WO2023178141A2 - Combination of stat3 targeting oligonucleotides and pd-l1 inhibitors - Google Patents

Combination of stat3 targeting oligonucleotides and pd-l1 inhibitors Download PDF

Info

Publication number
WO2023178141A2
WO2023178141A2 PCT/US2023/064381 US2023064381W WO2023178141A2 WO 2023178141 A2 WO2023178141 A2 WO 2023178141A2 US 2023064381 W US2023064381 W US 2023064381W WO 2023178141 A2 WO2023178141 A2 WO 2023178141A2
Authority
WO
WIPO (PCT)
Prior art keywords
seq
nos
oligonucleotide
nucleotides
antisense strand
Prior art date
Application number
PCT/US2023/064381
Other languages
French (fr)
Other versions
WO2023178141A3 (en
Inventor
Shanthi Ganesh
Marc Abrams
Original Assignee
Dicerna Pharmaceuticals, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Dicerna Pharmaceuticals, Inc. filed Critical Dicerna Pharmaceuticals, Inc.
Publication of WO2023178141A2 publication Critical patent/WO2023178141A2/en
Publication of WO2023178141A3 publication Critical patent/WO2023178141A3/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7088Compounds having three or more nucleosides or nucleotides
    • A61K31/7125Nucleic acids or oligonucleotides having modified internucleoside linkage, i.e. other than 3'-5' phosphodiesters
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • 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
    • C12N15/1135Non-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 oncogenes or tumor suppressor genes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/10Type of nucleic acid
    • C12N2310/11Antisense
    • 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
    • C12N2310/312Phosphonates
    • C12N2310/3125Methylphosphonates
    • 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
    • C12N2310/35Nature of the modification
    • C12N2310/351Conjugate
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2320/00Applications; Uses
    • C12N2320/30Special therapeutic applications
    • C12N2320/31Combination therapy

Definitions

  • MDR multidrug resistance
  • cancer research has often been focused on tumor cells even though the effect of the tumor microenvironment and the ‘normal’ or non-cancerous cells within it that have been shown to play a key role in tumor progression, development and MDR (Klemm et al., TRENDS CELL BIOL (2015) 25(4): 198-213). Novel therapies that target different facets of the TME that contribute to tumor growth are needed.
  • the disclosure is based, in part, on the discovery that a combination of a STAT3 oligonucleotide and a PD-L1 inhibitor provides synergistic anti-tumor efficacy for tumors of varying tumor microenvironments.
  • a STAT3 oligonucleotide conjugated to a lipid when delivered in combination with an anti-PD-L1 antibody, reduced tumor volume in vivo in immunosuppressive and inflamed tumor models.
  • the combination of a STAT3 oligonucleotide and PD-L1 inhibitor induced an anti-tumor memory response as when mice were re-challenged with cancer cells, no tumors were established.
  • the efficacy of the STAT3 oligonucleotide and PD- L1 inhibitor was dependent on the presence of CD8+ T cells.
  • the disclosure provides a method of treating cancer in a subject that has received or is receiving a PD-L1 inhibitor, the method comprising administering an RNAi oligonucleotide comprising an antisense strand of 15 to 30 nucleotides in length and a sense strand of 15 to 40 nucleotides in length, wherein the antisense and sense strands form a duplex region, wherein the antisense strand comprises a region of complementarity to a STAT3 mRNA target sequence, and wherein the region of complementarity is at least 15 contiguous nucleotides in length, thereby treating cancer in the subject.
  • the disclosure provides a method of treating cancer in a subject that has received or is receiving an RNAi oligonucleotide, the method comprising administering a PD-L1 inhibitor, wherein the oligonucleotide comprises an antisense strand of 15 to 30 nucleotides in length and a sense strand of 15 to 40 nucleotides in length, wherein the antisense and sense strands form a duplex region, wherein the antisense strand comprises a region of complementarity to a STAT3 mRNA target sequence, and wherein the region of complementarity is at least 15 contiguous nucleotides in length, thereby treating cancer in the subject.
  • the disclosure provides a method of treating a disease, disorder or condition associated with activated STAT3 expression, comprising administering to a subject in need thereof an RNAi oligonucleotide, wherein the subject has received or is receiving a PD-L1 inhibitor, wherein the oligonucleotide comprises an antisense strand of 15 to 30 nucleotides in length and a sense strand of 15 to 40 nucleotides in length, wherein the antisense and sense strands form a duplex region, wherein the antisense strand comprises a region of complementarity to a STAT3 mRNA target sequence, and wherein the region of complementarity is at least 15 contiguous nucleotides in length, thereby treating the disease, disorder, or condition associated with activated STAT3 expression.
  • the disclosure provides a kit comprising an RNAi oligonucleotide, an optional pharmaceutically acceptable carrier, and a package insert comprising instructions for administering the oligonucleotide to a subject in need thereof that has received or is receiving a PD-L1 inhibitor, wherein the oligonucleotide comprises an antisense strand of 15 to 30 nucleotides in length and a sense strand of 15 to 40 nucleotides in length, wherein the antisense and sense strands form a duplex region, wherein the antisense strand comprises a region of complementarity to a STAT3 mRNA target sequence, and wherein the region of complementarity is at least 15 contiguous nucleotides in length.
  • kits comprising a PD-L1 inhibitor, an optional pharmaceutically acceptable carrier, and a package insert comprising instructions for administering the antibody to a subject in need thereof that has received or is receiving an RNAi oligonucleotide comprising an antisense strand of 15 to 30 nucleotides in length and a sense strand of 15 to 40 nucleotides in length, wherein the antisense and sense strands form a duplex region, wherein the antisense strand comprises a region of complementarity to a STAT3 mRNA target sequence, and wherein the region of complementarity is at least 15 contiguous nucleotides in length.
  • an RNAi oligonucleotide comprising an antisense strand of 15 to 30 nucleotides in length and a sense strand of 15 to 40 nucleotides in length, wherein the antisense and sense strands form a duplex region, wherein the antisense strand comprises a region of complementarity to a STAT
  • a subject has a disease, disorder, or condition associated with activated STAT3 expression.
  • the disease, disorder, or condition associated with activated STAT3 expression is a cancer.
  • the cancer is selected from carcinoma, sarcoma, melanoma, lymphoma, and leukemia, prostate cancer, breast cancer, hepatocellular carcinoma (HCC), colorectal cancer, pancreatic cancer and glioblastoma.
  • the cancer comprises an immunosuppressive tumor microenvironment.
  • the immunosuppressive tumor microenvironment has low levels of CD8+ T cell infiltration and high levels of MDSCs.
  • the cancer comprises an inflamed tumor microenvironment.
  • the inflamed tumor microenvironment comprises infiltrating T cells.
  • the inflamed tumor microenvironment comprises high levels of CD8+ T cell infiltration and high levels of MDSCs.
  • the cancer comprises a tumor resistant to immune checkpoint therapy.
  • the cancer comprises a tumor partially resistant to immune checkpoint therapy.
  • the cancer comprises a tumor sensitive to immune checkpoint therapy.
  • the methods described herein treat spontaneous tumor metastasis.
  • the methods described herein reduce immune suppressive genes and enhance immune activation genes.
  • immune suppressive genes comprise checkpoint inhibitors, STAT3 mediated genes, suppressive cytokines, suppressive chemokines, and angiogenesis and matrix remodeling related genes.
  • immune activation genes comprise gene related to T cell migration, T cell activation, T cell memory, and/or T cell cytotoxicity.
  • the PD-L1 inhibitor is an antibody.
  • the antibody is an anti-PD-L1 antibody.
  • the anti- PDL1 antibody is selected from FAZ053, atezolizumab, avelumab, durvalumab, envafolimab, and BMS-936559.
  • the antibody is an anti-PD-1 antibody.
  • the anti-PD-1 antibody is selected from nivolumab, pembrolizumab, and cemiplimab.
  • the PD-L1 inhibitor is a small molecule inhibitor.
  • the PD-L1 inhibitor is a peptide.
  • the PD-L1 inhibitor is a nucleic acid molecule.
  • the nucleic acid molecule is selected from an antisense oligonucleotide, an siRNA, or an miRNA.
  • the STAT3 mRNA target sequence comprises any one of SEQ ID NOs: 89-280.
  • the region of complementarity is fully complementary to the STAT3 mRNA target sequence. In some aspects, the region of complementarity comprises no more than 4 mismatches to the STAT3 mRNA target sequence.
  • the antisense strand is 19 to 27 nucleotides in length. In some aspects, the antisense strand is 21 to 27 nucleotides in length, optionally wherein the antisense strand is 22 nucleotides in length. [0017] In some or any of the foregoing or related aspects, the sense strand is 19 to 40 nucleotides in length, optionally wherein the sense strand is 36 nucleotides in length.
  • the duplex region is at least 19 nucleotides in length. In some aspects, the duplex region is at least 20 nucleotides in length, optionally wherein the duplex region is 21 nucleotides in length. [0019] In some or any of the foregoing or related aspects, the region of complementarity to STAT3 is at least 19 contiguous nucleotides in length. In some aspects, the region of complementarity to STAT3 is at least 21 contiguous nucleotides in length.
  • the sense strand comprises at its 3′ end a stem-loop set forth as: S1-L-S2, wherein S1 is complementary to S2, and wherein L forms a loop between S1 and S2 of 3 to 5 nucleotides in length.
  • L is a tetraloop, optionally wherein L is 4 nucleotides in length.
  • L comprises a sequence set forth as GAAA.
  • the antisense strand comprises a 3’ overhang sequence of one or more nucleotides in length, optionally wherein the 3’ overhang sequence is 2 nucleotides in length, optionally wherein the 3’ overhang sequence is GG.
  • the oligonucleotide comprises at least one modified nucleotide. In some aspects, the modified nucleotide comprises a 2′- modification.
  • the 2′-modification is a modification selected from 2′- aminoethyl, 2′-fluoro, 2′-O-methyl, 2′-O-methoxyethyl, and 2′-deoxy-2′-fluoro- ⁇ -d- arabinonucleic acid.
  • about 10-15%, 10%, 11%, 12%, 13%, 14% or 15% of the nucleotides of the sense strand comprise a 2’-fluoro modification.
  • about 25-35%, 25%, 26%, 27%, 28%, 29%, 30%, 31%, 32%, 33%, 34% or 35% of the nucleotides of the antisense strand comprise a 2’-fluoro modification.
  • nucleotides of the oligonucleotide comprise a 2’-fluoro modification.
  • the sense strand comprises 36 nucleotides with positions 1-36 from 5’ to 3 ’, wherein positions 8-11 comprise a 2’-fluoro modification.
  • the antisense strand comprises 22 nucleotides with positions 1-22 from 3’ to 5’, and wherein positions 2, 3, 4, 5, 7, 10 and 14 comprise a 2’-fluoro modification.
  • the remaining nucleotides comprise a 2’-O-methyl modification.
  • the oligonucleotide comprises at least one modified internucleotide linkage. In some aspects, the at least one modified internucleotide linkage is a phosphorothioate linkage.
  • the 4′-carbon of the sugar of the 5′-nucleotide of the antisense strand comprises a phosphate analog. In some aspects, the phosphate analog is oxymethylphosphonate, vinylphosphonate or malonylphosphonate.
  • At least one nucleotide of the oligonucleotide is conjugated to one or more targeting ligands.
  • the nucleotide is conjugated to more than one targeting ligands, wherein the targeting ligands are the same or are different.
  • the one or more targeting ligands is selected from carbohydrate, amino sugar, cholesterol, polypeptide, or lipid.
  • the one or more targeting ligands is a saturated or unsaturated fatty acid moiety.
  • the targeting ligand is a saturated fatty acid moiety that ranges in size from C10 to C24 long.
  • the targeting ligand is a C16 saturated fatty acid moiety. In some aspects, the targeting ligand is a C18 saturated fatty acid moiety. In some aspects, the targeting ligand is a C22 saturated fatty acid moiety. In some aspects, the targeting ligand comprises a N-acetylgalactosamine (GalNAc) moiety. In some aspects, the GalNAc moiety is a monovalent GalNAc moiety, a bivalent GalNAc moiety, a trivalent GalNAc moiety or a tetravalent GalNAc moiety. In some aspects, up to 4 nucleotides of L of the stem-loop are each conjugated to a monovalent GalNAc moiety.
  • the sense strand comprises a sequence as set forth in SEQ ID NOs: 857-946.
  • the antisense strand comprises a sequence as set for in SEQ ID NOs: 947-1036.
  • the sense strand and antisense strand comprise nucleotide sequences selected from the group consisting of: (a) SEQ ID NOs: 9 and 10, respectively; (b) SEQ ID NOs: 37 and 38, respectively; (c) SEQ ID NOs: 65 and 66, respectively; and (d) SEQ ID NOs: 69 and 70, respectively.
  • the sense strand and antisense strand comprise nucleotide sequences selected from the group consisting of: (a) SEQ ID NOs: 861 and 951, respectively; (b) SEQ ID NOs: 857 and 947, respectively; (c) SEQ ID NOs: 858 and 948, respectively; (d) SEQ ID NOs: 859 and 949, respectively; (e) SEQ ID NOs: 860 and 950, respectively; (f) SEQ ID NOs: 862 and 952, respectively; (g) SEQ ID NOs: 863 and 953, respectively; (h) SEQ ID NOs: 864 and 954, respectively; (i) SEQ ID NOs: 865 and 955, respectively; (j) SEQ ID NOs: 866 and 956, respectively; (k) SEQ ID NOs: 867 and 957, respectively; (l) SEQ ID NOs: 868 and 958, respectively; (m) SEQ ID NOs: 869 and 9
  • the sense strand comprises the nucleotide sequence of SEQ ID NO: 862 and the antisense strand comprises the nucleotide sequence of SEQ ID NO: 952.
  • the sense strand comprises the nucleotide sequence of SEQ ID NO: 875 and the antisense strand comprises the nucleotide sequence of SEQ ID NO: 965.
  • the sense strand comprises the nucleotide sequence of SEQ ID NO: 876 and the antisense strand comprises the nucleotide sequence of SEQ ID NO: 966.
  • sense strand comprises the nucleotide sequence of SEQ ID NO: 920 and the antisense strand comprises the nucleotide sequence of SEQ ID NO: 1010.
  • the sense strand and antisense strand comprise nucleotide sequences selected from the group consisting of: (a) SEQ ID NOs: 901 and 991, respectively; (b) SEQ ID NOs: 910 and 1000, respectively; (c) SEQ ID NOs: 899 and 989, respectively; (d) SEQ ID NOs: 896 and 986, respectively; (e) SEQ ID NOs: 892 and 982, respectively; (f) SEQ ID NOs: 890 and 980, respectively; and (g) SEQ ID NOs: 889 and 979, respectively.
  • the sense strand and antisense strand comprise nucleotide sequences selected from the group consisting of: (a) SEQ ID NOs: 940 and 1030, respectively; (b) SEQ ID NOs: 937 and 1027, respectively; and (c) SEQ ID NOs: 939 and 1029, respectively.
  • the sense strand and antisense strand comprise nucleotide sequences selected from the group consisting of: (a) SEQ ID NOs: 915 and 1005, respectively; (b) SEQ ID NOs: 924 and 1014, respectively; (c) SEQ ID NOs: 913 and 1003, respectively; and (d) SEQ ID NOs: 920 and 1010, respectively.
  • the sense strand comprises a nucleotide sequence of any one of SEQ ID NOs: 11, 39, 67 and 71.
  • the antisense strand comprises a nucleotide sequence of any one of SEQ ID NOs: 12, 40, 68 and 72.
  • the sense strand and antisense strand comprise nucleotide sequences selected from the group consisting of: (a) SEQ ID NOs: 11 and 12, respectively; (b) SEQ ID NOs: 39 and 40, respectively; (c) SEQ ID NOs: 67 and 68, respectively; and (d) SEQ ID NOs: 71 and 72, respectively.
  • the sense strand comprises a nucleotide sequence selected from SEQ ID NOs: 1042, 1055, 1056, and 1100.
  • the antisense strand comprises a nucleotide sequence selected from SEQ ID NOs: 1132, 1145, 1146, and 1190.
  • the sense strand and antisense strand comprise nucleotide sequences selected from the group consisting of: (a) SEQ ID NOs: 1041 and 1131, respectively; (b) SEQ ID NOs: 1037 and 1127, respectively; (c) SEQ ID NOs: 1038 and 1128, respectively; (d) SEQ ID NOs: 1039 and 1129, respectively; (e) SEQ ID NOs: 1040 and 1130, respectively; (f) SEQ ID NOs: 1042 and 1132, respectively; (g) SEQ ID NOs: 1043 and 1133, respectively; (h) SEQ ID NOs: 1044 and 1134, respectively; (i) SEQ ID NOs: 1045 and 1135, respectively; (j) SEQ ID NOs: 1046 and 1136, respectively; (k) SEQ ID NOs: 1047 and 1137, respectively; (l) SEQ ID NOs: 1048 and 1138, respectively; (m) SEQ ID NOs
  • the sense strand comprises the nucleotide sequence of SEQ ID NO: 1042 and the antisense strand comprises the nucleotide sequence of SEQ ID NO: 1132.
  • the sense strand comprises the nucleotide sequence of SEQ ID NO: 1055 and the antisense strand comprises the nucleotide sequence of SEQ ID NO: 1145.
  • the sense strand comprises the nucleotide sequence of SEQ ID NO: 1056 and the antisense strand comprises the nucleotide sequence of SEQ ID NO: 1146.
  • the sense strand comprises the nucleotide sequence of SEQ ID NO: 1100 and the antisense strand comprises the nucleotide sequence of SEQ ID NO: 1190.
  • the sense strand and antisense strand comprise nucleotide sequences selected from the group consisting of: (a) SEQ ID NOs: 1081 and 1171, respectively; (b) SEQ ID NOs: 1090 and 1180, respectively; (c) SEQ ID NOs: 1079 and 1169, respectively; (d) SEQ ID NOs: 1076 and 1166, respectively; (e) SEQ ID NOs: 1072 and 1162, respectively; (f) SEQ ID NOs: 1070 and 1160, respectively; and (g) SEQ ID NOs: 1069 and 1159, respectively.
  • the sense strand and antisense strand comprise nucleotide sequences selected from the group consisting of: (a) SEQ ID NOs: 1120 and 1210, respectively; (b) SEQ ID NOs: 1117 and 1207, respectively; and (c) SEQ ID NOs: 1119 and 1209, respectively.
  • the sense strand and antisense strand comprise nucleotide sequences selected from the group consisting of: (a) SEQ ID NOs: 1095 and 1185, respectively; (b) SEQ ID NOs: 1104 and 1194, respectively; (c) SEQ ID NOs: 1093 and 1183, respectively; and (d) SEQ ID NOs: 1100 and 1190, respectively.
  • the disclosure provides a method of treating cancer in a subject that has received or is receiving an anti-PD-L1 antibody, the method comprising administering an RNAi oligonucleotide comprising an antisense strand comprising a nucleotide sequence set forth in SEQ ID NO: 84, wherein the antisense strand comprises a region of complementarity to a STAT3 mRNA target sequence, and a sense strand comprising a nucleotide sequence set forth in SEQ ID NO: 83, thereby treating cancer in the subject.
  • the disclosure provides a method of treating cancer in a subject that has received or is receiving an RNAi oligonucleotide, the method comprising administering an anti-PD-L1 antibody, wherein the oligonucleotide comprises an antisense strand comprising a nucleotide sequence set forth in SEQ ID NO: 84, wherein the antisense strand comprises a region of complementarity to a STAT3 mRNA target sequence, and a sense strand comprising a nucleotide sequence set forth in SEQ ID NO: 83, thereby treating cancer in the subject.
  • the disclosure provides a method of treating a disease, disorder or condition associated with activated STAT3 expression, comprising administering to a subject in need thereof an RNAi oligonucleotide, wherein the subject has received or is receiving an anti-PD-L1 antibody, wherein the oligonucleotide comprises an antisense strand comprising a nucleotide sequence set forth in SEQ ID NO: 84, wherein the antisense strand comprises a region of complementarity to a STAT3 mRNA target sequence, and a sense strand comprising a nucleotide sequence set forth in SEQ ID NO: 83, thereby treating the disease, disorder, or condition associated with activated STAT3 expression.
  • the disclosure provides a kit comprising an RNAi oligonucleotide, an optional pharmaceutically acceptable carrier, and a package insert comprising instructions for administering the oligonucleotide to a subject in need thereof that has received or is receiving an anti-PD-L1 antibody, wherein the oligonucleotide comprises an antisense strand comprising a nucleotide sequence set forth in SEQ ID NO: 84, wherein the antisense strand comprises a region of complementarity to a STAT3 mRNA target sequence, and a sense strand comprising a nucleotide sequence set forth in SEQ ID NO: 83.
  • kits comprising an anti-PD-L1 antibody, an optional pharmaceutically acceptable carrier, and a package insert comprising instructions for administering the antibody to a subject in need thereof that has received or is receiving an RNAi oligonucleotide comprising an antisense strand comprising a nucleotide sequence set forth in SEQ ID NO: 84, wherein the antisense strand comprises a region of complementarity to a STAT3 mRNA target sequence, and a sense strand comprising a nucleotide sequence set forth in SEQ ID NO: 83.
  • the disclosure provides a method of treating cancer in a subject that has received or is receiving an anti-PD-L1 antibody, the method comprising administering an RNAi oligonucleotide comprising an antisense strand comprising a nucleotide sequence set forth in SEQ ID NO: 70, wherein the antisense strand comprises a region of complementarity to a STAT3 mRNA target sequence, and a sense strand comprising a nucleotide sequence set forth in SEQ ID NO: 69, thereby treating cancer in the subject.
  • the disclosure provides a method of treating cancer in a subject that has received or is receiving an RNAi oligonucleotide, the method comprising administering an anti-PD-L1 antibody, wherein the oligonucleotide comprises an antisense strand comprising a nucleotide sequence set forth in SEQ ID NO: 70, wherein the antisense strand comprises a region of complementarity to a STAT3 mRNA target sequence, and a sense strand comprising a nucleotide sequence set forth in SEQ ID NO: 69, thereby treating cancer in the subject.
  • the disclosure provides a method of treating a disease, disorder or condition associated with activated STAT3 expression, comprising administering to a subject in need thereof an RNAi oligonucleotide, wherein the subject has received or is receiving an anti-PD-L1 antibody, wherein the oligonucleotide comprises an antisense strand comprising a nucleotide sequence set forth in SEQ ID NO: 70, wherein the antisense strand comprises a region of complementarity to a STAT3 mRNA target sequence, and a sense strand comprising a nucleotide sequence set forth in SEQ ID NO: 69, thereby treating the disease, disorder, or condition associated with activated STAT3 expression.
  • the disclosure provides a kit comprising an RNAi oligonucleotide, an optional pharmaceutically acceptable carrier, and a package insert comprising instructions for administering the oligonucleotide to a subject in need thereof that has received or is receiving an anti-PD-L1 antibody, wherein the oligonucleotide comprises an antisense strand comprising a nucleotide sequence set forth in SEQ ID NO: 70, wherein the antisense strand comprises a region of complementarity to a STAT3 mRNA target sequence, and a sense strand comprising a nucleotide sequence set forth in SEQ ID NO: 69.
  • kits comprising an anti-PD-L1 antibody, an optional pharmaceutically acceptable carrier, and a package insert comprising instructions for administering the antibody to a subject in need thereof that has received or is receiving an RNAi oligonucleotide comprising an antisense strand comprising a nucleotide sequence set forth in SEQ ID NO: 70, wherein the antisense strand comprises a region of complementarity to a STAT3 mRNA target sequence, and a sense strand comprising a nucleotide sequence set forth in SEQ ID NO: 69.
  • FIG.1A provides structures of RNAi oligonucleotide molecules having chemical modifications with GalNAc (top) or lipid (e.g., C18 hydrocarbon chain) (bottom) conjugated to the oligonucleotide molecule to generate oligonucleotide-ligand conjugates.
  • FIG.1B provides structures of lipid tails suitable for conjugation to RNAi oligonucleotide molecules.
  • FIGs.2A and 2B are graphs showing remaining mouse Stat3 mRNA levels in the livers of mice treated with GalXC-STAT3-conjugates (GalNAc conjugates) targeting different regions of Stat3 mRNA.
  • FIGs.3A and 3B are graphs showing mouse Stat3 mRNA expression 3 days after treatment with GalXC-STAT3-C18 conjugates in G-MDSCs and M-MDSCs derived from Pan02 xenografts implanted in mice. Tumors were dosed at 25 mg/kg (FIG.3A) and 50 mg/kg (FIG.3B).
  • FIGs.4A and 4B are graphs showing mouse Stat3 mRNA expression after treatment of Pan02 xenograft mice with GalXC-STAT3-C18 conjugates in bulk tumor (TME) (FIG.4A) and tumor draining lymph nodes (TdLNs) (FIG.4B) at doses of 25 and 50 mg/kg.
  • FIG.5A provides graphs showing the effect of GalXC-STAT3-C18-4123 on Stat3 and Pdl1 mRNA levels in G/M-MDSCs in TME and TdLNs of Pan02 xenograft mice 3 days after a dose of 25 or 50 mg/kg of the conjugated oligonucleotide.
  • FIG.5B provides graphs showing the effect of GalXC-STAT3-C18-4123 on Stat3 and Pdl1 mRNA levels in TdLN of Pan02 xenograft mice 7 days after a 25mg/kg dose of the conjugated oligonucleotide.
  • FIGs.6A and 6B are graphs showing the in vivo effect of subcutaneous treatment of a total dose of 50 mg/kg GalXC-STAT3-C18-4123 on tumor volume over time in immunocompetent mice bearing Pan02 murine pancreatic tumors.
  • FIG.7 provides a graph depicting the percent (%) of human STAT3 mRNA remaining in Huh7 cells endogenously expressing human STAT3, after 24-hour treatment with 1nM of DsiRNA targeting various regions of the STAT3 gene.192 DsiRNAs were designed and screened. Two primer pairs were used.
  • FIGs.8A and 8B provide graphs depicting the percent (%) of human STAT3 mRNA remaining in Huh7 cells endogenously expressing human STAT3, after 24-hour treatment with 0.05nM, 0.3nM, or 1nM of DsiRNA targeting various regions of the STAT3 gene.48 GalNAc-conjugated STAT3 oligonucleotides were assayed in FIG.8A and 34 of those oligonucleotides were selected for further testing in vivo (FIG.8B).
  • FIGs.9A and 9B provide graphs depicting the percent (%) of human STAT3 mRNA remaining in liver of mice exogenously expressing human STAT3 (hydrodynamic injection model) after treatment with GalNAc-conjugated STAT3 oligonucleotides.
  • Mice were dosed subcutaneously with 1mg/kg of the indicated GalNAc-STAT3 oligonucleotides formulated in PBS.
  • Three days post-dose mice were hydrodynamically injected (HDI) with a DNA plasmid encoding human STAT3.
  • the level of human STAT3 mRNA was determined from livers collected 18 hours after injection. Arrows indicate oligonucleotides selected for dose response analysis.
  • FIG.10 provides a graph depicting the dose response of GalNAc-conjugated STAT3 oligonucleotides.
  • the level of human STAT3 mRNA was determined from livers collected 18 hours after injection with plasmid encoding human STAT3.
  • FIG.11 provides a graph depicting the normalized (to Ppib) relative mouse STAT3 mRNA remaining in liver of mice endogenously expressing mouse STAT3 after treatment with GalNAc-conjugated STAT3 oligonucleotides. Mice were dosed subcutaneously with 3mg/kg of the indicated GalNAc-STAT3 oligonucleotides formulated in PBS. Five days post-dose liver was collected and the level of mouse STAT3 mRNA was determined. Arrows indicate top oligonucleotides and those selected for dose response study.
  • FIG.12 provides a graph depicting the normalized (to Ppib) relative mouse STAT3 mRNA remaining in liver of mice endogenously expressing mouse STAT3 after treatment with GalNAc-conjugated STAT3 oligonucleotides.
  • Mice were dosed subcutaneously with 3mg/kg of the indicated GalNAc-STAT3 oligonucleotides formulated in PBS. Five days post-dose liver was collected and the level of mouse STAT3 mRNA was determined. Arrows indicate oligonucleotides selected for dose response study.
  • FIGs.13A and 13B provide graphs depicting the dose response of GalNAc- conjugated STAT3 oligonucleotides.
  • FIG.14 provides a graph depicting the percent (%) of human STAT3 mRNA remaining in liver of mice exogenously expressing human STAT3 (hydrodynamic injection model) after treatment with GalNAc-conjugated STAT3 oligonucleotides.
  • FIG.15 provides a graph depicting the dose response of GalNAc-conjugated STAT3 oligonucleotides.
  • mice The percent (%) of human STAT3 mRNA remaining in liver of mice exogenously expressing human STAT3 (hydrodynamic injection model) after treatment with GalNAc-conjugated STAT3 oligonucleotides.
  • Mice were dosed subcutaneously with three doses (0.3mg/kg, 1mg/kg, and 3mg/kg) of the indicated GalNAc-STAT3 oligonucleotides formulated in PBS.
  • Three days post-dose mice were hydrodynamically injected (HDI) with a DNA plasmid encoding human STAT3.
  • the level of human STAT3 mRNA was determined from livers collected 18 hours after injection.
  • FIG.16 provides a graph depicting the dose response of GalNAc-conjugated STAT3 oligonucleotides. The percent (%) of human STAT3 mRNA remaining in liver of mice exogenously expressing human STAT3 (hydrodynamic injection model) after treatment with GalNAc-conjugated STAT3 oligonucleotides. Mice were dosed subcutaneously with two doses (0.3mg/kg and 1mg/kg) of the indicated GalNAc-STAT3 oligonucleotides formulated in PBS.
  • FIG.17 provides a graph depicting the percent (%) remaining human STAT1 mRNA in Huh7 cells endogenously expressing STAT3 and STAT1 treated with GalNAc- conjugated STAT3 oligonucleotides. Cells were treated for 24 hours with three doses (0.05nM, 0.3nM, and 1nM) of oligonucleotide.
  • FIG.18A provides a graph depicting tumor volume after administration of a GalXC-STAT3-C18 oligonucleotide alone or in combination with an anti-PD-L1 mAb.
  • Immunocompetent mice bearing Pan02 murine pancreatic tumors were dosed subcutaneously (s.c.) with 25 mg/kg of GalXC-STAT3-C18-4123 with intraperitoneal (i.p.) treatment of 10 mg/kg of anti-PD-L1 mAb.
  • FIG.18B provides a graph depicting tumor volume after administration of GalXC-STAT3-C18 oligonucleotide in combination with anti-PD-L1 mAb.
  • Placebo treated mice from FIG.18A were dosed subcutaneously (s.c.) with 25 mg/kg of GalXC-STAT3- C18-4123 with intraperitoneal (i.p.) treatment of 10 mg/kg of anti-PD-L1 mAb at Day 59.
  • FIGs.19A-19C provide graphs depicting tumor volume after administration of a GalXC-STAT3-C18 oligonucleotide alone or in combination with an anti-PD-L1 mAb or GalXC-Placebo alone or in combination with anti-PD-L1 mAb in tumors with different immunophenotypes.4T1 (triple negative breast, checkpoint resistant) (FIG.19A), MC-38 (Colon carcinoma, partially checkpoint sensitive) (FIG.19B), or Hepa1-6 (Hepatocellular carcinoma, checkpoint sensitive) (FIG.19C) cells were implanted into mice. Tumor bearing mice were dosed s.c.
  • FIG.20 provides a graph depicting the effect of Hepa1-6 re-challenge in the completely eradicated tumors.
  • mice were rechallenged on day 51 with Hepa1-6 cells (2e6 cells/mouse) on the opposite flank of the mice and tumor volume was monitored (FIG.20).
  • Arrow (5/5 CR) All mice remained tumor free even after the re-challenge.
  • FIG.21 provides a graph depicting tumor volume after administration of GalXC- STAT3-C18 oligonucleotide alone or in combination with an anti-PD-L1 mAb in immunocompromised mice with no functional CD8+ T cells.
  • Mice bearing 4T1 tumors were dosed s.c. with GalXC-STAT3-C18-4123 (25 mg/kg, three times with each dose three days apart (q3d x 3)) and i.p. with anti-PD-L1 mAb (10 mg/kg, q3d x 3).
  • FIG.22 provides images showing the appearance of tumors (with cell death) from mice assayed in FIG.19A, and perforin staining for positive cytotoxic CD8+ T cells in the tumors at the end of the study.
  • FIG.23 provides images showing lung tumor metastasis after administration of GalXC-STAT3-C18-4123 oligonucleotide alone or in combination with an anti-PD-L1 mAb as treated in FIGs.19A and 21.
  • mice (immunocompetent or immunocompromised) bearing 4T1 tumors were dosed s.c. with GalXC-STAT3-C18-4123 (50 mg/kg, q3d x 3) and i.p. with anti-PD-L1 mAb (10 mg/kg, q3d x 3).
  • Controls included GalXC-Placebo, GalXC-STAT3- C18-4123 at 50 mg/kg or GalXC-Placebo at 50 mg/kg in combination with anti-PD-L1 mAb at 10 mg/kg.
  • FIG.24 provides a heat map showing the regulation of targets involved in immune modulation observed in CT26 tumors upon combination treatment of GalXC- STAT3-C18-4123 (s.c, 25 mg/kg, q3d x 3) and anti-PD-L1 mAb (i.p. at 10 mg/kg, q3d x 3) compared to controls including GalXC-Placebo, GalXC-STAT3-C18-4123 at 25 mg/kg or GalXC-Placebo at 25 mg/kg in combination with anti-PD-L1 mAb at 10 mg/kg.
  • the term “and/or” includes any and all combinations of one or more of the associated listed items. Further, the singular forms and the articles “a”, “an” and “the” are intended to include the plural forms as well, unless expressly stated otherwise. It will be further understood that the terms: includes, comprises, including and/or comprising, when used in this specification, specify the presence of stated features, integers, steps, operations, elements, and/or components, but do not preclude the presence or addition of one or more other features, integers, steps, operations, elements, components, and/or groups thereof.
  • PCR polymerase chain reaction
  • LCR ligase chain reaction
  • Q.beta.-replicase amplification RNA polymerase mediated techniques
  • NASBA RNA polymerase mediated techniques
  • cancer or tumor includes, but is not limited to, solid tumors and blood borne tumors. These terms include diseases of the skin, tissues, organs, bone, cartilage, blood, and vessels. These terms further encompass primary and metastatic cancers.
  • PD-1 refers to a protein found on T cells that helps keep the immune responses in check. When PD-1 is bound to another protein called PD-L1, it helps keep T cells from killing other cells, including cancer cells. Some anticancer drugs, called immune checkpoint inhibitors, are used to block PD-1.
  • STAT3 refers to Signal transducer and activator of transcription 3 (STAT3) which is a transcription factor which in humans is encoded by the STAT3 gene (STAT3 Human (Hs) NM_001369512.1 Genbank RefSeq #, or NM_139276.3). STAT3 mediates the expression of a variety of genes in response to cell stimuli, and thus plays a key role in many cellular processes such as cell growth and apoptosis, as well as the growth and progression of cancer.
  • STAT3 Signal transducer and activator of transcription 3
  • the term "cold tumor” or “non-inflamed tumor” refers to a tumor or tumor microenvironment wherein there is minimal to no presence of anti-tumor immune cells, such as tumor infiltrating lymphocytes (TILs), and/or contain cell subsets associated with immune suppression including regulatory T cells (Treg), myeloid-derived suppressor cells (MDSCs) and M2 macrophages.
  • TILs tumor infiltrating lymphocytes
  • a cold tumor is characterized by a low number or even absence of infiltration of anti-tumor immune cells that such cells may be present but remain stuck in the surrounding stroma, thus unable to colonize the tumor microenvironment to provide their antitumor functions.
  • complementary refers to a structural relationship between two nucleotides (e.g., on two opposing nucleic acids or on opposing regions of a single nucleic acid strand) that permits the two nucleotides to form base pairs with one another.
  • a purine nucleotide of one nucleic acid that is complementary to a pyrimidine nucleotide of an opposing nucleic acid may base pair together by forming hydrogen bonds with one another.
  • complementary nucleotides can base pair in the Watson-Crick manner or in any other manner that allows for the formation of stable duplexes.
  • two nucleic acids may have regions of multiple nucleotides that are complementary with each other to form regions of complementarity, as described herein.
  • “species cross-reactive oligonucleotide” refers to an oligonucleotide capable of inhibiting expression of a target mRNA in more than one species.
  • a species cross-reactive oligonucleotide is capable of inhibiting expression of a target mRNA in human and non-human primates.
  • Example species include but is not limited to human, non-human primates, mouse, and rat.
  • species cross-reactive oligonucleotides are capable of targeting and inhibiting mRNA in at least two, at least three, or at least four species.
  • deoxyribonucleotide refers to a nucleotide having a hydrogen in place of a hydroxyl at the 2′ position of its pentose sugar when compared with a ribonucleotide.
  • a modified deoxyribonucleotide is a deoxyribonucleotide having one or more modifications or substitutions of atoms other than at the 2′ position, including modifications or substitutions in or of the sugar, phosphate group or base.
  • double-stranded RNA or “dsRNA” refers to an RNA oligonucleotide that is substantially in a duplex form.
  • the complementary base-pairing of duplex region(s) of a dsRNA oligonucleotide is formed between antiparallel sequences of nucleotides of covalently separate nucleic acid strands.
  • complementary base-pairing of duplex region(s) of a dsRNA formed between antiparallel sequences of nucleotides of nucleic acid strands that are covalently linked.
  • complementary base-pairing of duplex region(s) of a dsRNA is formed from single nucleic acid strand that is folded (e.g., via a hairpin) to provide complementary antiparallel sequences of nucleotides that base pair together.
  • a dsRNA comprises two covalently separate nucleic acid strands that are fully duplexed with one another.
  • a dsRNA comprises two covalently separate nucleic acid strands that are partially duplexed (e.g., having overhangs at one or both ends).
  • a dsRNA comprises antiparallel sequence of nucleotides that are partially complementary, and thus, may have one or more mismatches, which may include internal mismatches or end mismatches.
  • duplex in reference to nucleic acids (e.g., oligonucleotides), refers to a structure formed through complementary base pairing of two antiparallel sequences of nucleotides.
  • excipient refers to a non-therapeutic agent that may be included in a composition, for example, to provide or contribute to a desired consistency or stabilizing effect.
  • the term "hot tumor” or “inflamed tumor” refers to a tumor or tumor microenvironment wherein there is a considerable presence of anti-tumor immune cells especially TILs and thus are typically immuno-stimulatory.
  • “loop” refers to an unpaired region of a nucleic acid (e.g., oligonucleotide) that is flanked by two antiparallel regions of the nucleic acid that are sufficiently complementary to one another, such that under appropriate hybridization conditions (e.g., in a phosphate buffer, in a cells), the two antiparallel regions, which flank the unpaired region, hybridize to form a duplex (referred to as a “stem”).
  • a nucleic acid e.g., oligonucleotide
  • the loop may refer to a loop comprising four nucleotides as a tetraloop (tetraL).
  • the loop may refer to a loop comprising three nucleotides as a triloop (triL).
  • modified internucleotide linkage refers to an internucleotide linkage having one or more chemical modifications when compared with a reference internucleotide linkage comprising a phosphodiester bond.
  • a modified nucleotide is a non-naturally occurring linkage.
  • a modified internucleotide linkage confers one or more desirable properties to a nucleic acid in which the modified internucleotide linkage is present.
  • modified nucleotide may improve thermal stability, resistance to degradation, nuclease resistance, solubility, bioavailability, bioactivity, reduced immunogenicity, etc.
  • modified nucleotide refers to a nucleotide having one or more chemical modifications when compared with a corresponding reference nucleotide selected from: adenine ribonucleotide, guanine ribonucleotide, cytosine ribonucleotide, uracil ribonucleotide, adenine deoxyribonucleotide, guanine deoxyribonucleotide, cytosine deoxyribonucleotide and thymidine deoxyribonucleotide.
  • a modified nucleotide is a non-naturally occurring nucleotide.
  • a modified nucleotide has one or more chemical modification in its sugar, nucleobase and/or phosphate group.
  • a modified nucleotide has one or more chemical moieties conjugated to a corresponding reference nucleotide.
  • a modified nucleotide confers one or more desirable properties to a nucleic acid in which the modified nucleotide is present.
  • a modified nucleotide may improve thermal stability, resistance to degradation, nuclease resistance, solubility, bioavailability, bioactivity, reduced immunogenicity, etc.
  • RNAi oligonucleotide refers to a structure of a RNAi oligonucleotide that is characterized by separate sense (passenger) and antisense (guide) strands, in which the sense strand has a region of complementarity with the antisense strand, and in which at least one of the strands, generally the sense strand, has a tetraloop configured to stabilize an adjacent stem region formed within the at least one strand.
  • oligonucleotide refers to a short nucleic acid (e.g., less than about 100 nucleotides in length).
  • An oligonucleotide may be single stranded (ss) or double- stranded (ds). An oligonucleotide may or may not have duplex regions. An oligonucleotide may comprise deoxyribonucleotides, ribonucleosides, or a combination of both. In some embodiments, a double-stranded oligonucleotide comprising ribonucleotides is referred to as “dsRNA”.
  • an oligonucleotide may be, but is not limited to, a small interfering RNA (siRNA), microRNA (miRNA), short hairpin RNA (shRNA), dicer substrate interfering RNA (dsiRNA), antisense oligonucleotide, short siRNA or ss siRNA.
  • a double-stranded RNA dsRNA is an RNAi oligonucleotide.
  • RNAi oligonucleotide conjugate and “oligonucleotide-ligand conjugate” are used interchangeably and refer to an oligonucleotide comprising one or more nucleotides conjugated with one or more targeting ligands.
  • overhang refers to terminal non-base pairing nucleotide(s) resulting from one strand or region extending beyond the terminus of a complementary strand with which the one strand or region forms a duplex.
  • an overhang comprises one or more unpaired nucleotides extending from a duplex region at the 5′ terminus or 3′ terminus of a dsRNA.
  • the overhang is a 3′ or 5′ overhang on the antisense strand or sense strand of a dsRNA.
  • phosphate analog refers to a chemical moiety that mimics the electrostatic and/or steric properties of a phosphate group.
  • a phosphate analog is positioned at the 5′ terminal nucleotide of an oligonucleotide in place of a 5′-phosphate, which is often susceptible to enzymatic removal.
  • a 5′ phosphate analog contains a phosphatase-resistant linkage.
  • phosphate analogs include, but are not limited to, 5′ phosphonates, such as 5′ methylene phosphonate (5′-MP) and 5′-(E)-vinylphosphonate (5′-VP).
  • an oligonucleotide has a phosphate analog at a 4′-carbon position of the sugar (referred to as a “4′-phosphate analog”) at a 5′-terminal nucleotide.
  • An example of a 4′-phosphate analog is oxymethylphosphonate, in which the oxygen atom of the oxymethyl group is bound to the sugar moiety (e.g., at its 4′- carbon) or analog thereof.
  • reduced expression of a gene refers to a decrease in the amount or level of RNA transcript (e.g., STAT3 mRNA) or protein encoded by the gene and/or a decrease in the amount or level of activity of the gene in a cell, a population of cells, a sample, or a subject, when compared to an appropriate reference (e.g., a reference cell, population of cells, sample, or subject).
  • an appropriate reference e.g., a reference cell, population of cells, sample, or subject.
  • the act of contacting a cell with an oligonucleotide herein may result in a decrease in the amount or level of STAT3 mRNA, protein and/or activity (e.g., via degradation of STAT3 mRNA by the RNAi pathway) when compared to a cell that is not treated with the dsRNA.
  • reducing expression refers to an act that results in reduced expression of a gene (e.g., STAT3).
  • “reduction of STAT3 expression” refers to a decrease in the amount or level of STAT3 mRNA, STAT3 protein and/or STAT3 activity in a cell, a population of cells, a sample or a subject when compared to an appropriate reference (e.g., a reference cell, population of cells, sample, or subject).
  • region of complementarity refers to a sequence of nucleotides of a nucleic acid (e.g., a dsRNA) that is sufficiently complementary to an antiparallel sequence of nucleotides to permit hybridization between the two sequences of nucleotides under appropriate hybridization conditions (e.g., in a phosphate buffer, in a cell, etc.).
  • an oligonucleotide herein comprises a targeting sequence having a region of complementary to a mRNA target sequence.
  • ribonucleotide refers to a nucleotide having a ribose as its pentose sugar, which contains a hydroxyl group at its 2′ position.
  • a modified ribonucleotide is a ribonucleotide having one or more modifications or substitutions of atoms other than at the 2′ position, including modifications or substitutions in or of the ribose, phosphate group or base.
  • RNAi oligonucleotide refers to either (a) a dsRNA having a sense strand (passenger) and antisense strand (guide), in which the antisense strand or part of the antisense strand is used by the Argonaute 2 (Ago2) endonuclease in the cleavage of a target mRNA or (b) a ss oligonucleotide having a single antisense strand, where that antisense strand (or part of that antisense strand) is used by the Ago2 endonuclease in the cleavage of a target mRNA.
  • Ago2 Argonaute 2
  • strand refers to a single, contiguous sequence of nucleotides linked together through internucleotide linkages (e.g., phosphodiester linkages or phosphorothioate linkages). In some embodiments, a strand has two free ends (e.g., a 5′ end and a 3′ end).
  • subject means any mammal, including mice, rabbits, non- human primates (NHP), and humans. In one embodiment, the subject is a human or NHP.
  • “synthetic” refers to a nucleic acid or other molecule that is artificially synthesized (e.g., using a machine (e.g., a solid-state nucleic acid synthesizer)) or that is otherwise not derived from a natural source (e.g., a cell or organism) that normally produces the molecule.
  • targeting ligand refers to a molecule or “moiety” (e.g., a carbohydrate, amino sugar, cholesterol, polypeptide, or lipid) that selectively binds to a cognate molecule (e.g., a receptor) of a tissue or cell of interest and/or that is conjugatable to another substance for purposes of targeting the other substance to the tissue or cell of interest.
  • a targeting ligand may be conjugated to an oligonucleotide for purposes of targeting the oligonucleotide to a specific tissue or cell of interest.
  • a targeting ligand selectively binds to a cell surface receptor.
  • a targeting ligand when conjugated to an oligonucleotide facilitates delivery of the oligonucleotide into a particular cell through selective binding to a receptor expressed on the surface of the cell and endosomal internalization by the cell of the complex comprising the oligonucleotide, targeting ligand and receptor.
  • a targeting ligand is conjugated to an oligonucleotide via a linker that is cleaved following or during cellular internalization such that the oligonucleotide is released from the targeting ligand in the cell.
  • loop refers to a loop that increases stability of an adjacent duplex formed by hybridization of flanking sequences of nucleotides.
  • the increase in stability is detectable as an increase in melting temperature (Tm) of an adjacent stem duplex that is higher than the T m of the adjacent stem duplex expected, on average, from a set of loops of comparable length consisting of randomly selected sequences of nucleotides.
  • Tm melting temperature
  • a loop e.g., a tetraloop or triloop
  • a T m of at least about 50°C, at least about 55°C, at least about 56°C, at least about 58°C, at least about 60°C, at least about 65°C or at least about 75°C in 10 mM NaHPO 4 to a hairpin comprising a duplex of at least 2 base pairs (bp) in length.
  • a loop e.g., a tetraloop
  • a loop comprises or consists of 3 to 6 nucleotides and is typically 4 to 5 nucleotides. In certain embodiments, a loop comprises or consists of 3, 4, 5 or 6 nucleotides, which may or may not be modified (e.g., which may or may not be conjugated to a targeting moiety).
  • a tetraloop comprises or consists of 3 to 6 nucleotides and is typically 4 to 5 nucleotides. In certain embodiments, a tetraloop comprises or consists of 3, 4, 5 or 6 nucleotides, which may or may not be modified (e.g., which may or may not be conjugated to a targeting moiety). In one embodiment, a loop consisting of 4 nucleotides is a tetraloop. Any nucleotide may be used in the loop (e.g., a tetraloop) and standard IUPAC- IUB symbols for such nucleotides may be used as described in Cornish-Bowden ((1985) NUCLEIC ACIDS RES.13:3021-3030).
  • the letter “N” may be used to mean that any base may be in that position
  • the letter “R” may be used to show that A (adenine) or G (guanine) may be in that position
  • “B” may be used to show that C (cytosine), G (guanine), or T (thymine) may be in that position.
  • tetraloops include the UNCG family of tetraloops (e.g., UUCG), the GNRA family of tetraloops (e.g., GAAA), and the CUUG tetraloop (Woese et al., (1990) PROC. NATL. ACAD. SCI.
  • DNA tetraloops include the d(GNNA) family of tetraloops (e.g., d(GTTA), the d(GNRA)) family of tetraloops, the d(GNAB) family of tetraloops, the d(CNNG) family of tetraloops, and the d(TNCG) family of tetraloops (e.g., d(TTCG)).
  • the tetraloop is contained within a nicked tetraloop structure.
  • treat refers to the act of providing care to a subject in need thereof, for example, by administering a therapeutic agent (e.g., an oligonucleotide herein) to the subject, for purposes of improving the health and/or well-being of the subject with respect to an existing condition (e.g., a disease, disorder) or to prevent or decrease the likelihood of the occurrence of a condition.
  • a therapeutic agent e.g., an oligonucleotide herein
  • treatment involves reducing the frequency or severity of at least one sign, symptom or contributing factor of a condition (e.g., disease, disorder) experienced by a subject.
  • tumor microenvironment relates to the cellular environment in which any given tumor exists, including the tumor stroma, surrounding blood vessels, immune cells, fibroblasts, other cells, signaling molecules, and the ECM. It is understood that the tumor microenvironment harbors and/or surrounds the tumor cells with which it interacts.
  • Methods of Use Combination of STAT3 Oligonucleotide and PD-L1 Inhibitors [00116] In some embodiments, the disclosure provides STAT3 oligonucleotides for use, or adaptable for use, to treat a subject (e.g., a human having a disease, disorder or condition associated with STAT3 expression) that has received or is receiving a PD-L1 inhibitor.
  • methods described herein comprise selecting a subject having a disease, disorder or condition associated with STAT3 expression and/or PD-L1 expression or is predisposed to the same.
  • the methods can include selecting an individual having a marker for a disease associated with STAT3 expression and/or PD-L1 expression such as cancer or other chronic lymphoproliferative disorders.
  • the methods also may include steps such as measuring or obtaining a baseline value for a marker of STAT3 expression and/or PD-L1 expression, and then comparing such obtained value to one or more other baseline values or values obtained after being administered the oligonucleotide to assess the effectiveness of treatment.
  • the disclosure provides methods of treating a subject having, suspected of having, or at risk of developing a disease, disorder, or condition with a STAT3 oligonucleotide herein, wherein the subject has received or is receiving a PD-L1 inhibitor. In some embodiments, the disclosure provides methods of treating a subject having, suspected of having, or at risk of developing a disease, disorder, or condition with a PD-L1 inhibitor described herein, wherein the subject has received or is receiving a STAT3 oligonucleotide described herein.
  • the disclosure provides methods of treating or attenuating the onset or progression of a disease, disorder or condition associated with STAT3 expression using a STAT3 oligonucleotide herein in combination with a PD-L1 inhibitor.
  • the disclosure provides methods to achieve one or more therapeutic benefits in a subject having a disease, disorder or condition associated with STAT3 expression using a STAT3 oligonucleotide herein in combination with a PD-L1 inhibitor.
  • the subject is treated by administering a therapeutically effective amount of a STAT3 oligonucleotide herein in combination with a PD-L1 inhibitor.
  • the subject is treated by administering a therapeutically effective amount of a STAT3 oligonucleotide herein to a subject that has received or is receiving a PD-L1 inhibitor. In some embodiments of the methods herein, the subject is treated by administering a therapeutically effective amount of a PD-L1 inhibitor to a subject that has received or is receiving a STAT3 oligonucleotide herein. In some embodiments, the subject is treated therapeutically. In some embodiments, the subject is treated prophylactically.
  • one or more STAT3 oligonucleotides herein, or a pharmaceutical composition comprising one or more STAT3 oligonucleotides is administered to a subject having a disease, disorder or condition associated with STAT3 expression that has received or is receiving a PD-L1 inhibitor, such that STAT3 expression is reduced in the subject, thereby treating the subject.
  • an amount or level of STAT3 mRNA is reduced in the subject.
  • an amount or level of STAT3 and/or protein is reduced in the subject.
  • one or more STAT3 oligonucleotides herein, or a pharmaceutical composition comprising one or more STAT3 oligonucleotides is administered to a subject having a disease, disorder or condition associated with STAT3 expression that has received or is receiving a PD-L1 inhibitor such that STAT3 expression and PD-L1 signaling is reduced in the subject, thereby treating the subject.
  • an amount or level of STAT3 mRNA and PD-L1 signaling is reduced in the subject.
  • an amount or level of STAT3 and/or protein is reduced in the subject and PD-L1 signaling is reduced in the subject.
  • a therapeutically effective amount of a STAT3 oligonucleotide and/or PD-L1 inhibitor is administered to a subject.
  • a therapeutically acceptable amount may be an amount that can therapeutically treat a disease or disorder.
  • the appropriate dosage for any one subject will depend on certain factors, including the subject′s size, body surface area, age, the particular composition to be administered, the active ingredient(s) in the composition, time and route of administration, general health, and other drugs being administered concurrently.
  • a subject is administered any one of the compositions herein either enterally (e.g., orally, by gastric feeding tube, by duodenal feeding tube, via gastrostomy or rectally), parenterally (e.g., subcutaneous injection, intravenous injection or infusion, intra-arterial injection or infusion, intraosseous infusion, intramuscular injection, intracerebral injection, intracerebroventricular injection, intrathecal), topically (e.g., epicutaneous, inhalational, via eye drops, or through a mucous membrane), or by direct injection into a target organ (e.g., the liver of a subject).
  • oligonucleotides herein are administered intravenously or subcutaneously.
  • the oligonucleotides herein would typically be administered quarterly (once every three months), bi-monthly (once every two months), monthly or weekly.
  • the oligonucleotides may be administered every week or at intervals of two, or three weeks.
  • the oligonucleotides may be administered daily.
  • a subject is administered one or more loading doses of the oligonucleotide followed by one or more maintenance doses of the oligonucleotide.
  • a PD-L1 inhibitor e.g., an anti-PD-L1 antibody
  • a PD-L1 inhibitor is administered quarterly (once every three months), bi-monthly (once every two months), monthly or weekly.
  • the inhibitor is administered every week or at intervals of two, or three weeks.
  • the inhibitor is administered daily.
  • the oligonucleotides herein are administered in combination with a PD-L1 inhibitor.
  • the oligonucleotide and inhibitor are administered in combination concurrently, sequentially (in any order), or intermittently.
  • the oligonucleotide and inhibitor may be co-administered concurrently.
  • the oligonucleotide may be administered and followed any amount of time later (e.g., one hour, one day, one week or one month) by the administration of the inhibitor, or vice versa.
  • the subject to be treated is a human or non-human primate or other mammalian subject.
  • Other exemplary subjects include domesticated animals such as dogs and cats; livestock such as horses, cattle, pigs, sheep, goats, and chickens; and animals such as mice, rats, guinea pigs, and hamsters.
  • the disclosure provides a method of treating cancer in a subject that has received or is receiving a PD-L1 inhibitor, the method comprising administering an RNAi oligonucleotide comprising a sense strand and an antisense strand, wherein the sense strand and antisense strand comprise nucleotide sequences selected from the group consisting of: (a) SEQ ID NOs: 9 and 10, respectively; (b) SEQ ID NOs: 37 and 38, respectively; (c) SEQ ID NOs: 65 and 66, respectively; and (d) SEQ ID NOs: 69 and 70, respectively, thereby treating cancer in the subject.
  • the disclosure provides a method of treating cancer in a subject that has received or is receiving a PD-L1 inhibitor, the method comprising administering an RNAi oligonucleotide comprising a sense strand and an antisense strand, wherein the sense strand and antisense strand comprise nucleotide sequences selected from the group consisting of: (a) SEQ ID NOs: 9 and 10, respectively; (b) SEQ ID NOs: 37 and 38, respectively; (c) SEQ ID NOs: 65 and 66, respectively; and (d) SEQ ID NOs: 69 and 70, respectively, and wherein a lipid moiety is conjugated to a nucleotide of the sense strand, thereby treating cancer in the subject.
  • the disclosure provides a method of treating cancer in a subject that has received or is receiving a PD-L1 inhibitor, the method comprising administering an RNAi oligonucleotide comprising a sense strand and an antisense strand, wherein the sense strand comprises the nucleotide sequence of SEQ ID NO: 9 and antisense strand comprises the nucleotide sequence of SEQ ID NO: 10, thereby treating cancer in the subject.
  • the disclosure provides a method of treating cancer in a subject that has received or is receiving a PD-L1 inhibitor, the method comprising administering an RNAi oligonucleotide comprising a sense strand and an antisense strand, wherein the sense strand comprises the nucleotide sequence of SEQ ID NO: 9 and antisense strand comprises the nucleotide sequence of SEQ ID NO: 10, and wherein a lipid moiety is conjugated to a nucleotide of the sense strand, thereby treating cancer in the subject.
  • the disclosure provides a method of treating cancer in a subject that has received or is receiving a PD-L1 inhibitor, the method comprising administering an RNAi oligonucleotide comprising a sense strand and an antisense strand, wherein the sense strand comprises the nucleotide sequence of SEQ ID NO: 37 and antisense strand comprises the nucleotide sequence of SEQ ID NO: 38, thereby treating cancer in the subject.
  • the disclosure provides a method of treating cancer in a subject that has received or is receiving a PD-L1 inhibitor, the method comprising administering an RNAi oligonucleotide comprising a sense strand and an antisense strand, wherein the sense strand comprises the nucleotide sequence of SEQ ID NO: 37 and antisense strand comprises the nucleotide sequence of SEQ ID NO: 38, and wherein a lipid moiety is conjugated to a nucleotide of the sense strand, thereby treating cancer in the subject.
  • the disclosure provides a method of treating cancer in a subject that has received or is receiving a PD-L1 inhibitor, the method comprising administering an RNAi oligonucleotide comprising a sense strand and an antisense strand, wherein the sense strand comprises the nucleotide sequence of SEQ ID NO: 65 and antisense strand comprises the nucleotide sequence of SEQ ID NO: 66, thereby treating cancer in the subject.
  • the disclosure provides a method of treating cancer in a subject that has received or is receiving a PD-L1 inhibitor, the method comprising administering an RNAi oligonucleotide comprising a sense strand and an antisense strand, wherein the sense strand comprises the nucleotide sequence of SEQ ID NO: 65 and antisense strand comprises the nucleotide sequence of SEQ ID NO: 66, and wherein a lipid moiety is conjugated to a nucleotide of the sense strand, thereby treating cancer in the subject.
  • the disclosure provides a method of treating cancer in a subject that has received or is receiving a PD-L1 inhibitor, the method comprising administering an RNAi oligonucleotide comprising a sense strand and an antisense strand, wherein the sense strand comprises the nucleotide sequence of SEQ ID NO: 69 and antisense strand comprises the nucleotide sequence of SEQ ID NO: 70, thereby treating cancer in the subject.
  • the disclosure provides a method of treating cancer in a subject that has received or is receiving a PD-L1 inhibitor, the method comprising administering an RNAi oligonucleotide comprising a sense strand and an antisense strand, wherein the sense strand comprises the nucleotide sequence of SEQ ID NO: 69 and antisense strand comprises the nucleotide sequence of SEQ ID NO: 70, and wherein a lipid moiety is conjugated to a nucleotide of the sense strand, thereby treating cancer in the subject.
  • the disclosure provides a method of treating cancer in a subject that has received or is receiving an RNAi oligonucleotide, the method comprising administering a PD-L1 inhibitor, wherein the oligonucleotide comprises a sense strand and an antisense strand, wherein the sense strand and antisense strand comprise nucleotide sequences selected from the group consisting of: (a) SEQ ID NOs: 9 and 10, respectively; (b) SEQ ID NOs: 37 and 38, respectively; (c) SEQ ID NOs: 65 and 66, respectively; and (d) SEQ ID NOs: 69 and 70, respectively, thereby treating cancer in the subject.
  • the disclosure provides a method of treating cancer in a subject that has received or is receiving an RNAi oligonucleotide, the method comprising administering a PD-L1 inhibitor, wherein the oligonucleotide comprises a sense strand and an antisense strand, wherein the sense strand and antisense strand comprise nucleotide sequences selected from the group consisting of: (a) SEQ ID NOs: 9 and 10, respectively; (b) SEQ ID NOs: 37 and 38, respectively; (c) SEQ ID NOs: 65 and 66, respectively; and (d) SEQ ID NOs: 69 and 70, respectively, and wherein a lipid moiety is conjugated to a nucleotide of the sense strand, thereby treating cancer in the subject.
  • the disclosure provides a method of treating cancer in a subject that has received or is receiving an RNAi oligonucleotide, the method comprising administering a PD-L1 inhibitor, wherein the oligonucleotide comprises a sense strand and an antisense strand, wherein the sense strand comprises the nucleotide sequence of SEQ ID NO: 9 and antisense strand comprises the nucleotide sequence of SEQ ID NO: 10, thereby treating cancer in the subject.
  • the disclosure provides a method of treating cancer in a subject that has received or is receiving an RNAi oligonucleotide, the method comprising administering a PD-L1 inhibitor, wherein the oligonucleotide comprises a sense strand and an antisense strand, wherein the sense strand comprises the nucleotide sequence of SEQ ID NO: 9 and antisense strand comprises the nucleotide sequence of SEQ ID NO: 10, and wherein a lipid moiety is conjugated to a nucleotide of the sense strand, thereby treating cancer in the subject.
  • the disclosure provides a method of treating cancer in a subject that has received or is receiving an RNAi oligonucleotide, the method comprising administering a PD-L1 inhibitor, wherein the oligonucleotide comprises a sense strand and an antisense strand, wherein the sense strand comprises the nucleotide sequence of SEQ ID NO: 37 and antisense strand comprises the nucleotide sequence of SEQ ID NO: 38, thereby treating cancer in the subject.
  • the disclosure provides a method of treating cancer in a subject that has received or is receiving an RNAi oligonucleotide, the method comprising administering a PD-L1 inhibitor, wherein the oligonucleotide comprises a sense strand and an antisense strand, wherein the sense strand comprises the nucleotide sequence of SEQ ID NO: 37 and antisense strand comprises the nucleotide sequence of SEQ ID NO: 38, and wherein a lipid moiety is conjugated to a nucleotide of the sense strand, thereby treating cancer in the subject.
  • the disclosure provides a method of treating cancer in a subject that has received or is receiving an RNAi oligonucleotide, the method comprising administering a PD-L1 inhibitor, wherein the oligonucleotide comprises a sense strand and an antisense strand, wherein the sense strand comprises the nucleotide sequence of SEQ ID NO: 65 and antisense strand comprises the nucleotide sequence of SEQ ID NO: 66, thereby treating cancer in the subject.
  • the disclosure provides a method of treating cancer in a subject that has received or is receiving an RNAi oligonucleotide, the method comprising administering a PD-L1 inhibitor, wherein the oligonucleotide comprises a sense strand and an antisense strand, wherein the sense strand comprises the nucleotide sequence of SEQ ID NO: 65 and antisense strand comprises the nucleotide sequence of SEQ ID NO: 66, and wherein a lipid moiety is conjugated to a nucleotide of the sense strand, thereby treating cancer in the subject.
  • the disclosure provides a method of treating cancer in a subject that has received or is receiving an RNAi oligonucleotide, the method comprising administering a PD-L1 inhibitor, wherein the oligonucleotide comprises a sense strand and an antisense strand, wherein the sense strand comprises the nucleotide sequence of SEQ ID NO: 69 and antisense strand comprises the nucleotide sequence of SEQ ID NO: 70, thereby treating cancer in the subject.
  • the disclosure provides a method of treating cancer in a subject that has received or is receiving an RNAi oligonucleotide, the method comprising administering a PD-L1 inhibitor, wherein the oligonucleotide comprises a sense strand and an antisense strand, wherein the sense strand comprises the nucleotide sequence of SEQ ID NO: 69 and antisense strand comprises the nucleotide sequence of SEQ ID NO: 70, and wherein a lipid moiety is conjugated to a nucleotide of the sense strand, thereby treating cancer in the subject.
  • the disclosure provides a method of treating a disease, disorder or condition associated with activated STAT3 expression, comprising administering to a subject in need thereof an RNAi oligonucleotide, wherein the subject has received or is receiving a PD-L1 inhibitor, wherein the oligonucleotide comprises a sense strand and an antisense strand, wherein the sense strand and antisense strand comprise nucleotide sequences selected from the group consisting of: (a) SEQ ID NOs: 9 and 10, respectively; (b) SEQ ID NOs: 37 and 38, respectively; (c) SEQ ID NOs: 65 and 66, respectively; and (d) SEQ ID NOs: 69 and 70, respectively, thereby treating cancer in the subject.
  • the disclosure provides a method of treating a disease, disorder or condition associated with activated STAT3 expression, comprising administering to a subject in need thereof an RNAi oligonucleotide, wherein the subject has received or is receiving a PD-L1 inhibitor, wherein the oligonucleotide comprises a sense strand and an antisense strand, wherein the sense strand and antisense strand comprise nucleotide sequences selected from the group consisting of: (a) SEQ ID NOs: 9 and 10, respectively; (b) SEQ ID NOs: 37 and 38, respectively; (c) SEQ ID NOs: 65 and 66, respectively; and (d) SEQ ID NOs: 69 and 70, respectively, and wherein a lipid moiety is conjugated to a nucleotide of the sense strand, thereby treating cancer in the subject.
  • the disclosure provides a method of treating a disease, disorder or condition associated with activated STAT3 expression, comprising administering to a subject in need thereof an RNAi oligonucleotide, wherein the subject has received or is receiving a PD-L1 inhibitor, wherein the oligonucleotide comprises a sense strand and an antisense strand, wherein the sense strand comprises the nucleotide sequence of SEQ ID NO: 9 and antisense strand comprises the nucleotide sequence of SEQ ID NO: 10, thereby treating cancer in the subject.
  • the disclosure provides a method of treating a disease, disorder or condition associated with activated STAT3 expression, comprising administering to a subject in need thereof an RNAi oligonucleotide, wherein the subject has received or is receiving a PD-L1 inhibitor, wherein the oligonucleotide comprises a sense strand and an antisense strand, wherein the sense strand comprises the nucleotide sequence of SEQ ID NO: 9 and antisense strand comprises the nucleotide sequence of SEQ ID NO: 10, and wherein a lipid moiety is conjugated to a nucleotide of the sense strand, thereby treating cancer in the subject.
  • the disclosure provides a method of treating a disease, disorder or condition associated with activated STAT3 expression, comprising administering to a subject in need thereof an RNAi oligonucleotide, wherein the subject has received or is receiving a PD-L1 inhibitor, wherein the oligonucleotide comprises a sense strand and an antisense strand, wherein the sense strand comprises the nucleotide sequence of SEQ ID NO: 37 and antisense strand comprises the nucleotide sequence of SEQ ID NO: 38, thereby treating cancer in the subject.
  • the disclosure provides a method of treating a disease, disorder or condition associated with activated STAT3 expression, comprising administering to a subject in need thereof an RNAi oligonucleotide, wherein the subject has received or is receiving a PD-L1 inhibitor, wherein the oligonucleotide comprises a sense strand and an antisense strand, wherein the sense strand comprises the nucleotide sequence of SEQ ID NO: 37 and antisense strand comprises the nucleotide sequence of SEQ ID NO: 38, and wherein a lipid moiety is conjugated to a nucleotide of the sense strand, thereby treating cancer in the subject.
  • the disclosure provides a method of treating a disease, disorder or condition associated with activated STAT3 expression, comprising administering to a subject in need thereof an RNAi oligonucleotide, wherein the subject has received or is receiving a PD-L1 inhibitor, wherein the oligonucleotide comprises a sense strand and an antisense strand, wherein the sense strand comprises the nucleotide sequence of SEQ ID NO: 65 and antisense strand comprises the nucleotide sequence of SEQ ID NO: 66, thereby treating cancer in the subject.
  • the disclosure provides a method of treating a disease, disorder or condition associated with activated STAT3 expression, comprising administering to a subject in need thereof an RNAi oligonucleotide, wherein the subject has received or is receiving a PD-L1 inhibitor, wherein the oligonucleotide comprises a sense strand and an antisense strand, wherein the sense strand comprises the nucleotide sequence of SEQ ID NO: 65 and antisense strand comprises the nucleotide sequence of SEQ ID NO: 66, and wherein a lipid moiety is conjugated to a nucleotide of the sense strand, thereby treating cancer in the subject.
  • the disclosure provides a method of treating a disease, disorder or condition associated with activated STAT3 expression, comprising administering to a subject in need thereof an RNAi oligonucleotide, wherein the subject has received or is receiving a PD-L1 inhibitor, wherein the oligonucleotide comprises a sense strand and an antisense strand, wherein the sense strand comprises the nucleotide sequence of SEQ ID NO: 69 and antisense strand comprises the nucleotide sequence of SEQ ID NO: 70, thereby treating cancer in the subject.
  • the disclosure provides a method of treating a disease, disorder or condition associated with activated STAT3 expression, comprising administering to a subject in need thereof an RNAi oligonucleotide, wherein the subject has received or is receiving a PD-L1 inhibitor, wherein the oligonucleotide comprises a sense strand and an antisense strand, wherein the sense strand comprises the nucleotide sequence of SEQ ID NO: 69 and antisense strand comprises the nucleotide sequence of SEQ ID NO: 70, and wherein a lipid moiety is conjugated to a nucleotide of the sense strand, thereby treating cancer in the subject.
  • the methods of the disclosure increase expression of immune activation genes.
  • administering an RNAi oligonucleotide described herein with a PD-L1 inhibitor increases expression of immune activation genes.
  • the methods described herein increase expression of one or more of CD8B1, IL2R ⁇ , TNFRSF9, CCL19, CCL27a, CD40Ig, CXCR5, CD44, STAT4, GZMB, PFR1, FASL, and HMGB1.
  • the methods of the disclosures decrease expression of immuno-suppressive genes.
  • administering an RNAi oligonucleotide described herein with a PD-L1 inhibitor decreases expression of immune-suppressive genes.
  • the methods described herein decrease expression of one or more of CD274, LAG3, TIGIT, PDSD1IG2, CD163, PIK3R5, FOXP3, TGFBR2, IDO1, CD200R1, CCL22, CCL8, CCR9, IL10, IL33, CXCL3, CSF2RD, TNFRSF18, MMP9, LOX12, FGF18, and FAP.
  • the STAT3 oligonucleotide and PD-L1 inhibitor target are used to treat a cancer or a tumor.
  • the tumor is a primary tumor.
  • the tumor is a metastatic tumor.
  • the tumor is a refractory tumor.
  • the tumor is a Stage I, Stage II, Stage III, or Stage IV tumor.
  • the tumor is a solid-tumor.
  • Solid-tumors refer to conditions where the cancer forms a mass [00160]
  • the cancer is a thyroid cancer, papillary thyroid carcinoma, head and neck cancer, liver cancer, colorectal cancer, pancreatic cancer, breast cancer, ovarian cancer, lung cancer, carcinoma, blastoma, medulloblastoma, retinoblastoma, sarcoma, liposarcoma, synovial cell sarcoma, neuroendocrine tumors, carcinoid tumors, gastrinoma, islet cell cancer, mesothelioma, schwannoma, acoustic neuroma, meningioma, adenocarcinoma, lymphoid malignancies, squamous cell cancer, epithelial squamous cell cancer, small-cell lung cancer (SCLC), non-small cell lung cancer (NSCLC), adenocarcinoma of the lung, squamous carcinoma of the lung, cancer of the
  • the cancer is refractory to anti-PD1, anti-PDL1 and/or anti-CTLA4 therapy.
  • the cancer is a pancreatic cancer or lung cancer.
  • the cancer comprises tumors with immunosuppressive tumor microenvironments.
  • the cancer is resistant to immune checkpoint therapy.
  • the cancer is partially resistant to immune checkpoint therapy.
  • the cancer is sensitive to immune checkpoint therapy.
  • the STAT3 oligonucleotide and PD-L1 inhibitor reduces tumor volume. Tumor volume is measured using methods know to one of skill in the art. For example, extracted tumors are measured manually using calipers. Other methods include imagine methods such as ultrasound and MRI.
  • the oligonucleotide conjugate reduces tumor volume by at least 5%, at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, or at least 90% compared to an untreated tumor.
  • Oligonucleotide Inhibitors of STAT3 [00162]
  • the disclosure provides, inter alia, oligonucleotides that reduce or inhibit STAT3 expression.
  • an oligonucleotide that inhibits STAT3 expression herein is targeted to a STAT3 mRNA.
  • STAT3 is a known target for conventional cancer therapies.
  • STAT3 is a known target for conventional cancer therapies.
  • the tolerogenic activities of MDSCs are controlled by an oncogenic transcription factor, signal transducer and activator of transcription 3 (STAT3) (Su et al., INT J. MOL SCI (2016) 19(6): 1803).
  • STAT3 is also known to be highly expressed across a range of cancer types and in in vitro and in vivo preclinical models (Huynh et al., NAT. REV. CANCER (2019) 19: 82-96).
  • STAT3 leads to the selective apoptosis of tumor cells and tumor growth inhibition through modulation of downstream target genes (Wang et al., INTERNATIONAL JOURNAL OF BIOLOGICAL SCIENCES, 15(3): 668–79 (2019)).
  • STAT3 is of particular interest in immuno-oncology due to its well documented contributions to an immunosuppressive tumor microenvironment.
  • STAT3 contributes to an immunosuppressive tumor microenvironment by upregulating the inhibitory receptor expressed by T-cells, and via expression of its ligand (PD-1/PD-L1), through increased secretion of IFN ⁇ (Bu et al., JOURNAL OF DENTAL RESEARCH, 96(9): 1027–34 (2017)).
  • reduction of STAT3 expression can be determined by an appropriate assay or technique to evaluate one or more properties or characteristics of a cell or population of cells associated with STAT3 expression (e.g., using an STAT3 expression biomarker) or by an assay or technique that evaluates molecules that are directly indicative of STAT3 expression (e.g., STAT3 mRNA or STAT3 protein).
  • an appropriate assay or technique to evaluate one or more properties or characteristics of a cell or population of cells associated with STAT3 expression (e.g., using an STAT3 expression biomarker) or by an assay or technique that evaluates molecules that are directly indicative of STAT3 expression (e.g., STAT3 mRNA or STAT3 protein).
  • an oligonucleotide herein reduces STAT3 expression is evaluated by comparing STAT3 expression in a cell or population of cells contacted with the oligonucleotide to an appropriate control (e.g., an appropriate cell or population of cells not contacted with the oligonucleotide or contacted with a control oligonucleotide).
  • an appropriate control level of mRNA expression into protein, after delivery of a RNAi molecule may be a predetermined level or value, such that a control level need not be measured every time.
  • the predetermined level or value can take a variety of forms.
  • a predetermined level or value can be single cut-off value, such as a median or mean.
  • administration of an oligonucleotide herein results in a reduction in STAT3 expression in a cell or population of cells.
  • the reduction in STAT3 or STAT3 expression is about 1% or lower, about 5% or lower, about 10% or lower, about 15% or lower, about 20% or lower, about 25% or lower, about 30% or lower, about 35% or lower, about 40% or lower, about 45% or lower, about 50% or lower, about 55% or lower, about 60% or lower, about 70% or lower, about 80% or lower, or about 90% or lower when compared with an appropriate control level of mRNA.
  • the appropriate control level may be a level of mRNA expression and/or protein translation in a cell or population of cells that has not been contacted with an oligonucleotide herein.
  • the effect of delivery of an oligonucleotide to a cell according to a method herein is assessed after a finite period.
  • levels of mRNA may be analyzed in a cell at least about 8 hours, about 12 hours, about 18 hours, about 24 hours; or at least about 1, 2, 3, 4, 5, 6, 7 or even up to 14 days after introduction of the oligonucleotide into the cell.
  • an oligonucleotide is delivered in the form of a transgene that is engineered to express in a cell the oligonucleotide or strands comprising the oligonucleotide (e.g., its sense and antisense strands).
  • an oligonucleotide is delivered using a transgene engineered to express any oligonucleotide disclosed herein.
  • Transgenes may be delivered using viral vectors (e.g., adenovirus, retrovirus, vaccinia virus, poxvirus, adeno-associated virus, or herpes simplex virus) or non- viral vectors (e.g., plasmids or synthetic mRNAs).
  • viral vectors e.g., adenovirus, retrovirus, vaccinia virus, poxvirus, adeno-associated virus, or herpes simplex virus
  • non- viral vectors e.g., plasmids or synthetic mRNAs.
  • transgenes can be injected directly to a subject.
  • STAT3 Target Sequences [00167]
  • the oligonucleotide is targeted to a target sequence comprising a STAT3 mRNA.
  • the oligonucleotide, or a portion, fragment, or strand thereof binds or anneals to a target sequence comprising a STAT3 mRNA, thereby inhibiting STAT3 expression.
  • the oligonucleotide is targeted to a STAT3 target sequence for the purpose of inhibiting STAT3 expression in vivo.
  • the amount or extent of inhibition of STAT3 expression by an oligonucleotide targeted to a STAT3 target sequence correlates with the potency of the oligonucleotide.
  • the amount or extent of inhibition of STAT3 expression by an oligonucleotide targeted to a STAT3 target sequence correlates with the amount or extent of therapeutic benefit in a subject or patient having a disease, disorder or condition associated with the expression of STAT3 treated with the oligonucleotide.
  • a sense strand of an oligonucleotide (e.g., a dsRNA) described herein comprises a STAT3 target sequence.
  • a portion or region of the sense strand of a dsRNA described herein comprises a STAT3 target sequence.
  • a STAT3 mRNA target sequence comprises, or consists of, a sequence of SEQ ID NO 85.
  • a STAT3 mRNA target sequence comprises, or consists of, a sequence of SEQ ID NO: 1217.
  • a STAT3 mRNA target sequence comprises, or consists of, a sequence of any one of SEQ ID NOs: 89-280.
  • a STAT3 mRNA target sequence comprises, or consists of, the sequence set forth in SEQ ID NO: 108. In some embodiments, a STAT3 mRNA target sequence comprises, or consists of, the sequence set forth in SEQ ID NO: 140. In some embodiments, a STAT3 mRNA target sequence comprises, or consists of, the sequence set forth in SEQ ID NO: 141. In some embodiments, a STAT3 mRNA target sequence comprises, or consists of, the sequence set forth in SEQ ID NO: 147.
  • the oligonucleotides herein have regions of complementarity to STAT3 mRNA (e.g., within a target sequence of STAT3 mRNA) for purposes of targeting the mRNA in cells and reducing or inhibiting its expression.
  • the oligonucleotides herein comprise a STAT3 targeting sequence (e.g., an antisense strand or a guide strand of a dsRNA) having a region of complementarity that binds or anneals to a STAT3 target sequence by complementary (Watson-Crick) base pairing.
  • the targeting sequence or region of complementarity is generally of a suitable length and base content to enable binding or annealing of the oligonucleotide (or a strand thereof) to a STAT3 mRNA for purposes of inhibiting its expression.
  • the targeting sequence or region of complementarity is at least about 12, at least about 13, at least about 14, at least about 15, at least about 16, at least about 17, at least about 18, at least about 19, at least about 20, at least about 21, at least about 22, at least about 23, at least about 24, at least about 25, at least about 26, at least about 27, at least about 28, at least about 29 or at least about 30 nucleotides in length.
  • the targeting sequence or region of complementarity is about 12 to about 30 (e.g., 12 to 30, 12 to 22, 15 to 25, 17 to 21, 18 to 27, 19 to 27, or 15 to 30) nucleotides in length. In some embodiments, the targeting sequence or region of complementarity is about 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29 or 30 nucleotides in length. In some embodiments, the targeting sequence or region of complementarity is 18 nucleotides in length. In some embodiments, the targeting sequence or region of complementarity is 19 nucleotides in length. In some embodiments, the targeting sequence or region of complementarity is 20 nucleotides in length.
  • the targeting sequence or region of complementarity is 21 nucleotides in length. In some embodiments, the targeting sequence or region of complementarity is 22 nucleotides in length. In some embodiments, the targeting sequence or region of complementarity is 23 nucleotides in length. In some embodiments, the targeting sequence or region of complementarity is 24 nucleotides in length. In some embodiments, an oligonucleotide comprises a target sequence or region of complementarity complementary to a sequence of any one of SEQ ID NOs: 89-280 , and the targeting sequence or region of complementarity is 18 nucleotides in length.
  • an oligonucleotide comprises a target sequence or region of complementarity complementary to a sequence of any one of SEQ ID NOs: 89-280, and the targeting sequence or region of complementarity is 19 nucleotides in length. In some embodiments, an oligonucleotide comprises a target sequence or region of complementarity complementary to a sequence of any one of SEQ ID NOs: 473-664, and the targeting sequence or region of complementarity is 20 nucleotides in length.
  • an oligonucleotide comprises a targeting sequence or region of complementarity complementary to a sequence of any one of SEQ ID NOs: 473-664, and the targeting sequence or region of complementarity is 21 nucleotides in length. In some embodiments, an oligonucleotide comprises a targeting sequence or region of complementarity complementary to a sequence of any one of SEQ ID NOs: 473-664, and the targeting sequence or region of complementarity is 22 nucleotides in length. In some embodiments, an oligonucleotide comprises a targeting sequence or region of complementarity complementary to a sequence of any one of SEQ ID NOs: 473-664, and the targeting sequence or region of complementarity is 23 nucleotides in length.
  • an oligonucleotide comprises a targeting sequence or region of complementarity complementary to a sequence of any one of SEQ ID NOs: 473-664 and the targeting sequence or region of complementarity is 24 nucleotides in length.
  • an oligonucleotide herein comprises a targeting sequence or a region of complementarity (e.g., an antisense strand or a guide strand of a double-stranded oligonucleotide) that is fully complementary to a STAT3 target sequence.
  • the targeting sequence or region of complementarity is partially complementary to a STAT3 target sequence.
  • the oligonucleotide comprises a targeting sequence or region of complementarity that is fully complementary to a sequence of STAT3 or STAT3. In some embodiments, the oligonucleotide comprises a targeting sequence or region of complementarity that is partially complementary to a sequence of STAT3 or STAT3. [00171] In some embodiments, the oligonucleotide comprises a targeting sequence or region of complementarity that is fully complementary to a sequence of any one of SEQ ID NOs: 89-280. In some embodiments, the oligonucleotide comprises a targeting sequence or region of complementarity that is fully complementary to the sequence set forth in SEQ ID NOs: 108, 140, 141, and 147.
  • the oligonucleotide comprises a targeting sequence or region of complementarity that is partially complementary to a sequence of any one of SEQ ID NOs: 89-280. In some embodiments, the oligonucleotide comprises a targeting sequence or region of complementarity that is partially complementary to the sequence set forth in SEQ ID NOs: 108, 140, 141, and 147.
  • the oligonucleotide herein comprises a targeting sequence or region of complementarity that is complementary to a contiguous sequence of nucleotides comprising a STAT3 mRNA, wherein the contiguous sequence of nucleotides is about 12 to about 30 nucleotides in length (e.g., 12 to 30, 12 to 28, 12 to 26, 12 to 24, 12 to 20, 12 to 18, 12 to 16, 14 to 22, 16 to 20, 18 to 20 or 18 to 19 nucleotides in length).
  • the oligonucleotide comprises a targeting sequence or region of complementarity that is complementary to a contiguous sequence of nucleotides comprising a STAT3 mRNA, wherein the contiguous sequence of nucleotides is 10, 11, 12, 13, 14, 15, 16, 17, 18, 19 or 20 nucleotides in length. In some embodiments, the oligonucleotide comprises a targeting sequence or region of complementarity that is complementary to a contiguous sequence of nucleotides comprising a STAT3 mRNA, wherein the contiguous sequence of nucleotides is 19 nucleotides in length.
  • an oligonucleotide herein (e.g., an RNAi oligonucleotide) comprises a targeting sequence or a region of complementary that is complementary to a contiguous sequence of nucleotides of any one of SEQ ID NOs: 89-280, optionally wherein the contiguous sequence of nucleotides is 19 nucleotides in length.
  • the oligonucleotide comprises a targeting sequence or a region of complementary that is complementary to a contiguous sequence of nucleotides of any one of SEQ ID NOs: 108, 140, 141, and 147, wherein the contiguous sequence of nucleotides is 19 nucleotides in length.
  • the oligonucleotide comprises a targeting sequence or a region of complementary that is complementary to a contiguous sequence of nucleotides of any one of SEQ ID NOs: 473-664, wherein the contiguous sequence of nucleotides is 20 nucleotides in length. In some embodiments, the oligonucleotide comprises a targeting sequence or a region of complementary that is complementary to a contiguous sequence of nucleotides of any one of SEQ ID NOs: 492, 524, 525, and 531, wherein the contiguous sequence of nucleotides is 20 nucleotides in length.
  • a targeting sequence or region of complementarity of an oligonucleotide that is complementary to contiguous nucleotides of STAT3 or STAT3 target sequence spans the entire length of an antisense strand. In some embodiments, a region of complementarity of an oligonucleotide that is complementary to contiguous nucleotides of STAT3 or STAT3 target sequence spans a portion of the entire length of an antisense strand.
  • an oligonucleotide herein comprises a region of complementarity (e.g., on an antisense strand of a dsRNA) that is at least partially (e.g., fully) complementary to a contiguous stretch of nucleotides spanning nucleotides 1-20 of a target sequence of STAT3 or STAT3.
  • a targeting sequence or region of complementarity of an oligonucleotide herein is complementary to a contiguous sequence of nucleotides of any one of SEQ ID NOs: 89-280 and spans the entire length of an antisense strand.
  • a targeting sequence or region of complementarity of the oligonucleotide is complementary to a contiguous sequence of nucleotides of SEQ ID NOs: 89-280 and spans a portion of the entire length of an antisense strand.
  • an oligonucleotide herein (e.g., an RNAi oligonucleotide) comprises a region of complementarity (e.g., on an antisense strand of a dsRNA) that is at least partially (e.g., fully) complementary to a contiguous stretch of nucleotides spanning nucleotides 1-19 or 1- 20 of a sequence as set forth in any one of SEQ ID NOs: 473-664.
  • an oligonucleotide herein comprises a targeting sequence or region of complementarity having one or more bp mismatches with the corresponding STAT3 target sequence.
  • the targeting sequence or region of complementarity may have up to about 1, up to about 2, up to about 3, up to about 4, up to about 5, etc. mismatches with the corresponding STAT3 target sequence provided that the ability of the targeting sequence or region of complementarity to bind or anneal to the STAT3 mRNA under appropriate hybridization conditions and/or the ability of the oligonucleotide to inhibit STAT3 expression is maintained.
  • the targeting sequence or region of complementarity may have no more than 1, no more than 2, no more than 3, no more than 4, or no more than 5 mismatches with the corresponding STAT3 target sequence provided that the ability of the targeting sequence or region of complementarity to bind or anneal to the STAT3 mRNA under appropriate hybridization conditions and/or the ability of the oligonucleotide to inhibit STAT3 expression is maintained.
  • the oligonucleotide comprises a targeting sequence or region of complementarity having 1 mismatch with the corresponding target sequence.
  • the oligonucleotide comprises a targeting sequence or region of complementarity having 2 mismatches with the corresponding target sequence.
  • the oligonucleotide comprises a targeting sequence or region of complementarity having 3 mismatches with the corresponding target sequence. In some embodiments, the oligonucleotide comprises a targeting sequence or region of complementarity having 4 mismatches with the corresponding target sequence. In some embodiments, the oligonucleotide comprises a targeting sequence or region of complementarity having 5 mismatches with the corresponding target sequence.
  • the oligonucleotide comprises a targeting sequence or region of complementarity more than one mismatch (e.g., 2, 3, 4, 5 or more mismatches) with the corresponding target sequence, wherein at least 2 (e.g., all) of the mismatches are positioned consecutively (e.g., 2, 3, 4, 5 or more mismatches in a row), or where in the mismatches are interspersed throughout the targeting sequence or region of complementarity.
  • mismatch e.g., 2, 3, 4, 5 or more mismatches
  • the oligonucleotide comprises a targeting sequence or a region of complementary that is complementary to a contiguous sequence of nucleotides of any one of SEQ ID NOs: 89-280, wherein the targeting sequence or region of complementarity may have up to about 1, up to about 2, up to about 3, up to about 4, up to about 5, etc. mismatches with the corresponding STAT3 target sequence.
  • the oligonucleotide comprises a targeting sequence or a region of complementary that is complementary to a contiguous sequence of nucleotides of any one of SEQ ID NOs: 89-280, wherein the targeting sequence or region of complementarity may have no more than 1, no more than 2, no more than 3, no more than 4, or no more than 5 mismatches with the corresponding STAT3 target sequence.
  • the oligonucleotide comprises a targeting sequence or a region of complementary that is complementary to a contiguous sequence of nucleotides of any one of SEQ ID NOs: 108, 140, 141, and 147, wherein the targeting sequence or region of complementarity may have up to about 1, up to about 2, up to about 3, up to about 4, up to about 5, etc. mismatches with the corresponding STAT3 target sequence.
  • the oligonucleotide comprises a targeting sequence or a region of complementary that is complementary to a contiguous sequence of nucleotides of any one of SEQ ID NOs: 108, 140, 141, and 147, wherein the targeting sequence or region of complementarity may have no more than 1, no more than 2, no more than 3, no more than 4, or no more than 5 mismatches with the corresponding STAT3 target sequence.
  • Types of Oligonucleotides [00177] A variety of oligonucleotide types and/or structures are useful for targeting a target sequence in the methods herein including, but not limited to, RNAi oligonucleotides, antisense oligonucleotides, miRNAs, etc.
  • RNAi RNA interference pathway upstream or downstream of Dicer involvement.
  • RNAi oligonucleotides have been developed with each strand having sizes of about 19-25 nucleotides with at least one 3′ overhang of 1 to 5 nucleotides (see, e.g., US Patent No.8,372,968).
  • RNAi products Longer oligonucleotides also have been developed that are processed by Dicer to generate active RNAi products (see, e.g., US Patent No.8,883,996). Further work produced extended dsRNAs where at least one end of at least one strand is extended beyond a duplex targeting region, including structures where one of the strands includes a thermodynamically-stabilizing tetraloop structure (see, e.g., US Patent Nos.8,513,207 and 8,927,705, as well as Intl. Patent Application Publication No. WO 2010/033225). Such structures may include ss extensions (on one or both sides of the molecule) as well as ds extensions.
  • the oligonucleotides herein engage with the RNAi pathway downstream of the involvement of Dicer (e.g., Dicer cleavage).
  • the oligonucleotides described herein are Dicer substrates.
  • double-stranded nucleic acids of 19-23 nucleotide sin length capable of reducing target mRNA expression are produced.
  • the oligonucleotide has an overhang (e.g., of 1, 2, or 3 nucleotides in length) in the 3′ end of the sense strand.
  • the oligonucleotide (e.g., siRNA) comprises a 21-nucleotide guide strand that is antisense to a target RNA and a complementary passenger strand, in which both strands anneal to form a 19-bp duplex and 2 nucleotide overhangs at either or both 3′ ends.
  • oligonucleotide designs also are available including oligonucleotides having a guide strand of 23 nucleotides and a passenger strand of 21 nucleotides, where there is a blunt end on the right side of the molecule (3′ end of passenger strand/5′ end of guide strand) and a two nucleotide 3′-guide strand overhang on the left side of the molecule (5′ end of the passenger strand/3′ end of the guide strand). In such molecules, there is a 21 bp duplex region. See, e.g., US Patent Nos.9,012,138; 9,012,621 and 9,193,753.
  • the oligonucleotides herein comprise sense and antisense strands that are both in the range of about 17 to 26 (e.g., 17 to 26, 20 to 25 or 21- 23) nucleotides in length. In some embodiments, the oligonucleotides herein comprise sense and antisense strands that are both in the range of about 17 to 36 (e.g., 17 to 36, 20 to 25 or 21-23) nucleotides in length.
  • the oligonucleotides described herein comprise an antisense strand of 19-30 nucleotides in length and a sense strand of 19-50 nucleotides in length, wherein the antisense and sense strands are separate strands which form an asymmetric duplex region having an overhand of 1-4 nucleotides at the 3’ terminus of the antisense strand.
  • an oligonucleotide herein comprises a sense and antisense strand that are both in the range of about 19-22 nucleotides in length.
  • the sense and antisense strands are of equal length.
  • an oligonucleotide comprises sense and antisense strands, such that there is a 3′-overhang on either the sense strand or the antisense strand, or both the sense and antisense strand.
  • a 3′ overhang on the sense, antisense, or both sense and antisense strands is 1 or 2 nucleotides in length.
  • the oligonucleotide has a guide strand of 22 nucleotides and a passenger strand of 20 nucleotides, where there is a blunt end on the right side of the molecule (3′ end of passenger strand/5′ end of guide strand) and a 2 nucleotide 3′-guide strand overhang on the left side of the molecule (5′ end of the passenger strand/3′ end of the guide strand). In such molecules, there is a 20 bp duplex region.
  • Other oligonucleotide designs for use with the compositions and methods herein include: 16-mer siRNAs (see, e.g., NUCLEIC ACIDS IN CHEMISTRY AND BIOLOGY.
  • shRNAs e.g., having 19 bp or shorter stems; (see, e.g., Moore et al., (2010) METHODS MOL. BIOL.629:141-58), blunt siRNAs (e.g., of 19 bps in length; see, e.g., Kraynack and Baker (2006) RNA 12:163-76), asymmetrical siRNAs (aiRNA; see, e.g., Sun et al., (2008) NAT. BIOTECHNOL.26:1379-82), asymmetric shorter-duplex siRNA (see, e.g., Chang et al., (2009) MOL.
  • THER.17:725-32 fork siRNAs (see, e.g., Hohjoh (2004) FEBS LETT.557:193-98), ss siRNAs (Elsner (2012) NAT. BIOTECHNOL.30:1063), dumbbell-shaped circular siRNAs (see, e.g., Abe et al., (2007) J. AM. CHEM. SOC.129:15108-09), and small internally segmented interfering RNA (siRNA; see, e.g., Bramsen et al., (2007) NUCLEIC ACIDS RES.35:5886-97).
  • siRNA small internally segmented interfering RNA
  • an oligonucleotide structures that may be used in some embodiments to reduce or inhibit the expression of STAT3 are microRNA (miRNA), short hairpin RNA (shRNA) and short siRNA (see, e.g., Hamilton et al., (2002) EMBO J.21:4671-79; see also, US Patent Application Publication No.2009/0099115).
  • miRNA microRNA
  • shRNA short hairpin RNA
  • siRNA see, e.g., Hamilton et al., (2002) EMBO J.21:4671-79; see also, US Patent Application Publication No.2009/0099115.
  • ss an oligonucleotide for reducing or inhibiting expression of a target sequence herein.
  • Such structures may include but are not limited to ss RNAi molecules.
  • oligonucleotides herein are antisense oligonucleotides (ASOs).
  • An antisense oligonucleotide is a ss oligonucleotide that has a nucleobase sequence which, when written in the 5′ to 3′ direction, comprises the reverse complement of a targeted segment of a particular nucleic acid and is suitably modified (e.g., as a gapmer) to induce RNaseH-mediated cleavage of its target RNA in cells or (e.g., as a mixmer) to inhibit translation of the target mRNA in cells.
  • ASOs for use herein may be modified in any suitable manner known in the art including, for example, as shown in US Patent No.9,567,587 (including, e.g., length, sugar moieties of the nucleobase (pyrimidine, purine), and alterations of the heterocyclic portion of the nucleobase). Further, ASOs have been used for decades to reduce expression of specific target genes (see, e.g., Bennett et al., (2017) ANNU. REV. PHARMACOL.57:81-105). [00183] In some embodiments, the antisense oligonucleotide shares a region of complementarity with a target mRNA.
  • the antisense oligonucleotide is 15-50 nucleotides in length. In some embodiments, the antisense oligonucleotide is 15-25 nucleotides in length. In some embodiments, the antisense oligonucleotide is 22 nucleotides in length. In some embodiments, the antisense oligonucleotide is at least 15 contiguous nucleotides in length. In some embodiments, the antisense oligonucleotide is at least 19 contiguous nucleotides in length. In some embodiments, the antisense oligonucleotide is at least 20 contiguous nucleotides in length.
  • the antisense oligonucleotide differs by 1, 2, or 3 nucleotides from the target sequence.
  • Double-Stranded Oligonucleotides [00184]
  • the disclosure provides double-stranded dsRNAs for targeting and inhibiting expression of a target sequence (e.g., via the RNAi pathway) comprising a sense strand (also referred to herein as a passenger strand) and an antisense strand (also referred to herein as a guide strand).
  • the sense strand and antisense strand are separate strands and are not covalently linked.
  • the sense strand and antisense strand are covalently linked.
  • the sense strand and antisense strand form a duplex region, wherein the sense strand and antisense strand, or a portion thereof, binds with one another in a complementary fashion (e.g., by Watson-Crick base pairing).
  • the sense strand has a first region (R1) and a second region (R2), wherein R2 comprises a first subregion (S1), a loop (L), such as a tetraloop (tetraL) or triloop (triL), and a second subregion (S2), wherein L, tetraL, or triL is located between S1 and S2, and wherein S1 and S2 form a second duplex (D2).
  • D2 may have various length. In some embodiments, D2 is about 1-6 bp in length. In some embodiments, D2 is 2-6, 3-6, 4-6, 5-6, 1-5, 2-5, 3-5 or 4-5 bp in length. In some embodiments, D2 is 1, 2, 3, 4, 5 or 6 bp in length. In some embodiments, D2 is 6 bp in length. [00186] In some embodiments, R1 of the sense strand and the antisense strand form a first duplex (D1). In some embodiments, D1 is at least about 15 (e.g., at least 15, at least 16, at least 17, at least 18, at least 19, at least 20 or at least 21) nucleotides in length.
  • D1 is at least about 15 (e.g., at least 15, at least 16, at least 17, at least 18, at least 19, at least 20 or at least 21) nucleotides in length.
  • D1 is in the range of about 12 to 30 nucleotides in length (e.g., 12 to 30, 12 to 27, 15 to 22, 18 to 22, 18 to 25, 18 to 27, 18 to 30 or 21 to 30 nucleotides in length). In some embodiments, D1 is at least 12 nucleotides in length (e.g., at least 12, at least 15, at least 20, at least 25, or at least 30 nucleotides in length). In some embodiments, D1 is 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29 or 30 nucleotides in length. In some embodiments, D1 is 20 nucleotides in length.
  • D1 comprising sense strand and antisense strand does not span the entire length of the sense strand and/or antisense strand. In some embodiments, D1 comprising the sense strand and antisense strand spans the entire length of either the sense strand or antisense strand or both. In certain embodiments, D1 comprising the sense strand and antisense strand spans the entire length of both the sense strand and the antisense strand. [00187] It should be appreciated that, in some embodiments, sequences presented in the Sequence Listing may be referred to in describing the structure of an oligonucleotide or other nucleic acid.
  • the actual oligonucleotide or other nucleic acid may have one or more alternative nucleotides (e.g., an RNA counterpart of a DNA nucleotide or a DNA counterpart of an RNA nucleotide) and/or one or more modified nucleotides and/or one or more modified internucleotide linkages and/or one or more other modification when compared with the specified sequence while retaining essentially same or similar complementary properties as the specified sequence.
  • alternative nucleotides e.g., an RNA counterpart of a DNA nucleotide or a DNA counterpart of an RNA nucleotide
  • modified nucleotides and/or one or more modified internucleotide linkages and/or one or more other modification when compared with the specified sequence while retaining essentially same or similar complementary properties as the specified sequence.
  • a double-stranded RNA herein comprises a 25-nucleotide sense strand and a 27-nucleotide antisense strand that when acted upon by a Dicer enzyme results in an antisense strand that is incorporated into the mature RISC.
  • the sense strand of the dsRNA is longer than 27 nucleotides (e.g., 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39 or 40 nucleotides).
  • the sense strand of the dsRNA is longer than 27 nucleotides (e.g., 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, or 50 nucleotides). In some embodiments, the sense strand of the dsRNA is longer than 25 nucleotides (e.g., 26, 27, 28, 29 or 30 nucleotides). [00189] In some embodiments, oligonucleotides herein have one 5′ end that is thermodynamically less stable when compared to the other 5′ end.
  • an asymmetry oligonucleotide that includes a blunt end at the 3′ end of a sense strand and a 3′-overhang at the 3′ end of an antisense strand.
  • the 3′- overhang on the antisense strand is about 1-8 nucleotides in length (e.g., 1, 2, 3, 4, 5, 6, 7 or 8 nucleotides in length).
  • an oligonucleotide for RNAi has a two-nucleotide overhang on the 3′ end of the antisense (guide) strand. However, other overhangs are possible.
  • an overhang is a 3′-overhang comprising a length of between 1 and 6 nucleotides, optionally 1 to 5, 1 to 4, 1 to 3, 1 to 2, 2 to 6, 2 to 5, 2 to 4, 2 to 3, 3 to 6, 3 to 5, 3 to 4, 4 to 6, 4 to 5, 5 to 6 nucleotides, or 1, 2, 3, 4, 5 or 6 nucleotides.
  • the overhang is a 5′-overhang comprising a length of between 1 and 6 nucleotides, optionally 1 to 5, 1 to 4, 1 to 3, 1 to 2, 2 to 6, 2 to 5, 2 to 4, 2 to 3, 3 to 6, 3 to 5, 3 to 4, 4 to 6, 4 to 5, 5 to 6 nucleotides, or 1, 2, 3, 4, 5 or 6 nucleotides.
  • two terminal nucleotides on the 3′ end of an antisense strand are modified.
  • the two terminal nucleotides on the 3′ end of the antisense strand are complementary with the target mRNA.
  • the two terminal nucleotides on the 3′ end of the antisense strand are not complementary with the target mRNA.
  • the two terminal nucleotides on the 3′ end of the antisense strand of an oligonucleotide herein comprise an unpaired GG.
  • the two (2) terminal nucleotides on the 3′ end of an antisense strand of an oligonucleotide herein are not complementary to the target mRNA.
  • two terminal nucleotides on each 3′ end of an oligonucleotide in the nicked tetraloop structure are GG.
  • one or both of the two (2) terminal GG nucleotides on each 3′ end of an oligonucleotide herein is not complementary with the target mRNA.
  • one or both two terminal GG nucleotides on each 3′ end of an oligonucleotide is not complementary with the target.
  • there is one or more (e.g., 1, 2, 3, 4 or 5) mismatch between a sense and antisense strand If there is more than one mismatch between a sense and antisense strand, they may be positioned consecutively (e.g., 2, 3 or more in a row), or interspersed throughout the region of complementarity.
  • the 3′ end of the sense strand contains one or more mismatches. In one embodiment, two mismatches are incorporated at the 3′ end of the sense strand. In some embodiments, base mismatches, or destabilization of segments at the 3′ end of the sense strand of the oligonucleotide improved the potency of synthetic duplexes in RNAi, possibly through facilitating processing by Dicer. a.
  • a dsRNA comprises an antisense strand of up to about 40 nucleotides in length (e.g., up to 40, up to 35, up to 30, up to 27, up to 25, up to 21, up to 19, up to 17 or up to 12 nucleotides in length).
  • an oligonucleotide herein e.g., an RNAi oligonucleotide
  • comprises an antisense strand of up to about 50 nucleotides in length e.g., up to 50, up to 40, up to 35, up to 30, up to 27, up to 25, up to 21, up to 19, up to 17 or up to 12 nucleotides in length).
  • an oligonucleotide may have an antisense strand of at least about 12 nucleotides in length (e.g., at least 12, at least 15, at least 19, at least 21, at least 22, at least 25, at least 27, at least 30, at least 35 or at least 38 nucleotides in length).
  • an oligonucleotide may have an antisense strand in a range of about 12 to about 40 (e.g., 12 to 40, 12 to 36, 12 to 32, 12 to 28, 15 to 40, 15 to 36, 15 to 32, 15 to 28, 17 to 22, 17 to 25, 19 to 27, 19 to 30, 20 to 40, 22 to 40, 25 to 40 or 32 to 40) nucleotides in length.
  • an oligonucleotide comprises antisense strand of 15 to 30 nucleotides in length. In some embodiments, an oligonucleotide may have an antisense strand of 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 in length.
  • an antisense strand of an oligonucleotide may be referred to as a “guide strand.”
  • a guide strand For example, if an antisense strand can engage with RNA- induced silencing complex (RISC) and bind to an Argonaute protein such as Ago2, or engage with or bind to one or more similar factors, and direct silencing of a target gene, it may be referred to as a guide strand.
  • RISC RNA- induced silencing complex
  • an oligonucleotide disclosed herein for targeting STAT3 comprises an antisense strand comprising or consisting of a sequence as set forth in any one of SEQ ID NOs: 281-472.
  • an oligonucleotide herein comprises an antisense strand comprising at least about 12 (e.g., at least 12, at least 13, at least 14, at least 15, at least 16, at least 17, at least 18, at least 19, at least 20, at least 21, at least 22 or at least 23) contiguous nucleotides of a sequence as set forth in any one of SEQ ID NOs: 281-472.
  • an oligonucleotide disclosed herein e.g., an RNAi oligonucleotide for targeting STAT3 comprises an antisense strand comprising or consisting of a sequence as set forth in any one of SEQ ID NOs: 665-856.
  • an oligonucleotide herein comprises an antisense strand comprising at least about 12 (e.g., at least 12, at least 13, at least 14, at least 15, at least 16, at least 17, at least 18, at least 19, at least 20, at least 21, at least 22 or at least 23) contiguous nucleotides of a sequence as set forth in any one of SEQ ID NOs: 665-856.
  • an oligonucleotide disclosed herein for targeting STAT3 comprises an antisense strand comprising or consisting of a sequence as set forth in any one of SEQ ID NOs: 947-1036.
  • an oligonucleotide herein comprises an antisense strand comprising at least about 12 (e.g., at least 12, at least 13, at least 14, at least 15, at least 16, at least 17, at least 18, at least 19, at least 20, at least 21, at least 22 or at least 23) contiguous nucleotides of a sequence as set forth in any one of SEQ ID NOs: 947-1036.
  • an oligonucleotide disclosed herein for targeting STAT3 comprises an antisense strand comprising or consisting of a sequence as set forth in any one of SEQ ID NOs: 10, 38, 66, and 70.
  • an oligonucleotide herein comprises an antisense strand comprising at least about 12 (e.g., at least 12, at least 13, at least 14, at least 15, at least 16, at least 17, at least 18, at least 19, at least 20, at least 21, at least 22 or at least 23) contiguous nucleotides of a sequence as set forth in any one of SEQ ID NOs: 10, 38, 66, and 70.
  • an oligonucleotide disclosed herein for targeting STAT3 comprises an antisense strand comprising or consisting of a sequence as set forth in any one of SEQ ID NOs: 281-472.
  • an oligonucleotide herein comprises an antisense strand comprising at least about 12 (e.g., at least 12, at least 13, at least 14, at least 15, at least 16, at least 17, at least 18, at least 19, at least 20, at least 21, at least 22 or at least 23) contiguous nucleotides of a sequence as set forth in any one of SEQ ID NOs: 281-472.
  • an oligonucleotide disclosed herein for targeting STAT3 comprises an antisense strand comprising or consisting of a sequence as set forth in any one of SEQ ID NOs: 952, 965, 966, and 1010.
  • an oligonucleotide herein comprises an antisense strand comprising at least about 12 (e.g., at least 12, at least 13, at least 14, at least 15, at least 16, at least 17, at least 18, at least 19, at least 20, at least 21, at least 22 or at least 23) contiguous nucleotides of a sequence as set forth in any one of SEQ ID NOs: 952, 965, 966, and 1010.
  • an oligonucleotide disclosed herein for targeting STAT3 comprises an antisense strand comprising or consisting of a sequence as set forth in any one of SEQ ID NOs: 991, 1000, 989, 986, 982, 980, and 979.
  • an oligonucleotide herein comprises an antisense strand comprising at least about 12 (e.g., at least 12, at least 13, at least 14, at least 15, at least 16, at least 17, at least 18, at least 19, at least 20, at least 21, at least 22 or at least 23) contiguous nucleotides of a sequence as set forth in any one of SEQ ID NOs: 991, 1000, 989, 986, 982, 980, and 979.
  • an oligonucleotide disclosed herein for targeting STAT3 comprises an antisense strand comprising or consisting of a sequence as set forth in any one of SEQ ID NOs: 1030, 1027, and 1029.
  • an oligonucleotide herein comprises an antisense strand comprising at least about 12 (e.g., at least 12, at least 13, at least 14, at least 15, at least 16, at least 17, at least 18, at least 19, at least 20, at least 21, at least 22 or at least 23) contiguous nucleotides of a sequence as set forth in any one of SEQ ID NOs: 1030, 1027, and 1029.
  • an oligonucleotide disclosed herein for targeting STAT3 comprises an antisense strand comprising or consisting of a sequence as set forth in any one of SEQ ID NOs: 1005, 1014, 1003, and 1010.
  • an oligonucleotide herein comprises an antisense strand comprising at least about 12 (e.g., at least 12, at least 13, at least 14, at least 15, at least 16, at least 17, at least 18, at least 19, at least 20, at least 21, at least 22 or at least 23) contiguous nucleotides of a sequence as set forth in any one of SEQ ID NOs: 1005, 1014, 1003, and 1010. [00195] b.
  • an oligonucleotide disclosed herein for targeting STAT3 mRNA and inhibiting STAT3 expression comprises a sense strand sequence as set forth in in any one of SEQ ID NOs: 89-280.
  • an oligonucleotide herein has a sense strand that comprise at least about 12 (e.g., at least 13, at least 14, at least 15, at least 16, at least 17, at least 18, at least 19, at least 20, at least 21, at least 22 or at least 23) contiguous nucleotides of a sequence as set forth in in any one of SEQ ID NOs: 89-280.
  • an oligonucleotide disclosed herein for targeting STAT3 mRNA and inhibiting STAT3 expression comprises a sense strand sequence a set forth in any one of SEQ ID NOs: 473- 664.
  • an oligonucleotide herein has a sense strand that comprise at least about 12 (e.g., at least 13, at least 14, at least 15, at least 16, at least 17, at least 18, at least 19, at least 20, at least 21, at least 22 or at least 23) contiguous nucleotides of a sequence as set forth in in any one of SEQ ID NOs: 473-664.
  • an oligonucleotide disclosed herein for targeting STAT3 mRNA and inhibiting STAT3 expression comprises a sense strand sequence as set forth in any one of SEQ ID NOs: 857-946.
  • an oligonucleotide herein has a sense strand comprised of least about 12 (e.g., at least 13, at least 14, at least 15, at least 16, at least 17, at least 18, at least 19, at least 20, at least 21, at least 22 or at least 23) contiguous nucleotides of a sequence as set forth in any one of SEQ ID NOs: 857-946.
  • an oligonucleotide disclosed herein for targeting STAT3 mRNA and inhibiting STAT3 expression comprises a sense strand sequence as set forth in any one of SEQ ID NOs: 9, 37, 65, and 69.
  • an oligonucleotide herein has a sense strand comprised of least about 12 (e.g., at least 13, at least 14, at least 15, at least 16, at least 17, at least 18, at least 19, at least 20, at least 21, at least 22 or at least 23) contiguous nucleotides of a sequence as set forth in any one of SEQ ID NOs: 9, 37, 65, and 69.
  • an oligonucleotide disclosed herein for targeting STAT3 mRNA and inhibiting STAT3 expression comprises a sense strand sequence as set forth in any one of SEQ ID NOs: 862, 875, 876, and 920.
  • an oligonucleotide herein has a sense strand that comprise at least about 12 (e.g., at least 13, at least 14, at least 15, at least 16, at least 17, at least 18, at least 19, at least 20, at least 21, at least 22 or at least 23) contiguous nucleotides of a sequence as set forth in any one of SEQ ID NOs: 862, 875, 876, and 920.
  • an oligonucleotide disclosed herein for targeting STAT3 mRNA and inhibiting STAT3 expression comprises a sense strand sequence as set forth in any one of SEQ ID NOs: 901, 910, 899, 896, 892, 890, and 889.
  • an oligonucleotide herein has a sense strand that comprise at least about 12 (e.g., at least 13, at least 14, at least 15, at least 16, at least 17, at least 18, at least 19, at least 20, at least 21, at least 22 or at least 23) contiguous nucleotides of a sequence as set forth in any one of SEQ ID NOs: 901, 910, 899, 896, 892, 890, and 889.
  • an oligonucleotide disclosed herein for targeting STAT3 comprises an antisense strand comprising or consisting of a sequence as set forth in any one of SEQ ID NOs: 940, 937, and 939.
  • an oligonucleotide herein comprises an antisense strand comprising at least about 12 (e.g., at least 12, at least 13, at least 14, at least 15, at least 16, at least 17, at least 18, at least 19, at least 20, at least 21, at least 22 or at least 23) contiguous nucleotides of a sequence as set forth in any one of SEQ ID NOs: 940, 937, and 939.
  • an oligonucleotide disclosed herein for targeting STAT3 comprises an antisense strand comprising or consisting of a sequence as set forth in any one of SEQ ID NOs: 915, 924, 913, and 920.
  • an oligonucleotide herein comprises an antisense strand comprising at least about 12 (e.g., at least 12, at least 13, at least 14, at least 15, at least 16, at least 17, at least 18, at least 19, at least 20, at least 21, at least 22 or at least 23) contiguous nucleotides of a sequence as set forth in any one of SEQ ID NOs: 915, 924, 913, and 920.
  • an oligonucleotide comprises a sense strand (or passenger strand) of up to about 40 nucleotides in length (e.g., up to 40, up to 36, up to 30, up to 27, up to 25, up to 21, up to 19, up to 17 or up to 12 nucleotides in length).
  • an oligonucleotide may have a sense strand of at least about 12 nucleotides in length (e.g., at least 12, at least 15, at least 19, at least 21, at least 25, at least 27, at least 30, at least 36 or at least 38 nucleotides in length).
  • an oligonucleotide may have a sense strand in a range of about 12 to about 40 (e.g., 12 to 40, 12 to 36, 12 to 32, 12 to 28, 15 to 40, 15 to 36, 15 to 32, 15 to 28, 17 to 21, 17 to 25, 19 to 27, 19 to 30, 20 to 40, 22 to 40, 25 to 40 or 32 to 40) nucleotides in length.
  • an oligonucleotide herein comprises a sense strand of 15 to 50 nucleotides in length.
  • an oligonucleotide herein comprises a sense strand of 18 to 36 nucleotides in length.
  • an oligonucleotide may have a sense strand of 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 in length.
  • an oligonucleotide comprises a sense strand of 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, 40,41, 42, 43, 44, 45, 46, 47, 48, 49, or 50 nucleotides in length.
  • an oligonucleotide herein comprises a sense strand of 36 nucleotides in length.
  • an oligonucleotide provided herein comprises a sense strand comprising a stem-loop structure at the 3′ end of the sense strand.
  • the stem-loop is formed by intrastrand base pairing.
  • a sense strand comprises a stem-loop structure at its 5′ end.
  • the stem of the stem-loop comprises a duplex of 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13 or 14 nucleotides in length.
  • the stem of the stem-loop comprises a duplex of 2 nucleotides in length.
  • the stem of the stem-loop comprises a duplex of 3 nucleotides in length. In some embodiments, the stem of the stem-loop comprises a duplex of 4 nucleotides in length. In some embodiments, the stem of the stem- loop comprises a duplex of 5 nucleotides in length. In some embodiments, the stem of the stem-loop comprises a duplex of 6 nucleotides in length. In some embodiments, the stem of the stem-loop comprises a duplex of 7 nucleotides in length. In some embodiments, the stem of the stem-loop comprises a duplex of 8 nucleotides in length.
  • the stem of the stem-loop comprises a duplex of 9 nucleotides in length. In some embodiments, the stem of the stem-loop comprises a duplex of 10 nucleotides in length. In some embodiments, the stem of the stem-loop comprises a duplex of 11 nucleotides in length. In some embodiments, the stem of the stem-loop comprises a duplex of 12 nucleotides in length. In some embodiments, the stem of the stem-loop comprises a duplex of 13 nucleotides in length. In some embodiments, the stem of the stem-loop comprises a duplex of 14 nucleotides in length.
  • a stem-loop provides the oligonucleotide protection against degradation (e.g., enzymatic degradation), facilitates or improves targeting and/or delivery to a target cell, tissue, or organ (e.g., the liver), or both.
  • the loop of a stem-loop is comprised of nucleotides comprising one or more modifications that facilitate, improve, or increase targeting to a target, inhibition of target gene expression, and/or delivery, uptake, and/or penetrance into a target cell, tissue, or organ (e.g., the liver), or a combination thereof.
  • the stem-loop itself or modification(s) to the stem-loop do not affect or do not substantially affect the inherent gene expression inhibition activity of the oligonucleotide, but facilitates, improves, or increases stability (e.g., provides protection against degradation) and/or delivery, uptake, and/or penetrance of the oligonucleotide to a target cell, tissue, or organ.
  • an oligonucleotide herein comprises a sense strand comprising (e.g., at its 3′ end) a stem-loop set forth as: S1-L-S2, in which S1 is complementary to S2, and in which L forms a single- stranded loop of linked nucleotides between S1 and S2 of up to about 10 nucleotides in length (e.g., 3, 4, 5, 6, 7, 8, 9 or 10 nucleotides in length).
  • the loop (L) is 3 nucleotides in length (referred to herein as “triloop”.
  • the loop (L) is 4 nucleotides in length (referred to herein as “tetraloop”).
  • the loop (L) is 5 nucleotides in length. In some embodiments, the loop (L) is 6 nucleotides in length. In some embodiments, the loop (L) is 7 nucleotides in length. In some embodiments, the loop (L) is 8 nucleotides in length. In some embodiments, the loop (L) is 9 nucleotides in length. In some embodiments, the loop (L) is 10 nucleotides in length.
  • an oligonucleotide provided herein comprises a targeting sequence or a region of complementary that is complementary to a contiguous sequence of nucleotides of any one of SEQ ID NOs: 89-280, and the oligonucleotide comprises a sense strand comprising (e.g., at its 3′ end) a stem-loop set forth as: S1-L-S2, in which S1 is complementary to S2, and in which L forms a single- stranded loop between S1 and S2 of up to about 10 nucleotides in length (e.g., 3, 4, 5, 6, 7, 8, 9 or 10 nucleotides in length).
  • the oligonucleotide comprises a targeting sequence or a region of complementary that is complementary to a contiguous sequence of nucleotides of any one of SEQ ID NOs: 89-280, and the oligonucleotide comprises a sense strand comprising (e.g., at its 3′ end) a stem-loop set forth as: S1-L-S2, in which S1 is complementary to S2, and in which L forms a single-stranded loop between S1 and S2 of 4 nucleotides in length.
  • the tetraloop comprises the sequence 5’-GAAA-3’.
  • the stem loop comprises the sequence 5’-GCAGCCGAAAGGCUGC-3’ (SEQ ID NO: 86).
  • a sense strand comprises a stem-loop structure at its 3′ end.
  • a sense strand comprises a stem-loop structure at its 5′ end.
  • a stem is a duplex of 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13 or 14 bp in length.
  • a stem-loop provides the molecule protection against degradation (e.g., enzymatic degradation) and facilitates targeting characteristics for delivery to a target cell.
  • a loop provides added nucleotides on which modification can be made without substantially affecting the gene expression inhibition activity of an oligonucleotide.
  • an oligonucleotide is herein in which the sense strand comprises (e.g., at its 3′ end) a stem-loop set forth as: S1-L- S2, in which S1 is complementary to S2, and in which L forms a loop between S1 and S2 of up to about 10 nucleotides in length (e.g., 3, 4, 5, 6, 7, 8, 9 or 10 nucleotides in length).
  • FIG. 1A depicts non-limiting examples of such an oligonucleotide.
  • a loop (L) of a stem-loop having the structure S1-L-S2 as described herein is a triloop.
  • the triloop comprises ribonucleotides, deoxyribonucleotides, modified nucleotides, ligands (e.g., delivery ligands), and combinations thereof.
  • a loop of a stem-loop is a tetraloop (e.g., within a nicked tetraloop structure).
  • a tetraloop may contain ribonucleotides, deoxyribonucleotides, modified nucleotides and combinations thereof.
  • a tetraloop has 4 to 5 nucleotides.
  • Duplex Length [00205] In some embodiments, a duplex formed between a sense and antisense strand is at least 12 (e.g., at least 15, at least 16, at least 17, at least 18, at least 19, at least 20, or at least 21) nucleotides in length. In some embodiments, a duplex formed between a sense and antisense strand is in the range of 12-30 nucleotides in length (e.g., 12 to 30, 12 to 27, 12 to 22, 15 to 25, 18 to 30, 18 to 22, 18 to 25, 18 to 27, 18 to 30, 19 to 30 or 21 to 30 nucleotides in length).
  • a duplex formed between a sense and antisense strand is 12, 13, 14, 15, 16, 17, 18, 19, 29, 21, 22, 23, 24, 25, 26, 27, 28, 29 or 30 nucleotides in length. In some embodiments, a duplex formed between a sense and antisense strand is 12 nucleotides in length. In some embodiments, a duplex formed between a sense and antisense strand is 13 nucleotides in length. In some embodiments, a duplex formed between a sense and antisense strand is 14 nucleotides in length. In some embodiments, a duplex formed between a sense and antisense strand is 15 nucleotides in length.
  • a duplex formed between a sense and antisense strand is 16 nucleotides in length. In some embodiments, a duplex formed between a sense and antisense strand is 17 nucleotides in length. In some embodiments, a duplex formed between a sense and antisense strand is 18 nucleotides in length. In some embodiments, a duplex formed between a sense and antisense strand is 19 nucleotides in length. In some embodiments, a duplex formed between a sense and antisense strand is 20 nucleotides in length. In some embodiments, a duplex formed between a sense and antisense strand is 21 nucleotides in length.
  • a duplex formed between a sense and antisense strand is 22 nucleotides in length. In some embodiments, a duplex formed between a sense and antisense strand is 23 nucleotides in length. In some embodiments, a duplex formed between a sense and antisense strand is 24 nucleotides in length. In some embodiments, a duplex formed between a sense and antisense strand is 25 nucleotides in length. In some embodiments, a duplex formed between a sense and antisense strand is 26 nucleotides in length. In some embodiments, a duplex formed between a sense and antisense strand is 27 nucleotides in length.
  • a duplex formed between a sense and antisense strand is 28 nucleotides in length. In some embodiments, a duplex formed between a sense and antisense strand is 29 nucleotides in length. In some embodiments, a duplex formed between a sense and antisense strand is 30 nucleotides in length. In some embodiments, a duplex formed between a sense and antisense strand does not span the entire length of the sense strand and/or antisense strand. In some embodiments, a duplex between a sense and antisense strand spans the entire length of either the sense or antisense strands.
  • a duplex between a sense and antisense strand spans the entire length of both the sense strand and the antisense strand. [00206] In some embodiments, a duplex between a sense and antisense strand spans the entire length of both the sense strand and the antisense strand.
  • the sense and antisense strands of an oligonucleotide comprise nucleotides sequences selected from the group consisting of: (a) SEQ ID NOs: 861 and 951, respectively; (b) SEQ ID NOs: 857 and 947, respectively; (c) SEQ ID NOs: 858 and 948, respectively; (d) SEQ ID NOs: 859 and 949, respectively; (e) SEQ ID NOs: 860 and 950, respectively; (f) SEQ ID NOs: 862 and 952, respectively; (g) SEQ ID NOs: 863 and 953, respectively; (h) SEQ ID NOs: 864 and 954, respectively; (i) SEQ ID NOs: 865 and 955, respectively; (j) SEQ ID NOs: 866 and 956, respectively; (k) SEQ ID NOs: 867 and 957, respectively; (l) SEQ ID NOs: 868 and 958, respectively; (m) SEQ ID NOs: 869 and 959, respectively;
  • a duplex between a sense and antisense strand spans the entire length of both the sense strand and the antisense strand.
  • the sense and antisense strands of an oligonucleotide comprise nucleotides sequences selected from the group consisting of: (a) SEQ ID NOs: 857 and 947, respectively; (b) SEQ ID NOs: 858 and 948, respectively; (c) SEQ ID NOs: 859 and 949, respectively; (d) SEQ ID NOs: 860 and 950, respectively; (e) SEQ ID NOs: 862 and 952, respectively; (f) SEQ ID NOs: 867 and 957, respectively; (g) SEQ ID NOs: 875 and 965, respectively; and (h) SEQ ID NOs: 876 and 966, respectively, wherein a duplex formed between a sense and antisense strand is in the range of 12-30 nucleotides in length (e.g.,
  • a duplex between a sense and antisense strand spans the entire length of both the sense strand and the antisense strand.
  • the sense and antisense strands of an oligonucleotide comprise nucleotides sequences selected from the group consisting of: (a) SEQ ID NOs: 9 and 10, respectively; (b) SEQ ID NOs: 37 and 38, respectively; (c) SEQ ID NOs: 65 and 66, respectively; and (d) SEQ ID NOs: 69 and 70, respectively.
  • a duplex formed between a sense and antisense strand is in the range of 12-30 nucleotides in length (e.g., 12 to 30, 12 to 27, 12 to 22, 15 to 25, 18 to 30, 18 to 22, 18 to 25, 18 to 27, 18 to 30, 19 to 30 or 21 to 30 nucleotides in length).
  • a duplex between a sense and antisense strand spans the entire length of both the sense strand and the antisense strand.
  • the sense and antisense strands of an oligonucleotide comprise nucleotides sequences selected from the group consisting of: (a) SEQ ID NOs: 901 and 991, respectively; (b) SEQ ID NOs: 910 and 1000, respectively; (c) SEQ ID NOs: 899 and 989, respectively; (d) SEQ ID NOs: 896 and 986, respectively; (e) SEQ ID NOs: 892 and 982, respectively; (f) SEQ ID NOs: 890 and 980, respectively; and (g) SEQ ID NOs: 889 and 979, respectively.
  • a duplex formed between a sense and antisense strand is in the range of 12-30 nucleotides in length (e.g., 12 to 30, 12 to 27, 12 to 22, 15 to 25, 18 to 30, 18 to 22, 18 to 25, 18 to 27, 18 to 30, 19 to 30 or 21 to 30 nucleotides in length).
  • a duplex between a sense and antisense strand spans the entire length of both the sense strand and the antisense strand.
  • the sense and antisense strands of an oligonucleotide comprise nucleotides sequences selected from the group consisting of: (a) SEQ ID NOs: 940 and 1030, respectively; (b) SEQ ID NOs: 937 and 1027, respectively; and (c) SEQ ID NOs: 939 and 1029, respectively. wherein a duplex formed between a sense and antisense strand is in the range of 12-30 nucleotides in length (e.g., 12 to 30, 12 to 27, 12 to 22, 15 to 25, 18 to 30, 18 to 22, 18 to 25, 18 to 27, 18 to 30, 19 to 30 or 21 to 30 nucleotides in length).
  • a duplex between a sense and antisense strand spans the entire length of both the sense strand and the antisense strand.
  • the sense and antisense strands of an oligonucleotide comprise nucleotides sequences selected from the group consisting of: (a) SEQ ID NOs: 915 and 1005, respectively; (b) SEQ ID NOs: 924 and 1014, respectively; (c) SEQ ID NOs: 913 and 1003, respectively; and (d) SEQ ID NOs: 920 and 1010, respectively.
  • an oligonucleotide disclosed herein comprises a sense strand and an antisense strand, wherein the termini of either or both strands comprise a blunt end.
  • an oligonucleotide herein comprises sense and antisense strands that are separate strands which form an asymmetric duplex region having an overhang at the 3’ terminus of the antisense strand.
  • an oligonucleotide herein comprises a sense strand and an antisense strand, wherein the termini of either or both strands comprise an overhang comprising one or more nucleotides.
  • the one or more nucleotides comprising the overhang are unpaired nucleotides.
  • an oligonucleotide herein comprises a sense strand and an antisense strand, wherein the 3’ termini of the sense strand and the 5’ termini of the antisense strand comprise a blunt end. In some embodiments, an oligonucleotide herein comprises a sense strand and an antisense strand, wherein the 5’ termini of the sense strand and the 3’ termini of the antisense strand comprise a blunt end. [00213] In some embodiments, an oligonucleotide herein comprises a sense strand and an antisense strand, wherein the 3’ terminus of either or both strands comprise a 3’-overhang comprising one or more nucleotides.
  • an oligonucleotide herein comprises a sense strand and an antisense strand, wherein the sense strand comprises a 3’- overhang comprising one or more nucleotides. In some embodiments, an oligonucleotide herein comprises a sense strand and an antisense strand, wherein the antisense strand comprises a 3’-overhang comprising one or more nucleotides. In some embodiments, an oligonucleotide herein comprises a sense strand and an antisense strand, wherein both the sense strand and the antisense strand comprises a 3’-overhang comprising one or more nucleotides.
  • the 3’-overhang is about one (1) to twenty (20) nucleotides in length (e.g., about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or about 20 nucleotides in length).
  • the 3’ overhang is about one (1) to nineteen (19), one (1) to eighteen (18), one (1) to seventeen (17), one (1) to sixteen (16), one (1) to fifteen (15), one (1) to fourteen (14), one (1) to thirteen (13), one (1) to twelve (12), one (1) to eleven (11), one (1) to ten (10), one (1) to nine (9), one (1) to eight (8), one (1) to seven (7), one (1) to six (6), one (1) to five (5), one (1) to four (4), one (1) to three (3), or about one (1) to two (2) nucleotides in length.
  • the 3’-overhang is (1) nucleotide in length. In some embodiments, the 3’-overhang is two (2) nucleotides in length.
  • the 3’-overhang is three (3) nucleotides in length. In some embodiments, the 3’-overhang is four (4) nucleotides in length. In some embodiments, the 3’- overhang is five (5) nucleotides in length. In some embodiments, the 3’-overhang is six (6) nucleotides in length. In some embodiments, the 3’-overhang is seven (7) nucleotides in length. In some embodiments, the 3’-overhang is eight (8) nucleotides in length. In some embodiments, the 3’-overhang is nine (9) nucleotides in length. In some embodiments, the 3’- overhang is ten (10) nucleotides in length.
  • the 3’-overhang is eleven (11) nucleotides in length. In some embodiments, the 3’-overhang is twelve (12) nucleotides in length. In some embodiments, the 3’-overhang is thirteen (13) nucleotides in length. In some embodiments, the 3’-overhang is fourteen (14) nucleotides in length. In some embodiments, the 3’-overhang is fifteen (15) nucleotides in length. In some embodiments, the 3’-overhang is sixteen (16) nucleotides in length. In some embodiments, the 3’-overhang is seventeen (17) nucleotides in length. In some embodiments, the 3’-overhang is eighteen (18) nucleotides in length.
  • an oligonucleotide herein comprises a sense strand and an antisense strand, wherein the 5’ terminus of either or both strands comprise a 5’-overhang comprising one or more nucleotides. In some embodiments, an oligonucleotide herein comprises a sense strand and an antisense strand, wherein the sense strand comprises a 5’- overhang comprising one or more nucleotides.
  • an oligonucleotide herein comprises a sense strand and an antisense strand, wherein the antisense strand comprises a 5’-overhang comprising one or more nucleotides.
  • an oligonucleotide herein comprises a sense strand and an antisense strand, wherein both the sense strand and the antisense strand comprises a 5’-overhang comprising one or more nucleotides.
  • the 5’-overhang is about one (1) to twenty (20) nucleotides in length (e.g., about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or about 20 nucleotides in length).
  • the 5’ overhang is about one (1) to nineteen (19), one (1) to eighteen (18), one (1) to seventeen (17), one (1) to sixteen (16), one (1) to fifteen (15), one (1) to fourteen (14), one (1) to thirteen (13), one (1) to twelve (12), one (1) to eleven (11), one (1) to ten (10), one (1) to nine (9), one (1) to eight (8), one (1) to seven (7), one (1) to six (6), one (1) to five (5), one (1) to four (4), one (1) to three (3), or about one (1) to two (2) nucleotides in length.
  • the 5’-overhang is (1) nucleotide in length.
  • the 5’-overhang is two (2) nucleotides in length.
  • the 5’-overhang is three (3) nucleotides in length. In some embodiments, the 5’-overhang is four (4) nucleotides in length. In some embodiments, the 5’- overhang is five (5) nucleotides in length. In some embodiments, the 5’-overhang is six (6) nucleotides in length. In some embodiments, the 5’-overhang is seven (7) nucleotides in length. In some embodiments, the 5’-overhang is eight (8) nucleotides in length. In some embodiments, the 5’-overhang is nine (9) nucleotides in length. In some embodiments, the 5’- overhang is ten (10) nucleotides in length.
  • the 5’-overhang is eleven (11) nucleotides in length. In some embodiments, the 5’-overhang is twelve (12) nucleotides in length. In some embodiments, the 5’-overhang is thirteen (13) nucleotides in length. In some embodiments, the 5’-overhang is fourteen (14) nucleotides in length. In some embodiments, the 5’-overhang is fifteen (15) nucleotides in length. In some embodiments, the 5’-overhang is sixteen (16) nucleotides in length. In some embodiments, the 5’-overhang is seventeen (17) nucleotides in length. In some embodiments, the 5’-overhang is eighteen (18) nucleotides in length.
  • the 5’-overhang is nineteen (19) nucleotides in length. In some embodiments, the 5’-overhang is twenty (20) nucleotides in length. [00217] In some embodiments, one or more (e.g., 2, 3, 4, 5, or more) nucleotides comprising the 3’ terminus or 5’ terminus of a sense and/or antisense strand are modified. For example, in some embodiments, one or two terminal nucleotides of the 3’ terminus of the antisense strand are modified.
  • the last nucleotide at the 3’ terminus of an antisense strand is modified, such that it comprises 2’ modification, or it comprises, a 2’- O-methoxyethyl.
  • the last one or two terminal nucleotides at the 3’ terminus of an antisense strand are complementary with the target.
  • the last one or two nucleotides at the 3’ terminus of the antisense strand are not complementary with the target.
  • an oligonucleotide disclosed herein comprises a sense strand and an antisense strand, wherein the 3’ terminus of the sense strand comprises a step-loop described herein and the 3’ terminus of the antisense strand comprises a 3’-overhang described herein.
  • an oligonucleotide herein (e.g., an RNAi oligonucleotide) comprises a sense strand and an antisense strand that form a nicked tetraloop structure described herein, wherein the 3’ terminus of the sense strand comprises a stem-loop, wherein the loop is a tetraloop described herein, and wherein the 3’ terminus of the antisense strand comprises a 3’-overhang described herein.
  • the 3’-overhang is two (2) nucleotides in length.
  • the two (2) nucleotides comprising the 3’-overhang both comprise guanine (G) nucleobases.
  • a modified sugar (also referred herein to a sugar analog) includes a modified deoxyribose or ribose moiety in which, for example, one or more modifications occur at the 2′, 3′, 4′ and/or 5′ carbon position of the sugar.
  • a modified sugar may also include non-natural alternative carbon structures such as those present in locked nucleic acids (“LNA”; see, e.g., Koshkin et al., (1998) TETRAHEDON 54:3607-3630), unlocked nucleic acids (“UNA”; see, e.g., Snead et al., (2013) MOL. THER-NUCL. ACIDS 2:e103) and bridged nucleic acids (“BNA”; see, e.g., Imanishi and Obika (2002) CHEM COMMUN. (CAMB) 21:1653-1659).
  • LNA locked nucleic acids
  • NDA unlocked nucleic acids
  • BNA bridged nucleic acids
  • a nucleotide modification in a sugar comprises a 2′- modification.
  • a 2′-modification may be 2′-O-propargyl, 2′-O- propylamin, 2′-amino, 2′-ethyl, 2′-fluoro (2′-F), 2′-aminoethyl (EA), 2′-O-methyl (2′-OMe), 2′-O-methoxyethyl (2′-MOE), 2′-O-[2-(methylamino)-2-oxoethyl] (2′-O-NMA) or 2′-deoxy- 2′-fluoro- ⁇ -d-arabinonucleic acid (2′-FANA).
  • the modification is 2′- F, 2′-OMe or 2′-MOE.
  • a modification in a sugar comprises a modification of the sugar ring, which may comprise modification of one or more carbons of the sugar ring.
  • a modification of a sugar of a nucleotide may comprise a 2′- oxygen of a sugar is linked to a 1′-carbon or 4′-carbon of the sugar, or a 2′-oxygen is linked to the 1′-carbon or 4′-carbon via an ethylene or methylene bridge.
  • a modified nucleotide has an acyclic sugar that lacks a 2′-carbon to 3′-carbon bond.
  • a modified nucleotide has a thiol group, e.g., in the 4′ position of the sugar.
  • the oligonucleotide described herein comprises at least about 1 modified nucleotide (e.g., at least 1, at least 5, at least 10, at least 15, at least 20, at least 25, at least 30, at least 35, at least 40, at least 45, at least 50, at least 55, at least 60, or more).
  • the sense strand of the oligonucleotide comprises at least about 1 modified nucleotide (e.g., at least 1, at least 5, at least 10, at least 15, at least 20, at least 25, at least 30, at least 35, or more).
  • the antisense strand of the oligonucleotide comprises at least about 1 modified nucleotide (e.g., at least 1, at least 5, at least 10, at least 15, at least 20, or more). [00222] In some embodiments, all the nucleotides of the sense strand of the oligonucleotide are modified. In some embodiments, all the nucleotides of the antisense strand of the oligonucleotide are modified. In some embodiments, all the nucleotides of the oligonucleotide (i.e., both the sense strand and the antisense strand) are modified.
  • the modified nucleotide comprises a 2′-modification (e.g., a 2′-F or 2′-OMe, 2′-MOE, and 2′-deoxy-2′-fluoro- ⁇ -d-arabinonucleic acid).
  • the modified nucleotide comprises a 2′-modification (e.g., a 2′-F or 2′-OMe).
  • the disclosure provides oligonucleotides having different modification patterns.
  • an oligonucleotide herein comprises a sense strand having a modification pattern as set forth in the Examples and Sequence Listing and an antisense strand having a modification pattern as set forth in the Examples and Sequence Listing.
  • an oligonucleotide disclosed herein e.g., an RNAi oligonucleotide
  • an oligonucleotide herein comprises an antisense strand comprising nucleotides that are modified with 2′-F and 2′-OMe.
  • an oligonucleotide disclosed herein comprises a sense strand having nucleotides that are modified with 2′-F. In some embodiments, an oligonucleotide disclosed herein comprises a sense strand comprises nucleotides that are modified with 2′-F and 2′-OMe. [00225] In some embodiments, an oligonucleotide described herein comprises a sense strand with about 10-15%, 10%, 11%, 12%, 13%, 14% or 15% of the nucleotides of the sense strand comprising a 2’-fluoro modification. In some embodiments, about 11% of the nucleotides of the sense strand comprise a 2-fluoro modification.
  • an oligonucleotide described herein comprises an antisense strand with about 25-35%, 25%, 26%, 27%, 28%, 29%, 30%, 31%, 32%, 33%, 34% or 35% of the nucleotides of the antisense strand comprising a 2’-fluoro modification. In some embodiments, about 32% of the nucleotides of the antisense strand comprise a 2’-fluoro modification. In some embodiments, the oligonucleotide has about 15-25%, 15%, 16%, 17%, 18%, 19%, 20%, 21%, 22%, 23%, 24%, or 25% of its nucleotides comprising a 2’-fluoro modification.
  • the modified oligonucleotides comprise a sense strand sequence having a modification pattern as set forth in FIG 1A or Example 7 and an antisense strand having a modification pattern as set forth in FIG 1A or Example 7.
  • one or more of positions 8, 9, 10 or 11 of the sense strand is modified with a 2′-F group.
  • the sugar moiety at each of nucleotides at positions 1-7 and 12-20 in the sense strand is modified with a 2′-OMe.
  • the antisense strand has 3 nucleotides that are modified at the 2′-position of the sugar moiety with a 2′-F.
  • the sugar moiety at positions 2, 5 and 14 and optionally up to 3 of the nucleotides at positions 1, 3, 7 and 10 of the antisense strand are modified with a 2′-F.
  • the sugar moiety at positions 2, 5 and 14 and optionally up to 3 of the nucleotides at positions 3, 4, 7 and 10 of the antisense strand are modified with a 2′-F.
  • the sugar moiety at each of the positions at positions 2, 5 and 14 of the antisense strand is modified with the 2′-F.
  • the sugar moiety at each of the positions at positions 1, 2, 5 and 14 of the antisense strand is modified with the 2′-F.
  • the sugar moiety at each of the positions at positions 2, 4, 5 and 14 of the antisense strand is modified with the 2′-F.
  • the sugar moiety at each of the positions at positions 1, 2, 3, 5, 7 and 14 of the antisense strand is modified with the 2′-F. In other embodiments, the sugar moiety at each of the positions at positions 2, 3, 4, 5, 7 and 14 of the antisense strand is modified with the 2′-F. In yet another embodiment, the sugar moiety at each of the positions at positions 1, 2, 3, 5, 10 and 14 of the antisense strand is modified with the 2′-F. In other embodiments, the sugar moiety at each of the positions at positions 2, 3, 4, 5, 10 and 14 of the antisense strand is modified with the 2′-F.
  • an oligonucleotide provided herein comprises an antisense strand having the sugar moiety at position 1, position 2, position 3, position 4, position 5, position 6, position 7, position 8, position 9, position 10, position 11, position 12, position 13, position 14, position 15, position 16, position 17, position 18, position 19, position 20, position 21, or position 22 modified with 2′-F.
  • an oligonucleotide provided herein comprises an antisense strand having the sugar moiety at position 1, position 2, position 3, position 4, position 5, position 6, position 7, position 8, position 9, position 10, position 11, position 12, position 13, position 14, position 15, position 16, position 17, position 18, position 19, position 20, position 21, or position 22 modified with 2′-OMe.
  • an oligonucleotide provided herein comprises an antisense strand having the sugar moiety at position 1, position 2, position 3, position 4, position 5, position 6, position 7, position 8, position 9, position 10, position 11, position 12, position 13, position 14, position 15, position 16, position 17, position 18, position 19, position 20, position 21, or position 22 modified with a modification selected from the group consisting of 2′-O-propargyl, 2′-O-propylamin, 2′-amino, 2′-ethyl, 2’-aminoethyl (EA), 2′-O- methyl (2′-OMe), 2′-O-methoxyethyl (2′-MOE), 2′-O-[2-(methylamino)-2-oxoethyl] (2′-O- NMA), and 2′-deoxy-2′-fluoro- ⁇ -d-arabinonucleic acid (2′-FANA).
  • an oligonucleotide provided herein comprises a sense strand having the sugar moiety at positions 8-11 modified with 2′-F. In some embodiments, an oligonucleotide provided herein comprises a sense strand having the sugar moiety at positions 3, 8, 9, 10, 12, 13 and 17 modified with 2′-F. In some embodiments, an oligonucleotide provided herein comprises a sense strand having the sugar moiety at positions 1-7 and 12-17 or 12-20 modified with 2’OMe. In some embodiments, an oligonucleotide provided herein comprises a sense strand having the sugar moiety at positions 1-7, 12-27 and 31-36 modified with 2’OMe.
  • an oligonucleotide provided herein comprises a sense strand having the sugar moiety of each of the nucleotides at positions 1-7 and 12-17 or 12-20 of the sense strand modified with a modification selected from the group consisting of 2′-O-propargyl, 2′-O-propylamin, 2′-amino, 2′-ethyl, 2’-aminoethyl (EA), 2′-O- methyl (2′-OMe), 2′-O-methoxyethyl (2′-MOE), 2′-O-[2-(methylamino)-2-oxoethyl] (2′-O- NMA), and 2′-deoxy-2′-fluoro- ⁇ -d-arabinonucleic acid (2′-FANA).
  • an oligonucleotide provided herein comprises a sense strand having the sugar moiety at positions 1-2, 4-7, 11, 14-16 and 18-20 modified with 2’OMe.
  • an oligonucleotide provided herein comprises a sense strand having the sugar moiety of each of the nucleotides at positions 1-2, 4-7, 11, 14-16 and 18-20 of the sense strand modified with a modification selected from the group consisting of 2′-O-propargyl, 2′-O-propylamin, 2′- amino, 2′-ethyl, 2’-aminoethyl (EA), 2′-O-methyl (2′-OMe), 2′-O-methoxyethyl (2′-MOE), 2′-O-[2-(methylamino)-2-oxoethyl] (2′-O-NMA), and 2′-deoxy-2′-fluoro- ⁇ -d-arabinonucleic acid (2′-FANA).
  • an oligonucleotide provided herein comprises a sense strand having the sugar moiety at position 1, position 2, position 3, position 4, position 5, position 6, position 7, position 8, position 9, position 10, position 11, position 12, position 13, position 14, position 15, position 16, position 17, position 18, position 19, position 20, position 21, position 22, position 23, position 24, position 25, position 26, position 27, position 28, position 29, position 30, position 31, position 32, position 33, position 34, position 35, or position 36 modified with 2′-F.
  • an oligonucleotide provided herein comprises a sense strand having the sugar moiety at position 1, position 2, position 3, position 4, position 5, position 6, position 7, position 8, position 9, position 10, position 11, position 12, position 13, position 14, position 15, position 16, position 17, position 18, position 19, position 20, position 21, position 22, position 23, position 24, position 25, position 26, position 27, position 28, position 29, position 30, position 31, position 32, position 33, position 34, position 35, or position 36 modified with 2′-OMe.
  • an oligonucleotide provided herein comprises a sense strand having the sugar moiety at position 1, position 2, position 3, position 4, position 5, position 6, position 7, position 8, position 9, position 10, position 11, position 12, position 13, position 14, position 15, position 16, position 17, position 18, position 19, position 20, position 21, position 22, position 23, position 24, position 25, position 26, position 27, position 28, position 29, position 30, position 31, position 32, position 33, position 34, position 35, or position 36 modified with a modification selected from the group consisting of 2′-O-propargyl, 2′-O-propylamin, 2′-amino, 2′-ethyl, 2’-aminoethyl (EA), 2′-O-methyl (2′- OMe), 2′-O-methoxyethyl (2′-MOE), 2′-O-[2-(methylamino)-2-oxoethyl] (2′-O-NMA), and 2′-deoxy-2′-fluoro- ⁇ -d-arabinonu
  • 5′-terminal phosphate groups of oligonucleotides enhance the interaction with Ago2.
  • oligonucleotides comprising a 5′-phosphate group may be susceptible to degradation via phosphatases or other enzymes, which can limit their bioavailability in vivo.
  • oligonucleotides include analogs of 5′ phosphates that are resistant to such degradation.
  • a phosphate analog may be oxymethylphosphonate, vinylphosphonate or malonyl phosphonate.
  • an oligonucleotide strand is attached to chemical moiety that mimics the electrostatic and steric properties of a natural 5′-phosphate group (“phosphate mimic”).
  • phosphate mimic a natural 5′-phosphate group
  • an oligonucleotide has a phosphate analog at a 4′- carbon position of the sugar (referred to as a “4′-phosphate analog”). See, e.g., Intl. Patent Application Publication No. WO 2018/045317.
  • an oligonucleotide herein comprises a 4′-phosphate analog at a 5′-terminal nucleotide.
  • a phosphate analog is an oxymethylphosphonate, in which the oxygen atom of the oxymethyl group is bound to the sugar moiety (e.g., at its 4′-carbon) or analog thereof.
  • a 4′-phosphate analog is a thiomethyl phosphonate or an amino methyl phosphonate, in which the sulfur atom of the thiomethyl group or the nitrogen atom of the amino methyl group is bound to the 4′-carbon of the sugar moiety or analog thereof.
  • a 4′-phosphate analog is an oxymethyl phosphonate.
  • an oxymethyl phosphonate is represented by the formula –O–CH 2 –PO(OH) 2 or –O–CH 2 –PO(OR) 2 , in which R is independently selected from H, CH 3 , an alkyl group, CH 2 CH 2 CN, CH 2 OCOC(CH3)3, CH 2 OCH 2 CH 2 Si (CH3)3 or a protecting group.
  • the alkyl group is CH 2 CH 3 . More typically, R is independently selected from H, CH3 or CH 2 CH3.
  • an oligonucleotide provided herein comprises an antisense strand comprising a 4′-phosphate analog at the 5′-terminal nucleotide, wherein 5’- terminal nucleotide comprises the following structure (Chem 1):
  • an oligonucleotide may comprise a modified internucleoside linkage.
  • phosphate modifications or substitutions may result in an oligonucleotide that comprises at least about 1 (e.g., at least 1, at least 2, at least 3 or at least 5) modified internucleotide linkage.
  • any one of the oligonucleotides disclosed herein comprises about 1 to about 10 (e.g., 1 to 10, 2 to 8, 4 to 6, 3 to 10, 5 to 10, 1 to 5, 1 to 3 or 1 to 2) modified internucleotide linkages. In some embodiments, any one of the oligonucleotides disclosed herein comprises 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10 modified internucleotide linkages.
  • a modified internucleotide linkage may be a phosphorodithioate linkage, 4′- O-methylene phosphonate linkage, a phosphorothioate linkage, a phosphotriester linkage, a thionoalkylphosphonate linkage, a thionalkylphosphotriester linkage, a phosphoramidite linkage, a phosphonate linkage or a boranophosphate linkage.
  • at least one modified internucleotide linkage of any one of the oligonucleotides as disclosed herein is a phosphorothioate linkage.
  • At least one modified internucleotide linkage of any one of the oligonucleotides as disclosed herein is a 4′-O-methylene phosphonate linkage.
  • the oligonucleotide described herein has a phosphorothioate linkage between one or more of positions 1 and 2 of the sense strand, positions 1 and 2 of the antisense strand, positions 2 and 3 of the antisense strand, positions 3 and 4 of the antisense strand, positions 20 and 21 of the antisense strand, and positions 21 and 22 of the antisense strand.
  • the oligonucleotide described herein has a phosphorothioate linkage between each of positions 1 and 2 of the sense strand, positions 1 and 2 of the antisense strand, positions 2 and 3 of the antisense strand, positions 20 and 21 of the antisense strand, and positions 21 and 22 of the antisense strand.
  • Base Modifications [00241]
  • oligonucleotides herein have one or more modified nucleobases.
  • modified nucleobases also referred to herein as base analogs
  • a modified nucleobase is a nitrogenous base.
  • a modified nucleobase does not contain nitrogen atom. See, e.g., US Patent Application Publication No. 2008/0274462.
  • a modified nucleotide comprises a universal base. However, in certain embodiments, a modified nucleotide does not contain a nucleobase (abasic).
  • a universal base is a heterocyclic moiety located at the 1′ position of a nucleotide sugar moiety in a modified nucleotide, or the equivalent position in a nucleotide sugar moiety substitution, that, when present in a duplex, can be positioned opposite more than one type of base without substantially altering structure of the duplex.
  • a single-stranded nucleic acid containing a universal base forms a duplex with the target nucleic acid that has a lower T m than a duplex formed with the complementary nucleic acid.
  • the single- stranded nucleic acid containing the universal base forms a duplex with the target nucleic acid that has a higher T m than a duplex formed with the nucleic acid comprising the mismatched base.
  • Non-limiting examples of universal-binding nucleotides include, but are not limited to, inosine, 1- ⁇ -D-ribofuranosyl-5-nitroindole and/or 1- ⁇ -D-ribofuranosyl-3- nitropyrrole (see, US Patent Application Publication No.2007/0254362; Van Aerschot et al., (1995) NUCLEIC ACIDS RES.23:4363-4370; Loakes et al., (1995) NUCLEIC ACIDS RES. 23:2361-66; and Loakes and Brown (1994) NUCLEIC ACIDS RES.22:4039-43). e.
  • a reversibly modified nucleotide comprises a glutathione-sensitive moiety.
  • nucleic acid molecules have been chemically modified with cyclic disulfide moieties to mask the negative charge created by the internucleotide diphosphate linkages and improve cellular uptake and nuclease resistance.
  • cyclic disulfide moieties to mask the negative charge created by the internucleotide diphosphate linkages and improve cellular uptake and nuclease resistance.
  • these larger chemical groups can be engineered to confer various advantages to the nucleotide or oligonucleotide, such as nuclease resistance, lipophilicity, charge, thermal stability, specificity, and reduced immunogenicity.
  • the structure of the glutathione-sensitive moiety can be engineered to modify the kinetics of its release. [00247]
  • a glutathione-sensitive moiety is attached to the sugar of the nucleotide.
  • a glutathione-sensitive moiety is attached to the 2′- carbon of the sugar of a modified nucleotide.
  • the glutathione-sensitive moiety is located at the 5′-carbon of a sugar, particularly when the modified nucleotide is the 5′-terminal nucleotide of the oligonucleotide. In some embodiments, the glutathione-sensitive moiety is located at the 3′-carbon of sugar, particularly when the modified nucleotide is the 3′-terminal nucleotide of the oligonucleotide. In some embodiments, the glutathione-sensitive moiety comprises a sulfonyl group.
  • Targeting Ligands it is desirable to target the STAT3 targeting oligonucleotides of the disclosure to one or more cells or one or more organs. Such a strategy can help to avoid undesirable effects in other organs or avoid undue loss of the oligonucleotide to cells, tissue or organs that would not benefit from the oligonucleotide. Targeting of oligonucleotides to one or more cells or one or more organs can be achieved through a variety of approaches.
  • Conjugation of oligonucleotides to tissue or cell specific antibodies, small molecules or targeting ligands can facilitate delivery to and modify accumulation of the oligonucleotide in one or more target cells or tissues (Chernolovskaya et al., (2019) FRONT PHARMACOL.10:444).
  • conjugation of an oligonucleotide to a saturated fatty acid may facilitate delivery to cells or tissues like adipose tissue or immune cells which uptake such ligands more readily than conventional oligonucleotide ligands.
  • oligonucleotides disclosed herein are modified to facilitate targeting and/or delivery of a tissue, cell, or organ (e.g., to facilitate delivery of the oligonucleotide to the liver).
  • oligonucleotides disclosed herein are modified to facilitate delivery of the oligonucleotide to cells of the immune system.
  • oligonucleotides disclosed herein are modified to facilitate delivery of the oligonucleotide to myeloid derived suppressor cells.
  • an oligonucleotide comprises at least one nucleotide (e.g., 1, 2, 3, 4, 5, 6 or more nucleotides) conjugated to one or more targeting ligand(s).
  • the targeting ligand comprises a carbohydrate, amino sugar, cholesterol, peptide, polypeptide, protein, or part of a protein (e.g., an antibody or antibody fragment), or lipid.
  • the targeting ligand is an aptamer.
  • a targeting ligand may be an RGD peptide that is used to target tumor vasculature or glioma cells, CREKA peptide to target tumor vasculature or stoma, transferring, lactoferrin, or an aptamer to target transferrin receptors expressed on CNS vasculature, or an anti-EGFR antibody to target EGFR on glioma cells.
  • the targeting ligand is one or more GalNAc moieties.
  • 1 or more (e.g., 1, 2, 3, 4, 5 or 6) nucleotides of an oligonucleotide are each conjugated to a separate targeting ligand.
  • 2 to 4 nucleotides of an oligonucleotide are each conjugated to a separate targeting ligand.
  • targeting ligands are conjugated to 2 to 4 nucleotides at either ends of the sense or antisense strand (e.g., targeting ligands are conjugated to a 2 to 4 nucleotide overhang or extension on the 5′ or 3′ end of the sense or antisense strand) such that the targeting ligands resemble bristles of a toothbrush and the oligonucleotide resembles a toothbrush.
  • an oligonucleotide may comprise a stem-loop at either the 5′ or 3′ end of the sense strand and 1, 2, 3 or 4 nucleotides of the loop of the stem may be individually conjugated to a targeting ligand.
  • an oligonucleotide e.g., a dsRNA
  • an oligonucleotide provided by the disclosure comprises a stem-loop at the 3′ end of the sense strand, wherein the loop of the stem-loop comprises a triloop or a tetraloop, and wherein the 3 or 4 nucleotides comprising the triloop or tetraloop, respectfully, are individually conjugated to a targeting ligand.
  • an oligonucleotide provided by the disclosure comprises a stem-loop at the 3′ terminus of the sense strand, wherein the loop of the stem-loop comprises a tetraloop, and wherein 3 nucleotides of the tetraloop are individually conjugated to a targeting ligand.
  • GalNAc is a high affinity ligand for the ASGPR, which is primarily expressed on the sinusoidal surface of hepatocyte cells and has a major role in binding, internalizing and subsequent clearing circulating glycoproteins that contain terminal galactose or GalNAc residues (asialoglycoproteins).
  • Conjugation (either indirect or direct) of GalNAc moieties to oligonucleotides of the instant disclosure can be used to target these oligonucleotides to the ASGPR expressed on cells.
  • an oligonucleotide of the instant disclosure is conjugated to at least one or more GalNAc moieties, wherein the GalNAc moieties target the oligonucleotide to an ASGPR expressed on human liver cells (e.g., human hepatocytes).
  • the GalNAc moiety target the oligonucleotide to the liver.
  • an oligonucleotide of the instant disclosure is conjugated directly or indirectly to a monovalent GalNAc.
  • the oligonucleotide is conjugated directly or indirectly to more than one monovalent GalNAc (i.e., is conjugated to 2, 3 or 4 monovalent GalNAc moieties, and is typically conjugated to 3 or 4 monovalent GalNAc moieties).
  • an oligonucleotide is conjugated to one or more bivalent GalNAc, trivalent GalNAc or tetravalent GalNAc moieties.
  • nucleotides of an oligonucleotide are each conjugated to a GalNAc moiety.
  • 2 to 4 nucleotides of a tetraloop are each conjugated to a separate GalNAc.
  • 1 to 3 nucleotides of a triloop are each conjugated to a separate GalNAc.
  • targeting ligands are conjugated to 2 to 4 nucleotides at either ends of the sense or antisense strand (e.g., ligands are conjugated to a 2 to 4 nucleotide overhang or extension on the 5′ or 3′ end of the sense or antisense strand) such that the GalNAc moieties resemble bristles of a toothbrush and the oligonucleotide resembles a toothbrush.
  • GalNAc moieties are conjugated to a nucleotide of the sense strand.
  • GalNAc moieties can be conjugated to nucleotides in the tetraloop of the sense strand where each GalNAc moiety is conjugated to 1 nucleotide.
  • the tetraloop is any combination of adenine and guanine nucleotides.
  • an oligonucleotide herein comprises a monovalent GalNAc attached to a guanine nucleotide referred to as [ademG-GalNAc] or 2′- aminodiethoxymethanol-Guanine-GalNAc, as depicted below in Chem 4: [00258] In some embodiments, an oligonucleotide herein comprises a monovalent GalNAc attached to an adenine nucleotide, referred to as [ademA-GalNAc] or 2′- aminodiethoxymethanol-Adenine-GalNAc, as depicted below in Chem 5:
  • a targeting ligand is conjugated to a nucleotide using a click linker.
  • an acetal-based linker is used to conjugate a targeting ligand to a nucleotide of any one of the oligonucleotides described herein. Acetal-based linkers are disclosed, for example, in Intl. Patent Application Publication No. WO 2016/100401.
  • the linker is a labile linker. However, in other embodiments, the linker is stable.
  • a loop comprising from 5′ to 3′ the nucleotides GAAA, in which GalNAc moieties are attached to nucleotides of the loop using an acetal linker (Chem 7 and Chem 8).
  • a loop may be present, for example, at positions 27-30 of the any one of the sense strand as shown in FIG. 1.
  • a targeting ligand is conjugated to a nucleotide using a click linker.
  • an acetal-based linker is used to conjugate a targeting ligand to a nucleotide of any one of the oligonucleotides described herein. Acetal-based linkers are disclosed, for example, in Intl. Patent Application Publication No. WO 2016/100401.
  • the linker is a labile linker.
  • the linker is a stable linker.
  • a duplex extension (e.g., of up to 3, 4, 5 or 6 bp in length) is provided between a targeting ligand (e.g., a GalNAc moiety) and a dsRNA.
  • a targeting ligand e.g., a GalNAc moiety
  • a dsRNA e.g., a dsRNA.
  • the oligonucleotides herein do not have a GalNAc conjugated thereto.
  • a STAT3 targeting oligonucleotide described herein comprises a nucleotide sequence having a region of complementarity to a STAT3 mRNA target sequence and one or more targeting ligands, wherein the nucleotide sequence comprises one or more nucleosides (nucleic acids) conjugated with one or more targeting ligands represented by formula I-a: or a pharmaceutically acceptable salt thereof, wherein: B is a nucleobase or hydrogen; R 1 and R 2 are independently hydrogen, halogen, R A , -CN, -S(O)R, -S(O) 2 R, -Si(OR) 2 R, - Si(OR)R2, or -SiR3; or R 1 and R 2 on the same carbon are taken together with their intervening atoms to form a 3-7 membered saturated or partially unsaturated ring having 0-3 hetero
  • the STAT3 targeting oligonucleotide comprises one or more nucleic acids conjugated with targeting ligands represented by formula II-a: or a pharmaceutically acceptable salt thereof.
  • the STAT3 targeting oligonucleotide comprises one or more nucleic acids conjugated with targeting ligands represented by formula II-b or II-c: II-c or a pharmaceutically acceptable salt thereof, wherein: L 1 is a covalent bond, a monovalent or a bivalent saturated or unsaturated, straight or branched C 1-50 hydrocarbon chain, wherein 0-10 methylene units of the hydrocarbon chain are independently replaced by -Cy-, -O-, -C(O)NR-, -NR-, -S-, -C(O)-, -C(O)O-, -S(O) 2 -, -P(O)OR-, -P(S)OR-, -P(S)OR-, -P(S)OR-, -P(S
  • R 5 is . In some . , embodiments, . In some embodiments, R 5 is some embodiments, R 5 is some embodiments, R 5 is . In some embodiments, R 5 is . [00269] In some embodiments, the STAT3 targeting oligonucleotide comprises one or more nucleic acids conjugated with targeting ligands represented by formula II-Ib or II-Ic:
  • B is a nucleobase or hydrogen; m is 1-50; X 1 is -O-, or -S-; Y is hydrogen, R 3 is hydrogen, or a suitable protecting group; X 2 is O, or S; X 3 is -O-, -S-, or a covalent bond; Y 1 is a linking group attaching to the 2′- or 3′-terminal of a nucleoside, a nucleotide, or an oligonucleotide; Y 2 is hydrogen, a phosphoramidite analogue, an internucleotide linking group attaching to the 5′-terminal of a nucleoside, a nucleotide, or an oligonucleotide, or a linking group attaching to a solid support; R 5 is adamantyl, or a saturated or unsaturated, straight, or branched C 1-50 hydrocarbon chain, wherein 0-10
  • R 5 is selected from . [00271] In some embodiments, R 5 is . [00273] In some embodiments, the nucleotide sequence of the STAT3 targeting oligonucleotide comprises 1-10 targeting ligands. In some embodiments, the nucleotide sequence comprises 1, 2 or 3 targeting ligands. [00274] In some embodiments, the STAT3 targeting oligonucleotide is a double- stranded molecule. In some embodiments, the STAT3 targeting oligonucleotide is an RNAi molecule. In some embodiments, the STAT3 targeting double stranded oligonucleotide comprises a stem loop.
  • the ligand is conjugated to any of the nucleotides in the stem loop. In some embodiments, the ligand is conjugated to the first nucleotide from 5’ to 3’, in the stem loop. In some embodiments, the ligand is conjugated to the second nucleotide from 5’ to 3’ in the stem loop. In some embodiments, the ligand is conjugated to the third nucleotide from 5’ to 3’ in the stem loop. In some embodiments, the ligand is conjugated to the fourth nucleotide from 5’ to 3’ in the stem loop. In some embodiments, the ligand is conjugated to one, two, three, or four of the nucleotides in the stem loop.
  • the ligand is conjugated to three of the nucleotides in the stem loop.
  • the STAT3 targeting double stranded oligonucleotide comprises a stem loop, wherein one or more lipids are conjugated to one or more nucleotides of the stem loop.
  • the STAT3 targeting double stranded oligonucleotide comprises a stem loop, wherein one or more C16 lipids are conjugated to one or more nucleotides of the stem loop.
  • the STAT3 targeting double stranded oligonucleotide comprises a stem loop, wherein one or more C18 lipids are conjugated to one or more nucleotides of the stem loop.
  • the STAT3 targeting oligonucleotide comprises a sense strand of 36 nucleotides with positions numbered 1-36 from 5’ to 3’.
  • the STAT3 targeting oligonucleotide comprises a lipid conjugated to position 27 of a 36- nucleotide sense strand.
  • STAT3 targeting oligonucleotide comprises a lipid conjugated to position 28 of a 36-nucleotide sense strand.
  • the STAT3 targeting oligonucleotide comprises a lipid conjugated to position 29 of a 36- nucleotide sense strand. In some embodiments, the STAT3 targeting oligonucleotide comprises a lipid conjugated to position 30 of a 36-nucleotide sense strand. In some embodiments, a 36-nucleotide sense strand forms a stem loop having a loop with positions 27-30. In some embodiments, a lipid is conjugated to more than one position of the loop (e.g., positions 27 and 28 of a 36-nucleotide sense strand).
  • the STAT3 targeting oligonucleotide comprises a C16 lipid conjugated to position 27 of a 36-nucleotide sense strand. In some embodiments, STAT3 targeting oligonucleotide comprises a C16 lipid conjugated to position 28 of a 36- nucleotide sense strand. In some embodiments, the STAT3 targeting oligonucleotide comprises a C16 lipid conjugated to position 29 of a 36-nucleotide sense strand. In some embodiments, the STAT3 targeting oligonucleotide comprises a C16 lipid conjugated to position 30 of a 36-nucleotide sense strand.
  • a 36-nucleotide sense strand forms a stem loop having a loop with positions 27-30.
  • a C16 lipid is conjugated to more than one position of the loop (e.g., positions 27 and 28 of a 36- nucleotide sense strand).
  • the STAT3 targeting oligonucleotide comprises a C18 lipid conjugated to position 27 of a 36-nucleotide sense strand.
  • STAT3 targeting oligonucleotide comprises a C18 lipid conjugated to position 28 of a 36- nucleotide sense strand.
  • the STAT3 targeting oligonucleotide comprises a C18 lipid conjugated to position 29 of a 36-nucleotide sense strand. In some embodiments, the STAT3 targeting oligonucleotide comprises a C18 lipid conjugated to position 30 of a 36-nucleotide sense strand. In some embodiments, a 36-nucleotide sense strand forms a stem loop having a loop with positions 27-30. In some embodiments, a C18 lipid is conjugated to more than one position of the loop (e.g., positions 27 and 28 of a 36- nucleotide sense strand).
  • a STAT3 targeting oligonucleotide comprises an antisense strand of 15 to 30 nucleotides and a sense strand of 15 to 40 nucleotide, wherein the sense and antisense strands form a duplex region, wherein the antisense strand comprises a region of complementarity to a STAT3 mRNA target sequence expressed in an immune cell associated with a tumor microenvironment, wherein the sense strand comprises at its 3’ end a stem-loop comprising a tetraloop comprising 4 nucleosides, wherein one or more of the 4 nucleosides is represented by formula II-Ib: , wherein B is selected from an adenine and a guanine nucleobase, and wherein R 5 is a hydrocarbon chain.
  • m is 1, X1 is O, Y2 is an internucleotide linking group attaching to the 5’ terminal of a nucleoside, Y 1 Y is represented by , Y1 is a linking group attaching to the 2’ or 3’ terminal of a nucleotide, X2 is O, X3 is O, and R3 is H.
  • the hydrocarbon chain is a C8-C30 hydrocarbon chain. In some embodiments, the hydrocarbon chain is a C16 hydrocarbon chain. In some embodiments, the C16 hydrocarbon chain is represented by . In some embodiments, the hydrocarbon chain is a C18 hydrocarbon chain. In some embodiments, the C18 hydrocarbon chain is represented by .
  • the oligonucleotide comprises a sense strand comprising a sequence selected from SEQ ID NOs: 89-280, wherein the sense strand comprises a C18 lipid.
  • the 4 nucleosides of the tetraloop are numbered 1-4 from 5’ to 3’ and position 1 is represented by formula II-Ib.
  • position 2 is represented by formula II-Ib.
  • position 3 is represented by formula II-Ib.
  • position 4 is represented by formula II- Ib.
  • the sense strand is 36 nucleotides with positions numbered 1-36 from 5’ to 3’, wherein the stem-loop comprises nucleotides at positions 21-36, and wherein one or more nucleosides at positions 27-30 are represented by formula II-Ib.
  • the antisense strand is 22 nucleotides.
  • an oligonucleotide targeting STAT3 comprises a sense strand and an antisense strand as set forth in Tables 1, 2, 3, 8, 9, 10, 11, and 12, wherein the oligonucleotide comprises a stem loop structure having a double-stranded stem of about 2-6 base pairs and a loop of 3-4 nucleotides, and wherein the sense and antisense strands comprise the modification pattern set forth in FIG.1A or Example 7.
  • an oligonucleotide targeting STAT3 comprises a sense strand and an antisense strand as set forth in Tables 1, 2, 3, 8, 9, 10, 11, and 12, wherein the oligonucleotide comprises a stem loop structure having a double-stranded stem of about 2-6 base pairs and a loop of 3-4 nucleotides, wherein the sense and antisense strands comprise the modification pattern set forth in FIG.1A, and wherein antisense strand is modified with an oxymethylphosphonate at the 4’ carbon of the 5’ terminal nucleotide.
  • the oligonucleotide comprises a stem loop comprising the nucleotide sequence of SEQ ID NO: 86.
  • the oligonucleotide comprises a double-stranded stem of 6 base pairs and a stem loop of 4 nucleotides comprising one, two, three or four GalNAc conjugated nucleotides.
  • the GalNAc conjugated nucleotide is a monovalent GalNAc conjugated to an adenine nucleotide, referred to as [ademA-GalNAc] or 2′- aminodiethoxymethanol-Adenine-GalNAc, as depicted below: [00283]
  • the stem loop comprises a double-stranded stem of 6 base pairs and a loop comprising the nucleotide sequence GAAA, wherein each adenine nucleotide is ademA-GalNAc.
  • an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense and antisense strand comprising nucleotide sequences selected from: (a) SEQ ID NOs: 9 and 10, respectively; (b) SEQ ID NOs: 37 and 38, respectively; (c) SEQ ID NOs: 65 and 66, respectively; and (d) SEQ ID NOs: 69 and 70, respectively.
  • an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense strand and an antisense strand comprising nucleotide sequences selected from: (a) SEQ ID NOs: 9 and 10, respectively; (b) SEQ ID NOs: 37 and 38, respectively; (c) SEQ ID NOs: 65 and 66, respectively; and (d) SEQ ID NOs: 69 and 70, respectively, wherein the sense and antisense strands are modified based on the pattern below Sense Strand: [mXs][mX][mX][mX][mX][mX][mX][mX][fX][fX][fX][mX][mX][mX][mX][mX][mX][mX][mX][mX][mX][mX][mX][mX][mX][mX][mX][mX][mX][mX][mX
  • an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense strand and an antisense strand comprising nucleotide sequences selected from: (a) SEQ ID NOs: 862 and 952, respectively; (b) SEQ ID NOs: 875 and 965, respectively; (c) SEQ ID NOs: 876 and 966, respectively; and (d) SEQ ID NOs: 920 and 1010, respectively, wherein the sense and antisense strands are modified based on the pattern below Sense Strand: [mXs][mX][mX][mX][mX][mX][mX][mX][fX][fX][fX][mX][mX][mX][mX][mX][mX][mX][mX][mX][mX][mX][mX][mX][mX][mX][mX][mX][mX][
  • an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense strand and an antisense strand comprising nucleotide sequences selected from: (a) SEQ ID NOs: 862 and 952, respectively; (b) SEQ ID NOs: 875 and 965, respectively; (c) SEQ ID NOs: 876 and 966, respectively; and (d) SEQ ID NOs: 920 and 1010, respectively, wherein the sense and antisense strands are modified based on the pattern below Sense Strand: [mXs][mX][mX][mX][mX][mX][mX][mX][fX][fX][fX][mX][mX][mX][mX][mX][mX][mX][mX][mX][mX][mX][mX][mX][mX][mX][mX][mX][mX][
  • an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense and antisense strand comprising nucleotide sequences selected from: (a) SEQ ID NOs: 11 and 12, respectively; (b) SEQ ID NOs: 39 and 40, respectively; (c) SEQ ID NOs: 67 and 68, respectively; and (d) SEQ ID NOs: 71 and 72, respectively.
  • an oligonucleotide for reducing expression of STAT3 mRNA comprises the sequence set forth in SEQ ID NO: 81.
  • an oligonucleotide for reducing expression of STAT3 mRNA comprises the sequence set forth in SEQ ID NO: 82. In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises the sequence set forth in SEQ ID NO: 83. In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises the sequence set forth in SEQ ID NO: 84. [00290] In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense and antisense strand having nucleotide sequences set forth in SEQ ID NOs: 87 and 68, respectively.
  • an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense and antisense strand having nucleotide sequences set forth in SEQ ID NOs: 88 and 71, respectively.
  • an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense strand sequence selected from SEQ ID NOs: 89-280.
  • an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense strand sequence selected from SEQ ID NOs: 857-946.
  • an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense strand sequence selected from SEQ ID NOs: 857-888. In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense strand sequence selected from SEQ ID NOs: 889-912. In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense strand sequence selected from SEQ ID NOs: 913-934. In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense strand sequence selected from SEQ ID NOs: 935-946.
  • an oligonucleotide for reducing expression of STAT3 mRNA comprises an antisense strand sequence selected from SEQ ID NOs: 947-1036. In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises an antisense strand sequence selected from SEQ ID NOs: 947-978. In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises an antisense strand sequence selected from SEQ ID NOs: 979-1002. In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises an antisense strand sequence selected from SEQ ID NOs: 1003-1024.
  • an oligonucleotide for reducing expression of STAT3 mRNA comprises an antisense strand sequence selected from SEQ ID NOs: 1025-1036.
  • an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense strand sequence selected from SEQ ID NOs: 857-946 and an antisense strand selected from SEQ ID NOs: 947-1036.
  • an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense strand sequence selected from SEQ ID NOs: 857-888 and an antisense strand selected from SEQ ID NOs: 947-978.
  • an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense strand sequence selected from SEQ ID NOs: 889-912 and an antisense strand selected from SEQ ID NOs: 979-1002.
  • an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense strand sequence selected from SEQ ID NOs: 913-934 and an antisense strand selected from SEQ ID NOs:1003-1024.
  • an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense strand sequence selected from SEQ ID NOs: 935-946 and an antisense strand selected from SEQ ID NOs:1025-1036.
  • an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense strand sequence selected from SEQ ID NOs: 1037-1126. In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense strand sequence selected from SEQ ID NOs: 1037-1068. In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense strand sequence selected from SEQ ID NOs:1069-1092. In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense strand sequence selected from SEQ ID NOs: 1093-1114.
  • an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense strand sequence selected from SEQ ID NOs:1115-1126. [00295] In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises an antisense strand sequence selected from SEQ ID NOs: 1127-1216. In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises an antisense strand sequence selected from SEQ ID NOs: 1127-1158. In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises an antisense strand sequence selected from SEQ ID NOs: 1159-1182.
  • an oligonucleotide for reducing expression of STAT3 mRNA comprises an antisense strand sequence selected from SEQ ID NOs:1183-1204. In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises an antisense strand sequence selected from SEQ ID NOs:1205-1216. [00296] In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense strand sequence selected from SEQ ID NOs: 1037-1126 and an antisense strand selected from SEQ ID NOs: 1127-1216.
  • an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense strand sequence selected from SEQ ID NOs: 1037-1068 and an antisense strand selected from SEQ ID NOs: 1127-1182. In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense strand sequence selected from SEQ ID NOs: 1069-1092 and an antisense strand selected from SEQ ID NOs: 1159-1182. In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense strand sequence selected from SEQ ID NOs: 1093-1114 and an antisense strand selected from SEQ ID NOs:1183-1204.
  • an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense strand sequence selected from SEQ ID NOs: 1115-1126 and an antisense strand selected from SEQ ID NOs:1205-1216.
  • an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense and antisense strand comprising nucleotide sequences selected from: (a) SEQ ID NOs: 857 and 947, respectively; (b) SEQ ID NOs: 858 and 948, respectively; (c) SEQ ID NOs: 859 and 949, respectively; (d) SEQ ID NOs: 860 and 950, respectively; (e) SEQ ID NOs: 862 and 952, respectively; (f) SEQ ID NOs: 867 and 957, respectively; (g) SEQ ID NOs: 875 and 965, respectively; and (h) SEQ ID NOs: 876 and 966, respectively.
  • an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense and antisense strand comprising nucleotide sequences selected from: (a) SEQ ID NOs: 901 and 991, respectively; (b) SEQ ID NOs: 910 and 1000, respectively; (c) SEQ ID NOs: 899 and 989, respectively; (d) SEQ ID NOs: 896 and 986, respectively; (e) SEQ ID NOs: 892 and 982, respectively; (f) SEQ ID NOs: 890 and 980, respectively; and (g) SEQ ID NOs: 889 and 979, respectively.
  • an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense and antisense strand comprising nucleotide sequences selected from: (a) SEQ ID NOs: 940 and 1030, respectively; (b) SEQ ID NOs: 937 and 1027, respectively; and (c) SEQ ID NOs: 939 and 1029, respectively.
  • an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense and antisense strand comprising nucleotide sequences selected from: (a) SEQ ID NOs: 915 and 1005, respectively; (b) SEQ ID NOs: 924 and 1014, respectively; (c) SEQ ID NOs: 913 and 1003, respectively; and (d) SEQ ID NOs: 920 and 1010, respectively.
  • an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense and antisense strand comprising nucleotide sequences selected from: (a) SEQ ID NOs: 862 and 952, respectively; (b) SEQ ID NOs: 875 and 965, respectively; (c) SEQ ID NOs: 876 and 966, respectively; and (d) SEQ ID NOs: 920 and 1010, respectively.
  • the sense strand comprises the sequence of SEQ ID NO: 862 and the antisense strand comprises the sequence of SEQ ID NO: 952.
  • the sense strand comprises the sequence of SEQ ID NO: 875 and the antisense strand comprises the sequence of SEQ ID NO: 965. [00304] In some embodiments, the sense strand comprises the sequence of SEQ ID NO: 876 and the antisense strand comprises the sequence of SEQ ID NO: 966. [00305] In some embodiments, the sense strand comprises the sequence of SEQ ID NO: 920 and the antisense strand comprises the sequence of SEQ ID NO: 1010.
  • an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense and antisense strand comprising nucleotide sequences selected from: (a) SEQ ID NOs: 1037 and 1127, respectively; (b) SEQ ID NOs: 1038 and 1128, respectively; (c) SEQ ID NOs: 1039 and 1129, respectively; (d) SEQ ID NOs: 1040 and 1130, respectively; (e) SEQ ID NOs: 1042 and 1132, respectively; (f) SEQ ID NOs: 1047 and 1137, respectively; (g) SEQ ID NOs: 1055 and 1145, respectively; and (h) SEQ ID NOs: 1056 and 1146, respectively.
  • an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense and antisense strand comprising nucleotide sequences selected from: (a) SEQ ID NOs: 1081 and 1171, respectively; (b) SEQ ID NOs: 1090 and 1180, respectively; (c) SEQ ID NOs: 1079 and 1169, respectively; (d) SEQ ID NOs: 1076 and 1166, respectively; (e) SEQ ID NOs: 1072 and 1162, respectively; (f) SEQ ID NOs: 1070 and 1160, respectively; and (g) SEQ ID NOs: 1069 and 1159, respectively.
  • an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense and antisense strand comprising nucleotide sequences selected from: (a) SEQ ID NOs: 1120 and 1210, respectively; (b) SEQ ID NOs: 1117 and 1207, respectively; and (c) SEQ ID NOs: 1119 and 1209, respectively.
  • an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense and antisense strand comprising nucleotide sequences selected from: (a) SEQ ID NOs: 1095 and 1185, respectively; (b) SEQ ID NOs: 1104 and 1194, respectively; (c) SEQ ID NOs: 1093 and 1183, respectively; and (d) SEQ ID NOs: 1100 and 1190, respectively.
  • an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense and antisense strand comprising nucleotide sequences selected from: (a) SEQ ID NOs: 1042 and 1132, respectively; (b) SEQ ID NOs: 1055 and 1145, respectively; (c) SEQ ID NOs: 1056 and 1146, respectively; and (d) SEQ ID NOs: 1100 and 1190, respectively.
  • the sense strand comprises the sequence of SEQ ID NO: 1042 and the antisense strand comprises the sequence of SEQ ID NO: 1132.
  • the sense strand comprises the sequence of SEQ ID NO: 1055 and the antisense strand comprises the sequence of SEQ ID NO: 1145. [00313] In some embodiments, the sense strand comprises the sequence of SEQ ID NO: 1056 and the antisense strand comprises the sequence of SEQ ID NO: 1146. [00314] In some embodiments, the sense strand comprises the sequence of SEQ ID NO: 1100 and the antisense strand comprises the sequence of SEQ ID NO: 1190.
  • an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense and antisense strand comprising nucleotide sequences selected from: (a) SEQ ID NOs: 1042 and 4, respectively; (b) SEQ ID NOs: 1055 and 5, respectively; (c) SEQ ID NOs: 1056 and 6, respectively; and (d) SEQ ID NOs: 1100 and 7, respectively.
  • an oligonucleotide for reducing expression of STAT3 mRNA described herein comprises minimal off-target effects.
  • an oligonucleotide described herein reduces STAT3 expression and does not reduce STAT1 expression or reduces STAT1 expression less than STAT3 expression.
  • the oligonucleotide comprises a sense strand comprising the nucleotide sequence set forth in SEQ ID NO: 862 and an antisense strand comprising the nucleotide sequence set forth in SEQ ID NO: 952, wherein the oligonucleotide reduces STAT3 expression and does not reduce STAT1 expression or reduces STAT1 expression less than STAT3 expression.
  • the oligonucleotide comprises a sense strand comprising the nucleotide sequence set forth in SEQ ID NO: 1042 and an antisense strand comprising the nucleotide sequence set forth in SEQ ID NO: 1132, wherein the oligonucleotide reduces STAT3 expression and does not reduce STAT1 expression or reduces STAT1 expression less than STAT3 expression.
  • the oligonucleotide comprises a sense strand comprising the nucleotide sequence set forth in SEQ ID NO: 875 and an antisense strand comprising the nucleotide sequence set forth in SEQ ID NO: 965, wherein the oligonucleotide reduces STAT3 expression and does not reduce STAT1 expression or reduces STAT1 expression less than STAT3 expression.
  • the oligonucleotide comprises a sense strand comprising the nucleotide sequence set forth in SEQ ID NO: 1055 and an antisense strand comprising the nucleotide sequence set forth in SEQ ID NO: 1145, wherein the oligonucleotide reduces STAT3 expression and does not reduce STAT1 expression or reduces STAT1 expression less than STAT3 expression.
  • an oligonucleotide for reducing expression of STAT3 mRNA described herein is a species cross-reactive oligonucleotide.
  • an oligonucleotide described herein is capable of reducing expression of STAT3 mRNA of at least two different species.
  • an oligonucleotide described herein is capable of reducing expression of STAT3 mRNA of at least two different species but does not cross-react with non-STAT3 mRNA (e.g., STAT1).
  • an oligonucleotide for reducing expression of STAT3 mRNA is cross-reactive between at least two species.
  • an oligonucleotide for reducing expression of STAT3 cross-reacts with human, non-human primate, and mouse STAT3 mRNA.
  • an oligonucleotide for reducing expression of STAT3 mRNA cross-reacts with human and mouse STAT3 mRNA.
  • an oligonucleotide for reducing expression of STAT3 mRNA cross-reacts with human and non-human primate STAT3 mRNA.
  • an oligonucleotide for reducing expression of STAT3 mRNA reduces STAT3 mRNA by at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, or at least 95%.
  • an oligonucleotide for reducing expression of STAT3 mRNA reduces STAT3 mRNA by at least 50% to at least 75% in human, non-human primate, and mouse (i.e. the oligonucleotide is a species cross-reactive oligonucleotide).
  • an oligonucleotide for reducing expression of STAT3 mRNA reduces STAT3 mRNA by at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, or at least 75% in human, non-human primate, and mouse (i.e. the oligonucleotide is a species cross-reactive oligonucleotide).
  • an oligonucleotide for reducing expression of STAT3 mRNA reduces STAT3 mRNA by at least 80%, at least 85%, at least 90%, or at least 95% in human, non-human primate, and mouse (i.e. the oligonucleotide is a species cross-reactive oligonucleotide).
  • an oligonucleotide for reducing expression of STAT3 mRNA reduces STAT3 mRNA by at least 50% to at least 75% in human and non-human primate (i.e. the oligonucleotide is a species cross-reactive oligonucleotide).
  • an oligonucleotide for reducing expression of STAT3 mRNA reduces STAT3 mRNA by at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, or at least 75% in human and non-human primate (i.e. the oligonucleotide is a species cross-reactive oligonucleotide).
  • an oligonucleotide for reducing expression of STAT3 mRNA reduces STAT3 mRNA by at least 80%, at least 85%, at least 90%, or at least 95% in human and non-human primate (i.e. the oligonucleotide is a species cross-reactive oligonucleotide).
  • an oligonucleotide for reducing expression of STAT3 mRNA reduces STAT3 mRNA by at least 50% to at least 75% in human and mouse (i.e. the oligonucleotide is a species cross-reactive oligonucleotide).
  • an oligonucleotide for reducing expression of STAT3 mRNA reduces STAT3 mRNA by at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, or at least 75% in human and mouse (i.e. the oligonucleotide is a species cross-reactive oligonucleotide).
  • an oligonucleotide for reducing expression of STAT3 mRNA reduces STAT3 mRNA by at least 80%, at least 85%, at least 90%, or at least 95% in human and mouse (i.e. the oligonucleotide is a species cross-reactive oligonucleotide).
  • an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense and antisense strand selected from: (a) SEQ ID NOs: 901 and 991, respectively; (b) SEQ ID NOs: 910 and 1000, respectively; (c) SEQ ID NOs: 899 and 989, respectively; (d) SEQ ID NOs: 896 and 986, respectively; (e) SEQ ID NOs: 892 and 982, respectively; (f) SEQ ID NOs: 890 and 980, respectively; and (g) SEQ ID NOs: 889 and 979, respectively, wherein the oligonucleotide reduces STAT3 mRNA in humans, non-human primates, and mice (i.e.
  • an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense and antisense strand selected from: (a) SEQ ID NOs: 940 and 1030, respectively; (b) SEQ ID NOs: 937 and 1027, respectively; and (c) SEQ ID NOs: 939 and 1029, respectively, wherein the oligonucleotide reduces STAT3 mRNA in humans, non-human primates, and mice (i.e. the oligonucleotide is a species cross-reactive oligonucleotide).
  • an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense and antisense strand selected from: (a) SEQ ID NOs: 915 and 1005, respectively; (b) SEQ ID NOs: 924 and 1014, respectively; (c) SEQ ID NOs: 913 and 1003, respectively; and (d) SEQ ID NOs: 920 and 1010, respectively, wherein the oligonucleotide reduces STAT3 mRNA in humans, non-human primates, and mice (i.e. the oligonucleotide is a species cross-reactive oligonucleotide).
  • an oligonucleotide for reducing expression of STAT3 mRNA comprises the sense strand sequence of SEQ ID NO: 862 and the antisense strand sequence of SEQ ID NO: 952, wherein the oligonucleotide reduces STAT3 mRNA in humans and non-human primates (i.e. the oligonucleotide is a species cross-reactive oligonucleotide).
  • an oligonucleotide for reducing expression of STAT3 mRNA comprises the sense strand sequence of SEQ ID NO: 875 and the antisense strand sequence of SEQ ID NO: 965, wherein the oligonucleotide reduces STAT3 mRNA in humans.
  • an oligonucleotide for reducing expression of STAT3 mRNA comprises the sense strand sequence of SEQ ID NO: 876 and the antisense strand sequence of SEQ ID NO: 966, wherein the oligonucleotide reduces STAT3 mRNA in humans.
  • an oligonucleotide for reducing expression of STAT3 mRNA comprises the sense strand sequence of SEQ ID NO: 920 and the antisense strand sequence of SEQ ID NO: 1010, wherein the oligonucleotide reduces STAT3 mRNA in humans.
  • an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense and antisense strand selected from: (a) SEQ ID NOs: 857 and 947, respectively; (b) SEQ ID NOs: 858 and 948, respectively; (c) SEQ ID NOs: 859 and 949, respectively; (d) SEQ ID NOs: 860 and 950, respectively; (e) SEQ ID NOs: 862 and 952, respectively; (f) SEQ ID NOs: 867 and 957, respectively; (g) SEQ ID NOs: 875 and 965, respectively; and (h) SEQ ID NOs: 876 and 966, respectively, wherein the oligonucleotide reduces STAT3 mRNA by at least 75%.
  • an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense and antisense strand selected from: (a) SEQ ID NOs: 901 and 991, respectively; (b) SEQ ID NOs: 910 and 1000, respectively; (c) SEQ ID NOs: 899 and 989, respectively; (d) SEQ ID NOs: 896 and 986, respectively; (e) SEQ ID NOs: 892 and 982, respectively; (f) SEQ ID NOs: 890 and 980, respectively; and (g) SEQ ID NOs: 889 and 979, respectively, wherein the oligonucleotide reduces STAT3 mRNA by at least 75%.
  • an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense and antisense strand selected from: (a) SEQ ID NOs: 940 and 1030, respectively; (b) SEQ ID NOs: 937 and 1027, respectively; and (c) SEQ ID NOs: 939 and 1029, respectively, wherein the oligonucleotide reduces STAT3 mRNA by at least 75%.
  • an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense and antisense strand selected from: (a) SEQ ID NOs: 915 and 1005, respectively; (b) SEQ ID NOs: 924 and 1014, respectively; (c) SEQ ID NOs: 913 and 1003, respectively; and (d) SEQ ID NOs: 920 and 1010, respectively, wherein the oligonucleotide reduces STAT3 mRNA by at least 75%.
  • an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense and antisense strand selected from: (a) SEQ ID NOs: 862 and 952, respectively; (b) SEQ ID NOs: 875 and 965, respectively; (c) SEQ ID NOs: 876 and 966, respectively; and (d) SEQ ID NOs: 920 and 1010, respectively, wherein the oligonucleotide reduces STAT3 mRNA by at least 75%.
  • an oligonucleotide for reducing expression of STAT3 mRNA comprises the sense strand sequence of SEQ ID NO: 862 and the antisense strand sequence of SEQ ID NO: 952, wherein the oligonucleotide reduces STAT3 mRNA by at least 75%.
  • an oligonucleotide for reducing expression of STAT3 mRNA comprises the sense strand sequence of SEQ ID NO: 875 and the antisense strand sequence of SEQ ID NO: 965, wherein the oligonucleotide reduces STAT3 mRNA by at least [00335] In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises the sense strand sequence of SEQ ID NO: 876 and the antisense strand sequence of SEQ ID NO: 966, wherein the oligonucleotide reduces STAT3 mRNA by at least 75%.
  • an oligonucleotide for reducing expression of STAT3 mRNA comprises the sense strand sequence of SEQ ID NO: 920 and the antisense strand sequence of SEQ ID NO: 1010, wherein the oligonucleotide reduces STAT3 mRNA by at least 75%.
  • an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense and antisense strand selected from: (a) SEQ ID NOs: 857 and 947, respectively; (b) SEQ ID NOs: 858 and 948, respectively; (c) SEQ ID NOs: 859 and 949, respectively; (d) SEQ ID NOs: 860 and 950, respectively; (e) SEQ ID NOs: 862 and 952, respectively; (f) SEQ ID NOs: 867 and 957, respectively; (g) SEQ ID NOs: 875 and 965, respectively; and (h) SEQ ID NOs: 876 and 966, respectively, wherein the oligonucleotide is conjugated to a lipid.
  • an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense and antisense strand selected from: (a) SEQ ID NOs: 901 and 991, respectively; (b) SEQ ID NOs: 910 and 1000, respectively; (c) SEQ ID NOs: 899 and 989, respectively; (d) SEQ ID NOs: 896 and 986, respectively; (e) SEQ ID NOs: 892 and 982, respectively; (f) SEQ ID NOs: 890 and 980, respectively; and (g) SEQ ID NOs: 889 and 979, respectively, wherein the oligonucleotide is conjugated to a lipid on the sense strand.
  • an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense and antisense strand selected from: (a) SEQ ID NOs: 940 and 1030, respectively; (b) SEQ ID NOs: 937 and 1027, respectively; and (c) SEQ ID NOs: 939 and 1029, respectively, wherein the oligonucleotide is conjugated to a lipid on the sense strand.
  • an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense and antisense strand selected from: (a) SEQ ID NOs: 915 and 1005, respectively; (b) SEQ ID NOs: 924 and 1014, respectively; (c) SEQ ID NOs: 913 and 1003, respectively; and (d) SEQ ID NOs: 920 and 1010, respectively, wherein the oligonucleotide is conjugated to a lipid on the sense strand.
  • an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense and antisense strand selected from: (a) SEQ ID NOs: 862 and 952, respectively; (b) SEQ ID NOs: 875 and 965, respectively; (c) SEQ ID NOs: 876 and 966, respectively; and (d) SEQ ID NOs: 920 and 1010, respectively, wherein the oligonucleotide is conjugated to a lipid on the sense strand.
  • an oligonucleotide for reducing expression of STAT3 mRNA comprises the sense strand sequence of SEQ ID NO: 862 and the antisense strand sequence of SEQ ID NO: 952, wherein the oligonucleotide is conjugated to a lipid on the sense strand.
  • an oligonucleotide for reducing expression of STAT3 mRNA comprises the sense strand sequence of SEQ ID NO: 875 and the antisense strand sequence of SEQ ID NO: 965, wherein the oligonucleotide is conjugated to a lipid on the sense strand.
  • an oligonucleotide for reducing expression of STAT3 mRNA comprises the sense strand sequence of SEQ ID NO: 876 and the antisense strand sequence of SEQ ID NO: 966, wherein the oligonucleotide is conjugated to a lipid on the sense strand.
  • an oligonucleotide for reducing expression of STAT3 mRNA comprises the sense strand sequence of SEQ ID NO: 920 and the antisense strand sequence of SEQ ID NO: 1010, wherein the oligonucleotide is conjugated to a lipid on the sense strand.
  • an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense and antisense strand selected from: (a) SEQ ID NOs: 857 and 947, respectively; (b) SEQ ID NOs: 858 and 948, respectively; (c) SEQ ID NOs: 859 and 949, respectively; (d) SEQ ID NOs: 860 and 950, respectively; (e) SEQ ID NOs: 862 and 952, respectively; (f) SEQ ID NOs: 867 and 957, respectively; (g) SEQ ID NOs: 875 and 965, respectively; and (h) SEQ ID NOs: 876 and 966, respectively, wherein the oligonucleotide is conjugated to a C18 lipid on the sense strand.
  • an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense and antisense strand selected from: (a) SEQ ID NOs: 901 and 991, respectively; (b) SEQ ID NOs: 910 and 1000, respectively; (c) SEQ ID NOs: 899 and 989, respectively; (d) SEQ ID NOs: 896 and 986, respectively; (e) SEQ ID NOs: 892 and 982, respectively; (f) SEQ ID NOs: 890 and 980, respectively; and (g) SEQ ID NOs: 889 and 979, respectively, wherein the oligonucleotide is conjugated to a C18 lipid on the sense strand.
  • an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense and antisense strand selected from: (a) SEQ ID NOs: 940 and 1030, respectively; (b) SEQ ID NOs: 937 and 1027, respectively; and (c) SEQ ID NOs: 939 and 1029, respectively, wherein the oligonucleotide is conjugated to a C18 lipid on the sense strand.
  • an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense and antisense strand selected from: (a) SEQ ID NOs: 915 and 1005, respectively; (b) SEQ ID NOs: 924 and 1014, respectively; (c) SEQ ID NOs: 913 and 1003, respectively; and (d) SEQ ID NOs: 920 and 1010, respectively, wherein the oligonucleotide is conjugated to a C18 lipid on the sense strand.
  • an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense and antisense strand selected from: (a) SEQ ID NOs: 862 and 952, respectively; (b) SEQ ID NOs: 875 and 965, respectively; (c) SEQ ID NOs: 876 and 966, respectively; and (d) SEQ ID NOs: 920 and 1010, respectively, wherein the oligonucleotide is conjugated to a C18 lipid on the sense strand.
  • an oligonucleotide for reducing expression of STAT3 mRNA comprises the sense strand sequence of SEQ ID NO: 862 and the antisense strand sequence of SEQ ID NO: 952, wherein the oligonucleotide is conjugated to a C18 lipid on the sense strand.
  • an oligonucleotide for reducing expression of STAT3 mRNA comprises the sense strand sequence of SEQ ID NO: 875 and the antisense strand sequence of SEQ ID NO: 965, wherein the oligonucleotide is conjugated to a C18 lipid on the sense strand.
  • an oligonucleotide for reducing expression of STAT3 mRNA comprises the sense strand sequence of SEQ ID NO: 876 and the antisense strand sequence of SEQ ID NO: 966, wherein the oligonucleotide is conjugated to a C18 lipid on the sense strand.
  • an oligonucleotide for reducing expression of STAT3 mRNA comprises the sense strand sequence of SEQ ID NO: 920 and the antisense strand sequence of SEQ ID NO: 1010, wherein the oligonucleotide is conjugated to a C18 lipid on the sense strand.
  • an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense and antisense strand selected from: (a) SEQ ID NOs: 901 and 991, respectively; (b) SEQ ID NOs: 910 and 1000, respectively; (c) SEQ ID NOs: 899 and 989, respectively; (d) SEQ ID NOs: 896 and 986, respectively; (e) SEQ ID NOs: 892 and 982, respectively; (f) SEQ ID NOs: 890 and 980, respectively; and (g) SEQ ID NOs: 889 and 979, respectively, wherein the oligonucleotide reduces STAT3 mRNA in humans, non-human primates, and mice (i.e.
  • an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense and antisense strand selected from: (a) SEQ ID NOs: 940 and 1030, respectively; (b) SEQ ID NOs: 937 and 1027, respectively; and (c) SEQ ID NOs: 939 and 1029, respectively, wherein the oligonucleotide reduces STAT3 mRNA in humans and mice (i.e. the oligonucleotide is a species cross-reactive oligonucleotide) by at least 75%.
  • an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense and antisense strand selected from: (a) SEQ ID NOs: 915 and 1005, respectively; (b) SEQ ID NOs: 924 and 1014, respectively; (c) SEQ ID NOs: 913 and 1003, respectively; and (d) SEQ ID NOs: 920 and 1010, respectively, wherein the oligonucleotide reduces STAT3 mRNA in humans by at least 75%.
  • an oligonucleotide for reducing expression of STAT3 mRNA comprises the sense strand sequence of SEQ ID NO: 862 and the antisense strand sequence of SEQ ID NO: 952, wherein the oligonucleotide reduces STAT3 mRNA in humans and non-human primates (i.e. the oligonucleotide is a species cross-reactive oligonucleotide) by at least 75%.
  • an oligonucleotide for reducing expression of STAT3 mRNA comprises the sense strand sequence of SEQ ID NO: 875 and the antisense strand sequence of SEQ ID NO: 965, wherein the oligonucleotide reduces STAT3 mRNA in humans by at least 75%.
  • an oligonucleotide for reducing expression of STAT3 mRNA comprises the sense strand sequence of SEQ ID NO: 876 and the antisense strand sequence of SEQ ID NO: 966, wherein the oligonucleotide reduces STAT3 mRNA in humans by at least 75%.
  • an oligonucleotide for reducing expression of STAT3 mRNA comprises the sense strand sequence of SEQ ID NO: 920 and the antisense strand sequence of SEQ ID NO: 1010, wherein the oligonucleotide reduces STAT3 mRNA in humans by at least 75%.
  • an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense and antisense strand selected from: (a) SEQ ID NOs: 901 and 991, respectively; (b) SEQ ID NOs: 910 and 1000, respectively; (c) SEQ ID NOs: 899 and 989, respectively; (d) SEQ ID NOs: 896 and 986, respectively; (e) SEQ ID NOs: 892 and 982, respectively; (f) SEQ ID NOs: 890 and 980, respectively; and (g) SEQ ID NOs: 889 and 979, respectively, wherein the oligonucleotide is conjugated to a lipid on the sense strand and reduces STAT3 mRNA in humans, non-human primates, and mice (i.e.
  • an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense and antisense strand selected from: (a) SEQ ID NOs: 940 and 1030, respectively; (b) SEQ ID NOs: 937 and 1027, respectively; and (c) SEQ ID NOs: 939 and 1029, respectively, wherein the oligonucleotide is conjugated to a lipid on the sense strand and reduces STAT3 mRNA in humans and mice (i.e. the oligonucleotide is a species cross-reactive oligonucleotide).
  • an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense and antisense strand selected from: (a) SEQ ID NOs: 915 and 1005, respectively; (b) SEQ ID NOs: 924 and 1014, respectively; (c) SEQ ID NOs: 913 and 1003, respectively; and (d) SEQ ID NOs: 920 and 1010, respectively, wherein the oligonucleotide is conjugated to a lipid on the sense strand and reduces STAT3 mRNA in humans.
  • an oligonucleotide for reducing expression of STAT3 mRNA comprises the sense strand sequence of SEQ ID NO: 862 and the antisense strand sequence of SEQ ID NO: 952, wherein the oligonucleotide is conjugated to a lipid on the sense strand and reduces STAT3 mRNA in humans and non-human primates (i.e. the oligonucleotide is a species cross-reactive oligonucleotide).
  • an oligonucleotide for reducing expression of STAT3 mRNA comprises the sense strand sequence of SEQ ID NO: 875 and the antisense strand sequence of SEQ ID NO: 965, wherein the oligonucleotide is conjugated to a lipid on the sense strand and reduces STAT3 mRNA in humans.
  • an oligonucleotide for reducing expression of STAT3 mRNA comprises the sense strand sequence of SEQ ID NO: 876 and the antisense strand sequence of SEQ ID NO: 966, wherein the oligonucleotide is conjugated to a lipid on the sense strand and reduces STAT3 mRNA in humans.
  • an oligonucleotide for reducing expression of STAT3 mRNA comprises the sense strand sequence of SEQ ID NO: 920 and the antisense strand sequence of SEQ ID NO: 1010, wherein the oligonucleotide is conjugated to a lipid on the sense strand and reduces STAT3 mRNA in humans.
  • an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense and antisense strand selected from: (a) SEQ ID NOs: 901 and 991, respectively; (b) SEQ ID NOs: 910 and 1000, respectively; (c) SEQ ID NOs: 899 and 989, respectively; (d) SEQ ID NOs: 896 and 986, respectively; (e) SEQ ID NOs: 892 and 982, respectively; (f) SEQ ID NOs: 890 and 980, respectively; and (g) SEQ ID NOs: 889 and 979, respectively, wherein the oligonucleotide is conjugated to a C18 lipid on the sense strand and reduces STAT3 mRNA in humans, non-human primates, and mice (i.e.
  • an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense and antisense strand selected from: (a) SEQ ID NOs: 940 and 1030, respectively; (b) SEQ ID NOs: 937 and 1027, respectively; and (c) SEQ ID NOs: 939 and 1029, respectively, wherein the oligonucleotide is conjugated to a C18 lipid on the sense strand and reduces STAT3 mRNA in humans and mice (i.e. the oligonucleotide is a species cross-reactive oligonucleotide).
  • an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense and antisense strand selected from: (a) SEQ ID NOs: 915 and 1005, respectively; (b) SEQ ID NOs: 924 and 1014, respectively; (c) SEQ ID NOs: 913 and 1003, respectively; and (d) SEQ ID NOs: 920 and 1010, respectively, wherein the oligonucleotide is conjugated to a C18 lipid on the sense strand and reduces STAT3 mRNA in humans.
  • an oligonucleotide for reducing expression of STAT3 mRNA comprises the sense strand sequence of SEQ ID NO: 862 and the antisense strand sequence of SEQ ID NO: 952, wherein the oligonucleotide is conjugated to a C18 lipid on the sense strand and reduces STAT3 mRNA in humans and non-human primates (i.e. the oligonucleotide is a species cross-reactive oligonucleotide).
  • an oligonucleotide for reducing expression of STAT3 mRNA comprises the sense strand sequence of SEQ ID NO: 875 and the antisense strand sequence of SEQ ID NO: 965, wherein the oligonucleotide is conjugated to a C18 on the sense strand lipid and reduces STAT3 mRNA in humans.
  • an oligonucleotide for reducing expression of STAT3 mRNA comprises the sense strand sequence of SEQ ID NO: 876 and the antisense strand sequence of SEQ ID NO: 966, wherein the oligonucleotide is conjugated to a C18 lipid on the sense strand and reduces STAT3 mRNA in humans.
  • an oligonucleotide for reducing expression of STAT3 mRNA comprises the sense strand sequence of SEQ ID NO: 920 and the antisense strand sequence of SEQ ID NO: 1010, wherein the oligonucleotide is conjugated to a C18 lipid on the sense strand and reduces STAT3 mRNA in humans.
  • an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense and antisense strand selected from: (a) SEQ ID NOs: 901 and 991, respectively; (b) SEQ ID NOs: 910 and 1000, respectively; (c) SEQ ID NOs: 899 and 989, respectively; (d) SEQ ID NOs: 896 and 986, respectively; (e) SEQ ID NOs: 892 and 982, respectively; (f) SEQ ID NOs: 890 and 980, respectively; and (g) SEQ ID NOs: 889 and 979, respectively, wherein the oligonucleotide is conjugated to a lipid on the sense strand and reduces STAT3 mRNA in humans, non-human primates, and mice (i.e.
  • an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense and antisense strand selected from: (a) SEQ ID NOs: 940 and 1030, respectively; (b) SEQ ID NOs: 937 and 1027, respectively; and (c) SEQ ID NOs: 939 and 1029, respectively, wherein the oligonucleotide is conjugated to a lipid on the sense strand and reduces STAT3 mRNA in humans and mice (i.e.
  • an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense and antisense strand selected from: (a) SEQ ID NOs: 915 and 1005, respectively; (b) SEQ ID NOs: 924 and 1014, respectively; (c) SEQ ID NOs: 913 and 1003, respectively; and (d) SEQ ID NOs: 920 and 1010, respectively, wherein the oligonucleotide is conjugated to a lipid on the sense strand and reduces STAT3 mRNA in humans by at least 75%.
  • an oligonucleotide for reducing expression of STAT3 mRNA comprises the sense strand sequence of SEQ ID NO: 862 and the antisense strand sequence of SEQ ID NO: 952, wherein the oligonucleotide is conjugated to a lipid on the sense strand and reduces STAT3 mRNA in humans and non-human primates (i.e. the oligonucleotide is a species cross-reactive oligonucleotide) by at least 75%.
  • an oligonucleotide for reducing expression of STAT3 mRNA comprises the sense strand sequence of SEQ ID NO: 875 and the antisense strand sequence of SEQ ID NO: 965, wherein the oligonucleotide is conjugated to a lipid on the sense strand and reduces STAT3 mRNA in humans by at least 75%.
  • an oligonucleotide for reducing expression of STAT3 mRNA comprises the sense strand sequence of SEQ ID NO: 876 and the antisense strand sequence of SEQ ID NO: 966, wherein the oligonucleotide is conjugated to a lipid on the sense strand and reduces STAT3 mRNA in humans by at least 75%.
  • an oligonucleotide for reducing expression of STAT3 mRNA comprises the sense strand sequence of SEQ ID NO: 920 and the antisense strand sequence of SEQ ID NO: 1010, wherein the oligonucleotide is conjugated to a lipid on the sense strand and reduces STAT3 mRNA in humans by at least 75%.
  • an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense and antisense strand selected from: (a) SEQ ID NOs: 901 and 991, respectively; (b) SEQ ID NOs: 910 and 1000, respectively; (c) SEQ ID NOs: 899 and 989, respectively; (d) SEQ ID NOs: 896 and 986, respectively; (e) SEQ ID NOs: 892 and 982, respectively; (f) SEQ ID NOs: 890 and 980, respectively; and (g) SEQ ID NOs: 889 and 979, respectively, wherein the oligonucleotide is conjugated to a C18 lipid on the sense strand and reduces STAT3 mRNA in humans, non-human primates, and mice (i.e.
  • an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense and antisense strand selected from: (a) SEQ ID NOs: 940 and 1030, respectively; (b) SEQ ID NOs: 937 and 1027, respectively; and (c) SEQ ID NOs: 939 and 1029, respectively, wherein the oligonucleotide is conjugated to a C18 lipid on the sense strand and reduces STAT3 mRNA in humans and mice (i.e.
  • an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense and antisense strand selected from: (a) SEQ ID NOs: 915 and 1005, respectively; (b) SEQ ID NOs: 924 and 1014, respectively; (c) SEQ ID NOs: 913 and 1003, respectively; and (d) SEQ ID NOs: 920 and 1010, respectively, wherein the oligonucleotide is conjugated to a C18 lipid on the sense strand and reduces STAT3 mRNA in humans by at least 75%.
  • an oligonucleotide for reducing expression of STAT3 mRNA comprises the sense strand sequence of SEQ ID NO: 862 and the antisense strand sequence of SEQ ID NO: 952, wherein the oligonucleotide is conjugated to a C18 lipid on the sense strand and reduces STAT3 mRNA in humans and non-human primates (i.e. the oligonucleotide is a species cross-reactive oligonucleotide) by at least 75%.
  • an oligonucleotide for reducing expression of STAT3 mRNA comprises the sense strand sequence of SEQ ID NO: 875 and the antisense strand sequence of SEQ ID NO: 965, wherein the oligonucleotide is conjugated to a C18 lipid on the sense strand and reduces STAT3 mRNA in humans by at least 75%.
  • an oligonucleotide for reducing expression of STAT3 mRNA comprises the sense strand sequence of SEQ ID NO: 876 and the antisense strand sequence of SEQ ID NO: 966, wherein the oligonucleotide is conjugated to a C18 lipid on the sense strand and reduces STAT3 mRNA in humans by at least 75%.
  • an oligonucleotide for reducing expression of STAT3 mRNA comprises the sense strand sequence of SEQ ID NO: 920 and the antisense strand sequence of SEQ ID NO: 1010, wherein the oligonucleotide is conjugated to a C18 lipid on the sense strand and reduces STAT3 mRNA in humans by at least 75%.
  • Formulations [00390] Various formulations have been developed to facilitate oligonucleotide use. For example, oligonucleotides can be delivered to a subject or a cellular environment using a formulation that minimizes degradation, facilitates delivery and/or uptake, or provides another beneficial property to the oligonucleotides in the formulation.
  • an oligonucleotide is formulated in buffer solutions such as phosphate buffered saline solutions, liposomes, micellar structures, and capsids.
  • buffer solutions such as phosphate buffered saline solutions, liposomes, micellar structures, and capsids.
  • cationic lipids such as lipofectin, cationic glycerol derivatives, and polycationic molecules (e.g., polylysine, can be used.
  • Suitable lipids include Oligofectamine, Lipofectamine (Life Technologies), NC388 (Ribozyme Pharmaceuticals, Inc., Boulder, Colo.), or FuGene 6 (Roche) all of which can be used according to the manufacturer′s instructions.
  • a formulation comprises a lipid nanoparticle.
  • an excipient comprises a liposome, a lipid, a lipid complex, a microsphere, a microparticle, a nanosphere or a nanoparticle, or may be otherwise formulated for administration to the cells, tissues, organs, or body of a subject in need thereof (see, e.g., Remington: THE SCIENCE AND PRACTICE OF PHARMACY, 22nd edition, Pharmaceutical Press, 2013).
  • the formulations herein comprise an excipient.
  • an excipient confers to a composition improved stability, improved absorption, improved solubility and/or therapeutic enhancement of the active ingredient.
  • an excipient is a buffering agent (e.g., sodium citrate, sodium phosphate, a tris base, or sodium hydroxide) or a vehicle (e.g., a buffered solution, petrolatum, dimethyl sulfoxide, or mineral oil).
  • a buffering agent e.g., sodium citrate, sodium phosphate, a tris base, or sodium hydroxide
  • a vehicle e.g., a buffered solution, petrolatum, dimethyl sulfoxide, or mineral oil.
  • an oligonucleotide is lyophilized for extending its shelf-life and then made into a solution before use (e.g., administration to a subject).
  • an excipient in a composition comprising any one of the oligonucleotides described herein may be a lyoprotectant (e.g., mannitol, lactose, polyethylene glycol or polyvinylpyrrolidone) or a collapse temperature modifier (e.g., dextran, FicollTM or gelatin).
  • a pharmaceutical composition is formulated to be compatible with its intended route of administration. Examples of routes of administration include parenteral (e.g., intravenous, intramuscular, intraperitoneal, intradermal, subcutaneous), oral (e.g., inhalation), transdermal (e.g., topical), transmucosal and rectal administration.
  • compositions suitable for injectable use include sterile aqueous solutions (where water soluble) or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersion.
  • suitable carriers include physiological saline, bacteriostatic water, Cremophor ELTM (BASF, Parsippany, N.J.) or phosphate buffered saline (PBS).
  • the carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (e.g., glycerol, propylene glycol, and liquid polyethylene glycol, and the like), and suitable mixtures thereof.
  • a composition may contain at least about 0.1% of the therapeutic agent or more, although the percentage of the active ingredient(s) may be between about 1% to about 80% or more of the weight or volume of the total composition.
  • PD-L1 Programmed Death Ligand 1
  • the disclosure provides a PD-L1 inhibitor for use in combination with an oligonucleotide described herein.
  • a PD-L1 inhibitor is a small molecule, a peptide, a protein, an antibody, or nucleic acid molecule such as an siRNA, miRNA, or an antisense RNA. In some embodiments, the PD-L1 inhibitor inhibits association of PD-L1 and PD-1. In some embodiments, the PD-L1 inhibitor is specific for PD-L1. [00399] In some embodiments, the PD-L1 inhibitor is an anti-PD-L1 antibody. In some embodiments, the PD-L1 inhibitor is specific for PD-1. In some embodiments, the PD-L1 inhibitor is an anti-PD-1 antibody. In some embodiments, the antibody is a full-length antibody.
  • the antibody is an antibody fragment.
  • the PD- L1 inhibitor is a small molecule.
  • the anti-PD-L1 antibody is atezolizumab.
  • Atezolizumab MPDL3280A
  • the anti-PD-L1 antibody is avelumab.
  • Avelumab (MSB0010718C) is a fully humanized, engineered IgG1 monoclonal antibody to PD-L1.
  • the anti-PD-L1 antibody is envafolimab.
  • the anti- PD-L1 antibody is durvalumab.
  • Duvalumab (MEDI4736) is a human monoclonal antibody to PD-L1.
  • the anti-PD-L1 antibody is TSR-042.
  • TSR-042 refers to an engineered chimeric antibody directed against the PD-1/PD-L1 pathway.
  • the anti-PD-L1 antibody is KD-033.
  • KD-033 refers to a bifunctional anti-PD- L1/IL-15 fusion protein, wherein the anti-PD-L1 antibody binds to the cytokine IL-15 by the sushi domain of the IL-15 receptor connected at the tail.
  • the anti-PD- L1 antibody is STI-1014.
  • STI-1014 refers to an anti-PD-L1 antibody.
  • the anti-PD-L1 antibody is KY-1003.
  • KY-1003 is a monoclonal antibody against PD-L1.
  • the anti-PD-L1 antibody is YW243.55.S70.
  • Antibody YW243.55.S70 is an anti-PDL1 described in U.S. Patent No.9,920,123, the contents of which are incorporated herein in their entirety.
  • the anti-PD-L1 antibody is MDX-1106.
  • MDX- 1106, also known as MDX-1106-04, ONO-4538 or BMS-936558 is an anti-PD 1 antibody described in U.S.
  • the anti-PD-L1 antibody is Merck 3745.
  • Merck 3745 also known as MK-3475 or SCH-900475, is an anti-PD1 antibody described in U.S. Patent No. 8,168,757, the contents of which are incorporated herein in their entirety.
  • the anti-PD-L1 antibody is CT-011.
  • CT-011, also known as hBAT or hBAT-1 is an anti-PD1 antibody described in U.S. Patent No.8,747,847, the contents of which are incorporated herein in their entirety.
  • the anti-PD-L1 antibody is AMP- 224.
  • the anti-PD-L1 antibody is any anti-PD-L1 antibody known in the art, including, but not limited to, the anti-PD-L1 antibodies disclosed in Akinleye & Rasool “Immune checkpoint inhibitors of PD-L1 as cancer therapeutics” J. of Hematology & Oncology.12(92): 2019.
  • the anti-PD-L1 antibody is BMS-936559.
  • BMS-936559 (MDX-1105) is a fully human IgG4 monoclonal antibody against PD-L1 and is described in U.S. Patent No.7,943,743, the contents of which are incorporated herein in their entirety.
  • the anti-PD-L1 antibody is CK- 301.
  • the anti-PD-L1 antibody is CS-1001.
  • the anti-PD-L1 antibody is SHR-1316.
  • the anti-PD-L1 antibody is BG- A333.
  • the anti-PD-L1 antibody is an antibody disclosed in International Pub. No. WO 2021/231741, the contents of which are incorporated herein in their entirety.
  • the anti-PD-1 antibody is nivolumab.
  • Nivolumab (BMS-936558) is an IgG4 monoclonal antibody described in U.S. Patent No.8,008,449, the contents of which are incorporated herein in their entirety.
  • the anti- PD-1 antibody is pembrolizumab.
  • Pembrolizumab is an IgG4 monoclonal antibody described in U.S. Patent No.8,354,509, the contents of which are incorporated herein in their entirety.
  • the anti-PD-1 antibody is cemiplimab.
  • Cemiplimab (REGN2810) is an IgG4 monoclonal antibody described in U.S.
  • the anti-PD-L1 antibody described herein binds to PD-L1 with an affinity of about 30nM to about 100nM. In some embodiments, the anti-PD-L1 antibody described herein binds to PD-L1 with an affinity of about 30nM. In some embodiments, the anti-PD-L1 antibody described herein binds to PD-L1 with an affinity of about 40nM. In some embodiments, the anti-PD-L1 antibody described herein binds to PD-L1 with an affinity of about 50nM.
  • the anti-PD-L1 antibody described herein binds to PD-L1 with an affinity of about 60nM. In some embodiments, the anti-PD-L1 antibody described herein binds to PD-L1 with an affinity of about 70nM. In some embodiments, the anti-PD-L1 antibody described herein binds to PD-L1 with an affinity of about 80nM. In some embodiments, the anti-PD-L1 antibody described herein binds to PD-L1 with an affinity of about 90nM. In some embodiments, the anti-PD-L1 antibody described herein binds to PD-L1 with an affinity of about 100nM.
  • the anti-PD-1 antibody described herein binds to PD-1 with an affinity of about 30nM to about 100nM. In some embodiments, the anti-PD-1 antibody described herein binds to PD-1 with an affinity of about 30nM. In some embodiments, the anti- PD-1 antibody described herein binds to PD-1 with an affinity of about 40nM. In some embodiments, the anti-PD-1 antibody described herein binds to PD-1 with an affinity of about 50nM. In some embodiments, the anti-PD-1 antibody described herein binds to PD-1 with an affinity of about 60nM. In some embodiments, the anti-PD-1 antibody described herein binds to PD-1 with an affinity of about 70nM.
  • the anti-PD-1 antibody described herein binds to PD-1 with an affinity of about 80nM. In some embodiments, the anti- PD-1 antibody described herein binds to PD-1 with an affinity of about 90nM. In some embodiments, the anti-PD-1 antibody described herein binds to PD-1 with an affinity of about 100nM. [00405] In some embodiments, the antibody is generated using display technologies. Display technologies used to generate antibody polypeptides include any of the display techniques (e.g. display library screening techniques). In some embodiments, synthetic antibodies are designed, selected, or optimized by screening target antigens using display technologies (e.g. phage display technologies).
  • Phage display libraries may comprise millions to billions of phage vectors, each expressing unique antibody fragments on their viral coats. Such libraries may provide richly diverse resources that are used to select potentially hundreds of antibody fragments with diverse levels of affinity for one or more antigens of interest (McCafferty, et al., 1990. Nature.348:552-4; Edwards, B.M. et al., 2003. JMB.334: 103-18; Schofield, D. et al., 2007. Genome Biol.8, R254 and Pershad, K. et al., 2010. Protein Engineering Design and Selection.23:279-88; the contents of each of which are herein incorporated by reference in their entirety).
  • the antibody fragments present in such libraries comprise scFv antibody fragments, comprising a fusion protein of V H and VL antibody domains joined by a flexible linker.
  • scFvs may contain the same sequence with the exception of unique sequences encoding variable loops of the CDRs.
  • scFvs are expressed as fusion proteins, linked to viral coat proteins (e.g. the N- terminus of the viral pill coat protein). VL chains may be expressed separately for assembly with VH chains in the periplasm prior to complex incorporation into viral coats. Precipitated library members may be sequenced from the bound phage to obtain cDNA encoding desired scFvs.
  • Antibody variable domains or CDRs from such sequences may be directly incorporated into antibody sequences for recombinant antibody production or mutated and utilized for further optimization through in vitro affinity maturation.
  • the sequences of the polypeptides to be encoded in the viral genomes are produced using yeast surface display technology.
  • recombinant antibodies are developed by displaying the antibody fragment of interest as a fusion to on the surface of the yeast, where the protein interacts with proteins and small molecules in a solution.
  • scFvs with affinity toward desired receptors may be isolated from the yeast surface using magnetic separation and flow cytometry. Several cycles of yeast surface display and isolation may be done to attain scFvs with desired properties through directed evolution.
  • An exemplary method for determining binding affinity employs surface plasmon resonance.
  • Surface plasmon resonance is an optical phenomenon that allows for the analysis of realtime biospecific interactions by detection of alterations in protein concentrations within a biosensor matrix, for example using the BIAcore system (Pharmacia Biosensor AB, Uppsala, Sweden and Piscataway, N.J.).
  • BIAcore Phharmacia Biosensor AB, Uppsala, Sweden and Piscataway, N.J.
  • the PD-L1 inhibitor is an siRNA molecule.
  • the PD-L1 inhibitor is ALN-PDL.
  • the PD-L1 inhibitor is a PD-L1 inhibitor described in U.S. Patent No.10,889,813, U.S. Patent No.10,745,704, U.S. Pub. No.2021/0277403, U.S. Pub. No.2006/0276422A1, U.S.2021/0277403, International Pub. No. WO2019/000149, or International Pub.
  • the PD-L1 inhibitor is an siRNA molecule described in Barati, M., et al. A review of PD-1/PD-L1 siRNA delivery systems in immune T cells and cancer cells, International Immunopharmacology, 2022, vol.111.
  • the siRNA molecule for inhibition of PD-L1 is an siRNA molecule generated using methods known to those of skill in the art.
  • the PD-L1 inhibitor is an antisense oligonucleotide.
  • the PD-L1 inhibitor is an antisense oligonucleotide described in U.S.
  • the antisense oligonucleotide for inhibition of PD-L1 is an antisense oligonucleotide generated using methods known to those of skill in the art.
  • the PD-L1 inhibitor is a small molecule.
  • the small molecule is CA-170.
  • CA-170 is a small molecule antagonist of PD- L1 and VISTA.
  • the small molecule is CA-327.
  • CA-327 refers to small molecule antagonists of PD-L1 and TIM3.
  • the small molecule is BMS-1001. In some embodiments, the small molecule is BMS-1166. In some embodiments, the small molecule is BMS-8. In some embodiments, the small molecule is BMS-37. In some embodiments, the small molecule is BMS-202. In some embodiments, the small molecule is BMS-200. In some embodiments, the small molecule is Incyte-001. In some embodiments, the small molecule is Incyte-011. In some embodiments, the small molecule is INCB086550. In some embodiments, the small molecule is LH1306. In some embodiments, the small molecule is LH1307.
  • the small molecule is ARB-272572.
  • the PD-L1 inhibitor is a small molecule described in Sasikumar, P. et al. Small Molecule Agents Targeting PD-1 Checkpoint Pathway for Cancer Immunotherapy: Mechanisms of Action and Other Considerations for Their Advanced Development, Front. Immunol.2022, 13:752065.
  • the PD-L1 inhibitor is a small molecule described in U.S. Patent No.11,130,740, U.S. Patent No.10,590,105, International Pub. No. WO2019/076343, U.S. Patent No.11,555,029, or U.S. Pub.
  • the small molecule is a small molecule described in Wu, Q. et al. Small molecule inhibitors targeting the PD- 1/PD-L1 signaling pathway, Acta Pharmacologica Sinica.2021, 42: 1-9.
  • the small molecule is a small molecule described in Wang, Y. et al. A Small Molecule Antagonist of PD-1/PD-L1 Interactions Acts as an Immune Checkpoint Inhibitor for NSCLC and Melanoma Immunotherapy, Front. Immuno.2021. Vol.12.
  • the small molecule inhibitor is a stereoisomer, a tautomer, a pharmaceutically acceptable salt, a hydrate, or a solvent of a small molecule described herein.
  • the small molecule for inhibition of PD-L1 is a small molecule generated using methods known to those of skill in the art.
  • the PD-L1 inhibitor is a peptide.
  • the PD-L1 inhibitor is a peptide inhibitor described in U.S. Patent No.9,422,339 or U.S. Patent No.9,850,283, the contents of which are incorporated herein in their entirety.
  • the PD-L1 inhibitor is a peptide inhibitor described in Yin, H. et al. Rational Design of Potent Peptide Inhibitors of the PD-1:PD-L1 Interaction for Cancer Immunotherapy, J. Am. Chem. Soc.2021, 143: 44, 18536-18547.
  • the PD-L1 inhibitor is a peptide inhibitor described in Lin, X., et al. Progress in PD-1/PD-L1 pathway inhibitors: From biomacromolecules to small molecules, Euro. J. Med. Chem.2020, vol.186.
  • the inhibitor peptide for inhibition of PD-L1 is a peptide generated using methods known to those of skill in the art.
  • the person skilled in the art knows how to determine whether a compound is a PD1 and/or PDL1 inhibitor by testing it in an appropriate assay. Binding of inhibitors to PD1 and/or PDL1 and/or PDL2 can e.g., be measured in ELISA-type assays that are well known in the art. Bioassays to measure the biological effect of PD1 and/or PDL1 and/or PDL2 inhibition are well known by the person skilled in the art.
  • kits comprising a STAT3 oligonucleotide herein, and instructions for administering the STAT3 oligonucleotide to a subject that has received or is receiving a PD-L1 inhibitor.
  • the kit comprises, in a suitable container, an oligonucleotide herein, one or more controls, and various buffers, reagents, enzymes and other standard ingredients well known in the art.
  • the container comprises at least one vial, well, test tube, flask, bottle, syringe, or other container means, into which the oligonucleotide is placed, and in some instances, suitably aliquoted.
  • the kit contains additional containers into which this component is placed.
  • the kits can also include a means for containing the oligonucleotide and any other reagent in close confinement for commercial sale.
  • Such containers may include injection or blow-molded plastic containers into which the desired vials are retained.
  • Containers and/or kits can include labeling with instructions for use and/or warnings.
  • a kit comprises a STAT3 oligonucleotide herein, and a pharmaceutically acceptable carrier, or a pharmaceutical composition comprising the oligonucleotide and instructions for treating or delaying progression of a disease, disorder or condition associated with STAT3 expression in a subject in need thereof, wherein the subject has received or is receiving a PD-L1 inhibitor.
  • a kit comprises a STAT3 oligonucleotide herein, and a pharmaceutically acceptable carrier, or a pharmaceutical composition comprising the oligonucleotide and instructions for treating or delaying progression of a cancer in a subject in need thereof, wherein the subject has received or is receiving a PD-L1 inhibitor.
  • a kit comprises a PD-L1 inhibitor, and a pharmaceutically acceptable carrier, or a pharmaceutical composition comprising the oligonucleotide and instructions for treating or delaying progression of a disease, disorder or condition in a subject in need thereof, wherein the subject has received or is receiving a STAT3 oligonucleotide described herein.
  • a kit comprises a PD-L1 inhibitor, and a pharmaceutically acceptable carrier, or a pharmaceutical composition comprising the oligonucleotide and instructions for treating or delaying progression of a cancer in a subject in need thereof, wherein the subject has received or is receiving a STAT3 oligonucleotide described herein.
  • nucleic acid or analogues thereof of the present disclosure are either commercially available or can be produced by organic synthesis methods known to one of ordinary skill in the art (METHODS OF ORGANIC SYNTHESIS, Thieme, Volume 21 (Houben-Weyl 4th Ed.1952)). Further, the nucleic acid or analogues thereof of the present disclosure can be produced by organic synthesis methods known to one of ordinary skill in the art as shown in the following examples. [00419] All reactions are carried out under nitrogen or argon unless otherwise stated.
  • Example ⁇ 1a ⁇ Synthesis ⁇ of ⁇ 2 ⁇ (2 ⁇ ((((6aR,8R,9R,9aR) ⁇ 8 ⁇ (6 ⁇ benzamido ⁇ 9H ⁇ purin ⁇ 9 ⁇ yl) ⁇ 2,2,4,4 ⁇ tetraisopropyltetrahydro ⁇ 6H ⁇ furo[3,2 ⁇ f][1,3,5,2,4]trioxadisilocin ⁇ 9 ⁇ yl)oxy)methoxy)ethoxy) ⁇ ethan ⁇ 1 ⁇ ammonium ⁇ formate ⁇ (1 ⁇ 6) [00422] A solution of compound 1-1 (25.00 g, 67.38 mmol) in 20 mL of DMF was treated with pyridine (11 mL, 134.67 mmol) and tetraisopropyldisiloxane dichloride (22.63 mL, 70.75 mmol) at 10 °C.
  • Example 1b Synthesis of (2R,3R,4R,5R) ⁇ 5 ⁇ (6 ⁇ benzamido ⁇ 9H ⁇ purin ⁇ 9 ⁇ yl) ⁇ 2 ⁇ ((bis(4 ⁇ methoxyphenyl)(phenyl)methoxy)methyl) ⁇ 4 ⁇ ((2 ⁇ (2 ⁇ [lipid] ⁇ amidoethoxy)ethoxy)methoxy) tetrahydrofuran ⁇ 3 ⁇ yl (2 ⁇ cyanoethyl) ⁇ diisopropylphosphoramidite(2 ⁇ 4a ⁇ to ⁇ 2 ⁇ 4e) ⁇
  • R1COOH group represents fatty acid C8:0, C10:0, C11:0, C12:0, C14:0, C16:0, C17:0, C18:0, C18:1, C18:2, C22:5, C22:0, C24:0, C26:0, C22:6, C24:1, diacyl C16:0 or diacyl C18:1
  • Synthesis Sense 1 and Antisense 1 were prepared by solid-phase synthesis. Synthesis of Conjugated Sense 1a-1i. [00435] Conjugated Sense 1a was synthesized through post-syntenic conjugation approach.
  • Eppendorf tube 1 a solution of octanoic acid (0.58 mg, 4 umol) in DMA (0.75 mL) was treated with HATU (1.52 mg, 4 umol) at rt.
  • Eppendorf tube 2 a solution of oligo Sense 1 (10.00 mg, 0.8 umol) in H 2 O (0.25 mL) was treated with DIPEA (1.39 uL, 8 umol). The solution in Eppendorf tube 1 was added to the Eppendorf tube 2 and mixed using Thermomixer at rt.
  • reaction mixture was diluted with 5 mL of water and purified by revers phase XBridge C18 column using a 5-95% gradient of 100 mM TEAA in ACN and H 2 O.
  • the product fractions were concentrated under reduced pressure using Genevac.
  • the combined residual solvent was dialyzed against water (1 X), saline (1 X), and water (3 X) using Amicon® Ultra-15 Centrifugal (3K).
  • the Amicon membrane was washed with water (3 X 2 mL) and the combined solvents were then lyophilized to afford an amorphous white solid of Conjugated Sense 1a (6.43 mg, 64% yield).
  • Conjugated Sense 1b-1i were prepared using similar procedures as described for the synthesis of Conjugated Sense 1a and obtained in 42%-69% yields. Annealing of Duplex 1a-1j. [00437] Conjugated Sense 1a (10 mg, measured by weight) was dissolved in 0.5 mL deionized water to prepare a 20 mg/mL solution. Antisense 1 (10 mg, measured by OD) was dissolved in 0.5 mL deionized water to prepare a 20 mg/mL solution, which was used for the titration of the conjugated sense and quantification of the duplex amount.
  • Duplex 1b-1i were prepared using the same procedures as described for the annealing of Duplex 1a (C8).
  • the following Scheme 1-2 depicts the synthesis of Nicked tetraloop GalXC conjugates with mono-lipid on the loop. Post-synthetic conjugation was realized through Cu- catalyzed alkyne-azide cycloaddition reaction.
  • Sense 1B and Antisense 1B were prepared by solid-phase synthesis. Synthesis of Conjugated Sense 1j. [00441] In Eppendorf tube 1, a solution of oligo (10.00 mg, 0.8 umol) in a 3:1 mixture of DMA/ H 2 O (0.5 mL) was treated with the lipid linker azide (11.26 mg, 4 umol). In Eppendorf tube 2, CuBr dimethyl sulfide (1.64 mg, 8 umol) was dissolved in ACN (0.5 mL). Both solutions were degassed for 10 min by bubbling N 2 through them. The ACN solution of CuBrSMe2 was then added into tube 1 and the resulting mixture was stirred at 40 °C.
  • reaction mixture was diluted with 0.5 M EDTA (2 mL) and dialyzed against water (2 X) using a Amicon® Ultra-15 Centrifugal (3K).
  • the reaction crude was purified by revers phase XBridge C18 column using a 5-95% gradient of 100 mM TEAA in ACN (with 30% IPA spiked in) and H 2 O.
  • the product fractions were concentrated under reduced pressure using Genevac.
  • the combined residual solvent was dialyzed against water (1 X), saline (1 X), and water (3 X) using Amicon® Ultra-15 Centrifugal (3K).
  • Duplex 1j (PEG2K-diacyl C18) was prepared using the same procedures as described for the annealing of Duplex 1a (C8).
  • Scheme 1-3 depicts the synthesis of Nicked tetraloop GalXC conjugates with di-lipid on the loop using post-synthetic conjugation approach.
  • Scheme1-3 Sense 2 and Antisense 2 were prepared by solid-phase synthesis.
  • Conjugated Sense 2a and 2b were prepared using similar procedures as described for the synthesis of Conjugated Sense 1a but with 10 eq of lipid, 10 eq of HATU, and 20 eq of DIPEA.
  • Duplex 2a (2XC11) and 2b (2XC22) were prepared using the same procedures as described for the annealing of Duplex 1a (C8).
  • the following Scheme 1-4 depicts the synthesis of GalXC of fully phosphorothioated stem-loop conjugated with mono-lipid using post-synthetic conjugation approach.
  • Scheme1-4 Sense 3 and Antisense 3 were prepared by solid-phase synthesis.
  • Conjugated Sense 3a was prepared using similar procedures as described for the synthesis of Conjugated Sense 1a and obtained in a 65% yield.
  • Duplex 3a (PS-C22) was prepared using the same procedures as described for the annealing of Duplex 1a (C8).
  • the following Scheme1-5 depicts the synthesis of GalXC of short sense conjugated with mono-lipid using post-synthetic conjugation approach.
  • Scheme 1-5 Sense 4 and Antisense 4 were prepared by solid-phase synthesis.
  • Conjugated Sense 4a was prepared using similar procedures as described for the synthesis of Conjugated Sense 1a and obtained in a 74% yield.
  • Duplex 4a (SS-C22) was prepared using the same procedures as described for the annealing of Duplex 1a (C8).
  • the following Scheme 1-6 depicts the synthesis of Nicked tetraloop GalXC conjugated with tri-adamantane moiety on the loop using post-synthetic conjugation approach.
  • Sense 5 and Antisense 5 were prepared by solid-phase synthesis.
  • Conjugated Sense 5a and 5b were prepared using similar procedures as described for the synthesis of Conjugated Sense 1a and obtained in 42%-73% yields.
  • Duplex 5a (3Xadamantane) and Duplex 5b (3Xacetyladamantane) were prepared using the same procedures as described for the annealing of Duplex 1a (C8).
  • the following scheme 1-7 depicts an example of solid phase synthesis of Nicked tetraloop GalXC conjugated with lipid(s) on the loop.
  • Conjugated Sense 6 was prepared by solid-phase synthesis using a commercial oligo synthesizer. The oligonucleotides were synthesized using 2’-modified nucleoside phosphoramidites, such as 2’-F or 2’-OMe, and 2'-diethoxymethanol linked fatty acid amide nucleoside phosphoramidites. Oligonucleotide synthesis was conducted on a solid support in the 3’ to 5’direction using a standard oligonucleotide synthesis protocol. In these efforts, 5-ethylthio-1H-tetrazole (ETT) was used as an activator for the coupling reaction.
  • ETT 5-ethylthio-1H-tetrazole
  • Duplex 6 was prepared using the same procedures as described for the annealing of Duplex 1a (C8). Scheme ⁇ 8.
  • Scheme1-10 depicts the synthesis of GalXC of short sense and short stem loop conjugated with mono-lipid using post-synthetic conjugation approach.
  • Scheme 1-10 Synthesis of Sense 9a [00464] Conjugated Sense 9a was obtained using the same method or a substantially similar method to the synthesis of Conjugated Sense 5. Synthesis example of Duplex 9a [00465] Duplex 9a was obtained using the same method or a substantially similar method to the synthesis of Duplex 5. [00466] The following Scheme1-11 depicts the synthesis of GalXC conjugated with mono-lipid at 5’-end using post-synthetic conjugation approach.
  • a passenger strand with 2’-amine linkers [ademA] was used for post solid phase conjugation.
  • Different types of lipids were conjugated using the same chemistry to generate a series of conjugates (FIG.1A and 1B).
  • SAR studies were performed to identify a lipid conjugate that could be used to deliver payloads to the tissues of interest in order to mediate target knockdown.
  • Example 3 Tissue Specific Targets in MDSC Cell Populations and Tumor Draining Lymph Nodes.
  • STAT3 is involved in immune suppression with examples abundantly reported in literature. Targeting STAT3 transcription through an RNAi mechanism could potentially overcome the challenges in the development of pharmacological STAT3 inhibitors.
  • STAT3 was selected as a proof-of-concept target to demonstrate tissue specific activity in the tissues of interest, such as myeloid derived suppressor cells (MDSCs).
  • STAT3 sequences were designed in the GalXC format with described modification patterns and screening for target knockdown in liver tissue was performed in normal CD-1 mice.
  • Eighteen STAT3-GalXC conjugates (Table 1) were dosed once subcutaneously at 3 mg/kg.
  • Table 1 GalXC Compound Candidates for Identifying Tool Compounds for Proof-of- concept Studies in Mice:
  • livers were collected and subjected to mRNA analysis by qPCR.
  • four sequences (GalXC -STAT3-838, GalXC- STAT3-1402, GalXC-STAT3-4110 and GalXC-STAT3-4123) that showed >85% target knockdown in liver were selected for further evaluation (FIG.2A).
  • FOG.2A four sequences that showed >85% target knockdown in liver were selected for further evaluation.
  • three were identified as mouse specific and one was identified as human-mouse cross-reactive.
  • These 4 sequences were further screened in CD-1 mice at 3 different doses (0.3, 1 and 3 mg/kg) to assess the dose response.
  • GalXC-STAT3-4110 and 4123 were identified as the most potent sequences after the dose response screen, each with ED50 of 0.3 mg/kg and thus these molecules were selected for further studies (FIG.2B).
  • C18 lipid conjugation was performed for both GalXC-STAT3-4110 or 4123 for proof-of-concept studies (Table 2).
  • Table 2 GalXC-STAT3 Lipid Conjugates
  • mice were implanted in nude mice and upon reaching sufficient tumor volume mice were subjected to randomization as previously described. Mice received either a single dose of GalXC- STAT3-C184110 and 4123 subcutaneously at 25 mg/kg, 50 mg/kg, or PBS. At 3 days post injection, bulk tumors were collected and MDSC subsets were isolated.
  • MDSCs are characterized by the co-expression of cell surface or mRNA markers CD11b (a marker for the myeloid cells of the macrophage lineage) and Gr-1(a marker for the myeloid lineage differentiation antigen) and denoted as CD11b + Gr-1 + cells.
  • Gr-1 is further comprised of 2 components Ly6G and Ly6C.
  • MDSCs consist of two subsets: Granulocytic MDSC (G- MDSC), further characterized as CD11b + Ly6G + Ly6C lo , and monocytic MDSC (M-MDSC) characterized as CD11b + Ly6G-Ly6C hi .
  • G- MDSC Granulocytic MDSC
  • M-MDSC monocytic MDSC
  • CD11b positive cells in the single cell suspension were then magnetically labeled with MACS microbeads and enriched by passing through MACS columns and subsequently eluting the retained labeled cells in the column as positively selected fractions (CD11b MicroBeads UltraPure, mouse kit Cat# 130- 126-725).
  • non-target cells in the cell suspension were magnetically labeled with a cocktail of microbeads and passed through the MACS columns. During this process, the unwanted labeled cells were retained in the column and the unlabeled target cells (tumor cells) were collected in the flow-through as pure fraction. (Tumor Cell Isolation Kit, human Cat # 130-108-339).
  • Pan02 tumor bearing mice were treated with a single subcutaneous dose of either GalXC-STAT3-C18-4123 at 50 mg/kg, or PBS and Stat3 mRNA levels were measured after 3 days.
  • the Stat3 knockdown in G-MDSC was not significantly altered as compared to the knockdown observed at the 25 mg/kg dose, however there was a significant improvement in Stat3 silencing observed in M- MDSC subset at this same dose level.
  • Stat3 knockdown was assessed in bulk tumors and TdLNs on day 7 (FIGs.4A and 4B). Dose dependent Stat3 mRNA knockdown was observed in bulk tumor with both GalXC- STAT3-C18 sequences.
  • Example 4 STAT3 Inhibition Decreases the PD-L1 Levels in MDSCs and Mediates Acute Tumor Effects [00479]
  • the transcriptional signature of phosphorylated STAT3 has been positively correlated with PD-L1 expression in tumors (Song et al, JOURNAL OF CELL PHYSIOLOGY (2020), Zerdes et al, CANCERS (2019), Song et al, BLOOD (2018).
  • isolated populations of MDSCs treated with either PBS or a GalXC-STAT3 conjugate were assayed for Pdl1 mRNA.
  • Pdl1 mRNA levels were decreased by ⁇ 80% in both G-MDSC and M-MDSC populations treated with either 25 or 50 mg/kg of a GalXC-STAT3 (FIG.5A).
  • the Pdl1 levels were also dramatically reduced in TdLN after treatment with the GalXC-STAT3 conjugate, specifically GalXC-STAT3-C18- 4123 (FIG.5B).
  • Pan02 murine pancreatic syngeneic model
  • GalXC-STAT3- C18 conjugate following a split dosing model where all animals received a total dose of 50 mg/kg, dosed as either 25 mg/kg x 2 doses or 12.5 mg/kg x 4 doses.
  • Tumors treated using the 25 mg/kg split dose showed acute tumor regression, even after the first dose (FIG.6B).
  • the second dose of 25 mg/kg tumors from 3 out of 4 mice regressed to sizes that were too small to be collected for further processing.
  • dsRNAi oligonucleotides were synthesized using solid phase oligonucleotide synthesis methods as described for 19-23mer siRNAs (see, e.g., Scaringe et al. (1990) Nucleic Acids Res.18:5433-5441 and Usman et al. (1987) J. Am. Chem.
  • dsRNAi oligonucleotides having a 19mer core sequence were formatted into constructs having a 25mer sense strand and a 27mer antisense strand to allow for processing by the RNAi machinery.
  • the 19mer core sequence is complementary to a region in the STAT3 mRNA.
  • RNA oligonucleotides were synthesized using solid phase phosphoramidite chemistry, deprotected and desalted on NAP-5 columns (Amersham Pharmacia Biotech; Piscataway, NJ) using standard techniques (Damha & Olgivie (1993) Methods Mol. Biol.20:81-114; Wincott et al. (1995) Nucleic Acids Res.23:2677-2684).
  • the oligomers were purified using ion-exchange high performance liquid chromatography (IE-HPLC) on an Amersham Source 15Q column (1.0 cm ⁇ 25 cm; Amersham Pharmacia Biotech) using a 15 min step-linear gradient.
  • the gradient varied from 90:10 Buffers A:B to 52:48 Buffers A:B, where Buffer A is 100 mM Tris pH 8.5 and Buffer B is 100 mM Tris pH 8.5, 1 M NaCl.
  • Samples were monitored at 260 nm and peaks corresponding to the full-length oligonucleotide species were collected, pooled, desalted on NAP-5 columns, and lyophilized. [00483]
  • the purity of each oligomer was determined by capillary electrophoresis (CE) on a Beckman PACE 5000 (Beckman Coulter, Inc.; Fullerton, CA).
  • the CE capillaries have a 100 ⁇ m inner diameter and contain ssDNA 100R Gel (Beckman-Coulter). Typically, about 0.6 nmole of oligonucleotide was injected into a capillary, run in an electric field of 444 V/cm and was detected by UV absorbance at 260 nm. Denaturing Tris-Borate-7 M-urea running buffer was purchased from Beckman-Coulter. Oligoribonucleotides were obtained that were at least 90% pure as assessed by CE for use in experiments described below.
  • RNA oligomers were resuspended (e.g., at 100 ⁇ M concentration) in duplex buffer consisting of 100 mM potassium acetate, 30 mM HEPES, pH 7.5.
  • RNA buffer IDT
  • dsRNA oligonucleotides were stored at ⁇ 20° C.
  • Single strand RNA oligomers were stored lyophilized or in nuclease-free water at ⁇ 80° C.
  • Example 6 Generation of STAT3-Targeting Double-Stranded RNAi Oligonucleotides Identification of STAT3 ⁇ mRNA Target Sequences
  • STAT3 Signal transducer and activator of transcription 3
  • a computer-based algorithm was used to computationally identify STAT3 mRNA target sequences suitable for assaying inhibition of STAT3 expression by the RNAi pathway.
  • RNAi oligonucleotide guide (antisense) strand sequences each having a region of complementarity to a suitable STAT3 target sequence of human STAT3 mRNA (e.g., SEQ ID NO:1217; Table 4).
  • STAT3 RNAi oligonucleotides comprising a region of complementarity to homologous STAT3 mRNA target sequences with nucleotide sequence similarity are predicted to have the ability to target homologous STAT3 mRNAs.
  • RNAi oligonucleotides (formatted as DsiRNA oligonucleotides) were generated as described in Example 5 for evaluation in vitro. Each DsiRNA was generated with the same modification pattern, and each with a unique guide strand having a region of complementarity to a STAT3 target sequence identified by SEQ ID NOs: 89-280.
  • human hepatocyte (Huh7) cells expressing endogenous human STAT3 gene were transfected with each of the DsiRNAs listed in Table 5 at 1 nM in separate wells of a multi-well cell-culture plate. Cells were maintained for 24 hours following transfection with the modified DsiRNA, and then the amount of remaining STAT3 mRNA from the transfected cells was determined using TAQMAN®-based qPCR assays.
  • Two qPCR assays a 3′ assay and 5’ assay (Forward 1- SEQ ID NO:1219), Reverse 1- SEQ ID NO:1220, Probe 1- SEQ ID NO: 1221; Forward 2- SEQ ID NO: 1, Reverse 2- SEQ ID NO: 2, Probe 2- SEQ ID NO: 3) were used to determine STAT3 mRNA levels as measured using PCR probes conjugated to 6-carboxy-fluorescein (FAM). Each primer pair was assayed for % remaining RNA as shown in Table 5 and FIG. 7. DsiRNAs resulting in less than or equal to 10% STAT3 mRNA remaining in DsiRNA- transfected cells when compared to mock-transfected cells were considered DsiRNA “hits”.
  • FAM 6-carboxy-fluorescein
  • the Huh7 cell-based assay evaluating the ability of the DsiRNAs listed in Table 5 to inhibit STAT3 expression identified several candidate DsiRNAs. [00488] Taken together, these results show that DsiRNAs designed to target human STAT3 mRNA inhibit STAT3 expression in cells, as determined by a reduced amount of STAT3 mRNA in DsiRNA-transfected cells relative to control cells. These results demonstrate that the nucleotide sequences comprising the DsiRNA are useful for generating RNAi oligonucleotides to inhibit STAT3 expression. Further, these results demonstrate that multiple STAT3 mRNA target sequences are suitable for the RNAi-mediated inhibition of STAT3 expression. Table 5. Analysis of STAT3 mRNA in Huh7 cells
  • RNAi oligonucleotides To confirm the ability of the RNAi oligonucleotides to knockdown STAT3 in vivo, an HDI mouse model was used. A subset of the DsiRNAs identified in Example 6 were used to generate corresponding double-stranded RNAi oligonucleotides comprising a nicked tetraloop GalNAc-conjugated structure (referred to herein as “GalNAc-conjugated STAT3 oligonucleotides” or “GalNAc- STAT3 oligonucleotides”) having a 36-mer passenger strand and a 22-mer guide strand (Table 8 and Table 9).
  • GalNAc-conjugated STAT3 oligonucleotides referred to herein as “GalNAc-conjugated STAT3 oligonucleotides” or “GalNAc- STAT3 oligonucleotides” having a 36-mer passenger strand
  • nucleotide sequences comprising the passenger strand and guide strand have a distinct pattern of modified nucleotides and phosphorothioate linkages.
  • Three of the nucleotides comprising the tetraloop were each conjugated to a GalNAc moiety (CAS#14131-60-3).
  • the modification patterns used are illustrated below: Pattern 1 Sense Strand: 5’ mX-S-mX-mX-mX-mX-mX-mX-mX-mX-mX-mX-mX-fX-fX[-mX-]16-[ademX-GalNAc]- [ademX-GalNAc]-[ademX-GalNAc]-mX-mX-mX-mX-mX-mX 3’.
  • HDI hydrodynamically injected
  • CMV ubiquitous cytomegalovirus
  • RNA derived from these HDI mice were subjected to qRT-PCR analysis to determine STAT3 mRNA levels as described in Example 6. mRNA levels were measured for human mRNA. The values were normalized for transfection efficiency using the NeoR gene included on the DNA plasmid.
  • a benchmark control (STAT3-1388) comprising a different modification pattern, was used for both assays (Sense Strand SEQ ID NO: 1100; Antisense Strand SEQ ID NO: 1190).
  • STAT3-1388 comprising a different modification pattern
  • FIGs.9A and 9B demonstrate that GalNAc-conjugated STAT3 oligonucleotides designed to target human STAT3 mRNA inhibited human STAT3 mRNA expression in HDI mice, as determined by a reduction in the amount of human STAT3 mRNA expression in liver samples from HDI mice treated with GalNAc-conjugated STAT3 oligonucleotides relative to control HDI mice treated with only PBS.
  • FIGs.9A and 9B demonstrate that GalNAc-conjugated STAT3 oligonucleotides designed to target human STAT3 mRNA inhibited human STAT3 mRNA expression in HDI mice, as determined by a reduction in the amount of human STAT3 mRNA expression in liver samples from HDI mice treated with GalNAc-conjugated STAT3 oligonucleotides relative to control HDI mice treated with only PBS.
  • 9A and 9B were further validated in a dosing study. Specifically, dosing studies were carried out using nine GalNAc-conjugated STAT3 oligonucleotides (STAT3-715, STAT3-716, STAT3-717, STAT3-720, STAT3-721, STAT3-1145, STAT3- 1286, STAT3-1286, and STAT3-1287). Mice were hydrodynamically injected as described above and treated with 0.1mg/kg, 0.3mg/kg, or 1mg/kg of oligonucleotide. Livers were collected after one day, and STAT3 expression was measured to determine a potent dose (FIG.10).
  • RNAi oligonucleotides were able to reduce STAT3 expression at a 1mg/kg dose and STAT3-1286 was able to reduce expression at a 0.3mg/kg dose.
  • the HDI studies identified several potential GalNAc-conjugated STAT3 oligonucleotides for inhibiting STAT3 expression in liver.
  • Example 8 Species Specific RNAi Oligonucleotide Inhibition of STAT3 In Vivo [00494] To confirm the ability of RNAi oligonucleotides to knockdown STAT3 in vivo, several cross species and species specific GalNAc-conjugated STAT3 oligonucleotides were generated.
  • Hs/Mf/Mm triple common (targeting human, non-human primate, and mouse; Hs/Mf/Mm), human/mouse (Hs/Mm), and human specific (Hs) oligonucleotides were evaluated.
  • Hs/Mf/Mm and Hs/Mm Commons [00495] Mice expressing endogenous mouse STAT3 in the liver were subcutaneously injected at a dose of 3mg/kg with the GalNAc-conjugated STAT3 oligonucleotides set forth in Table 10. Livers were collected after five days, and STAT3 expression was measured.
  • a subset of the GalNAc-conjugated STAT3 oligonucleotides tested in FIGs. 11 and 12 were further validated in a dosing study. Specifically, dosing studies were carried out using ten GalNAc-conjugated STAT3 oligonucleotides (STAT3-2626, STAT3-2627, STAT3-2408, STAT3-2412, STAT3-2139, STAT3-4909, STAT3- 461, STAT3-678, STAT3- 2148, and STAT3-2144). Mice endogenously expressing mouse STAT3 were subcutaneously injected with 0.3mg/kg, 1mg/kg, or 3mg/kg oligonucleotide.
  • mice were administered only PBS. Three days later (72 hours), the mice were hydrodynamically injected (HDI) with a DNA plasmid encoding the full human STAT3 gene (25 ⁇ g) under control of a ubiquitous cytomegalovirus (CMV) promoter sequence. One day after introduction of the DNA plasmid, liver samples from HDI mice were collected. Total RNA derived from these HDI mice were subjected to qRT-PCR analysis to determine STAT3 mRNA levels. Table 12. GalNAc-Conjugated Human STAT3 RNAi Oligonucleotides for Exogenous STAT3 Screen.
  • HDI hydrodynamically injected
  • CMV ubiquitous cytomegalovirus
  • FIG.14 demonstrate that GalNAc-conjugated STAT3 oligonucleotides designed to target human STAT3 mRNA inhibited human STAT3 mRNA expression in HDI mice, as determined by a reduction in the amount of human STAT3 mRNA expression in liver samples from HDI mice treated with GalNAc-conjugated STAT3 oligonucleotides relative to control HDI mice treated with only PBS.
  • a subset of the GalNAc-conjugated STAT3 oligonucleotides tested in FIG.14 were further validated in a dosing study.
  • mice were hydrodynamically injected as described above and treated with 0.3mg/kg, 1 mg/kg, or 3mg/kg of oligonucleotide. Livers were collected after one day, and human STAT3 expression was measured to determine a potent dose (FIG. 15). A dose of 1mg/kg was capable of reducing STAT3 mRNA by about 75%, thereby identifying several potential GalNAc-conjugated STAT3 oligonucleotides for inhibiting STAT3 expression in liver.
  • Example 9 Specific STAT3 Inhibition by GalNAc-Conjugated STAT3 Oligonucleotides [00501] The specificity of the GalNAc-conjugated STAT3 oligonucleotides to inhibit STAT3 rather than a family member (e.g. STAT1) was measured.
  • Huh7 cells expressing endogenous STAT1 were treated for 24 hours with 0.05nM, 0.3nM, or 1nM of a GalNAc-conjugated STAT3 oligonucleotide (STAT3-721, STAT3-1286, and STAT3-1388) using lipofectamine as transfection agent.
  • the percent (%) remaining mRNA was measured compared to a mock control (PBS; no lipofectamine or siRNA) and UTR (un-transfected; treated with lipofectamine but no siRNA) (Table 13 and FIG.17).
  • STAT3721 and 1286 did not downregulate human STAT1 but STAT31388 did (Table 13).
  • mice received either a single dose of GalXC- STAT3-C18-4123 subcutaneously at 25 mg/kg as single agent or in combination with an anti- PD-L1 mAb (anti-mouse PD-L1 mAb (B7-H1), Clone 10F.9G2) at 10 mg/kg (i.p.). Mice were first administered two doses three days apart, and two weeks later were administered two more doses three days apart [(q3dx2)x2]. Control groups were treated with either GalXC- Placebo as single agent or in combination with the anti-PD-L1 mAb as described for the GalXC-STAT3-C18-4123 compound. Two weeks after the last dose, the same dose regimen was repeated.
  • Tumor sizes were measured twice a week throughout the study period.
  • the tumors that received GalXC-Placebo or GalXC- Placebo + mAb treatments continued to grow to the same extent.
  • the group that received GalXC-STAT3 demonstrated anti-tumor efficacy after the first round of treatment, but they continued to grow despite receiving a second dose.
  • the group that received a combination of GalXC-STAT3 and mAb demonstrated significantly more tumor regression as compared to the single agent treatment. This demonstrates that combination therapy with a checkpoint inhibitor can achieve improved anti-tumor efficacy.
  • Example 11 Correlation Between Treatment With a Combination of GalXC-STAT3 and PD-L1 mAb With Tumor Immune Phenotypes
  • tumor types with different phenotypes were selected for implantation in mice.
  • Selected tumor types included Pan02 (FIG.18A, checkpoint resistant tumors), 4T1 (triple negative breast, checkpoint resistant tumors), MC-38 (Colon Carcinoma, partially checkpoint sensitive tumors) and Hepa1-6 (Hepatocellular Carcinoma, checkpoint sensitive tumors).
  • Pan02 (5e6 cells + matrigel, FIG.18A) MC-38 (5e6 cells) and Hepa1-6 tumors (2e6 cells) were grown in C57BL/6 mice (7-8 weeks old) and 4T1 tumors (7-8 weeks old) were grown in Balb/c mice.
  • Example 5.4T1 tumors were treated three times with each dose three days apart (q3dx3), with a combination of subcutaneous GalXC- STAT3-C18-4123 with an anti-PD-L1 mAb or single agents GalXC-Placebo, GalXC- STAT3-C18-4123, or GalXC-Placebo with the mAb, as shown in FIG.19A.
  • Tumor volumes were measured twice a week throughout the study period.
  • MC-38 and Hepa1-6 tumors were treated with a combination of subcutaneous GalXC-STAT3-C18-4123 with an anti-PD-L1 mAb or single agents GalXC-Placebo, GalXC-STAT3-C18-4123, or GalXC-Placebo with the mAb (2 doses at 3 days apart for 2 weeks) as shown in FIGs.19B and 19C.
  • Combination treatment demonstrated synergistic efficacy in the resistant tumor types where the tumors expected to have very little or no CD8+ T cell infiltration in the TME and a larger population of MDSCs (CD8 low MDSC high ) (FIGs.18A and 19A).
  • the combination treatment showed improved efficacy compared to checkpoint alone treatment in partially sensitive tumors where the tumors had slightly higher levels of CD8+ T-cell infiltration and modest levels of MDSCs (CD8 med MDSC med / high ) (FIG.19B). Interestingly, the combination treatment led to complete regression of the sensitive tumors (CD8 high MDSC high ) (FIG.19C). Tumors with higher levels of CD8+ T cell infiltration and MDSCs, when treated with the combination of GalXC-STAT3-C18-4123 + anti-PD-L1 mAb, were completely eradicated.
  • Example 12 Treatment Mediated Tumor Regression and Generation of Tumor Specific Memory [00507] To evaluate if the combination treatment demonstrating complete regression also led to the generation of memory T-cells in treated mice, tumors that were completely regressed in FIG.19C were re-challenged with Hep1-6 cells (2e6 cells) on the opposite flank of the mice on day 51. As shown in FIG.20, even after the re-challenge, all mice remained tumor-free and survived for the period that they were kept and maintained ( ⁇ 2 months). These data demonstrate strong therapeutic antitumor efficacy of combination treatment leading to long term immunological memory.
  • Example 13 CD8+ T Cell Mediated Combination Efficacy is Also Perforin Dependent [00508]
  • the efficacy study described in FIG.19A was repeated in immunocompromised nude mice bearing 4T1 tumors. As shown in FIG.19A, there was synergistic efficacy with combination treatment of GalXC-STAT3-C18-4123 plus anti-PD- L1 mAb in tumor bearing immunocompetent mice, but no efficacy observed in nude mice bearing 4T1 tumors (FIG.21), suggesting that there is a key role for CD8+ T cells in mediating anti-tumor efficacy.
  • Example 14 Effect of Combination Treatment on Spontaneous Tumor Metastasis in a Highly Metastatic Tumor Model
  • mice showed tumor metastases throughout the whole organ whereas the mice administered the combination treatment (GalXC-STAT3- C18-4123 + anti-PD-L1 mAb) showed no visible metastases in the lungs of all five mice, suggesting that the treatment not only reduced the local tumor growth as shown in the figure, but also reduced the spontaneous metastases to lung.
  • the same experiment was repeated in nude mice also shown in FIG.23. All the lungs, including those from the mice that received the combination treatment had tumor metastases, further confirming the role of CD8+ T cells in anti-tumor efficacy.
  • Example 15 Treatment Mediated Immune Modulation in Tumors [00510] To understand how the combination treatment of GalXC-STAT3-C18-4123 with an anti-PD-L1 mAb changes the immune profile in tumor, CT26 tumors were implanted in Balb/c mice. These tumors are partially sensitive to checkpoint inhibitors and have the profile similar to MC38 (CD8 med MDSC med/high ).

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Medicinal Chemistry (AREA)
  • Epidemiology (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Molecular Biology (AREA)
  • Biochemistry (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

The subject matter disclosed herein is directed to modulating gene expression using siRNA compositions and methods directed to affecting key cell populations supporting the growth and metastasis of cancer to affect the beneficial treatment, remission or removal of the underlying tumor in a patient.

Description

COMBINATION OF STAT3 TARGETING OLIGONUCLEOTIDES AND PD-L1 INHIBITORS REFERENCE TO CROSS-RELATED APPLICATIONS [001] This application claims the priority to and the benefit of U.S. Provisional Patent Application Serial No. 63,320,163 filed March 15, 2022. The entire contents of which is incorporated herein by this reference. REFERENCE TO SEQUENCE LISTING [002] The contents of the electronic sequence listing (DICN_021_001WO_SeqList_ST26.xml; Size: 4,153,073 bytes; and Date of Creation: March 8, 2023) are herein incorporated by reference in its entirety. BACKGROUND OF THE DISCLOSURE [003] Currently, chemotherapy is the leading cancer therapy worldwide, often combined with surgery, or surgery and radiotherapy, depending on tumor type and stage (Abbas et al., AN OVERVIEW OF CANCER TREATMENT MODALITIES/INTECHOPEN, 2018). Since the discovery of several important mutations that contribute to carcinogenesis (e.g., epidermal cell alterations (Yamaoka et al., INT. J. MOL. SCI. (2017) 18(11): 2420)) these mutations and the proteins they represent have been extensively used as targets for the development of more selective drugs and drug combinations to treat cancer patients. Despite the effectiveness of these drugs, multidrug resistance (MDR) is often seen in patients, which often results in tumor relapse, limited therapeutic options and low quality of life for patients. In addition, cancer research has often been focused on tumor cells even though the effect of the tumor microenvironment and the ‘normal’ or non-cancerous cells within it that have been shown to play a key role in tumor progression, development and MDR (Klemm et al., TRENDS CELL BIOL (2015) 25(4): 198-213). Novel therapies that target different facets of the TME that contribute to tumor growth are needed. BRIEF SUMMARY OF THE DISCLOSURE [004] The disclosure is based, in part, on the discovery that a combination of a STAT3 oligonucleotide and a PD-L1 inhibitor provides synergistic anti-tumor efficacy for tumors of varying tumor microenvironments. Specifically, as demonstrated herein, a STAT3 oligonucleotide conjugated to a lipid, when delivered in combination with an anti-PD-L1 antibody, reduced tumor volume in vivo in immunosuppressive and inflamed tumor models. Further, as shown herein, the combination of a STAT3 oligonucleotide and PD-L1 inhibitor induced an anti-tumor memory response as when mice were re-challenged with cancer cells, no tumors were established. In addition, the efficacy of the STAT3 oligonucleotide and PD- L1 inhibitor was dependent on the presence of CD8+ T cells. [005] Accordingly, in some aspects, the disclosure provides a method of treating cancer in a subject that has received or is receiving a PD-L1 inhibitor, the method comprising administering an RNAi oligonucleotide comprising an antisense strand of 15 to 30 nucleotides in length and a sense strand of 15 to 40 nucleotides in length, wherein the antisense and sense strands form a duplex region, wherein the antisense strand comprises a region of complementarity to a STAT3 mRNA target sequence, and wherein the region of complementarity is at least 15 contiguous nucleotides in length, thereby treating cancer in the subject. [006] In other aspects, the disclosure provides a method of treating cancer in a subject that has received or is receiving an RNAi oligonucleotide, the method comprising administering a PD-L1 inhibitor, wherein the oligonucleotide comprises an antisense strand of 15 to 30 nucleotides in length and a sense strand of 15 to 40 nucleotides in length, wherein the antisense and sense strands form a duplex region, wherein the antisense strand comprises a region of complementarity to a STAT3 mRNA target sequence, and wherein the region of complementarity is at least 15 contiguous nucleotides in length, thereby treating cancer in the subject. [007] In yet other aspects, the disclosure provides a method of treating a disease, disorder or condition associated with activated STAT3 expression, comprising administering to a subject in need thereof an RNAi oligonucleotide, wherein the subject has received or is receiving a PD-L1 inhibitor, wherein the oligonucleotide comprises an antisense strand of 15 to 30 nucleotides in length and a sense strand of 15 to 40 nucleotides in length, wherein the antisense and sense strands form a duplex region, wherein the antisense strand comprises a region of complementarity to a STAT3 mRNA target sequence, and wherein the region of complementarity is at least 15 contiguous nucleotides in length, thereby treating the disease, disorder, or condition associated with activated STAT3 expression. [008] In further aspects, the disclosure provides a kit comprising an RNAi oligonucleotide, an optional pharmaceutically acceptable carrier, and a package insert comprising instructions for administering the oligonucleotide to a subject in need thereof that has received or is receiving a PD-L1 inhibitor, wherein the oligonucleotide comprises an antisense strand of 15 to 30 nucleotides in length and a sense strand of 15 to 40 nucleotides in length, wherein the antisense and sense strands form a duplex region, wherein the antisense strand comprises a region of complementarity to a STAT3 mRNA target sequence, and wherein the region of complementarity is at least 15 contiguous nucleotides in length. [009] In other aspects, the disclosure provides, a kit comprising a PD-L1 inhibitor, an optional pharmaceutically acceptable carrier, and a package insert comprising instructions for administering the antibody to a subject in need thereof that has received or is receiving an RNAi oligonucleotide comprising an antisense strand of 15 to 30 nucleotides in length and a sense strand of 15 to 40 nucleotides in length, wherein the antisense and sense strands form a duplex region, wherein the antisense strand comprises a region of complementarity to a STAT3 mRNA target sequence, and wherein the region of complementarity is at least 15 contiguous nucleotides in length. [0010] In some or any of the foregoing or related aspects, a subject has a disease, disorder, or condition associated with activated STAT3 expression. In some aspects, the disease, disorder, or condition associated with activated STAT3 expression is a cancer. In some aspects, the cancer is selected from carcinoma, sarcoma, melanoma, lymphoma, and leukemia, prostate cancer, breast cancer, hepatocellular carcinoma (HCC), colorectal cancer, pancreatic cancer and glioblastoma. In some aspects, the cancer comprises an immunosuppressive tumor microenvironment. In some aspects, the immunosuppressive tumor microenvironment has low levels of CD8+ T cell infiltration and high levels of MDSCs. In other aspects, the cancer comprises an inflamed tumor microenvironment. In some aspects, the inflamed tumor microenvironment comprises infiltrating T cells. In some aspects, the inflamed tumor microenvironment comprises high levels of CD8+ T cell infiltration and high levels of MDSCs. In some aspects, the cancer comprises a tumor resistant to immune checkpoint therapy. In some aspects, the cancer comprises a tumor partially resistant to immune checkpoint therapy. In some aspects, the cancer comprises a tumor sensitive to immune checkpoint therapy. [0011] In some or any of the foregoing or related aspects, the methods described herein treat spontaneous tumor metastasis. [0012] In some or any of the foregoing or related aspects, the methods described herein reduce immune suppressive genes and enhance immune activation genes. In some aspects, immune suppressive genes comprise checkpoint inhibitors, STAT3 mediated genes, suppressive cytokines, suppressive chemokines, and angiogenesis and matrix remodeling related genes. In some aspects, immune activation genes comprise gene related to T cell migration, T cell activation, T cell memory, and/or T cell cytotoxicity. [0013] In some or any of the foregoing or related aspects, the PD-L1 inhibitor is an antibody. In some aspects, the antibody is an anti-PD-L1 antibody. In some aspects, the anti- PDL1 antibody is selected from FAZ053, atezolizumab, avelumab, durvalumab, envafolimab, and BMS-936559. In some aspects, the antibody is an anti-PD-1 antibody. In some aspects, the anti-PD-1 antibody is selected from nivolumab, pembrolizumab, and cemiplimab. [0014] In some aspects, the PD-L1 inhibitor is a small molecule inhibitor. In some aspects, the PD-L1 inhibitor is a peptide. In some aspects, the PD-L1 inhibitor is a nucleic acid molecule. In some aspects, the nucleic acid molecule is selected from an antisense oligonucleotide, an siRNA, or an miRNA. [0015] In some or any of the foregoing or related aspects, the STAT3 mRNA target sequence comprises any one of SEQ ID NOs: 89-280. In some aspects, the region of complementarity is fully complementary to the STAT3 mRNA target sequence. In some aspects, the region of complementarity comprises no more than 4 mismatches to the STAT3 mRNA target sequence. [0016] In some or any of the foregoing or related aspects, the antisense strand is 19 to 27 nucleotides in length. In some aspects, the antisense strand is 21 to 27 nucleotides in length, optionally wherein the antisense strand is 22 nucleotides in length. [0017] In some or any of the foregoing or related aspects, the sense strand is 19 to 40 nucleotides in length, optionally wherein the sense strand is 36 nucleotides in length. [0018] In some or any of the foregoing or related aspects, the duplex region is at least 19 nucleotides in length. In some aspects, the duplex region is at least 20 nucleotides in length, optionally wherein the duplex region is 21 nucleotides in length. [0019] In some or any of the foregoing or related aspects, the region of complementarity to STAT3 is at least 19 contiguous nucleotides in length. In some aspects, the region of complementarity to STAT3 is at least 21 contiguous nucleotides in length. [0020] In some or any of the foregoing or related aspects, the sense strand comprises at its 3′ end a stem-loop set forth as: S1-L-S2, wherein S1 is complementary to S2, and wherein L forms a loop between S1 and S2 of 3 to 5 nucleotides in length. In some aspects, L is a tetraloop, optionally wherein L is 4 nucleotides in length. In some aspects, L comprises a sequence set forth as GAAA. [0021] In some or any of the foregoing or related aspects, the antisense strand comprises a 3’ overhang sequence of one or more nucleotides in length, optionally wherein the 3’ overhang sequence is 2 nucleotides in length, optionally wherein the 3’ overhang sequence is GG. [0022] In some or any of the foregoing or related aspects, the oligonucleotide comprises at least one modified nucleotide. In some aspects, the modified nucleotide comprises a 2′- modification. In some aspects, the 2′-modification is a modification selected from 2′- aminoethyl, 2′-fluoro, 2′-O-methyl, 2′-O-methoxyethyl, and 2′-deoxy-2′-fluoro-β-d- arabinonucleic acid. In some aspects, about 10-15%, 10%, 11%, 12%, 13%, 14% or 15% of the nucleotides of the sense strand comprise a 2’-fluoro modification. In some aspects, about 25-35%, 25%, 26%, 27%, 28%, 29%, 30%, 31%, 32%, 33%, 34% or 35% of the nucleotides of the antisense strand comprise a 2’-fluoro modification. In some aspects, about 25-35%, 25%, 26%, 27%, 28%, 29%, 30%, 31%, 32%, 33%, 34% or 35% of the nucleotides of the oligonucleotide comprise a 2’-fluoro modification. In some aspects, the sense strand comprises 36 nucleotides with positions 1-36 from 5’ to 3 ’, wherein positions 8-11 comprise a 2’-fluoro modification. In some aspects, the antisense strand comprises 22 nucleotides with positions 1-22 from 3’ to 5’, and wherein positions 2, 3, 4, 5, 7, 10 and 14 comprise a 2’-fluoro modification. In some aspects, the remaining nucleotides comprise a 2’-O-methyl modification. In some aspects, all of the nucleotides of the oligonucleotide are modified. [0023] In some or any of the foregoing or related aspects, the oligonucleotide comprises at least one modified internucleotide linkage. In some aspects, the at least one modified internucleotide linkage is a phosphorothioate linkage. [0024] In some or any of the foregoing or related aspects, the 4′-carbon of the sugar of the 5′-nucleotide of the antisense strand comprises a phosphate analog. In some aspects, the phosphate analog is oxymethylphosphonate, vinylphosphonate or malonylphosphonate. [0025] In some or any of the foregoing or related aspects, at least one nucleotide of the oligonucleotide is conjugated to one or more targeting ligands. In some aspects, the nucleotide is conjugated to more than one targeting ligands, wherein the targeting ligands are the same or are different. In some aspects, the one or more targeting ligands is selected from carbohydrate, amino sugar, cholesterol, polypeptide, or lipid. In some aspects, the one or more targeting ligands is a saturated or unsaturated fatty acid moiety. In some aspects, the targeting ligand is a saturated fatty acid moiety that ranges in size from C10 to C24 long. In some aspects, the targeting ligand is a C16 saturated fatty acid moiety. In some aspects, the targeting ligand is a C18 saturated fatty acid moiety. In some aspects, the targeting ligand is a C22 saturated fatty acid moiety. In some aspects, the targeting ligand comprises a N-acetylgalactosamine (GalNAc) moiety. In some aspects, the GalNAc moiety is a monovalent GalNAc moiety, a bivalent GalNAc moiety, a trivalent GalNAc moiety or a tetravalent GalNAc moiety. In some aspects, up to 4 nucleotides of L of the stem-loop are each conjugated to a monovalent GalNAc moiety. [0026] In some or any of the foregoing or related aspects, In some or any of the foregoing or related aspects, the sense strand comprises a sequence as set forth in SEQ ID NOs: 857-946. In some aspects, the antisense strand comprises a sequence as set for in SEQ ID NOs: 947-1036. [0027] In some or any of the foregoing or related aspects, the sense strand and antisense strand comprise nucleotide sequences selected from the group consisting of: (a) SEQ ID NOs: 9 and 10, respectively; (b) SEQ ID NOs: 37 and 38, respectively; (c) SEQ ID NOs: 65 and 66, respectively; and (d) SEQ ID NOs: 69 and 70, respectively. [0028] In some or any of the foregoing or related aspects, the sense strand and antisense strand comprise nucleotide sequences selected from the group consisting of: (a) SEQ ID NOs: 861 and 951, respectively; (b) SEQ ID NOs: 857 and 947, respectively; (c) SEQ ID NOs: 858 and 948, respectively; (d) SEQ ID NOs: 859 and 949, respectively; (e) SEQ ID NOs: 860 and 950, respectively; (f) SEQ ID NOs: 862 and 952, respectively; (g) SEQ ID NOs: 863 and 953, respectively; (h) SEQ ID NOs: 864 and 954, respectively; (i) SEQ ID NOs: 865 and 955, respectively; (j) SEQ ID NOs: 866 and 956, respectively; (k) SEQ ID NOs: 867 and 957, respectively; (l) SEQ ID NOs: 868 and 958, respectively; (m) SEQ ID NOs: 869 and 959, respectively; (n) SEQ ID NOs: 870 and 960, respectively; (o) SEQ ID NOs: 871 and 961, respectively; (p) SEQ ID NOs: 872 and 962, respectively; (q) SEQ ID NOs: 873 and 963, respectively; (r) SEQ ID NOs: 874 and 964, respectively; (s) SEQ ID NOs: 875 and 965, respectively; (t) SEQ ID NOs: 876 and 966, respectively; (u) SEQ ID NOs: 877 and 967, respectively; (v) SEQ ID NOs: 878 and 968, respectively; (w) SEQ ID NOs: 879 and 969, respectively; (x) SEQ ID NOs: 880 and 970, respectively; (y) SEQ ID NOs: 881and 971, respectively; (z) SEQ ID NOs: 882 and 972, respectively; (aa) SEQ ID NOs: 883 and 973, respectively; (bb) SEQ ID NOs: 884 and 974, respectively; (cc) SEQ ID NOs: 885 and 975, respectively; (dd) SEQ ID NOs: 886 and 976, respectively; (ee) SEQ ID NOs: 887 and 977, respectively; (ff) SEQ ID NOs: 888 and 978, respectively; (gg) SEQ ID NOs: 940 and 1030, respectively; (hh) SEQ ID NOs: 896 and 986, respectively; and (ii) SEQ ID NOs: 920 and 1010, respectively. [0029] In some or any of the foregoing or related aspects, the sense strand comprises the nucleotide sequence of SEQ ID NO: 862 and the antisense strand comprises the nucleotide sequence of SEQ ID NO: 952. In some aspects, the sense strand comprises the nucleotide sequence of SEQ ID NO: 875 and the antisense strand comprises the nucleotide sequence of SEQ ID NO: 965. In some aspects, the sense strand comprises the nucleotide sequence of SEQ ID NO: 876 and the antisense strand comprises the nucleotide sequence of SEQ ID NO: 966. In some aspects, sense strand comprises the nucleotide sequence of SEQ ID NO: 920 and the antisense strand comprises the nucleotide sequence of SEQ ID NO: 1010. [0030] In some or any of the foregoing or related aspects, the sense strand and antisense strand comprise nucleotide sequences selected from the group consisting of: (a) SEQ ID NOs: 901 and 991, respectively; (b) SEQ ID NOs: 910 and 1000, respectively; (c) SEQ ID NOs: 899 and 989, respectively; (d) SEQ ID NOs: 896 and 986, respectively; (e) SEQ ID NOs: 892 and 982, respectively; (f) SEQ ID NOs: 890 and 980, respectively; and (g) SEQ ID NOs: 889 and 979, respectively. [0031] In some or any of the foregoing or related aspects, the sense strand and antisense strand comprise nucleotide sequences selected from the group consisting of: (a) SEQ ID NOs: 940 and 1030, respectively; (b) SEQ ID NOs: 937 and 1027, respectively; and (c) SEQ ID NOs: 939 and 1029, respectively. [0032] In some or any of the foregoing or related aspects, the sense strand and antisense strand comprise nucleotide sequences selected from the group consisting of: (a) SEQ ID NOs: 915 and 1005, respectively; (b) SEQ ID NOs: 924 and 1014, respectively; (c) SEQ ID NOs: 913 and 1003, respectively; and (d) SEQ ID NOs: 920 and 1010, respectively. [0033] In some or any of the foregoing or related aspects, the sense strand comprises a nucleotide sequence of any one of SEQ ID NOs: 11, 39, 67 and 71. In some aspects, the antisense strand comprises a nucleotide sequence of any one of SEQ ID NOs: 12, 40, 68 and 72. [0034] In some or any of the foregoing or related aspects, the sense strand and antisense strand comprise nucleotide sequences selected from the group consisting of: (a) SEQ ID NOs: 11 and 12, respectively; (b) SEQ ID NOs: 39 and 40, respectively; (c) SEQ ID NOs: 67 and 68, respectively; and (d) SEQ ID NOs: 71 and 72, respectively. [0035] In some or any of the foregoing or related aspects, the sense strand comprises a nucleotide sequence selected from SEQ ID NOs: 1042, 1055, 1056, and 1100. In some aspects, the antisense strand comprises a nucleotide sequence selected from SEQ ID NOs: 1132, 1145, 1146, and 1190. [0036] In some or any of the foregoing or related aspects, the sense strand and antisense strand comprise nucleotide sequences selected from the group consisting of: (a) SEQ ID NOs: 1041 and 1131, respectively; (b) SEQ ID NOs: 1037 and 1127, respectively; (c) SEQ ID NOs: 1038 and 1128, respectively; (d) SEQ ID NOs: 1039 and 1129, respectively; (e) SEQ ID NOs: 1040 and 1130, respectively; (f) SEQ ID NOs: 1042 and 1132, respectively; (g) SEQ ID NOs: 1043 and 1133, respectively; (h) SEQ ID NOs: 1044 and 1134, respectively; (i) SEQ ID NOs: 1045 and 1135, respectively; (j) SEQ ID NOs: 1046 and 1136, respectively; (k) SEQ ID NOs: 1047 and 1137, respectively; (l) SEQ ID NOs: 1048 and 1138, respectively; (m) SEQ ID NOs: 1049 and 1139, respectively; (n) SEQ ID NOs: 1050 and 1140, respectively; (o) SEQ ID NOs: 1051 and 1141, respectively; (p) SEQ ID NOs: 1052 and 1142, respectively; (q) SEQ ID NOs: 1053 and 1143, respectively; (r) SEQ ID NOs: 1054 and 1144, respectively; (s) SEQ ID NOs: 1055 and 1145, respectively; (t) SEQ ID NOs: 1056 and 1146, respectively; (u) SEQ ID NOs: 1057 and 1147, respectively; (v) SEQ ID NOs: 1058 and 1148, respectively; (w) SEQ ID NOs: 1059 and 1149, respectively; (x) SEQ ID NOs: 1060 and 1150, respectively; (y) SEQ ID NOs: 1061 and 1151, respectively; (z) SEQ ID NOs: 1062 and 1152, respectively; (aa) SEQ ID NOs: 1063 and 1153, respectively; (bb) SEQ ID NOs: 1064 and 1154, respectively; (cc) SEQ ID NOs: 1065 and 1155, respectively; (dd) SEQ ID NOs: 1066 and 1156, respectively; (ee) SEQ ID NOs: 1067 and 1157, respectively; (ff) SEQ ID NOs: 1068 and 1158, respectively; (gg) SEQ ID NOs: 1120 and 1210, respectively; (hh) SEQ ID NOs: 1076 and 1166, respectively; and (ii) SEQ ID NOs: 1100 and 1190, respectively. [0037] In some or any of the foregoing or related aspects, the sense strand comprises the nucleotide sequence of SEQ ID NO: 1042 and the antisense strand comprises the nucleotide sequence of SEQ ID NO: 1132. In some aspects, the sense strand comprises the nucleotide sequence of SEQ ID NO: 1055 and the antisense strand comprises the nucleotide sequence of SEQ ID NO: 1145. In some aspects, the sense strand comprises the nucleotide sequence of SEQ ID NO: 1056 and the antisense strand comprises the nucleotide sequence of SEQ ID NO: 1146. In some aspects, the sense strand comprises the nucleotide sequence of SEQ ID NO: 1100 and the antisense strand comprises the nucleotide sequence of SEQ ID NO: 1190. [0038] In some or any of the foregoing or related aspects, the sense strand and antisense strand comprise nucleotide sequences selected from the group consisting of: (a) SEQ ID NOs: 1081 and 1171, respectively; (b) SEQ ID NOs: 1090 and 1180, respectively; (c) SEQ ID NOs: 1079 and 1169, respectively; (d) SEQ ID NOs: 1076 and 1166, respectively; (e) SEQ ID NOs: 1072 and 1162, respectively; (f) SEQ ID NOs: 1070 and 1160, respectively; and (g) SEQ ID NOs: 1069 and 1159, respectively. [0039] In some or any of the foregoing or related aspects, wherein the sense strand and antisense strand comprise nucleotide sequences selected from the group consisting of: (a) SEQ ID NOs: 1120 and 1210, respectively; (b) SEQ ID NOs: 1117 and 1207, respectively; and (c) SEQ ID NOs: 1119 and 1209, respectively. [0040] In some or any of the foregoing or related aspects, the sense strand and antisense strand comprise nucleotide sequences selected from the group consisting of: (a) SEQ ID NOs: 1095 and 1185, respectively; (b) SEQ ID NOs: 1104 and 1194, respectively; (c) SEQ ID NOs: 1093 and 1183, respectively; and (d) SEQ ID NOs: 1100 and 1190, respectively. [0041] In some aspects, the disclosure provides a method of treating cancer in a subject that has received or is receiving an anti-PD-L1 antibody, the method comprising administering an RNAi oligonucleotide comprising an antisense strand comprising a nucleotide sequence set forth in SEQ ID NO: 84, wherein the antisense strand comprises a region of complementarity to a STAT3 mRNA target sequence, and a sense strand comprising a nucleotide sequence set forth in SEQ ID NO: 83, thereby treating cancer in the subject. [0042] In other aspects, the disclosure provides a method of treating cancer in a subject that has received or is receiving an RNAi oligonucleotide, the method comprising administering an anti-PD-L1 antibody, wherein the oligonucleotide comprises an antisense strand comprising a nucleotide sequence set forth in SEQ ID NO: 84, wherein the antisense strand comprises a region of complementarity to a STAT3 mRNA target sequence, and a sense strand comprising a nucleotide sequence set forth in SEQ ID NO: 83, thereby treating cancer in the subject. [0043] In yet other aspects, the disclosure provides a method of treating a disease, disorder or condition associated with activated STAT3 expression, comprising administering to a subject in need thereof an RNAi oligonucleotide, wherein the subject has received or is receiving an anti-PD-L1 antibody, wherein the oligonucleotide comprises an antisense strand comprising a nucleotide sequence set forth in SEQ ID NO: 84, wherein the antisense strand comprises a region of complementarity to a STAT3 mRNA target sequence, and a sense strand comprising a nucleotide sequence set forth in SEQ ID NO: 83, thereby treating the disease, disorder, or condition associated with activated STAT3 expression. [0044] In further aspects, the disclosure provides a kit comprising an RNAi oligonucleotide, an optional pharmaceutically acceptable carrier, and a package insert comprising instructions for administering the oligonucleotide to a subject in need thereof that has received or is receiving an anti-PD-L1 antibody, wherein the oligonucleotide comprises an antisense strand comprising a nucleotide sequence set forth in SEQ ID NO: 84, wherein the antisense strand comprises a region of complementarity to a STAT3 mRNA target sequence, and a sense strand comprising a nucleotide sequence set forth in SEQ ID NO: 83. [0045] In other aspects, the disclosure provides, a kit comprising an anti-PD-L1 antibody, an optional pharmaceutically acceptable carrier, and a package insert comprising instructions for administering the antibody to a subject in need thereof that has received or is receiving an RNAi oligonucleotide comprising an antisense strand comprising a nucleotide sequence set forth in SEQ ID NO: 84, wherein the antisense strand comprises a region of complementarity to a STAT3 mRNA target sequence, and a sense strand comprising a nucleotide sequence set forth in SEQ ID NO: 83. [0046] In some aspects, the disclosure provides a method of treating cancer in a subject that has received or is receiving an anti-PD-L1 antibody, the method comprising administering an RNAi oligonucleotide comprising an antisense strand comprising a nucleotide sequence set forth in SEQ ID NO: 70, wherein the antisense strand comprises a region of complementarity to a STAT3 mRNA target sequence, and a sense strand comprising a nucleotide sequence set forth in SEQ ID NO: 69, thereby treating cancer in the subject. [0047] In other aspects, the disclosure provides a method of treating cancer in a subject that has received or is receiving an RNAi oligonucleotide, the method comprising administering an anti-PD-L1 antibody, wherein the oligonucleotide comprises an antisense strand comprising a nucleotide sequence set forth in SEQ ID NO: 70, wherein the antisense strand comprises a region of complementarity to a STAT3 mRNA target sequence, and a sense strand comprising a nucleotide sequence set forth in SEQ ID NO: 69, thereby treating cancer in the subject. [0048] In yet other aspects, the disclosure provides a method of treating a disease, disorder or condition associated with activated STAT3 expression, comprising administering to a subject in need thereof an RNAi oligonucleotide, wherein the subject has received or is receiving an anti-PD-L1 antibody, wherein the oligonucleotide comprises an antisense strand comprising a nucleotide sequence set forth in SEQ ID NO: 70, wherein the antisense strand comprises a region of complementarity to a STAT3 mRNA target sequence, and a sense strand comprising a nucleotide sequence set forth in SEQ ID NO: 69, thereby treating the disease, disorder, or condition associated with activated STAT3 expression. [0049] In further aspects, the disclosure provides a kit comprising an RNAi oligonucleotide, an optional pharmaceutically acceptable carrier, and a package insert comprising instructions for administering the oligonucleotide to a subject in need thereof that has received or is receiving an anti-PD-L1 antibody, wherein the oligonucleotide comprises an antisense strand comprising a nucleotide sequence set forth in SEQ ID NO: 70, wherein the antisense strand comprises a region of complementarity to a STAT3 mRNA target sequence, and a sense strand comprising a nucleotide sequence set forth in SEQ ID NO: 69. [0050] In other aspects, the disclosure provides, a kit comprising an anti-PD-L1 antibody, an optional pharmaceutically acceptable carrier, and a package insert comprising instructions for administering the antibody to a subject in need thereof that has received or is receiving an RNAi oligonucleotide comprising an antisense strand comprising a nucleotide sequence set forth in SEQ ID NO: 70, wherein the antisense strand comprises a region of complementarity to a STAT3 mRNA target sequence, and a sense strand comprising a nucleotide sequence set forth in SEQ ID NO: 69. BRIEF DESCRIPTION OF THE DRAWINGS [0051] FIG.1A provides structures of RNAi oligonucleotide molecules having chemical modifications with GalNAc (top) or lipid (e.g., C18 hydrocarbon chain) (bottom) conjugated to the oligonucleotide molecule to generate oligonucleotide-ligand conjugates. [0052] FIG.1B provides structures of lipid tails suitable for conjugation to RNAi oligonucleotide molecules. [0053] FIGs.2A and 2B are graphs showing remaining mouse Stat3 mRNA levels in the livers of mice treated with GalXC-STAT3-conjugates (GalNAc conjugates) targeting different regions of Stat3 mRNA. Mice were administered a single dose (3mg/kg) (FIG.2A) or varying doses (0.3, 1.0 or 3.0 mg/kg) to determine dose responsiveness (FIG.2B). Arrows indicate constructs selected for further study. [0054] FIGs.3A and 3B are graphs showing mouse Stat3 mRNA expression 3 days after treatment with GalXC-STAT3-C18 conjugates in G-MDSCs and M-MDSCs derived from Pan02 xenografts implanted in mice. Tumors were dosed at 25 mg/kg (FIG.3A) and 50 mg/kg (FIG.3B). [0055] FIGs.4A and 4B are graphs showing mouse Stat3 mRNA expression after treatment of Pan02 xenograft mice with GalXC-STAT3-C18 conjugates in bulk tumor (TME) (FIG.4A) and tumor draining lymph nodes (TdLNs) (FIG.4B) at doses of 25 and 50 mg/kg. [0056] FIG.5A provides graphs showing the effect of GalXC-STAT3-C18-4123 on Stat3 and Pdl1 mRNA levels in G/M-MDSCs in TME and TdLNs of Pan02 xenograft mice 3 days after a dose of 25 or 50 mg/kg of the conjugated oligonucleotide. [0057] FIG.5B provides graphs showing the effect of GalXC-STAT3-C18-4123 on Stat3 and Pdl1 mRNA levels in TdLN of Pan02 xenograft mice 7 days after a 25mg/kg dose of the conjugated oligonucleotide. [0058] FIGs.6A and 6B are graphs showing the in vivo effect of subcutaneous treatment of a total dose of 50 mg/kg GalXC-STAT3-C18-4123 on tumor volume over time in immunocompetent mice bearing Pan02 murine pancreatic tumors. Mice were treated with either four 12.5 mg/kg (FIG.6A) or two 25mg/kg (FIG.6B) doses of the conjugated oligonucleotide. [0059] FIG.7 provides a graph depicting the percent (%) of human STAT3 mRNA remaining in Huh7 cells endogenously expressing human STAT3, after 24-hour treatment with 1nM of DsiRNA targeting various regions of the STAT3 gene.192 DsiRNAs were designed and screened. Two primer pairs were used. Expression was normalized between samples using the HPRT and SFRS9 housekeeping genes (Forward 1- SEQ ID NO: 1219, Reverse 1- SEQ ID NO: 1220; Probe 1- SEQ ID NO: 1221; Forward 2- SEQ ID NO: 1222, Reverse 2- SEQ ID NO: 1223; Probe 2- SEQ ID NO: 1224). [0060] FIGs.8A and 8B provide graphs depicting the percent (%) of human STAT3 mRNA remaining in Huh7 cells endogenously expressing human STAT3, after 24-hour treatment with 0.05nM, 0.3nM, or 1nM of DsiRNA targeting various regions of the STAT3 gene.48 GalNAc-conjugated STAT3 oligonucleotides were assayed in FIG.8A and 34 of those oligonucleotides were selected for further testing in vivo (FIG.8B). [0061] FIGs.9A and 9B provide graphs depicting the percent (%) of human STAT3 mRNA remaining in liver of mice exogenously expressing human STAT3 (hydrodynamic injection model) after treatment with GalNAc-conjugated STAT3 oligonucleotides. Mice were dosed subcutaneously with 1mg/kg of the indicated GalNAc-STAT3 oligonucleotides formulated in PBS. Three days post-dose mice were hydrodynamically injected (HDI) with a DNA plasmid encoding human STAT3. The level of human STAT3 mRNA was determined from livers collected 18 hours after injection. Arrows indicate oligonucleotides selected for dose response analysis. Hs/Mf = human/monkey common sequence; Hs/Mm= human/mouse common sequence; Hs/Mf/Mm= human/monkey/mouse triple common sequence. [0062] FIG.10 provides a graph depicting the dose response of GalNAc-conjugated STAT3 oligonucleotides. The percent (%) of human STAT3 mRNA remaining in liver of mice exogenously expressing STAT3 (HDI model) after treatment with human GalNAc-conjugated STAT3 oligonucleotides at two different doses (0.3mg/kg or 1mg/kg,) was measured. The level of human STAT3 mRNA was determined from livers collected 18 hours after injection with plasmid encoding human STAT3. Arrows indicate oligonucleotides selected for dose response analysis. Hs/Mf = human/monkey common sequence; Hs/Mm= human/mouse common sequence [0063] FIG.11 provides a graph depicting the normalized (to Ppib) relative mouse STAT3 mRNA remaining in liver of mice endogenously expressing mouse STAT3 after treatment with GalNAc-conjugated STAT3 oligonucleotides. Mice were dosed subcutaneously with 3mg/kg of the indicated GalNAc-STAT3 oligonucleotides formulated in PBS. Five days post-dose liver was collected and the level of mouse STAT3 mRNA was determined. Arrows indicate top oligonucleotides and those selected for dose response study. [0064] FIG.12 provides a graph depicting the normalized (to Ppib) relative mouse STAT3 mRNA remaining in liver of mice endogenously expressing mouse STAT3 after treatment with GalNAc-conjugated STAT3 oligonucleotides. Mice were dosed subcutaneously with 3mg/kg of the indicated GalNAc-STAT3 oligonucleotides formulated in PBS. Five days post-dose liver was collected and the level of mouse STAT3 mRNA was determined. Arrows indicate oligonucleotides selected for dose response study. [0065] FIGs.13A and 13B provide graphs depicting the dose response of GalNAc- conjugated STAT3 oligonucleotides. The percent (%) of mouse STAT3 mRNA remaining in liver of mice endogenously expressing STAT3 after treatment with human GalNAc- conjugated STAT3 oligonucleotides at three doses (0.3mg/kg, 1mg/kg, and 3mg/kg) was measured. The level of mouse STAT3 mRNA was determined from livers collected 5 days later. TC = triple common (mouse/human/monkey); Hs_Mm = human/mouse. [0066] FIG.14 provides a graph depicting the percent (%) of human STAT3 mRNA remaining in liver of mice exogenously expressing human STAT3 (hydrodynamic injection model) after treatment with GalNAc-conjugated STAT3 oligonucleotides. Mice were dosed subcutaneously with 1mg/kg of the indicated GalNAc-STAT3 oligonucleotides formulated in PBS. Three days post-dose mice were hydrodynamically injected (HDI) with a DNA plasmid encoding human STAT3. The level of human STAT3 mRNA was determined from livers collected 18 hours after injection. Arrows indicate oligonucleotides selected for dose response study. [0067] FIG.15 provides a graph depicting the dose response of GalNAc-conjugated STAT3 oligonucleotides. The percent (%) of human STAT3 mRNA remaining in liver of mice exogenously expressing human STAT3 (hydrodynamic injection model) after treatment with GalNAc-conjugated STAT3 oligonucleotides. Mice were dosed subcutaneously with three doses (0.3mg/kg, 1mg/kg, and 3mg/kg) of the indicated GalNAc-STAT3 oligonucleotides formulated in PBS. Three days post-dose mice were hydrodynamically injected (HDI) with a DNA plasmid encoding human STAT3. The level of human STAT3 mRNA was determined from livers collected 18 hours after injection. TC = triple common (mouse/human/monkey); Hs_Mm = human/mouse; Hs = human. [0068] FIG.16 provides a graph depicting the dose response of GalNAc-conjugated STAT3 oligonucleotides. The percent (%) of human STAT3 mRNA remaining in liver of mice exogenously expressing human STAT3 (hydrodynamic injection model) after treatment with GalNAc-conjugated STAT3 oligonucleotides. Mice were dosed subcutaneously with two doses (0.3mg/kg and 1mg/kg) of the indicated GalNAc-STAT3 oligonucleotides formulated in PBS. Three days post-dose mice were hydrodynamically injected (HDI) with a DNA plasmid encoding human STAT3. The level of human STAT3 mRNA was determined from livers collected 18 hours after injection. [0069] FIG.17 provides a graph depicting the percent (%) remaining human STAT1 mRNA in Huh7 cells endogenously expressing STAT3 and STAT1 treated with GalNAc- conjugated STAT3 oligonucleotides. Cells were treated for 24 hours with three doses (0.05nM, 0.3nM, and 1nM) of oligonucleotide. [0070] FIG.18A provides a graph depicting tumor volume after administration of a GalXC-STAT3-C18 oligonucleotide alone or in combination with an anti-PD-L1 mAb. Immunocompetent mice bearing Pan02 murine pancreatic tumors were dosed subcutaneously (s.c.) with 25 mg/kg of GalXC-STAT3-C18-4123 with intraperitoneal (i.p.) treatment of 10 mg/kg of anti-PD-L1 mAb. Controls included GalXC-Placebo (an HBV siRNA with identical chemistry and lipid conjugation as GalXC-STAT3 oligonucleotides), GalXC-STAT3-C18- 4123 at 25 mg/kg or GalXC-Placebo at 25 mg/kg in combination with anti-PD-L1 mAb at 10 mg/kg. Mice were first administered two doses three days apart, and two weeks later were administered two more doses three days apart [(q3dx2) x2]. Arrows indicate days doses were administered. [0071] FIG.18B provides a graph depicting tumor volume after administration of GalXC-STAT3-C18 oligonucleotide in combination with anti-PD-L1 mAb. Placebo treated mice from FIG.18A were dosed subcutaneously (s.c.) with 25 mg/kg of GalXC-STAT3- C18-4123 with intraperitoneal (i.p.) treatment of 10 mg/kg of anti-PD-L1 mAb at Day 59. [0072] FIGs.19A-19C provide graphs depicting tumor volume after administration of a GalXC-STAT3-C18 oligonucleotide alone or in combination with an anti-PD-L1 mAb or GalXC-Placebo alone or in combination with anti-PD-L1 mAb in tumors with different immunophenotypes.4T1 (triple negative breast, checkpoint resistant) (FIG.19A), MC-38 (Colon carcinoma, partially checkpoint sensitive) (FIG.19B), or Hepa1-6 (Hepatocellular carcinoma, checkpoint sensitive) (FIG.19C) cells were implanted into mice. Tumor bearing mice were dosed s.c. with 25 mg/kg of GalXC-STAT3-C18-4123 with i.p. treatment of 10 mg/kg of anti-PD-L1 mAb. Controls included GalXC-Placebo, GalXC-STAT3-C18-4123 at 25 mg/kg or GalXC-Placebo at 25 mg/kg in combination with anti-PD-L1 mAb at 10 mg/kg.. Mice bearing MC-38 and Hepa1-6 tumors were administered two doses three days apart at 25 mg/kg and the same regimen was repeated the following week. Mice bearing 4T1 tumors were administered three doses each three days apart (q3d x 3). Arrow (5/5 CR) = All mice treated were complete responders. [0073] FIG.20 provides a graph depicting the effect of Hepa1-6 re-challenge in the completely eradicated tumors. After tumors in all 5 mice were completely regressed with the treatment of GalXC-STAT3-C18 (25 mg/kg, s.c.) and anti-PD-L1 mAb (10 mg/kg, i.p.) in FIG.19C, mice were rechallenged on day 51 with Hepa1-6 cells (2e6 cells/mouse) on the opposite flank of the mice and tumor volume was monitored (FIG.20). Arrow (5/5 CR) = All mice remained tumor free even after the re-challenge. [0074] FIG.21 provides a graph depicting tumor volume after administration of GalXC- STAT3-C18 oligonucleotide alone or in combination with an anti-PD-L1 mAb in immunocompromised mice with no functional CD8+ T cells. Mice bearing 4T1 tumors were dosed s.c. with GalXC-STAT3-C18-4123 (25 mg/kg, three times with each dose three days apart (q3d x 3)) and i.p. with anti-PD-L1 mAb (10 mg/kg, q3d x 3). Controls included GalXC-Placebo, GalXC-STAT3-C18-4123 at 25 mg/kg or GalXC-Placebo at 25 mg/kg in combination with anti-PD-L1 mAb at 10 mg/kg. [0075] FIG.22 provides images showing the appearance of tumors (with cell death) from mice assayed in FIG.19A, and perforin staining for positive cytotoxic CD8+ T cells in the tumors at the end of the study. [0076] FIG.23 provides images showing lung tumor metastasis after administration of GalXC-STAT3-C18-4123 oligonucleotide alone or in combination with an anti-PD-L1 mAb as treated in FIGs.19A and 21. Mice (immunocompetent or immunocompromised) bearing 4T1 tumors were dosed s.c. with GalXC-STAT3-C18-4123 (50 mg/kg, q3d x 3) and i.p. with anti-PD-L1 mAb (10 mg/kg, q3d x 3). Controls included GalXC-Placebo, GalXC-STAT3- C18-4123 at 50 mg/kg or GalXC-Placebo at 50 mg/kg in combination with anti-PD-L1 mAb at 10 mg/kg. [0077] FIG.24 provides a heat map showing the regulation of targets involved in immune modulation observed in CT26 tumors upon combination treatment of GalXC- STAT3-C18-4123 (s.c, 25 mg/kg, q3d x 3) and anti-PD-L1 mAb (i.p. at 10 mg/kg, q3d x 3) compared to controls including GalXC-Placebo, GalXC-STAT3-C18-4123 at 25 mg/kg or GalXC-Placebo at 25 mg/kg in combination with anti-PD-L1 mAb at 10 mg/kg. DETAILED DESCRIPTION [0078] The present disclosure now will be described more fully hereinafter with reference to the accompanying drawings, in which illustrative embodiments of the disclosure are shown. The disclosure may, however, be embodied in many different forms and should not be construed as limited to the embodiments set forth herein; rather, these embodiments are provided so that this disclosure will be thorough and complete, and will fully convey the scope of the disclosure to those skilled in the art. Definitions [0079] The publications discussed throughout the text are provided solely for their disclosure prior to the filing date of the present application. Nothing herein is to be construed as an admission that the inventors are not entitled to antedate such disclosure by virtue of prior disclosure. [0080] As used herein, the term "and/or" includes any and all combinations of one or more of the associated listed items. Further, the singular forms and the articles "a", "an" and "the" are intended to include the plural forms as well, unless expressly stated otherwise. It will be further understood that the terms: includes, comprises, including and/or comprising, when used in this specification, specify the presence of stated features, integers, steps, operations, elements, and/or components, but do not preclude the presence or addition of one or more other features, integers, steps, operations, elements, components, and/or groups thereof. Further, it will be understood that when an element, including component or subsystem, is referred to and/or shown as being connected or coupled to another element, it can be directly connected or coupled to the other element or intervening elements may be present. [0081] Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood to one of ordinary skill in the art to which this disclosure belongs. Although methods and materials similar or equivalent to those described herein can be used in the practice of the disclosed methods and compositions, exemplary methods, and materials are described herein. [0082] General texts which describe molecular biological techniques useful herein, including the use of vectors, promoters and many other relevant topics, include Berger and Kimmel, GUIDE TO MOLECULAR CLONING TECHNIQUES, METHODS IN ENZYMOLOGY, volume 152, (Academic Press, Inc., San Diego, Calif.) ("Berger"); Sambrook et al., MOLECULAR CLONING--A LABORATORY MANUAL, 2d ed., Vol.1-3, Cold Spring Harbor Laboratory, Cold Spring Harbor, 1989 ("Sambrook") and CURRENT PROTOCOLS IN MOLECULAR BIOLOGY, F.M. Ausubel et al., eds., CURRENT PROTOCOLS, A JOINT VENTURE BETWEEN GREENE PUBLISHING ASSOCIATES, INC. AND JOHN WILEY AND SONS, INC., (supplemented through 1999) ("Ausubel"). Examples of protocols sufficient to direct persons of skill through in vitro amplification methods, including the polymerase chain reaction (PCR), the ligase chain reaction(LCR), Q.beta.-replicase amplification and other RNA polymerase mediated techniques (e.g., NASBA), e.g., for the production of the homologous nucleic acids of the disclosure are found in Berger, Sambrook, and Ausubel, as well as in Mullis et al., (1987) U.S. Pat. No.4,683,202; Innis et al., eds. (1990); PCR PROTOCOLS: A GUIDE TO METHODS AND APPLICATIONS (Academic Press Inc. San Diego, Calif.) ("Innis"); Arnheim and Levinson (Oct.1, 1990) Cand EN 36-47; J. NIH RES. (1991) 3:81-94; Kwoh et al., (1989) PROC. NATL. ACAD. SCI. USA 86: 1173; Guatelliet et al., (1990) PROC. NAT'L. ACAD. SCI. USA 87: 1874; Lomell et al., (1989) J. CLIN. CHEM 35: 1826; Landegren et al., (1988) SCIENCE 241: 1077- 80; Van Brunt (1990) BIOTECHNOLOGY 8: 291-94; Wu and Wallace (1989) GENE 4:560; Barringer et al., (1990) GENE 89:117; and, Sooknanan and Malek(1995) BIOTECHNOLOGY 13: 563-564. Improved methods for cloning in vitro amplified nucleic acids are described in Wallace et al., U.S. Pat. No.5,426,039. Improved methods for amplifying large nucleic acids by PCR are summarized in Cheng et al., (1994) NATURE 369: 684-85 and the references cited therein, in which PCR amplicons of up to 40 kb are generated. [0083] As used in the specification and the appended claims, the singular forms "a," "an" and "the" include plural referents unless the context clearly dictates otherwise. Thus, for example, reference to "a pharmaceutical carrier" includes mixtures of two or more such carriers, and the like. [0084] Ranges can be expressed herein as from "about" one value, and/or to "about" another value. When such a range is expressed, another embodiment includes from the one value and/or to the other value. Similarly, when values are expressed as approximations, by use of the antecedent "about," it will be understood that the value forms another embodiment. It will be further understood that the endpoints of each of the ranges are significant both in relation to the other endpoint, and independently of the other endpoint. It is also understood that there are several values disclosed herein, and that each value is also herein disclosed as "about" that value in addition to the value itself. For example, if the value "10" is disclosed, then "about 10" is also disclosed. It is also understood that when a value is disclosed that "less than or equal to" the value, "greater than or equal to the value" and possible ranges between values are also disclosed, as appropriately understood by the skilled artisan. For example, if the value "10" is disclosed the "less than or equal to 10" as well as "greater than or equal to 10" is also disclosed. It is also understood that the throughout the application, data is provided in several different formats, and that this data, represents endpoints and starting points, and ranges for any combination of the data points. For example, if a particular datapoint "10" and a particular data point 15 are disclosed, it is understood that greater than, greater than or equal to, less than, less than or equal to, and equal to 10 and 15 are considered disclosed as well as between 10 and 15. It is also understood that each unit between two particular units are also disclosed. For example, if 10 and 15 are disclosed, then 11, 12, 13, and 14 are also disclosed. [0085] In this specification and in the claims, which follow, reference will be made to several terms which shall be defined to have the following meanings: [0086] The term "cancer" or "tumor" includes, but is not limited to, solid tumors and blood borne tumors. These terms include diseases of the skin, tissues, organs, bone, cartilage, blood, and vessels. These terms further encompass primary and metastatic cancers. [0087] The term “PD-1” refers to a protein found on T cells that helps keep the immune responses in check. When PD-1 is bound to another protein called PD-L1, it helps keep T cells from killing other cells, including cancer cells. Some anticancer drugs, called immune checkpoint inhibitors, are used to block PD-1. When this protein is prevented from acting on T cells, they can act to kill cancer cells. [0088] The term “STAT3” refers to Signal transducer and activator of transcription 3 (STAT3) which is a transcription factor which in humans is encoded by the STAT3 gene (STAT3 Human (Hs) NM_001369512.1 Genbank RefSeq #, or NM_139276.3). STAT3 mediates the expression of a variety of genes in response to cell stimuli, and thus plays a key role in many cellular processes such as cell growth and apoptosis, as well as the growth and progression of cancer. [0089] As used herein, the term "cold tumor" or "non-inflamed tumor" refers to a tumor or tumor microenvironment wherein there is minimal to no presence of anti-tumor immune cells, such as tumor infiltrating lymphocytes (TILs), and/or contain cell subsets associated with immune suppression including regulatory T cells (Treg), myeloid-derived suppressor cells (MDSCs) and M2 macrophages. Specifically, in some embodiments, a cold tumor is characterized by a low number or even absence of infiltration of anti-tumor immune cells that such cells may be present but remain stuck in the surrounding stroma, thus unable to colonize the tumor microenvironment to provide their antitumor functions. [0090] As used herein, “complementary” refers to a structural relationship between two nucleotides (e.g., on two opposing nucleic acids or on opposing regions of a single nucleic acid strand) that permits the two nucleotides to form base pairs with one another. For example, a purine nucleotide of one nucleic acid that is complementary to a pyrimidine nucleotide of an opposing nucleic acid may base pair together by forming hydrogen bonds with one another. In some embodiments, complementary nucleotides can base pair in the Watson-Crick manner or in any other manner that allows for the formation of stable duplexes. In some embodiments, two nucleic acids may have regions of multiple nucleotides that are complementary with each other to form regions of complementarity, as described herein. [0091] As used herein, “species cross-reactive oligonucleotide” refers to an oligonucleotide capable of inhibiting expression of a target mRNA in more than one species. For example, in some embodiments a species cross-reactive oligonucleotide is capable of inhibiting expression of a target mRNA in human and non-human primates. Example species include but is not limited to human, non-human primates, mouse, and rat. In some embodiments, species cross-reactive oligonucleotides are capable of targeting and inhibiting mRNA in at least two, at least three, or at least four species. [0092] As used herein, “deoxyribonucleotide” refers to a nucleotide having a hydrogen in place of a hydroxyl at the 2′ position of its pentose sugar when compared with a ribonucleotide. A modified deoxyribonucleotide is a deoxyribonucleotide having one or more modifications or substitutions of atoms other than at the 2′ position, including modifications or substitutions in or of the sugar, phosphate group or base. [0093] As used herein, “double-stranded RNA” or “dsRNA” refers to an RNA oligonucleotide that is substantially in a duplex form. In some embodiments, the complementary base-pairing of duplex region(s) of a dsRNA oligonucleotide is formed between antiparallel sequences of nucleotides of covalently separate nucleic acid strands. In some embodiments, complementary base-pairing of duplex region(s) of a dsRNA formed between antiparallel sequences of nucleotides of nucleic acid strands that are covalently linked. In some embodiments, complementary base-pairing of duplex region(s) of a dsRNA is formed from single nucleic acid strand that is folded (e.g., via a hairpin) to provide complementary antiparallel sequences of nucleotides that base pair together. In some embodiments, a dsRNA comprises two covalently separate nucleic acid strands that are fully duplexed with one another. However, in some embodiments, a dsRNA comprises two covalently separate nucleic acid strands that are partially duplexed (e.g., having overhangs at one or both ends). In some embodiments, a dsRNA comprises antiparallel sequence of nucleotides that are partially complementary, and thus, may have one or more mismatches, which may include internal mismatches or end mismatches. [0094] As used herein, “duplex,” in reference to nucleic acids (e.g., oligonucleotides), refers to a structure formed through complementary base pairing of two antiparallel sequences of nucleotides. [0095] As used herein, “excipient” refers to a non-therapeutic agent that may be included in a composition, for example, to provide or contribute to a desired consistency or stabilizing effect. [0096] As used herein, the term "hot tumor" or "inflamed tumor" refers to a tumor or tumor microenvironment wherein there is a considerable presence of anti-tumor immune cells especially TILs and thus are typically immuno-stimulatory. [0097] As used herein, “loop” refers to an unpaired region of a nucleic acid (e.g., oligonucleotide) that is flanked by two antiparallel regions of the nucleic acid that are sufficiently complementary to one another, such that under appropriate hybridization conditions (e.g., in a phosphate buffer, in a cells), the two antiparallel regions, which flank the unpaired region, hybridize to form a duplex (referred to as a “stem”). The loop may refer to a loop comprising four nucleotides as a tetraloop (tetraL). The loop may refer to a loop comprising three nucleotides as a triloop (triL). [0098] As used herein, “modified internucleotide linkage” refers to an internucleotide linkage having one or more chemical modifications when compared with a reference internucleotide linkage comprising a phosphodiester bond. In some embodiments, a modified nucleotide is a non-naturally occurring linkage. Typically, a modified internucleotide linkage confers one or more desirable properties to a nucleic acid in which the modified internucleotide linkage is present. For example, a modified nucleotide may improve thermal stability, resistance to degradation, nuclease resistance, solubility, bioavailability, bioactivity, reduced immunogenicity, etc. [0099] As used herein, “modified nucleotide” refers to a nucleotide having one or more chemical modifications when compared with a corresponding reference nucleotide selected from: adenine ribonucleotide, guanine ribonucleotide, cytosine ribonucleotide, uracil ribonucleotide, adenine deoxyribonucleotide, guanine deoxyribonucleotide, cytosine deoxyribonucleotide and thymidine deoxyribonucleotide. In some embodiments, a modified nucleotide is a non-naturally occurring nucleotide. In some embodiments, a modified nucleotide has one or more chemical modification in its sugar, nucleobase and/or phosphate group. In some embodiments, a modified nucleotide has one or more chemical moieties conjugated to a corresponding reference nucleotide. Typically, a modified nucleotide confers one or more desirable properties to a nucleic acid in which the modified nucleotide is present. For example, a modified nucleotide may improve thermal stability, resistance to degradation, nuclease resistance, solubility, bioavailability, bioactivity, reduced immunogenicity, etc. [00100] As used herein, “nicked tetraloop structure” refers to a structure of a RNAi oligonucleotide that is characterized by separate sense (passenger) and antisense (guide) strands, in which the sense strand has a region of complementarity with the antisense strand, and in which at least one of the strands, generally the sense strand, has a tetraloop configured to stabilize an adjacent stem region formed within the at least one strand. [00101] As used herein, “oligonucleotide” refers to a short nucleic acid (e.g., less than about 100 nucleotides in length). An oligonucleotide may be single stranded (ss) or double- stranded (ds). An oligonucleotide may or may not have duplex regions. An oligonucleotide may comprise deoxyribonucleotides, ribonucleosides, or a combination of both. In some embodiments, a double-stranded oligonucleotide comprising ribonucleotides is referred to as “dsRNA”. As a set of non-limiting examples, an oligonucleotide may be, but is not limited to, a small interfering RNA (siRNA), microRNA (miRNA), short hairpin RNA (shRNA), dicer substrate interfering RNA (dsiRNA), antisense oligonucleotide, short siRNA or ss siRNA. In some embodiments, a double-stranded RNA (dsRNA) is an RNAi oligonucleotide. [00102] The terms “RNAi oligonucleotide conjugate” and “oligonucleotide-ligand conjugate” are used interchangeably and refer to an oligonucleotide comprising one or more nucleotides conjugated with one or more targeting ligands. [00103] As used herein, “overhang” refers to terminal non-base pairing nucleotide(s) resulting from one strand or region extending beyond the terminus of a complementary strand with which the one strand or region forms a duplex. In some embodiments, an overhang comprises one or more unpaired nucleotides extending from a duplex region at the 5′ terminus or 3′ terminus of a dsRNA. In certain embodiments, the overhang is a 3′ or 5′ overhang on the antisense strand or sense strand of a dsRNA. [00104] As used herein, “phosphate analog” refers to a chemical moiety that mimics the electrostatic and/or steric properties of a phosphate group. In some embodiments, a phosphate analog is positioned at the 5′ terminal nucleotide of an oligonucleotide in place of a 5′-phosphate, which is often susceptible to enzymatic removal. In some embodiments, a 5′ phosphate analog contains a phosphatase-resistant linkage. Examples of phosphate analogs include, but are not limited to, 5′ phosphonates, such as 5′ methylene phosphonate (5′-MP) and 5′-(E)-vinylphosphonate (5′-VP). In some embodiments, an oligonucleotide has a phosphate analog at a 4′-carbon position of the sugar (referred to as a “4′-phosphate analog”) at a 5′-terminal nucleotide. An example of a 4′-phosphate analog is oxymethylphosphonate, in which the oxygen atom of the oxymethyl group is bound to the sugar moiety (e.g., at its 4′- carbon) or analog thereof. See, e.g., US Provisional Patent Application Nos.62/383,207 (filed on 2 September 2016) and 62/393,401 (filed on 12 September 2016). Other modifications have been developed for the 5′ end of oligonucleotides (see, e.g., Intl. Patent Application No. WO 2011/133871; US Patent No.8,927,513; and Prakash et al., (2015) NUCLEIC ACIDS RES.43:2993-3011). [00105] As used herein, “reduced expression” of a gene (e.g., STAT3) refers to a decrease in the amount or level of RNA transcript (e.g., STAT3 mRNA) or protein encoded by the gene and/or a decrease in the amount or level of activity of the gene in a cell, a population of cells, a sample, or a subject, when compared to an appropriate reference (e.g., a reference cell, population of cells, sample, or subject). For example, the act of contacting a cell with an oligonucleotide herein (e.g., an oligonucleotide comprising an antisense strand having a nucleotide sequence that is complementary to a nucleotide sequence comprising STAT3 mRNA) may result in a decrease in the amount or level of STAT3 mRNA, protein and/or activity (e.g., via degradation of STAT3 mRNA by the RNAi pathway) when compared to a cell that is not treated with the dsRNA. Similarly, and as used herein, “reducing expression” refers to an act that results in reduced expression of a gene (e.g., STAT3). As used herein, “reduction of STAT3 expression” refers to a decrease in the amount or level of STAT3 mRNA, STAT3 protein and/or STAT3 activity in a cell, a population of cells, a sample or a subject when compared to an appropriate reference (e.g., a reference cell, population of cells, sample, or subject). [00106] As used herein, “region of complementarity” refers to a sequence of nucleotides of a nucleic acid (e.g., a dsRNA) that is sufficiently complementary to an antiparallel sequence of nucleotides to permit hybridization between the two sequences of nucleotides under appropriate hybridization conditions (e.g., in a phosphate buffer, in a cell, etc.). In some embodiments, an oligonucleotide herein comprises a targeting sequence having a region of complementary to a mRNA target sequence. [00107] As used herein, “ribonucleotide” refers to a nucleotide having a ribose as its pentose sugar, which contains a hydroxyl group at its 2′ position. A modified ribonucleotide is a ribonucleotide having one or more modifications or substitutions of atoms other than at the 2′ position, including modifications or substitutions in or of the ribose, phosphate group or base. [00108] As used herein, “RNAi oligonucleotide” refers to either (a) a dsRNA having a sense strand (passenger) and antisense strand (guide), in which the antisense strand or part of the antisense strand is used by the Argonaute 2 (Ago2) endonuclease in the cleavage of a target mRNA or (b) a ss oligonucleotide having a single antisense strand, where that antisense strand (or part of that antisense strand) is used by the Ago2 endonuclease in the cleavage of a target mRNA. [00109] As used herein, “strand” refers to a single, contiguous sequence of nucleotides linked together through internucleotide linkages (e.g., phosphodiester linkages or phosphorothioate linkages). In some embodiments, a strand has two free ends (e.g., a 5′ end and a 3′ end). [00110] As used herein, “subject” means any mammal, including mice, rabbits, non- human primates (NHP), and humans. In one embodiment, the subject is a human or NHP. Moreover, “individual” or “patient” may be used interchangeably with “subject.” [00111] As used herein, “synthetic” refers to a nucleic acid or other molecule that is artificially synthesized (e.g., using a machine (e.g., a solid-state nucleic acid synthesizer)) or that is otherwise not derived from a natural source (e.g., a cell or organism) that normally produces the molecule. [00112] As used herein, “targeting ligand” refers to a molecule or “moiety” (e.g., a carbohydrate, amino sugar, cholesterol, polypeptide, or lipid) that selectively binds to a cognate molecule (e.g., a receptor) of a tissue or cell of interest and/or that is conjugatable to another substance for purposes of targeting the other substance to the tissue or cell of interest. For example, in some embodiments, a targeting ligand may be conjugated to an oligonucleotide for purposes of targeting the oligonucleotide to a specific tissue or cell of interest. In some embodiments, a targeting ligand selectively binds to a cell surface receptor. Accordingly, in some embodiments, a targeting ligand when conjugated to an oligonucleotide facilitates delivery of the oligonucleotide into a particular cell through selective binding to a receptor expressed on the surface of the cell and endosomal internalization by the cell of the complex comprising the oligonucleotide, targeting ligand and receptor. In some embodiments, a targeting ligand is conjugated to an oligonucleotide via a linker that is cleaved following or during cellular internalization such that the oligonucleotide is released from the targeting ligand in the cell. [00113] As used herein, “loop”, “triloop”, or “tetraloop” refers to a loop that increases stability of an adjacent duplex formed by hybridization of flanking sequences of nucleotides. The increase in stability is detectable as an increase in melting temperature (Tm) of an adjacent stem duplex that is higher than the Tm of the adjacent stem duplex expected, on average, from a set of loops of comparable length consisting of randomly selected sequences of nucleotides. For example, a loop (e.g., a tetraloop or triloop) can confer a Tm of at least about 50°C, at least about 55°C, at least about 56°C, at least about 58°C, at least about 60°C, at least about 65°C or at least about 75°C in 10 mM NaHPO4 to a hairpin comprising a duplex of at least 2 base pairs (bp) in length. In some embodiments, a loop (e.g., a tetraloop) may stabilize a bp in an adjacent stem duplex by stacking interactions. In addition, interactions among the nucleotides in a tetraloop include, but are not limited to, non-Watson- Crick base pairing, stacking interactions, hydrogen bonding and contact interactions (Cheong et al., (1990) NATURE 346:680-82; Heus and Pardi (1991) SCIENCE 253:191-94). In some embodiments, a loop comprises or consists of 3 to 6 nucleotides and is typically 4 to 5 nucleotides. In certain embodiments, a loop comprises or consists of 3, 4, 5 or 6 nucleotides, which may or may not be modified (e.g., which may or may not be conjugated to a targeting moiety). In some embodiments, a tetraloop comprises or consists of 3 to 6 nucleotides and is typically 4 to 5 nucleotides. In certain embodiments, a tetraloop comprises or consists of 3, 4, 5 or 6 nucleotides, which may or may not be modified (e.g., which may or may not be conjugated to a targeting moiety). In one embodiment, a loop consisting of 4 nucleotides is a tetraloop. Any nucleotide may be used in the loop (e.g., a tetraloop) and standard IUPAC- IUB symbols for such nucleotides may be used as described in Cornish-Bowden ((1985) NUCLEIC ACIDS RES.13:3021-3030). For example, the letter “N” may be used to mean that any base may be in that position, the letter “R” may be used to show that A (adenine) or G (guanine) may be in that position, and “B” may be used to show that C (cytosine), G (guanine), or T (thymine) may be in that position. Examples of tetraloops include the UNCG family of tetraloops (e.g., UUCG), the GNRA family of tetraloops (e.g., GAAA), and the CUUG tetraloop (Woese et al., (1990) PROC. NATL. ACAD. SCI. USA 87:8467-71; Antao et al., (1991) NUCLEIC ACIDS RES.19:5901-05). Examples of DNA tetraloops include the d(GNNA) family of tetraloops (e.g., d(GTTA), the d(GNRA)) family of tetraloops, the d(GNAB) family of tetraloops, the d(CNNG) family of tetraloops, and the d(TNCG) family of tetraloops (e.g., d(TTCG)). (See, e.g., Nakano et al., (2002) BIOCHEM.41:4281-92; Shinji et al., (2000) NIPPON KAGAKKAI KOEN YOKOSHU 78:731). In some embodiments, the tetraloop is contained within a nicked tetraloop structure. [00114] As used herein, “treat” or “treating” refers to the act of providing care to a subject in need thereof, for example, by administering a therapeutic agent (e.g., an oligonucleotide herein) to the subject, for purposes of improving the health and/or well-being of the subject with respect to an existing condition (e.g., a disease, disorder) or to prevent or decrease the likelihood of the occurrence of a condition. In some embodiments, treatment involves reducing the frequency or severity of at least one sign, symptom or contributing factor of a condition (e.g., disease, disorder) experienced by a subject. [00115] As used herein, the term "tumor microenvironment" relates to the cellular environment in which any given tumor exists, including the tumor stroma, surrounding blood vessels, immune cells, fibroblasts, other cells, signaling molecules, and the ECM. It is understood that the tumor microenvironment harbors and/or surrounds the tumor cells with which it interacts. Methods of Use Combination of STAT3 Oligonucleotide and PD-L1 Inhibitors [00116] In some embodiments, the disclosure provides STAT3 oligonucleotides for use, or adaptable for use, to treat a subject (e.g., a human having a disease, disorder or condition associated with STAT3 expression) that has received or is receiving a PD-L1 inhibitor. [00117] In some embodiments, methods described herein comprise selecting a subject having a disease, disorder or condition associated with STAT3 expression and/or PD-L1 expression or is predisposed to the same. In some instances, the methods can include selecting an individual having a marker for a disease associated with STAT3 expression and/or PD-L1 expression such as cancer or other chronic lymphoproliferative disorders. [00118] Likewise, and as detailed herein, the methods also may include steps such as measuring or obtaining a baseline value for a marker of STAT3 expression and/or PD-L1 expression, and then comparing such obtained value to one or more other baseline values or values obtained after being administered the oligonucleotide to assess the effectiveness of treatment. [00119] In some embodiments, the disclosure provides methods of treating a subject having, suspected of having, or at risk of developing a disease, disorder, or condition with a STAT3 oligonucleotide herein, wherein the subject has received or is receiving a PD-L1 inhibitor. In some embodiments, the disclosure provides methods of treating a subject having, suspected of having, or at risk of developing a disease, disorder, or condition with a PD-L1 inhibitor described herein, wherein the subject has received or is receiving a STAT3 oligonucleotide described herein. [00120] In some aspects, the disclosure provides methods of treating or attenuating the onset or progression of a disease, disorder or condition associated with STAT3 expression using a STAT3 oligonucleotide herein in combination with a PD-L1 inhibitor. In other aspects, the disclosure provides methods to achieve one or more therapeutic benefits in a subject having a disease, disorder or condition associated with STAT3 expression using a STAT3 oligonucleotide herein in combination with a PD-L1 inhibitor. In some embodiments of the methods herein, the subject is treated by administering a therapeutically effective amount of a STAT3 oligonucleotide herein in combination with a PD-L1 inhibitor. In some embodiments of the methods herein, the subject is treated by administering a therapeutically effective amount of a STAT3 oligonucleotide herein to a subject that has received or is receiving a PD-L1 inhibitor. In some embodiments of the methods herein, the subject is treated by administering a therapeutically effective amount of a PD-L1 inhibitor to a subject that has received or is receiving a STAT3 oligonucleotide herein. In some embodiments, the subject is treated therapeutically. In some embodiments, the subject is treated prophylactically. [00121] In some embodiments of the methods herein, one or more STAT3 oligonucleotides herein, or a pharmaceutical composition comprising one or more STAT3 oligonucleotides, is administered to a subject having a disease, disorder or condition associated with STAT3 expression that has received or is receiving a PD-L1 inhibitor, such that STAT3 expression is reduced in the subject, thereby treating the subject. In some embodiments, an amount or level of STAT3 mRNA is reduced in the subject. In some embodiments, an amount or level of STAT3 and/or protein is reduced in the subject. In some embodiments of the methods herein, one or more STAT3 oligonucleotides herein, or a pharmaceutical composition comprising one or more STAT3 oligonucleotides, is administered to a subject having a disease, disorder or condition associated with STAT3 expression that has received or is receiving a PD-L1 inhibitor such that STAT3 expression and PD-L1 signaling is reduced in the subject, thereby treating the subject. In some embodiments, an amount or level of STAT3 mRNA and PD-L1 signaling is reduced in the subject. In some embodiments, an amount or level of STAT3 and/or protein is reduced in the subject and PD-L1 signaling is reduced in the subject. [00122] In some embodiments, a therapeutically effective amount of a STAT3 oligonucleotide and/or PD-L1 inhibitor is administered to a subject. A therapeutically acceptable amount may be an amount that can therapeutically treat a disease or disorder. The appropriate dosage for any one subject will depend on certain factors, including the subject′s size, body surface area, age, the particular composition to be administered, the active ingredient(s) in the composition, time and route of administration, general health, and other drugs being administered concurrently. [00123] In some embodiments, a subject is administered any one of the compositions herein either enterally (e.g., orally, by gastric feeding tube, by duodenal feeding tube, via gastrostomy or rectally), parenterally (e.g., subcutaneous injection, intravenous injection or infusion, intra-arterial injection or infusion, intraosseous infusion, intramuscular injection, intracerebral injection, intracerebroventricular injection, intrathecal), topically (e.g., epicutaneous, inhalational, via eye drops, or through a mucous membrane), or by direct injection into a target organ (e.g., the liver of a subject). Typically, oligonucleotides herein are administered intravenously or subcutaneously. [00124] As a non-limiting set of examples, the oligonucleotides herein would typically be administered quarterly (once every three months), bi-monthly (once every two months), monthly or weekly. For example, the oligonucleotides may be administered every week or at intervals of two, or three weeks. Alternatively, the oligonucleotides may be administered daily. In some embodiments, a subject is administered one or more loading doses of the oligonucleotide followed by one or more maintenance doses of the oligonucleotide. [00125] In some embodiments, a PD-L1 inhibitor (e.g., an anti-PD-L1 antibody) herein is administered quarterly (once every three months), bi-monthly (once every two months), monthly or weekly. For example, the inhibitor is administered every week or at intervals of two, or three weeks. Alternatively, the inhibitor is administered daily. [00126] In some embodiments the oligonucleotides herein are administered in combination with a PD-L1 inhibitor. In some embodiments the oligonucleotide and inhibitor are administered in combination concurrently, sequentially (in any order), or intermittently. For example, the oligonucleotide and inhibitor may be co-administered concurrently. Alternatively, the oligonucleotide may be administered and followed any amount of time later (e.g., one hour, one day, one week or one month) by the administration of the inhibitor, or vice versa. [00127] In some embodiments, the subject to be treated is a human or non-human primate or other mammalian subject. Other exemplary subjects include domesticated animals such as dogs and cats; livestock such as horses, cattle, pigs, sheep, goats, and chickens; and animals such as mice, rats, guinea pigs, and hamsters. In some embodiments, the disclosure provides a method of treating cancer in a subject that has received or is receiving a PD-L1 inhibitor, the method comprising administering an RNAi oligonucleotide comprising a sense strand and an antisense strand, wherein the sense strand and antisense strand comprise nucleotide sequences selected from the group consisting of: (a) SEQ ID NOs: 9 and 10, respectively; (b) SEQ ID NOs: 37 and 38, respectively; (c) SEQ ID NOs: 65 and 66, respectively; and (d) SEQ ID NOs: 69 and 70, respectively, thereby treating cancer in the subject. [00128] In some embodiments, the disclosure provides a method of treating cancer in a subject that has received or is receiving a PD-L1 inhibitor, the method comprising administering an RNAi oligonucleotide comprising a sense strand and an antisense strand, wherein the sense strand and antisense strand comprise nucleotide sequences selected from the group consisting of: (a) SEQ ID NOs: 9 and 10, respectively; (b) SEQ ID NOs: 37 and 38, respectively; (c) SEQ ID NOs: 65 and 66, respectively; and (d) SEQ ID NOs: 69 and 70, respectively, and wherein a lipid moiety is conjugated to a nucleotide of the sense strand, thereby treating cancer in the subject. [00129] In some embodiments, the disclosure provides a method of treating cancer in a subject that has received or is receiving a PD-L1 inhibitor, the method comprising administering an RNAi oligonucleotide comprising a sense strand and an antisense strand, wherein the sense strand comprises the nucleotide sequence of SEQ ID NO: 9 and antisense strand comprises the nucleotide sequence of SEQ ID NO: 10, thereby treating cancer in the subject. [00130] In some embodiments, the disclosure provides a method of treating cancer in a subject that has received or is receiving a PD-L1 inhibitor, the method comprising administering an RNAi oligonucleotide comprising a sense strand and an antisense strand, wherein the sense strand comprises the nucleotide sequence of SEQ ID NO: 9 and antisense strand comprises the nucleotide sequence of SEQ ID NO: 10, and wherein a lipid moiety is conjugated to a nucleotide of the sense strand, thereby treating cancer in the subject. [00131] In some embodiments, the disclosure provides a method of treating cancer in a subject that has received or is receiving a PD-L1 inhibitor, the method comprising administering an RNAi oligonucleotide comprising a sense strand and an antisense strand, wherein the sense strand comprises the nucleotide sequence of SEQ ID NO: 37 and antisense strand comprises the nucleotide sequence of SEQ ID NO: 38, thereby treating cancer in the subject. [00132] In some embodiments, the disclosure provides a method of treating cancer in a subject that has received or is receiving a PD-L1 inhibitor, the method comprising administering an RNAi oligonucleotide comprising a sense strand and an antisense strand, wherein the sense strand comprises the nucleotide sequence of SEQ ID NO: 37 and antisense strand comprises the nucleotide sequence of SEQ ID NO: 38, and wherein a lipid moiety is conjugated to a nucleotide of the sense strand, thereby treating cancer in the subject. [00133] In some embodiments, the disclosure provides a method of treating cancer in a subject that has received or is receiving a PD-L1 inhibitor, the method comprising administering an RNAi oligonucleotide comprising a sense strand and an antisense strand, wherein the sense strand comprises the nucleotide sequence of SEQ ID NO: 65 and antisense strand comprises the nucleotide sequence of SEQ ID NO: 66, thereby treating cancer in the subject. [00134] In some embodiments, the disclosure provides a method of treating cancer in a subject that has received or is receiving a PD-L1 inhibitor, the method comprising administering an RNAi oligonucleotide comprising a sense strand and an antisense strand, wherein the sense strand comprises the nucleotide sequence of SEQ ID NO: 65 and antisense strand comprises the nucleotide sequence of SEQ ID NO: 66, and wherein a lipid moiety is conjugated to a nucleotide of the sense strand, thereby treating cancer in the subject. [00135] In some embodiments, the disclosure provides a method of treating cancer in a subject that has received or is receiving a PD-L1 inhibitor, the method comprising administering an RNAi oligonucleotide comprising a sense strand and an antisense strand, wherein the sense strand comprises the nucleotide sequence of SEQ ID NO: 69 and antisense strand comprises the nucleotide sequence of SEQ ID NO: 70, thereby treating cancer in the subject. [00136] In some embodiments, the disclosure provides a method of treating cancer in a subject that has received or is receiving a PD-L1 inhibitor, the method comprising administering an RNAi oligonucleotide comprising a sense strand and an antisense strand, wherein the sense strand comprises the nucleotide sequence of SEQ ID NO: 69 and antisense strand comprises the nucleotide sequence of SEQ ID NO: 70, and wherein a lipid moiety is conjugated to a nucleotide of the sense strand, thereby treating cancer in the subject. [00137] In some embodiments, the disclosure provides a method of treating cancer in a subject that has received or is receiving an RNAi oligonucleotide, the method comprising administering a PD-L1 inhibitor, wherein the oligonucleotide comprises a sense strand and an antisense strand, wherein the sense strand and antisense strand comprise nucleotide sequences selected from the group consisting of: (a) SEQ ID NOs: 9 and 10, respectively; (b) SEQ ID NOs: 37 and 38, respectively; (c) SEQ ID NOs: 65 and 66, respectively; and (d) SEQ ID NOs: 69 and 70, respectively, thereby treating cancer in the subject. [00138] In some embodiments, the disclosure provides a method of treating cancer in a subject that has received or is receiving an RNAi oligonucleotide, the method comprising administering a PD-L1 inhibitor, wherein the oligonucleotide comprises a sense strand and an antisense strand, wherein the sense strand and antisense strand comprise nucleotide sequences selected from the group consisting of: (a) SEQ ID NOs: 9 and 10, respectively; (b) SEQ ID NOs: 37 and 38, respectively; (c) SEQ ID NOs: 65 and 66, respectively; and (d) SEQ ID NOs: 69 and 70, respectively, and wherein a lipid moiety is conjugated to a nucleotide of the sense strand, thereby treating cancer in the subject. [00139] In some embodiments, the disclosure provides a method of treating cancer in a subject that has received or is receiving an RNAi oligonucleotide, the method comprising administering a PD-L1 inhibitor, wherein the oligonucleotide comprises a sense strand and an antisense strand, wherein the sense strand comprises the nucleotide sequence of SEQ ID NO: 9 and antisense strand comprises the nucleotide sequence of SEQ ID NO: 10, thereby treating cancer in the subject. [00140] In some embodiments, the disclosure provides a method of treating cancer in a subject that has received or is receiving an RNAi oligonucleotide, the method comprising administering a PD-L1 inhibitor, wherein the oligonucleotide comprises a sense strand and an antisense strand, wherein the sense strand comprises the nucleotide sequence of SEQ ID NO: 9 and antisense strand comprises the nucleotide sequence of SEQ ID NO: 10, and wherein a lipid moiety is conjugated to a nucleotide of the sense strand, thereby treating cancer in the subject. [00141] In some embodiments, the disclosure provides a method of treating cancer in a subject that has received or is receiving an RNAi oligonucleotide, the method comprising administering a PD-L1 inhibitor, wherein the oligonucleotide comprises a sense strand and an antisense strand, wherein the sense strand comprises the nucleotide sequence of SEQ ID NO: 37 and antisense strand comprises the nucleotide sequence of SEQ ID NO: 38, thereby treating cancer in the subject. [00142] In some embodiments, the disclosure provides a method of treating cancer in a subject that has received or is receiving an RNAi oligonucleotide, the method comprising administering a PD-L1 inhibitor, wherein the oligonucleotide comprises a sense strand and an antisense strand, wherein the sense strand comprises the nucleotide sequence of SEQ ID NO: 37 and antisense strand comprises the nucleotide sequence of SEQ ID NO: 38, and wherein a lipid moiety is conjugated to a nucleotide of the sense strand, thereby treating cancer in the subject. [00143] In some embodiments, the disclosure provides a method of treating cancer in a subject that has received or is receiving an RNAi oligonucleotide, the method comprising administering a PD-L1 inhibitor, wherein the oligonucleotide comprises a sense strand and an antisense strand, wherein the sense strand comprises the nucleotide sequence of SEQ ID NO: 65 and antisense strand comprises the nucleotide sequence of SEQ ID NO: 66, thereby treating cancer in the subject. [00144] In some embodiments, the disclosure provides a method of treating cancer in a subject that has received or is receiving an RNAi oligonucleotide, the method comprising administering a PD-L1 inhibitor, wherein the oligonucleotide comprises a sense strand and an antisense strand, wherein the sense strand comprises the nucleotide sequence of SEQ ID NO: 65 and antisense strand comprises the nucleotide sequence of SEQ ID NO: 66, and wherein a lipid moiety is conjugated to a nucleotide of the sense strand, thereby treating cancer in the subject. [00145] In some embodiments, the disclosure provides a method of treating cancer in a subject that has received or is receiving an RNAi oligonucleotide, the method comprising administering a PD-L1 inhibitor, wherein the oligonucleotide comprises a sense strand and an antisense strand, wherein the sense strand comprises the nucleotide sequence of SEQ ID NO: 69 and antisense strand comprises the nucleotide sequence of SEQ ID NO: 70, thereby treating cancer in the subject. [00146] In some embodiments, the disclosure provides a method of treating cancer in a subject that has received or is receiving an RNAi oligonucleotide, the method comprising administering a PD-L1 inhibitor, wherein the oligonucleotide comprises a sense strand and an antisense strand, wherein the sense strand comprises the nucleotide sequence of SEQ ID NO: 69 and antisense strand comprises the nucleotide sequence of SEQ ID NO: 70, and wherein a lipid moiety is conjugated to a nucleotide of the sense strand, thereby treating cancer in the subject. [00147] In some embodiments, the disclosure provides a method of treating a disease, disorder or condition associated with activated STAT3 expression, comprising administering to a subject in need thereof an RNAi oligonucleotide, wherein the subject has received or is receiving a PD-L1 inhibitor, wherein the oligonucleotide comprises a sense strand and an antisense strand, wherein the sense strand and antisense strand comprise nucleotide sequences selected from the group consisting of: (a) SEQ ID NOs: 9 and 10, respectively; (b) SEQ ID NOs: 37 and 38, respectively; (c) SEQ ID NOs: 65 and 66, respectively; and (d) SEQ ID NOs: 69 and 70, respectively, thereby treating cancer in the subject. [00148] In some embodiments, the disclosure provides a method of treating a disease, disorder or condition associated with activated STAT3 expression, comprising administering to a subject in need thereof an RNAi oligonucleotide, wherein the subject has received or is receiving a PD-L1 inhibitor, wherein the oligonucleotide comprises a sense strand and an antisense strand, wherein the sense strand and antisense strand comprise nucleotide sequences selected from the group consisting of: (a) SEQ ID NOs: 9 and 10, respectively; (b) SEQ ID NOs: 37 and 38, respectively; (c) SEQ ID NOs: 65 and 66, respectively; and (d) SEQ ID NOs: 69 and 70, respectively, and wherein a lipid moiety is conjugated to a nucleotide of the sense strand, thereby treating cancer in the subject. [00149] In some embodiments, the disclosure provides a method of treating a disease, disorder or condition associated with activated STAT3 expression, comprising administering to a subject in need thereof an RNAi oligonucleotide, wherein the subject has received or is receiving a PD-L1 inhibitor, wherein the oligonucleotide comprises a sense strand and an antisense strand, wherein the sense strand comprises the nucleotide sequence of SEQ ID NO: 9 and antisense strand comprises the nucleotide sequence of SEQ ID NO: 10, thereby treating cancer in the subject. [00150] In some embodiments, the disclosure provides a method of treating a disease, disorder or condition associated with activated STAT3 expression, comprising administering to a subject in need thereof an RNAi oligonucleotide, wherein the subject has received or is receiving a PD-L1 inhibitor, wherein the oligonucleotide comprises a sense strand and an antisense strand, wherein the sense strand comprises the nucleotide sequence of SEQ ID NO: 9 and antisense strand comprises the nucleotide sequence of SEQ ID NO: 10, and wherein a lipid moiety is conjugated to a nucleotide of the sense strand, thereby treating cancer in the subject. [00151] In some embodiments, the disclosure provides a method of treating a disease, disorder or condition associated with activated STAT3 expression, comprising administering to a subject in need thereof an RNAi oligonucleotide, wherein the subject has received or is receiving a PD-L1 inhibitor, wherein the oligonucleotide comprises a sense strand and an antisense strand, wherein the sense strand comprises the nucleotide sequence of SEQ ID NO: 37 and antisense strand comprises the nucleotide sequence of SEQ ID NO: 38, thereby treating cancer in the subject. [00152] In some embodiments, the disclosure provides a method of treating a disease, disorder or condition associated with activated STAT3 expression, comprising administering to a subject in need thereof an RNAi oligonucleotide, wherein the subject has received or is receiving a PD-L1 inhibitor, wherein the oligonucleotide comprises a sense strand and an antisense strand, wherein the sense strand comprises the nucleotide sequence of SEQ ID NO: 37 and antisense strand comprises the nucleotide sequence of SEQ ID NO: 38, and wherein a lipid moiety is conjugated to a nucleotide of the sense strand, thereby treating cancer in the subject. [00153] In some embodiments, the disclosure provides a method of treating a disease, disorder or condition associated with activated STAT3 expression, comprising administering to a subject in need thereof an RNAi oligonucleotide, wherein the subject has received or is receiving a PD-L1 inhibitor, wherein the oligonucleotide comprises a sense strand and an antisense strand, wherein the sense strand comprises the nucleotide sequence of SEQ ID NO: 65 and antisense strand comprises the nucleotide sequence of SEQ ID NO: 66, thereby treating cancer in the subject. [00154] In some embodiments, the disclosure provides a method of treating a disease, disorder or condition associated with activated STAT3 expression, comprising administering to a subject in need thereof an RNAi oligonucleotide, wherein the subject has received or is receiving a PD-L1 inhibitor, wherein the oligonucleotide comprises a sense strand and an antisense strand, wherein the sense strand comprises the nucleotide sequence of SEQ ID NO: 65 and antisense strand comprises the nucleotide sequence of SEQ ID NO: 66, and wherein a lipid moiety is conjugated to a nucleotide of the sense strand, thereby treating cancer in the subject. [00155] In some embodiments, the disclosure provides a method of treating a disease, disorder or condition associated with activated STAT3 expression, comprising administering to a subject in need thereof an RNAi oligonucleotide, wherein the subject has received or is receiving a PD-L1 inhibitor, wherein the oligonucleotide comprises a sense strand and an antisense strand, wherein the sense strand comprises the nucleotide sequence of SEQ ID NO: 69 and antisense strand comprises the nucleotide sequence of SEQ ID NO: 70, thereby treating cancer in the subject. [00156] In some embodiments, the disclosure provides a method of treating a disease, disorder or condition associated with activated STAT3 expression, comprising administering to a subject in need thereof an RNAi oligonucleotide, wherein the subject has received or is receiving a PD-L1 inhibitor, wherein the oligonucleotide comprises a sense strand and an antisense strand, wherein the sense strand comprises the nucleotide sequence of SEQ ID NO: 69 and antisense strand comprises the nucleotide sequence of SEQ ID NO: 70, and wherein a lipid moiety is conjugated to a nucleotide of the sense strand, thereby treating cancer in the subject. [00157] In some embodiments, the methods of the disclosure increase expression of immune activation genes. In some embodiments, administering an RNAi oligonucleotide described herein with a PD-L1 inhibitor increases expression of immune activation genes. In some embodiments, the methods described herein increase expression of one or more of CD8B1, IL2Rα, TNFRSF9, CCL19, CCL27a, CD40Ig, CXCR5, CD44, STAT4, GZMB, PFR1, FASL, and HMGB1. [00158] In some embodiments, the methods of the disclosures decrease expression of immuno-suppressive genes. In some embodiments, administering an RNAi oligonucleotide described herein with a PD-L1 inhibitor decreases expression of immune-suppressive genes. In some embodiments, the methods described herein decrease expression of one or more of CD274, LAG3, TIGIT, PDSD1IG2, CD163, PIK3R5, FOXP3, TGFBR2, IDO1, CD200R1, CCL22, CCL8, CCR9, IL10, IL33, CXCL3, CSF2RD, TNFRSF18, MMP9, LOX12, FGF18, and FAP. Cancers [00159] In some embodiments, the STAT3 oligonucleotide and PD-L1 inhibitor target are used to treat a cancer or a tumor. In some embodiments, the tumor is a primary tumor. In some embodiments, the tumor is a metastatic tumor. In some embodiments, the tumor is a refractory tumor. In some embodiments, the tumor is a Stage I, Stage II, Stage III, or Stage IV tumor. In some embodiments, the tumor is a solid-tumor. Solid-tumors refer to conditions where the cancer forms a mass [00160] In some embodiments, the cancer is a thyroid cancer, papillary thyroid carcinoma, head and neck cancer, liver cancer, colorectal cancer, pancreatic cancer, breast cancer, ovarian cancer, lung cancer, carcinoma, blastoma, medulloblastoma, retinoblastoma, sarcoma, liposarcoma, synovial cell sarcoma, neuroendocrine tumors, carcinoid tumors, gastrinoma, islet cell cancer, mesothelioma, schwannoma, acoustic neuroma, meningioma, adenocarcinoma, lymphoid malignancies, squamous cell cancer, epithelial squamous cell cancer, small-cell lung cancer (SCLC), non-small cell lung cancer (NSCLC), adenocarcinoma of the lung, squamous carcinoma of the lung, cancer of the peritoneum, hepatocellular cancer, gastric or stomach cancer, gastrointestinal cancer, glioblastoma, cervical cancer, bladder cancer, hepatoma, metastatic breast cancer, colon cancer, rectal cancer, endometrial or uterine carcinoma, salivary gland carcinoma, kidney or renal cancer, prostate cancer, vulval cancer, hepatic carcinoma, anal carcinoma, penile carcinoma, Merkel cell cancer, testicular cancer, esophageal cancer, or tumors of the biliary tract. In some embodiments, the cancer is refractory to anti-PD1, anti-PDL1 and/or anti-CTLA4 therapy. In some embodiments, the cancer is a pancreatic cancer or lung cancer. In some embodiments, the cancer comprises tumors with immunosuppressive tumor microenvironments. In some embodiments, the cancer is resistant to immune checkpoint therapy. In some embodiments, the cancer is partially resistant to immune checkpoint therapy. In some embodiments, the cancer is sensitive to immune checkpoint therapy. [00161] In some embodiments, the STAT3 oligonucleotide and PD-L1 inhibitor reduces tumor volume. Tumor volume is measured using methods know to one of skill in the art. For example, extracted tumors are measured manually using calipers. Other methods include imagine methods such as ultrasound and MRI. In some embodiments, the oligonucleotide conjugate reduces tumor volume by at least 5%, at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, or at least 90% compared to an untreated tumor. Oligonucleotide Inhibitors of STAT3 [00162] In some aspects, the disclosure provides, inter alia, oligonucleotides that reduce or inhibit STAT3 expression. In some embodiments, an oligonucleotide that inhibits STAT3 expression herein is targeted to a STAT3 mRNA. The sequence of human STAT3 mRNA (NM_001369512.1) is set forth as SEQ ID NO: 85 or NM_139276.3 (SEQ ID NO: 1217). STAT3 is a known target for conventional cancer therapies. [00163] The tolerogenic activities of MDSCs are controlled by an oncogenic transcription factor, signal transducer and activator of transcription 3 (STAT3) (Su et al., INT J. MOL SCI (2018) 19(6): 1803). STAT3 is also known to be highly expressed across a range of cancer types and in in vitro and in vivo preclinical models (Huynh et al., NAT. REV. CANCER (2019) 19: 82-96). The inhibition of STAT3 leads to the selective apoptosis of tumor cells and tumor growth inhibition through modulation of downstream target genes (Wang et al., INTERNATIONAL JOURNAL OF BIOLOGICAL SCIENCES, 15(3): 668–79 (2019)). STAT3 is of particular interest in immuno-oncology due to its well documented contributions to an immunosuppressive tumor microenvironment. STAT3 contributes to an immunosuppressive tumor microenvironment by upregulating the inhibitory receptor expressed by T-cells, and via expression of its ligand (PD-1/PD-L1), through increased secretion of IFNɣ (Bu et al., JOURNAL OF DENTAL RESEARCH, 96(9): 1027–34 (2017)). It has long been known that inhibition of STAT3 signaling in antigen presenting cells (APCs) results in priming of antigen-specific CD4+ T cells in response to otherwise tolerogenic stimuli (Cheng et al., IMMUNITY, 19: 425–36 (2003)). In addition, phosphorylated STAT3 on MDSCs directly contributes to the modulation of the suppressive tumor microenvironment by regulating suppressive components such as the amino acid arginine, through transcriptional control (Vasques-Dunndel et al., J. CLIN. INVEST., 15(3): 668–79 (2013)). Over the years several methodologies have been explored to therapeutically target STAT3. While direct targeting of the protein is attractive, the true target is a protein-protein interaction that has been held up as an example of an ‘undruggable’ target due historical data showing that multiple classes of compounds have failed to effectively inhibit its activity (Lau et al., CANCERS (2019) 11(11): 1681, Zou et al., MOL CANCER (2020) 19: 145). In addition, ubiquitous expression of STAT3 across several tissues have led to concerns about severe on-target toxicities (Wong et al., EXPERT OPINION ON INVESTIGATIONAL DRUGS, 26 (8):883-87 (2017), (Kortylewski et al., CANCER IMMUNOL IMMUNOTHER (2017) 66(8): 979-88). [00164] In some embodiments, reduction of STAT3 expression can be determined by an appropriate assay or technique to evaluate one or more properties or characteristics of a cell or population of cells associated with STAT3 expression (e.g., using an STAT3 expression biomarker) or by an assay or technique that evaluates molecules that are directly indicative of STAT3 expression (e.g., STAT3 mRNA or STAT3 protein). In some embodiments, the extent to which an oligonucleotide herein reduces STAT3 expression is evaluated by comparing STAT3 expression in a cell or population of cells contacted with the oligonucleotide to an appropriate control (e.g., an appropriate cell or population of cells not contacted with the oligonucleotide or contacted with a control oligonucleotide). In some embodiments, an appropriate control level of mRNA expression into protein, after delivery of a RNAi molecule may be a predetermined level or value, such that a control level need not be measured every time. The predetermined level or value can take a variety of forms. In some embodiments, a predetermined level or value can be single cut-off value, such as a median or mean. [00165] In some embodiments, administration of an oligonucleotide herein results in a reduction in STAT3 expression in a cell or population of cells. In some embodiments, the reduction in STAT3 or STAT3 expression is about 1% or lower, about 5% or lower, about 10% or lower, about 15% or lower, about 20% or lower, about 25% or lower, about 30% or lower, about 35% or lower, about 40% or lower, about 45% or lower, about 50% or lower, about 55% or lower, about 60% or lower, about 70% or lower, about 80% or lower, or about 90% or lower when compared with an appropriate control level of mRNA. The appropriate control level may be a level of mRNA expression and/or protein translation in a cell or population of cells that has not been contacted with an oligonucleotide herein. In some embodiments, the effect of delivery of an oligonucleotide to a cell according to a method herein is assessed after a finite period. For example, levels of mRNA may be analyzed in a cell at least about 8 hours, about 12 hours, about 18 hours, about 24 hours; or at least about 1, 2, 3, 4, 5, 6, 7 or even up to 14 days after introduction of the oligonucleotide into the cell. [00166] In some embodiments, an oligonucleotide is delivered in the form of a transgene that is engineered to express in a cell the oligonucleotide or strands comprising the oligonucleotide (e.g., its sense and antisense strands). In some embodiments, an oligonucleotide is delivered using a transgene engineered to express any oligonucleotide disclosed herein. Transgenes may be delivered using viral vectors (e.g., adenovirus, retrovirus, vaccinia virus, poxvirus, adeno-associated virus, or herpes simplex virus) or non- viral vectors (e.g., plasmids or synthetic mRNAs). In some embodiments, transgenes can be injected directly to a subject. STAT3 Target Sequences [00167] In some embodiments, the oligonucleotide is targeted to a target sequence comprising a STAT3 mRNA. In some embodiments, the oligonucleotide, or a portion, fragment, or strand thereof (e.g., an antisense strand or a guide strand of a dsRNA) binds or anneals to a target sequence comprising a STAT3 mRNA, thereby inhibiting STAT3 expression. In some embodiments, the oligonucleotide is targeted to a STAT3 target sequence for the purpose of inhibiting STAT3 expression in vivo. In some embodiments, the amount or extent of inhibition of STAT3 expression by an oligonucleotide targeted to a STAT3 target sequence correlates with the potency of the oligonucleotide. In some embodiments, the amount or extent of inhibition of STAT3 expression by an oligonucleotide targeted to a STAT3 target sequence correlates with the amount or extent of therapeutic benefit in a subject or patient having a disease, disorder or condition associated with the expression of STAT3 treated with the oligonucleotide. [00168] Through examination of the nucleotide sequence of mRNAs encoding STAT3, including mRNAs of multiple different species (e.g., human, cynomolgus monkey, mouse, and rat; see, e.g., Example 6) and as a result of in vitro and in vivo testing (see, e.g., Example 7 and Example 8), it has been discovered that certain nucleotide sequences of STAT3 mRNA are more amenable than others to oligonucleotide-based inhibition and are thus useful as target sequences for the oligonucleotides herein. In some embodiments, a sense strand of an oligonucleotide (e.g., a dsRNA) described herein comprises a STAT3 target sequence. In some embodiments, a portion or region of the sense strand of a dsRNA described herein comprises a STAT3 target sequence. In some embodiments, a STAT3 mRNA target sequence comprises, or consists of, a sequence of SEQ ID NO 85. In some embodiments, a STAT3 mRNA target sequence comprises, or consists of, a sequence of SEQ ID NO: 1217. In some embodiments, a STAT3 mRNA target sequence comprises, or consists of, a sequence of any one of SEQ ID NOs: 89-280. In some embodiments, a STAT3 mRNA target sequence comprises, or consists of, the sequence set forth in SEQ ID NO: 108. In some embodiments, a STAT3 mRNA target sequence comprises, or consists of, the sequence set forth in SEQ ID NO: 140. In some embodiments, a STAT3 mRNA target sequence comprises, or consists of, the sequence set forth in SEQ ID NO: 141. In some embodiments, a STAT3 mRNA target sequence comprises, or consists of, the sequence set forth in SEQ ID NO: 147. STAT3 Targeting Sequences [00169] In some embodiments, the oligonucleotides herein have regions of complementarity to STAT3 mRNA (e.g., within a target sequence of STAT3 mRNA) for purposes of targeting the mRNA in cells and reducing or inhibiting its expression. In some embodiments, the oligonucleotides herein comprise a STAT3 targeting sequence (e.g., an antisense strand or a guide strand of a dsRNA) having a region of complementarity that binds or anneals to a STAT3 target sequence by complementary (Watson-Crick) base pairing. The targeting sequence or region of complementarity is generally of a suitable length and base content to enable binding or annealing of the oligonucleotide (or a strand thereof) to a STAT3 mRNA for purposes of inhibiting its expression. In some embodiments, the targeting sequence or region of complementarity is at least about 12, at least about 13, at least about 14, at least about 15, at least about 16, at least about 17, at least about 18, at least about 19, at least about 20, at least about 21, at least about 22, at least about 23, at least about 24, at least about 25, at least about 26, at least about 27, at least about 28, at least about 29 or at least about 30 nucleotides in length. In some embodiments, the targeting sequence or region of complementarity is about 12 to about 30 (e.g., 12 to 30, 12 to 22, 15 to 25, 17 to 21, 18 to 27, 19 to 27, or 15 to 30) nucleotides in length. In some embodiments, the targeting sequence or region of complementarity is about 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29 or 30 nucleotides in length. In some embodiments, the targeting sequence or region of complementarity is 18 nucleotides in length. In some embodiments, the targeting sequence or region of complementarity is 19 nucleotides in length. In some embodiments, the targeting sequence or region of complementarity is 20 nucleotides in length. In some embodiments, the targeting sequence or region of complementarity is 21 nucleotides in length. In some embodiments, the targeting sequence or region of complementarity is 22 nucleotides in length. In some embodiments, the targeting sequence or region of complementarity is 23 nucleotides in length. In some embodiments, the targeting sequence or region of complementarity is 24 nucleotides in length. In some embodiments, an oligonucleotide comprises a target sequence or region of complementarity complementary to a sequence of any one of SEQ ID NOs: 89-280 , and the targeting sequence or region of complementarity is 18 nucleotides in length. In some embodiments, an oligonucleotide comprises a target sequence or region of complementarity complementary to a sequence of any one of SEQ ID NOs: 89-280, and the targeting sequence or region of complementarity is 19 nucleotides in length. In some embodiments, an oligonucleotide comprises a target sequence or region of complementarity complementary to a sequence of any one of SEQ ID NOs: 473-664, and the targeting sequence or region of complementarity is 20 nucleotides in length. In some embodiments, an oligonucleotide comprises a targeting sequence or region of complementarity complementary to a sequence of any one of SEQ ID NOs: 473-664, and the targeting sequence or region of complementarity is 21 nucleotides in length. In some embodiments, an oligonucleotide comprises a targeting sequence or region of complementarity complementary to a sequence of any one of SEQ ID NOs: 473-664, and the targeting sequence or region of complementarity is 22 nucleotides in length. In some embodiments, an oligonucleotide comprises a targeting sequence or region of complementarity complementary to a sequence of any one of SEQ ID NOs: 473-664, and the targeting sequence or region of complementarity is 23 nucleotides in length. In some embodiments, an oligonucleotide comprises a targeting sequence or region of complementarity complementary to a sequence of any one of SEQ ID NOs: 473-664 and the targeting sequence or region of complementarity is 24 nucleotides in length. [00170] In some embodiments, an oligonucleotide herein comprises a targeting sequence or a region of complementarity (e.g., an antisense strand or a guide strand of a double-stranded oligonucleotide) that is fully complementary to a STAT3 target sequence. In some embodiments, the targeting sequence or region of complementarity is partially complementary to a STAT3 target sequence. In some embodiments, the oligonucleotide comprises a targeting sequence or region of complementarity that is fully complementary to a sequence of STAT3 or STAT3. In some embodiments, the oligonucleotide comprises a targeting sequence or region of complementarity that is partially complementary to a sequence of STAT3 or STAT3. [00171] In some embodiments, the oligonucleotide comprises a targeting sequence or region of complementarity that is fully complementary to a sequence of any one of SEQ ID NOs: 89-280. In some embodiments, the oligonucleotide comprises a targeting sequence or region of complementarity that is fully complementary to the sequence set forth in SEQ ID NOs: 108, 140, 141, and 147. In some embodiments, the oligonucleotide comprises a targeting sequence or region of complementarity that is partially complementary to a sequence of any one of SEQ ID NOs: 89-280. In some embodiments, the oligonucleotide comprises a targeting sequence or region of complementarity that is partially complementary to the sequence set forth in SEQ ID NOs: 108, 140, 141, and 147. [00172] In some embodiments, the oligonucleotide herein comprises a targeting sequence or region of complementarity that is complementary to a contiguous sequence of nucleotides comprising a STAT3 mRNA, wherein the contiguous sequence of nucleotides is about 12 to about 30 nucleotides in length (e.g., 12 to 30, 12 to 28, 12 to 26, 12 to 24, 12 to 20, 12 to 18, 12 to 16, 14 to 22, 16 to 20, 18 to 20 or 18 to 19 nucleotides in length). In some embodiments, the oligonucleotide comprises a targeting sequence or region of complementarity that is complementary to a contiguous sequence of nucleotides comprising a STAT3 mRNA, wherein the contiguous sequence of nucleotides is 10, 11, 12, 13, 14, 15, 16, 17, 18, 19 or 20 nucleotides in length. In some embodiments, the oligonucleotide comprises a targeting sequence or region of complementarity that is complementary to a contiguous sequence of nucleotides comprising a STAT3 mRNA, wherein the contiguous sequence of nucleotides is 19 nucleotides in length. [00173] In some embodiments, an oligonucleotide herein (e.g., an RNAi oligonucleotide) comprises a targeting sequence or a region of complementary that is complementary to a contiguous sequence of nucleotides of any one of SEQ ID NOs: 89-280, optionally wherein the contiguous sequence of nucleotides is 19 nucleotides in length. In some embodiments, the oligonucleotide comprises a targeting sequence or a region of complementary that is complementary to a contiguous sequence of nucleotides of any one of SEQ ID NOs: 108, 140, 141, and 147, wherein the contiguous sequence of nucleotides is 19 nucleotides in length. In some embodiments, the oligonucleotide comprises a targeting sequence or a region of complementary that is complementary to a contiguous sequence of nucleotides of any one of SEQ ID NOs: 473-664, wherein the contiguous sequence of nucleotides is 20 nucleotides in length. In some embodiments, the oligonucleotide comprises a targeting sequence or a region of complementary that is complementary to a contiguous sequence of nucleotides of any one of SEQ ID NOs: 492, 524, 525, and 531, wherein the contiguous sequence of nucleotides is 20 nucleotides in length. [00174] In some embodiments, a targeting sequence or region of complementarity of an oligonucleotide that is complementary to contiguous nucleotides of STAT3 or STAT3 target sequence spans the entire length of an antisense strand. In some embodiments, a region of complementarity of an oligonucleotide that is complementary to contiguous nucleotides of STAT3 or STAT3 target sequence spans a portion of the entire length of an antisense strand. In some embodiments, an oligonucleotide herein comprises a region of complementarity (e.g., on an antisense strand of a dsRNA) that is at least partially (e.g., fully) complementary to a contiguous stretch of nucleotides spanning nucleotides 1-20 of a target sequence of STAT3 or STAT3. [00175] In some embodiments, a targeting sequence or region of complementarity of an oligonucleotide herein (e.g., an RNAi oligonucleotide) is complementary to a contiguous sequence of nucleotides of any one of SEQ ID NOs: 89-280 and spans the entire length of an antisense strand. In some embodiments, a targeting sequence or region of complementarity of the oligonucleotide is complementary to a contiguous sequence of nucleotides of SEQ ID NOs: 89-280 and spans a portion of the entire length of an antisense strand. In some embodiments, an oligonucleotide herein (e.g., an RNAi oligonucleotide) comprises a region of complementarity (e.g., on an antisense strand of a dsRNA) that is at least partially (e.g., fully) complementary to a contiguous stretch of nucleotides spanning nucleotides 1-19 or 1- 20 of a sequence as set forth in any one of SEQ ID NOs: 473-664. [00176] In some embodiments, an oligonucleotide herein comprises a targeting sequence or region of complementarity having one or more bp mismatches with the corresponding STAT3 target sequence. In some embodiments, the targeting sequence or region of complementarity may have up to about 1, up to about 2, up to about 3, up to about 4, up to about 5, etc. mismatches with the corresponding STAT3 target sequence provided that the ability of the targeting sequence or region of complementarity to bind or anneal to the STAT3 mRNA under appropriate hybridization conditions and/or the ability of the oligonucleotide to inhibit STAT3 expression is maintained. Alternatively, the targeting sequence or region of complementarity may have no more than 1, no more than 2, no more than 3, no more than 4, or no more than 5 mismatches with the corresponding STAT3 target sequence provided that the ability of the targeting sequence or region of complementarity to bind or anneal to the STAT3 mRNA under appropriate hybridization conditions and/or the ability of the oligonucleotide to inhibit STAT3 expression is maintained. In some embodiments, the oligonucleotide comprises a targeting sequence or region of complementarity having 1 mismatch with the corresponding target sequence. In some embodiments, the oligonucleotide comprises a targeting sequence or region of complementarity having 2 mismatches with the corresponding target sequence. In some embodiments, the oligonucleotide comprises a targeting sequence or region of complementarity having 3 mismatches with the corresponding target sequence. In some embodiments, the oligonucleotide comprises a targeting sequence or region of complementarity having 4 mismatches with the corresponding target sequence. In some embodiments, the oligonucleotide comprises a targeting sequence or region of complementarity having 5 mismatches with the corresponding target sequence. In some embodiments, the oligonucleotide comprises a targeting sequence or region of complementarity more than one mismatch (e.g., 2, 3, 4, 5 or more mismatches) with the corresponding target sequence, wherein at least 2 (e.g., all) of the mismatches are positioned consecutively (e.g., 2, 3, 4, 5 or more mismatches in a row), or where in the mismatches are interspersed throughout the targeting sequence or region of complementarity. In some embodiments, the oligonucleotide comprises a targeting sequence or a region of complementary that is complementary to a contiguous sequence of nucleotides of any one of SEQ ID NOs: 89-280, wherein the targeting sequence or region of complementarity may have up to about 1, up to about 2, up to about 3, up to about 4, up to about 5, etc. mismatches with the corresponding STAT3 target sequence. In some embodiments, the oligonucleotide comprises a targeting sequence or a region of complementary that is complementary to a contiguous sequence of nucleotides of any one of SEQ ID NOs: 89-280, wherein the targeting sequence or region of complementarity may have no more than 1, no more than 2, no more than 3, no more than 4, or no more than 5 mismatches with the corresponding STAT3 target sequence. In some embodiments, the oligonucleotide comprises a targeting sequence or a region of complementary that is complementary to a contiguous sequence of nucleotides of any one of SEQ ID NOs: 108, 140, 141, and 147, wherein the targeting sequence or region of complementarity may have up to about 1, up to about 2, up to about 3, up to about 4, up to about 5, etc. mismatches with the corresponding STAT3 target sequence. In some embodiments, the oligonucleotide comprises a targeting sequence or a region of complementary that is complementary to a contiguous sequence of nucleotides of any one of SEQ ID NOs: 108, 140, 141, and 147, wherein the targeting sequence or region of complementarity may have no more than 1, no more than 2, no more than 3, no more than 4, or no more than 5 mismatches with the corresponding STAT3 target sequence. Types of Oligonucleotides [00177] A variety of oligonucleotide types and/or structures are useful for targeting a target sequence in the methods herein including, but not limited to, RNAi oligonucleotides, antisense oligonucleotides, miRNAs, etc. Any of the oligonucleotide types described herein or elsewhere are contemplated for use as a framework to incorporate a targeting sequence herein. [00178] In some embodiments, the oligonucleotides herein inhibit expression of a target sequence by engaging with RNA interference (RNAi) pathways upstream or downstream of Dicer involvement. For example, RNAi oligonucleotides have been developed with each strand having sizes of about 19-25 nucleotides with at least one 3′ overhang of 1 to 5 nucleotides (see, e.g., US Patent No.8,372,968). Longer oligonucleotides also have been developed that are processed by Dicer to generate active RNAi products (see, e.g., US Patent No.8,883,996). Further work produced extended dsRNAs where at least one end of at least one strand is extended beyond a duplex targeting region, including structures where one of the strands includes a thermodynamically-stabilizing tetraloop structure (see, e.g., US Patent Nos.8,513,207 and 8,927,705, as well as Intl. Patent Application Publication No. WO 2010/033225). Such structures may include ss extensions (on one or both sides of the molecule) as well as ds extensions. [00179] In some embodiments, the oligonucleotides herein engage with the RNAi pathway downstream of the involvement of Dicer (e.g., Dicer cleavage). In some embodiments, the oligonucleotides described herein are Dicer substrates. In some embodiments, upon endogenous Dicer processing, double-stranded nucleic acids of 19-23 nucleotide sin length capable of reducing target mRNA expression are produced. In some embodiments, the oligonucleotide has an overhang (e.g., of 1, 2, or 3 nucleotides in length) in the 3′ end of the sense strand. In some embodiments, the oligonucleotide (e.g., siRNA) comprises a 21-nucleotide guide strand that is antisense to a target RNA and a complementary passenger strand, in which both strands anneal to form a 19-bp duplex and 2 nucleotide overhangs at either or both 3′ ends. Longer oligonucleotide designs also are available including oligonucleotides having a guide strand of 23 nucleotides and a passenger strand of 21 nucleotides, where there is a blunt end on the right side of the molecule (3′ end of passenger strand/5′ end of guide strand) and a two nucleotide 3′-guide strand overhang on the left side of the molecule (5′ end of the passenger strand/3′ end of the guide strand). In such molecules, there is a 21 bp duplex region. See, e.g., US Patent Nos.9,012,138; 9,012,621 and 9,193,753. [00180] In some embodiments, the oligonucleotides herein comprise sense and antisense strands that are both in the range of about 17 to 26 (e.g., 17 to 26, 20 to 25 or 21- 23) nucleotides in length. In some embodiments, the oligonucleotides herein comprise sense and antisense strands that are both in the range of about 17 to 36 (e.g., 17 to 36, 20 to 25 or 21-23) nucleotides in length. In some embodiments, the oligonucleotides described herein comprise an antisense strand of 19-30 nucleotides in length and a sense strand of 19-50 nucleotides in length, wherein the antisense and sense strands are separate strands which form an asymmetric duplex region having an overhand of 1-4 nucleotides at the 3’ terminus of the antisense strand. In some embodiments, an oligonucleotide herein comprises a sense and antisense strand that are both in the range of about 19-22 nucleotides in length. In some embodiments, the sense and antisense strands are of equal length. In some embodiments, an oligonucleotide comprises sense and antisense strands, such that there is a 3′-overhang on either the sense strand or the antisense strand, or both the sense and antisense strand. In some embodiments, for oligonucleotides that have sense and antisense strands that are both in the range of about 21-23 nucleotides in length, a 3′ overhang on the sense, antisense, or both sense and antisense strands is 1 or 2 nucleotides in length. In some embodiments, the oligonucleotide has a guide strand of 22 nucleotides and a passenger strand of 20 nucleotides, where there is a blunt end on the right side of the molecule (3′ end of passenger strand/5′ end of guide strand) and a 2 nucleotide 3′-guide strand overhang on the left side of the molecule (5′ end of the passenger strand/3′ end of the guide strand). In such molecules, there is a 20 bp duplex region. [00181] Other oligonucleotide designs for use with the compositions and methods herein include: 16-mer siRNAs (see, e.g., NUCLEIC ACIDS IN CHEMISTRY AND BIOLOGY. Blackburn (ed.), Royal Society of Chemistry, 2006), shRNAs (e.g., having 19 bp or shorter stems; (see, e.g., Moore et al., (2010) METHODS MOL. BIOL.629:141-58), blunt siRNAs (e.g., of 19 bps in length; see, e.g., Kraynack and Baker (2006) RNA 12:163-76), asymmetrical siRNAs (aiRNA; see, e.g., Sun et al., (2008) NAT. BIOTECHNOL.26:1379-82), asymmetric shorter-duplex siRNA (see, e.g., Chang et al., (2009) MOL. THER.17:725-32), fork siRNAs (see, e.g., Hohjoh (2004) FEBS LETT.557:193-98), ss siRNAs (Elsner (2012) NAT. BIOTECHNOL.30:1063), dumbbell-shaped circular siRNAs (see, e.g., Abe et al., (2007) J. AM. CHEM. SOC.129:15108-09), and small internally segmented interfering RNA (siRNA; see, e.g., Bramsen et al., (2007) NUCLEIC ACIDS RES.35:5886-97). Further non-limiting examples of an oligonucleotide structures that may be used in some embodiments to reduce or inhibit the expression of STAT3 are microRNA (miRNA), short hairpin RNA (shRNA) and short siRNA (see, e.g., Hamilton et al., (2002) EMBO J.21:4671-79; see also, US Patent Application Publication No.2009/0099115). [00182] Still, in some embodiments, an oligonucleotide for reducing or inhibiting expression of a target sequence herein is ss. Such structures may include but are not limited to ss RNAi molecules. Recent efforts have demonstrated the activity of ss RNAi molecules (see, e.g., Matsui et al., (2016) MOL. THER.24:946-55). However, in some embodiments, oligonucleotides herein are antisense oligonucleotides (ASOs). An antisense oligonucleotide is a ss oligonucleotide that has a nucleobase sequence which, when written in the 5′ to 3′ direction, comprises the reverse complement of a targeted segment of a particular nucleic acid and is suitably modified (e.g., as a gapmer) to induce RNaseH-mediated cleavage of its target RNA in cells or (e.g., as a mixmer) to inhibit translation of the target mRNA in cells. ASOs for use herein may be modified in any suitable manner known in the art including, for example, as shown in US Patent No.9,567,587 (including, e.g., length, sugar moieties of the nucleobase (pyrimidine, purine), and alterations of the heterocyclic portion of the nucleobase). Further, ASOs have been used for decades to reduce expression of specific target genes (see, e.g., Bennett et al., (2017) ANNU. REV. PHARMACOL.57:81-105). [00183] In some embodiments, the antisense oligonucleotide shares a region of complementarity with a target mRNA. In some embodiments, the antisense oligonucleotide is 15-50 nucleotides in length. In some embodiments, the antisense oligonucleotide is 15-25 nucleotides in length. In some embodiments, the antisense oligonucleotide is 22 nucleotides in length. In some embodiments, the antisense oligonucleotide is at least 15 contiguous nucleotides in length. In some embodiments, the antisense oligonucleotide is at least 19 contiguous nucleotides in length. In some embodiments, the antisense oligonucleotide is at least 20 contiguous nucleotides in length. In some embodiments, the antisense oligonucleotide differs by 1, 2, or 3 nucleotides from the target sequence. Double-Stranded Oligonucleotides [00184] In some embodiments, the disclosure provides double-stranded dsRNAs for targeting and inhibiting expression of a target sequence (e.g., via the RNAi pathway) comprising a sense strand (also referred to herein as a passenger strand) and an antisense strand (also referred to herein as a guide strand). In some embodiments, the sense strand and antisense strand are separate strands and are not covalently linked. In some embodiments, the sense strand and antisense strand are covalently linked. In some embodiments, the sense strand and antisense strand form a duplex region, wherein the sense strand and antisense strand, or a portion thereof, binds with one another in a complementary fashion (e.g., by Watson-Crick base pairing). [00185] In some embodiments, the sense strand has a first region (R1) and a second region (R2), wherein R2 comprises a first subregion (S1), a loop (L), such as a tetraloop (tetraL) or triloop (triL), and a second subregion (S2), wherein L, tetraL, or triL is located between S1 and S2, and wherein S1 and S2 form a second duplex (D2). D2 may have various length. In some embodiments, D2 is about 1-6 bp in length. In some embodiments, D2 is 2-6, 3-6, 4-6, 5-6, 1-5, 2-5, 3-5 or 4-5 bp in length. In some embodiments, D2 is 1, 2, 3, 4, 5 or 6 bp in length. In some embodiments, D2 is 6 bp in length. [00186] In some embodiments, R1 of the sense strand and the antisense strand form a first duplex (D1). In some embodiments, D1 is at least about 15 (e.g., at least 15, at least 16, at least 17, at least 18, at least 19, at least 20 or at least 21) nucleotides in length. In some embodiments, D1 is in the range of about 12 to 30 nucleotides in length (e.g., 12 to 30, 12 to 27, 15 to 22, 18 to 22, 18 to 25, 18 to 27, 18 to 30 or 21 to 30 nucleotides in length). In some embodiments, D1 is at least 12 nucleotides in length (e.g., at least 12, at least 15, at least 20, at least 25, or at least 30 nucleotides in length). In some embodiments, D1 is 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29 or 30 nucleotides in length. In some embodiments, D1 is 20 nucleotides in length. In some embodiments, D1 comprising sense strand and antisense strand does not span the entire length of the sense strand and/or antisense strand. In some embodiments, D1 comprising the sense strand and antisense strand spans the entire length of either the sense strand or antisense strand or both. In certain embodiments, D1 comprising the sense strand and antisense strand spans the entire length of both the sense strand and the antisense strand. [00187] It should be appreciated that, in some embodiments, sequences presented in the Sequence Listing may be referred to in describing the structure of an oligonucleotide or other nucleic acid. In such embodiments, the actual oligonucleotide or other nucleic acid may have one or more alternative nucleotides (e.g., an RNA counterpart of a DNA nucleotide or a DNA counterpart of an RNA nucleotide) and/or one or more modified nucleotides and/or one or more modified internucleotide linkages and/or one or more other modification when compared with the specified sequence while retaining essentially same or similar complementary properties as the specified sequence. [00188] In some embodiments, a double-stranded RNA (dsRNA) herein comprises a 25-nucleotide sense strand and a 27-nucleotide antisense strand that when acted upon by a Dicer enzyme results in an antisense strand that is incorporated into the mature RISC. In some embodiments, the sense strand of the dsRNA is longer than 27 nucleotides (e.g., 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39 or 40 nucleotides). In some embodiments, the sense strand of the dsRNA is longer than 27 nucleotides (e.g., 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, or 50 nucleotides). In some embodiments, the sense strand of the dsRNA is longer than 25 nucleotides (e.g., 26, 27, 28, 29 or 30 nucleotides). [00189] In some embodiments, oligonucleotides herein have one 5′ end that is thermodynamically less stable when compared to the other 5′ end. In some embodiments, an asymmetry oligonucleotide is provided that includes a blunt end at the 3′ end of a sense strand and a 3′-overhang at the 3′ end of an antisense strand. In some embodiments, the 3′- overhang on the antisense strand is about 1-8 nucleotides in length (e.g., 1, 2, 3, 4, 5, 6, 7 or 8 nucleotides in length). Typically, an oligonucleotide for RNAi has a two-nucleotide overhang on the 3′ end of the antisense (guide) strand. However, other overhangs are possible. In some embodiments, an overhang is a 3′-overhang comprising a length of between 1 and 6 nucleotides, optionally 1 to 5, 1 to 4, 1 to 3, 1 to 2, 2 to 6, 2 to 5, 2 to 4, 2 to 3, 3 to 6, 3 to 5, 3 to 4, 4 to 6, 4 to 5, 5 to 6 nucleotides, or 1, 2, 3, 4, 5 or 6 nucleotides. However, in some embodiments, the overhang is a 5′-overhang comprising a length of between 1 and 6 nucleotides, optionally 1 to 5, 1 to 4, 1 to 3, 1 to 2, 2 to 6, 2 to 5, 2 to 4, 2 to 3, 3 to 6, 3 to 5, 3 to 4, 4 to 6, 4 to 5, 5 to 6 nucleotides, or 1, 2, 3, 4, 5 or 6 nucleotides. [00190] In some embodiments, two terminal nucleotides on the 3′ end of an antisense strand are modified. In some embodiments, the two terminal nucleotides on the 3′ end of the antisense strand are complementary with the target mRNA. In some embodiments, the two terminal nucleotides on the 3′ end of the antisense strand are not complementary with the target mRNA. In some embodiments, the two terminal nucleotides on the 3′ end of the antisense strand of an oligonucleotide herein comprise an unpaired GG. In some embodiments, the two (2) terminal nucleotides on the 3′ end of an antisense strand of an oligonucleotide herein are not complementary to the target mRNA. In some embodiments, two terminal nucleotides on each 3′ end of an oligonucleotide in the nicked tetraloop structure are GG. In some embodiments, one or both of the two (2) terminal GG nucleotides on each 3′ end of an oligonucleotide herein is not complementary with the target mRNA. Typically, one or both two terminal GG nucleotides on each 3′ end of an oligonucleotide is not complementary with the target. [00191] In some embodiments, there is one or more (e.g., 1, 2, 3, 4 or 5) mismatch between a sense and antisense strand. If there is more than one mismatch between a sense and antisense strand, they may be positioned consecutively (e.g., 2, 3 or more in a row), or interspersed throughout the region of complementarity. In some embodiments, the 3′ end of the sense strand contains one or more mismatches. In one embodiment, two mismatches are incorporated at the 3′ end of the sense strand. In some embodiments, base mismatches, or destabilization of segments at the 3′ end of the sense strand of the oligonucleotide improved the potency of synthetic duplexes in RNAi, possibly through facilitating processing by Dicer. a. Antisense Strands [00192] In some embodiments, a dsRNA comprises an antisense strand of up to about 40 nucleotides in length (e.g., up to 40, up to 35, up to 30, up to 27, up to 25, up to 21, up to 19, up to 17 or up to 12 nucleotides in length). In some embodiments, an oligonucleotide herein (e.g., an RNAi oligonucleotide) comprises an antisense strand of up to about 50 nucleotides in length (e.g., up to 50, up to 40, up to 35, up to 30, up to 27, up to 25, up to 21, up to 19, up to 17 or up to 12 nucleotides in length). In some embodiments, an oligonucleotide may have an antisense strand of at least about 12 nucleotides in length (e.g., at least 12, at least 15, at least 19, at least 21, at least 22, at least 25, at least 27, at least 30, at least 35 or at least 38 nucleotides in length). In some embodiments, an oligonucleotide may have an antisense strand in a range of about 12 to about 40 (e.g., 12 to 40, 12 to 36, 12 to 32, 12 to 28, 15 to 40, 15 to 36, 15 to 32, 15 to 28, 17 to 22, 17 to 25, 19 to 27, 19 to 30, 20 to 40, 22 to 40, 25 to 40 or 32 to 40) nucleotides in length. In some embodiments, an oligonucleotide comprises antisense strand of 15 to 30 nucleotides in length. In some embodiments, an oligonucleotide may have an antisense strand of 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 in length. [00193] In some embodiments, an antisense strand of an oligonucleotide may be referred to as a “guide strand.” For example, if an antisense strand can engage with RNA- induced silencing complex (RISC) and bind to an Argonaute protein such as Ago2, or engage with or bind to one or more similar factors, and direct silencing of a target gene, it may be referred to as a guide strand. In some embodiments, a sense strand complementary to a guide strand may be referred to as a “passenger strand.” [00194] In some embodiments, an oligonucleotide disclosed herein for targeting STAT3 comprises an antisense strand comprising or consisting of a sequence as set forth in any one of SEQ ID NOs: 281-472. In some embodiments, an oligonucleotide herein comprises an antisense strand comprising at least about 12 (e.g., at least 12, at least 13, at least 14, at least 15, at least 16, at least 17, at least 18, at least 19, at least 20, at least 21, at least 22 or at least 23) contiguous nucleotides of a sequence as set forth in any one of SEQ ID NOs: 281-472. In some embodiments, an oligonucleotide disclosed herein (e.g., an RNAi oligonucleotide) for targeting STAT3 comprises an antisense strand comprising or consisting of a sequence as set forth in any one of SEQ ID NOs: 665-856. In some embodiments, an oligonucleotide herein comprises an antisense strand comprising at least about 12 (e.g., at least 12, at least 13, at least 14, at least 15, at least 16, at least 17, at least 18, at least 19, at least 20, at least 21, at least 22 or at least 23) contiguous nucleotides of a sequence as set forth in any one of SEQ ID NOs: 665-856. In some embodiments, an oligonucleotide disclosed herein for targeting STAT3 comprises an antisense strand comprising or consisting of a sequence as set forth in any one of SEQ ID NOs: 947-1036. In some embodiments, an oligonucleotide herein comprises an antisense strand comprising at least about 12 (e.g., at least 12, at least 13, at least 14, at least 15, at least 16, at least 17, at least 18, at least 19, at least 20, at least 21, at least 22 or at least 23) contiguous nucleotides of a sequence as set forth in any one of SEQ ID NOs: 947-1036. In some embodiments, an oligonucleotide disclosed herein for targeting STAT3 comprises an antisense strand comprising or consisting of a sequence as set forth in any one of SEQ ID NOs: 10, 38, 66, and 70. In some embodiments, an oligonucleotide herein comprises an antisense strand comprising at least about 12 (e.g., at least 12, at least 13, at least 14, at least 15, at least 16, at least 17, at least 18, at least 19, at least 20, at least 21, at least 22 or at least 23) contiguous nucleotides of a sequence as set forth in any one of SEQ ID NOs: 10, 38, 66, and 70. In some embodiments, an oligonucleotide disclosed herein for targeting STAT3 comprises an antisense strand comprising or consisting of a sequence as set forth in any one of SEQ ID NOs: 281-472. In some embodiments, an oligonucleotide herein comprises an antisense strand comprising at least about 12 (e.g., at least 12, at least 13, at least 14, at least 15, at least 16, at least 17, at least 18, at least 19, at least 20, at least 21, at least 22 or at least 23) contiguous nucleotides of a sequence as set forth in any one of SEQ ID NOs: 281-472. In some embodiments, an oligonucleotide disclosed herein for targeting STAT3 comprises an antisense strand comprising or consisting of a sequence as set forth in any one of SEQ ID NOs: 952, 965, 966, and 1010. In some embodiments, an oligonucleotide herein comprises an antisense strand comprising at least about 12 (e.g., at least 12, at least 13, at least 14, at least 15, at least 16, at least 17, at least 18, at least 19, at least 20, at least 21, at least 22 or at least 23) contiguous nucleotides of a sequence as set forth in any one of SEQ ID NOs: 952, 965, 966, and 1010. In some embodiments, an oligonucleotide disclosed herein for targeting STAT3 comprises an antisense strand comprising or consisting of a sequence as set forth in any one of SEQ ID NOs: 991, 1000, 989, 986, 982, 980, and 979. In some embodiments, an oligonucleotide herein comprises an antisense strand comprising at least about 12 (e.g., at least 12, at least 13, at least 14, at least 15, at least 16, at least 17, at least 18, at least 19, at least 20, at least 21, at least 22 or at least 23) contiguous nucleotides of a sequence as set forth in any one of SEQ ID NOs: 991, 1000, 989, 986, 982, 980, and 979. In some embodiments, an oligonucleotide disclosed herein for targeting STAT3 comprises an antisense strand comprising or consisting of a sequence as set forth in any one of SEQ ID NOs: 1030, 1027, and 1029. In some embodiments, an oligonucleotide herein comprises an antisense strand comprising at least about 12 (e.g., at least 12, at least 13, at least 14, at least 15, at least 16, at least 17, at least 18, at least 19, at least 20, at least 21, at least 22 or at least 23) contiguous nucleotides of a sequence as set forth in any one of SEQ ID NOs: 1030, 1027, and 1029. In some embodiments, an oligonucleotide disclosed herein for targeting STAT3 comprises an antisense strand comprising or consisting of a sequence as set forth in any one of SEQ ID NOs: 1005, 1014, 1003, and 1010. In some embodiments, an oligonucleotide herein comprises an antisense strand comprising at least about 12 (e.g., at least 12, at least 13, at least 14, at least 15, at least 16, at least 17, at least 18, at least 19, at least 20, at least 21, at least 22 or at least 23) contiguous nucleotides of a sequence as set forth in any one of SEQ ID NOs: 1005, 1014, 1003, and 1010. [00195] b. Sense Strands [00196] In some embodiments, an oligonucleotide disclosed herein (e.g., and RNAi oligonucleotide) for targeting STAT3 mRNA and inhibiting STAT3 expression comprises a sense strand sequence as set forth in in any one of SEQ ID NOs: 89-280. In some embodiments, an oligonucleotide herein has a sense strand that comprise at least about 12 (e.g., at least 13, at least 14, at least 15, at least 16, at least 17, at least 18, at least 19, at least 20, at least 21, at least 22 or at least 23) contiguous nucleotides of a sequence as set forth in in any one of SEQ ID NOs: 89-280. In some embodiments, an oligonucleotide disclosed herein (e.g., an RNAi oligonucleotide) for targeting STAT3 mRNA and inhibiting STAT3 expression comprises a sense strand sequence a set forth in any one of SEQ ID NOs: 473- 664. In some embodiments, an oligonucleotide herein has a sense strand that comprise at least about 12 (e.g., at least 13, at least 14, at least 15, at least 16, at least 17, at least 18, at least 19, at least 20, at least 21, at least 22 or at least 23) contiguous nucleotides of a sequence as set forth in in any one of SEQ ID NOs: 473-664. In some embodiments, an oligonucleotide disclosed herein for targeting STAT3 mRNA and inhibiting STAT3 expression comprises a sense strand sequence as set forth in any one of SEQ ID NOs: 857-946. In some embodiments, an oligonucleotide herein has a sense strand comprised of least about 12 (e.g., at least 13, at least 14, at least 15, at least 16, at least 17, at least 18, at least 19, at least 20, at least 21, at least 22 or at least 23) contiguous nucleotides of a sequence as set forth in any one of SEQ ID NOs: 857-946. In some embodiments, an oligonucleotide disclosed herein for targeting STAT3 mRNA and inhibiting STAT3 expression comprises a sense strand sequence as set forth in any one of SEQ ID NOs: 9, 37, 65, and 69. In some embodiments, an oligonucleotide herein has a sense strand comprised of least about 12 (e.g., at least 13, at least 14, at least 15, at least 16, at least 17, at least 18, at least 19, at least 20, at least 21, at least 22 or at least 23) contiguous nucleotides of a sequence as set forth in any one of SEQ ID NOs: 9, 37, 65, and 69. In some embodiments, an oligonucleotide disclosed herein for targeting STAT3 mRNA and inhibiting STAT3 expression comprises a sense strand sequence as set forth in any one of SEQ ID NOs: 862, 875, 876, and 920. In some embodiments, an oligonucleotide herein has a sense strand that comprise at least about 12 (e.g., at least 13, at least 14, at least 15, at least 16, at least 17, at least 18, at least 19, at least 20, at least 21, at least 22 or at least 23) contiguous nucleotides of a sequence as set forth in any one of SEQ ID NOs: 862, 875, 876, and 920. In some embodiments, an oligonucleotide disclosed herein for targeting STAT3 mRNA and inhibiting STAT3 expression comprises a sense strand sequence as set forth in any one of SEQ ID NOs: 901, 910, 899, 896, 892, 890, and 889. In some embodiments, an oligonucleotide herein has a sense strand that comprise at least about 12 (e.g., at least 13, at least 14, at least 15, at least 16, at least 17, at least 18, at least 19, at least 20, at least 21, at least 22 or at least 23) contiguous nucleotides of a sequence as set forth in any one of SEQ ID NOs: 901, 910, 899, 896, 892, 890, and 889. In some embodiments, an oligonucleotide disclosed herein for targeting STAT3 comprises an antisense strand comprising or consisting of a sequence as set forth in any one of SEQ ID NOs: 940, 937, and 939. In some embodiments, an oligonucleotide herein comprises an antisense strand comprising at least about 12 (e.g., at least 12, at least 13, at least 14, at least 15, at least 16, at least 17, at least 18, at least 19, at least 20, at least 21, at least 22 or at least 23) contiguous nucleotides of a sequence as set forth in any one of SEQ ID NOs: 940, 937, and 939. In some embodiments, an oligonucleotide disclosed herein for targeting STAT3 comprises an antisense strand comprising or consisting of a sequence as set forth in any one of SEQ ID NOs: 915, 924, 913, and 920. In some embodiments, an oligonucleotide herein comprises an antisense strand comprising at least about 12 (e.g., at least 12, at least 13, at least 14, at least 15, at least 16, at least 17, at least 18, at least 19, at least 20, at least 21, at least 22 or at least 23) contiguous nucleotides of a sequence as set forth in any one of SEQ ID NOs: 915, 924, 913, and 920. [00197] In some embodiments, an oligonucleotide comprises a sense strand (or passenger strand) of up to about 40 nucleotides in length (e.g., up to 40, up to 36, up to 30, up to 27, up to 25, up to 21, up to 19, up to 17 or up to 12 nucleotides in length). In some embodiments, an oligonucleotide may have a sense strand of at least about 12 nucleotides in length (e.g., at least 12, at least 15, at least 19, at least 21, at least 25, at least 27, at least 30, at least 36 or at least 38 nucleotides in length). In some embodiments, an oligonucleotide may have a sense strand in a range of about 12 to about 40 (e.g., 12 to 40, 12 to 36, 12 to 32, 12 to 28, 15 to 40, 15 to 36, 15 to 32, 15 to 28, 17 to 21, 17 to 25, 19 to 27, 19 to 30, 20 to 40, 22 to 40, 25 to 40 or 32 to 40) nucleotides in length. In some embodiments, an oligonucleotide herein comprises a sense strand of 15 to 50 nucleotides in length. In some embodiments, an oligonucleotide herein comprises a sense strand of 18 to 36 nucleotides in length. In some embodiments, an oligonucleotide may have a sense strand of 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 in length. In some embodiments, an oligonucleotide comprises a sense strand of 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, 40,41, 42, 43, 44, 45, 46, 47, 48, 49, or 50 nucleotides in length. In some embodiments, an oligonucleotide herein comprises a sense strand of 36 nucleotides in length. [00198] In some embodiments, an oligonucleotide provided herein (e.g., an RNAi oligonucleotide) comprises a sense strand comprising a stem-loop structure at the 3′ end of the sense strand. In some embodiments, the stem-loop is formed by intrastrand base pairing. In some embodiments, a sense strand comprises a stem-loop structure at its 5′ end. In some embodiments, the stem of the stem-loop comprises a duplex of 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13 or 14 nucleotides in length. In some embodiments, the stem of the stem-loop comprises a duplex of 2 nucleotides in length. In some embodiments, the stem of the stem-loop comprises a duplex of 3 nucleotides in length. In some embodiments, the stem of the stem-loop comprises a duplex of 4 nucleotides in length. In some embodiments, the stem of the stem- loop comprises a duplex of 5 nucleotides in length. In some embodiments, the stem of the stem-loop comprises a duplex of 6 nucleotides in length. In some embodiments, the stem of the stem-loop comprises a duplex of 7 nucleotides in length. In some embodiments, the stem of the stem-loop comprises a duplex of 8 nucleotides in length. In some embodiments, the stem of the stem-loop comprises a duplex of 9 nucleotides in length. In some embodiments, the stem of the stem-loop comprises a duplex of 10 nucleotides in length. In some embodiments, the stem of the stem-loop comprises a duplex of 11 nucleotides in length. In some embodiments, the stem of the stem-loop comprises a duplex of 12 nucleotides in length. In some embodiments, the stem of the stem-loop comprises a duplex of 13 nucleotides in length. In some embodiments, the stem of the stem-loop comprises a duplex of 14 nucleotides in length. [00199] In some embodiments, a stem-loop provides the oligonucleotide protection against degradation (e.g., enzymatic degradation), facilitates or improves targeting and/or delivery to a target cell, tissue, or organ (e.g., the liver), or both. For example, in some embodiments, the loop of a stem-loop is comprised of nucleotides comprising one or more modifications that facilitate, improve, or increase targeting to a target, inhibition of target gene expression, and/or delivery, uptake, and/or penetrance into a target cell, tissue, or organ (e.g., the liver), or a combination thereof. In some embodiments, the stem-loop itself or modification(s) to the stem-loop do not affect or do not substantially affect the inherent gene expression inhibition activity of the oligonucleotide, but facilitates, improves, or increases stability (e.g., provides protection against degradation) and/or delivery, uptake, and/or penetrance of the oligonucleotide to a target cell, tissue, or organ. In certain embodiments, an oligonucleotide herein comprises a sense strand comprising (e.g., at its 3′ end) a stem-loop set forth as: S1-L-S2, in which S1 is complementary to S2, and in which L forms a single- stranded loop of linked nucleotides between S1 and S2 of up to about 10 nucleotides in length (e.g., 3, 4, 5, 6, 7, 8, 9 or 10 nucleotides in length). In some embodiments, the loop (L) is 3 nucleotides in length (referred to herein as “triloop”. In some embodiments, the loop (L) is 4 nucleotides in length (referred to herein as “tetraloop”). In some embodiments, the loop (L) is 5 nucleotides in length. In some embodiments, the loop (L) is 6 nucleotides in length. In some embodiments, the loop (L) is 7 nucleotides in length. In some embodiments, the loop (L) is 8 nucleotides in length. In some embodiments, the loop (L) is 9 nucleotides in length. In some embodiments, the loop (L) is 10 nucleotides in length. [00200] In some embodiments, an oligonucleotide provided herein (e.g., an RNAi oligonucleotide) comprises a targeting sequence or a region of complementary that is complementary to a contiguous sequence of nucleotides of any one of SEQ ID NOs: 89-280, and the oligonucleotide comprises a sense strand comprising (e.g., at its 3′ end) a stem-loop set forth as: S1-L-S2, in which S1 is complementary to S2, and in which L forms a single- stranded loop between S1 and S2 of up to about 10 nucleotides in length (e.g., 3, 4, 5, 6, 7, 8, 9 or 10 nucleotides in length). In some embodiments, the oligonucleotide comprises a targeting sequence or a region of complementary that is complementary to a contiguous sequence of nucleotides of any one of SEQ ID NOs: 89-280, and the oligonucleotide comprises a sense strand comprising (e.g., at its 3′ end) a stem-loop set forth as: S1-L-S2, in which S1 is complementary to S2, and in which L forms a single-stranded loop between S1 and S2 of 4 nucleotides in length. [00201] In some embodiments, the tetraloop comprises the sequence 5’-GAAA-3’. In some embodiments, the stem loop comprises the sequence 5’-GCAGCCGAAAGGCUGC-3’ (SEQ ID NO: 86). [00202] In some embodiments, a sense strand comprises a stem-loop structure at its 3′ end. In some embodiments, a sense strand comprises a stem-loop structure at its 5′ end. In some embodiments, a stem is a duplex of 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13 or 14 bp in length. In some embodiments, a stem-loop provides the molecule protection against degradation (e.g., enzymatic degradation) and facilitates targeting characteristics for delivery to a target cell. For example, in some embodiments, a loop provides added nucleotides on which modification can be made without substantially affecting the gene expression inhibition activity of an oligonucleotide. In certain embodiments, an oligonucleotide is herein in which the sense strand comprises (e.g., at its 3′ end) a stem-loop set forth as: S1-L- S2, in which S1 is complementary to S2, and in which L forms a loop between S1 and S2 of up to about 10 nucleotides in length (e.g., 3, 4, 5, 6, 7, 8, 9 or 10 nucleotides in length). FIG. 1A depicts non-limiting examples of such an oligonucleotide. [00203] In some embodiments, a loop (L) of a stem-loop having the structure S1-L-S2 as described herein is a triloop. In some embodiments, the triloop comprises ribonucleotides, deoxyribonucleotides, modified nucleotides, ligands (e.g., delivery ligands), and combinations thereof. [00204] In some embodiments, a loop of a stem-loop is a tetraloop (e.g., within a nicked tetraloop structure). A tetraloop may contain ribonucleotides, deoxyribonucleotides, modified nucleotides and combinations thereof. Typically, a tetraloop has 4 to 5 nucleotides. Duplex Length [00205] In some embodiments, a duplex formed between a sense and antisense strand is at least 12 (e.g., at least 15, at least 16, at least 17, at least 18, at least 19, at least 20, or at least 21) nucleotides in length. In some embodiments, a duplex formed between a sense and antisense strand is in the range of 12-30 nucleotides in length (e.g., 12 to 30, 12 to 27, 12 to 22, 15 to 25, 18 to 30, 18 to 22, 18 to 25, 18 to 27, 18 to 30, 19 to 30 or 21 to 30 nucleotides in length). In some embodiments, a duplex formed between a sense and antisense strand is 12, 13, 14, 15, 16, 17, 18, 19, 29, 21, 22, 23, 24, 25, 26, 27, 28, 29 or 30 nucleotides in length. In some embodiments, a duplex formed between a sense and antisense strand is 12 nucleotides in length. In some embodiments, a duplex formed between a sense and antisense strand is 13 nucleotides in length. In some embodiments, a duplex formed between a sense and antisense strand is 14 nucleotides in length. In some embodiments, a duplex formed between a sense and antisense strand is 15 nucleotides in length. In some embodiments, a duplex formed between a sense and antisense strand is 16 nucleotides in length. In some embodiments, a duplex formed between a sense and antisense strand is 17 nucleotides in length. In some embodiments, a duplex formed between a sense and antisense strand is 18 nucleotides in length. In some embodiments, a duplex formed between a sense and antisense strand is 19 nucleotides in length. In some embodiments, a duplex formed between a sense and antisense strand is 20 nucleotides in length. In some embodiments, a duplex formed between a sense and antisense strand is 21 nucleotides in length. In some embodiments, a duplex formed between a sense and antisense strand is 22 nucleotides in length. In some embodiments, a duplex formed between a sense and antisense strand is 23 nucleotides in length. In some embodiments, a duplex formed between a sense and antisense strand is 24 nucleotides in length. In some embodiments, a duplex formed between a sense and antisense strand is 25 nucleotides in length. In some embodiments, a duplex formed between a sense and antisense strand is 26 nucleotides in length. In some embodiments, a duplex formed between a sense and antisense strand is 27 nucleotides in length. In some embodiments, a duplex formed between a sense and antisense strand is 28 nucleotides in length. In some embodiments, a duplex formed between a sense and antisense strand is 29 nucleotides in length. In some embodiments, a duplex formed between a sense and antisense strand is 30 nucleotides in length. In some embodiments, a duplex formed between a sense and antisense strand does not span the entire length of the sense strand and/or antisense strand. In some embodiments, a duplex between a sense and antisense strand spans the entire length of either the sense or antisense strands. In some embodiments, a duplex between a sense and antisense strand spans the entire length of both the sense strand and the antisense strand. [00206] In some embodiments, a duplex between a sense and antisense strand spans the entire length of both the sense strand and the antisense strand. In some embodiments, the sense and antisense strands of an oligonucleotide comprise nucleotides sequences selected from the group consisting of: (a) SEQ ID NOs: 861 and 951, respectively; (b) SEQ ID NOs: 857 and 947, respectively; (c) SEQ ID NOs: 858 and 948, respectively; (d) SEQ ID NOs: 859 and 949, respectively; (e) SEQ ID NOs: 860 and 950, respectively; (f) SEQ ID NOs: 862 and 952, respectively; (g) SEQ ID NOs: 863 and 953, respectively; (h) SEQ ID NOs: 864 and 954, respectively; (i) SEQ ID NOs: 865 and 955, respectively; (j) SEQ ID NOs: 866 and 956, respectively; (k) SEQ ID NOs: 867 and 957, respectively; (l) SEQ ID NOs: 868 and 958, respectively; (m) SEQ ID NOs: 869 and 959, respectively; (n) SEQ ID NOs: 870 and 960, respectively; (o) SEQ ID NOs: 871 and 961, respectively; (p) SEQ ID NOs: 872 and 962, respectively; (q) SEQ ID NOs: 873 and 963, respectively; (r) SEQ ID NOs: 874 and 964, respectively; (s) SEQ ID NOs: 875 and 965, respectively; (t) SEQ ID NOs: 876 and 966, respectively; (u) SEQ ID NOs: 877 and 967, respectively; (v) SEQ ID NOs: 878 and 968, respectively; (w) SEQ ID NOs: 879 and 969, respectively; (x) SEQ ID NOs: 880 and 970, respectively; (y) SEQ ID NOs: 881and 971, respectively; (z) SEQ ID NOs: 882 and 972, respectively; (aa) SEQ ID NOs: 883 and 973, respectively; (bb) SEQ ID NOs: 884 and 974, respectively; (cc) SEQ ID NOs: 885 and 975, respectively; (dd) SEQ ID NOs: 886 and 976, respectively; (ee) SEQ ID NOs: 887 and 977, respectively; (ff) SEQ ID NOs: 888 and 978, respectively; (gg) SEQ ID NOs: 940 and 1030, respectively; (hh) SEQ ID NOs: 896 and 986, respectively; and (ii) SEQ ID NOs: 920 and 1010, respectively, wherein a duplex formed between a sense and antisense strand is in the range of 12-30 nucleotides in length (e.g., 12 to 30, 12 to 27, 12 to 22, 15 to 25, 18 to 30, 18 to 22, 18 to 25, 18 to 27, 18 to 30, 19 to 30 or 21 to 30 nucleotides in length). [00207] In some embodiments, a duplex between a sense and antisense strand spans the entire length of both the sense strand and the antisense strand. In some embodiments, the sense and antisense strands of an oligonucleotide comprise nucleotides sequences selected from the group consisting of: (a) SEQ ID NOs: 857 and 947, respectively; (b) SEQ ID NOs: 858 and 948, respectively; (c) SEQ ID NOs: 859 and 949, respectively; (d) SEQ ID NOs: 860 and 950, respectively; (e) SEQ ID NOs: 862 and 952, respectively; (f) SEQ ID NOs: 867 and 957, respectively; (g) SEQ ID NOs: 875 and 965, respectively; and (h) SEQ ID NOs: 876 and 966, respectively, wherein a duplex formed between a sense and antisense strand is in the range of 12-30 nucleotides in length (e.g., 12 to 30, 12 to 27, 12 to 22, 15 to 25, 18 to 30, 18 to 22, 18 to 25, 18 to 27, 18 to 30, 19 to 30 or 21 to 30 nucleotides in length). [00208] In some embodiments, a duplex between a sense and antisense strand spans the entire length of both the sense strand and the antisense strand. In some embodiments, the sense and antisense strands of an oligonucleotide comprise nucleotides sequences selected from the group consisting of: (a) SEQ ID NOs: 9 and 10, respectively; (b) SEQ ID NOs: 37 and 38, respectively; (c) SEQ ID NOs: 65 and 66, respectively; and (d) SEQ ID NOs: 69 and 70, respectively. wherein a duplex formed between a sense and antisense strand is in the range of 12-30 nucleotides in length (e.g., 12 to 30, 12 to 27, 12 to 22, 15 to 25, 18 to 30, 18 to 22, 18 to 25, 18 to 27, 18 to 30, 19 to 30 or 21 to 30 nucleotides in length). [00209] In some embodiments, a duplex between a sense and antisense strand spans the entire length of both the sense strand and the antisense strand. In some embodiments, the sense and antisense strands of an oligonucleotide comprise nucleotides sequences selected from the group consisting of: (a) SEQ ID NOs: 901 and 991, respectively; (b) SEQ ID NOs: 910 and 1000, respectively; (c) SEQ ID NOs: 899 and 989, respectively; (d) SEQ ID NOs: 896 and 986, respectively; (e) SEQ ID NOs: 892 and 982, respectively; (f) SEQ ID NOs: 890 and 980, respectively; and (g) SEQ ID NOs: 889 and 979, respectively. wherein a duplex formed between a sense and antisense strand is in the range of 12-30 nucleotides in length (e.g., 12 to 30, 12 to 27, 12 to 22, 15 to 25, 18 to 30, 18 to 22, 18 to 25, 18 to 27, 18 to 30, 19 to 30 or 21 to 30 nucleotides in length). [00210] In some embodiments, a duplex between a sense and antisense strand spans the entire length of both the sense strand and the antisense strand. In some embodiments, the sense and antisense strands of an oligonucleotide comprise nucleotides sequences selected from the group consisting of: (a) SEQ ID NOs: 940 and 1030, respectively; (b) SEQ ID NOs: 937 and 1027, respectively; and (c) SEQ ID NOs: 939 and 1029, respectively. wherein a duplex formed between a sense and antisense strand is in the range of 12-30 nucleotides in length (e.g., 12 to 30, 12 to 27, 12 to 22, 15 to 25, 18 to 30, 18 to 22, 18 to 25, 18 to 27, 18 to 30, 19 to 30 or 21 to 30 nucleotides in length). [00211] In some embodiments, a duplex between a sense and antisense strand spans the entire length of both the sense strand and the antisense strand. In some embodiments, the sense and antisense strands of an oligonucleotide comprise nucleotides sequences selected from the group consisting of: (a) SEQ ID NOs: 915 and 1005, respectively; (b) SEQ ID NOs: 924 and 1014, respectively; (c) SEQ ID NOs: 913 and 1003, respectively; and (d) SEQ ID NOs: 920 and 1010, respectively. wherein a duplex formed between a sense and antisense strand is in the range of 12-30 nucleotides in length (e.g., 12 to 30, 12 to 27, 12 to 22, 15 to 25, 18 to 30, 18 to 22, 18 to 25, 18 to 27, 18 to 30, 19 to 30 or 21 to 30 nucleotides in length). Oligonucleotide Termini [00212] In some embodiments, an oligonucleotide disclosed herein (e.g., an RNAi oligonucleotide) comprises a sense strand and an antisense strand, wherein the termini of either or both strands comprise a blunt end. In some embodiments, an oligonucleotide herein comprises sense and antisense strands that are separate strands which form an asymmetric duplex region having an overhang at the 3’ terminus of the antisense strand. In some embodiments, an oligonucleotide herein comprises a sense strand and an antisense strand, wherein the termini of either or both strands comprise an overhang comprising one or more nucleotides. In some embodiments, the one or more nucleotides comprising the overhang are unpaired nucleotides. In some embodiments, an oligonucleotide herein comprises a sense strand and an antisense strand, wherein the 3’ termini of the sense strand and the 5’ termini of the antisense strand comprise a blunt end. In some embodiments, an oligonucleotide herein comprises a sense strand and an antisense strand, wherein the 5’ termini of the sense strand and the 3’ termini of the antisense strand comprise a blunt end. [00213] In some embodiments, an oligonucleotide herein comprises a sense strand and an antisense strand, wherein the 3’ terminus of either or both strands comprise a 3’-overhang comprising one or more nucleotides. In some embodiments, an oligonucleotide herein comprises a sense strand and an antisense strand, wherein the sense strand comprises a 3’- overhang comprising one or more nucleotides. In some embodiments, an oligonucleotide herein comprises a sense strand and an antisense strand, wherein the antisense strand comprises a 3’-overhang comprising one or more nucleotides. In some embodiments, an oligonucleotide herein comprises a sense strand and an antisense strand, wherein both the sense strand and the antisense strand comprises a 3’-overhang comprising one or more nucleotides. [00214] In some embodiments, the 3’-overhang is about one (1) to twenty (20) nucleotides in length (e.g., about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or about 20 nucleotides in length). In some embodiments, the 3’ overhang is about one (1) to nineteen (19), one (1) to eighteen (18), one (1) to seventeen (17), one (1) to sixteen (16), one (1) to fifteen (15), one (1) to fourteen (14), one (1) to thirteen (13), one (1) to twelve (12), one (1) to eleven (11), one (1) to ten (10), one (1) to nine (9), one (1) to eight (8), one (1) to seven (7), one (1) to six (6), one (1) to five (5), one (1) to four (4), one (1) to three (3), or about one (1) to two (2) nucleotides in length. In some embodiments, the 3’-overhang is (1) nucleotide in length. In some embodiments, the 3’-overhang is two (2) nucleotides in length. In some embodiments, the 3’-overhang is three (3) nucleotides in length. In some embodiments, the 3’-overhang is four (4) nucleotides in length. In some embodiments, the 3’- overhang is five (5) nucleotides in length. In some embodiments, the 3’-overhang is six (6) nucleotides in length. In some embodiments, the 3’-overhang is seven (7) nucleotides in length. In some embodiments, the 3’-overhang is eight (8) nucleotides in length. In some embodiments, the 3’-overhang is nine (9) nucleotides in length. In some embodiments, the 3’- overhang is ten (10) nucleotides in length. In some embodiments, the 3’-overhang is eleven (11) nucleotides in length. In some embodiments, the 3’-overhang is twelve (12) nucleotides in length. In some embodiments, the 3’-overhang is thirteen (13) nucleotides in length. In some embodiments, the 3’-overhang is fourteen (14) nucleotides in length. In some embodiments, the 3’-overhang is fifteen (15) nucleotides in length. In some embodiments, the 3’-overhang is sixteen (16) nucleotides in length. In some embodiments, the 3’-overhang is seventeen (17) nucleotides in length. In some embodiments, the 3’-overhang is eighteen (18) nucleotides in length. In some embodiments, the 3’-overhang is nineteen (19) nucleotides in length. In some embodiments, the 3’-overhang is twenty (20) nucleotides in length. [00215] In some embodiments, an oligonucleotide herein comprises a sense strand and an antisense strand, wherein the 5’ terminus of either or both strands comprise a 5’-overhang comprising one or more nucleotides. In some embodiments, an oligonucleotide herein comprises a sense strand and an antisense strand, wherein the sense strand comprises a 5’- overhang comprising one or more nucleotides. In some embodiments, an oligonucleotide herein comprises a sense strand and an antisense strand, wherein the antisense strand comprises a 5’-overhang comprising one or more nucleotides. In some embodiments, an oligonucleotide herein comprises a sense strand and an antisense strand, wherein both the sense strand and the antisense strand comprises a 5’-overhang comprising one or more nucleotides. [00216] In some embodiments, the 5’-overhang is about one (1) to twenty (20) nucleotides in length (e.g., about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or about 20 nucleotides in length). In some embodiments, the 5’ overhang is about one (1) to nineteen (19), one (1) to eighteen (18), one (1) to seventeen (17), one (1) to sixteen (16), one (1) to fifteen (15), one (1) to fourteen (14), one (1) to thirteen (13), one (1) to twelve (12), one (1) to eleven (11), one (1) to ten (10), one (1) to nine (9), one (1) to eight (8), one (1) to seven (7), one (1) to six (6), one (1) to five (5), one (1) to four (4), one (1) to three (3), or about one (1) to two (2) nucleotides in length. In some embodiments, the 5’-overhang is (1) nucleotide in length. In some embodiments, the 5’-overhang is two (2) nucleotides in length. In some embodiments, the 5’-overhang is three (3) nucleotides in length. In some embodiments, the 5’-overhang is four (4) nucleotides in length. In some embodiments, the 5’- overhang is five (5) nucleotides in length. In some embodiments, the 5’-overhang is six (6) nucleotides in length. In some embodiments, the 5’-overhang is seven (7) nucleotides in length. In some embodiments, the 5’-overhang is eight (8) nucleotides in length. In some embodiments, the 5’-overhang is nine (9) nucleotides in length. In some embodiments, the 5’- overhang is ten (10) nucleotides in length. In some embodiments, the 5’-overhang is eleven (11) nucleotides in length. In some embodiments, the 5’-overhang is twelve (12) nucleotides in length. In some embodiments, the 5’-overhang is thirteen (13) nucleotides in length. In some embodiments, the 5’-overhang is fourteen (14) nucleotides in length. In some embodiments, the 5’-overhang is fifteen (15) nucleotides in length. In some embodiments, the 5’-overhang is sixteen (16) nucleotides in length. In some embodiments, the 5’-overhang is seventeen (17) nucleotides in length. In some embodiments, the 5’-overhang is eighteen (18) nucleotides in length. In some embodiments, the 5’-overhang is nineteen (19) nucleotides in length. In some embodiments, the 5’-overhang is twenty (20) nucleotides in length. [00217] In some embodiments, one or more (e.g., 2, 3, 4, 5, or more) nucleotides comprising the 3’ terminus or 5’ terminus of a sense and/or antisense strand are modified. For example, in some embodiments, one or two terminal nucleotides of the 3’ terminus of the antisense strand are modified. In some embodiments, the last nucleotide at the 3’ terminus of an antisense strand is modified, such that it comprises 2’ modification, or it comprises, a 2’- O-methoxyethyl. In some embodiments, the last one or two terminal nucleotides at the 3’ terminus of an antisense strand are complementary with the target. In some embodiments, the last one or two nucleotides at the 3’ terminus of the antisense strand are not complementary with the target. [00218] In some embodiments, an oligonucleotide disclosed herein (e.g., an RNAi oligonucleotide) comprises a sense strand and an antisense strand, wherein the 3’ terminus of the sense strand comprises a step-loop described herein and the 3’ terminus of the antisense strand comprises a 3’-overhang described herein. In some embodiments, an oligonucleotide herein (e.g., an RNAi oligonucleotide) comprises a sense strand and an antisense strand that form a nicked tetraloop structure described herein, wherein the 3’ terminus of the sense strand comprises a stem-loop, wherein the loop is a tetraloop described herein, and wherein the 3’ terminus of the antisense strand comprises a 3’-overhang described herein. In some embodiments, the 3’-overhang is two (2) nucleotides in length. In some embodiments, the two (2) nucleotides comprising the 3’-overhang both comprise guanine (G) nucleobases. Typically, one or both of the nucleotides comprising the 3’-overhang of the antisense strand are not complementary with the target mRNA. Oligonucleotide Modifications a. Sugar Modifications [00219] In some embodiments, a modified sugar (also referred herein to a sugar analog) includes a modified deoxyribose or ribose moiety in which, for example, one or more modifications occur at the 2′, 3′, 4′ and/or 5′ carbon position of the sugar. In some embodiments, a modified sugar may also include non-natural alternative carbon structures such as those present in locked nucleic acids (“LNA”; see, e.g., Koshkin et al., (1998) TETRAHEDON 54:3607-3630), unlocked nucleic acids (“UNA”; see, e.g., Snead et al., (2013) MOL. THER-NUCL. ACIDS 2:e103) and bridged nucleic acids (“BNA”; see, e.g., Imanishi and Obika (2002) CHEM COMMUN. (CAMB) 21:1653-1659). [00220] In some embodiments, a nucleotide modification in a sugar comprises a 2′- modification. In some embodiments, a 2′-modification may be 2′-O-propargyl, 2′-O- propylamin, 2′-amino, 2′-ethyl, 2′-fluoro (2′-F), 2′-aminoethyl (EA), 2′-O-methyl (2′-OMe), 2′-O-methoxyethyl (2′-MOE), 2′-O-[2-(methylamino)-2-oxoethyl] (2′-O-NMA) or 2′-deoxy- 2′-fluoro-β-d-arabinonucleic acid (2′-FANA). In some embodiments, the modification is 2′- F, 2′-OMe or 2′-MOE. In some embodiments, a modification in a sugar comprises a modification of the sugar ring, which may comprise modification of one or more carbons of the sugar ring. For example, a modification of a sugar of a nucleotide may comprise a 2′- oxygen of a sugar is linked to a 1′-carbon or 4′-carbon of the sugar, or a 2′-oxygen is linked to the 1′-carbon or 4′-carbon via an ethylene or methylene bridge. In some embodiments, a modified nucleotide has an acyclic sugar that lacks a 2′-carbon to 3′-carbon bond. In some embodiments, a modified nucleotide has a thiol group, e.g., in the 4′ position of the sugar. [00221] In some embodiments, the oligonucleotide described herein comprises at least about 1 modified nucleotide (e.g., at least 1, at least 5, at least 10, at least 15, at least 20, at least 25, at least 30, at least 35, at least 40, at least 45, at least 50, at least 55, at least 60, or more). In some embodiments, the sense strand of the oligonucleotide comprises at least about 1 modified nucleotide (e.g., at least 1, at least 5, at least 10, at least 15, at least 20, at least 25, at least 30, at least 35, or more). In some embodiments, the antisense strand of the oligonucleotide comprises at least about 1 modified nucleotide (e.g., at least 1, at least 5, at least 10, at least 15, at least 20, or more). [00222] In some embodiments, all the nucleotides of the sense strand of the oligonucleotide are modified. In some embodiments, all the nucleotides of the antisense strand of the oligonucleotide are modified. In some embodiments, all the nucleotides of the oligonucleotide (i.e., both the sense strand and the antisense strand) are modified. In some embodiments, the modified nucleotide comprises a 2′-modification (e.g., a 2′-F or 2′-OMe, 2′-MOE, and 2′-deoxy-2′-fluoro-β-d-arabinonucleic acid). In some embodiments, the modified nucleotide comprises a 2′-modification (e.g., a 2′-F or 2′-OMe). [00223] In some embodiments, the disclosure provides oligonucleotides having different modification patterns. In some embodiments, an oligonucleotide herein comprises a sense strand having a modification pattern as set forth in the Examples and Sequence Listing and an antisense strand having a modification pattern as set forth in the Examples and Sequence Listing. [00224] In some embodiments, an oligonucleotide disclosed herein (e.g., an RNAi oligonucleotide) comprises an antisense strand having nucleotides that are modified with 2′- F. In some embodiments, an oligonucleotide herein comprises an antisense strand comprising nucleotides that are modified with 2′-F and 2′-OMe. In some embodiments, an oligonucleotide disclosed herein comprises a sense strand having nucleotides that are modified with 2′-F. In some embodiments, an oligonucleotide disclosed herein comprises a sense strand comprises nucleotides that are modified with 2′-F and 2′-OMe. [00225] In some embodiments, an oligonucleotide described herein comprises a sense strand with about 10-15%, 10%, 11%, 12%, 13%, 14% or 15% of the nucleotides of the sense strand comprising a 2’-fluoro modification. In some embodiments, about 11% of the nucleotides of the sense strand comprise a 2-fluoro modification. In some embodiments, an oligonucleotide described herein comprises an antisense strand with about 25-35%, 25%, 26%, 27%, 28%, 29%, 30%, 31%, 32%, 33%, 34% or 35% of the nucleotides of the antisense strand comprising a 2’-fluoro modification. In some embodiments, about 32% of the nucleotides of the antisense strand comprise a 2’-fluoro modification. In some embodiments, the oligonucleotide has about 15-25%, 15%, 16%, 17%, 18%, 19%, 20%, 21%, 22%, 23%, 24%, or 25% of its nucleotides comprising a 2’-fluoro modification. In some embodiments, about 19% of the nucleotides in the dsRNAi oligonucleotide comprise a 2’-fluoro modification. [00226] In some embodiments, the modified oligonucleotides comprise a sense strand sequence having a modification pattern as set forth in FIG 1A or Example 7 and an antisense strand having a modification pattern as set forth in FIG 1A or Example 7. In some embodiments, for these oligonucleotides, one or more of positions 8, 9, 10 or 11 of the sense strand is modified with a 2′-F group. In other embodiments, for these oligonucleotides, the sugar moiety at each of nucleotides at positions 1-7 and 12-20 in the sense strand is modified with a 2′-OMe. [00227] In some embodiments, the antisense strand has 3 nucleotides that are modified at the 2′-position of the sugar moiety with a 2′-F. In some embodiments, the sugar moiety at positions 2, 5 and 14 and optionally up to 3 of the nucleotides at positions 1, 3, 7 and 10 of the antisense strand are modified with a 2′-F. In some embodiments, the sugar moiety at positions 2, 5 and 14 and optionally up to 3 of the nucleotides at positions 3, 4, 7 and 10 of the antisense strand are modified with a 2′-F. In other embodiments, the sugar moiety at each of the positions at positions 2, 5 and 14 of the antisense strand is modified with the 2′-F. In other embodiments, the sugar moiety at each of the positions at positions 1, 2, 5 and 14 of the antisense strand is modified with the 2′-F. In other embodiments, the sugar moiety at each of the positions at positions 2, 4, 5 and 14 of the antisense strand is modified with the 2′-F. In still other embodiments, the sugar moiety at each of the positions at positions 1, 2, 3, 5, 7 and 14 of the antisense strand is modified with the 2′-F. In other embodiments, the sugar moiety at each of the positions at positions 2, 3, 4, 5, 7 and 14 of the antisense strand is modified with the 2′-F. In yet another embodiment, the sugar moiety at each of the positions at positions 1, 2, 3, 5, 10 and 14 of the antisense strand is modified with the 2′-F. In other embodiments, the sugar moiety at each of the positions at positions 2, 3, 4, 5, 10 and 14 of the antisense strand is modified with the 2′-F. In another embodiment, the sugar moiety at each of the positions at positions 2, 3, 5, 7, 10 and 14 of the antisense strand is modified with the 2′-F. In yet another embodiment, the sugar moiety at each of the positions at positions 2, 3, 4, 5, 7, 10 and 14 of the antisense strand is modified with the 2′-F. [00228] In some embodiments, an oligonucleotide provided herein comprises an antisense strand having the sugar moiety at position 1, position 2, position 3, position 4, position 5, position 6, position 7, position 8, position 9, position 10, position 11, position 12, position 13, position 14, position 15, position 16, position 17, position 18, position 19, position 20, position 21, or position 22 modified with 2′-F. [00229] In some embodiments, an oligonucleotide provided herein comprises an antisense strand having the sugar moiety at position 1, position 2, position 3, position 4, position 5, position 6, position 7, position 8, position 9, position 10, position 11, position 12, position 13, position 14, position 15, position 16, position 17, position 18, position 19, position 20, position 21, or position 22 modified with 2′-OMe. [00230] In some embodiments, an oligonucleotide provided herein comprises an antisense strand having the sugar moiety at position 1, position 2, position 3, position 4, position 5, position 6, position 7, position 8, position 9, position 10, position 11, position 12, position 13, position 14, position 15, position 16, position 17, position 18, position 19, position 20, position 21, or position 22 modified with a modification selected from the group consisting of 2′-O-propargyl, 2′-O-propylamin, 2′-amino, 2′-ethyl, 2’-aminoethyl (EA), 2′-O- methyl (2′-OMe), 2′-O-methoxyethyl (2′-MOE), 2′-O-[2-(methylamino)-2-oxoethyl] (2′-O- NMA), and 2′-deoxy-2′-fluoro-β-d-arabinonucleic acid (2′-FANA). [00231] In some embodiments, an oligonucleotide provided herein comprises a sense strand having the sugar moiety at positions 8-11 modified with 2′-F. In some embodiments, an oligonucleotide provided herein comprises a sense strand having the sugar moiety at positions 3, 8, 9, 10, 12, 13 and 17 modified with 2′-F. In some embodiments, an oligonucleotide provided herein comprises a sense strand having the sugar moiety at positions 1-7 and 12-17 or 12-20 modified with 2’OMe. In some embodiments, an oligonucleotide provided herein comprises a sense strand having the sugar moiety at positions 1-7, 12-27 and 31-36 modified with 2’OMe. In some embodiments, an oligonucleotide provided herein comprises a sense strand having the sugar moiety of each of the nucleotides at positions 1-7 and 12-17 or 12-20 of the sense strand modified with a modification selected from the group consisting of 2′-O-propargyl, 2′-O-propylamin, 2′-amino, 2′-ethyl, 2’-aminoethyl (EA), 2′-O- methyl (2′-OMe), 2′-O-methoxyethyl (2′-MOE), 2′-O-[2-(methylamino)-2-oxoethyl] (2′-O- NMA), and 2′-deoxy-2′-fluoro-β-d-arabinonucleic acid (2′-FANA). In some embodiments, an oligonucleotide provided herein comprises a sense strand having the sugar moiety at positions 1-2, 4-7, 11, 14-16 and 18-20 modified with 2’OMe. In some embodiments, an oligonucleotide provided herein comprises a sense strand having the sugar moiety of each of the nucleotides at positions 1-2, 4-7, 11, 14-16 and 18-20 of the sense strand modified with a modification selected from the group consisting of 2′-O-propargyl, 2′-O-propylamin, 2′- amino, 2′-ethyl, 2’-aminoethyl (EA), 2′-O-methyl (2′-OMe), 2′-O-methoxyethyl (2′-MOE), 2′-O-[2-(methylamino)-2-oxoethyl] (2′-O-NMA), and 2′-deoxy-2′-fluoro-β-d-arabinonucleic acid (2′-FANA). [00232] In some embodiments, an oligonucleotide provided herein comprises a sense strand having the sugar moiety at position 1, position 2, position 3, position 4, position 5, position 6, position 7, position 8, position 9, position 10, position 11, position 12, position 13, position 14, position 15, position 16, position 17, position 18, position 19, position 20, position 21, position 22, position 23, position 24, position 25, position 26, position 27, position 28, position 29, position 30, position 31, position 32, position 33, position 34, position 35, or position 36 modified with 2′-F. [00233] In some embodiments, an oligonucleotide provided herein comprises a sense strand having the sugar moiety at position 1, position 2, position 3, position 4, position 5, position 6, position 7, position 8, position 9, position 10, position 11, position 12, position 13, position 14, position 15, position 16, position 17, position 18, position 19, position 20, position 21, position 22, position 23, position 24, position 25, position 26, position 27, position 28, position 29, position 30, position 31, position 32, position 33, position 34, position 35, or position 36 modified with 2′-OMe. [00234] In some embodiments, an oligonucleotide provided herein comprises a sense strand having the sugar moiety at position 1, position 2, position 3, position 4, position 5, position 6, position 7, position 8, position 9, position 10, position 11, position 12, position 13, position 14, position 15, position 16, position 17, position 18, position 19, position 20, position 21, position 22, position 23, position 24, position 25, position 26, position 27, position 28, position 29, position 30, position 31, position 32, position 33, position 34, position 35, or position 36 modified with a modification selected from the group consisting of 2′-O-propargyl, 2′-O-propylamin, 2′-amino, 2′-ethyl, 2’-aminoethyl (EA), 2′-O-methyl (2′- OMe), 2′-O-methoxyethyl (2′-MOE), 2′-O-[2-(methylamino)-2-oxoethyl] (2′-O-NMA), and 2′-deoxy-2′-fluoro-β-d-arabinonucleic acid (2′-FANA). b. 5′ Terminal Phosphates [00235] In some embodiments, 5′-terminal phosphate groups of oligonucleotides enhance the interaction with Ago2. However, oligonucleotides comprising a 5′-phosphate group may be susceptible to degradation via phosphatases or other enzymes, which can limit their bioavailability in vivo. In some embodiments, oligonucleotides include analogs of 5′ phosphates that are resistant to such degradation. In some embodiments, a phosphate analog may be oxymethylphosphonate, vinylphosphonate or malonyl phosphonate. In certain embodiments, the 1′ end of an oligonucleotide strand is attached to chemical moiety that mimics the electrostatic and steric properties of a natural 5′-phosphate group (“phosphate mimic”). [00236] In some embodiments, an oligonucleotide has a phosphate analog at a 4′- carbon position of the sugar (referred to as a “4′-phosphate analog”). See, e.g., Intl. Patent Application Publication No. WO 2018/045317. In some embodiments, an oligonucleotide herein comprises a 4′-phosphate analog at a 5′-terminal nucleotide. In some embodiments, a phosphate analog is an oxymethylphosphonate, in which the oxygen atom of the oxymethyl group is bound to the sugar moiety (e.g., at its 4′-carbon) or analog thereof. In other embodiments, a 4′-phosphate analog is a thiomethyl phosphonate or an amino methyl phosphonate, in which the sulfur atom of the thiomethyl group or the nitrogen atom of the amino methyl group is bound to the 4′-carbon of the sugar moiety or analog thereof. In certain embodiments, a 4′-phosphate analog is an oxymethyl phosphonate. In some embodiments, an oxymethyl phosphonate is represented by the formula –O–CH2–PO(OH)2 or –O–CH2–PO(OR)2, in which R is independently selected from H, CH3, an alkyl group, CH2CH2CN, CH2OCOC(CH3)3, CH2OCH2CH2Si (CH3)3 or a protecting group. In certain embodiments, the alkyl group is CH2CH3. More typically, R is independently selected from H, CH3 or CH2CH3. [00237] In some embodiments, an oligonucleotide provided herein comprises an antisense strand comprising a 4′-phosphate analog at the 5′-terminal nucleotide, wherein 5’- terminal nucleotide comprises the following structure (Chem 1):
Figure imgf000071_0001
4’-O-monomethylphosphonate-2’-O-methyluridine phosphorothioate [MePhosphonate- 4O-mUs]. Chem 1 c. Modified Internucleotide Linkages [00238] In some embodiments, an oligonucleotide may comprise a modified internucleoside linkage. In some embodiments, phosphate modifications or substitutions may result in an oligonucleotide that comprises at least about 1 (e.g., at least 1, at least 2, at least 3 or at least 5) modified internucleotide linkage. In some embodiments, any one of the oligonucleotides disclosed herein comprises about 1 to about 10 (e.g., 1 to 10, 2 to 8, 4 to 6, 3 to 10, 5 to 10, 1 to 5, 1 to 3 or 1 to 2) modified internucleotide linkages. In some embodiments, any one of the oligonucleotides disclosed herein comprises 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10 modified internucleotide linkages. [00239] A modified internucleotide linkage may be a phosphorodithioate linkage, 4′- O-methylene phosphonate linkage, a phosphorothioate linkage, a phosphotriester linkage, a thionoalkylphosphonate linkage, a thionalkylphosphotriester linkage, a phosphoramidite linkage, a phosphonate linkage or a boranophosphate linkage. In some embodiments, at least one modified internucleotide linkage of any one of the oligonucleotides as disclosed herein is a phosphorothioate linkage. In some embodiments, at least one modified internucleotide linkage of any one of the oligonucleotides as disclosed herein is a 4′-O-methylene phosphonate linkage. [00240] In some embodiments, the oligonucleotide described herein has a phosphorothioate linkage between one or more of positions 1 and 2 of the sense strand, positions 1 and 2 of the antisense strand, positions 2 and 3 of the antisense strand, positions 3 and 4 of the antisense strand, positions 20 and 21 of the antisense strand, and positions 21 and 22 of the antisense strand. In some embodiments, the oligonucleotide described herein has a phosphorothioate linkage between each of positions 1 and 2 of the sense strand, positions 1 and 2 of the antisense strand, positions 2 and 3 of the antisense strand, positions 20 and 21 of the antisense strand, and positions 21 and 22 of the antisense strand. d. Base Modifications [00241] In some embodiments, oligonucleotides herein have one or more modified nucleobases. In some embodiments, modified nucleobases (also referred to herein as base analogs) are linked at the 1′ position of a nucleotide sugar moiety. In certain embodiments, a modified nucleobase is a nitrogenous base. In certain embodiments, a modified nucleobase does not contain nitrogen atom. See, e.g., US Patent Application Publication No. 2008/0274462. In some embodiments, a modified nucleotide comprises a universal base. However, in certain embodiments, a modified nucleotide does not contain a nucleobase (abasic). [00242] In some embodiments, a universal base is a heterocyclic moiety located at the 1′ position of a nucleotide sugar moiety in a modified nucleotide, or the equivalent position in a nucleotide sugar moiety substitution, that, when present in a duplex, can be positioned opposite more than one type of base without substantially altering structure of the duplex. In some embodiments, compared to a reference single-stranded nucleic acid (e.g., oligonucleotide) that is fully complementary to a target nucleic acid, a single-stranded nucleic acid containing a universal base forms a duplex with the target nucleic acid that has a lower Tm than a duplex formed with the complementary nucleic acid. However, in some embodiments, when compared to a reference single-stranded nucleic acid in which the universal base has been replaced with a base to generate a single mismatch, the single- stranded nucleic acid containing the universal base forms a duplex with the target nucleic acid that has a higher Tm than a duplex formed with the nucleic acid comprising the mismatched base. [00243] Non-limiting examples of universal-binding nucleotides include, but are not limited to, inosine, 1-β-D-ribofuranosyl-5-nitroindole and/or 1-β-D-ribofuranosyl-3- nitropyrrole (see, US Patent Application Publication No.2007/0254362; Van Aerschot et al., (1995) NUCLEIC ACIDS RES.23:4363-4370; Loakes et al., (1995) NUCLEIC ACIDS RES. 23:2361-66; and Loakes and Brown (1994) NUCLEIC ACIDS RES.22:4039-43). e. Reversible Modifications [00244] While certain modifications to protect an oligonucleotide from the in vivo environment before reaching target cells can be made, they can reduce the potency or activity of the oligonucleotide once it reaches the cytosol of the target cell. Reversible modifications can be made such that the molecule retains desirable properties outside of the cell, which are then removed upon entering the cytosolic environment of the cell. Reversible modification can be removed, for example, by the action of an intracellular enzyme or by the chemical conditions inside of a cell (e.g., through reduction by intracellular glutathione). [00245] In some embodiments, a reversibly modified nucleotide comprises a glutathione-sensitive moiety. Typically, nucleic acid molecules have been chemically modified with cyclic disulfide moieties to mask the negative charge created by the internucleotide diphosphate linkages and improve cellular uptake and nuclease resistance. See US Patent Application Publication No.2011/0294869, Intl. Patent Application Publication Nos. WO 2014/088920 and WO 2015/188197, and Meade et al., (2014) NAT. BIOTECHNOL.32:1256-63. This reversible modification of the internucleotide diphosphate linkages is designed to be cleaved intracellularly by the reducing environment of the cytosol (e.g., glutathione). Earlier examples include neutralizing phosphotriester modifications that were reported to be cleavable inside cells (see, Dellinger et al., (2003) J. AM. CHEM. SOC. 125:940-50). [00246] In some embodiments, such a reversible modification allows protection during in vivo administration (e.g., transit through the blood and/or lysosomal/endosomal compartments of a cell) where the oligonucleotide will be exposed to nucleases and other harsh environmental conditions (e.g., pH). When released into the cytosol of a cell where the levels of glutathione are higher compared to extracellular space, the modification is reversed, and the result is a cleaved oligonucleotide. Using reversible, glutathione-sensitive moieties, it is possible to introduce sterically larger chemical groups into the oligonucleotide of interest when compared to the options available using irreversible chemical modifications. This is because these larger chemical groups will be removed in the cytosol and, therefore, should not interfere with the biological activity of the oligonucleotides inside the cytosol of a cell. As a result, these larger chemical groups can be engineered to confer various advantages to the nucleotide or oligonucleotide, such as nuclease resistance, lipophilicity, charge, thermal stability, specificity, and reduced immunogenicity. In some embodiments, the structure of the glutathione-sensitive moiety can be engineered to modify the kinetics of its release. [00247] In some embodiments, a glutathione-sensitive moiety is attached to the sugar of the nucleotide. In some embodiments, a glutathione-sensitive moiety is attached to the 2′- carbon of the sugar of a modified nucleotide. In some embodiments, the glutathione- sensitive moiety is located at the 5′-carbon of a sugar, particularly when the modified nucleotide is the 5′-terminal nucleotide of the oligonucleotide. In some embodiments, the glutathione-sensitive moiety is located at the 3′-carbon of sugar, particularly when the modified nucleotide is the 3′-terminal nucleotide of the oligonucleotide. In some embodiments, the glutathione-sensitive moiety comprises a sulfonyl group. See, e.g., US Provisional Patent Application No.62/378,635, entitled Compositions Comprising Reversibly Modified Oligonucleotides and Uses Thereof, which was filed on August 23, 2016. Targeting Ligands [00248] In some embodiments, it is desirable to target the STAT3 targeting oligonucleotides of the disclosure to one or more cells or one or more organs. Such a strategy can help to avoid undesirable effects in other organs or avoid undue loss of the oligonucleotide to cells, tissue or organs that would not benefit from the oligonucleotide. Targeting of oligonucleotides to one or more cells or one or more organs can be achieved through a variety of approaches. Conjugation of oligonucleotides to tissue or cell specific antibodies, small molecules or targeting ligands can facilitate delivery to and modify accumulation of the oligonucleotide in one or more target cells or tissues (Chernolovskaya et al., (2019) FRONT PHARMACOL.10:444). For example, conjugation of an oligonucleotide to a saturated fatty acid (e.g., C22) may facilitate delivery to cells or tissues like adipose tissue or immune cells which uptake such ligands more readily than conventional oligonucleotide ligands. Accordingly, in some embodiments, oligonucleotides disclosed herein are modified to facilitate targeting and/or delivery of a tissue, cell, or organ (e.g., to facilitate delivery of the oligonucleotide to the liver). In certain embodiments, oligonucleotides disclosed herein are modified to facilitate delivery of the oligonucleotide to cells of the immune system. In certain embodiments, oligonucleotides disclosed herein are modified to facilitate delivery of the oligonucleotide to myeloid derived suppressor cells. In some embodiments, an oligonucleotide comprises at least one nucleotide (e.g., 1, 2, 3, 4, 5, 6 or more nucleotides) conjugated to one or more targeting ligand(s). [00249] In some embodiments, the targeting ligand comprises a carbohydrate, amino sugar, cholesterol, peptide, polypeptide, protein, or part of a protein (e.g., an antibody or antibody fragment), or lipid. In some embodiments, the targeting ligand is an aptamer. For example, a targeting ligand may be an RGD peptide that is used to target tumor vasculature or glioma cells, CREKA peptide to target tumor vasculature or stoma, transferring, lactoferrin, or an aptamer to target transferrin receptors expressed on CNS vasculature, or an anti-EGFR antibody to target EGFR on glioma cells. In certain embodiments, the targeting ligand is one or more GalNAc moieties. [00250] In some embodiments, 1 or more (e.g., 1, 2, 3, 4, 5 or 6) nucleotides of an oligonucleotide are each conjugated to a separate targeting ligand. In some embodiments, 2 to 4 nucleotides of an oligonucleotide are each conjugated to a separate targeting ligand. In some embodiments, targeting ligands are conjugated to 2 to 4 nucleotides at either ends of the sense or antisense strand (e.g., targeting ligands are conjugated to a 2 to 4 nucleotide overhang or extension on the 5′ or 3′ end of the sense or antisense strand) such that the targeting ligands resemble bristles of a toothbrush and the oligonucleotide resembles a toothbrush. For example, an oligonucleotide may comprise a stem-loop at either the 5′ or 3′ end of the sense strand and 1, 2, 3 or 4 nucleotides of the loop of the stem may be individually conjugated to a targeting ligand. In some embodiments, an oligonucleotide (e.g., a dsRNA) provided by the disclosure comprises a stem-loop at the 3′ end of the sense strand, wherein the loop of the stem-loop comprises a triloop or a tetraloop, and wherein the 3 or 4 nucleotides comprising the triloop or tetraloop, respectfully, are individually conjugated to a targeting ligand. In some embodiments, an oligonucleotide provided by the disclosure (e.g., a RNAi oligonucleotide) comprises a stem-loop at the 3′ terminus of the sense strand, wherein the loop of the stem-loop comprises a tetraloop, and wherein 3 nucleotides of the tetraloop are individually conjugated to a targeting ligand. [00251] GalNAc is a high affinity ligand for the ASGPR, which is primarily expressed on the sinusoidal surface of hepatocyte cells and has a major role in binding, internalizing and subsequent clearing circulating glycoproteins that contain terminal galactose or GalNAc residues (asialoglycoproteins). Conjugation (either indirect or direct) of GalNAc moieties to oligonucleotides of the instant disclosure can be used to target these oligonucleotides to the ASGPR expressed on cells. In some embodiments, an oligonucleotide of the instant disclosure is conjugated to at least one or more GalNAc moieties, wherein the GalNAc moieties target the oligonucleotide to an ASGPR expressed on human liver cells (e.g., human hepatocytes). In some embodiments, the GalNAc moiety target the oligonucleotide to the liver. [00252] In some embodiments, an oligonucleotide of the instant disclosure is conjugated directly or indirectly to a monovalent GalNAc. In some embodiments, the oligonucleotide is conjugated directly or indirectly to more than one monovalent GalNAc (i.e., is conjugated to 2, 3 or 4 monovalent GalNAc moieties, and is typically conjugated to 3 or 4 monovalent GalNAc moieties). In some embodiments, an oligonucleotide is conjugated to one or more bivalent GalNAc, trivalent GalNAc or tetravalent GalNAc moieties. [00253] In some embodiments, 1 or more (e.g., 1, 2, 3, 4, 5 or 6) nucleotides of an oligonucleotide are each conjugated to a GalNAc moiety. In some embodiments, 2 to 4 nucleotides of a tetraloop are each conjugated to a separate GalNAc. In some embodiments, 1 to 3 nucleotides of a triloop are each conjugated to a separate GalNAc. In some embodiments, targeting ligands are conjugated to 2 to 4 nucleotides at either ends of the sense or antisense strand (e.g., ligands are conjugated to a 2 to 4 nucleotide overhang or extension on the 5′ or 3′ end of the sense or antisense strand) such that the GalNAc moieties resemble bristles of a toothbrush and the oligonucleotide resembles a toothbrush. In some embodiments, GalNAc moieties are conjugated to a nucleotide of the sense strand. For example, 4 GalNAc moieties can be conjugated to nucleotides in the tetraloop of the sense strand where each GalNAc moiety is conjugated to 1 nucleotide. [00254] In some embodiments, the tetraloop is any combination of adenine and guanine nucleotides. [00255] In some embodiments, the tetraloop (tetraL) has a monovalent GalNAc moiety attached to any one or more guanine nucleotides of the tetraloop via any linker described herein, as depicted below in Chem 2 (X=heteroatom):
Figure imgf000076_0001
Chem 2 [00256] In some embodiments, the tetraloop (tetraL) has a monovalent GalNAc attached to any one or more adenine nucleotides of the tetraloop via any linker described herein, as depicted below in Chem 3 (X=heteroatom):
Figure imgf000077_0001
Chem 3 [00257] In some embodiments, an oligonucleotide herein comprises a monovalent GalNAc attached to a guanine nucleotide referred to as [ademG-GalNAc] or 2′- aminodiethoxymethanol-Guanine-GalNAc, as depicted below in Chem 4:
Figure imgf000077_0002
[00258] In some embodiments, an oligonucleotide herein comprises a monovalent GalNAc attached to an adenine nucleotide, referred to as [ademA-GalNAc] or 2′- aminodiethoxymethanol-Adenine-GalNAc, as depicted below in Chem 5:
Figure imgf000078_0001
Chem 5 [00259] An example of such conjugation is shown below (Chem 6) for a loop comprising from 5′ to 3′ the nucleotide sequence GAAA (L = linker, X = heteroatom) stem attachment points are shown. Such a loop may be present, for example, at positions 27-30 of the sense strand as shown in FIG.1A. In the chemical formula,
Figure imgf000078_0002
is used to describe an attachment point to the oligonucleotide strand (Chem 6).
Figure imgf000079_0001
Chem 6 [00260] Appropriate methods or chemistry (e.g., click chemistry) can be used to link a targeting ligand to a nucleotide. In some embodiments, a targeting ligand is conjugated to a nucleotide using a click linker. In some embodiments, an acetal-based linker is used to conjugate a targeting ligand to a nucleotide of any one of the oligonucleotides described herein. Acetal-based linkers are disclosed, for example, in Intl. Patent Application Publication No. WO 2016/100401. In some embodiments, the linker is a labile linker. However, in other embodiments, the linker is stable. Examples are shown below for a loop comprising from 5′ to 3′ the nucleotides GAAA, in which GalNAc moieties are attached to nucleotides of the loop using an acetal linker (Chem 7 and Chem 8). Such a loop may be present, for example, at positions 27-30 of the any one of the sense strand as shown in FIG. 1. In the chemical formula,
Figure imgf000080_0001
is an attachment point to the oligonucleotide strand (Chem 7 and Chem 8).
Figure imgf000080_0002
Figure imgf000081_0001
[00261] As mentioned, various appropriate methods or chemistry synthetic techniques (e.g., click chemistry) can be used to link a targeting ligand to a nucleotide. In some embodiments, a targeting ligand is conjugated to a nucleotide using a click linker. In some embodiments, an acetal-based linker is used to conjugate a targeting ligand to a nucleotide of any one of the oligonucleotides described herein. Acetal-based linkers are disclosed, for example, in Intl. Patent Application Publication No. WO 2016/100401. In some embodiments, the linker is a labile linker. However, in other embodiments, the linker is a stable linker. [00262] In some embodiments, a duplex extension (e.g., of up to 3, 4, 5 or 6 bp in length) is provided between a targeting ligand (e.g., a GalNAc moiety) and a dsRNA. In some embodiments, the oligonucleotides herein do not have a GalNAc conjugated thereto. Structure of Conjugated STAT3 Targeting Oligonucleotides [00263] In some embodiments, a STAT3 targeting oligonucleotide described herein comprises a nucleotide sequence having a region of complementarity to a STAT3 mRNA target sequence and one or more targeting ligands, wherein the nucleotide sequence comprises one or more nucleosides (nucleic acids) conjugated with one or more targeting ligands represented by formula I-a:
Figure imgf000082_0001
or a pharmaceutically acceptable salt thereof, wherein: B is a nucleobase or hydrogen; R1 and R2 are independently hydrogen, halogen, RA, -CN, -S(O)R, -S(O)2R, -Si(OR)2R, - Si(OR)R2, or -SiR3; or R1 and R2 on the same carbon are taken together with their intervening atoms to form a 3-7 membered saturated or partially unsaturated ring having 0-3 heteroatoms, independently selected from nitrogen, oxygen, and sulfur; each RA is independently an optionally substituted group selected from C1-6 aliphatic, phenyl, a 4-7 membered saturated or partially unsaturated heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, and sulfur, and a 5-6 membered heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur; each R is independently hydrogen, a suitable protecting group, or an optionally substituted group selected from C1-6 aliphatic, phenyl, a 4-7 membered saturated or partially unsaturated heterocyclic having 1-2 heteroatoms independently selected from nitrogen, oxygen, and sulfur, and a 5-6 membered heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur; or two R groups on the same atom are taken together with their intervening atoms to form a 4-7 membered saturated, partially unsaturated, or heteroaryl ring having 0-3 heteroatoms, independently selected from nitrogen, oxygen, silicon, and sulfur; each targeting ligand is selected from lipid conjugate moiety (LC), carbohydrate, amino sugar or GalNAc; and wherein each LC is independently a lipid conjugate moiety comprising a saturated or unsaturated, straight, or branched C1-50 hydrocarbon chain, wherein 0-10 methylene units of the hydrocarbon chain are independently replaced by -Cy-, -O-, - C(O)NR-, -NR-, -S-, -C(O)-, -C(O)O-, -S(O)-, -S(O)2-, -P(O)OR-, -P(S)OR-; each -Cy- is independently an optionally substituted bivalent ring selected from phenylenyl, an 8-10 membered bicyclic arylenyl, a 4-7 membered saturated or partially unsaturated carbocyclylenyl, a 4-11 membered saturated or partially unsaturated spiro carbocyclylenyl, an 8-10 membered bicyclic saturated or partially unsaturated carbocyclylenyl, a 4-7 membered saturated or partially unsaturated heterocyclylenyl having 1-3 heteroatoms independently selected from nitrogen, oxygen, and sulfur, a 4- 11 membered saturated or partially unsaturated spiro heterocyclylenyl having 1-2 heteroatoms independently selected from nitrogen, oxygen, and sulfur, an 8-10 membered bicyclic saturated or partially unsaturated heterocyclylenyl having 1-2 heteroatoms independently selected from nitrogen, oxygen, and sulfur, a 5-6 membered heteroarylenyl having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, or an 8-10 membered bicyclic heteroarylenyl having 1-5 heteroatoms independently selected from nitrogen, oxygen, or sulfur; n is 1-10; L is a covalent bond or a bivalent saturated or unsaturated, straight or branched C1-50 hydrocarbon chain, wherein 0-10 methylene units of the hydrocarbon chain are independently replaced by -Cy-, -O-, -C(O)NR-, -NR-, -S-, -C(O)-, -C(O)O-, -S(O)-, - S(O)2-, -P(O)OR-, -P(S)OR-, -V1CR2W1-, or
Figure imgf000083_0002
m is 1-50; X1, V1 and W1 are independently -C(R)2-, -OR, -O-, -S-, -Se-, or -NR-; Y is hydrogen, a suitable hydroxyl protecting group,
Figure imgf000083_0001
; R3 is hydrogen, a suitable protecting group, a suitable prodrug, or an optionally substituted group selected from C1-6 aliphatic, phenyl, a 4-7 membered saturated or partially unsaturated heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, and sulfur, and a 5-6 membered heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur; X2 is O, S, or NR; X3 is -O-, -S-, -BH2-, or a covalent bond; Y1 is a linking group attaching to the 2′- or 3′-terminal of a nucleoside, a nucleotide, or an oligonucleotide; Y2 is hydrogen, a suitable protecting group, a phosphoramidite analogue, an internucleotide linking group attaching to the 5′-terminal of a nucleoside, a nucleotide, or an oligonucleotide, or a linking group attaching to a solid support; and Z is -O-, -S-, -NR-, or -CR2-. [00264] In some embodiments, the STAT3 targeting oligonucleotide comprises one or more nucleic acids conjugated with targeting ligands represented by formula II-a:
Figure imgf000084_0001
or a pharmaceutically acceptable salt thereof. [00265] In some embodiments, the STAT3 targeting oligonucleotide comprises one or more nucleic acids conjugated with targeting ligands represented by formula II-b or II-c:
Figure imgf000084_0002
II-c or a pharmaceutically acceptable salt thereof, wherein: L1 is a covalent bond, a monovalent or a bivalent saturated or unsaturated, straight or branched C1-50 hydrocarbon chain, wherein 0-10 methylene units of the hydrocarbon chain are independently replaced by -Cy-, -O-, -C(O)NR-, -NR-, -S-, -C(O)-, -C(O)O-, -S(O)-, -S(O)2-, -P(O)OR-, -P(S)OR-, or
Figure imgf000084_0003
R4 is hydrogen, RA, or a suitable amine protection group; and R5 is adamantyl, or a saturated or unsaturated, straight, or branched C1-50 hydrocarbon chain, wherein 0-10 methylene units of the hydrocarbon chain are independently replaced by -O-, - C(O)NR-, -NR-, -S-, -C(O)-, -C(O)O-, -S(O)-, -S(O)2-, -P(O)OR-, or -P(S)OR. [00266] In some embodiments, R5 is selected from
Figure imgf000085_0001
[00267] In some embodiments, R5 is selected from:
Figure imgf000086_0001
[00268] In some embodiments, R5 is
Figure imgf000086_0002
. In some
Figure imgf000086_0004
. ,
Figure imgf000086_0005
embodiments,
Figure imgf000086_0003
. In some embodiments, R5 is
Figure imgf000087_0001
some embodiments, R5 is
Figure imgf000087_0002
some embodiments, R5 is
Figure imgf000087_0004
. In some embodiments, R5 is
Figure imgf000087_0003
. [00269] In some embodiments, the STAT3 targeting oligonucleotide comprises one or more nucleic acids conjugated with targeting ligands represented by formula II-Ib or II-Ic:
Figure imgf000088_0001
II-Ic or a pharmaceutically acceptable salt thereof; wherein B is a nucleobase or hydrogen; m is 1-50; X1 is -O-, or -S-; Y is hydrogen,
Figure imgf000088_0002
R3 is hydrogen, or a suitable protecting group; X2 is O, or S; X3 is -O-, -S-, or a covalent bond; Y1 is a linking group attaching to the 2′- or 3′-terminal of a nucleoside, a nucleotide, or an oligonucleotide; Y2 is hydrogen, a phosphoramidite analogue, an internucleotide linking group attaching to the 5′-terminal of a nucleoside, a nucleotide, or an oligonucleotide, or a linking group attaching to a solid support; R5 is adamantyl, or a saturated or unsaturated, straight, or branched C1-50 hydrocarbon chain, wherein 0-10 methylene units of the hydrocarbon chain are independently replaced by -O-, - C(O)NR-, -NR-, -S-, -C(O)-, -C(O)O-, -S(O)-, -S(O)2-, -P(O)OR-, or -P(S)OR-; and R is hydrogen, a suitable protecting group, or an optionally substituted group selected from C1-6 aliphatic, phenyl, a 4-7 membered saturated or partially unsaturated heterocyclic having 1-2 heteroatoms independently selected from nitrogen, oxygen, and sulfur, and a 5-6 membered heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur. [00270] In some embodiments, R5 is selected from
Figure imgf000089_0001
. [00271] In some embodiments, R5 is
Figure imgf000090_0001
. [00273] In some embodiments, the nucleotide sequence of the STAT3 targeting oligonucleotide comprises 1-10 targeting ligands. In some embodiments, the nucleotide sequence comprises 1, 2 or 3 targeting ligands. [00274] In some embodiments, the STAT3 targeting oligonucleotide is a double- stranded molecule. In some embodiments, the STAT3 targeting oligonucleotide is an RNAi molecule. In some embodiments, the STAT3 targeting double stranded oligonucleotide comprises a stem loop. In some embodiments, the ligand is conjugated to any of the nucleotides in the stem loop. In some embodiments, the ligand is conjugated to the first nucleotide from 5’ to 3’, in the stem loop. In some embodiments, the ligand is conjugated to the second nucleotide from 5’ to 3’ in the stem loop. In some embodiments, the ligand is conjugated to the third nucleotide from 5’ to 3’ in the stem loop. In some embodiments, the ligand is conjugated to the fourth nucleotide from 5’ to 3’ in the stem loop. In some embodiments, the ligand is conjugated to one, two, three, or four of the nucleotides in the stem loop. In some embodiments, the ligand is conjugated to three of the nucleotides in the stem loop. [00275] In some embodiments, the STAT3 targeting double stranded oligonucleotide comprises a stem loop, wherein one or more lipids are conjugated to one or more nucleotides of the stem loop. In some embodiments, the STAT3 targeting double stranded oligonucleotide comprises a stem loop, wherein one or more C16 lipids are conjugated to one or more nucleotides of the stem loop. In some embodiments, the STAT3 targeting double stranded oligonucleotide comprises a stem loop, wherein one or more C18 lipids are conjugated to one or more nucleotides of the stem loop. [00276] In some embodiments, the STAT3 targeting oligonucleotide comprises a sense strand of 36 nucleotides with positions numbered 1-36 from 5’ to 3’. In some embodiments, the STAT3 targeting oligonucleotide comprises a lipid conjugated to position 27 of a 36- nucleotide sense strand. In some embodiments, STAT3 targeting oligonucleotide comprises a lipid conjugated to position 28 of a 36-nucleotide sense strand. In some embodiments, the STAT3 targeting oligonucleotide comprises a lipid conjugated to position 29 of a 36- nucleotide sense strand. In some embodiments, the STAT3 targeting oligonucleotide comprises a lipid conjugated to position 30 of a 36-nucleotide sense strand. In some embodiments, a 36-nucleotide sense strand forms a stem loop having a loop with positions 27-30. In some embodiments, a lipid is conjugated to more than one position of the loop (e.g., positions 27 and 28 of a 36-nucleotide sense strand). [00277] In some embodiments, the STAT3 targeting oligonucleotide comprises a C16 lipid conjugated to position 27 of a 36-nucleotide sense strand. In some embodiments, STAT3 targeting oligonucleotide comprises a C16 lipid conjugated to position 28 of a 36- nucleotide sense strand. In some embodiments, the STAT3 targeting oligonucleotide comprises a C16 lipid conjugated to position 29 of a 36-nucleotide sense strand. In some embodiments, the STAT3 targeting oligonucleotide comprises a C16 lipid conjugated to position 30 of a 36-nucleotide sense strand. In some embodiments, a 36-nucleotide sense strand forms a stem loop having a loop with positions 27-30. In some embodiments, a C16 lipid is conjugated to more than one position of the loop (e.g., positions 27 and 28 of a 36- nucleotide sense strand). [00278] In some embodiments, the STAT3 targeting oligonucleotide comprises a C18 lipid conjugated to position 27 of a 36-nucleotide sense strand. In some embodiments, STAT3 targeting oligonucleotide comprises a C18 lipid conjugated to position 28 of a 36- nucleotide sense strand. In some embodiments, the STAT3 targeting oligonucleotide comprises a C18 lipid conjugated to position 29 of a 36-nucleotide sense strand. In some embodiments, the STAT3 targeting oligonucleotide comprises a C18 lipid conjugated to position 30 of a 36-nucleotide sense strand. In some embodiments, a 36-nucleotide sense strand forms a stem loop having a loop with positions 27-30. In some embodiments, a C18 lipid is conjugated to more than one position of the loop (e.g., positions 27 and 28 of a 36- nucleotide sense strand). [00279] In some embodiments, a STAT3 targeting oligonucleotide comprises an antisense strand of 15 to 30 nucleotides and a sense strand of 15 to 40 nucleotide, wherein the sense and antisense strands form a duplex region, wherein the antisense strand comprises a region of complementarity to a STAT3 mRNA target sequence expressed in an immune cell associated with a tumor microenvironment, wherein the sense strand comprises at its 3’ end a stem-loop comprising a tetraloop comprising 4 nucleosides, wherein one or more of the 4 nucleosides is represented by formula II-Ib:
Figure imgf000092_0001
, wherein B is selected from an adenine and a guanine nucleobase, and wherein R5 is a hydrocarbon chain. In some embodiments, m is 1, X1 is O, Y2 is an internucleotide linking group attaching to the 5’ terminal of a nucleoside, Y1 Y is represented by
Figure imgf000092_0002
, Y1 is a linking group attaching to the 2’ or 3’ terminal of a nucleotide, X2 is O, X3 is O, and R3 is H. [00280] In some embodiments, the hydrocarbon chain is a C8-C30 hydrocarbon chain. In some embodiments, the hydrocarbon chain is a C16 hydrocarbon chain. In some embodiments, the C16 hydrocarbon chain is represented by
Figure imgf000092_0003
. In some embodiments, the hydrocarbon chain is a C18 hydrocarbon chain. In some embodiments, the C18 hydrocarbon chain is represented by
Figure imgf000092_0004
. [00281] In some embodiments, the oligonucleotide comprises a sense strand comprising a sequence selected from SEQ ID NOs: 89-280, wherein the sense strand comprises a C18 lipid. In some embodiments, the 4 nucleosides of the tetraloop are numbered 1-4 from 5’ to 3’ and position 1 is represented by formula II-Ib. In some embodiments, position 2 is represented by formula II-Ib. In some embodiments, position 3 is represented by formula II-Ib. In some embodiments, position 4 is represented by formula II- Ib. In some embodiments, the sense strand is 36 nucleotides with positions numbered 1-36 from 5’ to 3’, wherein the stem-loop comprises nucleotides at positions 21-36, and wherein one or more nucleosides at positions 27-30 are represented by formula II-Ib. In some embodiments, the antisense strand is 22 nucleotides. Exemplary STAT3 Targeting Oligonucleotides [00282] In some embodiments, an oligonucleotide targeting STAT3 comprises a sense strand and an antisense strand as set forth in Tables 1, 2, 3, 8, 9, 10, 11, and 12, wherein the oligonucleotide comprises a stem loop structure having a double-stranded stem of about 2-6 base pairs and a loop of 3-4 nucleotides, and wherein the sense and antisense strands comprise the modification pattern set forth in FIG.1A or Example 7. In some embodiments, an oligonucleotide targeting STAT3 comprises a sense strand and an antisense strand as set forth in Tables 1, 2, 3, 8, 9, 10, 11, and 12, wherein the oligonucleotide comprises a stem loop structure having a double-stranded stem of about 2-6 base pairs and a loop of 3-4 nucleotides, wherein the sense and antisense strands comprise the modification pattern set forth in FIG.1A, and wherein antisense strand is modified with an oxymethylphosphonate at the 4’ carbon of the 5’ terminal nucleotide. In some embodiments, the oligonucleotide comprises a stem loop comprising the nucleotide sequence of SEQ ID NO: 86. In some embodiments, the oligonucleotide comprises a double-stranded stem of 6 base pairs and a stem loop of 4 nucleotides comprising one, two, three or four GalNAc conjugated nucleotides. In some embodiments, the GalNAc conjugated nucleotide is a monovalent GalNAc conjugated to an adenine nucleotide, referred to as [ademA-GalNAc] or 2′- aminodiethoxymethanol-Adenine-GalNAc, as depicted below:
Figure imgf000093_0001
[00283] In some embodiments, the stem loop comprises a double-stranded stem of 6 base pairs and a loop comprising the nucleotide sequence GAAA, wherein each adenine nucleotide is ademA-GalNAc. [00284] In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense and antisense strand comprising nucleotide sequences selected from: (a) SEQ ID NOs: 9 and 10, respectively; (b) SEQ ID NOs: 37 and 38, respectively; (c) SEQ ID NOs: 65 and 66, respectively; and (d) SEQ ID NOs: 69 and 70, respectively. [00285] In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense strand and an antisense strand comprising nucleotide sequences selected from: (a) SEQ ID NOs: 9 and 10, respectively; (b) SEQ ID NOs: 37 and 38, respectively; (c) SEQ ID NOs: 65 and 66, respectively; and (d) SEQ ID NOs: 69 and 70, respectively, wherein the sense and antisense strands are modified based on the pattern below Sense Strand: [mXs][mX][mX][mX][mX][mX][mX][fX][fX][fX][fX][mX][mX][mX][mX][mX][mX][ mX][mX][mX][mX][mX][mX][mX][mX][mX][mX][ademX-C#] [mX] [mX] [mX] [mX] [mX] [mX][mX][mX] Hybridized to Antisense Strand: [MePhosphonate-4O-mXs][fXs][fX][fX][fX][mX][fX][mX][mX] [fX][mX][mX][mX][fX][mX][mX][mX][mX][mX][mXs][mXs][mX] (key provided in Table 7). In some embodiments, C# is C16 or C18. [00286] In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense strand and an antisense strand comprising nucleotide sequences selected from: (a) SEQ ID NOs: 862 and 952, respectively; (b) SEQ ID NOs: 875 and 965, respectively; (c) SEQ ID NOs: 876 and 966, respectively; and (d) SEQ ID NOs: 920 and 1010, respectively, wherein the sense and antisense strands are modified based on the pattern below Sense Strand: [mXs][mX][mX][mX][mX][mX][mX][fX][fX][fX][fX][mX][mX][mX][mX][mX][mX][ mX][mX][mX][mX][mX][mX][mX][mX][mX][mX][ademX-C#] [mX] [mX] [mX] [mX] [mX] [mX][mX][mX] Hybridized to Antisense Strand: [MePhosphonate-4O-mXs][fXs][fX][fX][fX][mX][fX][mX][mX] [fX][mX][mX][mX][fX][mX][mX][mX][mX][mX][mXs][mXs][mX] (key provided in Table 7). In some embodiments, C# is C16 or C18. [00287] In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense strand and an antisense strand comprising nucleotide sequences selected from: (a) SEQ ID NOs: 862 and 952, respectively; (b) SEQ ID NOs: 875 and 965, respectively; (c) SEQ ID NOs: 876 and 966, respectively; and (d) SEQ ID NOs: 920 and 1010, respectively, wherein the sense and antisense strands are modified based on the pattern below Sense Strand: [mXs][mX][mX][mX][mX][mX][mX][fX][fX][fX][fX][mX][mX][mX][mX][mX][mX][ mX][mX][mX][mX][mX][mX][mX][mX][mX][mX][ademX-C#] [mX] [mX] [mX] [mX] [mX] [mX][mX][mX] Hybridized to Antisense Strand: [MePhosphonate-4O-mXs][fXs][fXs][fX][fX][mX][fX][mX][mX] [fX][mX][mX][mX][fX][mX][mX][mX][mX][mX][mXs][mXs][mX] (key provided in Table 7). In some embodiments, C# is C16 or C18. [00288] In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense and antisense strand comprising nucleotide sequences selected from: (a) SEQ ID NOs: 11 and 12, respectively; (b) SEQ ID NOs: 39 and 40, respectively; (c) SEQ ID NOs: 67 and 68, respectively; and (d) SEQ ID NOs: 71 and 72, respectively. [00289] In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises the sequence set forth in SEQ ID NO: 81. In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises the sequence set forth in SEQ ID NO: 82. In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises the sequence set forth in SEQ ID NO: 83. In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises the sequence set forth in SEQ ID NO: 84. [00290] In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense and antisense strand having nucleotide sequences set forth in SEQ ID NOs: 87 and 68, respectively. In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense and antisense strand having nucleotide sequences set forth in SEQ ID NOs: 88 and 71, respectively. [00291] In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense strand sequence selected from SEQ ID NOs: 89-280. In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense strand sequence selected from SEQ ID NOs: 857-946. In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense strand sequence selected from SEQ ID NOs: 857-888. In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense strand sequence selected from SEQ ID NOs: 889-912. In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense strand sequence selected from SEQ ID NOs: 913-934. In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense strand sequence selected from SEQ ID NOs: 935-946. [00292] In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises an antisense strand sequence selected from SEQ ID NOs: 947-1036. In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises an antisense strand sequence selected from SEQ ID NOs: 947-978. In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises an antisense strand sequence selected from SEQ ID NOs: 979-1002. In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises an antisense strand sequence selected from SEQ ID NOs: 1003-1024. In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises an antisense strand sequence selected from SEQ ID NOs: 1025-1036. [00293] In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense strand sequence selected from SEQ ID NOs: 857-946 and an antisense strand selected from SEQ ID NOs: 947-1036. In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense strand sequence selected from SEQ ID NOs: 857-888 and an antisense strand selected from SEQ ID NOs: 947-978. In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense strand sequence selected from SEQ ID NOs: 889-912 and an antisense strand selected from SEQ ID NOs: 979-1002. In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense strand sequence selected from SEQ ID NOs: 913-934 and an antisense strand selected from SEQ ID NOs:1003-1024. In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense strand sequence selected from SEQ ID NOs: 935-946 and an antisense strand selected from SEQ ID NOs:1025-1036. [00294] In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense strand sequence selected from SEQ ID NOs: 1037-1126. In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense strand sequence selected from SEQ ID NOs: 1037-1068. In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense strand sequence selected from SEQ ID NOs:1069-1092. In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense strand sequence selected from SEQ ID NOs: 1093-1114. In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense strand sequence selected from SEQ ID NOs:1115-1126. [00295] In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises an antisense strand sequence selected from SEQ ID NOs: 1127-1216. In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises an antisense strand sequence selected from SEQ ID NOs: 1127-1158. In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises an antisense strand sequence selected from SEQ ID NOs: 1159-1182. In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises an antisense strand sequence selected from SEQ ID NOs:1183-1204. In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises an antisense strand sequence selected from SEQ ID NOs:1205-1216. [00296] In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense strand sequence selected from SEQ ID NOs: 1037-1126 and an antisense strand selected from SEQ ID NOs: 1127-1216. In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense strand sequence selected from SEQ ID NOs: 1037-1068 and an antisense strand selected from SEQ ID NOs: 1127-1182. In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense strand sequence selected from SEQ ID NOs: 1069-1092 and an antisense strand selected from SEQ ID NOs: 1159-1182. In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense strand sequence selected from SEQ ID NOs: 1093-1114 and an antisense strand selected from SEQ ID NOs:1183-1204. In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense strand sequence selected from SEQ ID NOs: 1115-1126 and an antisense strand selected from SEQ ID NOs:1205-1216. [00297] In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense and antisense strand comprising nucleotide sequences selected from: (a) SEQ ID NOs: 857 and 947, respectively; (b) SEQ ID NOs: 858 and 948, respectively; (c) SEQ ID NOs: 859 and 949, respectively; (d) SEQ ID NOs: 860 and 950, respectively; (e) SEQ ID NOs: 862 and 952, respectively; (f) SEQ ID NOs: 867 and 957, respectively; (g) SEQ ID NOs: 875 and 965, respectively; and (h) SEQ ID NOs: 876 and 966, respectively. [00298] In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense and antisense strand comprising nucleotide sequences selected from: (a) SEQ ID NOs: 901 and 991, respectively; (b) SEQ ID NOs: 910 and 1000, respectively; (c) SEQ ID NOs: 899 and 989, respectively; (d) SEQ ID NOs: 896 and 986, respectively; (e) SEQ ID NOs: 892 and 982, respectively; (f) SEQ ID NOs: 890 and 980, respectively; and (g) SEQ ID NOs: 889 and 979, respectively. [00299] In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense and antisense strand comprising nucleotide sequences selected from: (a) SEQ ID NOs: 940 and 1030, respectively; (b) SEQ ID NOs: 937 and 1027, respectively; and (c) SEQ ID NOs: 939 and 1029, respectively. [00300] In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense and antisense strand comprising nucleotide sequences selected from: (a) SEQ ID NOs: 915 and 1005, respectively; (b) SEQ ID NOs: 924 and 1014, respectively; (c) SEQ ID NOs: 913 and 1003, respectively; and (d) SEQ ID NOs: 920 and 1010, respectively. [00301] In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense and antisense strand comprising nucleotide sequences selected from: (a) SEQ ID NOs: 862 and 952, respectively; (b) SEQ ID NOs: 875 and 965, respectively; (c) SEQ ID NOs: 876 and 966, respectively; and (d) SEQ ID NOs: 920 and 1010, respectively. [00302] In some embodiments, the sense strand comprises the sequence of SEQ ID NO: 862 and the antisense strand comprises the sequence of SEQ ID NO: 952. [00303] In some embodiments, the sense strand comprises the sequence of SEQ ID NO: 875 and the antisense strand comprises the sequence of SEQ ID NO: 965. [00304] In some embodiments, the sense strand comprises the sequence of SEQ ID NO: 876 and the antisense strand comprises the sequence of SEQ ID NO: 966. [00305] In some embodiments, the sense strand comprises the sequence of SEQ ID NO: 920 and the antisense strand comprises the sequence of SEQ ID NO: 1010. [00306] In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense and antisense strand comprising nucleotide sequences selected from: (a) SEQ ID NOs: 1037 and 1127, respectively; (b) SEQ ID NOs: 1038 and 1128, respectively; (c) SEQ ID NOs: 1039 and 1129, respectively; (d) SEQ ID NOs: 1040 and 1130, respectively; (e) SEQ ID NOs: 1042 and 1132, respectively; (f) SEQ ID NOs: 1047 and 1137, respectively; (g) SEQ ID NOs: 1055 and 1145, respectively; and (h) SEQ ID NOs: 1056 and 1146, respectively. [00307] In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense and antisense strand comprising nucleotide sequences selected from: (a) SEQ ID NOs: 1081 and 1171, respectively; (b) SEQ ID NOs: 1090 and 1180, respectively; (c) SEQ ID NOs: 1079 and 1169, respectively; (d) SEQ ID NOs: 1076 and 1166, respectively; (e) SEQ ID NOs: 1072 and 1162, respectively; (f) SEQ ID NOs: 1070 and 1160, respectively; and (g) SEQ ID NOs: 1069 and 1159, respectively. [00308] In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense and antisense strand comprising nucleotide sequences selected from: (a) SEQ ID NOs: 1120 and 1210, respectively; (b) SEQ ID NOs: 1117 and 1207, respectively; and (c) SEQ ID NOs: 1119 and 1209, respectively. [00309] In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense and antisense strand comprising nucleotide sequences selected from: (a) SEQ ID NOs: 1095 and 1185, respectively; (b) SEQ ID NOs: 1104 and 1194, respectively; (c) SEQ ID NOs: 1093 and 1183, respectively; and (d) SEQ ID NOs: 1100 and 1190, respectively. [00310] In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense and antisense strand comprising nucleotide sequences selected from: (a) SEQ ID NOs: 1042 and 1132, respectively; (b) SEQ ID NOs: 1055 and 1145, respectively; (c) SEQ ID NOs: 1056 and 1146, respectively; and (d) SEQ ID NOs: 1100 and 1190, respectively. [00311] In some embodiments, the sense strand comprises the sequence of SEQ ID NO: 1042 and the antisense strand comprises the sequence of SEQ ID NO: 1132. [00312] In some embodiments, the sense strand comprises the sequence of SEQ ID NO: 1055 and the antisense strand comprises the sequence of SEQ ID NO: 1145. [00313] In some embodiments, the sense strand comprises the sequence of SEQ ID NO: 1056 and the antisense strand comprises the sequence of SEQ ID NO: 1146. [00314] In some embodiments, the sense strand comprises the sequence of SEQ ID NO: 1100 and the antisense strand comprises the sequence of SEQ ID NO: 1190. [00315] In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense and antisense strand comprising nucleotide sequences selected from: (a) SEQ ID NOs: 1042 and 4, respectively; (b) SEQ ID NOs: 1055 and 5, respectively; (c) SEQ ID NOs: 1056 and 6, respectively; and (d) SEQ ID NOs: 1100 and 7, respectively. [00316] In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA described herein comprises minimal off-target effects. For example, in some embodiments, an oligonucleotide described herein reduces STAT3 expression and does not reduce STAT1 expression or reduces STAT1 expression less than STAT3 expression. In some embodiments, the oligonucleotide comprises a sense strand comprising the nucleotide sequence set forth in SEQ ID NO: 862 and an antisense strand comprising the nucleotide sequence set forth in SEQ ID NO: 952, wherein the oligonucleotide reduces STAT3 expression and does not reduce STAT1 expression or reduces STAT1 expression less than STAT3 expression. In some embodiments, the oligonucleotide comprises a sense strand comprising the nucleotide sequence set forth in SEQ ID NO: 1042 and an antisense strand comprising the nucleotide sequence set forth in SEQ ID NO: 1132, wherein the oligonucleotide reduces STAT3 expression and does not reduce STAT1 expression or reduces STAT1 expression less than STAT3 expression. In some embodiments, the oligonucleotide comprises a sense strand comprising the nucleotide sequence set forth in SEQ ID NO: 875 and an antisense strand comprising the nucleotide sequence set forth in SEQ ID NO: 965, wherein the oligonucleotide reduces STAT3 expression and does not reduce STAT1 expression or reduces STAT1 expression less than STAT3 expression. In some embodiments, the oligonucleotide comprises a sense strand comprising the nucleotide sequence set forth in SEQ ID NO: 1055 and an antisense strand comprising the nucleotide sequence set forth in SEQ ID NO: 1145, wherein the oligonucleotide reduces STAT3 expression and does not reduce STAT1 expression or reduces STAT1 expression less than STAT3 expression. [00317] In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA described herein is a species cross-reactive oligonucleotide. In some embodiments, an oligonucleotide described herein is capable of reducing expression of STAT3 mRNA of at least two different species. In some embodiments, an oligonucleotide described herein is capable of reducing expression of STAT3 mRNA of at least two different species but does not cross-react with non-STAT3 mRNA (e.g., STAT1). In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA is cross-reactive between at least two species. In some embodiments, an oligonucleotide for reducing expression of STAT3 cross-reacts with human, non-human primate, and mouse STAT3 mRNA. In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA cross-reacts with human and mouse STAT3 mRNA. In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA cross-reacts with human and non-human primate STAT3 mRNA. [00318] In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA reduces STAT3 mRNA by at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, or at least 95%. [00319] In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA reduces STAT3 mRNA by at least 50% to at least 75% in human, non-human primate, and mouse (i.e. the oligonucleotide is a species cross-reactive oligonucleotide). In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA reduces STAT3 mRNA by at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, or at least 75% in human, non-human primate, and mouse (i.e. the oligonucleotide is a species cross-reactive oligonucleotide). In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA reduces STAT3 mRNA by at least 80%, at least 85%, at least 90%, or at least 95% in human, non-human primate, and mouse (i.e. the oligonucleotide is a species cross-reactive oligonucleotide). [00320] In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA reduces STAT3 mRNA by at least 50% to at least 75% in human and non-human primate (i.e. the oligonucleotide is a species cross-reactive oligonucleotide). In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA reduces STAT3 mRNA by at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, or at least 75% in human and non-human primate (i.e. the oligonucleotide is a species cross-reactive oligonucleotide). In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA reduces STAT3 mRNA by at least 80%, at least 85%, at least 90%, or at least 95% in human and non-human primate (i.e. the oligonucleotide is a species cross-reactive oligonucleotide). [00321] In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA reduces STAT3 mRNA by at least 50% to at least 75% in human and mouse (i.e. the oligonucleotide is a species cross-reactive oligonucleotide). In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA reduces STAT3 mRNA by at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, or at least 75% in human and mouse (i.e. the oligonucleotide is a species cross-reactive oligonucleotide). In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA reduces STAT3 mRNA by at least 80%, at least 85%, at least 90%, or at least 95% in human and mouse (i.e. the oligonucleotide is a species cross-reactive oligonucleotide). In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense and antisense strand selected from: (a) SEQ ID NOs: 901 and 991, respectively; (b) SEQ ID NOs: 910 and 1000, respectively; (c) SEQ ID NOs: 899 and 989, respectively; (d) SEQ ID NOs: 896 and 986, respectively; (e) SEQ ID NOs: 892 and 982, respectively; (f) SEQ ID NOs: 890 and 980, respectively; and (g) SEQ ID NOs: 889 and 979, respectively, wherein the oligonucleotide reduces STAT3 mRNA in humans, non-human primates, and mice (i.e. the oligonucleotide is a species cross-reactive oligonucleotide). [00322] In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense and antisense strand selected from: (a) SEQ ID NOs: 940 and 1030, respectively; (b) SEQ ID NOs: 937 and 1027, respectively; and (c) SEQ ID NOs: 939 and 1029, respectively, wherein the oligonucleotide reduces STAT3 mRNA in humans, non-human primates, and mice (i.e. the oligonucleotide is a species cross-reactive oligonucleotide). [00323] In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense and antisense strand selected from: (a) SEQ ID NOs: 915 and 1005, respectively; (b) SEQ ID NOs: 924 and 1014, respectively; (c) SEQ ID NOs: 913 and 1003, respectively; and (d) SEQ ID NOs: 920 and 1010, respectively, wherein the oligonucleotide reduces STAT3 mRNA in humans, non-human primates, and mice (i.e. the oligonucleotide is a species cross-reactive oligonucleotide). [00324] In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises the sense strand sequence of SEQ ID NO: 862 and the antisense strand sequence of SEQ ID NO: 952, wherein the oligonucleotide reduces STAT3 mRNA in humans and non-human primates (i.e. the oligonucleotide is a species cross-reactive oligonucleotide). [00325] In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises the sense strand sequence of SEQ ID NO: 875 and the antisense strand sequence of SEQ ID NO: 965, wherein the oligonucleotide reduces STAT3 mRNA in humans. [00326] In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises the sense strand sequence of SEQ ID NO: 876 and the antisense strand sequence of SEQ ID NO: 966, wherein the oligonucleotide reduces STAT3 mRNA in humans. [00327] In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises the sense strand sequence of SEQ ID NO: 920 and the antisense strand sequence of SEQ ID NO: 1010, wherein the oligonucleotide reduces STAT3 mRNA in humans. [00328] In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense and antisense strand selected from: (a) SEQ ID NOs: 857 and 947, respectively; (b) SEQ ID NOs: 858 and 948, respectively; (c) SEQ ID NOs: 859 and 949, respectively; (d) SEQ ID NOs: 860 and 950, respectively; (e) SEQ ID NOs: 862 and 952, respectively; (f) SEQ ID NOs: 867 and 957, respectively; (g) SEQ ID NOs: 875 and 965, respectively; and (h) SEQ ID NOs: 876 and 966, respectively, wherein the oligonucleotide reduces STAT3 mRNA by at least 75%. [00329] In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense and antisense strand selected from: (a) SEQ ID NOs: 901 and 991, respectively; (b) SEQ ID NOs: 910 and 1000, respectively; (c) SEQ ID NOs: 899 and 989, respectively; (d) SEQ ID NOs: 896 and 986, respectively; (e) SEQ ID NOs: 892 and 982, respectively; (f) SEQ ID NOs: 890 and 980, respectively; and (g) SEQ ID NOs: 889 and 979, respectively, wherein the oligonucleotide reduces STAT3 mRNA by at least 75%. [00330] In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense and antisense strand selected from: (a) SEQ ID NOs: 940 and 1030, respectively; (b) SEQ ID NOs: 937 and 1027, respectively; and (c) SEQ ID NOs: 939 and 1029, respectively, wherein the oligonucleotide reduces STAT3 mRNA by at least 75%. [00331] In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense and antisense strand selected from: (a) SEQ ID NOs: 915 and 1005, respectively; (b) SEQ ID NOs: 924 and 1014, respectively; (c) SEQ ID NOs: 913 and 1003, respectively; and (d) SEQ ID NOs: 920 and 1010, respectively, wherein the oligonucleotide reduces STAT3 mRNA by at least 75%. [00332] In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense and antisense strand selected from: (a) SEQ ID NOs: 862 and 952, respectively; (b) SEQ ID NOs: 875 and 965, respectively; (c) SEQ ID NOs: 876 and 966, respectively; and (d) SEQ ID NOs: 920 and 1010, respectively, wherein the oligonucleotide reduces STAT3 mRNA by at least 75%. [00333] In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises the sense strand sequence of SEQ ID NO: 862 and the antisense strand sequence of SEQ ID NO: 952, wherein the oligonucleotide reduces STAT3 mRNA by at least 75%. [00334] In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises the sense strand sequence of SEQ ID NO: 875 and the antisense strand sequence of SEQ ID NO: 965, wherein the oligonucleotide reduces STAT3 mRNA by at least [00335] In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises the sense strand sequence of SEQ ID NO: 876 and the antisense strand sequence of SEQ ID NO: 966, wherein the oligonucleotide reduces STAT3 mRNA by at least 75%. [00336] In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises the sense strand sequence of SEQ ID NO: 920 and the antisense strand sequence of SEQ ID NO: 1010, wherein the oligonucleotide reduces STAT3 mRNA by at least 75%. [00337] In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense and antisense strand selected from: (a) SEQ ID NOs: 857 and 947, respectively; (b) SEQ ID NOs: 858 and 948, respectively; (c) SEQ ID NOs: 859 and 949, respectively; (d) SEQ ID NOs: 860 and 950, respectively; (e) SEQ ID NOs: 862 and 952, respectively; (f) SEQ ID NOs: 867 and 957, respectively; (g) SEQ ID NOs: 875 and 965, respectively; and (h) SEQ ID NOs: 876 and 966, respectively, wherein the oligonucleotide is conjugated to a lipid. [00338] In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense and antisense strand selected from: (a) SEQ ID NOs: 901 and 991, respectively; (b) SEQ ID NOs: 910 and 1000, respectively; (c) SEQ ID NOs: 899 and 989, respectively; (d) SEQ ID NOs: 896 and 986, respectively; (e) SEQ ID NOs: 892 and 982, respectively; (f) SEQ ID NOs: 890 and 980, respectively; and (g) SEQ ID NOs: 889 and 979, respectively, wherein the oligonucleotide is conjugated to a lipid on the sense strand. [00339] In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense and antisense strand selected from: (a) SEQ ID NOs: 940 and 1030, respectively; (b) SEQ ID NOs: 937 and 1027, respectively; and (c) SEQ ID NOs: 939 and 1029, respectively, wherein the oligonucleotide is conjugated to a lipid on the sense strand. [00340] In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense and antisense strand selected from: (a) SEQ ID NOs: 915 and 1005, respectively; (b) SEQ ID NOs: 924 and 1014, respectively; (c) SEQ ID NOs: 913 and 1003, respectively; and (d) SEQ ID NOs: 920 and 1010, respectively, wherein the oligonucleotide is conjugated to a lipid on the sense strand. [00341] In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense and antisense strand selected from: (a) SEQ ID NOs: 862 and 952, respectively; (b) SEQ ID NOs: 875 and 965, respectively; (c) SEQ ID NOs: 876 and 966, respectively; and (d) SEQ ID NOs: 920 and 1010, respectively, wherein the oligonucleotide is conjugated to a lipid on the sense strand. [00342] In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises the sense strand sequence of SEQ ID NO: 862 and the antisense strand sequence of SEQ ID NO: 952, wherein the oligonucleotide is conjugated to a lipid on the sense strand. [00343] In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises the sense strand sequence of SEQ ID NO: 875 and the antisense strand sequence of SEQ ID NO: 965, wherein the oligonucleotide is conjugated to a lipid on the sense strand. [00344] In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises the sense strand sequence of SEQ ID NO: 876 and the antisense strand sequence of SEQ ID NO: 966, wherein the oligonucleotide is conjugated to a lipid on the sense strand. [00345] In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises the sense strand sequence of SEQ ID NO: 920 and the antisense strand sequence of SEQ ID NO: 1010, wherein the oligonucleotide is conjugated to a lipid on the sense strand. [00346] In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense and antisense strand selected from: (a) SEQ ID NOs: 857 and 947, respectively; (b) SEQ ID NOs: 858 and 948, respectively; (c) SEQ ID NOs: 859 and 949, respectively; (d) SEQ ID NOs: 860 and 950, respectively; (e) SEQ ID NOs: 862 and 952, respectively; (f) SEQ ID NOs: 867 and 957, respectively; (g) SEQ ID NOs: 875 and 965, respectively; and (h) SEQ ID NOs: 876 and 966, respectively, wherein the oligonucleotide is conjugated to a C18 lipid on the sense strand. [00347] In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense and antisense strand selected from: (a) SEQ ID NOs: 901 and 991, respectively; (b) SEQ ID NOs: 910 and 1000, respectively; (c) SEQ ID NOs: 899 and 989, respectively; (d) SEQ ID NOs: 896 and 986, respectively; (e) SEQ ID NOs: 892 and 982, respectively; (f) SEQ ID NOs: 890 and 980, respectively; and (g) SEQ ID NOs: 889 and 979, respectively, wherein the oligonucleotide is conjugated to a C18 lipid on the sense strand. [00348] In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense and antisense strand selected from: (a) SEQ ID NOs: 940 and 1030, respectively; (b) SEQ ID NOs: 937 and 1027, respectively; and (c) SEQ ID NOs: 939 and 1029, respectively, wherein the oligonucleotide is conjugated to a C18 lipid on the sense strand. [00349] In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense and antisense strand selected from: (a) SEQ ID NOs: 915 and 1005, respectively; (b) SEQ ID NOs: 924 and 1014, respectively; (c) SEQ ID NOs: 913 and 1003, respectively; and (d) SEQ ID NOs: 920 and 1010, respectively, wherein the oligonucleotide is conjugated to a C18 lipid on the sense strand. [00350] In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense and antisense strand selected from: (a) SEQ ID NOs: 862 and 952, respectively; (b) SEQ ID NOs: 875 and 965, respectively; (c) SEQ ID NOs: 876 and 966, respectively; and (d) SEQ ID NOs: 920 and 1010, respectively, wherein the oligonucleotide is conjugated to a C18 lipid on the sense strand. [00351] In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises the sense strand sequence of SEQ ID NO: 862 and the antisense strand sequence of SEQ ID NO: 952, wherein the oligonucleotide is conjugated to a C18 lipid on the sense strand. [00352] In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises the sense strand sequence of SEQ ID NO: 875 and the antisense strand sequence of SEQ ID NO: 965, wherein the oligonucleotide is conjugated to a C18 lipid on the sense strand. [00353] In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises the sense strand sequence of SEQ ID NO: 876 and the antisense strand sequence of SEQ ID NO: 966, wherein the oligonucleotide is conjugated to a C18 lipid on the sense strand. [00354] In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises the sense strand sequence of SEQ ID NO: 920 and the antisense strand sequence of SEQ ID NO: 1010, wherein the oligonucleotide is conjugated to a C18 lipid on the sense strand. [00355] In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense and antisense strand selected from: (a) SEQ ID NOs: 901 and 991, respectively; (b) SEQ ID NOs: 910 and 1000, respectively; (c) SEQ ID NOs: 899 and 989, respectively; (d) SEQ ID NOs: 896 and 986, respectively; (e) SEQ ID NOs: 892 and 982, respectively; (f) SEQ ID NOs: 890 and 980, respectively; and (g) SEQ ID NOs: 889 and 979, respectively, wherein the oligonucleotide reduces STAT3 mRNA in humans, non-human primates, and mice (i.e. the oligonucleotide is a species cross-reactive oligonucleotide) by at least 75%. [00356] In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense and antisense strand selected from: (a) SEQ ID NOs: 940 and 1030, respectively; (b) SEQ ID NOs: 937 and 1027, respectively; and (c) SEQ ID NOs: 939 and 1029, respectively, wherein the oligonucleotide reduces STAT3 mRNA in humans and mice (i.e. the oligonucleotide is a species cross-reactive oligonucleotide) by at least 75%. [00357] In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense and antisense strand selected from: (a) SEQ ID NOs: 915 and 1005, respectively; (b) SEQ ID NOs: 924 and 1014, respectively; (c) SEQ ID NOs: 913 and 1003, respectively; and (d) SEQ ID NOs: 920 and 1010, respectively, wherein the oligonucleotide reduces STAT3 mRNA in humans by at least 75%. [00358] In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises the sense strand sequence of SEQ ID NO: 862 and the antisense strand sequence of SEQ ID NO: 952, wherein the oligonucleotide reduces STAT3 mRNA in humans and non-human primates (i.e. the oligonucleotide is a species cross-reactive oligonucleotide) by at least 75%. [00359] In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises the sense strand sequence of SEQ ID NO: 875 and the antisense strand sequence of SEQ ID NO: 965, wherein the oligonucleotide reduces STAT3 mRNA in humans by at least 75%. [00360] In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises the sense strand sequence of SEQ ID NO: 876 and the antisense strand sequence of SEQ ID NO: 966, wherein the oligonucleotide reduces STAT3 mRNA in humans by at least 75%. [00361] In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises the sense strand sequence of SEQ ID NO: 920 and the antisense strand sequence of SEQ ID NO: 1010, wherein the oligonucleotide reduces STAT3 mRNA in humans by at least 75%. [00362] In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense and antisense strand selected from: (a) SEQ ID NOs: 901 and 991, respectively; (b) SEQ ID NOs: 910 and 1000, respectively; (c) SEQ ID NOs: 899 and 989, respectively; (d) SEQ ID NOs: 896 and 986, respectively; (e) SEQ ID NOs: 892 and 982, respectively; (f) SEQ ID NOs: 890 and 980, respectively; and (g) SEQ ID NOs: 889 and 979, respectively, wherein the oligonucleotide is conjugated to a lipid on the sense strand and reduces STAT3 mRNA in humans, non-human primates, and mice (i.e. the oligonucleotide is a species cross- reactive oligonucleotide). [00363] In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense and antisense strand selected from: (a) SEQ ID NOs: 940 and 1030, respectively; (b) SEQ ID NOs: 937 and 1027, respectively; and (c) SEQ ID NOs: 939 and 1029, respectively, wherein the oligonucleotide is conjugated to a lipid on the sense strand and reduces STAT3 mRNA in humans and mice (i.e. the oligonucleotide is a species cross-reactive oligonucleotide). [00364] In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense and antisense strand selected from: (a) SEQ ID NOs: 915 and 1005, respectively; (b) SEQ ID NOs: 924 and 1014, respectively; (c) SEQ ID NOs: 913 and 1003, respectively; and (d) SEQ ID NOs: 920 and 1010, respectively, wherein the oligonucleotide is conjugated to a lipid on the sense strand and reduces STAT3 mRNA in humans. [00365] In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises the sense strand sequence of SEQ ID NO: 862 and the antisense strand sequence of SEQ ID NO: 952, wherein the oligonucleotide is conjugated to a lipid on the sense strand and reduces STAT3 mRNA in humans and non-human primates (i.e. the oligonucleotide is a species cross-reactive oligonucleotide). [00366] In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises the sense strand sequence of SEQ ID NO: 875 and the antisense strand sequence of SEQ ID NO: 965, wherein the oligonucleotide is conjugated to a lipid on the sense strand and reduces STAT3 mRNA in humans. [00367] In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises the sense strand sequence of SEQ ID NO: 876 and the antisense strand sequence of SEQ ID NO: 966, wherein the oligonucleotide is conjugated to a lipid on the sense strand and reduces STAT3 mRNA in humans. [00368] In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises the sense strand sequence of SEQ ID NO: 920 and the antisense strand sequence of SEQ ID NO: 1010, wherein the oligonucleotide is conjugated to a lipid on the sense strand and reduces STAT3 mRNA in humans. [00369] In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense and antisense strand selected from: (a) SEQ ID NOs: 901 and 991, respectively; (b) SEQ ID NOs: 910 and 1000, respectively; (c) SEQ ID NOs: 899 and 989, respectively; (d) SEQ ID NOs: 896 and 986, respectively; (e) SEQ ID NOs: 892 and 982, respectively; (f) SEQ ID NOs: 890 and 980, respectively; and (g) SEQ ID NOs: 889 and 979, respectively, wherein the oligonucleotide is conjugated to a C18 lipid on the sense strand and reduces STAT3 mRNA in humans, non-human primates, and mice (i.e. the oligonucleotide is a species cross-reactive oligonucleotide). [00370] In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense and antisense strand selected from: (a) SEQ ID NOs: 940 and 1030, respectively; (b) SEQ ID NOs: 937 and 1027, respectively; and (c) SEQ ID NOs: 939 and 1029, respectively, wherein the oligonucleotide is conjugated to a C18 lipid on the sense strand and reduces STAT3 mRNA in humans and mice (i.e. the oligonucleotide is a species cross-reactive oligonucleotide). [00371] In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense and antisense strand selected from: (a) SEQ ID NOs: 915 and 1005, respectively; (b) SEQ ID NOs: 924 and 1014, respectively; (c) SEQ ID NOs: 913 and 1003, respectively; and (d) SEQ ID NOs: 920 and 1010, respectively, wherein the oligonucleotide is conjugated to a C18 lipid on the sense strand and reduces STAT3 mRNA in humans. [00372] In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises the sense strand sequence of SEQ ID NO: 862 and the antisense strand sequence of SEQ ID NO: 952, wherein the oligonucleotide is conjugated to a C18 lipid on the sense strand and reduces STAT3 mRNA in humans and non-human primates (i.e. the oligonucleotide is a species cross-reactive oligonucleotide). [00373] In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises the sense strand sequence of SEQ ID NO: 875 and the antisense strand sequence of SEQ ID NO: 965, wherein the oligonucleotide is conjugated to a C18 on the sense strand lipid and reduces STAT3 mRNA in humans. [00374] In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises the sense strand sequence of SEQ ID NO: 876 and the antisense strand sequence of SEQ ID NO: 966, wherein the oligonucleotide is conjugated to a C18 lipid on the sense strand and reduces STAT3 mRNA in humans. [00375] In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises the sense strand sequence of SEQ ID NO: 920 and the antisense strand sequence of SEQ ID NO: 1010, wherein the oligonucleotide is conjugated to a C18 lipid on the sense strand and reduces STAT3 mRNA in humans. [00376] In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense and antisense strand selected from: (a) SEQ ID NOs: 901 and 991, respectively; (b) SEQ ID NOs: 910 and 1000, respectively; (c) SEQ ID NOs: 899 and 989, respectively; (d) SEQ ID NOs: 896 and 986, respectively; (e) SEQ ID NOs: 892 and 982, respectively; (f) SEQ ID NOs: 890 and 980, respectively; and (g) SEQ ID NOs: 889 and 979, respectively, wherein the oligonucleotide is conjugated to a lipid on the sense strand and reduces STAT3 mRNA in humans, non-human primates, and mice (i.e. the oligonucleotide is a species cross- reactive oligonucleotide) by at least 75%. [00377] In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense and antisense strand selected from: (a) SEQ ID NOs: 940 and 1030, respectively; (b) SEQ ID NOs: 937 and 1027, respectively; and (c) SEQ ID NOs: 939 and 1029, respectively, wherein the oligonucleotide is conjugated to a lipid on the sense strand and reduces STAT3 mRNA in humans and mice (i.e. the oligonucleotide is a species cross-reactive oligonucleotide) by at least 75%. [00378] In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense and antisense strand selected from: (a) SEQ ID NOs: 915 and 1005, respectively; (b) SEQ ID NOs: 924 and 1014, respectively; (c) SEQ ID NOs: 913 and 1003, respectively; and (d) SEQ ID NOs: 920 and 1010, respectively, wherein the oligonucleotide is conjugated to a lipid on the sense strand and reduces STAT3 mRNA in humans by at least 75%. [00379] In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises the sense strand sequence of SEQ ID NO: 862 and the antisense strand sequence of SEQ ID NO: 952, wherein the oligonucleotide is conjugated to a lipid on the sense strand and reduces STAT3 mRNA in humans and non-human primates (i.e. the oligonucleotide is a species cross-reactive oligonucleotide) by at least 75%. [00380] In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises the sense strand sequence of SEQ ID NO: 875 and the antisense strand sequence of SEQ ID NO: 965, wherein the oligonucleotide is conjugated to a lipid on the sense strand and reduces STAT3 mRNA in humans by at least 75%. [00381] In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises the sense strand sequence of SEQ ID NO: 876 and the antisense strand sequence of SEQ ID NO: 966, wherein the oligonucleotide is conjugated to a lipid on the sense strand and reduces STAT3 mRNA in humans by at least 75%. [00382] In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises the sense strand sequence of SEQ ID NO: 920 and the antisense strand sequence of SEQ ID NO: 1010, wherein the oligonucleotide is conjugated to a lipid on the sense strand and reduces STAT3 mRNA in humans by at least 75%. [00383] In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense and antisense strand selected from: (a) SEQ ID NOs: 901 and 991, respectively; (b) SEQ ID NOs: 910 and 1000, respectively; (c) SEQ ID NOs: 899 and 989, respectively; (d) SEQ ID NOs: 896 and 986, respectively; (e) SEQ ID NOs: 892 and 982, respectively; (f) SEQ ID NOs: 890 and 980, respectively; and (g) SEQ ID NOs: 889 and 979, respectively, wherein the oligonucleotide is conjugated to a C18 lipid on the sense strand and reduces STAT3 mRNA in humans, non-human primates, and mice (i.e. the oligonucleotide is a species cross-reactive oligonucleotide) by at least 75%. [00384] In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense and antisense strand selected from: (a) SEQ ID NOs: 940 and 1030, respectively; (b) SEQ ID NOs: 937 and 1027, respectively; and (c) SEQ ID NOs: 939 and 1029, respectively, wherein the oligonucleotide is conjugated to a C18 lipid on the sense strand and reduces STAT3 mRNA in humans and mice (i.e. the oligonucleotide is a species cross-reactive oligonucleotide) by at least 75%. [00385] In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises a sense and antisense strand selected from: (a) SEQ ID NOs: 915 and 1005, respectively; (b) SEQ ID NOs: 924 and 1014, respectively; (c) SEQ ID NOs: 913 and 1003, respectively; and (d) SEQ ID NOs: 920 and 1010, respectively, wherein the oligonucleotide is conjugated to a C18 lipid on the sense strand and reduces STAT3 mRNA in humans by at least 75%. [00386] In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises the sense strand sequence of SEQ ID NO: 862 and the antisense strand sequence of SEQ ID NO: 952, wherein the oligonucleotide is conjugated to a C18 lipid on the sense strand and reduces STAT3 mRNA in humans and non-human primates (i.e. the oligonucleotide is a species cross-reactive oligonucleotide) by at least 75%. [00387] In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises the sense strand sequence of SEQ ID NO: 875 and the antisense strand sequence of SEQ ID NO: 965, wherein the oligonucleotide is conjugated to a C18 lipid on the sense strand and reduces STAT3 mRNA in humans by at least 75%. [00388] In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises the sense strand sequence of SEQ ID NO: 876 and the antisense strand sequence of SEQ ID NO: 966, wherein the oligonucleotide is conjugated to a C18 lipid on the sense strand and reduces STAT3 mRNA in humans by at least 75%. [00389] In some embodiments, an oligonucleotide for reducing expression of STAT3 mRNA comprises the sense strand sequence of SEQ ID NO: 920 and the antisense strand sequence of SEQ ID NO: 1010, wherein the oligonucleotide is conjugated to a C18 lipid on the sense strand and reduces STAT3 mRNA in humans by at least 75%. Formulations [00390] Various formulations have been developed to facilitate oligonucleotide use. For example, oligonucleotides can be delivered to a subject or a cellular environment using a formulation that minimizes degradation, facilitates delivery and/or uptake, or provides another beneficial property to the oligonucleotides in the formulation. In some embodiments, an oligonucleotide is formulated in buffer solutions such as phosphate buffered saline solutions, liposomes, micellar structures, and capsids. [00391] Formulations of oligonucleotides with cationic lipids can be used to facilitate transfection of the oligonucleotides into cells. For example, cationic lipids, such as lipofectin, cationic glycerol derivatives, and polycationic molecules (e.g., polylysine, can be used. Suitable lipids include Oligofectamine, Lipofectamine (Life Technologies), NC388 (Ribozyme Pharmaceuticals, Inc., Boulder, Colo.), or FuGene 6 (Roche) all of which can be used according to the manufacturer′s instructions. [00392] Accordingly, in some embodiments, a formulation comprises a lipid nanoparticle. In some embodiments, an excipient comprises a liposome, a lipid, a lipid complex, a microsphere, a microparticle, a nanosphere or a nanoparticle, or may be otherwise formulated for administration to the cells, tissues, organs, or body of a subject in need thereof (see, e.g., Remington: THE SCIENCE AND PRACTICE OF PHARMACY, 22nd edition, Pharmaceutical Press, 2013). [00393] In some embodiments, the formulations herein comprise an excipient. In some embodiments, an excipient confers to a composition improved stability, improved absorption, improved solubility and/or therapeutic enhancement of the active ingredient. In some embodiments, an excipient is a buffering agent (e.g., sodium citrate, sodium phosphate, a tris base, or sodium hydroxide) or a vehicle (e.g., a buffered solution, petrolatum, dimethyl sulfoxide, or mineral oil). In some embodiments, an oligonucleotide is lyophilized for extending its shelf-life and then made into a solution before use (e.g., administration to a subject). Accordingly, an excipient in a composition comprising any one of the oligonucleotides described herein may be a lyoprotectant (e.g., mannitol, lactose, polyethylene glycol or polyvinylpyrrolidone) or a collapse temperature modifier (e.g., dextran, Ficoll™ or gelatin). [00394] In some embodiments, a pharmaceutical composition is formulated to be compatible with its intended route of administration. Examples of routes of administration include parenteral (e.g., intravenous, intramuscular, intraperitoneal, intradermal, subcutaneous), oral (e.g., inhalation), transdermal (e.g., topical), transmucosal and rectal administration. [00395] Pharmaceutical compositions suitable for injectable use include sterile aqueous solutions (where water soluble) or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersion. For intravenous administration, suitable carriers include physiological saline, bacteriostatic water, Cremophor EL™ (BASF, Parsippany, N.J.) or phosphate buffered saline (PBS). The carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (e.g., glycerol, propylene glycol, and liquid polyethylene glycol, and the like), and suitable mixtures thereof. In many cases, it will be preferable to include isotonic agents, for example, sugars, polyalcohol’s such as mannitol, sorbitol, sodium chloride in the composition. Sterile injectable solutions can be prepared by incorporating the oligonucleotides in a required amount in a selected solvent with one or a combination of ingredients enumerated above, as required, followed by filtered sterilization. [00396] In some embodiments, a composition may contain at least about 0.1% of the therapeutic agent or more, although the percentage of the active ingredient(s) may be between about 1% to about 80% or more of the weight or volume of the total composition. Factors such as solubility, bioavailability, biological half-life, route of administration, product shelf life, as well as other pharmacological considerations will be contemplated by one skilled in the art of preparing such pharmaceutical formulations, and as such, a variety of dosages and treatment regimens may be desirable. [00397] Even though several embodiments are directed to liver-targeted delivery of any of the oligonucleotides herein, targeting of other tissues is also contemplated. Programmed Death Ligand 1 (PD-L1) Inhibitors [00398] In some embodiments, the disclosure provides a PD-L1 inhibitor for use in combination with an oligonucleotide described herein. In some embodiments, a PD-L1 inhibitor is a small molecule, a peptide, a protein, an antibody, or nucleic acid molecule such as an siRNA, miRNA, or an antisense RNA. In some embodiments, the PD-L1 inhibitor inhibits association of PD-L1 and PD-1. In some embodiments, the PD-L1 inhibitor is specific for PD-L1. [00399] In some embodiments, the PD-L1 inhibitor is an anti-PD-L1 antibody. In some embodiments, the PD-L1 inhibitor is specific for PD-1. In some embodiments, the PD-L1 inhibitor is an anti-PD-1 antibody. In some embodiments, the antibody is a full-length antibody. In some embodiments, the antibody is an antibody fragment. In some embodiments, the PD- L1 inhibitor is a small molecule. [00400] In some embodiments, the anti-PD-L1 antibody is atezolizumab. Atezolizumab (MPDL3280A) is a fully humanized, engineered IgG1 monoclonal antibody to PD-L1. In some embodiments, the anti-PD-L1 antibody is avelumab. Avelumab (MSB0010718C) is a fully humanized, engineered IgG1 monoclonal antibody to PD-L1. In some embodiments, the anti-PD-L1 antibody is envafolimab. In some embodiments, the anti- PD-L1 antibody is durvalumab. Duvalumab (MEDI4736) is a human monoclonal antibody to PD-L1. In some embodiments, the anti-PD-L1 antibody is TSR-042. TSR-042 refers to an engineered chimeric antibody directed against the PD-1/PD-L1 pathway. In some embodiments, the anti-PD-L1 antibody is KD-033. KD-033 refers to a bifunctional anti-PD- L1/IL-15 fusion protein, wherein the anti-PD-L1 antibody binds to the cytokine IL-15 by the sushi domain of the IL-15 receptor connected at the tail. In some embodiments, the anti-PD- L1 antibody is STI-1014. STI-1014 refers to an anti-PD-L1 antibody. In some embodiments, the anti-PD-L1 antibody is KY-1003. KY-1003 is a monoclonal antibody against PD-L1. In some embodiments, the anti-PD-L1 antibody is YW243.55.S70. Antibody YW243.55.S70 is an anti-PDL1 described in U.S. Patent No.9,920,123, the contents of which are incorporated herein in their entirety. In some embodiments, the anti-PD-L1 antibody is MDX-1106. MDX- 1106, also known as MDX-1106-04, ONO-4538 or BMS-936558, is an anti-PD 1 antibody described in U.S. Patent No.8,008,449, the contents of which are incorporated herein in their entirety. In some embodiments, the anti-PD-L1 antibody is Merck 3745. Merck 3745, also known as MK-3475 or SCH-900475, is an anti-PD1 antibody described in U.S. Patent No. 8,168,757, the contents of which are incorporated herein in their entirety. In some embodiments, the anti-PD-L1 antibody is CT-011. CT-011, also known as hBAT or hBAT-1, is an anti-PD1 antibody described in U.S. Patent No.8,747,847, the contents of which are incorporated herein in their entirety. In some embodiments, the anti-PD-L1 antibody is AMP- 224. AMP-224, also known as B7-DClg, is a PDL2-Fc fusion soluble receptor described in U.S. Pub. No.2011/0223188 and U.S. Pub. No.2013/0017199, the contents of which are incorporated herein in their entirety. [00401] In some embodiments, the anti-PD-L1 antibody is any anti-PD-L1 antibody known in the art, including, but not limited to, the anti-PD-L1 antibodies disclosed in Akinleye & Rasool “Immune checkpoint inhibitors of PD-L1 as cancer therapeutics” J. of Hematology & Oncology.12(92): 2019. In some embodiments, the anti-PD-L1 antibody is BMS-936559. BMS-936559 (MDX-1105) is a fully human IgG4 monoclonal antibody against PD-L1 and is described in U.S. Patent No.7,943,743, the contents of which are incorporated herein in their entirety. In some embodiments, the anti-PD-L1 antibody is CK- 301. In some embodiments, the anti-PD-L1 antibody is CS-1001. In some embodiments, the anti-PD-L1 antibody is SHR-1316. In some embodiments, the anti-PD-L1 antibody is BG- A333. In some embodiments, the anti-PD-L1 antibody is an antibody disclosed in International Pub. No. WO 2021/231741, the contents of which are incorporated herein in their entirety. [00402] In some embodiments, the anti-PD-1 antibody is nivolumab. Nivolumab (BMS-936558) is an IgG4 monoclonal antibody described in U.S. Patent No.8,008,449, the contents of which are incorporated herein in their entirety. In some embodiments, the anti- PD-1 antibody is pembrolizumab. Pembrolizumab is an IgG4 monoclonal antibody described in U.S. Patent No.8,354,509, the contents of which are incorporated herein in their entirety. In some embodiments, the anti-PD-1 antibody is cemiplimab. Cemiplimab (REGN2810) is an IgG4 monoclonal antibody described in U.S. Patent No.9,987,500, the contents of which are incorporated herein in their entirety. [00403] In some embodiments, the anti-PD-L1 antibody described herein binds to PD-L1 with an affinity of about 30nM to about 100nM. In some embodiments, the anti-PD-L1 antibody described herein binds to PD-L1 with an affinity of about 30nM. In some embodiments, the anti-PD-L1 antibody described herein binds to PD-L1 with an affinity of about 40nM. In some embodiments, the anti-PD-L1 antibody described herein binds to PD-L1 with an affinity of about 50nM. In some embodiments, the anti-PD-L1 antibody described herein binds to PD-L1 with an affinity of about 60nM. In some embodiments, the anti-PD-L1 antibody described herein binds to PD-L1 with an affinity of about 70nM. In some embodiments, the anti-PD-L1 antibody described herein binds to PD-L1 with an affinity of about 80nM. In some embodiments, the anti-PD-L1 antibody described herein binds to PD-L1 with an affinity of about 90nM. In some embodiments, the anti-PD-L1 antibody described herein binds to PD-L1 with an affinity of about 100nM. [00404] In some embodiments, the anti-PD-1 antibody described herein binds to PD-1 with an affinity of about 30nM to about 100nM. In some embodiments, the anti-PD-1 antibody described herein binds to PD-1 with an affinity of about 30nM. In some embodiments, the anti- PD-1 antibody described herein binds to PD-1 with an affinity of about 40nM. In some embodiments, the anti-PD-1 antibody described herein binds to PD-1 with an affinity of about 50nM. In some embodiments, the anti-PD-1 antibody described herein binds to PD-1 with an affinity of about 60nM. In some embodiments, the anti-PD-1 antibody described herein binds to PD-1 with an affinity of about 70nM. In some embodiments, the anti-PD-1 antibody described herein binds to PD-1 with an affinity of about 80nM. In some embodiments, the anti- PD-1 antibody described herein binds to PD-1 with an affinity of about 90nM. In some embodiments, the anti-PD-1 antibody described herein binds to PD-1 with an affinity of about 100nM. [00405] In some embodiments, the antibody is generated using display technologies. Display technologies used to generate antibody polypeptides include any of the display techniques (e.g. display library screening techniques). In some embodiments, synthetic antibodies are designed, selected, or optimized by screening target antigens using display technologies (e.g. phage display technologies). Phage display libraries may comprise millions to billions of phage vectors, each expressing unique antibody fragments on their viral coats. Such libraries may provide richly diverse resources that are used to select potentially hundreds of antibody fragments with diverse levels of affinity for one or more antigens of interest (McCafferty, et al., 1990. Nature.348:552-4; Edwards, B.M. et al., 2003. JMB.334: 103-18; Schofield, D. et al., 2007. Genome Biol.8, R254 and Pershad, K. et al., 2010. Protein Engineering Design and Selection.23:279-88; the contents of each of which are herein incorporated by reference in their entirety). Often, the antibody fragments present in such libraries comprise scFv antibody fragments, comprising a fusion protein of VH and VL antibody domains joined by a flexible linker. In some cases, scFvs may contain the same sequence with the exception of unique sequences encoding variable loops of the CDRs. In some cases, scFvs are expressed as fusion proteins, linked to viral coat proteins (e.g. the N- terminus of the viral pill coat protein). VL chains may be expressed separately for assembly with VH chains in the periplasm prior to complex incorporation into viral coats. Precipitated library members may be sequenced from the bound phage to obtain cDNA encoding desired scFvs. Antibody variable domains or CDRs from such sequences may be directly incorporated into antibody sequences for recombinant antibody production or mutated and utilized for further optimization through in vitro affinity maturation. [00406] In some embodiments, the sequences of the polypeptides to be encoded in the viral genomes are produced using yeast surface display technology. In some embodiments, recombinant antibodies are developed by displaying the antibody fragment of interest as a fusion to on the surface of the yeast, where the protein interacts with proteins and small molecules in a solution. scFvs with affinity toward desired receptors may be isolated from the yeast surface using magnetic separation and flow cytometry. Several cycles of yeast surface display and isolation may be done to attain scFvs with desired properties through directed evolution. [00407] Methods for determining the affinity of an antibody for its antigen are known in the art. An exemplary method for determining binding affinity employs surface plasmon resonance. Surface plasmon resonance is an optical phenomenon that allows for the analysis of realtime biospecific interactions by detection of alterations in protein concentrations within a biosensor matrix, for example using the BIAcore system (Pharmacia Biosensor AB, Uppsala, Sweden and Piscataway, N.J.). For further descriptions, see Jonsson, U., et al. (1993) Ann. Biol. Clin.51: 19-26; Jonsson, U., i (1991) Biotechniques 11 :620-627; Johnsson, B., et al. (1995) J. Mol.Recognit.8: 125-131; and Johnsson, B., et al. (1991) Anal. Biochem.198:268-277. [00408] In some embodiments, the PD-L1 inhibitor is an siRNA molecule. In some embodiments, the PD-L1 inhibitor is ALN-PDL. In some embodiments, the PD-L1 inhibitor is a PD-L1 inhibitor described in U.S. Patent No.10,889,813, U.S. Patent No.10,745,704, U.S. Pub. No.2021/0277403, U.S. Pub. No.2006/0276422A1, U.S.2021/0277403, International Pub. No. WO2019/000149, or International Pub. No. WO2019/000149, the contents of which are incorporated herein in their entirety. In some embodiments, the PD-L1 inhibitor is an siRNA molecule described in Barati, M., et al. A review of PD-1/PD-L1 siRNA delivery systems in immune T cells and cancer cells, International Immunopharmacology, 2022, vol.111. In some embodiments, the siRNA molecule for inhibition of PD-L1 is an siRNA molecule generated using methods known to those of skill in the art. [00409] In some embodiments, the PD-L1 inhibitor is an antisense oligonucleotide. In some embodiments, the PD-L1 inhibitor is an antisense oligonucleotide described in U.S. Patent No.10,745,480, U.S. Patent. No.10,982,215, U.S. Pub. No.2021/0269797A1, or U.S. Pub. No. US20220220485A1, the contents of which are incorporated herein in their entirety. In some embodiments, the antisense oligonucleotide for inhibition of PD-L1 is an antisense oligonucleotide generated using methods known to those of skill in the art. [00410] In some embodiments, the PD-L1 inhibitor is a small molecule. In some embodiments, the small molecule is CA-170. CA-170 is a small molecule antagonist of PD- L1 and VISTA. In some embodiments, the small molecule is CA-327. CA-327 refers to small molecule antagonists of PD-L1 and TIM3. In some embodiments, the small molecule is BMS-1001. In some embodiments, the small molecule is BMS-1166. In some embodiments, the small molecule is BMS-8. In some embodiments, the small molecule is BMS-37. In some embodiments, the small molecule is BMS-202. In some embodiments, the small molecule is BMS-200. In some embodiments, the small molecule is Incyte-001. In some embodiments, the small molecule is Incyte-011. In some embodiments, the small molecule is INCB086550. In some embodiments, the small molecule is LH1306. In some embodiments, the small molecule is LH1307. In some embodiments, the small molecule is ARB-272572. In some embodiments, the PD-L1 inhibitor is a small molecule described in Sasikumar, P. et al. Small Molecule Agents Targeting PD-1 Checkpoint Pathway for Cancer Immunotherapy: Mechanisms of Action and Other Considerations for Their Advanced Development, Front. Immunol.2022, 13:752065. In some embodiments, the PD-L1 inhibitor is a small molecule described in U.S. Patent No.11,130,740, U.S. Patent No.10,590,105, International Pub. No. WO2019/076343, U.S. Patent No.11,555,029, or U.S. Pub. No.2018/0305315, the contents of which are incorporated herein in their entirety. In some embodiments, the small molecule is a small molecule described in Wu, Q. et al. Small molecule inhibitors targeting the PD- 1/PD-L1 signaling pathway, Acta Pharmacologica Sinica.2021, 42: 1-9. In some embodiments, the small molecule is a small molecule described in Wang, Y. et al. A Small Molecule Antagonist of PD-1/PD-L1 Interactions Acts as an Immune Checkpoint Inhibitor for NSCLC and Melanoma Immunotherapy, Front. Immuno.2021. Vol.12. In some embodiments, the small molecule inhibitor is a stereoisomer, a tautomer, a pharmaceutically acceptable salt, a hydrate, or a solvent of a small molecule described herein. In some embodiments, the small molecule for inhibition of PD-L1 is a small molecule generated using methods known to those of skill in the art. [00411] In some embodiments, the PD-L1 inhibitor is a peptide. In some embodiments, the PD-L1 inhibitor is a peptide inhibitor described in U.S. Patent No.9,422,339 or U.S. Patent No.9,850,283, the contents of which are incorporated herein in their entirety. In some embodiments, the PD-L1 inhibitor is a peptide inhibitor described in Yin, H. et al. Rational Design of Potent Peptide Inhibitors of the PD-1:PD-L1 Interaction for Cancer Immunotherapy, J. Am. Chem. Soc.2021, 143: 44, 18536-18547. In some embodiments, the PD-L1 inhibitor is a peptide inhibitor described in Lin, X., et al. Progress in PD-1/PD-L1 pathway inhibitors: From biomacromolecules to small molecules, Euro. J. Med. Chem.2020, vol.186. In some embodiments, the inhibitor peptide for inhibition of PD-L1 is a peptide generated using methods known to those of skill in the art. [00412] The person skilled in the art knows how to determine whether a compound is a PD1 and/or PDL1 inhibitor by testing it in an appropriate assay. Binding of inhibitors to PD1 and/or PDL1 and/or PDL2 can e.g., be measured in ELISA-type assays that are well known in the art. Bioassays to measure the biological effect of PD1 and/or PDL1 and/or PDL2 inhibition are well known by the person skilled in the art. Kits [00413] In some embodiments, the disclosure provides a kit comprising a STAT3 oligonucleotide herein, and instructions for administering the STAT3 oligonucleotide to a subject that has received or is receiving a PD-L1 inhibitor. In some embodiments, the kit comprises, in a suitable container, an oligonucleotide herein, one or more controls, and various buffers, reagents, enzymes and other standard ingredients well known in the art. In some embodiments, the container comprises at least one vial, well, test tube, flask, bottle, syringe, or other container means, into which the oligonucleotide is placed, and in some instances, suitably aliquoted. In some embodiments where an additional component is provided, the kit contains additional containers into which this component is placed. The kits can also include a means for containing the oligonucleotide and any other reagent in close confinement for commercial sale. Such containers may include injection or blow-molded plastic containers into which the desired vials are retained. Containers and/or kits can include labeling with instructions for use and/or warnings. [00414] In some embodiments, a kit comprises a STAT3 oligonucleotide herein, and a pharmaceutically acceptable carrier, or a pharmaceutical composition comprising the oligonucleotide and instructions for treating or delaying progression of a disease, disorder or condition associated with STAT3 expression in a subject in need thereof, wherein the subject has received or is receiving a PD-L1 inhibitor. In some embodiments, a kit comprises a STAT3 oligonucleotide herein, and a pharmaceutically acceptable carrier, or a pharmaceutical composition comprising the oligonucleotide and instructions for treating or delaying progression of a cancer in a subject in need thereof, wherein the subject has received or is receiving a PD-L1 inhibitor. [00415] In some embodiments, a kit comprises a PD-L1 inhibitor, and a pharmaceutically acceptable carrier, or a pharmaceutical composition comprising the oligonucleotide and instructions for treating or delaying progression of a disease, disorder or condition in a subject in need thereof, wherein the subject has received or is receiving a STAT3 oligonucleotide described herein. In some embodiments, a kit comprises a PD-L1 inhibitor, and a pharmaceutically acceptable carrier, or a pharmaceutical composition comprising the oligonucleotide and instructions for treating or delaying progression of a cancer in a subject in need thereof, wherein the subject has received or is receiving a STAT3 oligonucleotide described herein. EXAMPLES [00416] While the disclosure has been described with reference to the specific embodiments set forth in the following Examples, it should be understood by those skilled in the art that various changes may be made, and equivalents may be substituted without departing from the true spirit and scope of the disclosure. Further, the following Examples are offered by way of illustration and are not intended to limit the scope of the disclosure in any manner. In addition, modifications may be made to adapt to a situation, material, composition of matter, process, process step or steps, to the objective, spirit, and scope of the disclosure. All such modifications are intended to be within the scope of the disclosure. Standard techniques well known in the art or the techniques specifically described below were utilized. Abbreviations Ac: acetyl AcOH: acetic acid ACN: acetonitrile Ad: adamantyl AIBN: 2,2'-azo bisisobutyronitrile Anhyd: anhydrous Aq: aqueous B2Pin2: bis (pinacolato)diboron -4,4,4',4',5,5,5',5'-octamethyl-2,2'-bi(1,3,2- dioxaborolane) BINAP: 2,2'-bis(diphenylphosphino)-1,1'-binaphthyl BH3: Borane Bn: benzyl Boc: tert-butoxycarbonyl Boc2O: di-tert-butyl dicarbonate BPO: benzoyl peroxide BuOH: n-butanol CDI: carbonyldiimidazole COD: cyclooctadiene d: days DABCO: 1,4-diazobicyclo[2.2.2]octane DAST: diethylaminosulfur trifluoride dba: dibenzylideneacetone DBU: 1,8-diazobicyclo[5.4.0]undec-7-ene DCE: 1,2-dichloroethane DCM: dichloromethane DEA: diethylamine DHP: dihydropyran DIBAL-H: diisobutylaluminum hydride DIPA: diisopropylamine DIPEA or DIEA: N,N-diisopropylethylamine DMA: N,N-dimethylacetamide DME: 1,2-dimethoxyethane DMAP: 4-dimethylaminopyridine DMF: N,N-dimethylformamide DMP: Dess-Martin periodinane DMSO-dimethyl sulfoxide DMTr: 4,4’-dimethyoxytrityl DPPA: diphenylphosphoryl azide dppf: 1,1’-bis(diphenylphosphino)ferrocene EDC or EDCI: 1-(3-dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride ee: enantiomeric excess ESI: electrospray ionization EA: ethyl acetate EtOAc: ethyl acetate EtOH: ethanol FA: formic acid h or hrs: hours HATU: N,N,N’,N’-tetramethyl-O-(7-azabenzotriazol-1-yl)uronium hexafluorophosphate HCl: hydrochloric acid HPLC: high performance liquid chromatography HOAc: acetic acid IBX: 2-iodoxybenzoic acid IPA: isopropyl alcohol KHMDS: potassium hexamethyldisilazide K2CO3: potassium carbonate LAH: lithium aluminum hydride LDA: lithium diisopropylamide L-DBTA: dibenzoyl-L-tartaric acid m-CPBA: meta-chloroperbenzoic acid M: molar MeCN: acetonitrile MeOH: methanol Me2S: dimethyl sulfide MeONa: sodium methylate MeI: iodomethane min: minutes mL: milliliters mM: millimolar mmol: millimoles MPa: mega pascal MOMCl: methyl chloromethyl ether MsCl: methanesulfonyl chloride MTBE: methyl tert-butyl ether nBuLi: n-butyllithium NaNO2: sodium nitrite NaOH: sodium hydroxide Na2SO4: sodium sulfate NBS: N-bromosuccinimide NCS: N-chlorosuccinimide NFSI: N-Fluorobenzenesulfonimide NMO: N-methylmorpholine N-oxide NMP: N-methylpyrrolidine NMR: Nuclear Magnetic Resonance oC: degrees Celsius Pd/C: Palladium on Carbon Pd(OAc)2: Palladium Acetate PBS: phosphate buffered saline PE: petroleum ether POCl3: phosphorus oxychloride PPh3: triphenylphosphine PyBOP: (Benzotriazol-1-yloxy)tripyrrolidinophosphonium hexafluorophosphate Rel: relative R.T. or rt: room temperature s or sec: second sat: saturated SEMCl: chloromethyl-2-trimethylsilylethyl ether SFC: supercritical fluid chromatography SOCl2: sulfur dichloride tBuOK: potassium tert-butoxide TBAB: tetrabutylammonium bromide TBAF: tetrabutylammmonium fluoride TBAI: tetrabutylammonium iodide TEA: triethylamine Tf: trifluoromethanesulfonate TfAA, TFMSA or Tf2O: trifluoromethanesulfonic anhydride TFA: trifluoroacetic acid TIBSCl: 2,4,6-triisopropylbenzenesulfonyl chloride TIPS: triisopropylsilyl THF: tetrahydrofuran THP: tetrahydropyran TLC: thin layer chromatography TMEDA: tetramethylethylenediamine pTSA: para-toluenesulfonic acid UPLC: Ultra Performance Liquid Chromatography wt: weight Xantphos: 4,5-bis(diphenylphosphino)-9,9-dimethylxanthene Example 1: Preparation of Double-Stranded RNAi Oligonucleotides General Synthetic Methods [00417] The following examples are intended to illustrate the disclosure and are not to be construed as being limitations thereon. Temperatures are given in degrees centigrade (C). If not mentioned otherwise, all evaporations are performed under reduced pressure, preferably between about 15 mm Hg and 100 mm Hg (= 20-133 mbar). The structure of final products, intermediates and starting materials was confirmed by standard analytical methods, e.g., microanalysis and spectroscopic characteristics, e.g., MS, IR, NMR. Abbreviations used are those conventional in the art. [00418] All starting materials, building blocks, reagents, acids, bases, dehydrating agents, solvents, and catalysts utilized to synthesis the nucleic acid or analogues thereof of the present disclosure are either commercially available or can be produced by organic synthesis methods known to one of ordinary skill in the art (METHODS OF ORGANIC SYNTHESIS, Thieme, Volume 21 (Houben-Weyl 4th Ed.1952)). Further, the nucleic acid or analogues thereof of the present disclosure can be produced by organic synthesis methods known to one of ordinary skill in the art as shown in the following examples. [00419] All reactions are carried out under nitrogen or argon unless otherwise stated. [00420] Proton NMR (1H NMR) was conducted in deuterated solvent. In certain nucleic acid or analogues thereof disclosed herein, one or more 1H shifts overlap with residual proteo solvent signals; these signals have not been reported in the experimental provided hereinafter. [00421] As depicted in the Examples below, in certain exemplary embodiments, the nucleic acid or analogues thereof were prepared according to the following general procedures. It will be appreciated that, although the general methods depict the synthesis of certain nucleic acid or analogues thereof of the present disclosure, the following general methods, and other methods known to one of ordinary skill in the art, can be applied to all nucleic acid or analogues thereof and subclasses and species of each of these nucleic acid or analogues thereof, as described herein. Example^1a:^Synthesis^of^2‐(2‐((((6aR,8R,9R,9aR)‐8‐(6‐benzamido‐9H‐purin‐9‐yl)‐2,2,4,4‐ tetraisopropyltetrahydro‐6H‐furo[3,2‐f][1,3,5,2,4]trioxadisilocin‐9‐ yl)oxy)methoxy)ethoxy)^ethan‐1‐ammonium^formate^(1‐6)
Figure imgf000129_0001
[00422] A solution of compound 1-1 (25.00 g, 67.38 mmol) in 20 mL of DMF was treated with pyridine (11 mL, 134.67 mmol) and tetraisopropyldisiloxane dichloride (22.63 mL, 70.75 mmol) at 10 °C. The resulting mixture was stirred at 25 °C for 3 h and quenched with 20% citric acid (50 mL). The aqueous layer was extracted with EtOAc (3X50 mL) and the combined organic layers were concentrated in vacuo. The crude residue was recrystallized from a mixture of MTBE and n-heptane (1:15, 320 mL) to afford compound 1- 2 (37.20 g, 90%) as a white oily solid. [00423] A solution of compound 1-2 (37.00 g, 60.33 mmol) in 20 mL of DMSO was treated with AcOH (20 mL, 317.20 mmol) and Ac2O (15 mL, 156.68 mmol). The mixture was stirred at 25 °C for 15 h. The reaction was diluted with EtOAc (100 mL) and quenched with sat. K2CO3 (50 mL). The aqueous layer was extracted with EtOAc (3X50 mL). The combined organic layers were concentrated and recrystallized with ACN (30 mL) to afford compound 1-3 (15.65 g, 38.4%) as a white solid. [00424] A solution of compound 1-3 (20.00 g, 29.72 mmol) in 120 mL of DCM was treated with Fmoc-amino-ethoxy ethanol (11.67 g, 35.66 mmol) at 25 °C. The mixture was stirred to afford a clear solution and then treated with 4Å molecular sieves (20.0 g), N- iodosuccinimide (8.02 g, 35.66 mmol), and TfOH (5.25 mL, 59.44 mmol). The mixture was stirred at 30 °C until the HPLC analysis indicated >95% consumption of compound 1-3. The reaction was quenched with TEA (6 mL) and filtered. The filtrate was diluted with EtOAc, washed with sat. NaHCO3 (2X100 mL), sat. Na2SO3 (2X100 mL), and water (2X100 mL) and concentrated in vacuo to afford crude compound 1-4 (26.34 g, 93.9%) as a yellow solid, which was used directly for the next step without further purification. [00425] A solution of compound 1-4 (26.34 g, 27.62 mmol) in a mixture of DCM/water (10:7, 170 mL) was treated with DBU (7.00 mL, 45.08 mmol) at 5 °C. The mixture was stirred at 5-25 °C for 1 h. The organic layer was then separated, washed with water (100 mL), and diluted with DCM (130 mL). The solution was treated with fumaric acid (7.05 g, 60.76 mmol) and 4Å molecular sieves (26.34 g) in four portions. The mixture was stirred for 1 h, concentrated, and recrystallized from a mixture of MTBE and DCM (5:1) to afford compound 1-6 (14.74 g, 62.9%) as a white solid: 1H NMR (400 MHz, d6-DMSO) 8.73 (s, 1H), 8.58 (s, 1H), 8.15-8.02 (m, 2H), 7.65-7.60 (m, 1H), 7.59-7.51 (m, 2H), 6.52 (s, 2H), 6.15(s, 1H), 5.08-4.90 (m, 3H), 4.83-4.78 (m, 1H), 4.15-3.90 (m, 3H), 3.79-3.65 (m, 2H), 2.98-2.85 (m, 6H), 1.20-0.95 (m, 28H). Example 1b: Synthesis of (2R,3R,4R,5R)‐5‐(6‐benzamido‐9H‐purin‐9‐yl)‐2‐((bis(4‐ methoxyphenyl)(phenyl)methoxy)methyl)‐4‐((2‐(2‐[lipid]‐ amidoethoxy)ethoxy)methoxy) tetrahydrofuran‐3‐yl (2‐cyanoethyl)^ diisopropylphosphoramidite(2‐4a^to^2‐4e)^
Figure imgf000131_0001
[00426] A solution of compound 1-6 (50.00 g, 59.01 mmol) in 150 mL of 2- methyltetrahydrofuran was washed with ice cold aqueous K2HPO4 (6%, 100 mL) and brine (20%, 2X100 mL). The organic layer was separated and treated with hexanoic acid (10.33 mL, 82.61 mmol), HATU (33.66 g, 88.52 mmol), and DMAP (10.81 g, 147.52 mmol) at 0 °C. The resulting mixture was warmed to 25 °C and stirred for 1 h. The solution was washed with water (2X100 mL), brine (100 mL), and concentrated in vacuo to afford a crude residue. Flash chromatography on silica gel (1:1 hexanes/acetone) gave compound 2-1a (34.95 g, 71.5%) as a white solid. [00427] A mixture of compound 2-1a (34.95 g, 42.19 mmol) and TEA (9.28 mL, 126.58 mmol) in 80 mL of THF was treated with triethylamine trihydrofluoride (20.61 mL, 126.58 mmol) dropwise at 10 °C. The mixture was warmed to 25 °C and stirred for 2 h. The reaction was concentrated, dissolved in DCM (100 mL), and washed with sat. NaHCO3 (5X20 mL) and brine (50 mL). The organic layer was concentrated in vacuo to afford crude compound 2-2a (24.72 g, 99%), which was used directly for the next step without further purification. [00428] A solution of compound 2-2a (24.72 g, 42.18 mmol) in 50 mL of DCM was treated with N-methylmorpholine (18.54 mL, 168.67 mmol) and DMTr-Cl (15.69 g, 46.38 mmol). The mixture was stirred at 25 °C for 2 h and quenched with sat. NaHCO3 (50 mL). The organic layer was separated, washed with water, concentrated to afford a slurry crude. Flash chromatography on silica gel (1:1 hexanes/acetone) gave compound 2-3a (30.05 g, 33.8 mmol, 79.9%) as a white solid. [00429] A solution of compound 2-3a (25.00 g, 28.17 mmol) in 50 mL of DCM was treated with N-methylmorpholine (3.10 mL, 28.17 mmol) and tetrazole (0.67 mL, 14.09 mmol) under nitrogen atmosphere. Bis(diisopropylamino) chlorophosphine (9.02 g, 33.80 mmol) was added to the solution dropwise and the resulting mixture was stirred at 25 °C for 4 h. The reaction was quenched with water (15 mL), and the aqueous layer was extracted with DCM (3X50 mL). The combined organic layers were washed with sat. NaHCO3 (50 mL), concentrated to afford a crude solid that was recrystallized from a mixture of DCM/MTBE/n- hexane (1:4:40) to afford compound 2-4a (25.52 g, 83.4%) as a white solid: 1H NMR (400 MHz, d6-DMSO) 11.25 (s, 1H), 8.65-8.60 (m, 2 H), 8.09-8.02 (m, 2H), 7.71 (s, 1H), 7.67- 7.60 (m, 1H), 7.59-7.51 (m, 2H), 7.38-7.34 (m, 2H), 7.30-7.25 (m, 7H), 6.85-6.79 (m, 4H), 6.23-6.20 (m, 1H), 5.23-5.14 (m, 1H), 4.80-4.69 (m, 3H), 4.33-4.23 (m, 2H), 3.90-3.78 (m, 1H), 3.75 (s, 6H), 3.74-3.52 (m, 3H), 3.50-3.20 (m, 6H), 3.14-3.09 (m, 2H), 3.09 (s, 1H), 2.82-2.80 (m, 1H), 2.65-2.60 (m, 1H), 2.05-1.96 (m, 2H), 1.50-1.39 (m, 2H), 1.31-1.10 (m, 14H), 1.08-1.05 (m, 2 H), 0.85-0.79 (m, 3H); 31P NMR (162 MHz, d6-DMSO) 149.43, 149.18. [00430] Compound 2-4b, 2-4c, 2-4d, and 2-4e were prepared using similar procedures described above for compound 2-4a. Compound 2-4b was obtained (25.50 g, 85.4%) as a white solid: 1H NMR (400 MHz, d6-DMSO) 11.23 (s, 1H), 8.65-8.60 (m, 2 H), 8.05-8.02 (m, 2H), 7.73-7.70 (m, 1H), 7.67-7.60 (m, 1H), 7.59-7.51 (m, 2H), 7.38-7.34 (m, 2H), 7.30-7.25 (m, 7H), 6.89-6.80 (m, 4H), 6.21-6.15 (m, 1H), 5.23-5.17 (m, 1H), 4.80-4.69 (m, 3H), 4.40- 4.21 (m, 2H), 3.91-3.80 (m, 1H), 3.74 (s, 6H), 3.74-3.52 (m, 3H), 3.50-3.20 (m, 6H), 3.14- 3.09 (m, 2H), 3.09 (s, 1H), 2.83-2.79 (m, 1H), 2.68-2.62 (m, 1H), 2.05-1.97 (m, 2H), 1.50- 1.38 (m, 2H), 1.31-1.10 (m, 18H), 1.08-1.05 (m, 2H), 0.85-0.78 (m, 3H); 31P NMR (162 MHz, d6-DMSO) 149.43, 149.19. [00431] Compound 2-4c was obtained (36.60 g, 66.3%) as an off-white solid: 1H NMR (400 MHz, d6-DMSO) 11.22 (s, 1H), 8.64-8.59 (m, 2H), 8.05-8.00 (m, 2H), 7.73-7.70 (m, 1H), 7.67-7.60 (m, 1H), 7.59-7.51 (m, 2H), 7.38-7.34 (m, 2H), 7.30-7.25 (m, 7H), 6.89-6.80 (m, 4H), 6.21-6.15 (m, 1H), 5.25-5.17 (m, 1H), 4.80-4.69 (m, 3H), 4.40-4.21 (m, 2H), 3.91- 3.80 (m, 1H), 3.74 (s, 6H), 3.74-3.50 (m, 3H), 3.50-3.20 (m, 6H), 3.14-3.09 (m, 2H), 3.09 (s, 1H), 2.83-2.79 (m, 1H), 2.68-2.62 (m, 1H), 2.05-1.99 (m, 2H), 1.50-1.38 (m, 2H), 1.33-1.12 (m, 38H), 1.08-1.05 (m, 2 H), 0.86-0.80 (m, 3H); 31P NMR (162 MHz, d6-DMSO) 149.42, 149.17. [00432] Compound 2-4d was obtained (26.60 g, 72.9%) as an off-white solid: 1H NMR (400 MHz, d6-DMSO) 11.22 (s, 1H), 8.64-8.59 (m, 2H), 8.05-8.00 (m, 2H), 7.73-7.70 (m, 1H), 7.67-7.60 (m, 1H), 7.59-7.51 (m, 2H), 7.38-7.33 (m, 2H), 7.30-7.25 (m, 7H), 6.89- 6.80 (m, 4H), 6.21-6.15 (m, 1H), 5.22-5.17 (m, 1H), 4.80-4.69 (m, 3H), 4.40-4.21 (m, 2H), 3.91-3.80 (m, 1H), 3.74 (s, 6H), 3.74-3.52 (m, 3H), 3.50-3.20 (m, 6H), 3.14-3.09 (m, 2H), 3.09 (s, 1H), 2.83-2.79 (m, 1H), 2.68-2.62 (m, 1H), 2.05-1.99 (m, 2H), 1.50-1.38 (m, 2H), 1.35-1.08 (m, 38H), 1.08-1.05 (m, 2 H), 0.85-0.79 (m, 3H); 31P NMR (162 MHz, d6-DMSO) 149.47, 149.22. [00433] Compound 2-4e was obtained (38.10 g, 54.0%) as a white solid: 1H NMR (400 MHz, d6-DMSO) 11.21 (s, 1H), 8.64-8.59 (m, 2H), 8.05-8.00 (m, 2H), 7.73-7.70 (m, 1H), 7.67-7.60 (m, 1H), 7.59-7.51 (m, 2H), 7.38-7.34 (m, 2H), 7.30-7.25 (m, 7H), 6.89-6.80 (m, 4H), 6.21-6.15 (m, 1H), 5.23-5.17 (m, 1H), 4.80-4.69 (m, 3H), 4.40-4.21 (m, 2H), 3.91-3.80 (m, 1H), 3.73 (s, 6H), 3.74-3.52 (m, 3H), 3.47-3.22 (m, 6H), 3.14-3.09 (m, 2H), 3.09 (s, 1H), 2.83-2.79 (m, 1H), 2.68-2.62 (m, 1H), 2.05-1.99 (m, 2H), 1.50-1.38 (m, 2H), 1.35-1.06 (m, 46H), 1.08-1.06 (m, 2 H), 0.85-0.77 (m, 3H); 31P NMR (162 MHz, d6-DMSO) 149.41, 149.15. Example^2.^^Synthesis^of^GalXC^RNAi^Oligonucleotide‐Lipid^Conjugates Scheme^1. Synthesis of GalXC RNAi oligonucleotide-lipid conjugates with mono-lipid (linear and branched) conjugated to the tetraloop. Post-synthetic conjugation was realized through amide coupling reactions.
Figure imgf000134_0001
R1COOH group represents fatty acid C8:0, C10:0, C11:0, C12:0, C14:0, C16:0, C17:0, C18:0, C18:1, C18:2, C22:5, C22:0, C24:0, C26:0, C22:6, C24:1, diacyl C16:0 or diacyl C18:1
Figure imgf000134_0002
[00434] Synthesis Sense 1 and Antisense 1 were prepared by solid-phase synthesis. Synthesis of Conjugated Sense 1a-1i. [00435] Conjugated Sense 1a was synthesized through post-syntenic conjugation approach. In Eppendorf tube 1, a solution of octanoic acid (0.58 mg, 4 umol) in DMA (0.75 mL) was treated with HATU (1.52 mg, 4 umol) at rt. In Eppendorf tube 2, a solution of oligo Sense 1 (10.00 mg, 0.8 umol) in H2O (0.25 mL) was treated with DIPEA (1.39 uL, 8 umol). The solution in Eppendorf tube 1 was added to the Eppendorf tube 2 and mixed using Thermomixer at rt. After the reaction was completed indicated by LC-MS analysis, the reaction mixture was diluted with 5 mL of water and purified by revers phase XBridge C18 column using a 5-95% gradient of 100 mM TEAA in ACN and H2O. The product fractions were concentrated under reduced pressure using Genevac. The combined residual solvent was dialyzed against water (1 X), saline (1 X), and water (3 X) using Amicon® Ultra-15 Centrifugal (3K). The Amicon membrane was washed with water (3 X 2 mL) and the combined solvents were then lyophilized to afford an amorphous white solid of Conjugated Sense 1a (6.43 mg, 64% yield). [00436] Conjugated Sense 1b-1i were prepared using similar procedures as described for the synthesis of Conjugated Sense 1a and obtained in 42%-69% yields. Annealing of Duplex 1a-1j. [00437] Conjugated Sense 1a (10 mg, measured by weight) was dissolved in 0.5 mL deionized water to prepare a 20 mg/mL solution. Antisense 1 (10 mg, measured by OD) was dissolved in 0.5 mL deionized water to prepare a 20 mg/mL solution, which was used for the titration of the conjugated sense and quantification of the duplex amount. Based on the calculation of molar amounts of both conjugated sense and antisense, a proportion of required Antisense 1 was added to the Conjugated Sense 1a solution. The resulting mixture was stirred at 95 °C for 5 min and allowed to cool down to rt. The annealing progress was monitored by ion-exchange HPLC. Based on the annealing progress, several proportions of Antisense 1 were further added to complete the annealing with >95% purity. The solution was lyophilized to afford Duplex 1a (C8) and its amount was calculated based on the molar amount of the antisense consumed in the annealing. [00438] Duplex 1b-1i were prepared using the same procedures as described for the annealing of Duplex 1a (C8). [00439] The following Scheme 1-2 depicts the synthesis of Nicked tetraloop GalXC conjugates with mono-lipid on the loop. Post-synthetic conjugation was realized through Cu- catalyzed alkyne-azide cycloaddition reaction.
Figure imgf000136_0001
Scheme1-2 [00440] Sense 1B and Antisense 1B were prepared by solid-phase synthesis. Synthesis of Conjugated Sense 1j. [00441] In Eppendorf tube 1, a solution of oligo (10.00 mg, 0.8 umol) in a 3:1 mixture of DMA/ H2O (0.5 mL) was treated with the lipid linker azide (11.26 mg, 4 umol). In Eppendorf tube 2, CuBr dimethyl sulfide (1.64 mg, 8 umol) was dissolved in ACN (0.5 mL). Both solutions were degassed for 10 min by bubbling N2 through them. The ACN solution of CuBrSMe2 was then added into tube 1 and the resulting mixture was stirred at 40 °C. After the reaction was completed indicated by LC-MS analysis, the reaction mixture was diluted with 0.5 M EDTA (2 mL) and dialyzed against water (2 X) using a Amicon® Ultra-15 Centrifugal (3K). The reaction crude was purified by revers phase XBridge C18 column using a 5-95% gradient of 100 mM TEAA in ACN (with 30% IPA spiked in) and H2O. The product fractions were concentrated under reduced pressure using Genevac. The combined residual solvent was dialyzed against water (1 X), saline (1 X), and water (3 X) using Amicon® Ultra-15 Centrifugal (3K). The Amicon membrane was washed with water (3 X 2 mL) and the combined solvents were lyophilized to afford an amorphous white solid of Conjugated Sense 1j (6.90 mg, 57% yield). [00442] Duplex 1j (PEG2K-diacyl C18) was prepared using the same procedures as described for the annealing of Duplex 1a (C8). [00443] The following Scheme 1-3 depicts the synthesis of Nicked tetraloop GalXC conjugates with di-lipid on the loop using post-synthetic conjugation approach.
Figure imgf000137_0001
Scheme1-3 Sense 2 and Antisense 2 were prepared by solid-phase synthesis. [00444] Conjugated Sense 2a and 2b were prepared using similar procedures as described for the synthesis of Conjugated Sense 1a but with 10 eq of lipid, 10 eq of HATU, and 20 eq of DIPEA. [00445] Duplex 2a (2XC11) and 2b (2XC22) were prepared using the same procedures as described for the annealing of Duplex 1a (C8). [00446] The following Scheme 1-4 depicts the synthesis of GalXC of fully phosphorothioated stem-loop conjugated with mono-lipid using post-synthetic conjugation approach.
Figure imgf000138_0001
Scheme1-4 Sense 3 and Antisense 3 were prepared by solid-phase synthesis. [00447] Conjugated Sense 3a was prepared using similar procedures as described for the synthesis of Conjugated Sense 1a and obtained in a 65% yield. [00448] Duplex 3a (PS-C22) was prepared using the same procedures as described for the annealing of Duplex 1a (C8). [00449] The following Scheme1-5 depicts the synthesis of GalXC of short sense conjugated with mono-lipid using post-synthetic conjugation approach.
Figure imgf000139_0001
Scheme 1-5 Sense 4 and Antisense 4 were prepared by solid-phase synthesis. [00450] Conjugated Sense 4a was prepared using similar procedures as described for the synthesis of Conjugated Sense 1a and obtained in a 74% yield. [00451] Duplex 4a (SS-C22) was prepared using the same procedures as described for the annealing of Duplex 1a (C8). [00452] The following Scheme 1-6 depicts the synthesis of Nicked tetraloop GalXC conjugated with tri-adamantane moiety on the loop using post-synthetic conjugation approach.
Figure imgf000140_0001
Sense 5 and Antisense 5 were prepared by solid-phase synthesis. [00453] Conjugated Sense 5a and 5b were prepared using similar procedures as described for the synthesis of Conjugated Sense 1a and obtained in 42%-73% yields. [00454] Duplex 5a (3Xadamantane) and Duplex 5b (3Xacetyladamantane) were prepared using the same procedures as described for the annealing of Duplex 1a (C8). [00455] The following scheme 1-7 depicts an example of solid phase synthesis of Nicked tetraloop GalXC conjugated with lipid(s) on the loop.
Figure imgf000141_0001
Scheme 1-7 Synthesis of Conjugated Sense 6. [00456] Conjugated Sense 6 was prepared by solid-phase synthesis using a commercial oligo synthesizer. The oligonucleotides were synthesized using 2’-modified nucleoside phosphoramidites, such as 2’-F or 2’-OMe, and 2'-diethoxymethanol linked fatty acid amide nucleoside phosphoramidites. Oligonucleotide synthesis was conducted on a solid support in the 3’ to 5’direction using a standard oligonucleotide synthesis protocol. In these efforts, 5-ethylthio-1H-tetrazole (ETT) was used as an activator for the coupling reaction. Iodine solution was used for phosphite triester oxidation.3- (Dimethylaminomethylidene)amino-3H-1,2,4-dithiazole-3-thione (DDTT) was used for the formation of phosphorothioate linkages. Synthesized oligonucleotides were treated with concentrated aqueous ammonium for 10 h. The ammonia was removed from the suspension and the solid support residues were removed by filtration. The crude oligonucleotide was treated with TEAA, analyzed, and purified by strong anion exchange high performance liquid chromatography (SAX-HPLC). The fractions were combined and dialyzed against water (3 X), saline (1 X), and water (3 X) using Amicon® Ultra-15 Centrifugal (3K). The remaining solvent was then lyophilized to afford the desired Conjugated Sense 6. [00457] Duplex 6 was prepared using the same procedures as described for the annealing of Duplex 1a (C8). Scheme^8. Synthesis of Nicked tetraloop GalXC conjugated with one adamantane unit on the loop
Figure imgf000142_0001
N = 0: Adamantane Carboxylic Acid; n = 1: Adamantane Acetic Acid Scheme^1‐8 Synthesis of Conjugated Sense^7a^and^7b^ [00458] Conjugated Sense 7a and Sense 7b were obtained using the same method or a substantially similar method to the synthesis of Conjugated Sense 5. Synthesis example of Duplex 7a and 7b [00459] Duplex 7a and Duplex 7b were obtained using the same method or a substantially similar method to the synthesis of Duplex 5. [00460] Scheme 9. Synthesis of nicked tetraloop GalXC conjugated with two adamantane units on the loop via a post-synthetic conjugation approach.
Figure imgf000143_0001
8a, n = 0 Duplex 8 8b, n = 1 Scheme^1‐9 Synthesis of Conjugated Sense^8a^and^8b^ [00461] Conjugated Sense 8a and Sense 8b were obtained using the same method or a substantially similar method to the synthesis of Conjugated Sense 5. Synthesis example of Duplex 8a and 8b [00462] Duplex 8a and Duplex 8b were obtained using the same method or a substantially similar method to the synthesis of Duplex 5. [00463] The following Scheme1-10 depicts the synthesis of GalXC of short sense and short stem loop conjugated with mono-lipid using post-synthetic conjugation approach.
Figure imgf000144_0001
Scheme 1-10 Synthesis of Sense 9a [00464] Conjugated Sense 9a was obtained using the same method or a substantially similar method to the synthesis of Conjugated Sense 5. Synthesis example of Duplex 9a [00465] Duplex 9a was obtained using the same method or a substantially similar method to the synthesis of Duplex 5. [00466] The following Scheme1-11 depicts the synthesis of GalXC conjugated with mono-lipid at 5’-end using post-synthetic conjugation approach.
Figure imgf000145_0001
Scheme 1-11 Synthesis of Conjugated Sense 10a [00467] Conjugated Sense 10a was obtained using the same method or a substantially similar method to the synthesis of Conjugated Sense 5. Synthesis example of Duplex 10a [00468] Duplex 10a was obtained using the same method or a substantially similar method to the synthesis of Duplex 5. [00469] The following Scheme1-12a and 1-12b depict the synthesis of GalXC with blunt end conjugated with mono-lipid at 3’-end or 5’-end using post-synthetic conjugation approach.
Figure imgf000146_0001
Scheme 1-12a
Figure imgf000147_0001
Scheme 1-12b Synthesis of Conjugated Sense 11a and 12a [00470] Conjugated Sense 11a and 12a were obtained using the same method or a substantially similar method to the synthesis of Conjugated Sense 5. Synthesis example of Duplex 11a and 12a [00471] Duplex 11a and 12a were obtained using the same method or a substantially similar method to the synthesis of Duplex 5. [00472] Conjugates Duplex 8D and Duplex 9D were obtained using the same method or a substantially similar method to the synthesis of Duplex 5. [00473] Later, acyl chains were conjugated to a nucleic acid inhibitor molecule that targets the STAT3 gene, a gene that is expressed in the tissues of interest. A passenger strand with 2’-amine linkers [ademA] was used for post solid phase conjugation. Different types of lipids were conjugated using the same chemistry to generate a series of conjugates (FIG.1A and 1B). SAR studies were performed to identify a lipid conjugate that could be used to deliver payloads to the tissues of interest in order to mediate target knockdown. Example 3: Tissue Specific Targets in MDSC Cell Populations and Tumor Draining Lymph Nodes. [00474] STAT3 is involved in immune suppression with examples abundantly reported in literature. Targeting STAT3 transcription through an RNAi mechanism could potentially overcome the challenges in the development of pharmacological STAT3 inhibitors. For these reasons STAT3 was selected as a proof-of-concept target to demonstrate tissue specific activity in the tissues of interest, such as myeloid derived suppressor cells (MDSCs). STAT3 sequences were designed in the GalXC format with described modification patterns and screening for target knockdown in liver tissue was performed in normal CD-1 mice. Eighteen STAT3-GalXC conjugates (Table 1) were dosed once subcutaneously at 3 mg/kg. Table 1: GalXC Compound Candidates for Identifying Tool Compounds for Proof-of- concept Studies in Mice:
Figure imgf000148_0001
Figure imgf000149_0001
[00475] Five days post injection, livers were collected and subjected to mRNA analysis by qPCR. As a result of the screen, four sequences (GalXC -STAT3-838, GalXC- STAT3-1402, GalXC-STAT3-4110 and GalXC-STAT3-4123) that showed >85% target knockdown in liver were selected for further evaluation (FIG.2A). Of these sequences three were identified as mouse specific and one was identified as human-mouse cross-reactive. These 4 sequences were further screened in CD-1 mice at 3 different doses (0.3, 1 and 3 mg/kg) to assess the dose response. GalXC-STAT3-4110 and 4123 were identified as the most potent sequences after the dose response screen, each with ED50 of 0.3 mg/kg and thus these molecules were selected for further studies (FIG.2B). C18 lipid conjugation was performed for both GalXC-STAT3-4110 or 4123 for proof-of-concept studies (Table 2). Table 2: GalXC-STAT3 Lipid Conjugates
Figure imgf000149_0002
Figure imgf000150_0001
Table 3: GalXC-STAT3 Lipid Conjugates
Figure imgf000150_0002
[00476] To evaluate the performance of GalXC-STAT3-C18 conjugates, Pan02 tumors were implanted in nude mice and upon reaching sufficient tumor volume mice were subjected to randomization as previously described. Mice received either a single dose of GalXC- STAT3-C184110 and 4123 subcutaneously at 25 mg/kg, 50 mg/kg, or PBS. At 3 days post injection, bulk tumors were collected and MDSC subsets were isolated. Collectively, MDSCs are characterized by the co-expression of cell surface or mRNA markers CD11b (a marker for the myeloid cells of the macrophage lineage) and Gr-1(a marker for the myeloid lineage differentiation antigen) and denoted as CD11b+Gr-1+ cells. Gr-1 is further comprised of 2 components Ly6G and Ly6C. MDSCs consist of two subsets: Granulocytic MDSC (G- MDSC), further characterized as CD11b+Ly6G+Ly6Clo, and monocytic MDSC (M-MDSC) characterized as CD11b+Ly6G-Ly6Chi. To isolate the CD11b positive cells, a single cell suspension of tumor was made using gentle MACS dissociator. CD11b positive cells in the single cell suspension were then magnetically labeled with MACS microbeads and enriched by passing through MACS columns and subsequently eluting the retained labeled cells in the column as positively selected fractions (CD11b MicroBeads UltraPure, mouse kit Cat# 130- 126-725). For tumor cell separation, non-target cells in the cell suspension were magnetically labeled with a cocktail of microbeads and passed through the MACS columns. During this process, the unwanted labeled cells were retained in the column and the unlabeled target cells (tumor cells) were collected in the flow-through as pure fraction. (Tumor Cell Isolation Kit, human Cat # 130-108-339). [00477] Following cell isolation mRNA was analyzed by qPCR (FIGs.3A and 3B). Stat3 mRNA levels were reduced by ~40% in G-MDSC and M-MDSCs by GalXC-STAT3- C18-4123. GalXC-STAT3-C18-4110 reduced the Stat3 mRNA levels only by 20% in both MDSC subsets. [00478] To understand how the dose level of GalXC-STAT3-C18 conjugates plays a role in trafficking of these molecules to different tissues and cell subsets, a follow-up study was performed as previously described with the same tumor model. Pan02 tumor bearing mice were treated with a single subcutaneous dose of either GalXC-STAT3-C18-4123 at 50 mg/kg, or PBS and Stat3 mRNA levels were measured after 3 days. The Stat3 knockdown in G-MDSC was not significantly altered as compared to the knockdown observed at the 25 mg/kg dose, however there was a significant improvement in Stat3 silencing observed in M- MDSC subset at this same dose level. In parallel study performed as previously described, Stat3 knockdown was assessed in bulk tumors and TdLNs on day 7 (FIGs.4A and 4B). Dose dependent Stat3 mRNA knockdown was observed in bulk tumor with both GalXC- STAT3-C18 sequences. In TdLNs Stat3 mRNA levels were reduced by ~60-65% by GalXC- STAT3-C18-4123, ~25-30% by GalXC-STAT3-C18-4110 at both doses suggesting a saturation effect at these dose levels. Based on the data, GalXC-STAT3-C18-4123 was selected for further efficacy evaluations in immunocompetent mice. Example 4: STAT3 Inhibition Decreases the PD-L1 Levels in MDSCs and Mediates Acute Tumor Effects [00479] The transcriptional signature of phosphorylated STAT3 has been positively correlated with PD-L1 expression in tumors (Song et al, JOURNAL OF CELL PHYSIOLOGY (2020), Zerdes et al, CANCERS (2019), Song et al, BLOOD (2018). To extrapolate this correlation to STAT3 expressed by MDSCs, isolated populations of MDSCs treated with either PBS or a GalXC-STAT3 conjugate were assayed for Pdl1 mRNA. Pdl1 mRNA levels were decreased by ~80% in both G-MDSC and M-MDSC populations treated with either 25 or 50 mg/kg of a GalXC-STAT3 (FIG.5A). The Pdl1 levels were also dramatically reduced in TdLN after treatment with the GalXC-STAT3 conjugate, specifically GalXC-STAT3-C18- 4123 (FIG.5B). These data suggest a potential for downstream immunomodulation of PD- L1 after knockdown of STAT3. [00480] In a separate study, a Pan02 (murine pancreatic syngeneic model) tumor bearing C57BL/6 mice (n=4 per group) were treated subcutaneously with GalXC-STAT3- C18 conjugate following a split dosing model where all animals received a total dose of 50 mg/kg, dosed as either 25 mg/kg x 2 doses or 12.5 mg/kg x 4 doses. Tumors treated using the 25 mg/kg split dose showed acute tumor regression, even after the first dose (FIG.6B). After the second dose of 25 mg/kg, tumors from 3 out of 4 mice regressed to sizes that were too small to be collected for further processing. The anti-tumor effect of the GalXC-STAT3 treatment was also observed in mice that received the 12.5 mg/kg split doses (FIG.6A). These data suggest that STAT3 mediated regulation of PD-L1 results in an acute and dramatic effect on tumor growth in the Pan02 tumor bearing immunocompetent mice. Example 5: Preparation of Double-Stranded RNAi Oligonucleotides Oligonucleotide Synthesis and Purification [00481] The double-stranded RNAi (dsRNA) oligonucleotides described in the foregoing Examples were chemically synthesized using methods described herein. Generally, dsRNAi oligonucleotides were synthesized using solid phase oligonucleotide synthesis methods as described for 19-23mer siRNAs (see, e.g., Scaringe et al. (1990) Nucleic Acids Res.18:5433-5441 and Usman et al. (1987) J. Am. Chem. Soc.109:7845-7845; see also, US Patent Nos.5,804,683; 5,831,071; 5,998,203; 6,008,400; 6,111,086; 6,117,657; 6,353,098; 6,362,323; 6,437,117 and 6,469,158) in addition to using known phosphoramidite synthesis (see, e.g. Hughes and Ellington (2017) Cold Spring Harb Perspect Biol. 9(1):a023812; Beaucage S.L., Caruthers M.H. Studies on Nucleotide Chemistry V: Deoxynucleoside Phosphoramidites—A New Class of Key Intermediates for Deoxypolynucleotide Synthesis. Tetrahedron Lett.1981;22:1859–1862. doi: 10.1016/S0040- 4039(01)90461-7). dsRNAi oligonucleotides having a 19mer core sequence were formatted into constructs having a 25mer sense strand and a 27mer antisense strand to allow for processing by the RNAi machinery. The 19mer core sequence is complementary to a region in the STAT3 mRNA. [00482] Individual RNA strands were synthesized and HPLC purified according to standard methods (Integrated DNA Technologies; Coralville, IA). For example, RNA oligonucleotides were synthesized using solid phase phosphoramidite chemistry, deprotected and desalted on NAP-5 columns (Amersham Pharmacia Biotech; Piscataway, NJ) using standard techniques (Damha & Olgivie (1993) Methods Mol. Biol.20:81-114; Wincott et al. (1995) Nucleic Acids Res.23:2677-2684). The oligomers were purified using ion-exchange high performance liquid chromatography (IE-HPLC) on an Amersham Source 15Q column (1.0 cm×25 cm; Amersham Pharmacia Biotech) using a 15 min step-linear gradient. The gradient varied from 90:10 Buffers A:B to 52:48 Buffers A:B, where Buffer A is 100 mM Tris pH 8.5 and Buffer B is 100 mM Tris pH 8.5, 1 M NaCl. Samples were monitored at 260 nm and peaks corresponding to the full-length oligonucleotide species were collected, pooled, desalted on NAP-5 columns, and lyophilized. [00483] The purity of each oligomer was determined by capillary electrophoresis (CE) on a Beckman PACE 5000 (Beckman Coulter, Inc.; Fullerton, CA). The CE capillaries have a 100 μm inner diameter and contain ssDNA 100R Gel (Beckman-Coulter). Typically, about 0.6 nmole of oligonucleotide was injected into a capillary, run in an electric field of 444 V/cm and was detected by UV absorbance at 260 nm. Denaturing Tris-Borate-7 M-urea running buffer was purchased from Beckman-Coulter. Oligoribonucleotides were obtained that were at least 90% pure as assessed by CE for use in experiments described below. Compound identity was verified by matrix-assisted laser desorption ionization time-of-flight (MALDI-TOF) mass spectroscopy on a Voyager DE™ Biospectometry Work Station (Applied Biosystems; Foster City, CA) following the manufacturer′s recommended protocol. Relative molecular masses of all oligomers were obtained, often within 0.2% of expected molecular mass. Preparation^of^Duplexes^ [00484] Single strand RNA oligomers were resuspended (e.g., at 100 μM concentration) in duplex buffer consisting of 100 mM potassium acetate, 30 mM HEPES, pH 7.5. Complementary sense and antisense strands were mixed in equal molar amounts to yield a final solution of, for example, 50 μM duplex. Samples were heated to 100°C for 5′ in RNA buffer (IDT) and were allowed to cool to room temperature before use. The dsRNA oligonucleotides were stored at −20° C. Single strand RNA oligomers were stored lyophilized or in nuclease-free water at −80° C. Example 6: Generation of STAT3-Targeting Double-Stranded RNAi Oligonucleotides Identification of STAT3^mRNA Target Sequences [00485] Signal transducer and activator of transcription 3 (STAT3) is a transcription factor involved in several development and disease functions. To generate RNAi oligonucleotide inhibitors of STAT3 expression, a computer-based algorithm was used to computationally identify STAT3 mRNA target sequences suitable for assaying inhibition of STAT3 expression by the RNAi pathway. The algorithm provided RNAi oligonucleotide guide (antisense) strand sequences each having a region of complementarity to a suitable STAT3 target sequence of human STAT3 mRNA (e.g., SEQ ID NO:1217; Table 4). Some of the guide strand sequences identified by the algorithm were also complementary to the corresponding STAT3 target sequence of monkey STAT3 mRNA (SEQ ID NO: 1218 Table 4) and/or mouse STAT3 mRNA. STAT3 RNAi oligonucleotides comprising a region of complementarity to homologous STAT3 mRNA target sequences with nucleotide sequence similarity are predicted to have the ability to target homologous STAT3 mRNAs. Table 4: Sequences of Human and Monkey STAT3 mRNA
Figure imgf000154_0001
[00486] RNAi oligonucleotides (formatted as DsiRNA oligonucleotides) were generated as described in Example 5 for evaluation in vitro. Each DsiRNA was generated with the same modification pattern, and each with a unique guide strand having a region of complementarity to a STAT3 target sequence identified by SEQ ID NOs: 89-280. Modifications for the sense and anti- sense DsiRNA included the following (X- any nucleotide ; m- 2’-O-methyl modified nucleotide; r- ribosyl modified nucleotide): Sense Strand: rXmXrXmXrXrXrXrXrXrXrXrXrXmXrXmXrXrXrXrXrXrXrXXX Anti-sense Strand: mXmXmXmXrXrXrXrXrXrXmXrXmXrXrXrXrXrXrXrXrXrXmXrXmXmXmX [00487] The ability of each of the modified DsiRNA in Table 5 to reduce STAT3 mRNA was measured using in vitro cell-based assays. Briefly, human hepatocyte (Huh7) cells expressing endogenous human STAT3 gene were transfected with each of the DsiRNAs listed in Table 5 at 1 nM in separate wells of a multi-well cell-culture plate. Cells were maintained for 24 hours following transfection with the modified DsiRNA, and then the amount of remaining STAT3 mRNA from the transfected cells was determined using TAQMAN®-based qPCR assays. Two qPCR assays, a 3′ assay and 5’ assay (Forward 1- SEQ ID NO:1219), Reverse 1- SEQ ID NO:1220, Probe 1- SEQ ID NO: 1221; Forward 2- SEQ ID NO: 1, Reverse 2- SEQ ID NO: 2, Probe 2- SEQ ID NO: 3) were used to determine STAT3 mRNA levels as measured using PCR probes conjugated to 6-carboxy-fluorescein (FAM). Each primer pair was assayed for % remaining RNA as shown in Table 5 and FIG. 7. DsiRNAs resulting in less than or equal to 10% STAT3 mRNA remaining in DsiRNA- transfected cells when compared to mock-transfected cells were considered DsiRNA “hits”. The Huh7 cell-based assay evaluating the ability of the DsiRNAs listed in Table 5 to inhibit STAT3 expression identified several candidate DsiRNAs. [00488] Taken together, these results show that DsiRNAs designed to target human STAT3 mRNA inhibit STAT3 expression in cells, as determined by a reduced amount of STAT3 mRNA in DsiRNA-transfected cells relative to control cells. These results demonstrate that the nucleotide sequences comprising the DsiRNA are useful for generating RNAi oligonucleotides to inhibit STAT3 expression. Further, these results demonstrate that multiple STAT3 mRNA target sequences are suitable for the RNAi-mediated inhibition of STAT3 expression. Table 5. Analysis of STAT3 mRNA in Huh7 cells
Figure imgf000155_0001
Figure imgf000156_0001
Figure imgf000157_0001
Figure imgf000158_0001
Figure imgf000159_0001
[00489] Following the initial in vitro screen, 48 constructs were selected for dosing studies. Huh7 cells were treated for 24 hours with 0.05nM, 0.3nM, or 1nM of oligonucleotide. mRNA was isolated and measured to determine a potent dose (FIG.8A). Of the tested oligonucleotides, 34 sequences were selected for further testing in vivo (Table 6 and FIG.8B). Table 6. Analysis of STAT3 mRNA in Huh7 Dosing Study
Figure imgf000159_0002
Figure imgf000160_0001
Example 7: RNAi Oligonucleotide Inhibition of STAT3 In Vivo [00490] The in vitro screening assay in Example 6 validated the ability of STAT3- targeting DsiRNAs to knock-down target mRNA. To confirm the ability of the RNAi oligonucleotides to knockdown STAT3 in vivo, an HDI mouse model was used. A subset of the DsiRNAs identified in Example 6 were used to generate corresponding double-stranded RNAi oligonucleotides comprising a nicked tetraloop GalNAc-conjugated structure (referred to herein as “GalNAc-conjugated STAT3 oligonucleotides” or “GalNAc- STAT3 oligonucleotides”) having a 36-mer passenger strand and a 22-mer guide strand (Table 8 and Table 9). Further, the nucleotide sequences comprising the passenger strand and guide strand have a distinct pattern of modified nucleotides and phosphorothioate linkages. Three of the nucleotides comprising the tetraloop were each conjugated to a GalNAc moiety (CAS#14131-60-3). The modification patterns used are illustrated below: Pattern 1 Sense Strand: 5’ mX-S-mX-mX-mX-mX-mX-mX-fX-fX-fX-fX[-mX-]16-[ademX-GalNAc]- [ademX-GalNAc]-[ademX-GalNAc]-mX-mX-mX-mX-mX-mX 3’. Hybridized to: Antisense Strand: 5’ [MePhosphonate-4O-mX]-S-fX-S-fX-fX-fX-mX-fX-mX-mX-fX-mX- mX-mX-fX-mX-mX-mX-mX-mX-mX-S-mX-S-mX 3’. Or, represented as: Sense Strand: [mXs][mX][mX][mX][mX][mX][mX][fX][fX][fX][fX][mX][mX][mX] [mX][mX][mX][mX][mX][mX][mX][mX][mX][mX][mX][mX][mX][ademA- GalNAc][ademA-GalNAc][ademA-GalNAc][mX][mX][mX][mX][mX][mX] Hybridized to: Antisense Strand: [MePhosphonate-4O-mXs][fXs][fX][fX][fX][mX][fX][mX] [mX][fX][mX][mX][mX][fX][mX][mX][mX][mX][mX][mXs][mXs][mX] Pattern 2 Sense Strand: 5’ mX-S-mX-mX-mX-mX-mX-mX-fX-fX-fX-fX[-mX-]16-[ademX-GalNAc]- [ademX-GalNAc]-[ademX-GalNAc]-mX-mX-mX-mX-mX-mX 3’. Hybridized to: Antisense Strand: 5’ [MePhosphonate-4O-mX]-S-fX-S-fX-S-fX-fX-mX-fX-mX-mX-fX- mX-mX-mX-fX-mX-mX-mX-mX-mX-mX-S-mX-S-mX 3’. Or, represented as: Sense Strand: [mXs][mX][mX][mX][mX][mX][mX][fX][fX][fX][fX][mX][mX][mX] [mX][mX][mX][mX][mX][mX][mX][mX][mX][mX][mX][mX][mX][ademA- GalNAc][ademA-GalNAc][ademA-GalNAc][mX][mX][mX][mX][mX][mX] Hybridized to: Antisense Strand: [MePhosphonate-4O-mXs][fXs][fXs][fX][fX][mX][fX][mX] [mX][fX][mX][mX][mX][fX][mX][mX][mX][mX][mX][mXs][mXs][mX] (Modification key: Table 7).
Figure imgf000162_0001
[00491] Oligonucleotides in Table 8 and Table 9 were evaluated in mice engineered to transiently express human STAT3 mRNA in hepatocytes of the mouse liver. Briefly, 6-8- week-old female CD-1 mice (n = 4-5) were subcutaneously administered the indicated GalNAc-conjugated STAT3 oligonucleotides at a dose of 1mg/kg formulated in PBS. A control group of mice (n = 3-4) were administered only PBS. Three days later (72 hours), the mice were hydrodynamically injected (HDI) with a DNA plasmid encoding the full human STAT3 gene (25µg) under control of a ubiquitous cytomegalovirus (CMV) promoter sequence. One day after introduction of the DNA plasmid, liver samples from HDI mice were collected. Total RNA derived from these HDI mice were subjected to qRT-PCR analysis to determine STAT3 mRNA levels as described in Example 6. mRNA levels were measured for human mRNA. The values were normalized for transfection efficiency using the NeoR gene included on the DNA plasmid. A benchmark control (STAT3-1388) comprising a different modification pattern, was used for both assays (Sense Strand SEQ ID NO: 1100; Antisense Strand SEQ ID NO: 1190). Table 8. GalNAc-Conjugated STAT3 RNAi Oligonucleotides for HDI screen
Figure imgf000163_0001
Table 9. GalNAc-Conjugated STAT3 RNAi Oligonucleotides for HDI screen
Figure imgf000163_0002
Figure imgf000164_0001
[00492] The results in FIGs.9A and 9B demonstrate that GalNAc-conjugated STAT3 oligonucleotides designed to target human STAT3 mRNA inhibited human STAT3 mRNA expression in HDI mice, as determined by a reduction in the amount of human STAT3 mRNA expression in liver samples from HDI mice treated with GalNAc-conjugated STAT3 oligonucleotides relative to control HDI mice treated with only PBS. [00493] A subset of the GalNAc-conjugated STAT3 oligonucleotides tested in FIGs. 9A and 9B were further validated in a dosing study. Specifically, dosing studies were carried out using nine GalNAc-conjugated STAT3 oligonucleotides (STAT3-715, STAT3-716, STAT3-717, STAT3-720, STAT3-721, STAT3-1145, STAT3- 1286, STAT3-1286, and STAT3-1287). Mice were hydrodynamically injected as described above and treated with 0.1mg/kg, 0.3mg/kg, or 1mg/kg of oligonucleotide. Livers were collected after one day, and STAT3 expression was measured to determine a potent dose (FIG.10). All GalNAc- conjugated STAT3 oligonucleotides were able to reduce STAT3 expression at a 1mg/kg dose and STAT3-1286 was able to reduce expression at a 0.3mg/kg dose. Overall, the HDI studies identified several potential GalNAc-conjugated STAT3 oligonucleotides for inhibiting STAT3 expression in liver. Example 8: Species Specific RNAi Oligonucleotide Inhibition of STAT3 In Vivo [00494] To confirm the ability of RNAi oligonucleotides to knockdown STAT3 in vivo, several cross species and species specific GalNAc-conjugated STAT3 oligonucleotides were generated. Specifically, triple common (targeting human, non-human primate, and mouse; Hs/Mf/Mm), human/mouse (Hs/Mm), and human specific (Hs) oligonucleotides were evaluated. Hs/Mf/Mm and Hs/Mm Commons [00495] Mice expressing endogenous mouse STAT3 in the liver were subcutaneously injected at a dose of 3mg/kg with the GalNAc-conjugated STAT3 oligonucleotides set forth in Table 10. Livers were collected after five days, and STAT3 expression was measured. Overall, the study identified several potential Hs/Mf/Mm GalNAc-conjugated STAT3 oligonucleotides for inhibiting STAT3 expression in liver (FIG.11). Table 10. GalNAc-Conjugated Human/Monkey/Mouse STAT3 RNAi Oligonucleotides for Endogenous STAT3 screen.
Figure imgf000165_0001
[00496] Human/Mouse GalNAc-conjugated STAT3 oligonucleotides set forth in Table 11 were tested in mice endogenously expressing mouse STAT3. As described above, mice were subcutaneously injected at a dose of 3mg/kg with oligonucleotide. Livers were collected after five days, and mouse STAT3 expression was measured. Overall, the study identified several potential Hs/Mm GalNAc-conjugated STAT3 oligonucleotides for inhibiting STAT3 expression in liver (FIG.12). ^ Table 11. GalNAc-Conjugated Human/Mouse STAT3 RNAi Oligonucleotides for Endogenous STAT3 Screen.
Figure imgf000166_0001
[00497] A subset of the GalNAc-conjugated STAT3 oligonucleotides tested in FIGs. 11 and 12 were further validated in a dosing study. Specifically, dosing studies were carried out using ten GalNAc-conjugated STAT3 oligonucleotides (STAT3-2626, STAT3-2627, STAT3-2408, STAT3-2412, STAT3-2139, STAT3-4909, STAT3- 461, STAT3-678, STAT3- 2148, and STAT3-2144). Mice endogenously expressing mouse STAT3 were subcutaneously injected with 0.3mg/kg, 1mg/kg, or 3mg/kg oligonucleotide. Livers were collected after five days, and mouse STAT3 expression was measured to determine a potent dose (FIGs.13A and 13B). Overall, the endogenous mouse STAT3 expression studies identified several potential GalNAc-conjugated STAT3 oligonucleotides for inhibiting mouse STAT3 expression in liver. Hs^Specific [00498] Using the HDI model described in Example 7, human specific GalNAc- conjugated STAT3 oligonucleotides were evaluated. Specifically, 6-8-week-old female CD-1 mice (n = 4-5) were subcutaneously administered the indicated GalNAc-conjugated STAT3 oligonucleotides (Table 12) at a dose of 1mg/kg formulated in PBS. A control group of mice (n = 3-4) were administered only PBS. Three days later (72 hours), the mice were hydrodynamically injected (HDI) with a DNA plasmid encoding the full human STAT3 gene (25µg) under control of a ubiquitous cytomegalovirus (CMV) promoter sequence. One day after introduction of the DNA plasmid, liver samples from HDI mice were collected. Total RNA derived from these HDI mice were subjected to qRT-PCR analysis to determine STAT3 mRNA levels. Table 12. GalNAc-Conjugated Human STAT3 RNAi Oligonucleotides for Exogenous STAT3 Screen.
Figure imgf000167_0001
[00499] The results in FIG.14 demonstrate that GalNAc-conjugated STAT3 oligonucleotides designed to target human STAT3 mRNA inhibited human STAT3 mRNA expression in HDI mice, as determined by a reduction in the amount of human STAT3 mRNA expression in liver samples from HDI mice treated with GalNAc-conjugated STAT3 oligonucleotides relative to control HDI mice treated with only PBS. [00500] A subset of the GalNAc-conjugated STAT3 oligonucleotides tested in FIG.14 were further validated in a dosing study. Specifically, dosing studies were carried out using five GalNAc-conjugated STAT3 oligonucleotides (STAT3-426, STAT3-432, STAT3-1068, STAT3-1388, and STAT3-2404). Mice were hydrodynamically injected as described above and treated with 0.3mg/kg, 1 mg/kg, or 3mg/kg of oligonucleotide. Livers were collected after one day, and human STAT3 expression was measured to determine a potent dose (FIG. 15). A dose of 1mg/kg was capable of reducing STAT3 mRNA by about 75%, thereby identifying several potential GalNAc-conjugated STAT3 oligonucleotides for inhibiting STAT3 expression in liver. The best 2 sequences from FIG.23 and the best sequence from FIG.28 are tested in the final HDI screen (FIG.16). Example 9: Specific STAT3 Inhibition by GalNAc-Conjugated STAT3 Oligonucleotides [00501] The specificity of the GalNAc-conjugated STAT3 oligonucleotides to inhibit STAT3 rather than a family member (e.g. STAT1) was measured. Specifically, Huh7 cells expressing endogenous STAT1 were treated for 24 hours with 0.05nM, 0.3nM, or 1nM of a GalNAc-conjugated STAT3 oligonucleotide (STAT3-721, STAT3-1286, and STAT3-1388) using lipofectamine as transfection agent. The percent (%) remaining mRNA was measured compared to a mock control (PBS; no lipofectamine or siRNA) and UTR (un-transfected; treated with lipofectamine but no siRNA) (Table 13 and FIG.17). STAT3721 and 1286 did not downregulate human STAT1 but STAT31388 did (Table 13). Oligonucleotides did not downregulate STAT1 expression demonstrating a specificity for STAT3 with limited off-target effects for STAT1. ^ Table 13. STAT1 Expression
Figure imgf000168_0001
Example 10: STAT3 Inhibition in Combination with Checkpoint Inhibition Significantly Improves Anti-Tumor Efficacy [00502] To evaluate the performance of GalXC-STAT3-C18 conjugates as single agent or in combination with a checkpoint inhibitor, anti-PD-L1 mAb, Pan02 tumors (2x106 cells) were implanted in 6-8 week old C57BL/6 mice and upon reaching 300-400 mm3 volume mice were subjected to randomization. Mice received either a single dose of GalXC- STAT3-C18-4123 subcutaneously at 25 mg/kg as single agent or in combination with an anti- PD-L1 mAb (anti-mouse PD-L1 mAb (B7-H1), Clone 10F.9G2) at 10 mg/kg (i.p.). Mice were first administered two doses three days apart, and two weeks later were administered two more doses three days apart [(q3dx2)x2]. Control groups were treated with either GalXC- Placebo as single agent or in combination with the anti-PD-L1 mAb as described for the GalXC-STAT3-C18-4123 compound. Two weeks after the last dose, the same dose regimen was repeated. Tumor sizes were measured twice a week throughout the study period. [00503] As shown in FIG.18A, the tumors that received GalXC-Placebo or GalXC- Placebo + mAb treatments, continued to grow to the same extent. However, the group that received GalXC-STAT3 demonstrated anti-tumor efficacy after the first round of treatment, but they continued to grow despite receiving a second dose. The group that received a combination of GalXC-STAT3 and mAb, demonstrated significantly more tumor regression as compared to the single agent treatment. This demonstrates that combination therapy with a checkpoint inhibitor can achieve improved anti-tumor efficacy. [00504] To assess whether larger tumors would still respond to combination treatment, the tumors that initially received placebo treatment (tumors averaging about 1000mm3) were treated on day 59 and day 62 with the combination of GalXC-STAT3 (25 mg/kg) and mAb (10 mg/kg). Rapid regression of larger tumors to the initial size of about 400 mm3 was observed within a week after one cycle of dosing (FIG.18B) suggesting the effect of combination therapy even in large size tumors. Example 11: Correlation Between Treatment With a Combination of GalXC-STAT3 and PD-L1 mAb With Tumor Immune Phenotypes [00505] To ascertain whether the combination efficacy pattern aligns with the tumor immune phenotype, tumor types with different phenotypes were selected for implantation in mice. Selected tumor types included Pan02 (FIG.18A, checkpoint resistant tumors), 4T1 (triple negative breast, checkpoint resistant tumors), MC-38 (Colon Carcinoma, partially checkpoint sensitive tumors) and Hepa1-6 (Hepatocellular Carcinoma, checkpoint sensitive tumors). Pan02 (5e6 cells + matrigel, FIG.18A) MC-38 (5e6 cells) and Hepa1-6 tumors (2e6 cells) were grown in C57BL/6 mice (7-8 weeks old) and 4T1 tumors (7-8 weeks old) were grown in Balb/c mice. When each tumor reached the sufficient tumor volume, they were sorted and subjected to treatment, as described in Example 5.4T1 tumors were treated three times with each dose three days apart (q3dx3), with a combination of subcutaneous GalXC- STAT3-C18-4123 with an anti-PD-L1 mAb or single agents GalXC-Placebo, GalXC- STAT3-C18-4123, or GalXC-Placebo with the mAb, as shown in FIG.19A. Tumor volumes were measured twice a week throughout the study period. MC-38 and Hepa1-6 tumors were treated with a combination of subcutaneous GalXC-STAT3-C18-4123 with an anti-PD-L1 mAb or single agents GalXC-Placebo, GalXC-STAT3-C18-4123, or GalXC-Placebo with the mAb (2 doses at 3 days apart for 2 weeks) as shown in FIGs.19B and 19C. [00506] Combination treatment demonstrated synergistic efficacy in the resistant tumor types where the tumors expected to have very little or no CD8+ T cell infiltration in the TME and a larger population of MDSCs (CD8low MDSChigh) (FIGs.18A and 19A). The combination treatment showed improved efficacy compared to checkpoint alone treatment in partially sensitive tumors where the tumors had slightly higher levels of CD8+ T-cell infiltration and modest levels of MDSCs (CD8med MDSCmed/high) (FIG.19B). Interestingly, the combination treatment led to complete regression of the sensitive tumors (CD8high MDSChigh) (FIG.19C). Tumors with higher levels of CD8+ T cell infiltration and MDSCs, when treated with the combination of GalXC-STAT3-C18-4123 + anti-PD-L1 mAb, were completely eradicated. Example 12: Treatment Mediated Tumor Regression and Generation of Tumor Specific Memory [00507] To evaluate if the combination treatment demonstrating complete regression also led to the generation of memory T-cells in treated mice, tumors that were completely regressed in FIG.19C were re-challenged with Hep1-6 cells (2e6 cells) on the opposite flank of the mice on day 51. As shown in FIG.20, even after the re-challenge, all mice remained tumor-free and survived for the period that they were kept and maintained (~2 months). These data demonstrate strong therapeutic antitumor efficacy of combination treatment leading to long term immunological memory. Example 13: CD8+ T Cell Mediated Combination Efficacy is Also Perforin Dependent [00508] To evaluate if the efficacy mediated by the combination treatment was CD8+ T cell mediated, the efficacy study described in FIG.19A was repeated in immunocompromised nude mice bearing 4T1 tumors. As shown in FIG.19A, there was synergistic efficacy with combination treatment of GalXC-STAT3-C18-4123 plus anti-PD- L1 mAb in tumor bearing immunocompetent mice, but no efficacy observed in nude mice bearing 4T1 tumors (FIG.21), suggesting that there is a key role for CD8+ T cells in mediating anti-tumor efficacy. To confirm that efficacy is mediated by cytotoxic CD8+ T cells, tumor samples from the terminal timepoint of the study in FIG.19A were stained for perforin. A significantly larger population of perforin positive cells in the tumors that received combination treatment, as shown in FIG.22, shows that the T cells involved in mediating efficacy were cytotoxic in nature. Example 14: Effect of Combination Treatment on Spontaneous Tumor Metastasis in a Highly Metastatic Tumor Model [00509] To evaluate whether combination treatment reduces the metastasis in a spontaneous metastatic tumor model, the mice from the terminal time point of the study in FIGs.19A and 21 were sacrificed and lungs were photographed. As shown in FIG.23, lungs from single agent or placebo treatments in Balb/c mice showed tumor metastases throughout the whole organ whereas the mice administered the combination treatment (GalXC-STAT3- C18-4123 + anti-PD-L1 mAb) showed no visible metastases in the lungs of all five mice, suggesting that the treatment not only reduced the local tumor growth as shown in the figure, but also reduced the spontaneous metastases to lung. The same experiment was repeated in nude mice also shown in FIG.23. All the lungs, including those from the mice that received the combination treatment had tumor metastases, further confirming the role of CD8+ T cells in anti-tumor efficacy. Example 15: Treatment Mediated Immune Modulation in Tumors [00510] To understand how the combination treatment of GalXC-STAT3-C18-4123 with an anti-PD-L1 mAb changes the immune profile in tumor, CT26 tumors were implanted in Balb/c mice. These tumors are partially sensitive to checkpoint inhibitors and have the profile similar to MC38 (CD8med MDSC med/high). When the tumors reached a sufficient size, they were treated with GalXC-Placebo, GalXC-STAT3-C18-4123, GalXC-Placebo + anti- PD-L1 mAb, or GalXC-STAT3-C18-4123 + anti-PD-L1 mAb (q3d x 2, 25 mg/kg or 10 mg/kg). Seven days post last dose, tumors were collected, subjected to homogenization, and nanostring analysis was performed (mRNA extracted from paraffin embedded samples and mRNA expression was analyzed via the ncounterRMouse Pancancer IO 360TM Panel (Nanostring Technologies, Seattle, WA). [00511] The analysis showed that the genes that are suppressive in nature (checkpoints, STAT3 mediated genes, suppressive cytokine/chemokines, angiogenesis & matrix remodeling related genes) were reduced and genes that favor T-cell activation (genes that involve in T-cell migration, activation, memory and cytotoxicity) increased after the combination treatment compared to the single agent or GalXC-Placebo, anti-PD-L1 mAb treatments suggesting that the combination treatment is changing the TME from suppressive to a favorable TME for T-cell infiltration (FIG.24). SEQUENCE LISTING
Figure imgf000172_0001
Figure imgf000173_0001
Figure imgf000174_0001
Figure imgf000175_0001
Figure imgf000176_0001
Figure imgf000177_0001
Figure imgf000178_0001
Figure imgf000179_0001
Figure imgf000180_0001
Figure imgf000181_0001
Figure imgf000182_0001
Figure imgf000183_0001
Figure imgf000184_0001
Figure imgf000185_0001
Figure imgf000186_0001
Figure imgf000187_0001
Figure imgf000188_0001
Figure imgf000189_0001
Figure imgf000190_0001
Figure imgf000191_0001
Figure imgf000192_0001
Figure imgf000193_0001
Figure imgf000194_0001
Figure imgf000195_0001
Figure imgf000196_0001
Figure imgf000197_0001
Figure imgf000198_0001
Figure imgf000199_0001
Figure imgf000200_0001
Figure imgf000201_0001
Figure imgf000202_0001
Figure imgf000203_0001
Figure imgf000204_0001
Figure imgf000205_0001
Figure imgf000206_0001
Figure imgf000207_0001
Figure imgf000208_0001
Figure imgf000209_0001
Figure imgf000210_0001
Figure imgf000211_0001
Figure imgf000212_0001
Figure imgf000213_0001
Figure imgf000214_0001
Figure imgf000215_0001
Figure imgf000216_0001
Figure imgf000217_0001
Figure imgf000218_0001
Figure imgf000219_0001
Figure imgf000220_0001
Figure imgf000221_0001
Figure imgf000222_0001
Figure imgf000223_0001
Figure imgf000224_0001
Figure imgf000225_0001
Figure imgf000226_0001
Figure imgf000227_0001
Figure imgf000228_0001
Figure imgf000229_0001
Figure imgf000230_0001
Figure imgf000231_0001
Figure imgf000232_0001
Figure imgf000233_0001
Figure imgf000234_0001
Figure imgf000235_0001
Figure imgf000236_0001
Figure imgf000237_0001
Figure imgf000238_0001
Figure imgf000239_0001
Figure imgf000240_0001
Figure imgf000241_0001
Figure imgf000242_0001
Figure imgf000243_0001
Figure imgf000244_0001
Figure imgf000245_0001
Figure imgf000246_0001
Figure imgf000247_0001
Figure imgf000248_0001
Figure imgf000249_0001
Figure imgf000250_0001
Figure imgf000251_0001
Figure imgf000252_0001
Figure imgf000253_0001
Figure imgf000254_0001
Figure imgf000255_0001
Figure imgf000256_0001
Figure imgf000257_0001
Figure imgf000258_0001
Figure imgf000259_0001
Figure imgf000260_0001
Figure imgf000261_0001

Claims

Claims 1. A method of treating cancer in a subject that has received or is receiving a PD-L1 inhibitor, the method comprising administering an RNAi oligonucleotide comprising an antisense strand of 15 to 30 nucleotides in length and a sense strand of 15 to 40 nucleotides in length, wherein the antisense and sense strands form a duplex region, wherein the antisense strand comprises a region of complementarity to a STAT3 mRNA target sequence, and wherein the region of complementarity is at least 15 contiguous nucleotides in length, thereby treating cancer in the subject.
2. A method of treating cancer in a subject that has received or is receiving an RNAi oligonucleotide, the method comprising administering a PD-L1 inhibitor, wherein the oligonucleotide comprises an antisense strand of 15 to 30 nucleotides in length and a sense strand of 15 to 40 nucleotides in length, wherein the antisense and sense strands form a duplex region, wherein the antisense strand comprises a region of complementarity to a STAT3 mRNA target sequence, and wherein the region of complementarity is at least 15 contiguous nucleotides in length, thereby treating cancer in the subject.
3. A method of treating a disease, disorder or condition associated with activated STAT3 expression, comprising administering to a subject in need thereof an RNAi oligonucleotide, wherein the subject has received or is receiving a PD-L1 inhibitor, wherein the oligonucleotide comprises an antisense strand of 15 to 30 nucleotides in length and a sense strand of 15 to 40 nucleotides in length, wherein the antisense and sense strands form a duplex region, wherein the antisense strand comprises a region of complementarity to a STAT3 mRNA target sequence, and wherein the region of complementarity is at least 15 contiguous nucleotides in length, thereby treating cancer in the subject.
4. The method of claim 3, wherein the disease, disorder, or condition associated with activated STAT3 expression is a cancer.
5. The method of any one of claims 1-2 and 4, wherein the cancer is selected from carcinoma, sarcoma, melanoma, lymphoma, and leukemia, prostate cancer, breast cancer, hepatocellular carcinoma (HCC), colorectal cancer, pancreatic cancer and glioblastoma.
6. The method of any one of claims 1-2 and 4-5, wherein the cancer comprises an immunosuppressive tumor microenvironment.
7. The method of any one of claims 1-2 and 4-5, wherein the cancer comprises an inflamed tumor microenvironment.
8. The method of claim 7, wherein the inflamed tumor microenvironment comprises infiltrating T cells.
9. The method of any one of claims 1-8, wherein the PD-L1 inhibitor is an antibody.
10. The method of claim 9, wherein the antibody is an anti-PD-L1 antibody.
11. The method of claim 10, wherein the anti-PDL1 antibody is selected from FAZ053, atezolizumab, avelumab, durvalumab, envafolimab, and BMS-936559.
12. The method of claim 9, wherein the antibody is an anti-PD-1 antibody.
13. The method of claim 12, wherein the anti-PD-1 antibody is selected from nivolumab, pembrolizumab, and cemiplimab.
14. The method of any one of claims 1-8, wherein the PD-L1 inhibitor is a small molecule inhibitor.
15. The method of any one of claims 1-8, wherein the PD-L1 inhibitor is a peptide.
16. The method of any one of claims 1-8, wherein the PD-L1 inhibitor is a nucleic acid molecule.
17. The method of claim 16, wherein the nucleic acid molecule is selected from an antisense oligonucleotide, an siRNA, or an miRNA.
18. The method of any one of claims 1-17, wherein the STAT3 mRNA target sequence comprises any one of SEQ ID NOs: 89-280.
19. The method of any one of claims 1-18, wherein the region of complementarity is fully complementary to the STAT3 mRNA target sequence.
20. The method of any one of claims 1-18, wherein the region of complementarity comprises no more than 4 mismatches to the STAT3 mRNA target sequence.
21. The method of any one of claims 1-20, wherein the antisense strand is 19 to 27 nucleotides in length.
22. The method of any one of claims 1-21, wherein the antisense strand is 21 to 27 nucleotides in length, optionally wherein the antisense strand is 22 nucleotides in length.
23. The method of any one of claims 1-22, wherein the sense strand is 19 to 40 nucleotides in length, optionally wherein the sense strand is 36 nucleotides in length.
24. The method of any one of claims 1-23, wherein the duplex region is at least 19 nucleotides in length.
25. The method of any one of claims 1-24, wherein the duplex region is at least 20 nucleotides in length, optionally wherein the duplex region is 21 nucleotides in length.
26. The method of any one of claims 1-25, wherein the region of complementarity to STAT3 is at least 19 contiguous nucleotides in length.
27. The method of any one of claims 1-26, wherein the region of complementarity to STAT3 is at least 21 contiguous nucleotides in length.
28. The method of any one of claims 1-27, wherein the sense strand comprises at its 3′ end a stem-loop set forth as: S1-L-S2, wherein S1 is complementary to S2, and wherein L forms a loop between S1 and S2 of 3 to 5 nucleotides in length.
29. The method of claim 28, wherein L is a tetraloop, optionally wherein L is 4 nucleotides in length.
30. The method of claim 28 or 29, wherein L comprises a sequence set forth as GAAA.
31. The method of any one of claims 1-30, wherein the antisense strand comprises a 3’ overhang sequence of one or more nucleotides in length, optionally wherein the 3’ overhang sequence is 2 nucleotides in length, optionally wherein the 3’ overhang sequence is GG.
32. The method of any one of claims 1-31, wherein the oligonucleotide comprises at least one modified nucleotide.
33. The method of claim 32, wherein the modified nucleotide comprises a 2′- modification.
34. The method of claim 33, wherein the 2′-modification is a modification selected from 2′-aminoethyl, 2′-fluoro, 2′-O-methyl, 2′-O-methoxyethyl, and 2′-deoxy-2′-fluoro-β- d-arabinonucleic acid.
35. The method of any one of claims 33-34, wherein about 10-15%, 10%, 11%, 12%, 13%, 14% or 15% of the nucleotides of the sense strand comprise a 2’-fluoro modification.
36. The method of any one of claims 33-35, wherein about 25-35%, 25%, 26%, 27%, 28%, 29%, 30%, 31%, 32%, 33%, 34% or 35% of the nucleotides of the antisense strand comprise a 2’-fluoro modification.
37. The method of any one of claims 33-36, wherein about 25-35%, 25%, 26%, 27%, 28%, 29%, 30%, 31%, 32%, 33%, 34% or 35% of the nucleotides of the oligonucleotide comprise a 2’-fluoro modification.
38. The method of any one of claims 33-37, wherein the sense strand comprises 36 nucleotides with positions 1-36 from 5’ to 3 ’, wherein positions 8-11 comprise a 2’- fluoro modification.
39. The method of any one of claims 33-38, wherein the antisense strand comprises 22 nucleotides with positions 1-22 from 3’ to 5’, and wherein positions 2, 3, 4, 5, 7, 10 and 14 comprise a 2’-fluoro modification.
40. The method of any one of claims 33-39, wherein the remaining nucleotides comprise a 2’-O-methyl modification.
41. The method of any one of claims 33-40, wherein all of the nucleotides of the oligonucleotide are modified.
42. The method of any one of claims 1-41, wherein the oligonucleotide comprises at least one modified internucleotide linkage.
43. The method of claim 42, wherein the at least one modified internucleotide linkage is a phosphorothioate linkage.
44. The method of any one of claims 1-43, wherein the 4′-carbon of the sugar of the 5′-nucleotide of the antisense strand comprises a phosphate analog.
45. The method of claim 44, wherein the phosphate analog is oxymethylphosphonate, vinylphosphonate or malonylphosphonate.
46. The method of claim 44, wherein the phosphate analog is oxymethylphosphonate.
47. The method of any one of claims 1-46, wherein at least one nucleotide of the oligonucleotide is conjugated to one or more targeting ligands.
48. The method of claim 47, wherein the nucleotide is conjugated to more than one targeting ligands, wherein the targeting ligands are the same or are different.
49. The method of claim 47 or 48, wherein the one or more targeting ligands is selected from carbohydrate, amino sugar, cholesterol, polypeptide, or lipid.
50. The method of claim 47 or 48, wherein the one or more targeting ligands is a saturated or unsaturated fatty acid moiety.
51. The method of claim 47 or 48, wherein the targeting ligand is a saturated fatty acid moiety that ranges in size from C10 to C24 long.
52. The method of claim 51, wherein the targeting ligand is a C16 saturated fatty acid moiety.
53. The method of claim 51, wherein the targeting ligand is a C18 saturated fatty acid moiety.
54. The method of claim 51, wherein the targeting ligand is a C22 saturated fatty acid moiety.
55. The method of claim 47 or 48, wherein the targeting ligand comprises a N- acetylgalactosamine (GalNAc) moiety.
56. The method of claim 55, wherein the GalNAc moiety is a monovalent GalNAc moiety, a bivalent GalNAc moiety, a trivalent GalNAc moiety or a tetravalent GalNAc moiety.
57. The method of any one of claims 28-48, wherein up to 4 nucleotides of L of the stem-loop are each conjugated to a monovalent GalNAc moiety.
58. The method of any one of claims 1-57, wherein the sense strand comprises a sequence as set forth in SEQ ID NOs: 857-946.
59. The method of any one of claims 1-58, wherein the antisense strand comprises a sequence as set for in SEQ ID NOs: 947-1036.
60. The method of any one of claims 1-57, wherein the sense strand and antisense strand comprise nucleotide sequences selected from the group consisting of: (a) SEQ ID NOs: 9 and 10, respectively; (b) SEQ ID NOs: 37 and 38, respectively; (c) SEQ ID NOs: 65 and 66, respectively; and (d) SEQ ID NOs: 69 and 70, respectively.
61. The method of any one of claims 1-57, wherein the sense strand and antisense strand comprise nucleotide sequences selected from the group consisting of: (a) SEQ ID NOs: 861 and 951, respectively; (b) SEQ ID NOs: 857 and 947, respectively; (c) SEQ ID NOs: 858 and 948, respectively; (d) SEQ ID NOs: 859 and 949, respectively; (e) SEQ ID NOs: 860 and 950, respectively; (f) SEQ ID NOs: 862 and 952, respectively; (g) SEQ ID NOs: 863 and 953, respectively; (h) SEQ ID NOs: 864 and 954, respectively; (i) SEQ ID NOs: 865 and 955, respectively; (j) SEQ ID NOs: 866 and 956, respectively; (k) SEQ ID NOs: 867 and 957, respectively; (l) SEQ ID NOs: 868 and 958, respectively; (m) SEQ ID NOs: 869 and 959, respectively; (n) SEQ ID NOs: 870 and 960, respectively; (o) SEQ ID NOs: 871 and 961, respectively; (p) SEQ ID NOs: 872 and 962, respectively; (q) SEQ ID NOs: 873 and 963, respectively; (r) SEQ ID NOs: 874 and 964, respectively; (s) SEQ ID NOs: 875 and 965, respectively; (t) SEQ ID NOs: 876 and 966, respectively; (u) SEQ ID NOs: 877 and 967, respectively; (v) SEQ ID NOs: 878 and 968, respectively; (w) SEQ ID NOs: 879 and 969, respectively; (x) SEQ ID NOs: 880 and 970, respectively; (y) SEQ ID NOs: 881and 971, respectively; (z) SEQ ID NOs: 882 and 972, respectively; (aa) SEQ ID NOs: 883 and 973, respectively; (bb) SEQ ID NOs: 884 and 974, respectively; (cc) SEQ ID NOs: 885 and 975, respectively; (dd) SEQ ID NOs: 886 and 976, respectively; (ee) SEQ ID NOs: 887 and 977, respectively; (ff) SEQ ID NOs: 888 and 978, respectively; (gg) SEQ ID NOs: 940 and 1030, respectively; (hh) SEQ ID NOs: 896 and 986, respectively; and (ii) SEQ ID NOs: 920 and 1010, respectively.
62. The method of any one of claims 1-57, wherein the sense strand comprises the nucleotide sequence of SEQ ID NO: 862 and the antisense strand comprises the nucleotide sequence of SEQ ID NO: 952.
63. The method of any one of claims 1-57, wherein the sense strand comprises the nucleotide sequence of SEQ ID NO: 875 and the antisense strand comprises the nucleotide sequence of SEQ ID NO: 965.
64. The method of any one of claims 1-57, wherein the sense strand comprises the nucleotide sequence of SEQ ID NO: 876 and the antisense strand comprises the nucleotide sequence of SEQ ID NO: 966.
65. The method of any one of claims 1-57, wherein the sense strand comprises the nucleotide sequence of SEQ ID NO: 920 and the antisense strand comprises the nucleotide sequence of SEQ ID NO: 1010.
66. The method of any one of claims 1-57, wherein the sense strand and antisense strand comprise nucleotide sequences selected from the group consisting of: (a) SEQ ID NOs: 901 and 991, respectively; (b) SEQ ID NOs: 910 and 1000, respectively; (c) SEQ ID NOs: 899 and 989, respectively; (d) SEQ ID NOs: 896 and 986, respectively; (e) SEQ ID NOs: 892 and 982, respectively; (f) SEQ ID NOs: 890 and 980, respectively; and (g) SEQ ID NOs: 889 and 979, respectively.
67. The method of any one of claims 1-57, wherein the sense strand and antisense strand comprise nucleotide sequences selected from the group consisting of: (a) SEQ ID NOs: 940 and 1030, respectively; (b) SEQ ID NOs: 937 and 1027, respectively; and (c) SEQ ID NOs: 939 and 1029, respectively.
68. The method of any one of claims 1-57, wherein the sense strand and antisense strand comprise nucleotide sequences selected from the group consisting of: (a) SEQ ID NOs: 915 and 1005, respectively; (b) SEQ ID NOs: 924 and 1014, respectively; (c) SEQ ID NOs: 913 and 1003, respectively; and (d) SEQ ID NOs: 920 and 1010, respectively.
69. The method of any one of claims 1-57, wherein the sense strand comprises a nucleotide sequence of any one of SEQ ID NOs: 11, 39, 67 and 71.
70. The method of any one of claims 1-57, wherein the antisense strand comprises a nucleotide sequence of any one of SEQ ID NOs: 12, 40, 68 and 72.
71. The method of any one of claims 1-57, wherein the sense strand and antisense strand comprise nucleotide sequences selected from the group consisting of: (a) SEQ ID NOs: 11 and 12, respectively; (b) SEQ ID NOs: 39 and 40, respectively; (c) SEQ ID NOs: 67 and 68, respectively; and (d) SEQ ID NOs: 71 and 72, respectively.
72. The method of any one of claims 1-57, wherein the sense strand comprises a nucleotide sequence selected from SEQ ID NOs: 1042, 1055, 1056, and 1100.
73. The method of any one of claims 1-57 and 72, wherein the antisense strand comprises a nucleotide sequence selected from SEQ ID NOs: 1132, 1145, 1146, and 1190.
74. The method of any one of claims 1-57, wherein the sense strand and antisense strand comprise nucleotide sequences selected from the group consisting of: (a) SEQ ID NOs: 1041 and 1131, respectively; (b) SEQ ID NOs: 1037 and 1127, respectively; (c) SEQ ID NOs: 1038 and 1128, respectively; (d) SEQ ID NOs: 1039 and 1129, respectively; (e) SEQ ID NOs: 1040 and 1130, respectively; (f) SEQ ID NOs: 1042 and 1132, respectively; (g) SEQ ID NOs: 1043 and 1133, respectively; (h) SEQ ID NOs: 1044 and 1134, respectively; (i) SEQ ID NOs: 1045 and 1135, respectively; (j) SEQ ID NOs: 1046 and 1136, respectively; (k) SEQ ID NOs: 1047 and 1137, respectively; (l) SEQ ID NOs: 1048 and 1138, respectively; (m) SEQ ID NOs: 1049 and 1139, respectively; (n) SEQ ID NOs: 1050 and 1140, respectively; (o) SEQ ID NOs: 1051 and 1141, respectively; (p) SEQ ID NOs: 1052 and 1142, respectively; (q) SEQ ID NOs: 1053 and 1143, respectively; (r) SEQ ID NOs: 1054 and 1144, respectively; (s) SEQ ID NOs: 1055 and 1145, respectively; (t) SEQ ID NOs: 1056 and 1146, respectively; (u) SEQ ID NOs: 1057 and 1147, respectively; (v) SEQ ID NOs: 1058 and 1148, respectively; (w) SEQ ID NOs: 1059 and 1149, respectively; (x) SEQ ID NOs: 1060 and 1150, respectively; (y) SEQ ID NOs: 1061 and 1151, respectively; (z) SEQ ID NOs: 1062 and 1152, respectively; (aa) SEQ ID NOs: 1063 and 1153, respectively; (bb) SEQ ID NOs: 1064 and 1154, respectively; (cc) SEQ ID NOs: 1065 and 1155, respectively; (dd) SEQ ID NOs: 1066 and 1156, respectively; (ee) SEQ ID NOs: 1067 and 1157, respectively; (ff) SEQ ID NOs: 1068 and 1158, respectively; (gg) SEQ ID NOs: 1120 and 1210, respectively; (hh) SEQ ID NOs: 1076 and 1166, respectively; and (ii) SEQ ID NOs: 1100 and 1190, respectively.
75. The method of any one of claims 1-57, wherein the sense strand comprises the nucleotide sequence of SEQ ID NO: 1042 and the antisense strand comprises the nucleotide sequence of SEQ ID NO: 1132.
76. The method of any one of claims 1-57, wherein the sense strand comprises the nucleotide sequence of SEQ ID NO: 1055 and the antisense strand comprises the nucleotide sequence of SEQ ID NO: 1145.
77. The method of any one of claims 1-57, wherein the sense strand comprises the nucleotide sequence of SEQ ID NO: 1056 and the antisense strand comprises the nucleotide sequence of SEQ ID NO: 1146.
78. The method of any one of claims 1-57, wherein the sense strand comprises the nucleotide sequence of SEQ ID NO: 1100 and the antisense strand comprises the nucleotide sequence of SEQ ID NO: 1190.
79. The method of any one of claims 1-57, wherein the sense strand and antisense strand comprise nucleotide sequences selected from the group consisting of: (a) SEQ ID NOs: 1081 and 1171, respectively; (b) SEQ ID NOs: 1090 and 1180, respectively; (c) SEQ ID NOs: 1079 and 1169, respectively; (d) SEQ ID NOs: 1076 and 1166, respectively; (e) SEQ ID NOs: 1072 and 1162, respectively; (f) SEQ ID NOs: 1070 and 1160, respectively; and (g) SEQ ID NOs: 1069 and 1159, respectively.
80. The method of any one of claims 1-57, wherein the sense strand and antisense strand comprise nucleotide sequences selected from the group consisting of: (a) SEQ ID NOs: 1120 and 1210, respectively; (b) SEQ ID NOs: 1117 and 1207, respectively; and (c) SEQ ID NOs: 1119 and 1209, respectively.
81. The method of any one of claims 1-57, wherein the sense strand and antisense strand comprise nucleotide sequences selected from the group consisting of: (a) SEQ ID NOs: 1095 and 1185, respectively; (b) SEQ ID NOs: 1104 and 1194, respectively; (c) SEQ ID NOs: 1093 and 1183, respectively; and (d) SEQ ID NOs: 1100 and 1190, respectively.
82. A kit comprising an RNAi oligonucleotide, an optional pharmaceutically acceptable carrier, and a package insert comprising instructions for administering the oligonucleotide to a subject in need thereof that has received or is receiving a PD-L1 inhibitor, wherein the oligonucleotide comprises an antisense strand of 15 to 30 nucleotides in length and a sense strand of 15 to 40 nucleotides in length, wherein the antisense and sense strands form a duplex region, wherein the antisense strand comprises a region of complementarity to a STAT3 mRNA target sequence, and wherein the region of complementarity is at least 15 contiguous nucleotides in length.
83. A kit comprising a PD-L1 inhibitor, an optional pharmaceutically acceptable carrier, and a package insert comprising instructions for administering the antibody to a subject in need thereof that has received or is receiving an RNAi oligonucleotide comprising an antisense strand of 15 to 30 nucleotides in length and a sense strand of 15 to 40 nucleotides in length, wherein the antisense and sense strands form a duplex region, wherein the antisense strand comprises a region of complementarity to a STAT3 mRNA target sequence, and wherein the region of complementarity is at least 15 contiguous nucleotides in length.
84. The kit of 82 or 83, wherein the subject has a disease, disorder, or condition associated with activated STAT3 expression.
85. The kit of any one of claims 82-84, wherein the subject has cancer.
86. The kit of claim 85, wherein the cancer is selected from carcinoma, sarcoma, melanoma, lymphoma, and leukemia, prostate cancer, breast cancer, hepatocellular carcinoma (HCC), colorectal cancer, pancreatic cancer and glioblastoma.
87. The kit of any one of claims 82-86, wherein the cancer comprises an immunosuppressive tumor microenvironment.
88. The kit of any one of claims 82-86, wherein the cancer comprises an inflamed tumor microenvironment.
89. The kit of claim 88, wherein the inflamed tumor microenvironment comprises infiltrating T cells.
90. The kit of any one of claims 82-89, wherein the PD-L1 inhibitor is an antibody.
91. The kit of claim 90, wherein the antibody is an anti-PD-L1 antibody.
92. The kit of claim 91, wherein the anti-PDL1 antibody is selected from FAZ053, atezolizumab, avelumab, durvalumab, envafolimab, and BMS-936559.
93. The kit of claim 90, wherein the antibody is an anti-PD-1 antibody.
94. The kit of claim 93, wherein the anti-PD-1 antibody is selected from nivolumab, pembrolizumab, and cemiplimab.
95. The kit of any one of claims 82-89, wherein the PD-L1 inhibitor is a small molecule inhibitor.
96. The kit of any one of claims 82-89, wherein the PD-L1 inhibitor is a peptide.
97. The kit of any one of claims 82-89, wherein the PD-L1 inhibitor is a nucleic acid molecule.
98. The kit of any one of claims 82-89, wherein the nucleic acid molecule is selected from an antisense oligonucleotide, an siRNA, or an miRNA.
99. The kit of any one of claims 82-94, wherein the STAT3 mRNA target sequence comprises any one of SEQ ID NOs: 89-280.
100. The kit of any one of claims 82-99, wherein the region of complementarity is fully complementary to the STAT3 mRNA target sequence.
101. The kit of any one of claims 82-99, wherein the region of complementarity comprises no more than 4 mismatches to the STAT3 mRNA target sequence.
102. The kit of any one of claims 82-101, wherein the antisense strand is 19 to 27 nucleotides in length.
103. The kit of any one of claims 82-102, wherein the antisense strand is 21 to 27 nucleotides in length, optionally wherein the antisense strand is 22 nucleotides in length.
104. The kit of any one of claims 82-103, wherein the sense strand is 19 to 40 nucleotides in length, optionally wherein the sense strand is 36 nucleotides in length.
105. The kit of any one of claims 82-104, wherein the duplex region is at least 19 nucleotides in length.
106. The kit of any one of claims 82-105, wherein the duplex region is at least 20 nucleotides in length, optionally wherein the duplex region is 21 nucleotides in length.
107. The kit of any one of claims 82-106, wherein the region of complementarity to STAT3 is at least 19 contiguous nucleotides in length.
108. The kit of any one of claims 82-107, wherein the region of complementarity to STAT3 is at least 21 contiguous nucleotides in length.
109. The kit of any one of claims 82-108, wherein the sense strand comprises at its 3′ end a stem-loop set forth as: S1-L-S2, wherein S1 is complementary to S2, and wherein L forms a loop between S1 and S2 of 3 to 5 nucleotides in length.
110. The kit of claim 109, wherein L is a tetraloop, optionally wherein L is 4 nucleotides in length.
111. The kit of claim 109 or 110, wherein L comprises a sequence set forth as GAAA.
112. The kit of any one of claims 82-111, wherein the antisense strand comprises a 3’ overhang sequence of one or more nucleotides in length, optionally wherein the 3’ overhang sequence is 2 nucleotides in length, optionally wherein the 3’ overhang sequence is GG.
113. The kit of any one of claims 82-112, wherein the oligonucleotide comprises at least one modified nucleotide.
114. The kit of claim 113, wherein the modified nucleotide comprises a 2′- modification.
115. The kit of claim 114, wherein the 2′-modification is a modification selected from 2′-aminoethyl, 2′-fluoro, 2′-O-methyl, 2′-O-methoxyethyl, and 2′-deoxy-2′-fluoro-β-d- arabinonucleic acid.
116. The kit of any one of claims 114-115, wherein about 10-15%, 10%, 11%, 12%, 13%, 14% or 15% of the nucleotides of the sense strand comprise a 2’-fluoro modification.
117. The kit of any one of claims 114-116, wherein about 25-35%, 25%, 26%, 27%, 28%, 29%, 30%, 31%, 32%, 33%, 34% or 35% of the nucleotides of the antisense strand comprise a 2’-fluoro modification.
118. The kit of any one of claims 114-117, wherein about 25-35%, 25%, 26%, 27%, 28%, 29%, 30%, 31%, 32%, 33%, 34% or 35% of the nucleotides of the oligonucleotide comprise a 2’-fluoro modification.
119. The kit of any one of claims 114-118, wherein the sense strand comprises 36 nucleotides with positions 1-36 from 5’ to 3 ’, wherein positions 8-11 comprise a 2’- fluoro modification.
120. The kit of any one of claims 114-119, wherein the antisense strand comprises 22 nucleotides with positions 1-22 from 3’ to 5’, and wherein positions 2, 3, 4, 5, 7, 10 and 14 comprise a 2’-fluoro modification.
121. The kit of any one of claims 114-120, wherein the remaining nucleotides comprise a 2’-O-methyl modification.
122. The kit of any one of claims 114-121, wherein all of the nucleotides of the oligonucleotide are modified.
123. The kit of any one of claims 82-122, wherein the oligonucleotide comprises at least one modified internucleotide linkage.
124. The kit of claim 123, wherein the at least one modified internucleotide linkage is a phosphorothioate linkage.
125. The kit of any one of claims 82-124, wherein the 4′-carbon of the sugar of the 5′- nucleotide of the antisense strand comprises a phosphate analog.
126. The kit of claim 125, wherein the phosphate analog is oxymethylphosphonate, vinylphosphonate or malonylphosphonate.
127. The kit of any one of claims 82-126, wherein at least one nucleotide of the oligonucleotide is conjugated to one or more targeting ligands.
128. The kit of claim 127, wherein the nucleotide is conjugated to more than one targeting ligands, wherein the targeting ligands are the same or are different.
129. The kit of claim 127 or 128, wherein the one or more targeting ligands is selected from carbohydrate, amino sugar, cholesterol, polypeptide, or lipid.
130. The kit of claim 127 or 128, wherein the one or more targeting ligands is a saturated or unsaturated fatty acid moiety.
131. The kit of claim 127 or 128, wherein the targeting ligand is a saturated fatty acid moiety that ranges in size from C10 to C24 long.
132. The kit of claim 131, wherein the targeting ligand is a C16 saturated fatty acid moiety.
133. The kit of claim 131, wherein the targeting ligand is a C18 saturated fatty acid moiety.
134. The kit of claim 131, wherein the targeting ligand is a C22 saturated fatty acid moiety.
135. The kit of claim 127 or 128, wherein the targeting ligand comprises a N- acetylgalactosamine (GalNAc) moiety.
136. The kit of claim 135, wherein the GalNAc moiety is a monovalent GalNAc moiety, a bivalent GalNAc moiety, a trivalent GalNAc moiety or a tetravalent GalNAc moiety.
137. The kit of any one of claims 109-129, wherein up to 4 nucleotides of L of the stem-loop are each conjugated to a monovalent GalNAc moiety.
138. The kit of any one of claims 82-137, wherein the sense strand comprises a sequence as set forth in SEQ ID NOs: 857-946.
139. The kit of any one of claims 82-138, wherein the antisense strand comprises a sequence as set for in SEQ ID NOs: 947-1036.
140. The kit of any one of claims 82-137, wherein the sense strand and antisense strand comprise nucleotide sequences selected from the group consisting of: (a) SEQ ID NOs: 9 and 10, respectively; (b) SEQ ID NOs: 37 and 38, respectively; (c) SEQ ID NOs: 65 and 66, respectively; and (d) SEQ ID NOs: 69 and 70, respectively.
141. The kit of any one of claims 82-137, wherein the sense strand and antisense strand comprise nucleotide sequences selected from the group consisting of: (a) SEQ ID NOs: 861 and 951, respectively; (b) SEQ ID NOs: 857 and 947, respectively; (c) SEQ ID NOs: 858 and 948, respectively; (d) SEQ ID NOs: 859 and 949, respectively; (e) SEQ ID NOs: 860 and 950, respectively; (f) SEQ ID NOs: 862 and 952, respectively; (g) SEQ ID NOs: 863 and 953, respectively; (h) SEQ ID NOs: 864 and 954, respectively; (i) SEQ ID NOs: 865 and 955, respectively; (j) SEQ ID NOs: 866 and 956, respectively; (k) SEQ ID NOs: 867 and 957, respectively; (l) SEQ ID NOs: 868 and 958, respectively; (m) SEQ ID NOs: 869 and 959, respectively; (n) SEQ ID NOs: 870 and 960, respectively; (o) SEQ ID NOs: 871 and 961, respectively; (p) SEQ ID NOs: 872 and 962, respectively; (q) SEQ ID NOs: 873 and 963, respectively; (r) SEQ ID NOs: 874 and 964, respectively; (s) SEQ ID NOs: 875 and 965, respectively; (t) SEQ ID NOs: 876 and 966, respectively; (u) SEQ ID NOs: 877 and 967, respectively; (v) SEQ ID NOs: 878 and 968, respectively; (w) SEQ ID NOs: 879 and 969, respectively; (x) SEQ ID NOs: 880 and 970, respectively; (y) SEQ ID NOs: 881and 971, respectively; (z) SEQ ID NOs: 882 and 972, respectively; (aa) SEQ ID NOs: 883 and 973, respectively; (bb) SEQ ID NOs: 884 and 974, respectively; (cc) SEQ ID NOs: 885 and 975, respectively; (dd) SEQ ID NOs: 886 and 976, respectively; (ee) SEQ ID NOs: 887 and 977, respectively; (ff) SEQ ID NOs: 888 and 978, respectively; (gg) SEQ ID NOs: 940 and 1030, respectively; (hh) SEQ ID NOs: 896 and 986, respectively; and (ii) SEQ ID NOs: 920 and 1010, respectively.
142. The kit of any one of claims 82-137, wherein the sense strand comprises the nucleotide sequence of SEQ ID NO: 862 and the antisense strand comprises the nucleotide sequence of SEQ ID NO: 952.
143. The kit of any one of claims 82-137, wherein the sense strand comprises the nucleotide sequence of SEQ ID NO: 875 and the antisense strand comprises the nucleotide sequence of SEQ ID NO: 965.
144. The kit of any one of claims 82-137, wherein the sense strand comprises the nucleotide sequence of SEQ ID NO: 876 and the antisense strand comprises the nucleotide sequence of SEQ ID NO: 966.
145. The kit of any one of claims 82-137, wherein the sense strand comprises the nucleotide sequence of SEQ ID NO: 920 and the antisense strand comprises the nucleotide sequence of SEQ ID NO: 1010.
146. The kit of any one of claims 82-137, wherein the sense strand and antisense strand comprise nucleotide sequences selected from the group consisting of: (a) SEQ ID NOs: 901 and 991, respectively; (b) SEQ ID NOs: 910 and 1000, respectively; (c) SEQ ID NOs: 899 and 989, respectively; (d) SEQ ID NOs: 896 and 986, respectively; (e) SEQ ID NOs: 892 and 982, respectively; (f) SEQ ID NOs: 890 and 980, respectively; and (g) SEQ ID NOs: 889 and 979, respectively.
147. The kit of any one of claims 82-137, wherein the sense strand and antisense strand comprise nucleotide sequences selected from the group consisting of: (a) SEQ ID NOs: 940 and 1030, respectively; (b) SEQ ID NOs: 937 and 1027, respectively; and (c) SEQ ID NOs: 939 and 1029, respectively.
148. The kit of any one of claims 82-137, wherein the sense strand and antisense strand comprise nucleotide sequences selected from the group consisting of: (a) SEQ ID NOs: 915 and 1005, respectively; (b) SEQ ID NOs: 924 and 1014, respectively; (c) SEQ ID NOs: 913 and 1003, respectively; and (d) SEQ ID NOs: 920 and 1010, respectively.
149. The kit of any one of claims 82-137, wherein the sense strand comprises a nucleotide sequence of any one of SEQ ID NOs: 11, 39, 67 and 71.
150. The kit of any one of claims 82-137, wherein the antisense strand comprises a nucleotide sequence of any one of SEQ ID NOs: 12, 40, 68 and 72.
151. The kit of any one of claims 82-137, wherein the sense strand and antisense strand comprise nucleotide sequences selected from the group consisting of: (a) SEQ ID NOs: 11 and 12, respectively; (b) SEQ ID NOs: 39 and 40, respectively; (c) SEQ ID NOs: 67 and 68, respectively; and (d) SEQ ID NOs: 71 and 72, respectively.
152. The kit of any one of claims 82-137, wherein the sense strand comprises a nucleotide sequence selected from SEQ ID NOs: 1042, 1055, 1056, and 1100.
153. The kit of any one of claims 82-137 and 152, wherein the antisense strand comprises a nucleotide sequence selected from SEQ ID NOs: 1132, 1145, 1146, and 11
154. The kit of any one of claims 82-137, wherein the sense strand and antisense strand comprise nucleotide sequences selected from the group consisting of: (a) SEQ ID NOs: 1041 and 1131, respectively; (b) SEQ ID NOs: 1037 and 1127, respectively; (c) SEQ ID NOs: 1038 and 1128, respectively; (d) SEQ ID NOs: 1039 and 1129, respectively; (e) SEQ ID NOs: 1040 and 1130, respectively; (f) SEQ ID NOs: 1042 and 1132, respectively; (g) SEQ ID NOs: 1043 and 1133, respectively; (h) SEQ ID NOs: 1044 and 1134, respectively; (i) SEQ ID NOs: 1045 and 1135, respectively; (j) SEQ ID NOs: 1046 and 1136, respectively; (k) SEQ ID NOs: 1047 and 1137, respectively; (l) SEQ ID NOs: 1048 and 1138, respectively; (m) SEQ ID NOs: 1049 and 1139, respectively; (n) SEQ ID NOs: 1050 and 1140, respectively; (o) SEQ ID NOs: 1051 and 1141, respectively; (p) SEQ ID NOs: 1052 and 1142, respectively; (q) SEQ ID NOs: 1053 and 1143, respectively; (r) SEQ ID NOs: 1054 and 1144, respectively; (s) SEQ ID NOs: 1055 and 1145, respectively; (t) SEQ ID NOs: 1056 and 1146, respectively; (u) SEQ ID NOs: 1057 and 1147, respectively; (v) SEQ ID NOs: 1058 and 1148, respectively; (w) SEQ ID NOs: 1059 and 1149, respectively; (x) SEQ ID NOs: 1060 and 1150, respectively; (y) SEQ ID NOs: 1061 and 1151, respectively; (z) SEQ ID NOs: 1062 and 1152, respectively; (aa) SEQ ID NOs: 1063 and 1153, respectively; (bb) SEQ ID NOs: 1064 and 1154, respectively; (cc) SEQ ID NOs: 1065 and 1155, respectively; (dd) SEQ ID NOs: 1066 and 1156, respectively; (ee) SEQ ID NOs: 1067 and 1157, respectively; (ff) SEQ ID NOs: 1068 and 1158, respectively; (gg) SEQ ID NOs: 1120 and 1210, respectively; (hh) SEQ ID NOs: 1076 and 1166, respectively; and (ii) SEQ ID NOs: 1100 and 1190, respectively.
155. The kit of any one of claims 82-137, wherein the sense strand comprises the nucleotide sequence of SEQ ID NO: 1042 and the antisense strand comprises the nucleotide sequence of SEQ ID NO: 1132.
156. The kit of any one of claims 82-137, wherein the sense strand comprises the nucleotide sequence of SEQ ID NO: 1055 and the antisense strand comprises the nucleotide sequence of SEQ ID NO: 1145.
157. The kit of any one of claims 82-137, wherein the sense strand comprises the nucleotide sequence of SEQ ID NO: 1056 and the antisense strand comprises the nucleotide sequence of SEQ ID NO: 1146.
158. The kit of any one of claims 82-137, wherein the sense strand comprises the nucleotide sequence of SEQ ID NO: 1100 and the antisense strand comprises the nucleotide sequence of SEQ ID NO: 1190.
159. The kit of any one of claims 82-137, wherein the sense strand and antisense strand comprise nucleotide sequences selected from the group consisting of: (a) SEQ ID NOs: 1081 and 1171, respectively; (b) SEQ ID NOs: 1090 and 1180, respectively; (c) SEQ ID NOs: 1079 and 1169, respectively; (d) SEQ ID NOs: 1076 and 1166, respectively; (e) SEQ ID NOs: 1072 and 1162, respectively; (f) SEQ ID NOs: 1070 and 1160, respectively; and (g) SEQ ID NOs: 1069 and 1159, respectively.
160. The kit of any one of claims 82-137, wherein the sense strand and antisense strand comprise nucleotide sequences selected from the group consisting of: (a) SEQ ID NOs: 1120 and 1210, respectively; (b) SEQ ID NOs: 1117 and 1207, respectively; and (c) SEQ ID NOs: 1119 and 1209, respectively.
161. The kit of any one of claims 82-137, wherein the sense strand and antisense strand comprise nucleotide sequences selected from the group consisting of: (a) SEQ ID NOs: 1095 and 1185, respectively; (b) SEQ ID NOs: 1104 and 1194, respectively; (c) SEQ ID NOs: 1093 and 1183, respectively; and (d) SEQ ID NOs: 1100 and 1190, respectively.
PCT/US2023/064381 2022-03-15 2023-03-15 Combination of stat3 targeting oligonucleotides and pd-l1 inhibitors WO2023178141A2 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US202263320163P 2022-03-15 2022-03-15
US63/320,163 2022-03-15

Publications (2)

Publication Number Publication Date
WO2023178141A2 true WO2023178141A2 (en) 2023-09-21
WO2023178141A3 WO2023178141A3 (en) 2023-10-19

Family

ID=88024405

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2023/064381 WO2023178141A2 (en) 2022-03-15 2023-03-15 Combination of stat3 targeting oligonucleotides and pd-l1 inhibitors

Country Status (2)

Country Link
TW (1) TW202406558A (en)
WO (1) WO2023178141A2 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN117890603A (en) * 2024-01-17 2024-04-16 南通大学附属医院 Application of PD-L1 as drug target in screening drugs for inhibiting tendon adhesion

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050196781A1 (en) * 2001-05-18 2005-09-08 Sirna Therapeutics, Inc. RNA interference mediated inhibition of STAT3 gene expression using short interfering nucleic acid (siNA)
CN107106590B (en) * 2014-10-24 2022-10-18 阿斯利康(瑞典)有限公司 Combination (I)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN117890603A (en) * 2024-01-17 2024-04-16 南通大学附属医院 Application of PD-L1 as drug target in screening drugs for inhibiting tendon adhesion

Also Published As

Publication number Publication date
WO2023178141A3 (en) 2023-10-19
TW202406558A (en) 2024-02-16

Similar Documents

Publication Publication Date Title
US10675295B2 (en) Compositions and methods for inhibiting HMGB1 expression
JP2022551269A (en) Chemical modification of small interfering RNA with minimal fluorine content
AU2022256742A1 (en) Compositions and methods for inhibiting ketohexokinase (khk)
WO2023178141A2 (en) Combination of stat3 targeting oligonucleotides and pd-l1 inhibitors
US20220364098A1 (en) Compositions and methods for modulating pnpla3 expression
US20220072024A1 (en) Compositions and methods for inhibiting hmgb1 expression
US20240124875A1 (en) Rnai conjugates and uses thereof
WO2023081500A2 (en) RNAi OLIGONUCLEOTIDE CONJUGATES
US20240117351A1 (en) Compositions and methods for inhibiting gene expression in the central nervous system
WO2024107993A1 (en) Stat3 targeting oligonucleotides and uses thereof
WO2022031850A2 (en) Compositions and methods for the treatment of metabolic syndrome
CN117015384A (en) RNAI conjugates and uses thereof
US20230416743A1 (en) Compositions and methods for inhibiting snca expression
US20230416742A1 (en) Compositions and methods for inhibiting mapt expression
EP4337773A1 (en) Lipid conjugation for targeting neurons of the central nervous system
TW202330920A (en) Compositions and methods for modulating apoc3 expression
TW202345873A (en) Compositions and methods for modulatingscapactivity
WO2024040041A1 (en) Regulation of activity of rnai molecules
TW202334414A (en) Compositions and methods for inhibiting melanocortin 2 receptor and cytochrome p450 11b1 expression

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: 23771609

Country of ref document: EP

Kind code of ref document: A2