CN117203338A - Transmembrane protease serine 6 (TMPRSS 6) IRNA compositions and methods of use thereof - Google Patents

Transmembrane protease serine 6 (TMPRSS 6) IRNA compositions and methods of use thereof Download PDF

Info

Publication number
CN117203338A
CN117203338A CN202280030748.9A CN202280030748A CN117203338A CN 117203338 A CN117203338 A CN 117203338A CN 202280030748 A CN202280030748 A CN 202280030748A CN 117203338 A CN117203338 A CN 117203338A
Authority
CN
China
Prior art keywords
nucleotides
dsrna agent
ome
antisense strand
seq
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
CN202280030748.9A
Other languages
Chinese (zh)
Inventor
A·M·迪顿
J·M·甘斯纳尔
J·D·麦金因克
M·K·施莱格尔
B·P·加芬克尔
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Alnylam Pharmaceuticals Inc
Original Assignee
Alnylam Pharmaceuticals Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Alnylam Pharmaceuticals Inc filed Critical Alnylam Pharmaceuticals Inc
Priority claimed from PCT/US2022/026097 external-priority patent/WO2022231999A1/en
Publication of CN117203338A publication Critical patent/CN117203338A/en
Pending legal-status Critical Current

Links

Landscapes

  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The present application relates to RNAi agents, e.g., double-stranded RNA (dsRNA) agents, targeting the transmembrane protease serine 6 (TMPRSS 6) gene. The application also relates to methods of inhibiting expression of TMPRSS6 genes using such RNAi agents, and to methods of preventing and treating TMPRSS 6-related disorders, e.g., disorders associated with iron overload and/or disorders of ineffective erythropoiesis, e.g., hereditary hemochromatosis, beta-thalassemia (e.g., severe beta-thalassemia and intermediate beta-thalassemia), polycythemia vera, myelodysplastic syndrome, congenital erythropoiesis anemia, pyruvate kinase deficiency, erythropoiesis porphyria, parkinson's Disease, alzheimer's Disease, or Friedreich's Ataxia.

Description

Transmembrane protease serine 6 (TMPRSS 6) IRNA compositions and methods of use thereof
Cross reference to related applications
The present application claims the benefit of priority from U.S. provisional application No. 63/179,607, filed on month 26 of 2021, and U.S. provisional application No. 63/278,227, filed on month 11 of 2021. The entire contents of each of the foregoing applications are incorporated herein by reference.
Sequence listing
The present application contains a sequence listing that has been electronically submitted in ASCII format and is incorporated herein by reference in its entirety. The ASCII copy created at month 18 of 2022 is named 121301_15420_sl.txt and is 673,291 bytes in size.
Background
TMPRSS6 (transmembrane protease serine 6), also known as Ma Qu Tase-2, is a type II serine protease. It is expressed predominantly in the liver, although high levels of TMPRSS6mRNA are also found in the kidney, with lower levels in the uterus and much smaller amounts detected in many other tissues (Beliveau et al, 2019, cytochemistry biology (Cell Chemical Biology) 26, 1559-1572). TMPRSS6 plays a key role in iron homeostasis by regulating hepcidin expression. Hepcidin is a liver-derived peptide hormone, known as a central regulator of systemic iron homeostasis, whose imbalance is produced to contribute to the onset of a range of iron disorders. Hepcidin works by blocking intestinal absorption of dietary iron and release of iron by macrophages and hepatocytes (Ganz t.2011, blood, volume 117, 17, 4425-4433). Hepcidin gene expression can be stimulated in response to iron by BMP/SMAD dependent signal transduction cascades mediated by BMP co-receptor Hemojuvelin (HJV). TMPRSS6 inhibits BMP-mediated upregulation of hepcidin by cleaving BMP co-receptor HJV, thereby preventing BMP signaling, SMAD transfer to the nucleus and hepcidin transcriptional activation, which down-regulates hepcidin levels (fiberg, k.e. et al 2010, blood 115,3817-3826; wang, c.y. et al, 2014 pharmacological front (front. Pharmacol.) 5,114).
Thus, inhibition of TMPRSS6 increases hepcidin levels, making it an attractive pharmacological target for conditions associated with iron overload and inappropriate low hepcidin or conditions where iron limitation is desired. Many disorders, such as thalassemia, hemochromatosis, and certain types of myelodysplastic syndromes (MDS), are associated with iron overload, a condition characterized by increased levels of iron. Iron overload can cause excessive iron deposition in various tissues and can ultimately lead to tissue and organ damage. Additionally, iron limitation is desirable in certain conditions such as polycythemia vera.
Current treatments for conditions associated with iron overload and conditions where iron limitation is desired (e.g., polycythemia vera) include phlebotomy (phlebotomy) or phlebotomy (venesection), a treatment that removes iron-rich blood from the body; splenectomy; iron chelation therapy; and (5) diet. However, these treatments are not always effective. Accordingly, there is a need in the art for alternative treatments for subjects suffering from conditions associated with iron overload.
Disclosure of Invention
The present invention provides iRNA compositions that affect RNA-induced silencing complex (RISC) -mediated cleavage of an RNA transcript of a gene encoding a transmembrane protease serine 6 (TMPRSS 6). The TMPRSS6 gene may be within a cell, e.g., a cell in a subject (e.g., a human subject). The invention also provides methods for using the iRNA compositions of the invention for inhibiting expression of a TMPRSS6 gene and/or for treating a subject who would benefit from inhibiting or reducing expression of a TMPRSS6 gene, e.g., a subject suffering from or susceptible to a TMPRSS 6-related disorder, e.g., an iron overload-related disorder and/or a disorder that does not have erythropoiesis, such as thalassemia, e.g., β -thalassemia, hemochromatosis, myelodysplastic syndrome (MDS), or polycythemia vera.
Accordingly, in one aspect, the invention provides a double stranded ribonucleic acid (dsRNA) for inhibiting expression of a transmembrane protease serine 6 (TMPRSS 6) in a cell, wherein the dsRNA comprises a sense strand and an antisense strand forming a double stranded region, wherein the antisense strand comprises a region complementary to an mRNA encoding TMPRSS6, and wherein the region of complementarity comprises at least 15, e.g. 15, 16, 17, 18, 19 or 20 consecutive 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.
In one embodiment, the dsRNA agent comprises a sense strand comprising a contiguous nucleotide sequence having at least 85%, e.g., 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% nucleotide sequence identity over its entire length to any of the nucleotide sequences of the sense strand in any of tables 2-7 and an antisense strand comprising a contiguous nucleotide sequence having at least 85%, e.g., 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% nucleotide sequence identity over its entire length to any of the nucleotide sequences of the antisense strand in any of tables 2-7.
In one embodiment, the dsRNA agent comprises a sense strand comprising at least 15, e.g., 15, 16, 17, 18, 19, 20, or 21 consecutive nucleotides differing by no more than three nucleotides from any of the nucleotide sequences of the sense strand in any of tables 2-7, and an antisense strand comprising at least 15, e.g., 15, 16, 17, 18, 19, 20, or 21 consecutive nucleotides differing by no more than three nucleotides from any of the nucleotide sequences of the antisense strand in any of tables 2-7.
In one embodiment, the dsRNA agent comprises a sense strand comprising at least 15, e.g., 15, 16, 17, 18, 19, 20, or 21 consecutive nucleotides differing by no more than two nucleotides from any of the nucleotide sequences of the sense strand in any of tables 2-7, and an antisense strand comprising at least 15, e.g., 15, 16, 17, 18, 19, 20, or 21 consecutive nucleotides differing by no more than two nucleotides from any of the nucleotide sequences of the antisense strand in any of tables 2-7.
In one embodiment, the dsRNA agent comprises a sense strand comprising at least 15, e.g., 15, 16, 17, 18, 19, 20, or 21 consecutive nucleotides differing by no more than one nucleotide from any of the nucleotide sequences of the sense strand in any of tables 2-7, and an antisense strand comprising at least 15, e.g., 15, 16, 17, 18, 19, 20, or 21 consecutive nucleotides differing by no more than one nucleotide from any of the nucleotide sequences of the antisense strand in any of tables 2-7.
In one embodiment, the dsRNA agent comprises or consists of a sense strand comprising or consisting of a nucleotide sequence selected from the group consisting of any one of the nucleotide sequences of the sense strand in any one of tables 2-7 and an antisense strand comprising or consisting of a nucleotide sequence selected from the group consisting of any one of the nucleotide sequences of the antisense strand in any one of tables 2-7.
In one aspect, the invention provides a double-stranded ribonucleic acid (dsRNA) for inhibiting expression of a transmembrane protease serine 6 (TMPRSS 6) in a cell, wherein the dsRNA comprises a sense strand and an antisense strand forming a double-stranded region, wherein the sense strand comprises at least 15, e.g. 15, 16, 17, 18, 19, 20 or 21 consecutive nucleotides differing by NO more than three, e.g. 3, 2, 1 or 0 nucleotides from any of the nucleotide sequences of the following nucleotides of SEQ ID NO: 1: 187-210;227-254;322-363;362-390;398-420;404-429;410-435;439-461;443-467;448-474;460-483;466-488;496-519;519-542;526-548;557-593;641-671;652-676;687-713;725-762;757-794;886-908;921-951;956-987;1051-1082;1233-1269;1279-1313;1313-1341;1327-1351;1415-1439;1447-1480;1464-1486;1486-1509;1559-1589;1571-1595;1579-1609;1707-1735;1738-1764;1806-1828;1864-1886;1934-1966;1967-1991;2008-2031;2015-2043;2042-2072;2287-2311;2297-2354;2336-2361;2360-2384;2416-2438;2481-2510;2496-2527;2526-2558;2665-2693;2693-2719;2707-2729;2799-2821;2851-2874;2971-2999;2981-3006 and 3155-3195, and the antisense strand comprises at least 15 consecutive nucleotides differing by NO more than 3, e.g. 3, 2, 1 or 0 nucleotides from the corresponding nucleotide sequence of SEQ ID No. 2.
In one aspect, the invention provides a double-stranded ribonucleic acid (dsRNA) agent for inhibiting expression of a transmembrane protease serine 6 (TMPRSS 6) 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, e.g. 15, 16, 17, 18, 19, 20 or 21 consecutive nucleotides differing by NO more than three, e.g. 3, 2, 1 or 0 nucleotides from any of the nucleotide sequences of the following nucleotides of SEQ ID NO: 1: 230-252, 324-346, 560-578, 560-582, 2338-2360, 3163-3185, 3169-3191 and 3172-3194, and the antisense strand comprises at least 15, e.g. 15, 16, 17, 18, 19, 20 or 21 consecutive nucleotides differing by NO more than three, e.g. 3, 2, 1 or 0 nucleotides from the corresponding nucleotide sequence of SEQ ID No. 2.
In one aspect, the invention provides a double-stranded ribonucleic acid (dsRNA) agent for inhibiting expression of a transmembrane protease serine 6 (TMPRSS 6) 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, e.g. 15, 16, 17, 18, 19, 20 or 21 consecutive nucleotides differing by NO more than three, e.g. 3, 2, 1 or 0 nucleotides from any of the nucleotide sequences of nucleotides 560-578, 2338-2360 and 3169-3191 of SEQ ID NO:1, and the antisense strand comprises at least 15, e.g. 15, 16, 17, 18, 19, 20 or 21 consecutive nucleotides differing by NO more than 3, e.g. 3, 2, 1 or 0 nucleotides from the corresponding nucleotide sequence of SEQ ID NO: 2.
In some embodiments, the antisense strand comprises at least 15, e.g., 15, 16, 17, 18, 19, 20, or 21 consecutive nucleotides that differ by no more than three, e.g., 3, 2, 1, or 0 nucleotides from any of the antisense strand nucleotide sequences of the duplex selected from the group consisting of: AD-1556360, AD-1571158, AD-1571033, AD-1554875, AD-1571160, AD-1555117, AD-1554911 and AD-1556915.
In some embodiments, the antisense strand comprises at least 15, e.g., 15, 16, 17, 18, 19, 20, or 21 consecutive nucleotides that differ by no more than three, e.g., 3, 2, 1, or 0 nucleotides from any of the antisense strand nucleotide sequences of the duplex selected from the group consisting of: AD-1556360, AD-1571158 and AD-1571033.
In one embodiment, the dsRNA agent comprises at least one modified nucleotide.
In one embodiment, substantially all of the nucleotides of the sense strand comprise modifications; substantially all of the nucleotides of the antisense strand comprise modifications; or substantially all nucleotides of the sense strand and substantially all nucleotides of the antisense strand comprise modifications.
In one embodiment, all nucleotides of the sense strand comprise modifications; all nucleotides of the antisense strand include modifications; or all nucleotides of the sense strand and all nucleotides of the antisense strand comprise modifications.
In one embodiment, at least one of the modified nucleotides is selected from the group consisting of: deoxynucleotides, 3' -terminal deoxythymine (dT) nucleotides, 2' -O-methyl modified nucleotides, 2' -fluoro modified nucleotides, 2' -deoxymodified nucleotides, locked nucleotides, unlocked nucleotides, conformationally restricted nucleotides, constrained ethyl nucleotides, abasic nucleotides, 2' -amino modified nucleotides, 2' -O-allyl modified nucleotides, 2' -C-alkyl modified nucleotides, 2' -hydroxy modified nucleotides, 2' -methoxyethyl modified nucleotides, 2' -O-alkyl modified nucleotides, morpholino nucleotides, phosphoramidates, non-natural bases including nucleotides, tetrahydropyran modified nucleotides, 1, 5-anhydrohexitol modified nucleotides, cyclohexenyl modified nucleotides, nucleotides including phosphorothioate groups, nucleotides including methylphosphonate groups, nucleotides including 5' -phosphate esters, nucleotides including 5' -phosphate ester mimetics, thermally labile nucleotides, ethylene glycol modified nucleotides (GNA), nucleotides including 2' phosphate esters, and 2-O- (N-methyl) acetamides; and combinations thereof.
In one embodiment, the modification on the nucleotide is 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' -hydroxy and ethylene glycol; and combinations thereof.
In one embodiment, at least one of the modified nucleotides is selected from the group consisting of: deoxynucleotides, 2 '-O-methyl modified nucleotides, 2' -fluoro modified nucleotides, 2 '-deoxymodified nucleotides, ethylene glycol modified nucleotides (GNAs), e.g., ggn, cgn, tgn or Agn, nucleotides with 2' phosphates, e.g., G2p, C2p, A2p or U2p, and vinyl phosphonate nucleotides; and combinations thereof.
In some embodiments, the modified nucleotide comprises a short sequence of 3' terminal deoxythymidines (dT).
In some embodiments, the dsRNA agent further comprises at least one phosphorothioate internucleotide linkage. In some embodiments, the dsRNA agent comprises 6 to 8 phosphorothioate internucleotide linkages. In one embodiment, the internucleotide linkage of the phosphorothioate or methylphosphonate is located at the 3' end of one strand. Optionally, the strand is the antisense strand. In another embodiment, the strand is the sense strand. In a related embodiment, the phosphorothioate or methylphosphonate internucleotide linkage is located at the 5' end of one strand. Optionally, the strand is the antisense strand. In another embodiment, the strand is the sense strand. In another embodiment, the phosphorothioate or methylphosphonate internucleotide linkages are located at both the 5 'and 3' ends of one strand. Optionally, the strand is the antisense strand. In another embodiment, the strand is the sense strand.
The double-stranded region may be 19 to 30 nucleotide pairs in length; 19 to 25 nucleotide pairs in length; 19 to 23 nucleotide pairs in length; 23 to 27 nucleotide pairs in length; or 21 to 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 complementary region may be at least 17 nucleotides in length; 19 to 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 conjugated to the 5' 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 linked by a monovalent, divalent or trivalent branched linker.
In one embodiment, the ligand is
In one embodiment, the dsRNA agent is conjugated to the ligand, as shown in the following schematic
And wherein X is O or S.
In one embodiment, the X is O.
In one embodiment, the dsRNA agent is conjugated to the ligand, as shown in the following schematic
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 located on one strand, for example at the 3' terminus of the antisense strand or sense strand.
In another embodiment, the phosphorothioate or methylphosphonate internucleotide linkage is located on one strand, for example at the 5' end of the antisense strand or sense strand.
In one embodiment, the phosphorothioate or methylphosphonate internucleotide linkages are located at both the 5 'and 3' ends of one strand. In one embodiment, the strand is an antisense strand.
In one embodiment, the base pair at position 1 of the 5' -end of the antisense strand of the duplex is an AU base pair.
In one embodiment, the sense strand comprises at least 17 consecutive nucleotides that differ from the nucleotide sequence of 5'-GACGCCACGCAUGCUGUGUGU-3' (SEQ ID NO: 119) by NO more than 0, 1, 2 or 3 nucleotides.
In one embodiment, the sense strand comprises at least 19 consecutive nucleotides that differ from the nucleotide sequence of 5'-GACGCCACGCAUGCUGUGUGU-3' (SEQ ID NO: 119) by NO more than 0, 1, 2 or 3 nucleotides.
In one embodiment, the sense strand comprises or consists of the nucleotide sequence of 5'-GACGCCACGCAUGCUGUGUGU-3' (SEQ ID NO: 119).
In one embodiment, the antisense strand comprises at least 17 consecutive nucleotides that differ from the nucleotide sequence of 5'-ACACACAGCAUGCGUGGCGUCAC-3' (SEQ ID NO: 245) by NO more than 0, 1, 2 or 3 nucleotides.
In one embodiment, the antisense strand comprises at least 19 consecutive nucleotides that differ from the nucleotide sequence of 5'-ACACACAGCAUGCGUGGCGUCAC-3' (SEQ ID NO: 245) by NO more than 0, 1, 2 or 3 nucleotides.
In one embodiment, the antisense strand comprises at least 21 consecutive nucleotides that differ from the nucleotide sequence of 5'-ACACACAGCAUGCGUGGCGUCAC-3' (SEQ ID NO: 245) by NO more than 0, 1, 2 or 3 nucleotides.
In one embodiment, the antisense strand comprises or consists of the nucleotide sequence of 5'-ACACACAGCAUGCGUGGCGUCAC-3' (SEQ ID NO: 245).
In one embodiment, the sense strand comprises the nucleotide sequence of 5'-GACGCCACGCAUGCUGUGUGU-3' (SEQ ID NO: 119) and the antisense strand comprises the nucleotide sequence of 5'-ACACACAGCAUGCGUGGCGUCAC-3' (SEQ ID NO: 245).
In one embodiment, the sense strand differs by NO more than 3, such as 0, 1, 2 or 3 modified nucleotides from the nucleotide sequence of 5 '-gsascgccacgcfcfafafugugugu-3' (SEQ ID NO: 371), wherein a, g, c and U are 2 '-O-methyl (2' -OMe) A, G, C and U, respectively; af. Gf, cf and Uf are 2' -fluoro A, G, C and U, respectively; and s is a phosphorothioate bond.
In one embodiment, the antisense strand differs from the nucleotide sequence of 5 '-asdcsacdacacagcaudcgcgfuggcgucsasc-3' (SEQ ID NO: 497) by NO more than 3, e.g., 0, 1, 2, or 3 modified nucleotides, wherein a, g, c, and U are 2 '-O-methyl (2' -OMe) A, G, C and U, respectively; af. Gf, cf and Uf are 2' -fluoro A, G, C and U, respectively; wherein dA, dG and dC are 2 '-deoxyadenosine-3' -phosphate, 2 '-deoxyguanosine-3' -phosphate and 2 '-deoxycytidine-3' -phosphate, respectively; and s is a phosphorothioate bond.
In one embodiment, the sense strand comprises the nucleotide sequence of 5 '-gsascgccacgcfcfaffugcugugugu-3' (SEQ ID NO: 371), and the antisense strand comprises the nucleotide sequence of 5 '-asdcsacdacdacgcaudcgcgcgfggcgucasc-3' (SEQ ID NO: 497), wherein a, g, c, and U are 2 '-O-methyl (2' -OMe) A, G, C and U, respectively; af. Gf, cf and Uf are 2' -fluoro A, G, C and U, respectively; wherein dA, dG and dC are 2 '-deoxyadenosine-3' -phosphate, 2 '-deoxyguanosine-3' -phosphate and 2 '-deoxycytidine-3' -phosphate, respectively; and s is a phosphorothioate bond.
In one embodiment, the sense strand comprises the nucleotide sequence of 5 '-gsascgccacgcfcfaffugcugugul 96-3' (SEQ ID NO: 371), and the antisense strand comprises the nucleotide sequence of 5 '-asdcsacdacdacgcaudcgcgfuggcascasc-3' (SEQ ID NO: 497), wherein a, g, c, and U are 2 '-O-methyl (2' -OMe) A, G, C and U, respectively; af. Gf, cf and Uf are 2' -fluoro A, G, C and U, respectively; wherein dA, dG and dC are 2 '-deoxyadenosine-3' -phosphate, 2 '-deoxyguanosine-3' -phosphate and 2 '-deoxycytidine-3' -phosphate, respectively; s is a phosphorothioate linkage, and L96 is N- [ tris (GalNAc-alkyl) -amidodecanoyl) ] -4-hydroxyproline.
In one embodiment, the sense strand comprises the nucleotide sequence of 5 '-gsascgccacgcfcfaffugcugugugu-3' (SEQ ID NO: 371), and the antisense strand comprises the nucleotide sequence of 5 '-asdcsacdacdacgcaudcgcgcgfggcgucasc-3' (SEQ ID NO: 497), wherein a, g, c, and U are 2 '-O-methyl (2' -OMe) A, G, C and U, respectively; af. Gf, cf and Uf are 2' -fluoro A, G, C and U, respectively; wherein dA, dG and dC are 2 '-deoxyadenosine-3' -phosphate, 2 '-deoxyguanosine-3' -phosphate and 2 '-deoxycytidine-3' -phosphate, respectively; and s is a phosphorothioate linkage, wherein the 3' end of the sense strand is conjugated to the ligand, as shown in the following schematic:
And wherein X is O.
In one embodiment, the sense strand comprises at least 17 consecutive nucleotides that differ from the nucleotide sequence of 5'-CCUUUGGAAUAAAGCUGCCUU-3' (SEQ ID NO: 844) by NO more than 0, 1, 2 or 3 nucleotides.
In one embodiment, the sense strand comprises at least 19 consecutive nucleotides that differ from the nucleotide sequence of 5'-CCUUUGGAAUAAAGCUGCCUU-3' (SEQ ID NO: 844) by NO more than 0, 1, 2 or 3 nucleotides.
In one embodiment, the sense strand comprises or consists of the nucleotide sequence of 5'-CCUUUGGAAUAAAGCUGCCUU-3' (SEQ ID NO: 844).
In one embodiment, the antisense strand comprises at least 17 consecutive nucleotides that differ from the nucleotide sequence of 5'-AAGGCAGCUUUAUUCCAAAGGGC-3' (SEQ ID NO: 1868) by NO more than 0, 1, 2 or 3 nucleotides.
In one embodiment, the antisense strand comprises at least 19 consecutive nucleotides that differ from the nucleotide sequence of 5'-AAGGCAGCUUUAUUCCAAAGGGC-3' (SEQ ID NO: 1868) by NO more than 0, 1, 2 or 3 nucleotides.
In one embodiment, the antisense strand comprises at least 21 consecutive nucleotides that differ from the nucleotide sequence of 5'-AAGGCAGCUUUAUUCCAAAGGGC-3' (SEQ ID NO: 1868) by NO more than 0, 1, 2 or 3 nucleotides.
In one embodiment, the antisense strand comprises or consists of the nucleotide sequence of 5'-AAGGCAGCUUUAUUCCAAAGGGC-3' (SEQ ID NO: 1868).
In one embodiment, the sense strand comprises the nucleotide sequence of 5'-CCUUUGGAAUAAAGCUGCCUU-3' (SEQ ID NO: 844) and the antisense strand comprises the nucleotide sequence of 5'-AAGGCAGCUUUAUUCCAAAGGGC-3' (SEQ ID NO: 1868).
In one embodiment, the sense strand differs by NO more than 3, e.g., 0, 1, 2, or 3 modified nucleotides from the nucleotide sequence of 5'-cscsu ug gfafafafaagccugcuu-3' (SEQ ID NO: 2095), wherein a, g, c, and U are 2 '-O-methyl (2' -OMe) A, G, C and U, respectively; af. Gf, cf and Uf are 2' -fluoro A, G, C and U, respectively; and s is a phosphorothioate bond.
In one embodiment, the antisense strand differs from the nucleotide sequence of 5'-asAfsggdCa (G2 p) cuuauufcfaaaggsgsc-3' (SEQ ID NO: 2324) by NO more than 3, e.g., 0, 1, 2, or 3 modified nucleotides, wherein a, G, c, and U are 2 '-O-methyl (2' -OMe) A, G, C and U, respectively; af. Gf, cf and Uf are 2' -fluoro A, G, C and U, respectively; wherein G2p is guanosine-2' -phosphate; and s is a phosphorothioate bond.
In one embodiment, the sense strand comprises the nucleotide sequence of 5 '-cscsuuuggfafafaaagcugccuu-3' (SEQ ID NO: 2095), and the antisense strand comprises the nucleotide sequence of 5'-asAfsggdCa (G2 p) cuuuafaaagsgsc-3' (SEQ ID NO: 2324), wherein a, G, c, and U are 2 '-O-methyl (2' -OMe) A, G, C and U, respectively; af. Gf, cf and Uf are 2' -fluoro A, G, C and U, respectively; wherein G2p is guanosine-2' -phosphate; and s is a phosphorothioate bond.
In one embodiment, the sense strand comprises the nucleotide sequence of 5 '-cscsuuuggfafafaaagcugccul 96-3' (SEQ ID NO: 2095), and the antisense strand comprises the nucleotide sequence of 5'-asAfsggdCa (G2 p) cuuauufcfaaaggsgsc-3' (SEQ ID NO: 2324), wherein a, G, c, and U are 2 '-O-methyl (2' -OMe) A, G, C and U, respectively; af. Gf, cf and Uf are 2' -fluoro A, G, C and U, respectively; wherein G2p is guanosine-2' -phosphate; s is a phosphorothioate linkage, and L96 is N- [ tris (GalNAc-alkyl) -amidodecanoyl) ] -4-hydroxyproline.
In one embodiment, the sense strand comprises the nucleotide sequence of 5 '-cscsuuuggfafafaaagcugccuu-3' (SEQ ID NO: 2095), and the antisense strand comprises the nucleotide sequence of 5'-asAfsggdCa (G2 p) cuuuafaaagsgsc-3' (SEQ ID NO: 2324), wherein a, G, c, and U are 2 '-O-methyl (2' -OMe) A, G, C and U, respectively; af. Gf, cf and Uf are 2' -fluoro A, G, C and U, respectively; wherein G2p is guanosine-2 '-phosphate and s is a phosphorothioate linkage, and wherein the 3' end of the sense strand is conjugated to the ligand, as shown in the following schematic:
and wherein X is O.
In one embodiment, the sense strand comprises at least 17 consecutive nucleotides that differ from the nucleotide sequence of 5'-UCACCUGCUUCUUCUGGUU-3' (SEQ ID NO: 1686) by NO more than 0, 1, 2, or 3 nucleotides.
In one embodiment, the sense strand comprises or consists of the nucleotide sequence of 5'-UCACCUGCUUCUUCUGGUU-3' (SEQ ID NO: 1686).
In one embodiment, the antisense strand comprises at least 17 consecutive nucleotides that differ from the nucleotide sequence of 5'-AACCAGAAGAAGCAGGUGA-3' (SEQ ID NO: 1790) by NO more than 0, 1, 2 or 3 nucleotides.
In one embodiment, the antisense strand comprises or consists of the nucleotide sequence of 5'-AACCAGAAGAAGCAGGUGA-3' (SEQ ID NO: 1790).
In one embodiment, the sense strand comprises the nucleotide sequence of 5'-UCACCUGCUUCUUCUGGUU-3' (SEQ ID NO: 1686) and the antisense strand comprises the nucleotide sequence of 5'-AACCAGAAGAAGCAGGUGA-3' (SEQ ID NO: 1790).
In one embodiment, the sense strand differs from the nucleotide sequence of 5 '-UfcAfcCfuUfcUfuUfcUfcUfcUfgGfsusuf-3' (SEQ ID NO: 1974) by NO more than 3, e.g., 0, 1, 2, or 3 modified nucleotides, wherein a, g, c, and U are 2 '-O-methyl (2' -OMe) A, G, C and U, respectively; af. Gf, cf and Uf are 2' -fluoro A, G, C and U, respectively; and s is a phosphorothioate bond.
In one embodiment, the antisense strand differs from the nucleotide sequence of 5 '-asafscfagfafafafafafafafafcggfusgfsa-3' (SEQ ID NO:) by NO more than 3, e.g., 0, 1, 2, or 3 modified nucleotides, wherein a, g, c, and U are 2 '-O-methyl (2' -OMe) A, G, C and U, respectively; af. Gf, cf and Uf are 2' -fluoro A, G, C and U, respectively; and s is a phosphorothioate bond.
In one embodiment, the sense strand comprises the nucleotide sequence of 5 '-UfcAfcCfuUfcUfuUfcUfcUfgGfsusuf-3' (SEQ ID NO: 2203), and the antisense strand comprises the nucleotide sequence of 5 '-asafscFaAfaAfaGAfaGfGfcAfgGfsGfsa-3' (SEQ ID NO:), wherein a, g, c, and U are 2 '-O-methyl (2' -OMe) A, G, C and U, respectively; af. Gf, cf and Uf are 2' -fluoro A, G, C and U, respectively; and s is a phosphorothioate bond.
In one embodiment, the sense strand comprises the nucleotide sequence of 5'-Q191 sUfcCfuGfcUfuUfcUfcUfcUfgGfsusuf-3' (SEQ ID NO: 1974), and the antisense strand comprises the nucleotide sequence of 5 '-asafscFaAfaAfaGfAfaGfcAfGfugsGfsa-3' (SEQ ID NO: 2203), wherein a, g, c, and U are 2 '-O-methyl (2' -OMe) A, G, C and U, respectively; af. Gf, cf and Uf are 2' -fluoro A, G, C and U, respectively; s is a phosphorothioate linkage, and Q191 is N- [ tris (GalNAc-alkyl) -amidododecanoyl ] - (S) -pyrrolidin-3-ol-phosphorothioate (p-C12- (GalNAc-alkyl) 3).
In one embodiment, the sense strand comprises the nucleotide sequence of 5 '-UfcAfcCfuUfcUfuUfcUfcUfgGfsusuf-3' (SEQ ID NO: 1974), and the antisense strand comprises the nucleotide sequence of 5 '-asafscFaAfaAfaGAfaGfGfcAfgGfsGfsa-3' (SEQ ID NO: 2203), wherein a, g, c, and U are 2 '-O-methyl (2' -OMe) A, G, C and U, respectively; af. Gf, cf and Uf are 2' -fluoro A, G, C and U, respectively; and s is a phosphorothioate linkage, wherein the 5' end of the sense strand is conjugated to the ligand, as shown in the following schematic:
In another embodiment, the RNAi agent is a pharmaceutically acceptable salt thereof. The "pharmaceutically acceptable salt" of each of the RNAi agents herein includes, but is not limited to, sodium, calcium, lithium, potassium, ammonium, magnesium salts, and mixtures thereof. Those of skill in the art will appreciate that when the RNAi agent is provided in the form of a polycationic salt, the polycationic salt has one cation per free acid group of the optionally modified phosphodiester backbone and/or any other acidic modification (e.g., a 5' terminal phosphonate group). For example, an oligonucleotide of length "n" nucleotides contains n-1 optionally modified bisphosphates, so an oligonucleotide of length 21nt can be provided as a salt with up to 20 cations (e.g., 20 sodium cations). Similarly, RNAi agents having a sense strand of 21nt in length and an antisense strand of 23nt in length can be provided in the form of salts having up to 42 cations (e.g., 42 sodium cations). In the foregoing examples, wherein the RNAi agent further comprises a 5 'terminal phosphate or 5' terminal vinyl phosphonate group, the RNAi agent can be provided in the form of a salt having up to 44 cations (e.g., 44 sodium cations).
The invention also provides cells comprising 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 comprise the dsRNA agent in an unbuffered solution, e.g. saline or water, or the pharmaceutical composition of the invention may comprise the dsRNA agent in a buffered solution, e.g. a buffered solution comprising: acetate, citrate, prolamin, carbonate, or phosphate or any combination thereof; or Phosphate Buffered Saline (PBS).
In one aspect, the invention provides a method of inhibiting the expression of a transmembrane protease serine 6 (TMPRSS 6) gene in a cell. The method comprises contacting the cell with any dsRNA of the invention or any pharmaceutical composition of the invention, thereby inhibiting expression of the TMPRSS6 gene in the cell.
In one embodiment, the cells are in a subject, e.g., a human subject, e.g., a subject having a transmembrane protease serine 6 (TMPRSS 6) -related disorder, such as a disorder associated with iron overload and/or a disorder that is not erythropoietic, e.g., hereditary hemochromatosis, beta-thalassemia (e.g., severe beta-thalassemia and intermediate beta-thalassemia), polycythemia vera, myelodysplastic syndrome, congenital erythropoiesis-dysanemia, pyruvate kinase deficiency, erythropoiesis porphyria, parkinson's Disease, alzheimer's Disease, or friedrich's Ataxia.
In some embodiments, the TMPRSS 6-related disorder is β -thalassemia. In one embodiment, the TMPRSS 6-related disorder is severe beta-thalassemia. In another embodiment, the TMPRSS 6-related disorder is inter-type β -thalassemia. In some embodiments, the TMPRSS 6-related disorder is polycythemia vera.
In certain embodiments, the TMPRSS6 expression is inhibited by at least about 30%, 40%, 50%, 60%, 70%, 80%, 90%, or 95%. In one embodiment, inhibiting expression of TMPRSS6 reduces TMPRSS6 protein levels in serum of the subject by at least 30%, 40%, 50%, 60%, 70%, 80%, 90% or 95%.
In certain embodiments, contacting the cell with the dsRNA agent increases expression of hepcidin by at least 50%, 60%, 70%, 80%, 90% or 95%. In one embodiment, increasing expression of hepcidin increases hepcidin protein levels in serum of the subject by at least 50%, 60%, 70%, 80%, 90% or 95%.
In one aspect, the invention provides a method of treating a subject suffering from a disorder that would benefit from reduced expression of the transmembrane protease serine 6 (TMPRSS 6). The method comprises administering to the subject a therapeutically effective amount of any dsRNA of the invention or any pharmaceutical composition of the invention, thereby treating the subject with a disorder that would benefit from reduced expression of TMPRSS 6.
In another aspect, the invention provides a method of preventing at least one symptom in a subject suffering from a disorder that would benefit from reduced expression of the transmembrane protease serine 6 (TMPRSS 6). The method comprises administering to the subject a prophylactically effective amount of any dsRNA of the invention or any pharmaceutical composition of the invention, thereby preventing at least one symptom of the subject suffering from a disorder that would benefit from reduced expression of TMPRSS 6.
In certain embodiments, the disorder is a transmembrane protease serine 6 (TMPRSS 6) related disorder, e.g., a disorder associated with iron overload and/or a disorder that does not have erythropoiesis, e.g., hereditary hemochromatosis, β -thalassemia (e.g., severe β -thalassemia and intermediate β -thalassemia), polycythemia vera, myelodysplastic syndrome, congenital erythropoiesis anemia, pyruvate kinase deficiency, erythropoiesis porphyria, parkinson's disease, alzheimer's disease, or friedel-crafts ataxia.
In some embodiments, the TMPRSS 6-related disorder is β -thalassemia. In one embodiment, the TMPRSS 6-related disorder is severe beta-thalassemia. In another embodiment, the TMPRSS 6-related disorder is inter-type β -thalassemia. In some embodiments, the TMPRSS 6-related disorder is polycythemia vera.
In certain embodiments, administering the dsRNA to the subject results in a decrease in iron levels, ferritin levels, and/or transferrin saturation levels, and/or TMPRSS6 protein accumulation in the subject. In some embodiments, administering the dsRNA to the subject increases the subject's hemoglobin level and/or hematocrit level.
In a further aspect, the invention also provides a method of inhibiting TMPRSS6 expression in a subject. The method comprises administering to the subject a therapeutically effective amount of any dsRNA provided herein, thereby inhibiting expression of TMPRSS6 in the subject.
In one embodiment, the subject is a human.
In one embodiment, the dsRNA agent is administered to the subject at a dose of about 0.01mg/kg to about 50 mg/kg.
In one embodiment, the dsRNA agent is administered to the subject subcutaneously or intravenously.
In one embodiment, the method of the invention further comprises determining the level of TMPRSS6 in a sample from the subject.
In one embodiment, the level of TMPRSS6 in the subject sample is a level of TMPRSS6 protein in a blood, serum or liver sample.
In one embodiment, the method of the invention further comprises determining the level of iron and/or hepcidin in a sample from the subject.
In certain embodiments, the methods of the invention further comprise administering to the subject an additional therapeutic agent. In one embodiment, the methods of the invention further comprise administering to the subject an iron chelator, such as deferiprone, deferoxamine, and deferasirox (deferasirox).
The invention also provides a kit comprising any dsRNA of the invention or any pharmaceutical composition of the invention, and optionally instructions for use. In one embodiment, the invention provides a kit for performing a method of inhibiting expression of a TMPRSS6 gene in a cell by contacting a cell with an amount of a double stranded RNAi agent of the invention effective to inhibit expression of TMPRSS6 in the cell. The kit includes an RNAi agent and instructions for use, and optionally a method of administering the RNAi agent to a subject.
The invention also provides an RNA-induced silencing complex (RISC) comprising the antisense strand of any of the dsRNA agents of the invention.
Drawings
Fig. 1 is a schematic diagram depicting a study plan for determining the efficacy of dsRNA agents disclosed herein in cynomolgus monkeys.
Fig. 2 is a graph showing the percentage of serum TMPRSS6 mRNA remaining in cynomolgus monkeys (n=3 per group) administered indicated dsRNA duplex at single 3mg/kg or 10mg/kg dose subcutaneously on days 21, 22, 57 and 85 post-dosing. TMPRSS6 mRNA levels are shown relative to control levels obtained from cynomolgus monkeys administered PBS as a control.
Fig. 3 is a graph showing plasma iron levels as a percentage of pre-dose levels in cynomolgus monkeys (n=3 per group) administered indicated dsRNA duplex at single 3mg/kg or 10mg/kg doses subcutaneously on days 1, 8, 15, 22, 29, 36, 43, 50, 57, 64, 71, 78 and 85 post-dose.
Fig. 4 is a graph showing the percentage of transferrin saturation levels in cynomolgus monkeys (n=3 per group) administered indicated dsRNA duplex at single 3mg/kg or 10mg/kg doses subcutaneously on days 1, 8, 15, 22, 29, 36, 43, 50, 57, 64, 71, 78 and 85 post-dosing.
Detailed Description
The present invention provides iRNA compositions that affect RNA-induced silencing complex (RISC) -mediated cleavage of an RNA transcript of the transmembrane protease serine 6 (TMPRSS 6) gene. The gene may be within a cell, e.g., a cell in a subject (e.g., human). The use of these irnas enables targeted degradation of mRNA of the corresponding gene (TMPRSS 6) in mammals.
The iRNA of the invention is designed to target the human transmembrane protease serine 6 (TMPRSS 6) gene, including portions of the gene that are conserved among TMPRSS6 orthologs of other mammalian species. Without intending to be limited by theory, it is believed that the combination or subcombination of the above properties and specific target sites or specific modifications of these irnas impart improved efficacy, stability, potency, durability, and safety to the irnas of the present invention.
Accordingly, the present invention provides methods for treating and preventing transmembrane protease serine 6 (TMPRSS 6) -related disorders, such as disorders associated with iron overload and/or disorders that do not have erythropoiesis, such as hereditary hemochromatosis, beta-thalassemia (e.g., severe beta-thalassemia and intermediate beta-thalassemia), polycythemia vera, myelodysplastic syndrome, congenital erythropoiesis anemia, pyruvate kinase deficiency, using iRNA compositions that affect RNA-induced silencing complex (RISC) -mediated cleavage of RNA transcripts of the TMPRSS6 gene.
The iRNA of the present invention comprises an RNA strand (antisense strand) having a region of up to about 30 nucleotides or less in length, such as 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, that is substantially complementary to at least a portion of the mRNA transcript of the TMPRSS6 gene.
In certain embodiments, one or both strands of a double stranded RNAi agent of the invention are up to 66 nucleotides, e.g., 36-66, 26-36, 25-36, 31-60, 22-43, or 27-53 nucleotides in length, with a region of at least 19 consecutive nucleotides that is substantially complementary to at least a portion of an mRNA transcript of the TMPRSS6 gene. In some embodiments, such iRNA agents having an antisense strand of longer length can, for example, comprise a second RNA strand (sense strand) of 20 to 60 nucleotides in length, wherein the sense strand and the antisense strand form a duplex of 18 to 30 consecutive nucleotides.
The use of the iRNA of the invention enables targeted degradation of mRNA of a corresponding gene (TMPRSS 6 gene) in mammals. Using in vitro assays, the inventors have demonstrated that iRNA targeting the TMPRSS6 gene can effectively mediate RNAi, thereby significantly inhibiting expression of the TMPRSS6 gene. Thus, methods and compositions comprising these irnas are useful for treating a subject having a TMPRSS 6-related disorder, e.g., a disorder associated with iron overload and/or a disorder that is not erythropoietic, e.g., hereditary hemochromatosis, β -thalassemia (e.g., severe β -thalassemia and intermediate β -thalassemia), polycythemia vera, myelodysplastic syndrome, congenital erythropoiesis anemia, pyruvate kinase deficiency, erythropoiesis porphyria, parkinson's disease, alzheimer's disease, or friedel-crafts ataxia.
Accordingly, the present invention provides methods and combination therapies for treating a subject having a disorder that would benefit from inhibiting or reducing expression of a TMPRSS6 gene, e.g., a transmembrane protease serine 6 (TMPRSS 6) -related disorder, such as a disorder associated with iron overload and/or a disorder that is not erythropoietic, e.g., hereditary hemochromatosis, β -thalassemia (e.g., severe β -thalassemia and intermediate β -thalassemia), polycythemia vera, myelodysplastic syndrome, congenital erythropoiesis aplastic anemia, pyruvate kinase deficiency, using iRNA compositions that affect RNA-induced silencing complex (RISC) -mediated cleavage of RNA transcripts of the TMPRSS6 gene.
The invention also provides methods for preventing at least one symptom of a subject having a disorder that would benefit from inhibiting or reducing expression of a TMPRSS6 gene, e.g., a disorder associated with iron overload and/or a disorder that does not have erythropoiesis, e.g., hereditary hemochromatosis, beta-thalassemia (e.g., severe beta-thalassemia and intermediate beta-thalassemia), polycythemia vera, myelodysplastic syndrome, congenital erythropoiesis anemia, pyruvate kinase deficiency, erythropoiesis porphyria, parkinson's disease, alzheimer's disease, or friedel's ataxia.
The following detailed description discloses how to make and use iRNA-containing compositions to inhibit expression of TMPRSS6 genes, as well as compositions, uses, and methods for treating subjects that would benefit from inhibition and/or reduction of expression of TMPRSS6 genes, e.g., subjects that are susceptible to or diagnosed with a TMPRSS 6-related disorder.
I. Definition of the definition
For easier understanding of the present invention, certain terms are first defined. In addition, it should be noted that whenever a value or range of values for a parameter is referred to, it is intended that values and ranges intermediate to the values recited are also intended to be part of the present 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. For example, "an element" refers to one element or more than one element, e.g., a plurality of elements.
The term "comprising" 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 the context clearly indicates otherwise. For example, "sense strand or antisense strand" is understood to be "sense strand or antisense strand or sense and antisense strand".
The term "about" is used herein to mean within typical tolerances in the art. For example, "about" may be understood as about 2 standard deviations of the mean. In certain embodiments, about ±10%. In certain embodiments, about ±5%. When "about" occurs before a series of numbers or ranges, it is to be understood that "about" can modify each of the numbers in the series or ranges.
The term "at least", "not less than" or more "preceding a number or series of numbers is to be understood to include the number adjacent to the term" at least ", as well as all subsequent numbers or integers that may be logically included, as will be clear from context. For example, the number of nucleotides in a nucleic acid molecule must be an integer. For example, "at least 19 nucleotides in a 21 nucleotide nucleic acid molecule" means that 19, 20, or 21 nucleotides have the indicated properties. When "at least" occurs before a series of numbers or ranges, it is to be understood that "at least" can modify each of the numbers in the series or ranges.
As used herein, "no more than" or less "is understood to mean values adjacent to the phrase and logically lower values or integers, from logical to zero in the context. For example, a duplex with an "up to 2 nucleotides" overhang has a 2, 1 or 0 nucleotide overhang. When "no more than" occurs before a series of numbers or ranges, it is to be understood that "no more than" can modify each of the numbers in the series or ranges. As used herein, a range includes both upper and lower limits.
As used herein, a detection method may comprise determining that the amount of analyte present is below the detection level of the method.
In the event of a conflict between the indicated target site and the nucleotide sequence of the sense strand 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 sequences described in the specification will control.
As used herein, "transmembrane protease serine 6", which is used interchangeably with the term "TMPRSS6", refers to a type II plasma membrane serine protease (TTSP) gene or protein. TMPRSS6 is also known as Malatitase-2, IRIDA (iron refractory iron deficiency anemia), transmembrane protease serine 6, type II transmembrane serine protease 6, and membrane bound mosaic serine protease Malatitase-2. TMPRSS6 is a serine protease type II transmembrane protein of about 899 amino acids in length. TMPRSS6 contains multiple domains, e.g., a short endo-domain, a transmembrane domain, a SEA urchin sperm protein/intestinal peptidase domain/collectin (SEA) domain, two complement factors/SEA urchin embryo growth factors/BMP domains (CUB), three LDL-R a class domains (LDLa), and a trypsin-like serine protease domain with a conserved His-Asp-Ser triplet (HDS).
The sequence of the human TMPRSS6 mRNA transcript can be found, for example, in the following: genBank accession number GI 1755203660 (NM-153609.4;SEQ ID NO:1; reverse complement, SEQ ID NO: 2). The sequence of mouse TMPRSS6 mRNA can be found, for example, in the following: genBank accession number GI 125656151 (NM-027902.2;SEQ ID NO:3; reverse complement, SEQ ID NO: 4). The sequence of rat TMPRSS6 mRNA can be found, for example, in the following: genBank accession number GI 194474097 (NM-001130556.1;SEQ ID NO:5; reverse complement, SEQ ID NO: 6). The sequence of cynomolgus TMPRSS6 mRNA can be found, for example, in the following: genBank accession number GI:982272225 (XM_ 005567384.2;SEQ ID NO:7; reverse complement, SEQ ID NO: 8). The sequence of rhesus TMPRSS6 mRNA can be found, for example, in the following: genBank accession number GI:1622838152 (XM_ 015150283.2;SEQ ID NO:9; reverse complement, SEQ ID NO: 10).
Additional examples of TMPRSS6 mRNA sequences can be readily obtained by disclosing available databases such as GenBank, uniProt, OMIM and Macaca genome project sites.
Additional information about TMPRSS6 may be found, for example, at www.ncbi.nlm.nih.gov/gene/? term = TMPRSS 6.
By the date of filing the present application, the entire contents of each of the GenBank accession number and the gene database number described above are incorporated herein by reference.
As used herein, the term TMPRSS6 also refers to variants of the TMPRSS6 gene, including variants provided in the SNP database. Many sequence variants within the TMPRSS6 gene have been identified and can be found, for example, in NCBI dbSNP and UniProt (see, for example, www.ncbi.nlm.nih.gov/snp/.
As used herein, "target sequence" refers to a contiguous portion of the nucleotide sequence of an mRNA molecule formed during transcription of the TMPRSS6 gene, comprising mRNA that is the RNA processing product of the primary transcript. In one embodiment, the target portion of the sequence will be at least long enough to serve as a substrate for iRNA directed cleavage at or near the portion of the nucleotide sequence of the mRNA molecule formed during transcription of the TMPRSS6 gene.
The target sequence may be about 19 to 36 nucleotides in length, for example about 19 to 30 nucleotides in length. For example, the target sequence may 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 certain embodiments, the target sequence is 19 to 23 nucleotides in length, optionally 21 to 23 nucleotides in length. Ranges and lengths intermediate to those described above are also considered part of the present disclosure.
As used herein, the term "strand comprising a sequence" refers to an oligonucleotide comprising a chain of nucleotides described by a sequence referred to using standard nucleotide nomenclature.
"G", "C", "A", "T" and "U" generally represent nucleotides containing guanine, cytosine, adenine, thymine and uracil, respectively, as bases. However, it is understood that the term "ribonucleotide" or "nucleotide" may also refer to modified nucleotides, as described in further detail below, or alternative substitute parts (see e.g. table 1). It will be apparent to those skilled in the art that guanine, cytosine, adenine and uracil can be substituted with other moieties without substantially altering the base pairing properties of oligonucleotides including nucleotides containing such substituted moieties. For example, but not limited to, a nucleotide that includes inosine as its base may be base paired with a nucleotide containing adenine, cytosine, or uracil. Thus, in the nucleotide sequence of the dsRNA characteristic of the invention, the nucleotide containing uracil, guanine or adenine may be substituted with a nucleotide containing, for example, inosine. In another example, adenine and cytosine at any positions in the oligonucleotide may be substituted with guanine and uracil, respectively, to form a G-U wobble base pairing with the target mRNA. Sequences containing such substituted moieties are suitable for use in compositions and methods featuring the invention.
The terms "iRNA," "RNAi agent," "iRNA agent," and "RNA interfering agent" as used interchangeably herein refer to agents that contain RNA as defined herein, and which mediate targeted cleavage of RNA transcripts through an RNA-induced silencing complex (RISC) pathway. iRNA directs sequence-specific degradation of mRNA by a process known as RNA interference (RNAi). iRNA modulates, e.g., inhibits, expression of a TMPRSS6 gene in a cell, e.g., a cell in a subject (e.g., a mammalian subject).
In one embodiment, the RNAi agents of the invention comprise single-stranded RNA that interacts with a target RNA sequence (e.g., TMPRSS6 target mRNA sequence) to direct cleavage of the target RNA. Without wishing to be bound by theory, it is believed that long double stranded RNA introduced into the cells is decomposed into siRNA by a type III endonuclease called Dicer (Sharp et al, (2001) Gene and development (Genes Dev.)) (15:485). Dicer, a ribonuclease-III like enzyme, uses a characteristic two base 3' overhang to process dsRNA into 19 to 23 base pair short interfering RNA (Bernstein et al, (2001) Nature (Nature) 409:363). The siRNA is then incorporated into an RNA-induced silencing complex (RISC), wherein one or more helices cleave the siRNA duplex, thereby enabling the complementary antisense strand to direct target recognition (Nykanen et al, (2001) Cell (Cell) 107:309). Upon binding to the appropriate target mRNA, one or more endonucleases within RISC cleave the target to induce silencing (Elbashir et al, (2001) Gene and development 15:188). Thus, in one aspect, the invention relates to single stranded RNAs (sirnas) produced in cells, and which promote the formation of RISC complexes to affect silencing of a target gene, namely TMPRSS6 gene. Thus, the term "siRNA" is also used herein to refer to iRNA as described above.
In certain embodiments, the RNAi agent can be a single stranded siRNA (ssRNAi) that is introduced into a cell or organism to inhibit a target mRNA. The single stranded RNAi agent binds to RISC endonuclease Argonaute2, which then cleaves the target mRNA. Single-stranded siRNA is typically 15 to 30 nucleotides and is chemically modified. The design and testing of single stranded siRNA is described in U.S. Pat. No. 8,101,348, good and Lima et al, (2012) cell 150:883-894, the entire contents of each of which are incorporated herein by reference. Any of the antisense nucleotide sequences described herein can be used as a single stranded siRNA described herein or chemically modified by the method described in Lima et al, (2012) cell 150:883-894.
In certain embodiments, the "iRNA" used in the compositions, uses, and methods of the invention is 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 antiparallel and substantially complementary nucleic acid strands having "sense" and "antisense" orientations relative to a target RNA (i.e., TMPRSS6 gene). In some embodiments of the invention, double-stranded RNA (dsRNA) triggers degradation of target RNA (e.g., mRNA) by a post-transcriptional gene silencing mechanism referred to herein as RNA interference or RNAi.
In general, the majority of the nucleotides of each strand of a dsRNA molecule are ribonucleotides, but as described in detail herein, each or both strands may also comprise one or more non-ribonucleotides, such as deoxyribonucleotides or modified nucleotides. In addition, as used in this specification, "iRNA" may comprise ribonucleotides with chemical modification; the iRNA may comprise substantial modifications at multiple nucleotides. As used herein, the term "modified nucleotide" refers to a nucleotide that independently has a modified sugar moiety, a modified internucleotide linkage, or a modified nucleobase, or any combination thereof. Thus, the term modified nucleotide encompasses substitution, addition or removal of an internucleoside linkage, sugar moiety or nucleobase, e.g., a functional group or atom. Modifications suitable for use in the agents of the invention include all types of modifications disclosed herein or known in the art. For the purposes of the present specification and claims, as used in siRNA-type molecules, any such modification is encompassed by "iRNA" or "RNAi agent".
In certain embodiments of the present disclosure, the inclusion of deoxynucleotides may be considered to constitute modified nucleotides if present within an RNAi agent.
The duplex region may be any length that allows for specific degradation of the desired target RNA through the RISC pathway, and the length may be in the range of about 19 to 36 base pairs, e.g., about 19 to 30 base pairs in length, e.g., 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 certain embodiments, the duplex region is 19 to 21 base pairs in length, e.g., 21 base pairs in length. Ranges and lengths intermediate to those described above are also considered part of the present disclosure.
The two strands forming the duplex structure may be different portions of one larger RNA molecule, or they may be separate RNA molecules. If the two strands are part of one larger molecule and are thus connected by an uninterrupted nucleotide chain between the 3 'end of one strand and the 5' end of the corresponding other strand forming a duplex structure, the connected RNA strand is referred to as a "hairpin loop". The hairpin loop may include at least one unpaired nucleotide. In some embodiments, the hairpin loop may include at least 2, 3, 4, 5, 6, 7, 8, 9, 10, 20, 23, or more unpaired nucleotides. In some embodiments, the hairpin loop may be 10 nucleotides or less. In some embodiments, the hairpin loop may be 8 or fewer unpaired nucleotides. In some embodiments, the hairpin loop may be 4 to 10 unpaired nucleotides. In some embodiments, the hairpin loop may be 4 to 8 nucleotides.
Where the two substantially complementary strands of the dsRNA are made up of separate RNA molecules, those molecules need not be, but can be, covalently linked. When two strands are covalently linked by other means than an uninterrupted nucleotide chain between the 3 'end of one strand and the 5' end of the corresponding other strand forming a duplex structure, the linking structure is referred to as a "adaptor" RNA strand may have the same or different numbers of nucleotides. The maximum number of base pairs is the number of nucleotides in the shortest strand of the dsRNA minus any overhangs present in the duplex. In addition to duplex structures, RNAi can include 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 other embodiments, both the 3 'and 5' ends of one strand of the RNAi agent comprise an overhang of at least 1 nucleotide.
In certain embodiments, the iRNA agent of the invention is a dsRNA, each strand of which comprises 19-23 nucleotides, that interacts with a target RNA sequence, such as the TMPRSS6 gene, to directly cleave the target RNA.
In some embodiments, an iRNA of the invention is a 24-30 nucleotide dsRNA that interacts with a target RNA sequence (e.g., TMPRSS6 target mRNA sequence) to guide 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 the 3 'end of one strand of a dsRNA extends beyond the 5' end of the other strand, or vice versa, a nucleotide overhang is present. The dsRNA may include an overhang of at least one nucleotide; alternatively, the overhang may include at least two nucleotides, at least three nucleotides, at least four nucleotides, at least five nucleotides, or more. Nucleotide overhangs may include or consist of: nucleotide/nucleoside analogs comprising deoxynucleotides/nucleosides. The overhang may be on the sense strand, the antisense strand, or any combination thereof. Furthermore, the overhanging nucleotides may be present on the 5 'end, 3' end or both ends of the antisense strand or sense strand of the dsRNA.
In one embodiment, the antisense strand of the dsRNA has 1 to 10 nucleotides, e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 nucleotides, at the 3 'end or the 5' end of the overhang. In one embodiment, the overhang of the sense strand of the dsRNA at the 3 'end or 5' end has 1 to 10 nucleotides, e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 nucleotides. In another embodiment, one or more nucleotides in the overhang are replaced with a nucleoside thiophosphate.
In certain embodiments, the antisense strand of the dsRNA has 1 to 10 nucleotides, e.g., 0 to 3, 1 to 3, 2 to 4, 2 to 5, 4 to 10, 5 to 10, e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 nucleotides at the 3 'end or the 5' end of the overhang. In one embodiment, the overhang of the sense strand of the dsRNA at the 3 'end or 5' end has 1 to 10 nucleotides, e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 nucleotides. In another embodiment, one or more nucleotides in the overhang are replaced with a nucleoside thiophosphate.
In certain embodiments, the antisense strand of the dsRNA has 1 to 10 nucleotides, e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 nucleotides, at the 3 'end or the 5' end of the overhang. In certain embodiments, the overhang on the sense strand or the antisense strand, or both, can comprise an extension length longer than 10 nucleotides in length, such as 1 to 30 nucleotides in length, 2 to 30 nucleotides in length, 10 to 25 nucleotides in length, 10 to 20 nucleotides in length, or 10 to 15 nucleotides in length. In certain embodiments, the extended overhang is on the sense strand of the duplex. In certain embodiments, there is an extended overhang on the 3' end of the sense strand of the duplex. In certain embodiments, there is an extended overhang on the 5' end of the sense strand of the duplex. In certain embodiments, the extended overhang is on the antisense strand of the duplex. In certain embodiments, there is an extended overhang on the 3' end of the antisense strand of the duplex. In certain embodiments, there is an extended overhang on the 5' end of the antisense strand of the duplex. In certain embodiments, one or more nucleotides in the extended overhang are replaced with nucleoside thiophosphates. In certain embodiments, the overhang comprises a self-complementary portion such that the overhang is capable of forming a hairpin structure that is stable under physiological conditions.
By "blunt" or "blunt end" is meant that there are no unpaired nucleotides at the end of the double stranded RNA agent, i.e. no nucleotide overhangs. A "blunt-ended" double-stranded RNA agent is double-stranded throughout its length, i.e., no nucleotide overhangs are present at either end of the molecule. The RNAi agents of the invention include RNAi agents that have no nucleotide overhangs at one end (i.e., agents having one overhang and one blunt end) or no nucleotide overhangs at both ends. The most common such molecules will be double stranded throughout their length.
The term "antisense strand" or "guide strand" refers to a strand of an iRNA, e.g., a dsRNA, that comprises a region that is substantially complementary to a target sequence (e.g., TMPRSS6 mRNA).
As used herein, the term "complementary region" refers to a region on the antisense strand that is substantially complementary to a sequence defined herein (e.g., a target sequence, such as a TMPRSS6 nucleotide sequence). In the case where the complementary region is not perfectly complementary to the target sequence, the mismatch may be in the internal or terminal region of the molecule. Typically, the most tolerable mismatches are within the terminal region, e.g., 5, 4, or 3 nucleotides of the 5 '-or 3' -end of the iRNA. In some embodiments, the double stranded RNA agent of the invention comprises a nucleotide mismatch in the antisense strand. In some embodiments, the antisense strand of a double-stranded RNA agent of the invention comprises no more than 4 mismatches to the target mRNA, e.g., the antisense strand comprises 4, 3, 2, 1, or 0 mismatches to the target mRNA. In some embodiments, an antisense strand double-stranded RNA agent of the invention comprises no more than 4 mismatches with the sense strand, e.g., the antisense strand comprises 4, 3, 2, 1, or 0 mismatches with the sense strand. In some embodiments, the double stranded RNA agents of the invention comprise nucleotide mismatches in the sense strand. In some embodiments, the sense strand of a double-stranded RNA agent of the invention comprises no more than 4 mismatches with the antisense strand, e.g., the sense strand comprises 4, 3, 2, 1, or 0 mismatches with the antisense strand. In some embodiments, the nucleotide mismatches are within, for example, 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 is not in the seed region.
Thus, an RNAi agent as described herein can contain one or more mismatches with a 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 a mismatch to the target sequence, the mismatch can optionally be limited to the last 5 nucleotides from the 5 '-or 3' -end of the complementary region. For example, in such embodiments, for a 23 nucleotide RNAi agent, the strand complementary to the region of the TMPRSS6 gene typically does not contain any mismatches within the center 13 nucleotides. The methods described herein or known in the art can be used to determine whether an RNAi agent containing a mismatch to a target sequence is effective in inhibiting expression of the TMPRSS6 gene. Considering the efficacy of RNAi agents with mismatches in inhibiting TMPRSS6 gene expression is important, especially if specific complementary regions in the TMPRSS6 gene are known to have polymorphic sequence variations within the population.
As used herein, the term "sense strand" or "follower strand" refers to an iRNA strand comprising a region that is substantially complementary to a region of the antisense strand of the term as defined herein.
As used herein, "substantially all nucleotides are modified" to a large extent but not completely modified, and may comprise no more than 5, 4, 3, 2, or 1 unmodified nucleotides.
As used herein, the term "cleavage region" refers to the region immediately adjacent to the cleavage site. The cleavage site is the site on the target where cleavage occurs. In some embodiments, the cleavage region comprises three bases on either end of the cleavage site and immediately adjacent to the cleavage site. In some embodiments, the cleavage region comprises two bases on either end of the cleavage site and immediately adjacent to the cleavage site. In some embodiments, the cleavage site occurs specifically at the site where nucleotides 10 and 11 of the antisense strand bind, 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 relative to a second nucleotide sequence refers to the ability of an oligonucleotide or polynucleotide comprising the first nucleotide sequence to hybridize under certain conditions to an oligonucleotide or polynucleotide comprising the second nucleotide sequence and form a duplex structure, as will be understood by the skilled artisan. For example, such conditions may be stringent conditions, where stringent conditions may comprise: 400mM NaCl,40mM PIPES,pH 6.4,1mM EDTA,50 ℃or 70℃for 12 to 16 hours, and then washed (see, for example, molecular cloning: laboratory Manual (Molecular Cloning: A Laboratory Manual), sambrook et al, (1989) Cold spring harbor laboratory Press (Cold Spring Harbor Laboratory Press)). Other conditions may be applied, such as physiologically relevant conditions that may be encountered inside an organism. The skilled person will be able to determine the set of conditions most suitable for the complementarity test of the two sequences depending on the end use of the hybridizing nucleotides.
Complementary sequences within an iRNA (e.g., within a dsRNA as described herein) comprise base pairing of an oligonucleotide or polynucleotide comprising a first nucleotide sequence with an oligonucleotide or polynucleotide comprising a second nucleotide sequence over the entire length of one or both nucleotide sequences. Such sequences may be referred to herein as being "fully complementary" to each other. However, when a first sequence is referred to herein as "substantially complementary" to a second sequence, the two sequences may be fully complementary, or they may form one or more, but typically no more than 5, 4, 3 or 2 mismatched base pairs after hybridization for up to 30 base pairs of the duplex, while retaining the ability to hybridize under conditions most relevant to its end use, such as in vitro or in vivo inhibition of gene expression. However, where two oligonucleotides are designed to form one or more single stranded overhangs upon hybridization, such overhangs should not be considered as a defined mismatch with respect to complementarity. For example, a dsRNA comprising one oligonucleotide of 21 nucleotides in length and another oligonucleotide of 23 nucleotides in length, wherein the longer oligonucleotide comprises a sequence of 21 nucleotides that is fully complementary to the shorter oligonucleotide, may still be referred to as "fully complementary" for purposes described herein.
As used herein, a "complementary" sequence may also comprise or be formed entirely of non-Watson-Crick base pairs (non-Watson-Crick base pairs) or base pairs formed from non-natural and modified nucleotides, so long as the above requirements regarding its hybridization ability are met. Such non-Watson-Crick base pairs include, but are not limited to, G: U wobble base pairing or Holstein base pairing (Hoogsteen base pairing).
The terms "complementary," "fully complementary," and "substantially complementary" herein may be used with respect to base matching between the sense and antisense strands of a dsRNA, or base matching between two oligonucleotides or polynucleotides (e.g., the antisense strand and target sequence of a double-stranded RNA agent), as understood in the context of its use.
As used herein, a polynucleotide that is "at least partially substantially complementary" to a messenger RNA (mRNA) refers to a polynucleotide that is substantially complementary to a contiguous portion of an mRNA of interest (e.g., an mRNA encoding the TMPRSS6 gene). For example, if the sequence is substantially complementary to an uninterrupted portion of an mRNA encoding the TMPRSS6 gene, the polynucleotide is complementary to at least a portion of the TMPRSS6 mRNA.
Thus, in some embodiments, the antisense polynucleotides disclosed herein are fully complementary to the target TMPRSS6 sequence. In other embodiments, an antisense polynucleotide disclosed herein is substantially complementary to a target TMPRSS6 sequence and comprises a contiguous nucleotide sequence that is at least 80% complementary over its entire length to the nucleotide sequence of any of SEQ ID NOS: 1, 3, 5, 7, or 9 or to an equivalent region of a fragment of any of SEQ ID NOS: 1, 3, 5, 7, or 9, 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%.
In some embodiments, an antisense polynucleotide disclosed herein is substantially complementary to a fragment of a target TMPRSS6 sequence and comprises a contiguous nucleotide sequence that is at least 80% complementary over its entire length to a fragment of SEQ ID NO:1 selected from the group consisting of: nucleotides 187-210 of SEQ ID NO. 1; 227-254;322-363;362-390;398-420;404-429;410-435;439-461;443-467;448-474;460-483;466-488;496-519;519-542;526-548;557-593;641-671;652-676;687-713;725-762;757-794;886-908;921-951;956-987;1051-1082;1233-1269;1279-1313;1313-1341;1327-1351;1415-1439;1447-1480;1464-1486;1486-1509;1559-1589;1571-1595;1579-1609;1707-1735;1738-1764;1806-1828;1864-1886;1934-1966;1967-1991;2008-2031;2015-2043;2042-2072;2287-2311;2297-2354;2336-2361;2360-2384;2416-2438;2481-2510;2496-2527;2526-2558;2665-2693;2693-2719;2707-2729;2799-2821;2851-2874;2971-2999;2981-3006; and 3155-3195, e.g., 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, an antisense polynucleotide disclosed herein is substantially complementary to a fragment of a target TMPRSS6 sequence and comprises a contiguous nucleotide sequence that is at least 80% complementary over its entire length to a fragment of SEQ ID NO:1 selected from the group consisting of: nucleotides 230-252, 324-346, 560-578, 560-582, 2338-2360, 3163-3185, 3169-3191 and 3172-3194 of SEQ ID NO. 1 are 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, an antisense polynucleotide disclosed herein is substantially complementary to a fragment of a target TMPRSS6 sequence and comprises a contiguous nucleotide sequence that is at least 80% complementary over its entire length to a fragment of SEQ ID NO:1 selected from the group consisting of: nucleotides 560-578, 2338-2360 and 3169-3191 of SEQ ID NO. 1, e.g., 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 other embodiments, an antisense polynucleotide disclosed herein is substantially complementary to a target TMPRSS6 sequence and comprises a contiguous nucleotide sequence that is at least about 80% complementary over its entire length to any one of the sense strand nucleotide sequences in any one of the tables 2-7, or a fragment of any one of the sense strand nucleotide sequences in any one of the tables 2-7, such as about 85%, about 90%, about 91%, about 92%, about 93%, about 94%, about 95%, about 96%, about 97%, about 98%, about 99% or 100% complementary.
In one embodiment, the RNAi agents of the present disclosure comprise a sense strand that is substantially complementary to an antisense polynucleotide, which in turn is identical to a target TMPRSS6 sequence, and wherein the sense strand polynucleotide comprises a contiguous nucleotide sequence that is at least about 80% complementary, such as about 85%, about 90%, about 91%, about 92%, about 93%, about 94%, about 95%, about 96%, about 97%, about 98%, about 99% or 100% complementary, over its entire length to SEQ ID NO:2, 4, 6, 8 or 10, or a fragment of any of SEQ ID NOs: 2, 4, 6, 8 or 10.
In some embodiments, an iRNA of the invention comprises a sense strand that is substantially complementary to an antisense polynucleotide that in turn is complementary to a target TMPRSS6 sequence, and wherein the sense strand polynucleotide comprises a contiguous nucleotide sequence that is at least about 80% complementary over its entire length to any one of the antisense strand nucleotide sequences in any one of tables 2-7 or a fragment of any one of the antisense strand nucleotide sequences in any one of tables 2-7, such as about 85%, about 90%, about 91%, about 92%, about 93%, about 94%, about 95%, about 96%, about 97%, about 98%, about 99% or 100% complementary.
In certain embodiments, the sense and antisense strands are selected from any one of the following: duplex AD-1556360, AD-1571158, AD-1571033, AD-1554875, AD-1571160, AD-1555117, AD-1554911 and AD-1556915.
In certain embodiments, the sense and antisense strands are selected from any one of the following: AD-1556360, AD-1571158 and AD-1571033.
In general, "iRNA" comprises ribonucleotides with chemical modification. Such modifications may include all types of modifications disclosed herein or known in the art. For the purposes of the present specification and claims, as used in dsRNA molecules, any such modifications are encompassed by "iRNA".
In certain embodiments of the present disclosure, the inclusion of deoxynucleotides may be considered to constitute modified nucleotides if present within an RNAi agent.
In one aspect of the invention, the agents used in the methods and compositions of the invention are single stranded antisense oligonucleotide molecules that inhibit target mRNA by an antisense inhibition mechanism. The single stranded antisense oligonucleotide molecule is complementary to a sequence within the target mRNA. Single-stranded antisense oligonucleotides can inhibit translation in a stoichiometric manner by base pairing with mRNA and physically blocking the translation machinery, see Dias, N.et al, (2002) molecular Cancer therapeutics (Mol Cancer Ther) 1:347-355. The single stranded antisense oligonucleotide molecule can be about 14 to about 30 nucleotides in length and have a sequence complementary to the target sequence. For example, a single stranded antisense oligonucleotide molecule can comprise a sequence of at least about 14, 15, 16, 17, 18, 19, 20, or more consecutive nucleotides from any of the antisense sequences described herein.
As used herein, the phrase "contacting a cell with an iRNA (e.g., dsRNA)" includes contacting a cell by any possible means. Contacting the cell with the iRNA comprises contacting the cell with the iRNA in vitro or contacting the cell with the iRNA in vivo. The contacting may be performed directly or indirectly. Thus, for example, an iRNA may be brought into physical contact with a cell by an individual performing the method, or alternatively, the iRNA may be brought into a condition that allows or brings it into subsequent contact with the cell.
In vitro contacting of cells can be performed, for example, by incubating the cells with iRNA. Contacting cells in vivo may be performed, for example, by injecting iRNA into or near the tissue in which the cells are located, or by injecting iRNA into another area, such as the blood stream or subcutaneous space, such that the agent then reaches the tissue in which the cells are to be contacted. For example, the iRNA may contain or be conjugated to a ligand (e.g., galNAc) that directs the iRNA to a site of interest, such as the liver. Combinations of in vitro and in vivo contact methods are also possible. For example, the cells can also be contacted with an iRNA in vitro and subsequently transplanted into a subject.
In certain embodiments, contacting the cell with the iRNA comprises "introducing" or "delivering" the iRNA into the cell by promoting or affecting uptake or uptake by the cell. The uptake or uptake of iRNA can occur by unassisted diffusion or active cellular processes, or by adjuvants or devices. The introduction of the iRNA into the cell may be in vitro or in vivo. For example, for in vivo introduction, the iRNA may be injected into a tissue site or administered systemically. In vitro introduction into cells comprises methods known in the art, such as electroporation and lipofection. Additional methods are described 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. LNP is described, for example, in U.S. patent nos. 6,858,225, 6,815,432, 8,158,601, and 8,058,069, the entire contents of which are hereby incorporated by reference.
As used herein, a "subject" is an animal, such as a mammal, that endogenously or heterologously expresses a target gene, including primates (e.g., humans, non-human primates, e.g., monkeys and chimpanzees), non-primates (e.g., cows, pigs, horses, goats, rabbits, sheep, hamsters, guinea pigs, cats, dogs, rats or mice), or birds. In one embodiment, the subject is a human, such as a human being treated or evaluated for a disease or disorder that would benefit from reduced expression of TMPRSS 6; a human at risk of a disease or disorder that would benefit from reduced TMPRSS6 expression; a human suffering from a disease or disorder that would benefit from reduced expression of TMPRSS 6; or a person being treated who would benefit from a disease or disorder in which TMPRSS6 expression is reduced 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 term "treatment" or "treatment" refers to a beneficial or desired outcome, such as reducing at least one sign or symptom of a TMPRSS 6-related disorder in a subject. Treatment also includes reducing one or more signs or symptoms associated with undesired TMPRSS6 expression; reducing the extent of undesired TMPRSS6 activation or stabilization; improving or moderating undesired TMPRSS6 activation or stabilization. "treatment" may also mean an extension of survival compared to the expected survival without treatment. In the context of TMPRSS6 levels in a subject or disease marker or symptom, the term "lower" means that such levels are statistically significantly reduced. The decrease may 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, the reduction is at least 20%. In certain embodiments, the decrease in the level of a disease marker, e.g., protein or gene expression, is at least 50%. A "lower" level of TMPRSS6 in a subject is an acceptable level that decreases to within the normal range of individuals without such a disorder. In certain embodiments, "lower" is a decrease in the difference between the level of a marker or symptom of a subject with a disease and the level that the individual receives in the normal range, e.g., the level of weight loss between an obese individual and an individual with weight received in the normal range.
As used herein, "prevention" or "prophylaxis" when used in reference to a disease, disorder, or condition thereof, may be treated or ameliorated by reducing expression of a TMPRSS6 gene, and refers to reducing the likelihood that a subject will develop symptoms associated with such disease, disorder, or condition (e.g., symptoms of undesired or excessive TMPRSS6 expression, such as elevated levels of iron or iron imbalance). The likelihood of developing elevated levels of iron or iron imbalance is reduced, for example, when an individual having one or more risk factors for elevated levels of iron or iron imbalance fails to develop elevated levels of iron or suffers from elevated levels of iron or iron imbalance to a lesser extent than a population having the same risk factors and not receiving the treatment described herein. Failure to develop a disease, disorder, or condition, or reduced development of symptoms associated with such a disease, disorder, or condition (e.g., at least about 10% reduction on a clinically accepted scale of the disease or disorder), or delayed manifestation of symptoms (e.g., days, weeks, months, or years) is considered effective prophylaxis.
As used herein, the term "transmembrane protease serine 6-related disease" or "TMPRSS 6-related disease" is a disease or disorder caused by or associated with TMPRSS6 gene expression or TMPRSS6 protein production. The term "TMPRSS 6-related disease" encompasses a disease, disorder, or condition that would benefit from reduced expression, replication, or protein activity of a TMPRSS6 gene.
In some embodiments, the TMPRSS 6-related disease is a disorder associated with iron overload, a condition characterized by elevated levels of iron, or an iron imbalance. Iron overload can be caused, for example, by genetic conditions, increased iron uptake in the diet, or parenterally administered excess iron, including intravenous injection and transfusion iron overload of excess iron.
In some embodiments, the TMPRSS6 related disease is a disorder that is not erythropoietic. Ineffective erythropoiesis is an abnormal expansion of the number of erythrocyte progenitors, accompanied by ineffective synthesis of enucleated erythrocytes, resulting in anemia and hypoxia. In particular, an increase in erythroid cells does not produce a corresponding increase in erythrocytes. Thus, iron absorption is still increased in response to stress, but iron is deposited in the organ rather than for the production of more red blood cells.
In some embodiments, TMPRSS 6-related disorders include, but are not limited to, hereditary hemochromatosis, idiopathic hemochromatosis, primary hemochromatosis, secondary hemochromatosis, severe juvenile hemochromatosis, neonatal hemochromatosis, iron-mother's anemia, hemolytic anemia, erythropoiesis anemia, sickle cell anemia, thalassemia (e.g., β -thalassemia and α -thalassemia), polycythemia vera, myelodysplastic syndrome, congenital erythropoiesis anemia, pyruvate kinase deficiency, chronic liver disease, delayed skin porphyrins, erythropoiesis porphyria, metastatic ferriglobulinemia, hereditary tyrosine blood, cerebral liver and kidney syndrome, idiopathic pulmonary iron-containing hemochromatosis, renal iron-containing hemosiderosis.
In some embodiments, the TMPRSS 6-related disorder comprises a disorder related to oral administration of excess iron, transfusion iron overload, and intravenous injection of excess iron.
In other embodiments, TMPRSS6 related disorders also include disorders having symptoms associated with or likely caused by iron overload. Such symptoms include liver disease (cirrhosis, cancer), heart attack or heart failure, diabetes, osteoarthritis, osteoporosis, metabolic syndrome, hypothyroidism, hypogonadism, and in some cases an increased risk of premature death. In one embodiment, the TMPRSS 6-related disorder comprises a neurodegenerative disorder associated with iron overload and/or iron imbalance, such as alzheimer's disease, parkinson's disease, huntington's disease, friedel-crafts ataxia, epilepsy, and multiple sclerosis. Administration of TMPRSS 6-targeted iRNA, such as described in any of tables 2-7, can treat one or more of these symptoms, or prevent the development or progression of a disease or disorder that is exacerbated by increased levels of iron.
In one embodiment, the TMPRSS 6-related disorder is beta-thalassemia. Beta-thalassemia is any one of a group of genetic disorders characterized by a genetic defect in beta-globin chain synthesis. In the homozygous state, β -thalassemia ("thalassemia major") causes severe transfusion dependent anemia. In the heterozygous state, the β -thalassemia trait ("mild thalassemia") results in mild to moderate microcytic anemia. An "intermediate thalassemia" is a β -thalassemia that results in a subject with a clinical severity of the disease that is between mild β -thalassemia and mild symptoms of severe β -thalassemia. Several laboratory tests may be used to help detect and diagnose thalassemia, for example, whole blood count to determine the number of red blood cells and the number of hemoglobin, blood smear tests, hemoglobin electrophoresis, gene sequencing, or iron tests to check iron, ferritin, unshaped iron binding capacity, total iron binding capacity, or transferrin saturation level. The type and relative amount of hemoglobin present in red blood cells is another indicator of thalassemia. Beta-thalassemia disrupts the balance of beta and alpha hemoglobin chain formation and results in an increase in minor hemoglobin components. Thus, individuals with severe β -thalassemia typically have a greater percentage of Hb F. Individuals with mild beta-thalassemia typically have an elevated Hb A2 fraction.
In one embodiment, the beta-thalassemia is thalassemia major. In another embodiment, the beta-thalassemia is an intermediate thalassemia.
In some embodiments, the TMPRSS 6-related disorder is polycythemia vera. Polycythemia vera is a blood cancer that results in bone marrow producing excessive amounts of red blood cells. These excess cells often want to blood vessels, which makes it easier for the patient to develop a bleeding clot, as well as other complications such as stroke or heart attack. Several tests may be performed to aid in the detection and diagnosis of polycythemia vera, such as whole blood count, blood smear tests, erythropoietin level tests, bone marrow aspiration or biopsy, or gene sequencing.
As used herein, a "therapeutically effective amount" is intended to encompass an amount of RNAi agent sufficient to affect treatment of a disease (e.g., by reducing, ameliorating, or maintaining an existing disease or one or more symptoms of a disease) when administered to a subject having a TMPRSS 6-related disease. The "therapeutically effective amount" may vary depending on the RNAi agent, how the agent is administered, the disease and its severity, as well as the medical history, age, weight, family history, genetic composition, type of previous or concomitant therapy (if any), and other individual characteristics of the subject to be treated.
As used herein, a "prophylactically effective amount" is intended to encompass an amount of RNAi agent sufficient to prevent or ameliorate a disease or one or more symptoms of the disease when administered to a subject suffering from a TMPRSS 6-related disorder. Improving a disease comprises slowing the progression of the disease or reducing the severity of a subsequent disease. The "prophylactically effective amount" may vary depending on the RNAi agent, how the agent is administered, the degree of risk of the disease, as well as the medical history, age, body weight, family history, genetic composition, type of previous or concomitant therapy (if any), and other individual characteristics of the patient to be treated.
"therapeutically effective amount" or "prophylactically effective amount" also encompasses 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 invention can be administered in sufficient amounts to produce a reasonable benefit/risk ratio suitable for such treatment.
The phrase "pharmaceutically acceptable" is used herein to refer to those compounds, materials (including salts), compositions, or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human and animal subjects without excessive toxicity, irritation, allergic response, or other problem or complication commensurate with a reasonable benefit/risk ratio.
As used herein, the phrase "pharmaceutically acceptable carrier" refers to 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 stearic acid), or solvent encapsulating material (involving 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 deleterious to the subject being treated. Such vectors are known in the art. The pharmaceutically acceptable carrier comprises a carrier for administration by injection.
As used herein, the term "sample" includes a collection of similar fluids, cells, or tissues isolated from a subject, as well as fluids, cells, and tissues present in the subject. Examples of biological fluids include blood, serum and serosal fluids, plasma, cerebrospinal fluid, ocular fluid, lymph, urine, saliva, and the like. The tissue sample may comprise a sample from a tissue, organ or local area. For example, the sample may originate from a particular organ, a portion of an organ, or a fluid or cell within such an organ. In certain embodiments, the sample may be derived from the liver (e.g., whole liver or portions of the liver or certain types of cells in the liver, e.g., hepatocytes). In some embodiments, "sample derived from a subject" refers to urine obtained from a subject. "subject-derived sample" may refer to blood or blood-derived serum or plasma from a subject.
II. IRNA of the invention
The present invention provides iRNA that inhibits expression of TMPRSS6 gene. In certain embodiments, the iRNA comprises a double-stranded ribonucleic acid (dsRNA) molecule for inhibiting expression of a TMPRSS6 gene in a cell, such as a cell within a subject, e.g., a mammal, e.g., a human susceptible to a TMPRSS 6-related disorder, e.g., a disorder associated with iron overload and/or a disorder that is not erythropoietic, e.g., hereditary hemochromatosis, β -thalassemia (e.g., severe β -thalassemia and intermediate β -thalassemia), polycythemia vera, myelodysplastic syndrome, congenital erythropoiesis aplastic anemia, pyruvate kinase deficiency, erythropoiesis porphyrin, parkinson's disease, alzheimer's disease, or friedel's ataxia. The dsRNAi agent comprises an antisense strand with a complementary region complementary to at least a portion of the mRNA formed in expression of the TMPRSS6 gene. The complementary region is about 19 to 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 a TMPRSS6 gene, the iRNA inhibits expression of the TMPRSS6 gene (e.g., a human, primate, non-primate, or rat TMPRSS6 gene) by at least about 50%, as determined by, for example, PCR or branched DNA (bDNA) based methods or by protein based methods (e.g., by immunofluorescence analysis using, for example, western blot or flow cytometry techniques). In certain embodiments, inhibition of expression is determined by qPCR methods provided in examples herein, in a suitable organism cell line provided therein, with siRNA, e.g., at a concentration of 10 nM. In certain embodiments, inhibition of expression in vivo is determined by knocking down the human gene in a rodent that expresses the human gene, e.g., a mouse that expresses a human target gene or an AAV-infected mouse, e.g., when administered in a single dose, e.g., at 3mg/kg at the nadir of RNA expression.
The dsRNA comprises two RNA strands that are complementary and hybridize under conditions where the dsRNA will be used to form a duplex structure. One strand (the antisense strand) of the dsRNA comprises a region of complementarity that is substantially complementary and typically fully complementary to a target sequence. The target sequence may be derived from the sequence of mRNA formed during expression of the TMPRSS6 gene. The other strand (the sense strand) contains a region complementary to the antisense strand such that the two strands hybridize and form a duplex structure when combined under appropriate conditions. As described elsewhere herein and as known in the art, the complementary sequence of a dsRNA can also be included as a self-complementary region of a single nucleic acid molecule relative to being located on a separate oligonucleotide.
Typically, the duplex structure is 15 to 30 base pairs in length, for example 15 to 29, 15 to 28, 15 to 27, 15 to 26, 15 to 25, 15 to 24, 15 to 23, 15 to 22, 15 to 21, 15 to 20, 15 to 19, 15 to 18, 15 to 17, 18 to 30, 18 to 29, 18 to 28, 18 to 27, 18 to 26, 18 to 25, 18 to 24, 18 to 23, 18 to 22, 18 to 21, 18 to 20, 19 to 30, 19 to 29, 19 to 28 19 to 27, 19 to 26, 19 to 25, 19 to 24, 19 to 23, 19 to 22, 19 to 21, 19 to 20, 20 to 30, 20 to 29, 20 to 28, 20 to 27, 20 to 26, 20 to 25, 20 to 24,20 to 23, 20 to 22, 20 to 21, 21 to 30, 21 to 29, 21 to 28, 21 to 27, 21 to 26, 21 to 25, 21 to 24, 21 to 23, or 21 to 22 base pairs. In certain embodiments, the duplex structure is 18 to 25 base pairs in length, e.g., 18 to 25, 18 to 24, 18 to 23, 18 to 22, 18 to 21, 18 to 20, 19 to 25, 19 to 24, 19 to 23, 19 to 22, 19 to 21, 19 to 20, 20 to 25, 20 to 24,20 to 23, 20 to 22, 20 to 21, 21 to 25, 21 to 24, 21 to 23, 21 to 22, 22 to 25, 22 to 24, 22 to 23, 23 to 25, 23 to 24, or 24 to 25 base pairs in length, e.g., 19 to 21 base pairs in length. Ranges and lengths intermediate to those described above are also considered part of the present disclosure.
Similarly, the region complementary to the target sequence is 15 to 30 nucleotides in length, for example 15 to 29, 15 to 28, 15 to 27, 15 to 26, 15 to 25, 15 to 24, 15 to 23, 15 to 22, 15 to 21, 15 to 20, 15 to 19, 15 to 18, 15 to 17, 18 to 30, 18 to 29, 18 to 28, 18 to 27, 18 to 26, 18 to 25, 18 to 24, 18 to 23, 18 to 22, 18 to 21, 18 to 20, 19 to 30, 19 to 29, 19 to 28 19 to 27, 19 to 26, 19 to 25, 19 to 24, 19 to 23, 19 to 22, 19 to 21, 19 to 20, 20 to 30, 20 to 29, 20 to 28, 20 to 27, 20 to 26, 20 to 25, 20 to 24,20 to 23, 20 to 22, 20 to 21, 21 to 30, 21 to 29, 21 to 28, 21 to 27, 21 to 26, 21 to 25, 21 to 24, 21 to 23, or 21 to 22 nucleotides, for example 19 to 23 nucleotides in length or 21 to 23 nucleotides in length. Ranges and lengths intermediate to those described above are also considered part of the present disclosure.
In some embodiments, the duplex structure is 19 to 30 base pairs in length. Similarly, the region complementary 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, dsrnas are long enough to serve as substrates for Dicer enzymes. For example, it is well known in the art that dsrnas greater than about 21 to 23 nucleotides in length can be used as substrates for Dicer. As the skilled artisan will also recognize, the region targeted to the cleaved RNA will typically be part of a larger RNA molecule, typically an mRNA molecule. In related cases, a "portion" of an mRNA target is a contiguous sequence of the mRNA target that is sufficiently long to make it a substrate for RNAi-directed cleavage (i.e., cleavage via the RISC pathway).
Those of skill in the art will also recognize that the duplex region is a major functional portion of a dsRNA, e.g., a duplex region of about 19 to about 30 base pairs (e.g., about 19 to 30, 19 to 29, 19 to 28, 19 to 27, 19 to 26, 19 to 25, 19 to 24, 19 to 23, 19 to 22, 19 to 21, 19 to 20, 20 to 30, 20 to 29, 20 to 28, 20 to 27, 20 to 26, 20 to 25, 20 to 24,20 to 23, 20 to 22, 20 to 21, 21 to 30, 21 to 29, 21 to 28, 21 to 27, 21 to 26, 21 to 25, 21 to 24, 21 to 23, or 21 to 22 pairs). Thus, in one embodiment, the RNA molecule or RNA molecule complex having a duplex region of greater than 30 base pairs is a dsRNA to the extent that it is processed into a functional duplex of, for example, 15 to 30 base pairs that targets the desired RNA for cleavage. Thus, one of ordinary skill will recognize that in one embodiment, the miRNA is dsRNA. In another embodiment, the dsRNA is not a naturally occurring miRNA. In another embodiment, an iRNA agent useful for targeting TMPRSS6 gene expression is not produced in a target cell by cleavage of a larger dsRNA.
The dsRNA as described herein may further comprise one or more single-stranded nucleotide overhangs, e.g., 1 to 4, 2 to 4, 1 to 3, 2 to 3, 1, 2, 3, or 4 nucleotides. Dsrnas having at least one nucleotide overhang have better inhibitory properties relative to their blunt-ended counterparts. Nucleotide overhangs may include or consist of: nucleotide/nucleoside analogs comprising deoxynucleotides/nucleosides. The overhang may be on the sense strand, the antisense strand, or any combination thereof. Furthermore, the overhanging nucleotides may be present on the 5 'end, 3' end or both ends of the antisense strand or sense strand of the dsRNA.
dsRNA can be synthesized by standard methods known in the art. Double stranded RNAi compounds of the invention can be prepared using a two-step procedure. First, individual strands of a double-stranded RNA molecule are prepared separately. The component chains are then annealed. The individual strands of the siRNA compound may be prepared using either solution phase or solid phase organic synthesis or both. Organic synthesis offers the advantage that oligonucleotide chains comprising non-natural or modified nucleotides can be readily prepared. Similarly, single stranded oligonucleotides of the invention may be prepared using either solution phase or solid phase organic synthesis or both.
In one aspect, the dsRNA of the invention comprises at least two nucleotide sequences, a sense sequence and an antisense sequence. The sense strand is selected from the group consisting of the sequences provided in any of tables 2-7, and the corresponding antisense strand of the sense strand is selected from the group consisting of the sequences in any of tables 2-7. In this regard, one of the two sequences is complementary to the other of the two sequences, wherein one of the sequences is substantially complementary to an mRNA sequence produced in TMPRSS6 gene expression. Thus, in this regard, a dsRNA will comprise two oligonucleotides, one of which is described as the sense strand in any one of tables 2-7, and the second oligonucleotide is described as the corresponding antisense strand of the sense strand in any one of tables 2-7.
In certain embodiments, the substantially complementary sequence of the dsRNA is contained on an isolated oligonucleotide. In other embodiments, the substantially complementary sequence of the dsRNA is contained on a single oligonucleotide.
In one embodiment, the antisense strand comprises at least 15, e.g., 15, 16, 17, 18, 19, or 20 consecutive nucleotides that differ by no more than 0, 1, 2, or 3 nucleotides from any of the antisense strand nucleotide sequences in any of tables 2-7.
It should be appreciated that, for example, although the sequences in tables 3 or 5 are not described as modified or conjugated sequences, the RNAs of the invention, e.g., the dsRNA of the invention, may include any of the unmodified, unconjugated, or differently modified or conjugated sequences described therein, as described in any of tables 2-7. In other words, the invention encompasses dsRNA of tables 2-7, which are unmodified, unconjugated, modified or conjugated, as described herein.
It is well known to those skilled in the art that dsRNAs having duplex structures of about 20 to 23 base pairs (e.g., 21 base pairs) have been known to be particularly effective in inducing RNA interference (Elbashir et al, J. European molecular biological tissue (EMBO) 2001, 20:6877-6888). However, others have found that shorter or longer RNA duplex structures may also be effective (Chu and Rana (2007) RNA 14:1714-1719; kim et al, (2005) Nature Biotech (Nat Biotech) 23:222-226). In the above examples, the dsRNA described herein may comprise at least one strand of at least 21 nucleotides in length due to the nature of the oligonucleotide sequences provided in any of tables 2-7. It is reasonably expected that shorter duplexes with any one of the sequences of any one of tables 2-7 minus only few nucleotides at one or both ends may be similarly effective compared to the dsRNA described above. Thus, a dsRNA having a sequence of at least 19, 20, or more consecutive nucleotides derived from any one of the sequences of any one of tables 2-7, and whose ability to inhibit TMPRSS6 gene expression differs from a dsRNA comprising the complete sequence by no more than about 5%, 10%, 15%, 20%, 25%, or 30% inhibition, is considered to be within the scope of the present invention.
In addition, the RNAs provided in tables 2-7 identify sites in TMPRSS6 transcripts that are susceptible to RISC-mediated cleavage. Thus, the invention further features targeting an iRNA within one of these sites. As used herein, an iRNA is said to be within a particular site of a targeted RNA transcript if it facilitates cleavage of the transcript at any position within the particular site. Such irnas typically comprise at least about 19 contiguous nucleotides from any one of the sequences provided in any one of tables 2-7, coupled with additional nucleotide sequences taken from contiguous regions of a selected sequence in the TMPRSS6 gene.
III modified iRNA of the invention
In certain embodiments, the RNA, e.g., dsRNA, of the iRNA of the invention is unmodified and does not include chemical modifications or conjugation, e.g., as known in the art and described herein. In other embodiments, the RNAs, e.g., dsRNA, of the iRNA of the invention are chemically modified to enhance stability or other beneficial properties. 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 nucleotides of the iRNA or substantially all nucleotides of the iRNA are modified, i.e., no more than 5, 4, 3, 2, or 1 unmodified nucleotides are present in the strand of the iRNA.
The nucleic acids of the features of the invention may be synthesized or modified by art-recognized methods, such as those described in "Current protocols in nucleic acid chemistry (Current protocols in nucleic acid chemistry)," Beaucage, S.L et al, (editions), john Willi parent, N.Y., U.S. A., incorporated herein by reference. Modifications include, for example, terminal modifications, such as 5 'terminal modifications (phosphorylation, conjugation, reverse ligation, etc.), 3' terminal modifications (conjugation, DNA nucleotides, reverse ligation, etc.); base modification, e.g., substitution, removal (no base nucleotides) or conjugation of bases with stable bases, destabilizing bases or bases base pairing with extended partner pools; sugar modification (e.g., at the 2 'position or the 4' position) or sugar substitution; or backbone modification, including modification or substitution of phosphodiester bonds. Specific examples of iRNA compounds that can be used in the embodiments described herein include, but are not limited to, RNAs that contain modified backbones or do not contain natural internucleoside linkages. In addition, RNAs having modified backbones include those that do not have a phosphorus atom in the backbone. For the purposes of this specification, and as sometimes referred to in the art, modified RNAs that do not have phosphorus atoms in their internucleoside backbones can also be considered oligonucleotides. In some embodiments, the modified iRNA will have a phosphorus atom in its internucleoside backbone.
Modified RNA backbones include, for example, phosphorothioates, chiral phosphorothioates, phosphorodithioates, phosphotriesters, aminoalkyl phosphotriesters, methyl and other alkylphosphonates (including 3 '-alkylene phosphonates and chiral phosphonates), phosphinates, phosphoramidates (including 3' -phosphoramidate and aminoalkyl amine phosphates), thiocarbonylphosphoramidates, thiocarbonylalkyl phosphonates, thiocarbonylalkyl phosphotriesters, and borane phosphates with normal 3'-5' linkages, 2'-5' linked analogs of these esters, and those with reversed polarity, wherein adjacent pairs of nucleoside units are linked in 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 agent of the invention is in the free acid form. In other embodiments of the invention, the dsRNA agents of the invention are in salt form. In one embodiment, the dsRNA agent of the invention is in the form of a sodium salt. In certain embodiments, when the dsRNA agents of the invention are in the form of sodium salts, sodium ions are present in the agent as counter ions to substantially all of the phosphodiester and/or phosphorothioate groups present in the agent. Agents in which substantially all of the phosphodiester and/or phosphorothioate linkages have a sodium counterion comprise no more than 5, 4, 3, 2, or 1 phosphodiester and/or phosphorothioate linkages that do not have a sodium counterion. In some embodiments, when the dsRNA agents of the invention are in the form of sodium salts, sodium ions are present in the agent as counter ions to all phosphodiester and/or phosphorothioate groups present in the agent.
Representative U.S. patents teaching the preparation of the above-described phosphorus-containing bonds include, but are not limited to, U.S. Pat. nos. 3,687,808; 4,469,863; 4,476,301; no. 5,023,243; 5,177,195; 5,188,897; 5,264,423; U.S. Pat. No. 5,276,019; no. 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. patent RE39464, the entire contents of each of which are hereby incorporated by reference.
Wherein the modified RNA backbone that does not comprise a phosphorus atom has a backbone formed by: short chain alkyl or cycloalkyl internucleoside linkages, mixed heteroatoms and alkyl or cycloalkyl internucleoside linkages, or one or more short chain heteroatoms or heterocyclic internucleoside linkages. These include those having morpholino linkages (formed in part from the sugar moiety of the nucleoside); a siloxane backbone; sulfide, sulfoxide, and sulfone backbones; formylacetyl and thioacetylacetyl backbones; methylene formylacetyl and thioformylacetyl backbones; an olefin-containing backbone; a sulfamate backbone; methylene imino and methylene hydrazino backbones; sulfonate and sulfonamide backbones; an amide backbone; and other N, O, S and CH with mixing 2 Backbone of the component parts.
Representative U.S. patents teaching the preparation of the above oligonucleotides 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; no. 5,489,677; 5,541,307; 5,561,225; 5,596,086; no. 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 by reference.
It is contemplated that suitable RNA mimics are used in the iRNA provided herein, wherein the sugar and internucleoside linkages of the nucleotide units, i.e., the backbone, are replaced with new groups. The base unit is maintained to hybridize to the appropriate nucleic acid target compound. One such oligomeric compound, in which RNA mimics that have been shown to have excellent hybridization properties, is known as Peptide Nucleic Acid (PNA). In PNA compounds, the sugar backbone of RNA is replaced by an amide containing backbone, especially an aminoethylglycine backbone. The nucleobases are retained and bound directly or indirectly to the aza nitrogen atoms of the amide moiety of the backbone. Representative U.S. patents teaching the preparation of PNA compounds include, but are not limited to, U.S. patent nos. 5,539,082; no. 5,714,331; and U.S. Pat. No. 5,719,262, the entire contents of each of which are hereby incorporated by reference. Additional PNA compounds suitable for use in the iRNA of the present invention are described, for example, in Nielsen et al Science 1991,254,1497-1500.
Some embodiments of the inventive features include RNAs having phosphorothioate backbones and oligonucleotides having heteroatom backbones, particularly- -CH of U.S. Pat. No. 5,489,677 cited above 2 --NH--CH 2 -、--CH 2 --N(CH 3 )--O--CH 2 - - [ known as methylene (methylimino) or MMI backbone ]],--CH 2 --O--N(CH 3 )--CH 2 --,--CH 2 --N(CH 3 )--N(CH 3 )--CH 2 -and-N (CH) 3 )--CH 2 --CH 2 - - - - -, and the amide backbone of U.S. Pat. No. 5,602,240, cited above. In some embodiments, the RNAs characterized herein have a morpholino backbone structure of U.S. patent No. 5,034,506 cited above. The natural phosphodiester backbone may be represented as O-P (O) (OH) -OCH2-.
The modified RNA may also include one or more substituted sugar moieties. An iRNA, e.g., dsRNA, as characterized herein may comprise one of the following at the 2' position: OH; f, performing the process; o-, S-or N-alkyl; o-, S-or N-alkenyl; o-, S-or N-alkynyl; or O-alkyl-O-alkyl, wherein alkyl, alkenyl and alkynyl groups may be substituted or unsubstituted C 1 To C 10 Alkyl or C 2 To C 10 Alkenyl and alkynyl groups. Exemplary suitable modifications include O [ (CH) 2 ) n O] m CH 3 、O(CH 2 )、 n OCH 3 、O(CH 2 ) n NH 2 、O(CH 2 ) n CH 3 、O(CH 2 ) n ONH 2 And O (CH) 2 ) n ON[(CH 2 ) n CH 3 )] 2 Wherein n and m are from 1 to about 10. In other embodiments, the dsRNA comprises one of the following at the 2' position: c (C) 1 To C 10 Lower alkyl, substituted lower alkyl, alkylaryl, arylalkyl, O-alkylaryl or O-arylalkyl, SH, SCH 3 、OCN、Cl、Br、CN、CF 3 、OCF 3 、SOCH 3 、SO 2 CH 3 、ONO 2 、NO 2 、N 3 、NH 2 A heterocycloalkyl group, a heterocycloalkyl aryl group, an aminoalkylamino group, a polyalkylamino group, a substituted silyl group, an RNA cleavage 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, as well as other substituents having similar properties. In some embodiments, the modification comprises 2 '-methoxyethoxy (2' -O- -CH) 2 CH 2 OCH 3 ) Also known as 2'-O- (2-methoxyethyl) or 2' -MOE (Martin et al, proc. Swiss chemistry (Helv. Chim. Acta), 1995, 78:486-504), i.e., alkoxy-)An alkoxy group. Another exemplary modification is 2' -dimethylaminooxyethoxy, i.e., O (CH) 2 ) 2 ON(CH 3 ) 2 Groups, also known as 2' -DMAEE, as described in the examples below, and 2' -dimethylaminoethoxyethoxy (also known in the art as 2' -O-dimethylaminoethoxyethyl or 2' -DMAEOE), i.e., 2' -O- -CH 2 --O--CH 2 --N(CH 3 ) 2 . Additional exemplary modifications include: 5'-Me-2' -F nucleotide, 5'-Me-2' -OMe nucleotide, 5'-Me-2' -deoxynucleotide (both R and S isomers in these three families); 2' -alkoxyalkyl; and 2' -NMA (N-methylacetamide).
Other modifications include 2 '-methoxy (2' -OCH) 3 ) 2 '-aminopropoxy (2' -OCH) 2 CH 2 CH 2 NH 2 ) And 2 '-fluoro (2' -F). Similar modifications can also be made at other positions on the RNA of the iRNA, particularly at the 3 'position of the sugar on the 3' terminal nucleotide or at the 5 'position of the 2' -5 'linked dsRNA and 5' terminal nucleotide. The iRNA may also have a glycomimetic, such as a cyclobutyl moiety in place of the pentose sugar. Representative U.S. patents teaching the preparation of such modified sugar structures include, but are not limited to, U.S. patent No. 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, some of which are common to the present application. The entire contents of each of the foregoing are hereby incorporated by reference.
iRNA may also include nucleobase (commonly referred to in the art simply as "base") modifications or substitutions. As used herein, "unmodified" or "natural" nucleobases comprise the purine bases adenine (a) and guanine (G), as well as the pyrimidine bases thymine (T), cytosine (C) and uracil (U). Modified nucleobases include other synthetic and natural nucleobases such as deoxythymine (dT), 5-methylcytosine (5-me-C), 5-hydroxymethylcytosine, 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-propynyluracil and cytosine, 6-azouracil, cytosine and thymine, 5-uracil (pseudouracil), 4-thiouracil, 8-halo, 8-amino, 8-thio, 8-hydroxy and other 8-substituted adenine and guanine, 5-halo, in particular 5-bromo, 5-trifluoromethyl and other 5-substituted uracil and cytosine, 7-methylguanine and 7-methyladenine, 8-azaguanine and 8-azaguanine, 7-deaza and 7-adenine and 3-deaza. Additional nucleobases include those disclosed in U.S. Pat. No. 3,687,808, modified nucleosides in biochemistry, biotechnology and medicine (Modified Nucleosides in Biochemistry, biotechnology and Medicine), herdewijn, P. Editions, wiley-VCH Press (Wiley-VCH), 2008; those disclosed in polymeric science and engineering encyclopedia (The Concise Encyclopedia Of Polymer Science And Engineering), pages 858-859, kroschwitz, j.l editions, john wili father, 1990, englisch et al, chemistry applications international edition (Angewandte Chemie, international Edition), 1991,30,613, and Sanghvi, Y s, chapter 15, dsRNA research and applications (dsRNA Research and Applications), pages 289-302, croake, s.t. and lebleuu, b. editions, CRC Press, 1993. Certain of these nucleobase pairs are particularly useful in increasing the binding affinity of oligomeric compounds that are characteristic of the application. 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 ℃ to 1.2 ℃ (Sanghvi, y.s., rooke, s.t., and Lebleu, b. editions, dsRNA research and applications, CRC press, boca Raton, 1993, pages 276-278), and are exemplary base substitutions, even more particularly when combined with 2' -O-methoxyethyl sugar modifications.
Representative U.S. patents teaching the preparation of certain of the above-described modified nucleobases, as well as other modified nucleobases, include, but are not limited to, U.S. Pat. nos. 3,687,808, 4,845,205, supra; 5,130,30; 5,134,066; 5,175,273; 5,367,066; 5,432,272; 5,457,187; no. 5,459,255; 5,484,908; 5,502,177; 5,525,711; 5,552,540; 5,587,469; 5,594,121 and 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 by reference.
In some embodiments, RNAi agents of the present disclosure can also be modified to include one or more bicyclic sugar moieties. A "bicyclic sugar" is a ring-modified furanosyl ring formed by bridging two carbons, whether adjacent or not. A "bicyclic nucleoside" ("BNA") is a nucleoside having a sugar moiety that includes a ring formed by bridging two carbons of the sugar ring (whether adjacent or non-adjacent), thereby forming a bicyclic system. In certain embodiments, the bridge optionally connects the 4' -carbon and the 2' -carbon of the sugar ring through a 2' -acyclic oxygen atom. Thus, in some embodiments, the agents of the invention may comprise one or more Locked Nucleic Acids (LNAs). Locked nucleic acids are nucleotides having a modified ribose moiety, where the ribose moiety includes an additional bridge connecting the 2 'and 4' carbons. In other words, LNA is a nucleotide comprising a bicyclic sugar moiety comprising 4' -CH 2 -O-2' bridge. This structure effectively "locks" the ribose in the 3' -internal structure conformation. Addition of locked nucleic acids to siRNA has been shown to increase siRNA stability in serum and reduce off-target effects (Elmen, J. Et al, (2005) nucleic acids research (Nucleic Acids Research) 33 (1): 439-447; mook, OR. Et al, (2007) molecular cancer therapeutics (Mol Canc Ther)6 (3) 833-843; grunwiller, A. Et al, (2003) nucleic acids research 31 (12): 3185-3193). Examples of bicyclic nucleosides for polynucleotides of the invention include, but are not limited to nucleosides that include a bridge between the 4 'and 2' ribosyl ring atoms. In certain embodiments, antisense polynucleotide agents of the invention comprise one or more bicyclic nucleosides comprising a 4 'to 2' bridge.
The locked nucleoside can be represented by the following structure (stereochemistry omitted),
wherein B is a nucleobase or modified nucleobase and L is a linking group linking 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' - (CH) 2 )—O-2'(LNA);4'-(CH 2 )—S-2';4'-(CH 2 ) 2 —O-2'(ENA);4'-CH(CH 3 ) -O-2 '(also referred to as "constrained ethyl" or "cEt") and 4' -CH (CH) 2 OCH 3 ) -O-2' (and analogues thereof; see, for example, U.S. patent No. 7,399,845); 4' -C (CH) 3 )(CH 3 ) -O-2' (and analogues thereof; see, for example, U.S. patent No. 8,278,283); 4' -CH 2 —N(OCH 3 ) -2' (and analogues thereof; see, for example, U.S. patent No. 8,278,425); 4' -CH 2 —O—N(CH 3 ) -2' (see, e.g., U.S. patent publication No. 2004/0171570); 4' -CH 2 -N (R) -O-2', wherein R is H, C1-C12 alkyl or a nitrogen protecting group (see e.g. us patent 7,427,672); 4' -CH 2 —C(H)(CH 3 ) -2' (see e.g. chattopladhyaya et al, journal of organic chemistry (j. Org. Chem.)), 2009,74,118-134; 4' -CH 2 —C(═CH 2 ) -2' (and analogues thereof; see, for example, U.S. patent No. 8,278,426). The entire contents of each of the foregoing are hereby incorporated by reference.
Other representative U.S. patents and U.S. patent 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; no. 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; US2008/0039618; and US2009/0012281, the entire contents of each of which are hereby incorporated 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).
The RNA of the iRNA may also be modified to include one or more constrained ethyl nucleotides. As used herein, "constrained ethyl nucleotide" or "cEt" is intended to include a nucleotide comprising 4' -CH (CH 3 ) -a locked nucleic acid of the bicyclic sugar moiety of the O-2' bridge (i.e. L in the preceding structure). In one embodiment, the constrained ethyl nucleotide is in an S conformation referred to herein as "S-cEt".
The iRNA of the invention may also comprise one or more "conformational restriction nucleotides" ("CRNs"). CRNs are nucleotide analogs having a linker linking the C2' and C4' carbons of ribose or the C3 and-C5 ' carbons of ribose. CRN locks the ribose ring in a stable conformation and increases the hybridization affinity to mRNA. The linker is of sufficient length to place the oxygen in the optimal position for stability and affinity, resulting in less ribose ring wrinkling.
Representative publications that teach the preparation of certain CRNs described above include, but are not limited to, U.S. patent publication No. 2013/0190383; and PCT publication WO 2013/036868, the entire contents of each of which are hereby incorporated by reference.
In some embodiments, an iRNA of the invention includes one or more monomers that are UNA (unlocking nucleic acid) nucleotides. UNA is an unlocked acyclic nucleic acid in which any bonds of the sugar have been removed, forming an unlocked "sugar" residue. In one example, UNA also encompasses monomers where the bond between C1'-C4' has been removed (i.e., covalent carbon-oxygen-carbon bonds between C1 'and C4' carbons). In another example, the C2'-C3' bond of the sugar (i.e., the covalent carbon-carbon bond between the C2 'and C3' carbons) has been removed (see nucleic acid seminar cluster (nuc. Acids Symp. Series), 52,133-134 (2008) and fluidizer et al, molecular biological systems (mol. Biosystem.), 2009,10,1039, hereby incorporated by reference).
Representative U.S. disclosures teaching the preparation of UNA include, but are not limited to, U.S. patent No. 8,314,227; U.S. patent publication No. 2013/0096289; 2013/0011922; and 2011/0313020, the entire contents of each of which are hereby incorporated by reference.
Potentially stable modifications to the ends of RNA molecules may include N- (acetamidohexanoyl) -4-hydroxyproline (Hyp-C6-NHAc), N- (hexanoyl-4-hydroxyproline) (Hyp-C6), N- (acetyl-4-hydroxyproline) (Hyp-NHAc), thymidine-2 '-O-deoxythymidine (ether), N- (aminohexanoyl) -4-hydroxyproline (Hyp-C6-amino), 2-behenoyl-uridine-3' -phosphate, reverse 2 '-deoxymodified ribonucleotides such as reverse dT (idT), reverse dA (idA), reverse abasic 2' -deoxyribonucleotide (iAb), and the like. The disclosure of this modification can be found in WO 2011/005861.
In one example, the 3 'or 5' end of the oligonucleotide is linked to an inverted 2 '-deoxymodified ribonucleotide, such as an inverted dT (idT), an inverted dA (idA) or an inverted abasic 2' -deoxyribonucleotide (iAb). In one particular example, the reverse 2' -deoxy modified ribonucleotide is linked to the 3' -end of an oligonucleotide, as described herein, the 3' -end of the sense strand, wherein the linkage is through a 3' -3' -phosphodiester linkage or a 3' -3' -phosphorothioate linkage.
In another example, the 3' end of the sense strand is linked to an inverted abasic ribonucleotide (iAb) via a 3' -3' -phosphorothioate linkage. In another example, the 3' end of the sense strand is linked to the inverted dA (idA) via a 3' -3' -phosphorothioate linkage.
In one particular example, the reverse 2' -deoxy modified ribonucleotide is linked to the 3' -end of an oligonucleotide, as described herein, the 3' -end of the sense strand, wherein the linkage is through a 3' -3' -phosphodiester linkage or a 3' -3' -phosphorothioate linkage.
In another example, the 3' -terminal nucleotide of the sense strand is an inverted dA (idA) and is linked to the aforementioned nucleotide by a 3' -3' -linkage (e.g., a 3' -3' -phosphorothioate linkage).
Other modifications of the nucleotides of the iRNA of the invention include 5' phosphates or 5' phosphate mimics, e.g., 5' terminal phosphates or phosphate mimics on the antisense strand of the iRNA. Suitable phosphate ester mimetics are disclosed, for example, in U.S. patent publication 2012/0157511, the entire contents of which are incorporated herein by reference.
A. Modified iRNA comprising the motif of the invention
In certain aspects of the invention, the double stranded RNA agents of the invention comprise agents having 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, excellent results can be obtained by introducing one or more motifs with three identical modifications on three consecutive nucleotides into the sense strand or antisense strand of a dsRNAi agent, particularly at or near the cleavage site. In some embodiments, the sense and antisense strands of the dsRNAi agent can be fully modified in other ways. The introduction of these motifs interrupts the modification pattern of the sense or antisense strand, if present. The dsRNAi agent can optionally be conjugated to GalNAc derivative ligand, e.g., on the sense strand.
More specifically, gene silencing activity of a dsRNAi agent is observed when the sense and antisense strands of the dsRNAi agent are fully modified to have three identically modified one or more motifs on three consecutive nucleotides at or near the cleavage site of at least one strand of the dsRNAi agent.
Accordingly, the present invention provides a double-stranded RNA agent capable of inhibiting expression of a target gene (i.e., TMPRSS6 gene) in vivo. RNAi agents include a sense strand and an antisense strand. Each strand of the RNAi agent can be, for example, 17 to 30 nucleotides in length, 25 to 30 nucleotides in length, 27 to 30 nucleotides in length, 19 to 25 nucleotides in length, 19 to 23 nucleotides in length, 19 to 21 nucleotides in length, 21 to 25 nucleotides in length, or 21 to 23 nucleotides in length.
The sense and antisense strands typically form duplex double-stranded RNAs ("dsRNA"), also referred to herein as "dsRNAi agents. The duplex region of the dsRNAi agent can be, for example, 27 to 30 nucleotide pairs in length, 19 to 25 nucleotide pairs in length, 19 to 23 nucleotide pairs in length, 19 to 21 nucleotide pairs in length, 21 to 25 nucleotide pairs in length, or 21 to 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 can contain one or more overhanging regions or end capping groups at the 3 'end, 5' end, or both of one or both strands. The overhangs may independently be 1 to 6 nucleotides in length, for example 2 to 6 nucleotides in length, 1 to 5 nucleotides in length, 2 to 5 nucleotides in length, 1 to 4 nucleotides in length, 2 to 4 nucleotides in length, 1 to 3 nucleotides in length, 2 to 3 nucleotides in length, or 1 to 2 nucleotides in length. In certain embodiments, the protruding end region may comprise an extended protruding end region as provided above. An overhang may be the result of one strand being longer than the other, or the result of two strands of the same length being interleaved. The overhang may form a mismatch with the target mRNA, or it may be complementary to the gene sequence being targeted, or it may be another sequence. The first strand and the second strand may also be linked, for example by additional bases to form a hairpin, or by other non-base linkers.
In certain embodiments, the nucleotides in the protruding end region of the dsRNAi agent can each independently be a modified or unmodified nucleotide, including but not limited to 2 '-sugar modified, such as 2' -F, 2 '-O-methyl, thymidine (T), 2' -O-methoxyethyl-5-methyluridine (Teo), 2 '-O-methoxyethyl adenosine (Aeo), 2' -O-methoxyethyl-5-methylcytidine (m 5 Ceo), and any combination thereof.
For example, TT may be an overhang sequence at either end of either strand. The overhang may form a mismatch with the target mRNA, or it may be complementary to the gene sequence being targeted, or it may be another sequence.
The 5 '-or 3' -overhang on the sense, antisense, or both strands of the dsRNAi agent can be phosphorylated. In some embodiments, the overhang region contains two nucleotides with phosphorothioates between the two nucleotides, where the two nucleotides may be the same or different. In some embodiments, the overhang is present at the 3' end of the sense strand, the antisense strand, or both strands. In some embodiments, such 3' -overhangs are present in the antisense strand. In some embodiments, such a 3' -overhang is present in the sense strand.
dsRNAi agents may contain only a single overhang, which may enhance the interfering activity of RNAi without affecting its overall stability. For example, a single stranded overhang may be located at the 3 'end of the sense strand, or alternatively, at the 3' end of the antisense strand. 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) and vice versa. Typically, the antisense strand of a 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 the 3' overhang of the antisense strand facilitate the loading of the guide strand into the RISC process.
In certain embodiments, the dsRNAi agent is a double-ended passivating agent 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, 9 starting 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 starting from the 5' end.
In other embodiments, the dsRNAi agent is a double-ended passivating agent 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 starting 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 starting from the 5' end.
In still other embodiments, the dsRNAi agent is a double-ended passivating agent 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, 11 starting 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 starting from the 5' end.
In certain embodiments, a 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 starting from the 5' end, wherein one end of the RNAi agent is blunt and the other end comprises a 2 nucleotide overhang. In one embodiment, the 2 nucleotide overhang is located at the 3' end of the antisense strand.
When a 2 nucleotide overhang is located at the 3' end of the antisense strand, there may be two phosphorothioate internucleotide linkages between the terminal three nucleotides, two of which are the overhang nucleotides and the third is the pairing nucleotide next to the overhang nucleotide. In one embodiment, the RNAi agent has two additional phosphorothioate internucleotide linkages between the terminal three nucleotides of both the 5 'end of the sense strand and the 5' end of the antisense strand. In certain embodiments, each nucleotide in the sense and antisense strands of a dsRNAi agent, including a nucleotide that is part of a motif, is a modified nucleotide. In certain embodiments, each residue is independently modified with 2 '-O-methyl or 3' -fluoro, e.g., in alternating motifs. Optionally, the dsRNAi agent further comprises a ligand (e.g., galNAc 3 )。
In certain embodiments, the dsRNAi agent comprises a sense strand and an antisense strand, wherein the sense strand is 25-30 nucleotide residues in length, starting from nucleotide 1 to 23 at the 5' end of the first strand (position 1), comprising at least 8 ribonucleotides; the antisense strand is 36 to 66 nucleotide residues in length and, starting from the 3' terminal nucleotide, comprises at least 8 ribonucleotides in the position that pairs with positions 1 to 23 of the sense strand to form a duplex; wherein at least the 3' terminal nucleotide of the antisense strand is unpaired with the sense strand and at most 6 consecutive 3' terminal nucleotides are unpaired with the sense strand, thereby forming a 1 to 6 nucleotide 3' single stranded overhang; wherein the 5 'end of the antisense strand comprises 10 to 30 consecutive nucleotides that are unpaired with the sense strand, thereby forming a 10 to 30 nucleotide single strand 5' overhang; wherein when the sense strand and the antisense strand are aligned for maximum complementarity, at least the 5 'end and 3' end nucleotides of the sense strand are paired with the nucleotide bases of the antisense strand, thereby forming a substantially double-stranded region between the sense strand and the antisense strand; and the antisense strand is sufficiently complementary to the target RNA along at least 19 ribonucleotides of the antisense strand length to reduce target gene expression upon introduction of the double-stranded nucleic acid into a mammalian cell; and wherein the sense strand contains at least one motif modified by three 2' -F on three consecutive nucleotides, wherein 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 at or near the cleavage site at three consecutive nucleotides.
In certain embodiments, the dsRNAi agent comprises a sense strand and an antisense strand, wherein the dsRNAi agent comprises a first strand of at least 25 and up to 29 nucleotides in length and a second strand of up to 30 nucleotides in length, having at least one motif with three 2 '-O-methyl modifications on three consecutive nucleotides at positions 11, 12, 13 from the 5' end; wherein the 3 'end of the first strand and the 5' end of the second strand form blunt ends and the second strand is 1 to 4 nucleotides longer than the first strand at its 3 'end, wherein the duplex region is at least 25 nucleotides in length and the second strand is sufficiently complementary to a target mRNA along at least 19 nucleotides of the second strand length to reduce target gene expression upon introduction of the RNAi agent into a mammalian cell, and wherein Dicer cleavage of the dsRNAi agent produces siRNA comprising the 3' end of the second strand, thereby reducing expression of the target gene in a mammal. Optionally, the dsRNAi agent further comprises a ligand.
In certain embodiments, the sense strand of the dsRNAi agent contains at least one motif with three identical modifications on three consecutive nucleotides, one of which is present at a cleavage site in the sense strand.
In certain embodiments, the antisense strand of a dsRNAi agent can also contain at least one motif of three identical modifications on three consecutive nucleotides, with one motif occurring at or near the cleavage site in the antisense strand.
For dsRNAi agents having duplex regions of 19 to 23 nucleotides in length, the cleavage site of the antisense strand is typically near the 10, 11, and 12 positions from the 5' end. Thus, three identical modified motifs can occur at positions 9, 10, 11 of the antisense strand; 10. 11, 12 positions; 11. 12, 13 positions; 12. 13, 14 positions; or 13, 14, 15 positions, counting from the first nucleotide at the 5 'end of the antisense strand, or counting from the first paired nucleotide within the 5' duplex region of the antisense strand. The cleavage site in the antisense strand can also vary depending on the length of the duplex region of the dsRNAi agent from the 5' end.
The sense strand of a dsRNAi agent can contain at least one motif of three identical modifications at three consecutive nucleotides at the cleavage site of the strand; and the antisense strand can have at least one motif of three identical modifications at or near three consecutive nucleotides of the strand at the cleavage site. When the sense strand and the antisense strand form a dsRNA duplex, the sense strand and the antisense strand may be arranged such that one motif of three nucleotides on the sense strand overlaps one motif of three nucleotides on the antisense strand by at least one nucleotide, 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 a dsRNAi agent can contain more than one motif with three identical modifications on three consecutive nucleotides. The first motif may occur at or near the cleavage site of the strand, and the other motif may be a wing modification. The term "wing modification" herein refers to a motif occurring at another part of the strand, which part is separated from the motif at or near the cleavage site of the same strand. The wing modifications are either adjacent to the first motif or separated by at least one or more nucleotides. When the motifs are in close proximity to each other, then the chemical properties of the motifs are different from each other, and when the motifs are separated by one or more nucleotides, the chemical properties may be the same or different. There may be two or more wing modifications. For example, when two wing modifications are present, each wing modification may occur at one end relative to the first motif, which is located at or near the cleavage site, or on either side of the leader motif.
As with the sense strand, the antisense strand of a dsRNAi agent can contain more than one motif with three identical modifications on three consecutive nucleotides, with at least one motif occurring at or near the cleavage site of the strand. The antisense strand may also contain one or more wing modifications in an arrangement similar to the wing modifications that may be present on the sense strand.
In some embodiments, the wing modification on the sense or antisense strand of the dsRNAi agent generally does not comprise the first or first two terminal nucleotides at the 3', 5', or both ends of the strand.
In other embodiments, the wing modifications on the sense or antisense strand of a dsRNAi agent generally do not comprise the first or first two paired nucleotides within the duplex region at the 3', 5', or both ends of the strand.
When the sense and antisense strands of the dsRNAi agent each contain at least one flanking modification, the flanking modifications may fall on the same end of the duplex region and have an overlap of one, two, or three nucleotides.
When the sense and antisense strands of a dsRNAi agent each contain at least two winged modifications, the sense and antisense strands can be arranged such that the two modifications each from one strand fall at one end of the duplex region, with an overlap of one, two, or three nucleotides; two modifications each from one strand fall at the other end of the duplex region, with an overlap of one, two, or three nucleotides; two modified strands, one strand on each side of the leader, have an overlap of one, two or three nucleotides in the duplex region.
In some embodiments, each nucleotide in the sense and antisense strands of a dsRNAi agent, including a nucleotide that is part of a motif, can be modified. Each nucleotide may be modified with the same or different modifications, which modifications may comprise one or more changes of one or two non-linked phosphate oxygens or one or more linked phosphate oxygens; a change in the composition of ribose, e.g., a change in the 2' -hydroxyl group on ribose; large scale substitution of the phosphate moiety with a "dephosphorylation" linker; modification or substitution of naturally occurring bases; substitution or modification of the ribose-phosphate backbone.
Since nucleic acids are polymers of subunits, many modifications occur at repeated positions within the nucleic acid, such as modifications of bases or phosphate moieties, or non-linking O of phosphate moieties. In some cases, the modification will occur at all subject positions in the nucleic acid, but in many cases will not. For example, the modification may occur only at the 3 '-or 5' -terminal position, may occur only at the terminal region, e.g., at a position on the terminal nucleotide or in the last 2, 3, 4, 5 or 10 nucleotides of the strand. Modification may occur in the double-stranded region, the single-stranded region, or both. Modification may occur only in the double stranded region of the RNA, or may occur only in the single stranded region of the RNA. For example, phosphorothioate modifications at non-linked O positions may occur only at one or both ends, may occur only in the end regions, e.g., at positions on the end nucleotides or in the last 2, 3, 4, 5 or 10 nucleotides of the strand, or may occur in double-and single-stranded regions, particularly at the ends. One or more of the 5' ends may be phosphorylated.
For example, to enhance stability, a specific base may be included in the overhang, or a modified nucleotide or nucleotide substitute may be included in the single stranded overhang, such as in the 5 '-or 3' -overhang, or both. For example, it may be desirable to include purine nucleotides in the overhangs. In some embodiments, all or some of the bases in the 3 '-or 5' -overhang may be modified, e.g., by modification described herein. Modifications may include, for example, modifications at the 2' position of ribose sugar using modifications known in the art, for example, ribose with deoxyribonucleotide, 2' -deoxy-2 ' -fluoro (2 ' -F), or 2' -O-methyl modifications instead of nucleobases, and modifications of phosphate groups, for example phosphorothioate modifications. The overhangs need not be homologous to the target sequence.
In some embodiments, each residue of the sense and antisense strands is independently modified with LNA, CRN, cET, UNA, HNA, ceNA, 2' -methoxyethyl, 2' -O-methyl, 2' -O-allyl, 2' -C-allyl, 2' -deoxy, 2' -hydroxy, or 2' -fluoro. The chain may contain more than one modification. In one embodiment, each residue of the sense and antisense strands is independently modified with 2 '-O-methyl or 2' -fluoro.
There are typically at least two different modifications on the sense and antisense strands. These two modifications may be 2 '-O-methyl or 2' -fluoro modifications, or other modifications.
In certain embodiments, N a Or N b Including alternating patterns of modification. As used herein, the term "alternating motif" refers to a motif having one or more modifications, each modification occurring on alternating nucleotides of one strand. Alternate nucleotides may refer to one every other nucleotide or one every three nucleotides, or similar patterns. For example, if A, B and C each represent a type of modification of a nucleotide, the alternating motifs may be "ababababababab …", "AABBAABBAABB …", "aabababaabaab …", "AAABAAABAAAB …", "AAABBBAAABBB …" or "abccabcabc …", etc.
The types of modifications contained in the alternating motifs may be the same or different. For example, if A, B, C, D each represents a type of modification on a nucleotide, the alternating pattern, i.e., the modifications on every other nucleotide, may be identical, but each sense strand or antisense strand may be selected from several modification possibilities within the alternating motif, such as "ABABAB …", "ACACAC …", "bdbd …" or "CDCDCD …", etc.
In some embodiments, the dsRNAi agents of the invention include a modification pattern of an alternate motif on the sense strand that is offset relative to the modification pattern of an alternate motif on the antisense strand. The offset may be such that the modified group of nucleotides of the sense strand corresponds to a different modified group of nucleotides of the antisense strand, and vice versa. For example, when the sense strand is paired with the antisense strand in a dsRNA duplex, the alternating motifs in the sense strand may start with "abababa" from 5 'to 3' of the strand, and the alternating motifs 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 there is a complete or partial transfer of modification pattern between the sense and antisense strands.
In some embodiments, the dsRNAi agent comprises a pattern of alternating motifs of 2 '-O-methyl modification and 2' -F modification on the sense strand initially offset from the pattern of alternating motifs of 2 '-O-methyl modification and 2' -F modification on the antisense strand, i.e., 2 '-O-methyl modified nucleotides on the bases of the sense strand pair with 2' -F modified nucleotides on the antisense strand, and vice versa. The 1 position of the sense strand may start with a 2'-F modification and the 1 position of the antisense strand may start with a 2' -O-methyl modification.
Introducing one or more motifs of three identical modifications on three consecutive nucleotides into the sense strand or the antisense strand interrupts the initial modification pattern present in the sense strand or the antisense strand. The pattern of modification of the sense strand or antisense strand may be interrupted by introducing three identical modified one or more motifs on three consecutive nucleotides into the sense strand or antisense strand, which may enhance the gene silencing activity against the target gene.
In some embodiments, when three identically modified motifs on three consecutive nucleotides are introduced into any strand, the modification of the nucleotide next to the motif is a modification that is different from the modification of the motif. For example, the motif-containing sequence portion is "… N a YYYN b … ", wherein" Y "represents a modification of three identical modified motifs on three consecutive nucleotides, and" N a "AND" N b "means modification of a nucleotide beside the motif" YYY "other than Y, andwherein N is a And N b May be the same or different modifications. Alternatively, when wing modifications are present, N a Or N b May be present or absent.
The iRNA may further comprise at least one phosphorothioate or methylphosphonate internucleotide linkage. Phosphorothioate or methylphosphonate internucleotide linkage modifications may occur on any nucleotide of the sense strand, the antisense strand or both strands in any position of the strand. For example, internucleotide linkage modifications may occur on each nucleotide on the sense or antisense strand; each internucleotide linkage modification may occur in an alternating pattern on the sense strand or the antisense strand; or the sense or antisense strand may contain two internucleotide linkage modifications in an alternating pattern. The alternating pattern of internucleotide linkage modifications on the sense strand may be the same as or different from the antisense strand, and the alternating pattern of internucleotide linkage modifications on the sense strand may be offset relative to the alternating pattern of internucleotide linkage on the antisense strand. In one embodiment, the double stranded RNAi agent comprises 6 to 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 the 5 'end or the 3' end.
In some embodiments, the dsRNAi agent comprises phosphorothioate or methylphosphonate internucleotide linkage modifications in the protruding end region. For example, the overhang region may contain two nucleotides with phosphorothioate or methylphosphonate internucleotide linkages therebetween. Internucleotide linkage modifications may also be made to link the overhanging nucleotides to terminal pairing nucleotides within the duplex region. For example, at least 2, 3, 4 or all of the overhang nucleotides can be linked by phosphorothioate or methylphosphonate internucleotide linkages, and optionally, additional phosphorothioate or methylphosphonate internucleotide linkages can be present, linking the overhang nucleotide to the paired nucleotide next to the overhang nucleotide. For example, there may be at least two phosphorothioate internucleotide linkages between the terminal three nucleotides, two of which are the overhang nucleotides and the third is the pairing nucleotide adjacent to the overhang nucleotide. These terminal three nucleotides may be located 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, when a 2-nucleotide overhang is located 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 the paired nucleotide next to the overhang nucleotide. Optionally, the dsRNAi agent can additionally have two phosphorothioate internucleotide linkages between the terminal three nucleotides of both the 5 'end of the sense strand and the 5' end of the antisense strand.
In one embodiment, the dsRNAi agent comprises a mismatch to the target, within the duplex, or a combination thereof. The mismatch may occur in the overhang region or the duplex region. Base pairs may be ordered based on their propensity to promote dissociation or melting (e.g., based on the free energy of association or dissociation of a particular pairing, the simplest approach is to examine the pairing on a single pairing basis, although the next nearest point or similar analysis may also be used). In promoting dissociation: a is better than G and C; g is better than G and C; and I: C is better than G: C (i=inosine). Mismatches, such as non-canonical or canonical exopairs (as described elsewhere herein) are better than canonical (A: T, A: U, G: C) pairs; and pairing involving universal bases is preferred over canonical pairing.
In certain embodiments, the dsRNAi agent comprises at least one of the first 1, 2, 3, 4, or 5 base pairs within a duplex region at the 5' end of the antisense strand, said duplex region being independently selected from the group consisting of: a: u, G: U, I C and mismatch pairs, e.g., non-canonical or canonical exopairs or pairs that contain universal bases, to promote dissociation of the antisense strand at the 5' end of the duplex.
In certain embodiments, the nucleotide at position 1 within the duplex region starting 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 pairs within the duplex region starting from the 5' end of the antisense strand is an AU base pair. For example, the first base pair in 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 deoxythymine (dT), or the nucleotide at the 3' end of the antisense strand is deoxythymine (dT). For example, there is a short sequence of deoxythymidines, e.g., two dT nucleotides on the 3' end of the sense, antisense or both strands.
In certain embodiments, the sense strand sequence can be represented by formula (I):
5'n p -N a -(X X X) i -N b -Y Y Y-N b -(Z Z Z) j -N a -n q 3'(I)
wherein:
i and j are each independently 0 or 1;
p and q are each independently 0 to 6;
each N a Independently representing oligonucleotide sequences comprising from 0 to 25 modified nucleotides, each sequence comprising at least two different modified nucleotides;
each N b Independently represents an oligonucleotide sequence comprising 0 to 10 modified nucleotides;
each n p And n q Independently represent an overhang nucleotide;
wherein Nb and Y do not have the same modification; and is also provided with
XXX, YYY and ZZZ each independently represents a motif of three identical modifications on three consecutive nucleotides. In one embodiment, YYY is all 2' -F modified nucleotides.
In some embodiments, N a Or N b Including alternating patterns of modification.
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 to 23 nucleotides in length, the YYY motif can occur at or near the cleavage site of the sense strand (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), counting from the first nucleotide, starting from the 5' end; or optionally counting from the 5' end, from the first paired nucleotide within the duplex region.
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. Thus, the sense strand can be represented by the formula:
5'n p -N a -YYY-N b -ZZZ-N a -n q 3' (Ib);
5'n p -N a -XXX-N b -YYY-N a -n q 3' (Ic); or (b)
5'n p -N a -XXX-N b -YYY-N b -ZZZ-N a -n q 3' (Id)。
When the sense strand is represented by formula (Ib), N b Represents an oligonucleotide sequence comprising 0 to 10, 0 to 7, 0 to 5, 0 to 4, 0 to 2 or 0 modified nucleotides. Each N a An oligonucleotide sequence comprising 2 to 20, 2 to 15 or 2 to 10 modified nucleotides may be represented independently.
When the sense strand is represented by formula (Ic), N b Represents an oligonucleotide sequence comprising 0 to 10, 0 to 7, 0 to 5, 0 to 4, 0 to 2 or 0 modified nucleotides. Each N a An oligonucleotide sequence comprising 2 to 20, 2 to 15 or 2 to 10 modified nucleotides may be represented independently.
When the sense strand is represented by formula (Id), each N b Independently represent oligonucleotide sequences comprising 0 to 10, 0 to 7, 0 to 5, 0 to 4, 0 to 2 or 0 modified nucleotides. In one embodiment, N b 0, 1, 2, 3, 4, 5 or 6. Each N a An oligonucleotide sequence comprising 2 to 20, 2 to 15 or 2 to 10 modified nucleotides may be represented independently.
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 can be represented by the following formula:
5'n p -N a -YYY-N a -n q 3' (Ia)。
when the sense strand is represented by formula (Ia), each N a An oligonucleotide sequence comprising 2 to 20, 2 to 15 or 2 to 10 modified nucleotides may be represented independently.
In one embodiment, the antisense strand sequence of RNAi can be represented by formula (II):
5'n q' -N a '-(Z'Z'Z') k -N b '-Y'Y'Y'-N b '-(X'X'X') l -N' a -n p '3' (II)
wherein:
k and l are each independently 0 or 1;
p 'and q' are each independently 0 to 6;
each N a ' independently represents an oligonucleotide sequence comprising 0 to 25 modified nucleotides, each sequence comprising at least two different modified nucleotides;
each N b ' independently represents an oligonucleotide sequence comprising 0 to 10 modified nucleotides;
each n p ' and n q ' independently represents an overhang nucleotide;
wherein N is b 'and Y' do not have the same modification; and is also provided with
X ' X ' X ', Y ' Y ' Y ' and Z ' Z ' Z ' each independently represent a motif of three identical modifications on three consecutive nucleotides.
In some embodiments, N a ' or N b ' includes an alternating pattern of modification.
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 to 23 nucleotides in length, the Y' motif can occur at positions 9, 10, 11 of the antisense strand; 10. 11, 12; 11. 12, 13; 12. 13, 14; or 13, 14, 15, counting from the first nucleotide, from the 5' end; or optionally counting from the 5' end, from the first paired nucleotide within the duplex region. In one embodiment, the Y ' Y ' Y ' motif occurs at positions 11, 12, 13.
In certain embodiments, the Y 'Y' Y 'motifs are 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.
Thus, the antisense strand can be represented by the formula:
5'n q' -N a '-Z'Z'Z'-N b '-Y'Y'Y'-N a '-n p' 3' (IIb);
5'n q' -N a ′-Y′Y′Y′-N b ′-X′X′X′-n p' 3' (IIc); or (b)
5'n q' -N a ′-Z′Z′Z′-N b ′-Y′Y′Y′-N b ′-X′X′X′-N a ′-n p' 3' (IId)。
When the antisense strand is represented by formula (IIb), N b ' means an oligonucleotide sequence comprising 0 to 10, 0 to 7, 0 to 5, 0 to 4, 0 to 2 or 0 modified nucleotides. Each N a ' independently means an oligonucleotide sequence comprising 2 to 20, 2 to 15 or 2 to 10 modified nucleotides.
When the antisense strand is represented by formula (IIc), N b Represents an oligonucleotide sequence comprising 0 to 10, 0 to 7, 0 to 5, 0 to 4, 0 to 2 or 0 modified nucleotides. Each N a ' independently means an oligonucleotide sequence comprising 2 to 20, 2 to 15 or 2 to 10 modified nucleotides.
When the antisense strand is represented by formula (IId), each N b ' independently means an oligonucleotide sequence comprising 0 to 10, 0 to 7, 0 to 5, 0 to 4, 0 to 2 or 0 modified nucleotides. Each N a ' independently means an oligonucleotide sequence comprising 2 to 20, 2 to 15 or 2 to 10 modified nucleotides. In one embodiment,N b 0, 1, 2, 3, 4, 5 or 6.
In other embodiments, k is 0 and l is 0, and the antisense strand can be represented by the formula:
5'n p' -N a' -Y'Y'Y'-N a' -n q' 3' (Ia)。
when the antisense strand is represented by formula (IIa), each N a ' independently means an oligonucleotide sequence comprising 2 to 20, 2 to 15 or 2 to 10 modified nucleotides.
Each of X ', Y ', and Z ' may be the same or different from each other.
Each nucleotide of the sense and antisense strands may be independently modified with LNA, CRN, UNA, cEt, HNA, ceNA, 2 '-methoxyethyl, 2' -O-methyl, 2 '-O-allyl, 2' -C-allyl, 2 '-hydroxy or 2' -fluoro. For example, each nucleotide of the sense strand and the antisense strand is independently modified with 2 '-O-methyl or 2' -fluoro. Specifically, each X, Y, Z, X ', Y ' and Z ' may represent a 2' -O-methyl modification or a 2' -fluoro modification.
In some embodiments, when the duplex region is 21nt, the sense strand of the dsRNAi agent can contain YYY motifs present at positions 9, 10, and 11 of the strand, counting from the first nucleotide at the 5 'end, or optionally, counting from the first paired nucleotide within the duplex region at the 5' end; and Y represents a 2' -F modification. The sense strand may additionally contain an XXX motif or a ZZZ motif as a wing modification at the opposite end of the duplex region; and XXX and ZZZ each independently represent a 2'-OMe modification or a 2' -F modification.
In some embodiments, the antisense strand may contain a Y ' motif present at positions 11, 12, 13 of the strand, counting from the first nucleotide at the 5' end, or optionally, counting from the first paired nucleotide within the duplex region at the 5' end; and Y represents a 2' -O-methyl modification. The antisense strand may additionally contain an X 'motif or a Z' motif as a wing modification at opposite ends of the duplex region; and X 'X' X 'and Z' Z 'Z' each independently represent a 2'-OMe modification or a 2' -F modification.
The sense strand represented by any of the above formulas (Ia), (Ib), (Ic) and (Id) forms a duplex with the antisense strand represented by any of the formulas (IIa), (IIb), (IIc) and (IId), respectively.
Thus, a dsRNAi agent for use in the methods of the invention can comprise a sense strand and an antisense strand, each strand having 14 to 30 nucleotides, an iRNA duplex represented by formula (III):
sense: 5'n p -N a -(X X X) i -N b -Y Y Y-N b -(Z Z Z) j -N a -n q 3'
Antisense: 3' n p '-N a '-(X'X'X') k -N b '-Y'Y'Y'-N b '-(Z'Z'Z') l -N a '-n q '5'
(III)
Wherein:
i. j, k and l are each independently 0 or 1;
p, p ', q and q' are each independently 0 to 6;
each N a And N a ' independently represents an oligonucleotide sequence comprising 0 to 25 modified nucleotides, each sequence comprising at least two different modified nucleotides;
Each N b And N b ' independently represents an oligonucleotide sequence comprising 0 to 10 modified nucleotides;
wherein each n may or may not be present p '、n p 、n q ' and n q Independently represent an overhang nucleotide; and is also provided with
XXX, YYY, ZZZ, X ' X ' X ', Y ' Y ' Y ' and Z ' Z ' Z ' each independently represent a 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 i and j are both 0; or i and j are both 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 k and l are both 0; or k and l are both 1.
An exemplary combination of sense and antisense strands that form an iRNA duplex comprises the formula:
5'n p -N a -Y Y Y-N a -n q 3'
3'n p '-N a '-Y'Y'Y'-N a 'n q '5'
(IIIa)
5'n p -N a -Y Y Y-N b -Z Z Z-N a -n q 3'
3'n p '-N a '-Y'Y'Y'-N b '-Z'Z'Z'-N a 'n q '5'
(IIIb)
5'n p -N a -X X X-N b -Y Y Y-N a -n q 3'
3'n p '-N a '-X'X'X'-N b '-Y'Y'Y'-N a '-n q '5'
(IIIc)
5'n p -N a -X X X-N b -Y Y Y-N b -Z Z Z-N a -n q 3'
3'n p '-N a '-X'X'X'-N b '-Y'Y'Y'-N b '-Z'Z'Z'-N a -n q '5'
(IIId)
when the dsRNAi agent is represented by formula (IIIa), each N a Independently represent oligonucleotide sequences comprising 2 to 20, 2 to 15 or 2 to 10 modified nucleotides.
When the dsRNAi agent is represented by formula (IIIb), each N b Independently represent oligonucleotide sequences comprising 1 to 10, 1 to 7, 1 to 5 or 1 to 4 modified nucleotides. Each N a Independently represent oligonucleotide sequences comprising 2 to 20, 2 to 15 or 2 to 10 modified nucleotides.
When dsRNAi agent is represented by formula (IIIc), each N b 、N b ' independently means an oligonucleotide sequence comprising 0 to 10, 0 to 7, 0 to 5, 0 to 4, 0 to 2 or 0 modified nucleotides. Each N a Independently means including 2 to 20Oligonucleotide sequences of 2 to 15 or 2 to 10 modified nucleotides.
When the dsRNAi agent is represented by formula (IIId), each N b 、N b ' independently means an oligonucleotide sequence comprising 0 to 10, 0 to 7, 0 to 5, 0 to 4, 0 to 2 or 0 modified nucleotides. Each N a 、N a ' independently means an oligonucleotide sequence comprising 2 to 20, 2 to 15 or 2 to 10 modified nucleotides. N (N) a 、N a '、N b And N b Each of the' independently includes an alternating pattern of modification.
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 formulas (III), (IIIa), (IIIb), (IIIc), and (IIId), at least one Y nucleotide can form a base pair with at least one Y' nucleotide. Alternatively, at least two Y nucleotides form base pairs with corresponding Y' nucleotides; or all three Y nucleotides form base pairs with the corresponding Y' nucleotide.
When the dsRNAi agent is represented by formula (IIIb) or (IIId), at least one Z nucleotide can form a base pair with one Z' nucleotide. Alternatively, at least two Z nucleotides form base pairs with corresponding Z' nucleotides; or all three Z nucleotides form base pairs with the corresponding Z' nucleotide.
When the dsRNAi agent is represented by formula (IIIc) or (IIId), at least one X nucleotide can form a base pair with one X' nucleotide. Alternatively, at least two X nucleotides form base pairs with corresponding X' nucleotides; or all three X nucleotides form base pairs with the corresponding X' nucleotide.
In certain embodiments, the modification on the Y nucleotide is different from the modification on the Y ' nucleotide, the modification on the Z nucleotide is different from the modification on the Z ' nucleotide, or the modification on the X nucleotide is different from the modification on the X ' nucleotide.
In certain embodiments, when the dsRNAi agent is represented by formula (IIId), N a ModificationIs 2 '-O-methyl or 2' -fluoro. In other embodiments, when the RNAi agent is represented by formula (IIId), N a The modification is a 2 '-O-methyl or 2' -fluoro modification, and n p ′>0, and at least one n p ' via phosphorothioate linkages to adjacent nucleotide a. In still other embodiments, when the RNAi agent is represented by formula (IIId), N a The modification is a 2 '-O-methyl or 2' -fluoro modification, and n p ′>0, and at least one n p ' linked to adjacent nucleotides via phosphorothioate linkages, and the sense strand conjugated to one or more GalNAc derivatives linked via a divalent or trivalent branched linker (described below). In other embodiments, when the RNAi agent is represented by formula (IIId), N a The modification is a 2 '-O-methyl or 2' -fluoro modification, and n p ′>0, and at least one n p ' linked to adjacent nucleotides by phosphorothioate linkages, the sense strand comprises at least one phosphorothioate linkage, and the sense strand is conjugated to one or more GalNAc derivatives linked by a divalent or trivalent branched linker.
In some embodiments, when the dsRNAi agent is represented by formula (IIIa), N a The modification is a 2 '-O-methyl or 2' -fluoro modification, and n p ′>0, and at least one n p ' linked to adjacent nucleotides by phosphorothioate linkages, the sense strand comprises at least one phosphorothioate linkage, and the sense strand is conjugated to one or more GalNAc derivatives linked by a divalent or trivalent branched linker.
In some embodiments, the dsRNAi agent is a multimer comprising at least two duplex represented by formulas (III), (IIIa), (IIIb), (IIIc), and (IIId), wherein the duplex is connected by a linker. The linker may be cleavable or non-cleavable. Optionally, the multimer further comprises a ligand. Each duplex may target the same gene or two different genes; or each duplex may target the same gene at two different target sites.
In some embodiments, the dsRNAi agent is a multimer comprising three, four, five, six, or more duplex represented by formulas (III), (IIIa), (IIIb), (IIIc), and (IIId), wherein the duplex is connected by a linker. The linker may be cleavable or non-cleavable. Optionally, the multimer further comprises a ligand. Each duplex may target the same gene or two different genes; or each duplex may target the same gene at two different target sites.
In one embodiment, the 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 3' ends, and optionally conjugated to a ligand. Each agent may target the same gene or two different genes; or each agent may target the same gene at two different target sites.
In certain embodiments, RNAi agents of the invention can contain a small amount of nucleotides containing a 2 '-fluoro modification, e.g., 10 or fewer nucleotides with a 2' -fluoro modification. For example, an RNAi agent can contain 10, 9, 8, 7, 6, 5, 4, 3, 2, 1, or 0 nucleotides with 2' -fluoro modifications. In a specific embodiment, the RNAi agents of the invention contain 10 nucleotides with 2' -fluoro modifications, e.g., 4 nucleotides with 2' -fluoro modifications in the sense strand and 6 nucleotides with 2' -fluoro modifications in the antisense strand. In another specific embodiment, the RNAi agents of the invention contain 6 nucleotides with 2' -fluoro modifications, e.g., 4 nucleotides with 2' -fluoro modifications in the sense strand and 2 nucleotides with 2' -fluoro modifications in the antisense strand.
In other embodiments, RNAi agents of the invention can contain an ultra-low amount of nucleotides containing 2 '-fluoro modifications, e.g., 2 or fewer nucleotides containing 2' -fluoro modifications. For example, an RNAi agent can contain 2, 1 out of 0 nucleotides with 2' -fluoro modifications. In particular embodiments, the RNAi agent can contain 2 nucleotides with 2 '-fluoro modifications, e.g., 0 nucleotides with 2-fluoro modifications in the sense strand and 2 nucleotides with 2' -fluoro modifications 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. patent No. 7,858,769, WO2010/141511, WO2007/117686, WO2009/014887 and WO2011/031520, the entire contents of each of which are hereby incorporated by reference.
In certain embodiments, the compositions and methods of the present disclosure comprise a Vinyl Phosphonate (VP) modification of RNAi agents as described herein. In exemplary embodiments, the 5' vinylphosphonate modified nucleotides of the present disclosure have the following structure:
wherein X is O or S;
r is hydrogen, hydroxy, fluoro or C 1-20 Alkoxy (e.g., methoxy or n-hexadecyloxy);
R 5 ' is =c (H) -P (O) (OH) 2 And C5' carbon is with R 5 The double bond between' is in the E or Z direction (e.g., E direction); and is also provided with
B is a nucleobase or modified nucleobase, optionally wherein B is adenine, guanine, cytosine, thymine or uracil.
The vinyl phosphonate of the present disclosure may be linked to the antisense strand or sense strand of the dsRNA of the present disclosure. In certain embodiments, the vinyl phosphonate of the present disclosure is linked to the antisense strand of the dsRNA, optionally at the 5' end of the antisense strand of the dsRNA.
Vinyl phosphonate modifications are also contemplated for use in the compositions and methods of the present disclosure. Exemplary vinyl phosphonate structures include the foregoing structures, wherein R5' is =c (H) -OP (O) (OH) 2, and the double bond between the C5' carbon and R5' is in the E or Z direction (e.g., E direction).
As described in more detail below, an iRNA that contains one or more carbohydrate moieties conjugated to the iRNA can optimize one or more characteristics of the iRNA. In many cases, the carbohydrate moiety will be linked 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 (e.g., cyclic) carrier linked to a carbohydrate ligand. Ribonucleotide subunits in which the ribose sugar of the subunit has been so replaced are referred to herein as Ribose Replacement Modified Subunits (RRMS). The 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 heteroatoms, such as nitrogen, oxygen, sulfur. The cyclic carrier may be a single ring system or may contain two or more rings, such as fused rings. The cyclic support may be a fully saturated ring system or it may contain one or more double bonds.
The ligand may be linked to the polynucleotide by a vector. The carrier comprises (i) at least one "backbone attachment point", such as two "backbone attachment points", and (ii) at least one "tether attachment point". As used herein, "backbone attachment point" refers to a functional group, such as a hydroxyl group, or a bond that is generally useful and suitable for incorporating the carrier into a backbone, such as a phosphate or modified phosphate (e.g., sulfur-containing) backbone of ribonucleic acid. In some embodiments, "tethered attachment point" (TAP) refers to a constituent ring atom of a cyclic carrier that attaches to a selected moiety, such as a carbon atom or a heteroatom (other than the atom providing the backbone attachment point). The moiety may be, for example, a carbohydrate, such as a monosaccharide, disaccharide, trisaccharide, tetrasaccharide, oligosaccharide or polysaccharide. Optionally, the selected moiety is attached to the circular carrier via an intermediate tether. Thus, the cyclic support will typically comprise a functional group, such as an amino group, or will typically provide a bond suitable for binding or tethering of another chemical entity (e.g., a ligand that makes up a ring).
The iRNA may be conjugated to the ligand via a carrier, wherein the carrier may be a cyclic group or an acyclic group. In one embodiment, the cyclic group is selected from pyrrolidinyl, pyrazolinyl, pyrazolidinyl, imidazolinyl, imidazolidinyl, piperidinyl, piperazinyl, [1,3] dioxolane, oxazolidinyl, isoxazolidinyl, morpholinyl, thiazolidinyl, isothiazolidinyl, quinoxalinyl, pyridazinonyl, tetrahydrofuranyl, and decalin. In one embodiment, the acyclic group is a serine alcohol backbone or a diethanolamine backbone.
i. Thermally labile modifications
In certain embodiments, RNA interference of dsRNA molecules can be optimized by incorporating thermally labile modifications in the seed region of the antisense strand. As used herein, "seed region" means positions 2-9 of the 5 'end of the reference strand or positions 2-8 of the 5' end of the reference strand. For example, thermally labile modifications can be incorporated into the seed region of the antisense strand to reduce or inhibit off-target gene silencing.
The term "heat labile modification" encompasses the total melting temperature (T m ) T lower than dsRNA without such modification m Is modified by the above-mentioned modification(s). For example, a heat labile modification can modify the T of a dsRNA m Lowering the temperature by 1 to 4 degrees celsius, such as one, two, three, or four degrees celsius. Also, the term "thermally labile nucleotide" refers to a nucleotide containing one or more thermally labile modifications.
It has been found that dsRNA with an antisense strand comprising at least one thermostable modification of a duplex within the first 9 nucleotide positions, counting from the 5' end of the antisense strand, has the effect of reducing off-target gene silencing activity. Thus, in some embodiments, the antisense strand includes a thermally labile modification of at least one (e.g., one, two, three, four, five, or more) duplex within the first 9 nucleotide positions of the 5' region of the antisense strand. In some embodiments, the one or more thermally labile modifications of the duplex are located at positions 2 to 9, such as positions 4 to 8, from the 5' end of the antisense strand. In some further embodiments, the thermally labile modification of the duplex is located at position 6, 7, or 8 from the 5' end of the antisense strand. In still other embodiments, the thermostable modification of the duplex is located at position 7 from the 5' end of the antisense strand. In some embodiments, the thermostable modification of the duplex is located at position 2, 3, 4, 5 or 9 from the 5' end of the antisense strand.
The iRNA agent includes a sense strand and an antisense strand, each strand having 14 to 40 nucleotides. The RNAi agent can be represented by formula (L):
in formula (L), B1, B2, B3, B1', B2', B3', and B4' are each independently a modified nucleotide containing a nucleotide 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 a 2' -OMe modification. In one embodiment, B1, B2, B3, B1', B2', B3', and B4' each contain a 2'-OMe or 2' -F modification. In one embodiment, at least one of B1, B2, B3, B1', B2', B3', and B4' contains a 2' -O-N-methylacetamido (2 ' -O-NMA, 2' O-CH2C (O) N (Me) H) modification.
C1 is a thermally labile nucleotide located at a site opposite the seed region of the antisense strand (i.e., at positions 2-8 of the 5 'end of the antisense strand or at positions 2-9 of the 5' end of the antisense strand). For example, C1 is located at a position of the sense strand that is paired with nucleotides at positions 2 to 8 of the 5' end of the antisense strand. In one example, C1 is located at position 15 from the 5' end of the sense strand. C1 nucleotides have a thermally labile modification, which may comprise an abasic modification; mismatches with the opposite nucleotide in the duplex; and sugar modifications, such as 2 '-deoxy modifications or acyclic nucleotides, for example Unlocking Nucleic Acids (UNA) or Glycerol Nucleic Acids (GNA) or 2' -5 '-linked ribonucleotides ("3' -RNA"). In one embodiment, C1 has a thermally labile modification selected from the group consisting of: i) Mismatches with the opposite nucleotide in the antisense strand; ii) an abasic modification selected from the group consisting of:
And iii) a sugar modification selected from the group consisting of:
wherein B is a modified or unmodified nucleobase, R 1 And R is 2 Independently H, halogen, OR 3 Or alkyl; and R is 3 Is H, alkyl, cycloalkyl, arylAlkyl, heteroaryl or sugar. In one embodiment, the thermally labile modification in C1 is a mismatch selected from the group consisting of: 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' -deoxynucleobase. In one example, the heat labile modification in C1 is GNA or
T1, T1', T2', and T3 'each independently represent a spatial volume comprising a modification of a nucleotide that provides the nucleotide with a spatial volume that is less than or equal to the spatial volume of the 2' -OMe modification. The spatial volume refers to the sum of the spatial effects of the modification. Methods for determining the spatial effect of nucleotide modifications are known to those skilled in the art. The modification may be at the 2' position of the ribose sugar of the nucleotide, or a modification to a non-ribonucleotide, an acyclic nucleotide, or the backbone of a nucleotide that is similar or equivalent to the 2' position of the ribose sugar, and provides the nucleotide with a steric bulk that is less than or equal to the steric bulk of the 2' -OMe modification. For example, 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.
n 1 、n 3 And q 1 Independently 4 to 15 nucleotides in length.
n 5 、q 3 And q 7 Independently 1 to 6 nucleotides in length.
n 4 、q 2 And q 6 Independently 1 to 3 nucleotides in length; alternatively, n 4 Is 0.
q 5 Independently 0 to 10 nucleotides in length.
n 2 And q 4 Independently 0 to 3 nucleotides in length.
Alternatively, n 4 Is 0 to 3 nucleotides in length.
In one embodiment, n 4 May be 0. In one example, n 4 Is 0 and q 2 And q 6 Is 1. In another example, n 4 Is 0 and q 2 And q 6 Is 1, there are two phosphorothioate internucleotide linkage modifications in positions 1 to 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 in positions 18 to 23 of the antisense strand (counting from the 5' end of the antisense strand).
In one embodiment, n 4 、q 2 And q 6 Each 1.
In one embodiment, n 2 、n 4 、q 2 、q 4 And q 6 Each 1.
In one embodiment, the sense strand is 19 to 22 nucleotides in length, and n 4 In the case of 1, C1 is located at positions 14 to 17 of the 5' -end of the sense strand. In one embodiment, C1 is located at position 15 of the 5' end of the sense strand
In one embodiment, T3 'starts at position 2 of the 5' end of the antisense strand. In one example, T3 'is located at position 2 from the 5' end of the antisense strand, and q 6 Equal to 1.
In one embodiment, T1 'begins at position 14 of the 5' end of the antisense strand. In one example, T1 'is located at position 14 from the 5' end of the antisense strand, and q 2 Equal to 1.
In one exemplary embodiment, T3 'starts at position 2 in the 5' end of the antisense strand and T1 'starts at position 14 in the 5' end of the antisense strand. In one example, T3 'starts at position 2 in the 5' end of the antisense strand, and q 6 Equal to 1, and T1 'starts from position 14 in the 5' end of the antisense strand, and q 2 Equal to 1.
In one embodiment, T1 'and T3' are separated by 11 nucleotides in length (i.e., T1 'and T3' nucleotides are not counted).
In one embodiment, T1 'is located at position 14 from the 5' end of the antisense strand. In a real worldIn the example, T1 'is located at position 14 from the 5' end of the antisense strand, and q 2 Equal to 1, and the modification is at the 2 'position or at a position in the non-ribose, acyclic, or backbone that provides less steric bulk than the 2' -OMe ribose.
In one embodiment, T3 'is located at position 2 from the 5' end of the antisense strand. In one example, T3 'is located at position 2 from the 5' end of the antisense strand, and q 6 Equal to 1, and the modification is at the 2 'position or at a position in the non-ribose, acyclic, or backbone that provides less or equal spatial volume than the 2' -OMe ribose.
In one embodiment, T1 is located at the cleavage site of the sense strand. In one example, the sense strand is 19 to 22 nucleotides in length, and n 2 In the case of 1, T1 is located at position 11 from the 5' end of the sense strand. In one exemplary embodiment, the sense strand is 19 to 22 nucleotides in length, and n 2 When 1, T1 is located at the sense strand cleavage site at position 11 from the 5' end of the sense strand,
in one embodiment, T2 'starts at position 6 of the 5' end of the antisense strand. In one example, T2 'is located at positions 6 to 10 from the 5' end of the antisense strand, and q 4 1.
In one exemplary embodiment, the sense strand is 19 to 22 nucleotides in length, and n 2 For 1, T1 is located at the cleavage site of the sense strand, e.g., at position 11 from the 5' end of the sense strand; t1 'is located at position 14 from the 5' end of the antisense strand, and q 2 1 and the modification to T1' is located at the 2' position of the ribose sugar, or at a position in the non-ribose, acyclic, or backbone that provides less steric bulk than 2' -OMe ribose; t2 'is located at positions 6 to 10 from the 5' end of the antisense strand, and q 4 1 is shown in the specification; and T3 'is located at position 2 from the 5' end of the antisense strand, and q 6 Equal to 1, and the modification to T3' is at the 2' position or at a position in the non-ribose, acyclic, or backbone that provides less or equal spatial volume than the 2' -OMe ribose.
In one embodiment, T2 'starts at position 8 of the 5' end of the antisense strand. At the position ofIn one example, T2 'starts at position 8 of the 5' end of the antisense strand and q 4 2.
In one embodiment, T2 'starts at position 9 of the 5' end of the antisense strand. In one example, T2 'is located at position 9 from the 5' end of the antisense strand, and q 4 1.
In one embodiment, B1' is 2' -OMe or 2' -F, q 1 Is 9, T1 'is 2' -F, q 2 Is 1, B2' is 2' -OMe or 2' -F, q 3 Is 4, T2 'is 2' -F, q 4 Is 1, B3' is 2' -OMe or 2' -F, q 5 Is 6, T3 'is 2' -F, q 6 Is 1, B4 'is 2' -OMe, and q 7 Is 1; there are two phosphorothioate internucleotide linkage modifications within positions 1 to 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 to 23 of the antisense strand (counting from the 5' end of the antisense strand).
In one embodiment, n 4 Is 0, B3 is 2' -OMe, n 5 Is 3, B1' is 2' -OMe or 2' -F, q 1 Is 9, T1 'is 2' -F, q 2 Is 1, B2' is 2' -OMe or 2' -F, q 3 Is 4, T2 'is 2' -F, q 4 Is 1, B3' is 2' -OMe or 2' -F, q 5 Is 6, T3 'is 2' -F, q 6 Is 1, B4 'is 2' -OMe, and q 7 Is 1; there are two phosphorothioate internucleotide linkage modifications within positions 1 to 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 to 23 of the antisense strand (counting from the 5' end of the antisense strand).
In one embodiment, B1 is 2'-OMe or 2' -F, n 1 Is 8, T1 is 2' F, n 2 Is 3, B2 is 2' -OMe, n 3 Is 7, n 4 Is 0, B3 is 2' OMe, n 5 Is 3, B1' is 2' -OMe or 2' -F, q 1 Is 9, T1 'is 2' -F, q 2 Is 1, B2' is 2' -OMe or 2' -F, q 3 Is 4, T2 'is 2' -F, q 4 Is 2', B3' is 2'-OMe or 2' -F, q 5 Is 5, T3 'is 2' -F, q 6 Is 1, B4'Is 2' -OMe, and q 7 Is 1.
In one embodiment, B1 is 2'-OMe or 2' -F, n 1 Is 8, T1 is 2' F, n 2 Is 3, B2 is 2' -OMe, n 3 Is 7, n 4 Is 0, B3 is 2' -OMe, n 5 Is 3, B1' is 2' -OMe or 2' -F, q 1 Is 9, T1 'is 2' -F, q 2 Is 1, B2' is 2' -OMe or 2' -F, q 3 Is 4, T2 'is 2' -F, q 4 Is 2, B3' is 2' -OMe or 2' -F, q 5 Is 5, T3 'is 2' -F, q 6 Is 1, B4 'is 2' -OMe, and q 7 Is 1; there are two phosphorothioate internucleotide linkage modifications within positions 1 to 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 to 23 of the antisense strand (counting from the 5' end of the antisense strand).
In one embodiment, B1 is 2'-OMe or 2' -F, n 1 Is 6, T1 is 2' F, n 2 Is 3, B2 is 2' -OMe, n 3 Is 7, n 4 Is 0, B3 is 2' OMe, n 5 Is 3, B1' is 2' -OMe or 2' -F, q 1 Is 7, T1 'is 2' -F, q 2 Is 1, B2' is 2' -OMe or 2' -F, q 3 Is 4, T2 'is 2' -F, q 4 Is 2', B3' is 2'-OMe or 2' -F, q 5 Is 5, T3 'is 2' -F, q 6 Is 1, B4 'is 2' -OMe, and q 7 Is 1.
In one embodiment, B1 is 2'-OMe or 2' -F, n 1 Is 6, T1 is 2' F, n 2 Is 3, B2 is 2' -OMe, n 3 Is 7, n 4 Is 0, B3 is 2' -OMe, n 5 Is 3, B1' is 2' -OMe or 2' -F, q 1 Is 7, T1 'is 2' -F, q 2 Is 1, B2' is 2' -OMe or 2' -F, q 3 Is 4, T2 'is 2' -F, q 4 Is 2, B3' is 2' -OMe or 2' -F, q 5 Is 5, T3 'is 2' -F, q 6 Is 1, B4 'is 2' -OMe, and q 7 Is 1; having two phosphorothioate internucleotide linkage modifications in positions 1 to 5 of the sense strand (counting from the 5 'end of the sense strand) and two phosphorothioate internucleotide linkage modifications in positions 1 and 2, and in positions 18 to 23 of the antisense strand (counting from the 5' end of the antisense strand)Number) has two phosphorothioate internucleotide linkage modifications.
In one embodiment, B1 is 2'-OMe or 2' -F, n 1 Is 8, T1 is 2' F, n 2 Is 3, B2 is 2' -OMe, n 3 Is 7, n 4 Is 0, B3 is 2' OMe, n 5 Is 3, B1' is 2' -OMe or 2' -F, q 1 Is 9, T1 'is 2' -F, q 2 Is 1, B2' is 2' -OMe or 2' -F, q 3 Is 4, T2 'is 2' -F, q 4 Is 1, B3' is 2' -OMe or 2' -F, q 5 Is 6, T3 'is 2' -F, q 6 Is 1, B4 'is 2' -OMe, and q 7 Is 1.
In one embodiment, B1 is 2'-OMe or 2' -F, n 1 Is 8, T1 is 2' F, n 2 Is 3, B2 is 2' -OMe, n 3 Is 7, n 4 Is 0, B3 is 2' -OMe, n 5 Is 3, B1' is 2' -OMe or 2' -F, q 1 Is 9, T1 'is 2' -F, q 2 Is 1, B2' is 2' -OMe or 2' -F, q 3 Is 4, T2 'is 2' -F, q 4 Is 1, B3' is 2' -OMe or 2' -F, q 5 Is 6, T3 'is 2' -F, q 6 Is 1, B4 'is 2' -OMe, and q 7 Is 1; there are two phosphorothioate internucleotide linkage modifications within positions 1 to 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 to 23 of the antisense strand (counting from the 5' end of the antisense strand).
In one embodiment, B1 is 2'-OMe or 2' -F, n 1 Is 8, T1 is 2' F, n 2 Is 3, B2 is 2' -OMe, n 3 Is 7, n 4 Is 0, B3 is 2' OMe, n 5 Is 3, B1' is 2' -OMe or 2' -F, q 1 Is 9, T1 'is 2' -F, q 2 Is 1, B2' is 2' -OMe or 2' -F, q 3 Is 5, T2 'is 2' -F, q 4 Is 1, B3' is 2' -OMe or 2' -F, q 5 Is 5, T3 'is 2' -F, q 6 Is 1, B4 'is 2' -OMe, and q 7 Is 1; optionally having at least 2 additional TTs at the 3' end of the antisense strand.
In one embodiment, B1 is 2'-OMe or 2' -F, n 1 Is 8, T1 is 2' F, n 2 Is 3, B2 is 2' -OMe, n 3 Is 7, n 4 Is 0, B3 is 2' -OMe, n 5 Is 3, B1' is 2' -OMe or 2' -F, q 1 Is 9, T1 'is 2' -F, q 2 Is 1, B2' is 2' -OMe or 2' -F, q 3 Is 5, T2 'is 2' -F, q 4 Is 1, B3' is 2' -OMe or 2' -F, q 5 Is 5, T3 'is 2' -F, q 6 Is 1, B4 'is 2' -OMe, and q 7 Is 1; optionally having at least 2 additional TTs at the 3' end of the antisense strand; there are two phosphorothioate internucleotide linkage modifications within positions 1 to 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 to 23 of the antisense strand (counting from the 5' end of the antisense strand).
In one embodiment, B1 is 2'-OMe or 2' -F, n 1 Is 8, T1 is 2' F, n 2 Is 3, B2 is 2' -OMe, n 3 Is 7, n 4 Is 0, B3 is 2' OMe, n 5 Is 3, B1' is 2' -OMe or 2' -F, q 1 Is 9, T1 'is 2' -F, q 2 Is 1, B2' is 2' -OMe or 2' -F, q 3 Is 4, q 4 Is 0, B3' is 2' -OMe or 2' -F, q 5 Is 7, T3 'is 2' -F, q 6 Is 1, B4 'is 2' -OMe, and q 7 Is 1.
In one embodiment, B1 is 2'-OMe or 2' -F, n 1 Is 8, T1 is 2' F, n 2 Is 3, B2 is 2' -OMe, n 3 Is 7, n 4 Is 0, B3 is 2' -OMe, n 5 Is 3, B1' is 2' -OMe or 2' -F, q 1 Is 9, T1 'is 2' -F, q 2 Is 1, B2' is 2' -OMe or 2' -F, q 3 Is 4, q 4 Is 0, B3' is 2' -OMe or 2' -F, q 5 Is 7, T3 'is 2' -F, q 6 Is 1, B4 'is 2' -OMe, and q 7 Is 1; there are two phosphorothioate internucleotide linkage modifications in positions 1 to 5 (counting from the 5 'end) of the sense strand, and two phosphorothioate internucleotide linkage modifications in positions 1 and 2, and two phosphorothioate internucleotide linkage modifications in positions 18 to 23 (counting from the 5' end) of the antisense strand.
In one embodiment, B1 is 2'-OMe or 2' -F, n 1 Is 8, T1 is 2' F, n 2 Is 3, B2 is 2' -OMe, n 3 Is 7, n 4 Is 0, B3 is 2' OMe, n 5 Is 3, B1' is 2' -OMe or 2' -F, q 1 Is 9, T1 'is 2' -F, q 2 Is 1, B2' is 2' -OMe or 2' -F, q 3 Is 4, T2 'is 2' -F, q 4 Is 2', B3' is 2'-OMe or 2' -F, q 5 Is 5, T3 'is 2' -F, q 6 Is 1, B4 'is 2' -F, and q 7 Is 1.
In one embodiment, B1 is 2'-OMe or 2' -F, n 1 Is 8, T1 is 2' F, n 2 Is 3, B2 is 2' -OMe, n 3 Is 7, n 4 Is 0, B3 is 2' -OMe, n 5 Is 3, B1' is 2' -OMe or 2' -F, q 1 Is 9, T1 'is 2' -F, q 2 Is 1, B2' is 2' -OMe or 2' -F, q 3 Is 4, T2 'is 2' -F, q 4 Is 2, B3' is 2' -OMe or 2' -F, q 5 Is 5, T3 'is 2' -F, q 6 Is 1, B4 'is 2' -F, and q 7 Is 1; there are two phosphorothioate internucleotide linkage modifications within positions 1 to 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 to 23 of the antisense strand (counting from the 5' end of the antisense strand).
In one embodiment, B1 is 2'-OMe or 2' -F, n 1 Is 8, T1 is 2' F, n 2 Is 3, B2 is 2' -OMe, n 3 Is 7, n 4 Is 0, B3 is 2' OMe, n 5 Is 3, B1' is 2' -OMe or 2' -F, q 1 Is 9, T1 'is 2' -F, q 2 Is 1, B2' is 2' -OMe or 2' -F, q 3 Is 4, q 4 Is 0, B3' is 2' -OMe or 2' -F, q 5 Is 7, T3 'is 2' -F, q 6 Is 1, B4 'is 2' -F, and q 7 Is 1.
In one embodiment, B1 is 2'-OMe or 2' -F, n 1 Is 8, T1 is 2' F, n 2 Is 3, B2 is 2' -OMe, n 3 Is 7, n 4 Is 0, B3 is 2' -OMe, n 5 Is 3, B1' is 2' -OMe or 2' -F, q 1 Is 9, T1 'is 2' -F, q 2 Is 1, B2' is 2' -OMe or 2' -F, q 3 Is 4, q 4 Is 0, B3' is 2' -OMe or 2' -F, q 5 Is 7, T3 'is 2' -F, q 6 Is 1, B4 'is 2' -F, and q 7 Is 1; there are two phosphorothioate internucleotide linkage modifications within positions 1 to 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 to 23 of the antisense strand (counting from the 5' end of the antisense strand).
RNAi agents may include a phosphorus-containing group at the 5' end of the sense strand or antisense strand. The phosphorus-containing group at the 5' end can be 5' -phosphate (5 ' -P), 5' -phosphorothioate (5 ' -PS), 5' -phosphorothioate diester (5 ' -PS) 2 ) 5' -terminal vinyl phosphonate (5 ' -VP), 5' -terminal methylphosphonate (MePhos) or 5' -deoxy-5 ' -C-malonylWhen the 5' -terminal phosphorus-containing group is a 5' -terminal vinyl phosphonate (5 ' -VP), the 5' -VP may be the 5' -E-VP isomer (i.e., trans-vinyl phosphonate- >) The 5' -Z-VP isomer (i.e., cis-vinyl phosphate>) Or a mixture 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 5' -P. In one embodiment, the RNAi agent comprises 5' -P in the antisense strand.
In one embodiment, the RNAi agent comprises 5' -PS. In one embodiment, the RNAi agent comprises 5' -PS in the antisense strand.
In one embodiment, the RNAi agent comprises 5' -VP. In one embodiment, the RNAi agent comprises a 5' -VP in the antisense strand. In one embodiment, the RNAi agent comprises 5' -E-VP in the antisense strand. In one embodiment, the RNAi agent comprises 5' -Z-VP in the antisense strand.
In one embodiment, the RNAi agent comprises 5' -PS 2 . In one embodiment, the RNAi agent comprises 5' -PS in the antisense strand 2
In one embodiment, the RNAi agent comprises 5' -PS 2 . In one embodiment, the RNAi agent comprises 5 '-deoxy-5' -C-malonyl in the antisense strand.
In one embodiment, B1 is 2'-OMe or 2' -F, n 1 Is 8, T1 is 2' F, n 2 Is 3, B2 is 2' -OMe, n 3 Is 7, n 4 Is 0, B3 is 2' OMe, n 5 Is 3, B1' is 2' -OMe or 2' -F, q 1 Is 9, T1 'is 2' -F, q 2 Is 1, B2' is 2' -OMe or 2' -F, q 3 Is 4, T2 'is 2' -F, q 4 Is 2', B3' is 2'-OMe or 2' -F, q 5 Is 5, T3 'is 2' -F, q 6 Is 1, B4 'is 2' -OMe, and q 7 Is 1.RNAi agents also include 5' -PS.
In one embodiment, B1 is 2'-OMe or 2' -F, n 1 Is 8, T1 is 2' F, n 2 Is 3, B2 is 2' -OMe, n 3 Is 7, n 4 Is 0, B3 is 2' OMe, n 5 Is 3, B1' is 2' -OMe or 2' -F, q 1 Is 9, T1 'is 2' -F, q 2 Is 1, B2' is 2' -OMe or 2' -F, q 3 Is 4, T2 'is 2' -F, q 4 Is 2', B3' is 2'-OMe or 2' -F, q 5 Is 5, T3 'is 2' -F, q 6 Is 1, B4 'is 2' -OMe, and q 7 Is 1.RNAi agents also include 5' -P.
In one embodiment, B1 is 2'-OMe or 2' -F, n 1 Is 8, T1 is 2' F, n 2 Is 3, B2 is 2' -OMe, n 3 Is 7, n 4 Is 0, B3 is 2' OMe, n 5 Is 3, B1' is 2' -OMe or 2' -F, q 1 Is 9, T1 'is 2' -F, q 2 Is 1, B2' is 2' -OMe or 2' -F, q 3 Is 4, T2 'is 2' -F, q 4 Is 2', B3' is 2'-OMe or 2' -F, q 5 Is 5, T3 'is 2' -F, q 6 Is 1, B4 'is 2' -OMe, and q 7 Is 1.RNAi agents also include 5' -VP. The 5' -VP may be 5' -E-VP, 5' -Z-VP, or a combination thereof.
In one embodiment, B1 is 2'-OMe or 2' -F, n 1 Is 8, T1 is 2' F, n 2 Is 3, B2 is 2' -OMe,n 3 is 7, n 4 Is 0, B3 is 2' OMe, n 5 Is 3, B1' is 2' -OMe or 2' -F, q 1 Is 9, T1 'is 2' -F, q 2 Is 1, B2' is 2' -OMe or 2' -F, q 3 Is 4, T2 'is 2' -F, q 4 Is 2', B3' is 2'-OMe or 2' -F, q 5 Is 5, T3 'is 2' -F, q 6 Is 1, B4 'is 2' -OMe, and q 7 Is 1.RNAi agents also include 5' -PS 2
In one embodiment, B1 is 2'-OMe or 2' -F, n 1 Is 8, T1 is 2' F, n 2 Is 3, B2 is 2' -OMe, n 3 Is 7, n 4 Is 0, B3 is 2' OMe, n 5 Is 3, B1' is 2' -OMe or 2' -F, q 1 Is 9, T1 'is 2' -F, q 2 Is 1, B2' is 2' -OMe or 2' -F, q 3 Is 4, T2 'is 2' -F, q 4 Is 2', B3' is 2'-OMe or 2' -F, q 5 Is 5, T3 'is 2' -F, q 6 Is 1, B4 'is 2' -OMe, and q 7 Is 1.RNAi agents also include 5 '-deoxy-5' -C-malonyl.
In one embodiment, B1 is 2'-OMe or 2' -F, n 1 Is 8, T1 is 2' F, n 2 Is 3, B2 is 2' -OMe, n 3 Is 7, n 4 Is 0, B3 is 2' -OMe, n 5 Is 3, B1' is 2' -OMe or 2' -F, q 1 Is 9, T1 'is 2' -F, q 2 Is 1, B2' is 2' -OMe or 2' -F, q 3 Is 4, T2 'is 2' -F, q 4 Is 2, B3' is 2' -OMe or 2' -F, q 5 Is 5, T3 'is 2' -F, q 6 Is 1, B4 'is 2' -OMe, and q 7 Is 1; there are two phosphorothioate internucleotide linkage modifications within positions 1 to 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 to 23 of the antisense strand (counting from the 5' end of the antisense strand). RNAi agents also include 5' -P.
In one embodiment, B1 is 2'-OMe or 2' -F, n 1 Is 8, T1 is 2' F, n 2 Is 3, B2 is 2' -OMe, n 3 Is 7, n 4 Is 0, B3 is 2' -OMe, n 5 Is 3, B1' is 2' -OMe or 2' -F, q 1 Is 9, T1 'is 2' -F, q 2 Is 1, B2' is 2' -OMe or 2' -F, q 3 Is 4, T2 'is 2' -F, q 4 Is 2, B3' is 2' -OMe or 2' -F, q 5 Is 5, T3 'is 2' -F, q 6 Is 1, B4 'is 2' -OMe, and q 7 Is 1; there are two phosphorothioate internucleotide linkage modifications within positions 1 to 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 to 23 of the antisense strand (counting from the 5' end of the antisense strand). RNAi agents also include 5' -PS.
In one embodiment, B1 is 2'-OMe or 2' -F, n 1 Is 8, T1 is 2' F, n 2 Is 3, B2 is 2' -OMe, n 3 Is 7, n 4 Is 0, B3 is 2' -OMe, n 5 Is 3, B1' is 2' -OMe or 2' -F, q 1 Is 9, T1 'is 2' -F, q 2 Is 1, B2' is 2' -OMe or 2' -F, q 3 Is 4, T2 'is 2' -F, q 4 Is 2, B3' is 2' -OMe or 2' -F, q 5 Is 5, T3 'is 2' -F, q 6 Is 1, B4 'is 2' -OMe, and q 7 Is 1; there are two phosphorothioate internucleotide linkage modifications within positions 1 to 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 to 23 of the antisense strand (counting from the 5' end of the antisense strand). RNAi agents also include 5' -VP. The 5' -VP may be 5' -E-VP, 5' -Z-VP, or a combination thereof.
In one embodiment, B1 is 2'-OMe or 2' -F, n 1 Is 8, T1 is 2' F, n 2 Is 3, B2 is 2' -OMe, n 3 Is 7, n 4 Is 0, B3 is 2' -OMe, n 5 Is 3, B1' is 2' -OMe or 2' -F, q 1 Is 9, T1 'is 2' -F, q 2 Is 1, B2' is 2' -OMe or 2' -F, q 3 Is 4, T2 'is 2' -F, q 4 Is 2, B3' is 2' -OMe or 2' -F, q 5 Is 5, T3 'is 2' -F, q 6 Is 1, B4 'is 2' -OMe, and q 7 Is 1; having two phosphorothioate internucleotide linkage modifications in positions 1 to 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 in the antisense strandTwo phosphorothioate internucleotide linkage modifications are located in positions 18 to 23 (counting from the 5' end of the antisense strand). RNAi agents also include 5' -PS 2
In one embodiment, B1 is 2'-OMe or 2' -F, n 1 Is 8, T1 is 2' F, n 2 Is 3, B2 is 2' -OMe, n 3 Is 7, n 4 Is 0, B3 is 2' -OMe, n 5 Is 3, B1' is 2' -OMe or 2' -F, q 1 Is 9, T1 'is 2' -F, q 2 Is 1, B2' is 2' -OMe or 2' -F, q 3 Is 4, T2 'is 2' -F, q 4 Is 2, B3' is 2' -OMe or 2' -F, q 5 Is 5, T3 'is 2' -F, q 6 Is 1, B4 'is 2' -OMe, and q 7 Is 1; there are two phosphorothioate internucleotide linkage modifications within positions 1 to 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 to 23 of the antisense strand (counting from the 5' end of the antisense strand). RNAi agents also include 5 '-deoxy-5' -C-malonyl.
In one embodiment, B1 is 2'-OMe or 2' -F, n 1 Is 8, T1 is 2' F, n 2 Is 3, B2 is 2' -OMe, n 3 Is 7, n 4 Is 0, B3 is 2' OMe, n 5 Is 3, B1' is 2' -OMe or 2' -F, q 1 Is 9, T1 'is 2' -F, q 2 Is 1, B2' is 2' -OMe or 2' -F, q 3 Is 4, q 4 Is 0, B3' is 2' -OMe or 2' -F, q 5 Is 7, T3 'is 2' -F, q 6 Is 1, B4 'is 2' -OMe, and q 7 Is 1.RNAi agents also include 5' -P.
In one embodiment, B1 is 2'-OMe or 2' -F, n 1 Is 8, T1 is 2' F, n 2 Is 3, B2 is 2' -OMe, n 3 Is 7, n 4 Is 0, B3 is 2' OMe, n 5 Is 3, B1' is 2' -OMe or 2' -F, q 1 Is 9, T1 'is 2' -F, q 2 Is 1, B2' is 2' -OMe or 2' -F, q 3 Is 4, q 4 Is 0, B3' is 2' -OMe or 2' -F, q 5 Is 7, T3 'is 2' -F, q 6 Is 1, B4 'is 2' -OMe, and q 7 Is 1.dsRNA agents also include 5' -PS.
In one embodiment, B1 is 2' -OMe or2'-F,n 1 Is 8, T1 is 2' F, n 2 Is 3, B2 is 2' -OMe, n 3 Is 7, n 4 Is 0, B3 is 2' OMe, n 5 Is 3, B1' is 2' -OMe or 2' -F, q 1 Is 9, T1 'is 2' -F, q 2 Is 1, B2' is 2' -OMe or 2' -F, q 3 Is 4, q 4 Is 0, B3' is 2' -OMe or 2' -F, q 5 Is 7, T3 'is 2' -F, q 6 Is 1, B4 'is 2' -OMe, and q 7 Is 1.RNAi agents also include 5' -VP. The 5' -VP may be 5' -E-VP, 5' -Z-VP, or a combination thereof.
In one embodiment, B1 is 2'-OMe or 2' -F, n 1 Is 8, T1 is 2' F, n 2 Is 3, B2 is 2' -OMe, n 3 Is 7, n 4 Is 0, B3 is 2' OMe, n 5 Is 3, B1' is 2' -OMe or 2' -F, q 1 Is 9, T1 'is 2' -F, q 2 Is 1, B2' is 2' -OMe or 2' -F, q 3 Is 4, q 4 Is 0, B3' is 2' -OMe or 2' -F, q 5 Is 7, T3 'is 2' -F, q 6 Is 1, B4 'is 2' -OMe, and q 7 Is 1.RNAi agents also include 5' -PS 2
In one embodiment, B1 is 2'-OMe or 2' -F, n 1 Is 8, T1 is 2' F, n 2 Is 3, B2 is 2' -OMe, n 3 Is 7, n 4 Is 0, B3 is 2' OMe, n 5 Is 3, B1' is 2' -OMe or 2' -F, q 1 Is 9, T1 'is 2' -F, q 2 Is 1, B2' is 2' -OMe or 2' -F, q 3 Is 4, q 4 Is 0, B3' is 2' -OMe or 2' -F, q 5 Is 7, T3 'is 2' -F, q 6 Is 1, B4 'is 2' -OMe, and q 7 Is 1.RNAi agents also include 5 '-deoxy-5' -C-malonyl.
In one embodiment, B1 is 2'-OMe or 2' -F, n 1 Is 8, T1 is 2' F, n 2 Is 3, B2 is 2' -OMe, n 3 Is 7, n 4 Is 0, B3 is 2' -OMe, n 5 Is 3, B1' is 2' -OMe or 2' -F, q 1 Is 9, T1 'is 2' -F, q 2 Is 1, B2' is 2' -OMe or 2' -F, q 3 Is 4, q 4 Is 0, B3' is 2' -OMe or 2' -F, q 5 Is 7, T3 'is 2' -F, q 6 Is 1, B4 'is 2' -OMe, and q 7 Is 1; having two phosphorothioates in positions 1 to 5 (counting from the 5' end) of the sense strand Ester internucleotide linkage modifications and two phosphorothioate internucleotide linkage modifications at positions 1 and 2 and two phosphorothioate internucleotide linkage modifications within positions 18 to 23 (counting from the 5' end) of the antisense strand. RNAi agents also include 5' -P.
In one embodiment, B1 is 2'-OMe or 2' -F, n 1 Is 8, T1 is 2' F, n 2 Is 3, B2 is 2' -OMe, n 3 Is 7, n 4 Is 0, B3 is 2' -OMe, n 5 Is 3, B1' is 2' -OMe or 2' -F, q 1 Is 9, T1 'is 2' -F, q 2 Is 1, B2' is 2' -OMe or 2' -F, q 3 Is 4, q 4 Is 0, B3' is 2' -OMe or 2' -F, q 5 Is 7, T3 'is 2' -F, q 6 Is 1, B4 'is 2' -OMe, and q 7 Is 1; there are two phosphorothioate internucleotide linkage modifications in positions 1 to 5 (counting from the 5 'end) of the sense strand, and two phosphorothioate internucleotide linkage modifications in positions 1 and 2, and two phosphorothioate internucleotide linkage modifications in positions 18 to 23 (counting from the 5' end) of the antisense strand. RNAi agents also include 5' -PS.
In one embodiment, B1 is 2'-OMe or 2' -F, n 1 Is 8, T1 is 2' F, n 2 Is 3, B2 is 2' -OMe, n 3 Is 7, n 4 Is 0, B3 is 2' -OMe, n 5 Is 3, B1' is 2' -OMe or 2' -F, q 1 Is 9, T1 'is 2' -F, q 2 Is 1, B2' is 2' -OMe or 2' -F, q 3 Is 4, q 4 Is 0, B3' is 2' -OMe or 2' -F, q 5 Is 7, T3 'is 2' -F, q 6 Is 1, B4 'is 2' -OMe, and q 7 Is 1; there are two phosphorothioate internucleotide linkage modifications in positions 1 to 5 (counting from the 5 'end) of the sense strand, and two phosphorothioate internucleotide linkage modifications in positions 1 and 2, and two phosphorothioate internucleotide linkage modifications in positions 18 to 23 (counting from the 5' end) of the antisense strand. RNAi agents also include 5' -VP. The 5' -VP may be 5' -E-VP, 5' -Z-VP, or a combination thereof.
In one embodiment, B1 is 2'-OMe or 2' -F, n 1 Is 8, T1 is 2' F, n 2 Is 3, B2 is 2' -OMe, n 3 Is 7, n 4 Is 0, B3 is 2' -OMe, n 5 Is 3, B1' is 2' -OMe or 2' -F, q 1 Is 9, T1 'is 2' -F, q 2 Is 1, B2' is 2' -OMe or 2' -F, q 3 Is 4, q 4 Is 0, B3' is 2' -OMe or 2' -F, q 5 Is 7, T3 'is 2' -F, q 6 Is 1, B4 'is 2' -OMe, and q 7 Is 1; there are two phosphorothioate internucleotide linkage modifications in positions 1 to 5 (counting from the 5 'end) of the sense strand, and two phosphorothioate internucleotide linkage modifications in positions 1 and 2, and two phosphorothioate internucleotide linkage modifications in positions 18 to 23 (counting from the 5' end) of the antisense strand. RNAi agents also include 5' -PS 2
In one embodiment, B1 is 2'-OMe or 2' -F, n 1 Is 8, T1 is 2' F, n 2 Is 3, B2 is 2' -OMe, n 3 Is 7, n 4 Is 0, B3 is 2' -OMe, n 5 Is 3, B1' is 2' -OMe or 2' -F, q 1 Is 9, T1 'is 2' -F, q 2 Is 1, B2' is 2' -OMe or 2' -F, q 3 Is 4, q 4 Is 0, B3' is 2' -OMe or 2' -F, q 5 Is 7, T3 'is 2' -F, q 6 Is 1, B4 'is 2' -OMe, and q 7 Is 1; there are two phosphorothioate internucleotide linkage modifications in positions 1 to 5 (counting from the 5 'end) of the sense strand, and two phosphorothioate internucleotide linkage modifications in positions 1 and 2, and two phosphorothioate internucleotide linkage modifications in positions 18 to 23 (counting from the 5' end) of the antisense strand. RNAi agents also include 5 '-deoxy-5' -C-malonyl.
In one embodiment, B1 is 2'-OMe or 2' -F, n 1 Is 8, T1 is 2' F, n 2 Is 3, B2 is 2' -OMe, n 3 Is 7, n 4 Is 0, B3 is 2' OMe, n 5 Is 3, B1' is 2' -OMe or 2' -F, q 1 Is 9, T1 'is 2' -F, q 2 Is 1, B2' is 2' -OMe or 2' -F, q 3 Is 4, T2 'is 2' -F, q 4 Is 2', B3' is 2'-OMe or 2' -F, q 5 Is 5, T3 'is 2' -F, q 6 Is 1, B4 'is 2' -F, and q 7 Is 1.RNAi agents also include 5' -P.
In one embodiment, B1 is 2'-OMe or 2' -F, n 1 Is 8, T1 is 2' F, n 2 Is 3, B2 is 2' -OMe, n 3 Is 7, n 4 Is 0, B3 is 2' OMe, n 5 Is 3, B1' is 2' -OMe or 2' -F, q 1 Is 9, T1 'is 2' -F, q 2 Is 1, B2' is 2' -OMe or 2' -F, q 3 Is 4, T2 'is 2' -F, q 4 Is 2', B3' is 2'-OMe or 2' -F, q 5 Is 5, T3 'is 2' -F, q 6 Is 1, B4 'is 2' -F, and q 7 Is 1.RNAi agents also include 5' -PS.
In one embodiment, B1 is 2'-OMe or 2' -F, n 1 Is 8, T1 is 2' F, n 2 Is 3, B2 is 2' -OMe, n 3 Is 7, n 4 Is 0, B3 is 2' OMe, n 5 Is 3, B1' is 2' -OMe or 2' -F, q 1 Is 9, T1 'is 2' -F, q 2 Is 1, B2' is 2' -OMe or 2' -F, q 3 Is 4, T2 'is 2' -F, q 4 Is 2', B3' is 2'-OMe or 2' -F, q 5 Is 5, T3 'is 2' -F, q 6 Is 1, B4 'is 2' -F, and q 7 Is 1.RNAi agents also include 5' -VP. The 5' -VP may be 5' -E-VP, 5' -Z-VP, or a combination thereof.
In one embodiment, B1 is 2'-OMe or 2' -F, n 1 Is 8, T1 is 2' F, n 2 Is 3, B2 is 2' -OMe, n 3 Is 7, n 4 Is 0, B3 is 2' OMe, n 5 Is 3, B1' is 2' -OMe or 2' -F, q 1 Is 9, T1 'is 2' -F, q 2 Is 1, B2' is 2' -OMe or 2' -F, q 3 Is 4, T2 'is 2' -F, q 4 Is 2', B3' is 2'-OMe or 2' -F, q 5 Is 5, T3 'is 2' -F, q 6 Is 1, B4 'is 2' -F, and q 7 Is 1.dsRNAi RNA agents also include 5' -PS 2
In one embodiment, B1 is 2'-OMe or 2' -F, n 1 Is 8, T1 is 2' F, n 2 Is 3, B2 is 2' -OMe, n 3 Is 7, n 4 Is 0, B3 is 2' OMe, n 5 Is 3, B1' is 2' -OMe or 2' -F, q 1 Is 9, T1 'is 2' -F, q 2 Is 1, B2' is 2' -OMe or 2' -F, q 3 Is 4, T2 'is 2' -F, q 4 Is 2', B3' is 2'-OMe or 2' -F, q 5 Is 5, T3 'is 2' -F, q 6 Is 1, B4 'is 2' -F, and q 7 Is 1.RNAi agents also include 5 '-deoxy-5' -C-malonyl.
In one embodiment, B1 is 2'-OMe or 2'-F,n 1 Is 8, T1 is 2' F, n 2 Is 3, B2 is 2' -OMe, n 3 Is 7, n 4 Is 0, B3 is 2' -OMe, n 5 Is 3, B1' is 2' -OMe or 2' -F, q 1 Is 9, T1 'is 2' -F, q 2 Is 1, B2' is 2' -OMe or 2' -F, q 3 Is 4, T2 'is 2' -F, q 4 Is 2, B3' is 2' -OMe or 2' -F, q 5 Is 5, T3 'is 2' -F, q 6 Is 1, B4 'is 2' -F, and q 7 Is 1; there are two phosphorothioate internucleotide linkage modifications within positions 1 to 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 to 23 of the antisense strand (counting from the 5' end of the antisense strand). RNAi agents also include 5' -P.
In one embodiment, B1 is 2'-OMe or 2' -F, n 1 Is 8, T1 is 2' F, n 2 Is 3, B2 is 2' -OMe, n 3 Is 7, n 4 Is 0, B3 is 2' -OMe, n 5 Is 3, B1' is 2' -OMe or 2' -F, q 1 Is 9, T1 'is 2' -F, q 2 Is 1, B2' is 2' -OMe or 2' -F, q 3 Is 4, T2 'is 2' -F, q 4 Is 2, B3' is 2' -OMe or 2' -F, q 5 Is 5, T3 'is 2' -F, q 6 Is 1, B4 'is 2' -F, and q 7 Is 1; there are two phosphorothioate internucleotide linkage modifications within positions 1 to 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 to 23 of the antisense strand (counting from the 5' end of the antisense strand). RNAi agents also include 5' -PS.
In one embodiment, B1 is 2'-OMe or 2' -F, n 1 Is 8, T1 is 2' F, n 2 Is 3, B2 is 2' -OMe, n 3 Is 7, n 4 Is 0, B3 is 2' -OMe, n 5 Is 3, B1' is 2' -OMe or 2' -F, q 1 Is 9, T1 'is 2' -F, q 2 Is 1, B2' is 2' -OMe or 2' -F, q 3 Is 4, T2 'is 2' -F, q 4 Is 2, B3' is 2' -OMe or 2' -F, q 5 Is 5, T3 'is 2' -F, q 6 Is 1, B4 'is 2' -F, and q 7 Is 1; in the sense strandHas two phosphorothioate internucleotide linkage modifications in positions 1 to 5 (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 in positions 18 to 23 of the antisense strand (counting from the 5' end of the antisense strand). RNAi agents also include 5' -VP. The 5' -VP may be 5' -E-VP, 5' -Z-VP, or a combination thereof.
In one embodiment, B1 is 2'-OMe or 2' -F, n 1 Is 8, T1 is 2' F, n 2 Is 3, B2 is 2' -OMe, n 3 Is 7, n 4 Is 0, B3 is 2' -OMe, n 5 Is 3, B1' is 2' -OMe or 2' -F, q 1 Is 9, T1 'is 2' -F, q 2 Is 1, B2' is 2' -OMe or 2' -F, q 3 Is 4, T2 'is 2' -F, q 4 Is 2, B3' is 2' -OMe or 2' -F, q 5 Is 5, T3 'is 2' -F, q 6 Is 1, B4 'is 2' -F, and q 7 Is 1; there are two phosphorothioate internucleotide linkage modifications within positions 1 to 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 to 23 of the antisense strand (counting from the 5' end of the antisense strand). RNAi agents also include 5' -PS 2
In one embodiment, B1 is 2'-OMe or 2' -F, n 1 Is 8, T1 is 2' F, n 2 Is 3, B2 is 2' -OMe, n 3 Is 7, n 4 Is 0, B3 is 2' -OMe, n 5 Is 3, B1' is 2' -OMe or 2' -F, q 1 Is 9, T1 'is 2' -F, q 2 Is 1, B2' is 2' -OMe or 2' -F, q 3 Is 4, T2 'is 2' -F, q 4 Is 2, B3' is 2' -OMe or 2' -F, q 5 Is 5, T3 'is 2' -F, q 6 Is 1, B4 'is 2' -F, and q 7 Is 1; there are two phosphorothioate internucleotide linkage modifications within positions 1 to 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 to 23 of the antisense strand (counting from the 5' end of the antisense strand). RNAi agents also include 5 '-deoxy-5' -C-malonyl.
In one embodiment, B1 is 2'-OMe or 2' -F, n 1 Is 8, T1 is 2' F, n 2 Is 3, B2 is 2' -OMe, n 3 Is 7, n 4 Is 0, B3 is 2' OMe, n 5 Is 3, B1' is 2' -OMe or 2' -F, q 1 Is 9, T1 'is 2' -F, q 2 Is 1, B2' is 2' -OMe or 2' -F, q 3 Is 4, q 4 Is 0, B3' is 2' -OMe or 2' -F, q 5 Is 7, T3 'is 2' -F, q 6 Is 1, B4 'is 2' -F, and q 7 Is 1.RNAi agents also include 5' -P.
In one embodiment, B1 is 2'-OMe or 2' -F, n 1 Is 8, T1 is 2' F, n 2 Is 3, B2 is 2' -OMe, n 3 Is 7, n 4 Is 0, B3 is 2' OMe, n 5 Is 3, B1' is 2' -OMe or 2' -F, q 1 Is 9, T1 'is 2' -F, q 2 Is 1, B2' is 2' -OMe or 2' -F, q 3 Is 4, q 4 Is 0, B3' is 2' -OMe or 2' -F, q 5 Is 7, T3 'is 2' -F, q 6 Is 1, B4 'is 2' -F, and q 7 Is 1.RNAi agents also include 5' -PS.
In one embodiment, B1 is 2'-OMe or 2' -F, n 1 Is 8, T1 is 2' F, n 2 Is 3, B2 is 2' -OMe, n 3 Is 7, n 4 Is 0, B3 is 2' OMe, n 5 Is 3, B1' is 2' -OMe or 2' -F, q 1 Is 9, T1 'is 2' -F, q 2 Is 1, B2' is 2' -OMe or 2' -F, q 3 Is 4, q 4 Is 0, B3' is 2' -OMe or 2' -F, q 5 Is 7, T3 'is 2' -F, q 6 Is 1, B4 'is 2' -F, and q 7 Is 1.RNAi agents also include 5' -VP. The 5' -VP may be 5' -E-VP, 5' -Z-VP, or a combination thereof.
In one embodiment, B1 is 2'-OMe or 2' -F, n 1 Is 8, T1 is 2' F, n 2 Is 3, B2 is 2' -OMe, n 3 Is 7, n 4 Is 0, B3 is 2' OMe, n 5 Is 3, B1' is 2' -OMe or 2' -F, q 1 Is 9, T1 'is 2' -F, q 2 Is 1, B2' is 2' -OMe or 2' -F, q 3 Is 4, q 4 Is 0, B3' is 2' -OMe or 2' -F, q 5 Is 7, T3 'is 2' -F, q 6 Is 1, B4 'is 2' -F, and q 7 Is 1.RNAi agents also include 5' -PS 2
In one embodiment, B1 is 2'-OMe or 2' -F, n 1 Is 8, T1 is 2' F, n 2 Is 3, B2 is 2' -OMe, n 3 Is 7, n 4 Is 0, B3 is 2' OMe, n 5 Is 3, B1' is 2' -OMe or 2' -F, q 1 Is 9, T1 'is 2' -F, q 2 Is 1, B2' is 2' -OMe or 2' -F, q 3 Is 4, q 4 Is 0, B3' is 2' -OMe or 2' -F, q 5 Is 7, T3 'is 2' -F, q 6 Is 1, B4 'is 2' -F, and q 7 Is 1.RNAi agents also include 5 '-deoxy-5' -C-malonyl.
In one embodiment, B1 is 2'-OMe or 2' -F, n 1 Is 8, T1 is 2' F, n 2 Is 3, B2 is 2' -OMe, n 3 Is 7, n 4 Is 0, B3 is 2' -OMe, n 5 Is 3, B1' is 2' -OMe or 2' -F, q 1 Is 9, T1 'is 2' -F, q 2 Is 1, B2' is 2' -OMe or 2' -F, q 3 Is 4, q 4 Is 0, B3' is 2' -OMe or 2' -F, q 5 Is 7, T3 'is 2' -F, q 6 Is 1, B4 'is 2' -F, and q 7 Is 1; there are two phosphorothioate internucleotide linkage modifications within positions 1 to 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 to 23 of the antisense strand (counting from the 5' end of the antisense strand). RNAi agents also include 5' -P.
In one embodiment, B1 is 2'-OMe or 2' -F, n 1 Is 8, T1 is 2' F, n 2 Is 3, B2 is 2' -OMe, n 3 Is 7, n 4 Is 0, B3 is 2' -OMe, n 5 Is 3, B1' is 2' -OMe or 2' -F, q 1 Is 9, T1 'is 2' -F, q 2 Is 1, B2' is 2' -OMe or 2' -F, q 3 Is 4, q 4 Is 0, B3' is 2' -OMe or 2' -F, q 5 Is 7, T3 'is 2' -F, q 6 Is 1, B4 'is 2' -F, and q 7 Is 1; having two phosphorothioate internucleotide linkage modifications in positions 1 to 5 of the sense strand (counting from the 5 'end of the sense strand) and two phosphorothioate internucleotide linkage modifications in positions 1 and 2, and two phosphorothioate nucleosides in positions 18 to 23 of the antisense strand (counting from the 5' end of the antisense strand)Modification of the acid bond. RNAi agents also include 5' -PS.
In one embodiment, B1 is 2'-OMe or 2' -F, n 1 Is 8, T1 is 2' F, n 2 Is 3, B2 is 2' -OMe, n 3 Is 7, n 4 Is 0, B3 is 2' -OMe, n 5 Is 3, B1' is 2' -OMe or 2' -F, q 1 Is 9, T1 'is 2' -F, q 2 Is 1, B2' is 2' -OMe or 2' -F, q 3 Is 4, q 4 Is 0, B3' is 2' -OMe or 2' -F, q 5 Is 7, T3 'is 2' -F, q 6 Is 1, B4 'is 2' -F, and q 7 Is 1; there are two phosphorothioate internucleotide linkage modifications within positions 1 to 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 to 23 of the antisense strand (counting from the 5' end of the antisense strand). RNAi agents also include 5' -VP. The 5' -VP may be 5' -E-VP, 5' -Z-VP, or a combination thereof.
In one embodiment, B1 is 2'-OMe or 2' -F, n 1 Is 8, T1 is 2' F, n 2 Is 3, B2 is 2' -OMe, n 3 Is 7, n 4 Is 0, B3 is 2' -OMe, n 5 Is 3, B1' is 2' -OMe or 2' -F, q 1 Is 9, T1 'is 2' -F, q 2 Is 1, B2' is 2' -OMe or 2' -F, q 3 Is 4, q 4 Is 0, B3' is 2' -OMe or 2' -F, q 5 Is 7, T3 'is 2' -F, q 6 Is 1, B4 'is 2' -F, and q 7 Is 1; there are two phosphorothioate internucleotide linkage modifications within positions 1 to 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 to 23 of the antisense strand (counting from the 5' end of the antisense strand). RNAi agents also include 5' -PS 2
In one embodiment, B1 is 2'-OMe or 2' -F, n 1 Is 8, T1 is 2' F, n 2 Is 3, B2 is 2' -OMe, n 3 Is 7, n 4 Is 0, B3 is 2' -OMe, n 5 Is 3, B1' is 2' -OMe or 2' -F, q 1 Is 9, T1 'is 2' -F, q 2 Is 1, B2' is 2' -OMe or 2' -F, q 3 Is 4, q 4 Is 0, B3' is 2' -OMe or 2' -F, q 5 Is 7, T3 'is 2' -F, q 6 Is 1, B4 'is 2' -F, and q 7 Is 1; there are two phosphorothioate internucleotide linkage modifications within positions 1 to 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 to 23 of the antisense strand (counting from the 5' end of the antisense strand). RNAi agents also include 5 '-deoxy-5' -C-malonyl.
In one embodiment, B1 is 2'-OMe or 2' -F, n 1 Is 8, T1 is 2' F, n 2 Is 3, B2 is 2' -OMe, n 3 Is 7, n 4 Is 0, B3 is 2' -OMe, n 5 Is 3, B1' is 2' -OMe or 2' -F, q 1 Is 9, T1 'is 2' -F, q 2 Is 1, B2' is 2' -OMe or 2' -F, q 3 Is 4, T2 'is 2' -F, q 4 Is 2, B3' is 2' -OMe or 2' -F, q 5 Is 5, T3 'is 2' -F, q 6 Is 1, B4 'is 2' -OMe, and q 7 Is 1; there are two phosphorothioate internucleotide linkage modifications within positions 1 to 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 to 23 of the antisense strand (counting from the 5' end of the antisense strand). RNAi agents also include 5' -P and targeting ligands. In one embodiment, the 5' -P is located at the 5' end of the antisense strand and the targeting ligand is located at the 3' end of the sense strand.
In one embodiment, B1 is 2'-OMe or 2' -F, n 1 Is 8, T1 is 2' F, n 2 Is 3, B2 is 2' -OMe, n 3 Is 7, n 4 Is 0, B3 is 2' -OMe, n 5 Is 3, B1' is 2' -OMe or 2' -F, q 1 Is 9, T1 'is 2' -F, q 2 Is 1, B2' is 2' -OMe or 2' -F, q 3 Is 4, T2 'is 2' -F, q 4 Is 2, B3' is 2' -OMe or 2' -F, q 5 Is 5, T3 'is 2' -F, q 6 Is 1, B4 'is 2' -OMe, and q 7 Is 1; having two phosphorothioate internucleotide linkage modifications in positions 1 to 5 of the sense strand (counting from the 5' end of the sense strand) and two at positions 1 and 2Phosphorothioate internucleotide linkage modifications and there are two phosphorothioate internucleotide linkage modifications within positions 18 to 23 of the antisense strand (counting from the 5' end of the antisense strand). RNAi agents also include 5' -PS and targeting ligands. In one embodiment, the 5' -PS is located at the 5' end of the antisense strand and the targeting ligand is located at the 3' end of the sense strand.
In one embodiment, B1 is 2'-OMe or 2' -F, n 1 Is 8, T1 is 2' F, n 2 Is 3, B2 is 2' -OMe, n 3 Is 7, n 4 Is 0, B3 is 2' -OMe, n 5 Is 3, B1' is 2' -OMe or 2' -F, q 1 Is 9, T1 'is 2' -F, q 2 Is 1, B2' is 2' -OMe or 2' -F, q 3 Is 4, T2 'is 2' -F, q 4 Is 2, B3' is 2' -OMe or 2' -F, q 5 Is 5, T3 'is 2' -F, q 6 Is 1, B4 'is 2' -OMe, and q 7 Is 1; there are two phosphorothioate internucleotide linkage modifications within positions 1 to 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 to 23 of the antisense strand (counting from the 5' end of the antisense strand). RNAi agents also include 5' -VP (e.g., 5' -E-VP, 5' -Z-VP, or a combination thereof) and targeting ligand.
In one embodiment, the 5' -VP is located at the 5' end of the antisense strand and the targeting ligand is located at the 3' end of the sense strand.
In one embodiment, B1 is 2'-OMe or 2' -F, n 1 Is 8, T1 is 2' F, n 2 Is 3, B2 is 2' -OMe, n 3 Is 7, n 4 Is 0, B3 is 2' -OMe, n 5 Is 3, B1' is 2' -OMe or 2' -F, q 1 Is 9, T1 'is 2' -F, q 2 Is 1, B2' is 2' -OMe or 2' -F, q 3 Is 4, T2 'is 2' -F, q 4 Is 2, B3' is 2' -OMe or 2' -F, q 5 Is 5, T3 'is 2' -F, q 6 Is 1, B4 'is 2' -OMe, and q 7 Is 1; having two phosphorothioate internucleotide linkage modifications in positions 1 to 5 of the sense strand (counting from the 5 'end of the sense strand) and two phosphorothioate internucleotide linkage modifications in positions 1 and 2, and in positions 18 to 23 of the antisense strand (counting from the 5' end of the antisense strand)Initial count) of two phosphorothioate internucleotide linkage modifications. RNAi agents also include 5' -PS 2 And a targeting ligand. In one embodiment, 5' -PS 2 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, n 1 Is 8, T1 is 2' F, n 2 Is 3, B2 is 2' -OMe, n 3 Is 7, n 4 Is 0, B3 is 2' -OMe, n 5 Is 3, B1' is 2' -OMe or 2' -F, q 1 Is 9, T1 'is 2' -F, q 2 Is 1, B2' is 2' -OMe or 2' -F, q 3 Is 4, T2 'is 2' -F, q 4 Is 2, B3' is 2' -OMe or 2' -F, q 5 Is 5, T3 'is 2' -F, q 6 Is 1, B4 'is 2' -OMe, and q 7 Is 1; there are two phosphorothioate internucleotide linkage modifications within positions 1 to 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 to 23 of the antisense strand (counting from the 5' end of the antisense strand). RNAi agents also include 5 '-deoxy-5' -C-malonyl and targeting ligands. In one embodiment, the 5 '-deoxy-5' -C-malonyl is located at the 5 'end of the antisense strand and the targeting ligand is located at the 3' end of the sense strand.
In one embodiment, B1 is 2'-OMe or 2' -F, n 1 Is 8, T1 is 2' F, n 2 Is 3, B2 is 2' -OMe, n 3 Is 7, n 4 Is 0, B3 is 2' -OMe, n 5 Is 3, B1' is 2' -OMe or 2' -F, q 1 Is 9, T1 'is 2' -F, q 2 Is 1, B2' is 2' -OMe or 2' -F, q 3 Is 4, q 4 Is 0, B3' is 2' -OMe or 2' -F, q 5 Is 7, T3 'is 2' -F, q 6 Is 1, B4 'is 2' -OMe, and q 7 Is 1; there are two phosphorothioate internucleotide linkage modifications in positions 1 to 5 (counting from the 5 'end) of the sense strand, and two phosphorothioate internucleotide linkage modifications in positions 1 and 2, and two phosphorothioate internucleotide linkage modifications in positions 18 to 23 (counting from the 5' end) of the antisense strand. RNAi agents also include 5' -P and targeting ligands. In one embodiment, 5' -P is located 5 of the antisense strand At the 'end, and the targeting ligand is located at the 3' end of the sense strand.
In one embodiment, B1 is 2'-OMe or 2' -F, n 1 Is 8, T1 is 2' F, n 2 Is 3, B2 is 2' -OMe, n 3 Is 7, n 4 Is 0, B3 is 2' -OMe, n 5 Is 3, B1' is 2' -OMe or 2' -F, q 1 Is 9, T1 'is 2' -F, q 2 Is 1, B2' is 2' -OMe or 2' -F, q 3 Is 4, q 4 Is 0, B3' is 2' -OMe or 2' -F, q 5 Is 7, T3 'is 2' -F, q 6 Is 1, B4 'is 2' -OMe, and q 7 Is 1; there are two phosphorothioate internucleotide linkage modifications in positions 1 to 5 (counting from the 5 'end) of the sense strand, and two phosphorothioate internucleotide linkage modifications in positions 1 and 2, and two phosphorothioate internucleotide linkage modifications in positions 18 to 23 (counting from the 5' end) of the antisense strand. RNAi agents also include 5' -PS and targeting ligands. In one embodiment, the 5' -PS is located at the 5' end of the antisense strand and the targeting ligand is located at the 3' end of the sense strand.
In one embodiment, B1 is 2'-OMe or 2' -F, n 1 Is 8, T1 is 2' F, n 2 Is 3, B2 is 2' -OMe, n 3 Is 7, n 4 Is 0, B3 is 2' -OMe, n 5 Is 3, B1' is 2' -OMe or 2' -F, q 1 Is 9, T1 'is 2' -F, q 2 Is 1, B2' is 2' -OMe or 2' -F, q 3 Is 4, q 4 Is 0, B3' is 2' -OMe or 2' -F, q 5 Is 7, T3 'is 2' -F, q 6 Is 1, B4 'is 2' -OMe, and q 7 Is 1; there are two phosphorothioate internucleotide linkage modifications in positions 1 to 5 (counting from the 5 'end) of the sense strand, and two phosphorothioate internucleotide linkage modifications in positions 1 and 2, and two phosphorothioate internucleotide linkage modifications in positions 18 to 23 (counting from the 5' end) of the antisense strand. RNAi agents also include 5' -VP (e.g., 5' -E-VP, 5' -Z-VP, or a combination thereof) and targeting ligand. In one embodiment, the 5' -VP is located at the 5' end of the antisense strand and the targeting ligand is located at the 3' end of the sense strand.
In one embodiment, B1 is 2'-OMe or 2' -F, n 1 Is 8, T1 is 2' F, n 2 Is 3, B2 is 2' -OMe, n 3 Is 7, n 4 Is 0, B3 is 2' -OMe, n 5 Is 3, B1' is 2' -OMe or 2' -F, q 1 Is 9, T1 'is 2' -F, q 2 Is 1, B2' is 2' -OMe or 2' -F, q 3 Is 4, q 4 Is 0, B3' is 2' -OMe or 2' -F, q 5 Is 7, T3 'is 2' -F, q 6 Is 1, B4 'is 2' -OMe, and q 7 Is 1; there are two phosphorothioate internucleotide linkage modifications in positions 1 to 5 (counting from the 5 'end) of the sense strand, and two phosphorothioate internucleotide linkage modifications in positions 1 and 2, and two phosphorothioate internucleotide linkage modifications in positions 18 to 23 (counting from the 5' end) of the antisense strand. RNAi agents also include 5' -PS 2 And a targeting ligand. In one embodiment, 5' -PS 2 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, n 1 Is 8, T1 is 2' F, n 2 Is 3, B2 is 2' -OMe, n 3 Is 7, n 4 Is 0, B3 is 2' -OMe, n 5 Is 3, B1' is 2' -OMe or 2' -F, q 1 Is 9, T1 'is 2' -F, q 2 Is 1, B2' is 2' -OMe or 2' -F, q 3 Is 4, q 4 Is 0, B3' is 2' -OMe or 2' -F, q 5 Is 7, T3 'is 2' -F, q 6 Is 1, B4 'is 2' -OMe, and q 7 Is 1; there are two phosphorothioate internucleotide linkage modifications in positions 1 to 5 (counting from the 5 'end) of the sense strand, and two phosphorothioate internucleotide linkage modifications in positions 1 and 2, and two phosphorothioate internucleotide linkage modifications in positions 18 to 23 (counting from the 5' end) of the antisense strand. RNAi agents also include 5 '-deoxy-5' -C-malonyl and targeting ligands. In one embodiment, the 5 '-deoxy-5' -C-malonyl is located at the 5 'end of the antisense strand and the targeting ligand is located at the 3' end of the sense strand.
In one embodiment, B1 is 2'-OMe or 2' -F, n 1 Is 8, T1 is 2' F, n 2 Is 3, B2 is 2' -OMe, n 3 Is 7, n 4 Is 0, B3 is 2' -OMe, n 5 Is 3, B1' is 2' -OMe or 2' -F, q 1 Is 9, T1 'is 2' -F, q 2 Is 1, B2' is 2' -OMe or 2' -F, q 3 Is 4T2 'is 2' -F, q 4 Is 2, B3' is 2' -OMe or 2' -F, q 5 Is 5, T3 'is 2' -F, q 6 Is 1, B4 'is 2' -F, and q 7 Is 1; there are two phosphorothioate internucleotide linkage modifications within positions 1 to 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 to 23 of the antisense strand (counting from the 5' end of the antisense strand). RNAi agents also include 5' -P and targeting ligands. In one embodiment, the 5' -P is located at the 5' end of the antisense strand and the targeting ligand is located at the 3' end of the sense strand.
In one embodiment, B1 is 2'-OMe or 2' -F, n 1 Is 8, T1 is 2' F, n 2 Is 3, B2 is 2' -OMe, n 3 Is 7, n 4 Is 0, B3 is 2' -OMe, n 5 Is 3, B1' is 2' -OMe or 2' -F, q 1 Is 9, T1 'is 2' -F, q 2 Is 1, B2' is 2' -OMe or 2' -F, q 3 Is 4, T2 'is 2' -F, q 4 Is 2, B3' is 2' -OMe or 2' -F, q 5 Is 5, T3 'is 2' -F, q 6 Is 1, B4 'is 2' -F, and q 7 Is 1; there are two phosphorothioate internucleotide linkage modifications within positions 1 to 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 to 23 of the antisense strand (counting from the 5' end of the antisense strand). RNAi agents also include 5' -PS and targeting ligands. In one embodiment, the 5' -PS is located at the 5' end of the antisense strand and the targeting ligand is located at the 3' end of the sense strand.
In one embodiment, B1 is 2'-OMe or 2' -F, n 1 Is 8, T1 is 2' F, n 2 Is 3, B2 is 2' -OMe, n 3 Is 7, n 4 Is 0, B3 is 2' -OMe, n 5 Is 3, B1' is 2' -OMe or 2' -F, q 1 Is 9, T1 'is 2' -F, q 2 Is 1, B2' is 2' -OMe or 2' -F, q 3 Is 4, T2 'is 2' -F, q 4 Is 2, B3' is 2' -OMe or 2' -F, q 5 Is 5, T3 'is 2' -F, q 6 Is 1, B4 'is 2' -F, and q 7 Is 1; in positions 1 to 5 of the sense strand (fromThe 5 'end of the sense strand starts counting) has two phosphorothioate internucleotide linkage modifications, and two phosphorothioate internucleotide linkage modifications at positions 1 and 2, and two phosphorothioate internucleotide linkage modifications in positions 18 to 23 of the antisense strand (counting from the 5' end of the antisense strand). RNAi agents also include 5' -VP (e.g., 5' -E-VP, 5' -Z-VP, or a combination thereof) and targeting ligand. In one embodiment, the 5' -VP is located at the 5' end of the antisense strand and the targeting ligand is located at the 3' end of the sense strand.
In one embodiment, B1 is 2'-OMe or 2' -F, n 1 Is 8, T1 is 2' F, n 2 Is 3, B2 is 2' -OMe, n 3 Is 7, n 4 Is 0, B3 is 2' -OMe, n 5 Is 3, B1' is 2' -OMe or 2' -F, q 1 Is 9, T1 'is 2' -F, q 2 Is 1, B2' is 2' -OMe or 2' -F, q 3 Is 4, T2 'is 2' -F, q 4 Is 2, B3' is 2' -OMe or 2' -F, q 5 Is 5, T3 'is 2' -F, q 6 Is 1, B4 'is 2' -F, and q 7 Is 1; there are two phosphorothioate internucleotide linkage modifications within positions 1 to 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 to 23 of the antisense strand (counting from the 5' end of the antisense strand). RNAi agents also include 5' -PS 2 And a targeting ligand. In one embodiment, 5' -PS 2 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, n 1 Is 8, T1 is 2' F, n 2 Is 3, B2 is 2' -OMe, n 3 Is 7, n 4 Is 0, B3 is 2' -OMe, n 5 Is 3, B1' is 2' -OMe or 2' -F, q 1 Is 9, T1 'is 2' -F, q 2 Is 1, B2' is 2' -OMe or 2' -F, q 3 Is 4, T2 'is 2' -F, q 4 Is 2, B3' is 2' -OMe or 2' -F, q 5 Is 5, T3 'is 2' -F, q 6 Is 1, B4 'is 2' -F, and q 7 Is 1; having two phosphorothioate internucleotide linkage modifications in positions 1 to 5 of the sense strand (counting from the 5' end of the sense strand) and two at positions 1 and 2Phosphorothioate internucleotide linkage modifications and there are two phosphorothioate internucleotide linkage modifications within positions 18 to 23 of the antisense strand (counting from the 5' end of the antisense strand). RNAi agents also include 5 '-deoxy-5' -C-malonyl and targeting ligands. In one embodiment, the 5 '-deoxy-5' -C-malonyl is located at the 5 'end of the antisense strand and the targeting ligand is located at the 3' end of the sense strand.
In one embodiment, B1 is 2'-OMe or 2' -F, n 1 Is 8, T1 is 2' F, n 2 Is 3, B2 is 2' -OMe, n 3 Is 7, n 4 Is 0, B3 is 2' -OMe, n 5 Is 3, B1' is 2' -OMe or 2' -F, q 1 Is 9, T1 'is 2' -F, q 2 Is 1, B2' is 2' -OMe or 2' -F, q 3 Is 4, q 4 Is 0, B3' is 2' -OMe or 2' -F, q 5 Is 7, T3 'is 2' -F, q 6 Is 1, B4 'is 2' -F, and q 7 Is 1; there are two phosphorothioate internucleotide linkage modifications within positions 1 to 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 to 23 of the antisense strand (counting from the 5' end of the antisense strand). RNAi agents also include 5' -P and targeting ligands. In one embodiment, the 5' -P is located at the 5' end of the antisense strand and the targeting ligand is located at the 3' end of the sense strand.
In one embodiment, B1 is 2'-OMe or 2' -F, n 1 Is 8, T1 is 2' F, n 2 Is 3, B2 is 2' -OMe, n 3 Is 7, n 4 Is 0, B3 is 2' -OMe, n 5 Is 3, B1' is 2' -OMe or 2' -F, q 1 Is 9, T1 'is 2' -F, q 2 Is 1, B2' is 2' -OMe or 2' -F, q 3 Is 4, q 4 Is 0, B3' is 2' -OMe or 2' -F, q 5 Is 7, T3 'is 2' -F, q 6 Is 1, B4 'is 2' -F, and q 7 Is 1; there are two phosphorothioate internucleotide linkage modifications within positions 1 to 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 to 23 of the antisense strand (counting from the 5' end of the antisense strand). RNAi drugsAgents also include 5' -PS and targeting ligands. In one embodiment, the 5' -PS is located at the 5' end of the antisense strand and the targeting ligand is located at the 3' end of the sense strand.
In one embodiment, B1 is 2'-OMe or 2' -F, n 1 Is 8, T1 is 2' F, n 2 Is 3, B2 is 2' -OMe, n 3 Is 7, n 4 Is 0, B3 is 2' -OMe, n 5 Is 3, B1' is 2' -OMe or 2' -F, q 1 Is 9, T1 'is 2' -F, q 2 Is 1, B2' is 2' -OMe or 2' -F, q 3 Is 4, q 4 Is 0, B3' is 2' -OMe or 2' -F, q 5 Is 7, T3 'is 2' -F, q 6 Is 1, B4 'is 2' -F, and q 7 Is 1; there are two phosphorothioate internucleotide linkage modifications within positions 1 to 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 to 23 of the antisense strand (counting from the 5' end of the antisense strand). RNAi agents also include 5' -VP (e.g., 5' -E-VP, 5' -Z-VP, or a combination thereof) and targeting ligand. In one embodiment, the 5' -VP is located at the 5' end of the antisense strand and the targeting ligand is located at the 3' end of the sense strand.
In one embodiment, B1 is 2'-OMe or 2' -F, n 1 Is 8, T1 is 2' F, n 2 Is 3, B2 is 2' -OMe, n 3 Is 7, n 4 Is 0, B3 is 2' -OMe, n 5 Is 3, B1' is 2' -OMe or 2' -F, q 1 Is 9, T1 'is 2' -F, q 2 Is 1, B2' is 2' -OMe or 2' -F, q 3 Is 4, q 4 Is 0, B3' is 2' -OMe or 2' -F, q 5 Is 7, T3 'is 2' -F, q 6 Is 1, B4 'is 2' -F, and q 7 Is 1; there are two phosphorothioate internucleotide linkage modifications within positions 1 to 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 to 23 of the antisense strand (counting from the 5' end of the antisense strand). RNAi agents also include 5' -PS 2 And a targeting ligand. In one embodiment, 5' -PS 2 At the 5 'end of the antisense strand and a targeting ligand at the 3' end of the sense strand。
In one embodiment, B1 is 2'-OMe or 2' -F, n 1 Is 8, T1 is 2' F, n 2 Is 3, B2 is 2' -OMe, n 3 Is 7, n 4 Is 0, B3 is 2' -OMe, n 5 Is 3, B1' is 2' -OMe or 2' -F, q 1 Is 9, T1 'is 2' -F, q 2 Is 1, B2' is 2' -OMe or 2' -F, q 3 Is 4, q 4 Is 0, B3' is 2' -OMe or 2' -F, q 5 Is 7, T3 'is 2' -F, q 6 Is 1, B4 'is 2' -F, and q 7 Is 1; there are two phosphorothioate internucleotide linkage modifications within positions 1 to 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 to 23 of the antisense strand (counting from the 5' end of the antisense strand). RNAi agents also include 5 '-deoxy-5' -C-malonyl and targeting ligands. In one embodiment, the 5 '-deoxy-5' -C-malonyl is located at the 5 'end of the antisense strand and the targeting ligand is located at the 3' end of the sense strand.
In a particular embodiment, the RNAi agent of the invention comprises:
(a) A sense strand having:
(i) A length of 21 nucleotides;
(ii) An ASGPR ligand linked to the 3' end, wherein the ASGPR ligand comprises three GalNAc derivatives linked by 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 (counting from the 5' end) at positions 2, 4, 6, 8, 12, 14 to 16, 18 and 20;
and
(b) An antisense strand, said 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 (counting from the 5' end) between nucleotide positions 21 and 22 and between nucleotide positions 22 and 23;
wherein the dsRNA agent has 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, the RNAi agent of the invention comprises:
(a) A sense strand having:
(i) A length of 21 nucleotides;
(ii) An ASGPR ligand linked to the 3' end, wherein the ASGPR ligand comprises three GalNAc derivatives linked by 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 (counting from the 5' end) between nucleotide positions 1 and 2 and between nucleotide positions 2 and 3;
and
(b) An antisense strand, said 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 (counting from the 5' end) 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;
wherein the RNAi agent has two nucleotide overhangs at the 3 'end of the antisense strand and a blunt end at the 5' end of the antisense strand.
In another particular embodiment, the RNAi agent of the invention comprises:
(a) A sense strand having:
(i) A length of 21 nucleotides;
(ii) An ASGPR ligand linked to the 3' end, wherein the ASGPR ligand comprises three GalNAc derivatives linked by 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 deoxynucleotides (e.g., dT) at position 11 (counted from the 5' end); and
(iv) Phosphorothioate internucleotide linkages (counting from the 5' end) between nucleotide positions 1 and 2 and between nucleotide positions 2 and 3;
and
(b) An antisense strand, said 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 (counting from the 5' end) 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;
wherein the RNAi agent has two nucleotide overhangs at the 3 'end of the antisense strand and a blunt end at the 5' end of the antisense strand.
In another particular embodiment, the RNAi agent of the invention comprises:
(a) A sense strand having:
(i) A length of 21 nucleotides;
(ii) An ASGPR ligand linked to the 3' end, wherein the ASGPR ligand comprises three GalNAc derivatives linked by 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 (counting from the 5' end) between nucleotide positions 1 and 2 and between nucleotide positions 2 and 3;
and
(b) An antisense strand, said 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 (counting from the 5' end) 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;
wherein the RNAi agent has two nucleotide overhangs at the 3 'end of the antisense strand and a blunt end at the 5' end of the antisense strand.
In another particular embodiment, the RNAi agent of the invention comprises:
(a) A sense strand having:
(i) A length of 21 nucleotides;
(ii) An ASGPR ligand linked to the 3' end, wherein the ASGPR ligand comprises three GalNAc derivatives linked by 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 (counting from the 5' end) between nucleotide positions 1 and 2 and between nucleotide positions 2 and 3;
and
(b) An antisense strand, said 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 (counting from the 5' end) 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;
wherein the RNAi agent has two nucleotide overhangs at the 3 'end of the antisense strand and a blunt end at the 5' end of the antisense strand.
In another particular embodiment, the RNAi agent of the invention comprises:
(a) A sense strand having:
(i) A length of 21 nucleotides;
(ii) An ASGPR ligand linked to the 3' end, wherein the ASGPR ligand comprises three GalNAc derivatives linked by 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 (counting from the 5' end) between nucleotide positions 1 and 2 and between nucleotide positions 2 and 3;
and
(b) An antisense strand, said 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 (counting from the 5' end) 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;
wherein the RNAi agent has two nucleotide overhangs at the 3 'end of the antisense strand and a blunt end at the 5' end of the antisense strand.
In another particular embodiment, the RNAi agent of the invention comprises:
(a) A sense strand having:
(i) A length of 21 nucleotides;
(ii) An ASGPR ligand linked to the 3' end, wherein the ASGPR ligand comprises three GalNAc derivatives linked by 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 (counting from the 5' end) between nucleotide positions 1 and 2 and between nucleotide positions 2 and 3;
and
(b) An antisense strand, said 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 deoxynucleotides (e.g., dT) at positions 24 and 25 (counted from the 5' end); and
(iii) Phosphorothioate internucleotide linkages (counting from the 5' end) 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;
wherein the RNAi agent has 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, the RNAi agent of the invention comprises:
(a) A sense strand having:
(i) A length of 21 nucleotides;
(ii) An ASGPR ligand linked to the 3' end, wherein the ASGPR ligand comprises three GalNAc derivatives linked by 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 (counting from the 5' end) between nucleotide positions 1 and 2 and between nucleotide positions 2 and 3;
and
(b) An antisense strand, said 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 (counting from the 5' end) 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;
Wherein the RNAi agent has two nucleotide overhangs at the 3 'end of the antisense strand and a blunt end at the 5' end of the antisense strand.
In another particular embodiment, the RNAi agent of the invention comprises:
(a) A sense strand having:
(i) A length of 21 nucleotides;
(ii) An ASGPR ligand linked to the 3' end, wherein the ASGPR ligand comprises three GalNAc derivatives linked by 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 (counting from the 5' end) between nucleotide positions 1 and 2 and between nucleotide positions 2 and 3;
and
(b) An antisense strand, said 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 (counting from the 5' end) 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;
wherein the RNAi agent has two nucleotide overhangs at the 3 'end of the antisense strand and a blunt end at the 5' end of the antisense strand.
In another particular embodiment, the RNAi agent of the invention comprises:
(a) A sense strand having:
(i) A length of 19 nucleotides;
(ii) An ASGPR ligand linked to the 3' end, wherein the ASGPR ligand comprises three GalNAc derivatives linked by 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 (counting from the 5' end) between nucleotide positions 1 and 2 and between nucleotide positions 2 and 3;
and
(b) An antisense strand, said 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 (counting from the 5' end) 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;
wherein the RNAi agent has two nucleotide overhangs 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 used in the methods of the invention is an agent selected from the agents listed in any one of tables 2-7. These agents may further comprise a ligand.
iRNA conjugated to ligand
Another modification of the RNAs of the iRNAs of the present invention involves chemically linking one or more ligands, moieties, or conjugates to the iRNA that enhance the activity, cellular distribution, or cellular uptake (e.g., into cells) of the iRNA. Such moieties include, but are not limited to, lipid moieties, such as cholesterol moieties (Letsinger et al, proc. Natl. Acid. Sci. USA, 1989, 86:6553-6556). In other embodiments, the ligand is cholic acid (Manoharan et al, biological organic and pharmaceutical chemistry flash (Biorg. Med. Chem. Let.), (1994, 4: 1053-1060), a thioether, e.g., a chlorohydrin-S-triphenylmethyl thiol (Manoharan et al, new York academy of sciences (Ann. N. Y. Acad. Sci.), 1992,660:306-309; manoharan et al, biological organic and pharmaceutical chemistry flash (1993, 3: 2765-2770), thiocholesterol (Obohauser et al, nucleic acids research (1992, 20: 533-538)), an aliphatic chain, e.g., dodecanediol or undecyl residues (Saison-Behmaras et al, european journal of molecular biology (EMBO), 1991,10: 1111-Kabav et al, FEBS (Lert. 1990: 330, lert. 330), biochemistry (Biochimie), 1993, 75:49-54), phospholipids such as hexacosyl-rac-glycerol or triethylammonium 1, 2-di-O-hexadecyl-rac-glycerol-3-phosphonate (Manoharan et al, tetrahedron Lett.) 1995,36:3651-3654; shea et al, nucleic acids research 1990, 18:3777-3783), phospholipids, polyamines or polyethylene glycol chains (Manoharan et al, nucleosides & Nucleotides (Nucleosides) 1995, 14:969-973) or adamantane acetic acid (Manoharan et al, tetrahedron et al, 1995, 36:3651-3654), palmitoyl moieties (Mira et al, biochemistry and biophysics acta, 1990, 18:3777-3783), phospholipids, polyamines or polyethylene glycol chains (Manoharan et al, nucleosides & Nucleotides & Nucleosides), 1995, 14:969-973) or adamantane acetic acid (Manohara et al, 1995, 36:3651-3654), palmitoyl moieties (Mira et al, biochemistry and biophysics, equiz. Acids, 1994, oxygen-carbonyl moieties (Crohexanoes) or cholesterol et al, journal of pharmacology and experimental therapeutics (J.Pharmacol. Exp. Ther.), 1996, 277:923-937.
In certain embodiments, the ligand alters the distribution, targeting, or lifetime of the iRNA agent into which it is incorporated. In some embodiments, the ligand provides enhanced affinity for a selected target (e.g., a molecule, cell, or cell type), compartment (e.g., a cell or organ compartment, tissue, organ, or body region), for example, as compared to a species in which such ligand is not present. In some embodiments, the ligand does not participate in duplex pairing in duplex nucleic acids.
The ligand may comprise naturally occurring substances, such as proteins (e.g., human Serum Albumin (HSA), low Density Lipoprotein (LDL), or globulin); carbohydrates (e.g., dextran, pullulan, chitin, chitosan, inulin, cyclodextrin, N-acetylglucosamine, N-acetylgalactosamine, or hyaluronic acid); or a lipid. The ligand may also be a recombinant molecule or a synthetic molecule, such as a synthetic polymer, e.g. a synthetic polyamino acid. Examples of the polyamino acid include polyamino acids, namely Polylysine (PLL), poly L-aspartic acid, poly L-glutamic acid, styrene-maleic anhydride copolymer, poly (L-lactide-co-glycolide) copolymer, divinyl ether-maleic anhydride copolymer, N- (2-hydroxypropyl) methacrylamide copolymer (HMPA), polyethylene glycol (PEG), polyvinyl alcohol (PVA), polyurethane, poly (2-ethylacrylic acid), N-isopropylacrylamide polymer or polyphosphazine. Examples of polyamines include: polyethyleneimine, polylysine (PLL), spermine, spermidine, polyamine, pseudopeptide-polyamine, peptidomimetic polyamine, dendrimer polyamine, arginine, amidine, protamine, cationic lipid, cationic porphyrin, quaternary salts of polyamines, or alpha helical peptides.
The ligand may also comprise a targeting group, such as a cell or tissue targeting agent, e.g. a lectin, glycoprotein, lipid or protein, e.g. an antibody that binds to a specific cell type (e.g. kidney cells). The targeting group may be thyrotropin, melanotropin, lectin, glycoprotein, surfactant protein a, mucin carbohydrate, multivalent lactose, multivalent galactose, N-acetyl-galactosamine, N-acetyl-glucosamine multivalent mannose, multivalent fucose, glycosylated polyamino acid, multivalent galactose, transferrin, bisphosphonate, polyglutamate, polyaspartate, lipid, cholesterol, steroid, bile acid, folic acid, vitamin B12, vitamin a, biotin, or RGD peptide mimetic. In certain embodiments, the ligand is a multivalent galactose, e.g., N-acetyl-galactosamine.
Other examples of ligands include dyes, intercalators (e.g., acridine), crosslinkers (e.g., psoralene, mitomycin C (mitomycin C)), porphyrins (TPPC 4, texaphyrin (texaphyrin), ring-extended porphyrins (saphyrin)), polycyclic aromatic hydrocarbons (e.g., phenazine, dihydrophenazine), artificial endonucleases (e.g., EDTA), lipophilic molecules (e.g., cholesterol, cholic acid, adamantaneacetic acid, 1-pyrene butyric acid, dihydrotestosterone, 1, 3-bis-O (hexadecyl) glycerol, geranyloxyhexyl, hexadecyl glycerol, borneol, menthol, 1, 3-propanediol) Heptadecyl, palmitic acid, myristic acid, O3- (oleoyl) lithocholic acid, O3- (oleoyl) cholanic acid, dimethoxytrityl or phenoxazine), peptide conjugates (e.g., antennapedia mutant peptides, tat peptides), alkylating agents, phosphates, amino groups, sulfhydryl groups, PEG (e.g., PEG-40K), MPEG, [ MPEG ]] 2 Polyamino groups, alkyl groups, substituted alkyl groups, radiolabelled markers, enzymes, haptens (e.g., biotin), transport/absorption enhancers (e.g., aspirin (aspirin), vitamin E, folic acid), synthetic ribonucleases (e.g., imidazole, bisimidazole, histamine, imidazole clusters, acridine-imidazole conjugates, eu3+ complexes of tetraazamacrocyclic compounds), dinitrophenyl, HRP or AP.
The ligand may be a protein (e.g., glycoprotein) or peptide (e.g., a molecule having a specific affinity for the co-ligand) or an antibody (e.g., an antibody that binds to a particular cell type, such as a hepatocyte). The ligand may also comprise a hormone and a hormone receptor. The ligand may also comprise non-peptide substances such as lipids, lectins, carbohydrates, vitamins, cofactors, multivalent lactose, multivalent galactose, N-acetyl-galactosamine, N-acetyl-glucosamine multivalent mannose or multivalent fucose. The ligand may be, for example, lipopolysaccharide, an activator of p38MAP kinase or an activator of NF- κB.
The ligand may be a substance, such as a drug, which may increase uptake of the iRNA agent into the cell, for example, by disrupting the cytoskeleton of the cell, such as by disrupting microtubules, microfilaments or intermediate filaments of the cell. The drug may be, for example, taxol, vincristine, vinblastine, cytochalasin, nocodazole, jestide, lappachinolide, ragalolin A (latrunculin A), phalloidin, ste Wen Heli A (swinholide A), yin Dannuo octyl (indanocine) or myosin.
In some embodiments, the ligand linked to an iRNA as described herein acts as a pharmacokinetic modulator (PK modulator). PK modulators include lipophilic substances, bile acids, steroids, phospholipid analogs, peptides, protein binders, PEG, vitamins, and the like. Exemplary PK modulators include, but are not limited to, cholesterol, fatty acids, cholic acid, lithocholic acid, dialkyl glycerides, diacylglycerides, phospholipids, sphingolipids, naproxen (naproxen), ibuprofen (ibuprofen), vitamin E, biotin. Serum protein-binding oligonucleotides comprising a number of phosphorothioate linkages are also known, and thus short oligonucleotides comprising a number of phosphorothioate linkages in the backbone (e.g., oligonucleotides having about 5 bases, 10 bases, 15 bases, or 20 bases) are also suitable for use in the present invention as ligands (e.g., as PK modulating ligands). In addition, aptamers that bind to serum components (e.g., serum proteins) are also suitable for use as PK modulating ligands in the embodiments described herein.
The ligand-conjugated iRNA of the invention can be synthesized by using oligonucleotides with side-reactive functions, such as those derived from linking molecules attached to oligonucleotides (as described below). Such reactive oligonucleotides may be reacted directly with commercially available ligands, synthetic ligands with any of a variety of protecting groups, or ligands having a linking moiety attached thereto.
The oligonucleotides used in the conjugates of the invention may be conveniently and routinely prepared by well known solid phase synthesis techniques. The equipment for such synthesis is sold by several suppliers, e.g. including Applied(Foster City, california, calif.). Any other method known in the art for such synthesis may additionally or alternatively be employed. Other oligonucleotides (e.g., phosphorothioates and alkylated derivatives) are also known to be prepared using similar techniques.
In the ligand-conjugated iRNA and ligand molecules with sequence-specific linked nucleosides of the invention, the oligonucleotides and oligonucleotides can be assembled on a suitable DNA synthesizer using standard nucleotides or nucleoside precursors or nucleotides or nucleoside conjugate precursors already bearing a linking moiety, ligand-nucleotides or nucleoside conjugate precursors already bearing a ligand molecule or non-nucleoside ligands bearing a building block.
When using nucleotide conjugate precursors that already carry a linking moiety, synthesis of the sequence-specific linked nucleoside is typically accomplished, and then the ligand molecule reacts with the linking moiety to form a ligand conjugated oligonucleotide. In some embodiments, the oligonucleotides or linked nucleosides of the invention are synthesized by an automated synthesizer using a phosphoramidite derived from a ligand-nucleoside conjugate as well as standard and non-standard phosphoramidites commercially available and conventionally used for oligonucleotide synthesis.
A. Lipid conjugates
In certain embodiments, the ligand or conjugate is a lipid or lipid-based molecule. In one embodiment, such lipids or lipid-based molecules bind to serum proteins, such as Human Serum Albumin (HSA). HSA binding ligands allow the conjugate to be distributed to target tissue, e.g., non-kidney target tissue of the body. For example, the target tissue may be the liver, including parenchymal cells of the liver. Other molecules that can bind to HSA can also be used as ligands. For example, naproxen or aspirin may be used. The lipid or lipid-based ligand may (a) increase resistance to conjugate degradation, (b) increase targeting or transport into a target cell or cell membrane, or (c) may be used to modulate binding of HSA to a serum protein, such as HSA.
Lipid-based ligands can be used to inhibit (e.g., control) binding of the conjugate to a target tissue. For example, lipids or lipid-based ligands that bind more strongly to HSA will be less likely to be targeted to the kidneys and therefore less likely to be cleared from the body. Lipids or lipid-based ligands that bind less strongly to HSA can be used to target the conjugate to the kidney.
In certain embodiments, the lipid-based ligand binds to HSA. In one embodiment, it binds to HSA with sufficient affinity such that the conjugate will distribute to non-kidney tissue. However, it is preferred that the affinity is not so strong that HSA ligand binding cannot be reversed.
In other embodiments, the lipid-based ligand binds to HSA weakly or not at all. In one embodiment, the conjugate will be distributed to the kidneys. Other moieties targeted to kidney cells may also be used instead of or in addition to lipid-based ligands.
On the other hand, the ligand is a moiety, such as a vitamin, that is taken up by the target cell (e.g., proliferating cell). These are particularly useful for treating diseases characterized by undesired cell proliferation, such as malignant or non-malignant types of diseases, such as cancer cells. Exemplary vitamins include vitamins A, E and K. Other exemplary vitamins that are included are B vitamins such as folic acid, B12, riboflavin, biotin, pyridoxal, or other vitamins or nutrients that are taken up by target cells (e.g., hepatocytes). HSA and Low Density Lipoprotein (LDL) are also included.
B. Cell penetrating agent
In another aspect, the ligand is a cell penetrating agent, such as a helical cell penetrating agent. In one embodiment, the agent is amphiphilic. Exemplary agents are peptides, such as tat or antennapedia mutant peptides. If the agent is a peptide, it may be modified, including peptidomimetics, inversion bodies, non-peptide or pseudopeptide bonds, and the use of D-amino acids. In one embodiment, the helices are alpha helices that have a lipophilic and lipophobic phase.
The ligand may be a peptide or a peptidomimetic. A peptidomimetic (also referred to herein as an oligopeptide mimetic) is a molecule capable of folding into a defined three-dimensional structure similar to a natural peptide. The attachment of peptides and peptidomimetics to iRNA agents can affect the pharmacokinetic profile of the iRNA, such as by enhancing cell recognition and uptake. The peptide or peptidomimetic moiety can be about 5 to 50 amino acids in length, for example about 5, 10, 15, 20, 25, 30, 35, 40, 45, or 50 amino acids in length.
The peptide or peptidomimetic can be, for example, a cell penetrating peptide, a cationic peptide, an amphiphilic peptide, or a hydrophobic peptide (e.g., consisting essentially of Tyr, trp, or Phe). The peptide moiety may be a dendrimer peptide, a constraint peptide or a cross-linked peptide. In another alternative, the peptide moiety may comprise a hydrophobic Membrane Translocation Sequence (MTS). An exemplary hydrophobic MTS-containing peptide is RFGF having the amino acid sequence AAVALLPAVLLALLAP (SEQ ID NO: 14). RFGF analogs (e.g., amino acid sequence AALLPVLLAAP (SEQ ID NO: 15)) containing a hydrophobic MTS can also be targeting moieties. The peptide moiety may be a "delivery" peptide that can carry a large polar molecule including peptides, oligonucleotides and proteins across the cell membrane. For example, it has been found that sequences from the HIV Tat protein (GRKKRRQRRRRRPPQ (SEQ ID NO: 16) and drosophila antennapedia mutein (RQIKIWFQNRRMKWKK (SEQ ID NO: 17)) can act as delivery peptides.
RGD peptides 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 particular tissue. RGD-containing peptides and peptide dimers may comprise D-amino acids and synthetic RGD mimics. In addition to RGD, other moieties that target integrin ligands, such as PECAM-1 or VEGF, may also be used.
"cell penetrating peptide" is capable of penetrating a cell, such as a microbial cell (e.g., a bacterial or fungal cell) or a mammalian cell (e.g., a human cell). The microbial cell penetrating peptide may be, for example, an alpha-helical linear peptide (e.g., LL-37 or cerpin P1), a disulfide-containing peptide (e.g., an alpha-defensin, a beta-defensin, or a bacteriocin), or a peptide containing only one or two major amino acids (e.g., PR-39 or indomethacin). Cell penetrating peptides may also comprise Nuclear Localization Signals (NLS). For example, the cell penetrating peptide may be a bipartite amphiphilic peptide, such as MPG, derived from the fusion peptide domain of HIV-1gp41 and NLS of the SV40 large T antigen (Simeoni et al, nucleic acids research 31:2717-2724,2003).
C. Carbohydrate conjugates
In some embodiments of the compositions and methods of the invention, the iRNA further comprises a carbohydrate. Carbohydrate conjugated iRNA is advantageous for in vivo delivery of nucleic acids and compositions suitable for in vivo therapeutic use as described herein. As used herein, "carbohydrate" refers to a compound that is a carbohydrate that itself is comprised of one or more monosaccharide units having at least 6 carbon atoms (which may be linear, branched, or cyclic), wherein an oxygen, nitrogen, or sulfur atom is bound to each carbon atom; or a compound having as part thereof a carbohydrate moiety made up of one or more monosaccharide units each having at least six carbon atoms (which may be linear, branched or cyclic), wherein an oxygen, nitrogen or sulfur atom is bonded to each carbon atom. Representative carbohydrates include sugars (monosaccharides, disaccharides, trisaccharides, and oligosaccharides containing about 4, 5, 6, 7, 8, or 9 monosaccharide units) and polysaccharides such as starch, glycogen, cellulose, and polysaccharide gums. Specific monosaccharides include C5 and above (e.g., C5, C6, C7, or C8) sugars; disaccharides and trisaccharides include saccharides having two or three monosaccharide units (e.g., C5, C6, C7, or C8).
In certain embodiments, the carbohydrate conjugates used in the compositions and methods of the invention are monosaccharides.
In certain embodiments, the monosaccharide is N-acetylgalactosamine (GalNAc). GalNAc conjugates comprising one or more N-acetylgalactosamine (GalNAc) derivatives are described, for example, in US 8,106,022, the entire contents of which are hereby incorporated by reference. In some embodiments, galNAc conjugates are used as ligands to target iRNA to a particular cell. In some embodiments, galNAc conjugates target iRNA to hepatocytes, for example, by acting as a ligand for an asialoglycoprotein receptor of a hepatocyte (e.g., hepatocyte).
In some embodiments, the carbohydrate conjugate includes one or more GalNAc derivatives. GalNAc derivatives may be linked by a linker, for example a divalent 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) through a linker, such as the linkers 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) through a linker, such as the linkers described herein.
In certain embodiments of the invention, galNAc or GalNAc derivative is linked to an iRNA agent of the invention by a monovalent linker. In some embodiments, galNAc or GalNAc derivative is linked to an iRNA agent of the invention through a divalent linker. In still other embodiments of the invention, galNAc or GalNAc derivative is linked to the iRNA agent of the invention through a trivalent linker. In other embodiments of the invention, galNAc or GalNAc derivative is linked to the iRNA agent of the invention through a tetravalent linker.
In certain embodiments, the double stranded RNAi agents of the invention comprise one GalNAc or GalNAc derivative linked to an iRNA agent. In certain embodiments, the double stranded RNAi agents of the invention comprise a plurality (e.g., 2, 3, 4, 5, or 6) galnacs or GalNAc derivatives, each of which is independently linked to a plurality of nucleotides of the double stranded RNAi agent by a plurality of monovalent linkers.
In some embodiments, for example, when two strands of an iRNA agent of the invention are part of one larger molecule, the larger molecule is joined by an uninterrupted nucleotide strand between the 3 'end of one strand and the 5' end of the corresponding other strand, forming a hairpin loop comprising a plurality of unpaired nucleotides, each unpaired nucleotide within the hairpin loop can independently comprise GalNAc or a GalNAc derivative joined by 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 two strands of an iRNA agent of the invention are part of one larger molecule, the larger molecule is joined by an uninterrupted nucleotide strand between the 3 'end of one strand and the 5' end of the corresponding other strand, forming a hairpin loop comprising a plurality of unpaired nucleotides, each unpaired nucleotide within the hairpin loop can independently comprise GalNAc or a GalNAc derivative joined by a monovalent linker. The hairpin loop may also be formed by an extended overhang in one strand of the duplex.
In one embodiment, the carbohydrate conjugates used in the compositions and methods of the invention are selected from the group consisting of:
/>
/>
/>
/>
wherein Y is O or S and n is 3 to 6 (formula XXIV); />
Wherein Y is O or S, and n is 3 to 6 (formula XXV);
wherein X is O or S (formula XXVII); />
/>
In another embodiment, the carbohydrate conjugates used in the compositions and methods of the invention are monosaccharides. In one embodiment, the monosaccharide is N-acetylgalactosamine, e.g
/>
In some embodiments, the RNAi agent is linked to the carbohydrate conjugate via a linker, as shown in the following schematic, wherein X is O or S
In some embodiments, the RNAi agent is conjugated to L96 as defined in table 1, and is as follows:
Another representative carbohydrate conjugate for use in the embodiments described herein includes, but is not limited to:
(formula XXXVI) when one of X or Y is an oligonucleotide, the other is hydrogen.
In some embodiments, suitable ligands are those disclosed in WO 2019/055633, the entire contents of which are incorporated herein by reference. In one embodiment, the ligand comprises the following structure:
in certain embodiments of the invention, galNAc or GalNAc derivative is linked to an iRNA agent of the invention by a monovalent linker. In some embodiments, galNAc or GalNAc derivative is linked to an iRNA agent of the invention through a divalent linker. In still other embodiments of the invention, galNAc or GalNAc derivative is linked to the iRNA agent of the invention through a trivalent linker.
In one embodiment, the double stranded RNAi agents of the invention comprise one or more GalNAc or GalNAc derivatives linked to an iRNA agent. GalNAc can be linked to any nucleotide by a linker on the sense or antisense strand. GalNac can be linked 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, galNAc is linked to the 3' end of the sense strand, for example, by 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) galnacs or GalNAc derivatives, each of which is independently linked to a plurality of nucleotides of the double stranded RNAi agent by a plurality of linkers (e.g., monovalent linkers).
In some embodiments, for example, when two strands of an iRNA agent of the invention are part of one larger molecule, the larger molecule is joined by an uninterrupted nucleotide strand between the 3 'end of one strand and the 5' end of the corresponding other strand, forming a hairpin loop comprising a plurality of unpaired nucleotides, each unpaired nucleotide within the hairpin loop can independently comprise GalNAc or a GalNAc derivative joined by 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 penetrating 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. Joint
In some embodiments, the conjugates or ligands described herein can be attached to an iRNA oligonucleotide through various linkers, which may be cleavable or non-cleavable.
The term "linker" or "linking group" means an organic moiety that connects two moieties of a compound, e.g., covalently connects two moieties of a compound. The linker typically includes a direct bond or atom, such as oxygen or sulfur; units, e.g. NR8, C (O) NH, SO 2 、SO 2 NH or an atomic chain such as, but not limited to, substituted or unsubstituted alkyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl, aralkyl, aralkenyl, aralkynyl, heteroarylalkyl, heteroarylalkenyl, heteroarylalkynyl, heterocycloalkynyl, heterocycloalkenyl, heterocycloalkynyl, aryl, heteroaryl, heterocycloalkynyl, cycloalkyl, cycloalkenyl, alkylarylalkyl, alkylarylakenyl, alkylarylalkynyl, alkenylaralkyl, alkenylarylalkenyl, alkenylarylalkynyl, alkynylarylalkenyl, alkynylarylalkynyl, alkylheteroarylalkyl, alkylheteroarylalkenyl, alkylheteroarylalkynyl, alkenylheteroarylalkyl, alkenylheteroarylalkenyl, alkenylheteroarylalkynyl, alkynylheteroarylalkyl, alkynylheteroarylalkenyl, alkynylheteroarylalkynyl, alkylheterocycloalkyl, alkylheterocycloalkenyl, alkenylheterocycloalkenyl, alkenylheterocycloalkynyl, alkynylheterocycloalkynyl, alkylaryl, alkenylaryl, alkynylaryl, alkylheteroaryl, alkenylheteroaryl, alkynylheteroaryl, one or more of which may be blocked by: o, S, S (O), SO 2 N (R8), C (O), substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl, or substituted or unsubstituted heterocyclyl; wherein R8 is hydrogen, acyl, aliphatic or substituted aliphatic. In one embodiment, the linker is about 1 to 24 atoms, 2 to 24, 3 to 24, 4 to 24, 5 to 24,6 to 24, 6 to 18, 7 to 18, 8 to 18, 7 to 17, 8 to 17, 6 to 16, 7 to 17, or 8 to 16 atoms.
The cleavable linking group is one that is sufficiently stable outside the cell, but which cleaves after entry into the target cell to release the two parts of the linker that remain together. In exemplary embodiments, cleavage of the cleavable linking group in a target cell or under a first reference condition (which may, for example, be selected to mimic or represent an intracellular condition) is at least about 10-fold, 20-fold, 30-fold, 40-fold, 50-fold, 60-fold, 70-fold, 80-fold, 90-fold, or more, or at least 100-fold greater than cleavage rate in a subject's blood or under a second reference condition (which may, for example, be selected to mimic or represent a condition found in blood or serum).
Cleavable linking groups are susceptible to cleavage by a cleavage agent (e.g., pH, redox potential, or the presence of a degrading molecule). Generally, cleavage agents are more prevalent inside cells than in serum or blood, or are found at higher levels or activities. Examples of such degradation agents include: redox agents selected for a particular substrate or not having substrate specificity, including, for example, an oxidation or reduction enzyme or reducing agent present in the cell, such as a thiol, which can cleave a redox cleavable linking group by reductive degradation; an esterase; endosomes or agents that can produce an acidic environment, e.g., those that produce a pH of five or less; enzymes that hydrolyze or degrade acid cleavable linkers can be used as broad acids, peptidases (which may be substrate specific), and phosphatases.
Cleavable linkage groups, such as disulfide linkages, may be pH sensitive. The pH of human serum was 7.4, while the average intracellular pH was slightly lower, ranging from about 7.1 to 7.3. Endosomes have a more acidic pH in the range of 5.5-6.0, and lysosomes have an even more acidic pH of about 5.0. Some linkers will have cleavable linking groups that cleave at a selected pH, thereby releasing the cationic lipid from the ligand within the cell, or into a desired compartment of the cell.
The linker may comprise a cleavable linking group cleavable by a specific enzyme. The type of cleavable linking group incorporated into the linker may depend on the cell targeted. For example, the liver targeting ligand may be linked to the cationic lipid through a linker comprising an ester group. Hepatocytes are rich in esterases and thus the linker will cleave more efficiently in hepatocytes than in non-esterase-rich cell types. Other esterase-enriched cell types include cells in the lung, kidney cortex and testes.
When targeting peptidase-rich cell types, such as hepatocytes and synovial cells, linkers containing peptide bonds may be used.
In general, the suitability of a candidate cleavable linking group can be assessed by testing the ability of the degrading agent (or condition) to cleave the candidate linking group. It would also be desirable to test candidate cleavable linking groups for their ability to resist cleavage in blood or when in contact with other non-target tissues. Thus, a relative susceptibility to cleavage between a first condition and a second condition may be determined, wherein the first condition is selected to indicate cleavage in a target cell and the second condition is selected to indicate cleavage in other tissue or biological fluid, such as blood or serum. The evaluation can be performed in a cell-free system, in cells, in cell culture, in organ or tissue culture, or in whole animals. Initial evaluation was performed under cell-free or culture conditions and confirmed to be useful by further evaluation in whole animals. In certain embodiments, the cleavage of a useful candidate compound in a cell (or under in vitro conditions selected to mimic intracellular conditions) is at least about 2-fold, 4-fold, 10-fold, 20-fold, 30-fold, 40-fold, 50-fold, 60-fold, 70-fold, 80-fold, 90-fold, or 100-fold faster than the cleavage rate in blood or serum (or under in vitro conditions selected to mimic extracellular conditions).
i. Redox cleavable linking groups
In certain embodiments, the cleavable linking group is a redox cleavable linking group that cleaves upon reduction or oxidation. An example of a reducing cleavable linking group is a disulfide linking group (-S-). To determine whether a candidate cleavable linking group is a suitable "reductive cleavable linking group," or whether it is suitable for use with a particular iRNA moiety and a particular targeting agent, for example, reference may be made to the methods described herein. For example, candidates can be evaluated by incubation with Dithiothreitol (DTT) or other reducing agent using reagents known in the art, which mimic the cleavage rate that would be observed in a cell (e.g., a target cell). Candidates may also be evaluated under conditions selected to mimic blood or serum conditions. In one, the candidate compound is cleaved in the blood up to about 10%. In other embodiments, the degradation of a useful candidate compound in a cell (or under in vitro conditions selected to mimic intracellular conditions) is at least about 2-fold, 4-fold, 10-fold, 20-fold, 30-fold, 40-fold, 50-fold, 60-fold, 70-fold, 80-fold, 90-fold, or about 100-fold greater than the cleavage rate in blood (or under in vitro conditions selected to mimic extracellular conditions). The cleavage rate of the candidate compound can be determined using standard enzymatic kinetic assays under conditions selected to mimic intracellular media and compared to conditions selected to mimic extracellular media.
Phosphate-based cleavable linking groups
In other embodiments, the cleavable linker comprises a phosphate-based cleavable linking group. The phosphate-based cleavable linking group is cleaved by an agent that degrades or hydrolyzes the phosphate group. Examples of agents in cells that cleave phosphate groups are enzymes in cells, such as phosphatases. 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-O (ORk) -O-, O-O (ORk) O-, O-O (O) S-O-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 may be independently at each occurrence C1-C20 alkyl, C1-C20 haloalkyl, C6-C10 aryl, or C7-C12 aralkyl. -S-P (O) (OH) -O- -O-P (O) (OH) -S-, -S-P (O) (OH) -O-, -O-P (O) (OH) -S-, and-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, the phosphate-based linking group is-O-P (O) (OH) -O-. These candidates can be evaluated using methods similar to those described above.
Acid cleavable linking groups
In other embodiments, the cleavable linker comprises an acid-based cleavable linking group. An acid cleavable linking group is a linking group that is cleaved under acidic conditions. In certain embodiments, the acid-cleavable linking group is cleaved in an acidic environment at a pH of about 6.5 or less (e.g., about 6.0, 5.5, 5.0 or less) or by an agent that can act as a generalized acid, such as an enzyme. In cells, specific low pH organelles, such as endosomes and lysosomes, can provide a cleavage environment for acid cleavable linkers. Examples of acid cleavable linking groups include, but are not limited to, hydrazones, esters, and esters of amino acids. The acid cleavable group may have the general formula-c=nn-, C (O) O or-OC (O). Exemplary embodiments are those wherein the carbon (alkoxy) attached to the oxygen of the ester is aryl, substituted alkyl or tertiary alkyl, such as dimethylpentyl or tertiary butyl. These candidates can be evaluated using methods similar to those described above.
Ester-based linking groups
In other embodiments, the cleavable linker comprises an ester-based cleavable linking group. The cleavable ester-based linking group is cleaved by enzymes in the cell, such as esterases and amidases. Examples of ester-based cleavable linking groups include, but are not limited to, esters of alkylene, alkenylene, and alkynylene. The ester cleavable linking group has the general formula-C (O) O-or-OC (O) -. These candidates can be evaluated using methods similar to those described above.
v. peptide-based cleavage groups
In still other embodiments, the cleavable linker comprises a peptide-based cleavable linking group. The peptide-based cleavable linking group is cleaved by enzymes in the cell, such as peptidases and proteases. The peptide-based cleavable linking group is a peptide bond formed between amino acids to produce oligopeptides (e.g., dipeptides, tripeptides, etc.) and polypeptides. The peptide-based cleavable group does not comprise an amide group (-C (O) NH-). The amide groups may be formed between any alkylene, alkenylene or alkynylene groups. Peptide bonds are a special type of amide bond formed between amino acids to produce peptides and proteins. The peptide-based cleavage groups are typically limited to peptide bonds (i.e., amide bonds) formed between the amino acids that produce the peptide and protein and do not contain an entire amide functionality. The peptide-based cleavable linking group has the general formula-NHCHRAC (O) NHCHRBC (O) -, wherein RA and RB are R groups of two adjacent amino acids. These candidates can be evaluated using methods similar to those described above.
In some embodiments, an iRNA of the invention is conjugated to a carbohydrate through a linker. Non-limiting examples of linkers for iRNA carbohydrate conjugates with the compositions and methods of the invention include, but are not limited to:
/>
(formula XLIV) when one of X or Y is an oligonucleotide, the other is hydrogen.
In certain embodiments of the compositions and methods of the present invention, the ligand is one or more "GalNAc" (N-acetylgalactosamine) derivatives linked by a divalent or trivalent branched linker.
In one embodiment, the dsRNA of the invention is conjugated to a divalent or trivalent branched linker selected from the group consisting of structures shown in any one of formulas (XLV) to (XLVI):
/>
wherein:
q2A, q2B, q3A, q3B, q4A, q4B, q5A, q5B and q5C independently represent from 0 to 20 at each occurrence, and wherein the repeating units may be the same or different;
P 2A 、P 2B 、P 3A 、P 3B 、P 4A 、P 4B 、P 5A 、P 5B 、P 5C 、T 2A 、T 2B 、T 3A 、T 3B 、T 4A 、T 4B 、T 4A 、T 5B 、T 5C each occurrence is independently absent, CO, NH, O, S, OC (O), NHC (O), CH 2 、CH 2 NH or CH 2 O;
Q 2A 、Q 2B 、Q 3A 、Q 3B 、Q 4A 、Q 4B 、Q 5A 、Q 5B 、Q 5C Independently at each occurrence is absent, alkylene, substituted alkylene, wherein one or more methylene groups may be interrupted or capped by one or more of the following: o, S, S (O), SO 2 、N(R N ) C (R')=c (R), c≡c, or C (O);
R 2A 、R 2B 、R 3A 、R 3B 、R 4A 、R 4B 、R 5A 、R 5B 、R 5C each occurrence is independently absent, NH, O, S, CH 2 、C(O)O、C(O)NH、NHCH(R a )C(O)、-C(O)-CH(R a )-NH-、CO、CH=N-O、 Or a heterocyclic group;
L 2A 、L 2B 、L 3A 、L 3B 、L 4A 、L 4B 、L 5A 、L 5B and L 5C Represents a ligand; i.e., each occurrence is independently a monosaccharide (e.g., galNAc), disaccharide, trisaccharide, tetrasaccharide, oligosaccharide, or polysaccharide; and R is a Is H or an amino acid side chain. Trivalent conjugated GalThe NAc derivatives are particularly useful in combination with RNAi agents to inhibit expression of a target gene, such as the expression of formula (XLIX):
wherein L is 5A 、L 5B And L 5C Represents a monosaccharide such as GalNAc derivatives.
Examples of suitable divalent and trivalent branched linker groups for conjugation to GalNAc derivatives include, but are not limited to, the structures of formula II, formula VII, formula XI, formula X, and formula XIII mentioned above.
Representative U.S. patents teaching the preparation of RNA conjugates include, but are not limited to, U.S. patent No. 4,828,979; 4,948,882; 5,218,105; 5,525,465; 5,541,313; 5,545,730; 5,552,538; 5,578,717 and 5,580,731; 5,591,584; 5,109,124; 5,118,802; no. 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; no. 5,082,830; 5,112,963; 5,214,136; no. 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 and 5,391,723; 5,416,203 and 5,451,463; 5,510,475; 5,512,667; 5,514,785; 5,565,552; no. 5,567,810; no. 5,574,142; no. 5,585,481; 5,587,371; 5,595,726; no. 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 by reference.
It is not necessary to modify all positions in a given compound uniformly, and in fact, more than one of the modifications described above may be incorporated into a single compound or even at a single nucleoside within an iRNA. The invention also encompasses iRNA compounds as chimeric compounds.
In the context of the present invention, a "chimeric" iRNA compound or "chimera" is an iRNA compound, such as a dsRNAi agent, that contains two or more chemically distinct regions, each region being 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 in which the RNA is modified to confer increased resistance to nuclease degradation, increased cellular uptake, or increased binding affinity to a target nucleic acid. The additional region of the iRNA can serve as a substrate for an enzyme capable of cleaving RNA: DNA or RNA: RNA hybrids. For example, RNase H is a cellular endonuclease that cleaves RNA strands of RNA: DNA duplex. Thus, activation of RNase H cleaves the RNA target, thereby greatly enhancing the efficiency of iRNA repressor gene expression. Thus, comparable results are generally obtained with shorter irnas when chimeric dsRNA is used, as compared to phosphorothioate deoxydsrna hybridized to the same target region. Cleavage of the RNA target can be routinely detected by gel electrophoresis and, if necessary, related nucleic acid hybridization techniques known in the art.
In some cases, the RNA of the iRNA can be modified with a non-ligand group. Many non-ligand molecules have been conjugated to iRNA to enhance the activity, cellular distribution or cellular uptake of iRNA, and procedures for performing such conjugation 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.)," biochem. Comm.), 2007,365 (1): 54-61; letsinger et al, proc. Natl. Acad. Sci. USA, 1989, 86:6553), cholic acid (Manoharan et al, quick report of bioorganic and pharmaceutical chemistry, 1994, 4:1053), thioethers, for example hexyl-S-triphenylmethyl mercaptan (Manoharan et al, new York science annual. 1992,660:306; manoharan et al, quick report of bioorganic and pharmaceutical chemistry, 1993, 3:2765), thiocholesterol (Obohauser et al, 1992, 20:533), aliphatic chains, for example dodecanediol or undecyl residues (Saison-Haras et al, J. European molecular biological tissue, 1991,10:111; kanov et al, BS quick report, 1990,259:327, svihuk et al, biochemistry et al, 1993, 75:49), phospholipids, such as di-hexadecyl-rac-glycerol or triethylammonium 1, 2-di-O-hexadecyl-rac-glycerol-3-H-phosphonate (Manoharan et al, 1995,36:3651; shea et al, nucleic acids research, 1990, 18:3777), phospholipids, polyamines or polyethylene glycol chains (Manoharan et al, nucleosides and nucleotides, 1995, 14:969) or adamantaneacetic acid (Manoharan et al, tetrahedral flash, 1995, 36:3651), palmityl moieties (Mishra et al, biochemistry and biophysics flash, 1995, 1264:229), or octadecylamine or hexylamino-carbonyl-oxo-cholesterol moieties (Croo et al, journal of pharmacology and experimental therapeutics, 1996,277:923). Representative U.S. patents teaching the preparation of such RNA conjugates are listed above. Typical conjugation protocols involve the synthesis of RNAs with amino linkers at one or more positions in the sequence. The amino group is then reacted with the conjugated molecule using an appropriate coupling or activating reagent. The conjugation reaction may be carried out with the RNA still bound to the solid support or in the solution phase after cleavage of the RNA. Purification of the RNA conjugate by HPLC generally yields the pure conjugate.
Delivery of iRNA of the present invention
The delivery of an iRNA of the invention to a cell, e.g., a cell in a subject, e.g., a human subject (e.g., a subject in need thereof, e.g., a subject susceptible to or diagnosed with a TMPRSS 6-related disorder, e.g., a disorder associated with iron overload and/or a disorder of ineffective erythropoiesis) can be accomplished in a number of different ways. For example, delivery can be by contacting a cell with an iRNA of the invention in vitro or in vivo. In vivo delivery may also be directly performed by administering a composition (e.g., dsRNA) comprising iRNA to a subject. Alternatively, in vivo delivery may be performed indirectly by administering one or more vectors encoding and directing 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) may be suitable for use with the iRNA of the present invention (see, e.g., akhtar S. And Julian RL. (1992) [ Trends in cell. Biol.) ] 2 (5): 139-144 and WO94/02595, which are incorporated herein by reference in their entirety). For in vivo delivery, factors that need to be considered for delivery of the iRNA molecule include, for example, biostability of the delivered molecule, prevention of non-specific effects, and accumulation of the delivered molecule in the target tissue. RNA interference has also been shown to be successful in local delivery to the CNS by direct injection (Dorn, G. Et al, (2004) Nucleic Acids (Nucleic Acids) 32:49; tan, PH. Et al (2005) Gene therapy (Gene Ther.) 12:59-66; makimura, H. Et al (2002) BMC Neuroscience (BMC Neuroscience.)) 3:18; shishkina, GT. Et al (2004) Neuroscience (Neuroscience) 129:521-528; thak, ER. Et al (2004) national academy of sciences 101:17270-17275; akaneya, Y. Et al (2005) neurophysiological journal (J. Neurohysis.)) 93:594-602). Modification of the RNA or drug carrier can also allow the iRNA to target the target tissue and avoid undesirable off-target effects. The iRNA molecule can be modified by chemical conjugation to a lipophilic group (e.g., cholesterol) to enhance cellular uptake and prevent degradation. For example, iRNA directed against ApoB conjugated to a lipophilic cholesterol moiety is injected systemically into mice and results in knockdown of ApoB mRNA in both liver and jejunum (Soutschek, j. Et al (2004) Nature 432:173-178).
In alternative embodiments, the iRNA can be delivered using a drug delivery system (e.g., nanoparticle, dendrimer, polymer, liposome, or cationic delivery system). The positively charged cation delivery system promotes binding of (negatively charged) iRNA molecules and also enhances interactions at negatively charged cell membranes to allow efficient uptake of iRNA by cells. The cationic lipid, dendrimer, or polymer can bind to or be induced to form vesicles or micelles that encapsulate the iRNA (see, e.g., kimsh et al (2008) journal of controlled release (Journal of Controlled Release) 129 (2): 107-116). When administered systemically, the formation of vesicles or micelles further prevents degradation of the iRNA. Methods for preparing and administering cationic iRNA complexes are well within the capabilities of those skilled in the art (see, e.g., sorensen, DR et al (2003) [ journal of molecular biology (J. Mol. Biol) [ 327:761-766 ]; verma, UN et al (2003) [ clinical cancer research (Clin. Cancer Res.)) ] [ 9:1291-1300 ], arnold, AS et al (2007) [ journal of neurophysiology (J. Hypertens.) ] 25:197-205, which is incorporated herein by reference in its entirety). Some non-limiting examples of drug delivery systems that can be used for systemic delivery of iRNA include DOTAP (Sorensen, DR. et al (2003), supra; verma, UN et al (2003), supra), "solid nucleic acid lipid particles (solid nucleic acid lipid particles)" (Zimmermann, TS et al (2006), "Nature 441:111-114), cardiolipin (Chien, PY et al (2005)," Cancer Gene therapy (Cancer Gene Ther.)) (12:321-328; pal, A et al (2005), "International journal of oncology (Int J. Oncol.)) (26:1087-1091), polyethylenimine (Bonnet ME et al (2008)," pharmaceutical research (pharm. Res.) (8 months 16. Month-day-before-printing electronic publications; aigner, A.) (2006) biomedical and biotechnology journal (J. Biohnol.) (71659), peptide (Liu, S.) (2006) molecular medicine (mol.) (3:487-1091), polyethyleneimine (Bonnet ME et al (2008), "pharmaceutical research (Pharm. Res.) (1994) and Biotechnology journal (J. Biohnol.)) (35:35-35, et al.) (35:35, yu.) (35:. In some embodiments, the iRNA forms a complex with cyclodextrin for systemic administration. Methods of administration, as well as pharmaceutical compositions of iRNA and cyclodextrin, can be found in U.S. patent No. 7,427,605, which is incorporated herein by reference in its entirety.
A. Vector-encoded iRNA of the invention
The iRNA targeting the TMPRSS6 gene can be expressed from transcriptional units inserted into DNA or RNA vectors (see, e.g., couture, A et al, (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 may be transient (on the order of hours to weeks) or continuous (weeks to months or longer), depending on the particular construct and target tissue or cell type used. These transgenes may be introduced as linear constructs, circular plasmids, or viral vectors, which may be integrating or non-integrating vectors. Transgenes may also be constructed to allow them to be inherited as extrachromosomal plasmids (Gassmann et al, proc. Natl. Acad. Sci. USA (1995) 92:1292).
Viral vector systems that may be used with the methods and compositions described herein include, but are not limited to, (a) adenoviral vectors; (b) Retroviral vectors, including but not limited to lentiviral vectors, moronella leukemia virus (moloney murine leukemia virus), and the like; (c) an adeno-associated viral vector; (d) a herpes simplex virus vector; (e) SV 40 vector; (f) polyomavirus vectors; (g) papillomavirus vectors; (h) a picornaviral vector; (i) Poxvirus vectors such as orthopoxes (e.g., vaccinia virus vectors) or fowlpox (e.g., canary pox or fowlpox); and (j) helper-dependent or entero-free adenoviruses. Replication-defective viruses may also be advantageous. The different vectors will or will not be incorporated into the genome of the cell. If desired, the construct may comprise viral sequences for transfection. Alternatively, the construct may be incorporated into vectors capable of episomal replication, such as EPV and EBV vectors. Constructs for recombinant expression of iRNA will typically require regulatory elements, such as promoters, enhancers, and the like, to ensure expression of the iRNA in the target cell. Other aspects to be considered for vectors and constructs are known in the art.
V. pharmaceutical composition of the invention
The invention also encompasses pharmaceutical compositions and formulations comprising the iRNA of the invention. In one embodiment, provided herein are pharmaceutical compositions comprising an iRNA as described herein and a pharmaceutically acceptable carrier. Pharmaceutical compositions containing iRNA are useful for preventing or treating TMPRSS6 related disorders, such as disorders associated with iron overload and/or disorders of ineffective erythropoiesis. Such pharmaceutical compositions are formulated based on the mode of delivery. One example is the formulation of a composition for systemic administration by parenteral delivery, such as by Subcutaneous (SC), intramuscular (IM), or Intravenous (IV) delivery. The pharmaceutical compositions of the invention may be administered in a dose sufficient to inhibit expression of the TMPRSS6 gene.
In some embodiments, the pharmaceutical compositions of the present invention are sterile. In another embodiment, the pharmaceutical composition of the invention is pyrogen-free.
The pharmaceutical compositions of the invention may be administered in a dose sufficient to inhibit expression of the TMPRSS6 gene. Typically, suitable dosages of iRNA of the invention will range from about 0.001 to about 200.0 mg/kg body weight/day of the subject, typically ranging from about 1 to 50 mg/kg body weight/day. Generally, suitable dosages of the iRNA of the present invention will range from about 0.1mg/kg to about 5.0mg/kg, such as about 0.3mg/kg and about 3.0mg/kg. Repeated dose regimens may comprise periodic administration of a therapeutic amount of an iRNA, such as once a month, 3 months to 6 months, or once a year. In certain embodiments, the iRNA is administered from about once a month to about once every six months.
After the initial treatment regimen, the treatment may be administered on a less frequent basis. The duration of treatment may be determined based on the severity of the disease.
In other embodiments, a single dose of the pharmaceutical composition may be durable such that the dose is administered at intervals of no more than 1 month, 2 months, 3 months, or 4 months. In some embodiments of the invention, a single dose of the pharmaceutical composition of the invention is administered about once a month. In other embodiments of the invention, a single dose of the pharmaceutical composition of the invention is administered quarterly (i.e., about every three months). In other embodiments of the invention, a single dose of the pharmaceutical composition of the invention is administered twice a year (i.e., about once every six months).
Those of skill in the art will appreciate that certain factors may affect the dosage and timing required to effectively treat a subject, including, but not limited to, mutations present in the subject, previous treatments, the overall health or age of the subject, and other diseases present. Furthermore, treating a subject with a prophylactically or therapeutically effective amount of the composition (as the case may be) may comprise a single treatment or a series of treatments.
iRNA can be delivered in a manner that targets a specific tissue (e.g., hepatocytes).
Pharmaceutical compositions of the invention include, but are not limited to, solutions, emulsions, and liposome-containing formulations. These compositions may be produced from a variety of components including, but not limited to, preformed liquids, self-emulsifying solids, and self-emulsifying semisolids. The formulation comprises a liver-targeting formulation.
The pharmaceutical formulations of the present invention may conveniently be presented in unit dosage form and may be prepared according to conventional techniques well known in the pharmaceutical industry. Such techniques include the step of associating the active ingredient with a pharmaceutical carrier or excipient. In general, formulations are prepared by uniformly and intimately bringing into association the active ingredient with liquid carriers.
A. Additional formulations
i. Emulsion
The compositions of the present invention may be prepared and formulated as emulsions. Emulsions are generally heterogeneous systems in which one liquid is dispersed in another liquid in the form of droplets, typically exceeding 0.1 μm in diameter (see, e.g., ansel's Pharmaceutical Dosage Forms and Drug Delivery Systems for pharmaceutical dosage forms and drug delivery systems (Ansel's Pharmaceutical Dosage Forms and Drug Delivery Systems), allen, LV., popovich ng. And Ansel HC.,2004, lipsotrichia wilsons Wilkins publication (Lippincott Williams & Wilkins, new York, NY) (8 th edition); idson, pharmaceutical dosage form (Pharmaceutical Dosage Forms), lieberman, rieger and Banker (editions), 1988, marcel Dekker, inc., new York, N.Y.), volume 1, page 199, rosoff, pharmaceutical dosage form, lieberman, rieger and Banker (editions), 1988, marcel Dekker, rieger and Banker (editions), volume 1, page 245, block, pharmaceutical dosage form, lieberman, rieger and Banker (editions), 1988, marcel Dekker, volume 2, page 335, higuchi et al, remington pharmaceutical science (Remington's Pharmaceutical Sciences), marcel Puston, p.Pust. Pusing, 1985, page 301. Emulsions are typically two-phase systems comprising two immiscible liquid phases intimately mixed and dispersed with each other. Typically, the emulsion may be of the water-in-oil (w/w) or oil-in-water (w/w) type. When the aqueous phase is finely divided into tiny droplets and dispersed into the bulk oil phase, the resulting composition is referred to as a water-in-oil (w/o) emulsion. Alternatively, when the oil phase is finely divided into tiny droplets and dispersed into the bulk aqueous phase, the resulting composition is referred to as an oil-in-water (o/w) emulsion. In addition to the dispersed phase and the active agent, the emulsion may contain additional components, which may be present in the aqueous phase, in the oil phase or as a separate phase itself, in the form of a solution. Pharmaceutical excipients, such as emulsifiers, stabilizers, dyes and antioxidants, may also be present in the emulsion as desired. The pharmaceutical emulsion may also be a variety of emulsions comprising more than two phases, for example in the case of oil-in-water (o/w/o) and water-in-oil-in-water (w/o/w) emulsions. Such complex formulations generally provide certain advantages over simple binary emulsions. Multiple emulsions in which the individual oil droplets of the o/w emulsion surround the water droplets constitute the w/o/w emulsion. Similarly, an oil droplet system surrounded by stable water droplets in an oily continuous phase provides an o/w/o emulsion.
Emulsions are characterized by little or no thermodynamic stability. Typically, the dispersed or discontinuous phase of the emulsion disperses well into the external or continuous phase and this form is maintained by the viscosity of the emulsifier or formulation. Other methods of stabilizing emulsions require the use of emulsifiers that can be incorporated into either phase of the emulsion. Emulsifiers can be broadly divided into four categories: synthetic surfactants, naturally occurring emulsifiers, absorbing bases, and finely divided solids (see, e.g., ansair's pharmaceutical dosage form and drug delivery System, allen, LV., popovich NG. And Ansel HC.,2004, liPindect Williams Wills publishing company (8 th edition), idson, lieberman, rieger and Banker, editions), 1988, masailder, volume 1, page 199, new York).
Synthetic surfactants (also known as surfactants) have found wide applicability in emulsion formulations and have been reviewed in the literature (see, e.g., ansel's pharmaceutical dosage form and drug delivery system, allen, LV., popovich ng. And Ansel HC.,2004, litopent wilcox wilkins publication company (8 th edition) in new york, new york; rieger, pharmaceutical dosage form, lieberman, rieger and Banker (edit), 1988, majordson, lieberman, rieger and Banker (edit), 1988, page 199, new york). Surfactants are generally amphiphilic and include hydrophilic and hydrophobic portions. The ratio of hydrophilicity to hydrophobicity of a surfactant is known as the hydrophilic/lipophilic balance (HLB) and is a valuable tool for classifying and selecting surfactants in the preparation of formulations. Surfactants can be classified into different categories based on the nature of the hydrophilic group: nonionic, anionic, cationic and amphoteric (see, e.g., allen, LV., popovich ng. And Ansel HC.,2004, lepidopter willust wilkins publication (8 th edition), rieger, pharmaceutical dosage form, lieberman, rieger and Banker, 1988, volume 1, page 285, new york).
A variety of non-emulsifying materials are also included in the emulsion formulation and contribute to the characteristics of the emulsion. These include fats, oils, waxes, fatty acids, fatty alcohols, fatty acid esters, humectants, hydrocolloids, preservatives and antioxidants (Block, pharmaceutical dosage form, lieberman, rieger and Banker (eds.), 1988, marseidel, new York, vol.1, page 335; idson, pharmaceutical dosage form, lieberman, rieger and Banker (eds.), 1988, marseidel, new York, vol.1, page 199).
The use of emulsion formulations through the skin, oral and parenteral routes, and methods of making them, have been reviewed in the literature (see, e.g., ansel's pharmaceutical dosage form and drug delivery system, allen, LV., popovich ng. And Ansel HC.,2004, litscott, willi wilkins publication company (8 th edition), idson, pharmaceutical dosage form, lieberman, rieger and Banker, 1988, majelddel company, volume 1, page 199, new york).
Microemulsion(s)
In one embodiment of the invention, the composition of iRNA and nucleic acid is formulated as a microemulsion. Microemulsions may be defined as systems of water, oil and amphiphiles which are single optically isotropic and thermodynamically stable liquid solutions (see, e.g., allen, LV., popovich ng. And anse HC.,2004, lipping willust wilkins publishing company (8 th edition), rosoff, pharmaceutical dosage form, lieberman, rieger and Banker, 1988, makinder, volume 1, page 245, new york). Generally, microemulsions are systems prepared by first dispersing the oil in an aqueous surfactant solution and then adding a sufficient amount of a fourth component (typically a medium chain length alcohol) to form a transparent system. Microemulsions are therefore also described as thermodynamically stable, isotropically transparent dispersions of two immiscible liquids stabilized by interfacial films of surface active molecules (Leung and Shah, controlled release of drugs: polymer and aggregation System (Controlled Release of Drugs: polymers and Aggregate Systems)), rosoff, M.editions, 1989, VCH Publishers, new York, pages 185-215.
Micro-particles
The iRNA of the invention can be incorporated into particles, such as microparticles. Microparticles may 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 permeation enhancer
In one embodiment, the present invention uses various permeation enhancers to achieve efficient delivery of nucleic acids, particularly iRNA, to the skin of an animal. Most drugs exist in solution in both ionized and non-ionized forms. However, only lipid-soluble or lipophilic drugs are generally easy to cross cell membranes. It has been found that even non-lipophilic drugs can cross cell membranes if the membrane to be crossed is treated with a permeation enhancer. In addition to facilitating diffusion of the non-lipophilic drug across the cell membrane, the permeation enhancer may also enhance the permeability of the lipophilic drug.
Permeation enhancers can be divided into one of five major classes, namely surfactants, fatty acids, bile salts, chelating agents, and non-chelating non-surfactants (see, e.g., malmsten, m. surfactants and polymers in drug delivery (Surfactants and polymers in drug delivery), infroma medical health corporation of New York, NY, 2002; lee et al, critical reviews of therapeutic drug carrier systems (Critical Reviews in Therapeutic Drug Carrier Systems), 1991, page 92). Each of the above classes of permeation enhancers and their use in the manufacture of pharmaceutical compositions and delivery agents are well known in the art.
v. excipient
In contrast to carrier compounds, "pharmaceutical carriers" or "excipients" are pharmaceutically acceptable solvents, suspending agents, or any other pharmacologically inert vehicle for delivering one or more nucleic acids to an animal. Excipients may be liquid or solid and are selected in consideration of the intended mode of administration so as to provide the desired volume, consistency, etc. when combined with the nucleic acid and other components of a given pharmaceutical composition. Such agents are well known in the art.
Other components
The compositions of the present invention may additionally contain other accessory components conventionally present in pharmaceutical compositions at the level of use established in the art. Thus, for example, the compositions may contain additional compatible pharmaceutically active materials, e.g., antipruritics, astringents, local anesthetics, or anti-inflammatory agents, or may contain additional materials such as dyes, flavors, preservatives, antioxidants, opacifying agents, thickening agents, and stabilizers useful in physically formulating the various dosage forms of the compositions of the present invention. However, such materials, when added, should not unduly interfere with the biological activity of the components of the compositions of the present invention. The formulation may be sterilized and, if desired, mixed with adjuvants which do not deleteriously interact with the nucleic acids of the formulation, such as lubricants, preservatives, stabilizers, wetting agents, emulsifiers, salts for influencing osmotic pressure, buffers, colorants, flavoring or aromatic substances, and the like.
The aqueous suspension may contain substances which increase the viscosity of the suspension, including for example sodium carboxymethyl cellulose, sorbitol, or dextran. The suspension may also contain stabilizers.
In some embodiments, the pharmaceutical compositions of the invention feature (a) one or more irnas and (b) one or more agents that act through a non-iRNA mechanism and are useful in treating TMPRSS 63-related disorders, e.g., disorders associated with iron overload and/or disorders that do not have erythropoiesis, e.g., hereditary hemochromatosis, β -thalassemia (e.g., severe β -thalassemia and intermediate β -thalassemia), polycythemia vera, myelodysplastic syndrome, congenital erythropoiesis aplastic anemia, pyruvate kinase deficiency, erythropoiesis porphyrin, parkinson's disease, alzheimer's disease, or friedel's ataxia.
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 effect and therapeutic effect is the therapeutic index and the therapeutic index can be expressed as the ratio LD50/ED50. Compounds exhibiting 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 the compositions characterized herein in the present invention is generally in the circulating concentration range comprising the ED50 (e.g., ED80 or ED 90) with little or no toxicity. The dosage may vary within this range depending upon the dosage form employed and the route of administration utilized. For any compound used in the methods of the invention, a prophylactically effective dose may be estimated initially from a cell culture assay. Dosages may be formulated in animal models to achieve a circulating plasma concentration range of the compound, or where appropriate, of the polypeptide product of the target sequence (e.g., to achieve a reduction in polypeptide concentration), which range comprises an IC50 or higher level of inhibition (i.e., the concentration of the test compound that achieves half the maximum inhibition of symptoms) as determined in cell culture. Such information can be used to more accurately determine useful doses in the human body. The level in the plasma may be measured, for example, by high performance liquid chromatography.
In addition to their administration, as discussed above, the irnas featured by the invention can be administered in combination with other known drugs for preventing or treating TMPRSS 6-related disorders, e.g., disorders associated with iron overload and/or disorders of ineffective erythropoiesis. In any event, the administering physician can adjust the amount and time of iRNA administration based on the results observed using standard measurements of efficacy known in the art or described herein.
Methods for inhibiting TMPRSS6 expression
The invention also provides methods of inhibiting expression of a TMPRSS6 gene in a cell. The method comprises contacting the cell with an RNAi agent, e.g., a double-stranded RNA agent, in an amount effective to inhibit expression of TMPRSS6 in the cell, thereby inhibiting expression of TMPRSS6 in the cell.
The contacting of the cells with an iRNA, e.g., a double stranded RNA agent, can be performed in vitro or in vivo. Contacting a cell with an iRNA in vivo comprises contacting a cell or group of cells in a subject (e.g., a human subject) with the iRNA. Combinations of in vitro and in vivo methods of contacting cells are also possible. As discussed above, contacting the cells may be direct or indirect. Furthermore, contacting the cells may be achieved by targeting the ligand, including any ligand described herein or known in the art. In some embodiments, the targeting ligand is a carbohydrate moiety, such as GalNAc 3 A ligand, or any other ligand that directs an RNAi agent to a site of interest.
As used herein, the term "inhibit" may be used interchangeably with "reduce," "silence," "down-regulate," "inhibit," and other similar terms, and includes any level of inhibition.
The phrase "inhibiting expression of TMPRSS 6" is intended to mean inhibiting expression of any TMPRSS6 gene (e.g., a mouse TMPRSS6 3 gene, a rat TMPRSS6 gene, a monkey TMPRSS6 gene, or a human TMPRSS6 gene) or a variant or mutant of a TMPRSS6 gene. Thus, in the context of a genetically manipulated cell, group of cells, or organism, the TMPRSS6 gene may be a wild-type TMPRSS6 gene, a mutant TMPRSS6 gene, or a transgenic TMPRSS6 gene.
"inhibiting expression of a TMPRSS6 gene" includes any level of inhibition of a TMPRSS6 gene, e.g., at least partially inhibiting expression of a TMPRSS6 gene. Expression of the TMPRSS6 gene, e.g., TMPRSS6 mRNA levels or TMPRSS6 protein levels, may be assessed based on the level or change in the level of any variable associated with TMPRSS6 gene expression. Expression of TMPRSS6 may also be assessed indirectly based on hepcidin mRNA levels, hepcidin protein levels, or levels of iron in tissues or serum. This level can be assessed in a single cell or a group of cells, including, for example, a sample derived from a subject. It is understood that TMPRSS6 is mainly expressed in the liver.
Inhibition may be assessed by a decrease in the absolute or relative level of one or more variables associated with TMPRSS6 expression compared to a control level. The control level may be any type of control level used in the art, e.g., a pre-dosing baseline level, or a level determined from a similar subject, cell, or sample that has not been treated or treated with a control (e.g., a buffer-only control or an inactive agent control).
In some embodiments of the methods of the invention, expression of the TMPRSS6 gene is inhibited by at least 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90% or 95%, or below the level of detection determined. In some embodiments, expression of the TMPRSS6 gene is inhibited by at least 70%. It will be further appreciated that in certain tissues, e.g., liver, inhibition of TMPRSS6 expression may be desirable, while in other tissues, e.g., brain, not significantly inhibited expression. In some embodiments, the expression level is determined in a suitable species-matched cell line at a concentration of 10nM siRNA using the assay provided in example 2.
In certain embodiments, inhibition of expression in vivo is determined by knocking down a human gene in an AAV-infected mouse expressing the human gene, e.g., expressing the human target gene (i.e., TMPRSS 6), e.g., when administered in a single dose, e.g., at 3mg/kg at the nadir of RNA expression. Knock-down of endogenous gene expression in the model animal system can also be determined, for example, after a single dose of, for example, 3mg/kg is administered at the nadir of RNA expression. Such a system is useful when the nucleic acid sequences of the human gene and the model animal gene are sufficiently close that the human iRNA provides an effective knock-down of the model animal gene. RNA expression in liver was determined using the PCR method provided in example 2.
Inhibition of TMPRSS6 gene expression may be manifested by a reduction in the amount of mRNA expressed by a first cell or group of cells (e.g., such cells may be present in a sample derived from a subject) in which the TMPRSS6 gene is transcribed and has been treated (e.g., by contacting one or more cells with an iRNA of the invention, or by administering an iRNA of the invention to a subject in which the cells are present or have been present), such that expression of the TMPRSS6 gene is inhibited compared to a second cell or group of cells substantially identical to the first cell or group of cells but not so treated (control cells not treated with an iRNA or not treated with an iRNA targeting a gene of interest). In some embodiments, inhibition is assessed by the method provided in example 2, using a 10nM siRNA concentration in a species matched cell line, and expressing mRNA levels in the treated cells as a percentage of mRNA levels in control cells using the following formula:
in other embodiments, inhibition of TMPRSS6 gene expression may be assessed according to a parameter functionally related to TMPRSS6 gene expression, e.g., a decrease in TMPRSS6 protein level in blood or serum from a subject. TMPRSS6 gene silencing can be determined in any cell expressing TMPRSS6, whether endogenous or heterologous from the expression construct, and by any assay known in the art.
Inhibition of expression of TMPRSS6 protein may be manifested by a decrease in the level of TMPRSS6 protein expressed in a cell or group of cells or in a sample of the subject (e.g., protein level in a blood sample derived from the subject). As explained above, to assess mRNA inhibition, inhibition of protein expression levels in a treated cell or group of cells can be similarly expressed as a percentage of protein levels in a control cell or group of cells, or as a change in protein levels in a subject sample (e.g., blood or serum derived therefrom).
Control cells, cell groups, or subject samples that can be used to assess inhibition of expression of TMPRSS6 genes include cells, cell groups, or test sample that have not been contacted with an RNAi agent of the invention. For example, a control cell, group of cells, or subject sample can be derived from an individual subject (e.g., a human or animal subject) prior to treatment of the subject with an RNAi agent or a suitably matched population control.
The level of TMPRSS6 mRNA 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 expression level of TMPRSS6 in the sample is determined by detecting the mRNA of a transcribed polynucleotide, or a portion thereof, e.g., the TMPRSS6 gene. RNA can be extracted from cells using RNA extraction techniques, including, for example, using phenol/guanidine isothiocyanate extraction (RNAzol B; biogenesis), RNeasy TM RNA preparation kitOr PAXgene TM (PreAnalytix in Switzerland) TM ). Typical assay formats utilizing ribonucleic acid hybridization include nuclear run assays, RT-PCR, RNase protection assays, northern blots, in situ hybridization and microarray analysis.
In some embodiments, the expression level of TMPRSS6 is determined using a nucleic acid probe. As used herein, the term "probe" refers to any molecule capable of selectively binding to a particular TMPRSS 6. Probes may be synthesized by those skilled in the art or derived from appropriate biological agents. Probes may be specifically designed to be labeled. Examples of molecules that can be used as probes include, but are not limited to, RNA, DNA, proteins, antibodies, and organic molecules.
The isolated mRNA can be used in hybridization or amplification assays, including but not limited toIn Southern or northern analysis, polymerase Chain Reaction (PCR) analysis and probe arrays. One method for determining mRNA levels involves contacting the isolated mRNA with a nucleic acid molecule (probe) that can hybridize to TMPRSS6 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 is immobilized on a solid surface and the mRNA is contacted with the probe, e.g., in In a gene chip array. Known mRNA detection methods can be readily used by those skilled in the art to determine TMPRSS6 mRNA levels.
Alternative methods for determining the expression level of TMPRSS6 in a sample involve the process of nucleic acid amplification or reverse transcriptase (to prepare cDNA) of e.g.mRNA in the sample, for example by RT-PCR (Mullis, 1987, experimental examples described in 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. Sci. USA, 87:1874-1878), transcription amplification system (Kwoh et al (1989), Q-beta replicase (Lizardi et al (1988), bio/Technology (Bio/Technology) 6:1197), rolling circle replication (Lizard et al, U.S. Pat. No. 5,854,033) or any other method of nucleic acid amplification followed by the use of the well known techniques for detecting molecules. These detection schemes are particularly useful for detecting nucleic acid molecules if such nucleic acid molecules are present in very low amounts. In a particular aspect of the invention, the expression level of TMPRSS6 is determined by quantitative fluorescent RT-PCR (i.e., taqMan TM System) determination. In some embodiments, expression levels are determined in species-matched cell lines by the method provided in example 2, using, for example, 10nM siRNA concentration.
The expression level of TMPRSS6 mRNA may be monitored using membrane blotting (e.g., for hybridization analysis, such as northern, southern, spots, etc.) or microwells, sample tubes, gels, beads, or fibers (or any solid support including bound nucleic acids). See U.S. patent nos. 5,770,722, 5,874,219, 5,744,305, 5,677,195, and 5,445,934, which are incorporated herein by reference. Determination of TMPRSS6 expression levels may also include the use of nucleic acid probes in solution.
In some embodiments, the level of mRNA expression is assessed using a branched DNA (bDNA) assay or real-time PCR (qPCR). The use of these methods is described and illustrated in the examples given herein. In some embodiments, expression levels are determined in species-matched cell lines by the method provided in example 2, using, for example, 10nM siRNA concentration.
TMPRSS6 protein expression levels may be determined using any method known in the art for measuring protein levels. Such methods include, for example, electrophoresis, capillary electrophoresis, high Performance Liquid Chromatography (HPLC), thin Layer Chromatography (TLC), super-diffusion chromatography, fluid or gel precipitation reactions, absorption spectroscopy, colorimetric assays, spectrophotometric assays, flow cytometry, immunodiffusion (single or double), immunoelectrophoresis, western blotting, radioimmunoassay (RIA), enzyme-linked immunosorbent assay (ELISA), immunofluorescent assays, electrochemiluminescent assays, and the like.
In some embodiments, the efficacy of the methods of the invention is assessed by a decrease in TMPRSS6 mRNA or protein levels (e.g., in liver biopsy).
In some embodiments of the methods of the invention, an iRNA is administered to a subject, thereby delivering the iRNA to a specific site within the subject. Inhibition of TMPRSS6 expression may be assessed using measurement of levels or changes in levels of TMPRSS6 mRNA or TMPRSS6 protein in a sample of fluid or tissue derived from a specific site (e.g., liver or blood) in a subject.
As used herein, the term detecting or determining the level of an analyte is understood to be performing a step to determine whether a material (e.g., protein, RNA) is present. As used herein, a method of detecting or determining comprises detecting or determining an analyte level that is lower than the detection level of the method used.
The prevention and treatment methods of the invention
The invention also provides methods of using the iRNA of the invention or compositions containing the iRNA of the invention to inhibit expression of TMPRSS6, thereby preventing or treating a TMPRSS 6-related disorder, such as a disorder associated with iron overload and/or a disorder of ineffective erythropoiesis. In the methods of the invention, the cells can be contacted with the siRNA in vitro or in vivo, i.e., the cells can be in a subject.
The cells suitable for treatment using the methods of the invention may be any cells expressing the TMPRSS6 gene, e.g., liver cells. Cells suitable for use in the methods of the invention may be mammalian cells, such as primate cells (e.g., human cells, including human cells in chimeric non-human animals, or non-human primate cells, such as monkey cells or chimpanzee cells) or non-primate cells. In certain embodiments, the cell is a human cell, such as a human hepatocyte. In the methods of the invention, expression of TMPRSS6 in the cell is inhibited by at least 50, 55, 60, 65, 70, 75, 80, 85, 90, or 95, or to a level below the level of detection determined.
The in vivo methods of the invention can comprise administering to a subject a composition comprising an iRNA, wherein the iRNA comprises a nucleotide sequence that is complementary to at least a portion of an RNA transcript of a TMPRSS6 gene of a mammal to which the RNAi agent is to be administered. The compositions may 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), intranasal, rectal, and topical (including buccal and sublingual) administration. In certain embodiments, the composition is administered by intravenous infusion or injection. In certain embodiments, the composition is administered by subcutaneous injection. In certain embodiments, the composition is administered by intramuscular injection.
In one aspect, the invention also provides a method for inhibiting expression of a TMPRSS6 gene in a mammal. The method comprises administering to the mammal a composition comprising dsRNA targeting a TMPRSS6 gene in a cell of the mammal and maintaining the mammal for a time sufficient to obtain degradation of mRNA transcripts of the TMPRSS6 gene, thereby inhibiting expression of the TMPRSS6 gene in the cell. The reduction in gene expression can be assessed by any method known in the art and by the methods described herein (e.g., in example 2) (e.g., qRT-PCR). The reduction in protein production may be assessed by any method known in the art, such as ELISA. In certain embodiments, a needle liver biopsy is used as a tissue material for monitoring a decrease in TMPRSS6 gene or protein expression. In other embodiments, the blood sample is used as a subject sample for monitoring a decrease in TMPRSS6 protein expression.
The invention further provides methods of treating a subject in need thereof, e.g., a subject diagnosed with a TMPRSS 6-related disorder, such as a disorder associated with iron overload and/or a disorder that is not erythropoietic, e.g., hereditary hemochromatosis, beta-thalassemia (e.g., severe beta-thalassemia and intermediate beta-thalassemia), polycythemia vera, myelodysplastic syndrome, congenital erythropoiesis anemia, pyruvate kinase deficiency, erythropoiesis porphyria, parkinson's disease, alzheimer's disease, or friedel-crafts ataxia. In one embodiment, a subject having a TMPRSS 6-related disorder has hereditary hemochromatosis. In another embodiment, a subject having a TMPRSS 6-related disorder has β -thalassemia. In another embodiment, a subject having a TMPRSS 6-related disorder has polycythemia vera.
The invention further provides a method of prophylaxis in a subject in need thereof. The methods of treatment of the invention comprise administering the iRNA of the invention to a subject, e.g., a subject who would benefit from reduced expression of TMPRSS6, in a prophylactically effective amount of a dsRNA targeting a TMPRSS6 gene or a pharmaceutical composition comprising a dsRNA targeting a TMPRSS6 gene.
In one aspect, the invention provides methods of treating a subject having a disorder that would benefit from reduced expression of TMPRSS6, e.g., a TMPRSS 6-related disorder, such as a disorder associated with iron overload and/or a disorder that is not erythropoietic, e.g., hereditary hemochromatosis, beta-thalassemia (e.g., severe beta-thalassemia and intermediate beta-thalassemia), polycythemia vera, myelodysplastic syndrome, congenital erythropoiesis anemia, pyruvate kinase deficiency, erythropoiesis porphyria, parkinson's disease, alzheimer's disease, or friedel's ataxia. Treatment of a subject who would benefit from a reduction and/or inhibition of TMPRSS6 gene expression includes both therapeutic treatment (e.g., a subject having an elevated iron level) and prophylactic treatment (e.g., a subject not having an elevated iron level or a subject may be at risk of developing an elevated iron level).
The iRNA of the invention can be administered as "free iRNA". The free iRNA is administered in the absence of the pharmaceutical composition. The naked iRNA can be in a suitable buffer solution. The buffer solution may include acetate, citrate, prolamin, carbonate, or phosphate, or any combination thereof. In one embodiment, the buffer solution is Phosphate Buffered Saline (PBS). The pH and osmolarity of the iRNA-containing buffer solution can be adjusted so that it is suitable for administration to a subject.
Alternatively, the iRNA of the invention may be administered as a pharmaceutical composition, such as a dsRNA liposome formulation.
The subject who would benefit from inhibition of TMPRSS6 gene expression is a subject who is susceptible to or diagnosed with a TMPRSS 6-related disorder, such as a disorder associated with iron overload and/or a disorder of ineffective erythropoiesis, for example, hereditary hemochromatosis, beta-thalassemia (e.g., severe beta-thalassemia and intermediate beta-thalassemia), polycythemia vera, myelodysplastic syndrome, congenital erythropoiesis anemia, pyruvate kinase deficiency, erythropoiesis porphyria, parkinson's disease, alzheimer's disease, or friedel-crafts disease. In one embodiment, the method comprises administering a composition as characterized herein such that expression of the target a TMPRSS6 gene is reduced, e.g., about 1, 2, 3, 4, 5, 6, 1-3, or 3-6 months per dose. In certain embodiments, the composition is administered once every 3 months to 6 months.
In one embodiment, the iRNA useful in the methods and compositions characterized herein specifically targets RNA (primary or processed) of a target TMPRSS6 gene. Compositions and methods for inhibiting expression of these genes using iRNA can be prepared and performed as described herein.
Administration of iRNA according to the methods of the invention can prevent or treat TMPRSS 6-related disorders, e.g., disorders associated with iron overload and/or disorders that do not have erythropoiesis, e.g., hereditary hemochromatosis, beta-thalassemia (e.g., severe beta-thalassemia and intermediate beta-thalassemia), polycythemia vera, myelodysplastic syndrome, congenital erythropoiesis anemia, pyruvate kinase deficiency, erythropoiesis porphyrin, parkinson's disease, alzheimer's disease, or friedel's ataxia. A therapeutic amount of iRNA, such as about 0.01mg/kg to about 200mg/kg, can be administered to a subject.
In one embodiment, the iRNA is administered subcutaneously, i.e., by subcutaneous injection. In another embodiment, the iRNA is administered intravenously, i.e., by intravenous injection. One or more injections may be used to deliver a desired dose of iRNA to a subject. The injection may be repeated over a period of time.
The administration may be repeated periodically. In certain embodiments, after an initial treatment regimen, the treatment may be administered on a less frequent basis. Repeated dose regimens may include periodic administration of a therapeutic amount of an iRNA, such as once a month to once a year. In certain embodiments, the iRNA is administered from about once a month to about once every three months, or from about once every three months to about once every six months.
The invention also provides methods and uses of iRNA agents or pharmaceutical compositions thereof for treating subjects that would benefit from a reduction and/or inhibition of TMPRSS6 gene expression, e.g., subjects suffering from a TMPRSS6 related disorder, in combination with other drugs and/or other therapies, e.g., in combination with known drugs and/or known therapies, e.g., those drugs or therapies currently used to treat such disorders.
Thus, in some aspects of the invention, a method comprising a single iRNA agent of the invention further comprises administering one or more additional therapeutic agents to a subject.
For example, in certain embodiments, TMPRSS 6-targeted iRNA is administered in combination with an agent that is useful, for example, in the treatment of conditions associated with iron overload. For example, additional agents suitable for treating subjects who would benefit from reduced TMPRSS6 expression, e.g., subjects with conditions associated with iron overload, may include iron chelators (e.g., deferoxamine), folic acid, blood transfusion, phlebotomy, agents to treat ulcers, agents to increase fetal hemoglobin levels (e.g., hydroxyurea), agents to control infection (e.g., antibiotics and antiviral agents), agents to treat thrombotic status, or stem cell or bone marrow transplantation. Stem cell transplantation may utilize stem cells from the umbilical cord, such as from a relative, e.g., sibling. Exemplary iron chelators include deferoxamine, deferasirox (exjack), deferiprone, vitamin E, wheat germ oil, tocassam (tocopheryl) and opuntia ficus-indica.
The iRNA agent and the additional therapeutic agent and/or treatment may be administered simultaneously and/or in the same combination, e.g., parenterally, or the additional therapeutic agent may be administered as part of a separate composition or at a separate time and/or by another method known in the art or described herein.
VIII kit
In certain aspects, the present disclosure provides kits comprising suitable containers containing a pharmaceutical formulation of an siRNA compound (e.g., a double stranded siRNA compound or an siRNA compound (e.g., a precursor, such as a larger siRNA compound that can be processed into a ssiRNA compound, or DNA encoding an siRNA compound, such as a double stranded siRNA compound or an siRNA compound, or a precursor thereof)).
Such kits comprise one or more dsRNA agents and instructions for use, e.g., instructions for administering a prophylactically or therapeutically effective amount of the dsRNA agent. The dsRNA agent may be in a vial or pre-filled syringe. The kit may optionally further comprise a device for administering the dsRNA agent (e.g., an injection device, such as a pre-filled syringe), or a device for measuring the inhibition of TMPRSS6 (e.g., a device for measuring the inhibition of TMPRSS6 mRNA, TMPRSS6 protein, and/or TMPRSS6 activity). Such devices for measuring inhibition of TMPRSS6 may include devices for obtaining a sample, e.g., a plasma sample, from a subject. The kits of the invention may optionally further comprise means for determining a therapeutically effective amount or a prophylactically effective amount.
In certain embodiments, the individual components of the pharmaceutical formulation may be provided in one container, such as a vial or prefilled 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 the siRNA compound formulation and at least one other for the carrier compound. The kits may be packaged in many different configurations, such as one or more containers in a single box. The different components may be combined, for example, according to instructions provided with the kit. These components may be combined according to the methods described herein, for example, to prepare and administer a pharmaceutical composition. The kit may further comprise a delivery device.
The application is further illustrated by the following examples, which should not be construed as limiting. All references, patents and published patent applications cited in this application are hereby incorporated by reference in their entirety into the informal sequence listing and drawings.
Examples
EXAMPLE 1 iRNA Synthesis
Reagent source
Where the source of the agent is not specifically set forth herein, such agents may be obtained from any molecular biological agent provider, the quality/purity criteria of which are applicable to molecular biology.
SiRNA design
siRNAs targeting the human transmembrane protease serine 6 (TMPRSS 6) gene (human: NCBI refseqID NM-153609.4,NCBI GeneID:164656) were designed using custom R and Python scripts. Human NM-153609.4 REFSEQ mRNA, has a length of 3197 bases.
A detailed list of unmodified TMPRSS6 sense and antisense strand nucleotide sequences is shown in tables 2,4 and 6. A detailed list of modified TMPRSS6 sense and antisense strand nucleotide sequences is shown in tables 3, 5 and 7.
It should be appreciated that throughout the application, duplex names without a decimal are equivalent to duplex names with a decimal having a lot number referencing only duplex. For example, AD-959917 is equivalent to AD-959917.1.
siRNA synthesis
siRNA was designed, synthesized and prepared using methods known in the art.
Briefly, siRNA sequences were synthesized on a 1 μmol scale using a Mermade 192 synthesizer (BioAutomation) having phosphoramidite chemistry on a solid support. The solid support being a controlled pore glassLoaded with a custom GalNAc ligand (3' -GalNAc conjugate), a universal solid support (AM Chemicals (AM chemical company)), or a first nucleotide of interest. Auxiliary synthesis reagents and standard 2-cyanoethyl phosphoramidite monomers (2 ' -deoxy-2 ' -fluoro, 2' -O-methyl, RNA, DNA) are obtained from sameifeier corporation (Thermo Fisher) (Milwaukee, WI), megawatt technology development limited (honene) (China)) or Chemgenes corporation (Chemgenes) (Wilmington, MA, USA). Additional phosphoramidite monomer is purchased from commercial suppliers, prepared internally, or purchased using custom synthesis from various CMOs. Phosphoramidites were prepared at a concentration of 100mM in acetonitrile or 9:1 acetonitrile in DMF and coupled using 5-ethylthio-1H-tetrazole (ETT, 0.25M in acetonitrile) at a reaction time of 400 seconds. Phosphorothioate linkages were generated using a 100mM solution of 3- ((dimethylamino-methylene) amino) -3H-1,2, 4-dithiazole-3-thione (DDTT, obtained from Chemgenes, inc. (Wilmington, mass.) in anhydrous acetonitrile/pyridine (9:1 v/v). The oxidation time was 5 minutes. All sequences were synthesized with final removal of DMT groups (“DMT-Off”)。
After completion of the solid phase synthesis, the solid phase supported oligoribonucleotides were treated with 300 μl of methylamine (40% aqueous solution) in 96-well plates at room temperature for about 2 hours to cleave from the solid phase support and subsequently remove all additional base labile protecting groups. For sequences containing any natural ribonucleotide bond (2' -OH) protected with a tert-butyldimethylsilyl (TBDMS) group, the second deprotection step was performed using tea.3hf (triethylamine trihydrofluoride). 200. Mu.L of dimethyl sulfoxide (DMSO) and 300. Mu.L of TEA.3HF were added to each oligonucleotide solution in aqueous methylamine solution, and the solution was incubated at 60℃for about 30 minutes. After incubation, the plates were brought to room temperature and the crude oligonucleotides were precipitated by adding 1mL of 9:1 acetonitrile: ethanol or 1:1 ethanol: isopropanol. The plates were then centrifuged at 4 ℃ for 45 minutes and the supernatant carefully decanted with the aid of a multichannel pipette. The oligonucleotide particles were resuspended in 20mM NaOAc and then desalted using a HiTrap size exclusion column (5 mL, general electric medical Co., ltd.) on an Agilent LC system equipped with an autosampler, UV detector, conductivity meter and fraction collector. Desalted samples were collected in 96-well plates and then analyzed by LC-MS and UV spectroscopy to confirm identity and quantification of the materials, respectively.
Single-stranded double-sided folding was performed on a Tecan liquid handling robot. In 96-well plates, sense and antisense single strands were combined at equimolar ratio to a final concentration of 10 μm in 1x PBS, the plates were sealed, incubated at 100 ℃ for 10 minutes, and then allowed to slowly recover to room temperature over 2 to 3 hours. The concentration and identity of each duplex was confirmed and then used in an in vitro screening assay.
Example 2 in vitro screening methods
Cell culture and 384 well transfection
For transfection, hep3b cells (American type culture Collection (ATCC, manassas, va.) of Marassas, virginia) were 5% CO at 37 ℃C 2 Grown to near confluence in Eagle minimum essential medium (Ji Boke company (Gibco)) supplemented with 10% FBS (ATCC),and then released from the plate by trypsin digestion. Transfection was performed by adding 7.5 μl of Opti-MEM to 0.1 μl of Lipofectamine RNAiMax per well (Invitrogen, carlsbad ca, catalog No. 13778-150) to 2.5 μl of each siRNA duplex in a single well in 384 well plates. The mixture was then incubated at room temperature for 15 minutes. Forty. Mu.l of antibiotic-free containing approximately 1.5X10 4 Complete growth medium for individual cells was added to the siRNA mixture. Cells were incubated for 24 hours prior to RNA purification. Single dose experiments were performed at final duplex concentrations of 10nM, 1nM and 0.1 nM.
TM Total RNA isolation Using DYNABEADS mRNA isolation kit (Invitrogen, part # 610-12)
Cells were lysed in 75 μl lysis/binding buffer containing 3 μl of beads per well and mixed for 10 min on an electrostatic oscillator. The washing step was automated on a Biotek EL406 using a magnetic plate support. The beads were washed once in buffer a (in 90 μl), once in buffer B, and twice in buffer E with a pumping step in between. After final aspiration, the entire 10 μl RT mix was added to each well, as described below.
Using ABI high capacity cDNA reverse transcription kit (applied biosystems, inc. of foster, calif.) (Applied Biosystems, foster City, calif.), catalog number 4368813) for cDNA Synthesis
Mu.l 10 Xbuffer, 0.4. Mu.l 25 XdNTP, 1. Mu.l random primer, 0.5. Mu.l reverse transcriptase, 0.5. Mu.l RNase inhibitor and 6.6. Mu.l H were added per well per reaction 2 A master mix of O. The plates were sealed, stirred on an electrostatic oscillator for 10 minutes, and then incubated at 37℃for 2 hours. After this, the plate was stirred at 80 ℃ for 8 minutes.
Real-time PCR
Two microliters (μl) of cDNA was added to a master mix of 384 well plates (Roche, cat# 04887301001) containing 0.5 μl of human GAPDH TaqMan probe (4326317E), 0.5 μl of human TMPRSS6, 2 μl of nuclease free water, and 5 μl of Lightcycle 480 probe master mix (Roche, cat# 04887301001) per well. Real-time PCR was performed in the LightCycler480 real-time PCR system (Roche).
To calculate the relative fold change, the data were analyzed using the ΔΔct method and normalized to the assay performed with 10nM AD-1955 transfected cells or mock transfected cells. Calculation of IC using 4-parameter fitting model of XLFit 50 And normalized to cells transfected or mock transfected with AD-1955. The sense and antisense sequences of AD-1955 are: sense: cuAcGcuGAGcuUCCGAdTsdT (SEQ ID NO: 18) and antisense UCGAAGuACUAGCGuAAGdTsdT (SEQ ID NO: 19).
The single dose screening results of the agents in tables 2, 3, 6 and 7 in Hep3b cells are shown in table 8.
Table 1: abbreviations for nucleotide monomers used for nucleic acid sequence representation. It will be appreciated that when these monomers are present in the oligonucleotide, they are linked to each other by a 5'-3' -phosphodiester linkage; and it will be appreciated that when the nucleotide contains a 2' -fluoro modification then fluoro replaces the hydroxy group at that position in the parent nucleotide (i.e. it is a 2' -deoxy-2 ' -fluoro nucleotide).
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
Table 8: single dose screening in Hep3b cells
/>
/>
/>
/>
/>
/>
/>
Example 3: efficacy of dsRNA duplex in non-human primate (NHP)
Selected duplexes of interest identified from the in vitro studies described above were evaluated in non-human primate animals. Fig. 1 provides a depiction of a study design.
In particular, 15 male cynomolgus monkeys were divided into 5 groups of 3, and AD-1556360 at a single 3mg/kg dose, AD-1556360 at a single 10mg/kg dose, AD-1571158 at a single 3mg/kg dose, or AD-1571033 at a single 3mg/kg dose or PBS were subcutaneously administered as controls (see table 9). For each animal, two liver biopsy samples (one per leaf) of approximately 100mg each were collected after 12 hours of fasting on day 22, day 57, and/or day 85. Liver biopsies and serum samples were also collected from animals 21 days prior to dosing. From day 1, one mL of blood was collected weekly into an anticoagulant-free tube for hepcidin level, iron level, transferrin saturation level and Red Blood Cell (RBC) count determination. After coagulation, serum was aliquoted and stored at-80 ℃.
Tissue mRNA was extracted and analyzed by RT-QPCR. TMPRSS6 mRNA levels were compared to the levels of housekeeping gene GAPDH. These values were then normalized to the average value of the PBS vehicle control group. Data are expressed as a percentage of baseline values and as mean plus standard deviation.
Iron and transferrin saturation levels were determined using a commercially available kit from roche company.
The results shown in fig. 2-4 demonstrate that all three exemplary duplexes, AD-1556360, AD-1571158 and AD-1571033, effectively and permanently inhibited TMPRSS6 messenger RNA expression in vivo (fig. 2), effectively and permanently reduced plasma iron levels (fig. 3), and effectively and permanently reduced transferrin saturation levels (fig. 4). Transferrin saturation is a measure of the amount of iron bound to serum transferrin and corresponds to the ratio of serum iron to total iron binding capacity.
Table 9: treatment group
Group number Duplex body Dosage level (mg/kg) Number of males
1 PBS (control) 0 3
2 AD-1556360 3 3
3 AD-1556360 10 3
4 AD-1571158 3 3
5 AD-1571033 (reference comparator duplex) 3 3
Equivalent forms
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 (135)

1. A double-stranded ribonucleic acid (dsRNA) agent for inhibiting expression of a transmembrane protease serine 6 (TMPRSS 6) in a cell, wherein the dsRNA agent comprises a sense strand and an antisense strand that form a double-stranded region, wherein the antisense strand comprises a region that is complementary to an mRNA encoding TMPRSS6, and wherein the complementary region comprises at least 15 contiguous nucleotides that differ by no more than 3 nucleotides from any one of the antisense nucleotide sequences in any one of tables 2-7.
2. The dsRNA agent of claim 1, wherein the dsRNA agent comprises a sense strand comprising at least 15 contiguous nucleotides differing by no more than three nucleotides from any one of the nucleotide sequences of the sense strand in any one of tables 2-7 and an antisense strand comprising at least 15 contiguous nucleotides differing by no more than three nucleotides from any one of the nucleotide sequences of the antisense strand in any one of tables 2-7.
3. The dsRNA agent of claim 1, wherein the dsRNA agent comprises a sense strand comprising at least 15 consecutive nucleotides differing by no more than two nucleotides from any one of the nucleotide sequences of the sense strand in any one of tables 2-7 and an antisense strand comprising at least 15 consecutive nucleotides differing by no more than two nucleotides from any one of the nucleotide sequences of the antisense strand in any one of tables 2-7.
4. The dsRNA agent of claim 1, wherein the dsRNA agent comprises a sense strand comprising at least 15 contiguous nucleotides differing by no more than one nucleotide from any one of the nucleotide sequences of the sense strand in any one of tables 2-7 and an antisense strand comprising at least 15 contiguous nucleotides differing by no more than one nucleotide from any one of the nucleotide sequences of the antisense strand in any one of tables 2-7.
5. The dsRNA agent of claim 1, wherein the dsRNA agent comprises a sense strand comprising a nucleotide sequence selected from the group consisting of any one of the nucleotide sequences of the sense strand in any one of tables 2-7 and an antisense strand comprising a nucleotide sequence selected from the group consisting of any one of the nucleotide sequences of the antisense strand in any one of tables 2-7.
6. A double-stranded ribonucleic acid (dsRNA) agent for inhibiting expression of a transmembrane protease serine 6 (TMPRSS 6) 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 consecutive nucleotides differing by NO more than three nucleotides from any one of the nucleotide sequences of the following nucleotides of SEQ ID NO: 1: 187-210;227-254;322-363;362-390;398-420;404-429;410-435;439-461;443-467;448-474;460-483;466-488;496-519;519-542;526-548;557-593;641-671;652-676;687-713;725-762;757-794;886-908;921-951;956-987;1051-1082;1233-1269;1279-1313;1313-1341;1327-1351;1415-1439;1447-1480;1464-1486;1486-1509;1559-1589;1571-1595;1579-1609;1707-1735;1738-1764;1806-1828;1864-1886;1934-1966;1967-1991;2008-2031;2015-2043;2042-2072;2287-2311;2297-2354;2336-2361;2360-2384;2416-2438;2481-2510;2496-2527;2526-2558;2665-2693;2693-2719;2707-2729;2799-2821;2851-2874;2971-2999;2981-3006; and 3155-3195, and the antisense strand comprises at least 15 consecutive nucleotides from the corresponding nucleotide sequence of SEQ ID NO. 2.
7. A double-stranded ribonucleic acid (dsRNA) agent for inhibiting expression of a transmembrane protease serine 6 (TMPRSS 6) 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 consecutive nucleotides differing by NO more than three nucleotides from any one of the nucleotide sequences of the following nucleotides of SEQ ID NO: 1: 230-252, 324-346, 560-578, 560-582, 2338-2360, 3163-3185, 3169-3191 and 3172-3194, and the antisense strand comprises at least 15 consecutive nucleotides of the corresponding nucleotide sequence from SEQ ID No. 2.
8. A double-stranded ribonucleic acid (dsRNA) agent for inhibiting expression of a transmembrane protease serine 6 (TMPRSS 6) 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 three nucleotides from any one of the nucleotide sequences of nucleotides 560-578, 2338-2360, and 3169-3191 of SEQ ID No. 1, and the antisense strand comprises at least 15 contiguous nucleotides from a corresponding nucleotide sequence of SEQ ID No. 2.
9. The dsRNA agent of any one of claims 1 to 8, wherein the antisense strand comprises at least 15 consecutive nucleotides differing by no more than three nucleotides from any one of the antisense strand nucleotide sequences of a duplex selected from the group consisting of: AD-1556360, AD-1571158, AD-1571033, AD-1554875, AD-1571160, AD-1555117, AD-1554911 and AD-1556915.
10. The dsRNA agent of any one of claims 1 to 9, wherein the antisense strand comprises at least 15 consecutive nucleotides differing by no more than three nucleotides from any one of the antisense strand nucleotide sequences of a duplex selected from the group consisting of: AD-1556360, AD-1571158 and AD-1571033.
11. The dsRNA agent of any one of claims 1 to 10, wherein the dsRNA agent comprises at least one modified nucleotide.
12. The dsRNA agent of any one of claims 1 to 11, wherein substantially all nucleotides of the sense strand comprise a modification; substantially all of the nucleotides of the antisense strand comprise modifications; or substantially all nucleotides of the sense strand and substantially all nucleotides of the antisense strand comprise modifications.
13. The dsRNA agent of any one of claims 1 to 12, wherein all nucleotides of the sense strand comprise a modification; all nucleotides of the antisense strand include modifications; or all nucleotides of the sense strand and all nucleotides of the antisense strand comprise modifications.
14. The dsRNA agent of any one of claims 11 to 13, wherein at least one of the modified nucleotides is selected from the group of: deoxynucleotides, 3' -terminal deoxythymine (dT) nucleotides, 2' -O-methyl modified nucleotides, 2' -fluoro modified nucleotides, 2' -deoxymodified nucleotides, 2' -5' -linked ribonucleotides (3 ' -RNA), locked nucleotides, unlocked nucleotides, conformationally restricted nucleotides, constrained ethyl nucleotides, abasic nucleotides, 2' -amino modified nucleotides, 2' -O-allyl modified nucleotides, 2' -C-alkyl modified nucleotides, 2' -methoxyethyl modified nucleotides, 2' -O-alkyl modified nucleotides, morpholino nucleotides, phosphoramidates, non-natural bases including nucleotides, tetrahydropyran modified nucleotides, 1, 5-anhydrohexitol modified nucleotides, cyclohexenyl modified nucleotides, nucleotides including 5' -phosphorothioate groups, nucleotides including 5' -methylphosphonate groups, nucleotides including 5' phosphate or 5' phosphate mimics, nucleotides including vinylphosphonate, ethylene Glycol Nucleic Acid (GNA), ethylene glycol nucleic acid (S-thymine) including 2' -hydroxy-3 ' -deoxyguanylate, and 3' -hydroxy-3 ' -phosphoester derivatives including the hydroxy-2 ' -methyl-guanylate and the 3' -hydroxy-3 ' -phospho-terminal phospho-ester derivatives thereof; and combinations thereof.
15. The dsRNA agent of any one of claims 11 to 13, wherein the modification on the nucleotide is 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' -hydroxy and ethylene glycol; and combinations thereof.
16. The dsRNA agent of any one of claims 11 to 13, wherein at least one of the modified nucleotides is selected from the group consisting of: deoxynucleotides, 2 '-O-methyl modified nucleotides, 2' -fluoro modified nucleotides, 2 '-deoxy modified nucleotides, ethylene glycol modified nucleotides (GNAs), nucleotides including 2' phosphates and vinyl phosphonate nucleotides; and combinations thereof.
17. The dsRNA agent of any one of claims 1 to 16, wherein the double-stranded region is 19-30 nucleotide pairs in length.
18. The dsRNA agent of claim 17, wherein the double-stranded region is 19-25 nucleotide pairs in length.
19. The dsRNA agent of claim 17, wherein the double-stranded region is 19-23 nucleotide pairs in length.
20. The dsRNA agent of claim 17, wherein the double-stranded region is 23-27 nucleotide pairs in length.
21. The dsRNA agent of claim 17, wherein the double-stranded region is 21-23 nucleotide pairs in length.
22. The dsRNA agent of any one of claims 1 to 21, wherein each strand independently is no more than 30 nucleotides in length.
23. The dsRNA agent of any one of claims 1 to 22, wherein the sense strand is 21 nucleotides in length and the antisense strand is 23 nucleotides in length.
24. The dsRNA agent of any one of claims 1 to 23, wherein the complementary region is at least 17 nucleotides in length.
25. The dsRNA agent of any one of claims 1 to 24, wherein the length of the complementary region is between 19 and 23 nucleotides.
26. The dsRNA agent of any one of claims 1 to 25, wherein the complementary region is 19 nucleotides in length.
27. The dsRNA agent of any one of claims 1 to 26, wherein at least one strand comprises a 3' overhang of at least 1 nucleotide.
28. The dsRNA agent of any one of claims 1 to 26, wherein at least one strand comprises a 3' overhang of at least 2 nucleotides.
29. The dsRNA agent of any one of claims 1 to 28, further comprising a ligand.
30. The dsRNA agent of claim 29, wherein the ligand is conjugated to the 3' end of the sense strand of the dsRNA agent.
31. The dsRNA agent of claim 29, wherein the ligand is conjugated to the 5' end of the sense strand of the dsRNA agent.
32. The dsRNA agent of any one of claims 29 to 31, wherein the ligand is an N-acetylgalactosamine (GalNAc) derivative.
33. The dsRNA agent of any one of claims 29 to 32, wherein the ligand is one or more GalNAc derivatives linked by a monovalent, divalent or trivalent branched linker.
34. The dsRNA agent of claim 32 or 33, wherein the ligand is
35. The dsRNA agent of claim 34, wherein the dsRNA agent is conjugated to the ligand as shown in the following schematic diagram
And wherein X is O or S.
36. The dsRNA agent of claim 35, wherein the X is O.
37. The dsRNA agent of claim 34, wherein the dsRNA agent is conjugated to the ligand as shown in the following schematic diagram
38. The dsRNA agent of any one of claims 1 to 37, wherein the dsRNA agent further comprises at least one phosphorothioate or methylphosphonate internucleotide linkage.
39. The dsRNA agent of claim 38, wherein the phosphorothioate or methylphosphonate internucleotide linkage is located at the 3' terminus of one strand.
40. The dsRNA agent of claim 39, wherein the strand is the antisense strand.
41. The dsRNA agent of claim 39, wherein the strand is the sense strand.
42. The dsRNA agent of claim 39, wherein the phosphorothioate or methylphosphonate internucleotide linkage is located at the 5' end of one strand.
43. The dsRNA agent of claim 42, wherein the strand is the antisense strand.
44. The dsRNA agent of claim 42, wherein the strand is the sense strand.
45. The dsRNA agent of claim 38, wherein the phosphorothioate or methylphosphonate internucleotide linkages are located at both the 5 'and 3' ends of one strand.
46. The dsRNA agent of claim 45, wherein the strand is the antisense strand.
47. The dsRNA agent of any one of claims 1 to 46, comprising 6-8 phosphorothioate or methylphosphonate internucleotide linkages.
48. The dsRNA agent of any one of claims 1 to 47, wherein the base pair at position 1 of the 5' end of the antisense strand of the duplex is an AU base pair.
49. The dsRNA agent of any one of claims 1 to 48, wherein the sense strand comprises at least 17 consecutive nucleotides differing by NO more than 3 nucleotides from the nucleotide sequence of 5'-GACGCCACGCAUGCUGUGUGU-3' (SEQ ID NO: 119).
50. The dsRNA agent of any one of claims 1 to 49, wherein the sense strand comprises at least 19 consecutive nucleotides differing by NO more than 3 nucleotides from the nucleotide sequence of 5'-GACGCCACGCAUGCUGUGUGU-3' (SEQ ID NO: 119).
51. The dsRNA agent of any one of claims 1 to 50, wherein the sense strand comprises a nucleotide sequence of 5'-GACGCCACGCAUGCUGUGUGU-3' (SEQ ID NO: 119).
52. The dsRNA agent of any one of claims 1 to 51, wherein the sense strand consists of a nucleotide sequence of 5'-GACGCCACGCAUGCUGUGUGU-3' (SEQ ID NO: 119).
53. The dsRNA agent of any one of claims 1 to 52, wherein the antisense strand comprises at least 17 consecutive nucleotides differing by NO more than 3 nucleotides from the nucleotide sequence of 5'-ACACACAGCAUGCGUGGCGUCAC-3' (SEQ ID NO: 245).
54. The dsRNA agent of any one of claims 1 to 53, wherein the antisense strand comprises at least 19 consecutive nucleotides differing by NO more than 3 nucleotides from the nucleotide sequence of 5'-ACACACAGCAUGCGUGGCGUCAC-3' (SEQ ID NO: 245).
55. The dsRNA agent of any one of claims 1 to 54, wherein the antisense strand comprises at least 21 consecutive nucleotides differing by NO more than 3 nucleotides from the nucleotide sequence of 5'-ACACACAGCAUGCGUGGCGUCAC-3' (SEQ ID NO: 245).
56. The dsRNA agent of any one of claims 1 to 55, wherein the antisense strand comprises a nucleotide sequence of 5'-ACACACAGCAUGCGUGGCGUCAC-3' (SEQ ID NO: 245).
57. The dsRNA agent of any one of claims 1 to 56, wherein the antisense strand consists of a nucleotide sequence of 5'-ACACACAGCAUGCGUGGCGUCAC-3' (SEQ ID NO: 245).
58. The dsRNA agent of any one of claims 1 to 57, wherein the sense strand comprises a nucleotide sequence of 5'-GACGCCACGCAUGCUGUGUGU-3' (SEQ ID NO: 119) and the antisense strand comprises a nucleotide sequence of 5'-ACACACAGCAUGCGUGGCGUCAC-3' (SEQ ID NO: 245).
59. The dsRNA agent of any one of claims 1 to 58, wherein the sense strand differs from the nucleotide sequence of 5 '-gsascgccacgfcfafafugcugugugugu-3' (SEQ ID NO: 371) by NO more than 3 modified nucleotides, wherein a, g, c and U are 2 '-O-methyl (2' -OMe) A, G, C and U, respectively; af. Gf, cf and Uf are 2' -fluoro A, G, C and U, respectively; and s is a phosphorothioate bond.
60. The dsRNA agent of any one of claims 1 to 59, wherein said antisense strand differs from the nucleotide sequence of 5 '-asdcsacdaccaudgcgfuggcgcascasc-3' (SEQ ID NO: 497) by NO more than 3 modified nucleotides, wherein a, g, c and U are 2 '-O-methyl (2' -OMe) A, G, C and U, respectively; af. Gf, cf and Uf are 2' -fluoro A, G, C and U, respectively; wherein dA, dG and dC are 2 '-deoxyadenosine-3' -phosphate, 2 '-deoxyguanosine-3' -phosphate and 2 '-deoxycytidine-3' -phosphate, respectively; and s is a phosphorothioate bond.
61. The dsRNA agent of any one of claims 1 to 60, wherein the sense strand comprises a nucleotide sequence of 5 '-gsascgccgfcafafugcugugugugu-3' (SEQ ID NO: 371) and the antisense strand comprises a nucleotide sequence of 5 '-asdcsacdacacdaccaudgcgcgfuggcgucsasc-3' (SEQ ID NO: 497) wherein a, g, c and U are 2 '-O-methyl (2' -OMe) A, G, C and U, respectively; af. Gf, cf and Uf are 2' -fluoro A, G, C and U, respectively; wherein dA, dG and dC are 2 '-deoxyadenosine-3' -phosphate, 2 '-deoxyguanosine-3' -phosphate and 2 '-deoxycytidine-3' -phosphate, respectively; and s is a phosphorothioate bond.
62. The dsRNA agent of any one of claims 1 to 61, wherein the sense strand comprises a nucleotide sequence of 5 '-gsascgccgfcaffugcugugul 96-3' (SEQ ID NO: 371) and the antisense strand comprises a nucleotide sequence of 5 '-asdcsacdacacgcaadcgcgcgfuggcgcascasc-3' (SEQ ID NO: 497), wherein a, g, c and U are 2 '-O-methyl (2' -OMe) A, G, C and U, respectively; af. Gf, cf and Uf are 2' -fluoro A, G, C and U, respectively; wherein dA, dG and dC are 2 '-deoxyadenosine-3' -phosphate, 2 '-deoxyguanosine-3' -phosphate and 2 '-deoxycytidine-3' -phosphate, respectively; s is a phosphorothioate linkage, and L96 is N- [ tris (GalNAc-alkyl) -amidodecanoyl) ] -4-hydroxyproline.
63. The dsRNA agent of any one of claims 1 to 61, wherein the sense strand comprises a nucleotide sequence of 5 '-gsascgccgfcafafugcugugugugu-3' (SEQ ID NO: 371) and the antisense strand comprises a nucleotide sequence of 5 '-asdcsacdacacdaccaudgcgcgfuggcgucsasc-3' (SEQ ID NO: 497) wherein a, g, c and U are 2 '-O-methyl (2' -OMe) A, G, C and U, respectively; af. Gf, cf and Uf are 2' -fluoro A, G, C and U, respectively; wherein dA, dG and dC are 2 '-deoxyadenosine-3' -phosphate, 2 '-deoxyguanosine-3' -phosphate and 2 '-deoxycytidine-3' -phosphate, respectively; and s is a phosphorothioate linkage, wherein the 3' end of the sense strand is conjugated to the ligand, as shown in the following schematic:
and wherein X is O.
64. The dsRNA agent of any one of claims 1 to 48, wherein the sense strand comprises at least 17 consecutive nucleotides differing by NO more than 3 nucleotides from the nucleotide sequence of 5'-CCUUUGGAAUAAAGCUGCCUU-3' (SEQ ID NO: 844).
65. The dsRNA agent of any one of claims 1 to 48 and 64, wherein the sense strand comprises at least 19 consecutive nucleotides differing by NO more than 3 nucleotides from the nucleotide sequence of 5'-CCUUUGGAAUAAAGCUGCCUU-3' (SEQ ID NO: 844).
66. The dsRNA agent of any one of claims 1 to 48, 64 and 65, wherein the sense strand comprises a nucleotide sequence of 5'-CCUUUGGAAUAAAGCUGCCUU-3' (SEQ ID NO: 844).
67. The dsRNA agent of any one of claims 1 to 48 and 64 to 66, wherein the sense strand consists of a nucleotide sequence of 5'-CCUUUGGAAUAAAGCUGCCUU-3' (SEQ ID NO: 844).
68. The dsRNA agent of any one of claims 1 to 48 and 64 to 67, wherein the antisense strand comprises at least 17 consecutive nucleotides differing by NO more than 3 nucleotides from the nucleotide sequence of 5'-AAGGCAGCUUUAUUCCAAAGGGC-3' (SEQ ID NO: 1868).
69. The dsRNA agent of any one of claims 1 to 48 and 64 to 68, wherein the antisense strand comprises at least 19 consecutive nucleotides differing by NO more than 3 nucleotides from the nucleotide sequence of 5'-AAGGCAGCUUUAUUCCAAAGGGC-3' (SEQ ID NO: 1868).
70. The dsRNA agent of any one of claims 1 to 48 and 64 to 69, wherein the antisense strand comprises at least 21 consecutive nucleotides differing by NO more than 3 nucleotides from the nucleotide sequence of 5'-AAGGCAGCUUUAUUCCAAAGGGC-3' (SEQ ID NO: 1868).
71. The dsRNA agent of any one of claims 1 to 48 and 64 to 70, wherein the antisense strand comprises a nucleotide sequence of 5'-AAGGCAGCUUUAUUCCAAAGGGC-3' (SEQ ID NO: 1868).
72. The dsRNA agent of any one of claims 1 to 48 and 64 to 71, wherein the antisense strand consists of a nucleotide sequence of 5'-AAGGCAGCUUUAUUCCAAAGGGC-3' (SEQ ID NO: 1868).
73. The dsRNA agent of any one of claims 1 to 48 and 64 to 72, wherein the sense strand comprises a nucleotide sequence of 5'-CCUUUGGAAUAAAGCUGCCUU-3' (SEQ ID NO: 844) and the antisense strand comprises a nucleotide sequence of 5'-AAGGCAGCUUUAUUCCAAAGGGC-3' (SEQ ID NO: 1868).
74. The dsRNA agent of any one of claims 1 to 48 and 64 to 73, wherein the sense strand differs from the nucleotide sequence of 5 '-cscsuuggfafafaagcugccuu-3' (SEQ ID NO: 2095) by NO more than 3 modified nucleotides, wherein a, g, c and U are 2 '-O-methyl (2' -OMe) A, G, C and U, respectively; af. Gf, cf and Uf are 2' -fluoro A, G, C and U, respectively; and s is a phosphorothioate bond.
75. The dsRNA agent of any one of claims 1 to 48 and 64 to 74, wherein said antisense strand differs from the nucleotide sequence of 5'-asAfsggdCa (G2 p) cuuuuufccfaaaggsgsc-3' (SEQ ID NO: 2324) by NO more than 3 modified nucleotides, wherein a, G, c and U are 2 '-O-methyl (2' -OMe) A, G, C and U, respectively; af. Gf, cf and Uf are 2' -fluoro A, G, C and U, respectively; wherein G2p is guanosine-2' -phosphate; and s is a phosphorothioate bond.
76. The dsRNA agent of any one of claims 1 to 48 and 64 to 75, wherein the sense strand comprises a nucleotide sequence of 5 '-cscsuuuggfafaagcugccuu-3' (SEQ ID NO: 2095) and the antisense strand comprises a nucleotide sequence of 5'-asAfsggdCa (G2 p) cuuauuuauauaaaggsgscsc-3' (SEQ ID NO: 2324) wherein a, G, c and U are 2 '-O-methyl (2' -OMe) A, G, C and U, respectively; af. Gf, cf and Uf are 2' -fluoro A, G, C and U, respectively; wherein G2p is guanosine-2' -phosphate; and s is a phosphorothioate bond.
77. The dsRNA agent of any one of claims 1 to 48 and 64 to 76, wherein the sense strand comprises a nucleotide sequence of 5 '-cscsuuuggfafaaagcugccuul 96-3' (SEQ ID NO: 2095) and the antisense strand comprises a nucleotide sequence of 5'-asAfsggdCa (G2 p) cuuauuuauaaggsgssc-3' (SEQ ID NO: 2324) wherein a, G, c and U are 2 '-O-methyl (2' -OMe) A, G, C and U, respectively; af. Gf, cf and Uf are 2' -fluoro A, G, C and U, respectively; wherein G2p is guanosine-2' -phosphate; s is a phosphorothioate linkage, and L96 is N- [ tris (GalNAc-alkyl) -amidodecanoyl) ] -4-hydroxyproline.
78. The dsRNA agent of any one of claims 1 to 48 and 64 to 76, wherein the sense strand comprises a nucleotide sequence of 5 '-cscsuuuggfafaagcugccuu-3' (SEQ ID NO: 2095) and the antisense strand comprises a nucleotide sequence of 5'-asAfsggdCa (G2 p) cuuauuuauauaaaggsgscsc-3' (SEQ ID NO: 2324) wherein a, G, c and U are 2 '-O-methyl (2' -OMe) A, G, C and U, respectively; af. Gf, cf and Uf are 2' -fluoro A, G, C and U, respectively; wherein G2p is guanosine-2 '-phosphate and s is a phosphorothioate linkage, and wherein the 3' end of the sense strand is conjugated to the ligand, as shown in the following schematic:
And wherein X is O.
79. The dsRNA agent of any one of claims 1 to 48, wherein the sense strand comprises at least 17 consecutive nucleotides differing by NO more than 3 nucleotides from the nucleotide sequence of 5'-UCACCUGCUUCUUCUGGUU-3' (SEQ ID NO: 1686).
80. The dsRNA agent of any one of claims 1 to 48 and 79, wherein the sense strand comprises a nucleotide sequence of 5'-UCACCUGCUUCUUCUGGUU-3' (SEQ ID NO: 1686).
81. The dsRNA agent of any one of claims 1 to 48, 79 and 80, wherein the sense strand consists of a nucleotide sequence of 5'-UCACCUGCUUCUUCUGGUU-3' (SEQ ID NO: 1686).
82. The dsRNA agent of any one of claims 1 to 48 and 79 to 81, wherein said antisense strand comprises at least 17 consecutive nucleotides differing by NO more than 3 nucleotides from the nucleotide sequence of 5'-AACCAGAAGAAGCAGGUGA-3' (SEQ ID NO: 1790).
83. The dsRNA agent of any one of claims 1 to 48 and 79 to 82, wherein the antisense strand comprises a nucleotide sequence of 5'-AACCAGAAGAAGCAGGUGA-3' (SEQ ID NO: 1790).
84. The dsRNA agent of any one of claims 1 to 48 and 79 to 83, wherein said antisense strand consists of a nucleotide sequence of 5'-AACCAGAAGAAGCAGGUGA-3' (SEQ ID NO: 1790).
85. The dsRNA agent of any one of claims 1 to 48 and 79 to 84, wherein the sense strand comprises a nucleotide sequence of 5'-UCACCUGCUUCUUCUGGUU-3' (SEQ ID NO: 1686) and the antisense strand comprises a nucleotide sequence of 5'-AACCAGAAGAAGCAGGUGA-3' (SEQ ID NO: 1790).
86. The dsRNA agent of any one of claims 1 to 48 and 79 to 85, wherein said sense strand differs from the nucleotide sequence of 5 '-uffccffugffucffuufgfgcgfsusuf-3' (SEQ ID NO: 1974) by NO more than 3 modified nucleotides, wherein a, g, c and U are 2 '-O-methyl (2' -OMe) A, G, C and U, respectively; af. Gf, cf and Uf are 2' -fluoro A, G, C and U, respectively; and s is a phosphorothioate bond.
87. The dsRNA agent of any one of claims 1 to 48 and 79 to 86, wherein said antisense strand differs from the nucleotide sequence of 5 '-asafscfagfafafafafafafafcggfusgfsa-3' (SEQ ID NO: 2203) by NO more than 3 modified nucleotides, wherein a, g, c and U are 2 '-O-methyl (2' -OMe) A, G, C and U, respectively; af. Gf, cf and Uf are 2' -fluoro A, G, C and U, respectively; and s is a phosphorothioate bond.
88. The dsRNA agent of any one of claims 1 to 48 and 79 to 87, wherein said sense strand comprises a nucleotide sequence of 5 '-uffccffugffucffuufgfgcgfsusuf-3' (SEQ ID NO: 1974) and said antisense strand comprises a nucleotide sequence of 5 '-asafscfafafafafafafafaffagfgfgcgfussgsa-3' (SEQ ID NO: 2203) wherein a, g, c and U are 2 '-O-methyl (2' -OMe) A, G, C and U, respectively; af. Gf, cf and Uf are 2' -fluoro A, G, C and U, respectively; and s is a phosphorothioate bond.
89. The dsRNA agent of any one of claims 1 to 48 and 79 to 88, wherein said sense strand comprises a nucleotide sequence of 5'-Q191s ufca fccfcu fucu fuu fcufgfgsuf-3' (SEQ ID NO: 1974) and said antisense strand comprises a nucleotide sequence of 5 '-asafscfaafaffaafafafaffagfgfgsgsgsa-3' (SEQ ID NO: 2203) wherein a, g, c and U are 2 '-O-methyl (2' -OMe) A, G, C and U, respectively; af. Gf, cf and Uf are 2' -fluoro A, G, C and U, respectively; s is a phosphorothioate linkage, and Q191 is N- [ tris (GalNAc-alkyl) -amidododecanoyl ] - (S) -pyrrolidin-3-ol-phosphorothioate (p-C12- (GalNAc-alkyl) 3).
90. The dsRNA agent of any one of claims 1 to 48 and 79 to 88, wherein said sense strand comprises a nucleotide sequence of 5 '-uffccffugffucffuufgfgcgfsusuf-3' (SEQ ID NO: 1974) and said antisense strand comprises a nucleotide sequence of 5 '-asafscfafafafafafafafaffagfgfgcgfussgsa-3' (SEQ ID NO: 2203) wherein a, g, c and U are 2 '-O-methyl (2' -OMe) A, G, C and U, respectively; af. Gf, cf and Uf are 2' -fluoro A, G, C and U, respectively; and s is a phosphorothioate linkage, wherein the 5' end of the sense strand is conjugated to the ligand, as shown in the following schematic:
91. A cell containing the dsRNA agent of any one of claims 1 to 90.
92. A pharmaceutical composition for inhibiting expression of a gene encoding a transmembrane protease serine 6 (TMPRSS 6), the pharmaceutical composition comprising the dsRNA agent of any one of claims 1 to 90.
93. The pharmaceutical composition of claim 92, wherein the dsRNA agent is in an unbuffered solution.
94. The pharmaceutical composition of claim 93, wherein the unbuffered solution is saline or water.
95. The pharmaceutical composition of claim 92, wherein the dsRNA agent is in a buffer solution.
96. The pharmaceutical composition of claim 95, wherein the buffer solution comprises acetate, citrate, prolamin, carbonate, or phosphate, or any combination thereof.
97. The pharmaceutical composition of claim 96, wherein the buffer solution is Phosphate Buffered Saline (PBS).
98. A method of inhibiting expression of a transmembrane protease serine 6 (TMPRSS 6) gene in a cell, the method comprising contacting the cell with the dsRNA agent of any one of claims 1 to 90 or the pharmaceutical composition of any one of claims 92 to 97, thereby inhibiting expression of the TMPRSS6 gene in the cell.
99. The method of claim 98, wherein the cell is in a subject.
100. The method of claim 98, wherein the subject is a human.
101. The method of claim 98 or 100, wherein the subject has a TMPRSS 6-related disorder.
102. The method of claim 101, wherein the TMPRSS6 related disorder is a disorder associated with iron overload and/or a disorder of ineffective erythropoiesis.
103. The method of claim 101, wherein the TMPRSS6 related disorder is selected from the group consisting of: hereditary hemochromatosis, beta-thalassemia, polycythemia vera, myelodysplastic syndrome, congenital erythropoiesis anemia, pyruvate kinase deficiency, erythropoietic porphyrin, parkinson's Disease, alzheimer's Disease, and Friedreich's Ataxia.
104. The method of claim 101, wherein the TMPRSS 6-related disorder is β -thalassemia.
105. The method of claim 104, wherein the beta-thalassemia is thalassemia major.
106. The method of claim 104, wherein the beta-thalassemia is an intermediate thalassemia.
107. The method of claim 101, wherein the TMPRSS6 related disorder is polycythemia vera.
108. The method of any one of claims 98-107, wherein contacting the cell with the dsRNA agent inhibits expression of TMPRSS6 by at least 50%, 60%, 70%, 80%, 90%, or 95%.
109. The method of any one of claims 98-108, wherein inhibiting expression of TMPRSS6 reduces TMPRSS6 protein levels in serum of the subject by at least 50%, 60%, 70%, 80%, 90% or 95%.
110. The method of any one of claims 98 to 109, wherein contacting the cell with the dsRNA agent increases expression of hepcidin by at least 50%, 60%, 70%, 80%, 90% or 95%.
111. The method of any one of claims 98-110, wherein increasing expression of hepcidin increases hepcidin protein levels in serum of the subject by at least 50%, 60%, 70%, 80%, 90%, or 95%.
112. A method of treating a subject suffering from a disorder that would benefit from reduced expression of the transmembrane protease serine 6 (TMPRSS 6), the method comprising administering to the subject a therapeutically effective amount of the dsRNA agent of any one of claims 1-58 or the pharmaceutical composition of any one of claims 50-55, thereby treating the subject suffering from the disorder that would benefit from reduced expression of TMPRSS 6.
113. A method of preventing at least one symptom of a subject suffering from a disorder that would benefit from reduced expression of the transmembrane protease serine 6 (TMPRSS 6), the method comprising administering to the subject a prophylactically effective amount of the dsRNA agent of any one of claims 1-48 or the pharmaceutical composition of any one of claims 50-55, thereby preventing at least one symptom of the subject suffering from the disorder that would benefit from reduced expression of TMPRSS 6.
114. The method of claim 112 or 113, wherein the disorder is a TMPRSS6 related disorder.
115. The method of claim 114, wherein the TMPRSS6 related disorder is a disorder associated with iron overload and/or a disorder of ineffective erythropoiesis.
116. The method of claim 115, wherein the TMPRSS6 related disorder is selected from the group consisting of: hereditary hemochromatosis, beta-thalassemia, polycythemia vera, myelodysplastic syndrome, congenital erythropoiesis anemia, pyruvate kinase deficiency, erythropoietic porphyrin, parkinson's disease, alzheimer's disease, and friedreich's ataxia.
117. The method of claim 114, wherein the TMPRSS 6-related disorder is β -thalassemia.
118. The method of claim 117, wherein the beta-thalassemia is thalassemia major.
119. The method of claim 117, wherein the beta-thalassemia is an intermediate thalassemia.
120. The method of claim 114, wherein the TMPRSS6 related disorder is polycythemia vera.
121. The method of any one of claims 112-120, wherein the subject is a human.
122. The method of any one of claims 112-121, wherein administration of the agent to the subject results in reduced iron levels, reduced ferritin levels, reduced transferrin saturation levels, increased hemoglobin levels, increased hematocrit levels, and/or reduced TMPRSS6 protein accumulation.
123. The method of any one of claims 112-122, wherein the dsRNA agent is administered to the subject at a dose of about 0.01mg/kg to about 50 mg/kg.
124. The method of any one of claims 112-123, wherein the dsRNA agent is administered subcutaneously to the subject.
125. The method of any one of claims 112-123, wherein the dsRNA agent is administered intravenously to the subject.
126. The method of any one of claims 112-125, further comprising determining a level of TMPRSS6 in a sample from the subject.
127. The method of claim 126, wherein the level of TMPRSS6 in the subject sample is a level of TMPRSS6 protein in a blood, serum or liver sample.
128. The method of any one of claims 112-127, further comprising determining the level of iron and/or hepcidin in a sample from the subject.
129. The method of any one of claims 112-128, further comprising administering to the subject an additional therapeutic agent for treating a TMPRSS6 related disorder.
130. The method of claim 129, wherein the additional therapeutic agent is an iron chelator.
131. The method of claim 130, wherein the iron chelator is selected from the group consisting of: deferiprone, deferoxamine and deferasirox (deferasirox).
132. A kit comprising the dsRNA agent of any one of claims 1 to 90 or the pharmaceutical composition of any one of claims 92 to 97.
133. A vial comprising the dsRNA agent of any one of claims 1 to 90 or the pharmaceutical composition of any one of claims 92 to 97.
134. A syringe comprising the dsRNA agent of any one of claims 1 to 90 or the pharmaceutical composition of any one of claims 92 to 97.
135. An RNA-induced silencing complex (RISC) comprising the antisense strand of the dsRNA agent of any one of claims 1-90.
CN202280030748.9A 2021-04-26 2022-04-25 Transmembrane protease serine 6 (TMPRSS 6) IRNA compositions and methods of use thereof Pending CN117203338A (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US63/179,607 2021-04-26
US202163278227P 2021-11-11 2021-11-11
US63/278,227 2021-11-11
PCT/US2022/026097 WO2022231999A1 (en) 2021-04-26 2022-04-25 Transmembrane protease, serine 6 (tmprss6) irna compositions and methods of use thereof

Publications (1)

Publication Number Publication Date
CN117203338A true CN117203338A (en) 2023-12-08

Family

ID=88983688

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202280030748.9A Pending CN117203338A (en) 2021-04-26 2022-04-25 Transmembrane protease serine 6 (TMPRSS 6) IRNA compositions and methods of use thereof

Country Status (1)

Country Link
CN (1) CN117203338A (en)

Similar Documents

Publication Publication Date Title
US11326166B1 (en) Xanthine dehydrogenase (XDH) iRNA compositions and methods of use thereof
US11866710B2 (en) Transmembrane protease, serine 6 (TMPRSS6) iRNA compositions and methods of use thereof
JP2024515423A (en) Ketohexokinase (KHK) iRNA compositions and methods of use thereof
TW202138559A (en) Patatin-like phospholipase domain containing 3 (pnpla3) irna compositions and methods of use thereof
JP2023516095A (en) Ketohexokinase (KHK) iRNA compositions and methods of use thereof
US20230133637A1 (en) Angiopoietin-like 3 (angptl3) irna compositions and methods of use thereof
CN117203338A (en) Transmembrane protease serine 6 (TMPRSS 6) IRNA compositions and methods of use thereof
US20240182905A1 (en) TRANSMEMBRANE PROTEASE, SERINE 6 (TMPRSS6) iRNA COMPOSITIONS AND METHODS OF USE THEREOF
US20240132896A1 (en) FACTOR XII (F12) iRNA COMPOSITIONS AND METHODS OF USE THEREOF
TW202237841A (en) Coagulation factor v (f5) irna compositions and methods of use thereof
CN116964204A (en) Angiopoietin-like 3 (ANGPTL 3) iRNA compositions and methods of use thereof
CN116529372A (en) Coagulation factor V (F5) iRNA compositions and methods of use thereof
CN117751187A (en) Ketohexokinase (KHK) iRNA compositions and methods of use thereof
TW202333749A (en) Complement factor b (cfb) irna compositions and methods of use thereof
WO2022212153A1 (en) Proline dehydrogenase 2 (prodh2) irna compositions and methods of use thereof
EP4045652A1 (en) Solute carrier family member irna compositions and methods of use thereof
CN117795074A (en) Transthyretin (TTR) iRNA compositions and methods of use thereof
CN117651769A (en) Beta catenin (CTNNB 1) iRNA compositions and methods of use thereof

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination