COMPLEMENT FACTOR B (CFB) IRNA COMPOSITIONS AND METHODS OF USE THEREOF

Information

  • Patent Application
  • 20240018515
  • Publication Number
    20240018515
  • Date Filed
    October 27, 2022
    a year ago
  • Date Published
    January 18, 2024
    4 months ago
Abstract
The present invention relates to RNAi agents, e.g., dsRNA agents, targeting the complement factor B (CFB) gene. The invention also relates to methods of using such RNAi agents to inhibit expression of a CFB gene and to methods of treating or preventing a CFB-associated disease in a subject.
Description
SEQUENCE LISTING

The instant application contains a Sequence Listing which has been submitted electronically in XML file format and is hereby incorporated by reference in its entirety. Said XML copy, created on Jun. 1, 2023, is named 121301-11404_SL.xml and is 9,553,806 bytes in size.


BACKGROUND OF THE INVENTION

Complement was first discovered in the 1890s when it was found to aid or “complement” the killing of bacteria by heat-stable antibodies present in normal serum (Walport, M. J. (2001) N Engl J Med. 344:1058). The complement system consists of more than 30 proteins that are either present as soluble proteins in the blood or are present as membrane-associated proteins. Activation of complement leads to a sequential cascade of enzymatic reactions, known as complement activation pathways resulting in the formation of the potent anaphylatoxins C3a and C5a that elicit a plethora of physiological responses that range from chemoattraction to apoptosis. Initially, complement was thought to play a major role in innate immunity where a robust and rapid response is mounted against invading pathogens. However, recently it is becoming increasingly evident that complement also plays an important role in adaptive immunity involving T and B cells that help in elimination of pathogens (Dunkelberger J R and Song W C. (2010) Cell Res. 20:34; Molina H, et al. (1996) Proc Natl Acad Sci USA. 93:3357), in maintaining immunologic memory preventing pathogenic re-invasion, and is involved in numerous human pathological states (Qu, H, et al. (2009) Mol Immunol. 47:185; Wagner, E. and Frank M M. (2010) Nat Rev Drug Discov. 9:43).


Complement activation is known to occur through three different pathways: alternate, classical and lectin (FIG. 1) involving proteins that mostly exist as inactive zymogens that are then sequentially cleaved and activated.


The classical pathway is often activated by antibody-antigen complexes or by the C-reactive protein (CRP), both of which interact with complement component C1q. In addition, the classical pathway can be activated by phosphatidyl serine present in apoptotic bodies in the absence of immune complexes.


The lectin pathway is initiated by the mannose-binding lectins (MBL) that bind to complex carbohydrate residues on the surface of pathogens. The activation of the classical pathway or the lectin pathway leads to activation of the (C4b2b) C3 convertase.


The alternate pathway is activated by the binding of C3b, which is spontaneously generated by the hydrolysis of C3, on targeted surfaces. This surface-bound C3b is then recognized by factor B, forming the complex C3bB. The C3bB complex, in turn, is cleaved by factor D to yield the active form of the C3 convertase of the AP (C3bBb). Both types of C3 convertases will cleave C3, forming C3b. C3b then either binds to more factor B, enhancing the complement activation through the AP (the so-called alternative or amplification loop), or leads to the formation of the active C5 convertase (C3bBbC3b or C4bC2bC3b), which cleaves C5 and triggers the late events that result in the formation of the membrane attack complex (MAC) (C5b-9).


Inappropriate activation of the complement system is responsible for propagating or initiating pathology in many different diseases, including, for example, C3 glomerulopathy, systemic lupus erythematosus (SLE), e.g., Lupus Nephritis, IgA nephropathy, diabetic nephropathy, polycystic kidney disease, membranous nephropathy, age-related macular degeneration, atypical hemolytic uremic syndrome, thrombotic microangiopathy, myasthenia gravis, ischemia and reperfusion injury, paroxysmal nocturnal hemoglobinuria, and rheumatoid arthritis.


To date, only one therapeutic that targets the alternate pathway, e.g., the C5-C5a axis, is available for the treatment of complement component-associated diseases, the anti-C5 antibody, eculizumab (Soliris®). Although eculizumab has been shown to be effective for the treatment of paroxysmal nocturnal hemoglobinuria (PNH), atypical hemolytic uremic syndrome (aHUS), and Myasthenia Gravis, and is currently being evaluated in clinical trials for additional complement component-associated diseases, eculizumab therapy requires weekly high dose infusions followed by biweekly maintenance infusions at a high cost. Furthermore, approximately 50% of eculizumab-treated PNH subjects have low level of hemolysis and require residual transfusions (Hill A, et al. (2010) Haematologica 95(4):567-73).


Accordingly, there is a need in the art for compositions and methods for treating diseases, disorders, and conditions associated with complement activation by, for example, activation of complement factor B activity.


SUMMARY OF THE INVENTION

The present invention provides iRNA compositions which affect the RNA-induced silencing complex (RISC)-mediated cleavage of RNA transcripts of a gene encoding complement factor B (CFB). The complement factor B (CFB) may be within a cell, e.g., a cell within a subject, such as a human subject.


Accordingly, in one aspect the invention provides a double stranded ribonucleic acid (dsRNA) agent for inhibiting expression of complement factor B (CFB) in a cell, wherein the dsRNA agent comprises a sense strand and an antisense strand forming a double stranded region, wherein the sense strand comprises at least 15 contiguous nucleotides differing by no more than 0, 1, 2, or 3 nucleotides from the nucleotide sequence of SEQ ID NO:1 and the antisense strand comprises at least 15 contiguous nucleotides differing by no more than 1, 2, or 3 nucleotides from the nucleotide sequence of SEQ ID NO:8. In certain embodiments, the sense strand comprises at least 15 contiguous nucleotides of the nucleotide sequence of SEQ ID NO:1 and the antisense strand comprises at least 15 contiguous nucleotides of the nucleotide sequence of SEQ ID NO:8. In certain embodiments, the sense strand comprises at least 17 contiguous nucleotides of the nucleotide sequence of SEQ ID NO:1 and the antisense strand comprises at least 17 contiguous nucleotides of the nucleotide sequence of SEQ ID NO:8. In certain embodiments, the sense strand comprises at least 19 contiguous nucleotides of the nucleotide sequence of SEQ ID NO:1 and the antisense strand comprises at least 19 contiguous nucleotides of the nucleotide sequence of SEQ ID NO:8.


In another aspect, the present invention provides a double stranded ribonucleic acid (dsRNA) for inhibiting expression of complement factor B (CFB) in a cell, wherein said dsRNA comprises a sense strand and an antisense strand forming a double stranded region, wherein the antisense strand comprises a region of complementarity to an mRNA encoding complement factor B (CFB), and wherein the region of complementarity comprises at least 15 contiguous nucleotides differing by no more than 0, 1, 2, or 3 nucleotides from any one of the antisense nucleotide sequences in any one of Tables 2-7, 13, 16, 19 and 20. In certain embodiments, the region of complementarity comprises at least 15 contiguous nucleotides of any one of the antisense nucleotide sequences in any one of Tables 2-7, 13, 16, 19 and 20. In certain embodiments, the region of complementarity comprises at least 17 contiguous nucleotides of any one of the antisense nucleotide sequences in any one of Tables 2-7, 13, 16, 19 and 20. In certain embodiments, the region of complementarity comprises at least 19 contiguous nucleotides of any one of the antisense nucleotide sequences in any one of Tables 2-7, 13, 16, 19 and 20. In certain embodiments, the region of complementarity comprises at least 20 contiguous nucleotides of any one of the antisense nucleotide sequences in any one of Tables 2-7, 13, 16, 19 and 20. In certain embodiments, the region of complementarity comprises at least 21 contiguous nucleotides of any one of the antisense nucleotide sequences in any one of Tables 2-7, 13, 16, 19 and 20.


In one aspect, the present invention provides a double stranded ribonucleic acid (dsRNA) for inhibiting expression of complement factor B (CFB) in a cell, wherein said dsRNA comprises a sense strand and an antisense strand forming a double stranded region, wherein the sense strand comprises at least 15 contiguous nucleotides differing by no more than three nucleotides from any one of the nucleotide sequence of nucleotides 633-665, 1133-1185, 1133-1173, 1133-1167, 1143-1173, 1540-1563, 1976-2002, 2386-2438, 2386-2418, 2386-2413, and 2389-1418 of SEQ ID NO: 1, and the antisense strand comprises at least 15 contiguous nucleotides from the corresponding nucleotide sequence of SEQ ID NO:8, where a substitution of a T with a U in either SEQ ID NO: 1 or SEQ ID NO: 8 does not count as a difference.


In another aspect, the present invention provides a double stranded ribonucleic acid (dsRNA) for inhibiting expression of complement factor B (CFB) in a cell, wherein said dsRNA comprises a sense strand and an antisense strand forming a double stranded region, wherein the sense strand comprises at least 15 contiguous nucleotides differing by no more than 0, 1, 2, or 3 nucleotides e.g., at least 15 nucleotides, at least 17 nucleotides, at least 19 nucleotides, or at least 20 nucleotides, from any one of the nucleotide sequence of nucleotides 633-655, 643-665, 928-950, 1133-1155, 1140-1162, 1141-1163, 1143-1165, 1145-1167, 1148-1170, 1150-1172, 1151-1173, 1185-1207, 1306-1328, 1534-1556, 1540-1562, 1541-1563, 1976-1998, 1979-2001, 1980-2002, 2078-2100, 2386-2408, 2388-2410, 2389-2411, 2391-2413, 2393-2415, 2395-2417, 2396-2418, 2438-2460, 2602-2624 of SEQ ID NO: 1, and the antisense strand comprises at least 15 contiguous nucleotides differing by no more than 0, 1, 2, or 3 nucleotides, e.g., at least 15 nucleotides, at least 17 nucleotides, at least 19 nucleotides, or at least 20 nucleotides, from the corresponding nucleotide sequence of SEQ ID NO:8, where a substitution of a T with a U in either SEQ ID NO: 1 or SEQ ID NO: 8 does not count as a difference.


In one embodiment, the antisense strand comprises at least 15 contiguous nucleotides differing by nor more than three nucleotides from any one of the antisense strand nucleotide sequences of a duplex selected from the group consisting of AD-560018, AD-559375, AD-559160, AD-559374, AD-559060, AD-559721, AD-559026, AD-558225, AD-557069, AD-558068, AD-557422, AD-558063, AD-558066, AD-556701, AD-558657, AD-559020, AD-559023, AD-558860, AD-560019, AD-560016, AD-559008, AD-559717, AD-557072, AD-558097, AD-557774, AD-557070, AD-558065, AD-557853, and AD-557079. In certain embodiments, the antisense strand comprises at least 15 contiguous nucleotides of any one of the selected duplexes. In certain embodiments, the antisense strand comprises at least 17 contiguous nucleotides of any one of the selected duplexes. In certain embodiments, the antisense strand comprises at least 19 contiguous nucleotides of any one of the selected duplexes. In certain embodiments, the antisense strand comprises at least 20 contiguous nucleotides of any one of the selected duplexes. In certain embodiments, the antisense strand comprises at least 21 contiguous nucleotides of any one of the selected duplexes.


In one embodiment, the sense strand comprises at least 15 contiguous nucleotides differing by nor more than three nucleotides from any one of the antisense strand nucleotide sequences of a duplex selected from the group consisting of AD-560018, AD-559375, AD-559160, AD-559374, AD-559060, AD-559721, AD-559026, AD-558225, AD-557069, AD-558068, AD-557422, AD-558063, AD-558066, AD-556701, AD-558657, AD-559020, AD-559023, AD-558860, AD-560019, AD-560016, AD-559008, AD-559717, AD-557072, AD-558097, AD-557774, AD-557070, AD-558065, AD-557853, and AD-557079. In certain embodiments, the sense strand comprises at least 15 contiguous nucleotides of any one of the selected duplexes. In certain embodiments, the sense strand comprises at least 17 contiguous nucleotides of any one of the selected duplexes. In certain embodiments, the sense strand comprises at least 19 contiguous nucleotides of any one of the selected duplexes. In certain embodiments, the sense strand comprises at least 20 contiguous nucleotides of any one of the selected duplexes. In certain embodiments, the sense strand comprises at least 21 contiguous nucleotides of any one of the selected duplexes.


In one aspect, the present invention provides a double stranded ribonucleic acid (dsRNA) for inhibiting expression of complement factor B (CFB) in a cell, wherein said dsRNA comprises a sense strand and an antisense strand forming a double stranded region, wherein the sense strand comprises at least 15 contiguous nucleotides differing by no more than 0, 1, 2, or 3 nucleotides e.g., at least 15 nucleotides, at least 17 nucleotides, at least 19 nucleotides, or at least 20 nucleotides, from any one of the nucleotide sequence of nucleotides 153-175; 202-224; 219-241; 254-276; 304-326; 321-343; 347-369; 402-424; 418-440; 447-469; 491-513; 528-550; 549-571; 566-588; 591-613; 792-814; 819-841; 967-989; 1042-1064; 1234-1256; 1250-1272; 1269-1291; 1335-1357; 1354-1376; 1372-1394; 1422-1444; 1496-1518; 1670-1692; 1716-1738; 1757-1779; 1774-1796; 1793-1815; 1844-1866; 1871-1893; 1909-1931; 1924-1947; 1947-1969; 2161-2183; 2310-2332; 2330-2352; 2355-2377; 2494-2516; and 2527-2549 of SEQ ID NO: 1, and the antisense strand comprises at least 15 contiguous nucleotides differing by no more than 0, 1, 2, or 3 nucleotides, e.g., at least 15 nucleotides, at least 17 nucleotides, at least 19 nucleotides, or at least 20 nucleotides, from the corresponding nucleotide sequence of SEQ ID NO:8, where a substitution of a T with a U in either SEQ ID NO: 1 or SEQ ID NO: 8 does not count as a difference.


In one embodiment, the antisense strand comprises at least 15 contiguous nucleotides differing by nor more than three nucleotides from any one of the antisense strand nucleotide sequences of a duplex selected from the group consisting of AD-560132.1; AD-560099.1; AD-559998.1; AD-559993.1; AD-559973.1; AD-559882.1; AD-559706.1; AD-559704.1; AD-559688.1; AD-559668.1; AD-559641.1; AD-559609.1; AD-559590.1; AD-559573.1; AD-559532.1; AD-559486.1; AD-559330.1; AD-559274.1; AD-559226.1; AD-559208.1; AD-559189.1; AD-559124.1; AD-559105.1; AD-559089.1; AD-558935.1; AD-558879.1; AD-558777.1; AD-558750.1; AD-558637.1; AD-558612.1; AD-558595.1; AD-558574.1; AD-558555.1; AD-558511.1; AD-558482.1; AD-558466.1; AD-558450.1; AD-558424.1; AD-558407.1; AD-558393.1; AD-558378.1; AD-558361.1; AD-558312.1. In certain embodiments, the antisense strand comprises at least 15 contiguous nucleotides of any one of the selected duplexes. In certain embodiments, the antisense strand comprises at least 17 contiguous nucleotides of any one of the selected duplexes. In certain embodiments, the antisense strand comprises at least 19 contiguous nucleotides of any one of the selected duplexes. In certain embodiments, the antisense strand comprises at least 20 contiguous nucleotides of any one of the selected duplexes. In certain embodiments, the antisense strand comprises at least 21 contiguous nucleotides of any one of the selected duplexes.


In one embodiment, the sense strand comprises at least 15 contiguous nucleotides differing by nor more than three nucleotides from any one of the antisense strand nucleotide sequences of a duplex selected from the group consisting of AD-560132.1; AD-560099.1; AD-559998.1; AD-559993.1; AD-559973.1; AD-559882.1; AD-559706.1; AD-559704.1; AD-559688.1; AD-559668.1; AD-559641.1; AD-559609.1; AD-559590.1; AD-559573.1; AD-559532.1; AD-559486.1; AD-559330.1; AD-559274.1; AD-559226.1; AD-559208.1; AD-559189.1; AD-559124.1; AD-559105.1; AD-559089.1; AD-558935.1; AD-558879.1; AD-558777.1; AD-558750.1; AD-558637.1; AD-558612.1; AD-558595.1; AD-558574.1; AD-558555.1; AD-558511.1; AD-558482.1; AD-558466.1; AD-558450.1; AD-558424.1; AD-558407.1; AD-558393.1; AD-558378.1; AD-558361.1; AD-558312.1. In certain embodiments, the sense strand comprises at least 15 contiguous nucleotides of any one of the selected duplexes. In certain embodiments, the sense strand comprises at least 17 contiguous nucleotides of any one of the selected duplexes. In certain embodiments, the sense strand comprises at least 19 contiguous nucleotides of any one of the selected duplexes. In certain embodiments, the sense strand comprises at least 20 contiguous nucleotides of any one of the selected duplexes. In certain embodiments, the sense strand comprises at least 21 contiguous nucleotides of any one of the selected duplexes.


In one embodiment, the dsRNA agent comprises at least one modified nucleotide.


In one embodiment, substantially all of the nucleotides of the sense strand; substantially all of the nucleotides of the antisense strand comprise a modification; or substantially all of the nucleotides of the sense strand and substantially all of the nucleotides of the antisense strand comprise a modification.


In one embodiment, all of the nucleotides of the sense strand comprise a modification; all of the nucleotides of the antisense strand comprise a modification; or all of the nucleotides of the sense strand and all of the nucleotides of the antisense strand comprise a modification.


In one embodiment, at least one of the modified nucleotides is selected from the group consisting of a deoxy-nucleotide, a 3′-terminal deoxythimidine (dT) nucleotide, a 2′-O-methyl modified nucleotide, a 2′-fluoro modified nucleotide, a 2′-deoxy-modified nucleotide, a locked nucleotide, an unlocked nucleotide, a conformationally restricted nucleotide, a constrained ethyl nucleotide, an abasic nucleotide, a 2′-amino-modified nucleotide, a 2′-O-allyl-modified nucleotide, 2′-C-alkyl-modified nucleotide, a 2′-methoxyethyl modified nucleotide, a 2′-O-alkyl-modified nucleotide, a morpholino nucleotide, a phosphoramidate, a non-natural base comprising nucleotide, a tetrahydropyran modified nucleotide, a 1,5-anhydrohexitol modified nucleotide, a cyclohexenyl modified nucleotide, a nucleotide comprising a phosphorothioate group, a nucleotide comprising a methylphosphonate group, a nucleotide comprising a 5′-phosphate, a nucleotide comprising a 5′-phosphate mimic, a nucleotide comprising a 2′-phosphate group, e.g., cytidine-2′-phosphate (C2p); guanosine-2′-phosphate (G2p); uridine-2′-phosphate (U2p); adenosine-2′-phosphate (A2p); a thermally destabilizing nucleotide, a glycol modified nucleotide (GNA), and a 2-O—(N-methylacetamide) modified nucleotide; and combinations thereof.


In one embodiment, the modifications on the nucleotides are selected from the group consisting of LNA, HNA, CeNA, 2′-methoxyethyl, 2′-O-alkyl, 2′-O-allyl, 2′-C-allyl, 2′-fluoro, 2′-deoxy, 2′-hydroxyl, and glycol; and combinations thereof.


In one embodiment, at least one of the modified nucleotides is selected from the group consisting of a deoxy-nucleotide, a 2′-O-methyl modified nucleotide, a 2′-fluoro modified nucleotide, a 2′-deoxy-modified nucleotide, a glycol modified nucleotide (GNA), e.g., Ggn, Cgn, Tgn, or Agn, a nucleotide comprising a 2′-phosphate group, and, a vinyl-phosphonate nucleotide; and combinations thereof.


In another embodiment, at least one of the modifications on the nucleotides is a thermally destabilizing nucleotide modification.


In one embodiment, the thermally destabilizing nucleotide modification is selected from the group consisting of an abasic modification; a mismatch with the opposing nucleotide in the duplex; and destabilizing sugar modification, a 2′-deoxy modification, an acyclic nucleotide, an unlocked nucleic acid (UNA), and a glycerol nucleic acid (GNA).


The double stranded region may be 19-30 nucleotide pairs in length; 19-25 nucleotide pairs in length; 19-23 nucleotide pairs in length; 23-27 nucleotide pairs in length; or 21-23 nucleotide pairs in length.


In one embodiment, each strand is independently no more than 30 nucleotides in length.


In one embodiment, the sense strand is 21 nucleotides in length and the antisense strand is 23 nucleotides in length.


The region of complementarity may be at least 17 nucleotides in length; 19-23 nucleotides in length; or 19 nucleotides in length.


In one embodiment, at least one strand comprises a 3′ overhang of at least 1 nucleotide. In another embodiment, at least one strand comprises a 3′ overhang of at least 2 nucleotides.


In one embodiment, the dsRNA agent further comprises a ligand.


In one embodiment, the ligand is conjugated to the 3′ end of the sense strand of the dsRNA agent.


In one embodiment, the ligand is an N-acetylgalactosamine (GalNAc) derivative.


In one embodiment, the ligand is one or more GalNAc derivatives attached through a monovalent, bivalent, or trivalent branched linker.


In one embodiment, the ligand is




embedded image


In one embodiment, the dsRNA agent is conjugated to the ligand as shown in the following schematic




embedded image


and, wherein X is O or S.


In one embodiment, the X is O.


In one embodiment, the dsRNA agent further comprises at least one phosphorothioate or methylphosphonate internucleotide linkage.


In one embodiment, the phosphorothioate or methylphosphonate internucleotide linkage is at the 3′-terminus of one strand, e.g., the antisense strand or the sense strand.


In another embodiment, the phosphorothioate or methylphosphonate internucleotide linkage is at the 5′-terminus of one strand, e.g., the antisense strand or the sense strand.


In one embodiment, the phosphorothioate or methylphosphonate internucleotide linkage is at the both the 5′- and 3′-terminus of one strand. In one embodiment, the strand is the antisense strand.


In one embodiment, the base pair at the 1 position of the 5′-end of the antisense strand of the duplex is an AU base pair.


The present invention also provides cells containing any of the dsRNA agents of the invention and pharmaceutical compositions comprising any of the dsRNA agents of the invention.


The pharmaceutical composition of the invention may include dsRNA agent in an unbuffered solution, e.g., saline or water, or the pharmaceutical composition of the invention may include the dsRNA agent is in a buffer solution, e.g., a buffer solution comprising acetate, citrate, prolamine, carbonate, or phosphate or any combination thereof; or phosphate buffered saline (PBS).


In one aspect, the present invention provides a method of inhibiting expression of a complement factor B (CFB) gene in a cell. The method includes contacting the cell with any of the dsRNAs of the invention or any of the pharmaceutical compositions of the invention, thereby inhibiting expression of the CFB gene in the cell.


In one embodiment, the cell is within a subject, e.g., a human subject, e.g., a subject having a complement factor B-associated disorder. Such disorders are typically associated with inflammation or immune system activation, e.g., membrane attack complex-mediated lysis, anaphylaxis, or hemolysis. Non-limiting examples of complement factor B-associated disorders include paroxysmal nocturnal hemoglobinuria (PNH), atypical hemolytic uremic syndrome (aHUS), asthma, rheumatoid arthritis (RA); antiphospholipid antibody syndrome; lupus nephritis; ischemia-reperfusion injury; typical or infectious hemolytic uremic syndrome (tHUS); dense deposit disease (DDD); neuromyelitis optica (NMO); multifocal motor neuropathy (MMN); multiple sclerosis (MS); macular degeneration (e.g., age-related macular degeneration (AMD)); hemolysis, elevated liver enzymes, and low platelets (HELLP) syndrome; thrombotic thrombocytopenic purpura (TTP); spontaneous fetal loss; Pauci-immune vasculitis; epidermolysis bullosa; recurrent fetal loss; pre-eclampsia, traumatic brain injury, myasthenia gravis, cold agglutinin disease, dermatomyositis bullous pemphigoid, Shiga toxin E. coli-related hemolytic uremic syndrome, C3 neuropathy, anti-neutrophil cytoplasmic antibody-associated vasculitis (e.g., granulomatosis with polyangiitis (previously known as Wegener granulomatosis), Churg-Strauss syndrome, and microscopic polyangiitis), humoral and vascular transplant rejection, graft dysfunction, myocardial infarction (e.g., tissue damage and ischemia in myocardial infarction), an allogenic transplant, sepsis (e.g., poor outcome in sepsis), Coronary artery disease, dermatomyositis, Graves' disease, atherosclerosis, Alzheimer's disease, systemic inflammatory response sepsis, septic shock, spinal cord injury, glomerulonephritis, Hashimoto's thyroiditis, type I diabetes, psoriasis, pemphigus, autoimmune hemolytic anemia (AIHA), ITP, Goodpasture syndrome, Degos disease, antiphospholipid syndrome (APS), catastrophic APS (CAPS), a cardiovascular disorder, myocarditis, a cerebrovascular disorder, a peripheral (e.g., musculoskeletal) vascular disorder, a renovascular disorder, a mesenteric/enteric vascular disorder, vasculitis, Henoch-Schönlein purpura nephritis, systemic lupus erythematosus-associated vasculitis, vasculitis associated with rheumatoid arthritis, immune complex vasculitis, Takayasu's disease, dilated cardiomyopathy, diabetic angiopathy, Kawasaki's disease (arteritis), venous gas embolus (VGE), and restenosis following stent placement, rotational atherectomy, and percutaneous transluminal coronary angioplasty (PTCA) (see, e.g., Holers (2008) Immunological Reviews 223:300-316; Holers and Thurman (2004) Molecular Immunology 41:147-152; U.S. Patent Publication No. 20070172483).


In one embodiment, the complement factor B-associate disease is selected from the group consisting of C3 glomerulopathy, systemic lupus erythematosus (SLE), e.g., Lupus Nephritis, IgA nephropathy, diabetic nephropathy, polycystic kidney disease, membranous nephropathy, age-related macular degeneration, atypical hemolytic uremic syndrome, thrombotic microangiopathy, myasthenia gravis, ischemia and reperfusion injury, paroxysmal nocturnal hemoglobinuria, and rheumatoid arthritis


In another embodiment, the complement factor B-associate disease is selected from the group consisting of C3 glomerulopathy, systemic lupus erythematosus (SLE), e.g., Lupus Nephritis, IgA nephropathy, diabetic nephropathy, and polycystic kidney disease.


In one embodiment, contacting the cell with the dsRNA agent inhibits the expression of CFB by at least 50%, 60%, 70%, 80%, 90%, or 95%.


In one embodiment, inhibiting expression of CFB decreases CFB protein level in serum of the subject by at least 50%, 60%, 70%, 80%, 90%, or 95%.


In one aspect, the present invention provides a method of treating a subject having a disorder that would benefit from reduction in complement factor B (CFB) expression. The method includes administering to the subject a therapeutically effective amount of any of the dsRNAs of the invention or any of the pharmaceutical compositions of the invention, thereby treating the subject having the disorder that would benefit from reduction in CFB expression.


In another aspect, the present invention provides a method of preventing development of a disorder that would benefit from reduction in complement factor B (CFB) expression in a subject having at least one sign or symptom of a disorder who does not yet meet the diagnostic criteria for that disorder. The method includes administering to the subject a prophylactically effective amount of any of the dsRNAs of the invention or any of the pharmaceutical compositions of the invention, thereby preventing the subject progressing to meet the diagnostic criteria of the disorder that would benefit from reduction in CFB expression.


In one embodiment, the disorder is a complement factor B- (CFB)-associated disorder.


In one embodiment, the subject is human.


In one embodiment, the dsRNA agent is administered to the subject at a dose of about 0.01 mg/kg to about 50 mg/kg.


In one embodiment, the dsRNA agent is administered to the subject subcutaneously.


In one embodiment, the level of CFB in the subject sample(s) is a CFB protein level in a blood or serum sample(s).


In one embodiment, the administration of the agent to the subject causes a decrease in hemolysis or a decrease in CFB protein accumulation.


In certain embodiments, the methods of the invention further comprise administering to the subject an additional therapeutic agent.


In some aspects, the additional therapeutic agent is an iRNA agent targeting a C5 gene, such as those described in U.S. Pat. No. 9,249,415, the entire contents of which are hereby incorporated herein by reference.


In other aspects, the additional therapeutic agent is an iRNA agent targeting a complement factor B (CFB) gene, such as those described in U.S. Pat. No. 10,465,194, the entire contents of which are hereby incorporated herein by reference.


In other aspects, the additional therapeutic agent is an inhibitor of C5, such as an anti-complement component C5 antibody, or antigen-binding fragment thereof (e.g., eculizumab, ravulizumab-cwvz, or pozelimab (REGN3918)) or a C5 peptide inhibitor (e.g., zilucoplan). Eculizumab is a humanized monoclonal IgG2/4, kappa light chain antibody that specifically binds complement component C5 with high affinity and inhibits cleavage of C5 to C5a and C5b, thereby inhibiting the generation of the terminal complement complex C5b-9. Eculizumab is described in U.S. Pat. No. 6,355,245, the entire contents of which are incorporated herein by reference. Ravulizumab-cwvz is a humanized IgG2/4 monoclonal antibody that specifically binds complement component C5 with high affinity and inhibits cleavage of C5 to C5a and C5b, thereby inhibiting the generation of the terminal complement complex C5b-9. Ravulizumab-cwvz is described in WO2015134894, the entire contents of which are incorporated herein by reference. Pozelimab (also known as H4H12166P, described in US20170355757, the entire contents of which are incorporated herein by reference) is a fully-human IgG4 monoclonal antibody designed to block complement factor C5. Zilucoplan is a synthetic, macrocyclic peptide that binds complement component 5 (C5) with sub-nanomolar affinity and allosterically inhibits its cleavage into C5a and C5b upon activation of the classical, alternative, or lectin pathways (see, e.g., WO2017105939, the entire contents of which are incorporated herein by reference).


In yet other aspects, the additional therapeutic is a C3 peptide inhibitor, or analog thereof. In one embodiment, the C3 peptide inhibitor is compstatin. Compstatin is a cyclic tridecapeptide with potent and selective C3 inhibitory activity. Compstatin, and its analogs, are described in U.S. Pat. Nos. 7,888,323, 7,989,589, and 8,442,776, in U.S. Patent Publication No. 2012/0178694 and 2013/0053302, and in PCT Publication Nos. WO 2012/174055, WO 2012/2178083, WO 2013/036778, the entire contents of each of which are incorporated herein by reference.


In certain embodiments, treatments known in the art for the various CFB-associated diseases are used in combination with the RNAi agents of the invention.


The present invention also provides kits comprising any of the dsRNAs of the invention or any of the pharmaceutical compositions of the invention, and optionally, instructions for use.





BRIEF DESCRIPTION OF THE DRAWINGS


FIG. 1 depicts the three complement pathways: alternative, classical and lectin.



FIG. 2A is a heatmap depicting the effect of dual targeting of C3 and CFB on alternative hemolytic activity in human sera in vitro.



FIG. 2B is a heatmap depicting the effect of dual targeting of C3 and C5 on alternative hemolytic activity in human sera in vitro.



FIG. 2C is a heatmap depicting the effect of dual targeting of C3 and C5 on classical hemolytic activity in human sera in vitro.



FIG. 2D is a heatmap depicting the effect of dual targeting of C3 and C5 on classical hemolytic activity as determined by the Wieslab® Complement Classical Pathway (CCP) assay in human sera in vitro.



FIG. 3A is a graph depicting the effect of administration of a single 6 mg/kg dose of a dsRNA agent targeting C3; or a dsRNA agent targeting CFB; or a dsRNA agent targeting C5; or a single 6 mg/kg dose of a dsRNA agent targeting C3 and a single 6 mg/kg dose of a dsRNA agent targeting CFB; or a single 6 mg/kg dose of a dsRNA agent targeting C3 and a single 6 mg/kg dose of a dsRNA agent targeting C5; or a single 6 mg/kg dose of a dsRNA agent targeting CFB and a single 6 mg/kg dose of a dsRNA agent targeting C5 on C3 protein levels, CFB protein levels or C5 protein levels in the sera of non-human primates.



FIG. 3B is a graph depicting the effect of administration of a single 6 mg/kg dose of a dsRNA agent targeting C3; or a dsRNA agent targeting CFB; or a dsRNA agent targeting C5; or a single 6 mg/kg dose of a dsRNA agent targeting C3 and a single 6 mg/kg dose of a dsRNA agent targeting CFB; or a single 6 mg/kg dose of a dsRNA agent targeting C3 and a single 6 mg/kg dose of a dsRNA agent targeting C5; or a single 6 mg/kg dose of a dsRNA agent targeting CFB and a single 6 mg/kg dose of a dsRNA agent targeting C5 on alternative hemolytic activity in the sera of non-human primates.



FIG. 3C is a graph depicting the effect of administration of a single 6 mg/kg dose of a dsRNA agent targeting C3; or a dsRNA agent targeting CFB; or a dsRNA agent targeting C5; or a single 6 mg/kg dose of a dsRNA agent targeting C3 and a single 6 mg/kg dose of a dsRNA agent targeting CFB; or a single 6 mg/kg dose of a dsRNA agent targeting C3 and a single 6 mg/kg dose of a dsRNA agent targeting C5; or a single 6 mg/kg dose of a dsRNA agent targeting CFB and a single 6 mg/kg dose of a dsRNA agent targeting C5 on classical hemolytic activity in the sera of non-human primates.



FIG. 3D is a graph depicting the effect of administration of a single 6 mg/kg dose of a dsRNA agent targeting C3; or a dsRNA agent targeting CFB; or a dsRNA agent targeting C5; or a single 6 mg/kg dose of a dsRNA agent targeting C3 and a single 6 mg/kg dose of a dsRNA agent targeting CFB; or a single 6 mg/kg dose of a dsRNA agent targeting C3 and a single 6 mg/kg dose of a dsRNA agent targeting C5; or a single 6 mg/kg dose of a dsRNA agent targeting CFB and a single 6 mg/kg dose of a dsRNA agent targeting C5 on alternative hemolytic activity as determined by the Wieslab® Complement Alternative Pathway (CAP) assay in the sera of non-human primates.



FIG. 3E is a graph depicting the effect of administration of a single 6 mg/kg dose of a dsRNA agent targeting C3; or a dsRNA agent targeting CFB; or a dsRNA agent targeting C5; or a single 6 mg/kg dose of a dsRNA agent targeting C3 and a single 6 mg/kg dose of a dsRNA agent targeting CFB; or a single 6 mg/kg dose of a dsRNA agent targeting C3 and a single 6 mg/kg dose of a dsRNA agent targeting C5; or a single 6 mg/kg dose of a dsRNA agent targeting CFB and a single 6 mg/kg dose of a dsRNA agent targeting C5 on alternative hemolytic activity as determined by the Wieslab® Complement Classical Pathway (CCP) assay in the sera of non-human primates.





DETAILED DESCRIPTION OF THE INVENTION

The present invention provides iRNA compositions which effect the RNA-induced silencing complex (RISC)-mediated cleavage of RNA transcripts of a complement factor B (CFB) gene. The gene may be within a cell, e.g., a cell within a subject, such as a human. The use of these iRNAs enables the targeted degradation of mRNAs of the corresponding gene (complement factor B gene) in mammals.


The iRNAs of the invention have been designed to target the human complement factor B gene, including portions of the gene that are conserved in the complement factor B orthologs of other mammalian species. Without intending to be limited by theory, it is believed that a combination or sub-combination of the foregoing properties and the specific target sites or the specific modifications in these iRNAs confer to the iRNAs of the invention improved efficacy, stability, potency, durability, and safety.


Accordingly, the present invention provides methods for treating and preventing a complement factor B-associated disorder, disease, or condition, e.g., a disorder, disease, or condition with inflammation or immune system activation, e.g., membrane attack complex-mediated lysis, anaphylaxis, or hemolysis, e.g., C3 glomerulopathy, systemic lupus erythematosus (SLE), e.g., Lupus Nephritis, IgA nephropathy, diabetic nephropathy, and polycystic kidney disease, using iRNA compositions which effect the RNA-induced silencing complex (RISC)-mediated cleavage of RNA transcripts of a complement factor B gene.


The iRNAs of the invention include an RNA strand (the antisense strand) having a region which is up to about 30 nucleotides or less in length, e.g., 19-30, 19-29, 19-28, 19-27, 19-26, 19-25, 19-24, 19-23, 19-22, 19-21, 19-20, 20-30, 20-29, 20-28, 20-27, 20-26, 20-25, 20-24, 20-23, 20-22, 20-21, 21-30, 21-29, 21-28, 21-27, 21-26, 21-25, 21-24, 21-23, or 21-22 nucleotides in length, which region is substantially complementary to at least part of an mRNA transcript of a complement factor B gene. In certain embodiments, the RNAi agents of the disclosure include an RNA strand (the antisense strand) having a region which is about 21-23 nucleotides in length, which region is substantially complementary to at least part of an mRNA transcript of a complement factor B gene.


In certain embodiments, one or both of the strands of the double stranded RNAi agents of the invention is up to 66 nucleotides in length, e.g., 36-66, 26-36, 25-36, 31-60, 22-43, 27-53 nucleotides in length, with a region of at least 19 contiguous nucleotides that is substantially complementary to at least a part of an mRNA transcript of a complement factor B gene. In some embodiments, such iRNA agents having longer length antisense strands may include a second RNA strand (the sense strand) of 20-60 nucleotides in length wherein the sense and antisense strands form a duplex of 18-30 contiguous nucleotides.


The use of iRNAs of the invention enables the targeted degradation of mRNAs of the corresponding gene (complement factor B gene) in mammals. Using in vitro and in vivo assays, the present inventors have demonstrated that iRNAs targeting a complement factor B gene can potently mediate RNAi, resulting in significant inhibition of expression of a complement factor B gene. Thus, methods and compositions including these iRNAs are useful for treating a subject having a complement factor B-associated disorder, e.g., C3 glomerulopathy, systemic lupus erythematosus (SLE), e.g., Lupus Nephritis, IgA nephropathy, diabetic nephropathy, and polycystic kidney disease.


Accordingly, the present invention provides methods and combination therapies for treating a subject having a disorder that would benefit from inhibiting or reducing the expression of a complement factor B gene, e.g., a complement factor B-associated disease, such as C3 glomerulopathy, systemic lupus erythematosus (SLE), e.g., Lupus Nephritis, IgA nephropathy, diabetic nephropathy, and polycystic kidney disease, using iRNA compositions which effect the RNA-induced silencing complex (RISC)-mediated cleavage of RNA transcripts of a CFB gene.


The present invention also provides methods for preventing at least one symptom in a subject having a disorder that would benefit from inhibiting or reducing the expression of a complement factor B gene, e.g., C3 glomerulopathy, systemic lupus erythematosus (SLE), e.g., Lupus Nephritis, IgA nephropathy, diabetic nephropathy, and polycystic kidney disease.


In certain embodiments, the administration of the dsRNA to the subject causes a decrease in CFB mRNA level, CFB protein level, CH50 activity (a measure of total hemolytic complement), AH50 (a measure the hemolytic activity of the alternate pathway of complement), lactate dehydrogenase (LDH) (a measure of intravascular hemolysis), hemoglobin levels; the level of any one or more of C3, C9, C5, C5a, C5b, and soluble C5b-9 complex.


The following detailed description discloses how to make and use compositions containing iRNAs to inhibit the expression of a complement factor B gene as well as compositions, uses, and methods for treating subjects that would benefit from inhibition or reduction of the expression of a complement factor B gene, e.g., subjects susceptible to or diagnosed with a complement factor B-associated disorder.


I. Definitions

In order that the present invention may be more readily understood, certain terms are first defined. In addition, it should be noted that whenever a value or range of values of a parameter are recited, it is intended that values and ranges intermediate to the recited values are also intended to be part of this invention.


The articles “a” and “an” are used herein to refer to one or to more than one (i.e., to at least one) of the grammatical object of the article. By way of example, “an element” means one element or more than one element, e.g., a plurality of elements.


The term “including” is used herein to mean, and is used interchangeably with, the phrase “including but not limited to”.


The term “or” is used herein to mean, and is used interchangeably with, the term “and/or,” unless context clearly indicates otherwise. For example, “sense strand or antisense strand” is understood as “sense strand or antisense strand or sense strand and antisense strand.”


The term “about” is used herein to mean within the typical ranges of tolerances in the art. For example, “about” can be understood as about 2 standard deviations from the mean. In certain embodiments, about means ±10%. In certain embodiments, about means ±5%. When about is present before a series of numbers or a range, it is understood that “about” can modify each of the numbers in the series or range.


The term “at least”, “no less than” or “or more” prior to a number or series of numbers is understood to include the number adjacent to the term “at least”, and all subsequent numbers or integers that could logically be included, as clear from context. For example, the number of nucleotides in a nucleic acid molecule must be an integer. For example, “at least 19 nucleotides of a 21 nucleotide nucleic acid molecule” means that 19, 20, or 21 nucleotides have the indicated property. When at least is present before a series of numbers or a range, it is understood that “at least” can modify each of the numbers in the series or range.


As used herein, “no more than” or “or less” is understood as the value adjacent to the phrase and logical lower values or integers, as logical from context, to zero. For example, a duplex with an overhang of “no more than 2 nucleotides” has a 2, 1, or 0 nucleotide overhang. When “no more than” is present before a series of numbers or a range, it is understood that “no more than” can modify each of the numbers in the series or range. As used herein, ranges include both the upper and lower limit.


As used herein, methods of detection can include determination that the amount of analyte present is below the level of detection of the method.


In the event of a conflict between an indicated target site and the nucleotide sequence for a sense or antisense strand, the indicated sequence takes precedence.


In the event of a conflict between a sequence and its indicated site on a transcript or other sequence, the nucleotide sequence recited in the specification takes precedence.


As used herein, the term “Complement Factor B,” used interchangeably with the term “CFB,” refers to the well-known gene and polypeptide, also known in the art as AHUS, BF, CFAB, BFD, FB, GBG, FBI12, B-Factor, Properdin, H2-Bf, Glycine-Rich Beta Glycoprotein, C3 Proaccelerator, Properdin Factor 2B, C3 Proactivator, PBF2, Glycine-Rich Beta-Glycoprotein, C3/C5 Convertase, EC 3.4.21, and EC 3.4.21.473.


The term “CFB” includes human CFB, the amino acid and nucleotide sequence of which may be found in, for example, GenBank Accession No. GI:189181756; mouse CFB, the amino acid and nucleotide sequence of which may be found in, for example, GenBank Accession Nos. GI:218156288 and GI:218156290; rat CFB, the amino acid and nucleotide sequence of which may be found in, for example, GenBank Accession No. GI:218156284; and chimpanzee CFB, the amino acid and nucleotide sequence of which may be found in, for example, GenBank Accession No. GI:57114201.


The term “CFB” also includes Macacafascicularis CFB, the amino acid and nucleotide sequence of which may be found in, for example, GenBank Accession No. GI:544428919 and in the entry for the gene, ENSMMUP00000000985 (locus=scaffold3881:47830:53620), in the Macaca genome project web site (macque.genomics.org.cn/page/species/index.jsp). Additional examples of CFB mRNA sequences are readily available using, e.g., GenBank, UniProt, OMIM, and the Macaca genome project web site.


Exemplary CFB nucleotide sequences may also be found in SEQ ID NOs:1-7. SEQ ID NOs:8-14 are the antisense sequences of SEQ ID NOs: 1-7, respectively.


The term “CFB,” as used herein, also refers to naturally occurring DNA sequence variations of the CFB gene. Non-limiting examples of sequence variations within the CFB gene include 1598A>G in exon 12, which results in a lysine being changed to an arginine at amino acid residue 533; 858C>G in exon 6, which results in a phenylalanine being changed to a leucine at amino acid residue 286; and 967A>G in exon 7, which results in a lysine being changed to an alanine at amino acid residue 323 (Tawadrous H. et al. (2010) Pediatr Nephrol. 25:947; Goicoechea de Jorge E et al. (2007) Proc Natl Acad Sci. USA 104:240). The term “CFB,” as used herein, also refers to single nucleotide polymorphisms in the CFB gene. Numerous sequence variations within the CFB gene have been identified and may be found at, for example, NCBI dbSNP and UniProt (see, e.g., ncbi.nlm.nih.gov/snp).


Further information on CFB can be found, for example, at www.ncbi.nlm.nih.gov/gene/629.


Additional examples of CFB mRNA sequences are readily available through publicly available databases, e.g., GenBank, UniProt, OMIM, and the Macaca genome project web site.


The entire contents of each of the foregoing GenBank Accession numbers and the Gene database numbers are incorporated herein by reference as of the date of filing this application.


As used herein, “target sequence” refers to a contiguous portion of the nucleotide sequence of an mRNA molecule formed during the transcription of a complement factor B gene, including mRNA that is a product of RNA processing of a primary transcription product. The target portion of the sequence will be at least long enough to serve as a substrate for iRNA-directed cleavage at or near that portion of the nucleotide sequence of an mRNA molecule formed during the transcription of a CFB gene. In one embodiment, the target sequence is within the protein coding region of CFB.


The target sequence may be from about 19-36 nucleotides in length, e.g., about 19-30 nucleotides in length. For example, the target sequence can be about 19-30 nucleotides, 19-30, 19-29, 19-28, 19-27, 19-26, 19-25, 19-24, 19-23, 19-22, 19-21, 19-20, 20-30, 20-29, 20-28, 20-27, 20-26, 20-25, 20-24, 20-23, 20-22, 20-21, 21-30, 21-29, 21-28, 21-27, 21-26, 21-25, 21-24, 21-23, or 21-22 nucleotides in length. In some embodiments, the target sequence is about 19 to about 30 nucleotides in length. In other embodiments, the target sequence is about 19 to about 25 nucleotides in length. In still other embodiments, the target sequence is about 19 to about 23 nucleotides in length. In some embodiments, the target sequence is about 21 to about 23 nucleotides in length. Ranges and lengths intermediate to the above recited ranges and lengths are also contemplated to be part of the invention.


As used herein, the term “strand comprising a sequence” refers to an oligonucleotide comprising a chain of nucleotides that is described by the sequence referred to using the standard nucleotide nomenclature.


“G,” “C,” “A,” “T,” and “U” each generally stand for a nucleotide that contains guanine, cytosine, adenine, thymidine, and uracil as a base, respectively. However, it will be understood that the term “ribonucleotide” or “nucleotide” can also refer to a modified nucleotide, as further detailed below, or a surrogate replacement moiety (see, e.g., Table 1). The skilled person is well aware that guanine, cytosine, adenine, and uracil can be replaced by other moieties without substantially altering the base pairing properties of an oligonucleotide comprising a nucleotide bearing such replacement moiety. For example, without limitation, a nucleotide comprising inosine as its base can base pair with nucleotides containing adenine, cytosine, or uracil. Hence, nucleotides containing uracil, guanine, or adenine can be replaced in the nucleotide sequences of dsRNA featured in the invention by a nucleotide containing, for example, inosine. In another example, adenine and cytosine anywhere in the oligonucleotide can be replaced with guanine and uracil, respectively to form G-U Wobble base pairing with the target mRNA. Sequences containing such replacement moieties are suitable for the compositions and methods featured in the invention.


The terms “iRNA”, “RNAi agent,” “iRNA agent,”, “RNA interference agent” as used interchangeably herein, refer to an agent that contains RNA as that term is defined herein, and which mediates the targeted cleavage of an RNA transcript via an RNA-induced silencing complex (RISC) pathway. iRNA directs the sequence-specific degradation of mRNA through a process known as RNA interference (RNAi). The iRNA modulates, e.g., inhibits, the expression of a complement factor B gene in a cell, e.g., a cell within a subject, such as a mammalian subject.


In one embodiment, an RNAi agent of the invention includes a single stranded RNA that interacts with a target RNA sequence, e.g., a complement factor B target mRNA sequence, to direct the cleavage of the target RNA. Without wishing to be bound by theory it is believed that long double stranded RNA introduced into cells is broken down into siRNA by a Type III endonuclease known as Dicer (Sharp et al. (2001) Genes Dev. 15:485). Dicer, a ribonuclease-III-like enzyme, processes the dsRNA into 19-23 base pair short interfering RNAs with characteristic two base 3′ overhangs (Bernstein, et al., (2001) Nature 409:363). The siRNAs are then incorporated into an RNA-induced silencing complex (RISC) where one or more helicases unwind the siRNA duplex, enabling the complementary antisense strand to guide target recognition (Nykanen, et al., (2001) Cell 107:309). Upon binding to the appropriate target mRNA, one or more endonucleases within the RISC cleave the target to induce silencing (Elbashir, et al., (2001) Genes Dev. 15:188). Thus, in one aspect the invention relates to a single stranded RNA (siRNA) generated within a cell and which promotes the formation of a RISC complex to effect silencing of the target gene, i.e., a complement factor B (CFB) gene. Accordingly, the term “siRNA” is also used herein to refer to an iRNA as described above.


In certain embodiments, the RNAi agent may be a single-stranded siRNA (ssRNAi) that is introduced into a cell or organism to inhibit a target mRNA. Single-stranded RNAi agents bind to the RISC endonuclease, Argonaute 2, which then cleaves the target mRNA. The single-stranded siRNAs are generally 15-30 nucleotides and are chemically modified. The design and testing of single-stranded siRNAs are described in U.S. Pat. No. 8,101,348 and in Lima et al., (2012) Cell 150:883-894, the entire contents of each of which are hereby incorporated herein by reference. Any of the antisense nucleotide sequences described herein may be used as a single-stranded siRNA as described herein or as chemically modified by the methods described in Lima et al., (2012) Cell 150:883-894.


In certain embodiments, an “iRNA” for use in the compositions, uses, and methods of the invention is a double stranded RNA and is referred to herein as a “double stranded RNA agent,” “double stranded RNA (dsRNA) molecule,” “dsRNA agent,” or “dsRNA”. The term “dsRNA”, refers to a complex of ribonucleic acid molecules, having a duplex structure comprising two anti-parallel and substantially complementary nucleic acid strands, referred to as having “sense” and “antisense” orientations with respect to a target RNA, i.e., a complement factor B (CFB) gene. In some embodiments of the invention, a double stranded RNA (dsRNA) triggers the degradation of a target RNA, e.g., an mRNA, through a post-transcriptional gene-silencing mechanism referred to herein as RNA interference or RNAi.


As used herein, the term “modified nucleotide” refers to a nucleotide having, independently, a modified sugar moiety, a modified internucleotide linkage, or modified nucleobase, or any combination thereof. Thus, the term modified nucleotide encompasses substitutions, additions or removal of, e.g., a functional group or atom, to internucleoside linkages, sugar moieties, or nucleobases. The modifications suitable for use in the agents of the invention include all types of modifications disclosed herein or known in the art. Any such modifications, as used in a siRNA type molecule, are encompassed by “iRNA” or “RNAi agent” for the purposes of this specification and claims.


In certain embodiments of the instant disclosure, inclusion of a deoxy-nucleotide—which is acknowledged as a naturally occurring form of nucleotide—if present within a RNAi agent can be considered to constitute a modified nucleotide.


The duplex region may be of any length that permits specific degradation of a desired target RNA through a RISC pathway, and may range from about 19 to 36 base pairs in length, e.g., about 19-30 base pairs in length, for example, about 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, or 36 base pairs in length, such as about 19-30, 19-29, 19-28, 19-27, 19-26, 19-25, 19-24, 19-23, 19-22, 19-21, 19-20, 20-30, 20-29, 20-28, 20-27, 20-26, 20-25, 20-24, 20-23, 20-22, 20-21, 21-30, 21-29, 21-28, 21-27, 21-26, 21-25, 21-24, 21-23, or 21-22 base pairs in length. In certain embodiments, the duplex region is 19-21 base pairs in length, e.g., 21 base pairs in length. Ranges and lengths intermediate to the above recited ranges and lengths are also contemplated to be part of the disclosure.


The two strands forming the duplex structure may be different portions of one larger RNA molecule, or they may be separate RNA molecules. Where the two strands are part of one larger molecule, and therefore are connected by an uninterrupted chain of nucleotides between the 3′-end of one strand and the 5′-end of the respective other strand forming the duplex structure, the connecting RNA chain is referred to as a “hairpin loop.” A hairpin loop can comprise at least one unpaired nucleotide. In some embodiments, the hairpin loop can comprise at least 4, 5, 6, 7, 8, 9, 10, 20, 23 or more unpaired nucleotides. In some embodiments, the hairpin loop can be 10 or fewer nucleotides. In some embodiments, the hairpin loop can be 8 or fewer unpaired nucleotides. In some embodiments, the hairpin loop can be 4-10 unpaired nucleotides. In some embodiments, the hairpin loop can be 4-8 nucleotides.


In certain embodiment, the two strands of double-stranded oligomeric compound can be linked together. The two strands can be linked to each other at both ends, or at one end only. By linking at one end is meant that 5′-end of first strand is linked to the 3′-end of the second strand or 3′-end of first strand is linked to 5′-end of the second strand. When the two strands are linked to each other at both ends, 5′-end of first strand is linked to 3′-end of second strand and 3′-end of first strand is linked to 5′-end of second strand. The two strands can be linked together by an oligonucleotide linker including, but not limited to, (N)n; wherein N is independently a modified or unmodified nucleotide and n is 3-23. In some embodiments, n is 3-10, e.g., 3, 4, 5, 6, 7, 8, 9, or 10. In some embodiments, the oligonucleotide linker is selected from the group consisting of GNRA, (G)4, (U)4, and (dT)4, wherein N is a modified or unmodified nucleotide and R is a modified or unmodified purine nucleotide. Some of the nucleotides in the linker can be involved in base-pair interactions with other nucleotides in the linker. The two strands can also be linked together by a non-nucleosidic linker, e.g. a linker described herein. It will be appreciated by one of skill in the art that any oligonucleotide chemical modifications or variations describe herein can be used in the oligonucleotide linker.


Hairpin and dumbbell type oligomeric compounds will have a duplex region equal to or at least 14, 15, 15, 16, 17, 18, 19, 29, 21, 22, 23, 24, or 25 nucleotide pairs. The duplex region can be equal to or less than 200, 100, or 50, in length. In some embodiments, ranges for the duplex region are 15-30, 17 to 23, 19 to 23, and 19 to 21 nucleotides pairs in length.


The hairpin oligomeric compounds can have a single strand overhang or terminal unpaired region, in some embodiments at the 3′, and in some embodiments on the antisense side of the hairpin. In some embodiments, the overhangs are 1-4, more generally 2-3 nucleotides in length. The hairpin oligomeric compounds that can induce RNA interference are also referred to as “shRNA” herein.


Where the two substantially complementary strands of a dsRNA are comprised by separate RNA molecules, those molecules need not be, but can be covalently connected. Where the two strands are connected covalently by means other than an uninterrupted chain of nucleotides between the 3′-end of one strand and the 5′-end of the respective other strand forming the duplex structure, the connecting structure is referred to as a “linker.” The RNA strands may have the same or a different number of nucleotides. The maximum number of base pairs is the number of nucleotides in the shortest strand of the dsRNA minus any overhangs that are present in the duplex. In addition to the duplex structure, an RNAi may comprise one or more nucleotide overhangs. In one embodiment of the RNAi agent, at least one strand comprises a 3′ overhang of at least 1 nucleotide. In another embodiment, at least one strand comprises a 3′ overhang of at least 2 nucleotides, e.g., 2, 3, 4, 5, 6, 7, 9, 10, 11, 12, 13, 14, or 15 nucleotides. In other embodiments, at least one strand of the RNAi agent comprises a 5′ overhang of at least 1 nucleotide. In certain embodiments, at least one strand comprises a 5′ overhang of at least 2 nucleotides, e.g., 2, 3, 4, 5, 6, 7, 9, 10, 11, 12, 13, 14, or 15 nucleotides. In still other embodiments, both the 3′ and the 5′ end of one strand of the RNAi agent comprise an overhang of at least 1 nucleotide.


In certain embodiments, an iRNA agent of the invention is a dsRNA, each strand of which comprises 19-23 nucleotides, that interacts with a target RNA sequence, e.g., a complement factor B (CFB) gene, to direct cleavage of the target RNA.


In some embodiments, an iRNA of the invention is a dsRNA of 24-30 nucleotides that interacts with a target RNA sequence, e.g., a CFB target mRNA sequence, to direct the cleavage of the target RNA.


As used herein, the term “nucleotide overhang” refers to at least one unpaired nucleotide that protrudes from the duplex structure of a double stranded iRNA. For example, when a 3′-end of one strand of a dsRNA extends beyond the 5′-end of the other strand, or vice versa, there is a nucleotide overhang. A dsRNA can comprise an overhang of at least one nucleotide; alternatively, the overhang can comprise at least two nucleotides, at least three nucleotides, at least four nucleotides, at least five nucleotides or more. A nucleotide overhang can comprise or consist of a nucleotide/nucleoside analog, including a deoxynucleotide/nucleoside. The overhang(s) can be on the sense strand, the antisense strand, or any combination thereof. Furthermore, the nucleotide(s) of an overhang can be present on the 5′-end, 3′-end, or both ends of either an antisense or sense strand of a dsRNA.


In one embodiment of the dsRNA, at least one strand comprises a 3′ overhang of at least 1 nucleotide. In another embodiment, at least one strand comprises a 3′ overhang of at least 2 nucleotides, e.g., 2, 3, 4, 5, 6, 7, 9, 10, 11, 12, 13, 14, or 15 nucleotides. In other embodiments, at least one strand of the RNAi agent comprises a 5′ overhang of at least 1 nucleotide. In certain embodiments, at least one strand comprises a 5′ overhang of at least 2 nucleotides, e.g., 2, 3, 4, 5, 6, 7, 9, 10, 11, 12, 13, 14, or 15 nucleotides. In still other embodiments, both the 3′ and the 5′ end of one strand of the RNAi agent comprise an overhang of at least 1 nucleotide.


In one embodiment, the antisense strand of a dsRNA has a 1-10 nucleotide, e.g., a 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 nucleotide, overhang at the 3′-end or the 5′-end. In one embodiment, the sense strand of a dsRNA has a 1-10 nucleotide, e.g., a 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 nucleotide, overhang at the 3′-end or the 5′-end. In another embodiment, one or more of the nucleotides in the overhang is replaced with a nucleoside thiophosphate.


In certain embodiments, the antisense strand of a dsRNA has a 1-10 nucleotides, e.g., a 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 nucleotide, overhang at the 3′-end or the 5′-end. In certain embodiments, the overhang on the sense strand or the antisense strand, or both, can include extended lengths longer than 10 nucleotides, e.g., 1-30 nucleotides, 2-30 nucleotides, 10-30 nucleotides, 10-25 nucleotides, 10-20 nucleotides, or 10-15 nucleotides in length. In certain embodiments, an extended overhang is on the sense strand of the duplex. In certain embodiments, an extended overhang is present on the 3′ end of the sense strand of the duplex. In certain embodiments, an extended overhang is present on the 5′ end of the sense strand of the duplex. In certain embodiments, an extended overhang is on the antisense strand of the duplex. In certain embodiments, an extended overhang is present on the 3′end of the antisense strand of the duplex. In certain embodiments, an extended overhang is present on the 5′end of the antisense strand of the duplex. In certain embodiments, one or more of the nucleotides in the extended overhang is replaced with a nucleoside thiophosphate. In certain embodiments, the overhang includes a self-complementary portion such that the overhang is capable of forming a hairpin structure that is stable under physiological conditions.


“Blunt” or “blunt end” means that there are no unpaired nucleotides at that end of the double stranded RNA agent, i.e., no nucleotide overhang. A “blunt ended” double stranded RNA agent is double stranded over its entire length, i.e., no nucleotide overhang at either end of the molecule. The RNAi agents of the invention include RNAi agents with no nucleotide overhang at one end (i.e., agents with one overhang and one blunt end) or with no nucleotide overhangs at either end. Most often such a molecule will be double-stranded over its entire length.


The term “antisense strand” or “guide strand” refers to the strand of an iRNA, e.g., a dsRNA, which includes a region that is substantially complementary to a target sequence, e.g., a CFB mRNA.


As used herein, the term “region of complementarity” refers to the region on the antisense strand that is substantially complementary to a sequence, for example a target sequence, e.g., a complement factor B nucleotide sequence, as defined herein. Where the region of complementarity is not fully complementary to the target sequence, the mismatches can be in the internal or terminal regions of the molecule. Generally, the most tolerated mismatches are in the terminal regions, e.g., within 5, 4, or 3 nucleotides of the 5′- or 3′-end of the iRNA. In some embodiments, a double stranded RNA agent of the invention includes a nucleotide mismatch in the antisense strand. In some embodiments, the antisense strand of the double stranded RNA agent of the invention includes no more than 4 mismatches with the target mRNA, e.g., the antisense strand includes 4, 3, 2, 1, or 0 mismatches with the target mRNA. In some embodiments, the antisense strand double stranded RNA agent of the invention includes no more than 4 mismatches with the sense strand, e.g., the antisense strand includes 4, 3, 2, 1, or 0 mismatches with the sense strand. In some embodiments, a double stranded RNA agent of the invention includes a nucleotide mismatch in the sense strand. In some embodiments, the sense strand of the double stranded RNA agent of the invention includes no more than 4 mismatches with the antisense strand, e.g., the sense strand includes 4, 3, 2, 1, or 0 mismatches with the antisense strand. In some embodiments, the nucleotide mismatch is, for example, within 5, 4, 3 nucleotides from the 3′-end of the iRNA. In another embodiment, the nucleotide mismatch is, for example, in the 3′-terminal nucleotide of the iRNA agent. In some embodiments, the mismatch(s) is not in the seed region.


Thus, an RNAi agent as described herein can contain one or more mismatches to the target sequence. In one embodiment, a RNAi agent as described herein contains no more than 3 mismatches (i.e., 3, 2, 1, or 0 mismatches). In one embodiment, an RNAi agent as described herein contains no more than 2 mismatches. In one embodiment, an RNAi agent as described herein contains no more than 1 mismatch. In one embodiment, an RNAi agent as described herein contains 0 mismatches. In certain embodiments, if the antisense strand of the RNAi agent contains mismatches to the target sequence, the mismatch can optionally be restricted to be within the last 5 nucleotides from either the 5′- or 3′-end of the region of complementarity. For example, in such embodiments, for a 23 nucleotide RNAi agent, the strand which is complementary to a region of a CFB gene, generally does not contain any mismatch within the central 13 nucleotides. The methods described herein or methods known in the art can be used to determine whether an RNAi agent containing a mismatch to a target sequence is effective in inhibiting the expression of a CFB gene. Consideration of the efficacy of RNAi agents with mismatches in inhibiting expression of a CFB gene is important, especially if the particular region of complementarity in a CFB gene is known to have polymorphic sequence variation within the population.


The term “sense strand” or “passenger strand” as used herein, refers to the strand of an iRNA that includes a region that is substantially complementary to a region of the antisense strand as that term is defined herein.


As used herein, “substantially all of the nucleotides are modified” are largely but not wholly modified and can include not more than 5, 4, 3, 2, or 1 unmodified nucleotides.


As used herein, the term “cleavage region” refers to a region that is located immediately adjacent to the cleavage site. The cleavage site is the site on the target at which cleavage occurs. In some embodiments, the cleavage region comprises three bases on either end of, and immediately adjacent to, the cleavage site. In some embodiments, the cleavage region comprises two bases on either end of, and immediately adjacent to, the cleavage site. In some embodiments, the cleavage site specifically occurs at the site bound by nucleotides 10 and 11 of the antisense strand, and the cleavage region comprises nucleotides 11, 12 and 13.


As used herein, and unless otherwise indicated, the term “complementary,” when used to describe a first nucleotide sequence in relation to a second nucleotide sequence, refers to the ability of an oligonucleotide or polynucleotide comprising the first nucleotide sequence to hybridize and form a duplex structure under certain conditions with an oligonucleotide or polynucleotide comprising the second nucleotide sequence, as will be understood by the skilled person. Such conditions can be, for example, “stringent conditions”, where stringent conditions can include: 400 mM NaCl, 40 mM PIPES pH 6.4, 1 mM EDTA, 50° C. or 70° C. for 12-16 hours followed by washing (see, e.g., “Molecular Cloning: A Laboratory Manual, Sambrook, et al. (1989) Cold Spring Harbor Laboratory Press). Other conditions, such as physiologically relevant conditions as can be encountered inside an organism, can apply. The skilled person will be able to determine the set of conditions most appropriate for a test of complementarity of two sequences in accordance with the ultimate application of the hybridized nucleotides.


Complementary sequences within an iRNA, e.g., within a dsRNA as described herein, include base-pairing of the oligonucleotide or polynucleotide comprising a first nucleotide sequence to an oligonucleotide or polynucleotide comprising a second nucleotide sequence over the entire length of one or both nucleotide sequences. Such sequences can be referred to as “fully complementary” with respect to each other herein. However, where a first sequence is referred to as “substantially complementary” with respect to a second sequence herein, the two sequences can be fully complementary, or they can form one or more, but generally not more than 5, 4, 3, or 2 mismatched base pairs upon hybridization for a duplex up to 30 base pairs, while retaining the ability to hybridize under the conditions most relevant to their ultimate application, e.g., inhibition of gene expression, in vitro or in vivo. However, where two oligonucleotides are designed to form, upon hybridization, one or more single stranded overhangs, such overhangs shall not be regarded as mismatches with regard to the determination of complementarity. For example, a dsRNA comprising one oligonucleotide 21 nucleotides in length and another oligonucleotide 23 nucleotides in length, wherein the longer oligonucleotide comprises a sequence of 21 nucleotides that is fully complementary to the shorter oligonucleotide, can yet be referred to as “fully complementary” for the purposes described herein.


“Complementary” sequences, as used herein, can also include, or be formed entirely from, non-Watson-Crick base pairs or base pairs formed from non-natural and modified nucleotides, in so far as the above requirements with respect to their ability to hybridize are fulfilled. Such non-Watson-Crick base pairs include, but are not limited to, G:U Wobble or Hoogsteen base pairing.


The terms “complementary,” “fully complementary” and “substantially complementary” herein can be used with respect to the base matching between the sense strand and the antisense strand of a dsRNA, or between two oligonucleotides or polynucleotides, such as the antisense strand of a double stranded RNA agent and a target sequence, as will be understood from the context of their use.


As used herein, a polynucleotide that is “substantially complementary to at least part of” a messenger RNA (mRNA) refers to a polynucleotide that is substantially complementary to a contiguous portion of the mRNA of interest (e.g., an mRNA encoding a complement factor B gene). For example, a polynucleotide is complementary to at least a part of a complement factor B mRNA if the sequence is substantially complementary to a non-interrupted portion of an mRNA encoding a complement factor B gene.


Accordingly, in some embodiments, the antisense polynucleotides disclosed herein are fully complementary to the target CFB sequence.


In other embodiments, the antisense polynucleotides disclosed herein are substantially complementary to the target CFB sequence and comprise a contiguous nucleotide sequence which is at least 80% complementary over its entire length to the equivalent region of the nucleotide sequence of any one of SEQ ID NOs:1, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, 33 or 35, or a fragment of any one of SEQ ID NOs:1, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, 33 or 35, such as about 85%, about 90%, about 91%, about 92%, about 93%, about 94%, about 95%, about 96%, about 97%, about 98%, or about 99% complementary.


In some embodiments, the antisense polynucleotides disclosed herein are substantially complementary to a fragment of a target CFB sequence and comprise a contiguous nucleotide sequence which is at least 80% complementary over its entire length to a fragment of SEQ ID NO: 1 selected from the group of nucleotides 943-965; 788-810; 734-756; 1016-1038; 1013-1035; 1207-1229; 1149-1171; 574-596; 1207-1229 or 828-850 of SEQ ID NO: 1, such as about 85%, about 90%, about 95%, or fully complementary.


In some embodiments, the antisense polynucleotides disclosed herein are substantially complementary to a fragment of a target CFB sequence and comprise a contiguous nucleotide sequence which is at least 80% complementary over its entire length to a fragment of SEQ ID NO: 1 selected from the group of nucleotides 153-175; 202-224; 219-241; 254-276; 304-326; 321-343; 347-369; 402-424; 418-440; 447-469; 491-513; 528-550; 549-571; 566-588; 591-613; 792-814; 819-841; 967-989; 1042-1064; 1234-1256; 1250-1272; 1269-1291; 1335-1357; 1354-1376; 1372-1394; 1422-1444; 1496-1518; 1670-1692; 1716-1738; 1757-1779; 1774-1796; 1793-1815; 1844-1866; 1871-1893; 1909-1931; 1924-1947; 1947-1969; 2161-2183; 2310-2332; 2330-2352; 2355-2377; 2494-2516; and 2527-2549 of SEQ ID NO: 1, such as about 85%, about 90%, about 95%, or fully complementary.


In other embodiments, the antisense polynucleotides disclosed herein are substantially complementary to the target CFB sequence and comprise a contiguous nucleotide sequence which is at least about 80% complementary over its entire length to any one of the sense strand nucleotide sequences in any one of any one of Tables 2-7, 13, 16, 19 and 20, or a fragment of any one of the sense strand nucleotide sequences in any one of Tables 2-7, 13, 16, 19 and 20, such as about 85%, about 90%, about 95%, or fully complementary.


In one embodiment, an RNAi agent of the disclosure includes a sense strand that is substantially complementary to an antisense polynucleotide which, in turn, is the same as a target CFB sequence, and wherein the sense strand polynucleotide comprises a contiguous nucleotide sequence which is at least about 80% complementary over its entire length to the equivalent region of the nucleotide sequence of SEQ ID NOs: 2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, 34 or 36, or a fragment of any one of SEQ ID NOs:2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, 34 or 36, such as about 85%, about 90%, about 95%, or fully complementary.


In some embodiments, an iRNA of the invention includes a sense strand that is substantially complementary to an antisense polynucleotide which, in turn, is complementary to a target complement factor B sequence, and wherein the sense strand polynucleotide comprises a contiguous nucleotide sequence which is at least about 80% complementary over its entire length to any one of the antisense strand nucleotide sequences in any one of any one of Tables 2-7, 13, 16, 19 and 20, or a fragment of any one of the antisense strand nucleotide sequences in any one of Tables 2-7, 13, 16, 19 and 20, such as about 85%, about 90%, about 95%, or fully complementary. In certain embodiments, the sense and antisense strands are selected from any one of the chemically modified duplexes AD-560018, AD-559375, AD-559160, AD-559374, AD-559060, AD-559721, AD-559026, AD-558225, AD-557069, AD-558068, AD-557422, AD-558063, AD-558066, AD-556701, AD-558657, AD-559020, AD-559023, AD-558860, AD-560019, AD-560016, AD-559008, AD-559717, AD-557072, AD-558097, AD-557774, AD-557070, AD-558065, AD-557853, AD-557079.


In certain embodiments, the sense and antisense strands are selected from any one of the chemically modified duplexes AD-560132.1; AD-560099.1; AD-559998.1; AD-559993.1; AD-559973.1; AD-559882.1; AD-559706.1; AD-559704.1; AD-559688.1; AD-559668.1; AD-559641.1; AD-559609.1; AD-559590.1; AD-559573.1; AD-559532.1; AD-559486.1; AD-559330.1; AD-559274.1; AD-559226.1; AD-559208.1; AD-559189.1; AD-559124.1; AD-559105.1; AD-559089.1; AD-558935.1; AD-558879.1; AD-558777.1; AD-558750.1; AD-558637.1; AD-558612.1; AD-558595.1; AD-558574.1; AD-558555.1; AD-558511.1; AD-558482.1; AD-558466.1; AD-558450.1; AD-558424.1; AD-558407.1; AD-558393.1; AD-558378.1; AD-558361.1; AD-558312.1 In some embodiments, the double-stranded region of a double-stranded iRNA agent is equal to or at least, 17, 18, 19, 20, 21, 22, 23, 23, 24, 25, 26, 27, 28, 29, 30 or more nucleotide pairs in length.


In some embodiments, the antisense strand of a double-stranded iRNA agent is equal to or at least 17, 18, 19, 20, 21, 22, 23, 23, 24, 25, 26, 27, 28, 29, or 30 nucleotides in length.


In some embodiments, the sense strand of a double-stranded iRNA agent is equal to or at least 17, 18, 19, 20, 21, 22, 23, 23, 24, 25, 26, 27, 28, 29, or 30 nucleotides in length.


In one embodiment, the sense and antisense strands of the double-stranded iRNA agent are each 18 to 30 nucleotides in length.


In one embodiment, the sense and antisense strands of the double-stranded iRNA agent are each 19 to 25 nucleotides in length.


In one embodiment, the sense and antisense strands of the double-stranded iRNA agent are each 21 to 23 nucleotides in length.


In one embodiment, the sense strand of the iRNA agent is 21-nucleotides in length, and the antisense strand is 23-nucleotides in length, wherein the strands form a double-stranded region of 21 consecutive base pairs having a 2-nucleotide long single stranded overhangs at the 3-end.


In some embodiments, the majority of nucleotides of each strand are ribonucleotides, but as described in detail herein, each or both strands can also include one or more non-ribonucleotides, e.g., a deoxyribonucleotide or a modified nucleotide. In addition, an “iRNA” may include ribonucleotides with chemical modifications. Such modifications may include all types of modifications disclosed herein or known in the art. Any such modifications, as used in an iRNA molecule, are encompassed by “iRNA” for the purposes of this specification and claims.


In certain embodiments of the instant disclosure, inclusion of a deoxy-nucleotide if present within an RNAi agent can be considered to constitute a modified nucleotide.


In one embodiment, at least partial suppression of the expression of a CFB gene, is assessed by a reduction of the amount of CFB mRNA which can be isolated from or detected in a first cell or group of cells in which a CFB gene is transcribed and which has or have been treated such that the expression of a CFB gene is inhibited, as compared to a second cell or group of cells substantially identical to the first cell or group of cells but which has or have not been so treated (control cells). The degree of inhibition may be expressed in terms of:









(

mRNA


in


control


cells

)

-

(

mRNA


in


treated


cells

)



(

mRNA


in


control


cells

)



•100

%




The phrase “contacting a cell with an iRNA,” such as a dsRNA, as used herein, includes contacting a cell by any possible means. Contacting a cell with an iRNA includes contacting a cell in vitro with the iRNA or contacting a cell in vivo with the iRNA. The contacting may be done directly or indirectly. Thus, for example, the iRNA may be put into physical contact with the cell by the individual performing the method, or alternatively, the iRNA may be put into a situation that will permit or cause it to subsequently come into contact with the cell.


Contacting a cell in vitro may be done, for example, by incubating the cell with the iRNA. Contacting a cell in vivo may be done, for example, by injecting the iRNA into or near the tissue where the cell is located, or by injecting the iRNA into another area, e.g., the bloodstream or the subcutaneous space, such that the agent will subsequently reach the tissue where the cell to be contacted is located. For example, the iRNA may contain or be coupled to a ligand, e.g., GalNAc, that directs the iRNA to a site of interest, e.g., the liver. Combinations of in vitro and in vivo methods of contacting are also possible. For example, a cell may also be contacted in vitro with an iRNA and subsequently transplanted into a subject.


In certain embodiments, contacting a cell with an iRNA includes “introducing” or “delivering the iRNA into the cell” by facilitating or effecting uptake or absorption into the cell. Absorption or uptake of an iRNA can occur through unaided diffusion or active cellular processes, or by auxiliary agents or devices. Introducing an iRNA into a cell may be in vitro or in vivo. For example, for in vivo introduction, iRNA can be injected into a tissue site or administered systemically. In vitro introduction into a cell includes methods known in the art such as electroporation and lipofection. Further approaches are described herein below or are known in the art.


The term “lipid nanoparticle” or “LNP” is a vesicle comprising a lipid layer encapsulating a pharmaceutically active molecule, such as a nucleic acid molecule, e.g., an iRNA or a plasmid from which an iRNA is transcribed. LNPs are described in, for example, U.S. Pat. Nos. 6,858,225, 6,815,432, 8,158,601, and 8,058,069, the entire contents of which are hereby incorporated herein by reference.


As used herein, a “subject” is an animal, such as a mammal, including a primate (such as a human, a non-human primate, e.g., a monkey, and a chimpanzee), a non-primate (such as a rabbit, a sheep, a hamster, a guinea pig, a dog, a rat, or a mouse), or a bird that expresses the target gene, either endogenously or heterologously. In an embodiment, the subject is a human, such as a human being treated or assessed for a disease or disorder that would benefit from reduction in CFB expression; a human at risk for a disease or disorder that would benefit from reduction in CFB expression; a human having a disease or disorder that would benefit from reduction in CFB expression; or human being treated for a disease or disorder that would benefit from reduction in CFB expression as described herein. In some embodiments, the subject is a female human. In other embodiments, the subject is a male human. In one embodiment, the subject is an adult subject. In another embodiment, the subject is a pediatric subject.


As used herein, the terms “treating” or “treatment” refer to a beneficial or desired result, such as reducing at least one sign or symptom of a CFB-associated disorder in a subject. Treatment also includes a reduction of one or more sign or symptoms associated with unwanted CFB expression; diminishing the extent of unwanted CFB activation or stabilization; amelioration or palliation of unwanted CFB activation or stabilization. “Treatment” can also mean prolonging survival as compared to expected survival in the absence of treatment.


The term “lower” in the context of the level of CFB in a subject or a disease marker or symptom refers to a statistically significant decrease in such level. The decrease can be, for example, at least 10%, 15%, 20%, 25%, 30%, %, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or more. In certain embodiments, a decrease is at least 20%. In certain embodiments, the decrease is at least 50% in a disease marker, e.g., protein or gene expression level. “Lower” in the context of the level of CFB in a subject is a decrease to a level accepted as within the range of normal for an individual without such disorder. In certain embodiments, the expression of the target is normalized, i.e., decreased towards or to a level accepted as within the range of normal for an individual without such disorder, e.g., normalization of body weight, blood pressure, or a serum lipid level. As used here, “lower” in a subject can refer to lowering of gene expression or protein production in a cell in a subject does not require lowering of expression in all cells or tissues of a subject. For example, as used herein, lowering in a subject can include lowering of gene expression or protein production in the liver of a subject.


The term “lower” can also be used in association with normalizing a symptom of a disease or condition, i.e. decreasing the difference between a level in a subject suffering from a CFB-associated disease towards or to a level in a normal subject not suffering from a CFB-associated disease. For example, if a subject with a normal weight of 70 kg weighs 90 kg prior to treatment (20 kg overweight) and 80 kg after treatment (10 kg overweight), the subject's weight is lowered towards a normal weight by 50% (10/20×100%). Similarly, if the HDL level of a woman is increased from 50 mg/dL (poor) to 57 mg/dL, with a normal level being 60 mg/dL, the difference between the prior level of the subject and the normal level is decreased by 70% (difference of 10 mg/dL between subject level and normal is decreased by 7 mg/dL, 7/10×100%). As used herein, if a disease is associated with an elevated value for a symptom, “normal” is considered to be the upper limit of normal. If a disease is associated with a decreased value for a symptom, “normal” is considered to be the lower limit of normal.


As used herein, “prevention” or “preventing,” when used in reference to a disease, disorder or condition thereof, that would benefit from a reduction in expression of a CFB gene or production of CFB protein, refers to preventing a subject who has at least one sign or symptom of a disease from developing further signs and symptoms thereby meeting the diagnostic criteria for that disease. In certain embodiments, prevention includes delayed progression to meeting the diagnostic criteria of the disease by days, weeks, months or years as compared to what would be predicted by natural history studies or the typical progression of the disease.


As used herein, the term “complement factor B disease” or “CFB-associated disease,” is a disease or disorder that is caused by, or associated with, complement activation. The term “CFB-associated disease” includes a disease, disorder or condition that would benefit from a decrease in CFB gene expression, replication, or protein activity. Non-limiting examples of CFB-associated diseases include, for example, paroxysmal nocturnal hemoglobinuria (PNH), atypical hemolytic uremic syndrome (aHUS), asthma, rheumatoid arthritis (RA); antiphospholipid antibody syndrome; lupus nephritis; ischemia-reperfusion injury; typical or infectious hemolytic uremic syndrome (tHUS); dense deposit disease (DDD); neuromyelitis optica (NMO); multifocal motor neuropathy (MMN); multiple sclerosis (MS); macular degeneration (e.g., age-related macular degeneration (AMD)); hemolysis, elevated liver enzymes, and low platelets (HELLP) syndrome; thrombotic thrombocytopenic purpura (TTP); spontaneous fetal loss; Pauci-immune vasculitis; epidermolysis bullosa; recurrent fetal loss; pre-eclampsia, traumatic brain injury, myasthenia gravis, cold agglutinin disease, dermatomyositis bullous pemphigoid, Shiga toxin E. coli-related hemolytic uremic syndrome, C3 neuropathy, anti-neutrophil cytoplasmic antibody-associated vasculitis (e.g., granulomatosis with polyangiitis (previously known as Wegener granulomatosis), Churg-Strauss syndrome, and microscopic polyangiitis), humoral and vascular transplant rejection, graft dysfunction, myocardial infarction (e.g., tissue damage and ischemia in myocardial infarction), an allogenic transplant, sepsis (e.g., poor outcome in sepsis), Coronary artery disease, dermatomyositis, Graves' disease, atherosclerosis, Alzheimer's disease, systemic inflammatory response sepsis, septic shock, spinal cord injury, glomerulonephritis, Hashimoto's thyroiditis, type I diabetes, psoriasis, pemphigus, autoimmune hemolytic anemia (AIHA), ITP, Goodpasture syndrome, Degos disease, antiphospholipid syndrome (APS), catastrophic APS (CAPS), a cardiovascular disorder, myocarditis, a cerebrovascular disorder, a peripheral (e.g., musculoskeletal) vascular disorder, a renovascular disorder, a mesenteric/enteric vascular disorder, vasculitis, Henoch-Schönlein purpura nephritis, systemic lupus erythematosus-associated vasculitis, vasculitis associated with rheumatoid arthritis, immune complex vasculitis, Takayasu's disease, dilated cardiomyopathy, diabetic angiopathy, Kawasaki's disease (arteritis), venous gas embolus (VGE), and restenosis following stent placement, rotational atherectomy, and percutaneous transluminal coronary angioplasty (PTCA) (see, e.g., Holers (2008) Immunological Reviews 223:300-316; Holers and Thurman (2004) Molecular Immunology 41:147-152; U.S. Patent Publication No. 20070172483).


In one embodiment, the complement factor B-associate disease is selected from the group consisting of C3 glomerulopathy, systemic lupus erythematosus (SLE), e.g., Lupus Nephritis, IgA nephropathy, diabetic nephropathy, polycystic kidney disease, membranous nephropathy, age-related macular degeneration, atypical hemolytic uremic syndrome, thrombotic microangiopathy, myasthenia gravis, ischemia and reperfusion injury, paroxysmal nocturnal hemoglobinuria, and rheumatoid arthritis


In another embodiment, the complement factor B-associate disease is selected from the group consisting of C3 glomerulopathy, systemic lupus erythematosus (SLE), e.g., Lupus Nephritis, IgA nephropathy, diabetic nephropathy, and polycystic kidney disease.


Further details regarding signs and symptoms of the various diseases or conditions are provided herein and are well known in the art.


“Therapeutically effective amount,” as used herein, is intended to include the amount of an RNAi agent that, when administered to a subject having a CFB-associated disease, is sufficient to effect treatment of the disease (e.g., by diminishing, ameliorating, or maintaining the existing disease or one or more symptoms of disease). The “therapeutically effective amount” may vary depending on the RNAi agent, how the agent is administered, the disease and its severity and the history, age, weight, family history, genetic makeup, the types of preceding or concomitant treatments, if any, and other individual characteristics of the subject to be treated.


“Prophylactically effective amount,” as used herein, is intended to include the amount of an RNAi agent that, when administered to a subject having at least one sign or symptom of a CFB-associated disorder, is sufficient to prevent or delay the subject's progression to meeting the full diagnostic criteria of the disease. Prevention of the disease includes slowing the course of progression to full blown disease. The “prophylactically effective amount” may vary depending on the RNAi agent, how the agent is administered, the degree of risk of disease, and the history, age, weight, family history, genetic makeup, the types of preceding or concomitant treatments, if any, and other individual characteristics of the patient to be treated.


A “therapeutically-effective amount” or “prophylactically effective amount” also includes an amount of an RNAi agent that produces some desired effect at a reasonable benefit/risk ratio applicable to any treatment. The iRNA employed in the methods of the present invention may be administered in a sufficient amount to produce a reasonable benefit/risk ratio applicable to such treatment.


The phrase “pharmaceutically acceptable” is employed herein to refer to those compounds, materials, compositions, or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human subjects and animal subjects without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.


The phrase “pharmaceutically-acceptable carrier” as used herein means a pharmaceutically-acceptable material, composition, or vehicle, such as a liquid or solid filler, diluent, excipient, manufacturing aid (e.g., lubricant, talc magnesium, calcium or zinc stearate, or steric acid), or solvent encapsulating material, involved in carrying or transporting the subject compound from one organ, or portion of the body, to another organ, or portion of the body. Each carrier must be “acceptable” in the sense of being compatible with the other ingredients of the formulation and not injurious to the subject being treated. Such carriers are known in the art. Pharmaceutically acceptable carriers include carriers for administration by injection.


The term “sample,” as used herein, includes a collection of similar fluids, cells, or tissues isolated from a subject, as well as fluids, cells, or tissues present within a subject. Examples of biological fluids include blood, serum and serosal fluids, plasma, cerebrospinal fluid, ocular fluids, lymph, urine, saliva, and the like. Tissue samples may include samples from tissues, organs, or localized regions. For example, samples may be derived from particular organs, parts of organs, or fluids or cells within those organs. In certain embodiments, samples may be derived from the liver (e.g., whole liver or certain segments of liver or certain types of cells in the liver, such as, e.g., hepatocytes). In some embodiments, a “sample derived from a subject” refers to urine obtained from the subject. A “sample derived from a subject” can refer to blood or blood derived serum or plasma from the subject.


II. iRNAs of the Invention

The present invention provides iRNAs which inhibit the expression of a complement factor B gene. In certain embodiments, the iRNA includes double stranded ribonucleic acid (dsRNA) molecules for inhibiting the expression of a CFB gene in a cell, such as a cell within a subject, e.g., a mammal, such as a human susceptible to developing a complement factor B-associated disorder. The dsRNAi agent includes an antisense strand having a region of complementarity which is complementary to at least a part of an mRNA formed in the expression of a CFB gene. The region of complementarity is about 19-30 nucleotides in length (e.g., about 30, 29, 28, 27, 26, 25, 24, 23, 22, 21, 20, or 19 nucleotides in length). Upon contact with a cell expressing the CFB gene, the iRNA inhibits the expression of the CFB gene (e.g., a human, a primate, a non-primate, or a rat CFB gene) by at least about 50% as assayed by, for example, a PCR or branched DNA (bDNA)-based method, or by a protein-based method, such as by immunofluorescence analysis, using, for example, western blotting or flow cytometric techniques. In certain embodiments, inhibition of expression is determined by the qPCR method provided in the examples herein with the siRNA at, e.g., a 10 nM concentration, in an appropriate organism cell line provided therein. In certain embodiments, inhibition of expression in vivo is determined by knockdown of the human gene in a rodent expressing the human gene, e.g., a mouse or an AAV-infected mouse expressing the human target gene, e.g., when administered as single dose, e.g., at 3 mg/kg at the nadir of RNA expression.


A dsRNA includes two RNA strands that are complementary and hybridize to form a duplex structure under conditions in which the dsRNA will be used. One strand of a dsRNA (the antisense strand) includes a region of complementarity that is substantially complementary, and generally fully complementary, to a target sequence. The target sequence can be derived from the sequence of an mRNA formed during the expression of a CFB gene. The other strand (the sense strand) includes a region that is complementary to the antisense strand, such that the two strands hybridize and form a duplex structure when combined under suitable conditions. As described elsewhere herein and as known in the art, the complementary sequences of a dsRNA can also be contained as self-complementary regions of a single nucleic acid molecule, as opposed to being on separate oligonucleotides.


Generally, the duplex structure is 15 to 30 base pairs in length, e.g., 15-29, 15-28, 15-27, 15-26, 15-25, 15-24, 15-23, 15-22, 15-21, 15-20, 15-19, 15-18, 15-17, 18-30, 18-29, 18-28, 18-27, 18-26, 18-25, 18-24, 18-23, 18-22, 18-21, 18-20, 19-30, 19-29, 19-28, 19-27, 19-26, 19-25, 19-24, 19-23, 19-22, 19-21, 19-20, 20-30, 20-29, 20-28, 20-27, 20-26, 20-25, 20-24, 20-23, 20-22, 20-21, 21-30, 21-29, 21-28, 21-27, 21-26, 21-25, 21-24, 21-23, or 21-22 base pairs in length. In certain embodiments, the duplex structure is 18 to 25 base pairs in length, e.g., 18-25, 18-24, 18-23, 18-22, 18-21, 18-20, 19-25, 19-24, 19-23, 19-22, 19-21, 19-20, 20-25, 20-24, 20-23, 20-22, 20-21, 21-25, 21-24, 21-23, 21-22, 22-25, 22-24, 22-23, 23-25, 23-24 or 24-25 base pairs in length, for example, 19-21 basepairs in length. Ranges and lengths intermediate to the above recited ranges and lengths are also contemplated to be part of the disclosure.


Similarly, the region of complementarity to the target sequence is 15 to 30 nucleotides in length, e.g., 15-29, 15-28, 15-27, 15-26, 15-25, 15-24, 15-23, 15-22, 15-21, 15-20, 15-19, 15-18, 15-17, 18-30, 18-29, 18-28, 18-27, 18-26, 18-25, 18-24, 18-23, 18-22, 18-21, 18-20, 19-30, 19-29, 19-28, 19-27, 19-26, 19-25, 19-24, 19-23, 19-22, 19-21, 19-20, 20-30, 20-29, 20-28, 20-27, 20-26, 20-25, 20-24,20-23, 20-22, 20-21, 21-30, 21-29, 21-28, 21-27, 21-26, 21-25, 21-24, 21-23, or 21-22 nucleotides in length, for example 19-23 nucleotides in length or 21-23 nucleotides in length. Ranges and lengths intermediate to the above recited ranges and lengths are also contemplated to be part of the disclosure.


In some embodiments, the duplex structure is 19 to 30 base pairs in length. Similarly, the region of complementarity to the target sequence is 19 to 30 nucleotides in length.


In some embodiments, the dsRNA is about 19 to about 23 nucleotides in length, or about 25 to about 30 nucleotides in length. In general, the dsRNA is long enough to serve as a substrate for the Dicer enzyme. For example, it is well-known in the art that dsRNAs longer than about 21-23 nucleotides in length may serve as substrates for Dicer. As the ordinarily skilled person will also recognize, the region of an RNA targeted for cleavage will most often be part of a larger RNA molecule, often an mRNA molecule. Where relevant, a “part” of an mRNA target is a contiguous sequence of an mRNA target of sufficient length to allow it to be a substrate for RNAi-directed cleavage (i.e., cleavage through a RISC pathway).


One of skill in the art will also recognize that the duplex region is a primary functional portion of a dsRNA, e.g., a duplex region of about 19 to about 30 base pairs, e.g., about 19-30, 19-29, 19-28, 19-27, 19-26, 19-25, 19-24, 19-23, 19-22, 19-21, 19-20, 20-30, 20-29, 20-28, 20-27, 20-26, 20-25, 20-24, 20-23, 20-22, 20-21, 21-30, 21-29, 21-28, 21-27, 21-26, 21-25, 21-24, 21-23, or 21-22 base pairs. Thus, in one embodiment, to the extent that it becomes processed to a functional duplex, of e.g., 15-30 base pairs, that targets a desired RNA for cleavage, an RNA molecule or complex of RNA molecules having a duplex region greater than 30 base pairs is a dsRNA. Thus, an ordinarily skilled artisan will recognize that in one embodiment, a miRNA is a dsRNA. In another embodiment, a dsRNA is not a naturally occurring miRNA. In another embodiment, an iRNA agent useful to target complement factor B gene expression is not generated in the target cell by cleavage of a larger dsRNA.


A dsRNA as described herein can further include one or more single-stranded nucleotide overhangs e.g., 1-4, 2-4, 1-3, 2-3, 1, 2, 3, or 4 nucleotides. dsRNAs having at least one nucleotide overhang can have superior inhibitory properties relative to their blunt-ended counterparts. A nucleotide overhang can comprise or consist of a nucleotide/nucleoside analog, including a deoxynucleotide/nucleoside. The overhang(s) can be on the sense strand, the antisense strand, or any combination thereof. Furthermore, the nucleotide(s) of an overhang can be present on the 5′-end, 3′-end, or both ends of an antisense or sense strand of a dsRNA.


A dsRNA can be synthesized by standard methods known in the art. Double stranded RNAi compounds of the invention may be prepared using a two-step procedure. First, the individual strands of the double stranded RNA molecule are prepared separately. Then, the component strands are annealed. The individual strands of the siRNA compound can be prepared using solution-phase or solid-phase organic synthesis or both. Organic synthesis offers the advantage that the oligonucleotide strands comprising unnatural or modified nucleotides can be easily prepared. Similarly, single-stranded oligonucleotides of the invention can be prepared using solution-phase or solid-phase organic synthesis or both.


Regardless of the method of synthesis, the siRNA preparation can be prepared in a solution (e.g., an aqueous or organic solution) that is appropriate for formulation. For example, the siRNA preparation can be precipitated and redissolved in pure double-distilled water, and lyophilized. The dried siRNA can then be resuspended in a solution appropriate for the intended formulation process.


In an aspect, a dsRNA of the invention includes at least two nucleotide sequences, a sense sequence and an anti-sense sequence. The sense strand is selected from the group of sequences provided in any one of Tables 2-7, 13, 16, 19 and 20, and the corresponding antisense strand of the sense strand is selected from the group of sequences of any one of Tables 2-7, 13, 16, 19 and 20. In this aspect, one of the two sequences is complementary to the other of the two sequences, with one of the sequences being substantially complementary to a sequence of an mRNA generated in the expression of a complement factor B gene. As such, in this aspect, a dsRNA will include two oligonucleotides, where one oligonucleotide is described as the sense strand in any one of Tables 2-7, 13, 16, 19 and 20, and the second oligonucleotide is described as the corresponding antisense strand of the sense strand in any one of Tables 2-7, 13, 16, 19 and 20.


In certain embodiments, the substantially complementary sequences of the dsRNA are contained on separate oligonucleotides. In other embodiments, the substantially complementary sequences of the dsRNA are contained on a single oligonucleotide.


In certain embodiments, the sense or antisense strand is selected from the sense or antisense strand of any one of duplexes AD-560018, AD-559375, AD-559160, AD-559374, AD-559060, AD-559721, AD-559026, AD-558225, AD-557069, AD-558068, AD-557422, AD-558063, AD-558066, AD-556701, AD-558657, AD-559020, AD-559023, AD-558860, AD-560019, AD-560016, AD-559008, AD-559717, AD-557072, AD-558097, AD-557774, AD-557070, AD-558065, AD-557853, or AD-557079.


It will be understood that, although the sequences in Tables 2, 4, 6 and 19 are not described as modified or conjugated sequences, the RNA of the iRNA of the invention e.g., a dsRNA of the invention, may comprise any one of the sequences set forth in any one of Tables 2-7, 13, 16, 19 and 20 that is un-modified, un-conjugated, or modified or conjugated differently than described therein. In other words, the invention encompasses dsRNA of Tables 2-7, 13, 16, 19 and 20 which are un-modified, un-conjugated, modified, or conjugated, as described herein.


The skilled person is well aware that dsRNAs having a duplex structure of about 20 to 23 base pairs, e.g., 21, base pairs have been hailed as particularly effective in inducing RNA interference (Elbashir et al., EMBO 2001, 20:6877-6888). However, others have found that shorter or longer RNA duplex structures can also be effective (Chu and Rana (2007) RNA 14:1714-1719; Kim et al. (2005) Nat Biotech 23:222-226). In the embodiments described above, by virtue of the nature of the oligonucleotide sequences provided in any one of Tables 2-7, 13, 16, 19 and 20, dsRNAs described herein can include at least one strand of a length of minimally 21 nucleotides. It can be reasonably expected that shorter duplexes having any one of the sequences in any one of Tables 2-7, 13, 16, 19 and 20 minus only a few nucleotides on one or both ends can be similarly effective as compared to the dsRNAs described above. Hence, dsRNAs having a sequence of at least 19, 20, or more contiguous nucleotides derived from any one of the sequences of any one of Tables 2-7, 13, 16, 19 and 20, and differing in their ability to inhibit the expression of a complement factor B gene by not more than about 5, 10, 15, 20, 25, or 30% inhibition from a dsRNA comprising the full sequence, are contemplated to be within the scope of the present invention.


In addition, the RNAs provided in Tables 2-7, 13, 16, 19 and 20 identify a site(s) in a complement factor B transcript that is susceptible to RISC-mediated cleavage. As such, the present invention further features iRNAs that target within one of these sites. As used herein, an iRNA is said to target within a particular site of an RNA transcript if the iRNA promotes cleavage of the transcript anywhere within that particular site. Such an iRNA will generally include at least about 19 contiguous nucleotides from any one of the sequences provided in any one of Tables 2-7, 13, 16, 19 and 20 coupled to additional nucleotide sequences taken from the region contiguous to the selected sequence in a complement factor B gene.


An RNAi agent as described herein can contain one or more mismatches to the target sequence. In one embodiment, an RNAi agent as described herein contains no more than 3 mismatches (i.e., 3, 2, 1, or 0 mismatches). In one embodiment, an RNAi agent as described herein contains no more than 2 mismatches. In one embodiment, an RNAi agent as described herein contains no more than 1 mismatch. In one embodiment, an RNAi agent as described herein contains 0 mismatches. In certain embodiments, if the antisense strand of the RNAi agent contains mismatches to the target sequence, the mismatch can optionally be restricted to be within the last 5 nucleotides from either the 5′- or 3′-end of the region of complementarity. For example, in such embodiments, for a 23 nucleotide RNAi agent, the strand which is complementary to a region of a CFB gene generally does not contain any mismatch within the central 13 nucleotides. The methods described herein or methods known in the art can be used to determine whether an RNAi agent containing a mismatch to a target sequence is effective in inhibiting the expression of a CFB gene. Consideration of the efficacy of RNAi agents with mismatches in inhibiting expression of a CFB gene is important, especially if the particular region of complementarity in a CFB gene is known to have polymorphic sequence variation within the population.


III. Modified iRNAs of the Invention

In certain embodiments, the RNA of the iRNA of the invention e.g., a dsRNA, is un-modified, and does not comprise, e.g., chemical modifications or conjugations known in the art and described herein. In other embodiments, the RNA of an iRNA of the invention, e.g., a dsRNA, is chemically modified to enhance stability or other beneficial characteristics. In certain embodiments of the invention, substantially all of the nucleotides of an iRNA of the invention are modified. In other embodiments of the invention, all of the nucleotides of an iRNA or substantially all of the nucleotides of an iRNA are modified, i.e., not more than 5, 4, 3, 2, or 1 unmodified nucleotides are present in a strand of the iRNA.


The nucleic acids featured in the invention can be synthesized or modified by methods well established in the art, such as those described in “Current protocols in nucleic acid chemistry,” Beaucage, S. L. et al. (Edrs.), John Wiley & Sons, Inc., New York, NY, USA, which is hereby incorporated herein by reference. Modifications include, for example, end modifications, e.g., 5′-end modifications (phosphorylation, conjugation, inverted linkages) or 3′-end modifications (conjugation, DNA nucleotides, inverted linkages, etc.); base modifications, e.g., replacement with stabilizing bases, destabilizing bases, or bases that base pair with an expanded repertoire of partners, removal of bases (abasic nucleotides), or conjugated bases; sugar modifications (e.g., at the 2′-position or 4′-position) or replacement of the sugar; or backbone modifications, including modification or replacement of the phosphodiester linkages. Specific examples of iRNA compounds useful in the embodiments described herein include, but are not limited to RNAs containing modified backbones or no natural internucleoside linkages. RNAs having modified backbones include, among others, those that do not have a phosphorus atom in the backbone. For the purposes of this specification, and as sometimes referenced in the art, modified RNAs that do not have a phosphorus atom in their internucleoside backbone can also be considered to be oligonucleosides. In some embodiments, a modified iRNA will have a phosphorus atom in its internucleoside backbone.


Modified RNA backbones include, for example, phosphorothioates, chiral phosphorothioates, phosphorodithioates, phosphotriesters, aminoalkylphosphotriesters, methyl and other alkyl phosphonates including 3′-alkylene phosphonates and chiral phosphonates, phosphinates, phosphoramidates including 3′-amino phosphoramidate and aminoalkylphosphoramidates, thionophosphoramidates, thionoalkylphosphonates, thionoalkylphosphotriesters, and boranophosphates having normal 3′-5′ linkages, 2′-5′-linked analogs of these, and those having inverted polarity wherein the adjacent pairs of nucleoside units are linked 3′-5′ to 5′-3′ or 2′-5′ to 5′-2′. Various salts, mixed salts and free acid forms are also included. In some embodiments of the invention, the dsRNA agents of the invention are in a free acid form. In other embodiments of the invention, the dsRNA agents of the invention are in a salt form. In one embodiment, the dsRNA agents of the invention are in a sodium salt form. In certain embodiments, when the dsRNA agents of the invention are in the sodium salt form, sodium ions are present in the agent as counterions for substantially all of the phosphodiester or phosphorothiotate groups present in the agent. Agents in which substantially all of the phosphodiester or phosphorothioate linkages have a sodium counterion include not more than 5, 4, 3, 2, or 1 phosphodiester or phosphorothioate linkages without a sodium counterion. In some embodiments, when the dsRNA agents of the invention are in the sodium salt form, sodium ions are present in the agent as counterions for all of the phosphodiester or phosphorothiotate groups present in the agent.


Representative U.S. patents that teach the preparation of the above phosphorus-containing linkages include, but are not limited to, U.S. Pat. Nos. 3,687,808; 4,469,863; 4,476,301; 5,023,243; 5,177,195; 5,188,897; 5,264,423; 5,276,019; 5,278,302; 5,286,717; 5,321,131; 5,399,676; 5,405,939; 5,453,496; 5,455,233; 5,466,677; 5,476,925; 5,519,126; 5,536,821; 5,541,316; 5,550,111; 5,563,253; 5,571,799; 5,587,361; 5,625,050; 6,028,188; 6,124,445; 6,160,109; 6,169,170; 6,172,209; 6,239,265; 6,277,603; 6,326,199; 6,346,614; 6,444,423; 6,531,590; 6,534,639; 6,608,035; 6,683,167; 6,858,715; 6,867,294; 6,878,805; 7,015,315; 7,041,816; 7,273,933; 7,321,029; and U.S. Pat. No. RE39,464, the entire contents of each of which are hereby incorporated herein by reference.


Modified RNA backbones that do not include a phosphorus atom therein have backbones that are formed by short chain alkyl or cycloalkyl internucleoside linkages, mixed heteroatoms and alkyl or cycloalkyl internucleoside linkages, or one or more short chain heteroatomic or heterocyclic internucleoside linkages. These include those having morpholino linkages (formed in part from the sugar portion of a nucleoside); siloxane backbones; sulfide, sulfoxide and sulfone backbones; formacetyl and thioformacetyl backbones; methylene formacetyl and thioformacetyl backbones; alkene containing backbones; sulfamate backbones; methyleneimino and methylenehydrazino backbones; sulfonate and sulfonamide backbones; amide backbones; and others having mixed N, O, S, and CH2 component parts.


Representative U.S. patents that teach the preparation of the above oligonucleosides include, but are not limited to, U.S. Pat. Nos. 5,034,506; 5,166,315; 5,185,444; 5,214,134; 5,216,141; 5,235,033; 5,64,562; 5,264,564; 5,405,938; 5,434,257; 5,466,677; 5,470,967; 5,489,677; 5,541,307; 5,561,225; 5,596,086; 5,602,240; 5,608,046; 5,610,289; 5,618,704; 5,623,070; 5,663,312; 5,633,360; 5,677,437; and 5,677,439, the entire contents of each of which are hereby incorporated herein by reference.


Suitable RNA mimetics are contemplated for use in iRNAs provided herein, in which both the sugar and the internucleoside linkage, i.e., the backbone, of the nucleotide units are replaced with novel groups. The base units are maintained for hybridization with an appropriate nucleic acid target compound. One such oligomeric compound in which an RNA mimetic that has been shown to have excellent hybridization properties is referred to as a peptide nucleic acid (PNA). In PNA compounds, the sugar backbone of an RNA is replaced with an amide containing backbone, in particular an aminoethylglycine backbone. The nucleobases are retained and are bound directly or indirectly to aza nitrogen atoms of the amide portion of the backbone. Representative US patents that teach the preparation of PNA compounds include, but are not limited to, U.S. Pat. Nos. 5,539,082; 5,714,331; and 5,719,262, the entire contents of each of which are hereby incorporated herein by reference. Additional PNA compounds suitable for use in the iRNAs of the invention are described in, for example, in Nielsen et al., Science, 1991, 254, 1497-1500.


Some embodiments featured in the invention include RNAs with phosphorothioate backbones and oligonucleosides with heteroatom backbones, and in particular —CH2—NH—CH2—, —CH2—N(CH3)—O—CH2— [known as a methylene (methylimino) or MMI backbone], —CH2—O—N(CH3)—CH2—, —CH2—N(CH3)—N(CH3)—CH2— and —N(CH3)—CH2—CH2— of the above-referenced U.S. Pat. No. 5,489,677, and the amide backbones of the above-referenced U.S. Pat. No. 5,602,240. In some embodiments, the RNAs featured herein have morpholino backbone structures of the above-referenced U.S. Pat. No. 5,034,506. The native phosphodiester backbone can be represented as O—P(O)(OH)—OCH2—.


Modified RNAs can also contain one or more substituted sugar moieties. The iRNAs, e.g., dsRNAs, featured herein can include one of the following at the 2′-position: OH; F; O-, S-, or N-alkyl; O-, S-, or N-alkenyl; O-, S- or N-alkynyl; or O-alkyl-O-alkyl, wherein the alkyl, alkenyl and alkynyl can be substituted or unsubstituted C1 to C10 alkyl or C2 to C10 alkenyl and alkynyl. Exemplary suitable modifications include O[(CH2)nO]mCH3, O(CH2)nOCH3, O(CH2)nNH2, O(CH2)nCH3, O(CH2)nONH2, and O(CH2)nON[(CH2)·CH3)]2, where n and m are from 1 to about 10. In other embodiments, dsRNAs include one of the following at the 2′ position: C1 to C10 lower alkyl, substituted lower alkyl, alkaryl, aralkyl, O-alkaryl or O-aralkyl, SH, SCH3, OCN, Cl, Br, CN, CF3, OCF3, SOCH3, SO2CH3, ONO2, NO2, N3, NH2, heterocycloalkyl, heterocycloalkaryl, aminoalkylamino, polyalkylamino, substituted silyl, an RNA cleaving group, a reporter group, an intercalator, a group for improving the pharmacokinetic properties of an iRNA, or a group for improving the pharmacodynamic properties of an iRNA, and other substituents having similar properties. In some embodiments, the modification includes a 2′-methoxyethoxy (2′-O—CH2CH2OCH3, also known as 2′-O-(2-methoxyethyl) or 2′-MOE) (Martin et al., Helv. Chim. Acta, 1995, 78:486-504) i.e., an alkoxy-alkoxy group. Another exemplary modification is 2′-dimethylaminooxyethoxy, i.e., a O(CH2)2ON(CH3)2 group, also known as 2′-DMAOE, as described in examples herein below, and 2′-dimethylaminoethoxyethoxy (also known in the art as 2′-O-dimethylaminoethoxyethyl or 2′-DMAEOE), i.e., 2′-O—CH2—O—CH2—N(CH2)2. Further exemplary modifications include: 5′-Me-2′-F nucleotides, 5′-Me-2′-OMe nucleotides, 5′-Me-2′-deoxynucleotides, (both R and S isomers in these three families); 2′-alkoxyalkyl; and 2′-NMA (N-methylacetamide).


Other modifications include 2′-methoxy (2′-OCH3), 2′-aminopropoxy (2′-OCH2CH2CH2NH2) and 2′-fluoro (2′-F). Similar modifications can also be made at other positions on the RNA of an iRNA, particularly the 3′ position of the sugar on the 3′ terminal nucleotide or in 2′-5′ linked dsRNAs and the 5′ position of 5′ terminal nucleotide. iRNAs can also have sugar mimetics such as cyclobutyl moieties in place of the pentofuranosyl sugar. Representative US patents that teach the preparation of such modified sugar structures include, but are not limited to, U.S. Pat. Nos. 4,981,957; 5,118,800; 5,319,080; 5,359,044; 5,393,878; 5,446,137; 5,466,786; 5,514,785; 5,519,134; 5,567,811; 5,576,427; 5,591,722; 5,597,909; 5,610,300; 5,627,053; 5,639,873; 5,646,265; 5,658,873; 5,670,633; and 5,700,920, certain of which are commonly owned with the instant application. The entire contents of each of the foregoing are hereby incorporated herein by reference.


An iRNA can also include nucleobase (often referred to in the art simply as “base”) modifications or substitutions. As used herein, “unmodified” or “natural” nucleobases include the purine bases adenine (A) and guanine (G), and the pyrimidine bases thymine (T), cytosine (C), and uracil (U). Modified nucleobases include other synthetic and natural nucleobases such as deoxythimidine (dT), 5-methylcytosine (5-me-C), 5-hydroxymethyl cytosine, xanthine, hypoxanthine, 2-aminoadenine, 6-methyl and other alkyl derivatives of adenine and guanine, 2-propyl and other alkyl derivatives of adenine and guanine, 2-thiouracil, 2-thiothymine and 2-thiocytosine, 5-halouracil and cytosine, 5-propynyl uracil and cytosine, 6-azo uracil, cytosine and thymine, 5-uracil (pseudouracil), 4-thiouracil, 8-halo, 8-amino, 8-thiol, 8-thioalkyl, 8-hydroxyl anal other 8-substituted adenines and guanines, 5-halo, particularly 5-bromo, 5-trifluoromethyl and other 5-substituted uracils and cytosines, 7-methylguanine and 7-methyladenine, 8-azaguanine and 8-azaadenine, 7-deazaguanine and 7-daazaadenine and 3-deazaguanine and 3-deazaadenine. Further nucleobases include those disclosed in U.S. Pat. No. 3,687,808, those disclosed in Modified Nucleosides in Biochemistry, Biotechnology and Medicine, Herdewijn, P. ed. Wiley-VCH, 2008; those disclosed in The Concise Encyclopedia Of Polymer Science And Engineering, pages 858-859, Kroschwitz, J. L, ed. John Wiley & Sons, 1990, these disclosed by Englisch et al., Angewandte Chemie, International Edition, 1991, 30, 613, and those disclosed by Sanghvi, Y S., Chapter 15, dsRNA Research and Applications, pages 289-302, Crooke, S. T. and Lebleu, B., Ed., CRC Press, 1993. Certain of these nucleobases are particularly useful for increasing the binding affinity of the oligomeric compounds featured in the invention. These include 5-substituted pyrimidines, 6-azapyrimidines and N-2, N-6 and 0-6 substituted purines, including 2-aminopropyladenine, 5-propynyluracil and 5-propynylcytosine. 5-methylcytosine substitutions have been shown to increase nucleic acid duplex stability by 0.6-1.2° C. (Sanghvi, Y. S., Crooke, S. T. and Lebleu, B., Eds., dsRNA Research and Applications, CRC Press, Boca Raton, 1993, pp. 276-278) and are exemplary base substitutions, even more particularly when combined with 2′-O-methoxyethyl sugar modifications.


Representative U.S. patents that teach the preparation of certain of the above noted modified nucleobases as well as other modified nucleobases include, but are not limited to, the above noted U.S. Pat. Nos. 3,687,808, 4,845,205; 5,130,30; 5,134,066; 5,175,273; 5,367,066; 5,432,272; 5,457,187; 5,459,255; 5,484,908; 5,502,177; 5,525,711; 5,552,540; 5,587,469; 5,594,121, 5,596,091; 5,614,617; 5,681,941; 5,750,692; 6,015,886; 6,147,200; 6,166,197; 6,222,025; 6,235,887; 6,380,368; 6,528,640; 6,639,062; 6,617,438; 7,045,610; 7,427,672; and 7,495,088, the entire contents of each of which are hereby incorporated herein by reference.


In some embodiments, an iRNA agent of the disclosure can also be modified to include one or more bicyclic sugar moieties. A “bicyclic sugar” is a furanosyl ring modified by the bridging of two atoms. A “bicyclic sugar” is a furanosyl ring modified by a ring formed by the bridging of two carbons, whether adjacent or non-adjacent atoms. A “bicyclic nucleoside” (“BNA”) is a nucleoside having a sugar moiety comprising a bridge a ring formed by bridging connecting two carbons, whether adjacent or non-adjacent, atoms of the sugar ring, thereby forming a bicyclic ring system. In certain embodiments, the bridge connects the 4′-carbon and the 2′-carbon of the sugar ring, optionally, via the 2′-acyclic oxygen atoms. Thus, in some embodiments an agent of the invention may include one or more locked nucleic acids (LNA). A locked nucleic acid is a nucleotide having a modified ribose moiety in which the ribose moiety comprises an extra bridge connecting the 2′ and 4′ carbons. In other words, an LNA is a nucleotide comprising a bicyclic sugar moiety comprising a 4′-CH2—O-2′ bridge. This structure effectively “locks” the ribose in the 3′-endo structural conformation. The addition of locked nucleic acids to siRNAs has been shown to increase siRNA stability in serum, and to reduce off-target effects (Elmen, J. et al., (2005) Nucleic Acids Research 33(1):439-447; Mook, O R. et al., (2007) Mol Canc Ther 6(3):833-843; Grunweller, A. et al., (2003) Nucleic Acids Research 31(12):3185-3193). Examples of bicyclic nucleosides for use in the polynucleotides of the invention include without limitation nucleosides comprising a bridge between the 4′ and the 2′ ribosyl ring atoms. In certain embodiments, the antisense polynucleotide agents of the invention include one or more bicyclic nucleosides comprising a 4′ to 2′ bridge.


A locked nucleoside can be represented by the structure (omitting stereochemistry),




embedded image




    • wherein B is a nucleobase or modified nucleobase and L is the linking group that joins the 2′-carbon to the 4′-carbon of the ribose ring.





Examples of such 4′ to 2′ bridged bicyclic nucleosides, include but are not limited to 4′-(CH2)—O-2′ (LNA); 4′-(CH2)—S-2′; 4′-(CH2)2—O-2′ (ENA); 4′-CH(CH3)—O-2′ (also referred to as “constrained ethyl” or “cEt”) and 4′-CH(CH2OCH3)—O-2′ (and analogs thereof; see, e.g., U.S. Pat. No. 7,399,845); 4′-C(CH3)(CH3)—O-2′ (and analogs thereof; see e.g., U.S. Pat. No. 8,278,283); 4′-CH2—N(OCH3)-2′ (and analogs thereof; see e.g., U.S. Pat. No. 8,278,425); 4′-CH2—O—N(CH3)-2′ (see, e.g., U.S. Patent Publication No. 2004/0171570); 4′-CH2—N(R)—O-2′, wherein R is H, C1-C12 alkyl, or a nitrogen protecting group (see, e.g., U.S. Pat. No. 7,427,672); 4′-CH2—C(H)(CH3)-2′ (see, e.g., Chattopadhyaya et al., J. Org. Chem., 2009, 74, 118-134); and 4′-CH2—C(═CH2)-2′ (and analogs thereof; see, e.g., U.S. Pat. No. 8,278,426). The entire contents of each of the foregoing are hereby incorporated herein by reference.


Additional representative U.S. patents and U.S. patenttent Publications that teach the preparation of locked nucleic acid nucleotides include, but are not limited to, the following: U.S. Pat. Nos. 6,268,490; 6,525,191; 6,670,461; 6,770,748; 6,794,499; 6,998,484; 7,053,207; 7,034,133; 7,084,125; 7,399,845; 7,427,672; 7,569,686; 7,741,457; 8,022,193; 8,030,467; 8,278,425; 8,278,426; 8,278,283; US 2008/0039618; and US 2009/0012281, the entire contents of each of which are hereby incorporated herein by reference.


Any of the foregoing bicyclic nucleosides can be prepared having one or more stereochemical sugar configurations including for example α-L-ribofuranose and β-D-ribofuranose (see WO 99/14226).


An iRNA agent of the disclosure can also be modified to include one or more constrained ethyl nucleotides. As used herein, a “constrained ethyl nucleotide” or “cEt” is a locked nucleic acid comprising a bicyclic sugar moiety comprising a 4′-CH(CH3)—O-2′ bridge (i.e., L in the preceding structure). In one embodiment, a constrained ethyl nucleotide is in the S conformation referred to herein as “S-cEt.”


An iRNA of the invention may also include one or more “conformationally restricted nucleotides” (“CRN”). CRN are nucleotide analogs with a linker connecting the C2′ and C4′ carbons of ribose or the CFB and —C5′ carbons of ribose. CRN lock the ribose ring into a stable conformation and increase the hybridization affinity to mRNA. The linker is of sufficient length to place the oxygen in an optimal position for stability and affinity resulting in less ribose ring puckering.


Representative publications that teach the preparation of certain of the above noted CRN include, but are not limited to, US2013/0190383; and WO2013/036868, the entire contents of each of which are hereby incorporated herein by reference.


In some embodiments, an iRNA of the invention comprises one or more monomers that are UNA (unlocked nucleic acid) nucleotides. UNA is unlocked acyclic nucleic acid, wherein any of the bonds of the sugar has been removed, forming an unlocked “sugar” residue. In one example, UNA also encompasses monomer with bonds between C1′-C4′ have been removed (i.e. the covalent carbon-oxygen-carbon bond between the C1′ and C4′ carbons). In another example, the C2′-C3′ bond (i.e. the covalent carbon-carbon bond between the C2′ and C3′ carbons) of the sugar has been removed (see Nuc. Acids Symp. Series, 52, 133-134 (2008) and Fluiter et al., Mol. Biosyst., 2009, 10, 1039 hereby incorporated by reference).


Representative U.S. publications that teach the preparation of UNA include, but are not limited to, U.S. Pat. No. 8,314,227; and US2013/0096289; US2013/0011922; and US2011/0313020, the entire contents of each of which are hereby incorporated herein by reference.


Potentially stabilizing modifications to the ends of RNA molecules can include N-(acetylaminocaproyl)-4-hydroxyprolinol (Hyp-C6-NHAc), N-(caproyl-4-hydroxyprolinol (Hyp-C6), N-(acetyl-4-hydroxyprolinol (Hyp-NHAc), thymidine-2′-O-deoxythymidine (ether), N-(aminocaproyl)-4-hydroxyprolinol (Hyp-C6-amino), 2-docosanoyl-uridine-3″-phosphate, inverted base dT(idT) and others. Disclosure of this modification can be found in WO2011/005861.


Other modifications of the nucleotides of an iRNA of the invention include a 5′ phosphate or 5′ phosphate mimic, e.g., a 5′-terminal phosphate or phosphate mimic on the antisense strand of an iRNA. Suitable phosphate mimics are disclosed in, for example US2012/0157511, the entire contents of which are incorporated herein by reference.


A. Modified iRNAs Comprising Motifs of the Invention


In certain aspects of the invention, the double stranded RNA agents of the invention include agents with chemical modifications as disclosed, for example, in WO2013/075035, the entire contents of each of which are incorporated herein by reference. As shown herein and in WO2013/075035, one or more motifs of three identical modifications on three consecutive nucleotides may be introduced into a sense strand or antisense strand of a dsRNAi agent, particularly at or near the cleavage site. In some embodiments, the sense strand and antisense strand of the dsRNAi agent may otherwise be completely modified. The introduction of these motifs interrupts the modification pattern, if present, of the sense or antisense strand. The dsRNAi agent may be optionally conjugated with a GalNAc derivative ligand, for instance on the sense strand.


More specifically, when the sense strand and antisense strand of the double stranded RNA agent are completely modified to have one or more motifs of three identical modifications on three consecutive nucleotides at or near the cleavage site of at least one strand of a dsRNAi agent, the gene silencing activity of the dsRNAi agent was observed.


Accordingly, the invention provides double stranded RNA agents capable of inhibiting the expression of a target gene (i.e., CFB gene) in vivo. The RNAi agent comprises a sense strand and an antisense strand. Each strand of the RNAi agent may be, for example, 17-30 nucleotides in length, 25-30 nucleotides in length, 27-30 nucleotides in length, 19-25 nucleotides in length, 19-23 nucleotides in length, 19-21 nucleotides in length, 21-25 nucleotides in length, or 21-23 nucleotides in length.


The sense strand and antisense strand typically form a duplex double stranded RNA (“dsRNA”), also referred to herein as “dsRNAi agent.” The duplex region of a dsRNAi agent may be, for example, the duplex region can be 27-30 nucleotide pairs in length, 19-25 nucleotide pairs in length, 19-23 nucleotide pairs in length, 19-21 nucleotide pairs in length, 21-25 nucleotide pairs in length, or 21-23 nucleotide pairs in length. In another example, the duplex region is selected from 19, 20, 21, 22, 23, 24, 25, 26, and 27 nucleotides in length.


In certain embodiments, the dsRNAi agent may contain one or more overhang regions or capping groups at the 3′-end, 5′-end, or both ends of one or both strands. The overhang can be, independently, 1-6 nucleotides in length, for instance 2-6 nucleotides in length, 1-5 nucleotides in length, 2-5 nucleotides in length, 1-4 nucleotides in length, 2-4 nucleotides in length, 1-3 nucleotides in length, 2-3 nucleotides in length, or 1-2 nucleotides in length. In certain embodiments, the overhang regions can include extended overhang regions as provided above. The overhangs can be the result of one strand being longer than the other, or the result of two strands of the same length being staggered. The overhang can form a mismatch with the target mRNA or it can be complementary to the gene sequences being targeted or can be another sequence. The first and second strands can also be joined, e.g., by additional bases to form a hairpin, or by other non-base linkers.


In certain embodiments, the nucleotides in the overhang region of the dsRNAi agent can each independently be a modified or unmodified nucleotide including, but no limited to 2′-sugar modified, such as, 2′-F, 2′-O-methyl, thymidine (T), 2′-O-methoxyethyl-5-methyluridine (Teo), 2′-O-methoxyethyladenosine (Aeo), 2′-O-methoxyethyl-5-methylcytidine (m5Ceo), and any combinations thereof.


For example, TT can be an overhang sequence for either end on either strand. The overhang can form a mismatch with the target mRNA or it can be complementary to the gene sequences being targeted or can be another sequence.


The 5′- or 3′-overhangs at the sense strand, antisense strand, or both strands of the dsRNAi agent may be phosphorylated. In some embodiments, the overhang region(s) contains two nucleotides having a phosphorothioate between the two nucleotides, where the two nucleotides can be the same or different. In some embodiments, the overhang is present at the 3′-end of the sense strand, antisense strand, or both strands. In some embodiments, this 3′-overhang is present in the antisense strand. In some embodiments, this 3′-overhang is present in the sense strand.


The RNAi agent may contain only a single overhang, which can strengthen the interference activity of the RNAi, without affecting its overall stability. For example, the single-stranded overhang may be located at the 3′-end of the sense strand or, alternatively, at the 3-end of the antisense strand. The RNAi may also have a blunt end, located at the 5′-end of the antisense strand (i.e., the 3′-end of the sense strand) or vice versa. Generally, the antisense strand of the dsRNAi agent has a nucleotide overhang at the 3′-end, and the 5′-end is blunt. While not wishing to be bound by theory, the asymmetric blunt end at the 5′-end of the antisense strand and 3′-end overhang of the antisense strand favor the guide strand loading into RISC process.


In certain embodiments, the dsRNAi agent is a double blunt-ended of 19 nucleotides in length, wherein the sense strand contains at least one motif of three 2′-F modifications on three consecutive nucleotides at positions 7, 8, and 9 from the 5′end. The antisense strand contains at least one motif of three 2′-O-methyl modifications on three consecutive nucleotides at positions 11, 12, and 13 from the 5′end.


In other embodiments, the dsRNAi agent is a double blunt-ended of 20 nucleotides in length, wherein the sense strand contains at least one motif of three 2′-F modifications on three consecutive nucleotides at positions 8, 9, and 10 from the 5′end. The antisense strand contains at least one motif of three 2′-O-methyl modifications on three consecutive nucleotides at positions 11, 12, and 13 from the 5′end.


In yet other embodiments, the dsRNAi agent is a double blunt-ended of 21 nucleotides in length, wherein the sense strand contains at least one motif of three 2′-F modifications on three consecutive nucleotides at positions 9, 10, and 11 from the 5′end. The antisense strand contains at least one motif of three 2′-O-methyl modifications on three consecutive nucleotides at positions 11, 12, and 13 from the 5′end.


In certain embodiments, the dsRNAi agent comprises a 21 nucleotide sense strand and a 23 nucleotide antisense strand, wherein the sense strand contains at least one motif of three 2′-F modifications on three consecutive nucleotides at positions 9, 10, and 11 from the 5′end; the antisense strand contains at least one motif of three 2′-O-methyl modifications on three consecutive nucleotides at positions 11, 12, and 13 from the 5′end, wherein one end of the RNAi agent is blunt, while the other end comprises a two nucleotide overhang. in one embodiment, the two nucleotide overhang is at the 3′-end of the antisense strand.


When the two nucleotide overhang is at the 3′-end of the antisense strand, there may be two phosphorothioate internucleotide linkages between the terminal three nucleotides, wherein two of the three nucleotides are the overhang nucleotides, and the third nucleotide is a paired nucleotide next to the overhang nucleotide. In one embodiment, the RNAi agent additionally has two phosphorothioate internucleotide linkages between the terminal three nucleotides at both the 5′-end of the sense strand and at the 5′-end of the antisense strand. In certain embodiments, every nucleotide in the sense strand and the antisense strand of the dsRNAi agent, including the nucleotides that are part of the motifs are modified nucleotides. In certain embodiments each residue is independently modified with a 2′-O-methyl or 2′-fluoro, e.g., in an alternating motif. Optionally, the dsRNAi agent further comprises a ligand (such as, GalNAc).


In certain embodiments, the dsRNAi agent comprises a sense and an antisense strand, wherein the sense strand is 25-30 nucleotide residues in length, wherein starting from the 5′ terminal nucleotide (position 1) positions 1 to 23 of the first strand comprise at least 8 ribonucleotides; the antisense strand is 36-66 nucleotide residues in length and, starting from the 3′ terminal nucleotide, comprises at least 8 ribonucleotides in the positions paired with positions 1-23 of sense strand to form a duplex; wherein at least the 3′ terminal nucleotide of antisense strand is unpaired with sense strand, and up to 6 consecutive 3′ terminal nucleotides are unpaired with sense strand, thereby forming a 3′ single stranded overhang of 1-6 nucleotides; wherein the 5′ terminus of antisense strand comprises from 10-30 consecutive nucleotides which are unpaired with sense strand, thereby forming a 10-30 nucleotide single stranded 5′ overhang; wherein at least the sense strand 5′ terminal and 3′ terminal nucleotides are base paired with nucleotides of antisense strand when sense and antisense strands are aligned for maximum complementarity, thereby forming a substantially duplexed region between sense and antisense strands; and antisense strand is sufficiently complementary to a target RNA along at least 19 ribonucleotides of antisense strand length to reduce target gene expression when the double stranded nucleic acid is introduced into a mammalian cell; and wherein the sense strand contains at least one motif of three 2′-F modifications on three consecutive nucleotides, where at least one of the motifs occurs at or near the cleavage site. The antisense strand contains at least one motif of three 2′-O-methyl modifications on three consecutive nucleotides at or near the cleavage site.


In certain embodiments, the dsRNAi agent comprises sense and antisense strands, wherein the dsRNAi agent comprises a first strand having a length which is at least 25 and at most 29 nucleotides and a second strand having a length which is at most 30 nucleotides with at least one motif of three 2′-O-methyl modifications on three consecutive nucleotides at position 11, 12, and 13 from the 5′ end; wherein the 3′ end of the first strand and the 5′ end of the second strand form a blunt end and the second strand is 1-4 nucleotides longer at its 3′ end than the first strand, wherein the duplex region which is at least 25 nucleotides in length, and the second strand is sufficiently complementary to a target mRNA along at least 19 nucleotide of the second strand length to reduce target gene expression when the RNAi agent is introduced into a mammalian cell, and wherein Dicer cleavage of the dsRNAi agent results in an siRNA comprising the 3′-end of the second strand, thereby reducing expression of the target gene in the mammal. Optionally, the dsRNAi agent further comprises a ligand.


In certain embodiments, the sense strand of the dsRNAi agent contains at least one motif of three identical modifications on three consecutive nucleotides, where one of the motifs occurs at the cleavage site in the sense strand.


In certain embodiments, the antisense strand of the dsRNAi agent can also contain at least one motif of three identical modifications on three consecutive nucleotides, where one of the motifs occurs at or near the cleavage site in the antisense strand.


For a dsRNAi agent having a duplex region of 19-23 nucleotides in length, the cleavage site of the antisense strand is typically around the 10, 11, and 12 positions from the 5′-end. Thus the motifs of three identical modifications may occur at the 9, 10, and 11 positions; the 10, 11, and 12 positions; the 11, 12, and 13 positions; the 12, 13, and 14 positions; or the 13, 14, and 15 positions of the antisense strand, the count starting from the first nucleotide from the 5′-end of the antisense strand, or, the count starting from the first paired nucleotide within the duplex region from the 5′-end of the antisense strand. The cleavage site in the antisense strand may also change according to the length of the duplex region of the dsRNAi agent from the 5′-end.


The sense strand of the dsRNAi agent may contain at least one motif of three identical modifications on three consecutive nucleotides at the cleavage site of the strand; and the antisense strand may have at least one motif of three identical modifications on three consecutive nucleotides at or near the cleavage site of the strand. When the sense strand and the antisense strand form a dsRNA duplex, the sense strand and the antisense strand can be so aligned that one motif of the three nucleotides on the sense strand and one motif of the three nucleotides on the antisense strand have at least one nucleotide overlap, i.e., at least one of the three nucleotides of the motif in the sense strand forms a base pair with at least one of the three nucleotides of the motif in the antisense strand. Alternatively, at least two nucleotides may overlap, or all three nucleotides may overlap.


In some embodiments, the sense strand of the dsRNAi agent may contain more than one motif of three identical modifications on three consecutive nucleotides. The first motif may occur at or near the cleavage site of the strand and the other motifs may be a wing modification. The term “wing modification” herein refers to a motif occurring at another portion of the strand that is separated from the motif at or near the cleavage site of the same strand. The wing modification is either adjacent to the first motif or is separated by at least one or more nucleotides. When the motifs are immediately adjacent to each other then the chemistries of the motifs are distinct from each other, and when the motifs are separated by one or more nucleotide than the chemistries can be the same or different. Two or more wing modifications may be present. For instance, when two wing modifications are present, each wing modification may occur at one end relative to the first motif which is at or near cleavage site or on either side of the lead motif.


Like the sense strand, the antisense strand of the dsRNAi agent may contain more than one motifs of three identical modifications on three consecutive nucleotides, with at least one of the motifs occurring at or near the cleavage site of the strand. This antisense strand may also contain one or more wing modifications in an alignment similar to the wing modifications that may be present on the sense strand.


In some embodiments, the wing modification on the sense strand or antisense strand of the dsRNAi agent typically does not include the first one or two terminal nucleotides at the 3′-end, 5′-end, or both ends of the strand.


In other embodiments, the wing modification on the sense strand or antisense strand of the dsRNAi agent typically does not include the first one or two paired nucleotides within the duplex region at the 3′-end, 5′-end, or both ends of the strand.


When the sense strand and the antisense strand of the dsRNAi agent each contain at least one wing modification, the wing modifications may fall on the same end of the duplex region, and have an overlap of one, two, or three nucleotides.


When the sense strand and the antisense strand of the dsRNAi agent each contain at least two wing modifications, the sense strand and the antisense strand can be so aligned that two modifications each from one strand fall on one end of the duplex region, having an overlap of one, two, or three nucleotides; two modifications each from one strand fall on the other end of the duplex region, having an overlap of one, two or three nucleotides; two modifications one strand fall on each side of the lead motif, having an overlap of one, two or three nucleotides in the duplex region.


In some embodiments, every nucleotide in the sense strand and antisense strand of the dsRNAi agent, including the nucleotides that are part of the motifs, may be modified. Each nucleotide may be modified with the same or different modification which can include one or more alteration of one or both of the non-linking phosphate oxygens or of one or more of the linking phosphate oxygens; alteration of a constituent of the ribose sugar, e.g., of the 2′-hydroxyl on the ribose sugar; wholesale replacement of the phosphate moiety with “dephospho” linkers; modification or replacement of a naturally occurring base; and replacement or modification of the ribose-phosphate backbone.


As nucleic acids are polymers of subunits, many of the modifications occur at a position which is repeated within a nucleic acid, e.g., a modification of a base, or a phosphate moiety, or a non-linking O of a phosphate moiety. In some cases the modification will occur at all of the subject positions in the nucleic acid but in many cases it will not. By way of example, a modification may only occur at a 3′- or 5′ terminal position, may only occur in a terminal region, e.g., at a position on a terminal nucleotide or in the last 2, 3, 4, 5, or 10 nucleotides of a strand. A modification may occur in a double strand region, a single strand region, or in both. A modification may occur only in the double strand region of an RNA or may only occur in a single strand region of a RNA. For example, a phosphorothioate modification at a non-linking O position may only occur at one or both termini, may only occur in a terminal region, e.g., at a position on a terminal nucleotide or in the last 2, 3, 4, 5, or 10 nucleotides of a strand, or may occur in double strand and single strand regions, particularly at termini. The 5′-end or ends can be phosphorylated.


It may be possible, e.g., to enhance stability, to include particular bases in overhangs, or to include modified nucleotides or nucleotide surrogates, in single strand overhangs, e.g., in a 5′- or 3′-overhang, or in both. For example, it can be desirable to include purine nucleotides in overhangs. In some embodiments all or some of the bases in a 3′- or 5′-overhang may be modified, e.g., with a modification described herein. Modifications can include, e.g., the use of modifications at the 2′ position of the ribose sugar with modifications that are known in the art, e.g., the use of deoxyribonucleotides, 2′-deoxy-2′-fluoro (2′-F) or 2′-O-methyl modified instead of the ribosugar of the nucleobase, and modifications in the phosphate group, e.g., phosphorothioate modifications. Overhangs need not be homologous with the target sequence.


In some embodiments, each residue of the sense strand and antisense strand is independently modified with LNA, CRN, cET, UNA, HNA, CeNA, 2′-methoxyethyl, 2′-O-methyl, 2′-O-allyl, 2′-C-allyl, 2′-deoxy, 2′-hydroxyl, or 2′-fluoro. The strands can contain more than one modification. In one embodiment, each residue of the sense strand and antisense strand is independently modified with 2′-O-methyl or 2′-fluoro.


At least two different modifications are typically present on the sense strand and antisense strand. Those two modifications may be the 2′-O-methyl or 2′-fluoro modifications, or others.


In certain embodiments, the Na or Nb comprise modifications of an alternating pattern. The term “alternating motif” as used herein refers to a motif having one or more modifications, each modification occurring on alternating nucleotides of one strand. The alternating nucleotide may refer to one per every other nucleotide or one per every three nucleotides, or a similar pattern. For example, if A, B and C each represent one type of modification to the nucleotide, the alternating motif can be “ABABABABABAB . . . ,” “AABBAABBAABB . . . ,” “AABAABAABAAB . . . ,” “AAABAAABAAAB . . . ,” “AAABBBAAABBB . . . ,” or “ABCABCABCABC . . . ,” etc.


The type of modifications contained in the alternating motif may be the same or different. For example, if A, B, C, D each represent one type of modification on the nucleotide, the alternating pattern, i.e., modifications on every other nucleotide, may be the same, but each of the sense strand or antisense strand can be selected from several possibilities of modifications within the alternating motif such as “ABABAB . . . ”, “ACACAC . . . ” “BDBDBD . . . ” or “CDCDCD . . . ,” etc.


In some embodiments, the dsRNAi agent of the invention comprises the modification pattern for the alternating motif on the sense strand relative to the modification pattern for the alternating motif on the antisense strand is shifted. The shift may be such that the modified group of nucleotides of the sense strand corresponds to a differently modified group of nucleotides of the antisense strand and vice versa. For example, the sense strand when paired with the antisense strand in the dsRNA duplex, the alternating motif in the sense strand may start with “ABABAB” from 5′ to 3′ of the strand and the alternating motif in the antisense strand may start with “BABABA” from 5′ to 3′ of the strand within the duplex region. As another example, the alternating motif in the sense strand may start with “AABBAABB” from 5′ to 3′ of the strand and the alternating motif in the antisense strand may start with “BBAABBAA” from 5′ to 3′ of the strand within the duplex region, so that there is a complete or partial shift of the modification patterns between the sense strand and the antisense strand.


In some embodiments, the dsRNAi agent comprises the pattern of the alternating motif of 2′-O-methyl modification and 2′-F modification on the sense strand initially has a shift relative to the pattern of the alternating motif of 2′-O-methyl modification and 2′-F modification on the antisense strand initially, i.e., the 2′-O-methyl modified nucleotide on the sense strand base pairs with a 2′-F modified nucleotide on the antisense strand and vice versa. The 1 position of the sense strand may start with the 2′-F modification, and the 1 position of the antisense strand may start with the 2′-O-methyl modification.


The introduction of one or more motifs of three identical modifications on three consecutive nucleotides to the sense strand or antisense strand interrupts the initial modification pattern present in the sense strand or antisense strand. This interruption of the modification pattern of the sense or antisense strand by introducing one or more motifs of three identical modifications on three consecutive nucleotides to the sense or antisense strand may enhance the gene silencing activity against the target gene.


In some embodiments, when the motif of three identical modifications on three consecutive nucleotides is introduced to any of the strands, the modification of the nucleotide next to the motif is a different modification than the modification of the motif. For example, the portion of the sequence containing the motif is “ . . . NaYYYNb . . . ,” where “Y” represents the modification of the motif of three identical modifications on three consecutive nucleotide, and “Na” and “Nb” represent a modification to the nucleotide next to the motif “YYY” that is different than the modification of Y, and where Na and Nb can be the same or different modifications. Alternatively, Na or Nb may be present or absent when there is a wing modification present.


The iRNA may further comprise at least one phosphorothioate or methylphosphonate internucleotide linkage. The phosphorothioate or methylphosphonate internucleotide linkage modification may occur on any nucleotide of the sense strand, antisense strand, or both strands in any position of the strand. For instance, the internucleotide linkage modification may occur on every nucleotide on the sense strand or antisense strand; each internucleotide linkage modification may occur in an alternating pattern on the sense strand or antisense strand; or the sense strand or antisense strand may contain both internucleotide linkage modifications in an alternating pattern. The alternating pattern of the internucleotide linkage modification on the sense strand may be the same or different from the antisense strand, and the alternating pattern of the internucleotide linkage modification on the sense strand may have a shift relative to the alternating pattern of the internucleotide linkage modification on the antisense strand. In one embodiment, a double-stranded RNAi agent comprises 6-8 phosphorothioate internucleotide linkages. In some embodiments, the antisense strand comprises two phosphorothioate internucleotide linkages at the 5′-end and two phosphorothioate internucleotide linkages at the 3′-end, and the sense strand comprises at least two phosphorothioate internucleotide linkages at either the 5′-end or the 3′-end.


In some embodiments, the dsRNAi agent comprises a phosphorothioate or methylphosphonate internucleotide linkage modification in the overhang region. For example, the overhang region may contain two nucleotides having a phosphorothioate or methylphosphonate internucleotide linkage between the two nucleotides. Internucleotide linkage modifications also may be made to link the overhang nucleotides with the terminal paired nucleotides within the duplex region. For example, at least 2, 3, 4, or all the overhang nucleotides may be linked through phosphorothioate or methylphosphonate internucleotide linkage, and optionally, there may be additional phosphorothioate or methylphosphonate internucleotide linkages linking the overhang nucleotide with a paired nucleotide that is next to the overhang nucleotide. For instance, there may be at least two phosphorothioate internucleotide linkages between the terminal three nucleotides, in which two of the three nucleotides are overhang nucleotides, and the third is a paired nucleotide next to the overhang nucleotide. These terminal three nucleotides may be at the 3′-end of the antisense strand, the 3′-end of the sense strand, the 5′-end of the antisense strand, or the 5′end of the antisense strand.


In some embodiments, the 2-nucleotide overhang is at the 3′-end of the antisense strand, and there are two phosphorothioate internucleotide linkages between the terminal three nucleotides, wherein two of the three nucleotides are the overhang nucleotides, and the third nucleotide is a paired nucleotide next to the overhang nucleotide. Optionally, the dsRNAi agent may additionally have two phosphorothioate internucleotide linkages between the terminal three nucleotides at both the 5′-end of the sense strand and at the 5′-end of the antisense strand.


In one embodiment, the dsRNAi agent comprises mismatch(es) with the target, within the duplex, or combinations thereof. The mismatch may occur in the overhang region or the duplex region. The base pair may be ranked on the basis of their propensity to promote dissociation or melting (e.g., on the free energy of association or dissociation of a particular pairing, the simplest approach is to examine the pairs on an individual pair basis, though next neighbor or similar analysis can also be used). In terms of promoting dissociation: A:U is preferred over G:C; G:U is preferred over G:C; and I:C is preferred over G:C (I=inosine). Mismatches, e.g., non-canonical or other than canonical pairings (as described elsewhere herein) are preferred over canonical (A:T, A:U, G:C) pairings; and pairings which include a universal base are preferred over canonical pairings.


In certain embodiments, the dsRNAi agent comprises at least one of the first 1, 2, 3, 4, or 5 base pairs within the duplex regions from the 5′-end of the antisense strand independently selected from the group of: A:U, G:U, I:C, and mismatched pairs, e.g., non-canonical or other than canonical pairings or pairings which include a universal base, to promote the dissociation of the antisense strand at the 5′-end of the duplex.


In certain embodiments, the nucleotide at the 1 position within the duplex region from the 5′-end in the antisense strand is selected from A, dA, dU, U, and dT. Alternatively, at least one of the first 1, 2, or 3 base pair within the duplex region from the 5′-end of the antisense strand is an AU base pair. For example, the first base pair within the duplex region from the 5′-end of the antisense strand is an AU base pair.


In other embodiments, the nucleotide at the 3′-end of the sense strand is deoxythimidine (dT) or the nucleotide at the 3′-end of the antisense strand is deoxythimidine (dT). For example, there is a short sequence of deoxythimidine nucleotides, for example, two dT nucleotides on the 3′-end of the sense, antisense strand, or both strands.


In certain embodiments, the sense strand sequence may be represented by formula (I):





5′ np-Na—(XXX)i—Nb—YYY—Nb—(ZZZ)j—Na-nq 3′  (I)

    • wherein:
    • i and j are each independently 0 or 1;
    • p and q are each independently 0-6;
    • each Na independently represents an oligonucleotide sequence comprising 0-25 modified nucleotides, each sequence comprising at least two differently modified nucleotides;
    • each Nb independently represents an oligonucleotide sequence comprising 0-10 modified nucleotides;
    • each np and nq independently represent an overhang nucleotide;
    • wherein Nb and Y do not have the same modification; and
    • XXX, YYY, and ZZZ each independently represent one motif of three identical modifications on three consecutive nucleotides. In one embodiment, YYY is all 2′-F modified nucleotides.


In some embodiments, the Na or Nb comprises modifications of alternating pattern.


In some embodiments, the YYY motif occurs at or near the cleavage site of the sense strand. For example, when the dsRNAi agent has a duplex region of 17-23 nucleotides in length, the YYY motif can occur at or the vicinity of the cleavage site (e.g.: can occur at positions 6, 7, 8; 7, 8, 9; 8, 9, 10; 9, 10, 11; 10, 11, 12; or 11, 12, 13) of the sense strand, the count starting from the first nucleotide, from the 5′-end; or optionally, the count starting at the first paired nucleotide within the duplex region, from the 5′-end.


In one embodiment, i is 1 and j is 0, or i is 0 and j is 1, or both i and j are 1. The sense strand can therefore be represented by the following formulas:





5′ np-Na—YYY—Nb—ZZZ—Na-nq 3′  (Ib);





5′ np-Na—XXX—Nb—YYY—Na-nq 3′  (Ic); or





5′ np-Na—XXX—Nb—YYY—Nb—ZZZ—Na-nq 3′  (Id).


When the sense strand is represented by formula (Ib), Nb represents an oligonucleotide sequence comprising 0-10, 0-7, 0-5, 0-4, 0-2, or 0 modified nucleotides. Each Na independently can represent an oligonucleotide sequence comprising 2-20, 2-15, or 2-10 modified nucleotides.


When the sense strand is represented as formula (Ic), Nb represents an oligonucleotide sequence comprising 0-10, 0-7, 0-10, 0-7, 0-5, 0-4, 0-2, or 0 modified nucleotides. Each Na can independently represent an oligonucleotide sequence comprising 2-20, 2-15, or 2-10 modified nucleotides.


When the sense strand is represented as formula (Id), each Nb independently represents an oligonucleotide sequence comprising 0-10, 0-7, 0-5, 0-4, 0-2, or 0 modified nucleotides. In one embodiment, Nb is 0, 1, 2, 3, 4, 5, or 6. Each Na can independently represent an oligonucleotide sequence comprising 2-20, 2-15, or 2-10 modified nucleotides.


Each of X, Y and Z may be the same or different from each other.


In other embodiments, i is 0 and j is 0, and the sense strand may be represented by the formula:





5′ np-Na—YYY—Na-nq 3′  (Ia).


When the sense strand is represented by formula (Ia), each Na independently can represent an oligonucleotide sequence comprising 2-20, 2-15, or 2-10 modified nucleotides.


In one embodiment, the antisense strand sequence of the RNAi may be represented by formula (II):





5′ nq′-Na′—(Z′Z′Z′)k—Nb′—Y′Y′Y′—Nb′—(X′X′X′)l—N′a-np′ 3′  (II)


wherein:

    • k and l are each independently 0 or 1;
    • p′ and q′ are each independently 0-6;
    • each Na′ independently represents an oligonucleotide sequence comprising 0-25 modified nucleotides, each sequence comprising at least two differently modified nucleotides;
    • each Nb′ independently represents an oligonucleotide sequence comprising 0-10 modified nucleotides;
    • each np′ and nq′ independently represent an overhang nucleotide;
    • wherein Nb′ and Y′ do not have the same modification; and
    • X′X′X′, Y′Y′Y′, and Z′Z′Z′ each independently represent one motif of three identical modifications on three consecutive nucleotides.


In some embodiments, the Na′ or Nb′ comprises modifications of alternating pattern.


The Y′Y′Y′ motif occurs at or near the cleavage site of the antisense strand. For example, when the dsRNAi agent has a duplex region of 17-23 nucleotides in length, the Y′Y′Y′ motif can occur at positions 9, 10, 11; 10, 11, 12; 11, 12, 13; 12, 13, 14; or 13, 14, 15 of the antisense strand, with the count starting from the first nucleotide, from the 5′-end; or optionally, the count starting at the first paired nucleotide within the duplex region, from the 5′-end. In one embodiment, the Y′Y′Y′ motif occurs at positions 11, 12, 13.


In certain embodiments, Y′Y′Y′ motif is all 2′-OMe modified nucleotides.


In certain embodiments, k is 1 and l is 0, or k is 0 and l is 1, or both k and l are 1.


The antisense strand can therefore be represented by the following formulas:





5′ nq′-Na′—Z′Z′Z′—Nb′—Y′Y′Y′—Na′-np′ 3′  (IIb);





5′ nq′-Na′—Y′Y′Y′—Nb′—X′X′X′-np′ 3′  (IIc); or





5′ nq′-Na′—Z′Z′Z′—Nb′—Y′Y′Y′—Nb′—X′X′X′—Na′-np′ 3′  (IId).


When the antisense strand is represented by formula (IIb), Nb′ represents an oligonucleotide sequence comprising 0-10, 0-7, 0-10, 0-7, 0-5, 0-4, 0-2, or 0 modified nucleotides. Each Na′ independently represents an oligonucleotide sequence comprising 2-20, 2-15, or 2-10 modified nucleotides.


When the antisense strand is represented as formula (IIc), Nb′ represents an oligonucleotide sequence comprising 0-10, 0-7, 0-10, 0-7, 0-5, 0-4, 0-2, or 0 modified nucleotides. Each Na′ independently represents an oligonucleotide sequence comprising 2-20, 2-15, or 2-10 modified nucleotides.


When the antisense strand is represented as formula (IId), each Nb′ independently represents an oligonucleotide sequence comprising 0-10, 0-7, 0-10, 0-7, 0-5, 0-4, 0-2, or 0 modified nucleotides. Each Na′ independently represents an oligonucleotide sequence comprising 2-20, 2-15, or 2-10 modified nucleotides. In one embodiment, Nb is 0, 1, 2, 3, 4, 5, or 6.


In other embodiments, k is 0 and 1 is 0 and the antisense strand may be represented by the formula:





5′ np′-Na′—Y′Y′Y′—Na′-nq′ 3′  (Ia).


When the antisense strand is represented as formula (IIa), each Na′ independently represents an oligonucleotide sequence comprising 2-20, 2-15, or 2-10 modified nucleotides.


Each of X′, Y′ and Z′ may be the same or different from each other.


Each nucleotide of the sense strand and antisense strand may be independently modified with LNA, CRN, UNA, cEt, glycol nucleic acid (GNA), hexitol nucleic acid (HNA) CeNA, 2′-methoxyethyl, 2′-O-methyl, 2′-O-allyl, 2′-C-allyl, 2′-hydroxyl, or 2′-fluoro. For example, each nucleotide of the sense strand and antisense strand is independently modified with 2′-O-methyl or 2′-fluoro. Each X, Y, Z, X′, Y′, and Z′, in particular, may represent a 2′-O-methyl modification or a 2′-fluoro modification.


In some embodiments, the sense strand of the dsRNAi agent may contain YYY motif occurring at 9, 10, and 11 positions of the strand when the duplex region is 21 nt, the count starting from the first nucleotide from the 5′-end, or optionally, the count starting at the first paired nucleotide within the duplex region, from the 5′-end; and Y represents 2′-F modification. The sense strand may additionally contain XXX motif or ZZZ motifs as wing modifications at the opposite end of the duplex region; and XXX and ZZZ each independently represents a 2′-OMe modification or 2′-F modification.


In some embodiments the antisense strand may contain Y′Y′Y′ motif occurring at positions 11, 12, 13 of the strand, the count starting from the first nucleotide from the 5′-end, or optionally, the count starting at the first paired nucleotide within the duplex region, from the 5′-end; and Y′ represents 2′-O-methyl modification. The antisense strand may additionally contain X′X′X′ motif or Z′Z′Z′ motifs as wing modifications at the opposite end of the duplex region; and X′X′X′ and Z′Z′Z′ each independently represents a 2′-OMe modification or 2′-F modification.


The sense strand represented by any one of the above formulas (Ia), (Ib), (Ic), and (Id) forms a duplex with an antisense strand being represented by any one of formulas (IIa), (IIb), (IIc), and (IId), respectively.


Accordingly, the dsRNAi agents for use in the methods of the invention may comprise a sense strand and an antisense strand, each strand having 14 to 30 nucleotides, the iRNA duplex represented by formula (III):





sense: 5′ np-Na—(XXX)i—Nb—YYY—Nb—(ZZZ)j—Na-nq 3′





antisense: 3′ np′—Na′—(X′X′X′)k—Nb′—Y′Y′Y′—Nb′—(Z′Z′Z′)iNa′-nq′ 5′   (III)


wherein:

    • i, j, k, and l are each independently 0 or 1;
    • p, p′, q, and q′ are each independently 0-6;
    • each Na and Na′ independently represents an oligonucleotide sequence comprising 0-25 modified nucleotides, each sequence comprising at least two differently modified nucleotides; each Nb and Nb′ independently represents an oligonucleotide sequence comprising 0-10 modified nucleotides;
    • wherein each np′, np, nq′, and nq, each of which may or may not be present, independently represents an overhang nucleotide; and
    • XXX, YYY, ZZZ, X′X′X′, Y′Y′Y′, and Z′Z′Z′ each independently represent one motif of three identical modifications on three consecutive nucleotides.


In one embodiment, i is 0 and j is 0; or i is 1 and j is 0; or i is 0 and j is 1; or both i and j are 0; or both i and j are 1. In another embodiment, k is 0 and l is 0; or k is 1 and l is 0; k is 0 and l is 1; or both k and l are 0; or both k and l are 1.


Exemplary combinations of the sense strand and antisense strand forming an iRNA duplex include the formulas below:





5′ np-Na—YYY—Na-nq 3′





3′ np′—Na′—Y′Y′Y′—Na′-nq′ 5′   (IIIa)





5′ np-Na—YYY—Nb—ZZZ—Na-nq 3′





3′ np′—Na′—Y′Y′Y′—Nb′—Z′Z′Z′—Na′-nq′ 5′   (IIIb)





5′ np-Na—XXX—Nb—YYY—Na-nq 3′





3′ np′—Na′—X′X′X′—Nb′—Y′Y′Y′—Na′-nq′ 5′   (IIIc)





5′ np-Na—XXX—Nb—YYY—Nb—ZZZ—Na-nq 3′





3′ np′—Na′—X′X′X′—Nb′—Y′Y′Y′—Nb′—Z′Z′Z′—Na-nq′ 5′   (IIId)


When the dsRNAi agent is represented by formula (IIIa), each Na independently represents an oligonucleotide sequence comprising 2-20, 2-15, or 2-10 modified nucleotides.


When the dsRNAi agent is represented by formula (IIIb), each Nb independently represents an oligonucleotide sequence comprising 1-10, 1-7, 1-5, or 1-4 modified nucleotides. Each Na independently represents an oligonucleotide sequence comprising 2-20, 2-15, or 2-10 modified nucleotides.


When the dsRNAi agent is represented as formula (IIIc), each Nb, Nb′ independently represents an oligonucleotide sequence comprising 0-10, 0-7, 0-10, 0-7, 0-5, 0-4, 0-2, or 0 modified nucleotides. Each Na independently represents an oligonucleotide sequence comprising 2-20, 2-15, or 2-10 modified nucleotides.


When the dsRNAi agent is represented as formula (IIId), each Nb, Nb′ independently represents an oligonucleotide sequence comprising 0-10, 0-7, 0-10, 0-7, 0-5, 0-4, 0-2, or 0 modified nucleotides. Each Na, Na′ independently represents an oligonucleotide sequence comprising 2-20, 2-15, or 2-10 modified nucleotides. Each of Na, Na′, Nb, and Nb′ independently comprises modifications of alternating pattern.


Each of X, Y, and Z in formulas (III), (IIIa), (IIIb), (IIIc), and (IIId) may be the same or different from each other.


When the dsRNAi agent is represented by formula (III), (IIIa), (IIIb), (IIIc), and (IIId), at least one of the Y nucleotides may form a base pair with one of the Y′ nucleotides. Alternatively, at least two of the Y nucleotides form base pairs with the corresponding Y′ nucleotides; or all three of the Y nucleotides all form base pairs with the corresponding Y′ nucleotides.


When the dsRNAi agent is represented by formula (IIIb) or (IIId), at least one of the Z nucleotides may form a base pair with one of the Z′ nucleotides. Alternatively, at least two of the Z nucleotides form base pairs with the corresponding Z′ nucleotides; or all three of the Z nucleotides all form base pairs with the corresponding Z′ nucleotides.


When the dsRNAi agent is represented as formula (IIIc) or (IIId), at least one of the X nucleotides may form a base pair with one of the X′ nucleotides. Alternatively, at least two of the X nucleotides form base pairs with the corresponding X′ nucleotides; or all three of the X nucleotides all form base pairs with the corresponding X′ nucleotides.


In certain embodiments, the modification on the Y nucleotide is different than the modification on the Y′ nucleotide, the modification on the Z nucleotide is different than the modification on the Z′ nucleotide, or the modification on the X nucleotide is different than the modification on the X′ nucleotide.


In certain embodiments, when the dsRNAi agent is represented by formula (IIId), the Na modifications are 2′-O-methyl or 2′-fluoro modifications. In other embodiments, when the RNAi agent is represented by formula (IIId), the Na modifications are 2′-O-methyl or 2′-fluoro modifications and np′>0 and at least one np′ is linked to a neighboring nucleotide a via phosphorothioate linkage. In yet other embodiments, when the RNAi agent is represented by formula (IIId), the Na modifications are 2′-O-methyl or 2′-fluoro modifications, np′>0 and at least one np′ is linked to a neighboring nucleotide via phosphorothioate linkage, and the sense strand is conjugated to one or more GalNAc derivatives attached through a bivalent or trivalent branched linker (described below). In other embodiments, when the RNAi agent is represented by formula (IIId), the Na modifications are 2′-O-methyl or 2′-fluoro modifications, np′>0 and at least one np′ is linked to a neighboring nucleotide via phosphorothioate linkage, the sense strand comprises at least one phosphorothioate linkage, and the sense strand is conjugated to one or more GalNAc derivatives attached through a bivalent or trivalent branched linker.


In some embodiments, when the dsRNAi agent is represented by formula (IIIa), the Na modifications are 2′-O-methyl or 2′-fluoro modifications, np′>0 and at least one np′ is linked to a neighboring nucleotide via phosphorothioate linkage, the sense strand comprises at least one phosphorothioate linkage, and the sense strand is conjugated to one or more GalNAc derivatives attached through a bivalent or trivalent branched linker.


In some embodiments, the dsRNAi agent is a multimer containing at least two duplexes represented by formula (III), (IIIa), (IIIb), (IIIc), and (IIId), wherein the duplexes are connected by a linker. The linker can be cleavable or non-cleavable. Optionally, the multimer further comprises a ligand. Each of the duplexes can target the same gene or two different genes; or each of the duplexes can target same gene at two different target sites.


In some embodiments, the dsRNAi agent is a multimer containing three, four, five, six, or more duplexes represented by formula (III), (IIIa), (IIIb), (IIIc), and (IIId), wherein the duplexes are connected by a linker. The linker can be cleavable or non-cleavable. Optionally, the multimer further comprises a ligand. Each of the duplexes can target the same gene or two different genes; or each of the duplexes can target same gene at two different target sites.


In one embodiment, two dsRNAi agents represented by at least one of formulas (III), (IIIa), (IIIb), (IIIc), and (IIId) are linked to each other at the 5′ end, and one or both of the 3′ ends, and are optionally conjugated to a ligand. Each of the agents can target the same gene or two different genes; or each of the agents can target same gene at two different target sites.


In certain embodiments, an RNAi agent of the invention may contain a low number of nucleotides containing a 2′-fluoro modification, e.g., 10 or fewer nucleotides with 2′-fluoro modification. For example, the RNAi agent may contain 10, 9, 8, 7, 6, 5, 4, 3, 2, 1 or 0 nucleotides with a 2′-fluoro modification. In a specific embodiment, the RNAi agent of the invention contains 10 nucleotides with a 2′-fluoro modification, e.g., 4 nucleotides with a 2′-fluoro modification in the sense strand and 6 nucleotides with a 2′-fluoro modification in the antisense strand. In another specific embodiment, the RNAi agent of the invention contains 6 nucleotides with a 2′-fluoro modification, e.g., 4 nucleotides with a 2′-fluoro modification in the sense strand and 2 nucleotides with a 2′-fluoro modification in the antisense strand.


In other embodiments, an RNAi agent of the invention may contain an ultra low number of nucleotides containing a 2′-fluoro modification, e.g., 2 or fewer nucleotides containing a 2′-fluoro modification. For example, the RNAi agent may contain 2, 1 of 0 nucleotides with a 2′-fluoro modification. In a specific embodiment, the RNAi agent may contain 2 nucleotides with a 2′-fluoro modification, e.g., 0 nucleotides with a 2-fluoro modification in the sense strand and 2 nucleotides with a 2′-fluoro modification in the antisense strand.


Various publications describe multimeric iRNAs that can be used in the methods of the invention. Such publications include WO2007/091269, U.S. Pat. No. 7,858,769, WO2010/141511, WO2007/117686, WO2009/014887, and WO2011/031520 the entire contents of each of which are hereby incorporated herein by reference.


In certain embodiments, the compositions and methods of the disclosure include a vinyl phosphonate (VP) modification of an RNAi agent as described herein. In exemplary embodiments, a 5′-vinyl phosphonate modified nucleotide of the disclosure has the structure:




embedded image


wherein X is O or S;

    • R is hydrogen, hydroxy, fluoro, or C1-20alkoxy (e.g., methoxy or n-hexadecyloxy);
    • R5′ is ═C(H)—P(O)(OH)2 and the double bond between the C5′ carbon and R5′ is in the E or Z orientation (e.g., E orientation); and
    • B is a nucleobase or a modified nucleobase, optionally where B is adenine, guanine, cytosine, thymine, or uracil.


A vinyl phosphonate of the instant disclosure may be attached to either the antisense or the sense strand of a dsRNA of the disclosure. In certain embodiments, a vinyl phosphonate of the instant disclosure is attached to the antisense strand of a dsRNA, optionally at the 5′ end of the antisense strand of the dsRNA.


Vinyl phosphate modifications are also contemplated for the compositions and methods of the instant disclosure. An exemplary vinyl phosphate structure includes the preceding structure, where R5′ is ═C(H)—OP(O)(OH)2 and the double bond between the C5′ carbon and R5′ is in the E or Z orientation (e.g., E orientation).


As described in more detail below, the iRNA that contains conjugations of one or more carbohydrate moieties to an iRNA can optimize one or more properties of the iRNA. In many cases, the carbohydrate moiety will be attached to a modified subunit of the iRNA. For example, the ribose sugar of one or more ribonucleotide subunits of an iRNA can be replaced with another moiety, e.g., a non-carbohydrate (such as, cyclic) carrier to which is attached a carbohydrate ligand. A ribonucleotide subunit in which the ribose sugar of the subunit has been so replaced is referred to herein as a ribose replacement modification subunit (RRMS). A cyclic carrier may be a carbocyclic ring system, i.e., all ring atoms are carbon atoms, or a heterocyclic ring system, i.e., one or more ring atoms may be a heteroatom, e.g., nitrogen, oxygen, sulfur. The cyclic carrier may be a monocyclic ring system, or may contain two or more rings, e.g. fused rings. The cyclic carrier may be a fully saturated ring system, or it may contain one or more double bonds.


The ligand may be attached to the polynucleotide via a carrier. The carriers include (i) at least one “backbone attachment point,” such as, two “backbone attachment points” and (ii) at least one “tethering attachment point.” A “backbone attachment point” as used herein refers to a functional group, e.g. a hydroxyl group, or generally, a bond available for, and that is suitable for incorporation of the carrier into the backbone, e.g., the phosphate, or modified phosphate, e.g., sulfur containing, backbone, of a ribonucleic acid. A “tethering attachment point” (TAP) in some embodiments refers to a constituent ring atom of the cyclic carrier, e.g., a carbon atom or a heteroatom (distinct from an atom which provides a backbone attachment point), that connects a selected moiety. The moiety can be, e.g., a carbohydrate, e.g. monosaccharide, disaccharide, trisaccharide, tetrasaccharide, oligosaccharide, or polysaccharide. Optionally, the selected moiety is connected by an intervening tether to the cyclic carrier. Thus, the cyclic carrier will often include a functional group, e.g., an amino group, or generally, provide a bond, that is suitable for incorporation or tethering of another chemical entity, e.g., a ligand to the constituent ring.


The iRNA may be conjugated to a ligand via a carrier, wherein the carrier can be cyclic group or acyclic group. In some embodiments, the cyclic group is selected from pyrrolidinyl, pyrazolinyl, pyrazolidinyl, imidazolinyl, imidazolidinyl, piperidinyl, piperazinyl, [1,3]dioxolane, oxazolidinyl, isoxazolidinyl, morpholinyl, thiazolidinyl, isothiazolidinyl, quinoxalinyl, pyridazinonyl, tetrahydrofuryl, and decalin. In some embodiments, the acyclic group is a serinol backbone or diethanolamine backbone.


i. Thermally Destabilizing Modifications


In certain embodiments, a dsRNA molecule can be optimized for RNA interference by incorporating thermally destabilizing modifications in the seed region of the antisense strand. As used herein “seed region” means at positions 2-9 of the 5′-end of the referenced strand. For example, thermally destabilizing modifications can be incorporated in the seed region of the antisense strand to reduce or inhibit off-target gene silencing.


The term “thermally destabilizing modification(s)” includes modification(s) that would result with a dsRNA with a lower overall melting temperature (Tm) than the Tm of the dsRNA without having such modification(s). For example, the thermally destabilizing modification(s) can decrease the Tm of the dsRNA by 1-4° C., such as one, two, three or four degrees Celcius. And, the term “thermally destabilizing nucleotide” refers to a nucleotide containing one or more thermally destabilizing modifications.


It has been discovered that dsRNAs with an antisense strand comprising at least one thermally destabilizing modification of the duplex within the first 9 nucleotide positions, counting from the 5′ end, of the antisense strand have reduced off-target gene silencing activity. Accordingly, in some embodiments, the antisense strand comprises at least one (e.g., one, two, three, four, five or more) thermally destabilizing modification of the duplex within the first 9 nucleotide positions of the 5′ region of the antisense strand. In some embodiments, one or more thermally destabilizing modification(s) of the duplex is/are located in positions 2-9, such as, positions 4-8, from the 5′-end of the antisense strand. In some further embodiments, the thermally destabilizing modification(s) of the duplex is/are located at position 6, 7 or 8 from the 5′-end of the antisense strand. In still some further embodiments, the thermally destabilizing modification of the duplex is located at position 7 from the 5′-end of the antisense strand. In some embodiments, the thermally destabilizing modification of the duplex is located at position 2, 3, 4, 5 or 9 from the 5′-end of the antisense strand.


The thermally destabilizing modifications can include, but are not limited to, abasic modification; mismatch with the opposing nucleotide in the opposing strand; and sugar modification such as 2′-deoxy modification or acyclic nucleotide, e.g., unlocked nucleic acids (UNA) or glycol nucleic acid (GNA).


An iRNA agent comprises a sense strand and an antisense strand, each strand having 14 to 40 nucleotides. The RNAi agent may be represented by formula (L):




embedded image


In formula (L), B1, B2, B3, B1′, B2′, B3′, and B4′ each are independently a nucleotide containing a modification selected from the group consisting of 2′-O-alkyl, 2′-substituted alkoxy, 2′-substituted alkyl, 2′-halo, ENA, and BNA/LNA. In one embodiment, B1, B2, B3, B1′, B2′, B3′, and B4′ each contain 2′-OMe modifications. In one embodiment, B1, B2, B3, B1′, B2′, B3′, and B4′ each contain 2′-OMe or 2′-F modifications. In one embodiment, at least one of B1, B2, B3, B1′, B2′, B3′, and B4′ contain 2′-O—N-methylacetamido (2′-O-NMA) modification.


C1 is a thermally destabilizing nucleotide placed at a site opposite to the seed region of the antisense strand (i.e., at positions 2-8 of the 5′-end of the antisense strand). For example, C1 is at a position of the sense strand that pairs with a nucleotide at positions 2-8 of the 5′-end of the antisense strand. In one example, C1 is at position 15 from the 5′-end of the sense strand. C1 nucleotide bears the thermally destabilizing modification which can include abasic modification; mismatch with the opposing nucleotide in the duplex; and sugar modification such as 2′-deoxy modification or acyclic nucleotide e.g., unlocked nucleic acids (UNA) or glycerol nucleic acid (GNA). In one embodiment, C1 has thermally destabilizing modification selected from the group consisting of: i) mismatch with the opposing nucleotide in the antisense strand; ii) abasic modification selected from the group consisting of:




embedded image


and iii) sugar modification selected from the group consisting of:




embedded image


wherein B is a modified or unmodified nucleobase, R1 and R2 independently are H, halogen, OR3, or alkyl; and R3 is H, alkyl, cycloalkyl, aryl, aralkyl, heteroaryl or sugar. In one embodiment, the thermally destabilizing modification in C1 is a mismatch selected from the group consisting of G:G, G:A, G:U, G:T, A:A, A:C, C:C, C:U, C:T, U:U, T:T, and U:T; and optionally, at least one nucleobase in the mismatch pair is a 2′-deoxy nucleobase. In one example, the thermally destabilizing modification in C1 is GNA or




embedded image


T1, T1′, T2′, and T3′ each independently represent a nucleotide comprising a modification providing the nucleotide a steric bulk that is less or equal to the steric bulk of a 2′-OMe modification. A steric bulk refers to the sum of steric effects of a modification. Methods for determining steric effects of a modification of a nucleotide are known to one skilled in the art. The modification can be at the 2′ position of a ribose sugar of the nucleotide, or a modification to a non-ribose nucleotide, acyclic nucleotide, or the backbone of the nucleotide that is similar or equivalent to the 2′ position of the ribose sugar, and provides the nucleotide a steric bulk that is less than or equal to the steric bulk of a 2′-OMe modification. For example, T1, T1′, T2′, and T3′ are each independently selected from DNA, RNA, LNA, 2′-F, and 2′-F-5′-methyl. In one embodiment, T1 is DNA. In one embodiment, T1′ is DNA, RNA or LNA. In one embodiment, T2′ is DNA or RNA. In one embodiment, T3′ is DNA or RNA.


n1, n3, and q1 are independently 4 to 15 nucleotides in length.


n5, q3, and q7 are independently 1-6 nucleotide(s) in length.


n4, q2, and q6 are independently 1-3 nucleotide(s) in length; alternatively, n4 is 0.


q5 is independently 0-10 nucleotide(s) in length.


n2 and q4 are independently 0-3 nucleotide(s) in length.


Alternatively, n4 is 0-3 nucleotide(s) in length.


In one embodiment, n4 can be 0. In one example, n4 is 0, and q2 and q6 are 1. In another example, n4 is 0, and q2 and q6 are 1, with two phosphorothioate internucleotide linkage modifications within position 1-5 of the sense strand (counting from the 5′-end of the sense strand), and two phosphorothioate internucleotide linkage modifications at positions 1 and 2 and two phosphorothioate internucleotide linkage modifications within positions 18-23 of the antisense strand (counting from the 5′-end of the antisense strand).


In one embodiment, n4, q2, and q6 are each 1.


In one embodiment, n2, n4, q2, q4, and q6 are each 1.


In one embodiment, C1 is at position 14-17 of the 5′-end of the sense strand, when the sense strand is 19-22 nucleotides in length, and n4 is 1. In one embodiment, C1 is at position 15 of the 5′-end of the sense strand


In one embodiment, T3′ starts at position 2 from the 5′ end of the antisense strand. In one example, T3′ is at position 2 from the 5′ end of the antisense strand and q6 is equal to 1.


In one embodiment, T1′ starts at position 14 from the 5′ end of the antisense strand. In one example, T1′ is at position 14 from the 5′ end of the antisense strand and q2 is equal to 1.


In an exemplary embodiment, T3′ starts from position 2 from the 5′ end of the antisense strand and T1′ starts from position 14 from the 5′ end of the antisense strand. In one example, T3′ starts from position 2 from the 5′ end of the antisense strand and q6 is equal to 1 and T1′ starts from position 14 from the 5′ end of the antisense strand and q2 is equal to 1.


In one embodiment, T1′ and T3′ are separated by 11 nucleotides in length (i.e. not counting the T1′ and T3′ nucleotides).


In one embodiment, T1′ is at position 14 from the 5′ end of the antisense strand. In one example, T1′ is at position 14 from the 5′ end of the antisense strand and q2 is equal to 1, and the modification at the 2′ position or positions in a non-ribose, acyclic or backbone that provide less steric bulk than a 2′-OMe ribose.


In one embodiment, T3′ is at position 2 from the 5′ end of the antisense strand. In one example, T3′ is at position 2 from the 5′ end of the antisense strand and q6 is equal to 1, and the modification at the 2′ position or positions in a non-ribose, acyclic or backbone that provide less than or equal to steric bulk than a 2′-OMe ribose.


In one embodiment, T1 is at the cleavage site of the sense strand. In one example, T1 is at position 11 from the 5′ end of the sense strand, when the sense strand is 19-22 nucleotides in length, and n2 is 1. In an exemplary embodiment, T1 is at the cleavage site of the sense strand at position 11 from the 5′ end of the sense strand, when the sense strand is 19-22 nucleotides in length, and n2 is 1,


In one embodiment, T2′ starts at position 6 from the 5′ end of the antisense strand. In one example, T2′ is at positions 6-10 from the 5′ end of the antisense strand, and q4 is 1.


In an exemplary embodiment, T1 is at the cleavage site of the sense strand, for instance, at position 11 from the 5′ end of the sense strand, when the sense strand is 19-22 nucleotides in length, and n2 is 1; T1′ is at position 14 from the 5′ end of the antisense strand, and q2 is equal to 1, and the modification to T1′ is at the 2′ position of a ribose sugar or at positions in a non-ribose, acyclic or backbone that provide less steric bulk than a 2′-OMe ribose; T2′ is at positions 6-10 from the 5′ end of the antisense strand, and q4 is 1; and T3′ is at position 2 from the 5′ end of the antisense strand, and q6 is equal to 1, and the modification to T3′ is at the 2′ position or at positions in a non-ribose, acyclic or backbone that provide less than or equal to steric bulk than a 2′-OMe ribose.


In one embodiment, T2′ starts at position 8 from the 5′ end of the antisense strand. In one example, T2′ starts at position 8 from the 5′ end of the antisense strand, and q4 is 2.


In one embodiment, T2′ starts at position 9 from the 5′ end of the antisense strand. In one example, T2′ is at position 9 from the 5′ end of the antisense strand, and q4 is 1.


In one embodiment, B1′ is 2′-OMe or 2′-F, q1 is 9, T1′ is 2′-F, q2 is 1, B2′ is 2′-OMe or 2′-F, q3 is 4, T2′ is 2′-F, q4 is 1, B3′ is 2′-OMe or 2′-F, q5 is 6, T3′ is 2′-F, q6 is 1, B4′ is 2′-OMe, and q7 is 1; with two phosphorothioate internucleotide linkage modifications within positions 1-5 of the sense strand (counting from the 5′-end of the sense strand), and two phosphorothioate internucleotide linkage modifications at positions 1 and 2 and two phosphorothioate internucleotide linkage modifications within positions 18-23 of the antisense strand (counting from the 5′-end of the antisense strand).


In one embodiment, n4 is 0, B3 is 2′-OMe, n5 is 3, B1′ is 2′-OMe or 2′-F, q1 is 9, T1′ is 2′-F, q2 is 1, B2′ is 2′-OMe or 2′-F, q3 is 4, T2′ is 2′-F, q4 is 1, B3′ is 2′-OMe or 2′-F, q5 is 6, T3′ is 2′-F, q6 is 1, B4′ is 2′-OMe, and q7 is 1; with two phosphorothioate internucleotide linkage modifications within positions 1-5 of the sense strand (counting from the 5′-end of the sense strand), and two phosphorothioate internucleotide linkage modifications at positions 1 and 2 and two phosphorothioate internucleotide linkage modifications within positions 18-23 of the antisense strand (counting from the 5′-end of the antisense strand).


In one embodiment, B1 is 2′-OMe or 2′-F, n1 is 8, T1 is 2′F, n2 is 3, B2 is 2′-OMe, n3 is 7, n4 is 0, B3 is 2′OMe, n5 is 3, B1′ is 2′-OMe or 2′-F, q1 is 9, T1′ is 2′-F, q2 is 1, B2′ is 2′-OMe or 2′-F, q3 is 4, T2′ is 2′-F, q4 is 2, B3′ is 2′-OMe or 2′-F, q5 is 5, T3′ is 2′-F, q6 is 1, B4′ is 2′-OMe, and q7 is 1.


In one embodiment, B1 is 2′-OMe or 2′-F, n1 is 8, T1 is 2′F, n2 is 3, B2 is 2′-OMe, n3 is 7, n4 is 0, B3 is 2′-OMe, n5 is 3, B1′ is 2′-OMe or 2′-F, q1 is 9, T1′ is 2′-F, q2 is 1, B2′ is 2′-OMe or 2′-F, q3 is 4, T2′ is 2′-F, q4 is 2, B3′ is 2′-OMe or 2′-F, q5 is 5, T3′ is 2′-F, q6 is 1, B4′ is 2′-OMe, and q7 is 1; with two phosphorothioate internucleotide linkage modifications within positions 1-5 of the sense strand (counting from the 5′-end of the sense strand), and two phosphorothioate internucleotide linkage modifications at positions 1 and 2 and two phosphorothioate internucleotide linkage modifications within positions 18-23 of the antisense strand (counting from the 5′-end of the antisense strand).


In one embodiment, B1 is 2′-OMe or 2′-F, n1 is 6, T1 is 2′F, n2 is 3, B2 is 2′-OMe, n3 is 7, n4 is 0, B3 is 2′OMe, n5 is 3, B1′ is 2′-OMe or 2′-F, q1 is 7, T1′ is 2′-F, q2 is 1, B2′ is 2′-OMe or 2′-F, q3 is 4, T2′ is 2′-F, q4 is 2, B3′ is 2′-OMe or 2′-F, q5 is 5, T3′ is 2′-F, q6 is 1, B4′ is 2′-OMe, and q7 is 1.


In one embodiment, B1 is 2′-OMe or 2′-F, n1 is 6, T1 is 2′F, n2 is 3, B2 is 2′-OMe, n3 is 7, n4 is 0, B3 is 2′-OMe, n5 is 3, B1′ is 2′-OMe or 2′-F, q1 is 7, T1′ is 2′-F, q2 is 1, B2′ is 2′-OMe or 2′-F, q3 is 4, T2′ is 2′-F, q4 is 2, B3′ is 2′-OMe or 2′-F, q5 is 5, T3′ is 2′-F, q6 is 1, B4′ is 2′-OMe, and q7 is 1; with two phosphorothioate internucleotide linkage modifications within positions 1-5 of the sense strand (counting from the 5′-end of the sense strand), and two phosphorothioate internucleotide linkage modifications at positions 1 and 2 and two phosphorothioate internucleotide linkage modifications within positions 18-23 of the antisense strand (counting from the 5′-end of the antisense strand).


In one embodiment, B1 is 2′-OMe or 2′-F, n1 is 8, T1 is 2′F, n2 is 3, B2 is 2′-OMe, n3 is 7, n4 is 0, B3 is 2′OMe, n5 is 3, B1′ is 2′-OMe or 2′-F, q1 is 9, T1′ is 2′-F, q2 is 1, B2′ is 2′-OMe or 2′-F, q3 is 4, T2′ is 2′-F, q4 is 1, B3′ is 2′-OMe or 2′-F, q5 is 6, T3′ is 2′-F, q6 is 1, B4′ is 2′-OMe, and q7 is 1.


In one embodiment, B1 is 2′-OMe or 2′-F, n1 is 8, T1 is 2′F, n2 is 3, B2 is 2′-OMe, n3 is 7, n4 is 0, B3 is 2′-OMe, n5 is 3, B1′ is 2′-OMe or 2′-F, q1 is 9, T1′ is 2′-F, q2 is 1, B2′ is 2′-OMe or 2′-F, q3 is 4, T2′ is 2′-F, q4 is 1, B3′ is 2′-OMe or 2′-F, q5 is 6, T3′ is 2′-F, q6 is 1, B4′ is 2′-OMe, and q7 is 1; with two phosphorothioate internucleotide linkage modifications within positions 1-5 of the sense strand (counting from the 5′-end of the sense strand), and two phosphorothioate internucleotide linkage modifications at positions 1 and 2 and two phosphorothioate internucleotide linkage modifications within positions 18-23 of the antisense strand (counting from the 5′-end of the antisense strand).


In one embodiment, B1 is 2′-OMe or 2′-F, n1 is 8, T1 is 2′F, n2 is 3, B2 is 2′-OMe, n3 is 7, n4 is 0, B3 is 2′OMe, n5 is 3, B1′ is 2′-OMe or 2′-F, q1 is 9, T1′ is 2′-F, q2 is 1, B2′ is 2′-OMe or 2′-F, q3 is 5, T2′ is 2′-F, q4 is 1, B3′ is 2′-OMe or 2′-F, q5 is 5, T3′ is 2′-F, q6 is 1, B4′ is 2′-OMe, and q7 is 1; optionally with at least 2 additional TT at the 3′-end of the antisense strand.


In one embodiment, B1 is 2′-OMe or 2′-F, n1 is 8, T1 is 2′F, n2 is 3, B2 is 2′-OMe, n3 is 7, n4 is 0, B3 is 2′-OMe, n5 is 3, B1′ is 2′-OMe or 2′-F, q1 is 9, T1′ is 2′-F, q2 is 1, B2′ is 2′-OMe or 2′-F, q3 is 5, T2′ is 2′-F, q4 is 1, B3′ is 2′-OMe or 2′-F, q5 is 5, T3′ is 2′-F, q6 is 1, B4′ is 2′-OMe, and q7 is 1; optionally with at least 2 additional TT at the 3′-end of the antisense strand; with two phosphorothioate internucleotide linkage modifications within positions 1-5 of the sense strand (counting from the 5′-end of the sense strand), and two phosphorothioate internucleotide linkage modifications at positions 1 and 2 and two phosphorothioate internucleotide linkage modifications within positions 18-23 of the antisense strand (counting from the 5′-end of the antisense strand).


In one embodiment, B1 is 2′-OMe or 2′-F, n1 is 8, T1 is 2′F, n2 is 3, B2 is 2′-OMe, n3 is 7, n4 is 0, B3 is 2′-OMe, n5 is 3, B1′ is 2′-OMe or 2′-F, q1 is 9, T1′ is 2′-F, q2 is 1, B2′ is 2′-OMe or 2′-F, q3 is 4, q4 is 0, B3′ is 2′-OMe or 2′-F, q5 is 7, T3′ is 2′-F, q6 is 1, B4′ is 2′-OMe, and q7 is 1.


In one embodiment, B1 is 2′-OMe or 2′-F, n1 is 8, T1 is 2′F, n2 is 3, B2 is 2′-OMe, n3 is 7, n4 is 0, B3 is 2′-OMe, n5 is 3, B1′ is 2′-OMe or 2′-F, q1 is 9, T1′ is 2′-F, q2 is 1, B2′ is 2′-OMe or 2′-F, q3 is 4, q4 is 0, B3′ is 2′-OMe or 2′-F, q5 is 7, T3′ is 2′-F, q6 is 1, B4′ is 2′-OMe, and q7 is 1; with two phosphorothioate internucleotide linkage modifications within positions 1-5 of the sense strand (counting from the 5′-end), and two phosphorothioate internucleotide linkage modifications at positions 1 and 2 and two phosphorothioate internucleotide linkage modifications within positions 18-23 of the antisense strand (counting from the 5′-end).


In one embodiment, B1 is 2′-OMe or 2′-F, n1 is 8, T1 is 2′F, n2 is 3, B2 is 2′-OMe, n3 is 7, n4 is 0, B3 is 2′OMe, n5 is 3, B1′ is 2′-OMe or 2′-F, q1 is 9, T1′ is 2′-F, q2 is 1, B2′ is 2′-OMe or 2′-F, q3 is 4, T2′ is 2′-F, q4 is 2, B3′ is 2′-OMe or 2′-F, q5 is 5, T3′ is 2′-F, q6 is 1, B4′ is 2′-F, and q7 is 1.


In one embodiment, B1 is 2′-OMe or 2′-F, n1 is 8, T1 is 2′F, n2 is 3, B2 is 2′-OMe, n3 is 7, n4 is 0, B3 is 2′-OMe, n5 is 3, B1′ is 2′-OMe or 2′-F, q1 is 9, T1′ is 2′-F, q2 is 1, B2′ is 2′-OMe or 2′-F, q3 is 4, T2′ is 2′-F, q4 is 2, B3′ is 2′-OMe or 2′-F, q5 is 5, T3′ is 2′-F, q6 is 1, B4′ is 2′-F, and q7 is 1; with two phosphorothioate internucleotide linkage modifications within positions 1-5 of the sense strand (counting from the 5′-end of the sense strand), and two phosphorothioate internucleotide linkage modifications at positions 1 and 2 and two phosphorothioate internucleotide linkage modifications within positions 18-23 of the antisense strand (counting from the 5′-end of the antisense strand).


In one embodiment, B1 is 2′-OMe or 2′-F, n1 is 8, T1 is 2′F, n2 is 3, B2 is 2′-OMe, n3 is 7, n4 is 0, B3 is 2′-OMe, n5 is 3, B1′ is 2′-OMe or 2′-F, q1 is 9, T1′ is 2′-F, q2 is 1, B2′ is 2′-OMe or 2′-F, q3 is 4, q4 is 0, B3′ is 2′-OMe or 2′-F, q5 is 7, T3′ is 2′-F, q6 is 1, B4′ is 2′-F, and q7 is 1.


In one embodiment, B1 is 2′-OMe or 2′-F, n1 is 8, T1 is 2′F, n2 is 3, B2 is 2′-OMe, n3 is 7, n4 is 0, B3 is 2′-OMe, n5 is 3, B1′ is 2′-OMe or 2′-F, q1 is 9, T1′ is 2′-F, q2 is 1, B2′ is 2′-OMe or 2′-F, q3 is 4, q4 is 0, B3′ is 2′-OMe or 2′-F, q5 is 7, T3′ is 2′-F, q6 is 1, B4′ is 2′-F, and q7 is 1; with two phosphorothioate internucleotide linkage modifications within positions 1-5 of the sense strand (counting from the 5′-end of the sense strand), and two phosphorothioate internucleotide linkage modifications at positions 1 and 2 and two phosphorothioate internucleotide linkage modifications within positions 18-23 of the antisense strand (counting from the 5′-end of the antisense strand).


The RNAi agent can comprise a phosphorus-containing group at the 5′-end of the sense strand or antisense strand. The 5′-end phosphorus-containing group can be 5′-end phosphate (5′-P), 5′-end phosphorothioate (5′-PS), 5′-end phosphorodithioate (5′-PS2), 5′-end vinylphosphonate (5′-VP), 5′-end methylphosphonate (MePhos), or 5′-deoxy-5′-C-malonyl




embedded image


When the 5′-end phosphorus-containing group is 5′-end vinylphosphonate (5′-VP), the 5′-VP can be either 5′-E-VP isomer (i.e., trans-vinylphosphate,




embedded image


5′-Z-VP isomer (i.e., cis-vinylphosphate,




embedded image


or mixtures thereof.


In one embodiment, the RNAi agent comprises a phosphorus-containing group at the 5′-end of the sense strand. In one embodiment, the RNAi agent comprises a phosphorus-containing group at the 5′-end of the antisense strand.


In one embodiment, the RNAi agent comprises a 5′-P. In one embodiment, the RNAi agent comprises a 5′-P in the antisense strand.


In one embodiment, the RNAi agent comprises a 5′-PS. In one embodiment, the RNAi agent comprises a 5′-PS in the antisense strand.


In one embodiment, the RNAi agent comprises a 5′-VP. In one embodiment, the RNAi agent comprises a 5′-VP in the antisense strand. In one embodiment, the RNAi agent comprises a 5′-E-VP in the antisense strand. In one embodiment, the RNAi agent comprises a 5′-Z-VP in the antisense strand.


In one embodiment, the RNAi agent comprises a 5′-PS2. In one embodiment, the RNAi agent comprises a 5′-PS2 in the antisense strand.


In one embodiment, the RNAi agent comprises a 5′-PS2. In one embodiment, the RNAi agent comprises a 5′-deoxy-5′-C-malonyl in the antisense strand.


In one embodiment, B1 is 2′-OMe or 2′-F, n1 is 8, T1 is 2′F, n2 is 3, B2 is 2′-OMe, n3 is 7, n4 is 0, B3 is 2′OMe, n5 is 3, B1′ is 2′-OMe or 2′-F, q1 is 9, T1′ is 2′-F, q2 is 1, B2′ is 2′-OMe or 2′-F, q3 is 4, T2′ is 2′-F, q4 is 2, B3′ is 2′-OMe or 2′-F, q5 is 5, T3′ is 2′-F, q6 is 1, B4′ is 2′-OMe, and q7 is 1. The RNAi agent also comprises a 5′-PS.


In one embodiment, B1 is 2′-OMe or 2′-F, n1 is 8, T1 is 2′F, n2 is 3, B2 is 2′-OMe, n3 is 7, n4 is 0, B3 is 2′OMe, n5 is 3, B1′ is 2′-OMe or 2′-F, q1 is 9, T1′ is 2′-F, q2 is 1, B2′ is 2′-OMe or 2′-F, q3 is 4, T2′ is 2′-F, q4 is 2, B3′ is 2′-OMe or 2′-F, q5 is 5, T3′ is 2′-F, q6 is 1, B4′ is 2′-OMe, and q7 is 1. The RNAi agent also comprises a 5′-P.


In one embodiment, B1 is 2′-OMe or 2′-F, n1 is 8, T1 is 2′F, n2 is 3, B2 is 2′-OMe, n3 is 7, n4 is 0, B3 is 2′OMe, n5 is 3, B1′ is 2′-OMe or 2′-F, q1 is 9, T1′ is 2′-F, q2 is 1, B2′ is 2′-OMe or 2′-F, q3 is 4, T2′ is 2′-F, q4 is 2, B3′ is 2′-OMe or 2′-F, q5 is 5, T3′ is 2′-F, q6 is 1, B4′ is 2′-OMe, and q7 is 1. The RNAi agent also comprises a 5′-VP. The 5′-VP may be 5′-E-VP, 5′-Z-VP, or combination thereof.


In one embodiment, B1 is 2′-OMe or 2′-F, n1 is 8, T1 is 2′F, n2 is 3, B2 is 2′-OMe, n3 is 7, n4 is 0, B3 is 2′OMe, n5 is 3, B1′ is 2′-OMe or 2′-F, q1 is 9, T1′ is 2′-F, q2 is 1, B2′ is 2′-OMe or 2′-F, q3 is 4, T2′ is 2′-F, q4 is 2, B3′ is 2′-OMe or 2′-F, q5 is 5, T3′ is 2′-F, q6 is 1, B4′ is 2′-OMe, and q7 is 1. The RNAi agent also comprises a 5′-PS2.


In one embodiment, B1 is 2′-OMe or 2′-F, n1 is 8, T1 is 2′F, n2 is 3, B2 is 2′-OMe, n3 is 7, n4 is 0, B3 is 2′OMe, n5 is 3, B1′ is 2′-OMe or 2′-F, q1 is 9, T1′ is 2′-F, q2 is 1, B2′ is 2′-OMe or 2′-F, q3 is 4, T2′ is 2′-F, q4 is 2, B3′ is 2′-OMe or 2′-F, q5 is 5, T3′ is 2′-F, q6 is 1, B4′ is 2′-OMe, and q7 is 1. The RNAi agent also comprises a 5′-deoxy-5′-C-malonyl.


In one embodiment, B1 is 2′-OMe or 2′-F, n1 is 8, T1 is 2′F, n2 is 3, B2 is 2′-OMe, n3 is 7, n4 is 0, B3 is 2′-OMe, n5 is 3, B1′ is 2′-OMe or 2′-F, q1 is 9, T1′ is 2′-F, q2 is 1, B2′ is 2′-OMe or 2′-F, q3 is 4, T2′ is 2′-F, q4 is 2, B3′ is 2′-OMe or 2′-F, q5 is 5, T3′ is 2′-F, q6 is 1, B4′ is 2′-OMe, and q7 is 1; with two phosphorothioate internucleotide linkage modifications within position 1-5 of the sense strand (counting from the 5′-end of the sense strand), and two phosphorothioate internucleotide linkage modifications at positions 1 and 2 and two phosphorothioate internucleotide linkage modifications within positions 18-23 of the antisense strand (counting from the 5′-end of the antisense strand). The RNAi agent also comprises a 5′-P.


In one embodiment, B1 is 2′-OMe or 2′-F, n1 is 8, T1 is 2′F, n2 is 3, B2 is 2′-OMe, n3 is 7, n4 is 0, B3 is 2′-OMe, n5 is 3, B1′ is 2′-OMe or 2′-F, q1 is 9, T1′ is 2′-F, q2 is 1, B2′ is 2′-OMe or 2′-F, q3 is 4, T2′ is 2′-F, q4 is 2, B3′ is 2′-OMe or 2′-F, q5 is 5, T3′ is 2′-F, q6 is 1, B4′ is 2′-OMe, and q7 is 1; with two phosphorothioate internucleotide linkage modifications within position 1-5 of the sense strand (counting from the 5′-end of the sense strand), and two phosphorothioate internucleotide linkage modifications at positions 1 and 2 and two phosphorothioate internucleotide linkage modifications within positions 18-23 of the antisense strand (counting from the 5′-end of the antisense strand). The RNAi agent also comprises a 5′-PS.


In one embodiment, B1 is 2′-OMe or 2′-F, n1 is 8, T1 is 2′F, n2 is 3, B2 is 2′-OMe, n3 is 7, n4 is 0, B3 is 2′-OMe, n5 is 3, B1′ is 2′-OMe or 2′-F, q1 is 9, T1′ is 2′-F, q2 is 1, B2′ is 2′-OMe or 2′-F, q3 is 4, T2′ is 2′-F, q4 is 2, B3′ is 2′-OMe or 2′-F, q5 is 5, T3′ is 2′-F, q6 is 1, B4′ is 2′-OMe, and q7 is 1; with two phosphorothioate internucleotide linkage modifications within position 1-5 of the sense strand (counting from the 5′-end of the sense strand), and two phosphorothioate internucleotide linkage modifications at positions 1 and 2 and two phosphorothioate internucleotide linkage modifications within positions 18-23 of the antisense strand (counting from the 5′-end of the antisense strand). The RNAi agent also comprises a 5′-VP. The 5′-VP may be 5′-E-VP, 5′-Z-VP, or combination thereof.


In one embodiment, B1 is 2′-OMe or 2′-F, n1 is 8, T1 is 2′F, n2 is 3, B2 is 2′-OMe, n3 is 7, n4 is 0, B3 is 2′-OMe, n5 is 3, B1′ is 2′-OMe or 2′-F, q1 is 9, T1′ is 2′-F, q2 is 1, B2′ is 2′-OMe or 2′-F, q3 is 4, T2′ is 2′-F, q4 is 2, B3′ is 2′-OMe or 2′-F, q5 is 5, T3′ is 2′-F, q6 is 1, B4′ is 2′-OMe, and q7 is 1; with two phosphorothioate internucleotide linkage modifications within position 1-5 of the sense strand (counting from the 5′-end of the sense strand), and two phosphorothioate internucleotide linkage modifications at positions 1 and 2 and two phosphorothioate internucleotide linkage modifications within positions 18-23 of the antisense strand (counting from the 5′-end of the antisense strand). The RNAi agent also comprises a 5′-PS2.


In one embodiment, B1 is 2′-OMe or 2′-F, n1 is 8, T1 is 2′F, n2 is 3, B2 is 2′-OMe, n3 is 7, n4 is 0, B3 is 2′-OMe, n5 is 3, B1′ is 2′-OMe or 2′-F, q1 is 9, T1′ is 2′-F, q2 is 1, B2′ is 2′-OMe or 2′-F, q3 is 4, T2′ is 2′-F, q4 is 2, B3′ is 2′-OMe or 2′-F, q5 is 5, T3′ is 2′-F, q6 is 1, B4′ is 2′-OMe, and q7 is 1; with two phosphorothioate internucleotide linkage modifications within position 1-5 of the sense strand (counting from the 5′-end of the sense strand), and two phosphorothioate internucleotide linkage modifications at positions 1 and 2 and two phosphorothioate internucleotide linkage modifications within positions 18-23 of the antisense strand (counting from the 5′-end of the antisense strand). The RNAi agent also comprises a 5′-deoxy-5′-C-malonyl.


In one embodiment, B1 is 2′-OMe or 2′-F, n1 is 8, T1 is 2′F, n2 is 3, B2 is 2′-OMe, n3 is 7, n4 is 0, B3 is 2′-OMe, n5 is 3, B1′ is 2′-OMe or 2′-F, q1 is 9, T1′ is 2′-F, q2 is 1, B2′ is 2′-OMe or 2′-F, q3 is 4, q4 is 0, B3′ is 2′-OMe or 2′-F, q5 is 7, T3′ is 2′-F, q6 is 1, B4′ is 2′-OMe, and q7 is 1. The RNAi agent also comprises a 5′-P.


In one embodiment, B1 is 2′-OMe or 2′-F, n1 is 8, T1 is 2′F, n2 is 3, B2 is 2′-OMe, n3 is 7, n4 is 0, B3 is 2′-OMe, n5 is 3, B1′ is 2′-OMe or 2′-F, q1 is 9, T1′ is 2′-F, q2 is 1, B2′ is 2′-OMe or 2′-F, q3 is 4, q4 is 0, B3′ is 2′-OMe or 2′-F, q5 is 7, T3′ is 2′-F, q6 is 1, B4′ is 2′-OMe, and q7 is 1. The dsRNA agent also comprises a 5′-PS.


In one embodiment, B1 is 2′-OMe or 2′-F, n1 is 8, T1 is 2′F, n2 is 3, B2 is 2′-OMe, n3 is 7, n4 is 0, B3 is 2′-OMe, n5 is 3, B1′ is 2′-OMe or 2′-F, q1 is 9, T1′ is 2′-F, q2 is 1, B2′ is 2′-OMe or 2′-F, q3 is 4, q4 is 0, B3′ is 2′-OMe or 2′-F, q5 is 7, T3′ is 2′-F, q6 is 1, B4′ is 2′-OMe, and q7 is 1. The RNAi agent also comprises a 5′-VP. The 5′-VP may be 5′-E-VP, 5′-Z-VP, or combination thereof.


In one embodiment, B1 is 2′-OMe or 2′-F, n1 is 8, T1 is 2′F, n2 is 3, B2 is 2′-OMe, n3 is 7, n4 is 0, B3 is 2′-OMe, n5 is 3, B1′ is 2′-OMe or 2′-F, q1 is 9, T1′ is 2′-F, q2 is 1, B2′ is 2′-OMe or 2′-F, q3 is 4, q4 is 0, B3′ is 2′-OMe or 2′-F, q5 is 7, T3′ is 2′-F, q6 is 1, B4′ is 2′-OMe, and q7 is 1. The RNAi agent also comprises a 5′-PS2.


In one embodiment, B1 is 2′-OMe or 2′-F, n1 is 8, T1 is 2′F, n2 is 3, B2 is 2′-OMe, n3 is 7, n4 is 0, B3 is 2′-OMe, n5 is 3, B1′ is 2′-OMe or 2′-F, q1 is 9, T1′ is 2′-F, q2 is 1, B2′ is 2′-OMe or 2′-F, q3 is 4, q4 is 0, B3′ is 2′-OMe or 2′-F, q5 is 7, T3′ is 2′-F, q6 is 1, B4′ is 2′-OMe, and q7 is 1. The RNAi agent also comprises a 5′-deoxy-5′-C-malonyl.


In one embodiment, B1 is 2′-OMe or 2′-F, n1 is 8, T1 is 2′F, n2 is 3, B2 is 2′-OMe, n3 is 7, n4 is 0, B3 is 2′-OMe, n5 is 3, B1′ is 2′-OMe or 2′-F, q1 is 9, T1′ is 2′-F, q2 is 1, B2′ is 2′-OMe or 2′-F, q3 is 4, q4 is 0, B3′ is 2′-OMe or 2′-F, q5 is 7, T3′ is 2′-F, q6 is 1, B4′ is 2′-OMe, and q7 is 1; with two phosphorothioate internucleotide linkage modifications within position 1-5 of the sense strand (counting from the 5′-end), and two phosphorothioate internucleotide linkage modifications at positions 1 and 2 and two phosphorothioate internucleotide linkage modifications within positions 18-23 of the antisense strand (counting from the 5′-end). The RNAi agent also comprises a 5′-P.


In one embodiment, B1 is 2′-OMe or 2′-F, n1 is 8, T1 is 2′F, n2 is 3, B2 is 2′-OMe, n3 is 7, n4 is 0, B3 is 2′-OMe, n5 is 3, B1′ is 2′-OMe or 2′-F, q1 is 9, T1′ is 2′-F, q2 is 1, B2′ is 2′-OMe or 2′-F, q3 is 4, q4 is 0, B3′ is 2′-OMe or 2′-F, q5 is 7, T3′ is 2′-F, q6 is 1, B4′ is 2′-OMe, and q7 is 1; with two phosphorothioate internucleotide linkage modifications within position 1-5 of the sense strand (counting from the 5′-end), and two phosphorothioate internucleotide linkage modifications at positions 1 and 2 and two phosphorothioate internucleotide linkage modifications within positions 18-23 of the antisense strand (counting from the 5′-end). The RNAi agent also comprises a 5′-PS.


In one embodiment, B1 is 2′-OMe or 2′-F, n1 is 8, T1 is 2′F, n2 is 3, B2 is 2′-OMe, n3 is 7, n4 is 0, B3 is 2′-OMe, n5 is 3, B1′ is 2′-OMe or 2′-F, q1 is 9, T1′ is 2′-F, q2 is 1, B2′ is 2′-OMe or 2′-F, q3 is 4, q4 is 0, B3′ is 2′-OMe or 2′-F, q5 is 7, T3′ is 2′-F, q6 is 1, B4′ is 2′-OMe, and q7 is 1; with two phosphorothioate internucleotide linkage modifications within position 1-5 of the sense strand (counting from the 5′-end), and two phosphorothioate internucleotide linkage modifications at positions 1 and 2 and two phosphorothioate internucleotide linkage modifications within positions 18-23 of the antisense strand (counting from the 5′-end). The RNAi agent also comprises a 5′-VP. The 5′-VP may be 5′-E-VP, 5′-Z-VP, or combination thereof.


In one embodiment, B1 is 2′-OMe or 2′-F, n1 is 8, T1 is 2′F, n2 is 3, B2 is 2′-OMe, n3 is 7, n4 is 0, B3 is 2′-OMe, n5 is 3, B1′ is 2′-OMe or 2′-F, q1 is 9, T1′ is 2′-F, q2 is 1, B2′ is 2′-OMe or 2′-F, q3 is 4, q4 is 0, B3′ is 2′-OMe or 2′-F, q5 is 7, T3′ is 2′-F, q6 is 1, B4′ is 2′-OMe, and q7 is 1; with two phosphorothioate internucleotide linkage modifications within position 1-5 of the sense strand (counting from the 5′-end), and two phosphorothioate internucleotide linkage modifications at positions 1 and 2 and two phosphorothioate internucleotide linkage modifications within positions 18-23 of the antisense strand (counting from the 5′-end). The RNAi agent also comprises a 5′-PS2.


In one embodiment, B1 is 2′-OMe or 2′-F, n1 is 8, T1 is 2′F, n2 is 3, B2 is 2′-OMe, n3 is 7, n4 is 0, B3 is 2′-OMe, n5 is 3, B1′ is 2′-OMe or 2′-F, q1 is 9, T1′ is 2′-F, q2 is 1, B2′ is 2′-OMe or 2′-F, q3 is 4, q4 is 0, B3′ is 2′-OMe or 2′-F, q5 is 7, T3′ is 2′-F, q6 is 1, B4′ is 2′-OMe, and q7 is 1; with two phosphorothioate internucleotide linkage modifications within position 1-5 of the sense strand (counting from the 5′-end), and two phosphorothioate internucleotide linkage modifications at positions 1 and 2 and two phosphorothioate internucleotide linkage modifications within positions 18-23 of the antisense strand (counting from the 5′-end). The RNAi agent also comprises a 5′-deoxy-5′-C-malonyl.


In one embodiment, B1 is 2′-OMe or 2′-F, n1 is 8, T1 is 2′F, n2 is 3, B2 is 2′-OMe, n3 is 7, n4 is 0, B3 is 2′OMe, n5 is 3, B1′ is 2′-OMe or 2′-F, q1 is 9, T1′ is 2′-F, q2 is 1, B2′ is 2′-OMe or 2′-F, q3 is 4, T2′ is 2′-F, q4 is 2, B3′ is 2′-OMe or 2′-F, q5 is 5, T3′ is 2′-F, q6 is 1, B4′ is 2′-F, and q7 is 1. The RNAi agent also comprises a 5′-P.


In one embodiment, B1 is 2′-OMe or 2′-F, n1 is 8, T1 is 2′F, n2 is 3, B2 is 2′-OMe, n3 is 7, n4 is 0, B3 is 2′OMe, n5 is 3, B1′ is 2′-OMe or 2′-F, q1 is 9, T1′ is 2′-F, q2 is 1, B2′ is 2′-OMe or 2′-F, q3 is 4, T2′ is 2′-F, q4 is 2, B3′ is 2′-OMe or 2′-F, q5 is 5, T3′ is 2′-F, q6 is 1, B4′ is 2′-F, and q7 is 1. The RNAi agent also comprises a 5′-PS.


In one embodiment, B1 is 2′-OMe or 2′-F, n1 is 8, T1 is 2′F, n2 is 3, B2 is 2′-OMe, n3 is 7, n4 is 0, B3 is 2′OMe, n5 is 3, B1′ is 2′-OMe or 2′-F, q1 is 9, T1′ is 2′-F, q2 is 1, B2′ is 2′-OMe or 2′-F, q3 is 4, T2′ is 2′-F, q4 is 2, B3′ is 2′-OMe or 2′-F, q5 is 5, T3′ is 2′-F, q6 is 1, B4′ is 2′-F, and q7 is 1. The RNAi agent also comprises a 5′-VP. The 5′-VP may be 5′-E-VP, 5′-Z-VP, or combination thereof.


In one embodiment, B1 is 2′-OMe or 2′-F, n1 is 8, T1 is 2′F, n2 is 3, B2 is 2′-OMe, n3 is 7, n4 is 0, B3 is 2′OMe, n5 is 3, B1′ is 2′-OMe or 2′-F, q1 is 9, T1′ is 2′-F, q2 is 1, B2′ is 2′-OMe or 2′-F, q3 is 4, T2′ is 2′-F, q4 is 2, B3′ is 2′-OMe or 2′-F, q5 is 5, T3′ is 2′-F, q6 is 1, B4′ is 2′-F, and q7 is 1. The dsRNAi RNA agent also comprises a 5′-PS2.


In one embodiment, B1 is 2′-OMe or 2′-F, n1 is 8, T1 is 2′F, n2 is 3, B2 is 2′-OMe, n3 is 7, n4 is 0, B3 is 2′OMe, n5 is 3, B1′ is 2′-OMe or 2′-F, q1 is 9, T1′ is 2′-F, q2 is 1, B2′ is 2′-OMe or 2′-F, q3 is 4, T2′ is 2′-F, q4 is 2, B3′ is 2′-OMe or 2′-F, q5 is 5, T3′ is 2′-F, q6 is 1, B4′ is 2′-F, and q7 is 1.


The RNAi agent also comprises a 5′-deoxy-5′-C-malonyl.


In one embodiment, B1 is 2′-OMe or 2′-F, n1 is 8, T1 is 2′F, n2 is 3, B2 is 2′-OMe, n3 is 7, n4 is 0, B3 is 2′-OMe, n5 is 3, B1′ is 2′-OMe or 2′-F, q1 is 9, T1′ is 2′-F, q2 is 1, B2′ is 2′-OMe or 2′-F, q3 is 4, T2′ is 2′-F, q4 is 2, B3′ is 2′-OMe or 2′-F, q5 is 5, T3′ is 2′-F, q6 is 1, B4′ is 2′-F, and q7 is 1; with two phosphorothioate internucleotide linkage modifications within position 1-5 of the sense strand (counting from the 5′-end of the sense strand), and two phosphorothioate internucleotide linkage modifications at positions 1 and 2 and two phosphorothioate internucleotide linkage modifications within positions 18-23 of the antisense strand (counting from the 5′-end of the antisense strand). The RNAi agent also comprises a 5′-P.


In one embodiment, B1 is 2′-OMe or 2′-F, n1 is 8, T1 is 2′F, n2 is 3, B2 is 2′-OMe, n3 is 7, n4 is 0, B3 is 2′-OMe, n5 is 3, B1′ is 2′-OMe or 2′-F, q1 is 9, T1′ is 2′-F, q2 is 1, B2′ is 2′-OMe or 2′-F, q3 is 4, T2′ is 2′-F, q4 is 2, B3′ is 2′-OMe or 2′-F, q5 is 5, T3′ is 2′-F, q6 is 1, B4′ is 2′-F, and q7 is 1; with two phosphorothioate internucleotide linkage modifications within position 1-5 of the sense strand (counting from the 5′-end of the sense strand), and two phosphorothioate internucleotide linkage modifications at positions 1 and 2 and two phosphorothioate internucleotide linkage modifications within positions 18-23 of the antisense strand (counting from the 5′-end of the antisense strand). The RNAi agent also comprises a 5′-PS.


In one embodiment, B1 is 2′-OMe or 2′-F, n1 is 8, T1 is 2′F, n2 is 3, B2 is 2′-OMe, n3 is 7, n4 is 0, B3 is 2′-OMe, n5 is 3, B1′ is 2′-OMe or 2′-F, q1 is 9, T1′ is 2′-F, q2 is 1, B2′ is 2′-OMe or 2′-F, q3 is 4, T2′ is 2′-F, q4 is 2, B3′ is 2′-OMe or 2′-F, q5 is 5, T3′ is 2′-F, q6 is 1, B4′ is 2′-F, and q7 is 1; with two phosphorothioate internucleotide linkage modifications within position 1-5 of the sense strand (counting from the 5′-end of the sense strand), and two phosphorothioate internucleotide linkage modifications at positions 1 and 2 and two phosphorothioate internucleotide linkage modifications within positions 18-23 of the antisense strand (counting from the 5′-end of the antisense strand). The RNAi agent also comprises a 5′-VP. The 5′-VP may be 5′-E-VP, 5′-Z-VP, or combination thereof.


In one embodiment, B1 is 2′-OMe or 2′-F, n1 is 8, T1 is 2′F, n2 is 3, B2 is 2′-OMe, n3 is 7, n4 is 0, B3 is 2′-OMe, n5 is 3, B1′ is 2′-OMe or 2′-F, q1 is 9, T1′ is 2′-F, q2 is 1, B2′ is 2′-OMe or 2′-F, q3 is 4, T2′ is 2′-F, q4 is 2, B3′ is 2′-OMe or 2′-F, q5 is 5, T3′ is 2′-F, q6 is 1, B4′ is 2′-F, and q7 is 1; with two phosphorothioate internucleotide linkage modifications within position 1-5 of the sense strand (counting from the 5′-end of the sense strand), and two phosphorothioate internucleotide linkage modifications at positions 1 and 2 and two phosphorothioate internucleotide linkage modifications within positions 18-23 of the antisense strand (counting from the 5′-end of the antisense strand). The RNAi agent also comprises a 5′-PS2.


In one embodiment, B1 is 2′-OMe or 2′-F, n1 is 8, T1 is 2′F, n2 is 3, B2 is 2′-OMe, n3 is 7, n4 is 0, B3 is 2′-OMe, n5 is 3, B1′ is 2′-OMe or 2′-F, q1 is 9, T1′ is 2′-F, q2 is 1, B2′ is 2′-OMe or 2′-F, q3 is 4, T2′ is 2′-F, q4 is 2, B3′ is 2′-OMe or 2′-F, q5 is 5, T3′ is 2′-F, q6 is 1, B4′ is 2′-F, and q7 is 1; with two phosphorothioate internucleotide linkage modifications within position 1-5 of the sense strand (counting from the 5′-end of the sense strand), and two phosphorothioate internucleotide linkage modifications at positions 1 and 2 and two phosphorothioate internucleotide linkage modifications within positions 18-23 of the antisense strand (counting from the 5′-end of the antisense strand). The RNAi agent also comprises a 5′-deoxy-5′-C-malonyl.


In one embodiment, B1 is 2′-OMe or 2′-F, n1 is 8, T1 is 2′F, n2 is 3, B2 is 2′-OMe, n3 is 7, n4 is 0, B3 is 2′-OMe, n5 is 3, B1′ is 2′-OMe or 2′-F, q1 is 9, T1′ is 2′-F, q2 is 1, B2′ is 2′-OMe or 2′-F, q3 is 4, q4 is 0, B3′ is 2′-OMe or 2′-F, q5 is 7, T3′ is 2′-F, q6 is 1, B4′ is 2′-F, and q7 is 1. The RNAi agent also comprises a 5′-P.


In one embodiment, B1 is 2′-OMe or 2′-F, n1 is 8, T1 is 2′F, n2 is 3, B2 is 2′-OMe, n3 is 7, n4 is 0, B3 is 2′-OMe, n5 is 3, B1′ is 2′-OMe or 2′-F, q1 is 9, T1′ is 2′-F, q2 is 1, B2′ is 2′-OMe or 2′-F, q3 is 4, q4 is 0, B3′ is 2′-OMe or 2′-F, q5 is 7, T3′ is 2′-F, q6 is 1, B4′ is 2′-F, and q7 is 1. The RNAi agent also comprises a 5′-PS.


In one embodiment, B1 is 2′-OMe or 2′-F, n1 is 8, T1 is 2′F, n2 is 3, B2 is 2′-OMe, n3 is 7, n4 is 0, B3 is 2′-OMe, n5 is 3, B1′ is 2′-OMe or 2′-F, q1 is 9, T1′ is 2′-F, q2 is 1, B2′ is 2′-OMe or 2′-F, q3 is 4, q4 is 0, B3′ is 2′-OMe or 2′-F, q5 is 7, T3′ is 2′-F, q6 is 1, B4′ is 2′-F, and q7 is 1. The RNAi agent also comprises a 5′-VP. The 5′-VP may be 5′-E-VP, 5′-Z-VP, or combination thereof.


In one embodiment, B1 is 2′-OMe or 2′-F, n1 is 8, T1 is 2′F, n2 is 3, B2 is 2′-OMe, n3 is 7, n4 is 0, B3 is 2′-OMe, n5 is 3, B1′ is 2′-OMe or 2′-F, q1 is 9, T1′ is 2′-F, q2 is 1, B2′ is 2′-OMe or 2′-F, q3 is 4, q4 is 0, B3′ is 2′-OMe or 2′-F, q5 is 7, T3′ is 2′-F, q6 is 1, B4′ is 2′-F, and q7 is 1. The RNAi agent also comprises a 5′-PS2.


In one embodiment, B1 is 2′-OMe or 2′-F, n1 is 8, T1 is 2′F, n2 is 3, B2 is 2′-OMe, n3 is 7, n4 is 0, B3 is 2′-OMe, n5 is 3, B1′ is 2′-OMe or 2′-F, q1 is 9, T1′ is 2′-F, q2 is 1, B2′ is 2′-OMe or 2′-F, q3 is 4, q4 is 0, B3′ is 2′-OMe or 2′-F, q5 is 7, T3′ is 2′-F, q6 is 1, B4′ is 2′-F, and q7 is 1. The RNAi agent also comprises a 5′-deoxy-5′-C-malonyl.


In one embodiment, B1 is 2′-OMe or 2′-F, n1 is 8, T1 is 2′F, n2 is 3, B2 is 2′-OMe, n3 is 7, n4 is 0, B3 is 2′-OMe, n5 is 3, B1′ is 2′-OMe or 2′-F, q1 is 9, T1′ is 2′-F, q2 is 1, B2′ is 2′-OMe or 2′-F, q3 is 4, q4 is 0, B3′ is 2′-OMe or 2′-F, q5 is 7, T3′ is 2′-F, q6 is 1, B4′ is 2′-F, and q7 is 1; with two phosphorothioate internucleotide linkage modifications within position 1-5 of the sense strand (counting from the 5′-end of the sense strand), and two phosphorothioate internucleotide linkage modifications at positions 1 and 2 and two phosphorothioate internucleotide linkage modifications within positions 18-23 of the antisense strand (counting from the 5′-end of the antisense strand). The RNAi agent also comprises a 5′-P.


In one embodiment, B1 is 2′-OMe or 2′-F, n1 is 8, T1 is 2′F, n2 is 3, B2 is 2′-OMe, n3 is 7, n4 is 0, B3 is 2′-OMe, n5 is 3, B1′ is 2′-OMe or 2′-F, q1 is 9, T1′ is 2′-F, q2 is 1, B2′ is 2′-OMe or 2′-F, q3 is 4, q4 is 0, B3′ is 2′-OMe or 2′-F, q5 is 7, T3′ is 2′-F, q6 is 1, B4′ is 2′-F, and q7 is 1; with two phosphorothioate internucleotide linkage modifications within position 1-5 of the sense strand (counting from the 5′-end of the sense strand), and two phosphorothioate internucleotide linkage modifications at positions 1 and 2 and two phosphorothioate internucleotide linkage modifications within positions 18-23 of the antisense strand (counting from the 5′-end of the antisense strand). The RNAi agent also comprises a 5′-PS.


In one embodiment, B1 is 2′-OMe or 2′-F, n1 is 8, T1 is 2′F, n2 is 3, B2 is 2′-OMe, n3 is 7, n4 is 0, B3 is 2′-OMe, n5 is 3, B1′ is 2′-OMe or 2′-F, q1 is 9, T1′ is 2′-F, q2 is 1, B2′ is 2′-OMe or 2′-F, q3 is 4, q4 is 0, B3′ is 2′-OMe or 2′-F, q5 is 7, T3′ is 2′-F, q6 is 1, B4′ is 2′-F, and q7 is 1; with two phosphorothioate internucleotide linkage modifications within position 1-5 of the sense strand (counting from the 5′-end of the sense strand), and two phosphorothioate internucleotide linkage modifications at positions 1 and 2 and two phosphorothioate internucleotide linkage modifications within positions 18-23 of the antisense strand (counting from the 5′-end of the antisense strand). The RNAi agent also comprises a 5′-VP. The 5′-VP may be 5′-E-VP, 5′-Z-VP, or combination thereof.


In one embodiment, B1 is 2′-OMe or 2′-F, n1 is 8, T1 is 2′F, n2 is 3, B2 is 2′-OMe, n3 is 7, n4 is 0, B3 is 2′-OMe, n5 is 3, B1′ is 2′-OMe or 2′-F, q1 is 9, T1′ is 2′-F, q2 is 1, B2′ is 2′-OMe or 2′-F, q3 is 4, q4 is 0, B3′ is 2′-OMe or 2′-F, q5 is 7, T3′ is 2′-F, q6 is 1, B4′ is 2′-F, and q7 is 1; with two phosphorothioate internucleotide linkage modifications within position 1-5 of the sense strand (counting from the 5′-end of the sense strand), and two phosphorothioate internucleotide linkage modifications at positions 1 and 2 and two phosphorothioate internucleotide linkage modifications within positions 18-23 of the antisense strand (counting from the 5′-end of the antisense strand). The RNAi agent also comprises a 5′-PS2.


In one embodiment, B1 is 2′-OMe or 2′-F, n1 is 8, T1 is 2′F, n2 is 3, B2 is 2′-OMe, n3 is 7, n4 is 0, B3 is 2′-OMe, n5 is 3, B1′ is 2′-OMe or 2′-F, q1 is 9, T1′ is 2′-F, q2 is 1, B2′ is 2′-OMe or 2′-F, q3 is 4, q4 is 0, B3′ is 2′-OMe or 2′-F, q5 is 7, T3′ is 2′-F, q6 is 1, B4′ is 2′-F, and q7 is 1; with two phosphorothioate internucleotide linkage modifications within position 1-5 of the sense strand (counting from the 5′-end of the sense strand), and two phosphorothioate internucleotide linkage modifications at positions 1 and 2 and two phosphorothioate internucleotide linkage modifications within positions 18-23 of the antisense strand (counting from the 5′-end of the antisense strand). The RNAi agent also comprises a 5′-deoxy-5′-C-malonyl.


In one embodiment, B1 is 2′-OMe or 2′-F, n1 is 8, T1 is 2′F, n2 is 3, B2 is 2′-OMe, n3 is 7, n4 is 0, B3 is 2′-OMe, n5 is 3, B1′ is 2′-OMe or 2′-F, q1 is 9, T1′ is 2′-F, q2 is 1, B2′ is 2′-OMe or 2′-F, q3 is 4, T2′ is 2′-F, q4 is 2, B3′ is 2′-OMe or 2′-F, q5 is 5, T3′ is 2′-F, q6 is 1, B4′ is 2′-OMe, and q7 is 1; with two phosphorothioate internucleotide linkage modifications within position 1-5 of the sense strand (counting from the 5′-end of the sense strand), and two phosphorothioate internucleotide linkage modifications at positions 1 and 2 and two phosphorothioate internucleotide linkage modifications within positions 18-23 of the antisense strand (counting from the 5′-end of the antisense strand). The RNAi agent also comprises a 5′-P and a targeting ligand. In one embodiment, the 5′-P is at the 5′-end of the antisense strand, and the targeting ligand is at the 3′-end of the sense strand.


In one embodiment, B1 is 2′-OMe or 2′-F, n1 is 8, T1 is 2′F, n2 is 3, B2 is 2′-OMe, n3 is 7, n4 is 0, B3 is 2′-OMe, n5 is 3, B1′ is 2′-OMe or 2′-F, q1 is 9, T1′ is 2′-F, q2 is 1, B2′ is 2′-OMe or 2′-F, q3 is 4, T2′ is 2′-F, q4 is 2, B3′ is 2′-OMe or 2′-F, q5 is 5, T3′ is 2′-F, q6 is 1, B4′ is 2′-OMe, and q7 is 1; with two phosphorothioate internucleotide linkage modifications within position 1-5 of the sense strand (counting from the 5′-end of the sense strand), and two phosphorothioate internucleotide linkage modifications at positions 1 and 2 and two phosphorothioate internucleotide linkage modifications within positions 18-23 of the antisense strand (counting from the 5′-end of the antisense strand). The RNAi agent also comprises a 5′-PS and a targeting ligand. In one embodiment, the 5′-PS is at the 5′-end of the antisense strand, and the targeting ligand is at the 3′-end of the sense strand.


In one embodiment, B1 is 2′-OMe or 2′-F, n1 is 8, T1 is 2′F, n2 is 3, B2 is 2′-OMe, n3 is 7, n4 is 0, B3 is 2′-OMe, n5 is 3, B1′ is 2′-OMe or 2′-F, q1 is 9, T1′ is 2′-F, q2 is 1, B2′ is 2′-OMe or 2′-F, q3 is 4, T2′ is 2′-F, q4 is 2, B3′ is 2′-OMe or 2′-F, q5 is 5, T3′ is 2′-F, q6 is 1, B4′ is 2′-OMe, and q7 is 1; with two phosphorothioate internucleotide linkage modifications within position 1-5 of the sense strand (counting from the 5′-end of the sense strand), and two phosphorothioate internucleotide linkage modifications at positions 1 and 2 and two phosphorothioate internucleotide linkage modifications within positions 18-23 of the antisense strand (counting from the 5′-end of the antisense strand). The RNAi agent also comprises a 5′-VP (e.g., a 5′-E-VP, 5′-Z-VP, or combination thereof), and a targeting ligand.


In one embodiment, the 5′-VP is at the 5′-end of the antisense strand, and the targeting ligand is at the 3′-end of the sense strand.


In one embodiment, B1 is 2′-OMe or 2′-F, n1 is 8, T1 is 2′F, n2 is 3, B2 is 2′-OMe, n3 is 7, n4 is 0, B3 is 2′-OMe, n5 is 3, B1′ is 2′-OMe or 2′-F, q1 is 9, T1′ is 2′-F, q2 is 1, B2′ is 2′-OMe or 2′-F, q3 is 4, T2′ is 2′-F, q4 is 2, B3′ is 2′-OMe or 2′-F, q5 is 5, T3′ is 2′-F, q6 is 1, B4′ is 2′-OMe, and q7 is 1; with two phosphorothioate internucleotide linkage modifications within position 1-5 of the sense strand (counting from the 5′-end of the sense strand), and two phosphorothioate internucleotide linkage modifications at positions 1 and 2 and two phosphorothioate internucleotide linkage modifications within positions 18-23 of the antisense strand (counting from the 5′-end of the antisense strand). The RNAi agent also comprises a 5′-PS2 and a targeting ligand. In one embodiment, the 5′-PS2 is at the 5′-end of the antisense strand, and the targeting ligand is at the 3′-end of the sense strand.


In one embodiment, B1 is 2′-OMe or 2′-F, n1 is 8, T1 is 2′F, n2 is 3, B2 is 2′-OMe, n3 is 7, n4 is 0, B3 is 2′-OMe, n5 is 3, B1′ is 2′-OMe or 2′-F, q1 is 9, T1′ is 2′-F, q2 is 1, B2′ is 2′-OMe or 2′-F, q3 is 4, T2′ is 2′-F, q4 is 2, B3′ is 2′-OMe or 2′-F, q5 is 5, T3′ is 2′-F, q6 is 1, B4′ is 2′-OMe, and q7 is 1; with two phosphorothioate internucleotide linkage modifications within position 1-5 of the sense strand (counting from the 5′-end of the sense strand), and two phosphorothioate internucleotide linkage modifications at positions 1 and 2 and two phosphorothioate internucleotide linkage modifications within positions 18-23 of the antisense strand (counting from the 5′-end of the antisense strand). The RNAi agent also comprises a 5′-deoxy-5′-C-malonyl and a targeting ligand. In one embodiment, the 5′-deoxy-5′-C-malonyl is at the 5′-end of the antisense strand, and the targeting ligand is at the 3′-end of the sense strand.


In one embodiment, B1 is 2′-OMe or 2′-F, n1 is 8, T1 is 2′F, n2 is 3, B2 is 2′-OMe, n3 is 7, n4 is 0, B3 is 2′-OMe, n5 is 3, B1′ is 2′-OMe or 2′-F, q1 is 9, T1′ is 2′-F, q2 is 1, B2′ is 2′-OMe or 2′-F, q3 is 4, q4 is 0, B3′ is 2′-OMe or 2′-F, q5 is 7, T3′ is 2′-F, q6 is 1, B4′ is 2′-OMe, and q7 is 1; with two phosphorothioate internucleotide linkage modifications within position 1-5 of the sense strand (counting from the 5′-end), and two phosphorothioate internucleotide linkage modifications at positions 1 and 2 and two phosphorothioate internucleotide linkage modifications within positions 18-23 of the antisense strand (counting from the 5′-end). The RNAi agent also comprises a 5′-P and a targeting ligand. In one embodiment, the 5′-P is at the 5′-end of the antisense strand, and the targeting ligand is at the 3′-end of the sense strand.


In one embodiment, B1 is 2′-OMe or 2′-F, n1 is 8, T1 is 2′F, n2 is 3, B2 is 2′-OMe, n3 is 7, n4 is 0, B3 is 2′-OMe, n5 is 3, B1′ is 2′-OMe or 2′-F, q1 is 9, T1′ is 2′-F, q2 is 1, B2′ is 2′-OMe or 2′-F, q3 is 4, q4 is 0, B3′ is 2′-OMe or 2′-F, q5 is 7, T3′ is 2′-F, q6 is 1, B4′ is 2′-OMe, and q7 is 1; with two phosphorothioate internucleotide linkage modifications within position 1-5 of the sense strand (counting from the 5′-end), and two phosphorothioate internucleotide linkage modifications at positions 1 and 2 and two phosphorothioate internucleotide linkage modifications within positions 18-23 of the antisense strand (counting from the 5′-end). The RNAi agent also comprises a 5′-PS and a targeting ligand. In one embodiment, the 5′-PS is at the 5′-end of the antisense strand, and the targeting ligand is at the 3′-end of the sense strand.


In one embodiment, B1 is 2′-OMe or 2′-F, n1 is 8, T1 is 2′F, n2 is 3, B2 is 2′-OMe, n3 is 7, n4 is 0, B3 is 2′-OMe, n5 is 3, B1′ is 2′-OMe or 2′-F, q1 is 9, T1′ is 2′-F, q2 is 1, B2′ is 2′-OMe or 2′-F, q3 is 4, q4 is 0, B3′ is 2′-OMe or 2′-F, q5 is 7, T3′ is 2′-F, q6 is 1, B4′ is 2′-OMe, and q7 is 1; with two phosphorothioate internucleotide linkage modifications within position 1-5 of the sense strand (counting from the 5′-end), and two phosphorothioate internucleotide linkage modifications at positions 1 and 2 and two phosphorothioate internucleotide linkage modifications within positions 18-23 of the antisense strand (counting from the 5′-end). The RNAi agent also comprises a 5′-VP (e.g., a 5′-E-VP, 5′-Z-VP, or combination thereof) and a targeting ligand. In one embodiment, the 5′-VP is at the 5′-end of the antisense strand, and the targeting ligand is at the 3′-end of the sense strand.


In one embodiment, B1 is 2′-OMe or 2′-F, n1 is 8, T1 is 2′F, n2 is 3, B2 is 2′-OMe, n3 is 7, n4 is 0, B3 is 2′-OMe, n5 is 3, B1′ is 2′-OMe or 2′-F, q1 is 9, T1′ is 2′-F, q2 is 1, B2′ is 2′-OMe or 2′-F, q3 is 4, q4 is 0, B3′ is 2′-OMe or 2′-F, q5 is 7, T3′ is 2′-F, q6 is 1, B4′ is 2′-OMe, and q7 is 1; with two phosphorothioate internucleotide linkage modifications within position 1-5 of the sense strand (counting from the 5′-end), and two phosphorothioate internucleotide linkage modifications at positions 1 and 2 and two phosphorothioate internucleotide linkage modifications within positions 18-23 of the antisense strand (counting from the 5′-end). The RNAi agent also comprises a 5′-PS2 and a targeting ligand. In one embodiment, the 5′-PS2 is at the 5′-end of the antisense strand, and the targeting ligand is at the 3′-end of the sense strand.


In one embodiment, B1 is 2′-OMe or 2′-F, n1 is 8, T1 is 2′F, n2 is 3, B2 is 2′-OMe, n3 is 7, n4 is 0, B3 is 2′-OMe, n5 is 3, B1′ is 2′-OMe or 2′-F, q1 is 9, T1′ is 2′-F, q2 is 1, B2′ is 2′-OMe or 2′-F, q3 is 4, q4 is 0, B3′ is 2′-OMe or 2′-F, q5 is 7, T3′ is 2′-F, q6 is 1, B4′ is 2′-OMe, and q7 is 1; with two phosphorothioate internucleotide linkage modifications within position 1-5 of the sense strand (counting from the 5′-end), and two phosphorothioate internucleotide linkage modifications at positions 1 and 2 and two phosphorothioate internucleotide linkage modifications within positions 18-23 of the antisense strand (counting from the 5′-end). The RNAi agent also comprises a 5′-deoxy-5′-C-malonyl and a targeting ligand. In one embodiment, the 5′-deoxy-5′-C-malonyl is at the 5′-end of the antisense strand, and the targeting ligand is at the 3′-end of the sense strand.


In one embodiment, B1 is 2′-OMe or 2′-F, n1 is 8, T1 is 2′F, n2 is 3, B2 is 2′-OMe, n3 is 7, n4 is 0, B3 is 2′-OMe, n5 is 3, B1′ is 2′-OMe or 2′-F, q1 is 9, T1′ is 2′-F, q2 is 1, B2′ is 2′-OMe or 2′-F, q3 is 4, T2′ is 2′-F, q4 is 2, B3′ is 2′-OMe or 2′-F, q5 is 5, T3′ is 2′-F, q6 is 1, B4′ is 2′-F, and q7 is 1; with two phosphorothioate internucleotide linkage modifications within position 1-5 of the sense strand (counting from the 5′-end of the sense strand), and two phosphorothioate internucleotide linkage modifications at positions 1 and 2 and two phosphorothioate internucleotide linkage modifications within positions 18-23 of the antisense strand (counting from the 5′-end of the antisense strand). The RNAi agent also comprises a 5′-P and a targeting ligand. In one embodiment, the 5′-P is at the 5′-end of the antisense strand, and the targeting ligand is at the 3′-end of the sense strand.


In one embodiment, B1 is 2′-OMe or 2′-F, n1 is 8, T1 is 2′F, n2 is 3, B2 is 2′-OMe, n3 is 7, n4 is 0, B3 is 2′-OMe, n5 is 3, B1′ is 2′-OMe or 2′-F, q1 is 9, T1′ is 2′-F, q2 is 1, B2′ is 2′-OMe or 2′-F, q3 is 4, T2′ is 2′-F, q4 is 2, B3′ is 2′-OMe or 2′-F, q5 is 5, T3′ is 2′-F, q6 is 1, B4′ is 2′-F, and q7 is 1; with two phosphorothioate internucleotide linkage modifications within position 1-5 of the sense strand (counting from the 5′-end of the sense strand), and two phosphorothioate internucleotide linkage modifications at positions 1 and 2 and two phosphorothioate internucleotide linkage modifications within positions 18-23 of the antisense strand (counting from the 5′-end of the antisense strand). The RNAi agent also comprises a 5′-PS and a targeting ligand. In one embodiment, the 5′-PS is at the 5′-end of the antisense strand, and the targeting ligand is at the 3′-end of the sense strand.


In one embodiment, B1 is 2′-OMe or 2′-F, n1 is 8, T1 is 2′F, n2 is 3, B2 is 2′-OMe, n3 is 7, n4 is 0, B3 is 2′-OMe, n5 is 3, B1′ is 2′-OMe or 2′-F, q1 is 9, T1′ is 2′-F, q2 is 1, B2′ is 2′-OMe or 2′-F, q3 is 4, T2′ is 2′-F, q4 is 2, B3′ is 2′-OMe or 2′-F, q5 is 5, T3′ is 2′-F, q6 is 1, B4′ is 2′-F, and q7 is 1; with two phosphorothioate internucleotide linkage modifications within position 1-5 of the sense strand (counting from the 5′-end of the sense strand), and two phosphorothioate internucleotide linkage modifications at positions 1 and 2 and two phosphorothioate internucleotide linkage modifications within positions 18-23 of the antisense strand (counting from the 5′-end of the antisense strand). The RNAi agent also comprises a 5′-VP (e.g., a 5′-E-VP, 5′-Z-VP, or combination thereof) and a targeting ligand. In one embodiment, the 5′-VP is at the 5′-end of the antisense strand, and the targeting ligand is at the 3′-end of the sense strand.


In one embodiment, B1 is 2′-OMe or 2′-F, n1 is 8, T1 is 2′F, n2 is 3, B2 is 2′-OMe, n3 is 7, n4 is 0, B3 is 2′-OMe, n5 is 3, B1′ is 2′-OMe or 2′-F, q1 is 9, T1′ is 2′-F, q2 is 1, B2′ is 2′-OMe or 2′-F, q3 is 4, T2′ is 2′-F, q4 is 2, B3′ is 2′-OMe or 2′-F, q5 is 5, T3′ is 2′-F, q6 is 1, B4′ is 2′-F, and q7 is 1; with two phosphorothioate internucleotide linkage modifications within position 1-5 of the sense strand (counting from the 5′-end of the sense strand), and two phosphorothioate internucleotide linkage modifications at positions 1 and 2 and two phosphorothioate internucleotide linkage modifications within positions 18-23 of the antisense strand (counting from the 5′-end of the antisense strand). The RNAi agent also comprises a 5′-PS2 and a targeting ligand. In one embodiment, the 5′-PS2 is at the 5′-end of the antisense strand, and the targeting ligand is at the 3′-end of the sense strand.


In one embodiment, B1 is 2′-OMe or 2′-F, n1 is 8, T1 is 2′F, n2 is 3, B2 is 2′-OMe, n3 is 7, n4 is 0, B3 is 2′-OMe, n5 is 3, B1′ is 2′-OMe or 2′-F, q1 is 9, T1′ is 2′-F, q2 is 1, B2′ is 2′-OMe or 2′-F, q3 is 4, T2′ is 2′-F, q4 is 2, B3′ is 2′-OMe or 2′-F, q5 is 5, T3′ is 2′-F, q6 is 1, B4′ is 2′-F, and q7 is 1; with two phosphorothioate internucleotide linkage modifications within position 1-5 of the sense strand (counting from the 5′-end of the sense strand), and two phosphorothioate internucleotide linkage modifications at positions 1 and 2 and two phosphorothioate internucleotide linkage modifications within positions 18-23 of the antisense strand (counting from the 5′-end of the antisense strand). The RNAi agent also comprises a 5′-deoxy-5′-C-malonyl and a targeting ligand. In one embodiment, the 5′-deoxy-5′-C-malonyl is at the 5′-end of the antisense strand, and the targeting ligand is at the 3′-end of the sense strand.


In one embodiment, B1 is 2′-OMe or 2′-F, n1 is 8, T1 is 2′F, n2 is 3, B2 is 2′-OMe, n3 is 7, n4 is 0, B3 is 2′-OMe, n5 is 3, B1′ is 2′-OMe or 2′-F, q1 is 9, T1′ is 2′-F, q2 is 1, B2′ is 2′-OMe or 2′-F, q3 is 4, q4 is 0, B3′ is 2′-OMe or 2′-F, q5 is 7, T3′ is 2′-F, q6 is 1, B4′ is 2′-F, and q7 is 1; with two phosphorothioate internucleotide linkage modifications within position 1-5 of the sense strand (counting from the 5′-end of the sense strand), and two phosphorothioate internucleotide linkage modifications at positions 1 and 2 and two phosphorothioate internucleotide linkage modifications within positions 18-23 of the antisense strand (counting from the 5′-end of the antisense strand). The RNAi agent also comprises a 5′-P and a targeting ligand. In one embodiment, the 5′-P is at the 5′-end of the antisense strand, and the targeting ligand is at the 3′-end of the sense strand.


In one embodiment, B1 is 2′-OMe or 2′-F, n1 is 8, T1 is 2′F, n2 is 3, B2 is 2′-OMe, n3 is 7, n4 is 0, B3 is 2′-OMe, n5 is 3, B1′ is 2′-OMe or 2′-F, q1 is 9, T1′ is 2′-F, q2 is 1, B2′ is 2′-OMe or 2′-F, q3 is 4, q4 is 0, B3′ is 2′-OMe or 2′-F, q5 is 7, T3′ is 2′-F, q6 is 1, B4′ is 2′-F, and q7 is 1; with two phosphorothioate internucleotide linkage modifications within position 1-5 of the sense strand (counting from the 5′-end of the sense strand), and two phosphorothioate internucleotide linkage modifications at positions 1 and 2 and two phosphorothioate internucleotide linkage modifications within positions 18-23 of the antisense strand (counting from the 5′-end of the antisense strand). The RNAi agent also comprises a 5′-PS and a targeting ligand. In one embodiment, the 5′-PS is at the 5′-end of the antisense strand, and the targeting ligand is at the 3′-end of the sense strand.


In one embodiment, B1 is 2′-OMe or 2′-F, n1 is 8, T1 is 2′F, n2 is 3, B2 is 2′-OMe, n3 is 7, n4 is 0, B3 is 2′-OMe, n5 is 3, B1′ is 2′-OMe or 2′-F, q1 is 9, T1′ is 2′-F, q2 is 1, B2′ is 2′-OMe or 2′-F, q3 is 4, q4 is 0, B3′ is 2′-OMe or 2′-F, q5 is 7, T3′ is 2′-F, q6 is 1, B4′ is 2′-F, and q7 is 1; with two phosphorothioate internucleotide linkage modifications within position 1-5 of the sense strand (counting from the 5′-end of the sense strand), and two phosphorothioate internucleotide linkage modifications at positions 1 and 2 and two phosphorothioate internucleotide linkage modifications within positions 18-23 of the antisense strand (counting from the 5′-end of the antisense strand). The RNAi agent also comprises a 5′-VP (e.g., a 5′-E-VP, 5′-Z-VP, or combination thereof) and a targeting ligand. In one embodiment, the 5′-VP is at the 5′-end of the antisense strand, and the targeting ligand is at the 3′-end of the sense strand.


In one embodiment, B1 is 2′-OMe or 2′-F, n1 is 8, T1 is 2′F, n2 is 3, B2 is 2′-OMe, n3 is 7, n4 is 0, B3 is 2′-OMe, n5 is 3, B1′ is 2′-OMe or 2′-F, q1 is 9, T1′ is 2′-F, q2 is 1, B2′ is 2′-OMe or 2′-F, q3 is 4, q4 is 0, B3′ is 2′-OMe or 2′-F, q5 is 7, T3′ is 2′-F, q6 is 1, B4′ is 2′-F, and q7 is 1; with two phosphorothioate internucleotide linkage modifications within position 1-5 of the sense strand (counting from the 5′-end of the sense strand), and two phosphorothioate internucleotide linkage modifications at positions 1 and 2 and two phosphorothioate internucleotide linkage modifications within positions 18-23 of the antisense strand (counting from the 5′-end of the antisense strand). The RNAi agent also comprises a 5′-PS2 and a targeting ligand. In one embodiment, the 5′-PS2 is at the 5′-end of the antisense strand, and the targeting ligand is at the 3′-end of the sense strand.


In one embodiment, B1 is 2′-OMe or 2′-F, n1 is 8, T1 is 2′F, n2 is 3, B2 is 2′-OMe, n3 is 7, n4 is 0, B3 is 2′-OMe, n5 is 3, B1′ is 2′-OMe or 2′-F, q1 is 9, T1′ is 2′-F, q2 is 1, B2′ is 2′-OMe or 2′-F, q3 is 4, q4 is 0, B3′ is 2′-OMe or 2′-F, q5 is 7, T3′ is 2′-F, q6 is 1, B4′ is 2′-F, and q7 is 1; with two phosphorothioate internucleotide linkage modifications within position 1-5 of the sense strand (counting from the 5′-end of the sense strand), and two phosphorothioate internucleotide linkage modifications at positions 1 and 2 and two phosphorothioate internucleotide linkage modifications within positions 18-23 of the antisense strand (counting from the 5′-end of the antisense strand). The RNAi agent also comprises a 5′-deoxy-5′-C-malonyl and a targeting ligand. In one embodiment, the 5′-deoxy-5′-C-malonyl is at the 5′-end of the antisense strand, and the targeting ligand is at the 3′-end of the sense strand.


In a particular embodiment, an RNAi agent of the present invention comprises:

    • (a) a sense strand having:
      • (i) a length of 21 nucleotides;
      • (ii) an ASGPR ligand attached to the 3′-end, wherein said ASGPR ligand comprises three GalNAc derivatives attached through a trivalent branched linker; and
      • (iii) 2′-F modifications at positions 1, 3, 5, 7, 9 to 11, 13, 17, 19, and 21, and 2′-OMe modifications at positions 2, 4, 6, 8, 12, 14 to 16, 18, and 20 (counting from the 5′ end); and
    • (b) an antisense strand having:
      • (i) a length of 23 nucleotides;
      • (ii) 2′-OMe modifications at positions 1, 3, 5, 9, 11 to 13, 15, 17, 19, 21, and 23, and 2′F modifications at positions 2, 4, 6 to 8, 10, 14, 16, 18, 20, and 22 (counting from the 5′ end); and
      • (iii) phosphorothioate internucleotide linkages between nucleotide positions 21 and 22, and between nucleotide positions 22 and 23 (counting from the 5′ end);
    • wherein the dsRNA agents have a two nucleotide overhang at the 3′-end of the antisense strand, and a blunt end at the 5′-end of the antisense strand.


In another particular embodiment, an RNAi agent of the present invention comprises:

    • (a) a sense strand having:
      • (i) a length of 21 nucleotides;
      • (ii) an ASGPR ligand attached to the 3′-end, wherein said ASGPR ligand comprises three GalNAc derivatives attached through a trivalent branched linker;
      • (iii) 2′-F modifications at positions 1, 3, 5, 7, 9 to 11, 13, 15, 17, 19, and 21, and 2′-OMe modifications at positions 2, 4, 6, 8, 12, 14, 16, 18, and 20 (counting from the 5′ end); and
      • (iv) phosphorothioate internucleotide linkages between nucleotide positions 1 and 2, and between nucleotide positions 2 and 3 (counting from the 5′ end);
    • and
    • (b) an antisense strand having:
      • (i) a length of 23 nucleotides;
      • (ii) 2′-OMe modifications at positions 1, 3, 5, 7, 9, 11 to 13, 15, 17, 19, and 21 to 23, and 2′F modifications at positions 2, 4, 6, 8, 10, 14, 16, 18, and 20 (counting from the 5′ end); and
      • (iii) phosphorothioate internucleotide linkages between nucleotide positions 1 and 2, between nucleotide positions 2 and 3, between nucleotide positions 21 and 22, and between nucleotide positions 22 and 23 (counting from the 5′ end);


        wherein the RNAi agents have a two nucleotide overhang at the 3′-end of the antisense strand, and a blunt end at the 5′-end of the antisense strand.


In another particular embodiment, a RNAi agent of the present invention comprises:

    • (a) a sense strand having:
      • (i) a length of 21 nucleotides;
      • (ii) an ASGPR ligand attached to the 3′-end, wherein said ASGPR ligand comprises three GalNAc derivatives attached through a trivalent branched linker;
      • (iii) 2′-OMe modifications at positions 1 to 6, 8, 10, and 12 to 21, 2′-F modifications at positions 7, and 9, and a deoxy-nucleotide (e.g. dT) at position 11 (counting from the 5′ end); and
      • (iv) phosphorothioate internucleotide linkages between nucleotide positions 1 and 2, and between nucleotide positions 2 and 3 (counting from the 5′ end);
    • and
    • (b) an antisense strand having:
      • (i) a length of 23 nucleotides;
      • (ii) 2′-OMe modifications at positions 1, 3, 7, 9, 11, 13, 15, 17, and 19 to 23, and 2′-F modifications at positions 2, 4 to 6, 8, 10, 12, 14, 16, and 18 (counting from the 5′ end); and
      • (iii) phosphorothioate internucleotide linkages between nucleotide positions 1 and 2, between nucleotide positions 2 and 3, between nucleotide positions 21 and 22, and between nucleotide positions 22 and 23 (counting from the 5′ end);


        wherein the RNAi agents have a two nucleotide overhang at the 3′-end of the antisense strand, and a blunt end at the 5′-end of the antisense strand.


In another particular embodiment, a RNAi agent of the present invention comprises:

    • (a) a sense strand having:
      • (i) a length of 21 nucleotides;
      • (ii) an ASGPR ligand attached to the 3′-end, wherein said ASGPR ligand comprises three GalNAc derivatives attached through a trivalent branched linker;
      • (iii) 2′-OMe modifications at positions 1 to 6, 8, 10, 12, 14, and 16 to 21, and 2′-F modifications at positions 7, 9, 11, 13, and 15; and
      • (iv) phosphorothioate internucleotide linkages between nucleotide positions 1 and 2, and between nucleotide positions 2 and 3 (counting from the 5′ end);
    • and
    • (b) an antisense strand having:
      • (i) a length of 23 nucleotides;
      • (ii) 2′-OMe modifications at positions 1, 5, 7, 9, 11, 13, 15, 17, 19, and 21 to 23, and 2′-F modifications at positions 2 to 4, 6, 8, 10, 12, 14, 16, 18, and 20 (counting from the 5′ end); and
      • (iii) phosphorothioate internucleotide linkages between nucleotide positions 1 and 2, between nucleotide positions 2 and 3, between nucleotide positions 21 and 22, and between nucleotide positions 22 and 23 (counting from the 5′ end);


        wherein the RNAi agents have a two nucleotide overhang at the 3′-end of the antisense strand, and a blunt end at the 5′-end of the antisense strand.


In another particular embodiment, a RNAi agent of the present invention comprises:

    • (a) a sense strand having:
      • (i) a length of 21 nucleotides;
      • (ii) an ASGPR ligand attached to the 3′-end, wherein said ASGPR ligand comprises three GalNAc derivatives attached through a trivalent branched linker;
      • (iii) 2′-OMe modifications at positions 1 to 9, and 12 to 21, and 2′-F modifications at positions 10, and 11; and
      • (iv) phosphorothioate internucleotide linkages between nucleotide positions 1 and 2, and between nucleotide positions 2 and 3 (counting from the 5′ end);
    • and
    • (b) an antisense strand having:
      • (i) a length of 23 nucleotides;
      • (ii) 2′-OMe modifications at positions 1, 3, 5, 7, 9, 11 to 13, 15, 17, 19, and 21 to 23, and 2′-F modifications at positions 2, 4, 6, 8, 10, 14, 16, 18, and 20 (counting from the 5′ end); and
      • (iii) phosphorothioate internucleotide linkages between nucleotide positions 1 and 2, between nucleotide positions 2 and 3, between nucleotide positions 21 and 22, and between nucleotide positions 22 and 23 (counting from the 5′ end);


        wherein the RNAi agents have a two nucleotide overhang at the 3′-end of the antisense strand, and a blunt end at the 5′-end of the antisense strand.


In another particular embodiment, a RNAi agent of the present invention comprises:

    • (a) a sense strand having:
      • (i) a length of 21 nucleotides;
      • (ii) an ASGPR ligand attached to the 3′-end, wherein said ASGPR ligand comprises three GalNAc derivatives attached through a trivalent branched linker;
      • (iii) 2′-F modifications at positions 1, 3, 5, 7, 9 to 11, and 13, and 2′-OMe modifications at positions 2, 4, 6, 8, 12, and 14 to 21; and
      • (iv) phosphorothioate internucleotide linkages between nucleotide positions 1 and 2, and between nucleotide positions 2 and 3 (counting from the 5′ end);
    • and
    • (b) an antisense strand having:
      • (i) a length of 23 nucleotides;
      • (ii) 2′-OMe modifications at positions 1, 3, 5 to 7, 9, 11 to 13, 15, 17 to 19, and 21 to 23, and 2′-F modifications at positions 2, 4, 8, 10, 14, 16, and 20 (counting from the 5′ end); and
      • (iii) phosphorothioate internucleotide linkages between nucleotide positions 1 and 2, between nucleotide positions 2 and 3, between nucleotide positions 21 and 22, and between nucleotide positions 22 and 23 (counting from the 5′ end);


        wherein the RNAi agents have a two nucleotide overhang at the 3′-end of the antisense strand, and a blunt end at the 5′-end of the antisense strand.


In another particular embodiment, a RNAi agent of the present invention comprises:

    • (a) a sense strand having:
      • (i) a length of 21 nucleotides;
      • (ii) an ASGPR ligand attached to the 3′-end, wherein said ASGPR ligand comprises three GalNAc derivatives attached through a trivalent branched linker;
      • (iii) 2′-OMe modifications at positions 1, 2, 4, 6, 8, 12, 14, 15, 17, and 19 to 21, and 2′-F modifications at positions 3, 5, 7, 9 to 11, 13, 16, and 18; and
      • (iv) phosphorothioate internucleotide linkages between nucleotide positions 1 and 2, and between nucleotide positions 2 and 3 (counting from the 5′ end);
    • and
    • (b) an antisense strand having:
      • (i) a length of 25 nucleotides;
      • (ii) 2′-OMe modifications at positions 1, 4, 6, 7, 9, 11 to 13, 15, 17, and 19 to 23, 2′-F modifications at positions 2, 3, 5, 8, 10, 14, 16, and 18, and desoxy-nucleotides (e.g. dT) at positions 24 and 25 (counting from the 5′ end); and
      • (iii) phosphorothioate internucleotide linkages between nucleotide positions 1 and 2, between nucleotide positions 2 and 3, between nucleotide positions 21 and 22, and between nucleotide positions 22 and 23 (counting from the 5′ end);


        wherein the RNAi agents have a four nucleotide overhang at the 3′-end of the antisense strand, and a blunt end at the 5′-end of the antisense strand.


In another particular embodiment, a RNAi agent of the present invention comprises:

    • (a) a sense strand having:
      • (i) a length of 21 nucleotides;
      • (ii) an ASGPR ligand attached to the 3′-end, wherein said ASGPR ligand comprises three GalNAc derivatives attached through a trivalent branched linker;
      • (iii) 2′-OMe modifications at positions 1 to 6, 8, and 12 to 21, and 2′-F modifications at positions 7, and 9 to 11; and
      • (iv) phosphorothioate internucleotide linkages between nucleotide positions 1 and 2, and between nucleotide positions 2 and 3 (counting from the 5′ end);
    • and
    • (b) an antisense strand having:
      • (i) a length of 23 nucleotides;
      • (ii) 2′-OMe modifications at positions 1, 3 to 5, 7, 8, 10 to 13, 15, and 17 to 23, and 2′-F modifications at positions 2, 6, 9, 14, and 16 (counting from the 5′ end); and
      • (iii) phosphorothioate internucleotide linkages between nucleotide positions 1 and 2, between nucleotide positions 2 and 3, between nucleotide positions 21 and 22, and between nucleotide positions 22 and 23 (counting from the 5′ end);


        wherein the RNAi agents have a two nucleotide overhang at the 3′-end of the antisense strand, and a blunt end at the 5′-end of the antisense strand.


In another particular embodiment, a RNAi agent of the present invention comprises:

    • (a) a sense strand having:
      • (i) a length of 21 nucleotides;
      • (ii) an ASGPR ligand attached to the 3′-end, wherein said ASGPR ligand comprises three GalNAc derivatives attached through a trivalent branched linker;
      • (iii) 2′-OMe modifications at positions 1 to 6, 8, and 12 to 21, and 2′-F modifications at positions 7, and 9 to 11; and
      • (iv) phosphorothioate internucleotide linkages between nucleotide positions 1 and 2, and between nucleotide positions 2 and 3 (counting from the 5′ end);
    • and
    • (b) an antisense strand having:
      • (i) a length of 23 nucleotides;
      • (ii) 2′-OMe modifications at positions 1, 3 to 5, 7, 10 to 13, 15, and 17 to 23, and 2′-F modifications at positions 2, 6, 8, 9, 14, and 16 (counting from the 5′ end); and
      • (iii) phosphorothioate internucleotide linkages between nucleotide positions 1 and 2, between nucleotide positions 2 and 3, between nucleotide positions 21 and 22, and between nucleotide positions 22 and 23 (counting from the 5′ end);


        wherein the RNAi agents have a two nucleotide overhang at the 3′-end of the antisense strand, and a blunt end at the 5′-end of the antisense strand.


In another particular embodiment, a RNAi agent of the present invention comprises:

    • (a) a sense strand having:
      • (i) a length of 19 nucleotides;
      • (ii) an ASGPR ligand attached to the 3′-end, wherein said ASGPR ligand comprises three GalNAc derivatives attached through a trivalent branched linker;
      • (iii) 2′-OMe modifications at positions 1 to 4, 6, and 10 to 19, and 2′-F modifications at positions 5, and 7 to 9; and
      • (iv) phosphorothioate internucleotide linkages between nucleotide positions 1 and 2, and between nucleotide positions 2 and 3 (counting from the 5′ end);
    • and
    • (b) an antisense strand having:
      • (i) a length of 21 nucleotides;
      • (ii) 2′-OMe modifications at positions 1, 3 to 5, 7, 10 to 13, 15, and 17 to 21, and 2′-F modifications at positions 2, 6, 8, 9, 14, and 16 (counting from the 5′ end); and
      • (iii) phosphorothioate internucleotide linkages between nucleotide positions 1 and 2, between nucleotide positions 2 and 3, between nucleotide positions 19 and 20, and between nucleotide positions 20 and 21 (counting from the 5′ end);


        wherein the RNAi agents have a two nucleotide overhang at the 3′-end of the antisense strand, and a blunt end at the 5′-end of the antisense strand.


In certain embodiments, the iRNA for use in the methods of the invention is an agent selected from agents listed in any one of Tables 2-7, 13, 16, 19 and 20. These agents may further comprise a ligand.


III. iRNAs Conjugated to Ligands

Another modification of the RNA of an iRNA of the invention involves chemically linking to the iRNA one or more ligands, moieties or conjugates that enhance the activity, cellular distribution, or cellular uptake of the iRNA e.g., into a cell. Such moieties include but are not limited to lipid moieties such as a cholesterol moiety (Letsinger et al., Proc. Natl. Acid. Sci. USA, 1989, 86: 6553-6556). In other embodiments, the ligand is cholic acid (Manoharan et al., Biorg. Med. Chem. Let., 1994, 4:1053-1060), a thioether, e.g., beryl-S-tritylthiol (Manoharan et al., Ann. N.Y. Acad. Sci., 1992, 660:306-309; Manoharan et al., Biorg. Med. Chem. Let., 1993, 3:2765-2770), a thiocholesterol (Oberhauser et al., Nucl. Acids Res., 1992, 20:533-538), an aliphatic chain, e.g., dodecandiol or undecyl residues (Saison-Behmoaras et al., EMBO J, 1991, 10:1111-1118; Kabanov et al., FEBS Lett., 1990, 259:327-330; Svinarchuk et al., Biochimie, 1993, 75:49-54), a phospholipid, e.g., di-hexadecyl-rac-glycerol or triethyl-ammonium 1,2-di-O-hexadecyl-rac-glycero-3-phosphonate (Manoharan et al., Tetrahedron Lett., 1995, 36:3651-3654; Shea et al., Nucl. Acids Res., 1990, 18:3777-3783), a polyamine or a polyethylene glycol chain (Manoharan et al., Nucleosides & Nucleotides, 1995, 14:969-973), or adamantane acetic acid (Manoharan et al., Tetrahedron Lett., 1995, 36:3651-3654), a palmityl moiety (Mishra et al., Biochim. Biophys. Acta, 1995, 1264:229-237), or an octadecylamine or hexylamino-carbonyloxycholesterol moiety (Crooke et al., J. Pharmacol. Exp. Ther., 1996, 277:923-937).


In certain embodiments, a ligand alters the distribution, targeting, or lifetime of an iRNA agent into which it is incorporated. In certain embodiments a ligand provides an enhanced affinity for a selected target, e.g., molecule, cell or cell type, compartment, e.g., a cellular or organ compartment, tissue, organ or region of the body, as, e.g., compared to a species absent such a ligand. In some embodiments, ligands do not take part in duplex pairing in a duplexed nucleic acid.


Ligands can include a naturally occurring substance, such as a protein (e.g., human serum albumin (HSA), low-density lipoprotein (LDL), or globulin); carbohydrate (e.g., a dextran, pullulan, chitin, chitosan, inulin, cyclodextrin, N-acetylglucosamine, N-acetylgalactosamine, or hyaluronic acid); or a lipid. The ligand can also be a recombinant or synthetic molecule, such as a synthetic polymer, e.g., a synthetic polyamino acid. Examples of polyamino acids include polyamino acid is a polylysine (PLL), poly L-aspartic acid, poly L-glutamic acid, styrene-maleic acid anhydride copolymer, poly(L-lactide-co-glycolied) copolymer, divinyl ether-maleic anhydride copolymer, N-(2-hydroxypropyl)methacrylamide copolymer (HMPA), polyethylene glycol (PEG), polyvinyl alcohol (PVA), polyurethane, poly(2-ethylacryllic acid), N-isopropylacrylamide polymers, or polyphosphazine. Example of polyamines include: polyethylenimine, polylysine (PLL), spermine, spermidine, polyamine, pseudopeptide-polyamine, peptidomimetic polyamine, dendrimer polyamine, arginine, amidine, protamine, cationic lipid, cationic porphyrin, quaternary salt of a polyamine, or an alpha helical peptide.


Ligands can also include targeting groups, e.g., a cell or tissue targeting agent, e.g., a lectin, glycoprotein, lipid or protein, e.g., an antibody, that binds to a specified cell type such as a kidney cell. A targeting group can be a thyrotropin, melanotropin, lectin, glycoprotein, surfactant protein A, Mucin carbohydrate, multivalent lactose, multivalent galactose, N-acetyl-galactosamine, N-acetyl-glucosamine multivalent mannose, multivalent fucose, glycosylated polyaminoacids, multivalent galactose, transferrin, bisphosphonate, polyglutamate, polyaspartate, a lipid, cholesterol, a steroid, bile acid, folate, vitamin B12, vitamin A, biotin, or an RGD peptide or RGD peptide mimetic. In certain embodiments, the ligand is a multivalent galactose, e.g., an N-acetyl-galactosamine.


Other examples of ligands include dyes, intercalating agents (e.g. acridines), cross-linkers (e.g. psoralene, mitomycin C), porphyrins (TPPC4, texaphyrin, Sapphyrin), polycyclic aromatic hydrocarbons (e.g., phenazine, dihydrophenazine), artificial endonucleases (e.g. EDTA), lipophilic molecules, e.g., cholesterol, cholic acid, adamantane acetic acid, 1-pyrene butyric acid, dihydrotestosterone, 1,3-Bis-O(hexadecyl)glycerol, geranyloxyhexyl group, hexadecylglycerol, borneol, menthol, 1,3-propanediol, heptadecyl group, palmitic acid, myristic acid, O3-(oleoyl)lithocholic acid, O3-(oleoyl)cholenic acid, dimethoxytrityl, or phenoxazine) and peptide conjugates (e.g., antennapedia peptide, Tat peptide), alkylating agents, phosphate, amino, mercapto, PEG (e.g., PEG-40K), MPEG, [MPEG]2, polyamino, alkyl, substituted alkyl, radiolabeled markers, enzymes, haptens (e.g. biotin), transport/absorption facilitators (e.g., aspirin, vitamin E, folic acid), synthetic ribonucleases (e.g., imidazole, bisimidazole, histamine, imidazole clusters, acridine-imidazole conjugates, Eu3+ complexes of tetraazamacrocycles), dinitrophenyl, HRP, or AP.


Ligands can be proteins, e.g., glycoproteins, or peptides, e.g., molecules having a specific affinity for a co-ligand, or antibodies e.g., an antibody, that binds to a specified cell type such as a hepatic cell. Ligands can also include hormones and hormone receptors. They can also include non-peptidic species, such as lipids, lectins, carbohydrates, vitamins, cofactors, multivalent lactose, multivalent galactose, N-acetyl-galactosamine, N-acetyl-glucosamine multivalent mannose, or multivalent fucose. The ligand can be, for example, a lipopolysaccharide, an activator of p38 MAP kinase, or an activator of NF-κB.


The ligand can be a substance, e.g., a drug, which can increase the uptake of the iRNA agent into the cell, for example, by disrupting the cell's cytoskeleton, e.g., by disrupting the cell's microtubules, microfilaments, or intermediate filaments. The drug can be, for example, taxol, vincristine, vinblastine, cytochalasin, nocodazole, japlakinolide, latrunculin A, phalloidin, swinholide A, indanocine, or myoservin.


In some embodiments, a ligand attached to an iRNA as described herein acts as a pharmacokinetic modulator (PK modulator). PK modulators include lipophiles, bile acids, steroids, phospholipid analogues, peptides, protein binding agents, PEG, vitamins, etc. Exemplary PK modulators include, but are not limited to, cholesterol, fatty acids, cholic acid, lithocholic acid, dialkylglycerides, diacylglyceride, phospholipids, sphingolipids, naproxen, ibuprofen, vitamin E, biotin. Oligonucleotides that comprise a number of phosphorothioate linkages are also known to bind to serum protein, thus short oligonucleotides, e.g., oligonucleotides of about 5 bases, 10 bases, 15 bases, or 20 bases, comprising multiple of phosphorothioate linkages in the backbone are also amenable to the present invention as ligands (e.g. as PK modulating ligands). In addition, aptamers that bind serum components (e.g. serum proteins) are also suitable for use as PK modulating ligands in the embodiments described herein.


Ligand-conjugated iRNAs of the invention may be synthesized by the use of an oligonucleotide that bears a pendant reactive functionality, such as that derived from the attachment of a linking molecule onto the oligonucleotide (described below). This reactive oligonucleotide may be reacted directly with commercially-available ligands, ligands that are synthesized bearing any of a variety of protecting groups, or ligands that have a linking moiety attached thereto.


The oligonucleotides used in the conjugates of the present invention may be conveniently and routinely made through the well-known technique of solid-phase synthesis. Equipment for such synthesis is sold by several vendors including, for example, Applied Biosystems® (Foster City, Calif.). Any other methods for such synthesis known in the art may additionally or alternatively be employed. It is also known to use similar techniques to prepare other oligonucleotides, such as the phosphorothioates and alkylated derivatives.


In the ligand-conjugated iRNAs and ligand-molecule bearing sequence-specific linked nucleosides of the present invention, the oligonucleotides and oligonucleosides may be assembled on a suitable DNA synthesizer utilizing standard nucleotide or nucleoside precursors, or nucleotide or nucleoside conjugate precursors that already bear the linking moiety, ligand-nucleotide or nucleoside-conjugate precursors that already bear the ligand molecule, or non-nucleoside ligand-bearing building blocks.


When using nucleotide-conjugate precursors that already bear a linking moiety, the synthesis of the sequence-specific linked nucleosides is typically completed, and the ligand molecule is then reacted with the linking moiety to form the ligand-conjugated oligonucleotide. In some embodiments, the oligonucleotides or linked nucleosides of the present invention are synthesized by an automated synthesizer using phosphoramidites derived from ligand-nucleoside conjugates in addition to the standard phosphoramidites and non-standard phosphoramidites that are commercially available and routinely used in oligonucleotide synthesis.


A. Lipid Conjugates


In certain embodiments, the ligand or conjugate is a lipid or lipid-based molecule. Such a lipid or lipid-based molecule may bind a serum protein, e.g., human serum albumin (HSA). An HSA binding ligand allows for distribution of the conjugate to a target tissue, e.g., a non-kidney target tissue of the body. For example, the target tissue can be the liver, including parenchymal cells of the liver. Other molecules that can bind HSA can also be used as ligands. For example, naproxen or aspirin can be used. A lipid or lipid-based ligand can (a) increase resistance to degradation of the conjugate, (b) increase targeting or transport into a target cell or cell membrane, or (c) can be used to adjust binding to a serum protein, e.g., HSA.


A lipid based ligand can be used to inhibit, e.g., control the binding of the conjugate to a target tissue. For example, a lipid or lipid-based ligand that binds to HSA more strongly will be less likely to be targeted to the kidney and therefore less likely to be cleared from the body. A lipid or lipid-based ligand that binds to HSA less strongly can be used to target the conjugate to the kidney.


In certain embodiments, the lipid based ligand binds HSA. In one embodiment, it binds HSA with a sufficient affinity such that the conjugate will be distributed to a non-kidney tissue. However, it is preferred that the affinity not be so strong that the HSA-ligand binding cannot be reversed.


In other embodiments, the lipid based ligand binds HSA weakly or not at all, such that the conjugate will be distributed to the kidney. Other moieties that target to kidney cells can also be used in place of, or in addition to, the lipid based ligand.


In another aspect, the ligand is a moiety, e.g., a vitamin, which is taken up by a target cell, e.g., a proliferating cell. These are particularly useful for treating disorders characterized by unwanted cell proliferation, e.g., of the malignant or non-malignant type, e.g., cancer cells. Exemplary vitamins include vitamin A, E, and K. Other exemplary vitamins include are B vitamin, e.g., folic acid, B12, riboflavin, biotin, pyridoxal or other vitamins or nutrients taken up by target cells such as liver cells. Also included are HSA and low density lipoprotein (LDL).


B. Cell Permeation Agents


In another aspect, the ligand is a cell-permeation agent, such as, a helical cell-permeation agent. In one embodiment, the agent is amphipathic. An exemplary agent is a peptide such as tat or antennopedia. If the agent is a peptide, it can be modified, including a peptidylmimetic, invertomers, non-peptide or pseudo-peptide linkages, and use of D-amino acids. In one embodiment, the helical agent is an alpha-helical agent, for example, having a lipophilic and a lipophobic phase.


The ligand can be a peptide or peptidomimetic. A peptidomimetic (also referred to herein as an oligopeptidomimetic) is a molecule capable of folding into a defined three-dimensional structure similar to a natural peptide. The attachment of peptide and peptidomimetics to iRNA agents can affect pharmacokinetic distribution of the iRNA, such as by enhancing cellular recognition and absorption. The peptide or peptidomimetic moiety can be about 5-50 amino acids long, e.g., about 5, 10, 15, 20, 25, 30, 35, 40, 45, or 50 amino acids long.


A peptide or peptidomimetic can be, for example, a cell permeation peptide, cationic peptide, amphipathic peptide, or hydrophobic peptide (e.g., consisting primarily of Tyr, Trp, or Phe). The peptide moiety can be a dendrimer peptide, constrained peptide or crosslinked peptide. In another alternative, the peptide moiety can include a hydrophobic membrane translocation sequence (MTS).


An exemplary hydrophobic MTS-containing peptide is RFGF having the amino acid sequence AAVALLPAVLLALLAP (SEQ ID NO:15). An RFGF analogue (e.g., amino acid sequence AALLPVLLAAP (SEQ ID NO:16) containing a hydrophobic MTS can also be a targeting moiety. The peptide moiety can be a “delivery” peptide, which can carry large polar molecules including peptides, oligonucleotides, and protein across cell membranes. For example, sequences from the HIV Tat protein (GRKKRRQRRRPPQ (SEQ ID NO:17) and the Drosophila Antennapedia protein (RQIKIWFQNRRMKWKK (SEQ ID NO:18) have been found to be capable of functioning as delivery peptides. A peptide or peptidomimetic can be encoded by a random sequence of DNA, such as a peptide identified from a phage-display library, or one-bead-one-compound (OBOC) combinatorial library (Lam et al., Nature, 354:82-84, 1991). Examples of a peptide or peptidomimetic tethered to a dsRNA agent via an incorporated monomer unit for cell targeting purposes is an arginine-glycine-aspartic acid (RGD)-peptide, or RGD mimic. A peptide moiety can range in length from about 5 amino acids to about 40 amino acids. The peptide moieties can have a structural modification, such as to increase stability or direct conformational properties. Any of the structural modifications described below can be utilized.


An RGD peptide for use in the compositions and methods of the invention may be linear or cyclic, and may be modified, e.g., glycosylated or methylated, to facilitate targeting to a specific tissue(s). RGD-containing peptides and peptidiomimemtics may include D-amino acids, as well as synthetic RGD mimics. In addition to RGD, one can use other moieties that target the integrin ligand, such as, PECAM-1 or VEGF.


A “cell permeation peptide” is capable of permeating a cell, e.g., a microbial cell, such as a bacterial or fungal cell, or a mammalian cell, such as a human cell. A microbial cell-permeating peptide can be, for example, an α-helical linear peptide (e.g., LL-37 or Ceropin P1), a disulfide bond-containing peptide (e.g., α-defensin, β-defensin or bactenecin), or a peptide containing only one or two dominating amino acids (e.g., PR-39 or indolicidin). A cell permeation peptide can also include a nuclear localization signal (NLS). For example, a cell permeation peptide can be a bipartite amphipathic peptide, such as MPG, which is derived from the fusion peptide domain of HIV-1 gp41 and the NLS of SV40 large T antigen (Simeoni et al., Nucl. Acids Res. 31:2717-2724, 2003).


C. Carbohydrate Conjugates


In some embodiments of the compositions and methods of the invention, an iRNA further comprises a carbohydrate. The carbohydrate conjugated iRNA is advantageous for the in vivo delivery of nucleic acids, as well as compositions suitable for in vivo therapeutic use, as described herein. As used herein, “carbohydrate” refers to a compound which is either a carbohydrate per se made up of one or more monosaccharide units having at least 6 carbon atoms (which can be linear, branched or cyclic) with an oxygen, nitrogen or sulfur atom bonded to each carbon atom; or a compound having as a part thereof a carbohydrate moiety made up of one or more monosaccharide units each having at least six carbon atoms (which can be linear, branched or cyclic), with an oxygen, nitrogen or sulfur atom bonded to each carbon atom. Representative carbohydrates include the sugars (mono-, di-, tri-, and oligosaccharides containing from about 4, 5, 6, 7, 8, or 9 monosaccharide units), and polysaccharides such as starches, glycogen, cellulose and polysaccharide gums. Specific monosaccharides include C5 and above (e.g., C5, C6, C7, or C8) sugars; di- and trisaccharides include sugars having two or three monosaccharide units (e.g., C5, C6, C7, or C8).


In certain embodiments, a carbohydrate conjugate for use in the compositions and methods of the invention is a monosaccharide.


In certain embodiments, the monosaccharide is an N-acetylgalactosamine (GalNAc). GalNAc conjugates, which comprise one or more N-acetylgalactosamine (GalNAc) derivatives, are described, for example, in U.S. Pat. No. 8,106,022, the entire content of which is hereby incorporated herein by reference. In some embodiments, the GalNAc conjugate serves as a ligand that targets the iRNA to particular cells. In some embodiments, the GalNAc conjugate targets the iRNA to liver cells, e.g., by serving as a ligand for the asialoglycoprotein receptor of liver cells (e.g., hepatocytes).


In some embodiments, the carbohydrate conjugate comprises one or more GalNAc derivatives. The GalNAc derivatives may be attached via a linker, e.g., a bivalent or trivalent branched linker. In some embodiments the GalNAc conjugate is conjugated to the 3′ end of the sense strand. In some embodiments, the GalNAc conjugate is conjugated to the iRNA agent (e.g., to the 3′ end of the sense strand) via a linker, e.g., a linker as described herein. In some embodiments the GalNAc conjugate is conjugated to the 5′ end of the sense strand. In some embodiments, the GalNAc conjugate is conjugated to the iRNA agent (e.g., to the 5′ end of the sense strand) via a linker, e.g., a linker as described herein.


In certain embodiments of the invention, the GalNAc or GalNAc derivative is attached to an iRNA agent of the invention via a monovalent linker. In some embodiments, the GalNAc or GalNAc derivative is attached to an iRNA agent of the invention via a bivalent linker. In yet other embodiments of the invention, the GalNAc or GalNAc derivative is attached to an iRNA agent of the invention via a trivalent linker. In other embodiments of the invention, the GalNAc or GalNAc derivative is attached to an iRNA agent of the invention via a tetravalent linker.


In certain embodiments, the double stranded RNAi agents of the invention comprise one GalNAc or GalNAc derivative attached to the iRNA agent. In certain embodiments, the double stranded RNAi agents of the invention comprise a plurality (e.g., 2, 3, 4, 5, or 6) GalNAc or GalNAc derivatives, each independently attached to a plurality of nucleotides of the double stranded RNAi agent through a plurality of monovalent linkers.


In some embodiments, for example, when the two strands of an iRNA agent of the invention are part of one larger molecule connected by an uninterrupted chain of nucleotides between the 3′-end of one strand and the 5′-end of the respective other strand forming a hairpin loop comprising, a plurality of unpaired nucleotides, each unpaired nucleotide within the hairpin loop may independently comprise a GalNAc or GalNAc derivative attached via a monovalent linker. The hairpin loop may also be formed by an extended overhang in one strand of the duplex.


In some embodiments, for example, when the two strands of an iRNA agent of the invention are part of one larger molecule connected by an uninterrupted chain of nucleotides between the 3′-end of one strand and the 5′-end of the respective other strand forming a hairpin loop comprising, a plurality of unpaired nucleotides, each unpaired nucleotide within the hairpin loop may independently comprise a GalNAc or GalNAc derivative attached via a monovalent linker. The hairpin loop may also be formed by an extended overhang in one strand of the duplex.


In one embodiment, a carbohydrate conjugate for use in the compositions and methods of the invention is selected from the group consisting of:




embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


wherein Y is O or S and n is 3-6 (Formula XXIV);




embedded image


wherein Y is O or S and n is 3-6 (Formula XXV);




embedded image


wherein X is O or S (Formula XXVII);




embedded image


embedded image


embedded image


In another embodiment, a carbohydrate conjugate for use in the compositions and methods of the invention is a monosaccharide. In one embodiment, the monosaccharide is an N-acetylgalactosamine, such as




embedded image


In some embodiments, the RNAi agent is attached to the carbohydrate conjugate via a linker as shown in the following schematic, wherein X is O or S




embedded image


In some embodiments, the RNAi agent is conjugated to L96 as defined in Table 1 and shown below:




embedded image


Another representative carbohydrate conjugate for use in the embodiments described herein includes, but is not limited to,




embedded image


when one of X or Y is an oligonucleotide, the other is a hydrogen.


In some embodiments, a suitable ligand is a ligand disclosed in WO 2019/055633, the entire contents of which are incorporated herein by reference. In one embodiment the ligand comprises the structure below:




embedded image


In certain embodiments of the invention, the GalNAc or GalNAc derivative is attached to an iRNA agent of the invention via a monovalent linker. In some embodiments, the GalNAc or GalNAc derivative is attached to an iRNA agent of the invention via a bivalent linker. In yet other embodiments of the invention, the GalNAc or GalNAc derivative is attached to an iRNA agent of the invention via a trivalent linker.


In one embodiment, the double stranded RNAi agents of the invention comprise one or more GalNAc or GalNAc derivative attached to the iRNA agent. The GalNAc may be attached to any nucleotide via a linker on the sense strand or antsisense strand. The GalNac may be attached to the 5′-end of the sense strand, the 3′ end of the sense strand, the 5′-end of the antisense strand, or the 3′-end of the antisense strand. In one embodiment, the GalNAc is attached to the 3′ end of the sense strand, e.g., via a trivalent linker.


In other embodiments, the double stranded RNAi agents of the invention comprise a plurality (e.g., 2, 3, 4, 5, or 6) GalNAc or GalNAc derivatives, each independently attached to a plurality of nucleotides of the double stranded RNAi agent through a plurality of linkers, e.g., monovalent linkers.


In some embodiments, for example, when the two strands of an iRNA agent of the invention is part of one larger molecule connected by an uninterrupted chain of nucleotides between the 3′-end of one strand and the 5′-end of the respective other strand forming a hairpin loop comprising, a plurality of unpaired nucleotides, each unpaired nucleotide within the hairpin loop may independently comprise a GalNAc or GalNAc derivative attached via a monovalent linker.


In some embodiments, the carbohydrate conjugate further comprises one or more additional ligands as described above, such as, but not limited to, a PK modulator or a cell permeation peptide.


Additional carbohydrate conjugates and linkers suitable for use in the present invention include those described in PCT Publication Nos. WO 2014/179620 and WO 2014/179627, the entire contents of each of which are incorporated herein by reference.


D. Linkers


In some embodiments, the conjugate or ligand described herein can be attached to an iRNA oligonucleotide with various linkers that can be cleavable or non-cleavable.


The term “linker” or “linking group” means an organic moiety that connects two parts of a compound, e.g., covalently attaches two parts of a compound. Linkers typically comprise a direct bond or an atom such as oxygen or sulfur, a unit such as NR8, C(O), C(O)NH, SO, SO2, SO2NH or a chain of atoms, such as, but not limited to, substituted or unsubstituted alkyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl, arylalkyl, arylalkenyl, arylalkynyl, heteroarylalkyl, heteroarylalkenyl, heteroarylalkynyl, heterocyclylalkyl, heterocyclylalkenyl, heterocyclylalkynyl, aryl, heteroaryl, heterocyclyl, cycloalkyl, cycloalkenyl, alkylarylalkyl, alkylarylalkenyl, alkylarylalkynyl, alkenylarylalkyl, alkenylarylalkenyl, alkenylarylalkynyl, alkynylarylalkyl, alkynylarylalkenyl, alkynylarylalkynyl, alkylheteroarylalkyl, alkylheteroarylalkenyl, alkylheteroarylalkynyl, alkenylheteroarylalkyl, alkenylheteroarylalkenyl, alkenylheteroarylalkynyl, alkynylheteroarylalkyl, alkynylheteroarylalkenyl, alkynylheteroarylalkynyl, alkylheterocyclylalkyl, alkylheterocyclylalkenyl, alkylhererocyclylalkynyl, alkenylheterocyclylalkyl, alkenylheterocyclylalkenyl, alkenylheterocyclylalkynyl, alkynylheterocyclylalkyl, alkynylheterocyclylalkenyl, alkynylheterocyclylalkynyl, alkylaryl, alkenylaryl, alkynylaryl, alkylheteroaryl, alkenylheteroaryl, alkynylhereroaryl, which one or more methylenes can be interrupted or terminated by O, S, S(O), SO2, N(R8), C(O), substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl, or substituted or unsubstituted heterocyclic; where R8 is hydrogen, acyl, aliphatic, or substituted aliphatic. In one embodiment, the linker is about 1-24 atoms, 2-24, 3-24, 4-24, 5-24, 6-24, 6-18, 7-18, 8-18, 7-17, 8-17, 6-16, 7-17, or 8-16 atoms.


A cleavable linking group is one which is sufficiently stable outside the cell, but which upon entry into a target cell is cleaved to release the two parts the linker is holding together. In one embodiment, the cleavable linking group is cleaved at least about 10 times, 20, times, 30 times, 40 times, 50 times, 60 times, 70 times, 80 times, 90 times, or more, or at least 100 times faster in a target cell or under a first reference condition (which can, e.g., be selected to mimic or represent intracellular conditions) than in the blood of a subject, or under a second reference condition (which can, e.g., be selected to mimic or represent conditions found in the blood or serum).


Cleavable linking groups are susceptible to cleavage agents, e.g., pH, redox potential, or the presence of degradative molecules. Generally, cleavage agents are more prevalent or found at higher levels or activities inside cells than in serum or blood. Examples of such degradative agents include: redox agents which are selected for particular substrates or which have no substrate specificity, including, e.g., oxidative or reductive enzymes or reductive agents such as mercaptans, present in cells, that can degrade a redox cleavable linking group by reduction; esterases; endosomes or agents that can create an acidic environment, e.g., those that result in a pH of five or lower; enzymes that can hydrolyze or degrade an acid cleavable linking group by acting as a general acid, peptidases (which can be substrate specific), and phosphatases.


A cleavable linkage group, such as a disulfide bond can be susceptible to pH. The pH of human serum is 7.4, while the average intracellular pH is slightly lower, ranging from about 7.1-7.3. Endosomes have a more acidic pH, in the range of 5.5-6.0, and lysosomes have an even more acidic pH at around 5.0. Some linkers will have a cleavable linking group that is cleaved at a selected pH, thereby releasing a cationic lipid from the ligand inside the cell, or into the desired compartment of the cell.


A linker can include a cleavable linking group that is cleavable by a particular enzyme. The type of cleavable linking group incorporated into a linker can depend on the cell to be targeted. For example, a liver-targeting ligand can be linked to a cationic lipid through a linker that includes an ester group. Liver cells are rich in esterases, and therefore the linker will be cleaved more efficiently in liver cells than in cell types that are not esterase-rich. Other cell-types rich in esterases include cells of the lung, renal cortex, and testis.


Linkers that contain peptide bonds can be used when targeting cell types rich in peptidases, such as liver cells and synoviocytes.


In general, the suitability of a candidate cleavable linking group can be evaluated by testing the ability of a degradative agent (or condition) to cleave the candidate linking group. It will also be desirable to also test the candidate cleavable linking group for the ability to resist cleavage in the blood or when in contact with other non-target tissue. Thus, one can determine the relative susceptibility to cleavage between a first and a second condition, where the first is selected to be indicative of cleavage in a target cell and the second is selected to be indicative of cleavage in other tissues or biological fluids, e.g., blood or serum. The evaluations can be carried out in cell free systems, in cells, in cell culture, in organ or tissue culture, or in whole animals. It can be useful to make initial evaluations in cell-free or culture conditions and to confirm by further evaluations in whole animals. In certain embodiments, useful candidate compounds are cleaved at least about 2, 4, 10, 20, 30, 40, 50, 60, 70, 80, 90, or 100 times faster in the cell (or under in vitro conditions selected to mimic intracellular conditions) as compared to blood or serum (or under in vitro conditions selected to mimic extracellular conditions).


i. Redox Cleavable Linking Groups


In certain embodiments, a cleavable linking group is a redox cleavable linking group that is cleaved upon reduction or oxidation. An example of reductively cleavable linking group is a disulphide linking group (—S—S—). To determine if a candidate cleavable linking group is a suitable “reductively cleavable linking group,” or for example is suitable for use with a particular iRNA moiety and particular targeting agent one can look to methods described herein. For example, a candidate can be evaluated by incubation with dithiothreitol (DTT), or other reducing agent using reagents know in the art, which mimic the rate of cleavage which would be observed in a cell, e.g., a target cell. The candidates can also be evaluated under conditions which are selected to mimic blood or serum conditions. In one, candidate compounds are cleaved by at most about 10% in the blood. In other embodiments, useful candidate compounds are degraded at least about 2, 4, 10, 20, 30, 40, 50, 60, 70, 80, 90, or about 100 times faster in the cell (or under in vitro conditions selected to mimic intracellular conditions) as compared to blood (or under in vitro conditions selected to mimic extracellular conditions). The rate of cleavage of candidate compounds can be determined using standard enzyme kinetics assays under conditions chosen to mimic intracellular media and compared to conditions chosen to mimic extracellular media.


ii. Phosphate-Based Cleavable Linking Groups


In other embodiments, a cleavable linker comprises a phosphate-based cleavable linking group. A phosphate-based cleavable linking group is cleaved by agents that degrade or hydrolyze the phosphate group. An example of an agent that cleaves phosphate groups in cells are enzymes such as phosphatases in cells. Examples of phosphate-based linking groups are —O—P(O)(ORk)-O—, —O—P(S)(ORk)-O—, —O—P(S)(SRk)-O—, —S—P(O)(ORk)-O—, —O—P(O)(ORk)-S—, —S—P(O)(ORk)-S—, —O—P(S)(ORk)-S—, —S—P(S)(ORk)-O—, —O—P(O)(Rk)-O—, —O—P(S)(Rk)-O—, —S—P(O)(Rk)-O—, —S—P(S)(Rk)-O—, —S—P(O)(Rk)-S—, —O—P(S)(Rk)-S—, wherein Rk at each occurrence can be, independently, C1-C20 alkyl, C1-C20 haloalkyl, C6-C10 aryl, or C7-C12 aralkyl. Exemplary embodiments include —O—P(O)(OH)—O—, —O—P(S)(OH)—O—, —O—P(S)(SH)—O—, —S—P(O)(OH)—O—, —O—P(O)(OH)—S—, —S—P(O)(OH)—S—, —O—P(S)(OH)—S—, —S—P(S)(OH)—O—, —O—P(O)(H)—O—, —O—P(S)(H)—O—, —S—P(O)(H)—O, —S—P(S)(H)—O—, —S—P(O)(H)—S—, and —O—P(S)(H)—S—. In certain embodiments, a phosphate-based linking group is —O—P(O)(OH)—O—. These candidates can be evaluated using methods analogous to those described above.


iii. Acid Cleavable Linking Groups


In other embodiments, a cleavable linker comprises an acid cleavable linking group. An acid cleavable linking group is a linking group that is cleaved under acidic conditions. In certain embodiments acid cleavable linking groups are cleaved in an acidic environment with a pH of about 6.5 or lower (e.g., about 6.0, 5.5, 5.0, or lower), or by agents such as enzymes that can act as a general acid. In a cell, specific low pH organelles, such as endosomes and lysosomes can provide a cleaving environment for acid cleavable linking groups. Examples of acid cleavable linking groups include but are not limited to hydrazones, esters, and esters of amino acids. Acid cleavable groups can have the general formula —C═NN—, C(O)O, or —OC(O). An exemplary embodiment is when the carbon attached to the oxygen of the ester (the alkoxy group) is an aryl group, substituted alkyl group, or tertiary alkyl group such as dimethyl pentyl or t-butyl. These candidates can be evaluated using methods analogous to those described above.


iv. Ester-Based Linking Groups


In other embodiments, a cleavable linker comprises an ester-based cleavable linking group. An ester-based cleavable linking group is cleaved by enzymes such as esterases and amidases in cells. Examples of ester-based cleavable linking groups include, but are not limited to, esters of alkylene, alkenylene and alkynylene groups. Ester cleavable linking groups have the general formula —C(O)O—, or —OC(O)—. These candidates can be evaluated using methods analogous to those described above.


v. Peptide-Based Cleaving Groups


In yet other embodiments, a cleavable linker comprises a peptide-based cleavable linking group. A peptide-based cleavable linking group is cleaved by enzymes such as peptidases and proteases in cells. Peptide-based cleavable linking groups are peptide bonds formed between amino acids to yield oligopeptides (e.g., dipeptides, tripeptides etc.) and polypeptides. Peptide-based cleavable groups do not include the amide group (—C(O)NH—). The amide group can be formed between any alkylene, alkenylene or alkynelene. A peptide bond is a special type of amide bond formed between amino acids to yield peptides and proteins. The peptide based cleavage group is generally limited to the peptide bond (i.e., the amide bond) formed between amino acids yielding peptides and proteins and does not include the entire amide functional group. Peptide-based cleavable linking groups have the general formula —NHCHRAC(O)NHCHRBC(O)—, where RA and RB are the R groups of the two adjacent amino acids. These candidates can be evaluated using methods analogous to those described above.


In some embodiments, an iRNA of the invention is conjugated to a carbohydrate through a linker. Non-limiting examples of iRNA carbohydrate conjugates with linkers of the compositions and methods of the invention include, but are not limited to,




embedded image


embedded image


embedded image


when one of X or Y is an oligonucleotide, the other is a hydrogen.


In certain embodiments of the compositions and methods of the invention, a ligand is one or more “GalNAc” (N-acetylgalactosamine) derivatives attached through a bivalent or trivalent branched linker.


In one embodiment, a dsRNA of the invention is conjugated to a bivalent or trivalent branched linker selected from the group of structures shown in any of formula (XLV)-(XLVI):




embedded image


wherein:

    • q2A, q2B, q3A, q3B, q4A, q4B, q5A, q5B and q5C represent independently for each occurrence 0-20 and wherein the repeating unit can be the same or different;
    • P2A, P2B, P3A, P3B, P4A, P4B, P5A, P5B, P5C, T2A, T2B, T3A, T3B, T4A, T4B, T4A, T5B, T5C are each independently for each occurrence absent, CO, NH, O, S, OC(O), NHC(O), CH2, CH2NH or CH2O;
    • Q2A, Q2B, Q3A, Q3B, Q4A, Q4B, Q5A, Q5B, Q5C are independently for each occurrence absent, alkylene, substituted alkylene wherein one or more methylenes can be interrupted or terminated by one or more of O, S, S(O), SO2, N(RN), C(R′)═C(R″), C≡C or C(O);
    • R2A, R2B, R3A, R3B, R4A, R4B, R5A, R5B, R5C are each independently for each occurrence absent, NH, O, S, CH2, C(O)O, C(O)NH, NHCH(Ra)C(O), —C(O)—CH(Ra)—NH—, CO, CH═N—O,




embedded image


or heterocyclyl;

    • L2A, L2B, L3A, L3B, L4A, L4B, L5A, L5B and L5C represent the ligand; i.e. each independently for each occurrence a monosaccharide (such as GalNAc), disaccharide, trisaccharide, tetrasaccharide, oligosaccharide, or polysaccharide; and Ra is H or amino acid side chain. Trivalent conjugating GalNAc derivatives are particularly useful for use with RNAi agents for inhibiting the expression of a target gene, such as those of formula (XLIX):




embedded image


wherein L5A, L5B and L5C represent a monosaccharide, such as GalNAc derivative.


Examples of suitable bivalent and trivalent branched linker groups conjugating GalNAc derivatives include, but are not limited to, the structures recited above as formulas II, VII, XI, X, and XIII.


Representative U.S. patents that teach the preparation of RNA conjugates include, but are not limited to, U.S. Pat. Nos. 4,828,979; 4,948,882; 5,218,105; 5,525,465; 5,541,313; 5,545,730; 5,552,538; 5,578,717, 5,580,731; 5,591,584; 5,109,124; 5,118,802; 5,138,045; 5,414,077; 5,486,603; 5,512,439; 5,578,718; 5,608,046; 4,587,044; 4,605,735; 4,667,025; 4,762,779; 4,789,737; 4,824,941; 4,835,263; 4,876,335; 4,904,582; 4,958,013; 5,082,830; 5,112,963; 5,214,136; 5,082,830; 5,112,963; 5,214,136; 5,245,022; 5,254,469; 5,258,506; 5,262,536; 5,272,250; 5,292,873; 5,317,098; 5,371,241, 5,391,723; 5,416,203, 5,451,463; 5,510,475; 5,512,667; 5,514,785; 5,565,552; 5,567,810; 5,574,142; 5,585,481; 5,587,371; 5,595,726; 5,597,696; 5,599,923; 5,599,928; 5,688,941; 6,294,664; 6,320,017; 6,576,752; 6,783,931; 6,900,297; 7,037,646; and 8,106,022, the entire contents of each of which are hereby incorporated herein by reference.


It is not necessary for all positions in a given compound to be uniformly modified, and in fact more than one of the aforementioned modifications can be incorporated in a single compound or even at a single nucleoside within an iRNA. The present invention also includes iRNA compounds that are chimeric compounds.


“Chimeric” iRNA compounds or “chimeras,” in the context of this invention, are iRNA compounds, such as, dsRNAi agents, that contain two or more chemically distinct regions, each made up of at least one monomer unit, i.e., a nucleotide in the case of a dsRNA compound. These iRNAs typically contain at least one region wherein the RNA is modified so as to confer upon the iRNA increased resistance to nuclease degradation, increased cellular uptake, or increased binding affinity for the target nucleic acid. An additional region of the iRNA can serve as a substrate for enzymes capable of cleaving RNA:DNA or RNA:RNA hybrids. By way of example, RNase H is a cellular endonuclease which cleaves the RNA strand of an RNA:DNA duplex. Activation of RNase H, therefore, results in cleavage of the RNA target, thereby greatly enhancing the efficiency of iRNA inhibition of gene expression. Consequently, comparable results can often be obtained with shorter iRNAs when chimeric dsRNAs are used, compared to phosphorothioate deoxy dsRNAs hybridizing to the same target region. Cleavage of the RNA target can be routinely detected by gel electrophoresis and, if necessary, associated nucleic acid hybridization techniques known in the art.


In certain instances, the RNA of an iRNA can be modified by a non-ligand group. A number of non-ligand molecules have been conjugated to iRNAs in order to enhance the activity, cellular distribution or cellular uptake of the iRNA, and procedures for performing such conjugations are available in the scientific literature. Such non-ligand moieties have included lipid moieties, such as cholesterol (Kubo, T. et al., Biochem. Biophys. Res. Comm., 2007, 365(1):54-61; Letsinger et al., Proc. Natl. Acad. Sci. USA, 1989, 86:6553), cholic acid (Manoharan et al., Bioorg. Med. Chem. Lett., 1994, 4:1053), a thioether, e.g., hexyl-S-tritylthiol (Manoharan et al., Ann. N.Y. Acad. Sci., 1992, 660:306; Manoharan et al., Bioorg. Med. Chem. Let., 1993, 3:2765), a thiocholesterol (Oberhauser et al., Nucl. Acids Res., 1992, 20:533), an aliphatic chain, e.g., dodecandiol or undecyl residues (Saison-Behmoaras et al., EMBO J., 1991, 10:111; Kabanov et al., FEBS Lett., 1990, 259:327; Svinarchuk et al., Biochimie, 1993, 75:49), a phospholipid, e.g., di-hexadecyl-rac-glycerol or triethylammonium 1,2-di-O-hexadecyl-rac-glycero-3-H-phosphonate (Manoharan et al., Tetrahedron Lett., 1995, 36:3651; Shea et al., Nucl. Acids Res., 1990, 18:3777), a polyamine or a polyethylene glycol chain (Manoharan et al., Nucleosides & Nucleotides, 1995, 14:969), or adamantane acetic acid (Manoharan et al., Tetrahedron Lett., 1995, 36:3651), a palmityl moiety (Mishra et al., Biochim. Biophys. Acta, 1995, 1264:229), or an octadecylamine or hexylamino-carbonyl-oxycholesterol moiety (Crooke et al., J. Pharmacol. Exp. Ther., 1996, 277:923). Representative United States patents that teach the preparation of such RNA conjugates have been listed above. Typical conjugation protocols involve the synthesis of RNAs bearing an aminolinker at one or more positions of the sequence. The amino group is then reacted with the molecule being conjugated using appropriate coupling or activating reagents. The conjugation reaction can be performed either with the RNA still bound to the solid support or following cleavage of the RNA, in solution phase. Purification of the RNA conjugate by HPLC typically affords the pure conjugate.


IV. Delivery of an iRNA of the Invention

The delivery of an iRNA of the invention to a cell e.g., a cell within a subject, such as a human subject (e.g., a subject in need thereof, such as a subject susceptible to or diagnosed with a complement factor B-associated disorder) can be achieved in a number of different ways. For example, delivery may be performed by contacting a cell with an iRNA of the invention either in vitro or in vivo. In vivo delivery may also be performed directly by administering a composition comprising an iRNA, e.g., a dsRNA, to a subject. Alternatively, in vivo delivery may be performed indirectly by administering one or more vectors that encode and direct the expression of the iRNA. These alternatives are discussed further below.


In general, any method of delivering a nucleic acid molecule (in vitro or in vivo) can be adapted for use with an iRNA of the invention (see e.g., Akhtar S. and Julian R L. (1992) Trends Cell. Biol. 2(5):139-144 and WO94/02595, which are incorporated herein by reference in their entireties). For in vivo delivery, factors to consider in order to deliver an iRNA molecule include, for example, biological stability of the delivered molecule, prevention of non-specific effects, and accumulation of the delivered molecule in the target tissue. RNA interference has also shown success with local delivery to the CNS by direct injection (Dorn, G., et al. (2004) Nucleic Acids 32:e49; Tan, P H., et al (2005) Gene Ther. 12:59-66; Makimura, H., et al (2002) BMC Neurosci. 3:18; Shishkina, G T., et al (2004) Neuroscience 129:521-528; Thakker, E R., et al (2004) Proc. Natl. Acad. Sci. U.S.A. 101:17270-17275; Akaneya, Y., et al (2005) J. Neurophysiol. 93:594-602). Modification of the RNA or the pharmaceutical carrier can also permit targeting of the iRNA to the target tissue and avoid undesirable off-target effects. iRNA molecules can be modified by chemical conjugation to lipophilic groups such as cholesterol to enhance cellular uptake and prevent degradation. For example, an iRNA directed against ApoB conjugated to a lipophilic cholesterol moiety was injected systemically into mice and resulted in knockdown of apoB mRNA in both the liver and jejunum (Soutschek, J., et al (2004) Nature 432:173-178).


In an alternative embodiment, the iRNA can be delivered using drug delivery systems such as a nanoparticle, a dendrimer, a polymer, liposomes, or a cationic delivery system. Positively charged cationic delivery systems facilitate binding of an iRNA molecule (negatively charged) and also enhance interactions at the negatively charged cell membrane to permit efficient uptake of an iRNA by the cell. Cationic lipids, dendrimers, or polymers can either be bound to an iRNA, or induced to form a vesicle or micelle (see e.g., Kim S H, et al (2008) Journal of Controlled Release 129(2):107-116) that encases an iRNA. The formation of vesicles or micelles further prevents degradation of the iRNA when administered systemically. Methods for making and administering cationic-iRNA complexes are well within the abilities of one skilled in the art (see e.g., Sorensen, D R, et al (2003) J. Mol. Biol 327:761-766; Verma, U N, et al (2003) Clin. Cancer Res. 9:1291-1300; Arnold, A S et al (2007) J. Hypertens. 25:197-205, which are incorporated herein by reference in their entirety). Some non-limiting examples of drug delivery systems useful for systemic delivery of iRNAs include DOTAP (Sorensen, D R., et al (2003), supra; Verma, U N, et al (2003), supra), “solid nucleic acid lipid particles” (Zimmermann, T S, et al (2006) Nature 441:111-114), cardiolipin (Chien, P Y, et al (2005) Cancer Gene Ther. 12:321-328; Pal, A, et al (2005) Int J. Oncol. 26:1087-1091), polyethyleneimine (Bonnet M E, et al (2008) Pharm. Res. August 16 Epub ahead of print; Aigner, A. (2006) J. Biomed. Biotechnol. 71659), Arg-Gly-Asp (RGD) peptides (Liu, S. (2006) Mol. Pharm. 3:472-487), and polyamidoamines (Tomalia, D A, et al (2007) Biochem. Soc. Trans. 35:61-67; Yoo, H., et al (1999) Pharm. Res. 16:1799-1804). In some embodiments, an iRNA forms a complex with cyclodextrin for systemic administration. Methods for administration and pharmaceutical compositions of iRNAs and cyclodextrins can be found in U.S. Pat. No. 7,427,605, which is herein incorporated by reference in its entirety.


A. Vector encoded iRNAs of the Invention


iRNA targeting the complement factor B gene can be expressed from transcription units inserted into DNA or RNA vectors (see, e.g., Couture, A, et al., TIG. (1996), 12:5-10; Skillern, A, et al., International PCT Publication No. WO 00/22113, Conrad, International PCT Publication No. WO 00/22114, and Conrad, U.S. Pat. No. 6,054,299). Expression can be transient (on the order of hours to weeks) or sustained (weeks to months or longer), depending upon the specific construct used and the target tissue or cell type. These transgenes can be introduced as a linear construct, a circular plasmid, or a viral vector, which can be an integrating or non-integrating vector. The transgene can also be constructed to permit it to be inherited as an extrachromosomal plasmid (Gassmann, et al., Proc. Natl. Acad. Sci. USA (1995) 92:1292).


Viral vector systems which can be utilized with the methods and compositions described herein include, but are not limited to, (a) adenovirus vectors; (b) retrovirus vectors, including but not limited to lentiviral vectors, moloney murine leukemia virus, etc.; (c) adeno-associated virus vectors; (d) herpes simplex virus vectors; (e) SV 40 vectors; (f) polyoma virus vectors; (g) papilloma virus vectors; (h) picornavirus vectors; (i) pox virus vectors such as an orthopox, e.g., vaccinia virus vectors or avipox, e.g. canary pox or fowl pox; and (j) a helper-dependent or gutless adenovirus. Replication-defective viruses can also be advantageous. Different vectors will or will not become incorporated into the cells' genome. The constructs can include viral sequences for transfection, if desired. Alternatively, the construct can be incorporated into vectors capable of episomal replication, e.g. EPV and EBV vectors. Constructs for the recombinant expression of an iRNA will generally require regulatory elements, e.g., promoters, enhancers, etc., to ensure the expression of the iRNA in target cells. Other aspects to consider for vectors and constructs are known in the art.


V. Pharmaceutical Compositions of the Invention

The present invention also includes pharmaceutical compositions and formulations which include the iRNAs of the invention. In one embodiment, provided herein are pharmaceutical compositions containing an iRNA, as described herein, and a pharmaceutically acceptable carrier. The pharmaceutical compositions containing the iRNA are useful for preventing or treating a complement factor B-associated disorder. Such pharmaceutical compositions are formulated based on the mode of delivery. One example is compositions that are formulated for systemic administration via parenteral delivery, e.g., by subcutaneous (SC), intramuscular (IM), or intravenous (IV) delivery. The pharmaceutical compositions of the invention may be administered in dosages sufficient to inhibit expression of a complement factor B gene.


In some embodiments, the pharmaceutical compositions of the invention are sterile. In another embodiment, the pharmaceutical compositions of the invention are pyrogen free.


The pharmaceutical compositions of the invention may be administered in dosages sufficient to inhibit expression of a complement factor B gene. In general, a suitable dose of an iRNA of the invention will be in the range of about 0.001 to about 200.0 milligrams per kilogram body weight of the recipient per day, generally in the range of about 1 to 50 mg per kilogram body weight per day. Typically, a suitable dose of an iRNA of the invention will be in the range of about 0.1 mg/kg to about 5.0 mg/kg, such as, about 0.3 mg/kg and about 3.0 mg/kg. A repeat-dose regimen may include administration of a therapeutic amount of iRNA on a regular basis, such as every month, once every 3-6 months, or once a year. In certain embodiments, the iRNA is administered about once per month to about once per six months.


After an initial treatment regimen, the treatments can be administered on a less frequent basis. Duration of treatment can be determined based on the severity of disease.


In other embodiments, a single dose of the pharmaceutical compositions can be long lasting, such that doses are administered at not more than 1, 2, 3, or 4 month intervals. In some embodiments of the invention, a single dose of the pharmaceutical compositions of the invention is administered about once per month. In other embodiments of the invention, a single dose of the pharmaceutical compositions of the invention is administered quarterly (i.e., about every three months). In other embodiments of the invention, a single dose of the pharmaceutical compositions of the invention is administered twice per year (i.e., about once every six months).


The skilled artisan will appreciate that certain factors can influence the dosage and timing required to effectively treat a subject, including but not limited to mutations present in the subject, previous treatments, the general health or age of the subject, and other diseases present. Moreover, treatment of a subject with a prophylactically or therapeutically effective amount, as appropriate, of a composition can include a single treatment or a series of treatments.


The iRNA can be delivered in a manner to target a particular tissue (e.g., hepatocytes).


Pharmaceutical compositions of the present invention include, but are not limited to, solutions, emulsions, and liposome-containing formulations. These compositions can be generated from a variety of components that include, but are not limited to, preformed liquids, self-emulsifying solids, and self-emulsifying semisolids. Formulations include those that target the liver.


The pharmaceutical formulations of the present invention, which can conveniently be presented in unit dosage form, can be prepared according to conventional techniques well known in the pharmaceutical industry. Such techniques include the step of bringing into association the active ingredients with the pharmaceutical carrier(s) or excipient(s). In general, the formulations are prepared by uniformly and intimately bringing into association the active ingredients with liquid carriers.


A. Additional Formulations


i. Emulsions


The compositions of the present invention can be prepared and formulated as emulsions. Emulsions are typically heterogeneous systems of one liquid dispersed in another in the form of droplets usually exceeding 0.1 μm in diameter (see e.g., Ansel's Pharmaceutical Dosage Forms and Drug Delivery Systems, Allen, L V., Popovich N G., and Ansel H C., 2004, Lippincott Williams & Wilkins (8th ed.), New York, NY; Idson, in Pharmaceutical Dosage Forms, Lieberman, Rieger and Banker (Eds.), 1988, Marcel Dekker, Inc., New York, N.Y., volume 1, p. 199; Rosoff, in Pharmaceutical Dosage Forms, Lieberman, Rieger and Banker (Eds.), 1988, Marcel Dekker, Inc., New York, N.Y., Volume 1, p. 245; Block in Pharmaceutical Dosage Forms, Lieberman, Rieger and Banker (Eds.), 1988, Marcel Dekker, Inc., New York, N.Y., volume 2, p. 335; Higuchi et al., in Remington's Pharmaceutical Sciences, Mack Publishing Co., Easton, Pa., 1985, p. 301). Emulsions are often biphasic systems comprising two immiscible liquid phases intimately mixed and dispersed with each other. In general, emulsions can be of either the water-in-oil (w/o) or the oil-in-water (o/w) variety. When an aqueous phase is finely divided into and dispersed as minute droplets into a bulk oily phase, the resulting composition is called a water-in-oil (w/o) emulsion. Alternatively, when an oily phase is finely divided into and dispersed as minute droplets into a bulk aqueous phase, the resulting composition is called an oil-in-water (o/w) emulsion. Emulsions can contain additional components in addition to the dispersed phases, and the active drug which can be present as a solution either in the aqueous phase, oily phase or itself as a separate phase. Pharmaceutical excipients such as emulsifiers, stabilizers, dyes, and anti-oxidants can also be present in emulsions as needed. Pharmaceutical emulsions can also be multiple emulsions that are comprised of more than two phases such as, for example, in the case of oil-in-water-in-oil (o/w/o) and water-in-oil-in-water (w/o/w) emulsions. Such complex formulations often provide certain advantages that simple binary emulsions do not. Multiple emulsions in which individual oil droplets of an o/w emulsion enclose small water droplets constitute a w/o/w emulsion. Likewise a system of oil droplets enclosed in globules of water stabilized in an oily continuous phase provides an o/w/o emulsion.


Emulsions are characterized by little or no thermodynamic stability. Often, the dispersed or discontinuous phase of the emulsion is well dispersed into the external or continuous phase and maintained in this form through the means of emulsifiers or the viscosity of the formulation. Other means of stabilizing emulsions entail the use of emulsifiers that can be incorporated into either phase of the emulsion. Emulsifiers can broadly be classified into four categories: synthetic surfactants, naturally occurring emulsifiers, absorption bases, and finely dispersed solids (see e.g., Ansel's Pharmaceutical Dosage Forms and Drug Delivery Systems, Allen, LV., Popovich N G., and Ansel H C., 2004, Lippincott Williams & Wilkins (8th ed.), New York, NY; Idson, in Pharmaceutical Dosage Forms, Lieberman, Rieger and Banker (Eds.), 1988, Marcel Dekker, Inc., New York, N.Y., volume 1, p. 199).


Synthetic surfactants, also known as surface active agents, have found wide applicability in the formulation of emulsions and have been reviewed in the literature (see e.g., Ansel's Pharmaceutical Dosage Forms and Drug Delivery Systems, Allen, LV., Popovich N G., and Ansel H C., 2004, Lippincott Williams & Wilkins (8th ed.), New York, NY; Rieger, in Pharmaceutical Dosage Forms, Lieberman, Rieger and Banker (Eds.), 1988, Marcel Dekker, Inc., New York, N.Y., volume 1, p. 285; Idson, in Pharmaceutical Dosage Forms, Lieberman, Rieger and Banker (Eds.), Marcel Dekker, Inc., New York, N.Y., 1988, volume 1, p. 199). Surfactants are typically amphiphilic and comprise a hydrophilic and a hydrophobic portion. The ratio of the hydrophilic to the hydrophobic nature of the surfactant has been termed the hydrophile/lipophile balance (HLB) and is a valuable tool in categorizing and selecting surfactants in the preparation of formulations. Surfactants can be classified into different classes based on the nature of the hydrophilic group: nonionic, anionic, cationic, and amphoteric (see e.g., Ansel's Pharmaceutical Dosage Forms and Drug Delivery Systems, Allen, LV., Popovich N G., and Ansel H C., 2004, Lippincott Williams & Wilkins (8th ed.), New York, NY Rieger, in Pharmaceutical Dosage Forms, Lieberman, Rieger and Banker (Eds.), 1988, Marcel Dekker, Inc., New York, N.Y., volume 1, p. 285).


A large variety of non-emulsifying materials are also included in emulsion formulations and contribute to the properties of emulsions. These include fats, oils, waxes, fatty acids, fatty alcohols, fatty esters, humectants, hydrophilic colloids, preservatives, and antioxidants (Block, in Pharmaceutical Dosage Forms, Lieberman, Rieger and Banker (Eds.), 1988, Marcel Dekker, Inc., New York, N.Y., volume 1, p. 335; Idson, in Pharmaceutical Dosage Forms, Lieberman, Rieger and Banker (Eds.), 1988, Marcel Dekker, Inc., New York, N.Y., volume 1, p. 199).


The application of emulsion formulations via dermatological, oral, and parenteral routes, and methods for their manufacture have been reviewed in the literature (see e.g., Ansel's Pharmaceutical Dosage Forms and Drug Delivery Systems, Allen, LV., Popovich N G., and Ansel H C., 2004, Lippincott Williams & Wilkins (8th ed.), New York, NY; Idson, in Pharmaceutical Dosage Forms, Lieberman, Rieger and Banker (Eds.), 1988, Marcel Dekker, Inc., New York, N.Y., volume 1, p. 199).


ii. Microemulsions


In one embodiment of the present invention, the compositions of iRNAs and nucleic acids are formulated as microemulsions. A microemulsion can be defined as a system of water, oil, and amphiphile which is a single optically isotropic and thermodynamically stable liquid solution (see e.g., Ansel's Pharmaceutical Dosage Forms and Drug Delivery Systems, Allen, LV., Popovich N G., and Ansel H C., 2004, Lippincott Williams & Wilkins (8th ed.), New York, NY; Rosoff, in Pharmaceutical Dosage Forms, Lieberman, Rieger and Banker (Eds.), 1988, Marcel Dekker, Inc., New York, N.Y., volume 1, p. 245). Typically microemulsions are systems that are prepared by first dispersing an oil in an aqueous surfactant solution and then adding a sufficient amount of a fourth component, generally an intermediate chain-length alcohol to form a transparent system. Therefore, microemulsions have also been described as thermodynamically stable, isotropically clear dispersions of two immiscible liquids that are stabilized by interfacial films of surface-active molecules (Leung and Shah, in: Controlled Release of Drugs: Polymers and Aggregate Systems, Rosoff, M., Ed., 1989, VCH Publishers, New York, pages 185-215).


iii. Microparticles


An iRNA of the invention may be incorporated into a particle, e.g., a microparticle. Microparticles can be produced by spray-drying, but may also be produced by other methods including lyophilization, evaporation, fluid bed drying, vacuum drying, or a combination of these techniques.


iv. Penetration Enhancers


In one embodiment, the present invention employs various penetration enhancers to effect the efficient delivery of nucleic acids, particularly iRNAs, to the skin of animals. Most drugs are present in solution in both ionized and nonionized forms. However, usually only lipid soluble or lipophilic drugs readily cross cell membranes. It has been discovered that even non-lipophilic drugs can cross cell membranes if the membrane to be crossed is treated with a penetration enhancer. In addition to aiding the diffusion of non-lipophilic drugs across cell membranes, penetration enhancers also enhance the permeability of lipophilic drugs.


Penetration enhancers can be classified as belonging to one of five broad categories, i.e., surfactants, fatty acids, bile salts, chelating agents, and non-chelating non-surfactants (see e.g., Malmsten, M. Surfactants and polymers in drug delivery, Informa Health Care, New York, NY, 2002; Lee et al., Critical Reviews in Therapeutic Drug Carrier Systems, 1991, p. 92). Each of the above mentioned classes of penetration enhancers and their use in manufacture of pharmaceutical compositions and delivery of pharmaceutical agents are well known in the art.


v. Excipients


In contrast to a carrier compound, a “pharmaceutical carrier” or “excipient” is a pharmaceutically acceptable solvent, suspending agent, or any other pharmacologically inert vehicle for delivering one or more nucleic acids to an animal. The excipient can be liquid or solid and is selected, with the planned manner of administration in mind, so as to provide for the desired bulk, consistency, etc., when combined with a nucleic acid and the other components of a given pharmaceutical composition. Such agent are well known in the art.


vi. Other Components


The compositions of the present invention can additionally contain other adjunct components conventionally found in pharmaceutical compositions, at their art-established usage levels. Thus, for example, the compositions can contain additional, compatible, pharmaceutically-active materials such as, for example, antipruritics, astringents, local anesthetics or anti-inflammatory agents, or can contain additional materials useful in physically formulating various dosage forms of the compositions of the present invention, such as dyes, flavoring agents, preservatives, antioxidants, opacifiers, thickening agents and stabilizers. However, such materials, when added, should not unduly interfere with the biological activities of the components of the compositions of the present invention. The formulations can be sterilized and, if desired, mixed with auxiliary agents, e.g., lubricants, preservatives, stabilizers, wetting agents, emulsifiers, salts for influencing osmotic pressure, buffers, colorings, flavorings, or aromatic substances, and the like which do not deleteriously interact with the nucleic acid(s) of the formulation.


Aqueous suspensions can contain substances which increase the viscosity of the suspension including, for example, sodium carboxymethylcellulose, sorbitol, or dextran. The suspension can also contain stabilizers.


In some embodiments, pharmaceutical compositions featured in the invention include (a) one or more iRNA and (b) one or more agents which function by a non-iRNA mechanism and which are useful in treating a complement factor B-associated disorder.


Toxicity and prophylactic efficacy of such compounds can be determined by standard pharmaceutical procedures in cell cultures or experimental animals, e.g., for determining the LD50 (the dose lethal to 50% of the population) and the ED50 (the dose prophylactically effective in 50% of the population). The dose ratio between toxic and therapeutic effects is the therapeutic index and it can be expressed as the ratio LD50/ED50. Compounds that exhibit high therapeutic indices are preferred.


The data obtained from cell culture assays and animal studies can be used in formulating a range of dosage for use in humans. The dosage of compositions featured herein in the invention lies generally within a range of circulating concentrations that include the ED50, such as an ED80 or ED90, with little or no toxicity. The dosage can vary within this range depending upon the dosage form employed and the route of administration utilized. For any compound used in the methods featured in the invention, the prophylactically effective dose can be estimated initially from cell culture assays. A dose can be formulated in animal models to achieve a circulating plasma concentration range of the compound or, when appropriate, of the polypeptide product of a target sequence (e.g., achieving a decreased concentration of the polypeptide) that includes the IC50 (i.e., the concentration of the test compound which achieves a half-maximal inhibition of symptoms) or higher levels of inhibition as determined in cell culture. Such information can be used to more accurately determine useful doses in humans. Levels in plasma can be measured, for example, by high performance liquid chromatography.


In addition to their administration, as discussed above, the iRNAs featured in the invention can be administered in combination with other known agents used for the prevention or treatment of a complement factor B-associated disorder. In any event, the administering physician can adjust the amount and timing of iRNA administration on the basis of results observed using standard measures of efficacy known in the art or described herein.


VI. Methods for Inhibiting Complement Factor B Expression

The present invention also provides methods of inhibiting expression of a CFB gene in a cell. The methods include contacting a cell with an RNAi agent, e.g., double stranded RNA agent, in an amount effective to inhibit expression of CFB in the cell, thereby inhibiting expression of CFB in the cell.


Contacting of a cell with an iRNA, e.g., a double stranded RNA agent, may be done in vitro or in vivo. Contacting a cell in vivo with the iRNA includes contacting a cell or group of cells within a subject, e.g., a human subject, with the iRNA. Combinations of in vitro and in vivo methods of contacting a cell are also possible. Contacting a cell may be direct or indirect, as discussed above. Furthermore, contacting a cell may be accomplished via a targeting ligand, including any ligand described herein or known in the art. In certain embodiments, the targeting ligand is a carbohydrate moiety, e.g., a GalNAc ligand, or any other ligand that directs the RNAi agent to a site of interest.


The term “inhibiting,” as used herein, is used interchangeably with “reducing,” “silencing,” “downregulating”, “suppressing”, and other similar terms, and includes any level of inhibition.


The phrase “inhibiting expression of a complement factor B gene” is intended to refer to inhibition of expression of any complement factor B gene (such as, e.g., a mouse complement factor B gene, a rat complement factor B gene, a monkey complement factor B gene, or a human complement factor B gene) as well as variants or mutants of a complement factor B gene. Thus, the complement factor B gene may be a wild-type complement factor B gene, a mutant complement factor B gene, or a transgenic complement factor B gene in the context of a genetically manipulated cell, group of cells, or organism.


“Inhibiting expression of a complement factor B gene” includes any level of inhibition of a complement factor B gene, e.g., at least partial suppression of the expression of a complement factor B gene, such as a clinically relevant level of supression. The expression of the complement factor B gene may be assessed based on the level, or the change in the level, of any variable associated with complement factor B gene expression, e.g., complement factor B mRNA level or complement factor B protein level, or, for example, CH50 activity as a measure of total hemolytic complement, AH50 to measure the hemolytic activity of the alternate pathway of complement, or lactate dehydrogenase (LDH) levels as a measure of intravascular hemolysis, or hemoglobin levels. Levels of C3, C9, C5, C5a, C5b, and soluble C5b-9 complex may also be measured to assess CFB expression. Inhibition may be assessed by a decrease in an absolute or relative level of one or more of these variables compared with a control level. This level may be assessed in an individual cell or in a group of cells, including, for example, a sample derived from a subject. It is understood that complement factor B is expressed predominantly in the liver, and is present in circulation.


Inhibition may be assessed by a decrease in an absolute or relative level of one or more variables that are associated with complement factor B expression compared with a control level. The control level may be any type of control level that is utilized in the art, e.g., a pre-dose baseline level, or a level determined from a similar subject, cell, or sample that is untreated or treated with a control (such as, e.g., buffer only control or inactive agent control).


In some embodiments of the methods of the invention, expression of a complement factor B gene is inhibited by at least 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, or 95%, or to below the level of detection of the assay. In certain embodiments, expression of a complement factor B gene is inhibited by at least 70%. It is further understood that inhibition of complement factor B expression in certain tissues, e.g., in gall bladder, without a significant inhibition of expression in other tissues, e.g., brain, may be desirable. In certain embodiments, expression level is determined using the assay method provided in Example 2 with a 10 nM siRNA concentration in the appropriate species matched cell line.


In certain embodiments, inhibition of expression in vivo is determined by knockdown of the human gene in a rodent expressing the human gene, e.g., an AAV-infected mouse expressing the human target gene (i.e., complement factor B), e.g., when administered as a single dose, e.g., at 3 mg/kg at the nadir of RNA expression. Knockdown of expression of an endogenous gene in a model animal system can also be determined, e.g., after administration of a single dose at, e.g., 3 mg/kg at the nadir of RNA expression. Such systems are useful when the nucleic acid sequence of the human gene and the model animal gene are sufficiently close such that the human iRNA provides effective knockdown of the model animal gene. RNA expression in liver is determined using the PCR methods provided in Example 2.


Inhibition of the expression of a complement factor B gene may be manifested by a reduction of the amount of mRNA expressed by a first cell or group of cells (such cells may be present, for example, in a sample derived from a subject) in which a complement factor B gene is transcribed and which has or have been treated (e.g., by contacting the cell or cells with an iRNA of the invention, or by administering an iRNA of the invention to a subject in which the cells are or were present) such that the expression of a complement factor B gene is inhibited, as compared to a second cell or group of cells substantially identical to the first cell or group of cells but which has not or have not been so treated (control cell(s) not treated with an iRNA or not treated with an iRNA targeted to the gene of interest). In certain embodiments, the inhibition is assessed by the method provided in Example 2 using a 10 nM siRNA concentration in the species matched cell line and expressing the level of mRNA in treated cells as a percentage of the level of mRNA in control cells, using the following formula:









(

mRNA


in


control


cells

)

-

(

mRNA


in


treated


cells

)



(

mRNA


in


control


cells

)



•100

%




In other embodiments, inhibition of the expression of a complement factor B gene may be assessed in terms of a reduction of a parameter that is functionally linked to complement factor B gene expression, e.g., complement factor B protein level in blood or serum from a subject. Complement factor B gene silencing may be determined in any cell expressing complement factor B, either endogenous or heterologous from an expression construct, and by any assay known in the art.


Inhibition of the expression of a complement factor B protein may be manifested by a reduction in the level of the complement factor B protein that is expressed by a cell or group of cells or in a subject sample (e.g., the level of protein in a blood sample derived from a subject). As explained above, for the assessment of mRNA suppression, the inhibition of protein expression levels in a treated cell or group of cells may similarly be expressed as a percentage of the level of protein in a control cell or group of cells, or the change in the level of protein in a subject sample, e.g., blood or serum derived therefrom.


A control cell, a group of cells, or subject sample that may be used to assess the inhibition of the expression of a complement factor B gene includes a cell, group of cells, or subject sample that has not yet been contacted with an RNAi agent of the invention. For example, the control cell, group of cells, or subject sample may be derived from an individual subject (e.g., a human or animal subject) prior to treatment of the subject with an RNAi agent or an appropriately matched population control.


The level of complement factor B mRNA that is expressed by a cell or group of cells may be determined using any method known in the art for assessing mRNA expression. In one embodiment, the level of expression of complement factor B in a sample is determined by detecting a transcribed polynucleotide, or portion thereof, e.g., mRNA of the complement factor B gene. RNA may be extracted from cells using RNA extraction techniques including, for example, using acid phenol/guanidine isothiocyanate extraction (RNAzol B; Biogenesis), RNeasy™ RNA preparation kits (Qiagen®) or PAXgene™ (PreAnalytix™, Switzerland). Typical assay formats utilizing ribonucleic acid hybridization include nuclear run-on assays, RT-PCR, RNase protection assays, northern blotting, in situ hybridization, and microarray analysis.


In some embodiments, the level of expression of complement factor B is determined using a nucleic acid probe. The term “probe”, as used herein, refers to any molecule that is capable of selectively binding to a specific complement factor B. Probes can be synthesized by one of skill in the art, or derived from appropriate biological preparations. Probes may be specifically designed to be labeled. Examples of molecules that can be utilized as probes include, but are not limited to, RNA, DNA, proteins, antibodies, and organic molecules.


Isolated mRNA can be used in hybridization or amplification assays that include, but are not limited to, Southern or northern analyses, polymerase chain reaction (PCR) analyses and probe arrays. One method for the determination of mRNA levels involves contacting the isolated mRNA with a nucleic acid molecule (probe) that can hybridize to complement factor B mRNA. In one embodiment, the mRNA is immobilized on a solid surface and contacted with a probe, for example by running the isolated mRNA on an agarose gel and transferring the mRNA from the gel to a membrane, such as nitrocellulose. In an alternative embodiment, the probe(s) are immobilized on a solid surface and the mRNA is contacted with the probe(s), for example, in an Affymetrix® gene chip array. A skilled artisan can readily adapt known mRNA detection methods for use in determining the level of complement factor B mRNA.


An alternative method for determining the level of expression of complement factor B in a sample involves the process of nucleic acid amplification or reverse transcriptase (to prepare cDNA) of for example mRNA in the sample, e.g., by RT-PCR (the experimental embodiment set forth in Mullis, 1987, U.S. Pat. No. 4,683,202), ligase chain reaction (Barany (1991) Proc. Natl. Acad. Sci. USA 88:189-193), self sustained sequence replication (Guatelli et al. (1990) Proc. Natl. Acad. Sci. USA 87:1874-1878), transcriptional amplification system (Kwoh et al. (1989) Proc. Natl. Acad. Sci. USA 86:1173-1177), Q-Beta Replicase (Lizardi et al. (1988) Bio/Technology 6:1197), rolling circle replication (Lizardi et al., U.S. Pat. No. 5,854,033) or any other nucleic acid amplification method, followed by the detection of the amplified molecules using techniques well known to those of skill in the art. These detection schemes are especially useful for the detection of nucleic acid molecules if such molecules are present in very low numbers. In particular aspects of the invention, the level of expression of CFB is determined by quantitative fluorogenic RT-PCR (i.e., the TaqMan™ System). In certain embodiments, expression level is determined by the method provided in Example 2 using, e.g., a 10 nM siRNA concentration, in the species matched cell line.


The expression levels of complement factor B mRNA may be monitored using a membrane blot (such as used in hybridization analysis such as northern, Southern, dot, and the like), or microwells, sample tubes, gels, beads or fibers (or any solid support comprising bound nucleic acids). See U.S. Pat. Nos. 5,770,722, 5,874,219, 5,744,305, 5,677,195 and 5,445,934, which are incorporated herein by reference. The determination of complement factor B expression level may also comprise using nucleic acid probes in solution.


In certain embodiments, the level of mRNA expression is assessed using branched DNA (bDNA) assays or real time PCR (qPCR). The use of these methods is described and exemplified in the Examples presented herein. In certain embodiments, expression level is determined by the method provided in Example 2 using a 10 nM siRNA concentration in the species matched cell line.


The level of CFB protein expression may be determined using any method known in the art for the measurement of protein levels. Such methods include, for example, electrophoresis, capillary electrophoresis, high performance liquid chromatography (HPLC), thin layer chromatography (TLC), hyperdiffusion chromatography, fluid or gel precipitin reactions, absorption spectroscopy, a colorimetric assays, spectrophotometric assays, flow cytometry, immunodiffusion (single or double), immunoelectrophoresis, western blotting, radioimmunoassay (RIA), enzyme-linked immunosorbent assays (ELISAs), immunofluorescent assays, electrochemiluminescence assays, and the like.


In some embodiments, the efficacy of the methods of the invention are assessed by a decrease in CFB mRNA or protein level (e.g., in a liver biopsy).


In some embodiments of the methods of the invention, the iRNA is administered to a subject such that the iRNA is delivered to a specific site within the subject. The inhibition of expression of complement factor B may be assessed using measurements of the level or change in the level of complement factor B mRNA or complement factor B protein in a sample derived from fluid or tissue from the specific site within the subject (e.g., liver or blood).


As used herein, the terms detecting or determining a level of an analyte are understood to mean performing the steps to determine if a material, e.g., protein, RNA, is present. As used herein, methods of detecting or determining include detection or determination of an analyte level that is below the level of detection for the method used.


VII. Prophylactic and Treatment Methods of the Invention

The present invention also provides methods of using an iRNA of the invention or a composition containing an iRNA of the invention to inhibit expression of complement factor B, thereby preventing or treating a complement factor B-associated disorder, e.g., paroxysmal nocturnal hemoglobinuria (PNH), atypical hemolytic uremic syndrome (aHUS), asthma, rheumatoid arthritis (RA); antiphospholipid antibody syndrome; lupus nephritis; ischemia-reperfusion injury; typical or infectious hemolytic uremic syndrome (tHUS); dense deposit disease (DDD); neuromyelitis optica (NMO); multifocal motor neuropathy (MMN); multiple sclerosis (MS); macular degeneration (e.g., age-related macular degeneration (AMD)); hemolysis, elevated liver enzymes, and low platelets (HELLP) syndrome; thrombotic thrombocytopenic purpura (TTP); spontaneous fetal loss; Pauci-immune vasculitis; epidermolysis bullosa; recurrent fetal loss; pre-eclampsia, traumatic brain injury, myasthenia gravis, cold agglutinin disease, dermatomyositis bullous pemphigoid, Shiga toxin E. coli-related hemolytic uremic syndrome, C3 neuropathy, anti-neutrophil cytoplasmic antibody-associated vasculitis (e.g., granulomatosis with polyangiitis (previously known as Wegener granulomatosis), Churg-Strauss syndrome, and microscopic polyangiitis), humoral and vascular transplant rejection, graft dysfunction, myocardial infarction (e.g., tissue damage and ischemia in myocardial infarction), an allogenic transplant, sepsis (e.g., poor outcome in sepsis), Coronary artery disease, dermatomyositis, Graves' disease, atherosclerosis, Alzheimer's disease, systemic inflammatory response sepsis, septic shock, spinal cord injury, glomerulonephritis, Hashimoto's thyroiditis, type I diabetes, psoriasis, pemphigus, autoimmune hemolytic anemia (AIHA), ITP, Goodpasture syndrome, Degos disease, antiphospholipid syndrome (APS), catastrophic APS (CAPS), a cardiovascular disorder, myocarditis, a cerebrovascular disorder, a peripheral (e.g., musculoskeletal) vascular disorder, a renovascular disorder, a mesenteric/enteric vascular disorder, vasculitis, Henoch-Schönlein purpura nephritis, systemic lupus erythematosus-associated vasculitis, vasculitis associated with rheumatoid arthritis, immune complex vasculitis, Takayasu's disease, dilated cardiomyopathy, diabetic angiopathy, Kawasaki's disease (arteritis), venous gas embolus (VGE), and restenosis following stent placement, rotational atherectomy, and percutaneous transluminal coronary angioplasty (PTCA) (see, e.g., Holers (2008) Immunological Reviews 223:300-316; Holers and Thurman (2004) Molecular Immunology 41:147-152; U.S. Patent Publication No. 20070172483).


In one embodiment, the complement factor B-associate disease is selected from the group consisting of C3 glomerulopathy, systemic lupus erythematosus (SLE), e.g., Lupus Nephritis, IgA nephropathy, diabetic nephropathy, polycystic kidney disease, membranous nephropathy, age-related macular degeneration, atypical hemolytic uremic syndrome, thrombotic microangiopathy, myasthenia gravis, ischemia and reperfusion injury, paroxysmal nocturnal hemoglobinuria, and rheumatoid arthritis


In another embodiment, the complement factor B-associate disease is selected from the group consisting of C3 glomerulopathy, systemic lupus erythematosus (SLE), e.g., Lupus Nephritis, IgA nephropathy, diabetic nephropathy, and polycystic kidney disease.


In the methods of the invention the cell may be contacted with the siRNA in vitro or in vivo, i.e., the cell may be within a subject.


A cell suitable for treatment using the methods of the invention may be any cell that expresses a complement factor B gene, e.g., a liver cell, a brain cell, a gall bladder cell, a heart cell, or a kidney cell. In one embodiment, the cell is a liver cell. A cell suitable for use in the methods of the invention may be a mammalian cell, e.g., a primate cell (such as a human cell, including human cell in a chimeric non-human animal, or a non-human primate cell, e.g., a monkey cell or a chimpanzee cell), or a non-primate cell. In certain embodiments, the cell is a human cell, e.g., a human liver cell. In the methods of the invention, complement factor B expression is inhibited in the cell by at least 50, 55, 60, 65, 70, 75, 80, 85, 90, or 95, or to a level below the level of detection of the assay.


The in vivo methods of the invention may include administering to a subject a composition containing an iRNA, where the iRNA includes a nucleotide sequence that is complementary to at least a part of an RNA transcript of the complement factor B gene of the mammal to which the RNAi agent is to be administered. The composition can be administered by any means known in the art including, but not limited to oral, intraperitoneal, or parenteral routes, including intracranial (e.g., intraventricular, intraparenchymal, and intrathecal), intravenous, intramuscular, subcutaneous, transdermal, airway (aerosol), nasal, rectal, and topical (including buccal and sublingual) administration. In certain embodiments, the compositions are administered by intravenous infusion or injection. In certain embodiments, the compositions are administered by subcutaneous injection. In certain embodiments, the compositions are administered by intramuscular injection.


In some embodiments, the administration is via a depot injection. A depot injection may release the iRNA in a consistent way over a prolonged time period. Thus, a depot injection may reduce the frequency of dosing needed to obtain a desired effect, e.g., a desired inhibition of CFB, or a therapeutic or prophylactic effect. A depot injection may also provide more consistent serum concentrations. Depot injections may include subcutaneous injections or intramuscular injections. In certain embodiments, the depot injection is a subcutaneous injection.


In some embodiments, the administration is via a pump. The pump may be an external pump or a surgically implanted pump. In certain embodiments, the pump is a subcutaneously implanted osmotic pump. In other embodiments, the pump is an infusion pump. An infusion pump may be used for intravenous, subcutaneous, arterial, or epidural infusions. In certain embodiments, the infusion pump is a subcutaneous infusion pump. In other embodiments, the pump is a surgically implanted pump that delivers the iRNA to the liver.


The mode of administration may be chosen based upon whether local or systemic treatment is desired and based upon the area to be treated. The route and site of administration may be chosen to enhance targeting.


In one aspect, the present invention also provides methods for inhibiting the expression of a complement factor B gene in a mammal. The methods include administering to the mammal a composition comprising a dsRNA that targets a complement factor B gene in a cell of the mammal and maintaining the mammal for a time sufficient to obtain degradation of the mRNA transcript of the complement factor B gene, thereby inhibiting expression of the complement factor B gene in the cell. Reduction in gene expression can be assessed by any methods known in the art and by methods, e.g. qRT-PCR, described herein, e.g., in Example 2. Reduction in protein production can be assessed by any methods known it the art, e.g. ELISA. In certain embodiments, a puncture liver biopsy sample serves as the tissue material for monitoring the reduction in the complement factor B gene or protein expression. In other embodiments, a blood sample serves as the subject sample for monitoring the reduction in the complement factor B protein expression.


The present invention further provides methods of treatment in a subject in need thereof, e.g., a subject diagnosed with a complement factor B-associated disorder, such as, C3 glomerulopathy, systemic lupus erythematosus (SLE), e.g., Lupus Nephritis, IgA nephropathy, diabetic nephropathy, and polycystic kidney disease.


The present invention further provides methods of prophylaxis in a subject in need thereof. The treatment methods of the invention include administering an iRNA of the invention to a subject, e.g., a subject that would benefit from a reduction of complement factor B expression, in a prophylactically effective amount of an iRNA targeting a complement factor B gene or a pharmaceutical composition comprising an iRNA targeting a complement factor B gene.


In one embodiment, a complement factor B-associated disease is selected from the group consisting of paroxysmal nocturnal hemoglobinuria (PNH), atypical hemolytic uremic syndrome (aHUS), asthma, rheumatoid arthritis (RA); antiphospholipid antibody syndrome; lupus nephritis; ischemia-reperfusion injury; typical or infectious hemolytic uremic syndrome (tHUS); dense deposit disease (DDD); neuromyelitis optica (NMO); multifocal motor neuropathy (MMN); multiple sclerosis (MS); macular degeneration (e.g., age-related macular degeneration (AMD)); hemolysis, elevated liver enzymes, and low platelets (HELLP) syndrome; thrombotic thrombocytopenic purpura (TTP); spontaneous fetal loss; Pauci-immune vasculitis; epidermolysis bullosa; recurrent fetal loss; pre-eclampsia, traumatic brain injury, myasthenia gravis, cold agglutinin disease, dermatomyositis bullous pemphigoid, Shiga toxin E. coli-related hemolytic uremic syndrome, C3 neuropathy, anti-neutrophil cytoplasmic antibody-associated vasculitis (e.g., granulomatosis with polyangiitis (previously known as Wegener granulomatosis), Churg-Strauss syndrome, and microscopic polyangiitis), humoral and vascular transplant rejection, graft dysfunction, myocardial infarction (e.g., tissue damage and ischemia in myocardial infarction), an allogenic transplant, sepsis (e.g., poor outcome in sepsis), Coronary artery disease, dermatomyositis, Graves' disease, atherosclerosis, Alzheimer's disease, systemic inflammatory response sepsis, septic shock, spinal cord injury, glomerulonephritis, Hashimoto's thyroiditis, type I diabetes, psoriasis, pemphigus, autoimmune hemolytic anemia (AIHA), ITP, Goodpasture syndrome, Degos disease, antiphospholipid syndrome (APS), catastrophic APS (CAPS), a cardiovascular disorder, myocarditis, a cerebrovascular disorder, a peripheral (e.g., musculoskeletal) vascular disorder, a renovascular disorder, a mesenteric/enteric vascular disorder, vasculitis, Henoch-Schonlein purpura nephritis, systemic lupus erythematosus-associated vasculitis, vasculitis associated with rheumatoid arthritis, immune complex vasculitis, Takayasu's disease, dilated cardiomyopathy, diabetic angiopathy, Kawasaki's disease (arteritis), venous gas embolus (VGE), and restenosis following stent placement, rotational atherectomy, and percutaneous transluminal coronary angioplasty (PTCA) (see, e.g., Holers (2008) Immunological Reviews 223:300-316; Holers and Thurman (2004) Molecular Immunology 41:147-152; US20070172483).


In one embodiment, the complement factor B-associate disease is selected from the group consisting of C3 glomerulopathy, systemic lupus erythematosus (SLE), e.g., Lupus Nephritis, IgA nephropathy, diabetic nephropathy, polycystic kidney disease, membranous nephropathy, age-related macular degeneration, atypical hemolytic uremic syndrome, thrombotic microangiopathy, myasthenia gravis, ischemia and reperfusion injury, paroxysmal nocturnal hemoglobinuria, and rheumatoid arthritis


In another embodiment, the complement factor B-associate disease is selected from the group consisting of C3 glomerulopathy, systemic lupus erythematosus (SLE), e.g., Lupus Nephritis, IgA nephropathy, diabetic nephropathy, and polycystic kidney disease.


An iRNA of the invention may be administered as a “free iRNA.” A free iRNA is administered in the absence of a pharmaceutical composition. The naked iRNA may be in a suitable buffer solution. The buffer solution may comprise acetate, citrate, prolamine, carbonate, or phosphate, or any combination thereof. In one embodiment, the buffer solution is phosphate buffered saline (PBS). The pH and osmolarity of the buffer solution containing the iRNA can be adjusted such that it is suitable for administering to a subject.


Alternatively, an iRNA of the invention may be administered as a pharmaceutical composition, such as a dsRNA liposomal formulation.


Subjects that would benefit from an inhibition of complement factor B gene expression are subjects susceptible to or diagnosed with a CFB-associated disorder, e.g., C3 glomerulopathy, systemic lupus erythematosus (SLE), e.g., Lupus Nephritis, IgA nephropathy, diabetic nephropathy, and polycystic kidney disease.


In an embodiment, the method includes administering a composition featured herein such that expression of the target complement component B gene is decreased, such as for about 1, 2, 3, 4, 5, 6, 1-6, 1-3, or 3-6 months per dose. In certain embodiments, the composition is administered once every 3-6 months.


In one embodiment, the iRNAs useful for the methods and compositions featured herein specifically target RNAs (primary or processed) of the target complement factor B gene. Compositions and methods for inhibiting the expression of these genes using iRNAs can be prepared and performed as described herein.


Administration of the iRNA according to the methods of the invention may result prevention or treatment of a complement factor B-associated disorder, e.g., C3 glomerulopathy, systemic lupus erythematosus (SLE), e.g., Lupus Nephritis, IgA nephropathy, diabetic nephropathy, and polycystic kidney disease.


Subjects can be administered a therapeutic amount of iRNA, such as about 0.01 mg/kg to about 200 mg/kg. Subjects can be administered a therapeutic amount of iRNA, such as about 5 mg to about 1000 mg as a fixed dose, regardless of body weight.


In some embodiment, the iRNA is administered subcutaneously, i.e., by subcutaneous injection. One or more injections may be used to deliver the desired dose of iRNA to a subject. The injections may be repeated over a period of time.


The administration may be repeated on a regular basis. In certain embodiments, after an initial treatment regimen, the treatments can be administered on a less frequent basis. A repeat-dose regimen may include administration of a therapeutic amount of iRNA on a regular basis, such as once per month to once a year. In certain embodiments, the iRNA is administered about once per month to about once every three months, or about once every three months to about once every six months.


The invention further provides methods and uses of an iRNA agent or a pharmaceutical composition thereof for treating a subject that would benefit from reduction or inhibition of CFB gene expression, e.g., a subject having a CFB-associated disease, in combination with other pharmaceuticals or other therapeutic methods, e.g., with known pharmaceuticals or known therapeutic methods, such as, for example, those which are currently employed for treating these disorders.


Accordingly, in some aspects of the invention, the methods which include either a single iRNA agent of the invention, further include administering to the subject one or more additional therapeutic agents. The iRNA agent and an additional therapeutic agent or treatment may be administered at the same time or in the same combination, e.g., parenterally, or the additional therapeutic agent can be administered as part of a separate composition or at separate times or by another method known in the art or described herein.


In one embodiment, an iRNA agent of the invention is administered in combination with an anti-complement component C5 antibody, or antigen-binding fragment thereof (e.g., eculizumab or ravulizumab-cwvz), an iRNA agent targeting complement component C5, an iRNA agent targeting complement component C3, or a C3 peptide inhibitor (e.g., compstatin). In one embodiment, the iRNA agent of the invention is administered to the patient, and then the additional therapeutic agent is administered to the patient (or vice versa). In another embodiment, the iRNA agent of the invention and the additional therapeutic agent are administered at the same time.


The iRNA agent of the invention and an additional therapeutic agent or treatment may be administered at the same time or in the same combination, e.g., parenterally, or the additional therapeutic agent can be administered as part of a separate composition or at separate times or by another method known in the art or described herein.


VIII. Kits

In certain aspects, the instant disclosure provides kits that include a suitable container containing a pharmaceutical formulation of a siRNA compound, e.g., a double-stranded siRNA compound, or ssiRNA compound, (e.g., a precursor, e.g., a larger siRNA compound which can be processed into a ssiRNA compound, or a DNA which encodes an siRNA compound, e.g., a double-stranded siRNA compound, or ssiRNA compound, or precursor thereof).


Such kits include one or more dsRNA agent(s) and instructions for use, e.g., instructions for administering a prophylactically or therapeutically effective amount of a dsRNA agent(s). The dsRNA agent may be in a vial or a pre-filled syringe. The kits may optionally further comprise means for administering the dsRNA agent (e.g., an injection device, such as a pre-filled syringe), or means for measuring the inhibition of CFB (e.g., means for measuring the inhibition of CFB mRNA, CFB protein, or CFB activity). Such means for measuring the inhibition of CFB may comprise a means for obtaining a sample from a subject, such as, e.g., a plasma sample. The kits of the invention may optionally further comprise means for determining the therapeutically effective or prophylactically effective amount.


In certain embodiments the individual components of the pharmaceutical formulation may be provided in one container, e.g., a vial or a pre-filled syringe. Alternatively, it may be desirable to provide the components of the pharmaceutical formulation separately in two or more containers, e.g., one container for a siRNA compound preparation, and at least another for a carrier compound. The kit may be packaged in a number of different configurations such as one or more containers in a single box. The different components can be combined, e.g., according to instructions provided with the kit. The components can be combined according to a method described herein, e.g., to prepare and administer a pharmaceutical composition. The kit can also include a delivery device.


This invention is further illustrated by the following examples which should not be construed as limiting. The entire contents of all publications, patents and published patent applications cited throughout this application, as well as the informal Sequence Listing and Figures, are hereby incorporated herein by reference.


EXAMPLES
Example 1. iRNA Synthesis
Source of Reagents

Where the source of a reagent is not specifically given herein, such reagent can be obtained from any supplier of reagents for molecular biology at a quality/purity standard for application in molecular biology.


siRNA Design


siRNAs targeting the human complement factor B (CFB) gene, (human: NCBI refseqID NM_001710.5; NCBI GeneID: 629) was designed using custom R and Python scripts. The human NM_001710 REFSEQ mRNA, version 5, has a length of 2646 bases. Detailed lists of the unmodified CFB sense and antisense strand nucleotide sequences are shown in Tables 2, 4, and 6. Detailed lists of the modified CFB sense and antisense strand nucleotide sequences are shown in Tables 3, 5, and 7.


It is to be understood that, throughout the application, a duplex name without a decimal is equivalent to a duplex name with a decimal which merely references the batch number of the duplex. For example, AD-959917 is equivalent to AD-959917.1.


siRNA Synthesis


siRNAs were synthesized and annealed using routine methods known in the art.


Example 2. In Vitro Screening Methods
Cell Culture and 384-Well Transfections

Hep3b cells (ATCC, Manassas, VA) were grown to near confluence at 37° C. in an atmosphere of 5% CO2 in Eagle's Minimum Essential Medium (Gibco) supplemented with 10% FBS (ATCC) before being released from the plate by trypsinization. Primary mouse hepatocytes (PMH) were freshly isolated less than 1 hour prior to transfections and grown in primary hepatocyte media. For both Hep3B and PMH, transfection was carried out by adding 5 μl of Opti-MEM plus 0.1 μl of Lipofectamine RNAiMax per well (Invitrogen, Carlsbad CA. cat #13778-150) to 5 μl of each siRNA duplex to an individual well in a 384-well plate. The mixture was then incubated at room temperature for 15 minutes. Forty μl of Eagle's Minimum Essential Medium (ATCC Cat #30-2003) containing ˜5×103 Hep3B cells or PMH were then added to the siRNA mixture. Cells were incubated for 24 hours prior to RNA purification. Single dose experiments were performed at 10 nM, 1 nM, and 0.1 nM.


Total RNA Isolation Using DYNABEADS mRNA Isolation Kit (Invitrogen™, Part #: 610-12)


RNA was isolated using an automated protocol on a BioTek-EL406 platform using DYNABEADs (Invitrogen, cat #61012). Briefly, 70 μl of Lysis/Binding Buffer and 10 μl of lysis buffer containing 3 μl of magnetic beads were added to the plate with cells. Plates were incubated on an electromagnetic shaker for 10 minutes at room temperature and then magnetic beads were captured and the supernatant was removed. Bead-bound RNA was then washed 2 times with 150 μl Wash Buffer A and once with Wash Buffer B. Beads were then washed with 150 μl Elution Buffer, re-captured and supernatant removed.


cDNA Synthesis Using ABI High Capacity cDNA Reverse Transcription Kit (Applied Biosystems, Foster City, CA, Cat #4368813)


A master mix of 1.2 μl 10× Buffer, 0.48 μl 25×dNTPs, 1.2 μl Random primers, 0.6 μl Reverse Transcriptase, 0.6 μl RNase inhibitor and 7.92 μl of H2O per reaction were added per well. Plates were sealed, mixed, and then incubated on an electromagnetic shaker for 10 minutes at room temperature, followed by incubation at 37° C. for 2 hours.


Real Time PCR

Two μl of cDNA were added to a master mix containing 0.5 μl of human GAPDH TaqMan Probe (4326317E), and 0.5 μl CFB human probe (Hs01071998_m1) and 5 μl Lightcycler 480 probe master mix (Roche Cat #04887301001) per well in a 384 well plates (Roche cat #04887301001). Real time PCR was done in a LightCycler480 Real Time PCR system (Roche). Each duplex was tested at least two times and data were normalized to cells transfected with a non-targeting control siRNA. To calculate relative fold change, real time data were analyzed using the ΔΔCt method and normalized to assays performed with cells transfected with a non-targeting control siRNA.


The results of the single dose screen of the dsRNA agents in Tables 2 are 3 in Hep3B cells are shown in Table 8. The results of the single dose screen of the dsRNA agents Tables 4 and 5 in Hep3B cells are shown in Table 9. The results of the single dose screen of the dsRNA agents Tables 4 and 5 in PMH cells are shown in Table 10. The results of the single dose screen of the dsRNA agents Tables 6 and 7 in PMH cells are shown in Table 11. The results of the single dose screen of the dsRNA agents Tables 6 and 7 in Hep3B cells are shown in Table 12.


ELISA Assay

Human CFB protein levels were determined using a quantitative sandwich enzyme immunoassay (Human Complement Factor B AssayMax ELISA Kit—AssayPro). Briefly, samples were diluted 1:1000 and 50 μl of sample was added to a well of a microtiter plate. Samples were incubated for two hours and subsequently washed. Fifty μl of biotinylated anti-CFB antibody was added to each well and incubated 1 hour. Samples were then washed and 50 μl of streptavidin-peroxidase conjugate was added to each well and incubated for 30 minutes. Following another wash, 50 μl of peroxidase enzyme substrate per well was added and samples were incubated for 15 minutes after which 50 μl of Stop Solution per well was added. Samples were read at 450 nm immediately and the amount of human CFB protein was determined by comparing the reading to a standard curve (0-280 ng of human CFB protein).









TABLE 1







Abbreviations of nucleotide monomers used in nucleic acid sequence representation. It will


be understood that these monomers, when present in an oligonucleotide, are mutually linked by 5′-3′-


phosphodiester bonds; and it is understood that when the nucleotide contains a 2′-fluoro modification,


then the fluoro replaces the hydroxy at that position in the parent nucleotide (i.e., it is a 2′-deoxy-2′-


fluoronucleotide).








Abbreviation
Nucleotide(s)





A
Adenosine-3′-phosphate


Ab
beta-L-adenosine-3′-phosphate


Abs
beta-L-adenosine-3′-phosphorothioate


Af
2′-fluoroadenosine-3′-phosphate


Afs
2′-fluoroadenosine-3′-phosphorothioate


As
adenosine-3′-phosphorothioate


C
cytidine-3′-phosphate


Cb
beta-L-cytidine-3′-phosphate


Cbs
beta-L-cytidine-3′-phosphorothioate


Cf
2′-fluorocytidine-3′-phosphate


Cfs
2′-fluorocytidine-3′-phosphorothioate


Cs
cytidine-3′-phosphorothioate


G
guanosine-3′-phosphate


Gb
beta-L-guanosine-3′-phosphate


Gbs
beta-L-guanosine-3′-phosphorothioate


Gf
2′-fluoroguanosine-3′-phosphate


Gfs
2′-fluoroguanosine-3′-phosphorothioate


Gs
guanosine-3′-phosphorothioate


T
5′-methyluridine-3′-phosphate


Tf
2′-fluoro-5-methyluridine-3′-phosphate


Tfs
2′-fluoro-5-methyluridine-3′-phosphorothioate


Ts
5-methyluridine-3′-phosphorothioate


U
Uridine-3′-phosphate


Uf
2′-fluorouridine-3′-phosphate


Ufs
2′-fluorouridine -3′-phosphorothioate


Us
uridine-3′-phosphorothioate


N
any nucleotide, modified or unmodified


a
2′-O-methyladenosine-3′-phosphate


as
2′-O-methyladenosine-3′-phosphorothioate


C
2′-O-methylcytidine-3′-phosphate


CS
2′-O-methylcytidine-3′-phosphorothioate


g
2′-O-methylguanosine-3′-phosphate


gs
2′-O-methylguanosine-3′-phosphorothioate


t
2′-O-methyl-5-methyluridine-3′-phosphate


ts
2′-O-methyl-5-methyluridine-3′-phosphorothioate


U
2′-O-methyluridine-3′-phosphate


us
2′-O-methyluridine-3′-phosphorothioate


S
phosphorothioate linkage


L10
N-(cholesterylcarboxamidocaproyl)-4-hydroxyprolinol (Hyp-C6-Chol)





L96
N-[tris(GalNAc-alkyl)-amidodecanoyl)]-4-hydroxyprolinol (Hyp-(GalNAc-alkyl)3) embedded image





Y34
2-hydroxymethyl-tetrahydrofurane-4-methoxy-3-phosphate (abasic 2′-OMe



furanose)


Y44
inverted abasic DNA (2-hydroxymethyl-tetrahydrofurane-5-phosphate)


(Agn)
Adenosine-glycol nucleic acid (GNA)


(Cgn)
Cytidine-glycol nucleic acid (GNA)


(Ggn)
Guanosine-glycol nucleic acid (GNA)


(Tgn)
Thymidine-glycol nucleic acid (GNA) S-Isomer


P
Phosphate


VP
Vinyl-phosphonate


dA
2′-deoxyadenosine-3′-phosphate


dAs
2′-deoxyadenosine-3′-phosphorothioate


dC
2′-deoxycytidine-3′-phosphate


dCs
2′-deoxycytidine-3′-phosphorothioate


dG
2′-deoxyguanosine-3′-phosphate


dGs
2′-deoxyguanosine-3′-phosphorothioate


dT
2′-deoxythymidine-3′-phosphate


dTs
2′-deoxythymidine-3′-phosphorothioate


dU
2′-deoxyuridine


dUs
2′-deoxyuridine-3′-phosphorothioate


(C2p)
cytidine-2′-phosphate


(G2p)
guanosine-2′-phosphate


(U2p)
uridine-2′-phosphate


(A2p)
adenosine-2 -phosphate


(Chd)
2′-O-hexadecyl-cytidine-3′-phosphate


(Ahd)
2′-O-hexadecyl-adenosine-3′-phosphate


(Ghd)
2′-O-hexadecyl-guanosine-3′-phosphate


(Uhd)
2′-O-hexadecyl-uridine-3′-phosphate
















TABLE 2







Unmodified Sense and Antisense Strand Sequences of Complement 


Factor B dsRNA Agents















SEQ
Range

SEQ
Range


Duplex 
Sense Sequence
ID
(NM_
Antiense Sequence
ID
(NM_


Name
5′ to 3′
NO:
001710.5)
5′ to 3′
NO:
001710.5)





AD-557072.1
CAAAAAGUGUCUAGUCAACUU
 19
1145-1165
AAGUUGACUAGACACUUUUUGGC
105
1143-1165





AD-558097.1
UAGUGGAUGUCUGCAAAAACU
 20
2440-2460
AGUUUUTGCAGACAUCCACUACU
106
2438-2460





AD-557774.1
GAAUUCCUGAAUUUUAUGACU
 21
1981-2001
AGUCAUAAAAUUCAGGAAUUCCU
107
1979-2001





AD-557070.1
GCCAAAAAGUGUCUAGUCAAU
 22
1143-1163
AUUGACTAGACACUUUUUGGCUC
108
1141-1163





AD-558068.1
UCACAAGAGAAGUCGUUUCAU
 23
2393-2413
AUGAAACGACUUCUCUUGUGAAC
109
2391-2413





AD-557068.1
GAGCCAAAAAGUGUCUAGUCU
 24
1141-1161
AGACUAGACACUUUUUGGCUCCU
110
1139-1161





AD-556701.1
CUACUACAAUGUGAGUGAUGU
 25
 635-655
ACAUCACUCACAUUGUAGUAGGG
111
 633-655





AD-558076.1
GAAGUCGUUUCAUUCAAGUUU
 26
2401-2421
AAACUUGAAUGAAACGACUUCUC
112
2399-2421





AD-558065.1
AGUUCACAAGAGAAGUCGUUU
 27
2390-2410
AAACGACUUCUCUUGUGAACUAU
113
2388-2410





AD-557859.1
CAACUCGAGCUUUGAGGCUUU
 28
2086-2106
AAAGCCTCAAAGCUCGAGUUGUU
114
2084-2106





AD-558096.1
GUAGUGGAUGUCUGCAAAAAU
 29
2439-2459
AUUUUUGCAGACAUCCACUACUC
115
2437-2459





AD-557422.1
GAGGAUUAUCUGGAUGUCUAU
 30
1536-1556
AUAGACAUCCAGAUAAUCCUCCC
116
1534-1556





AD-556919.1
AGACUCCUUCAUGUACGACAU
 31
 935-955
AUGUCGTACAUGAAGGAGUCUUG
117
 933-955





AD-558069.1
CACAAGAGAAGUCGUUUCAUU
 32
2394-2414
AAUGAAACGACUUCUCUUGUGAA
118
2392-2414





AD-558063.1
AUAGUUCACAAGAGAAGUCGU
 33
2388-2408
ACGACUTCUCUUGUGAACUAUCA
119
2386-2408





AD-557069.1
AGCCAAAAAGUGUCUAGUCAU
 34
1142-1162
AUGACUAGACACUUUUUGGCUCC
120
1140-1162





AD-558225.1
UGAAUUAAAACAGCUGCGACU
 35
2604-2624
AGUCGCAGCUGUUUUAAUUCAAU
121
2602-2624





AD-557853.1
AGGGAACAACUCGAGCUUUGU
 36
2080-2100
ACAAAGCUCGAGUUGUUCCCUCG
122
2078-2100





AD-558012.1
GACAAAGUCAAGGACAUCUCU
 37
2277-2297
AGAGAUGUCCUUGACUUUGUCAU
123
2275-2297





AD-558061.1
UGAUAGUUCACAAGAGAAGUU
 38
2386-2406
AACUUCTCUUGUGAACUAUCAAG
124
2384-2406





AD-557079.1
UGUCUAGUCAACUUAAUUGAU
 39
1152-1172
AUCAAUTAAGUUGACUAGACACU
125
1150-1172





AD-558066.1
GUUCACAAGAGAAGUCGUUUU
 40
2391-2411
AAAACGACUUCUCUUGUGAACUA
126
2389-2411





AD-557353.1
GACCCAAUUACUGUCAUUGAU
 41
1467-1487
AUCAAUGACAGUAAUUGGGUCCC
127
1465-1487





AD-557782.1
GAAUUUUAUGACUAUGACGUU
 42
1989-2009
AACGUCAUAGUCAUAAAAUUCAG
128
1987-2009





AD-556390.1
CUGGAGUUUCAGCUUGGACAU
 43
 179-199
AUGUCCAAGCUGAAACUCCAGAC
129
 177-199





AD-557078.1
GUGUCUAGUCAACUUAAUUGU
 44
1151-1171
ACAAUUAAGUUGACUAGACACUU
130
1149-1171





AD-557475.1
UUCCAAGAAAGACAAUGAGCU
 45
1607-1627
AGCUCATUGUCUUUCUUGGAAGC
131
1605-1627





AD-556734.1
CUGCUAUGACGGUUACACUCU
 46
 668-688
AGAGUGTAACCGUCAUAGCAGUG
132
 666-688





AD-557084.1
AGUCAACUUAAUUGAGAAGGU
 47
1157-1177
ACCUUCTCAAUUAAGUUGACUAG
133
1155-1177





AD-557498.1
AUGUGUUCAAAGUCAAGGAUU
 48
1630-1650
AAUCCUTGACUUUGAACACAUGU
134
1628-1650





AD-556788.1
GACAGCGAUCUGUGACAACGU
 49
 740-760
ACGUUGTCACAGAUCGCUGUCUG
135
 738-760





AD-557067.1
GGAGCCAAAAAGUGUCUAGUU
 50
1140-1160
AACUAGACACUUUUUGGCUCCUG
136
1138-1160





AD-557852.1
GAGGGAACAACUCGAGCUUUU
 51
2079-2099
AAAAGCTCGAGUUGUUCCCUCGG
137
2077-2099





AD-556733.1
ACUGCUAUGACGGUUACACUU
 52
 667-687
AAGUGUAACCGUCAUAGCAGUGG
138
 665-687





AD-556725.1
CUCUUUCCACUGCUAUGACGU
 53
 659-679
ACGUCATAGCAGUGGAAAGAGAU
139
 657-679





AD-557860.1
AACUCGAGCUUUGAGGCUUCU
 54
2087-2107
AGAAGCCUCAAAGCUCGAGUUGU
140
2085-2107





AD-556786.1
CAGACAGCGAUCUGUGACAAU
 55
 738-758
AUUGUCACAGAUCGCUGUCUGCC
141
 736-758





AD-556581.1
CCUUCUGGCUUCUACCCGUAU
 56
 465-485
AUACGGGUAGAAGCCAGAAGGAC
142
 463-485





AD-556963.1
ACCAUAGAAGGAGUCGAUGCU
 57
 999-1019
AGCAUCGACUCCUUCUAUGGUCU
143
 997-1019





AD-557450.1
ACCAAGUGAACAUCAAUGCUU
 58
1582-1602
AAGCAUTGAUGUUCACUUGGUUC
144
1580-1602





AD-557204.1
UGAAAUCAAUUAUGAAGACCU
 59
1298-1318
AGGUCUTCAUAAUUGAUUUCAUU
145
1296-1318





AD-558062.1
GAUAGUUCACAAGAGAAGUCU
 60
2387-2407
AGACUUCUCUUGUGAACUAUCAA
146
2385-2407





AD-557602.1
AAGGGUACCGAUUACCACAAU
 61
1734-1754
AUUGUGGUAAUCGGUACCCUUCC
147
1732-1754





AD-556917.1
CAAGACUCCUUCAUGUACGAU
 62
 933-953
AUCGUACAUGAAGGAGUCUUGGC
148
 931-953





AD-557969.1
CUGACUCGGAAGGAGGUCUAU
 63
2196-2216
AUAGACCUCCUUCCGAGUCAGCU
149
2194-2216





AD-558064.1
UAGUUCACAAGAGAAGUCGUU
 64
2389-2409
AACGACTUCUCUUGUGAACUAUC
150
2387-2409





AD-558105.1
GUCUGCAAAAACCAGAAGCGU
 65
2448-2468
ACGCUUCUGGUUUUUGCAGACAU
151
2446-2468





AD-556791.1
AGCGAUCUGUGACAACGGAGU
 66
 743-763
ACUCCGTUGUCACAGAUCGCUGU
152
 741-763





AD-557972.1
ACUCGGAAGGAGGUCUACAUU
 67
2199-2219
AAUGUAGACCUCCUUCCGAGUCA
153
2197-2219





AD-556920.1
GACUCCUUCAUGUACGACACU
 68
 936-956
AGUGUCGUACAUGAAGGAGUCUU
154
 934-956





AD-558078.1
AGUCGUUUCAUUCAAGUUGGU
 69
2403-2423
ACCAACTUGAAUGAAACGACUUC
155
2401-2423





AD-557861.1
ACUCGAGCUUUGAGGCUUCCU
 70
2088-2108
AGGAAGCCUCAAAGCUCGAGUUG
156
2086-2108





AD-557856.1
GAACAACUCGAGCUUUGAGGU
 71
2083-2103
ACCUCAAAGCUCGAGUUGUUCCC
157
2081-2103





AD-557041.1
UGGUGCUAGAUGGAUCAGACU
 72
1096-1116
AGUCUGAUCCAUCUAGCACCAGG
158
1094-1116





AD-556874.1
GUGUCAGGAAGGUGGCUCUUU
 73
 890-910
AAAGAGCCACCUUCCUGACACGU
159
 888-910





AD-557345.1
CUGAUGGAUUGCACAACAUGU
 74
1441-1461
ACAUGUTGUGCAAUCCAUCAGUC
160
1439-1461





AD-557839.1
CAGACUAUCAGGCCCAUUUGU
 75
2046-2066
ACAAAUGGGCCUGAUAGUCUGGC
161
2044-2066





AD-556962.1
GACCAUAGAAGGAGUCGAUGU
 76
 998-1018
ACAUCGACUCCUUCUAUGGUCUC
162
 996-1018





AD-557066.1
AGGAGCCAAAAAGUGUCUAGU
 77
1139-1159
ACUAGACACUUUUUGGCUCCUGU
163
1137-1159





AD-556961.1
AGACCAUAGAAGGAGUCGAUU
 78
 997-1017
AAUCGACUCCUUCUAUGGUCUCU
164
 995-1017





AD-557226.1
AAGUUGAAGUCAGGGACUAAU
 79
1320-1340
AUUAGUCCCUGACUUCAACUUGU
165
1318-1340





AD-557865.1
GAGCUUUGAGGCUUCCUCCAU
 80
2092-2112
AUGGAGGAAGCCUCAAAGCUCGA
166
2090-2112





AD-557209.1
UCAAUUAUGAAGACCACAAGU
 81
1303-1323
ACUUGUGGUCUUCAUAAUUGAUU
167
1301-1323





AD-556918.1
AAGACUCCUUCAUGUACGACU
 82
 934-954
AGUCGUACAUGAAGGAGUCUUGG
168
 932-954





AD-557868.1
CUUUGAGGCUUCCUCCAACUU
 83
2095-2115
AAGUUGGAGGAAGCCUCAAAGCU
169
2093-2115





AD-557873.1
AGGCUUCCUCCAACUACCACU
 84
2100-2120
AGUGGUAGUUGGAGGAAGCCUCA
170
2098-2120





AD-556787.1
AGACAGCGAUCUGUGACAACU
 85
 739-759
AGUUGUCACAGAUCGCUGUCUGC
171
 737-759





AD-558004.1
CAGGCUAUGACAAAGUCAAGU
 86
2269-2289
ACUUGACUUUGUCAUAGCCUGGG
172
2267-2289





AD-557867.1
GCUUUGAGGCUUCCUCCAACU
 87
2094-2114
AGUUGGAGGAAGCCUCAAAGCUC
173
2092-2114





AD-557788.1
UAUGACUAUGACGUUGCCCUU
 88
1995-2015
AAGGGCAACGUCAUAGUCAUAAA
174
1993-2015





AD-558106.1
UCUGCAAAAACCAGAAGCGGU
 89
2449-2469
ACCGCUTCUGGUUUUUGCAGACA
175
2447-2469





AD-556724.1
UCUCUUUCCACUGCUAUGACU
 90
 658-678
AGUCAUAGCAGUGGAAAGAGAUC
176
 656-678





AD-556739.1
AUGACGGUUACACUCUCCGGU
 91
 673-693
ACCGGAGAGUGUAACCGUCAUAG
177
 671-693





AD-557417.1
CAAGGGAGGAUUAUCUGGAUU
 92
1531-1551
AAUCCAGAUAAUCCUCCCUUGGG
178
1529-1551





AD-556790.1
CAGCGAUCUGUGACAACGGAU
 93
 742-762
AUCCGUTGUCACAGAUCGCUGUC
179
 740-762





AD-557872.1
GAGGCUUCCUCCAACUACCAU
 94
2099-2119
AUGGUAGUUGGAGGAAGCCUCAA
180
2097-2119





AD-556738.1
UAUGACGGUUACACUCUCCGU
 95
 672-692
ACGGAGAGUGUAACCGUCAUAGC
181
 670-692





AD-557857.1
AACAACUCGAGCUUUGAGGCU
 96
2084-2104
AGCCUCAAAGCUCGAGUUGUUCC
182
2082-2104





AD-556726.1
UCUUUCCACUGCUAUGACGGU
 97
 660-680
ACCGUCAUAGCAGUGGAAAGAGA
183
 658-680





AD-557495.1
AACAUGUGUUCAAAGUCAAGU
 98
1627-1647
ACUUGACUUUGAACACAUGUUGC
184
1625-1647





AD-557016.1
CCUUCAGGCUCCAUGAACAUU
 99
1071-1091
AAUGUUCAUGGAGCCUGAAGGGU
185
1069-1091





AD-558074.1
GAGAAGUCGUUUCAUUCAAGU
100
2399-2419
ACUUGAAUGAAACGACUUCUCUU
186
2397-2419





AD-557604.1
GGGUACCGAUUACCACAAGCU
101
1736-1756
AGCUUGTGGUAAUCGGUACCCUU
187
1734-1756





AD-557346.1
UGAUGGAUUGCACAACAUGGU
102
1442-1462
ACCAUGTUGUGCAAUCCAUCAGU
188
1440-1462





AD-557851.1
CGAGGGAACAACUCGAGCUUU
103
2078-2098
AAAGCUCGAGUUGUUCCCUCGGU
189
2076-2098





AD-557786.1
UUUAUGACUAUGACGUUGCCU
104
1993-2013
AGGCAACGUCAUAGUCAUAAAAU
190
1991-2013
















TABLE 3







Modified Sense and Antisense Strand Sequences of Complement Factor B dsRNA Agents















SEQ

SEQ

SEQ




ID
Antisense
ID

ID


Duplex Name
Sense Sequence 5′ to 3′
NO:
Sequence 5′ to 3′
NO:
mRNA Target Sequence
NO:





AD-557072.1
csasaaaaGfuGfUfCfuagucaacuuL96
191
asAfsguug(Agn)cua
277
GCCAAAAAGUGUCUAGUCAACUU
363





gacAfcUfuuuugsgsc








AD-558097.1
usasguggAfuGfUfCfugcaaaaacuL96
192
asGfsuuuu(Tgn)gca
278
AGUAGUGGAUGUCUGCAAAAACC
364





gacAfuCfcacuascsu








AD-557774.1
gsasauucCfuGfAfAfuuuuaugacuL96
193
asGfsucau(Agn)aaa
279
AGGAAUUCCUGAAUUUUAUGACU
365





uucAfgGfaauucscsu








AD-557070.1
gscscaaaAfaGfUfGfucuagucaauL96
194
asUfsugac(Tgn)aga
280
GAGCCAAAAAGUGUCUAGUCAAC
366





cacUfuUfuuggcsusc








AD-558068.1
uscsacaaGfaGfAfAfgucguuucauL96
195
asUfsgaaa(Cgn)gac
281
GUUCACAAGAGAAGUCGUUUCAU
367





uucUfcUfugugasasc








AD-557068.1
gsasgccaAfaAfAfGfugucuagucuL96
196
asGfsacua(Ggn)aca
282
AGGAGCCAAAAAGUGUCUAGUCA
368





cuuUfuUfggcucscsu








AD-556701.1
csusacuaCfaAfUfGfugagugauguL96
197
asCfsauca(Cgn)uca
283
CCCUACUACAAUGUGAGUGAUGA
369





cauUfgUfaguagsgsg








AD-558076.1
gsasagucGfuUfUfCfauucaaguuuL96
198
asAfsacuu(Ggn)aau
284
GAGAAGUCGUUUCAUUCAAGUUG
370





gaaAfcGfacuucsusc








AD-557859.1
csasacucGfaGfCfUfuugaggcuuuL96
200
asAfsagcc(Tgn)caa
286
AACAACUCGAGCUUUGAGGCUUC
372





agcUfcGfaguugsusu








AD-558096.1
gsusagugGfaUfGfUfcugcaaaaauL96
201
asUfsuuuu(Ggn)cag
287
GAGUAGUGGAUGUCUGCAAAAAC
373





acaUfcCfacuacsusc








AD-557422.1
gsasggauUfaUfCfUfggaugucuauL96
202
asUfsagac(Agn)ucc
288
GGGAGGAUUAUCUGGAUGUCUAU
374





agaUfaAfuccucscsc








AD-556919.1
asgsacucCfuUfCfAfuguacgacauL96
203
asUfsgucg(Tgn)aca
289
CAAGACUCCUUCAUGUACGACAC
375





ugaAfgGfagucususg








AD-558069.1
csascaagAfgAfAfGfucguuucauuL96
204
asAfsugaa(Agn)cga
290
UUCACAAGAGAAGUCGUUUCAUU
376





cuuCfuCfuugugsasa








AD-558063.1
asusaguuCfaCfAfAfgagaagucguL96
205
asCfsgacu(Tgn)cuc
291
UGAUAGUUCACAAGAGAAGUCGU
377





uugUfgAfacuauscsa








AD-557069.1
asgsccaaAfaAfGfUfgucuagucauL96
206
asUfsgacu(Agn)gac
292
GGAGCCAAAAAGUGUCUAGUCAA
378





acuUfuUfuggcuscsc








AD-558225.1
usgsaauuAfaAfAfCfagcugcgacuL96
207
asGfsucgc(Agn)gcu
293
AUUGAAUUAAAACAGCUGCGACA
379





guuUfuAfauucasasu








AD-557853.1
asgsggaaCfaAfCfUfcgagcuuuguL96
208
asCfsaaag(Cgn)ucg
294
CGAGGGAACAACUCGAGCUUUGA
380





aguUfgUfucccuscsg








AD-558012.1
gsascaaaGfuCfAfAfggacaucucuL96
209
asGfsagau(Ggn)ucc
295
AUGACAAAGUCAAGGACAUCUCA
381





uugAfcUfuugucsasu








AD-558061.1
usgsauagUfuCfAfCfaagagaaguuL96
210
asAfscuuc(Tgn)cuu
296
CUUGAUAGUUCACAAGAGAAGUC
382





gugAfaCfuaucasasg








AD-557079.1
usgsucuaGfuCfAfAfcuuaauugauL96
211
asUfscaau(Tgn)aag
297
AGUGUCUAGUCAACUUAAUUGAG
383





uugAfcUfagacascsu








AD-558066.1
gsusucacAfaGfAfGfaagucguuuuL96
212
asAfsaacg(Agn)cuu
298
UAGUUCACAAGAGAAGUCGUUUC
384





cucUfuGfugaacsusa








AD-557353.1
gsascccaAfuUfAfCfugucauugauL96
213
asUfscaau(Ggn)aca
299
GGGACCCAAUUACUGUCAUUGAU
385





guaAfuUfgggucscsc








AD-557782.1
gsasauuuUfaUfGfAfcuaugacguuL96
214
asAfscguc(Agn)uag
300
CUGAAUUUUAUGACUAUGACGUU
386





ucaUfaAfaauucsasg








AD-556390.1
csusggagUfuUfCfAfgcuuggacauL96
215
asUfsgucc(Agn)agc
301
GUCUGGAGUUUCAGCUUGGACAC
387





ugaAfaCfuccagsasc








AD-557078.1
gsusgucuAfgUfCfAfacuuaauuguL96
216
asCfsaauu(Agn)agu
302
AAGUGUCUAGUCAACUUAAUUGA
388





ugaCfuAfgacacsusu








AD-557475.1
ususccaaGfaAfAfGfacaaugagcuL96
217
asGfscuca(Tgn)ugu
303
GCUUCCAAGAAAGACAAUGAGCA
389





cuuUfcUfuggaasgsc











AD-556734.1
csusgcuaUfgAfCfGfguuacacucuL96
218
asGfsagug(Tgn)aac
304
CACUGCUAUGACGGUUACACUCU
390





cguCfaUfagcagsusg








AD-557084.1
asgsucaaCfuUfAfAfuugagaagguL96
219
asCfscuuc(Tgn)caa
305
CUAGUCAACUUAAUUGAGAAGGU
391





uuaAfgUfugacusasg








AD-557498.1
asusguguUfcAfAfAfgucaaggauuL96
220
asAfsuccu(Tgn)gac
306
ACAUGUGUUCAAAGUCAAGGAUA
392





uuuGfaAfcacausgsu








AD-556788.1
gsascagcGfaUfCfUfgugacaacguL96
221
asCfsguug(Tgn)cac
307
CAGACAGCGAUCUGUGACAACGG
393





agaUfcGfcugucsusg








AD-557067.1
gsgsagccAfaAfAfAfgugucuaguuL96
222
asAfscuag(Agn)cac
308
CAGGAGCCAAAAAGUGUCUAGUC
394





uuuUfuGfgcuccsusg








AD-557852.1
gsasgggaAfcAfAfCfucgagcuuuuL96
223
asAfsaagc(Tgn)cga
309
CCGAGGGAACAACUCGAGCUUUG
395





guuGfuUfcccucsgsg








AD-556733.1
ascsugcuAfuGfAfCfgguuacacuuL96
224
asAfsgugu(Agn)acc
310
CCACUGCUAUGACGGUUACACUC
396





gucAfuAfgcagusgsg








AD-556725.1
csuscuuuCfcAfCfUfgcuaugacguL96
225
asCfsguca(Tgn)agc
311
AUCUCUUUCCACUGCUAUGACGG
397





aguGfgAfaagagsasu








AD-557860.1
asascucgAfgCfUfUfugaggcuucuL96
226
asGfsaagc(Cgn)uca
312
ACAACUCGAGCUUUGAGGCUUCC
398





aagCfuCfgaguusgsu








AD-556786.1
csasgacaGfcGfAfUfcugugacaauL96
227
asUfsuguc(Agn)cag
313
GGCAGACAGCGAUCUGUGACAAC
399





aucGfcUfgucugscsc








AD-556581.1
cscsuucuGfgCfUfUfcuacccguauL96
228
asUfsacgg(Ggn)uag
314
GUCCUUCUGGCUUCUACCCGUAC
400





aagCfcAfgaaggsasc








AD-556963.1
ascscauaGfaAfGfGfagucgaugcuL96
229
asGfscauc(Ggn)acu
315
AGACCAUAGAAGGAGUCGAUGCU
401





ccuUfcUfaugguscsu








AD-557450.1
ascscaagUfgAfAfCfaucaaugcuuL96
230
asAfsgcau(Tgn)gau
316
GAACCAAGUGAACAUCAAUGCUU
402





guuCfaCfuuggususc








AD-557204.1
usgsaaauCfaAfUfUfaugaagaccuL96
231
asGfsgucu(Tgn)cau
317
AAUGAAAUCAAUUAUGAAGACCA
403





aauUfgAfuuucasusu








AD-558062.1
gsasuaguUfcAfCfAfagagaagucuL96
232
asGfsacuu(Cgn)ucu
318
UUGAUAGUUCACAAGAGAAGUCG
404





uguGfaAfcuaucsasa








AD-557602.1
asasggguAfcCfGfAfuuaccacaauL96
233
asUfsugug(Ggn)uaa
319
GGAAGGGUACCGAUUACCACAAG
405





ucgGfuAfcccuuscsc








AD-556917.1
csasagacUfcCfUfUfcauguacgauL96
234
asUfscgua(Cgn)aug
320
GCCAAGACUCCUUCAUGUACGAC
406





aagGfaGfucuugsgsc








AD-557969.1
csusgacuCfgGfAfAfggaggucuauL96
235
asUfsagac(Cgn)ucc
321
AGCUGACUCGGAAGGAGGUCUAC
407





uucCfgAfgucagscsu








AD-558064.1
usasguucAfcAfAfGfagaagucguuL96
236
asAfscgac(Tgn)ucu
322
GAUAGUUCACAAGAGAAGUCGUU
408





cuuGfuGfaacuasusc








AD-558105.1
gsuscugcAfaAfAfAfccagaagcguL96
237
asCfsgcuu(Cgn)ugg
323
AUGUCUGCAAAAACCAGAAGCGG
409





uuuUfuGfcagacsasu








AD-556791.1
asgscgauCfuGfUfGfacaacggaguL96
238
asCfsuccg(Tgn)ugu
324
ACAGCGAUCUGUGACAACGGAGC
410





cacAfgAfucgcusgsu








AD-557972.1
ascsucggAfaGfGfAfggucuacauuL96
239
asAfsugua(Ggn)acc
325
UGACUCGGAAGGAGGUCUACAUC
411





uccUfuCfcgaguscsa








AD-556920.1
gsascuccUfuCfAfUfguacgacacuL96
240
asGfsuguc(Ggn)uac
326
AAGACUCCUUCAUGUACGACACC
412





augAfaGfgagucsusu








AD-558078.1
asgsucguUfuCfAfUfucaaguugguL96
241
asCfscaac(Tgn)uga
327
GAAGUCGUUUCAUUCAAGUUGGU
413





augAfaAfcgacususc








AD-557861.1
ascsucgaGfcUfUfUfgaggcuuccuL96
242
asGfsgaag(Cgn)cuc
328
CAACUCGAGCUUUGAGGCUUCCU
414





aaaGfcUfcgagususg








AD-557856.1
gsasacaaCfuCfGfAfgcuuugagguL96
243
asCfscuca(Agn)agc
329
GGGAACAACUCGAGCUUUGAGGC
415





ucgAfgUfuguucscsc








AD-557041.1
usgsgugcUfaGfAfUfggaucagacuL96
244
asGfsucug(Agn)ucc
330
CCUGGUGCUAGAUGGAUCAGACA
416





aucUfaGfcaccasgsg








AD-556874.1
gsusgucaGfgAfAfGfguggcucuuuL96
245
asAfsagag(Cgn)cac
331
ACGUGUCAGGAAGGUGGCUCUUG
417





cuuCfcUfgacacsgsu








AD-557345.1
csusgaugGfaUfUfGfcacaacauguL96
246
asCfsaugu(Tgn)gug
332
GACUGAUGGAUUGCACAACAUGG
418





caaUfcCfaucagsusc








AD-557839.1
csasgacuAfuCfAfGfgcccauuuguL96
247
asCfsaaau(Ggn)ggc
333
GCCAGACUAUCAGGCCCAUUUGU
419





cugAfuAfgucugsgsc








AD-556962.1
gsasccauAfgAfAfGfgagucgauguL96
248
asCfsaucg(Agn)cuc
334
GAGACCAUAGAAGGAGUCGAUGC
420





cuuCfuAfuggucsusc








AD-557066.1
asgsgagcCfaAfAfAfagugucuaguL96
249
asCfsuaga(Cgn)acu
335
ACAGGAGCCAAAAAGUGUCUAGU
421





uuuUfgGfcuccusgsu








AD-556961.1
asgsaccaUfaGfAfAfggagucgauuL96
250
asAfsucga(Cgn)ucc
336
AGAGACCAUAGAAGGAGUCGAUG
422





uucUfaUfggucuscsu








AD-557226.1
asasguugAfaGfUfCfagggacuaauL96
251
asUfsuagu(Cgn)ccu
337
ACAAGUUGAAGUCAGGGACUAAC
423





gacUfuCfaacuusgsu








AD-557865.1
gsasgcuuUfgAfGfGfcuuccuccauL96
252
asUfsggag(Ggn)aag
338
UCGAGCUUUGAGGCUUCCUCCAA
424





ccuCfaAfagcucsgsa








AD-557209.1
uscsaauuAfuGfAfAfgaccacaaguL96
253
asCfsuugu(Ggn)guc
339
AAUCAAUUAUGAAGACCACAAGU
425





uucAfuAfauugasusu








AD-556918.1
asasgacuCfcUfUfCfauguacgacuL96
254
asGfsucgu(Agn)cau
340
CCAAGACUCCUUCAUGUACGACA
426





gaaGfgAfgucuusgsg








AD-557868.1
csusuugaGfgCfUfUfccuccaacuuL96
255
asAfsguug(Ggn)agg
341
AGCUUUGAGGCUUCCUCCAACUA
427





aagCfcUfcaaagscsu








AD-557873.1
asgsgcuuCfcUfCfCfaacuaccacuL96
256
asGfsuggu(Agn)guu
342
UGAGGCUUCCUCCAACUACCACU
428





ggaGfgAfagccuscsa








AD-556787.1
asgsacagCfgAfUfCfugugacaacuL96
257
asGfsuugu(Cgn)aca
343
GCAGACAGCGAUCUGUGACAACG
429





gauCfgCfugucusgsc








AD-558004.1
csasggcuAfuGfAfCfaaagucaaguL96
258
asCfsuuga(Cgn)uuu
344
CCCAGGCUAUGACAAAGUCAAGG
430





gucAfuAfgccugsgsg








AD-557867.1
gscsuuugAfgGfCfUfuccuccaacuL96
259
asGfsuugg(Agn)gga
345
GAGCUUUGAGGCUUCCUCCAACU
431





agcCfuCfaaagcsusc








AD-557788.1
usasugacUfaUfGfAfcguugcccuuL96
260
asAfsgggc(Agn)acg
346
UUUAUGACUAUGACGUUGCCCUG
432





ucaUfaGfucauasasa








AD-558106.1
uscsugcaAfaAfAfCfcagaagcgguL96
261
asCfscgcu(Tgn)cug
347
UGUCUGCAAAAACCAGAAGCGGC
433





guuUfuUfgcagascsa








AD-556724.1
uscsucuuUfcCfAfCfugcuaugacuL96
262
asGfsucau(Agn)gca
348
GAUCUCUUUCCACUGCUAUGACG
434





gugGfaAfagagasusc








AD-556739.1
asusgacgGfuUfAfCfacucuccgguL96
263
asCfscgga(Ggn)agu
349
CUAUGACGGUUACACUCUCCGGG
435





guaAfcCfgucausasg








AD-557417.1
csasagggAfgGfAfUfuaucuggauuL96
264
asAfsucca(Ggn)aua
350
CCCAAGGGAGGAUUAUCUGGAUG
436





aucCfuCfccuugsgsg








AD-556790.1
csasgcgaUfcUfGfUfgacaacggauL96
265
asUfsccgu(Tgn)guc
351
GACAGCGAUCUGUGACAACGGAG
437





acaGfaUfcgcugsusc








AD-557872.1
gsasggcuUfcCfUfCfcaacuaccauL96
266
asUfsggua(Ggn)uug
352
UUGAGGCUUCCUCCAACUACCAC
438





gagGfaAfgccucsasa








AD-556738.1
usasugacGfgUfUfAfcacucuccguL96
267
asCfsggag(Agn)gug
353
GCUAUGACGGUUACACUCUCCGG
439





uaaCfcGfucauasgsc








AD-557857.1
asascaacUfcGfAfGfcuuugaggcuL96
268
asGfsccuc(Agn)aag
354
GGAACAACUCGAGCUUUGAGGCU
440





cucGfaGfuuguuscsc








AD-556726.1
uscsuuucCfaCfUfGfcuaugacgguL96
269
asCfscguc(Agn)uag
355
UCUCUUUCCACUGCUAUGACGGU
441





cagUfgGfaaagasgsa








AD-557495.1
asascaugUfgUfUfCfaaagucaaguL96
270
asCfsuuga(Cgn)uuu
356
GCAACAUGUGUUCAAAGUCAAGG
442





gaaCfaCfauguusgsc








AD-557016.1
cscsuucaGfgCfUfCfcaugaacauuL96
271
asAfsuguu(Cgn)aug
357
ACCCUUCAGGCUCCAUGAACAUC
443





gagCfcUfgaaggsgsu








AD-558074.1
gsasgaagUfcGfUfUfucauucaaguL96
272
asCfsuuga(Agn)uga
358
AAGAGAAGUCGUUUCAUUCAAGU
444





aacGfaCfuucucsusu








AD-557604.1
gsgsguacCfgAfUfUfaccacaagcuL96
273
asGfscuug(Tgn)ggu
359
AAGGGUACCGAUUACCACAAGCA
445





aauCfgGfuacccsusu








AD-557346.1
usgsauggAfuUfGfCfacaacaugguL96
274
asCfscaug(Tgn)ugu
360
ACUGAUGGAUUGCACAACAUGGG
446





gcaAfuCfcaucasgsu








AD-557851.1
csgsagggAfaCfAfAfcucgagcuuuL96
275
asAfsagcu(Cgn)gag
361
ACCGAGGGAACAACUCGAGCUUU
447





uugUfuCfccucgsgsu
















TABLE 4







Unmodified Sense and Antisense Sequences of Complement Factor B dsRNA Agents















SEQ


SEQ




Sense
ID
Range
Antiense
ID
Range


Duplex Name
Sequence 5′ to 3′
NO:
(NM_001710.5)
Sequence 5′ to 3′
NO:
(NM_001710.5)





AD-558860.1
UGCCAAGACUCCUUCAUGUAU
449
930-950
AUACAUGAAGGAGUCUUGGCAGG
520
928-950





AD-560018.1
AAGAGAAGUCGUUUCAUUCAU
450
2397-2417
AUGAAUGAAACGACUUCUCUUGU
521
2395-2417





AD-560019.1
AGAGAAGUCGUUUCAUUCAAU
451
2398-2418
AUUGAAUGAAACGACUUCUCUUG
522
2396-2418





AD-559160.1
UAUGAAGACCACAAGUUGAAU
452
1308-1328
AUUCAACUUGUGGUCUUCAUAAU
523
1306-1328





AD-559921.1
CGGAAGGAGGUCUACAUCAAU
453
2202-2222
AUUGAUGUAGACCUCCUUCCGAG
524
2200-2222





AD-559755.1
AUCAAGCUCAAGAAUAAGCUU
454
2016-2036
AAGCUUAUUCUUGAGCUUGAUCA
525
2014-2036





AD-560017.1
CAAGAGAAGUCGUUUCAUUCU
455
2396-2416
AGAAUGAAACGACUUCUCUUGUG
526
2394-2416





AD-559614.1
CUGUGGUGUCUGAGUACUUUU
456
1819-1839
AAAAGUACUCAGACACCACAGCC
527
1817-1839





AD-559435.1
AUGAGCAACAUGUGUUCAAAU
457
1621-1641
AUUUGAACACAUGUUGCUCAUUG
528
1619-1641





AD-560016.1
ACAAGAGAAGUCGUUUCAUUU
458
2395-2415
AAAUGAAACGACUUCUCUUGUGA
529
2393-2415





AD-559451.1
CAAAGUCAAGGAUAUGGAAAU
459
1637-1657
AUUUCCAUAUCCUUGACUUUGAA
530
1635-1657





AD-559617.1
UGGUGUCUGAGUACUUUGUGU
460
1822-1842
ACACAAAGUACUCAGACACCACA
531
1820-1842





AD-560021.1
AGAAGUCGUUUCAUUCAAGUU
461
2400-2420
AACUUGAAUGAAACGACUUCUCU
532
2398-2420





AD-559449.1
UUCAAAGUCAAGGAUAUGGAU
462
1635-1655
AUCCAUAUCCUUGACUUUGAACA
533
1633-1655





AD-559450.1
UCAAAGUCAAGGAUAUGGAAU
463
1636-1656
AUUCCAUAUCCUUGACUUUGAAC
534
1634-1656





AD-559925.1
AGGAGGUCUACAUCAAGAAUU
464
2206-2226
AAUUCUUGAUGUAGACCUCCUUC
535
2204-2226





AD-559440.1
CAACAUGUGUUCAAAGUCAAU
465
1626-1646
AUUGACUUUGAACACAUGUUGCU
536
1624-1646





AD-559788.1
ACUAUCAGGCCCAUUUGUCUU
466
2049-2069
AAGACAAAUGGGCCUGAUAGUCU
537
2047-2069





AD-559437.1
GAGCAACAUGUGUUCAAAGUU
467
1623-1643
AACUUUGAACACAUGUUGCUCAU
538
1621-1643





AD-559369.1
AGGAUUAUCUGGAUGUCUAUU
468
1537-1557
AAUAGACAUCCAGAUAAUCCUCC
539
1535-1557





AD-559446.1
GUGUUCAAAGUCAAGGAUAUU
469
1632-1652
AAUAUCCUUGACUUUGAACACAU
540
1630-1652





AD-559924.1
AAGGAGGUCUACAUCAAGAAU
470
2205-2225
AUUCUUGAUGUAGACCUCCUUCC
541
2203-2225





AD-558965.1
UCAGGCUCCAUGAACAUCUAU
471
1074-1094
AUAGAUGUUCAUGGAGCCUGAAG
542
1072-1094





AD-560045.1
GUGGAUGUCUGCAAAAACCAU
472
2442-2462
AUGGUUUUUGCAGACAUCCACUA
543
2440-2462





AD-559946.1
GUGAGAGAGAUGCUCAAUAUU
473
2245-2265
AAUAUUGAGCAUCUCUCUCACAG
544
2243-2265





AD-558697.1
AUCGCACCUGCCAAGUGAAUU
474
703-723
AAUUCACUUGGCAGGUGCGAUUG
545
701-723





AD-559008.1
UCACAGGAGCCAAAAAGUGUU
475
1135-1155
AACACUUUUUGGCUCCUGUGAAG
546
1133-1155





AD-559357.1
AAAACCCAAGGGAGGAUUAUU
476
1525-1545
AAUAAUCCUCCCUUGGGUUUUUG
547
1523-1545





AD-559020.1
AAAAGUGUCUAGUCAACUUAU
477
1147-1167
AUAAGUUGACUAGACACUUUUUG
548
1145-1167





AD-559143.1
CAGCUCAAUGAAAUCAAUUAU
478
1290-1310
AUAAUUGAUUUCAUUGAGCUGCU
549
1288-1310





AD-559374.1
UAUCUGGAUGUCUAUGUGUUU
479
1542-1562
AAACACAUAGACAUCCAGAUAAU
550
1540-1562





AD-560161.1
GGCGUGGGAUUGAAUUAAAAU
480
2594-2614
AUUUUAAUUCAAUCCCACGCCCC
551
2592-2614





AD-559947.1
UGAGAGAGAUGCUCAAUAUGU
481
2246-2266
ACAUAUUGAGCAUCUCUCUCACA
552
2244-2266





AD-559616.1
GUGGUGUCUGAGUACUUUGUU
482
1821-1841
AACAAAGUACUCAGACACCACAG
553
1819-1841





AD-559142.1
GCAGCUCAAUGAAAUCAAUUU
483
1289-1309
AAAUUGAUUUCAUUGAGCUGCUU
554
1287-1309





AD-558639.1
CGGUCUCCCUACUACAAUGUU
484
627-647
AACAUUGUAGUAGGGAGACCGGG
555
625-647





AD-560166.1
GGGAUUGAAUUAAAACAGCUU
485
2599-2619
AAGCUGUUUUAAUUCAAUCCCAC
556
2597-2619





AD-559359.1
AACCCAAGGGAGGAUUAUCUU
486
1527-1547
AAGAUAAUCCUCCCUUGGGUUUU
557
1525-1547





AD-558657.1
GUGAGUGAUGAGAUCUCUUUU
487
645-665
AAAAGAGAUCUCAUCACUCACAU
558
643-665





AD-559442.1
ACAUGUGUUCAAAGUCAAGGU
488
1628-1648
ACCUUGACUUUGAACACAUGUUG
559
1626-1648





AD-559023.1
AGUGUCUAGUCAACUUAAUUU
489
1150-1170
AAAUUAAGUUGACUAGACACUUU
560
1148-1170





AD-560160.1
GGGCGUGGGAUUGAAUUAAAU
490
2593-2613
AUUUAAUUCAAUCCCACGCCCCU
561
2591-2613





AD-559398.1
CAAGUGAACAUCAAUGCUUUU
491
1584-1604
AAAAGCAUUGAUGUUCACUUGGU
562
1582-1604





AD-559722.1
AUUCCUGAAUUUUAUGACUAU
492
1983-2003
AUAGUCAUAAAAUUCAGGAAUUC
563
1981-2003





AD-559146.1
CUCAAUGAAAUCAAUUAUGAU
493
1293-1313
AUCAUAAUUGAUUUCAUUGAGCU
564
1291-1313





AD-558267.1
GGAUGUUCCGGGAAAGUGAUU
494
55-75
AAUCACUUUCCCGGAACAUCCAA
565
53-75





AD-559074.1
GAUAUGGUCUAGUGACAUAUU
495
1201-1221
AAUAUGUCACUAGACCAUAUCUU
566
1199-1221





AD-560162.1
GCGUGGGAUUGAAUUAAAACU
496
2595-2615
AGUUUUAAUUCAAUCCCACGCCC
567
2593-2615





AD-559021.1
AAAGUGUCUAGUCAACUUAAU
497
1148-1168
AUUAAGUUGACUAGACACUUUUU
568
1146-1168





AD-559144.1
AGCUCAAUGAAAUCAAUUAUU
498
1291-1311
AAUAAUUGAUUUCAUUGAGCUGC
569
1289-1311





AD-559147.1
CAAUGAAAUCAAUUAUGAAGU
499
1295-1315
ACUUCAUAAUUGAUUUCAUUGAG
570
1293-1315





AD-560164.1
GUGGGAUUGAAUUAAAACAGU
500
2597-2617
ACUGUUUUAAUUCAAUCCCACGC
571
2595-2617





AD-559714.1
AAGCAGGAAUUCCUGAAUUUU
501
1975-1995
AAAAUUCAGGAAUUCCUGCUUCU
572
1973-1995





AD-560165.1
UGGGAUUGAAUUAAAACAGCU
502
2598-2618
AGCUGUUUUAAUUCAAUCCCACG
573
2596-2618





AD-559300.1
ACCCAAUUACUGUCAUUGAUU
503
1468-1488
AAUCAAUGACAGUAAUUGGGUCC
574
1466-1488





AD-559866.1
CCCUGCACAGGAUAUCAAAGU
504
2147-2167
ACUUUGAUAUCCUGUGCAGGGAG
575
2145-2167





AD-559302.1
CCAAUUACUGUCAUUGAUGAU
505
1470-1490
AUCAUCAAUGACAGUAAUUGGGU
576
1468-1490





AD-560163.1
CGUGGGAUUGAAUUAAAACAU
506
2596-2616
AUGUUUUAAUUCAAUCCCACGCC
577
2594-2616





AD-559718.1
AGGAAUUCCUGAAUUUUAUGU
507
1979-1999
ACAUAAAAUUCAGGAAUUCCUGC
578
1977-1999





AD-559721.1
AAUUCCUGAAUUUUAUGACUU
508
1982-2002
AAGUCAUAAAAUUCAGGAAUUCC
579
1980-2002





AD-559026.1
GUCUAGUCAACUUAAUUGAGU
509
1153-1173
ACUCAAUUAAGUUGACUAGACAC
580
1151-1173





AD-559719.1
GGAAUUCCUGAAUUUUAUGAU
510
1980-2000
AUCAUAAAAUUCAGGAAUUCCUG
581
1978-2000





AD-559060.1
UGGUGUGAAGCCAAGAUAUGU
511
1187-1207
ACAUAUCUUGGCUUCACACCAUA
582
1185-1207





AD-559864.1
CUCCCUGCACAGGAUAUCAAU
512
2145-2165
AUUGAUAUCCUGUGCAGGGAGCA
583
2143-2165





AD-559059.1
AUGGUGUGAAGCCAAGAUAUU
513
1186-1206
AAUAUCUUGGCUUCACACCAUAA
584
1184-1206





AD-559865.1
UCCCUGCACAGGAUAUCAAAU
514
2146-2166
AUUUGAUAUCCUGUGCAGGGAGC
585
2144-2166





AD-559148.1
AAUGAAAUCAAUUAUGAAGAU
515
1296-1316
AUCUUCAUAAUUGAUUUCAUUGA
586
1294-1316





AD-559375.1
AUCUGGAUGUCUAUGUGUUUU
516
1543-1563
AAAACACAUAGACAUCCAGAUAA
587
1541-1563





AD-559393.1
UGAACCAAGUGAACAUCAAUU
517
1579-1599
AAUUGAUGUUCACUUGGUUCACC
588
1577-1599





AD-559717.1
CAGGAAUUCCUGAAUUUUAUU
518
1978-1998
AAUAAAAUUCAGGAAUUCCUGCU
589
1976-1998





AD-559392.1
GUGAACCAAGUGAACAUCAAU
519
1578-1598
AUUGAUGUUCACUUGGUUCACCA
590
1576-1598
















TABLE 5







Modified Sense and Antisense Strand Sequences of Complement Factor B dsRNA Agents















SEQ

SEQ

SEQ




ID
Antisense
ID

ID


Duplex Name
Sense Sequence 5′ to 3′
NO:
Sequence 5′ to 3′
NO:
mRNA Target Sequence
NO:





AD-558860.1
usgsccaaGfaCfUfCfcuucauguauL96
591
asUfsacaUfgAfAfgg
662
CCUGCCAAGACUCCUUCAUGUAC
733





agUfcUfuggcasgsg








AD-560018.1
asasgagaAfgUfCfGfuuucauucauL96
592
asUfsgaaUfgAfAfac
663
ACAAGAGAAGUCGUUUCAUUCAA
734





gaCfuUfcucuusgsu








AD-560019.1
asgsagaaGfuCfGfUfuucauucaauL96
593
asUfsugaAfuGfAfaa
664
CAAGAGAAGUCGUUUCAUUCAAG
735





cgAfcUfucucususg








AD-559160.1
usasugaaGfaCfCfAfcaaguugaauL96
594
asUfsucaAfcUfUfgu
665
AUUAUGAAGACCACAAGUUGAAG
736





ggUfcUfucauasasu








AD-559921.1
csgsgaagGfaGfGfUfcuacaucaauL96
595
asUfsugaUfgUfAfga
666
CUCGGAAGGAGGUCUACAUCAAG
737





ccUfcCfuuccgsasg








AD-559755.1
asuscaagCfuCfAfAfgaauaagcuuL96
596
asAfsgcuUfaUfUfcu
667
UGAUCAAGCUCAAGAAUAAGCUG
738





ugAfgCfuugauscsa








AD-560017.1
csasagagAfaGfUfCfguuucauucuL96
597
asGfsaauGfaAfAfcg
668
CACAAGAGAAGUCGUUUCAUUCA
739





acUfuCfucuugsusg








AD-559614.1
csusguggUfgUfCfUfgaguacuuuuL96
598
asAfsaagUfaCfUfca
669
GGCUGUGGUGUCUGAGUACUUUG
740





gaCfaCfcacagscsc








AD-559435.1
asusgagcAfaCfAfUfguguucaaauL96
599
asUfsuugAfaCfAfca
670
CAAUGAGCAACAUGUGUUCAAAG
741





ugUfuGfcucaususg








AD-560016.1
ascsaagaGfaAfGfUfcguuucauuuL96
600
asAfsaugAfaAfCfga
671
UCACAAGAGAAGUCGUUUCAUUC
742





cuUfcUfcuugusgsa








AD-559451.1
csasaaguCfaAfGfGfauauggaaauL96
601
asUfsuucCfaUfAfuc
672
UUCAAAGUCAAGGAUAUGGAAAA
743





cuUfgAfcuuugsasa








AD-559617.1
usgsguguCfuGfAfGfuacuuuguguL96
602
asCfsacaAfaGfUfac
673
UGUGGUGUCUGAGUACUUUGUGC
744





ucAfgAfcaccascsa








AD-560021.1
asgsaaguCfgUfUfUfcauucaaguuL96
603
asAfscuuGfaAfUfga
674
AGAGAAGUCGUUUCAUUCAAGUU
745





aaCfgAfcuucuscsu








AD-559449.1
ususcaaaGfuCfAfAfggauauggauL96
604
asUfsccaUfaUfCfcu
675
UGUUCAAAGUCAAGGAUAUGGAA
746





ugAfcUfuugaascsa








AD-559450.1
uscsaaagUfcAfAfGfgauauggaauL96
605
asUfsuccAfuAfUfcc
676
GUUCAAAGUCAAGGAUAUGGAAA
747





uuGfaCfuuugasasc








AD-559925.1
asgsgaggUfcUfAfCfaucaagaauuL96
606
asAfsuucUfuGfAfug
677
GAAGGAGGUCUACAUCAAGAAUG
748





uaGfaCfcuccususc








AD-559440.1
csasacauGfuGfUfUfcaaagucaauL96
607
asUfsugaCfuUfUfga
678
AGCAACAUGUGUUCAAAGUCAAG
749





acAfcAfuguugscsu








AD-559788.1
ascsuaucAfgGfCfCfcauuugucuuL96
608
asAfsgacAfaAfUfgg
679
AGACUAUCAGGCCCAUUUGUCUC
750





gcCfuGfauaguscsu








AD-559437.1
gsasgcaaCfaUfGfUfguucaaaguuL96
609
asAfscuuUfgAfAfca
680
AUGAGCAACAUGUGUUCAAAGUC
751





caUfgUfugcucsasu








AD-559369.1
asgsgauuAfuCfUfGfgaugucuauuL96
610
asAfsuagAfcAfUfcc
681
GGAGGAUUAUCUGGAUGUCUAUG
752





agAfuAfauccuscsc








AD-559446.1
gsusguucAfaAfGfUfcaaggauauuL96
611
asAfsuauCfcUfUfga
682
AUGUGUUCAAAGUCAAGGAUAUG
753





cuUfuGfaacacsasu








AD-559924.1
asasggagGfuCfUfAfcaucaagaauL96
612
asUfsucuUfgAfUfgu
683
GGAAGGAGGUCUACAUCAAGAAU
754





agAfcCfuccuuscsc








AD-558965.1
uscsaggcUfcCfAfUfgaacaucuauL96
613
asUfsagaUfgUfUfca
684
CUUCAGGCUCCAUGAACAUCUAC
755





ugGfaGfccugasasg








AD-560045.1
gsusggauGfuCfUfGfcaaaaaccauL96
614
asUfsgguUfuUfUfgc
685
UAGUGGAUGUCUGCAAAAACCAG
756





agAfcAfuccacsusa








AD-559946.1
gsusgagaGfaGfAfUfgcucaauauuL96
615
asAfsuauUfgAfGfca
686
CUGUGAGAGAGAUGCUCAAUAUG
757





ucUfcUfcucacsasg








AD-558697.1
asuscgcaCfcUfGfCfcaagugaauuL96
616
asAfsuucAfcUfUfgg
687
CAAUCGCACCUGCCAAGUGAAUG
758





caGfgUfgcgaususg








AD-559008.1
uscsacagGfaGfCfCfaaaaaguguuL96
617
asAfscacUfuUfUfug
688
CUUCACAGGAGCCAAAAAGUGUC
759





gcUfcCfugugasasg








AD-559357.1
asasaaccCfaAfGfGfgaggauuauuL96
618
asAfsuaaUfcCfUfcc
689
CAAAAACCCAAGGGAGGAUUAUC
760





cuUfgGfguuuususg








AD-559020.1
asasaaguGfuCfUfAfgucaacuuauL96
619
asUfsaagUfuGfAfcu
690
CAAAAAGUGUCUAGUCAACUUAA
761





agAfcAfcuuuususg








AD-559143.1
csasgcucAfaUfGfAfaaucaauuauL96
620
asUfsaauUfgAfUfuu
691
AGCAGCUCAAUGAAAUCAAUUAU
762





caUfuGfagcugscsu








AD-559374.1
usasucugGfaUfGfUfcuauguguuuL96
621
asAfsacaCfaUfAfga
692
AUUAUCUGGAUGUCUAUGUGUUU
763





caUfcCfagauasasu








AD-560161.1
gsgscgugGfgAfUfUfgaauuaaaauL96
622
asUfsuuuAfaUfUfca
693
GGGGCGUGGGAUUGAAUUAAAAC
764





auCfcCfacgccscsc








AD-559947.1
usgsagagAfgAfUfGfcucaauauguL96
623
asCfsauaUfuGfAfgc
694
UGUGAGAGAGAUGCUCAAUAUGC
765





auCfuCfucucascsa








AD-559616.1
gsusggugUfcUfGfAfguacuuuguuL96
624
asAfscaaAfgUfAfcu
695
CUGUGGUGUCUGAGUACUUUGUG
766





caGfaCfaccacsasg








AD-559142.1
gscsagcuCfaAfUfGfaaaucaauuuL96
625
asAfsauuGfaUfUfuc
696
AAGCAGCUCAAUGAAAUCAAUUA
767





auUfgAfgcugcsusu








AD-558639.1
csgsgucuCfcCfUfAfcuacaauguuL96
626
asAfscauUfgUfAfgu
697
CCCGGUCUCCCUACUACAAUGUG
768





agGfgAfgaccgsgsg








AD-560166.1
gsgsgauuGfaAfUfUfaaaacagcuuL96
627
asAfsgcuGfuUfUfua
698
GUGGGAUUGAAUUAAAACAGCUG
769





auUfcAfaucccsasc








AD-559359.1
asascccaAfgGfGfAfggauuaucuuL96
628
asAfsgauAfaUfCfcu
699
AAAACCCAAGGGAGGAUUAUCUG
770





ccCfuUfggguususu








AD-558657.1
gsusgaguGfaUfGfAfgaucucuuuuL96
629
asAfsaagAfgAfUfcu
700
AUGUGAGUGAUGAGAUCUCUUUC
771





caUfcAfcucacsasu








AD-559442.1
ascsauguGfuUfCfAfaagucaagguL96
630
asCfscuuGfaCfUfuu
701
CAACAUGUGUUCAAAGUCAAGGA
772





gaAfcAfcaugususg








AD-559023.1
asgsugucUfaGfUfCfaacuuaauuuL96
631
asAfsauuAfaGfUfug
702
AAAGUGUCUAGUCAACUUAAUUG
773





acUfaGfacacususu








AD-560160.1
gsgsgcguGfgGfAfUfugaauuaaauL96
632
asUfsuuaAfuUfCfaa
703
AGGGGCGUGGGAUUGAAUUAAAA
774





ucCfcAfcgcccscsu








AD-559398.1
csasagugAfaCfAfUfcaaugcuuuuL96
633
asAfsaagCfaUfUfga
704
ACCAAGUGAACAUCAAUGCUUUG
775





ugUfuCfacuugsgsu








AD-559722.1
asusuccuGfaAfUfUfuuaugacuauL96
634
asUfsaguCfaUfAfaa
705
GAAUUCCUGAAUUUUAUGACUAU
776





auUfcAfggaaususc








AD-559146.1
csuscaauGfaAfAfUfcaauuaugauL96
635
asUfscauAfaUfUfga
706
AGCUCAAUGAAAUCAAUUAUGAA
777





uuUfcAfuugagscsu








AD-558267.1
gsgsauguUfcCfGfGfgaaagugauuL96
636
asAfsucaCfuUfUfcc
707
UUGGAUGUUCCGGGAAAGUGAUG
778





cgGfaAfcauccsasa








AD-559074.1
gsasuaugGfuCfUfAfgugacauauuL96
637
asAfsuauGfuCfAfcu
708
AAGAUAUGGUCUAGUGACAUAUG
779





agAfcCfauaucsusu








AD-560162.1
gscsguggGfaUfUfGfaauuaaaacuL96
638
asGfsuuuUfaAfUfuc
709
GGGCGUGGGAUUGAAUUAAAACA
780





aaUfcCfcacgcscsc








AD-559021.1
asasagugUfcUfAfGfucaacuuaauL96
639
asUfsuaaGfuUfGfac
710
AAAAAGUGUCUAGUCAACUUAAU
781





uaGfaCfacuuususu








AD-559144.1
asgscucaAfuGfAfAfaucaauuauuL96
640
asAfsuaaUfuGfAfuu
711
GCAGCUCAAUGAAAUCAAUUAUG
782





ucAfuUfgagcusgsc








AD-559147.1
csasaugaAfaUfCfAfauuaugaaguL96
641
asCfsuucAfuAfAfuu
712
CUCAAUGAAAUCAAUUAUGAAGA
783





gaUfuUfcauugsasg








AD-560164.1
gsusgggaUfuGfAfAfuuaaaacaguL96
642
asCfsuguUfuUfAfau
713
GCGUGGGAUUGAAUUAAAACAGC
784





ucAfaUfcccacsgsc








AD-559714.1
asasgcagGfaAfUfUfccugaauuuuL96
643
asAfsaauUfcAfGfga
714
AGAAGCAGGAAUUCCUGAAUUUU
785





auUfcCfugcuuscsu








AD-560165.1
usgsggauUfgAfAfUfuaaaacagcuL96
644
asGfscugUfuUfUfaa
715
CGUGGGAUUGAAUUAAAACAGCU
786





uuCfaAfucccascsg








AD-559300.1
ascsccaaUfuAfCfUfgucauugauuL96
645
asAfsucaAfuGfAfca
716
GGACCCAAUUACUGUCAUUGAUG
787





guAfaUfuggguscsc








AD-559866.1
cscscugcAfcAfGfGfauaucaaaguL96
646
asCfsuuuGfaUfAfuc
717
CUCCCUGCACAGGAUAUCAAAGC
788





cuGfuGfcagggsasg








AD-559302.1
cscsaauuAfcUfGfUfcauugaugauL96
647
asUfscauCfaAfUfga
718
ACCCAAUUACUGUCAUUGAUGAG
789





caGfuAfauuggsgsu








AD-560163.1
csgsugggAfuUfGfAfauuaaaacauL96
648
asUfsguuUfuAfAfuu
719
GGCGUGGGAUUGAAUUAAAACAG
790





caAfuCfccacgscsc








AD-559718.1
asgsgaauUfcCfUfGfaauuuuauguL96
649
asCfsauaAfaAfUfuc
720
GCAGGAAUUCCUGAAUUUUAUGA
791





agGfaAfuuccusgsc








AD-559721.1
asasuuccUfgAfAfUfuuuaugacuuL96
650
asAfsgucAfuAfAfaa
721
GGAAUUCCUGAAUUUUAUGACUA
792





uuCfaGfgaauuscsc








AD-559026.1
gsuscuagUfcAfAfCfuuaauugaguL96
651
asCfsucaAfuUfAfag
722
GUGUCUAGUCAACUUAAUUGAGA
793





uuGfaCfuagacsasc








AD-559719.1
gsgsaauuCfcUfGfAfauuuuaugauL96
652
asUfscauAfaAfAfuu
723
CAGGAAUUCCUGAAUUUUAUGAC
794





caGfgAfauuccsusg








AD-559060.1
usgsguguGfaAfGfCfcaagauauguL96
653
asCfsauaUfcUfUfgg
724
UAUGGUGUGAAGCCAAGAUAUGG
795





cuUfcAfcaccasusa








AD-559864.1
csuscccuGfcAfCfAfggauaucaauL96
654
asUfsugaUfaUfCfcu
725
UGCUCCCUGCACAGGAUAUCAAA
796





guGfcAfgggagscsa








AD-559059.1
asusggugUfgAfAfGfccaagauauuL96
655
asAfsuauCfuUfGfgc
726
UUAUGGUGUGAAGCCAAGAUAUG
797





uuCfaCfaccausasa








AD-559865.1
uscsccugCfaCfAfGfgauaucaaauL96
656
asUfsuugAfuAfUfcc
727
GCUCCCUGCACAGGAUAUCAAAG
798





ugUfgCfagggasgsc








AD-559148.1
asasugaaAfuCfAfAfuuaugaagauL96
657
asUfscuuCfaUfAfau
728
UCAAUGAAAUCAAUUAUGAAGAC
799





ugAfuUfucauusgsa








AD-559375.1
asuscuggAfuGfUfCfuauguguuuuL96
658
asAfsaacAfcAfUfag
729
UUAUCUGGAUGUCUAUGUGUUUG
800





acAfuCfcagausasa








AD-559393.1
usgsaaccAfaGfUfGfaacaucaauuL96
659
asAfsuugAfuGfUfuc
730
GGUGAACCAAGUGAACAUCAAUG
801





acUfuGfguucascsc








AD-559717.1
csasggaaUfuCfCfUfgaauuuuauuL96
660
asAfsuaaAfaUfUfca
731
AGCAGGAAUUCCUGAAUUUUAUG
802





ggAfaUfuccugscsu








AD-559392.1
gsusgaacCfaAfGfUfgaacaucaauL96
661
asUfsugaUfgUfUfca
732
UGGUGAACCAAGUGAACAUCAAU
803





cuUfgGfuucacscsa
















TABLE 6







Unmodified Sense and Antisense Strand Sequences of Complement Factor B dsRNA Agents















SEQ


SEQ



Duplex
Sense
ID

Antisense
ID
Antisense


Name
Sequence 5′ to 3′
NO:
Sense Source Name
Sequence 5′ to 3′
NO:
Source Name





AD-
UGCCAAGAUUCCUUCAUG
804
NM_008198.2_1051-
AUACAUGAAGGAAUCUUGG
849
NM_008198.2_1049-


560969.1
UAU

1071_s
CAGG

1071_as





AD-
AUGUGUUUAAAGUCAAGG
805
NM_008198.2_1751-
AAUCCUTGACUUUAAACACA
850
NM_008198.2_1749-


561537.1
AUU

1771_A21U_s
UGA

1771_U1A_as





AD-
AAAGAUGAGGAUUUGGGU
806
NM_008198.2_2668-
AAAACCCAAAUCCUCAUCUU
851
NM_008198.2_2666-


562262.1
UUU

2688_s
UGA

2688_as





AD-
AAGGAUGUCAAAGCUCUG
807
NM_008198.2_2275-
AAACAGAGCUUUGACAUCC
852
NM_008198.2_2273-


561960.1
UUU

2295_s
UUCA

2295_as





AD-
CUACCAAAUGAUUGAUGA
808
NM_008198.2_1794-
AUUUCATCAAUCAUUUGGU
853
NM_008198.2_1792-


561580.1
AAU

1814_C21U_s
AGAA

1814_G1A_as





AD-
AUCAGUUAUGAAGACCAC
809
NM_008198.2_1423-
AUUGUGGUCUUCAUAACUG
854
NM_008198.2_1421-


561254.1
AAU

1443_G21U_s
AUUU

1443_C1A_as





AD-
UUCUACCAAAUGAUUGAU
810
NM_008198.2_1792-
AUCAUCAAUCAUUUGGUAG
855
NM_008198.2_1790-


561578.1
GAU

1812_A21U_s
AAAA

1812_U1A_as





AD-
UGUGUUUAAAGUCAAGGA
811
NM_008198.2_1752-
AUAUCCTUGACUUUAAACAC
856
NM_008198.2_1750-


561538.1
UAU

1772_s
AUG

1772_as





AD-
AAAUCAGUUAUGAAGACC
812
NM_008198.2_1421-
AGUGGUCUUCAUAACUGAU
857
NM_008198.2_1419-


561252.1
ACU

1441_A21U_s
UUGG

1441_U1A_as





AD-
GAUGUCAAAGCUCUGUUU
813
NM_008198.2_2278-
AACAAACAGAGCUUUGACA
858
NM_008198.2_2276-


561963.1
GUU

2298_A21U_s
UCCU

2298_U1A_as





AD-
CCAGUUGUGAGAGAGAUG
814
NM_008198.2_2360-
AAGCAUCUCUCUCACAACUG
859
NM_008198.2_2358-


562027.1
CUU

2380_A21U_s
GCU

2380_U1A_as





AD-
AGCCAAGAUCUCAGUCAC
815
NM_008198.2_1887-
AGAGUGACUGAGAUCUUGG
860
NM_008198.2_1885-


561653.1
UCU

1907_G21U_s
CUUG

1907_C1A_as





AD-
CGCUUCAUUCAAGUUGGU
816
NM_008198.2_2527-
AACACCAACUUGAAUGAAG
861
NM_008198.2_2525-


562137.1
GUU

2547_G21U_s
CGGC

2547_C1A_as





AD-
CAUGUGUUUAAAGUCAAG
817
NM_008198.2_1750-
AUCCUUGACUUUAAACACA
862
NM_008198.2_1748-


561536.1
GAU

1770_s
UGAU

1770_as





AD-
AAUCAGUUAUGAAGACCA
818
NM_008198.2_1422-
AUGUGGTCUUCAUAACUGA
863
NM_008198.2_1420-


561253.1
CAU

1442 A21U_s
UUUG

1442_U1A_as





AD-
GAAGGAUGUCAAAGCUCU
819
NM_008198.2_2274-
AACAGAGCUUUGACAUCCU
864
NM_008198.2_2272-


561959.1
GUU

2294_s
UCAC

2294_as





AD-
AUGUUUUCUACCAAAUGA
820
NM_008198.2_1787-
AAAUCATUUGGUAGAAAAC
865
NM_008198.2_1785-


561573.1
UUU

1807_G21U_s
AUUC

1807_C1A_as





AD-
CAAGCCAAGAUCUCAGUC
821
NM_008198.2_1885-
AGUGACTGAGAUCUUGGCU
866
NM_008198.2_1883-


561651.1
ACU

1905_s
UGCC

1905_as





AD-
ACCAACUUGAUUGAGAAG
822
NM_008198.2_1279-
AACCUUCUCAAUCAAGUUG
867
NM_008198.2_1277-


561148.1
GUU

1299_G21U_s
GUGA

1299_C1A_as





AD-
GGAUGUCAAAGCUCUGUU
823
NM_008198.2_2277-
ACAAACAGAGCUUUGACAU
868
NM_008198.2_2275-


561962.1
UGU

2297_s
CCUU

2297_as





AD-
AGAUGAGGAUUUGGGUUU
824
NM_001710.5_2549-
AGAAAACCCAAAUCCUCAUC
869
NM_008198.2_2668-


562237.1
UCU

2569_s
UUU

2690_as





AD-
GCCAAGAUCUCAGUCACU
825
NM_008198.2_1888-
ACGAGUGACUGAGAUCUUG
870
NM_008198.2_1886-


561654.1
CGU

1908_C21U_s
GCUU

1908_G1A_as





AD-
CCGCUUCAUUCAAGUUGG
826
NM_008198.2_2526-
ACACCAACUUGAAUGAAGC
871
NM_008198.2_2524-


562136.1
UGU

2546_s
GGCU

2546_as





AD-
GCCAGUUGUGAGAGAGAU
827
NM_008198.2_2359-
AGCAUCTCUCUCACAACUGG
872
NM_008198.2_2357-


562026.1
GCU

2379_s
CUU

2379_as





AD-
UCCAUGAAUAUCUACCUG
828
NM_008198.2_1201-
AACCAGGUAGAUAUUCAUG
873
NM_008198.2_1199-


561101.1
GUU

1221_G21U_s
GAGC

1221_C1A_as





AD-
GUGUUUAAAGUCAAGGAU
829
NM_008198.2_1753-
AAUAUCCUUGACUUUAAAC
874
NM_008198.2_1751-


561539.1
AUU

1773_G21U_s
ACAU

1773_C1A_as





AD-
AGGAUGUCAAAGCUCUGU
830
NM_008198.2_2276-
AAAACAGAGCUUUGACAUC
875
NM_008198.2_2274-


561961.1
UUU

2296_G21U_s
CUUC

2296_C1A_as





AD-
UCAUGUGUUUAAAGUCAA
831
NM_008198.2_1749-
ACCUUGACUUUAAACACAU
876
NM_008198.2_1747-


561535.1
GGU

1769_A21U_s
GAUG

1769_U1A_as





AD-
AAGCCAAGAUCUCAGUCA
832
NM_008198.2_1886-
AAGUGACUGAGAUCUUGGC
877
NM_008198.2_1884-


561652.1
CUU

1906_C21U_s
UUGC

1906_G1A_as





AD-
UCAAAGAUGAGGAUUUGG
833
NM_008198.2_2666-
AACCCAAAUCCUCAUCUUUG
878
NM_008198.2_2664-


562260.1
GUU

2686_s
AGC

2686_as





AD-
UGGUGCUAGAUGGAUCAG
 72
NM_001710.5_1096-
AGUCUGAUCCAUCUAGCACC
158
NM_001710.5_1094-


557041.2
ACU

1116_A21U_s
AGG

1116_U1A_as





AD-
CCAAGAUCUCAGUCACUC
834
NM_008198.2_1889-
AGCGAGTGACUGAGAUCUU
879
NM_008198.2_1887-


561655.1
GCU

1909_C21U_s
GGCU

1909_G1A_as





AD-
GUGCUAGAUGGAUCAGAC
835
NM_001710.5_1098-
ACUGUCTGAUCCAUCUAGCA
880
NM_001710.5_1096-


557043.1
AGU

1118_C21U_s
CCA

1118_G1A_as





AD-
CAAAUCUCUGAGUCUCUG
836
NM_008198.2_1815-
ACACAGAGACUCAGAGAUU
881
NM_008198.2_1813-


561603.1
UGU

1835_G21U_s
UGGU

1835_C1A_as





AD-
UGCUAGAUGGAUCAGACA
837
NM_001710.5_1099-
AGCUGUCUGAUCCAUCUAGC
882
NM_001710.5_1097-


557044.1
GCU

1119_A21U_s
ACC

1119_U1A_as





AD-
GACCACAAGCUGAAGUCA
838
NM_008198.2_1435-
ACCUGACUUCAGCUUGUGG
883
NM_008198.2_1433-


561266.1
GGU

1455_G21U_s
UCUU

1455_C1A_as





AD-
GUUUAAAGUCAAGGAUAU
839
NM_008198.2_1755-
ACCAUATCCUUGACUUUAAA
884
NM_008198.2_1753-


561541.1
GGU

1775_A21U_s
CAC

1775_U1A_as





AD-
AGCUCAAAGAUGAGGAUU
840
NM_008198.2_2663-
ACAAAUCCUCAUCUUUGAGC
885
NM_008198.2_2661-


562257.1
UGU

2683_G21U_s
UUG

2683_C1A_as





AD-
GAGGGAGUAGAGAUCAAA
841
NM_008198.2_517-
ACCUUUGAUCUCUACUCCCU
886
NM_008198.2_515-


560538.1
GGU

537_C21U_s
CCA

537_G1A_as





AD-
UCUACCAAAUGAUUGAUG
842
NM_008198.2_1793-
AUUCAUCAAUCAUUUGGUA
887
NM_008198.2_1791-


561579.1
AAU

1813_A21U_s
GAAA

1813_U1A_as





AD-
CUCAAAGAUGAGGAUUUG
843
NM_008198.2_2665-
ACCCAAAUCCUCAUCUUUGA
888
NM_008198.2_2663-


562259.1
GGU

2685_s
GCU

2685_as





AD-
CUGCCAAGAUUCCUUCAU
844
NM_008198.2_1050-
AACAUGAAGGAAUCUUGGC
889
NM_008198.2_1048-


560968.1
GUU

1070_A21U_s
AGGA

1070_U1A_as





AD-
AUGGCAAGCCAAGAUCUC
845
NM_008198.2_1881-
ACUGAGAUCUUGGCUUGCC
890
NM_008198.2_1879-


561647.1
AGU

1901_s
AUGG

1901_as





AD-
CCUGGAUGUGUAUGUGUU
846
NM_008198.2_1665-
ACAAACACAUACACAUCCAG
891
NM_008198.2_1663-


561469.1
UGU

1685_G21U_s
GUA

1685_C1A_as





AD-
UGUUUAAAGUCAAGGAUA
847
NM_008198.2_1754-
ACAUAUCCUUGACUUUAAA
892
NM_008198.2_1752-


561540.1
UGU

1774_G21U_s
CACA

1774_C1A_as





AD-
GCCGCUUCAUUCAAGUUG
848
NM_008198.2_2525-
AACCAACUUGAAUGAAGCG
893
NM_008198.2_2523-


562135.1
GUU

2545_G21U_s
GCUU

2545_C1A_as
















TABLE 7







Modified Sense and Antisense Strand Sequences of Complement Factor B dsRNA Agents















SEQ

SEQ

SEQ




ID
Antisense
ID

ID


Duplex Name
Sense Sequence 5′ to 3′
NO:
Sequence 5′ to 3′
NO:
mRNA Target Sequence
NO:





AD-560969.1
usgsccaaGfaUfUfCfcuucauguauL96
894
asUfsacau(Ggn)aag
939
CCUGCCAAGAUUCCUUCAUGUAU
 984





gaaUfcUfuggcasgsg








AD-561537.1
asusguguUfuAfAfAfgucaaggauuL96
895
asAfsuccu(Tgn)gac
940
UCAUGUGUUUAAAGUCAAGGAUA
 985





uuuAfaAfcacausgsa








AD-562262.1
asasagauGfaGfGfAfuuuggguuuuL96
896
asAfsaacc(Cgn)aaa
941
UCAAAGAUGAGGAUUUGGGUUUU
 986





uccUfcAfucuuusgsa








AD-561960.1
asasggauGfuCfAfAfagcucuguuuL96
897
asAfsacag(Agn)gcu
942
UGAAGGAUGUCAAAGCUCUGUUU
 987





uugAfcAfuccuuscsa








AD-561580.1
csusaccaAfaUfGfAfuugaugaaauL96
898
asUfsuuca(Tgn)caa
943
UUCUACCAAAUGAUUGAUGAAAC
 988





ucaUfuUfgguagsasa








AD-561254.1
asuscaguUfaUfGfAfagaccacaauL96
899
asUfsugug(Ggn)ucu
944
AAAUCAGUUAUGAAGACCACAAG
 989





ucaUfaAfcugaususu








AD-561578.1
ususcuacCfaAfAfUfgauugaugauL96
900
asUfscauc(Agn)auc
945
UUUUCUACCAAAUGAUUGAUGAA
 990





auuUfgGfuagaasasa








AD-561538.1
usgsuguuUfaAfAfGfucaaggauauL96
901
asUfsaucc(Tgn)uga
946
CAUGUGUUUAAAGUCAAGGAUAU
 991





cuuUfaAfacacasusg








AD-561252.1
asasaucaGfuUfAfUfgaagaccacuL96
902
asGfsuggu(Cgn)uuc
947
CCAAAUCAGUUAUGAAGACCACA
 992





auaAfcUfgauuusgsg








AD-561963.1
gsasugucAfaAfGfCfucuguuuguuL96
903
asAfscaaa(Cgn)aga
948
AGGAUGUCAAAGCUCUGUUUGUA
 993





gcuUfuGfacaucscsu








AD-562027.1
cscsaguuGfuGfAfGfagagaugcuuL96
904
asAfsgcau(Cgn)ucu
949
AGCCAGUUGUGAGAGAGAUGCUA
 994





cucAfcAfacuggscsu








AD-561653.1
asgsccaaGfaUfCfUfcagucacucuL96
905
asGfsagug(Agn)cug
950
CAAGCCAAGAUCUCAGUCACUCG
 995





agaUfcUfuggcususg








AD-562137.1
csgscuucAfuUfCfAfaguugguguuL96
906
asAfscacc(Agn)acu
951
GCCGCUUCAUUCAAGUUGGUGUG
 996





ugaAfuGfaagcgsgsc








AD-561536.1
csasugugUfuUfAfAfagucaaggauL96
907
asUfsccuu(Ggn)acu
952
AUCAUGUGUUUAAAGUCAAGGAU
 997





uuaAfaCfacaugsasu








AD-561253.1
asasucagUfuAfUfGfaagaccacauL96
908
asUfsgugg(Tgn)cuu
953
CAAAUCAGUUAUGAAGACCACAA
 998





cauAfaCfugauususg








AD-561959.1
gsasaggaUfgUfCfAfaagcucuguuL96
909
asAfscaga(Ggn)cuu
954
GUGAAGGAUGUCAAAGCUCUGUU
 999





ugaCfaUfccuucsasc








AD-561573.1
asusguuuUfcUfAfCfcaaaugauuuL96
910
asAfsauca(Tgn)uug
955
GAAUGUUUUCUACCAAAUGAUUG
1000





guaGfaAfaacaususc








AD-561651.1
csasagccAfaGfAfUfcucagucacuL96
911
asGfsugac(Tgn)gag
956
GGCAAGCCAAGAUCUCAGUCACU
1001





aucUfuGfgcuugscsc








AD-561148.1
ascscaacUfuGfAfUfugagaagguuL96
912
asAfsccuu(Cgn)uca
957
UCACCAACUUGAUUGAGAAGGUG
1002





aucAfaGfuuggusgsa








AD-561962.1
gsgsauguCfaAfAfGfcucuguuuguL96
913
asCfsaaac(Agn)gag
958
AAGGAUGUCAAAGCUCUGUUUGU
1003





cuuUfgAfcauccsusu








AD-562237.1
asgsaugaGfgAfUfUfuggguuuucuL96
914
asGfsaaaa(Cgn)cca
959
AAGAUGAGGAUUUGGGUUUUCU
1004





aauCfcUfcaucususu








AD-561654.1
gscscaagAfuCfUfCfagucacucguL96
915
asCfsgagu(Ggn)acu
960
AAGCCAAGAUCUCAGUCACUCGC
1005





gagAfuCfuuggcsusu








AD-562136.1
cscsgcuuCfaUfUfCfaaguugguguL96
916
asCfsacca(Agn)cuu
961
AGCCGCUUCAUUCAAGUUGGUGU
1006





gaaUfgAfagcggscsu








AD-562026.1
gscscaguUfgUfGfAfgagagaugcuL96
917
asGfscauc(Tgn)cuc
962
AAGCCAGUUGUGAGAGAGAUGCU
1007





ucaCfaAfcuggcsusu








AD-561101.1
uscscaugAfaUfAfUfcuaccugguuL96
918
asAfsccag(Ggn)uag
963
GCUCCAUGAAUAUCUACCUGGUG
1008





auaUfuCfauggasgsc








AD-561539.1
gsusguuuAfaAfGfUfcaaggauauuL96
919
asAfsuauc(Cgn)uug
964
AUGUGUUUAAAGUCAAGGAUAUG
1009





acuUfuAfaacacsasu








AD-561961.1
asgsgaugUfcAfAfAfgcucuguuuuL96
920
asAfsaaca(Ggn)agc
965
GAAGGAUGUCAAAGCUCUGUUUG
1010





uuuGfaCfauccususc








AD-561535.1
uscsauguGfuUfUfAfaagucaagguL96
921
asCfscuug(Agn)cuu
966
CAUCAUGUGUUUAAAGUCAAGGA
1011





uaaAfcAfcaugasusg








AD-561652.1
asasgccaAfgAfUfCfucagucacuuL96
922
asAfsguga(Cgn)uga
967
GCAAGCCAAGAUCUCAGUCACUC
1012





gauCfuUfggcuusgsc








AD-562260.1
uscsaaagAfuGfAfGfgauuuggguuL96
923
asAfsccca(Agn)auc
968
GCUCAAAGAUGAGGAUUUGGGUU
1013





cucAfuCfuuugasgsc








AD-557041.2
usgsgugcUfaGfAfUfggaucagacuL96
244
asGfsucug(Agn)ucc
330
CCUGGUGCUAGAUGGAUCAGACA
416





aucUfaGfcaccasgsg








AD-561655.1
cscsaagaUfcUfCfAfgucacucgcuL96
924
asGfscgag(Tgn)gac
969
AGCCAAGAUCUCAGUCACUCGCC
1014





ugaGfaUfcuuggscsu








AD-557043.1
gsusgcuaGfaUfGfGfaucagacaguL96
925
asCfsuguc(Tgn)gau
970
UGGUGCUAGAUGGAUCAGACAGC
1015





ccaUfcUfagcacscsa








AD-561603.1
csasaaucUfcUfGfAfgucucuguguL96
926
asCfsacag(Agn)gac
971
ACCAAAUCUCUGAGUCUCUGUGG
1016





ucaGfaGfauuugsgsu








AD-557044.1
usgscuagAfuGfGfAfucagacagcuL96
927
asGfscugu(Cgn)uga
972
GGUGCUAGAUGGAUCAGACAGCA
1017





uccAfuCfuagcascsc








AD-561266.1
gsasccacAfaGfCfUfgaagucagguL96
928
asCfscuga(Cgn)uuc
973
AAGACCACAAGCUGAAGUCAGGG
1018





agcUfuGfuggucsusu








AD-561541.1
gsusuuaaAfgUfCfAfaggauaugguL96
929
asCfscaua(Tgn)ccu
974
GUGUUUAAAGUCAAGGAUAUGGA
1019





ugaCfuUfuaaacsasc








AD-562257.1
asgscucaAfaGfAfUfgaggauuuguL96
930
asCfsaaau(Cgn)cuc
975
CAAGCUCAAAGAUGAGGAUUUGG
1020





aucUfuUfgagcususg








AD-560538.1
gsasgggaGfuAfGfAfgaucaaagguL96
931
asCfscuuu(Ggn)auc
976
UGGAGGGAGUAGAGAUCAAAGGC
1021





ucuAfcUfcccucscsa








AD-561579.1
uscsuaccAfaAfUfGfauugaugaauL96
932
asUfsucau(Cgn)aau
977
UUUCUACCAAAUGAUUGAUGAAA
1022





cauUfuGfguagasasa








AD-562259.1
csuscaaaGfaUfGfAfggauuuggguL96
933
asCfsccaa(Agn)ucc
978
AGCUCAAAGAUGAGGAUUUGGGU
1023





ucaUfcUfuugagscsu








AD-560968.1
csusgccaAfgAfUfUfccuucauguuL96
934
asAfscaug(Agn)agg
979
UCCUGCCAAGAUUCCUUCAUGUA
1024





aauCfuUfggcagsgsa








AD-561647.1
asusggcaAfgCfCfAfagaucucaguL96
935
asCfsugag(Agn)ucu
980
CCAUGGCAAGCCAAGAUCUCAGU
1025





uggCfuUfgccausgsg








AD-561469.1
cscsuggaUfgUfGfUfauguguuuguL96
936
asCfsaaac(Agn)cau
981
UACCUGGAUGUGUAUGUGUUUGG
1026





acaCfaUfccaggsusa








AD-561540.1
usgsuuuaAfaGfUfCfaaggauauguL96
937
asCfsauau(Cgn)cuu
982
UGUGUUUAAAGUCAAGGAUAUGG
1027





gacUfuUfaaacascsa








AD-562135.1
gscscgcuUfcAfUfUfcaaguugguuL96
938
asAfsccaa(Cgn)uug
983
AAGCCGCUUCAUUCAAGUUGGUG
1028





aauGfaAfgcggcsusu
















TABLE 8







Complement Factor B In Vitro Single Dose Screens


in Hep3B cells













10 nM
1 nM
0.1 nM




% of
% of
% of




Message
Message
Message



DuplexID
Remaining
Remaining
Remaining
















AD-557072.1
4.99
8.87
40.99



AD-558097.1
5.00
11.75
45.33



AD-557068.1
9.00
22.78
53.22



AD-557774.1
6.55
17.23
52.48



AD-557070.1
7.33
21.06
54.78



AD-558225.1
19.10
29.43
106.87



AD-558065.1
12.21
24.28
61.24



AD-557853.1
9.39
35.28
55.01



AD-556919.1
20.01
27.64
138.66



AD-557859.1
13.46
25.92
104.56



AD-557069.1
11.26
29.35
54.36



AD-558068.1
14.59
21.63
48.67



AD-557422.1
14.87
27.15
55.45



AD-558096.1
10.81
26.65
76.39



AD-557084.1
28.07
54.88
138.96



AD-558076.1
13.06
24.15
42.77



AD-558063.1
20.24
28.87
54.49



AD-558069.1
17.70
27.78
53.41



AD-558061.1
19.59
36.04
48.88



AD-558066.1
23.98
44.12
51.72



AD-556581.1
22.46
67.53
186.97



AD-557079.1
9.97
41.07
71.40



AD-558012.1
17.26
35.39
112.72



AD-556701.1
9.91
23.66
62.85



AD-557782.1
13.23
45.66
53.87



AD-557498.1
28.16
56.57
131.91



AD-556788.1
33.88
58.98
118.02



AD-557078.1
18.18
50.25
80.91



AD-557852.1
40.94
60.36
167.53



AD-557353.1
23.37
44.41
62.52



AD-557041.1
26.00
87.78
110.85



AD-556786.1
51.74
66.59
90.96



AD-556734.1
29.10
54.88
70.13



AD-557475.1
26.95
50.54
65.87



AD-557972.1
63.07
77.45
71.99



AD-556390.1
31.40
46.72
72.19



AD-556963.1
42.99
68.75
146.93



AD-558078.1
30.67
83.78
62.14



AD-557204.1
40.18
69.41
199.62



AD-556962.1
67.05
92.06
80.95



AD-556733.1
34.07
61.86
62.03



AD-556724.1
43.85
114.37
225.16



AD-557067.1
42.27
59.20
67.56



AD-557602.1
39.99
70.66
155.25



AD-557345.1
45.66
91.09
186.86



AD-557969.1
70.94
72.23
72.25



AD-557867.1
43.92
108.43
178.41



AD-557860.1
49.94
63.71
134.13



AD-557868.1
52.22
99.81
112.51



AD-558064.1
61.63
72.39
59.83



AD-556725.1
45.46
62.37
68.67



AD-556787.1
58.40
102.98
163.60



AD-558062.1
55.53
70.65
156.02



AD-556874.1
83.34
90.67
62.04



AD-557066.1
75.61
93.55
168.75



AD-556791.1
64.56
74.17
75.36



AD-557450.1
56.23
68.84
77.40



AD-557865.1
66.83
94.99
153.50



AD-556961.1
69.87
93.70
206.00



AD-556920.1
52.86
81.48
188.46



AD-558074.1
74.73
132.35
71.18



AD-556738.1
72.67
123.81
206.91



AD-558004.1
100.55
103.41
190.48



AD-556918.1
85.53
97.13
199.73



AD-557209.1
84.13
96.02
140.72



AD-557861.1
66.07
84.41
120.27



AD-557226.1
83.39
94.85
123.74



AD-557856.1
84.68
85.69
137.01



AD-557839.1
114.79
91.84
69.49



AD-557873.1
84.73
102.93
65.71



AD-557417.1
88.08
115.91
225.81



AD-558106.1
95.90
110.10
217.75



AD-557788.1
87.87
109.59
129.42



AD-556917.1
79.04
71.81
78.26



AD-556739.1
87.37
115.81
70.32



AD-556726.1
96.01
129.96
221.48



AD-556790.1
109.55
118.05
193.15



AD-557604.1
74.95
133.00
177.11



AD-557495.1
96.32
130.00
254.48



AD-557016.1
106.90
131.43
110.73



AD-558105.1
77.28
73.82
85.07



AD-557872.1
102.16
123.17
172.64



AD-557346.1
113.18
138.10
228.78



AD-557851.1
107.51
138.22
178.85



AD-557786.1
117.86
149.34
168.00



AD-557857.1
118.52
127.67
145.90

















TABLE 9







Complement Factor B In Vitro Single Dose Screens in Hep3B cells














10 nM

1 nM

0.1 nM




Dose % of

Dose % of

Dose % of




Message

Message

Message



DuplexID
Remaining
STDEV
Remaining
STDEV
Remaining
STDEV
















AD-558657.1
4.81
0.55
6.98
1.27
24.75
5.46


AD-559020.1
5.18
0.75
7.21
1.31
17.95
3.88


AD-559023.1
7.71
0.93
9.85
2.87
32.65
5.12


AD-558860.1
8.35
1.69
14.34
1.97
33.70
7.41


AD-559143.1
8.57
1.09
11.19
1.02
28.52
7.22


AD-559021.1
8.65
3.21
13.97
4.56
42.99
5.93


AD-559144.1
9.25
0.94
16.75
3.32
33.52
2.97


AD-559435.1
10.47
0.85
13.57
1.00
38.94
5.33


AD-560019.1
10.57
0.67
12.96
1.19
17.67
1.23


AD-559722.1
11.24
1.35
19.04
1.63
42.16
2.85


AD-560016.1
11.42
1.30
17.54
2.39
28.72
3.63


AD-559008.1
11.51
3.34
17.60
0.78
45.58
15.60


AD-560018.1
11.76
1.89
16.87
2.51
28.83
4.60


AD-560045.1
12.13
1.71
28.22
11.60
53.32
14.16


AD-559375.1
12.45
1.28
21.86
1.78
61.75
7.34


AD-559142.1
13.01
2.52
20.98
3.88
43.82
6.89


AD-559160.1
13.06
3.15
19.02
1.35
47.19
7.99


AD-559374.1
13.17
3.28
27.58
5.37
72.78
11.80


AD-559369.1
13.75
0.79
25.72
0.58
62.74
6.62


AD-559148.1
13.81
1.72
25.39
1.60
53.03
5.36


AD-560163.1
14.11
0.87
20.31
1.31
49.05
4.99


AD-559398.1
14.23
3.97
18.11
2.34
46.53
6.07


AD-559060.1
14.39
3.41
32.15
3.07
62.25
5.65


AD-559392.1
14.69
3.59
21.68
2.40
55.88
8.59


AD-559451.1
14.84
3.18
25.50
3.19
43.71
5.05


AD-559721.1
15.23
3.73
23.03
3.34
51.33
10.51


AD-559146.1
15.26
5.05
28.14
1.99
71.88
8.41


AD-560165.1
15.74
2.70
25.08
2.14
65.45
14.31


AD-559921.1
15.99
1.86
19.91
4.59
38.78
9.90


AD-560021.1
16.02
3.62
37.73
4.08
58.59
3.73


AD-560164.1
16.23
2.00
22.73
3.67
63.13
4.12


AD-559614.1
16.27
4.70
31.75
4.73
49.34
6.16


AD-559393.1
16.46
3.99
31.63
4.04
61.56
10.13


AD-559717.1
16.68
1.69
20.78
3.33
42.46
10.02


AD-560017.1
16.71
2.32
28.04
3.35
57.72
16.22


AD-559440.1
16.90
1.64
27.94
4.84
50.75
9.54


AD-560166.1
17.02
4.84
20.01
3.95
58.88
13.01


AD-559059.1
17.61
5.82
33.74
3.87
59.15
7.45


AD-559300.1
17.90
5.18
34.67
2.86
67.79
6.31


AD-559946.1
18.11
1.61
36.74
5.04
62.70
7.20


AD-559147.1
18.33
2.11
47.39
7.94
85.39
10.23


AD-559755.1
19.99
5.12
26.88
2.30
52.33
10.49


AD-559446.1
20.58
2.25
30.84
7.47
64.13
7.62


AD-559026.1
21.11
2.49
41.40
14.50
65.91
11.08


AD-559714.1
22.71
0.91
32.56
2.47
69.69
6.49


AD-560160.1
22.85
5.07
28.30
2.24
65.27
9.41


AD-559437.1
25.76
4.50
44.87
12.58
78.13
8.91


AD-560162.1
25.95
4.26
43.29
5.64
71.14
20.99


AD-560161.1
26.34
3.19
38.91
4.83
68.19
5.53


AD-559924.1
26.34
8.91
36.46
3.90
57.07
6.32


AD-559865.1
27.49
2.47
47.05
6.32
75.04
14.94


AD-559449.1
28.57
3.20
54.33
4.83
70.39
14.64


AD-559788.1
28.70
6.51
62.57
7.45
79.25
8.07


AD-559617.1
29.43
9.48
67.18
6.78
88.54
10.81


AD-559719.1
30.38
3.11
55.82
5.69
83.18
12.99


AD-559718.1
32.51
3.42
57.77
8.99
82.88
18.79


AD-558697.1
32.69
5.66
44.32
3.45
58.56
7.73


AD-559357.1
33.33
3.84
33.06
4.91
59.41
16.08


AD-558965.1
35.13
2.66
34.85
11.29
51.66
15.94


AD-558267.1
37.43
7.93
35.43
6.56
40.76
7.84


AD-558639.1
37.99
5.49
46.82
7.78
44.37
7.45


AD-559925.1
39.15
17.96
74.82
7.86
75.18
4.17


AD-559450.1
40.47
6.34
63.09
4.35
53.12
12.34


AD-559302.1
41.61
5.63
59.86
3.32
79.54
9.58


AD-559074.1
41.79
3.21
57.94
3.73
62.36
5.75


AD-559947.1
43.30
2.71
66.78
11.57
73.55
7.33


AD-559442.1
47.58
8.47
69.78
10.91
92.36
7.58


AD-559359.1
48.10
2.37
86.83
11.09
102.01
6.31


AD-559616.1
50.68
6.63
93.13
11.78
88.54
12.26


AD-559864.1
63.01
11.07
91.70
12.15
89.75
13.66


AD-559866.1
85.15
11.86
94.33
6.79
79.14
12.77
















TABLE 10







Complement Factor B In Vitro Single Dose Screens in Primary Mouse Hepatocytes


(PMH)














10 nM

1 nM

0.1 nM




Dose % of

Dose % of

Dose % of




Message

Message

Message



DuplexID
Remaining
STDEV
Remaining
STDEV
Remaining
STDEV
















AD-558860.1
10.33
2.38
40.10
5.03
94.64
11.07


AD-560018.1
13.28
3.25
64.35
11.23
114.97
8.99


AD-560019.1
18.44
3.65
75.46
18.65
118.42
14.85


AD-559160.1
19.42
3.01
47.55
4.18
125.03
7.48


AD-559921.1
20.12
3.89
67.51
6.36
123.87
9.76


AD-559755.1
20.65
5.45
77.24
11.02
117.00
9.54


AD-560017.1
22.11
5.43
82.72
11.41
123.23
4.98


AD-559614.1
23.18
7.36
67.15
7.61
123.18
19.54


AD-559435.1
25.13
3.68
66.19
8.46
118.78
11.85


AD-560016.1
25.40
5.70
91.81
22.08
128.76
16.87


AD-559451.1
27.47
9.44
71.89
12.25
137.27
20.14


AD-559617.1
29.70
10.00
84.66
4.39
140.37
6.69


AD-560021.1
32.74
4.65
91.51
4.21
126.40
12.31


AD-559449.1
37.90
5.63
82.95
9.33
130.52
19.04


AD-559450.1
38.55
8.96
87.87
22.39
129.90
7.64


AD-559925.1
38.62
3.39
90.35
10.90
104.75
5.55


AD-559440.1
39.15
5.87
82.63
10.21
124.84
7.74


AD-559788.1
42.67
7.18
114.25
12.73
138.37
4.54


AD-559437.1
45.26
10.40
94.10
7.91
130.12
11.84


AD-559369.1
47.64
10.56
83.98
9.74
136.96
5.52


AD-559446.1
49.32
10.27
104.76
13.05
141.33
23.45


AD-559924.1
56.47
10.68
101.50
14.50
131.17
5.61


AD-558965.1
58.30
14.89
94.61
10.27
109.17
5.25


AD-560045.1
68.96
6.99
94.93
5.17
98.24
5.15


AD-559946.1
74.67
7.51
116.30
7.12
108.08
6.00


AD-558697.1
79.21
8.58
103.00
11.53
141.24
23.63


AD-559008.1
79.41
8.78
103.11
8.12
103.48
5.21


AD-559357.1
81.03
4.45
95.92
5.13
110.83
8.24


AD-559020.1
82.26
11.63
98.50
12.69
102.20
4.21


AD-559143.1
82.37
15.28
97.44
5.47
104.94
5.93


AD-559374.1
85.24
7.46
89.59
3.03
130.94
10.46


AD-560161.1
85.46
17.44
91.49
7.11
106.52
4.01


AD-559947.1
86.06
13.19
115.94
7.06
118.65
5.89


AD-559616.1
86.75
7.61
115.52
9.47
134.57
3.86


AD-559142.1
87.22
10.07
88.47
5.77
96.92
5.92


AD-558639.1
87.64
13.26
99.95
4.27
122.33
12.04


AD-560166.1
88.71
22.04
104.01
13.19
97.42
5.90


AD-559359.1
89.05
14.80
104.69
7.55
123.70
6.43


AD-558657.1
89.59
11.42
95.66
6.61
109.56
9.31


AD-559442.1
90.05
26.12
107.09
17.30
152.65
17.71


AD-559023.1
93.44
12.18
96.28
8.79
116.18
4.76


AD-560160.1
93.98
12.77
106.07
14.37
96.30
1.99


AD-559398.1
94.15
14.19
89.68
5.33
141.12
7.36


AD-559722.1
94.20
7.15
114.34
13.25
128.86
17.84


AD-559146.1
94.84
7.07
94.68
6.79
150.36
18.96


AD-558267.1
96.73
10.58
101.69
8.01
107.42
7.77


AD-559074.1
99.75
10.40
91.77
6.74
132.38
20.72


AD-560162.1
100.49
17.85
98.34
9.70
99.71
7.13


AD-559021.1
102.22
20.82
100.57
9.81
143.53
24.85


AD-559144.1
105.28
4.70
98.16
9.44
149.08
20.99


AD-559147.1
106.45
27.57
89.01
4.30
147.67
22.58


AD-560164.1
106.73
13.72
97.25
8.25
103.76
7.45


AD-559714.1
107.80
24.01
130.26
16.11
139.71
8.34


AD-560165.1
108.93
14.81
100.31
4.51
95.90
5.35


AD-559300.1
111.25
11.78
90.58
4.56
134.36
10.29


AD-559866.1
112.76
7.74
123.50
10.25
143.37
8.58


AD-559302.1
113.89
12.96
90.69
3.34
112.12
19.35


AD-560163.1
114.10
15.54
92.44
9.83
107.52
5.99


AD-559718.1
114.19
9.34
139.34
10.12
143.95
10.17


AD-559721.1
115.30
17.06
116.42
5.04
143.90
13.34


AD-559026.1
115.85
15.27
95.64
12.87
132.32
13.74


AD-559719.1
116.22
12.55
130.31
6.06
150.76
26.42


AD-559060.1
117.05
20.30
95.06
4.36
141.26
25.56


AD-559864.1
118.44
9.57
134.91
10.92
143.92
22.59


AD-559059.1
120.63
14.40
94.88
3.85
154.23
9.79


AD-559865.1
123.03
9.71
134.45
14.17
142.24
10.43


AD-559148.1
123.28
10.25
91.36
4.38
156.95
14.94


AD-559375.1
125.42
7.77
96.09
8.53
148.89
19.81


AD-559393.1
126.82
14.36
97.97
6.13
142.67
16.86


AD-559717.1
131.20
6.00
131.80
6.57
148.89
12.67


AD-559392.1
134.26
21.19
96.35
2.77
130.60
8.29
















TABLE 11







Complement Factor B In Vitro Single Dose Screens in


Primary Mouse Hepatocytes (PMH)














10 nM

1 nM

0.1 nM




Dose % of

Dose % of

Dose % of




Message

Message

Message



DuplexID
Remaining
STDEV
Remaining
STDEV
Remaining
STDEV
















AD-560969.1
1.76
0.44
12.85
2.32
72.08
7.11


AD-561537.1
1.97
0.30
13.11
1.38
57.66
1.85


AD-562262.1
2.07
0.40
14.20
2.60
54.79
3.33


AD-561960.1
2.82
0.45
15.86
1.84
76.74
10.88


AD-561580.1
2.88
1.59
18.55
1.98
69.39
11.71


AD-561254.1
2.89
0.86
15.62
4.05
61.84
5.51


AD-561578.1
2.95
0.42
25.93
1.50
76.75
7.73


AD-561538.1
3.50
0.97
4.45
1.11
31.69
5.04


AD-561252.1
3.60
1.65
16.00
2.58
59.33
6.64


AD-561963.1
3.62
0.30
29.50
8.22
81.62
4.93


AD-562027.1
3.89
0.78
23.89
5.04
70.54
7.81


AD-561653.1
4.01
0.74
23.82
3.53
80.62
7.07


AD-562137.1
4.14
1.55
24.06
11.00
69.61
6.62


AD-561536.1
4.21
1.10
29.07
5.79
80.38
5.28


AD-561253.1
5.04
1.15
27.31
6.10
68.27
12.61


AD-561959.1
5.17
1.81
50.97
13.35
90.71
8.16


AD-561573.1
5.38
0.27
33.50
11.42
91.06
13.14


AD-561651.1
6.36
1.01
49.06
2.79
92.69
9.37


AD-561148.1
6.36
0.87
38.00
1.38
89.30
12.00


AD-561962.1
7.16
1.19
36.21
6.11
91.19
5.39


AD-562237.1
8.80
2.07
53.16
6.31
86.16
11.24


AD-561654.1
9.91
1.61
55.56
7.65
101.67
5.04


AD-562136.1
10.27
0.81
62.46
9.28
97.86
7.44


AD-562026.1
11.34
1.47
60.69
2.81
89.22
3.89


AD-561101.1
11.78
2.75
62.67
4.18
84.92
9.50


AD-561539.1
12.00
1.31
53.65
10.81
84.80
12.57


AD-561961.1
14.01
2.50
58.58
7.03
101.01
6.13


AD-561535.1
14.27
3.13
70.96
10.08
103.90
12.70


AD-561652.1
16.40
3.40
64.57
7.97
96.09
9.38


AD-562260.1
19.73
4.82
77.55
5.09
91.07
8.78


AD-557041.2
25.76
5.84
50.26
6.69
93.59
0.72


AD-561655.1
26.16
3.41
80.98
7.80
102.69
10.61


AD-557043.1
27.03
3.02
69.48
3.50
95.16
3.84


AD-561603.1
31.99
4.57
78.55
6.25
86.39
7.35


AD-557044.1
38.05
3.97
53.45
3.97
90.84
7.63


AD-561266.1
38.64
2.80
79.00
6.75
89.14
5.01


AD-561541.1
45.60
6.83
92.73
1.82
107.60
8.31


AD-562257.1
52.27
2.66
93.01
8.78
100.58
4.10


AD-560538.1
57.85
5.71
92.26
2.62
102.77
6.06


AD-561579.1
65.71
10.54
91.74
8.83
101.05
6.91


AD-562259.1
66.28
3.34
91.38
7.85
96.23
5.92


AD-560968.1
73.08
6.97
87.16
9.40
100.89
2.47


AD-561647.1
74.09
6.95
96.10
5.34
103.62
8.94


AD-561469.1
78.98
10.38
91.17
7.11
102.25
6.72


AD-561540.1
84.14
6.35
95.72
4.13
104.96
9.90


AD-562135.1
87.86
10.90
102.43
11.38
102.47
11.43
















TABLE 12







Complement Factor B In Vitro Single Dose Screens in Hep3B Cells














10 nM

1 nM

0.1 nM




Dose % of

Dose % of

Dose % of




Message

Message

Message



DuplexID
Remaining
STDEV
Remaining
STDEV
Remaining
STDEV
















AD-560969.1
29.34
1.71
71.22
4.40
88.98
14.10


AD-561537.1
86.96
7.93
84.92
6.56
97.67
11.01


AD-562262.1
19.99
1.52
43.30
9.67
77.20
2.33


AD-561960.1
101.89
7.12
93.06
6.78
86.95
11.28


AD-561580.1
77.04
4.88
72.79
13.33
99.51
7.87


AD-561254.1
47.27
3.23
59.31
9.89
80.05
5.60


AD-561578.1
17.62
1.52
54.39
5.50
86.17
9.50


AD-561538.1
14.90
6.97
24.45
3.87
42.19
9.11


AD-561252.1
93.27
11.00
96.17
6.74
78.24
13.72


AD-561963.1
40.07
4.40
63.17
10.68
84.66
8.57


AD-562027.1
77.41
4.07
84.74
1.64
95.86
11.49


AD-561653.1
44.48
8.74
68.46
11.39
101.86
10.10


AD-562137.1
12.14
2.49
35.01
5.57
63.19
12.92


AD-561536.1
81.78
6.45
107.20
6.41
72.90
5.20


AD-561253.1
83.43
9.54
84.78
15.70
70.37
8.01


AD-561959.1
78.06
6.19
70.68
2.99
89.62
9.10


AD-561573.1
62.25
8.08
74.47
7.81
69.74
13.65


AD-561651.1
65.86
6.99
94.78
14.92
86.37
10.00


AD-561148.1
85.70
6.64
93.17
12.36
96.96
10.73


AD-561962.1
73.24
4.15
104.85
7.06
105.02
11.38


AD-562237.1
35.58
3.42
62.94
2.85
82.87
12.89


AD-561654.1
68.27
6.59
84.86
8.20
88.86
16.82


AD-562136.1
43.50
5.85
96.79
6.07
97.96
8.53


AD-562026.1
78.57
11.58
92.78
9.79
100.53
12.79


AD-561101.1
80.42
8.87
88.94
8.58
78.42
5.79


AD-561539.1
73.75
9.48
70.46
13.02
81.45
11.45


AD-561961.1
93.29
14.09
102.04
3.89
82.92
14.39


AD-561535.1
90.31
7.87
100.89
9.09
89.58
10.53


AD-561652.1
99.83
14.12
86.03
11.59
99.32
15.07


AD-562260.1
72.75
4.48
93.46
5.11
91.71
18.08


AD-557041.2
18.62
0.85
50.98
12.96
78.19
10.26


AD-561655.1
81.07
2.83
92.73
6.74
73.97
11.80


AD-557043.1
28.86
4.75
81.11
7.28
91.72
10.13


AD-561603.1
46.27
14.44
99.22
11.97
86.19
7.73


AD-557044.1
23.45
2.59
74.62
9.75
79.77
7.73


AD-561266.1
105.79
5.46
95.99
11.36
99.59
2.26


AD-561541.1
100.77
11.65
98.43
4.85
93.30
4.25


AD-562257.1
101.31
9.81
97.55
15.36
95.26
7.79


AD-560538.1
103.91
11.00
104.39
8.70
86.71
20.47


AD-561579.1
87.65
10.59
80.92
11.54
83.54
7.48


AD-562259.1
86.75
12.13
98.75
19.03
90.33
10.56


AD-560968.1
94.61
7.70
107.14
4.84
88.97
10.45


AD-561647.1
99.94
9.55
110.74
11.11
98.97
13.25


AD-561469.1
84.71
4.50
88.17
6.08
82.50
8.92


AD-561540.1
84.25
4.08
88.70
14.19
110.80
11.10


AD-562135.1
89.38
4.70
95.02
2.81
98.61
12.79









Example 3. In Vivo Screening of dsRNA Duplexes in Mice

Duplexes of interest, identified from the above in vitro studies, were evaluated in vivo.


In particular, 6-8-week old wild-type mice (C57BL/6) were administered 100 ml of a 2×1011 viral particles/ml solution of an adeno-associated virus 8 (AAV8) vector encoding human complement factor B (hCFB AAV) by intravenous tail vein injection at Day −14.


At day 0, mice were subcutaneously administered a single 2 mg/kg dose of a duplex of interest or PBS control (n=3/group). Table 13 provides the duplexes that were administered to the mice.


At day 0, 7, and 14 post dose, blood was collected and plasma was prepared for ELISA assay. At day 14 post-dose, animals were sacrificed, liver samples were collected and snap-frozen in liquid nitrogen and tissue mRNA was extracted.


The level of human CFB protein was determined by quantitative sandwich enzyme immunoassay (AssayMax™, Human Complement Factor B ELISA Kit). Table 14 shows that the protein levels of human CFB were reduced upon treatment with a single dose of siRNA targeting hCFB at 2 mg/kg.


The level of human CFB expression was measured by RT-QPCR, as described above. Human CFB mRNA levels were compared to the mRNA level of the housekeeping gene GAPDH. The values were then normalized to the average of PBS vehicle control group. The data were expressed as percent of baseline value, and were presented as mean plus standard deviation. As shown in Table 15, human CFB mRNA levels were reduced upon treatment with a single dose of siRNA targeting hCFB at 2 mg/kg.









TABLE 13







dsRNA Duplexes for In Vivo Screening
















SEQ
Range in


Duplex ID
Oligo ID
Strand
Sequence 5′ to 3′
ID NO:
NM_001710.5





AD-558657.2
A-1072067.1
sense
gsusgaguGfaUfGfAfgaucucuuuuL96
629
643-665



A-1075535.1
antis
asAfsaagAfgAfUfcucaUfcAfcucacsasu
700






AD-559020.2
A-1072793.1
sense
asasaaguGfuCfUfAfgucaacuuauL96
619
1145-1167



A-1075898.1
antis
asUfsaagUfuGfAfcuagAfcAfcuuuususg
690






AD-559023.2
A-1072799.1
sense
asgsugucUfaGfUfCfaacuuaauuuL96
631
1148-1170



A-1075901.1
antis
asAfsauuAfaGfUfugacUfaGfacacususu
702






AD-558860.2
A-1072473.1
sense
usgsccaaGfaCfUfCfcuucauguauL96
591
928-950



A-1075738.1
antis
asUfsacaUfgAfAfggagUfcUfuggcasgsg
662






AD-560019.2
A-1074791.1
sense
asgsagaaGfuCfGfUfuucauucaauL96
593
2396-2418



A-1076897.1
antis
asUfsugaAfuGfAfaacgAfcUfucucususg
664






AD-560016.2
A-1074785.1
sense
ascsaagaGfaAfGfUfcguuucauuuL96
600
2393-2415



A-1076894.1
antis
asAfsaugAfaAfCfgacuUfcUfcuugusgsa
671






AD-559008.2
A-1072769.1
sense
uscsacagGfaGfCfCfaaaaaguguuL96
617
1133-1155



A-1075886.1
antis
asAfscacUfuUfUfuggcUfcCfugugasasg
688






AD-559717.2
A-1074187.1
sense
csasggaaUfuCfCfUfgaauuuuauuL96
660
1976-1998



A-1076595.1
antis
asAfsuaaAfaUfUfcaggAfaUfuccugscsu
731






AD-557072.2
A-1072789.1
sense
csasaaaaGfuGfUfCfuagucaacuuL96
191
1143-1165



A-1072790.1
antis
asAfsguug(Agn)cuagacAfcUfuuuugsgsc
277






AD-558097.2
A-1074839.1
sense
usasguggAfuGfUfCfugcaaaaacuL96
192
2438-2460



A-1074840.1
antis
asGfsuuuu(Tgn)gcagacAfuCfcacuascsu
278






AD-557774.2
A-1074193.1
sense
gsasauucCfuGfAfAfuuuuaugacuL96
193
1979-2001



A-1074194.1
antis
asGfsucau(Agn)aaauucAfgGfaauucscsu
279






AD-557070.2
A-1072785.1
sense
gscscaaaAfaGfUfGfucuagucaauL96
194
1141-1163



A-1072786.1
antis
asUfsugac(Tgn)agacacUfuUfuuggcsusc
280






AD-558065.2
A-1074775.1
sense
asgsuucaCfaAfGfAfgaagucguuuL96
199
2388-2410



A-1074776.1
antis
asAfsacga(Cgn)uucucuUfgUfgaacusasu
285






AD-557853.2
A-1074351.1
sense
asgsggaaCfaAfCfUfcgagcuuuguL96
208
2078-2100



A-1074352.1
antis
asCfsaaag(Cgn)ucgaguUfgUfucccuscsg
294






AD-557079.2
A-1072803.1
sense
usgsucuaGfuCfAfAfcuuaauugauL96
211
1150-1172



A-1072804.1
antis
asUfscaau(Tgn)aaguugAfcUfagacascsu
297
















TABLE 14







Human CFB siRNA In Vivo Screening










ELISA Results D7
ELISA Results D14












Avg hCFB

Avg hCFB




Protein

Protein




Remaining
SD
Remaining
SD














PBS
110.13
22.47
104.31
17.38


Naïve
96.75
21.00
92.31
6.54


AD-558657.2
29.32
8.38
24.76
5.08


AD-559020.2
24.26
4.07
25.05
1.22


AD-559023.2
28.56
7.34
26.35
3.44


AD-558860.2
52.69
4.25
50.31
17.27


AD-560019.2
26.68
8.43
20.07
6.59


AD-560016.2
44.06
2.59
45.94
2.69


AD-559008.2
46.07
6.47
34.55
5.26


AD-559717.2
57.72
11.41
51.14
13.28


AD-557072.2
34.29
8.88
22.19
6.45


AD-558097.2
31.01
9.59
29.55
10.60


AD-557774.2
70.26
6.03
64.26
12.89


AD-557070.2
91.58
21.23
71.11
18.76


AD-558065.2
120.96
14.77
82.66
44.55


AD-557853.2
94.22
24.50
81.47
18.23


AD-557079.2
101.05
17.40
97.09
15.44
















TABLE 15







Human CFB siRNA In Vivo Screening


qPCR Results D14










% message




remaining
SD












PBS
100.19
7.12


Naïve
88.59
6.21


AD-558657.2
37.82
11.34


AD-559020.2
56.19
2.64


AD-559023.2
37.50
3.28


AD-558860.2
65.89
10.68


AD-560019.2
22.01
1.70


AD-560016.2
68.31
5.22


AD-559008.2
53.91
12.03


AD-559717.2
63.13
7.55


AD-557072.2
39.39
1.21


AD-558097.2
43.35
11.63


AD-557774.2
81.10
4.79


AD-557070.2
83.09
13.83


AD-558065.2
86.18
9.28


AD-557853.2
84.42
6.60


AD-557079.2
98.00
15.34









Example 4. In Vivo Screening of dsRNA Duplexes in Mice

Additional duplexes of interest, identified from the above in vitro studies, were evaluated in vivo.


In particular, 6-8-week old wild-type mice (C57BL/6) were administered 100 ml of a 2×1011 viral particles/ml solution of an adeno-associated virus 8 (AAV8) vector encoding human complement factor B (hCFB AAV) by intravenous tail vein injection at Day −14.


At day 0, mice were subcutaneously administered a single 2 mg/kg dose of a duplex of interest or PBS control (n=3/group). Table 16 provides the duplexes that were administered to the mice.


At day 0, 7, and 14 post dose, blood was collected and plasma was prepared for ELISA assay. At day 14 post-dose, animals were sacrificed, liver samples were collected and snap-frozen in liquid nitrogen and tissue mRNA was extracted.


The level of human CFB protein was determined by quantitative sandwich enzyme immunoassay (AssayMax™, Human Complement Factor B ELISA Kit). Table 17 shows that the protein levels of human CFB were reduced upon treatment with a single dose of siRNA targeting hCFB at 2 mg/kg.


The level of human CFB expression was measured by RT-QPCR, as described above. Human CFB mRNA levels were compared to the mRNA level of the housekeeping gene GAPDH. The values were then normalized to the average of PBS vehicle control group. The data were expressed as percent of baseline value, and were presented as mean plus standard deviation. As shown in Table 18, human CFB mRNA levels were reduced upon treatment with a single dose of siRNA targeting hCFB at 2 mg/kg.









TABLE 16







dsRNA Duplexes for In Vivo Screening
















SEQ
Range in


DuplexID
OligoID
Strand
Sequence 5′ to 3′
ID NO:
NM_001710.5





AD-560018.2
A-1074789.1
sense
asasgagaAfgUfCfGfuuucauucauL96
592
2395-2417



A-1076896.1
antis
asUfsgaaUfgAfAfacgaCfuUfcucuusgsu
663






AD-559375.2
A-1073503.1
sense
asuscuggAfuGfUfCfuauguguuuuL96
658
1541-1563



A-1076253.1
antis
asAfsaacAfcAfUfagacAfuCfcagausasa
729






AD-559160.2
A-1073073.1
sense
usasugaaGfaCfCfAfcaaguugaauL96
594
1306-1328



A-1076038.1
antis
asUfsucaAfcUfUfguggUfcUfucauasasu
665






AD-559374.2
A-1073501.1
sense
usasucugGfaUfGfUfcuauguguuuL96
621
1540-1562



A-1076252.1
antis
asAfsacaCfaUfAfgacaUfcCfagauasasu
692






AD-559060.2
A-1072873.1
sense
usgsguguGfaAfGfCfcaagauauguL96
653
1185-1207



A-1075938.1
antis
asCfsauaUfcUfUfggcuUfcAfcaccasusa
724






AD-559721.2
A-1074195.1
sense
asasuuccUfgAfAfUfuuuaugacuuL96
650
1980-2002



A-1076599.1
antis
asAfsgucAfuAfAfaauuCfaGfgaauuscsc
721






AD-559026.2
A-1072805.1
sense
gsuscuagUfcAfAfCfuuaauugaguL96
651
1151-1173



A-1075904.1
antis
asCfsucaAfuUfAfaguuGfaCfuagacsasc
722






AD-558225.2
A-1075095.1
sense
usgsaauuAfaAfAfCfagcugcgacuL96
207
2602-2624



A-1075096.1
antis
asGfsucgc(Agn)gcuguuUfuAfauucasasu
293






AD-557069.2
A-1072783.1
sense
asgsccaaAfaAfGfUfgucuagucauL96
206
1140-1162



A-1072784.1
antis
asUfsgacu(Agn)gacacuUfuUfuggcuscsc
292






AD-558068.2
A-1074781.1
sense
uscsacaaGfaGfAfAfgucguuucauL96
195
2391-2413



A-1074782.1
antis
asUfsgaaa(Cgn)gacuucUfcUfugugasasc
281






AD-557422.2
A-1073489.1
sense
gsasggauUfaUfCfUfggaugucuauL96
202
1534-1556



A-1073490.1
antis
asUfsagac(Agn)uccagaUfaAfuccucscsc
288






AD-558063.2
A-1074771.1
sense
asusaguuCfaCfAfAfgagaagucguL96
205
2386-2408



A-1074772.1
antis
asCfsgacu(Tgn)cucuugUfgAfacuauscsa
291






AD-558066.2
A-1074777.1
sense
gsusucacAfaGfAfGfaagucguuuuL96
212
2389-2411



A-1074778.1
antis
asAfsaacg(Agn)cuucucUfuGfugaacsusa
298






AD-556701.2
A-1072047.1
sense
csusacuaCfaAfUfGfugagugauguL96
197
633-655



A-1072048.1
antis
asCfsauca(Cgn)ucacauUfgUfaguagsgsg
283
















TABLE 17







Human CFB siRNA In Vivo Screening










ELISA Results D7
ELISA Results D14












Avg hCFB

Avg hCFB




Protein

Protein




Remaining
SD
Remaining
SD














PBS
115.67
31.44
117.68
25.52


Naïve
110.11
4.46
108.16
15.25


AD-560018.2
16.29
2.97
15.23
4.63


AD-559375.2
42.43
8.09
44.31
14.32


AD-559160.2
39.66
10.78
49.87
3.52


AD-559374.2
43.88
10.14
41.19
9.12


AD-559060.2
63.43
15.73
50.37
6.51


AD-559721.2
30.89
2.80
28.32
5.76


AD-559026.2
70.20
7.55
53.71
3.75


AD-558225.2
42.06
1.30
41.12
6.52


AD-557069.2
94.78
13.32
71.48
6.01


AD-558068.2
94.83
9.22
97.31
20.15


AD-557422.2
74.07
1.97
63.49
9.48


AD-558063.2
77.64
3.66
81.77
4.84


AD-558066.2
76.19
5.77
65.77
8.00


AD-556701.2
69.44
3.28
62.51
21.07
















TABLE 18







Human CFB siRNA In Vivo Screening


qPCR Results D14










% message




remaining
SD












PBS
100.4
9.3


Naïve
77.4
13.2


AD-560018.2
20.0
1.7


AD-559375.2
66.4
10.1


AD-559160.2
60.4
6.4


AD-559374.2
38.3
12.3


AD-559060.2
67.7
4.4


AD-559721.2
38.6
7.2


AD-559026.2
69.5
22.8


AD-558225.2
58.7
12.7


AD-557069.2
74.2
20.7


AD-558068.2
56.4
18.2


AD-557422.2
92.5
10.5


AD-558063.2
91.9
26.6


AD-558066.2
62.3
8.2


AD-556701.2
58.4
3.2









Example 5. Design and Synthesis of Additional dsRNA Duplexes

Additional siRNAs were designed, synthesized and annealed using methods known in the art and described above in Example 1.


Detailed lists of the additional unmodified CFB sense and antisense strand nucleotide sequences are shown in Table 19. Detailed lists of the modified CFB sense and antisense strand nucleotide sequences are shown in Table 20.


In vitro and in vivo single dose screens of these agents in HepG2 cells were performed as described in the Examples above. Briefly, HepG2 cells were transfected by adding 5 μl of Opti-MEM plus 0.25 μl of Lipofectamine RNAiMax per well (Invitrogen, Carlsbad CA. cat #13778-150) to 5 μl of each siRNA duplex to an individual well in a 96-well plate. The mixture was then incubated at room temperature for 15 minutes. Forty μl of Eagle's Minimum Essential Medium (ATCC Cat #30-2003) containing ˜2×104 HepG2 cells were then added to the siRNA mixture. Cells were incubated for 24 hours prior to RNA purification. Single dose experiments were performed at 10 nM. The assays were performed as quadruplicates.


The results of the single dose screens of the dsRNA agents listed in Tables 19 and 20 in HepG2 cells are shown in Table 21. The results are presented as the mean percentage of message remaining.









TABLE 19







Unmodified Sense and Antisense Strand Sequences of Complement Factor B dsRNA Agents




















Start
End






SEQ

SEQ
site
site




Duplex
Sense
ID
Antiense
ID
in NM_
in NM_




Name
Sequence 5′ to 3′
NO:
Sequence 5′ to 3′
NO:
001710.5
001710.5
Region
Exon


















AD-558312
AAGGGAAUGUGACCAGGUCUU
1029
AAGACCUGGUCACAUUCCCUUCC
1121
 153
 175
5′UTR
 1





AD-558336
CUGGAGUUUCAGCUUGGACAU
  43
AUGUCCAAGCUGAAACUCCAGAC
 129
 177
 199
5′UTR
 1





AD-558361
CCAAGCAGACAAGCAAAGCAU
1030
AUGCUUUGCUUGUCUGCUUGGCU
1122
 202
 224
5′UTR
 1





AD-558382
GCCAGGACACACCAUCCUGCU
1031
AGCAGGAUGGUGUGUCCUGGCUU
1123
 223
 245
5′UTR
 1





AD-558389
CCAGCUUCUCUCCUGCCUUCU
1032
AGAAGGCAGGAGAGAAGCUGGGC
1124
 250
 272
5′UTR
 1





AD-558407
UGCCUGAUGCCCUUUAUCUUU
1033
AAAGAUAAAGGGCAUCAGGCAGA
1125
 304
 326
CDS
 1





AD-558426
UGGGCCUCUUGUCUGGAGGUU
1034
AACCUCCAGACAAGAGGCCCAAG
1126
 323
 345
CDS
1-2





AD-558450
CCACCACUCCAUGGUCUUUGU
1035
ACAAAGACCAUGGAGUGGUGGUC
1127
 347
 369
CDS
 2





AD-558482
UCCUUCCGACUUCUCCAAGAU
1036
AUCUUGGAGAAGUCGGAAGGAGC
1128
 418
 440
CDS
 2





AD-558507
AGGCACUGGAGUACGUGUGUU
1037
AACACACGUACUCCAGUGCCUGG
1129
 443
 465
CDS
 2





AD-558522
UGUGUCCUUCUGGCUUCUACU
1038
AGUAGAAGCCAGAAGGACACACG
1130
 458
 480
CDS
 2





AD-558539
UACCCGUACCCUGUGCAGACU
1039
AGUCUGCACAGGGUACGGGUAGA
1131
 475
 497
CDS
 2





AD-558555
AGACACGUACCUGCAGAUCUU
1040
AAGAUCUGCAGGUACGUGUCUGC
1132
 491
 513
CDS
 2





AD-558579
AGACUCAAGACCAAAAGACUU
1041
AAGUCUUUUGGUCUUGAGUCUUC
1133
 533
 555
CDS
 2





AD-558612
AGUGCAGAGCAAUCCACUGUU
1042
AACAGUGGAUUGCUCUGCACUCU
1134
 566
 588
CDS
2-3





AD-558637
ACCACACGACUUCGAGAACGU
1043
ACGUUCUCGAAGUCGUGUGGUCU
1135
 591
 613
CDS
 3





AD-558646
CCUACUACAAUGUGAGUGAUU
1044
AAUCACUCACAUUGUAGUAGGGA
1136
 632
 654
CDS
 3





AD-558662
UGAUGAGAUCUCUUUCCACUU
1045
AAGUGGAAAGAGAUCUCAUCACU
1137
 648
 670
CDS
 3





AD-558679
ACUGCUAUGACGGUUACACUU
  52
AAGUGUAACCGUCAUAGCAGUGG
 138
 665
 687
CDS
 3





AD-558701
CACCUGCCAAGUGAAUGGCCU
1046
AGGCCAUUCACUUGGCAGGUGCG
1138
 705
 727
CDS
 3





AD-558737
AGCGAUCUGUGACAACGGAGU
  66
ACUCCGUUGUCACAGAUCGCUGU
1139
 741
 763
CDS
3-4





AD-558747
CAUUGGCACAAGGAAGGUGGU
1047
ACCACCUUCCUUGUGCCAAUGGG
1140
 789
 811
CDS
 4





AD-558778
CGCCUUGAAGACAGCGUCACU
1048
AGUGACGCUGUCUUCAAGGCGGU
1141
 820
 842
CDS
 4





AD-558813
GGCGAACGUGUCAGGAAGGUU
1049
AACCUUCCUGACACGUUCGCCGC
1142
 881
 903
CDS
 4





AD-558859
CUGCCAAGACUCCUUCAUGUU
1050
AACAUGAAGGAGUCUUGGCAGGA
1143
 927
 949
CDS
4-5





AD-558879
GCCGAAGCUUUCCUGUCUUCU
1051
AGAAGACAGGAAAGCUUCGGCCA
1144
 967
 989
CDS
 5





AD-558896
UUCCCUGACAGAGACCAUAGU
1052
ACUAUGGUCUCUGUCAGGGAAGA
1145
 984
1006
CDS
 5





AD-558918
GGAGUCGAUGCUGAGGAUGGU
1053
ACCAUCCUCAGCAUCGACUCCUU
1146
1006
1028
CDS
 5





AD-558935
CAACAGAAGCGGAAGAUCGUU
1054
AACGAUCUUCCGCUUCUGUUGUU
1147
1042
1064
CDS
 6





AD-558965
UCAGGCUCCAUGAACAUCUAU
 471
AUAGAUGUUCAUGGAGCCUGAAG
 542
1072
1094
CDS
 6





AD-558993
UAGAUGGAUCAGACAGCAUUU
1055
AAAUGCUGUCUGAUCCAUCUAGC
1148
1100
1122
CDS
 6





AD-558998
GCCAGCAACUUCACAGGAGCU
1056
AGCUCCUGUGAAGUUGCUGGCCC
1149
1123
1145
CDS
 6





AD-559023
AGUGUCUAGUCAACUUAAUUU
 489
AAAUUAAGUUGACUAGACACUUU
 560
1148
1170
CDS
 6





AD-559040
AUUGAGAAGGUGGCAAGUUAU
1057
AUAACUUGCCACCUUCUCAAUUA
1150
1165
1187
CDS
6-7





AD-559059
AUGGUGUGAAGCCAAGAUAUU
 513
AAUAUCUUGGCUUCACACCAUAA
 584
1184
1206
CDS
 7





AD-559074
GAUAUGGUCUAGUGACAUAUU
 495
AAUAUGUCACUAGACCAUAUCUU
 566
1199
1221
CDS
 7





AD-559089
AUUUGGGUCAAAGUGUCUGAU
1058
AUCAGACACUUUGACCCAAAUUU
1151
1234
1256
CDS
 7





AD-559112
AGACAGCAGUAAUGCAGACUU
1059
AAGUCUGCAUUACUGCUGUCUGC
1152
1257
1279
CDS
 7





AD-559143
CAGCUCAAUGAAAUCAAUUAU
 478
AUAAUUGAUUUCAUUGAGCUGCU
 549
1288
1310
CDS
 7





AD-559163
GAAGACCACAAGUUGAAGUCU
1060
AGACUUCAACUUGUGGUCUUCAU
1153
1309
1331
CDS
7-8





AD-559184
GGGACUAACACCAAGAAGGCU
1061
AGCCUUCUUGGUGUUAGUCCCUG
1154
1330
1352
CDS
 8





AD-559208
CAGGCAGUGUACAGCAUGAUU
1062
AAUCAUGCUGUACACUGCCUGGA
1155
1354
1376
CDS
 8





AD-559233
GGCCAGAUGACGUCCCUCCUU
1063
AAGGAGGGACGUCAUCUGGCCAG
1156
1379
1401
CDS
 8





AD-559274
UGUCAUCAUCCUCAUGACUGU
1064
ACAGUCAUGAGGAUGAUGACAUG
1157
1422
1444
CDS
 8





AD-559290
ACUGAUGGAUUGCACAACAUU
1065
AAUGUUGUGCAAUCCAUCAGUCA
1158
1438
1460
CDS
8-9





AD-559306
UUACUGUCAUUGAUGAGAUCU
1066
AGAUCUCAUCAAUGACAGUAAUU
1159
1472
1494
CDS
 9





AD-559329
GACUUGCUAUACAUUGGCAAU
1067
AUUGCCAAUGUAUAGCAAGUCCC
1160
1495
1517
CDS
 9





AD-559359
AACCCAAGGGAGGAUUAUCUU
 486
AAGAUAAUCCUCCCUUGGGUUUU
 557
1525
1547
CDS
 9





AD-559374
UAUCUGGAUGUCUAUGUGUUU
 479
AAACACAUAGACAUCCAGAUAAU
 550
1540
1562
CDS
 9-10





AD-559383
GGGCCUUUGGUGAACCAAGUU
1068
AACUUGGUUCACCAAAGGCCCGA
1161
1567
1589
CDS
10





AD-559398
CAAGUGAACAUCAAUGCUUUU
 491
AAAAGCAUUGAUGUUCACUUGGU
 562
1582
1604
CDS
10





AD-559421
UUCCAAGAAAGACAAUGAGCU
  45
AGCUCAUUGUCUUUCUUGGAAGC
1162
1605
1627
CDS
10





AD-559438
AGCAACAUGUGUUCAAAGUCU
1069
AGACUUUGAACACAUGUUGCUCA
1163
1622
1644
CDS
10





AD-559455
GUCAAGGAUAUGGAAAACCUU
1070
AAGGUUUUCCAUAUCCUUGACUU
1164
1639
1661
CDS
10





AD-559476
GAAGAUGUUUUCUACCAAAUU
1071
AAUUUGGUAGAAAACAUCUUCCA
1165
1660
1682
CDS
10





AD-559497
AUCGAUGAAAGCCAGUCUCUU
1072
AAGAGACUGGCUUUCAUCGAUCA
1166
1681
1703
CDS
10-11





AD-559512
UCUCUGAGUCUCUGUGGCAUU
1073
AAUGCCACAGAGACUCAGAGACU
1167
1696
1718
CDS
11





AD-559536
UGGGAACACAGGAAGGGUACU
1074
AGUACCCUUCCUGUGUUCCCAAA
1168
1720
1742
CDS
11





AD-559556
CGAUUACCACAAGCAACCAUU
1075
AAUGGUUGCUUGUGGUAAUCGGU
1169
1740
1762
CDS
11





AD-559590
UCAGUCAUUCGCCCUUCAAAU
1076
AUUUGAAGGGCGAAUGACUGAGA
1170
1774
1796
CDS
11-12





AD-559610
GGGACACGAGAGCUGUAUGGU
1077
ACCAUACAGCUCUCGUGUCCCUU
1171
1794
1816
CDS
12





AD-559616
GUGGUGUCUGAGUACUUUGUU
 482
AACAAAGUACUCAGACACCACAG
 553
1819
1841
CDS
12





AD-559641
CAGCAGCACAUUGUUUCACUU
1078
AAGUGAAACAAUGUGCUGCUGUC
1172
1844
1866
CDS
12





AD-559670
AAGGAACACUCAAUCAAGGUU
1079
AACCUUGAUUGAGUGUUCCUUGU
1173
1873
1895
CDS
12





AD-559704
UAGAAGUAGUCCUAUUUCACU
1080
AGUGAAAUAGGACUACUUCUAUC
1174
1925
1947
CDS
13





AD-559706
CAACUACAACAUUAAUGGGAU
1081
AUCCCAUUAAUGUUGUAGUUGGG
1175
1947
1969
CDS
13





AD-559722
AUUCCUGAAUUUUAUGACUAU
 492
AUAGUCAUAAAAUUCAGGAAUUC
 563
1981
2003
CDS
13





AD-559740
UAUGACGUUGCCCUGAUCAAU
1082
AUUGAUCAGGGCAACGUCAUAGU
1176
1999
2021
CDS
13





AD-559760
GCUCAAGAAUAAGCUGAAAUU
1083
AAUUUCAGCUUAUUCUUGAGCUU
1177
2019
2041
CDS
13





AD-559788
ACUAUCAGGCCCAUUUGUCUU
 466
AAGACAAAUGGGCCUGAUAGUCU
 537
2047
2069
CDS
13-14





AD-559799
AGGGAACAACUCGAGCUUUGU
  36
ACAAAGCUCGAGUUGUUCCCUCG
 122
2078
2100
CDS
14





AD-559823
UUCCUCCAACUACCACUUGCU
1084
AGCAAGUGGUAGUUGGAGGAAGC
1178
2102
2124
CDS
14





AD-559838
CUUGCCAGCAACAAAAGGAAU
1085
AUUCCUUUUGUUGCUGGCAAGUG
1179
2117
2139
CDS
14-15





AD-559873
CAGGAUAUCAAAGCUCUGUUU
1086
AAACAGAGCUUUGAUAUCCUGUG
1180
2152
2174
CDS
15





AD-559892
UUGUGUCUGAGGAGGAGAAAU
1087
AUUUCUCCUCCUCAGACACAAAC
1181
2171
2193
CDS
15





AD-559926
GGAGGUCUACAUCAAGAAUGU
1088
ACAUUCUUGAUGUAGACCUCCUU
1182
2205
2227
CDS
15





AD-559946
GUGAGAGAGAUGCUCAAUAUU
 473
AAUAUUGAGCAUCUCUCUCACAG
 544
2243
2265
CDS
16





AD-559958
GACAAAGUCAAGGACAUCUCU
  37
AGAGAUGUCCUUGACUUUGUCAU
 123
2275
2297
CDS
16





AD-559973
UCGGUUCCUUUGUACUGGAGU
1089
ACUCCAGUACAAAGGAACCGAGG
1183
2310
2332
CDS
16





AD-559993
GAGUGAGUCCCUAUGCUGACU
1090
AGUCAGCAUAGGGACUCACUCCU
1184
2330
2352
CDS
16





AD-559998
UACUUGCAGAGGUGAUUCUGU
1091
ACAGAAUCACCUCUGCAAGUAUU
1185
2355
2377
CDS
16-17





AD-560011
AGUUCACAAGAGAAGUCGUUU
  27
AAACGACUUCUCUUGUGAACUAU
 113
2388
2410
CDS
17





AD-560031
UCAUUCAAGUUGGUGUAAUCU
1092
AGAUUACACCAACUUGAAUGAAA
1186
2408
2430
CDS
17-18





AD-560040
GAGUAGUGGAUGUCUGCAAAU
1093
AUUUGCAGACAUCCACUACUCCC
1187
2435
2457
CDS
18





AD-560060
AACCAGAAGCGGCAAAAGCAU
1094
AUGCUUUUGCCGCUUCUGGUUUU
1188
2455
2477
CDS
18





AD-560099
GACUUUCACAUCAACCUCUUU
1095
AAAGAGGUUGAUGUGAAAGUCUC
1189
2494
2516
CDS
18





AD-560114
CUCUUUCAAGUGCUGCCCUGU
1096
ACAGGGCAGCACUUGAAAGAGGU
1190
2509
2531
CDS
18





AD-560138
AAGGAGAAACUCCAAGAUGAU
1097
AUCAUCUUGGAGUUUCUCCUUCA
1191
2533
2555
CDS
18





AD-560156
GAGGAUUUGGGUUUUCUAUAU
1098
AUAUAGAAAACCCAAAUCCUCAU
1192
2551
2573
CDS
18





AD-560163
CGUGGGAUUGAAUUAAAACAU
 506
AUGUUUUAAUUCAAUCCCACGCC
 577
2594
2616
3′UTR
18





AD-558378
GCAAGCCAGGACACACCAUCU
1099
AGAUGGUGUGUCCUGGCUUGCUU
1193
 219
 241







AD-558393
CUUCUCUCCUGCCUUCCAACU
1100
AGUUGGAAGGCAGGAGAGAAGCU
1194
 254
 276







AD-558424
CUUGGGCCUCUUGUCUGGAGU
1101
ACUCCAGACAAGAGGCCCAAGAU
1195
 321
 343







AD-558466
AGAGAUCAAAGGCGGCUCCUU
1102
AAGGAGCCGCCUUUGAUCUCUAC
1196
 402
 424







AD-558511
ACUGGAGUACGUGUGUCCUUU
1103
AAAGGACACACGUACUCCAGUGC
1197
 447
 469







AD-558574
CCUGAAGACUCAAGACCAAAU
1104
AUUUGGUCUUGAGUCUUCAGGGU
1198
 528
 550







AD-558595
GACUGUCAGGAAGGCAGAGUU
1105
AACUCUGCCUUCCUGACAGUCUU
1199
 549
 571







AD-558750
UGGCACAAGGAAGGUGGGCAU
1106
AUGCCCACCUUCCUUGUGCCAAU
1200
 792
 814







AD-558777
CCGCCUUGAAGACAGCGUCAU
1107
AUGACGCUGUCUUCAAGGCGGUA
1201
 819
 841







AD-559105
CUGAAGCAGACAGCAGUAAUU
1108
AAUUACUGCUGUCUGCUUCAGAC
1202
1250
1272







AD-559124
UGCAGACUGGGUCACGAAGCU
1109
AGCUUCGUGACCCAGUCUGCAUU
1203
1269
1291







AD-559189
UAACACCAAGAAGGCCCUCCU
1110
AGGAGGGCCUUCUUGGUGUUAGU
1204
1335
1357







AD-559226
AUGAGCUGGCCAGAUGACGUU
1111
AACGUCAUCUGGCCAGCUCAUCA
1205
1372
1394







AD-559330
ACUUGCUAUACAUUGGCAAGU
1112
ACUUGCCAAUGUAUAGCAAGUCC
1206
1496
1518







AD-559486
UCUACCAAAUGAUCGAUGAAU
1113
AUUCAUCGAUCAUUUGGUAGAAA
1207
1670
1692







AD-559532
GGUUUGGGAACACAGGAAGGU
1114
ACCUUCCUGUGUUCCCAAACCAU
1208
1716
1738







AD-559573
CAUGGCAGGCCAAGAUCUCAU
1115
AUGAGAUCUUGGCCUGCCAUGGU
1209
1757
1779







AD-559609
AGGGACACGAGAGCUGUAUGU
1116
ACAUACAGCUCUCGUGUCCCUUU
1210
1793
1815







AD-559668
ACAAGGAACACUCAAUCAAGU
1117
ACUUGAUUGAGUGUUCCUUGUCA
1211
1871
1893







AD-559688
AAGCGGGACCUGGAGAUAGAU
1118
AUCUAUCUCCAGGUCCCGCUUCU
1212
1909
1931







AD-559882
AAAGCUCUGUUUGUGUCUGAU
1119
AUCAGACACAAACAGAGCUUUGA
1213
2161
2183







AD-560123
UGGCUGAAGGAGAAACUCCAU
1120
AUGGAGUUUCUCCUUCAGCCAGG
1214
2527
2549
















TABLE 20







Modified Sense and Antisense Strand Sequences of Complement Factor B dsRNA Agents















SEQ

SEQ

SEQ


Duplex

ID
Antisense
ID

ID


Name
Sense Sequence 5′ to 3′
NO:
Sequence 5′ to 3′
NO:
mRNA Target Sequence
NO:





AD-558312
asasgggaAfuGfUfGfaccaggucuuL96
1215
asAfsgacCfuGfGfuc
1307
GGAAGGGAATGTGACCAGGTCTA
1406





acAfuUfcccuuscsc








AD-558336
csusggagUfuUfCfAfgcuuggacauL96
215
asUfsgucCfaAfGfcu
1308
GTCTGGAGTTTCAGCTTGGACAC
1407





gaAfaCfuccagsasc








AD-558361
cscsaagcAfgAfCfAfagcaaagcauL96
1216
asUfsgcuUfuGfCfuu
1309
AGCCAAGCAGACAAGCAAAGCAA
1408





guCfuGfcuuggscsu








AD-558382
gscscaggAfcAfCfAfccauccugcuL96
1217
asGfscagGfaUfGfgu
1310
AAGCCAGGACACACCATCCTGCC
1409





guGfuCfcuggcsusu








AD-558389
cscsagcuUfcUfCfUfccugccuucuL96
1218
asGfsaagGfcAfGfga
1311
GCCCAGCTTCTCTCCTGCCTTCC
1410





gaGfaAfgcuggsgsc








AD-558407
usgsccugAfuGfCfCfcuuuaucuuuL96
1219
asAfsagaUfaAfAfgg
1312
TCTGCCTGATGCCCTTTATCTTG
1411





gcAfuCfaggcasgsa








AD-558426
usgsggccUfcUfUfGfucuggagguuL96
1220
asAfsccuCfcAfGfac
1313
CTTGGGCCTCTTGTCTGGAGGTG
1412





aaGfaGfgcccasasg








AD-558450
cscsaccaCfuCfCfAfuggucuuuguL96
1221
asCfsaaaGfaCfCfau
1314
GACCACCACTCCATGGTCTTTGG
1413





ggAfgUfgguggsusc








AD-558482
uscscuucCfgAfCfUfucuccaagauL96
1222
asUfscuuGfgAfGfaa
1315
GCTCCTTCCGACTTCTCCAAGAG
1414





guCfgGfaaggasgsc








AD-558507
asgsgcacUfgGfAfGfuacguguguuL96
1223
asAfscacAfcGfUfac
1316
CCAGGCACTGGAGTACGTGTGTC
1415





ucCfaGfugccusgsg








AD-558522
usgsugucCfuUfCfUfggcuucuacuL96
1224
asGfsuagAfaGfCfca
1317
CGTGTGTCCTTCTGGCTTCTACC
1416





gaAfgGfacacascsg








AD-558539
usascccgUfaCfCfCfugugcagacuL96
1225
asGfsucuGfcAfCfag
1318
TCTACCCGTACCCTGTGCAGACA
1417





ggUfaCfggguasgsa








AD-558555
asgsacacGfuAfCfCfugcagaucuuL96
1226
asAfsgauCfuGfCfag
1319
GCAGACACGTACCTGCAGATCTA
1418





guAfcGfugucusgsc








AD-558579
asgsacucAfaGfAfCfcaaaagacuuL96
1227
asAfsgucUfuUfUfgg
1320
GAAGACTCAAGACCAAAAGACTG
1419





ucUfuGfagucususc








AD-558612
asgsugcaGfaGfCfAfauccacuguuL96
1228
asAfscagUfgGfAfuu
1321
AGAGTGCAGAGCAATCCACTGTC
1420





gcUfcUfgcacuscsu








AD-558637
ascscacaCfgAfCfUfucgagaacguL96
1229
asCfsguuCfuCfGfaa
1322
AGACCACACGACTTCGAGAACGG
1421





guCfgUfgugguscsu








AD-558646
cscsuacuAfcAfAfUfgugagugauuL96
1230
asAfsucaCfuCfAfca
1323
TCCCTACTACAATGTGAGTGATG
1422





uuGfuAfguaggsgsa








AD-558662
usgsaugaGfaUfCfUfcuuuccacuuL96
1231
asAfsgugGfaAfAfga
1324
AGTGATGAGATCTCTTTCCACTG
1423





gaUfcUfcaucascsu








AD-558679
ascsugcuAfuGfAfCfgguuacacuuL96
224
asAfsgugUfaAfCfcg
1325
CCACTGCTATGACGGTTACACTC
1424





ucAfuAfgcagusgsg








AD-558701
csasccugCfcAfAfGfugaauggccuL96
1232
asGfsgccAfuUfCfac
1326
CGCACCTGCCAAGTGAATGGCCG
1425





uuGfgCfaggugscsg








AD-558737
asgscgauCfuGfUfGfacaacggaguL96
 238
asCfsuccGfuUfGfuc
1327
ACAGCGATCTGTGACAACGGAGC
1426





acAfgAfucgcusgsu








AD-558747
csasuuggCfaCfAfAfggaaggugguL96
1233
asCfscacCfuUfCfcu
1328
CCCATTGGCACAAGGAAGGTGGG
1427





ugUfgCfcaaugsgsg








AD-558778
csgsccuuGfaAfGfAfcagcgucacuL96
1234
asGfsugaCfgCfUfgu
1329
ACCGCCTTGAAGACAGCGTCACC
1428





cuUfcAfaggcgsgsu








AD-558813
gsgscgaaCfgUfGfUfcaggaagguuL96
1235
asAfsccuUfcCfUfga
1330
GCGGCGAACGTGTCAGGAAGGTG
1429





caCfgUfucgccsgsc








AD-558859
csusgccaAfgAfCfUfccuucauguuL96
1236
asAfscauGfaAfGfga
1331
TCCTGCCAAGACTCCTTCATGTA
1430





guCfuUfggcagsgsa








AD-558879
gscscgaaGfcUfUfUfccugucuucuL96
1237
asGfsaagAfcAfGfga
1332
TGGCCGAAGCTTTCCTGTCTTCC
1431





aaGfcUfucggcscsa








AD-558896
ususcccuGfaCfAfGfagaccauaguL96
1238
asCfsuauGfgUfCfuc
1333
TCTTCCCTGACAGAGACCATAGA
1432





ugUfcAfgggaasgsa








AD-558918
gsgsagucGfaUfGfCfugaggaugguL96
1239
asCfscauCfcUfCfag
1334
AAGGAGTCGATGCTGAGGATGGG
1433





caUfcGfacuccsusu








AD-558935
csasacagAfaGfCfGfgaagaucguuL96
1240
asAfscgaUfcUfUfcc
1335
AACAACAGAAGCGGAAGATCGTC
1434





gcUfuCfuguugsusu








AD-558965
uscsaggcUfcCfAfUfgaacaucuauL96 
 613
asUfsagaUfgUfUfca
 684
CTTCAGGCTCCATGAACATCTAC
1435





ugGfaGfccugasasg








AD-558993
usasgaugGfaUfCfAfgacagcauuuL96
1241
asAfsaugCfuGfUfcu
1336
GCTAGATGGATCAGACAGCATTG
1436





gaUfcCfaucuasgsc








AD-558998
gscscagcAfaCfUfUfcacaggagcuL96
1242
asGfscucCfuGfUfga
1337
GGGCCAGCAACTTCACAGGAGCC
1437





agUfuGfcuggcscsc








AD-559023
asgsugucUfaGfUfCfaacuuaauuuL96
 631
asAfsauuAfaGfUfug
 702
AAAGTGTCTAGTCAACTTAATTG
1438





acUfaGfacacususu








AD-559040
asusugagAfaGfGfUfggcaaguuauL96
1243
asUfsaacUfuGfCfca
1338
TAATTGAGAAGGTGGCAAGTTAT
1439





ccUfuCfucaaususa








AD-559059
asusggugUfgAfAfGfccaagauauuL96
 655
asAfsuauCfuUfGfgc
 726
TTATGGTGTGAAGCCAAGATATG
1440





uuCfaCfaccausasa








AD-559074
gsasuaugGfuCfUfAfgugacauauuL96
 637
asAfsuauGfuCfAfcu
 708
AAGATATGGTCTAGTGACATATG
1441





agAfcCfauaucsusu








AD-559089
asusuuggGfuCfAfAfagugucugauL96
1244
asUfscagAfcAfCfuu
1339
AAATTTGGGTCAAAGTGTCTGAA
1442





ugAfcCfcaaaususu








AD-559112
asgsacagCfaGfUfAfaugcagacuuL96
1245
asAfsgucUfgCfAfuu
1340
GCAGACAGCAGTAATGCAGACTG
1443





acUfgCfugucusgsc








AD-559143
csasgcucAfaUfGfAfaaucaauuauL96
 620
asUfsaauUfgAfUfuu
 691
AGCAGCTCAATGAAATCAATTAT
1444





caUfuGfagcugscsu








AD-559163
gsasagacCfaCfAfAfguugaagucuL96
1246
asGfsacuUfcAfAfcu
1341
ATGAAGACCACAAGTTGAAGTCA
1445





ugUfgGfucuucsasu








AD-559184
gsgsgacuAfaCfAfCfcaagaaggcuL96
1247
asGfsccuUfcUfUfgg
1342
CAGGGACTAACACCAAGAAGGCC
1446





ugUfuAfgucccsusg








AD-559208
csasggcaGfuGfUfAfcagcaugauuL96
1248
asAfsucaUfgCfUfgu
1343
TCCAGGCAGTGTACAGCATGATG
1447





acAfcUfgccugsgsa








AD-559233
gsgsccagAfuGfAfCfgucccuccuuL96
1249
asAfsggaGfgGfAfcg
1344
CTGGCCAGATGACGTCCCTCCTG
1448





ucAfuCfuggccsasg








AD-559274
usgsucauCfaUfCfCfucaugacuguL96
1250
asCfsaguCfaUfGfag
1345
CATGTCATCATCCTCATGACTGA
1449





gaUfgAfugacasusg








AD-559290
ascsugauGfgAfUfUfgcacaacauuL96
1251
asAfsuguUfgUfGfca
1346
TGACTGATGGATTGCACAACATG
1450





auCfcAfucaguscsa








AD-559306
ususacugUfcAfUfUfgaugagaucuL96
1252
asGfsaucUfcAfUfca
1347
AATTACTGTCATTGATGAGATCC
1451





auGfaCfaguaasusu








AD-559329
gsascuugCfuAfUfAfcauuggcaauL96
1253
asUfsugcCfaAfUfgu
1348
GGGACTTGCTATACATTGGCAAG
1452





auAfgCfaagucscsc








AD-559359
asascccaAfgGfGfAfggauuaucuuL96
 628
asAfsgauAfaUfCfcu
 699
AAAACCCAAGGGAGGATTATCTG
1453





ccCfuUfggguususu








AD-559374
usasucugGfaUfGfUfcuauguguuuL96
 621
asAfsacaCfaUfAfga
 692
ATTATCTGGATGTCTATGTGTTT
1454





caUfcCfagauasasu








AD-559383
gsgsgccuUfuGfGfUfgaaccaaguuL96
1254
asAfscuuGfgUfUfca
1349
TCGGGCCTTTGGTGAACCAAGTG
1455





ccAfaAfggcccsgsa








AD-559398
csasagugAfaCfAfUfcaaugcuuuuL96
 633
asAfsaagCfaUfUfga
 704
ACCAAGTGAACATCAATGCTTTG
1456





ugUfuCfacuugsgsu








AD-559421
ususccaaGfaAfAfGfacaaugagcuL96
 217
asGfscucAfuUfGfuc
1350
GCTTCCAAGAAAGACAATGAGCA
1457





uuUfcUfuggaasgsc








AD-559438
asgscaacAfuGfUfGfuucaaagucuL96
1255
asGfsacuUfuGfAfac
1351
TGAGCAACATGTGTTCAAAGTCA
1458





acAfuGfuugcuscsa








AD-559455
gsuscaagGfaUfAfUfggaaaaccuuL96
1256
asAfsgguUfuUfCfca
1352
AAGTCAAGGATATGGAAAACCTG
1459





uaUfcCfuugacsusu








AD-559476
gsasagauGfuUfUfUfcuaccaaauuL96
1257
asAfsuuuGfgUfAfga
1353
TGGAAGATGTTTTCTACCAAATG
1460





aaAfcAfucuucscsa








AD-559497
asuscgauGfaAfAfGfccagucucuuL96
1258
asAfsgagAfcUfGfgc
1354
TGATCGATGAAAGCCAGTCTCTG
1461





uuUfcAfucgauscsa








AD-559512
uscsucugAfgUfCfUfcuguggcauuL96
1259
asAfsugcCfaCfAfga
1355
AGTCTCTGAGTCTCTGTGGCATG
1462





gaCfuCfagagascsu








AD-559536
usgsggaaCfaCfAfGfgaaggguacuL96
1260
asGfsuacCfcUfUfcc
1356
TTTGGGAACACAGGAAGGGTACC
1463





ugUfgUfucccasasa








AD-559556
csgsauuaCfcAfCfAfagcaaccauuL96
1261
asAfsuggUfuGfCfuu
1357
ACCGATTACCACAAGCAACCATG
1464





guGfgUfaaucgsgsu








AD-559590
uscsagucAfuUfCfGfcccuucaaauL96
1262
asUfsuugAfaGfGfgc
1358
TCTCAGTCATTCGCCCTTCAAAG
1465





gaAfuGfacugasgsa








AD-559610
gsgsgacaCfgAfGfAfgcuguaugguL96
1263
asCfscauAfcAfGfcu
1359
AAGGGACACGAGAGCTGTATGGG
1466





cuCfgUfgucccsusu








AD-559616
gsusggugUfcUfGfAfguacuuuguuL96
 624
asAfscaaAfgUfAfcu
 695
CTGTGGTGTCTGAGTACTTTGTG
1467





caGfaCfaccacsasg








AD-559641
csasgcagCfaCfAfUfuguuucacuuL96
1264
asAfsgugAfaAfCfaa
1360
GACAGCAGCACATTGTTTCACTG
1468





ugUfgCfugcugsusc








AD-559670
asasggaaCfaCfUfCfaaucaagguuL96
1265
asAfsccuUfgAfUfug
1361
ACAAGGAACACTCAATCAAGGTC
1469





agUfgUfuccuusgsu








AD-559704
usasgaagUfaGfUfCfcuauuucacuL96
1266
asGfsugaAfaUfAfgg
1362
GATAGAAGTAGTCCTATTTCACC
1470





acUfaCfuucuasusc








AD-559706
csasacuaCfaAfCfAfuuaaugggauL96
1267
asUfscccAfuUfAfau
1363
CCCAACTACAACATTAATGGGAA
1471





guUfgUfaguugsgsg








AD-559722
asusuccuGfaAfUfUfuuaugacuauL96
 634
asUfsaguCfaUfAfaa
 705
GAATTCCTGAATTTTATGACTAT
1472





auUfcAfggaaususc








AD-559740
usasugacGfuUfGfCfccugaucaauL96
1268
asUfsugaUfcAfGfgg
1364
ACTATGACGTTGCCCTGATCAAG
1473





caAfcGfucauasgsu








AD-559760
gscsucaaGfaAfUfAfagcugaaauuL96
1269
asAfsuuuCfaGfCfuu
1365
AAGCTCAAGAATAAGCTGAAATA
1474





auUfcUfugagcsusu








AD-559788
ascsuaucAfgGfCfCfcauuugucuuL96
 608
asAfsgacAfaAfUfgg
 679
AGACTATCAGGCCCATTTGTCTC
1475





gcCfuGfauaguscsu








AD-559799
asgsggaaCfaAfCfUfcgagcuuuguL96
 208
asCfsaaaGfcUfCfga
1366
CGAGGGAACAACTCGAGCTTTGA
1476





guUfgUfucccuscsg








AD-559823
ususccucCfaAfCfUfaccacuugcuL96
1270
asGfscaaGfuGfGfua
1367
GCTTCCTCCAACTACCACTTGCC
1477





guUfgGfaggaasgsc








AD-559838
csusugccAfgCfAfAfcaaaaggaauL96
1271
asUfsuccUfuUfUfgu
1368
CACTTGCCAGCAACAAAAGGAAG
1478





ugCfuGfgcaagsusg








AD-559873
csasggauAfuCfAfAfagcucuguuuL96
1272
asAfsacaGfaGfCfuu
1369
CACAGGATATCAAAGCTCTGTTT
1479





ugAfuAfuccugsusg








AD-559892
ususguguCfuGfAfGfgaggagaaauL96
1273
asUfsuucUfcCfUfcc
1370
GTTTGTGTCTGAGGAGGAGAAAA
1480





ucAfgAfcacaasasc








AD-559926
gsgsagguCfuAfCfAfucaagaauguL96
1274
asCfsauuCfuUfGfau
1371
AAGGAGGTCTACATCAAGAATGG
1481





guAfgAfccuccsusu








AD-559946
gsusgagaGfaGfAfUfgcucaauauuL96
 615
asAfsuauUfgAfGfca
 686
CTGTGAGAGAGATGCTCAATATG
1482





ucUfcUfcucacsasg








AD-559958
gsascaaaGfuCfAfAfggacaucucuL96
 209
asGfsagaUfgUfCfcu
1372
ATGACAAAGTCAAGGACATCTCA
1483





ugAfcUfuugucsasu








AD-559973
uscsgguuCfcUfUfUfguacuggaguL96
1275
asCfsuccAfgUfAfca
1373
CCTCGGTTCCTTTGTACTGGAGG
1484





aaGfgAfaccgasgsg








AD-559993
gsasgugaGfuCfCfCfuaugcugacuL96
1276
asGfsucaGfcAfUfag
1374
AGGAGTGAGTCCCTATGCTGACC
1485





ggAfcUfcacucscsu








AD-559998
usascuugCfaGfAfGfgugauucuguL96
1277
asCfsagaAfuCfAfcc
1375
AATACTTGCAGAGGTGATTCTGG
1486





ucUfgCfaaguasusu








AD-560011
asgsuucaCfaAfGfAfgaagucguuuL96
 199
asAfsacgAfcUfUfcu
1376
ATAGTTCACAAGAGAAGTCGTTT
1487





cuUfgUfgaacusasu








AD-560031
uscsauucAfaGfUfUfgguguaaucuL96
1278
asGfsauuAfcAfCfca
1377
TTTCATTCAAGTTGGTGTAATCA
1488





acUfuGfaaugasasa








AD-560040
gsasguagUfgGfAfUfgucugcaaauL96
1279
asUfsuugCfaGfAfca
1378
GGGAGTAGTGGATGTCTGCAAAA
1489





ucCfaCfuacucscsc








AD-560060
asasccagAfaGfCfGfgcaaaagcauL96
1280
asUfsgcuUfuUfGfcc
1379
AAAACCAGAAGCGGCAAAAGCAG
1490





gcUfuCfugguususu








AD-560099
gsascuuuCfaCfAfUfcaaccucuuuL96
1281
asAfsagaGfgUfUfga
1380
GAGACTTTCACATCAACCTCTTT
1491





ugUfgAfaagucsusc








AD-560114
csuscuuuCfaAfGfUfgcugcccuguL96
1282
asCfsaggGfcAfGfca
1381
ACCTCTTTCAAGTGCTGCCCTGG
1492





cuUfgAfaagagsgsu








AD-560138
asasggagAfaAfCfUfccaagaugauL96
1283
asUfscauCfuUfGfga
1382
TGAAGGAGAAACTCCAAGATGAG
1493





guUfuCfuccuuscsa








AD-560156
gsasggauUfuGfGfGfuuuucuauauL96
1284
asUfsauaGfaAfAfac
1383
ATGAGGATTTGGGTTTTCTATAA
1494





ccAfaAfuccucsasu








AD-560163
csgsugggAfuUfGfAfauuaaaacauL96
 648
asUfsguuUfuAfAfuu
 719
GGCGTGGGATTGAATTAAAACAG
1495





caAfuCfccacgscsc








AD-558378
gscsaagcCfaGfGfAfcacaccaucuL96
1285
asGfsaugGfuGfUfgu
1384
AAGCAAGCCAGGACACACCAUCC
1496





ccUfgGfcuugcsusu








AD-558393
csusucucUfcCfUfGfccuuccaacuL96
1286
asGfsuugGfaAfGfgc
1385
AGCUUCUCUCCUGCCUUCCAACG
1497





agGfaGfagaagscsu








AD-558424
csusugggCfcUfCfUfugucuggaguL96
1287
asCfsuccAfgAfCfaa
1386
AUCUUGGGCCUCUUGUCUGGAGG
1498





gaGfgCfccaagsasu








AD-558466
asgsagauCfaAfAfGfgcggcuccuuL96
1288
asAfsggaGfcCfGfcc
1387
GUAGAGAUCAAAGGCGGCUCCUU
1499





uuUfgAfucucusasc








AD-558511
ascsuggaGfuAfCfGfuguguccuuuL96
1289
asAfsaggAfcAfCfac
1388
GCACUGGAGUACGUGUGUCCUUC
1500





guAfcUfccagusgsc








AD-558574
cscsugaaGfaCfUfCfaagaccaaauL96
1290
asUfsuugGfuCfUfug
1389
ACCCUGAAGACUCAAGACCAAAA
1501





agUfcUfucaggsgsu








AD-558595
gsascuguCfaGfGfAfaggcagaguuL96
1291
asAfscucUfgCfCfuu
1390
AAGACUGUCAGGAAGGCAGAGUG
1502





ccUfgAfcagucsusu








AD-558750
usgsgcacAfaGfGfAfaggugggcauL96
1292
asUfsgccCfaCfCfuu
1391
AUUGGCACAAGGAAGGUGGGCAG
1503





ccUfuGfugccasasu








AD-558777
cscsgccuUfgAfAfGfacagcgucauL96
1293
asUfsgacGfcUfGfuc
1392
UACCGCCUUGAAGACAGCGUCAC
1504





uuCfaAfggcggsusa








AD-559105
csusgaagCfaGfAfCfagcaguaauuL96
1294
asAfsuuaCfuGfCfug
1393
GUCUGAAGCAGACAGCAGUAAUG
1505





ucUfgCfuucagsasc








AD-559124
usgscagaCfuGfGfGfucacgaagcuL96
1295
asGfscuuCfgUfGfac
1394
AAUGCAGACUGGGUCACGAAGCA
1506





ccAfgUfcugcasusu








AD-559189
usasacacCfaAfGfAfaggcccuccuL96
1296
asGfsgagGfgCfCfuu
1395
ACUAACACCAAGAAGGCCCUCCA
1507





cuUfgGfuguuasgsu








AD-559226
asusgagcUfgGfCfCfagaugacguuL96
1297
asAfscguCfaUfCfug
1396
UGAUGAGCUGGCCAGAUGACGUC
1508





gcCfaGfcucauscsa








AD-559330
ascsuugcUfaUfAfCfauuggcaaguL96
1298
asCfsuugCfcAfAfug
1397
GGACUUGCUAUACAUUGGCAAGG
1509





uaUfaGfcaaguscsc








AD-559486
uscsuaccAfaAfUfGfaucgaugaauL96
1299
asUfsucaUfcGfAfuc
1398
UUUCUACCAAAUGAUCGAUGAAA
1510





auUfuGfguagasasa








AD-559532
gsgsuuugGfgAfAfCfacaggaagguL96
1300
asCfscuuCfcUfGfug
1399
AUGGUUUGGGAACACAGGAAGGG
1511





uuCfcCfaaaccsasu








AD-559573
csasuggcAfgGfCfCfaagaucucauL96
1301
asUfsgagAfuCfUfug
1400
ACCAUGGCAGGCCAAGAUCUCAG
1512





gcCfuGfccaugsgsu








AD-559609
asgsggacAfcGfAfGfagcuguauguL96
1302
asCfsauaCfaGfCfuc
1401
AAAGGGACACGAGAGCUGUAUGG
1513





ucGfuGfucccususu








AD-559668
ascsaaggAfaCfAfCfucaaucaaguL96
1303
asCfsuugAfuUfGfag
1402
UGACAAGGAACACUCAAUCAAGG
1514





ugUfuCfcuuguscsa








AD-559688
asasgcggGfaCfCfUfggagauagauL96
1304
asUfscuaUfcUfCfca
1403
AGAAGCGGGACCUGGAGAUAGAA
1515





ggUfcCfcgcuuscsu








AD-559882
asasagcuCfuGfUfUfugugucugauL96
1305
asUfscagAfcAfCfaa
1404
UCAAAGCUCUGUUUGUGUCUGAG
1516





acAfgAfgcuuusgsa
















TABLE 21







In Vitro Single Dose Screen in HepG2 cells










CFB/gapdh




10 nM












% average





message

Target region in


Duplex Name
remaining
SD
NM_001710.5













AD-560132.1
9.927
1.092
2527-2549


AD-560099.1
9.213
1.335
2494-2516


AD-559998.1
11.297
1.165
2355-2377


AD-559993.1
11.852
1.080
2330-2352


AD-559973.1
53.866
5.431
2310-2332


AD-559882.1
10.290
1.623
2161-2183


AD-559706.1
11.537
1.805
1947-1969


AD-559704.1
15.239
3.563
1924-1947


AD-559688.1
38.443
9.924
1909-1931


AD-559668.1
10.169
0.593
1871-1893


AD-559641.1
29.195
3.820
1844-1866


AD-559609.1
88.678
6.099
1793-1815


AD-559590.1
35.152
4.022
1774-1796


AD-559573.1
38.212
6.701
1757-1779


AD-559532.1
74.766
13.877
1716-1738


AD-559486.1
24.644
9.120
1670-1692


AD-559330.1
95.265
4.050
1496-1518


AD-559274.1
13.751
2.733
1422-1444


AD-559226.1
102.524
5.658
1372-1394


AD-559208.1
16.066
2.069
1354-1376


AD-559189.1
16.100
2.031
1335-1357


AD-559124.1
15.423
2.069
1269-1291


AD-559105.1
9.673
1.193
1250-1272


AD-559089.1
10.663
2.329
1234-1256


AD-558935.1
13.757
1.222
1042-1064


AD-558879.1
14.752
3.406
967-989


AD-558777.1
14.623
2.074
819-841


AD-558750.1
16.390
2.306
792-814


AD-558637.1
24.702
7.767
591-613


AD-558612.1
12.506
1.594
566-588


AD-558595.1
11.413
1.630
549-571


AD-558574.1
12.253
1.051
528-550


AD-558555.1
12.550
1.821
491-513


AD-558511.1
10.434
4.050
447-469


AD-558482.1
47.746
10.594
418-440


AD-558466.1
17.794
3.389
402-424


AD-558450.1
11.521
2.865
347-369


AD-558424.1
23.751
5.256
321-343


AD-558407.1
14.394
1.584
304-326


AD-558393.1
14.017
1.825
254-276


AD-558378.1
18.296
0.945
219-241


AD-558361.1
15.926
1.921
202-224


AD-558312.1
27.703
7.359
153-175









Example 6. Combinations of dsRNA Agents Targeting Combinations of Complement Components

In order to determine whether hemolytic activity can be strongly suppressed using a combination of a dsRNA agent targeting complement component C3 (C3) and a dsRNA agent targeting complement component C5 (C5) or a dsRNA agent targeting complement component factor B (CFB) as compared to use of a single dsRNA targeting C3, C5, or CFB alone, in vitro double reconstitution studies were performed.


Briefly, in vitro complement combination modeling was conducted using sera depleted of two complement components and adding individual proteins back at various concentrations. All reagents were purchased from Complement Technology (Tyler, Texas), unless otherwise stated. An Alternative Hemolysis (AH) assay was performed by reconstituting C3 and CFB Complement component depleted human sera with a range of concentrations of C3 and CFB protein. Ten percent reconstituted serum was added to GVBE and 5 mM MgEGTA with 25% rabbit erythrocytes (Er). Samples were incubated at 37° C. for one hour, with shaking. Hemolysis was stopped with addition of GVBE at a 1:1 ratio to samples. Samples were centrifuged, supernatants were transferred, and absorbance measured at 541 nm. Hemolytic activity was calculated by subtracting negative control sample and normalizing to positive control samples, where negative control was buffer and Er only and positive control was water and Er only.


The results of dual targeting of C3 and CFB are depicted in FIG. 2A. In particular, the hemolytic activity (alternative hemolysis, AH) in human sera depleted of C3 and CFB is shown as a heatmap with higher hemolysis levels being medium gray (top left corner, “normal range”) and lower being darker gray. The concentration of C3 was plotted on the Y axis and the concentration of CFB on the X axis.


Normal levels of CFB and C3 yielded full hemolytic activity. Decreasing either C3 or CFB level decreased hemopytic activity. siRNA administration in non-human promates showed that C3 levels could be suppressed to 200 μg/ml levels; a similar reduction is expected in human sera. Assuming 200 μg/ml as the initial level of C3, FIG. 2A demonstrates that suppression of CFB to about 40 μg/ml (about 80% silencing) reduces hemolytic activity to levels below 10%. Therefore, dual targeting of C3 and CFB can achieve near-complete suppression of AH.


It should be noted that CFB suppression does not impact Classical Hemolysis (CH), therefore no combination data was generated for CH.



FIG. 2B depicts the results of dual dose response of C3 and C5 in depleted serum reconstituted with varying levels of C3 and C5 and assayed for AH as described above. C3 was plotted on the Y-axis and C5 on the X-axis. Normal levels of both C3 and C5 produced high levels of hemolytic activity; decreasing C5 levels lowered the AH. A dose response for C3 was also observed. Although it is known that administration of a dsRNA agent targeting C5, cemdisiran, to human subjects can achieve C5 silencing down to the range of about 1-3 μg/ml, further decrease in hemolytic activity can be achieved by concurrently decreasing the level of C3 protein.


The effect of dual targeting C3 and C5 on classical hemolytic activity was also determined using a Classical Hemolysis (CH) assay. C3 and C5 depleted human sera was reconstituted with a range of concentrations of C3 and C5 proteins. Reconstituted serum (0.7%) was added to GVB++ with 13.4% antibody sensitized sheep erythrocytes (EA). Samples were incubated at 37° C. for one hour, with shaking. Samples were centrifuged, supernatants were transferred, and absorbance measured at 541 nm Hemolytic activity was calculated by subtracting negative control sample and normalizing to positive control samples, where negative control was buffer and Er only and positive control was water and Er only.



FIG. 2C depicts the results of C3 and C5 CH reconstitution experiments; it demonstrates that targeting both C3 and C5 resulted in benefit on CH. The observed effect of C3 suppression on CH activity when C5 is ≤3 μg/ml could not by resolved by determining the level of active C5b-9 formation to assess classical hemolysis activity using the Wieslab® Complement Classical Pathway (CCP) assay (FIG. 2D). The CCP assay (Wieslab® COMPL CP310, IBL America) was conducted according to manufacturer's protocol. Briefly, in vitro complement combination modeling was conducted using sera depleted of two complement components and adding individual proteins back at various concentrations. C3 and C5 Complement component depleted human sera was reconstituted with a range of concentrations of C3 and C5 protein. Reconstituted depleted sera were diluted to a final sera concentration of 1:101. Samples were added to wells and incubated for 1 hour at 37° C. Plates were washed three times and then 100 μl of conjugate solution was added to each well. Plates were incubated for 30 minutes at room temperature then washed three times. Substrate solution (100 μl) was added to each well and incubated for 30 minutes at room temperature. Reaction was stopped with 100 μl of 5 mM EDTA and absorbance was read at 405 nm. Activity was calculated by subtracting blank control from all values and then normalizing to positive control.


The ability of a combination of a dsRNA agent targeting complement component C3 (C3) and a dsRNA agent targeting complement component C5 (C5) or complement component factor B (CFB) to further suppress hemolytic activity as compared to use of a single dsRNA targeting C3, C5, or CFB alone was also assessed in vivo in non-human primates (NHPs), cynomologus monkeys (Macaca fascicularis).


Cynomologus monkeys were subcutaneously administered a single 6 mg/kg dose of a dsRNA agent targeting C3; or a dsRNA agent targeting CFB; or a dsRNA agent targeting C5; or a single 6 mg/kg dose of a dsRNA agent targeting C3 and 6 mg/kg dose of a dsRNA agent targeting CFB; or a single 6 mg/kg dose of a dsRNA agent targeting C3 and 6 mg/kg dose of a dsRNA agent targeting C5; or a single 6 mg/kg dose of a dsRNA agent targeting CFB and 6 mg/kg dose of a dsRNA agent targeting C5 on Day 1. The study design in depicted in the Table below.
























Number







Dose
Dose
of Male
Dose
End of


Group
Target(s)
Test Article
Route
(mg/kg)
Animals
Regimen
Study






















2
C3
AD-570714
SubQ
6
3
Day 1
Day 29


3
CFB
AD-560018
SubQ
6
3
Day 1
Day 29


4
C5
AD-61679
SubQ
6
3
Day 1
Day 29


5
C3
AD-570714
SubQ
6
3
Day 1
Day 29



CFB
AD-560018

6





6
C3
AD-570714
SubQ
6
3
Day 1
Day 29



C5
AD-61679

6





7
CFB
AD-560018
SubQ
6
3
Day 1
Day 29



C5
AD-61679

6









On Day −6, 1 pre-dose, and Days 8, 15, 22, and 29 post-dose, serum samples were obtained from the NHPs and the levels of C3, C5, and CFB protein and the alternative and classical hemolytic activities and the Wieslab® CAP and CCP activities were determined.


The level of C3 protein was determined by ELISA. Briefly, C3 protein was measured by a cynomolgus cross-reactive ELISA (C3 Human ELISA, Hycult HK366) according to the manufacturer's protocol. Serum was diluted by 1:40,000. C3 levels were normalized to individual animal's pre-dose levels to determine percent of C3 remaining.


The level of C5 protein was also determined by ELISA. Briefly, C5 was measured by a cynomolgus cross-reactive ELISA (Human Complement C5 ELISA Kit, Abcam ab125963) according to the manufacturer's protocol. Serum was diluted by 1:20,000 for pre-dose and day 8 samples and 1:5,000 for silenced samples days 12, 22, and 29. C5 levels were normalized to individual animal's pre-dose levels to determine % C5 remaining.


Serum CFB was measured at 1:20 dilution by quantitative analysis of western blot, using 4-12% Bis-Tris gels and imaged on Li-Cor Odyssey CLx. (1°: ProteinTech 10170-1-AP 1:50, 2°: Goat anti-Rabbit HRP).


The alternative and classical hemolytic activities were determined. The NHP alternative hemolysis was performed as briefly described. Serum (5.6%) was added to GVB° (Complement Technology, Tyler, Texas) and 5 mM MgEGTA with 25% rabbit erythrocytes (Er, Complement Technology, Tyler, Texas). Samples were incubated at 37° C. for 1 hour, with shaking. Hemolysis was stopped with addition of GVBE (Complement Technology, Tyler, Texas) at a 1:1 ratio to samples. Samples were centrifuged, supernatants were transferred, and absorbance measured at 541 nm. Hemolytic activity was calculated by subtracting negative control sample and normalizing to positive control samples, where negative control was buffer and Er only and positive control was water and Er only. Then individual animal samples were normalized to their average pre-dose samples. The NHP classical pathway hemolysis was preformed as briefly described. 1.77% serum was added to GVB++ (Complement Technology, Tyler, Texas) with 13.4% antibody sensitized sheep erythrocytes (EA, Complement Technology, Tyler, Texas). Samples were incubated at 37° C. for 1 hour, with shaking. Samples were centrifuged, supernatants were transferred, and absorbance measured at 541 nm. Hemolytic activity was calculated by subtracting negative control sample and normalizing to positive control samples, where negative control was buffer and Er only and positive control was water and EA only. Then individual animal samples were normalized to their average pre-dose samples.


The level of active C5b-9 formation to assess alternative hemolysis activity and classical hemolysis activity was also determined using the Wieslab® Complement Classical Pathway (CCP) assay as described above and the Wieslab® Complement Alternative Pathway (CAP) assay. The CAP assay (COMPL AP330 RUO, IBL America) was conducted according to manufacturer's protocol. Sera were diluted to a final sera concentration of 1:18. Samples were added to wells and incubated for 1 hour at 37° C. Plates were washed 3 times and then 100 μl of conjugate solution was added to each well. Plates were incubated for 30 minutes at room temperature then washed 3 times. Substrate solution (100 μl) was added to each well and incubated for 30 minutes at room temperature. Reaction was stopped with 100 μl of 5 mM EDTA and absorbance was read at 405 nm. Activity was calculated by subtracting blank control from all values and then normalizing to positive control. For both CAP and CCP activity values were then normalized to individual animals' average pre-dose.


The results of these assays are depicted in FIGS. 3A-3E. In particular, FIG. 3A demonstrates that a single 6 mg/kg dose of a dsRNA agent targeting C3 suppressed C3 protein up to 90% (˜110 μg/ml) in serum. As expected, silencing CFB with a dsRNA agent targeting CFB caused C3 protein levels to slightly increase. FIG. 3A also demonstrates that silencing C5 with a dsRNA agent targeting C5 left C3 protein levels unaffected and that neither a dsRNA agent targeting C3 nor a dsRNA agent targeting CFB affected C5 protein levels.



FIG. 3B demonstrates that a single 6 mg/kg dose of a dsRNA agent targeting C3 or a single 6 mg/kg dose of a dsRNA agent targeting CFB showed similar suppression of alternative hemolysis (about 60% suppression) and the combination of a single 6 mg/kg dose of a dsRNA agent targeting C3 and a 6 mg/kg dose of a dsRNA agent targeting CFB suppressed alternative hemolysis by about 90%. FIG. 3C demonstrates that the combination of a single 6 mg/kg dose of a dsRNA agent targeting C3 and 6 mg/kg dose of a dsRNA agent targeting C5 have the greatest impact on classical hemolysis.


As depicted in FIG. 3D, using the Wieslab® Complement alternative Pathway (CAP) assay, silencing either CFB or C3 or silencing CFB and C3, CFB and C5, and C3 and C5 inhibited alternative pathway activity. Silencing C5 alone had an intermediate effect in the Wieslab® CAP assay.



FIG. 3E demonstrates that C3 or CFB suppression did not confer a benefit on silencing classical pathway activity beyond that of C5 silencing.


In summary, the combination of a dsRNA agent targeting C3 and a dsRNA agent targeting CFB effectively suppressed alternative hemolysis activity to less than about 10%, while the combination of a dsRNA agent targeting C3 and a dsRNA agent targeting C5 effectively suppresses classical hemolysis activity.


EQUIVALENTS

Those skilled in the art will recognize, or be able to ascertain using no more than routine experimentation, many equivalents to the specific embodiments and methods described herein. Such equivalents are intended to be encompassed by the scope of the following claims.

Claims
  • 1. A double stranded ribonucleic acid (dsRNA) for inhibiting expression of complement factor B (CFB) in a cell, (a) wherein said dsRNA comprises a sense strand and an antisense strand forming a double stranded region, wherein the antisense strand comprises a region of complementarity to an mRNA encoding CFB, and wherein the region of complementarity comprises at least 15 contiguous nucleotides differing by no more than 3 nucleotides from any one of the antisense nucleotide sequences in any one of Tables 2-7, 13, 16, 19 and 20; or(b) wherein said dsRNA comprises a sense strand and an antisense strand forming a double stranded region, wherein the sense strand comprises at least 15 contiguous nucleotides differing by no more than three nucleotides from any one of the nucleotide sequence of nucleotides 633-665, 1133-1185, 1133-1173, 1133-1167, 1143-1173, 1540-1563, 1976-2002, 2386-2438, 2386-2418, 2386-2413, and 2389-1418 of SEQ ID NO: 1, and the antisense strand comprises at least 15 contiguous nucleotides from the corresponding nucleotide sequence of SEQ ID NO:8, where a substitution of a T with a U in either SEQ ID NO: 1 or SEQ ID NO: 8 does not count as a difference; or(c) wherein said dsRNA comprises a sense strand and an antisense strand forming a double stranded region, wherein the sense strand comprises at least 15 contiguous nucleotides differing by no more than three nucleotides from any one of the nucleotide sequence of nucleotides 633-655, 643-665, 928-950, 1133-1155, 1140-1162, 1141-1163, 1143-1165, 1145-1167, 1148-1170, 1150-1172, 1151-1173, 1185-1207, 1306-1328, 1534-1556, 1540-1562, 1541-1563, 1976-1998, 1979-2001, 1980-2002, 2078-2100, 2386-2408, 2388-2410, 2389-2411, 2391-2413, 2393-2415, 2395-2417, 2396-2418, 2438-2460, and 2602-2624 of SEQ ID NO: 1, and the antisense strand comprises at least 15 contiguous nucleotides from the corresponding nucleotide sequence of SEQ ID NO:8, where a substitution of a T with a U in either SEQ ID NO: 1 or SEQ ID NO: 8 does not count as a difference; or(d) wherein said dsRNA comprises a sense strand and an antisense strand forming a double stranded region, wherein the sense strand comprises at least 15 contiguous nucleotides differing by no more than three nucleotides from any one of the nucleotide sequence of nucleotides 153-175; 202-224; 219-241; 254-276; 304-326; 321-343; 347-369; 402-424; 418-440; 447-469; 491-513; 528-550; 549-571; 566-588; 591-613; 792-814; 819-841; 967-989; 1042-1064; 1234-1256; 1250-1272; 1269-1291; 1335-1357; 1354-1376; 1372-1394; 1422-1444; 1496-1518; 1670-1692; 1716-1738; 1757-1779; 1774-1796; 1793-1815; 1844-1866; 1871-1893; 1909-1931; 1924-1947; 1947-1969; 2161-2183; 2310-2332; 2330-2352; 2355-2377; 2494-2516; and 2527-2549 of SEQ ID NO; 1, and the antisense strand comprises at least 15 contiguous nucleotides from the corresponding nucleotide sequence of SEQ ID NO:8, where a substitution of a T with a U in either SEQ ID NO: 1 or SEQ ID NO: 8 does not count as a difference.
  • 2-6. (canceled)
  • 7. The dsRNA agent of claim 1, wherein at least one nucleotide of the dsRNA agent comprises a nucleotide modification.
  • 8. (canceled)
  • 9. The dsRNA agent of claim 1, wherein all of the nucleotides of the sense strand comprise a modification; all of the nucleotides of the antisense strand comprise a modification; or all of the nucleotides of the sense strand and all of the nucleotides of the antisense strand comprise a modification.
  • 10. The dsRNA agent of claim 7, wherein at least one of the nucleotide modifications is selected from the group consisting of a deoxy-nucleotide modification, a 3′-terminal deoxythimidine (dT) nucleotide modification, a 2′-O-methyl nucleotide modification, a 2′-fluoro nucleotide modification, a 2′-deoxy-nucleotide modification, a locked nucleotide modification, an unlocked nucleotide modification, a conformationally restricted nucleotide modification, a constrained ethyl nucleotide modification, an abasic nucleotide modification, a 2′-amino-nucleotide modification, a 2′-O-allyl-nucleotide modification, 2′-C-alkyl-nucleotide modification, a 2′-methoxyethyl nucleotide modification, a 2′-O-alkyl-nucleotide modification, a morpholino nucleotide modification, a phosphoramidate modification, a non-natural base comprising nucleotide modification, a tetrahydropyran nucleotide modification, a 1,5-anhydrohexitol nucleotide modification, a cyclohexenyl nucleotide modification, a nucleotide comprising a phosphorothioate group modification, a nucleotide comprising a methylphosphonate group modification, a nucleotide comprising a 2′-phosphate modification, a nucleotide comprising a 5′-phosphate modification, a nucleotide comprising a 5′-phosphate mimic modification, a thermally destabilizing nucleotide modification, a glycol modified nucleotide (GNA) modification, and a 2-O—(N-methylacetamide) modified nucleotide modification; and combinations thereof.
  • 11.-14. (canceled)
  • 15. The dsRNA agent of claim 1, wherein the double stranded region is 19-30 nucleotide pairs in length.
  • 16.-19. (canceled)
  • 20. The dsRNA agent of claim 1, wherein each strand is independently no more than 30 nucleotides in length.
  • 21.-24. (canceled)
  • 25. The dsRNA agent of claim 1, wherein at least one strand comprises a 3′ overhang of at least 1 nucleotide; or at least one strand comprises a 3′ overhang of at least 2 nucleotides.
  • 26. (canceled)
  • 27. The dsRNA agent of claim 1, further comprising a ligand.
  • 28. The dsRNA agent of claim 27, wherein the ligand is conjugated to the 3′ end of the sense strand of the dsRNA agent.
  • 29. The dsRNA agent of claim 27, wherein the ligand is an N-acetylgalactosamine (GalNAc) derivative.
  • 30. The dsRNA agent of claim 27, wherein the ligand is one or more GalNAc derivatives attached through a monovalent, bivalent, or trivalent branched linker.
  • 31. The dsRNA agent of claim 29, wherein the ligand is
  • 32. The dsRNA agent of claim 31, wherein the dsRNA agent is conjugated to the ligand as shown in the following schematic
  • 33. The dsRNA agent of claim 32, wherein the X is O.
  • 34. The dsRNA agent of claim 1, wherein the dsRNA agent further comprises at least one phosphorothioate or methylphosphonate internucleotide linkage.
  • 35.-43. (canceled)
  • 44. An isolated cell containing the dsRNA agent of claim 1.
  • 45. A pharmaceutical composition for inhibiting expression of a gene encoding complement factor B (CFB) comprising the dsRNA agent of claim 1.
  • 46.-50. (canceled)
  • 51. An in vitro method of inhibiting expression of a complement factor B (CFB) gene in a cell, the method comprising contacting the cell with the dsRNA agent of claim 1, thereby inhibiting expression of the CFB gene in the cell.
  • 52.-56. (canceled)
  • 57. A method of treating a subject having a disorder that would benefit from reduction in complement factor B expression, comprising administering to the subject a therapeutically effective amount of the dsRNA agent of claim 1, thereby treating the subject having the disorder that would benefit from reduction in complement factor B expression.
  • 58. (canceled)
  • 59. The method of claim 57, wherein the disorder is a complement factor B-associated disorder.
  • 60.-72. (canceled)
RELATED APPLICATIONS

This application is a 35 § U.S.C. 111(a) continuation application which claims the benefit of priority to PCT/US2021/029872, filed on Apr. 29, 2021, which, in turn, claims the benefit of priority to U.S. Provisional Application No. 63/017,725, filed on Apr. 30, 2020, U.S. Provisional Application No. 63/119,009, filed on Nov. 30, 2020, and U.S. Provisional Application No. 63/157,899, filed on Mar. 8, 2021. The entire contents of each of the foregoing applications are incorporated herein by reference.

Provisional Applications (3)
Number Date Country
63157899 Mar 2021 US
63119009 Nov 2020 US
63017725 Apr 2020 US
Continuations (1)
Number Date Country
Parent PCT/US2021/029872 Apr 2021 US
Child 17974598 US