OA21336A - Modified short interfering nucleic acid (siNA) molecules and uses thereof. - Google Patents

Modified short interfering nucleic acid (siNA) molecules and uses thereof. Download PDF

Info

Publication number
OA21336A
OA21336A OA1202200362 OA21336A OA 21336 A OA21336 A OA 21336A OA 1202200362 OA1202200362 OA 1202200362 OA 21336 A OA21336 A OA 21336A
Authority
OA
OAPI
Prior art keywords
nucléotide
nucléotides
sina
sequence
fluoro
Prior art date
Application number
OA1202200362
Inventor
Leonid Beigelman
Vivek Kumar Rajwanshi
Jin Hong
Rajendra K. Pandey
Markus Hossbach
Lax man ELTE PU
Saul MARTINEZ MONTERO
N. Tilani S. DE COSTA
Original Assignee
Aligos Therapeutics, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Aligos Therapeutics, Inc. filed Critical Aligos Therapeutics, Inc.
Publication of OA21336A publication Critical patent/OA21336A/en

Links

Abstract

Disclosed herein are short interfering nucleic acid (siNA) molecules comprising modified nucleotides and uses therof. The siNA molecules may be double stranded and comprise modified nucleotides selected from 2’-O-methyl nucleotides and 2’-fluoro nucleotides. Further disclosed herein are siNA molecules comprising (a) a phosphorylation blocker, conjugated moiety, or 5’- stabilized end cap; and (b) a short interfering nucleic acid (siNA).

Description

Modifiée! Short Interfering Nucleic Acid (siNA) Molécules and Uses Thereof CROSS-REFERENCE TO RELATED APPLICATIONS
This application daims priority to U.S. Provisional Application No. 62/986,150, filed March 6, 2020, and U.S. Provisional Application No. 63/109,196, filed November 3, 2020, the disclosures of which are hereby incorporated by reference in their entireties.
SEQUENCE LISTING
The instant application contains a Sequence Listing which has been submitted electronically in ASCII format and is hereby incorporated by reference in its entirety. Said ASCII copy, created on May 13, 2021, is named 122400-0154_SL.txt and is 178,906 bytes in size. .
FIELD OF THE INVENTION
Described are short interfering nucleic acid (siNA) molécules comprising modified nucléotides, compositions, and uses thereof.
BACKGROUND OF THE INVENTION
RNA interférence (RNAi) is a biological response to double-stranded RNA that médiates résistance to both endogenous parasitic and exogenous pathogenic nucleic acids, and régulâtes the expression of protein-coding genes. The short interfering nucleic acids (siNA), such as siRNA, hâve been developed for RNAi therapy to treat a variety of diseases. For instance, RNAi therapy has been proposed for the treatment of metabolic diseases, neurodegenerative diseases, cancer, and pathogenic infections (See e.g., Rondindone, Biotechniques, 2018, 40(4S), doi.org/10.2144/000112163, Boudreau and Davidson, Curr Top Dev Biol, 2006, 75:73-92, Chalbatani et al., Int JNanomedicine, 2019, 14:3111-3128, Arbuthnot, DrugNews Perspect, 2010, 23(6):341-50, and Chemikov et. al., Front. Pharmacol., 2019, :
doi.org/10.3389/fphar.2019.00444, each of which are incorporated by reference in their entirety). However, major limitations of RNAi therapy are the ability to effectively deliver siRNA to target cells and the dégradation of the siRNA.
The présent disclosure improves the delivery and stability of siNA molécules by providing siNA molécules comprising modified nucléotides. The siNA molécules of the présent disclosure provide optimized combinations and numbers of modified nucléotides, nucléotide lengths, design (e.g., blunt ends or overhangs, intemucleoside linkages, conjugales), and modification patterns for improving the delivery and stability of siNA molécules.
SUMMARY OF THE INVENTION
Disclosed herein is a short interfering nucleic acid (siNA) molécule comprising: (a) a sense strand comprising a first nucléotide sequence that is at least about 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% identical to an RNA corresponding to a target gene, wherein the first nucléotide sequence: (i) is 15 to 30 nucléotides in length; and (ii) comprises 15 or more modified nucléotides independently selected from a 2’-(9-methyl nucléotide and a 2’-fluoro nucléotide, wherein at least one modified nucléotide is a 2’-O-methyl nucléotide and the nucléotide at position 3, 5, 7, 8, 9, 10, 11, 12, 14, 17, and/or 19 from the 5’ end of the first nucléotide sequence is a 2’-fluoro nucléotide; and (b) an antisense strand comprising a second nucléotide sequence that is at least about 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% complementary to the RNA corresponding to the target gene, wherein the second nucléotide sequence: (i) is 15 to 30 nucléotides in length; and (ii) comprises 15 or more modified nucléotides independently selected from a 2’-O-methyl nucléotide and a 2’-fluoro nucléotide, wherein at least one modified nucléotide is a 2’-(2-methyl nucléotide and at least one modified nucléotide is a 2’-fluoro nucléotide.
Disclosed herein is a short interfering nucleic acid (siNA) molécule comprising: (a) a sense strand comprising a first nucléotide sequence that is at least about 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% identical to an RNA corresponding to a target gene, wherein the first nucléotide sequence: (i) is 15 to 30 nucléotides in length; and (ii) comprises 15 or more modified nucléotides independently selected from a 2’-O-methyl nucléotide and a 2’-fluoro nucléotide, wherein at least one modified nucléotide is a 2’-(9-methyl nucléotide and at least one modified nucléotide is a 2’-fluoro nucléotide; and (b) an antisense strand comprising a second nucléotide sequence that is at least about 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% complementary to the RNA corresponding to the target gene, wherein the second nucléotide sequence: (i) is 15 to 30 nucléotides in length; and (ii) comprises 15 or more modified nucléotides independently selected from a 2’-O-methyl nucléotide and a 2’-fluoro nucléotide, wherein at least one modified nucléotide is a 2’-(9-methyl nucléotide and the nucléotide at position 2, 5, 6, 8, 10, 14,16, 17, and/or 18 from the 5’ end of the second nucléotide sequence is a 2’-fluoro nucléotide.
In some embodiments, the first nucléotide sequence comprises 16, 17, 18, 19, 20, 21, 22, 23, or more modified nucléotides independently selected from a 2’-(2-methyl nucléotide and a 2' fluoro nucléotide. In some embodiments, 70%, 75%, 80%, 85%, 90%, 95% or 100% of the nucléotides in the first nucléotide sequence are modified nucléotides independently selected from a 2’-O-methyl nucléotide and a 2’-fluoro nucléotide. In some embodiments, between 2 to 15 modified nucléotides of the first nucléotide sequence are 2’-fluoro nucléotides. In some embodiments, between 2 to 10 modified nucléotides of the first nucléotide sequence are 2’fluoro nucléotides. In some embodiments, between 2 to 6 modified nucléotides of the first nucléotide sequence are 2’-fluoro nucléotides. In some embodiments, at least 2, 3, 4, 5, or 6 modified nucléotides of the first nucléotide sequence are 2’-fluoro nucléotides. In some embodiments, less than or equal to 10, 9, 8, 7, 6, 5, 4, 3, or 2 modified nucléotides of the first nucléotide sequence are 2’-fluoro nucléotides. In some embodiments, between about 2 to 25 modified nucléotides of the first nucléotide sequence are 2’-<9-methyl nucléotides. In some embodiments, between about 2 to 20 modified nucléotides of the first nucléotide sequence are 2’-(9-methyl nucléotides. In some embodiments, between about 5 to 25 modified nucléotides of the first nucléotide sequence are 2’-(9-methyl nucléotides. In some embodiments, between about 10 to 25 modified nucléotides of the first nucléotide sequence are 2’-(9-methyl nucléotides. In some embodiments, between about 12 to 25 modified nucléotides of the first nucléotide sequence are 2’-(9-methyl nucléotides. In some embodiments, at least 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, or 22 modified nucléotides of the first nucléotide sequence are 2’-(9-methyl nucléotides. In some embodiments, less than or equal to 25, 24, 23, 22, 21, 20, 19, 18, 17, 16, 15, 14, 13, 12, 11, 10, 9, 8, 7, 6, 5, 4, 3, or 2 modified nucléotides of the first nucléotide sequence are 2’-O-methyl nucléotides.
In some embodiments, the second nucléotide sequence comprises 16, 17, 18, 19, 20, 21, 22, 23, or more modified nucléotides independently selected from a 2’-O-methyl nucléotide and a 2'-fluoro nucléotide. In some embodiments, 70%, 75%, 80%, 85%, 90%, 95% or 100% of the nucléotides in the second nucléotide sequence are modified nucléotides independently selected from a 2’-O-methyl nucléotide and a 2’-fluoro nucléotide. In some embodiments, between 2 to 15 modified nucléotides of the second nucléotide sequence are 2’-fluoro nucléotides. In some embodiments, between 2 to 10 modified nucléotides of the second nucléotide sequence are 2’fluoro nucléotides. In some embodiments, between 2 to 6 modified nucléotides of the second nucléotide sequence are 2’-fluoro nucléotides. In some embodiments, at least 2, 3, 4, 5, or 6 modified nucléotides of the second nucléotide sequence are 2’-fluoro nucléotides. In some embodiments, less than or equal to 10, 9, 8, 7, 6, 5, 4, 3, or 2 modified nucléotides of the second nucléotide sequence are 2’-fluoro nucléotides. In some embodiments, between about 2 to 25 modified nucléotides of the second nucléotide sequence are 2’-(9-methyl nucléotides. In some embodiments, between about 2 to 20 modified nucléotides of the second nucléotide sequence are 2’-O-methyl nucléotides. In some embodiments, between about 5 to 25 modified nucléotides of the second nucléotide sequence are 2’-O-methyl nucléotides. In some embodiments, between about 10 to 25 modified nucléotides of the second nucléotide sequence are 2’-(9-methyl nucléotides. In some embodiments, between about 12 to 25 modified nucléotides of the second nucléotide sequence are 2’-(9-methyl nucléotides. In some embodiments, at least 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, or 22 modified nucléotides of the second nucléotide sequence are 2’-(?-methyl nucléotides. In some embodiments, less than or equal to 25, 24, 23, 22, 21, 20, 19, 18, 17, 16, 15, 14, 13, 12, 11, 10, 9, 8, 7, 6, 5, 4, 3, or 2 modified nucléotides of the second nucléotide sequence are 2’-O-methyl nucléotides.
Disclosed herein is a short interfering nucleic acid (siNA) molécule comprising: (a) a sense strand comprising a first nucléotide sequence that is at least about 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% identical to an RNA corresponding to a target gene, wherein the first nucléotide sequence: (i) is 15 to 30 nucléotides in length; (ii) comprises 15 or more modified nucléotides independently selected from a 2’-O-methyl nucléotide and a 2’-fluoro nucléotide, wherein at least one modified nucléotide is a 2’-O-methyl nucléotide and at least one modified nucléotide is a 2’-fluoro nucléotide; and (iii) comprises 1 or more phosphorothioate intemucleoside linkage; and (b) an antisense strand comprising a second nucléotide sequence that is at least about 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% complementary to the RNA corresponding to the target gene, wherein the second nucléotide sequence: (i) is 15 to 30 nucléotides in length; (ii) comprises 15 or more modified nucléotides independently selected from a 2’-(9-methyl nucléotide and a 2’-fluoro nucléotide, wherein at least one modified nucléotide is a 2’-(9-methyl nucléotide and at least one modified nucléotide is a 2’-fluoro nucléotide; and (iii) comprises 1 or more phosphorothioate intemucleoside linkage.
Disclosed herein is a short interfering nucleic acid (siNA) molécule comprising: (a) a sense strand comprising a first nucléotide sequence that is at least about 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% identical to an RNA corresponding to a target gene, wherein the first nucléotide sequence: (i) is 15 to 30 nucléotides in length; and (ii) comprises 15 or more modified nucléotides independently selected from a 2’-O-methyl nucléotide and a 2’-fluoro nucléotide, wherein at least one modified nucléotide is a 2’-O-methyl nucléotide and at least one modified nucléotide is a 2’-fluoro nucléotide; and (b) an antisense strand comprising a second nucléotide sequence that is at least about 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% complementary to the RNA corresponding to the target gene, wherein the second nucléotide sequence: (i) is 15 to 30 nucléotides in length; and (ii) comprises 15 or more modified nucléotides independently selected from a 2’-O-methyl nucléotide and a 2’-fluoro nucléotide, wherein at least one modified nucléotide is a 2’-<9-methyl nucléotide and at least one modified nucléotide is a 2’-fluoro nucléotide, wherein the siNA further comprises a phosphorylation blocker, a galactosamine, or 5’-stabilized end cap.
In some embodiments, at least 1, 2, 3, 4, 5, 6, or 7 nucléotides at position 3, 5, 7, 8, 9, 10, 11, 12, and/or 17 from the 5’ end of the first nucléotide sequence is a 2’-fluoro nucléotide. In some embodiments, the nucléotide at position 3 from the 5’ end of the first nucléotide sequence is a 2’-fluoro nucléotide. In some embodiments, the nucléotide at position 5 from the 5’ end of the first nucléotide sequence is a 2’-fluoro nucléotide. In some embodiments, the nucléotide at position 7 from the 5’ end of the first nucléotide sequence is a 2’-fluoro nucléotide. In some embodiments, the nucléotide at position 8 from the 5’ end of the first nucléotide sequence is a 2’fluoro nucléotide. In some embodiments, the nucléotide at position 9 from the 5’ end of the first nucléotide sequence is a 2’-fluoro nucléotide. In some embodiments, the nucléotide at position 12 from the 5’ end of the first nucléotide sequence is a 2’-fluoro nucléotide. In some embodiments, the nucléotide at position 17 from the 5’ end of the first nucléotide sequence is a 2’-fluoro nucléotide. In some embodiments, nucléotide at position 10 from the 5’ end of the first nucléotide sequence is a 2’-fluoro nucléotide. In some embodiments, the nucléotide at position 11 from the 5’ end of the first nucléotide sequence is a 2’-fluoro nucléotide.
In some embodiments, at least 1, 2, 3, 4, 5, 6, 7, 8, or 9 nucléotides at position 2, 5, 6, 8, 10, 14, 16, 17, and/or 18 from the 5’ end of the second nucléotide sequence is a 2’-fluoro nucléotide. In some embodiments, the nucléotide at position 2 from the 5’ end of the second nucléotide sequence is a 2’-fluoro nucléotide. In some embodiments, the nucléotide at position 5 from the 5’ end of the second nucléotide sequence is a 2’-fluoro nucléotide. In some embodiments, the nucléotide at position 6 from the 5’ end of the second nucléotide sequence is a 2’-fluoro nucléotide. In some embodiments, the nucléotide at position 8 from the 5’ end of the second nucléotide sequence is a 2’-fluoro nucléotide. In some embodiments, the nucléotide at position 10 from the 5’ end of the second nucléotide sequence is a 2’-fluoro nucléotide. In some embodiments, the nucléotide at position 14 from the 5’ end of the second nucléotide sequence is a 2’-fluoro nucléotide. In some embodiments, the nucléotides at position 16 from the 5’ end of the second nucléotide sequence is a 2’-fluoro nucléotide. In some embodiments, the nucléotide at position 17 from the 5’ end of the second nucléotide sequence is a 2’-fluoro nucléotide. In some embodiments, the nucléotide at position 18 from the 5’ end of the second nucléotide sequence is a 2’-fluoro nucléotide.
In some embodiments, the nucléotides in the second nucléotide sequence are arranged in an altemating 1:3 modification pattern, and wherein 1 nucléotide is a 2’-fluoro nucléotide and 3 nucléotides are 2’-(9-methyl nucléotides. In some embodiments, the altemating 1:3 modification pattern occurs 2-5 times. In some embodiments, at least two of the altemating 1:3 modification pattern occur consecutively. In some embodiments, at least two of the altemating 1:3 modification pattern occurs nonconsecutively. In some embodiments, at least 1, 2, 3, 4, or 5 altemating 1:3 modification pattern begins at nucléotide position 2, 6, 10, 14, and/or 18 ffom the 5’ end of the antisense strand. In some embodiments, at least one altemating 1:3 modification pattern begins at nucléotide position 2 from the 5’ end of the antisense strand. In some embodiments, at least one altemating 1:3 modification pattern begins at nucléotide position 6 from the 5’ end of the antisense strand. In some embodiments, at least one altemating 1:3 modification pattern begins at nucléotide position 10 ffom the 5’ end of the antisense strand. In some embodiments, at least one altemating 1:3 modification pattern begins at nucléotide position 14 from the 5’ end of the antisense strand. In some embodiments, at least one altemating 1:3 modification pattern begins at nucléotide position 18 from the 5’ end of the antisense strand.
In some embodiments, the nucléotides in the second nucléotide sequence are arranged in an altemating 1:2 modification pattern, and wherein 1 nucléotide is a 2’-fluoro nucléotide and 2 nucléotides are 2’-O-methyl nucléotides. In some embodiments, the altemating 1:2 modification pattern occurs 2-5 times. In some embodiments, at least two of the altemating 1:2 modification pattern occurs consecutively. In some embodiments, at least two of the altemating 1:2 modification pattern occurs nonconsecutively. In some embodiments, at least 1, 2, 3, 4, or 5 altemating 1:2 modification pattern begins at nucléotide position 2, 5, 8, 14, and/or 17 from the 5’ end of the antisense strand. In some embodiments, at least one altemating 1:2 modification pattern begins at nucléotide position 2 from the 5’ end of the antisense strand. In some embodiments, at least one altemating 1:2 modification pattern begins at nucléotide position 5 from the 5’ end of the antisense strand. In some embodiments, at least one altemating 1:2 modification pattern begins at nucléotide position 8 from the 5’ end of the antisense strand. In some embodiments, at least one altemating 1:2 modification pattern begins at nucléotide position 14 from the 5’ end of the antisense strand. In some embodiments, at least one altemating 1:2 modification pattern begins at nucléotide position 17 from the 5’ end of the antisense strand.
Disclosed herein is a short interfering nucleic acid (siNA) molécule represented by Formula (VIII):
’-An%2An3Bn4An5Bn6An7Bn8An9-3 ’ 3’-Cq1Aq2Bq3Aq4Bq5Aq6Bq7Aq8Bq9Aq10Bq11Aq12-5’ wherein:
the top strand is a sense strand comprising a first nucléotide sequence that is at least about 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% identical to an RNA corresponding to a target gene, wherein the first nucléotide sequence comprises 15 to 30 nucléotides;
the bottom strand is an antisense strand comprising a second nucléotide sequence that is at least about 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% complementary to the RNA corresponding to the target gene, wherein the second nucléotide sequence comprises 15 to 30 nucléotides;
each A is independently a 2’-O-methyl nucléotide or a nucléotide comprising a 5’-stabilized end cap or a phosphorylation blocker;
B is a 2’-fluoro nucléotide;
C represents overhanging nucléotides and is a 2’-O-methyl nucléotide;
n1 = 1-4 nucléotides in length;
each n2, n6, n8, q3, q5, q7, q9, q11, and q12 is independently 0-1 nucléotides in length;
each n3 and n4 is independently 1-3 nucléotides in length;
n5 is 1-10 nucléotides in length;
n7 is 0-4 nucléotides in length;
each n9, q1, and q2 is independently 0-2 nucléotides in length;
q4 is 0-3 nucléotides in length;
q6 is 0-5 nucléotides in length;
q8 is 2-7 nucléotides in length; and q10 is 2-11 nucléotides in length.
Disclosed herein is a short interfering nucleic acid (siNA) molécule represented by
Formula (IX):
5’-A2-4BiAi-3 B2-3 A2-ioBo-iAo-4Bo-iAo-2-3’
3’-C2Ao-2Bo-iAo-3Bo-iAo-5Bo-iA2-7BiA2-iiBiAi-5’ wherein:
the top strand is a sense strand comprising a first nucléotide sequence that is at least about 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% identical to an RNA corresponding to a target gene, wherein the first nucléotide sequence comprises 15 to 30 nucléotides;
the bottom strand is an antisense strand comprising a second nucléotide sequence that is at least about 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% complementary to the RNA corresponding to the target gene, wherein the second nucléotide sequence comprises 15 to 30 nucléotides;
each A is independently a 2’-(9-methyl nucléotide or a nucléotide comprising a 5’-stabilized end cap or a phosphorylation blocker;
B is a 2’-fluoro nucléotide;
C represents overhanging nucléotides and is a 2’-O-methyl nucléotide.
Disclosed herein is a short interfering nucleic acid (siNA) molécule comprising (a) a sense strand comprising a first nucléotide sequence consisting of 17 to 23 nucléotides, wherein 2’-fluoro nucléotides are at positions 3,7-9, 12, and 17 from the 5’ end of the first nucléotide sequence, and wherein 2’-O-methyl nucléotides are at positions 1, 2, 4-6, 10, 11, and 13-16 from the 5’ end of the first nucléotide sequence; and (b) an antisense strand comprising a second nucléotide sequence consisting of 17 to 23 nucléotides, wherein 2’-fluoro nucléotides are at positions 2 and 14 from the 5’ end of the second nucléotide sequence, and wherein 2’-O-methyl nucléotides are at positions 1, 3-13, and 15-17 from the 5’ end of the second nucléotide sequence. In some embodiments, the first nucléotide sequence consists of 19 nucléotides. In some embodiments, 2’-O-methyl nucléotides are at positions 18 and 19 from the 5’ end of the first nucléotide sequence. In some embodiments, the second nucléotide sequence consists of 21 nucléotides. In some embodiments, 2’-O-methyl nucléotides are at positions 18-21 from the 5’ end of the second nucléotide sequence.
Disclosed herein is a short interfering nucleic acid (siNA) molécule comprising (a) a sense strand comprising a first nucléotide sequence consisting of 17 to 23 nucléotides, wherein 2’-fluoro nucléotides are at positions 3, 7, 8, and 17 from the 5’ end of the first nucléotide sequence, and wherein 2’-O-methyl nucléotides are at positions 1, 2, 4-6, and 9-16 from the 5’ end of the first nucléotide sequence; and (b) an antisense strand comprising a second nucléotide sequence consisting of 17 to 23 nucléotides, wherein 2’-fluoro nucléotides are at positions 2 and 14 from the 5’ end of the first nucléotide sequence; and wherein 2’-(9-methyl nucléotides are at positions 1,3-13, and 15-17 from the 5’ end of the first nucléotide sequence. In some embodiments, the first nucléotide sequence consists of 19 nucléotides. In some embodiments, 2’O-methyl nucléotides are at positions 18 and 19 from the 5’ end of the first nucléotide sequence. In some embodiments, the second nucléotide sequence consists of 21 nucléotides. In some embodiments, 2’-O-methyl nucléotides are at positions 18-21 from the 5’ end of the second nucléotide sequence.
Disclosed herein is a short interfering nucleic acid (siNA) molécule comprising (a) a sense strand comprising a first nucléotide sequence consisting of 17 to 23 nucléotides, wherein 2’-fluoro nucléotides are at positions 3, 7-9, 12 and 17 from the 5’ end of the first nucléotide sequence, and wherein 2’-O-methyl nucléotides are at positions 1, 2, 4-6, 10, 11, and 13-16 from the 5’ end of the first nucléotide sequence; and (b) an antisense strand comprising a second nucléotide sequence consisting of 17 to 23 nucléotides, wherein the nucléotides in the second nucléotide sequence are arranged in an altemating 1:3 modification pattern, and wherein 1 nucléotide is a 2’-fluoro nucléotide and 3 nucléotides are 2’-O-methyl nucléotides. In some embodiments, the first nucléotide sequence consists of 19 nucléotides. In some embodiments, 2’O-methyl nucléotides are at positions 18 and 19 from the 5’ end of the first nucléotide sequence. In some embodiments, the second nucléotide sequence consists of 21 nucléotides. In some embodiments, 2’-O-methyl nucléotides are at positions 19-21 from the 5’ end of the second nucléotide sequence. In some embodiments, the altemating 1:3 modification pattern occurs 2-5 times. In some embodiments, at least two of the altemating 1:3 modification pattern occur consecutively. In some embodiments, at least two of the altemating 1:3 modification pattern occurs nonconsecutively. In some embodiments, at least 1, 2, 3, 4, or 5 altemating 1:3 modification pattern begins at nucléotide position 2, 6, 10, 14, and/or 18 from the 5’ end of the antisense strand. In some embodiments, at least one altemating 1:3 modification pattern begins at nucléotide position 2 from the 5’ end of the antisense strand. In some embodiments, at least one altemating 1:3 modification pattern begins at nucléotide position 6 from the 5’ end of the antisense strand. In some embodiments, at least one altemating 1:3 modification pattern begins at nucléotide position 10 from the 5’ end of the antisense strand. In some embodiments, at least one altemating 1:3 modification pattern begins at nucléotide position 14 from the 5’ end of the antisense strand. In some embodiments, at least one altemating 1:3 modification pattern begins at nucléotide position 18 from the 5’ end of the antisense strand.
Disclosed herein is a short interfering nucleic acid (siNA) molécule comprising (a) a sense strand comprising a first nucléotide sequence consisting of 17 to 23 nucléotides, wherein 2’-fluoro nucléotides are at positions 5 and 7-9 from the 5’ end of the first nucléotide sequence, and wherein 2’-O-methyl nucléotides are at positions 1-4, 6, and 10-17 from the 5’ end of the first nucléotide sequence; and (b) an antisense strand comprising a second nucléotide sequence consisting of 17 to 23 nucléotides, wherein the nucléotides in the second nucléotide sequence are arranged in an altemating 1:3 modification pattern, and wherein 1 nucléotide is a 2’-fluoro nucléotide and 3 nucléotides are 2’-(9-methyl nucléotides. In some embodiments, the first nucléotide sequence consists of 19 nucléotides. In some embodiments, 2’-O-methyl nucléotides are at positions 18 and 19 from the 5’ end of the first nucléotide sequence. In some embodiments, the second nucléotide sequence consists of 21 nucléotides. In some embodiments, 2’-<9-methyl nucléotides are at positions 19-21 from the 5’ end of the second nucléotide sequence. In some embodiments, the altemating 1:3 modification pattern occurs 2-5 times. In some embodiments, at least two of the altemating 1:3 modification pattern occur consecutively. In some embodiments, at least two of the altemating 1:3 modification pattern occurs nonconsecutively. In some embodiments, at least 1, 2, 3, 4, or 5 altemating 1:3 modification pattern begins at nucléotide position 2, 6, 10, 14, and/or 18 from the 5’ end of the antisense strand. In some embodiments, at least one altemating 1:3 modification pattern begins at nucléotide position 2 from the 5’ end of the antisense strand. In some embodiments, at least one altemating 1:3 modification pattern begins at nucléotide position 6 from the 5’ end of the antisense strand. In some embodiments, at least one altemating 1:3 modification pattern begins at nucléotide position 10 from the 5’ end of the antisense strand. In some embodiments, at least one altemating 1:3 modification pattern begins at nucléotide position 14 from the 5’ end of the antisense strand. In some embodiments, at least one altemating 1:3 modification pattern begins at nucléotide position 18 from the 5’ end of the antisense strand.
Disclosed herein is a short interfering nucleic acid (siNA) molécule comprising (a) a sense strand comprising a first nucléotide sequence consisting of 17 to 23 nucléotides, wherein 2’-fluoro nucléotides are at positions 5 and 7-9 from the 5’ end of the first nucléotide sequence, and wherein 2’-O-methyl nucléotides are at positions 1-4, 6, and 10-17 from the 5’ end of the first nucléotide sequence; and (b) an antisense strand comprising a second nucléotide sequence consisting of 17 to 23 nucléotides, wherein the nucléotides in the second nucléotide sequence are arranged in an altemating 1:2 modification pattern, and wherein 1 nucléotide is a 2’-fluoro nucléotide and 2 nucléotides are 2’-O-methyl nucléotides. In some embodiments, the first nucléotide sequence consists of 19 nucléotides. In some embodiments, 2’-(9-methyl nucléotides are at positions 18 and 19 from the 5’ end of the first nucléotide sequence. In some embodiments, the second nucléotide sequence consists of 21 nucléotides. In some embodiments, 2’-O-methyl nucléotides are at positions 18-21 from the 5’ end of the second nucléotide sequence. In some embodiments, the altemating 1:2 modification pattern occurs 2-5 times. In some embodiments, at least two of the altemating 1:2 modification pattern occur consecutively. In some embodiments, at least two of the altemating 1:2 modification pattern occurs nonconsecutively. In some embodiments, at least 1, 2, 3, 4, or 5 altemating 1:2 modification pattern begins at nucléotide position 2, 5, 8, 14, and/or 17 from the 5’ end of the antisense strand. In some embodiments, at least one altemating 1:2 modification pattern begins at nucléotide position 2 from the 5’ end of the antisense strand. In some embodiments, at least one altemating 1:2 modification pattern begins at nucléotide position 5 from the 5’ end of the antisense strand. In some embodiments, at least one altemating 1:2 modification pattern begins at nucléotide position 8 from the 5’ end of the antisense strand. In some embodiments, at least one altemating
1:2 modification pattern begins at nucléotide position 14 from the 5’ end of the antisense strand. In some embodiments, at least one altemating 1:2 modification pattern begins at nucléotide position 17 from the 5’ end of the antisense strand.
Disclosed herein is a short interfering nucleic acid (siNA) molécule comprising (a) a sense strand comprising a first nucléotide sequence consisting of 17 to 23 nucléotides, wherein 2’-fluoro nucléotides are at positions 5 and 7-9 from the 5’ end of the first nucléotide sequence, and wherein 2’-O-methyl nucléotides are at positions 1-4, 6, and 10-17 from the 5’ end of the first nucléotide sequence; and (b) an antisense strand comprising a second nucléotide sequence consisting of 17 to 23 nucléotides, wherein 2’-fluoro nucléotides are at positions 2, 6, 14, and 16 from the 5’ end of the second nucléotide sequence, and wherein 2’-O-methyl nucléotides are at positions 1, 3-5, 7-13, 15, and 17 from the 5’ end the second nucléotide sequence. In some embodiments, the first nucléotide sequence consists of 19 nucléotides. In some embodiments, 2’O-methyl nucléotides are at positions 18 and 19 from the 5’ end of the first nucléotide sequence. In some embodiments, the second nucléotide sequence consists of 21 nucléotides. In some embodiments, 2’-(9-methyl nucléotides are at positions 18-21 from the 5’ end of the second nucléotide sequence.
Disclosed herein is a short interfering nucleic acid (siNA) molécule comprising: (a) a sense strand comprising a first nucléotide sequence consisting of 17 to 23 nucléotides, wherein 2’-fluoro nucléotides are at positions 5, 9-11, and 14 from the 5’ end of the first nucléotide sequence, and wherein 2’-O-methyl nucléotides are at positions 1-4, 6-8, and 12-17 from the 5’ end of the first nucléotide sequence; and (b) an antisense strand comprising a second nucléotide sequence consisting of 17 to 23 nucléotides, wherein 2’-fluoro nucléotides are at positions 2 and 14 from the 5’ end of the second nucléotide sequence, and wherein 2’-O-methyl nucléotides are at positions 1, 3-13, and 15-17 from the 5’ end the second nucléotide sequence. In some embodiments, the first nucléotide sequence consists of 21 nucléotides. In some embodiments, 2’(9-methyl nucléotides are at positions 18-21 from the 5’ end of the first nucléotide sequence. In some embodiments, the second nucléotide sequence consists of 23 nucléotides. In some embodiments, 2’-O-methyl nucléotides are at positions 18-23 from the 5’ end of the second nucléotide sequence.
In some embodiments, any of the sense strands disclosed herein further comprise a TT sequence adjacent to the first nucléotide sequence.
In some embodiments, any of the sense strands disclosed herein further comprise at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15 or more phosphorothioate intemucleoside linkages. In some embodiments, at least one phosphorothioate intemucleoside linkage is between the nucléotides at positions 1 and 2 from the 5’ end of the first nucléotide sequence. In some embodiments, at least one phosphorothioate intemucleoside linkage is between the nucléotides at positions 2 and 3 from the 5’ end of the first nucléotide sequence.
In some embodiments, any of the antisense strands disclosed herein further comprise TT sequence adjacent to the second nucléotide sequence. In some embodiments, the antisense strand further comprises at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15 or more phosphorothioate intemucleoside linkages. In some embodiments, at least one phosphorothioate intemucleoside linkage is between the nucléotides at positions 1 and 2 from the 5’ end of the second nucléotide sequence. In some embodiments, at least one phosphorothioate intemucleoside linkage is between the nucléotides at positions 2 and 3 from the 5’ end of the second nucléotide sequence. In some embodiments, at least one phosphorothioate intemucleoside linkage is between the nucléotides at positions 1 and 2 from the 3’ end of the second nucléotide sequence. In some embodiments, at least one phosphorothioate intemucleoside linkage is between the nucléotides at positions 2 and 3 from the 3 ’ end of the second nucléotide sequence.
In some embodiments, the first nucléotide from the 5’ end of any of the first nucléotide sequences disclosed herein comprises a 5’ stabilizing end cap.
In some embodiments, the first nucléotide from the 5’ end of any of the second nucléotide sequences disclosed herein comprise a 5’ stabilizing end cap.
' In some embodiments, the first nucléotide from the 5’ end of any of the first nucléotide sequences dislosed herein comprises a phosphorylation blocker.
In some embodiments, the first nucléotide from the 5’ end of any of the second nucléotide sequences dislosed herein comprises a phosphorylation blocker.
In some embodiments, any of the first nucléotide sequences or second nucléotide sequences disclosed herein comprise at least one modified nucléotide selected from
O<=O -Æo -oy-N (LNA), (ScpBNA or “cp”); 0 (AmNA), where R is H
or alkyl (or AmNA(N-Me)) when R is alkyl);
h2n (GuNA); and
GuNA(N-R), R = Me, Et, iPr, tBu, wherein B is a nucleobase.
Disclosed herein is a short-interfering nucleic acid (siNA) molécule comprising: (a) a R4\Î/R1 phosphorylation blocker of Formula (IV): ~J— , wherein
R1 is a nucleobase,
R4is -O-R30 or-NR31R32,
R30 is Ci-Cs substituted or unsubstituted alkyl; and
R31 and R32 together with the nitrogen to which they are attached form a substituted or unsubstituted heterocyclic ring; and (b) a short interfering nucleic acid (siNA). In some embodiments, the siNA is any of the siNAs disclosed herein. In some embodiments, the siNA comprises any of the sense strands, first nucléotide sequences, antisense strands, or second nucléotide sequences disclosed herein. In some embodiments, the siNA comprises any of the the sense strands disclosed herein. In some embodiments, the siNA comprises any of the antisense strand disclosed herein. In some embodiments, the siNA comprises a first nucléotide sequence selected from any one of SEQ ID NOs: 1-56, 103-158, and 205-260. In some embodiments, the siNA comprises a second nucléotide sequence selected from any one of SEQ ID NOs: 57-102, 159-204, and 261-306. In some embodiments, the siNA comprises a sense sequence selected from any one of SEQ ID NOs: 307-362 and 415-444. In some embodiments, the siNA comprises an antisense sequence selected from any one of SEQ ID NOs: 363-409, 445-533, and 536-539. In some embodiments, the siNA comprises a ds-siNA sequence selected from any one of ds-siNA-001 to ds-siNA-0178. In some embodiments, the siNA further comprises any of the 5’ end caps disclosed herein. In some embodiments, the siNA further comprises any of the conjugated moieties disclosed herein. In some embodiments, the siNA further comprises any of the destabilizing nucléotides disclosed herein. In some embodiments, the siNA further comprises any of the modified nucléotides disclosed herein.
Disclosed herein is a short-interfering nucleic acid (siNA) molécule comprising: (a) a 5’ stabilized end cap of Formula (la):
wherein
R1 is a nucleobase, aryl, heteroaryl, or H,
H
H ,N. / i S // Λ
O O 5 zOCD3 o , -CH=CD-Z, -CD=CH-Z, -CD=CD-Z, -(CR2IR22)n-Z, or -(C2-C6 alkenylene)-Z and R20 is hydrogen; or
R2 and R20 together form a 3- to 7-membered carbocyclic ring substituted with -(CR21R22)n-Z or
-(C2-C6 alkenylene)-Z;
n is 1, 2, 3, or 4;
Z is -ONR23R24, -OP(O)OH(CH2)mCO2R23, -OP(S)OH(CH2)mCO2R23, -P(O)(OH)2, P(O)(OH)(OCH3), -P(O)(OH)(OCD3), -SO2(CH2)mP(O)(OH)2, -SO2NR23R25, -nr23r24, R21 and R22 are independently hydrogen or Ci-Cô alkyl; R21 and R22 together form an oxo group; R23 is hydrogen or Ci-Cô alkyl;
R24 is -SO2R25 or -C(O)R25; or
R23 and R24 together with the nitrogen to which they are attached form a substituted or unsubstituted heterocyclic ring;
R25 is Ci-Cô alkyl; and m is 1, 2, 3, or 4; and (b) a short interfering nucleic acid (siNA). In some embodiments, the siNA comprises any of the the sense strands disclosed herein. In some embodiments, the siNA comprises any of the antisense strand disclosed herein. In some embodiments, the siNA comprises a first nucléotide sequence selected from any one of SEQ ID NOs: 1-56, 103-158, and 205-260. In some embodiments, the siNA comprises a second nucléotide sequence selected from any one of SEQ 14
ID NOs: 57-102, 159-204, and 261-306. In some embodiments, the siNA comprises a sense sequence selected from any one of SEQ ID NOs: 307-362 and 415-444. In some embodiments, the siNA comprises an antisense sequence selected from any one of SEQ ID NOs: 363-409, 445533, and 536-539. In some embodiments, the siNA comprises a ds-siNA sequence selected from any one of ds-siNA-001 to ds-siNA-0178. In some embodiments, the siNA further comprises any of the phosphorylation blockers disclosed herein. In some embodiments, the siNA further comprises any of the conjugated moieties disclosed herein. In some embodiments, the siNA further comprises any of the destabilizing nucléotides disclosed herein. In some embodiments, the siNA further comprises any of the modified nucléotides disclosed herein.
Disclosed herein is a short-interfering nucleic acid (siNA) molécule comprising: (a) a 5’ stabilized end cap of Formula (Ib):
wherein
R1 is a nucleobase, aryl, heteroaryl, or H,
O
II
PX-OH OH
HO, ZS 9 O, OH O, OCH3
.. pz J-L p Y
Y O ^ OH Y YH Y YH
O,, zocd3
-CH=CD-Z, -CD=CH-Z, -CD=CD-Z, -(CR21R22)n-Z, or -(C2-C6 alkenylene)-Z and R20 is hydrogen; or
R2 and R20 together form a 3- to 7-membered carbocyclic ring substituted with -(CR21R22)n-Z or
-(C2-C6 alkenylene)-Z;
n is 1, 2, 3, or 4;
Z is -ONR23R24, -OP(O)OH(CH2)mCO2R23, -OP(S)OH(CH2)mCO2R23, -P(O)(OH)2, P(O)(OH)(OCH3), -P(O)(OH)(OCD3), -SO2(CH2)mP(O)(OH)2, -SO2NR23R25, -nr23r24,
R21 and R22 are independently hydrogen or Ci-Cô alkyl; R21 and R22 together form an oxo group;
R23 is hydrogen or Ci-Cô alkyl;
R24 is -SO2R25 or -C(O)R25; or
R23 and R24 together with the nitrogen to which they are attached form a substituted or unsubstituted heterocyclic ring;
R25 is Ci-Cô alkyl; and m is 1, 2, 3, or 4; and (b) a short interfering nucleic acid (siNA). In some embodiments, the siNA comprises any of the the sense strands disclosed herein. In some embodiments, the siNA comprises any of the antisense strand disclosed herein. In some embodiments, the siNA comprises a first nucléotide sequence selected from any one of SEQ ID NOs: 1-56, 103-158, and 205-260. In some embodiments, the siNA comprises a second nucléotide sequence selected from any one of SEQ ID NOs: 57-102, 159-204, and 261-306. In some embodiments, the siNA comprises a sense sequence selected from any one of SEQ ID NOs: 307-362 and 415-444. In some embodiments, the siNA comprises an antisense sequence selected from any one of SEQ ID NOs: 363-409, 445533, and 536-539. In some embodiments, the siNA comprises a ds-siNA sequence selected from any one of ds-siNA-001 to ds-siNA-0178. In some embodiments, the siNA further comprises any of the phosphorylation blockers disclosed herein. In some embodiments, the siNA further comprises any of the conjugated moieties disclosed herein. In some embodiments, the siNA further comprises any of the destabilizing nucléotides disclosed herein. In some embodiments, the siNA further comprises any of the modified nucléotides disclosed herein.
Disclosed herein is a short-interfering nucleic acid (siNA) molécule comprising: (a) a 5’stabilized end cap selected from the group consisting of Formula (1) to Formula (15), Formula (9X) to Formula (12X), and Formula (9Y) to Formula (12Y):
Formula (8) Formula (9) Formula (9X) Formula (9Y)
Formula (10Y)
Formula (10)
Formula (10X)
Formula (11 Y)
Formula (11) Formula (11X)
Formula (12X) Formula (12Y)
Formula (12)
Formula (13)
Formula (14)
Formula (15) , wherein R1 is a nucleobase, aryl, heteroaryl, or H; and (b) a short interfering nucleic acid (siNA). In some embodiments, the siNA comprises any of the the sense strands disclosed herein. In some embodiments, the siNA comprises any of the antisense strand disclosed herein. In some embodiments, the siNA comprises a first nucléotide sequence selected from any one of SEQ ID NOs: 1-56, 103-158, and 205-260. In some embodiments, the siNA comprises a second nucléotide sequence selected from any one of SEQ ID NOs: 57-102, 159-204, and 261-306. In some embodiments, the siNA comprises a sense sequence selected from any one of SEQ ID NOs: 307-362 and 415-444. In some embodiments, the siNA comprises an antisense sequence selected from any one of SEQ ID NOs: 363-409, 445-533, and 536-539. In some embodiments, the siNA comprises a ds-siNA sequence selected from any one of ds-siNA-001 to ds-siNA-0178. In some embodiments, the siNA fiirther comprises any of the phosphorylation blockers disclosed herein. In some embodiments, the siNA fiirther comprises any of the conjugated moieties disclosed herein. In some embodiments, the siNA fiirther comprises any of the destabilizing nucléotides disclosed herein. In some embodiments, the siNA fiirther comprises any of the modified nucléotides disclosed herein.
Disclosed herein is a short-interfering nucleic acid (siNA) molécule comprising: (a) a 5’stabilized end cap selected from the group consisting of Formulas (1 A)-(15A), Formulas (9B)(12B), Formulas (9AX)-(12AX), Formulas (9AY)-(12AY), Formulas (9BX)-(12BX), and Formulas (9BY)-(12BY):
Formula (8A)
Formula (9A) Formula (9AX) Formula (9AY)
Formula (9B) Formula (9BX) Formula (9BY)
Formula (10A)
Formula (10AX)
Formula (10B)
Formula (10BX)
Formula (10BY)
Formula (11A)
Formula (11AX)
Formula (11AY)
Formula (11 B)
Formula (11BX)
Formula (11BY)
Formula (12AX)
Formula (12AY)
Formula (12A)
Formula (12BX)
Formula (12BY)
interfering nucleic acid (siNA). In some embodiments, the siNA comprises any of the the sense strands disclosed herein. In some embodiments, the siNA comprises any of the antisense strand disclosed herein. In some embodiments, the siNA comprises a first nucléotide sequence selected from any one of SEQ ID NOs: 1-56, 103-158, and 205-260. In some embodiments, the siNA comprises a second nucléotide sequence selected from any one of SEQ ID NOs: 57-102, 159204, and 261-306. In some embodiments, the siNA comprises a sense sequence selected from any one of SEQ ID NOs: 307-362 and 415-444. In some embodiments, the siNA comprises an antisense sequence selected from any one of SEQ ID NOs: 363-409, 445-533, and 536-539. In some embodiments, the siNA comprises a ds-siNA sequence selected from any one of ds-siNA001 to ds-siNA-0178. In some embodiments, the siNA further comprises any of the phosphorylation blockers disclosed herein. In some embodiments, the siNA further comprises any of the conjugated moieties disclosed herein. In some embodiments, the siNA further comprises any of the destabilizing nucléotides disclosed herein. In some embodiments, the siNA further comprises any of the modified nucléotides disclosed herein.
Disclosed herein is a short-interfering nucleic acid (siNA) molécule comprising: (a) a 5’ stabilized end cap of Formula (le):
wherein
R1 is a nucleobase, aryl, heteroaryl, or H,
O II PX-OH OH ZOH Οχ, ZOCH3 zOCD3 ? 5
HOX zzS 9
Y^op^A'oh
o , -CH=CD-Z, -CD=CH-Z, -CD=CD-Z, -(CR21R22)n-Z, or -(C2-C6 alkenylene)-Z and R20 is hydrogen; or
R2 and R20 together form a 3- to 7-membered carbocyclic ring substituted with -(CR21R22)n-Z or
-(C2-C6 alkenylene)-Z;
nis 1, 2, 3, or 4;
Z is -ONR23R24, -OP(O)OH(CH2)mCO2R23, -OP(S)OH(CH2)mCO2R23, -P(O)(OH)2, P(O)(OH)(OCH3), -P(O)(OH)(OCD3), -SO2(CH2)mP(O)(OH)2, -SO2NR23R25, -NR23R24, orNR23SO2R24;
R21 and R22 either are independently hydrogen or Ci-Ce alkyl, or R21 and R22 together form an oxo group;
R23 is hydrogen or Ci-Ce alkyl;
R24 is -SO2R25 or -C(O)R25; or
R23 and R24 together with the nitrogen to which they are attached form a substituted or unsubstituted heterocyclic ring;
R25 is Ci-C6 alkyl; and m is 1, 2, 3, or 4; and (b) a short interfering nucleic acid (siNA). In some embodiments, the siNA comprises any of the the sense strands disclosed herein. In some embodiments, the siNA comprises any of the antisense strand disclosed herein. In some embodiments, the siNA comprises a first nucléotide sequence selected from any one of SEQ ID NOs: 1-56, 103-158, and 205-260. In some embodiments, the siNA comprises a second nucléotide sequence selected from any one of SEQ ID NOs: 57-102, 159-204, and 261-306. In some embodiments, the siNA comprises a sense sequence selected from any one of SEQ ID NOs: 307-362 and 415-444. In some embodiments, the siNA comprises an antisense sequence selected from any one of SEQ ID NOs: 363-409, 445533, and 536-539. In some embodiments, the siNA comprises a ds-siNA sequence selected from any one of ds-siNA-001 to ds-siNA-0178. In some embodiments, the siNA further comprises any of the phosphorylation blockers disclosed herein. In some embodiments, the siNA further comprises any of the conjugated moieties disclosed herein. In some embodiments, the siNA further comprises any of the destabilizing nucléotides disclosed herein. In some embodiments, the siNA further comprises any of the modified nucléotides disclosed herein.
Disclosed herein is a short-interfering nucleic acid (siNA) molécule comprising: (a) a 5’stabilized end cap selected from the group consisting of Formula (21) to Formula (35):
Formula (25)
Formula (26)
Formula (27)
Formula (31) Formula (32) Formula (33)
Formula (34)
Formula (35) , wherein R1 is a nucleobase, aryl, heteroaryl, or
H; and (b) a short interfering nucleic acid (siNA). In some embodiments, the siNA comprises any of the the sense strands disclosed herein. In some embodiments, the siNA comprises any of the antisense strand disclosed herein. In some embodiments, the siNA comprises a first nucléotide sequence selected from any one of SEQ ID NOs: 1-56, 103-158, and 205-260. In some embodiments, the siNA comprises a second nucléotide sequence selected from any one of SEQ ID NOs: 57-102, 159-204, and 261-306. In some embodiments, the siNA comprises a sense sequence selected from any one of SEQ ID NOs: 307-362 and 415-444. In some embodiments, the siNA comprises an antisense sequence selected from any one of SEQ ID NOs: 363-409, 445533, and 536-539. In some embodiments, the siNA comprises a ds-siNA sequence selected from any one of ds-siNA-001 to ds-siNA-0178. In some embodiments, the siNA further comprises any of the phosphorylation blockers disclosed herein. In some embodiments, the siNA further comprises any of the conjugated moieties disclosed herein. In some embodiments, the siNA further comprises any of the destabilizing nucléotides disclosed herein. In some embodiments, the siNA further comprises any of the modified nucléotides disclosed herein.
Disclosed herein is a short-interfering nucleic acid (siNA) molécule comprising: (a) a 5’stabilized end cap selected from the group consisting of Formulas (21A)-(35A), Formulas (29B)(32B), Formulas (29AX)-(32AX), Formulas (29AY)-(32AY), Formulas (29BX)-(32BX), and Formulas (29BY)-(32BY):
Formula (28A)
Formula (29B)
Formula (29AY)
Formula (29A)
Formula (29AX)
Formula (29BX)
Formula (29BY)
Formula (31AX)
Formula (31BX)
interfering nucleic acid (siNA). In some embodiments, the siNA comprises any of the the sense strands disclosed herein. In some embodiments, the siNA comprises any of the antisense strand disclosed herein. In some embodiments, the siNA comprises a first nucléotide sequence selected from any one of SEQ ID NOs: 1-56, 103-158, and 205-260. In some embodiments, the siNA comprises a second nucléotide sequence selected from any one of SEQ ID NOs: 57-102, 159204, and 261-306. In some embodiments, the siNA comprises a sense sequence selected from any one of SEQ ID NOs: 307-362 and 415-444. In some embodiments, the siNA comprises an antisense sequence selected from any one of SEQ ID NOs: 363-409, 445-533, and 536-539. In some embodiments, the siNA comprises a ds-siNA sequence selected from any one of ds-siNA001 to ds-siNA-0178. In some embodiments, the siNA further comprises any of the phosphorylation blockers disclosed herein. In some embodiments, the siNA further comprises any of the conjugated moieties disclosed herein. In some embodiments, the siNA further comprises any of the destabilizing nucléotides disclosed herein. In some embodiments, the siNA further comprises any of the modified nucléotides disclosed herein.
Disclosed herein is a short interfering nucleic acid (siNA) molécule.comprising: (a) a sense strand comprising a first nucléotide sequence that is at least about 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% identical to an RNA corresponding to a target gene, wherein the first nucléotide sequence comprises a nucléotide sequence of any one SEQ ID NOs: 1-56, 103158, and 205-260; and (b) an antisense strand comprising a second nucléotide sequence that is at least about 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% complementary to the RNA corresponding to the target gene, wherein the second nucléotide sequence comprises a nucléotide sequence of any one of SEQ ID NOs: 57-102, 159-204, and 261-306. In some embodiments, the siNA further comprises any of the 5’ end caps disclosed herein. In some embodiments, the siNA fürther comprises any of the phosphorylation blockers disclosed herein. In some emboiments, the siNA further comprises any of the conjugated moieties disclosed herein. In some embodiments, the siNA further comprises any of the destabilizing nucléotides disclosed herein. In some embodiments, the siNA further comprises any of the modified nucléotides disclosed herein.
Disclosed herein is a interfering nucleic acid (siNA) molécule comprising: (a) a sense strand comprising a nucléotide sequence of any one of SEQ ID NOs: 307-362 and 415-444; and (b) an antisense strand comprising a nucléotide sequence of any one of SEQ ID NOs: 363-409, 445-533, and 536-539.
In some embodiments, any of the siNA disclosed herein further comprise a phosphorylation blocker.
In some embodiments, the phosphorylation blocker has the structure of Formula (IV):
, wherein
R1 is a nucleobase,
R4 is -O-R30 or -NR31R32, R30 is Ci-Cs substituted or unsubstituted alkyl; and
R31 and R32 together with the nitrogen to which they are attached form a substituted or unsubstituted heterocyclic ring.
In some embodiments, R4 is -OCH3 or -N(CH2CH2)2O.
In some embodiments, the phosphorylation blocker is attached to the 5’ end of the sense strand.
In some embodiments, the phosphorylation blocker is attached to the 5’ end of the sense strand via one or more linkers independently selected from a phosphodiester linker, phosphorothioate linker, and phosphorodithioate linker.
In some embodiments, the phosphorylation blocker is attached to the 3 ’ end of the sense strand.
In some embodiments, the phosphorylation blocker is attached to the 3’ end of the sense strand via one or more linkers independently selected from a phosphodiester linker, phosphorothioate linker, and phosphorodithioate linker.
In some embodiments, the phosphorylation blocker is attached to the 5’ end of the antisense strand. In some embodiments, the phosphorylation blocker is attached to the 5’ end of the antisense strand via one or more linkers independently selected from a phosphodiester linker, phosphorothioate linker, and phosphorodithioate linker. In some embodiments, the phosphorylation blocker is attached to the 3’ end of the antisense strand. In some embodiments, the phosphorylation blocker is attached to the 3 ’ end of the antisense strand via one or more linkers independently selected from a phosphodiester linker, phosphorothioate linker, and phosphorodithioate linker.
In some embodiments, any of the siNAs disclosed herein further comprise a conjugated moiety. In some embodiments, the conjugated moiety comprises a galactosamine. In some embodiments, the galactosamine is N-acetylgalactosamine (GalNAc) of Formula (VII):
R = OH or SH wherein each n is independently 1 or 2. In some embodiments, the galactosamine is Nacetylgalactosamine (GalNAc) of Formula (VI):
m is 1, 2, 3, 4, or 5;
each n is independently 1 or 2; p is 0 or 1 ;
each R is independently H;
each Y is independently selected from -O-P(=O)(SH)-, -O-P(=O)(O)-, -O-P(=O)(OH)-, and O-P(S)S-;
Z is H or a second protecting group;
either L is a linker or L and Y in combination are a linker; and
A is H, OH, a third protecting group, an activated group, or an oligonucleotide. In some embodiments, wherein A is an oligonucleotide. In some embodiments, A is 1-2 oligonucleotides. In some embodiments, the oligonucleotide is dTdT. In some embodiments, the galactosamine is attached to the 3’ end of the sense strand. In some embodiments, the galactosamine is attached to the 3 ’ end of the sense strand via one or more linkers independently selected from a phosphodiester linker, phosphorothioate linker, or phosphorodithioate linker. In some embodiments, the galactosamine is attached to the 5’ end of the sense strand. In some embodiments, the galactosamine is attached to the 5’ end of the sense strand via one or more linkers independently selected from a phosphodiester linker, phosphorothioate linker, or phosphorodithioate linker. In some embodiments, the galactosamine is attached to the 3 ’ end of the antisense strand. In some embodiments, the galactosamine is attached to the 3 ’ end of the atnisense strand via one or more linkers independently selected from a phosphodiester linker, phosphorothioate linker, or phosphorodithioate linker. In some embodiments, the galactosamine is attached to the 5’ end of the antisense strand. In some embodiments, the galactosamine is attached to the 5’ end of the atnisense strand via one or more linkers independently selected from a phosphodiester linker, phosphorothioate linker, or phosphorodithioate linker.
In some embodiments, any of the siNAs disclosed herein further comprise a 5’-stabilized end cap. In some embodiments, the 5’-stabilized end cap is a 5’ vinyl phosphonate or deuterated
5’ vinyl phosphonate. In some embodiments, the 5’-stabilized end cap has the structure of
R2
R20' 0'
Formula (la): — °\^R1 (DCH3 , wherein
R1 is a nucleobase, aryl, heteroaryl, or H,
O, ZO
R2 is H
9'P
H ;s-N o' SO
O. O
O, ZO
H o,xzp 9
J:s^px-oh
OH
HO, zs 9
O, OH ZOCH3
ΌΗ
Os OCD3 ^PxOH
H ,N. / 1 S // Ά
O O 5
O , -CH=CD-Z, -CD=CH-Z, -CD=CD-Z, -(CR21R22)n-Z, or -(C2-C6 alkenylene)-Z and R20 is hydrogen; or
R2 and R20 together form a 3- to 7-membered carbocyclic ring substituted with -(CR21R22)n-Z or
-(C2-C6 alkenylene)-Z;
n is 1, 2, 3, or 4;
Z is -ONR23R24, -OP(O)OH(CH2)mCO2R23, -OP(S)OH(CH2)mCO2R23, -P(O)(OH)2, P(O)(OH)(OCH3), -P(O)(OH)(OCD3), -SO2(CH2)mP(O)(OH)2, -SO2NR23R25, -NR23R24, orNR23SO2R24;
R21 and R22 either are independently hydrogen or Ci-Cô alkyl, or R21 and R22 together form an oxo group;
R23 is hydrogen or Ci-Cô alkyl;
R24 is -SO2R25 or-C(O)R25; or
R23 and R24 together with the nitrogen to which they are attached form a substituted or unsubstituted heterocyclic ring;
R25 is Ci-Cô alkyl; and m is 1, 2, 3, or 4. In some embodiments, the 5’-stabilized end cap has the structure of Formula rvr1 R2ovy
0' zocd3 (Ib): —I— , wherein
R1 is a nucleobase, aryl, heteroaryl, or H,
Zz° ° HO. ZS 9 Ox DH O, och3 O, ocd3 's'/R-OH , p' A u A . A _ A
Oh Y O OH V OH Y OH Y OH
O , -CH=CD-Z, -CD=CH-Z, -CD=CD-Z, -(CR21R22)n-Z, or -(C2-C6 alkenylene)-Z and R20 is hydrogen; or
R2 and R20 together form a 3- to 7-membered carbocyclic ring substituted with -(CR21R22)n-Z or
-(C2-Cô alkenylene)-Z;
n is 1, 2, 3, or 4;
Z is -ONR23R24, -OP(O)OH(CH2)mCO2R23, -OP(S)OH(CH2)mCO2R23, -P(O)(OH)2, P(O)(OH)(OCH3), -P(O)(OH)(OCD3), -SO2(CH2)mP(O)(OH)2, -SO2NR23R25, -NR23R24, orNR23SO2R24;
R21 and R22 either are independently hydrogen or Ci-Ce alkyl, or R21 and R22 together form an oxo group;
R23 is hydrogen or Ci-Ce alkyl;
R24 is -SO2R25 or -C(O)R25; or
R23 and R24 together with the nitrogen to which they are attached form a substituted or unsubstituted heterocyclic ring;
R25 is Ci-Cô alkyl; and m is 1, 2, 3, or 4. In some embodiments, the 5’-stabilized end cap has the structure of Formula
0' F (le): —L- , wherein
R1 is a nucleobase, aryl, heteroaryl, or H,
Os θ HO. ZS 9 ox OH O. och3 O. ocd3
Js^r-oh x A A
OH V O OH Y OH Y OH Y OH ? 9 5 5
Ο , -CH=CD-Z, -CD=CH-Z, -CD=CD-Z, -(CR2 ‘R22)n-Z, or -(C2-C6 alkenylene)-Z and R20 is hydrogen; or
R2 and R20 together form a 3- to 7-membered carbocyclic ring substituted with -(CR21R22)n-Z or
-(C2-C6 alkenylene)-Z;
n is 1, 2, 3, or 4;
Z is -ONR23R24, -OP(O)OH(CH2)mCO2R23, -OP(S)OH(CH2)mCO2R23, -P(O)(OH)2, P(O)(OH)(OCH3), -P(O)(OH)(OCD3), -SO2(CH2)mP(O)(OH)2, -SO2NR23R25, -NR23R24, orNR23SO2R24;
R21 and R22 either are independently hydrogen or Ci-Cô alkyl, or R21 and R22 together form an oxo group;
R23 is hydrogen or Ci-Cô alkyl;
R24 is -SO2R25 or -C(O)R25; or
R23 and R24 together with the nitrogen to which they are attached form a substituted or unsubstituted heterocyclic ring;
R25 is Ci-Cô alkyl; and m is 1, 2, 3, or 4. In some embodiments, R1 is an aryl. In some embodiments, the aryl is a phenyl. In some embodiments, the 5’-stabilized end cap is selected from the group consisting of Formula (1) to Formula (15), Formula (9X) to Formula (12X), and Formula (9Y) to Formula (12Y):
Formula (5)
Formula (6)
Formula (7)
Formula (11) Formula (11X) Formula (11 Y)
Formula (12) Formula (12X) Formula (12Y)
Formula (13) Formula (14)
Formula (15) , wherein R1 independently is a nucleobase, aryl, heteroaryl, or H. In some embodiments, the 5’-stabilized end cap is selected from the group consisting of Formulas (1A)-(15A), Formulas (9B)-(12B), Formulas (9AX)-(12AX), Formulas (9AY)-(12AY), Formulas (9BX)-(12BX), and Formulas (9BY)(12BY):
Formula (5A)
Formula (8A)
Formula (9AX) Formula (9AY)
Formula (9B)
Formula (10A)
Formula (9A)
Formula (9BX)
cf Î)CH3
Formula (9BY)
Formula (10B) Formula (10BX) Formula (10BY)
Formula (11AX)
Formula (11 B)
Formula (11BX)
Formula (11BY)
Formula (12A) Formula (12AX) Formula (12AY)
the 5’-stabilized end cap is selected from the group consisting of Formula (21) to Formula (35):
Formula (25)
Formula (26)
Formula (27)
Formula (28) Formula (29) Formula (30)
Formula (31) Formula (32) Formula (33)
Formula (34) Formula (35) , wherein R1 is a nucleobase, aryl, heteroaryl, or
H. In some embodiments, the 5’-stabilized end cap is selected from the group consisting of
Formulas (21A)-(35A), Formulas (29B)-(32B), Formulas (29AX)-(32AX), Formulas (29AY)(32AY), Formulas (29BX)-(32BX), and Formulas (29BY)-(32BY):
Formula (28A)
Formula (29B)
Formula (31A)
Formula (29A) Formula (29AX) Formula (29AY)
Formula (29BX)
Formula (29BY)
Formula (30AX)
Formula (30BY)
Formula (31AX)
Formula (31BY)
Formula (32A) Formula (32AX) Formula (32AY)
Formula (33A) Formula (34A) Formula (35A) . j-q sq^ic embodiments the 5’-stabilized end cap is attached to the 5’ end of the antisense strand. In some embodiments, the 5’-stabilized end cap is attached to the 5’ end of the antisense strand via one or more linkers independently selected from a phosphodiester linker, phosphorothioate linker, or phosphorodithioate linker. In some embodiments, the 5’-stabilized end cap is attached to the 5’ end of the sense strand. In some embodiments, the 5’-stabilized end cap is attached to the 5’ end of the sense strand via one or more linkers independently selected from a phosphodiester linker, phosphorothioate linker, or phosphorodithioate linker.
In some embodiments, any of the siNAs, sense strands, first nucléotide sequences, antisense strands, or second nucléotide sequences disclosed herein further comprise at least one thermally destabilizing nucléotides. In some embodiments, any of the antisense strands disclosed herein further comprise at least one thermally destabilizing nucléotide selected from:
. In some embodiments, any of the sense strands disclosed herein comprise at least one thermally destabilizing nucléotide selected from:
herein further comprise at least one thermally destabilizing nucléotide selected from:
herein further comprise at least one thermally destabilizing nucléotide selected from:
0' F . In some embodiments, any of the modified nucléotides disclosed herein is a thermally destabilizing nucléotide.
In some embodiments, any of the siNAs disclosed herein specifically downregulate or reduce expression of a target gene. In some embodiments, the target gene is a viral gene. In some embodiments, the viral gene is from a DNA virus. In some embodiments, the DNA virus is a double-stranded DNA (dsDNA) virus. In some embodiments, the dsDNA virus is a hepadnavirus. In some embodiments, the hepadnavirus is a hepatitis B virus (HBV). In some embodiments, the HBV is selected from HBV génotypes A-J. In some embodiments, the target gene is selected from the S gene or X gene of the HBV.
In some embodiments, the second nucléotide sequence is at least about 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% complementary to 15 to 30 nucléotides within positions 200-720 or 1100-1700 of SEQ ID NO: 410. In some embodiments, the second nucléotide sequence is at least about 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% complementary to 15 to 30 nucléotides within positions 200-280, 300-445, 460-510, 650-720, 1170-1220, 1250-1300, or 1550-1630 of SEQ ID NO: 410. In some embodiments, the second nucléotide sequence is at least about 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% complementary to 15 to 30 nucléotides within positions 200-230, 250-280, 300-330, 370-400, 405-445, 460-500, 670-700, 1180-1210, 1260-1295, 1520-1550, or 1570-1610 of SEQ ID NO: 410. In some embodiments, the second nucléotide sequence is at least about 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% complementary to 15 to 30 nucléotides starting at position 203, 206, 254, 305, 375, 409, 412, 415, 416, 419, 462, 466, 467, 674, 676, 1182, 1262, 1263, 1268, 1526, 1577, 1578, 1580, 1581, 1583, or 1584 of SEQ ID NO: 410.
In some embodiments, the first nucléotide sequence is at least about 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% identical to 15 to 30 nucléotides within positions 200-720 or 1100-1700 of SEQ ID NO: 410. In some embodiments, the first nucléotide sequence is at least about 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% identical to 15 to 30 nucléotides within positions 200-280, 300-445, 460-510, 650-720, 1170-1220, 1250-1300, or 1550-1630 of SEQ ID NO: 410. In some embodiments, the first nucléotide sequence is at least about 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% identical to 15 to 30 nucléotides within positions 200-230, 250-280, 300-330, 370-400, 405-445, 460-500, 670-700, 1180-1210, 12601295, 1520-1550, or 1570-1610 of SEQ ID NO: 410. In some embodiments, the first nucléotide sequence is at least about 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% identical to 15 to 30 nucléotides starting at position 203, 206, 254, 305, 375, 409, 412, 415, 416, 419, 462, 466, 467, 674, 676, 1182, 1262, 1263, 1268, 1526, 1577, 1578, 1580, 1581, 1583, or 1584 ofSEQ ID NO: 410.
In some embodiments, the first nucléotide sequence comprises a nucléotide sequence of any one SEQ ID NOs: 1-56, 103-158, and 205-260.
In some embodiments, the second nucléotide sequence comprises a nucléotide sequence of any one ofSEQ ID NOs: 57-102, 159-204, and 261-306.
In some embodiments, the sense strand comprises a nucléotide sequence of any one of SEQ ID NOs: 307-362 and 415-444.
In some embodiments, the antisense strand comprises a nucléotide sequence of any one ofSEQ ID NOs: 363-409, 445-533, and 536-539.
In some embodiments, at least one end of the siNA is a blunt end.
In some embodiments, at least one end of the siNA comprises an overhang, wherein the overhang comprises at least one nucléotide.
In some embodiments, both ends of the siNA comprise an overhang, wherein the overhang comprises at least one nucléotide.
In some embodiments, the siNA is selected from ds-siNA-001 to ds-siNA-0178.
In some embodiments, at least one 2’-fluoro nucléotide or 2’-O-methyl nucléotide is a 2’fluoro or 2-O-methyl nucléotide mimic of Formula (V):
q2 r5 , wherein .
R1 is independently a nucleobase, aryl, heteroaryl, or H, Q1 and Q2 are independently S or O,
R5 is independently -OCD3, -F, or -OCH3, and
R6 and R7 are independently H, D, or CD3.
In some embodiments, the 2’-fluoro or 2’-(9-methyl nucléotide mimic is a nucléotide mimic of Formula (16) - Formula (20):
O R2 S R2 d R2 0' ôcd3 d ÔCD3 % \ \ \ %
Formula (16) Formula (17) Formula (18) Formula (19) Formula (20) wherein R1 is a nucleobase and R2 is independently F or -OCH3.
Further disclosed herein are compositions comprising any of the siNAs disclosed herein. In some embodiments, the siNA targets an S gene of HBV. In some embodiments, the siNA specifically downregulates or reduces expression of the S gene of HBV. In some embodiments, the siNA targets an X gene of HBV. In some embodiments, the siNA specifically downregulates or reduces expression of the X gene of HBV. In some embodiments, the siNA comprises a first nucléotide sequence. In some embodiments, the first nucléotide sequence comprises a nucléotide sequence of any one SEQ ID NOs: 1-56, 103-158, and 205-260. In some embodiments, the siNA comprises a second nucléotide sequence. In some embodiments, the second nucléotide sequence comprises a nucléotide sequence of any one of SEQ ID NOs: 57-102, 159-204, and 261-306. In some embodiments, the siNA comprises a sense strand. In some embodiments, the sense strand comprises a nucléotide sequence of any one of SEQ ID NOs: 307-362 and 415-444. In some embodiments, the siNA comprises an antisense strand. In some embodiments, the antisense strand comprises a nucléotide sequence of any one of SEQ ID NOs: 363-409, 445-533, and 536539. In some embodiments, the siNA further comprises any of the 5’ end caps disclosed herein. In some embodiments, the siNA further comprises any of the phosphorylation blockers disclosed herein. In some embodiments, the siNA further comprises any of the conjugated moieties disclosed herein. In some embodiments, the siNA further comprises any of the destabilized nucléotides disclosed herein. In some embodiments, the siNA further comprises any of the modified nucléotides disclosed herein.
Further disclosed herein are compositions comprising 2, 3, 4, 5, 6, 7, 8, 9, 10 or more of any of the siNAs disclosed herein. In some embodiments, at least 1, 2, 3, 4, 5, or more siNAs target an S gene of HBV. In some embodiments, at least 1, 2, 3, 4, 5, or more siNAs specifically downregulate or reduce expression of the S gene of HBV. In some embodiments, at least 1, 2, 3, 4, 5, or more siNAs target an X gene of HBV. In some embodiments, at least 1, 2, 3, 4, 5, or more siNAs specifically downregulate or reduce expression of the X gene of HBV. In some embodiments, the siNA comprises a first nucléotide sequence. In some embodiments, the first nucléotide sequence comprises a nucléotide sequence of any one SEQ ID NOs: 1-56, 103-158, and 205-260. In some embodiments, the siNA comprises a second nucléotide sequence. In some embodiments, the second nucléotide sequence comprises a nucléotide sequence of any one of SEQ ID NOs: 57-102, 159-204, and 261-306. In some embodiments, the siNA comprises a sense strand. In some embodiments, the sense strand comprises a nucléotide sequence of any one of SEQ ID NOs: 307-362 and 415-444. In some embodiments, the siNA comprises an antisense strand. In some embodiments, the antisense strand comprises a nucléotide sequence of any one of SEQ ID NOs: 363-409, 445-533, and 536-539. In some embodiments, the siNA further comprises any of the 5 ’ end caps disclosed herein. In some embodiments, the siNA fürther comprises any of the phosphorylation blockers disclosed herein. In some embodiments, the siNA further comprises any of the conjugated moieties disclosed herein. In some embodiments, the siNA further comprises any of the destabilized nucléotides disclosed herein. In some embodiments, the siNA further comprises any of the modified nucléotides disclosed herein.
In some embodiments, any of the compositions disclosed herein further comprise an additional HBV treatment agent. In some embodiments, the additional HBV treatment agent is selected from a nucléotide analog, nucleoside analog, a capsid assembly modulator (CAM), a recombinant interferon, an entry inhibitor, a small molécule immunomodulator and oligonucleotide therapy. In some embodiments, the oligonucleotide therapy is an additional siNA. In some embodiments, the additional siNA is selected from any of ds-sîNA-001 to dssiNA-0178. In some embodiments, the oligonucleotide therapy is an antisense oligonucleotide (ASO), NAPs, or STOPs. In some embodiments, the ASO is ASO 1 or ASO 2. In some embodiments, the ASO specifically targets the S gene of HBV. In some embodiments, the ASO specifically targets the X gene of HBV. In some embodiments, the additional HBV treatment agent is selected from HBV STOPS™ ALG-010133, HBV CAM ALG-000184, ASO 1, recombinant interferon alpha 2b, IFN-a, PEG-IFN-a-2a, lamivudine, telbivudine, adefovir dipivoxil, clevudine, entecavir, tenofovir alafenamide, tenofovir disoproxil, NVR3-778, BAY414109, JNJ-632, JNJ-3989 (ARO-HBV), RG6004, GSK3228836, REP-2139, REP-2165, AB-729, VIR-2218, RG6346 (DCR-HBVS), JNJ-6379, GLS4, ABI-HO731, JNJ-440, NZ-4, RG7907, EDP-514, AB-423, AB-506, ABI-H03733 and ABI-H2158.
In some embodiments, any of the compositions disclosed herein further comprise a liver disease treatment agent. In some embodiments, the liver disease treatment agent is selected from a peroxisome proliferator-activator receptor (PPAR) agonist, famesoid X receptor (FXR) agonist, lipid-altering agent, and incretin-based therapy. In some embodiments, the PPAR agonist is selected from a PPARa agonist, dual PPARa/δ agonist, PPARy agonist, and dual PPARa/γ agonist. In some embodiments, the dual PPARa agonist is a fibrate. In some embodiments, the PPARa/δ agonist is elafibranor. In some embodiments, the PPARy agonist is a thiazolidinedione (TZD). In some embodiments, TZD is pioglitazone. In some embodiments, the dual PPARa/γ agonist is saroglitazar. In some embodiments, the FXR agonist is obeticholic acis (OCA). In some embodiments, the lipid-altering agent is aramchol. In some embodiments, the incretin-based therapy is a glucagon-like peptide 1 (GLP-1) receptor agonist or dipeptidyl peptidase 4 (DPP-4) inhibitor. In some embodiments, the GLP-1 receptor agonist is exenatide or liraglutide. In some embodiments, the DPP-4 inhibitor is sitagliptin or vildapliptin.
Further disclosed herein are methods of treating a disease in a subject in need thereof, comprising administering to the subject any of the siNAs disclosed herein. In some embodiments, the siNA comprises a first nucléotide sequence. In some embodiments, the first nucléotide sequence comprises a nucléotide sequence of any one SEQ ID NOs: 1-56, 103-158, and 205-260. In some embodiments, the siNA comprises a second nucléotide sequence. In some embodiments, the second nucléotide sequence comprises a nucléotide sequence of any one of SEQ ID NOs: 57-102, 159-204, and 261-306. In some embodiments, the siNA comprises a sense strand. In some embodiments, the sense strand comprises a nucléotide sequence of any one of SEQ ID NOs: 307-362 and 415-444. In some embodiments, the siNA comprises an antisense strand. In some embodiments, the antisense strand comprises a nucléotide sequence of any one of SEQ ID NOs: 363-409, 445-533, and 536-539. In some embodiments, the siNA further comprises any of the 5 ’ end caps disclosed herein. In some embodiments, the siNA further comprises any of the phosphorylation blockers disclosed herein. In some embodiments, the siNA further comprises any of the conjugated moieties disclosed herein. In some embodiments, the siNA further comprises any of the destabilized nucléotides disclosed herein. In some embodiments, the siNA further comprises any of the modified nucléotides disclosed herein.
Further disclosed herein are methods of treating a disease in a subject in need thereof, comprising administering to the subject any of the compositions disclosed herein. In some embodiments, the compositon comprises at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 or more of any of the siNAs disclosed herein. In some embodiments, the siNA comprises a first nucléotide sequence. In some embodiments, the first nucléotide sequence comprises a nucléotide sequence of any one SEQ ID NOs: 1-56, 103-158, and 205-260. In some embodiments, the siNA comprises a second nucléotide sequence. In some embodiments, the second nucléotide sequence comprises a nucléotide sequence of any one of SEQ ID NOs: 57-102, 159-204, and 261-306. In some embodiments, the siNA comprises a sense strand. In some embodiments, the sense strand comprises a nucléotide sequence of any one of SEQ ID NOs: 307-362 and 415-444. In some embodiments, the siNA comprises an antisense strand. In some embodiments, the antisense strand comprises a nucléotide sequence of any one of SEQ ID NOs: 363-409, 445-533, and 536539. In some embodiments, the siNA further comprises any of the 5’ end caps disclosed herein. In some embodiments, the siNA further comprises any of the phosphorylation blockers disclosed herein. In some embodiments, the siNA further comprises any of the conjugated moieties disclosed herein. In some embodiments, the siNA further comprises any of the destabilized nucléotides disclosed herein. In some embodiments, the siNA further comprises any of the modified nucléotides disclosed herein. In some embodiments, the composition further comprises any of the additional HBV treatment agents disclosed herein. In some embodiments, the disease is a viral disease. In some embodiments, the viral disease is caused by a DNA virus. In some embodiments, the DNA virus is a double stranded DNA (dsDNA) virus. In some embodiments, the dsDNA virus is a hepadnavirus. In some embodiments, the hepadnavirus is a hepatitis B virus (HBV). In some embodiments, the HBV is selected from HBV génotypes A-J. In some embodiments, the method further comprises administering an additional HBV treatment agent. In some embodiments, the siNA or the composition and the additional HBV treatment agent are administered concurrently. In some embodiments, the siNA or the composition and the additional HBV treatment agent are administered sequentially. In some embodiments, the siNA or the composition is administered prior to administering the additional HBV treatment agent. In some embodiments, the siNA or the composition is administered after administering the additional HBV treatment agent. In some embodiments, the additional HBV treatment agent is selected from a nucléotide analog, nucleoside analog, a capsid assembly modulator (CAM), a recombinant interferon, an entry inhibitor, a small molécule immunomodulator and oligonucleotide therapy. In some embodiments, the oligonucleotide therapy is an additional siNA. In some embodiments, the additional siNA is selected from any of ds-siNA-001 to dssiNA-0178. In some embodiments, the oligonucleotide therapy is an antisense oligonucleotide (ASO), NAPs, or STOPs. In some embodiments, the ASO is ASO 1 or ASO 2. In some embodiments, the additional HBV treatment agent is selected from HBV STOPS™ ALG010133, HBV CAM ALG-000184, ASO 1, recombinant interferon alpha 2b, IFN-a, PEG-IFN-a2a, lamivudine, telbivudine, adefovir dipivoxil, clevudine, entecavir, tenofovir alafenamide, tenofovir disoproxil, NVR3-778, BAY41-4109, JNJ-632, JNJ-3989 (ARO-HBV), RG6004, GSK3228836, REP-2139, REP-2165, AB-729, VIR-2218, RG6346 (DCR-HBVS), JNJ-6379, GLS4, ABI-HO731, JNJ-440, NZ-4, RG7907, EDP-514, AB-423, AB-506, ABI-H03733 and ABI-H2158.
In some embodiments, the disease is a liver disease. In some embodiments, the liver disease is a nonalcoholic fatty liver disease (NAFLD) or hepatocellular carcinoma (HCC). In some embodiments, the NAFLD is nonalcoholic steatohepatitis (NASH). In some embodiments, the method further comprises administering to the subject a liver disease treatment agent. In some embodiments, the liver disease treatment agent is selected from a peroxisome proliferatoractivator receptor (PPAR) agonist, famesoid X receptor (FXR) agonist, lipid-altering agent, and incretin-based therapy. In some embodiments, the PPAR agonist is selected from a PPARa agonist, dual PPARa/δ agonist, PPARy agonist, and dual PPARa/γ agonist. In some embodiments, the dual PPARa agonist is a fibrate. In some embodiments, the PPARa/δ agonist is elafibranor. In some embodiments, the PPARy agonist is a thiazolidinedione (TZD). In some embodiments, TZD is pioglitazone. In some embodiments, the dual PPARa/γ agonist is saroglitazar. In some embodiments, the FXR agonist is obeticholic acis (OCA). In some embodiments, the lipid-altering agent is aramchol. In some embodiments, the incretin-based therapy is a glucagon-like peptide 1 (GLP-1) receptor agonist or dipeptidyl peptidase 4 (DPP-4) inhibitor. In some embodiments, the GLP-1 receptor agonist is exenatide or liraglutide. In some embodiments, the DPP-4 inhibitor is sitagliptin or vildapliptin. In some embodiments, the siNA or composition and the liver disease treatment agent are administered concurrently. In some embodiments, the siNA or composition and the liver disease treatment agent are administered sequentially. In some embodiments, the siNA or composition is administered prior to administering the liver disease treatment agent. In some embodiments, the siNA or composition is administered after administering the liver disease treatment agent.
In some embodiments, the siNA or the composition is administered at a dose of at least 1 mg/kg, 2 mg/kg, 3 mg/kg, 4 mg/kg, 5 mg/kg, 6 mg/kg, 7 mg/kg, 8 mg/kg, 9 mg/kg, 10 mg/kg, 11 mg/kg, 12 mg/kg, 13 mg/kg 14 mg/kg, or 15 mg/kg. In some embodiments, the siNA or the composition is administered at a dose of between 0.5 mg/kg to 50 mg/kg, 0.5 mg/kg to 40 mg/kg 0.5 mg/kg to 30 mg/kg, 1 mg/kg to 50 mg/kg, 1 mg/kg to 40 mg/kg, 1 mg/kg to 30 mg/kg, 1 mg/kg to 20 mg/kg, 3 mg/kg to 50 mg/kg, 3 mg/kg to 40 mg/kg, 3 mg/kg to 30 mg/kg, 3 mg/kg to 20 mg/kg, 3 mg/kg to 15 mg/kg, 3 mg/kg to 10 mg/kg, 4 mg/kg to 50 mg/kg, 4 mg/kg to 40 mg/kg, 4 mg/kg to 30 mg/kg, 4 mg/kg to 20 mg/kg, 4 mg/kg to 15 mg/kg, 4 mg/kg to 10 mg/kg, 5 mg/kg to 50 mg/kg, 5 mg/kg to 40 mg/kg, 5 mg/kg to 30 mg/kg, 5 mg/kg to 20 mg/kg, 5 mg/kg to 15 mg/kg, or 5 mg/kg to 10 mg/kg.
In some embodiments, the siNA or the composition is administered at least 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 times. In some embodiments, the siNA or the composition is administered at least 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 times a day, at least 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 times a week, or at least 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 times a month. In some embodiments, the siNA or the composition are administered at least once every 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, or 21 days. In some embodiments, the siNA or the composition is administered for aperiod of at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, or 21 days, or at least 1,2, 3,4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19,20, 21,22, 23, 24, 25, 26, 27, 28, 29, 30, 35, 40, 45, 50, 51, 52, 53, 54, or 55 weeks.
In some embodiments, the siNA or the composition is administered at a single dose of 5 mg/kg. In some embodiments, the siNA or the composition is administered at a single dose of 10 mg/kg. In some embodiments, the siNA or the composition is administered at three doses of 10 mg/kg once a week. In some embodiments, the siNA or the composition is administered at three doses of 10 mg/kg once every three days. In some embodiments, the siNA or the composition is administered at five doses of 10 mg/kg once every three days. In some embodiments, the siNA or the composition is administered at six doses of ranging from 1 mg/kg to 15 mg/kg, 1 mg/kg to 10 mg/kg, 2 mg/kg to 15 mg/kg, 2 mg/kg to 10 mg/kg, 3 mg/kg to 15 mg/kg, or 3 mg/kg to 10 mg/kg. In some embodiments, the first dose and second dose are administered at least 3 days apart. In some embodiments, the second dose and third dose are administered at least 4 days apart. In some embodiments, the third dose and fourth dose, fourth dose and fifth dose, or fifth dose and sixth dose are administered at least 7 days apart.
In some embodiments, any of the siNAs or the compositions disclosed herein are formulated as a particle or viral vector. In some embodiments, the siNA or the composition are administered in a particle or viral vector. In some embodiments, the viral vector is a vector of adenovirus, adeno-associated virus (AAV), alphavirus, flavivirus, herpes simplex virus, lentivirus, measles virus, picomavirus, poxvirus, retrovirus, or rhabdovirus. In some embodiments, the viral vector is a recombinant viral vector. In some embodiments, the viral vector is selected from AAVrh.74, AAVrh.10, AAVrh.20, AAV-1, AAV-2, AAV-3, AAV-4, AAV-5, AAV-6, AAV-7, AAV-8, AAV-9, AAV-10, AAV-11, AAV-12 and AAV-13. In some embodiments, the siNA or the composition is administered systemically. In some embodiments, the siNA or the composition is administered locally. In some embodiments, the siNA or the composition is administered intravenously, subcutaneously, or intramuscularly.
In some embodiments, any of the siRNAs or compositions disclosed herein are used in the manufacture of a médicament for treating a disease. In some embodiments, the disease is a viral disease. In some embodiments, the viral disease is caused by a DNA virus. In some embodiments, the DNA virus is a double stranded DNA (dsDNA virus). In some embodiments, the dsDNA virus is a hepadnavirus. In some embodiments, the hepadnavirus is a hepatitis B virus (HBV). In some embodiments, the HBV is selected from HBV génotypes A-J. In some embodiments, an additional HBV treatment agent is fùrther used in the manufacture of the médicament. In some embodiments, the additional HBV treatment agent is selected from a nucléotide analog, nucleoside analog, a capsid assembly modulator (CAM), a recombinant interferon, an entry inhibitor, a small molécule immunomodulator and oligonucleotide therapy. In some embodiments, the oligonucleotide therapy is an additional siNA. In some embodiments, the additional siNA is selected from any of ds-siNA-001 to ds-siNA-0178. In some embodiments, the oligonucleotide therapy is an antisense oligonucleotide (ASO), NAPs, or STOPs. In some embodiments, the ASO is ASO 1 or ASO 2. In some embodiments, the additional HBV treatment agent is selected from HBV STOPS™ ALG-010133, HBV CAM ALG-000184, ASO 1, recombinant interferon alpha 2b, IFN-a, PEG-IFN-a-2a, lamivudine, telbivudine, adefovir dipivoxil, clevudine, entecavir, tenofovir alafenamide, tenofovir disoproxil, NVR3-778, BAY41-4109, JNJ-632, JNJ-3989 (ARO-HBV), RG6004, GSK3228836, REP-2139, REP-2165, AB-729, VIR-2218, RG6346 (DCR-HBVS), JNJ-6379, GLS4, ABI-HO731, JNJ440, NZ-4, RG7907, EDP-514, AB-423, AB-506, ABI-H03733 and ABI-H2158.
In some embodiments, any of the siRNAs or compositions disclosed herein are used in the manufacture of a médicament for treating a disease. In some embodiments, the disease is a liver disease. In some embodiments, the liver disease is a nonalcoholic fatty liver disease (NAFLD) or hepatocellular carcinoma (HCC). In some embodiments, the NAFLD is nonalcoholic steatohepatitis (NASH). In some embodiments, the siNA comprises a first nucléotide sequence. In some embodiments, the first nucléotide sequence comprises a nucléotide sequence of any one SEQ ID NOs: 1-56, 103-158, and 205-260. In some embodiments, the siNA comprises a second nucléotide sequence. In some embodiments, the second nucléotide sequence comprises a nucléotide sequence of any one of SEQ ID NOs: 57-102, 159-204, and 261-306. In some embodiments, the siNA comprises a sense strand. In some embodiments, the sense strand comprises a nucléotide sequence of any one of SEQ ID NOs: 307-362 and 415-444. In some embodiments, the siNA comprises an antisense strand. In some embodiments, the antisense strand comprises a nucléotide sequence of any one of SEQ ID NOs: 363-409, 445-533, and 536539. In some embodiments, the siNA further comprises any of the 5’ end caps disclosed herein. In some embodiments, the siNA further comprises any of the phosphorylation blockers disclosed herein. In some embodiments, the siNA further comprises any of the conjugated moieties disclosed herein. In some embodiments, the siNA further comprises any of the destabilized nucléotides disclosed herein. In some embodiments, the siNA further comprises any of the modified nucléotides disclosed herein. In some embodiments, a liver disease treatment agent is further used in the manufacture of the médicament. In some embodiments, the liver disease treatment agent is selected from a peroxisome proliferator-activator receptor (PPAR) agonist, famesoid X receptor (FXR) agonist, lipid-altering agent, and incretin-based therapy. In some embodiments, the PPAR agonist is selected from a PPARa agonist, dual PPARa/δ agonist, PPARy agonist, and dual PPARa/γ agonist. In some embodiments, the dual PPARa agonist is a fibrate. In some embodiments, the PPARa/δ agonist is elafibranor. In some embodiments, the PPARy agonist is a thiazolidinedione (TZD). In some embodiments, TZD is pioglitazone. In some embodiments, the dual PPARa/γ agonist is saroglitazar. In some embodiments, the FXR agonist is obeticholic acis (OCA). In some embodiments, the lipid-altering agent is aramchol. In some embodiments, the incretin-based therapy is a glucagon-like peptide 1 (GLP-1) receptor agonist or dipeptidyl peptidase 4 (DPP-4) inhibitor. In some embodiments, the GLP-1 receptor agonist is exenatide or liraglutide. In some embodiments, the DPP-4 inhibitor is sitagliptin or vildapliptin.
In some embodiments, any of the siNAs disclosed herein is used as a médicament. In some embodiments, the siNA comprises a first nucléotide sequence. In some embodiments, the first nucléotide sequence comprises a nucléotide sequence of any one SEQ ID NOs: 1-56, 103158, and 205-260. In some embodiments, the siNA comprises a second nucléotide sequence. In some embodiments, the second nucléotide sequence comprises a nucléotide sequence of any one of SEQ ID NOs: 57-102, 159-204, and 261-306. In some embodiments, the siNA comprises a sense strand. In some embodiments, the sense strand comprises a nucléotide sequence of any one of SEQ ID NOs: 307-362 and 415-444. In some embodiments, the siNA comprises an antisense strand. In some embodiments, the antisense strand comprises a nucléotide sequence of any one of SEQ ID NOs: 363-409, 445-533, and 536-539. In some embodiments, the siNA further comprises any of the 5’ end caps disclosed herein. In some embodiments, the siNA further comprises any of the phosphorylation blockers disclosed herein. In some embodiments, the siNA fürther comprises any of the conjugated moieties disclosed herein. In some embodiments, the siNA further comprises any of the destabilized nucléotides disclosed herein. In some embodiments, the siNA further comprises any of the modified nucléotides disclosed herein.
In some embodiments, any of the compositions disclosed herein are used as a médicament. In some embodiments, the composition comprises any of the siNAs disclosed herein. In some embodiments, the siNA comprises a first nucléotide sequence. In some embodiments, the first nucléotide sequence comprises a nucléotide sequence of any one SEQ ID NOs: 1-56, 103-158, and 205-260. In some embodiments, the siNA comprises a second nucléotide sequence. In some embodiments, the second nucléotide sequence comprises a nucléotide sequence of any one of SEQ ID NOs: 57-102, 159-204, and 261-306. In some embodiments, the siNA comprises a sense strand. In some embodiments, the sense strand comprises a nucléotide sequence of any one of SEQ ID NOs: 307-362 and 415-444. In some embodiments, the siNA comprises an antisense strand. In some embodiments, the antisense strand comprises a nucléotide sequence of any one of SEQ ID NOs: 363-409, 445-533, and 536539. In some embodiments, the siNA further comprises any of the 5’ end caps disclosed herein. In some embodiments, the siNA further comprises any of the phosphorylation blockers disclosed herein. In some embodiments, the siNA further comprises any of the conjugated moieties disclosed herein. In some embodiments, the siNA further comprises any of the destabilized nucléotides disclosed herein. In some embodiments, the siNA further comprises any of the modified nucléotides disclosed herein.
In some embodiments, any of the siNAs disclosed herein are used in the treatment of a disease. In some embodiments, the siNA comprises a first nucléotide sequence. In some embodiments, the first nucléotide sequence comprises a nucléotide sequence of any one SEQ ID NOs: 1-56, 103-158, and 205-260. In some embodiments, the siNA comprises a second nucléotide sequence. In some embodiments, the second nucléotide sequence comprises a nucléotide sequence of any one of SEQ ID NOs: 57-102, 159-204, and 261-306. In some embodiments, the siNA comprises a sense strand. In some embodiments, the sense strand comprises a nucléotide sequence of any one of SEQ ID NOs: 307-362 and 415-444. In some embodiments, the siNA comprises an antisense strand. In some embodiments, the antisense strand comprises a nucléotide sequence of any one of SEQ ID NOs: 363-409, 445-533, and 536539. In some embodiments, the siNA further comprises any of the 5’ end caps disclosed herein. In some embodiments, the siNA further comprises any of the phosphorylation blockers disclosed herein. In some embodiments, the siNA fùrther comprises any of the conjugated moieties disclosed herein. In some embodiments, the siNA further comprises any of the destabilized nucléotides disclosed herein. In some embodiments, the siNA further comprises any of the modified nucléotides disclosed herein. In some embodiments, the disease is a viral disease. In some embodiments, the viral disease is caused by a DNA virus. In some embodiments, the DNA virus is a double stranded DNA (dsDNA virus). In some embodiments, the dsDNA virus is a hepadnavirus. In some embodiments, the hepadnavirus is a hepatitis B virus (HBV). In some embodiments, the HBV is selected from HBV génotypes A-J. In some embodiments, the disease is a liver disease. In some embodiments, the liver disease is a nonalcoholic fatty liver disease (NAFLD) or hepatocellular carcinoma (HCC). In some embodiments, the NAFLD is nonalcoholic steatohepatitis (NASH).
In some embodiments, any of the compositions disclosed herein are used in the treatment of a disease. In some embodiments, the composition comprises any of the siNAs disclosed herein. In some embodiments, the siNA comprises a first nucléotide sequence. In some embodiments, the first nucléotide sequence comprises a nucléotide sequence of any one SEQ ID NOs: 1-56, 103-158, and 205-260. In some embodiments, the siNA comprises a second nucléotide sequence. In some embodiments, the second nucléotide sequence comprises a nucléotide sequence of any one of SEQ ID NOs: 57-102, 159-204, and 261-306. In some embodiments, the siNA comprises a sense strand. In some embodiments, the sense strand comprises a nucléotide sequence of any one of SEQ ID NOs: 307-362 and 415-444. In some embodiments, the siNA comprises an antisense strand. In some embodiments, the antisense strand comprises a nucléotide sequence of any one of SEQ ID NOs: 363-409, 445-533, and 536539. In some embodiments, the siNA further comprises any of the 5’ end caps disclosed herein. In some embodiments, the siNA further comprises any of the phosphorylation blockers disclosed herein. In some embodiments, the siNA further comprises any of the conjugated moieties disclosed herein. In some embodiments, the siNA further comprises any of the destabilized nucléotides disclosed herein. In some embodiments, the siNA further comprises any of the modified nucléotides disclosed herein. In some embodiments, the disease is a viral disease. In some embodiments, the viral disease is caused by a DNA virus. In some embodiments, the DNA virus is a double stranded DNA (dsDNA virus). In some embodiments, the dsDNA virus is a hepadnavirus. In some embodiments, the hepadnavirus is a hepatitis B virus (HBV). In some embodiments, the HBV is selected from HBV génotypes A-J. In some embodiments, the disease is a liver disease. In some embodiments, the liver disease is a nonalcoholic fatty liver disease (NAFLD) or hepatocellular carcinoma (HCC). In some embodiments, the NAFLD is nonalcoholic steatohepatitis (NASH).
BRIEF DESCRIPTION OF THE DRAWINGS
FIG. 1 illustrâtes an exemplary siNA molécule.
FIG. 2 illustrâtes an exemplary siNA molécule.
FIGs. 3A-3G illustrate exemplary double-stranded siNA molécules.
FIG. 4 shows a graph of the change in sérum HBsAg from AAV-HBV mice treated with ds-siNA-0160, ds-siNA-0165, ds-siNA-0163, or ds-siNA-0166.
FIG. 5A shows a graph of the change in sérum HBsAg from AAV-HBV mice treated with vehicle (G01) or ds-siNA-0160 (G03).
FIG. 5B shows a graph of the change in sérum HBsAg from AAV-HBV mice treated with vehicle (G01) or ds-siNA-0160 (G 15).
FIG. 5C shows a graph of the change in sérum HBsAg from AAV-HBV mice treated with vehicle (G01) or ds-siNA-0160 (G03).
FIG. 5D shows a graph of the change in sérum HBsAg from AAV-HBV mice treated with vehicle (G01), ds-siNA-0160 (G03), or ds-siNA-0109 (G09).
FIGs. 5E-5F show a graph of the change in sérum HBsAg from AAV-HBV mice treated with vehicle (G01) or ds-siNA-0169 (G 18).
FIG. 5G shows a graph of the change in sérum HBsAg from AAV-HBV mice treated with vehicle (G01) or ds-siNA-0169 (G04).
FIG. 5H shows a graph of the change in sérum HBsAg from AAV-HBV mice treated with vehicle (G01) or ds-siNA-0169 (G04).
FIG. 51 shows a graph of the change in sérum HBsAg from AAV-HBV mice treated with vehicle (G01), ds-siNA-0169 (G04), or ds-siNA-0147 (G08).
FIG. 5J shows a graph of the change in sérum HBsAg from AAV-HBV mice treated with vehicle (G01), ds-siNA-0166 (G06), or ds-siNA-0153 (G14).
FIG. 5K shows a graph of the change in sérum HBsAg from AAV-HBV mice treated with vehicle (G01), ds-siNA-0163 (G05), or ds-siNA-0119 (G13).
FIG. 6A shows a graph of the change in sérum HBsAg from AAV-HBV mice treated with vehicle (G01), ds-siNA-0160 (G15), or ds-siNA-080 (G14).
FIG. 6B shows a graph of the change in sérum HBsAg from AAV-HBV mice treated with vehicle (G01), ds-siNA-0169 (G16), or ds-siNA-081 (G13).
FIG. 7A shows a graph of the change in sérum HBsAg from AAV-HBV mice treated with vehicle (G01), ds-siNA-0165 (G18), or ds-siNA-0127 (G17).
FIG. 7B shows a graph of the change in sérum HBsAg from AAV-HBV mice treated with vehicle (G01), ds-siNA-0168 (G20), or ds-siNA-0150 (G19).
FIG. 8A shows a graph of the change in sérum HBsAg from AAV-HBV mice treated with vehicle (G01), ds-siNA-0160 (G06), ASO 1 (G18), or a combination of ds-siNA-0160 and ASO 1 (G20).
FIG. 8B shows a graph of the change in sérum HBsAg from AAV-HBV mice treated with vehicle (G01), ds-siNA-0160 (G06), ASO 1 (G18), or a combination of ds-siNA-0160 and ASO 1 (G20).
FIG. 8C shows a graph of a synergy analysis of a combination therapy with unconjugated forms of ds-siNA-0164 and ASO 2 (e.g., ds-siNA-0160 and ASO 1 without GalNac).
FIG. 9 shows a graph of the change in sérum HBsAg from AAV-HBV mice treated with vehicle (G01), ds-siNA-0166 (G03), ds-siNA-0155 (G08), or ds-siNA-0157.
FIG. 10 shows a graph of the change in sérum HBsAg from AAV-HBV mice treated with vehicle (G01), ds-siNA-0165 (G10), ds-siNA-0160 (G06), or a combination therapy with ds-siNA-0160 and ds-siNA-0165 (G14).
FIG. 11 shows a graph of the change in sérum HBsAg from AAV-HBV mice treated with vehicle (G01), ds-siNA-0165 (G05), or ds-siNA-0144 (G11).
FIG. 12 shows a graph of the change in sérum HBsAg from AAV-HBV mice treated with vehicle (G01), ds-siNA-0163 (G04), ds-siNA-0122 (G09), or ds-siNA-0123 (G13).
FIG. 13 shows a graph of the change in sérum HBsAg from AAV-HBV mice treated with vehicle (G 15) or ds-siNA-0147 (G 19).
FIG. 14 shows a graph of the change in sérum HBsAg from AAV-HBV mice treated with vehicle (G 15, square), ds-siNA-0109 (G 21, circle), or ds-siNA-0172 (G 27, diamond).
FIG. 15 shows a graph of the change in sérum HBsAg from AAV-HBV mice treated with vehicle (G 01, circle), ds-siNA-0109 (G 07, square), ds-siNA-0119 (G 11, triangle), or dssiNA-0153 (G 13, diamond).
FIG. 16 shows a graph of the change in sérum HBsAg from AAV-HBV mice treated with vehicle (G 01, circle), ASO 1 (G 20, square), ds-siNA-0147 (G 24, diamond), or a combination of ASO 1 and ds-siNA-0147 (G 25, triangle).
FIG. 17 shows a graph of the change in sérum HBsAg from AAV-HBV mice treated with vehicle (G 01, circle), ASO 1 (G 20, square), ds-siNA-0109 (G 26, diamond), or a combination of ASO 1 and ds-siNA-0109 (G 27, triangle).
DETAILED DESCRIPTION OF THE INVENTION
Disclosed herein are short interfering nucleic acid (siNA) molécules comprising modified nucléotides. The siNA molécules described herein may be double-stranded siNA (ds-siNA) molécules. The siNA molécules described herein may comprise modified nucléotides selected from 2’-(9-methyl nucléotides and 2’-fluoro nucléotides. The siNA molécules described herein may comprise 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 or more phosphorothioate intemucleoside linkages. The siNA molécules described herein may comprise a phosphorylation blocker. The siNA molécules described herein may comprise a 5’-stabilized end cap. The siNA molécules described herein may comprise a galactosamine. The siNA molécules described herein may comprise one or more blunt ends. The siNA molécules described herein may comprise one or more overhangs.
Further disclosed herein are short interfering nucleic acid (siNA) molécules comprising (a) a phosphorylation blocker; and (b) a short interfering nucleic acid (siNA). The siNA may comprise at least 5 nucléotides. The nucléotides may be modified nucléotides, non-modified nucléotides, or any combination thereof. The nucléotides may be ribonucleotides, deoxyribonucleotides, or any combination thereof. The siNA may be single-stranded. Altematively, the siNA is double-stranded. The double-stranded siNA may comprise one or more blunt ends. The double-stranded siNA may comprise one or more overhangs. The doublestranded siNA may comprise a blunt end and an overhang.
Further disclosed herein are short interfering nucleic acid (siNA) molécules comprising (a) a conjugated moiety; and (b) a short interfering nucleic acid (siNA). The siNA may comprise at least 5 nucléotides. The nucléotides may be modified nucléotides, non-modified nucléotides, or any combination thereof. The nucléotides may be ribonucleotides, deoxyribonucleotides, or any combination thereof. The siNA may be single-stranded. Altematively, the siNA is doublestranded. The double-stranded siNA may comprise one or more blunt ends. The double-stranded siNA may comprise one or more overhangs. The double-stranded siNA may comprise a blunt end and an overhang.
Further disclosed herein are short interfering nucleic acid (siNA) molécules comprising (a) a 5’-stabilized end cap; and (b) a short interfering nucleic acid (siNA). The siNA may comprise at least 5 nucléotides. The nucléotides may be modified nucléotides, non-modified nucléotides, or any combination thereof. The nucléotides may be ribonucleotides, deoxyribonucleotides, or any combination thereof. The siNA may be single-stranded. Altematively, the siNA is double-stranded. The double-stranded siNA may comprise one or more blunt ends. The double-stranded siNA may comprise one or more overhangs. The doublestranded siNA may comprise a blunt end and an overhang.
Further disclosed herein are short interfering nucleic acid (siNA) molécules comprising (a) at least one phosphorylation blocker, conjugated moiety, or 5’-stabilized end cap; and (b) a short interfering nucleic acid (siNA). The siNA may comprise at least 5 nucléotides. The nucléotides may be modified nucléotides, non-modified nucléotides, or any combination thereof. The nucléotides may be ribonucleotides, deoxyribonucleotides, or any combination thereof. The siNA may be single-stranded. Altematively, the siNA is double-stranded. The double-stranded siNA may comprise one or more blunt ends. The double-stranded siNA may comprise one or more overhangs. The double-stranded siNA may comprise a blunt end and an overhang.
An exemplary siNA molécule of the présent disclosure is shown in FIG. 1. As shown in FIG. 1, an exemplary siNA molécule comprises a sense strand (101) and an antisense strand (102). The sense strand (101) may comprise a first oligonucleotide sequence (103). The first oligonucleotide sequence (103) may comprise one or more phosphorothioate intemucleoside linkages (109). The phosphorothioate intemucleoside linkage (109) may be between the nucléotides at the 5’ or 3’ terminal end of the first oligonucleotide sequence (103). The phosphorothioate intemucleoside linkage (109) may be between the first three nucléotides from the 5’ end of the first oligonucleotide sequence (103). The first oligonucleotide sequence (103) may comprise one or more 2’-fluoro nucléotides (110). The first oligonucleotide sequence (103) may comprise one or more 2’-(9-methyl nucléotides (111). The first oligonucleotide sequence (103) may comprise 15 or more modified nucléotides independently selected from 2’-fluoro nucléotides (110) and 2’-<9-methyl nucléotides (111). The sense strand (101) may further comprise a phosphorylation blocker (105). The sense strand (101) may further comprise a galactosamine (106). The antisense strand (102) may comprise a second oligonucleotide sequence (104). The second oligonucleotide sequence (104) may comprise one or more phophorothioate intemucleoside linkages (109). The phosphorothioate intemucleoside linkage (109) may be between the nucléotides at the 5’ or 3’ terminal end of the second oligonucleotide sequence (104). The phosphorothioate intemucleoside linkage (109) may be between the first three nucléotides from the 5’ end of the second oligonucleotide sequence (104). The phosphorothioate intemucleoside linkage (109) may be between the first three nucléotides from the 3’ end of the second oligonucleotide sequence (104). The second oligonucleotide sequence (104) may comprise one or more 2’-fluoro nucléotides (110). The second oligonucleotide sequence (104) may comprise one or more 2’-O-methyl nucléotides (111). The second oligonucleotide sequence (104) may comprise 15 or more modified nucléotides independently selected from 2’-fluoro nucléotides (110) and 2’-O-methyl nucléotides (111). The antisense strand (102) may further comprise a 5’-stabilized end cap (107). The siNA may further comprise one or more blunt ends. Altematively, or additionally, one end of the siNA may comprise an overhang (108). The overhang (108) may be part of the sense strand (101). The overhang (108) may be part of the antisense strand (102). The overhang (108) may be distinct from the first nucléotide sequence (103). The overhang (108) may be distinct from the second nucléotide sequence (104). The overhang (108) may be part of the first nucléotide sequence (103). The overhang (108) may be part of the second nucléotide sequence (104). The overhang (108) may comprise 1 or more nucléotides. The overhang (108) may comprise 1 or more deoxyribonucleotides. The overhang (108) may comprise 1 or more modified nucléotides. The overhang (108) may comprise 1 or more modified ribonucleotides. The sense strand (101) may be shorter than the antisense strand (102). The sense strand (101) may be the same length as the antisense strand (102). The sense strand (101) may be longer than the antisense strand (102).
An exemplary siNA molécule of the présent disclosure is shown in FIG. 2. As shown in FIG. 2, an exemplary siNA molécule comprises a sense strand (201) and an antisense strand (202). The sense strand (201) may comprise a first oligonucleotide'sequence (203). The first oligonucleotide sequence (203) may comprise one or more phophorothioate intemucleoside linkages (209). The phosphorothioate intemucleoside linkage (209) may be between the nucléotides at the 5’ or 3’ terminal end of the first oligonucleotide sequence (203). The phosphorothioate intemucleoside linkage (209) may be between the first three nucléotides from the 5’ end of the first oligonucleotide sequence (203). The first oligonucleotide sequence (203) may comprise one or more 2’-fluoro nucléotides (210). The first oligonucleotide sequence (203) may comprise one or more 2’-O-methyl nucléotides (211). The first oligonucleotide sequence (203) may comprise 15 or more modified nucléotides independently selected from 2’-fluoro nucléotides (210) and 2’-(9-methyl nucléotides (211). The sense strand (201) may further comprise a phosphorylation blocker (205). The sense strand (201) may further comprise a galactosamine (206). The antisense strand (202) may comprise a second oligonucleotide sequence (204). The second oligonucleotide sequence (204) may comprise one or more phophorothioate intemucleoside linkages (209). The phosphorothioate intemucleoside linkage (209) may be between the nucléotides at the 5’ or 3’ terminal end of the second oligonucleotide sequence (204). The phosphorothioate intemucleoside linkage (209) may be between the first three nucléotides from the 5’ end of the second oligonucleotide sequence (204). The phosphorothioate intemucleoside linkage (209) may be between the first three nucléotides from the 3’ end of the second oligonucleotide sequence (204). The second oligonucleotide sequence (204) may comprise one or more 2’-fluoro nucléotides (210). The second oligonucleotide sequence (204) may comprise one or more 2’-O-methyl nucléotides (211). The second oligonucleotide sequence (204) may comprise 15 or more modified nucléotides independently selected from 2’-fluoro nucléotides (210) and 2’-O-methyl nucléotides (211). The antisense strand (202) may further comprise a 5’-stabilized end cap (207). The siNA may further comprise one or more overhangs (208). The overhang (208) may be part of the sense strand (201). The overhang (208) may be part of the antisense strand. (202). The overhang (208) may be distinct from the first nucléotide sequence (203). The overhang (208) may be distinct from the second nucléotide sequence (204). The overhang (208) may be part of the first nucléotide sequence (203). The overhang (208) may be part of the second nucléotide sequence (204). The overhang (208) may be adjacent to the 3’ end of the first nucléotide sequence (203). The overhang (208) may be adjacent to the 5’ end of the first nucléotide sequence (203). The overhang (208) may be adjacent to the 3’ end of the second nucléotide sequence (204). The overhang (208) may be adjacent to the 5’ end of the second nucléotide sequence (204). The overhang (208) may comprise 1 or more nucléotides. The overhang (208) may comprise 1 or more deoxyribonucleotides. The overhang (208) may comprise a TT sequence. The overhang (208) may comprise 1 or more modified nucléotides. The overhang (208) may comprise 1 or more modified nucléotides disclosed herein (e.g., 2-fluoro nucléotide, 2’-O-methyl nucléotide, 2’ fluoro nucléotide mimic, 2’-(9-methyl nucléotide mimic, or a nucléotide comprising a modified nucleobase). The overhang (208) may comprise 1 or more modified ribonucleotides. The sense strand (201) may be shorter than the antisense strand (202). The sense strand (201) may be the same length as the antisense strand (202). The sense strand (201) may be longer than the antisense strand (202).
FIGs. 3A-3G depict exemplary ds-siNA modification patterns. As shown in FIGs. 3A3 G, an exemplary ds-siNA molécule may hâve the following formula: S’-An’Bn^BnUn^An^An9^’
3’-Cq 1Aq2Bq 3Aq4Bq5Aq 6Bq7Aq8Bq 9AqI0Bq11Aq12-5’ wherein:
the top strand is a sense strand comprising a first nucléotide sequence that is at least about 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% identical to an RNA corresponding to a target gene, wherein the first nucléotide sequence comprises 15 to 30 nucléotides;
the bottom strand is an antisense strand comprising a second nucléotide sequence that is at least about 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% complementary to the RNA corresponding to the target gene, wherein the second nucléotide sequence comprises 15 to 30 nucléotides;
each A is independently a 2’-(9-methyl nucléotide or a nucléotide comprising a 5’ stabilized end cap or phosphorylation blocker;
B is a 2’-fluoro nucléotide;
C represents overhanging nucléotides and is a 2’-O-methyl nucléotide;
n1 = 1-4 nucléotides in length;
each n2, n6, n8, q3, q5, q7, q9, q11, and q12 is independently 0-1 nucléotides in length;
each n3 and n4 is independently 1-3 nucléotides in length;
n5 is 1-10 nucléotides in length;
n7 is 0-4 nucléotides in length;
each n9, q1, and q2 is independently 0-2 nucléotides in length;
q4 is 0-3 nucléotides in length;
q6 is 0-5 nucléotides in length;
q8 is 2-7 nucléotides in length; and q10 is 2-11 nucléotides in length.
The ds-siNA may further comprise a conjugated moiety. The conjugated moiety may comprise any of the galactosamines disclosed herein. The ds-siNA may further comprise (i) phosphorothioate intemucleoside linkages between the nucléotides at positions 1 and 2 and positions 2 and 3 from the 5’ end of the sense strand; and (ii) phosphorothioate intemucleoside linkages between the nucléotides at positions 1 and 2; positions 2 and 3; positions 19 and 20; and positions 20 and 21 from the 5’ end of the antisense strand. The ds-siNA may further comprise a 5’-stabilizing end cap. The 5’-stabilizing end cap may be a vinyl phosphonate. The 5’-stabilizing end cap may be attached to the 5’ end of the antisense strand. In some embodiments, the 2’-Omethyl nucléotide at position 1 from the 5’ end of the sense strand is further modifïed to contain a 5’ stabilizing end cap. In some embodiments, the 2’-O-methyl nucléotide at position 1 from the 5’ end of the antisense strand is further modifïed to contain a 5’ stabilizing end cap. In some embodiments, the 2’-O-methyl nucléotide at position 1 from the 5’ end of the sense strand is further modifïed to contain a phosphorylation blocker. In some embodiments, the 2’-(9-methyl nucléotide at position 1 from the 3 ’ end of the sense strand is further modifïed to contain a phosphorylation blocker. In some embodiments, the 2’-(9-methyl nucléotide at position 1 from the 5’ end of the antisense strand is further modifïed to contain a phosphorylation blocker. In some embodiments, the 2’-O-methyl nucléotide at position 1 from the 3’ end of the antisense strand is further modifïed to contain a phosphorylation blocker. An exemplary ds-siNA molécule may hâve the following formula:
5’-A2-4 BiAi-3 B2-3 A2-io B0-1A0-4B0-1 Ao-2-3’ ’ -C2 Aq-2Bo- 1 Ao-3Bq- 1A0-5B0-1A2-7 B1A2.11 B1A1 -5 ’ wherein:
the top strand is a sense strand comprising a first nucléotide sequence that is at least about 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% identical to an RNA corresponding to a target gene, wherein the first nucléotide sequence comprises 15 to 30 nucléotides;
the bottom strand is an antisense strand comprising a second nucléotide sequence that is at least about 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% complementary to the RNA corresponding to the target gene, wherein the second nucléotide sequence comprises 15 to 30 nucléotides;
each A is independently a 2’-O-methyl nucléotide or a nucléotide comprising a 5’ stabilized end cap or phosphorylation blocker;
B is a 2’-fluoro nucléotide;
C represents overhanging nucléotides and is a 2’-O-methyl nucléotide.
The ds-siNA may further comprise a conjugated moiety. The conjugated moiety may comprise any of the galactosamines disclosed herein. The ds-siNA may further comprise (i) phosphorothioate intemucleoside linkages between the nucléotides at positions 1 and 2 and positions 2 and 3 from the 5’ end of the sense strand; and (ii) phosphorothioate intemucleoside linkages between the nucléotides at positions 1 and 2; positions 2 and 3; positions 19 and 20; and positions 20 and 21 from the 5’ end of the antisense strand. The ds-siNA may further comprise a 5’-stabilizing end cap. The 5’-stabilizing end cap may be a vinyl phosphonate. The vinyl phosphonate may be a deuterated vinyl phosphonate. The deuterated vinyl phosphonate may be a mono-deuterated vinyl phosphonate. The deuterated vinyl phosphonate may be a mono-dideuterated vinyl phosphonate.The 5’-stabilizing end cap may be attached to the 5’ end of the antisense strand. The 5’-stabilizing end cap may be attached to the 3’ end of the antisense strand. The 5’-stabilizing end cap may be attached to the 5’ end of the sense strand. The 5’-stabilizing end cap may be attached to the 3’ end of the sense strand. In some embodiments, the 2’-Omethyl nucléotide at position 1 from the 5’ end of the sense strand is further modified to contain a 5’ stabilizing end cap. In some embodiments, the 2’-(9-methyl nucléotide at position 1 from the 5’ end of the antisense strand is further modified to contain a 5’ stabilizing end cap. In some embodiments, the 2’-O-methyl nucléotide at position 1 from the 5’ end of the sense strand is further modified to contain a phosphorylation blocker. In some embodiments, the 2’-(9-methyl nucléotide at position 1 from the 3 ’ end of the sense strand is fùrther modified to contain a phosphorylation blocker. In some embodiments, the 2’-(9-methyl nucléotide at position 1 from the 5’ end of the antisense strand is further modified to contain a phosphorylation blocker. In some embodiments, the 2’-(9-methyl nucléotide at position 1 from the 3’ end of the antisense strand is further modified to contain a phosphorylation blocker.
The exemplary ds-siNA shown in FIGs. 3A-3G comprise (i) a sense strand comprising 19-21 nucléotides; and (ii) an antisense strand comprising 21-23 nucléotides. The ds-siNA may further comprise (iii) a conjugated moiety, wherein the conjugated moiety is attached to the 3’ end of the antisense strand. The ds-siNA may comprise a 2 nucléotide overhang consisting of nucléotides at positions 20 and 21 from the 5’ end of the antisense strand. The ds-siNA may comprise a 2 nucléotide overhang consisting of nucléotides at positions 22 and 23 from the 5’ end of the antisense strand. The ds-siNA may further comprise 1, 2, 3, 4, 5, 6 or more phosphorothioate (ps) intemucleoside linkages. At least one phosphorothioate intemucleoside linkage may be between the nucléotides at positions 1 and 2 or positions 2 and 3 from the 5’ end of the sense strand. At least one phosphorothioate intemucleoside linkage may be between the nucléotides at positions 1 and 2 or positions 2 and 3 from the 5’ end of the antisense strand. At least one phosphorothioate intemucleoside linkage may be between the nucléotides at positions 19 and 20, positions 20 and 21, positions 21 and 22, or positions 22 and 23 from the 5’ end of the antisense strand. As shown in FIGs. 3A-3G, 4-6 nucléotides in the sense strand may be 2’-fluoro nucléotides. As shown in FIGs. 3A-3G, 2-5 nucléotides in the antisense strand may be 2’-fluoro nucléotides. As shown in FIGs. 3A-3G, 13-15 nucléotides in the sense strand may be 2’-(9methyl nucléotides. As shown in FIGs. 3A-3G, 14-19 nucléotides in the antisense strand may be 2’-(9-methyl nucléotides. As shown in FIGs. 3A-3G, the ds-siNA does not contain a base pair between 2’-fluoro nucléotides on the sense and antisense strands. In some embodiments, the 2’(2-methyl nucléotide at position 1 from the 5’ end of the sense strand is further modified to contain a 5’ stabilizing end cap. In some embodiments, the 2’-O-methyl nucléotide at position 1 from the 5’ end of the antisense strand is further modified to contain a 5’ stabilizing end cap. In some embodiments, the 2’-O-methyl nucléotide at position 1 from the 5’ end of the sense strand is further modified to contain a phosphorylation blocker. In some embodiments, the 2’-O-methyl nucléotide at position 1 from the 3 ’ end of the sense strand is further modified to contain a phosphorylation blocker. In some embodiments, the 2’-(2-methyl nucléotide at position 1 from the 5’ end of the antisense strand is further modified to contain a phosphorylation blocker. In some embodiments, the 2’-O-methyl nucléotide at position 1 from the 3’ end of the antisense strand is further modified to contain a phosphorylation blocker.
As shown in FIG. 3 A, a ds-siNA may comprise (a) a sense strand consisting of 19 nucléotides, wherein 2’-fluoro nucléotides are at positions 3, 7-9, 12, and 17 from the 5’ end of the sense strand, and wherein 2’-O-methyl nucléotides are at positions 1, 2, 4-6, 10, 11, 13-16, 18, and 19 from the 5’ end of the sense strand; (b) an antisense strand consisting of 21 nucléotides, wherein nucléotides at positions 2 and 14 from the 5’ end of the antisense strand are 2’-fluoro nucléotides; and wherein nucléotides at positions 1, 3-13, and 15-21 are 2’-O-methyl nucléotides. The ds-siNA may further comprise a conjugated moiety attached to the 3’ end of the sense strand. The ds-siNA may further comprise (i) phosphorothioate intemucleoside linkages between the nucléotides at positions 1 and 2 and positions 2 and 3 from the 5’ end of the sense strand; and (ii) phosphorothioate intemucleoside linkages between the nucléotides at positions 1 and 2; positions 2 and 3; positions 19 and 20; and positions 20 and 21 from the 5’ end of the antisense strand. In some embodiments, the 2’-O-methyl nucléotide at position 1 from the 5’ end of the sense strand is further modified to contain a 5’ stabilizing end cap. In some embodiments, the 2’-O-methyl nucléotide at position 1 from the 5’ end of the antisense strand is further modified to contain a 5’ stabilizing end cap. In some embodiments, the 2’-O-methyl nucléotide at position 1 from the 5’ end of the sense strand is further modified to contain a phosphorylation blocker. In some embodiments, the 2’-O-methyl nucléotide at position 1 from the 3’ end of the sense strand is further modified to contain a phosphorylation blocker. In some embodiments, the 2’-O-methyl nucléotide at position 1 from the 5’ end of the antisense strand is further modified to contain a phosphorylation blocker. In some embodiments, the 2’-O-methyl nucléotide at position 1 from the 3 ’ end of the antisense strand is further modified to contain a phosphorylation blocker. In some embodiments, the 2’-O-methyl nucléotide at position 1 from the 5’ end of the sense strand is a d2vd3 nucléotide. In some embodiments, the 2’-O-methyl nucléotide at position 1 from the 5’ end of the antisense strand is a d2vd3 nucléotide. In some embodiments, the 2’-(9-methyl nucléotide at position 1 from the 3’ end of the sense strand is a d2vd3 nucléotide. In some embodiments, the 2’-O-methyl nucléotide at position 1 from the 3’ end of the antisense strand is a d2vd3 nucléotide. In some embodiments, at least 1, 2, 3, 4 or more 2’-fluoro nucléotides on the sense strand or antisense strand is a 2’-fluoro nucléotide mimic.In some embodiments, at least 1, 2, 3, 4 or more 2’-fluoro nucléotides on the sense strand or antisense strand is a f4P, f2P, or IX nucléotide. In some embodiments, at least 1, 2, 3, 4 or more 2’-(9-methyl nucléotide on the sense or antisense strand is a 2’-O-methyl nucléotide mimic.
As shown in FIG. 3B, a ds-siNA may comprise (a) a sense strand consisting of 19 nucléotides, wherein 2’-fluoro nucléotides are at positions 3, 7, 8, and 17 from the 5’ end of the sense strand, and wherein 2’-O-methyl nucléotides are at positions 1, 2, 4-6, 9-16, 18, and 19 from the 5’ end of the sense strand; (b) an antisense strand consisting of 21 nucléotides, wherein nucléotides at positions 2 and 14 from the 5’ end of the antisense strand are 2’-fluoro nucléotides; and wherein nucléotides at positions 1,3-13, and 15-21 are 2’-O-methyl nucléotides. The ds-siNA may further comprise a conjugated moiety attached to the 3’ end of the sense strand. The ds-siNA may further comprise (i) phosphorothioate intemucleoside linkages between the nucléotides at positions 1 and 2 and positions 2 and 3 from the 5’ end of the sense strand; and (ii) phosphorothioate intemucleoside linkages between the nucléotides at positions 1 and 2; positions 2 and 3; positions 19 and 20; and positions 20 and 21 from the 5’ end of the antisense strand. In some embodiments, the 2’-O-methyl nucléotide at position 1 from the 5’ end of the sense strand is further modified to contain a 5’ stabilizing end cap. In some embodiments, the 2’-(9-methyl nucléotide at position 1 from the 5’ end of the antisense strand is further modified to contain a 5’ stabilizing end cap. In some embodiments, the 2’-O-methyl nucléotide at position 1 from the 5’ end of the sense strand is further modified to contain a phosphorylation blocker. In some embodiments, the 2’-(9-methyl nucléotide at position 1 from the 3’ end of the sense strand is further modified to contain a phosphorylation blocker. In some embodiments, the 2’-(9-methyl nucléotide at position 1 from the 5’ end of the antisense strand is further modified to contain a phosphorylation blocker. In some embodiments, the 2’-O-methyl nucléotide at position 1 from the 3 ’ end of the antisense strand is further modified to contain a phosphorylation blocker. In some embodiments, the 2’-O-methyl nucléotide at position 1 from the 5’ end of the sense strand is a d2vd3 nucléotide. In some embodiments, the 2’-(9-methyl nucléotide at position 1 from the 5’ end of the antisense strand is a d2vd3 nucléotide. In some embodiments, the 2’-O-methyl nucléotide at position 1 from the 3’ end of the sense strand is a d2vd3 nucléotide. In some embodiments, the 2’-O-methyl nucléotide at position 1 from the 3’ end of the antisense strand is a d2vd3 nucléotide. In some embodiments, at least 1, 2, 3, 4 or more 2’-fluoro nucléotides on the sense strand or antisense strand is a 2’-fluoro nucléotide mimic. In some embodiments, at least 1, 2, 3, 4 or more 2’-fluoro nucléotides on the sense strand or antisense strand is a f4P, f2P, or IX nucléotide. In some embodiments, at least 1, 2, 3, 4 or more 2’-(9-methyl nucléotide on the sense or antisense strand is a 2’-O-methyl nucléotide mimic.
As shown in FIG. 3C, a ds-siNA may comprise (a) a sense strand consisting of 19 nucléotides, wherein 2’-fluoro nucléotides are at positions 3, 7-9, 12 and 17 from the 5’ end of the sense strand, and wherein 2’-O-methyl nucléotides are at positions 1, 2, 4-6, 10, 11, 13-16, 18, and 19 from the 5’ end of the sense strand; (b) an antisense strand consisting of 21 nucléotides, wherein the nucléotides in the antisense strand comprise an altemating 1:3 modification pattern, and wherein 1 nucléotide is a 2’-fluoro nucléotide and 3 nucléotides are 2’O-methyl nucléotides. The ds-siNA may further comprise a conjugated moiety attached to the 3’ end of the sense strand. The ds-siNA may further comprise (i) phosphorothioate intemucleoside linkages between the nucléotides at positions 1 and 2 and positions 2 and 3 from the 5’ end of the sense strand; and (ii) phosphorothioate intemucleoside linkages between the nucléotides at positions 1 and 2; positions 2 and 3; positions 19 and 20; and positions 20 and 21 from the 5’ end of the antisense strand. The ds-siNA may comprise 2-5 altemating 1:3 modification patterns on the antisense strand. In some embodiments, the 2’-O-methyl nucléotide at position 1 from the 5’ end of the sense strand is further modified to contain a 5’ stabilizing end cap. In some embodiments, the 2’-(9-methyl nucléotide at position 1 from the 5’ end of the antisense strand is further modified to contain a 5’ stabilizing end cap. In some embodiments, the 2’-O-methyl nucléotide at position 1 from the 5’ end of the sense strand is further modified to contain a phosphorylation blocker. In some embodiments, the 2’-O-methyl nucléotide at position 1 from the 3’ end of the sense strand is further modified to contain a phosphorylation blocker. In some embodiments, the 2’-(9-methyl nucléotide at position 1 from the 5’ end of the antisense strand is further modified to contain a phosphorylation blocker. In some embodiments, the 2’-O-methyl nucléotide at position 1 from the 3 ’ end of the antisense strand is further modified to contain a phosphorylation blocker. In some embodiments, the 2’-(9-methyl nucléotide at position 1 from the 5’ end of the sense strand is a d2vd3 nucléotide. In some embodiments, the 2’-O-methyl nucléotide at position 1 from the 5’ end of the antisense strand is a d2vd3 nucléotide. In some embodiments, the 2’-O-methyl nucléotide at position 1 from the 3’ end of the sense strand is a d2vd3 nucléotide. In some embodiments, the 2’-O-methyl nucléotide at position 1 from the 3’ end of the antisense strand is a d2vd3 nucléotide. In some embodiments, at least 1, 2, 3, 4 or more 2’-fluoro nucléotides on the sense strand or antisense strand is a 2’-fluoro nucléotide mimic. In some embodiments, at least 1, 2, 3, 4 or more 2’-fluoro nucléotides on the sense strand is a f4P, f2P, or fX nucléotide. In some embodiments, at least 1, 2, 3, 4 or more 2’-fluoro nucléotides on the antisense strand is a f4P, f2P, or fX nucléotide. In some embodiments, at least 1, 2, 3, 4 or more 2’-(9-methyl nucléotide on the sense or antisense strand is a 2’-O-methyl nucléotide mimic.
As shown in FIG. 3D, a ds-siNA may comprise (a) a sense strand consisting of 19 nucléotides, wherein 2’-fluoro nucléotides are at positions 5 and 7-9 from the 5’ end of the sense strand, and wherein 2’-O-methyl nucléotides are at positions 1-4, 6, and 10-19 from the 5’ end of the sense strand; (b) an antisense strand consisting of 21 nucléotides, wherein the nucléotides in the antisense strand comprise an altemating 1:3 modification pattern, and wherein 1 nucléotide is a 2’-fluoro nucléotide and 3 nucléotides are 2’-O-methyl nucléotides. The ds-siNA may further comprise a conjugated moiety attached to the 3’ end of the sense strand. The ds-siNA may fùrther comprise (i) phosphorothioate intemucleoside linkages between the nucléotides at positions 1 and 2 and positions 2 and 3 from the 5’ end of the sense strand; and (ii) phosphorothioate intemucleoside linkages between the nucléotides at positions 1 and 2; positions 2 and 3; positions 19 and 20; and positions 20 and 21 from the 5’ end of the antisense strand. The ds-siNA may comprise 2-5 altemating 1:3 modification patterns on the antisense strand. The altemating 1:3 modification pattern may start at the nucléotide at any of positions 2, 6, 10, 14, and/or 18 from the 5’ end of the antisense strand. In some embodiments, the 2’-O-methyl nucléotide at position 1 from the 5’ end of the sense strand is further modified to contain a 5’ stabilizing end cap. In some embodiments, the 2’-(9-methyl nucléotide at position 1 from the 5’ end of the antisense strand is further modified to contain a 5’ stabilizing end cap. In some embodiments, the 2’-O-methyl nucléotide at position 1 from the 5’ end of the sense strand is further modified to contain a phosphorylation blocker. In some embodiments, the 2’-O-methyl nucléotide at position 1 from the 3 ’ end of the sense strand is further modified to contain a phosphorylation blocker. In some embodiments, the 2’-(9-methyl nucléotide at position 1 from the 5’ end of the antisense strand is further modified to contain a phosphorylation blocker. In some embodiments, the 2’-O-methyl nucléotide at position 1 from the 3’ end of the antisense strand is fùrther modified to contain a phosphorylation blocker. In some embodiments, the 2’-Omethyl nucléotide at position 1 from the 5’ end of the sense strand is a d2vd3 nucléotide. In some embodiments, the 2’-O-methyl nucléotide at position 1 from the 5’ end of the antisense strand is a d2vd3 nucléotide. In some embodiments, the 2’-O-methyl nucléotide at position 1 from the 3’ end of the sense strand is a d2vd3 nucléotide. In some embodiments, the 2’-O-methyl nucléotide at position 1 from the 3 ’ end of the antisense strand is a d2vd3 nucléotide. In some embodiments, at least 1, 2, 3, 4 or more 2’-fluoro nucléotides on the sense strand or antisense strand is a 2’fluoro nucléotide mimic. In some embodiments, at least 1, 2, 3, 4 or more 2’-fluoro nucléotides on the sense strand is a f4P, f2P, or fX nucléotide. In some embodiments, at least 1, 2, 3, 4 or more 2’-fluoro nucléotides on the antisense strand is a f4P, f2P, or fX nucléotide. In some embodiments, at least 1, 2, 3, 4 or more 2’-O-methyl nucléotide on the sense or antisense strand is a 2’-O-methyl nucléotide mimic.
As shown in FIG. 3E, a ds-siNA may comprise (a) a sense strand consisting of 19 nucléotides, wherein 2’-fluoro nucléotides are at positions 5 and 7-9 from the 5’ end of the sense strand, and wherein 2’-(9-methyl nucléotides are at positions 1-4, 6, and 10-19 from the 5’ end of the sense strand; (b) an antisense strand consisting of 21 nucléotides, wherein the nucléotides in the antisense strand comprise an altemating 1:2 modification pattern, and wherein 1 nucléotide is a 2’-fluoro nucléotide and 2 nucléotides are 2’-O-methyl nucléotides. The ds-siNA may further comprise a conjugated moiety attached to the 3 ’ end of the sense strand. The ds-siNA may further comprise (i) phosphorothioate intemucleoside linkages between the nucléotides at positions 1 and 2 and positions 2 and 3 from the 5’ end of the sense strand; and (ii) phosphorothioate intemucleoside linkages between the nucléotides at positions 1 and 2; positions 2 and 3; positions 19 and 20; and positions 20 and 21 from the 5’ end of the antisense strand. The ds-siNA may comprise 2-5 altemating 1:2 modification patterns on the antisense strand. The altemating 1:2 modification pattern may start at the nucléotide at any of positions 2, 5, 8, 14, and/or 17 from the 5’ end of the antisense strand. In some embodiments, the ds-siNA comprises (a) a sense strand consisting of 19 nucléotides, wherein 2’-fluoro nucléotides are at positions 5 and 7-9 from the 5’ end of the sense strand, and wherein 2’-O-methyl nucléotides are at positions 1-4, 6, and 10-19 from the 5’ end of the sense strand; (b) an antisense strand consisting of 21 nucléotides, wherein 2’-fluoro nucléotides are at positions 2, 5, 8, 14, and 17 from the 5’ end of the antisense strand, and wherein 2’-O-methyl nucléotides are at positions 1,3,4, 6, 7, 9-13, 15, 16, and 18-21 from the 5’ end of the sense strand. In some embodiments, the 2’-(9-methyl nucléotide at position 1 from the 5’ end of the sense strand is further modified to contain a 5’ stabilizing end cap. In some embodiments, the 2’-O-methyl nucléotide at position 1 from the 5’ end of the antisense strand is further modified to contain a 5’ stabilizing end cap. In some embodiments, the 2’-O-methyl nucléotide at position 1 from the 5’ end of the sense strand is further modified to contain a phosphorylation blocker. In some embodiments, the 2’-O-methyl nucléotide at position 1 from the 3 ’ end of the sense strand is further modified to contain a phosphorylation blocker. In some embodiments, the 2’-(9-methyl nucléotide at position 1 from the 5’ end of the antisense strand is further modified to contain a phosphorylation blocker. In some embodiments, the 2’-(2-methyl nucléotide at position 1 from the 3’ end of the antisense strand is further modified to contain a phosphorylation blocker. In some embodiments, the T-Omethyl nucléotide at position 1 from the 5’ end of the sense strand is a d2vd3 nucléotide. In some embodiments, the 2’-O-methyl nucléotide at position 1 from the 5’ end of the antisense strand is a d2vd3 nucléotide. In some embodiments, the 2’-(9-methyl nucléotide at position 1 from the 3’ end of the sense strand is a d2vd3 nucléotide. In some embodiments, the 2’-(9-methyl nucléotide at position 1 from the 3 ’ end of the antisense strand is a d2vd3 nucléotide. In some embodiments, at least 1, 2, 3, 4 or more 2’-fluoro nucléotides on the sense strand or antisense strand is a 2’fluoro nucléotide mimic. In some embodiments, at least 1, 2, 3, 4 or more 2’-fluoro nucléotides on the sense strand is a f4P, f2P, or fX nucléotide. In some embodiments, at least 1, 2, 3, 4 or more 2’-fluoro nucléotides on the antisense strand is a f4P, f2P, or fX nucléotide. In some embodiments, at least 1, 2, 3, 4 or more 2’-O-methyl nucléotide on the sense or antisense strand is a 2’-(9-methyl nucléotide mimic.
As shown in FIG. 3 F, a ds-siNA may comprise (a) a sense strand consisting of 19 nucléotides, wherein 2’-fluoro nucléotides are at positions 5 and 7-9 from the 5’ end of the sense strand, and wherein 2’-O-methyl nucléotides are at positions 1-4, 6, and 10-19 from the 5’ end of the sense strand; (b) an antisense strand consisting of 21 nucléotides, wherein 2’-fluoro nucléotides are at positions 2, 6, 14, and 16 from the 5’ end of the antisense strand, and wherein 2’-O-methyl nucléotides are at positions 1, 3-5, 7-13, 15, and 17-21 from the 5’ end of the antisense strand. The ds-siNA may further comprise a conjugated moiety attached to the 3’ end of the sense strand. The ds-siNA may further comprise (i) phosphorothioate intemucleoside linkages between the nucléotides at positions 1 and 2 and positions 2 and 3 from the 5’ end of the sense strand; and (ii) phosphorothioate intemucleoside linkages between the nucléotides at positions 1 and 2; positions 2 and 3; positions 19 and 20; and positions 20 and 21 from the 5’ end of the antisense strand. In some embodiments, at least 1, 2, 3, 4 or more 2’-fluoro nucléotides on the sense strand or antisense strand is a f4P, f2P, or fX nucléotide. In some embodiments, at least 1, 2, 3, 4 or more 2’-fluoro nucléotides on the sense strand or antisense strand is a f4P nucléotide. In some embodiments, at least 1, 2, 3, or 4 of the 2’-fluoro-nucleotides at positions 2, 6, 14, and 16 from the 5’ end of the antisense strand is a f4P nucléotide. In some embodiments, at least one of the 2’-fluoro-nucleotides at positions 2, 6, 14, and 16 from the 5’ end of the antisense strand is a f4P nucléotide. In some embodiments, at least two of the 2’-fluoro63 nucléotides at positions 2, 6, 14, and 16 from the 5’ end of the antisense strand is a f4P nucléotide. In some embodiments, less than or equal to 3 of the 2’-fluoro-nucleotides at positions 2, 6, 14, and 16 from the 5’ end of the antisense strand is a f4P nucléotide. In some embodiments, less than or equal to 2 of the 2’-fluoro-nucleotides at positions 2, 6, 14, and 16 from the 5’ end of the antisense strand is a f4P nucléotide. In some embodiments, the 2’-fluoronucleotide at position 2 from the 5’ end of the antisense strand is a f4P nucléotide. In some embodiments, the 2’-fluoro-nucleotide at position 6 from the 5’ end of the antisense strand is a f4P nucléotide. In some embodiments, the 2’-fluoro-nucleotide at position 14 from the 5’ end of the antisense strand is a f4P nucléotide. In some embodiments, the 2’-fluoro-nucleotide at position 16 from the 5’ end of the antisense strand is a f4P nucléotide. In some embodiments, at least 1, 2, 3, 4 or more 2’-fluoro nucléotides on the sense strand or antisense strand is a f2P nucléotide. In some embodiments, at least 1, 2, 3, or 4 of the 2’-fluoro-nucleotides at positions 2, 6, 14, and 16 from the 5’ end of the antisense strand is a f2P nucléotide. In some embodiments, at least one of the 2’-fluoro-nucleotides at positions 2, 6, 14, and 16 from the 5’ end of the antisense strand is a f2P nucléotide. In some embodiments, at least two of the 2’-fluoronucleotides at positions 2, 6, 14, and 16 from the 5’ end of the antisense strand is a f2P nucléotide. In some embodiments, less than or equal to 3 of the 2’-fluoro-nucleotides at positions 2, 6, 14, and 16 from the 5’ end of the antisense strand is a f2P nucléotide. In some embodiments, less than or equal to 2 of the 2’-fluoro-nucleotides at positions 2, 6, 14, and 16 from the 5’ end of the antisense strand is a I2P nucléotide. In some embodiments, the 2’-fluoronucleotide at position 2 from the 5’ end of the antisense strand is a f2P nucléotide. In some embodiments, the 2’-fluoro-nucleotide at position 6 from the 5’ end of the antisense strand is a f2P nucléotide. In some embodiments, the 2’-fluoro-nucleotide at position 14 from the 5’ end of the antisense strand is a f2P nucléotide. In some embodiments, the 2’-fluoro-nucleotide at position 16 from the 5’ end of the antisense strand is a f2P nucléotide. In some embodiments, at least 1, 2, 3, 4 or more 2’-fluoro nucléotides on the sense strand or antisense strand is a fX nucléotide. In some embodiments, at least 1, 2, 3, or 4 of the 2’-fluoro-nucleotides at positions 2, 6, 14, and 16 from the 5’ end of the antisense strand is a fX nucléotide. In some embodiments, at least one of the 2’-fluoro-nucleotides at positions 2, 6, 14, and 16 from the 5’ end of the antisense strand is a fX nucléotide. In some embodiments, at least two of the 2’-fluoronucleotides at positions 2, 6, 14, and 16 from the 5’ end of the antisense strand is a fX nucléotide. In some embodiments, less than or equal to 3 of the 2’-fluoro-nucleotides at positions 2, 6, 14, and 16 from the 5’ end of the antisense strand is a fX nucléotide. In some embodiments, less than or equal to 2 of the 2’-fluoro-nucleotides at positions 2, 6, 14, and 16 from the 5’ end of the antisense strand is a fX nucléotide. In some embodiments, the 2’-fluoro-nucleotide at position 2 from the 5’ end of the antisense strand is a fX nucléotide. In some embodiments, the 2’-fluoro-nucleotide at position 6 from the 5’ end of the antisense strand is a fX nucléotide. In some embodiments, the 2’-fluoro-nucleotide at position 14 from the 5’ end of the antisense strand is a fX nucléotide. In some embodiments, the 2’-fluoro-nucleotide at position 16 from the 5’ end of the antisense strand is a fX nucléotide. In some embodiments, the 2’-O-methyl nucléotide at position 1 from the 5’ end of the sense strand is further modified to contain a 5’ stabilizing end cap. In some embodiments, the 2’-O-methyl nucléotide at position 1 from the 5’ end of the antisense strand is further modified to contain a 5’ stabilizing end cap. In some embodiments, the 2’-(9-methyl nucléotide at position 1 from the 5’ end of the sense strand is further modified to contain a phosphorylation blocker. In some embodiments, the 2’-O-methyl nucléotide at position 1 from the 3 ’ end of the sense strand is further modified to contain a phosphorylation blocker. In some embodiments, the 2’-O-methyl nucléotide at position 1 from the 5’ end of the antisense strand is further modified to contain a phosphorylation blocker. In some embodiments, the 2’-O-methyl nucléotide at position 1 from the 3’ end of the antisense strand is further modified to contain a phosphorylation blocker. In some embodiments, the 2’-(9methyl nucléotide at position 1 from the 5’ end of the sense strand is a d2vd3 nucléotide. In some embodiments, the 2’-O-methyl nucléotide at position 1 from the 5’ end of the antisense strand is a d2vd3 nucléotide. In some embodiments, the 2’-O-methyl nucléotide at position 1 from the 3’ end of the sense strand is a d2vd3 nucléotide. In some embodiments, the 2’-O-methyl nucléotide at position 1 from the 3 ’ end of the antisense strand is a d2vd3 nucléotide. In some embodiments, at least 1, 2, 3, 4 or more 2’-fluoro nucléotides on the sense strand or antisense strand is a 2’fluoro nucléotide mimic. In some embodiments, at least 1, 2, 3, 4 or more 2’-fluoro nucléotides on the sense strand is a f4P, f2P, or fX nucléotide. In some embodiments, at least 1, 2, 3, 4 or more 2’-fluoro nucléotides on the antisense strand is a f4P, f2P, or fX nucléotide. In some embodiments, at least 1, 2, 3, 4 or more 2’-(9-methyl nucléotide on the sense or antisense strand is a 2’-O-methyl nucléotide mimic.
As shown in FIG. 3G, a ds-siNA may comprise (a) a sense strand consisting of 21 nucléotides, wherein 2’-fluoro nucléotides are at positions 5, 9-11, 14, and 19 from the 5’ end of the sense strand, and wherein 2’-<9-methyl nucléotides are at positions 1-4, 6-8, 12, 13, 15-18, 20, and 21 from the 5’ end of the sense strand; and (b) an antisense strand consisting of 23 nucléotides, wherein 2’-flouro nucleodies are at positions 2 and 14 from the 5’ end of the antisense strand, and wherein 2’-O-methyl nucléotides are at positions 1, 3-13, and 15-23 from the 5’ end of the antisense strand. The ds-siNA may further comprise a conjugated moiety attached to the 3’ end of the sense strand. The ds-siNA may further comprise (i) phosphorothioate intemucleoside linkages between the nucléotides at positions 1 and 2 and positions 2 and 3 from the 5’ end of the sense strand; and (ii) phosphorothioate intemucleoside linkages between the nucléotides at positions 1 and 2; positions 2 and 3; positions 19 and 20; and positions 20 and 21 from the 5’ end of the antisense strand. In some embodiments, the 2’-Omethyl nucléotide at position 1 from the 5’ end of the sense strand is further modified to contain a 5’ stabilizing end cap. In some embodiments, the 2’-O-methyl nucléotide at position 1 from the 5’ end of the antisense strand is further modified to contain a 5’ stabilizing end cap. In some embodiments, the 2’-(9-methyl nucléotide at position 1 from the 5’ end of the sense strand is further modified to contain a phosphorylation blocker. In some embodiments, the 2’-(9-methyl nucléotide at position 1 from the 3 ’ end of the sense strand is further modified to contain a phosphorylation blocker. In some embodiments, the 2’-O-methyl nucléotide at position 1 from the 5’ end of the antisense strand is further modified to contain a phosphorylation blocker. In some embodiments, the 2’-O-methyl nucléotide at position 1 from the 3’ end of the antisense strand is further modified to contain a phosphorylation blocker. In some embodiments, the 2’-(9methyl nucléotide at position 1 from the 5’ end of the sense strand is a d2vd3 nucléotide. In some embodiments, the 2’-O-methyl nucléotide at position 1 from the 5’ end of the antisense strand is a d2vd3 nucléotide. In some embodiments, the 2’-(2-methyl nucléotide at position 1 from the 3’ end of the sense strand is a d2vd3 nucléotide. In some embodiments, the 2’-O-methyl nucléotide at position 1 from the 3 ’ end of the antisense strand is a d2vd3 nucléotide. In some embodiments, at least 1, 2, 3, 4 or more 2’-fluoro nucléotides on the sense strand or antisense strand is a 2’fluoro nucléotide mimic. In some embodiments, at least 1, 2, 3, 4 or more 2’-fluoro nucléotides on the sense strand is a f4P, f2P, or fX nucléotide. In some embodiments, at least 1, 2, 3, 4 or more 2’-fluoro nucléotides on the antisense strand is a f4P, f2P, or fX nucléotide. In some embodiments, at least 1, 2, 3, 4 or more 2’-O-methyl nucléotide on the sense or antisense strand is a 2’-O-methyl nucléotide mimic.
Any of the siNAs disclosed herein may comprise a sense strand and an antisense strand. The sense strand may comprise a first nucléotide sequence that is 15 to 30 nucléotides in length. The antisense strand may comprise a second nucléotide sequence that is 15 to 30 nucléotides in length.
In some embodiments, the double-stranded short interfering nucleic acid (ds-siNA) molécule comprises: (a) a sense strand comprising a first nucléotide sequence that is at least about 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% identical to an RNA corresponding to a target gene, wherein the first nucléotide sequence: (i) is 15 to 30 nucléotides in length; and (ii) comprises 15 or more modified nucléotides independently selected from a 2’-O-methyl nucléotide and a 2’-fluoro nucléotide, wherein at least one modified nucléotide is a 2’-O-methyl nucléotide and the nucléotide at position 3, 5, 7, 8, 9, 10, 11, 12, 14, 17, and/or 19 from the 5’ end of the first nucléotide sequence is a 2’-fluoro nucléotide; and (b) an antisense strand comprising a second nucléotide sequence that is at least about 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% complementary to the RNA corresponding to the target gene, wherein the second nucléotide sequence: (i) is 15 to 30 nucléotides in length; and (ii) comprises 15 or more modified nucléotides independently selected from a 2’-O-methyl nucléotide and a 2’-fluoro nucléotide, wherein at least one modified nucléotide is a 2’-O-methyl nucléotide and at least one modified nucléotide is a 2’-fluoro nucléotide.
In some embodiments, the double-stranded short interfering nucleic acid (ds-siNA) molécule comprises: (a) a sense strand comprising a first nucléotide sequence that is at least about 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% identical to an RNA corresponding to a target gene, wherein the first nucléotide sequence: (i) is 15 to 30 nucléotides in length; and (ii) comprises 15 or more modified nucléotides independently selected from a 2’-(9-methyl nucléotide and a 2’-fluoro nucléotide, wherein at least one modified nucléotide is a 2’-O-methyl nucléotide and the nucléotide at position 3, 5, 7, 8, 9, 10, 11, 12, 14, 17, and/or 19 from the 5’ end of the first nucléotide sequence is a 2’-fluoro nucléotide; and (b) an antisense strand comprising a second nucléotide sequence that is at least about 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% complementary to the RNA corresponding to the target gene, wherein the second nucléotide sequence: (i) is 15 to 30 nucléotides in length; and (ii) comprises 15 or more modified nucléotides independently selected from a 2’-O-methyl nucléotide and a 2’-fluoro nucléotide, wherein at least one modified nucléotide is a 2’-(9-methyl nucléotide and at least one modified nucléotide is a 2’-fluoro nucléotide.
In some embodiments, the double-stranded short interfering nucleic acid (ds-siNA) molécule comprises: (a) a sense strand comprising a first nucléotide sequence that is at least about 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% identical to an RNA corresponding to a target gene, wherein the first nucléotide sequence: (i) is 15 to 30 nucléotides in length; and (ii) comprises 15 or more modified nucléotides independently selected from a 2’-O-methyl nucléotide and a 2’-fluoro nucléotide, wherein at least one modified nucléotide is a 2’-O-methyl nucléotide and the nucléotide at position 7 from the 5’ end of the first nucléotide sequence is a 2’-fluoro nucléotide; and (b) an antisense strand comprising a second nucléotide sequence that is at least about 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% complementary to the RNA corresponding to the target gene, wherein the second nucléotide sequence: (i) is 15 to 30 nucléotides in length; and (ii) comprises 15 or more modified nucléotides independently selected from a 2’-(9-methyl nucléotide and a 2’-fluoro nucléotide, wherein at least one modified nucléotide is a 2’-O-methyl nucléotide and at least one modified nucléotide is a 2’-fluoro nucléotide.
In some embodiments, the double-stranded short interfering nucleic acid (ds-siNA) molécule comprises: (a) a sense strand comprising a first nucléotide sequence that is at least about 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% identical to an RNA corresponding to a target gene, wherein the first nucléotide sequence: (i) is 15 to 30 nucléotides in length; and (ii) comprises 15 or more modified nucléotides independently selected from a 2’-O-methyl nucléotide and a 2’-fluoro nucléotide, wherein at least one modified nucléotide is a 2’-O-methyl nucléotide and the nucléotide at position 7, 9, 10, and/or 11 from the 5’ end of the first nucléotide sequence is a 2’-fluoro nucléotide; and (b) an antisense strand comprising a second nucléotide sequence that is at least about 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% complementary to the RNA corresponding to the target gene, wherein the second nucléotide sequence: (i) is 15 to 30 nucléotides in length; and (ii) comprises 15 or more modified nucléotides independently selected from a 2’-O-methyI nucléotide and a 2’-fluoro nucléotide, wherein at least one modified nucléotide is a 2’-O-methyl nucléotide and at least one modified nucléotide is a 2’-fluoro nucléotide.
In some embodiments, the double-stranded short interfering nucleic acid (ds-siNA) molécule comprises: (a) a sense strand comprising a first nucléotide sequence that is at least about 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% identical to an RNA corresponding to a target gene, wherein the first nucléotide sequence: (i) is 15 to 30 nucléotides in length; and (ii) comprises 15 or more modified nucléotides independently selected from a 2’-(9-methyl nucléotide and a 2’-fluoro nucléotide, wherein at least one modified nucléotide is a 2’-O-methyl nucléotide and at least one modified nucléotide is a 2’-fluoro nucléotide; and (b) an antisense strand comprising a second nucléotide sequence that is at least about 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% complementary to the RNA corresponding to the target gene, wherein the second nucléotide sequence: (i) is 15 to 30 nucléotides in length; and (ii) comprises 15 or more modified nucléotides independently selected from a 2’-(9-methyl nucléotide and a 2’fluoro nucléotide, wherein at least one modified nucléotide is a 2’-O-methyl nucléotide and the nucléotide at position 2, 5, 6, 8, 10, 14, 16, 17, and/or 18 from the 5’ end of the second nucléotide sequence is a 2’-fluoro nucléotide.
In some embodiments, the double-stranded short interfering nucleic acid (ds-siNA) molécule comprises: (a) a sense strand comprising a first nucléotide sequence that is at least about 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% identical to an RNA corresponding to a target gene, wherein the first nucléotide sequence: (i) is 15 to 30 nucléotides in length; and (ii) comprises 15 or more modified nucléotides independently selected from a 2’-O-methyl nucléotide and a 2’-fluoro nucléotide, wherein at least one modified nucléotide is a 2’-O-methyl nucléotide and at least one modified nucléotide is a 2’-fluoro nucléotide; and (b) an antisense strand comprising a second nucléotide sequence that is at least about 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% complementary to the RNA corresponding to the target gene, wherein the second nucléotide sequence: (i) is 15 to 30 nucléotides in length; and (ii) comprises 15 or more modified nucléotides independently selected from a 2’-O-methyl nucléotide and a 2’fluoro nucléotide, wherein at least one modified nucléotide is a 2’-O-methyl nucléotide and the nucléotide at position 2 of the second nucléotide sequence is a 2’-fluoro nucléotide.
In some embodiments, the double-stranded short interfering nucleic acid (ds-siNA) molécule comprises: (a) a sense strand comprising a first nucléotide sequence that is at least about 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% identical to an RNA corresponding to a target gene, wherein the first nucléotide sequence: (i) is 15 to 30 nucléotides in length; (ii) comprises 15 or more modified nucléotides independently selected from a 2’-O-methyl nucléotide and a 2’-fluoro nucléotide, wherein at least one modified nucléotide is a 2’-O-methyl nucléotide and at least one modified nucléotide is a 2’-fluoro nucléotide; and (iii) comprises 1 or more phosphorothioate intemucleoside linkage; and (b) an antisense strand comprising a second nucléotide sequence that is at least about 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% complementary to the RNA corresponding to the target gene, wherein the second nucléotide sequence: (i) is 15 to 30 nucléotides in length; (ii) comprises 15 or more modified nucléotides independently selected from a 2’-O-methyl nucléotide and a 2’-fluoro nucléotide, wherein at least one modified nucléotide is a 2’-(9-methyl nucléotide and at least one modified nucléotide is a 2’-fluoro nucléotide; and (iii) comprises 1 or more phosphorothioate intemucleoside linkage.
In some embodiments, the double-stranded short interfering nucleic acid (ds-siNA) molécule comprises: (a) a sense strand comprising a first nucléotide sequence that is at least about 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% identical to an RNA corresponding to a target gene, wherein the first nucléotide sequence: (i) is 15 to 30 nucléotides in length; and (ii) comprises 15 or more modified nucléotides independently selected from a 2’-O-methyl nucléotide and a 2’-fluoro nucléotide, wherein at least one modified nucléotide is a 2’-O-methyl nucléotide and at least one modified nucléotide is a 2’-fluoro nucléotide; and (b) an antisense strand comprising a second nucléotide sequence that is at least about 60%, 65%, 70%, 75%,
80%, 85%, 90%, 95%, or 100% complementary to the RNA corresponding to the target gene, wherein the second nucléotide sequence: (i) is 15 to 30 nucléotides in length; and (ii) comprises 15 or more modified nucléotides independently selected from a 2’-O-methyl nucléotide and a 2’fluoro nucléotide, wherein at least one modified nucléotide is a 2’-O-methyl nucléotide and at least one modified nucléotide is a 2’-fluoro nucléotide, wherein the ds-siNA may further comprise a phosphorylation blocker, a galactosamine, or 5’-stabilized end cap.
In some embodiments, the double-stranded short interfering nucleic acid (ds-siNA) molécule comprises: (I) a sense strand comprising (A) a first nucléotide sequence that is at least about 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% identical to an RNA corresponding to a target gene, wherein the first nucléotide sequence: (i) is 15 to 30 nucléotides in length; and (ii) comprises 15 or more modified nucléotides independently selected from a 2’-O-methyl nucléotide and a 2’-fluoro nucléotide, wherein at least one modified nucléotide is a 2’-O-methyl nucléotide and at least one modified nucléotide is a 2’-fluoro nucléotide; and (B) a phosphorylation blocker or a galactosamine; and (II) an antisense strand comprising a second nucléotide sequence that is at least about 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% complementary to the RNA corresponding to the target gene, wherein the second nucléotide sequence: (a) is 15 to 30 nucléotides in length; and (b) comprises 15 or more modified nucléotides independently selected from a 2’-O-methyl nucléotide and a 2’-fluoro nucléotide, wherein at least one modified nucléotide is a 2’-(2-methyl nucléotide and at least one modified nucléotide is a 2’-fluoro nucléotide.
In some embodiments, the double-stranded short interfering nucleic acid (ds-siNA) molécule comprises: (I) a sense strand comprising a first nucléotide sequence that is at least about 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% identical to an RNA corresponding to a target gene, wherein the first nucléotide sequence: (a) is 15 to 30 nucléotides in length; and (b) comprises 15 or more modified nucléotides independently selected from a 2’-O-methyl nucléotide and a 2’-fluoro nucléotide, wherein at least one modified nucléotide is a 2’-O-methyl nucléotide and at least one modified nucléotide is a 2’-fluoro nucléotide; and (II) an antisense strand comprising (A) a second nucléotide sequence that is at least about 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% complementary to the RNA corresponding to the target gene, wherein the second nucléotide sequence: (i) is 15 to 30 nucléotides in length; and (ii) comprises 15 or more modified nucléotides independently selected from a 2’-O-methyl nucléotide and a 2’fhioro nucléotide, wherein at least one modified nucléotide is a 2’-(9-methyl nucléotide and at least one modified nucléotide is a 2’-fluoro nucléotide; and (B) a 5’-stabilized end cap.
In some embodiments, the double-stranded short interfering nucleic acid (ds-siNA) molécule comprises: (I) a sense strand comprising (A) a first nucléotide sequence that is at least about 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% identical to an RNA corresponding to a target gene, wherein the first nucléotide sequence: (i) is 15 to 30 nucléotides in length; and (ii) comprises 15 or more modified nucléotides independently selected from a 2’-O-methyl nucléotide and a 2’-fluoro nucléotide, wherein at least one modified nucléotide is a 2’-O-methyl nucléotide and at least one modified nucléotide is a 2’-fluoro nucléotide; and (B) a phosphorylation blocker or a galactosamine; and (II) an antisense strand comprising (A) a second nucléotide sequence that is at least about 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% complementary to the RNA corresponding to the target gene, wherein the second nucléotide sequence: (i) is 15 to 30 nucléotides in length; and (ii) comprises 15 or more modified nucléotides independently selected from a 2’-O-methyl nucléotide and a 2’-fluoro nucléotide, wherein at least one modified nucléotide is a 2’-O-methyl nucléotide and at least one modified nucléotide is a 2’-fluoro nucléotide; and (B) a 5’-stabilized end cap.
In some embodiments, the double-stranded short interfering nucleic acid (ds-siNA) molécule comprises: (a) a sense strand comprising a first nucléotide sequence that is at least about 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% identical to an RNA corresponding to a target gene, wherein the first nucléotide sequence comprises a nucléotide sequence of any one SEQ ID NOs: 1-56, 103-158, and 205-260; and (b) an antisense strand comprising a second nucléotide sequence that is at least about 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% complementary to the RNA corresponding to the target gene, wherein the second nucléotide sequence comprises a nucléotide sequence of any one of SEQ ID NOs: 57-102, 159-204, and 261-306. In some embodiments, the double-stranded short interfering nucleic acid (ds-siNA) molécule comprises: (a) a sense strand comprising a first nucléotide sequence that is at least about 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% identical to an RNA corresponding to a target gene, wherein the first nucléotide sequence comprises a nucléotide sequence as shown in Tables 1-3; and (b) an antisense strand comprising a second nucléotide sequence that is at least about 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% complementary to the RNA corresponding to the target gene, wherein the second nucléotide sequence comprises a nucléotide sequence as shown in Tables 1-3.
In some embodiments, the double-stranded short interfering nucleic acid (ds-siNA) molécule comprises: (a) a sense strand comprising a nucléotide sequence of any one of SEQ ID NOs: 307-362 and 415-444; and (b) an antisense strand comprising a nucléotide sequence of any one of SEQ ID NOs: 363-409, 445-533, and 536-539. In some embodiments, the ds-siNA molécule comprises a double-stranded molécule as identified by the duplex ID (e.g., ds-siNA001 to ds-siNA-0178) shown in Tables 6 and 10.
Further disclosed herein are compositions comprising two or more of the siNA molécules described herein.
Further disclosed herein are compositions comprising any of the siNA molécule described and a pharmaceutically acceptable carrier or diluent.
Further disclosed herein are compositions comprising two or more of the siNA molécules described herein for use as a médicament.
Further disclosed herein are compositions comprising any of the siNA molécule described and a pharmaceutically acceptable carrier or diluent for use as a médicament.
Further disclosed herein are methods of treating a disease in a subject in need thereof, the method comprising administering to the subject any of the siNA molécules described herein.
Further disclosed herein are uses of any of the siNA molécules described herein in the manufacture of a médicament for treating a disease.
Short interfering nucleic acid (siNA) molécules
As indicated above, the présent disclosure provides siNA molécules comprising modifïed nucléotides. Any of the siNA molécules described herein may be double-stranded siNA (dssiNA) molécules. The terms “siNA molécules” and “ds-siNA molécules” may be used interchangeably. In some embodiments, the ds-siNA molécules comprise a sense strand and an antisense strand.
Further disclosed herein are siNA molécules comprising (a) at least one phosphorylation blocker, conjugated moiety, or 5’-stabilized end cap; and (b) a short interfering nucleic acid (siNA). In some embodiments, the phosphorylation blocker is a phosphorylation blocker disclosed herein. In some embodiments, the conjugated moiety is a galactosamine disclosed herein. In some embodiments, the 5’-stabilized end cap is a 5’-stabilized end cap disclosed herein. The siNA may comprise any of the first nucléotide, second nucléotide, sense strand, or antisense strand sequences disclosed herein. The siNA may comprise 5 to 100, 5 to 90, 10 to 100, 10 to 90, 10 to 80, 10 to 70, 10 to 60, 10 to 50, 10 to 30, 10 to 25, 15 to 100, 15 to 90, 15 to 80, 15 to 70, 15 to 60, 15 to 50, 15 to 30, or 15 to 25 nucléotides. The siNA may comprise at least 5, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20,21,22, 23,24, 25,26, 27, 28, 29,30,31,32,33, 34, 35, 36, 37, 38, 39, or 40 nucléotides. The siNA may comprise less than or equal to 50, 45, 40, 39, 38, 37, 36, 35, 34, 33, 32, 31, 30, 29, 28, 27, 26, 25, 24, 23, 22, 21, 20, or 19 nucléotides. The nucléotides may be modifïed nucléotides. The siNA may be single stranded. The siNA may be double stranded. The siNA may comprise (a) a sense strand comprising 15 to 30, 15 to 25, 15 to
24, 15 to 23, 15 to 22, 15 to 21, 17 to 30, 17 to 25, 17 to 24, 17 to 23, 17 to 22, 17 to 21, 18 to
30, 18 to 25, 18 to 24, 18 to 23, 18 to 22, 18 to 21, 19 to 30, 19 to 25, 19 to 24, 19 to 23, 19 to
22, 19 to 21, 20 to 25, 20 to 24, 20 to 23, 21 to 25, 21 to 24, or 21 to 23 nucléotides; and (b) an antisense strand comprising 15 to 30, 15 to 25, 15 to 24, 15 to 23, 15 to 22, 15 to 21, 17 to 30, 17 to 25, 17 to 24, 17 to 23, 17 to 22, 17 to 21, 18 to 30, 18 to 25, 18 to 24, 18 to 23, 18 to 22, 18 to 21, 19 to 30, 19 to 25, 19 to 24, 19 to 23, 19 to 22, 19 to 21, 20 to 25, 20 to 24, 20 to 23, 21 to 25, 21 to 24, or 21 to 23 nucléotides. The siNA may comprise (a) a sense strand comprising about 15, 16, 17, 18, 19, 20, 21, 22, or 23 nucléotides; and (b) an antisense strand comprising about 15, 16, 17, 18, 19, 20, 21, 22, or 23 nucléotides. The siNA may comprise (a) a sense strand comprising about 19 nucléotides; and (b) an antisense strand comprising about 21 nucléotides. The siNA may comprise (a) a sense strand comprising about 21 nucléotides; and (b) an antisense strand comprising about 23 nucléotides.
In some embodiments, any of the siNA molécules disclosed herein further comprise one or more linkers independently selected from a phosphodiester (PO) linker, phosphorothioate (PS) linker, phosphorodithioate linker, and PS-mimic linker. In some embodiments, the PS-mimic linker is a sulfur linker. In some embodiments, the linkers are intemucleoside linkers. Altematively, or additionally, the linkers connect a nucléotide of the siNA molécule to at least one phosphorylation blocker, conjugated moiety, or 5’-stabilized end cap. In some embodiments, the linkers connect a conjugated moiety to a phosphorylation blocker or 5’stabilized end cap. siNA sense strand
Any of the siNA molécules described herein may comprise a sense strand. The sense strand may comprise a first nucléotide sequence. The first nucléotide sequence may be 15 to 30, 15 to 25, 15 to 23, 17 to 23, 19 to 23, or 19 to 21 nucléotides in length. In some embodiments, the first nucléotide sequence is 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30 nucléotides in length. In some embodiments, the first nucléotide sequence is at least 19 nucléotides in length. In some embodiments, the first nucléotide sequence is at least 21 nucléotides in length.
In some embodiments, the sense strand is the same length as the first nucléotide sequence. In some embodiments, the sense strand is longer than the first nucléotide sequence. In some embodiments, the sense strand may further comprise 1, 2, 3, 4, or 5 or more nucléotides than the first nucléotide sequence. In some embodiments, the sense strand may further comprise a deoxyribonucleic acid (DNA). In some embodiments, the DNA is thymine (T). In some embodiments, the sense strand may further comprise a TT sequence. In some embodiments, the
I sense strand may further comprise one or more modified nucléotides that are adjacent to the first nucléotide sequence. In some embodiments, the one or more modified nucléotides are independently selected from any of the modified nucléotides disclosed herein (e.g., 2’-fluoro nucléotide, 2’-O-methyl nucléotide, 2’-fluoro nucléotide mimic, 2’-O-methyl nucléotide mimic, or a nucléotide comprising a modified nucleobase).
In some embodiments, the first nucléotide sequence comprises 15, 16, 17, 18, 19, 20, 21, 22, 23, or more modified nucléotides independently selected from a 2’-(9-methyl nucléotide and a 2'-fluoro nucléotide. In some embodiments, 70%, 75%, 80%, 85%, 90%, 95% or 100% of the nucléotides in the first nucléotide sequence are modified nucléotides independently selected from a 2’-O-methyl nucléotide and a 2’-fluoro nucléotide. In some embodiments, 100% of the nucléotides in the first nucléotide sequence are modified nucléotides independently selected from a 2’-O-methyl nucléotide and a 2’-fluoro nucléotide. In some embodiments, the 2’-O-methyl nucléotide is a 2’-(9-methyl nucléotide mimic. In some embodiments, the 2’-fluoro nucléotide is a 2’-fluoro nucléotide mimic.
In some embodiments, between about 15 to 30, 15 to 25, 15 to 24, 15 to 23, 15 to 22, 15 to 21, 17 to 30, 17 to 25, 17 to 24, 17 to 23, 17 to 22, 17 to 21, 18 to 30, 18 to 25, 18 to 24, 18 to 23, 18 to 22, 18 to 21, 19 to 30, 19 to 25, 19 to 24, 19 to 23, 19 to 22, 19 to 21, 20 to 25, 20 to 24, 20 to 23, 21 to 25, 21 to 24, or 21 to 23 modified nucléotides of the first nucléotide sequence are 2’-O-methyl nucléotides. In some embodiments, between about 2 to 20 modified nucléotides of the first nucléotide sequence are 2’-(9-methyl nucléotides. In some embodiments, between about 5 to 25 modified nucléotides of the first nucléotide sequence are 2’-O-methyl nucléotides. In some embodiments, between about 10 to 25 modified nucléotides of the first nucléotide sequence are 2’-O-methyl nucléotides. In some embodiments, between about 12 to 25 modified nucléotides of the first nucléotide sequence are 2’-O-methyl nucléotides. In some embodiments, at least about 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, or 22 modified nucléotides of the first nucléotide sequence are 2’-O-methyl nucléotides. In some embodiments, at least about 12 modified nucléotides of the first nucléotide sequence are 2’-O-methyl nucléotides. In some embodiments, at least about 13 modified nucléotides of the first nucléotide sequence are 2’-Omethyl nucléotides. In some embodiments, at least about 14 modified nucléotides of the first nucléotide sequence are 2’-(9-methyl nucléotides. In some embodiments, at least about 15 modified nucléotides of the first nucléotide sequence are 2’-O-methyl nucléotides. In some embodiments, at least about 16 modified nucléotides of the first nucléotide sequence are T-Omethyl nucléotides. In some embodiments, at least about 17 modified nucléotides of the first nucléotide sequence are 2’-(9-methyl nucléotides. In some embodiments, at least about 18 modified nucléotides of the first nucléotide sequence are 2’-O-methyl nucléotides. In some embodiments, at least about 19 modified nucléotides of the first nucléotide sequence are 2’-(2methyl nucléotides. In some embodiments, less than or equal to 25, 24, 23, 22, 21, 20, 19, 18, 17, 16, 15, 14, 13, 12, 11, 10, 9, 8, 7, 6, 5, 4, 3, or 2 modified nucléotides of the first nucléotide sequence are 2’-O-methyl nucléotides. In some embodiments, less than or equal to 21 modified nucléotides of the first nucléotide sequence are 2’-O-methyl nucléotides. In some embodiments, less than or equal to 20 modified nucléotides of the first nucléotide sequence are 2’-(2-methyl nucléotides. In some embodiments, less than or equal to 19 modified nucléotides of the first nucléotide sequence are 2’-(9-methyl nucléotides. In some embodiments, less than or equal to 18 modified nucléotides of the first nucléotide sequence are 2’-O-methyl nucléotides. In some embodiments, less than or equal to 17 modified nucléotides of the first nucléotide sequence are 2’-O-methyl nucléotides. In some embodiments, less than or equal to 16 modified nucléotides of the first nucléotide sequence are 2’-O-methyl nucléotides. In some embodiments, less than or equal to 15 modified nucléotides of the first nucléotide sequence are 2’-O-methyl nucléotides. In some embodiments, less than or equal to 14 modified nucléotides of the first nucléotide sequence are 2’-O-methyl nucléotides. In some embodiments, less than or equal to 13 modified nucléotides of the first nucléotide sequence are 2’-O-methyl nucléotides. In some embodiments, at least one modified nucléotide of the first nucléotide sequence is a 2’-O-methyl pyrimidine. In some embodiments, at least 5, 6, 7, 8, 9, or 10 modified nucléotides of the first nucléotide sequence are 2’-O-methyl pyrimidines. In some embodiments, at least one modified nucléotide of the first nucléotide sequence is a 2’-O-methyl purine. In some embodiments, at least 5, 6, 7, 8, 9, or 10 modified nucléotides of the first nucléotide sequence are 2’-(9-methyl purines. In some embodiments, the 2’-(9-methyl nucléotide is a 2’-O-methyl nucléotide mimic.
In some embodiments, between 2 to 15 modified nucléotides of the first nucléotide sequence are 2’-fluoro nucléotides. In some embodiments, between 2 to 10 modified nucléotides of the first nucléotide sequence are 2’-fluoro nucléotides. In some embodiments, between 2 to 6 modified nucléotides of the first nucléotide sequence are 2’-fluoro nucléotides. In some embodiments, 1 to 6, 1 to 5, 1 to 4, or 1 to 3 modified nucléotides of the first nucléotide sequence are 2’-fluoro nucléotides. In some embodiments, at least 1, 2, 3, 4, 5, or 6 modified nucléotides of the first nucléotide sequence are 2’-fluoro nucléotides. In some embodiments, at least 1 modified nucléotide of the first nucléotide sequence is a 2’-fluoro nucléotide. In some embodiments, at least 2 modified nucléotides of the first nucléotide sequence are 2’-fluoro nucléotides. In some embodiments, at least 3 modified nucléotides of the first nucléotide sequence are 2’-fluoro nucléotides. In some embodiments, at least 4 modified nucléotides of the first nucléotide sequence are 2’-fluoro nucléotides. In some embodiments, at least 5 modified nucléotides of the first nucléotide sequence are 2’-fluoro nucléotides. In some embodiments, at least 6 modified nucléotides of the first nucléotide sequence are 2’-fluoro nucléotides. In some embodiments, 10, 9, 8, 7, 6, 5, 4, 3 or fewer modified nucléotides of the first nucléotide sequence are 2’-fluoro nucléotides. In some embodiments, 10 or fewer modified nucléotides of the first nucléotide sequence are 2’-fluoro nucléotides. In some embodiments, 7 or fewer modified nucléotides of the first nucléotide sequence are 2’-fluoro nucléotides. In some embodiments, 6 or fewer modified nucléotides of the first nucléotide sequence are 2’-fluoro nucléotides. In some embodiments, 5 or fewer modified nucléotides of the first nucléotide sequence are 2’-fluoro nucléotides. In some embodiments, 4 or fewer modified nucléotides of the first nucléotide sequence are 2’-fluoro nucléotides. In some embodiments, 3 or fewer modified nucléotides of the first nucléotide sequence are 2’-fluoro nucléotides. In some embodiments, 2 or fewer modified nucléotides of the first nucléotide sequence are 2’-fluoro nucléotides. In some embodiments, at least one modified nucléotide of the first nucléotide sequence is a 2’-fluoro pyrimidine. In some embodiments, 1, 2, 3, 4, 5, or 6 modified nucléotides of the first nucléotide sequence are 2’fluoro pyrimidines. In some embodiments, at least one modified nucléotide of the first nucléotide sequence is a 2’-fluoro purine. In some embodiments, 1, 2, 3, 4, 5, or 6 modified nucléotides of the first nucléotide sequence are 2’-fluoro purines. In some embodiments, the 2’-fluoro nucléotide is a 2’-fluoro nucléotide mimic.
In some embodiments, the nucléotide at position 3, 5, 7, 8, 9, 10, 11, 12, 14, 17, and/or 19 from the 5’ end of the first nucléotide sequence is a 2’-fluoro nucléotide. In some embodiments, at least two nucléotides at positions 3, 5, 7, 8, 9, 10, 11, 12, 14, 17, and/or 19 from the 5’ end of the first nucléotide sequence are 2’-fluoro nucléotides. In some embodiments, at least three nucléotides at positions 3, 5, 7, 8, 9, 10, 11, 12, 14, 17, and/or 19 from the 5’ end of the first nucléotide sequence are 2’-fluoro nucléotides. In some embodiments, at least four nucléotides at positions 3, 5, 7, 8, 9, 10, 11, 12, 14, 17, and/or 19 from the 5’ end of the first nucléotide sequence are 2’-fhioro nucléotides. In some embodiments, at least five nucléotides at positions 3, 5, 7, 8, 9, 10, 11, 12, 14, 17, and/or 19 from the 5’ end of the first nucléotide sequence are 2’fluoro nucléotides. In some embodiments, the nucléotides at positions 3, 5, 7, 8, 9, 10, 11, 12, 14, 17, and/or 19 from the 5’ end of the first nucléotide sequence are 2’-fluoro nucléotides. In some embodiments, the nucléotide at position 3 from the 5’ end of the first nucléotide sequence is a 2’fluoro nucléotide. In some embodiments, the nucléotide at position 7 from the 5’ end of the first nucléotide sequence is a 2’-fluoro nucléotide. In some embodiments, the nucléotide at position 8 from the 5’ end of the first nucléotide sequence is a 2’-fluoro nucléotide. In some embodiments, the nucléotide at position 9 from the 5’ end of the first nucléotide sequence is a 2’-fluoro nucléotide. In some embodiments, the nucléotide at position 12 from the 5’ end of the first nucléotide sequence is a 2’-fluoro nucléotide. In some embodiments, the nucléotide at position 17 from the 5’ end of the first nucléotide sequence is a 2’-fluoro nucléotide. In some embodiments, the 2’-fluoro nucléotide is a 2’-fluoro nucléotide mimic.
In some embodiments, at least 1, 2, 3, 4, 5, 6, or 7 nucléotides at position 3, 5, 7, 8, 9, 10, 11, 12, 14, 17, and/or 19 from the 5’ end of the first nucléotide sequence is a 2’-fluoro nucléotide. In some embodiments, the nucléotide at positions 3, 5, 7, 8, 9, 10, 11, 12, 14, 17, and/or 19 from the 5’ end of the first nucléotide sequence is a 2’-fluoro nucléotide. In some embodiments, at least two nucléotides at positions 3, 5, 7, 8, 9, 10, 11, 12, 14, 17, and/or 19 from the 5’ end of the first nucléotide sequence are 2’-fluoro nucléotides. In some embodiments, at least three nucléotides at positions 3, 5, 7, 8, 9, 10, 11, 12, 14, 17, and/or 19 from the 5’ end of the first nucléotide sequence are 2’-fluoro nucléotides. In some embodiments, the nucléotides at positions 3, 5, 7, 8, 9, 10, 11, 12, 14, 17, and/or 19 from the 5’ end of the first nucléotide sequence are 2’-fluoro nucléotides. In some embodiments, the nucléotide at position 3 from the 5’ end of the first nucléotide sequence is a 2’-fluoro nucléotide. In some embodiments, the nucléotide at position 5 from the 5’ end of the first nucléotide sequence is a 2’-fluoro nucléotide. In some embodiments, the nucléotide at position 7 from the 5’ end of the first nucléotide sequence is a 2’-fluoro nucléotide. In some embodiments, the nucléotide at position 8 from the 5’ end of the first nucléotide sequence is a 2’-fluoro nucléotide. In some embodiments, the nucléotide at position 9 from the 5’ end of the first nucléotide sequence is a 2’-fluoro nucléotide. In some embodiments, the nucléotide at position 10 from the 5’ end of the first nucléotide sequence is a 2’-fluoro nucléotide. In some embodiments, the nucléotide at position 11 from the 5’ end of the first nucléotide sequence is a 2’-fluoro nucléotide. In some embodiments, the nucléotide at position 12 from the 5’ end of the first nucléotide sequence is a 2’-fluoro nucléotide. In some embodiments, the nucléotide at position 14 from the 5’ end of the first nucléotide sequence is a 2’-fluoro nucléotide. In some embodiments, the nucléotide at position 17 from the 5’ end of the first nucléotide sequence is a 2’-fluoro nucléotide. In some embodiments, the nucléotide at position 19 from the 5’ end of the first nucléotide sequence is a 2’-fluoro nucléotide. In some embodiments, the nucléotide at position 3, 7, 8, 9, 12, and/or 17 from the 5’ end of the first nucléotide sequence is a 2’-fluoro nucléotide. In some embodiments, the nucléotide at position 3, 7, 8, and/or 17 from the 5’ end of the first nucléotide sequence is a 2’-fluoro nucléotide. In some embodiments, the nucléotide at position 3, 7, 8, 9, 12, and/or 17 from the 5’ end of the first nucléotide sequence is a 2’-fluoro nucléotide. In some embodiments, the nucléotide at position 5, 7, 8, and/or 9 from the 5’ end of the first nucléotide sequence is a 2’fluoro nucléotide. In some embodiments, the nucléotide at position 5, 9, 10, 11, 12, and/or 19 from the 5’ end of the first nucléotide sequence is a 2’-fluoro nucléotide. In some embodiments, the 2’-fluoro nucléotide is a 2’-fluoro nucléotide mimic.
In some embodiments, the 2’-fluoro nucléotide or 2’-O-methyl nucléotide is a 2’-fluoro or 2’-(9-methyl nucléotide mimic. In some embodiments, the 2’-fluoro or 2’-O-methyl nucléotide
Q2 R5 mimic is a nucléotide mimic of Formula (V): , wherein R1 is independently a nucleobase, aryl, heteroaryl, or H, Q1 and Q2 are independently S or O, R5 is independently OCD3, -F, or -OCH3, and R6 and R7 are independently H, D, or CD3. In some embodiments, the nucleobase is selected from cytosine, guanine, adenine, uracil, aryl, heteroaryl, and an analogue or dérivative thereof.
In some embodiments, the 2’-fluoro or 2’-O-methyl nucléotide mimic is a nucléotide mimic of Formula (16) - Formula (20):
Formula (18) Formula (19) Formula (20) wherein R1 is independently a nucleobase and R2 is F or -OCH3. In some embodiments, the nucleobase is selected from cytosine, guanine, adenine, uracil, aryl, heteroaryl, and an analogue or dérivative thereof.
In some embodiments, the first nucléotide sequence comprises, consists of, or consists essentially of ribonucleic acids (RNAs). In some embodiments, the first nucléotide sequence comprises, consists of, or consists essentially of modified RNAs. In some embodiments, the modified RNAs are selected from a 2’-(9-methyl RNA and 2’-fluoro RNA. In some embodiments, 15, 16, 17, 18, 19, 20, 21, 22, or 23 modified nucléotides of the first nucléotide sequence are independently selected from 2’-(2-methyl RNA and 2’-fluoro RNA.
In some embodiments, the sense strand may further comprise one or more intemucleoside linkages independently selected from a phosphodiester (PO) intemucleoside linkage, phosphorothioate (PS) intemucleoside linkage, phosphorodithioate intemucleoside linkage, and
PS-mimic intemucleoside linkage. In some embodiments, the PS-mimic intemucleoside linkage is a sulfo intemucleoside linkage.
In some embodiments, the sense strand may further comprise at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, or 15 or more phosphorothioate intemucleoside linkages. In some embodiments, the sense strand comprises 20, 19, 18, 17, 16, 15, 14, 13, 12, 11, 10, 9, 8, 7, 6, 5, 4, or 3 or fewer phosphorothioate intemucleoside linkages. In some embodiments, the sense strand comprises 2 to 10, 2 to 8, 2 to 6, 1 to 5, 1 to 4, 1 to 3, or 1 to 2 phosphorothioate intemucleoside linkages. In some embodiments, the sense strand comprises 1 to 2 phosphorothioate intemucleoside linkages. In some embodiments, the sense strand comprises 2 to 4 phosphorothioate intemucleoside linkages. In some embodiments, at least one phosphorothioate intemucleoside linkage is between the nucléotides at positions 1 and 2 from the 5’ end of the first nucléotide sequence. In some embodiments, at least one phosphorothioate intemucleoside linkage is between the nucléotides at positions 2 and 3 from the 5’ end of the first nucléotide sequence. In some embodiments, the sense strand comprises two phosphorothioate intemucleoside linkages between the nucléotides at positions 1 to 3 from the 5’ end of the first nucléotide sequence.
In some embodiments, any of the sense strands disclosed herein fiirther comprise a monomer selected from Examples 21-32, 36, 37, 40-42, and 44-46 monomers. In some embodiments, any of the sense strands disclosed herein fiirther comprise a 5’ end cap monomer. In some embodiments, the 5’ end cap monomer is selected from Examples 5-11, 33-35, 38, 39, 43, and 49-53 5’ end cap monomers. .
In some embodiments, any of the first nucléotide sequences disclosed herein fiirther comprise a monomer selected from Examples 21-32, 36, 37, 40-42, and 44-46 monomers. In some embodiments, any of the first nucléotide sequences disclosed herein fiirther comprise a 5’ end cap monomer. In some embodiments, the 5’ end cap monomer is selected from Examples 511, 33-35, 38, 39, 43, and 49-53 5’ end cap monomers. siNA antisense strand
Any of the siNA molécules described herein may comprise an antisense strand. The antisense strand may comprise a second nucléotide sequence. The second nucléotide sequence may be 15 to 30, 15 to 25, 15 to 23, 17 to 23, 19 to 23, or 19 to 21 nucléotides in length. In some embodiments, the second nucléotide sequence is 15, 16, 17, 18,19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30 nucléotides in length. In some embodiments, the second nucléotide sequence is at least 19 nucléotides in length. In some embodiments, the second nucléotide sequence is at least 21 nucléotides in length.
In some embodiments, the antisense strand is the same length as the second nucléotide sequence. In some embodiments, the antisense strand is longer than the second nucléotide sequence. In some embodiments, the antisense strand may further comprise 1, 2, 3, 4, or 5 or more nucléotides than the second nucléotide sequence. In some embodiments, the antisense strand is the same length as the sense strand. In some embodiments, the antisense strand is longer than the sense strand. In some embodiments, the antisense strand may further comprise 1, 2, 3, 4, or 5 or more nucléotides than the sense strand. In some embodiments, the antisense strand may further comprise a deoxyribonucleic acid (DNA). In some embodiments, the DNA is thymine (T). In some embodiments, the antisense strand may further comprise a TT sequence. In some embodiments, the antisense strand may further comprise one or more modified nucléotides that are adjacent to the second nucléotide sequence. In some embodiments, the one or more modified nucléotides are independently selected from any of the modified nucléotides disclosed herein (e.g., 2’-fluoro nucléotide, 2’-O-methyl nucléotide, 2’-fluoro nucléotide mimic, 2’-O-methyl nucléotide mimic, or a nucléotide comprising a modified nucleobase).
In some embodiments, the second nucléotide sequence comprises 15, 16, 17, 18, 19, 20, 21, 22, 23, or more modified nucléotides independently selected from a 2’-O-methyl nucléotide and a 2’-fluoro nucléotide. In some embodiments, 70%, 75%, 80%, 85%, 90%, 95% or 100% of the nucléotides in the second nucléotide sequence are modified nucléotides independently selected from a 2’-(9-methyl nucléotide and a 2’-fluoro nucléotide. In some embodiments, 100% of the nucléotides in the second nucléotide sequence are modified nucléotides independently selected from a 2’-O-methyl nucléotide and a 2’-fluoro nucléotide.
In some embodiments, between about 15 to 30, 15 to 25, 15 to 24, 15 to 23, 15 to 22, 15 to 21, 17 to 30, 17 to 25, 17 to 24, 17 to 23, 17 to 22, 17 to 21, 18 to 30, 18 to 25, 18 to 24, 18 to 23, 18 to 22, 18 to 21, 19 to 30, 19 to 25, 19 to 24, 19 to 23, 19 to 22, 19 to 21, 20 to 25, 20 to 24, 20 to 23, 21 to 25, 21 to 24, or 21 to 23 modified nucléotides of the second nucléotide sequence are 2’-(9-methyl nucléotides. In some embodiments, between about 2 to 20 modified nucléotides of the second nucléotide sequence are 2’-O-methyl nucléotides. In some embodiments, between about 5 to 25 modified nucléotides of the second nucléotide sequence are 2’-O-methyl nucléotides. In some embodiments, between about 10 to 25 modified nucléotides of the second nucléotide sequence are 2’-O-methyl nucléotides. In some embodiments, between about 12 to 25 modified nucléotides of the second nucléotide sequence are 2’-O-methyl nucléotides. In some embodiments, at least 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, or 22 modified nucléotides of the second nucléotide sequence are 2’-O-methyl nucléotides. In some embodiments, at least about 12 modified nucléotides of the second nucléotide sequence are 2’-O methyl nucléotides. In some embodiments, at least about 13 modified nucléotides of the second nucléotide sequence are 2’-O-methyl nucléotides. In some embodiments, at least about 14 modified nucléotides of the second nucléotide sequence are 2’-(9-methyl nucléotides. In some embodiments, at least about 15 modified nucléotides of the second nucléotide sequence are 2’-Omethyl nucléotides. In some embodiments, at least about 16 modified nucléotides of the second nucléotide sequence are 2’-O-methyl nucléotides. In some embodiments, at least about 17 modified nucléotides of the second nucléotide sequence are 2’-O-methyl nucléotides. In some embodiments, at least about 18 modified nucléotides of the second nucléotide sequence are 2’-Omethyl nucléotides. In some embodiments, at least about 19 modified nucléotides of the second nucléotide sequence are 2’-O-methyl nucléotides. In some embodiments, less than or equal to 25, 24, 23,22,21,20, 19, 18, 17, 16, 15, 14, 13, 12, 11, 10, 9, 8,7, 6, 5, 4, 3, or 2 modified nucléotides of the second nucléotide sequence are 2’-(9-methyl nucléotides. In some embodiments, less than or equal to 21 modified nucléotides of the second nucléotide sequence are 2’-O-methyl nucléotides. In some embodiments, less than or equal to 20 modified nucléotides of the second nucléotide sequence are 2’-(9-methyl nucléotides. In some embodiments, less than or equal to 19 modified nucléotides of the second nucléotide sequence are 2’-O-methyl nucléotides. In some embodiments, less than or equal to 18 modified nucléotides of the second nucléotide sequence are 2’-O-methyl nucléotides. In some embodiments, less than or equal to 17 modified nucléotides of the second nucléotide sequence are 2’-O-methyl nucléotides. In some embodiments, less than or equal to 16 modified nucléotides of the second nucléotide sequence are 2’-O-methyl nucléotides. In some embodiments, less than or equal to 15 modified nucléotides of the second nucléotide sequence are 2’-O-methyl nucléotides. In some embodiments, less than or equal to 14 modified nucléotides of the second nucléotide sequence are 2’-(9-methyl nucléotides. In some embodiments, less than or equal to 13 modified nucléotides of the second nucléotide sequence are 2’-O-methyl nucléotides. In some embodiments, at least one modified nucléotide of the second nucléotide sequence is a 2’-(9-methyl pyrimidine. In some embodiments, at least 5, 6, 7, 8, 9, or 10 modified nucléotides of the second nucléotide sequence are 2’-O-methyl pyrimidines. In some embodiments, at least one modified nucléotide of the second nucléotide sequence is a 2’-(2-methyl purine. In some embodiments, at least 5, 6, 7, 8, 9, or 10 modified nucléotides of the second nucléotide sequence are 2’-O-methyl purines. In some embodiments, the 2’-(9-methyl nucléotide is a 2’-(9-methyl nucléotide mimic.
In some embodiments, between 2 to 15 modified nucléotides of the second nucléotide sequence are 2’-fluoro nucléotides. In some embodiments, between 2 to 10 modified nucléotides of the second nucléotide sequence are 2’-fluoro nucléotides. In some embodiments, between 2 to 6 modified nucléotides of the second nucléotide sequence are 2’-fluoro nucléotides. In some embodiments, 1 to 6, 1 to 5, 1 to 4, or 1 to 3 modified nucléotides of the second nucléotide sequence are 2’-fluoro nucléotides. In some embodiments, at least 1, 2, 3, 4, 5, or 6 modified nucléotides of the second nucléotide sequence are 2’-fluoro nucléotides. In some embodiments, at least 1 modified nucléotide of the second nucléotide sequence is a 2’-fluoro nucléotide. In some embodiments, at least 2 modified nucléotides of the second nucléotide sequence are 2’fluoro nucléotides. In some embodiments, at least 3 modified nucléotides of the second nucléotide sequence are 2’-fluoro nucléotides. In some embodiments, at least 4 modified nucléotides of the second nucléotide sequence are 2’-fluoro nucléotides. In some embodiments, at least 5 modified nucléotides of the second nucléotide sequence are 2’-fluoro nucléotides. In some embodiments, 10, 9, 8, 7, 6, 5, 4, 3 or fewer modified nucléotides of the second nucléotide sequence are 2’-fluoro nucléotides. In some embodiments, 10 or fewer modified nucléotides of the second nucléotide sequence are 2’-fluoro nucléotides. In some embodiments, 7 or fewer modified nucléotides of the second nucléotide sequence are 2’-fluoro nucléotides. In some embodiments, 6 or fewer modified nucléotides of the second nucléotide sequence are 2’-fluoro nucléotides. In some embodiments, 5 or fewer modified nucléotides of the second nucléotide sequence are 2’-fluoro nucléotides. In some embodiments, 4 or fewer modified nucléotides of the second nucléotide sequence are 2’-fluoro nucléotides. In some embodiments, 3 or fewer modified nucléotides of the second nucléotide sequence are 2’-fluoro nucléotides. In some embodiments, 2 or fewer modified nucléotides of the second nucléotide sequence are 2’-fluoro nucléotides. In some embodiments, at least one modified nucléotide of the second nucléotide sequence is a 2’-fluoro pyrimidine. In some embodiments, 1, 2, 3, 4, 5, or 6 modified nucléotides of the second nucléotide sequence are 2’-fluoro pyrimidines. In some embodiments, at least one modified nucléotide of the second nucléotide sequence is a 2’-fluoro purine. In some embodiments, 1, 2, 3, 4, 5, or 6 modified nucléotides of the second nucléotide sequence are 2’fluoro purines. In some embodiments, the 2’-fluoro nucléotide is a 2’-fluoro nucléotide mimic.
In some embodiments, the 2’-fluoro nucléotide or 2’-(9-methyl nucléotide is a 2’-fluoro or 2’-O-methyl nucléotide mimic. In some embodiments, the 2’-fluoro or 2’-O-methyl nucléotide
Q2 R5 mimic is a nucléotide mimic of Formula (V): , wherein R1 is independently a nucleobase, aryl, heteroaryl, or H, Q1 and Q2 are independently S or O, R5 is independently
OCÜ3, -F, or -OCH3, and R6 and R7 are independently H, D, or CD3. In some embodiments, the nucleobase is selected from cytosine, guanine, adenine, uracil, aryl, heteroaryl, and an analogue or dérivative thereof.
In some embodiments, the 2’-fluoro or 2’-O-methyl nucléotide mimic is a nucléotide mimic of Formula (16) - Formula (20):
O'' R2 % SC R2 Cf' R2 οζ ôcd3 O' OCD % '
Formula (16) Formula (17) Formula (18) Formula (19) Formula (20)
wherein R1 is a nucleobase and R2 is independently F or -OCH3. In some embodiments, the nucleobase is selected from cytosine, guanine, adenine, uracil, aryl, heteroaryl, and an analogue or dérivative thereof.
In some embodiments, at least 1, 2, 3, 4, 5, 6, 7, 8, or 9 nucléotides at position 2, 5, 6, 8, 10, 14, 16, 17, and/or 18 from the 5’ end of the second nucléotide sequence is a 2’-fluoro nucléotide. In some embodiments, the nucléotide at position 2, 5, 6, 8, 10, 14, 16, 17, and/or 18 from the 5’ end of the second nucléotide sequence is a 2’-fluoro nucléotide. In some embodiments, at least two nucléotides at positions 2, 5, 6, 8, 10, 14, 16, 17, and/or 18 from the 5’ end of the second nucléotide sequence are 2’-fluoro nucléotides. In some embodiments, at least three nucléotides at positions 2, 5, 6, 8, 10, 14, 16, 17, and/or 18 from the 5’ end of the second nucléotide sequence are 2’-fluoro nucléotides. In some embodiments, at least four nucléotides at positions 2, 5, 6, 8, 10, 14, 16, 17, and/or 18 from the 5’ end of the second nucléotide sequence are 2’-fluoro nucléotides. In some embodiments, at least five nucléotides at positions 2, 5, 6, 8, 10, 14, 16, 17, and/or 18 from the 5’ end of the second nucléotide sequence are 2’-fluoro nucléotides. In some embodiments, the nucléotides at positions 2 and/or 14 from the 5’ end of the second nucléotide sequence are 2’-fluoro nucléotides. In some embodiments, the nucléotides at positions 2, 6, and/or 16 from the 5’ end of the second nucléotide sequence are 2’-fluoro nucléotides. In some embodiments, the nucléotides at positions 2, 6, 14, and/or 16 from the 5’ end of the second nucléotide sequence are 2’-fluoro nucléotides. In some embodiments, the nucléotides at positions 2, 6, 10, 14, and/or 18 from the 5’ end of the second nucléotide sequence are 2’-fluoro nucléotides. In some embodiments, the nucléotides at positions 2, 5, 8, 14, and/or 17 from the 5’ end of the second nucléotide sequence are 2’-fluoro nucléotides. In some embodiments, the nucléotide at position 2 from the 5’ end of the second nucléotide sequence is a 2’-fluoro nucléotide. In some embodiments, the nucléotide at position 5 from the 5’ end of the second nucléotide sequence is a 2’-fluoro nucléotide. In some embodiments, the nucléotide at position 6 from the 5’ end of the second nucléotide sequence is a 2’-fluoro nucléotide. In some embodiments, the nucléotide at position 8 from the 5’ end of the second nucléotide sequence is a 2’-fluoro nucléotide. In some embodiments, the nucléotide at position 10 from the 5’ end of the second nucléotide sequence is a 2’-fluoro nucléotide. In some embodiments, the nucléotide at position 14 from the 5’ end of the second nucléotide sequence is a 2’-fluoro nucléotide. In some embodiments, the nucléotide at position 16 from the 5’ end of the second nucléotide sequence is a 2’-fluoro nucléotide. In some embodiments, the nucléotide at position 17 from the 5’ end of the second nucléotide sequence is a 2’-fluoro nucléotide. In some embodiments, the nucléotide at position 18 from the 5’ end of the second nucléotide sequence is a 2’-fluoro nucléotide. In some embodiments, the 2’-fluoro nucléotide is a 2’-fluoro nucléotide mimic.
In some embodiments, the nucléotides in the second nucléotide sequence are arranged in an altemating 1:3 modification pattern, wherein 1 nucléotide is a 2’-fluoro nucléotide and 3 nucléotides are 2’-(9-methyl nucléotides, and wherein the altemating 1:3 modification pattern occurs at least 2 times. In some embodiments, the altemating 1:3 modification pattern occurs 2-5 times. In some embodiments, at least two of the altemating 1:3 modification pattern occur consecutively. In some embodiments, at least two of the altemating 1:3 modification pattern occurs nonconsecutively. In some embodiments, at least 1, 2, 3, 4, or 5 altemating 1:3 modification pattern begins at nucléotide position 2, 6, 10, 14, and/or 18 from the 5’ end of the antisense strand. In some embodiments, at least one altemating 1:3 modification pattern begins at nucléotide position 2 from the 5’ end of the antisense strand. In some embodiments, wherein at least one altemating 1:3 modification pattern begins at nucléotide position 6 from the 5’ end of the antisense strand. In some embodiments, at least one altemating 1:3 modification pattern begins at nucléotide position 10 from the 5’ end of the antisense strand. In some embodiments, at least one altemating 1:3 modification pattern begins at nucléotide position 14 from the 5’ end of the antisense strand. In some embodiments, at least one altemating 1:3 modification pattern begins at nucléotide position 18 from the 5’ end of the antisense strand. In some embodiments, the 2’-fluoro nucléotide is a 2’-fluoro nucléotide mimic.
In some embodiments, the nucléotides in the second nucléotide sequence are arranged in an altemating 1:2 modification pattern, wherein 1 nucléotide is a 2’-fluoro nucléotide and 2 nucléotides are 2’-O-methyl nucléotides, and wherein the altemating 1:2 modification pattern occurs at least 2 times. In some embodiments, the altemating 1:2 modification pattern occurs 2-5 times. In some embodiments, at least two of the altemating 1:2 modification pattern occurs consecutively. In some embodiments, at least two of the altemating 1:2 modification pattern occurs nonconsecutively. In some embodiments, at least 1, 2, 3, 4, or 5 altemating 1:2 modification pattern begins at nucléotide position 2, 5, 8, 14, and/or 17 from the 5’ end of the antisense strand. In some embodiments, at least one altemating 1:2 modification pattern begins at nucléotide position 2 from the 5’ end of the antisense strand. In some embodiments, at least one altemating 1:2 modification pattern begins at nucléotide position 5 from the 5’ end of the antisense strand. In some embodiments, at least one altemating 1:2 modification pattern begins at nucléotide position 8 from the 5’ end of the antisense strand. In some embodiments, at least one altemating 1:2 modification pattern begins at nucléotide position 14 from the 5’ end of the antisense strand. In some embodiments, at least one altemating 1:2 modification pattern begins at nucléotide position 17 from the 5’ end of the antisense strand. In some embodiments, the 2’fluoro nucléotide is a 2’-fluoro nucléotide mimic.
In some embodiments, the second nucléotide sequence comprises, consists of, or consists essentially of ribonucleic acids (RNAs). In some embodiments, the second nucléotide sequence comprises, consists of, or consists essentially of modified RNAs. In some embodiments, the modified RNAs are selected from a 2’-O-methyl RNA and 2’-fhioro RNA. In some embodiments, 15, 16, 17, 18, 19, 20, 21, 22, or 23 modified nucléotides of the second nucléotide sequence are independently selected from 2’-O-methyl RNA and 2’-fluoro RNA. In some embodiments, the 2’-fluoro nucléotide is a 2’-fluoro nucléotide mimic.
In some embodiments, the sense strand may further comprise one or more intemucleoside linkages independently selected from a phosphodiester (PO) intemucleoside linkage, phosphorothioate (PS) intemucleoside linkage, phosphorodithioate intemucleoside linkage, and PS-mimic intemucleoside linkage. In some embodiments, the PS-mimic intemucleoside linkage is a sulfo intemucleoside linkage.
In some embodiments, the antisense strand may further comprise at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, or 15 or more phosphorothioate intemucleoside linkages. In some embodiments, the antisense strand comprises 20, 19, 18, 17, 16, 15, 14, 13, 12, 11, 10, 9, 8, 7, 6, 5, 4, or 3 or fewer phosphorothioate intemucleoside linkages. In some embodiments, the antisense strand comprises 2 to 10, 2 to 8, 2 to 6, 1 to 5, 1 to 4, 1 to 3, or 1 to 2 phosphorothioate intemucleoside linkages. In some embodiments, the antisense strand comprises 2 to 10, 2 to 8, 2 to 6, 1 to 5, 1 to 4, 1 to 3, or 1 to 2 phosphorothioate intemucleoside linkages. In some embodiments, the antisense strand comprises 2 to 8 phosphorothioate intemucleoside linkages. In some embodiments, the antisense strand comprises 3 to 8 phosphorothioate intemucleoside linkages. In some embodiments, the antisense strand comprises 4 to 8 phosphorothioate intemucleoside linkages. In some embodiments, at least one phosphorothioate intemucleoside linkage is between the nucléotides at positions 1 and 2 from the 5’ end of the second nucléotide sequence. In some embodiments, at least one phosphorothioate intemucleoside linkage is between the nucléotides at positions 2 and 3 from the 5’ end of the second nucléotide sequence. In some embodiments, at least one phosphorothioate intemucleoside linkage is between the nucléotides at positions 1 and 2 from the 3 ’ end of the second nucléotide sequence. In some embodiments, at least one phosphorothioate intemucleoside linkage is between the nucléotides at positions 2 and 3 from the 3 ’ end of the second nucléotide sequence. In some embodiments, the antisense strand comprises two phosphorothioate intemucleoside linkages between the nucléotides at positions 1 to 3 from the 5’ end of the first nucléotide sequence. In some embodiments, the antisense strand comprises two phosphorothioate intemucleoside linkages between the nucléotides at positions 1 to 3 from the 3’ end of the first nucléotide sequence. In some embodiments, the antisense strand comprises (a) two phosphorothioate intemucleoside linkages between the nucléotides at positions 1 to 3 from the 5’ end of the first nucléotide sequence; and (b) two phosphorothioate intemucleoside linkages between the nucléotides at positions 1 to 3 from the 3 ’ end of the first nucléotide sequence.
In some embodiments, at least one end of the ds-siNA is a blunt end. In some embodiments, at least one end of the ds-siNA comprises an overhang, wherein the overhang comprises at least one nucléotide. In some embodiments, both ends of the ds-siNA comprise an overhang, wherein the overhang comprises at least one nucléotide. In some embodiments, the overhang comprises 1 to 5 nucléotides, 1 to 4 nucléotides, 1 to 3 nucléotides, or 1 to 2 nucléotides. In some embodiments, the overhang consists of 1 to 2 nucléotides.
In some embodiments, the first nucléotide sequence comprises a nucléotide sequence of any one SEQ ID NOs: 1-56, 103-158, and 205-260. In some embodiments, the second nucléotide sequence comprises a nucléotide sequence of any one of SEQ ID NOs: 57-102, 159-204, and 261-306. In some embodiments, the sense strand comprises a nucléotide sequence of any one of SEQ ID NOs: 307-362 and 415-444. In some embodiments, the antisense strand comprises a nucléotide sequence of any one of SEQ ID NOs: 363-409, 445-533, and 536-539.
In some embodiments, any of the antisense strands disclosed herein further comprise a monomer selected from Examples 21-32, 36, 37, 40-42, and 44-46 monomers. In some embodiments, any of the antisense strands disclosed herein further comprise a 5’ end cap monomer. In some embodiments, the 5’ end cap monomer is selected from Examples 5-11, 3335, 38, 39, 43, and 49-53 5’ end cap monomers.
In some embodiments, any of the secondnucleotide sequences disclosed herein further comprise a monomer selected from Examples 21-32, 36, 37, 40-42, and 44-46 monomers. In some embodiments, any of the second nucléotide sequences disclosed herein further comprise a
5’ end cap monomer. In some embodiments, the 5’ end cap monomer is selected from Examples 5-11, 33-35, 38, 39, 43, and 49-53 5’ end cap monomers.
Modified Nucléotides
Further disclosed herein are siNA molécules comprising one or more modified nucléotides. In some embodiments, any of the siNAs disclosed herein comprise one or more modified nucléotides. In some embodiments, any of the sense strands disclosed herein comprise one or more modified nucléotides. In some embodiments, any of the first nucléotide sequences disclosed herein comprise one or more modified nucléotides. In some embodiments, any of the antisense strands disclosed herein comprise one or more modified nucléotides. In some embodiments, any of the second nucléotide sequences disclosed herein comprise one or more modified nucléotides. In some embodiments, the one or more modified nucléotides is adjacent to the first nucléotide sequence. In some embodiments, at least one modified nucléotide is adjacent to the 5’ end of the first nucléotide sequence. In some embodiments, at least one modified nucléotide is adjacent to the 3’ end of the first nucléotide sequence. In some embodiments, at least one modified nucléotide is adjacent to the 5’ end of the first nucléotide sequence and at least one modified nucléotide is adjacent to the 3’ end of the first nucléotide sequence. In some embodiments, the one or more modified nucléotides is adjacent to the second nucléotide sequence. In some embodiments, at least one modified nucléotide is adjacent to the 5’ end of the second nucléotide sequence. In some embodiments, at least one modified nucléotide is adjacent to the 3 ’ end of the second nucléotide sequence. In some embodiments, at least one modified nucléotide is adjacent to the 5’ end of the second nucléotide sequence and at least one modified nucléotide is adjacent to the 3’ end of the second nucléotide sequence. In some embodiments, a 2’-(9-methyl nucléotide in any of sense strands or first nucléotide sequences disclosed herein is replaced with a modified nucléotide. In some embodiments, a 2’-O-methyl nucléotide in any of antisense strands or second nucléotide sequences disclosed herein is replaced with a modified nucléotide.
In some embodiments, any of the siNA molécules, siNAs, sense strands, first nucléotide sequences, antisense strands, and second nucléotide sequences disclosed herein comprise 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20,21,22, 23,24, 25, 26, 27,28,29, or 30 or more modified nucléotides. In some embodiments, 1%, 2%, 3%, 4%, 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 70%, 75%, 80%, 85%, 86%, 87%, 88%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% of the nucléotides in the siNA molécule, siNA, sense strand, first nucléotide sequence, antisense strand, or second nucléotide sequence are modified nucléotides.
In some embodiments, a modified nucléotide is selected from the group consisting of 2’fluoro nucléotide, 2’-O-methyl nucléotide, 2’-fluoro nucléotide mimic, 2’-(9-methyl nucléotide mimic, a locked nucleic acid, and a nucléotide comprising a modified nucleobase.
In some embodiments, any of the siRNAs disclosed herein comprise at least 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 or more 2’-fluoro or 2’-O-methyl nucléotide mimics. In some embodiments, any of the sense strands disclosed herein comprise at least 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 or more 2’fluoro or 2’-O-methyl nucléotide mimics. In some embodiments, any of the first nucléotide sequences disclosed herein comprise at least 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 or more 2’-fluoro or 2’O-methyl nucléotide mimics. In some embodiments, any of the antisense strand disclosed herein comprise at least 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 or more 2’-fluoro or 2’-O-methyl nucléotide mimics. In some embodiments, any of the second nucléotide sequences disclosed herein comprise at least 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 or more 2’-fhioro or 2’-O-methyl nucléotide mimics. In some embodiments, the 2’-fluoro or 2’-O-methyl nucléotide mimic is a nucléotide mimic of Formula (16) - Formula (20):
Formula (16)
Formula (17) Formula (18) Formula (19)
Formula (20) wherein R1 is a nucleobase and R2 is independently F or -OCH3. In some embodiments, the nucleobase is selected from cytosine, guanine, adenine, uracil, aryl, heteroaryl, and an analogue or dérivative thereof. In some embodiments, the siNA molécules disclosed herein comprise at least one 2’-fluoro nucléotide, at least one 2’-O-methyl nucléotide, and at least one 2’-fluoro or 2’-O-methyl nucléotide mimic. In some embodiments, the at least one 2’-fluoro or 2’-O-methyl nucléotide mimic is adjacent to the first nucléotide sequence. In some embodiments, the at least one 2’-fluoro or 2’-O-methyl nucléotide mimic is adjacent to the 5’ end of first nucléotide sequence. In some embodiments, the at least one 2’-fluoro or 2’-O-methyl nucléotide mimic is adjacent to the 3’ end of first nucléotide sequence. In some embodiments, the at least one 2’fluoro or 2’-(9-methyl nucléotide mimic is adjacent to the second nucléotide sequence. In some embodiments, the at least one 2’-fluoro or 2’-(9-methyl nucléotide mimic is adjacent to the 5’ end of second nucléotide sequence. In some embodiments, the at least one 2’-fluoro or 2’-Omethyl nucléotide mimic is adjacent to the 3’ end of second nucléotide sequence. In some embodiments, the first nucléotide sequence does not comprise a 2’-fluoro nucléotide mimic. In some embodiments, the first nucléotide sequence does not comprise a 2’-O-methyl nucléotide mimic. In some embodiments, the second nucléotide sequence does not comprise a 2’-fluoro nucléotide mimic. In some embodiments, the second nucléotide sequence does not comprise a
2’-O-methyl nucléotide mimic.
In some embodiments, any of the siRNAs disclosed herein comprise at least 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 or more locked nucleic acids. In some embodiments, any of the sense strands disclosed herein comprise at least 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 or more locked nucleic acids. In some embodiments, any of the first nucléotide sequences disclosed herein comprise at least 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 or more locked nucleic acids. In some embodiments, any of the antisense strand disclosed herein comprise at least 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 or more locked nucleic acids. In some embodiments, any of the second nucléotide sequences disclosed herein comprise at least 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 or more locked nucleic acids. In some embodiments, the B °^\ n /B
Voy y°y o^o locked nucleic acid is selected from * (LNA), (ScpBNA or “cp”);
(AmNA), where R is H or alkyl (or AmNA(N-Me)) when R is alkyl);
(GuNA); and GuNA(N-R), r - Me, Et, iPr, tBu, wherein B is a nucleobase. In some embodiments, any of the siRNAs, sense strands, first nucléotide sequences, antisense strands, or second nucléotide sequences disclosed herein comprise at least modified
nucléotide that is (LNA). In some embodiments, any of the siRNAs, sense strands, first nucléotide sequences, antisense strands, or second nucléotide sequences disclosed herein comprise at least modified nucléotide that is
(ScpBNA or “cp”). In some embodiments, any of the siRNAs, sense strands, first nucléotide sequences, antisense strands, or second nucléotide sequences disclosed herein comprise at least modified nucléotide that is
B
Voy
-o^-n,r 0 (AmNA), where R is H or alkyl (or AmNA(N-Me)) when R is alkyl). In some embodiments, any of the siRNAs, sense strands, first nucléotide sequences, antisense strands, or second nucléotide sequences disclosed herein comprise at least modified nucléotide that is
h2n (GuNA). In some embodiments, any of the siRNAs, sense strands, first nucléotide sequences, antisense strands, or second nucléotide sequences disclosed herein
comprise at least modified nucléotide that is GuNA(N-R), r Me, Et, iPr, tBu, wherein B is a nucleobase.
Phosphorylation blocker
Further disclosed herein are siNA molécules comprising a phosphorylation blocker. In some embodiments, a 2’-(9-methyl nucléotide in any of sense strands or first nucléotide sequences disclosed herein is replaced with a nucléotide containing a phosphorylation blocker. In some embodiments, a 2’-O-methyl nucléotide in any of antisense strands or second nucléotide sequences disclosed herein is replaced with a nucléotide containing a phosphorylation blocker. In some embodiments, a 2’-(9-methyl nucléotide in any of sense strands or first nucléotide sequences disclosed herein is further modified to contain a phosphorylation blocker. In some embodiments, a 2’-O-methyl nucléotide in any of antisense strands or second nucléotide sequences disclosed herein is further modified to contain a phosphorylation blocker.
In some embodiments, any of the siNA molécules disclosed herein comprise a R4yyR1 phosphorylation blocker of Formula (IV): , wherein R1 is a nucleobase, R4 is -OR30 or -NR31R32, R30 is Ci-Cs substituted or unsubstituted alkyl; and R31 and R32 together with the nitrogen to which they are attached form a substituted or unsubstituted heterocyclic ring.
In some embodiments, any of the siNA molécules disclosed herein comprise a
phosphorylation blocker of Formula (IV): —L Formula (IV), wherein R1 is a nucleobase, and R4 is -OCH3 or -N(CH2CH2)2O.
In some embodiments, a siNA molécule comprises (a) a phosphorylation blocker of R4\°/R1
Formula (IV): —J— , wherein R1 is a nucleobase, R4 is -O-R30 or-NR31R32, R30 is Ci-Cs substituted or unsubstituted alkyl; and R31 and R32 together with the nitrogen to which they are attached form a substituted or unsubstituted heterocyclic ring; and (b) a short interfering nucleic acid (siNA), wherein the phosphorylation blocker is conjugated to the siNA.
In some embodiments, a siNA molécule comprises (a) a phosphorylation blocker of R4A^°yR1 '^O^'O
Formula (IV): Formula (IV), wherein R1 is a nucleobase, and R4 is -OCH3 or N(CH2CH2)2O; and (b) a short interfering nucleic acid (siNA), wherein the phosphorylation blocker is conjugated to the siNA.
In some embodiments, the phosphorylation blocker is attached to the 3’ end of the sense strand or first nucléotide sequence. In some embodiments, the phosphorylation blocker is attached to the 3’ end of the sense strand or first nucléotide sequence via 1, 2, 3, 4, or 5 or more linkers. In some embodiments, the phosphorylation blocker is attached to the 5’ end of the sense strand or first nucléotide sequence. In some embodiments, the phosphorylation blocker is attached to the 5’ end of the sense strand or first nucléotide sequence via 1, 2, 3, 4, or 5 or more linkers. In some embodiments, the phosphorylation blocker is attached to the 3’ end of the antisense strand or second nucléotide sequence. In some embodiments, the phosphorylation blocker is attached to the 3’ end of the antisense strand or second nucléotide sequence via 1, 2, 3, 4, or 5 or more linkers. In some embodiments, the phosphorylation blocker is attached to the 5’ end of the antisense strand or second nucléotide sequence. In some embodiments, the phosphorylation blocker is attached to the 5’ end of the antisense strand or second nucléotide sequence via 1, 2, 3, 4, or 5 or more linkers. In some embodiments, the one or more linkers are independently selected from the group consisting of a phosphodiester linker, phosphorothioate linker, and phosphorodithioate linker.
Conjugated Moiety
Further disclosed herein are siNA molécules comprising a conjugated moiety. In some embodiments, the conjugated moiety is selected from galactosamine, peptides, proteins, sterols, lipids, phospholipids, biotin, phenoxazines, active diug substance, cholestérols, phenanthridine, anthraquinone, acridine, fluoresceins, rhodamines, coumarins, and dyes. In some embodiments, the conjugated moiety is attached to the 3’ end of the sense strand or first nucléotide sequence. In some embodiments, the conjugated moiety is attached to the 3’ end of the sense strand or first nucléotide sequence via 1, 2, 3, 4, or 5 or more linkers. In some embodiments, the conjugated moiety is attached to the 5’ end of the sense strand or first nucléotide sequence. In some embodiments, the conjugated moiety is attached to the 5’ end of the sense strand or first nucléotide sequence via 1, 2, 3, 4, or 5 or more linkers. In some embodiments, the conjugated moiety is attached to the 3 ’ end of the antisense strand or second nucléotide sequence. In some embodiments, the conjugated moiety is attached to the 3’ end of the antisense strand or second nucléotide sequence via 1, 2, 3, 4, or 5 or more linkers. In some embodiments, the conjugated moiety is attached to the 5’ end of the antisense strand or second nucléotide sequence. In some embodiments, the conjugated moiety is attached to the 5’ end of the antisense strand or second nucléotide sequence via 1, 2, 3, 4, or 5 or more linkers. In some embodiments, the one or more linkers are independently selected from the group consisting of a phosphodiester linker, phosphorothioate linker, and phosphorodithioate linker.
In some embodiments, the conjugated moiety is galactosamine. In some embodiments, any of the siNAs disclosed herein are attached to a conjugated moiety that is galactosamine. In some embodiments, the galactosamine is N-acetylgalactosamine (GalNAc). In some embodiments, any of the siNA molécules disclosed herein comprise GalNAc. In some embodiments, the GalNAc is of Formula (VI):
is independently 1 or 2; p is 0 or 1 ; each R is independently H or a first protecting group; each Y is independently selected from -O-P(=O)(SH) -, -O-P(=O)(O) -, -O-P(=O)(OH) -, -O-P(S)S-, and -O-; Z is H or a second protecting group; either L is a linker or L and Y in combination are a linker; and A is H, OH, a third protecting group, an activated group, or an oligonucleotide. In some embodiments, the first protecting group is acetyl. In some embodiments, the second protecting group is trimethoxytrityl (TMT). In some embodiments, the activated group is a phosphoramidite group. In some embodiments, the phosphoramidite group is a cyanoethoxy V A-diisopropylphosphoramidite group. In some embodiments, the linker is a C6-NH2 group. In some embodiments, A is a short interfering nucleic acid (siNA) or siNA molécule. In some embodiments, m is 3. In some embodiments, R is H, Z is H, and n is 1. In some embodiments, R is H, Z is H, and n is 2.
In some embodiments, the GalNAc is of Formula (VII):
R = OH or SH wherein each n is independently 1 or 2.
In some embodiments, the galactosamine is attached to the 3 ’ end of the sense strand or first nucléotide sequence. In some embodiments, the galactosamine is attached to the 3 ’ end of the sense strand or first nucléotide sequence via 1, 2, 3, 4, or 5 or more linkers. In some embodiments, the galactosamine is attached to the 5’ end of the sense strand or first nucléotide sequence. In some embodiments, the galactosamine is attached to the 5’ end of the sense strand or first nucléotide sequence via 1, 2, 3, 4, or 5 or more linkers. In some embodiments, the galactosamine is attached to the 3 ’ end of the antisense strand or second nucléotide sequence. In some embodiments, the galactosamine is attached to the 3 ’ end of the antisense strand or second nucléotide sequence via 1, 2, 3, 4, or 5 or more linkers. In some embodiments, the galactosamine is attached to the 5’ end of the antisense strand or second nucléotide sequence. In some embodiments, the galactosamine is attached to the 5’ end of the antisense strand or second nucléotide sequence via 1, 2, 3, 4, or 5 or more linkers. In some embodiments, the one or more linkers are independently selected from the group consisting of a phosphodiester (p or po) linker, phosphorothioate (ps) linker, phosphoramidite (HEG) linker, triethylene glycol (TEG) linker, and/or phosphorodithioate linker. In some embodiments, the one or more linkers are independently selected from the group consisting of p-(PS)2, (PS)2-p-TEG-p, (PS)2-p-HEG-p, and (PS)2-p-(HEG-p)2.
In some embodiments, the conjugated moiety is a lipid moiety. In some embodiments, any of the siNAs disclosed herein are attached to a conjugated moiety that is a lipid moiety. Examples of lipid moieties include, but are not limited to, a cholestérol moiety, a thioether, e.g., hexyl-S-tritylthiol, a thiocholestérol, an aliphatic chain, e.g., dodecandiol or undecyl residues a phospholipid, e.g., di-hexadecyl-rac-glycerol or triethylammonium 1-di-O-hexadecyl-racglycero-S-H-phosphonate, a polyamine or a polyethylene glycol chain, adamantane acetic acid, a palmityl moiety, or an octadecylamine or hexylamino-carbonyl-oxycholesterol moiety.
In some embodiments, the conjugated moiety is an active drug substance. In some embodiments, any of the siNAs disclosed herein are attached to a conjugated moiety that is an active drug substance. Examples of active drug substances include, but are not limited to, aspirin, warfarin, phenylbutazone, ibuprofen, suprofen, fenbufen, ketoprofen, (5)-(+)- pranoprofen, carprofen, dansylsarcosine, 2,3,5-triiodobenzoic acid, flufenamic acid, folinic acid, a benzothiadiazide, chlorothiazide, a diazepine, indomethicin, a barbiturate, a cephalosporin, a sulfa drug, an antidiabetic, an antibacterial or an antibiotic.
5’-Stabilized End Cap
Further disclosed herein are siNA molécules comprising a 5’-stabilized end cap. As used herein the ternis “5’-stabilized end cap” and “5’ end cap” are used interchangeably. In some embodiments, a 2’-O-methyl nucléotide in any of sense strands or first nucléotide sequences disclosed herein is replaced with a nucléotide containing a 5’-stabilized end cap. In some embodiments, a 2’-O-methyl nucléotide in any of antisense strands or second nucléotide sequences disclosed herein is replaced with a nucléotide containing a 5’-stabilized end cap. In some embodiments, a 2’-(9-methyl nucléotide in any of sense strands or first nucléotide sequences disclosed herein is further modified to contain a 5’-stabilized end cap. In some embodiments, a 2’-(9-methyl nucléotide in any of antisense strands or second nucléotide sequences disclosed herein is further modified to contain a 5’-stabilized end cap.
In some embodiments, the 5’-stabilized end cap is a 5’ phosphate mimic. In some embodiments, the 5’-stabilized end cap is a modified 5’ phosphate mimic. In some embodiments, the modified 5’ phosphate is a chemically modified 5’ phosphate. In some embodiments, the 5’stabilized end cap is a 5’-vinyl phosphonate. In some embodiments, the 5’-vinyl phosphonate is a 5’-(E)-vinyl phosphonate or 5’-(Z)-vinyl phosphonate. In some embodiments, the 5’-vinyl phosphonate is a deuterated vinyl phosphonate. In some embodiments, the deuterated vinyl phosphonate is a mono-deuterated vinyl phosphonate. In some embodiments, the deuterated vinyl phosphonate is a di-deuterated vinyl phosphonate. In some embodiments, the 5’-stabilized end cap is a phosphate mimic. Examples of phosphate mimics are disclosed in Parmar et al.,
2018, J Med Chem, 61(3):734-744, International Publication Nos. WO2018/045317 and
WO2018/044350, and U.S. Patent No. 10,087,210, each of which is incorporated by reference in its entirety.
In some embodiments, any of the siNA molécules, sense strands, first nucléotide sequences, antisense strands, or second nucléotide sequences disclosed herein comprise a 5’-
O (DCH3 , wherein R1 is H, a nucleobase, aryl, or stabilized end cap of Formula (la):
zocd3
ο , -CH CD-Z, -CD=CH-Z, -CD=CD-Z, (CR21R22)n-Z, or -(C2-C6 alkenylene)-Z and R20 is H; or R2 and R20 together form a 3- to 7membered carbocyclic ring substituted with -(CR21R22)n-Z or -(C2-C6 alkenylene)-Z; n is 1, 2, 3, or 4; Z is -ONR23R24, -OP(O)OH(CH2)mCO2R23, -OP(S)OH(CH2)mCO2R23, -P(O)(OH)2, P(O)(OH)(OCH3), -P(O)(OH)(OCD3), -SO2(CH2)mP(O)(OH)2, -so2nr23r25, -nr23r24, NR23SÛ2R24; either R21 and R22 are independently hydrogen or Ci-Cô alkyl, or R21 and R22 together form an oxo group; R23 is hydrogen or Ci-Cô alkyl; R24 is -SO2R25 or -C(O)R25; or R23 and R24 together with the nitrogen to which they are attached form a substituted or unsubstituted heterocyclic ring; R25 is Ci-Cg alkyl; and m is 1, 2, 3, or 4. In some embodiments, R1 is an aryl.
In some embodiments, the aryl is a phenyl.
In some embodiments, any of the siNA molécules, sense strands, first nucléotide sequences, antisense strands, or second nucléotide sequences disclosed herein comprise a 5’-
0' (DCD3 stabilized end cap of Formula (Ib): Ά , wherein R1 is H, a nucleobase, aryl, or heteroaryl; R2 is
O O
Ο
II
Ρχ-ΟΗ
OH
HO, ,S 9 O,, ZOH
Y'YCP'X~/OH Y^^^OH 9
O, OCH
zOCD ΌΗ
-CH=CD-Z, -CD=CH-Z, -CD=CD-Z, (CR2IR22)n-Z, or -(C2-C6 alkenylene)-Z and R20 is H; or R2 and R20 together form a 3- to 7membered carbocyclic ring substituted with -(CR21R22)n-Z or -(C2-Cô alkenylene)-Z; n is 1, 2, 3, or 4; Z is -ONR23R24, -OP(O)OH(CH2)mCO2R23, -OP(S)OH(CH2)mCO2R23, -P(O)(OH)2, P(O)(OH)(OCH3), -P(O)(OH)(OCD3), -SO2(CH2)mP(O)(OH)2, -SO2NR23R25, -NR23R24, NR23SO2R24; either R21 and R22 are independently hydrogen or Ci-Cô alkyl, or R21 and R22 together form an oxo group; R23 is hydrogen or Ci-Cô alkyl; R24 is -SO2R25 or -C(O)R25; or R23 and R24 together with the nitrogen to which they are attached form a substituted or unsubstituted heterocyclic ring; R25 is Ci-Cô alkyl; and m is 1, 2, 3, or 4. In some embodiments, R1 is an aryl.
In some embodiments, the aryl is a phenyl.
In some embodiments, any of the siNA molécules, sense strands, first nucléotide sequences, antisense strands, or second nucléotide sequences disclosed herein comprise a 5’stabilized end cap of Formula (le):
, wherein R1 is a nucleobase, aryl, heteroaryl, or H,
O
II
PX-OH
OH
HO, Z/S 9 O,, ZOH
ΥΥο'^^ΌΗ Ύ^Ρ^ΟΗ
O, och3 o, ocd3
Y OH Y OH 5
, -CH=CD-Z, -CD=CH-Z, -CD=CD-Z, -(CR2 ‘R22)n-Z, or -(C2-C6 alkenylene)-Z and R20 is hydrogen; or R2 and R20 together form a 3- to 7-membered carbocyclic ring substituted with -(CR21R22)n-Z or-(C2-Cô alkenylene)-Z; n is 1, 2, 3, or 4;
Z is -ONR23R24, -OP(O)OH(CH2)mCO2R23, -OP(S)OH(CH2)raCO2R23, -P(O)(OH)2, P(O)(OH)(OCH3), -P(O)(OH)(OCD3), -SO2(CH2)mP(O)(OH)2, -SO2NR23R25, -NR23R24, orNR23SO2R24; R21 and R22 either are independently hydrogen or Ci-Cô alkyl, or R21 and R22 together form an oxo group; R23 is hydrogen or Ci-Cô alkyl; R24 is -SO2R25 or -C(O)R25; or
R23 and R24 together with the nitrogen to which they are attached form a substituted or unsubstituted heterocyclic ring; R25 is Ci-Cô alkyl; and m is 1, 2, 3, or 4. In some embodiments, R1 is an aryl. In some embodiments, the aryl is a phenyl.
In some embodiments, any of the siNA molécules, sense strands, first nucléotide 5 sequences, antisense strands, or second nucléotide sequences disclosed herein comprise a 5’Rv°v-R’
O t>CH3 stabilized end cap of Formula (Ha): , wherein R1 is a nucleobase, aryl, heteroaryl,
COCH3, - - is a double or single bond, R10 = -CH2PO3H or -NHCH3, R11 is -CH2- or -CO-, 10 and R12 is H and R13 is CH3 or R12 and R13 together form -CH2CH2CH2-. In some embodiments, R1 is an aryl. In some embodiments, the aryl is a phenyl.
In some embodiments, any of the siNA molécules, sense strands, first nucléotide sequences, antisense strands, or second nucléotide sequences disclosed herein comprise a 5’-
0' OCD3 stabilized end cap of Formula (Ilb): -~L~ , wherein R1 is a nucleobase, aryl, heteroaryl,
HQ R Æ 'S=O ° N-R11 0 , -CH2SO2NHCH3, or R12^, R9 is -SO2CH3 orCOCH3, - - - is a double or single bond, R10 = -CH2PO3H or -NHCH3, R11 is -CH2- or -CO-, and R12 is H and R13 is CH3 or R12 and R13 together form -CH2CH2CH2- In some embodiments, R1 is an aryl. In some embodiments, the aryl is a phenyl.
In some embodiments, any of the siNA molécules, sense strands, first nucléotide sequences, antisense strands, or second nucléotide sequences disclosed herein comprise a 5’a/Lx^°^R1 θ' 'OCH3 stabilized end cap of Formula (III): ~-L~ , wherein R1 is a nucleobase, aryl, heteroaryl, or H, L is -CH2-, -CH=CH-, -CO-, or -CH2CH2-, and A is -ONHCOCH3, -ONHSO2CH3, PO3H, -OP(SOH)CH2CO2H, -SO2CH2PO3H, -SO2NHCH3, -NHSO2CH3, or N(SO2CH2CH2CH2). In some embodiments, R1 is an aryl. In some embodiments, the aryl is a phenyl.
In some embodiments, any of the siNA molécules, sense strands, first nucléotide sequences, antisense strands, or second nucléotide sequences disclosed herein comprise a 5’stabilized end cap selected from Examples 5-11, 33-35, 38, 39, 43, and 49-53 5’ end cap monomers.
Further disclosed herein are siNA molécules comprising (a) a 5’-stabilized end cap of
0' 'OCH3 , wherein R1 is a nucleobase, aryl, heteroaryl, or H; R2 is
Formula (la):
zP 9 HO. ZS 9 Ox OH O, OCH3 O, OCD3 ^JS^PX-OH A s/<X X 5/^jX l OH Y O OH X OH Y OH V OH
5 5 5
, -CH=CD-Z, -CD=CH-Z, -CD=CD-Z, -(CR21R22)n-Z, or-(C2-C6 alkenylene)-Z and R20 is H; or R2 and R20 together form a 3- to 7-membered carbocyclic ring substituted with -(CR21R22)n-Z or - (C2-Cô alkenylene)-Z; n is 1, 2, 3, or 4; Z is -ONR23R24, OP(O)OH(CH2)mCO2R23, -OP(S)OH(CH2)mCO2R23, -P(O)(OH)2, -P(O)(OH)(OCH3), P(O)(OH)(OCD3), -SO2(CH2)mP(O)(OH)2, -SO2NR23R25, -NR23R24, -NR23SO2R24; either R21 and R22 are independently hydrogen or Ci-Ce alkyl, or R21 and R22 together form an oxo group; R23 is hydrogen or Ci-Cô alkyl; R24 is -SO2R25 or -C(O)R25; or R23 and R24 together with the nitrogen to which they are attached form a substituted or unsubstituted heterocyclic ring; R25 is
Ci-Cô alkyl; and m is 1, 2, 3, or 4; and (b) a short interfering nucleic acid (siNA), wherein the 5’ stabilized end cap is conjugated to the siNA. In some embodiments, R1 is an aryl. In some embodiments, the aryl is a phenyl.
Further disclosed herein are siNA molécules comprising (a) a 5’-stabilized end cap of
0' ocd3
Ύ , wherein R1 is a nucleobase, aryl, heteroaryl, or H; R2 is
Formula (Ib):
O , -CH=CD-Z, -CD=CH-Z, -CD=CD-Z, -(CR21R22)n-Z, or -(C2-C6 alkenylene)-Z and R20 is H; or R2 and R20 together form a 3- to 7-membered carbocyclic ring substituted with -(CR21R22)n-Z or - (C2-Cô alkenylene)-Z; n is 1, 2, 3, or 4; Z is -ONR23R24, OP(O)OH(CH2)mCO2R23, -OP(S)OH(CH2)mCO2R23, -P(O)(OH)2, -P(O)(OH)(OCH3), P(O)(OH)(OCD3), -SO2(CH2)mP(O)(OH)2, -SO2NR23R25, -NR23R24, -NR23SO2R24; either R21 and R22 are independently hydrogen or Ci-Cô alkyl, or R21 and R22 together form an oxo group; R23 is hydrogen or Ci-Cô alkyl; R24 is -SO2R25 or -C(O)R25; or R23 and R24 together with the nitrogen to which they are attached form a substituted or unsubstituted heterocyclic ring; R25 is
Ci-Cô alkyl; and m is 1, 2, 3, or 4; and (b) a short interfering nucleic acid (siNA), wherein the 5’ stabilized end cap is conjugated to the siNA. In some embodiments, R1 is an aryl. In some embodiments, the aryl is a phenyl.
Further disclosed herein are siNA molécules comprising (a) a 5’-stabilized end cap of Ry°yR1
R20' VJ
0' F
Formula (le): ~~L- , wherein R1 is a nucleobase, aryl, heteroaryl, or H, R2 is
H ,Ν./ । S // Ά
O O ο , -CH=CD-Z, -CD=CH-Z, -CD=CD-Z, -(CR21R22)n-Z, or -(C2-C6 alkenylene)-Z and R20 is hydrogen; or R2 and R20 together form a 3- to 7-membered carbocyclic ring substituted with -(CR21R22)n-Z or -(C2-Cô alkenylene)-Z; n is 1, 2, 3, or 4;
Z is -ONR23R24, -OP(O)OH(CH2)mCO2R23, -OP(S)OH(CH2)mCO2R23, -P(O)(OH)2, P(O)(OH)(OCH3), -P(O)(OH)(OCD3), -SO2(CH2)mP(O)(OH)2, -SO2NR23R25, -NR23R24, orNR23SO2R24; R21 and R22 either are independently hydrogen or Ci-Cô alkyl, or R21 and R22 together form an oxo group; R23 is hydrogen or Ci-Cô alkyl; R24 is -SO2R25 or -C(O)R25; or R23 and R24 together with the nitrogen to which they are attached form a substituted or unsubstituted heterocyclic ring; R25 is Ci-Cô alkyl; and m is 1, 2, 3, or 4; and (b) a short interfering nucleic acid (siNA), wherein the 5’-stabilized end cap is conjugated to the siNA. In some embodiments, R1 is an aryl. In some embodiments, the aryl is a phenyl.
In some embodiments, a siNA molécule comprises (a) a 5’-stabilized end cap of Formula
R13 P R'S>0 'n-r11
CH2SO2NHCH3, or R12 , R9 is -SO2CH3 or -COCH3, wherein is a double or single bond, R10 = -CH2PO3H or -NHCH3, R11 is -CH2- or -CO-, and R12 is H and R13 is CH3 or R12 and R13 together form -CH2CH2CH2-; and (b) a short interfering nucleic acid (siNA), wherein the 5’-stabilized end cap is conjugated to the siNA. In some embodiments, R1 is an aryl. In some embodiments, the aryl is a phenyl.
In some embodiments, a siNA molécule comprises (a) a 5’-stabilized end cap of Formula
0' OCD3 hn.o (Ilb): —J— , wherein R1 is a nucleobase, aryl, heteroaryl, or H, R2 is
100
R13/9 R's=o 'n-r11
CH2SO2NHCH3, or R12 , R9 is -SO2CH3 or -COCH3, wherein is a double or single bond, R10 = -CH2PO3H or -NHCH3, R11 is -CH2- or -CO-, and R12 is H and R13 is CH3 or R12 and R13 together form -CH2CH2CH2-; and (b) a short interfering nucleic acid (siNA), wherein the 5’-stabilized end cap is conjugated to the siNA. In some embodiments, R1 is an aryl. In some 5 embodiments, the aryl is a phenyl.
In some embodiments, a siNA molécule comprises (a) a 5’-stabilized end cap of Formula a/Lx^°\^R1 ô' OCH3 (III): , wherein R1 is a nucleobase, aryl, heteroaryl, or H, L is -CH2-, -CH=CH-,
-CO-, or -CH2CH2-, and A is -ONHCOCH3, -ONHSO2CH3, -PO3H, -OP(SOH)CH2CO2H, SO2CH2PO3H, -SO2NHCH3, -NHSO2CH3, or-N(SO2CH2CH2CH2); and (b) a short interfering 10 nucleic acid (siNA), wherein the 5’-stabilized end cap is conjugated to the siNA. In some embodiments, R1 is an aryl. In some embodiments, the aryl is phenyl.
In some embodiments, any of the siNA molécules disclosed herein comprise a 5’stabilized end cap selected from the group consisting of Formula (1) to Formula (15), Formula (9X) to Formula (12X), and Formula (9Y) to Formula (12Y):
Formula (1) Formula (2) Formula (3) Formula (4)
Formula (5) Formula (6) Formula (7)
Formula (8)
Formula (9)
Formula (9X)
Formula (9Y)
101
Formula (11X) Formula (11Y)
Formula (11)
Formula (12) Formula (12X) Formula (12Y)
Formula (13) Formula (14)
Formula (15) , wherein R1 is a nucleobase, aryl, heteroaryl, or H. In some embodiments, R1 is an aryl. In some embodiments, the aryl is a phenyl.
In some embodiments, any of the siNA molécules disclosed herein comprise a 5’stabilized end cap selected from the group consisting of Formulas (1 A)-(15A), Formulas (9B)(12B), Formulas (9AX)-(12AX), Formulas (9AY)-(12AY), Formulas (9BX)-(12BX), and
Formulas (9BY)-(12BY):
102
Formula (9B)
Formula (9AX) Formula (9AY)
Formula (8A) Formula (9A)
Formula (9BX)
Formula (10AX)
Formula (9BY)
Formula (10B)
Formula (10BX)
Formula (10BY)
Formula (11 A)
Formula (11AX)
Formula (11AY)
103
Formula (11 B) Formula (11BX) Formula (11BY)
Formula (12A) Formula (12AX) Formula (12AY)
Formula (12B) Formula (12BX) Formula (12BY)
In some embodiments, any of the siNA molécules disclosed herein comprise a 5’stabilized end cap selected from the group consisting of Formula (21) to Formula (35):
Formula (25)
Formula (26)
Formula (27)
104
Formula (31) Formula (32) Formula (33)
Formula (34)
Formula (35) , wherein R1 is a nucleobase, aryl, heteroaryl, or
H. In some embodiments, R1 is an aryl. In some embodiments, the aryl is a phenyl.
In some embodiments, any of the siNA molécules disclosed herein comprise a 5’stabilized end cap selected from the group consisting of Formulas (21A)-(35A), Formulas (29B)(32B), Formulas (29AX)-(32AX), Formulas (29AY)-(32AY), Formulas (29BX)-(32BX), and Formulas (29BY)-(32BY):
105
Formula (28A)
Formula (29A)
Formula (29AX)
Formula (29AY)
Formula (29B)
Formula (29BX)
Formula (29BY)
Formula (30A)
Formula (30AX)
Formula (30AY)
Formula (30B) Formula (30BX) Formula (30BY)
Formula (31A)
Formula (31AX)
Formula (31AY)
Formula (31BY)
106
Formula (32A) Formula (32AX) Formula (32AY)
Formula (32B)
Formula (32BX)
In some embodiments, the 5’-stabilized end cap is attached to the 5’ end of the antisense strand. In some embodiments, the 5’-stabilized end cap is attached to the 5’ end of the antisense strand via 1, 2, 3, 4, or 5 or more linkers. In some embodiments, the one or more linkers are independently selected from the group consisting of a phosphodiester (p or po) linker, phosphorothioate (ps) linker (ps), phosphoramidite (HEG) linker, triethylene glycol (TEG) linker, and/or phosphorodithioate linker. In some embodiments, the one or more linkers are independently selected from the group consisting of p-(PS)2, (PS)2-p-TEG-p, (PS)2-p-HEG-p, and (PS)2-p-(HEG-p)2.
Linker
In some embodiments, any of the siRNAs, sense strands, first nucléotide sequences, antisense strands, and/or second nucléotide sequences disclosed herein comprise 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23 or more intemucleoside linkers. In some embodiments, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 or more intemucleoside linkers are independently selected from the group consisting of a phosphodiester (p or po) linker, phosphorothioate (ps) linker, or phosphorodithioate linker.
In some embodiments, any of the siRNAs, sense strands, first nucléotide sequences, antisense strands, and/or second nucléotide sequences disclosed herein further comprise 1, 2, 3, 4 or more linkers that attach a conjugated moiety, phosphorylation blocker, and/or 5’ end cap to
107 the siRNA, sense strand, first nucléotide sequence, antisense strand, and/or second nucléotide sequences. In some embodiments, the 1, 2, 3, 4 or more linkers are independently selected from the group consisting of a phosphodiester (p or po) linker, phosphorothioate (ps) linker, phosphoramidite (HEG) linker, triethylene glycol (TEG) linker, and/or phosphorodithioate linker. In some embodiments, the one or more linkers are independently selected from the group consisting of p-(PS)2, (PS)2-p-TEG-p, (PS)2-p-HEG-p, and (PS)2-p-(HEG-p)2.
Target Gene
Without wishing to be bound by theory, upon entry into a cell, any of the ds-siNA molécules disclosed herein may interact with proteins in the cell to form a RNA-Induced Silencing Complex (RISC). Once the ds-siNA is part of the RISC, the ds-siNA may be unwound to form a single-stranded siNA (ss-siNA). The ss-siNA may comprise the antisense strand of the ds-siNA. The antisense strand may bind to a complementary messenger RNA (mRNA), which results in silencing of the gene that encodes the mRNA.
The target gene may be any gene in a cell. In some embodiments, the target gene is a viral gene. In some embodiments, the viral gene is from a DNA virus. In some embodiments, the DNA virus is a double-stranded DNA (dsDNA) virus. In some embodiments, the dsDNA virus is a hepadnavirus. In some embodiments, the hepadnavirus is a hepatitis B virus (HBV). In some embodiments, the HBV is selected from HBV génotypes A-J.
In some embodiments, the target gene is selected from the S gene or X gene of the HBV. In some embodiments, the HBV has a genome sequence shown in the nucléotide sequence of SEQ ID NO: 410, which corresponds to the nucléotide sequence of GenBank Accession No. U95551.1, which is incorporated by reference in its entirety.
An exemplary HBV genome sequence is shown in SEQ ID NO: 596, corresponding to Genbank Accession No. KC315400.1, which is incorporated by reference in its entirety. Nucléotides 2307..3215,1..1623 of SEQ ID NO: 596 correspond to the polymerase/RT gene sequence, which encodes for the polymerase protein. Nucléotides 2848..3215,1..835 of SEQ ID NO: 596 correspond to the PreSl/S2/S gene sequence, which encodes for the large S protein. Nucléotides 3205..3215,1..835 of SEQ ID NO: 596 correspond to the PreS2/S gene sequence, which encodes for the middle S protein. Nucléotides 155..835 of SEQ ID NO: 596 correspond to the S gene sequence, which encodes the small S protein. Nucléotides 1374..1838 of SEQ ID NO: 596 correspond to the X gene sequence, which encodes the X protein. Nucléotides 1814..2452 of SEQ ID NO: 596 correspond to the PreC/C gene sequence, which encodes the precore/core protein. Nucléotides 1901..2452 of SEQ ID NO: 596 correspond to the C gene sequence, which encodes the core protein. The HBV genome further comprises viral regulatory éléments, such as
108 viral promoters (preS2, preSl, Core, and X) and enhancer éléments (ENH1 and ENH2). Nucléotides 1624..1771 ofSEQ ID NO: 596 correspond to ENH2. Nucléotides 1742..1849 of SEQ ID NO: 596 correspond to the Core promoter. Nucléotides 1818...3215,1..1930 ofSEQ ID NO: 596 correspond to the pregenomic RNA (pgRNA), which encodes the core and polymerase pro teins.
In some embodiments, the ASO is at least about 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% complementary or hybridizes to a viral target RNA sequence that begins in an X région of HBV or in an S région of HBV. The viral target may, e.g., begin at the 5'-end of targetsite in acc. KC315400.1 (génotype B, “gt B”), or in any one of génotypes A, C, or D. The skilled person would understand the HBV position, e.g., as described in Wing-Kin Sung, et al., Nature Genetics 44:765 (2012). In some embodiments, the S région is defmed as from the beginning of small S protein (in génotype B KC315400.1 isolate, position #155) to before beginning of X protein (in génotype B KC315400.1 isolate, position #1373). In some embodiments, the X région is defined as from the beginning X protein (in génotype B KC315400.1 isolate, position #1374) to end of DR2 site (in génotype B KC315400.1 isolate, position #1603).
In some embodiments, the second nucléotide sequence is at least about 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% complementary to 15 to 30, 15 to 25, 15 to 23, 15 to 22, 15 to 21, 17 to 25, 17 to 23, 17 to 22, 17 to 21, or 19 to 21 nucléotides within positions 200-720 or 1100-1700 of SEQ ID NO: 410. In some embodiments, the second nucléotide sequence is at least about 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% complementary to 15 to 30, 15 to 25, 15 to 23, 15 to 22, 15 to 21, 17 to 25, 17 to 23, 17 to 22, 17 to 21, or 19 to 21 nucléotides within positions 200-280, 300-445, 460-510, 650-720, 1170-1220, 1250-1300, or 1550-1630 of SEQ ID NO: 410. In some embodiments, the second nucléotide sequence is at least about 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% complementary to 15 to 30, 15 to 25, 15 to 23, 15 to 22, 15 to 21, 17 to 25, 17 to 23, 17 to 22, 17 to 21, or 19 to 21 nucléotides within positions 200-230, 250-280, 300-330, 370-400, 405-445, 460-500, 670-700, 1180-1210, 1260-1295, 15201550, or 1570-1610 ofSEQ ID NO: 410. In some embodiments, the second nucléotide sequence is at least about 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% complementary to 15 to 30, 15 to 25, 15 to 23, 15 to 22, 15 to 21, 17 to 25, 17 to 23, 17 to 22, 17 to 21, or 19 to 21 nucléotides starting at position 203, 206, 254, 305, 375, 409, 412, 415, 416, 419, 462, 466, 467, 674, 676, 1182, 1262, 1263, 1268, 1526, 1577, 1578, 1580, 1581, 1583, or 1584 ofSEQ ID NO: 410.
In some embodiments, the first nucléotide is at least about 60%, 65%, 70%, 75%, 80%, 85%>, 90%, 95%, or 100% identical to a nucléotide région within SEQ ID NO: 410, with the
109 exception that the thymines (Ts) in SEQ ID NO: 410 are replaced with uracil (U). In some embodiments, the first nucléotide sequence is at least about 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% identical to 15 to 30, 15 to 25, 15 to 23, 15 to 22, 15 to 21, 17 to 25, 17 to 23, 17 to 22, 17 to 21, or 19 to 21 nucléotides within positions 200-720 or 1100-1700 of SEQ ID NO: 410. In some embodiments, the first nucléotide sequence is at least about 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% identical to 15 to 30, 15 to 25, 15 to 23, 15 to 22, 15 to 21, 17 to 25, 17 to 23, 17 to 22, 17 to 21, or 19 to 21 nucléotides within positions 200-280, 300-445, 460-510, 650-720, 1170-1220, 1250-1300, or 1550-1630 of SEQ ID NO: 410. In some embodiments, the first nucléotide sequence is at least about 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% identical to 15 to 30, 15 to 25, 15 to 23, 15 to 22, 15 to 21, 17 to 25, 17 to 23, 17 to 22, 17 to 21, or 19 to 21 nucléotides within positions 200-230, 250-280, 300-330, 370400, 405-445,460-500, 670-700, 1180-1210, 1260-1295, 1520-1550, or 1570-1610 of SEQ ID NO: 410. In some embodiments, the first nucléotide sequence is at least about 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% identical to 15 to 30, 15 to 25, 15 to 23, 15 to 22, 15 to 21, 17 to 25, 17 to 23, 17 to 22, 17 to 21, or 19 to 21 nucléotides starting at position 203, 206, 254, 305, 375, 409, 412, 415, 416, 419, 462, 466, 467, 674, 676, 1182, 1262, 1263, 1268, 1526, 1577, 1578, 1580, 1581, 1583, or 1584 ofSEQ IDNO:410.
In some embodiments, the target gene is involved in liver metabolism. In some embodiments, the target gene is an inhibitor of the électron transport chain. In some embodiments, the target gene encodes the MC J protein (MCJ/DnaJC15 or MethylationControlled J protein). In some embodiments, the MC J protein is encoded by the mRNA sequence of SEQ ID NO: 411, which corresponds to the nucléotide sequence of GenBank Accession No. NM 013238.3, which is incorporated by reference in its entirety.
In some embodiments, the target gene is TAZ. In some embodiments, TAZ comprises the nucléotide sequence of SEQ ID NO: 412, which corresponds to the nucléotide sequence of GenBank Accession No. NM 000116.5, which is incorporated by reference in its entirety.
In some embodiments, the target gene is angiopoietin like 3 (ANGPTL3). In some embodiments, ANGPTL3 comprises the nucléotide sequence ofSEQ ID NO: 413, which corresponds to the nucléotide sequence of GenBank Accession No. NM_014495.4, which is incorporated by reference in its entirety.
In some embodiments, the target gene is diacylglycérol acyltransferase 2 (DGAT2). In some embodiments, DGAT2 comprises the nucléotide sequence ofSEQ ID NO: 414, which corresponds to the nucléotide sequence of GenBank Accession No. NM_001253891.1, which is incorporated by reference in its entirety.
110
Compositions
As indicated above, the présent disclosure provides compositions comprising any of the siNA molécules, sense strands, antisense strands, first nucléotide sequences, or second nucléotide sequences described herein. The compositions may comprise 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12 or more siNA molécules described herein. The compositions may comprise a first nucléotide sequence comprising a nucléotide sequence of any one SEQ ID NOs: 1-56, 103-158, and 205-260. In some embodiments, the composition comprises a second nucléotide sequence comprising a nucléotide sequence of any one of SEQ ID NOs: 57-102, 159-204, and 261-306. In some embodiments, the composition comprises a sense strand comprising a nucléotide sequence of any one of SEQ ID NOs: 307-362 and 415-444. In some embodiments, the composition comprises an antisense strand comprising a nucléotide sequence of any one of SEQ ID NOs: 363-409, 445-533, and 536-539.
Altematively, the compositions may comprise (a) a phosphorylation blocker; and (b) a short interfering nucleic acid (siNA). In some embodiments, the phosphorylation blocker is any of the phosphorylation blockers disclosed herein. In some embodiments, the siNA is any of the siNAs disclosed herein. In some embodiments, the siNA comprises any of the sense strands, antisense strands, first nucléotide sequences, or second nucléotide sequences described herein. In some embodiments, the siNA comprises any of the sense strands, antisense strands, first nucléotide sequences, or second nucléotide sequences described herein. In some embodiments, the siNA comprises one or more modified nucléotides. In some embodiments, the one or more modified nucléotides are independently selected from a 2’-fluoro nucléotide and a 2’-(9-methyl nucléotide. In some embodiments, the 2’-fluoro nucléotide or the 2’-(9-methyl nucléotide is independently selected from any of the 2’-fluoro or 2’-O-methyl nucléotide mimics disclosed herein. In some embodiments, the siNA comprises a nucléotide sequence comprising any of the modification patterns disclosed herein.
In some embodiments, the composition comprises (a) a conjugated moiety; and (b) a short interfering nucleic acid (siNA). In some embodiments, the conjugated moiety is any of the galactosamines disclosed herein. In some embodiments, the siNA is any of the siNAs disclosed herein. In some embodiments, the siNA comprises any of the sense strands, antisense strands, first nucléotide sequences, or second nucléotide sequences described herein. In some embodiments, the siNA comprises any of the sense strands, antisense strands, first nucléotide sequences, or second nucléotide sequences described herein. In some embodiments, the siNA comprises one or more modified nucléotides. In some embodiments, the one or more modified nucléotides are independently selected from a 2’-fluoro nucléotide and a 2’-O-methyl nucléotide.
111
In some embodiments, the 2’-fluoro nucléotide or the 2’-O-methyl nucléotide is independently selected from any of the 2’-fluoro or 2’-(9-methyl nucléotide mimics disclosed herein. In some embodiments, the siNA comprises a nucléotide sequence comprising any of the modification patterns disclosed herein.
In some embodiments, the composition comprises (a) a 5’-stabilized end cap; and (b) a short interfering nucleic acid (siNA). In some embodiments, the 5’-stabilized end cap is any of the 5-stabilized end caps disclosed herein. In some embodiments, the siNA is any of the siNAs disclosed herein. In some embodiments, the siNA comprises any of the sense strands, antisense strands, first nucléotide sequences, or second nucléotide sequences described herein. In some embodiments, the siNA comprises one or more modified nucléotides. In some embodiments, the one or more modified nucléotides are independently selected from a 2’-fluoro nucléotide and a 2’-O-methyl nucléotide. In some embodiments, the 2’-fluoro nucléotide or the 2’-(9-methyl nucléotide is independently selected from any of the 2’-fluoro or 2’-O-methyl nucléotide mimics disclosed herein. In some embodiments, the siNA comprises a nucléotide sequence comprising any of the modification patterns disclosed herein.
In some embodiments, the composition comprises (a) at least one phosphorylation blocker, conjugated moiety, or 5’-stabilized end cap; and (b) a short interfering nucleic acid (siNA). In some embodiments, the phosphorylation blocker is any of the phosphorylation blockers disclosed herein. In some embodiments, the conjugated moiety is any of the galactosamines disclosed herein. In some embodiments, the 5’-stabilized end cap is any of the 5stabilized end caps disclosed herein. In some embodiments, the siNA is any of the siNAs disclosed herein. In some embodiments, the siNA comprises any of the sense strands, antisense strands, first nucléotide sequences, or second nucléotide sequences described herein. In some embodiments, the siNA comprises one or more modified nucléotides. In some embodiments, the one or more modified nucléotides are independently selected from a 2’-fluoro nucléotide and a 2’-(9-methyl nucléotide. In some embodiments, the 2’-fluoro nucléotide or the 2’-O-methyl nucléotide is independently selected from any of the 2’-fluoro or 2’-(9-methyl nucléotide mimics disclosed herein. In some embodiments, the siNA comprises a nucléotide sequence comprising any of the modification patterns disclosed herein.
The composition may be a pharmaceutical composition. In some embodiments, the pharmaceutical composition comprises an amount of one or more of the siNA molécules described herein formulated with one or more pharmaceutically acceptable carriers (additives) and/or diluents. The pharmaceutical compositions may be specially formulated for administration in solid or liquid form, including those adapted for the following: (1) oral
112 administration, for example, drenches (aqueous or non-aqueous solutions or suspensions), tablets, e.g., those targeted for buccal, sublingual, and systemic absorption, boluses, powders, granules, pastes for application to the tongue; (2) parentéral administration, for example, by subcutaneous, intramuscular, intravenous or épidural injection as, for example, a stérile solution or suspension, or sustained-release formulation; (3) topical application, for example, as a cream, ointment, or a controlled-release patch or spray applied to the skin; (4) intravaginally or intrarectally, for example, as a pessary, cream or foam; (5) sublingually; (6) ocularly; (7) transdermally; or (8) nasally.
The phrase “therapeutically-effective amount” as used herein means that amount of a compound, material, or composition comprising a siNA of the présent disclosure which is effective for producing some desired therapeutic effect in at least a sub-population of cells in an animal at a reasonable benefit/risk ratio applicable to any medical treatment.
The phrase “pharmaceutically acceptable” is employed herein to refer to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animais without excessive toxicity, irritation, allergie response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.
Wetting agents, emulsifïers and lubricants, such as sodium lauryl sulfate and magnésium stéarate, as well as coloring agents, release agents, coating agents, sweetening, flavoring and perfuming agents, preservatives and antioxidants can also be présent in the compositions.
Examples of pharmaceutically-acceptable antioxidants include: (1) water soluble antioxidants, such as ascorbic acid, cysteine hydrochloride, sodium bisulfate, sodium metabisulfite, sodium sulfite and the like; (2) oil-soluble antioxidants, such as ascorbyl palmitate, butylated hydroxyanisole (BHA), butylated hydroxytoluene (BHT), lecithin, propyl gallate, alpha-tocopherol, and the like; and (3) métal chelating agents, such as citric acid, ethylenediamine tetraacetic acid (EDTA), sorbitol, tartaric acid, phosphoric acid, and the like.
Formulations of the présent disclosure include those suitable for oral, nasal, topical (including buccal and sublingual), rectal, vaginal and/or parentéral administration. The formulations may conveniently be presented in unit dosage form and may be prepared by any methods well known in the art of pharmacy. The amount of active ingrédient which can be combined with a carrier material to produce a single dosage form will vary depending upon the host being treated, the particular mode of administration. The amount of active ingrédient which can be combined with a carrier material to produce a single dosage form will generally be that amount of the compound (e.g., siNA molécule) which produces a therapeutic effect. Generally,
113 out of one hundred percent, this amount will range from about 0.1 percent to about ninety-nine percent of active ingrédient, preferably from about 5 percent to about 70 percent, most preferably from about 10 percent to about 30 percent.
In certain embodiments, a formulation of the présent disclosure. comprises an excipient selected from the group consisting of cyclodextrins, celluloses, liposomes, micelle forming agents, e.g., bile acids, and polymeric carriers, e.g., polyesters and polyanhydrides; and a compound (e.g., siNA molécule) of the présent disclosure. In certain embodiments, an aforementioned formulation renders orally bioavailable a compound (e.g., siNA molécule) of the présent disclosure.
Methods of preparing these formulations or compositions include the step of bringing into association a compound (e.g., siNA molécule) of the présent disclosure with the carrier and, optionally, one or more accessory ingrédients. In general, the formulations are prepared by uniformly and intimately bringing into association a compound (e.g., siNA molécule) of the présent disclosure with liquid carriers, or finely divided solid carriers, or both, and then, if necessary, shaping the product.
Formulations of the disclosure suitable for oral administration may be in the form of capsules, cachets, pills, tablets, lozenges (using a flavored basis, usually sucrose and acacia or tragacanth), powders, granules, or as a solution or a suspension in an aqueous or non-aqueous liquid, or as an oil-in-water or water-in-oil liquid émulsion, or as an élixir or syrup, or as pastilles (using an inert base, such as gelatin and glycerin, or sucrose and acacia) and/or as mouth washes and the like, each containing a predetermined amount of a compound (e.g, siNA molécule) of the présent disclosure as an active ingrédient. A compound (e.g, siNA molécule) of the présent disclosure may also be administered as a bolus, electuary or paste.
In solid dosage forms of the disclosure for oral administration (capsules, tablets, pills, dragees, powders, granules, tronches and the like), the active ingrédient is mixed with one or more pharmaceutically-acceptable carriers, such as sodium citrate or dicalcium phosphate, and/or any of the following: (1) fillers or extenders, such as starches, lactose, sucrose, glucose, mannitol, and/or silicic acid; (2) binders, such as, for example, carboxymethylcellulose, alginates, gelatin, polyvinyl pyrrolidone, sucrose and/or acacia; (3) humectants, such as glycerol; (4) disintegrating agents, such as agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates, and sodium carbonate; (5) solution retarding agents, such as paraffm; (6) absorption accelerators, such as quatemary ammonium compounds and surfactants, such as poloxamer and sodium lauryl sulfate; (7) wetting agents, such as, for example, cetyl alcohol, glycerol monostearate, and non-ionic surfactants; (8) absorbents, such as kaolin and bentonite
114 clay; (9) lubricants, such as talc, calcium stéarate, magnésium stéarate, solid polyethylene glycols, sodium lauryl sulfate, zinc stéarate, sodium stéarate, stearic acid, and mixtures thereof; (10) coloring agents; and (11) controlled release agents such as crospovidone or ethyl cellulose.
In the case of capsules, tablets and pills, the pharmaceutical compositions may also comprise buffering agents. Solid compositions of a similar type may also be employed as fillers in soft and hard-shelled gelatin capsules using such excipients as lactose or milk sugars, as well as high molecular weight polyethylene glycols and the like.
A tablet may be made by compression or molding, optionally with one or more accessory ingrédients. Compressed tablets may be prepared using binder (for example, gelatin or hydroxypropylmethyl cellulose), lubricant, inert diluent, preservative, disintegrant (for example, sodium starch glycolate or cross-linked sodium carboxymethyl cellulose), surface-active or dispersing agent. Molded tablets may be made by molding in a suitable machine a mixture of the powdered compound moistened with an inert liquid diluent.
The tablets, and other solid dosage forms of the pharmaceutical compositions of the présent disclosure, such as dragees, capsules, pills and granules, may optionally be scored or prepared with coatings and shells, such as enteric coatings and other coatings well known in the pharmaceutical-formulating art. They may also be formulated so as to provide slow or controlled release of the active ingrédient therein using, for example, hydroxypropylmethyl cellulose in varying proportions to provide the desired release profile, other polymer matrices, liposomes and/or microspheres. They may be formulated for rapid release, e.g., fireeze-dried.
They may be sterilized by, for example, filtration through a bacteria-retaining filter, or by incorporating sterilizing agents in the form of stérile solid compositions which can be dissolved in stérile water, or some other stérile injectable medium immediately before use. These compositions may also optionally contain opacifying agents and may be of a composition that they release the active ingredient(s) only, or preferentially, in a certain portion of the gastrointestinal tract, optionally, in a delayed manner. Examples of embedding compositions which can be used include polymeric substances and waxes. The active ingrédient can also be in micro-encapsulated form, if appropriate, with one or more of the above-described excipients.
Liquid dosage forms for oral administration of the compounds (e.g., siNA molécules) of the disclosure include pharmaceutically acceptable émulsions, microemulsions, solutions, suspensions, syrups and élixirs. In addition to the active ingrédient, the liquid dosage forms may contain inert diluents commonly used in the art, such as, for example, water or other solvents, solubilizing agents and emulsifiers, such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1,3-butylene glycol, oils (I
115 particular, cottonseed, groundnut, corn, germ, olive, castor and sesame oils), glycerol, tetrahydrofuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, and mixtures thereof.
Besides inert diluents, the oral compositions can also include adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, coloring, perfuming and preservative agents.
Suspensions, in addition to the active compounds (e.g., siNA molécules), may contain suspending agents as, for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, micro crystalline cellulose, aluminum metahydroxide, bentonite, agar-agar and tragacanth, and mixtures thereof.
Formulations of the pharmaceutical compositions of the disclosure for rectal or vaginal administration may be presented as a suppository, which may be prepared by mixing one or more compounds (e.g., siNA molécules) of the disclosure with one or more suitable nonirritating excipients or carriers comprising, for example, cocoa butter, polyethylene glycol, a suppository wax or a salicylate, and which is solid at room température, but liquid at body température and, therefore, will melt in the rectum or vaginal cavity and release the active compound (e.g., siNA molécule).
Formulations of the présent disclosure which are suitable for vaginal administration also include pessaries, tampons, creams, gels, pastes, foams or spray formulations containing such carriers as are known in the art to be appropriate.
Dosage forms for the topical or transdermal administration of a compound (e.g., siNA molécule) of this disclosure include powders, sprays, ointments, pastes, creams, lotions, gels, solutions, patches and inhalants. The active compound (e.g., siNA molécule) may be mixed under stérile conditions with a pharmaceutically acceptable carrier, and with any preservatives, buffers, or propellants which may be required.
The ointments, pastes, creams and gels may contain, in addition to an active compound (e.g., siNA molécule) of this disclosure, excipients, such as animal and vegetable fats, oils, waxes, paraffms, starch, tragacanth, cellulose dérivatives, polyethylene glycols, silicones, bentonites, silicic acid, talc and zinc oxide, or mixtures thereof.
Powders and sprays can contain, in addition to a compound (e.g., siNA molécule) of this disclosure, excipients such as lactose, talc, silicic acid, aluminum hydroxide, calcium silicates and polyamide powder, or mixtures of these substances. Sprays can additionally contain customary propellants, such as chlorofluorohydrocarbons and volatile unsubstituted hydrocarbons, such as butane and propane.
116
Transdermal patches hâve the added advantage of providing controlled delivery of a compound (e.g., siNA molécule) of the présent disclosure to the body. Such dosage forms can be made by dissolving or dispersing the compound (e.g., siNA molécule) in the proper medium. Absorption enhancers can also be used to increase the flux of the compound (e.g., siNA molécule) across the skin. The rate of such flux can be controlled by either providing a rate controlling membrane or dispersing the compound (e.g., siNA molécule) in a polymer matrix or gel.
Ophthalmic formulations, eye ointments, powders, solutions and the like, are also contemplated as being within the scope of this invention.
Pharmaceutical compositions of this disclosure suitable for parentéral administration comprise one or more compounds (e.g., siNA molécules) of the disclosure in combination with one or more pharmaceutically-acceptable stérile isotonie aqueous or nonaqueous solutions, dispersions, suspensions or émulsions, or stérile powders which may be reconstituted into stérile injectable solutions or dispersions just prior to use, which may contain sugars, alcohols, antioxidants, buffers, bacteriostats, solutés which render the formulation isotonie with the blood of the intended récipient or suspending or thickening agents.
Examples of suitable aqueous and nonaqueous carriers which may be employed in the pharmaceutical compositions of the disclosure include water, éthanol, polyols (such as glycerol, propylene glycol, polyethylene glycol, and the like), and suitable mixtures thereof, vegetable oils, such as olive oil, and injectable organic esters, such as ethyl oleate. Proper fluidity can be maintained, for example, by the use of coating materials, such as lecithin, by the maintenance of the required particle size in the case of dispersions, and by the use of surfactants.
These compositions may also contain adjuvants such as preservatives, wetting agents, emulsifying agents and dispersing agents. Prévention of the action of microorganisms upon the subject compounds may be ensured by the inclusion of varions antibacterial and antifungal agents, for example, paraben, chlorobutanol, phénol sorbic acid, and the like. It may also be désirable to include isotonie agents, such as sugars, sodium chloride, and the like into the compositions. In addition, prolonged absorption of the injectable pharmaceutical form may be brought about by the inclusion of agents which delay absorption such as aluminum monostearate and gelatin.
In some cases, in order to prolong the effect of a drug, it is désirable to slow the absorption of the drug from subeutaneous or intramuscular injection. This may be accomplished by the use of a liquid suspension of crystalline or amorphous material having poor water solubility. The rate of absorption of the drug then dépends upon its rate of dissolution which, in
117 tum, may dépend upon crystal size and crystalline form. Altematively, delayed absorption of a parenterally-administered drug form is accomplished by dissolving or suspending the drug in an oil vehicle.
Injectable depot forms are made by forming microencapsule matrices of the subject compounds (e.g., siNA molécules) in biodégradable polymers such as polylactide-polyglycolide. Depending on the ratio of drug to polymer, and the nature of the particular polymer employed, the rate of drug release can be controlled. Examples of other biodégradable polymers include poly(orthoesters) and poly(anhydrides). Depot injectable formulations are also prepared by entrapping the drug in liposomes or microemulsions which are compatible with body tissue.
When the compounds (e.g., siNA molécules) of the présent disclosure are administered as pharmaceuticals, to humans and animais, they can be given per se or as a pharmaceutical composition containing, for example, 0.1 to 99% (more preferably, 10 to 30%) of active ingrédient in combination with a pharmaceutically acceptable carrier.
Methods of Treatment and Administration
The siNA molécules of the présent disclosure may be used to treat a disease in a subject in need thereof. In some embodiments, a method of treating a disease in a subject in need thereof comprises administering to the subject any of the siNA molécules disclosed herein. In some embodiments, a method of treating a disease in a subject in need thereof comprises administering to the subject any of the compositions disclosed herein.
The préparations (e.g, siNA molécules or compositions) of the présent disclosure may be given orally, parenterally, topically, or rectally. They are of course given in forms suitable for each administration route. For example, they are administered in tablets or capsule form, administration by injection, infùsion or inhalation; topical by lotion or ointment; and rectal by suppositories. Oral administrations are preferred.
The phrases “parentéral administration” and “administered parenterally” as used herein means modes of administration other than enterai and topical administration, usually by injection, and includes, without limitation, intravenous, intramuscular, intraarterial, intrathecal, intracapsular, intraorbital, intracardiac, intradermal, intraperitoneal, transtracheal, subcutaneous, subcuticular, intraarticular, subcapsular, subarachnoid, intraspinal and intrastemal injection and infusion.
The phrases “systemic administration,” “administered systemically,” “peripheral administration” and “administered peripherally” as used herein mean the administration of a compound, drug or other material other than directly into the central nervous System, such that it
118 enters the patient’s System and, thus, is subject to metabolism and other like processes, for example, subcutaneous administration.
These compounds may be administered to humans and other animais for therapy by any suitable route of administration, including orally, nasally, as by, for example, a spray, rectally, intravaginally, parenterally, intracistemally and topically, as by powders, ointments or drops, including buccally and sublingually.
Regardless of the route of administration selected, the compounds (e.g., siNA molécules) of the présent disclosure, which may be used in a suitable hydrated form, and/or the pharmaceutical compositions of the présent disclosure, are formulated into pharmaceuticallyacceptable dosage forms by conventional methods known to those of skill in the art.
Actual dosage levels of the active ingrédients in the pharmaceutical compositions of this disclosure may be varied so as to obtain an amount of the active ingrédient which is effective to achieve the desired therapeutic response for a particular patient, composition, and mode of administration, without being toxic to the patient.
The selected dosage level will dépend upon a variety of factors including the activity of the particular compound (e.g., siNA molécule) of the présent disclosure employed, or the ester, sait or amide thereof, the route of administration, the time of administration, the rate of excrétion or metabolism of the particular compound being employed, the rate and extent of absorption, the duration of the treatment, other drugs, compounds and/or materials used in combination with the particular compound employed, the âge, sex, weight, condition, general health and prior medical history of the patient being treated, and like factors well known in the medical arts.
A physician or veterinarian having ordinary skill in the art can readily détermine and prescribe the effective amount of the pharmaceutical composition required. For example, the physician or veterinarian could start doses of the compounds (e.g., siNA molécules) of the disclosure employed in the pharmaceutical composition at levels lower than that required in order to achieve the desired therapeutic effect and gradually increase the dosage until the desired effect is achieved.
In general, a suitable daily dose of a compound (e.g., siNA molécule) of the disclosure is the amount of the compound that is the lowest dose effective to produce a therapeutic effect. Such an effective dose generally dépends upon the factors described above. Preferably, the compounds are administered at about 0.01 mg/kg to about 200 mg/kg, more preferably at about 0.1 mg/kg to about 100 mg/kg, even more preferably at about 0.5 mg/kg to about 50 mg/kg. In some embodiments, the compound is administered at a dose equal to or greater than 0.01, 0.02, 0.03, 0.04, 0.05, 0.06, 0.07, 0.08, 0.09, 0.10, 0.11, 0.12, 0.13, 0.14, 0.15, 0.16, 0.17, 0.18, 0.19,
119
0.20, 0.21, 0.22, 0.23, 0.24, 0.25, 0.26, 0.27, 0.28, 0.29, 0.30, 0.35, 0.40, 0.45, 0.50, 0.55, 0.60, 0.65, 0.7, 0.75, 0.8, 0.85, 0.9, 0.95, or 1 mg/kg. In some embodiments, the compound is administered at a dose equal to or less than 200, 190, 180, 170, 160, 150, 140, 130, 120, 110, 100, 95, 90, 85, 80, 75, 70, 65, 60, 55, 50, 45, 40, 35, 30, 25, 20, or 15 mg/kg. In some embodiments, the total daily dose of the compound is equal to or greater than 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 105, 110, 115, 120, 125, 130, 135, 140, 145, 150, 155, 160, 165, 170, 175, 180, 185, 190, 195, or 100 mg.
When the compounds (e.g., siNA molécules) described herein are co-administered with another, the effective amount may be less than when the compound is used alone.
If desired, the effective daily dose of the active compound (e.g., siNA molécule) may be administered as two, three, four, five, six or more sub-doses administered separately at appropriate intervals throughout the day, optionally, in unit dosage forms. Preferred dosing is one administration per day. In some embodiments, the compound is administered at least 1, 2, 3, 4, 5,6, 7, 8,9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, or 21 times a week. In some embodiments, the compound is administered at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, or 21 times a month. In some embodiments, the compound is administered onceevery 1,2, 3,4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, or 21 days. In some embodiments, the compound is administered once every 1, 2, 3, 4, 5, 6, 7, or 8 weeks.
Diseases
The siNA molécules and compositions described herein may be administered to a subject to treat a disease. Further disclosed herein are uses of any of the siNA molécules or compositions disclosed herein in the manufacture of a médicament for treating a disease.
In some embodiments, the disease is a viral disease. In some embodiments, the viral disease is caused by a DNA virus. In some embodiments, the DNA virus is a double stranded DNA (dsDNA virus). In some embodiments, the dsDNA virus is a hepadnavirus. In some embodiments, the hepadnavirus is a hepatitis B virus (HBV).
In some embodiments, the disease is a liver disease. In some embodiments, the liver disease is nonalcoholic fatty liver disease (NAFLD). In some embodiments, the NAFLD is nonalcoholic steatohepatitis (NASH). In some embodiments, the liver disease is hepatocellular carcinoma (HCC).
Administration of siNA
Administration of any of the siNAs disclosed herein may be conducted by methods known in the art. In some embodiments, the siNA is administered by subcutaneous (SC) or intravenous (IV) delivery. The préparations (e.g., siNAs or compositions) of the présent
120 disclosure may be given orally, parenterally, topically, or rectally. They are of course given in forms suitable for each administration route. For example, they are administered in tablets or capsule form, administration by injection, infusion or inhalation; topical by lotion or ointment; and rectal by suppositories. In some embodiments, subcutaneous administration is preferred.
The phrases “parentéral administration” and “administered parenterally” as used herein means modes of administration other than enterai and topical administration, usually by injection, and includes, without limitation, intravenous, intramuscular, intraarterial, intrathecal, intracapsular, intraorbital, intracardiac, intradermal, intraperitoneal, transtracheal, subcutaneous, subcuticular, intraarticular, subcapsular, subarachnoid, intraspinal and intrastemal injection and infusion.
The phrases “systemic administration,” “administered systemically,” “peripheral administration” and “administered peripherally” as used herein mean the administration of a compound, drug or other material other than directly into the central nervous System, such that it enters the patient’s System and, thus, is subject to metabolism and other like processes, for example, subcutaneous administration.
These compounds may be administered to humans and other animais for therapy by any suitable route of administration, including orally, nasally, as by, for example, a spray, rectally, intravaginally, parenterally, intracistemally and topically, as by powders, ointments or drops, including buccally and sublingually.
Regardless of the route of administration selected, the compounds (e.g., siNAs) of the présent disclosure, which may be used in a suitable hydrated form, and/or the pharmaceutical compositions of the présent disclosure, are formulated into pharmaceutically-acceptable dosage forms by conventional methods known to those of skill in the art.
Actual dosage levels of the active ingrédients in the pharmaceutical compositions of this disclosure may be varied so as to obtain an amount of the active ingrédient which is effective to achieve the desired therapeutic response for a particular patient, composition, and mode of administration, without being toxic to the patient.
The selected dosage level will dépend upon a variety of factors including the activity of the particular compound (e.g., siNA) of the présent disclosure employed, or the ester, sait or amide thereof, the route of administration, the time of administration, the rate of excrétion or metabolism of the particular compound being employed, the rate and extent of absorption, the duration of the treatment, other drugs, compounds and/or materials used in combination with the particular compound employed, the âge, sex, weight, condition, general health and prior medical history of the patient being treated, and like factors well known in the medical arts.
121
A physician or veterinarian having ordinary skill in the art can readily détermine and prescribe the effective amount of the pharmaceutical composition required. For example, the physician or veterinarian could start doses of the compounds (e.g., siNAs) of the disclosure employed in the pharmaceutical composition at levels lower than that required in order to achieve the desired therapeutic effect and gradually increase the dosage until the desired effect is achieved.
In general, a suitable daily dose of a compound (e.g., siNA) of the disclosure is the amount of the compound that is the lowest dose effective to produce a therapeutic effect. Such an effective dose generally dépends upon the factors described above. Preferably, the compounds are administered at about Ô.01 mg/kg to about 200 mg/kg, more preferably at about 0.1 mg/kg to about 100 mg/kg, even more preferably at about 0.5 mg/kg to about 50 mg/kg. In some embodiments, the compound is administered at about 1 mg/kg to about 40 mg/kg, about 1 mg/kg to about 30 mg/kg, about 1 mg/kg to about 20 mg/kg, about 1 mg/kg to about 15 mg/kg, or 1 mg/kg to about 10 mg/kg. In some embodiments, the compound is administered at a dose equal to or greater than 0.01, 0.02, 0.03, 0.04, 0.05, 0.06, 0.07, 0.08, 0.09, 0.10, 0.11, 0.12, 0.13, 0.14, 0.15, 0.16, 0.17, 0.18, 0.19, 0.20, 0.21, 0.22, 0.23, 0.24, 0.25, 0.26, 0.27, 0.28, 0.29, 0.30, 0.35, 0.40, 0.45, 0.50, 0.55, 0.60, 0.65, 0.7, 0.75, 0.8, 0.85, 0.9, 0.95, or 1 mg/kg. In some embodiments, the compound is administered at a dose equal to or greater than 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20,21,22, 23,24, 25, 26, 27, 28,29, or 30 mg/kg. In some embodiments, the compound is administered at a dose equal to or less than 200, 190, 180, 170, 160, 150, 140, 130, 120, 110, 100, 95, 90, 85, 80, 75, 70, 65, 60, 55, 50, 45, 40, 35, 30, 25, 20, or 15 mg/kg. In some embodiments, the total daily dose of the compound is equal to or greater than 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 105, 110, 115, 120, 125, 130, 135, 140, 145, 150, 155, 160, 165, 170, 175, 180, 185, 190, 195, or 100 mg.
If desired, the effective daily dose of the active compound (e.g., siNA) may be administered as two, three, four, five, six, seven, eight, nine, ten or more doses or sub-doses administered separately at appropriate intervals throughout the day, optionally, in unit dosage forms. In some embodiments, the compound is administered at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, or 15 times. Preferred dosing is one administration per day. In some embodiments, the compound is administered at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, or 21 times a week. In some embodiments, the compound is administered at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, or 21 times amonth. In some embodiments, the compound is administered once every 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, or 21 days. In some embodiments, the compound is administered
122 every 3 days. In some embodiments, the compound is administered once every 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, or 15 weeks. In some embodiments, the compound is administered every month. In some embodiments, the compound is administered once every 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, or 15 months. In some embodiments, the compound is administered at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, or 53 times overaperiod ofat least 1,2, 3,4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20,21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47,
48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, or 70 days. In some embodiments, the compound is administered at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39,
40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, or 53 times over aperiod of at least 1, 2, 3, 4, 5,
6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, or 53 weeks. In some embodiments, the compound is administered at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, or 53 times over aperiod of at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, or 53 months. In some embodiments, the compound is administered at least once a week for a period of at least 1, 2, 3, 4,5,6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20,21,22, 23,24, 25,26, 27,28,29,30,31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, or 70 weeks. In some embodiments, the compound is administered at least once a week for a period of at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39,
40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65,
66, 67, 68, 69, or 70 months. In some embodiments, the compound is administered at least twice a week for aperiod of at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21,
22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47,
48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, or 70 weeks.
In some embodiments, the compound is administered at least twice a week for a period of at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, or 70 months. In some embodiments, the
123 compound is administered at least once every two weeks for a period of at least 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20,21,22, 23,24, 25, 26, 27,28,29,30,31,32,33,34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, or 70 weeks. In some embodiments, the compound is administered at least once every two weeks for a period of at least 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, or 70 months. In some embodiments, the compound is administered at least once every four weeks for a period of at least 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44,
45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, or weeks. In some embodiments, the compound is administered at least once every four weeks for a period ofatleast4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20,21,22, 23,24, 25,
26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51,
52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, or 70 months.
In some embodiments, any one of the siNAs or compositions disclosed herein is administered in a particle or viral vector. In some embodiments, the viral vector is a vector of adenovirus, adeno-associated virus (AAV), alphavirus, flavivirus, herpes simplex virus, lentivirus, measles virus, picomavirus, poxvirus, rétro virus, or rhabdo virus. In some embodiments, the viral vector is a recombinant viral vector. In some embodiments, the viral vector is selected from AAVrh.74, AAVrh.10, AAVrh.20, AAV-1, AAV-2, AAV-3, AAV-4, AAV-5, AAV-6, AAV-7, AAV-8, AAV-9, AAV-10, AAV-11, AAV-12 and AAV-13.
The subject of the described methods may be a mammal, and it includes humans and non-human mammals. In some embodiments, the subject is a human, such as an adult human.
Some embodiments include a method for treating an HBV virus in a subject infected with the virus comprising administering a therapeutically effective amount of one or more siNA of the présent disclosure or a composition of the présent disclosure to the subject in need thereof thereby reducing the viral load of the virus in the subject and/or reducing a level of a virus antigen in the subject. The siNA may be complementary or hybridize to a portion of the target RNA in the virus, e.g., an X région and/or an S région of HBV.
Combination Thérapies
Any of the methods disclosed herein may further comprise administering to the subject an additional HBV treatment agent. Any of the compositions disclosed herein may further comprise an additional HBV treatment agent. In some embodiments, the additional HBV
124 treatment agent is selected from a nucléotide analog, nucleoside analog, a capsid assembly modulator (CAM), a recombinant interferon, an entry inhibitor, a small molécule immunomodulator and oligonucleotide therapy. In some embodiments, the additional HBV treatment agent is selected from HBV STOPS™ ALG-010133, HBV CAM ALG-000184, ASO 1, recombinant interferon alpha 2b, IFN-a, PEG-IFN-a-2a, lamivudine, telbivudine, adefovir dipivoxil, clevudine, entecavir, tenofovir alafenamide, tenofovir disoproxil, NVR3-778, BAY414109, JNJ-632, JNJ-3989 (ARO-HBV), RG6004, GSK3228836, REP-2139, REP-2165, AB-729, VIR-2218, RG6346 (DCR-HBVS), JNJ-6379, GLS4, ABI-HO731, JNJ-440, NZ-4, RG7907, EDP-514, AB-423, AB-506, ABI-H03733 and ABI-H2158. In some embodiments, the oligonucleotide therapy is selected from Nucleic Acid Polymers or S-Antigen Transportinhibiting Oligonucleotide Polymers (NAPs or STOPS), siRNA, and ASO. In some embodiments, the oligonucleotide therapy is an additional siNA. In some embodiments, the additional siNA is selected from any of ds-siNA-001 to ds-siNA-0178. In some embodiments, the oligonucleotide therapy is an antisense oligonucleotide (ASO). In some embodiments, the ASO is ASO 1. In some embodiments, any of the siNAs disclosed herein are co-administered with STOPS. Exemplary STOPS are described in International Publication No. W02020/097342 and U.S. Publication No. 2020/0147124, both of which are incorporated by reference in their entirety. In some embodiments, the STOPS is ALG-010133. In some embodiments, any of the siNAs disclosed herein are co-administered with tenofovir. In some embodiments, any of the siNAs disclosed herein are co-administered with a CAM. Exemplary CAMs are described in Berke et al., Antimicrob Agents Chemother, 2017, 61(8):e00560-17, Klumpp, et al., Gastroenterology, 2018, 154(3):652-662.e8, International Application Nos.
PCT/US2020/017974, PCT/US2020/026116, and PCT/US2020/028349 and U.S. Application Nos. 16/789,298, 16/837,515, and 16/849,851, each which is incorporated by reference in its entirety. In some embodiments, the CAM is ALG-000184, ALG-001075, ALG-001024, JNJ632, BAY41-4109, or NVR3-778. In some embodiments, the siNA and the HBV treatment agent are administered simultaneously. In some embodiments, the siNA and the HBV treatment agent are administered concurrently. In some embodiments, the siNA and the HBV treatment agent are administered sequentially. In some embodiments, the siNA is administered prior to administering the HBV treatment agent. In some embodiments, the siNA is administered after administering the HBV treatment agent. In some embodiments, the siNA and the HBV treatment agent are in separate containers. In some embodiments, the siNA and the HBV treatment agent are in the same container.
125
Any of the methods disclosed herein may further comprise administering to the subject a liver disease treatment agent. Any of the compositions disclosed herein may further comprise a liver disease treatment agent. In some embodiments, the liver disease treatment agent is selected from a peroxisome proliferator-activator receptor (PPAR) agonist, famesoid X receptor (FXR) agonist, lipid-altering agent, and incretin-based therapy. In some embodiments, the PPAR agonist is selected from a PPARa agonist, dual PPARa/δ agonist, PPARy agonist, and dual PPARa/γ agonist. In some embodiments, the dual PPARa agonist is a fïbrate. In some embodiments, the PPARa/δ agonist is elafibranor. In some embodiments, the PPARy agonist is a thiazolidinedione (TZD). In some embodiments, TZD is pioglitazone. In some embodiments, the dual PPARa/γ agonist is saroglitazar. In some embodiments, the FXR agonist is obeticholic acis (OCA). In some embodiments, the lipid-altering agent is aramchol. In some embodiments, the incretin-based therapy is a glucagon-like peptide 1 (GLP-1) receptor agonist or dipeptidyl peptidase 4 (DPP-4) inhibitor. In some embodiments, the GLP-1 receptor agonist is exenatide or liraglutide. In some embodiments, the DPP-4 inhibitor is sitagliptin or vildapliptin. In some embodiments, the siNA and the liver disease treatment agent are administered concurrently. In some embodiments, the siNA and the liver disease treatment agent are administered sequentially. In some embodiments, the siNA is administered prior to administering the liver disease treatment agent. In some embodiments, the siNA is administered after administering the liver disease treatment agent. In some embodiments, the siNA and the liver disease treatment agent are in separate containers. In some embodiments, the siNA and the liver disease treatment agent are in the same container.
Définitions
Unless defmed otherwise, ail technical and scientific terms used herein hâve the meaning commonly understood by a person skilled in the art to which this disclosure belongs. The following references provide one of skill with a general définition of many of the terms used in this invention: Singleton et al., Dictionary of Microbiology and Molecular Biology (2nd ed. 1994); The Cambridge Dictionary of Science and Technology (Walker ed., 1988); The Glossary of Genetics, 5th Ed., R. Rieger et al., (eds.), Springer Verlag (1991); and Haie & Marham, The Harper Collins Dictionary of Biology (1991). As used herein, the following terms hâve the meanings ascribed to them below, unless specified otherwise. The terminology used herein is for the purpose of describing particular embodiments only and is not intended to be limiting of the disclosure.
The terms “a” and “an” as used herein mean “one or more” and include the plural unless the context is inappropriate.
126
As used herein, the ternis “patient” and “subject” refer to organisms to be treated by the methods of the présent disclosure. Such organisms are preferably mammals (e.g., marines, simians, equines, bovines, porcinis, canines, felines, and the like), and more preferably humans.
As used herein, the terni “effective amount” refers to the amount of a compound (e.g., a siNA of the présent disclosure) sufficient to effect bénéficiai or desired results. An effective amount can be administered in one or more administrations, applications, or dosages and is not intended to be limited to a particular formulation or administration route.
As used herein, the term “treating” includes any effect, e.g., lessening, reducing, modulating, ameliorating or eliminating, that results in the improvement of the condition, disease, disorder, and the like, or ameliorating a symptom thereof.
As used herein, the ternis “alleviate” and “alleviating” refer to reducing the severity of the condition, such as reducing the severity by, for example, at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or 95%.
As used herein, the term “pharmaceutical composition” refers to the combination of an active agent with a carrier, inert or active, making the composition especially suitable for diagnostic or therapeutic use in vivo or ex vivo.
As used herein, the term “pharmaceutically acceptable carrier” refers to any of the standard pharmaceutical carriers, such as a phosphate buffered saline solution, water, émulsions (e.g., such as an oil/water or water/oil émulsions), and various types of wetting agents. The compositions also can include stabilizers and preservatives. For examples of carriers, stabilizers and adjuvants, see, for example, Martin, Remington's Pharmaceutical Sciences, 15th Ed., Mack Publ. Co., Easton, PA [1975],
The term “about” as used herein when referring to a measurable value (e.g., weight, time, and dose) is meant to encompass variations, such as ±10%, ±5% , ±1% , or ±0.1% of the specified value.
As used herein, the term “nucleobase” refers to a nitrogen-containing biological compound that forms a nucleoside. Examples of nucleobases include, but are not limited to, thymine, uracil, adenine, cytosine, guanine, aryl, heteroaryl, and an analogue or dérivative thereof.
Throughout the description, where compositions are described as having, including, or comprising spécifie components, or where processes and methods are described as having, including, or comprising spécifie steps, it is contemplated that, additionally, there are compositions of the présent disclosure that consist essentially of, or consist of, the recited
127 components, and that there are processes and methods according to the présent disclosure that consist essentially of, or consist of, the recited processing steps.
As a general matter, compositions specifying a percentage are by weight unless otherwise specified. Further, if a variable is not accompanied by a définition, then the previous définition of the variable Controls.
Ail publications and patents cited in this spécification are herein incorporated by reference as if each individual publication or patent were specifically and individually indicated to be incorporated by reference and are incorporated herein by reference to disclose and describe the methods and/or materials in connection with which the publications are cited. The citation of any publication is for its disclosure prior to the filing date and should not be construed as an admission that the présent invention is not entitled to antedate such publication by virtue of prior invention. Further, the dates of publication provided may be different from the actual publication dates that may need to be independently confirmed.
EXAMPLES
Example 1. siNA Synthesis
This example describes an exemplary method for synthesizing ds-siNAs, such as the siNAs disclosed in Table 6 (as identified by the ds-siNA ID).
The 2’-OMe phosphoramidite 5’-O-DMT-deoxy Adenosine (NH-Bz), 3’-O-(2cyanoethyl-N,N-diisopropyl phosphoramidite, 5’-O-DMT-deoxy Guanosine (NH-ibu), 3’-O-(2cyanoethyl-N,N-diisopropyl phosphoramidite, 5’-O-DMT-deoxy Cytosine (NH-Bz), 3’-O-(2cyanoethyl-N,N-diisopropyl phosphoramidite, 5’-O-DMT-Uridine 3’-O-(2-cyanoethyl-N,Ndiisopropyl phosphoramidite and solid supports were purchased from Chemgenes Corp. MA.
NC NC
NC NC
The 2’-F -5’-O-DMT-(NH-Bz) Adenosine-3’-O-(2-cyanoethyl-N,N-diisopropyl phosphoramidite, 2’-F -5’-O-DMT-(NH-ibu)- Guanosine, 3’-O-(2-cyanoethyl-N,N-diisopropyl
128 phosphoramidite, 5’-0-DMT-(NH-Bz)- Cytosine, 2’-F-3’-O-(2-cyanoethyl-N,N-diisopropyl phosphoramidite, 5’-0-DMT-Uridine, 2’-F-3’-O-(2-cyanoethyl-N,N-diisopropyl phosphoramidite and solid supports were purchased from Thermo Fischer Milwaukee WI, USA.
Ail the monomers were dried in vacuum desiccator with desiccants (P2O5, RT 24h). The solid supports (CPG) attached to the nucleosides and universal supports was obtained from LGC and Chemgenes. The Chemicals and solvents for post synthesis workflow were purchased from commercially available sources like VWR/Sigma and used without any purification or treatment. Solvent (Acetonitrile) and solutions (amidite and activator) were stored over molecular sieves during synthesis.
The oligonucleotides were synthesized on a DNA/RNA Synthesizers (Expedite 8909 or AB1-394) using standard oligonucleotide phosphoramidite chemistry starting from the 3' residue of the oligonucleotide preloaded on CPG support. An extended coupling of 0.1M solution of phosphoramidite in CH3CN in the presence of 5-(ethylthio)-177-tetrazole activator to a solid bound oligonucleotide followed by standard capping, oxidation and deprotection afforded modified oligonucleotides. The 0.1M I2, THF:Pyridine;Water-7:2:l was used as oxidizing agent while DDTT ((dimethylamino-methylidene) amino)-3H-l,2,4-dithiazaoline-3-thione was used as the sulfùr-transfer agent for the synthesis of oligoribonucleotide phosphorothioates. The stepwise coupling effïciency of ail modified phosphoramidites was more than 98%.
Reagents Detailed Description
Deblock Solution 3% Dichloroacetic acid (DCA) in Dichloromethane (DCM)
Amidite Concentration 0.1 M in Anhydrous Acetonitrile
Activator
0.25 M Ethyl-thio-Tetrazole (ETT)
129
Cap-A solution
Cap-B Solution
Oxidizing Solution
Sulfurizing Solution
Acetic anhydride in Pyridine/THF 16% 1-Methylimidazole in THF 0.02M I2, THF: Pyridine; Water-7:2:1
0.2 M DDTT in Pyridine/Acetonitrile 1:1
Cleavage and Deprotection:
Deprotection and cleavage from the solid support was achieved with mixture of ammonia methylamine (1:1, AMA) for 15 min at 65 °C, when the universal linker was used, the deprotection was left for 90 min at 65 °C or solid supports were heated with aqueous ammonia (28%) solution at 55 °C for 16 h to deprotect the base labile protecting groups.
Quantitation of Crude SiNA or Raw Analysis
Samples were dissolved in deionized water (l.OmL) and quantitated as follows: Blanking was first performed with water alone (2 ul) on Nanodrop then Oligo sample reading obtained at 260 nm. The crude material is dried down and stored at -20°C.
Crude HPLC/LC-MS analysis
The 0.1 OD of the crude samples were analyzed for crude HPLC and LC-MS analysis. After Confirming the crude LC-MS data then purification step was performed.
HPLC Purification
The unconjugated and GalNac modified oligonucleotides were purified by anionexchange HPLC. The buffers were 20 mM sodium phosphate in 10 % CH3CN, pH 8.5 (buffer A) and 20 mM sodium phosphate in 10% CH3CN, 1.0 M NaBr, pH 8.5 (buffer B). Fractions containing fiill-length oligonucleotides were pooled.
Desalting of Purified SiNA
The purified dry siNA was then desalted using Sephadex G-25 M (Amersham Biosciences). The cartridge was conditioned with 10 mL of deionized water thrice. Finally, the purified siNA dissolved thoroughly in 2.5mL RNAse free water was applied to the cartridge with very slow drop wise elution. The sait free siNA was eluted with 3.5 ml deionized water directly into a screw cap vial.
IEX HPLC and Electrospray LC/MS Analysis
Approximately 0.10 OD of siNA is dissolved in water and then pipetted in spécial vials for IEX-HPLC and LC/MS analysis. Analytical HPLC and ES LC-MS established the integrity of the compounds.
Duplex Préparation:
130
Single strand oligonucleotides (Sense and Antisense strands) were annealed (1:1 by molar équivalents, heat 90°C for 3 min followed by room température, 20 min) to give the duplex ds-siNA. The final compounds were analyzed on size exclusion chromatography (SEC).
Example 2. ds-siNA Activity
This example investigates the activity of the ds-siNAs synthesized in Example 1.
Homo sapiens HepG2.2.15 cells were cultured in Dulbecco’s Modified Eagle’s Medium (DMEM) (ATCC 30-2002) supplemented to also contain 10% fêtai calf sérum (FCS). Cells were incubated at 37°C in an atmosphère with 5% CO2 in a humidified incubator. For transfection of HepG2.2.15 cells with HBV targeting siRNAs, cells were seeded at a density of 15000 cells/well in 96-well regular tissue culture plates. Transfection of cells was carried out using RNAiMAX (Invitrogen/Life Technologies) according to the manufacturer’s instructions. Dose-response experiments were done with oligo concentrations of 40, 20, 10, 5, 2.5, 1.25, 0.625, 0.3125, 0.15625 and 0.07813nM. For each HBV targeting siRNA treatment (e.g., ds-siRNA, as identified by the ds-siNA ID in Table 6), four wells were transfected in parallel, and individual data points were collected from each well. After 24h of incubation with siRNA, media was removed, and cells were lysed and analyzed with a QuantiGene2.0 branched DNA (bDNA) probe set spécifie for HBV génotype D (also called Hepatitis B virus subtype ayw, complété genome of 3182 basepairs) as présent in cell line HepG2.2.15.
For each well, the HBV on-target mRNA levels were normalized to the GAPDH mRNA level. As shown in Table 6, the activity of the HBV targeting ds-siRNAs was expressed as EC50, 50% réduction of normalized HBV RNA level from no drug control. As shown in Table 6, the cytotoxicity of the HBV targeting ds-siRNAs was expressed by CC50 of 50% réduction of GAPDH mRNA from no drug control.
Example 3. Use of ds-siNAs to treat hepatitis B virus infection
In this example, the ds-siNAs synthesized in Example 1 are used to treat a hepatitis B virus infection in a subject. Generally, a composition comprising a ds-siNA from Table 6 (as identified by the ds-siNA ID) and a pharmaceutically acceptable carrier is administered to the subject suffering from hepatitis B virus. The ds-siNA from Table 6 is conjugated to Nacetylgalactosamine. The ds-siNA is administered at a dose of 0.3 to 5 mg/kg every three weeks by subeutaneous injection or intravenous infusion.
Example 4. ds-siNA Hepatitis B Clinical Trial
In this example, the ds-siNAs from Tables 6A and 6B (as identified by the ds-siNA ID) will be evaluated for safety and efficacy in healthy volunteers and chronic hepatitis B patients.
131 ds-siNAs are being developed for the treatment of chronic hepatitis B (CHB) in adults. The study will be conducted in 3 parts, a single ascending-dose (SAD) phase in healthy volunteers (Group A), a single-dose (SD) phase in patients with CHB (Group B), and a multiple ascending-dose (MAD) phase in patients with CHB (Group C).
Study Design
Study Type : Interventional (Clinical Trial)
Estimated Enrollment : 50 participants
Allocation: Randomized
Intervention Model: Sequential Assignment
Intervention Model Description: Progression from the SAD phase to the first cohort in the MAD phase is contingent upon the Safety Review Committee (SRC) review of a minimum of 14 days post-dose safety and tolerability data from ail HV in at least the first 2 SAD cohorts. The SRC will select one (or more) well-tolerated dose(s) from the SAD phase for administration in the SD and MAD phases. In ail study phases, dosing will be staggered with the use of sentinel participants to allow time for the assessment of safety before additional subjects are exposed to study drug.
Masking: Triple (Participant, Care Provider, Investigator)
Masking Description: This is a double-blind placebo-controlled study in which the study site team, the Sponsor, and the participants will be blinded to treatment assignment. The unblinded pharmacist will cover each syringe, prior to transport to the bedside, to ensure blinding. Participants will be centrally assigned to randomized study intervention using an Interactive Voice/Web Response System (IVRS/IWRS).
Primary Purpose: Treatment
Arms and Interventions
Arm Intervention/ treatment
Experimental: Cohort Al ds-siNA Drug: ds-siNA
132
Arm Intervention/treatment
Single dose, Subcutaneous injection of O.lmg/kg of ds-siNA (HV) ds-siNA is a synthetic ribonucleic acid interférence (RNAi) drug that consists of double-stranded oligonucleotides conjugated to an N-acetyl-Dgalactosamine (GalNAc) ligand. ds-siNA, stérile solution of the ds-siNA at a concentration of 185 mg/mL in water for injection (WFI).
Placebo Comparator: Cohort Al Placebo Single dose, Subcutaneous injection of O.lmg/kg of Placebo for ds-siNA (HV) Drug: Placebo for ds-siNA Stérile 9% saline for injection. Other Name: Placebo
Experimental: Cohort A2 ds-siNA Single dose, Subcutaneous injection of 1.5mg/kg of ds-siNA (HV) Drug: ds-siNA ds-siNA is a synthetic ribonucleic acid interférence (RNAi) drug that consists of double-stranded oligonucleotides conjugated to an N-acetyl-Dgalactosamine (GalNAc) ligand. ds-siNA, stérile solution of the ds-siNA at a concentration of 185 mg/mL in water for injection (WFI).
Placebo Comparator: Cohort A2 Placebo Single dose, Subcutaneous injection of 1.5mg/kg of Placebo for ds-siNA (HV) Drug: Placebo for ds-siNA Stérile 9% saline for injection. Other Name: Placebo
Experimental: Cohort A3 ds-siNA Single dose, Subcutaneous injection of 3mg/kg of ds-siNA (HV) Drug: ds-siNA ds-siNA is a synthetic ribonucleic acid interférence (RNAi) drug that consists of double-stranded oligonucleotides conjugated to an N-acetyl-Dgalactosamine (GalNAc) ligand. ds-siNA, stérile solution of the ds-siNA at a concentration of 185 mg/mL in water for injection (WFI).
Placebo Comparator: Cohort A3 Placebo Single dose, Subcutaneous injection of 3mg/kg of Placebo for ds-siNA (HV) Drug: Placebo for ds-siNA Stérile 9% saline for injection. Other Name: Placebo
133
Arm Intervention/treatment
Experimental: Cohort A4 ds-siNA Single dose, Subcutaneous injection of 6mg/kg of ds-siNA (HV) Drug: ds-siNA ds-siNA is a synthetic ribonucleic acid interférence (RNAi) drug that consists of double-stranded oligonucleotides conjugated to an N-acetyl-Dgalactosamine (GalNAc) ligand. ds-siNA, stérile solution of the ds-siNA at a concentration of 185 mg/mL in water for injection (WFI).
Placebo Comparator: Cohort A4 Placebo Single dose, Subcutaneous injection of 6mg/kg of Placebo for ds-siNA (HV) Drug: Placebo for ds-siNA Stérile 9% saline for injection. Other Name: Placebo
Experimental: Cohort A5 ds-siNA Single dose, Subcutaneous injection of 12mg/kg of ds-siNA (HV) Drug: ds-siNA ds-siNA is a synthetic ribonucleic acid interférence (RNAi) drug that consists of double-stranded oligonucleotides conjugated to an N-acetyl-Dgalactosamine (GalNAc) ligand. ds-siNA, stérile solution of the ds-siNA at a concentration of 185 mg/mL in water for injection (WFI).
Placebo Comparator: Cohort A5 Placebo Single dose, Subcutaneous injection of 12mg/kg of Placebo for ds-siNA (HV) Drug: Placebo for ds-siNA Stérile 9% saline for injection. Other Name: Placebo
Experimental: Cohort B ds-siNA Single dose, Subcutaneous injection of 3mg/kg of for ds-siNA (NUC naïve, CHB) Drug: ds-siNA ds-siNA is a synthetic ribonucleic acid interférence (RNAi) drug that consists of double-stranded oligonucleotides conjugated to an N-acetyl-Dgalactosamine (GalNAc) ligand. ds-siNA, stérile solution of the ds-siNA at a concentration of 185 mg/mL in water for injection (WFI).
Placebo Comparator: Cohort B Placebo Drug: Placebo for ds-siNA Stérile 9% saline for injection. Other Name: Placebo
134
Arm Intervention/treatment
Single dose, Subcutaneous injection of 3mg/kg of Placebo for ds-siNA (NUC naïve, CHB)
Experimental: Cohort Cl ds-siNA 4 doses- Subcutaneous injection of 1.5mg/kg of ds-siNA administered every 28 days (NUC experienced, CHB) Drug: ds-siNA ds-siNA is a synthetic ribonucleic acid interférence (RNAi) drug that consists of double-stranded oligonucleotides conjugated to an N-acetyl-Dgalactosamine (GalNAc) ligand. ds-siNA, stérile solution of the ds-siNA at a concentration of 185 mg/mL in water for injection (WFI).
Placebo Comparator: Cohort Cl Placebo 4 doses- Subcutaneous injection of 1.5mg/kg of Placebo for ds-siNA administered every 28 days (NUC experienced, CHB) Drug: Placebo for ds-siNA Stérile 9% saline for injection. Other Name: Placebo
Experimental: Cohort C2 ds-siNA 4 doses- Subcutaneous injection of 3mg/kg of ds-siNA administered every 28 days (NUC experienced, CHB) Drug: ds-siNA ds-siNA is a synthetic ribonucleic acid interférence (RNAi) drug that consists of double-stranded oligonucleotides conjugated to an N-acetyl-Dgalactosamine (GalNAc) ligand. ds-siNA, stérile solution of the ds-siNA at a concentration of 185 mg/mL in water for injection (WFI).
Placebo Comparator: Cohort C2 Placebo 4 doses- Subcutaneous injection of 3mg/kg of Placebo for ds-siNA administered every 28 days (NUC experienced, CHB) Drug: Placebo for ds-siNA Stérile 9% saline for injection. Other Name: Placebo
Experimental: Cohort C3 ds-siNA Drug: ds-siNA ds-siNA is a synthetic ribonucleic acid interférence (RNAi) drug that consists of double-stranded
135
Arm Intervention/treatment
4 doses- Subcutaneous injection of 6mg/kg of ds-siNA administered every 28 days (NUC experienced, CHB) oligonucleotides conjugated to an N-acetyl-Dgalactosamine (GalNAc) ligand. ds-siNA, stérile solution of the ds-siNA at a concentration of 185 mg/mL in water for injection (WFI).
Placebo Comparator: Cohort C3 Placebo 4 doses- Subcutaneous injection of 6mg/kg of Placebo for ds-siNA administered every 28 days (NUC experienced, CHB) Drug: Placebo for ds-siNA Stérile 9% saline for injection. Other Name: Placebo
Outcome Measures
Primary Outcome Measures :
Number of healthy volunteers with Adverse Events as assessed by CTCAE v5.0 [Time rame: 4 weeks]
Number of participants with abnormalities in vital signs, electrocardiogram (ECG), and clinically significant laboratory fmdings
Number participants with non-cirrhotic chronic Hepatitis B with Adverse Events as assessed by CTCAE v5.0 [Time Frame: 16 weeks]
Number of participants with abnormalities in vital signs, electrocardiogram (ECG), and clinically significant laboratory fmdings.
Secondary Outcome Measures :
To characterize the pharmacokinetics of ds-siNA in healthy volunteers by monitoring plasma pharmacokinetics profiles of [Time Frame: 4 weeks] Measure the amount of ds-siNA excreted in urine
To characterize the pharmacokinetics of ds-siNA in healthy volunteers by monitoring through concentrations of [Time Frame: 4 weeks]
Measure the amount of ds-siNA rénal clearance (CLR).
To characterize the pharmacokinetics of ds-siNA in participants with non-cirrhotic CHB by monitoring plasma pharmacokinetics profiles of ds-siNA. [Time Frame: 12 weeks]
Measure the amount of ds-siNA excreted in urine
To characterize the pharmacokinetics of ds-siNA in participants with non-cirrhotic CHB by monitoring through concentrations of ds-siNA. [Time Frame: 12 weeks]
Measure ds-siNA rénal clearance (CLR).
136
Other Outcome Measures:
To evaluate the preliminary antiviral efficacy of ds-siNA in participants with CHB by monitoring changes in sérum HBsAg levels (ail Group B and C participants) during and after single dose and 12 weeks of treatment with DCR HBVS. [Time Frame: 12 weeks]
Proportion of participants achieving at least a 1-log réduction in HBsAg and achieving a HBsAg level <100 lU/mL at last scheduled visit Time to HBsAg loss (Kaplan-Mayer) Time to anti-HBs séroconversion
To evaluate the preliminary antiviral efficacy of ds-siNA in participants with CHB by monitoring HBeAg levels (HBeAg+ participants only) during and after single dose and 12 weeks of treatment with DCR HBVS. [Time Frame: 12 weeks] % of participants with HBeAg loss and anti HBe at last scheduled visit (if HBeAg positive at study entry)
To evaluate the preliminary antiviral efficacy of ds-siNA in participants with CHB by monitoring HBV DNA levels (ail Group B and C participants) during and after single dose and 12 weeks of treatment with DCR HBVS. [Time Frame: 12 weeks]
Proportion of participants achieving HBV DNA < 2000 HJ/mL (if > 2,000 lU/mL at Baseline); and proportion of participants achieving PCR-nondetectable HBV DNA (if HBV DNA was détectable at Baseline).
To characterize the pharmacodynamies (PD) of ds-siNA on plasma levels of HBsAg and HBV inblood. [Time Frame: 12 weeks]
Track post-treatment duration of any observed efficacy effects.
Eligibility Criteria .
Ages Eligible for Study: 18 Years to 65 Years (Adult, Older Adult)
Sexes Eligible for Study: AU
Accepts Healthy Volunteers: Yes
Inclusion Criteria:
Healthy at the time of screening as determined by medical évaluation.
Capable of giving informed consent.
12-lead ECG within normal limits or with no clinically significant abnormalities.
Négative screen for alcohol or drugs of abuse.
Non-smokers for at least 3 months with a négative urinary cotinine concentration at screening.
BMI within range 18.0-32.0 kg/m2 (inclusive).
137
Female participants not prégnant, not breastfeeding, and not of childbearing potential or willing to follow contraceptive guidance.
Chronic hepatitis B infection (Group B and C only).
Clinical history compatible with compensated liver disease with no evidence of cirrhosis (Group B and C only).
Continuously on nucléotides (NUC) therapy for at least 12 weeks prior to screening (Group C only).
Exclusion Criteria:
History of any medical condition that may interfère with the absorption, distribution, or élimination of study drug.
Poorly controlled or unstable hypertension.
History of diabètes mellitus treated with insulin or hypoglycémie agents.
History of asthma requiring hospital admission within the preceding 12 months.
Evidence of G-6-PD deficiency.
Currently poorly controlled endocrine conditions, excluding thyroid conditions.
History of multiple drug allergies or history of allergie reaction to an oligonucleotide or GalNAc.
Clinically relevant surgical history.
Use of prescription médications (excluding contraception for women) within 4 weeks prior to the administration of study intervention.
Use of clinically relevant over-the-counter médication or suppléments (excluding routine vitamins) within 7 days of first dosing.
Has received an investigational agent within the 3 months prior to dosing or is in followup of another study.
Antiviral therapy (other than entecavir or tenofovir) within 3 months of screening or treatment with interferon in the last 3 years (Group B and C only).
Use within the last 6 months of anticoagulants or systemically administered corticosteroids, immunomodulators, or immunosuppressants (Group B and C only).
Example 5: Synthesis of 5’ End Cap Monomer
2 3
138
Oxone/MeOH. FLO
Example 5 monomer
Example 5 Monomer Synthesis Scheme
Préparation of (2): To a solution of 1 (15 g, 57.90 mmol) in DMF (150 mL) were added AcSK (11.24 g, 98.43 mmol) and TBAI (1.07 g, 2.89 mmol), and the mixture was stirred at 25 °C for 12 h. Upon completion as monitored by LCMS, the mixture was diluted with H2O (10 mL) and extracted with EA (200 mL * 3). The combined organic layers were washed with brine (200 mL * 3), dried over anhydrous NaiSCL, filtered and concentrated under reduced pressure to give 2 (14.5 g, 96.52% yield, 98% purity) as a colorless oil. ESI-LCMS: 254.28 [M+H]+; ‘H NMR (400 MHz, CDCI3) δ = 4.78 - 4.65 (m, 2H), 3.19 (d, .7=14.1 Hz, 2H), 2.38 (s, 3H), 1.32 (t, .7=6.7 Hz, 12H); 31P NMR (162 MHz, CDCI3) δ = 20.59.
Préparation of (3): To a solution of 2 (14.5 g, 57.02 mmol) in CH3CN (50 mL) and MeOH (25 mL) was added NaOH (3 M, 28.51 mL), and the mixture was stirred at 25 °C for 12 h under Ar. Upon completion as monitored by TLC, the reaction mixture was concentrated under reduced pressure to remove CH3CN and CH3OH. The residue was diluted with water (50 mL) and adjust pH=7 by 6M HCl, and the mixture was extracted with EA (50 mL * 3). The combined organic layers were washed with brine (50 mL * 3), dried over anhydrous Na2SÛ4, filtered and concentrated under reduced pressure to give 3 (12.1 g, crude) as a colorless oil.
139 .
Préparation of (4): To a solution of 3 (12.1 g, 57.01 mmol) in CH3CN (25 mL) and MeOH (25 mL) was added A (14.77 g, 57.01 mmol) dropwise at 25 °C, and the mixture was stirred at 25 °C under Ar for 12 h. Upon completion as monitored by LCMS, the reaction mixture was concentrated under reduced pressure to give 4 (19.5 g, 78.85% yield) as a colorless oil. Ή NMR (400 MHz, CDCI3) δ = 4.80 - 4.66 (m, 4H), 2.93 (d, J=\ 1.3 Hz, 4H), 1.31 (dd, >3.9, 6.1 Hz, 24H); 31P NMR (162 MHz, CDCI3) δ = 22.18.
Préparation of (5): To a solution of 4 (19.5 g, 49.95 mmol) in MeOH (100 mL) and H2O (100 mL) was added Oxone (61.41 g, 99.89 mmol) at 25 °C in portions, and the mixture was stirred at 25 °C for 12 h under Ar. Upon completion as monitored by LCMS, the reaction mixture was filtered, and the filtrate was concentrated under reduced pressure to remove MeOH. The residue was extracted with EA (50 mL *3). The combined organic layers were washed with brine (50 mL * 3), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure to give a residue. The crude product was triturated with i-Pr2O and n-Hexane (1:2, 100 mL) at 25 °C for 30 min to give 5 (15.6 g, 73.94% yield,) as a white solid. *H NMR (400 MHz, CDCI3) δ = 4.92 - 4.76 (m, 4H), 4.09 (d, >16.1 Hz, 4H), 1.37 (dd, >3.5, 6.3 Hz, 24H); 3IP NMR (162 MHz, CDCI3) δ = 10.17.
Préparation of (7): To a mixture of 5 (6.84 g, 16.20 mmol) in THF (20 mL) was added LiBr (937.67 mg, 10.80 mmol) until dissolved, followed by DIEA (1.40 g, 10.80 mmol, 1.88 mL) under argon at 15 °C. The mixture was stirred at 15 °C for 15 min. 6 (4 g, 10.80 mmol) were added. The mixture was stirred at 15 °C for 3 h. Upon completion as monitored by LCMS, the reaction mixture was quenched by addition of H2O (40 mL) and extracted with EA (40 mL * 3). The combined organic layers were washed with brine (100 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash reverse-phase chromatography (120 g C-18 Column, Eluent of 0-60% ACN/H2O gradient @ 80 mL/min) to give 7 (5.7 g, 61.95% yield) as a colorless oil. ESI-LCMS: 611.2 [M+H]+ ; *H NMR (400 MHz, CDCI3); δ = 9.26 (s, 1H), 7.50 (d, >8.1 Hz, 1H), 7.01 (s, 2H), 5.95 (d, >2.7 Hz, 1H), 5.80 (dd, >2.1, 8.2 Hz, 1H), 4.89 - 4.72 (m, 2H), 4.66 (d, >7.2 Hz, 1H), 4.09 - 4.04 (m, 1H), 3.77 (dd, >2.7, 4.9 Hz, 1H), 3.62 (d, >3.1 Hz, 1H), 3.58 (d, >3.1 Hz, 1H), 3.52 (s, 3H), 1.36 (td, >1.7, 6.1 Hz, 12H), 0.92 (s, 9H), 0.12 (s, 6H); 31P NMR (162 MHz, CDCh) δ = 9.02
Préparation of (8): To a mixture of 7 (5.4 g, 8.84 mmol) in THF (80 mL) was added Pd/C (5.4 g, 10% purity) under N2. The suspension was degassed under vacuum and purged with H2 several times. The mixture was stirred under H2 (15 psi) at 20 °C for 1 hr. Upon completion as monitored by LCMS, the reaction mixture was filtered, and the filtrate was concentrated to give 8 (5.12 g, 94.5% yield) as a white solid. ESI-LCMS: 613.3 [M+H]+ H NMR (400 MHz,
140
CD3CN) δ = 9.31 (s, 1H), 7.37 (d, 7=8.0 Hz, 1H), 5.80 - 5.69 (m, 2H), 4.87 - 4.75 (m, 2H), 4.11 4.00 (m, 1H), 3.93 - 3.85 (m, 1H), 3.80 - 3.74 (m, 1H), 3.66 - 3.60 (m, 1H), 3.57 - 3.52 (m, 1H), 3.49 (s, 3H), 3.46 - 3.38 (m, 1H), 2.35 - 2.24 (m, 1H), 2.16 - 2.03 (m, 1H), 1.89 - 1.80 (m, 1H), 1.37 - 1.34 (m, 12H), 0.90 (s, 9H), 0.09 (s, 6H); 3lP NMR (162 MHz, CD3CN) δ = 9.41.
Préparation of (9): To a solution of 8 (4.4 g, 7.18 mmol) in THF (7.2 mL) was added TBAF (1 M, 7.18 mL), and the mixture was stirred at 20 °C for 1 hr. Upon completion as monitored by LCMS, the reaction mixture was diluted with H2O (50 mL) and extracted with EA (50 mL*4). The combined organic layers were washed with brine (50 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (ISCO®; 40 g SepaFlash® Silica Flash Column, Eluent of 0~5%, MeOH/DCM gradient @ 40 mL/min) to give 9 (3.2 g, 88.50% yield) as a white solid. ESI-LCMS: 499.2 [M+H]+ ‘H NMR (400 MHz, CD3CN) δ = 9.21 (s, 1H), 7.36 (d, 7=8.3 Hz, 1H), 5.81 - 5.72 (m, 2H), 4.88 - 4.74 (m, 2H), 3.99 - 3.87 (m, 2H), 3.84 (dd, 7=1.9, 5.4 Hz, 1H), 3.66 - 3.47 (m, 7H), 2.98 (s, 1H), 2.44 - 2.15 (m, 2H), 1.36 (d, 7=6.0 Hz, 12H); 31P NMR (162 MHz, CD3CN) δ = 9.48.
Préparation of (Example 5 monomer): To a mixture of 9 (3.4 g, 6.82 mmol, 1 eq) and 4A MS (3.4 g) in MeCN (50 mL) was added PI (2.67 g, 8.87 mmol, 2.82 mL, 1.3 eq) at 0 °C, followed by addition of lH-imidazole-4,5-dicarbonitrile (886.05 mg, 7.50 mmol) at 0 °C. The mixture was stirred at 20 °C for 2 h. Upon completion as monitored by LCMS, the reaction mixture was quenched by addition of saturated aq. NaHCO3 (50 mL) and diluted with DCM (100 mL). The organic layer was washed with saturated aq. NaHCO3 (50 mL * 2), dried over Na2SC>4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC: column: YMC-Triart Prep C18 250*50 mm*10um; mobile phase: [water (10 mM NH4HCO3)-ACN]; B%: 15% to give a impure product. The impure product was fùrther purified by a flash silica gel column (0% to 5% i-PrOH in DCM with 0.5% TEA) to give Example 5 monomer (2.1 g, 43.18% yield) as a white solid. ESI-LCMS: 721.2 [M+Na]+;H NMR (400 MHz, CD3CN) δ = 9.29 (s,lH), 7.45 (d, 7=8.1 Hz, 1H), 5.81 (d, 7=4.2 Hz, 1H), 5.65 (d, 7=8.1 Hz, 1H), 4.79 - 4.67 (m, 2H), 4.26 - 4.05 (m, 2H), 4.00 - 3.94 (m, 1H), 3.89 - 3.63 (m, 6H), 3.53 - 3.33 (m, 5H), 2.77 - 2.61 (m, 2H), 2.31 - 2.21 (m, 1H), 2.16 - 2.07 (m, 1H), 1.33 - 1.28 (m, 12H), 1.22- 1.16 (m, 1H), 1.22-1.16 (m, 11H); 31P NMR (162 MHz, CD3CN) δ = 149.89, 149.78, 10.07, 10.02.
Example 6. Synthesis of 5’ End Cap Monomer
141
Example 6 Monomer
Example 6 Monomer Synthesis Scheme
Préparation of (2): To a solution of 1 (5 g, 13.42 mmol) in DMF (50 mL) were added PPI13 (4.58 g, 17.45 mmol) and 2-hydroxyisoindoline-l,3-dione (2.85 g, 17.45 mmol), followed by a solution of DIAD (4. 07 g, 20. 13 mmol, 3.91 mL) in DMF (10 mL) dropwise at 15°C. The resulting solution was stirred at 15°C for 18 hr. The reaction mixture was then diluted with DCM (50 mL), washed with H2O (60 mL*3) and brine (30 mL), dried over NazSCU, fïltered and evaporated to give a residue. The residue was then triturated with EtOH (55 mL) for 30 min, and the collected white powder was washed with EtOH (10 mL*2) and dried to give 2 (12.2 g, 85. 16% yield) as a white powder (the reaction was set up in two batches and combined) ESI-LCMS: 518.1 [M+H]+.
Préparation of (3): 2 (6 g, 11.59 mmol) was suspended in MeOH (50 mL), and then NH2NH2.H2O (3.48 g, 34. 74 mmol, 3.38 mL, 50% purity) was added dropwise at 20°C. The reaction mixture was stirred at 20°C for 4 hr. Upon completion, the reaction mixture was diluted with EA (20 mL) and washed with NaHCOs (10 mL*2) and brine (10 mL). The combined organic layers were then dried over Na2SO4, fïltered and evaporated to give 3 (8.3 g, 92.5% yield) as a white powder. (The reaction was set up in two batches and combined). ESI-LCMS: 388.0 [M+H]+ ; Ή NMR (400MHz, DMSO-d6) δ =11.39 (br s, 1H), 7.72 (d, .7=8.1 Hz, 1H), 6.24 6.09 (m, 2H), 5.80 (d,.7=4.9 Hz, 1H), 5.67 (d, .7=8.1 Ηζ,ΙΗ), 4.26 (t, .7=4.9 Hz, 1H), 4.03 -3.89 (m, 1H), 3.87 - 3.66 (m, 3H),3.33 (s, 3H), 0.88 (s, 9H), 0.09 (d, J=1.3 Hz, 6H)
Préparation of (4): To a solution of 3 (7 g, 18.06 mmol) and Py (1.43 g, 18.06 mmol, 1.46 mL) in DCM (130 mL) was added a solution of MsCl (2.48 g, 21.68 mmol, 1. 68 mL) in DCM (50 mL) dropwise at -78°C under N2. The reaction mixture was allowed to warm to 15°C in 30 min and stirred at 15°C for 3 h. The réaction mixture was quenched by addition of ice
142 water (70 mL) at 0°C, and then extracted with DCM (50 mL * 3). The combined organic layers were washed with saturated aq. NaHCO3 (50 mL) and brine (30 mL), dried over Na2SÜ4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (ISCO®; 30 g SepaFlash® Silica Flash Column, Eluent of 0~20% iPrOH/DCM gradient @ 30 mL/min to give 4 (6.9 g, 77.94% yield) as a white solid. ESLLCMS: 466.1 [M+H]+ ; 'H NMR (400MHz, DMSO-d6) δ = 11.41 (br s, 1H), 10. 15 (s, 1H), 7. 69 (d, J=8.1 Hz, 1H), 5.80 (d, J=4.4 Hz, 1H), 5.65 (d, J=8. 1 Hz, 1H), 4.24 (t, J=5.2 Hz, 1H), 4.16 3.98 (m, 3H), 3. 87 (t, J=4.8 Hz, 1H), 3.00 (s, 3H), 2.07 (s, 3H), 0.88 (s, 9H), 0. 10 (d, J=1.5 Hz, 6H)
Préparation of (5): To a solution of 4 (6.9 g, 14.82 mmol) in THF (70 mL) was added TBAF (1 M, 16.30 mL) at 15°C. The reaction mixture was stirred at 15°C for 18 hr, and then evaporated to give a residue. The residue was purified by flash silica gel chromatography (ISCO®; 24 g SepaFlash® Silica Flash Column, Eluent of 0~9% MeOH/Ethyl acetate gradient @ 30 mL/min) to give 5 (1.8 g, 50.8% yield) as a white solid. ESI-LCMS: 352.0 [M+H]+; Ή NMR (400MHz, DMSO-d6) δ = 11.40 (s, 1H), 10.13 (s, 1H), 7.66 (d, 7=8.1 Hz, 1H), 5.83 (d, 7=4. 9 Hz, 1H), 5.65 (dd, 7=1. 8, 8. 1 Hz, 1H), 5.36 (d, 7=6. 2 Hz, 1H), 4.13 - 4.00 (m, 4H), 3. 82 (t, 7=5.1 Hz, 1H), 3.36 (s, 3H), 3.00 (s, 3H)
Préparation of (Example 6 monomer): To a mixture of 5 (3 g, 8.54 mmol) and DIEA (2.21 g, 17.08 mmol, 2.97 mL) in ACN (90 mL) was added P2 (3.03 g, 12.81 mmol) dropwise at 15°C. The reaction mixture was stirred at 15°C for 5 h. Upon completion, the reaction mixture was diluted with EA (40 mL) and quenched with 5% NaHCO3 (20 mL). The organic layer was washed with brine (30 mL), dried over Na2SO4, filtered and evaporated to give a residue. The residue was purified by flash silica gel chromatography (ISCO®; 12 g SepaFlash® Silica Flash Column, Eluent of 0—15% i-PrOH/(DCM with 2% TEA) gradient @ 20 mL/min) to Example 6 monomer (2.1 g, 43.93% yield) as a white solid. ESI-LCMS: 552.3 [M+H]+; ‘H NMR (400 MHz, CD3CN) δ = 8.78 (br s, 1H), 7.57 (dd, J=4.6, 8.2 Hz, 1H), 5.97 - 5.80 (m, 1H), 5.67 (d, J=8. 3Hz, 1H), 4.46 - 4.11 (m, 4H), 3.95 -3.58 (m, 5H), 3.44 (d, J=16. 3 Hz, 3H), 3.02 (d, J=7. 5 Hz, 3H), 2. 73 -2.59 (m, 2H), 1.23 - 1.15 (m, 12H); 31P NMR (162 MHz, CD3CN) δ = 150.30, 150.10
Example 7: Synthesis of 5’ End Cap Monomer
143
TBAF
Example 7 Monomer
Example 7 Monomer Synthesis Scheme
Préparation of (2): To the solution of 1 (5 g, 12.90 mmol) and TEA (1.57 g, 15.48 mmol, 2.16 mL) in DCM (50 mL) was added P-4 (2.24 g, 15.48 mmol, 1.67 mL) in DCM (10 mL) dropwise at 15°C under N2. The reaction mixture was stirred at 15°C for 3 h. Upon completion as monitored by LCMS and TLC (PE: EtOAc = 0:1), the reaction mixture was concentrated to dryness, diluted with H2O (20 mL), and extracted with EA (50 mL*3). The combined organic layers were washed with brine (30 mL*3), dried over anhydrous Na2SC>4, filtered, and the filtrate was concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (ISCO®; 40 g SepaFlash® Silica Flash Column, Eluent of 0-95% Ethyl acetate/Petroleum ether gradient @ 60 mL/min) to give 2 (5.3 g, 71.3% yield) as a white solid. ESI-LCMS: 496.1 [M+H]+ ;H NMR (400 MHz, CDC13) δ= 0.10 (d, J=4.02 Hz, 6 H) 0.91 (s, 9 H) 3.42 - 3.54 (m, 3 H) 3.65 - 3.70 (m, 1 H) 3.76 - 3.89 (m, 6 H) 4.00 (dd, J=10.92, 2.89 Hz, 1 H) 4.08 - 4.13 (m, 1 H) 4.15 - 4.23 (m, 2 H) 5.73 (dd, J=8.28, 2.01 Hz, 1 H) 5.84 (d, J=2.76 Hz, 1 H) 6.86 (d, 1=15.81 Hz, 1 H) 7.72 (d, J=8.03 Hz, 1 H) 9.10 (s, 1 H); 31P NMR (162 MHz, CD3CN) δ = 9.65
Préparation of (3): To a solution of 2 (8.3 g, 16.75 mmol) in THF (50 mL) were added TBAF (1 M, 16.75 mL) and CH3COOH (1.01 g, 16.75 mmol, 957.95 uL). The mixture was stirred at 20 °C for 12 hr. Upon completion as monitored by LCMS, the reaction mixture was concentrated under reduced pressure. The residue was purified by column chromatography (SiO2, PE: EA = 0-100%; MeOH /EA= 0-10%) to give 3 (5 g, 77.51% yield) as a white solid.
144
ESI-LCMS: 382.1 [M+H]+; [H NMR (400 MHz, CDC13) δ= 3.35 (s, 3 H) 3.65 (br d, 7=2.76 Hz, 3 H) 3.68 (d, 7=2.76 Hz, 3 H) 3.77 (t, 7=5.08 Hz, 1 H) 3.84 - 4.10 (m, 4 H) 5.33 (br d, 7=5.52 Hz, 1 H) 5.62 (d, 7=7.77 Hz, 1 H) 5.83 (d, 7=4.94 Hz, 1 H) 7.69 (d, 7=7.71 Hz, 1 H) 9.08 (d, 7=16.81 Hz, 1 H) 11.39(brs, 1 H);3‘P NMR (162 MHz, CD3CN) δ = 15.41
Préparation of (Example 7 monomer): To a solution of 3 (2 g, 5.25 mmol) and DIPEA (2.03 g, 15.74 mmol, 2.74 mL, 3 eq) in MeCN (21 mL) and pyridine (7 mL) was added P2 (1.86 g, 7.87 mmol) dropwise at 20 °C, and the mixture was stirred at 20 °C for 3 hr. Upon completion as monitored by LCMS, the reaction mixture was diluted with water (20 mL) and extracted with EA (50 mL). The combined organic layers were washed with brine (30 mL), dried over anhydrous Na2SO4, filtered, and the filtrate was concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (ISCO®; 25 g SepaFlash® Silica Flash Column, Eluent of 0-45% (Ethyl acetate: EtOH=4:l)/Petroleum ether gradient) to give Example 7 monomer (1.2 g, 38.2% yield) as a white solid. ESI-LCMS: 604.1 [M+H]+; Ή NMR (400 MHz, CD3CN) δ= 1.12 - 1.24 (m, 12 H) 2.61 - 2.77 (m, 2 H) 3.43 (d, J=17.64 Hz, 3 H) 3.59 - 3.69 (m, 2 H) 3.71 - 3.78 (m, 6 H) 3.79 - 4.14 (m, 5 H) 4.16 - 4.28 (m, 1 H) 4.29 - 4.42 (m, 1 H) 5.59 - 5.72 (m, 1 H) 5.89 (t, J=4.53 Hz, 1 H) 7.48 (br d, J=12.76 Hz, 1 H) 7.62 - 7.74 (m, 1 H) 9.26 (br s, 1 H); 3IP NMR (162 MHz, CD3CN) δ = 150.57, 149.96, 9.87
Example 8: Synthesis of 5’ End Cap Monomer
I,. PI13P. CHjCN
HO'' 'OCH;
DMTrCl, AgNO3. trimethylpyridine
Example 8 Monomer
Example 8 Monomer Synthesis Scheme
Préparation of (2): To a solution of 1 (30 g, 101.07 mmol, 87% purity) in CH3CN (1.2 L) and Py (60 mL) were added I2 (33.35 g, 131.40 mmol, 26.47 mL) and PPI13 (37.11 g, 141.50 mmol) in one portion at 10 °C. The reaction was stirred at 25 °C for 48 h. Upon completion, the mixture was diluted with saturated aq.Na2S2O3 (300 mL) and saturated aq.NaHCCh (300 mL), concentrated to remove CH3CN, and extracted with EtOAc (300 mL * 3).
145
The combined organic layers were washed with brine (300 mL), dried over Na2SÛ4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (ISCO®; 330 g SepaFlash® Silica Flash Column, Eluent of 0~60% Methanol/Dichloromethane gradient @100 mL/min) to give 2 (28.2 g, 72 % yield) as abrown solid. ESI-LCMS: 369.1 [M+H]+ ;H NMR (400 MHz, DMSO-d6) δ = 11.43 (s, 1H), 7.68 (d, 7=8.1 Hz, 1H), 5.86 (d, 7=5.5 Hz, 1H), 5.69 (d, 7=8.1 Hz, 1H), 5.46 (d, .7=6.0 Hz, 1H), 4.08 3.96 (m, 2H), 3.90 - 3.81 (m, 1H), 3.60 - 3.51 (m, 1H), 3.40 (dd, 7=6.9, 10.6 Hz, 1H), 3.34 (s, 3H).
Préparation of (3): To the solution of 2 (12 g, 32.6 mmol) in DCM (150 mL) were added AgNCh (11.07 g, 65.20 mmol), 2,4,6-trimethylpyridine (11.85 g, 97.79 mmol, 12.92 mL), and DMTC1 (22.09 g, 65.20 mmol) at 10 °C, and the reaction mixture was stirred at 10 °C for 16 hr. Upon completion, the mixture was filtered and the filtrate was concentrated under reduced pressure. The residue was purified by flash silica gel chromatography (ISCO®; 120 g SepaFlash® Silica Flash Column, Eluent of 0-50% Ethyl acetate/Petroleum ethergradient @ 60 mL/min) to give 3 (17 g, 70.78% yield) as a yellow solid. ESI-LCMS: 693.1 [M+Na]+ [; H NMR (400 MHz, DMSO-dô) δ = 11.46 (s, 1H), 7.60 (d, 7=8.4 Hz, 1H), 7.49 (d, 7=7.2 Hz, 2H), 7.40 7.30 (m, 6H), 7.29 - 7.23 (m, 1H), 6.93 (d, 7=8.8 Hz, 4H), 5.97 (d, 7=6.0 Hz, 1H), 5.69 (d, 7=8.0 Hz, 1H), 4.05 - 4.02 (m, 1H), 3.75 (d, J=1.2 Hz, 6H), 3.57 (t, 7=5.6 Hz, 1H), 3.27 (s, 4H), 3.06 (t, 7=10.4 Hz, 1H), 2.98 - 2.89 (m, 1H).
Préparation of (4): To a solution of 3 (17 g, 25.35 mmol) in DMF (200 mL) was added AcSK (11.58 g, 101.42 mmol) at 25 °C, and the reaction was stirred at 60 °C for 2 hr. The mixture was diluted with H2O (600 mL) and extracted with EtOAc (300 mL * 4). The combined organic layers were washed with brine (300 mL), dried over Na2SO4, filtered, and concentrated under reduced pressure to give 4 (15.6 g, crude) as a brown solid, which was used directly without further purification. ESI-LCMS: 641.3 [M+H]+.
Préparation of (5): To a solution of 4 (15.6 g, 25.21 mmol) in CH3CN (200 mL) were added DTT (11.67 g, 75.64 mmol, 11.22 mL) and LiOH.H2O (1.06 g, 25.21 mmol) at 10 °C under Ar. The reaction was stirred at 10 °C for 1 hr. The mixture was concentrated under reduced pressure to remove CH3CN, and the residue was diluted with H2O (400 mL) and extracted with EtOAc (200 mL * 3). The combined organic layers were washed with brine (300 mL), dried over Na2SÜ4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (ISCO®; 220 g SepaFlash® Silica Flash Column, Eluent of 0-60% Ethyl acetate/Petroleum ether gradient @100 mL/min) to give 5 (8.6 g, 56.78% yield) as a white solid. ESI-LCMS: 599.3 [M+Na]+ ; 'H NMR (400 MHz, DMSO-dô)
146 δ = 8.79 (s, 1Η), 7.61 (d, .7=8.0 Hz, 1H), 7.56 - 7.46 (m, 2H), 7.45 - 7.37 (m, 4H), 7.36 - 7.27 (m, 3H), 6.85 (dd, .7=2.8, 8.8 Hz, 4H), 5.85 (d, .7=1.3 Hz, 1H), 5.68 (dd, .7=2.0, 8.2 Hz, 1H), 4.33 4.29 (m, 1H), 3.91 (dd, .7=4.8, 8.2 Hz, 1H), 3.81 (d, .7=1.6 Hz, 6H), 3.33 (s, 3H), 2.85 - 2.80 (m, 1H), 2.67-2.55 (m, 2H), 1.11 (t, .7=8.8 Hz, 1H).
Préparation of (Example 8 monomer): To a solution of 5 (6 g, 10.40 mmol) in DCM (120 mL) were added PI (4.08 g, 13.53 mmol, 4.30 mL) and DCI (1.35 g, 11.45 mmol) in one portion at 10 °C under Ar. The reaction was stirred at 10 °C for 2 hr. The reaction mixture was diluted with saturated aq.NaHCO3 (50 mL) and extracted with DCM (20 mL * 3). The combined organic layers were washed with brine (30 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC (column: YMC-Triart Prep C18 250*50 mm*10 um; mobile phase: [water(10mM NH4HCO3)-ACN]; B%: 35%-81%,20min) to give Example 8 monomer (3.54 g, 43.36% yield) as a yellow solid. ESILCMS: 776.4 [M+H]+; lH NMR (400 MHz, DMSO-d6) δ = 7.65 - 7.38 (m, 7H), 7.37 - 7.22 (m, 3H), 6.90 ( d, .7=8.4 Hz, 4H), 5.92 ( s, 1H), 5.66 ( t, .7=8.2 Hz, 1H), 4.13 ( d, .7=4.0 Hz, 1H), 4.00
- 3.88 (m, 1H), 3.87 - 3.59 (m, 10H), 3.33 ( d, .7=5.8 Hz, 3H), 3.12 - 2.94 (m,lH), 2.78 - 2.60 (m,
3H), 2.55-2.48 (m, 1H), 1.36 - 0.98 (m, 12H); 31P NMR (162 MHz, DMSO-d6) δ = 162.69.
Example 9: Synthesis of 5’ End Cap Monomer
NHBz O j ho IA0 A ____Oxidation TBSO 1 NHBz N-A'N (/ J j D ΗΟψϋ I NaBD4, CDjOD ~ L-M NHBz NHBz <z I zj <z J A ΗΟΌ MeOH, SOCI2 P TBSO TBSO O^ 2 3 NHBz <Z J J DMTrCI, D νΆν^ pyrldine DMTrO^i D I TBAF -----------------► T ,0-. -----------------►
TBSO 4 NHBz NZ D 'n-v Ta. DMTrO i D 1 /— r OH 6 TBSO Ά 5 NHBz p-i Dcl DMTroAD ? CN NC'^-X P Ar
147
Example 9 Monomer
Example 9 Monomer Synthesis Scheme
Préparation of (2): To a solution of 1 (22.6 g, 45.23 mmol) in DCM (500 mL) and H2O (125 mL) were added TEMPO (6.40 g, 40.71 mmol) and DIB (29.14 g, 90.47 mmol) at 0 °C. The mixture was stirred at 20 °C for 20 h. Upon completion as monitored by LCMS, saturated aq. NaHCO3 was added to the mixture to adjust pH >8. The mixture was diluted with H2O (200 mL) and washed with DCM (100 mL * 3). The aqueous layer was collected, adjusted to pH < 5 by HCl (4M), and extracted with DCM (200 mL * 3). The combined organic layers were washed with brine (300 mL), dried over Na2SO4, filtered, and concentrated under reduced pressure to give 2 (17.5 g, 68.55% yield) as a yellow solid. ESI-LCMS: 514.2 [M+H]+ ; 'H NMR (400 MHz, DMSO-d6) δ = 11.27 (s, 1H), 8.86 (s, 1H), 8.78 (s, 1H), 8.06 (d, 7=7.5 Hz, 2H), 7.68 - 7.62 (m, 1H), 7.59 - 7.52 (m, 2H), 6.28 (d, <7=6.8 Hz, 1H), 4.82 - 4.76 (m, 1H), 4.54 (dd, 7=4.1, 6.7 Hz, 1H), 4.48 (d, <7=1.8 Hz, 1H), 3.32 (s, 3H), 0.94 (s, 9H), 0.18 (d, <7=4.8 Hz, 6H).
Préparation of (3): To a solution of 2 (9.3 g, 18.11 mmol) in MeOH (20 mL) was added SOCI2 (3.23 g, 27.16 mmol, 1.97 mL) dropwise at 0 °C. The mixture was stirred at 20 °C for 0.5 hr. Upon completion as monitored by LCMS, the reaction mixture was quenched by addition of saturated aq. NaHCO3 (80 mL) and concentrated under reduced pressure to remove MeOH. The aqueous layer was extracted with DCM (80 mL * 3). The combined organic layers were washed with brine (200 mL), dried over Na2SÜ4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (ISCO®; 120 g SepaFlash® Silica Flash Column, Eluent of 0~5%, MeOH/DCM gradient @ 85 mL/min) to give 3 (5.8 g, 60 % yield) as a yellow solid. ESI-LCMS: 528.3 [M+H]+ ; Ή NMR (400 MHz, DMSO-d6) δ = 11.28 (s, 1H), 8.79 (d, 7=7.3 Hz, 2H), 8.06 (d, <7=7.5 Hz, 2H), 7.68 - 7.62 (m, 1H), 7.60 - 7.53 (m, 2H), 6.28 (d, J=6.6 Hz, 1H), 4.87 (dd, 7=2.4, 4.0 Hz, 1H), 4.61 (dd, 7=4.3, 6.5 Hz, 1H), 4.57 (d, <7=2.2 Hz, 1H), 3.75 (s, 3H), 3.32 (s, 3H), 0.94 (s, 9H), 0.17 (d, <7=2.2 Hz, 6H).
Préparation of (4): To a mixture of 3 (5.7 g, 10.80 mmol) in CD3OD (120 mL) was added NaBÜ4 (1.63 g, 43.21 mmol) in portions at 0 °C, and the mixture was stirred at 20 °C for 1 hr. Upon completion as monitored by LCMS, the reaction mixture was neutralized by AcOH (~ 10 mL) and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (ISCO®; 40 g SepaFlash® Silica Flash Column, Eluent of 0~5%, MeOH/DCM gradient @ 40 mL/min) to give 4 (4.15 g, 7.61 mmol, 70.45% yield) as a yellow solid. ESI-LCMS: 502.2 [M+H]+; ‘H NMR (400 MHz, DMSO-d6) δ = 11.23 (s, 1H), 8.76 (s, 2H), 8.04 (d, 7=7.3 Hz, 2H), 7.69 - 7.62 (m, 1H), 7.60 - 7.52 (m, 2H), 6.14 (d, 7=6.0 Hz, 1H),
148
5.18 (s, 1H), 4.60 - 4.51 (m, 2H), 3.98 (d, 7=3.0 Hz, 1H), 3.32 (s, 3H), 0.92 (s, 9H), 0.13 (d, 7=1.5 Hz, 6H).
Préparation of (5): To a solution of 4 (4.85 g, 9.67 mmol) in pyridine (50 mL) was added DMTrCl (5.90 g, 17.40 mmol) at 25 °C and the mixture was stirred for 2 hr. Upon completion as monitored by LCMS, the reaction mixture was concentrated under reduced pressure to remove pyridine. The residue was diluted with EtOAc (150 mL) and washed with H?O (50 mL * 3), dried over NazSCri, filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (ISCO®; 80 g SepaFlash® Silica Flash Column, Eluent of 0~70%, EA/PE gradient @ 60 mL/min) to give 5 (6.6 g, 84.06% yield) as a yellow solid. ESI-LCMS: 804.3 [M+H]+, Ή NMR (400 MHz, DMSO-d6) δ = 11.22 (s, 1H), 8.68 (d, 7=11.0 Hz, 2H), 8.03 (d, ./=7.3 Hz, 2H), 7.68 - 7.60 (m, 1H), 7.58 - 7.49 (m, 2H), 7.37 - 7.30 (m, 2H), 7.27 - 7.16 (m, 7H), 6.88 - 6.79 (m, 4H), 6.17 (d, 7=4.2 Hz, 1H), 4.72 (t, 7=5.0 Hz, 1H), 4.60 (t, 7=4.5 Hz, 1H), 4.03 - 3.98 (m, 1H), 3.71 (s, 6H), 0.83 (s, 9H), 0.12 - 0.03 (m, 6H).
Préparation of (6): To a solution of 5 (6.6 g, 8.21 mmol) in THF (16 mL) was added TBAF (1 M, 8.21 mL,), and the mixture was stirred at 20 °C for 2 hr. Upon completion as monitored by LCMS, the reaction mixture was diluted with EA (150 mL) and washed with H2O (50 mL*3). The organic layer was washed with brine (150 mL), dried over NasSCU, filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (ISCO®; 80 g SepaFlash® Silica Flash Column, Eluent of 10- 100%, EA/PE gradient @30 mL/min) to give 6 (5.4 g, 94.4 % yield) as a yellow solid. ESI-LCMS: 690.3 [M+Hf^HNMR (400 MHz, DMSO-d6) δ = 11.24 (s, 1H), 8.69 (s, 1H), 8.62 (s, 1H), 8.05 (d, 7=7.3 Hz, 2H), 7.69 - 7.62 (m, 1H), 7.60 - 7.52 (m, 2H), 7.40 - 7.33 (m, 2H), 7.30 - 7.18 (m, 7H), 6.84 (dd, 7=5.9, 8.9 Hz, 4H), 6.19 (d, 7=4.8 Hz, 1H), 5.36 (d, 7=6.0 Hz, 1H), 4.59 - 4.52 (m, 1H), 4.48 (q, 7=5.1 Hz, 1H), 4.11 (d, 7=4.8 Hz, 1 H), 3.72 (d, 7=1.0 Hz, 6H), 3.40 (s, 3H).
Préparation of (Example 9 monomer): To a solution of 6 (8.0 g, 11.60 mmol) in MeCN (150 mL) was added P-l (4.54 g, 15.08 mmol, 4.79 mL) at 0 °C, followed by DCI (1.51 g, 12.76 mmol) in one portion. The mixture was warmed to 20 °C and stirred for 2 h. Upon completion as monitored by LCMS, the reaction mixture was quenched by addition of saturated aq. NaHCCh (50 mL) and diluted with DCM (250 mL). The organic layer was washed with saturated aq.NaHCCh (50 mL * 2), dried over NazSCU, fdtered and concentrated under reduced pressure. The residue was purified by a flash silica gel column (0% to 60% EA in PE contain 0.5% TEA) to give Example 9 monomer (5.75 g, 55.37% yield, 99.4% purity) as a white solid. ESI-LCMS: 890.4 [M+H]+ ; Ή NMR (400 MHz, CD3CN) δ = 9.55 (s, 1H), 8.63 - 8.51 (m, 1H),
149
8.34 - 8.24 (m, 1H), 7.98 (br d, ./=7.5 Hz, 2H), 7.65 - 7.55 (m, 1H), 7.53 - 7.46 (m, 2H), 7.44 7.37 (m, 2H), 7.32 - 7.17 (m, 7H), 6.84 - 6.77 (m, 4H), 6.14 (d, J=4.3 Hz, 1H), 4.84 - 4.73 (m, 1H), 4.72 - 4.65 (m, 1H), 4.34 - 4.27 (m, 1 H), 3.91 - 3.61 (m, 9H), 3.50 - 3.43 (m, 3H), 2.72 2.61 (m, 1H), 2.50 (t, J=6.Q Hz, 1H), 1.21 - 1.15 (m, 10H), 1.09 (d, .7=6.8 Hz, 2H); 31P NMR (162 MHz, CD3CN) δ = 150.01, 149.65
Example 10: Synthesis of 5’ End Cap Monomer
Example 10 Monomer
Example 10 Monomer Synthesis Scheme
Préparation of (2): To a solution of 1 (10 g, 27.22 mmol) in CH3CN (200 mL) and H2O (50 mL) were added TEMPO (3.85 g, 24.50 mmol) and DIB (17.54 g, 54.44 mmol). The mixture was stirred at 25 °C for 12 h. Upon completion as monitored by LCMS, the reaction mixture was concentrated under reduced pressure to give a residue. The residue was triturated with EtOAc (600 mL) for 30 min. The resulting suspension was filtered and the collected solid was washed with EtOAc (300 mL*2) to give 2 (20.09 g, 91.5% yield) as a white solid. ESILCMS: 382.0 [M+H]+.
Préparation of (3): To a solution of 2 (6 g, 15.73 mmol) in MeOH (100 mL) was added SOCI2 (2.81 g, 23.60 mmol, 1.71 mL) dropwise at 0 °C. The mixture was stirred at 25 °C 150 for 12 h. Upon completion as monitored by LCMS, the reaction mixture was quenched by addition of NaHCCh (4 g) and stirred at 25 °C for 30 min. The reaction mixture was filtered and the filtrate was concentrated under reduced pressure to give 3 (18.8 g, 95.6% yield) as a white solid. The crude product was used for the next step without further purification. (The reaction was set up in parallel 3 batches and combined). ESI-LCMS: 396.1 [M+H]+;'H NMR (400 MHz, DMSO-d6) δ= 12.26 - 11.57 (m, 2H), 8.42 - 8.06 (m, 1H), 6.14 - 5.68 (m, 2H), 4.56 (s, 2H), 4.33 (dd, J=4.0, 7.3 Hz, 1H), 3.77 (m, 3H),, 3.30 (s, 3H), 2.81 - 2.69 (m, 1H), 1.11 (s, 6H)
Préparation of (4 & 5): To a mixture of 3 (10.1 g, 25.55 mmol) in CD3OD (120 mL) was added NaBÜ4 (3.29 g, 86.86 mmol, 3.4 eq) in portions at 0 °C. The mixture was stirrèd at 25 °C for 1 h. Upon completion as monitored by LCMS, the reaction mixture was neutralized with AcOH (~ 15 mL) and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (ISCO®; 120 g SepaFlash® Silica Flash Column, Eluent of 0~7.4%, MeOH/DCM gradient @ 80 mL/min) to give 4 (2.98 g, 6.88 mmol, 27% yield) as a yellow solid. ESI-LCMS: 370.1 [M+H]+ and 5 (10.9 g, crude) as a yellow solid. ESILCMS: 300.1[M+H]+; *H NMR (400MHz, CD3OD) δ = 7.85 (s, 1H), 5.87 (d, J=6.0 Hz, 1H), 4.46 - 4.39 (m, 1H), 4.34 (t, J=5.4 Hz, 1H), 4.08 (d, J=3.1 Hz, 1H), 3.49 - 3.38 (m, 4H)
Préparation of 6: To a solution of 4 (1.9 g, 4.58 mmol, 85.7% purity) in pyridine (19 mL) was added DMTrCl (2.02 g, 5.96 mmol). The mixture was stirred at 25 °C for 2 h under N2. Upon completion as monitored by LCMS, the reaction mixture was quenched by MeOH (10 mL) and concentrated under reduce pressure to give a residue. The residue was diluted with H2O (10 mL*3) and extracted with EA (20 mL*3). The combined organic layers were washed with brine (20 mL), dried over anhydrous NaiSCU, filtered and concentrated under reduce pressure to give a residue. The residue was purified by flash silica gel chromatography (ISCO®; 25 g SepaFlash® Silica Flash Column, Eluent of 0-77%, PE: (EA withlO%EtOH): 1%TEA@ 35 mL/min) to give 6 (2.6 g, 81.71% yield, 96.71% purity) as a white foam. ESI-LCMS: 672.2 [M+H]+; Ή NMR (400 MHz, CDCI3) δ= 12.02 ( s, 1H), 7.96 ( s, 1H), 7.83 (s, 1H),7.51 (d, J=7.4 Hz, 2H), 7.37(d, J=8.6 Hz, 4H), 7.25 - 7.17 (m, 2H),6.80 (t, J=8.4 Hz, 4H), 5.88 (d, >6.3 Hz, 1H), 4.69 (t, J=5.7 Ηζ,ΙΗ), 4.64 (s, 1H), 4.54 (s, 1H),4.19 (d, J=2.9 Hz, 1H), 3.77 (d, J=4.5 Hz, 6H), 3.60 - 3.38 (m, 3H),2.81 (s, 1H), 1.81 (td, >6.9, 13.7Hz, 1H), 0.97 (d, J=6.8 Hz, 3H),0.80 (d, J=6.9 Hz, 3H)
Préparation of Example 10 monomer: To a solution of 6 (8.4 g, 12.5 mmol) in MeCN (80 mL) was added P-l (4.9 g, 16.26 mmol, 5.16 mL) at 0°C, followed by addition of DCI (1.624 g, 13.76 mmol) in one portion at 0°C under Ar. The mixture was stirred at 25 °C for 2 h. Upon completion as monitored by LCMS, the reaction mixture was quenched with saturated aq.NaHCCh (20 mL) and extracted with DCM (50 mL*2). The combined organic
151 layers were dried over anhydrous NazSCU, fîltered and concentrated under reduce pressure to give a residue. The residue was purified by flash silica gel chromatography (ISCO®; 40 g SepaFlash® Silica Flash Column, Eluent of 0-52% PE: EA (10%EtOH): 5%TEA, @ 80 mL/min) to give Example 10 monomer (3.4 g, 72.1% yield,) as a white foam. ESI-LCMS:
872.4 [M+H]+; *H NMR (400 MHz, CD3CN) δ= 12.46 - 11.07 (m, 1H), 9.29 (s, 1H), 7.84 (d,
J=14.6 Hz, 1H), 7.42 (t, J=6.9 Hz, 2H), 7.34 - 7.17 (m, 7H), 6.85 - 6.77 (m, 4H), 5.95 - 5.77 (m, 1H), 4.56 - 4.40 (m, 2H), 4.24 (dd, J=4.0, 13.3 Hz, 1H), 3.72 (d, J=2.0 Hz, 7H), 3.66 - 3.53 (m, 3H), 3.42 (d, J=11.8Hz, 3H), 2.69-2.61 (m, 1H), 2.60 - 2.42 (m, 2H), 1.16 - 1.00 (m, 18H); 31P NMR (162 MHz, CD3CN) δ = 149.975, 149.9 ίο Example 11: Synthesis of 5’ End Cap Monomer
TBSCI
TBSO' OCHj
TFA/EtjSiH
NHBz
TBSO'' 'OCH;
TFA
P1/DCI
HO'' 'OCHj
Example 11 Monomer
Example 11 Monomer Synthesis Scheme
Préparation of (2): To a solution of 1 (40 g, 58.16 mmol) in DMF (60 mL) were added imidazole (11.88 g, 174.48 mmol), Nal (13.08 g, 87.24 mmol), and TBSCI (17.52 g, 116.32 mmol) at 20°C in one portion. The reaction mixture was stirred at 20°C for 12 h. Upon completion, the mixture was diluted with EA (200 mL). The organic layer was washed with
152 brine/water (80 mL/80 mL *4), dried over Na2SO4, filtered and evaporated to give 2 (50.8 g, crude) as yellow solid. ESI-LCMS: 802.3 [M+H]+
Préparation of (3): To a solution of 2 (8.4 g, 10.47 mmol) in DCM (120 mL) were added EtaSiH (3.06 g, 26.3 mmol, 4.2 mL) and TFA (1.29 g, 0.84 mL) dropwise at 0 °C. The reaction mixture was stirred at 20°C for 2 h. The reaction mixture was washed with saturated aq.NaHCCh (15 mL) and brine (80 mL). The organic layer was dried over Na2SO4, filtered and evaporated. The residue was purified by flash silica gel chromatography (ISCO®; 80 g SepaFlash® Silica Flash Column, Eluent of 0-83% EA/PE gradient @ 80 mL/min) to give 3 (2.92 g, 55.8% yield,) as a white solid. ESI-LCMS: 500.2 [M+H]+; Ή NMR (400 MHz, CDC13) δ= 8.79 (s, 1H), 8.14 (s, 1H), 8.02 (d, J=7.6 Hz, 2H), 7.64 - 7.58 (m,lH), 7.56 - 7.49 (m, 2H), 5.98 - 5.93 (m, 1H), 4.63 - 4.56 (m, 2H), 4.23 (s, 1H), 3.98 (dd, J=1.5, 13.1 Hz, 1H), 3.75 (dd, >1.5, 13.1 Hz, 1H), 3.28 (s, 3H), 2.06 - 1.99 (m, 1H), 1.00 - 0.90 (m, 9H), 0.15 (d, J=7.0 Hz, 6H).
Préparation of (4): 3 (6 g, 12.01 mmol) and teri-hwO^N-methylsulfonylcarbamate (3.52 g, 18.01 mmol) were co-evaporated with toluene (50 mL), dissolved in dry THF (100 mL), and cooled to 0°C. PPI13 (9.45 g, 36.03 mmol,) was then added, followed by dropwise addition of DIAD (7.28 g, 36.03 mmol, 7.00 mL) in dry THF (30 mL). The reaction mixture was stirred at 20°C for 18 h. Upon completion, the reaction mixture was then diluted with DCM (100 mL) and washed with water (70 mL) and brine (70 mL), dried over Na2SC>4, filtered and evaporated to give a residue. The residue was purified by flash silica gel chromatography (ISCO®; 80 g SepaFlash® Silica Flash Column, Eluent of 0-100% Ethyl acetate/Petroleum ether gradient @ 60 mL/min) followed by reverse-phase HP LC (0.1% NH3.H2O condition, eluent at 74%) to give 4 (2.88 g, 25 % yield) as a white solid. ESI-LCMS: 677.1 [M+H]+ ; Ή NMR (400MHz, CDCI3) δ= 9.24 (s, 1H), 8.84 (s, IH), 8.36 (s, 1H), 8.05 (br d,J=7.3 Hz, 2H), 7.66 - 7.42 (m, 4H), 6.16 (d,
J=5.0 Hz, 1H), 4.52 (br t, J=4.5 Hz, 1H), 4.25 - 4.10 (m, 1H), 3.97 (br dd, J=8.0, 14.8 Hz, 1H),
3.48 (s, 3H), 3.27 (s, 3H), 1.54 (s, 9H), 0.95 (s, 9H), 0.14 (d, J=0.8 Hz, 6H).
Préparation of (5): To a solution of 4 (2.8 g, 4.14 mmol) in THF (20 mL) was added TB AF (4 M, 1.03 mL) and the mixture was stirred at 20°C for 12 h. The reaction mixture was then evaporated. The residue was purified by flash silica gel chromatography (ISCO®; 12 g
SepaFlash® Silica Flash Column, Eluent of 0-6% MeOH/ethyl acetate gradient @ 20 mL/min) to give 5 (2.1 g, 83.92% yield) as a white solid. ESI-LCMS: 563.1 [M+H]+; Ή NMR (400MHz, CDCI3) δ= 8.85 - 8.77 (m, 1H), 8.38 (s, 1H), 8.11 - 7.99 (m, 2H), 7.64 -7.50 (m, 4H), 6.19 (d, J=2.8 Hz, 1H), 4.36 - 4.33 (m, 1H), 4.29 (br d, J=4.3 Hz, 1H), 4.22 - 4.02 (m, 2H), 3.65 - 3.59 (m, 3H), 3.28 (s, 3H), 1.54 (s, 9H).
153
Préparation of (6): To a solution of 5 (2.1 g, 3.73 mmol) in DCM (20 mL) was added TFA (7.70 g, 67.53 mmol, 5 mL) at 0°C. The reaction mixture was stirred at 20°C for 24 h. Upon completion, the reaction was quenched with saturated aq. NaHCCf to reach pH 7. The organic layer was dried over Na2SO4, filtered, and evaporated at low pressure. The residue was purified by flash silica gel chromatography (ISCO®; 12 g SepaFlash® Silica Flash Column, Eluent of 0~7% DCM/MeOH gradient @ 20 mL/min) to give 1.6 g (impure, 75% LCMS purity), followed by prep-HPLC [FA condition, column: Boston Uni Cl8 40*150*5um; mobile phase: [water (0.225%FA)-ACN]; B%: 8%-38%,7.7min.] to give 6 (1.04 g, 63.7 % yield) as a white solid. ESI-LCMS: 485.0 [M+Na]+; Ή NMR (400 MHz, DMSO-d6) δ= 11.27 - 11.21 (m, 1H), 8.77 (s, 1H), 8.74 (s, 1H), 8.05 (d, J=7.3 Hz, 2H), 7.68 -7.62 (m, 1H), 7.59 - 7.53 (m, 2H), 7.39 (t, J=6.3 Hz, 1H), 6.16 (d, J=6.0 Hz, 1H), 5.48 (d, J=5.5 Hz, 1H), 4.55 (t,J=5.5 Hz, 1H), 4.43 4.37 (m, 1H), 4.08 - 4.02 (m, 1H), 3.41 - 3.36 (m, 1H), 3.35 (s, 3H), 3.31 - 3.22 (m, 1H), 2.91(s, 3H).
Préparation of (Example 11 monomer): To a solution of 6 (1 g, 2.16 mmol) in DCM (30 mL) was added PI (977.58 mg, 3.24 mmol, 1.03 mL), followed by DCI (306.43 mg, 2.59 mmol) at 0°C in one portion under Ar atmosphère. The mixture was degassed and purged with Ar for 3 times, warmed to 20°C, and stirred for 2 hr under Ar atmosphère. Upon completion as monitored by LCMS and TLC (PE: EtOAc = 4:1), the reaction mixture was diluted with sat.aq. NaHCCL (30 mL) and extracted with DCM (50 mL*2). The combined organic layers were dried over anhydrous Na2SÛ4, filtered, and the filtrate was concentrated under reduced pressure to give a residue. The crude product was purified by reversed-phase HP LC (40 g Cl 8 column: neutral condition, Eluent of 0-57% of 0.3% NH4HCO3 in H2O/CH3CN ether gradient @35 mL/min) to give Example 11 monomer (0.49 g, 33.7% yield) as a white solid. ESI-LCMS: 663.1[M+H]+; ‘H NMR (400 MHz, CD3CN) δ= 1.19 - 1.29 (m, 12 H) 2.71 (q, J=5.77 Hz, 2 H) 2.94 (d, J=6.27 Hz, 3 H) 3.35 (d, J=15.56 Hz, 3 H) 3.40 - 3.52 (m, 2 H) 3.61 - 3.97 (m, 4 H) 4.23 - 4.45 (m, 1 H) 4.55 - 4.74 (m, 2 H) 6.02 (dd, J=10.67, 6.40 Hz, 1 H) 7.25 (br s, 1 H) 7.47 - 7.57 (m, 2 H) 7.59 - 7.68 (m, 1 H) 8.01 (d, J=7.78 Hz, 2 H) 8.28 (s, 1 H) 8.66 (s, 1 H) 9.69 (br s, 1 H); 31P NMR (162 MHz, CD3CN) δ = 150.92, 149.78.
Example 12. Synthesis of 5’-stabilized end cap modified oligonucleotides
This example provides an exemplary method for synthesizing the siNAs comprising a 5’stabilized end caps disclosed herein. The 5’-stabilized end cap and/or deuterated phosphoramidites were dissolved in anhydrous acetonitrile and oligonucleotide synthesis was performed on a Expedite 8909 Synthesizer using standard phosphoramidite chemistry. An extended coupling (12 minutes) of 0.12 M solution of phosphoramidite in anhydrous CH3CN in
154 the presence of Benzyl-thio-tetrazole (BTT) activator to a solid bound oligonucleotide followed by standard capping, oxidation and sulfurization produced modified oligonucleotides. The 0.02 M 12, THF: Pyridine; Water 7:2:1 was used as an oxidizing agent, while DDTT (dimethylaminomethylidene) amino)-3H-l,2,4-dithiazaoline-3-thione was used as the sulfur-transfer agent for the synthesis of oligoribonucleotide with a phosphorothioate backbone. The stepwise coupling efficiency of ail modified phosphoramidites was achieved around 98%. After synthesis the solid support was heated with aqueous ammonia (28%) solution at 45°C for 16h or 0.05 M K2CO3 in methanol was used to deprotect the base labile protecting groups. The crude oligonucleotides were precipitated with isopropanol and centrifùged (Eppendorf 581 OR, 3000g, 4°C, 15 min) to obtain a pellet. The crude product was then purified using ion exchange chromatography (TSK gel column, 20 mM NaH2PÜ4, 10% CH3CN, 1 M NaBr, gradient 20-60% B over 20 column volumes) and fractions were analyzed by ion change chromatography on an HPLC. Pure fractions were pooled and desalted by Sephadex G-25 column and evaporated to dryness. The purity and molecular weight were determined by HPLC analysis and ESI-MS analysis. Single strand RNA oligonucleotides (sense and antisense strand) were annealed (1:1 by molar équivalents) at 90°C for 3 min followed by RT 40 min) to produce the duplexes.
Example 13. siNA Activity Assays
This example provides exemplary methods for testing the activity of the siNAs disclosed herein.
In vitro Assay:
Homo sapiens HepG2.2.15 cells were cultured in Dulbecco’s Modified Eagle’s Medium (DMEM) (ATCC 30-2002) supplemented to also contain 10% fêtai calf sérum (FCS). Cells were incubated at 37°C in an atmosphère with 5% CO2 in a humidified incubator. For transfection of HepG2.2.15 cells with HBV targeting siRNAs, cells were seeded at a density of 15000 cells/well in 96-well regular tissue culture plates. Transfection of cells was carried out using RNAiMAX (Invitrogen/Life Technologies) according to the manufacturer’s instructions. Dose-response experiments were done with oligo concentrations of 40, 20, 10, 5, 2.5, 1.25, 0.625, 0.3125, 0.15625 and 0.07813nM. For each HBV targeting siRNA treatment (e.g., ds-siRNA, as identified by the ds-siNA ID in Table 6), four wells were transfected in parallel, and individual data points were collected from each well. After 24h of incubation with siRNA, media was removed, and cells were lysed and analyzed with a QuantiGene2.0 branched DNA (bDNA) probe set spécifie for HBV génotype D (also called Hepatitis B virus subtype ayw, complété genome of 3182 basepairs) as présent in cell line HepG2.2.15.
155
For each well, the HBV on-target mRNA levels were normalized to the GAPDH mRNA level. As shown in Table 6, the activity of the HBV targeting ds-siRNAs was expressed as EC50, 50% réduction of normalized HBV RNA level from no drug control. As shown in Table 6, the cytotoxicity of the HBV targeting ds-siRNAs was expressed by CC50 of 50% réduction of
GAPDH mRNA from no drug control.
Unconjugated siRNA 1) with or without a phosphorylation blocker; and 2) with or without end caps (e.g., 5’-stabilized end cap) are transfected into in vitro disease models or in vitro toxicity models. Afiter transfection, target réduction and/or cell viability is measured and compared after a period of incubation. For HBV, exemplary disease cell models include, but are 10 not limited to, HepG2.2.15, HepG2.117 or live HBV infected HepG2-NTCP or Primary Human Hépatocytes.
In vivo Assay:
GalNAc conjugated siRNA 1) with or without phosphorylation blocker; and 2) with or without 5’- end caps are dosed subcutaneously or intravenously in animal disease models. The 15 target knockdown magnitude and duration is measured from sérum or liver samples and compared to each other and/or control animais (e.g., non-treated diseased animais). In some instances, the toxicity of the siRNAs is compared through routine Clinpath or Histopath assays. For HBV, exemplary animal efficacy models include, but are not limited to, AAV-HBV mouse model, HBV transgenic mouse model, PXB or FRG mouse models.
Example 14. ds-siNA Testing in AAV-HBV Mouse Model
In this example, the efficacy of ds-siNAs in treating HBV in an adeno-associated virus (AAV)-HBV mouse model was evaluated. AAV-HBV mice were subcutaneously injected with a single dose of (a) 5mL/kg of vehicle; or (b) 5mg/kg a ds-siNA at day 0. The sequences of the dssiNA tested in this example are shown in Table 7.
FIG. 4 shows a graph of the change in sérum HBsAg from AAV-HBV mice treated with vehicle (G01), ds-siNA-0160 (G03), ds-siNA-0165 (G04), ds-siNA-0163 (G05), or ds-siNA0166 (G06). These results demonstrate that the ds-siNAs containing varions patterns of 2’-fluoro nucléotides and 2’-O-methyl nucléotides can effectively treated HBV.
Table 7. ds-siNA sequences tested in AAV-HBV mouse model
ds-siNA ID Sense strand sequence (5’-3 ’) Antisense strand sequence (5’-3’)
ds-siNA- 0160 mCpsmCpsfGmUmGmUfGfCfAmCmUf UmCmGmCmUfUmCmA-p-(PS)2- GalNAc4 (SEQ ID NO: 600) mUpsfGpsmAmAmGmCmGmAmAmGm UmGmCfAmCmAmCmGmGpsmUpsmC (SEQ ID NO: 272)
156
ds-siNA- 0165 mGpsmUpsfGmGmUmGfGfAfCmUmU fCmUmCmUmCfAmAmU-p-(PS)2GalNAc4 (SEQ ID NO: 601) mApsfUpsmUmGmAmGmAmGmAmA mGmUmCfCmAmCmCmAmCpsmGpsm A (SEQ ID NO: 292)
ds-siNA- 0163 mGpsmCpsmUmGmCmUfAmUfGfCfC mUmCmAmUmCmUmUmCmUmU-p(PS)2-GalNAc4 (SEQ ID NO: 602) mApsfApsmGmAmAfGmAmUmGmAm GmGmCfAmUfAmGmCmAmGmCpsmA psmG (SEQ ID NO: 287)
ds-siNA- 0166 mUpsmGpsfU mGmCm AfCfU mUmCm GmCmUmUmCmAfCmCmU-p-(PS)2GalNAc4 (SEQ ID NO: 603) mApsfGpsmGmUmGmAmAmGmCmGm AmAmGfUmGmCmAmCmApsmCpsmG (SEQ ID NO: 303)
Example 15. ds-siNA Activity Assay and Testing in AAV-HBV Mouse Model
This example investigates the in vitro and in vivo activity of ds-siNAs. The sequences of the ds-siNAs tested in this example are shown in Table 8. As shown in Table 8, the ds-siNAs comprise a sense and antisense strand comprising a mixture of 2’-fluoro and 2’-O-methyl nucléotides. The total number of 2’-fluoro nucléotides in the ds-siNAs are between 6-8. The 2’fluoro nucléotides may be at spécifie positions, such as nucléotide position 3, 5, 7, 8, 9, 10, 11, 12, and/or 17 from the 5’ end of the sense strand or 2, 5, 6, 8, 10, 14, 16, 17, and/or 18. The 2’fluoro nucléotides and 2’-(9-methyl nucléotides might occur at spécifie patterns on the antisense strand, such as an altemating 1:2 or 1:3 pattern, wherein 1 nucléotide is a 2’-fluoro nucléotide and 2 or 3 nucléotides are 2-O-methyl nucléotides.
In vitro Activity Assay
Homo sapiens HepG2.2.15 cells were cultured in Dulbecco’s Modified Eagle’s Medium (DMEM) (ATCC 30-2002) supplemented to also contain 10% fêtai calf sérum (FCS). Cells were incubated at 37°C in an atmosphère with 5% CO2 in a humidified incubator. For transfection of HepG2.2.15 cells with HBV targeting siRNAs, cells were seeded at a density of 15000 cells/well in 96-well regular tissue culture plates. Transfection of cells was carried out using RNAiMAX (Invitrogen/Life Technologies) according to the manufacturer’s instructions. Dose-response experiments were done with oligo concentrations of 40, 20, 10, 5, 2.5, 1.25, 0.625, 0.3125, 0.15625 and 0.07813nM. For each HBV targeting siRNA treatment (e.g., ds-siRNA, as identified by the ds-siNA ID in Table 8), four wells were transfected in parallel, and individual data points were collected from each well. After 24h of incubation with siRNA, media was removed, and cells were lysed and analyzed with a QuantiGene2.0 branched DNA (bDNA) probe set spécifie for HBV génotype D (also called Hepatitis B virus subtype ayw, complété genome of 3182 basepairs) as présent in cell line HepG2.2.15.
157
For each well, the HBV on-target mRNA levels were normalized to the GAPDH mRNA level. Table 8 shows the activity of the HBV targeting ds-siRNAs expressed as EC50, which is 50% réduction of normalized HBV RNA level from no drug control, where A = EC50 < 0.5 nM; B = 0.5 nM < EC50 < 1; and C = EC50 > 1.
In vivo testing in AAV-HBVmouse model:
AAV/HBV is a recombinant AAV carrying repli cable HBV genome. Taking advantage of the highly hepatotropic feature of génotype 8 AAV, the HBV genome can be efficiently delivered to the mouse liver cells. Infection of immune competent mouse with AAV/HBV can resuit in long term HBV viremia, which mimics chronic HBV infection in patients. The AAV/HBV model can be used to evaluate the in vivo activity of varions types of anti-HBV agents. Mice were infected with AAV-HBV on day -28 of the study. The test articles or négative control (PB S) were dosed subcutaneously (unless specifîed otherwise) as single dose on days 0 at 5 mg/kg. Serial blood collections were usually taken every 5 days on day 0, 5, 10 and 15 etc. until the termination of studies. Sérum HBV S antigen (HBsAg) was assayed through ELISA.
GalNAc conjugated ds-siNAs were further tested at a single dose of 5mg/kg at day 0 in the adeno-associated virus (AAV)-HBV mouse model. The resulting nadir logio réduction in sérum HBsAg is presented in Table 8, where X > 1 logio réduction in HBsAg, Y is 0.5 - 1 logio réduction in HBsAg, and Z is < 0.5 logio réduction in HBsAg.
FIG. 5A shows a graph of the change in sérum HBsAg from AAV-HBV mice treated with vehicle (G01) or ds-siNA-0160 (G03). AAV-HBV mice were subcutaneously injected with a single dose of 5mL/kg of vehicle or 5mg/kg of ds-siNA-0160 on day 0.
FIG. 5B shows a graph of the change in sérum HBsAg from AAV-HBV mice treated with vehicle (G01) or ds-siNA-0160 (G15). AAV-HBV mice were subcutaneously injected with a single dose of 5mL/kg of vehicle or 5mg/kg of ds-siNA-0160 on day 0.
FIG. 5C shows a graph of the change in sérum HBsAg from AAV-HBV mice treated with vehicle (G01) or ds-siNA-0160 (G03). AAV-HBV mice were subcutaneously injected with a single dose of 5mL/kg of vehicle or 5mg/kg of each ds-siNA on day 0.
FIG. 5D shows a graph of the change in sérum HBsAg from AAV-HBV mice treated with vehicle (G01), ds-siNA-0160 (G03), or ds-siNA-0109 (G09). AAV-HBV mice were subcutaneously injected with a single dose of 5mL/kg of vehicle or 5mg/kg of each ds-siNA on day 0.
158
FIGs. 5E-5F show a graph of the change in sérum HBsAg from AAV-HBV mice treated with vehicle (G01) or ds-siNA-0169 (G18). AAV-HBV mice were subcutaneously injected with a single dose of 5mL/kg of vehicle or 5mg/kg of ds-siNA-0169 on day 0.
FIG. 5G shows a graph of the change in sérum HBsAg from AAV-HBV mice treated with vehicle (G01) or ds-siNA-0169 (G04). AAV-HBV mice were subcutaneously injected with a single dose of 5mL/kg of vehicle or 5mg/kg of ds-siNA-0169 on day 0.
FIG. 5H shows a graph of the change in sérum HBsAg from AAV-HBV mice treated with vehicle (G01) or ds-siNA-0169 (G04). AAV-HBV mice were subcutaneously injected with a single dose of 5mL/kg of vehicle or 5mg/kg of each ds-siNA on day 0.
FIG. 51 shows a graph of the change in sérum HBsAg from AAV-HBV mice treated with vehicle (G01), ds-siNA-0169 (G04) or ds-siNA-0147 (G08). AAV-HBV mice were subcutaneously injected with a single dose of 5mL/kg of vehicle or 5mg/kg of each ds-siNA on day 0.
FIG. 5J shows a graph of the change in sérum HBsAg from AAV-HBV mice treated with 15 vehicle (G01), ds-siNA-0166 (G06), or ds-siNA-0153 (G14). AAV-HBV mice were subcutaneously injected with a single dose of 5mL/kg of vehicle or 5mg/kg of each ds-siNA on day 0.
FIG. 5K shows a graph of the change in sérum HBsAg from AAV-HBV mice treated with vehicle (G01), ds-siNA-0163 (G05), or ds-siNA-0119 (G13). AAV-HBV mice were subcutaneously injected with a single dose of 5mL/kg of vehicle or 5mg/kg of each ds-siNA on day 0.
These results demonstrate that ds-siNAs comprising combination of 2’-fluoro nucléotides and 2’-O-methyl nucléotides can be used to target HBV X and S gene sequences, which resulted in successfùl treatment of HBV.
As exemplified by ds-siNA-0160 and ds-siNA-0165, ds-siNAs comprising (a) a sense strand comprising 19 nucléotides, wherein 6 nucléotides are 2’-fluoro nucléotides and 13 nucléotides are 2’-O-methyl nucléotides; (b) an antisense strand comprising 21 nucléotides, wherein 2 nucléotides are 2’-fluoro nucléotides and 19 nucléotides are 2’-(9-methyl nucléotides; and (c) a conjugated moiety, wherein the conjugated moiety is attached to the 3 ’ end of the sense strand, resulted in successfùl treatment of HBV as evidenced by HBsAg réduction in sérum. See FIGs. 4 and 5A-5D, and Table 8. For ds-siNA-0160 and ds-siNA-0165, the 2’-fluoro nucléotides
159 were located at positions 3, 7-9, 12, and 17 from the 5’ end of the sense strand and at positions 2 and 14 from the 5’ end of the antisense strand.
As exemplified by ds-siNA-0166, ds-siNAs comprising (a) a sense strand comprising 19 nucléotides, wherein 4 nucléotides are 2’-fluoro nucléotides and 15 nucléotides are 2’-O-methyl 5 nucléotides; (b) an antisense strand comprising 21 nucléotides, wherein 2 nucléotides are 2’fluoro nucléotides and 19 nucléotides are 2’-O-methyl nucléotides; and (c) a conjugated moiety, wherein the conjugated moiety is attached to the 3’ end of the sense strand, resulted in successful treatment of HBV as evidenced by HBsAg réduction in sérum. See FIGs. 4 and 5J, and Table 8. For ds-siNA-0166, the 2’-fluoro nucléotides were located at positions 3, 7, 8, and 17 from the 5’ 10 end of the sense strand and at positions 2 and 14 from the 5’ end of the antisense strand.
As exemplified by ds-siNA-0153, ds-siNAs comprising (a) a sense strand comprising 19 nucléotides; (b) an antisense strand comprising 21 nucléotides, wherein the nucléotides in the antisense strand comprise at least two altemating 1:3 modification pattern, and wherein approximate 1 nucléotide is a 2’-fluoro nucléotide and 3 nucléotides are 2’-O-methyl nucléotides in repeat pattern; and (c) a conjugated moiety, wherein the conjugated moiety is attached to the 3’ end of the sense strand, resulted in successful treatment of HBV as evidenced by HBsAg réduction in sérum. See FIG. 5J. For ds-siNA-0153, the sense strand comprises 6 2’fluoro nucléotides at positions 3, 7-9, 12, and 17 from the 5’ end of the sense strand. In addition, the antisense strand comprises 5 repeats of the 1:3 modification pattern starting at position 2 from the 5’ end of the antisense strand.
As exemplified by ds-siNA-0109, ds-siNAs comprising (a) a sense strand comprising 19 nucléotides wherein 4 nucléotides are 2’-fluoro nucléotides and 15 nucléotides are 2’-O-methyl nucléotides; (b) an antisense strand comprising 21 nucléotides, wherein 4 nucléotides are 2’fluoro nucléotides and 17 nucléotides are 2’-O-methyl nucléotides; and (c) a conjugated moiety, 25 wherein the conjugated moiety is attached to the 3 ’ end of the sense strand, resulted in successful treatment of HBV as evidenced by HBsAg réduction in sérum. See FIG. 5D. For ds-siNA-0109 the sense strand comprises 4 2’-fluoro nucléotides at positions 5 and 7-9 from the 5’ end of the sense strand. In addition, the antisense strand comprises 5 repeats of the 1:2 modification pattern starting at positions 2, 5, 8, 14, and 17 from the 5’ end of the antisense strand.
As exemplified by ds-siNA-0147, ds-siNAs comprising (a) a sense strand comprising 19 nucléotides; (b) an antisense strand comprising 21 nucléotides, wherein the nucléotides in the antisense strand comprise at least two altemating 1:2 modification pattern, and wherein approximate 1 nucléotide is a 2’-fluoro nucléotide and 2 nucléotides are 2’-O-methyl
160 nucléotides in repeat pattern; and (c) a conjugated moiety, wherein the conjugated moiety is attached to the 3’ end of the sense strand, resulted in successful treatment of HBV as evidenced by HBsAg réduction in sérum. See FIG. 51. For ds-siNA-0147, the 2’-fluoro nucléotides were located at positions 5 and 7-9 from the 5’ end of the sense strand and at positions 2, 6, 14, and 16 5 from the 5’ end of the antisense strand.
Table 8. ds-siNA tested in AAV-HBV Mouse Model
ds- siNA ID Sense strand sequence (5’-3 ’) Antisense strand sequence (5’3’) EC50 HepG2. 2.15* HBsAg Nadir (Log)**
dssiNA- 0109 mCpsmCpsmGmUfGmUfGfCf AmCmUmUmCmGmCmUmU mCmA-p-(PS)2-GalNac4 (SEQ ID NO: 604) mUpsfGpsmAmAfGmCmGfA mAmGmUmGmCfAmCmAfC mGmGpsmUpsmC (SEQ ID NO: 605)
ds- siNA- 0119 mGpsmCpsmUmGfCmUmAm UfGfCfCmUmCfAmUmCmU mUfCmUmU-p-(PS)2-GalNac4 (SEQ ID NO: 606) mApsfApsmGmAmAmGmA mUmGmAmGmGmCfAmUm AmGmCmAmGmCpsmApsm G (SEQ ID NO: 495)
ds- siNA- 0147 mGpsmUpsmGmGfUmGfGfAf CmUmUmCmUmCmUmCmA mAmU-p-(PS)2-GalNac4 (SEQ ID NO: 607) mApsfUpsmUmGmAfGmAm GmAmAmGmUmCfCmAfCm CmAmCpsmGpsmA (SEQ ID NO: 608)
ds- siNA- 0153 mUpsmGpsfUmGmCmAfCfUf UmCmGfCmUmUmCmAfCm CmU-p-(PS)2-GalNac4 (SEQ ID NO: 609) mApsfGpsmGmUmGfAmAm GmCfGmAmAmGfUmGmC mAfCmApsmCpsmG (SEQ ID NO: 610)
dssiNA- 0167 mGpsmCpsfGmGmGmGfUfUf UmUmUfCmUmUmGmUfUm GmA-p-(PS)2-GalNac4 (SEQ ID NO: 611) mUpsfCpsmAmAmCmAmA mGmAmAmAmAmAfCmCm CmCmGmCpsmCpsmU (SEQ ID NO: 285) A X
ds- siNA- 0162 mGpsmCpsfGmGmGmGfU fU mUmUmUmCmUmUmGmUf UmGmA-p-(PS)2-GalNac4 (SEQ ID NO: 612) mUpsfCpsmAmAmCmAmA mGmAmAmAmAmAfCmCm CmCmGmCpsmCpsmU (SEQ ID NO: 285) C X
161
dssiNA- 0165 mGpsmUpsfGmGmUmGfGfAf CmUmUfCmUmCmUmCfAm AmU-p-(PS)2-GalNac4 (SEQ ID NO: 601) mApsfUpsmUmGmAmGmA mGmAmAmGmUmCfCmAm CmCmAmCpsmGpsmA (SEQ ID NO: 292) A X
dssiNA- 0168 mUpsmCpsmGmUmGmGfUm GfGfAfCmUmUmCmUmCmU mCmAmAmU-p-(PS)2GalNac4(SEQ ID NO: 613) . mApsfUpsmUmGmAfGmAm GmAmAmGmUmCfCmAfCm CmAmCmGmApsmGpsmU (SEQ ID NO: 298) A X
dssiNA- 0163 mGpsmCpsmUmGmCmUfAm UfGfCfCmUmCmAmUmCmU mUmCmUmU-p-(PS)2GalNac4 (SEQ ID NO: 602) mApsfApsmGmAmAfGmAm UmGmAmGmGmCfAmUfA mGmCmAmGmCpsmApsmG (SEQ ID NO: 287) A Y
ds- siNA- 0161 mCpsmUpsfGmCmUmAfUfGf CmCmUfCmAmUmCmUfUm CmU-p-(PS)2-GalNac4 (SEQ ID NO: 614) mApsfGpsmAmAmGmAmU mGmAmGmGmCmAfUmAm GmCmAmGpsmCpsmA (SEQ ID NO: 277) A
dssiNA- 0160 mCpsmCpsfGmUmGmUfGfCf AmCmUfUmCmGmCmUfUm CmA-p-(PS)2-GalNac4 (SEQ ID NO: 600) mUpsfGpsmAmAmGmCmG mAmAmGmUmGmCfAmCm AmCmGmGpsmUpsmC (SEQ ID NO: 272) A X
dssiNA- 0169 mCpsmCpsfGmUmGmUfGfCf AmCmU fümCmGmCmU fUm CmA-p-(PS)2-GalNac4 (SEQ ID NO: 600) mUpsfGpsmAmAmGmCmG mAmAmGmUmGmCfAmCm AmCmGmGpsTpsT (SEQ ID NO: 375) ' A X
dssiNA- 0170 mUpsmGpsfUmGmCmAfCfUf UmCmGfCmUmUmCmAfCm CmU- p-(PS)2-GalNac4 (SEQ ID NO: 609) mApsfGpsmGmUmGmAmA mGmCniGmAmAmGfU mGm CmAmCmApsmCpsmG (SEQ ID NO: 303) A X
dssiNA- 0166 mUpsmGpsfUmGmCmAfCfU mUmCmGmCmUmUmCmAfC mCmU-p-(PS)2-GalNAc4 (SEQIDNO: 615) mApsfGpsmGmUmGmAmA mGmCmGmAmAmGfUmGm CmAmCmApsmCpsmG (SEQ ID NO: 303) A X
162
ds- siNA- 0171 mUpsmGpsfUmGmCmAfCfU mUmCmGmCmUmUmCmAfC mCmU- p-(PS)2-GalNac4 (SEQ IDNO: 615) mApsfGpsmGmUmGmAmA mGmCmGmAmAmGfUmGm CmAmCmApsTpsT (SEQ ID NO: 407) A X
mX = 2’-O-methyl nucléotide; fX = 2’-fluoro nucléotide; ps= phosphorothioate linkage * For EC50, A = EC50 < 0.5 nM; B = 0.5 nM < EC50 < 1; and C = EC50 > 1. **For HBsAg Nadir, X > 1 logio réduction in HBsAg, Y is 0.5 - 1 logio réduction in HBsAg, and Z is < 0.5 logio réduction in HBsAg.
Example 16. Testing of ds-siNAs having a 5’-stabiIized end cap in AAV-HBV Mouse Model
This example investigates the in vivo activity of ds-siNAs having a 5’-stabilized end cap. 5 The sequences of the ds-siNAs tested in this example are shown in Table 9.
FIG. 6A shows a graph of the change in sérum HBsAg from AAV-HBV mice treated with vehicle (G01), ds-siNA-0160 (G15) (ds-siNA without a 5’-stabilized end cap, e.g., vinyl phosphonate), or ds-siNA-080 (G14) (ds-siNA with a 5’-stabilized end cap, e.g., vinyl phosphonate). AAV-HBV mice were subcutaneously injected with a single dose of 5mL/kg of 10 vehicle or 5mg/kg of each ds-siNA on day 0. The resulting nadir logio réduction in sérum
HBsAg is presented in Table 9, where X > 1 logio réduction in HBsAg, Y is 0.5 - 1 logio réduction in HBsAg, and Z is < 0.5 logio réduction in HBsAg.
FIG. 6B shows a graph of the change in sérum HBsAg from AAV-HBV mice treated with vehicle (G01), ds-siNA-0169 (G16) (ds-siNA without a 5’-stabilized end cap, e.g., vinyl 15 phosphonate), or ds-siNA-081 (G13) (ds-siNA with a 5’-stabilized end cap, e.g., vinyl phosphonate). AAV-HBV mice were subcutaneously injected with a single dose of 5mL/kg of vehicle or 5mg/kg of each ds-siNA on day 0. The resulting nadir logio réduction in sérum HBsAg is presented in Table 9, where X > 1 logio réduction in HBsAg, Y is 0.5 - 1 logio réduction in HBsAg, and Z is < 0.5 logio réduction in HBsAg.
FIG. 7A shows a graph of the change in sérum HBsAg from AAV-HBV mice treated with vehicle (G01), ds-siNA-0165 (G18) (ds-siNA without a 5’-stabilized end cap, e.g., vinyl phosphonate), or ds-siNA-0127 (G17) (ds-siNA with a 5’-stabilized end cap, e.g., vinyl phosphonate). AAV-HBV mice were subcutaneously injected with a single dose of 5mL/kg of vehicle or 5mg/kg of each ds-siNA on day 0. The resulting nadir logio réduction in sérum
HBsAg is presented in Table 9, where X > 1 logio réduction in HBsAg, Y is 0.5 - 1 logio réduction in HBsAg, and Z is < 0.5 logio réduction in HBsAg.
163
FIG. 7B shows a graph of the change in sérum HBsAg from AAV-HBV mice treated with vehicle (G01), ds-siNA-0168 (G20) (ds-siNA without a 5’-stabilized end cap, e.g., vinyl phosphonate), or ds-siNA-0150 (G19) (ds-siNA with a 5’-stabilized end cap, e.g., vinyl phosphonate). AAV-HBV mice were subcutaneously injected with a single dose of 5mL/kg of 5 vehicle or 5mg/kg of each ds-siNA on day 0. The resulting nadir logio réduction in sérum HBsAg is presented in Table 9, where X > 1 logio réduction in HBsAg, Y is 0.5 - 1 logio réduction in HBsAg, and Z is < 0.5 logio réduction in HBsAg.
These results demonstrate that the addition of a 5’-stabilized end cap can improve the efficacy of ds-siNAs without a 5’-stabilized end cap.
Table 9. ds-siNA sequences and HBsAg Nadir
ds-siNA ID Sense strand sequence (5’-3’) Antisense strand sequence (5’-3’) HBsAg Nadir (Log)*
ds-siNA- 0160 mCpsmCpsfGmUmGmUfGfC fAmCmUfUmCmGmCmUfU mCmA-(PS)2-p-GalNAc4 (SEQIDNO: 616) mUpsfGpsmAmAmGmCmGmAmAm GmUmGmCfAmCmAmCmGmGpsm UpsmC (SEQ ID NO: 272)
ds-siNA- 080 mCpsmCpsfGmUmGmUfGfC fAmCmUfUmCmGmCmUfU mCmA-(PS)2-p-GalNAc4 (SEQ ID NO: 616) VmUpsfGpsmAmAmGmCmGmAmA mGmUmGmCfAmCmAmCmGmGps mUpsmC (SEQ ID NO: 462) X
ds-siNA- 0169 mCpsmCpsfGmUmGmUfGfC fAmCmUfUmCmGmCmUfU mCmA-(PS)2-p-GalNAc4 (SEQ ID NO: 616) mUpsfGpsmAmAmGmCmGmAmAm GmUmGmCfAmCmAmCmGmGpsTp sT (SEQ ID NO: 375)
ds-siNA- 081 mCpsmCpsfGmUmGmUfGfC fAmCmUfUmCmGmCmUfU mCmA-(PS)2-p-GalNAc4 (SEQ ID NO: 616) VmUpsfGpsmAmAmGmCmGmAmA mGmUmGmCfAmCmAmCmGmGpsT psT (SEQ ID NO: 463) X
ds-siNA- 0165 mGpsmUpsfGmGmUmGfGfA fCmUmUfCmUmCmUmCfA mAmU-(PS)2-p-GalNAc4 (SEQ IDNO: 617) mApsfUpsmUmGmAmGmAmGmAm AmGmUmCfCmAmCmCmAmCpsmG psmA (SEQ ID NO: 292) X
164
ds-siNA- 0127 mGpsmUpsfGmGmUmGfGfA fCmUmUfCmUmCmUmCfA mAmU-(PS)2-p-GalNAc4 (SEQ ID NO: 617) vmApsfUpsmUmGmAmGmAmGmA mAmGmUmCfCmAmCmCmAmCpsm GpsmA (SEQ ID NO: 503) X
ds-siNA- 0168 mUpsmCpsmGmUmGmGfUm GfGfAfCmUmUmCmUmCm UmCmAmAmU-(PS)2-pGalNAc4 (SEQ ID NO: 618) mApsfUpsmUmGmAfGmAmGmAmA mGmUmCfCmAfCmCmAmCmGmAp smGpsmU (SEQ ID NO: 298)
ds-siNA- 0150 mUpsmCpsmGmUmGmGfUm GfGfAfCmUmUmCmUmCm UmCmAmAmU-(PS)2-pGalNAc4 (SEQ ID NO: 618) vmApsfUpsmUmGmAfGmAmGmAm AmGmUmCfCmAfCmCmAmCmGm ApsmGpsmU (SEQ ID NO: 523) X
mX = 2’-(2-methyl nucléotide; fX = 2’-fluoro nucléotide; ps= phosphorothioate linkage; VP = vinyl phosphonate *For HBsAg Nadir, X > 1 logio réduction in HBsAg, Y is 0.5 - 1 logio réduction in HBsAg, and Z is < 0.5 logio réduction in HBsAg.
Example 17. Efficacy of a Combination Therapy in AAV-HBV Mouse Model This example investigates the efficacy of a combination therapy comprising an antisense oligonucleotide (ASO 1, 5’ GalNAc4-ps-GalNAc4-ps-GalNAc4-po-mA-po- lnGpslnApslnTpslnApslnApsApsAps(5OH)CpsGps(5m)Cps(5m)CpsGps(5m)CpslnApslnGpsln Apscp(5m)C-3’(SEQ ID NO: 534)) and a ds-siNA-0160 for treating HBV in an AAV-HBV mouse model.
FIG. 8A shows a graph of the change in sérum HBsAg from AAV-HBV mice treated with vehicle (G01), ds-siNA-0160 (G06), ASO 1 (G18), or a combination of ds-siNA-0160 and
ASO 1 (G20). AAV-HBV mice were subcutaneously injected with (a) 5mL/kg of vehicle, three times a week, from days 0-42 (G01); (b) a single dose of 3mg/kg of ds-siNA-0160 on day 0 (G06); (c) 3mg/kg of ASO 1 on a weekly basis, from days 0-21 (G18); or (d) a combination of ASO 1 and ds-siNA-0160, wherein ASO 1 was administered at a dose of 3mg/kg on a weekly basis, from days 0-21; and ds-siNA-0160 was administered as a single dose of 3mg/kg at day 0.
FIG. 8B shows a graph of the change in sérum HBsAg from AAV-HBV mice treated with vehicle (G01), ds-siNA-0160 (G06), ASO 1 (G18), or a combination of ds-siNA-0160 and ASO 1 (G20). AAV-HBV mice were subcutaneously injected with (a) 5mL/kg of vehicle, three times a week, from days 0-42 (G01); (b) a single dose of lOmg/kg of ds-siNA-0160 on day 0
165 (G06); (c) lOmg/kg of ASO 1 on a weekly basis, from days 0-21 (G18); or (d) a combination of ASO 1 and ds-siNA-0160, wherein ASO 1 was administered at a dose of lOmg/kg on a weekly basis, from days 0-21; and ds-siNA-0160 was administered as a single dose of 3mg/kg at day 0.
FIG. 8C shows a graph of a synergy analysis of an in vitro combination therapy with the ASO 2 and ds-siNA-0164. For the ds-siNA-0164 combination studies with ASO 2, 35,000 cells per well were reverse transfected in a collagen I-coated 96-well plate (Corning, Biocoat; Catalog 356698). Test articles ds-siNA-0164 and ASO 2 were diluted in Opti-MEM™ I Reduced Sérum Medium (Thermo Fisher Scientific; Catalog 31985088) to 40* the desired final test concentration then serially diluted (1:3) up to 5 or 9 distinct concentrations, respectively. A 3.25-pL aliquot of each diluted compound was combined in a checkerboard fashion. This combination of compounds was mixed with 0.3 pL Lipofectamine® RNAiMAX Transfection Reagent (Thermo Fisher Scientific, Catalog 13778150) and 6.2 pL of Opti-MEM™ I Reduced Sérum Medium. After incubating for 20 minutes, the mixture was added to the cells. Space was also allotted for titrations of each compound alone as reference Controls. Cells were incubated with compounds for 3 days at 37°C in a 5% CO2 atmosphère. After that, HBsAg in the supematant of cell culture was assayed by ELIS A and cell viability was measured with Cell Titer Glow, the same procedures as in HepG2.2.15 in vitro assay section. The HBsAg réduction synergy between two test articles were analyzed using MacSynergy Software.
These results demonstrate that a combination therapy with ASO 1 and ds-siNA-0160 resulted in a greater réduction in sérum HBsAg as compared to treatment with ASO 1 or dssiNA-0160 alone.
Example 18. siNA Activity Assays
This example évaluâtes the activity of the siNAs disclosed in Table 10 (as identified by the ds-siNA ID). siRNAs were synthesized as described in Example 1. A conjugated moiety (e.g., ligand monomer) was further conjugated to the 3’ end of the sense strand (note: for dssiNA-067 and ds-siNA-083, the ligand monomer was conjugated to the 5’ end of the sense strand). A 5’-stabilized end cap was further attached to the 5’ end of the antisense strand of some siRNAs.
In vitro Assay.
Homo sapiens HepG2.2.15 cells were cultured in Dulbecco’s Modified Eagle’s Medium (DMEM) (ATCC 30-2002) supplemented to also contain 10% fêtai calf sérum (FCS). Cells were incubated at 37°C in an atmosphère with 5% CO2 in a humidified incubator. For transfection of HepG2.2.15 cells with HBV targeting siRNAs, cells were seeded at a density of 15000 cells/well
166 in 96-well regular tissue culture plates. Transfection of cells was carried out using RNAiMAX (Invitrogen/Life Technologies) according to the manufacturer’s instructions. Dose-response experiments were done with oligo concentrations of 40, 20, 10, 5, 2.5, 1.25, 0.625, 0.3125, 0.15625 and 0.07813nM. For each HBV targeting siRNA treatment (e.g., ds-siRNA, as identified by the ds-siNA ID in Table 6), four wells were transfected in parallel, and individual data points were collected from each well. After 24h of incubation with siRNA, media was removed, and cells were lysed and analyzed with a QuantiGene2.0 branched DNA (bDNA) probe set spécifie for HBV génotype D (also called Hepatitis B virus subtype ayw, complété genome of 3182 basepairs) as présent in cell line HepG2.2.15.
For each well, the HBV on-target mRNA levels were normalized to the GAPDH mRNA level. As shown in Table 10, the activity of the HBV targeting ds-siRNAs was expressed as EC50, 50% réduction of normalized HBV RNA level from no drug control, where A = EC50 < 5 nM; B = 5 nM < EC50 < 10; C = EC50 > 10. As shown in Table 10, the cytotoxicity of the HBV targeting ds-siRNAs was expressed by CC50 of 50% réduction of GAPDH mRNA from no drug control.
In vivo Assay:
GalNAc conjugated siRNA with or without 5’- stabilized end caps were subcutaneously injected at a single dose of 5mg/kg into AAV-HBV mi ce. The target knockdown magnitude was measured from sérum. The resulting max HBsAg knockdown (logio) is presented in Table 10, where X > 1 logio réduction in HBsAg, Y is 0.5 - 1 logio réduction in HBsAg, and Z is < 0.5 logio réduction in HBsAg.
Example 19: Analysis of 5’-stabilized end cap on the effïcacy of siNAs
In this example, the rôle of a 5’-stabilized end cap on the effïcacy of siNAs was investigated. Specifically, the first nucléotide on the 5’ end of the antisense strand was modified to contain a 5’-stabilized end cap. The ds-siNAs investigated in this example are shown in the table below:
ds-siNA Sense Strand Sequence (5’—>3 ’) Antisense Strand Sequence (5’—>3 ’)
ID
ds-siNA- mUpsmGpsfUmGmCmAfCfUmUmCmG mApsfGpsmGmUmGmAmAmGmCm
0166 mCmUmUmCmAfCmCmU-p-(PS)2- GmAmAmGfUmGmCmAmCmApsm
GalNAc4 (SEQ ID NO: 615) CpsmG (SEQ ID NO: 303)
ds-siNA- mUpsmGpsfUmGmCmAfCfUmUmCmG vmApsfGpsmGmUmGmAmAmGmC
0155 mCmUmUmCmAfCmCmU-p-(PS)2- mGm AmAmGfU mGmCm AmCm Aps
GalNAc4 (SEQ ID NO: 615) mCpsmG (SEQ ID NO: 525)
167 ds-siNA- mUpsmGpsfUmGmCmAfCfUmUmCmG d2vmApsfGpsmGmUmGmAmAmG
0157 mCmUmUmCmAfCmCmU-p-(PS)2- mCmGmAmAmGfUmGmCmAmCm
GalNAc4 (SEQ ID NO: 615) ApsmCpsmG (SEQ ID NO: 529) mX = 2’-O-methyl nucléotide; fX = 2’-fluoro nucléotide; vmA = 5’-vinyl phosphonate T-Omethyl adenosine; d2vmA = deuterated 5’ vinyl phosphonate adenosine; ps = phosphorothioate linkage
AAV-HBV mice were subcutaneously injected with vehicle or ds-siNAs. ds-siNA-0166, ds-siNA-0155, or ds-siNA-0157 were subcutaneously injected at a single dose of 5mg/kg into AAV-HBV mice. The target knockdown magnitude is measured from sérum. As shown in FIG.
9, the presence of the 5’ stabilized end cap in the first nucléotide from the 5’ end of the antisense strand in ds-siNA-0155 (triangle) and ds-siNA-0157 (square) improved the efficacy of the siNA (squares and triangles) as compared to the siNA without the 5’ stabilized end cap (ds-siNA-0166, diamond). In addition, the presence of the deuterated 5’ vinyl phosphonate in ds-siNA-0157 resulted in a greater improvement in efficacy of a ds-siNA as compared to the presence of the 5’ io vinylphosphanate in ds-siNA-0155. These results demonstrate that a 5’ stabilized end cap improves the efficacy of siNAs, with the greatest improvement seen in siNAs containing deuterated 5’ vinyl phosphonate.
Example 20: Analysis of HBV siRNA S and X Combination Therapy
In this example, combination therapy using an siNA targeting the S gene of HBV and an 15 siNA targeting the X gene of HBV was examined. AAV-HBV mice were treated with vehicle, a single siNA therapy, or a combination siNA therapy targeting the S gene and X gene of HBV. AAV-HBV mice were subcutaneously injected with a single dose of ds-siNA-0160 or ds-siNA0165 on day 0. For the combination siNA therapy, AAV-HBV mice were subcutaneously injected with a single dose of 1.5 mg/kg of ds-siNA-0165 (S trigger) and 1.5 mg/kg of ds-siNA-
0 1 60 (X trigger) on day 0. As shown in FIG. 10, the combination therapy with a siNA targeting the S gene and a siNA targeting the X gene was more potent than the single therapy with dssiNA-0165 or ds-siNA-0160.
168
Example 21. Synthesis of Monomer
Scheme 1
Préparation of (2a): To a solution of la (10.0 g, 29.5 mmol) in ACN (200.0 mL), KSAc (13.5 g, 118.6 mmol) was added at r.t., the mixture was stirred at r.t. for 15 h, TLC showed la was consumed completely. Mixture was fïltered by silica gel and filter cake was washed with DCM (100.0 mL), the filtrate was concentrated to give crude 2a (11.1 g) as an oil. 'H-NMR (400 MHz, CDCh): δ 7.32-7.24 (m, 5H), 7.16 (d, J= 8.9 Hz, 4H), 6.82 (d, J= 8.9 Hz, 4H), 3.82 (s, 6H), 2.28 (s, 3H).
Préparation of (3a): To a solution of crude 2a (11.1 g, 29.2 mmol) in THF (290.0 mL), L1AIH4 (2.0 g, 52.6 mmol) was added at 0°C and kept for 10 min, reaction was stirred at r.t. for 5 h under N2, TLC showed 2a was consumed completely. Mixture was put into aqueous NaHCCh solution and extracted with EA (500.0 mL*2), organic phase was concentrated to give crude which was purified by column chromatography (S1O2, PE/EA = 30:1 to 10:1) to give 3a (8.1g, 95% purity) as a white solid. ESI-LCMS: m/z 335.3 [M-H]’ ; 'H-NMR (400 MHz, CDCh): δ 7.33-7.24 (m, 5H), 7.19 (d, J= 8.8 Hz, 4H), 6.82 (d, J = 8.8 Hz, 4H), 3.83 (s, 6H), 3.09 (s, 1H).
Préparation of (2): To a solution of 1 (20.0 g, 81.3 mmol) inpyridine (400.0 mL), MsCl (10.23 g, 89.43 mmol) was added dropwise at -10°C, reaction was stirred at -10°C for 1 h, LCMS showed 1 was consumed completely, 100.0 mL aqueous NaHCCh solution was added and extracted with DCM (100.0 mL*2), organic phase was concentrated to give crude which was . purified by column chromatography (S1O2, DCM/MeOH = 30:1 to 10:1) to give 2 (9.5 g, 97% purity) as a white solid. ESI-LCMS: m/z 325.3 [M+H]+; 'H-NMR (400 MHz, DMSO-îZô): δ 11.45 (s, 1H), 7.64-7.62 (d, J= 8.0 Hz, 1H), 5.92-5.85 (m, 2H), 5.65-5.63 (d, J= 8.0 Hz, 1H), 5.26-5.11 (m, 1H), 4.53-4.37 (m, 2H), 4.27-4.16 (m, 1H), 4.10-4.04 (m, 1H), 3.23 (s, 3H).
169
Préparation of (3): Intermediate 3 was prepared by prepared according to reaction condition described in reference Helvetica Chimica Acta, 2004, 87. 2812. To a solution of 2 (9.2 g, 28.3 mmol) in dry DMSO (130.0 mL). DMTrSH (14.31 g, 42.5 mmol) was added, followed by tetramethylguanidine (3.6 g, 31.2 mmol) was added under N2, reaction was stirred at r.t. for 3 h, LCMS showed 2 was consumed completely. 100.0 mL H2O was added and extracted with EA (100.0 mL*2), organic phase was concentrated to give crude which was purified by column chromatography (SiO2, PE/EA = 5:1 to 1:1) to give 3 (12.0 g, 97% purity) as a white solid. ESILCMS: m/z 563.2 [Μ-H]’; ‘H-NMR (400 MHz, DMSO-t/6): δ 11.43-11.42 (d,7 = 4.0 Hz, 1H), 7.57-7.55 (d, J= 8.0 Hz, 1H), 7.33-7.17 (m, 9H), 6.89-6.86 (m, 4H), 5.80-5.74 (m, 1H), 5.6510 5.62 (m, 1H), 5.58-5.57 (d, J= 4.0 Hz, 1H), 5.16-5.01 (m, 1H), 3.98-3.90 (m, 1H), 3.73 (s, 6H),
3.73-3.67 (m, 1H), 2.50-2.37 (m, 2H).
Préparation of Example 21 monomer: To a solution of 3 (10.0 g, 17.7 mmol) in dichloromethane (120.0 mL) with an inert atmosphère of nitrogen was added CEOP[N(iPr)2]2 (6.4 g, 21.2 mmol) and DCI (1.8 g, 15.9 mmol) in order at room température. The resulting solution was stirred for 1.0 h at room température and diluted with 50 mL dichloromethane and washed with 2 x 50 mL of saturated aqueous sodium bicarbonate and 1 x 50 mL of saturated aqueous sodium chloride respectively. The organic phase was dried over anhydrous sodium sulfate, filtered and concentrated till no residual solvent left under reduced pressure. The residue was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, Cl8 silica gel; mobile phase, CH3CN/H2O (0.5% NH4HCO3) = 1/1 increasing to CH3CN/H2O (0.5% NH4HCO3) = 1/0 within 20 min, the eluted product was collected at CH3CN/ H2O (0.5% NH4HCO3) = 6/1; Detector, UV 254 nm. This resulted in to give Example 21 monomer (12.8 g, 98% purity, 93% yield) as an oil. ESI-LCMS: m/z 765.2 [M+H]+; ‘H-NMR (400 MHz, DMSOd6\. δ 11.44 (s, 1H), 7.70-7.66 (m, 1H), 7.32-7.18 (m, 9H), 6.89-6.85 (m, 4H), 5.80-5.64 (m, 2H),
5.38-5.22 (m, 1H), 4.38-4.15 (m, 1H), 3.81-3.70 (m, 8H), 3.61-3.43 (m, 3H), 2.76-2.73 (m, 1H),
2.66-2.63 (m, 1H), 2.50-2.41 (m, 2H), 1.12-1.05 (m, 9H), 0.97-0.95 (m, 3H); 3IP-NMR (162 MHz, DMSO-î/ô): δ 149.01, 148.97, 148.74, 148.67; 19F-NMR (376 MHz, DMSO-î/6): δ 149.01, 148.97, 148.74, 148.67.
Example 22. Synthesis of Monomer
170
Example 22 monomer
Scheme-2
Préparation of (2): To a stirred solution of 1 (2.0 g, 8.8 mmol) in pyridine (20 mL) were added DMTrCl (3.3 g, 9.7 mmol) at r.t. The reaction mixture was stirred at r.t. for 2.5 hrs. With ice-bath cooling, the reaction was quenched with water and the product was extracted with EA (100 mL). The organic phase was evaporated to dryness under reduced pressure to give a residue which was purified by silica gel column chromatography (eluent, DCM: MeOH=50:l~20:l) to give 2 (3.7 g, 7.2 mmol, 80.1%) as a white solid. ESI-LCMS: m/z 527 [M-H]'.
Préparation of (3): To the solution of 2 (2.8 g, 5.3 mmol) in dry DMF (56 mL) was added (CD3O)2Mg (2.9 g, 31.8 mmol) at r.t. under N2 atmosphère. The reaction mixture was stirred at 100°C for 15 hrs. With ice-bath cooling, the reaction was quenched with saturated aq. NH4CI and extracted with EA (300 mL). The combined organic layer was washed with water and brine, dried over Na2SO4, and concentrated to give a residue which was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, Cl8 silica gel; mobile phase, CH3CN/H2O (0.5% NH4HCO3) = 2/3 increasing to CH3CN/H2O (0.5% NH4HCO3) = 3/2 within 20 min, the eluted product was collected at CH3CN/ H2O (0.5% NEUHCCh) = 1/1; Detector, UV 254 nm. This resulted in to give 3 (2.0 g, 3.6 mmol, 67.9%) as a white solid. ESI-LCMS: m/z 562 [M-H]'; Ή-NMR (400 MHz, DMSO-76): δ 11.38 (s, 1H), 7.73 (d, J= 8 Hz, 1H), 7.46-7.19 (m, 9H), 6.91 (d, J= 7.4 Hz, 4H), 5.81-5.76 (AB, J= 20 Hz, 1H), 5.30 (d, J= 8 Hz, 1H), 5.22 (s, 1H), 4.254.15 (m, 1H), 3.99-3.92 (m, 1H), 3.85-3.79 (m, 1H), 3.74 (s, 6H), 3.34-3.18 (m, 31H).
Préparation of Example 22 monomer: To a suspension of 3 (2.0 g, 3.5 mmol) in DCM (20 mL) was added DCI (357 mg, 3.0 mmol) and CEP[N(iPr)2]2 (1.3 g, 4.3 mmol). The mixture was stirred at r.t. for 1 h. LC-MS showed 3 was consumed completely. The solution was washed with water twice and washed with brine and dried over Na2SO4. Then concentrated to give a residue which was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, Cl 8 silica gel; mobile phase, CH3CN/H2O (0.5% NH4HCO3) = 1/1 increasing to CH3CN/H2O (0.5% NH4HCO3) = 1/0 within 20 min, the eluted product was collected at CH3CN/ H2O (0.5% NH4HCO3) = 1/0; Detector, UV 254 nm. This resulted in to give Example 22 monomer (2.1 g, 2.7 mmol, 77.1%) as a white solid. ESI-LCMS: m/z 764 [M+H]+ ; 'H-NMR
171 (400 MHz, ACN-i/3): δ 9.45-8.90 (m, 1H, exchanged with D2O), 7.88-7.66 (m, 1H), 7.50-7.18 (m, 9H), 6.93-6.80 (m, 4H), 5.85 (d, J= 8.2 Hz, 1H),5.29-5.16 (m, 1H), 4.57-4.37 (m, 1H), 4.184.09 (m, 1H), 3.98-3.90 (m, 1H), 3.90-3.74 (m, 7H), 3.74-3.50 (m, 3H), 3.48-3.31 (m, 2H), 2.702.61 (m, 1H), 2.56-2.46 (m, 1H), 1.24-1.12 (m, 9H), 1.09-0.99 (m, 3H). 31P-NMR (162 MHz, 5 ACN-J3): δ= 149.87, 149.55.
Ëxample 23. Synthesis of Monomer
Example 23 monomer
Scheme-3
Préparation of (2): To the solution of 1 (39.2 g, 151.9 mmol) in DMF (390.0 mL) was added imidazole (33.0 g, 485.3 mmol) and TBSC1 (57.2 g, 379.6 mmol) at 0°C. The reaction mixture was stirred at room température for 15 hrs under N2 atmosphère. After addition of water, the resulting mixture was extracted with EA (500.0 mL). The combined organic layer was washed with water and brine, dried over Na2SO4, concentrated to give the crude 2 (85.6 g) as a white solid which was used directly for next step. ESI-LCMS: m/z 487.7 [M+H]+.
Préparation of (3): A solution of crude 2 (85.6 g) in a mixture solvent of TFA/H2O = 1/1 (400.0 mL) and THF (400.0 mL) was stirred at 0°C for 30 min. After completion of reaction, the resulting mixture was added con.NH3*H2O to pH = 7, and then extracted with EA (500.0 mL). The organic layer was washed with brine, dried over sodium sulfate and removed to give the residue was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, 172
Cl8 silica gel; mobile phase, CH3CN/H2O (0.5% NH4HCO3) = 2/3 increasing to CH3CN/H2O (0.5% NH4HCO3) = 3/2 within 20 min, the eluted product was collected at CH3CN/H2O (0.5% NH4HCO3) = 1/1; Detector, UV 254 nm. This resulted in to give 3 (36.6 g, 98.4 mmol, 64.7% over two step) as a white solid. ESI-LCMS: m/z 372.5 [M+H]+; [H-NMR (400 MHz, DMSO-<76): δ 11.36 (d, 1 Hz, 1H), 7.92 (d, J=8Hz, 1H), 5.83 (d, J=5Hz, 1H),
5.67-5.65 (m, 1H), 5.19 (s, 1H), 4.30 (t, J= 5 Hz, 1H), 3.85-3.83 (m, 2H), 3.68-3.52 (m, 2H), 0.88 (s, 9H), 0.09 (s, 6H).
Préparation of (4): To the solution of 3 (36.6 g, 98.4 mmol) in dry DCM (200.0 mL) and DMF (50.0 mL) was added PDC (73.9 g, 196.7 mmol), tert-butyl alcohol (188.0 mL) and AC2O (93.0 mL) at r.t under N2 atmosphère, the reaction mixture was stirred at r.t for 2 hrs. The solvent was removed to give a residue which was purified by silica gel column chromatography (eluent, PE/EA = 4:1 ~ 2:1) to give a residue which was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, Cl8 silica gel; mobile phase, CH3CN/H2O (0.5% NH4HCO3) = 1/1 increasing to CH3CN/H2O (0.5% NH4HCO3) = 1/0 within 20 min, the eluted product was collected at CH3CN/ H2O (0.5% NH4HCO3) = 1/0; Detector, UV 254 nm. This resulted in to give 4 (24.3 g, 54.9 mmol, 55.8%) as a white solid. ESI-LCMS: m/z 443.2 [M+H]+; *H-NMR (400 MHz, DMSO-cZô): δ 11.30 (d, J= 1 Hz, 1H), 7.92 (d, J = 8 Hz, 1H), 5.86 (d, J= 6 Hz, 1H), 5.675.65 (m, 1H), 4.33-4.31 (m, 1H), 4.13 (d, J= 3 Hz, 1H), 3.73-3.70 (m, 1H), 1.34 (s, 9H), 0.77 (s, 9H), 0.08 (s, 6H).
Préparation of (5): To the solution of 4 (18.0 g, 40.7 mmol) in dry THF/MeOD/D2O = 10/2/1 (145.0 mL) was added NaBÜ4 (5.1 g, 122.1 mmol) three times during an hour at 50°C, the reaction mixture was stirred at r.t. for 2 hrs. After completion of reaction, adjusted pH value to 7 with CH3COOD, after addition of water, the resulting mixture was extracted with EA (300.0 mL). The combined organic layer was washed with water and brine, dried over Na2SO4, concentrated to give a residue which was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, Cl 8 silica gel; mobile phase, CH3CN/H2O (0.5% NH4HCO3) = 2/3 increasing to CH3CN/H2O (0.5% NH4HCO3) = 3/2 within 20 min, the eluted product was collected at CH3CN/ H2O (0.5% NH4HCO3) = 1/1; Detector, UV 254 nm. This resulted in to give 5 (10.4 g, 27.8 mmol, 68.3%) as a white solid. ESI-LCMS: m/z 375.2 [M+H]+; ’H-NMR (400 MHz, DMSO-t/ô): δ 11.36 (d, J= 1 Hz, 1H), 7.92 (d, J= 8 Hz, 1H), 5.83 (d, J= 5 Hz, 1H), 5.675.65 (m, 1H), 5.19 (s, 1H), 4.30 (t, J= 5 Hz, 1H), 3.85-3.83 (m, 2H), 0.88 (s, 9H), 0.09 (s, 6H).
Préparation of (6): To a stirred solution of 5 (10.4 g, 27.8 mmol) in pyridine (100.0 mL) was added DMTrCl (12.2 g, 36.Immol) at r.t., The reaction mixture was stirred at r.t. for 2.5 hrs, the reaction was quenched with water and extracted with EA (200.0 mL). The organic phase was
173 evaporated to dryness under reduced pressure to give a residue which was purified by FlashPrep-HPLC with the following conditions (IntelFlash-1): Column, C18 silica gel; mobile phase, CH3CN/H2O (0.5% NH4HCO3) = 1/1 increasing to CH3CN/H2O (0.5% NH4HCO3) = 1/0 within 20 min, the eluted product was collected at CH3CN/ H2O (0.5% NH4HCO3) = 1/0; Detector, UV 254 nm. This resulted in to give 6 (13.5 g, 19.9 mmol, 71.6%) as a white solid. ESI-LCMS: m/z 677.8 [M+H]+; ‘H-NMR (400 MHz, DMSO-76): δ 11.39 (d, J= 1 Hz, 1H), 7.86 (d, J= 4 Hz, 1H), 7.35-7.21 (m, 9H), 6.90-6.88 (m, 4H), 5.78 (d, J= 2 Hz, 1H), 5.30-5.27 (m, 1H), 4.33-4.30 (m, 1H ), 3.91 (d, J= 7 Hz, 1H), 3.85-3.83 (m, 1H), 3.73 (s, 6H), 3.38 (s, 3H), 0.77 (s, 9H), 0.03 (s, 3H), 0.01 (s, 3H).
Préparation of (7): To a solution of 6 (13.5 g, 19.9 mmol) in THF (130.0 mL) was added 1 M TBAF solution (19.0 mL). The reaction mixture was stirred at r.t. for 1.5 hrs. LC-MS showed 6 was consumed completely. Water (500.0 mL) was added and extracted with EA (300.0 mL), the organic layer was washed with brine and dried over Na2SÛ4. Then the organic layer was concentrated to give a residue which was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, Cl 8 silica gel; mobile phase, CH3CN/H2O (0.5% NH4HCO3) = 2/3 increasing to CH3CN/H2O (0.5% NH4HCO3) = 3/2 within 20 min, the eluted product was collected at CH3CN/ H2O (0.5% NH4HCO3) = 1/1; Detector, UV 254 nm. This resulted in to give 7 (10.9 g, 19.4 mmol, 97.5%) as a white solid. ESI-LCMS: m/z 563.6 [M+H]+; ‘H-NMR (400 MHz, DMSO-î/ô): δ 11.39 (s, 1H), 7.23 (d, J= 8 Hz, 1H), 7.73 (d, J= 8 Hz, 1H), 7.36-7.23 (m, 9H), 6.90 (d, J= 8 Hz, 4H), 5.81 (d, J= 3 Hz, 1H), 5.30-5.28 (m, 1H), 5.22 (d, J= 7 Hz, 1H), 4.20 (q, 7=7Hz, 1H), 3.93 (d, 7= 7 Hz, 1H), 3.81 (t,7=5Hz, 1H), 3.74 (s, 6H), 3.41 (s, 3H).
Préparation of Example 23 monomer: To a suspension of 7 (10.9 g, 19.4 mmol) in DCM (100.0 mL) was added DCI (1.8 g, 15.7 mmol) and CEP[N(iPr)2]2 (6.1 g, 20.4 mmol). The mixture was stirred at r.t. for 1 h. LC-MS showed 7 was consumed completely. The mixture was washed with water twice and brine, dried over Na2SO4. Then concentrated to give a residue which was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, Cl 8 silica gel; mobile phase, CH3CN/H2O (0.5% NH4HCO3) = 1/1 increasing to CH3CN/H2O (0.5% NH4HCO3) = 1/0 within 20 min, the eluted product was collected at CH3CN/ H2O (0.5% NH4HCO3) = 1/0; Detector, UV 254 nm. This resulted in to give Example 23 monomer (12.5 g, 14.5 mmol, 74.7%) as a white solid. ESI-LCMS: m/z 863.6 [M+H]+; ‘H-NMR (400 MHz, DMSO-76): δ 11.39 (s, 1H), 7.81-7.55 (m, 1H), 7.40-7.22 (m, 9H), 6.92-6.87 (m, 4H), 5.83-5.80 (m, 1H), 5.32-5.25 (m, 1H), 4.46-4.34 (m, 1H), 4.10-3.98 (m, 2H), 3.84-3.73 (m, 7H), 3.60-3.50 (m, 3H), 3.42, 3.40 (s, 3H), 2.78 (t, 7= 6 Hz, 1H), 2.62-2.59 (m, 1H), 2.07 (s, 1H), 1.17-0.96 (m, 12H); 31P-NMR (162 MHz, DMSO-76): δ 149.37, 149.06.
174
Example 24. Synthesis of Monomer
HO' F imidazole
TBSC1
DMF__
THF/TFA/H2O
PDC,tert-Butanol
NaBD4
THF/MeOH-d/D2O
DMTrCl
Pyridine
5
TBSO' F
TBAF
THF
HO' F
DC1
CEP[N(iPr)2]2 DCM
Scheme-4
Préparation of (2): To the solution ofl (13.0 g, 52.8 mmol) in DMF (100 mL) was added imidazole (12.6 g, 184.8 mmol) and TBSC1 (19.8 g, 132.0 mmol) at 0 °C, and the reaction mixture was stirred at room température for 15 h under N2 atmosphère. After addition of water, the resulting product was extracted with EA (500 mL). The combined organic layer was washed with water and brine, dried over Na2SO4, and concentrated to give the crude 2 (30.6 g) as a white solid which was used directly for next step. ESI-LCMS: m/z 475 [M+H]+. W02017106710A1
Préparation of (3): A solution of crude 2 (30.6 g) in a mixture solvent of TFA/H2O = 1/1 (100 mL) and THF (100 mL) was stirred at 0 °C for 30 min. After completion of reaction, the resulting mixture was added con.NH3*H2O to pH = 7.5, and then the mixture was extracted with EA (500 mL), the organic layer was washed with brine, dried over Na2SÛ4 and removed to give the residue was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, Cl8 silica gel; mobile phase, CH3CN/H2O (0.5% NH4HCO3) = 2/3 increasing to CH3CN/H2O (0.5% NH4HCO3) = 3/2 within 20 min, the eluted product was collected at CH3CN/ H2O (0.5% NH4HCO3) = 1/1; Detector, UV 254 nm. This resulted in to give 3 (12.0 g, 33.3 mmol, 65.8% over two step) as a white solid. ESI-LCMS: m/z 361 [M+H]+; 'H-NMR (400 MHz,
175
DMSO-e/ô): δ 11.39 (s, J= 1 Hz, 1H, exchanged with D2O), 7.88 (d, J= 8 Hz, 1H), 5.91-5.86 (m, 1H), 5.66-5.62 (m, 1H), 5.21 (t, 5.2 Hz, 1H, exchanged with D2O), 5.18-5.03 (m, 1H), 4.37-
4.29 (m, 1H), 3.87-3.83 (m, 1H), 3.78-3.73 (m, 1H), 3.56-3.51 (m, 1H), 0.87 (s, 9H), 0.09 (s, 6H). W02017106710A1.
Préparation of (4): To the solution of 3 (11.0 g, 30.5 mmol) in dry DCM (60 mL) and DMF (15 mL) was added PDC (21. g, 61.0 mmol), tert-butyl alcohol (45 mL) and Ac2O (32 mL) at r.t under N2 atmosphère. And the reaction mixture was stirred at r.t for 2 h. The solvent was removed to give a residue which was purified by silica gel column chromatography (eluent, PE: EA=4:1~2:1) to give a residue which was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, Cl 8 silica gel; mobile phase, CH3CN/H2O (0.5% NH4HCO3) = 1/1 increasing to CH3CN/H2O (0.5% NH4HCO3) = 1/0 within 20 min, the eluted product was collected at CH3CN/ H2O (0.5% NH4HCO3) = 1/0; Detector, UV 254 nm. This resulted in to give 4 (9.5 g, 22.0 mmol, 72.3%) as a white solid. ESI-LCMS: m/z 431 [M+H]+ ; ‘H-NMR (400 MHz, DMSO-c/6): δ 11.45 (s, J= 1 Hz, 1H, exchanged with D2O), 7.93 (d, J= 8.5 Hz, 1H), 6.02-5.97 (m, 1H), 5.76-5.74 (m, 1H), 5.29-5.14 (m, 1H), 4.59-4.52 (m, 1H), 4.29-4.27 (m, 1H), 1.46 (s, 9H), 0.89 (s, 9H), 0.12 (s, 6H).
Préparation of (5): To the solution of 4 (8.5 g, 19.7 mmol) in dry THF/MeOD/D2O = 10/2/1 (80 mL) was added NaBÜ4 (2.5 g, 59.1 mmol) three times per an hour at 50°C. And the reaction mixture was stirred at r.t for 2 h. After completion of reaction, adjusted pH value to 7 with CH3COOD, after addition of water, the resulting mixture was extracted with EA (300 mL). The combined organic layer was washed with water and brine, dried over Na2SO4, and concentrated to give a residue which was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, Cl8 silica gel; mobile phase, CH3CN/H2O (0.5% NH4HCO3) = 2/3 increasing to CH3CN/H2O (0.5% NH4HCO3) = 3/2 within 20 min, the eluted product was collected at CH3CN/ H2O (0.5% NH4HCO3) = 1/1; Detector, UV 254 nm. This resulted in to give 5 (3.5 g, 9.7 mmol, 50.3%) as a white solid. ESI-LCMS: m/z 363 [M+H]+; Ή-NMR (400 MHz, DMSO-î/ô): δ 11.41 (s, J= 1 Hz, 1H, exchanged with D2O), 7.88 (d, J= 8 Hz, 1H), 5.91-5.86 (m, 1H), 5.66-5.62 (m, 1H), 5.19 (t, J= 5.2 Hz, 1H, exchanged with D2O), 5.18-5.03 (m, 1H), 4.374.29 (m, 1H), 3.87-3.83 (m, 1H), 0.88 (s, 9H), 0.10 (s, 6H).
Préparation of (6): To a stirred solution of 5 (3.4 g, 9.7 mmol) in pyridine (35 mL) were added DMTrCl (3.4 g, 10.Immol) at r.t. And the reaction mixture was stirred at r.t for 2.5 h. With ice-bath cooling, the reaction was quenched with water and the product was extracted with EA (200 mL). The organic phase was evaporated to dryness under reduced pressure to give a residue which was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1):
176
Column, Cl8 silica gel; mobile phase, CH3CN/H2O (0.5% NH4HCO3) = 1/1 increasing to CH3CN/H2O (0.5% NH4HCO3) = 1/0 within 20 min, the eluted product was collected at CH3CN/ H2O (0.5% NH4HCO3) = 1/0; Detector, UV 254 nm. This resulted in to give 6 (PCT Int. Appl., 2019173602), (5.5 g, 8.3 mmol, 85.3%) as a white solid. ESI-LCMS: m/z 665 [M+H]+; ’H-NMR (400 MHz, DMSO-Jô): δ 11.50 (d, J= 1 Hz, 1H, exchanged with D2O), 7.92 (d, J= 4 Hz, 1H), 7.44-7.27 (m, 9H), 6.96-6.93 (m, 4H), 5.94 (d, J= 20.5 Hz, 1H), 5.39-5.37 (m, 1H), 5.32-5.17 (m, 1H ), 4.60-4.51 (m, 1H ), 4.01 (d, J= 8.8 Hz, 1H), 3.80 (s, 6H), 0.80 (s, 9H), 0.09 (s, 3H), 0.05 (s, 3H).
Préparation of (7): To a solution of 6 (5.5 g, 8.3 mmol) in THF (50 mL) was added 1 M TBAF solution (9 mL). The reaction mixture was stirred at r.t. for 1.5 h. LC-MS showed 6 was consumed completely. Water (500 mL) was added. The product was extracted with EA (300 mL) and the organic layer was washed with brine and dried over Na2SO4. Then the organic layer was concentrated to give a residue which was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, Cl 8 silica gel; mobile phase, CH3CN/H2O (0.5% NH4HCO3) = 2/3 increasing to CH3CN/H2O (0.5% NH4HCO3) = 3/2 within 20 min, the eluted product was collected at CH3CN/ H2O (0.5% NH4HCO3) = 1/1; Detector, UV 254 nm. This resulted in to give 7 (4.1 g, 7.5 mmol, 90.0%) as a white solid. ESI-LCMS: m/z 551 [M+H]+; ’H-NMR (400 MHz, DMSO-î/ô): δ 11.42 (s, 1H, exchanged with D2O), 7.76 (d, J= 8.2 Hz, 1H), 7.39-7.22 (m, 9H), 6.90-6.88 (m, 4H), 5.83 (d, J= 20.5 Hz, 1H), 5.65 (d, J= 7.0 Hz, 1H, exchanged with D2O), 5.29 (d, J= 7.2 Hz, 1H), 5.18-5.03 (m, 1H), 4.40-4.28 (m, 1H), 4.01 (d, J= 8.8 Hz, 1H), 3.74 (s, 6H).
Préparation of Example 24 monomer: To a suspension of 7 (4.1 g, 7.5 mmol) in DCM (40 mL) was added DCI (0.7 g, 6.4 mmol) and CEP[N(iPr)2]2 (2.9 g, 9.7 mmol). The mixture was stirred at r.t. for 1 h. LC-MS showed 7 was consumed completely. The solution was washed with water twice and washed with brine and dried over Na2SO4. Then concentrated to give a residue which was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, Cl 8 silica gel; mobile phase, CH3CN/H2O (0.5% NH4HCO3) = 1/1 increasing to CH3CN/H2O (0.5% NH4HCO3) = 1/0 within 20 min, the eluted product was collected at CH3CN/ H2O (0.5% NH4HCO3) = .1/0; Detector, UV 254 nm. This resulted in to give Example 24 monomer (5.0 g, 6.6 mmol, 90.0%) as a white solid. ESI-LCMS: m/z 751 [M+H]+; ’H-NMR (400 MHz, DMSO-Jô): δ 11.43 (s, 1H), 7.85-7.82 (m, 1H), 7.40-7.23 (m, 9H), 6.90-6.85 (m, 4H), 5.94-5.86 (m, 1H), 5.40-5.24 (m, 2H), 4.74-4.49 (m, 1H), 4.12-4.09 (m, 2H), 3.79-3.47 (m, 10H), 2.78-2.59 (m, 2H), 1.14-0.93 (m, 12H). 31P-NMR (162 MHz, DMSO-J6): δ 149.67, 149.61, 149.32, 149.27.
177
Example 25. Synthesis of Monomer
imidazole
TBSC1 DMF
DCA
DCM
DMTrCI
Pyridine
TBSO' 'OCD3
NaBD4 THF/MeOD/D2O
Example 25 monomer
Scheme-5
Préparation of (4): To the solution of 3 (14.3 g, 25.4 mmol, Scheme 2) in pyridine (150 mL) was added imidazole (4.5 g, 66.6 mmol) and TBSC1 (6.0 g, 40.0 mmol) at 0 °C, and the reaction mixture was stirred at room température for 15 h under N2 atmosphère. After addition of water, the resulting mixture was extracted with EA (500 mL). The combined organic layer was washed with water and brine, dried over Na2SÜ4, and concentrated to give the crude 4 (18.0 g) as a white solid which was used directly for next step. ESI-LCMS: m/z 676 [M-H]’.
Préparation of (5): To the solution of crude 4 (18.0 g) in the solution of DCA (6%) in DCM (200 mL) was added TES (50 mL) at r.t, and the reaction mixture was stirred at room température for 5-10 min. After completion of reaction, the resulting mixture was added pyridine to pH = 7, and then the solvent was removed and the residue was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, Cl 8 silica gel; mobile phase, CH3CN/H2O (0.5% NH4HCO3) = 2/3 increasing to CH3CN/H2O (0.5% NH4HCO3) = 3/2 within 20 min, the eluted product was collected at CH3CN/ H2O (0.5% NH4HCO3) = 1/1; Detector, UV 254 nm. This resulted in to give 5 (6.5 g, 17.2 mmol, 67.7% for two step) as a white solid. ESI-LCMS: m/z 376 [M+H]+ ; ’H-NMR (400 MHz, DMSO-c76): δ 7.92 (d, J= 8 Hz, 1H), 5.82 (d, J= 5.2 Hz, 1H), 5.68-5.63 (m, 1H), 5.20-5.15 (m, 1H), 4.32-4.25 (m, 1H), 3.87-3.80 (m, 2H), 3.69-3.61 (m, 1H), 3.57-3.49 (m, 1H), 0.88 (s, 9H), 0.09 (s, 6H).
178
Préparation of (6): To the solution of 5 (6.5 g, 17.2 mmol) in dry DCM (35 mL) and DMF (9 mL) was added PDC (12.9 g, 34.3 mmol), tert-butyl alcohol (34 mL) and Ac2O (17 mL) at r.t under N2 atmosphère. And the reaction mixture was stirred at r.t for 2 hrs. The solvent was removed to give a residue which was purified by silica gel column chromatography (eluent, PE: EA = 4:1-2:1) to give a residue which was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, Cl 8 silica gel; mobile phase, CH3CN/H2O (0.5% NH4HCO3) = 1/1 increasing to CH3CN/H2O (0.5% NH4HCO3) = 1/0 within 20 min, the eluted product was collected at CH3CN/ H2O (0.5% NH4HCO3) = 1/0; Detector, UV 254 nm. This resulted in to give 6 (5.5 g, 12.3 mmol, 70.1%) as a white solid. ESI-LCMS: m/z 446 [M+H]+ ; ‘H-NMR (400 MHz, DMSO-Jé): δ = 11.29 (s, 1H), 7.91 (d, J= 8.4 Hz, 1H), 5.85 (d, J= 6.4 Hz, 1H), 5.71-5.61 (m, 1H), 4.35-4.28 (m, 1H), 4.12 (d, J= 3.2 Hz, 1H), 3.75-3.67 (m, 1H), 1.33 (s, 9H), 0.76 (s, 9H), 0.00 (d, J = 1.6 Hz, 6H).
Préparation of (7): To the solution of 6 (5.4 g, 12.1 mmol) in THF/MeOD/D2O= 10/2/1 (44 mL) was added NaBÜ4 (1.5 g, 36.3 mmol) at r.t. and the reaction mixture was stirred at 50°C for 2 hrs. After completion of reaction, adjusted pH value to 7 with CH3COOD. Water was added, the resulting mixture was extracted with EA (500 mL). The combined organic layer was washed with water and brine, dried over Na2SO4, and concentrated to give a residue which was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, Cl8 silica gel; mobile phase, CH3CN/H2O (0.5% NH4HCO3) = 2/3 increasing to CH3CN/H2O (0.5% NH4HCO3) = 3/2 within 20 min, the eluted product was collected at CH3CN/ H2O (0.5% NH4HCO3) = 1/1; Detector, UV 254 nm. This resulted in to give 7 (2.6 g, 6.8 mmol, 56.1%) as a white solid. ESI-LCMS: m/z 378 [M+H]+ ; ‘H-NMR (400 MHz, DMSO-t/6): δ 11.35 (s, 1H), 7.91 (d, J= 8.0 Hz, 1H), 5.82 (d, J= 5.2 Hz, 1H), 5.69-5.60 (m, 1H), 5.14 (s, 1H), 4.34-4.20 (m, 1H), 3.88-3.76 (m, 2H), 0.87 (s, 9H), 0.08 (s, 6H).
Préparation of (8): To a stirred solution of 7 (2.6 g, 6.8 mmol) in pyridine (30 mL) were added DMTrCl (3.5 g, 10.3 mmol) at r.t. And the reaction mixture was stirred at r.t. for 2.5 hrs. With ice-bath cooling, the reaction was quenched with water and the product was extracted into EA (200 mL). The organic phase was evaporated to dryness under reduced pressure to give a residue which was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, Cl8 silica gel; mobile phase, CH3CN/H2O (0.5% NH4HCO3) = 1/1 increasing to CH3CN/H2O (0.5% NH4HCO3) = 1/0 within 20 min, the eluted product was collected at CH3CN/ H2O (0.5% NH4HCO3) = 1/0; Detector, UV 254 nm. This resulted in to give 8 (4.3 g, 6.3 mmol, 90.1%) as a white solid. ESI-LCMS: m/z 678 [M-H]'; ‘H-NMR (400 MHz, DMSO-tZ6): δ 11.39 (s, 1H), 7.86 (d, J= 8.0 Hz, 1H), 7.42-7.17 (m, 9H), 6.96-6.83 (m, 4H), 5.82-5.69 (m, 2H), 5.29
179 (d, 8.4 Hz, 1H), 4.36-4.25 (m, 1H), 3.90 (d, J= 7.2 Hz, 1H), 3.86-3.80 (m, 1H), 3.73 (s, 6H),
0.75 (s, 9H), 0.02 (s, 3H), -0.04 (s, 3H).
Préparation of (9): To a solution of 8 (4.3 g, 6.3 mmol) in THF (45 mL) was added 1 M TBAF solution (6 mL). The reaction mixture was stirred at r.t. for 1.5 hrs. LCMS showed 8 was consumed completely. Water (200 mL) was added. The product was extracted with EA (200 mL) and the organic layer was washed with brine and dried over Na2SC>4. Then the organic layer was concentrated to give a residue which was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, Cl 8 silica gel; mobile phase, CH3CN/H2O (0.5% NH4HCO3) = 2/3 increasing to CH3CN/H2O (0.5% NH4HCO3) = 3/2 within 20 min, the eluted product was collected at CH3CN/ H2O (0.5% NH4HCO3) = 1/1; Detector, UV 254 nm. This resulted in to give 8 (3.5 g, 6.1 mmol, 90.1%) as a white solid. ESI-LCMS: m/z 678 [M-H]'; ‘H-NMR (400 MHz, DMSO-î/ê): δ 11.38 (d, J= 2.0 Hz, 1H), 7.23 (d, J= 8.0 Hz, 1H), 7.41-7.19 (m, 9H), 6.94-6.85 (m, 4H), 5.81 (d, J= 4.0 Hz, 1H), 5.33-5.26 (m, 1H), 5.21 (d, J= 7.2 Hz, 1H), 4.06-3.90 (m, 2H), 3.83-3.77 (m, 1H), 3.74 (s, 6H).
Préparation of Example 25 monomer: To a suspension of 9 (2.1 g, 3.7 mmol) in DCM (20 mL) was added DCI (373 mg, 3.1 mmol) and CEP[N(iPr)2]2 (1.3 g, 4.4 mmol). The mixture was stirred at r.t. for 1 h. LC-MS showed 9 was consumed completely. The solution was washed with water twice and washed with brine and dried over Na2SO4. Then concentrated to give a residue which was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, Cl8 silica gel; mobile phase, CH3CN/H2O (0.5% NH4HCO3) = 1/1 increasing to CH3CN/H2O (0.5% NH4HCO3) = 1/0 within 20 min, the eluted product was collected at CH3CN/ H2O (0.5% NH4HCO3) = 1/0; Detector, UV 254 nm. This resulted in to give Example 25 monomer (2.2 g, 3.5 mmol, 80%) as a white solid. ESI-LCMS: m/z 766 [M+H]+; 'H-NMR (400 MHz, ACN-i/3): δ 9.65-8.86 (m, 1H, exchanged with D2O), 7.93-7. 68 (m, 1H), 7.52-7.19 (m, 9H), 6.94-6.78 (m, 4H), 5.95-5.77 (m, 1H), 5.31-5.17 (m, 1H), 4.61-4.37 (m, 1H), 4.20-4.07 (m, 1H), 4.01-3.51 (m, 10H), 2.74-2.59 (m, 1H), 2.57-2.43 (m, 1H), 1.27-1.10 (m, 9H), 1.09-0.95 (m, 3H). 31P-NMR (162 MHz, ACN-J3): δ= 149.88, 149.55.
Example 26. Synthesis of Monomer
180
Example 26 monomer
Scheme-6
Préparation of (7): To a solution of 6 (17 g, 25.1 mmol, Scheme 3) in ACN (170 mL) was added DMAP (6.13 g, 50.3 mmol) and TEA (5.1 g, 50.3 mmol, 7.2 mL), Then added TPSC1 (11.4 g, 37.7 mmol) at 0 °C under N2 atmosphère and the mixture was stirred at r.t. for 3 h under N2 atmosphère. Then con. NH3.H2O (27.3 g, 233.7 mmol) was added at r.t. and the mixture was stirred at r.t. for 16 h. The reaction was quenched with water and the product was extracted with EA (200 mL). The organic phase was concentrated to give the crude 7 (17.0 g) as a white solid which was used directly for next step.
Préparation of (8): To a stirred solution of 7 (17.0 g, 25.1 mmol) in pyridine (170 mL) were added BzCl (4.3 g, 30. Immol) 0 °C under N2 atmosphère. And the reaction mixture was stirred at r.t for 2.5 h. With ice-bath cooling, the reaction was quenched with water and the product was extracted with EA (200 mL). The organic phase was evaporated to dryness under reduced pressure to give a residue which was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, Cl 8 silica gel; mobile phase, CH3CN/H2O (0.5% NH4HCO3) = 1/1 increasing to CH3CN/H2O (0.5% NH4HCO3) = 1/0 within 20 min, the eluted product was collected at CH3CN/ H2O (0.5% NH4HCO3) = 1/0; Detector, UV 254 nm. This resulted in to give 8 (19.0 g, 24.3 mmol, 95.6% over two step) as a white solid. ESI-LCMS: m/z 780 [M+H]+.
Préparation of (9): To a solution of 8 (19.0 g, 24.3 mmol) in THF (190 mL) was added 1 M TBAF solution (24 mL). The reaction mixture was stirred at r.t. for 1.0 h. LC-MS showed 8 was consumed completely. Water (500 mL) was added. The product was extracted with EA (300 mL) and the organic layer was washed with brine and dried over Na2SO4. Then the organic layer was concentrated to give a residue which was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, C18 silica gel; mobile phase, CH3CN/H2O (0.5% NH4HCO3) = 2/3 increasing to CH3CN/H2O (0.5% NH4HCO3) = 3/2 within 20 min, the eluted product was 181 collected at CH3CN/ H2O (0.5% NH4HCO3) = 1/1; Detector, UV 254 nm. This resulted in to give 9 (15.2 g, 23.1 mmol, 95.5%) as a white solid. ESI-LCMS: m/z 666 [M+H]+; ‘H-NMR (400 MHz, DMSO-î/6): δ 11.28 (s, 1H), 8.41 (m, 1H), 8.00-7.99 (m, 2H),7.63-7.15 (m, 13H), 6.936.89 (m, 4H), 5.87(s, 1H), 5.20(d, J= 7.4 Hz, 1H), 4.30 (m, 1H), 4.02 (m, 1H), 3.75 (s, 7H), 3.53 5 (s, 3H).
Préparation of Example 26 monomer: To a suspension of 9 (10.0 g, 15.0 mmol) in DCM (100 mL) was added DCI (1.5 g, 12.7 mmol) and CEP[N(iPr)2]2 (5.4 g, 18.0 mmol). The mixture was stirred at r.t. for 1 h. LC-MS showed 9 was consumed completely. The solution was washed with water twice and washed with brine and dried over Na2SO4. Then concentrated to give a residue which was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, Cl 8 silica gel; mobile phase, CH3CN/H2O (0.5% NH4HCO3) = 1/1 increasing to CH3CN/H2O (0.5% NH4HCO3) = 1/0 within 20 min, the eluted product was collected at CH3CN/ H2O (0.5% NH4HCO3) = 1/0; Detector, UV 254 nm. This resulted in to give Example 26 monomer (11.5 g, 13.5 mmol, 90.7%) as a white solid. ESI-LCMS: m/z 866 [M+H]+ ; *H-NMR (400 MHz, DMSO-î/ô): δ = 11.28 (s, 1H), 8.48-8.41 (m, 1H), 8.00-7.99 (m, 2H),7.63-7.11 (m,
13H), 6.93-6.89 (m, 4H), 5.92(m, 1H), 4.55-4.44 (m, 1H), 4.17 (m, 1H), 3.95 (m, 1H), 3.80-3.62 (m, 7H), 3.57-3.46 (m, 5H), 3.32 (s, 1H), 2.78 (m, 1H), 2.62-2.59 (m, 1H), 1.19-0.94 (m, 12H); 31P-NMR (162 MHz, DMSO-J6): δ= 149.52, 148.82.
Example 27. Synthesis of Monomer
I) TPSCl; TEA DMAP; ACN 2) NH4OH
TB AF THF
DMTrO
NHBz
CEP[N(iPr)2]2; DCI DCM
HO' ’bCD3
Example 27 monomer
Scheme-7
Préparation of (5): To the solution of 4 (18.8 g, Scheme 5) in dry ACN (200 mL) was added TPSCl (16.8 g, 65.2 mmol) and TEA (5.6 g, 65.2 mmol) and DMAP (6.8 g, 65.2 mmol), and the reaction mixture was stirred at room température for 3.5 hrs under N2 atmosphère. After 182 addition of water, the resulting mixture was extracted with EA (300 mL). The combined organic layer was washed with water and brine, dried over Na2SÜ4, and concentrated to give the crude 5 (22.0 g) as a white solid which was used directly for next step. ESI-LCMS: m/z 677 [M-H]+.
Préparation of (6): To a solution of 5 (22.0 g) in pyridine (150 mL) was added BzCl (6.8 5 g, 48.9 mmol) under ice bath. The reaction mixture was stirred at r.t. for 2.5 hrs. LCMS showed was consumed. The mixture was diluted with EA and water was added. The product was extracted with EA. The crude was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, Cl 8 silica gel; mobile phase, CH3CN/H2O (0.5% NH4HCO3) = 1/1 increasing to CH3CN/H2O (0.5% NFLHCCh) = 1/0 within 25 min, the eluted product was •10 collected at CH3CN/H2O (0.5% NEUHCCh) = 1/0; Detector, UV 254 nm. This resulted in to give the crude 6 (20.8 g, 26.7 mmol, 82% yield over two steps) as a white solid. ESI-LCMS: m/z 781 [M+H]+; ‘H-NMR (400 MHz, DMSO-î/6): δ 11.30 (s, 1H), 8.55 (d, J= 8.0 Hz, 1H), 8.00-7.98 (m, 2H), 7.74-7.66(m, 1H), 7.60-7.50(m, 2H), 7.47-7.31(m, 4H), 7.30-7.2(m, 5H), 7.20-7.1(m, 1H), 6.91 (d, J= 7.4 Hz, 4H), 5.91-5.86 (AB, J= 20.0 Hz, 1H), 4.30 (d, J= 8.0 Hz, 1H), 3.8715 3.78(s, 1H), 3.78-3.70 (m, 6H), 3.62-3.51 (m, 1H), 3.28-3.2 (m, 1H), 2.15-2.05 (m, 3H), 0.73 (s,
9H), 0.00 (m, 6H).
Préparation of (7): To a solution of 6 (20.8 g, 26.7 mmol) in THF (210 mL) was added 1 M TBAF solution (32 mL). The reaction mixture was stirred at r.t. for 1.5 hrs. LCMS showed 6 was consumed completely. Water (600 mL) was added. The product was extracted with EA (400 20 mL) and the organic layer was washed with brine and dried over Na2SO4. Then the organic layer was concentrated to give a residue which was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, Cl 8 silica gel; mobile phase, CH3CN/H2O (0.5% NH4HCO3) = 2/3 increasing to CH3CN/H2O (0.5% NH4HCO3) = 3/2 within 20 min, the eluted product was collected at CH3CN/ H2O (0.5% NH4HCO3) = 1/1; Detector, UV 254 nm. This resulted in to give 25 7 (12.4 g, 18.6 mmol, 70%) as a white solid. ESI-LCMS: m/z 667 [M+H]+; ‘H-NMR (400 MHz,
DMSO-îZê): δ 11.03 (m, 1H), 8.51-8.48 (m, 1H), 8.08-7.95 (m, 2H), 7.63-7.54(m, 1H), 7.52-7.19 (m, 9H), 7.16-7.07(m,lH), 6.94-6.89 (m, 3H), 5.95-5.87 (m, 1H), 5.31-5.17 (m, 1H), 4.61-4.37 (m, 1H), 4.20-4.07 (m, 1H), 3.82-3.47 (m, 7H), 2.57-2.42 (m, 2H).
Préparation of Example 27 monomer: To a suspension of 7 (12.4 g, 18.6 mmol) in DCM 30 (120 mL) was added DCI (1.7 g, 15.8 mmol) and CEP[N(iPr)2]2 (7.3 g, 24.2 mmol). The mixture was stirred at r.t. for 2 hrs. LC-MS showed 7 was consumed completely. The solution was washed with water twice and washed with brine and dried over Na2SC>4. Then concentrated to give a residue which was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, Cl8 silica gel; mobile phase, CH3CN/H2O (0.5% NH4HCO3) = 1/1
183 increasing to CH3CN/H2O (0.5% NH4HCO3) = 1/0 within 20 min, the eluted product was collected at CH3CN/ H2O (0.5% NH4HCO3) = 1/0; Detector, UV 254 nm. This resulted in to give Example 27 monomer (13.6 g, 15.7 mmol, 84.0%) as a white solid. ESI-LCMS: m/z 867 [M+H]+; ‘H-NMR (400 MHz, DMSO-t/6): δ 11.03 (m, 1H), 8.51-8.48 (m, 1H), 8.08-7.95 (m, 2H), 7.63-7.54(m, 1H), 7.52-7.19 (m, 9H), 7.16-7.07(m,lH), 6.94-6.89 (m, 3H), 5.95-5.87 (m, 1H), 5.31-5.17 (m, 1H), 4.61-4.37 (m, 1H), 4.20-4.07 (m, 1H), 3.82-3.47 (m, 10H), 2.74-2.59 (m, 1H), 2.57-2.43 (m, 1H), 1.27-1.10 (m, 9H), 1.09-0.95 (m, 3H). 31P-NMR (162 MHz, DMSOd6): δ 149.59, 148.85.
Example 28. Synthesis of Monomer
Example 28 monomer
Scheme-8
Préparation of (4): To a solution of 3 (13.1 g, 35.2 mmol, Scheme 3) in pyridine (130 mL) was added MsCl (4.8 g, 42.2 mmol) under -10~0°C. The reaction mixture was stirred at r.t. for 2.5 h under N2 atmosphère. TLC (DCM/MeOH =15:1) showed the reaction was consumed. The mixture was diluted with EA and water was added. The product was extracted with EA. The organic layer was washed with brine and dried over Na2SO4 and concentrated to give the crude. This resulted in to give the product 4 (14.2 g) which was used directly for the next step. ESI-LCMS: m/z 451 [M+H]+ ; ‘H-NMR (400 MHz, DMSO-î76) δ 11.43(m, 1H), 7.677.65(m, 1H), 5.90-5.80(m, 1H), 5.75-5.64(m, 1H), 4.52-4.21(m, 3H), 4.12-3.90(m, 2H), 3.483.21(m, 6H), 0.95-0.78(s, 9H), 0.13-0.03(s, 6H).
Préparation of (5): To a solution of 4 (14.2 g) in DMSO (200 mL) was added DMTrSH (19.6 g, 63.2 mmol) and tetramethylguanidine (5.1 g, 47.4 mmol) at r.t. The reaction mixture was stirred at r.t. for 3.5 h under N2 atmosphère. LCMS showed 4 the reaction was consumed. The mixture was diluted with EA and water was added. The product was extracted with EA. The
184 organic layer was washed with brine and dried over Na2SÛ4 and concentrated to give the crude. The crude was purified by silica gel column (S1O2, PE/EA = 10:1 — 1:1) to give 5 (14.2 g, 20.6 mmol, 58.5% yield over two steps) as a white solid. ESI-LCMS: m/z 689 [M+H]'; *H-NMR (400 MHz, DMSO-îZô) δ U.39(m, 1H), 7.63-7.61(d, J= 8.0 Hz, 1H), 7.45-7.1(m, 9H), 6.91-6.81(m, 4H), 5.80-5.70(m, 2H), 4.01-3.91(m, 1H), 3.85-3.78(m, 1H), 3.78-3.65(m, 6H), 3.60-3.51(m, 1H), 3.43-3.2(m, 3H), 2.50-2.32(m, 2H), 0.95-0.77(s, 9H), -0.00-0.02(s, 6H).
Préparation of (6): To a solution of 5 (14.2 g, 20.6 mmol) in THF (140 mL) was added 1 M TBAF solution (20 mL). The reaction mixture was stirred at r.t. under N2 atmosphère for 2.5 h. LCMS showed 5 was consumed completely. Water was added. The product was extracted with EA and the organic layer was washed with brine and dried over Na2SO4. Then the organic layer was concentrated to give a residue which was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, Cl 8 silica gel; mobile phase, CH3CN/H2O (0.5% NH4HCO3) = 2/3 increasing to CH3CN/H2O (0.5% NH4HCO3) = 3/2 within 20 min, the eluted product was collected at CH3CN/ H2O (0.5% NH4HCO3) = 1/1; Detector, UV 254 nm. This resulted in to give 6 (10.5 g, 18.2 mmol, 88.5%) as a white solid. ESI-LCMS: m/z 576 [M+H]+ ; Ή-NMR (400 MHz, DMSO-76) δ 11.38(m, 1H), 7.56-7.54(d, J= 8.0 Hz, 1H), 7.45-7.1(m, 9H), 6.91-6.81(m, 4H), 5.80-5.70(m, 2H), 4.05-4.00(m, 1H), 3.81-3.79(m, 1H), 3.74(m, 2H), 3.783.65(m, 6H), 3.60-3.51(m, 1H), 3.43-3.2(m, 3H), 2.40-2.32(m, 1H).
Préparation of Example 28 monomer: To a suspension of 9 (10.5 g, 18.2 mmol) in DCM (100 mL) was added DCI (1.7 g, 15.5 mmol) and CEP[N(iPr)2]2 (7.2 g, 23.7 mmol). The mixture was stirred at r.t. for 1 h. LC-MS showed 9 was consumed completely. The solution was washed with water twice and washed with brine and dried over Na2SO4. Then concentrated to give a residue which was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, Cl8 silica gel; mobile phase, CH3CN/H2O (0.5% NH4HCO3) = 1/1 increasing to CH3CN/H2O (0.5% NH4HCO3) = 1/0 within 20 min, the eluted product was collected at CH3CN/ H2O (0.5% NH4HCO3) = 1/0; Detector, UV 254 nm. This resulted in to give Example 28 monomer (12.5 g, 16.1 mmol, 88%) as a white solid. ESI-LCMS: m/z 776 [M+H]+; *H-NMR (400 MHz, DMSO-îZô) δ 11.41(m, 1H), 7.64-7.59(m, 1H), 7.40-7.25(m, 4H), 7.25-7.10(m, 5H), 6.89-6.86(m, 4H), 5.72-5.67(m, 2H), 4.02-4.00(m, 2H), 3.76-3.74(m, 8H), 3.74-3.73(m, 3H), 3.51-3.49(d, 7=8 Hz, 1H), 3.33-3.29(m, 1H), 2.77-2.73(m, 1H) , 2.63-2.60 (m, 1H), 2.502.47(m, 1H), 1.12-0.99(m, 12H). 31P-NMR (162 MHz, DMSO-76): δ 148.92, 148.84.
Example 29. Synthesis of Monomer .
185
TBSÔ F tpsci/nh4oh
Example 29 monomer
Scheme-9
Préparation of (7): To a solution of 6 (16 g, 24.1 mmol, Scheme 4) in ACN (160 mL) was added DMAP (5.9 g, 48.2 mmol) and TEA (4.8 g, 48.2 mmol), then added TPSC1 (10.9 g, 36.1 mmol) at 0 °C under N2 atmosphère and the mixture was stirred at r.t. for 5 hrs under N2 atmosphère. Then con. NH3.H2O (30 mL) was added at r.t. and the mixture was stirred at r.t. for 16 h. The reaction was quenched with water and the product was extracted with EA (200 mL). The organic phase was concentrated to give the crude 7 (16.0 g) as a white solid which was used directly for next step.
Préparation of (8): To a stirred solution of 7 (16.0 g, 24.1 mmol) in pyridine (160 mL) were added BzCl (4.1 g, 28.9 mmol) 0°C under N2 atmosphère. And the reaction mixture was stirred at r.t. for 2.5 h. With ice-bath cooling, the reaction was quenched with water and the product was extracted with EA (200 mL). The organic phase was evaporated to dryness under reduced pressure to give a residue which was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, Cl 8 silica gel; mobile phase, CFhCN/FhO (0.5% NH4HCO3) = 1/1 increasing to CFLCN/FLO (0.5% NH4HCO3) = 1/0 within 20 min, the eluted product was collected at CH3CN/ H2O (0.5% NH4HCO3) = 1/0; Detector, UV 254 nm. This resulted in to give 8 (18.0 g, 23.4 mmol, 97.0%) as a white solid. ESI-LCMS: m/z 768 [M+H]+ ; *H-NMR (400 MHz, DMSO-Jô): δ 11.31 (s, 1H), 8.47(d, J= 7.2 Hz, 1H), 7.99 (d, J= 7.6 Hz, 2H), 7.65-7.16 (m, 13H), 6.92 (d, J= 8.8 Hz, 4H), 6.01 (d, J= 18.4 Hz, 1H), 5.18-5.04 (dd, 1H), 4.58-4.52 (m, 1H), 4.07 (d, J= 9.6 Hz, 1H), 3.75 (s, 6H), 0.73 (s, 9H), 0.05 (s, 3H), -0.06 (s, 3H).
* Préparation of (9): To a solution of 8 (18.0 g, 23.4 mmol) in THF (180 mL) was added 1 M TB AF solution (23 mL). The reaction mixture was stirred at r.t. for 1.5 h. LC-MS showed 8 was consumed completely. Water (500 mL) was added. The product was extracted with EA (300
186 mL) and the organic layer was washed with brine and dried over NaiSCU. Then the organic layer was concentrated to give a residue which was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, Cl8 silica gel; mobile phase, CH3CN/H2O (0.5% NH4HCO3) = 2/3 increasing to CH3CN/H2O (0.5% NH4HCO3) = 3/2 within 20 min, the eluted product was collected at CH3CN/ H2O (0.5% NH4HCO3) = 1/1 ; Detector, UV 254 nm. This resulted in to give 7 (13.7 g, 21.1 mmol, 90.5%) as a white solid. ESI-LCMS: m/z 654.2 [M+H]+; ‘H-NMR (400 MHz, DMSO-ί/ό): δ 11.31 (s, 1H), 8.35(d, J= 7.4 Hz, 1H), 8.01 (m, 2H), 7.65-7.16 (m, 13H), 6.92 (d, J= 8.8 Hz, 4H), 5.94 (d, J= 18.0 Hz, 1H), 5.71 (d, J= 7.0 Hz, 1H), 5.12-4.98 (dd, 1H), 4.51-4.36 (m, 1H), 4.09 (d, J= 9.6 Hz, 1H), 3.75 (s, 6H).
Préparation of Example 29 monomer: To a suspension of 9 (10.6 g, 16.2 mmol) in DCM (100 mL) was added DCI (1.6 g, 13.7 mmol) and CEP[N(iPr)2]2 (5.8 g, 19.4 mmol). The mixture was stirred at r.t. for 1 h. LC-MS showed 9 was consumed completely. The solution was washed with water twice and washed with brine and dried over Na2SO4. Then concentrated to give a residue which was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1):
Column, Cl 8 silica gel; mobile phase, CH3CN/H2O (0.5% NH4HCO3) = 1/1 increasing to CH3CN/H2O (0.5% NH4HCO3) = 1/0 within 20 min, the eluted product was collected at CH3CN/ H2O (0.5% NH4HCO3) = 1/0; Detector, UV 254 nm. This resulted in to give Example 29 monomer (10.5 g, 14.5 mmol, 75.9%) as a white solid. ESI-LCMS: m/z 854.3 [M+H]+; ‘H-NMR (400 MHz, DMSO-î76): δ 11.31 (s, 1H), 8.41-8.37(m, 1H), 8.01 (d, J= 7.7 Hz, 2H), 7.65-7.16 (m, 13H), 6.92-6.88 (m, 4H), 6.06-5.98 (m, 1H), 5.33-5.15 (m, 1H), 4.78-4.58 (m, 1H), 4.234.19 (m, 1H), 3.81-3.73 (m, 6H), 3.60-3.50 (m, 3H), 3.32 (s, 1H), 2.76 (t, J= 6.0 Hz, 1H), 2.60 (t, .7=5.8 Hz, 1H), 1.15-0.94 (m, 12H) ; 31P-NMR (162 MHz, DMSO-îZ6): δ 150.23, 150.18, 149.43, 149.38.
Example 30. Synthesis of Monomer
187
TBAF THF
CEP[N(iPr),],; DCI DCM
HO' t)CD3
Example 30 tnonomer
Scheme-10
Préparation of (9): To a solution of 8 (18.8 g, 26.4 mmol, Scheme 5 ) in ACN (200 mL) was added TPSC1 (16.8 g, 55.3 mmol) and DMAP (5.6 g, 55.3 mmol) and TEA (6.8 g, 55.3 mmol). The reaction mixture was stirred at r.t. for 3.5 hrs. LCMS showed the reaction was consumed. The mixture was diluted with con. NH4OH (28 mL). The mixture was diluted with water and EA. The product was extracted with EA. The organic layer was washed with brine and dried over NajSCU and concentrated to give the crude 9 (18.5 g) wihch was used directly for the next step.
Préparation of (10): To a solution of 9 (18.8 g, 27.69 mmol) in pyridine (200 mL) was added BzCl (5.8 g, 41.5 mmol) under ice bath. The reaction mixture was stirred at r.t. for 2.5 hrs. LCMS showed 9 was consumed. The mixture was diluted with EA and water was added. The product was extracted with EA. The crude was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, Cl8 silica gel; mobile phase, CH3CN/H2O (0.5% NH4HCO3) = 1/1 increasing to CH3CN/H2O (0.5% NH4HCO3) = 1/0 within 25 min, the eluted product was collected at CH3CN/H2O (0.5% NH4HCO3) = 1/0; Detector, UV 254 nm. This resulted in to give 10 (19.8 g, 25.3 mmol, 91% yield) as a white solid. ESI-LCMS: m/z 783 [ΜΗ]’; Ή-NMR (400 MHz, DMSO-ri6): δ 11.29 (d, J= 2.0 Hz, 1H), 8.42 (d, J= 8.0 Hz, 1H), 8.028.00(m,2H), 7.64-7.62(m,lH), 7.60-7.41 (m,2H),7.47.41-7.19 (m, 9H), 6.94-6.85 (m, 4H), 5.81 (d, 4.0 Hz, 1H), 5.33-5.26 (m, 1H), 5.21 (d, J= 7.2 Hz, 1H), 4.06-3.90 (m, 2H), 3.83-3.77 (m, 1H), 3.74 (s, 6H).
Préparation of (11): To a solution of 10 (18.8 g, 26.4 mmol) in THF (190 mL) was added 1 M TBAF solution (28 mL). The reaction mixture was stirred at r.t. for 1.5 hrs. LCMS showed 10 was consumed completely. Water (200 mL) was added. The product was extracted with EA
188 (200 mL) and the organic layer was washed with brine and dried over Na2SO4. Then the organic layer was concentrated to give a residue which was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, Cl 8 silica gel; mobile phase, CH3CN/H2O (0.5% NH4HCO3) = 2/3 increasing to CH3CN/H2O (0.5% NH4HCO3) = 3/2 within 20 min, the eluted product was collected at CH3CN/ H2O (0.5% NH4HCO3) = 1/1; Detector, UV 254 nm. This resulted in to give 11 (17.1 g, 25.6 mmol, 96%) as a white solid. ESI-LCMS: m/z 669 [M-H]'; ‘H-NMR (400 MHz, DMSO-î/6): δ 11.29 (d, J= 2.0 Hz, 1H), 8.42 (d, J= 8.0 Hz, 1H), 8.028.00(m,2H), 7.64-7.62(m,lH), 7.60-7.41 (m,2H),7.47.41-7.19 (m, 9H), 6.94-6.85 (m, 4H), 5.81 (d, J= 4.0 Hz, 1H), 5.33-5.26 (m, 1H), 5.21 (d, J= 7.2 Hz, 1H), 4.06-3.90 (m, 2H), 3.83-3.77 (m, 1H), 3.74 (s, 6H).
Préparation of Example 30 monomer: To a suspension of 11 (10.8 g, 16.2 mmol) in DCM (100 mL) was added DCI (1.5 g, 13.7 mmol) and CEP[N(iPr)2]2 (5.8 g, 19.3 mmol). The mixture was stirred at r.t. for 2 hrs. LC-MS showed 11 was consumed completely. The solution was washed with water twice and washed with brine and dried over Na2SC>4. Then concentrated to give a residue which was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, Cl 8 silica gel; mobile phase, CH3CN/H2O (0.5% NH4HCO3) = 1/1 increasing to CH3CN/H2O (0.5% NH4HCO3) = 1/0 within 20 min, the eluted product was collected at CH3CN/ H2O (0.5% NH4HCO3) = 1/0; Detector, UV 254 nm. This resulted in to give Example 30 monomer (11.3 g, 13 mmol, 80%) as a white solid. ESI-LCMS: m/z 868 [M+H]+; ‘H-NMR (400 MHz, DMSO-J6): δ 11.03 (m, 1H), 8.51-8.48 (m, 1H), 8.08-7.95 (m, 2H), 7.637.54(m, 1H), 7.52-7.19 (m, 9H), 7.16-7.07(m,lH), 6.94-6.89 (m, 3H), 5.95-5.87 (m, 1H), 5.315.17 (m, 1H), 4.61-4.37 (m, 1H), 4.20-4.07 (m, 1H), 3.82-3.47 (m, 10H), 2.74-2.59 (m, 1H), 2.57-2.43 (m, 1H), 1.27-1.10 (m, 9H), 1.09-0.95 (m, 3H). 31P-NMR (162 MHz, DMSO-J6): δ 149.52, 148.81.
Example 31. Synthesis of Monomer
189
CEP[N(iPr)2],; DCI DCM
Scheme-11
Préparation of (2): To a stirred solution of 1 (100.0 g, 406.5 mmol) in pyridine (1000 mL) were added DMTrCl (151.2 g, 447.Immol) at r.t. And the reaction mixture was stirred at r.t. for 2.5 hrs. With ice-bath cooling, the reaction was quenched with water and the product was extracted with EA (3000 mL). The organic phase was evaporated to dryness under reduced pressure to give a residue which was purified by silica gel column chromatography (S1O2, dichloromethane: methanol = 100:1) to give 2 (210.0 g, 90%) as a white solid. ESI-LCMS: m/z 548.2 [M+H]+; 'H-NMR (400 MHz, DMSO-76): δ 11.43 (d, J= 1.8 Hz, 1H), 7.77 (d, 7 = 8.0 Hz, 1H), 7.40-7.21(m, 9H), 6.92-6.88(m, 4H), 5.89 (d, 7= 20.0 Hz, 1H), 5.31-5.29 (m, 1H), 5.195.04 (dd, 1H), 4.38-4.31 (m, 1H), 4.02-3.98 (m, 1H), 3.74(s, 6H), 3.30 (d, J= 3.2 Hz, 2H); 19FNMR (376 MHz, DMSO-76): δ -199.51.
Préparation of (3): To a stirred solution of 2 (100.0 g, 182.8 mmol) in pyridine (1000 mL) were added MsCl (31.2 g, 274.2 mmol) at 0°C under N2 atmosphère. And the reaction mixture was stirred at r.t for 2.5 h. With ice-bath cooling, the reaction was quenched with water and the product was extracted with EA (200 mL). The organic phase was evaporated to dryness under reduced pressure to give the crude (114.0 g) as a white solid which was used directly for next step. To the solution of the crude (114.0 g, 187.8 mmol) in DMF (2000 mL) was added K2CO3
190 (71.5 g, 548.4 mmol), and the reaction mixture was stirred at 90 °C for 15 h under N2 atmosphère. After addition of water, the resulting mixture was extracted with EA (500 mL). The combined organic layer was washed with water and brine, dried over Na2SO4, and concentrated to give a residue which was purified by silica gel column chromatography (SiO2, dichloromethane: methanol = 30:1) to give 3 (100.0 g, 90%) as a white solid. ESI-LCMS: m/z 531.2 [M+H]+; ‘H-NMR (400 MHz, DMSO-cZ6): δ 7.79 (d, J= 8.0 Hz, 1H), 7.40-7.21(m, 9H), 6.89-6.83(m, 4H), 6.14 (d, J= 5.4 Hz, 1H), 6.02-5.90 (dd, 1H), 5.87 (d, J= 20.0 Hz, 1H), 5.45 (m, 1H), 4.61 (m, 1H), 3.73(d, J= 1.9 Hz, 6H), 3.30-3.15 (m, 2H), 1.24-1.16 (m, 1H); 19F-NMR (376 MHz, DMSO-î/ô): δ -204.23.
ίο Préparation of (4): A solution of 3 (100 g, 187.8 mmol) in THF (1000 mL) was added 6N
NaOH (34 mL, 206.5 mmol). The mixture was stirred at r.t. for 6 h. After completion of reaction, the resulting mixture was added H2O, and then the mixture was extracted with EA, the organic layer was washed with brine, dried over sodium sulfate and removed to give the residue was purified by silica gel column chromatography (SiO2, dichloromethane: methanol = 30:1) to give
4 (90.4 g, 90%) as a white solid. ESI-LCMS: m/z 548.2 [M+H]+; 19F-NMR (376 MHz, DMSOde): δ -184.58.
Préparation of (5): To a stirred solution of 4 (90.4 g, 165.2 mmol) in pyridine (1000 mL) were added MsCl (61.5 g, 495.6 mmol) at 0°C under N2 atmosphère. And the reaction mixture was stirred at r.t for 16 hrs. With ice-bath cooling, the reaction was quenched with water and the 20 product was extracted with EA. the organic layer was washed with brine, dried over sodium sulfate and removed to give the residue was purified by silica gel column chromatography (SiO2, PE: EA = 1:1) to give 5 (75.0 g, 90%) as a white solid. ESI-LCMS: m/z 626.2 [M+H]+ ; ‘H-NMR (400 MHz, DMSO-de): δ 11.51 (d, 7= 1.6 Hz, 1H), 7.43-7.23(m, 10H), 6.92-6.88(m, 4H), 6.08 (d, J= 20.0 Hz, 1H), 5.55-5.39 (m, 2H), 4.59 (m, 1H), 3.74(s, 6H), 3.48-3.28 (m, 2H), 3.17 (s,
3H); I9F-NMR (376 MHz, DMSO-76): δ -187.72.
Préparation of (6): To the solution of 5 (75.0 g, 120.4 mmol) in DMF (1500 mL) was added KSAc (71.5 g, 548.4 mmol) at 110 °C under N2 atmosphère, After the reaction mixture was stirred at 110 °C for 3 h were added KSAc (7L5 g, 548.4 mmol) under N2 atmosphère. And the reaction mixture was stirred at r.t for 16 h. After addition of water, the resulting mixture was 30 extracted with EA. The combined organic layer was washed with water and brine, dried over Na2SC>4, and concentrated to give a residue which was purified by silica gel column chromatography (SiO2, PE: EA = 1:1) to give 6 (29.0 g, 90%) as a white solid. ESI-LCMS: m/z 605.2 [M+H]+ ; ‘H-NMR (400 MHz, DMSO-76): δ 11.45 (d, J= 1.9 Hz, 1H), 7.95(d, J= 8.0 Hz, 1H), 7.38-7.21 (m, 9H), 6.92-6.87 (m, 4H), 5.93 (m, 1H), 5.50-5.36 (dd, 1H), 5.25-5.23 (dd, 1H),
191
4.54-4.42 (m, 1H), 4.17-4.12 (m, 1H), 3.74 (m, 7H), 3.35-3.22 (m, 2H), 2.39 (s,lH); I9F-NMR (376 MHz, DMSO-Jô): δ -181.97.
Préparation of (7): A solution of 6 (22 g, 36.3 mmol) in a mixture solvent of THF /MeOH (1:1, 200 mL) was added IN NaOMe (70 mL, 72.6 mmol)was stirred at 20 °C for 4 h. After completion of reaction, the resulting mixture was added H2O, and then the mixture was extracted with EA, the organic layer was washed with brine, dried over sodium sulfate and removed to give the residue was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, Cl 8 silica gel; mobile phase, CH3CNÆUO (0.5% NH4HCO3) = 2/3 increasing to CH3CN/H2O (0.5% NH4HCO3) = 3/2 within 25 min, the eluted product was collected at CH3CN/ H2O (0.5% NH4HCO3) =4/3; Detector, UV 254 nm. This resulted in to give 7 (10.5 g, 14.5 mmol, 75.9%) as a white solid. ESI-LCMS: m/z 565.1 [M+H]+ ; *H-NMR (400 MHz, DMSO-cZô): δ 11.45 (s, 1H), 7.83(d, J= 8.0 Hz, 1H), 7.40-7.23 (m, 9H), 6.90 (d, J= 8.8 Hz, 4H), 5.88 (m, 1H), 5.29-5.15 (m, 2H), 3.72 (m, 7H), 3.43 (m, 2H), 2.78 (d, J= 10.6 Hz, 1H).
Préparation of Example 31 monomer: To a suspension of 7 (10.5 g, 18.6 mmol) in DCM (100 mL) was added DCI (1.8 g, 15.7 mmol) and CEP[N(iPr)2]2 (6.7 g, 22.3 mmol). The mixture was stirred at r.t. for 1 h. LC-MS showed 8 was consumed completely. The solution was washed with water twice and washed with brine and dried over Na2SO4. Then concentrated to give a residue which was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, Cl8 silica gel; mobile phase, CH3CN/H2O (0.5% NH4HCO3) = 1/1 increasing to CH3CN/H2O (0.5% NH4HCO3) = 1/0 within 20 min, the eluted product was collected at CH3CN/ H2O (0.5% NH4HCO3) = 1/0; Detector, UV 254 nm. This resulted in to give Example 31 monomer (10.5 g, 14.5 mmol, 75.9%) as a white solid. ESI-LCMS: m/z 765.3 [M+H]+; 'H-NMR (400 MHz, DMSO-t/ô): δ 11.40 (d, J= 12.2 Hz, 1H), 7.90-7.86(m, 1H), 7.41-7.24 (m, 9H), 6.916.89 (m, 4H), 5.97 (m, 1H), 5.33-5.10 (m, 2H), 4.18-4.16 (m, 1H), 3.91-3.39 (m, 17H), 2.81 (t, J = 5.6 Hz, 1H), 2.66 (t, J= 6.0 Hz, 1H), 1.33-0.97 (m, 12H) ; 31P-NMR (162 MHz, DMSO-76): δ 164.57, 160.13.
192
Example 32. Synthesis of Monomer
DMTrCl Pyridine
IJMsCl Pyridine
2) K2CO3 DMF
Example 32 monomer
Scheme-12
Préparation of (2): To a stirred solution of 1 (100.0 g, 387.5 mmol) in pyridine (1000 mL) was added DMTrCl (151.2 g, 447. Immol) at r.t. And the reaction mixture was stirred at r.t. for 2.5 hrs. With ice-bath cooling, the reaction was quenched with water and the product was extracted with EA (3000 mL). The organic phase was evaporated to dryness under reduced pressure to give a residue which was purified by silica gel column chromatography (SiO2, dichloromethane: methanol = 100:1) to give 2 (200.0 g, 90%) as a white solid. ESI-LCMS: m/z
561 [M+H]+.
Préparation of (3): To a stirred solution of 2 (73.0 g, 130.3 mmol) in pyridine (730 mL) were added MsCl (19.5 g, 169.2 mmol) at 0°C under N2 atmosphère. And the reaction mixture was stirred at r.t for 2.5 h. With ice-bath cooling, the reaction was quenched with water and the product was extracted with EA (200 mL). The organic phase was evaporated to dryness under reduced pressure to give the crude (80.0 g) as a white solid which was used directly for next step. To the solution of the crude (80.0 g, 130.3 mmol) in DMF (1600 mL) was added K2CCh (71.5 g, 390.9 mmol), and the reaction mixture was stirred at 90 °C for 15 h under N2 atmosphère. Aller addition of water, the resulting mixture was extracted with EA (500 mL). The combined organic layer was washed with water and brine, dried over Na2SÜ4, and concentrated to give a residue
193 which was purified by silica gel column chromatography (S1O2, dichloromethane: methanol = 30:1) to give 3 (55.0 g, 90%) as a white solid. ESI-LCMS: m/z 543. [M+H]+; ’H-NMR (400 MHz, DMSO-î/ô): δ 7.68 (d, J=8.0 Hz, 1H), 7.40-7.21(m, 9H), 6.89-6.83(m, 4H), 5.96(s, 1H), 5.83 (d, J= 5.4 Hz, 1H), 5.26 (s, 1H), 4.59 (s, 1H), 4.46 (t, J= 6.0 Hz, 1H), 3.72(s, 6H), 3.44(s, 3H), 3.18-3.12 (m, 2H).
Préparation of (4): A solution of 3 (55 g, 101.8 mmol) in THF (550 mL) was added 6N NaOH (34 mL, 206.5 mmol). The mixture was stirred at 20 °C for 6 hrs. After completion of reaction, the resulting mixture was added H2O, and then the mixture was extracted with EA, the organic layer was washed with brine, dried over sodium sulfate and removed to give the residue was purified by silica gel column chromatography (S1O2, dichloromethane: methanol = 30:1) to give 4 (57.4 g, 87%) as a white solid. ESI-LCMS: m/z 561 [M+H]+.
Préparation of (5): To a stirred solution of 4 (57.4 g, 101.8 mmol) in pyridine (550 mL) were added MsCl (61.5 g, 495.6 mmol) at 0°C under N2 atmosphère. And the reaction mixture was stirred at r.t for 16 h. With ice-bath cooling, the reaction was quenched with water and the product was extracted with EA. the organic layer was washed with brine, dried over sodium sulfate and removed to give the residue was purified by silica gel column chromatography (S1O2, PE: EA = 1:1) to give 5 (57.0 g, 90%) as a white solid. ESI-LCMS: m/z 639 [M+H]+
Préparation of (6): To the solution of 5 (57.0 g, 89.2 mmol) in DMF (600 mL) was added KSAc (71.5 g, 448.4 mmol) at 110 °C under N2 atmosphère, After the reaction mixture was stirred at 110 °C for 3 h were added KSAc (71.5 g, 448.4 mmol) under N2 atmosphère. And the reaction mixture was stirred at r.t for 16 h. After addition of water, the resulting mixture was extracted with EA. The combined organic layer was washed with water and brine, dried over Na2SO4, and concentrated to give a residue which was purified by silica gel column chromatography (S1O2, PE: EA = 1:1) to give 6 (29.0 g, 47%) as a white solid. ESI-LCMS: m/z 619.2 [M+H]+; ’H-NMR (400 MHz, DMSO-î/6): δ 11.41 (s, 1H), 8.06 (s, 1H), 7.40-7.23 (m, 9H), 6.90 (d, J= 8.8 Hz, 4H), 5.82 (s, 1H), 5.10-5.08 (dd, 1H), 4.38-4.34 (m, 1H), 4.08-4.02 (m, 3H), 3.74 (s, 6H), 3.45 (s, 3H),3.25 (m, 2H), 2.37 (s, 3H); ESI-LCMS: m/z 619 [M+H]+ .
Préparation of (7): A solution of 6 (22 g, 35.3 mmol) in a mixture solvent of THF /MeOH (1:1, 200 mL) was added IN NaOMe (70 mL, 72.6 mmol)was stirred at 20 °C for 4 h. After completion of reaction, the resulting mixture was added H2O, and then the mixture was extracted with EA, the organic layer was washed with brine, dried over sodium sulfate and removed to give the residue was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, Cl8 silica gel; mobile phase, CH3CN/H2O (0.5% NH4HCO3) = 2/3 increasing to CH3CN/H2O (0.5% NH4HCO3) = 3/2 within 25 min, the eluted product was collected at CH3CN/
194
H2O (0.5% NH4HCO3) =4/3; Detector, UV 254 nm. This resulted in to give 7 (14.0 g, 70.9%) as a white solid. ESI-LCMS: m/z 576.1 [M+H]+; ‘H-NMR (400 MHz, DMSO-J6): δ 11.38 (s, 1H), 7.90(d, 7= 8.0 Hz, 1H), 7.40-7.23 (m, 9H), 6.90 (d, 7=8.8 Hz, 4H), 5.80 (s, 1H), 5.15-5.13 (dd, 1H), 3.93 (m, 1H),3.87 (d, 7= 5.0 Hz, 1H), 3.74 (s, 6H), 3.59 (m, 2H), 3.49 (s, 3H),3.39 (d, 7 = 2.2 Hz, 2H), 2.40 (d, 7= 10.2 Hz, 1H).
Préparation of Example 32 monomer: To a suspension of 7 (10.5 g, 18.6 mmol) in DCM (100 mL) was added DCI (1.8 g, 15.7 mmol) and CEP[N(iPr)2]2 (6.7 g, 22.3 mmol). The mixture was stirred at r.t. for 1 h. LC-MS showed 7 was consumed completely. The solution was washed with water twice and washed with brine and dried over Na2SO4. Then concentrated to give a residue which was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, Cl 8 silica gel; mobile phase, CH3CN/H2O (0.5% NH4HCO3) = 1/1 increasing to CH3CN/H2O (0.5% NH4HCO3) = 1/0 within 20 min, the eluted product was collected at CH3CN/ H2O (0.5% NH4HCO3) = 1/0; Detector, UV 254 nm. This resulted in to give Example 32 monomer (10.5 g, 14.5 mmol, 75.9%) as a white solid. ESI-LCMS: m/z 776.3 [M+H]+; ‘H-NMR (400 MHz, DMSO-Jô): δ 11.40 (d, 7= 12.2 Hz, 1H), 8.04-7.96(dd, 1H), 7.43-7.24 (m, 9H), 6.926.87 (m, 4H), 5.84 (m, 1H), 4.93 (m, 1H), 4.13 (m, 1H), 3.91-3.39 (m, 17H), 2.82 (t, 7= 5.6 Hz, 1H), 2.68 (t, 7= 6.0 Hz, 1H), 1.22-0.97 (m, 12H) ; 3IP-NMR (162 MHz, DMSO-76): δ 165.06, 157.59.
Example 33. Synthesis of 5’ End Cap Monomer
Imidazole DMTrO—DCM DMTrO— H° /° TBS 1 O MOPO-p' ΜΟΡΟ HCOOH/H2O TBSO 'b / 5 H0 EDCI; Pyridine Toluene \ q 6%DCA in DCM TFA; DMSO , O=\.O. ________ \/ --x ··— OPOM b b TBS° /° TBSO' O MOPO-p=O / I OPOM 9 3 4 K 2 q' OPOM % \ O Π 0 / > MOPO-p' CEP[N(iPr)2]2; DCI _ù/ /^° Κ-χΟ ΜΟΡΟ DCM______ ΙΓ° )__/ O . °\ ° HO O \ ,P-0 / /—N \ c ) CN O Example 33 monomer Scheme-13
195
Préparation of (2): To a solution of 1 (11.2 g, 24.7 mmol) in DCM (120 mL), imidazole (4.2 g, 61.9 mmol) and TBSC1 (5.6 g, 37.1 mmol) were added at r.t., mixture was stirred at r.t. for 15 hrs, LCMS showed 1 was consumed completely. Mixture was added water (500 mL) and extracted with DCM (50 mL*2). The organic phase was dried over NaiSO4 and concentrated to give 2 (16.0 g) as an oil for the next step.
Préparation of (3): To a solution of 2 (16.0 g, 28.4 mmol) was added 6% DCA in DCM (160 mL) and triethylsilane (40 mL) at r.t. The reaction mixture was stirred at r.t. for 2 hrs. TLC showed 2 was consumed completely. Water (300 mL) was added, mixture was extracted with DCM (50 mL*4), organic phase was dried by NazSCfi, concentrated by reduce pressure to give crude which was purified by column chromatography (S1O2, PE/EA = 10:1 to 1:1) to give 3 (4.9 g, 65.9% yield) as an oil. ESI-LCMS: m/z 263 [M+HjjH-NMR (400 MHz, DMSO-76) δ 4.84-4.50(m, 1H), 4.3-4.09(m, 1H), 3.90-3.80(m, 1H), 3.75-3.67(m, 1H), 3.65-3.57(m, 2H), 3.50-3.44(m, 1H), 3.37-3.28(m, 4H), 0.95-0.78(s, 9H), O.13-O.O3(s, 6H).
Préparation of (4): To a solution of 3 (3.3 g, 12.6 mmol) in DMSO (33 mL) was added EDCI (7.2 g, 37.7 mmol) .The mixture was added pyridine (1.1 g, 13.8 mmol) and TFA (788.6 mg, 6.9 mmol). The reaction mixture was stirred at r.t. for 3 hrs. TLC (PE/EA = 4:1) showed 3 was consumed. The mixture was diluted with EA and water was added. The product was extracted with EA. The organic layer was washed with brine and dried over Na2SÜ4 and concentrated to give the crude. This resulted in to give 4 (3.23 g) as an oil for the next step.
Préparation of (5): To a solution of 4 (3.3 g, 12.6 mmol) in toluene (30 mL) was added POM ester 4a ( reference for 4a Journal of Médicinal Chemistry, 2018, 61 (3), 734-744) (7.9 g, 12.6 mmol) and KOH (1.3 g, 22.6 mmol) at r.t. The reaction mixture was stirred at 40 °C for 8 hrs. LCMS showed 4 was consumed. The mixture was diluted with water and EA was added. The product was extracted with EA. The organic layer was washed with brine and dried over Na2SÛ4 and concentrated to give the crude. The crude was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, Cl 8 silica gel; mobile phase, CH3CN/H2O (0.5% NH4HCO3) = 1/1 increasing to CH3CN/H2O (0.5% NH4HCO3) = 1/0 within 20 min, the eluted product was collected at CH3CN/H2O (0.5% NH4HCO3) = 91/9 Detector, UV 254 nm. This resulted in to give 5 (5.4 g, 9.5 mmol, 75.9% yield) as an oil. ESI-LCMS: m/z 567.2 [M+H]+; ‘H-NMR (400 MHz, CDCI3) δ 6.89-6.77(m, 1H), 6.07-5.96(m, 1H), 5.86-5.55(m, 4H), 4.85 -4.73(m, 1H), 4.36-4.27(m, 1H), 4.05-3.96(m, 1H), 3.95-3.85(m, 1H), 3.73-3.65(m, 1H), 3.44-3.35 (m, 3H), 1.30-1.25(s, 18H), 0.94-0.84(s, 9H), 0.14-0.05(s, 6H).31P-NMR (162 MHz, CDC13)ô 18.30, 15.11.
196
Préparation of (6): To a solution of 5 (5.4 g, 9.5 mmol) in HCOOH (30 mL) /H2O (30 mL) = 1:1 at r.t. The reaction mixture was stirred at r.t. for 15 hrs. LCMS showed the reaction was consumed. The mixture was diluted with con. NH4OH till pH = 7.5. The product was extracted with EA. The organic layer was washed with brine and dried over Na2SÛ4 and concentrated to give the crude. The crude was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, C18 silica gel; mobile phase, CH3CN/FEO (0.5%HCOOH) = 30/70 increasing to CH3CN/H2O (0.5% HCOOH) = 70/30 within 45 min, the eluted product was collected at CH3CN/ H2O (0.5% HCOOH) = 59/41 Detector, UV 220 nm. This resulted in to give 6 (2.4 g, 5.7 mmol, 59.4% yield) as an oil. ESI-LCMS: m/z 453.2 [M+H]+; *H-NMR (400 MHz, DMSO-î/ô) δ 6.84-6.68(m, 1H), 6.07-5.90(m, 1H), 5.64- 5.55(m, 4H), 5.32-5.24(m, 1H), 4.23-4.15(m, 1H), 4.00-3.90(m, 1H), 3.89-3.80(m, 1H), 3.78-3.69(m, 2H), 3.37-3.30(s, 3H), 1.30-1.10(s, 18H). 31P-NMR (162 MHz, DMSO-d6) δ 18.14.
Préparation of Example 33 monomer: To a solution of 6 (2.1 g, 4.5 mmol) in DCM (21 mL) were added DCI (452.5 mg, 3.8 mmol) and CEP[N(iPr)2]2 (1.8 g, 5.9 mmol) at r.t. The reaction mixture was stirred at r.t. for 15 hrs under N2 atmosphère. LCMS showed 6 was consumed. The mixture was diluted with water. The product was extracted with DCM (30 mL). The organic layer was washed with brine and dried over Na2SO4 and concentrated to give the crude. The crude was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, Cl 8 silica gel; mobile phase, CH3CN/H2O (0.5% NH4HCO3) = 1/1 increasing to CH3CN/H2O (0.5% NH4HCO3) = 1/0 within 28 min, the eluted product was collected at CH3CN/ H2O (0.5% NH4HCO3) = 80/20 Detector, UV 254 nm. This resulted in to give Example 33 monomer (2.8 g, 4.3 mmol, 95.2% yield) as an oil. ESI-LCMS: m/z 653.2 [M+H]+; ’H-NMR (400 MHz, DMSO-î/ô) δ 6.89-6.77(m, 1H), 6.11-5.96(m, 1H), 5.65-5.50(m, 4H), 4.39-4.34(d, J= 20 Hz, 1H), 4.18-3.95(m, 2H), 3.94-3.48(s, 6H), 3.40-3.28(m, 4H), 2.84-2.75 (m, 2H), 1.261.98(s, 30H). 3IP-NMR (162 MHz, DMSO-J6) δ 149.018, 148.736, 17.775, 17.508.
Example 34. Synthesis of 5’ End Cap Monomer
197
OPOM tbso' b
MOPO-p=o 1 f ,ΟΡΟΜ q'P'OPOM 5
CEP, DCI.DCM
Example 34 monomer
Scheme-14
Préparation of (2): To a solution of 1 (ref for 1 Tetrahedron , 2013, 69, 600-606) (10.60 g, 47.32 mmol) in DMF (106 mL), imidazole (11.26 g, 165.59 mmol) and TBSC1 (19.88 g, 132.53 mmol) were added. The mixture was stirred at r.t. for 3.5 hrs, LCMS showed 1 was consumed completely. Water was added and extracted with EA, dried over by Na^SCL. The filtrate was evaporated under reduced pressure to give 2 (20.80 g, 45.94 mmol, 97.19% yield) for the next step.
Préparation of (3): To a solution of 2 (20.80 g, 45.94mmol) in THF (248 mL), was added TFA (124 mL) and H2O (124 mL) at 0°C, reaction mixture was stirred for 30 min. LCMS showed 2 was consumed completely. Then was extracted with EA, washed with sat. NaCl (aq.), dried over by Na2SO4. The filtrate was evaporated under reduced pressure to give the crude product which was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, Cl8 silica gel; mobile phase, CH3CN/H2O (0.5% NH4HCO3) = 1/1 increasing to CH3CN/H2O (0.5% NH4HCO3) = 1/0 within 20 min, the eluted product was collected at CH3CN/ H2O (0.5% NH4HCO3) = 1/0; Detector, UV 254 nm. This resulted in to give 3 (10.00 g, 29.59 mmol, 64.31% yield). ‘H-NMR (400 MHz, DMSO-J6): δ 7.33-7.18(m, 5H), 4.83-4.80(m, 1H), 4.61-4.59(m, 1H), 4.21-4.19(m, 1H), 3.75-3.74(m, 1H), 3.23(m, 3H), 3.13(m, 3H),2.41-2.40(m, 1H), 0.81(m, 9H), 0.00(m, 6H).
Préparation of (4): To a solution of 3(3.70 g, 10.95 mmol) in DMSO (37 mL) was added EDCI (6.30 g, 32.84 mmol). Then pyridine (0.95 g, 12.05 mmol) and TFA (0.69 g, 6.02 mmol) was added in N2 atmosphère. The mixture was stirred for 3 hrs at r.t. LCMS showed 3 was consumed completely. Water was poured into and extracted with EA, washed with sat. NaCl (aq.), dried over by Na2SC>4. The filtrate was evaporated under reduced pressure to give the crude product which was directly used for next step.
Préparation of (5): To a solution of 4 in toluene (100.00 mL), was added 4a (6.93 g, 10.97 mmol) and KOH (1.11 g, 19.78 mmol). It was stirred for 3.5 hrs at 40°C in N2 atmosphère.
198
TLC and LCMS showed 4 was consumed completely. Then was extracted with EA, washed with water and sat. NaCl (aq.), dried over by Na2SÜ4. The fdtrate was evaporated under reduced pressure to give the crude product which was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, Cl8 silica gel; mobile phase, CH3CN/H2O (0.5% NH4HCO3) = 1/1 increasing to CH3CN/H2O (0.5% NH4HCO3) = 1/0 within 20 min, the eluted product was collected at CH3CN/ H2O (0.5% NH4HCO3) = 1/0; Detector, UV 254 nm. This resulted in to give 5 (4.30 g, 6.70 mmol, 61.17% yield). ‘H-NMR (400 MHz, CDCI3): δ 7.27-7,26(m, 4H), 7.17(m, 1H), 6.94-6.82(m, 1H), 6.13-6.02(m, 1H), 5.63-5.56(m, 4H), 4.90-4.89(m, 1H), 4,45-4.41(m, 1H), 3.98-3.95(m, 1H), 3.39-3.29(m, 4H), 1.90(m, 1H), 1.12-0.83(m, 29H), 0.00(m, 7H); 31PNMR (162 MHz, CDCI3): δ 18.021, 14.472.
Préparation of (6): To a solution of 5 (4.30 g, 6.70 mmol) in THF (43.00 mL) was added HCOOH (100 mL) and H2O (100 mL). It was stirred ovemight at r.t. LCMS showed 5 was consumed completely. NH4OH was poured into it and was extracted with EA, washed with sat. NaCl (aq.), dried over by Na2SC>4. The filtrate was evaporated under reduced pressure to give the crude product which was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, Cl 8 silica gel; mobile phase, CH3CN/H2O (0.5% NH4HCO3) = 1/1 increasing to CH3CN/H2O (0.5% NH4HCO3) = 1/0 within 20 min, the eluted product was collected at CH3CN/ H2O (0.5% NH4HCO3) = 1/0; Detector, UV 254 nm. This resulted in to give 6 (2.10 g, 3.98 mmol, 59.32% yield). ‘H-NMR (400 MHz, CDCI3): δ 7.40-7.28(m, 5H), 7.117.00(m, 1H), 6.19-6.14(m, 1H), 5.71-5.68(m, 4H), 4.95-4.94(m, 1H), 4.48-4.47(m, 1H), 4.054.03(m, 1H), 3.62-3.61(m, 1H), 3.46(m, 3H), 3.00-2.99(m, 1H), 1.22(m, 18H);31P-NMR (162 MHz, CDCI3): δ 18.134.
Préparation of Example 34 monomer: To a solution of 6 (2.10 g, 3.98 mmol) in DCM (21 mL) was added DCI (410 mg, 3.47 mmol). CEP (1.40 g, 4.65 mmol) was added in a N2 atmosphère. LCMS showed 6 was consumed completely. DCM and H2O was poured, the organic phase was washed with water and sat. NaCl (aq.), dried over by Na2SO4. The filtrate was evaporated under reduced pressure at 40°C to give the crude product which was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, Cl 8 silica gel; mobile phase, CH3CN/H2O (0.5% NH4HCO3) = 1/1 increasing to CH3CN/H2O (0.5% NH4HCO3) = 1/0 within 20 min, the eluted product was collected at CH3CN/ H2O (0.5% NH4HCO3) = 1/0; Detector, UV 254 nm. This resulted in to give Example 34 monomer (2.10 g, 2.88 mmol). 'HNMR (400 MHz, DMSO-c/6): δ 7.39-7.32(m, 6H), 6.21-6.1 l(m, 1H), 5.64-5.61(m, 4H), 4.914.85(m, 1H), 4.59(m, 1H), 4.28-4.25(m, 1H), 3.84-3.60(m, 5H), 3.36-3.36(m, 2H), 2.83-2.79(m, 2H), 1.18-1.14(m, 29H); 31P-NMR (162. MHz, DMSO-76): δ 149.588, 148.920, 17.355, 17.010.
199
Example 35. Synthesis of 5’ End Cap Monomer
Scheme-15
Préparation of (2): To a solution of 1 (5.90 g, 21.50 mmol) in DMF (60.00 mL), imidazole (4.39 g, 64.51 mmol) and TBSO (7.63 g, 49.56 mmol) were added. The mixture was stirred at r.t. for 3.5 hrs, LCMS showed 1 was consumed completely. Water was added and extracted with EA, dried over by Na2SC>4. The filtrate was evaporated under reduced pressure to give 2 (11.00 g, 21.91 mmol, 98.19% yield) for the next step. ESI-LCMS: m/z 225.1 [M+H]+.
Préparation of (3): To a solution of 2 (11.00 g, 21.91mmol) in THF (55.00 mL) was added TFA (110.00 mL) and H2O (55.00 mL) at 0°C,reaction mixture was stirred for 30 min. LCMS showed 2 was consumed completely. Then was extracted with EA, washed with sat. NaCl (aq.), dried over by Na2SO4. The filtrate was evaporated under reduced pressure to give the crude product which was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, Cl 8 silica gel; mobile phase, CH3CN/H2O (0.5% NH4HCO3) = 1/1 increasing to CH3CN/H2O (0.5% NH4HCO3) = 1/0 within 20 min, the eluted product was collected at CH3CN/ H2O (0.5% NH4HCO3) = 1/0; Detector, UV 254 nm. This resulted in to give 3 (6.20 g, 16.32 mmol, 72.94 % yield). ESI-LCMS: m/z 411.2 [M+H]+.
Préparation of (4): To a solution of 3 (3.50 g, 9.02 mmol) in DMSO (35.00 mL) was added EDCI (5.19 g, 27.06 mmol). Then pyridine (0.78 g, 9.92 mmol) and TFA (0.57 g, 4.96 mmol) was added in N2 atmosphère. The mixture was stirred for 3h at r.t. Water was poured into it and was extracted with EA, washed with sat. NaCl (aq.), dried over by Na2SC>4. The filtrate was
200 evaporated under reduced pressure to give the crude product which was directly used for next step. ESI-LCMS: m/z 406.2 [M+H]+.
Préparation of (5): To a solution of 4 in toluene (100.00 mL) was added 4a (5.73 g, 9.07 mmol) and KOH (916.3 g, 16.33 mmol). It was stirred for 3.5h at 40°C in N2 atmosphère. Then was extracted with EA, washed with water and sat. NaCl (aq.), dried over by Na2SO4. The filtrate was evaporated under reduced pressure to give the crude product which was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, Cl 8 silica gel; mobile phase, CH3CN/H2O (0.5% NH4HCO3) = 1/1 increasing to CH3CN/H2O (0.5% NH4HCO3) = 1/0 within 20 min, the eluted product was collected at CH3CN/ H2O (0.5% NH4HCO3) = 1/0; Detector, UV 254 nm. This resulted in to give 5 (5.02 g, 7.25 mmol, 80.44% yield). ESI-LCMS: m/z 693.2 [M+H]+;3IP-NMR (162 MHz, DMSO-</6): δ 17.811
Préparation of (6): To a solution of 5 (4.59 g, 6.63 mmol) in THF (46.00 mL) was added HCOOH (92.00 mL) and H2O (92.00 mL). It was stirred ovemight at r.t. NH4OH was poured into it and extracted with EA, washed with sat. NaCl (aq.), dried over by Na2SO4. The filtrate was evaporated under reduced pressure to give the crude product which was purified by FlashPrep-HPLC with the following conditions (IntelFlash-1): Column, Cl8 silica gel; mobile phase, CH3CN/H2O (0.5% NH4HCO3) = 1/1 increasing to CH3CN/H2O (0.5% NH4HCO3) = 1/0 within 20 min, the eluted product was collected at CH3CN/ H2O (0.5% NH4HCO3) = 1/0; Detector, UV 254 nm. This resulted in to give 6 (2.52 g, 4.36 mmol, 65.80% yield).
Préparation of Example 35 monomer: To a solution of 6 (2.00 g, 3.46 mmol) in DCM (21.00 mL) was added DCI (370.00 mg, 3.11 mmol) and CEP (1.12 g, 4.15 mmol) was added in N2 atmosphère. DCM and H2O was poured, the organic phase was washed with water and sat. NaCl (aq.), dried over by Na2SÜ4. The filtrate was evaporated under reduced pressure at 38°C to give the crude product which was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, Cl8 silica gel; mobile phase, CH3CN/H2O (0.5% NH4HCO3) = 1/1 increasing to CH3CN/H2O (0.5% NH4HCO3) = 1/0 within 20 min, the eluted product was collected at CH3CN/ H2O (0.5% NH4HCO3) = 1/0; Detector, UV 254 nm. This resulted in to give Example 35 monomer (2.10 g, 2.70 mmol, 78.07% yield). ‘H-NMR (400 MHz, DMSO-Jd): δ 7.39-7.32(m, 6H), 6.21-6.1 l(m, 1H), 5.64-5.61(m, 4H), 4.91-4.85(m, 1H), 4.59(m, 1H), 4.284.25(m, 1H), 3.84-3.60(m, 5H), 3.36-3.36(m, 2H), 2.83-2.79(m, 2H), 1.18-1.14(m, 29H). 31PNMR (162 MHz, DMSO-î76): δ 149.588, 148.920, 17.355, 17.010.
Example 36. Synthesis of Monomer
201
1) iBuCI, Pyridine
2) 0.5 N NaOH in pyr/MeOH/H2O
Example 36 monomer
Scheme-16
Préparation of (2): To a solution of 1 (35.0 g, 53.2 mmol) in DMF (350 mL) was added imidazole (9.0 g, 133.0 mmol) then added TBSC1 (12.0 g, 79.8 mmol) at 0°C. The mixture was stirred at r.t. for 14 hrs. TLC showed 1 was consumed completely. Water was added to the reaction. The product was extracted with EA, The organic layer was washed with NaHCCL and brine. Then the solution was concentrated under reduced pressure the crude 2 (41.6
g) as a white solid which was used directly for next step. ESI-LCMS: m/z 772 [M+H]+. Préparation of (3): To a solution of 2 (41.0 g, 53.1 mmol) in 3% DCA (53.1 mmol, 350 mL) and EtsSiH (53.1 mmol, 100 mL) at 0°C. The mixture was stirred at 0°C for 0.5 h.
TLC showed 2 was consumed completely. NaHCCL was added to the reaction. The product was extracted with EA, The organic layer was washed with NaHCCh and brine. Then the solution was concentrated under reduced pressure. The residue silica gel column chromatography (eluent, DCM/MeOH = 100:1-20:1). This resulted in to give 3 (20.0 g, 41.7 mmol, 78.6% over two step) as a white solid. ESI-LCMS: m/z 470 [M+H]+ ; 'H-NMR (400 MHz, DMSO-</6): δ 12.12 (s, 1H), 11.67 (s, 1H), 8.28 (s, 1H), 6.12-6.07 (dd, J=15Hz, 1H), 5.75 (d,J=5Hz, 1 H), 5.48-5.24 (m,
202
2H), 4.55-4.49 (m, 1H), 3.97 (s, 1H), 3.75-3.55 (m, 2H), 2.79-2.76 (m, 1H), 1.12 (d, J = 6 Hz, 6H), 0.88 (s, 9H), 0.1 l(d, J= 6 Hz, 6H).
Préparation of (4): To the solution of 3 (20 g, 42.6 mmol) in dry DCM (100 mL) and DMF (60 mL) was added PDC (20. g, 85.1 mmol), tert-butyl alcohol (63.1 g, 851.8 mmol) and AciO (43.4 g, 425.9 mmol) at r.t. under N2 atmosphère. And the reaction mixture was stirred at r.t. for 2 h. The solvent was removed to give a residue which was purified by silica gel column chromatography (eluent, PE: EA = 4:1-2:1) to give a residue which was purified by Flash-PrepHPLC with the following conditions (IntelFlash-1): Column, Cl8 silica gel; mobile phase, CH3CN/H2O (0.5% NH4HCO3) = 1/1 increasing to CH3CN/H2O (0.5% NH4HCO3) = 1/0 within 20 min, the eluted product was collected at CH3CN/ H2O (0.5% NH4HCO3) = 1/0; Detector, UV 254 nm. This resulted in to give 4 (16.0 g, 29.0 mmol, 68.2% yield) as a white solid. ESI-LCMS: m/z 540 [M+H]+ ; ‘H-NMR (400 MHz, DMSO-tZ6): δ 12.12 (s, 1H), 11.69 (s, 1H), 8.28 (s, 1H), 6.21-6.17 (dd, J= 15 Hz, 1H), 5.63-5.55 (m, 1H), 4.75-4.72 (m, 1H), 4.41 (d, J= 5 Hz, 1H), 2.79-2.76 (m, 1H), 1.46 (s, 9H), 1.13-1.11 (m, 6H), 0.90 (s, 9H), 0.14(d, J= 2 Hz, 6H).
Préparation of (5): To the solution of 4 (16.0 g, 29.6 mmol) in dry THF/MeODÆhO = 10/2/1 (195 mL) was added NaBÜ4 (3.4 g, 88.9 mmol) at r.t. and the reaction mixture was stirred at 50 °C for 2 h. After completion of reaction, adjusted pH value to 7 with CH3COOD, after addition of water, the resulting mixture was extracted with EA (300 mL). The combined organic layer was washed with water and brine, dried over Na2SO4, Then the solution was concentrated under reduced pressure the crude 5 (11.8 g) as a white solid which was used directly for next step. ESI-LCMS: m/z 402 [M+H]+.
Préparation of (6): To a solution of 5 (5.0 g, 12.4 mmol) in pyridine (50 mL) was added iBuCl (2.6 g, 24.9 mmol) at 0°C under N2 atmosphère. The mixture was stirred at r.t. for 14 h. TLC showed 5 was consumed completely. Then the solution diluted with EA. The organic layer was washed with NaHCCh and brine. Then the solution was concentrated under reduced pressure to give the crude. To a solution of the crude in pyridine (50 mL) was added 2N NaOH (MeOH/H2O=4:l, 15 mL) at 0°C. The mixture was stirred at 0°C for 10 min. Then the solution diluted with EA .The organic layer was washed with NH4CI and brine. Then the solution was concentrated under reduced pressure the residue was purified by Flash-Prep-HPLC with the following conditions(IntelFlash-l): Column, Cl 8 silica gel; mobile phase, CH3CN/H2O (0.5% NH4HCO3) =1/3 increasing to CH3CN/H2O (0.5% NH4HCO3)=4/1 within 25 min, the eluted product was collected at CH3CN/ H2O (0.5% NH4HCO3) =3/2; Detector, UV 254 nm. This resulted in to give 6 (6 g, 10.86 mmol, 87.17% yield) as a white solid. ESI-LCMS: m/z 472.2 [M+H]+; ‘H-NMR (400 MHz, DMSO-J6): δ 12.12 (s, 1H), 11.67 (s, 1H), 8.28 (s, 1H), 6.12-6.07
203 (dd, J= 15 Hz, 1H), 5.48-5.24 (m, 2H), 5.22 (s, 1H), 4.55-4.49 (m, 1H), 3.97 (d, J= 5 Hz, 1H), 2.79-2.76 (m, 1H), 1.12 (d, J= 6 Hz, 6H), 0.88 (s, 9H), 0.1 l(d, J= 6 Hz, 6H).
Préparation of (7): To a solution of 6 (3.8 g, 8.1 mmol) in pyridine (40 mL) was added DMTrCl (4.1 g, 12.1 mmol) at 20°C. The mixture was stirred at 20°C for 1 h. TLC showed 7 was consumed completely. Water was added to the reaction. The product was extracted with EA, The organic layer was washed with NaHCCh and brine. Then the solution was concentrated under reduced pressure to give the crude product of 7 (6 g, 7.6 mmol, 94.3% yield) as a yellow solid. ESI-LCMS: m/z 775 [M+H]+.
Préparation of (8): To a solution of 7 (6.0 g, 7.75 mmol) in THF (60 mL) was added TBAF (2.4 g, 9.3 mmol). The mixture was stirred at r.t. for 1 h. TLC showed 7 was consumed completely. Water was added to the reaction. The product was extracted with EA, The organic layer was washed with NaHCCL and brine. Then the solution was concentrated under reduced pressure, the residue was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, C18 silica gel; mobile phase, CH3CN/H2O (0.5% NH4HCO3) =1/1 increasing to CH3CN/H2O (0.5% NH4HCO3) =1/0 within 25 min, the eluted product was collected at CH3CN/ H2O (0.5% NH4HCO3) =4/1; Detector, UV 254 nm. This resulted in to give 8 (4.0 g, 5.9 mmol, 76.6% yield) as a white solid. ESI-LCMS: m/z 660 [M+H]+; Ή-NMR (400 MHz, DMSO-d6): δ 12.12 (s, 1H), 11.67 (s, 1H), 8.12 (s, 1H), 7.34-7.17 (m, 9H), 6.83-6.78 (m, 4H), 6.23-6.18 (m, 1H), 5.66 (d, J = 7 Hz, 1H), 5.48-5.35 (m, 1H), 4.654.54 (m, 1H), 3.72 (d, J = 2 Hz, 6H), 2.79-2.73 (m, 1H), 1.19-1.06 (m, 6H).
Préparation of Example 36 monomer: To a solution of 9 (4.0 g, 6.1 mmol) in DCM (40 mL) was added DCI (608 mg, 5.1 mmol) and CEP (2.2 g, 7.3 mmol) under N2 pro. The mixture was stirred at 20°C for 0.5 h. TLC showed 9 was consumed completely. The product was extracted with DCM, The organic layer was washed with H2O and brine. Then the solution was concentrated under reduced pressure and the residue was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, Cl8 silica gel; mobile phase, CH3CN/H2O (0.5% NH4HCO3) =1/1 increasing to CH3CN/H2O (0.5% NH4HCO3) = 1/0 within 25 min, the eluted product was collected at CH3CN/ H2O (0.5% NH4HCO3) =1/0; Detector, UV 254 nm. This resulted in to give Example 36 monomer (5.1 g, 5.81 mmol, 95.8% yield) as a white solid. ESILCMS: m/z 860 [M+H]+; *H-NMR (400 MHz, DMSO-tZô): δ 12.12 (s, 1H), 11.67 (s, 1H), 8.12 (s, 1H), 7.34-7.17 (m, 9H), 6.83-6.78 (m, 4H), 6.23-6.18 (m, 1H), 5.67-5.54 (m, 1H), 4.70-4.67 (m, 1H), 4.23-4.20 (m, 1H), 3.72 (m, 6H), 3.60-3.48 (m, 3H), 2.79-2.58 (m, 3H), 1.13-0.94 (m, 18H); 3IP-NMR (162 MHz, DMSO-î/6): δ 150.31, 150.26, 140.62, 149.57.
204
[0001] Ëxample 37: Synthesis of Monomer
TEMPO
ACN/H2O
DAIB
SOC12
MeOH
TBSCl Imidazole DMF
NaBD4
THF/CH3OD/D2O
Example 37 monomer
Scheme-17
Préparation of (2): To a solution of 1 (35 g, 130.2 mmol) in DMF (350 mL) was added imidazole (26.5 g, 390.0 mmol) then added TBSCl (48.7 g, 325.8 mmol) at 0°C. The mixture was stirred at r.t. for 14 h. TLC showed 1 was consumed completely. Water was added to the reaction. The product was extracted with EA, The organic layer was washed with NaHCCh and brine. Then the solution was concentrated under reduced pressure the crude 2 (64.6 g) as a white solid which was used directly for next step. ESI-LCMS: m/z 498 [M+H]+.
Préparation of (3): To a solution of 2 (64.6 g, 130.2 mmol) in THF (300 mL) and added TFA/H2O (1:1, 300 mL) at 0°C. The mixture was stirred at 0°C for 2 h. TLC showed 2 was consumed completely. NaHCCh was added to the reaction. The product was extracted with EA, The organic layer was washed with NaHCCb and brine. Then the solution was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent, DCM: MEOH = 100:1-20:1). This resulted in to give 3 (31.3 g, 81.7 mmol, 62.6% over two step) as a white solid. ESI-LCMS: m/z 384 [M+H]+.
205
Préparation of (4): To a solution of 3 (31.3 g, 81.7 mmol) in ACN/ H2O (1:1,350 mL) was added DAIB (78.0 g, 244.0 mmol) and Tempo (3.8 g, 24.4 mmol). The mixture was stirred at 40°C for 2 h. TLC showed 3 was consumed completely. Then filtered to give 4 (22.5 g, 55.5 mmol, 70.9%) as a white solid. ESI-LCMS: m/z 398 [M+H]+.
Préparation of (5): To a solution of 4 (22.5 g, 55.5 mmol) in MeOH (225 mL) held at 15° C with an ice/MeOH bath was added SOCI2 (7.6 mL, 94.5 mmol), dropwise at such a rate that the reaction temp did not exceed 7° C. After the addition was complété, cooling was removed, the reaction was allowed to stir at room temp. The mixture was stirred at r.t. for 14 h. TLC showed 4 was consumed completely. Then the solution was concentrated under reduced pressure to get crude 5 (23.0 g) as a white solid which was used directly for next step. ESILCMS: m/z 298 [M+H]+.
Préparation of (6): To a solution of 5 (23 g, 55.5 mmol) in DMF (220 mL) was added imidazole (11.6 g, 165.0 mmol) then added TBSC1 (12.3 g, 82.3 mmol) at 0°C. The mixture was stirred at 20°C for 14 h. TLC showed 1 was consumed completely. Water was added to the reaction. The product was extracted with EA, The organic layer was washed with NaHCCL and brine. Then the solution was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent, DCM: MEOH = 100:1—20:1). This resulted in to give 6 (21.3 g, 51.1 mmol, 90 % over two step) as a white solid. ESI-LCMS: m/z 412 [M+H]+.
Préparation of (7): To the solution of 6 (21.0 g, 51.0 mmol) in dry THF/MeODÆhO = 10/2/1 (260.5 mL) was added NaBÜ4 (6.4 g, 153.1 mmol) at r.t. and the reaction mixture was stirred at 50°C for 2 h. After completion of reaction, the resulting mixture was added CH3COOD to pH = 7, after addition of water, the resulting mixture was extracted with EA (300 mL). The combined organic layer was washed with water and brine, dried over Na2SO4. Then the solution was concentrated under reduced pressure and the residue was used for next step without further purification. ESI-LCMS: m/z 386 [M+H]+.
Préparation of (8): To a stirred solution of 7 (14.0 g, 35 mmol) in pyridine (50 mL) were added BzCl (17.2 g, 122.5 mmol) at 0°C under N2 atmosphère. The mixture was stirred at r.t. for 14 h. TLC showed 7 was consumed completely. Then the solution diluted with EA .The organic layer was washed with NaHCCh and brine. Then the solution was concentrated under reduced pressure and the residue was used for next step without further purification. To a solution of the crude in pyridine (300 mL) then added 2M NaOH (MeOH: H2O=4:1, 60 mL) at 0°C. The mixture was stirred at 0°C for 10 min. Then the solution diluted with EA. The organic layer was washed with NH4CI and brine. Then the solution was concentrated under reduced pressure and
206 the residue was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, Cl 8 silica gel; mobile phase, CH3CN/H2O (0.5% NH4HCO3) =1/3 increasing to CH3CN/H2O (0.5% NH4HCO3) =4/1 within 25 min, the eluted product was collected at CH3CN/ H2O (0.5% NH4HCO3) =3/2; Detector, UV 254 nm. This resulted in to give 8 (14 g, 28.02 mmol, 69.21% yield) as a white solid. ESI-LCMS: m/z 490 [M+H]+; ‘H-NMR (400 MHz, DMSO-76): δ 11.24 (s, 1H), 8.76 (s, 1H), 8.71 (m, 1H), 8.04 (d, J= 7 Hz, 2H),7.66-7.10 (m, 5H), 6.40-6.35 (dd, 1H), 5.71-5.56 (m, 1H), 5.16 (s, 1H), 4.79-4.72 (m, 1H), 4.01 (m, 1H), 0.91 (s, 9H), 0.14 (m, 6H).
Préparation of (9): To a solution of 8 (5.1 g, 10.4 mmol) in pyridine (50 mL) was added DMTrCl (5.3 g, 15.6 mmol). The mixture was stirred at r.t. for 1 h. TLC showed 8 was consumed completely. Water was added to the reaction. The product was extracted with EA, The organic layer was washed with NaHCCh and brine. Then the solution was concentrated under reduced pressure and the residue was used for next step without further purification. ESILCMS: m/z 792 [M+H]+.
Préparation of (10): To a solution of 9 (7.9 g, 10.0 mmol) in THF (80 mL) was added IM TB AF in THF (12 mL). The mixture was stirred at r.t. for 1 h. TLC showed 9 was consumed completely. Water was added to the reaction. The product was extracted with EA, The organic layer was washed with NaHCO3 and brine. Then the solution was concentrated under reduced pressure the residue was purified by Flash-Prep-HPLC with the following conditions (IntelFlash1): Column, Cl 8 silica gel; mobile phase, CH3CN/H2O (0.5% NH4HCO3) =1/1 increasing to CH3CN/H2O (0.5% NH4HCO3) =1/0 within 25 min, the eluted product was collected at CH3CN/ H2O (0.5% NH4HCO3) =4/1; Detector, UV 254 nm. This resulted in to give 10 as a white solid. ESI-LCMS: m/z 678 [M+H]+; ‘H-NMR (400 MHz, DMSO-76): δ 11.25 (s, 1H), 8.74 (s, 1H), 8.62 (s, 1H), 8.04 (d, J= 7 Hz, 2H),7.66-7.53 (m, 3H), 7.33-7.15 (m, 9H), 6.82-6.78 (m, 4H), 6.43 (d, J= 20 Ηζ,ΙΗ), 5.76-5.60 (m, 1H), 4.88-4.80 (m, 1H), 4.13 (d, J= 8 Hz, 1H), 3.71 (m, 6H).
Préparation of Example 37 monomer: To a solution of 10 (6.2 g, 9.1 mmol) in DCM (60 mL) was added DCI (1.1 g, 9.4 mmol) and CEP (3.3 g, 10.9 mmol) under N2 pro. The mixture was stirred at 20°C for 0.5 h. TLC showed 10 was consumed completely. The product was extracted with DCM, The organic layer was washed with H2O and brine. Then the solution was concentrated under reduced pressure and the residue was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, Cl8 silica gel; mobile phase, CH3CN/H2O (0.5% NH4HCO3) = 1/1 increasing to CH3CN/H2O (0.5% NH4HCO3) = 1/0 within 20 min, the eluted product was collected at CH3CN/ H2O (0.5% NH4HCO3) = 1/0; Detector, UV 254 nm. This
207 resulted in to give Example 37 monomer (7.5 g, 8.3 mmol, 90.7%) as a white solid. ESI-LCMS: m/z 878 [M+H]+; ‘H-NMR (400 MHz, DMSO-d6): δ 11.25 (s, 1H), 8.68-8.65 (dd, 2H), 8.04 (m, 2H),7.66-7.53 (m, 3H), 7.33-7.15 (m, 9H), 6.82-6.78 (m, 4H), 6.53-6.43 (m, 1H), 5.96-5.81 (m, 1H), 5.36-5.15 (m, 1H), 4.21 (m, 1H), 3.86-3.52 (m, 10H), 2.79-2.61 (m, 2H), 1.21-0.99 (m,
12H); 31P-NMR (162 MHz, DMSO-îZ6): δ 149.60, 149.56, 149.48.
Example 38. Synthesis of End Cap Monomer
TBSCl Imidazole DMF ,
NaBD4
THF/MeOD/D,O
TEMPO, DA1B
ACN/H,O= 1/1 _
S
TMSC1 BzCl NH4OH
OPOM MOPO-p=o
D—1 OPOM n ,ps D q' OPOM
HCOOH
CEP, PCI, DCM
OPOM MOPO~P=o D_^ pPOM D o OPOM
O Me MeO-p=Q f zOMe Px q OMe
PivCl, Nal ACN
9a
OPOM MOPO~p=o f pPOM o PxOPOM
9b h2, thf/d2o
Scheme-18
Préparation of (2): To a solution of 1 (20.0 g, 71.2 mmol) in dry pyridine (200.0 mL) was 10 added TBSCl (26.8 g, 177.9 mmol) and imidazole (15.6 g, 227.8 mmol). The mixture was stirred at r.t. for 15 h. TLC showed 1 was consumed completely. The reaction mixture was concentrated to give residue. The residue was quenched with DCM (300.0 mL). The DCM layer was washed with H2O (100.0 mL*2) and brine. The DCM layer concentrated to give crude 2 (45.8 g) as a yellow oil. The crude used to next step directly. ESI-LCMS m/z 510.5 [M+H]+.
208
Préparation of (3): To a mixture solution of 2 (45.8 g) in THF (300.0 mL) was added mixture of H2O (100.0 mL) and TFA (100.0 mL) at 0°C over 30min. Then the reaction mixture was stirred at 0°C for 4 h. TLC showed the 2 was consumed completely. The reaction mixture pH was adjusted to 7-8 with NH3.H2O (100 mL). Then the mixture was extracted with EA (500.0 mL*2). The combined EA layer was washed with brine and concentrated to give crude which was purified by c.c. (PE:EA = 5:1 - 1:0) to give compound 3 (21.0 g, 53.2 mmol, 74.7% yield over 2 steps) as a white solid. ESI-LCMS m/z 396.2 [M+H]+.
Préparation of (4): To a solution of 3 (21.0 g, 53.2 mmol) in ACN (100.0 mL) and water (100.0 mL) were added (diacetoxyiodo)benzene (51.0 g, 159.5 mmol) and TEMPO (2.5 g, 15.9 mmol), The reaction mixture was stirred at 40°C for 1 h. TLC showed the 3 was consumed completely. The reaction mixture was cooled down to r.t. and filtered, the filtrate was concentrated to give crude which was purified by crystallization (ACN) to give 4 (14.5 g, 35.4 mmol, 66.2% yield). ESI-LCMS m/z 410.1[M+H]+.
Préparation of (5): To a solution of 4 (14.5 g, 35.4 mmol) in toluene (90.0 mL) and MeOH (60.0 mL) was added trimethylsilyldiazomethane (62.5 mL, 2.0 M, 141.8 mmol) at 0°C, then stirred at r.t. for 2h. TLC showed the 4 was consumed completely. The solvent was removed under reduce pressure, the residue was purified by crystallization (ACN) to give 5 (10.0 g, 23.6 mmol, 66.6% yield). ESI-LCMS m/z 424.2 [M+H]+
Préparation of (6): To the solution of 5 (10.0 g, 23.6 mmol) in dry THF/MeODÆhO = 10/2/1 (100.0 mL) was added NaBÜ4 (2.98 g, 70.9 mmol) three times during an hour at 40°C, the reaction mixture was stirred at r.t. for 2.0 h. The resulting mixture was added CH3COOD change pH = 7.5, after addition of water, the resulting mixture was extracted with EA (50.0 mL*3). The combined organic layer was washed with water and brine, dried over Na2SCU, concentrated to give a residue which was purified by c.c. (PE/EA =1:1-1:0). This resulted in to give 6 (6.1 g, 15.4 mmol, 65.3% yield) as a white solid. ESI-LCMS m/z 398.1 [M+H]+; 'H-NMR (400 MHz, DMSO-Jô) δ 8.28 (s, 1H), 8.02 (s, 1H), 7.23 (s, 2H), 5.86 (d, J= 6.4 Hz, 1H), 5.26 (s, 1H), 4.424.41(m, 1H), 4.35-4.32 (m,lH), 3.82 (d, J= 2.6 Hz, 1H), 3.14 (s, 3H), 0.78 (s, 9H), 0.00 (d, J= 0.9 Hz, 6H).
Préparation of (7): To a solution of 6 (6.1 g, 15.4 mmol) in pyridine (60.0 mL) was added the benzoyl chloride (6.5 g, 46.2 mmol) drop wise at 5°C. The reaction mixture was stirred at r.t. for 2 h. TLC showed the 6 was consumed completely. The reaction mixture was cooled down to 10°C and quenched with H2O (20.0 mL), extracted with EA (200.0 mL*2), combined the EA layer. The organic phase was washed with brine and dried over Na2SO4, concentrated to give the crude (12.0 g) which was dissolved in pyridine (60.0 mL), cooled to 0°C, 20.0 mL NaOH (2 M
209 in methanol : H2O = 4:1) was added and stirred for 10 min. The reaction was quenched by saturated solution of ammonium chloride, the aqueous layer was extracted with EA (200.0 mL*2), combined the EA layer, washed with brine and dried over Na2SÛ4, concentrated. The residue was purified by c.c. (PE/EA = 10:1 - 1:1)to give 7 (7.0 g, 13.9 mmol, 90.2% yield). ESI-LCMS m/z 502.2 [M+H]+; ‘H-NMR (400 MHz,DMSO-î76) δ 11.24 (s, 1H, exchanged with D2O) 8.77 (s, 2H), 8.04-8.06 (m, 2H), 7.64-7.66 (m, 2H), 7.54-7.58 (m, 2H), 6.14-6.16 (d, J = 5.9 Hz, 1H), 5.20-5.23 (m, 1H),4.58-4.60 (m, 1H), 4.52-4.55 (m,lH), 3.99-4.01 (m, 1H), 3.34 (s, 4H), 0.93 (s, 9H), 0.14-0.15 (d, J = 1.44 Hz, 6H).
Préparation of (8): To a stirred solution of 7 (5.5 g, 10.9 mmol) in DMSO (55.0 mL) was added EDCI (6.3 g, 32.9 mmol), pyridine (0.9g, 10.9mmol) and TFA(0.6 g,5.5mmol), the reaction mixture was stirred at r.t. for 15 h. The reaction was quenched with water and extracted with EA (100.0 mL). The organic phase was washed by brine, dried over Na2SO4, The organic phase was evaporated to dryness under reduced pressure to give a residue 8 (4.8 g) which was used directly to next step. ESI-LCMS: m/z 517.1 [M+H2O]+
Préparation of (9b): A solution of 9a (35.0 g, 150.8 mmol) and Nal (90.5 g, 603.4 mmol) in dry ACN (180.0 mL) was added chloromethyl pivalate (113.6 g, 754.3 mmol) at r.t., the reaction was stirred at 80°C for 4 h. The reaction was cooled to r.t. and quenched by water, then the mixture was extracted with EA (500.0 mL *3), combined the organic layer was washed with saturated solution of ammonium chloride, followed by with brine and dried over Na2SO4. Then the organic layer was concentrated to give a residue which was purified by c.c., this resulted in to give 9b (38.0 g, 60.1mmol, 39.8% yield) as a white solid. ESI-LCMS m/z 655.2 [M+Na]+; ΉNMR (400 MHz, CDC13): δ 5.74-5.67 (m, 8H), 2.67 (t, J= 21.6 Hz, 2H), 1.23 (s, 36H).
Préparation of (9): 3.8 g 10% Pd/C was washed with dry THF (30.0 mL) three times.
Then transferred into a round-bottom flask charged with 9b (38.0 g, 60.Immol) and solvent (dry THF:D2O=5:1, 400.0 mL), the mixture was stirred at 80°C under IL H2balloon for 15 h. The reaction was cooled to r.t. and extracted with EA (500.0 mL *3), combined the organic layer was washed with brine and dried over Na2SO4. The residue 9 (3.0 g, 3.7 mmol, 38.8% yield) as a white solid was used directly to next step without further purification. ESI-LCMS m/z 657.2 [M+Na]+; ‘H-NMR (400 MHz, CDC13): δ 5.74-5.67 (m, 8H), 1.23 (s, 36H).
Préparation of (10): A solution of 8 (4.8 g, 9.6 mmol), 9 (7.3 g, 11.5 mmol) and K2CO3 (4.0 g, 38.8 mmol) in dry THF (60.0 mL) and D2O (20.0 mL) was stirred at r.t. 18h. LC-MS showed 8 was consumed completely. The product was extracted with EA (300.0 mL) and the organic layer was washed with brine and dried over Na2SO4. Then the organic layer was concentrated to give a residue which was purified by c.c. (PE/EA = 5:1-1:1) and MP LC. This
210 resulted in to give 10 (3.0 g, 3.7 mmol, 38.8% yield) as a white solid. ESI-LCMS m/z 806.4[M+H]+; *H-NMR (400 MHz, DMSO-76): δ 11.25 (s, 1H, exchanged with D2O) 8.75 (s, 2H), 8.07-8.05 (d, J= 8.0 Hz, 2H), 7.67-7.54 (m, 3H), 6.05 (d, J= 5.1 Hz, 1H), 5.65-5.58 (m, 4H), 4.80-4.70 (m, 2H), 4.59-4.57 (m,lH), 3.36 (s, 3H), 1.11 (s, 9H), 1.10 (s, 9H), 0.94 (s, 9H), 0.17-0.16 (m, 6H); 3IP NMR (162 MHz, DMSO-d6) δ 17.02.
Préparation of (11): To a round-bottom flask was added 10 (3.0 g, 3.7 mmol) in a mixture of H2O (30.0 mL), HCOOH (30.0 mL). The reaction mixture was stirred at 40°C for 15 hrs. LCMS showed the 10 was consumed completely. The reaction mixture was adjusted the pH = 6-7 with con. NH3.H2O (100.0 mL). Then the mixture was extracted with DCM (100.0 mL*3). The combined DCM layer was dried over Na2SO4. Filtered and filtrate was concentrated to give crude which was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, C18 silica gel; mobile phase, CH3CN/H2O (0.5% NH4HCO3) = 1/2 increasing to CH3CN/ H2O (0.5% NH4HCO3) = 1/0 within 20 min, the eluted product was collected at CH3CN/ H2O (0.5% NH4HCO3) = 3/2; Detector, UV 254 nm. To give product 11 (1.8 g, 2.6 mmol, 70.3% yield). ESI-LCMS m/z = 692.2[M+H]+; ‘H-NMR (400 MHz, DMSO-J6): δ 11.11 (s, 1H, exchanged with D2O) 8.71-8.75 (d, 7=14.4, 2H), 8.04-8.06 (m, 2H), 7.64-7.65 (m, 1H), 7.54-7.58 (m, 2H), 6.20-6.22 (d, 7=5.4, 2H), 5.74-5.75 (d, 7=5.72, 2H), 5.56-5.64 (m, 4H), 4.644.67 (m, 1H), 4.58-4.59(m, 1H), 4.49-4.52 (m, 1H), 3.37 (s, 3H), 1.09-1.10 (d, 7=1.96, 18H); 31P NMR (162 MHz, DMSO-J6) δ 17.46.
Préparation of Example 38 monomer: To a solution of 11 (1.8 g, 2.6 mmol) in DCM (18.0 mL) was added the DCI (276.0 mg, 2.3 mmol), then CEP[N(ipr)2]2 (939.5 mg, 3.1 mmol) was added. The mixture was stirred at r.t. for Ih. TLC showed 11 consumed completely. The reaction mixture was washed with H2O (50.0 mL*2) and brine (50.0 mL*2), dried over Na2SÛ4 and concentrated to give crude which was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, C18 silica gel; mobile phase, CTECNÆbO (0.5% NH4HCO3) = 1/1 increasing to CH3CN/ H2O (0.5% NH4HCO3) = 1/0 within 20 min, the eluted product was collected at CH3CN/ H2O (0.5% NH4HCO3) = 9/1; Detector, UV 254 nm. The product was concentrated to give Example 38 monomer (2.0 g, 2.2 mmol, 86.2% yield) as a white solid. ESILCMS m/z 892.3[M+H]+; 'H-NMR (400 MHz, DMSO-76): δ 11.27 (s, 1H, exchanged with D2O) 8.72-8.75 (m, 2H), 8.04-8.06 (m, 2H), 7.54-7.68 (m, 3H), 6.20-6.26 (m, 1H), 5.57-5.64 (m, 4H), 4.70-4.87 (m, 3H), 3.66-3.88 (m, 4H), 3.37-3.41 (m, 3H),2.82-2.86 (m, 2H) , 1.20-1.21 (m, 12H), 1.08-1.09 (m, 18H); 3IP-NMR (162 MHz, DMSO-76): δ 150.03, 149.19, 17.05, 16.81.
211
Example 39. Synthesis of 5’ End Cap Monomer
EDCI, Pyridine, TFA DMSO
OPOM
MOPO-p=Q
D-l ,ΟΡΟΜ 7 η ,Ρχ D OPOM
K2CO3,THF.D2O.
HCOOH,H2O^
CEPCI.DCl DCM
Scheme-19
Préparation of (6): To a stirred solution of 5 (8.0 g, 21.3 mmol, Scheme 3) in DMSO (80.0 mL) were added EDCI(12.2 g, 63.9mmol), pyridine(1.7 g,21.3mmol),TFA(1.2 g,10.6mmol) at r.t. And the reaction mixture was stirred at r.t. for 1.5 h. The reaction was quenched with water and extracted with EA (200.0 mL). The organic phase was washed by brine, dried over Na2SÛ4, The organic phase was evaporated to dryness under reduced pressure to give a residue 6 which was used directly to next step. ESI-LCMS: m/z 372.3 [M+H]+.
Préparation of (8): To a solution of K2CO3 (5.5 g, 8.3 mmol) in dry THF (60.0 mL) and D2O (20.0 mL) was added a solution of 6 (8.0 g, 21.5mmol) in dry THF(10.0 mL). The reaction mixture was stirred at r.t. ovemight. LC-MS showed 6 was consumed completely. The product was extracted with EA (300.0 mL) and the organic layer was washed with brine and dried over Na2SO4. Then the organic layer was concentrated to give a residue which was purified by FlashPrep-HPLC with the following conditions (IntelFlash-1): Column, Cl8 silica gel; mobile phase, CH3CN/H2O (0.5% NH4HCO3) = 2/3 increasing to CH3CN/H2O (0.5% NH4HCO3) = 3/2 within 20 min, the eluted product was collected at CH3CN/ H2O (0.5% NH4HCO3) = 1/1; Detector, UV 254 nm. This resulted in to give 8 (5.0 g, 7.3 mmol, 40.0%) as a white solid. ESI-LCMS: m/z 679.3 [M+H]+; ‘H-NMR (400 MHz, Chloroform-d): δ 9.91 (s, 1H), 7.29 (d, J= 8.1 Hz, 1H), 5.82 (d, J= 2.7 Hz, 1H), 5.72 (d, J= 8.1 Hz, 1H), 5.65 - 5.54 (m, 4H), 4.43 (dd, J= 7.2, 3.2 Hz, 1H), 3.92 (dd, J= 7.2, 5.0 Hz, 1H), 3.65 (dd, J= 5.1, 2.7 Hz, 1H), 3.44 (s, 3H), 1.13 (s, 18H), 0.82 (s, 9H), 0.01 (d, J= 4.8 Hz, 6H); 31P NMR (162 MHz, Chloroform-d): δ 16.40.
Préparation of (9): To a solution of HCOOH (50.0 mL) and H2O (50.0 mL) was added 8 (5.0 g,7.3 mmol). The reaction mixture was stirred at 40°C ovemight. LC-MS showed 8 was 212 consumed completely. A solution of NaHCCh (500.0 mL) was added. The product was extracted with EA (300.0 mL) and the organic layer was washed with brine and dried over Na2SO4. Then the organic layer was concentrated to give a residue which was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, Cl8 silica gel; mobile phase, CH3CN/H2O (0.5% NH4HCO3) = 2/3 increasing to CH3CN/H2O (0.5% NH4HCO3) = 3/2 within 20 min, the eluted product was collected at CH3CN/ H2O (0.5% NH4HCO3) = 1/1; Detector, UV 254 nm. This resulted in to give 9 (3.0 g, 5.4 mmol, 73.2%) as a white solid. ESI-LCMS: m/z 565.2 [M+H]+; 'H-NMR (400 MHz, DMSO-J6): δ 11.43 (s, 1H), 7.64 (d, J= 8.1 Hz, 1H), 5.83 (d, J = 4.3 Hz, 1H), 5.69 - 5.56 (m, 5H), 5.54 (d, J= 6.7 Hz, 1H), 4.37 (dd, J= 6.1, 2.9 Hz, 1H), 4.12 (q, .7=6.1 Hz, 1H), 3.96 (dd,J=5.4, 4.3 Hz, 1H), 3.39 (s, 3H), 1.16 (s, 18H); 31P NMR (162 MHz, DMSO-î/ô): δ 17.16.
Préparation of Example 39 monomer: To a suspension of 9 (2.6 g, 4.6 mmol) in DCM (40.0 mL) was added DCI (0.5 g, 5.6 mmol) and CEP[N(iPr)2]2 (1.7 g, 5.6 mmol). The mixture was stirred at r.t. for 1.0 h. LC-MS showed 9 was consumed completely. The solution was washed with water twice and washed with brine and dried over Na2SO4. Then concentrated to give a residue which was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, C18 silica gel; mobile phase, CH3CN/H2O (0.5% NH4HCO3) = 1/1 increasing to CH3CN/H2O (0.5% NH4HCO3) = 1/0 within 20 min, the eluted product was collected at CH3CN/ H2O (0.5% NH4HCO3) = 1/0; Detector, UV 254 nm. This resulted in to give Example 39 monomer (3.0 g, 3.9 mmol, 85.2%) as a white solid. .ESI-LCMS: m/z 765.3 [M+H]+; ‘H-NMR (400 MHz, DMSO-î76): δ 11.44 (s, 1H), 7.71 (dd, J= 8.1, 3.8 Hz, 1H), 5.81 (dd, J= 4.4, 2.5 Hz, 1H), 5.74-5.53 (m, 5H), 4.59-4.33 (m, 2H), 4.20-4.14 (m, 1H), 3.88-3.53 (m, 4H), 3.39 (d, J= 16.2 Hz, 3H), 2.80 (td, J= 5.9, 2.9 Hz,2H), 1.16 (d, J= 1.9 Hz, 30H); 31PNMR (162 MHz, DMSO-tZô): δ 147.68, 149.16, 16.84, 16.55.
Example 40. Synthesis of Monomer
213
BzCl Pyridine
IM NaOH Pyridine
DMTrCI
Pyridine
TBSC) qCD3
TBAF THF
DCI; CEP[N(iPr)2]2 DCM
Example 40 monomer
Scheme-20
Préparation of (2): To a solution of 1 (26.7 g*2, 0.1 mol) in DMF (400 mL) was added sodium hydride (4.8 g, 0.1 mol) for 30 min, then was added CD3I (16 g, O.lmol) at 0°C for 2.5 hr (ref. for sélective 2’-O-alkylation reaction conditions , J. Org. Chem. 1991, 56, 5846-5859). The mixture was stirring at r.t. for another Ih. LCMS showed the reaction was consumed. The mixture was filtered and the clear solution was evaporated to dryness and was evaporated with CH3OH. The crude was purified by slica gel column (SiO2, DCM/MeOH = 50:1-15:1). This resulted in to give the product 2 (35.5 g, 124.6 mmol, 62% yield) as a solid. ESI-LCMS: m/z 285 [M+H]+ .
Préparation of (3): To a solution of 2 (35.5 g, 124.6 mmol) in pyridine (360 mL) was added imidazole (29.7 g, 436.1 mmol) and TBSC1 (46.9 g, 311.5 mmol). The mixture was stirred at r.t. over night. LCMS showed 2 was consumed completely. The reaction was quenched with water (500 mL). The product was extracted into ethyl acetate (1 L). The organic layer was washed with brine and dried over anhydrous Na2SO4. The crude was purified by slica gel column (SiO2, PE/EA = 4:1-1:1). This resulted in to give the product 3 (20.3 g, 39.6 mmol, 31.8% yield) as a solid. ESI-LCMS: m/z 513 [M+H]+; Ή-NMR (400 MHz, DMSO-î/6): δ 8.32 (m, 1H), 8.13 (m, 1H), 7.31 (m, 2H), 6.02-6.01(d, J= 4.0 Hz, 1H), 4.60-4.58 (m, 1H), 4.49-4.47(m,lH), 3.96-3.86 (m, 2H), 3.72-3.68 (m, 1H), 0.91-0.85 (m, 18H), 0.13-0.01 (m, 12H).
Préparation of (4): To a solution of 3 (20.3 g, 39.6 mmol) in THF (80 mL) was added TFA (20 mL) and water (20 mL) at 0°C. The reaction mixture was stirred at 0°C for 5 h. LC-MS showed 3 was consumed completely. Con. NH4OH was added to the mixture at 0°C to quench the reaction until the pH = 7.5. The product was extracted into ethyl acetate (200 mL). The organic layer was washed with brine and dried over anhydrous Na2SO4. The solution was then concentrated under reduced pressure and the residue was washed by PE/EA = 5:1. This resulted in to give 4 (10.5 g, 26.4 mmol, 66.6% yield) as a white solid. ESI-LCMS: m/z 399 [M+H]+; ’H
214
NMR (400 MHz, DMSO-76): δ 8.41 (m, 1H), 8.14 (m, 1H), 7.37 (m, 2H), 5.99-5.97(d, J= 8.0 Hz, 1H), 5.43 (m, 1H), 4.54-4.44 (m,2H), 3.97-3.94 (m, 1H), 3.70-3.53 (m, 2H), 0.91 (m, 9H), 0.13-0.12 (m, 6H).
Préparation of (5): To a solution of 4 (10.5 g, 26.4 mmol) in ACN/H2O = 1:1 (100 mL) was added DAIB (25.4 g, 79.2 mmol) and TEMPO (1.7 g, 7.9 mmol). The reaction mixture was stirred at 40°C for 2 h. LCMS showed 4 was consumed. The mixture was diluted with EA and water was added. The product was extracted with EA. The organic layer was washed with brine and dried over anhydrous Na2SO4. The solution was then concentrated under reduced pressure and the residue was washed by ACN. This resulted in to give 5 (6.3 g, 15.3 mmol, 57.9% yield) as a white solid. ESI-LCMS: m/z 413 [M+H]+; Y-NMR (400 MHz, DMSO-î76): δ = 8.48 (m, 1H), 8.16 (m, 1H), 7.41 (m, 2H), 6.12-6.10(d, J= 8.0 Hz, 1H), 4.75-4.73 (m, 1H), 4.42-4.36 (m, 2H), 3.17 (m, 6H), 2.07 (m, 2H), 0.93 (m, 9H), 0.17-0.15 (m, 6H).
Préparation of (6): To a solution of 5 (6.3 g, 15.3 mmol) in toluene (36 mL) and methanol (24 mL) was added (trimethylsilyl)diazomethane (7.0 g, 61.2 mmol) till the yellow color not disappear at r.t. for 2 min. LCMS showed the reaction was consumed. The solvent was removed to give the cured 6 (6.0 g) as a solid witch used for the next step. ESI-LCMS: m/z 427 [M+H]+ ; ‘H-NMR (400 MHz, DMSO-J6): δ 8.45 (m, 1H), 8.15 (m, 1H), 7.35 (m, 2H), 6.12-6.10(d, J= 8.0 Hz, 1H), 4.83-4.81 (m, 1H), 4.50-4.46 (m, 1H), 3.73 (m, 3H), 3.31 (m, 1H), 0.93 (m, 9H), 0.15-0.14 (m, 6H).
Préparation of (7): To the solution of 6 (6 g) in dry THF/MeODÆbO = 10/2/1 (78 mL) was added NaBÜ4 (2.3 g, 54.8 mmol) at r.t. And the reaction mixture was stirred at r.t for 2.5 hr. After completion of reaction, adjusted pH value to 7 with CH3COOD, after addition of water, the resulting mixture was extracted with EA (100 mL). The combined organic layer was washed with water and brine, dried over Na2SC>4, and concentrated to give 7 (5.7 g) which was used for the next step. ESI-LCMS: m/z 401 [M+H]+ .
Préparation of (8): To a solution of 7 (5.7 g) in pyridine (60 mL) was added BzCl (10.0 g, 71.3 mmol) under ice bath. The reaction mixture was stirred at r.t. for 2.5 hrs. LCMS showed 7 was consumed. The mixture was diluted with EA and water was added. The product was extracted with EA. The crude was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, C18 silica gel; mobile phase, CH3CN/H2O (0.5% NH4HCO3) = 1/1 increasing to CH3CN/H2O (0.5% NH4HCO3) = 1/0 within 25 min, the eluted product was collected at CH3CN/H2O (0.5% NH4HCO3) = 7/3; Detector, UV 254 nm. This resulted in to give the crude 8 (6.2 g, 8.7 mmol, 57% yield, over two steps) as a white solid. ESI-LCMS: m/z 713 [M+H]+.
215
Préparation of (9): To a solution of 8 (6.2 g, 8.7 mmol) in pyridine (70 mL) and was added IM NaOH (MeOH/HiO = 4/1) (24 mL). LCMS showed 8 was consumed. The mixture was added saturated NH4CI till pH = 7.5. The mixture was diluted with water and EA. The organic layer was washed with brine and dried over Na?SO4 and concentrated to give the crude. The crude was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, Cl8 silica gel; mobile phase, CH3CN/H2O (0.5% NH4HCO3) = 1/1 increasing to CH3CN/H2O (0.5% NH4HCO3) = 1/0 within 25 min, the eluted product was collected at CH3CN/ H2O (0.5% NH4HCO3) = 67/33 Detector, UV 254 nm. This resulted in to give the product 10 (4.3 g, 8.5 mmol, 98% yield) as a white solid. ESI-LCMS: m/z 505 [M+H]+; 'H-NMR (400 MHz, DMSO-Jâ): δ 11.23 (m, 1H), 8.77 (m, 2H), 8.06-8.04 (m, 2H), 7.66-7.63 (m, 2H), 7.577.53 (m, 3H), 6.16-6.14 (d, J= 8.0 Hz, 1H), 5.17 (m, 1H), 4.60-4.52 (m, 2H), 3.34 (m, 1H), 0.93 (m, 9H), 0.14 (m, 6H).
Préparation of (10): To a stirred solution of 9 (4.3 g, 8.5 mmol) in pyridine (45 mL) were added DMTrCl (3.3 g, 9.8 mmol) at r.t. And the reaction mixture was stirred at r.t for 2.5 hr. With ice-bath cooling, the reaction was quenched with water and the product was extracted into EA. The organic layer was washed with brine and dried over Na2SO4 and concentrated to give the crude. The crude was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, Cl8 silica gel; mobile phase, CH3CN/H2O (0.5% NH4HCO3) = 1/1 increasing to CH3CN/H2O (0.5% NH4HCO3) = 1/0 within 25 min, the eluted product was collected at CH3CN/ H2O (0.5% NH4HCO3) =97/3 Detector, UV 254 nm. This resulted in to give the product 10 (6.5 g, 8.1 mmol, 95% yield) as a white solid. ESI-LCMS: m/z 807 [M+H]+; ’HNMR (400 MHz, DMSO-J6): δ 11.23 (m, 1H), 8.70-8.68 (m, 2H), 8.04-8.02 (m, 2H), 7.66-7.62 (m, 1H), 7.56-7.52 (m, 2H), 7.35-7.26 (m, 2H), 7.25-7.17 (m, 7H), 6.85-6.82 (m, 4H), 6.18-6.16 (d, J= 8.0 Hz, 1H), 4.73-4.70 (m, 1H), 4.61-4.58 (m, 1H), 3.71 (m, 6H), 3.32 (m, 1H), 0.83 (m, 9H), 0.09-0.03 (m, 6H).
Préparation of (11): To a solution of 10 (3.5 g, 4.3 mmol) in THF (35 mL) was added 1 M TBAF solution (5 mL). The reaction mixture was stirred at r.t. for 1.5 h. LCMS showed 10 was consumed completely. Water (100 mL) was added. The product was extracted with EA (100 mL) and the organic layer was washed with brine and dried over Na2SO4. Then the organic layer was concentrated to give a residue which was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, C18 silica gel; mobile phase, CH3CN/H2O (0.5% NH4HCO3) = 2/3 increasing to CH3CN/H2O (0.5% NH4HCO3) = 1/0 within 20 min, the eluted product was collected at CH3CN/H2O (0.5% NH4HCO3) = 62/38; Detector, UV 254 nm. This resulted in to give 11 (2.7 g, 3.9 mmol, 90.7%) as a white solid. ESI-LCMS: m/z 693 [M+H]+ .
216
Préparation of Example 40 monomer: To a suspension of 11 (2.7 g, 3.9 mmol) in DCM (30 mL) was added DCI (0.39 g, 3.3 mmol) and CEP[N(iPr)2]2 (1.4 g, 4.7 mmol). The mixture was stirred at r.t. for 2 h. LC-MS showed 11 was consumed completely. The solution was washed with water twice and washed with brine and dried over Na2SO4. Then concentrated to give a residue which was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, Cl 8 silica gel; mobile phase, CH3CN/H2O (0.5% NH4HCO3) = 1/1 increasing to CH3CN/H2O (0.5% NH4HCO3) = 1/0 within 20 min, the eluted product was collected at CH3CN/ H2O (0.5% NH4HCO3) = 73/27; Detector, UV 254 nm. This resulted in to give Example 40 monomer (3.3 g, 3.7 mmol, 94.9%) as a white solid. ESI-LCMS: m/z 893
[M+H]+; ‘H-NMR (400 MHz, DMSO-î/6): δ = 11.24 (m, 1H), 8.66-8.64 (m, 2H), 8.06-8.03 (m,
2H), 7.65-7.53(m, 3H), 7.42-7.38 (m, 2H), 7.37-7.34 (m, 2H), 7.25-7.19 (m, 7H), 6.86-6.80 (m, 4H), 6.20-6.19 (d, J= 4.0 Hz, 1H), 4.78 (m, 2H), 4.22-4.21 (m, 1H), 3.92-3.83 (m, 1H), 3.72 (m, 6H), 3.62-3.57 (m, 3H), 2.81-2.78 (m, 1H), 2.64-2.61 (m, 1H), 1.17-1.04 (m, 12H); 31P-NMR (162 MHz, DMSO-îZô): δ 149.51, 149.30.
Example 41. Synthesis of Monomer
217
BzO' ΟΒζ
ch3nh.
DPC
NalICOj DMF
AgNO3 collidine
TrtCl Pyridine
DAST Pyridine DCM
TrtÔ
9
Pyridine
BzCl DCM
Scheme-21
Préparation of (3): To the solution of 1 (70 g, 138.9 mmol) in dry acetonitrile (700 mL) was added 2 (27.0 g, 166.7 mmol), BSA (112.8 g, 555.5 mmol). The mixture was stirred at 50°C for 1 h. Then the mixture was cooled to -5°C and TMSOTf (46.2 g, 208.3 mmol) slowly added to the mixture. Then the reaction mixture was stirred at r.t for 48 h. Then the solution was cooled to 0°C and saturated aq. NaHCCh was added and the resulting mixture was extracted with EA. The combined organic layer was washed with water and brine, dried over NaiSCU, and concentrated under reduced pressure to give a residue which was purified by silica gel column chromatography (eluent, PE: EA=3:1~1:1) to give 3 (70 g, 115.3 mmol, 81.6%) as a white solid. ESI-LCMS: m/z 605 [M-H]+ .
Préparation of (4): To the solution of 3 (70.0 g, 115.3 mmol) in méthylammonium solution (1 M, 700 mL), and the reaction mixture was stirred at 40 °C for 15 h. After completion of reaction, the resulting mixture was concentrated. The residue was crystallized from EA. Solid was isolated by filtration, washed with PE and dried ovemight at 45°Cin vacuum to give 4 (31.0
218 g, 105.4 mmol, 91.1%) as a white solid. ESI-LCMS: m/z 295 [M+H]+; Ή-NMR (400 MHz, DMSO): δ 11.63 (s, 1H) , 8.07-7.99 (m, 1H),7.81 (d, J= 8.4 Hz, 1 H), 7.72-7.63 (m, 1H), 7.347.26 (m, 1H), 6.18 (d, J= 6.4 Hz, 1H), 5.24 (s, 1H), 5.00 (s, 2H), 4.58-4.47 (m, 1H), 4.19^1.10 (m, 1H), 3.85-3.77 (m, 1H), 3.75-3.66 (m, 1H), 3.66-3.57 (m, 1H).
Préparation of (5): To the solution of 4 (20.0 g, 68.0 mmol) in dry DMF (200 mL) was added DPC (18.9 g, 88.0 mmol) and NaHCCh (343 mg, 4 mmol) at r.t, and the reaction mixture was stirred at 150°C for 35 min. After completion of reaction, the resulting mixture was poured into tert-Butyl methyl ether (4 L). Solid was isolated by filtration, washed with PE and dried□ in vacuum to give crude 5 (21.0 g) as a brown solid which was used directly for next step (ref for 5, Journal of Organic Chemistry, 1989, vol. 33, p. 1219 - 1225). ESI-LCMS: m/z 275 [M-H]'.
Préparation of (6): To the solution of 5 (crude, 21.0 g) in Pyridine (200 mL) was added AgNCL (31.0 g, 180.0 mmol) and collidine (88.0 g, 720 mmol) and TrtCl (41.5 g, 181 mmol) at r.t, and the reaction mixture was stirred at r.t for 15 h. After addition of water, the resulting mixture was extracted with EA. The combined organic layer was washed with water and brine, dried over Na2SÜ4, and concentrated to give the crude. The crude was by Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, Cl 8 silica gel; mobile phase, CH3CN/H2O (0.5% NH4HCO3) = 1/1 increasing to CH3CN/H2O (0.5% NH4HCO3) = 1/0 within 20 min, the eluted product was collected at CH3CN/ H2O (0.5% NH4HCO3) = 1/0; Detector, UV 254 nm. This resulted in to give 6 (10.0 g, 13.1 mmol, 20% yield over 3 steps) as a white solid. ESILCMS: m/z 761 [M+H]+.
Préparation of (7): To the solution of 6 (10.0 g, 13.1 mmol) in THF (100 mL) was added 6 N NaOH (30 mL) at r.t, and the reaction mixture was stirred at r.t for 1 hr. After addition of NH4CI, the resulting mixture was extracted with EA. The combined organic layer was washed with water and brine, dried over Na2SO4, and concentrated under reduced pressure and the residue was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, Cl 8 silica gel; mobile phase, CH3CN/H2O (0.5% NH4HCO3) = 1/1 increasing to CH3CN/H2O (0.5% NH4HCO3) = 1/0 within 20 min, the eluted product was collected at CH3CN/ H2O (0.5% NH4HCO3) = 9/1; Detector, UV 254 nm. This resulted in to give 7 (9.3 g, 11.9 mmol, 90%) as a white solid. ESI-LCMS: m/z 777 [M-H]’; Ή-NMR (400 MHz, DMSO-J6): δ 11.57 (s, 1H), 8.02 (d, J= 8.7 Hz, 1H), 7.88-7.81 (m, 1H), 7.39-7.18 (m, 30H), 7.09-6.99 (m, 30H), 6.92-6.84 (m, 30H), 6.44 (d, J= 4.0 Hz, 1H), 4.87 (d, J= 4.0 Hz, 1H), 4.37^1.29 (m, 1H), 4.003.96 (m, 1H), 3.76-3.70 (m, 1H), 3.22-3.13 (m, 1H), 3.13-3.04 (m, 1H).
Préparation of (8): To the solution of 7 (8.3 g, 10.7 mmol) in dry DCM (80 mL) was added Pyridine (5.0 g, 64.2 mmol) and DAST (6.9 g, 42.8 mmol) at 0°C, and the reaction
219 mixture was stirred at r.t for 15 hr. After addition of NH4CI, the resulting mixture was extracted with DCM. The combined organic layer was washed with water and brine, dried over NasSCU, and concentrated under reduced pressure and the residue was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, C18 silica gel; mobile phase, CH3CN/H2O (0.5% NH4HCO3) = 1/1 increasing to CH3CN/H2O (0.5% NH4HCO3) = 1/0 within 20 min, the eluted product was collected at CH3CN/ H2O (0.5% NH4HCO3) = 1/0; Detector, UV 254 nm. This resulted in to give 8 (6.8 g, 8.7 mmol, 81.2%) as a white solid. ESI-LCMS: m/z 779 [ΜΗ]4·; 19F-NMR (376 MHz, DMSO-î/6): δ -183.05.
Préparation of (9): To the solution of 8 (5.8 g, 7.5 mmol) in dry ACN (60 mL) was added TEA (1.5 g, 15.1 mmol), DMAP (1.84 g, 15.1 mmol) and TPSC1 (4.1 g, 13.6 mmol) at r.t, and the reaction mixture was stirred at room température for 3 h under N2 atmosphère. After completion of reaction, the mixture was added NH3.H2O (12 mL). After addition of water, the resulting mixture was extracted with EA. The combined organic layer was washed with water and brine, dried over Na2SO4, and concentrated under reduced pressure and the residue was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, Cl8 silica gel; mobile phase, CH3CN/H2O (0.5% NH4HCO3) = 1/1 increasing to CH3CN/H2O (0.5% NH4HCO3) = 1/0 within 20 min, the eluted product was collected at CH3CN/ H2O (0.5% NH4HCO3) = 1/0; Detector, UV 254 nm. This resulted in to give 9 (5.5 g, 7 mmol, 90.2%) as a white solid. ESI-LCMS: m/z 780 [M+H]+.
Préparation of (10): To a solution of 9 (5.5 g, 7 mmol) in DCM (50 mL) with an inert atmosphère of nitrogen was added pyridine (5.6 g, 70.0 mmol) and BzCl (1.2 g, 8.5 mmol) in order at 0°C. The reaction solution was stirred for 30 minutes at room température. The solution was diluted with DCM (100 mL) and the combined organic layer was washed with water and brine, dried over Na2SC>4, and concentrated under reduced pressure to give a residue which was purified by silica gel column chromatography (eluent, PE: EA=5:1~2:1) to give 10 (5.4 g, 6.1 mmol, 90.6%) as a white solid. ESI-LCMS: m/z 884 [M+H]+; 19F-NMR (376 MHz, DMSO-76): δ-183.64.
Préparation of (11): To the solution of 10 (5.4 g, 6.1 mmol) in the solution of DCA (6%) in DCM (60 mL) was added TES (15 mL) at r.t, and the reaction mixture was stirred at room température for 5-10 min. After completion of reaction, the resulting mixture was added NaHCCE, the resulting mixture was extracted with DCM. The combined organic layer was washed with water and brine, dried over Na2SC>4, and concentrated under reduced pressure and the residue was crystallized from EA. Solid was isolated by filtration, washed with PE and dried
220 ovemight at 45° J in vacuum to give 11 (2.0 g, 5.0 mmol, 83.2%) as a white solid. ESI-LCMS: m/z 400 [M+H]+ .
Préparation of (12): To a solution of 11 (2.0 g, 5.0 mmol) in dry Pyridine (20 mL) was added DMTrCl (2.0 g, 6.0 mmol). The reaction mixture was stirred at r.t. for 2.5 h. LCMS showed 11 was consumed and water (200 mL) was added. The product was extracted with EA (200 mL) and the organic layer was washed with brine and dried over Na2SO4 and concentrated to give the crude. The crude was purified by c.c. (PE: EA = 4:1-1:1) to give crude 12. The crude was further purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, Cl 8 silica gel; mobile phase, CH3CN/H2O (0.5% NH4HCO3) = 1/1 increasing to CH3CN/H2O (0.5% NH4HCO3) = 1/0 within 20 min, the eluted product was collected at CH3CN/ H2O (0.5% NH4HCO3) = 1/0; Detector, UV 254 nm. This resulted in to give 12 (2.1 g, 3 mmol, 60%) as a white solid. ESI-LCMS: m/z 702 [M+H]+; *H-NMR (400 MHz, DMSO-î/6): δ 12.63 (s, 1H), 8.54 (d, J= 7.8 Hz, 1H), 8.25 (d, J= 7.2 Hz, 2H), 7.82 (d, J= 3.6 Hz, 2H), 7.67-7.58 (m, 1H), 7.57-7.49 (m, 2H), 7.49-7.39 (m, 1H), 7.39-7.31 (m, 2H), 7.27-7.09 (m, 7H), 6.82-6.69 (m, 4H), 6.23 (d, J= 26.1 Hz, 1H), 5.59-5.49 (m, 1H), 4.83-4.61 (m, 1H), 4.15-4.01 (m, 1H), 3.743.59 (m, 6H), 3.33-3.28 (m, 1H), 3.16-3.05 (m, 1H). 19F-NMR (376 MHz, DMSO-76): δ -191.66.
Préparation of Example 41 monomer: To a suspension of 12 (2.1 g, 3.0 mmol) in DCM (20 mL) was added DCI (310 mg, 2.6 mmol) and CEP[N(iPr)2]2 (1.1 g, 3.7 mmol). The mixture was stirred at r.t. for 1 h. LC-MS showed 12 was consumed completely. The solution was washed with water twice and washed with brine and dried over Na2SO4. Then concentrated to give the crude. The crude was by Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, Cl 8 silica gel; mobile phase, CH3CN/H2O (0.5% NH4HCO3) = 1/1 increasing to CH3CN/H2O (0.5% NH4HCO3) = 1/0 within 20 min, the eluted product was collected at CH3CN/ H2O (0.5% NH4HCO3) = 1/0; Detector, UV 254 nm. This resulted in to give Example 41 monomer (2.1 g, 2.3 mmol, 80.0%) as a white solid. ESI-LCMS: m/z 902 [M+H]+; *H-NMR (400 MHz, DMSO-Jô): δ 12.64 (s, 1H), 8.54 (d, J= 7.6 Hz, 1H), 8.24 (d, J= 7.7 Hz, 2H), 7.937.88 (m, 2H), 7.67-7.58 (m, 1H), 7.56-7.42 (m, 3H), 7.41-7.29 (m, 2H), 7.27-7.08 (m, 7H), 6.826.64 (m, 4H), 6.37-6.18 (m, 1H), 6.03-5.72 (m, 1H), 5.26-4.83 (m, 1H), 4.28-4.12 (m, 1H), 3.883.72 (m, 1H), 3.71-3.37 (m, 9H), 3.15-3.00 (m, 1H), 2.83-2.75 (m, 1H), 2.66-2.57 (m, 1H), 1.210.88 (m, 12H). 19F-NMR (376 MHz, DMSO-îZ6): δ -189.71.31P-NMR (162 MHz, DMSO-îZô): δ
149.48, 149.50, 148.95, 148.88.
221
Ëxample 42. Synthesis of Monomer
BzO' OBz
H
1a
BSA
TMSOTf
ACN
BzO' OBz ch3nh2
TrtCl
DMAP Pyridine
TrtO' OH
TrtCl collidine AgNO3
DMF
Tf-Cl Et3N DCM
TrtO' 'OH ho' 'oh
KOAc; DMF
CH3NH2
DAST; Pyridine
DCM
TrtO' V
DMTrCl Pyridine
Example 42 monomer
Scheme-22
Préparation of (2): To a solution of 1 (40.0 g, 79.3 mmol), la (7.6 g, 80.1 mmol) in ACN (100 mL). Then added BSA (35.2 g, 174.4 mmol) under N2 atmosphère. The mixture was stirred at 50°C for 1 h until the solution was clear. Then cool down to 0°C and dropped TMSOTf (18.5 g, 83.2 mmol).The mixture was stirred at 75°C for 1 h, TLC showed 1 was consumed completely. Then the solution was diluted.with EA, washed with H2O twice. The solvent was concentrated under reduced pressure and the residue was used for next step. ESILCMS: m/z 540 [M+H]+.
Préparation of (3): To a solution of 2 (37.1 g, 68.7 mmol) in 30%CH2NH2/MeOH solution (200 mL). The mixture was stirred at 25°C for 2 h. TLC showed 2 was consumed completely. The solvent was concentrated under reduced pressure and the residue was washed with EA twice to give 3 (12.5 g, 55.2 mmol) ( ref. for intermediate 3 Bioorganic & Médicinal Chemistry Letters, 1996, Vol. 6, No. 4, pp. 373-378,) which was used directly for the next step. ESI-LCMS: m/z 228 [M+H]+.
222
Préparation of (4): To a solution of 3 (12.5 g, 55.2 mmol) in pyridine (125 mL) and added DMAP (1.3 g, 11.0 mmol), TrtCl (30.7 g, 110.5 mmol). The mixture was stirred at r.t. for 24 h. TLC showed 3 was consumed completely. H2O was added to the mixture. Then filtered and the solution diluted with EA. The organic layer was washed with NaHCO3 and brine. The solvent was concentrated under reduced pressure and then added ACN, filtered to give 4a (17.0 g, 35.4 mmol, 64% yield) as a white solid.
To a solution of 4a (17.0 g, 35.4 mmol) in DMF (200 mL), collidine (5.2 g, 43.5 mmol), TrCl (13.1 g, 47.1 mmol) were added after 2h and then again after 3h TrCl (13.1 g, 47.1 mmol), AgNO3 (8.0 g, 47.1 mmol). The mixture was stirred at 25°C for 24 h. TLC showed 4a was consumed completely. Then filtered and the solution diluted with EA. The organic layer was washed with NaHCO3 and brine. The solvent was concentrated under reduced pressure and then added ACN, filtered to get 4 (14.2 g, 19.5 mmol, 54% yield) as a white solid. ESI-LCMS: m/z 712 [M+H]+ ; 'H-NMR (400 MHz, DMSO-76): δ 7.83 (d, J= 8 Hz, 2H), 7.42-7.20 (m, 30H), 6.18 (d, J= 7 Hz, 1H), 6.09 (d, 7= 8 Hz, 2H), 5.60 (d, 7= 7 Hz, 1H), 4.22 (m, 1H), 3.90 (d, 7= 5 Hz, 1H), 2.85 (d, 7= 10 Hz, 1H), 2.76 (s, 1H), 2.55-2.50 (dd, 1H).
Préparation of (5): To a solution of 4 (14.2 g, 19.9 mmol) in DCM (150 mL), DMAP (2.4 g, 19.9 mmol), TEA (4.0 g, 39.9 mmol, 5.6 mL) were added. Then cool down to 0°C, TfCl (6.7 g, 39.9 mmol) dissolved in DCM (150 mL) were dropped. The mixture was stirred at 25°C for 1 h. TLC showed 4 was consumed completely. Then filtered and the solution diluted with EA. The organic layer was washed with NaHCO3 and brine. The solvent was concentrated under reduced pressure to get 5 (16.8 g, 19.9 mmol) as a brown solid. ESI-LCMS: m/z 844 [M+H]+.
Préparation of (6): To a solution of 5 (16.8 g, 19.9 mmol) in DMF (200 mL), KOAc (9.7 g, 99.6 mmol) were added, The mixture was stirred at 25°C for 14 h and 50°C for 3 h, TLC showed 5 was consumed completely. Then filtered and the solution diluted with EA. The organic layer was washed with H2O and brine. The solvent was concentrated under reduced pressure to get 6a (15.0 g, 18.9 mmol, 90% yield) as a brown solid. To a solution of 6a (15.0 g, 19.9 mmol) in 30% CH3NH2/MeOH solution (100 mL) were added. The mixture was stirred at 25°C for 2 h, TLC showed 6a was consumed completely. Then the solvent was concentrated under reduced pressure and the residue was purified by cc (0-5% MeOH in DCM) to give 6 (11.6 g, 16.3 mmol, 82% yield) as a yellow solid. ESI-LCMS: m/z 712 [M+H]+; 'H-NMR (400 MHz, DMSO-î/ô): δ 7.59 (d, J= 8 Hz, 2H), 7.37-7.22 (m, 30H), 6.01 (d, 7= 8 Hz, 2H), 5.84 (d, 7= 3 Hz, 1H), 5.42 (d, 7= 4 Hz, 1H), 3.78-3.70 (m, 3H), 3.10 (t, 7= 9 Hz, 1H), 2.53 (d, 7= 4 Hz, 6H), 1.77 (s, 6H).
223
Préparation of (7): To a solution of 6 (11.6 g, 16.32 mmol) in DCM (200 mL), DAST (7.9 g, 48.9 mmol)were added at 0°C, The mixture was stirred at 25°C for 16 h, TLC showed 6 was consumed completely. Then the solution was diluted with EA, washed with NaHCCh twice, The solvent was concentrated under reduced pressure the residue purified by Flash-Prep-HPLC with the following conditions(IntelFlash-l): Column, Cl8 silica gel; mobile phase, CH3CN/H2O (0.5% NH4HCO3) =1/1 increasing to CH3CN/H2O (0.5% NH4HCO3)=l/0 within 25 min, the eluted product was collected at CH3CN/ H2O (0.5% NH4HCO3) =4/1; Detector, UV 254 nm. This resulted in to give 7 (11.6 g, 13.8 mmol, 84 % yield) as a white solid. ESI-LCMS: m/z 714 [M+H]+ .
Préparation of (8): To a solution of 7 (11.6 g, 16.2 mmol) in DCM (100 mL) was added TFA (10 mL). The mixture was stirred at 20°C for 1 h. TLC showed 7 was consumed completely. Then the solution was concentrated under reduced pressure the residue was purified by silica gel column (0~20% MeOH in DCM) and Flash-Prep-HPLC with the following conditions(IntelFlash-l): Column, C18 silica gel; mobile phase, CH3CN/H2O (0.5% NH4HCO3) =0/1 increasing to CH3CN/H2O (0.5% NH4HCO3)=l/3 within 25 min, the eluted product was collected at CH3CN/ H2O (0.5% NH4HCO3) =0/1; Detector, UV 254 nm. This resulted in to give 9 (1.7 g, 7.2 mmol, 45% yield) as a white solid. ESI-LCMS: m/z 229.9 [M+H]+; 'H-NMR (400 MHz, DMSO-76): δ 7.91 (d, J= 8 Hz, 2H), 6.14 (d, J= 8 Hz, 2H), 5.81-5.76 (m, 2H), 5.28 (t, J= 5 Hz, 1H), 5.13-4.97 (t, J= 4 Hz, 1H), 4.23 (m, 1H), 3.97 (m, 1H), 3.74-3.58 (m, 2H); 19F-NMR (376 MHz, DMSO-c/ô): δ -206.09.
Préparation of (9): To a solution of 8 (1.4 g, 6.1 mmol) in pyridine (14 mL) was added DMTrCl (2.5 g, 7.3 mmol) at 20°C. The mixture was stirred at 20°C for 1 h. TLC showed 8 was consumed completely. Water was added to the reaction. The product was extracted with EA, The organic layer was washed with NaHCCh and brine. Then the solution was concentrated under reduced pressure and the residue was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, C18 silica gel; mobile phase, CH3CN/H2O (0.5% NH4HCO3) = 1/3 increasing to CH3CN/H2O (0.5% NH4HCO3) = 4/1 within 25 min, the eluted product was collected at CH3CN/ H2O (0.5% NH4HCO3) = 1/1; Detector, UV 254 nm. This resulted in to give 9 (2.5 g, 4.6 mmol, 76 yield) as a white solid. ESI-LCMS: m/z 532.2 [M+H]+; 'H-NMR (400 MHz, DMSO-î/ô): δ 7.87-7.84 (m, 2H), 7.40-7.22 (m, 9H), 6.91-6.87(m, 4H), 5.98-5.95 (m, 2H), 5.88-5.77 (m, 2H), 5.16-5.02 (m, 1H), 4.42 (m, 1H), 4.05 (m, 1H), 3.74 (s, 6H), 3.35 (m, 2H); 19F-NMR (376 MHz, DMSO-J6): δ -202.32.
Préparation of Example 42 monomer: To a solution of 9 (2.2 g, 4.1 mmol) in DCM (20 mL) was added DCI (415 mg, 3.5 mmol) and CEP (1.5 g, 4.9 mmol) under N2 pro. The mixture
224 was stirred at 20°C for 0.5 h. TLC showed 9 was consumed completely. The product was extracted with DCM, The organic layer was washed with H2O and brine. Then the solution was concentrated under reduced pressure and the residue was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, Cl8 silica gel; mobile phase, CH3CN/H2O (0.5% NH4HCO3) = 1/3 increasing to CH3CN/H2O (0.5% NH4HCO3) = 1/0 within 25 min, the eluted product was collected at CH3CN/ H2O (0.5% NH4HCO3) = 1/0; Detector, UV 254 nm. This resulted in to give Example 42 monomer (2.6 g, 3.5 mmol, 85% yield) as a white solid. ESILCMS: m/z 732.2 [M+H]+; ‘H-NMR (400 MHz, DMSO-J6): δ 7.87-7.84 (m, 2H), 7.40-7.22 (m, 9H), 6.91-6.87(m, 4H), 5.98-5.95 (m, 2H), 5.90-5.88 (m, 1H), 5.30-5.17 (m, 1H), 4.62 (m, 1H), 4.19 (m, 1H), 3.78-3.73 (m, 7H), 3.62-3.35 (m, 5H), 2.78 (t, J= 5 Hz, 1H), 2.63 (t, J= 6 Hz, 1H),1.14-0.96 (m, 12H); 19F-NMR (376 MHz, DMSO-J6): δ -200.77, 200.80, 201.62, 201.64. 31P-NMR (162 MHz, DMSO-îZ6): δ 150.31, 150.24, 149.66, 149.60.
Example 43. Synthesis of End Cap Monomer
EDCI; TFA Pyridine DMSO
OPOM
MOPO-p=O 9 Dy ,ΟΡΟΜ D Q' OPOM
O
MOPO-p' D ΜΟΡΟ 11
TBSO' bcfè .0.
O
HCOOH
K,CO3
THF/D2O
CEP[N(iPr)2]2; DCI DCM
Example 43 monomer
Scheme-23
Préparation of (8): To a stirred solution of 7 (13.4 g, 35.5 mmol, Scheme 5) in DMSO (135 mL) were added EDCI (6.3 g, 32.9 mmol) and pyridine (0.9g, 10.9 mmol), TFA (0.6 g, 5.5 mmol) at r.t. And the reaction mixture was stirred at r.t for 2 h. LCMS showed 7 consumed completely. The reaction was quenched with water and the product was extracted with EA (1800 mL). The organic phase was washed by brine, dried over Na2SO4, The organic phase was evaporated to dryness under reduced pressure to give a residue 8 (13.2 g, 35.3 mmol, 99.3% yield). Which was used directly to next step. ESI-LCMS: m/z =375 [M+H2O]+
Préparation of (10): A solution of 8 (13.2 g, 35.3 mmol), 9 (26.8 g, 42.3 mmol, Scheme 18 ) and K2CO3 (19.5 g, 141.0 mmol) in dry THF (160 mL) and D2O (53 mL) was stirred at 225
r.t. 17 h. LCMS showed most of 8 was consumed. The product was extracted with EA (2500 mL) and the organic layer was washed with brine and dried over Na2SÛ4. Then the organic layer was concentrated to give a residue which was purified by c.c. (PE: EA = 10:1 ~ 1:2) to give product 10 (8.1 g, 11.8mmol, 33.4% yield) as a white solid. ESI-LCMS m/z = 682 [M+H]+; ’HNMR (400 MHz, DMSO-dô): δ 11.42 (s, 1H), 7.69-7.71 (d, 7=8.1 Hz, 1H), 5.78-5.79 (d, 7=3.7 Hz, 1H), 5.65-5.67 (m, 1H), 5.59-5.63 (m, 4H), 4.29-4.35 (m, 2H), 3.97-3.99 (m, 1H), 1.15 (s, 18H), 0.87 (s, 9H), 0.07-0.08 (d, 7=5.1 Hz, 6H). 31P-NMR (162 MHz, DMS O-76) δ 16.62.
Préparation of (11): To a round-bottom flask was added 10 (7.7 g, 11.1 mmol) in a mixture of HCOOH (80 mL) and H2O (80 mL). The reaction mixture was stirred at 40°C for 3 h. LCMS showed the 10 was consumed completely. The reaction mixture was adjusted the pH = 7.0 with con.NH3.H2O (100 mL). Then the mixture was extracted with DCM (100 mL*3). The combined DCM layer was dried over Na2SÜ4. Fïltered and filtrate was concentrated to give crude which was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, Cl 8 silica gel; mobile phase, CH3CN/H2O (0.5% NH4HCO3) = 1/2 increasing to CH3CN/ H2O (0.5% NH4HCO3) = 1/1 within 20 min, the eluted product was collected at CH3CN/ H2O (0.5% NH4HCO3) = 1/0; Detector, UV 254 nm. To give product 11 (5.5 g, 9.6 mmol, 86.1% yield) as a white solid. ESI-LCMS m/z = 568 [M+H]+; ‘H-NMR (400 MHz,DMSO-d6): δ 11.42 (s, 1H, exchanged with D2O), 7.62-7.64 (d, 7=8.1, 1H), 5.81-5.82 (d, 7=4.3, 1H), 5.58-5.66 (m, 5H), 5.52-5.53 (d, 7=6.6, 1H), 4.34-4.37 (m, 1H), 4.09-4.13 (m, 1H), 3.94-3.96 (t, 7=9.7, 1H), 1.15 (s, 18H), 0 (s, 1H). 31P NMR (162 MHz, DMSO-76) δ 17.16.
Préparation of Example 43 monomer: To a solution of 11 (5.3 g, 9.3 mmol) in DCM (40 mL) was added the DCI (1.1 g, 7.9 mmol), then CEP[N(ipr)2]2 (3.4 g, 11.2 mmol) was added. The mixture was stirred at r.t. for 1 h. LCMS showed 11 consumed completely. The reaction mixture was washed with H2O (50 mL*2) and brine (50 mL*l). Dried over Na2SC>4 and concentrated to give crude which was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, Cl 8 silica gel; mobile phase, CH3CN/H2O (0.5% NH4HCO3) = 1/3 increasing to CH3CN/ H2O (0.5% NH4HCO3) = 1/0 within 20 min, the eluted product was collected at CH3CN/ H2O (0.5% NH4HCO3) = 1/0; Detector, UV 254 nm. The product was concentrated to give Example 43 monomer (6.2 g, 8.0 mmol, 85.6% yield) as a white solid. ESILCMS m/z = 768 [M+H]+; ‘H-NMR (400 MHz, DMSO-76): δ 11.43 (s, 1H), 7.68-7.71 (m, 1H), 5.79-5.81 (m, 1H), 5.58-5.67 (m, 5H), 4.34-4.56 (m, 2H), 4.14-4.17 (m, 1H), 3.54-3.85 (m, 4H), 2.78-2.81 (m, 2H), 1.13-1.17 (m, 30H). 31P-NMR (162 MHz, DMSO-d6): δ 149.66, 149.16, 16.84, 16.56.
226
Example 44. Synthesis of Monomer
Example 44 monomer
Scheme-24
Préparation of (2): To a solution of 1 (20.0 g, 66.4 mmol) in dry DMF (400 mL) was added sodium hydride (1.9 g, 79.7 mmol) for 30 min, then was added CD3I (9.1 g, 79.7 mmol) in dry DCM (40 mL) at -20°C for 5.5 hr. LCMS showed the reaction was consumed. The mixture was filtered and the clear solution was evaporated to dryness and was evaporated with CH3OH. The crude was purified by silica gel column (SiCh, DCM/MeOH = 50:1-10:1). This resulted in to give the product 2 (7.5 g, 23.5 mmol, 35.5% yield) as a solid. ESI-LCMS: m/z 319 [M+H]+ ; ‘H-NMR (400 MHz, DMSO-J3): δ = 8.38 (m, 1H), 6.97 (m, 2H), 5.93-5.81 (m, 1H), 5.27-5.26 (d, J=4Hz, 1H), 5.13-5.11 (m, 1H), 4.39-4.31 (m, 1H), 4.31-4.25 (m, 1 H), 3.96-3.94 (m, 1H), 3.66-3.63 (m, 1H), 3.63-3.56 (m, 1H).
Préparation of (3): To a solution of 2 (7.5 g, 23.5 mmol) in dry DMF (75 mL) was added Imidazole (5.6 g, 82.3 mmol) and TBS.C1 (8.9 g, 58.8 mmol). The mixture was stirred at r.t. over 227 night. LCMS showed 2 was consumed completely. The reaction was quenched with water (300 mL). The product was extracted into ethyl acetate (100 mL). The organic layer was washed with brine and dried over anhydrous Na2SO4. The solvent was removed to give the cured 3 (9.8 g) as a solid witch used for the next step. ESI-LCMS: m/z 547 [M+H]+ .
Préparation of (4): To a solution of 3 (9.8 g) in THF (40 mL) was added TFA (10 mL) and water (10 mL) at 0°C. The reaction mixture was stirred at 0°C for 5 h. LC-MS showed 3 was consumed completely. Con. NH4OH was added to the mixture at 0°C to quench the reaction until the pH = 7.5. The product was extracted into ethyl acetate (200 mL). The organic layer was washed with brine and dried over anhydrous Na2SO4. The solvent was removed to give the cured 4 (8.4 g) as a solid witch used for the next step. ESI-LCMS: m/z 433 [M+H]+ .
Préparation of (5): To a solution of 4 (8.4 g) in DCM/H2O = 2:1 (84 mL) was added DAIB (18.8 g, 58.4 mmol) and TEMPO (0.87 g, 5.8 mmol). The reaction mixture was stirred at 40°C for 2 h. LCMS showed 4 was consumed. The mixture was diluted with DCM and water was added. The product was extracted with DCM. The organic layer was washed with brine and dried over anhydrous Na2SO4. The solution was then concentrated under reduced pressure. This resulted in to give 5 (14.4 g) as a white solid. ESI-LCMS: m/z 447 [M+H]+ .
Préparation of (6): To a solution of 5 (14.4 g) in toluene (90 mL) and methanol (60 mL) was added 2M TMSCHN2 (8.9 g, 78.1 mmol) till the yellow color not disappear at r.t. for 10 min. LCMS showed 5 was consumed. The crude was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, Cl8 silica gel; mobile phase, CH3CN/H2O (0.5% NH4HCO3) =1/1 increasing to CH3CN/H2O (0.5% NH4HCO3) = 1/0 within 25 min, the eluted product was collected at CH3CN/ H2O (0.5% NH4HCO3) =65/35 Detector, UV 254 nm. This resulted in to give the product 6 (3.5 g, 7.6 mmol, 32.3% yield over three steps, 70% purity) as a white solid. ESI-LCMS: m/z 461 [M+H]+ .
Préparation of (7): To the solution of 6 (3.5 g, 7.6 mmol) in dry THF/MeOD/D2O = 10/2/1 (45 mL) was added NaBÜ4 (0.96 g, 22.8 mmol). And the reaction mixture was stirred at r.t for 2.5 hr. Aller completion of reaction, the resulting mixture was added CH3COOD to pH = 7, aller addition of water, the resulting mixture was extracted with EA (100 mL). The combined organic layer was washed with water and brine, dried over Na2SO4, and concentrated to give 7 (3.3 g) which was used for the next step. ESI-LCMS: m/z 435 [M+H]+ .
Préparation of (8): To a solution of 7 (3.3 g) in dry DCM (30 mL) was added pyridine (5.9 g, 74.5 mmol) and iBuCl (2.4 g, 22.4 mmol) in DCM (6 mL) under ice bath. The reaction mixture was stirred at 0°C for 2.5 hr. LCMS showed 7 was consumed. The mixture was diluted with EA and water was added. The product was extracted with EA. The crude was purified by
228
Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, Cl8 silica gel; mobile phase, CH3CN/H2O (0.5% NH4HCO3) = 1/1 increasing to CH3CN/H2O (0.5% NH4HCO3) = 1/0 within 25 min, the eluted product was collected at CH3CN/H2O (0.5% NH4HCO3) = 87/13; Detector, UV 254 nm. This resulted in to give the crude 8 (1.6 g, 2.8 mmol, 36.8% yield over two steps) as a white solid. ESI-LCMS: m/z 575 [M+H]+ .
Préparation of (9): To a solution of 8 (1.6 g, 2.8 mmol,) in H2O/dioxane =1:1 (30 ml) was added K2CÜ3 (772.8 mg, 5.6 mmol) and DABCO (739.2 mg, 2.9 mmol). The reaction mixture was stirred at 50°C for 3 hr. LCMS showed 8 was consumed. The mixture was diluted with EA and water was added. The product was extracted with EA. The combined organic layer was washed with water and brine, dried over Na2SO4, and concentrated to give 9 (1.8 g) which was used for the next step. ESI-LCMS: m/z 557 [M+H]+ .
Préparation of (10): To a solution of 9 (1.8 g) in pyridine (20 mL) and was added 2M NaOH (MeOH/H2O = 4/1) (5 mL) at 0°C for 1 h. LCMS showed 9 was consumed. The mixture was added saturated NH4CI till pH = 7.5. The mixture was diluted with water and EA. The organic layer was washed with brine and dried over Na2SC>4 and concentrated to give the crude. This resulted in to give the product 10(1.5 g) as a white solid which was used for the next step. ESI-LCMS: m/z 487 [M+H]+ .
Préparation of (11): To a stirred solution of 10 (1.5 g) in pyridine (20 mL) were added DMTrCl (1.1 g, 3 mmol) at r.t. And the reaction mixture was stirred at r.t for 2.5 hr. With icebath cooling, the reaction was quenched with water and the product was extracted into EA. The organic layer was washed with brine and dried over Na2SÜ4 and concentrated to give the crude. The crude was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, C18 silica gel; mobile phase, CH3CN/H2O (0.5% NH4HCO3) = 1/1 increasing to CH3CN/H2O (0.5% NH4HCO3) = 1/0 within 25 min, the eluted product was collected at CH3CN/ H2O (0.5% NH4HCO3) = 7/3 Detector, UV 254 nm. This resulted in to give the product 11 (1.9 g, 2.4 mmol, 85.7% yield over two steps) as a white solid. ESI-LCMS: m/z 789.3 [M+H]+; *HNMR (400 MHz, DMSO-76): δ 12.10 (m, 1H), 11.63 (m, 1H), 8.20 (m, 1H), 7.35 -7.33 (m, 2H), 7.29-7.19 (m, 7H), 6.86-6.83 (m, 4H), 5.89-5.88 (d, J= 4 Hz, 1H), 4.40-4.28 (m, 2H), 3.72 (m, 6H), 2.81-2.76 (m, 1H), 1.13-1.11 (m, 6H), 0.80 (m, 9H), 0.05—0.01(m, 7H).
Préparation of (12): To a solution of 11 (1.9 g, 2.4 mmol) in THF (20 mL) was added 1 M TBAF solution (3 mL). The reaction mixture was stirred at r.t. for 1.5 h. LCMS showed 11 was consumed completely. Water (100 mL) was added. The product was extracted with EA (50 mL) and the organic layer was washed with brine and dried over Na2SO4. Then the organic layer was concentrated to give a residue which was purified by Flash-Prep-HPLC with the following
229 conditions (IntelFlash-1): Column, Cl8 silica gel; mobile phase, CH3CN/H2O (0.5% NH4HCO3) = 2/3 increasing to CH3CN/H2O (0.5% NH4HCO3) = 1/0 within 20 min, the eluted product was collected at CH3CN/ H2O (0.5% NH4HCO3) = 58/42; Detector, UV 254 nm. This resulted in to give 12 (1.5 g, 2.2 mmol, 91.6% yield) as a white solid. ESI-LCMS: m/z 675.3 [M+H]+; ’HNMR (400 MHz, DMSO-J6): δ 12.09 (m, 1H), 11.60 (m, 1H), 8.14 (m, 1H), 7.35 -7.27 (m, 2H), 7.25-7.20 (m, 7H), 6.85-6.80 (m, 4H), 5.96-5.94 (d, J= 8 Hz, 1H), 5.26-5.24 (m, 1H), 4.35-4.28 (m, 2H), 3.72 (m, 6H), 3.32 (m, 1H), 2.79-2.72 (m, 1H), 1.13-1.11 (m, 6H).
Préparation of Example 44 monomer: To a suspension of 11 (1.5 g, 2.2 mmol) in DCM (15 mL) was added DCI (220.8 mg, 1.9 mmol) and CEP[N(iPr)2]2 (795.7 mg, 2.6 mmol) under N2 pro. The mixture was stirred at r.t. for 2 h. LCMS showed 11 was consumed completely. The solution was washed with water twice and washed with brine and dried over Na2SO4. Then concentrated to give a residue which was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, C18 silica gel; mobile phase, CH3CN/H2O (0.5% NH4HCO3) = 1/1 increasing to CH3CN/H2O (0.5% NH4HCO3) = 1/0 within 20 min, the eluted product was collected at CH3CN/ H2O (0.5% NH4HCO3) = 4/1 ; Detector, UV 254 nm. This resulted in to give Example 44 monomer (1.6 g, 1.8 mmol, 83% yield) as a white solid. ESI-LCMS: m/z 875 [M+H]+; ‘H-NMR (400 MHz, DMSO-J6): δ 12.12 (m, 1H), 11.60 (m, 1H), 8.15 (m, 1H), 7.37 7.29 (m, 2H), 7.27-7.20 (m, 7H), 6.86-6.81 (m, 4H), 5.94-5.88 (m, 1H), 4.54-4.51 (m, 2H), 4.214.20 (m, 1H), 3.73-3.54 (m, 10H), 2.80-2.75 (m, 1H), 2.61-2.58 (m, 1 H), 1.19-1.11 (m, 19H). 31P-NMR (162 MHz, DMSO-î/6): δ= 149.77, 149.71.
Example 45. Synthesis of Monomer
230
2
TrtCl, AgNO3 collidine, DMF
TfCl, TEA
DMAP, DCM
Scheme-25
6%
DCA7DCM
Example 45 monomer
Préparation of (2): To a solution of 1 (50.0 g, 99.2 mmol) and la (11.3 g, 119.0 mmol) in ACN (500.0 mL). Then added BSA (53.2 g, 218.0 mmol) under N2 Pro. The mixture was stirred at 50°C for 1 h until the solution was clear. Then cool down to 0°C and dropped TMSOTf (26.4 g, 119.0 mmol).The mixture was stirred at 75°C for 1 h, TLC showed 1 was consumed completely. The reaction was quenched by sodium bicarbonate solution at 0°C, then the solution was diluted with EA, washed with H2O twice. The solvent was concentrated under reduced pressure and the crude 2 (60.1 g) was used for next step. ESI-LCMS: m/z 540.2
[M+H]+.
Préparation of (3): To a solution of 2 (60.1 g) in CHaNEb/ethanol (500.0 mL). The mixture was stirred at 25°C for 2 h. TLC showed 2 was consumed completely. The solvent was concentrated under reduced pressure and the residue was purified by c.c. (MeOH:DCM = 50:1 ~ 10:1) to give 3 (22.0 g, 96.9 mmol, 97.3% yield over two steps).
ESI-LCMS: m/z 228.0 [M+H]+; ‘H-NMR (400 MHz, DMSO-d6): δ 8.01-7.98 (m, 1H), 7.43-7.38 (m, 1H), 6.37-6.35 (m, 1H), 6.27-6.23 (m, 1H), 6.03 (d, J= 3.5 Hz, 1H), 5.39 (d, J= 4.2 Hz, 1H), 5.11 (t, J=5.1 Hz, 1H), 5.03 (d, .7=5.1 Hz, 1H), 3.98-3.95 (m, 2H), 3.91-3.88 (m, 1H), 3.74-3.57 (m, 2H).
231
Préparation of (4): To a solution of 3 (22.0 g, 96.9 mmol) in pyridine (250.0 mL), TrtCl (30.7 g, 110.5 mmol) was added. The mixture was stirred at 25°C for 24 h. TLC showed 3 was consumed completely, H2O was added to the mixture. Then filtered and the filtrate diluted with EA, the organic layer was washed with NaHCCh and brine. The solvent was concentrated under reduced pressure and then purified by c.c. (PE/EA = 5:1 ~ 0:1) to give 4 (38.8 g, 82.5 mmol, 85.1% yield) as a white solid. ESI-LCMS: m/z 470.1 [M+H]+.
Préparation of (5): To a solution of 4 (38.8 g, 82.5 mmol) in DMF (500.0 mL), collidine (10.0 g, 107.3 mmol), TrtCl (27.6 g, 99.1 mmol) were added followed by AgNCh (18.0 g, 105.1 mmol). The mixture was stirred at 25°C for 4 h. TLC showed 4 was consumed completely. Then filtered and the filtrate diluted with EA. The organic layer was washed with NaHCCh and brine. The solvent was concentrated under reduced pressure and then purified by c.c. (PE/EA = 5:1 ~ 1:1) to give a mixture of 5 (52.3 g, 73.5 mmol, 86.3% yield) as white solid. ESI-LCMS: m/z 711.1 [M+H]+.
Préparation of (6): To a solution of 5 (52.3 g, 73.5 mmol) in DCM (500.0 mL), DMAP (8.9 g, 73.5 mmol), TEA (14.9 g, 147.3 mmol, 20.6 mL) were added, cool down to 0°C, TfCl (16.1 g, 95.6 mmol) dissolved in DCM (100.0 mL) were dropped. The mixture was stirred at 25°C for 1 h. TLC showed 5 was consumed completely. Then filtered and the solution diluted with EA. The organic layer was washed with NaHCCh and brine. The solvent was concentrated under reduced pressure to get crude 6 (60.2 g) as a brown solid. ESI-LCMS: m/z 844.2 [M+H]+.
Préparation of (7): To a solution of 6 (60.2 g) in DMF (500.0 mL), KOAc (36.1 g, 367.8 mmol) were added, The mixture was stirred at 25°C for 14 h and 50°C for 3 h, TLC showed 6 was consumed completely. Then filtered and the solution diluted with EA. The organic layer was washed with H2O and brine. The solvent was concentrated under reduced pressure, residue was purified by c.c. (PE/EA = 5:1-1:1) to give 7 (28.0 g, 39.3 mmol, 53.5% yield) as yellow solid. ESI-LCMS: m/z 710.2 [M-H]'; Ή-NMR (400 MHz, DMSO-î/6): δ 7.37-7.25 (m, 33H), 6.34-6.31 (m, 2H), 6.13-6.10 (m, 1H), 5.08 (d, J=4.2 Hz, 1H), 3.99 (d, .7=7.6 Hz, 1H), 3.74 (s, 1H), 3.12 (t, J= 9.2 Hz, 1H), 2.72-2.69 (m, 1H).
Préparation of (8): To a solution of 7 (28.0 g, 39.3 mmol) in DCM (300.0 mL), DAST (31.6 g, 196.6 mmol) was added at 0°C, the mixture was stirred at 25°C for 16 h, TLC showed 7 was consumed completely. Then the solution was diluted with EA, washed with NaHCCL twice, the solvent was removed under reduced pressure, residue was purified by c.c. (PE/EA = 5:1 ~ 3:1) to give 8 (5.0 g, 7.0 mmol, 17.8% yield) as a white solid. ESI-LCMS: m/z 748.2 [M+2NH4]+; ‘H-NMR (400 MHz, DMSO-î/6): δ 7.57-7.18 (m, 35H), 6.30 (d, J= 8.8 Hz, 1H),
232
6.00 (d, J= 19.5 Hz, 1H), 5.92-5.88 (m, 1H), 4.22-4.17 (m, 2H), 3.94 (s, 0.5H), 3.80 (s, 0.5H), 3.35-3.31 (m, 1H), 3.14-3.10 (m, 1H); 19F-NMR(376 MHz, DMSO-î76): δ-193.54.
Préparation of (9): To a solution of 8 (5.0 g, 7.0 mmol) in DCM (60.0 mL) was added DCA (3.6 mL) and TES (15.0 mL). The mixture was stirred at 20°C for 1 h, TLC showed 8 was consumed completely. Then the solution was concentrated under reduced pressure, the residue was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, Cl8 silica gel; mobile phase, CH3CN/H2O (0.5% NH4HCO3) =0/1 increasing to CH3CN/H2O (0.5% NH4HCO3) = 1/3 within 25 min, the eluted product was collected at CH3CN/ H2O (0.5% NH4HCO3) =0/1; Detector, UV 254 nm. This resulted in to give 9 (1.6 g, 6.9 mmol, 98.5% yield) as a white solid. ESI-LCMS: m/z 229.9 [M+H]+; ‘H-NMR (400 MHz, DMSO-î/6): δ 8.06-8.04 (m, 1H), 7.48-7.43 (m, 1H), 6.39 (d, J= 9.0 Hz, 1H), 6.31-6.27 (m, 1H), 6.16-6.11 (m, 1H), 5.63 (s, 1H), 5.26 (s, 1H), 4.95-4.81 (m, 1H), 4.20-411 (m, 1H), 3.95 (d, J= 8.2 Hz, 1H), 3.84 (d, J =12.4 Hz, 1H), 3.64 (d, .7=12.1 Hz, 1H); 19F-NMR (376 MHz, DMSO-J6): δ -201.00.
Préparation of (10): To a solution of 9 (1.6 g, 6.9 mmol) in pyridine (20.0 mL) was added DMTrCl (3.5 g, 10.5 mmol) at 20°C and stirred for 1 h. TLC showed 9 was consumed completely. Water was added and extracted with EA, the organic layer was washed with NaHCO3 and brine. Then the solution was concentrated under reduced pressure and the residue was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, Cl8 silica gel; mobile phase, CH3CN/H2O (0.5% NH4HCO3) = 1/3 increasing to CH3CN/H2O (0.5% NH4HCO3) =4/1 within 25 min, the eluted product was collected at CH3CN/ H2O (0.5% NH4HCO3) =1/1; Detector, UV 254 nm. This resulted in to give 10 (2.2 g, 4.2 mmol, 60.8% yield) as a white solid. ESI-LCMS: m/z 530.1 [M-H]’; 'H-NMR (400 MHz, DMSO-îZ6): δ 7.937.91 (m, 1H), 7.47-7.23 (m, 10H), 6.91-6.89 (m, 4H), 6.41 (d, .7=8.8 Hz, 1H), 6.13 (d, .7=18.8 Hz, 1H), 6.00-5.96 (m, 1H), 5.68 (d, J= 6.6 Hz, 1H), 5.01 (d, J= 4.2 Hz, 0.5H), 4.88 (d, J= 4.2 Hz, 0.5H), 4.42-4.31 (m, 1H), 4.10-4.08 (m, 1H), 3.74 (s, 6H),3.40-3.34 (m, 2H); I9F-NMR (376 MHz, DMSO-de): δ -199.49.
Préparation of Example 45 monomer: To a solution of 10 (2.2 g, 4.2 mmol) in DCM (20.0 mL) was added DCI (415 mg, 3.5 mmol) and CEP (1.5 g, 4.9 mmol) under N2 pro. The mixture was stirred at 20°C for 0.5 h. TLC showed 10 was consumed completely. The product was extracted with DCM, the organic layer was washed with H2O and brine. Then the solution was concentrated under reduced pressure and the residue was purified by cc (PE/EA = 5:1 ~ 1:1) and Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, Cl8 silica gel; mobile phase, CH3CN/H2O (0.5% NH4HCO3) =1/3 increasing to CH3CN/H2O (0.5% NH4HCO3)=l/0 within 25 min, the eluted product was collected at CH3CN/ H2O (0.5%
233
NH4HCO3) =1/0; Detector, UV 254 nm. This resulted in to give Example 45 monomer (2.1 g, 3.0 mmol, 73.1% yield) as a white solid. ESI- ESI-LCMS: m/z 732.2 [M+H]+; 'H-NMR (400 MHz, DMSO-î/ô): δ 7.98-7.92 (m, 1H), 7.42-7.24 (m, 10H), 6.91-6.85 (m, 4H), 6.43-6.39 (m, 1H), 6.18-6.11 (m, 1H), 6.01-5.97 (m, 1H), 5.22-5.19 (m, 0.5H), 5.09-5.06 (m, 0.5H), 4.73-4.52 (m, 1H), 4.21-4.19 (m, 1H), 3.79-3.62 (m, 7H), 3.57-3.47 (m, 4H), 3.32-3.28 (m, 1H), 2.75-2.58 (m, 1H), 1.13-0.92 (m, 12H); 19F-NMR (376 MHz, DMSO-76): δ -196.82, -196.84, -197.86, 197.88; 31P-NMR (162 MHz, DMSO-76): δ 149.88, 149.83, 149.39, 149.35.
Example 46. Synthesis of Monomer
Example 46 monomer
Scheme-26
Préparation of (2): To the solution of Bromobenzene (2.1 g, 13.6 mmol) in dry THF (15 mL) was added 1.6 M n-BuLi (7 mL, 11.8 mmol) drop wise at -78°C. The mixture was stirred at -78°C for 0.5 h. Then the 1 (3.0 g, 9.1 mmol,Wang, Guangyi et al, Journal of Médicinal Chemistry, 2016,59(10), 4611-4624) was dissolved in THF (15 mL) and added to the mixture drop wise with keeping at -78°C. Then the reaction mixture was stirred at -78°C for 1 hr. LC-MS showed 1 was consumed completely. Then the solution was added to saturated aq. NH4CI and the resulting mixture was extracted with EA. The combined organic layer was washed with water and brine, dried over Na2SÜ4, and concentrated under reduced pressure to give a residue which was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, Cl8 silica gel; mobile phase, CH3CN/H2O (0.5% NH4HCO3) = 2/3 increasing to CH3CN/H2O (0.5% NH4HCO3) = 4/1 within 25 min, the eluted product was collected at CH3CN/ H2O (0.5% NH4HCO3) = 3/2; Detector, UV 254 nm. This resulted in to give 2 (3.0 g, 7.3 mmol, 80.0%) as a white solid. ESI-LCMS: m/z 391 [M-OH]’.
234
Préparation of (3): To the solution of 2 (4.0 g, 9.8 mmol) in DCM (40 mL) was added TES (1.9 g, 11.7 mmol) at -78°C, and the mixture was added BFa.OEt? (2.1 g, 14.7 mmol) drop wise at -78°C. The mixture was stirred at -40°C for 1 hr. LC-MS showed 2 was consumed completely. Then the solution was added to saturated aq. NaHCOs and the resulting mixture was extracted with DCM. The combined organic layer was washed with water and brine, dried over Na2SÛ4, and concentrated under reduced pressure to give a residue which was purified by FlashPrep-HPLC with the following conditions (IntelFlash-1): Column, Cl 8 silica gel; mobile phase, CH3CN/H2O (0.5% NH4HCO3) = 2/3 increasing to CH3CN/H2O (0.5% NH4HCO3) = 4/1 within 25 min, the eluted product was collected at CH3CN/ H2O (0.5% NH4HCO3) = 7/3; Detector, UV 254 nm. This resulted in to give 3 (3.1 g, 5.3 mmol, 54.0%) as a water clear oil. ESI-LCMS: m/z 410 [M+H2O]+; ’H-NMR (400 MHz, CDCI3: δ 7.48-7.25 (m, 15H), 5.24-5.13 (m, 1H), 4.93-4.74 (m, 1H), 4.74-4.46 (m, 4H), 4.37-4.25 (m, 1H), 4.19-4.05 (m, 1H), 4.00-3.80 (m, 1H), 3.77-3.63 (m, 1H). 19F-NMR (376 MHz, CDCI3): δ -196.84.
Préparation of (4): To the solution of 3 (2.1 g, 5.3 mmol) in dry DCM (20 mL) was added 1 M BCI3 (25 mL, 25.5 mmol) drop wise at -78°C, and the reaction mixture was stirred at -78°C for 0.5 hr. LC-MS showed 3 was consumed completely. After completion of reaction, the resulting mixture was poured into water (50 mL). The solution was extracted with DCM and the combined organic layer was concentrated under reduced pressure to give a crude. The crude in MeOH (4 mL) was added 1 M NaOH (15 mL), and the mixture was stirred at r.t for 5—10 min. The mixture was extracted with EA. The combined organic layer was washed with brine, dried over Na2SÜ4, and concentrated under reduced pressure to give a residue which was purified by silica gel column chromatography (eluent, DCM: MeOH = 40:1-15:1) to give 4 (1.0 g, 4.7 mmol, 88.6%) as a water clear oil. ESI-LCMS: m/z 211 [M-H]’; ‘H-NMR (400 MHz, DMSOd6): δ 7.58-7.19 (m, 5H), 5.41 (d, J= 6.1 Hz, 1H), 5.09-5.95 (m, 1H), 5.95-4.84 (m, 1H), 4.824.59 (m, 1H), 4.14-3.94 (m, 1H), 3.89-3.80 (m, 1H), 3.78-3.67 (m, 1H), 3.65-3.53 (m, 1H). 19FNMR (376 MHz, DMSO-îZ6): δ -196.46.
Préparation of (5): To a solution of 4 (1.0 g, 4.7 mmol) in Pyridine (10 mL) was added DMTrCl (2.0 g, 5.7 mmol). The reaction mixture was stirred at r.t. for 2 hr. LCMS showed 4 was consumed and water (100 mL) was added. The product was extracted with EA (100 mL) and the organic layer was washed with brine and dried over Na2SÛ4 and concentrated to give the crude. The crude was further purified by Flash-Prep-HPLC with the following conditions (IntelFlash1): Column, Cl 8 silica gel; mobile phase, CH3CN/H2O (0.5% NH4HCO3) = 1/1 increasing to CH3CN/H2O (0.5% NH4HCO3) = 1/0 within 20 min, the eluted product was collected at CH3CN/ H2O (0.5% NH4HCO3) = 9/1; Detector, UV 254 nm. This resulted in to give 5 (2.1 g, 4.1 mmol,
235
87.0%) as a red oil. ESI-LCMS: m/z 513 [M-H]'; ‘H-NMR (400 MHz, DMSO-î/6): δ 7.56-7.16 (m, 14H), 6.94-9.80 (m, 4H), 5.45 (d, J= 6.3 Hz, 1H), 5.21-5.09 (m, 1H), 4.89-4.68 (m, 1H), 4.18-4.03 (m, 2H), 3.74 (s, 6H), 3.33-3.29 (m, 1H), 3.26-3.17 (m, 1H). 19F-NMR (376 MHz, DMSO-î76): δ -194.08.
Préparation of Example 46 monomer: To a suspension of 5 (2.1 g, 4.1 mmol) in DCM (20 mL) was added DCI (410 mg, 3.4 mmol) and CEP[N(iPr)2]2 (1.5 g, 4.9 mmol). The mixture was stirred at r.t. for 1 h. LC-MS showed 5 was consumed completely. The solution was washed with water twice and washed with brine and dried over Na2SO4. Then concentrated to give the crude. The crude was purification by Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, Cl 8 silica gel; mobile phase, CH3CN/H2O (0.5% NH4HCO3) = 1/1 increasing to CH3CN/H2O (0.5% NH4HCO3) = 1/0 within 20 min, the eluted product was collected at CH3CN/ H2O (0.5% NH4HCO3) = 1/0; Detector, UV 254 nm. This resulted in to give Example 46 monomer (2.1 g, 2.9 mmol, 70.0%) as a white solid. ESI-LCMS: m/z 715 [M+H]+; ‘H-NMR (400 MHz, DMSO-îZ6): δ 7.59-7.16 (m, 14H), 6.94-9.80 (m, 4H), 5.26-5.12 (m, 1H), 5.06-4.77 (m, 1H), 4.50-4.20 (m, 1H), 4.20-4.10 (m, 1H), 3.83-3.63 (m, 7H), 3.59-3.37 (m, 4H), 3.25-3.13 (m, 1H), 2.80-2.66 (m, 1H), 2.63-2.53 (m, 1H), 1.18-0.78 (m, 12H). 19F-NMR (376 MHz, DMSO-c/ό): δ -194.40, -194.42, -194.50, -194.53.31P-NMR (162 MHz, DMSO-tZ6): δ 149.38, 149.30, 149.02, 148.98.
Example 47. Deuterated vinyl phosphonate improves potency of siNA
This example investigates whether a deuterated vinyl phosphonate improves potency of siNA in an AAV-HBV mouse. AAV-HBV mice were subcutaneously injected with vehicle, dssiNA-0165 (e.g., siNA without a deuterated vinyl phosphonate), or ds-siNA-0144 (e.g., siNA with a deuterated vinyl phosphonate). For siNA-treated AAV-HBV mice, AAV-HBV mice were subcutaneously injected with a single dose of 5 mg/kg of siNA. As shown in FIG. 11, siNA molécules having 2’-fluoro nucléotides at positions 3, 7-9, 12, and 17 from the 5’ end of the sense strand and 2’-fluoro nucléotides at positions 2 and 14 from the 5’ end of the antisense strand resulted in at least a 0.5-log réduction in HBsAg, with the greatest réduction in HBsAg found in mice treated with the deuterated vinylphopshonate siNA (ds-siNA-0165). Thus, FIG. 11 demonstrates that the presence of a deuterated vinyl phosphonate improves potency of the siNA.
Example 48. Deuterated vinyl phosphonate results in a greater réduction in sérum HBsAg
AAV-HBV mice were subcutaneously injected with vehicle, ds-siNA-0163 (e.g., siNA without a vinyl phosphonate), ds-siNA-0122 (e.g., siNA with a vinyl phosphonate), or ds-siNA0123 (e.g., siNA with a deuterated vinyl phosphonate). For siNA-treated AAV-HBV mice,
236
AAV-HBV mice were subcutaneously injected with a single dose of 5 mg/kg of siNA. As shown in FIG. 12, siNA molécules having 2’-fluoro nucléotides at positions 7, 9-11 from the 5’ end of the sense strand and 2’-fluoro nucléotides at positions 2 and 14 from the 5’ end of the antisense strand resulted in at least a 0.5-log réduction in HBsAg, with the greatest réduction in HBsAg found in mice treated with the deuterated vinylphopshonate siNA (ds-siNA-0165). Thus, FIG. 12 demonstrates that the presence of a deuterated vinyl phosphonate improves potency of the siNA, as compared to the siNA without a vinyl phosphonate and the siNA with the vinyl phosphonate.
Example 49: Synthesis of 5’ End Cap Monomer
Example 49 Monomer Synthesis Scheme
Préparation of (2): 1 (15 g, 58.09 mmol) and fôrt-butyl N-methylsulfonylcarbamate (17.01 g, 87.13 mmol) were dissolved in THF (250 mL), and PPha (30.47 g, 116.18 mmol) was added followed by dropwise addition of DIAD (23.49 g, 116.18 mmol, 22.59 mL) at 0°C. The reaction mixture was stirred at 15°C for 12 h. Upon completion as monitored by TLC (DCM/MeOH=10/l), the reaction mixture was evaporated to give a residue. The residue was purified by flash silica gel chromatography (ISCO®; 120 g SepaFlash® Silica Flash Column, Eluent of 0-20% MeOH/DCM gradient @ 60 mL/min) to give 2 (6.9 g, 24.28% yield) as a white solid. ESI-LCMS: m/z 457.9 [M+Na]+; Ή NMR (400 MHz, CDCI3) δ = 8.64 (br s, 1H), 7.64 (d, J=8.2 Hz, 1H), 5.88 (d, J=1.9 Hz, 1H), 5.80 (dd, J=2.2, 8.2 Hz, 1H), 4.19 - 4.01 (m, 3H), 3.90 237 (dt, J=5.5, 8.2 Hz, 1H), 3.82 - 3.78 (m, 1H), 3.64 (s, 3H), 3.32 (s, 3H), 2.75 (d, J=8.9 Hz, 1H), 1.56 (s, 9H).
Préparation of (3): 2 (6.9 g, 15.85 mmol) was dissolved in MeOH (40 mL), and a solution of HCl/MeOH (4 M, 7.92 mL) was added dropwise. The reaction mixture was stirred at 15°C for 12 h, and then evaporated to give a residue. The residue was purified by flash silica gel chromatography (ISCO®; 40 g SepaFlash® Silica Flash Column, Eluent of 0-10% MeOH/DCM gradient @ 40 mL/min) to give 3 (2.7 g, 50.30% yield) as a white solid. ESI-LCMS: m/z 336.0 [M+H]+; ‘H NMR (400 MHz, CD3CN) δ = 9.20 (br s, 1H), 7.52 (d, J=8.1 Hz, 1H), 5.75 (d, J=3.8 Hz, 1H), 5.64 (dd, J=2.0, 8.1 Hz, 1H), 5.60 - 5.52 (m, 1H), 4.15 - 3.99 (m, 1H), 3.96 - 3.81 (m, 2H), 3.46 (s, 3H), 3.44 - 3.35 (m, 1H), 3.34 - 3.26 (m, 1H), 2.92 (s, 3H).
Préparation of (Example 49 monomer): To a solution of 3 (2.14 g, 6.38 mmol) in DCM (20 mL) was added dropwise 3-bis(diisopropylamino)phosphanyloxypropanenitrile (2.50 g, 8.30 mmol, 2.63 mL) at 0°C, followed by lH-imidazole-4, 5-dicarbonitrile (829 mg, 7.02 mmol), and the mixture was purged under Ar for 3 times. The reaction mixture was stirred at 15 °C for 2 h. Upon completion, the mixture was quenched with 5% NaHCO3 (20 mL), extracted with DCM (20 mL*2), washed with brine (15 mL), dried over NazSCU, filtered, and evaporated to give a residue. The residue was purified by flash silica gel chromatography (ISCO®; 40 g SepaFlash® Silica Flash Column, Eluent of 0-10% (Phase B: i-PrOH/DCM=l/2)/Phase A: DCM with 5% TEA gradient @ 40 mL/min) to give Example 49 monomer (1.73 g, 48.59% yield) as a white solid. ESI-LCMS: m/z 536.3 [M+H]+; ‘H NMR (400 MHz, CD3CN) δ = 7.58 7.48 (m, 1H), 5.83 - 5.78 (m, 1H), 5.71 - 5.64 (m, 1H), 4.40 - 4.29 (m, 1H), 4.19 - 4.07 (m, 1H), 3.98 (td, J=5.3, 13.3 Hz, 1H), 3.90 - 3.78 (m, 2H), 3.73 - 3.59 (m, 3H), 3.41 (d, J=14.8 Hz, 4H), 2.92 (br d, J=7.0 Hz, 3H), 2.73 - 2.63 (m, 2H), 1.23 - 1.11 (m, 12H); 31P NMR (162 MHz, CD3CN) δ = 149.81, 150.37.
Example 50: Synthesis of 5’ End Cap Monomer
NaNj
Hj/Pd/C
238
Cl
TBSO' OCH3 TBSO' bCHj HO' t)CHj
6 7
Example 50 Monomer
Example 50 Monomer Synthesis Scheme
Préparation of (2): To a solution of 1 (10 g, 27.16 mmol) in DMF (23 mL) were added imidazole (3.70 g, 54.33 mmol) and TBSC1 (8.19 g, 54.33 mmol) at 25 °C. The mixture was stirred at 25 °C for 2 hr. Upon completion, the reaction mixture was diluted with Η2Ο (20 mL) and extracted with EA (30 mL * 2). The combined organic layers were washed with brine (20 mL * 2), dried over Na2SO4, filtered and concentrated under reduced pressure to give 2(13 g, 99.2% yield) as a white solid. ESI-LCMS: m/z 482.9 [M+H]+. .
Préparation of (3): To a solution of 2 (35.00 g, 72.56 mmol) in DMF (200 mL) was added NaN3 (14.15 g, 217.67 mmol). The mixture was stirred at 60 °C for 17 h. Upon completion, the reaction mixture was diluted with H2O (200 mL) and extracted with EA (200 mL* 2). The combined organic layers were washed with brine (100 mL * 2), dried over Na2SO4, filtered and concentrated under reduced pressure to give 3 (31.8 g, crude) as a yellow solid. ESILCMS: m/z 398.1 [M+H]+; Ή NMR (400 MHz, DMSO-d6) δ=11.21 (d, J=1.3 Hz, 1H), 7.50 (d, >8.1 Hz, 1H), 5.57 (d, >4.5 Ηζ,ΙΗ), 5.46 (dd, >2.1, 8.0 Hz, 1H), 4.06 (t, >5.2 Hz, 1H), 3.81 - 3.64 (m, 2H), 3.44 - 3.30 (m, 2H), 2.31 -2.25 (m, 3H), 0.65 (s, 9H), -0.13 (s, 6H).
Préparation of (4): To a solution of 3 (7 g, 17.61 mmol) in THF (60 mL) was added Pd/C (2 g) at 25 °C. The reaction mixture was stirred at 25 °C for 3 h under H2 atmosphère (15 PSI). The reaction mixture was filtered, and the filtrate was concentrated to give 4 (5.4 g, 75.11% yield) as a gray solid. ESI-LCMS: m/z 372.1 [M+H]+ ; *H NMR (400 MHz, DMSO-d6) δ =7.93 (d, >8.0 Hz, 1H), 5.81 (d, J=5.5 Hz, 1H), 5.65 (d, >8.3 Ηζ,ΙΗ), 4.28 (t,
239 >4.6 Hz, 1H), 3.88 (t, >5.3 Hz, 1H), 3.74 (q, >4.6 Hz, 1 H), 3.31 (s, 3H), 2.83 - 2.66 (m,2H), 0.88 (s, 9H), 0.09 (s, 6H).
Préparation of (5): To a solution of 4 (3 g, 8.08 mmol) in DCM (30 mL) was added TEA (2.45 g, 24.23 mmol, 3.37 mL) followed by dropwise addition of 3-chloropropane-lsulfonyl chloride (1.50 g, 8.48 mmol, 1.03 mL) at 25 °C. The reaction mixture was stirred at 25 °C for 18 h under N? atmosphère. Upon completion, the reaction mixture was diluted with H2O (50 mL) and extracted with DCM (50 mL * 2). The combined organic layers were washed with brine (50 mL* 2), dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by flash silica gel chromatography (ISCO®; 24 g SepaFlash® Silica Flash Column, Eluent of 0~30% MeOH/DCM @ 50 mL/min) to give 5 (3.6 g, 84.44% yield) as a white solid. ESI-LCMS: m/z 512.1 [M+H]+ ; ‘H NMR (400 MHz, DMSO-d6) δ =11.42 (s, 1H), 7.75 (d, >8.1 Ηζ,ΙΗ), 7.49 (t, >6.2 Hz, 1H), 5.83 (d, >5.8 Hz, 1H), 5.70 - 5.61 (m, 1H), 4.33 4.23 (m, 1H), 3.95 (t,>5.5Hz, 1H), 3.90 - 3.78 (m, 1H), 3.73(t, >6.5 Hz, 2H), 3.30 (s, 3H), 3.26- 3.12 (m, 4H), 2.14 - 2.02 (m, 2H), 0.88 (s, 9H), 0.11 (d,>3.3 Hz, 6H).
Préparation of (6): To a solution of 5 (5 g, 9.76 mmol) in DMF (45 mL) was added DBU (7.43 g, 48.82 mmol, 7.36 mL). The mixture was stirred at 25 °C for 16 h. The reaction mixture was concentrated to give a residue, diluted with H2O (50 mL) and extracted with EA (50 mL * 2). The combined organic layers were washed with brine (50 mL * 2), dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by flash silica gel chromatography (ISCO®; 24 g SepaFlash® Silica Flash Column, Eluent of 0-80% EA/PE @ 40 mL/min) to give 6 (4.4 g, 89.06% yield) as a white solid. ESI-LCMS: m/z 476.1 [M+H]+ ; 'H NMR (400 MHz, DMSO-d6) δ =11.43 (d, >1.7 Hz, 1H), 7.72 (d, >8.1 Hz, 1H), 5.82 (d,>4.8 Ηζ,ΙΗ), 5.67 (dd, >2.1, 8.1 Hz, 1H), 4.22 (t, >5.1 Hz, 1H), 3.99 - 3.87 (m, 2H), 3.33 - 3.27 (m, 6H), 3.09 (dd, >6.6, 14.7 Hz, 1H), 2.26 - 2.16 (m, 2H), 0.88 (s, 9H), 0.10 (d, >3.8 Hz, 6H).
Préparation of (7): To a solution of 6 (200 mg, 420.49 umol) in MeOH (2 mL) was added NH4F (311.48 mg, 8.41 mmol, 20 eq), and the mixture was stirred at 80 °C for 2 h. The mixture was filtered and concentrated to give a residue, which was purified by flash silica gel chromatography (ISCO®; 4 g SepaFlash® Silica Flash Column, Eluent of 0-50% MeOH/DCM @ 20 mL/min) to give 7 (120 mg, 76.60% yield) as a white solid. ESI-LCMS: m/z 362.1 [M+H]+ ; 'H NMR (400 MHz, DMSO-d6) δ =11.37 (br s, 1H), 7.68 (d, >8.1 Ηζ,ΙΗ), 5.81 (d, >4.6 Hz, 1H), 5.65 (d, >8.0 Hz, 1H), 4.02 (q, >5.6 Ηζ,ΙΗ), 3.95 - 3.83 (m, 2H), 3.34 (s, 9H), 3.09 (dd, >6.9, 14.6 Hz, 1H), 2.26 - 2.14 (m, 2H).
Préparation of (Example 50 monomer): To a solution of 7 (1.5 g, 4.15 mmol) in CH3CN (12 mL) were added 3-bis(diisopropylamino)phosphanyloxypropanenitrile (1.63 g, 5.40
240 mmol, 1.71 mL) and lH-imidazole-4,5-dicarbonitrile (539.22 mg, 4.57 mmol) in one portion at 0 °C. The reaction mixture was gradually warmed to 25 °C. The reaction mixture was stirred at 25 °C for 2 h under N2 atmosphère. Upon completion, the reaction mixture was diluted with NaHCCh (20 mL) and extracted with DCM (20 mL * 2). The combined organic layers were washed with brine (20 mL * 2), dried over Na2SÜ4, filtered and concentrated under reduced pressure to give a residue, which was purified by flash silica gel chromatography (ISCO®; 12 g SepaFlash® Silica Flash Column, Eluent of 0-85% EA /PE with 0.5% TEA @ 30 mL/min to give Example 50 monomer (800 mg, 33.6% yield, ) as a white solid. ESI-LCMS: m/z 562.3 [M+H]+; [H NMR (400 MHz, CD3CN) δ = 9.28 (br s,lH), 7.55 (br dd, 7=8.3, 12.8 Ηζ,ΙΗ), 5.86 (br d, 7=3.9 Hz, 1H), 5.65(br d, 7=8.0 Hz, 1H), 4.33 - 4.06 (m, 2H), 4.00 - 3.89 (m, 1H), 4.08 3.86(m, 1H), 3.89 - 3.72 (m, 4H), 3.43 (br d, 7=15.1 Hz, 6H), 3.23 - 3.05 (m, 3H), 2.69 (br s, 2H), 2.36 - 2.24 (m, 2H), 1.26 - 1.10 (m, 12H) ; 31P NMR (162 MHz, CD3CN) δ = 149.94 , 149.88.
Example 51: Synthesis of 5’ End Cap Monomer
Example 51 Monomer
Example 51 Monomer Synthesis Scheme
Préparation of (2): To a solution of 1 (30 g, 101.07 mmol, 87% purity) in CH3CN (1.2 L) and Py (60 mL) were added I2 (33.35 g, 131.40 mmol, 26.47 mL) and PPI13 (37.11 g, 141.50 mmol) in one portion at 10 °C. The reaction was stirred at 25 °C for another 48 h. The mixture 241 was diluted with aq.Na2S2O3 (300 mL) and aq.NaHCO3 (300 mL), concentrated to remove CH3CN, and then extracted with EtOAc (300 mL * 3). The combined organic layers were washed with brine (300 mL), dried over Na2SC>4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (ISCO®; 330 g SepaFlash® Silica Flash Column, Eluent of 0~60% Methanol/Dichloromethane gradient @ 100 mL/min) to give 2 (28.2 g, 72.00% yield, 95% purity) as a brown solid. ESILCMS: m/z 369.1 [M+H]+ ; ‘H NMR (400 MHz, DMSO-d6) δ = 11.43 (s, 1H), 7.68 (d, >8.1 Hz, 1H), 5.86 (d, >5.5 Hz, 1H), 5.69 (d, >8.1 Hz, 1H), 5.46 (d, >6.0 Hz, 1H), 4.08 -3.96 (m, 2H), 3.90 - 3.81 (m, 1H), 3.60 - 3.51 (m, 1H), 3.40 (dd, >6.9, 10.6 Hz, 1H), 3.34 (s, 3H).
Préparation of (3): To a solution of 2 in DMF (90 mL) were added imidazole (4.25 g, 62.48 mmol) and TBSC1 (6.96 g, 46.18 mmol) in one portion at 15°C. The mixture was stirred at 15 °C for 6 h. The reaction mixture was quenched by addition of H2O (300 mL) and extracted with EtOAc (300 mL * 2). The combined organic layers were washed with brine (300 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to give 3 (13.10 g, crude) as a white solid. ESI-LCMS: m/z 483.0 [M+H]+.
Préparation of (4): To a solution of 3 (10 g, 20.73 mmol) in MeOH (20 mL), H2O (80 mL), and dioxane (20 mL) was added Na2SO3 (15.68 g, 124.38 mmol), and the mixture was stirred at 80 °C for 24 h. The reaction mixture was concentrated under reduced pressure to remove MeOH. The aqueous layer was extracted with EtOAc (80 mL * 2) and concentrated under reduced pressure to give a residue. The residue was triturated with MeOH (100*3 mL) to give 4 (9.5 g, 94.48% yield, 90% purity) as a white solid. ESI-LCMS: m/z 437.0 [M+H]+.
Préparation of (5): To a solution of 4 (11 g, 21.42 mmol, 85% purity) in DCM (120 mL) was added DMF (469.65 mg, 6.43 mmol, 494.37 uL) at 0 °C, followed by dropwise addition of oxalyl dichloride (13.59 g, 107.10 mmol, 9.37 mL). The mixture was stirred at 20 °C for 2 h. The reaction mixture was quenched by addition of water (60 mL) and the organic layer 5 (0.1125 M, 240 mL DCM) was used directly for next step. (This reaction was set up for two batches and combined) ESI-LCMS: m/z 455.0 [M+H]+.
Préparation of (6): 5 (186.4 mL, 0.1125 M in DCM) was diluted with DCM (60 mL) and treated with methylamine (3.26 g, 41.93 mmol, 40% purity). The mixture was stirred at 20 °C for 2 h. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (ISCO®; 40 g SepaFlash® Silica Flash Column, Eluent of 0-10%, MeOH/DCM gradient @ 40 mL/min) to give AGS-9-3-008 (1.82 g, 18.53% yield, 96% purity) as a yellow solid. ESI-LCMS: m/z 472.0 [M+Na]+ ; ‘H NMR (400 MHz, CDC13) δ = 9.08 (s, 1H), 7.31 (d, >8.1 Hz, 1H), 5.78 (d, >8.1 Hz, 1H), 5.57 (d, >3.8
242
Hz, 1H), 4.61 - 4.48 (m, 1H), 4.41 - 4.27 (m, 2H), 4.13 - 4.03 (m, 1H), 3.46 (s, 3H), 3.43 - 3.33 (m, 2H), 2.78 (d, 7=5.2 Hz, 3H), 0.92 (s, 9H), 0.13 (s, 6H).
Préparation of (7): To a solution of 6 (2.3 g, 5.12 mmol) in MeOH (12 mL) was added HCl/MeOH (4 M, 6.39 mL). The mixture was stirred at 20 °C for 2 h. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (ISCO®; 24 g SepaFlash® Silica Flash Column, Eluent of 0-15%, MeOH/DCM gradient @ 30 mL/min) to give 7 (1.4 g, 79.98% yield) as a pink solid. ESI-LCMS: m/z 336.1 [M+H]+ ; Ή NMR (400 MHz, CDC13) δ = 9.12 (s, 1H), 7.39 (d, 7=8.0 Hz, 1H), 5.79 (d, .7=3.3 Hz, 1H), 5.66 (dd, 7=2.1, 8.2 Hz, 1H), 5.13 (s, 1H), 4.13 (t, 7=4.0, 7.4 Hz, 1H), 4.07 - 4.02 (m, 1H), 3.87 (dd, 7=3.3, 5.5 Hz, 1H), 3.47 (s, 3H), 3.43 - 3.37 (m, 2H), 2.65 (d, 7=4.5 Hz, 3H).
Préparation of (Example 51 monomer): To a mixture of 7 (1.7 g, 5.07 mmol) and 4A MS (1.4 g) in MeCN (18 mL) was added 3-bis(diisopropylamino)phosphanyloxypropanenitrile (1.99 g, 6.59 mmol, 2.09 mL) at 0 °C, followed by addition of lH-imidazole-4,5-dicarbonitrile (658.57 mg, 5.58 mmol) in one portion at 0 °C. The mixture was stirred at 20 °C for 2 h. Upon completion, the reaction mixture was quenched by addition of sat. NaHCO3 solution (20 mL) and diluted with DCM (40 mL). The organic layer was washed with sat. NaHCO3 (20 mL * 2), dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by a flash silica gel column (0% to 5% i-PrOH in DCM with 5% TEA) to give Example 51 monomer (1.30 g, 46.68% yield) as a white solid. ESI-LCMS: m/z 536.2 [M+H]+ ; Ή NMR (400 MHz, CD3CN) δ = 9.00 (s, 1H), 7.40 (d, 7=8.0 Hz, 1H), 5.85 - 5.76 (m, 1H), 5.64 (d, 7=8.0 Hz, 1H), 5.08 (d, 7=5.0 Hz, 1H), 4.42 - 4.21 (m, 2H), 4.00 (td, 7=4.6, 9.3 Hz, 1H), 3.89 - 3.61 (m, 4H), 3.47-3.40 (m, 4H), 3.37 - 3.22 (m, 1H), 2.71 - 2.60 (m, 5H), 1.21 - 1.16 (m, HH), 1.21 - 1.16 (m, 1H); 31P NMR (162 MHz, CD3CN) δ = 150.07, 149.97
Example 52: Synthesis of 5’ End Cap Monomer
243
Example 52 Monomer Synthesis Scheme
Préparation of (2): To a solution of 1 (13.10 g, 27.16 mmol) in THF (100 mL) was added DBU (20.67 g, 135.78 mmol, 20.47 mL). The mixture was stirred at 60 °C for 6 h. Upon completion, the reaction mixture was quenched by addition of sat.NHÆl solution (600 mL) and extracted with EA (600 mL * 2). The combined organic layers were washed with brine (100 ml), dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (ISCO®; 120 g SepaFlash® Silica Flash Column, Eluent of 0-50% (Phase B: ethyl acetate: dichloromethane=l : 1) / Phase A: petroleum ethergradient@ 45 mL/min) to give 2 (5.9 g, 60.1% yield, ) as a white solid. ESI-LCMS: m/z 355.1 [M+H]+ ; Ή NMR (400 MHz, DMSO-d6) δ = 11.61 - 11.30 (m, 1H), 7.76 - 7.51 (m, 1H), 6.04 (d, 7=5.4 Hz, 1H), 5.75 (s, 1H), 5.73 - 5.67 (m, 1H), 4.78 (d, 7=4.9 Hz, 1H), 4.41 (d, 7=1.1 Hz, 1H), 4.30 (t, 7=4.8 Hz, 1H), 4.22 (d, 7=1.4 Hz, 1H), 4.13 (t, 7=5.1 Hz, 1H), 4.06 - 3.97 (m, 1H), 3.94 - 3.89 (m, 1H), 3.82 - 3.75 (m, 1H), 3.33 (s, 3H), 3.30 (s, 2H), 1.17 (t, 7=7.2 Hz, 1H), 0.89 (s, 9H), 0.16 - 0.09 (m, 6H).
Préparation of (3): To a solution of 2 (4 g, 11.28 mmol) in DCM (40 mL) was added Ru(II)-Pheox (214.12 mg, 338.53 umol) in one portion followed by addition of diazo(dimethoxyphosphoryl)methane (2.54 g, 16.93 mmol) dropwise at 0°C under N2. The reaction was stirred at 20 °C for 16 h. Upon completion, the reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (ISCO®; 80 g SepaFlash® Silica Flash Column, Eluent of 0-4% MeOH/DCM@ 60 mL/min) to give 3 (5 g, 86.47% yield) as a red liquid. ESI-LCMS: m/z 477.1 244
[M+H]+ ; *H NMR (400 MHz, DMSO-dô) δ = 11.46 (s, 1H), 7.49 (d, 7=8.0 Hz, 1H), 6.01 -5.87 (m, 1H), 5.75 (dd, 7=2.0, 8.0 Hz, 1H), 4.58 (d, 7=3.8 Hz, 1H), 4.23 (dd, 7=3.8, 7.8 Ηζ,ΙΗ), 3.80 3.68 (m, 6H), 3.30 (s, 3H), 1.65 - 1.46 (m, 2H), 1.28 - 1.16 (m, 1H), 0.91 (s, 9H), 0.10 (d, 7=4.3 Hz, 6H);3'P NMR (162 MHz, DMSO-d6) δ = 27.5
Préparation of (4): To a mixture of 3 (2.8 g, 5.88 mmol) and Nal (1.76 g, 11.75 mmol) in CH3CN (30 mL) was added chloromethyl 2,2-dimethylpropanoate (2.21 g, 14.69 mmol, 2.13 mL) at 25°C. The mixture was stirred at 80 °C for 40 h under Ar. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (ISCO®; 40 g SepaFlash® Silica Flash Column, Eluent of 0~50% Ethylacetate/Petroleum ether gradient @ 40 mL/min) to give 4 (2.1 g, 51.23% yield, 97% purity) as a yellow solid. ESI-LCMS: 677.3 [M+H]+.
Préparation of (5): A mixture of 4 (2.09 g, 3.09 mmol) in H2O (1.5 mL) and HCOOH (741.81 mg, 15.44 mmol, 6 mL) was stirred at 15°C for 40 h. Upon completion, the reaction mixture was quenched by saturated aq.NaHCO3 (300 mL) and extracted with EA (300 mL * 2). The combined organic layers were washed with brine (300 mL), dried over Na2SÛ4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (ISCO®; 20 g SepaFlash® Silica Flash Column, Eluent of 0~5% Methanol/Dichloromethane@ 45 mL/min) to give 5 (1.51 g, 85.19% yield) as a yellow solid. ESI-LCMS: 585.1 [M+Na]+ ; ‘H NMR (400 MHz, DMSO-d6) δ = 11.45 (d, J=1.8 Hz, 1H), 7.44 (d, J=8.2 Hz, 1H), 6.04 (d, J=7.5 Ηζ,ΙΗ), 5.78 -5.51 (m, 6H), 4.39 (t, J=4.4 Hz, 1H), 4.15 (dd, J=4.3, 7.4 Hz, 1H), 4.03 (q, J=7.1 Hz, 1H),1.99 (s, 1H), 1.66 (dd, J=8.6, 10.8 Hz, 1H), 1.55 1.29 (m, 2H), 1.18 (d, J=2.0 Hz, 18H).
Préparation of (Example 52 monomer): To a solution of 5 (2.5 g, 4.44 mmol) in MeCN (30 mL) was added 3-bis(diisopropylamino)phosphanyloxypropanenitrile (1.74 g, 5.78 mmol, 1.84 mL) at 0 °C, followed by lH-imidazole-4,5-dicarbonitrile (577.36 mg, 4.89 mmol) in one portion under Ar. The mixture was gradually warmed to 20 °C and stirred at 20 °C for 1 h. The reaction mixture was quenched by addition of sat.NaHCCh solution (50 mL) and diluted with DCM (250 mL). The organic layer was washed with sat.NaHCCh solution (50 mL * 2), dried over Na2SCU, filtered and concentrated under reduced pressure to give a residue. The residue was purified by a flash silica gel column (0% to 50% EA / PE with 0.5% TEA) to give Example 52 monomer (1.85 g, 54.1% yield) as a white solid. ESI-LCMS: 785.2 [M+Na]+ ;'H NMR (400 MHz, CD3CN) δ = 9.18 (s, 1H), 7.31 (d, 7=8.3 Hz, 1H), 6.06 (d, 7=7.8 Hz, 1H), 5.72 - 5.60 (m, 5H), 4.85 - 4.76 (m, 1H), 4.27 (m, 1H), 3.93 - 3.64 (m, 4H), 3.41 (d, 7=16.6 Hz, 3H),
245
2.80 - 2.62 (m, 2H), 1.76 - 1.49 (m, 3H), 1.23 - 1.19 (m, 30H); 31P NMR (162 MHz, CD3CN) δ = 150.66 (s), 150.30,24.77,24.66.
Example 53: Synthesis of 5’ End Cap Monomer
Example 53 Monomer
Example 53 Monomer Synthesis Scheme
Préparation of (2): To a solution of 1 (15 g, 137.43 mmol) in DCM (75 mL) were added BOC2O (31.49 g, 144.30 mmol, 33.15 mL) and DMAP (839.47 mg, 6.87 mmol, 0.05 eq) at 0 °C. The mixture was stirred at 20 °C for 16 hr, and concentrated under reduced pressure to give 2 (29.9 g, crude) as a yellow oil. ‘H NMR (400MHz, CDCI3) δ = 3.23 (s, 3H), 3.16 (s, 3H), 1.51 (s, 9H).
Préparation of (3): To a solution of 2 (24.9 g, 118.99 mmol) in THF (250 mL) was added n-BuLi (2.5 M, 47.60 mL) dropwise at -78 °C under Ar and stirred at -78 °C for 1 hr. P-3 (17.19 g, 118.99 mmol, 12.83 mL) was added at 0 °C and stirred for 1 hr. The reaction mixture was quenched by saturated aq. NH4CI (100 mL), and then extracted with EA (100 mL * 2). The combined organic layers were washed with brine (100 mL * 2), dried over Na?SO4, fïltered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (ISCO®; 80 g SepaFlash® Silica Flash Column, Eluent of 0-50% Ethylacetate/Petroleum ethergradient @ 65 mL/min) to give 3 (7.1 g, 18.62% yield) as a yellow oil. ESI-LCMS: 339.9 [M+Na]+ ; ‘H NMR (400 MHz, CDCh) δ = 4.12 (s, 1H), 4.08 (s, 1H), 3.83 (s,3H), 3.81 (s, 3H), 3.22 (s, 3H), 1.51 (s, 9H).
246
Préparation of (5): To a mixture of 4 (15 g, 40.27 mmol) and PPTS (10.12 g, 40.27 mmol) in DMSO (75 mL) was added EDCI (23.16 g, 120.81 mmol) at 20 °C. The mixture was stirred at 20 °C for 4 hr. The reaction mixture was diluted with water (150 mL) and extracted with EA (150 mL*2). The combined organic layers were washed with brine (150 mL*2), dried over Na2SC>4, filtered and concentrated under reduced pressure to give 5 (12 g, crude) as a white solid. ESI-LCMS: 371.2[M+H]+; ‘H NMR (400MHz, CDC13) δ = 9.77 (s, 1H), 7.62 (d, >8.1 Hz, 1H), 5.83 - 5.76 (m, 2H), 4.53 (d, J=4.3 Hz, 1H), 4.43 (br t, >4.4 Hz, 1H), 3.95 (br t, >4.7 Hz, 1H), 3.47 - 3.35 (m, 5H), 0.92 (s, 9H), 0.13 (d, >5.8 Hz, 6H).
Préparation of (6): To a solution of P4 (8.02 g, 25.27 mmol) in THF (40 mL) was added n-BuLi (2.5 M, 8.42 mL) dropwise under Ar at -78 °C, and the mixture was stirred at -78 °C for 0.5 hr. A solution of 4 (7.8 g, 21.05 mmol) in THF (40 mL) was added dropwise. The mixture was allowed to warm to 0 °C and stirred for another 2 hr. The reaction mixture was quenched by saturated aq. NH4CI solution (80 mL) and extracted with EA (80 mL). The combined organic layers were washed with brine (80 mL * 2), dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (ISCO®; 80 g SepaFlash® Silica Flash Column, Eluent of 0~38% ethylacetate/petroleum ether gradient @ 60 mL/min) to give 7 (7.7 g, 13.43 mmol, 63.8% yield) as a white solid. ESI-LCMS: 506.2 [M-tBu]+; ‘H NMR (400MHz, CDC13) δ = 8.97 (s, 1H), 7.25 (d, >8.3 Hz, 1H), 6.95 - 6.88 (m, 1H), 6.87 - 6.81 (m, 1H), 5.83 - 5.77 (m, 2H), 4.58 (dd, >4.4, 6.7 Hz, 1H), 4.05 (dd, >5.0, 7.5 Hz, 1H), 3.82 - 3.77 (m, 1H), 3.53 (s, 3H), 3.20 (s, 3H), 1.50 (s, 9H), 0.91 (s, 9H), 0.11 (d, >2.5 Hz, 6H).
Préparation of (7): To a solution of 6 (7.7 g, 13.71 mmol) in MeOH (10 mL) was added HCl/MeOH (4 M, 51.40 mL) at 20 °C. The mixture was stirred at 20 °C for 16 hr. Upon completion, the reaction mixture was concentrated under reduced pressure to remove MeOH. The residue was purified by flash silica gel chromatography (ISCO®; 80 g SepaFlash® Silica Flash Column, Eluent of 0~4% MeOH/DCM @ 60 mL/min) to give 7 (4.1 g, 86.11% yield) as a white solid. ESI-LCMS: 369.9 [M+Na]+; Ή NMR (400MHz, DMSO-d6) δ = 11.44 (s, 1H), 7.66 (d, >8.3 Hz, 1H), 7.11 (q, >4.9 Hz, 1H), 6.69 (dd, >6.0, 15.1 Hz, 1H), 6.56 - 6.47 (m, 1H), 5.82 (d, >4.0 Hz, 1H), 5.67 (dd, >2.0, 8.0 Hz, 1H), 5.56 (br s, 1H), 4.42 (t, >6.1 Hz, 1H), 4.13 (t, >5.8 Hz, 1H), 3.97 (t, >4.8 Hz, 1H), 3.39 (s, 3H), 2.48 (d, >5.3 Hz, 3H)
Préparation of (8): To a solution of 7 (2.5 g, 7.20 mmol) in THF (25 mL) was added Pd/C (2.5 g, 10% purity) under H2 atmosphère, and the suspension was degassed and purged with H2 for 3 times. The mixture was stirred under H2 (15 Psi) at 20 °C for 1 hr. Upon completion, the reaction mixture was filtered and concentrated under reduced pressure to give a
247 residue. The residue was purified by flash silica gel chromatography (ISCO®; 25 g SepaFlash® Silica Flash Column, Eluent of 0-5% Ethylacetate/Petroleum ethergradient @ 50 mL/min) to give 8 (2.2 g, 87.49% yield, ) as a white solid. ESI-LCMS: 372.1 [M+Na]+; Ή NMR (400 MHz, DMSO-d6) δ = 11.40 (s, 1H), 7.62 (d, J=8.0 Hz, 1H), 6.93 (q, J=4.9 Hz, 1H), 5.76 (d, J=4.5 Hz, 1H), 5.66 (d, J=8.0 Hz, 1H), 5.26 (d, J=6.3 Hz, 1H), 3.97 (q, J=5.9 Hz, 1H), 3.91 - 3.79 (m, 2H), 3.36 (s, 3H), 3.14 - 3.00 (m, 2H), 2.56 (d, J=5.0 Hz, 3H), 2.07 - 1.87 (m, 2H).
Préparation of (Example 53 monomer): To a solution of 8 (2.2 g, 6.30 mmol, 1 eq) in CH3CN (25 mL) was added P-l (2.47 g, 8.19 mmol, 2.60 mL, 1.3 eq) at 0 °C, and then 1Himidazole-4,5-dicarbonitrile (818.07 mg, 6.93 mmol, 1.1 eq) was added in one portion at 0°C under Ar. The mixture was stirred at 20 °C for 2 hr. Upon completion, the reaction mixture was quenched by saturated aq. NaHCO3 (25 mL), and extracted with DCM (25 mL * 2). The combined organic layers were washed with brine (25 mL * 2), dried over NaiSCL, filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (ISCO®; 40 g SepaFlash® Silica Flash Column, Eluent of 40-85% ethylacetate/petroleum ether gradient @ 40 mL/min) to give Example 53 monomer (2.15 g, 61.32% yield) as a white solid. ESI-LCMS: 572.2 [M+Na]+ ;‘H NMR (400MHz, CD3CN) δ = 9.32 (br s, 1H), 7.39 (d, J=8.1 Hz, 1H), 5.82 - 5.75 (m, 1H), 5.66 (dd, J=0.7, 8.1 Hz, 1H), 5.14 (qd, J=4.9, 9.4 Hz, 1H), 4.24 - 4.02 (m, 2H), 3.99 - 3.93 (m, 1H), 3.90 - 3.60 (m, 4H), 3.43 (d, J=17.5 Hz, 3H), 3.18 -3.08 (m, 2H), 2.74 - 2.61 (m, 5H), 2.19 - 2.11 (m, 1 H), 2.09 - 1.98 (m, 1H), 1.19 (ddd, J=2.4, 4.0, 6.6 Hz, 12H).31P NMR (162 MHz, CD3CN) δ = 149.77 (s), 149.63 (br s).
Example 54. Long-term Efficacy of siNA in an AAV-HBV mouse model
AAV/HBV is a recombinant AAV carrying replicable HBV genome. Taking advantage of the highly hepatotropic feature of génotype 8 AAV, the HBV genome can be efficiently delivered to the mouse liver cells. Infection of immune competent mouse with AAV/HBV can resuit in long terni HBV viremia, which mimics chronic HBV infection in patients. The AAV/HBV model can be used to evaluate the in vivo activity of varions types of anti-HBV agents. Mice were infected with AAV-HBV on day -28 of the study. AAV-HBV mice were subcutaneously injected with a single dose of 5mL/kg of vehicle or 5mg/kg of ds-siNA0147 on day 0. Serial blood collections were usually taken every 5 days on day 0, 5, 10, and 15, etc. until the termination of the study. Sérum HBV S antigen (HBsAg) was assayed through ELIS A. FIG. 13 shows a graph of the change in sérum HBsAg from AAV-HBV mice treated with vehicle (G 15) or ds-siNA-0147 (G 19). As shown in FIG. 13, ds-siNA-0147 was effective in reducing sérum HBsAg levels and the réduction in sérum HBsAg levels was observed for the
248 duration of the study (i.e., 100 days). Thus, FIG. 13 demonstrates that ds-siNA-0147 is effective and durable after a single dose of 5 mg/kg.
ds-siNA ID Strand Sequence SEQ ID NO:
ds-siNA- 0147 Sense 5 ’ -mGpsmUpsmGmGfUmGfGfAfCmUmUmCmUmCmU mCmAmAmU-p-ps2-GalNAc4-3 ’ 438
Antisense 3 ’ -mApsmGpsmCm AmCfCmAfCmCmUmGmAm AmG mAfGmAmGmUpsfUpsmA-5 ’ 501
Example 55. Deuterated vinyl phosphonate improves potency of siNA
This example investigates whether a deuterated vinyl phosphonate improves potency of siNA in an AAV-HBV mouse. AAV-HBV mice were subcutaneously injected with vehicle, dssiNA-0109 (e.g., siNA without a deuterated vinyl phosphonate), or ds-siNA-0172 (e.g., siNA with a deuterated vinyl phosphonate). AAV-HBV mice were subcutaneously injected with a single dose of 5 mL/kg of vehicle or 5 mg/kg of ds-siNA-0149 or ds-siNA-0172 at day 0. Serial 10 blood collections were usually taken every 5 days on day 0, 5, 10, and 15, etc. until the termination of the study. Sérum HBV S antigen (HBsAg) was assayed through ELISA.
As shown in FIG. 14, siNA molécules having 2’-fluoro nucléotides at positions 5 and 7-9 from the 5’ end of the sense strand and 2’-fluoro nucléotides at positions 2, 5, 8, 14, and 17 from the 5’ end of the antisense strand resulted in greater than a 0.5-log réduction in HBsAg, with the 15 greatest réduction in HBsAg found in mice treated with the deuterated vinylphopshonate siNA (ds-siNA-0172). In addition, the duration of the réduction in sérum HBsAg levels was significantly longer for the deuterated vinylphosphonate siNA (ds-siNA-0172). Thus, FIG. 14 demonstrates that the presence of a deuterated vinyl phosphonate improves potency and durability of the siNA.
ds-siNA ID Strand Sequence SEQID NO:
ds-siNA- 0109 Sense 5’-mCpsmCpsmGmUfGmUfGfCfAmCmUmUmCmGmC mUmUmCmAp-ps2-GalNAc4 424
Antisense 3 ’-mCpsmUpsmGmGfCmAmCfAmCmGmUmGmAfAmG mCfGmAmApsfGpsmU-5 ’ 485
ds-siNA- 0172 Sense 5’-mCpsmCpsmGmUfGmUfGfCfAmCmUmUmCmGmC mUmUmCmA-p-ps2-GalNAc4-3 ’ 424
249
Antisense 3’-mCpsmUpsmGmGfCmAmCfAmCmGmUmGmAfAmG mCfGmAmApsfGpsd2vd3U-5’ 536
O Ho^' D HO Ο dZv/ 0 0' OCDî d2vd3U =
Example 56. Comparison of siNAs
AAV/HBV is a recombinant AAV carrying repli cable HBV genome. Taking advantage of the highly hepatotropic feature of génotype 8 AAV, the HBV genome can be efficiently delivered to the mouse liver cells. Infection of immune competent mouse with AAV/HBV can resuit in long term HBV viremia, which mimics chronic HBV infection in patients. The AAV/HBV model can be used to evaluate the in vivo activity of various types of anti-HBV agents. Mice were infected with AAV-HBV on day -28 of the study. AAV-HBV mice were subcutaneously injected with a single dose of 5mL/kg of vehicle or 5mg/kg of ds-siNA-
0109, ds-siNA-0119, or ds-siNA-0153 on day 0. Serial blood collections were usually taken every 5 days on day 0, 5, 10, and 15, etc. until the termination of the study. Sérum HBV S antigen (HBsAg) was assayed through ELIS A. FIG. 15 shows a graph of the change in sérum HBsAg from AAV-HBV mice treated with vehicle (G 01, circle), ds-siNA-0109 (G 07, square), ds-siNA-0119 (G11, triangle), or ds-siNA-0153 (G13, diamond). As shown in FIG. 14, ail three ds-siNAs were effective in reducing sérum HBsAg levels and the réduction in sérum HBsAg levels was observed for the duration of the study (i.e., 100 days), with the best potency and durability observed for ds-siNA-0153. Thus, FIG. 15 demonstrates that ds-siNA-0109, ds-siNA0119, and ds-siNA-0153 were effective and durable after a single dose of 5 mg/kg.
ds-siNA ID Strand Sequence SEQID NO:
ds-siNA- 0109 Sense 5’-mCpsmCpsmGmUfGmUfGfCfAmCmUmUmCmGmC mUmUmCmAp-ps2-GalNAc4 424
Antisense 3 ’ -mCpsmUpsmGmGfCm AmCfAmCmGmUmGm AfAmG mCfGmAmApsfGpsmU-5 ’ 485
ds-siNA- 0119 Sense 5’-mGpsmCpsmUmGfCmUmAmUfGfCfCmUmCfAmU mCmUmUfCmUmU-p-ps2-GalNAc4 430
250
Antisense 3’-mGpsmApsmCmGmAmCmGmAmUfAmCmGmGmA mGmUmAmGmAm AmGpsfApsm A-5 ’ 595
ds-siNA- 0153 Sense 5’-mUpsmGpsfUmGmCmAfCfUfUmCmGfCmUmUmC mAfCmCmU-p-ps2-GalNAc4-3 ’ 441
Antisense 3’-mGpsmCpsmAfCmAmCmGfUmGmAmAfGmCmGmA fAmGmUmGpsfGpsmA-5 526
Example 57. Efficacy of a Combination Therapy in AAV-HBV Mouse Model
This example investigates the efficacy of a combination therapy comprising an antisense oligonucleotide (ASO 1,5’ GalNAc4-ps-GalNAc4-ps-GalNAc4-po-mA-polnGpslnApslnTpslnApslnApsApsAps(5OH)CpsGps(5m)Cps(5m)CpsGps(5m)CpslnApslnGpsln Apscp(5m)C-3’(SEQ ID NO: 534)) and a ds-siNA-0147 for treating HBV in an AAV-HBV mouse model.
AAV-HBV mice were subcutaneously injected with (a) 5mL/kg of vehicle, three times a week, on days 0, 7, and 14 (G 01); (b) 5 mg/kg of ASO 1 on a weekly basis, on days 0, 7, and 14 (G 20); (c) a single dose of 5 mg/kg of ds-siNA-0147 on day 0 (G 24); or (d) a combination of ASO 1 and ds-siNA-0147, wherein ASO 1 was administered at a dose of 5 mg/kg on a weekly basis, on days 0, 7, and 14; and ds-siNA-0160 was administered as a single dose of 5 mg/kg at day 0 (G25). Serial blood collections were usually taken every 5 days on day 0, 5, 10, and 15, etc. until the termination of the study. Sérum HBV S antigen (HBsAg) was assayed through ELIS A. FIG. 16 shows a graph of the change in sérum HBsAg from AAV-HBV mice treated with vehicle (G 01, circle), ASO 1 (G 20, square), ds-siNA-0147 (G 24, diamond), or a combination of ds-siNA-0147 and ASO 1 (G 25, triangle). As shown in FIG. 16, treatment with ASO 1, ds-siNA-0147, or a combination of ASO 1 and ds-siNA-0147 resulted in a réduction in sérum, with the greatest réduction observed in mice treated with the combination of ASO 1 and ds-siNA-0147.
ds-siNA ID Strand Sequence SEQ ID NO:
ds-siNA- 0147 Sense 5 ’ -mGpsmUpsmGmGfUmGfGfAfCmUmUmCmUmCmU mCmAmAmU-p-ps2-GalNAc4-3 ’ 438
Antisense 3 ’ -mApsmGpsmCm AmCfCmAfCmCmUmGmAmAmG mAfGmAmGmUpsfUpsmA-5 ’ 501
Example 58. Efficacy of a Combination Therapy in AAV-HBV Mouse Model
251
This example investigates the efficacy of a combination therapy comprising an antisense oligonucleotide (ASO 1,5’ GalNAc4-ps-GalNAc4-ps-GalNAc4-po-mA-polnGpslnApslnTpslnApslnApsApsAps(5OH)CpsGps(5m)Cps(5m)CpsGps(5m)CpslnApslnGpsln Apscp(5m)C-3’(SEQ ID NO: 534)) and a ds-siNA-0109 for treating HBV in an AAV-HBV mouse model.
AAV-HBV mice were subcutaneously injected with (a) 5mL/kg of vehicle, three times a week, on days 0, 7, and 14 (G 01); (b) 5 mg/kg of ASO 1 on a weekly basis, on days 0, 7, and 14 (G 20); (c) a single dose of 5 mg/kg of ds-siNA-0109 on day 0 (G 26); or (d) a combination of ASO 1 and ds-siNA-0109, wherein ASO 1 was administered at a dose of 5 mg/kg on a weekly basis, on days 0, 7, and 14; and ds-siNA-0160 was administered as a single dose of 5 mg/kg at day 0 (G27). Serial blood collections were usually taken every 5 days on day 0, 5, 10, and 15, etc. until the termination of the study. Sérum HBV S antigen (HBsAg) was assayed through ELISA. FIG. 17 shows a graph of the change in sérum HBsAg from AAV-HBV mice treated with vehicle (G 01, circle), ASO 1 (G 20, square), ds-siNA-0109 (G 26, diamond), or a combination of ds-siNA-0109 and ASO 1 (G 27, triangle). As shown in FIG. 17, treatment with ASO 1, ds-siNA-0109, or a combination of ASO 1 and ds-siNA-0109 resulted in a réduction in sérum, with the greatest réduction observed in mice treated with the combination of ASO 1 and ds-siNA-0109.
ds-siNA ID Strand Sequence SEQID NO:
ds-siNA- 0109 Sense 5 ’ -mCpsmCpsmGmUfGmUfGfCfAmCmUmUmCmGmC mUmUmCmAp-ps2-GalNAc4 424
Antisense 3 ’ -mCpsmUpsmGmGfCmAmCfAmCmGmUmGmAfAmG mCfGmAmApsfGpsmU-5 ’ 485
Example 59. Rôle of 2’-Fluoro Mimics on siNA Activity
This example investigates the rôle of 2’-fluoro mimics, f4P and f2P monomers, on siNA activity. The f4P monomer was produced as described in Example 42. The f2P monomer was produced as described in Example 45.
The activity of ds-siNA-0173, ds-siNA-0174, and ds-siNA-0175 was assayed using an in vitro HBsAg sécrétion assay with HepG2.2.15 cells. Generally, HepG2.2.15 cells were maintained in DMEM medium with 10% fêtai bovine sérum (FBS) and 1% penicillin/streptomycin, 1% Glutamine, 1% non-essential amino acids, 1% Sodium Pyruvate and 250 ug/ml G418. Cells were maintained at 37°C in a 5% CO2 atmosphère. For HBsAg release
252 assay, an assay medium was made that DMEM with 5% FBS, 1% penicillin/streptomycin, 1% Glutamine and 1% DMSO. The day before the assay, HepG2.2.15 cells were trypsinized and washed with Assay Medium once, then spun at 250g x 5min, resuspended with Assay Medium. The resuspenced cells were seeded at 50,000/well in assay medium in collagen coated 96 well plates. On the next day, siRNA was diluted with Opti-MEM, 9-pt, 3-fold dilution and dilute Lipofectamine RNAiMAX (Invitrogen) according manufacturées manual. siRNA dilution and RNAiMAX dilution were mixed and incubated at room température for 5 minutes. 15 μΐ of the siRNA/RNAiMax mixture was added each well of the collagen coated 96 well plate. The plates were placed in a 37°C, 5% CO2 incubator for 4 days. After incubation, the supematant was harvested and measured for HBsAg with ELIS A kit (Diasino). The cell viability was measured with CellTiter-Glo (Promega). The EC50, the concentration of the drug required for reducing HBsAg sécrétion by 50% in relation to the untreated cell control, was calculated using the Prism Graphpad. The CC50, the concentration of the drug required for reducing cell viability by 50% in relation to the untreated cell control, was calculated with the same software. The EC50 and
CC50 values are shown in Table 11.
Table 11. siNA Activity
ds-siNA ID Strand Sequence SEQ ID NO: EC50 (nM)s CC50 (nM)
ds-siNA- 0173 Sense 5 ’ -mGpsmUpsmGmGfUmGfGfAfC mUmUmCmUmCmUmCmAmAmU 438 C >1
Antisense 3 ’-mApsmGpsmCmAmCfCmA fCmCmUmGmAmAmGmAfGmAmG mUpsf4PpsmA-5 ’ 537
ds-siNA- 0174 Sense 5 ’ -mGpsmUpsmGmGfUmGfGfAfC mUmUmCmUmCmUmCmAmAmU 438 A >1
Anti- sense 3 ’-mApsmGpsmCmAmCfCmAf2P mCmUmGmAmAmGmAfGmAmGm UpsfUpsmA-5’ 538
ds-siNA0175 (control) Sense 5 ’ -mGpsmUpsmGmGfUmGfGfAfC mUmUmCmUmCmUmCmAmAmU 438 B >1
Anti- sense 5 ’ -mApsfUpsmUmGmAfGm AmG mAmAmGmUmCfCmAfCmCmAmC psmGpsmA-3 ’ 501
*A = EC50 < 0.2 nM; B = 0.2 nM < EC50 < 0.1 nm; C = EC50 > 0.1 nm
253
Example 60. Rôle of 2’-Fluoro Mimics on siNA Activity
This example investigates the rôle of 2’-fluoro mimics, f4P, f2P, and fx monomers, on siNA activity of GalNAc4 conjugated siNAs. The f4P monomer was produced as described in
Example 42. The f2P monomer was produced as described in Example 45. The fx monomer was produced as described in Example 41.
ds-siNA ID Strand Sequence SEQID NO:
ds-siNA- 0176 Sense 5 ’ -mGpsmUpsmGmGfUmGfGfAfCmUmUmCmUmCmU mCmAmAmU-p-(PS)2-GalNAc4 438
Antisense 3 ’ -m ApsmGpsmCmAmCfCm AfCmCmUmGm Am AmGmA fGmAmGmUpsf4PpsmA-5 ’ 537
ds-siNA- 0177 Sense 5 ’ -mGpsmUpsmGmGfUmGfGfAfCmUmUmCmUmCmU mCmAmAmU-p-(PS)2-GalNAc4 438
Antisense 3 ’-mApsmGpsmCmAmCfCmAf2PmCmUmGmAmAmGm AfGmAmGmUpsfUpsmA-5 ’ 538
ds-siNA- 0178 Sense 5 ’ -mGpsmUpsmGmGfUmGfGfAfCmUmUmCmUmCmU mCmAmAmU-p-(PS)2-GalNAc4 438
Antisense 3 ’ -mApsmGpsmCmAmCfCmAfXmCmUmGmAmAmG mAfGmAmGmUpsfUpsmA-5 ’ 539
ds-siNA- 0147 Sense 5 ’ -mGpsmUpsmGmGfUmGfGfAfCmUmUmCmUmCmU mCmAmAmU-p-ps2-GalNAc4 438
Antisense 3 ’ -m ApsmGpsmCmAmCfCm AfCmCmUmGm Am AmG mAfGmAmGmUpsfUpsmA-5 ’ 501
3? U II o vw>O J ►ϋ II / O WV O f -Xk Z £ II O oX Z •‘N Z T ΙΌ
254
The activity of ds-siNA-017, ds-siNA-017, ds-siNA-017, and ds-siNA-0147 can be assayed using in vitro or in vivo methods. An exemplary in vitro assay can be performed as follows:
Homo sapiens HepG2.2.15 cells are cultured in Dulbecco’s Modified Eagle’s Medium (DMEM) (ATCC 30-2002) supplemented to also contain 10% fêtai calf sérum (FCS). Cells were incubated at 37°C in an atmosphère with 5% CO2 in a humidified incubator. For transfection of HepG2.2.15 cells with HBV targeting siRNAs, cells are seeded at a density of 15000 cells/well in 96-well regular tissue culture plates. Transfection of cells is carried out using RNAiMAX (Invitrogen/Life Technologies) according to the manufacturer’s instructions. Dose-response experiments are done with oligo concentrations of 40, 20, 10, 5, 2.5, 1.25, 0.625, 0.3125, 0.15625 and 0.07813nM. For each HBV targeting siRNA treatment (e.g., ds-siNA-0176, dssiNA-0177, ds-siNA-0178, or ds-siNA-0147), four wells are transfected in parallel, and individual data points were collected from each well. After 24h of incubation with siRNA, media is removed, and cells are lysed and analyzed with a QuantiGene2.0 branched DNA (bDNA) probe set spécifie for HBV génotype D (also called Hepatitis B virus subtype ayw, complété genome of 3182 base-pairs) as présent in cell line HepG2.2.15.
For each well, the HBV on-target mRNA levels is normalized to the GAPDH mRNA level. The activity of the HBV targeting ds-siNAs can be expressed as EC50, 50% réduction of normalized HBV RNA level from no drug control. The cytotoxicity of the HBV targeting dssiRNAs can be expressed by CC50 of 50% réduction of GAPDH mRNA from no drug control.
The AAV/HBV model can be used to evaluate the in vivo activity of the siRNA treatment (e.g., ds-siNA-0173, ds-siNA-0174, ds-siNA-0175, and ds-siNA-0147). Mice are infected with AAV-HBV on day -28 of the study. AAV-HBV mice are subcutaneously injected with a single dose of 5mL/kg of vehicle or 5mg/kg of ds-siNA-0173, ds-siNA-0174, ds-siNA0175, or ds-siNA-0147 on day 0. Serial blood collections can be taken every 5 days on day 0, 5, 10, and 15, etc. until the termination of the study. Sérum HBV S antigen (HBsAg) can be assayed through ELISA.
Exemplary Embodiments
Exemplary embodiments are provided below:
1. A short interfering nucleic acid (siNA) molécule comprising:
(a) a sense strand comprising a first nucléotide sequence that is at least about 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% identical to an RNA corresponding to a target gene, wherein the first nucléotide sequence:
(i) is 15 to 30 nucléotides in length; and
255 (ii) comprises 15 or more modified nucléotides independently selected from a 2’-O-methyl nucléotide and a 2’-fluoro nucléotide, wherein at least one modified nucléotide is a 2’-O-methyl nucléotide and the nucléotide at positions, 5, 7, 8, 9, 10, 11, 12, 14, 17, and/or 19 from the 5’ end ofthe first nucléotide sequence is a 2’-fluoro nucléotide; and (b) an antisense strand comprising a second nucléotide sequence that is at least about 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% complementary to the RNA corresponding to the target gene, wherein the second nucléotide sequence:
(i) is 15 to 30 nucléotides in length; and (ii) comprises 15 or more modified nucléotides independently selected from a 2’-O-methyl nucléotide and a 2’-fluoro nucléotide, wherein at least one modified nucléotide is a 2’-O-methyl nucléotide and at least one modified nucléotide is a 2’-fluoro nucléotide.
2. A short interfering nucleic acid (siNA) molécule comprising:
(a) a sense strand comprising a first nucléotide sequence that is at least about 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% identical to an RNA corresponding to a target gene, wherein the first nucléotide sequence:
(i) is 15 to 30 nucléotides in length; and (ii) comprises 15 or more modified nucléotides independently selected from a 2’-(9-methyl nucléotide and a 2’-fluoro nucléotide, wherein at least one modified nucléotide is a 2’-O-methyl nucléotide and at least one modified nucléotide is a 2’-fluoro nucléotide; and (b) an antisense strand comprising a second nucléotide sequence that is at least about 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% complementary to the RNA corresponding to the target gene, wherein the second nucléotide sequence:
(i) is 15 to 30 nucléotides in length; and (ii) comprises 15 or more modified nucléotides independently selected from a 2’-(9-methyl nucléotide and a 2’-fluoro nucléotide, wherein at least one modified nucléotide is a 2’-O-methyl nucléotide and the nucléotide at position 2, 5, 6, 8, 10, 14, 16, 17, and/or 18 from the 5’ end of the second nucléotide sequence is a 2’-fluoro nucléotide.
256
3. The siNA of embodiment 1 or 2, wherein the first nucléotide sequence comprises 16, 17, 18, 19, 20, 21, 22, 23, or more modified nucléotides independently selected from a 2’-O-methyl nucléotide and a 2'-fluoro nucléotide.
4. The siNA of embodiment 1 or 2, wherein 70%, 75%, 80%, 85%, 90%, 95% or 100% of the nucléotides in the first nucléotide sequence are modified nucléotides independently selected from a 2’-O-methyl nucléotide and a 2'-fluoro nucléotide.
5. The siNA of any one of embodiments 1-4, wherein at least 2, 3, 4, 5, or 6 modified nucléotides of the first nucléotide sequence are 2’-fluoro nucléotides.
6. The siNA of any one of embodiments 1-5, wherein no more than 10, 9, 8, 7, 6, 5, 4, 3, or 2 modified nucléotides of the first nucléotide sequence are 2’-fluoro nucléotides.
7. The siNA of any one of embodiments 1-6, wherein at least 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, or 22 modified nucléotides of the first nucléotide sequence are 2’-O-methyl nucléotides.
8. The siNA of any one of embodiments 1-7, wherein no more than 25, 24, 23, 22, 21, 20, 19, 18, 17, 16, 15, 14, 13, 12, 11, 10, 9, 8, 7, 6, 5, 4, 3, or 2 modified nucléotides of the first nucléotide sequence are 2’-O-methyl nucléotides.
9. The siRNA of any one of embodiments 1-8, wherein the second nucléotide sequence comprises 16, 17, 18, 19, 20, 21, 22, 23, or more modified nucléotides independently selected from a 2’-O-methyl nucléotide and a 2'-fluoro nucléotide.
10. The siNA of any one of embodiments 1-9, wherein 70%, 75%, 80%, 85%, 90%, 95% or 100% of the nucléotides in the second nucléotide sequence are modified nucléotides independently selected from a 2’-O-methyl nucléotide and a 2’-fluoro nucléotide.
11. The siNA of any one of embodiments 1-10, wherein at least 2, 3, 4, 5, or 6 modified nucléotides of the second nucléotide sequence are 2’-fluoro nucléotides.
12. The siNA of any one of embodiments 1-11, wherein less than or equal to 10, 9, 8, 7, 6, 5, 4, 3, or 2 modified nucléotides of the second nucléotide sequence are 2’-fluoro nucléotides.
13. The siNA of any one of embodiments 1-12, wherein at least 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, or 22 modified nucléotides of the second nucléotide sequence are 2’-O-methyl nucléotides.
257
14. The siNA of any one of embodiments 1-12, wherein less than or equal to 25, 24, 23, 22, 21, 20, 19, 18, 17, 16, 15, 14, 13, 12, 11, 10, 9, 8, 7, 6, 5, 4, 3, or 2 modified nucléotides ofthe second nucléotide sequence are 2’-O-methyl nucléotides.
15. A short interfering nucleic acid (siNA) molécule comprising:
(a) a sense strand comprising a first nucléotide sequence that is at least about 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% identical to an RNA corresponding to a target gene, wherein the first nucléotide sequence:
(i) is 15 to 30 nucléotides in length;
(ii) comprises 15 or more modified nucléotides independently selected from a 2’-O-methyl nucléotide and a 2’-fluoro nucléotide, wherein at least one modified nucléotide is a 2’-O-methyl nucléotide and at least one modified nucléotide is a 2’-fluoro nucléotide; and (iii) comprises 1 or more phosphorothioate intemucleoside linkage; and (b) an antisense strand comprising a second nucléotide sequence that is at least about 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% complementary to the RNA corresponding to the target gene, wherein the second nucléotide sequence:
(i) is 15 to 30 nucléotides in length;
(ii) comprises 15 or more modified nucléotides independently selected from a 2’-(9-methyl nucléotide and a 2’-fluoro nucléotide, wherein at least one modified nucléotide is a 2’-O-methyl nucléotide and at least one modified nucléotide is a 2’-fluoro nucléotide; and (iii) comprises 1 or more phosphorothioate intemucleoside linkage.
16. A short interfering nucleic acid (siNA) molécule comprising:
(a) a sense strand comprising a first nucléotide sequence that is at least about 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% identical to an RNA corresponding to a target gene, wherein the first nucléotide sequence:
(i) is 15 to 30 nucléotides in length; and (ii) comprises 15 or more modified nucléotides independently selected from a 2’-O-methyl nucléotide and a 2’-fluoro nucléotide, wherein at least one modified nucléotide is a 2’-O-methyl nucléotide and at least one modified nucléotide is a 2’-fluoro nucléotide; and
258 (b) an antisense strand comprising a second nucléotide sequence that is at least about 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% complementary to the RNA corresponding to the target gene, wherein the second nucléotide sequence:
(i) is 15 to 30 nucléotides in length; and (ii) comprises 15 or more modified nucléotides independently selected from a 2’-O-methyl nucléotide and a 2’-fluoro nucléotide, wherein at least one modified nucléotide is a 2’-O-methyl nucléotide and at least one modified nucléotide is a 2’-fluoro nucléotide, wherein the siNA further comprises a phosphorylation blocker, a galactosamine, or 5’stabilized end cap.
17. The siNA according to any preceding embodiment, wherein at least 1, 2, 3, 4, 5, 6, or 7 nucléotides at position 3, 5, 7, 8, 9, 10, 11, 12, and/or 17 from the 5’ end of the first nucléotide sequence is a 2’-fluoro nucléotide.
18. The siNA according to any preceding embodiment, wherein the nucléotide at position 3 from the 5’ end of the first nucléotide sequence is a 2’-fluoro nucléotide.
19. The siNA according to any preceding embodiment, wherein the nucléotide at position 5 from the 5’ end of the first nucléotide sequence is a 2’-fluoro nucléotide.
20. The siNA according to any preceding embodiment, wherein the nucléotide at position 7 from the 5’ end of the first nucléotide sequence is a 2’-fluoro nucléotide.
21. The siNA according to any preceding embodiment, wherein the nucléotide at position 8 from the 5’ end of the first nucléotide sequence is a 2’-fluoro nucléotide.
22. The siNA according to any preceding embodiment, wherein the nucléotide at position 9 from the 5’ end of the first nucléotide sequence is a 2’-fluoro nucléotide.
23. The siNA according to any preceding embodiment, wherein the nucléotide at position 12 from the 5’ end of the first nucléotide sequence is a 2’-fluoro nucléotide.
24. The siNA according to any preceding embodiment, wherein the nucléotide at position 17 from the 5’ end of the first nucléotide sequence is a 2’-fluoro nucléotide.
25. The siNA according to any preceding embodiment, wherein the nucléotide at position 10 from the 5’ end of the first nucléotide sequence is a 2’-fluoro nucléotide.
259
26. The siNA according to any preceding embodiment, wherein the nucléotide at position 11 from the 5’ end of the first nucléotide sequence is a 2’-fluoro nucléotide.
27. The siNA according to any preceding embodiment, wherein at least 1, 2, 3, 4, 5, 6, 7, 8, or 9 nucléotides at position 2, 5, 6, 8, 10, 14, 16, 17, and/or 18 from the 5’ end of the second nucléotide sequence is a 2’-fhioro nucléotide. .
28. The siNA according to any preceding embodiment, wherein the nucléotide at position 2 from the 5’ end of the second nucléotide sequence is a 2’-fluoro nucléotide.
29. The siNA according to any preceding embodiment, wherein the nucléotide at position 5 from the 5’ end of the second nucléotide sequence is a 2’-fluoro nucléotide.
30. The siNA according to any preceding embodiment, wherein the nucléotide at position 6 from the 5’ end of the second nucléotide sequence is a 2’-fluoro nucléotide.
31. The siNA according to any preceding embodiment, wherein the nucléotide at position 8 from the 5’ end of the second nucléotide sequence is a 2’-fluoro nucléotide.
32. The siNA according to any preceding embodiment, wherein the nucléotide at position 10 from the 5’ end of the second nucléotide sequence is a 2’-fluoro nucléotide.
33. The siNA according to any preceding embodiment, wherein the nucléotide at position 14 from the 5’ end of the second nucléotide sequence is a 2’-fluoro nucléotide.
34. The siNA according to any preceding embodiment, wherein the nucléotides at position 16 from the 5’ end of the second nucléotide sequence is a 2’-fluoro nucléotide.
35. The siNA according to any preceding embodiment, wherein the nucléotide at position 17 from the 5’ end of the second nucléotide sequence is a 2’-fluoro nucléotide.
36. The siNA according to any preceding embodiment, wherein the nucléotide at position 18 from the 5’ end of the second nucléotide sequence is a 2’-fluoro nucléotide.
37. The siNA according to any preceding embodiment, wherein the nucléotides in the second nucléotide sequence are arranged in an altemating 1:3 modification pattern, and wherein 1 nucléotide is a 2’-fluoro nucléotide and 3 nucléotides are 2’-O-methyl nucléotides.
260
38. The siNA of embodiment 37, wherein the altemating 1:3 modification pattern occurs 2-5 times.
39. The siNA according to embodiment 37 or 38, wherein at least two of the altemating 1:3 modification pattern occur consecutively.
40. The siNA according to any of embodiments 37-39, wherein at least two of the altemating 1:3 modification pattern occurs nonconsecutively.
41. The siNA according to any of clams 37-40, wherein at least 1, 2, 3, 4, or 5 altemating 1:3 modification pattern begins at nucléotide position 2, 6, 10, 14, and/or 18 from the 5’ end of the antisense strand.
42. The siNA according to any of clams 37-41, wherein at least one altemating 1:3 modification pattern begins at nucléotide position 2 from the 5’ end of the antisense strand.
43. The siNA according to any of clams 37-42, wherein at least one altemating 1:3 modification pattern begins at nucléotide position 6 from the 5’ end of the antisense strand.
44. The siNA according to any of clams 37-43, wherein at least one altemating 1:3 modification pattern begins at nucléotide position 10 from the 5’ end of the antisense strand.
45. The siNA according to any of clams 37-44, wherein at least one altemating 1:3 modification pattern begins at nucléotide position 14 from the 5’ end of the antisense strand.
46. The siNA according to any of clams 37-45, wherein at least one altemating 1:3 modification pattern begins at nucléotide position 18 from the 5’ end of the antisense strand.
47. The siNA according to any one of embodiments 1 -37, wherein the nucléotides in the second nucléotide sequence are arranged in an altemating 1:2 modification pattern, and wherein nucléotide is a 2’-fluoro nucléotide and 2 nucléotides are 2’-(9-methyl nucléotides.
48. The siNA of embodiment 47, wherein the altemating 1:2 modification pattern occurs 2-5 times.
49. The siNA according to embodiment 47 or 48, wherein at least two of the altemating 1:2 modification pattern occurs consecutively.
261
50. The siNA according to any of embodiments 47-49, wherein at least two of the altemating 1:2 modification pattern occurs nonconsecutively.
51. The siNA according to any of clams 47-50, wherein at least 1, 2, 3, 4, or 5 altemating 1:2 modification pattern begins at nucléotide position 2, 5, 8, 14, and/or 17 from the 5’ end of the antisense strand.
52. The siNA according to any of clams 47-51, wherein at least one altemating 1:2 modification pattern begins at nucléotide position 2 from the 5’ end of the antisense strand.
53. The siNA according to any of clams 47-52, wherein at least one altemating 1:2 modification pattern begins at nucléotide position 5 from the 5’ end of the antisense strand.
54. The siNA according to any of clams 47-53, wherein at least one altemating 1:2 modification pattern begins at nucléotide position 8 from the 5’ end of the antisense strand.
55. The siNA according to any of clams 47-54, wherein at least one altemating 1:2 modification pattern begins at nucléotide position 14 from the 5’ end of the antisense strand.
56. The siNA according to any of clams 47-55, wherein at least one altemating 1:2 modification pattern begins at nucléotide position 17 from the 5’ end of the antisense strand.
57. A short interfering nucleic acid (siNA) molécule represented by Formula (VIII):
’-An1Bn2An3Bn4An5Bn6An7Bn8An 9-3 ’
3’-CqIAq2Bq3Aq4Bq5Aq6Bq7Aq8Bq9Aq10Bq11Aq12-5’ wherein:
the top strand is a sense strand comprising a first nucléotide sequence that is at least about 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% identical to an RNA corresponding to a target gene, wherein the first nucléotide sequence comprises 15 to 30 nucléotides;
the bottom strand is an antisense strand comprising a second nucléotide sequence that is at least about 60%, 65%>, 70%, 75%, 80%, 85%, 90%, 95%, or 100% complementary to the RNA corresponding to the target gene, wherein the second nucléotide sequence comprises 15 to 30 nucléotides;
each A is independently a 2’-O-methyl nucléotide or a nucléotide comprising a 5’stabilized end cap or a phosphorylation blocker;
B is a 2’-fluoro nucléotide;
C represents overhanging nucléotides and is a 2’-O-methyl nucléotide;
262 η1 = 1-4 nucléotides in length;
each n2, n6, n8, q3, q5, q7, q9, q11, and q12 is independently 0-1 nucléotides in length;
each n3 and n4 is independently 1-3 nucléotides in length;
n5 is 1-10 nucléotides in length;
n7 is 0-4 nucléotides in length;
each n9, q1, and q2 is independently 0-2 nucléotides in length;
q4 is 0-3 nucléotides in length;
q6 is 0-5 nucléotides in length;
q8 is 2-7 nucléotides in length; and q10 is 2-11 nucléotides in length.
58. A short interfering nucleic acid (siNA) molécule represented by Formula (IX): 5’-A2-4BlAl-3 B2-3 Α2-ΐθΒθ-1Αθ-4Βθ-1Αθ-2-3’ ’ -C2 A0-2B0-1A0-3B0-1A0-5B0-1 Α2-γΒ i A2-11B i A i -5 ’ wherein:
the top strand is a sense strand comprising a first nucléotide sequence that is at least about 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% identical to an RNA corresponding to a target gene, wherein the first nucléotide sequence comprises 15 to 30 nucléotides;
the bottom strand is an antisense strand comprising a second nucléotide sequence that is at least about 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% complementary to the RNA corresponding to the target gene, wherein the second nucléotide sequence comprises 15 to 30 nucléotides;
each A is independently a 2’-(7-methyl nucléotide or a nucléotide comprising a 5’stabilized end cap or a phosphorylation blocker;
B is a 2’-fluoro nucléotide;
C represents overhanging nucléotides and is a 2’-O-methyl nucléotide.
59. A short interfering nucleic acid (siNA) molécule comprising (a) a sense strand comprising a first nucléotide sequence consisting of 17 to 23 nucléotides, wherein 2’-fluoro nucléotides are at positions 3, 7-9, 12, and 17 from the 5’ end of the first nucléotide sequence, and wherein 2’-(2-methyl nucléotides are at positions 1, 2, 4-6, 10, 11, and 13-16 from the 5’ end of the first nucléotide sequence; and (b) an antisense strand comprising a second nucléotide sequence consisting of 17 to 23 nucléotides, wherein 2’-fluoro nucléotides are at positions 2 and 14 from the 5’
263 end of the second nucléotide sequence, and wherein 2’-(9-methyl nucléotides are at positions 1, 3-13, and 15-17 from the 5’ end of the second nucléotide sequence.
60. The siNA molécule of embodiment 59, wherein the first nucléotide sequence consists of 19 nucléotides.
61. The siNA molécule of embodiment 60, wherein 2’-(9-methyl nucléotides are at positions 18 and 19 from the 5’ end of the first nucléotide sequence.
62. The siNA molécule according to any one of embodiments 59-61, wherein the second nucléotide sequence consists of 21 nucléotides.
63. The siNA molécule of embodiment 62, wherein 2’-O-methyl nucléotides are at positions 18-21 from the 5’ end of the second nucléotide sequence.
64. A short interfering nucleic acid (siNA) molécule comprising (a) a sense strand comprising a first nucléotide sequence consisting of 17 to 23 nucléotides, wherein 2’-fluoro nucléotides are at positions 3, 7, 8, and 17 from the 5’ end of the first nucléotide sequence, and wherein 2’-O-methyl nucléotides are at positions 1, 2, 4-6, and 9-16 from the 5’ end of the first nucléotide sequence; and (b) an antisense strand comprising. a second nucléotide sequence consisting of 17 to 23 nucléotides, wherein 2’-fluoro nucléotides are at positions 2 and 14 from the 5’ end of the first nucléotide sequence; and wherein 2’-O-methyl nucléotides are at positions 1, 3-13, and 15-17 from the 5’ end of the first nucléotide sequence.
65. The siNA molécule of embodiment 64, wherein the first nucléotide sequence consists of 19 nucléotides.
66. The siNA molécule of embodiment 65, wherein 2’-(9-methyl nucléotides are at positions 18 and 19 from the 5’ end of the first nucléotide sequence.
67. The siNA molécule according to any one of embodiments 64-66, wherein the second nucléotide sequence consists of 21 nucléotides.
68. The siNA molécule of embodiment 67, wherein 2’-O-methyl nucléotides are at positions 18-21 from the 5’ end of the second nucléotide sequence.
69. A short interfering nucleic acid (siNA) molécule comprising
264 (a) a sense strand comprising a first nucléotide sequence consisting of 17 to 23 nucléotides, wherein 2’-fluoro nucléotides are at positions 3, 7-9, 12 and 17 from the 5’ end of the first nucléotide sequence, and wherein 2’-O-methyl nucléotides are at positions 1, 2, 4-6, 10, 11, and 13-16 from the 5’ end of the first nucléotide sequence; and (b) an antisense strand comprising a second nucléotide sequence consisting of 17 to 23 nucléotides, wherein the nucléotides in the second nucléotide sequence are arranged in an altemating 1:3 modification pattern, and wherein 1 nucléotide is a 2’-fluoro nucléotide and 3 nucléotides are 2’-O-methyl nucléotides.
70. The siNA molécule of embodiment 69, wherein the first nucléotide sequence consists of 19 nucléotides.
71. The siNA molécule of embodiment 70, wherein 2’-O-methyl nucléotides are at positions 18 and 19 from the 5’ end of the first nucléotide sequence.
72. The siNA molécule according to any one of embodiments 69-71, wherein the second nucléotide sequence consists of 21 nucléotides.
73. The siNA molécule of embodiment 72, wherein 2’-O-methyl nucléotides are at positions 19-21 from the 5’ end of the second nucléotide sequence.
74. The siRNA molécule according to any one of embodiments 69-73, wherein the altemating 1:3 modification pattern occurs 2-5 times.
75. The siRNA molécule according to any one of embodiments 69-74, wherein at least two of the altemating 1:3 modification pattern occur consecutively.
76. The siRNA molécule according to any one of embodiments 69-75, wherein at least two of the altemating 1:3 modification pattern occurs nonconsecutively.
77. The siNA according to any one of embodiments 69-76, wherein at least 1, 2, 3, 4, or 5 altemating 1:3 modification pattern begins at nucléotide position 2, 6, 10, 14, and/or 18 from the 5’ end of the antisense strand.
78. The siNA according to any one of embodiments 69-77, wherein at least one altemating 1:3 modification pattern begins at nucléotide position 2 from the 5’ end of the antisense strand.
265
79. The siNA according to any one of embodiments 69-78, wherein at least one altemating 1:3 modification pattern begins at nucléotide position 6 from the 5’ end of the antisense strand.
80. The siNA according to any one of embodiments 69-79, wherein at least one altemating 1:3 modification pattern begins at nucléotide position 10 from the 5’ end of the antisense strand.
81. The siNA according to any one of embodiments 69-80, wherein at least one altemating 1:3 modification pattern begins at nucléotide position 14 from the 5’ end of the antisense strand.
82. The siNA according to any one of embodiments 69-81, wherein at least one altemating 1:3 modification pattern begins at nucléotide position 18 from the 5’ end of the antisense strand.
83. A short interfering nucleic acid (siNA) molécule comprising (a) a sense strand comprising a first nucléotide sequence consisting of 17 to 23 nucléotides, wherein 2’-fluoro nucléotides are at positions 5 and 7-9 from the 5’ end of the first nucléotide sequence, and wherein 2’-O-methyl nucléotides are at positions 1-4, 6, and 10-17 from the 5’ end of the first nucléotide sequence; and (b) an antisense strand comprising a second nucléotide sequence consisting of 17 to 23 nucléotides, wherein the nucléotides in the second nucléotide sequence are arranged in an altemating 1:3 modification pattern, and wherein 1 nucléotide is a 2’-fluoro nucléotide and 3 nucléotides are 2’-O-methyl nucléotides.
84. The siNA molécule of embodiment 83, wherein the first nucléotide sequence consists of 19 nucléotides.
85. The siNA molécule of embodiment 84, wherein 2’-O-methyl nucléotides are at positions 18 and 19 from the 5’ end of the first nucléotide sequence.
86. The siNA molécule according to any one of embodiments 83-85, wherein the second nucléotide sequence consists of 21 nucléotides.
87. The siNA molécule of embodiment 86, wherein 2’-O-methyl nucléotides are at positions 19-21 from the 5’ end of the second nucléotide sequence.
88. The siRNA molécule according to any one of embodiments 83-87, wherein the altemating 1:3 modification pattern occurs 2-5 times.
89. The siRNA molécule according to any one of embodiments 83-88, wherein at least two of the altemating 1:3 modification pattern occur consecutively.
266
90. The siRNA molécule according to any one of embodiments 83-89, wherein at least two of the altemating 1:3 modification pattern occurs nonconsecutively.
91. The siNA according to any one of embodiments 83-90, wherein at least 1, 2, 3, 4, or 5 altemating 1:3 modification pattern begins at nucléotide position 2, 6, 10, 14, and/or 18 from the 5’ end of the antisense strand.
92. The siNA according to any one of embodiments 83-91, wherein at least one altemating 1:3 modification pattern begins at nucléotide position 2 from the 5’ end of the antisense strand.
93. The siNA according to any one of embodiments 83-92, wherein at least one altemating 1:3 modification pattern begins at nucléotide position 6 from the 5’ end of the antisense strand.
94. The siNA according to any one of embodiments 83-93, wherein at least one altemating 1:3 modification pattern begins at nucléotide position 10 from the 5’ end of the antisense strand.
95. The siNA according to any one of embodiments 83-94, wherein at least one altemating 1:3 modification pattern begins at nucléotide position 14 from the 5’ end of the antisense strand.
96. The siNA according to any one of embodiments 83-95, wherein at least one altemating 1:3 modification pattern begins at nucléotide position 18 from the 5’ end of the antisense strand.
97. A short interfering nucleic acid (siNA) molécule comprising (a) a sense strand comprising a first nucléotide sequence consisting of 17 to 23 nucléotides, wherein 2’-fluoro nucléotides are at positions 5 and 7-9 from the 5’ end of the first nucléotide sequence, and wherein 2’-O-methyl nucléotides are at positions 1-4, 6, and 10-17 from the 5’ end of the first nucléotide sequence; and (b) an antisense strand comprising a second nucléotide sequence consisting of 17 to 23 nucléotides, wherein the nucléotides in the second nucléotide sequence are arranged in an altemating 1:2 modification pattern, and wherein 1 nucléotide is a 2’-fluoro nucléotide and 2 nucléotides are 2’-(9-methyl nucléotides.
98. The siNA molécule of embodiment 97, wherein the first nucléotide sequence consists of 19 nucléotides.
99. The siNA molécule of embodiment 98, wherein 2’-(9-methyl nucléotides are at positions 18 and 19 from the 5’ end of the first nucléotide sequence.
267
100. The siNA molécule according to any one of embodiments 97-99, wherein the second nucléotide sequence consists of 21 nucléotides.
101. The siNA molécule of embodiment 100, wherein 2’-O-methyl nucléotides are at positions 18-21 from the 5’ end of the second nucléotide sequence.
102. The siRNA molécule according to any one of embodiments 97-101, wherein the altemating 1:2 modification pattern occurs 2-5 times.
103. The siRNA molécule according to any one of embodiments 97-102, wherein at least two of the altemating 1:2 modification pattern occur consecutively.
104. The siRNA molécule according to any one of embodiments 97-103, wherein at least two of the altemating 1:2 modification pattern occurs nonconsecutively.
105. The siNA according to any one of embodiments 97-104, wherein at least 1, 2, 3, 4, or 5 altemating 1:2 modification pattern begins at nucléotide position 2, 5, 8, 14, and/or 17 from the 5’ end of the antisense strand.
106. The siNA according to any one of embodiments 97-105, wherein at least one altemating 1:2 modification pattern begins at nucléotide position 2 from the 5’ end of the antisense strand.
107. The siNA according to any one of embodiments 97-106, wherein at least one altemating 1:2 modification pattern begins at nucléotide position 5 from the 5’ end of the antisense strand.
108. The siNA according to any one of embodiments 97-107, wherein at least one altemating 1:2 modification pattern begins at nucléotide position 8 from the 5’ end of the antisense strand.
109. The siNA according to any one of embodiments 74-85, wherein at least one altemating 1:2 modification pattern begins at nucléotide position 14 from the 5’ end of the antisense strand.
110. The siNA according to any one of embodiments 97-109, wherein at least one altemating 1:2 modification pattern begins at nucléotide position 17 from the 5’ end of the antisense strand.
111. A short interfering nucleic acid (siNA) molécule comprising (a) a sense strand comprising a first nucléotide sequence consisting of 17 to 23 nucléotides, wherein 2’-fluoro nucléotides are at positions 5 and 7-9 from the 5’ end of the first nucléotide sequence, and wherein 2’-O-methyl nucléotides are at positions 1-4, 6, and 10-17 from the 5’ end of the first nucléotide sequence; and
268 (b) an antisense strand comprising a second nucléotide sequence consisting of 17 to 23 nucléotides, wherein 2’-fluoro nucléotides are at positions 2, 6, 14, and 16 from the 5’ end of the second nucléotide sequence, and wherein 2’-O-methyl nucléotides are at positions 1, 3-5, 7-13, 15, and 17 from the 5’ end the second nucléotide sequence.
112. The siNA molécule of embodiment 111, wherein the first nucléotide sequence consists of 19 nucléotides.
113. The siNA molécule of embodiment 112, wherein 2’-O-methyl nucléotides are at positions 18 and 19 from the 5’ end of the first nucléotide sequence.
114. The siNA molécule according to any one of embodiments 111-113, wherein the second nucléotide sequence consists of 21 nucléotides.
115. The siNA molécule of embodiment 114, wherein 2’-O-methyl nucléotides are at positions 18-21 from the 5’ end of the second nucléotide sequence.
116. A short interfering nucleic acid (siNA) molécule comprising:
(a) a sense strand comprising a first nucléotide sequence consisting of 17 to 23 nucléotides, wherein 2’-fluoro nucléotides are at positions 5, 9-11, and 14 from the 5’ end of the first nucléotide sequence, and wherein 2’-O-methyl nucléotides are at positions 1-4, 6-8, and 12-17 from the 5’ end of the first nucléotide sequence; and (b) an antisense strand comprising a second nucléotide sequence consisting of 17 to 23 nucléotides, wherein 2’-fluoro nucléotides are at positions 2 and 14 from the 5’ end of the second nucléotide sequence, and wherein 2’-(9-methyl nucléotides are at positions 1, 3-13, and 15-17 from the 5’ end the second nucléotide sequence.
117. The siNA molécule of embodiment 116, wherein the first nucléotide sequence consists of 21 nucléotides.
118. The siNA molécule of embodiment 117, wherein 2’-(9-methyl nucléotides are at positions 18-21 from the 5’ end of the first nucléotide sequence.
119. The siNA molécule according to any one of embodiments 116-118, wherein the second nucléotide sequence consists of 23 nucléotides.
269
120. The siNA molécule of embodiment 119, wherein 2’-O-methyl nucléotides are at positions 18-23 from the 5’ end of the second nucléotide sequence.
121. The siNA according to any preceding embodiment, wherein the sense strand fùrther comprises TT sequence adjacent to the first nucléotide sequence.
122. The siNA according to any preceding embodiment, wherein the sense strand further comprises at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15 or more phosphorothioate intemucleoside linkages.
123. The siNA of embodiment 122, wherein at least one phosphorothioate intemucleoside linkage is between the nucléotides at positions 1 and 2 from the 5’ end of the first nucléotide sequence.
124. The siNA of embodiment 122 or 123, wherein at least one phosphorothioate intemucleoside linkage is between the nucléotides at positions 2 and 3 from the 5’ end of the first nucléotide sequence.
125. The siNA according to any preceding embodiment, wherein the antisense strand further comprises TT sequence adjacent to the second hucleotide sequence.
126. The siNA according to any preceding embodiment, wherein the antisense strand further comprises at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15 or more phosphorothioate intemucleoside linkages.
127. The siNA of embodiment 126, wherein at least one phosphorothioate intemucleoside linkage is between the nucléotides at positions 1 and 2 from the 5’ end of the second nucléotide sequence.
128. The siNA of embodiment 126 or 127, wherein at least one phosphorothioate intemucleoside linkage is between the nucléotides at positions 2 and 3 from the 5’ end of the second nucléotide sequence.
129. The siNA of any one of embodiments 126-128, wherein at least one phosphorothioate intemucleoside linkage is between the nucléotides at positions 1 and 2 from the 3 ’ end of the second nucléotide sequence.
270
130. The siNA of any one of embodiments 126-129, wherein at least one phosphorothioate intemucleoside linkage is between the nucléotides at positions 2 and 3 from the 3’ end of the second nucléotide sequence.
131. The siNA according to any preceding embodiment, wherein the first nucléotide from the 5 5’ end of the first nucléotide sequence comprises a 5’ stabilizing end cap.
132. The siNA according to any preceding embodiment, wherein the first nucléotide from the 5’ end of the second nucléotide sequence comprises a 5’ stabilizing end cap.
133. The siNA according to any preceding embodiment, wherein the first nucléotide from the 5’ end of the first nucléotide sequence comprises a phosphorylation blocker.
134. The siNA according to any preceding embodiment, wherein the first nucléotide from the
5’ end of the second nucléotide sequence comprises a phosphorylation blocker.
135. The siNA according to any preceding embodiment, wherein the first nucléotide sequence or second nucléotide sequence comprises at least one modified nucléotide selected from
or alkyl (or AmNA(N-Me)) when R is alkyl);
(AmNA), where R is H (GuNA); and
GuNA(N-R), R = Me, Et, iPr, tBu, wherein B is a nucleobase.
136. A short-interfering nucleic acid (siNA) molécule comprising:
(a) a phosphorylation blocker of Formula (IV):
wherein
271
R1 is a nucleobase,
R4 is-O-R30 or-NR3IR32,
R30 is Ci-C8 substituted or unsubstituted alkyl; and
R31 and R32 together with the nitrogen to which they are attached form a substituted or unsubstituted heterocyclic ring; and (b) a short interfering nucleic acid (siNA).
137. A short-interfering nucleic acid (siNA) molécule comprising:
(a) a 5’-stabilized end cap of Formula (la): wherein
R1 is a nucleobase, aryl, heteroaryl, or H, ry°vr1
R20' VJ
0' t>CH3
Οχ zo , Vn-s'
R2 is H fi 0, .0
H o' o /0 ^SN H
Oxx /0 9 HO .s 9 ^S^PX-OH X A
OH \ Ο ^OH
O. .OH ^PxOH ox och3
Ox zOCD3
H ,N, / i S
O O ?
O , -CH=CD-Z,-CD=CH-Z,-CD=CD-Z,
-(CR21R22)n-Z, or -(C2-Cé alkenylene)-Z and R20 is hydrogen; or
R2 and R20 together form a 3- to 7-membered carbocyclic ring substituted with (CR21R22)n-Z or -(C2-C6 alkenylene)-Z;
n is 1, 2, 3, or 4;
Z is -ONR23R24, -OP(O)OH(CH2)mCO2R23, -OP(S)OH(CH2)mCO2R23, -P(O)(OH)2, P(O)(OH)(OCH3), -P(O)(OH)(OCD3), -SO2(CH2)mP(O)(OH)2, -so2nr23r25, -nr23r24, R21 and R22 are independently hydrogen or Ci-Cô alkyl; R21 and R22 together form an oxo group;
R23 is hydrogen or Ci-Cô alkyl;
R24 is -SO2R25 or -C(O)R25; or
R23 and R24 together with the nitrogen to which they are attached form a substituted or unsubstituted heterocyclic ring;
R25 is Ci-Cô alkyl; and m is 1, 2, 3, or 4; and
272 (b) a short interfering nucleic acid (siNA).
138. A short-interfering nucleic acid (siNA) molécule comprising:
(a) a 5’-stabilized end cap of Formula (Ib):
wherein
R1 is a nucleobase, aryl, heteroaryl, or H,
/0 il HOV .S 9 Ox OH Os OCH3
JS^Pç-OH
OH X O OH \ OH OH ? 5 5
-CH=CD-Z, -CD=CH-Z, -CD=CD-Z,
-(CR21R22)n-Z, or -(C2-C6 alkenylene)-Z and R20 is hydrogen; or
R2 and R20 together form a 3- to 7-membered carbocyclic ring substituted with (CR21R22)n-Z or -(C2-C6 alkenylene)-Z;
n is 1, 2, 3, or 4;
Z is -ONR23R24, -OP(O)OH(CH2)mCO2R23, -OP(S)OH(CH2)mCO2R23, -P(O)(OH)2, P(O)(OH)(OCH3), -P(O)(OH)(OCD3), -SO2(CH2)mP(O)(OH)2, -so2nr23r25, -nr23r24,
R21 and R22 are independently hydrogen or Ci-Cô alkyl; R21 and R22 together form an oxo group;
R23 is hydrogen or Ci-Cô alkyl;
R24 is -SO2R25 or -C(O)R25; or
R23 and R24 together with the nitrogen to which they are attached form a substituted or unsubstituted heterocyclic ring;
R25 is Ci-Cô alkyl; and m is 1, 2, 3, or 4; and (b) a short interfering nucleic acid (siNA).
139. A short-interfering nucleic acid (siNA) molécule comprising:
273 (a) a 5’-stabilized end cap selected from the group consisting of Formula (1) to Formula (15), Formula (9X) to Formula (12X), and Formula (9Y) to Formula (12Y):
Formula (5) Formula (6) Formula (7)
Formula (8)
Formula (9) Formula (9X) Formula (9Y)
Formula (11 Y)
Formula (11)
Formula (11X)
Formula (12)
Formula (12X)
Formula (12Y)
274
Formula (13) Formula (14) Formula (15) wherein R1 is a nucleobase, aryl, heteroaryl, or H; and (b) a short interfering nucleic acid (siNA).
140. A short-interfering nucleic acid (siNA) molécule comprising:
(a) a 5’-stabilized end cap selected from the group consisting of Formulas (1A)-(15A),
Formulas (9B)-(12B), Formulas (9AX)-(12AX), Formulas (9AY)-(12AY), Formulas (9BX)-(12BX), and Formulas (9BY)-(12BY):
cf och3 och3 och3
Formula (5A) Formula (6A) Formula (7A)
0 //^ nh FJj' hohcx<S H3co.p<f 0 / \ ' och3 οχ och3 Formula (8A) Formula (9A) H3CCV°/^/°\ D3C% HO Λ__/ HO \ / HO ci och3 ci ôch3 x y Formula (9B) Formula (9BX) Q O d3C0>z-° ho' '^\_j cT och3 ci och3 Formula (9AX) Formula (9AY) cT îoch3 Formula (9BY)
275
Formula (10A)
Formula (10AX)
Formula (10AY)
Formula (10B)
Formula (10BX)
Formula (10BY)
Formula (11 A)
Formula (11AX)
Formula (11AY)
Formula (11 B)
Formula (11 BY)
Formula (11BX)
Formula (12B)
Formula (12BX)
Formula (12BY)
276
Formula (13A) Formula (14A) Formula (15A) anj (b) a short interfering nucleic acid (siNA).
141. The siNA molécule according to any one of embodiments 136-140, wherein the siNA comprises the sense strand of any one of embodiments 1-135.
142. The siNA molécule according to any one of embodiments 136-141, wherein the siNA comprises the antisense strand of any one of embodiments 1-135.
143. A short interfering nucleic acid (siNA) molécule comprising:
(a) a sense strand comprising a first nucléotide sequence that is at least about 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% identical to an RNA corresponding to a target gene, wherein the first nucléotide sequence comprises a nucléotide sequence of any one SEQ ID NOs: 1-56, 103-158, and 205-260; and (b) an antisense strand comprising a second nucléotide sequence that is at least about 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% complementary to the RNA corresponding to the target gene, wherein the second nucléotide sequence comprises a nucléotide sequence of any one of SEQ ID NOs: 57-102, 159-204, and 261-306.
144. A interfering nucleic acid (siNA) molécule comprising:
(a) a sense strand comprising a nucléotide sequence of any one of SEQ ID NOs: 307362 and 415-444; and (b) an antisense strand comprising a nucléotide sequence of any one of SEQ ID NOs: 363-409, 445-533, and 536-539.
145. The siNA according to any one of embodiments 1-132, 135, and 137-144, wherein the siNA further comprises a phosphorylation blocker.
277
146. The siNA according to any one of embodiments 16, 133, 134, and 145, wherein the R4A^°yR1 phosphorylation blocker has the structure of Formula (IV): ~~L~ , wherein
R1 is a nucleobase,
R4 is -O-R30 or -NR31R32, R30 is Ci-Cs substituted or unsubstituted alkyl; and
R31 and R32 together with the nitrogen to which they are attached form a substituted or unsubstituted heterocyclic ring.
147. The siNA of embodiment 136 or 146, wherein R4 is -OCH3 or -N(CH2CH2)2O.
148. The siNA according to any one of embodiments 16, 133, 134, 136, and 145-147, wherein the phosphorylation blocker is attached to the 5’ end of the sense strand.
149. The siNA of embodiment 148, wherein the phosphorylation blocker is attached to the 5’ end of the sense strand via one or more linkers independently selected from a phosphodiester linker, phosphorothioate linker, and phosphorodithioate linker.
150. The siNA according to any one of embodiments 16, 133, 134, 136, and 145-147, wherein the phosphorylation blocker is attached to the 3’ end of the sense strand.
151. The siNA of embodiment 150, wherein the phosphorylation blocker is attached to the 3 ’ end of the sense strand via one or more linkers independently selected from a phosphodiester linker, phosphorothioate linker, and phosphorodithioate linker.
152. The siNA according to any one of embodiments 16, 133, 134, 136, and 145-147, wherein the phosphorylation blocker is attached to the 5’ end of the antisense strand.
153. The siNA of embodiment 152, wherein the phosphorylation blocker is attached to the 5’ end of the antisense strand via one or more linkers independently selected from a phosphodiester linker, phosphorothioate linker, and phosphorodithioate linker.
154. The siNA according to any one of embodiments 16, 133, 134, 136, and 144-147, wherein the phosphorylation blocker is attached to the 3 ’ end of the antisense strand.
278
155. The siNA of embodiment 154, wherein the phosphorylation blocker is attached to the 3 ’ end of the antisense strand via one or more linkers independently selected from a phosphodiester linker, phosphorothioate linker, and phosphorodithioate linker.
156. The siNA according to any preceding embodiment, wherein the siNA further comprises a galactosamine.
157. The siNA of embodiment 16 or 156, wherein the galactosamine is N-acetylgalactosamine (GalNAc) of Formula (VII):
R = OH or SH wherein each n is independently 1 or 2.
158. The siNA of embodiment 16 or 156, wherein the galactosamine is N-acetylgalactosamine (GalNAc) of Formula (VI): wherein
m is 1, 2, 3, 4, or 5;
each n is independently 1 or 2;
p is 0 or 1 ;
each R is independently H;
each Y is independently selected from -O-P(=O)(SH)-, -O-P(=O)(O)-, -O-P(=O)(OH)-, and O-P(S)S-;
Z is H or a second protecting group;
either L is a linker or L and Y in combination are a linker; and
A is H, OH, a third protecting group, an activated group, or an oligonucleotide.
279
159. The siNA of embodiment 158, wherein A is an oligonucleotide. ·
160. The siNA of embodiment 158, wherein A is 1-2 oligonucleotides.
161. The siNA of any one of embodiments 158-160, wherein the oligonucleotide is dTdT.
162. The siNA according to any one of embodiments 16 and 156-161, wherein the galactosamine is attached to the 3’ end of the sense strand.
163. The siNA of embodiment 162, wherein the galactosamine is attached to the 3’ end of the sense strand via one or more linkers independently selected from a phosphodiester linker, phosphorothioate linker, or phosphorodithioate linker.
164. The siNA according to any one of embodiments 16 and 156-161, wherein the galactosamine is attached to the 5’ end of the sense strand.
165. The siNA of embodiment 164, wherein the galactosamine is attached to the 5’ end of the sense strand via one or more linkers independently selected from a phosphodiester linker, phosphorothioate linker, or phosphorodithioate linker.
166. The siNA according to any one of embodiments 16 and 156-161, wherein the galactosamine is attached to the 3’ end of the antisense strand.
167. The siNA of embodiment 166, wherein the galactosamine is attached to the 3’ end of the atnisense strand via one or more linkers independently selected from a phosphodiester linker, phosphorothioate linker, or phosphorodithioate linker.
168. The siNA according to any one of embodiments 16 and 156-161, wherein the galactosamine is attached to the 5’ end of the antisense strand.
169. The siNA of embodiment 168, wherein the galactosamine is attached to the 5’ end of the atnisense strand via one or more linkers independently selected from a phosphodiester linker, phosphorothioate linker, or phosphorodithioate linker.
170. The siNA according to any one of embodiments 1-130, 133-136, and 139-169, wherein the siNA further comprises a 5’-stabilized end cap.
171. The siNA according to any one of embodiments 16, 131, 132, and 170, wherein the 5’stabilized end cap is a 5’ vinyl phosphonate or deuterated 5’ vinyl phosphonate.
280
172. The siNA according to any one of embodiments 16, 131, 132, and 170, wherein the 5’- stabilized end cap has the structure of Formula (la):
wherein
R1 is a nucleobase, aryl, heteroaryl, or H,
/0 9 HO ZS 9 Ox OH Ox OCH3 Ox ocd3
JsQ^pcoh
OH Y O OH V OH Y OH Y OH
O , -CH=CD-Z, -CD=CH-Z, -CD=CD-Z, -(CR21R22)n-Z, or -(C2-C6 alkenylene)-Z and R20 is hydrogen; or
R2 and R20 together form a 3- to 7-membered carbocyclic ring substituted with -(CR21R22)n-Z or
-(C2-C6 alkenylene)-Z;
nis 1, 2, 3, or 4;
Z is -ONR23R24, -OP(O)OH(CH2)mCO2R23, -OP(S)OH(CH2)mCO2R23, -P(O)(OH)2, P(O)(OH)(OCH3), -P(O)(OH)(OCD3), -SO2(CH2)mP(O)(OH)2, -SO2NR23R25, -NR23R24, or NR23SO2R24;
R21 and R22 either are independently hydrogen or Ci-Cô alkyl, or R21 and R22 together form an oxo group;
R23 is hydrogen or Ci-Ce alkyl;
R24 is -SO2R25 or -C(O)R25; or
R23 and R24 together with the nitrogen to which they are attached form a substituted or unsubstituted heterocyclic ring;
R25 is Ci-Cô alkyl; and m is 1, 2, 3, or 4.
173. The siNA according to any one of embodiments 131, 132, and 170, wherein the 5’- stabilized end cap has the structure of Formula (Ib):
wherein
281
R1 is a nucleobase, aryl, heteroaryl, or H,
O
II
PX-OH
OH
HO, Z/S 9 O,, ZOH
ΧΑ/Χόη V^%h
O, OCH3 O, OCD3
Y OH Y OH 5
, -CH=CD-Z, -CD=CH-Z, -CD=CD-Z, -(CR21R22)n-Z, or-(C2-C6 alkenylene)-Z and R20 is hydrogen; or
R2 and R20 together form a 3- to 7-membered carbocyclic ring substituted with -(CR21R22)n-Z or
-(C2-Cô alkenylene)-Z;
n is 1, 2, 3, or 4;
Z is -ONR23R24, -OP(O)OH(CH2)mCO2R23, -OP(S)OH(CH2)raCO2R23, -P(O)(OH)2, P(O)(OH)(OCH3), -P(O)(OH)(OCD3), -SO2(CH2)mP(O)(OH)2, -SO2NR23R25, -NR23R24, orNR23SO2R24;
R21 and R22 either are independently hydrogen or Ci-Cô alkyl, or R21 and R22 together form an oxo group;
R23 is hydrogen or Ci-Cô alkyl;
R24 is -SO2R25 or-C(O)R25; or
R23 and R24 together with the nitrogen to which they are attached form a substituted or unsubstituted heterocyclic ring;
R25 is Ci-Cô alkyl; and m is 1, 2, 3, or 4.
174. The siNA of embodiment 172 or 173, wherein R1 is an aryl.
175. The siNA of embodiment 174, wherein the aryl is a phenyl.
176. The siNA according to any one of embodiments 16, 131, 132, and 170, wherein the 5’stabilized end cap is selected from the group consisting of Formula (1) to Formula (15), Formula (9X) to Formula (12X), and Formula (9Y) to Formula (12Y):
282
Formula (10) Formula (10X) Formula (1 ΟΥ)
Formula (11) Formula (11X) Formula (11Y)
Formula (12)
Formula (12X) Formula (12Y)
Formula (13) Formula (14) Formula (15) , wherein R1 is a nucleobase, aryl, heteroaryl, or H.
283
177. The siNA according to any one of embodiments 16, 131, 132, and 170, wherein the 5’stabilized end cap is selected from the group consisting of Formulas (1A)-(15A), Formulas (9B)(12B), Formulas (9AX)-(12AX), Formulas (9AY)-(12AY), Formulas (9BX)-(12BX), and Formulas (9BY)-(12BY):
Formula (9AX)
Formula (8A)
Formula (9A)
Formula (9AY) D 3COyO n.
HO /
O\ OCH3
Formula (9BY)
Formula (9BX)
Formula (9B)
Formula (10AX)
284
Formula (10B)
Formula (10BX)
Formula (10BY)
Formula (11 A)
Formula (11AX)
Formula (11AY)
Formula (11 B) Formula (11BX) Formula (11 BY)
Formula (12A) Formula (12AX) Formula (12AY)
Formula (12BY)
Formula (12B) Formula (12BX)
285
178. The siNA according to any one of embodiments 131, 132, and 170, wherein the 5’- stabilized end cap has the structure of Formula (le):
, wherein
R1 is a nucleobase, aryl, heteroaryl, or H,
<P 9 HO. ZS 9 Ox OH Ox OCH3 Ox ocd3
Js^px-oh . ss/XpC ï/XC
OH Y O OH OH Y OH Y OH
5 5 5
o , -CH=CD-Z, -CD=CH-Z, -CD=CD-Z, -(CR21 R22)n-Z, or -(C2-C6 alkenylene)-Z and R20 is hydrogen; or
R2 and R20 together form a 3- to 7-membered carbocyclic ring substituted with -(CR21R22)n-Z or
-(C2-Cô alkenylene)-Z;
n is 1, 2, 3, or 4;
Z is -ONR23R24, -OP(O)OH(CH2)mCO2R23, -OP(S)OH(CH2)mCO2R23, -P(O)(OH)2, P(O)(OH)(OCH3), -P(O)(OH)(OCD3), -SO2(CH2)mP(O)(OH)2, -SO2NR23R25, -NR23R24, orNR23SO2R24;
R21 and R22 either are independently hydrogen or Ci-Cô alkyl, or R21 and R22 together form an oxo group;
R23 is hydrogen or Ci-Cô alkyl;
R24 is -SO2R25 or -C(O)R25; or
R23 and R24 together with the nitrogen to which they are attached form a substituted or unsubstituted heterocyclic ring;
R25 is Ci-Cô alkyl; and m is 1, 2, 3, or 4.
179. The siNA of embodiment 178, wherein R1 is an aryl.
180. The siNA of embodiment 179, wherein the aryl is a phenyl.
181. The siNA according to any one of embodiments 16, 131, 132, and 170, wherein the 5’stabilized end cap is selected from the group consisting of Formula (21) to Formula (35):
286
Formula (28) Formula (29) Formula (30)
Formula (31) Formula (32) Formula (33)
Formula (34) Formula (35) , wherein R1 is a nucleobase, aryl, heteroaryl, or
H.
182. The siNA according to any one of embodiments 16, 131, 132, and 170, wherein the 5’stabilized end cap is selected from the group consisting of Formulas (21A)-(35A), Formulas (29B)-(32B), Formulas (29AX)-(32AX), Formulas (29AY)-(32AY), Formulas (29BX)-(32BX), io and Formulas (29BY)-(32BY):
287
Formula (22A)
Formula (23A)
Formula (24A)
Formula (27A)
Formula (28A) Formula (29A)
Formula (29AX) Formula (29AY)
Formula (29B)
Formula (29BX)
Formula (29BY)
Formula (30A)
Formula (30AX)
Formula (30AY)
288
Formula (31A)
Formula (31AY)
Formula (31 B)
Formula (32A)
183. The siNA according to any one of embodiments 1-182, wherein the antisense strand comprises at least one thermally destabilizing nucléotide selected from:
289
184. The siNA according to any one of embodiments 1-182, wherein the sense strand comprises at least one thermally destabilizing nucléotide selected from:
185. The siNA according to any one of embodiments 1-182, wherein the first nucléotide sequence comprises at least one thermally destabilizing nucléotide selected from:
290
186. The siNA according to any one of embodiments 1-182, wherein the second nucléotide sequence comprises at least one thermally destabilizing nucléotide selected from:
187. The siNA according to any one of embodiments 16, 131, 132, and 170-186, wherein the
5’-stabilized end cap is attached to the 5’ end of the antisense strand.
188. The siNA of embodiment 187, wherein the 5’-stabilized end cap is attached to the 5’ end of the antisense strand via one or more linkers independently selected from a phosphodiester linker, phosphorothioate linker, or phosphorodithioate linker.
189. The siNA according to any one of embodiments 16, 131, 132, and 170-186, wherein the 5’-stabilized end cap is attached to the 5’ end of the sense strand.
190. The siNA of embodiment 189, wherein the 5’-stabilized end cap is attached to the 5’ end of the sense strand via one or more linkers independently selected from a phosphodiester linker, phosphorothioate linker, or phosphorodithioate linker.
191. The siNA according to any preceding embodiment, wherein the target gene is a viral gene.
192. The siNA of embodiment 191, wherein the viral gene is from a DNA virus.
193. The siNA of embodiment 192, wherein the DNA virus is a double-stranded DNA (dsDNA) virus.
194. The siNA of embodiment 193, wherein the dsDNA virus is a hepadnavirus.
195. The siNA of embodiment 194, wherein the hepadnavirus is a hepatitis B virus (HBV).
291
196. The siNA of embodiment 195, wherein the HBV is selected from HBV génotypes A-J.
197. The siNA of embodiment 195 or 196, wherein the target gene is selected from the S gene or X gene of the HBV.
198. The siNA according to any one of embodiments 1-197, wherein the second nucléotide sequence is at least about 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% complementary to 15 to 30 nucléotides within positions 200-720 or 1100-1700 of SEQ ID NO: 410.
199. The siNA according to any one of embodiments 1-197, wherein the second nucléotide sequence is at least about 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% complementary to 15 to 30 nucléotides within positions 200-280, 300-445, 460-510, 650-720, 1170-1220, 1250-1300, or 1550-1630 of SEQ ID NO: 410.
200. The siNA according to any one of embodiments 1-197, wherein the second nucléotide sequence is at least about 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% complementary to 15 to 30 nucléotides within positions 200-230, 250-280, 300-330, 370-400, 405-445, 460-500, 670-700, 1180-1210, 1260-1295, 1520-1550, or 1570-1610 of SEQ ID NO: 410.
201. The siNA according to any one of embodiments 1-197, wherein the second nucléotide sequence is at least about 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% complementary to 15 to 30 nucléotides starting at position 203, 206, 254, 305, 375, 409, 412, 415, 416, 419, 462, 466, 467, 674, 676, 1182, 1262, 1263, 1268, 1526, 1577, 1578, 1580, 1581, 1583, or 1584 of SEQ ID NO: 410.
202. The siNA according to any one of embodiments 1-201, wherein the first nucléotide sequence is at least about 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% identical to 15 to 30 nucléotides within positions 200-720 or 1100-1700 of SEQ ID NO: 410.
203. The siNA according to any one of embodiments 1-201, wherein the first nucléotide sequence is at least about 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% identical to 15 to 30 nucléotides within positions 200-280, 300-445, 460-510, 650-720, 1170-1220, 1250-1300, or 1550-1630 of SEQ ID NO: 410.
204. The siNA according to any one of embodiments 1-201, wherein the first nucléotide sequence is at least about 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% identical to 15
292 to 30 nucléotides within positions 200-230, 250-280, 300-330, 370-400, 405-445, 460-500, 670700, 1180-1210, 1260-1295, 1520-1550, or 1570-1610 ofSEQ IDNO:410.
205. The siNA according to any one of embodiments 1-201, wherein the first nucléotide sequence is at least about 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% identical to 15 to 30 nucléotides starting at position 203, 206, 254, 305, 375, 409, 412, 415, 416, 419, 462, 466, 467, 674, 676, 1182, 1262, 1263, 1268, 1526, 1577, 1578, 1580, 1581, 1583, or 1584 ofSEQ ID NO: 410.
206. The siNA according to any preceding embodiment, wherein the first nucléotide sequence comprises a nucléotide sequence of any one SEQ ID NOs: 1-56, 103-158, and 205-260.
207. The siNA according to any preceding embodiment, wherein the second nucléotide sequence comprises a nucléotide sequence of any one ofSEQ ID NOs: 57-102, 159-204, and 261-306.
208. The siNA according to any preceding embodiment, wherein the sense strand comprises a nucléotide sequence of any one of SEQ ID NOs: 307-362 and 415-444.
209. The siNA according to any preceding embodiment, wherein the antisense strand comprises a nucléotide sequence of any one of SEQ ID NOs: 363-409, 445-533, and 536-539.
210. The siNA according to any preceding embodiment, wherein at least one end of the siNA is a blunt end.
211. The siNA according to any preceding embodiment, wherein at least one end of the siNA comprises an overhang, wherein the overhang comprises at least one nucléotide.
212. The siNA according to any one of embodiments 1-209, wherein both ends of the siNA comprise an overhang, wherein the overhang comprises at least one nucléotide.
213. The siNA according to any preceding embodiment, wherein the siNA is selected from dssiNA-001 to ds-siNA-0178.
214. The siNA according to any preceding embodiment, wherein at least one 2’-fluoro nucléotide or 2’-O-methyl nucléotide is a 2’-fluoro or 2-O-methyl nucléotide mimic of Formula (V):
293
R1 is independently a nucleobase, aryl, heteroaryl, or H, Q1 and Q2 are independently S or
O,
R5 is independently-OCD3, -F, or-OCH3, and
R6 and R7 are independently H, D, or CD3.
215. The siNA of embodiment 214, wherein the 2’-fluoro or 2’-O-methyl nucléotide mimic is a nucléotide mimic of Formula (16) - Formula (20):
Formula (16)
S'' R2
Formula (17)
Formula (18) Formula (19)
Formula (20) wherein R1 is a nucleobase and R2 is independently F or -OCH3.
216. The siNA according to any preceding embodiment, wherein at least one 2’-fluoro nucléotide is a 2’-fluoro nucléotide mimic.
217. The siNA according to embodiment 216, wherein at least one 2’-fluoro nucléotide on the antisense strand or the second nucléotide sequence is a 2’-fluoro nucléotide mimic.
218. The siNA according to embodiment 216 or 217, wherein the nucléotide at position 2 from the 5’ end of the antisense strand or the second nucléotide sequence is a 2’-fluoro nucléotide mimic.
219. The siNA according to any one of embodiments 216-218, wherein the nucléotide at position 5 from the 5’ end of the antisense strand or the second nucléotide sequence is a 2’-fluoro nucléotide mimic.
220. The siNA according to any one of embodiments 216-219, wherein the nucléotide at position 6 from the 5’ end of the antisense strand or the second nucléotide sequence is a 2’-fluoro nucléotide mimic.
294
221. The siNA according any one of embodiments 216-220, wherein the nucléotide at position from the 5’ end of the antisense strand or the second nucléotide sequence is a 2’-fluoro nucléotide mimic.
222. The siNA according to any one of embodiments 216-221, wherein the nucléotide at position 10 from the 5’ end of the antisense strand or the second nucléotide sequence is a 2’fluoro nucléotide mimic.
223. The siNA according to any one of embodiments 216-222, wherein the nucléotide at position 14 from the 5’ end of the antisense strand or the second nucléotide sequence is a 2’fluoro nucléotide mimic.
224. The siNA according to any one of embodiments 216-223, wherein the nucléotide at position 16 from the 5’ end of the antisense strand or the second nucléotide sequence is a 2’fluoro nucléotide mimic.
225. The siNA according to any one of embodiments 216-224, wherein the nucléotide at position 17 from the 5’ end of the antisense strand or the second nucléotide sequence is a 2’fluoro nucléotide mimic.
226. The siNA according to any one of embodiments 216-225, wherein at least one 2’-fluoro nucléotide on the sense strand or the first nucléotide sequence is a 2’-fluoro nucléotide mimic.
227. The siNA according to any one of embodiments 216-226, wherein the nucléotide at position 3 from the 5’ end of the sense strand or the first nucléotide sequence is a 2’-fluoro nucléotide mimic.
228. The siNA according to any one of embodiments 216-227, wherein the nucléotide at position 5 from the 5’ end of the sense strand or the first nucléotide sequence is a 2’-fluoro nucléotide mimic.
229. The siNA according to any one of embodiments 216-228, wherein the nucléotide at position 7 from the 5’ end of the sense strand or the first nucléotide sequence is a 2’-fluoro nucléotide mimic.
230. The siNA according to any one of embodiments 216-229, wherein the nucléotide at position 8 from the 5’ end of the sense strand or the first nucléotide sequence is a 2’-fluoro nucléotide mimic.
295
231. The siNA according to any one of embodiments 216-230, wherein the nucléotide at position 9 from the 5’ end of the sense strand or the first nucléotide sequence is a 2’-fluoro nucléotide mimic.
232. The siNA according to any one of embodiments 216-231, wherein the nucléotide at position 10 from the 5’ end of the sense strand or the first nucléotide sequence is a 2’-fluoro nucléotide mimic.
233. The siNA according to any one of embodiments 216-232, wherein the nucléotide at position 11 from the 5’ end of the sense strand or the first nucléotide sequence is a 2’-fluoro nucléotide mimic.
234. The siNA according to any one of embodiments 216-233, wherein the nucléotide at position 12 from the 5’ end of the sense strand or the first nucléotide sequence is a 2’-fluoro nucléotide mimic. '
235. The siNA according to any one of embodiments 216-234, wherein the nucléotide at position 14 from the 5’ end of the sense strand or the first nucléotide sequence is a 2’-fluoro nucléotide mimic.
236. The siNA according to any one of embodiments 216-235, wherein the nucléotide at position 17 from the 5’ end of the sense strand or the first nucléotide sequence is a 2’-fluoro nucléotide mimic.
237. The siNA according to any one of embodiments 216-236, wherein at least 1, 2, 3, 4, 5, 6,
or more 2’-fluoro nucléotide mimics is a f4P nucléotide (
238. The siNA according to any one of embodiments 216-237, wherein less than or equal to
10, 9, 8, 7, 6, 5, 4, 3, or 2 2’-fluoro nucléotide mimics is a f4P nucléotide (
296
239. The siNA according to any one of embodiments 216-238, wherein 1, 2, 3, 4, 5, 6, or more
2’-fluoro nucléotide mimics is a f2P nucléotide (
240. The siNA according to any one of embodiments 216-239, wherein less than or equal to
10, 9, 8, 7, 6, 5, 4, 3, or 2 2’-fluoro nucléotide mimics is a f2P nucléotide ( ).
241. The siNA according to any one of embodiments 216-240, wherein 1, 2, 3,. 4, 5, 6, or more nh2
2’-fluoro nucléotide mimics is a fX nucléotide ( ).
242. The siNA according to any one of embodiments 216-241, wherein less than or equal to
o' F
10, 9, 8, 7, 6, 5, 4, 3, or 2 2’-fluoro nucléotide mimics is a fX nucléotide ( ).
243. The siNA according to any preceding embodiment, wherein the first nucléotide from the 10 5’ end of the sense strand or first nucléotide sequence is a d2vd3 nucléotide (
297
244. The siNA according to any preceding embodiment, wherein the first nucléotide from the
3’ end of the sense strand or first nucléotide sequence is a d2vd3 nucléotide (
245. The siNA according to any preceding embodiment, wherein the first nucléotide from the
5’ end of the antisense strand or second nucléotide sequence is a d2vd3 nucléotide (
246. The siNA according to any preceding embodiment, wherein the first nucléotide from the ’ end of the antisense strand or second nucléotide sequence is a d2vd3 nucléotide (
io 247. A composition comprising the siNA according to any one of embodiments 1-246.
248. A composition comprising 2, 3, 4, 5, 6, 7, 8, 9, 10 or more siNAs according to any one of embodiments 1-246.
249. The composition of embodiment 248, wherein at least 1, 2, 3, 4, 5, or more siNAs target an S gene of HBV.
250. The composition of embodiment 248 or 249, wherein at least 1, 2, 3, 4, 5, or more siNAs target an X gene of HBV.
251. The composition according to any one of embodiments 247-250, further comprising an additional HBV treatment agent.
298
252. The composition of embodiment 251, wherein the additional HBV treatment agent is selected from a nucléotide analog, nucleoside analog, a capsid assembly modulator (CAM), a recombinant interferon, an entry inhibitor, a small molécule immunomodulator and oligonucleotide therapy.
253. The composition of embodiment 252, wherein the oligonucleotide therapy is an additional siNA.
254. The composition of embodiment 253, wherein the additional siNA is selected from any of ds-siNA-001 to ds-siNA-0178.
255. The composition of embodiment 252, wherein the oligonucleotide therapy is an antisense oligonucleotide (ASO), NAPs, or STOPS™
256. The composition of embodiment 255, wherein the ASO is ASO 1 or ASO 2.
257. The composition of embodiment 251 or 252, wherein the additional HBV treatment agent is selected from HBV STOPS™ ALG-010133, HBV CAM ALG-000184, ASO 1, recombinant interferon alpha 2b, IFN-a, PEG-IFN-a-2a, lamivudine, telbivudine, adefovir dipivoxil, clevudine, entecavir, tenofovir alafenamide, tenofovir disoproxil, NVR3-778, BAY41-4109, JNJ-632, JNJ-3989 (ARO-HBV), RG6004, GSK3228836, REP-2139, REP-2165, AB-729, VIR2218, RG6346 (DCR-HBVS), JNJ-6379, GLS4, ABI-HO731, JNJ-440, NZ-4, RG7907, EDP514, AB-423, AB-506, ABI-H03733 and ABI-H2158.
258. A method of treating a disease in a subject in need thereof, comprising administering to the subject the siNA according to any one of embodiments 1-246.
259. A method of treating a disease in a subject in need thereof, comprising administering to the subject the composition according to any one of embodiments 247-257.
260. The method of embodiment 258 or 259, wherein the disease is a viral disease.
261. The method of embodiment 260, wherein the viral disease is caused by a DNA virus.
262. The method of embodiment 261, wherein the DNA virus is a double stranded DNA (dsDNA) virus.
263. The method of embodiment 262, wherein the dsDNA virus is a hepadnavirus.
299
264. The method of embodiment 263, wherein the hepadnavirus is a hepatitis B virus (HBV).
265. The method of embodiment 264, wherein the HBV is selected from HBV génotypes A-J.
266. The method of any of embodiments 258-265, further comprising administering an additional HBV treatment agent.
267. The method of embodiment 266, wherein the siNA or the composition and the additional HBV treatment agent are administered concurrently.
268. The method of embodiment 266, wherein the siNA or the composition and the additional HBV treatment agent are administered sequentially.
269. The method of embodiment 266, wherein the siNA or the composition is administered prior to administering the additional HBV treatment agent.
270. The method of embodiment 266, wherein the siNA or the composition is administered after administering the additional HBV treatment agent.
271. The method of any one of embodiments 266-270, wherein the additional HBV treatment agent is selected from a nucléotide analog, nucleoside analog, a capsid assembly modulator (CAM), a recombinant interferon, an entry inhibitor, a small molécule immunomodulator and oligonucleotide therapy.
272. The method of embodiment 271, wherein the oligonucleotide therapy is an additional siNA.
273. The method of embodiment 272, wherein the additional siNA is selected from any of dssiNA-001 to ds-siNA-0178.
274. The method of embodiment 271, wherein the oligonucleotide therapy is an antisense oligonucleotide (ASO), NAPs, or STOPs.
275. The method of embodiment 274, wherein the ASO is ASO 1 or ASO 2.
276. The method of embodiment 270 or 271, wherein the additional HBV treatment agent is selected from HBV STOPS™ ALG-010133, HBV CAM ALG-000184, ASO 1, recombinant interferon alpha 2b, IFN-a, PEG-IFN-a-2a, lamivudine, telbivudine, adefovir dipivoxil, clevudine, entecavir, tenofovir alafenamide, tenofovir disoproxil, NVR3-778, BAY41-4109,
300
JNJ-632, JNJ-3989 (ARO-HBV), RG6004, GSK3228836, REP-2139, REP-2165, AB-729, VIR2218, RG6346 (DCR-HBVS), JNJ-6379, GLS4, ABI-HO731, JNJ-440, NZ-4, RG7907, EDP514, AB-423, AB-506, ABI-H03733 and ABI-H2158.
277. The method of embodiment 258 or 259, wherein the disease is a liver disease.
278. The method of embodiment 277, wherein the liver disease is a nonalcoholic fatty liver disease (NAFLD) or hepatocellular carcinoma (HCC).
279. The method of embodiment 278, wherein the NAFLD is nonalcoholic steatohepatitis (NASH).
280. The method of any of embodiments 277-279 further comprising administering to the subject a liver disease treatment agent.
281. The method of embodiment 280, wherein the liver disease treatment agent is selected from a peroxisome proliferator-activator receptor (PPAR) agonist, famesoid X receptor (FXR) agonist, lipid-altering agent, and incretin-based therapy.
282. The method of embodiment 281, wherein the PPAR agonist is selected from a PPARa agonist, dual PPARa/δ agonist, PPARy agonist, and dual PPARa/γ agonist.
283. The method of embodiment 282, wherein the dual PPARa agonist is a fibrate.
284. The method of embodiment 282, wherein the PPARa/δ agonist is elafibranor.
285. The method of embodiment 282, wherein the PPARy agonist is a thiazolidinedione (TZD).
286. The method of embodiment 282, wherein TZD is pioglitazone.
287. The method of embodiment 282, wherein the dual PPARa/γ agonist is saroglitazar.
288. The method of embodiment 281, wherein the FXR agonist is obeticholic acis (OCA).
289. The method of embodiment 281, wherein the lipid-altering agent is aramchol.
290. The method of embodiment 281, wherein the incretin-based therapy is a glucagon-like peptide 1 (GLP-1) receptor agonist or dipeptidyl peptidase 4 (DPP-4) inhibitor.
301
291. The method of embodiment 290, wherein the GLP-1 receptor agonist is exenatide or liraglutide.
292. The method of embodiment 290, wherein the DPP-4 inhibitor is sitagliptin or vildapliptin.
293. The method of any one of embodiments 280-292, wherein the siNA or composition and the liver disease treatment agent are administered concurrently.
294. The method of any one of embodiments 280-292, wherein the siNA or composition and the liver disease treatment agent are administered sequentially.
295. The method of any one of embodiments 280-292, wherein the siNA or composition is administered prior to administering the liver disease treatment agent.
296. The method of any one of embodiments 280-292, wherein the siNA or composition is administered after administering the liver disease treatment agent.
297. The method of any of one embodiments 258-296, wherein the siNA or the composition is administered at a dose of at least 1 mg/kg, 2 mg/kg, 3 mg/kg, 4 mg/kg, 5 mg/kg, 6 mg/kg, 7 mg/kg, 8 mg/kg, 9 mg/kg, 10 mg/kg, 11 mg/kg, 12 mg/kg, 13 mg/kg 14 mg/kg, or 15 mg/kg.
298. The method of any of one embodiments 258-296, wherein the siNA or the composition is administered at a dose of between 0.5 mg/kg to 50 mg/kg, 0.5 mg/kg to 40 mg/kg 0.5 mg/kg to 30 mg/kg, 1 mg/kg to 50 mg/kg, 1 mg/kg to 40 mg/kg, 1 mg/kg to 30 mg/kg, 1 mg/kg to 20 mg/kg, 3 mg/kg to 50 mg/kg, 3 mg/kg to 40 mg/kg, 3 mg/kg to 30 mg/kg, 3 mg/kg to 20 mg/kg, 3 mg/kg to 15 mg/kg, 3 mg/kg to 10 mg/kg, 4 mg/kg to 50 mg/kg, 4 mg/kg to 40 mg/kg, 4 mg/kg to 30 mg/kg, 4 mg/kg to 20 mg/kg, 4 mg/kg to 15 mg/kg, 4 mg/kg to 10 mg/kg, 5 mg/kg to 50 mg/kg, 5 mg/kg to 40 mg/kg, 5 mg/kg to 30 mg/kg, 5 mg/kg to 20 mg/kg, 5 mg/kg to 15 mg/kg, or 5 mg/kg to 10 mg/kg.
299. The method of any of one embodiments 258-298, wherein the siNA or the composition is administered at least 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 times.
300. The method of any of one embodiments 258-298, wherein the siNA or the composition is administered at least 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 times a day, at least 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 times a week, or at least 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 times a month.
302
301. The method of any of one embodiments 258-300, wherein the siNA or the composition are administered at least once every 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, or 21 days.
302. The method of any of one embodiments 258-301, wherein the siNA or the composition is administered for aperiod of at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, or 21 days, or at least 1,2, 3,4, 5, 6,7, 8,9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19,20, 21,22, 23, 24, 25, 26, 27, 28, 29, 30, 35, 40, 45, .50, 51, 52, 53, 54, or 55 weeks.
303. The method of any of one embodiments 258-302, wherein the siNA or the composition is administered at a single dose of 5 mg/kg.
304. The method of any of one embodiments 258-302, wherein the siNA or the composition is administered at a single dose of 10 mg/kg.
305. The method of any of one embodiments 258-302, wherein the siNA or the composition is administered at three doses of 10 mg/kg once a week.
306. The method of any of one embodiments 258-302, wherein the siNA or the composition is administered at three doses of 10 mg/kg once every three days.
307. The method of any of one embodiments 258-302, wherein the siNA or the composition is administered at fïve doses of 10 mg/kg once every three days.
308. The method of any of one embodiments 258-302, wherein the siNA or the composition is administered at six doses of ranging from 1 mg/kg to 15 mg/kg, 1 mg/kg to 10 mg/kg, 2 mg/kg to 15 mg/kg, 2 mg/kg to 10 mg/kg, 3 mg/kg to 15 mg/kg, or 3 mg/kg to 10 mg/kg.
309. The method of embodiment 308, wherein the first dose and second dose are administered at least 3 days apart.
310. The method of embodiment 308 or 309, wherein the second dose and third dose are administered at least 4 days apart.
311. The method of any one of embodiments 308-310, wherein the third dose and fourth dose, fourth dose and fifth dose, or fifth dose and sixth dose are administered at least 7 days apart.
312. The method of any one of embodiments 258-311, wherein the siNA or the composition are administered in a particle or viral vector.
303
313. The method of embodiment 312, wherein the viral vector is a vector of adenovirus, adenoassociated virus (AAV), alphavirus, flavivirus, herpes simplex virus, lentivirus, measles virus, picomavirus, poxvirus, rétro virus, or rhabdo virus.
314. The method of embodiment 312, wherein the viral vector is a recombinant viral vector.
315. The method according to any one of embodiments 312-314, wherein the viral vector is selected from AAVrh.74, AAVrh.10, AAVrh.20, AAV-1, AAV-2, AAV-3, AAV-4, AAV-5, AAV-6, AAV-7, AAV-8, AAV-9, AAV-10, AAV-11, AAV-12 and AAV-13.
316. The method according to any one of embodiments 258-315, wherein the siNA or the composition is administered systemically.
317. The method according to any one of embodiments 258-315, wherein the siNA or the composition is administered locally.
318. The method according to any one of embodiments 258-317, wherein the siNA or the composition is administered intravenously, subcutaneously, or intramuscularly.
319. Use of the siNA according to any one of embodiments 1-246 or the composition according to any one of embodiments 247-257 in the manufacture of a médicament for treating a disease.
320. The use of embodiment 319, wherein the disease fs a viral disease.
321. The use of embodiment 320, wherein the viral disease is caused by a DNA virus.
322. The use of embodiment 321, wherein the DNA virus is a double stranded DNA (dsDNA virus).
323. The use of embodiment 321, wherein the dsDNA virus is a hepadnavirus.
324. The use of embodiment 323, wherein the hepadnavirus is a hepatitis B virus (HBV).
325. The use of embodiment 324, wherein the HBV is selected from HBV génotypes A-J.
326. The use of any of one of embodiments 319-325, further comprising an additional HBV treatment agent in the manufacture of the médicament.
304
327. The use of embodiment 326, wherein the additional HBV treatment agent is selected from a nucléotide analog, nucleoside analog, a capsid assembly modulator (CAM), a recombinant interferon, an entry inhibitor, a small molécule immunomodulator and oligonucleotide therapy.
328. The use of embodiment 327, wherein the oligonucleotide therapy is an additional siNA.
329. The use of embodiment 328, wherein the additional siNA is selected from any of ds- siNA-001 to ds-siNA-0178.
330. The use of embodiment 327, wherein the oligonucleotide therapy is an antisense oligonucleotide (ASO), NAPs, or STOPs.
331. The use of embodiment 330, wherein the ASO is ASO 1 or ASO2.
332. The use of embodiment 326 or 327, wherein the additional HBV treatment agent is selected from HBV STOPS™ ALG-010133, HBV CAM ALG-000184, ASO 1, recombinant interferon alpha 2b, IFN-a, PEG-IFN-a-2a, lamivudine, telbivudine, adefovir dipivoxil, clevudine, entecavir, tenofovir alafenamide, tenofovir disoproxil, NVR3-778, BAY41-4109, JNJ-632, JNJ-3989 (ARO-HBV), RG6004, GSK3228836, REP-2139, REP-2165, AB-729, VIR2218, RG6346 (DCR-HBVS), JNJ-6379, GLS4, ABI-HO731, JNJ-440, NZ-4, RG7907, EDP514, AB-423, AB-506, ABI-H03733 and ABI-H2158.
333. The use of embodiment 319, wherein the disease is a liver disease.
334. The use of embodiment 333, wherein the liver disease is a nonalcoholic fatty liver disease (NAFLD) or hepatocellular carcinoma (HCC).
335. The use of embodiment 334, wherein the NAFLD is nonalcoholic steatohepatitis (NASH).
336. The use of any of embodiments 333-335, further comprising a liver disease treatment agent in the manufacture of the médicament.
337. The use of embodiment 336, wherein the liver disease treatment agent is selected from a peroxisome proliferator-activator receptor (PPAR) agonist, famesoid X receptor (FXR) agonist, lipid-altering agent, and incretin-based therapy.
305
338. The use of embodiment 337, wherein the PPAR agonist is selected from a PPARa agonist, dual PPARa/δ agonist, PPARy agonist, and dual PPARa/γ agonist.
339. The use of embodiment 338, wherein the dual PPARa agonist is a fibrate.
340. The use of embodiment 338, wherein the PPARa/δ agonist is elafïbranor.
341. The use of embodiment 338, wherein the PPARy agonist is a thiazolidinedione (TZD).
342. The use of embodiment 341, wherein TZD is pioglitazone.
343. The use of embodiment 338, wherein the dual PPARa/γ agonist is saroglitazar.
344. The use of embodiment 337, wherein the FXR agonist is obeticholic acis (OCA).
345. The use of embodiment 337, wherein the lipid-altering agent is aramchol.
346. The use of embodiment 337, wherein the incretin-based therapy is a glucagon-like peptide 1 (GLP-1) receptor agonist or dipeptidyl peptidase 4 (DPP-4) inhibitor.
347. The use of embodiment 346, wherein the GLP-1 receptor agonist is exenatide or liraglutide.
348. The use of embodiment 346, wherein the DPP-4 inhibitor is sitagliptin or vildapliptin.
349. The siNA according to any one of embodiments 1-246 for use as a médicament.
350. The composition according to any one of embodiments 247-257 for use as a médicament.
351. The siNA according to any one of embodiments 1 -246 for use in the treatment of a disease.
352. The siNA of embodiment 351, wherein the disease is a viral disease.
353. The siNA of embodiment 352, wherein the viral disease is caused by a DNA virus.
354. The siNA of embodiment 353, wherein the DNA virus is a double stranded DNA (dsDNA virus).
355. The siNA of embodiment 354, wherein the dsDNA virus is a hepadnavirus.
356. The siNA of embodiment 355, wherein the hepadnavirus is a hepatitis B virus (HBV).
306
357. The siNA of embodiment 356, wherein the HBV is selected from HBV génotypes A-J.
358. The siNA of embodiment 351, wherein the disease is a liver disease.
359. The siNA of embodiment 358, wherein the liver disease is a nonalcoholic fatty liver disease (NAFLD) or hepatocellular carcinoma (HCC).
360. The siNA of embodiment 359, wherein the NAFLD is nonalcoholic steatohepatitis (NASH).
361. The composition according to any one of embodiments 247-257, for use in the treatment of a disease.
362. The composition of embodiment 361, wherein the disease is a viral disease.
363. The composition of embodiment 362, wherein the viral disease is caused by a DNA virus.
364. The composition of embodiment 363, wherein the DNA virus is a double stranded DNA (dsDNA virus).
365. The composition of embodiment 364, wherein the dsDNA virus is a hepadnavirus.
366. The composition of embodiment 365, wherein the hepadnavirus is a hepatitis B virus (HBV).
367. The composition of embodiment 366, wherein the disease is a liver disease.
368. The composition of embodiment 367, wherein the liver disease is a nonalcoholic fatty liver disease (NAFLD) or hepatocellular carcinoma (HCC).
369. The composition of embodiment 368, wherein the NAFLD is nonalcoholic steatohepatitis (NASH).
Tables
Table 1. Non-modifïed Nucléotide Sequences
SEQ ID NO. First Nucléotide Sequence (5’-3’) SEQ ID NO. Second Nucléotide Sequence(5’-3’
1 ACCGUGUGCACUUCGCUUC 57 GAAGCGAAGUGCACACGGUCC
2 ACCGUGUGCACUUCGCUUC 58 GAAGCGAAGUGCACACGGU
307
Table 1. Non-modifïed Nucléotide Sequences
SEQ ID NO. First Nucléotide Sequence (5’-3’) SEQ ID NO. Second Nucléotide Sequence(5’-3’
3 ACUUCGCUUCACCUCUGCA 59 UGCAGAGGUGAAGCGAAGUGC
4 AGUGUUUGCUGACGCAACC 60 .GGUUGCGUCAGCAAACACUUG
5 CAGGCGGGGUUUUUCUUGU 61 ACAAGAAAAACCCCGCCUGUA
6 CAGGCGGGGUUUUUCUUGU 62 ACAAGAAAAAACCCCGCCUG
7 CAGUUUACUAGUGCCAUUU 63 AAAUGGCACUAGUAAACUGAG
8 CAGUUUACUAGUGCCAUUU 64 AAAUGGCACUAGUAAACUG
9 CAUCCUGCUGCUAUGCCUC 65 GAGGCAUAGCAGCAGGAUGAA
10 CAUCCUGCUGCUAUGCCUCAU 66 AUGAGGCAUAGCAGCAGGAUGAA
11 CAUCCUGCUGCUAUGCCUC 67 GAGGCAUAGCAGCAGGAUG
12 CCGUGUGCACUUCGCUUCA 68 UGAAGCGAAGUGCACACGGUC
13 CCGUGUGCACUUCGCUUCA 69 UGAAGCGAAGUGCACACGG
14 CCUGCUGCUAUGCCUCAUCUU 70 AAGAUGAGGCAUAGCAGCAGGAU
15 CUCAGUUUACUAGUGCCAU 71 AUGGCACUAGUAAACUGAGCC
16 CUCAGUUUACUAGUGCCAU 71 AUGGCACUAGUAAACUGAGCC
17 CUCAGUUUACUAGUGCCAU 71 AUGGCACUAGUAAACUGAGCC
18 CUCAGUUUACUAGUGCCAU 72 AUGGCACUAGUAAACUGAG
19 CUCAGUUUACUAGUGCCAU 72 AUGGCACUAGUAAACUGAG
20 CUGCUAUGCCUCAUCUUCU 73 AGAAGAUGAGGCAUAGCAGCA
21 CUGCUAUGCCUCAUCUUCU 73 AGAAGAUGAGGCAUAGCAGCA
22 CUGCUAUGCCUCAUCUUCU 74 AGAAGAUGAGGCAUAGCAG
23 CUGCUAUGCCUCAUCUUCU 74 AGAAGAUGAGGCAUAGCAG
24 CUGCUGCUAUGCCUCAUCU 75 AGAUGAGGCAUAGCAGCAGGA
25 CUGCUGCUAUGCCUCAUCU 76 AGAUGAGGCAUAGCAGCAG
26 CUGCUGCUAUGCCUCAUCU 76 AGAUGAGGCAUAGCAGCAG
27 CUUCGCUUCACCUCUGCACGU 77 ACGUGCAGAGGUGAAGCGAAGUG
28 GCACUUCGCUUCACCUCUGCA 78 UGCAGAGGUGAAGCGAAGUGCAC
29 GCCGAUCCAUACUGCGGAA 79 UUCCGCAGUAUGGAUCGGCAG
30 GCCGGGUUUUUCUUGUUGA 80 UUCCGCAGUAUGGAUCGGC
31 GCGGGGUUUUUCUUGUUGA 81 UCAACAAGAAAAACCCCGCCU
32 GCGGGGUUUUUCUUGUUGA 81 UCAACAAGAAAAACCCCGCCU
33 GCGGGGUUUUUCUUGUUGA 82 UCAACAAGAAAAACCCCGC
34 GCGGGGUUUUUCUUGUUGA 82 UCAACAAGAAAAACCCCGC
35 GCUGCUAUGCCUCAUCUUCUU 83 AAGAAGAUGAGGCAUAGCAGCAG
36 GGAUGUGUCUGCGGCGUUUUA 84 UAAAACGCCGCAGACACAUCCAG
37 GGCCAAAAUUCGCAGUCCC 85 GGGACUGCGAAUUUUGGCCAA
38 GGCGCACCUCUCUUUACGC 86 GCGUAAAGAGAGGUGCGCCCC
39 GUAUGUUGCCCGUUUGUCC 87 GGACAAACGGGCAACAUACCU
40 GUGGUGGACUUCUCUCAAU 88 AUUGAGAGAAGUCCACCACGA
41 GUGUGCACUUCGCUUCACC 89 GGUGAAGCGAAGUGCACACGG
308
Table 1. Non-modified Nucléotide Sequences
SEQ ID NO. First Nucléotide Sequence (5’-3’) SEQ ID NO. Second Nucléotide Sequence(5’-3’
42 GUUGCCCGUUUGUCCUCUA 90 UAGAGGACAAACGGGCAACAU
43 GUUGCCCGUUUGUCCUCUA 91 UAGAGGACAAACGGGCAAC
44 UCCAUACUGCGGAACUCCU 92 AGGAGUUCCGCAGUAUGGAUC
45 UCCAUACUGCGGAACUCCU 93 AGGAGUUCCGCAGUAUGGA
46 UCGUGGUGGACUUCUCUCAAU 94 AUUGAGAGAAGUCCACCACGAGU
47 UGCACUUCGCUUCACCUCU 95 AGAGGUGAAGCGAAGUGCACA
48 UGCCGAUCCAUACUGCGGA 96 UCCGCAGUAUGGAUCGGCAGA
49 UGCCGAUCCAUACUGCGGA 97 UCCGCAGUAUGGAUCGGCA
50 UGCUAUGCCUCAUCUUCUU 98 AAGAAGAUGAGGCAUAGCAGC
51 UGUGCACUUCGCUUCACCU 99 AGGUGAAGCGAAGUGCACACG
52 UGUGCACUUCGCUUCACCU 99 AGGUGAAGCGAAGUGCACACG
53 UGUGCACUUCGCUUCACCU 100 AGGUGAAGCGAAGUGCACA
54 UGUGCACUUCGCUUCACCU 100 AGGUGAAGCGAAGUGCACA
55 UUGCCCGUUUGUCCUCUAA 101 UUAGAGGACAAACGGGCAACA
56 UUGCCCGUUUGUCCUCUAA 102 UUAGAGGACAAACGGGCAA
Table 2. 2’-OMe and 2’-F Modified Nucléotide Sequences
SEQ ID NO. First Nucléotide Sequence (5’3’) SEQID NO. Second Nucléotide Sequence(5’3’)
103 mAmCfCmGmUmGfUfGfCmA mCfUmUmCmGmCfUmUmC 159 mGfAmAmGmCmGmAmAmGm UmGmCmAfCmAmCmGmGmUm CmC
104 mAmCfCmGmUmGfUfGfCmA mCfUmUmCmGmCfUmUmC 160 mGfAmAmGmCmGmAmAmGm UmGmCmAfCmAmCmGmGmU
105 mAmCfUmUmCmGfCfUfUmC mAfCmCmUmCmUfGmCmA 161 mU fGmCm AmGm AmGmGmUm GmAmAmGfCmGmAmAmGmU mGmC
106 mAmGfUmGmUmUfUfGfCmU mGfAmCmGmCmAfAmCmC 162 mGfGmUmUmGmCmGmUmCmA mGmCmAfAmAmCmAmCmUmU mG
107 mCmAfGmGmCmGfGfGfGmU mU fUmUmUmCmU fU mGmU 163 mAfCmAmAmGmAmAmAmAm AmCmCmCfCmGmCmCmUmGm UmA
108 mCmAfGmGmCmGfGfGfGmU mUfUmUmUmCmU fU mGmU 164 mAfCmAmAmGmAmAmAmAm AmAmCmCmCfCmGmCmCmUm G
109 mCmAfGmUmUmUfAfCfUmA mGfUmGmCmCmAfUmUmU 165 mAfAmAmUmGmGmCmAmCmU mAmGmUfAmAmAmCmUmGm AmG
309
Table 2. 2’-OMe and 2*-F Modified Nucléotide Sequences
SEQ ID NO. First Nucléotide Sequence (5’3’) SEQ ID NO. Second Nucléotide Sequence(5’3’)
110 mCm AfGmUmUmU fAfCfUm A mGfUmGmCmCmAfUmUmU 166 mAfAmAmUmGmGmCmAmCmU m AmGmU fAmAm AmCmUmG
111 mCmAfUmCmCmUfGfCfUmG mCfUmAmUmGmCfCmUmC 167 mGfAmGmGmCmAmUmAmGmC mAmGmCfAmGmGmAmUmGm AmA
112 mCm AmUmCmCm U fGmCfU fG fCmUmAmUmGmCmCmUmCm AmU 168 mAfUmGmAmGfGmCmAmUmA mGmCmAfGmCfAmGmGmAmU mGmAmA
113 mCmAfUmCmCmUfGfCfUmG mCfUmAmUmGmCfCmUmC 169 mGfAmGmGmCmAmUmAmGmC mAmGmCfAmGmGmAmUmG
114 mCmCfGmUmGmUfGfCfAmC mUfUmCmGmCmUfUmCmA 170 mU fGm AmAmGmCmGmAm Am GmUmGmCfAmCmAmCmGmGm UmC
115 mCmCfGmUmGmUfGfCfAmC mUfUmCmGmCmU fUmCm A 171 mUfGm AmAmGmCmGmAmAm GmUmGmCfAmCmAmCmGmG
116 mCmCmUmGmCmUfGmCfUfA fUmGmCmCmUmCmAmUmCm UmU 172 m AfAmGm AmU fGm AmGmGmC mAmUmAfGmCfAmGmCmAmG mGmAmU
117 mCmU fCmAmGmU fU fU fAmC mU fAmGmUmGmCfCm AmU 173 mAfUmGmGmCmAmCmUmAmG mUmAmAfAmCmUmGmAmGmC mC
118 mCmUmCmAmGmUfUmUmAm CmUmAmGmUmGmCmCmAm U 173 mAfU mGmGmCm AmCmUm AmG mUmAmAfAmCmUmGmAmGmC mC
119 mCmU fCmAmGmU fU fUmAmC mUmAmGmUmGmCfCmAmU 173 mAfUmGmGmCmAmCmUmAmG mUmAmAfAmCmUmGmAmGmC mC
120 mCmUfCmAmGmUfUfUfAmC mU fAmGmUmGmCfCmAmU 174 mAfUmGmGmCmAmCmUmAmG mUmAmAfAmCmUmGmAmG
121 mCmU fCmAmGmU fU fU m AmC mUmAmGmUmGmCfCmAmU 174 mAfUmGmGmCmAmCmUmAmG mUmAmAfAmCmUmGmAmG
122 mCmUfGmCmUmAfUfGfCmC mU fCm AmUmCmU fUmCmU 175· mAfGm Am AmGm AmU mGm Am GmGmCmAfUmAmGmCmAmGm CmA
123 mCmUfGmCmUmAfUfGmCmC mUmCmAmUmCmUfUmCmU 175 mAfGmAmAmGmAmUmGmAm GmGmCm AfU m AmGmCmAmGm CmA
124 mCmUfGmCmUmAfUfGfCmC mUfCmAmUmCmUfUmCmU 176 mAfGmAmAmGmAmUmGmAm GmGmCmAfUmAmGmCmAmG
125 mCmUfGmCmUmAfUfGmCmC mUmCm AmUmCmU fUmCmU 176 mAfGmAmAmGmAmUmGmAm GmGmCmAfUmAmGmCmAmG
310
Table 2. 2’-OMe and 2’-F Modified Nucléotide Sequences
SEQ ID NO. First Nucléotide Sequence (5’3’) SEQ ID NO. Second Nucléotide Sequence(5’3’)
126 mCmUfGmCmUmGfCfUfAmU mGfCmCmUmCmAfUmCmU 177 mAfGmAmUmGmAmGmGmCm AmUmAmGfCmAmGmCmAmGm GmA
127 mCmUfGmCmUmGfCfUfAmU mGfCmCmUmCmAfUmCmU 178 mAfGmAmUmGmAmGmGmCm AmUmAmGfCmAmGmCmAmG
128 mCmUfGmCmUmGfCfUmAmU mGmCmCmUmCmAfUmCmU 178 mAfGmAmUmGmAmGmGmCm AmUmAmGfCmAmGmCmAmG
129 mCmUmUmCmGmCfUmUfCfA fCmCmUmCmUmGmCmAmCm GmU 179 mAfCmGmUmGfCmAmGmAmG mGmUmGfAmAfGmCmGmAmA mGmUmG
130 mGmCmAmCmUmUfCmGfCfU fUmCmAmCmCmUmCmUmGm CmA 180 mU fGmCm AmGfAmGmGmUmG mAmAmGfCmGfAmAmGmUmG mCmAmC
131 mGmCfCmGmAmUfCfCfAmU mAfCmUmGmCmGfGmAmA 181 mUfUmCmCmGmCmAmGmUmA mUmGmGfAmUmCmGmGmCmA mG
132 mGmCfCmGmGmGfUfUfUmU mUfCmUmUmGmUfUmGmA 182 mUfUmCmCmGmCmAmGmUmA mUmGmGfAmUmCmGmGmC
133 mGmCfGmGmGmGfUfUfUmU mUfCmUmUmGmUfUmGmA 183 mU fCm Am AmCmAm AmGmAm A mAmAmAfCmCmCmCmGmCmC mU
134 mGmCfGmGmGmGfUfUmUmU mUmCmUmUmGmUfUmGmA 183 mU fCm Am AmCm AmAmGm AmA mAmAmAfCmCmCmCmGmCmC mU
135 mGmCfGmGmGmGfUfUfUmU mUfCmUmUmGmUfUmGmA 184 mU fCm Am AmCmAm AmGmAm A mAmAmAfCmCmCmCmGmC
136 mGmCfGmGmGmGfUfUmUmU mUmCmUmUmGmU fU mGm A 184 mUfCmAmAmCmAm AmGmAm A mAmAmAfCmCmCmCmGmC
137 mGmCmUmGmCmUfAmUfGfC fCmUmCmAmUmCmUmUmCm UmU 185 mAfAmGmAmAfGmAmUmGmA mGmGmCfAmUfAmGmCmAmG mCmAmG
138 mGmGmAmUmGmUfGmUfCfU fGmCmGmGmCmGmUmUmUm UmA 186 mU fAmAm Am AfCmGmCmCmG mCmAmGfAmCfAmCmAmUmC mCmAmG .
139 mGmGfCmCmAmAfAfAfUmU mCfGmCmAmGmUfCmCmC 187 mGfGmGmAmCmUmGmCmGmA mAmUmUfUmUmGmGmCmCm A mA
140 mGmGfCmGmCmAfCfCfUmCm UfCmUmUmUmAfCmGmC 188 mGfCmGmUmAmAmAmGmAm GmAmGmGfUmGmCmGmCmCm CmC
311
Table 2. 2’-OMe and 2’-F Modified Nucléotide Sequences
SEQ IDNO. First Nucléotide Sequence (5’3’) SEQ ID NO. Second Nucléotide Sequence(5’3’)
141 mGmUfAmUmGmUfUfGfCmC mCfGmUmUmUmGfUmCmC 189 mGfGmAmCmAmAmAmCmGmG mGmCmAfAmCmAmUmAmCmC mU
142 mGmUfGmGmUmGfGfAfCmU mUfCmUmCmUmCfAmAmU 190 mAfUmUmGmAmGmAmGmAm AmGmUmCfCmAmCmCmAmCm GmA
143 mGmUfGmUmGmCfAfCfUmU mCfGmCmUmUmCfAmCmC 191 mGfGmUmGmAmAmGmCmGm AmAmGmU fGmCm AmCmAmCm GmG
144 mGmUfUmGmCmCfCfGfUmU mUfGmUmCmCmUfCmUmA 192 mU fAmGm AmGmGm AmCmAm AmAmCmGfGmGmCmAmAmCm AmU
145 mGmUfUmGmCmCfCfGfUmU mUfGmUmCmCmUfCmUmA 193 mU fAmGm AmGmGm AmCmAm AmAmCmGfGmGmCmAmAmC
146 mUmCfCmAmUmAfCfUfGmC mGfGm Am AmCmU fCmCmU 194 mAfGmGmAmGmUmUmCmCmG mCmAmGfUmAmUmGmGmAm UmC
147 mUmCfCmAmUmAfCfUfGmC mGfGm Am AmCmU fCmCmU 195 mAfGmGmAmGmUmUmCmCmG mCmAmGfUmAmUmGmGmA
148 mUmCmGmUmGmGfUmGfGfA fCmUmUmCmUmCmUmCmAm AmU 196 mAfUmUmGmAfGmAmGmAmA mGmUmCfCmAfCmCmAmCmG mAmGmU
149 mUmGfCmAmCmUfUfCfGmC mUfUmCmAmCmCfUmCmU 197 mAfGmAmGmGmUmGmAmAm GmCmGmAfAmGmUmGmCmAm CmA
150 mUmGfCmCmGmAfUfCfCmA mU fAmCmUmGmCfGmGm A 198 mUfCmCmGmCmAmGmUmAmU mGmGmAfUmCmGmGmCmAmG mA
151 mUmGfCmCmGmAfU fCfCm A mU fAmCmUmGmCfGmGm A 199 mUfCmCmGmCmAmGmUmAmU mGmGmAfUmCmGmGmCmA
152 mUmGfCmUmAmUfGfCfCmU mCfAmUmCmUmUfCmUmU 200 mAfAmGmAmAmGmAmUmGm AmGmGmCfAmUmAmGmCmAm GmC
153 mUmGfUmGmCmAfCfUfUmC mGfCmUmUmCmAfCmCmU 201 mAfGmGmUmGmAmAmGmCm GmAm AmGfU mGmCm AmCmAm CmG
154 mUmGfUmGmCmAfCfUmUmC mGmCmUmUmCm AfCmCmU 201 mAfGmGmUmGmAmAmGmCm GmAmAmGfUmGmCmAmCmAm CmG
155 mUmGfUmGmCmAfCfU fUmC mGfCmUmUmCmAfCmCmU 202 mAfGmGmUmGmAmAmGmCm GmAmAmGfUmGmCmAmCmA
312
Table 2. 2’-OMe and 2’-F Modified Nucléotide Sequences
SEQ IDNO. First Nucléotide Sequence (5’3’) SEQ ID NO. Second Nucléotide Sequence(5’3’)
156 mUmGfUmGmCmAfCfUmUmC mGmCmUmUmCmAfCmCmU 202 mAfGmGmUmGmAmAmGmCm GmAmAmGfUmGmCmAmCmA
157 mUmUfGmCmCmCfGfUmUmU mGmUmCmCmUmCfUmAmA 203 mU fUmAmGm AmGmGm AmCm AmAmAmCfGmGmGmCmAmAm CmA
158 mUmUfGmCmCmCfGfUfUmU mGfUmCmCmUmCfUmAmA 204 mU fUmAmGm AmGmGm AmCm AmAmAmCfGmGmGmCmAmA
mX = 2’-O-methyl nucléotide; fX = 2’-fluoro nucléotide
Table 3. 2’-O-methyl and 2’-fluoro Modified Nucléotide Sequences with Phosphorothioate Linkages
SEQ ID NO. First Nucléotide Sequence (5’3’) SEQ ID NO. Second Nucléotide Sequence (5’-3’
205 mApsmCpsfCmGmUmGfUfGfC mAmCfUmUmCmGmCfUmUmC 261 mGpsfApsmAmGmCmGmAmAmGm UmGmCmAfCmAmCmGmGmUpsmC psmC
206 mApsmCpsfCmGmUmGfUfGfC mAmCfUmUmCmGmCfUmUmC 262 mGpsfApsmAmGmCmGmAmAmGm UmGmCmAfCmAmCmGmGmU
207 mApsmCpsfU mUmCmGfCfU fU mCmAfCmCmUmCmUfGmCmA 263 mUpsfGpsmCmAmGmAmGmGmUm GmAmAmGfCmGmAmAmGmUpsm GpsmC
208 mApsmGpsfUmGmUmU fU fGfC mUmGfAmCmGmCmAfAmCmC 264 mGpsfGpsmUmUmGmCmGmUmCm AmGmCmAfAmAmCmAmCmUpsmU psmG
209 mCpsmApsfGmGmCmGfGfGfG mUmU fU mUmUmCmUfUmGm U 265 mApsfCpsmAmAmGmAmAmAmAm AmCmCmCfCmGmCmCmUmGpsmU psmA
210 mCpsmApsfGmGmCmGfGfGfG mUmUfUmUmUmCmUfUmGm U 266 mApsfCpsmAmAmGmAmAmAmAm AmAmCmCmCfCmGmCmCmUmG
211 mCpsmApsfGmUmUmUfAfCfU mAmGfUmGmCmCmAfUmUm U 267 mApsfApsmAmUmGmGmCmAmCm UmAmGmU fAm Am AmCmUmGpsm ApsmG
212 mCpsmApsfGmUmUmUfAfCfU mAmGfUmGmCmCmAfUmUm U 268 mApsfApsmAmUmGmGmCmAmCm UmAmGmU fAm AmAmCmUmG
213 mCpsmApsfUmCmCmUfGfCfU mGmCfUmAmUmGmCfCmUmC 269 mGpsfApsmGmGmCmAmUmAmGm CmAmGmCfAmGmGmAmUmGpsm ApsmA
313
Table 3. 2’-O-methyl and 2’-fluoro Modified Nucléotide Sequences with Phosphorothioate Linkages
SEQ ID NO. First Nucléotide Sequence (5’3’) SEQ ID NO. Second Nucléotide Sequence (5’-3’
214 mCpsmApsmUmCmCmUfGmCf UfGfCmUmAmUmGmCmCmUm CmAmU 270 mApsfUpsmGmAmGfGmCmAmUmA mGmCmAfGmCfAmGmGmAmUmGp smApsmA
215 mCpsmApsfUmCmCmUfGfCfU mGmCfUmAmUmGmCfCmUmC 271 mGpsfApsmGmGmCmAmUmAmGm CmAmGmCfAmGmGmAmUmG
216 mCpsmCpsfGmUmGmUfGfCfA mCmUfUmCmGmCmUfUmCmA 272 mUpsfGpsmAmAmGmCmGmAmAm GmUmGmCfAmCmAmCmGmGpsmU psmC
217 mCpsmCpsfGmUmGmUfGfCfA mCmU fU mCmGmCmUfUmCm A 273 mUpsfGpsmAmAmGmCmGmAmAm GmUmGmCfAmCmAmCmGmG
218 mCpsmCpsmUmGmCmUfGmCf U fAfU mGmCmCmUmCm AmUm CmUmU 274 mApsfApsmGmAmUfGmAmGmGmC mAmUmAfGmCfAmGmCmAmGmGp smApsmU
219 mCpsmUpsfCmAmGmUfUfUfA mCmUfAmGmUmGmCfCmAmU 275 mApsfUpsmGmGmCmAmCmUmAm GmUmAmAfAmCmUmGmAmGpsm CpsmC
220 mCpsmUpsmCmAmGmUfUmU mAmCmUmAmGmUmGmCmC mAmU 275 mApsfUpsmGmGmCmAmCmUmAm GmUmAmAfAmCmUmGmAmGpsm CpsmC
221 mCpsmUpsfCmAmGmUfUfUmA mCmUmAmGmUmGmCfCmAm U 275 mApsfUpsmGmGmCmAmCmUmAm GmUmAmAfAmCmUmGmAmGpsm CpsmC
222 mCpsmUpsfCmAmGmUfUfUfA mCmU fAmGmUmGmCfCm AmU 276 mApsfUpsmGmGmCmAmCmUmAm GmUmAmAfAmCmUmGmAmG
223 mCpsmUpsfCmAmGmUfUfUmA mCmUmAmGmUmGmCfCmAm U 276 mApsfUpsmGmGmCmAmCmUmAm GmUmAmAfAmCmUmGmAmG
224 mCpsmUpsfGmCmUmAfUfGfC mCmU fCm AmUmCmU fUmCmU 277 mApsfGpsmAmAmGmAmUmGmAm GmGmCmAfUmAmGmCmAmGpsmC psmA
225 mCpsmUpsfGmCmUmAfUfGmC mCmUmCmAmUmCmUfUmCm U 277 mApsfGpsmAmAmGmAmUmGmAm GmGmCmAfUmAmGmCmAmGpsmC psmA
226 mCpsmUpsfGmCmUmAfUfGfC mCmU fCm AmUmCmU fU mCmU 278 mApsfGpsmAmAmGmAmUmGmAm GmGmCmAfUmAmGmCmAmG
227 mCpsmUpsfGmCmUmAfUfGmC mCmUmCm AmUmCm U fU mCm U 278 mApsfGpsmAmAmGmAmUmGmAm GmGmCmAfUmAmGmCmAmG
314
Table 3. 2’-O-methyl and 2’-fluoro Modified Nucléotide Sequences with Phosphorothioate Linkages
SEQ ID NO. First Nucléotide Sequence (5’3’) SEQ ID NO. Second Nucléotide Sequence (5’-3’
228 mCpsmUpsfGmCmUmGfCfUfA mUmGfCmCmUmCmAfUmCmU 279 mApsfGpsmAmUmGmAmGmGmCm AmUmAmGfCmAmGmCmAmGpsm GpsmA
229 mCpsmUpsfGmCmUmGfCfUfA mUmGfCmCmUmCmAfUmCmU 280 mApsfGpsmAmUmGmAmGmGmCm AmUmAmGfCmAmGmCmAmG
230 mCpsmUpsfGmCmUmGfCfUmA mUmGmCmCmUmCmAfUmCm U . 280 mApsfGpsmAmUmGmAmGmGmCm AmUmAmGfCmAmGmCmAmG
231 mCpsmUpsmUmCmGmCfUmUf CfAfCmCmUmCmUmGmCmAm CmGmU 281 mApsfCpsmGmUmGfCmAmGmAmG mGmUmGfAmAfGmCmGmAmAmG psmUpsmG
232 mGpsmCpsmAmCmUmUfCmGf CfUfUmCmAmCmCmUmCmUm GmCmA 282 mUpsfGpsmCmAmGfAmGmGmUmG mAmAmGfCmGfAmAmGmUmGmCp smApsmC
233 mGpsmCpsfCmGmAmUfCfCfA mUmAfCmUmGmCmGfGmAm A 283 mUpsfUpsmCmCmGmCmAmGmUm AmUmGmGfAmUmCmGmGmCpsm ApsmG
234 mGpsmCpsfCmGmGmGfU fU fU mUmUfCmUmUmGmUfUmGm A 284 mUpsfUpsmCmCmGmCmAmGmUm AmUmGmGfAmUmCmGmGmC
235 mGpsmCpsfGmGmGmGfU fU fU mUmU fCmUmUmGmU fUmGm A 285 mUpsfCpsmAmAmCmAmAmGmAm AmAmAmAfCmCmCmCmGmCpsmC psmU
236 mGpsmCpsfGmGmGmGfU fU mU mUmUmCmUmUmGmUfUmGm A 285 mUpsfCpsmAmAmCmAmAmGmAm AmAmAmAfCmCmCmCmGmCpsmC psmU
237 mGpsmCpsfGmGmGmGfUfUfU mUmU fCmUmUmGmU fUmGm A 286 mUpsfCpsmAmAmCmAmAmGmAm AmAmAmAfCmCmCmCmGmC
238 mGpsmCpsfGmGmGmGfUfUmU mUmUmCmUmUmGmU fUmGm A 286 mUpsfCmAmAmCmAmAmGmAmA mAmAmAfCmCmCmCmGmC
239 mGpsmCpsmUmGmCmUfAmUf GfCfCmUmCmAmUmCmUmUm CmUmU 287 mApsfApsmGmAmAfGmAmUmGmA mGmGmCfAmUfAmGmCm AmGmCp smApsmG
240 mGpsmGpsmAmUmGmUfGmUf CfUfGmCmGmGmCmGmUmUm UmUmA 288 mUpsfApsmAmAmAfCmGmCmCmG mCmAmGfAmCfAmCmAmUmCmCp smApsmG
315
Table 3. 2’-O-methyl and 2’-fluoro Modified Nucléotide Sequences with Phosphorothioate Linkages
SEQ ID NO. First Nucléotide Sequence (5’3’) SEQ ID NO. Second Nucléotide Sequence (5’-35
241 mGpsmGpsfCmCmAmAfAfAfU mUmCfGmCmAmGmUfCmCmC 289 mGpsfGpsmGmAmCmUmGmCmGm AmAmUmUfUmUmGmGmCmCpsm ApsmA
242 mGpsmGpsfCmGmCmAfCfCfU mCmUfCmUmUmUmAfCmGmC 290 mGpsfCpsmGmUmAmAmAmGmAm GmAmGmGfUmGmCmGmCmCpsmC psmC
243 mGpsmUpsfAmUmGmUfUfGfC mCmCfGmUmUmUmGfUmCmC 291 mGpsfGpsmAmCmAmAmAmCmGm GmGmCmAfAmCmAmUmAmCpsmC psmU
244 mGpsmUpsfGmGmUmGfGfAfC mUmUfCmUmCmUmCfAmAmU 292 mApsfUpsmUmGmAmGmAmGmAm AmGmUmCfCmAmCmCmAmCpsmG psmA
245 mGpsmUpsfGmUmGmCfAfCfU mUmCfGmCmUmUmCfAmCmC 293 mGpsfGpsmUmGmAmAmGmCmGm AmAmGmUfGmCmAmCmAmCpsmG psmG
246 mGpsmUpsfUmGmCmCfCfGfU mUmUfGmUmCmCmUfCmUmA 294 mUpsfApsmGmAmGmGmAmCmAm AmAmCmGfGmGmCmAmAmCpsmA psmU
247 mGpsmUpsfUmGmCmCfCfGfU mUmUfGmUmCmCmUfCmUmA 295 mUpsfApsmGmAmGmGmAmCmAm AmAmCmGfGmGmCmAmAmC
248 mUpsmCpsfCmAmUmAfCfUfG mCmGfGmAmAmCmU fCmCmU 296 mApsfGpsmGmAmGmUmUmCmCm GmCmAmGfUmAmUmGmGmApsm UpsmC
249 mUpsmCpsfCmAmUmAfCfUfG mCmGfGmAmAmCmU fCmCmU 297 mApsfGpsmGmAmGmUmUmCmCm GmCmAmGfUmAmUmGmGmA
250 mUpsmCpsmGmUmGmGfUmGf GfAfCmUmUmCmUmCmUmCm AmAmU 298 mApsfUpsmUmGmAfGmAmGmAmA mGmUmCfCmAfCmCmAmCmGmAp smGpsmU
251 mUpsmGpsfCmAmCmUfUfCfG mCmUfUmCmAmCmCfUmCmU 299 mApsfGpsmAmGmGmUmGmAmAm GmCmGmAfAmGmUmGmCmApsmC psmA
252 mUpsmGpsfCmCmGmAfUfCfC mAmU fAmCmUmGmCfGmGm A 300 mUpsfCpsmCmGmCmAmGmUmAm UmGmGmAfUmCmGmGmCmApsm GpsmA
253 mUpsmGpsfCmCmGmAfUfCfC m AmU fAmCmUmGmCfGmGm A 301 mUpsfCpsmCmGmCmAmGmUmAm UmGmGmAfUmCmGmGmCmA
316
Table 3. 2’-O-methyl and 2’-fluoro Modified Nucléotide Sequences with Phosphorothioate Linkages
SEQ ID NO. First Nucléotide Sequence (5’3’) SEQ ID NO. Second Nucléotide Sequence (5’-3’
254 mUpsmGpsfCmUmAmUfGfCfC mUmCfAmUmCmUmUfCmUmU 302 mApsfApsmGmAmAmGmAmUmGm AmGmGmCfAmUmAmGmCmApsm GpsmC
255 mUpsmGpsfUmGmCmAfCfUfU mCmGfCmUmUmCmAfCmCmU 303 mApsfGpsmGmUmGmAmAmGmCm GmAmAmGfUmGmCmAmCmApsmC psmG
256 mUpsmGpsfUmGmCmAfCfUmU mCmGmCmUmUmCmAfCmCm U 303 mApsfGpsmGmUmGmAmAmGmCm Gm AmAmGfU mGmCm AmCmApsmC psmG
257 mUpsmGpsfUmGmCmAfCfUfU mCmGfCmUmUmCmAfCmCmU 304 mApsfGpsmGmUmGmAmAmGmCm Gm Am AmGfU mGmCm AmCrnA
258 mUpsmGpsfUmGmCmAfCfUmU mCmGmCmUmUmCmAfCmCm U 304 mApsfGpsmGmUmGmAmAmGmCm Gm AmAmGfU mGmCm AmCm A
259 mUpsmUpsfGmCmCmCfGfUmU mUmGniUmCmCmUmCfUmAm A 305 mUpsfUpsmAmGmAmGmGmAmCm AmAmAmCfGmGmGmCmAmApsmC psmA
260 mUpsmUpsfGmCmCmCfGfU fU mUmGfUmCmCmUmCfUmAmA 306 mUpsfUpsmAmGmAmGmGmAmCm AmAmAmCfGmGmGmCmAmA
mX = 2’-O-methyl nucléotide; fX = 2’-fluoro nucleotit e; ps= phosphorothioate linkage
Table 4. siNA Sequences
SEQ ID NO. Sense Sequence (5'-3*) SEQ ID NO. Antisense Sequence (5’-3*)
307 mApsmCpsfCmGmUmGfU fGfC mAmCfUmUmCmGmCfUmUmC 363 mGpsfApsmAmGmCmGmAmAmGmU mGmCmAfCmAmCmGmGmUpsmCps mC
308 mApsmCpsfCmGmUmGfU fGfC mAmCfUmUmCmGmCfUmUmC TT 364 mGpsfApsmAmGmCmGmAmAmGmU mGmCmAfCmAmCmGmGmUpsTpsT
309 mApsmCpsfUmUmCmGfCfU fU mCm AfCmCmUmCmU fGmCmA 365 mUpsfGpsmCmAmGmAmGmGmUmG mAmAmGfCmGmAmAmGmUpsmGps mC
310 mApsmGpsfUmGmUmUfUfGfC mUmGfAmCmGmCmAfAmCmC 366 mGpsfGpsmUmUmGmCmGmUmCmA mGmCmAfAmAmCmAmCmUpsmUps mG
317
Table 4. siNA Sequences
SEQ ID NO. Sense Sequence (5'-3') SEQ ID NO. Antisense Sequence (5’-3*)
311 mCpsmApsfGmGmCmGfGfGfG mUmUfUmUmUmCmUfUmGm U 367 mApsfCpsmAmAmGmAmAmAmAmA mCmCmCfCmGmCmCmUmGpsmUps mA
312 mCpsmApsfGmGmCmGfGfGfG mUmUfUmUmUmCmUfUmGm UTT 368 mApsfCpsmAmAmGmAmAmAmAmA mAmCmCmCfCmGmCmCmUmGpsTp sT
313 mCpsmApsfGmUmUmUfAfCfU mAmGfUmGmCmCmAfUmUm U 369 mApsfApsmAmUmGmGmCmAmCmU mAmGmUfAmAmAmCmUmGpsmAps mG
314 mCpsmApsfGmUmUmUfAfCfU mAmGfUmGmCmCmAfUmUm UTT 370 mApsfApsmAmUmGmGmCmAmCmU mAmGmUfAmAmAmCmUmGpsTpsT
315 mCpsmApsfUmCmCmUfGfCfU mGmCfUmAmUmGmCfCmUmC 371 mGpsfApsmGmGmCmAmUmAmGmC mAmGmCfAmGmGmAmUmGpsmAps mA .
316 mCpsmApsmUmCmCmUfGmCf UfGfCmUmAmUmGmCmCmUm CmAmU 372 mApsfUpsmGmAmGfGmCmAmUmA mGmCmAfGmCfAmGmGmAmUmGps mApsmA
317 mCpsmApsfUmCmCmUfGfCfU mGmCfUmAmUmGmCfCmUmC TT 373 mGpsfApsmGmGmCmAmUmAmGmC mAmGmCfAmGmGmAmUmGpsTpsT
318 mCpsmCpsfGmUmGmUfGfCfA mCmU fUmCmGmCm U fUmCm A 374 mUpsfGpsmAmAmGmCmGmAmAmG mUmGmCfAmCmAmCmGmGpsmUps mC
319 mCpsmCpsfGmUmGmUfGfCfA mCmU fUmCmGmCmUfUmCm A TT 375 mUpsfGpsmAmAmGmCmGmAmAmG mUmGmCfAmCmAmCmGmGpsTpsT
320 mCpsmCpsmUmGmCmUfGmCf U fAfU mGmCmCmUmCm AmUm CmUmU 376 m ApsfApsmGm AmU fGmAmGmGmC mAmUmAfGmCfAmGmCmAmGmGps mApsmU
321 mCpsmUpsfCm AmGmU fUfUfA mCmUfAmGmUmGmCfCmAmU 377 mApsfUpsmGmGmCmAmCmUmAmG mUmAmAfAmCmUmGmAmGpsmCps mC
322 mCpsmUpsmCmAmGmUfUmU mAmCmUmAmGmUmGmCmC mAmU 377 mApsfUpsmGmGmCmAmCmUmAmG mUmAmAfAmCmUmGmAmGpsmCps mC
323 mCpsmUpsfCmAmGmUfUfUmA mCmUmAmGmUmGmCfCmAm U 377 mApsfUpsmGmGmCmAmCmUmAmG mUmAmAfAmCmUmGmAmGpsmCps mC
318
Table 4. siNA Sequences
SEQ ID NO. Sense Sequence (5'-3’) SEQ ID NO. Antisense Sequence (5'-3')
324 mCpsmUpsfCm AmGmUfUfUfA mCmUfAmGmUmGmCfCmAmU TT 378 mApsfUpsmGmGmCmAmCmUmAmG mUmAmAfAmCmUmGmAmGpsTpsT
325 mCpsmUpsfCmAmGmUfUfUmA mCmUmAmGmUmGmCfCmAm UTT 378 mApsfUpsmGmGmCmAmCmUmAmG mUmAmAfAmCmUmGmAmGpsTpsT
326 mCpsmUpsfGmCmUmAfUfGfC mCmU fCm AmUmCmUfUmCmU 379 mApsfGpsmAmAmGmAmUmGmAmG mGmCmAfUmAmGmCmAmGpsmCps mA
327 mCpsmUpsfGmCmUmAfUfGmC mCmUmCmAmUmCmUfUmCm U 379 mApsfGpsmAmAmGmAmUmGmAmG mGmCmAfUmAmGmCmAmGpsmCps mA
. 328 mCpsmUpsfGmCmUmAfUfGfC mCmU fCm AmUmCmU fUmCmU TT 380 mApsfGpsmAmAmGmAmUmGmAmG mGmCmAfUmAmGmCmAmGpsTpsT
329 mCpsmUpsfGmCmUmAfUfGmC mCmUmCmAmUmCmUfUmCm UTT 380 mApsfGpsmAmAmGmAmUmGmAmG mGmCmAfUmAmGmCmAmGpsTpsT
330 mCpsmUpsfGmCmUmGfCfUfA mUmGfCmCmUmCm AfU mCmU 381 mApsfGpsmAmUmGmAmGmGmCmA mUmAmGfCmAmGmCmAmGpsmGps mA
331 mCpsmUpsfGmCmUmGfCfUfA mUmGfCmCmUmCmAfUmCmU TT 382 mApsfGpsmAmUmGmAmGmGmCmA mUmAmGfCmAmGmCmAmGpsTpsT
332 mCpsmUpsfGmCmUmGfCfUmA mUmGmCmCmUmCmAfUmCm UTT 383 mApsfGpsmAmUmGmAmGmGmCmA mUmAmGfCmAmGmCmAmGpsTpsT
333 mCpsmUpsmUmCmGmCfUmUf CfAfCmCmUmCmUmGmCmAm CmGmU 384 mApsfCpsmGmUmGfCmAmGmAmG mGmUmGfAmAfGmCmGmAmAmGp smUpsmG
334 mGpsmCpsmAmCmUmUfCmGf CfUfU mCmAmCmCm UmCmUm GmCmA 385 mUpsfGpsmCmAmGfAmGmGmUmG mAmAmGfCmGfAmAmGmUmGmCps mApsmC
335 mGpsmCpsfCmGmAmUfCfCfA mUmAfCmUmGrnCmGfGmAm A 386 mUpsfUpsmCmCmGmCmAmGmUmA mUmGmGfAmUmCmGmGmCpsmAps mG
336 mGpsmCpsfCmGmGmGfUfUfU mUmUfCmUmUmGmUfUmGm ATT 387 mUpsfUpsmCmCmGmCmAmGmUmA mUmGmGfAmUmCmGmGmCpsTpsT
319
Table 4. siNA Sequences
SEQ ID NO. Sense Sequence (5'-3') SEQ ID NO. Antisense Sequence (5’-3*)
337 mGpsmCpsfGmGmGmGfU fU fU mUmUfCmUmUmGmUfUmGm A 388 mUpsfCpsmAmAmCmAmAmGmAmA mAmAmAfCmCmCmCmGmCpsmCps mU
338 mGpsmCpsfGmGmGmGfU fUmU mUmUmCmUmUmGmUfUmGm A 388 mUpsfCpsmAmAmCmAmAmGmAmA mAmAmAfCmCmCmCmGmCpsmCps mU
339 mGpsmCpsfGmGmGmGfUfUfU mUmUfCmUmUmGmUfUmGm ATT 389 mUpsfCpsmAmAmCmAmAmGmAmA mAmAmAfCmCmCmCmGmCpsTpsT
340 mGpsmCpsfGmGmGmGfU fU mU mUmUmCmUmUmGmUfUmGm ATT 389 mUpsfCmAmAmCmAmAmGmAmAm AmAmAfCmCmCmCmGmCpsTpsT
341 mGpsmCpsmUmGmCmUfAmUf GfCfCmUmCmAmUmCmUmUm CmUmU 390 mApsfApsmGmAmAfGmAmUmGmA mGmGmCfAmUfAmGmCmAmGmCps mApsmG
342 mGpsmGpsm AmUmGmU fGmU f CfU fGmCmGmGmCmGmUmUm UmUmA 391 mUpsfApsmAmAmAfCmGmCmCmG mCmAmGfAmCfAmCmAmUmCmCps mApsmG
343 mGpsmGpsfCmCmAmAfAfAfU mUmCfGmCmAmGmU fCmCmC 392 mGpsfGpsmGmAmCmUmGmCmGmA mAmUmU fU mUmGmGmCmCpsm Aps mA
344 mGpsmGpsfCmGmCmAfCfCfU mCmU fCmUmUmUm AfCmGmC 393 mGpsfCpsmGmUmAmAmAmGmAmG mAmGmGfUmGmCmGmCmCpsmCps mC
345 mGpsmUpsfAmUmGmUfUfGfC mCmCfGmUmUmUmGfUmCmC 394 mGpsfGpsmAmCmAmAmAmCmGmG mGmCmAfAmCmAmUmAmCpsmCps mU
346 mGpsmUpsfGmGmUmGfGfAfC mUmUfCmUmCmUmCfAmAmU 395 mApsfUpsmUmGmAmGmAmGmAmA mGmUmCfCmAmCmCmAmCpsmGps mA
347 mGpsmUpsfGmUmGmCfAfCfU mUmCfGmCmUmUmCfAmCmC 396 mGpsfGpsmUmGmAmAmGmCmGmA m AmGmU fGmCm AmCm AmCpsmGps mG
348 mGpsmUpsfUmGmCmCfCfGfU mUmU fGmUmCmCmU fCmUm A 397 mUpsfApsmGmAmGmGmAmCmAmA mAmCmGfGmGmCmAmAmCpsmAps mU
349 mGpsmUpsfUmGmCmCfCfGfU mUmUfGmUmCmCmUfCmUmA TT 398 mUpsfApsmGmAmGmGmAmCmAmA m AmCmGfGmGmCm Am AmCpsT psT
320
Table 4. siNA Sequences
SEQ ID NO. Sense Sequence (5'-3') SEQ ID NO. Antisense Sequence (5'-3')
350 mUpsmCpsfCm AmUmAfCfUfG mCmGfGm Am AmCmU fCmCmU 399 mApsfGpsmGmAmGmUmUmCmCmG mCmAmGfUmAmUmGmGmApsmUps mC
351 mUpsmCpsfCmAmUmAfCfUfG mCmGfGm AmAmCmU fCmCmU TT 400 mApsfGpsmGmAmGmUmUmCmCmG mCmAmGfUmAmUmGmGmApsTpsT
352 mUpsmCpsmGmUmGmGfUmGf GfAfCmUmUmCmUmCmUmCm AmAmU 401 mApsfUpsmUmGmAfGmAmGmAmA mGmUmCfCmAfCmCmAmCmGmAps mGpsmU
353 mUpsmGpsfCmAmCmUfUfCfG mCmU fU mCm AmCmCfUmCmU 402 mApsfGpsmAmGmGmUmGmAmAmG mCmGmAfAmGmUmGmCmApsmCps mA
354 mUpsmGpsfCmCmGmAfUfCfC mAmUfAmCmUmGmCfGmGm A 403 mUpsfCpsmCmGmCmAmGmUmAmU mGmGmAfUmCmGmGmCmApsmGps mA
355 mUpsmGpsfCmCmGmAfU fCfC mAmUfAmCmUmGmCfGmGm ATT 404 mUpsfCpsmCmGmCmAmGmUmAmU mGmGmAfUmCmGmGmCmApsTpsT
356 mUpsmGpsfCmUmAmUfGfCfC mUmCfAmUmCmUmUfCmUmU 405 mApsfApsmGmAmAmGmAmUmGmA mGmGmCfAmUmAmGmCmApsmGps mC
357 mUpsmGpsfUmGmCmAfCfUfU mCmGfCmUmUmCmAfCmCmU 406 mApsfGpsmGmUmGmAmAmGmCmG mAmAmGfUmGmCmAmCmApsmCps mG
358 mUpsmGpsfUmGmCmAfCfUmU mCmGmCmUmUmCmAfCmCm U 406 mApsfGpsmGmUmGmAmAmGmCmG mAmAmGfUmGmCmAmCmApsmCps mG
359 mUpsmGpsfUmGmCmAfCfUfU mCmGfCmUmUmCmAfCmCmU TT 407 mApsfGpsmGmUmGmAmAmGmCmG mAmAmGfUmGmCmAmCmApsTpsT
360 mUpsmGpsfUmGmCmAfCfUmU mCmGmCmUmUmCmAfCmCm UTT 407 mApsfGpsmGmUmGmAmAmGmCmG mAmAmGfUmGmCmAmCmApsTpsT
361 mUpsmUpsfGmCmCmCfGfUmU mUmGmUmCmCmUmCfUmAm A 408 mUpsfUpsmAmGmAmGmGmAmCmA mAmAmCfGmGmGmCmAmApsmCps mA
362 mUpsmUpsfGmCmCmCfGfUfU mUmGfUmCmCmUmCfUmAmA TT 409 mUpsfUpsmAmGmAmGmGmAmCmA mAmAmCfGmGmGmCmAmApsTpsT
321
Table 4. siNA Sequences
SEQ ID NO. Sense Sequence (5'-3') SEQ ID NO. Antisense Sequence (5'-3')
415 5dcd3Cps5dcd3CpsfG5dcd3UmG 5dcd3UfG5dfCfA5dcd3C5dcd3U 5dfU5dcd3CmG5dcd3C5dcd3U5d fU5dcd3CmA 445 5 dcd3UpsfGpsmAmAmG5 dcd3 CmGm AmAmG5dcd3UmG5dcd3CfA5dcd3C mA5dcd3CmGmGps5dcd3Ups5dcd3U
415 5dcd3Cps5dcd3CpsfG5dcd3UmG 5dcd3UfG5dfCfA5dcd3C5dcd3U 5dfU5dcd3CmG5dcd3C5dcd3U5d fU5dcd3CmA 446 5dcd3UpsfGpsmAmAmGmCmGmAm AmGmUmGmCfAmCmAmCmGmGps 5dcd3Ups5dcd3U
415 5dcd3Cps5dcd3CpsfG5dcd3UmG 5dcd3UfG5dfCfA5dcd3C5dcd3U 5dfU5dcd3CmG5dcd3C5dcd3U5d fU5dcd3CmA 447 mUpsfGpsmAmAmGmCmGmAmAmG mUmGmCfAmCmAmCmGmGpsmUps mU
416 fCpsmCpsfGmUfGmUfGfCfAmC fUmUfCmGfCmUfUmCfA 448 mUpsfGpsmAfAmGfCmGfAmAfGmU mGmCfAmCfAmCfGmGpsmUpsmC
416 fCpsmCpsfGmUfGmUfGfCfAmC fUmUfCmGfCmUfUmCfA 449 vmUpsfGpsmAfAmGfCmGfAmAfGm UmGmCfAmCfAmCfGmGpsmUpsmC
416 fCpsmCpsfGmU fGmU fGmCfAm CfUmUfCmGfCmUfUmCfA 450 mUpsfGpsmAfAmGfCmGfAmAfGmU f GmCfAmCfAmCfGmGpsmUpsmC
416 fCpsmCpsfGmU fGmU fGmCfAm CfUmUfCmGfCmUfUmCfA 451 VmUpsfGpsmAfAmGfCmGfAmAfGm UfGmCfAmCfAmCfGmGpsmUpsmC
417 fCpsmUpsfGmCfUmAfUmGfCm CfUmCfAmUfCmUfUmCfU 452 mApsfGpsmAfAmGfAmUfGmAfGmGf CmAfUmAfGmCfAmGpsmUpsmU
417 fCpsmUpsfGmCfUmAfUmGfCm CfUmCfAmUfCmUfUmCfU 452 mApsfGpsmAfAmGfAmUfGmAfGmGf CmAfUmAfGmCfAmGpsmUpsmU
417 fCpsmUpsfGmCfUmAfUmGfCm CfUmCfAmUfCmUfUmCfU 452 mApsfGpsmAfAmGfAmUfGmAfGmGf CmAfUmAfGmCfAmGpsmUpsmU
417 fCpsmUpsfGmCfUmAfUmGfCm CfUmCfAmUfCmUfUmCfU 452 mApsfGpsmAfAmGfAmUfGmAfGmGf CmAfUmAfGmCfAmGpsmUpsmU
418 fGpsmUpsfGmGfUmGfGfAfCm UfUmCfUmCfUmCfAmAfU 453 mApsfUpsmUfGmAfGmAfGmAfAmG mUmCfCmAfCmCfAmCpsmGpsmA
418 fGpsmUpsfGmGfUmGfGfAfCm UfUmCfUmCfUmCfAmAfU 454 vmApsfUpsmUfGmAfGmAfGmAfAm GmUmCfCmAfCmCfAmCpsmGpsmA
419 fGpsmUpsfGmGfUmGfGmAfCm UfUmCfUmCfUmCfAmAfU 455 mApsfUpsmUfGmAfGmAfGmAfAmGf UmCfCmAfCmCfAmCpsmGpsmA
419 fGpsmUpsfGmGfUmGfGmAfCm UfUmCfUmCfUmCfAmAfU 456 VmApsfUpsmUfGmAfGmAfGmAfAm GfUmCfCmAfCmCfAmCpsmGpsmA
420 mCpsmCpsfGmUfGmUfGfCfAm CmUmUmCmGmCmUmUmCm A 457 mUpsfGpsmAmAmGfCmGmAmAmG mUmGmCfAmCfAmCmGmGpsmUps mC
322
Table 4. siNA Sequences
SEQ ID NO. Sense Sequence (5'-3') SEQ ID NO. Antisense Sequence (5’-3*)
421 mCpsmCpsfGmUmGmUfGfCfA mAmUfUmCmGmCmUfUmCmA 458 mUpsfGpsmAmAmGmCmGmAmAmG mUmGmCfAmCmAmCmGmGpsmUps mC
422 mCpsmCpsfGmUmGmUfGfCfA mCmUfUmCmGmCmUfUmCmA 459 mUpsfGpsmAmAmGmCmGmAmAmG mUmGmCfAmCmAmCmGmGpsmU3 s mU
423 mCpsmCpsfGmUmGmUfGfCfA mCmUfUmCmGmCmUfUmCmA 460 mUpsfGpsmAmAmGmCmGmAmAmG mUmGmCfAmCmAmCmGmG5smU5s mU
423 mCpsmCpsfGmUmGmUfGfCfA mCmUfUmCmGmCmU fUmCm A 461 mUpsfGpsmAmAmGmCmGmAmAmG mUmGmCfAmCmAmCmGmGpsmU 5 s mU
423 mCpsmCpsfGmUmGmUfGfCfA mCmUfUmCmGmCmUfUmCmA 458 mUpsfGpsmAmAmGmCmGmAmAmG mUmGmCfAmCmAmCmGmGpsmUps mC
423 mCpsmCpsfGmUmGmUfGfCfA mCmUfUmCmGmCmUfUmCmA 458 mUpsfGpsmAmAmGmCmGmAmAmG mUmGmCfAmCmAmCmGmGpsmUps mC
423 mCpsmCpsfGmUmGmUfGfCfA mCmU fUmCmGmCmUfUmCmA 458 mUpsfGpsmAmAmGmCmGmAmAmG mUmGmCfAmCmAmCmGmGpsmUps mC
423 mCpsmCpsfGmUmGmUfGfCfA mCmU fU mCmGmCm U fUmCm A 462 VmUpsfGpsmAmAmGmCmGmAmAm GmUmGmCfAmCmAmCmGmGpsmU psmC
423 mCpsmCpsfGmUmGmUfGfCfA mCmUfUmCmGmCmUfUmCmA 463 VmUpsfGpsmAmAmGmCmGmAmAm GmUmGmCfAmCmAmCmGmGpsTps T
423 mCpsmCpsfGmUmGmUfGfCfA mCmU fUmCmGmCmU fUmCm A 447 mUpsfGpsmAmAmGmCmGmAmAmG mUmGmCfAmCmAmCmGmGpsmUps mU
423 mCpsmCpsfGmUmGmUfGfCfA mCmUfUmCmGmCmUfUmCmA 458 mUpsfGpsmAmAmGmCmGmAmAmG mUmGmCfAmCmAmCmGmGpsmUps mC
423 mCpsmCpsfGmUmGmU fGfCfA mCmU fU mCmGmCmU fUmCm A 457 mUpsfGpsmAmAmGfCmGmAmAmG mUmGmCfAmCfAmCmGmGpsmUps mC
423 mCpsmCpsfGmUmGmUfGfCfA mCmU fUmCmGmCmU fU mCm A 464 mUpsfGpsmAmAmGfCmGmAmAfGm UmGmCfAmCmAmCfGmGpsmUpsmC
323
Table 4. siNA Sequences
SEQ ID NO. Sense Sequence (5'-3') SEQ ID NO. Antisense Sequence (5'-3')
423 mCpsmCpsfGmUmGmUfGfCfA mCmUfUmCmGmCmUfUmCmA 465 vmBpsfGpsmAmAmGmCmGmAmAm GmUmGmCfAmCmAmCmGmGpsmU psmC
423 mCpsmCpsfGmUmGmUfGfCfA mCmUfUmCmGmCmUfUmCmA 466 mesnmUpsfGpsmAmAmGmCmGmAm AmGmUmGmCfAmCmAmCmGmGps mUpsmC
423 mCpsmCpsfGmUmGmUfGfCfA mCmUfUmCmGmCmU fU mCm A 467 cmUpsfGpsmAmAmGmCmGmAmAm GmUmGmCfAmCmAmCmGmGpsmU psmC
423 mCpsmCpsfGmUmGmUfGfCfA mCmUfUmCmGmCmU fUmCm A 468 mesnomUpsfGpsmAmAmGmCmGmA inAmGmUmGmCfAmCmAmCmGmGp smUpsmC
423 mCpsmCpsfGmUmGmUfGfCfA mCmUfUmCmGmCmUfUmCmA 469 mUpsfGpsmAmAmGfCmGmAmAmG mUmGmCfAmCmAmCmGmGpsmUps mC
423 mCpsmCpsfGmUmGmUfGfCfA mCmUfUmCmGmCmUfUmCmA 470 mUpsfGpsmAmAmGmCmGmAmAmG fUmGmCfAmCmAmCmGmGpsmUps mC
423 mCpsmCpsfGmUmGmUfGfCfA mCmU fU mCmGmCmUfUmCm A 471 mUpsfGpsmAmAmGmCmGmAmAmG mUfGmCfAmCmAmCmGmGpsmUps mC
423 mCpsmCpsfGmUmGmUfGfCfA mCmUfUmCmGmCmU fUmCm A 472 mUpsfGpsmAmAmGmCmGmAmAmG mUmGmCfAmCfAmCmGmGpsmUps mC
423 mCpsmCpsfGmUmGmUfGfCfA mCmU fUmCmGmCmU fUmCm A 473 mUpsfGpsmAmAmGmCmGmAmAmG mUmGmCfAmCmAfCmGmGpsmUps mC
423 mCpsmCpsfGmUmGmUfGfCfA mCmU fUmCmGmCmU fUmCm A 474 mUpsfGpsmAmAmGmCmGmAmAmG mUmGmCfAmCmAmCfGmGpsmUps mC
423 mCpsmCpsfGmUmGmUfGfCfA mCmUfUmCmGmCmUfUmCmA 475 mUpsfGpsmAmAmGmCmGmAmAmG mUmGmCfAmCmAmCmGfGpsmUps mC
423 mCpsmCpsfGmUmGmUfGfCfA mCmUfUmCmGmCmUfUmCmA 476 mUpsfGpsmAfAmGmCmGmAmAmG mUmGmCfAmCmAmCmGmGpsmUps mC
423 mCpsmCpsfGmUmGmUfGfCfA mCmUfUmCmGmCmUfUmCmA 477 mUpsfGpsmAmAfGmCmGmAmAmG mUmGmCfAmCmAmCmGmGpsmUps mC
324
Table 4. siNA Sequences
SEQ ID NO. Sense Sequence (5'-3') SEQ ID NO. Antisense Sequence (5'-3')
423 mCpsmCpsfGmUmGmUfGfCfA mCmUfUmCm GmCm U fU mCm A 478 mUpsfGpsmAmAmGmCfGmAmAmG mUmGmCfAmCmAmCmGmGpsmUps mC
423 mCpsmCpsfGmUmGmUfGfCfA mCmU fUmCmGmCmUfUmCm A 479 mUpsfGpsmAmAmGmCmGfAmAmG mUmGmCfAmCmAmCmGmGpsmUps mC
423 mCpsmCpsfGmUmGmUfGfCfA mCmUfUmCmGmCmUfUmCmA 480 mUpsfGpsmAmAmGmCmGmAfAmG mUmGmCfAmCmAmCmGmGpsmUps mC
423 mCpsmCpsfGmUmGmUfGfCfA mCmUfUmCmGmCmU fUmCm A 481 mUpsfGpsmAmAmGmCmGmAmAfG mUmGmCfAmCmAmCmGmGpsmUps mC
423 mCpsmCpsfGmUmGmUfGfCfA mCmUfUmCmGmCmUfUmCmA 482 d2vmUpsfGpsmAmAmGmCmGmAmA mGmUmGmCfAmCmAmCmGmGpsm UpsmC
423 mCpsmCpsfGmUmGmUfGfCfA mCmUmUmCmGmCmUfUmCm A 483 mUpsfGpsmAfAmGfCmGfAmAmGm UmGmCfAmCmAmCmGmGpsmUpsm C
597 mCpsmCpsfGmUmGmUfGfCfC mCmU fU mCmGmCmU fUmCm A 458 mUpsfGpsmAmAmGmCmGmAmAmG mUmGmCfAmCmAmCmGmGpsmUps mC
423 mCpsmCpsfGmUmGmUfGfCmA mCmUmUmCmGmCmUfUmCm A 458 mUpsfGpsmAmAmGmCmGmAmAmG mUmGmCfAmCmAmCmGmGpsmUps mC
424 mCpsmCpsmGmUfGmUfGfCfA mCmUmUmCmGmCmUmUmC mA 457 mUpsfGpsmAmAmGfCmGmAmAmG mUmGmCfAmCfAmCmGmGpsmUps mC
424 mCpsmCpsmGmUfGmUfGfCfA mCmUmUmCmGmCmUmUmC mA 484 mUpsfGpsmAmAmGfCmGfAfAmGm UmGmCfAmCfAmCmGmGpsmUpsmC
424 mCpsmCpsmGmUfGmUfGfCfA mCmUmUmCmGmCmUmUmC mA 485 mUpsfGpsmAmAfGmCmGfAmAmGm UmGmCfAmCmAfCmGmGpsmUpsmC
425 mCpsmUpsmGmCfUmAfUfGfC mCmUmCmAmUmCmUmUmC mU 486 mApsfGpsmAmAmGfAmUmGmAfGm GmCmAfUmAmGmCfAmGpsmCpsm A
425 mCpsmUpsmGmCfUmAfUfGfC mCmUmCmAmUmCmUmUmC mU 487 mApsfGpsmAmAmGfAmUmGmAmG mGmCmAfUmAfGmCmAmGpsmCps mA
325
Table 4. siNA Sequences
SEQ ID NO. Sense Sequence (5'-3') SEQ ID NO. Antisense Sequence (5'-3')
425 mCpsmUpsmGmCfUmAfUfGfC mCmUmCmAmUmCmUmUmC mU 488 mApsfGpsmAmAfGmAmUfGmAmGm GmCmAfUmAmGfCmAmGpsmCpsm A
426 mCpsmUpsmGmCfUmAfUmGfC mCmUfCmAmUmCmUfUmCmU 489 mApsfGpsmAmAmGmAmUmGmAmG inGmCmAfUmAmGmCmAmGpsmCps mA
427 HiGpsmCpsfGmGmGmGfU fU fU mUmU fCmUmUmGmU fU mGm A 490 vmUpsfCpsmAmAmCmAmAmGmAm AmAmAmAfCmCmCmCmGmCpsmCp smU
428 mGpsmCpsfGmGmGmGfUmUm UmUmUmCmUmUmGmUfUmG mA 491 mUpsfCpsmAmAmCmAmAmGmAmA mAmAmAfCmCmCmCmGmCpsmCps mU
429 mGpsmCpsmGmGfGmGfU fU fU mUmUmCmUmUmGmUmUmG mA 492 mUpsfCpsmAmAmCfAmAinGmAfAm AmAmAfCmCmCmCfGmCpsmCpsmU
429 mGpsmCpsmGmGfGmGfUfUfU mUmUmCmUmUmGmUmUmG mA 493 mUpsfCpsmAmAmCfAmAmGmAmA mAmAmAfCmCfCmCmGmCpsmCpsm U
429 mGpsmCpsmGmGfGmGfUfUfU mUmUmCmUmUmGmUmUmG mA 494 mUpsfCpsmAmAfCmAmAfGmAmAm AmAmAfCmCmCfCmGmCpsmCpsmU
430 mGpsmCpsmUmGfCmUmAmUf GfCfCmUmCfAmUmCmUmUfC mUmU 495 mApsfApsmGmAmAmGmAmUmGmA mGmGmCfAmUmAmGmCmAmGmCp smApsmG
430 mGpsmCpsmUmGfCmUmAmU f GfCfCmUmCfAmUmCmUmUfC mUmU 496 mApsfApsmGmAmAfGmAmUmGmA mGmGmCfAmUfAmGmCmAmGmCps mApsmG
431 mGpsmCpsmUmGmCmU fAmU f GfCfCmUmCmAmUfCmUmUfC mUmU 497 m ApsfApsmGm AmAfGm AfU fGm Am GmGmCfAmU fAmGmCm AmGmCpsm ApsmG
432 mGpsmCpsmUmGmCmU fAmU f GfCfCmUmCmAmUmCmUmUm CmUmU 498 vmApsfApsmGmAmAfGmAmUmGmA mGmGmCfAmUfAmGmCmAmGmCps mApsmG
432 mGpsmCpsmUmGmCmUfAmUf GfCfCmUmCmAmUmCmUmUm CmUmU 500 d2vmApsfApsmGmAmAfGmAmUmG mAmGmGmCfAmU fAmGmCmAmGm CpsmApsmG
433 mGpsmUpsfGmGfUmGfGfAfCm UmUmCmUmCmUmCmAmAm U 501 mApsfUpsmUmGmAfGmAmGmAmA mGmUmCfCmAfCmCmAmCpsmGpsm A
326
Table 4. siNA Sequences
SEQ ID NO. Sense Sequence (5*-3*) SEQ ID NO. Antisense Sequence (5'-3')
434 mGpsmUpsfGmGmUmGfGfAfA mUmUfCmUmCmUmCfAmAmU 502 mApsfUpsmUmGmAmGmAmGmAmA mGmUmCfCmAmCmCmAmCpsmGps mA
598 mGpsmUpsfGmGmUmGfGfAfC mGmUfCmUmCmUmCfAmAmU 502 mApsfUpsmUmGmAmGmAmGmAmA mGmUmCfCmAmCmCmAmCpsmGps mA
435 mGpsmUpsfGmGmUmGfGfAfC mUmUfCmUmCmUmCfAmAmU 503 vmApsfUpsmUmGmAmGmAmGmAm AmGmUmCfCmAmCmCmAmCpsmGp smA
435 mGpsmUpsfGmGmUmGfGfAfC mUmUfCmUmCmUmCfAmAmU 501 mApsfUpsmUmGmAfGmAmGmAmA mGmUmCfCmAfCmCmAmCpsmGpsm A
435 mGpsmUpsfGmGmUmGfGfAfC mUmUfCmUmCmUmCfAmAmU 504 mApsfUpsmUmGmAfGmAmGmAfAm GmUmCfCmAmCmCfAmCpsmGpsmA
435 mGpsmUpsfGmGmUmGfGfAfC mUmUfCmUmCmUmCfAmAmU 505 vmNpsfUpsmUmGmAmGmAmGmAm AmGmUmCfCmAmCmCmAmCpsmGp smA
435 mGpsmUpsfGmGmUmGfGfAfC mUmUfCmUmCmUmCfAmAmU 506 vmUpsfUpsmUmGmAmGmAmGmAm AmGmUmCfCmAmCmCmAmCpsmGp smA
435 mGpsmUpsfGmGmUmGfGfAfC mUmUfCmUmCmUmCfAmAmU 507 cmUpsfUpsmUmGmAmGmAmGmAm AmGmUmCfCmAmCmCmAmCpsmGp smA
435 mGpsmUpsfGmGmUmGfGfAfC mUmUfCmUmCmUmCfAmAmU 508 mesnmUpsfUpsmUmGmAmGmAmGm AmAmGmUmCfCmAmCmCmAmCps mGpsmA
435 mGpsmUpsfGmGmUmGfGfAfC mUmUfCmUmCmUmCfAmAmU 509 mesnomUpsfUpsmUmGmAmGmAmG mAmAmGmUmCfCmAmCmCmAmCp smGpsmA
435 mGpsmUpsfGmGmUmGfGfAfC mUmUfCmUmCmUmCfAmAmU 510 mApsfUpsmUmGmAfGmAmGmAmA mGmUmCfCmAmCmCmAmCpsmGps mA
435 mGpsmUpsfGmGmUmGfGfAfC mUmUfCmUmCmUmCfAmAmU 511 m ApsfUpsmU fGm AmGrn AmGmAmA mGmUmCfCmAmCmCmAmCpsmGps mA
435 mGpsmUpsfGmGmUmGfGfAfC mUmUfCmUmCmUmCfAmAmU 512 mApsfUpsmUmGfAmGmAmGmAmA mGmUmCfCmAmCmCmAmCpsmGps mA
327
Table 4. siNA Sequences
SEQ ID NO. Sense Sequence (5'-3') SEQ ID NO. Antisense Sequence (5'-3')
435 mGpsmUpsfGmGmUmGfGfAfC mUmUfCmUmCmUmCfAmAmU 513 mApsfUpsmUmGmAmGfAmGmAmA mGmUmCfCmAmCmCmAmCpsmGps mA
435 mGpsmUpsfGmGmUmGfGfAfC mUmUfCmUmCmUmCfAmAmU 514 mApsfUpsmUmGmAmGmAmGfAmA mGmUmCfCmAmCmCmAmCpsmGps mA
435 mGpsmUpsfGmGmUmGfGfAfC mUmUfCmUmCmUmCfAmAmU 515 mApsfUpsmUmGmAmGmAmGmAfA mGmUmCfCmAmCmCmAmCpsmGps mA
435 mGpsmUpsfGmGmUmGfGfAfC mUmUfCmUmCmUmCfAmAmU 516 mApsfUpsmUmGmAmGmAmGmAmA mGmUmCfCmAfCmCmAmCpsmGpsm A
435 mGpsmUpsfGmGmUmGfGfAfC mUmUfCmUmCmUmCfAmAmU 517 mApsfUpsmUmGmAmGmAmGmAmA mGmUmCfCmAmCmCfAmCpsmGpsm A
435 mGpsmUpsfGmGmUmGfGfAfC mUmUfCmUmCmUmCfAmAmU 518 mApsfUpsmUmGmAmGmAmGmAmA mGmUmCfCmAmCmCmAfCpsmGpsm A
435 mGpsmUpsfGmGmUmGfGfAfC mUmUfCmUmCmUmCfAmAmU 519 d2vmApsfUpsmUmGmAmGmAmGmA mAmGmUmCfCmAmCmCmAmCpsm GpsmA
436 mGpsmUpsfGmGmUmGfGfAfC mUmUmCmUmCmUmCfUmAm U 520 mApsfUpsmUfGmAfGmAfGmAmAm GmUmCfCmAmCmCmAmCpsmGpsm A
437 mGpsmUpsfGmGmUmGfGfAmC mUmUmCmUmCmUmCfAmAm U 502 mApsfUpsmUmGmAmGmAmGmAmA mGmUmCfCmAmCmCmAmCpsmGps mA
438 mGpsmUpsmGmGfUmGfGfAfC mUmUmCmUmCmUmCmAmA mU 501 mApsfUpsmUmGmAfGmAmGmAmA mGmUmCfCmAfCmCmAmCpsmGpsm A
438 mGpsmUpsmGmGfUmGfGfAfC mUmUmCmUmCmUmCmAmA mU 521 mApsfUpsmUmGmAfGmAfGfAmAm GmUmCfCmAfCmCmAmCpsmGpsmA
438 mGpsmUpsmGmGfUmGfGfAfC mUmUmCmUmCmUmCmAmA mU 522 mApsfUpsmUmGfAmGmAfGmAmAm GmUmCfCmAmCfCmAmCpsmGpsmA
439 mUpsmCpsmGmUmGmGfUmGf GfAfCmUmUmCmUmCmUmCm AmAmU 523 vmApsfUpsmUmGmAfGmAmGmAmA mGmUmCfCmAfCmCmAmCmGmAps mGpsmU
328
Table 4. siNA Set uences
SEQ ID NO. Sense Sequence (5'-3') SEQ ID NO. Antisense Sequence (5'-3')
440 mUpsmGpsfCmCmGm AfU fCfC mAmUfAmCmUmGmCfGmGm A 524 vmUpsfCpsmCmGmCmAmGmUmAm UmGmGmAfUmCmGmGmCmApsmG psmA
441 mUpsmGpsfUmGmCm AfCfU fU mCmGfCmUmUmCmAfCmCmU 525 VmApsfGpsmGmUmGmAmAmGmCm GmAmAmGfUmGmCmAmCmApsmC psmG
441 mUpsmGpsfUmGmCmAfCfUfU mCmGfCmUmUmCmAfCmCmU 526 mApsfGpsmGmUmGfAmAmGmCfGm AmAmGfUmGmCmAfCmApsmCpsm G
441 mUpsmGpsfUmGmCmAfCfUfU mCmGfCmUmUmCmAfCmCmU 527 mApsfGpsmGmUmGfAmAmGmCmG mAmAmGfUmGfCmAmCmApsmCps mG
442 mUpsmGpsfUmGmCmAfCfUmU mCmGmCmUmUmCmAfCmCm U 525 VmApsfGpsmGmUmGmAmAmGmCm GmAmAmGfUmGmCmAmCmApsmC psmG
442 mUpsmGpsfUmGmCmAfCfUmU mCmGmCmUmUmCmAfCmCm U 528 VmApsfGpsmGmUmGmAmAmGmCm GmAmAmGfUmGmCmAmCmApsTps T
442 mUpsmGpsfUmGmCmAfCfUmU mCmGmCmUmUmCmAfCmCm U 529 d2vmApsfGpsmGmUmGmAmAmGmC mGmAmAmGfUmGmCmAmCmApsm CpsmG
443 unCpsmCpsfGmUmGmU fGfCfA mCmUfUmCmGmCmUfUmCmA 458 mUpsfGpsmAmAmGmCmGmAmAmG mUmGmCfAmCmAmCmGmGpsmUps mC
444 unGpsmUpsfGmGmUmGfGfAfC mUmUfCmUmCmUmCfAmAmU 502 mApsfUpsmUmGmAmGmAmGmAmA mGmUmCfCmAmCmCmAmCpsmGps mA
423 mCpsmCpsfGmUmGmUfGfCfA mCmU fUmCmGmCmU fUmCmA 458 mUpsfGpsmAmAmGmCmGmAmAmG mUmGmCfAmCmAmCmGmGpsmUps mC
224 mCpsmUpsfGmCmUmAfUfGfC mCmU fCmAmUmCmU fU mCmU 489 mApsfGpsmAmAmGmAmUmGmAmG mGmCmAfUmAmGmCmAmGpsmCps mA
236 mGpsmCpsfGmGmGmGfUfUmU mUmUmCmUmUmGmUfUmGm A 491 mUpsfCpsmAmAmCmAmAmGmAmA mAmAmAfCmCmCmCmGmCpsmCps mU
432 mGpsmCpsmUmGmCmUfAmUf GfCfCmUmCmAmUmCmUmUm CmUmU 496 mApsfApsmGmAmAfGmAmUmGmA mGmGmCfAmU fAmGmCm AmGmCps mApsmG
329
Table 4. siNA Sequences
SEQ ID NO. Sense Sequence (5'-3') SEQ ID NO. Antisense Sequence (5'-3')
435 mGpsmUpsfGmGmUmGfGfAfC mUmUfCmUmCmUmCfAmAmU 502 mApsfUpsmUmGmAmGmAmGmAmA mGmUmCfCmAmCmCmAmCpsmGps mA
442 mUpsmGpsfUmGmCmAfCfUmU mCmGmCmUmUmCmAfCmCm U 530 mApsfGpsmGmUmGmAmAmGmCmG mAmAmGfUmGmCmAmCmApsmCps mG
439 mUpsmCpsmGmUmGmGfUmGf GfAfCmUmUmCmUmCmUmCm AmAmU 531 mApsfUpsmUmGmAfGmAmGmAmA mGmUmCfCmAfCmCmAmCmGmAps mGpsmU
423 mCpsmCpsfGmUmGmUfGfCfA mCmU fU mCmGmCmU fUmCm A 532 mUpsfGpsmAmAmGmCmGmAmAmG mUmGmCfAmCmAmCmGmGpsTpsT
441 mUpsmGpsfUmGmCmAfCfUfU mCmGfCmUmUmCmAfCmCmU 530 mApsfGpsmGmUmGmAmAmGmCmG mAmAmGfUmGmCmAmCmApsmCps mG
442 mUpsmGpsfUmGmCmAfCfUmU mCmGmCmUmUmCmAfCmCm U 533 mApsfGpsmGmUmGmAmAmGmCmG mAmAmGfUmGmCmAmCmApsTpsT
424 mCpsmCpsmGmUfGmUfGfCfA mCmUmUmCmGmCmUmUmC mA 536 d2vd3UpsfGpsmAmAfGmCmGfAmAm GmUmGmCfAmCmAfCmGmGpsmUp smC
438 mGpsmUpsmGmGfUmGfGfAfC mUmUmCmUmCmUmCmAmA mU 537 mApsf4PpsmUmGmAfGmAmGmAmA mGmUmCfCmAfCmCmAmCpsmGpsm A
438 mGpsmUpsmGmGfUmGfGfAfC mUmUmCmUmCmUmCmAmA mU 538 mApsfUpsmUmGmAfGmAmGmAmA mGmUmCfZPmAfCmCmAmCpsmGps mA
438 mGpsmUpsmGmGfUmGfGfAfC mUmUmCmUmCmUmCmAmA mU 599 mApsfUpsmUmGmAfGmAmGmAmA mGmUmCfCmAfXmCmAmCpsmGps mA
mX = 2’-O-methyl nucléotide; fX = 2’-fluoro nucléotide; 5dcd3X = nucléotide of Formula 17; 5dfX = nucléotide of Formula 16; vX= 5’ vinyl phosphonate nucléotide; d2vX = deuterated 5’ vinyl phosphonate nucléotide; vmX = 5’ vinyl phosphonate, 2’-O-methyl nucléotide; vmB = 0 ho.£ HO ? HO„° HO X 0 HO 0 N—( H0 / Y\ \ / Y <· -> A // v—-, <z ? P V-' 0 OMe î /° 1 f 1 . -vU ; vmN = · ymu =
330
Table 4. siNA Sequences
SEQ ID NO. Sense Sequence (5'-3') SEQ ID NO. Antisense Sequence (5*-3*)
0' tOMe 0' OC H 3 cmU = ; mesnmU = zwvw ; mesnomU =
o bcH3 9 OMe ; d2vmU = zvwwx · J2vmA =
O
phosphorothioate linkage
Table 5
SEQ ID NO: Description Sequence+
410 Hepatitis B virus (Genbank Accession No. U95551.1) aattccacaacctttcaccaaactctgcaagatcccagagtgagaggcctgtatttccctgctggtgg ctccagttcaggagcagtaaaccctgttccgactactgcctctcccttatcgtcaatcttctcgaggatt ggggaccctgcgctgaacatggagaacatcacatcaggattcctaggaccccttctcgtgttacagg cggggtttttcttgttgacaagaatcctcacaataccgcagagtctagactcgtggtggacttctctca attttctagggggaactaccgtgtgtcttggccaaaattcgcagtccccaacctccaatcactcacca acctcctgtcctccaacttgtcctggttatcgctggatgtgtctgcggcgttttatcatcttcctcttcatc
331
Table 5
SEQ ID NO: Description Sequence+
ctgctgctatgcctcatcttcttgttggttcttctggactatcaaggtatgttgcccgtttgtcctctaattc caggatcctcaaccaccagcacgggaccatgccgaacctgcatgactactgctcaaggaacctcta tgtatccctcctgttgctgtaccaaaccttcggacggaaattgcacctgtattcccatcccatcatcctg ggctttcggaaaattcctatgggagtgggcctcagcccgtttctcctggctcagtttactagtgccattt gttcagtggttcgtagggctttcccccactgtttggctttcagttatatggatgatgtggtattgggggc caagtctgtacagcatcttgagtccctttttaccgctgttaccaattttcttttgtctttgggtatacatttaa accctaacaaaacaaagagatggggttactctctgaattttatgggttatgtcattggaagttatgggtc cttgccacaagaacacatcatacaaaaaatcaaagaatgttttagaaaacttcctattaacaggcctat tgattggaaagtatgtcaacgaattgtgggtcttttgggttttgctgccccatttacacaatgtggttatc ctgcgttaatgcccttgtatgcatgtattcaatctaagcaggctttcactttctcgccaacttacaaggcc tttctgtgtaaacaatacctgaacctttaccccgttgcccggcaacggccaggtctgtgccaagtgttt gctgacgcaacccccactggctggggcttggtcatgggccatcagcgcgtgcgtggaaccttttcg gctcctctgccgatccatactgcggaactcctagccgcttgttttgctcgcagcaggtctggagcaaa cattatcgggactgataactctgttgtcctctcccgcaaatatacatcgtatccatggctgctaggctgt gctgccaactggatcctgcgcgggacgtcctttgtttacgtcccgtcggcgctgaatcctgcggacg acccttctcggggtcgcttgggactctctcgtccccttctccgtctgccgttccgaccgaccacgggg cgcacctctctttacgcggactccccgtctgtgccttctcatctgccggaccgtgtgcacttcgcttca cctctgcacgtcgcatggagaccaccgtgaacgcccaccgaatgttgcccaaggtcttacataaga ggactcttggactctctgcaatgtcaacgaccgaccttgaggcatacttcaaagactgtttgtttaaag actgggaggagttgggggaggagattagattaaaggtctttgtactaggaggctgtaggcataaatt ggtctgcgcaccagcaccatgcaactttttcacctctgcctaatcatctcttgttcatgtcctactgttca agcctccaagctgtgccttgggtggctttggggcatggacatcgacccttataaagaatttggagcta ctgtggagttactctcgtttttgccttctgacttctttccttcagtacgagatcttctagataccgcctcag ctctgtatcgggaagccttagagtctcctgagcattgttcacctcaccatactgcactcaggcaagca attctttgctggggggaactaatgactctagctacctgggtgggtgttaatttggaagatccagcatct agagacctagtagtcagttatgtcaacactaatatgggcctaaagttcaggcaactcttgtggtttcac atttcttgtctcacttttggaagagaaaccgttatagagtatttggtgtctttcggagtgtggattcgcact cctccagcttatagaccaccaaatgcccctatcctatcaacacttccggaaactactgttgttagacga cgaggcaggtcccctagaagaagaactccctcgcctcgcagacgaaggtctcaatcgccgcgtcg cagaagatctcaatctcgggaacctcaatgttagtattccttggactcataaggtggggaactttactg gtctttattcttctactgtacctgtctttaatcctcattggaaaacaccatcttttcctaatatacatttacacc aagacattatcaaaaaatgtgaacagtttgtaggcccacttacagttaatgagaaaagaagattgcaa ttgattatgcctgctaggttttatccaaaggttaccaaatatttaccattggataagggtattaaaccttat tatccagaacatctagttaatcattacttccaaactagacactatttacacactctatggaaggcgggta tattatataagagagaaacaacacatagcgcctcattttgtgggtcaccatattcttgggaacaagatc tacagcatggggcagaatctttccaccagcaatcctctgggattctttcccgaccaccagttggatcc agccttcagagcaaacacagcaaatccagattgggacttcaatcccaacaaggacacctggccag acgccaacaaggtaggagctggagcattcgggctgggtttcaccccaccgcacggaggccttttg gggtggagecetcaggcteagggcatactacaaactttgccagcaaatccgcctcctgcctccacc aatcgccagacaggaaggcagcctaccccgctgtctccacctttgagaaacactcatcctcaggcc atgcagtgg
332
Table 5
SEQ ID NO: Description Sequence+
411 MCJ mRNA (GenBank Accession No. NM_01323 8.3) agtcactgccgcggcgccttgagtctccgggccgccttgccatggctgcccgtggtgtcatcgctc cagttggcgagagtttgcgctacgctgagtacttgcagccctcggccaaacggccagacgccgac gtcgaccagcagagactggtaagaagtttgatagctgtaggactgggtgttgcagctcttgcatttgc aggtcgctacgcatttcggatctggaaacctctagaacaagttatcacagaaactgcaaagaagattt caactcctagcttttcatcctactataaaggaggatttgaacagaaaatgagtaggcgagaagctggt cttattttaggtgtaagcccatctgctggcaaggctaagattagaacagctcataggagagtcatgatt ttgaatcacccagataaaggtggatctccttacgtagcagccaaaataaatgaagcaaaagacttgct agaaacaaccaccaaacattgatgcttaaggaccacactgaaggaaaaaaaaagaggggacttcg aaaaaaaaaaaagccctgcaaaatattctaaaacatggtcttcttaattttctatatggattgaccacag tcttatcttccaccattaagctgtataacaataaaatgttaatagtcttgctttttattatcttttaaagatctc cttaaattctataactgatcttttttcttattttgtttgtgacattcatacatttttaagatttttgttatgttctgaa ttcccccctacacacacacacacacàcacacacacacacacgtgcaaaaaatatgatcaagaatgc aattgggatttgtgagcaatgagtagacctcttattgtttatatttgtaccctcattgtcaatttttttttagg gaatttgggactctgcctatataaggtgttttaaatgtcttgagaacaagcactggctgatacctcttgg agatatgatctgaaatgtaatggaatttattaaatggtgtttagtaaagtaggggttaaggacttgttaaa gaaccccactatctctgagaccctatagccaaagcatgaggacttggagagctactaaaatgattca ggtttacaaaatgagccctgtgaggaaaggttgagagaagtctgaggagtttgtatttaattatagtctt ccagtactgtatattcattcattactcattctacaaatatttattgaccccttttgatgtgcaaggcactatc gtgcgtcccctgagagttgcaagtatgaagcagtcatggatcatgaaccaaaggaacttatatgtag aggaaggataaatcacaaatagtgaatactgttagatacagatgatatattttaaaagttcaaaggaag aaaagaatgtgttaaacactgcatgagaggaggaataagtggcatagagctaggctttagaaaaga aaaatattccgataccatatgattggtgaggtaagtgttattctgagatgagaattagcagaaatagat atatcaatcggagtgattagagtgcagggtttctggaaagcaaggtttggacagagtggtcatcaaa ggccagccctgtgacttacactgcattaaattaatttcttagaacatagtccctgatcattatcactttact attccaaaggtgagagaacagattcagatagagtgccagcattgtttcccagtattcctttacaaatctt gggttcattccaggtaaactgaactactgcattgtttctatcttaaaatactttttagatatcctagatgcat ctttcaacttctaacattctgtagtttaggagttctcaaccttggcattattgacatgttaggccaaataatt ttttttgtgggaggtctcttgtgcgttttagatgattagcaataatccctgacctgttatctactaaagact agtcgtttctcatcagttgtgacaacaaaaatggttccagatattgccaaatgccctttagaggacagt aatcgcccccagttgagaaccatttcagtaaaactttaattactattttttcttttggtttataaaataatgat cctgaattaaattgatggaaccttgaagtcgataaaatatatttcttgctttaaagtccccatacgtgtcct actaattttctcatgctttagtgttttcacttttctcctgttatccttgtacctaagaatgccatcccaatccc cagatgtccacctgcccaaagtctaggcatagctgaaggccaagctaaaatgtatccctctttttctgg tacatgcagcaaaagtaatatgaattatcagctttctgagagcaggcattgtatctgtcttgtttggtgtt acattggcacccaataaatatttgttgagtgaatgaataaattcccatagcactttattcttcacatggta cataactataggggctatagcttggtaccttgtgaagcaactcttggtgtaacataccttatttctcatac taaaatgcaagaacctttagagcaaggatcttgccattcatctttgtaacctctttactctggagcacttg catttagcaggcatcataaagttttacgtaccaagaaaatgttgctgttttctgaatactatgcatcaaaa aatgttaccactaatttttaaagctctgctaaggaatattggggcaccctcagatgcaccttttaattgat gtcatattttcctaatccatactttattcatgagaatttgagtcaccccagcattagcttggaatttccttatt tcccatttgctttgcaggtgccttggagtcagatctggttttgaatactatcttcctgttatgtgatcttgg
333
Table 5
SEQ ID NO: Description Sequence+
gcagttacttaattttctagtcaataacccgtatctataaaatagagaaaataatcctacacaccgggg cctgttgtggggcggggagaggggggagggatcgcatttggagatatactaatgtaaatgacaagt taattggtgcagcacaccaacatggctcatgtctacatatgtaacaaacctgcacgttgtgcacatgtg ccctagaacttaaagtataataaaaagaaattttaaaaaatcctgtcaaataaggttatagtagagaata aggatgtgtaaagcatttagtcacgtaaatgcttaaaaaaatgtaatttttacttctttcactgcctcattta attagttttatctttaataataccttggattcagggtaaagtttcagttatgtcccagtaatcatttattttacc ctcgaatctgcaatttggatagaacatggtggggacagctcgtctctattccttgcagcattaacagg ctggaggcaccacttctctggccagcaagttgggcctggttgttggctgagagcctcagttcctttct gcacaggttcctctttacataggcttctcaacagggctactagagcatcgtcaccatagcagctgtctt ataacagagagtggtcggtctgagagacaaaaaatggaagctgccaaattgttctgggtctggaaa ctgtcagggcatcacttgtgccatattcagttggcctaagaattacagagcctgcctcgattcaaagg gagaggatagagaggactgaaggaatcagtgctcatctttaatatgcagcaggacaggtttgggatt ttttttcccccttgagtctgtgaaggcattacttaagaacaaagtcaggcatgtataattgaactacagtt acttgaaatataagcccagaaagtttcagataataaatacaactatttttctgctgttacccttgtacctaa agatgccatcctaatccccagatctccacaactatacctacatagtagaaggttaaaatgtatccctctt tttctggtgcatccagcaaaagtaatatcatgaattatgagctctctgagagcaaggatcatatcagtct tgtttattgttgcagtgaacaagtacagttgcagatattcaggagtaattatctaaatggcagtaggctt ataaaactgaattttcaccagccacaccctccccccaactccttatctgtaaaaagcttatttgagtggt tacctgtcttcagtaaagattgcgcttgcatatttgctgtcattgcatattctgcttaattaagctctgttga tattgcagtttctgtgcatacttacatcttagatgcaatctgagggcctaggaaggccttttaaaaataa aacaattccgattgcagagaaagtgtaagtcaaggacagttaattcaaggggaacatagaaagctat ttagattttagttgatggtgccagtcttcagcgtaaagtcaaaagtggagggaagtttagtaaggaaa aaatgttgggcttggaatacattgtttagtcttcaaagcactttactttttatgaaatatattttagacattca gcaaatattgaatacttactatatcaggcagtaaagatataaattcattcttaaaatgtgcaacatgttca aactgaaaaaaatacattcttaaacaggaaactttttccttcatactttttaattaacaagacatataaga gttgcattaatgggcgtgcttatgattgatcacccagcagcatcattagaaataatatattttattcatgt gcagaaatcttttggttgtcctggggaaccttgaacacagaaaagagcttttattgataaggtaattga acacacttgacaattagcttaatatggtttaataccatttgtgggagaagatgaatcagccaggctcttt acgtcaagaatatgaagtttctcttgagtcaaccaacttaagatgagctacggagactgcagtgaaaa gttaaatatccaagtacaccagccaatttcacacagtggaaccatgctgtcctcgggcaccctgcac ctcgcccaacagtcatcaactagatggaggctcctggctgcaaggaggatttgatgggaatgagta aatgtgtcagcatagtccgtcccttctaatggaaaagcaacccaaagagcaaatcctattaatggctg gatcagtatcatctacttgtcaaaaacattccatgaattatgagtcaaaattttatttatggtggcattaca cacattaagagatgaggacttctgttagcataatttattagctggaaaagttgagaaggttctctggact catttttataggtggaacctaagtgatctggataattgcccaccagcaaaattgctgggcatggtgga caaagaaaatgttccttctaatgattttttatgagctgagtagctattgttcccagctgagtgctcttttcct ctttttattgttgctgagcaaaagaatttataaaaagctctttcttttgtattaaaaaccctgctcaattgaa atgcaagttcattaagtaatcttcatttctcttcctgccataataaccctttccctctctgttcgattcaaca gtatctagcagcactgctccaaattttaagtctgaacagactatattacatagatgtagagaaatactca atcttcagcattaagagggagcttaatttcacacgggtggaatatgatcactcaggctagatgttggc cataaatttcaaattagtatctcaacttagcaggggggatcaacagtggcaaacttcaattatgacagg
334
Table 5
SEQ ID NO: Description Sequence+
ataaaaatcacatagagatattggttcaatatggacatctaaactataatgctaaaagccaataattaga ataagttcattttaagaaaagcattaataatattagctaacgtttagtacctgtgccaaacattctacctat gttaccttgattttcatagccagcctaagaggtactattatgtatccccattttacaggttaagaaacagg ctcagaggagtttaggatcttttccaagattacatagccagtaagtggtggcactaggaaccaaattc agactctgaatcgcatgctgtttatattatattgcactcattctaaatatgtgggaatcagaatgaaggg gcttgtatgacttttggctcattttttgatgcatgtgacctgggattataaatgtgaaattaggtttacgaa aggatccagtgtcattgtgcatcatgggcaaggagtacctaatctctttaattcttccctggaagcttac gatgtccatccaagtgcacatagcaaaagttctgttgtaaagtttagcagagtgactttctttgactcag agtgatgacggaggaagctttgataagattttatctgaaatgttcatggacaagagctttcaaggaga acatccagagcaaggttctgaagacagctcatgaaggtgaagcagcagacctggcacaagaaatg aagagagagctcagtgtattaaagatgaaaacaagaaaaccgaatatattgaaaggagcagagag gcaatgaaaacaagacaactgaaatgaggtaacttgcagcaattgaaagggaatttcagtacttttat agaattcttaaaaattgtttcctgctgtttattttcaattttgaacagggttatttgtccatgccatacttttttt gccaaattccaaaattgtgtatagttctatagttgtctggtggagtcaatggaactttagttaccagtcta agaatgtgtctttgagattgtccagttaattctctatttccagtagctgtaataaatggtgaaaaggtttct gactcctggagaaagtttctaactccttatgactaatattcataacagacttgtgagttccttgaacatgg atacacctatatgcaagagtgtattccaaagctaactcagtgatctttccatttatctattcttggattagt ggtgcctttgctctttccttctgtaaatgtgaatagttaagagttgactgcagaagtgtttacactttggct tccatgcctctggaatgtttgtgctttggtggtgagatgtgagactatatttgtatagtctgcatctctcag gctgccccagaatgttgtacagtgcagtgctgaagaaagcagcaggtacacacagaaatgcagcc tttcctggttaaccctgcttggatctgagttacactttgtttcctgacttcttgggacttaggtaatcagttt gccttctactctatctcattttgtactcgcttacatactacattcttgtttgggctttcgtttcttcttgtaagc agagattttttaaaatccaatatgtgaaaatacggatgcactacaattaaataaataaaatgctgttgtgt ttgttttgctttaaaattgtaaaggataaacaataagatagttttatctatgtggttttcccgatgcagttaa aataaaacctaatctgctaaaattgaa
412 TAZ (GenBank Accession No. NM-00011 6.5) gctttccggcggttgcaccgggccggggtgccagcgcccgccttcccgtttcctcccgttccgcag cgcgcccacggcctgtgaccccggcgaccgctccccagtgacgagagagcggggccgggcgc tgctccggcctgacctgcgaagggacctcggtccagtcccctgttgcgccgcgcccccgtccgtcc gtgcgcgggccagtcaggggccagtgtctcgagcggtcgaggtcgcagacctagaggcgcccc acaggccggcccggggcgctgggagcgccggccgcgggccgggtggggatgcctctgcacgt gaagtggccgttccccgcggtgccgccgctcacctggaccctggccagcagcgtcgtcatgggct tggtgggcacctacagctgcttctggaccaagtacatgaaccacctgaccgtgcacaacagggag gtgctgtacgagctcatcgagaagcgaggcccggccacgcccctcatcaccgtgtccaatcacca gtcctgcatggacgaccctcatctctgggggatcctgaaactccgccacatctggaacctgaagttg atgcgttggacccctgcagctgcagacatctgcttcaccaaggagctacactcccacttcttcagctt gggcaagtgtgtgcctgtgtgccgaggagcagaatttttccaagcagagaatgaggggaaaggtg ttctagacacaggcaggcacatgccaggtgctggaaaaagaagagagaaaggagatggcgtcta ccagaaggggatggacttcattttggagaagctcaaccatggggactgggtgcatatcttcccagaa gggaaagtgaacatgagttccgaattcctgcgtttcaagtggggaatcgggcgcctgattgctgagt gtcatctcaaccccatcatcctgcccctgtggcatgtcggaatgaatgacgtccttcctaacagtccg ccctacttcccccgctttggacagaaaatcactgtgctgatcgggaagcccttcagtgccctgcctgt
335
Table 5
SEQ ID NO: Description Sequence’
actcgagcggctccgggcggagaacaagtcggctgtggagatgcggaaagccctgacggacttc attcaagaggaattccagcatctgaagactcaggcagagcagctccacaaccacctccagcctgg gagataggccttgcttgctgccttctggattcttggcccgcacagagctggggctgagggatggact gatgcttttagctcaaacgtggcttttagacagatttgttcatagaccctctcaagtgccctctccgagc tggtaggcattccagctcctccgtgcttcctcagttacacaaaggacctcagctgcttctcccacttgg ccaagcagggaggaagaagcttaggcagggctctctttccttcttgccttcagatgttctctcccagg ggctggcttcaggagggagcatagaaggcaggtgagcaaccagttggctaggggagcaggggg cccaccagagctgtggagaggggaccctaagactcctcggcctggctcctacccaccgcccttgc cgaaccaggagctgctcactacctcctcagggatggccgttggccacgtcttccttctgcctgagctt cccccccaccacaggccctttcctcaggcaaggtctggcctcaggtgggccgcaggcgggaaaa gcagcccttggccagaagtcaagcccagccacgtggagcctagagtgagggcctgaggtctggc tgcttgcccccatgctggcgccaacaacttctccatcctttctgcctctcaacatcacttgaatcctagg gcctgggttttcatgtttttgaaacagaaccataaagcatatgtgttggcttgttgtaaaa
413 ANGPTL3 (GenBank Accession No. NM_01449 5.4) agaagaaaacagttccacgttgcttgaaattgaaaatcaagataaaaatgttcacaattaagctccttc tttttattgttcctctagttatttcctccagaattgatcaagacaattcatcatttgattctctatctccagag ccaaaatcaagatttgctatgttagacgatgtaaaaattttagccaatggcctccttcagttgggacatg gtcttaaagactttgtccataagacgaagggccaaattaatgacatatttcaaaaactcaacatatttga tcagtctttttatgatctatcgctgcaaaccagtgaaatcaaagaagaagaaaaggaactgagaaga actacatataaactacaagtcaaaaatgaagaggtaaagaatatgtcacttgaactcaactcaaaactt gaaagcctcctagaagaaaaaattctacttcaacaaaaagtgaaatatttagaagagcaactaactaa cttaattcaaaatcaacctgaaactccagaacacccagaagtaacttcacttaaaacttttgtagaaaa acaagataatagcatcaaagaccttctccagaccgtggaagaccaatataaacaattaaaccaacag catagtcaaataaaagaaatagaaaatcagctcagaaggactagtattcaagaacccacagaaattt ctctatcttccaagccaagagcaccaagaactactccctttcttcagttgaatgaaataagaaatgtaa aacatgatggcattcctgctgaatgtaccaccatttataacagaggtgaacatacaagtggcatgtat gccatcagacccagcaactctcaagtttttcatgtctactgtgatgttatatcaggtagtccatggacatt aattcaacatcgaatagatggatcacaaaacttcaatgaaacgtgggagaactacaaatatggttttg ggaggcttgatggagaattttggttgggcctagagaagatatactccatagtgaagcaatctaattat gttttacgaattgagttggaagactggaaagacaacaaacattatattgaatattctttttacttgggaaa tcacgaaaccaactatacgctacatctagttgcgattactggcaatgtccccaatgcaatcccggaaa acaaagatttggtgttttctacttgggatcacaaagcaaaaggacacttcaactgtccagagggttatt caggaggctggtggtggcatgatgagtgtggagaaaacaacctaaatggtaaatataacaaaccaa gagcaaaatctaagccagagaggagaagaggattatcttggaagtctcaaaatggaaggttatactc tataaaatcaaccaaaatgttgatccatccaacagattcagaaagctttgaatgaactgaggcaaattt aaaaggcaataatttaaacattaacctcattccaagttaatgtggtctaataatctggtattaaatccttaa gagaaagcttgagaaatagattttttttatcttaaagtcactgtctatttaagattaaacatacaatcacat aaccttaaagaataccgtttacatttctcaatcaaaattcttataatactatttgttttaaattttgtgatgtg ggaatcaattttagatggtcacaatctagattataatcaataggtgaacttattaaataacttttctaaata aaaaatttagagacttttattttaaaaggcatcatatgagctaatatcacaactttcccagtttaaaaaact agtactcttgttaaaactctaaacttgactaaatacagaggactggtaattgtacagttcttaaatgttgt agtattaatttcaaaactaaaaatcgtcagcacagagtatgtgtaaaaatctgtaatacaaatttttaaac
336
Table 5
SEQ ID NO: Description Sequence4-
tgatgcttcattttgctacaaaataatttggagtaaatgtttgatatgatttatttatgaaacctaatgaagc agaattaaatactgtattaaaataagttcgctgtctttaaacaaatggagatgactactaagtcacattg actttaacatgaggtatcactataccttatttgttaaaatatatactgtatacattttatatattttaacactta atactatgaaaacaaataattgtaaaggaatcttgtcagattacagtaagaatgaacatatttgtggcat cgagttaaagtttatatttcccctaaatatgctgtgattctaatacattcgtgtaggttttcaagtagaaat aaacctcgtaacaagttactgaacgtttaaacagcctgacaagcatgtatatatgtttaaaattcaataa acaaagacccagtccctaaattatagaaatttaaattattcttgcatgtttatcgacatcacaacagatcc ctaaatccctaaatccctaaagattagatacaaattttttaccacagtatcacttgtcagaatttatttttaa atatgattttttaaaactgccagtaagaaattttaaattaaacccatttgttaaaggatatagtgcccaagt tatatggtgacctacctttgtcaatacttagcattatgtatttcaaattatccaatatacatgtcatatatattt ttatatgtcacatatataaaagatatgtatgatctatgtgaatcctaagtaaatattttgttccagaaaagt acaaaataataaaggtaaaaataatctataattttcaggaccacagactaagctgtcgaaattaacgct gatttttttagggccagaataccaaaatggctcctctcttcccccaaaattggacaatttcaaatgcaaa ataattcattatttaatatatgagttgcttcctctatttggtttcc
414 DGAT2 (GenBank Accession No. NM_00125 3891.1) tgccccgttgtgaggtgataaagtgttgcgctccgggacgccagcgccgcggctgccgcctctgct ggggtctaggctgtttctctcgcgccaccactggccgccggccgcagctccaggtgtcctagccgc ccagcctcgacgccgtcccgggacccctgtgctctgcgcgaagccctggccccgggggccggg gcatgggccaggggcgcggggtgaagcggcttcccgcggggccgtgactgggcgggcttcagc catgaagaccctcatagccgcctactccggggtcctgcgcggcgagcgtcaggccgaggctgac cggagccagcgctctcacggaggacctgcgctgtcgcgcgaggggtctgggagatggggagtg gcctgcagtgccatcctcatgtacatattctgcactgattgctggctcatcgctgtgctctacttcacttg gctggtgtttgactggaacacacccaagaaaggtggcaggaggtcacagtgggtccgaaactggg ctgtgtggcgctactttcgagactactttcccatccagctggtgaagacacacaacctgctgaccacc aggaactatatctttggataccacccccatggtatcatgggcctgggtgccttctgcaacttcagcac agaggccacagaagtgagcaagaagttcccaggcatacggccttacctggctacactggcaggca acttccgaatgcctgtgttgagggagtacctgatgtctggaggtatctgccctgtcagccgggacac catagactatttgctttcaaagaatgggagtggcaatgctatcatcatcgtggtcgggggtgcggctg agtctctgagctccatgcctggcaagaatgcagtcaccctgcggaaccgcaagggctttgtgaaact ggccctgcgtcatggagctgacctggttcccatctactcctttggagagaatgaagtgtacaagcag gtgatcttcgaggagggctcctggggccgatgggtccagaagaagttccagaaatacattggtttcg ccccatgcatcttccatggtcgaggcctcttctcctccgacacctgggggctggtgccctactccaag cccatcaccactgttgtgggagagcccatcaccatccccaagctggagcacccaacccagcaaga catcgacctgtaccacaccatgtacatggaggccctggtgaagctcttcgacaagcacaagaccaa gttcggcctcccggagactgaggtcctggaggtgaactgagccagccttcggggccaattccctg gaggaaccagctgcaaatcacttttttgctctgtaaatttggaagtgtcatgggtgtctgtgggttattta aaagaaattataacaattttgctaaaccattacaatgttaggtcttttttaagaaggaaaaagtcagtattt caagttctttcacttccagcttgccctgttctaggtggtggctaaatctgggcctaatctgggtggctca gctaacctctcttcttcccttcctgaagtgacaaaggaaactcagtcttcttggggaagaaggattgcc attagtgacttggaccagttagatgattcactttttgcccctagggatgagaggcgaaagccacttctc atacaagcccctttattgccactaccccacgctcgtctagtcctgaaactgcaggaccagtttctctgc caaggggaggagttggagagcacagttgccccgttgtgtgagggcagtagtaggcatctggaatg
337
Table 5
SEQ ID NO: Description Sequence+
ctccagtttgatctcccttctgccacccctacctcacccctagtcactcatatcggagcctggactggc ctccaggatgaggatgggggtggcaatgacaccctgcaggggaaaggactgccccccatgcacc attgcagggaggatgccgccaccatgagctaggtggagtaactggtttttcttgggtggctgatgac atggatgcagcacagactcagccttggcctggagcacatgcttactggtggcctcagtttaccttccc cagatcctagattctggatgtgaggaagagatccctcttcagaaggggcctggccttctgagcagca gattagttccaaagcaggtggcccccgaacccaagcctcacttttctgtgccttcctgagggggttg ggccggggaggaaacccaaccctctcctgtgtgttctgttatctcttgatgagatcattgcaccatgtc agacttttgtatatgccttgaaaataaatgaaagtgagaatcctctaaaaaaaaaaaa
596 HBV Genbank Accession No. KC315400. 1 ctccaccactttccaccaaactcttcaagatcccagagtcagggccctgtactttcctgctggtggctc aagttccggaacagtaaaccctgctccgactactgcctctcccatatcgtcaatcttctcgaggactg gggaccctgtaccgaatatggagagcaccacatcaggattcctaggacccctgctcgtgttacagg cggggtttttcttgttgacaagaatcctcacaataccacagagtctagactcgtggtggacttctctca attttctagggggagcacccacgtgtcctggccaaaatttgcagtccccaacctccaatcactcacca acctcttgtcctccaatttgtcctggttatcgctggatgtgtctgcggcgttttatcatcttcctcttcatcc tgctgctatgcctcatcttcttgttggttcttctggactaccaaggtatgttgcccgtttgtcctctacttcc aggaacatcaactaccagcaccggaccatgcaaaacctgcacaactactgctcaagggacctctat gtttccctcatgttgctgtacaaaacctacggacggaaactgcacctgtattcccatcccatcatcttgg gctttcgcaaaatacctatgggagtgggcctcagtccgtttctcttggctcagtttactagtgccatttgt tcagtggttcgtagggctttcccccactgtctggctttcagttatatggatgatgtggttttgggggcca agtctgtacaacatcttgagtccctttataccgctgttaccaattttcttttatctttgggtatacatttaaac cctcacaaaacaaaaagatggggatattcccttaacttcatgggatatgtaattgggagttggggcac tttgcctcaggaacatattgtacaaaaaatcaagcaatgttttaggaaacttcctgtaaacaggcctatt gattggaaagtatgtcaacraattgtgggtcttttggggtttgccgcccctttcacgcaatgtggatatc ctgctttaatgcctttatatgcatgtatacaagctaagcaggcttttactttctcgccaacttacaaggcct ttctgtgtaaacaatatctgaacctttaccccgttgctcggcaacggtcaggtctttgccaagtgtttgct gacgcaacccccactggttggggcttggccataggccatcagcgcatgcgtggaacctttgtggct cctctgccgatccatactgcggaactcctagcagcttgttttgctcgcagccggtctggagcaaaact tatcggcaccgacaactctgttgtcctctctcggaaatacacctcctttccatggctgctaggatgtgct gccaactggatcctgcgcgggacgtcctttgtctacgtcccgtcggcgctgaatcccgcggacgac ccatctcggggccgtttgggactctaccgtccccttctgcgtctgccgttccgcccgaccacggggc gcacctctctttacgcggtctccccgtctgtgccttctcatctgccggaccgtgtgcacttcgcttcacc tctgcacgtcgcatggagaccaccgtgaacgcccacgggaacctgcccaaggtcttgcataagag gactcttggactttcagcaatgtcaacgaccgaccttgaggcatacttcaaagactgtgtgtttactga gtgggaggagttgggggaggaggttaggttaaaggtctttgtactaggaggctgtaggcataaattg gtgtgttcaccagcaccatgcaactttttcacctctgcctaatcatctcatgttcatgtcctactgttcaag cctccaagctgtgccttgggtggctttggggcatggacattgacccgtataaagaatttggagcttct gtggagttactctcttttttgccttctgacttctttccttctattcgagatctcctcgacaccgcctctgctct gtatcgggaggccttagagtctccggaacattgttcacctcaccatacggcactcaggcaagcaatt ctgtgttggggtgagttaatgaatctagccacctgggtgggaagtaatttggaagatccagcatcca gggaattagtagtcagctatgtcaacgttaatatgggcctaaaaatcagacaactattgtggtttcaca tttcctgtcttacttttgggagagaaactgttcttgaatatttggtgtcttttggagtgtggattcgcactcc
338
Table 5
SEQ ID NO: Description Sequence+
tcctgcatatagaccacaaaatgcccctatcttatcaacacttccggaaactactgttgttagacgaag aggcaggtcccctagaagaagaactccctcgcctcgcagacgaaggtctcaatcgccgcgtcgca gaagatctcaatctcgggaatctcaatgttagtattccttggacacataaggtgggaaactttacggg gctttattcttctacggtaccttgctttaatcctaaatggcaaactccttcttttcctgacattcatttgcag gaggacattgttgatagatgtaagcaatttgtggggccccttacagtaaatgaaaacaggagacttaa attaattatgcctgctaggttttatcccaatgttactaaatatttgcccttagataaagggatcaaaccgta ttatccagagtatgtagttaatcattacttccagacgcgacattatttacacactctttggaaggcgggg atcttatataaaagagagtccacacgtagcgcctcattttgcgggtcaccatattcttgggaacaagat ctacagcatgggaggttggtcttccaaacctcgaaaaggcatggggacaaatctttctgtccccaatc ccctgggattcttccccgatcatcagttggaccctgcattcaaagccaactcagaaaatccagattgg gacctcaacccacacaaggacaactggccggacgccaacaaggtgggagtgggagcattcggg ccagggttcacccctcctcatgggggactgttggggtggagccctcaggctcagggcatattcaca acagtgccagcagctcctcctcctgcctccaccaatcggcagtcaggaaggcagcctactcccttct ctccacctctaagagacactcatcctcaggccatgcagtggaa
534 ASO 1 GalNAc4-ps-GalNAc4-ps-GalNAc4-po-mA-po- lnGpslnApslnTpslnApslnApsApsAps(5OH)CpsGps(5m)Cps(5m)Cps Gps(5m)CpslnApslnGpslnApscp(5m)C
535 ASO 2 mA-po- lnGpslnApslnTpslnApslnApsApsAps(5OH)CpsGps(5m)Cps(5m)Cps Gps(5m)CpslnApslnGpslnApscp(5m)C
0^ B Voy cr—o +ln = Locked nucleic acid (LNA) = ; InA = Locked nucleic acid (LNA) A; ln(5m)C = ln(5m)C =Locked nucleic acid (LNA)-5 methyl C; lnG= Locked nucleic acid (LNA) G; lnT= Locked nucleic acid (LNA) T; (5m)C=5 methyl C; cp = sep = cyclopropyl; cpC = scpC NH2 ho. /L VI JWV . , , | N 0 CX poU 0 = cyclopropyl C; scp(5m)C = cyclopropyl-5 methyl C; (50H)C ; po = phosphodiester linkage; ps = phosphorothioate linkage
Table 6. siNA Activity
ds-siNA ID Sense Strand SEQ ID NO. Antisense Strand SEQ ID NO. HepG2.2.15 in vitro EC50* HepG2.2.15 in vitro CC50 (nM)
ds-siNA-001 307 363 A >40
ds-siNA-002 308 364 A >40
339
Table 6. siNA Activity
ds-siNA ID Sense Strand SEQ ID NO. Antisense Strand SEQ ID NO. HepG2.2.15 in vitro EC50* HepG2.2.15 in vitro CC50 (nM)
ds-siNA-003 309 365 B >40
ds-siNA-004 310 366 B >40
ds-siNA-005 311 367 B >40
ds-siNA-006 312 368 C >40
ds-siNA-007 313 369 Ά >40
ds-siNA-008 314 370 A >40
ds-siNA-009 315 371 B >40
ds-siNA-010 316 372 A >40
ds-siNA-011 317 373 B ; >40
ds-siNA-012 318 374 A >40
ds-siNA-013 319 375 A >40
ds-siNA-014 320 376 B >40
ds-siNA-015 321 377 A >40
ds-siNA-016 322 377 C >40
ds-siNA-017 323 377 A >40
ds-siNA-018 324 378 A >40
ds-siNA-019 325 378 A >40
ds-siNA-020 326 379 A >40
ds-siNA-021 327 379 B >40
ds-siNA-022 328 380 A >40
ds-siNA-023 329 380 B >40
ds-siNA-024 330 381 A >40
ds-siNA-025 331 382 A >40
ds-siNA-026 332 383 C >40
ds-siNA-027 333 384 A >40
ds-siNA-028 334 385 B >40
ds-siNA-029 335 386 A >40
ds-siNA-030 336 387 C >40
ds-siNA-031 337 388 A >40
ds-siNA-032 338 388 C >40
ds-siNA-033 339 389 B >40
ds-siNA-034 340 389 C >40
ds-siNA-035 341 390 A >40
ds-siNA-036 342 391 A >40
ds-siNA-037 343 392 B >40
ds-siNA-038 344 393 A >40
ds-siNA-039 345 394 A >40
ds-siNA-040 346 395 A >40
ds-siNA-041 347 396 C >40
ds-siNA-042 348 397 A >40
340
Table 6. siNA Activity
ds-siNA ID Sense Strand SEQ ID NO. Antisense Strand SEQ ID NO. HepG2.2.15 in vitro EC50* HepG2.2.15 in vitro CC50 (nM)
ds-siNA-043 349 398 B >40
ds-siNA-044 350 399 A >40
ds-siNA-045 351 400 A >40
ds-siNA-046 352 401 A >40
ds-siNA-047 353 402 A >40
ds-siNA-048 354 403 A >40
ds-siNA-049 355 404 B >40
ds-siNA-050 356 405 A >40
ds-siNA-051 357 406 A >40
ds-siNA-052 358 406 A >40
ds-siNA-053 359 407 A >40
ds-siNA-054 360 407 A >40
ds-siNA-055 361 408 A >40
ds-siNA-056 362 409 A >40
ds-siNA-0164 423 482
*A = EC50 < 0.5 nM; B = 0.5 nM [ < EC50 < 1; C = EC50 > 1 nm
Table 10. siNA Activity
ds-siNA ID Sense Strand SEQ ID NO 3' Ligand Monomer+ Antisense Strand SEQ ID NO HepG2.2.15 EC50* HepG2.2.15 CC50 (nM) Max HBsAg Knock Down (Logio) **
ds-siNA-057 415 p-(PS)2-GalNac4 445 ND ND X
ds-siNA-058 415 p-(PS)2-GalNac4 446 ND ND X
ds-siNA-059 415 p-(PS)2-GalNac4 447 ND ND Y
ds-siNA-060 416 p-(PS)2-GalNac4 448 ND ND Y
ds-siNA-061 416 p-(PS)2-GalNac4 449 ND ND Y
ds-siNA-062 416 p-(PS)2-GalNac4 450 ND ND Y
ds-siNA-063 416 p-(PS)2-GalNac4 451 ND ND X
ds-siNA-064 417 5’-GalNAc4(PS)2-p-TEG-p 452 ND ND Y
ds-siNA-065 417 5’-GalNAc4(PS)2-p-HEG-p 452 ND ND Y
ds-siNA-066 417 5’-GalNAc4(PS)2-p-(HEGP)2 452 ND ND Y
341
Table 10. siNA Activity
ds-siNA ID Sense Strand SEQ ID NO 3' Ligand Monomer+ Antisense Strand SEQ ID NO HepG2.2.15 EC50* HepG2.2.15 CC50 (nM) Max HBsAg Knock Down (Logio) **
ds-siNA-067 417 5'-GalNAc4(PS)2-p-(HEGP)2 452 ND ND Z
ds-siNA-068 418 p-(PS)2-GalNac4 453 ND ND Y
ds-siNA-069 418 p-(PS)2-GalNac4 454 ND ND Y
ds-siNA-070 419 p-(PS)2-GalNac4 455 ND ND Y
ds-siNA-071 419 p-(PS)2-GalNac4 456 ND ND Y
ds-siNA-072 420 p-(PS)2-GalNac4 457 ND ND X
ds-siNA-073 421 p-(PS)2-GalNac4 458 ND ND X
ds-siNA-074 422 p-(PS)2-GalNac4 459 ND ND Y
ds-siNA-075 423 p-(PS)2-GalNac4 460 ND ND Y
ds-siNA-076 423 p-(PS)2-GalNac4 461 ND ND Y
ds-siNA-077 423 5’-GalNAc4(PS)2-p-TEG-p 458 ND ND Y
ds-siNA-078 423 5’-GalNAc4(PS)2-p-HEG-p 458 ND ND X
ds-siNA-079 423 5’-GalNAc4(PS)2-p-(HEGP)2 458 ND ND Y
ds-siNA-080 423 p-(PS)2-GalNac4 462 ND ND X
ds-siNA-081 423 p-(PS)2-GalNac4 463 ND ND X
ds-siNA-082 423 p-(PS)2-GalNac4 447 ND ND X
ds-siNA-083 423 5'-GalNAc4(PS)2-p-(HEGP)2 458 ND ND Z
ds-siNA-084 423 p-(PS)2-GalNac4 457 ND ND X
ds-siNA-085 423 p-(PS)2-GalNac4 464 ND ND X
ds-siNA-086 423 p-(PS)2-GalNac4 465 ND ND X
ds-siNA-087 423 p-(PS)2-GalNac4 466 ND ND Y
ds-siNA-088 423 p-(PS)2-GalNac4 467 ND ND Z
ds-siNA-089 423 p-(PS)2-GalNac4 468 ND ND Z
ds-siNA-090 423 p-(PS)2-GalNac4 469 B >1000 X
ds-siNA-091 423 p-(PS)2-GalNac4 470 C >1000 ND
ds-siNA-092 423 p-(PS)2-GalNac4 471 B >1000 ND
ds-siNA-093 423 p-(PS)2-GalNac4 472 B >1000 ND
ds-siNA-094 423 p-(PS)2-GalNac4 473 B >1000 ND
342
Table 10. siNA Activity
ds-siNA ID Sense Strand SEQ ID NO 3' Ligand Monomer+ Antisense Strand SEQ ID NO HepG2.2.15 EC50* HepG2.2.15 CC50 (nM) Max HBsAg Knock Down (Logio) **
ds-siNA-095 423 p-(PS)2-GalNac4 474 C >1000 ND
ds-siNA-096 423 p-(PS)2-GalNac4 475 B >1000 ND
ds-siNA-097 423 p-(PS)2-GalNac4 476 B >1000 ND
ds-siNA-098 423 p-(PS)2-GalNac4 477 A >1000 ND
ds-siNA-099 423 p-(PS)2-GalNac4 478 B >1000 ND
ds-siNA0100 423 p-(PS)2-GalNac4 479 B >1000 ND
ds-siNA0101 423 p-(PS)2-GalNac4 480 B >1000 ND
ds-siNA0102 423 p-(PS)2-GalNac4 481 A >1000 ND
ds-siNA0103 423 p-(PS)2-GalNac4 482 ND ND ND
ds-siNA0104 423 p-(PS)2-GalNac4 483 ND ND ND
ds-siNA0105 423 p-(PS)2-GalNac4 458 ND ND Z
ds-siNA0106 423 p-(PS)2-GalNac4 458 ND ND Y
ds-siNA0107 424 p-(PS)2-GalNac4 457 ND ND X
ds-siNA0108 424 p-(PS)2-GalNac4 484 ND ND X
ds-siNA0109 424 p-(PS)2-GalNac4 485 ND ND X
ds-siNA0110 425 p-(PS)2-GalNac4 486 ND ND ND
ds-siNA0111 425 p-(PS)2-GalNac4 487 ND ND ND
ds-siNA0112 425 p-(PS)2-GalNac4 488 ND ND ND
ds-siNA0113 426 p-(PS)2-GalNac4 489 ND ND ND
ds-siNA0114 427 p-(PS)2-GalNac4 490 ND ND X
ds-siNA0115 428 p-(PS)2-GalNac4 491 ND ND Y
343
Table 10. siNA Activity
ds-siNA ID Sense Strand SEQ ID NO 3' Ligand Monomer+ Antisense Strand SEQID NO HepG2.2.15 EC50* HepG2.2.15 CC50 (nM) Max HBsAg Knock Down (Logio) **
ds-siNA0116 429 p-(PS)2-GalNac4 492 ND ND ND
ds-siNA0117 429 p-(PS)2-GalNac4 493 ND ND ND
ds-siNA0118 429 p-(PS)2-GalNac4 494 ND ND ND
ds-siNA0119 430 p-(PS)2-GalNac4 495 ND ND X
ds-siNA0120 430 p-(PS)2-GalNac4 496 ND ND ND
ds-siNA0121 431 p-(PS)2-GalNac4 497 ND ND Y
ds-siNA0122 432 p-(PS)2-GalNac4 498 ND ND ND
ds-siNA0123 432 p-(PS)2-GalNac4 500 ND ND ND
ds-siNA0124 433 p-(PS)2-GalNac4 501 ND ND ND
ds-siNA0125 434 p-(PS)2-GalNac4 502 ND ND Y
ds-siNA0126 435 p-(PS)2-GalNac4 502 ND ND Y
ds-siNA0127 435 p-(PS)2-GalNac4 503 ND ND X
ds-siNA0128 435 p-(PS)2-GalNac4 501 ND ND X
ds-siNA0129 435 p-(PS)2-GalNac4 504 ND ND Y
ds-siNA0130 435 p-(PS)2-GalNac4 505 ND ND Z
ds-siNA0131 435 p-(PS)2-GalNac4 506 ND ND Y
ds-siNA0132 435 p-(PS)2-GalNac4 507 ND ND Z
ds-siNA0133 435 p-(PS)2-GalNac4 508 ND ND Z
344
Table 10. siNA Activity
ds-siNA ID Sense Strand SEQ ID NO 3' Ligand Monomer+ Antisense Strand SEQ ID NO HepG2.2.15 EC50* HepG2.2.15 CC50 (nM) Max HBsAg Knock Down (Logio) **
ds-siNA0134 435 p-(PS)2-GalNac4 509 ND ND Z
ds-siNA0135 435 p-(PS)2-GalNac4 510 ND ND Y
ds-siNA0136 435 p-(PS)2-GalNac4 511 B >1000 ND
ds-siNA0137 435 p-(PS)2-GalNac4 512 B >1000 ND
ds-siNA0138 435 p-(PS)2-GalNac4 513 A >1000 ND
ds-siNA0139 435 p-(PS)2-GalNac4 514 B >1000 ND
ds-siNA0140 435 p-(PS)2-GalNac4 515 C >1000 ND
ds-siNA0141 435 p-(PS)2-GalNac4 516 A >1000 ND
ds-siNA0142 435 p-(PS)2-GalNac4 517 C >1000 ND
ds-siNA0143 435 p-(PS)2-GalNac4 518 C >1000 ND
ds-siNA0144 435 p-(PS)2-GalNac4 519 ND ND ND
ds-siNA0145 436 p-(PS)2-GalNac4 520 ND ND ND
ds-siNA0146 437 p-(PS)2-GalNac4 502 ND ND Y
ds-siNA0147 438 p-(PS)2-GalNac4 501 ND ND X
ds-siNA0148 438 p-(PS)2-GalNac4 521 ND ND X
ds-siNA0149 438 p-(PS)2-GalNac4 522 ND ND X
ds-siNA0150 439 p-(PS)2-GalNac4 523 ND ND X
ds-siNA0151 440 p-(PS)2-GalNac4 524 ND ND Y
345
Table 10. siNA Activity
ds-siNA ID Sense Strand SEQ ID NO 3' Ligand Monomer+ Antisense Strand SEQ ID NO HepG2.2.15 EC50* HepG2.2.15 CC50 (nM) Max HBsAg Knock Down (Logio) **
ds-siNA0152 441 p-(PS)2-GalNac4 525 ND ND Y
ds-siNA0153 441 p-(PS)2-GalNac4 526 ND ND X
ds-siNA0154 441 p-(PS)2-GalNac4 527 ND ND ND
ds-siNA0155 442 p-(PS)2-GalNac4 525 ND ND X
ds-siNA0156 442 p-(PS)2-GalNac4 528 ND ND Y
ds-siNA0157 442 p-(PS)2-GalNac4 529 ND ND ND
ds-siNA0158 443 p-(PS)2-GalNac4 458 ND ND Y
ds-siNA0159 444 p-(PS)2-GalNac4 502 ND ND Y
ds-siNA0160 423 p-(PS)2-GalNac4 458 ND ND ND
ds-siNA0161 533 p-(PS)2-GalNac4 489 ND ND ND
ds-siNA0162 534 p-(PS)2-GalNac4 491 ND ND ND
ds-siNA0163 432 p-(PS)2-GalNac4 496 ND ND ND
ds-siNA0165 435 p-(PS)2-GalNac4 502 ND ND ND
ds-siNA0166 442 p-(PS)2-GalNac4 530 ND ND ND
ds-siNA0167 427 p-(PS)2-GalNAc4 491 ND ND ND
ds-siNA0168 439 p-(PS)2-GalNac4 531 ND ND ND
ds-siNA0169 423 p-(PS)2-GalNac4 532 ND ND ND
ds-siNA0170 441 p-(PS)2-GalNAc4 530 ND ND ND
346
Table 10. siNA Activity
ds-siNA ID Sense Strand SEQ ID NO 3' Ligand Monomer+ Antisense Strand SEQID NO HepG2.2.15 EC50* HepG2.2.15 CC50 (nM) Max HBsAg Knock Down (Logio) **
ds-siNA0171 442 p-(PS)2-GalNAc4 533 ND ND ND
ds-siNA0172 424 p-(PS)2-GalNAc4 536 A >1 ND
ds-siNA0173 438 None 537
ds-siNA0174 438 None 538
ds-siNA0175 438 None 501
ds-siNA0176 438 p-(PS)2-GalNAc4 537
ds-siNA0177 438 p-(PS)2-GalNAc4 538
ds-siNA0178 438 p-(PS)2-GalNAc4 539
+Ligand monomers are attached to the 3 ’ end of the sense strand, unless the ligand monomer is annotated with 5’, in which the ligand monomer is attached to the 5’ end of the sense strand. Linkers are represented as p-(PS)2, (PS)2-p-TEG-p, (PS)2-p-HEG-p, or (PS)2-p-(HEG-p)2. *For EC50, A = EC50 < 5 nM; B = 5 nM < EC50 < 10; C = EC50 > 10. **For Max HBsAg knock down, X > 1 logio réduction in HBsAg, Y is 0.5 - 1 logio réduction in HBsAg, and Z is < 0.5 logio réduction in HBsAg.

Claims (34)

1. A short interfering nucleic acid (siNA) molécule comprising:
(a) a sense strand comprising a first nucléotide sequence that is at least about 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% identical to an RNA corresponding to a target gene, wherein the first nucléotide sequence:
(i) is 15 to 30 nucléotides in length; and (ii) comprises 15 or more modified nucléotides independently selected from a 2’-O-methyl nucléotide and a 2’-fluoro nucléotide, wherein at least one modified nucléotide is a 2’-O-methyl nucléotide and the nucléotide at position 3, 5, 7, 8, 9, 10,11, 12, 14, 17, and/or 19 from the 5’ end of the first nucléotide sequence is a 2’-fluoro nucléotide; and an antisense strand comprising a second nucléotide sequence that is at least about 60%, 65%, 70%, 75%, 80%, 85%, 90% 95%, or 100% complementary to the RNA corresponding to the target gene, wherein the second nucléotide sequence:
(iii) is 15 to 30 nucléotides in length; and (iv) comprises 15 or more modified nucléotides independently selected from a 2’-O-methyl nucléotide and a 2’-fluoro nucléotide, wherein at least one modified nucléotide is a 2’-O-methyl nucléotide and the nucléotide at position 2, 5, 6, 8, 10, 14, 16, 17, and/or 18 from the 5’ end of the second nucléotide sequence is a 2’-fluoro nucléotide or wherein at least one modified nucléotide is a 2’-(9-methyl nucléotide and at least one modified nucléotide is a 2’-fluoro nucléotide; or (b) a sense strand comprising a first nucléotide sequence that is at least about 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% identical to an RNA corresponding to a target gene, wherein the first nucléotide sequence:
(i) is 15 to 30 nucléotides in length; and (ii) comprises 15 or more modified nucléotides independently selected from a 2’-(9-methyl nucléotide and a 2’-fluoro nucléotide, wherein at least one modified nucléotide is a 2’-û-methyl nucléotide and at least one modified nucléotide is a 2’-fluoro nucléotide; andan antisense strand comprising a second nucléotide sequence that is at least about 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% complementary to the RNA corresponding to the target gene, wherein the second nucléotide sequence:
348 (iii) is 15 to 30 nucléotides in length; and (iv) comprises 15 or more modified nucléotides independently selected from a 2’-(9-methyl nucléotide and a 2’-fluoro nucléotide, wherein at least one modified nucléotide is a 2’-O-methyl nucléotide and the nucléotide at position 2, 5, 6, 8, 10, 14, 16, 17, and/or 18 from the 5’ end of the second nucléotide sequence is a 2’-fluoro nucléotide.
2. The siNA of claim 1, wherein:
(i) at least 2, 3, 4, 5, or 6 modified nucléotides of the first nucléotide sequence are 2’-fluoro nucléotides;
(ii) no more than 10, 9, 8, 7, 6, 5, 4, 3, or 2 modified nucléotides of the first nucléotide sequence are 2’-fluoro nucléotides;
(iii) at least 10, 11, 12, 13, 14, 15, 16, 17,18, 19, 20, 21, or 22 modified nucléotides of the first nucléotide sequence are 2’-O-methyl nucléotides;
(iv) no more than 25, 24, 23, 22, 21, 20,19, 18, 17, 16, 15, 14, 13, 12, 11, 10, 9, 8, 7, 6, 5, 4, 3, or 2 modified nucléotides of the first nucléotide sequence are 2’O-methyl nucléotides;
(v) at least 2, 3, 4, 5, or 6 modified nucléotides of the second nucléotide sequence are 2’-fluoro nucléotides;
(vi) less than or equal to 10, 9, 8, 7, 6, 5, 4, 3, or 2 modified nucléotides of the second nucléotide sequence are 2’-fluoro nucléotides;
(vii) at least 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, or 22 modified nucléotides of the second nucléotide sequence are 2’-O-methyl nucléotides; and/or (viii) less than or equal to 25, 24, 23, 22, 21, 20, 19, 18, 17, 16, 15, 14, 13,12, 11, 10, 9, 8, 7, 6, 5, 4, 3, or 2 modified nucléotides of the second nucléotide sequence are 2’-(9-methyl nucléotides.
3. The siNA according to claim 1 or 2, wherein at least 1, 2, 3, 4, 5, 6, or 7 nucléotides at position 3, 5, 7, 8, 9, 10, 11, 12, and/or 17 from the 5’ end of the first nucléotide sequence is a 2’-fluoro nucléotide, and/or wherein at least 1, 2, 3, 4, 5, 6, 7, 8, or 9 nucléotides at position 2, 5, 6, 8, 10, 14, 16, 17, and/or 18 from the 5’ end of the second nucléotide sequence is a 2’-fluoro nucléotide.
4. The siNA according to any preceding claim, wherein the sense strand, the antisense strand, or both further comprises at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15 or more phosphorothioate intemucleoside linkages.
349
5. The siNA of claim 4, wherein:
(i) at least one phosphorothioate intemucleoside linkage is between the nucléotides at positions 1 and 2 from the 5’ end of the first nucléotide sequence;
(ii) at least one phosphorothioate intemucleoside linkage is between the nucléotides at positions 2 and 3 from the 5’ end of the first nucléotide sequence;
(iii) at least one phosphorothioate intemucleoside linkage is between the nucléotides at positions 1 and 2 from the 5’ end of the second nucléotide sequence;
(iv) at least one phosphorothioate intemucleoside linkage is between the nucléotides at positions 2 and 3 from the 5’ end of the second nucléotide sequence;
(v) at least one phosphorothioate intemucleoside linkage is between the nucléotides at positions 1 and 2 from the 3’ end of the second nucléotide sequence; and/or (vi) at least one phosphorothioate intemucleoside linkage is between the nucléotides at positions 2 and 3 from the 3’ end of the second nucléotide sequence.
6. The siNA according to any preceding claim, wherein:
(i) the first nucléotide from the 5’ end of the first nucléotide sequence comprises a 5’ stabilizing end cap;
(ii) the first nucléotide from the 5’ end of the second nucléotide sequence comprises a 5’ stabilizing end cap;
(iii) the first nucléotide from the 5’ end of the first nucléotide sequence comprises a phosphorylation blocker; and/or (iv) the first nucléotide from the 5’ end of the second nucléotide sequence comprises a phosphorylation blocker.
7. The siNA according to any preceding claim, wherein the first nucléotide sequence or second nucléotide sequence comprises at least one modified nucléotide selected from i (LNA), i-* (ScpBNA or “cp”); 0 (AmNA), where R is
350
H or alkyl (or AmNA(N-Me)) when R is alkyl);
h2n (GuNA); and
GuNA(N-R), R = Me, Et, iPr, tBu , wherein B is a nucleobase.
8. The siNA according to any preceding claim, wherein the siNA further comprises a galactosamine.
5
9. The. siNA of claim 8, wherein the galactosamine is N-acetylgalactosamine (GalNAc) of
Formula (VII):
R = OH or SH wherein each n is independently 1 or 2.
10. The siNA of claim 8, wherein the galactosamine is N-acetylgalactosamine (GalNAc) of
Formula (VI):
wherein m is 1, 2, 3, 4, or 5;
each n is independently 1 or 2;
p is 0 or 1 ;
each R is independently H;
351 each Y is independently selected from -O-P(=O)(SH)-, -O-P(=O)(O)-, -O-P(=O)(OH)-, and O-P(S)S-;
Z is H or a second protecting group;
either L is a linker or L and Y in combination are a linker; and
A is H, OH, a third protecting group, an activated group, or an oligonucleotide.
11. The siNA according to any one of daims 1-10, wherein the antisense strand, sense strand, first nucléotide sequence, and/or second nucléotide sequence comprises at least one thermally
destabilizing nucléotide selected from:
, and
12. The siNA according to any preceding claim, wherein;
(i) the target gene is a viral gene;
(ii) the target gene is a gene is from a DNA virus.
(iii) the target gene is a gene from a double-stranded DNA (dsDNA) virus;.
(iv) the target gene is a gene from a hepadnavirus;
(v) the target gene is a gene from a hepatitis B virus (HBV);
(vi) the target gene is a gene from a HBV of any one of génotypes A-J; or (vii) the target gene is selected from the S gene or X gene of a HBV.
13. Use of the siNA according to any one of daims 1-12 in the manufacture of a médicament for treating a disease.
14. The use of claim 13, wherein:
(i) the disease is a viral disease;
(ii) the disease is caused by a DNA virus;
(iii) the disease is caused by a double stranded DNA (dsDNA virus);
(iv) the disease is caused by a hepadnavirus;
(v) the disease is caused by a hepatitis B virus (HBV); or
352 (vi) the disease is caused by a HBV of any one of HBV génotypes A-J.
15. The use of claim 13, wherein:
(i) the disease is a liver disease;
(ii) the disease is a nonalcoholic fatty liver disease (NAFLD) or hepatocellular carcinoma (HCC); or (iii) the disease is nonalcoholic steatohepatitis (NASH).
16. A double stranded short interfering nucleic acid (siNA) comprising:
(a) a sense strand comprising 19-21 nucléotides in a nucleic acid sequence that is at least 80% identical to SEQ ID NO: 40, wherein 15 or more of the nucléotides are modified nucléotides independently selected from a 2’-(9-methyl nucléotide and a 2’-fluoro nucléotide, and wherein at least 1 lof the modified nucléotides are a 2’-O-methyl nucléotide and at least 4 of the modified nucléotides are a 2’-fluoro nucléotide; and (b) an antisense strand comprising 19-21 nucléotides in a nucleic acid sequence that is at least 80% complementary to SEQ ID NO: 40, wherein 15 or more of the nucléotides are modified nucléotides independently selected from a 2’-(9-methyl nucléotide and a 2’fluoro nucléotide, and wherein at least 11 of the modified nucléotides are a 2’-O-methyl nucléotide and 4 to 6 of the modified nucléotides are a 2’-fluoro nucléotide.
17. The double stranded siNA of claim 16, wherein:
(a) the nucleotide(s) at position 3, 5, 7, 8, 9, 10, 11, 12, 14,17, or 19 from the 5’ end of the sense strand is a 2’-fluoro nucléotide; and (b) the nucleotide(s) at position 2, 5, 6, 8, 10, 14, 16, 17, or 18 from the 5’ end of the antisense strand is a 2’-fluoro nucléotide.
18. The double stranded siNA of claim 16 or 17, wherein the sense strand comprises SEQ ID NO: 438 or SEQ ID NO: 435, and the antisense strand comprises any one of SEQ ID NOs: 501-519, SEQ ID NO: 537, SEQ ID NO: 538, or SEQ ID NO: 539.
19. A double stranded short interfering nucleic acid (siNA) comprising:
(a) a sense strand comprising 19 nucléotides, wherein 2’-fluoro nucléotides are at positions 5 and 7-9 from the 5’ end of the sense strand, and wherein 2’-O-methyl nucléotides are at positions 1-4, 6, and 10-19 from the 5’ end of the sense strand; and (b) an antisense strand comprising 21 nucléotides, wherein 2’-fluoro nucléotides are at positions 2, 6, 14, and 16 from the 5’ end of the antisense strand, and wherein T-O-
353 methyl nucléotides are at positions 1,3-5,7-13, 15, and 17-21 from the 5’ end of the antisense strand.
20. The double stranded siNA of claim 19 further comprising (i) the nucléotides at positions
1 and 2 and positions 2 and 3 from the 5’ end of the sense strand are connected by
5 phosphorothioate intemucleoside linkages; and (ii) the nucléotides at positions 1 and 2;
positions 2 and 3; positions 19 and 20; and positions 20 and 21 from the 5’ end of the antisense strand are connected by phosphorothioate intemucleoside linkages.
21. The double stranded siNA of claim 19 or 21 further comprising a conjugated moiety attached to the sense strand.
10
22. The double stranded siNA of claim 21, wherein the conjugated moiety is N- acetylgalactosamine (GalNAc).
23. The double stranded siNA of claim 19 or 20, wherein the sense strand comprises SEQ ID NO: 438, and the antisense strand comprises any one of SEQ ID NO: 501, SEQ ID NO: 537, SEQ ID NO: 538, or SEQ ID NO: 539.
15
24. The double stranded siNA of claim 23, wherein the sense strand comprises SEQ ID NO:
438 and the antisense comprises SEQ ID NO: 501.
25. The double stranded siNA of claim 24 further comprising a N-acetylgalactosamine (GalNAc) attached to the 3’ of the sense strand, wherein the GalNAc comprises a structure of Formula (VII):
wherein n is 1, and R is OH.
26. The double stranded siNA of claim 23, wherein the sense strand comprises SEQ ID NO:
438 and the antisense strand comprises any one of SEQ ID NO: 537-539.
354
27. The double stranded siNA of claim 19 or 20, wherein the sense strand is at least 80% identical to SEQ ID NO: 40 and the antisense strand is at least 80% complementary to SEQ ID NO: 40.
28. The double stranded siNA of claim 19 or 20, wherein the sense strand or the antisense strand comprises at least one overhang consisting of 1 or 2 nucléotides.
29. A double stranded short interfering nucleic acid (siNA) molécule comprising:
(a) a sense strand comprising a nucleic acid sequence consisting of SEQ ID NO: 438 or SEQ ID NO: 435, and (b) an antisense strand comprising a nucleic acid sequence consisting of any one of SEQ ID NO: 501, SEQ ID NO: 505, SEQ ID NO: 506, SEQ ID NO: 537, SEQ ID NO: 538, and SEQ ID NO: 539.
30. The double stranded siNA molécule of claim 29 further comprising a Nacetylgalactosamine (GalNAc) attached to the 3 ’ of the sense strand.
31. The double stranded siNA molécule of claim 30, wherein the GalNAc comprises a structure of Formula (VII):
wherein n is 1, and R is OH.
32. A double stranded short interfering nucleic acid (siNA) comprising:
(a) a sense strand of 5’- mGpsmUpsmGmGfUmGfGfAfCmUmUmCmUmCmUmCmAmAmU-p-ps2GalNAc-3’ (SEQ ID NO: 438) and an antisense strand of 5’mApsfUpsmUmGmAfGmAmGmAmAmGmUmCfCmAfCmCmAmCpsmGpsm
A-3’ (SEQ ID NO: 501);
355 (b) a sense strand of 5’- mGpsmUpsmGmGfUmGfGfAfCmUmUmCmUmCmUmCmAmAmU-p-ps2GalNAc-3’ (SEQ ID NO: 438) and an antisense strand of 5’mApsfUpsmUmGmAfGmAfGfAmAmGmUmCfCmAfCmCmAmCpsmGpsmA3’ (SEQ ID NO: 521);
(c) a sense strand of 5’- mGpsmUpsmGmGfUmGfGfAfCmUmUmCmUmCmUmCmAmAmU-p-ps2GalNAc-3’ (SEQ ID NO: 438) and an antisense strand of 5’mApsfUpsmUmGfAmGmAfGmAmAmGmUmCfCmAmCfCmAmCpsmGpsmA3’ (SEQ IDNO: 522);
(d) a sense strand of 5’- mGpsmUpsmGmGfUmGfGfAfCmUmUmCmUmCmUmCmAmAmU-p-ps2GalNAc-3’ (SEQ ID NO: 438) and an antisense strand of 5’mApsf4PpsmUmGmAfGmAmGmAmAmGmUmCfCmAfCmCmAmCpsmGpsm
A-3’ (SEQ ID NO: 537);
(e) a sense strand of 5’- mGpsmUpsmGmGfUmGfGfAfCmUmUmCmUmCmUmCmAmAmU-p-ps2GalNAc-3’ (SEQ ID NO: 438) and an antisense strand of 5’mApsfUpsmUmGmAfGmAmGmAniAmGmUmCf2PmAfCmCmAmCpsmGpsm
A-3’ (SEQ ID NO: 538); or (f) a sense strand of 5’- mGpsmUpsmGmGfUmGfGfAfCmUmUmCmUmCmUmCmAmAmU-p-ps2GalNAc-3’ (SEQ ID NO: 438) and an antisense strand of 5’mApsfUpsmUmGmAfGmAmGmAmAmGmUmCfCmAfXmCmAmCpsmGpsm
A-3’ (SEQ ID NO: 539), wherein fX is
wherein GalNAc comprises a structure of Formula (VII):
356
wherein n is 1, and R is OH.
33. The double stranded siNA of claim 32, comprising a sense strand of 5’mGpsmUpsmGmGfUmGfGfAfCmUmUmCmUmCmUmCmAmAmU-p-ps2- GalNAc-3 ’
5 (SEQ ID NO: 438) and an antisense strand of 5’mApsfUpsmUmGmAfGmAmGmAmAmGmUmCfCmAfCmCmAmCpsmGpsmA-3’ (SEQ ID NO: 501).
34. Use of the double stranded siNA according to any one of claims 16-33 in the manufacture of a médicament for treating hepatitis B virus (HBV).
OA1202200362 2020-03-06 2021-03-05 Modified short interfering nucleic acid (siNA) molecules and uses thereof. OA21336A (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US62/986,150 2020-03-06
US63/109,196 2020-11-03

Publications (1)

Publication Number Publication Date
OA21336A true OA21336A (en) 2024-05-10

Family

ID=

Similar Documents

Publication Publication Date Title
US11549110B2 (en) Modified short interfering nucleic acid (siNA) molecules and uses thereof
RU2612521C2 (en) Novel prodrugs of nucleic acids and their application methods
CA2812962C (en) Azido nucleosides and nucleotide analogs
WO2020097342A1 (en) S-antigen transport inhibiting oligonucleotide polymers and methods
US20210189392A1 (en) S-antigen transport inhibiting oligonucleotide polymers and methods
AU2014217892A1 (en) Design of short oligonucleotides as vaccine adjuvants and therapeutic agents
EP4133080A1 (en) Short interfering nucleic acid (sina) molecules and uses thereof for coronavirus diseases
EP4232455A2 (en) Novel ligands for asialoglycoprotein receptor
US20230159929A1 (en) MODIFIED SHORT INTERFERING NUCLEIC ACID (siNA) MOLECULES AND USES THEREOF
WO2023039076A1 (en) Modified short interfering nucleic acid (sina) molecules and uses thereof
OA21336A (en) Modified short interfering nucleic acid (siNA) molecules and uses thereof.
TW202321449A (en) Oligonucleotide-based delivery vehicle for oligonucleotides agents and methods of use thereof
WO2024097674A2 (en) Modified short interfering nucleic acid (sina) molecules and uses thereof
KR20240099159A (en) Modified short interfering nucleic acid (SINA) molecules and uses thereof
AU2022339846A1 (en) Pnpla3-targeting short interfering rna (sirna) molecules and uses thereof
CN118234862A (en) Modified short interfering nucleic acid molecules (siNA) and uses thereof
WO2023177808A1 (en) Modified gapmer oligomers and methods of use thereof
JP2024515194A (en) siRNA and siRNA complexes targeting 17β-hydroxysteroid dehydrogenase type 13
CN116615542A (en) Systemic delivery of oligonucleotides