For ELISA, serum was separated from blood samples. Levels human ANGPTL3 protein in sera were quantified using ELISA (R&D Systems, DANL30) according to the manufacturer’s protocol. Results are presented as percent change in expression relative to the expression levels of negative vehicle control samples (FIG. 3) The results indicated that dsNA-244 showed greater target protein knockdown than comparative control dsNA-283 at all 3 dose levels (0.3, 1.5, or 5.0 mg/kg) and that dsNA- 247, dsNA-248 or dsNA-249 also were more capable of knocking down the target protein at 5 mg/kg than comparative control dsNA-283. Example 8. PD Study in Human ANGPTL3 Knock-In Mice. To evaluate certain ANGPTL3 siNA agents, a knock-in mouse model that expresses human ANGPTL3 (KI-ANGPTL3) was used. Mice (n=6/group) were subcutaneously administered a single dose (5 mg/kg) of siNA test article. Blood was collected at various timepoints throughout the study for ELISA and lipid quantification. Twenty-eight days after administration, the animals were sacrificed; the right, lateral liver lobe of each animal was collected for RT-qPCR and blood of each animal was collected for ELISA and lipid quantification. (Table 14 and 15). Table 14 – Treatment Groups for Example 8.
Figure imgf000156_0001
Table 15 – Experimental Timeline for Example 8.
Figure imgf000156_0002
Figure imgf000157_0001
For RT-qPCR, RNA was extracted from the liver samples and cDNA was synthesized. Gene expression was measured using TaqMan Fast Advanced Master Mix (Applied Biosystems, 4444964), and the following TaqMan Gene Expression assays (Applied Biosystems, 4331182): Actb (Mm00607939_s1), ANGPTL3 (Hs00205581_m1), B2m (Mm00437762_m1), and Hmbs (Mm01143545_m1). Actb, B2m, and Hmbs served as the endogenous control genes. RT-qPCR reactions were run on the QuantStudio™ 6 Pro Real- Time PCR System (Applied Biosystems). The RQ of gene expression was calculated via the 2−ΔΔCt (RQ) method. Results are presented as percent change in expression relative to the expression levels of negative vehicle control samples. Knockdown of ANGPTL3 RNA in the liver samples is shown in FIG. 5. Serum was separated from blood samples. Human ANGPTL3 protein levels in the serum were quantified using ELISA (Abcam, ab254510) (FIG. 4). Triglycerides and total cholesterol levels in the serum were also quantified using a Roche Cobas 6000 c501 Chemistry Analyzer. Absolute protein and lipid concentrations are reported (Table 16; FIG. 4). Table 16. Maximum Percent Serum Lipid Reduction vs. Vehicle-Only Negative Control During 28-Day Treatment Period
Figure imgf000157_0002
Figure imgf000158_0001
The results of the liver ANGPTL3 RNA knockdown experiment showed that dsNA- 240, dsNA-244, dsNA-245 and dsNA-247 showed greater target RNA knockdown than comparative control dsNA-283, while dsNA-243 and dsNA-246 showed less target RNA knockdown than the comparative control siNA. In addition, the results of the serum ANGPTL3 protein knockdown exhibited a similar trend with dsNA-240, dsNA-244 and dsNA-247 showing a higher percentage of target protein knockdown than comparative control dsNA-283, while dsNA-243, dsNA-245 and dsNA-246 showed similar target protein knockdown as the comparative control. Finally, the results of the serum lipid reduction study showed that dsNA-240, dsNA-244 and dsNA-245 had overall better lipid reduction profiles than that of comparative control dsNA-283, but dsNA-243, dsNA-246 and dsNA-247 did not show significant improvement when compared with comparative control dsNA-283. Example 9. RNA-Seq Off-Target Analysis. To assess the off-target profile, 10 nM siNA was transfected into PHH and after 24 hours, total RNA was isolated. On target RNA knockdown (human ANGPTL3) in total RNA was confirmed by RT-PCR and standard RNA-Seq was carried out by Genewiz/Azenta. The raw FASTQ files were trimmed using Trimmomatic (v.0.39) to eliminate adapter sequences, then reads were aligned and mapped to genes from the Homo sapiens GRCh38.104 Ensembl annotation using STAR. The aligned/mapped reads were counted/summarized using featureCounts. Differential expressed gene (DEG) analysis was carried out using DESeq2 in R. Genes with fold change greater than 2 and adjusted p-value less than 0.05 were counted as DE genes. The results of this analysis are shown in Table 17. Table 17 – Quantification of RNA-Seq Off-Target Analysis
Figure imgf000159_0001
All siNA showed good human ANGPTL3 target RNA knockdown at 10 nM in Primary Human Hepatocytes (PHH). However, dsNA-244 (DEG = 1), dsNA-243 (DEG = 3) and dsNA-241 (DEG = 5) have fewer off targets than comparative control dsNA-283 (DEG = 7). While dsNA-240 has good potency and knockdown capability, it does not have ideal off target profile as it has 45 DE genes identified in the RNA-Seq analysis. Thus, the seed region of dsNA-240 would be an ideal candidate for testing chemical modifications to reduce off targets while preserving the potency. Example 10. Modifying siNA candidate dsNA-240 by applying Aligos chemistries to destabilize the seed region. In order to improve dsNA-240 to maintain potency while reducing off-target binding, a seed destabilizing strategy was applied. This strategy involves incorporation of various nucleotide modifications, including UNA, seco, 3-OH, 3-OCP, or xylo-F chemistries, into key positions in the antisense seed region, which include positions 6 and 7 from the 5’ end of the antisense strand. When the modified siNAs generated via this strategy were tested, all candidates maintained in vitro potency (Table 18). Additional seed destabilized siNAs were generated in a second subsequent round (Table 19), This second round of testing further confirmed that seed destabilization does not significantly affect compound potency. Table 18 – Seed Destabilized siNA Round 1 EC50 Results
Figure imgf000160_0001
Table 19 – Seed Destabilized siNA Round 2 EC50 Results
Figure imgf000160_0002
Figure imgf000161_0001
Example 11. Plasmid Based On- and Off-Target Luciferase Assay. Two plasmids were constructed by inserting either on-target or off target sequence in the region of multiple cloning sites downstream of Renilla luciferase gene in psiCheck plasmid (C8021, Promega). On-target plasmid contains 87 nucleotide short sequence that is the complete sequence match (CM) of human ANGPTL3 with the target site of the specific siNA under investigation. The off-target plasmid contains the sequence match of the seed region (SM) of the siNA under investigation. Seed region of siNA is referring to nucleotide #2 to #8 of the antisense strand numbering from 5’ end. To enhance the signal generated from off target effect, we put in 4 tandem repeats of siNA seed region in psiCheck. On the day of experiment siNA under investigation with serial dilutions were transfected into Cos7 cells along with constant concentration of either on-target plasmid or off-target plasmid. Cos-7 cells containing on- or off- target plasmid, treated with siNA were incubated for 72 hours. Following the incubation, Renilla luciferase assay was carried out according to Promega’s protocol. Prism software was used to plot on-target (CM) and off-target (SM) dose response curves. The results of the luciferase assay demonstrate that UNA at position 7 on AS (dsNA- 262) and xylo-F at position 6 on AS (dsNA-266) resulted in greatest decrease in seed-related, off-target activity, while conserving on-target potency in comparison to parent compound dsNA-240 (FIG. 6). Example 12. AAV-hANGPTL8 Mouse Study Design To evaluate certain ANGPTL8 siNA agents, an AAV-ANGPTL8 mouse model was used. Mice (n=4/group) were injected with an adeno-associated virus (AAV) expressing human ANGPTL8. Seven days after AAV injection, mice were subcutaneously administered a single dose (5 mg/kg) of an siNA. Twenty-eight days after administration, the animals were sacrificed; the right, lateral liver lobe of each animal was collected for RT-qPCR. For RT-qPCR, RNA was extracted using the Rneasy Mini Kit (Qiagen, 74106), according to the manufacturer’s protocol. RNA quantity and quality was analyzed with a NanoDrop™ Lite Spectrophotometer (Thermo Scientific), and cDNA was synthesized using the SuperScript IV VILO Master Mix (Invitrogen, 11756500), according to the manufacturer’s protocol. Gene expression was measured using TaqMan Fast Advanced Master Mix (Applied Biosystems, 4444964), and the following TaqMan Gene Expression assays (Applied Biosystems, 4331182): Actb (Mm00607939_s1), ANGPTL8 (Hs00218820_m1), B2m (Mm00437762_m1), and Hmbs (Mm01143545_m1). Actb, B2m, and Hmbs served as the endogenous control genes. RT-qPCR reactions were run on the QuantStudio™ 6 Pro Real-Time PCR System (Applied Biosystems). The RQ of gene expression was calculated via the 2−ΔΔCt (RQ) method. Results are presented as percent change in expression relative to the expression levels of negative vehicle control samples (FIG. 7). The results of the experiment showed that ANGPTL8 siNA dsNA-149, dsNA-150, dsNA-151, dsNA-152, dsNA-153 provided greater human ANGPTL8 RNA knockdown than comparative control siNA dsNA-285 (FIG. 7). Example 13. hANGPTL3 siNA and hANGPTL8 siNA Combination in human ANGPTL3/8 Double Knock-In Mouse Study To evaluate certain ANGPTL3 and ANGPTL8 siNA agents, a knock-in mouse model that expresses both human ANGPTL3 and ANGPTL8 (KI-ANGPTL3/8) was used. Mice (n=4/group) were subcutaneously administered a single dose (1 or 5 mg/kg) of one siNA or a combination of two siNAs. Thus, combination of ANGPTL3 and ANGPTL8 siNA was achieved by co-formulation of two siNAs in a single injection in this example. Blood was collected at various timepoints throughout the study for lipid quantification. Twenty-eight days after administration, the animals were sacrificed; the right, lateral liver lobe of each animal was collected for RT-qPCR and blood of each animal was collected for ELISA and lipid quantification.(Tables 20 and 21) Table 20 – Treatment Groups for Example 13.
Figure imgf000163_0001
Table 21 – Experimental Timeline for Example 13.
Figure imgf000163_0002
For RT-qPCR, RNA was extracted from the liver samples and cDNA was synthesized. Gene expression was measured using TaqMan Fast Advanced Master Mix (Applied Biosystems, 4444964), and the following TaqMan Gene Expression assays (Applied Biosystems, 4331182): Actb (Mm00607939_s1), ANGPTL3 (Hs00205581_m1), ANGPTL8 (Hs00218820_m1), B2m (Mm00437762_m1), and Hmbs (Mm01143545_m1). Actb, B2m, and Hmbs served as the endogenous control genes. RT-qPCR reactions were run on the QuantStudio™ 6 Pro Real-Time PCR System (Applied Biosystems). The RQ of gene expression was calculated via the 2−ΔΔCt (RQ) method. Results are presented as percent change in expression relative to the expression levels of negative vehicle control samples. Serum was separated from blood samples. Human ANGPTL3 protein levels in the serum were quantified using ELISA (R&D Systems, DANL30). Results are presented as percent change in expression of each treatment group at day 28 relative to each respective group’s expression level pre-dose. Triglycerides and total cholesterol levels in the serum were also quantified using a Roche Cobas 6000 c501 Chemistry Analyzer. Absolute lipid concentrations are reported. The results of the serum tests for total cholesterol and triglycerides are shown in FIG. 8. Some combinations of the ANGPTL3 and ANGPTL8 siNAs exhibited a greater reduction in total cholesterol levels as compared to the comparative control siNA and single siNA treatments on Day 7 post-dose (FIG. 8A). The combination of dsNA-244 and dsNA-152 showed a reduction in total cholesterol levels of 32%, while dsNA-244 alone only exhibited 19% reduction and dsNA-152 alone exhibited 29% reduction. This combination’s reduction was also significantly more than the comparative control dsNA-283 alone, which exhibited a reduction of only 25%. Similarly, the combination of dsNA-244 and dsNA-150 together reduced total cholesterol levels 43%, which is significantly more than dsNA-283 and greater than the reduction seen for dsNA-244 alone or dsNA-150 alone (28%). Finally, the combination of dsNA-241 and dsNA-150 resulted in a 37% reduction of total cholesterol levels, which is also significantly different from the comparative control and greater than the reduction seen with only dsNA-241 (19%) or dsNA-150. Like the results seen at Day 7 post-dose, certain combinations displayed reductions in total cholesterol levels on Day 14 post-dose (FIG. 8B). The combination treatment of dsNA- 241 and dsNA-152 had a reduction of total cholesterol levels of 40%, which is a greater percent reduction as compared to dsNA-241 alone (31%) and ds-NA-152 alone (37%). The combination of dsNA-244 and ds-152 also had a higher reduction of total cholesterol (40%) than either dsNA-244 (37%) or dsNA-152 alone. Finally, knockdown of both ANGPTL3 and ANGPTL8 by dsNA-244 and dsNA-150 respectively showed a 49% reduction in total cholesterol, as compared to dsNA-244 alone or to dsNA-150 alone (39%). All three combinations were significantly better at reducing total cholesterol than the comparative control dsNA-283 (24%). A similar trend for total cholesterol levels observed at Day 21 post-dose (FIG. 8C). The combination of dsNA-241 and dsNA-150 resulted in a 41% reduction in total cholesterol levels, while dsNA-241 alone had only 30% reduction and dsNA-150 alone had 28% reduction. The combination of dsNA-244 and dsNA-152 together had a reduction of 37% of total cholesterol levels, which is greater than only dsNA-244 (35%) or dsNA-152 (29%). Finally, the combination of dsNA-244 and dsNA-150 result in a likewise greater reduction in total cholesterol (39%) than either dsNA-244 or dsNA-150 alone. All of these combinations had a significantly greater reduction in total cholesterol levels than the comparative control dsNA-283 (21%). The increase in the ability of combinations of siNAs to reduce total cholesterol levels was also seen at the final timepoint of Day 28 post-dose (FIG. 8D). The combination of dsNA-241 and dsNA-150 reduced total cholesterol levels by 53% at Day 28, whereas dsNA- 241 alone and dsNA-150 alone both showed 41%. dsNA-244 and dsNA-150 together also had an increased reduction in total cholesterol levels (45%) as compared to dsNA-244 alone (43%) and ds-150 alone. Both combinations showed significantly greater reductions in total cholesterol than comparative control dsNA-283 alone (-32%). A similar effect is seen for the combination siNA treatments when serum triglyceride levels were analyzed at Day 14 post-dose (FIG. 8E). The combination of dsNA-241 and dsNA-152 resulted in a 48% reduction in serum triglyceride levels as compared to dsNA-241 alone (40%) or dsNA-152 alone (19%). Treatment with both dsNA-244 and dsNA-152 also had a grater reduction in triglyceride levels (30%) as compared to only dsNA-244 (24%) and dsNA-152. Finally, the combination of dsNA-244 and ds-150 resulted in a 31% reduction, which is greater than the reduction seen for dsNA-244 or dsNA-150 (23%) alone. These combinations all exhibited significantly higher reduction in triglyceride levels than the comparative control dsNA-283 alone (-21%). Example 14. Base modification of 5’-vinyl phosphonate end cap. In order to further enhance in vitro potency, the 5’-vinyl phosphonate end cap was modified. The vm(5mim)U modification was capable of slightly improving the in vitro potency as compared to the parent siNA (dsNA-244) when tested via the same method as in Example 3 (Table 22). Table 22 – In Vitro Activities of siNA with Base-Modified End Cap
Figure imgf000166_0002
Example 15. Production of Example 15 Monomer The Report of Production Process for intermediate 1b
Figure imgf000166_0001
Preparation of (1b): To a solution of 1a (100.0 g, 787.4 mmol) in HCONH2 (800 mL) was stirred at 150°C for 8 hour. LC-MS and TLC show SM was completely consumed. The mixture was cooling to 0~5°C, and was added water (400 mL), the solid was collected by filtration. The filter cake was rinsed with MeOH (2 x 500 mL), drying by rotary evaporator to give 1b (108.0 g, 80%yeild) as a yellow solid without further purified and used directly for the next step. ESI-LCMS m/z 165.0 [M+H]+. 1H NMR (400 MHz, DMSO-d6) δ 11.09 (s, 2H), 9.03 – 8.91 (m, 2H). The Report of Production Process for Example 15 Monomer from Intermediate 1b
Figure imgf000167_0001
Preparation of (2): To a solution of 1b (130.0 g, 793.7 mmol) in HMDS (1.3 L) was added (NH4)2SO4 (5.2 g, 39.7 mmol) and TMSCl (650.0 g, 5.9 mol) at room temperature. The mixture solution was stirred at 140°C for 4 h. The mixture solution was removed under reduced pressure, the residue was removed with toluene three times under reduced pressure. The residue in ACN (4.0 L) was added 1 (400.0 g, 793.7 mmol) at room temperature. The mixture solution was cooled to 0℃, the mixture was added TMSOTf (264.3 g, 1.2 mol) drop wise at 0 °C. The mixture solution was stirred at r.t for 3 h. TLC and LC-MS show SM was completely consumed. Then the solution was added Na2HCO3 (2000 mL), the product was extracted with EA, and was washed with water. Then washed the organic phase once with saturated brine and dried over by Na2SO4. Then the solution was concentrated under reduced pressure. The residue was isolated by silica gel column chromatography (eluent, PE/EA = 2:1~1:1). This resulted in 2 (180.0 g, 296.0 mmol, 36% yield) as a yellow solid. ESI-LCMS: m/z 609.1 [M+H]-. 1H NMR (400 MHz, CDCl3-d) δ 9.33 (s, 1H), 9.13 - 9.00 (m, 2H), 8.07 – 7.84 (m, 6H), 7.61 - 7.27 (m, 9H), 7.04 (s, 1H), 6.35 – 6.13 (m, 2H), 4.91 – 4.60 (m, 3H). Preparation of (3): To a solution of 2 (180.0 g, 296.0 mmol) was dissolved in 33% CH3NH2 in MeOH (1.5 L) at room temperature. The mixture solution was stirred at r.t for 4 h. TLC and LC-MS show 3 was completely consumed. Then the solution was removed under reduced pressure. The residue was purified by slurry with EA/PE = 1:1. This resulted in 3 (80.0 g) as a white solid. ESI-LCMS: m/z 318.1 [M+Na]-. 1H NMR (400 MHz, DMSO-d6) δ 11.28 (s, 1H), 9.26 – 9.04 (m, 2H), 6.55 (s, 1H), 5.38 – 4.15 (m, 5H), 3.79 – 3.41 (m, 3H). Preparation of (4): To a solution of 3 (75.0 g, 253.4 mmol) in dry DMF (750 mL) was added Ph2CO (60.0 g, 329.4 mmol) and NaHCO3 (1.4 g, 17.7 mmol) at room temperature. The mixture solution was stirred at 140℃ for 1 h. LC-MS show 4 was completely consumed. The mixture solution was concentrated under reduced pressure. This resulting product was purified by silica gel column chromatograph (eluent, DCM/MeOH = 30:1~10:1). This resulted in 6 (22.0 g, 27% yield) as a brown solid. ESI-LCMS: m/z 279.1 [M+H]-. Preparation of (5): To a solution of 4 (21.0 g, 75.5 mmol) in pyridine (200 mL) was added DMTrCl (30.6 g, 90.6 mmol) at 25 °C. The mixture solution was stirred at 25 °C for 2 h. LC-MS show SM was completely consumed. The mixture was diluted with EA (500 mL) and was washed with water. Then washed the organic phase once with saturated brine and dried over by Na2SO4. Then the solution was concentrated under reduced pressure. The residue was purified by silica gel column chromatograph (eluent, EA/PE = 1:1). This resulted in 5 (31.0 g, 53.4 mmol, 66% yield) as a yellow solid. ESI-LCMS: m/z 581.1 [M+H]-. 1H NMR (400 MHz, DMSO-d6) δ 9.38 (s, 1H), 9.23 (s, 1H), 7.28 – 6.98 (m, 10H), 6.77 – 6.63 (m, 4H), 6.09 – 6.01 (m, 1H), 5.40 – 5.32 (m, 1H), 4.48 – 4.30 (m, 2H), 3.75 – 3.661 (m, 6H), 3.12 – 3.04 (m, 1H), 2.81 – 2.73 (m, 1H). Preparation of (6): To the solution of 5 (30.0 g, 21.7 mmol) in dry DMF (300 mL) was added (MeO)2Mg (26.7 g, 310.3 mmol) at room temperature. And the reaction mixture was stirred at 100℃ for 2 hr. With ice-bath cooling, the reaction was quenched with saturated aq. NH4Cl and the product was extracted into EA (500 mL). The combined organic layer was washed with water and brine, dried over Na2SO4, and concentrated to give a residue which was purified by silica gel column chromatograph (eluent, EA/PE = 2:1). This resulted in 6 (23.0 g, 37.5 mmol, 72%) as a white solid. ESI-LCMS: m/z 611.1 [M-H]-. 1H NMR (400 MHz, DMSO-d6) δ 12.04 (s, 1H), 9.16 (s, 1H), 9.09 (s, 1H), 7.46 – 6.46 (m, 14H), 5.02 – 4.87 (m, 1H), 4.48 – 3.86 (m, 3H), 3.71 (s, 6H), 3.35 (s, 3H), 3.18 (s, 2H). Preparation of (7): To a solution of 6 (18.0 g, 29.4 mmol) in DMF (180 mL) was added imidazole (4.0 g, 58.8 mmol) and TBSCl (5.3 g, 35.3 mmol) at room temperature. The mixture solution was stirred at 25 °C for 15 h. LC-MS show 6 was completely consumed. Then the mixture solution was added water, and the product was extracted with EA. And the organic phase was washed with water and saturated brine and dried over by Na2SO4. Then the solution was concentrated under reduced pressure. This resulted in crude 7 (19.5 g) as a white solid without further purified and used directly for the next step. ESI-LCMS: m/z 725.2 [M- H]-. Preparation of (8): To a solution of crude 7 (19.5 g) was dissolved in 4% DCA in DCM (190 mL) at room temperature. The mixture solution was stirred at r.t for 20 min. LC- MS show SM was completely consumed. Then the solution was diluted with NaHCO3.aq (200 mL) and the product was extracted with DCM and washed with water. Then washed the organic phase once with saturated brine and dried over by Na2SO4. Then the solution was concentrated under reduced pressure. The residue was purified by silica gel column chromatograph (eluent, DCM/MeOH = 50:1). This resulted in giving 8 (6.5 g, 15.3 mmol, 40% yield over 2 steps) as a white solid. ESI-LCMS: m/z 425.1 [M+H]-. 1H NMR (400 MHz, DMSO-d6) δ 11.95 (s, 1H), 9.12 – 9.13 (m, 2H), 6.49 (s, 1H), 4.59 – 4.16 (m, 3H), 3.68 – 3.28 (m, 3H), 3.19 (s, 3H), 0.80 (s, 9H), 0.03 - -003 (m, 6H). Preparation of (9): To a solution of 8 (6.0 g, 14.1 mmol) and EDCI (8.1 g, 42.3 mmol) in DMSO (60 mL) was added Pyridine (1.3 g, 15.6 mmol) and TFA (888 mg, 7.8 mmol) at 0- 5℃. The mixture was stirred at room temperature for 2 h, LC-MS showed the 8 was consumed completely and the reaction was quenched by water at 0° to obtain a mixture was diluted with EA which was separated by a funnel and aqueous phase was extracted with EA (100 mL*2) and was washed with water. Then washed the organic phase once with saturated brine and dried over by Na2SO4. Then the solution was concentrated under reduced pressure. This resulted in a crude 4 (6.5 g) as yellow solid was used directly in the next step. ESI- LCMS: m/z 423.1 [M+H]-. Preparation of (10): To a solution of 9 (6.5 g) in toluene (65 mL) was added KOH (1.3 g, 24.0 mmol) and POM ester (8.1 g, 12.9 mmol) at room temperature under N2 atmosphere. The reaction mixture was stirred at room temperature. for 2.5 h. LC-MS showed the 9 was consumed completely. Then the solution was diluted with EA (100 mL) and the product was extracted with EA and washed with water. Then washed the organic phase once with saturated brine and dried over by Na2SO4. Then the solution was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent, PE: EA = 1:1~1:2) to give 10 (5.0 g, 6.9 mmol E/Z = 76:23 by PNMR) as a yellow solid. ESI- LCMS: m/z 729.2 [M+H]-. 1H NMR (400 MHz, DMSO-d6) δ 12.09 (s, 1H), 9.18 (s, 2H), 6.80 – 6.43 (m, 2H), 6.18 – 5.91 (m, 1H), 5.68 – 5.47 (m, 4H), 4.93 – 4.23 (m, 3H), 3.40 – 3.20 (m, 3H), 1.23 – 1.05 (m, 18H), 0.89 (s, 9H), 0.08 (s, 6H). 31P NMR (600 MHz, DMSO- d6) δ 16.887, 14.34. Preparation of (11): A solution of 10 (5.0 g, 6.9 mmol) in HCOOH (25 mL) and water (25 mL) was stirred at room temperature overnight. LC-MS showed 10 was consumed completely. Con. NH4OH was added to the mixture at 0°C to quench the reaction until the pH = 7.5. The product was extracted into ethyl acetate (100 mL). The product was extracted with EA and was washed with water. Then washed the organic phase once with saturated brine and dried over by Na2SO4. Then the solution was concentrated under reduced pressure. The residue was purified twice by silica gel column chromatography (eluent, DCM: MeOH = 50:1~30:1) to give 11 (1.6 g, 2.6 mmol, 18% yield over 3 steps, >99%purity by PNMR) as a white solid. ESI-LCMS: m/z 616.1 [M+H]-. 1H NMR (400 MHz, DMSO-d6) δ 12.08 (s, 1H), 9.17 (s, 2H), 6.89 – 6.50 (m, 2H), 6.12 – 5.91 (m, 1H), 5.65 – 5.51 (m, 4H), 5.43 – 5.23 (m, 1H), 4.58 – 4.23 (m, 3H), 3.37 (s, 3H), 1.12 (s, 18H). 31P NMR (600 MHz, DMSO-d6) δ 17.60. Preparation of (Example 15 monomer): To a solution of 11 (1.6 g, 2.6 mmol) in DCM (20 mL) was added DCI (261 mg, 2.2 mmol) and CEP[N(iPr)2]2 (945 mg, 3.1 mmol) at r.t at N2. The mixture was stirred at r.t at N2 for 2 h. LC-MS showed all precursor was consumed completely. The mixture was added NaHCO3 aqueous (100 mL) and extracted with DCM (100 mL). Then the organic layer was washed with water (200 mL) and brine (200 mL) and dried over Na2SO4. The solution was filtered, and the filtrate was concentrated to give the crude. The crude was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, C18 silica gel; mobile phase, CH3CN/H2O (0.05% NH4HCO3) = 3/2 increasing to CH3CN/H2O (0.05% NH4HCO3) = 1/0 within 20 min, the eluted product was collected at CH3CN/ H2O (0.05% NH4HCO3) = 1/0; Detector, UV 254 nm. This resulted in Example 15 monomer (1.6 g, 1.9 mmol, 75% yield, 99% purity) as a white solid. ESI- LCMS: m/z 815.2 [M+H]-. 1H NMR (400 MHz, DMSO-d6) δ 12.09 (s, 1H), 9.27 – 9.06 (m, 2H), 6.98 – 6.54 (m, 2H), 6.21 – 6.00 (m, 1H), 5.67 – 5.45 (m, 4H), 4.92 – 4.35 (m, 3H), 3.89 – 3.51 (m, 4H), 3.43 – 3.34 (m, 3H), 2.87 – 2.74 (m, 2H), 1.24 – 1.06 (m, 30H). 31P NMR (600 MHz, DMSO-d6) δ 148.79, 147.97, 17.28, 16.92. Example 16. Production of Example 16 Monomer.
Figure imgf000171_0001
Preparation of (2): To a solution of 1 (100.0 g, 387.3 mmol) in ACN (1.0 L). Then the mixture was added Ceric ammonium nitrate (106.2 g, 193.6 mmol) and I2 (59.0 g, 232.4 mmol). The mixture was stirred at r.t for 3 h, the solid was collected by filtration. The filter cake was rinsed with ACN (2 x 500 mL), drying by rotary evaporator to give crude 2 (120.0 g) as a white solid without further purified and used directly for the next step. ESI-LCMS: m/z 384.9 [M+H]+. Preparation of (3): To a solution of 2 (120.0 g, 312.5 mmol) in DMF (1.0 L). Then the mixture was added Imidazole (63.8 g, 937.6 mmol), TBSCl (98.9 g, 656.3 mmol) under N2 atmosphere. The mixture was stirred at room temperature for 16 h. Then the solution was diluted with EA. And the organic phase was washed with water and saturated brine and dried over by Na2SO4. Then the solution was concentrated under reduced pressure. The residue was purified by silica gel column chromatograph (eluent, PE/EA = 100:1~ 1:1). This resulted in 3 (150.0 g, 245.1 mmol, 78.4% yield over two steps) as a solid. ESI-LCMS: m/z 613.1 [M+H]+. Preparation of (4): To a stirred solution of 3 (150.0 g, 245.1 mmol) and (E)-2- ethoxyvinylboronic acid pinacol ester (97.1 g, 490.2 mmol) in 1, 4-Dioxane (1.5 L) and H2O (187 mL) was added Cs2CO3 (175.7 g, 539.2 mmol), [PdCl2 (dppf)] CH2Cl2 (19.8 g, 24.5 mmol). The resulting mixture was stirred overnight at 50°C under argon atmosphere. The reaction was cooled to room temperature and was extracted with EA (2 x 1.0 L). And the organic phase was washed with water and saturated brine and dried over by Na2SO4. Then the solution was concentrated under reduced pressure. The residue was purified by silica gel column chromatograph (eluent, PE/EA = 100:1~ 1:1). This resulted in giving 4 (107.0 g, 192.1 mmol) as a solid. ESI-LCMS: m/z 557.2 [M+H]+. Preparation of (5): To the solution of 4 (107.0 g, 192.1 mmol) in THF (1.0 L) followed by the addition of Hg(OAc)2 (110.0 g, 345.8 mmol) was dissolved in ice-water (500 mL ). The resulting mixture was stirred for 6 h at 0°C. The reaction was quenched with sat. NH4Cl (aq.) at 0°C. The resulting mixture was extracted with EA. And the organic phase was washed with water and saturated brine and dried over by Na2SO4. Then the solution was concentrated under reduced pressure. The residue was purified by silica gel column chromatograph (eluent, PE/EA = 100:1~ 1:1). This resulted in 5 (41.0 g, 77.5 mmol) as a solid. ESI-LCMS: m/z 529.2 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ: 11.59 (s, 1H), 9.59 (d, J = 1.3 Hz, 1H), 7.56 (s, 1H), 5.85 (d, J = 5.5 Hz, 1H), 4.23 (q, J = 6.9, 5.6 Hz, 1H), 3.94 – 3.75 (m, 2H), 3.75 – 3.65 (m, 1H), 3.40 – 3.32 (m, 4H), 0.89 (t, J = 4.0 Hz, 18H), 0.16 – 0.03 (m, 12H). Preparation of (6): To a stirred solution 5 (41.0 g, 77.5 mmol) in dry EtOH (800 mL) was added KCN (756 mg, 11.6 mmol) and Tos-MIC (16.6 g, 85.2 mmol) at room temperature for 30 min. The reaction was concentrated under reduced pressure. The residue was dissolved in 7N NH3 ^MeOH (400 mL) in a High-pressure kettle. The resulting mixture was stirred for 4 h at 100°C. The reaction was quenched with sat. NH4Cl (aq.) at 0°C. The resulting mixture was extracted with EA. And the organic phase was washed with water and saturated brine and dried over by Na2SO4. Then the solution was concentrated under reduced pressure. The residue was purified by silica gel column chromatograph (eluent, PE/EA = 100:1~ 1:1). The residue was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, C18 silica gel; mobile phase, CH3CN/H2O (0.5% NH4HCO3) = 1/3 increasing to CH3CN/H2O (0.5% NH4HCO3) = 1/0 within 25 min, the eluted product was collected at CH3CN/ H2O (0.5% NH4HCO3) = 58/42; Detector, UV 254 nm. This resulted in giving 6 (10.3 g, 18.2 mmol) as a solid. ESI-LCMS: m/z 567.2 [M+H]+. Preparation of (7): To a solution of 6 (10.3 g, 18.2 mmol) in THF (100 mL) and was added TFA/H2O=1/1 (40 ml). The mixture was stirred at 0°C for 1 h. TLC showed 6 was consumed completely. NaHCO3 (aq.) was added to the mixture at 0°C. Then the solution diluted with EA. The organic layer was washed with brine, dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent, DCM: MeOH = 100:1 ~ 30:1). The residue was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, C18 silica gel; mobile phase, CH3CN/H2O (0.5% NH4HCO3) = 1/3 increasing to CH3CN/H2O (0.5% NH4HCO3) = 1/0 within 25 min, the eluted product was collected at CH3CN/ H2O (0.5% NH4HCO3) = 58/42; Detector, UV 254 nm. This resulted in giving 7 (3.0 g, 6.6 mmol) as a white solid. ESI-LCMS: m/z 453.3 [M+H]+. Preparation of (8): To a stirred mixture of 7 (3.0 g, 6.6 mmol) in DCM (30 mL) was added TEA (1.3 g, 13.2 mmol) and TrTCl (2.2 g, 7.9 mmol) at room temperature under N2 atmosphere. The resulting mixture was stirred for 2 h at room temperature under argon atmosphere. The reaction was quenched by the addition of sat. NaHCO3 (aq.). The resulting mixture was extracted with EA. The organic layer was washed with brine, dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent, PE (0.5% TEA)/ EA =50:1~ 2:3). This resulted in giving 8 (2.8 g, 4.0 mmol, 61% yield) as an off-white solid. ESI-LCMS: m/z 695.2 [M+H]+. Preparation of (9): To a stirred mixture of 8 (2.8 g, 4.0 mmol) in DMSO (30 mL) was added EDCI (2.3 g, 12.0 mmol), pyridine (0.35 g, 4.4 mmol) and was dropped TFA (0.25 g, 2.2 mmol) at 0°C under N2 atmosphere. The resulting mixture was stirred for 2 h at room temperature under argon atmosphere. The reaction was quenched by the addition of sat. NaHCO3 (aq.). The resulting mixture was extracted with EA. The organic layer was washed with brine, dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. This resulted in crude 9 (4.0 g) as yellow solid used directly for the next step. ESI-LCMS: m/z 711.2 [M+H2O]+. Preparation of (10): To a stirred mixture of 9 (4.0 g, 4.0 mmol) in Toluene (30 mL) was added KOH (0.41 g, 6.8 mmol) and POM (2.8 g, 4.4 mmol) at 0°C under N2 atmosphere. The resulting mixture was stirred for 2 h at room temperature under argon atmosphere. The reaction was quenched by the addition of sat. NaHCO3 (aq.). The resulting mixture was extracted with EA. The organic layer was washed with brine, dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, C18 silica gel; mobile phase, CH3CN/H2O (0.5% NH4HCO3) = 1/3 increasing to CH3CN/H2O (0.5% NH4HCO3) = 1/0 within 25 min, the eluted product was collected at CH3CN/ H2O (0.5% NH4HCO3) = 85/15; Detector, UV 254 nm. This resulted in giving 10 (3.1 g, 3.1 mmol) as a white solid. ESI-LCMS: m/z 999.2 [M+H]+. Preparation of (11): To a stirred mixture of 10 (3.1 g, 3.1 mmol) in THF (6 mL) and MeOH (4 mL) was added Formic acid (15 mL) and H2O (15 mL) at 0°C. The resulting mixture was stirred for 1 h at 0°C. The reaction was quenched by the addition of sat. NaHCO3 (aq.). The resulting mixture was extracted with EA. The organic layer was washed with brine, dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, C18 silica gel; mobile phase, CH3CN/H2O (0.5% NH4HCO3) = 1/3 increasing to CH3CN/H2O (0.5% NH4HCO3) = 1/0 within 25 min, the eluted product was collected at CH3CN/ H2O (0.5% NH4HCO3) = 75/25; Detector, UV 254 nm. The residue was purified by SFC. This resulted in giving 11 (1.4 g, 2.1 mmol) as a white solid. ESI-LCMS: m/z 643.5 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ: 11.78 (s, 1H), 11.44 (s, 1H), 7.54 (d, J = 20.3 Hz, 2H), 6.90 – 6.59 (m, 2H), 6.10 (ddd, J = 22.2, 17.1, 1.5 Hz, 1H), 5.80 (d, J = 4.0 Hz, 1H), 5.62 (dt, J = 13.7, 1.0 Hz, 4H), 5.53 (d, J = 6.7 Hz, 1H), 4.39 – 4.30 (m, 1H), 3.89 (dd, J = 5.4, 4.0 Hz, 1H), 3.45 (s, 2H), 3.38 (s, 3H), 3.33 (s, 2H), 1.14 (d, J = 3.8 Hz, 18H); 31P NMR (162 MHz, DMSO-d6) δ: 17.22 . Preparation of (Example 16 Monomer): To a solution of 11 (1.4 g, 2.1 mmol) in DCM (20 mL) was added DCI (210 mg, 1.7 mmol) and CEP[N(iPr)2]2 (0.76 g, 2.5 mmol) under N2. The mixture was stirred at 25 °C for 2.5 h. LCMS showed 11 was consumed completely. The product was extracted with DCM. The organic layer was washed with H2O and brine. Then the solution was concentrated under reduced pressure and the residue was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, C18 silica gel; mobile phase, CH3CN/H2O (0.5% NH4HCO3) = 1/3 increasing to CH3CN/H2O (0.5% NH4HCO3) = 1/0 within 25 min, the eluted product was collected at CH3CN/ H2O (0.5% NH4HCO3) = 1/0; Detector, UV 254 nm. This resulted in Example 16 Monomer (600 mg, 6.4 mmol) as a white solid. ESI-LCMS: m/z 843.2 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ: 11.78 (s, 1H), 11.45 (s, 1H), 7.57 (d, J = 15.1 Hz, 2H), 6.81 – 6.73 (m, 1H), 6.16 (ddt, J = 21.8, 17.1, 1.3 Hz, 1H), 5.82 – 5.73 (m, 1H), 5.67 – 5.55 (m, 4H), 4.58 – 4.44 (m, 1H), 4.35 (ddt, J = 43.0, 11.0, 5.6 Hz, 1H), 4.11 (q, J = 4.5 Hz, 1H), 3.85 – 3.67 (m, 1H), 3.65 – 3.53 (m, 1H), 3.38 (q, J = 17.0, 16.0 Hz, 8H), 2.86 – 2.74 (m, 2H), 1.17 – 1.10 (m, 30H); 31P NMR (162 MHz, DMSO-d6) δ: 149.63 , 149.07 , 16.92 , 16.62. Example 17. Production of Example 17 Monomer.
Figure imgf000176_0001
Preparation of (2): To a solution of 1 (300.0 g, 1.1 mol) in DMF (3.0 L) was added imidazole (408.0 g, 6.0 mol) and TBSCl (453.0 g, 3.0 mol) at room temperature. The mixture solution was stirred at 50°C for 15 h. LC-MS show 1 was completely consumed. Then the mixture solution was added water, and the product was extracted with EA. And the organic phase was washed with water and saturated brine and dried over by Na2SO4. Then the solution was concentrated under reduced pressure. The residue was purified by silica gel column chromatograph (eluent, PE/EA = 5:1). This resulted in giving 2 (500.0 g, 1.0 mol, 90% yield) as a white solid. ESI-LCMS: m/z 487.3 [M+H]-. 1H NMR (400 MHz, DMSO-d6) δ 11.30 (s, 1H), 7.71 (d, J = 8.1 MHz , 1H), 5.71 (d, J = 8.2 MHz , 1H), 5.47 (d, J = 12.1 MHz , 1H), 4.19 – 4.09 (m, 1H), 3.87 – 3.54 (m, 4H), 3.28 (s, 3H), 0.86 – 0.74 (m, 18H), 0.02- -0.01 (m, 12H). Preparation of (3): To a solution of 2 (530.0 g, 1.1 mol) in dry THF (2.5 L) was added 2M LDA (1.7 L, 3.3 mol) drop wise at -78oC. The mixture solution was stirred at -78°C for 1 h. The mixture was added I2 (1.1 kg, 4.4 mol) drop wise at -78°C. The mixture solution was stirred at -78°C for 2 h. TLC and LC-MS show SM was completely consumed. Then the solution was added NH4Cl.aq (500 mL), the product was extracted with EA, and was washed with water. Then washed the organic phase once with saturated brine and dried over by Na2SO4. Then the solution was concentrated under reduced pressure. The residue was to give crude 3 (635.0 g) as a brown solid without further purified and used directly for the next step. ESI-LCMS: m/z 613.13 [M+H]-. Preparation of (4): To a solution of 3 (635.0 g) in DMF (6.5 L) was added NaN3 (107.0 g, 1.7 mol) at 25 °C. The mixture solution was stirred at r.t for 15 h. LC-MS show SM was completely consumed. Then the solution was added H2O (500 mL), the product was extracted with EA, and was washed with water. Then washed the organic phase once with saturated brine and dried over by Na2SO4. Then the solution was concentrated under reduced pressure. The residue was to give crude 4 (580.0 g) as a yellow solid without further purified and used directly for the next step. ESI-LCMS: m/z 528.1 [M+H]-. Preparation of (5): To a solution of 4 (580.0 g) in THF (5.6 L) was added 10% Pd/C (58.4 g) at 25 °C. The mixture solution was stirred at r.t at H2 for 5 h. LC-MS show SM was completely consumed. Filtered out the solid, and the solution was concentrated under reduced pressure. The residue was purified by silica gel column chromatograph (eluent, PE/EA = 2:1~1:1). This resulted in giving 5 (60.0 g, 119.8 mmol, 12% yield over 3 steps) as a yellow solid. ESI-LCMS: m/z 503.0 [M+H]-. 1H NMR (400 MHz, DMSO-d6) δ 10.65 – 10.54 (m, 1H), 6.59 (s, 2H), 6.13 – 6.02 (m , 1H), 4.70 (d, J = 4.0 MHz , 1H), 4.42 – 4.02 (m, 2H), 3.89 – 3.66 (m, 3H), 3.26 (s, 3H), 0.92 – 0.82 (m, 18H), 0.15- 0.03 (m, 12H). Preparation of (6): To a solution of 4 (60.0 g, 119.8 mmol) in THF/AcOH/H2O =1:1:1 (600 mL) was added NaNO2 (12.4 g, 179.8 mmol) at 25 °C. The mixture solution was stirred at 50oC for 1 h. LC-MS show SM was completely consumed. The solution was diluted with EA (600 mL) and washed with water. Then washed the organic phase once with saturated brine and dried over by Na2SO4. Then the solution was concentrated under reduced pressure. The residue was to give crude 4 (60.0 g) as a yellow solid without further purified and used directly for the next step. ESI-LCMS: m/z 531.0 [M+H]-. Preparation of (7): To a solution of 6 (60.0 g) in THF/MeOH/H2O =1:1:1 (600 mL) was added Na2S2O4 (89.64 g, 515.8 mmol) at 25 °C. The mixture solution was stirred at r.t for 1 h. LC-MS show SM was completely consumed. The solution was diluted with EA (600 mL) and washed with water. Then washed the organic phase once with saturated brine and dried over by Na2SO4. Then the solution was concentrated under reduced pressure. The residue was purified by silica gel column chromatograph (eluent, PE/EA = 1:1~1:2). This resulted in giving 7 (31.0 g, 60.0 mmol, 50% yield over 2 steps) as a yellow solid. ESI- LCMS: m/z 517.2 [M+H]-. 1H NMR (400 MHz, DMSO-d6) δ 10.87 (s, 1H), 6.09 (d, J = 8.0 MHz, 1H), 5.74 (s, 2H), 4.48 – 4.37 (m, 1H), 4.12 – 3.67 (m, 4H), 3.25 (s, 3H), 3.23 – 2.97 (m, 1H), 0.88 (s, 18H), 0.10 – 0.05 (m, 12H). Preparation of (8): To a solution of 7 (30.0 g, 58.0 mmol) in MeOEtOH (300 mL) was added BrCN (29.9 L, 250.0 mmol) at 0oC. The mixture solution was stirred at 25°C for 15 min. The mixture was added water, and the mixture was filtered, and the filter residue was concentrated. The residue was dissolved in DMF (300 mL). The mixture was supplied with NH3 gas, and the mixture solution was stirred at 25°C for 15 h. TLC and LC-MS show SM was completely consumed. Then the solution was added NH4Cl.aq (500 mL), the product was extracted with EA, and was washed with water. Then washed the organic phase once with saturated brine and dried over by Na2SO4. Then the solution was concentrated under reduced pressure. This resulted was purified by silica gel column chromatograph (eluent, PE: EA = 2:1~1:2). The residue was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, C18 silica gel; mobile phase, CH3CN/H2O (0.05% NH4HCO3) = 1/3 increasing to CH3CN/H2O (0.05% NH4HCO3) = 4/1 within 25 min, the eluted product was collected at CH3CN/ H2O (0.05% NH4HCO3) = 3/2; Detector, UV 254 nm. This resulted in giving 8 (4.5 g, 8.3 mmol, 14% yield) as a yellow solid. ESI-LCMS: m/z 540.2 [M+H]-. 1H NMR (400 MHz, DMSO-d6) δ 11.36 (s, 1H), 10.65 (s, 1H), 6.78 – 5.87 (m, 3H), 4.66 – 4.46 (m, 2H), 3.88 – 3.54 (m, 3H), 3.27 (s, 3H), 1.75 (s, 1H), 0.95 – 0.77 (m, 18H), 0.14 – -0.05 (m, 12H). Preparation of (9): To a solution of 8 (4.5 g, 8.3 mmol) in DMF (45 mL) was added Bz2O (2.1 g, 9.1 mmol) and DMAP (101 mg, 0.8 mmol) at room temperature. The mixture was stirred at r.t for 4 h. TLC and LC-MS show the reaction was complete. The mixture was poured into NaHCO3.aq, the product was extracted with EA. Then washed the organic phase once with saturated brine and dried over by Na2SO4. Then the solution was concentrated under reduced pressure. This resulted was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, C18 silica gel; mobile phase, CH3CN/H2O (0.05% NH4HCO3) = 2/3 increasing to CH3CN/H2O (0.05% NH4HCO3) = 1/0 within 30 min, the eluted product was collected at CH3CN/ H2O (0.05% NH4HCO3) = 4/1; Detector, UV 254 nm. This resulted in 9 (3.3 g, 5.1 mmol, 61% yield, 98% purity) as a white solid. ESI-LCMS: m/z 646.2 [M+H]-. 1H NMR (400 MHz, DMSO-d6) δ 12.70 (s, 1H), 11.83 (s, 1H), 11.13 (s, 1H), 8.12 – 7.44 (m, 5H), 6.15 – 6.01 (m, 1H), 4.81 – 4.50 (m, 2H), 3.91 – 3.25 (m, 6H), 0.98 – 0.77 (m, 18H), 0.17 – -0.05 (m, 12H). Preparation of (10): To a solution of 9 (3.3 g, 5.1 mmol) in THF (10 mL) and was added TFA/H2O=1/1 (20 ml). The mixture was stirred at 0°C for 1 h. TLC showed 9 was consumed completely. NaHCO3 (aq.) was added to the mixture at 0°C. Then the solution diluted with EA. The organic layer was washed with brine, dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent, PE: EA = 3:1~1:1). The residue was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, C18 silica gel; mobile phase, CH3CN/H2O (0.5% NH4HCO3) = 2/3 increasing to CH3CN/H2O (0.5% NH4HCO3) = 1/0 within 25 min, the eluted product was collected at CH3CN/ H2O (0.5% NH4HCO3) = 58/42; Detector, UV 254 nm. This resulted in 10 (1.9 g, 3.6 mmol) as a white solid. ESI-LCMS: m/z 530.3 [M+H]-. 1H NMR (400 MHz, DMSO-d6) δ 12.74 (s, 1H), 11.83 (s, 1H), 11.17 (s, 1H), 8.08 – 7.50 (m, 5H), 6.15 – 6.01 (m, 1H), 4.84 – 4.45 (m, 3H), 3.84 – 3.22 (m, 6H), 0.90 (s, 18H), 0.12 – 0.0 (m, 6H). Preparation of (11): To a solution of 10 (1.9 g, 3.6 mmol) and EDCI (2.1 g, 10.7 mmol) in DMSO (20 mL) was added pyridine (310 mg, 3.9 mmol) and TFA (223 mg, 1.9 mmol) at 0-5℃. The mixture was stirred at room temperature for 2 h, LC-MS showed the 10 was consumed completely and the reaction was quenched by water at 0° to obtain a mixture was diluted with EA which was separated by a funnel and aqueous phase was extracted with EA (100 mL*2) and was washed with water. Then washed the organic phase once with saturated brine and dried over by Na2SO4. Then the solution was concentrated under reduced pressure. This resulted in a crude 11 (1.8 g) as yellow solid, which was used directly to next step. ESI-LCMS: m/z 528.3 [M+H]-. Preparation of (12): To a solution of 11 (1.8 g) in toluene (20 mL) was added KOH (323 g, 5.8 mmol) and POM seter (2.4 g, 3.7 mmol) at room temperature under N2 atmosphere. The reaction mixture was stirred at room temperature for 2.5 h. LC-MS showed the 11 was consumed completely. Then the solution was diluted with EA (100 mL) and the product was extracted with EA and washed with water. Then washed the organic phase once with saturated brine and dried over by Na2SO4. Then the solution was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent, PE: EA = 2:1~1:1). The residue was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, C18 silica gel; mobile phase, CH3CN/H2O (0.5% NH4HCO3) = 2/3 increasing to CH3CN/H2O (0.5% NH4HCO3) = 1/0 within 25 min, the eluted product was collected at CH3CN/ H2O (0.5% NH4HCO3) = 4/1; Detector, UV 254 nm. This resulted in 12 (600 mg, 0.7 mmol, E/Z > 99% by PNMR) as a white solid. ESI-LCMS: m/z 834.2 [M-H]-. 1H NMR (400 MHz, DMSO-d6) δ 12.73 (s, 1H), 11.81 (s, 1H), 11.18 (s, 1H), 8.13 – 7.47 (m, 5H), 6.98 – 6.67 (m, 1H), 6.21 – 6.00 (m, 2H), 5.67 – 5.48 (m, 4H), 4.69 – 4.28 (m, 3H), 3.34 (s, 3H), 1.13 – 1.09 (m, 18H), 0.89 (s, 9H), 0.11 – 0.06 (m, 6H). 31P NMR (600 MHz, DMSO-d6) δ 17.28. Preparation of (13): A solution of 12 (600 mg, 0.7 mmol) in HCOOH (3 mL) and water (3 mL) was stirred at room temperature overnight. LC-MS showed 12 was consumed completely. Con. NH4OH was added to the mixture at 0°C to quench the reaction until the pH = 7.5. The product was extracted into ethyl acetate (100 mL). The product was extracted with EA and was washed with water. Then washed the organic phase once with saturated brine and dried over by Na2SO4. Then the solution was concentrated under reduced pressure. The residue was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, C18 silica gel; mobile phase, CH3CN/H2O (0.5% NH4HCO3) = 2/3 increasing to CH3CN/H2O (0.5% NH4HCO3) = 1/0 within 25 min, the eluted product was collected at CH3CN/ H2O (0.5% NH4HCO3) = 4/1; Detector, UV 254 nm. This resulted in 13 (330 mg, 0.4 mmol, 57% yield, >98%purity by HPLC & PNMR) as a white solid. ESI-LCMS: m/z 720.2 [M-H]-. 1H NMR (400 MHz, DMSO-d6) δ 12.67 (s, 1H), 11.78 (s, 1H), 11.18 (s, 1H), 8.13 – 7.45 (m, 5H), 6.99 – 6.77 (m, 1H), 6.21 – 6.00 (m, 2H), 5.64 – 5.28 (m, 5H), 4.50 (s, 2H), 4.33 (s, 1H), 3.37 (s, 3H), 1.15 – 1.02 (m, 18H). 31P NMR (600 MHz, DMSO-d6) δ 17.91. Preparation of (Example 17 Monomer): To a solution of 13 (330 mg, 0.4 mmol) in DCM (20 mL) was added DCI (46 mg, 0.4 mmol) and CEP[N(iPr)2]2 (165 mg, 0.5 mmol) under N2. The mixture was stirred at 25 °C for 2.5 h. LCMS showed 13 was consumed completely. The product was extracted with DCM. The organic layer was washed with H2O and brine. Then the solution was concentrated under reduced pressure and the residue was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, C18 silica gel; mobile phase, CH3CN/H2O (0.5% NH4HCO3) = 1/3 increasing to CH3CN/H2O (0.5% NH4HCO3) = 1/0 within 25 min, the eluted product was collected at CH3CN/ H2O (0.5% NH4HCO3) = 1/0; Detector, UV 254 nm. This resulted in Example 17 Monomer (310 mg, 0.3 mmol) as a white solid. ESI-LCMS: m/z 920.1 [M-H]-. 1H NMR (400 MHz, DMSO- d6) δ 12.68 (s, 1H), 11.78 (s, 1H), 11.19 (s, 1H), 8.14 – 7.48 (m, 5H), 7.10 – 6.71 (m, 1H), 6.26 – 6.03 (m, 2H), 5.67 – 5.48 (m, 4H), 4.79 – 4.40 (m, 3H), 3.89 – 3.35 (m, 7H), 2.87 – 2.72 (m, 2H), 1.25 – 1.02 (m, 30H). 31P NMR (600 MHz, DMSO-d6) δ 149.24, 148.66, 17.64, 17.26. * * * * These examples are provided for illustrative purposes only and not to limit the scope of the claims provided herein. While certain embodiments have been illustrated and described, it should be understood that changes and modifications can be made therein in accordance with ordinary skill in the art without departing from the technology in its broader aspects as defined in the following claims. The embodiments, illustratively described herein may suitably be practiced in the absence of any element or elements, limitation or limitations, not specifically disclosed herein. Thus, for example, the terms “comprising,” “including,” “containing,” etc. shall be read expansively and without limitation. Additionally, the terms and expressions employed herein have been used as terms of description and not of limitation, and there is no intention in the use of such terms and expressions of excluding any equivalents of the features shown and described or portions thereof, but it is recognized that various modifications are possible within the scope of the claimed technology. Additionally, the phrase “consisting essentially of” will be understood to include those elements specifically recited and those additional elements that do not materially affect the basic and novel characteristics of the claimed technology. The phrase “consisting of” excludes any element not specified. In addition, where features or aspects of the disclosure are described in terms of Markush groups, those skilled in the art will recognize that the disclosure is also thereby described in terms of any individual member or subgroup of members of the Markush group. As will be understood by one skilled in the art, for any and all purposes, particularly in terms of providing a written description, all ranges disclosed herein also encompass any and all possible subranges and combinations of subranges thereof, inclusive of the endpoints. Any listed range can be easily recognized as sufficiently describing and enabling the same range being broken down into at least equal halves, thirds, quarters, fifths, tenths, etc. As a non-limiting example, each range discussed herein can be readily broken down into a lower third, middle third and upper third, etc. As will also be understood by one skilled in the art all language such as “up to,” “at least,” “greater than,” “less than,” and the like, include the number recited and refer to ranges which can be subsequently broken down into subranges as discussed above. Finally, as will be understood by one skilled in the art, a range includes each individual member. All publications, patent applications, issued patents, and other documents referred to in this specification are herein incorporated by reference as if each individual publication, patent application, issued patent, or other document was specifically and individually indicated to be incorporated by reference in its entirety. Definitions that are contained in text incorporated by reference are excluded to the extent that they contradict definitions in this disclosure.

Claims

WHAT IS CLAIMED IS: 1. A short interfering nucleic acid (siNA) molecule comprising: (a) a sense strand comprising a first nucleotide sequence that is at least about 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% identical to an RNA corresponding to a target gene, wherein the first nucleotide sequence: (i) is 15 to 30 nucleotides in length; and (ii) comprises 15 or more modified nucleotides independently selected from a 2’-O-methyl nucleotide and a 2’-fluoro nucleotide, wherein at least one modified nucleotide is a 2’-O-methyl nucleotide and the nucleotide at position 3, 5, 7, 8, 9, 10, 11, 12, 14, 17, and/or 19 from the 5’ end of the first nucleotide sequence is a 2’-fluoro nucleotide or wherein at least one modified nucleotide is a 2’-O-methyl nucleotide and at least one modified nucleotide is a 2’-fluoro nucleotide; and an antisense strand comprising a second nucleotide sequence that is at least about 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% complementary to the RNA corresponding to the target gene, wherein the second nucleotide sequence: (iii) is 15 to 30 nucleotides in length; and (iv) comprises 15 or more modified nucleotides independently selected from a 2’-O-methyl nucleotide and a 2’-fluoro nucleotide, wherein at least one modified nucleotide is a 2’-O-methyl nucleotide and at least one modified nucleotide is a 2’-fluoro nucleotide; or (b) a sense strand comprising a first nucleotide sequence that is at least about 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% identical to an RNA corresponding to a target gene, wherein the first nucleotide sequence: (i) is 15 to 30 nucleotides in length; and (ii) comprises 15 or more modified nucleotides independently selected from a 2’-O-methyl nucleotide and a 2’-fluoro nucleotide, wherein at least one modified nucleotide is a 2’-O-methyl nucleotide and at least one modified nucleotide is a 2’-fluoro nucleotide; and an antisense strand comprising a second nucleotide sequence that is at least about 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% complementary to the RNA corresponding to the target gene, wherein the second nucleotide sequence: (iii) is 15 to 30 nucleotides in length; and (iv) comprises 15 or more modified nucleotides independently selected from a 2’-O-methyl nucleotide and a 2’-fluoro nucleotide, wherein at least one modified nucleotide is a 2’-O-methyl nucleotide and the nucleotide at position 2, 5, 6, 8, 10, 14, 16, 17, and/or 18 from the 5’ end of the second nucleotide sequence is a 2’-fluoro nucleotide; wherein the target gene is a gene encoding ANGPTL8 or a gene that controls expression of ANGPTL8.
2. A short interfering nucleic acid (siNA) molecule comprising: (a) a sense strand comprising a first nucleotide sequence that is at least about 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% identical to an RNA corresponding to a target gene, wherein the first nucleotide sequence: (i) is 15 to 30 nucleotides in length; and (ii) comprises 15 or more modified nucleotides independently selected from a 2’-O-methyl nucleotide and a 2’-fluoro nucleotide, wherein at least one modified nucleotide is a 2’-O-methyl nucleotide and the nucleotide at position 3, 5, 7, 8, 9, 10, 11, 12, 14, 17, and/or 19 from the 5’ end of the first nucleotide sequence is a 2’-fluoro nucleotide or wherein at least one modified nucleotide is a 2’-O-methyl nucleotide and at least one modified nucleotide is a 2’-fluoro nucleotide; and an antisense strand comprising a second nucleotide sequence that is at least about 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% complementary to the RNA corresponding to the target gene, wherein the second nucleotide sequence: (iii) is 15 to 30 nucleotides in length; and (iv) comprises 15 or more modified nucleotides independently selected from a 2’-O-methyl nucleotide and a 2’-fluoro nucleotide, wherein at least one modified nucleotide is a 2’-O-methyl nucleotide and at least one modified nucleotide is a 2’-fluoro nucleotide; or (b) a sense strand comprising a first nucleotide sequence that is at least about 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% identical to an RNA corresponding to a target gene, wherein the first nucleotide sequence: (i) is 15 to 30 nucleotides in length; and (ii) comprises 15 or more modified nucleotides independently selected from a 2’-O-methyl nucleotide and a 2’-fluoro nucleotide, wherein at least one modified nucleotide is a 2’-O-methyl nucleotide and at least one modified nucleotide is a 2’-fluoro nucleotide; and an antisense strand comprising a second nucleotide sequence that is at least about 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% complementary to the RNA corresponding to the target gene, wherein the second nucleotide sequence: (iii) is 15 to 30 nucleotides in length; and (iv) comprises 15 or more modified nucleotides independently selected from a 2’-O-methyl nucleotide and a 2’-fluoro nucleotide, wherein at least one modified nucleotide is a 2’-O-methyl nucleotide and the nucleotide at position 2, 5, 6, 8, 10, 14, 16, 17, and/or 18 from the 5’ end of the second nucleotide sequence is a 2’-fluoro nucleotide; wherein the target gene is a gene encoding ANGPTL3 or a gene that controls expression of ANGPTL3.
3. A double-stranded short interfering nucleic acid (ds-siNA) molecule comprising: (a) a sense strand comprising a first nucleotide sequence, wherein the first nucleotide sequence is 15 to 30 nucleotides in length and comprises a nucleotide sequence as set forth in in any one of SEQ ID NOs: 1-316, and an antisense strand comprising a second nucleotide sequence, wherein the second nucleotide sequence is 15 to 30 nucleotides in length and comprises a nucleotide sequence that is a substantial reverse complement of the first nucleotide sequence; or (b) a sense strand comprising a first nucleotide sequence, wherein the first nucleotide sequence is 15 to 30 nucleotides in length and comprises a nucleotide sequence that is a substantial reverse complement of a second nucleotide sequence; and an antisense strand comprising the second nucleotide sequence, wherein the second nucleotide sequence is 15 to 30 nucleotides in length and comprises a nucleotide sequence as set forth in in any one of SEQ ID NOs: 317-632.
4. A double-stranded short interfering nucleic acid (ds-siNA) molecule comprising: (a) a sense strand comprising a first nucleotide sequence, wherein the first nucleotide sequence is 15 to 30 nucleotides in length and comprises a nucleotide sequence as set forth in in any one of SEQ ID NOs: 633-878, and an antisense strand comprising a second nucleotide sequence, wherein the second nucleotide sequence is 15 to 30 nucleotides in length and comprises a nucleotide sequence that is a substantial reverse complement of the first nucleotide sequence; or (b) a sense strand comprising a first nucleotide sequence, wherein the first nucleotide sequence is 15 to 30 nucleotides in length and comprises a nucleotide sequence that is a substantial reverse complement of a second nucleotide sequence; and an antisense strand comprising the second nucleotide sequence, wherein the second nucleotide sequence is 15 to 30 nucleotides in length and comprises a nucleotide sequence as set forth in any one of SEQ ID NOs: 879-1125.
5. A double-stranded short interfering nucleic acid (ds-siNA) molecule comprising (a) a sense strand comprising a nucleotide sequence as set forth in any one of SEQ ID NOs: 1-316, and (b) an antisense strand.
6. A double-stranded short interfering nucleic acid (ds-siNA) molecule comprising (a) a sense strand comprising a nucleotide sequence as set forth in any one of SEQ ID NOs: 633- 878, and (b) an antisense strand.
7. A double-stranded short interfering nucleic acid (ds-siNA) molecule comprising (a) an antisense strand comprising a nucleotide sequence as set forth in any one of SEQ ID NOs: 317-632, and (b) a sense strand.
8. A double-stranded short interfering nucleic acid (ds-siNA) molecule comprising (a) an antisense strand comprising a nucleotide sequence as set forth in any one of SEQ ID NOs: 879-1125, and (b) a sense strand.
9. The ds-siNA of any one of claims 2, 4, 6 or 8, wherein the sense strand comprises SEQ ID NOs: 633, 634, 642, 653, 654, 674, 687, 688, 690-694, 708-713, 756-757, 765, 776, 777, 797, 810, 811, 813-817, or 831-836, and the antisense strand comprises SEQ ID NOs: 879, 880, 888, 899, 900, 920, 933, 934, 936-940, 954-959, 1003-1004, 1012, 1023, 1024, 1044, 1057, 1058, 1060-1064, or 1078-1083, respectively.
10. The ds-siNA of any one of claims 2, 4, 6, 8 , wherein the sense strand comprises SEQ ID NOs: 633, 654, 688, 690, 708, 713, 756, 777, 811, 813, 831, or 836, and the antisense strand comprises SEQ ID NOs: 879, 900, 934, 936, 954, 959, 1003, 1024, 1058, 1060, 1078, or 1083, respectively.
11. The ds-siNA of any one of claims 1-10, wherein the sense and/or antisense strand further comprises a TT sequence at 3’ end.
12. The ds-siNA of any one of claims 1-11, wherein at least one end of the ds-siNA is a blunt end.
13. The ds-siNA of any one of claims 1-11, wherein at least one end of the ds-siNA comprises an overhang, wherein the overhang comprises at least one nucleotide, optionally wherein the at least one nucleotide comprises a 2’-O-methyl nucleotide, a deoxy nucleotide, or a uracil analogue or derivative.
14. The ds-siNA of any one of claims 1-11, wherein both ends of the ds-siNA comprise an overhang, wherein the overhang comprises at least one nucleotide, optionally wherein the at least one nucleotide comprises a 2’-O-methyl nucleotide, a deoxy nucleotide, or a uracil analogue or derivative.
15. The ds-siNA of any one of claims 1-14, wherein the sense strand and/or the antisense strand independently comprise 1 or more mesyl phosphoramidate internucleoside linkages, or 1 or more phosphorothioate internucleoside linkages.
16. The ds-siNA molecule according to any one of claims 1-15, wherein the sense strand comprises at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15 or more phosphorothioate internucleoside linkages.
17. The ds-siNA molecule of claim 16, wherein: (i) at least one phosphorothioate internucleoside linkage in the sense strand is between the nucleotides at positions 1 and 2 from the 5’ end of the first nucleotide sequence; (ii) at least one phosphorothioate internucleoside linkage is between the nucleotides at positions 2 and 3 from the 5’ end of the first nucleotide sequence.
18. The ds-siNA molecule according to any one of claims 1-17, wherein the antisense strand further comprises at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15 or more phosphorothioate internucleoside linkages.
19. The ds-siNA molecule of claim 18, wherein: (i) at least one phosphorothioate internucleoside linkage in the antisense strand is between the nucleotides at positions 1 and 2 from the 5’ end of the second nucleotide sequence; (ii) at least one phosphorothioate internucleoside linkage in the antisense strand is between the nucleotides at positions 2 and 3 from the 5’ end of the second nucleotide sequence; (iii) at least one phosphorothioate internucleoside linkage in the antisense strand is between the nucleotides at positions 1 and 2 from the 3’ end of the second nucleotide sequence; and/or (iv) at least one phosphorothioate internucleoside linkage is between the nucleotides at positions 2 and 3 from the 3’ end of the second nucleotide sequence.
20. The ds-siNA molecule according to any one of claims 1-19, wherein the sense strand comprises at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15 or more mesyl phosphoramidate internucleoside linkages.
21. The ds-siNA molecule of claim 20, wherein: (i) at least one mesyl phosphoramidate internucleoside linkage in the sense strand is between the nucleotides at positions 1 and 2 from the 5’ end of the first nucleotide sequence; (ii) at least one mesyl phosphoramidate internucleoside linkage is between the nucleotides at positions 2 and 3 from the 5’ end of the first nucleotide sequence.
22. The ds-siNA molecule according to any one of claims 1-21, wherein the antisense strand further comprises at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15 or more mesyl phosphoramidate internucleoside linkages.
23. The ds-siNA molecule of claim 22, wherein: (i) at least one mesyl phosphoramidate internucleoside linkage in the antisense strand is between the nucleotides at positions 1 and 2 from the 5’ end of the second nucleotide sequence; (ii) at least one mesyl phosphoramidate internucleoside linkage in the antisense strand is between the nucleotides at positions 2 and 3 from the 5’ end of the second nucleotide sequence; (iii) at least one mesyl phosphoramidate internucleoside linkage in the antisense strand is between the nucleotides at positions 1 and 2 from the 3’ end of the second nucleotide sequence; and/or (iv) at least one mesyl phosphoramidate internucleoside linkage is between the nucleotides at positions 2 and 3 from the 3’ end of the second nucleotide sequence.
24. The ds-siNA of any one of claims 1-23, wherein the antisense strand comprises one or more modified nucleotides.
25. The ds-siNA of any one of claims 1-24, wherein the sense strand comprises one or more modified nucleotides.
26. The ds-siNA of any one of claims 1-24, wherein the sense strand and the antisense strand each independently comprise one or more modified nucleotides.
27. The ds-siNA of any one of claims 24-26, wherein the modified nucleotides are independently selected from 2’-O-methyl nucleotides and 2’-fluoro nucleotides.
28. The ds-siNA of claim 27, wherein at least one 2’-fluoro nucleotide or 2’-O-methyl nucleotide is a 2’-fluoro or 2-O-methyl nucleotide mimic of Formula (V):
Figure imgf000190_0001
, wherein R1 is a nucleobase, aryl, heteroaryl, or H, Q1 and Q2 are independently S or O, R5 is –OCD3 , –F, or –OCH3, and R6 and R7 are independently H or D.
29. The siNA molecule according to any one of the preceding claims, wherein the antisense strand, sense strand, first nucleotide sequence, and/or second nucleotide sequence comprises at least one modified nucleotide selected from: , wherein Rx is a nucleobase, aryl, heteroaryl, or H,
Figure imgf000190_0002
Figure imgf000190_0003
(mun34), wherein Ry is a nucleobase,
Figure imgf000190_0004
Figure imgf000190_0005
Figure imgf000191_0001
Figure imgf000192_0001
(vm(56amim)U, wherein R is H or Bz), wherein B is a
Figure imgf000192_0002
nucleobase, aryl, heteroaryl, or H, and wherein
Figure imgf000192_0003
or
Figure imgf000192_0004
represents a phosphodiester linkage, a phosphorothioate linkage, a mesyl phosphoramidate linkage, or H.
30. The ds-siNA of any of claims 1-29, wherein the ds-siNA further comprises a phosphorylation blocker, a 5’-stabilized end cap, or a combination thereof.
31. The ds-siNA of claim 30, wherein the phosphorylation blocker is attached to the 5’ end of the sense strand, and optionally, wherein the phosphorylation blocker is attached to the 5’ end of the sense strand via one or more of a phosphodiester linkage, a phosphorothioate linkage, a mesyl phosphoramidate linkage, or a phosphorodithioate linkage.
32. The ds-siNA of claim 30 or 31, wherein the 5’-stabilized end cap is a 5’ vinyl phosphonate.
33. The ds-siNA of any of claims 30-32, wherein the 5’-stabilized end cap is attached to the 5’ end of the antisense strand, and optionally, wherein the 5’-stabilized end cap is attached to the 5’ end of the antisense strand with one or more of a phosphodiester linkage, a phosphorothioate linkage, a mesyl phosphoramidate linkage, or a phosphorodithioate linkage.
34. The ds-siNA according to any one of the preceding claims, wherein the ds-siNA further comprises a galactosamine.
35. The ds-siNA according to claim 34, wherein the galactosamine is an N- acetylgalactosamine comprising a structure of:
Figure imgf000193_0001
wherein m is 1, 2, 3, 4, or 5; each n is independently 1 or 2; p is 0 or 1; each R is independently H; each Y is independently selected from –O-P(=O)(SH)–, –O-P(=O)(O)–, –O-P(=O)(OH)–, and -O-P(S)S-; Z is H or a second protecting group; either L is a linker or L and Y in combination are a linker; and A is H, OH, a third protecting group, an activated group, or an oligonucleotide.
36. The siNA according to claim 34, wherein the galactosamine is an N- acetylgalactosamine comprising a structure of:
Figure imgf000194_0001
wherein Rz is OH or SH; and each n is independently 1 or 2.
37. The ds-siNA of any one of the preceding claims, wherein the sense and/or anti-sense strand of the ds-siNA is conjugated to GalNAc, Folate, Cholesterol, or Palmitic Acid.
38. The ds-siNA of any one of the preceding claims, wherein the ds-siNA reduces ANGPTL8 or ANGPTL3 expression by more than about 10 %, more than about 20% more than about 30%, more than about 40%, more than about 50%, more than about 60%, more than about 70%, more than about 80%, more than about 90%, more than about 95%, or 100%.
39. The ds-siNA of any one of the preceding claims, wherein the ds-siNA reduces ANGPTL8 or ANGPTL3 expression or activity with a EC50 value of 50 pM or less, of 40 pM or less, of 30 pM or less, of 20 pM or less, 15 pM or less, 10 pM or less, 5 pM or less, 2 pM or less, or 1 pM or less.
40. The ds-siNA of any one of the preceding claims, wherein the ds-siNA has a CC50 value of more than 1 µM.
41. The ds-siNA of any one of the preceding claims, wherein the ds-siNA comprises RNA nucleotides.
42. A ds-siNA, comprising a sense strand and an antisense strand, wherein the antisense strand comprises at its 5’ end one of the following:
Figure imgf000195_0001
nucleobase),
Figure imgf000195_0002
(vm(5mim)),
Figure imgf000195_0003
(vm(56mido)), and
Figure imgf000195_0004
(vm(56amim)U, wherein R is H or Bz)
43. A pharmaceutical composition comprising at least one ds-siNA according to any one of claims 1-42 and a pharmaceutically acceptable carrier or diluent.
44. A pharmaceutical composition comprising two or more ds-siNA according to any one of claims 1-42.
45. A pharmaceutical composition comprising at least a first siNA that reduces ANGPTL3 expression and a second siNA that reduces ANGPTL8 expression and a pharmaceutically acceptable carrier.
46. The pharmaceutical composition of claim 45, wherein the first siNA that reduces ANGPTL3 expression comprises a nucleic acid sequence disclosed in Table 3, and the second siNA that reduces ANGPTL8 expression comprises a nucleic acid sequence disclosed in Table 1.
47. The pharmaceutical composition of claim 45 or 46, wherein the first siNA that reduces ANGPTL3 expression is a ds-siNA disclosed in Table 4, and the second siNA that reduces ANGPTL8 expression is a ds-siNA disclosed in Table 2.
48 The pharmaceutical composition of any one of claims 45-47, wherein a weight ratio of the first siNA and the second siNA is selected from 1:10, 1:9, 1:8, 1:7, 1:6, 1:5, 1:4, 1:3, 1:2, 1:1, 2:1, 3:1, 4:1, 5:1, 6:1, 7:1, 8:1, 9:1, or 10:1.
49. The pharmaceutical composition of any one of claims 45-48, wherein a molar ration of the first siNA and the second siNA is selected from 1:10, 1:9, 1:8, 1:7, 1:6, 1:5, 1:4, 1:3, 1:2, 1:1, 2:1, 3:1, 4:1, 5:1, 6:1, 7:1, 8:1, 9:1, or 10:1.
50. The pharmaceutical composition of any one of claims 45-49, wherein the first siNA and the second siNA are connected by a linker.
51. The pharmaceutical composition of claim 50, wherein the linker is selected from a nucleotide-based linker or a non-nucleotide based linker.
52. The pharmaceutical composition of claim 51, wherein the nucleotide-based linker comprises between about 1 to about 15 nucleotides.
53. The pharmaceutical composition of claim 51, wherein the linker comprises at least one N-acetylgalactosamine.
54. The pharmaceutical composition of any one of claims 43-53, wherein the pharmaceutical composition is formulated for parenteral, ocular, nasal, transdermal, pulmonary, or topical administration or comprises a liposome that encapsulates the ds-siNA.
55. Use of the ds-siNA according to any one of claims 1-42 in the manufacture of a medicament for treating cardiovascular disease or nonalcoholic fatty liver disease.
56. Use of two or more ds-siNA according to any one of claims 1-42 in the manufacture of a medicament for treating cardiovascular disease or nonalcoholic fatty liver disease.
57. The ds-siNA according to any one of claims 1-42 or the pharmaceutical composition according to any one of claims 43-54 for use in treating cardiovascular disease or nonalcoholic fatty liver disease.
58. Two or more ds-siNA according to any one of claims 1-42 or the pharmaceutical composition according to any one of claims 43-54 for use in treating cardiovascular disease or nonalcoholic fatty liver disease
59. A method of treating a disease in a subject in need thereof, comprising administering to the subject the ds-siNA according to any one of claims 1-42 or the pharmaceutical composition according to any one of claims 43-54, wherein the disease is cardiovascular disease or nonalcoholic fatty liver disease.
60. A method of treating a disease in a subject in need thereof, comprising administering to the subject two or more ds-siNA according to any one of claims 1-42 or the pharmaceutical composition according to any one of claims 43-54, wherein the disease is cardiovascular disease or nonalcoholic fatty liver disease.
61. A method of treating a disease in a subject in need thereof, comprising administering to the subject a first pharmaceutical composition comprising at least a first ds-siNA that reduces ANGPTL3 expression and a second pharmaceutical composition comprising at least a second ds-siNA that reduces ANGPTL8 expression, wherein the disease is cardiovascular disease or nonalcoholic fatty liver disease.
62. The method of any one of claims 59-61, wherein the cardiovascular disease is Hypertriglyceridemia (HTG) or Familial Hypercholesterolemia (FH).
63. The method according to any one of claims 59-62, wherein the subject is a mammal, and optionally wherein the mammal is a human or a non-human primate.
64. The method of any one of claims 59-63, wherein the ds-siNA or pharmaceutical composition(s) are administered intravenously, subcutaneously, or via inhalation.
65. The method of any one of claims 59-64, wherein the subject has been treated with one or more additional therapeutic agents.
66. A method of reducing ANGPTL8 and/or ANGPTL3 expression or activity in a tissue, an organ, or a cell of a subject in need thereof, wherein the method comprises delivering to the subject one or more of the ds-siNA according to any one of claims 1-42 or the pharmaceutical composition according to any one of claims 43-54.
67. A modified nucleoside selected from
Figure imgf000198_0001
(vm, wherein B is a nucleobase),
Figure imgf000198_0002
(vm(5mim)),
Figure imgf000198_0003
(vm(56mido)), and
Figure imgf000198_0004
(vm(56amim)U, wherein R is H or Bz).
PCT/US2024/026619 2023-04-27 2024-04-26 Short interfering nucleic acid (sina) molecules targeting angptl 3 and angptl 8 and methods of using the same WO2024227059A2 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US202363462456P 2023-04-27 2023-04-27
US63/462,456 2023-04-27

Publications (2)

Publication Number Publication Date
WO2024227059A2 true WO2024227059A2 (en) 2024-10-31
WO2024227059A3 WO2024227059A3 (en) 2024-12-26

Family

ID=91185069

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2024/026619 WO2024227059A2 (en) 2023-04-27 2024-04-26 Short interfering nucleic acid (sina) molecules targeting angptl 3 and angptl 8 and methods of using the same

Country Status (1)

Country Link
WO (1) WO2024227059A2 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN119367545A (en) * 2025-01-02 2025-01-28 山东第一医科大学(山东省医学科学院) Application of targeted intervention of ANGPTL8 mRNA in the preparation of drugs for the treatment of hyperhomocysteinemia

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2018044350A1 (en) 2016-09-02 2018-03-08 Arrowhead Pharmaceuticals, Inc Targeting ligands
WO2018045317A1 (en) 2016-09-02 2018-03-08 Dicerna Pharmaceuticals, Inc. 4'-phosphate analogs and oligonucleotides comprising the same
US10087210B2 (en) 2010-04-28 2018-10-02 Ionis Pharmaceuticals, Inc. Modified nucleosides, analogs thereof and oligomeric compounds prepared therefrom

Family Cites Families (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
TWI723986B (en) * 2015-04-13 2021-04-11 美商阿尼拉製藥公司 ANGIOPOIETIN-LIKE 3 (ANGPTL3) iRNA COMPOSITIONS AND METHODS OF USE THEREOF
ES2896107T3 (en) * 2016-04-08 2022-02-23 Regeneron Pharma Methods of treating hyperlipidemia with an ANGPTL8 inhibitor and an ANGPTL3 inhibitor
US11560563B2 (en) * 2018-04-05 2023-01-24 Silence Therapeutics Gmbh SiRNAs with vinylphosphonate at the 5′ end of the antisense strand
US20220025367A1 (en) * 2018-11-23 2022-01-27 Sanofi Novel rna compositions and methods for inhibiting angptl8
EP4013767A4 (en) * 2019-08-15 2023-10-25 Ionis Pharmaceuticals, Inc. Linkage modified oligomeric compounds and uses thereof
JP2024523565A (en) * 2021-06-24 2024-06-28 イーライ リリー アンド カンパニー Novel RNA Therapeutics and Uses Thereof
AU2022343115A1 (en) * 2021-09-08 2024-03-14 Aligos Therapeutics, Inc. Modified short interfering nucleic acid (sina) molecules and uses thereof
JP2024536838A (en) * 2021-09-23 2024-10-08 シャンハイ アーゴ バイオファーマシューティカル カンパニー リミテッド Compositions and methods for inhibiting expression of angiopoietin-like protein 3 (ANGPTS3)

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10087210B2 (en) 2010-04-28 2018-10-02 Ionis Pharmaceuticals, Inc. Modified nucleosides, analogs thereof and oligomeric compounds prepared therefrom
WO2018044350A1 (en) 2016-09-02 2018-03-08 Arrowhead Pharmaceuticals, Inc Targeting ligands
WO2018045317A1 (en) 2016-09-02 2018-03-08 Dicerna Pharmaceuticals, Inc. 4'-phosphate analogs and oligonucleotides comprising the same

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
MARTIN: "Remington's Pharmaceutical Sciences", 1975, MACK PUBL. CO.
PARMAR ET AL., J MED CHEM, vol. 201861, no. 3, pages 734 - 744

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN119367545A (en) * 2025-01-02 2025-01-28 山东第一医科大学(山东省医学科学院) Application of targeted intervention of ANGPTL8 mRNA in the preparation of drugs for the treatment of hyperhomocysteinemia

Also Published As

Publication number Publication date
WO2024227059A3 (en) 2024-12-26

Similar Documents

Publication Publication Date Title
US11549110B2 (en) Modified short interfering nucleic acid (siNA) molecules and uses thereof
AU2017229469B2 (en) Targeting ligands for therapeutic compounds
CN107266391B (en) Amine cationic lipids and uses thereof
JP6832285B2 (en) Antisense-oligonucleotide as a TGF-R signaling inhibitor
EA031110B1 (en) Boronic acid conjugates of oligonucleotide analogues
JP2024533262A (en) Modified short interfering nucleic acid (SINA) molecules and uses thereof
US20230159929A1 (en) MODIFIED SHORT INTERFERING NUCLEIC ACID (siNA) MOLECULES AND USES THEREOF
EP3377630A1 (en) Treatment of age-related macular degeneration using rna complexes that target myd88 or tlr3
WO2020146521A2 (en) Rnai agents for inhibiting expression of hif-2 alpha (epas1), compositions thereof, and methods of use
CN112384222A (en) Exon skipping oligomer conjugates for muscular dystrophy
WO2024227059A2 (en) Short interfering nucleic acid (sina) molecules targeting angptl 3 and angptl 8 and methods of using the same
JPWO2020027227A1 (en) Small Cell Lung Cancer Therapeutic Agent Containing Oligonucleotides
US20230140736A1 (en) Antisense nucleic acid inducing skipping of exon 51
WO2023122316A2 (en) SPLICE SWITCH OLIGONUCLEOTIDE (SSO) MOLECULES FOR TARGETING β-CATENIN AND USES THEREOF
JP2024533173A (en) PNPLA3-Targeted Small Interfering RNA (siRNA) Molecules and Uses Thereof
US20240182889A1 (en) Microrna-27b inhibitors
WO2024097674A2 (en) Modified short interfering nucleic acid (sina) molecules and uses thereof
WO2025040139A1 (en) Rnai agent for inhibiting diacylglycerol-o-acyltransferase 2 expression, and application thereof
WO2023097251A1 (en) Oligonucleotide-containing transcription factor targeting chimeras
CN119563025A (en) A dsRNA molecule for inhibiting LP(a) gene expression and its application
WO2024182446A2 (en) Pnpla3-targeting short interfering rna (sirna) molecules and uses thereof
TW202448485A (en) Dystrophin exon skipping oligonucleotides
NZ785880A (en) Targeting ligands for therapeutic compounds

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: 24727119

Country of ref document: EP

Kind code of ref document: A2