Movatterモバイル変換


[0]ホーム

URL:


WO2025072672A2 - Slc6a19-targeting modulatory nucleic acid agents - Google Patents

Slc6a19-targeting modulatory nucleic acid agents
Download PDF

Info

Publication number
WO2025072672A2
WO2025072672A2PCT/US2024/048852US2024048852WWO2025072672A2WO 2025072672 A2WO2025072672 A2WO 2025072672A2US 2024048852 WUS2024048852 WUS 2024048852WWO 2025072672 A2WO2025072672 A2WO 2025072672A2
Authority
WO
WIPO (PCT)
Prior art keywords
nucleotides
seq
nos
nucleotide
aso
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
PCT/US2024/048852
Other languages
French (fr)
Other versions
WO2025072672A3 (en
Inventor
Brian Bettencourt
Steve ROBINETTE
Jeremy CUNIFF
Alfica Sehgal
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.)
Judo Bio Inc
Original Assignee
Judo Bio 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 Judo Bio IncfiledCriticalJudo Bio Inc
Publication of WO2025072672A2publicationCriticalpatent/WO2025072672A2/en
Publication of WO2025072672A3publicationCriticalpatent/WO2025072672A3/en
Pendinglegal-statusCriticalCurrent
Anticipated expirationlegal-statusCritical

Links

Classifications

Definitions

Landscapes

Abstract

This disclosure relates to oligonucleotides, compositions and methods useful for reducing SLC6A19 mRNA and/or B0AT1 protein expression, particularly in kidney cells. Disclosed oligonucleotides for the reduction of SLC6A19/B0AT1 expression may be double-stranded or single-stranded and may be modified for improved characteristics such as stronger resistance to nucleases and lower immunogenicity. Disclosed oligonucleotides for the reduction of SLC6A19/B0AT1 expression may also include targeting ligands to target a particular cell or organ, such as the proximal tubular epithelial cells and podocytes of the kidney, and may be used to treat diseases or disorders, including renal diseases and disorders (e.g., PKU, urea cycle disorders, chronic kidney disease, renal tubular acidosis, etc.).

Description

SLC6A19-TARGETING MODULATORY NUCLEIC ACID AGENTS
CROSS-REFERENCE TO RELATED APPLICATIONS
This application claims the benefit of and priority to U.S. Provisional Patent Application No. 63/585,781, filed on September 27, 2023, and U.S. Provisional Patent Application No. 63/655,344, filed on June 3, 2024, the contents of which are incorporated by reference in their entireties herein.
FIELD OF THE INVENTION
The instant disclosure relates generally to SLC6A19-targeting modulatory nucleic acid agents and methods.
BACKGROUND OF THE INVENTION
Modulatory nucleic acid therapeutics have demonstrated significant clinical and commercial success in treating liver disease, at least in part because fenestrations in liver sinusoidal epithelial cells render such cells particularly accessible to injected nucleic acids (especially those carrying N-acetylgalactosamine (GalNAc) modifications). Non-liver tissues, including kidney, have tended to be less accessible to modulatory nucleic acid therapeutics. However, recent discoveries have described successful use of moieties targeting kidney cell surface factors to promote efficient delivery of moiety-linked modulatory nucleic acids to kidney cells (see, e.g., PCT/US23/16319).
The human Solute Carrier Family 6 Member 19 (SLC6A19) gene encodes the sodiumdependent neutral amino acid transporter B°AT1. B°AT1 has been shown to mediate absorption of neutral amino acids in the small intestine and in the kidneys, where it is primarily expressed in early proximal tubules; and a role for SLC6A19 knockout or inhibition in treatment of metabolic disorders, including nephropathy, has been suggested.
While likely involvement of SLC6A19 and the encoded B°AT1 protein in kidney disease has recently been described, there remains a need for an agent possessing high biological activity and in vivo stability - particularly a kidney cell-directed agent - that can selectively and efficiently modulate (e.g., inhibit) the SLC6A19 gene. BRIEF SUMMARY OF THE INVENTION
The present disclosure provides modulatory nucleic acid agent compositions which block, degrade, promote or inhibit specific splicing and/or otherwise alter expression of a SLC6AI9 target RNA with sequence-specific targeting precision. Without wishing to be bound by theory', the SLC6A19-targeting nucleic acid agents of the instant disclosure can act via a direct antisense oligonucleotide (ASO) mechanism upon RNA transcripts of a SLC6A19 gene, can induce RNA- induced silencing complex (RlSC)-mediated cleavage of RNA transcripts of a SLC6A19 gene, and/or can otherwise modulate expression (e.g., by altering splice site selection) of RNA transcripts of a SLC6A19 gene. The SLC6A19 gene may be within a cell, e.g., a cell within a subject, such as a human. In certain embodiments, the modulatory nucleic acid agent is designed and directed for knockdown of SLC6A19 in kidney cells and/or tissues. The present disclosure also provides nucleic acid agents that are conjugated to kidney -targeting moieties, particularly vitamin carrier proteins and other proteins capable of binding Megalin and/or Cubilin and promoting endocytic uptake of an associated (e.g., linked/conjugated) modulatory nucleic acid agent of the instant disclosure. The instant disclosure additionally provides methods of using the modulatory nucleic acid agent compositions of the disclosure for altering (e.g., inhibiting) the expression of a SLC6A19 gene or B°AT1 protein for treating a subject who would benefit from altering (e.g., inhibiting or reducing) the expression of a SLC6A19 gene, e.g., a subject suffering or prone to suffering from a SLC6A19-associated disease or disorder, e.g., a metabolic disease and/or a kidney disease, e.g., a glomerular disorder, a renal tubular disorder, other renal disorders, an inborn error of metabolism, a systemic metabolic disorder, a disorder of the thyroid, a disorder of the parathyroid, a disorder of the inner ear, a neurological disorder, or a viral infection, or any combination thereof.
In one aspect, the instant disclosure provides a modulatory nucleic acid agent that includes a first strand of 12 to 60 linked nucleosides in length targeted to a human solute carrier family 6 member 19 (SLC6A19) RNA, where the human SLC6A19 RNA is the human SLC6A19 mRNA of SEQ ID NO: 1. In one embodiment, the modulatory nucleic acid agent is an antisense oligonucleotide (ASO).
In one embodiment, the modulatory nucleic acid agent of the disclosure (e g., ASO, dsRNA, etc.) is at least about 80% complementarity to a target region of the human SLC6A19 RNA. In some embodiments, the antisense strand hybridizes with 12 or more nucleotides of nucleotides 3 to 25, nucleotides 8 to 30, nucleotides 13 to 35, nucleotides 38 to 60, nucleotides 43 to 65, nucleotides 48 to 70, nucleotides 93 to 115, nucleotides 98 to 120, nucleotides 103 to 125, nucleotides 108 to 130, nucleotides 111 to 133, nucleotides 113 to 135, nucleotides 118 to 140, nucleotides 214 to 236, nucleotides 219 to 241, nucleotides 244 to 266, nucleotides 249 to 271, nucleotides 252 to 274, nucleotides 252 to 274, nucleotides 253 to 275, nucleotides 254 to 276, nucleotides 257 to 286, nucleotides 259 to 281, nucleotides 264 to 286, nucleotides 289 to 311, nucleotides 294 to 316, nucleotides 339 to 361, nucleotides 397 to 419, nucleotides 460 to 480, nucleotides 461 to 481, nucleotides 481 to 503, nucleotides 486 to 508, nucleotides 491 to 513, nucleotides 557 to 579, nucleotides 561 to 583, nucleotides 562 to 584, nucleotides 564 to 586, nucleotides 566 to 586, nucleotides 567 to 589, nucleotides 616 to 638, nucleotides 621 to 643, nucleotides 626 to 648, nucleotides 670 to 692, nucleotides 671 to 691, nucleotides 672 to 694, nucleotides 674 to 694, nucleotides 675 to 697, nucleotides 680 to 702, nucleotides 685 to 707, nucleotides 690 to 712, nucleotides 695 to 717, nucleotides 700 to 722, nucleotides 705 to 727, nucleotides 710 to 732, nucleotides 715 to 737, nucleotides 720 to 742, nucleotides 725 to 747, nucleotides 730 to 752, nucleotides 735 to 757, nucleotides 740 to 762, nucleotides 745 to 767, nucleotides 750 to 772, nucleotides 751 to 776, nucleotides 755 to 777, nucleotides 760 to 782, nucleotides 765 to 787, nucleotides 770 to 792, nucleotides 775 to 797, nucleotides 798 to 823, nucleotides 800 to 822, nucleotides 825 to 847, nucleotides 890 to 912, nucleotides 895 to 917, nucleotides 900 to 922, nucleotides 905 to 927, nucleotides 910 to 932, nucleotides 911 to 937, nucleotides 915 to 937, nucleotides 972 to 994, nucleotides 973 to 995, nucleotides 977 to 1000, nucleotides 977 to 999, nucleotides 978 to 1000, nucleotides 983 to 1005, nucleotides 1035 to 1057, nucleotides 1058 to 1080, nucleotides 1059 to 1081, nucleotides 1060 to 1082, nucleotides 1063 to 1082, nucleotides 1065 to 1087, nucleotides 1068 to 1087, nucleotides 1070 to 1094, nucleotides 1070 to 1092, nucleotides 1074 to 1096, nucleotides 1075 to 1097, nucleotides 1077 to 1096, nucleotides 1080 to 1102, nucleotides 1085 to 1107, nucleotides 1086 to 1106, nucleotides 1090 to 1112, nucleotides 1095 to 1117, nucleotides 1100 to 1122, nucleotides 1105 to 1127, nucleotides 1110 to 1132, nucleotides 1115 to 1137, nucleotides 1115 to 1137, nucleotides 1117 to 1139, nucleotides 1177 to 1199, nucleotides 1182 to 1204, nucleotides 1187 to 1209, nucleotides 1191 to 1214, nucleotides 1192 to 1214, nucleotides 1195 to 1217, nucleotides 1234 to 1256, nucleotides 1239 to 1261, nucleotides 1242 to 1264, nucleotides 1244 to 1266, nucleotides 1247 to 1268, nucleotides 1249 to 1271, nucleotides 1251 to 1270, nucleotides 1254 to 1276, nucleotides 1259 to 1281, nucleotides 1263 to 1282, nucleotides 1264 to 1286, nucleotides 1266 to 1285, nucleotides 1269 to 1291, nucleotides 1326 to 1348, nucleotides 1331 to 1353, nucleotides 1334 to 1361, nucleotides 1334 to 1356, nucleotides 1334 to 1356, nucleotides 1335 to 1357, nucleotides 1336 to 1358, nucleotides 1337 to 1359, nucleotides 1337 to 1359, nucleotides 1338 to 1360, nucleotides 1340 to 1362, nucleotides 1341 to 1363, nucleotides 1346 to 1368, nucleotides 1351 to 1373, nucleotides 1353 to 1372, nucleotides 1377 to 1399, nucleotides 1382 to 1404, nucleotides 1383 to 1403, nucleotides 1387 to 1409, nucleotides 1392 to 1414, nucleotides 1397 to 1419, nucleotides 1402 to 1424, nucleotides 1407 to 1429, nucleotides 1412 to 1434, nucleotides 1417 to 1439, nucleotides 1456 to 1478, nucleotides 1458 to 1477, nucleotides 1481 to 1503, nucleotides 1486 to 1508, nucleotides 1491 to 1513, nucleotides 1496 to 1518, nucleotides 1521 to 1543, nucleotides 1568 to 1590, nucleotides 1569 to 1591, nucleotides 1574 to 1596, nucleotides 1578 to 1621, nucleotides 1579 to 1601, nucleotides 1583 to 1605, nucleotides 1583 to 1605, nucleotides 1584 to 1606, nucleotides 1584 to 1606, nucleotides 1585 to 1607, nucleotides 1585 to 1607, nucleotides 1587 to 1609, nucleotides 1588 to 1610, nucleotides 1589 to 1611, nucleotides 1594 to 1616, nucleotides 1595 to 1617, nucleotides 1599 to 1621, nucleotides 1601 to 1620, nucleotides 1602 to 1624, nucleotides 1642 to 1664, nucleotides 1749 to 1775, nucleotides 1750 to 1772, nucleotides 1755 to 1777, nucleotides 1757 to 1779, nucleotides 1758 to 1780, nucleotides 1760 to 1782, nucleotides 1761 to 1792, nucleotides 1764 to 1783, nucleotides 1765 to 1787, nucleotides 1770 to 1792, nucleotides 1809 to 1831, nucleotides 1810 to 1829, nucleotides 1812 to 1832, nucleotides 1814 to 1836, nucleotides 1852 to 1881, nucleotides 1853 to 1875, nucleotides 1854 to 1876, nucleotides 1858 to 1880, nucleotides 2168 to 2188, nucleotides 2333 to 2353, nucleotides 2454 to 2474, nucleotides 2507 to 2527, nucleotides 2593 to 2613, nucleotides 3244 to 3264, nucleotides 3288 to 3308, nucleotides 3304 to 3324, nucleotides 3332 to 3352, nucleotides 3333 to 3353, nucleotides 3501 to 3523, nucleotides 3595 to 3615, nucleotides 3595 to 3617, nucleotides 3643 to 3671, nucleotides 3646 to 3668, nucleotides 3646 to 3668, nucleotides 3647 to 3669, nucleotides 3648 to 3668, nucleotides 3648 to 3670, nucleotides 3648 to 3670, nucleotides 3649 to 3671, nucleotides 3651 to 3670, nucleotides 3651 to 3673, nucleotides 3651 to 3673, nucleotides 3652 to 3671, nucleotides 3712 to 3733, nucleotides 3713 to 3735, nucleotides 3763 to 3786, nucleotides 3763 to 3785, nucleotides 3764 to 3786, nucleotides 3791 to 3811, nucleotides 3796 to 3816, nucleotides 3801 to 3829, nucleotides 3801 to 3820, nucleotides 3802 to 3821, nucleotides 3804 to 3826, nucleotides 3809 to 3831, nucleotides 3810 to 3832, nucleotides 3812 to 3833, nucleotides 3812 to 3834, nucleotides 3813 to 3835, nucleotides 3814 to 3833, nucleotides 3814 to 3836, nucleotides 3814 to 3836, nucleotides 3839 to 3859, nucleotides 3840 to 3862, nucleotides 3902 to 3924, nucleotides 3965 to 3987, nucleotides 3995 to 4015, nucleotides 3998 to 4020, nucleotides 4100 to 4124, nucleotides 4102 to 4124, nucleotides 4141 to 4163, nucleotides 4141 to4163, nucleotides 4142 to 4164, nucleotides 4143 to 4165, nucleotides 4144 to 4166, nucleotides 4146 to 4168, nucleotides 4150 to 4172, nucleotides 4151 to 4173, nucleotides 4192 to 4214, nucleotides 4197 to 4219, nucleotides 4201 to 4223, nucleotides 4265 to 4287, nucleotides 4270 to 4292, nucleotides 4318 to 4338, nucleotides 4319 to 4341, nucleotides 4329 to 4349, nucleotides 4348 to 4386, nucleotides 4349 to 4371, nucleotides 4350 to 4369, nucleotides 4350 to 4372, nucleotides 4350 to 4372, nucleotides 4351 to 4370, nucleotides 4352 to 4371, nucleotides 4352 to 4374, nucleotides 4353 to 4372, nucleotides 4353 to 4375, nucleotides 4354 to 4376, nucleotides 4359 to 4381, nucleotides 4361 to 4383, nucleotides 4362 to 4384, nucleotides 4362 to 4384, nucleotides 4364 to 4386, nucleotides 4365 to 4387, nucleotides 4395 to 4415, nucleotides 4396 to 4416, nucleotides 4409 to 4431, nucleotides 4410 to 4432, nucleotides 4412 to 4434, nucleotides 4637 to 4657, nucleotides 4638 to 4658, nucleotides 4640 to 4660, nucleotides 5103 to 5123, nucleotides 5124 to 5144, nucleotides 5126 to 5146, or nucleotides 5127 to 5147 of SEQ ID NO: 1.
In another embodiment, the ASO includes a second strand of 15 to 60 nucleobases in length and complementary to the first strand
In certain embodiments, the ASO includes a complementary strand of 15 to 60 nucleobases in length.
In some embodiments, the compound is an antisense oligonucleotide (ASO).
In one embodiment, the modulatory nucleic acid agent of the disclosure (e.g., antisense compound, dsRNA, etc.) has at least one modified internucleoside linkage, sugar moiety, or nucleobase. Optionally, the modified internucleoside linkage is or includes a phosphorothioate linkage.
In certain embodiments, the at least one modified internucleoside linkage, sugar moiety, or nucleobase includes one or more of the following modifications: a deoxy-nucleoside, a 3 '-terminal deoxy-thymine (dT) nucleoside, a 2'-O-methyl (2'-0Me) modified nucleoside, a 2‘~fluoro (2’-F) modified nucleoside, a 2'-deoxy-modified nucleoside, a locked nucleotide (LNA), an unlocked nucleoside, a conformationally restricted nucleoside, a constrained ethyl nucleoside, an abasic nucleoside, a 2'-O,4'-C-ethylene-bridged nucleic acid (ENA), a 2’-amino-modified nucleoside, a 2’-O-allyl-modified nucleoside, 2’-C-alkyl-modified nucleoside, 2"-hydroxly-modified nucleoside, a 2’-methoxy ethyl (2'- MOI i) modified nucleoside, a 2’-O-alkylmodified nucleoside, a morpholino nucleoside, a phospboramidate, a non-natural base including nucleoside, a tetrahydropyran modified nucleoside, a 1,5-anhydrohexitol modified nucleoside, a cyclohexenyl modified nucleoside, a nucleotide including a 5'-phosphorothioate group, a nucleotide including a 5'-methylphosphonate group, a nucleotide including a 5’ phosphate or 5’ phosphate mimic, a nucleotide including vinyl phosphonate, a nucleoside including adenosine-glycol nucleic acid (GN A), a nucleoside including thymidine-glycol nucleic acid (GN A) S-Isomer, a nucleotide including 2-hydroxymethyl-tetrahydrofurane-5-phosphate, a nucleotide including 2 ’ deoxy thymi di ne-3’ phosphate, a nucleotide including 2 ’-deoxyguanosine-3 ’-phosphate, and a terminal nucleoside linked to a cholesteryl derivative and a dodecanoic acid bisdecylamide group; and combinations thereof.
In certain embodiments, the modified nucleobase is or includes 5-methylcytosine.
Another aspect of the instant disclosure provides a double stranded ribonucleic acid (dsRNA) agent for inhibiting expression of human solute carrier family 6 member 19 (SLC6A19), where the dsRNA agent includes a sense strand and an antisense strand forming a double stranded region, where the antisense strand includes a region of complementarity to a SLC6A19 RNA, and where the region of complementarity includes at least 15 contiguous nucleotides differing by no more than 3 nucleotides from any one of the antisense nucleotide sequences of Tables 7-11, 14 or 17.
An additional aspect of the instant disclosure provides a double stranded ribonucleic acid (dsRNA) agent for inhibiting expression of human solute carrier family 6 member 19 (SLC6A19) in a cell, where the dsRNA agent includes a sense strand and an antisense strand forming a double stranded region, where the sense strand includes at least 15 contiguous nucleotides differing by no more than three nucleotides from SEQ ID NO: 1.
In one embodiment, the sense strand includes at least 15 contiguous nucleotides differing by no more than three nucleotides from any one of the nucleotide sequences of nucleotides 3 to 25, nucleotides 8 to 30, nucleotides 13 to 35, nucleotides 38 to 60, nucleotides 43 to 65, nucleotides 48 to 70, nucleotides 93 to 115, nucleotides 98 to 120, nucleotides 103 to 125, nucleotides 108 to 130, nucleotides 1 11 to 133, nucleotides 113 to 135, nucleotides 1 18 to 140, nucleotides 214 to
236, nucleotides 219 to 241, nucleotides 244 to 266, nucleotides 249 to 271, nucleotides 252 to
274, nucleotides 252 to 274, nucleotides 253 to 275, nucleotides 254 to 276, nucleotides 257 to
286, nucleotides 259 to 281, nucleotides 264 to 286, nucleotides 289 to 311, nucleotides 294 to
316, nucleotides 339 to 361, nucleotides 397 to 419, nucleotides 460 to 480, nucleotides 461 to
481, nucleotides 481 to 503, nucleotides 486 to 508, nucleotides 491 to 513, nucleotides 557 to
579, nucleotides 561 to 583, nucleotides 562 to 584, nucleotides 564 to 586, nucleotides 566 to
586, nucleotides 567 to 589, nucleotides 616 to 638, nucleotides 621 to 643, nucleotides 626 to
648, nucleotides 670 to 692, nucleotides 671 to 691, nucleotides 672 to 694, nucleotides 674 to
694, nucleotides 675 to 697, nucleotides 680 to 702, nucleotides 685 to 707, nucleotides 690 to
712, nucleotides 695 to 717, nucleotides 700 to 722, nucleotides 705 to 727, nucleotides 710 to
732, nucleotides 715 to 737, nucleotides 720 to 742, nucleotides 725 to 747, nucleotides 730 to
752, nucleotides 735 to 757, nucleotides 740 to 762, nucleotides 745 to 767, nucleotides 750 to
772, nucleotides 751 to 776, nucleotides 755 to 777, nucleotides 760 to 782, nucleotides 765 to
787, nucleotides 770 to 792, nucleotides 775 to 797, nucleotides 798 to 823, nucleotides 800 to
822, nucleotides 825 to 847, nucleotides 890 to 912, nucleotides 895 to 917, nucleotides 900 to
922, nucleotides 905 to 927, nucleotides 910 to 932, nucleotides 911 to 937, nucleotides 915 to
937, nucleotides 972 to 994, nucleotides 973 to 995, nucleotides 977 to 1000, nucleotides 977 to 999, nucleotides 978 to 1000, nucleotides 983 to 1005, nucleotides 1035 to 1057, nucleotides 1058 to 1080, nucleotides 1059 to 1081, nucleotides 1060 to 1082, nucleotides 1063 to 1082, nucleotides 1065 to 1087, nucleotides 1068 to 1087, nucleotides 1070 to 1094, nucleotides 1070 to 1092, nucleotides 1074 to 1096, nucleotides 1075 to 1097, nucleotides 1077 to 1096, nucleotides 1080 to 1102, nucleotides 1085 to 1107, nucleotides 1086 to 1106, nucleotides 1090 to 1112, nucleotides 1095 to 1117, nucleotides 1100 to 1122, nucleotides 1105 to 1127, nucleotides 1110 to 1132, nucleotides 1115 to 1137, nucleotides 1115 to 1137, nucleotides 1117 to 1139, nucleotides 1177 to 1199, nucleotides 1182 to 1204, nucleotides 1187 to 1209, nucleotides 1191 to 1214, nucleotides 1192 to 1214, nucleotides 1195 to 1217, nucleotides 1234 to 1256, nucleotides 1239 to 1261, nucleotides 1242 to 1264, nucleotides 1244 to 1266, nucleotides 1247 to 1268, nucleotides 1249 to 1271, nucleotides 1251 to 1270, nucleotides 1254 to 1276, nucleotides 1259 to 1281, nucleotides 1263 to 1282, nucleotides 1264 to 1286, nucleotides 1266 to 1285, nucleotides 1269 to 1291, nucleotides 1326 to 1348, nucleotides 1331 to 1353, nucleotides 1334 to 1361, nucleotides 1334 to 1356, nucleotides 1334 to 1356, nucleotides 1335 to 1357, nucleotides 1336 to 1358, nucleotides 1337 to 1359, nucleotides 1337 to 1359, nucleotides 1338 to 1360, nucleotides 1340 to 1362, nucleotides 1341 to 1363, nucleotides 1346 to 1368, nucleotides 1351 to 1373, nucleotides 1353 to 1372, nucleotides 1377 to 1399, nucleotides 1382 to 1404, nucleotides 1383 to 1403, nucleotides 1387 to 1409, nucleotides 1392 to 1414, nucleotides 1397 to 1419, nucleotides 1402 to 1424, nucleotides 1407 to 1429, nucleotides 1412 to 1434, nucleotides 1417 to 1439, nucleotides 1456 to 1478, nucleotides 1458 to 1477, nucleotides 1481 to 1503, nucleotides 1486 to 1508, nucleotides 1491 to 1513, nucleotides 1496 to 1518, nucleotides 1521 to 1543, nucleotides 1568 to 1590, nucleotides 1569 to 1591, nucleotides 1574 to 1596, nucleotides 1578 to 1621, nucleotides 1579 to 1601, nucleotides 1583 to 1605, nucleotides 1583 to 1605, nucleotides 1584 to 1606, nucleotides 1584 to 1606, nucleotides 1585 to 1607, nucleotides 1585 to 1607, nucleotides 1587 to 1609, nucleotides 1588 to 1610, nucleotides 1589 to 1611, nucleotides 1594 to 1616, nucleotides 1595 to 1617, nucleotides 1599 to 1621, nucleotides 1601 to 1620, nucleotides 1602 to 1624, nucleotides 1642 to 1664, nucleotides 1749 to 1775, nucleotides 1750 to 1772, nucleotides 1755 to 1777, nucleotides 1757 to 1779, nucleotides 1758 to 1780, nucleotides 1760 to 1782, nucleotides 1761 to 1792, nucleotides 1764 to 1783, nucleotides 1765 to 1787, nucleotides 1770 to 1792, nucleotides 1809 to 1831, nucleotides 1810 to 1829, nucleotides 1812 to 1832, nucleotides 1814 to 1836, nucleotides 1852 to 1881, nucleotides 1853 to 1875, nucleotides 1854 to 1876, nucleotides 1858 to 1880, nucleotides 2168 to 2188, nucleotides 2333 to 2353, nucleotides 2454 to 2474, nucleotides 2507 to 2527, nucleotides 2593 to 2613, nucleotides 3244 to 3264, nucleotides 3288 to 3308, nucleotides 3304 to 3324, nucleotides 3332 to 3352, nucleotides 3333 to 3353, nucleotides 3501 to 3523, nucleotides 3595 to 3615, nucleotides 3595 to 3617, nucleotides 3643 to 3671, nucleotides 3646 to 3668, nucleotides 3646 to 3668, nucleotides 3647 to 3669, nucleotides 3648 to 3668, nucleotides 3648 to 3670, nucleotides 3648 to 3670, nucleotides 3649 to 3671, nucleotides 3651 to 3670, nucleotides 3651 to 3673, nucleotides 3651 to 3673, nucleotides 3652 to 3671, nucleotides 3712 to 3733, nucleotides 3713 to 3735, nucleotides 3763 to 3786, nucleotides 3763 to 3785, nucleotides 3764 to 3786, nucleotides 3791 to 3811, nucleotides 3796 to 3816, nucleotides 3801 to 3829, nucleotides 3801 to 3820, nucleotides 3802 to 3821, nucleotides 3804 to 3826, nucleotides 3809 to 3831, nucleotides 3810 to 3832, nucleotides 3812 to 3833, nucleotides 3812 to 3834, nucleotides 3813 to 3835, nucleotides 3814 to 3833, nucleotides 3814 to 3836, nucleotides 3814 to 3836, nucleotides 3839 to 3859, nucleotides 3840 to 3862, nucleotides 3902 to 3924, nucleotides 3965 to 3987, nucleotides 3995 to 4015, nucleotides 3998 to 4020, nucleotides 4100 to 4124, nucleotides 4102 to 4124, nucleotides 4141 to 4163, nucleotides 4141 to 4163, nucleotides 4142 to 4164, nucleotides 4143 to 4165, nucleotides 4144 to 4166, nucleotides 4146 to 4168, nucleotides 4150 to 4172, nucleotides 4151 to 4173, nucleotides 4192 to 4214, nucleotides 4197 to 4219, nucleotides 4201 to 4223, nucleotides 4265 to 4287, nucleotides 4270 to 4292, nucleotides 4318 to 4338, nucleotides 4319 to 4341, nucleotides 4329 to 4349, nucleotides 4348 to 4386, nucleotides 4349 to 4371, nucleotides 4350 to 4369, nucleotides 4350 to 4372, nucleotides 4350 to 4372, nucleotides 4351 to 4370, nucleotides 4352 to 4371, nucleotides 4352 to 4374, nucleotides 4353 to4372, nucleotides 4353 to 4375, nucleotides 4354 to 4376, nucleotides 4359 to 4381, nucleotides 4361 to 4383, nucleotides 4362 to 4384, nucleotides 4362 to 4384, nucleotides 4364 to 4386, nucleotides 4365 to 4387, nucleotides 4395 to 4415, nucleotides 4396 to 4416, nucleotides 4409 to 4431, nucleotides 4410 to 4432, nucleotides 4412 to 4434, nucleotides 4637 to 4657, nucleotides 4638 to 4658, nucleotides 4640 to 4660, nucleotides 5103 to 5123, nucleotides 5124 to 5144, nucleotides 5126 to 5146, or nucleotides 5127 to 5147 of SEQ ID NO: 1 Optionally, the antisense strand includes at least 15 contiguous nucleotides from the corresponding nucleotide sequence of SEQ ID NO: 2.
In one embodiment, the antisense strand includes at least 15 contiguous nucleotides differing by not more than three nucleotides from any one of the antisense strand nucleotide sequences of the following duplexes: SEQ ID NOs: 2318 and 2430; SEQ ID NOs: 2319 and 2431; SEQ ID NOs: 2320 and 2432; SEQ ID NOs: 2321 and 2433; SEQ ID NOs: 2322 and 2434; SEQ ID NOs: 2323 and 2435; SEQ ID NOs: 2324 and 2436; SEQ ID NOs: 2325 and 2437; SEQ ID NOs: 2326 and 2438; SEQ ID NOs: 2327 and 2439; SEQ ID NOs: 2328 and 2440; SEQ ID NOs: 2329 and 2441; SEQ ID NOs: 2330 and 2442; SEQ ID NOs: 2331 and 2443; SEQ ID NOs: 2332 and 2444; SEQ ID NOs: 2333 and 2445; SEQ ID NOs: 2334 and 2446; SEQ ID NOs: 2335 and 2447; SEQ ID NOs: 2336 and 2448; SEQ ID NOs: 2337 and 2449; SEQ ID NOs: 2338 and 2450; SEQ ID NOs: 2339 and 2451; SEQ ID NOs: 2340 and 2452; SEQ ID NOs: 2341 and 2453; SEQ ID NOs: 2342 and 2454; SEQ ID NOs: 2343 and 2455; SEQ ID NOs: 2344 and 2456; SEQ ID NOs: 2345 and 2457; SEQ ID NOs: 2346 and 2458; SEQ ID NOs: 2347 and 2459; SEQ ID NOs: 2348 and 2460; SEQ ID NOs: 2349 and 2461; SEQ ID NOs: 2350 and 2462; SEQ ID NOs: 2351 and 2463; SEQ ID NOs: 2352 and 2464; SEQ ID NOs: 2353 and 2465; SEQ ID NOs: 2354 and 2466; SEQ ID NOs: 2355 and 2467; SEQ ID NOs: 2356 and 2468; SEQ ID NOs: 2357 and 2469; SEQ ID NOs: 2358 and 2470; SEQ ID NOs: 2359 and 2471 ; SEQ ID NOs: 2360 and 2472; SEQ ID NOs: 2361 and 2473; SEQ ID NOs: 2362 and 2474; SEQ ID NOs: 2363 and 2475; SEQ ID NOs: 2364 and 2476; SEQ ID NOs: 2365 and 2477; SEQ ID NOs: 2366 and 2478; SEQ ID NOs: 2367 and 2479; SEQ ID NOs: 2368 and 2480; SEQ ID NOs: 2369 and 2481; SEQ ID NOs: 2370 and 2482; SEQ ID NOs: 2371 and 2483; SEQ ID NOs: 2372 and 2484; SEQ ID NOs: 2373 and 2485; SEQ ID NOs: 2374 and 2486; SEQ ID NOs: 2375 and 2487; SEQ ID NOs: 2376 and 2488; SEQ ID NOs: 2377 and 2489; SEQ ID NOs: 2378 and 2490; SEQ ID NOs: 2379 and 2491; SEQ ID NOs: 2380 and 2492; SEQ ID NOs: 2381 and 2493; SEQ ID NOs: 2382 and 2494; SEQ ID NOs: 2383 and 2495; SEQ ID NOs: 2384 and 2496; SEQ ID NOs: 2385 and 2497; SEQ ID NOs: 2386 and 2498; SEQ ID NOs: 2387 and 2499; SEQ ID NOs: 2388 and 2500; SEQ ID NOs: 2389 and 2501; SEQ ID NOs: 2390 and 2502; SEQ ID NOs: 2391 and 2503; SEQ ID NOs: 2392 and 2504; SEQ ID NOs: 2393 and 2505; SEQ ID NOs: 2394 and 2506; SEQ ID NOs: 2395 and 2507; SEQ ID NOs: 2396 and 2508; SEQ ID NOs: 2397 and 2509; SEQ ID NOs: 2398 and 2510; SEQ ID NOs: 2399 and 2511; SEQ ID NOs: 2400 and 2512; SEQ ID NOs: 2401 and 2513; SEQ ID NOs: 2402 and 2514; SEQ ID NOs: 2403 and 2515; SEQ ID NOs: 2404 and 2516; SEQ ID NOs: 2405 and 2517; SEQ ID NOs: 2406 and 2518; SEQ ID NOs: 2407 and 2519; SEQ ID NOs: 2408 and 2520; SEQ ID NOs: 2409 and 2521; SEQ ID NOs: 2410 and 2522; SEQ ID NOs: 2411 and 2523; SEQ ID NOs: 2412 and 2524; SEQ ID NOs: 2413 and 2525; SEQ ID NOs: 2414 and 2526; SEQ ID NOs: 2415 and 2527; SEQ ID NOs: 2416 and 2528; SEQ ID NOs: 2417 and 2529; SEQ ID NOs: 2418 and 2530; SEQ ID NOs: 2419 and 2531; SEQ ID NOs: 2420 and 2532; SEQ ID NOs: 2421 and 2533; SEQ ID NOs: 2422 and 2534; SEQ ID NOs: 2423 and 2535; SEQ ID NOs: 2424 and 2536; SEQ ID NOs: 2425 and 2537; SEQ ID NOs: 2426 and 2538; SEQ ID NOs: 2427 and 2539; SEQ ID NOs: 2428 and 2540; SEQ ID NOs: 2429 and 2541; SEQ ID NO: 2720 and 2840; SEQ ID NO: 2721 and 2841; SEQ ID NO: 2722 and 2842; SEQ ID NO: 2723 and 2843; SEQ ID NO: 2724 and 2844; SEQ ID NO: 2725 and 2845; SEQ ID NO: 2726 and 2846; SEQ ID NO: 2727 and 2847; SEQ ID NO: 2728 and 2848; SEQ ID NO: 2729 and 2849; SEQ ID NO: 2730 and 2850; SEQ ID NO: 2731 and 2851; SEQ ID NO: 2732 and 2852; SEQ ID NO: 2733 and 2853; SEQ ID NO: 2734 and 2854; SEQ ID NO: 2735 and 2855; SEQ ID NO: 2736 and 2856; SEQ ID NO: 2737 and 2857; SEQ ID NO: 2738 and 2858; SEQ ID NO: 2739 and 2859; SEQ ID NO: 2740 and 2860; SEQ ID NO: 2741 and 2861; SEQ ID NO: 2742 and 2862; SEQ ID NO: 2743 and 2863; SEQ ID NO: 2744 and 2864; SEQ ID NO: 2745 and 2865; SEQ ID NO: 2746 and 2866; SEQ ID NO: 2747 and 2867; SEQ ID NO: 2748 and 2868; SEQ ID NO: 2749 and 2869; SEQ ID NO: 2750 and 2870; SEQ ID NO: 2751 and 2871; SEQ ID NO: 2752 and 2872; SEQ ID NO: 2753 and 2873; SEQ ID NO: 2754 and 2874; SEQ ID NO: 2755 and 2875; SEQ ID NO: 2756 and 2876; SEQ ID NO: 2757 and 2877; SEQ ID NO: 2758 and 2878; SEQ ID NO: 2759 and 2879; SEQ ID NO: 2760 and 2880; SEQ ID NO: 2761 and 2881; SEQ ID NO: 2762 and 2882; SEQ ID NO: 2763 and 2883; SEQ ID NO: 2764 and 2884; SEQ ID NO: 2765 and 2885; SEQ ID NO: 2766 and 2886; SEQ ID NO: 2767 and 2887; SEQ ID NO: 2768 and 2888; SEQ ID NO: 2769 and 2889; SEQ ID NO: 2770 and 2890; SEQ ID NO: 2771 and 2891; SEQ ID NO: 2772 and 2892; SEQ ID NO: 2773 and 2893; SEQ ID NO: 2774 and 2894; SEQ ID NO: 2775 and 2895; SEQ ID NO: 2776 and 2896; SEQ ID NO: 2777 and 2897; SEQ ID NO: 2778 and 2898; SEQ ID NO: 2779 and 2899; SEQ ID NO: 2960 and 3340; SEQ ID NO: 2961 and 3341; SEQ ID NO: 2962 and 3342; SEQ ID NO: 2963 and 3343; SEQ ID NO: 2964 and 3344; SEQ ID NO: 2965 and 3345; SEQ ID NO: 2966 and 3346; SEQ ID NO: 2967 and 3347; SEQ ID NO: 2968 and 3348; SEQ ID NO: 2969 and 3349; SEQ ID NO: 2970 and 3350; SEQ ID NO: 2971 and 3351; SEQ ID NO: 2972 and 3352; SEQ ID NO: 2973 and 3353; SEQ ID NO: 2974 and 3354; SEQ ID NO: 2975 and 3355; SEQ ID NO: 2976 and 3356; SEQ ID NO: 2977 and 3357; SEQ ID NO: 2978 and 3358; SEQ ID NO: 2979 and 3359; SEQ ID NO: 2980 and 3360; SEQ ID NO: 2981 and 3361; SEQ ID NO: 2982 and 3362; SEQ ID NO: 2983 and 3363; SEQ ID NO: 2984 and 3364; SEQ ID NO: 2985 and 3365; SEQ ID NO: 2986 and 3366; SEQ ID NO: 2987 and 3367; SEQ ID NO: 2988 and 3368; SEQ ID NO: 2989 and 3369; SEQ ID NO: 2990 and 3370; SEQ ID NO: 2991 and 3371; SEQ ID NO: 2992 and 3372; SEQ ID NO: 2993 and 3373; SEQ ID NO: 2994 and 3374; SEQ ID NO: 2995 and 3375; SEQ ID NO: 2996 and 3376; SEQ ID NO: 2997 and 3377; SEQ ID NO: 2998 and 3378; SEQ ID NO: 2999 and 3379; SEQ ID NO: 3000 and 3380; SEQ ID NO: 3001 and 3381; SEQ ID NO: 3002 and 3382; SEQ ID NO: 3003 and 3383; SEQ ID NO: 3004 and 3384; SEQ ID NO: 3005 and 3385; SEQ ID NO: 3006 and 3386; SEQ ID NO: 3007 and 3387; SEQ ID NO: 3008 and 3388; SEQ ID NO: 3009 and 3389; SEQ ID NO: 3010 and 3390; SEQ ID NO: 3011 and 3391; SEQ ID NO: 3012 and 3392; SEQ ID NO: 3013 and 3393; SEQ ID NO: 3014 and 3394; SEQ ID NO: 3015 and 3395; SEQ ID NO: 3016 and 3396; SEQ ID NO: 3017 and 3397; SEQ ID NO: 3018 and 3398; SEQ ID NO: 3019 and 3399; SEQ ID NO: 3020 and 3400; SEQ ID NO: 3021 and 3401; SEQ ID NO: 3022 and 3402; SEQ ID NO: 3023 and 3403; SEQ ID NO: 3024 and 3404; SEQ ID NO: 3025 and 3405; SEQ ID NO: 3026 and 3406; SEQ ID NO: 3027 and 3407; SEQ ID NO: 3028 and 3408; SEQ ID NO: 3029 and 3409; SEQ ID NO: 3030 and 3410; SEQIDNO: 3031 and 3411; SEQIDNO: 3032 and 3412; SEQIDNO: 3033 and 3413; SEQ ID NO: 3034 and 3414; SEQ ID NO: 3035 and 3415; SEQ ID NO: 3036 and 3416; SEQ ID NO: 3037 and 3417; SEQIDNO: 3038 and 3418; SEQIDNO: 3039 and 3419; SEQIDNO: 3040 and 3420; SEQIDNO: 3041 and 3421; SEQIDNO: 3042 and 3422; SEQIDNO: 3043 and 3423; SEQ ID NO: 3044 and 3424; SEQ ID NO: 3045 and 3425; SEQ ID NO: 3046 and 3426; SEQ ID NO: 3047 and 3427; SEQ ID NO: 3048 and 3428; SEQ ID NO: 3049 and 3429; SEQ ID NO: 3050 and 3430; SEQIDNO: 3051 and 3431; SEQIDNO: 3052 and 3432; SEQIDNO: 3053 and 3433; SEQ ID NO: 3054 and 3434; SEQ ID NO: 3055 and 3435; SEQ ID NO: 3056 and 3436; SEQ ID NO: 3057 and 3437; SEQ ID NO: 3058 and 3438; SEQ ID NO: 3059 and 3439; SEQ ID NO: 3060 and 3440; SEQIDNO: 3061 and 3441; SEQIDNO: 3062 and 3442; SEQIDNO: 3063 and 3443; SEQ ID NO: 3064 and 3444; SEQ ID NO: 3065 and 3445; SEQ ID NO: 3066 and 3446; SEQ ID NO: 3067 and 3447; SEQ ID NO: 3068 and 3448; SEQ ID NO: 3069 and 3449; SEQ ID NO: 3070 and 3450; SEQIDNO: 3071 and 3451; SEQIDNO: 3072 and 3452; SEQIDNO: 3073 and 3453; SEQ ID NO: 3074 and 3454; SEQ ID NO: 3075 and 3455; SEQ ID NO: 3076 and 3456; SEQ ID NO: 3077 and 3457; SEQ ID NO: 3078 and 3458; SEQ ID NO: 3079 and 3459; SEQ ID NO: 3080 and 3460; SEQIDNO: 3081 and 3461; SEQIDNO: 3082 and 3462; SEQIDNO: 3083 and 3463; SEQ ID NO: 3084 and 3464; SEQ ID NO: 3085 and 3465; SEQ ID NO: 3086 and 3466; SEQ ID NO: 3087 and 3467; SEQ ID NO: 3088 and 3468; SEQ ID NO: 3089 and 3469; SEQ ID NO: 3090 and 3470; SEQIDNO: 3091 and 3471; SEQIDNO: 3092 and 3472; SEQIDNO: 3093 and 3473; SEQ ID NO: 3094 and 3474; SEQ ID NO: 3095 and 3475; SEQ ID NO: 3096 and 3476; SEQ ID NO: 3097 and 3477; SEQ ID NO: 3098 and 3478; SEQ ID NO: 3099 and 3479; SEQ ID NO: 3100 and 3480; SEQIDNO: 3101 and 3481; SEQIDNO: 3102 and 3482; SEQIDNO: 3103 and 3483; SEQ ID NO: 3104 and 3484; SEQ ID NO: 3105 and 3485; SEQ ID NO: 3106 and 3486; SEQ ID NO: 3107 and 3487; SEQIDNO: 3108 and 3488; SEQIDNO: 3109 and 3489; SEQIDNO: 3110 and 3490; SEQIDNO: 3111 and 3491; SEQ ID NO: 3112 and 3492; SEQ ID NO: 3113 and 3493; SEQIDNO: 3114 and 3494; SEQIDNO: 3115 and 3495; SEQIDNO: 3116 and 3496; SEQ ID NO: 3117 and 3497; SEQIDNO: 3118 and 3498; SEQIDNO: 3119 and 3499; SEQIDNO: 3120 and 3500; SEQIDNO: 3121 and 3501; SEQ ID NO: 3122 and 3502; SEQ ID NO: 3123 and 3503; SEQIDNO: 3124 and 3504; SEQIDNO: 3125 and 3505; SEQIDNO: 3126 and 3506; SEQ ID NO: 3127 and 3507; SEQIDNO: 3128 and 3508; SEQIDNO: 3129 and 3509; SEQIDNO: 3130 and 3510; SEQIDNO: 3131 and 3511; SEQ ID NO: 3132 and 3512; SEQ ID NO: 3133 and 3513; SEQ ID NO: 3134 and 3514; SEQ ID NO: 3135 and 3515; SEQ ID NO: 3136 and 3516; SEQ ID NO: 3137 and 3517; SEQ ID NO: 3138 and 3518; SEQ ID NO: 3139 and 3519; SEQ ID NO: 3140 and 3520; SEQ ID NO: 3141 and 3521; SEQ ID NO: 3142 and 3522; SEQ ID NO: 3143 and 3523; SEQ ID NO: 3144 and 3524; SEQ ID NO: 3145 and 3525; SEQ ID NO: 3146 and 3526; SEQ ID NO: 3147 and 3527; SEQ ID NO: 3148 and 3528; and SEQ ID NO: 3149 and 3529.
In another aspect, the instant disclosure provides a double stranded ribonucleic acid (dsRNA) agent for inhibiting expression of human solute carrier family 6 member 19 (SLC6A19) in a cell, where the dsRNA agent includes a sense strand and an antisense strand forming a double stranded region, where the sense strand includes at least 15 contiguous nucleotides differing by no more than 0, 1, 2, or 3 nucleotides from any one of the nucleotide sequences of nucleotides 3 to 25, nucleotides 8 to 30, nucleotides 13 to 35, nucleotides 38 to 60, nucleotides 43 to 65, nucleotides 48 to 70, nucleotides 93 to 115, nucleotides 98 to 120, nucleotides 103 to 125, nucleotides 108 to
130, nucleotides 111 to 133, nucleotides 113 to 135, nucleotides 118 to 140, nucleotides 214 to 236, nucleotides 219 to 241, nucleotides 244 to 266, nucleotides 249 to 271, nucleotides 252 to 274, nucleotides 252 to 274, nucleotides 253 to 275, nucleotides 254 to 276, nucleotides 257 to 286, nucleotides 259 to 281, nucleotides 264 to 286, nucleotides 289 to 311, nucleotides 294 to 316, nucleotides 339 to 361, nucleotides 397 to 419, nucleotides 460 to 480, nucleotides 461 to 481, nucleotides 481 to 503, nucleotides 486 to 508, nucleotides 491 to 513, nucleotides 557 to 579, nucleotides 561 to 583, nucleotides 562 to 584, nucleotides 564 to 586, nucleotides 566 to 586, nucleotides 567 to 589, nucleotides 616 to 638, nucleotides 621 to 643, nucleotides 626 to 648, nucleotides 670 to 692, nucleotides 671 to 691, nucleotides 672 to 694, nucleotides 674 to 694, nucleotides 675 to 697, nucleotides 680 to 702, nucleotides 685 to 707, nucleotides 690 to 712, nucleotides 695 to 717, nucleotides 700 to 722, nucleotides 705 to 727, nucleotides 710 to 732, nucleotides 715 to 737, nucleotides 720 to 742, nucleotides 725 to 747, nucleotides 730 to 752, nucleotides 735 to 757, nucleotides 740 to 762, nucleotides 745 to 767, nucleotides 750 to 772, nucleotides 751 to 776, nucleotides 755 to 777, nucleotides 760 to 782, nucleotides 765 to 787, nucleotides 770 to 792, nucleotides 775 to 797, nucleotides 798 to 823, nucleotides 800 to 822, nucleotides 825 to 847, nucleotides 890 to 912, nucleotides 895 to 917, nucleotides 900 to 922, nucleotides 905 to 927, nucleotides 910 to 932, nucleotides 911 to 937, nucleotides 915 to 937, nucleotides 972 to 994, nucleotides 973 to 995, nucleotides 977 to ; .000, nucleotides 977 to 999, nucleotides 978 to 1000, nucleotides 983 to 1005, nucleotides 1035 ) 1057, nucleotides 1058 to 1080, nucleotides 1059 to 1081, nucleotides 1060 to 1082, nucleotides 1063 to 1082, nucleotides 1065 to 1087, nucleotides 1068 to 1087, nucleotides 1070 to 1094, nucleotides 1070 to 1092, nucleotides 1074 to 1096, nucleotides 1075 to 1097, nucleotides 1077 to 1096, nucleotides 1080 to 1102, nucleotides 1085 to 1107, nucleotides 1086 to 1106, nucleotides 1090 to 1112, nucleotides 1095 to 1117, nucleotides 1100 to 1122, nucleotides 1105 to 1127, nucleotides 1110 to 1132, nucleotides 1115 to 1137, nucleotides 1115 to 1137, nucleotides 1117 to 1139, nucleotides 1177 to 1199, nucleotides 1182 to 1204, nucleotides 1187 to 1209, nucleotides 1191 to 1214, nucleotides 1192 to 1214, nucleotides 1195 to 1217, nucleotides 1234 to 1256, nucleotides 1239 to 1261, nucleotides 1242 to 1264, nucleotides 1244 to 1266, nucleotides 1247 to 1268, nucleotides 1249 to 1271, nucleotides 1251 to 1270, nucleotides 1254 to 1276, nucleotides 1259 to 1281, nucleotides 1263 to 1282, nucleotides 1264 to 1286, nucleotides 1266 to 1285, nucleotides 1269 to 1291, nucleotides 1326 to 1348, nucleotides 1331 to 1353, nucleotides 1334 to 1361, nucleotides 1334 to 1356, nucleotides 1334 to 1356, nucleotides 1335 to 1357, nucleotides 1336 to 1358, nucleotides 1337 to 1359, nucleotides 1337 to 1359, nucleotides 1338 to 1360, nucleotides 1340 to 1362, nucleotides 1341 to 1363, nucleotides 1346 to 1368, nucleotides 1351 to 1373, nucleotides 1353 to 1372, nucleotides 1377 to 1399, nucleotides 1382 to 1404, nucleotides 1383 to 1403, nucleotides 1387 to 1409, nucleotides 1392 to 1414, nucleotides 1397 to 1419, nucleotides 1402 to 1424, nucleotides 1407 to 1429, nucleotides 1412 to 1434, nucleotides 1417 to 1439, nucleotides 1456 to 1478, nucleotides 1458 to 1477, nucleotides 1481 to 1503, nucleotides 1486 to 1508, nucleotides 1491 to 1513, nucleotides 1496 to 1518, nucleotides 1521 to 1543, nucleotides 1568 to 1590, nucleotides 1569 to 1591, nucleotides 1574 to 1596, nucleotides 1578 to 1621, nucleotides 1579 to 1601, nucleotides 1583 to 1605, nucleotides 1583 to 1605, nucleotides 1584 to 1606, nucleotides 1584 to 1606, nucleotides 1585 to 1607, nucleotides 1585 to 1607, nucleotides 1587 to 1609, nucleotides 1588 to 1610, nucleotides 1589 to 1611, nucleotides 1594 to 1616, nucleotides 1595 to 1617, nucleotides 1599 to 1621, nucleotides 1601 to 1620, nucleotides 1602 to 1624, nucleotides 1642 to 1664, nucleotides 1749 to 1775, nucleotides 1750 to 1772, nucleotides 1755 to 1777, nucleotides 1757 to 1779, nucleotides 1758 to 1780, nucleotides 1760 to 1782, nucleotides 1761 to 1792, nucleotides 1764 to 1783, nucleotides 1765 to 1787, nucleotides 1770 to 1792, nucleotides 1809 to 1831, nucleotides 1810 to 1829, nucleotides 1812 to 1832, nucleotides 1814 to 1836, nucleotides 1852 to 1881, nucleotides 1853 to 1875, nucleotides 1854 to 1876, nucleotides 1858 to 1880, nucleotides 2168 to 2188, nucleotides 2333 to 2353, nucleotides 2454 to 2474, nucleotides 2507 to 2527, nucleotides 2593 to 2613, nucleotides 3244 to 3264, nucleotides 3288 to 3308, nucleotides 3304 to 3324, nucleotides 3332 to 3352, nucleotides 3333 to 3353, nucleotides 3501 to 3523, nucleotides 3595 to 3615, nucleotides 3595 to 3617, nucleotides 3643 to 3671, nucleotides 3646 to 3668, nucleotides 3646 to 3668, nucleotides 3647 to 3669, nucleotides 3648 to 3668, nucleotides 3648 to 3670, nucleotides 3648 to 3670, nucleotides 3649 to 3671, nucleotides 3651 to 3670, nucleotides 3651 to 3673, nucleotides 3651 to 3673, nucleotides 3652 to 3671, nucleotides 3712 to 3733, nucleotides 3713 to 3735, nucleotides 3763 to 3786, nucleotides 3763 to 3785, nucleotides 3764 to 3786, nucleotides 3791 to 3811, nucleotides 3796 to 3816, nucleotides 3801 to 3829, nucleotides 3801 to 3820, nucleotides 3802 to 3821, nucleotides 3804 to 3826, nucleotides 3809 to 3831, nucleotides 3810 to 3832, nucleotides 3812 to 3833, nucleotides 3812 to 3834, nucleotides 3813 to 3835, nucleotides 3814 to 3833, nucleotides 3814 to 3836, nucleotides 3814 to 3836, nucleotides 3839 to 3859, nucleotides 3840 to 3862, nucleotides 3902 to 3924, nucleotides 3965 to 3987, nucleotides 3995 to 4015, nucleotides 3998 to 4020, nucleotides 4100 to 4124, nucleotides 4102 to 4124, nucleotides 4141 to 4163, nucleotides 4141 to 4163, nucleotides 4142 to 4164, nucleotides 4143 to 4165, nucleotides 4144 to 4166, nucleotides 4146 to 4168, nucleotides 4150 to 4172, nucleotides 4151 to 4173, nucleotides 4192 to 4214, nucleotides 4197 to 4219, nucleotides 4201 to 4223, nucleotides 4265 to 4287, nucleotides 4270 to 4292, nucleotides 4318 to 4338, nucleotides 4319 to 4341, nucleotides 4329 to 4349, nucleotides 4348 to 4386, nucleotides 4349 to 4371, nucleotides 4350 to 4369, nucleotides 4350 to 4372, nucleotides 4350 to 4372, nucleotides 4351 to 4370, nucleotides 4352 to 4371, nucleotides 4352 to 4374, nucleotides 4353 to4372, nucleotides 4353 to 4375, nucleotides 4354 to 4376, nucleotides 4359 to 4381, nucleotides 4361 to 4383, nucleotides 4362 to 4384, nucleotides 4362 to 4384, nucleotides 4364 to 4386, nucleotides 4365 to 4387, nucleotides 4395 to 4415, nucleotides 4396 to 4416, nucleotides 4409 to 4431, nucleotides 4410 to 4432, nucleotides 4412 to 4434, nucleotides 4637 to 4657, nucleotides 4638 to 4658, nucleotides 4640 to 4660, nucleotides 5103 to 5123, nucleotides 5124 to 5144, nucleotides 5126 to 5146, or nucleotides 5127 to 5147 of SEQ ID NO: 1, and the antisense strand includes at least 19 contiguous nucleotides from the corresponding nucleotide sequence of SEQ ID NO: 2.
In certain embodiments, the double stranded region is 17-30 nucleotide pairs in length; the double stranded region is 17-25 nucleotide pairs in length; the double stranded region is 17-23 nucleotide pairs in length; each strand is independently no more than 30 nucleotides in length; the sense strand is 21 nucleotides in length and the antisense strand is 23 nucleotides in length; the region of complementarity is at least 17 nucleotides in length, optionally where the region of complementarity is between 19 and 23 nucleotides in length, optionally where the region of complementarity is 19 nucleotides in length; and/or at least one strand includes a 3’ overhang of at least 2 nucleotides.
In one aspect, the disclosure provides an exon skipping antisense oligonucleotide including any of SEQ ID NOs: 2542-2719.
In a related embodiment, the exon skipping antisense oligonucleotide has at least one modified internucleoside linkage, sugar moiety, or nucleobase. Optionally, the modified internucleoside linkage includes a phosphorothioate linkage. In certain embodiments, the at least one modified intemucleoside linkage, sugar moiety, or nucleobase includes a modification selected from the following: a deoxy -nucleoside, a 3 ’-terminal deoxy-thymine (dT) nucleoside, a 2'-O- methyl (2'-0Me) modified nucleoside, a 2'-fluoro (2'-F) modified nucleoside, a 2 -deoxy -modified nucleoside, a locked nucleotide (LNA), an unlocked nucleoside, a conformationally restricted nucleoside, a constrained ethyl nucleoside, an abasic nucleoside, a 2'-O,4'-C-ethylene-bridged nucleic acid (ENA), a 2’-amino-modified nucleoside, a 2’-O-allyl-modified nucleoside, 2’-C- alkyl-modified nucleoside, 2’-hydroxly-modified nucleoside, a 2’-methoxyethyl (2 -MOE) modified nucleoside, a 2’-O-alkylmodified nucleoside, a morpholino nucleoside, a pbosphoramidate, a non-natural base comprising nucleoside, a tetrahydropyran modified nucleoside, a 1,5-anhydrohexitol modified nucleoside, a cyclohexenyl modified nucleoside, a nucleotide comprising a 5'-pbosphorothioate group, a nucleotide comprising a 5'- methyiphosphonate group, a nucleotide comprising a 5’ phosphate or 5’ phosphate mimic, a nucleotide comprising vinyl phosphonate, a nucleoside comprising adenosine-glycol nucleic acid (GNA), a nucleoside comprising thymidine-glycol nucleic acid (GNA) S-Isomer, a nucleotide comprising 2-hydroxymethyl-tetrahydrofurane-5-phosphate, a nucleotide comprising 2 ’deoxy thymidine-3’ phosphate, a nucleotide comprising 2 ’-deoxy guanosine- 3 ’-phosphate, and a terminal nucleoside linked to a cholesteryl derivative and a dodecanoic acid bisdecylamide group; and combinations thereof. Optionally, the antisense compound, dsRNA agent, or exon skipping antisense oligonucleotide includes one or more 2’-methoxyethyl (2'-M0E) modified nucleosides. In certain embodiments, all nucleosides of the exon skipping antisense oligonucleotide are 2’ - methoxyethyl (2'-M0E) modified nucleosides. In some embodiments, the ASO, dsRNA agent, and/or exon skipping antisense oligonucleotide includes at least one modified nucleotide. Optionally, substantially all of the nucleotides of the sense strand of the dsRNA agent; or substantially all of the nucleotides of the antisense strand of the antisense compound or the dsRNA agent include a modification; or substantially all of the nucleotides of the sense strand and substantially all of the nucleotides of the antisense strand of the dsRNA agent include a modification.
In certain embodiments, all of the nucleotides of the sense strand of the dsRNA agent include a modification; all of the nucleotides of the antisense strand of the ASO (ASOs including exon skipping antisense oligonucleotides) or the dsRNA agent include a modification; or all of the nucleotides of the sense strand and all of the nucleotides of the antisense strand of the dsRNA agent include a modification.
In one embodiment, at least one of the modified nucleotides is a deoxy-nucleotide, a 3’- terminal deoxy-thymine (dT) nucleotide, a 2'-O-methyl modified nucleotide, a 2'-fluoro modified nucleotide, a 2'-deoxy-modified nucleotide, a locked nucleotide, an unlocked nucleotide, a conformationally restricted nucleotide, a constrained ethyl nucleotide, an abasic nucleotide, a 2’- amino-modified nucleotide, a 2’ -O-allyl -modified nucleotide, 2’-C-alkyl-modified nucleotide, 2’- hydroxly-modified nucleotide, a 2’ -methoxyethyl modified nucleotide, a 2 ’-O-alkyl -modified nucleotide, a morpholino nucleotide (e g., a phosphorodiamidate morpholino nucleic acid (PMO)), a phosphoramidate, a non-natural base including nucleotide, a tetrahydropyran modified nucleotide, a 1,5-anhydrohexitol modified nucleotide, a cyclohexenyl modified nucleotide, a nucleotide including a phosphorothioate group, a nucleotide including a phosphoryl guanidine- based backbone, a nucleotide including a methylphosphonate group, a nucleotide including a 5’- phosphate, a nucleotide including a 5 ’-phosphate mimic, a thermally destabilizing nucleotide, a glycol modified nucleotide (GNA), and/or a 2-O-(N-methylacetamide) modified nucleotide; and/or combinations thereof.
In some embodiments, modifications on the nucleotides are selected from among the following: LNA, HNA, CeNA, 2'-methoxyethyl, 2'-O-alkyl, 2'-O-allyl, 2'-C- allyl, 2'-fluoro, 2'- deoxy, 2’-hydroxyl, and glycol; and combinations thereof; a C7-modified deaza-adenine, a C7- modified deaza-guanosine, a C5-modified cytosine, a C5-modified uridine, Nl-methyl- pseudouridine (mly), 1-ethyl-pseudouridine (el\p), 5-methoxy-uridine (mo5U), 5-methyl- cytidine (m5C), pseudouridine (y), 5-methoxymethyl uridine, 5-methylthio uridine, 1- methoxymethyl pseudouridine, 5-methyl cytidine, 5-methoxy cytidine, or any combination thereof; a phosphorothioate (PS) modification, a phosphoryl guanidine (PN) modification, a borano-phosphate modification, an alkyl phosphonate nucleic acid (phNA), a peptide nucleic acid (PNA), or any combination thereof; a deoxyribonucleic acid (DNA), optionally where the DNA is or includes a DNA analog, optionally where the DNA analog includes one or more morpholino subunits linked together by phosphorus-containing linkage(s), optionally where the DNA analog is or includes a phosphorodiamidate morpholino nucleic acid (PMO), optionally where the PMO includes about 12-40 nucleotides; a peptide nucleic acid (PNA) modification; and/or one or more modification to a 5’ end of the antisense compound or dsRNA agent, optionally where the modification to the 5' end is a 5’ amino modification.
In certain embodiments, at least one of the modified nucleotides is selected from the following: a deoxy-nucleotide, a 2'-O-methyl modified nucleotide, a 2'-fluoro modified nucleotide, a 2'-deoxy-modified nucleotide, a glycol modified nucleotide (GNA), and/or a vinyl-phosphonate nucleotide; and/or combinations thereof.
In some embodiments, at least one of the modified nucleotides is selected from the following: a deoxy-nucleotide, a 2'-O-methyl modified nucleotide, and/or a 2'-fluoro modified nucleotide; and/or combinations thereof.
In an embodiment, the ASO, dsRNA agent, or exon skipping antisense oligonucleotide of the disclosure includes one or more deoxyribonucleotides. Optionally, the antisense compound or the exon skipping antisense oligonucleotide is a component of a heteroduplex oligonucleotide (HDO) and/or the dsRNA agent of the disclosure is a HDO.
In one embodiment, the modulatory nucleic acid (e.g., ASO or dsRNA agent) is directly or indirectly conjugated to a targeting moiety. Optionally, the ASO or dsRNA agent and the targeting moiety are indirectly conjugated by way of a linker.
In some embodiments, the targeting moiety specifically binds a cell surface factor. Optionally, the targeting moiety specifically binds a kidney cell surface factor.
In certain embodiments, the cell surface factor is internalized when bound by the targeting moiety.
In one embodiment, the kidney cell surface factor is a receptor. Optionally, the kidney cell surface factor is megalin or cubilin. In some embodiments, the targeting moiety is chosen from: a polypeptide, an aminoglycoside, an endogenous ligand (e.g., a ligand disclosed in Table 1 or Table 2), a xenobiotic, an antibody or a fragment thereof, an aptamer, a small molecule, or any combination thereof. Optionally, the targeting moiety is or includes an endogenous ligand, e.g., a ligand disclosed in Table 1 or Table 2.
In one embodiment, the targeting moiety is or includes a vitamin. Optionally, the targeting moiety is or includes a vitamin provided in Table 1. Optionally, the vitamin is or includes vitamin B12.
In some embodiments, the targeting moiety is or includes a polypeptide. Optionally, the polypeptide is chosen from: a fragment of receptor associated protein (RAP), a peptide derived from a radiopharmaceutical conjugate such as ocreotide, ocreotate, exendin, minigastrin, and/or neurotensin; or any combination thereof. Optionally, the polypeptide includes a RAP fragment, or a variant thereof. Optionally, the RAP fragment includes a polypeptide having residues 219-323 of RAP.
In certain embodiments, the polypeptide is or includes a peptide derived from a radiopharmaceutical conjugate such as ocreotide, ocreotate, exendin, minigastrin, and/or neurotensin.
In related embodiments, the polypeptide is or includes a knotted peptide.
In one embodiment, the targeting moiety is or includes an aminoglycoside. Optionally, the aminoglycoside is chosen from one or more, or all of: streptomycin, neomycin, kanamycin, paromomycin, gentamicin, G-418 (geneticin) ELX-202, tobramycin, amikacin, netilmicin, spectinomycin, sisomicin, dibekacin, isepamicin, framycetin, paromomycin, apramycin, fradiomycin, arbekacin, plazomicin, or a derivative, or a fragment, or a variant thereof.
In another embodiment, the targeting moiety is or includes a xenobiotic. Optionally, the xenobiotic is or includes polymixin, aprotinin, trichosanthin, or any combination thereof.
In certain embodiments, the targeting moiety is or includes an antibody or a fragment thereof, optionally where the antibody or fragment thereof selectively binds Megalin, Cubilin, or both. In a related embodiment, the antibody or fragment thereof specifically binds Megalin. In another related embodiment, the antibody or fragment thereof specifically binds Cubilin.
In one embodiment, the antibody or fragment thereof is a bispecific antibody or a multispecific antibody. In some embodiments, the antibody includes one or more modifications of an Fc domain, e.g., an Fc variant.
In certain embodiments, the ASO or dsRNA agent is characterized in that when delivered to a cell expressing the SLC6A 19 target, reduced expression and/or activity of the SLC6A19 target is observed as compared to a cell which has not been delivered the anti sense compound or dsRNA agent or to a cell which does not express the SLC6A19 target.
In one embodiment, the linker is a cleavable linker. Optionally, the linker becomes cleaved when exposed to a cell-internal environment.
In another embodiment, the ASO, dsRNA agent, or exon skipping antisense oligonucleotide includes one or more extended nucleic acid ("exNA") modifications. Optionally, the one or more exNA modification(s) is/are positioned at or near a 3'-end of the nucleic acid.
In an additional embodiment, the ASO, dsRNA agent, or exon skipping antisense oligonucleotide includes one or more phosphoryl guanidine-containing backbone ("PN backbone") and/or mesyl phosphoramidate modifications.
Another aspect of the instant disclosure provides a pharmaceutical composition including an ASO or dsRNA agent of the disclosure and a pharmaceutically acceptable penetration enhancer, carrier, or diluent. Optionally, the pharmaceutical composition is formulated for intravenous, subcutaneous, intramuscular, parenteral, or oral delivery.
An additional aspect of the instant disclosure provides a method for the prevention, amelioration, or treatment of a kidney disease or condition in a subject, the method involving administration of the ASO, dsRNA or pharmaceutical composition of the instant disclosure to the subject in need of such inters7 ention.
A further aspect of the instant disclosure provides a method for the prevention, amelioration, or treatment of a kidney disease or condition in a subject, the method involving administration of an inhibitory ASO, dsRNA, or exon skipping antisense oligonucleotide of the disclosure to the subject in need of such intervention, where the inhibitory antisense compound, dsRNA, or exon skipping antisense oligonucleotide targets a human SLC6A19 mRNA. Optionally, the SLC6A19 mRNA of SI X) ID NO: 1 is targeted.
In one embodiment, the kidney disease or condition is a glomerular disorder, a renal tubular disorder, Chronic Kidney Disease, other renal disorders, an inborn error of metabolism, a systemic metabolic disorder, a disorder of the thyroid, a disorder of the parathyroid, a disorder of the inner ear, a neurological disorder, or a viral infection, or any combination thereof.
In an embodiment, the kidney disease or condition is phenylketonuria (PKU).
In another embodiment, the kidney disease or condition is urea cycle disorder.
In one embodiment, the glomerular disorder is chosen from: Lupus nephritis, Goodpasture syndrome, IgA nephropathy, Alport syndrome, glomerulosclerosis, diabetic nephropathy, focal segmental glomerulosclerosis, membranous nephropathy, minimal change disease, ApoLl nephropathy, post-infection glomerulonephritis, membranoproliferative glomerulonephritis, mesangioproliferative glomerulonephritis, nephrotic syndrome, nephritic syndrome, Anti-LRP2 nephropathy, C3 glomerulopathy, or any combination thereof.
In an additional embodiment, the renal tubular disorder is chosen from: Fanconi syndrome, cystinuria, Lowe syndrome, Dent syndrome, Light Chain Proximal Tubulopathy, Gitelman syndrome, renal tubular acidosis, nephrogenic diabetes insipidus, Bartter syndrome, Liddle syndrome, hereditary aminoaciduria, hereditary salt wasting disorders, hereditary phosphate wasting disorders, porphyria associated renal disease, nephropathic cystinosis, autosomal dominant tubulointerstitial kidney disease, or any combination thereof.
In another embodiment, the other renal disorders are chosen from: Autosomal dominant polycystic kidney disease (ADPKD), Autosomal recessive polycystic kidney disease (ARPKD), Nephronophthisis, Chronic Kidney Disease (CKD), nephrolithiasis, acute kidney injury, Alagille syndrome, cardiorenal syndrome, renal cell carcinoma, renal osteodystrophy, or any combination thereof.
In a further embodiment, the inborn error of metabolism is chosen from: phenylketonuria, urea cycle disorder, maple syrup urine disease, galactosemia, hereditary tyrosinemia, glutamic academia, isovaleric acidemia, very long/long/medium/short chain acyl-CoA dehydrogenase deficiency, methylmalonic academia, primary hyperoxaluria, propionic academia, porphyria, Wilson disease, Pyruvate dehydrogenase deficiency, homocystinuria, hereditary fructose intolerance, nonketotic hyperglycinemia, or any combination thereof.
Another aspect of the disclosure provides a method for reducing SLC6A19 rnRNA or B°AT1 polypeptide levels in a cell, the method involving contacting the cell with a modulatory' nucleic acid agent of the instant disclosure in an amount sufficient to reduce SLC6A19 mRNA or B°AT1 polypeptide levels in the cell. In certain embodiments, the cell is chosen from: immune cells; nervous system cells; muscle cells; small intestine cells; colon cells; adipocytes; kidney cells; liver cells; lung cells; splenic cells; stomach cells; esophagus cells; bladder cells; pancreas cells; thyroid cells; salivary gland cells; adrenal gland cells; pituitary gland cells; breast cells; skin cells; ovary cells; uterus cells; placenta cells; prostate cells; parathyroid cells; cells of the inner ear; or testis cells, or any combination thereof, optionally where the cell is a kidney cell, optionally where the kidney cell is a proximal tubular epithelial cell and/or a podocyte.
In another embodiment, the cell is a mammalian cell. Optionally, the cell is a human cell. Optionally, the cell is a human cell of a subject. Optionally, the cell is a human cell of a subject in situ (in vivo)
An additional aspect of the invention provides for use of an ASO, dsRNA or pharmaceutical composition of the instant disclosure in the manufacture of a medicament for the treatment, prevention or amelioration of a kidney disease or condition, where the medicament reduces the expression of a nucleic acid molecule encoding B°AT1.
In certain embodiments, the disease or condition includes a glomerular disorder, a renal tubular disorder, Chronic Kidney Disease, other renal disorders, an inborn error of metabolism, a systemic metabolic disorder, a disorder of the thyroid, a disorder of the parathyroid, a disorder of the inner ear, a neurological disorder, or a viral infection, or any combination thereof.
A further aspect of the instant disclosure provides a kit for performing a method disclosed herein, the kit including a modulatory nucleic acid agent disclosed herein, and instructions for its use. Optionally, the kit also includes a means for administering the modulatory nucleic acid agent to a subject.
Definitions
That the present disclosure may be more readily understood, certain terms are first defined. In addition, it should be noted that whenever a value or range of values of a parameter are recited, it is intended that values and ranges intermediate to the recited values are also intended to be part of this disclosure.
The articles “a” and “an” are used herein to refer to one or to more than one (i.e., to at least one) of the grammatical object of the article. By way of example, “an element” means one element or more than one element, e.g., a plurality of elements. The term "including" is used herein to mean, and is used interchangeably with, the phrase "including but not limited to".
The term "or" is used herein to mean, and is used interchangeably with, the term "and/or," unless context clearly indicates otherwise.
The term “about” is used herein to mean within the typical ranges of tolerances in the art. For example, “about” can be understood as about 2 standard deviations from the mean. In certain embodiments, about means ±10%. In certain embodiments, about means ±5%. When about is present before a series of numbers or a range, it is understood that “about” can modify each of the numbers in the series or range.
The term “at least” prior to a number or series of numbers is understood to include the number adjacent to the term “at least”, and all subsequent numbers or integers that could logically be included, as clear from context. For example, the number of nucleotides in a nucleic acid molecule must be an integer. For example, “at least 18 nucleotides of a 21 nucleotide nucleic acid molecule” means that 18, 19, 20, or 21 nucleotides have the indicated property. When at least is present before a series of numbers or a range, it is understood that “at least” can modify each of the numbers in the series or range.
As used herein, “no more than” or “less than” is understood as the value adjacent to the phrase and logical lower values or integers, as logical from context, to zero. For example, a duplex with an overhang of “no more than 2 nucleotides” has a 2, 1, or 0 nucleotide overhang. When “no more than” is present before a series of numbers or a range, it is understood that “no more than” can modify each of the numbers in the series or range. As used herein, ranges include both the upper and lower limit.
As used herein, methods of detection can include determination that the amount of analyte present is below the level of detection of the method.
In the event of a conflict between an indicated target site and the nucleotide sequence for a sense or antisense strand, the indicated sequence takes precedence.
In the event of a conflict between a chemical structure and a chemical name, the chemical structure takes precedence.
The term "SLC6A19" refers to a gene encoding a system B° transmembrane protein that actively transports most neutral amino acids across the apical membrane of epithelial cells, as well as the B°AT1 proteins encoded by that gene. The human SLC6A19 gene region covers approximately 25 kb. The SLC6A19 transcript contains 12 exons, and at least one mRNA isoform (XM_054352501.1) has been identified or otherwise predicted as produced. Nucleotide and amino acid sequences of SLC6A19 may be found, for example, at GenBank Accession No. NM_001003841.3, noting that SLC6A19 variants (abbreviated elsewhere herein to " NM 1003841.3"), including a predicted transcript variant XI found, e.g., at accession number XM_054352501.1, have also been described. XM 045393498.1 is further noted as the Macaca fascicularis SLC6A19 reference sequence
In certain embodiments, antisense compounds, including antisense oligonucleotides and other antisense compounds for use in modulating the expression of nucleic acid molecules encoding SLC6A19 mRNA and/or the sodium-dependent neutral amino acid transporter B°AT1, are disclosed. Without wishing to be bound by theory, SLC6A19/B°AT1 expression modulation is accomplished by providing antisense compounds that hybridize with one or more target nucleic acid molecules encoding SLC6A19/B°AT1.
In some embodiments, double-stranded RNA (dsRNA) agents (e.g., siRNAs) for use in modulating the expression of nucleic acid molecules encoding SLC6A19 mRNA and/or B°AT1, are disclosed. Without wishing to be bound by theory, SLC6A19/B°AT1 expression modulation is accomplished in certain embodiments by providing dsRNA agents that engage the RNA interference machinery (specifically the RNA-induced silencing complex (RISC)) to direct sequence-specific cleavage and degradation of one or more target nucleic acid molecules encoding SLC6A19/B°AT1. In certain other embodiments, dsRNA compositions of the instant disclosure (e.g., heteroduplex oligonucleotides (HDOs), which are known in the art and which include an antisense oligonucleotide strand (typically a "gapmer" antisense oligonucleotide structure) that is duplexed with a complementary RNA oligonucleotide strand) can accomplish expression modulation of a target SLC6A19/B°AT1 nucleic acid via an antisense oligonucleotide (ASO) mechanism that does not involve RISC.
The terms “iRNA”, “RNAi agent,” “iRNA agent,”, “RNA interference agent” as used interchangeably herein, refer to an agent that contains RNA as that term is defined herein, and which mediates the targeted cleavage of an RNA transcript via an RNA-induced silencing complex (RISC) pathway. iRNA directs the sequence-specific degradation of mRNA through a process known as RNA interference (RNAi). The iRNA modulates, e.g., inhibits, the expression of a SLC6A19 gene in a cell, e.g , a cell within a subject, such as a mammalian subject. In one embodiment, an RNAi agent of the present disclosure includes a single stranded RNA that interacts with a target RNA sequence, e.g., a SLC6A19 target mRNA sequence, to direct the cleavage of the target RNA, Without wishing to be bound by theory, it is believed that, long double stranded RNA introduced into cells is broken down into siRNA by a Type III endonuclease known as Dicer (Sharp et al. (2001) Genes Dev.15:485). Dicer, a ribonuclease-III-like enzyme, processes the dsRNA into 19-23 base pair short interfering RNAs with characteristic two base 3' overhangs (Bernstein, et al., (2001) Nature 409:363). The siRNAs are then incorporated into an RNA-induced silencing complex (RISC) where one or more helicases unwind the siRNA duplex, enabling the complementary antisense strand to guide target recognition (Nykanen, et al., (2001) Cell 107:309). Upon binding to the appropriate target mRNA, one or more endonucleases within the RISC cleave the target to induce silencing (Elbashir, et al., (2001) Genes Dev.15: 188). Thus, in one aspect the present disclosure relates to a single stranded RNA (siRNA) generated within a cell and which promotes the formation of a RISC complex to effect silencing of the target gene, i.e., a SLC6A19 gene. Accordingly, the term “siRNA” is also used herein to refer to an iRNA as described above.
In certain embodiments, the modulatory nucleic acid agent may be a single-stranded antisense oligonucleotide (ASO), or may be a single- stranded siRNA (ssRN.Ai), either of which is introduced into a cell or organism to inhibit a target mRNA. Without wishing to be bound by theory, single-stranded RNAi agents bind to the RISC endonuclease, Argonaute 2, which then cleaves the target mRNA Single-stranded siRNAs are generally 15-30 nucleotides and are chemically modified. Design and testing of single-stranded siRNAs are described in U.S. Patent No.8, 101, 348 and in Lima et al., (2012) Cell 150:883-894, the entire contents of each of which are hereby incorporated herein by reference. Any of the antisense nucleotide sequences described herein may be used as a single-stranded siRNA as described herein or as chemically modified by the methods described in Lima et al., (2012) Cell 150:883-894.
In certain embodiments, an “iRNA” for use in the compositions, uses, and methods of the present disclosure is a double stranded RNA and is referred to herein as a “double stranded RNA agent,” “double stranded RNA (dsRNA) molecule,” “dsRNA agent,” or “dsRNA”. The term “dsRNA”, refers to a complex of ribonucleic acid molecules (optionally including modified nucleotides, as defined herein, in substitution at one, multiple, or all such ribonucleic acids of one or both strands of such dsRNA agent), having a duplex structure comprising two anti-parallel and substantially complementary nucleic acid strands, referred to as having “sense” and “antisense” orientations with respect to a target RNA, i.e., a SLC6A19 gene. In some embodiments of the present disclosure, a double stranded RNA (dsRNA) triggers the degradation of a target RNA, e.g., a SLC6A 19 mRNA, through a post-transcriptional gene-silencing mechanism referred to herein as RNA interference or RNAi.
As used herein, the term “modulatory nucleic acid” refers to antisense oligonucleotides (ASOs), double stranded RNA (dsRNA), exon skipping agent, exon inclusion agent, or other splicing modulator.
As used herein, the term “antisense oligonucleotide (ASO)” refers to an oligonucleotide that is capable of interacting with and/or hybridizing to a pre-mRNA or an mRNA having a complementary nucleotide sequence thereby modifying gene expression.
As used herein, the term “exon skipping” refers to the modification of pre-mRNA splicing by the targeting of splice donor and/or acceptor and branch sites within a pre-mRNA with one or more complementary antisense oligonucleotide(s) (ASOs). By blocking access of a spliceosome to one or more splice donor, acceptor or branch site, an ASO can prevent a splicing reaction thereby causing the exclusion of one or more exons from a fully-processed mRNA. Exon skipping is achieved in the nucleus during the maturation process of pre-mRNAs. It includes the masking of key sequences involved in the splicing of targeted exons by using antisense oligonucleotides (ASO) that are complementary to splice donor/ acceptor, branch-point sequences and/or by overlapping ESE (in exon)/ISE (in intron) within a pre-mRNA.
In general, the majority of nucleotides of each strand of a dsRNA molecule are ribonucleotides, but as described in detail herein, each or both strands can also include one or more non-ribonucleotides, e g., a deoxyribonucleotide or a modified nucleotide. In addition, as used in this specification, an “iRNA”, or an ASO, may include ribonucleotides with chemical modifications; an iRNA, or an ASO, may include substantial modifications at multiple nucleotides.
As used herein, the term “modified nucleotide” refers to a nucleotide having, independently, a modified sugar moiety, a modified internucleotide linkage, or modified nucleobase, or any combination thereof. Thus, the term modified nucleotide encompasses substitutions, additions or removal of, e.g., a functional group or atom, to internucleoside linkages, sugar moieties, or nucleobases. The modifications suitable for use in the agents of the present disclosure include all types of modifications disclosed herein or known in the art Any such modifications, as used in a siRNA type molecule, are encompassed by “iRNA” or “RNAi agent”, or as used in an ASO type molecule, are encompassed by "antisense oligonucleotide" or "ASO", for the purposes of this specification and claims.
In certain embodiments of the instant disclosure, inclusion of a deoxy-nucleotide if present within an RNAi agent, or within an ASO or other modulatory nucleic acid agent, can be considered to constitute a modified nucleotide.
The term “targeting moiety” as used herein, refers to a moiety that, when contacted with a system that includes one or more target cells of interest (e.g., in culture, in a tissue, and/or in an organism) binds specifically with such target cells. In many embodiments, a targeting moiety binds to a cell surface factor (e.g., to a factor that is preferentially or specifically found on surface(s) of such target cells of interest). In some embodiments, binding of a targeting moiety to a cell surface factor results in internalization of a targeting moiety. Typically, a targeting moiety useful in accordance with the present disclosure retains its specific binding character when included in a conjugate agent as described herein; in some embodiments, binding of such a conjugate agent to a relevant cell surface factor results in internalization of a conjugate agent. In some embodiments, a targeting moiety binds specifically to a factor on the surface of kidney cells. In some embodiments, a targeting moiety binds specifically to Cubilin. In some embodiments, a targeting moiety binds specifically to Megalin.
The term “cell surface factor” as used herein, refers to a factor (e.g., that is or comprises a polypeptide) that is present on the surface of cell(s) of interest (e.g., of target cell(s) as described herein which, in many embodiments, may be kidney cells). In some embodiments, a cell surface factor is preferentially present on the surface of target cell(s) (e.g., kidney cells) as compared with cells of one or more other tissues. In some embodiments, a cell surface factor is present on certain non-target cells in addition to target cells. In some embodiments, a cell surface factor is not preferentially or specifically present on relevant target cells of interest. In some embodiments, a cell surface factor is or comprises a receptor. In some embodiments, a cell surface factor is internalized when bound by one or more particular ligands (e.g., with a targeting moiety as described herein). In some embodiments, a cell surface factor may interact with (e.g., bind to, form a complex with, etc.) one or more other components of a cell (e.g., with one or more cell membrane components and/or one or more cell surface components and/or one or more cell-internal components) on whose surface it is found. In some embodiments, a cell surface factor, and/or a particular form or variant thereof, and/or a cell surface factor of any of the foregoing, may be associated with a particular cell state or condition (e.g., stage of development, disease state, etc.).
In some embodiments, a "conjugate agent" has a structure represented by the following formula: (Xnl - Yn2 -Zn3), wherein X is a targeting moiety and nl is an integer (i.e. 1 or greater, typically less than 5); Y is a linker and n2 is 0 or an integer (i.e., 1 or greater, typically less than 5); and Z is a modulatory nucleic acid of the instant disclosure and n3 is an integer (i.e. 1 or greater, typically less than 5); in many embodiments, n2 = nl and/or n2 = n3. In many embodiments, nl and/or n3 is/are 1. In many embodiments, a conjugate agent has a structure represented by the formula (X-Y-Z). In some embodiments, a conjugate agent has a structure represented by a formula of: (X — Y)n-Z, wherein n is an integer greater than 1, and a conjugate agent comprises more than one targeting moiety. In some embodiments, a conjugate agent has structure represented by a formula of: X — (Y-Z)n, wherein n is an integer greater than 1, and a conjugate agent comprises more than one modulatory nucleic acid.
The term “Megalin,” as used herein refers to a receptor which is a member of the low- density lipoprotein receptor (LDLR) family. Megalin is encoded by the LRP2 gene. Amino acid sequences for full length Megalin, and/or for nucleic acids that encode it can be found in a public database such as GenBank, UniProt and Swiss-Prot. For example, the amino acid sequence of human Megalin (SEQ ID NO: 5, for which residues 27-4411 represent an extracellular domain comprising LDL Receptor Class A domains, LDL Receptor Class B domains, and EGF-like domains; residues 4589-4602 represent a DAB2 interaction domain; and residues 4453-4622 represent a cytoplasmic domain comprising NPXY motifs, SH2 binding domains, SH3 binding domains, and proline-rich domains) can be found as UniProt/Swiss-Prot Accession No. P98164 and the nucleic acid sequence encoding human Megalin can be found at Accession No. NM_ 004525.3. Megalin is also known, for example, as Low-density lipoprotein receptor-related protein 2 (LRP2), Glycoprotein 330 (Gp330), Calcium Sensor Protein, or Heymann Nephritis Antigen Homolog. Those skilled in the art will appreciate that the sequence of SEQ ID NO: 5 is exemplary, and certain variations (including, for example, conservative substitutions in SEQ ID NO: 5, as well as codon-optimized variants of associated nucleic acid sequences, etc.) are understood to also be or encode human Megalin. Additionally, those skilled in the art will appreciate that homologs and orthologs of human Megalin are known and/or knowable through the exercise or ordinary skill, for example, based on degree of sequence identity, presence of one or more characteristic sequence elements, and/or one or more shared activities. In some embodiments, Megalin comprises full- length Megalin, or a variant or a fragment thereof. In some embodiments, Megalin that is targeted in accordance with the present disclosure is a Megalin expressed by particular target cell(s) and/or tissue(s) of interest (e.g., in an organism of interest). In some embodiments, a Megalin that is targeted in accordance with the present disclosure is an engineered Megalin. In many embodiments, a Megalin that is targeted in accordance with the present disclosure is present on the surface of target cell(s) of interest (e.g., in target tissue(s) of interest, such as kidney) and that becomes internalized by such cell upon binding of a Megalin binding moiety as described herein. Megalin has been reported to be expressed in one or more of the following tissues and/or cells: immune cells (e.g., bone marrow cells, lymph node cells, thymic cells, peripheral blood mononuclear cells [e.g., myeloid and/or lymphoid cells], erythrocytes, eosinophils, neutrophils, and/or platelets); nervous system cells (e.g., brain tissue, cortex, cerebellum, retinal cells, spinal cord cells, nerve cells, neurons, and/or supporting cells); endothelial cells; muscle (e.g., heart muscle, smooth muscle, and/or skeletal muscle); small intestine; colon; adipocytes; kidney; liver; lung; spleen; stomach; esophagus; bladder; pancreas; thyroid; salivary gland; adrenal gland; pituitary gland; breast; skin; ovary; uterus; placenta; prostate; and testis. Megalin expression is reported to be enriched (e.g., high relative to one or more other tissues) in the following tissues and/or cells in particular: renal tissue, thyroid tissue, parathyroid tissue, cells of the inner ear, and nervous system tissue. Megalin has been specifically reported to be expressed (e.g., at relatively high level(s)) on surfaces of kidney cells such a proximal tubular epithelial cells and podocytes. See Nielsen R. et al. (2016), Kidney Int. 89(1): 58-67.
The term “Megalin-binding moiety” as used herein refers to a moiety that binds to Megalin when contacted therewith. Typically, a Megalin-binding moiety useful in accordance with the present disclosure binds specifically to Megalin under the circumstances of the contacting. In some embodiments, a Megalin-binding moiety is or comprises: a peptide, an aminoglycoside, an endogenous ligand (e.g., a ligand disclosed in Table 1 or an analog or variant thereof), a xenobiotic, an antibody or a fragment thereof, or any combination thereof. In some embodiments, a Megalin-binding moiety is internalized upon binding to Megalin on a cell surface.
The term “Cubilin,” as used herein refers to a receptor encoded by the CUBN gene. Amino acid sequences for full length Cubilin, and/or for nucleic acids that encode it can be found in a public database such as GenBank, UniProt and Swiss-Prot. For example, the amino acid sequence of human Cubilin (SEQ ID NO: 6, for which residues 1 -23 represent a signal peptide; residues 24- 35 represent a propeptide which can be removed in the mature form, and residues 36-3623 represent a mature Cubilin polypeptide) can be found as UniProt/Swiss-Prot Accession No. 060494 and the nucleic acid sequence encoding human Cubilin can be found at Accession No. NM_001081.3. Cubilin is also known, for example, as IFCR, Gp280, Intrinsic Factor-Vitamin B 12 Receptor, MGA1, or IGS1. Those skilled in the art will appreciate that the Cubilin sequence of SEQ ID NO: 6 is exemplary, and certain variations (including, for example, conservative substitutions in SEQ ID NO: 6, codon-optimized variants of Cubilin-encoding nucleic acid sequences, etc.) are understood to also be or encode human Cubilin. Additionally, those skilled in the art will appreciate that homologs and orthologs of human Cubilin are known and/or knowable through the exercise or ordinary skill, for example, based on degree of sequence identity, presence of one or more characteristic sequence elements, and/or one or more shared activities. In some embodiments, Cubilin comprises full-length Cubilin, or a variant or a fragment thereof. In some embodiments, Cubilin that is targeted in accordance with the present disclosure is a Cubilin expressed by particular target cell(s) and/or tissue(s) of interest (e.g., in an organism of interest). In some embodiments, a Cubilin that is targeted in accordance with the present disclosure is an engineered Cubilin. In many embodiments, a Cubilin that is targeted in accordance with the present disclosure is present on the surface of target cell(s) of interest (e.g., in target tissue(s) of interest) and that becomes internalized by such cell upon binding of a Cubilin binding moiety as described herein. Cubilin has been reported to be expressed in one or more of the following tissues and/or cells: immune cells (e.g., bone marrow cells, lymph node cells, thymic cells, peripheral blood mononuclear cells [e.g., myeloid and/or lymphoid cells], erythrocytes, eosinophils, neutrophils, and/or platelets); nervous system (e.g., brain tissue, cortex, cerebellum, retinal cells, spinal cord cells, nerve cells, neurons, and/or supporting cells; endothelial cells; muscle (e.g., heart muscle, smooth muscle, and/or skeletal muscle); small instetine; colon; adipocytes; kidney; liver; lung; spleen; stomach; esophagus; bladder; pancreas; thyroid; salivary gland; adrenal gland; pituitary gland; breast; skin; ovary; uterus; placenta; prostate; and testis. Cubilin expression is reported to be enriched (e.g., high relative to one or more other tissues) in the following tissues and/or cells in particular: renal tissue, thyroid tissue, parathyroid tissue, cells of the inner ear, and nervous system tissue. Cubilin has been specifically reported to be expressed (e.g., at relatively high level(s)) on surfaces of kidney cells such a proximal tubular epithelial cells and podocytes. See Nielsen R. et al. (2016), Kidney hit. 89(l):58-67.
The term “Cubilin-binding moiety” as used herein refers to a moiety that binds to Cubilin when contacted therewith. Typically, a Cubilin-binding moiety useful in accordance with the present disclosure binds specifically to Cubilin under the circumstances of the contacting. In some embodiments, a Cubilin-binding moiety is or comprises a ligand provided in Table 2. In some embodiments, a Cubilin-binding moiety is internalized upon binding to Cubilin on a cell surface.
The phrase “cell associated with a kidney” as used herein refers to a cell that is or can be found in a kidney (e.g., during development, during tissue homeostasis, or in the course of a disease or disorder). In some embodiments, a cell associated with a kidney is also referred to as a "kidney cell" herein. In some embodiments, a cell associated with a kidney includes any one or all of the following cell types: a proximal tubule epithelial cell, a podocyte, a parietal epithelial cell, a mesangial cell, a renal stem cell, an epithelial progenitor cell, a fibroblast, a myo-fibroblast, a pericyte, an ascending loop of Henle cell, a descending loop of Henle cell, a distal tubule cell, a connecting tubule cell, an intercalated cell, a principal cell. Exemplary renal cell populations are provided in Schumacher etal., (2021) npj Regen Med 6, 45, the entire contents of which are hereby incorporated by reference. In some embodiments, a kidney cell is or comprises a cell derived from a kidney, e.g., a kidney tumor cell and/or a metastatic kidney tumor cell.
As used herein, the term “combination therapy” refers to those situations in which a subject is simultaneously exposed to two or more therapeutic regimens (e.g., two or more therapeutic agents). In some embodiments, the two or more regimens may be administered simultaneously; in some embodiments, such regimens may be administered sequentially (e.g., all “doses” of a first regimen are administered prior to administration of any doses of a second regimen); in some embodiments, such agents are administered in overlapping dosing regimens. In some embodiments, “administration” of combination therapy may involve administration of one or more agent(s) or modality(ies) to a subject receiving the other agent(s) or modality(ies) in the combination. For clarity, combination therapy does not require that individual agents be administered together in a single composition (or even necessarily at the same time), although in some embodiments, two or more agents, or active moieties thereof, may be administered together in a combination composition, or even in a combination compound (e.g., as part of a single chemical complex or covalent entity). A composition or method described herein as "comprising" one or more named elements or steps is open-ended, meaning that the named elements or steps are essential, but other elements or steps may be added within the scope of the composition or method. To avoid prolixity, it is also understood that any composition or method described as "comprising" (or which "comprises") one or more named elements or steps also describes the corresponding, more limited composition or method "consisting essentially of (or which "consists essentially of) the same named elements or steps, meaning that the composition or method includes the named essential elements or steps and may also include additional elements or steps that do not materially affect the basic and novel character! stic(s) of the composition or method. It is also understood that any composition or method described herein as "comprising" or "consisting essentially of one or more named elements or steps also describes the corresponding, more limited, and closed-ended composition or method "consisting of (or "consists of) the named elements or steps to the exclusion of any other unnamed element or step. In any composition or method disclosed herein, known or disclosed equivalents of any named essential element or step may be substituted for that element or step.
As used herein, the term “pharmaceutical composition” refers to a composition in which an active agent (e.g., a modulatory nucleic acid agent as disclosed herein) is formulated together with one or more pharmaceutically acceptable carriers. In some embodiments, the active agent is present in unit dose amount appropriate for administration in a therapeutic regimen that shows a statistically significant probability of achieving a predetermined therapeutic effect when administered to a relevant population. In some embodiments, a pharmaceutical composition may be specially formulated for administration in a particular form (e.g., in a solid form or a liquid form), and/or may be specifically adapted for, for example: oral administration (for example, as a drenche [aqueous or non-aqueous solutions or suspensions], tablet, capsule, bolus, powder, granule, paste, etc, which may be formulated specifically for example for buccal, sublingual, or systemic absorption); parenteral administration (for example, by subcutaneous, intramuscular, intravenous or epidural injection as, for example, a sterile solution or suspension, or sustained- release formulation, etc); topical application (for example, as a cream, ointment, patch or spray applied for example to skin, lungs, or oral cavity); intravaginal or intrarectal administration (for example, as a pessary, suppository, cream, or foam); ocular administration; nasal or pulmonary administration, etc. As used herein, the term “subject” refers to an organism, for example, a mammal (e.g., a human, a non-human mammal, a non-human primate, a primate, a laboratory animal, a mouse, a rat, a hamster, a gerbil, a cat, a dog). In some embodiments, a human subject is an adult, adolescent, or pediatric subject. In some embodiments, a subject is suffering from a disease, disorder or condition, e.g., a disease, disorder or condition that can be treated as provided herein. In some embodiments, a subject is susceptible to a disease, disorder, or condition; in some embodiments, a susceptible subject is predisposed to and/or shows an increased risk (as compared to the average risk observed in a reference subject or population) of developing the disease, disorder or condition. In some embodiments, a subject displays one or more symptoms of a disease, disorder or condition. In some embodiments, a subject does not display a particular symptom (e.g., clinical manifestation of disease) or characteristic of a disease, disorder, or condition. In some embodiments, a subject does not display any symptom or characteristic of a disease, disorder, or condition. In some embodiments, a subject is a patient. In some embodiments, a subject is an individual to whom diagnosis and/or therapy is and/or has been administered. In an embodiment, the subject is a human, such as a human being treated or assessed for a disease or disorder that would benefit from reduction in SLC6A19 expression; a human at risk for a disease or disorder that would benefit from reduction in SLC6A19 expression; a human having a disease or disorder that would benefit from reduction in SLC6A19 expression; or human being treated for a disease or disorder that would benefit from reduction in SLC6A19 expression as described herein. In some embodiments, the subject is a female human In other embodiments, the subject is a male human. In one embodiment, the subject is an adult subject. In another embodiment, the subject is a pediatric subject.
As used herein, “complementary” refers to a nucleic acid molecule that can form hydrogen bond(s) with another nucleic acid molecule by either traditional Watson-Crick base pairing or other non-traditional types of pairing (e.g., Hoogsteen or reversed Hoogsteen hydrogen bonding) between complementary nucleosides or nucleotides In reference to the oligonucleotides of the present disclosure, the binding free energy for an antisense oligonucleotide/antisense strand with its complementary sequence is sufficient to allow the relevant function of the oligonucleotide agent to proceed and there is a sufficient degree of complementarity to avoid non-specific binding of the antisense oligonucleotide/antisense strand to non- target, sequences under conditions in which specific binding is desired, i e., under physiological conditions in the case of in vivo therapeutic treatment. Determination of binding free energies for nucleic acid molecules is well known in the art (see e.g., Turner et ah, CSH Symp. Quant. Biol. 1/7 : 123-133 (1987); Frier et al, Proc. Nat. Acad. Sci. USA 83:9373-77 (1986); and Turner et al, J Am. Chem. Soc. 109:3783-3785 (1987)). Thus, “complementary” (or “specifically hybridizable”) are terms that indicate a sufficient degree of complementarity or precise pairing such that stable and specific binding occurs between an antisense oligonucleotide/antisense strand and a pre-mRNA or mRNA target. It is understood in the art that a nucleic acid molecule need not be 100% complementary to a target nucleic acid sequence to be specifically hybridizable. That is, two or more nucleic acid molecules may be less than fully complementary'.
Complementarity is indicated by a percentage of contiguous residues in a nucleic acid molecule that can form hydrogen bonds with a second nucleic acid molecule. For example, if a first nucleic acid molecule has 10 nucleotides and a second nucleic acid molecule has 10 nucleotides, then base pairing of 5, 6, 7, 8, 9, or 10 nucleotides between the first and second nucleic acid molecules represents 50%, 60%, 70%, 80%, 90%, and 100% complementarity, respectively “Perfectly” or “fully” complementary nucleic acid molecules means those in which all the contiguous residues of a first nucleic acid molecule will hydrogen bond with the same number of contiguous residues in a second nucleic acid molecule, wherein the nucleic acid molecules either both have the same number of nucleotides (i.e., have the same length) or the two molecules have different lengths
The term “variant”, as used herein, refers to a molecule or entity (e.g., that are or comprise a nucleic acid, protein, or small molecule) that shows significant structural identity with a reference molecule or entity but differs structurally from the reference molecule or entity, e.g., in the presence or absence or in the level of one or more chemical moieties as compared to the reference molecule or entity. In some embodiments, a “variant” may be referred to as a “derivative”. In some embodiments, a variant differs functionally from its reference molecule or entity. In many embodiments, whether a particular molecule or entity is properly considered to be a “variant” of a reference is based on its degree of structural identity with the reference molecule. As will be appreciated by those skilled in the art, a biological or chemical reference molecule in typically characterized by certain characteristic structural elements. A variant, by definition, is a distinct molecule or entity that shares one or more such characteristic structural elements but differs in at least one aspect from the reference molecule or entity. To give but a few examples, a polypeptide may have a characteristic sequence element comprised of a plurality of amino acids having designated positions relative to one another in linear or three-dimensional space and/or contributing to a particular structural motif and/or biological function; a nucleic acid may have a characteristic sequence element comprised of a plurality of nucleotide residues having designated positions relative to on another in linear or three-dimensional space. In some embodiments, a variant polypeptide or nucleic acid may differ from a reference polypeptide or nucleic acid as a result of one or more differences in amino acid or nucleotide sequence and/or one or more differences in chemical moi eties (e.g., carbohydrates, lipids, phosphate groups) that are covalently components of the polypeptide or nucleic acid (e.g., that are attached to the polypeptide or nucleic acid backbone). In some embodiments, a variant polypeptide or nucleic acid shows an overall sequence identity with a reference polypeptide or nucleic acid that is at least 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, or 99%. In some embodiments, a variant polypeptide or nucleic acid does not share at least one characteristic sequence element with a reference polypeptide or nucleic acid. In some embodiments, a reference polypeptide or nucleic acid has one or more biological activities. In some embodiments, a variant polypeptide or nucleic acid shares one or more of the biological activities of the reference polypeptide or nucleic acid. In some embodiments, a variant polypeptide or nucleic acid lacks one or more of the biological activities of the reference polypeptide or nucleic acid. In some embodiments, a variant polypeptide or nucleic acid shows a reduced level of one or more biological activities as compared to the reference polypeptide or nucleic acid. In some embodiments, a polypeptide or nucleic acid of interest is considered to be a “variant” of a reference polypeptide or nucleic acid if it has an amino acid or nucleotide sequence that is identical to that of the reference but for a small number of sequence alterations at particular positions. Typically, fewer than about 20%, about 15%, about 10%, about 9%, about 8%, about 7%, about 6%, about 5%, about 4%, about 3%, or about 2% of the residues in a variant are substituted, inserted, or deleted, as compared to the reference. In some embodiments, a variant polypeptide or nucleic acid comprises about 10, about 9, about 8, about 7, about 6, about 5, about 4, about 3, about 2, or about 1 substituted residues as compared to a reference. Often, a variant polypeptide or nucleic acid comprises a very small number (e.g., fewer than about 5, about 4, about 3, about 2, or about 1) number of substituted, inserted, or deleted, functional residues (i.e., residues that participate in a particular biological activity) relative to the reference. In some embodiments, a variant polypeptide or nucleic acid comprises not more than about 5, about 4, about 3, about 2, or about 1 addition or deletion, and, in some embodiments, comprises no additions or deletions, as compared to the reference. In some embodiments, a variant polypeptide or nucleic acid comprises fewer than about 25, about 20, about 19, about 18, about 17, about 16, about 15, about 14, about 13, about 10, about 9, about 8, about 7, about 6, and commonly fewer than about 5, about 4, about 3, or about 2 additions or deletions as compared to the reference. In some embodiments, a reference polypeptide or nucleic acid is one found in nature. In some embodiments, a reference polypeptide or nucleic acid is a human polypeptide or nucleic acid.
In some embodiments, modulatory RNA compounds (e.g., inhibitory RNA compounds such as siRNAs, antisense compounds including both inhibitory antisense oligonucleotides and those designed to modulate splicing, etc.) are provided for the prevention, amelioration, and/or treatment of diseases or conditions for which delivery of a nucleic acid molecule capable of selectively and specifically modulating SLC6A19 expression to kidney cells or to cells of other organs of a subject can provide such prevention, amelioration, and/or treatment of the disease(s) or condition(s) in the subject.
.4s used herein, the terms “treating” or “treatment” refer to a beneficial or desired result, such as reducing at least one sign or symptom of a SLC6A19-associated disorder in a subject. Treatment also includes a reduction of one or more sign or symptoms associated with unwanted SLC6A19 and/or B°AT1 expression; diminishing the extent of unwanted SLC6A19 and/or B°AT1 activation or stabilization; amelioration or palliation of unwanted SLC6A19 and/or B°AT1 activation or stabilization. “Treatment” can also mean prolonging survival as compared to expected survival in the absence of treatment. The term “lower” in the context of the level of SLC6A19 and/or B°AT1 in a subject or a disease marker or symptom refers to a statistically significant decrease in such level. The decrease can be, for example, at least 10%, 15%, 20%, 25%, 30%, %, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or more. In certain embodiments, a decrease is at least 20%. In certain embodiments, the decrease is at least 50% in a disease marker, e.g., protein or gene expression level. “Lower” in the context of the level of SLC6A19 and/or B°AT1 in a subject is preferably down to a level accepted as within the range of normal for an individual without such disorder. In certain embodiments, “lower” is the decrease in the difference between the level of a marker or symptom for a subject suffering from a disease and a level accepted within the range of normal for an individual, e.g., the level of decrease in bodyweight between an obese individual and an individual having a weight accepted within the range of normal.
As used herein, “prevention” or “preventing,” when used in reference to a disease, disorder or condition thereof, may be treated or ameliorated by a reduction in expression of a SLC6A19 gene, refers to a reduction in the likelihood that a subject will develop a symptom associated with such a disease, disorder, or condition, e.g., a symptom of unwanted or excessive SLC6A19 and/or B°AT1 expression, such as a metabolic disorder and/or a kidney disease or condition, such as a glomerular disorder, a renal tubular disorder, other renal disorders, an inborn error of metabolism, a systemic metabolic disorder, a disorder of the thyroid, a disorder of the parathyroid, a disorder of the inner ear, a neurological disorder, a viral infection, etc. The likelihood of developing, e g., a kidney disease or condition, is reduced, for example, when an individual having one or more risk factors for a kidney disease or condition either fails to develop the kidney disease or condition or develops the a kidney disease or condition with less severity relative to a population having the same risk factors and not receiving treatment as described herein. The failure to develop a disease, disorder or condition, or the reduction in the development of a symptom associated with such a disease, disorder or condition (e.g., by at least about 10% on a clinically accepted scale for that disease or disorder), or the exhibition of delayed symptoms delayed (e.g., by days, weeks, months or years) is considered effective prevention.
As used herein, the term "SLC6AI9-associated disease” or “SLC6A19-associated disease,” is a disease, disorder or a condition that is caused by, or is associated with, unwanted or excessive SLC6A19 expression. The term "SLC6A19-associated disease” includes a disease, disorder or condition that may be treated or ameliorated by a reduction in SLC6A19 expression. The term SLC6A19-associated disease” includes metabolic disorders, including nephropathy and various other kidney diseases or conditions, such as glomerular disorders, renal tubular disorders, other renal disorders, inborn errors of metabolism, systemic metabolic disorders, disorders of the thyroid, disorders of the parathyroid, disorders of the inner ear, neurological disorders, viral infections, phenylketonuria (PKU) and related aminoacidopathies, etc.
"Therapeutically effective amount," as used herein, is intended to include the amount of a modulatory nucleic acid agent that, when administered to a subject having a SLC6A19-associated disease, is sufficient to effect treatment of the disease (e.g., by diminishing, ameliorating, or maintaining the existing disease or one or more symptoms of disease). The "therapeutically effective amount" may vary depending on the modulatory nucleic acid agent, how the agent is administered, the disease and its severity and the history, age, weight, family history, genetic makeup, the types of preceding or concomitant treatments, if any, and other individual characteristics of the subject to be treated.
“Prophylactically effective amount,’’ as used herein, is intended to include the amount of a modulatory nucleic acid agent that, when administered to a subject having a SLC6A19-associated disorder, is sufficient to prevent or ameliorate the disease or one or more symptoms of the disease. Ameliorating the disease includes slowing the course of the disease or reducing the severity of later-developing disease. The "prophylactically effective amount" may vary’ depending on the modulatory' nucleic acid agent, how the agent is administered, the degree of risk of disease, and the history, age, weight, family history, genetic makeup, the types of preceding or concomitant treatments, if any, and other individual characteristics of the patient to be treated.
A "therapeutically-effective amount" or “prophylactically effective amount” also includes an amount of a modulatory nucleic acid agent that produces some desired effect at. a reasonable benefit/risk ratio applicable to any treatment. The iRNA employed in the methods of the present disclosure may be administered in a sufficient amount to produce a reasonable benefit/risk ratio applicable to such treatment.
The phrase "pharmaceutically acceptable" is employed herein to refer to those compounds, materials, compositions, or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human subjects and animal subjects without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.
The phrase "pharmaceutically-acceptable carrier" as used herein means a pharmaceutically-acceptable material, composition, or vehicle, such as a liquid or solid filler, diluent, excipient, manufacturing aid (e.g., lubricant, talc magnesium, calcium or zinc stearate, or steric acid), or solvent encapsulating material, involved in carrying or transporting the subject compound from one organ, or portion of the body, to another organ, or portion of the body. Each carrier must be "acceptable" in the sense of being compatible with the other ingredients of the formulation and not injurious to the subject being treated. Such carriers are known in the art. Pharmaceutically acceptable carriers include carriers for administration by injection. The term “sample,” as used herein, includes a collection of similar fluids, cells, or tissues isolated from a subject, as well as fluids, cells, or tissues present within a subject. Examples of biological fluids include blood, serum and serosal fluids, plasma, cerebrospinal fluid, ocular fluids, lymph, urine, saliva, and the like. Tissue samples may include samples from tissues, organs, or localized regions. For example, samples may be derived from particular organs, parts of organs, or fluids or cells within those organs. In certain embodiments, samples may be derived from the kidney (e.g., whole kidney or certain segments of kidney or certain types of cells in the kidney, such as, e g., proximal tubular epithelial cells, podocytes, etc.). In some embodiments, a “sample derived from a subject” refers to urine obtained from the subject. A “sample derived from a subject” can refer to blood or blood derived serum or plasma from the subject.
As used herein, the terms “target nucleic acid”, “nucleic acid molecule encoding SLC6AI9” and "nucleic acid molecule encoding B°AT1" have been used for convenience to encompass RNA (including pre-mRNA and mRNA or portions thereof) transcribed from DNA encoding SLC6A19 and/or B°AT1, and also cDNA derived from such RNA. In a preferred embodiment, the target nucleic acid is an mRNA encoding for human B°AT1.
The term "target region" of a RNA refers either to: (1) for an ASO-targeted RNA, the sequence within the ASO-targeted RNA that is complementary to the ASO sequence or (2) for an RNAi agent-targeted RNA, the sequence within the RNAi agent-targeted RNA that is complementary to the antisense strand of the RNAi agent.
Modulation of expression of a target nucleic acid can be achieved through alteration of any number of nucleic acid (DNA or RNA) functions. “Modulation” or “Modulation of Expression” means a perturbation of function, for example, either an increase (stimulation or induction) or a decrease (inhibition or reduction) in expression of the target mRNA. As another example, modulation of expression can include perturbing splice site selection of pre-mRNA processing. “Expression” includes all the functions by which a gene's coded information is converted into structures present and operating in a cell. These structures include the products of transcription and translation. The functions of RNA to be modulated can include translocation functions, which include, but are not limited to, translocation of the RNA to a site of protein translation, translocation of the RNA to sites within the cell which are distant from the site of RNA synthesis, and translation of protein from the RNA. RNA processing functions that can be modulated include, but are not limited to, splicing of the RNA to yield one or more RNA species, capping of the RNA, 3' maturation of the RNA and catalytic activity or complex formation involving the RNA which may be engaged in or facilitated by the RNA. Modulation of expression can result in the increased level of one or more nucleic acid species or the decreased level of one or more nucleic acid species, either temporally or by net steady state level. One result of such interference with target nucleic acid function is modulation of the expression of SLC6A19 and/or B°AT1. Thus, in one embodiment, modulation of expression can mean increase or decrease in target RNA or protein levels. In another embodiment, modulation of expression can mean an increase or decrease of one or more RNA splice products, or a change in the ratio of two or more splice products.
The effect of modulatory RNA compounds on target nucleic acid expression can be tested in any of a variety of cell types provided that the target nucleic acid is present at measurable levels and can be routinely determined using, for example, PCR or Northern blot analysis. Cell lines are derived from both normal tissues and cell types and from cells associated with various disorders (e g., hyperproliferative disorders). Cell lines derived from multiple tissues and species can be obtained from American Type Culture Collection (ATCC, Manassas, Va.) and other public sources. Primary cells, or those cells which are isolated from an animal and not subjected to continuous culture, can be prepared according to methods known in the art, or obtained from various commercial suppliers. Additionally, primary cells include those obtained from donor human subjects in a clinical setting (i.e., blood donors, surgical patients). These techniques are well known to those skilled in the art.
BRIEF DESCRIPTION OF THE DRAWING
The following detailed description, given by way of example, but not intended to limit the disclosure solely to the specific embodiments described, may best be understood in conjunction with the accompanying drawing.
FIG. 1 shows results of screening human SLC6A19-targeting siRNAs for knockdown of huSLC6A19 expression in HEP3B217 cells in culture. Significant knockdown of SLC6A19 expression (>50%, dashed line) was observed for S003 and S005-S014 siRNAs, with doseresponse also observed for many of these siRNAs when comparing between 10 nM and 1 nM administrations of respective siRNAs.
The present disclosure is further illustrated by the following detailed description. DETAILED DESCRIPTION OF THE INVENTION
The present disclosure provides modulatory nucleic acid agent compositions, e.g., antisense oligonucleotides (ASOs) and/or inhibitory RNAs (iRNAs, e.g., siRNAs), capable of modulating (in certain embodiments, inhibiting) expression of a SLC6A19 target gene. Without wishing to be bound by theory, the ASOs provided herein are believed to trigger degradation of SLC6A19 gene target(s) by recruiting RNase H, while iRNAs (e.g., dsRNAs, e.g., siRNAs) provided herein are believed to effect the RNA-induced silencing complex (RlSC)-mediated cleavage of RNA transcript(s) of a SLC6A19 target gene. The SLC6A19 target gene and/or SLC6A19 transcript(s) may be within a cell, e.g., a cell within a subject, such as a human. The use of these modulatory nucleic acid agent compositions enables the targeted degradation of mRNAs of the corresponding SLC6A19 gene in mammals.
The modulatory nucleic acid agents of the disclosure have been designed to target the human SLC6A19 gene, including portions of the gene that are conserved in the SLC6A19 orthologs of other mammalian species Without intending to be limited by theory, it is believed that a combination or sub-combination of the foregoing properties and the specific target sites or the specific modifications in these modulator}' nucleic acid agents confer to the modulatory' nucleic acid agents of the disclosure improved efficacy, stability, potency, durability, and safety.
Accordingly, the present disclosure provides methods for treating and preventing a SLC6A19-associated disorder, e g., a glomerular disorder, a renal tubular disorder, other renal disorders, an inborn error of metabolism, a systemic metabolic disorder, a disorder of the thyroid, a disorder of the parathyroid, a disorder of the inner ear, a neurological disorder, or a viral infection, or any combination thereof, using modulatory nucleic acid agent compositions which effect the RNase H-mediated degradation and/or the RNA-induced silencing complex (RISC)- mediated cleavage of RNA transcripts of a SLC6A19 gene.
The modulatory nucleic acid agents of the disclosure include an RNA strand (the antisense strand) having a region which is up to about 30 nucleotides or less in length, e.g., 19-30, 19-29,
19-28, 19-27, 19-26, 19-25, 19-24, 19-23, 19-22, 19-21, 19-20, 20-30, 20-29, 20-28, 20-27, 20-26,
20-25, 20-24,20-23, 20-22, 20-21 , 21-30, 21 -29, 21-28, 21-27, 21 -26, 21 -25, 21-24, 21-23, or 21- 22 nucleotides in length, which region is substantially complementary' to at least part of an mRNA transcript of a SLC6A19 gene. In certain embodiments, an ASO and/or one or both of the strands of a double stranded RNAi agent of the disclosure is up to 66 nucleotides in length, e.g., 36-66, 26-36, 25-36, 31-60, 22-43, 27-53 nucleotides in length, with a region of at least 19 contiguous nucleotides that is substantially complementary to at least a part of an mRNA transcript of a SLC6A19 gene. In some embodiments, such ASO and/or iRNA agents having longer length antisense strands preferably may include a second RNA strand (the sense strand) of 20-60 nucleotides in length wherein the sense and antisense strands form a duplex of 18-30 contiguous nucleotides.
The use of ASOs and/or iRNAs of the disclosure enables the targeted inhibition and/or degradation of mRNAs of the corresponding gene (SLC6A19 gene) in mammals. Using in vitro assays, the present inventors demonstrate that ASOs and/or iRNAs targeting a SLC6A19 gene can potently mediate RNAi, resulting in significant inhibition of expression of a SLC6A19 gene. Thu s, methods and compositions including these ASOs and/or iRNAs are useful for treating a subject having a SLC6A19-associated disorder, e.g., a glomerular disorder, a renal tubular disorder, other renal disorders, an inborn error of metabolism, a systemic metabolic disorder, a disorder of the thyroid, a disorder of the parathyroid, a disorder of the inner ear, a neurological disorder, or a viral infection, or any combination thereof.
Accordingly, the present disclosure provides methods and combination therapies for treating a subject having a disorder that would benefit from modulating (e.g., inhibiting or reducing) the expression of a SLC6A19 gene, e.g., a SLC6A19-associated disease, such as a glomerular disorder, a renal tubular disorder, other renal disorders, an inborn error of metabolism, a systemic metabolic disorder, a disorder of the thyroid, a disorder of the parathyroid, a disorder of the inner ear, a neurological disorder, or a viral infection, or any combination thereof, using modulatory' nucleic acid agents, such as ASO and/or iRNA compositions which effect the RNase H-mediated degradation and/or the RNA-induced silencing complex (RlSC)-mediated cleavage of RNA transcripts of a SLC6A19 gene.
The present disclosure also provides methods for preventing at least one symptom in a subject having a disorder that would benefit from inhibiting or reducing the expression of a SLC6A19 gene, e.g , a glomerular disorder, a renal tubular disorder, other renal disorders, an inborn error of metabolism, a systemic metabolic disorder, a disorder of the thyroid, a disorder of the parathyroid, a disorder of the inner ear, a neurological disorder, or a viral infection, or any combination thereof The following detailed description discloses how to make and use compositions containing modulatory nucleic acid agents, e.g., ASOs and/or iRNAs, among others, to modulate (e.g., inhibit) the expression of a SLC6A19 gene as well as compositions, uses, and methods for treating subjects that would benefit from modulation (e.g., inhibition and/or reduction) of the expression of a SLC6A19 gene, e.g., subjects susceptible to or diagnosed with a SLC6A19-associated disorder
In some embodiments, a modulatory nucleic acid agent, e.g., an oligonucleotide agent, of the present disclosure may include one or more structural features or characteristics relevant to its mode of action. For example, those skilled in the art are aware of extensive literature regarding structural features of, for example, oligonucleotides that trigger degradation of their targets (e.g., by recruiting RNase H (such oligonucleotides often being referred to as “antisense” agents or “ASOs”) and/or Dicer and/or other elements of the RNA-Induced Silencing Complex (RISC) (such oligonucleotides often being referred to as “siRNA” agents) and/or that modulate splicing of target transcripts (e.g., to favor production of one splice form over another) and/or that act as guide RNAs to recruit other machinery (e.g., nucleases such as CRISPR/Cas or dsRNA binding proteins, or conjugates thereof, etc.) to particular nucleic acid sequences, or as aptamers that bind to particular targets, etc.
In some embodiments, a modulatory nucleic acid of the instant disclosure is or comprises an interfering RNA (RNAi) agent. In some embodiments, a modulatory nucleic acid agent is or comprises a short interfering RNA (siRNA) agent. In some embodiments, a modulatory nucleic acid agent is or comprises a micro RNA (miRNA) agent. In some embodiments, a modulatory nucleic acid agent is or comprises a guide RNA (gRNA) agent.
In some embodiments, a modulatory nucleic acid agent is or comprises a short interfering RNA (siRNA) agent. In some embodiments, a dsRNA modulatory nucleic acid agent can be linked to a targeting moiety (e.g., directly or indirectly) at a sense strand. In some embodiments, a dsRNA modulatory nucleic acid agent can be linked to a targeting moiety (e.g., directly or indirectly) at an antisense strand. In some embodiments, a dsRNA modulatory nucleic acid agent can be linked to a targeting moiety (e.g., directly or indirectly) at a 5’ end of a dsRNA. In some embodiments, a dsRNA modulatory nucleic acid agent can be linked to a targeting moiety (e.g., directly or indirectly) at a 3’ end of a dsRNA.
In some embodiments, a modulatory nucleic acid of the disclosure is or comprises an exon skipping agent, an exon inclusion agent, or other splicing modulator. In some embodiments, a modulatory nucleic acid of the disclosure is or comprises an aptamer agent.
In some embodiments, a modulatory nucleic acid agent of the disclosure is or comprises an antisense oligonucleotide (ASO). In some embodiments, an ASO modulates gene expression via RNase H-mediated mechanisms. In some embodiments, an ASO modulates gene expression via steric hindrance.
In some embodiments, a modulatory nucleic acid agent of the disclosure is or comprises a phosphorodiamidate morpholino oligonucleotide (PMO).
In some embodiments, a modulatory nucleic acid agent of the disclosure is or comprises a peptide-nucleic acid (PNA).
In some embodiments, a modulatory nucleic acid agent of the disclosure is or comprises a nucleic acid analog, e.g., an RNA analog or a DNA analog, or any combination thereof.
In some embodiments, a modulatory nucleic acid agent of the disclosure can be linked to a targeting moiety (e.g., directly or indirectly) at a sense strand. In some embodiments, a modulatory nucleic acid agent of the disclosure can be linked to a targeting moiety (e.g., directly or indirectly) at an antisense strand. In some embodiments, a modulatory nucleic acid agent of the disclosure can be linked to a targeting moiety (e.g., directly or indirectly) at a 5’ end of the modulatory nucleic acid. In some embodiments, a modulatory nucleic acid agent of the disclosure can be linked to a targeting moiety (e.g., directly or indirectly) at a 3 ’ end of the modulatory nucleic acid.
Additional detail regarding the compositions and methods of the instant disclosure is provided in the following sections.
Modulatory Nucleic Acid Agents
In certain aspects, the instant disclosure provides ASOs and/or iRNAs which inhibit the expression of a SLC6A19 gene. In certain embodiments, an iRNA includes double stranded ribonucleic acid (dsRNA) molecules for inhibiting the expression of a SLC6A19 gene in a cell, such as a cell within a subject, e.g., a mammal, such as a human susceptible to developing an SLC6A19-associated disorder, e.g., metabolic disorders, including nephropathy and various other kidney diseases or conditions.
In some embodiments, a modulatory nucleic acid agent, e.g., an oligonucleotide agent, of the present disclosure may include one or more structural features or characteristics relevant to its mode of action. For example, those skilled in the art are aware of extensive literature regarding structural features of, for example, oligonucleotides that trigger degradation of their targets (e.g., by recruiting RNase H (such oligonucleotides often being referred to as “antisense” agents or “ASOs”) and/or Dicer and/or other elements of the RNA-Induced Silencing Complex (RISC) (such oligonucleotides often being referred to as “siRNA” agents) and/or that modulate splicing of target transcripts (e.g., to favor production of one splice form over another) and/or that act as guide RNAs to recruit other machinery (e.g., nucleases such as CRISPR/Cas or dsRNA binding proteins, or conjugates thereof, etc.) to particular nucleic acid sequences, or as aptamers that bind to particular targets, etc.
In some embodiments, a modulatory nucleic acid of the instant disclosure is or comprises an interfering RNA (RNAi) agent. In some embodiments, a modulatory nucleic acid agent is or comprises a short interfering RNA (siRNA) agent. In some embodiments, a modulatory nucleic acid agent is or comprises a micro-RNA (miRNA) agent. In some embodiments, a modulatory nucleic acid agent is or comprises a guide RNA (gRNA) agent.
In some embodiments, a modulatory nucleic acid agent is or comprises a short interfering RNA (siRNA) agent. In some embodiments, a dsRNA modulatory nucleic acid agent can be linked to a targeting moiety (e.g., directly or indirectly) at a sense strand. In some embodiments, a dsRNA modulatory nucleic acid agent can be linked to a targeting moiety (e.g., directly or indirectly) at an antisense strand. In some embodiments, a dsRNA modulatory nucleic acid agent can be linked to a targeting moiety (e.g., directly or indirectly) at a 5’ end of a dsRNA. In some embodiments, a dsRNA modulatory nucleic acid agent can be linked to a targeting moiety (e.g., directly or indirectly) at a 3’ end of a dsRNA.
In some embodiments, a modulatory nucleic acid of the disclosure is or comprises an exon skipping agent, an exon inclusion agent, or other splicing modulator.
In some embodiments, a modulatory nucleic acid of the disclosure is or comprises an aptamer agent.
In some embodiments, a modulatory nucleic acid agent of the disclosure is or comprises an antisense oligo (ASO). In some embodiments, an ASO modulates gene expression via RNase H-mediated mechanisms. In some embodiments, an ASO modulates gene expression via steric hindrance. In some embodiments, a modulatory nucleic acid agent of the disclosure is or comprises a phosphorodiamidate morpholino oligonucleotide (PMO).
In some embodiments, a modulatory nucleic acid agent of the disclosure is or comprises a peptide-nucleic acid (PNA).
In some embodiments, a modulatory nucleic acid agent of the disclosure is or comprises a nucleic acid analog, e.g., an RNA analog or a DNA analog, or any combination thereof.
In some embodiments, a modulatory nucleic acid agent of the disclosure can be linked to a targeting moiety (e.g., directly or indirectly) at a sense strand. In some embodiments, a modulatory nucleic acid agent of the disclosure can be linked to a targeting moiety (e.g., directly or indirectly) at an antisense strand. In some embodiments, a modulatory nucleic acid agent of the disclosure can be linked to a targeting moiety (e.g., directly or indirectly) at a 5’ end of the modulatory nucleic acid. In some embodiments, a modulatory nucleic acid agent of the disclosure can be linked to a targeting moiety (e.g., directly or indirectly) at a 3 ’ end of the modulatory nucleic acid.
In some embodiments, a modulatory nucleic acid of the instant disclosure has a length and/or a strand length (for multi -stranded modulatory nucleic acids, e.g., dsRNAs) within a range of about 10-60 nucleotides, about 10-59 nucleotides, about 10-58 nucleotides, about 10-57 nucleotides, about 10-56 nucleotides, about 10-55 nucleotides, about 10-54 nucleotides, about 10- 53 nucleotides, about 10-52 nucleotides, about 10-51 nucleotides, about 10-50 nucleotides, about 10-49 nucleotides, about 10-48 nucleotides, about 10-47 nucleotides, about 10-46 nucleotides, about 10-45 nucleotides, about 10-44 nucleotides, about 10-43 nucleotides, about 10-42 nucleotides, about 10-41 nucleotides, about 10-40 nucleotides, about 10-39 nucleotides, about 10- 38 nucleotides, about 10-37 nucleotides, about 10-36 nucleotides, about 10-35 nucleotides, about 10-34 nucleotides, about 10-33 nucleotides, about 10-32 nucleotides, about 10-31 nucleotides, about 10-30 nucleotides, about 10-29 nucleotides, about 10-28 nucleotides, about 10-27 nucleotides, about 10-26 nucleotides, about 10-25 nucleotides, about 10-24 nucleotides, about 10- 23 nucleotides, about 10-22 nucleotides, about 10-21 nucleotides, about 10-20 nucleotides, about 10-19 nucleotides, about 10-18 nucleotides, about 10-17 nucleotides, about 10-16 nucleotides, about 10-15 nucleotides, about 10-14 nucleotides, about 10-13 nucleotides, about 10-12 nucleotides, about 10-11 nucleotides. In some embodiments, a modulatory nucleic acid of the instant disclosure has a length and/or a strand length (for multi -stranded modulatory nucleic acids, e g., dsRNAs) within a range of about 11-60 nucleotides, about 12-60 nucleotides, about 13-60 nucleotides, about 14-60 nucleotides, about 15-60 nucleotides, about 16-60 nucleotides, about 17- 60 nucleotides, about 18-60 nucleotides, about 19-60 nucleotides, about 20-60 nucleotides, about
21-60 nucleotides, about 22-60 nucleotides, about 23-60 nucleotides, about 24-60 nucleotides, about 25-60 nucleotides, about 26-60 nucleotides, about 27-60 nucleotides, about 28-60 nucleotides, about 29-60 nucleotides, about 30-60 nucleotides, about 31-60 nucleotides, about 32- 60 nucleotides, about 33-60 nucleotides, about 34-60 nucleotides, about 35-60 nucleotides, about
36-60 nucleotides, about 37-60 nucleotides, about 38-60 nucleotides, about 39-60 nucleotides, about 40-60 nucleotides, about 41-60 nucleotides, about 42-60 nucleotides, about 43-60 nucleotides, about 44-60 nucleotides, about 45-60 nucleotides, about 46-60 nucleotides, about 47- 60 nucleotides, about 48-60 nucleotides, about 49-60 nucleotides, about 50-60 nucleotides, about 51-60 nucleotides, about 52-60 nucleotides, about 53-60 nucleotides, about 54-60 nucleotides, about 55-60 nucleotides, about 56-60 nucleotides, about 57-60 nucleotides, about 58-60 nucleotides, about 59-60 nucleotides.
In some embodiments, a modulatory nucleic acid of the instant disclosure is about 10 nucleotides, about 11 nucleotides, about 12 nucleotides, about 13 nucleotides, about 14 nucleotides, about 15 nucleotides, about 16 nucleotides, about 17 nucleotides, about 18 nucleotides, about 19 nucleotides, about 20 nucleotides, about 21 nucleotides, about 22 nucleotides, about 23 nucleotides, about 24 nucleotides, about 25 nucleotides, about 26 nucleotides, about 27 nucleotides, about 28 nucleotides, about 29 nucleotides, about 30 nucleotides, about 31 nucleotides, about 32 nucleotides, about 33 nucleotides, about 34 nucleotides, about 35 nucleotides, about 36 nucleotides, about 37 nucleotides, about 38 nucleotides, about 39 nucleotides, about 40 nucleotides, about 41 nucleotides, about 42 nucleotides, about 43 nucleotides, about 44 nucleotides, about 45 nucleotides, about 46 nucleotides, about 47 nucleotides, about 48 nucleotides, about 49 nucleotides, about 50 nucleotides, about 51 nucleotides, about 52 nucleotides, about 53 nucleotides, about 54 nucleotides, about 55 nucleotides, about 56 nucleotides, about 57 nucleotides, about 58 nucleotides, about 59 nucleotides, about 60 nucleotides in length.
In some embodiments, a modulatory nucleic acid agent, e.g., an oligonucleotide agent, of the present disclosure may comprise a single strand. In some embodiments, a modulatory nucleic acid agent may comprise more than one strand. In some embodiments, a modulatory nucleic acid agent may comprise one or more double-stranded portions. In some such embodiments, some or all of such portion(s) may be formed by self-hybridization of sequences on a single strand; in some embodiments some or all of such portion(s) may be formed by hybridization of separate strands. In some embodiments, a modulatory nucleic acid agent that includes one or more double-stranded portions may include one or more nicks or gaps and/or one or more bulges or loops. dsRNA Agents
The duplex region of a dsRNA of the instant disclosure may be of any length that permits specific degradation of a desired target RNA through a RISC pathway, and may range from about 19 to 36 base pairs in length, e.g., about 19-30 base pairs in length, for example, about 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, or 36 base pairs in length, such as about 19-30, 19-29, 19-28, 19-27, 19-26, 19-25, 19-24, 19-23, 19-22, 19-21, 19-20, 20-30, 20-29, 20-28, 20-27, 20-26, 20-25, 20-24,20-23, 20-22, 20-21, 21-30, 21-29, 21-28, 21-27, 21-26, 21-25, 21-24, 21-23, or 21 -22 base pairs in length. In certain embodiments, the duplex region is 19-21 base pairs in length, e.g., 21 base pairs in length. Ranges and lengths intermediate to the above recited ranges and lengths are also contemplated to be part of the disclosure.
The two strands forming the duplex structure may be different portions of one larger RNA molecule, or they may be separate RNA molecules. Where the two strands are part of one larger molecule, and therefore are connected by an uninterrupted chain of nucleotides between the 3’- end of one strand and the 5’-end of the respective other strand forming the duplex structure, the connecting RNA chain is referred to as a “hairpin loop.” A hairpin loop can comprise at least one unpaired nucleotide. In some embodiments, the hairpin loop can comprise at least 2, 3, 4, 5, 6, 7, 8, 9, 10, 20, 23 or more unpaired nucleotides. In some embodiments, the hairpin loop can be 10 or fewer nucleotides. In some embodiments, the hairpin loop can be 8 or fewer unpaired nucleotides In some embodiments, the hairpin loop can be 4-10 unpaired nucleotides. In some embodiments, the hairpin loop can be 4-8 nucleotides.
Where the two substantially complementary' strands of a dsRNA are comprised by separate RNA molecules, those molecules need not be, but can be covalently connected. Where the two strands are connected covalently by means other than an uninterrupted chain of nucleotides between the 3 ’-end of one strand and the 5 ’-end of the respective other strand forming the duplex structure, the connecting structure is referred to as a “dsRN A linker,” The RNA strands may have the same or a different number of nucleotides. The maximum number of base pairs is the number of nucleotides in the shortest strand of the dsRNA minus any overhangs that are present in the duplex. In addition to the duplex structure, an RNAi may comprise one or more nucleotide overhangs. In one embodiment of the RNAi agent, at least one strand comprises a 3’ overhang of at least I 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, 1 1, 12, 13, 14, or 15 nucleotides. In other embodiments, at least one strand of the RNAi agent comprises a 5’ overhang of at least 1 nucleotide. In certain embodiments, at least one strand comprises a 5’ overhang of at least 2 nucleotides, e.g., 2, 3, 4, 5, 6, 7, 9, 10, 11, 12, 13, 14, or 15 nucleotides. In still other embodiments, both the 3’ and the 5’ end of one strand of the RNAi agent comprise an overhang of at least 1 nucleotide.
In certain embodiments, an iRNA agent of the present disclosure is a dsRNA, each strand of which comprises 19-23 nucleotides, that interacts with a target RNA sequence, e.g., a SLC6A19 gene, to direct cleavage of the target RNA.
In some embodiments, an iRNA of the present disclosure is a dsRNA of 24-30 nucleotides that interacts with a target RNA sequence, e.g., a SLC6A19 target mRNA sequence, to direct the cleavage of the target RNA.
As used herein, the term '"nucleotide overhang” refers to at least one unpaired nucleotide that protrudes from the duplex structure of a double stranded iRNA. For example, when a 3'-end of one strand of a dsRNA extends beyond the 5'-end of the other strand, or vice versa, there is a nucleotide overhang. A dsRNA can comprise an overhang of at least one nucleotide; alternatively, the overhang can comprise at least two nucleotides, at least three nucleotides, at least four nucleotides, at least five nucleotides or more. A nucleotide overhang can comprise or consist of a nucleotide/nucleoside analog, including a deoxynucleotide/nucleoside. The overhang(s) can be on the sense strand, the antisense strand, or any combination thereof. Furthermore, the nucleotide(s) of an overhang can be present on the 5!-end, 3'-end, or both ends of either an antisense or sense strand of a dsRNA .
In one embodiment, the antisense strand of a dsRNA has a 1-10 nucleotide, e.g., a 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 nucleotide, overhang at the 3’-end or the 5’-end. In one embodiment, the sense strand of a dsRNA has a 1-10 nucleotide, e.g., a 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 nucleotide, overhang at the 3 ’-end or the 5 ’-end. In another embodiment, one or more of the nucleotides in the overhang is replaced with a nucleoside thiophosphate In certain embodiments, the antisense strand of a dsRNA has a 1 -10 nucleotide, e.g., 0-3, 1-3, 2-4, 2-5, 4-10, 5-10, e.g., a 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 nucleotide, overhang at the 3 ’-end or the 5 ’-end. In one embodiment, the sense strand of a dsRNA has a 1-10 nucleotide, e.g., a 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 nucleotide, overhang at the 3 ’-end or the 5 ’-end. In another embodiment, one or more of the nucleotides in the overhang is replaced with a nucleoside thiophosphate.
In certain embodiments, the antisense strand of a dsRNA has a 1-10 nucleotides, e.g., a 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 nucleotide, overhang at the 3 ’-end or the 5 ’-end. In certain embodiments, the overhang on the sense strand or the antisense strand, or both, can include extended lengths longer than 10 nucleotides, e.g., 1-30 nucleotides, 2-30 nucleotides, 10-30 nucleotides, 10-25 nucleotides, 10-20 nucleotides, or 10-15 nucleotides in length. In certain embodiments, an extended overhang is on the sense strand of the duplex. In certain embodiments, an extended overhang is present on the 3’ end of the sense strand of the duplex. In certain embodiments, an extended overhang is present on the 5’ end of the sense strand of the duplex. In certain embodiments, an extended overhang is on the antisense strand of the duplex. In certain embodiments, an extended overhang is present on the 3’ end of the antisense strand of the duplex. In certain embodiments, an extended overhang is present on the 5’end of the antisense strand of the duplex In certain embodiments, one or more of the nucleotides in the extended overhang is replaced with a nucleoside thiophosphate. In certain embodiments, the overhang includes a self- complementary portion such that the overhang is capable of forming a hairpin structure that, is stable under physiological conditions.
“Blunt” or “blunt end” means that there are no unpaired nucleotides at that end of the double stranded RNA agent, i.e., no nucleotide overhang. A “blunt ended” double stranded RNA agent is double stranded over its entire length, i e., no nucleotide overhang at either end of the molecule. The RNAi agents of the present disclosure include RNAi agents with no nucleotide overhang at one end (i.e., agents with one overhang and one blunt end) or with no nucleotide overhangs at either end. Most often such a molecule will be double-stranded over its entire length.
The term “antisense strand” or "guide strand" refers to the strand of an iRNA, e.g., a dsRNA, which includes a region that is substantially complementary to a target sequence, e.g , a SLC6A19 mRNA.
As used herein, the term “region of complementarity” refers to the region on the antisense strand that is substantially complementary to a sequence, for example a target sequence, e.g., a SLC6A19 nucleotide sequence, as defined herein. Where the region of complementarity is not fully complementary to the target sequence, the mismatches can be in the internal or terminal regions of the molecule. Generally, the most tolerated mismatches are in the terminal regions, e.g., within 5, 4, or 3 nucleotides of the 5’- or 3’-end of the iRNA. In some embodiments, a double stranded RNA agent of the present disclosure includes a nucleotide mismatch in the antisense strand. In some embodiments, the antisense strand of the double stranded RNA agent of the present disclosure includes no more than 4 mismatches with the target mRNA, e.g., the antisense strand includes 4, 3, 2, 1 , or 0 mismatches with the target mRNA. In some embodiments, the antisense strand double stranded RNA agent of the present disclosure includes no more than 4 mismatches with the sense strand, e.g., the antisense strand includes 4, 3, 2, 1, or 0 mismatches with the sense strand. In some embodiments, a double stranded RNA agent of the present disclosure includes a nucleotide mismatch in the sense strand. In some embodiments, the sense strand of the double stranded RNA agent of the present disclosure includes no more than 4 mismatches with the antisense strand, e g., the sense strand includes 4, 3, 2, 1, or 0 mismatches with the antisense strand In some embodiments, the nucleotide mismatch is, for example, within 5, 4, 3 nucleotides from the 3 ’-end of the iRNA. In another embodiment, the nucleotide mismatch is, for example, in the 3’-terminal nucleotide of the iRNA agent. In some embodiments, the mismatch(s) is not in the seed region.
Thus, an RNAi agent as described herein can contain one or more mismatches to the target sequence. In one embodiment, a RNAi agent as described herein contains no more than 3 mismatches (i.e., 3, 2, 1, or 0 mismatches). In one embodiment, an RNAi agent as described herein contains no more than 2 mismatches. In one embodiment, an RNAi agent as described herein contains no more than 1 mismatch. In one embodiment, an RNAi agent as described herein contains 0 mismatches. In certain embodiments, if the antisense strand of the RNAi agent contains mismatches to the target sequence, the mismatch can optionally be restricted to be within the last 5 nucleotides from either the 5’- or 3 ’-end of the region of complementarity For example, in such embodiments, for a 23 nucleotide RNAi agent, the strand which is complementary to a region of a SLC6A19 gene, generally does not contain any mismatch within the central 13 nucleotides. The methods described herein or methods known in the art can be used to determine whether an RNAi agent containing a mismatch to a target sequence is effective in inhibiting the expression of a SLC6A19 gene. Consideration of the efficacy of RNAi agents with mismatches in inhibiting expression of a SLC6A19 gene is important, especially if the particular region of complementarity in a SLC6A19 gene is known to have polymorphic sequence variation within the population.
The terra “sense strand” or "passenger strand" as used herein, refers to the strand of an iRNA that includes a region that is substantially complementary to a region of the antisense strand as that term is defined herein.
.As used herein, “substantially all of the nucleotides are modified” are largely but not wholly modified and can include not more than 5, 4, 3, 2, or 1 unmodified nucleotides.
As used herein, the term “cleavage region” refers to a region that is located immediately adjacent to the cleavage site. The cleavage site is the site on the target at which cleavage occurs. In some embodiments, the cleavage region comprises three bases on either end of, and immediately adjacent to, the cleavage site. In some embodiments, the cleavage region comprises two bases on either end of and immediately adjacent to, the cleavage site In some embodiments, the cleavage site specifically occurs at the site bound by nucleotides 10 and 11 of the antisense strand, and the cleavage region comprises nucleotides 11, 12 and 13.
.As used herein, and unless otherwise indicated, the term “complementary,” when used to describe a first nucleotide sequence in relation to a second nucleotide sequence, refers to the ability of an oligonucleotide or polynucleotide comprising the first nucleotide sequence to hybridize and form a duplex structure under certain conditions with an oligonucleotide or polynucleotide comprising the second nucleotide sequence, as will be understood by the skilled person. Such conditions can, for example, be stringent conditions, where stringent conditions can include: 400 rnMNaCl, 40 mM PIPES pH 6.4, 1 mMEDTA, 50°C or 70°C for 12-16 hours followed by washing (see, e.g., “Molecular Cloning: A Laboratory' Manual, Sambrook, et al. (1989) Cold Spring Harbor Laboratory Press). Other conditions, such as physiologically relevant conditions as can be encountered inside an organism, can apply. The skilled person will be able to determine the set of conditions most appropriate for a test of complementarity of two seq uences in accordance with the ultimate application of the hybridized nucleotides.
Complementary' sequences within an iRNA, e.g., within a dsRNA as described herein, include base-pairing of the oligonucleotide or polynucleotide comprising a first nucleotide sequence to an oligonucleotide or polynucleotide comprising a second nucleotide sequence over the entire length of one or both nucleotide sequences. Such sequences can be referred to as “fully complementary” with respect to each other herein. However, where a first sequence is referred to as “substantially complementary” with respect to a second sequence herein, the two sequences can be fully complementary, or they can form one or more, but generally not more than 5, 4, 3, or 2 mismatched base pairs upon hybridization for a duplex up to 30 base pairs, while retaining the ability to hybridize under the conditions most relevant to their ultimate application, e.g., inhibition of gene expression via a RISC pathway. However, where two oligonucleotides are designed to form, upon hybridization, one or more single stranded overhangs, such overhangs shall not be regarded as mismatches with regard to the determination of complementarity For example, a dsRNA comprising one oligonucleotide 21 nucleotides in length and another oligonucleotide 23 nucleotides in length, wherein the longer oligonucleotide comprises a sequence of 21 nucleotides that is fully complementary to the shorter oligonucleotide, can yet be referred to as “fully complementary'” for the purposes described herein.
“Complementary” sequences, as used herein, can also include, or be formed entirely from, non-Watson-Crick base pairs or base pairs formed from non-natural and modified nucleotides, in so far as the above requirements with respect to their ability to hybridize are fulfilled. Such non- Watson-Crick base pairs include, but are not limited to, G:U Wobble or Hoogstein base pairing.
The terms “complementary',” “fully complementary” and “substantially complementary” herein can be used with respect to the base matching between the sense strand and the antisense strand of a dsRNA, or between the antisense strand of a double stranded RNA agent and a target sequence, as will be understood from the context of their use.
As used herein, a polynucleotide that is “substantially complementary' to at least part of” a messenger RNA (mRNA) refers to a polynucleotide that is substantially complementary to a contiguous portion of the mRNA of interest (e.g., an mRN.A encoding a B°AT1 polypeptide). For example, a polynucleotide is complementary to at least a part of a SLC6A19 mRNA if the sequence is substantially complementary' to a non-interrupted portion of an mRNA encoding a B°AT1 polypeptide.
Accordingly, in some embodiments, the antisense polynucleotides disclosed herein are fully complementary to the target SLC6A19 sequence. In other embodiments, the antisense polynucleotides disclosed herein are substantially complementary/ to the target SLC6A19 sequence and comprise a contiguous nucleotide sequence which is at least 80% complementary' over its entire length to the equivalent region of the nucleotide sequence of SEQ ID NO: 1, or a fragment of SEQ ID NO: 1, such as about 85%, about 90%, about 91%, about 92%, about 93%, about 94%, about 95%, about 96%, about 97%, about 98%, or about 99% complementary.
In some embodiments, the antisense polynucleotides disclosed herein are substantially complementary to a fragment of a target SLC6A19 sequence and comprise a contiguous nucleotide sequence which is at least 80% complementary over its entire length to a fragment of SEQ ID NO: 1 selected from the group of nucleotides: 3-25, 8-30, 13-35, 38-60, 43-65, 48-70, 93-115, 98-120, 103-125, 108-130, 111-133, 113-135, 118-140, 214-236, 219-241, 244-266, 249-271, 252-274,
252-274, 253-275, 254-276, 257-286, 259-281, 264-286, 289-311, 294-316, 339-361, 397-419,
460-480, 461-481, 481-503, 486-508, 491-513, 557-579, 561-583, 562-584, 564-586, 566-586,
567-589, 616-638, 621-643, 626-648, 670-692, 671-691, 672-694, 674-694, 675-697, 680-702,
685-707, 690-712, 695-717, 700-722, 705-727, 710-732, 715-737, 720-742, 725-747, 730-752,
735-757, 740-762, 745-767, 750-772, 751-776, 755-777, 760-782, 765-787, 770-792, 775-797,
798-823, 800-822, 825-847, 890-912, 895-917, 900-922, 905-927, 910-932, 911-937, 915-937,
972-994, 973-995, 977-1000, 977-999, 978-1000, 983-1005, 1035-1057, 1058-1080, 1059-1081,
1060-1082, 1063-1082, 1065-1087, 1068-1087, 1070-1094, 1070-1092, 1074-1096, 1075-1097, 1077-1096, 1080-1102, 1085-1107, 1086-1106, 1090-1112, 1095-1117, 1100-1122, 1105-1127, 1110-1132, 1115-1137, 1115-1137, 1117-1139, 1177-1199, 1182-1204, 1187-1209, 1191-1214, 1192-1214, 1195-1217, 1234-1256, 1239-1261, 1242-1264, 1244-1266, 1247-1268, 1249-1271, 1251-1270, 1254-1276, 1259-1281, 1263-1282, 1264-1286, 1266-1285, 1269-1291, 1326-1348, 1331-1353, 1334-1361, 1334-1356, 1334-1356, 1335-1357, 1336-1358, 1337-1359, 1337-1359, 1338-1360, 1340-1362, 1341-1363, 1346-1368, 1351-1373, 1353-1372, 1377-1399, 1382-1404, 1383-1403, 1387-1409, 1392-1414, 1397-1419, 1402-1424, 1407-1429, 1412-1434, 1417-1439, 1456-1478, 1458-1477, 1481-1503, 1486-1508, 1491-1513, 1496-1518, 1521-1543, 1568-1590, 1569-1591, 1574-1596, 1578-1621, 1579-1601, 1583-1605, 1583-1605, 1584-1606, 1584-1606, 1585-1607, 1585-1607, 1587-1609, 1588-1610, 1589-1611, 1594-1616, 1595-1617, 1599-1621, 1601-1620, 1602-1624, 1642-1664, 1749-1775, 1750-1772, 1755-1777, 1757-1779, 1758-1780, 1760-1782, 1761-1792, 1764-1783, 1765-1787, 1770-1792, 1809-1831, 1810-1829, 1812-1832, 1814-1836, 1852-1881, 1853-1875, 1854-1876, 1858-1880, 2168-2188, 2333-2353, 2454-2474, 2507-2527, 2593-2613, 3244-3264, 3288-3308, 3304-3324, 3332-3352, 3333-3353, 3501-3523, 3595-3615, 3595-3617, 3643-3671, 3646-3668, 3646-3668, 3647-3669, 3648-3668, 3648-3670, 3648-3670, 3649-3671, 3651-3670, 3651-3673, 3651-3673, 3652-3671, 3712-3733, 3713-3735, 3763-3786, 3763-3785, 3764-3786, 3791-3811 , 3796-3816, 3801-3829, 3801-3820, 3802-3821 , 3804-3826, 3809-3831, 3810-3832, 3812-3833, 3812-3834, 3813-3835, 3814-3833, 3814-3836, 3814-3836, 3839-3859, 3840-3862, 3902-3924, 3965-3987, 3995-4015, 3998-4020, 4100-4124, 4102-4124, 4141-4163, 4141-4163, 4142-4164, 4143-4165, 4144-4166, 4146-4168, 4150-4172, 4151-4173, 4192-4214, 4197-4219, 4201-4223, 4265-4287, 4270-4292, 4318-4338, 4319-4341, 4329-4349, 4348-4386, 4349-4371, 4350-4369, 4350-4372, 4350-4372, 4351-4370, 4352-4371, 4352-4374, 4353-4372, 4353-4375, 4354-4376, 4359-4381, 4361-4383, 4362-4384, 4362-4384, 4364-4386, 4365-4387, 4395-4415, 4396-4416, 4409-4431, 4410-4432, 4412-4434, 4637-4657, 4638-4658, 4640-4660, 5103-5123, 5124-5144, 5126-5146, and 5127-5147 of SEQ ID NO: 1, such as about 85%, about 90%, about 91%, about 92%, about 93%, about 94%, about 95%, about 96%, about 97%, about 98%, or about 99% complementary'.
In other embodiments, the antisense polynucleotides disclosed herein are substantially complementary' to the target SLC6A19 sequence and comprise a contiguous nucleotide sequence which is at least about 80% complementary over its entire length to any one of the sense strand nucleotide sequences in Tables 7-11, 14 or 17, or a fragment of any one of the sense strand nucleotide sequences in Tables 7-11, 14 or 17, 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%o complementary'.
In one embodiment, an RNAi agent, of the disclosure includes a sense strand that is substantially complementary' to an antisense polynucleotide which, in turn, is the same as a target SLC6A19 sequence, and wherein the sense strand polynucleotide comprises a contiguous nucleotide sequence which is at least about 80% complementary over its entire length to the equivalent region of the nucleotide sequence of SEQ ID NO; 2, or a fragment of SEQ ID NO: 2, 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 some embodiments, an 1R.NA of the present disclosure includes a sense strand that is substantially complementary' to an antisense polynucleotide which, in turn, is complementary to a target SLC6A19 sequence, and wherein the sense strand polynucleotide comprises a contiguous nucleotide sequence which is at least about 80% complementary' over its entire length to any' one of the antisense strand nucleotide sequences in Tables 7-11, 14 or 17, or a fragment of any one of the antisense strand nucleotide sequences in Tables 7-11, 14 or 17, 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.
Ln an aspect, a dsRNA of the present disclosure includes at least two nucleotide sequences, a sense sequence and an antisense sequence. The sense strand is selected from the group of sequences provided in Tables 7-11, 14 and 17, and the corresponding antisense strand of the sense strand is selected from the group of sequences of Tables 7-11, 14 and 17. In this aspect, one of the two sequences is complementary to the other of the two sequences, with one of the sequences being substantially complementary to a sequence of an rnRNA generated in the expression of a SLC6A19 gene. As such, in this aspect, a dsRNA will include two oligonucleotides, where one oligonucleotide is described as the sense strand in Tables 7-11, 14 or 17, and the second oligonucleotide is described as the corresponding antisense strand of the sense strand in Tables 7- 11, 14 or 17
In certain embodiments, the substantially complementary sequences of the dsRNA are contained on separate oligonucleotides. In other embodiments, the substantially complementary' sequences of the dsRNA are contained on a single oligonucleotide.
In certain embodiments, the antisense and/or sense strands are selected from the antisense and/or sense strand of any one of duplexes SEQ ID NOs: 2318 and 2430; SEQ ID NOs: 2319 and 2431; SEQ ID NOs: 2320 and 2432; SEQ ID NOs: 2321 and 2433; SEQ ID NOs: 2322 and 2434; SEQ ID NOs: 2323 and 2435; SEQ ID NOs: 2324 and 2436; SEQ ID NOs: 2325 and 2437; SEQ ID NOs: 2326 and 2438; SEQ ID NOs: 2327 and 2439; SEQ ID NOs: 2328 and 2440; SEQ ID NOs: 2329 and 2441; SEQ ID NOs: 2330 and 2442; SEQ ID NOs: 2331 and 2443; SEQ ID NOs: 2332 and 2444; SEQ ID NOs: 2333 and 2445; SEQ ID NOs: 2334 and 2446; SEQ ID NOs: 2335 and 2447; SEQ ID NOs: 2336 and 2448; SEQ ID NOs: 2337 and 2449; SEQ ID NOs: 2338 and 2450; SEQ ID NOs: 2339 and 2451; SEQ ID NOs: 2340 and 2452; SEQ ID NOs: 2341 and 2453; SEQ ID NOs: 2342 and 2454; SEQ ID NOs: 2343 and 2455; SEQ ID NOs: 2344 and 2456; SEQ ID NOs: 2345 and 2457; SEQ ID NOs: 2346 and 2458; SEQ ID NOs: 2347 and 2459; SEQ ID NOs: 2348 and 2460; SEQ ID NOs: 2349 and 2461; SEQ ID NOs: 2350 and 2462; SEQ ID NOs: 2351 and 2463; SEQ ID NOs: 2352 and 2464; SEQ ID NOs: 2353 and 2465; SEQ ID NOs: 2354 and 2466; SEQ ID NOs: 2355 and 2467; SEQ ID NOs: 2356 and 2468; SEQ ID NOs: 2357 and 2469; SEQ ID NOs: 2358 and 2470; SEQ ID NOs: 2359 and 2471; SEQ ID NOs: 2360 and 2472; SEQ ID NOs: 2361 and 2473; SEQ ID NOs: 2362 and 2474; SEQ ID NOs: 2363 and 2475; SEQ ID NOs: 2364 and 2476; SEQ ID NOs: 2365 and 2477; SEQ ID NOs: 2366 and 2478; SEQ ID NOs: 2367 and 2479; SEQ ID NOs: 2368 and 2480; SEQ ID NOs: 2369 and 2481; SEQ ID NOs: 2370 and 2482; SEQ ID NOs: 2371 and 2483; SEQ ID NOs: 2372 and 2484; SEQ ID NOs: 2373 and 2485; SEQ ID NOs: 2374 and 2486; SEQ ID NOs: 2375 and 2487; SEQ ID NOs: 2376 and 2488; SEQ ID NOs: 2377 and 2489; SEQ ID NOs: 2378 and 2490; SEQ ID NOs: 2379 and 2491; SEQ ID NOs: 2380 and 2492; SEQ ID NOs: 2381 and 2493; SEQ ID NOs: 2382 and 2494; SEQ ID NOs: 2383 and 2495; SEQ ID NOs: 2384 and 2496; SEQ ID NOs: 2385 and 2497; SEQ ID NOs: 2386 and 2498; SEQ ID NOs: 2387 and 2499; SEQ ID NOs: 2388 and 2500; SEQ ID NOs: 2389 and 2501; SEQ ID NOs: 2390 and 2502; SEQ ID NOs: 2391 and 2503; SEQ ID NOs: 2392 and 2504; SEQ ID NOs: 2393 and 2505; SEQ ID NOs: 2394 and 2506; SEQ ID NOs: 2395 and 2507; SEQ ID NOs: 2396 and 2508; SEQ ID NOs: 2397 and 2509; SEQ ID NOs: 2398 and 2510; SEQ ID NOs: 2399 and 2511; SEQ ID NOs: 2400 and 2512; SEQ ID NOs: 2401 and 2513; SEQ ID NOs: 2402 and 2514; SEQ ID NOs: 2403 and 2515; SEQ ID NOs: 2404 and 2516; SEQ ID NOs: 2405 and 2517; SEQ ID NOs: 2406 and 2518; SEQ ID NOs: 2407 and 2519; SEQ ID NOs: 2408 and 2520; SEQ ID NOs: 2409 and 2521; SEQ ID NOs: 2410 and 2522; SEQ ID NOs: 2411 and 2523; SEQ ID NOs: 2412 and 2524; SEQ ID NOs: 2413 and 2525; SEQ ID NOs: 2414 and 2526; SEQ ID NOs: 2415 and 2527; SEQ ID NOs: 2416 and 2528; SEQ ID NOs: 2417 and 2529; SEQ ID NOs: 2418 and 2530; SEQ ID NOs: 2419 and 2531; SEQ ID NOs: 2420 and 2532; SEQ ID NOs: 2421 and 2533; SEQ ID NOs: 2422 and 2534; SEQ ID NOs: 2423 and 2535; SEQ ID NOs: 2424 and 2536; SEQ ID NOs: 2425 and 2537; SEQ ID NOs: 2426 and 2538; SEQ ID NOs: 2427 and 2539; SEQ ID NOs: 2428 and 2540; SEQ ID NOs: 2429 and 2541; SEQ ID NO: 2720 and 2840; SEQ ID NO: 2721 and 2841; SEQ ID NO: 2722 and 2842; SEQ ID NO: 2723 and 2843; SEQ ID NO: 2724 and 2844; SEQ ID NO: 2725 and 2845; SEQ ID NO: 2726 and 2846; SEQ ID NO: 2727 and 2847; SEQ ID NO: 2728 and 2848; SEQ ID NO: 2729 and 2849; SEQ ID NO: 2730 and 2850; SEQ ID NO: 2731 and 2851; SEQ ID NO: 2732 and 2852; SEQ ID NO: 2733 and 2853; SEQ ID NO: 2734 and 2854; SEQ ID NO: 2735 and 2855; SEQ ID NO: 2736 and 2856; SEQ ID NO: 2737 and 2857; SEQ ID NO: 2738 and 2858; SEQ ID NO: 2739 and 2859; SEQ ID NO: 2740 and 2860; SEQ ID NO: 2741 and 2861; SEQ ID NO: 2742 and 2862; SEQ ID NO: 2743 and 2863; SEQ ID NO: 2744 and 2864; SEQ ID NO: 2745 and 2865; SEQ ID NO: 2746 and 2866; SEQ ID NO: 2747 and 2867; SEQ ID NO: 2748 and 2868; SEQ ID NO: 2749 and 2869; SEQ ID NO: 2750 and 2870; SEQ ID NO: 2751 and 2871; SEQ ID NO: 2752 and 2872; SEQ ID NO: 2753 and 2873; SEQ ID NO: 2754 and 2874; SEQ ID NO: 2755 and 2875; SEQ ID NO: 2756 and 2876; SEQ ID NO: 2757 and 2877; SEQ ID NO: 2758 and 2878; SEQ ID NO: 2759 and 2879; SEQ ID NO: 2760 and 2880; SEQ ID NO: 2761 and 2881; SEQ ID NO: 2762 and 2882; SEQ ID NO: 2763 and 2883; SEQ ID NO: 2764 and 2884; SEQ ID NO: 2765 and 2885; SEQ ID NO: 2766 and 2886; SEQ ID NO: 2767 and 2887; SEQ ID NO: 2768 and 2888; SEQ ID NO: 2769 and 2889; SEQ ID NO: 2770 and 2890; SEQ ID NO: 2771 and 2891; SEQ ID NO: 2772 and 2892; SEQ ID NO: 2773 and 2893; SEQ ID NO: 2774 and 2894; SEQ ID NO: 2775 and 2895; SEQ ID NO: 2776 and 2896; SEQ ID NO: 2777 and 2897; SEQ ID NO: 2778 and 2898; SEQ ID NO: 2779 and 2899; SEQ ID NO: 2960 and 3340; SEQ ID NO: 2961 and 3341; SEQ ID NO: 2962 and 3342; SEQ ID NO: 2963 and 3343; SEQ ID NO: 2964 and 3344; SEQ ID NO: 2965 and 3345; SEQ ID NO: 2966 and 3346; SEQ ID NO: 2967 and 3347; SEQ ID NO: 2968 and 3348; SEQ ID NO: 2969 and 3349; SEQ ID NO: 2970 and 3350; SEQ ID NO: 2971 and 3351; SEQ ID NO: 2972 and 3352; SEQ ID NO: 2973 and 3353; SEQ ID NO: 2974 and 3354; SEQ ID NO: 2975 and 3355; SEQ ID NO: 2976 and 3356; SEQ ID NO: 2977 and 3357; SEQ ID NO: 2978 and 3358; SEQ ID NO: 2979 and 3359; SEQ ID NO: 2980 and 3360; SEQ ID NO: 2981 and 3361; SEQ ID NO: 2982 and 3362; SEQ ID NO: 2983 and 3363; SEQ ID NO: 2984 and 3364; SEQ ID NO: 2985 and 3365; SEQ ID NO: 2986 and 3366; SEQ ID NO: 2987 and 3367; SEQ ID NO: 2988 and 3368; SEQ ID NO: 2989 and 3369; SEQ ID NO: 2990 and 3370; SEQ ID NO: 2991 and 3371; SEQ ID NO: 2992 and 3372; SEQ ID NO: 2993 and 3373; SEQ ID NO: 2994 and 3374; SEQ ID NO: 2995 and 3375; SEQ ID NO: 2996 and 3376; SEQ ID NO: 2997 and 3377; SEQ ID NO: 2998 and 3378; SEQ ID NO: 2999 and 3379; SEQ ID NO: 3000 and 3380; SEQ ID NO: 3001 and 3381; SEQ ID NO: 3002 and 3382; SEQ ID NO: 3003 and 3383; SEQ ID NO: 3004 and 3384; SEQ ID NO: 3005 and 3385; SEQ ID NO: 3006 and 3386; SEQ ID NO: 3007 and 3387; SEQ ID NO: 3008 and 3388; SEQ ID NO: 3009 and 3389; SEQ ID NO: 3010 and 3390; SEQ ID NO: 3011 and 3391; SEQ ID NO: 3012 and 3392; SEQ ID NO: 3013 and 3393; SEQ ID NO: 3014 and 3394; SEQ ID NO: 3015 and 3395; SEQ ID NO: 3016 and 3396; SEQ ID NO: 3017 and 3397; SEQ ID NO: 3018 and 3398; SEQ ID NO: 3019 and 3399; SEQ ID NO: 3020 and 3400; SEQ ID NO: 3021 and 3401; SEQ ID NO: 3022 and 3402; SEQ ID NO: 3023 and 3403; SEQ ID NO: 3024 and 3404; SEQ ID NO: 3025 and 3405; SEQ ID NO: 3026 and 3406; SEQ ID NO: 3027 and 3407; SEQ ID NO: 3028 and 3408; SEQ ID NO: 3029 and 3409; SEQ ID NO: 3030 and 3410; SEQ ID NO: 3031 and 3411; SEQ ID NO: 3032 and 3412; SEQ ID NO: 3033 and 3413; SEQ ID NO: 3034 and 3414; SEQ ID NO: 3035 and 3415; SEQ ID NO: 3036 and 3416; SEQ ID NO: 3037 and 3417; SEQ ID NO: 3038 and 3418; SEQ ID NO: 3039 and 3419; SEQ ID NO: 3040 and 3420; SEQ ID NO: 3041 and 3421; SEQ ID NO: 3042 and 3422; SEQ ID NO: 3043 and 3423; SEQ ID NO: 3044 and 3424; SEQ ID NO: 3045 and 3425; SEQ ID NO: 3046 and 3426; SEQ ID NO: 3047 and 3427; SEQ ID NO: 3048 and 3428; SEQ ID NO: 3049 and 3429; SEQ ID NO: 3050 and 3430; SEQ ID NO: 3051 and 3431; SEQ ID NO: 3052 and 3432; SEQ ID NO: 3053 and 3433; SEQ ID NO: 3054 and 3434; SEQ ID NO: 3055 and 3435; SEQ ID NO: 3056 and 3436; SEQ ID NO: 3057 and 3437; SEQ ID NO: 3058 and 3438; SEQ ID NO: 3059 and 3439; SEQ ID NO: 3060 and 3440; SEQ ID NO: 3061 and 3441; SEQ ID NO: 3062 and 3442; SEQ ID NO: 3063 and 3443; SEQ ID NO: 3064 and 3444; SEQ ID NO: 3065 and 3445; SEQ ID NO: 3066 and 3446; SEQ ID NO: 3067 and 3447; SEQ ID NO: 3068 and 3448; SEQ ID NO: 3069 and 3449; SEQ ID NO: 3070 and 3450; SEQ ID NO: 3071 and 3451; SEQ ID NO: 3072 and 3452; SEQ ID NO: 3073 and 3453; SEQ ID NO: 3074 and 3454; SEQ ID NO: 3075 and 3455; SEQ ID NO: 3076 and 3456; SEQ ID NO: 3077 and 3457; SEQ ID NO: 3078 and 3458; SEQ ID NO: 3079 and 3459; SEQ ID NO: 3080 and 3460; SEQ ID NO: 3081 and 3461; SEQ ID NO: 3082 and 3462; SEQ ID NO: 3083 and 3463; SEQ ID NO: 3084 and 3464; SEQ ID NO: 3085 and 3465; SEQ ID NO: 3086 and 3466; SEQ ID NO: 3087 and 3467; SEQ ID NO: 3088 and 3468; SEQ ID NO: 3089 and 3469; SEQ ID NO: 3090 and 3470; SEQ ID NO: 3091 and 3471; SEQ ID NO: 3092 and 3472; SEQ ID NO: 3093 and 3473; SEQ ID NO: 3094 and 3474; SEQ ID NO: 3095 and 3475; SEQ ID NO: 3096 and 3476; SEQ ID NO: 3097 and 3477; SEQ ID NO: 3098 and 3478; SEQ ID NO: 3099 and 3479; SEQ ID NO: 3100 and 3480; SEQ ID NO: 3101 and 3481; SEQ ID NO: 3102 and 3482; SEQ ID NO: 3103 and 3483; SEQ ID NO: 3104 and 3484; SEQ ID NO: 3105 and 3485; SEQ ID NO: 3106 and 3486; SEQ ID NO: 3107 and 3487; SEQ ID NO: 3108 and 3488; SEQ ID NO: 3109 and 3489; SEQ ID NO: 3110 and 3490; SEQ ID NO: 3111 and 3491; SEQ ID NO: 3112 and 3492; SEQ ID NO: 3113 and 3493; SEQ ID NO: 3114 and 3494; SEQ ID NO: 3115 and 3495; SEQ ID NO: 3116 and 3496; SEQ ID NO: 3117 and 3497; SEQ ID NO: 3118 and 3498; SEQ ID NO: 3119 and 3499; SEQ ID NO: 3120 and 3500; SEQ ID NO: 3121 and 3501; SEQ ID NO: 3122 and 3502; SEQ ID NO: 3123 and 3503; SEQ ID NO: 3124 and 3504; SEQ ID NO: 3125 and 3505; SEQ ID NO: 3126 and 3506; SEQ ID NO: 3127 and 3507; SEQ ID NO: 3128 and 3508; SEQ ID NO: 3129 and 3509; SEQ ID NO: 3130 and 3510; SEQ ID NO: 3131 and 3511; SEQ ID NO: 3132 and 3512; SEQ ID NO: 3133 and 3513; SEQ ID NO: 3134 and 3514; SEQ ID NO: 3135 and 3515; SEQ ID NO: 3136 and 3516; SEQ ID NO: 3137 and 3517; SEQ ID NO: 3138 and 3518; SEQ ID NO: 3139 and 3519; SEQ ID NO: 3140 and 3520; SEQ ID NO: 3141 and 3521 ; SEQ ID NO: 3142 and 3522; SEQ ID NO: 3143 and 3523; SEQ ID NO: 3144 and 3524; SEQ ID NO: 3145 and 3525; SEQ ID NO: 3146 and 3526; SEQ ID NO: 3147 and 3527; SEQ ID NO: 3148 and 3528; and SEQ ID NO: 3149 and 3529.
It will be understood that, although the sequences in Tables 3-11, 14, and 17 are not described as modified or conjugated sequences, the ASO and/or RNA of the iRNA of the present disclosure e.g., an ASO and/or a dsRNA of the present disclosure, may comprise any one of the sequences set forth in any one of Tables 3-15, and 17-18 that is modified or conjugated differently than described therein. In other words, the present disclosure encompasses ASOs and/or dsRNA of Tables 3-15, and 17-18 which are un-modified, un-conjugated, modified, or conjugated, as described herein.
The skilled person is well aware that dsRNAs having a duplex structure of about 20 to 23 base pairs, e.g., 21, base pairs have been hailed as particularly effective in inducing RNA interference (Elbashir et al., EMBO 2001, 20:6877-6888). However, others have found that shorter or longer RNA duplex structures can also be effective (Chu and Rana (2007) RNA 14:1714-1719; Kim et al. (2005) Nat Biotech 23:222-226). In the embodiments described herein, by virtue of the nature of the oligonucleotide sequences provided in any one of Tables 7-11, dsRNAs described herein can include at least one strand of a length of minimally 21 nucleotides. It can be reasonably expected that shorter duplexes having any one of the sequences in any one of Tables 7-11, 14, and 17 minus only a few nucleotides on one or both ends can be similarly effective as compared to the ASOs and/or dsRNAs described above. Hence, ASOs and/or dsRNAs having a sequence of at least 19, 20, or more contiguous nucleotides derived from any one of the sequences of any one of Tables 3-15, and 17-18, and differing in their ability to inhibit the expression of a SLC6A19 gene by not more than about 5, 10, 15, 20, 25, or 30% inhibition from an ASO and/or dsRNA comprising the full sequence, are contemplated to be within the scope of the instant disclosure.
In addition, the ASOs and/or dsRNAs provided in Tables 3-15, and 17-18 identify a site(s) in a SLC6A19 transcript that is susceptible to ASO-mediated inhibition (including exon-skipping ASO-mediated inhibition) and/or RISC-mediated cleavage. As such, the instant disclosure further features ASOs and/or IRNAs that target within one of these sites. As used herein, an ASO and/or iRNA is said to target within a particular site of an RNA transcript if the .ASO and/or iRNA promotes inhibition (ASOs) or cleavage (iRNAs) of the transcript anywhere within that particular site Such an ASO and/or iRNA will generally include at least about 19 contiguous nucleotides from any one of the sequences provided in any one of Tables 3-15, and 17-18 coupled to additional nucleotide sequences taken from the region contiguous to the selected sequence in a SLC6A19 gene.
In some embodiments, a modulatory nucleic acid agent of the disclosure comprises a structure comprising a first wing sequence, a gap sequence, and a second wing sequence. A nucleic acid comprising such a wing-gap-wing sequence is typically referred to as a gapmer. In some embodiments, a gap sequence is flanked by a first wing sequence and a second wing sequence. In some embodiments, a gap sequence comprises about 6-10 nucleotides. In some embodiments, a wing sequence comprises one or more nucleotides. In some embodiments, a wing sequence comprises one or more modified nucleotides, e.g., as disclosed herein. In some embodiments, a gapmer acts by recruiting RNaseH. In certain embodiments, a gapmer is a chimeric antisense oligonucleotide (ASO) that contains a central sequence of phosphorothioate DNA nucleotides (which form a "DNA gap") flanked by sequences of modified RNA residues at each end. Without wishing to be bound by theory, such wing sequence modified RNA residues are believed to protect the DNA gap region from nuclease degradation, whereas the central DNA gap region allows RNase-H-mediated cleavage of a target RNA.
In some embodiments, a modulatory nucleic acid agent of the disclosure comprises an overhang. In some embodiments, an overhang is a 3’ overhang or a 5’ overhang. In some embodiments, an overhang is a 3’ overhang. In some embodiments, an overhang comprises at least 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 nucleotides. In some embodiments, a modulatory nucleic acid agent of the disclosure is double-stranded and comprises an overhang.
In some embodiments, a modulatory nucleic acid agent of the disclosure comprises at least one stem-loop structure.
An oligonucleotide of a modulatory nucleic acid agent disclosed herein typically comprises at least one sequence element that hybridizes with a target sequence. In some embodiments, a modulatory nucleic acid agent of the disclosure, e.g., an oligonucleotide, is or comprises an antisense sequence element. In some embodiments, an antisense sequence element is complementary to at least a portion of one or more of: an exon, an intron, an untranslated region, a splice junction, a promoter region, an enhancer region, or a non-coding region, e.g., in a gene transcript. In some embodiments, an antisense sequence element is complementary to a portion of a target sequence in a sense strand. In some embodiments, a modulatory nucleic acid agent of the disclosure comprises a sequence element that is at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% complementary to a target sequence in a sense strand. In some embodiments, a modulatory nucleic acid agent of the disclosure comprises a sequence element that is complementary (i.e., 100% complementary) to a target sequence in a sense strand.
In some embodiments, a modulatory nucleic acid agent of the disclosure comprises a sequence element that is at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% complementary to a target sequence in an antisense strand. In some embodiments, a modulatory nucleic acid agent of the disclosure comprises a sequence element that is complementary (i.e., 100% complementary) to a target sequence in an antisense strand.
In some embodiments, a modulatory nucleic acid agent of the disclosure comprises at least one sequence element with at least 3, at least 4, at least 5, at least 6, at least 7, at least 8, at least 9, or at least 10 contiguous nucleotides having at least 80% complementarity to a portion of a target sequence. In some embodiments, a modulatory nucleic acid agent of the disclosure comprises at least one sequence element with at least 3, at least 4, at least 5, at least 6, at least 7, at least 8, at least 9, or at least 10 contiguous nucleotides having at least 85% complementarity to a portion of a target sequence. In some embodiments, a modulatory nucleic acid agent of the disclosure comprises at least one sequence element with at least 3, at least 4, at least 5, at least 6, at least 7, at least 8, at least 9, or at least 10 contiguous nucleotides having at least 90% complementarity to a portion of a target sequence. In some embodiments, a modulatory nucleic acid agent of the disclosure comprises at least one sequence element with at least 3, at least 4, at least 5, at least 6, at least 7, at least 8, at least 9, or at least 10 contiguous nucleotides having at least 95% complementarity to a portion of a target sequence. In some embodiments, a modulatory nucleic acid agent of the disclosure comprises at least one sequence element with at least 3, at least 4, at least 5, at least 6, at least 7, at least 8, at least 9, or at least 10 contiguous nucleotides having at least 96% complementarity to a portion of a target sequence. In some embodiments, a modulatory nucleic acid agent of the disclosure comprises at least one sequence element with at least 3, at least 4, at least 5, at least 6, at least 7, at least 8, at least 9, or at least 10 contiguous nucleotides having at least 97% complementarity to a portion of a target sequence. In some embodiments, a modulatory nucleic acid agent of the disclosure comprises at least one sequence element with at least 3, at least 4, at least 5, at least 6, at least 7, at least 8, at least 9, or at least 10 contiguous nucleotides having at least 98% complementarity to a portion of a target sequence. In some embodiments, a modulatory nucleic acid agent of the disclosure comprises at least one sequence element with at least 3, at least 4, at least 5, at least 6, at least 7, at least 8, at least 9, or at least 10 contiguous nucleotides having at least 99% complementarity to a portion of a target sequence. In some embodiments, a modulatory nucleic acid agent of the disclosure comprises at least one sequence element with at least 3, at least 4, at least 5, at least 6, at least 7, at least 8, at least 9, or at least 10 contiguous nucleotides having 100% complementarity to a portion of a target sequence.
In some embodiments, a modulatory nucleic acid agent of the disclosure comprises 2 or more sequence elements with at least 3, at least 4, at least 5, at least 6, at least 7, at least 8, at least 9, or at least 10 contiguous nucleotides having at least 80% complementarity to a portion of a target sequence.
In some embodiments, a modulatory nucleic acid agent of the disclosure binds to at least a portion of a target via Watson-Crick base pairing. In some embodiments, a modulatory nucleic acid agent of the disclosure binds to at least a portion of a target via Hoogsteen base pairing and/or other non-cannonical base pairing.
In general, an “iRNA” includes ribonucleotides with chemical modifications. Such modifications may include all types of modifications disclosed herein or known in the art. Any such modifications, as used in a dsRNA molecule, are encompassed by “iRNA” for the purposes of this specification and claims.
In certain embodiments of the instant disclosure, inclusion of a deoxy-nucleotide if present within an RNAi agent can be considered to constitute a modified nucleotide. dsRNA agents of the disclosure include those having an antisense strand including a region of complementarity which is complementary to at least a part of an mRNA formed in the expression of a SLC6A19 gene. In embodiments, the region of complementarity is about 19-30 nucleotides in length (e.g., about 30, 29, 28, 27, 26, 25, 24, 23, 22, 21, 20, or 19 nucleotides in length). Upon contact with a cell expressing the SLC6A19 gene, the iRNA inhibits the expression of the SLC6A19 gene (e.g., a human, a primate, a non-primate, or a rat SLC6A19 gene) by at least about 50% as assayed by, for example, a PCR or branched DNA (bDNA)-based method, or by a protein-based method, such as by immunofluorescence analysis, using, for example, western blotting or flow cytometric techniques. In certain embodiments, inhibition of expression is determined by a qPCR method known in the art with the siRNA at, e.g., a 10 nM concentration, in an appropriate organism cell line. In certain embodiments, inhibition of expression in vivo is determined by knockdown of the human gene in a rodent expressing the human gene, e.g., a mouse or an AAV-infected mouse expressing the human target gene, e.g., when administered as single dose, e.g., at 3 rng/kg at the nadir of RNA expression.
A dsRNA includes two RNA strands that are complementary and hybridize to form a duplex structure under conditions in which the dsRNA will be used. One strand of a dsRNA (the antisense strand) includes a region of complementarity that is substantially complementary/, and generally fully complementary’, to a target sequence. The target sequence can be derived from the sequence of an raRNA formed during the expression of a SLC6A19 gene The other strand (the sense strand) includes a region that is complementary to the antisense strand, such that the two strands hybridize and form a duplex structure when combined under suitable conditions. As described elsewdiere herein and as known in the art, the complementary' sequences of a dsRNA can also be contained as self-complementary regions of a single nucleic acid molecule, as opposed to being on separate oligonucleotides.
Generally, the duplex structure is 15 to 30 base pairs in length, e.g., 15-29, 15-28, 15-27,
15-26, 15-25, 15-24, 15-23, 15-22, 15-21 , 15-20, 15-19, 15-18, 15-17, 18-30, 18-29, 18-28, 18-27,
18-26, 18-25, 18-24, 18-23, 18-22, 18-21, 18-20, 19-30, 19-29, 19-28, 19-27, 19-26, 19-25, 19-24,
19-23, 19-22, 19-21 , 19-20, 20-30, 20-29, 20-28, 20-27, 20-26, 20-25, 20-24,20-23, 20-22, 20-21,
21-30, 21-29, 21-28, 21-27, 21-26, 21-25, 21-24, 21-23, or 21-22 base pairs in length. In certain embodiments, the duplex structure is 18 to 25 base pairs in length, e.g., 18-25, 18-24, 18-23, 18- 22, 18-21, 18-20, 19-25, 19-24, 19-23, 19-22, 19-21, 19-20, 20-25, 20-24,20-23, 20-22, 20-21, 21- 25, 21-24, 21-23, 21-22, 22-25, 22-24, 22-23, 23-25, 23-24 or 24-25 base pairs in length, for example, 19-21 base pairs in length. Ranges and lengths intermediate to the above recited ranges and lengths are also contemplated to be part of the disclosure.
Similarly, the region of complementarity to the target sequence is 15 to 30 nucleotides in length, e.g., 15-29, 15-28, 15-27, 15-26. 15-25. 15-24, 15-23, 15-22, 15-21, 15-20, 15-19. 15-18, 15-17, 18-30, 18-29, 18-28, 18-27, 18-26, 18-25, 18-24, 18-23, 18-22, 18-21, 18-20, 19-30, 19-29,
19-28, 19-27, 19-26, 19-25, 19-24, 19-23, 19-22, 19-21, 19-20, 20-30, 20-29, 20-28, 20-27, 20-26,
20-25, 20-24,20-23, 20-22, 20-21, 21-30, 21-29, 21-28, 21-27, 21-26, 21-25, 21-24, 21-23, or 21- 22 nucleotides in length, for example 19-23 nucleotides in length or 21-23 nucleotides in length. Ranges and lengths intermediate to the above recited ranges and lengths are also contemplated to be part of the disclosure.
Ln some embodiments, the duplex structure is 19 to 30 base pairs in length. Similarly, the region of complementarity to the target sequence is 19 to 30 nucleotides in length.
In some embodiments, the dsRNA is about 19 to about 23 nucleotides in length, or about 25 to about 30 nucleotides in length. In general, the dsRNA is long enough to serve as a substrate for the Dicer enzyme. For example, it is well-known in the art that dsRNAs longer than about 21- 23 nucleotides in length may serve as substrates for Dicer. As the ordinarily skilled person will also recognize, the region of an RNA targeted for cleavage will most often be part of a larger RNA molecule, often an mRNA molecule. Where relevant, a “part” of an mRNA target is a contiguous sequence of an mRNA target of sufficient length to allow it to be a substrate for RNAi-directed cleavage (i.e, cleavage through a RISC pathway).
One of skill in the art will also recognize that the duplex region is a primary functional portion of a dsRNA, e.g., a duplex region of about 19 to about 30 base pairs, e.g., about 19-30, 19- 29, 19-28, 19-27, 19-26, 19-25, 19-24, 19-23, 19-22, 19-21, 19-20, 20-30, 20-29, 20-28, 20-27, 20-26, 20-25, 20-24,20-23, 20-22, 20-21, 21-30, 21-29, 21-28, 21-27, 21-26, 21-25, 21-24, 21-23, or 21-22 base pairs. Thus, in one embodiment, to the extent that it becomes processed to a functional duplex, of e.g., 15-30 base pairs, that targets a desired RNA for cleavage, an RNA molecule or complex of RNA molecules having a duplex region greater than 30 base pairs is a dsRNA. Thus, an ordinarily skilled artisan will recognize that in one embodiment, a miRNA is a dsRNA In another embodiment, a dsRNA is not a naturally occurring miRNA. In another embodiment, an iRNA agent useful to target SLC6A19 gene expression is not generated in the target cell by cleavage of a larger dsRNA.
A dsRNA as described herein can further include one or more single-stranded nucleotide overhangs e.g., 1-4, 2-4, 1-3, 2-3, 1, 2, 3, or 4 nucleotides. dsRNAs having at least one nucleotide overhang can have superior inhibitory properties relative to their blunt-ended counterparts. A nucleotide overhang can comprise or consist of a nucleotide/nucleoside analog, including a deoxynucleotide/nucieoside. The overhang(s) can be on the sense strand, the antisense strand, or any combination thereof. Furthermore, the nucleotide(s) of an overhang can be present on the 5'- end, 3'-end, or both ends of an antisense or sense strand of a dsRNA. A dsRNA can be synthesized by standard methods known in the art. Double stranded RNAi compounds of the present disclosure may be prepared using a two-step procedure. First, the individual strands of the double stranded RNA molecule are prepared separately. Then, the component strands are annealed. The individual strands of the siRNA compound can be prepared using solution-phase or solid-phase organic synthesis or both Organic synthesis offers the advantage that the oligonucleotide strands comprising unnatural or modified nucleotides can be easily prepared. Similarly, single-stranded oligonucleotides of the present disclosure can be prepared using solution-phase or solid-phase organic synthesis or both.
Modified Modulatory Nucleic Acids of the Disclosure
In some embodiments, a modulatory nucleic acid agent includes one or more modified (relative to canonical DNA and/or RNA) nucleotides. In some embodiments, a modified nucleotide comprises one or more of: a modified backbone, a modified nucleobase, a modified sugar (e.g., a modified ribose, or a modified deoxyribose), or any combination thereof. In some embodiments, a modified nucleotide may be or comprise one or more naturally occurring modifications; in some embodiments a modified nucleotide may be or comprise one or more non-naturally-occurring modifications.
In some embodiments, a modulatory nucleic acid agent comprises one or more linkages that is not a phosphodi ester linkage (e.g., that is or comprises a phosphorothioate linkage or a phosphorodiamidate linkage).
In some embodiments, a modulatory nucleic acid agent comprises one or more morpholino subunits linked together by a phosphorus-containing linkage. In some embodiments, one or more morpholino subunits in an oligonucleotide agent is joined by a phosphorodiamidate linkage. The synthesis, structures, and binding characteristics of morpholino oligomers are detailed in U.S. Pat. Nos. 5,698,685, 5,217,866, 5,142,047, 5,034,506, 5,166,315, 5,521,063, and 5,506,337, and PCT Appn. Nos. PCT/US07/11435 (cationic linkages) and U.S. Ser. No. 08/012,804 (improved synthesis), all of which are incorporated herein by reference. Morpholino subunits linked by phosphorodiamidate linkages are disclosed in US Patent 11,071,749 the entire contents of which are hereby incorporated by reference. In some embodiments, a modulatory nucleic acid agent is or comprises a PMO. In some embodiments, a PMO is substantially uncharged, e.g., has a neutral charge.
In some embodiments, a modulatory nucleic acid agent has a negative charge. In some embodiments, a modulatory nucleic acid agent is substantially uncharged, e.g., has a neutral charge.
Those skilled in the art, reading the present disclosure, will appreciate that, in some embodiments, a modulatory nucleic acid agent of the present disclosure may include one or more DNA residues or analogs thereof, one or more RNA residues or analogs thereof, and/or combinations thereof. Furthermore, such skilled person will appreciate that, in some embodiments, a modulatory nucleic acid agent of the instant disclosure may include one or more, or entirely, phosphodiester linkages, phosphorothioate linkages, or other suitable linkages.
In some embodiments, a modulatory nucleic acid agent comprises natural residues, e.g., DNA residues and/or RNA residues.
In some embodiments a modulatory nucleic acid agent comprises one or more analogs, e g., DNA analogs and/or RNA analogs.
In some embodiments, a modulatory nucleic acid agent comprises DNA residues and/or RNA residues, e.g., natural residues or analogs.
In some embodiments, a modulatory nucleic acid of the disclosure comprises one or more chiral centers (e.g., as may be present in, for example, a phosphorothioate linkage). In some embodiments, a preparation of a modulatory nucleic acid having a chiral center is stereopure with respect to that center in that it includes only one stereoisomer of that center. In some embodiments, both stereoisomers are present. In some embodiments, the preparation represents a racemic mixture of stereoisomers at that position. In some embodiments, a preparation of a modulatory nucleic acid having more than one chiral linkage may be stereopure with respect to one or more centers and mixed (e.g., racemic) with respect to one or more others. In some embodiments, a preparation may be stereopure at all chiral centers. In some embodiments, a preparation may be racemic (e.g., at all chiral centers or overall).
In some embodiments, a modulatory nucleic acid of the disclosure comprises one or more modified nucleotides. In some embodiments, a modified nucleotide comprises one or more of: a modified backbone, a modified nucleobase, a modified ribose, a modified deoxyribose, or any combination thereof.
In some embodiments, a modified nucleotide is chosen from: a 2'-O-methyl modified nucleotide, a 5-methylcytidine, a 5-methyluridine, a nucleotide comprising a 5 '-phosphorothioate group, a morpholino nucleotide (e.g., a PMO), a terminal nucleotide linked to a cholesteryl derivative or a dodecanoic acid bisdecylamide group, a 2'-deoxy-2'-fluoro modified nucleotide, a 2'-deoxy-modified nucleotide, a locked nucleotide, an abasic nucleotide, 2'-amino-modified nucleotide, 2'-alkyl-modified nucleotide, morpholino nucleotide (e.g., PMO), a phosphoramidate, a phosphoryl guanidine (PN) based backbone, or a non-natural base comprising nucleotide, or any combination thereof.
In some embodiments, a modified nucleobase comprises a C7-modified deaza-adenine, a C7-modified deaza-guanosine, a C5-modified cytosine, a C5-modified uridine, N1 -methylpseudouridine (mly), 1-ethyl-pseudouridine (elvp), 5-methoxy-uridine (mo5U), 5-methyl- cytidine (m5C), pseudouridine (y), 5-methoxymethyl uridine, 5-methylthio uridine, 1- methoxymethyl pseudouridine, 5-methyl cytidine, 5-methoxy cytidine, or any combination thereof.
In some embodiments, a modified sugar (e g., a modified ribose, or a modified deoxyribose) comprises: a 2’fluoro modification, a 2’-O-methyl (2’0Me) modification, a locked nucleic acid (LNA), a 2’ -fluoro arabinose nucleic acid (FANA), a hexitol nucleic acid (HNA), a 2’ O-m ethoxy ethyl (2’MOE) modification, or any combination thereof.
In some embodiments, a modified backbone comprises a phosphorothioate (PS) modification, a phosphoryl guanidine (PN) modification, a borano-phosphate modification, an alkyl phosphonate nucleic acid (phNA), a peptide nucleic acid (PNA), or any combination thereof.
In some embodiments, a modulatory nucleic acid agent of the disclosure comprises one or more modifications, e g., to a 5’ end of an oligonucleotide. In some embodiments, a modulatory nucleic acid agent of the disclosure comprises a 5’ amino modification.
In some embodiments, a modulatory nucleic acid agent of the disclosure is partially modified (e.g., at least 5%) for a particular modification, e.g., throughout the length of a sequence.
In some embodiments, a modulatory nucleic acid agent of the disclosure is fully modified for a particular modification throughout the length of a sequence.
In some embodiments, at least 5% of a particular nucleotide (e.g., A, G, C, T, or U) is modified in an oligonucleotide.
In some embodiments, all (e.g., 100%) of a particular nucleotide (e.g., A, G, C, T, or U) is modified in an oligonucleotide.
In certain embodiments, the nucleotide sequence of a modulatory nucleic acid agent of the present disclosure e g., an ASO and/or dsRNA, is un-modified, and does not comprise, e.g , chemical modifications or conjugations known in the art and described herein. In other embodiments, the nucleotid e sequence of a modulatory nucleic acid agent of the present disclosure e.g., an ASO and/or dsRNA, is chemically modified to enhance stability or other beneficial characteristics. In certain embodiments of the present disclosure, substantially all of the nucleotides of an ASO and/or dsRNA of the present disclosure are modified. In other embodiments of the present disclosure, all of the nucleotides of an ASO and/or dsRNA or substantially all of the nucleotides of an ASO and/or dsRNA are modified, i.e., not more than 5, 4, 3, 2, or 1 unmodified nucleotides are present in a strand of the ASO and/or dsRNA.
The nucleic acids featured in the present disclosure can be synthesized or modified by methods well established in the art, such as those described in “Current protocols in nucleic acid chemistry',” Beaucage, S.L. et al. (Edrs.), John Wiley & Sons, Inc., New York, NY, USA, which is hereby incorporated herein by reference. Modifications include, for example, end modifications, e.g., 5’-end modifications (phosphorylation, conjugation, inverted linkages) or 3’-end modifications (conjugation, DNA nucleotides, inverted linkages, etc.); base modifications, e.g., replacement with stabilizing bases, destabilizing bases, or bases that base pair with an expanded repertoire of partners, removal of bases (abasic nucleotides), or conjugated bases; sugar modifications (e g., at the 2’-position or 4’-position) or replacement of the sugar; or backbone modifications, including modification or replacement of the phosphodiester linkages. Specific examples of ASO and/or dsRNA compounds useful in the embodiments described herein include, but are not limited to RNAs containing modified backbones or no natural internucleoside linkages. RN As having modified backbones include, among others, those that do not have a phosphorus atom in the backbone. For the purposes of this specification, and as sometimes referenced in the art, modified RNAs that do not have a phosphorus atom in their intemucleoside backbone can also be considered to be oligonucleosides. In some embodiments, a modified ASO and/or dsRNA will have a phosphorus atom in its internucleoside backbone.
Modified RNA backbones include, for example, phosphorothi oates, chiral phosphorothioates, phosphorodithioates, phosphotriesters, aminoalkylphosphotriesters, methyl and other alkyl phosphonates including 3'-alkylene phosphorates and chiral phosphorates, phosphinates, phosphoramidates including 3 '-amino phosphoramidate and aminoalkylphosphoramidates, thionophosphoramidates, thionoalkylphosphonates, thionoalkylphosphotriesters, and boranophosphates having normal 3'-5' linkages, 2'-5‘-linked analogs of these, and those having inverted polarity wherein the adjacent pairs of nucleoside units are linked 3'-5' to 5'-3! or 2'-5' to 5 -2'. Various salts, mixed salts and free acid forms are also included. In some embodiments of the present disclosure, the ASO and/or dsRNA agents of the present disclosure are in a free acid form. In other embodiments of the present disclosure, the ASO and/or dsRNA agents of the present disclosure are in a salt form. In one embodiment, the ASO and/or dsRNA agents of the present disclosure are in a sodium salt form. In certain embodiments, when the ASO and/or dsRNA agents of the present disclosure are in the sodium salt form, sodium ions are present in the agent as counterions for substantially all of the phosphodiester and/or phosphorothiotate groups present in the agent. Agents in which substantially all of the phosphodiester and/or phosphorothioate linkages have a sodium counterion include not more than 5, 4, 3, 2, or 1 phosphodiester and/or phosphorothioate linkages without a sodium counterion. In some embodiments, when the ASO and/or dsRNA agents of the present disclosure are in the sodium salt form, sodium ions are present in the agent as counterions for all of the phosphodiester and/or phosphorothiotate groups present in the agent.
Representative U.S. Patents that teach the preparation of the above phosphorus-containing linkages include, but are not limited to, U.S. Patent Nos.3, 687, 808; 4,469,863; 4,476,301, 5,023,243; 5,177,195; 5,188,897, 5,264,423, 5,276,019; 5,278,302; 5,286,717; 5,321,131,
5,399,676; 5,405,939; 5,453,496; 5,455,233; 5,466,677; 5,476,925; 5,519,126; 5,536,821;
5,541,316; 5,550,1 1 1; 5,563,253; 5,571 ,799; 5,587,361; 5,625,050; 6,028,188; 6,124,445;
6,160,109; 6,169,170; 6,172,209; 6, 239,265; 6,277,603; 6,326,199; 6,346,614; 6,444,423, 6,531,590, 6,534,639, 6,608,035, 6,683,167, 6,858,715; 6,867,294; 6,878,805, 7,015,315,
7,041,816; 7,273,933; 7,321,029; and U.S. Pat RE39464, the entire contents of each of which are hereby incorporated herein by reference.
Modified RNA backbones that do not include a phosphorus atom therein have backbones that are formed by short chain alkyl or cycloalkyl internucleoside linkages, mixed heteroatoms and alkyl or cycloalkyl internucleoside linkages, or one or more short chain heteroatomic or heterocyclic internucleoside linkages. These include those having morpholino linkages (formed in part from the sugar portion of a nucleoside); siloxane backbones; sulfide, sulfoxide and sulfone backbones; formacetyl and thioformacetyl backbones; methylene formacetyl and thioformacetyl backbones; alkene containing backbones; sulfamate backbones, methyleneimino and methylenehydrazino backbones; sulfonate and sulfonamide backbones; amide backbones; and others having mixed N, O, S, and CH.: component parts.
Representative U.S. Patents that teach the preparation of the above oligonucleosides include, but are not limited to, U.S. Patent Nos.5, 034, 506; 5,166,315; 5,185,444; 5,214,134; 5,216,141; 5,235,033, 5,64,562; 5,264,564; 5,405,938, 5,434,257; 5,466,677, 5,470,967; 5,489,677; 5,541,307; 5,561,225; 5,596,086; 5,602,240; 5,608,046; 5,610,289; 5,618,704; 5,623,070, 5,663,312; 5,633,360; 5,677,437; and 5,677,439, the entire contents of each of which are hereby incorporated herein by reference.
Suitable RNA mimetics are contemplated for use in ASO and/or dsRNA agents provided herein, in which both the sugar and the internucleoside linkage, i ,e., the backbone, of the nucleotide units are replaced with novel groups. The base units are maintained for hybridization with an appropriate nucleic acid target compound One such oligomeric compound in which an RNA mimetic that has been shown to have excellent hybridization properties is referred to as a peptide nucleic acid (PNA). In PNA compounds, the sugar backbone of an RNA is replaced with an amide containing backbone, in particular an aminoethyl glycine backbone. The nucleobases are retained and are bound directly or indirectly to aza nitrogen atoms of the amide portion of the backbone. Representative US patents that teach the preparation of PNA compounds include, but are not limited to, U.S. Patent Nos.5, 539, 082; 5,714,331; and 5,719,262, the entire contents of each of which are hereby incorporated herein by reference. Additional PNA compounds suitable for use in the ASO and/or dsRNA agents of the present disclosure are described in, for example, in Nielsen et al., Science, 1991, 254, 1497-1500.
In certain embodiments, modulatory7 nucleic acids of the disclosure include RNAs with phosphorothioate backbones and oligonucleosides with heteroatom backbones, and in particular - -CH?.— NH— CH?.-, — CH?.— N(CH?)— O--CH2“-[know’n as a methylene (methylimino) or MMI backbone], -CH2-O-N(CH3)-CH2-, --CH2--N(CH;0--N(CH3)--CH2-- and -N(CH3)-CH2- CH2--[wherein the native phosphodiester backbone is represented as — O--P— O--CH2— ] of U.S. Patent No.5,489,677, and the amide backbones of U.S. Patent No.5,602,240. In some embodiments, the RNAs featured herein have morpholino backbone structures of U S. Patent No. 5,034,506.
Modified RNAs can also contain one or more substituted sugar moieties. The modulatory nucleic acids, e.g., ASO and/or dsRNA agents, featured herein can include one of the following at the 2'-position: OH; F; O-, S-, or N-alkyl; O-, S-, or N-alkenyl; O-, S- or N-alkynyl; or O-alkyl-O- alkyl, wherein the alkyl, alkenyl and alkynyl can be substituted or unsubstituted Ci to Cio alkyl or C2 to Cio alkenyl and alkynyl. Exemplary/ suitable modifications include O[(CH2)nO]mCH3, O(CH2)nOCH3, O(CH2)nNH2, O(CH2)nCH3, O(CH2)nONH2, and O(CH2)nON[(CH2)nCH3)]2, where n and m are from 1 to about 10. In other embodiments, dsRNAs include one of the following at the 2' position: Ci to Cio lower alkyl, substituted lower alkyl, alkaryi, aralkyl, O-alkaryd or O- aralkyl, SH, SCIU, OCN, Cl, Br, CN, CF3, OCF3, SOCH3, SO2CH3, ONO2, NO2, N3, NHz, heterocycloalkyl, heterocycloalkaryl, ami noalkyl ami no, polyalkylamino, substituted silyl, an RNA cleaving group, a reporter group, an intercalator, a group for improving the pharmacokinetic properties of an ASO and/or an iRNA, or a group for improving the pharmacodynamic properties of an ASO and/or an iRNA, and other substituents having similar properties. In some embodiments, the modification includes a 2'-methoxyethoxy (2'-O— CH2CH2OCH3, also known as 2'-O-(2-methoxyethyl) or 2 -MOE) (Martin et al., Helv. Chim. Acta, 1995, 78:486-504) i.e., an alkoxy-alkoxy group. Another exemplary modification is 2‘-dimethylaminooxyethoxy, i.e., a O(CH2)2ON(CH3)2 group, also known as 2 -DMAOE, as described in examples herein below, and 2'-dimethylaminoethoxyethoxy (also known in the art as 2'-O-dimethylaminoethoxyethyl or 2'- DMAEOE), i.e., 2 -0— CH2— O--CH2— N(CH2)2. Further exemplary modifications include: 5’-Me- 2’-F nucleotides, 5’-Me-2’-OMe nucleotides, 5 ’-Me-2’ -deoxynucleotides, (both R and S isomers in these three families); 2’-alkoxyalkyl; and 2’-NM A (N-methylacetamide).
Other modifications include 2'-methoxy (2!-OCH3), 2'-aminopropoxy (2'~ OCH2CH2CH2NH2) and 2'-fluoro (2'-F). Similar modifications can also be made at other positions on the RNA of an ASO and/or iRNA, particularly the 3' position of the sugar on the 3' terminal nucleotide or in 2'-5' linked dsRNAs and the 5' position of 5' terminal nucleotide. ASOs and/or iRNAs can also have sugar mimetics such as cyclobutyl moieties in place of the pentofuranosyl sugar. Representative US patents that teach the preparation of such modified sugar structures include, but are not limited to, U.S. Patent Nos.4, 981 , 957; 5,118,800; 5,319,080; 5,359,044, 5,393,878; 5,446,137; 5,466,786; 5,514,785; 5,519,134; 5,567,811; 5,576,427; 5,591,722; 5,597,909; 5,610,300; 5,627,053; 5,639,873; 5,646,265; 5,658,873; 5,670,633; and 5,700,920, certain of which are commonly owned with the instant application. The entire contents of each of the foregoing are hereby incorporated herein by reference. An ASO and/or an iRNA can also include nucleobase (often referred to in the art simply as “base”) modifications or substitutions. As used herein, “unmodified” or “natural” nucleobases include the purine bases adenine (A) and guanine (G), and the pyrimidine bases thymine (T), cytosine (C), and uracil (U). Modified nucleobases include other synthetic and natural nucleobases such as deoxy-thymine (dT), 5-methylcytosine (5-me-C), 5-hydroxymethyl cytosine, xanthine, hypoxanthine, 2-aminoadenine, 6-methyl and other alkyl derivatives of adenine and guanine, 2- propyl and other alkyl derivatives of adenine and guanine, 2-thiouracil, 2-thiothymine and 2- thiocytosine, 5-halouracil and cytosine, 5-propynyl uracil and cytosine, 6-azo uracil, cytosine and thymine, 5-uracil (pseudouracil), 4-thiouracil, 8-halo, 8-amino, 8-thiol, 8-thioalkyl, 8-hydroxyl anal other 8-substituted adenines and guanines, 5-halo, particularly 5-bromo, 5-trifluoroniethyl and other 5-substituted uracils and cytosines, 7-methylguanine and 7-methyladenine, 8-azaguanine and 8-azaadenine, 7-deazaguanine and 7-daazaadenine and 3-deazaguanine and 3-deazaadenine. Further nucleobases include those disclosed in U.S. Pat. No.3, 687, 808, those disclosed in Modified Nucleosides in Biochemistry, Biotechnology and Medicine, Herdewijn, P. ed. Wiley-VCH, 2008; those disclosed in The Concise Encyclopedia Of Polymer Science And Engineering, pages 858- 859, Kroschwitz, J. L, ed. John Wiley & Sons, 1990, these disclosed by Englisch et al., Angewandte Chemi e, International Edition, 1991, 30, 613, and those disclosed by Sanghvi, Y S., Chapter 15, dsRNA Research and Applications, pages 289-302, Crooke, S. T. and Lebleu, B., Ed., CRC Press, 1993. Certain of these nucleobases are particularly useful for increasing the binding affinity of the oligomeric compounds featured in the present disclosure These include 5- substituted pyrimidines, 6-azapyrimidines and N-2, N-6 and 0-6 substituted purines, including 2- aminopropyladenine, 5-propynyluracil and 5-propynyicytosine.5-methylcytosine substitutions have been shown to increase nucleic acid duplex stability by 0.6-1.2°C (Sanghvi, Y S., Crooke, S. T. and Lebleu, B., Eds., dsRNA Research and Applications, CRC Press, Boca Raton, 1993, pp.276-278) and are exemplary base substitutions, even more particularly when combined with 2'- O-methoxyethyl sugar modifications.
Representative U.S. Patents that teach the preparation of certain of the above noted modified nucleobases as well as other modified nucleobases include, but are not limited to, the above noted U.S. Patent Nos.3, 687, 808, 4,845,205; 5,130,30; 5,134,066; 5,175,273; 5,367,066; 5,432,272, 5,457,187, 5,459,255, 5,484,908, 5,502,177, 5,525,711, 5,552,540, 5,587,469,
5,594,121, 5,596,091; 5,614,617; 5,681,941; 5,750,692; 6,015,886; 6,147,200; 6,166,197; 6,222,025; 6,235,887; 6,380,368; 6,528,640; 6,639,062; 6,617,438; 7,045,610; 7,427,672; and 7,495,088, the entire contents of each of which are hereby incorporated herein by reference.
The RNA of an ASO or iRNA agent can also be modified to include one or more locked nucleic acids (LNA). A locked nucleic acid is a nucleotide having a modified ribose moiety in which the ribose moiety comprises an extra bridge connecting the 2' and 4' carbons. This structure effectively "locks" the ribose in the 3'-endo structural conformation. The addition of locked nucleic acids to siRNAs has been shown to increase siRNA stability in serum, and to reduce off-target effects (Elmen, J. et al., (2005) Nucleic Acids Research 33(1 ):439-447; Mook, OR. et al., (2007) Mol Cane Ther 6(3): 833-843 ; Grunweller, A. et al., (2003) Nucleic Acids Research 31(12):3185- 3193).
The RNA of an ASO or iRNA agent can also be modified to include one or more constrained ethyl nucleotides. As used herein, a "constrained ethyl nucleotide" or "cEt" is a locked nucleic acid comprising a bicyclic sugar moiety comprising a 4'-CH(CH3)-O-2' bridge. In one embodiment, a constrained ethyl nucleotide is in the S conformation referred to herein as “S-cEt.” An ASO or iRNA agent of the present disclosure may also include one or more “conformationally restricted nucleotides” (“CRN”). CRN are nucleotide analogs with a linker connecting the C2’and C4’ carbons of ribose or the C3 and -C5’ carbons of ribose. CRN lock the ribose ring into a stable conformation and increase the hybridization affinity to mRNA. The linker is of sufficient length to place the oxygen in an optimal position for stability and affinity resulting in less ribose ring puckering.
Representati ve publications that teach the preparati on of certain of the above noted CRN 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 herein by reference.
In some embodiments, an ASO or iRNA agent of the present disclosure comprises one or more monomers that are UNA (unlocked nucleic acid) nucleotides. UNA is unlocked acyclic nucleic acid, wherein any of the bonds of the sugar has been removed, forming an unlocked "sugar" residue In one example, UNA also encompasses monomer with bonds between CT-C4’ have been removed (i.e. the covalent carbon-oxygen-carbon bond between the CT and C4' carbons). In another example, the C2 -C3' bond (i.e. the covalent carbon-carbon bond between the C2' and C.3' carbons) of the sugar has been removed (see Nuc. Acids Symp. Series, 52, 133-134 (2008) and Flutter et ai., Mol Biosyst, 2009, 10, 1039 hereby incorporated by reference). Representative U S. publications that teach the preparation of UNA include, but are not limited to, U.S. Patent No.8,314,227; and U.S. Patent Publication Nos.2013/0096289; 2013/001 1922; and 201 1/0313020, the entire contents of each of which are hereby incorporated herein by reference.
Potentially stabilizing modifications to the ends of RNA molecules can include N- (acetylaminocaproyl )-4-hydroxyproiinol (Hyp-Ce-NHAc), N-(caproyl-4-hydroxyprolinol (Hyp- Ce), N-(acetyl-4-hydroxyprolinol (Hyp-NHAc), thymidine-2’-O-deoxythymidine (ether), N- (aminocaproyl)-4-hydroxyprolinol (Hyp-Ce-amino), 2-docosanoyl-uridine-3 "-phosphate, inverted base dT(idT) and others. Disclosure of this modification can be found in PCT Publication No. WO 2011/005861.
Other modifications of the nucleotides of an ASO or iRNA agent of the present disclosure include a 5’ phosphate or 5’ phosphate mimic, e.g., a 5’-terminal phosphate or phosphate mimic on the antisense strand of an ASO or iRNA agent. Suitable phosphate mimics are disclosed in, for example U.S Patent Publication No. 2012/0157511, the entire contents of which are incorporated herein by reference.
In an aspect of the present disclosure, an agent for use in the methods and compositions of the present disclosure is a single-stranded antisense oligonucleotide (ASO) molecule that inhibits a target mRNA via an antisense inhibition mechanism. The single-stranded antisense oligonucleotide molecule is complementary to a sequence within the target. mRNA. The singlestranded antisense oligonucleotides can inhibit translation in a stoichiometric manner by base pairing to the mRNA and physically obstructing the translation machinery, see Dias, N. et al., (2002) Mol Cancer Ther 1 :347-355. The single-stranded antisense oligonucleotide molecule may be about 14 to about 30 nucleotides in length and have a sequence that i s complementary to a target sequence. For example, the single-stranded antisense oligonucleotide molecule may comprise a sequence that is at least about 14, 15, 16, 17, 18, 19, 20, or more contiguous nucleotides from anyone of the antisense sequences described herein.
The phrase “contacting a cell with an iRNA,” such as a dsRNA, as used herein, includes contacting a cell by any possible means. Contacting a cell with an iRNA includes contacting a cell in vitro with the iRNA or contacting a cell in vivo with the iRN.A. The contacting may be done directly or indirectly. Thus, for example, the iRNA may be put into physical contact with the cell by the individual performing the method, or alternatively, the iRNA may be put into a situation that will permit or cause it to subsequently come into contact with the cell.
Contacting a cell in vitro may be done, for example, by incubating the cell with the iRNA. Contacting a cell in vivo may be done, for example, by injecting the iRNA into or near the tissue where the cell is located, or by injecting the iRNA into another area, e.g., the bloodstream or the subcutaneous space, such that the agent will subsequently reach the tissue where the cell to be contacted is located. For example, the iRNA may contain or be coupled to a ligand, e.g., a Megalin- binding, Cubilin-binding, or other cell surface factor-binding moiety, that directs the iRNA to a site of interest, e.g., kidney cells. Combinations of in vitro and in vivo methods of contacting are also possible For example, a cell may also be contacted in vitro with an iRNA and subsequently transplanted into a subject.
In certain embodiments, contacting a cell with an ASO or iRNA agent includes “introducing” or “delivering the ASO or iRNA agent into the cell” by facilitating or effecting uptake or absorption into the cell. Absorption or uptake of an ASO or iRNA agent can occur through unaided diffusion or active cellular processes, or by auxiliary' agents or devices. Introducing an ASO or iRNA agent into a cell may be in vitro or in vivo. For example, for in vivo introduction, an ASO or iRNA agent can be injected into a tissue site or administered systemically. In vitro introduction into a cell includes methods known in the art such as electroporation and lipofection. Further approaches are described herein below or are known in the art
The term “lipid nanoparticle” or “LNP” is a vesicle comprising a lipid layer encapsulating a pharmaceutically active molecule, such as a nucleic acid molecule, e g., an iRNA or a plasmid from which an iRNA is transcribed. LNPs are described in, for example, U.S. Patent Nos.6, 858, 225, 6,815,432, 8,158,601, and 8,058,069, the entire contents of which are hereby incorporated herein by reference.
SLC6A19 mRNA and B°AT1 Protein
An exemplary' mRNA sequence of Homo sapiens solute carrier family 6 member 19 (SLC6A19) is provided below' (accession number NM_001003841.3; abbreviated elsewhere herein as NM 1003841.3): actcgccctccagcttctgccctgcctgctgtgtgcggagccgtccagcgaccaccatggtgaggctcgtgctgcccaaccccggcctaga cgcccggatcccgtccctggctgagctggagaccatcgagcaggaggaggccagctcccggccgaagtgggacaacaaggcgcagta catgctcacctgcctgggcttctgcgtgggcctcggcaacgtgtggcgcttcccctacctgtgtcagagccacggaggaggagccttcatg atcccgttcctcatcctgctggtcctggagggcatccccctgctgtacctggagttcgccatcgggcagcggctgcggcggggcagcctgg gtgtgtggagctccatccacccggccctgaagggcctaggcctggcctccatgctcacgtccttcatggtgggactgtattacaacaccatc atctcctggatcatgtggtacttattcaactccttccaggagcctctgccctggagcgactgcccgctcaacgagaaccagacagggtatgtg gacgagtgcgccaggagctcccctgtggactacttctggtaccgagagacgctcaacatctccacgtccatcagcgactcgggctccatcc agtggtggatgctgctgtgcctggcctgcgcatggagcgtcctgtacatgtgcaccatccgcggcatcgagaccaccgggaaggccgtgt acatcacctccacgctgccctatgtcgtcctgaccatcttcctcatccgaggcctgacgctgaagggcgccaccaatggcatcgtcttcctctt cacgcccaacgtcacggagctggcccagccggacacctggctggacgcgggcgcacaggtcttcttctccttctccctggccttcggggg cctcatctccttctccagctacaactctgtgcacaacaactgcgagaaggactcggtgattgtgtccatcatcaacggcttcacatcggtgtat gtggccatcgtggtctactccgtcattgggttccgcgccacacagcgctacgacgactgcttcagcacgaacatcctgaccctcatcaacgg gttcgacctgcctgaaggcaacgtgacccaggagaactttgtggacatgcagcagcggtgcaacgcctccgaccccgcggcctacgcgc agctggtgttccagacctgcgacatcaacgccttcctctcagaggccgtggagggcacaggcctggccttcatcgtcttcaccgaggccat caccaagatgccgttgtccccactgtggtctgtgctcttcttcattatgctcttctgcctggggctgtcatctatgtttgggaacatggagggcgt cgttgtgcccctgcaggacctcagagtcatccccccgaagtggcccaaggaggtgctcacaggcctcatctgcctggggacattcctcatt ggcttcatcttcacgctgaactccggccagtactggctctccctgctggacagctatgccggctccattcccctgctcatcatcgccttctgcg agatgttctctgtggtctacgtgtacggtgtggacaggttcaataaggacatcgagttcatgatcggccacaagcccaacatcttctggcaagt cacgtggcgcgtggtcagccccctgctcatgctgatcatcttcctcttcttcttcgtggtagaggtcagtcaggagctgacctacagcatctgg gaccctggctacgaggaatttcccaaatcccagaagatctcctacccgaactgggtgtatgtggtggtggtgattgtggctggagtgccctc cctcaccatccctggctatgccatctacaagctcatcaggaaccactgccagaagccaggggaccatcaggggctggtgagcacactgtc cacagcctccatgaacggggacctgaagtactgagaaggcccatcccacggcgtgccatacactggtgtcagggaaggaggaaccagc aagacctgtggggtgggggccgggctgcacctgcatgtgtgtaagcgtgagtgtatgctcgtgtgtgagtgtgtgtattgtacacgcatgtg ccatgtgtgcagatatgtatcgtgtgtgcatgtacatgcatgggcactgtgtgagtgtgcacgtgtatgcacacatatacatgtgtgtgggtgtg tgtattgtatgtgcatgtgccatgtgtgcagatgtgtcatgttgtgtgtgtgcatgtacatgtatggacattgtgtgagtgtgcaagtgtgcatgca tatacatgtgtgcgatatttgctgcccgtgtgtgtgcatgtatatatagacatacatgcctatgttgtgtgtggtgtgcatatgtgtgaacacacac gtgtatacatgcatgcacatgtgctcgtacaatgggtgtccacatgcacgtgtatatgtatatctgtgagtgtatatacatgcatgcaattgtgtgt atgtgtgttctgtgtgtgcgtttgcaagtatatatgcacatgtgtatatgtacatgtatgcctgtgtgacgtgtgtatatgtgagcatgtgtacgtgt gtgtatacgtgtgttgtgtatatgtgtgtgtctgtacctgtttgtgtatatgtgtgtgatgtgtgctcgtgtgtgtgcatattcaggcaggtgtgcattt gtgcatgccagtgtgtatgtatgtgcgcatatggacacgcatggacacgcatatggacacatatggacacacatatggacacgtgtggatat gtgtgcgtacacgtcgctgggacacatgcctgccactcggggcccagctgccctctgtgtttgtccttgccacagtcacggggtgcatgtgc agaggggagcagaccactggggacgtgctgtgccctgcacgtgcccgggggaagcggaagctgcagctggggtgggggcagcacct ctatgcttcatctctgtgggtggcaggagacaaaagcacagggtactatcttggctcctgggagcgactcttgctacccacccccacccatc cccttccccttggtgttgacctttgacctgggggttcccagagccctgtagccctcgacccggagcagcctctcggaagccggagtgggca gttgctggcgattctgagaaaacttggccgcatccaccggggccctgcctccagtcggccgctgccgagtctctgcgttctggccgcttccc ggcttaatgaatgccagccatttaatcattgctcctgccaccacaaatagatgagcagttaaataaaactcaacttggcataattcaaggcaaa taccactctgtgcattttcttaagaggacatgagctgtgtgaatttttagccagcctttggaaaagatgggttacagggtaactcaaccctggct gccatccttgggcactgtgtgtgtccagggcaccttggaggaccgtgcagcccccagaagcttccagctcccgcaccactcagtgaagcc cagcctggcgcctgccctgcccccgtcacgggatgggcccccattggggttcaacattccatcgcagccaaaggcagtcggcacttggga catctgcttccacggacaggtcacctccgctttgcacggaagaatctggatgcttacattaaactggtgttctgagagttcctacggacaggtc acctccgctttgcatggaagaatctggatgcttacattaaactggtgttctgagagttcctacggacaggtcacctctgctttccatagaagaat ctggacgcttacattaaactgatgttctgagaattcctacaggcaggactgaaagcctggtgtgtgccagtatgatgttccacccacagaaac ctggtcacaatcgtcccttccagcaccccatccagcagtgactgcacacactgagtcccctaccagcccctttcaccctgctgactgtcactg ggccctgggatgcgcaagactccacagcagcagaggtggggggacatatcacagcctctgcccccggctgtgatgccaccgaggggct cgcctgctgatggcttcaacagggtctcacctcatcttttcctgctctttggccctggatcgagaaaatttccatcagtgccccattaatatgctg ccctgtggcatctgcccaggaggccctgccaggcgtgcacaggtgtgcattggtgtaccctggcatgcacaggtgtgcactgatgtgccct ggcatccattggtgtaccctggtgtgcctgccataggaccctgggcgggagctcccatctcatctacatctcctgattcatgcgttgtttcatag gtttcaatgtctctgtaaatgtggtagaaatgcaggctttatgggcataaagtgtacatttctaaataaatcccttctattgagtatgctcaccctag aagttactgttgtccagacgtagagggatgagtgagccagtgacctcagacgggatggtggggacggcaggtccagctcctgcctcctcct ggggggtctggctttgggggcttgctccgaagaggccatggcccaggcctgtggcctcacaatggggaccaaccagctcttctcatcttctt ccctcacacttcctctcactcaaataagaaccttccaaaaatgtgtccacctgggcccctgccctgggactcatggatttggagttgtggccac acggttgaggggtgcagtgtccagtggaatggggcaattgcgggcctgggggcccttggcctgtccgtggcgggagcatctgcaaggag gagccccagagtccagggagcactgtggggagctccttagagctgaactcacccggcgtcaactcatcaaccctccacccatggacagg ggtgcccccagcacaggagaggactcagccctctgcccccacgcacggtgggtgcctgtcaccctgtcctgcccagcggcccgagggc agcagtgggtgtgagggcagcccccggcctcccaagagcagctgagaggatccctgcgggaatccgggcttcgggtgcatgcgatctg atctgagttgtttctgacagtgacagagtgacaatctataagtatctcaagatcaaatggttaaataaaacataagaaatttaaaacga (SEQ ID NO: 1)
The reverse complement sequence of the above exemplary mRNA sequence of Homo sapiens solute carrier family 6 member 19 (SLC6A19) is: tcgttttaaatttcttatgtttattaaccattgatcttgagatacttatagattgtcactctgtcactgtcagaaacaactcagatcagatcgcatgc acccgaagcccggattcccgcagggatcctctcagctgctcttgggaggccgggggctgccctcacacccactgctgccctcgggccgct gggcaggacagggtgacaggcacccaccgtgcgtgggggcagagggctgagtcctctcctgtgctgggggcacccctgtccatgggtg gagggtgatgagttgacgccgggtgagttcagctctaaggagctccccacagtgctccctggactctggggctcctccttgcagatgctcc cgccacggacaggccaagggcccccaggcccgcaattgccccattccactggacactgcacccctcaaccgtgtggccacaactccaaa tccatgagtcccagggcaggggcccaggtggacacatttttggaaggttcttatttgagtgagaggaagtgtgagggaagaagatgagaag agctggttggtccccattgtgaggccacaggcctgggccatggcctcttcggagcaagcccccaaagccagaccccccaggaggaggc aggagctggacctgccgtccccaccatcccgtctgaggtcactggctcactcatccctctacgtctggacaacagtaacttctagggtgagc atactcaatagaagggattatttagaaatgtacacttatgcccataaagcctgcattctaccacatttacagagacattgaaacctatgaaac aacgcatgaatcaggagatgtagatgagatgggagctcccgcccagggtcctatggcaggcacaccagggtacaccaatggatgccagg gcacatcagtgcacacctgtgcatgccagggtacaccaatgcacacctgtgcacgcctggcagggcctcctgggcagatgccacagggc agcatattaatggggcactgatggaaattttctcgatccagggccaaagagcaggaaaagatgaggtgagaccctgttgaagccatcagca ggcgagcccctcggtggcatcacagccgggggcagaggctgtgatatgtccccccacctctgctgctgtggagtctgcgcatcccaggg cccagtgacagtcagcagggtgaaaggggctggtaggggactcagtgtgtgcagtcactgctggatggggtgctggaagggacgattgt gaccaggttictgtgggtggaacatcatactggcacacaccaggctttcagtcctgcctgtaggaattctcagaacatcagtttaatgtaagcg tccagattcttctatggaaagcagaggtgacctgtccgtaggaactctcagaacaccagtttaatgtaagcatccagattcttccatgcaaagc ggaggtgacctgtccgtaggaactctcagaacaccagtttaatgtaagcatccagattcttccgtgcaaagcggaggtgacctgtccgtgga agcagatgtcccaagtgccgactgcctttggctgcgatggaatgttgaaccccaatgggggcccatcccgtgacgggggcagggcaggc gccaggctgggcttcactgagtggtgcgggagctggaagcttctgggggctgcacggtcctccaaggtgccctggacacacacagtgcc caaggatggcagccagggtgagttaccctgtaacccatcttttccaaaggctggctaaaaattcacacagctcatgtcctcttaagaaaatgc acagagtggtatttgccttgaattateccaagttgagttttatttaactgctcatctatttgtegtegcageagcaateattaaatgsctgecattc attaagccgggaagcggccagaacgcagagactcggcagcggccgactggaggcagggccccggtggatgcggccaagttttctcag aatcgccagcaactgcccactccggcttccgagaggctgctccgggtcgagggctacagggctctgggaacccccaggtcaaaggtcaa caccaaggggaaggggatgggtgggggtgggtagcaagagtcgctcccaggagccaagatagtaccctgtgctttgtctcctgccaccc acagagatgaagcatagaggtgctgcccccaccccagctgcagctccgctcccccgggcacgtgcagggcacagcacgtccccagtg gtctgctcccctctgcacatgcaccccgtgactgtggcaaggacaaacacagagggcagctgggccccgagtggcaggcatgtgtccca gcgacgtgtacgcacacatatccacacgtgtccatatgtgtgtccatatgtgtccatatgcgtgtccatgcgtgtccatatgcgcacatacatac acactggcatgcacaaatgcacacctgcctgaatatgcacacacacgagcacacatcacacacatatacacaaacaggtacagacacaca catatacacaacacacgtatacacacacgtacacatgctcacatatacacacgtcacacaggcatacatgtacatatacacatgtgcatatata cttgcaaacgcacacacagaacacacatacacacaattgcatgcatgtatatacactcacagatatacatatacacgtgcatgtggacaccca tgtacgagcacatgtgcatgcatgtatacacgtgtgtgttcacacatatgcacaccacacacaacataggcatgtatgtctatatatacatgca cacacacgggcagcaaatatcgcacacatgtatatgcatgcacacttgcacactcacacaatgtccatacatgtacatgcacacacacaaca tgacacatctgcacacatggcacatgcacatacaatacacacacccacacacatgtatatgtgtgcatacacgtgcacactcacacagtgcc catgcatgtacatgcacacacgatacatatctgcacacatggcacatgcgtgtacaatacacacactcacacacgagcatacactcacgctt acacacatgcaggtgcagcccggcccccaccccacaggtcttgctggttcctccttccctgacaccagtgtatggcacgccgtgggatggg ccttctc^tactcaggtccccgttcatggaggctgtggacagtgtgctcaccagcccctgatggtcccctggcttctggcagtggttcctga tgagcttgtagatggcatagccagggatggtgagggagggcactccagccacaatcaccaccaccacatacacccagttcgggtaggag atcttctgggattgggaaattcctcgtagccagggtcccagatgctgtaggtcagctcctgactgacctctaccacgaagaagaagaggaa gatgatcagcatgagcagggggctgaccacgcgccacgtgacttgccagaagatgtgggcttgtggccgatcatgaactcgatgtccttat tgaacctgtccacaccgtacacgtagaccacagagaacatctcgcagaaggcgatgatgagcaggggaatggagccggcatagctgtcc agcagggagagccagtactggccggagttcagcgtgaagatgaagccaatgaggaatgtccccaggcagatgaggcctgtgagcacct ccttgggccacttcggggggatgactctgaggtcctgcaggggcacaacgacgccctccatgttcccaaacatagatgacagccccaggc agaagagcataatgaagaagagcacagaccacagtggggacaacggcatcttggtgatggcctcggtgaagacgatgaaggccaggcc tgtgccctccacggcctctgagaggaaggcgtgatgtcgcaggtctggaacaccagctgcgcgtaggccgcggggtcggaggcgttgc accgctgctgcatgtccacaaagttctcctgggtcacgttgccttcaggcaggtcgaacccgttgatgagggtcaggatgttcgtgctgaag cagtcgtcgiagcgctgtgiggcgcggaacccaatgacggagtagaccacgatggccacatacaccgatgtgaagccgttgatgatggac acaatcaccgagtccttctcgcagtgttgtgcacagagttgtagctggagaaggagatgaggcccccgaaggccagggagaaggagaa gaagacctgtgcgcccgcgtccagccaggtgtccggctgggccagctccgtgacgttgggcgtgaagaggaagacgatgccattggtgg cgcccttcagcgtcaggcctcggatgaggaagatggtcaggacgacatagggcagcgtggaggtgatgtacacggccttcccggtggtct cgatgccgcggatggtgcacatgtacaggacgctccatgcgcaggccaggcacagcagcatccaccactggatggagcccgagtcgct gatggacgtggagatgtgagcgtctctcggtaccagaagtagtccacaggggagctcctggcgcactcgtccacataccctgtctggtct cgttgagcgggcagtcgctccagggcagaggctcctggaaggagttgaataagtaccacatgatccaggagatgatggtgtgtaatacag tcccaccatgaaggacgtgagcatggaggccaggcctaggccctcagggccgggtggatggagctccacacacccaggctgccccgc cgcagccgctgcccgatggcgaactccaggtacagcagggggatgccctccaggaccagcaggatgaggaacgggatcatgaaggct cctcctccgtggctctgacacaggtaggggaagcgccacacgtgccgaggcccacgcagaagcccaggcaggtgagcatgtactgcg ccttgtgtcccacttcggccgggagctggcctcctcctgctcgatggtctccagctcagccagggacgggatccgggcgtctaggccggg gttgggcagcacgagcctcaccatggtggtcgctggacggctccgcacacagcaggcagggcagaagctggagggcgagt (SEQ ID NO: 2)
Thel/omo sapiens SLC6A19 mRNA encodes for the sodium-dependent neutral amino acid transporter B°AT1 ofNP_001003841.1 : MVRLVLPNPGLDARIPSLAELETIEQEEASSRPKWDNKAQYMLTCLGFCVGLGNVWRFP YLCQSHGGGAFMIPFLILLVLEGIPLLYLEFAIGQRLRRGSLGVWSSIHPALKGLGLASML TSFMVGLYYNTIISWIMWYLFNSFQEPLPWSDCPLNENQTGYVDECARSSPVDYFWYRE TLNISTSISDSGSIQWWMLLCLACAWSVLYMCTIRGIETTGKAVYITSTLPYVVLTIFLIRG LTLKGATNGIVFLFTPNVTEL AQPDTWLD AGAQ VFF SF SL AFGGLISF S S YNS VHNNCEK DSVIVSIINGFTSVYVAIVVYSVIGFRATQRYDDCFSTNILTLINGFDLPEGNVTQENFVD MQQRCNASDPAAYAQLVFQTCDINAFLSEAVEGTGLAFIVFTEAITKMPLSPLWSVLFFI MLFCLGLSSMFGNMEGVVVPLQDLRVIPPKWPKEVLTGLICLGTFLIGFIFTLNSGQYWL SLLDSYAGSIPLLIIAFCEMFSVVYVYGVDRFNKDIEFMIGHKPNIFWQVTWRVVSPLLM LIIFLFFFVVEVSQELTYSIWDPGYEEFPKSQKISYPNWVYVVVVIVAGVPSLTIPGYAIYK LIRNHCQKPGDHQGLVSTLSTASMNGDLKY (SEQ ID NO: 3)
It is known in the art that alternative RNA transcripts can be produced from the same genomic region of DNA. These alternative transcripts are generally known as “variants.” More specifically, “pre-mRNA variants” are transcripts produced from the same genomic DNA that differ from other transcripts produced from the same genomic DNA in either their start or stop position and contain both intronic and exonic sequence. Variants can result in mRNA variants including, but not limited to, those with alternate splice junctions, or alternate initiation and termination codons. Variants in genomic and mRNA sequences can result in disease. For the human SLC6A19 target mRNA, predicted transcript variant XI can be found, e.g., at accession number XM54352501.1 : actcgccctccagcttctgccctgcctgctgtgtgcggagccgtccagcgaccaccatggtgaggctcgtgctgcccaaccccggcctaga cgcccggatcccgtccctggctgagctggagaccatcgagcaggaggaggccagctcccggccgaagtgggacaacaaggcgcagta catgctcacctgcctgggcttctgcgtgggcctcggcaacgtgtggcgcttcccctacctgtgtcagagccacggaggaggagccttcatg atcccgttcctcatcctgctggtcctggagggcatccccctgctgtacctggagttcgccatcgggcagcggctgcggcggggcagcctgg gcgtgtggagctccatccacccggccctgaagggcctaggcctggcctccatgctcacgtccttcatggtgggactgtattacaacaccatc atctcctggatcatgtggtacttattcaactccttccaggagcctctgccctggagcgactgcccgctcaacgagaaccagacagggtatgtg gacgagtgcgccaggagctcccctgtggactacttctggtaccgagagacgctcaacatctccacgtccatcagcgactcgggctccatcc agtggtggatgctgctgtgcctggcctgcgcatggagcgtcctgtacatgtgcaccatccgcggcatcgagaccaccgggaaggccgtgt acatcacctccacgctgccctatgtcgtcctgaccatcttcctcatccgaggcctgacgctgaagggcgccaccaatggcatcgtcttcctctt cacgcccaacgtcacggagctggcccagccggacacctggctggacgcgggcgcacaggtcttcttctccttctccctggccttcggggg cctcatctccttctccagctacaactctgtgcacaacaactgcgagaaggactcggtgattgtgtccatcatcaacggcttcacatcggtgtat gtggccatcgtggtctactccgtcattgggttccgcgccacgcagcgctacgacgactgcttcagcacgaacatcctgaccctcatcaacgg gttcgacctgcctgaaggcaacgtgacccaggagaactttgtggacatgcagcagcggtgcaacgcctccgaccccgcggcctacgcgc agctggtgttccagacctgcgacatcaacgccttcctctcagaggccgtggagggcacaggcctggccttcatcgtcttcaccgaggccat caccaagatgccgttgtccccactgtggtctgtgctcttcttcattatgctcttctgcctggggctgtcatctatgtttgggaacatggagggcgt cgttgtgcccctgcaggacctcagagtcatccccccgaagtggcccaaggaggtgctcacaggcctcatctgcctggggacattcctcatt ggcttcatcttcacgctgaactccggccagtactggctctccctgctggacagctatgccggctccattcccctgctcatcatcgccttctgcg agatgttctctgtggtctacgtgtacggtgtggacaggttcaataaggacatcgagttcatgatcggccacaagcccaacatcttctggcaagt cacgtggcgcgtggtcagccccctgctcatgctgatcatcttcctcttcttcttcgtggtagaggtcagtcaggagctgacctacagcatctgg gaccctggctacgaggaatttcccaaatcccagaagatctcctacccgaactgggtgtatgtggtggtggtgattgtggctggagtgccctc cctcaccatccctggctatgccatctacaagctcatcaggaaccactgccagaagccaggggaccatcaggggctggtgagcacactgtc cacagcctccatgaacggggacctgaagtactgagaaggcccatcccacggcgtgccatacactggtgtcagggaaggaggaaccagc aagacctgtggggtgggggccgggctgcacctgcatgtgtgtaagcgtgagtgtatgctcgtgtgtgagtgtgtgtattgtacacgcatgtg ccatgtgtgcagatatgtatcgtgtgtgcatgtacatgcatgggcactgtgtgagtgtgcacgtgtatgcacacatatacatgtgtgtgggtgtg tgtattgtatgtgcatgtgccatgtgtgcagatgtgtcatgttgtgtgtgtgcatgtacatgtatggacattgtgtgagtgtgcaagtgtgcatgca tatacatgtgtgcgatatttgctgcccgtgtgtgtgcatgtatatatagacatacatgcctatgttgtgtgtggtgtgcatatgtgtgaacacacac gtgtatacatgcatgcacatgtgctcgtacaatgggtgtccacatgcacgtgtatatgtatatctgtgagtgtatatacatgcatgcaattgtgtgt atgtgtgttctgtgtgtgcgtttgcaagtatatatgcacatgtgtatatgtacatgtatgcctgtgtgacgtgtgtatatgtgagcatgtgtacgtgt gtgtatacgtgtgttgtgtatatgtgtgtgtctgtacctgtttgtgtatatgtgtgtgatgtgtgctcgtgtgtgtgcatattcaggcaggtgtgcattt gtgcatgccagtgtgtatgtatgtgcgcatatggacacgcatggacacgcatatggacacatatggacacacatatggacacgtgtggatat gtgtgcgtacacgtcgctgggacacatgcctgccactcggggcccagctgccctctgtgtttgtccttgccacagtcacggggtgcatgtgc agaggggagcagaccactggggacgtgctgtgccctgcacgtgcccgggggaagcggaagctgcagctggggtgggggcagcacct ctatgcttcatctctgtgggtggcaggagacaaaagcacaggttactatcttggctcctgggagcgactcttgctacccacccccacccatcc ccttccccttggtgttgacctttgacctgggggttcccagagccctgtagccctcgacccggagcagcctctcggaagccggagtgggcag ttgctggcgattctgagaaaacttggccgcatccaccggggccctgcctccagtcggccgctgccgagtctctgcgttctggccgcttcccg gcttaatgaatgccagccatttaatcattgctcctgccaccacaaatagatgagcagttaaataaaactcaacttggcataattcaaggcaaata ccactctgtgcattttcttaagaggacatgagctgtgtgaatttttagccagcctttggaaaagatgggttacagggtaactcaaccctggctgc catccttgggcactgtgtgtgtccagggcaccttggaggaccgtgcagcccccagaagcttccagctcccgcaccactcagtgaagccca gcctggcgcctgccctgcccccgtcacgggatgggcccccattggggttcaacattccatcgcagccaaaggcagtcggcacttgggaca tctgcttccacggacaggtcacctccgctttgcacggaagaatctggatgcttacattaaactggtgttctgagagttcctacggacaggtcac ctccgctttgcacggaagaatctggatgcttacattaaactggtgttctgagagttcctacggacaggtcacctccgctttgcatggaagaatct ggatgcttacattaaactggtgttctgagagttcctacggacaggtcacctctgctttccatagaagaatctggacgcttacattaaactgatgtt ctgagaattcctacaggcaggactgaaagcctggtgtgtgccagtatgatgttccacccacagaaacctggtcacaatcgtcccttccagca ccccatccagcagtgactgcacacactgagccccctaccagcccctttcaccctgctgactgtcactgggccctgggatgtgcaagactcc acagcagcagaggtggggggaaatatcacagcctctgcccccggctgtgatgccaccgaggggctcgcctgctgatggcttcaacaggg tctcacctcatcttttcctgctccttggccctggatcgagaaaatttccatcagtgccccataatatgctgccctgtggcatctgcccaggagg ccctgccaggcgtgcacaggtgtgcattggtgtaccctgacatgcacaggtgtgcactgatgtgccctggcatccattggtgtaccctggtgt gcctgccataggaccctgggcgggagctcccatctcatctacatctcctgattcatgcgttgtttcgtaggtttcaatgtctctgtaaatgtggta gaaatgcaggctttatgggcataaagtgtacatttctaaataaatcccttctatttagtatgctca (SEQ ID NO: 4) Modulatory Nucleic Acids Conjugated to Ligands
Another modification of the RNA of an ASO or iRNA agent of the present disclosure involves chemically linking to the ASO or iRNA agent one or more ligands, moieties or conjugates that enhance the activity, cellular distribution, or cellular uptake of the ASO or iRNA agent e.g., into a cell. Such moieties include but are not limited to lipid moieties such as a cholesterol moiety (Letsinger et al.. Proc. Natl. Acid. Sci. USA, 1989, 86: 6553-6556). In other embodiments, the ligand is cholic acid (Manoharan et al., Biorg. Med. Chem. Let., 1994, 4:1053-1060), a thioether, e.g., beryl-S-trity1thiol (Manoharan et al., Ann. N.Y. Acad, Sci., 1992, 660:306-309; Manoharan et al., Biorg. Med. Chem. Let., 1993, 3:2765-2770), a thiocholesterol (Oberhauser et al., Nucl. Acids Res., 1992, 20:533-538), an aliphatic chain, e.g., dodecandiol or undecyl residues (Saison- Behmoaras et al., EMBO J, 1991, 10:1111-1118; Kabanov et al., FEBS Lett., 1990, 259:327-330; Svinarchuk et al., Biochimie, 1993, 75:49-54), a phospholipid, e.g., di-hexadecyl-rac-glycerol or triethyl -ammonium 1, 2-di-O-hexadecyl-rac-glycero-3 -phosphonate (Manoharan et al., Tetrahedron Lett., 1995, 36:3651-3654; Shea et al., Nucl. Acids Res., 1990, 18:3777-3783), a polyamine or a polyethylene glycol chain (Manoharan et al., Nucleosides & Nucleotides, 1995, 14:969-973), or adamantane acetic acid (Manoharan et al., Tetrahedron Lett., 1995, 36:3651- 3654), a palmityl moiety (Mishra et al , Biochim. Biophys. Acta, 1995, 1264:229-237), or an octadecylamine or hexylamino-carbonyloxy cholesterol moiety (Crooke et al., J. Pharmacol. Exp. Then, 1996, 277:923-937).
In certain embodiments, a ligand alters the distribution, targeting, or lifetime of an ASO or iRNA agent into which it is incorporated. In certain embodiments a ligand provides an enhanced affinity for a selected target, e.g., molecule, cell or cell type, compartment, e.g., a cellular or organ compartment, tissue, organ or region of the body, as, e.g., compared to a species absent such a ligand. Certain ligands do not take part in duplex pairing in a duplexed nucleic acid.
Ligands can include a naturally occurring substance, such as a protein (e.g., human serum albumin (HSA), low-density lipoprotein (LDL), or globulin); carbohydrate (e.g., a dextran, pullulan, chitin, chitosan, inulin, cyclodextrin, N-acetylglucosamine, N-acetylgalactosamine, or hyaluronic acid); or a lipid. The ligand can also be a recombinant or synthetic molecule, such as a synthetic polymer, e.g., a synthetic polyamino acid. Examples of polyamino acids include polyamino acid is a polylysine (PLL), poly L-aspartic acid, poly L-glutamic acid, styrene-maleic acid anhydride copolymer, poly (L-lactide-co-gly colied) copolymer, divinyl ether-maleic anhydride copolymer, N-(2-hydroxypropy1)methacry1amide copolymer (HMPA), polyethylene glycol (PEG), polyvinyl alcohol (PVA), polyurethane, poly(2-ethylacryllic acid), N- isopropylacrylamide polymers, or polyphosphazine. Examples of polyamines include: polyethylenimine, polylysine (PLL), spermine, spermidine, polyamine, pseudopeptide-polyamine, peptidomimetic polyamine, dendrimer polyamine, arginine, amidine, protamine, cationic lipid, cationic porphyrin, quaternary salt of a polyamine, or an alpha helical peptide.
Ligands can also include targeting groups, e.g., a cell or tissue targeting agent, e.g., a lectin, glycoprotein, lipid or protein, e.g , an antibody, that binds to a specified ceil type such as a kidney cell (kidney cell-targeting nioieties are considered in additional detail below). A targeting group can be a thyrotropin, melanotropin, lectin, glycoprotein, surfactant protein A, Mucin carbohydrate, multivalent lactose, multivalent galactose, N-acetyl-galactosamine, N-acetyl-glucosamine multivalent mannose, multivalent fucose, glycosylated polyaminoacids, multivalent galactose, transferrin, bisphosphonate, poly glutamate, polyaspartate, a lipid, cholesterol, a steroid, bile acid, folate, vitamin Bl 2, vitamin A, biotin, or an RGD peptide or RGD peptide mimetic. In certain embodiments, the ligand is a multivalent galactose, e.g., an N-acetyl-galactosamine.
Other examples of ligands include dyes, intercalating agents (e.g. acridines), cross-linkers (e.g psoralene, mitomycin C), porphyrins (TPPC4, texaphyrin, Sapphyrin), polycyclic aromatic hydrocarbons (e.g., phenazine, dihydrophenazine), artificial endonucleases (e.g. EDTA), lipophilic molecules, e.g., cholesterol, cholic acid, adamantane acetic acid, 1 -pyrene butyric acid, dihydrotestosterone, 1,3-Bis-O(hexadecyl)glycerol, geranyloxyhexyl group, hexadecylglycerol, borneol, menthol, 1,3 -propanediol, heptadecyl group, palmitic acid, myristic acid, 03- (oleoyl)lithocholic acid, O3-(oleoyl)cholenic acid, dimethoxytrityl, or phenoxazine)and peptide conjugates (e.g , antennapedia peptide, Tat peptide), alkylating agents, phosphate, amino, mercapto, PEG (e.g., PEG-40K), MPEG, [MPEG]2, polyamino, alkyl, substituted alkyl, radiolabeled markers, enzymes, haptens (e.g. biotin), Iran sport/ab sorption facilitators (e.g., aspirin, vitamin E, folic acid), synthetic ribonucleases (e.g , imidazole, bisimidazole, histamine, imidazole clusters, acridine-imidazole conjugates, Eu3+ complexes of tetraazamacrocycles), dinitrophenyl, HRP, or AP.
Ligands can be proteins, e.g., glycoproteins, or peptides, e.g., molecules having a specific affinity for a co-ligand, or antibodies e g., an antibody, that binds to a specified cell type such as a hepatic cell. Ligands can also include hormones and hormone receptors. They can also include non-peptidic species, such as lipids, lectins, carbohydrates, vitamins, cofactors, multivalent lactose, multivalent galactose, N-acetyl-galactosamine, N-acetyl-glucosamine multivalent mannose, or multivalent fucose. The ligand can be, for example, a lipopolysaccharide, an activator of p38 MAP kinase, or an activator of NF-KB.
The ligand can be a substance, e.g., a drug, which can increase the uptake of the ASO or iRNA agent into the cell, for example, by disrupting the cell’s cytoskeleton, e.g., by disrupting the cell’s microtubules, microfilaments, or intermediate filaments The drug can be, for example, taxol, vincristine, vinblastine, cytochalasin, nocodazole, japl akin ol ide, latrunculin A, phalloidin, swinholide A, indanocine, or myoservin.
In some embodiments, a ligand attached to an ASO or iRNA agent as described herein acts as a pharmacokinetic modulator (PK modulator). PK modulators include lipophiles, bile acids, steroids, phospholipid analogues, peptides, protein binding agents, PEG, vitamins, etc. Exemplary PK modulators include, but are not limited to, cholesterol, fatty acids, cholic acid, lithocholic acid, dialkylglycerides, diacyl glyceride, phospholipids, sphingolipids, naproxen, ibuprofen, vitamin E, biotin. Oligonucleotides that comprise a number of phosphorothioate linkages are also known to bind to serum protein, thus short oligonucleotides, e.g., oligonucleotides of about 5 bases, 10 bases, 15 bases, or 20 bases, comprising multiple of phosphorothioate linkages in the backbone are also amenable to the instant disclosure as ligands (e.g., as PK modulating ligands). In addition, aptamers that bind serum components (e.g., serum proteins) are also suitable for use as PK modulating ligands in the embodiments described herein
Ligand-conjugated ASOs or iRNA agents of the present disclosure may be synthesized by the use of an oligonucleotide that bears a pendant reactive functionality, such as that derived from the attachment of a linking molecule onto the oligonucleotide. This reactive oligonucleotide may be reacted directly with commercially-available ligands, ligands that are synthesized bearing any of a variety of protecting groups, or ligands that have a linking moiety attached thereto.
The oligonucleotides used in the conjugates of the instant disclosure may be conveniently and routinely made through the well-known technique of solid-phase synthesis. Equipment for such synthesis is sold by several vendors including, for example, Applied Biosystems® (Foster City, Calif.). Any other methods for such synthesis known in the art may additionally or alternatively be employed. It is also known to use similar techniques to prepare other oligonucleotides, such as the phosphorothioates and alkylated derivatives. In the ligand-conjugated ASOs or iRNA agents and ligand-molecule bearing sequencespecific linked nucleosides of the instant disclosure, the oligonucleotides and oligonucleosides may be assembled on a suitable DNA synthesizer utilizing standard nucleotide or nucleoside precursors, or nucleotide or nucleoside conjugate precursors that already bear the linking moiety, ligand-nucleotide or nucleoside-conjugate precursors that already bear the ligand molecule, or non-nucleoside ligand-bearing building blocks.
When using nucleotide-conjugate precursors that already bear a linking moiety, the synthesis of the sequence-specific linked nucleosides is typically completed, and the ligand molecule is then reacted with the linking moiety to form the ligand-conjugated oligonucleotide. In some embodiments, the oligonucleotides or linked nucleosides of the instant disclosure are synthesized by an automated synthesizer using phosphoramidites derived from ligand-nucleoside conjugates in addition to the standard phosphoramidites and non-standard phosphoramidites that are commercially available and routinely used in oligonucleotide synthesis.
Kidney Cell-Targeting Mote ties and Conjugates
In certain embodiments, a targeting moiety is employed that binds specifically to a factor present on the surface of target cell(s) of interest - e.g., kidney-associated cells. In some embodiments, provided technologies achieve targeted delivery of payload moieties to a target cell, tissue, organ or organism of interest, for example with minimal off-target effects. In some embodiments, a targeting moiety as described herein binds specifically to a factor that is preferentially present on the surface of target cell(s) or tissue(s) of interest - e.g., relative to one or more non-target cell(s) or tissue(s). In some embodiments, a targeting moiety as described herein binds specifically to a factor that is specific to target cell(s) or tissue(s) of interest.
In certain embodiments, targeting moieties associated with (e.g., conjugated with or otherwise linked to) modulatory nucleic acid agents of the instant disclosure target Megalin and/or Cubilin (kidney cell surface factor receptors), which have been previously described (see PCT/US23/16319) as particularly useful strategies for delivering nucleic acid agents into cells, especially into kidney-associated cells (e.g., kidney cells). Conjugate agents that include a Megalin-binding moiety conjugated (optionally by way of a linker) with a nucleic acid agent are particularly useful for delivering such nucleic acid agent into Megalin-expressing cells. Such conjugate agents are particularly useful for delivering nucleic acid agents to kidney cells. In other embodiments, targeting agents associated with modulatory nucleic acid agents of the disclosure can include a polypeptide, an aminoglycoside, an endogenous ligand (e.g., a ligand disclosed in Table 1 or Table 2), a xenobiotic, an antibody or a fragment thereof, an aptamer, a small molecule, or any combination thereof.
A targeting moiety for use as disclosed herein can bind to, e.g., selectively bind to, a surface factor (e.g., to a moiety or portion thereof, and/or to a particular form, such as a disease-associated form thereof) present on surfaces of target cell(s) of interest (e.g., of kidney cells) as disclosed herein.
Without wishing to be bound by theory, certain embodiments of the present disclosure provide for binding of a targeting moiety associated with a modulatory nucleic acid agent of the disclosure to a cell surface factor present on the surface of a relevant (e.g., kidney) cell, e.g., of a tissue, to achieve internalization of the cell surface factor, along with the bound targeting moiety (as part of a conjugate agent that includes the modulatory nucleic acid as described herein). In some embodiments, such internalization may mean that the relevant cell surface factor is no longer (at least for a period of time) available at the surface of the cell, e.g., of a tissue, for, e.g., signaling and/or binding to a ligand.
In some embodiments, a kidney cell surface factor is present on, e.g., can be detected on, a surface of a tissue associated with a kidney, e.g., a tissue that is part of or can be found in a kidney, e.g., during development, during tissue homeostasis, and/or in the course of a disease or disorder. In some embodiments, a kidney cell surface factor is present on, e.g., can be detected on, a proximal tubule epithelial cell and/or a podocyte. In some embodiments, an internalized conjugate agent (e.g., a portion thereof, e.g., a payload moiety), is delivered to a vesicle in a cell (e.g., a lysosome, an endosome, a clathrin coated pit, or an intracellular membranous organelle, or any combination thereof). In some embodiments, an internalized conjugate agent (e.g., a portion thereof, e.g., a payload moiety), is delivered to a compartment in a cell, e.g., a cytoplasm, a mitochondria, a ribosome, a nucleus, a nucleolus, or any other compartment in a cell, or any combination thereof. In some embodiments, an internalized conjugate agent (e.g., a portion thereof, e.g., a modulatory nucleic acid agent associated with a targeting moiety), in a cell (e.g., in a vesicle or a compartment in a cell) can reduce the expression and/or activity of a target of the modulatory nucleic acid agent. In some embodiments, internalization of a conjugate agent (e g., a portion thereof, e.g., a modulatory nucleic acid agent associated with a targeting moiety) into a cell (e.g., into a vesicle or a compartment in a cell) uncouples, e.g., separates, a targeting moiety from a modulatory nucleic acid agent. In some embodiments, a targeting moiety is uncoupled, e.g., separated, from a modulatory nucleic acid agent by a chemical reaction and/or mechanical separation. In some embodiments, a chemical reaction comprises an enzymatic reaction to cleave a linker linking a targeting moiety to a modulatory nucleic acid agent.
In some embodiments, internalization of a conjugate agent (e.g., a portion thereof, e.g., a modulatory nucleic acid agent associated with a targeting moiety) into a cell (e.g., into a vesicle or a compartment in a cell) uncouples a targeting moiety from a modulatory nucleic acid agent associated with the targeting moiety.
In some embodiments, a conjugate agent disclosed herein can be fdtered by a glomerular capillary, e.g., into a Bowman’s capsule. In some embodiments, a conjugate agent disclosed herein has a size, charge, conformation, and/or other properties that allows it to be filtered by a glomerular capillary. In some embodiments, a threshold for glomerular filtration is in the range of 30-50 kDa.
In some embodiments, a cell surface factor (e.g., a kidney cell surface factor) is or comprises a receptor chosen from Megalin, Cubilin, or both.
Megalin-Binding Moieties
Megalin is a receptor of about 600kDa (about 4655 amino acids) and belongs to the low- density lipoprotein receptor family (as disclosed in Nielsen R. et al. (2016), Kidney Int. 89( 1): 58- 67). Megalin is also known as LDL Receptor Related Protein 2 (LRP2), Glycoprotein 330 (Gp33O), Calcium Sensor Protein, or Heymann Nephritis Antigen Homolog.
Human Megalin protein sequence:
MDRGPAAVACTLLLALVACLAPASGQECDSAHFRCGSGHCIPADWRCDGTKDCSDDA DEIGCAVVTCQQGYFKCQSEGQCIPNSWVCDQDQDCDDGSDERQDCSQSTCSSHQITCS NGQCIPSEYRCDHVRDCPDGADENDCQYPTCEQLTCDNGACYNTSQKCDWKVDCRDS SDEINCTEICLHNEFSCGNGECIPRAYVCDHDNDCQDGSDEHACNYPTCGGYQFTCPSG RCIYQNWVCDGEDDCKDNGDEDGCESGPHDVHKCSPREWSCPESGRCISIYKVCDGILD CPGREDENNTSTGKYCSMTLCSALNCQYQCHETPYGGACFCPPGYIINHNDSRTCVEFD DCQIWGICDQKCESRPGRHLCHCEEGYILERGQYCKANDSFGEASIIFSNGRDLLIGDIHG RSFRILVESQNRGVAVGVAFHYHLQRVFWTDTVQNKVFSVDINGLNIQEVLNVSVETPE NLAVDWVNNKIYLVETKVNRIDMVNLDGSYRVTLITENLGHPRGIAVDPTVGYLFFSD WESLSGEPKLERAFMDGSNRKDLVKTKLGWPAGVTLDMISKRVYWVDSRFDYIETVTY DGIQRKTVVHGGSLIPHPFGVSLFEGQVFFTDWTKMAVLKANKFTETNPQVYYQASLRP YGVTVYHSLRQPYATNPCKDNNGGCEQVCVLSHRTDNDGLGFRCKCTFGFQLDTDER HCIAVQNFLIFSSQVAIRGIPFTLSTQEDVMVPVSGNPSFFVGIDFDAQDSTIFFSDMSKH MIFKQKIDGTGREILAANRVENVESLAFDWISKNLYWTDSHYKSISVMRLADKTRRTVV QYLNNPRSVVVHPFAGYLFFTDWFRPAKIMRAWSDGSHLLPVINTTLGWPNGLAIDWA ASRLYWVDAYFDKIEHSTFDGLDRRRLGHIEQMTHPFGLAIFGEHLFFTDWRLGAIIRVR KADGGEMTVIRSGIAYILHLKSYDVNIQTGSNACNQPTHPNGDCSHFCFPVPNFQRVCG CPYGMRLASNHLTCEGDPTNEPPTEQCGLFSFPCKNGRCVPNYYLCDGVDDCHDNSDE QLCGTLNNTC S S S AFTCGHGECIP AHWRCDKRNDC VDGSDEHNCPTHAP ASCLDTQ YT CDNHQCISKNWVCDTDNDCGDGSDEKNCNSTETCQPSQFNCPNHRCIDLSFVCDGDKD CVDGSDEVGCVLNCTASQFKCASGDKCIGVTNRCDGVFDCSDNSDEAGCPTRPPGMCH SDEFQCQEDGICIPNFWECDGHPDCLYGSDEHNACVPKTCPSSYFHCDNGNCIHRAWLC DRDNDCGDMSDEKDCPTQPFRCPSWQWQCLGHNICVNLSVVCDGIFDCPNGTDESPLC NGNSCSDFNGGCTHECVQEPFGAKCLCPLGFLLANDSKTCEDIDECDILGSCSQHCYNM RGSFRCSCDTGYMLESDGRTCKVTASESLLLLVASQNKIIADSVTSQVHNIYSLVENGSY IVAVDFDSISGRIFWSDATQGKTWSAFQNGTDRRVVFDSSIILTETIAIDWVGRNLYWTD YALETIEVSKIDGSHRTVLISKNLTNPRGLALDPRMNEHLLFWSDWGHHPRIERASMDG SMRTVIVQDKIFWPCGLTIDYPNRLLYFMDSYLDYMDFCDYNGHHRRQVIASDLIIRHP YALTLFEDSVYWTDRATRRVMRANKWHGGNQSVVMYNIQWPLGIVAVHPSKQPNSV NPCAF SRC SHLCLLS SQGPHF YSCVCPSGWSLSPDLLNCLRDDQPFLITVRQHIIFGISLNP EVKSNDAMVPIAGIQNGLDVEFDDAEQYIYWVENPGEIHRVKTDGTNRTVFASISMVGP
SMNLALDWISRNLYSTNPRTQSIEVLTLHGDIRYRKTLIANDGTALGVGFPIGITVDPARG KLYWSDQGTDSGVPAKIASANMDGTSVKTLFTGNLEHLECVTLDIEEQKLYWAVTGRG VIERGNVDGTDRMILVHQLSHPWGIAVHDSFLYYTDEQYEVIERVDKATGANKIVLRDN VPNLRGLQVYHRRNAAESSNGCSNNMNACQQICLPVPGGLFSCACATGFKLNPDNRSC SP YNSFIVVSMLS AIRGFSLELSDHSETMVP VAGQGRNALHVDVDVS SGFIYWCDF S S SV ASDNAIRRIKPDGSSLMNIVTHGIGENGVRGIAVDWVAGNLYFTNAFVSETLIEVLRINTT YRRVLLKVTVDMPRHIVVDPKNRYLFWADYGQRPKIERSFLDCTNRTVLVSEGIVTPRG LAVDRSDGYVYWVDDSLDIIARIRINGENSEVIRYGSRYPTPYGITVFENSIIWVDRNLKK IFQASKEPENTEPPTVIRDNINWLRDVTIFDKQVQPRSPAEVNNNPCLENNGGCSHLCFA LPGLHTPKCDCAFGTLQSDGKNCAISTENFLIFALSNSLRSLHLDPENHSPPFQTINVERT VMSLDYDSVSDRIYFTQNLASGVGQISYATLSSGIHTPTVIASGIGTADGIAFDWITRRIY YSDYLNQMINSMAEDGSNRTVIARVPKPRAIVLDPCQGYLYWADWDTHAKIERATLGG NFRVPIVNSSLVMPSGLTLDYEEDLLYWVDASLQRIERSTLTGVDREVIVNAAVHAFGL TLYGQYIYWTDLYTQRIYRANKYDGSGQIAMTTNLLSQPRGINTVVKNQKQQCNNPCE QFNGGCSHICAPGPNGAECQCPHEGNWYLANNRKHCIVDNGERCGASSFTCSNGRCISE EWKCDNDNDCGDGSDEMESVCALHTCSPTAFTCANGRCVQYSYRCDYYNDCGDGSDE AGCLFRDCNATTEFMCNNRRCIPREFICNGVDNCHDNNTSDEKNCPDRTCQSGYTKCH NSNICIPRVYLCDGDNDCGDNSDENPTYCTTHTCSSSEFQCASGRCIPQHWYCDQETDCF DASDEPASCGHSERTCLADEFKCDGGRCIPSEWICDGDNDCGDMSDEDKRHQCQNQNC SDSEFLCVNDRPPDRRCIPQSWVCDGDVDCTDGYDENQNCTRRTCSENEFTCGYGLCIP KIFRCDRHNDCGDYSDERGCLYQTCQQNQFTCQNGRCISKTFVCDEDNDCGDGSDELM HLCHTPEPTCPPHEFKCDNGRCIEMMKLCNHLDDCLDNSDEKGCGINECHDPSISGCDH
NCTDTLTSFYC SCRPGYKLMSDKRTC VDIDECTEMPF VC SQKCENVIGSYICKC APGYLR EPDGKTCRQNSNIEPYLIFSNRYYLRNLTIDGYFYSLILEGLDNVVALDFDRVEKRLYWI DTQRQVIERMFLNKTNKETIINHRLPAAESLAVDWVSRKLYWLDARLDGLFVSDLNGG HRRMLAQHCVDANNTFCFDNPRGLALHPQYGYLYWADWGHRAYIGRVGMDGTNKSV IISTKLEWPNGITIDYTNDLLYWADAHLGYIEYSDLEGHHRHTVYDGALPHPFAITIFEDT IYWTDWNTRTVEKGNKYDGSNRQTLVNTTHRPFDIHVYHPYRQPIVSNPCGTNNGGCS HLCLIKPGGKGFTCECPDDFRTLQLSGSTYCMPMCSSTQFLCANNEKCIPIWWKCDGQK DCSDGSDELALCPQRFCRLGQFQCSDGNCTSPQTLCNAHQNCPDGSDEDRLLCENHHC DSNEWQCANKRCIPESWQCDTFNDCEDNSDEDSSHCASRTCRPGQFRCANGRCIPQAW KCDVDNDCGDHSDEPIEECMSSAHLCDNFTEFSCKTNYRCIPKWAVCNGVDDCRDNSD EQGCEERTCHPVGDFRCKNHHCIPLRWQCDGQNDCGDNSDEENCAPRECTESEFRCVN QQCIPSRWICDHYNDCGDNSDERDCEMRTCHPEYFQCTSGHCVHSELKCDGSADCLDA SDEADCPTRFPDGAYCQATMFECKNHVCIPPYWKCDGDDDCGDGSDEELHLCLDVPCN SPNRFRCDNNRCIYSHEVCNGVDDCGDGTDETEEHCRKPTPKPCTEYEYKCGNGHCIPH DNVCDDADDCGDWSDELGCNKGKERTCAENICEQNCTQLNEGGFICSCTAGFETNVFD RTSCLDINECEQFGTCPQHCRNTKGSYECVCADGFTSMSDRPGKRCAAEGSSPLLLLPD NVRIRKYNLS SERF SEYLQDEEYIQ AVDYDWDPKDIGLS VVYYT VRGEGSRFGAIKRAYI PNFESGRNNLVQEVDLKLKYVMQPDGIAVDWVGRHIYWSDVKNKRIEVAKLDGRYRK WLISTDLDQPAAIAVNPKLGLMFWTDWGKEPKIESAWMNGEDRNILVFEDLGWPTGLSI DYLNNDRIYWSDFKEDVIETIKYDGTDRRVIAKEAMNPYSLDIFEDQLYWISKEKGEVW KQNKFGQGKKEKTLVVNPWLTQVRIFHQLRYNKSVPNLCKQICSHLCLLRPGGYSCAC PQGSSFIEGSTTECDAAIELPINLPPPCRCMHGGNCYFDETDLPKCKCPSGYTGKYCEMA F SKGISPGTTA VA VLLTILLIV VIGAL AIAGFFHYRRTGSLLP ALPKLP SL S SL VKP SENGN GVTFRSGADLNMDIGVSGFGPETAIDRSMAMSEDFVMEMGKQPIIFENPMYSARDSAVK VVQPIQVTVSENVDNKNYGSPINPSEIVPETNPTSPAADGTQVTKWNLFKRKSKQTTNFE NPIYAQMENEQKESVAATPPPSPSLPAKPKPPSRRDPTPTYSATEDTFKDTANLVKEDSE V (SEQ ID NO: 5)
The extracellular domain of Megalin includes clusters of cysteine-rich complement-type repeats. The repeats are separated by beta-propeller domains comprising YWTD motifs and EGF- type repeats. Megalin has one transmembrane domain which positions it in parts of the cell membrane that includes cholesterol and/or glycosphingolipids. Megalin also has an intracellular C-terminal cytoplasmic domain which can regulate receptor trafficking and/or endocytosis. The cytoplasmic domain of Megalin comprises NPXY motifs and several other domains such as proline-rich sequences and PDZ motifs. Megalin’ s cytoplasmic domain has been linked to receptor internalization. A typical structure of Megalin is disclosed in Figure 1 of Marzolo and Farfan (2011), Biol Res 44: 89-105, the entire contents of which are hereby incorporated by reference. The extracellular domain of Megalin may also include one or more post-translational modifications, such as glycosylation.
Megalin has been described to interact, at least in certain cases, with a co-receptor, Cubilin. Megalin has been identified on surfaces of one or more of the following tissues and/or cells: immune cells (e.g., bone marrow cells, lymph node cells, thymic cells, peripheral blood mononuclear cells [e.g., myeloid and/or lymphoid cells], erythrocytes, eosinophils, neutrophils, and/or platelets); nervous system (e.g., brain tissue, cortex, cerebellum, retinal cells, spinal cord cells, nerve cells, neurons, and/or supporting cells; endothelial cells; muscle (e.g., heart muscle, smooth muscle, and/or skeletal muscle); small intestine; colon; adipocytes; kidney; liver; lung; spleen; stomach; esophagus; bladder; pancreas; thyroid; salivary gland; adrenal gland; pituitary gland; breast; skin; ovary; uterus; placenta; prostate; and testis. . In kidney tissue, Megalin has been reported to be found on the surface of proximal tubular epithelial cells and podocytes. In proximal tubule epithelia cells of the kidney, Megalin expression has been observed in the brush border, in endocytic vesicles, dense apical tubules and/or lysosomes. Several ligands of Megalin have been identified, some of which are disclosed in Nielsen et al. 2016.
Table 1: Exemplary Megalin ligands
Figure imgf000094_0001
Figure imgf000095_0001
Additional exemplary Megalin binding moi eties or ligands are disclosed in U.S. Patent 7,560,431, U.S. Patent 8,877,714, U.S. Patent 8,795,627; International Patent Application WO 2006/138343, U.S. Patent 9,388,418, U.S. Patent 10,065,993, International Patent Application WO 2017/100700, and International Patent Application WO 2018/232122, the entire contents of each of which are hereby incorporated by reference.
In some embodiments, a targeted kidney cell surface factor for delivery of a targeting moiety-associated modulatory nucleic acid agent of the instant disclosure is Megalin, or a fragment, or a variant thereof.
In some embodiments, a targeting moiety is or comprises a megalin-binding moiety. In particular embodiments, a targeting moiety binds an extracellular domain (e.g., to a site on the extracellular domain, e.g., a site that is exposed when megalin is on a cell surface) of megalin. In other particular embodiments, a conjugate comprises a targeting moiety that binds an extracellular domain (e.g., to a site on an extracellular domain, e.g., a site that is exposed when megalin is on a cell surface) of megalin and, upon binding to megalin, causes the internalization of megalin. Cubilin-Binding Moieties
Cubilin is a receptor of about 460kDa. Cubilin is also known as IFCR, Gp280, Intrinsic Factor- Vitamin B12 Receptor, MGA1, or IGS1. As an extracellular protein, Cubilin can interact with other membrane proteins, e.g., Megalin. One of the functions of Cubilin is as a receptor for intrinsic factor-vitamin B12 complexes.
Human Cubilin protein sequence:
MMNMSLPFLWSLLTLLIFAEVNGEAGELELQRQKRSINLQQPRMATERGNLVFLTGSAQ NIEFRTGSLGKIKLNDEDLSECLHQIQKNKEDIIELKGSAIGLPQNISSQIYQLNSKLVDLE RKFQGLQQTVDKKVCSSNPCQNGGTCLNLHDSFFCICPPQWKGPLCSADVNECEIYSGT PLSCQNGGTCVNTMGSYSCHCPPETYGPQCASKYDDCEGGSVARCVHGICEDLMREQA GEPKYSCVCDAGWMFSPNSPACTLDRDECSFQPGPCSTLVQCFNTQGSFYCGACPTGW QGNGYICEDINECEINNGGCSVAPPVECVNTPGSSHCQACPPGYQGDGRVCTLTDICSVS NGGCHPDASCSSTLGSLPLCTCLPGYTGNGYGPNGCVQLSNICLSHPCLNGQCIDTVSGY FCKCDSGWTGVNCTENINECLSNPCLNGGTCVDGVDSFSCECTRLWTGALCQVPQQVC GESLSGINGSFSYRSPDVGYVHDVNCFWVIKTEMGKVLRITFTFFRLESMDNCPHEFLQV YDGDSSSAFQLGRFCGSSLPHELLSSDNALYFHLYSEHLRNGRGFTVRWETQQPECGGIL TGPYGSIKSPGYPGNYPPGRDCVWIVVTSPDLLVTFTFGTLSLEHHDDCNKDYLEIRDGP LYQDPLLGKFCTTFSVPPLQTTGPFARIHFHSDSQISDQGFHITYLTSPSDLRCGGNYTDP EGELFLPELSGPFTHTRQCVYMMKQPQGEQIQINFTHVELQCQSDSSQNYIEVRDGETLL GKVCGNGTISHIKSITNSVWIRFKIDASVEKASFRAVYQVACGDELTGEGVIRSPFFPNVY PGERTCRWTIHQPQSQVILLNFTVFEIGS S AHCETDYVEIGS S SILGSPENKKYCGTDIPSFI
TSVYNFLYVTFVKSSSTENHGFMAKFSAEDLACGEILTESTGTIQSPGHPNVYPHGINCT WHILVQPNHLIHLMFETFHLEFHYNCTNDYLEVYDTDSETSLGRYCGKSIPPSLTSSGNS LMLVF VTD SDL AYEGFLINYEAIS AATACLQD YTDDLGTFT SPNFPNNYPNNWECI YRIT VRTGQLIAVHFTNFSLEEAIGNYYTDFLEIRDGGYEKSPLLGIFYGSNLPPTIISHSNKLWL KFKSDQIDTRSGFSAYWDGSSTGCGGNLTTSSGTFISPNYPMPYYHSSECYWWLKSSHG SAFELEFKDFHLEHHPNCTLDYLAVYDGPSSNSHLLTQLCGDEKPPLIRSSGDSMFIKLRT DEGQQGRGFKAEYRQTCENVVIVNQTYGILESIGYPNPYSENQHCNWTIRATTGNTVNY TFLAFDLEHHINCSTDYLELYDGPRQMGRYCGVDLPPPGSTTSSKLQVLLLTDGVGRRE KGFQMQWFVYGCGGELSGATGSFSSPGFPNRYPPNKECIWYIRTDPGSSIQLTIHDFDVE YHSRCNFDVLEIYGGPDFHSPRIAQLCTQRSPENPMQVSSTGNELAIRFKTDLSINGRGFN ASWQAVTGGCGGIFQAPSGEIHSPNYPSPYRSNTDCSWVIRVDRNHRVLLNFTDFDLEP QDSCIMAYDGLSSTMSRLARTCGREQLANPIVSSGNSLFLRFQSGPSRQNRGFRAQFRQA CGGHILTS SFDTVS SPRFPANYPNNQNC SWIIQ AQPPLNHITLSFTHFELERSTTC ARDF VE ILDGGHEDAPLRGRYCGTDMPHPITSFSSALTLRFVSDSSISAGGFHTTVTASVSACGGTF YMAEGIFNSPGYPDIYPPNVECVWNIVSSPGNRLQLSFISFQLEDSQDCSRDFVEIREGNA TGHLVGRYCGNSFPLNYSSIVGHTLWVRFISDGSGSGTGFQATFMKIFGNDNIVGTHGK VASPFWPENYPHNSNYQWTVNVNASHVVHGRILEMDIEEIQNCYYDKLRIYDGPSIHAR LIGAYCGTQTESFSSTGNSLTFHFYSDSSISGKGFLLEWFAVDAPDGVLPTIAPGACGGFL RTGDAPVFLFSPGWPDSYSNRVDCTWLIQAPDSTVELNILSLDIESHRTCAYDSLVIRDG DNNLAQQLAVLCGREIPGPIRSTGEYMFIRFTSDSSVTRAGFNASFHKSCGGYLHADRGII T SPK YPET YP SNLNC S WHVL VQ S GLTI A VHFEQPF QIPNGD S S CNQGD YL VLRNGPDIC S PPLGPPGGNGHFCGSHASSTLFTSDNQMFVQFISDHSNEGQGFKIKYEAKSLACGGNVYI HD AD S AGYVT SPNHPHN YPPH A DC I W IL AAPPETRIQLQFEDRFDIE VTPNC TSNYLELR DGVDSDAPILSKFCGTSLPSSQWSSGEVMYLRFRSDNSPTHVGFKAKYSIAQCGGRVPG QSGVVESIGHPTLPYRDNLFCEWHLQGLSGHYLTISFEDFNLQNSSGCEKDFVEIWDNHT SGNILGRYCGNTIPDSIDTSSNTAVVRFVTDGSVTASGFRLRFESSMEECGGDLQGSIGTF TSPNYPNPNPHGRICEWRITAPEGRRITLMFNNLRLATHPSCNNEHVIVFNGIRSNSPQLE KLCSSVNVSNEIKSSGNTMKVIFFTDGSRPYGGFTASYTSSEDAVCGGSLPNTPEGNFTSP GYDGVRNYSRNLNCEWTLSNPNQGNSSISIHFEDFYLESHQDCQFDVLEFRVGDADGPL MWRLCGPSKPTLPLVIPYSQVWIHFVTNERVEHIGFHAKYSFTDCGGIQIGDSGVITSPNY PNAYD SLTHC S SLLE APQGHTITLTF SDFDIEPHTTC AWD S VT VRNGGSPE SPIIGQ YCGN SNPRTIQSGSNQLVVTFNSDHSLQGGGFYATWNTQTLGCGGIFHSDNGTIRSPHWPQNFP ENSRCSWTAITHKSKHLEISFDNNFLIPSGDGQCQNSFVKVWAGTEEVDKALLATGCGN VAPGPVITPSNTFTAVFQSQEAPAQGFSASFVSRCGSNFTGPSGYIISPNYPKQYDNNMN CTYVIEANPLSVVLLTFVSFHLEARSAVTGSCVNDGVHIIRGYSVMSTPFATVCGDEMPA PLTIAGPVLLNFYSNEQITDFGFKFSYRIISCGGVFNFSSGIITSPAYSYADYPNDMHCLYTI TVSDDKVIELKFSDFDVVPSTSCSHDYLAIYDGANTSDPLLGKFCGSKRPPNVKSSNNSM LLVFKTDSFQTAKGWKMSFRQTLGPQQGCGGYLTGSNNTFASPDSDSNGMYDKNLNC
VWIIIAPVNKVIHLTFNTFALEAASTRQRCLYDYVKLYDGDSENANLAGTFCGSTVPAPF ISSGNFLTVQFISDLTLEREGFNATYTIMDMPCGGTYNATWTPQNISSPNSSDPDVPFSICT WVIDSPPHQQVKITVWALQLTSQDCTQNYLQLQDSPQGHGNSRFQFCGRNASAVPVFY SSMSTAMVIFKSGVVNRNSRMSFTYQIADCNRDYHKAFGNLRSPGWPDNYDNDKDCT VTLTAPQNHTISLFFHSLGIENSVECRNDFLEVRNGSNSNSPLLGKYCGTLLPNPVFSQNN ELYLRFKSDSVTSDRGYEHWTSSPSGCGGTLYGDRGSFTSPGYPGTYPNNTYCEWVLVA PAGRLVTINFYFISIDDPGDCVQNYLTLYDGPNASSPSSGPYCGGDTSIAPFVASSNQVFI KFHADYARRPSAFRLTWDS (SEQ ID NO: 6)
The extracellular domain of Cubilin includes repeats of CUB domains (complement Clr/Cls, Uegf [epidermal growth factor-related sea urchin protein], and bone morphogenic protein 1) and EGF-type repeats. A typical structure of Cubilin is disclosed in Figure 1 of Marzolo and Farfan (2011), Biol Res 44: 89-105, the entire contents of which are hereby incorporated by reference. The extracellular domain of Cubilin may also include one or more post-translational modifications, such as glycosylation.
Cubilin has been reported to be found on surfaces of one or more of the following tissues and/or cells: immune cells (e.g., bone marrow cells, lymph node cells, thymic cells, peripheral blood mononuclear cells [e.g., myeloid and/or lymphoid cells], erythrocytes, eosinophils, neutrophils, and/or platelets); nervous system (e.g., brain tissue, cortex, cerebellum, retinal cells, spinal cord cells, nerve cells, neurons, and/or supporting cells; endothelial cells; muscle (e.g., heart muscle, smooth muscle, and/or skeletal muscle); small intestine; colon; adipocytes; kidney; liver; lung; splenic; stomach; esophagus; bladder; pancreas; thyroid; salivary gland; adrenal gland; pituitary gland; breast; skin; ovary; uterus; placenta; prostate; and testis. In kidney tissue, Cubilin has been reported to be found on the surface of proximal tubular epithelial cells and podocytes. Several ligands of Cubilin have been identified, some of which are disclosed in Nielsen et al. 2016.
Table 2: Exemplary Cubilin Ligands
Figure imgf000098_0001
Figure imgf000099_0001
Additional exemplary Cubilin binding moieties or ligands are disclosed in U.S. Patent 10,065,993, International Patent Application WO 2017/100700, International Patent Application WO 2018/232122, and International Patent Application WO 2015/027205, the entire contents of each of which are hereby incorporated by reference.
In some embodiments, a kidney cell surface factor is Cubilin, or a fragment, or a variant thereof.
In some embodiments, a targeting moiety is or comprises a Cubilin-binding moiety.
In some embodiments, a targeting moiety (e.g., a megalin binding moiety) is chosen from: a peptide, an aminoglycoside, an endogenous ligand (e.g., a ligand disclosed in Table 1 or Table 2, or an analog or variant thereof), a xenobiotic, an antibody or a fragment thereof, or any combination thereof.
In some embodiments, a targeting moiety (e.g., a megalin binding moiety) is or comprises a peptide. In some embodiments, a peptide is chosen from: a fragment of receptor associated protein (RAP), e.g., a RAP fragment comprising residues 219-323); a peptide derived from a radiopharmaceutical conjugate such as ocreotide, ocreotate, exendin, minigastrin, and/or neurotensin; or any combination thereof.
RAP (receptor-associated protein) is a cellular protein comprising about 300 amino acids and is encoded by the LRPAP1 gene. An exemplary RAP sequence is provided by NP_002328.1, and encoded by NM_002337.4. RAP has been shown to bind to Megalin to suppress the interaction of the Megalin receptor with one or more ligands (Willnow et al., EMBO J. 15, 2632-2639, 1996). Studies have shown that a minimal functional domain of RAP comprising about 104 amino acids retains RAP’s receptor binding and inhibition.
RAP protein sequence as provided by NM_002337.4:
MAPRRVRSFLRGLPALLLLLLFLGPWPAASHGGKYSREKNQPKPSPKRESGEEFRMEKL NQLWEKAQRLHLPPVRLAELHADLKIQERDELAWKKLKLDGLDEDGEKEARLIRNLNV ILAKYGLDGKKDARQVTSNSLSGTQEDGLDDPRLEKLWHKAKTSGKFSGEELDKLWRE FLHHKEKVHEYNVLLETLSRTEEIHENVISPSDLSDIKGSVLHSRHTELKEKLRSINQGLD RLRRVSHQGYSTEAEFEEPRVIDLWDLAQSANLTDKELEAFREELKHFEAKIEKHNHYQ KQLEIAHEKLRHAESVGDGERVSRSREKHALLEGRTKELGYTVKKHLQDLSGRISRARH NEL (SEQ ID NO: 7)
In some embodiments, a RAP fragment comprises a fragment of SEQ ID NO: 7, or a sequence with at least 90% identity thereto. In some embodiments, a RAP fragment comprises an LDL receptor binding domain of RAP. In some embodiments, a RAP fragment comprises a fragment of about -104 amino acids. In some embodiments, a RAP fragment is or comprises residues 219-323 of RAP.
Exemplary RAP fragments are disclosed in U.S. Patent Application US 2008/0153753A1, the entire contents of which are hereby incorporated by reference.
In some embodiments, a peptide disclosed herein further comprise one or more fragments, domains, and/or residues.
In some embodiments, a peptide disclosed herein comprises one or more modified amino acids.
In some embodiments, a peptide has one or more, or all of the following characteristics: (i) low molecular weight, e.g., 0.5kDa to lOkDa; (ii) limited charge at pH 7, e.g., -10 to +10; (iii) binding to a cell surface receptor, e g., Megalin, Cubilin, or both. Exemplary characteristics of peptides that may be useful in a targeting moiety are disclosed in Vegt, et al. Eur J Nucl Med Mol Imaging. 2011, and Wischnjow et al, Bioconjugate Chem. 2016, 27, 1050-1057, the entire contents of each of which are incorporated by reference herein.
In some embodiments, a targeting moiety (e.g., a megalin binding moiety) is or comprises an aminoglycoside. In some embodiments, an aminoglycoside is chosen from one or more, or all of: streptomycin, neomycin, kanamycin, paromomycin, gentamicin, G-418 (geneticin), ELX-02, tobramycin, amikacin, netilmicin, spectinomycin, sisomicin, dibekacin, isepamicin, framycetin, paromomycin, apramycin, fradiomycin, arbekacin, plazomicin, or a derivative or a variant thereof.
In some embodiments, an aminoglycoside disclosed herein has minimal bactericidal activity and/or toxicity, e.g., nephrotoxicity.
In some embodiments, an aminoglycoside comprises a variant having reduced toxicity, e.g., reduced nephrotoxicity as compared to an aminoglycoside without a variant. In some embodiments, an aminoglycoside comprises a variant having reduced bactericidal activity as compared to an aminoglycoside without a variant. In some embodiments, an aminoglycoside comprises a variant which retains activity, e.g., readthrough activity of premature termination codons, as compared to an aminoglycoside without a variant. In some embodiments, a variant of an aminoglycoside has reduced overall cationic charge as compared to an aminoglycoside without a variant. Exemplary aminoglycosides and variants thereof are disclosed in: Popadynec M. et al., (2021) ACS Med. Chem. Lett. 12( 9), 1486-1492; and in Brasell EJ et al., (2019), PLoS ONE 14(12): e0223954; the entire contents of each of which is hereby incorporated by reference.
In some embodiments, an aminoglycoside comprises an analog of an aminoglycoside having reduced antimicrobial activity (e.g., an aminoglycoside produced by resistance mutations in bacteria), and/or reduced endosomal or lysosomal stability, or both.
In some embodiments, an aminoglycoside has one or more, or all of the following characteristics: (i) high potency for binding to a cell surface factor, e.g., Megalin, Cubilin, or both; (ii) low nephrotoxicity; (iii) low ototoxicity; (iv) reduced endosomal or lysosomal stability; (v) reduced antimicrobial activity; or (vi) a combination of any one or all of (i) to (v).
In some embodiments, an aminoglycoside disclosed herein binds to one or more extracellular domains of a cell surface factor (e.g., Megalin, Cubilin, or both). In some embodiments, an aminoglycoside disclosed herein binds a cell surface receptor at or near one or more complement type repeats. Exemplary binding of an aminoglycoside to human Megalin is disclosed in Dagil R et al., (2013) Journal of Biological Chemistry; 288(6); 4424-4435; the entire contents of which are hereby incorporated by reference.
In some embodiments, a targeting moiety (e.g., a megalin binding moiety) is or comprises an endogenous ligand, e.g., a ligand disclosed in Table 1. In some embodiments, an endogenous ligand is chosen from: vitamin carrier proteins, apolioproteins, peptide hormones, or any combination thereof.
In some embodiments, when a targeting moiety (e.g., a megalin binding moiety) is or comprises a vitamin carrier protein, a vitamin carrier protein comprises a vitamin carried by a vitamin carrier protein. For example, for a vitamin carrier protein that carries Vitamin D, in some embodiments such a vitamin carrier protein comprises both a vitamin carrier protein and Vitamin D when used in a kidney-specific binding moiety described herein.
In some embodiments, a targeting moiety (e.g., a megalin binding moiety) is or comprises a ligand or substrate that binds to or is carried by another protein, e.g., a receptor or a carrier protein. Exemplary endogenous ligands are disclosed in Nielsen R. et al. (2016), Kidney Int. 89(l):58-67 the entire contents of which are hereby incorporated by reference.
In some embodiments, a targeting moiety (e.g., a megalin binding moiety) is or comprises a xenobiotic. In some embodiments, a xenobiotic is chosen from: Polymixins, aprotinin, trichosanthin, or any combination thereof.
In some embodiments, a targeting moiety (e.g., a megalin binding moiety) is or comprises an antibody or a fragment thereof. In some embodiments, a targeting moiety (e.g., a megalin binding moiety) is an antibody that binds to a relevant cell surface factor, e.g., Megalin. In some embodiments, an anti-Megalin antibody is a monoclonal antibody or a fragment thereof. In some embodiments, an anti-Megalin antibody is a polyclonal antibody or a fragment thereof. In some embodiments, an anti-Megalin antibody is a bispecific or multispecific antibody, or a fragment thereof. In some embodiments, a bispecific or multispecific antibody binds to Megalin and one or more additional antigens, e.g., a polypeptide present in podocytes.
Exemplary antibodies that bind Megalin include anti-Megalin autoantibodies found in patients having antibrush border antibodies and renal failure (ABBA disease), see e.g., Larsen C. et al., (2018) J Am Soc Nephrol. 29(2): 644-653. Anti-Megalin antibodies are also disclosed in: Perez-Gomez MV et al., (2020) Clin Kidney J., 13(3):281 -286; Dinesh KP et al., (2019) Am J Kidney Dis., 74(1): 132-137, Larsen CP et al., (2018) J Am Soc Nephrol., (2):644-653, and Gamayo A et al., (2019) Clin Kidney J., 13(3):468-472, the entire contents of each of which is hereby incorporated by reference.
Those skilled in the art would also appreciate various commercially available anti-Megalin antibodies (or fragments thereof) that can be utilized as a targeting moiety in a conjugate agent disclosed herein.
In some embodiments, an antibody used in a conjugate agent disclosed herein include one or more modifications of an Fc domain, e.g., an Fc variant. In some embodiments, an Fc variant comprises an effector null mutation. In some embodiments, an Fc variant has one or more of the following properties: (1) reduced effector function (e.g., reduced ADCC, ADCP and/or CDC); (2) reduced binding to one or more Fc receptors; and/or (3) reduced binding to Clq complement. In some embodiments, the reduction in any one, or all of properties (l)-(3 ) is compared to an otherwise similar antibody with a wildtype Fc region. In some embodiments, an Activin A antibody agent comprising a variant Fc region has reduced affinity to a human Fc receptor, e.g., FcyR I, FcyR II and/or FcyR III. Exemplary Fc region variants are disclosed in Saunders K.O., (2019) Frontiers in Immunology; vol 10, Article 296, the entire contents of which is hereby incorporated by reference. For example, a Fc region variant is or comprises a modification provided in Table 3 of Saunders KO (2019).
In some embodiments, an antibody used in a conjugate agent disclosed herein is a neutral binder, e.g., an antibody having no antagonism or blockage of binding sites for other substrates.
In some embodiments, an antibody which binds to Megalin and is used in a conjugate agent disclosed herein can be trafficked intracellularly with Megalin. An exemplary anti-Megalin antibody with such properties is the 20B monoclonal antibody disclosed in Shah M. et al. (2013), Journal of Cell Biology 202(1): 113-127, the entire contents of which are herein incorporated by reference.
In some embodiments, an antibody used in a conjugate agent disclosed herein comprises an Fc variant and is a neutral binder.
Linkers
In some embodiments, a conjugate agent has the structure of the following formula:
Figure imgf000103_0001
wherein a is 1-8; and each of the binding moiety, linker, and modulatory nucleic acid is as defined above and described herein.
The synthesis and application of conjugate agents (e.g., “bioconjugates”) as tools for life science research, as diagnostic reagents, and as therapeutic agents has exploded in recent years and development of conjugate agents remains an area of intensive activity. In some embodiments, a bioconjugate or conjugate agent comprises a modulatory nucleic acid that is chemically conjugated or linked covalently to a binding (e.g., targeting) moiety.
In some embodiments, a conjugate agent is prepared by conjugating or covalently linking a modulatory nucleic acid agent to a binding moiety. In some embodiments, the modulatory nucleic acid may be linked to a binding moiety by, for example, reaction of the modulatory nucleic acid in-solution with a binding moiety. The conjugate agent may also be prepared in a single synthesis, for example using known synthesis methods for preparation of GalNAc-conjugated nucleic acids by solid-phase means. (For example, U.S. Pat. No. 9,422,562, W02009073809, U.S. Pat. No. 8,106,022, U.S. Pat. No. 8,828,956, U.S. Pat. No. 9,133,461, and U.S. Pat. No., 10,131,907, each of which is incorporated by reference in its entirety).
Regardless of how produced and depending on the desired properties of the conjugate agent, it may or may not be advantageous to include a spacer or linker between the modulatory nucleic acid and the binding moiety. If it is advantageous to include a linker, then linkers can be of many different types and chemical compositions.
Generally, linkers are designated as “cleavable” or “non-cleavable”. Cleavable linkers are typically employed when it is desired that the payload and binding moiety to which it is conjugated be released so that either or both can better carry out their function (For example, U.S. Pat. No. 10,808,039 and U.S. Pat. No. 9,463,252, each of which is incorporated by reference in its entirety.) Non-cleavable linkers are typically employed to maintain the desired activity, performance and stability of the conjugate agent, for example enzymes linked to probes or (m)Abs to facilitate ELISA assays, to increase affinity, or bi-specificity, etc. Amongst the cleavable linkers are those that are cleaved chemically, for example by hydrolysis, change in pH, reduction or oxidation, and those that are cleaved enzymatically, for example by action of a protease, an esterase, a glucosidase, a glucuronidase, galactosidase, a phosphatase, phosphodiesterase, nuclease, lipase or any enzyme that is capable of cleaving a linker to liberate the biomolecule from the other compound.
In some embodiments, a cleavable linker is or comprises a disulfide linkage, an ester, a phosphodiester, a saccharide, or a lipid.
In some embodiments, a non-cleavable linker is chemically, enzymatically, or otherwise biochemically and physiologically stable. As such, a non-cleavable linker does not contain linkages that are chemically, biochemically, enzymatically cleavable or are otherwise physiologically unstable.
In some embodiments, whether cleavable or non-cleavable, a linker can be installed by a chemical linking reaction between the modulatory nucleic acid and the binding moiety to which it is being conjugated. The modulatory nucleic acid and binding moiety may or may not be first modified to increase or facilitate reactivity towards one another. Such modification can also increase or improve the specificity of the conjugation reaction and degree of conjugation when that is desired. The linkers may be installed in a single reaction or by stepwise reactions until the desired linker and modulatory nucleic acid have been prepared.
Non-limiting examples of chemical linking reactions to form conjugate agents include reaction of various thiols to form disulfides, reaction between thiols and alkyl halides or mal eimides to form thioethers, reaction of alkynes with azides to form triazoles (“Click Reaction”), reaction between aldehydes and hydrazides or amines, or aminoxy compounds to form hydrazones, imines and oxy imines, reaction between carboxylic acids and amines, thiols or alcohols (i.e., nucleophiles) to form amides, thioesters and esters. The carboxylic acids may be activated in situ in the presence of the amines, thiols or alcohols so as to be made reactive or may be pre-activated prior to addition of the nucleophile, for example by converting to activated esters of N- hydroxysuccinimide (NHS) or sulfonated-NHS. Many reviews of chemical linking reactions exist for example Spicer et al. (2018) Chem. Rev. 2018, 118, 16, 7702-7743.
The reaction of thiols with maleimides is very widely used, see for example (Revasco et al (2018) Chem. Eur. J. 10.1002/chem.201803174) as is the Click Reaction see for example (Fantoni et al., (2021) Chem. Rev., 121, 12, 7122-7154), as is hydrazide formation (See HyNic Peptide Conjugation Protocol, Dirksen et al (2006) J. Am. Chem. Soc., 128, 49, 15602-15603, Kozlov et al. (2004)Biopolymers73(5):621-30. doi: 10.1002/bip.20009). Numerous companies sell chemical compounds and kits with protocols that enable conjugate agents comprising various linkers to be prepared in a straightforward fashion.
As defined generally above and described herein, the linker is a bivalent group that connects or links the binding moiety to the modulatory nucleic acid moiety.
In some embodiments, the linker is or comprises a bivalent straight or branched C1-40 aliphatic chain, wherein one or more methylene units of the aliphatic chain are replaced by a group selected from -CH(R1)-, -C(R1)2-, -O-, -S-, -N(R)-, -C(=O)-, -C(=S)-, -C(=NR), -N(R)C(=O)-, -C(=O)N(R)-, -N(R)C(=S)-, -C(=S)N(R)-, -OC(=O)-, -C(=O)O-, -SC(=O)-, -C(=O)S-, -N(R)C(=O)N(R)-, -N(R)C(=O)O-, -OC(=O)N(R)-, -N(R)C(=O)S-, -SC(=O)N(R)-, -OC(=O)O-, -N(R)C(=NR)-, -N(R)C(=NR)N(R)-, a 3- to 6-membered saturated or partially unsaturated carbocyclic ring, phenyl, a 3- to 6-membered saturated or partially unsaturated heterocyclic ring having 1-3 heteroatoms independently selected from nitrogen, oxygen, and sulfur, a 5- to 6-membered heteroaryl ring having 1 -4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, wherein:
R1 is an amino acid side chain; and
R is selected from hydrogen or an optionally substituted Ci-6 aliphatic, a 3- to 6-membered saturated or partially unsaturated carbocyclic ring, phenyl, a 3 - to 6-membered saturated or partially unsaturated heterocyclic ring having 1-3 heteroatoms independently selected from nitrogen, oxygen, and sulfur, and a 5- to 6-membered heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur.
In some embodiments, the linker is or comprises a bivalent straight or branched C1-35 aliphatic chain, wherein one or more methylene units of the aliphatic chain are replaced by a group selected from -CHCR1)-, -C(R1)2-, -O-, -S-, -N(R)-, -C(=O)-, -C(=S)-, -C(=NR), -N(R)C(=O)-, -C(=O)N(R)-, -N(R)C(=S)-, -C(=S)N(R)-, -OC(=O)-, -C(=O)O-, -SC(=O)-, -C(=O)S-, -N(R)C(=O)N(R)-, -N(R)C(=O)O-, -OC(=O)N(R)-, -N(R)C(=O)S-, -SC(=O)N(R)-, -OC(=O)O-, -N(R)C(=NR)-, -N(R)C(=NR)N(R)-, a 3- to 6-membered saturated or partially unsaturated carbocyclic ring, phenyl, a 3- to 6-membered saturated or partially unsaturated heterocyclic ring having 1-3 heteroatoms independently selected from nitrogen, oxygen, and sulfur, a 5- to 6-membered heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur.
In some embodiments, the linker is or comprises a bivalent straight or branched C1-30 aliphatic chain, wherein one or more methylene units of the aliphatic chain are replaced by a group selected from -CHCR1)-, -C(R1)2-, -O-, -S-, -N(R)-, -C(=O)-, -C(=S)-, -C(=NR), -N(R)C(=O)-, -C(=O)N(R)-, -N(R)C(=S)-, -C(=S)N(R)-, -OC(=O)-, -C(=O)O-, -SC(=O)-, -C(=O)S-, -N(R)C(=O)N(R)-, -N(R)C(=O)O-, -OC(=O)N(R)-, -N(R)C(=O)S-, -SC(=O)N(R)-, -OC(=O)O-, -N(R)C(=NR)-, -N(R)C(=NR)N(R)-, a 3- to 6-membered saturated or partially unsaturated carbocyclic ring, phenyl, a 3- to 6-membered saturated or partially unsaturated heterocyclic ring having 1-3 heteroatoms independently selected from nitrogen, oxygen, and sulfur, a 5- to 6-membered heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur.
In some embodiments, the linker is or comprises a bivalent straight or branched C1-25 aliphatic chain, wherein one or more methylene units of the aliphatic chain are replaced by a group selected from -CHCR1)-, -QR1)!-, -O-, -S-, -N(R)-, -C(=O)-, -C(=S)-, -C(=NR), -N(R)C(=O)-, -C(=O)N(R)-, -N(R)C(=S)-, -C(=S)N(R)-, -OC(=O)-, -C(=O)O-, -SC(=O)-, -C(=O)S-, -N(R)C(=O)N(R)-, -N(R)C(=O)O-, -OC(=O)N(R)-, -N(R)C(=O)S-, -SC(=O)N(R)-, -OC(=O)O-, -N(R)C(=NR)-, -N(R)C(=NR)N(R)-, a 3- to 6-membered saturated or partially unsaturated carbocyclic ring, phenyl, a 3- to 6-membered saturated or partially unsaturated heterocyclic ring having 1-3 heteroatoms independently selected from nitrogen, oxygen, and sulfur, a 5- to 6-membered heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur.
In some embodiments, the linker is or comprises a bivalent straight or branched C1-20 aliphatic chain, wherein one or more methylene units of the aliphatic chain are replaced by a group selected from -CHCR1)-, -C(R1)2-, -O-, -S-, -N(R)-, -C(=O)-, -C(=S)-, -C(=NR), -N(R)C(=O)-, -C(=O)N(R)-, -N(R)C(=S)-, -C(=S)N(R)-, -OC(=O)-, -C(=O)O-, -SC(=O)-, -C(=O)S-, -N(R)C(=O)N(R)-, -N(R)C(=O)O-, -OC(=O)N(R)-, -N(R)C(=O)S-, -SC(=O)N(R)-, -OC(=O)O-, -N(R)C(=NR)-, -N(R)C(=NR)N(R)-, a 3- to 6-membered saturated or partially unsaturated carbocyclic ring, phenyl, a 3- to 6-membered saturated or partially unsaturated heterocyclic ring having 1-3 heteroatoms independently selected from nitrogen, oxygen, and sulfur, a 5- to 6-membered heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur.
In some embodiments, the linker is or comprises a bivalent straight or branched C1-15 aliphatic chain, wherein one or more methylene units of the aliphatic chain are replaced by a group selected from -CHCR1)-, -C(R1)2-, -O-, -S-, -N(R)-, -C(=O)-, -C(=S)-, -C(=NR), -N(R)C(=O)-, -C(=O)N(R)-, -N(R)C(=S)-, -C(=S)N(R)-, -OC(=O)-, -C(=O)O-, -SC(=O)-, -C(=O)S-, -N(R)C(=O)N(R)-, -N(R)C(=O)O-, -OC(=O)N(R)-, -N(R)C(=O)S-, -SC(=O)N(R)-, -OC(=O)O-, -N(R)C(=NR)-, -N(R)C(=NR)N(R)-, a 3- to 6-membered saturated or partially unsaturated carbocyclic ring, phenyl, a 3- to 6-membered saturated or partially unsaturated heterocyclic ring having 1-3 heteroatoms independently selected from nitrogen, oxygen, and sulfur, a 5- to 6-membered heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur.
In some embodiments, the linker is or comprises a bivalent straight or branched C1-10 aliphatic chain, wherein one or more methylene units of the aliphatic chain are replaced by a group selected from -CHCR1)-, -C(R1)2-, -O-, -S-, -N(R)-, -C(=O)-, -C(=S)-, -C(=NR), -N(R)C(=O)-, -C(=O)N(R)-, -N(R)C(=S)-, -C(=S)N(R)-, -OC(=O)-, -C(=O)O-, -SC(=O)-, -C(=O)S-, -N(R)C(=O)N(R)-, -N(R)C(=O)O-, -OC(=O)N(R)-, -N(R)C(=O)S-, -SC(=O)N(R)-, -OC(=O)O-, -N(R)C(=NR)-, -N(R)C(=NR)N(R)-, a 3- to 6-membered saturated or partially unsaturated carbocyclic ring, phenyl, a 3- to 6-membered saturated or partially unsaturated heterocyclic ring having 1-3 heteroatoms independently selected from nitrogen, oxygen, and sulfur, a 5- to 6-membered heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur.
In some embodiments, the linker is or comprises a bivalent straight or branched Ci-5 aliphatic chain, wherein one or more methylene units of the aliphatic chain are replaced by a group selected from -CHCR1)-, -C(R1)2-, -O-, -S-, -N(R)-, -C(=O)-, -C(=S)-, -C(=NR), -N(R)C(=O)-, -C(=O)N(R)-, -N(R)C(=S)-, -C(=S)N(R)-, -OC(=O)-, -C(=O)O-, -SC(=O)-, -C(=O)S-, -N(R)C(=O)N(R)-, -N(R)C(=O)O-, -OC(=O)N(R)-, -N(R)C(=O)S-, -SC(=O)N(R)-, -OC(=O)O-, -N(R)C(=NR)-, -N(R)C(=NR)N(R)-, a 3- to 6-membered saturated or partially unsaturated carbocyclic ring, phenyl, a 3- to 6-membered saturated or partially unsaturated heterocyclic ring having 1-3 heteroatoms independently selected from nitrogen, oxygen, and sulfur, a 5- to 6-membered heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur.
In some embodiments, the linker is or comprises a structure selected from
Figure imgf000108_0001
In some embodiments, the cleavable linker is a cathepsin-cleavable linker. In some such embodiments, the linker is or comprises a valine-citrulline (Val-Cit) motif:
Figure imgf000109_0001
wherein R is hydrogen or Ci-6 aliphatic.
In some embodiments, the valine-citrulline linker is or comprises
Figure imgf000109_0003
wherein R is hydrogen or Ci-6 aliphatic.
In some embodiments, the linker comprises a disulfide linkage. In some embodiments, the linker comprises a poly (ethyleneglycol) moiety (e.g., -(CH2CH2O)b-), wherein b is 1-50.
In some embodiments, the linker is or comprises a group selected from
Figure imgf000109_0002
Figure imgf000110_0001
Figure imgf000111_0001
Figure imgf000112_0001
wherein each of k, m, n, p, q, r, s, t, u, v, w, x, y, and z is 1-20; and
R is hydrogen or Ci-io aliphatic.
In some embodiments, k is 3.
In some embodiments, m is 3.
In some embodiments, n is 2. In some embodiments, n is 12.
In some embodiments, p is 3.
In some embodiments, each of m and p is 3.
In some embodiments, q is 1.
In some embodiments, r is 3. In some embodiments, r is 4. In some embodiments, r is 6.
In some embodiments, s is 3. In some embodiments, s is 4. In some embodiments, s is 6.
In some embodiments, each of r and s is 3. In some embodiments, each of r and s is 4. In some embodiments, each of r and s is 6.
In some embodiments, t is 3. In some embodiments, t is 5.
In some embodiments, u is 3. In some embodiments, u is 5.
In some embodiments, each of t and u is 3. In some embodiments, each of t and u is 5. In some embodiments, v is 3.
In some embodiments, w is 4.
In some embodiments, x is 8.
In some embodiments, y is 2.
In some embodiments, z is 1.
Other Conjugates
In certain embodiments, the ligand or conjugate is a lipid or lipid-based molecule Such a lipid or lipid-based molecule optionally binds a serum protein, e.g., human serum albumin (HSA). An HSA binding ligand allows for distribution of the conjugate to a target tissue, e.g., a non-kidney target tissue of the body. For example, the target tissue can be the liver, including parenchymal cells of the liver. Other molecules that can bind HSA can also be used as ligands. For example, naproxen or aspirin can be used. A lipid or lipid-based ligand can (a) increase resistance to degradation of the conjugate, (b) increase targeting or transport into a target cell or cell membrane, or (c) can be used to adjust binding to a serum protein, e.g., HSA.
.A lipid-based ligand can be used to inhibit, e.g., control the binding of the conjugate to a target tissue. For example, a lipid or lipid-based ligand that binds to HSA more strongly will be less likely to be targeted to the kidney and therefore less likely to be cleared from the body. A lipid or lipid-based ligand that binds to HSA less strongly can be used to target the conjugate to the kidney.
In certain embodiments, the lipid-based ligand binds HSA. Optionally, it binds HSA with a sufficient affinity such that the conjugate will be preferably distributed to a non-kidney tissue.
However, it is certain that the affinity not be so strong that the HSA-ligand binding cannot be reversed.
In other embodiments, the lipid-based ligand binds HSA weakly or not at all, such that the conjugate will be preferably distributed to the kidney. Other moieties that target to kidney cells can also be used in place of, or in addition to, the lipid-based ligand.
In another aspect, the ligand is a moiety, e.g., a vitamin, which is taken up by a target cell, e.g., a proliferating cell. These are particularly useful for treating disorders characterized by unwanted cell proliferation, e.g., of the malignant or non-malignant type, e.g., cancer cells.
I l l Exemplary vitamins include vitamin A, E, and K. Other exemplary vitamins include are B vitamin, e.g., folic acid, B 12, riboflavin, biotin, pyridoxal or other vitamins or nutrients taken up by target cells such as liver cells. Also included are HSA and low density lipoprotein (LDL).
Cell Permeation Agents
In another aspect, the ligand is a cell-permeation agent, optionally a helical cell-permeation agent. Optionally, the agent is amphipathic. An exemplary agent is a peptide such as tat or antennopedia. If the agent is a peptide, it can be modified, including a peptidylmimetic, invertomers, non-peptide or pseudo-peptide linkages, and use ofD-amino acids. The helical agent is preferably an alpha-helical agent, which preferably has a lipophilic and a lipophobic phase.
The ligand can be a peptide or peptidomimetic. A peptidomimetic (also referred to herein as an oligopeptidomimetic) is a molecule capable of folding into a defined three-dimensional structure similar to a natural peptide. The attachment of peptide and peptidomimetics to ASO or iRNA agents can affect pharmacokinetic distribution of the ASO or iRNA, such as by enhancing cellular recognition and absorption. The peptide or peptidomimetic moiety can be about 5-50 amino acids long, e.g., about 5, 10, 15, 20, 25, 30, 35, 40, 45, or 50 amino acids long.
A peptide or peptidomimetic can be, for example, a cell permeation peptide, cationic peptide, amphipathic peptide, or hydrophobic peptide (e.g., consisting primarily of Tyr, Trp, or Phe). The peptide moiety can be a dendrimer peptide, constrained peptide or crosslinked peptide. In another alternative, the peptide moiety can include a hydrophobic membrane translocation sequence (MTS).
A “cell permeation peptide” is capable of permeating a cell, e.g., a microbial cell, such as a bacterial or fungal cell, or a mammalian cell, such as a human cell. A microbial cell-permeating peptide can be, for example, an a-helical linear peptide (e.g., LL-37 or Ceropin Pl), a disulfide bond-containing peptide (e g., a -defensin, P-defensin or bactenecin), or a peptide containing only one or two dominating amino acids (e.g., PR-39 or indolicidin). A cell permeation peptide can also include a nuclear localization signal (NLS). For example, a cell permeation peptide can be a bipartite amphipathic peptide, such as MPG, which is derived from the fusion peptide domain of HIV-1 gp41 and the NLS of SV40 large T antigen (Simeoni et al., Nucl. Acids Res.31 :2717-2724, 2003). Carbohydrate Conjugates
In some embodiments of the compositions and methods of the present disclosure, an ASO or iRNA agent further comprises a carbohydrate. The carbohydrate conjugated ASO or iRNA agent is advantageous for the in vivo deliver}? of nucleic acids, as well as compositions suitable for in vivo therapeutic use. As used herein, “carbohydrate” refers to a compound which is either a carbohydrate per se made up of one or more monosaccharide units having at least 6 carbon atoms (which can be linear, branched or cyclic) with an oxygen, nitrogen or sulfur atom bonded to each carbon atom; or a compound having as a part thereof a carbohydrate moiety made up of one or more monosaccharide units each having at least six carbon atoms (which can be linear, branched or cyclic), with an oxygen, nitrogen or sulfur atom bonded to each carbon atom. Representative carbohydrates include the sugars (mono-, di-, tri-, and oligosaccharides containing from about 4, 5, 6, 7, 8, or 9 monosaccharide units), and polysaccharides such as starches, glycogen, cellulose and polysaccharide gums. Specific monosaccharides include C5 and above (e.g., C5, C6, C7, or C8) sugars; di- and tri saccharides include sugars having two or three monosaccharide units (e.g.. C5, C6, C7, or C8).
In certain embodiments, a carbohydrate conjugate for use in the compositions and methods of the present disclosure is a monosaccharide.
In certain embodiments, the monosaccharide is an N-acetylgalactosamine (GalNAc). GalNAc conjugates, which comprise one or more N-acetylgalactosamine (GalNAc) derivatives, are described, for example, in US 8, 106,022, the entire content of which is hereby incorporated herein by reference. In some embodiments, the GalNAc conjugate serves as a ligand that targets the ASO or iRNA agent to particular cells. In some embodiments, the GalNAc conjugate targets the ASO or iRNA agent to liver cell s, e.g , by serving as a ligand for the asial oglycoprotein receptor of liver cells (e.g., hepatocytes).
Other Nucleic Acid Modifications
In certain aspects, the kidney cell -targeting moieties and other modifications referenced herein can be used in combination with one or more of a variety of other nucleic acid modifications known in the art. Specific nucleic acid modifications explicitly contemplated for use with the SLC6A19-targeting nucleic acids of the instant disclosure (optionally in combination with other modifications as disclosed herein and/or as known in the art) include, without limitation, "extended nucleic acid" ("exNA") modifications, and a range of phosphoryl guanidine-containing backbone linkages ("PN chemistry"), among others.
"Extended nucleic acid" ("exNA") modifications typically refer to a form of modification that inserts a methyl group between the 3'-phosphate group of a first nucleoside and the 5'-carbon of a ribose of the next nucleoside (progressing from 5' to 3') in an oligonucleotide chain. Such canonical "exNA" modifications therefore have the following generic structure:
Figure imgf000116_0002
. In certain embodiments, "exNA" modifications that are optionally abasic and/or that possess longer extensions than methyl, and/or that are optionally substituted at one or more of the base, 2'-substituent and/or extended carbon chain, are also expressly contemplated. In some embodiments of the instant disclosure, the term "exNA" can therefore refer to any moiety having the structure:
Figure imgf000116_0001
, where: R1 is a nucleobase or is an abasic structure (e.g., H, CH3, or other modified structures, which are optionally substituted); R2 is a 2' substituent, including, e.g., without limitation, 2'-O-alkyl (e.g., 2'-O-methyl, etc.), 2'-F, etc.; and C1-10 refers to a 1-10 carbon chain that is optionally saturated or unsaturated and that harbors an optionally substituted alkyl (e g., optionally substituted methyl, ethyl, propyl, butyl, pentyl, hexyl, heptyl, octyl, nonyl, or decyl), alkenyl (e.g., optionally substituted methylene, ethylene, propylene, butylene, pentylene, hexylene, heptylene, octylene, nonylene, or decylene), or alkynyl (e.g., ethynyl, optionally substituted propynyl, butynyl, pentynyl, hexynyl, heptynyl, octynyl, nonynyl, or decynyl) chain.
In related embodiments, branching of the C1-10 region shown above is also contemplated for "exNA" modifications. For example, in some embodiments, an "exNA" modification of the
Figure imgf000117_0001
instant disclosure can also be represented by the formula , where R1 is a nucleobase or is an abasic structure (e.g., H, CH3, or other modified structures, which are optionally substituted); R2 is a 2' substituent, including, e.g., without limitation, 2'-O-alkyl (e.g., 2'-O-methyl, etc.), 2'-F, etc.; and the side substitution on the 5’- of the ribose can be 2-11 atoms, with various amounts of substituents at X and Y (from X=Y=H to long n- or branched alkyl structures).
In certain embodiments, "exNA" and/or other internucleoside modifications can be positioned at or near the 3'-end of an oligonucleotide therapeutic (e.g., at or near the 3'-end of the guide strand of a siRNA, at or near the 3'-end of an antisense agent, etc.), where, without wishing to be bound by theory, the exNA modification(s) can confer an exNA-modified nucleic acid payload with resistance against 3 '-exonuclease-mediated digestion (Yamada, K. et al., Nature Biotechnology 2024). In some embodiments, exNA-phosphorothioate internucleoside linkages can be positioned at the ultimate and optionally also at the penultimate intemucleoside linkage(s) of the 3'-terminus of a nucleic acid payload (e g., exNA-PS modification(s) positioned at the 3'- terminal region of the guide strand of a siRNA and/or ex-NA-PS modification(s) positioned at the 3'-terminal region of an antisense oligonucleotide). As disclosed in WO 2021/195533, exNA modifications can be used in combination with a range of other commonly used oligonucleotide modifications, including, without limitation, the following:
Figure imgf000117_0002
In such embodiments, it is also expressly contemplated that such exNA modifications can be used in concert with effectively any art-recognized base at the above-noted "Base" or R1 position, including, without limitation, the following representative natural and artificially modified bases known in the art:
Figure imgf000118_0001
Phosphoryl guanidine-containing backbone ("PN backbone") linkages are also expressly contemplated for use in combination with the kidney cell-targeting moieties of the instant disclosure. Such PN backbone modifications were initially identified in WO 2023/201095 as
Figure imgf000118_0002
members of the following genus: . Exemplified versions of PN backbone modifications include, without limitation, the following:
Figure imgf000118_0003
Figure imgf000119_0001
O or S. In particular embodiments, the PN backbone modifications are selected from among:
Figure imgf000119_0002
"nOOl" modification of WO 2023/201095). Exemplary PN backbone linkages are also disclosed, e.g., in U.S. Patent No. 11,208,430.
In addition to expressly contemplating herein use of the above-noted phosphoryl guanidine structures for modification of internucleoside linkages of the nucleic acid agents of the instant disclosure, a range of other guanidine-based moieties is also expressly contemplated herein for attachment to linkage phosphorous groups. Such other guanidine-based moieties include, without limitation, the following:
Figure imgf000120_0001
Figure imgf000121_0001
Figure imgf000122_0001
Figure imgf000123_0001
Figure imgf000124_0001
Figure imgf000125_0001
1z3l
Figure imgf000126_0001
Figure imgf000127_0001
RLS is independently -Cl, -Br, -F, N(Me)2, or NHCOCO3.
Other backbone modifications known in the art are also contemplated for use with the targeting moieties and payloads of the instant disclosure. In particular, inclusion of one or more mesyl phosphoramidate modification(s) and/or busyl phosphorami date (and/or other structurally related modification(s)) is expressly contemplated for the nucleic acid agents of the instant disclosure. Mesyl phosphoramidate and busyl phosphoramidate modifications have the following structure, as compared to phosphorothioate modifications (see Sergeeva et al. (2024) Front Chem., 12 - 2024 doi.org/10.3389/fchem.2024.1342178):
Figure imgf000128_0001
. In certain embodiments, oligomeric compounds
(including, e ,g., antisense oligonucleotides, siRNAs, etc., or portions thereof) comprise or consist of oligonucleotides consisting of linked nucleosides and having at least one intemucleoside linking group of the following formula:
Figure imgf000128_0002
, where X is selected from O or S, and R is selected from and, a substituted and, a heterocycle, a substituted heterocycle, an aromatic heterocycle, a substituted aromatic heterocycle, a diazole, a substituted diazole, a C1-C6 alkoxy, C1-C20 alkyl, C1-C6 alkenyl. C l- C6 alkynyl, substituted C1-C20 alkyl, substituted C1-C6 alkenyl substituted C1-C6 alkynyl, and a conjugate group. In certain embodiments, X is O and R is methyl, and the internucleoside linking group of the above formula is an internucleoside linking group of the formula immediately below.
In certain embodiments, oligomeric compounds (including, e.g., antisense oligonucleotides. siRNAs, etc , or portions thereof) comprise or consist of oligonucleotides consisting of linked nucleosides and having at least one intemucleoside linking group of the fol I owing formul a:
Figure imgf000129_0001
(a mesyl phosphorami date internucleoside linkage, see, e.g., WO
2023/278589).
In certain embodiments, oligomeric compounds (including, e.g., antisense oligonucleotides, siRNAs, etc., or portions thereof) comprise or consist of oligonucleotides consisting of linked nucleosides and having at least one internucleoside linking group of the fol 1 owi n g form ul a:
Figure imgf000129_0002
In certain embodiments, oligomeric compounds (including, e.g., antisense oligonucleotides, siRNAs, etc., or portions thereof) comprise or consist of oligonucleotides consisting of linked nucleosides and having at least one internucleoside linking group of the following formula:
Figure imgf000129_0003
. Modifications to the phosphate backbone may be positioned within an oligonucleotide agent of the instant disclosure in any position(s) known in the art and/or disclosed herein.
Modified oligonucleotides of the instant disclosure comprise at least one modification relative to an unmodified oligonucleotide (i.e., comprise at least one modified nucleoside (comprising a modified sugar moiety, a stereo-non-standard nucleoside, and/or a modified nucleobase) and/or at least one modified internucleoside linkage). In certain embodiments, the modified internucleoside linkage is a modified internucleoside linking group having any of the above formulas. In certain embodiments, compounds described herein are oligomeric compounds (including oligomeric compounds that are antisense agents, siRNAs, etc., or portions thereof) having at least one modified internucleoside linking group having any of the above formulas.
Assaying Modulation of Expression
Modulation of SLC6A19 and/or B°AT1 expression can be assayed in a variety of ways known in the art. SLC6A19 mRNA levels can be quantitated by, e.g., Northern blot analysis, competitive polymerase chain reaction (PCR), or real-time PCR. RNA analysis can be performed on total cellular RNA or poly(A)+mRNA by methods known in the art. Methods of RNA isolation are taught in, for example, Ausubel, F. M. et al., Current Protocols in Molecular Biology, Volume
1, pp. 4.1.1-4.2.9 and 4.5.1-4.5.3, John Wiley & Sons, Inc., 1993.
Northern blot analysis is routine in the art and is taught in, for example, Ausubel, F. M. et al., Current Protocols in Molecular Biology, Volume 1, pp. 4.2.1-4.2.9, John Wiley & Sons, Inc., 1996. Real-time quantitative (PCR) can be conveniently accomplished using the commercially available ABI PRISM™ 7700 Sequence Detection System, available from PE-Applied Biosystems, Foster City, Calif, and used according to manufacturer's instructions. The method of analysis of modulation of RNA levels is not a limitation of the instant invention.
Levels of a B°AT1 protein encoded by SLC6A19 mRNA or DNA can be quantitated in a variety of ways well known in the art, such as immunoprecipitation, Western blot analysis (immunoblotting), ELISA or fluorescence-activated cell sorting (FACS). Antibodies directed to a B°AT1 protein encoded by SLC6A19 can be identified and obtained from a variety of sources, such as the MSRS catalog of antibodies (Aerie Corporation, Birmingham, Mich.), or can be prepared via conventional antibody generation methods. Methods for preparation of polyclonal antisera are taught in, for example, Ausubel, F. M. et al., Current Protocols in Molecular Biology, Volume 2, pp. 11.12.1-11.12.9, John Wiley & Sons, Inc., 1997. Preparation of monoclonal antibodies is taught in, for example, Ausubel, F. M. et al., Current Protocols in Molecular Biology, Volume 2, pp. 11.4.1-11.11.5, John Wiley & Sons, Inc., 1997.
Immunoprecipitation methods are standard in the art and can be found at, for example, Ausubel, F. M. et al., Current Protocols in Molecular Biology, Volume 2, pp. 10.16.1-10.16.11, John Wiley & Sons, Inc., 1998. Western blot (immunoblot) analysis is standard in the art and can be found at, for example, Ausubel, F. M. et al., Current Protocols in Molecular Biology, Volume
2, pp. 10.8.1-10.8.21, John Wiley & Sons, Inc., 1997. Active Target Segments
The locations on the target nucleic acid (i.e., SLC6A19 mRNA) defined by having one or more active modulatory RNA compounds targeted thereto are referred to as “active target segments.” There may be substantial variation in activity (e.g., as defined by percent inhibition) of the modulatory RNA compounds within an active target segment. Active modulatory RNA compounds are those that are determined to modulate the expression of their target RNA. In some embodiments, active modulatory RNA compounds are inhibitory, optionally inhibiting expression of their target RNA at least about 50%, optionally at least about 70% and optionally at least about 80%, or more. In some embodiments, the level of inhibition required to define an active inhibitory RNA compound is defined based on the results from a screen used to define the active target segments. Those skilled in the art understand that the percent inhibition by an inhibitory RNA compound on a target mRNA will vary between assays due to factors relating to assay conditions.
Hybridization
As used herein, “hybridization” means the pairing of complementary strands of antisense compounds to their target sequence and/or the pairing of complementary strands of doublestranded nucleic acid molecules with one another. While not limited to a particular mechanism, the most common mechanism of pairing involves hydrogen bonding, which may be Watson-Crick, Hoogsteen or reversed Hoogsteen hydrogen bonding, between complementary nucleoside or nucleotide bases (nucleobases). For example, the natural base adenine is complementary to the natural nucleobases thymidine and uracil which pair through the formation of hydrogen bonds. The natural base guanine is complementary to the natural base 5-methyl cytosine and the artificial base known as a G-clamp. Hybridization can occur under varying circumstances.
A modulatory RNA compound is specifically hybridizable when there is a sufficient degree of complementarity to avoid non-specific binding of the modulatory RNA compound to non-target nucleic acid sequences under conditions in which specific binding is desired, i.e., under physiological conditions in the case of in vivo assays or therapeutic treatment, and under conditions in which assays are performed in the case of in vitro assays.
As used herein, “stringent hybridization conditions” or “stringent conditions” refers to conditions under which a modulatory RNA compound will hybridize to its target sequence, but to a minimal number of other sequences. Stringent conditions are sequence-dependent and will be different in different circumstances, and “stringent conditions” under which modulatory RNA compounds hybridize to a target sequence are determined by the nature and composition of the modulatory RNA compounds and the assays in which they are being investigated.
Complementarity
“Complementarity,” as used herein, refers to the capacity for precise pairing between two nucleobases on either two oligomeric compound strands or an antisense compound with its target nucleic acid. For example, if a nucleobase at a certain position of an antisense compound is capable of hydrogen bonding with a nucleobase at a certain position of a target nucleic acid, then the position of hydrogen bonding between the oligonucleotide and the target nucleic acid is considered to be a complementary position. The antisense compound and the further DNA or RNA are complementary to each other when a sufficient number of complementary positions in each molecule are occupied by nucleobases which can hydrogen bond with each other. Thus, “specifically hybridizable” and “complementary” are terms which are used to indicate a sufficient degree of precise pairing or complementarity over a sufficient number of nucleobases such that stable and specific binding occurs between the antisense compound and a target nucleic acid, or between respective sense and antisense strands (or subsequences thereof) of a double-stranded nucleic acid molecule.
Identity
Double-stranded RNA compounds, antisense compounds, or portions thereof, may have a defined percent identity to a target sequence and/or complement thereof. As used herein, a sequence is identical to the sequence disclosed herein if it has the same nucleobase pairing ability. For example, a RNA which contains uracil in place of thymidine in the disclosed sequences would be considered identical as they both pair with adenine. This identity may be over the entire length of the oligomeric compound, or in a portion of a given strand, e.g., in a portion of an antisense compound (e.g., nucleobases 1-20 of a 27-mer may be compared to a 20-mer to determine percent identity of the 27-mer across residues 1-20 to the comparator 20-mer (or to a defined 20 nucleotide sequence within a target nucleic acid molecule (e.g., SLC6A19 mRNA)). It is understood by those skilled in the art that a modulatory RNA compound (e.g., an siRNA compound or an antisense compound) need not have an identical sequence to those described herein to function similarly to the modulatory RNA compound described herein. Shortened or extended versions - on one or both strands where an siRNA compound - of modulatory RNA compounds taught herein, or nonidentical versions of the modulatory RNA compounds taught herein fall within the scope of this disclosure. Non-identical versions are those wherein each base does not have the same pairing activity as the modulatory RNA compounds disclosed herein. Bases do not have the same pairing activity by being shorter or having at least one abasic site. Alternatively, a non-identical version can include at least one base replaced with a different base with different pairing activity (e.g., G can be replaced by C, A, or T). Percent identity is calculated according to the number of bases that have identical base pairing corresponding to the reference sequence (be it a target sequence, a complementary strand of a dsRNA, an antisense oligonucleotide and/or antisense strand sequence, etc.) to which it is being compared. The non-identical bases may be adjacent to each other, dispersed throughout the oligonucleotide, or both.
For example, a 16-mer having the same sequence as nucleobases 2-17 of a 20-mer is 80% identical to the 20-mer across the entire length of the 20-mer. Alternatively, a 20-mer containing four nucleobases not identical to the 20-mer is also 80% identical to the 20-mer. A 14-mer having the same sequence as nucleobases 1-14 of an 18-mer is 78% identical to the 18-mer. Such calculations are well within the ability of those skilled in the art.
The percent identity is based on the percent of nucleobases in the original sequence present in a portion of the modified sequence. Therefore, a 30 nucleobase antisense compound comprising the full sequence of the complement of a 20 nucleobase active target segment would have a portion of 100% identity with the complement of the 20 nucleobase active target segment, while further comprising an additional 10 nucleobase portion. The complement of an active target segment may constitute a single portion. In certain embodiments, the oligonucleotides are at least about 80%, optionally at least about 85%, even more preferably at least about 90%, most preferably at least 95% identical to at least a portion of the complement of the active target segments presented herein.
It is well known by those skilled in the art that it is possible to increase or decrease the length of a modulatory RNA compound and/or introduce mismatch bases (with either target sequence, complementary strand(s) of a double- stranded RNA, or both) without eliminating activity. For example, in Woolf et al. (Proc. Natl. Acad. Sci. USA 89:7305-7309, 1992, incorporated herein by reference), a series of ASOs 13-25 nucleobases in length were tested for their ability to induce cleavage of a target RNA. ASOs 25 nucleobases in length with 8 or 11 mismatch bases near the ends of the ASOs were able to direct specific cleavage of the target mRNA, albeit to a lesser extent than the ASOs that contained no mismatches. Modulatory RNA compounds having a contiguous nucleobase composition that is shorter or longer (independently on either or both strands for double-stranded nucleic acid compounds) or that comprises mismatches are contemplated in the instant disclosure so long as the modulatory RNA activity towards target sequence (here, SLC6A19) is maintained.
Therapeutics
Modulatory nucleic acid agents of the instant disclosure can be used to modulate the expression of SLC6A19 and/or B°AT1 in an animal, such as a human. In one non-limiting embodiment, the methods comprise the step of administering to said animal in need of therapy for a disease or condition associated with SLC6A19 and/or B°AT1 an effective amount of a modulatory RNA compound that inhibits expression of SLC6A19 and/or B°AT1. A disease or condition associated with SLC6A19 and/or B°AT1 includes, but is not limited to, a glomerular disorder, a renal tubular disorder, other renal disorders, an inborn error of metabolism, a systemic metabolic disorder, a disorder of the thyroid, a disorder of the parathyroid, a disorder of the inner ear, a neurological disorder, or a viral infection, or combinations thereof. Phenylketonuria (PKU) and urea cycle disorders are specifically described in additional detail elsewhere herein.
In one embodiment, the modulatory RNA compounds effectively inhibit the levels or function of SLC6A19 mRNA. Because reduction in SLC6A19 mRNA levels can lead to alteration in B°AT1 protein products of expression as well, such resultant alterations can also be measured. Modulatory RNA compounds that effectively inhibit the level or function of SLC6A19 RNA or B°AT1 protein products of expression are considered active inhibitory RNA compounds. In one embodiment, the modulatory RNA compounds of the instant disclosure inhibit the expression of SLC6A19 causing a reduction of RNA by at least 10%, by at least 20%, by at least 25%, by at least 30%, by at least 40%, by at least 50%, by at least 60%, by at least 70%, by at least 75%, by at least 80%, by at least 85%, by at least 90%, by at least 95%, by at least 98%, by at least 99%, or by 100%.
For example, the reduction of the expression of SLC6A19 and/or B°AT1 can be measured in a bodily fluid, tissue or organ of the animal. Methods of obtaining samples for analysis, such as body fluids (e.g., blood, plasma, urine, saliva, etc.), tissues (e.g., biopsy), or organs, and methods of preparation of the samples to allow for analysis are well known to those skilled in the art. Methods for analysis of RNA and protein levels are discussed above and are well known to those skilled in the art. The effects of treatment can be assessed by measuring biomarkers associated with the SLC6A19 and/or B°AT1 expression in the aforementioned fluids, tissues or organs, collected from an animal contacted with one or more compounds, by routine clinical methods known in the art.
The modulatory nucleic acid agents of the instant disclosure can be utilized in pharmaceutical compositions by adding an effective amount of a compound to a suitable pharmaceutically acceptable diluent or carrier. Acceptable carriers and diluents are well known to those skilled in the art. Selection of a diluent or carrier is based on a number of factors, including, but not limited to, the solubility of the compound and the desired route of administration. Such considerations are well understood by those skilled in the art. In one aspect, the compounds inhibit the expression of SLC6A19 and/or B°AT1. The modulatory nucleic acid agents can also be used in the manufacture of a medicament for the treatment of diseases and conditions related to SLC6A19 and/or B° ATI expression.
Methods whereby bodily fluids, organs or tissues are contacted with an effective amount of one or more of the modulatory RNA compounds or compositions are also contemplated. Bodily fluids, organs or tissues can be contacted with one or more of the compounds disclosed herein resulting in modulation of SLC6A19 and/or B°AT1 expression in the cells of bodily fluids, organs or tissues.
Thus, provided herein is the use of an isolated single- or double-stranded modulatory nucleic acid agent targeted to SLC6A19 and/or B°AT1 in the manufacture of a medicament for the treatment of a disease or disorder by means of the method described above. In certain embodiments, the modulatory nucleic acid agent is a single-stranded antisense compound. In other embodiments, the modulatory nucleic acid agent is a double-stranded inhibitory RNA compound.
In some embodiments, a modulatory nucleic acid agent of the disclosure, e.g., an oligonucleotide, is characterized in that when a modulatory nucleic acid agent of the disclosure, a composition comprising a modulatory nucleic acid agent of the disclosure, or a conjugate agent comprising a modulatory nucleic acid agent of the disclosure is delivered to a cell, tissue, or organism expressing a target, reduced expression and/or activity of a target is observed as compared to a cell, tissue or organism which has not been delivered a modulatory nucleic acid agent, a composition comprising a modulatory nucleic acid agent, or a conjugate agent comprising a modulatory nucleic acid agent of the disclosure.
In some embodiments, a modulatory nucleic acid agent of the disclosure, e.g., an oligonucleotide, is characterized in that when a modulatory nucleic acid agent of the disclosure, a composition comprising a modulatory nucleic acid agent of the disclosure, or a conjugate agent comprising a modulatory nucleic acid agent of the disclosure is delivered to a cell, tissue, or organism expressing a target, reduced expression and/or activity of a target is observed as compared to a cell, tissue or organism which does not express a target (e.g., which has no detectable expression of a target).
In some embodiments, a modulatory nucleic acid agent of the disclosure, e.g., an oligonucleotide, is characterized in that when a modulatory nucleic acid agent of the disclosure, a composition comprising a modulatory nucleic acid agent of the disclosure, or a conjugate agent comprising a modulatory nucleic acid agent of the disclosure is delivered to a cell, tissue, or organism expressing a target, altered expression and/or activity of a target is observed relative to that observed with an appropriate reference agent known to have a specified impact on the target. In some embodiments, expression and/or activity of a target is altered in a manner and/or to an extent reasonably comparable to, or otherwise determined relative to, that observed with an appropriate reference agent known to have a specified impact on the target. In some embodiments, a reference agent may be a positive control reference agent. In some embodiments, a reference may be a negative control reference agent.
In some embodiments, a modulatory nucleic acid agent of the disclosure, e.g., an oligonucleotide, dsRNA, etc., is characterized in that when delivered to a cell, tissue, or organism expressing a target, expression and/or activity of a target is modulated, e.g., reduced, as compared to a cell, tissue, or organism, which has not been delivered a modulatory nucleic acid agent of the disclosure.
Without wishing to be bound by theory, it is believed that in some embodiments, a targeting moiety, e.g., a peptide as disclosed herein, can be conjugated to a modulatory nucleic acid agent of the disclosure, e.g., an oligonucleotide, dsRNA, etc.
Characterization of Conjugate Agents
In some embodiments, conjugate agent(s) as provided and/or utilized in accordance with the present disclosure are characterized in that, for example, when they are provided to a relevant system (e.g., comprising one or more cell(s), tissue(s), organ(s), or organism(s)) they impact expression and/or activity of one or more targets or form(s) thereof.
In some embodiments, a relevant agent is characterized by its impact on RNA (e.g., mRNA) and/or protein (e.g., encoded by an mRNA) targeted by its nucleic acid payload. In some such embodiments, such impact is assessed in vivo (i.e., in an organism). Alternatively or additionally, in some such embodiments, impact is assessed in vitro (e.g., in cell lines).
In some embodiments, conjugate agent(s) as described and/or utilized in accordance with the present disclosure are characterized relative to an unconjugated nucleic acid agent (as payload). In some embodiments, when assessed under comparable conditions, significantly greater impact is observed when an appropriate in vivo or in vitro system is contacted with a conjugate agent described herein than is observed when the system is contacted with an unconjugated nucleic acid agent under otherwise comparable conditions.
Pharmaceutical Compositions
The present disclosure, among other things, provides pharmaceutical compositions that comprise or otherwise deliver a modulatory nucleic acid agent; typically, such pharmaceutical compositions comprise an active agent (e g., an ASO or iRNA agent or a composition comprising the same) and one or more pharmaceutically or physiologically acceptable carriers, diluents, or excipients.
The pharmaceutical compositions containing the iRNA are useful for preventing or treating an SLC6A19-associated disorder, e.g., metabolic disorders, including nephropathy and various other kidney diseases or conditions. Such pharmaceutical compositions are formulated based on the mode of delivery'. One example is compositions that are formulated for systemic administration via parenteral delivery', e.g., by subcutaneous (SC), intramuscular (IM), or intravenous (IV) delivery. The pharmaceutical compositions of the present disclosure may be administered in dosages sufficient to inhibit expression of a SLC6A19 gene.
In some embodiments, the pharmaceutical compositions of the present disclosure are sterile. In another embodiment, the pharmaceutical compositions of the present disclosure are pyrogen free.
The pharmaceutical compositions of the present disclosure may be administered in dosages sufficient to inhibit expression of a SLC6A19 gene. In general, a suitable dose of an ASO or iRNA agent of the present disclosure will be in the range of about 0.001 to about 200.0 milligrams per kilogram body weight of the recipient per day, generally in the range of about 1 to 50 mg per kilogram body weight per day. Typically, a suitable dose of an ASO or iRNA agent of the present disclosure will be in the range of about 0.1 mg/kg to about 5 0 mg/kg, optionally about 0.3 mg/kg and about 3.0 mg/kg. A repeat-dose regimen may include administration of a therapeutic amount of ASO or iRNA agent on a regular basis, such as every month, once every' 3-6 months, or once a year. In certain embodiments, the ASO or iRNA agent is administered about once per month to about once per six months.
After an initial treatment regimen, the treatments can be administered on a less frequent basis. Duration of treatment can be determined based on the severity of disease.
In other embodiments, a single dose of the pharmaceutical compositions can be long lasting, such that doses are administered at not more than 1, 2, 3, or 4 month intervals. In some embodiments of the present disclosure, a single dose of the pharmaceutical compositions of the present disclosure is administered about once per month. In other embodiments of the present disclosure, a single dose of the pharmaceutical compositions of the present disclosure is administered quarterly (i.e., about every three months). In other embodiments of the present disclosure, a single dose of the pharmaceutical compositions of the present disclosure is administered twice per year (i.e., about once every' six months).
The skilled artisan will appreciate that certain factors can influence the dosage and timing required to effectively treat a subject, including but not limited to mutations present in the subject, previous treatments, the general health or age of the subject, and other diseases present. Moreover, treatment of a subject with a prophy lacti cal ly or therapeutically effective amount, as appropriate, of a composition can include a single treatment or a series of treatments.
The iRNA can be delivered in a manner to target a particular tissue (e.g., kidney cells).
Pharmaceutical compositions of the instant disclosure include, but are not limited to, solutions, emulsions, and liposome-containing formulations. These compositions can be generated from a variety of components that include, but are not limited to, preformed liquids, selfemulsifying solids, and self-emulsifying semisolids. Formulations include those that target the kidneys.
The pharmaceutical formulations of the instant disclosure, which can conveniently be presented in unit dosage form, can be prepared according to conventional techniques well known in the pharmaceutical industry. Such techniques include the step of bringing into association the active ingredients with the pharmaceutical carrier(s) or excipient(s). In general, the formulations are prepared by uniformly and intimately bringing into association the active ingredients with liquid carriers. In some embodiments, pharmaceutical compositions described herein may comprise buffers including neutral buffered saline or phosphate buffered saline (PBS); carbohydrates, such as glucose, mannose, sucrose, dextrans, or mannitol; proteins, polypeptides, or amino acids (e.g., glycine); antioxidants; chelating agents, such as EDTA or glutathione; adjuvants (e.g., aluminum hydroxide); and preservatives. In some embodiments, a pharmaceutical composition is substantially free of contaminants, e.g., there are no detectable levels of a contaminant (e.g., an endotoxin).
In some embodiments, pharmaceutical compositions described herein may be administered in a manner appropriate to the disease, disorder, or condition to be treated or prevented. In some embodiments, quantity and/or frequency of administration may be determined by such factors as condition of a patient, and/or type and/or severity of a patient’s disease, disorder, or condition, although appropriate dosages may be determined by clinical trials.
In some embodiments, a pharmaceutical composition provided by the present disclosure may be in a form such as, for example, liquid, semi-solid and solid dosage forms, such as liquid solutions (e.g., injectable and infusible solutions), dispersions or suspensions, liposomes, and suppositories. Typically, pharmaceutical compositions that comprise or deliver antibody agents are injectable or infusible solutions; in some such embodiments, such compositions can be formulated for administration intravenously, subcutaneously, intradermally, intratumorally, intranodally, intramedullary, intramuscularly, transarterially, sublingually, intranasally, topically or intraperitoneally. In some embodiments, provided pharmaceutical compositions are formulated for intravenous administration. In some embodiments, provided pharmaceutical compositions are formulated for subcutaneous administration.
Pharmaceutical compositions described herein can be formulated for administration by using infusion techniques that are commonly known in the field (See, e.g., Rosenberg et al., New Eng. J. of Med. 319: 1676, 1988, which is hereby incorporated by reference in its entirety).
In some embodiments, pharmaceutical compositions described herein are administered in combination with (e.g., before, simultaneously, or following) an additional therapy for a symptom, disease or disorder, e.g., a SOC therapy for a symptom, disease or disorder. In some embodiments, pharmaceutical compositions described herein may be administered before or following surgery. In some embodiments, a dosage of any aforementioned therapy to be administered to a subject will vary with a disease, disorder, or condition being treated and based on a specific subject. Scaling of dosages for human administration can be performed according to art-accepted practices.
Use of Modulatory Nucleic Acid Agents, Including Conjugate Agents
Cell type for delivery
In some embodiments, a modulatory nucleic acid agent of the instant disclosure - e.g., an ASO, a dsRNA and/or a conjugate agent that includes one or more modulatory nucleic acid agents disclosed herein - is delivered to a cell, e.g., of a tissue, optionally in which a cell surface factor is present.
In some embodiments, a cell is or comprises a cell (e.g., of a tissue) chosen from: immune cells (e.g., bone marrow cells, lymph node cells, thymic cells, peripheral blood mononuclear cells [e.g., myeloid and/or lymphoid cells], erythrocytes, eosinophils, neutrophils, and/or platelets); nervous system cells (e.g., brain tissue, cortex, cerebellum, retinal cells, spinal cord cells, nerve cells, neurons, and/or supporting cells; endothelial cells; muscle (e.g., heart muscle, smooth muscle, and/or skeletal muscle); small intestine cells; colon cells; adipocytes; kidney cells; liver cells; lung cells; splenic cells; stomach cells; esophagus cells; bladder cells; pancreas cells; thyroid cells; salivary gland cells; adrenal gland cells; pituitary gland cells; breast cells; skin cells; ovary cells; uterus cells; placenta cells; prostate cells; or testis cells, or any combination thereof.
In some embodiments, a cell is or comprises a cell (e.g., of a tissue) chosen from: renal cells, thyroid cells, parathyroid cells, cells of the inner ear, or nervous system cells.
In some embodiments, a cell is or comprises a kidney cell, e.g., as described herein. In some embodiments, a cell is or comprises a proximal tubular epithelial cell, a podocyte, or both.
In some embodiments, a cell to which a conjugate disclosed herein is delivered expresses both a cell surface factor (e.g., Megalin and/or Cubilin) and a target of a payload moiety.
In some embodiments, a modulatory nucleic acid agent of the instant disclosure - e.g., an ASO, a dsRNA and/or a conjugate agent that includes one or more modulatory nucleic acid agents disclosed herein - is administered to a subject having a disease or disorder, e.g., as disclosed herein. In some embodiments, a disease or disorder comprises a cell in which a surface cell factor (e.g., Megalin and/or Cubilin) and/or a SLC6A19 target RNA is present. Indications
In some embodiments, a modulatory nucleic acid agent of the instant disclosure - e g., an ASO, a dsRNA and/or a conjugate agent that includes one or more modulatory nucleic acid agents disclosed herein - is used to treat and/or prevent a symptom of, a disease or disorder disclosed herein.
In some embodiments, a disease or disorder to which a modulatory nucleic acid agent disclosed herein is provided, has elevated or aberrant expression of a cell surface factor such as Megalin and/or Cubilin.
In some embodiments, Megalin expression is reported to be enriched in the following tissues and/or cells in particular: renal tissue, thyroid tissue, parathyroid tissue, cells of the inner ear, and nervous system tissue. In some embodiments, Megalin is expressed (e.g., at relatively high level(s)) on surfaces of kidney cells such as proximal tubular epithelial cells and podocytes.
In some embodiments, a disease or disorder is chosen from: a glomerular disorder, a renal tubular disorder, other renal disorders, an inborn error of metabolism, a systemic metabolic disorder, a disorder of the thyroid, a disorder of the parathyroid, a disorder of the inner ear, a neurological disorder, or a viral infection, or any combination thereof.
In some embodiments, a disease or disorder is or comprises a glomerular disorder. In some embodiments, a glomerular disorder is chosen from: Lupus nephritis, Goodpasture syndrome, IgA nephropathy, Alport syndrome, glomerulosclerosis, diabetic nephropathy, focal segmental glomerulosclerosis, membranous nephropathy, minimal change disease, ApoLl nephropathy, post-infection glomerulonephritis, membranoproliferative glomerulonephritis, mesangioproliferative glomerulonephritis, nephrotic syndrome, nephritic syndrome, Anti-LRP2 nephropathy, C3 glomerulopathy, or any combination thereof.
In some embodiments, a disease or disorder is or comprises a renal tubular disorder. In some embodiments, a renal tubular disorder is chosen from: Fanconi syndrome, cystinuria, Lowe syndrome, Dent syndrome, Light Chain Proximal Tubulopathy, Gitelman syndrome, renal tubular acidosis, nephrogenic diabetes insipidus, Bartter syndrome, Liddle syndrome, hereditary aminoaciduria, hereditary salt wasting disorders, hereditary phosphate wasting disorders, porphyria associated renal disease, nephropathic cystinosis, autosomal dominant tubulointerstitial kidney disease, or any combination thereof. In some embodiments, a disease or disorder is or comprises other renal disorders. In some embodiments, other renal disoders are chosen from: ADPKD, ARPKD, Nephronophthisis, Chronic Kidney Disease, nephrolithiasis, acute kidney injury, Alagille syndrome, cardiorenal syndrome, renal cell carcinoma, renal osteodystrophy, or any combination thereof.
In some embodiments, a disease or disorder is or comprises an inborn error of metabolism. In some embodiments, an inborn error of metabolism is chosen from: phenylketonuria, urea cycle disorder, maple syrup urine disease, galactosemia, hereditary tyrosinemia, glutamic academia, isovaleric acidemia, very long/long/medium/short chain acyl-CoA dehydrogenase deficiency, methylmalonic academia, primary hyperoxaluria, propionic academia, porphyria, Wilson disease, Pyruvate dehydrogenase deficiency, homocystinuria, hereditary fructose intolerance, nonketotic hyperglycinemia, or any combination thereof.
In some embodiments, a disease or disorder is or comprises a systemic metabolic disorder. In some embodiments, a systemic metabolic disorder is chosen from: diabetes, obesity, hypertension, gout, polyneuropathy, hypoglycemia, vitamin B deficiencies, liver cirrhosis, coronary heart disease, stroke, lipodystrophy, or any combination thereof.
In some embodiments, a disease or disorder is or comprises a disorder of the thyroid. In some embodiments, a disorder of the thyroid is chosen from: Hashimoto disease, Graves' disease, hypothyroidism, hyperthyroidism, goiter, thyroid nodules, thyroiditis, thyroid cancer, thyrotropinoma, thyroid hormone resistance, MCT8 deficiency, Riedel’s thyroiditis, Pendred syndrome, sarcoidosis, McCune- Albright syndrome, familial dysalbuminemic hyperthyroxinemia, thyroxin binding globulin (TBG) deficiency, or any combination thereof.
In some embodiments, a disease or disorder is or comprises a disorder of the parathyroid. In some embodiments, a disorder of the parathyroid is chosen from: hyperparathyroidism/hypercalcemia, hypoparathyroidism/hypocalcemia, nephrolithiasis (kidney stone), pancreatitis, granulomatous disease, Addison’s disease, pernicious anemia (many of these belong to hyperparathyroidism and hypoparathyroidism).
In some embodiments, a disease or disorder is or comprises a disorder of the inner ear. In some embodiments, a disorder of the inner ear is chosen from: inherited sensorineural hearing loss, vestibular neuritis, Meniere’s syndrome, benign paroxysmal positional vertigo, tinnitus, age related hearing loss, bilateral vestibular loss, perilymphatic fistula (PLF), superior semicircular canal dehiscence syndrome (SCD), drug-induced ototoxicity, herpes zoster oticus, purulent labyrinthitis, vestibular schwannoma.
In some embodiments, a disease or disorder is or comprises a neurological disorder, e.g., a neurodegenerative disease. In some embodiments, a neurological disorder is chosen from: Alzheimer's disease, Parkinson's disease, Huntington's disease, A.L.S., multiple sclerosis, neuro- AIDS, brain cancer, stroke, brain injury, spinal cord injury, autism, lysosomal storage disorders, fragile X syndrome, inherited mental retardation, inherited ataxias, blindness, paralysis, stroke, traumatic brain injury and spinal cord injury, and lysosomal storage diseases such as MPS I, MPS II, MPS III A, MPS III B, Metachromatic Leukodystrophy, Gaucher, Krabbe, Pompe, CLN2, Niemann-Pick and Tay-Sachs disease, or any combination thereof.
In some embodiments, a disease or disorder is or comprises a viral infection. In some embodiments, a viral infection comprises a polyoma virus (e g., BK virusj-mediated nephropathy.
In certain embodiments, the instant disclosure provides a therapeutic agent for treatment of phenylketonuria (PKU) or a urea cycle disorder.
Phenylketonuria (PKU)
Phenylketonuria (PKU) is an inborn error of metabolism caused by mutations in phenylalanine hydroxylase (PAH), the enzyme responsible for metabolizing phenylalanine. PKU is an autosomal recessive metabolic disorder in which phenylalanine is not properly metabolized and results in abnormally high levels of plasma phenylalanine. People who have PKU have abnormally high blood levels of phenylalanine, which if untreated can lead to irreversible neurological damage resulting in a spectrum of complications such as intellectual disabilities, seizures, neurodevelopmental and behavioral disorders. PKU is difficult to treat because blood levels of phenylalanine are directly related to diet. Patients must adhere to a life-long and strict diet that impacts all aspects of patients’ lives. Current standard of care are enzyme co-factor and enzyme substitution therapy; but these therapies are not effective in all patients, and also carry potential risk for adverse events.
The enzyme responsible for metabolizing phenylalanine, and thus maintaining phenylalanine homeostasis is phenylalanine hydroxylase (PAH). Loss-of-function (LOF) mutations at PAH gene at chromosome 12q23.2 are known to cause most forms of PKU. These LOF mutations resulting in PKU can be diagnosed as classical PKU (the most severe form), and “mild PKU” or “hyperphe” a less severe form. In addition to PAH, mutations in other enyzmes that affect phenylalanine metabolism, such as dihydropteridine reductase (DHPR), the enzyme responsible for synthesis of co-factors required for PAH activity, may also result in elevated levels of phenylalanine. In addition to diet, blood amino acid levels, including levels of phenylalanine, are regulated by SLC6A19. SLC6A19 is located in the proximal tubule of the kidney and is responsible for reabsorption of amino acids back into the blood. Certain aspects of the instant disclosure therefore provide targeting of SLC6A19 in the kidney of a subject (e.g., in the proximal tubule of the kidney).
Urea Cycle Disorders
Urea cycle disorders are inborn errors of metabolism resulting from defects in one of the enzymes or transporter molecules involved in the hepatic removal of ammonia from the bloodstream. Removal of ammonia from the bloodstream normally occurs via its conversion to urea, which is then excreted by the kidneys. Consequently, urea cycle disorders lead to an accumulation of ammonia. Ammonia is extremely toxic, particularly to the central nervous system. Newborns with severe mutations in any one of the first four enzymes of the urea cycle can become catastrophically ill within 36 to 48 hours of birth despite appearing normal at birth. It is therefore possible for a newborn to be discharged from the hospital before signs of urea cycle disorders develop. The newborn may subsequently develop signs that go unrecognized at home. Hyperammonemia is key to the diagnosis of urea cycle disorders and treatment should not be delayed while a definitive diagnosis is sought.
The etiology of UCDs is complex since multiple proteins and two subcellular compartments are involved, i.e., the mitochondrial matrix and cytoplasm. The first two enzymatic steps in the urea cycle take place in the mitochondrial matrix. In step 1, carbamoylphosphate synthetase I (CPS1) ligates ammonia with bicarbonate forming carbamoyl phosphate which enters the urea cycle. Stressors such as sepsis can easily initiate defects in CPS1, which result in the most severe form of UCDs and hyperammonemia. It should be noted that N-acetylglutamate is an allosteric activator of CPS1. N-acetylglutamate is formed from acetyl-CoA and glutamate in a reaction catalyzed by N-acetylglutamate synthase (NAG), a mitochondrial enzyme. Defects in NAG, although very rare, mimic CPS1 UCDs and result in hyperammonemia.
In step 2, the liver enzyme, ornithine transcarb amylase (OTC) catalyzes the reaction of carbamoyl phosphate with ornithine to form citrulline. The OTC gene (SLC25A15) is located on the X chromosome, and OTC defects are inherited in an X-linked fashion. Defects in OTC are the most common cause of UCDs. OTC deficiency in males results in a severe form of UCD. In females, the severity is variable ranging from asymptomatic to severe depending upon the random nature of X chromosome inactivation. The ornithine needed for OTC must be transported into the mitochondrial matrix, and this task is accomplished by the mitochondrial ornithine translocase (0RNT1) protein. Defective 0RNT1, although very rare, can result in hyperammonemia which often manifests itself later in life.
Step 3 and all remaining enzymatic steps take place in the cytoplasm. In step 3, argininosuccinate synthase 1 (ASS1) catalyzes the formation of argininosuccinate from citrulline and aspartic acid. Defects in ASS1 result in both citrullinemia and hyperammonemia. The second mitochondrial transporter, citrin, contributes to the cytoplasmic pool of aspartic acid by transporting aspartic out of the mitochondria and into the cytoplasm where it is needed for the urea cycle. Citrin also transports glutamate from the cytoplasm into the mitochondria. Gene SLC25A13 codes for citrin and defects in this gene can result in both citrullinemia and hyperammonemia.
Step 4 of the urea cycle involves the cleavage of argininosuccinate to form fumaric acid and arginine, and this reaction is catalyzed by argininosuccinate lyase (ASL). Defects in ASL can cause a severe UCD in neonates as well as an adult-onset form. In contrast to the adult-onset form in which hyperammonemia is episodic, the neonatal form results in persistent hyperammonemia if left untreated. Elevated plasma levels of both citrulline and argininosuccinate are characteristic of ASL deficiency.
In the last step of the urea cycle, the arginine from step 4 is cleaved into urea and ornithine by the arginase enzyme which is coded for by the ARG1 gene. Defects in arginase can result in both argininemia and hyperammonemia. The signs and symptoms of arginase deficiency usually appear by 3 years of age and without a rapid onset of hyperammonemia.
Overall, the incidence of UCDs has recently been determined to be 1 in 35,000 births. Most State Screening Laboratories currently detect only 2 of all the conditions that cause UCDs. About two-thirds of all USDs are due to mutations in OTC, one-fifth due to ASS1, and one-tenth due to ASL.
While OTC deficiency (OTCD) is inherited in an X-linked manner, all other UCDs follow an autosomal recessive pattern. Moreover, there is evidence that even asymptomatic female carriers of OTCD can develop cognitive defects as a result of episodic hyperammonemia. Newborns with UCDs typically appear normal at birth and shortly after that can present with nonspecific signs and symptoms similar to many IEMS or an infection, i.e., lethargy, poor appetite, vomiting, and irritability. IEMs, in general, should be considered in the differential diagnosis of any infant with such signs. Except for arginase deficiency, most infants with severe UCDs rapidly develop cerebral edema, and subsequent neurological problems, resulting from acute hyperammonemia. Unless treated, hyperammonemia from severe UCDs can progress to coma and death. In cases of mild UCDs, hyperammonemia can result from a variety of stressors such as illness and surgery.
Accordingly, several enzyme deficiencies have been noted as contributing to urea cycle disorders. These include:
(a) N-acetyl glutamate synthetase deficiency, which causes neurologic deterioration due to elevated blood ammonia.
(b) Carbamyl phosphate synthetase (CPS) deficiency, which is often a lethal disease with death occurring in the first weeks of life.
(c) Ornithine transcarbamylase deficiency (OTID), which is inherited in a sexdinked dominant manner and is generally fatal in the newborn male.
(d) Argininosuccinic acid synthetase, which typically results in severe neurological impairment leading to mental retardation or death.
(e) Argininosuccinate lyase deficiencies, which result in clinical manifestations of retardation, spasticity, and episodes of convulsions. Plasma ammonia level are greatly elevated.
(f) Arginase deficiency, which results in severe neurologic deterioration over time. Plasma arginine concentrations are greatly elevated.
All of these disorders respond to some degree to restriction of protein intake. Acute episodes are usually precipitated by an increased protein intake, an infection or any incident that leads to a negative nitrogen balance. These acute episodes are best handled by the omission of protein and intravenous fluid therapy. Prolonged treatment of children by limiting protein intake to the minimal requirement together with adequate energy intake and supplements of essential amino acids has resulted in control of the plasma ammonia levels and alleviation of the clinical symptoms. No single panacea is available and nutritional support is specific to the individual disorder. A need therefore exists for targeted molecular treatments that are effective for managem ent/ cure ofUCDs.
Delivery of a Modulatory Nucleic Acid Agent
The delivery' of a modulatory' nucleic acid agent of the present disclosure to a cell e.g., a cell within a subject, such as a human subject (e.g., a subject in need thereof, such as a subject susceptible to or diagnosed with an SLC6A19-associated disorder, e.g., metabolic disorders, including nephropathy and various other kidney diseases or conditions) can be achieved in a number of different ways. For example, delivery may be performed by contacting a cell with an ASO or iRNA agent of the present disclosure either in vitro or in vivo. In vivo delivery may also be performed directly by administering a composition comprising an ASO or iRNA agent, e.g., a dsRNA, to a subject. Alternatively, in vivo delivery'’ may be performed indirectly by administering one or more vectors that encode and direct the expression of the ASO or iRNA agent.
In general, any method of delivering a nucleic acid molecule (in vitro or in vivo) can be adapted for use with an ASO or iRNA agent of the present disclosure (see e.g., Akhtar S. and Julian RL. (1992) Trends Cell. Biol.2(5): 139-144 and WO94/02595, which are incorporated herein by reference in their entireties). For in vivo delivery', factors to consider in order to deliver an ASO or iRNA molecule include, for example, biological stability of the delivered molecule, prevention of non-specific effects, and accumulation of the delivered molecule in the target tissue. RNA interference has also shown success with local delivery' to the CNS by direct injection (Dorn, G., et al. (2004) Nucleic Acids 32:e49; Tan, PH., et al (2005) Gene Ther.12:59-66; Makimura, H., et al (2002) BMC Neurosci.3:18; Shishkina, GT., et al (2004) Neuroscience 129:521-528, Thakker, ER., et al (2004) Proc. Natl. Acad. Sei. U.S.A. 101 : 17270-17275; Akaneya,Y., et al (2005) J. Neurophysiol.93: 594-602). Modification of the RNA or the pharmaceutical carrier can also permit targeting of the ASO or iRNzX agent to the target tissue and avoid undesirable off-target effects. ASO or iRNA agent molecules can be modified by chemical conjugation to lipophilic groups such as cholesterol to enhance cellular uptake and prevent degradation. For example, an .ASO or iRNA agent directed against ApoB conjugated to a lipophilic cholesterol moiety was injected systemical ly into mice and resulted in knockdown of apoB rn RNA in both the liver and jejunum (Soutschek, J., et al (2004) Nature 432: 173-178). In an alternative embodiment, the ASO or iRNA agent can be delivered using drug delivery systems such as a nanoparticle, a dendrimer, a polymer, liposomes, or a cationic delivery system. Positively charged cationic deliver}- systems facilitate binding of an ASO or iRNA agent molecule (negatively charged) and also enhance interactions at the negatively charged cell membrane to permit efficient uptake of an ASO or iRNA agent by the cell. Cationic lipids, dendrimers, or polymers can either be bound to an ASO or iRNA agent, or induced to form a vesicle or micelle (see e.g , Kim SH, et al (2008) Journal of Controlled Release 129(2): 107-1 16) that encases an ASO or iRNA agent. The formation of vesicles or micelles further prevents degradation of the ASO or iRNA agent when administered systemically. Methods for making and administering cationic- ASO or iRNA agent complexes are well within the abilities of one skilled in the art (see e.g., Sorensen, DR, et al (2003) J. Mol. Biol 327:761-766; Verma, UN, et al (2003) Clin. Cancer Res.9: 1291 -1300; Arnold, AS et al (2007) J. Hypertens.25: 197-205, which are incorporated herein by reference in their entirety). Some non-limiting examples of drug delivery- systems useful for systemic delivery of ASOs or iRNA agents include DOTAP (Sorensen, DR., et al (2003), supra; Verma, UN, et al (2003), supra), "solid nucleic acid lipid particles" (Zimmermann, TS, et al (2006) Nature 441 : 111-114), cardiolipin (Chien, PY, et al (2005) Cancer Gene Ther.12:321-328; Pal, A, et al (2005) Int J. Oncol.26: 1087-1091), polyethyleneimine (Bonnet ME, et al (2008) Pharm. Res. Aug 16 Epub ahead of print; Aigner, A. (2006) J. Biomed. Biotechnol.71659), Arg-Gly-Asp (RGD) peptides (Liu, S. (2006) Mol. Pharm. 3:472-487), and polyamidoamines (Tomalia, DA, et al (2007) Biochem. Soc. Trans.35:61-67; Yoo, H , et al (1999) Pharm Res 16: 1799- 1804). In some embodiments, an ASO or iRNA agent forms a complex with cyclodextrin for systemic administration. Methods for administration and pharmaceutical compositions of ASOs and iRNA agents and cyclodextrins can be found in U.S. Patent No.7, 427, 605, which is herein incorporated by reference in its entirety.
Vector-Encoded Modulatory Nucleic Acids
In embodiments, modulatory nucleic acid agents targeting the SLC6A19 gene can be expressed from transcription units inserted into DNA or RNA vectors (see, e.g., Couture, A, et al., TIG. (1996), 12:5-10; Skillem, A, et al., International PCT Publication No WO 00/22113, Conrad, International PCT Publication No. WO 00/22114, and Conrad, U S Patent No.6,054, 299) Expression can be transient (on the order of hours to weeks) or sustained (weeks to months or longer), depending upon the specific construct used and the target tissue or cell type. These transgenes can be introduced as a linear construct, a circular plasmid, or a viral vector, which can be an integrating or non-integrating vector. The transgene can also be constructed to permit it to be inherited as an extrachromosomal plasmid (Gassmann, et al., Proc. Natl. Acad. Sci USA (1995) 92: 1292).
Viral vector systems which can be utilized with the methods and compositions described herein include, but are not limited to, (a) adenovirus vectors; (b) retrovirus vectors, including but not limited to lenti viral vectors, moloney murine leukemia virus, etc.; (c) adeno- associated virus vectors, (d) herpes simplex virus vectors; (e) SV 40 vectors; (f) polyoma virus vectors; (g) papilloma virus vectors; (h) picomavirus vectors; (i) pox virus vectors such as an orthopox, e.g., vaccinia virus vectors or avipox, e.g. canary pox or fowl pox; and (j) a helper-dependent or gutless adenovirus. Replication-defective viruses can also be advantageous. Different vectors will or will not become incorporated into the cells’ genome. The constructs can include viral sequences for transfection, if desired.
Alternatively, the construct can be incorporated into vectors capable of episomal replication, e.g., EPV and EBV vectors. Constructs for the recombinant expression of an ASO or iRNA agent will generally require regulatory elements, e.g., promoters, enhancers, etc., to ensure the expression of the ASO or iRNA agent in target cells. Other aspects to consider for vectors and constructs are known in the art.
Dosing regimens
Those skilled in the art will be able to determine, according to known methods, the appropriate amount, dose or dosage of a conjugate agent, to administer to a patient, taking into account factors such as age, weight, general health, the route of administration, the nature of the symptom, disease or disorder requiring treatment, and the presence of other medications. For example, various dosing regimens for antibodies are disclosed in Hendrikx J et al. (2017) Oncologist 22(10): 1212-1221, PMID: 28754722, the entire contents of which is hereby incorporated by reference.
In some embodiments, a modulatory nucleic acid agent of the instant disclosure - e.g., an ASO, a dsRNA and/or a conjugate agent that includes one or more modulatory nucleic acid agents disclosed herein - is administered at a fixed dose, i.e. independent of body weight. In some embodiments, a fixed dose reduces interpatient variability, e.g., efficacy and/or PK/PD parameters. In some embodiments, a modulatory nucleic acid agent of the instant disclosure - e.g., an ASO, a dsRNA and/or a conjugate agent that includes one or more modulatory nucleic acid agents disclosed herein - is administered based on body weight, e.g., in a mg/kg dosing.
In some embodiments, a modulatory nucleic acid agent of the instant disclosure - e.g., an ASO, a dsRNA and/or a conjugate agent that includes one or more modulatory nucleic acid agents disclosed herein - is administered at an initial dose. In some embodiments, an initial dose may be followed by one or more subsequent doses. In some embodiments, one or more subsequent dose may be administered daily, weekly, or monthly, or at other intervals in between. In some embodiments, a dosing regimen disclosed herein may be repeated for one or more times.
Methods For Inhibiting SLC6A 19 Expression
The instant disclosure also provides methods of inhibiting expression of a SLC6A19 gene in a cell. The methods include contacting a cell with an ASO or RNAi agent, e.g., a double-stranded RNA agent, in an amount effective to inhibit, expression of SLC6A19 in the cell, thereby inhibiting expression of SLC6A19 in the cell.
Contacting of a cell with an ASO or iRNA agent, e.g., a double stranded RNA agent, may be done in vitro or in vivo. Contacting a cell in vivo with the ASO or iRNA agent includes contacting a cell or group of cells within a subject, e.g., a human subject, with the ASO or iRNA agent. Combinations of in vitro and in vivo methods of contacting a cell are also possible. Contacting a cell may be direct or indirect, as di scussed above. Furthermore, contacting a cell may be accomplished via a targeting ligand, including any ligand described herein or known in the art. In certain embodiments, the targeting ligand is a binder of a cell surface factor (e.g., binder of megalin or cubilin), or any other ligand that directs the ASO or i RNA agent to a site of interest.
The term “inhibiting,” as used herein, is used interchangeably with “reducing,”' “silencing,” “downregulating”, “suppressing”, and other similar terms, and includes any level of inhibition.
The phrase “inhibiting expression of a SLC6A19” is intended to refer to inhibition of expression of any SLC6A19 gene (such as, e.g., a mouse SLC6A19 gene, a rat SLC6A19 gene, a monkey SLC6.A19 gene, or a human SLC6A19 gene) as well as variants or mutants of a SLC6A19 gene. Thus, the SLC6A19 gene may be a wild-type SLC6A19 gene, a mutant SLC6A19 gene, or a transgenic SLC6A19 gene in the context of a genetically manipulated cell, group of cells, or organism. “Inhibiting expression of a SLC6A19 gene” includes any level of inhibition of a SLC6A19 gene, e.g., at least partial suppression of the expression of a SLC6A19 gene. The expression of the SLC6A19 gene may be assessed based on the level, or the change in the level, of any variable associated with SLC6A19 gene expression, e.g., SLC6A19 mRNA level or B°AT1 protein level. This level may be assessed in an individual cell or in a group of cells, including, for example, a sample derived from a subject. It is understood that SLC6A19/B°AT1 is expressed in the kidney, as well as in various other tissues.
Inhibition may be assessed by a decrease in an absolute or relative level of one or more variables that are associated with SLC6A19 expression compared with a control level. The control level may be any type of control level that is utilized in the art, e.g , a pre-dose baseline level, or a level determined from a similar subject, cell, or sample that is untreated or treated with a control (such as, e.g., buffer only control or inactive agent control)
In some embodiments of the methods of the present disclosure, expression of a SLC6A19 gene is inhibited by at least 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, or 95%, or to below' the level of detection of the assay. In certain embodiments, expression of a SLC6A19 gene is inhibited by at least 70%. It is further understood that inhibition of SLC6A19 expression in certain tissues, e.g., in liver, without a significant inhibition of expression in other tissues, e.g , brain, may be desirable.
In certain embodiments, inhibition of expression in vivo is determined by knockdown of the human gene in a rodent expressing the human gene, e g., an AAV-infected mouse expressing the human target gene (i.e., SLC6A19), e.g., when administered as a single dose, e.g., at 3 mg/kg at the nadir of RNA expression. Knockdown of expression of an endogenous gene in a model animal system can also be determined, e.g., after administration of a single dose at, e.g , 3 mg/kg at the nadir of RNA expression. Such systems are useful when the nucleic acid sequence of the human gene and the model animal gene are sufficiently close such that the human iRNA provides effective knockdown of the model animal gene. RNA expression in kidney is determined using art-recognized approaches.
Inhibition of the expression of a SLC6A19 gene may be manifested by a reduction of the amount of mRNA expressed by a first cell or group of cells (such cells may be present, for example, in a sample derived from a subject) in which a SLC6A19 gene is transcribed and which has or have been treated (e g., by contacting the cell or cells with an ASO or iRNA agent of the present disclosure, or by administering an ASO or iRNA agent of the present disclosure to a subject in which the cells are or were present) such that the expression of a SLC6A19 gene is inhibited, as compared to a second cell or group of cells substantially identical to the first cell or group of cells but which has not or have not been so treated (control cell(s) not treated with an ASO or iRNA agent or not treated with an ASO or iRNA agent targeted to SLC6A19).
In certain embodiments, inhibition of the expression of a SLC6A19 gene may be assessed in terms of a reduction of a parameter that is functionally linked to SLC6A19 gene expression, e.g., B°AT1 protein level in kidney cells or in a non-kidney tissue from a subject. SLC6A19 gene silencing may be determined in any cell expressing SLC6A19, either endogenous or heterologous from an expression construct, and by any assay known in the art.
Inhibition of the expression of a B°AT1 protein may be manifested by a reduction in the level of the B°AT1 protein that is expressed by a cell or group of cells or in a subject sample (e.g , the level of protein in a kidney cell or other sample derived from a subject). For the assessment of mRNA suppression, the inhibition of protein expression levels in a treated cell or group of cells may similarly be expressed as a percentage of the level of protein in a control cell or group of cells, or the change in the level of protein in a subject sample, e.g., kidney cells or other cells derived therefrom .
A control cell, a group of cells, or subject sample that may be used to assess the inhibition of the expression of a SLC6A19 gene includes a cell, group of cells, or subject sample that has not yet been contacted with an ASO or RNAi agent of the present disclosure. For example, the control cell, group of cells, or subject sample may be derived from an individual subject (e.g., a human or animal subject) prior to treatment of the subject with an ASO or RNAi agent or an appropriately matched population control.
The level of SLC6A19 mRNA that is expressed by a cell or group of cells may be determined using any method known in the art for assessing mRNA expression. In one embodiment, the level of expression of SLC6A19 in a sample is determined by detecting a transcribed polynucleotide, or portion thereof, e.g., mRNA of the SLC6A19 gene. RNA may be extracted from cells using RNA extraction techniques including, for example, using acid phen ol/guani dine isothiocyanate extraction (RNAzol B; Biogenesis), RNeasyIM RNA preparation kits (Qiagen®) or PAXgene1M (PreAnalytix1M, Switzerland). Typical assay formats utilizing ribonucleic acid hybridization include nuclear run-on assays, RT-PCR, RNase protection assays, northern blotting, in situ hybridization, and microarray analysis.
In some embodiments, the level of expression of SLC6A19 is determined using a nucleic acid probe. The term “probe”, as used herein, refers to any molecule that is capable of selectively binding to a specific SLC6A19. Probes can be synthesized by one of skill in the art, or derived from appropriate biological preparations. Probes may be specifically designed to be labeled. Examples of molecules that can be utilized as probes include, but are not limited to, RNA, DNA, proteins, antibodies, and organic molecules.
Isolated mRNA can be used in hybridization or amplification assays that include, but are not limited to. Southern or northern analyses, polymerase chain reaction (PCR) analyses and probe arrays. One method for the determination of mRNA levels involves contacting the isolated mRNA with a nucleic acid molecule (probe) that can hybridize to SLC6A19 mRNA. In one embodiment, the mRNA is immobilized on a solid surface and contacted with a probe, for example by running the isolated mRNA on an agarose gel and transferring the mRNA from the gel to a membrane, such as nitrocellulose. In an alternative embodiment, the probe(s) are immobilized on a solid surface and the mRNA is contacted with the probe(s), for example, in an Affymetrix® gene chip array. A skilled artisan can readily adapt known mRNA detection methods for use in determining the level of SLC6A19 mRNA.
An alternative method for determining the level of expression of SLC6A19 in a sample involves the process of nucleic acid amplification or reverse transcriptase (to prepare cDNA) of for example mRNA in the sample, e.g., by RT-PCR (the experimental embodiment set forth in Mullis, 1987, U.S. Patent No.4, 683, 202), ligase chain reaction (Barany (1991) Proc. Natl. Acad. Sci. USA 88: 189-193), self sustained sequence replication (Guatelli et al (1990) Proc. Natl. Acad. Sci. USA 87:1874-1878), transcriptional amplification system (Kwoh et al. (1989) Proc. Natl. Acad. Sci. USA 86: 1173-1177), Q-Beta Replicase (Lizardi et al. (1988) Bio/Technology 6: 1197), rolling circle replication (Lizardi et al., U.S Patent No.5, 854, 033) or any other nucleic acid amplification method, followed by the detection of the amplified molecules using techniques well known to those of skill in the art These detection schemes are especially useful for the detection of nucleic acid molecules if such molecules are present in very low7 numbers. In particular aspects of the present disclosure, the level of expression of SLC6A19 is determined by quantitative Anorogenic RT-PCR (i.e., the TaqMan1 M System). In certain embodiments, expression level is determined by the method provided in the Examples using, e.g., a lOnM siRNA concentration, in the species matched cell line.
The expression levels of SLC6AJ 9 mRNA may be monitored using a membrane blot (such as used in hybridization analysis such as northern, Southern, dot, and the like), or microwells, sample tubes, gels, beads or fibers (or any solid support comprising bound nucleic acids). See U S. Patent Nos.5, 770, 722, 5,874,219, 5,744,305, 5,677,195 and 5,445,934, which are incorporated herein by reference. The determination of SLC6A19 expression level may also comprise using nucleic acid probes in solution.
In certain embodiments, the level of mRNA expression is assessed using branched DNA (bDNA) assays or real time PCR (qPCR). The use of these methods is described and exemplified in the Examples presented herein. In certain embodiments, expression level is determined by the method provided in the Examples using a lOnM siRNA concentration in the species matched cell line.
The level of B°AT1 protein expression may be determined using any method known in the art for the measurement of protein levels. Such methods include, for example, electrophoresis, capillary electrophoresis, high performance liquid chromatography (HPLC), thin layer chromatography (TLC), hyperdiffusion chromatography, fluid or gel precipitin reactions, absorption spectroscopy, a colorimetric assays, spectrophotometric assays, flow cytometry, immunodiffusion (single or double), immunoelectrophoresis, western bloting, radioimmunoassay (RIA), enzyme-linked immunosorbent assays (ELISAs), immunofluorescent assays, electrochemiluminescence assays, and the like.
In some embodiments, the efficacy of the methods of the present disclosure are assessed by a decrease in SLC6A19 mRNA or B°AT1 protein level (e.g., in a kidney biopsy).
In some embodiments of the methods of the present disclosure, the ASO or iRNA agent is administered to a subject such that the ASO or iRNA agent is delivered to a specific site within the subject. The inhibition of expression of SLC6A19 may be assessed using measurements of the level or change in the level of SLC6A19 mRNA or B°AT1 protein in a sample derived from fluid or tissue from the specific site within the subject (e.g., kidney or blood).
As used herein, the terms detecting or determining a level of an analyte are understood to mean performing the steps to determine if a material, e.g., protein, RNA, is present. As used herein, methods of detecting or determining include detection or determination of an analyte level that is below the level of detection for the method used.
Prophylactic and Treatment Methods of the Disclosure
The instant disclosure also provides methods of using an ASO or iRNA agent of the present disclosure or a composition containing an ASO or iRNA agent of the present disclosure to inhibit expression of SLC6A19, thereby preventing or treating an SLC6A19-associated disorder, e.g., metabolic disorders, including a glomerular disorder, a renal tubular disorder, other renal disorders, an inborn error of metabolism, a systemic metabolic disorder, a disorder of the thyroid, a disorder of the parathyroid, a disorder of the inner ear, a neurological disorder, or a viral infection, or any combination thereof. Targeting of SLC6A19 in kidney via ASO was identified in a mouse model of phenylketonuria (PKU) to reduce circulating levels of phenylalanine (Phe) in such mice, indicating a role for inhibition of SLC6A19 in kidney as a therapy for PKU and related aminoacidopathies (Belanger et al. JCI Insight 3(14): el21762). In the methods of the present disclosure, the cell may be contacted with the ASO or siRNA in vitro or in vivo, i.e., the cell may be within a subject
.A cell suitable for treatment using the methods of the present disclosure may be any cell that expresses a SLC6A19 gene, e.g., a kidney cell, a urinary bladder cell, a gastrointestinal tract cell (e.g., a duodenum or small intestine cell), or a gall bladder cell, but optionally a kidney cell. A cell suitable for use in the methods of the present disclosure may be a mammalian cell, e.g., a primate cell (such as a human cell, including human cell in a chimeric non-human animal, or a non-human primate cell, e.g., a monkey cell or a chimpanzee cell), or a non-primate cell. In certain embodiments, the cell is a human cell, e.g., a human kidney cell. In the methods of the present disclosure, SLC6A19 expression is inhibited in the cell by at least 50, 55, 60, 65, 70, 75, 80, 85, 90, or 95, or to a level below the level of detection of the assay.
The in vivo methods of the present disclosure may include administering to a subject a composition containing an ASO or iRNA agent, where the ASO or iRNA agent includes a nucleotide sequence that is complementary to at least a part of an RNA transcript of the SLC6A19 gene of the mammal to which the ASO or iRNA agent is to be administered. The composition can be administered by any means known in the art including, but not limited to oral, intraperitoneal, or parenteral routes, including intracranial (e.g., intraventricular, intraparenchymal, and intrathecal), intravenous, intramuscular, subcutaneous, transdermal, airway (aerosol ), nasal, rectal, and topical (including buccal and sublingual) administration. In certain embodiments, the compositions are administered by intravenous infusion or injection. In certain embodiments, the compositions are administered by subcutaneous injection. In certain embodiments, the compositions are administered by intramuscular injection.
In one aspect, the instant disclosure also provides methods for inhibiting the expression of a SLC6A19 gene in a mammal. The methods include administering to the mammal a composition comprising an ASO or dsRNA that targets a SLC6A19 gene in a cell of the mammal and maintaining the mammal for a time sufficient to obtain degradation of the niRNA transcript of the SLC6A19 gene, thereby inhibiting expression of the SLC6A19 gene in the cell.
Reduction in gene expression can be assessed by any methods known in the art and by methods, e.g., qRT-PCR, described herein, e.g., in the Examples. Reduction in protein production can be assessed by any methods known it the art, e.g , ELISA. In certain embodiments, a kidney biopsy sample serves as the tissue material for monitoring the reduction in the SLC6A19 gene or protein expression. In other embodiments, a blood sample serves as the subject sample for monitoring the reduction in the B°AT1 protein expression.
The instant disclosure further provides methods of treatment in a subject in need thereof, e.g., a subject diagnosed with an SLC6A19-associated disorder, such as, a glomerular disorder, a renal tubular disorder, other renal disorders, an inborn error of metabolism, a systemic metabolic disorder, a disorder of the thyroid, a disorder of the parathyroid, a disorder of the inner ear, a neurological disorder, or a viral infection, or any combination thereof.
The instant disclosure further provides methods of prophylaxis in a subject in need thereof. The treatment methods of the present disclosure include administering an ASO or iRNA agent of the present disclosure to a subject, e.g , a subject that would benefit from a reduction of SLC6A19 expression, in a prophylactically effective amount of an ASO or iRNA agent targeting a SLC6A19 gene or a pharmaceutical composition comprising an ASO or iRNA agent targeting a SLC6A19 gene.
In one embodiment, an SLC6A19-associated disease is selected from the group consisting of kidney diseases or conditions, including glomerular disorders, renal tubular disorders, other renal disorders, inborn errors of metabolism, systemic metabolic disorders, disorders of the thyroid, disorders of the parathyroid, disorders of the inner ear, neurological disorders, or viral infections. In one embodiment, a SLC6A 19-associated disease is a glomerular disorder, including Lupus nephritis, Goodpasture syndrome, IgA nephropathy, Alport syndrome, glomerulosclerosis, diabetic nephropathy, focal segmental glomerulosclerosis, membranous nephropathy, minimal change disease, ApoLl nephropathy, post-infection glomerulonephritis, membranoproliferative glomerulonephritis, mesangioproliferative glomerulonephritis, nephrotic syndrome, nephritic syndrome, Anti-LRP2 nephropathy, C3 glomerulopathy, or any combination thereof.
In one embodiment, a SLC6A19-associated disease is a renal tubular disorder, including Fanconi syndrome, cystinuria, Lowe syndrome, Dent syndrome, Light Chain Proximal Tubulopathy, Gitelman syndrome, renal tubular acidosis, nephrogenic diabetes insipidus, Bartter syndrome, Liddle syndrome, hereditary aminoaciduria, hereditary salt wasting disorders, hereditary phosphate wasting disorders, porphyria associated renal disease, nephropathic cystinosis, autosomal dominant tubulointerstitial kidney disease, or any combination thereof.
In another embodiment, a SLC6A 19-associated disease is an other renal disorder, including Autosomal dominant polycystic kidney disease (ADPKD), Autosomal recessive polycystic kidney disease (ARPKD), Nephronophthisis, Chronic Kidney Disease, nephrolithiasis, acute kidney injury, Alagille syndrome, cardiorenal syndrome, renal cell carcinoma, renal osteodystrophy, or any combination thereof.
In a further embodiment, a SLC6A19-associated disease is an inborn error of metabolism, including phenylketonuria, urea cycle disorder, maple syrup urine disease, galactosemia, hereditary tyrosinemia, glutamic academia, isovaleric acidemia, very long/long/medium/short chain acyl-CoA dehydrogenase deficiency, methylmalonic academia, primary hyperoxaluria, propionic academia, porphyria, Wilson disease, Pyruvate dehydrogenase deficiency, homocystinuria, hereditary fructose intolerance, nonketotic hyperglycinemia, or any combination thereof.
Any disorder that may be a cause of kidney disease or disorder, including a glomerular disorder, a renal tubular disorder, other renal disorders, an inborn error of metabolism, a systemic metabolic disorder, a disorder of the thyroid, a disorder of the parathyroid, a disorder of the inner ear, a neurological disorder, or a viral infection, or any combination thereof, as well as various other kidney diseases or conditions is encompassed by the term ‘:SLC6A 19-associated disease”. Non-limiting examples of SLC6A 19-associated diseases include Lupus nephritis, Goodpasture syndrome, IgA nephropathy, Alport syndrome, glomerulosclerosis, diabetic nephropathy, focal segmental glomerulosclerosis, membranous nephropathy, minimal change disease, ApoLl nephropathy, post-infection glomerulonephritis, membranoproliferative glomerulonephritis, mesangioproliferative glomerulonephritis, nephrotic syndrome, nephritic syndrome, Anti-LRP2 nephropathy, C3 glomerulopathy, Fanconi syndrome, cystinuria, Lowe syndrome, Dent syndrome, Light Chain Proximal Tubulopathy, Gitelman syndrome, renal tubular acidosis, nephrogenic diabetes insipidus, Bartter syndrome, Liddle syndrome, hereditary aminoaciduria, hereditary salt wasting disorders, hereditary phosphate wasting disorders, porphyria associated renal disease, nephropathic cystinosis, autosomal dominant tubulointerstitial kidney disease, Autosomal dominant polycystic kidney disease (ADPKD), Autosomal recessive polycystic kidney disease (ARPKD), Nephronophthisis, Chronic Kidney Disease, nephrolithiasis, acute kidney injury, Alagille syndrome, cardiorenal syndrome, renal cell carcinoma, renal osteodystrophy, Autosomal dominant polycystic kidney disease (ADPKD), Autosomal recessive polycystic kidney disease (ARPKD), Nephronophthisis, Chronic Kidney Disease, nephrolithiasis, acute kidney injury, Alagille syndrome, cardiorenal syndrome, renal cell carcinoma, renal osteodystrophy, or any combination thereof.
An ASO or iRNA agent of the present disclosure may be administered as a "free ASO" or a ‘‘free iRNA.’1 A free ASO or free iRNA is administered in the absence of a pharmaceutical composition. The naked ASO or iRNA may be in a suitable buffer solution. The buffer solution may comprise acetate, citrate, prolamine, carbonate, or phosphate, or any combination thereof. In one embodiment, the buffer solution is phosphate buffered saline (PBS). The pH and osmolarity of the buffer solution containing the ASO or iRNA can be adjusted such that it is suitable for administering to a subject.
Alternatively, an ASO or iRNA agent of the present disclosure may be administered as a pharmaceutical composition, such as an ASO or dsRNA liposomal formulation.
Subjects that would benefit from an inhibition of SLC6A19 gene expression are subjects susceptible to or diagnosed with a SLC6A19-associated disorder, such as metabolic disorders, including nephropathy and various other kidney diseases or conditions, etc.
In an embodiment, the method includes administering a composition featured herein such that expression of the target SLC6A19 gene is decreased, such as for about 1, 2, 3, 4, 5, 6, 1-6, 1- 3, or 3-6 months per dose. In certain embodiments, the composition is administered once every 3- 6 months. Optionally, the ASOs or iRNAs useful for the methods and compositions featured herein specifically target RNAs (primary or processed) of the target SLC6A19 gene. Compositions and methods for inhibiting the expression of genes using ASOs or iRNAs can be prepared and performed as described herein.
Administration of the ASO or iRNA agent according to the methods of the present disclosure may result in prevention or treatment of an SLC6A19-associated disorder, e.g., metabolic disorders, including nephropathy and various other kidney diseases or conditions.
Subjects can be administered a therapeutic amount of iRNA, such as about 0.01 mg/kg to about 200 mg/kg.
The ASO or iRNA agent is optionally administered intravenously or subcutaneously, i.e., by intravenous or subcutaneous injection. One or more injections may be used to deliver the desired dose of ASO or iRNA agent to a subject. The injections may be repeated over a period of time.
The administration may be repeated on a regular basis. In certain embodiments, after an initial treatment regimen, the treatments can be administered on a less frequent basis. A repeatdose regimen may include administration of a therapeutic amount of ASO or iRNA agent on a regular basis, such as once per month to once a year. In certain embodiments, the ASO or iRNA agent is administered about once per month to about once every' three months, or about once every' three months to about once every six months.
The present disclosure provides methods and uses of an ASO or iRNA agent or a pharmaceutical composition thereof for treating a subject that would benefit from reduction and/or inhibition of SLC6A19 gene expression, e.g., a subject having a SLC6A19-associated disease, in combination with other pharmaceuticals and/or other therapeutic methods, e.g., with known pharmaceuticals and/or known therapeutic methods, such as, for example, those which are currently employed for treating these disorders.
Accordingly, in some aspects of the present disclosure, the methods which include either a single ASO or iRNA agent of the present disclosure, further include administering to the subject one or more additional therapeutic agents.
The ASO or iRNA agent and an additional therapeutic agent and/or treatment may be administered at the same time and/or in the same combination, e.g., parenterally, or the additional therapeutic agent can be administered as part of a separate composition or at separate times and/or by another method known in the art or described herein.
Examples of additional therapeutic agents include those known to treat metabolic disorders, including nephropathy and various other kidney diseases or conditions and other diseases that are caused by, associ ated with or are a consequence of metabolic disorders, including nephropathy and various other kidney diseases or conditions.
The ASO or iRNA agent and an additional therapeutic agent and/or treatment may be administered at the same time and/or in the same combination, e.g., parenterally, or the additional therapeutic agent can be administered as part of a separate composition or at separate times and/or by another method known in the art. or described herein.
Combination Therapies
In some embodiments, a modulatory nucleic acid agent of the instant disclosure - e.g., an ASO, a dsRNA and/or a conjugate agent that includes one or more modulatory nucleic acid agents disclosed herein, or a composition comprising the same - is administered in combination with an additional agent, e.g., additional therapy. In some embodiments, an additional therapy comprises a therapy for a disease or disorder, e.g., a standard of care (SOC) therapy, for a symptom, disease or disorder. In some embodiments, a modulatory nucleic acid agent is administered before, concurrently with or after administration of an additional therapy, e.g., a SOC therapy.
Kits
In certain aspects, the instant disclosure provides kits that include a suitable container containing a pharmaceutical formulation of an ASO or iRNA agent compound, e.g., an ASO, a double-stranded siRNA compound, or ssiRNA compound, (e.g., a precursor, e g., a larger siRNA compound which can be processed into a ssiRNA compound, or a DNA which encodes an ASO or dsRNA compound, e.g., a double-stranded siRNA compound, or ssiRNA compound, or precursor thereof)
Such kits include one or more dsRNA agent(s) and instructions for use, e.g., instructions for administering a prophy lacti cal ly or therapeutically effective amount of an ASO or dsRNA agent(s). The ASO or dsRNA agent may be in a vial or a pre-filled syringe. The kits may optionally further comprise means for administering the ASO or dsRNA agent, (e.g., an injection device, such as a pre-filled syringe), or means for measuring the inhibition of SLC6A19 (e.g , means for measuring the inhibition of SLC6A19 inRNA, SLC6A19 protein, and/or SLC6A19 activity) Such means for measuring the inhibition of SLC6A19 may comprise a means for obtaining a sample from a subject, such as, e.g., a plasma sample. The kits of the present disclosure may optionally further comprise means for determining the therapeutically effective or prophylactically effective amount.
In certain embodiments the individual components of the pharmaceutical formulation may be provided in one container, e.g., a vial or a pre-filled syringe. Alternatively, it may be desirable to provide the components of the pharmaceutical formulation separately in two or more containers, e.g., one container for an ASO or siRNA compound preparation, and at least another for a carrier compound. The kit may be packaged in a number of different configurations such as one or more containers in a single box. The different components can be combined, e.g., according to instructions provided with the kit. The components can be combined according to a method described herein, e.g., to prepare and administer a pharmaceutical composition The kit can also include a delivery7 device.
INCORPORATION BY REFERENCE
All publications, patent applications, patents, and other references mentioned herein, including GenBank Accession Numbers, are incorporated by reference in their entirety. In addition, the materials, methods, and examples are illustrative only and not intended to be limiting. Unless otherwise defined, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. Although methods and materials similar or equivalent to those described herein can be used in the practice or testing of the present invention, suitable methods and materials are described herein.
This present disclosure is further illustrated by the following examples which should not be construed as limiting the scope or content of the disclosure in any way. The entire contents of all references, patents and published patent applications cited throughout this application, as well as the informal Sequence Listing and Figures, are hereby incorporated herein by reference. EXAMPLES
Example 1: Materials and Methods
Screening of Unconjugated Modulatory Nucleic Acid Agents (e.g., ASOs, siRNAs, PMOs, exonskipping oligonucleotides)
HEP3B217 cells (ATCC, cat # HB-8064) were grown to near confluence at 37°C in an atmosphere of 5% CO?, in Eagle’s Minimum Essential Medium (Gibco® ) supplemented with 10% FBS (ATCC) before being released from the plate by trypsinization. Cells were counted and reseeded in a 96-well plate. Transfection was carried out by adding 0.2 pL of Lipofectamine1*1 RNAiMax (Invitrogeifo, Carlsbad, CA. cat # 13778-150) plus 1 nM and 10 nM of modulatory nucleic acid agent (e g., siRNA, as shown in FIG. 1, or ASC) or other agent(s)) in Opti-MEM. The mixture was incubated at room temperature for 20 minutes. Eighty pl of complete growth media without antibiotic containing ~2 xlO4 HEP3B217 cells was then added to 20 uL of modulatory nucleic acid agent mixture. Cells were placed in the incubator at 37°C and 5% CO?, and allowed to incubate for 48 hours. Primary screening was performed at 1 nM and 10 nM (FIG. 1), and doseresponse studies w7ere carried out at 9-point half-log dilution between 0.03 nM and 100 nM. A known human SLC6A19-targeting siRNA (Horizon: L-007352-01-0050) was used as a positive control of transfection
In alternative embodiments, for screening of PMOs, 6 pM Endo-Porter (GeneTools), a reagent for delivering Morpholino oligonucleotides into the cytosol of cultured cells, is used as a transfection agent, while other methods described above remain the same.
RT-PCR Readout
To prepare samples for RT-qPCR, the cells were washed with PBS and lysed using the Cells-to-CT™ 1-Step TaqMan™ Kit (Thermo Fi sher Scientific™). 50 pL of lysis buffer / DNase I reagent mixture was added into each well, mixed thoroughly and incubated for 5-10 minutes at room temperature. 5 uL of Stop Solution was added to each well and incubated for 5 minutes. The cell lysates were used for RT-qPCR analysis, to evaluate mRNA expression. RT-qPCR was performed on a Quant Studio 7 Pro Real-Time PCR System using a Cells-to-Cells-to-CT™ 1- Step TaqMan™ kit and TaqMan™ probes (Thermo Fisher Scientific™). Cell lysates from treatment and control wells were probed with Human SLC6A19-FAM (Thermo: HS01384157 M1, 4448489) and Human HMBS-VIC (Thermo: MM01143545 MI, 4448489). HMBS was designated as the house keeping gene for normalization The fold change of target gene relative to Non-targeting control wells (Horizon: D-001810-01-05) was calculated as 2A(- AACt). Relative expression was compiled and analyzed in GraphPad Prism.
Protein Readout
To prepare samples for western blotting, cells are washed with ice-cold PBS and 200 pL chilled complete RIP A lysis buffer (Thermo Scientific®, Cat. #89901) plus Hall Protease/Phosphatase Inhibitor Cocktail (Thermo Scientific®, Cat. #78440) is added to lyse the cells. Samples are mechanically sheared by passing the sample 5-6 times through a 21 -gauge needle. The lysates are incubated for 20 min in a cold room with gentle shaking and centrifuged at 14,000 g for 10 min at 4 °C. The supernatants are transferred to a pre-chilled microcentrifuge tube. Total protein is quantified using Pierce1M BCA Protein Assay Kit (Thermo Scientific®, Cat #23221) according to manufacturer’s instructions. Samples are prepared by adding NuPAGE1M LDS sample buffer (Thermo Scientific®, Cat #NP0007) and NuPAGE™ sample Reducing Agent (Thermo Scientific®, Cat. &NP0004) to the cell lysates. The mixture is heated at 95 °C for 10 min to denature proteins, spun down briefly and samples are used for western blotting or stored at -80 °C. Twenty pL of samples are loaded on NuPAGE™ 4 to 12%, Bis-Tris 1.0-1.5 mm, Mini Protein Gels along with 3 pL of Duo Pre-stained Protein Ladder (LI-COR Cat. #988-15653) and run at constant 200V for 45 min The gels are transferred to membrane using Invitrogen™ iBlot™ 2 Dry Blotting System. The membrane is blocked with Intercept®' Blocking Buffer (LI-COR™ Cat. #927-60001 ) for 1 hour at RT or 4 °C for overnight with gentle shaking, followed by incubation with anti-SLC6A19 primary antibody and appropriate secondary’ antibody. The antibody solutions are washed off' with TBST, and membranes are read on a LI-COR™ Odyssey® imaging system. Screening of Conjugated Modulatory Nucleic Acid Agents (e.g., ASOs, siRNAs, PMOs, exonskipping oligonucleotides)
For screening of conjugated modulatory nucleic acid agents, similar methods are followed as described above. However, the conjugated molecules are not subjected to transfection, but are instead administered to allow' free uptake. For free uptake cell culture experiments, cells are seeded in serum-free media and treated with conjugated modulatory nucleic acid agent without any transfection reagent for up to 4 hours. The media is then aspirated, cells are washed with PBS, then incubated in complete media for a further 24 -72 hours. The cells are then harvested for either RNA or protein detection according to the methods mentioned above. Example 2: Design and Screening of SLC6A19-Targeting Antisense Oligonucleotides and siRNAs
Human SLC6A19-targeting ASOs (including exon-skipping ASOs) and siRNAs were designed by art-recognized processes and are shown in Tables 3-15. Initial assessment of human SLC6A19-targeting siRNAs was performed using the methods of the above Example, and multiple human SLC6A19-targeting siRNAs that exhibited significant knockdown of SLC6A19 expression were thereby identified (FIG. 1). Dose-responsive behavior was also observed for many such siRNAs, when comparing between 10 nM and 1 nM administrations of respective siRNAs in HEP3B cell culture.
ASOs tested in screening experiments are shown in Table 5, while siRNAs tested in such screening assays are shown in Tables 10 and 11, and exon-skipping antisense oligonucleotides are presented in Tables 12 and 13. Highly effective inhibitory ASOs (including exon-skipping .ASOs) and additional siRNAs targeting SLC6A19 for knockdown are thereby identified.
Table 3. SLC6A19~Targeting Antisense Oligonucleotides
Figure imgf000164_0001
Figure imgf000165_0001
Figure imgf000166_0001
Figure imgf000167_0001
Figure imgf000168_0001
Figure imgf000169_0001
Figure imgf000170_0001
Figure imgf000171_0001
Figure imgf000172_0001
Figure imgf000173_0001
Figure imgf000174_0001
Figure imgf000175_0001
Figure imgf000176_0001
Figure imgf000177_0001
Figure imgf000178_0001
|
Figure imgf000179_0001
777 ~|
Table 4. Selected SLC6A19-Targeting Antisense Oligonuekotides
Figure imgf000179_0002
Figure imgf000180_0001
Figure imgf000181_0001
Figure imgf000182_0001
Figure imgf000183_0001
Table 5. Further Selected SLC6A 19- Targeting Antisense Oligonucleotides
Figure imgf000183_0002
Figure imgf000184_0001
Figure imgf000185_0001
Table 6. Screening Set Selection of SLC6A19-Targeting Antisense OHgonncleotides
Figure imgf000185_0002
Figure imgf000186_0001
Table 7. Selected SLC6A19-Targeting siRNAs
Figure imgf000186_0002
Figure imgf000187_0001
Figure imgf000188_0001
Figure imgf000189_0001
Figure imgf000190_0001
Figure imgf000191_0001
Figure imgf000192_0001
Figure imgf000193_0001
Figure imgf000194_0001
Figure imgf000195_0001
Figure imgf000196_0001
Figure imgf000197_0001
Figure imgf000198_0001
Figure imgf000199_0001
Figure imgf000200_0001
Table 8. Further Selected SLC6A19-Targeting siRNAs
Figure imgf000200_0002
Figure imgf000201_0001
Figure imgf000202_0001
Figure imgf000203_0001
Figure imgf000204_0001
Figure imgf000205_0001
Figure imgf000206_0001
Table 9. Additionally Selected SLC6A19-Targeting siRNAs
Figure imgf000206_0002
Figure imgf000207_0001
Figure imgf000208_0001
Figure imgf000209_0001
Tabk 10. Sub-sekrtion of SLC6A19~Targeting siRNAs
Figure imgf000209_0002
Table 11. Alternative Selection of SLC6A19"Targeting siRNAs
Figure imgf000210_0001
Figure imgf000211_0001
Figure imgf000212_0001
Table 12. SLC6A19 Exon-Skipping Antisense Oligonucleotides
Figure imgf000212_0002
Figure imgf000213_0001
Figure imgf000214_0001
Figure imgf000215_0001
m = 2'-methoxyethyl nucleotide
Table 13. Selected SLC6A19 Exon-Skipping Antisense Oligonucleotides
Figure imgf000215_0002
m = 2'-methoxyethyl nucleotide
Example 3: Screening of dsRNA Duplexes using Hep3B cells
Hep3B cells (ATCC Cat#: HB-8064) were cultured rn vitro Cells were seeded in 96-well plates using MEM (Gibco Cat#: 10370-021) supplemented with GlutaMAX and 10% FBS. Following plating, cells were treated with siRNA/transfection reagent (RNAiMAX) complexes (Table 15), at final concentrations of 1 nM or 10 nM, or with a control treatment. Cells were harvested 72 hours post-treatment for RNA isolation and cDNA synthesis, utilizing the TaqMan™ Fast Advanced Cells-to-CT™ Kit (ThermoFisher Cat#: A35378). Quantitative PCR (QPCR) analysis w-as performed using a Taqman probe at a concentration of 60X to measure SLC6A19 (ThermoFisher Cat#: Hs01384157_ml) expression. The expression levels of SLC6A19 wese normalized to ACTB (ThermoFisher Cat#: Hs01060665 m l) levels for analysis (Table 16).
Table 14. SLC6A19-Targeting siRNAs
Figure imgf000216_0001
Figure imgf000217_0001
Figure imgf000218_0001
Table 15. SLC6A19-Targeting siRNAs
Figure imgf000218_0002
Figure imgf000219_0001
Figure imgf000220_0001
Figure imgf000221_0001
modifications; and f and upper case letters denote 2' fluoro modifications
Table 16. SLC6A19-Targeting siRNAs Screening Results
Figure imgf000221_0002
Figure imgf000222_0001
Figure imgf000223_0001
Example 4: Screening of dsRNA Duplexes using Li-7 ceils
Li-7 cells (Riken, RCB1941) were cultured at 37°C in 5% CO2 using RPMI 1640 medium (Gibco, Cat #: C22400500CP) supplemented with 10% FBS (ExCell-Bio-FSP500) and 1% Penicillin-Streptomycin (Gibco, Cat #: 15140122). After plating, transfection was performed by adding RNAiMax (Invitrogen, Carlsbad, CA, Cat # 13778-150) along with 1 nM and 10 nM siRNA duplexes (Table 17). Cells were harvested 24 hours post-treatment for RNA analysis. Total RNA was extracted using the EZ-Press 96 RNA Purification Kit (EZBioscience, Cat #: EZ4001- L), and cDNA was generated with HiScript III RT SuperMix (Vazyme, Cat #: R323-01). Quantitative PCR (QPCR) analysis was conducted using a custom Taqman probe for SLC6A19 (Sangon Biotech, Shanghai, China), normalized to ACTB (Sangon Biotech, Shanghai, China) for evaluation (Table 19).
Figure imgf000223_0002
Table 17. SLC6A19-Targeting siRNAs
Figure imgf000223_0003
Figure imgf000224_0001
Figure imgf000225_0001
Figure imgf000226_0001
Figure imgf000227_0001
Figure imgf000228_0001
Figure imgf000229_0001
Figure imgf000230_0001
Table 18. SLC6A19-Targeting siRNAs
Figure imgf000230_0002
Figure imgf000231_0001
Figure imgf000232_0001
Figure imgf000233_0001
Figure imgf000234_0001
Figure imgf000235_0001
Figure imgf000236_0001
Figure imgf000237_0001
Figure imgf000238_0001
Figure imgf000239_0001
Figure imgf000240_0001
Asterisks (*) denote phosphorothioate linkages; lower case letters denote 2'-oMe modifications; and f and upper case letters denote 2' fluoro modifications
Table 19. SLC6A19-Targetmg siRNAs Screening Results
Figure imgf000240_0002
Figure imgf000241_0001
Figure imgf000242_0001
Figure imgf000243_0001
Figure imgf000244_0001
Figure imgf000245_0001
Example 5; In vivo screening of dsRNA Duplexes in Mice
Duplexes of interest, identified from the above in vitro studies, are evaluated in vivo.
At day 0, groups of three mice expressing human SLC6A19 mRNA are administered a single 3 mg/kg dose of the agents of interest (including conjugate agents as described herein) or PBS control via intravenous injection. At day 7 or day 14 post-dose, animals are sacrificed, kidney samples are collected and snap-frozen in liquid nitrogen. Kidney mRNA is extracted and analyzed by the RT-QPCR method.
Human SLC6A19 mRNA levels are compared to a housekeeping genes, like GAPDH, PPIB. The values are then normalized to the average of PBS vehicle control group. The data are expressed as percent of baseline value, and are presented as mean plus standard deviation. Results are expected to demonstrate that the exemplary duplex agents tested effectively reduce the level of the human SLC6A19 messenger RNA in vivo in targeted kidney cells.
EQUIVALENTS
Those skilled in the art will recognize, or be able to ascertain using no more than routine experimentation, many equivalents to the specific embodiments and methods described herein. Such equivalents are intended to be encompassed by the scope of the following claims.

Claims

We claim:
1. A modulatory' nucleic acid agent comprising a first strand of 12 to 60 linked nucleosides in length targeted to a human solute carrier family 6 member 19 (SLC6A19) RNA, wherein the human SLC6A19 RNA is the human SLC6A19 RNA of SEQ ID NO: 1.
2. The modulatory nucleic acid agent of claim 1, wherein the modulatory nucleic acid agent comprises an antisense oligonucleotide (ASO).
3. The ASO of claim 2, having at least about 80% complementarity to a target region of the human SLC6A19 RNA.
4. The antisense compound of claim 2, wherein the ASO hybridizes with 12 or more nucleotides of nucleotides 3 to 25, nucleotides 8 to 30, nucleotides 13 to 35, nucleotides 38 to 60, nucleotides 43 to 65, nucleotides 48 to 70, nucleotides 93 to 115, nucleotides 98 to 120, nucleotides
103 to 125, nucleotides 108 to 130, nucleotides 111 to 133, nucleotides 113 to 135, nucleotides 118 to 140, nucleotides 214 to 236, nucleotides 219 to 241, nucleotides 244 to 266, nucleotides 249 to 271, nucleotides 252 to 274, nucleotides 252 to 274, nucleotides 253 to 275, nucleotides 254 to 276, nucleotides 257 to 286, nucleotides 259 to 281, nucleotides 264 to 286, nucleotides 289 to 311, nucleotides 294 to 316, nucleotides 339 to 361, nucleotides 397 to 419, nucleotides 460 to 480, nucleotides 461 to 481, nucleotides 481 to 503, nucleotides 486 to 508, nucleotides 491 to 513, nucleotides 557 to 579, nucleotides 561 to 583, nucleotides 562 to 584, nucleotides 564 to 586, nucleotides 566 to 586, nucleotides 567 to 589, nucleotides 616 to 638, nucleotides 621 to 643, nucleotides 626 to 648, nucleotides 670 to 692, nucleotides 671 to 691, nucleotides 672 to 694, nucleotides 674 to 694, nucleotides 675 to 697, nucleotides 680 to 702, nucleotides 685 to 707, nucleotides 690 to 712, nucleotides 695 to 717, nucleotides 700 to 722, nucleotides 705 to 727, nucleotides 710 to 732, nucleotides 715 to 737, nucleotides 720 to 742, nucleotides 725 to 747, nucleotides 730 to 752, nucleotides 735 to 757, nucleotides 740 to 762, nucleotides 745 to 767, nucleotides 750 to 772, nucleotides 751 to 776, nucleotides 755 to 777, nucleotides 760 to 782, nucleotides 765 to 787, nucleotides 770 to 792, nucleotides 775 to 797, nucleotides 798 to 823, nucleotides 800 to 822, nucleotides 825 to 847, nucleotides 890 to 912, nucleotides 895 to 917, nucleotides 900 to 922, nucleotides 905 to 927, nucleotides 910 to 932, nucleotides 911 to 937, nucleotides 915 to 937, nucleotides 972 to 994, nucleotides 973 to 995, nucleotides 977 to 1000, nucleotides 977 to 999, nucleotides 978 to 1000, nucleotides 983 to 1005, nucleotides 1035 to 1057, nucleotides 1058 to 1080, nucleotides 1059 to 1081, nucleotides 1060 to 1082, nucleotides 1063 to 1082, nucleotides 1065 to 1087, nucleotides 1068 to 1087, nucleotides 1070 to 1094, nucleotides 1070 to 1092, nucleotides 1074 to 1096, nucleotides 1075 to 1097, nucleotides 1077 to 1096, nucleotides 1080 to 1102, nucleotides 1085 to 1107, nucleotides 1086 to 1106, nucleotides 1090 to 1112, nucleotides 1095 to 1117, nucleotides 1100 to 1122, nucleotides 1105 to 1127, nucleotides 1110 to 1132, nucleotides 1115 to 1137, nucleotides 1115 to 1137, nucleotides 1117 to 1139, nucleotides 1177 to 1199, nucleotides 1182 to 1204, nucleotides 1187 to 1209, nucleotides 1191 to 1214, nucleotides 1192 to 1214, nucleotides 1195 to 1217, nucleotides 1234 to 1256, nucleotides 1239 to 1261, nucleotides 1242 to 1264, nucleotides 1244 to 1266, nucleotides 1247 to 1268, nucleotides 1249 to 1271, nucleotides 1251 to 1270, nucleotides 1254 to 1276, nucleotides 1259 to 1281, nucleotides 1263 to 1282, nucleotides 1264 to 1286, nucleotides 1266 to 1285, nucleotides 1269 to 1291, nucleotides 1326 to 1348, nucleotides 1331 to 1353, nucleotides 1334 to 1361, nucleotides 1334 to 1356, nucleotides 1334 to 1356, nucleotides 1335 to 1357, nucleotides 1336 to 1358, nucleotides 1337 to 1359, nucleotides 1337 to 1359, nucleotides 1338 to 1360, nucleotides 1340 to 1362, nucleotides 1341 to 1363, nucleotides 1346 to 1368, nucleotides 1351 to 1373, nucleotides 1353 to 1372, nucleotides 1377 to 1399, nucleotides 1382 to 1404, nucleotides 1383 to 1403, nucleotides 1387 to 1409, nucleotides 1392 to 1414, nucleotides 1397 to 1419, nucleotides 1402 to 1424, nucleotides 1407 to 1429, nucleotides 1412 to 1434, nucleotides 1417 to 1439, nucleotides 1456 to 1478, nucleotides 1458 to 1477, nucleotides 1481 to 1503, nucleotides 1486 to 1508, nucleotides 1491 to 1513, nucleotides 1496 to 1518, nucleotides 1521 to 1543, nucleotides 1568 to 1590, nucleotides 1569 to 1591, nucleotides 1574 to 1596, nucleotides 1578 to 1621, nucleotides 1579 to 1601, nucleotides 1583 to 1605, nucleotides 1583 to 1605, nucleotides 1584 to 1606, nucleotides 1584 to 1606, nucleotides 1585 to 1607, nucleotides 1585 to 1607, nucleotides 1587 to 1609, nucleotides 1588 to 1610, nucleotides 1589 to 1611, nucleotides 1594 to 1616, nucleotides 1595 to 1617, nucleotides 1599 to 1621, nucleotides 1601 to 1620, nucleotides 1602 to 1624, nucleotides 1642 to 1664, nucleotides 1749 to 1775, nucleotides 1750 to 1772, nucleotides 1755 to 1777, nucleotides 1757 to 1779, nucleotides 1758 to 1780, nucleotides 1760 to 1782, nucleotides 1761 to 1792, nucleotides 1764 to 1783, nucleotides 1765 to 1787, nucleotides 1770 to 1792, nucleotides 1809 to 1831, nucleotides 1810 to 1829, nucleotides 1812 to 1832, nucleotides 1814 to 1836, nucleotides 1852 to 1881, nucleotides 1853 to 1875, nucleotides 1854 to 1876, nucleotides 1858 to 1880, nucleotides 2168 to 2188, nucleotides 2333 to 2353, nucleotides 2454 to 2474, nucleotides 2507 to 2527, nucleotides 2593 to 2613, nucleotides 3244 to 3264, nucleotides 3288 to 3308, nucleotides 3304 to 3324, nucleotides 3332 to 3352, nucleotides 3333 to 3353, nucleotides 3501 to 3523, nucleotides 3595 to 3615, nucleotides 3595 to 3617, nucleotides 3643 to 3671, nucleotides 3646 to 3668, nucleotides 3646 to 3668, nucleotides 3647 to 3669, nucleotides 3648 to 3668, nucleotides 3648 to 3670, nucleotides 3648 to 3670, nucleotides 3649 to 3671, nucleotides 3651 to 3670, nucleotides 3651 to 3673, nucleotides 3651 to 3673, nucleotides 3652 to 3671, nucleotides 3712 to 3733, nucleotides 3713 to 3735, nucleotides 3763 to 3786, nucleotides 3763 to 3785, nucleotides 3764 to 3786, nucleotides 3791 to 3811, nucleotides 3796 to 3816, nucleotides 3801 to 3829, nucleotides 3801 to 3820, nucleotides 3802 to 3821, nucleotides 3804 to 3826, nucleotides 3809 to 3831, nucleotides 3810 to 3832, nucleotides 3812 to 3833, nucleotides 3812 to 3834, nucleotides 3813 to 3835, nucleotides 3814 to 3833, nucleotides 3814 to 3836, nucleotides 3814 to 3836, nucleotides 3839 to 3859, nucleotides 3840 to 3862, nucleotides 3902 to 3924, nucleotides 3965 to 3987, nucleotides 3995 to 4015, nucleotides 3998 to 4020, nucleotides 4100 to 4124, nucleotides 4102 to 4124, nucleotides 4141 to 4163, nucleotides 4141 to 4163, nucleotides 4142 to 4164, nucleotides 4143 to 4165, nucleotides 4144 to 4166, nucleotides 4146 to 4168, nucleotides 4150 to 4172, nucleotides 4151 to 4173, nucleotides 4192 to 4214, nucleotides 4197 to 4219, nucleotides 4201 to 4223, nucleotides 4265 to 4287, nucleotides 4270 to 4292, nucleotides 4318 to 4338, nucleotides 4319 to 4341, nucleotides 4329 to 4349, nucleotides 4348 to 4386, nucleotides 4349 to 4371, nucleotides 4350 to 4369, nucleotides 4350 to 4372, nucleotides 4350 to 4372, nucleotides 4351 to 4370, nucleotides 4352 to 4371, nucleotides 4352 to 4374, nucleotides 4353 to 4372, nucleotides 4353 to 4375, nucleotides 4354 to 4376, nucleotides 4359 to 4381, nucleotides 4361 to 4383, nucleotides 4362 to 4384, nucleotides 4362 to4384, nucleotides 4364 to 4386, nucleotides 4365 to 4387, nucleotides 4395 to 4415, nucleotides 4396 to 4416, nucleotides 4409 to 4431, nucleotides 4410 to 4432, nucleotides 4412 to 4434, nucleotides 4637 to 4657, nucleotides 4638 to 4658, nucleotides 4640 to 4660, nucleotides 5103 to 5123, nucleotides 5124 to 5144, nucleotides 5126 to 5146, or nucleotides 5127 to 5147 of SEQ ID NO: 1.
5. The ASO of any one of the preceding claims, wherein the compound includes a second strand of 15 to 60 nucleobases in length and complementary to said first strand
6. The ASO of any one of claims 2-4-, wherein the compound is an antisense oligonucleotide.
7. The ASO of claim 6 having at least one modified internucleoside linkage, sugar moiety, or nucleobase.
8. The ASO of claim 7, wherein the modified intemucleoside linkage comprises a phosphorothi oate linkage.
9. The ASO of claim 8, wherein the at least one modified internucleoside linkage, sugar moiety, or nucleobase comprises a modification selected from the group consisting of a deoxynucleoside, a 3 ’-terminal deoxy-thymine (d'T) nucleoside, a 2'-O-methyl (2'-0Me) modified nucleoside, a 2'-fluoro (2'-F) modified nucleoside, a 2'-deoxy-modified nucleoside, a locked nucleotide (LNA), an unlocked nucleoside, a conformationally restricted nucleoside, a constrained ethyl nucleoside, an abasic nucleoside, a 2'-O,4'-C-ethylene-bridged nucleic acid (ENA), a 2’- amino-modified nucleoside, a2’-O-allyl-modified nucleoside, 2’-C-alkyl-modified nucleoside, 2’- hydroxly-modified nucleoside, a 2’-methoxyethyl (2 -MOE) modified nucleoside, a 2’-O- alkylmodified nucleoside, a morpholino nucleoside, a phosphoramidate, a non-natural base comprising nucleoside, a tetrahydropyran modified nucleoside, a 1,5-anhydrohexitol modified nucleoside, a cyclohexenyl modified nucleoside, a nucleotide comprising a 5'-phosphorothioate group, a nucleotide comprising a 5’-methylphosphonate group, a nucleotide comprising a 5’ phosphate or 5’ phosphate mimic, a nucleotide comprising vinyl phosphonate, a nucleoside comprising adenosine-glycol nucleic acid (GNA), a nucleoside comprising thymidine-glycol nucleic acid (GNA) S-Isomer, a nucleotide comprising 2-hydroxymetbyl-tetrahydrofurane-5- phosphate, a nucleotide comprising 2’deoxythymidine-3 ’phosphate, a nucleotide comprising 2 deoxyguanosine-3 ’-phosphate, and a terminal nucleoside linked to a cholesteryl derivative and a dodecanoic acid bisdecylamide group; and combinations thereof.
10. The ASO of claim 7 wherein the modified nucleobase comprises 5-methylcytosine.
11. A double stranded ribonucleic acid (dsRNA) agent for inhibiting expression of human solute carrier family 6 member 19 (SLC6A19), wherein said dsRNA agent comprises a sense strand and an antisense strand forming a double stranded region, wherein the antisense strand comprises a region of complementarity to a SLC6A19 RNA, and wherein the region of complementarity comprises at least 15 contiguous nucleotides differing by no more than 3 nucleotides from any one of the antisense nucleotide sequences of Tables 7-11, 14 or 17.
12. A double stranded ribonucleic acid (dsRNA) agent for inhibiting expression of human solute carrier family 6 member 19 (SLC6A19) in a cell, wherein said 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 SEQ ID NO: 1.
13. The dsRNA agent of claim I I or 12, 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 3 to 25, nucleotides 8 to 30, nucleotides 13 to 35, nucleotides 38 to 60, nucleotides 43 to 65, nucleotides 48 to 70, nucleotides 93 to 115, nucleotides 98 to 120, nucleotides 103 to 125, nucleotides 108 to 130, nucleotides 111 to 133, nucleotides 113 to 135, nucleotides
118 to 140, nucleotides 214 to 236, nucleotides 219 to 241, nucleotides 244 to 266, nucleotides
249 to 271, nucleotides 252 to 274, nucleotides 252 to 274, nucleotides 253 to 275, nucleotides
254 to 276, nucleotides 257 to 286, nucleotides 259 to 281, nucleotides 264 to 286, nucleotides
289 to 311, nucleotides 294 to 316, nucleotides 339 to 361, nucleotides 397 to 419, nucleotides
460 to 480, nucleotides 461 to 481, nucleotides 481 to 503, nucleotides 486 to 508, nucleotides
491 to 513, nucleotides 557 to 579, nucleotides 561 to 583, nucleotides 562 to 584, nucleotides
564 to 586, nucleotides 566 to 586, nucleotides 567 to 589, nucleotides 616 to 638, nucleotides
621 to 643, nucleotides 626 to 648, nucleotides 670 to 692, nucleotides 671 to 691, nucleotides
672 to 694, nucleotides 674 to 694, nucleotides 675 to 697, nucleotides 680 to 702, nucleotides
685 to 707, nucleotides 690 to 712, nucleotides 695 to 717, nucleotides 700 to 722, nucleotides
705 to 727, nucleotides 710 to 732, nucleotides 715 to 737, nucleotides 720 to 742, nucleotides
725 to 747, nucleotides 730 to 752, nucleotides 735 to 757, nucleotides 740 to 762, nucleotides
745 to 767, nucleotides 750 to 772, nucleotides 751 to 776, nucleotides 755 to 777, nucleotides
760 to 782, nucleotides 765 to 787, nucleotides 770 to 792, nucleotides 775 to 797, nucleotides
798 to 823, nucleotides 800 to 822, nucleotides 825 to 847, nucleotides 890 to 912, nucleotides
895 to 917, nucleotides 900 to 922, nucleotides 905 to 927, nucleotides 910 to 932, nucleotides
911 to 937, nucleotides 915 to 937, nucleotides 972 to 994, nucleotides 973 to 995, nucleotides
977 to 1000, nucleotides 977 to 999, nucleotides 978 to 1000, nucleotides 983 to 1005, nucleotides 1035 to 1057, nucleotides 1058 to 1080, nucleotides 1059 to 1081, nucleotides 1060 to 1082, nucleotides 1063 to 1082, nucleotides 1065 to 1087, nucleotides 1068 to 1087, nucleotides 1070 to 1094, nucleotides 1070 to 1092, nucleotides 1074 to 1096, nucleotides 1075 to 1097, nucleotides 1077 to 1096, nucleotides 1080 to 1102, nucleotides 1085 to 1107, nucleotides 1086 to 1106, nucleotides 1090 to 1112, nucleotides 1095 to 1117, nucleotides 1100 to 1122, nucleotides 1105 to 1127, nucleotides 1110 to 1132, nucleotides 1115 to 1137, nucleotides 1115 to 1137, nucleotides 1117 to 1139, nucleotides 1177 to 1199, nucleotides 1182 to 1204, nucleotides 1187 to 1209, nucleotides 1191 to 1214, nucleotides 1192 to 1214, nucleotides 1195 to 1217, nucleotides 1234 to 1256, nucleotides 1239 to 1261, nucleotides 1242 to 1264, nucleotides 1244 to 1266, nucleotides 1247 to 1268, nucleotides 1249 to 1271, nucleotides 1251 to 1270, nucleotides 1254 to 1276, nucleotides 1259 to 1281, nucleotides 1263 to 1282, nucleotides 1264 to 1286, nucleotides 1266 to 1285, nucleotides 1269 to 1291, nucleotides 1326 to 1348, nucleotides 1331 to 1353, nucleotides 1334 to 1361, nucleotides 1334 to 1356, nucleotides 1334 to 1356, nucleotides 1335 to 1357, nucleotides 1336 to 1358, nucleotides 1337 to 1359, nucleotides 1337 to 1359, nucleotides 1338 to 1360, nucleotides 1340 to 1362, nucleotides 1341 to 1363, nucleotides 1346 to 1368, nucleotides 1351 to 1373, nucleotides 1353 to 1372, nucleotides 1377 to 1399, nucleotides 1382 to 1404, nucleotides 1383 to 1403, nucleotides 1387 to 1409, nucleotides 1392 to 1414, nucleotides 1397 to 1419, nucleotides 1402 to 1424, nucleotides 1407 to 1429, nucleotides 1412 to 1434, nucleotides 1417 to 1439, nucleotides 1456 to 1478, nucleotides 1458 to 1477, nucleotides 1481 to 1503, nucleotides 1486 to 1508, nucleotides 1491 to 1513, nucleotides 1496 to 1518, nucleotides 1521 to 1543, nucleotides 1568 to 1590, nucleotides 1569 to 1591, nucleotides 1574 to 1596, nucleotides 1578 to 1621, nucleotides 1579 to 1601, nucleotides 1583 to 1605, nucleotides 1583 to 1605, nucleotides 1584 to 1606, nucleotides 1584 to 1606, nucleotides 1585 to 1607, nucleotides 1585 to 1607, nucleotides 1587 to 1609, nucleotides 1588 to 1610, nucleotides 1589 to 1611, nucleotides 1594 to 1616, nucleotides 1595 to 1617, nucleotides 1599 to 1621, nucleotides 1601 to 1620, nucleotides 1602 to 1624, nucleotides 1642 to 1664, nucleotides 1749 to 1775, nucleotides 1750 to 1772, nucleotides 1755 to 1777, nucleotides 1757 to 1779, nucleotides 1758 to 1780, nucleotides 1760 to 1782, nucleotides 1761 to 1792, nucleotides 1764 to 1783, nucleotides 1765 to 1787, nucleotides 1770 to 1792, nucleotides 1809 to 1831, nucleotides 1810 to 1829, nucleotides 1812 to 1832, nucleotides 1814 to 1836, nucleotides 1852 to 1881, nucleotides 1853 to 1875, nucleotides 1854 to 1876, nucleotides 1858 to 1880, nucleotides 2168 to 2188, nucleotides 2333 to 2353, nucleotides 2454 to 2474, nucleotides 2507 to 2527, nucleotides 2593 to 2613, nucleotides 3244 to 3264, nucleotides 3288 to 3308, nucleotides 3304 to 3324, nucleotides 3332 to 3352, nucleotides 3333 to 3353, nucleotides 3501 to 3523, nucleotides 3595 to 3615, nucleotides 3595 to 3617, nucleotides 3643 to 3671, nucleotides 3646 to 3668, nucleotides 3646 to 3668, nucleotides 3647 to 3669, nucleotides 3648 to 3668, nucleotides 3648 to 3670, nucleotides 3648 to 3670, nucleotides 3649 to 3671, nucleotides 3651 to 3670, nucleotides 3651 to 3673, nucleotides 3651 to 3673, nucleotides 3652 to 3671, nucleotides 3712 to 3733, nucleotides 3713 to 3735, nucleotides 3763 to 3786, nucleotides 3763 to 3785, nucleotides 3764 to 3786, nucleotides 3791 to 3811, nucleotides 3796 to 3816, nucleotides 3801 to 3829, nucleotides 3801 to 3820, nucleotides 3802 to 3821, nucleotides 3804 to 3826, nucleotides 3809 to 3831, nucleotides 3810 to 3832, nucleotides 3812 to 3833, nucleotides 3812 to 3834, nucleotides 3813 to 3835, nucleotides 3814 to 3833, nucleotides 3814 to 3836, nucleotides 3814 to 3836, nucleotides 3839 to 3859, nucleotides 3840 to 3862, nucleotides 3902 to 3924, nucleotides 3965 to 3987, nucleotides 3995 to 4015, nucleotides 3998 to 4020, nucleotides 4100 to 4124, nucleotides 4102 to 4124, nucleotides 4141 to 4163, nucleotides 4141 to 4163, nucleotides 4142 to 4164, nucleotides 4143 to 4165, nucleotides 4144 to 4166, nucleotides 4146 to 4168, nucleotides 4150 to 4172, nucleotides 4151 to 4173, nucleotides 4192 to 4214, nucleotides 4197 to 4219, nucleotides 4201 to 4223, nucleotides 4265 to 4287, nucleotides 4270 to 4292, nucleotides 4318 to 4338, nucleotides 4319 to 4341, nucleotides 4329 to 4349, nucleotides 4348 to 4386, nucleotides 4349 to 4371, nucleotides 4350 to 4369, nucleotides 4350 to 4372, nucleotides 4350 to 4372, nucleotides 4351 to 4370, nucleotides 4352 to 4371, nucleotides 4352 to 4374, nucleotides 4353 to 4372, nucleotides 4353 to 4375, nucleotides 4354 to 4376, nucleotides 4359 to 4381, nucleotides 4361 to 4383, nucleotides 4362 to 4384, nucleotides 4362 to 4384, nucleotides 4364 to 4386, nucleotides 4365 to 4387, nucleotides 4395 to 4415, nucleotides 4396 to 4416, nucleotides 4409 to 4431, nucleotides 4410 to 4432, nucleotides 4412 to 4434, nucleotides 4637 to 4657, nucleotides 4638 to 4658, nucleotides 4640 to 4660, nucleotides 5103 to 5123, nucleotides 5124 to 5144, nucleotides 5126 to 5146, or nucleotides 5127 to 5147 of SEQ ID NO: 1, and the antisense strand comprises at least 15 contiguous nucleotides from the corresponding nucleotide sequence of SEQ ID NO: 2.
14. The dsRNA agent of any one of claims 11-13, wherein the antisense strand comprises at least 15 contiguous nucleotides differing by not more than three nucleotides from any one of the antisense strand nucleotide sequences of a duplex selected from the group consisting of SEQ ID NOs: 2318 and 2430; SEQ ID NOs: 2319 and 2431 ; SEQ ID NOs: 2320 and 2432; SEQ ID NOs: 2321 and 2433; SEQ ID NOs: 2322 and 2434; SEQ ID NOs: 2323 and 2435; SEQ ID NOs: 2324 and 2436; SEQ ID NOs: 2325 and 2437; SEQ ID NOs: 2326 and 2438; SEQ ID NOs: 2327 and 2439; SEQ ID NOs: 2328 and 2440; SEQ ID NOs: 2329 and 2441; SEQ ID NOs: 2330 and 2442; SEQ ID NOs: 2331 and 2443; SEQ ID NOs: 2332 and 2444; SEQ ID NOs: 2333 and 2445; SEQ ID NOs: 2334 and 2446; SEQ ID NOs: 2335 and 2447; SEQ ID NOs: 2336 and 2448; SEQ ID NOs: 2337 and 2449; SEQ ID NOs: 2338 and 2450; SEQ ID NOs: 2339 and 2451; SEQ ID NOs: 2340 and 2452; SEQ ID NOs: 2341 and 2453; SEQ ID NOs: 2342 and 2454; SEQ ID NOs: 2343 and 2455; SEQ ID NOs: 2344 and 2456; SEQ ID NOs: 2345 and 2457; SEQ ID NOs: 2346 and 2458; SEQ ID NOs: 2347 and 2459; SEQ ID NOs: 2348 and 2460; SEQ ID NOs: 2349 and 2461; SEQ ID NOs: 2350 and 2462; SEQ ID NOs: 2351 and 2463; SEQ ID NOs: 2352 and 2464; SEQ ID NOs: 2353 and 2465; SEQ ID NOs: 2354 and 2466; SEQ ID NOs: 2355 and 2467; SEQ ID NOs: 2356 and 2468; SEQ ID NOs: 2357 and 2469; SEQ ID NOs: 2358 and 2470; SEQ ID NOs: 2359 and 2471; SEQ ID NOs: 2360 and 2472; SEQ ID NOs: 2361 and 2473; SEQ ID NOs: 2362 and 2474; SEQ ID NOs: 2363 and 2475; SEQ ID NOs: 2364 and 2476; SEQ ID NOs: 2365 and 2477; SEQ ID NOs: 2366 and 2478; SEQ ID NOs: 2367 and 2479; SEQ ID NOs: 2368 and 2480; SEQ ID NOs: 2369 and 2481; SEQ ID NOs: 2370 and 2482; SEQ ID NOs: 2371 and 2483; SEQ ID NOs: 2372 and 2484; SEQ ID NOs: 2373 and 2485; SEQ ID NOs: 2374 and 2486; SEQ ID NOs: 2375 and 2487; SEQ ID NOs: 2376 and 2488; SEQ ID NOs: 2377 and 2489; SEQ ID NOs: 2378 and 2490; SEQ ID NOs: 2379 and 2491; SEQ ID NOs: 2380 and 2492; SEQ ID NOs: 2381 and 2493; SEQ ID NOs: 2382 and 2494; SEQ ID NOs: 2383 and 2495; SEQ ID NOs: 2384 and 2496; SEQ ID NOs: 2385 and 2497; SEQ ID NOs: 2386 and 2498; SEQ ID NOs: 2387 and 2499; SEQ ID NOs: 2388 and 2500; SEQ ID NOs: 2389 and 2501; SEQ ID NOs: 2390 and 2502; SEQ ID NOs: 2391 and 2503; SEQ ID NOs: 2392 and 2504; SEQ ID NOs: 2393 and 2505; SEQ ID NOs: 2394 and 2506; SEQ ID NOs: 2395 and 2507; SEQ ID NOs: 2396 and 2508; SEQ ID NOs: 2397 and 2509; SEQ ID NOs: 2398 and 2510; SEQ ID NOs: 2399 and 2511; SEQ ID NOs: 2400 and 2512; SEQ ID NOs: 2401 and 2513; SEQ ID NOs: 2402 and 2514; SEQ ID NOs: 2403 and 2515; SEQ ID NOs: 2404 and 2516; SEQ ID NOs: 2405 and 2517; SEQ ID NOs: 2406 and 2518; SEQ ID NOs: 2407 and 2519; SEQ ID NOs: 2408 and 2520; SEQ ID NOs: 2409 and 2521; SEQ ID NOs: 2410 and 2522; SEQ ID NOs: 2411 and 2523; SEQ ID NOs: 2412 and 2524; SEQ ID NOs: 2413 and 2525; SEQ ID NOs: 2414 and 2526; SEQ ID NOs: 2415 and 2527; SEQ ID NOs: 2416 and 2528; SEQ ID NOs: 2417 and 2529; SEQ ID NOs: 2418 and 2530; SEQ ID NOs: 2419 and 2531; SEQ ID NOs: 2420 and 2532; SEQ ID NOs: 2421 and 2533; SEQ ID NOs: 2422 and 2534; SEQ ID NOs: 2423 and 2535; SEQ ID NOs: 2424 and 2536; SEQ ID NOs: 2425 and 2537; SEQ ID NOs: 2426 and 2538; SEQ ID NOs: 2427 and 2539; SEQ ID NOs: 2428 and 2540; SEQ ID NOs: 2429 and 2541; SEQ ID NO: 2720 and 2840; SEQ ID NO: 2721 and 2841; SEQ ID NO: 2722 and 2842; SEQ ID NO: 2723 and 2843; SEQ ID NO: 2724 and 2844; SEQ ID NO: 2725 and 2845; SEQ ID NO: 2726 and 2846; SEQ ID NO: 2727 and 2847; SEQ ID NO: 2728 and 2848; SEQ ID NO: 2729 and 2849; SEQ ID NO: 2730 and 2850; SEQ ID NO: 2731 and 2851; SEQ ID NO: 2732 and 2852; SEQ ID NO: 2733 and 2853; SEQ ID NO: 2734 and 2854; SEQ ID NO: 2735 and 2855; SEQ ID NO: 2736 and 2856; SEQ ID NO: 2737 and 2857; SEQ ID NO: 2738 and 2858; SEQ ID NO: 2739 and 2859; SEQ ID NO: 2740 and 2860; SEQ ID NO: 2741 and 2861; SEQ ID NO: 2742 and 2862; SEQ ID NO: 2743 and 2863; SEQ ID NO: 2744 and 2864; SEQ ID NO: 2745 and 2865; SEQ ID NO: 2746 and 2866; SEQ ID NO: 2747 and 2867; SEQ ID NO: 2748 and 2868; SEQ ID NO: 2749 and 2869; SEQ ID NO: 2750 and 2870; SEQ ID NO: 2751 and 2871; SEQ ID NO: 2752 and 2872; SEQ ID NO: 2753 and 2873; SEQ ID NO: 2754 and 2874; SEQ ID NO: 2755 and 2875; SEQ ID NO: 2756 and 2876; SEQ ID NO: 2757 and 2877; SEQ ID NO: 2758 and 2878; SEQ ID NO: 2759 and 2879; SEQ ID NO: 2760 and 2880; SEQ ID NO: 2761 and 2881; SEQ ID NO: 2762 and 2882; SEQ ID NO: 2763 and 2883; SEQ ID NO: 2764 and 2884; SEQ ID NO: 2765 and 2885; SEQ ID NO: 2766 and 2886; SEQ ID NO: 2767 and 2887; SEQ ID NO: 2768 and 2888; SEQ ID NO: 2769 and 2889; SEQ ID NO: 2770 and 2890; SEQ ID NO: 2771 and 2891; SEQ ID NO: 2772 and 2892; SEQ ID NO: 2773 and 2893; SEQ ID NO: 2774 and 2894; SEQ ID NO: 2775 and 2895; SEQ ID NO: 2776 and 2896; SEQ ID NO: 2777 and 2897; SEQ ID NO: 2778 and 2898; SEQ ID NO: 2779 and 2899; SEQ ID NO: 2960 and 3340; SEQ ID NO: 2961 and 3341; SEQ ID NO: 2962 and 3342; SEQ ID NO: 2963 and 3343; SEQ ID NO: 2964 and 3344; SEQ ID NO: 2965 and 3345; SEQ ID NO: 2966 and 3346; SEQ ID NO: 2967 and 3347; SEQ ID NO: 2968 and 3348; SEQ ID NO: 2969 and 3349; SEQ ID NO: 2970 and 3350; SEQ ID NO: 2971 and 3351; SEQ ID NO: 2972 and 3352; SEQ ID NO: 2973 and 3353; SEQ ID NO: 2974 and 3354; SEQ ID NO: 2975 and 3355; SEQ ID NO: 2976 and 3356; SEQ ID NO: 2977 and 3357; SEQ ID NO: 2978 and 3358; SEQ ID NO: 2979 and 3359; SEQ ID NO: 2980 and 3360; SEQ ID NO: 2981 and 3361; SEQ ID NO: 2982 and 3362; SEQ ID NO: 2983 and 3363; SEQ ID NO: 2984 and 3364; SEQ ID NO: 2985 and 3365; SEQ ID NO: 2986 and 3366; SEQ ID NO: 2987 and 3367; SEQ ID NO: 2988 and 3368; SEQ ID NO: 2989 and 3369; SEQ ID NO: 2990 and 3370; SEQ ID NO: 2991 and 3371 ; SEQ ID NO: 2992 and 3372; SEQ ID NO: 2993 and 3373; SEQ ID NO: 2994 and 3374; SEQ ID NO: 2995 and 3375; SEQ ID NO: 2996 and 3376; SEQ ID NO: 2997 and 3377; SEQ ID NO: 2998 and 3378; SEQ ID NO: 2999 and 3379; SEQ ID NO: 3000 and 3380; SEQ ID NO: 3001 and 3381; SEQ ID NO: 3002 and 3382; SEQ ID NO: 3003 and 3383; SEQ ID NO: 3004 and 3384; SEQ ID NO: 3005 and 3385; SEQ ID NO: 3006 and 3386; SEQ ID NO: 3007 and 3387; SEQ ID NO: 3008 and 3388; SEQ ID NO: 3009 and 3389; SEQ ID NO: 3010 and 3390; SEQ ID NO: 3011 and 3391; SEQ ID NO: 3012 and 3392; SEQ ID NO: 3013 and 3393; SEQ ID NO: 3014 and 3394; SEQ ID NO: 3015 and 3395; SEQ ID NO: 3016 and 3396; SEQ ID NO: 3017 and 3397; SEQ ID NO: 3018 and 3398; SEQ ID NO: 3019 and 3399; SEQ ID NO: 3020 and 3400; SEQ ID NO: 3021 and 3401; SEQ ID NO: 3022 and 3402; SEQ ID NO: 3023 and 3403; SEQ ID NO: 3024 and 3404; SEQ ID NO: 3025 and 3405; SEQ ID NO: 3026 and 3406; SEQ ID NO: 3027 and 3407; SEQ ID NO: 3028 and 3408; SEQ ID NO: 3029 and 3409; SEQ ID NO: 3030 and 3410; SEQ ID NO: 3031 and 3411; SEQ ID NO: 3032 and 3412; SEQ ID NO: 3033 and 3413; SEQ ID NO: 3034 and 3414; SEQ ID NO: 3035 and 3415; SEQ ID NO: 3036 and 3416; SEQ ID NO: 3037 and 3417; SEQ ID NO: 3038 and 3418; SEQ ID NO: 3039 and 3419; SEQ ID NO: 3040 and 3420; SEQ ID NO: 3041 and 3421; SEQ ID NO: 3042 and 3422; SEQ ID NO: 3043 and 3423; SEQ ID NO: 3044 and 3424; SEQ ID NO: 3045 and 3425; SEQ ID NO: 3046 and 3426; SEQ ID NO: 3047 and 3427; SEQ ID NO: 3048 and 3428; SEQ ID NO: 3049 and 3429; SEQ ID NO: 3050 and 3430; SEQ ID NO: 3051 and 3431; SEQ ID NO: 3052 and 3432; SEQ ID NO: 3053 and 3433; SEQ ID NO: 3054 and 3434; SEQ ID NO: 3055 and 3435; SEQ ID NO: 3056 and 3436; SEQ ID NO: 3057 and 3437; SEQ ID NO: 3058 and 3438; SEQ ID NO: 3059 and 3439; SEQ ID NO: 3060 and 3440; SEQ ID NO: 3061 and 3441; SEQ ID NO: 3062 and 3442; SEQ ID NO: 3063 and 3443; SEQ ID NO: 3064 and 3444; SEQ ID NO: 3065 and 3445; SEQ ID NO: 3066 and 3446; SEQ ID NO: 3067 and 3447; SEQ ID NO: 3068 and 3448; SEQ ID NO: 3069 and 3449; SEQ ID NO: 3070 and 3450; SEQ ID NO: 3071 and 3451; SEQ ID NO: 3072 and 3452; SEQ ID NO: 3073 and 3453; SEQ ID NO: 3074 and 3454; SEQ ID NO: 3075 and 3455; SEQ ID NO: 3076 and 3456; SEQ ID NO: 3077 and 3457; SEQ ID NO: 3078 and 3458; SEQ ID NO: 3079 and 3459; SEQ ID NO: 3080 and 3460; SEQ ID NO: 3081 and 3461; SEQ ID NO: 3082 and 3462; SEQ ID NO: 3083 and 3463; SEQ ID NO: 3084 and 3464; SEQ ID NO: 3085 and 3465; SEQ ID NO: 3086 and 3466; SEQ ID NO: 3087 and 3467; SEQ ID NO: 3088 and 3468; SEQ ID NO: 3089 and 3469; SEQ ID NO: 3090 and 3470; SEQ ID NO: 3091 and 3471; SEQ ID NO: 3092 and 3472; SEQ ID NO: 3093 and 3473; SEQ ID NO: 3094 and 3474; SEQ ID NO: 3095 and 3475; SEQ ID NO: 3096 and 3476; SEQ ID NO: 3097 and 3477; SEQ ID NO: 3098 and 3478; SEQ ID NO: 3099 and 3479; SEQ ID NO: 3100 and 3480; SEQ ID NO: 3101 and 3481; SEQ ID NO: 3102 and 3482; SEQ ID NO: 3103 and 3483; SEQ ID NO: 3104 and 3484; SEQ ID NO: 3105 and 3485; SEQ ID NO: 3106 and 3486; SEQ ID NO: 3107 and 3487; SEQ ID NO: 3108 and 3488; SEQ ID NO: 3109 and 3489; SEQ ID NO: 3110 and 3490; SEQ ID NO: 3111 and 3491; SEQ ID NO: 3112 and 3492; SEQ ID NO: 3113 and 3493; SEQ ID NO: 3114 and 3494; SEQ ID NO: 3115 and 3495; SEQ ID NO: 3116 and 3496; SEQ ID NO: 3117 and 3497; SEQ ID NO: 3118 and 3498; SEQ ID NO: 3119 and 3499; SEQ ID NO: 3120 and 3500; SEQ ID NO: 3121 and 3501; SEQ ID NO: 3122 and 3502; SEQ ID NO: 3123 and 3503; SEQ ID NO: 3124 and 3504; SEQ ID NO: 3125 and 3505; SEQ ID NO: 3126 and 3506; SEQ ID NO: 3127 and 3507; SEQ ID NO: 3128 and 3508; SEQ ID NO: 3129 and 3509; SEQ ID NO: 3130 and 3510; SEQ ID NO: 3131 and 3511; SEQ ID NO: 3132 and 3512; SEQ ID NO: 3133 and 3513; SEQ ID NO: 3134 and 3514; SEQ ID NO: 3135 and 3515; SEQ ID NO: 3136 and 3516; SEQ ID NO: 3137 and 3517; SEQ ID NO: 3138 and 3518; SEQ ID NO: 3139 and 3519; SEQ ID NO: 3140 and 3520; SEQ ID NO: 3141 and 3521; SEQ ID NO: 3142 and 3522; SEQ ID NO: 3143 and 3523; SEQ ID NO: 3144 and 3524; SEQ ID NO: 3145 and 3525; SEQ ID NO: 3146 and 3526; SEQ ID NO: 3147 and 3527; SEQ ID NO: 3148 and 3528; and SEQ ID NO: 3149 and 3529.
15. A double stranded ribonucleic acid (dsRNA) agent for inhibiting expression of human solute carrier family 6 member 19 (SLC6A19) in a cell, wherein the dsRNA agent comprises a sense strand and an antisense strand forming a double stranded region, wherein the sense strand comprises at least 15 contiguous nucleotides differing by no more than 0, 1, 2, or 3 nucleotides from any one of the nucleotide sequences of nucleotides 3 to 25, nucleotides 8 to 30, nucleotides 13 to 35, nucleotides 38 to 60, nucleotides 43 to 65, nucleotides 48 to 70, nucleotides 93 to 115, nucleotides 98 to 120, nucleotides 103 to 125, nucleotides 108 to 130, nucleotides 111 to 133, nucleotides 113 to 135, nucleotides 118 to 140, nucleotides 214 to 236, nucleotides 219 to 241, nucleotides 244 to 266, nucleotides 249 to 271, nucleotides 252 to 274, nucleotides 252 to 274, nucleotides 253 to 275, nucleotides 254 to 276, nucleotides 257 to 286, nucleotides 259 to 281, nucleotides 264 to 286, nucleotides 289 to 311, nucleotides 294 to 316, nucleotides 339 to 361, nucleotides 397 to 419, nucleotides 460 to 480, nucleotides 461 to 481, nucleotides 481 to 503, nucleotides 486 to 508, nucleotides 491 to 513, nucleotides 557 to 579, nucleotides 561 to 583, nucleotides 562 to 584, nucleotides 564 to 586, nucleotides 566 to 586, nucleotides 567 to 589, nucleotides 616 to 638, nucleotides 621 to 643, nucleotides 626 to 648, nucleotides 670 to 692, nucleotides 671 to 691, nucleotides 672 to 694, nucleotides 674 to 694, nucleotides 675 to 697, nucleotides 680 to 702, nucleotides 685 to 707, nucleotides 690 to 712, nucleotides 695 to 717, nucleotides 700 to 722, nucleotides 705 to 727, nucleotides 710 to 732, nucleotides 715 to 737, nucleotides 720 to 742, nucleotides 725 to 747, nucleotides 730 to 752, nucleotides 735 to 757, nucleotides 740 to 762, nucleotides 745 to 767, nucleotides 750 to 772, nucleotides 751 to 776, nucleotides 755 to 777, nucleotides 760 to 782, nucleotides 765 to 787, nucleotides 770 to 792, nucleotides 775 to 797, nucleotides 798 to 823, nucleotides 800 to 822, nucleotides 825 to 847, nucleotides 890 to 912, nucleotides 895 to 917, nucleotides 900 to 922, nucleotides 905 to 927, nucleotides 910 to 932, nucleotides 911 to 937, nucleotides 915 to 937, nucleotides 972 to 994, nucleotides 973 to 995, nucleotides 977 to 1000, nucleotides 977 to 999, nucleotides 978 to 1000, nucleotides 983 to 1005, nucleotides 1035 to 1057, nucleotides 1058 to 1080, nucleotides 1059 to 1081, nucleotides 1060 to 1082, nucleotides 1063 to 1082, nucleotides 1065 to 1087, nucleotides 1068 to 1087, nucleotides 1070 to 1094, nucleotides 1070 to 1092, nucleotides 1074 to 1096, nucleotides 1075 to 1097, nucleotides 1077 to 1096, nucleotides 1080 to 1102, nucleotides 1085 to 1107, nucleotides 1086 to 1106, nucleotides 1090 to 1112, nucleotides 1095 to 1117, nucleotides 1100 to 1122, nucleotides 1105 to 1127, nucleotides 1110 to 1132, nucleotides 1115 to 1137, nucleotides 1115 to 1137, nucleotides 1117 to 1139, nucleotides 1177 to 1199, nucleotides 1182 to 1204, nucleotides 1187 to 1209, nucleotides 1191 to 1214, nucleotides 1192 to 1214, nucleotides 1195 to 1217, nucleotides 1234 to 1256, nucleotides 1239 to 1261, nucleotides 1242 to 1264, nucleotides 1244 to 1266, nucleotides 1247 to 1268, nucleotides 1249 to 1271, nucleotides 1251 to 1270, nucleotides 1254 to 1276, nucleotides 1259 to 1281, nucleotides 1263 to 1282, nucleotides 1264 to 1286, nucleotides 1266 to 1285, nucleotides 1269 to 1291, nucleotides 1326 to 1348, nucleotides 1331 to 1353, nucleotides 1334 to 1361, nucleotides 1334 to 1356, nucleotides 1334 to 1356, nucleotides 1335 to 1357, nucleotides 1336 to 1358, nucleotides 1337 to 1359, nucleotides 1337 to 1359, nucleotides 1338 to 1360, nucleotides 1340 to 1362, nucleotides 1341 to 1363, nucleotides 1346 to 1368, nucleotides 1351 to 1373, nucleotides 1353 to 1372, nucleotides 1377 to 1399, nucleotides 1382 to 1404, nucleotides 1383 to 1403, nucleotides 1387 to 1409, nucleotides 1392 to 1414, nucleotides 1397 to 1419, nucleotides 1402 to 1424, nucleotides 1407 to 1429, nucleotides 1412 to 1434, nucleotides 1417 to 1439, nucleotides 1456 to 1478, nucleotides 1458 to 1477, nucleotides 1481 to 1503, nucleotides 1486 to 1508, nucleotides 1491 to 1513, nucleotides 1496 to 1518, nucleotides 1521 to 1543, nucleotides 1568 to 1590, nucleotides 1569 to 1591, nucleotides 1574 to 1596, nucleotides 1578 to 1621, nucleotides 1579 to 1601, nucleotides 1583 to 1605, nucleotides 1583 to 1605, nucleotides 1584 to 1606, nucleotides 1584 to 1606, nucleotides 1585 to 1607, nucleotides 1585 to 1607, nucleotides 1587 to 1609, nucleotides 1588 to 1610, nucleotides 1589 to 1611, nucleotides 1594 to 1616, nucleotides 1595 to 1617, nucleotides 1599 to 1621, nucleotides 1601 to 1620, nucleotides 1602 to 1624, nucleotides 1642 to 1664, nucleotides 1749 to 1775, nucleotides 1750 to 1772, nucleotides 1755 to 1777, nucleotides 1757 to 1779, nucleotides 1758 to 1780, nucleotides 1760 to 1782, nucleotides 1761 to 1792, nucleotides 1764 to 1783, nucleotides 1765 to 1787, nucleotides 1770 to 1792, nucleotides 1809 to 1831, nucleotides 1810 to 1829, nucleotides 1812 to 1832, nucleotides 1814 to 1836, nucleotides 1852 to 1881, nucleotides 1853 to 1875, nucleotides 1854 to 1876, nucleotides 1858 to 1880, nucleotides 2168 to 2188, nucleotides 2333 to 2353, nucleotides 2454 to 2474, nucleotides 2507 to 2527, nucleotides 2593 to 2613, nucleotides 3244 to 3264, nucleotides 3288 to 3308, nucleotides 3304 to 3324, nucleotides 3332 to 3352, nucleotides 3333 to 3353, nucleotides 3501 to 3523, nucleotides 3595 to 3615, nucleotides 3595 to 3617, nucleotides 3643 to 3671, nucleotides 3646 to 3668, nucleotides 3646 to 3668, nucleotides 3647 to 3669, nucleotides 3648 to 3668, nucleotides 3648 to 3670, nucleotides 3648 to 3670, nucleotides 3649 to 3671, nucleotides 3651 to 3670, nucleotides 3651 to 3673, nucleotides 3651 to 3673, nucleotides 3652 to 3671, nucleotides 3712 to 3733, nucleotides 3713 to 3735, nucleotides 3763 to 3786, nucleotides 3763 to 3785, nucleotides 3764 to 3786, nucleotides 3791 to 3811, nucleotides 3796 to 3816, nucleotides 3801 to 3829, nucleotides 3801 to 3820, nucleotides 3802 to 3821, nucleotides 3804 to 3826, nucleotides 3809 to 3831, nucleotides 3810 to 3832, nucleotides 3812 to 3833, nucleotides 3812 to 3834, nucleotides 3813 to 3835, nucleotides 3814 to 3833, nucleotides 3814 to 3836, nucleotides 3814 to 3836, nucleotides 3839 to 3859, nucleotides 3840 to 3862, nucleotides 3902 to 3924, nucleotides 3965 to 3987, nucleotides 3995 to 4015, nucleotides 3998 to 4020, nucleotides 4100 to 4124, nucleotides 4102 to 4124, nucleotides 4141 to 4163, nucleotides 4141 to 4163, nucleotides 4142 to 4164, nucleotides 4143 to 4165, nucleotides 4144 to 4166, nucleotides 4146 to 4168, nucleotides 4150 to 4172, nucleotides 4151 to 4173, nucleotides 4192 to 4214, nucleotides 4197 to 4219, nucleotides 4201 to 4223, nucleotides 4265 to 4287, nucleotides 4270 to 4292, nucleotides 4318 to 4338, nucleotides 4319 to 4341, nucleotides 4329 to 4349, nucleotides 4348 to 4386, nucleotides 4349 to 4371, nucleotides 4350 to 4369, nucleotides 4350 to 4372, nucleotides 4350 to 4372, nucleotides 4351 to 4370, nucleotides 4352 to 4371, nucleotides 4352 to 4374, nucleotides 4353 to 4372, nucleotides 4353 to4375, nucleotides 4354 to 4376, nucleotides 4359 to 4381, nucleotides 4361 to 4383, nucleotides 4362 to 4384, nucleotides 4362 to 4384, nucleotides 4364 to 4386, nucleotides 4365 to 4387, nucleotides 4395 to 4415, nucleotides 4396 to 4416, nucleotides 4409 to 4431, nucleotides 4410 to 4432, nucleotides 4412 to 4434, nucleotides 4637 to 4657, nucleotides 4638 to 4658, nucleotides 4640 to 4660, nucleotides 5103 to 5123, nucleotides 5124 to 5144, nucleotides 5126 to 5146, or nucleotides 5127 to 5147of SEQ ID NO: 1, and the antisense strand comprises at least 19 contiguous nucleotides from the corresponding nucleotide sequence of SEQ ID NO: 2.
16. The dsRNA agent of any one of claims 11-15, wherein: the double stranded region is 17-30 nucleotide pairs in length; the double stranded region is 17-25 nucleotide pairs in length; the double stranded region is 17-23 nucleotide pairs in length, each strand is independently no more than 30 nucleotides in length; the sense strand is 21 nucleotides in length and the antisense strand is 23 nucleotides in length; the region of complementarity is at least 17 nucleotides in length, optionally wherein the region of complementarity is between 19 and 23 nucleotides in length, optionally wherein the region of complementarity is 19 nucleotides in length; and/or at least one strand comprises a 3’ overhang of at least 2 nucleotides.
17. An exon skipping antisense oligonucleotide comprising any of SEQ ID NOs: 2542-2719.
18. The exon skipping antisense oligonucleotide of claim 17 having at least one modified internucleoside linkage, sugar moiety, or nucleobase.
19. The exon skipping antisense oligonucleotide of claim 18, wherein the modified internucleoside linkage comprises a phosphorothioate linkage.
20. The exon skipping antisense oligonucleotide of claim 19, wherein the at least one modified internucleoside linkage, sugar moiety, or nucleobase comprises a modification selected from the group consisting of a deoxy-nucleoside, a 3 ’-terminal deoxy -thymine (dT) nucleoside, a 2 -0- methyl (2'-0Me) modified nucleoside, a 2'-fluoro (2'-F ) modified nucleoside, a 2'-deoxy-modified nucleoside, a locked nucleotide (LNA), an unlocked nucleoside, a conformationally restricted nucleoside, a constrained ethyl nucleoside, an abasic nucleoside, a 2'-O,4'-C-ethylene-bridged nucleic acid (ENA), a 2’-amino-modified nucleoside, a 2’-O-allyl-modified nucleoside, 2’-C- alkyl-modified nucleoside, 2’-hydroxly-modified nucleoside, a 2’-methoxyethyl (2 -MOE) modified nucleoside, a 2’-O-alkylmodified nucleoside, a morpholino nucleoside, a phosphorami date, a non-natural base comprising nucleoside, a tetrahydropyran modified nucleoside, a 1,5-anhydrohexitol modified nucleoside, a cyclohexenyl modified nucleoside, a nucleotide comprising a 5 -phosphorothioate group, a nucleotide comprising a 5'- methylphosphonate group, a nucleotide comprising a 5’ phosphate or 5’ phosphate mimic, a nucleotide comprising vinyl phosphonate, a nucleoside comprising adenosine-glycol nucleic acid (GNA), a nucleoside comprising thymidine-glycol nucleic acid (GNA) S-Isomer, a nucleotide comprising 2-hydroxymethyl-tetrahydrofurane-5-phosphate, a nucleotide comprising 2’deoxythymidine-.3’phosphate, a nucleotide comprising 2 ’-deoxyguanosine-3 ’-phosphate, and a terminal nucleoside linked to a cholesteryl derivative and a dodecanoic acid bisdecylamide group; and combinations thereof, optionally wherein the exon skipping antisense oligonucleotide comprises one or more 2 ’-methoxy ethyl (2-MOE) modified nucleosides, optionally wherein all nucleosides of the exon skipping antisense oligonucleotide are 2’ -methoxy ethyl (2 -MOE) modified nucleosides.
21. The ASO, dsRNA agent, or exon skipping antisense oligonucleotide of any one of the preceding claims, wherein the antisense compound, dsRNA agent, or exon skipping antisense oligonucleotide comprises at least one modified nucleotide.
22. The ASO, dsRNA agent, or exon skipping antisense oligonucleotide of claim 21, wherein substantially all of the nucleotides of the sense strand of the dsRNA agent; or substantially all of the nucleotides of the antisense strand of the antisense compound, exon skipping antisense oligonucleotide or the dsRNA agent comprise a modification; or substantially all of the nucleotides of the sense strand and substantially all of the nucleotides of the antisense strand of the dsRNA agent comprise a modification.
23. The ASO, dsRNA agent, or exon skipping antisense oligonucleotide of claim 21 or claim 22, wherein all of the nucleotides of the sense strand of the dsRNA agent comprise a modification; all of the nucleotides of the antisense strand of the antisense compound, exon skipping antisense oligonucleotide or the dsRNA agent comprise a modification; or all of the nucleotides of the sense strand and all of the nucleotides of the antisense strand of the dsRNA agent comprise a modification.
24. The ASO, dsRNA agent, or exon skipping antisense oligonucleotide of any one of claims 21-23, wherein at least one of the modified nucleotides is selected from the group consisting of a deoxy-nucleotide, a 3 ’-terminal deoxy-thymine (dT) nucleotide, a 2'-O-methyl modified nucleotide, a 2'-fluoro modified nucleotide, a 2 -deoxy-modified nucleotide, a locked nucleotide, an unlocked nucleotide, a conformationally restricted nucleotide, a constrained ethyl nucleotide, an abasic nucleotide, a 2’-amino-modified nucleotide, a 2’-O-allyl-modified nucleotide, 2’-C- alkyl-modified nucleotide, 2’-hydroxly-modified nucleotide, a 2’ -methoxyethyl modified nucleotide, a 2’-O-alkyl-modified nucleotide, a morpholino nucleotide (e.g., a phosphorodiamidate morpholino nucleic acid (PMO)), a phosphorami date, a non-natural base comprising nucleotide, a tetrahydropyran modified nucleotide, a 1,5-anhydrohexitol modified nucleotide, a cyclohexenyl modified nucleotide, a nucleotide comprising a phosphorothioate group, a nucleotide comprising a phosphoryl guanidine-based backbone, a nucleotide comprising a methylphosphonate group, a nucleotide comprising a 5’-phosphate, a nucleotide comprising a 5’-phosphate mimic, a thermally destabilizing nucleotide, a glycol modified nucleotide (GNA), and a 2-O-(N-methylacetamide) modified nucleotide; and combinations thereof.
25. The ASO, dsRNA agent, or exon skipping antisense oligonucleotide of any one of claims 21-24, wherein the modifications on the nucleotides are selected from the group consisting of:
LNA, HNA, CeNA, 2 -methoxy ethyl, 2 -O-alkyl, 2'-O-allyl, 2'-C- allyl, 2 '-fluoro, 2'- deoxy, 2’ -hydroxyl, and glycol; and combinations thereof; a C7-modified deaza-adenine, a C7-modified deaza-guanosine, a C5-modified cytosine, a C5-modified uridine, Nl-methyl-pseudouridine (ml\|/), 1-ethyl-pseudouridine (ely), 5-methoxy- uridine (mo5U), 5-methyl-cytidine (m5C), pseudouridine (\|/), 5-methoxymethyl uridine, 5- methylthio uridine, 1 -methoxymethyl pseudouridine, 5-methyl cytidine, 5-methoxy cytidine, or any combination thereof; a phosphorothioate (PS) modification, a phosphoryl guanidine (PN) modification, a borano-phosphate modification, an alkyl phosphonate nucleic acid (phNA), a peptide nucleic acid (PNA), or any combination thereof; a deoxyribonucleic acid (DNA), optionally wherein the DNA is or comprises a DNA analog, optionally wherein the DNA analog comprises one or more morpholino subunits linked together by phosphorus-containing linkage(s), optionally wherein the DNA analog is or comprises a phosphorodiamidate morpholino nucleic acid (PMO), optionally wherein the PMO comprises about 12-40 nucleotides; a peptide nucleic acid (PNA) modification; and/or one or more modification to a 5’ end of the antisense compound or dsRNA agent, optionally wherein the modification to the 5' end is a 5’ amino modification.
26. The ASO, dsRNA agent, or exon skipping antisense oligonucleotide of any one of claims 21-25, wherein at least one of the modified nucleotides is selected from the group consisting of a deoxy-nucleotide, a 2'-O-methyl modified nucleotide, a 2'-fhioro modified nucleotide, a 2'-deoxy- modified nucleotide, a glycol modified nucleotide (GNA), and a vinyl-phosphonate nucleotide; and combinations thereof.
27. The ASO, dsRNA agent, or exon skipping antisense oligonucleotide of any one of claims 21-26, wherein at least one of the modified nucleotides is selected from the group consisting of a deoxy-nucleotide, a 2'-O-methyl modified nucleotide, and a 2'-fluoro modified nucleotide; and combinations thereof.
28. The ASO, dsRNA agent, or exon skipping antisense oligonucleotide of any one of the preceding claims comprising one or more deoxy-nucleotides, optionally wherein a heteroduplex oligonucleotide (HDO) comprises the antisense compound or the exon skipping antisense oligonucleotide and/or the dsRNA agent is a HDO.
29. The ASO, dsRNA agent, or exon skipping antisense oligonucleotide of any one of the preceding claims, directly or indirectly conjugated to a targeting moiety.
30. The ASO, dsRNA agent, or exon skipping antisense oligonucleotide of claim 29, wherein the antisense compound, exon skipping antisense oligonucleotide or dsRNA agent and the targeting moiety are indirectly conjugated by way of a linker.
31. The ASO, dsRNA agent, or exon skipping antisense oligonucleotide of claim 29 or claim 30, wherein the targeting moiety specifically binds a cell surface factor, optionally a kidney cell surface factor.
32. The ASO, dsRNA agent, or exon skipping antisense oligonucleotide of claim 31, wherein the cell surface factor is internalized when bound by the targeting moiety.
33. The ASO, dsRNA agent, or exon skipping antisense oligonucleotide of claim 31 or claim 32, wherein the kidney cell surface factor is a receptor, optionally megalin or cubilin.
24. The ASO, dsRNA agent, or exon skipping antisense oligonucleotide of any one of claims 29-33, wherein the targeting moiety is chosen from: a polypeptide, an aminoglycoside, an endogenous ligand (e.g., a ligand disclosed in Table 1 or Table 2), a xenobiotic, an antibody or a fragment thereof, an aptamer, a small molecule, or any combination thereof.
35. The ASO, dsRNA agent, or exon skipping antisense oligonucleotide of claim 34, wherein the targeting moiety is or comprises an endogenous ligand, e.g., a ligand disclosed in Table 1 or Table 2.
36. The ASO, dsRNA agent, or exon skipping antisense oligonucleotide of any one of claims 29-35, wherein the targeting moiety is or comprises a vitamin, optionally a vitamin provided in Table 1, optionally wherein the vitamin is or comprises vitamin B12.
37. The ASO, dsRNA agent, or exon skipping antisense oligonucleotide of claim 36, wherein the targeting moiety is or comprises a polypeptide.
38. The ASO, dsRNA agent, or exon skipping antisense oligonucleotide of claim 37, wherein the polypeptide is chosen from: a fragment of receptor associated protein (RAP), a peptide derived from a radiopharmaceutical conjugate such as ocreotide, ocreotate, exendin, minigastrin, and/or neurotensin; or any combination thereof.
39. The ASO, dsRNA agent, or exon skipping antisense oligonucleotide of claim 38, wherein the polypeptide comprises a RAP fragment, or a variant thereof, optionally wherein the RAP fragment comprises a polypeptide comprising residues 219-323 of RAP.
40. The ASO, dsRNA agent, or exon skipping antisense oligonucleotide of claim 37, wherein the polypeptide is or comprises a peptide derived from a radiopharmaceutical conjugate such as ocreotide, ocreotate, exendin, minigastrin, and/or neurotensin.
41. The ASO, dsRNA agent, or exon skipping anti sense oligonucleotide of any one of claims 37-40, wherein the polypeptide is or comprises a knotted peptide.
42. The ASO, dsRNA agent, or exon skipping antisense oligonucleotide of claim 29 or claim 30, wherein the targeting moiety is or comprises an aminoglycoside, optionally wherein the aminoglycoside is chosen from one or more, or all of: streptomycin, neomycin, kanamycin, paromomycin, gentamicin, G-418 (geneticin) ELX-202, tobramycin, amikacin, netilmicin, spectinomycin, sisomicin, dibekacin, isepamicin, framycetin, paromomycin, apramycin, fradiomycin, arbekacin, plazomicin, or a derivative, or a fragment, or a variant thereof.
43. The ASO, dsRNA agent, or exon skipping antisense oligonucleotide of claim 29 or claim 30, wherein the targeting moiety is or comprises a xenobiotic, optionally wherein the xenobiotic is or comprises polymixin, aprotinin, trichosanthin, or any combination thereof.
44. The ASO, dsRNA agent, or exon skipping antisense oligonucleotide of claim 29 or claim 30, wherein the targeting moiety is or comprises an antibody or a fragment thereof, optionally wherein the antibody or fragment thereof selectively binds Megalin, Cubilin, or both.
45. The ASO, dsRNA agent, or exon skipping antisense oligonucleotide of claim 44, wherein the antibody or fragment thereof specifically binds Megalin.
46. The ASO, dsRNA agent, or exon skipping antisense oligonucleotide of claim 44, wherein the antibody or fragment thereof specifically binds Cubilin.
47. The ASO, dsRNA agent, or exon skipping antisense oligonucleotide of any one of claims 43-46, wherein the antibody or fragment thereof is a bispecific antibody or a multi-specific antibody.
48. The ASO, dsRNA agent, or exon skipping antisense oligonucleotide of any one of claims 42-46, wherein the antibody comprises one or more modifications of an Fc domain, e.g., an Fc variant.
49. The ASO, dsRNA agent, or exon skipping antisense oligonucleotide of any one of the preceding claims, wherein the antisense compound or dsRNA agent is characterized in that when delivered to a cell expressing the target, reduced expression and/or activity of the target is observed as compared to a cell which has not been delivered the antisense compound, dsRNA agent, or exon skipping antisense oligonucleotide or to a cell which does not express the target.
50. The ASO, dsRNA agent, or exon skipping antisense oligonucleotide of any one of claims 30-49, wherein the linker is a cleavable linker, optionally wherein the linker becomes cleaved when exposed to a cell-internal environment.
51. The ASO, dsRNA agent, or exon skipping antisense oligonucleotide of any one of the preceding claims, wherein the antisense compound, dsRNA agent, or exon skipping antisense oligonucleotide comprises one or more extended nucleic acid ("exNA") modifications, optionally wherein the one or more exNA modification(s) is/are positioned at or near a 3'-end of the nucleic acid.
52. The ASO, dsRNA agent, or exon skipping antisense oligonucleotide of any one of the preceding claims, wherein the antisense compound, dsRNA agent, or exon skipping antisense oligonucleotide comprises one or more phosphoryl guanidine-containing backbone ("PN backbone") and/or mesyl phosphoramidate modifications.
53. A pharmaceutical composition comprising the ASO, dsRNA agent, or exon skipping antisense oligonucleotide of any one of the preceding claims and a pharmaceutically acceptable penetration enhancer, carrier, or diluent, optionally wherein the pharmaceutical composition is formulated for intravenous, subcutaneous, intramuscular, parenteral, or oral delivery
54. A method for the prevention, amelioration, or treatment of a kidney disease or condition in a subject comprising administration of the ASO, dsRNA, exon skipping antisense oligonucleotide, or pharmaceutical composition of any one of the preceding claims to the subject in need of such intervention.
55. A method for the prevention, amelioration, or treatment of a kidney disease or condition in a subject comprising administration of an inhibitory ASO, exon skipping antisense oligonucleotide, or dsRNA to the subject in need of such intervention, wherein the inhibitory' antisense compound, exon skipping antisense oligonucleotide, or dsRNA targets the human SLC6A19 m RN A of SEQ ID NO: 1 .
56. The method of claim 54 or claim 55, wherein the kidney disease or condition is a glomerular disorder, a renal tubular disorder, Chronic Kidney Disease, other renal disorders, an inborn error of metabolism, a systemic metabolic disorder, a disorder of the thyroid, a disorder of the parathyroid, a disorder of the inner ear, a neurological disorder, or a viral infection, or any combination thereof.
57. The method of claim 55, wherein the kidney disease or condition is phenylketonuria (PKU).
58. The method of claim 55, wherein the kidney disease or condition is a urea cycle disorder.
59. A method for reducing SI..C6 A19 mRNA or B°AT1 polypeptide levels in a cell, the method comprising contacting the cell with an ASO, dsRNA agent, or exon skipping antisense oligonucleotide of any one of claims 2-52 in an amount sufficient to reduce SLC6A19 mRNA or B°AT1 polypeptide levels in the cell.
60. The method of ciaim 59, wherein the cel 1 is chosen from: immune cells; nervous system cells; muscle cells; small intestine cells; colon cells; adipocytes; kidney cells; liver cells; lung cells; splenic cells; stomach cells; esophagus cells; bladder cells; pancreas cells; thyroid cells; salivary gland cells; adrenal gland cells; pituitary gland cells; breast cells; skin cells; ovary cells; uterus cells; placenta cells; prostate cells; parathyroid cells; cells of the inner ear; or testis cells, or any combination thereof, optionally wherein the cell is a kidney cell, optionally wherein the kidney cell is a proximal tubular epithelial cell and/or a podocyte.
61. The method of claim 59 or claim 60, wherein the cell is a mammalian cell, optionally a human cell, optionally a human cell of a subject, optionally a human cell of a subject in situ.
62. Use of an ASO, dsRNA, exon skipping antisense oligonucleotide, or pharmaceutical composition of any one of claims 2-53 in the manufacture of a medicament for the treatment, prevention or amelioration of a kidney disease or condition, wherein the medicament reduces the expression of a nucleic acid molecule encoding B°AT1.
63. The use of claim 62, wherein the kidney disease or condition comprises a glomerular disorder, a renal tubular disorder, Chronic Kidney Disease, other renal disorders, an inborn error of metabolism, a systemic metabolic disorder, a disorder of the thyroid, a disorder of the parathyroid, a disorder of the inner ear, a neurological disorder, or a viral infection, or any combination thereof.
64. The use of claim 62, wherein the kidney disease or condition is PKU.
65. The use of claim 62, wherein the kidney disease or condition is a urea cycle disorder.
66. A kit comprising the ASO, dsRNA, exon skipping antisense oligonucleotide, or pharmaceutical composition of any one of claims 2-53, and instructions for its use.
67. A vial comprising the ASO, dsRNA, exon skipping antisense oligonucleotide, or pharmaceutical composition of any one of claims 2-53.
68. A syringe comprising the ASO, dsRNA, exon skipping antisense oligonucleotide, or pharmaceutical composition of any one of claims 2-53.
PCT/US2024/0488522023-09-272024-09-27Slc6a19-targeting modulatory nucleic acid agentsPendingWO2025072672A2 (en)

Applications Claiming Priority (4)

Application NumberPriority DateFiling DateTitle
US202363585781P2023-09-272023-09-27
US63/585,7812023-09-27
US202463655344P2024-06-032024-06-03
US63/655,3442024-06-03

Publications (2)

Publication NumberPublication Date
WO2025072672A2true WO2025072672A2 (en)2025-04-03
WO2025072672A3 WO2025072672A3 (en)2025-05-08

Family

ID=93117481

Family Applications (1)

Application NumberTitlePriority DateFiling Date
PCT/US2024/048852PendingWO2025072672A2 (en)2023-09-272024-09-27Slc6a19-targeting modulatory nucleic acid agents

Country Status (1)

CountryLink
WO (1)WO2025072672A2 (en)

Citations (162)

* Cited by examiner, † Cited by third party
Publication numberPriority datePublication dateAssigneeTitle
US513030A (en)1894-01-16Machine for waxing or coating paper
US564562A (en)1896-07-21Joseph p
US1280493A (en)1916-05-011918-10-01Stewart S Kurtz JrMetallic piston-packing.
US3687808A (en)1969-08-141972-08-29Univ Leland Stanford JuniorSynthetic polynucleotides
US4469863A (en)1980-11-121984-09-04Ts O Paul O PNonionic nucleic acid alkyl and aryl phosphonates and processes for manufacture and use thereof
US4476301A (en)1982-04-291984-10-09Centre National De La Recherche ScientifiqueOligonucleotides, a process for preparing the same and their application as mediators of the action of interferon
US4683202A (en)1985-03-281987-07-28Cetus CorporationProcess for amplifying nucleic acid sequences
US4845205A (en)1985-01-081989-07-04Institut Pasteur2,N6 -disubstituted and 2,N6 -trisubstituted adenosine-3'-phosphoramidites
US4981957A (en)1984-07-191991-01-01Centre National De La Recherche ScientifiqueOligonucleotides with modified phosphate and modified carbohydrate moieties at the respective chain termini
US5023243A (en)1981-10-231991-06-11Molecular Biosystems, Inc.Oligonucleotide therapeutic agent and method of making same
US5034506A (en)1985-03-151991-07-23Anti-Gene Development GroupUncharged morpholino-based polymers having achiral intersubunit linkages
US5118800A (en)1983-12-201992-06-02California Institute Of TechnologyOligonucleotides possessing a primary amino group in the terminal nucleotide
US5134066A (en)1989-08-291992-07-28Monsanto CompanyImproved probes using nucleosides containing 3-dezauracil analogs
US5142047A (en)1985-03-151992-08-25Anti-Gene Development GroupUncharged polynucleotide-binding polymers
US5166315A (en)1989-12-201992-11-24Anti-Gene Development GroupSequence-specific binding polymers for duplex nucleic acids
US5175273A (en)1988-07-011992-12-29Genentech, Inc.Nucleic acid intercalating agents
US5177195A (en)1991-01-081993-01-05Imperial Chemical Industries PlcDisazo dyes
US5185444A (en)1985-03-151993-02-09Anti-Gene Deveopment GroupUncharged morpolino-based polymers having phosphorous containing chiral intersubunit linkages
US5188897A (en)1987-10-221993-02-23Temple University Of The Commonwealth System Of Higher EducationEncapsulated 2',5'-phosphorothioate oligoadenylates
US5214134A (en)1990-09-121993-05-25Sterling Winthrop Inc.Process of linking nucleosides with a siloxane bridge
US5216141A (en)1988-06-061993-06-01Benner Steven AOligonucleotide analogs containing sulfur linkages
US5217866A (en)1985-03-151993-06-08Anti-Gene Development GroupPolynucleotide assay reagent and method
US5235033A (en)1985-03-151993-08-10Anti-Gene Development GroupAlpha-morpholino ribonucleoside derivatives and polymers thereof
US5264564A (en)1989-10-241993-11-23Gilead SciencesOligonucleotide analogs with novel linkages
US5264423A (en)1987-03-251993-11-23The United States Of America As Represented By The Department Of Health And Human ServicesInhibitors for replication of retroviruses and for the expression of oncogene products
US5276019A (en)1987-03-251994-01-04The United States Of America As Represented By The Department Of Health And Human ServicesInhibitors for replication of retroviruses and for the expression of oncogene products
US5278302A (en)1988-05-261994-01-11University Patents, Inc.Polynucleotide phosphorodithioates
WO1994002595A1 (en)1992-07-171994-02-03Ribozyme Pharmaceuticals, Inc.Method and reagent for treatment of animal diseases
US5319080A (en)1991-10-171994-06-07Ciba-Geigy CorporationBicyclic nucleosides, oligonucleotides, process for their preparation and intermediates
US5321131A (en)1990-03-081994-06-14Hybridon, Inc.Site-specific functionalization of oligodeoxynucleotides for non-radioactive labelling
US5359044A (en)1991-12-131994-10-25Isis PharmaceuticalsCyclobutyl oligonucleotide surrogates
US5367066A (en)1984-10-161994-11-22Chiron CorporationOligonucleotides with selectably cleavable and/or abasic sites
US5399676A (en)1989-10-231995-03-21Gilead SciencesOligonucleotides with inverted polarity
US5405938A (en)1989-12-201995-04-11Anti-Gene Development GroupSequence-specific binding polymers for duplex nucleic acids
US5405939A (en)1987-10-221995-04-11Temple University Of The Commonwealth System Of Higher Education2',5'-phosphorothioate oligoadenylates and their covalent conjugates with polylysine
US5432272A (en)1990-10-091995-07-11Benner; Steven A.Method for incorporating into a DNA or RNA oligonucleotide using nucleotides bearing heterocyclic bases
US5434257A (en)1992-06-011995-07-18Gilead Sciences, Inc.Binding compentent oligomers containing unsaturated 3',5' and 2',5' linkages
US5446137A (en)1993-12-091995-08-29Syntex (U.S.A.) Inc.Oligonucleotides containing 4'-substituted nucleotides
US5445934A (en)1989-06-071995-08-29Affymax Technologies N.V.Array of oligonucleotides on a solid substrate
US5455233A (en)1989-11-301995-10-03University Of North CarolinaOligoribonucleoside and oligodeoxyribonucleoside boranophosphates
US5457187A (en)1993-12-081995-10-10Board Of Regents University Of NebraskaOligonucleotides containing 5-fluorouracil
US5459255A (en)1990-01-111995-10-17Isis Pharmaceuticals, Inc.N-2 substituted purines
US5466677A (en)1993-03-061995-11-14Ciba-Geigy CorporationDinucleoside phosphinates and their pharmaceutical compositions
US5466786A (en)1989-10-241995-11-14Gilead Sciences2'modified nucleoside and nucleotide compounds
US5470967A (en)1990-04-101995-11-28The Dupont Merck Pharmaceutical CompanyOligonucleotide analogs with sulfamate linkages
US5476925A (en)1993-02-011995-12-19Northwestern UniversityOligodeoxyribonucleotides including 3'-aminonucleoside-phosphoramidate linkages and terminal 3'-amino groups
US5484908A (en)1991-11-261996-01-16Gilead Sciences, Inc.Oligonucleotides containing 5-propynyl pyrimidines
US5489677A (en)1990-07-271996-02-06Isis Pharmaceuticals, Inc.Oligonucleoside linkages containing adjacent oxygen and nitrogen atoms
US5502177A (en)1993-09-171996-03-26Gilead Sciences, Inc.Pyrimidine derivatives for labeled binding partners
US5506337A (en)1985-03-151996-04-09Antivirals Inc.Morpholino-subunit combinatorial library and method
US5514785A (en)1990-05-111996-05-07Becton Dickinson And CompanySolid supports for nucleic acid hybridization assays
US5519134A (en)1994-01-111996-05-21Isis Pharmaceuticals, Inc.Pyrrolidine-containing monomers and oligomers
US5519126A (en)1988-03-251996-05-21University Of Virginia Alumni Patents FoundationOligonucleotide N-alkylphosphoramidates
US5521063A (en)1985-03-151996-05-28Antivirals Inc.Polynucleotide reagent containing chiral subunits and methods of use
US5525711A (en)1994-05-181996-06-11The United States Of America As Represented By The Secretary Of The Department Of Health And Human ServicesPteridine nucleotide analogs as fluorescent DNA probes
US5539082A (en)1993-04-261996-07-23Nielsen; Peter E.Peptide nucleic acids
US5541316A (en)1992-02-111996-07-30Henkel Kommanditgesellschaft Auf AktienProcess for the production of polysaccharide-based polycarboxylates
US5541307A (en)1990-07-271996-07-30Isis Pharmaceuticals, Inc.Backbone modified oligonucleotide analogs and solid phase synthesis thereof
US5550111A (en)1984-07-111996-08-27Temple University-Of The Commonwealth System Of Higher EducationDual action 2',5'-oligoadenylate antiviral derivatives and uses thereof
US5552540A (en)1987-06-241996-09-03Howard Florey Institute Of Experimental Physiology And MedicineNucleoside derivatives
US5561225A (en)1990-09-191996-10-01Southern Research InstitutePolynucleotide analogs containing sulfonate and sulfonamide internucleoside linkages
US5567811A (en)1990-05-031996-10-22Amersham International PlcPhosphoramidite derivatives, their preparation and the use thereof in the incorporation of reporter groups on synthetic oligonucleotides
US5571799A (en)1991-08-121996-11-05Basco, Ltd.(2'-5') oligoadenylate analogues useful as inhibitors of host-v5.-graft response
US5576427A (en)1993-03-301996-11-19Sterling Winthrop, Inc.Acyclic nucleoside analogs and oligonucleotide sequences containing them
US5587361A (en)1991-10-151996-12-24Isis Pharmaceuticals, Inc.Oligonucleotides having phosphorothioate linkages of high chiral purity
US5591722A (en)1989-09-151997-01-07Southern Research Institute2'-deoxy-4'-thioribonucleosides and their antiviral activity
US5594121A (en)1991-11-071997-01-14Gilead Sciences, Inc.Enhanced triple-helix and double-helix formation with oligomers containing modified purines
US5596086A (en)1990-09-201997-01-21Gilead Sciences, Inc.Modified internucleoside linkages having one nitrogen and two carbon atoms
US5596091A (en)1994-03-181997-01-21The Regents Of The University Of CaliforniaAntisense oligonucleotides comprising 5-aminoalkyl pyrimidine nucleotides
US5597909A (en)1994-08-251997-01-28Chiron CorporationPolynucleotide reagents containing modified deoxyribose moieties, and associated methods of synthesis and use
US5602240A (en)1990-07-271997-02-11Ciba Geigy Ag.Backbone modified oligonucleotide analogs
US5608046A (en)1990-07-271997-03-04Isis Pharmaceuticals, Inc.Conjugated 4'-desmethyl nucleoside analog compounds
US5610289A (en)1990-07-271997-03-11Isis Pharmaceuticals, Inc.Backbone modified oligonucleotide analogues
US5610300A (en)1992-07-011997-03-11Ciba-Geigy CorporationCarbocyclic nucleosides containing bicyclic rings, oligonucleotides therefrom, process for their preparation, their use and intermediates
US5614617A (en)1990-07-271997-03-25Isis Pharmaceuticals, Inc.Nuclease resistant, pyrimidine modified oligonucleotides that detect and modulate gene expression
US5618704A (en)1990-07-271997-04-08Isis Pharmacueticals, Inc.Backbone-modified oligonucleotide analogs and preparation thereof through radical coupling
US5623070A (en)1990-07-271997-04-22Isis Pharmaceuticals, Inc.Heteroatomic oligonucleoside linkages
US5625050A (en)1994-03-311997-04-29Amgen Inc.Modified oligonucleotides and intermediates useful in nucleic acid therapeutics
US5627053A (en)1994-03-291997-05-06Ribozyme Pharmaceuticals, Inc.2'deoxy-2'-alkylnucleotide containing nucleic acid
US5633360A (en)1992-04-141997-05-27Gilead Sciences, Inc.Oligonucleotide analogs capable of passive cell membrane permeation
US5639873A (en)1992-02-051997-06-17Centre National De La Recherche Scientifique (Cnrs)Oligothionucleotides
US5646265A (en)1990-01-111997-07-08Isis Pharmceuticals, Inc.Process for the preparation of 2'-O-alkyl purine phosphoramidites
US5658873A (en)1993-04-101997-08-19Degussa AktiengesellschaftCoated sodium percarbonate particles, a process for their production and detergent, cleaning and bleaching compositions containing them
US5663312A (en)1993-03-311997-09-02SanofiOligonucleotide dimers with amide linkages replacing phosphodiester linkages
US5670633A (en)1990-01-111997-09-23Isis Pharmaceuticals, Inc.Sugar modified oligonucleotides that detect and modulate gene expression
US5677439A (en)1990-08-031997-10-14SanofiOligonucleotide analogues containing phosphate diester linkage substitutes, compositions thereof, and precursor dinucleotide analogues
US5677195A (en)1991-11-221997-10-14Affymax Technologies N.V.Combinatorial strategies for polymer synthesis
US5677437A (en)1990-07-271997-10-14Isis Pharmaceuticals, Inc.Heteroatomic oligonucleoside linkages
US5681941A (en)1990-01-111997-10-28Isis Pharmaceuticals, Inc.Substituted purines and oligonucleotide cross-linking
US5714331A (en)1991-05-241998-02-03Buchardt, Deceased; OlePeptide nucleic acids having enhanced binding affinity, sequence specificity and solubility
US5719262A (en)1993-11-221998-02-17Buchardt, Deceased; OlePeptide nucleic acids having amino acid side chains
US5744305A (en)1989-06-071998-04-28Affymetrix, Inc.Arrays of materials attached to a substrate
US5750692A (en)1990-01-111998-05-12Isis Pharmaceuticals, Inc.Synthesis of 3-deazapurines
US5770722A (en)1994-10-241998-06-23Affymetrix, Inc.Surface-bound, unimolecular, double-stranded DNA
US5854033A (en)1995-11-211998-12-29Yale UniversityRolling circle replication reporter systems
US5874219A (en)1995-06-071999-02-23Affymetrix, Inc.Methods for concurrently processing multiple biological chip assays
US6015886A (en)1993-05-242000-01-18Chemgenes CorporationOligonucleotide phosphate esters
US6028188A (en)1993-11-162000-02-22Genta IncorporatedSynthetic oligomers having chirally pure phosphonate internucleosidyl linkages mixed with non-phosphonate internucleosidyl linkages
WO2000022113A1 (en)1998-10-092000-04-20Ingene, Inc.ENZYMATIC SYNTHESIS OF ssDNA
WO2000022114A1 (en)1998-10-092000-04-20Ingene, Inc.PRODUCTION OF ssDNA $i(IN VIVO)
US6054299A (en)1994-04-292000-04-25Conrad; Charles A.Stem-loop cloning vector and method
US6124445A (en)1994-11-232000-09-26Isis Pharmaceuticals, Inc.Phosphotriester oligonucleotides, amidities and method of preparation
US6147200A (en)1999-08-192000-11-14Isis Pharmaceuticals, Inc.2'-O-acetamido modified monomers and oligomers
US6160109A (en)1995-10-202000-12-12Isis Pharmaceuticals, Inc.Preparation of phosphorothioate and boranophosphate oligomers
US6166197A (en)1995-03-062000-12-26Isis Pharmaceuticals, Inc.Oligomeric compounds having pyrimidine nucleotide (S) with 2'and 5 substitutions
US6169170B1 (en)1994-03-182001-01-02Lynx Therapeutics, Inc.Oligonucleotide N3′→N5′Phosphoramidate Duplexes
US6172209B1 (en)1997-02-142001-01-09Isis Pharmaceuticals Inc.Aminooxy-modified oligonucleotides and methods for making same
US6222025B1 (en)1995-03-062001-04-24Isis Pharmaceuticals, Inc.Process for the synthesis of 2′-O-substituted pyrimidines and oligomeric compounds therefrom
US6235887B1 (en)1991-11-262001-05-22Isis Pharmaceuticals, Inc.Enhanced triple-helix and double-helix formation directed by oligonucleotides containing modified pyrimidines
US6239265B1 (en)1990-01-112001-05-29Isis Pharmaceuticals, Inc.Oligonucleotides having chiral phosphorus linkages
US6277603B1 (en)1991-12-242001-08-21Isis Pharmaceuticals, Inc.PNA-DNA-PNA chimeric macromolecules
US6326199B1 (en)1991-12-242001-12-04Isis Pharmaceuticals, Inc.Gapped 2′ modified oligonucleotides
US6346614B1 (en)1992-07-232002-02-12Hybridon, Inc.Hybrid oligonucleotide phosphorothioates
US6444423B1 (en)1996-06-072002-09-03Molecular Dynamics, Inc.Nucleosides comprising polydentate ligands
US6528640B1 (en)1997-11-052003-03-04Ribozyme Pharmaceuticals, IncorporatedSynthetic ribonucleic acids with RNAse activity
US6531590B1 (en)1998-04-242003-03-11Isis Pharmaceuticals, Inc.Processes for the synthesis of oligonucleotide compounds
US6534639B1 (en)1999-07-072003-03-18Isis Pharmaceuticals, Inc.Guanidinium functionalized oligonucleotides and method/synthesis
US6608035B1 (en)1994-10-252003-08-19Hybridon, Inc.Method of down-regulating gene expression
US6617438B1 (en)1997-11-052003-09-09Sirna Therapeutics, Inc.Oligoribonucleotides with enzymatic activity
US6639062B2 (en)1997-02-142003-10-28Isis Pharmaceuticals, Inc.Aminooxy-modified nucleosidic compounds and oligomeric compounds prepared therefrom
US6815432B2 (en)1995-06-072004-11-09Inex Pharmaceuticals Corp.Methods for encapsulating plasmids in lipid bilayers
US6858715B2 (en)1999-02-042005-02-22Isis Pharmaceuticals, Inc.Process for the synthesis of oligomeric compounds
US6858225B2 (en)1997-05-142005-02-22Inex Pharmaceuticals CorporationLipid-encapsulated polyanionic nucleic acid
US6867294B1 (en)1998-07-142005-03-15Isis Pharmaceuticals, Inc.Gapped oligomers having site specific chiral phosphorothioate internucleoside linkages
US6878805B2 (en)2002-08-162005-04-12Isis Pharmaceuticals, Inc.Peptide-conjugated oligomeric compounds
US7015315B1 (en)1991-12-242006-03-21Isis Pharmaceuticals, Inc.Gapped oligonucleotides
US7045610B2 (en)1998-04-032006-05-16Epoch Biosciences, Inc.Modified oligonucleotides for mismatch discrimination
WO2006138343A2 (en)2005-06-142006-12-28Raptor Pharmaceutical Inc.Compositions comprising receptor - associated protein (rap) variants specific for lrp2 and uses thereof
US7273933B1 (en)1998-02-262007-09-25Isis Pharmaceuticals, Inc.Methods for synthesis of oligonucleotides
US7321029B2 (en)2000-01-212008-01-22Geron Corporation2′-arabino-fluorooligonucleotide N3′→P5′ phosphoramidates: their synthesis and use
US20080153753A1 (en)2002-04-252008-06-26Thomas WillnowMethod of treating side effects induced by therapeutic agents
US7427605B2 (en)2005-03-312008-09-23Calando Pharmaceuticals, Inc.Inhibitors of ribonucleotide reductase subunit 2 and uses thereof
US7427672B2 (en)2003-08-282008-09-23Takeshi ImanishiArtificial nucleic acids of n-o bond crosslinkage type
US7495088B1 (en)1989-12-042009-02-24Enzo Life Sciences, Inc.Modified nucleotide compounds
WO2009073809A2 (en)2007-12-042009-06-11Alnylam Pharmaceuticals, Inc.Carbohydrate conjugates as delivery agents for oligonucleotides
US7560431B2 (en)2003-06-202009-07-14Raptor Pharmaceutical Inc.Methods of increasing delivery of active agents to brain comprising administering receptor associated protein (RAP) fragments conjugated to active agents
WO2011005861A1 (en)2009-07-072011-01-13Alnylam Pharmaceuticals, Inc.Oligonucleotide end caps
US8058069B2 (en)2008-04-152011-11-15Protiva Biotherapeutics, Inc.Lipid formulations for nucleic acid delivery
US20110313020A1 (en)2008-12-032011-12-22Marina Biotech, Inc.UsiRNA Complexes
US8101348B2 (en)2002-07-102012-01-24Max-Planck-Gesellschaft Zur Foerderung Der Wissenschaften E.V.RNA-interference by single-stranded RNA molecules
US8158601B2 (en)2009-06-102012-04-17Alnylam Pharmaceuticals, Inc.Lipid formulation
US20120157511A1 (en)2009-07-072012-06-21Alnylam Pharmaceuticals, Inc.5' phosphate mimics
US8314227B2 (en)2007-05-222012-11-20Marina Biotech, Inc.Hydroxymethyl substituted RNA oligonucleotides and RNA complexes
US20130011922A1 (en)2007-03-022013-01-10F/K/A Mdrna, Inc.Nucleic acid compounds for inhibiting gene expression and uses thereof
WO2013036868A1 (en)2011-09-072013-03-14Marina Biotech Inc.Synthesis and uses of nucleic acid compounds with conformationally restricted monomers
US20130190383A1 (en)2010-04-262013-07-25Marina Biotech, Inc.Nucleic acid compounds with conformationally restricted monomers and uses thereof
US8795627B2 (en)2007-03-212014-08-05Raptor Pharmaceuticals Inc.Treatment of liver disorders by administration of RAP conjugates
WO2015027205A1 (en)2013-08-222015-02-26Robert DoyleCompositions comprising vitamin b12 and intrinsic factor and methods of use thereof
US9133461B2 (en)2012-04-102015-09-15Alnylam Pharmaceuticals, Inc.Compositions and methods for inhibiting expression of the ALAS1 gene
US9388418B2 (en)2011-06-122016-07-12City Of HopeAptamer-mRNA conjugates for targeted protein or peptide expression and methods for their use
US9422562B2 (en)2010-04-062016-08-23Alnylam PharmaceuticalsCompositions and methods for inhibiting expression of CD274/PD-L1 gene
US9463252B2 (en)2008-03-182016-10-11Seattle Genetics, Inc.Auristatin drug linker conjugates
WO2017100700A2 (en)2015-12-112017-06-15Fred Hutchinson Cancer Research CenterPeptides for renal therapy
US10065993B2 (en)2013-05-072018-09-04Merck Patent GmbhPeptides and peptide/active compound conjugates for renal targeting
US10131907B2 (en)2007-04-232018-11-20Alnylam Pharmaceuticals, Inc.Glycoconjugates of RNA interference agents
WO2018232122A1 (en)2017-06-152018-12-20Blaze Bioscience, Inc.Renal-homing peptide conjugates and methods of use thereof
US10808039B2 (en)2003-11-062020-10-20Seattle Genetics Inc.Monomethylvaline compounds capable of conjugation to ligands
US11071749B2 (en)2011-12-082021-07-27Sarepta Therapeutics, Inc.Oligonucleotide analogues targeting human LMNA
WO2021195533A2 (en)2020-03-262021-09-30University Of MassachusettsSynthesis of modified oligonucleotides with increased stability
US11208430B2 (en)2014-08-222021-12-28Noogen LlcModified oligonucleotides and methods for their synthesis
WO2023278589A1 (en)2021-06-302023-01-05Ionis Pharmaceuticals, Inc.Method for synthesis of linkage modified oligomeric compounds
WO2023201095A2 (en)2022-04-152023-10-19Wave Life Sciences Ltd.Oligonucleotide compositions and methods relating thereto

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication numberPriority datePublication dateAssigneeTitle
WO2013173637A1 (en)*2012-05-162013-11-21Rana Therapeutics, Inc.Compositions and methods for modulating gene expression

Patent Citations (181)

* Cited by examiner, † Cited by third party
Publication numberPriority datePublication dateAssigneeTitle
US564562A (en)1896-07-21Joseph p
US513030A (en)1894-01-16Machine for waxing or coating paper
US1280493A (en)1916-05-011918-10-01Stewart S Kurtz JrMetallic piston-packing.
US3687808A (en)1969-08-141972-08-29Univ Leland Stanford JuniorSynthetic polynucleotides
US4469863A (en)1980-11-121984-09-04Ts O Paul O PNonionic nucleic acid alkyl and aryl phosphonates and processes for manufacture and use thereof
US5023243A (en)1981-10-231991-06-11Molecular Biosystems, Inc.Oligonucleotide therapeutic agent and method of making same
US4476301A (en)1982-04-291984-10-09Centre National De La Recherche ScientifiqueOligonucleotides, a process for preparing the same and their application as mediators of the action of interferon
US5118800A (en)1983-12-201992-06-02California Institute Of TechnologyOligonucleotides possessing a primary amino group in the terminal nucleotide
US5550111A (en)1984-07-111996-08-27Temple University-Of The Commonwealth System Of Higher EducationDual action 2',5'-oligoadenylate antiviral derivatives and uses thereof
US4981957A (en)1984-07-191991-01-01Centre National De La Recherche ScientifiqueOligonucleotides with modified phosphate and modified carbohydrate moieties at the respective chain termini
US5367066A (en)1984-10-161994-11-22Chiron CorporationOligonucleotides with selectably cleavable and/or abasic sites
US4845205A (en)1985-01-081989-07-04Institut Pasteur2,N6 -disubstituted and 2,N6 -trisubstituted adenosine-3'-phosphoramidites
US5521063A (en)1985-03-151996-05-28Antivirals Inc.Polynucleotide reagent containing chiral subunits and methods of use
US5034506A (en)1985-03-151991-07-23Anti-Gene Development GroupUncharged morpholino-based polymers having achiral intersubunit linkages
US5142047A (en)1985-03-151992-08-25Anti-Gene Development GroupUncharged polynucleotide-binding polymers
US5506337A (en)1985-03-151996-04-09Antivirals Inc.Morpholino-subunit combinatorial library and method
US5698685A (en)1985-03-151997-12-16Antivirals Inc.Morpholino-subunit combinatorial library and method
US5185444A (en)1985-03-151993-02-09Anti-Gene Deveopment GroupUncharged morpolino-based polymers having phosphorous containing chiral intersubunit linkages
US5217866A (en)1985-03-151993-06-08Anti-Gene Development GroupPolynucleotide assay reagent and method
US5235033A (en)1985-03-151993-08-10Anti-Gene Development GroupAlpha-morpholino ribonucleoside derivatives and polymers thereof
US4683202A (en)1985-03-281987-07-28Cetus CorporationProcess for amplifying nucleic acid sequences
US4683202B1 (en)1985-03-281990-11-27Cetus Corp
US5276019A (en)1987-03-251994-01-04The United States Of America As Represented By The Department Of Health And Human ServicesInhibitors for replication of retroviruses and for the expression of oncogene products
US5264423A (en)1987-03-251993-11-23The United States Of America As Represented By The Department Of Health And Human ServicesInhibitors for replication of retroviruses and for the expression of oncogene products
US5286717A (en)1987-03-251994-02-15The United States Of America As Represented By The Department Of Health And Human ServicesInhibitors for replication of retroviruses and for the expression of oncogene products
US5552540A (en)1987-06-241996-09-03Howard Florey Institute Of Experimental Physiology And MedicineNucleoside derivatives
US5405939A (en)1987-10-221995-04-11Temple University Of The Commonwealth System Of Higher Education2',5'-phosphorothioate oligoadenylates and their covalent conjugates with polylysine
US5188897A (en)1987-10-221993-02-23Temple University Of The Commonwealth System Of Higher EducationEncapsulated 2',5'-phosphorothioate oligoadenylates
US5519126A (en)1988-03-251996-05-21University Of Virginia Alumni Patents FoundationOligonucleotide N-alkylphosphoramidates
US5453496A (en)1988-05-261995-09-26University Patents, Inc.Polynucleotide phosphorodithioate
US5278302A (en)1988-05-261994-01-11University Patents, Inc.Polynucleotide phosphorodithioates
US5216141A (en)1988-06-061993-06-01Benner Steven AOligonucleotide analogs containing sulfur linkages
US5175273A (en)1988-07-011992-12-29Genentech, Inc.Nucleic acid intercalating agents
US5445934A (en)1989-06-071995-08-29Affymax Technologies N.V.Array of oligonucleotides on a solid substrate
US5744305A (en)1989-06-071998-04-28Affymetrix, Inc.Arrays of materials attached to a substrate
US5134066A (en)1989-08-291992-07-28Monsanto CompanyImproved probes using nucleosides containing 3-dezauracil analogs
US5591722A (en)1989-09-151997-01-07Southern Research Institute2'-deoxy-4'-thioribonucleosides and their antiviral activity
US5399676A (en)1989-10-231995-03-21Gilead SciencesOligonucleotides with inverted polarity
US5466786B1 (en)1989-10-241998-04-07Gilead Sciences2' Modified nucleoside and nucleotide compounds
US5466786A (en)1989-10-241995-11-14Gilead Sciences2'modified nucleoside and nucleotide compounds
US5264564A (en)1989-10-241993-11-23Gilead SciencesOligonucleotide analogs with novel linkages
US5455233A (en)1989-11-301995-10-03University Of North CarolinaOligoribonucleoside and oligodeoxyribonucleoside boranophosphates
US7495088B1 (en)1989-12-042009-02-24Enzo Life Sciences, Inc.Modified nucleotide compounds
US5405938A (en)1989-12-201995-04-11Anti-Gene Development GroupSequence-specific binding polymers for duplex nucleic acids
US5166315A (en)1989-12-201992-11-24Anti-Gene Development GroupSequence-specific binding polymers for duplex nucleic acids
US6239265B1 (en)1990-01-112001-05-29Isis Pharmaceuticals, Inc.Oligonucleotides having chiral phosphorus linkages
US5587469A (en)1990-01-111996-12-24Isis Pharmaceuticals, Inc.Oligonucleotides containing N-2 substituted purines
US5459255A (en)1990-01-111995-10-17Isis Pharmaceuticals, Inc.N-2 substituted purines
US5750692A (en)1990-01-111998-05-12Isis Pharmaceuticals, Inc.Synthesis of 3-deazapurines
US5646265A (en)1990-01-111997-07-08Isis Pharmceuticals, Inc.Process for the preparation of 2'-O-alkyl purine phosphoramidites
US5681941A (en)1990-01-111997-10-28Isis Pharmaceuticals, Inc.Substituted purines and oligonucleotide cross-linking
US5670633A (en)1990-01-111997-09-23Isis Pharmaceuticals, Inc.Sugar modified oligonucleotides that detect and modulate gene expression
US5563253A (en)1990-03-081996-10-08Worcester Foundation For Biomedical ResearchLinear aminoalkylphosphoramidate oligonucleotide derivatives
US5321131A (en)1990-03-081994-06-14Hybridon, Inc.Site-specific functionalization of oligodeoxynucleotides for non-radioactive labelling
US5536821A (en)1990-03-081996-07-16Worcester Foundation For Biomedical ResearchAminoalkylphosphorothioamidate oligonucleotide deratives
US5470967A (en)1990-04-101995-11-28The Dupont Merck Pharmaceutical CompanyOligonucleotide analogs with sulfamate linkages
US5567811A (en)1990-05-031996-10-22Amersham International PlcPhosphoramidite derivatives, their preparation and the use thereof in the incorporation of reporter groups on synthetic oligonucleotides
US5514785A (en)1990-05-111996-05-07Becton Dickinson And CompanySolid supports for nucleic acid hybridization assays
US5541307A (en)1990-07-271996-07-30Isis Pharmaceuticals, Inc.Backbone modified oligonucleotide analogs and solid phase synthesis thereof
US5608046A (en)1990-07-271997-03-04Isis Pharmaceuticals, Inc.Conjugated 4'-desmethyl nucleoside analog compounds
US5602240A (en)1990-07-271997-02-11Ciba Geigy Ag.Backbone modified oligonucleotide analogs
US5614617A (en)1990-07-271997-03-25Isis Pharmaceuticals, Inc.Nuclease resistant, pyrimidine modified oligonucleotides that detect and modulate gene expression
US5623070A (en)1990-07-271997-04-22Isis Pharmaceuticals, Inc.Heteroatomic oligonucleoside linkages
US5489677A (en)1990-07-271996-02-06Isis Pharmaceuticals, Inc.Oligonucleoside linkages containing adjacent oxygen and nitrogen atoms
US5618704A (en)1990-07-271997-04-08Isis Pharmacueticals, Inc.Backbone-modified oligonucleotide analogs and preparation thereof through radical coupling
US5610289A (en)1990-07-271997-03-11Isis Pharmaceuticals, Inc.Backbone modified oligonucleotide analogues
US5677437A (en)1990-07-271997-10-14Isis Pharmaceuticals, Inc.Heteroatomic oligonucleoside linkages
US5677439A (en)1990-08-031997-10-14SanofiOligonucleotide analogues containing phosphate diester linkage substitutes, compositions thereof, and precursor dinucleotide analogues
US5214134A (en)1990-09-121993-05-25Sterling Winthrop Inc.Process of linking nucleosides with a siloxane bridge
US5561225A (en)1990-09-191996-10-01Southern Research InstitutePolynucleotide analogs containing sulfonate and sulfonamide internucleoside linkages
US5596086A (en)1990-09-201997-01-21Gilead Sciences, Inc.Modified internucleoside linkages having one nitrogen and two carbon atoms
US5432272A (en)1990-10-091995-07-11Benner; Steven A.Method for incorporating into a DNA or RNA oligonucleotide using nucleotides bearing heterocyclic bases
US5177195A (en)1991-01-081993-01-05Imperial Chemical Industries PlcDisazo dyes
US5714331A (en)1991-05-241998-02-03Buchardt, Deceased; OlePeptide nucleic acids having enhanced binding affinity, sequence specificity and solubility
US5571799A (en)1991-08-121996-11-05Basco, Ltd.(2'-5') oligoadenylate analogues useful as inhibitors of host-v5.-graft response
US5587361A (en)1991-10-151996-12-24Isis Pharmaceuticals, Inc.Oligonucleotides having phosphorothioate linkages of high chiral purity
US5319080A (en)1991-10-171994-06-07Ciba-Geigy CorporationBicyclic nucleosides, oligonucleotides, process for their preparation and intermediates
US5393878A (en)1991-10-171995-02-28Ciba-Geigy CorporationBicyclic nucleosides, oligonucleotides, process for their preparation and intermediates
US5594121A (en)1991-11-071997-01-14Gilead Sciences, Inc.Enhanced triple-helix and double-helix formation with oligomers containing modified purines
US5677195A (en)1991-11-221997-10-14Affymax Technologies N.V.Combinatorial strategies for polymer synthesis
US5484908A (en)1991-11-261996-01-16Gilead Sciences, Inc.Oligonucleotides containing 5-propynyl pyrimidines
US6380368B1 (en)1991-11-262002-04-30Isis Pharmaceuticals, Inc.Enhanced triple-helix and double-helix formation with oligomers containing modified pyrimidines
US6235887B1 (en)1991-11-262001-05-22Isis Pharmaceuticals, Inc.Enhanced triple-helix and double-helix formation directed by oligonucleotides containing modified pyrimidines
US5359044A (en)1991-12-131994-10-25Isis PharmaceuticalsCyclobutyl oligonucleotide surrogates
US6277603B1 (en)1991-12-242001-08-21Isis Pharmaceuticals, Inc.PNA-DNA-PNA chimeric macromolecules
US7015315B1 (en)1991-12-242006-03-21Isis Pharmaceuticals, Inc.Gapped oligonucleotides
US6326199B1 (en)1991-12-242001-12-04Isis Pharmaceuticals, Inc.Gapped 2′ modified oligonucleotides
US5639873A (en)1992-02-051997-06-17Centre National De La Recherche Scientifique (Cnrs)Oligothionucleotides
US5541316A (en)1992-02-111996-07-30Henkel Kommanditgesellschaft Auf AktienProcess for the production of polysaccharide-based polycarboxylates
US5633360A (en)1992-04-141997-05-27Gilead Sciences, Inc.Oligonucleotide analogs capable of passive cell membrane permeation
US5434257A (en)1992-06-011995-07-18Gilead Sciences, Inc.Binding compentent oligomers containing unsaturated 3',5' and 2',5' linkages
US5610300A (en)1992-07-011997-03-11Ciba-Geigy CorporationCarbocyclic nucleosides containing bicyclic rings, oligonucleotides therefrom, process for their preparation, their use and intermediates
US5700920A (en)1992-07-011997-12-23Novartis CorporationCarbocyclic nucleosides containing bicyclic rings, oligonucleotides therefrom, process for their preparation, their use and intermediates
WO1994002595A1 (en)1992-07-171994-02-03Ribozyme Pharmaceuticals, Inc.Method and reagent for treatment of animal diseases
US6683167B2 (en)1992-07-232004-01-27University Of Massachusetts WorcesterHybrid oligonucleotide phosphorothioates
US6346614B1 (en)1992-07-232002-02-12Hybridon, Inc.Hybrid oligonucleotide phosphorothioates
US5476925A (en)1993-02-011995-12-19Northwestern UniversityOligodeoxyribonucleotides including 3'-aminonucleoside-phosphoramidate linkages and terminal 3'-amino groups
US5466677A (en)1993-03-061995-11-14Ciba-Geigy CorporationDinucleoside phosphinates and their pharmaceutical compositions
US5576427A (en)1993-03-301996-11-19Sterling Winthrop, Inc.Acyclic nucleoside analogs and oligonucleotide sequences containing them
US5663312A (en)1993-03-311997-09-02SanofiOligonucleotide dimers with amide linkages replacing phosphodiester linkages
US5658873A (en)1993-04-101997-08-19Degussa AktiengesellschaftCoated sodium percarbonate particles, a process for their production and detergent, cleaning and bleaching compositions containing them
US5539082A (en)1993-04-261996-07-23Nielsen; Peter E.Peptide nucleic acids
US6015886A (en)1993-05-242000-01-18Chemgenes CorporationOligonucleotide phosphate esters
US5502177A (en)1993-09-171996-03-26Gilead Sciences, Inc.Pyrimidine derivatives for labeled binding partners
US6028188A (en)1993-11-162000-02-22Genta IncorporatedSynthetic oligomers having chirally pure phosphonate internucleosidyl linkages mixed with non-phosphonate internucleosidyl linkages
US5719262A (en)1993-11-221998-02-17Buchardt, Deceased; OlePeptide nucleic acids having amino acid side chains
US5457187A (en)1993-12-081995-10-10Board Of Regents University Of NebraskaOligonucleotides containing 5-fluorouracil
US5446137B1 (en)1993-12-091998-10-06Behringwerke AgOligonucleotides containing 4'-substituted nucleotides
US5446137A (en)1993-12-091995-08-29Syntex (U.S.A.) Inc.Oligonucleotides containing 4'-substituted nucleotides
US5519134A (en)1994-01-111996-05-21Isis Pharmaceuticals, Inc.Pyrrolidine-containing monomers and oligomers
US6169170B1 (en)1994-03-182001-01-02Lynx Therapeutics, Inc.Oligonucleotide N3′→N5′Phosphoramidate Duplexes
US5596091A (en)1994-03-181997-01-21The Regents Of The University Of CaliforniaAntisense oligonucleotides comprising 5-aminoalkyl pyrimidine nucleotides
US5627053A (en)1994-03-291997-05-06Ribozyme Pharmaceuticals, Inc.2'deoxy-2'-alkylnucleotide containing nucleic acid
US5625050A (en)1994-03-311997-04-29Amgen Inc.Modified oligonucleotides and intermediates useful in nucleic acid therapeutics
US6054299A (en)1994-04-292000-04-25Conrad; Charles A.Stem-loop cloning vector and method
US5525711A (en)1994-05-181996-06-11The United States Of America As Represented By The Secretary Of The Department Of Health And Human ServicesPteridine nucleotide analogs as fluorescent DNA probes
US5597909A (en)1994-08-251997-01-28Chiron CorporationPolynucleotide reagents containing modified deoxyribose moieties, and associated methods of synthesis and use
US5770722A (en)1994-10-241998-06-23Affymetrix, Inc.Surface-bound, unimolecular, double-stranded DNA
US6608035B1 (en)1994-10-252003-08-19Hybridon, Inc.Method of down-regulating gene expression
US6124445A (en)1994-11-232000-09-26Isis Pharmaceuticals, Inc.Phosphotriester oligonucleotides, amidities and method of preparation
US6166197A (en)1995-03-062000-12-26Isis Pharmaceuticals, Inc.Oligomeric compounds having pyrimidine nucleotide (S) with 2'and 5 substitutions
US6222025B1 (en)1995-03-062001-04-24Isis Pharmaceuticals, Inc.Process for the synthesis of 2′-O-substituted pyrimidines and oligomeric compounds therefrom
US5874219A (en)1995-06-071999-02-23Affymetrix, Inc.Methods for concurrently processing multiple biological chip assays
US6815432B2 (en)1995-06-072004-11-09Inex Pharmaceuticals Corp.Methods for encapsulating plasmids in lipid bilayers
US6160109A (en)1995-10-202000-12-12Isis Pharmaceuticals, Inc.Preparation of phosphorothioate and boranophosphate oligomers
US5854033A (en)1995-11-211998-12-29Yale UniversityRolling circle replication reporter systems
US6444423B1 (en)1996-06-072002-09-03Molecular Dynamics, Inc.Nucleosides comprising polydentate ligands
US6639062B2 (en)1997-02-142003-10-28Isis Pharmaceuticals, Inc.Aminooxy-modified nucleosidic compounds and oligomeric compounds prepared therefrom
US6172209B1 (en)1997-02-142001-01-09Isis Pharmaceuticals Inc.Aminooxy-modified oligonucleotides and methods for making same
US6858225B2 (en)1997-05-142005-02-22Inex Pharmaceuticals CorporationLipid-encapsulated polyanionic nucleic acid
US6528640B1 (en)1997-11-052003-03-04Ribozyme Pharmaceuticals, IncorporatedSynthetic ribonucleic acids with RNAse activity
US6617438B1 (en)1997-11-052003-09-09Sirna Therapeutics, Inc.Oligoribonucleotides with enzymatic activity
US7273933B1 (en)1998-02-262007-09-25Isis Pharmaceuticals, Inc.Methods for synthesis of oligonucleotides
US7045610B2 (en)1998-04-032006-05-16Epoch Biosciences, Inc.Modified oligonucleotides for mismatch discrimination
US6531590B1 (en)1998-04-242003-03-11Isis Pharmaceuticals, Inc.Processes for the synthesis of oligonucleotide compounds
USRE39464E1 (en)1998-07-142007-01-09Isis Pharmaceuticals Inc.Oligonucleolotides having site specific chiral phosphorothioate internucleoside linkages
US6867294B1 (en)1998-07-142005-03-15Isis Pharmaceuticals, Inc.Gapped oligomers having site specific chiral phosphorothioate internucleoside linkages
WO2000022114A1 (en)1998-10-092000-04-20Ingene, Inc.PRODUCTION OF ssDNA $i(IN VIVO)
WO2000022113A1 (en)1998-10-092000-04-20Ingene, Inc.ENZYMATIC SYNTHESIS OF ssDNA
US7041816B2 (en)1999-02-042006-05-09Isis Pharmaceuticals, Inc.Process for the synthesis of oligomeric compounds
US6858715B2 (en)1999-02-042005-02-22Isis Pharmaceuticals, Inc.Process for the synthesis of oligomeric compounds
US6534639B1 (en)1999-07-072003-03-18Isis Pharmaceuticals, Inc.Guanidinium functionalized oligonucleotides and method/synthesis
US6147200A (en)1999-08-192000-11-14Isis Pharmaceuticals, Inc.2'-O-acetamido modified monomers and oligomers
US7321029B2 (en)2000-01-212008-01-22Geron Corporation2′-arabino-fluorooligonucleotide N3′→P5′ phosphoramidates: their synthesis and use
US20080153753A1 (en)2002-04-252008-06-26Thomas WillnowMethod of treating side effects induced by therapeutic agents
US8101348B2 (en)2002-07-102012-01-24Max-Planck-Gesellschaft Zur Foerderung Der Wissenschaften E.V.RNA-interference by single-stranded RNA molecules
US6878805B2 (en)2002-08-162005-04-12Isis Pharmaceuticals, Inc.Peptide-conjugated oligomeric compounds
US7560431B2 (en)2003-06-202009-07-14Raptor Pharmaceutical Inc.Methods of increasing delivery of active agents to brain comprising administering receptor associated protein (RAP) fragments conjugated to active agents
US7427672B2 (en)2003-08-282008-09-23Takeshi ImanishiArtificial nucleic acids of n-o bond crosslinkage type
US10808039B2 (en)2003-11-062020-10-20Seattle Genetics Inc.Monomethylvaline compounds capable of conjugation to ligands
US7427605B2 (en)2005-03-312008-09-23Calando Pharmaceuticals, Inc.Inhibitors of ribonucleotide reductase subunit 2 and uses thereof
WO2006138343A2 (en)2005-06-142006-12-28Raptor Pharmaceutical Inc.Compositions comprising receptor - associated protein (rap) variants specific for lrp2 and uses thereof
US8877714B2 (en)2005-06-142014-11-04Raptor Pharmaceutical Inc.Compositions comprising receptor-associated protein (RAP) variants specific for LRP2 and uses thereof
US20130011922A1 (en)2007-03-022013-01-10F/K/A Mdrna, Inc.Nucleic acid compounds for inhibiting gene expression and uses thereof
US8795627B2 (en)2007-03-212014-08-05Raptor Pharmaceuticals Inc.Treatment of liver disorders by administration of RAP conjugates
US10131907B2 (en)2007-04-232018-11-20Alnylam Pharmaceuticals, Inc.Glycoconjugates of RNA interference agents
US20130096289A1 (en)2007-05-222013-04-18Marina Biotech, Inc.Hydroxymethyl substituted rna oligonucleotides and rna complexes
US8314227B2 (en)2007-05-222012-11-20Marina Biotech, Inc.Hydroxymethyl substituted RNA oligonucleotides and RNA complexes
WO2009073809A2 (en)2007-12-042009-06-11Alnylam Pharmaceuticals, Inc.Carbohydrate conjugates as delivery agents for oligonucleotides
US8106022B2 (en)2007-12-042012-01-31Alnylam Pharmaceuticals, Inc.Carbohydrate conjugates as delivery agents for oligonucleotides
US8828956B2 (en)2007-12-042014-09-09Alnylam Pharmaceuticals, Inc.Carbohydrate conjugates as delivery agents for oligonucleotides
US9463252B2 (en)2008-03-182016-10-11Seattle Genetics, Inc.Auristatin drug linker conjugates
US8058069B2 (en)2008-04-152011-11-15Protiva Biotherapeutics, Inc.Lipid formulations for nucleic acid delivery
US20110313020A1 (en)2008-12-032011-12-22Marina Biotech, Inc.UsiRNA Complexes
US8158601B2 (en)2009-06-102012-04-17Alnylam Pharmaceuticals, Inc.Lipid formulation
WO2011005861A1 (en)2009-07-072011-01-13Alnylam Pharmaceuticals, Inc.Oligonucleotide end caps
US20120157511A1 (en)2009-07-072012-06-21Alnylam Pharmaceuticals, Inc.5' phosphate mimics
US9422562B2 (en)2010-04-062016-08-23Alnylam PharmaceuticalsCompositions and methods for inhibiting expression of CD274/PD-L1 gene
US20130190383A1 (en)2010-04-262013-07-25Marina Biotech, Inc.Nucleic acid compounds with conformationally restricted monomers and uses thereof
US9388418B2 (en)2011-06-122016-07-12City Of HopeAptamer-mRNA conjugates for targeted protein or peptide expression and methods for their use
WO2013036868A1 (en)2011-09-072013-03-14Marina Biotech Inc.Synthesis and uses of nucleic acid compounds with conformationally restricted monomers
US11071749B2 (en)2011-12-082021-07-27Sarepta Therapeutics, Inc.Oligonucleotide analogues targeting human LMNA
US9133461B2 (en)2012-04-102015-09-15Alnylam Pharmaceuticals, Inc.Compositions and methods for inhibiting expression of the ALAS1 gene
US10065993B2 (en)2013-05-072018-09-04Merck Patent GmbhPeptides and peptide/active compound conjugates for renal targeting
WO2015027205A1 (en)2013-08-222015-02-26Robert DoyleCompositions comprising vitamin b12 and intrinsic factor and methods of use thereof
US11208430B2 (en)2014-08-222021-12-28Noogen LlcModified oligonucleotides and methods for their synthesis
WO2017100700A2 (en)2015-12-112017-06-15Fred Hutchinson Cancer Research CenterPeptides for renal therapy
WO2018232122A1 (en)2017-06-152018-12-20Blaze Bioscience, Inc.Renal-homing peptide conjugates and methods of use thereof
WO2021195533A2 (en)2020-03-262021-09-30University Of MassachusettsSynthesis of modified oligonucleotides with increased stability
WO2023278589A1 (en)2021-06-302023-01-05Ionis Pharmaceuticals, Inc.Method for synthesis of linkage modified oligomeric compounds
WO2023201095A2 (en)2022-04-152023-10-19Wave Life Sciences Ltd.Oligonucleotide compositions and methods relating thereto

Non-Patent Citations (87)

* Cited by examiner, † Cited by third party
Title
"GenBank", Database accession no. NM_001003841.3
"Swiss-Prot", Database accession no. 060494
AIGNER, A., J. BIOMED. BIOTECHNOL., 2006, pages 71659
AKANEYA,Y. ET AL., J. NEUROPHYSIOL., vol. 93, 2005, pages 594 - 602
ARNOLD, A S ET AL., J. HYPERTENS., 2007, pages 197 - 205
AUSUBEL, F. M. ET AL.: "Current Protocols in Molecular Biology", vol. 2, 1997, JOHN WILEY & SONS, INC., pages: 1 - 21
BARANY, PROC. NATL. ACAD. SCI. USA, vol. 88, 1991, pages 189 - 19
BELANGER ET AL., JCI INSIGHT, vol. 3, no. 14, pages 121762
BERNSTEIN ET AL., NATURE, vol. 409, 2001, pages 363
BONNET ME ET AL., PHARM. RES., 16 August 2008 (2008-08-16)
BRASELL EJ ET AL., PLOS ONE, vol. 14, no. 12, 2019, pages 0223954
CHIEN, PY ET AL., CANCER GENE THER, vol. 12, 2005, pages 321 - 328
CHURANA, RNA, vol. 14, 2007, pages 1714 - 1719
COUTURE, A. ET AL., TIG, vol. 12, 1996, pages 5 - 10
CROOKE ET AL., J. PHARMACOL. EXP. THER., vol. 277, 1996, pages 923 - 937
DAGIL R ET AL., JOURNAL OF BIOLOGICAL CHEMISTRY, vol. 288, no. 6, 2013, pages 4424 - 4435
DARN, G. ET AL., NUCLEIC ACIDS, vol. 32, 2004, pages 49
DIAS, N. ET AL., MOL CANCER THER, vol. 1, 2002, pages 347 - 355
DINESH KP ET AL., AM J KIDNEY DIS, vol. 74, no. 1, 2019, pages 132 - 137
DIRKSEN ET AL., J. AM. CHEM. SOC., vol. 128, no. 49, 2006, pages 15602 - 15603
ELBASHIR ET AL., EMBO, vol. 20, 2001, pages 6877 - 6888
ELBASHIR ET AL., GENES DEV, vol. 15, 2001, pages 188
ELMEN, J. ET AL., NUCLEIC ACIDS RESEARCH, vol. 33, no. 1, 2005, pages 439 - 447
ENGLISCH ET AL., ANGEWANDTE CHEMIE, vol. 30, 1991, pages 613
FANTONI ET AL., CHEM. REV., vol. 121, no. 12, 2021, pages 7122 - 7154
FLUITER ET AL., MOL, vol. 10, 2009, pages 1039
FRIER ET AL., PROC. NAT. ACAD. SCI. USA, vol. 83, 1986, pages 9373 - 77
GAMAYO A ET AL., CLIN KIDNEY J, vol. 13, no. 3, 2019, pages 468 - 472
GASSMANN ET AL., PROC. NATL. ACAD. SCI. USA, vol. 92, 1995, pages 1292
GRUNWELLER, A. ET AL., NUCLEIC ACIDS RESEARCH, vol. 31, no. 12, 2003, pages 3185 - 3193
GUATELLI ET AL., PROC. NATL. ACAD. SCI. USA, vol. 87, 1990, pages 1874 - 1878
HENDRIKX J ET AL., ONCOLOGIST, vol. 22, no. 10, 2017, pages 1212 - 1221
KABANOV ET AL., FEES LETT, vol. 259, 1990, pages 327 - 330
KIM ET AL., NAT BIOTECH, vol. 23, 2005, pages 222 - 226
KIM SFI ET AL., JOURNAL OF CONTROLLED RELEASE, vol. 129, no. 2, 2008, pages 107 - 116
KOZLOV ET AL., BIOPOLYMERS, vol. 73, no. 5, 2004, pages 621 - 30
KWOH ET AL., PROC. NATL. ACAD. SCI. USA, vol. 86, pages 1173 - 1177
LARSEN C. ET AL., JAM SOC NEPHROL, vol. 29, no. 2, 2018, pages 644 - 653
LARSEN CP ET AL., J AM SOC NEPHROL, no. 2, 2018, pages 644 - 653
LETSINGER ET AL., PROC. NATL. ACID. SCI. USA, vol. 86, 1989, pages 6553 - 6556
LIU, S., MMOL. PHARM., vol. 3, 2006, pages 472 - 487
LIZARDI ET AL., BIO/TECHNOLOGY, vol. 6, no. 11, 1988, pages 7
MAKIMURA, H. ET AL., BMC NEUROSCI, vol. 3, 2002, pages 18
MANOHARAN ET AL., ANN. N.Y. ACAD. SCI., vol. 660, 1992, pages 300 - 309
MANOHARAN ET AL., BIORG. MED. CHEM. LET., vol. 3, 1993, pages 2765 - 2770
MANOHARAN ET AL., BIORG. MED. CHEM. LET., vol. 4, 1994, pages 1053 - 1060
MANOHARAN ET AL., NUCLEOSIDES & NUCLEOTIDES, vol. 14, 1995, pages 969 - 973
MANOHARAN ET AL., TETRAHEDRON LETT, vol. 36, 1995, pages 3651 - 3654
MARTIN ET AL., HELV. CHIM. ACTA, vol. 78, 1995, pages 486 - 504
MARZOLOFARFAN, BIOL RES, vol. 44, 2011, pages 89 - 105
MISHRA ET AL., BIOCHIM. BIOPHYS. ACTA, vol. 1264, 1995, pages 229 - 237
MOOK, OR. ET AL., MOL CANC THER, vol. 6, no. 3, 2007, pages 833 - 843
NIELSEN ET AL., SCIENCE, vol. 254, 1991, pages 1497 - 1500
NIELSEN R. ET AL., KIDNEY INT, vol. 89, no. 1, 2016, pages 58 - 67
NUC. ACIDS SYMP. SERIES, vol. 52, 2008, pages 133 - 134
NYKANEN ET AL., CELL, vol. 150, 2012, pages 883 - 894
OBERHAUSER ET AL., NUCL. ACIDS RES., vol. 20, 1992, pages 533 - 538
PAL, A ET AL., INT J. ONCOL., vol. 26, 2005, pages 1087 - 1091
PEREZ-GOMEZ MV ET AL., CLIN KIDNEY J, vol. 13, no. 3, 2020, pages 281 - 286
POPADYNEC M. ET AL., ACS MED. CHEM. LEFT., vol. 12, no. 9, 2021, pages 1486 - 1492
REVASCO ET AL., CHEM. EUR. J., 2018
ROSENBERG ET AL., NEW ENG. J. OF MED., vol. 319, 1988, pages 1676
SAISON-BEHMOARAS ET AL., EMBO J, vol. 10, 1991, pages 1111 - 1118
SAUNDERS K.O., FRONTIERS IN IMMUNOLOGY, vol. 10, 2019
SCHUMACHER ET AL., REGEN MED, vol. 6, 2021, pages 45
SERGEEVA ET AL., FRONT CHEM, vol. 12, 2024, pages 2024
SHAH M. ET AL., JOURNAL OF CELL BIOLOGY, vol. 202, no. 1, 2013, pages 113 - 127
SHEA ET AL., NUCL. ACIDS RES., vol. 18, 1990, pages 3777 - 3783
SHISHKINA, GT ET AL., NEUROSCIENCE, vol. 129, 2004, pages 521 - 528
SIMEONI ET AL., NUCL. ACIDS RES., vol. 31, 2003, pages 2717 - 2724
SORENSEN, DR ET AL., J. MOL. BIOL, vol. 327, 2003, pages 761 - 766
SOUTSCHEK, J. ET AL., NATURE, vol. 432, 2004, pages 173 - 178
SPICER ET AL., CHEM. REV., vol. 118, no. 16, 2018, pages 7702 - 7743
SVINARCHUK ET AL., BIOCHIMIE, vol. 75, 1993, pages 49 - 54
TAN, PH ET AL., GENE THER, vol. 12, 2005, pages 59 - 66
THAKKER, ER ET AL., PROC. NATL. ACAD. SCI. U.S.A., vol. 101, 2004, pages 17270 - 17275
TOMALIA, DA ET AL., BIOCHEM. SOC. TRANS., vol. 35, 2007, pages 61 - 67
TURNER ET AL., J. AM. CHEM. SOC, vol. 109, 1987, pages 3783 - 3785
TURNER, CSH SYMP. QUANT. BIOL., vol. 117, 1987, pages 123 - 133
VEGT ET AL., EUR J NUCL MED MOL IMAGING, 2011
VERMA, UN ET AL., CLIN. CANCER RES., vol. 9, 2003, pages 1291 - 1300
WILLNOW ET AL., EMBO J, vol. 15, 1996, pages 2632 - 2639
WISCHNJOW ET AL., BIOCONJUGATE CHEM, vol. 27, 2016, pages 1050 - 1057
WOOLF ET AL., PROC. NATL. ACAD. SCI. USA, vol. 89, 1992, pages 7305 - 7309
YAMADA, K. ET AL., NATURE BIOTECHNOLOGY, 2024
YOO, H. ET AL., PHARM. RES., vol. 16, 1999, pages 1799 - 1804
ZIMMERMANN, TS ET AL., NATURE, vol. 441, 2006, pages 111 - 114

Also Published As

Publication numberPublication date
WO2025072672A3 (en)2025-05-08

Similar Documents

PublicationPublication DateTitle
US20250154512A1 (en)Rnai constructs for inhibiting pnpla3 expression and methods of use thereof
US20220333104A1 (en)TRANSTHYRETIN (TTR) iRNA COMPOSITIONS AND METHODS OF USE THEREOF FOR TREATING OR PREVENTING TTR-ASSOCIATED OCULAR DISEASES
TWI694080B (en)Compositions and methods for inhibiting expression of the alas1 gene
CN112424355A (en)Ketohexokinase (KHK) iRNA compositions and methods of use thereof
EP3894555A2 (en)Rnai constructs for inhibiting pnpla3 expression
CA3184345A1 (en)Rnai constructs for inhibiting hsd17b13 expression and methods of use thereof
US20220307022A1 (en)Rnai constructs for inhibiting scap expression and methods of use thereof
US20240240187A1 (en)Sirna targeting tmprss6 for the treatment of myeloproliferative disorders
WO2025072672A2 (en)Slc6a19-targeting modulatory nucleic acid agents
CN117561334A (en)Human chromosome 9 open reading frame 72 (C9 ORF 72) iRNA pharmaceutical compositions and methods of use thereof
US20250207138A1 (en)Rnai constructs for inhibiting pnpla3 expression and methods of use thereof
WO2025072713A1 (en)Polymyxins for delivery of agents to the kidney
WO2025072699A1 (en)Aminoglycosides for delivery of agents to the kidney
AU2023395944A1 (en)Rnai constructs for inhibiting ttr expression and methods of use thereof
CA3150758A1 (en)Rnai constructs for inhibiting slc30a8 expression and methods of use thereof
CN117751187A (en)Ketohexokinase (KHK) iRNA compositions and methods of use thereof
CN118922543A (en)Compositions and methods for treating PCDH 19-related disorders
CN117651769A (en)Beta catenin (CTNNB 1) iRNA compositions and methods of use thereof
BR122025007695A2 (en) RNAI CONSTRUCTS FOR INHIBITTING PNPLA3 EXPRESSION, PHARMACEUTICAL COMPOSITION COMPRISING THE SAME AND USE OF THE RNAI CONSTRUCT OR COMPOSITION

Legal Events

DateCodeTitleDescription
121Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number:24790224

Country of ref document:EP

Kind code of ref document:A2


[8]ページ先頭

©2009-2025 Movatter.jp