WO2024002046A1 - Oligonucleotide delivery enhancing compounds, pharmaceutical compositions and methods using the same - Google Patents
Oligonucleotide delivery enhancing compounds, pharmaceutical compositions and methods using the same Download PDFInfo
- Publication number
- WO2024002046A1 WO2024002046A1 PCT/CN2023/102623 CN2023102623W WO2024002046A1 WO 2024002046 A1 WO2024002046 A1 WO 2024002046A1 CN 2023102623 W CN2023102623 W CN 2023102623W WO 2024002046 A1 WO2024002046 A1 WO 2024002046A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- alkylene
- alkyl
- arylene
- cycloalkylene
- group
- Prior art date
Links
- 150000001875 compounds Chemical class 0.000 title claims abstract description 321
- 108091034117 Oligonucleotide Proteins 0.000 title claims abstract description 225
- 238000000034 method Methods 0.000 title claims abstract description 54
- 239000008194 pharmaceutical composition Substances 0.000 title claims abstract description 29
- 230000002708 enhancing effect Effects 0.000 title claims description 66
- 108090000623 proteins and genes Proteins 0.000 claims abstract description 78
- 230000014509 gene expression Effects 0.000 claims abstract description 56
- JLCPHMBAVCMARE-UHFFFAOYSA-N [3-[[3-[[3-[[3-[[3-[[3-[[3-[[3-[[3-[[3-[[3-[[5-(2-amino-6-oxo-1H-purin-9-yl)-3-[[3-[[3-[[3-[[3-[[3-[[5-(2-amino-6-oxo-1H-purin-9-yl)-3-[[5-(2-amino-6-oxo-1H-purin-9-yl)-3-hydroxyoxolan-2-yl]methoxy-hydroxyphosphoryl]oxyoxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(5-methyl-2,4-dioxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxyoxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(5-methyl-2,4-dioxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(4-amino-2-oxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(5-methyl-2,4-dioxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(5-methyl-2,4-dioxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(4-amino-2-oxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(4-amino-2-oxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(4-amino-2-oxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(4-amino-2-oxopyrimidin-1-yl)oxolan-2-yl]methyl [5-(6-aminopurin-9-yl)-2-(hydroxymethyl)oxolan-3-yl] hydrogen phosphate Polymers Cc1cn(C2CC(OP(O)(=O)OCC3OC(CC3OP(O)(=O)OCC3OC(CC3O)n3cnc4c3nc(N)[nH]c4=O)n3cnc4c3nc(N)[nH]c4=O)C(COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3CO)n3cnc4c(N)ncnc34)n3ccc(N)nc3=O)n3cnc4c(N)ncnc34)n3ccc(N)nc3=O)n3ccc(N)nc3=O)n3ccc(N)nc3=O)n3cnc4c(N)ncnc34)n3cnc4c(N)ncnc34)n3cc(C)c(=O)[nH]c3=O)n3cc(C)c(=O)[nH]c3=O)n3ccc(N)nc3=O)n3cc(C)c(=O)[nH]c3=O)n3cnc4c3nc(N)[nH]c4=O)n3cnc4c(N)ncnc34)n3cnc4c(N)ncnc34)n3cnc4c(N)ncnc34)n3cnc4c(N)ncnc34)O2)c(=O)[nH]c1=O JLCPHMBAVCMARE-UHFFFAOYSA-N 0.000 claims abstract description 23
- 125000000217 alkyl group Chemical group 0.000 claims description 415
- 125000002947 alkylene group Chemical group 0.000 claims description 255
- 125000003837 (C1-C20) alkyl group Chemical group 0.000 claims description 175
- 125000004104 aryloxy group Chemical group 0.000 claims description 126
- 125000000753 cycloalkyl group Chemical group 0.000 claims description 126
- 125000003545 alkoxy group Chemical group 0.000 claims description 110
- IJGRMHOSHXDMSA-UHFFFAOYSA-N Atomic nitrogen Chemical compound N#N IJGRMHOSHXDMSA-UHFFFAOYSA-N 0.000 claims description 86
- 125000002993 cycloalkylene group Chemical group 0.000 claims description 85
- 125000002887 hydroxy group Chemical group [H]O* 0.000 claims description 83
- 125000000732 arylene group Chemical group 0.000 claims description 80
- 239000000203 mixture Substances 0.000 claims description 78
- 125000003118 aryl group Chemical group 0.000 claims description 71
- 125000001424 substituent group Chemical group 0.000 claims description 57
- 229910052799 carbon Inorganic materials 0.000 claims description 56
- 229910052739 hydrogen Inorganic materials 0.000 claims description 54
- 239000001257 hydrogen Substances 0.000 claims description 53
- 229910052757 nitrogen Inorganic materials 0.000 claims description 53
- 229910052760 oxygen Inorganic materials 0.000 claims description 53
- VYPSYNLAJGMNEJ-UHFFFAOYSA-N Silicium dioxide Chemical compound O=[Si]=O VYPSYNLAJGMNEJ-UHFFFAOYSA-N 0.000 claims description 52
- 239000001301 oxygen Substances 0.000 claims description 52
- QVGXLLKOCUKJST-UHFFFAOYSA-N atomic oxygen Chemical compound [O] QVGXLLKOCUKJST-UHFFFAOYSA-N 0.000 claims description 51
- 125000005549 heteroarylene group Chemical group 0.000 claims description 49
- 229910052717 sulfur Inorganic materials 0.000 claims description 49
- NINIDFKCEFEMDL-UHFFFAOYSA-N Sulfur Chemical compound [S] NINIDFKCEFEMDL-UHFFFAOYSA-N 0.000 claims description 48
- 210000004027 cell Anatomy 0.000 claims description 48
- 239000011593 sulfur Substances 0.000 claims description 48
- 230000008685 targeting Effects 0.000 claims description 46
- 108020004459 Small interfering RNA Proteins 0.000 claims description 44
- OKTJSMMVPCPJKN-UHFFFAOYSA-N Carbon Chemical compound [C] OKTJSMMVPCPJKN-UHFFFAOYSA-N 0.000 claims description 43
- 125000005647 linker group Chemical group 0.000 claims description 40
- 229910002027 silica gel Inorganic materials 0.000 claims description 40
- 239000000741 silica gel Substances 0.000 claims description 40
- 150000001413 amino acids Chemical class 0.000 claims description 37
- 239000004055 small Interfering RNA Substances 0.000 claims description 37
- HVYWMOMLDIMFJA-DPAQBDIFSA-N cholesterol Chemical compound C1C=C2C[C@@H](O)CC[C@]2(C)[C@@H]2[C@@H]1[C@@H]1CC[C@H]([C@H](C)CCCC(C)C)[C@@]1(C)CC2 HVYWMOMLDIMFJA-DPAQBDIFSA-N 0.000 claims description 34
- 125000004453 alkoxycarbonyl group Chemical group 0.000 claims description 33
- 125000003277 amino group Chemical group 0.000 claims description 33
- 125000003178 carboxy group Chemical group [H]OC(*)=O 0.000 claims description 33
- ORILYTVJVMAKLC-UHFFFAOYSA-N adamantane Chemical compound C1C(C2)CC3CC1CC2C3 ORILYTVJVMAKLC-UHFFFAOYSA-N 0.000 claims description 32
- SIKJAQJRHWYJAI-UHFFFAOYSA-N Indole Chemical class C1=CC=C2NC=CC2=C1 SIKJAQJRHWYJAI-UHFFFAOYSA-N 0.000 claims description 30
- 150000007523 nucleic acids Chemical class 0.000 claims description 30
- 125000004429 atom Chemical group 0.000 claims description 29
- 229940124447 delivery agent Drugs 0.000 claims description 29
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 claims description 29
- 125000005843 halogen group Chemical group 0.000 claims description 28
- 239000002253 acid Substances 0.000 claims description 27
- 125000000000 cycloalkoxy group Chemical group 0.000 claims description 27
- 125000004400 (C1-C12) alkyl group Chemical group 0.000 claims description 26
- RAXXELZNTBOGNW-UHFFFAOYSA-N imidazole Natural products C1=CNC=N1 RAXXELZNTBOGNW-UHFFFAOYSA-N 0.000 claims description 26
- 102000039446 nucleic acids Human genes 0.000 claims description 26
- 108020004707 nucleic acids Proteins 0.000 claims description 26
- 150000001720 carbohydrates Chemical class 0.000 claims description 25
- 235000014633 carbohydrates Nutrition 0.000 claims description 25
- 229920001223 polyethylene glycol Polymers 0.000 claims description 25
- 239000000074 antisense oligonucleotide Substances 0.000 claims description 24
- 238000012230 antisense oligonucleotides Methods 0.000 claims description 24
- -1 lipophile Chemical class 0.000 claims description 24
- 125000004435 hydrogen atom Chemical class [H]* 0.000 claims description 23
- QTBSBXVTEAMEQO-UHFFFAOYSA-N acetic acid Substances CC(O)=O QTBSBXVTEAMEQO-UHFFFAOYSA-N 0.000 claims description 22
- 150000002632 lipids Chemical class 0.000 claims description 22
- 235000001014 amino acid Nutrition 0.000 claims description 21
- QDKGOMZIPXGDDJ-UHFFFAOYSA-N 2,3-dihydro-1h-indazole Chemical class C1=CC=C2CNNC2=C1 QDKGOMZIPXGDDJ-UHFFFAOYSA-N 0.000 claims description 20
- IOJUPLGTWVMSFF-UHFFFAOYSA-N benzothiazole Chemical class C1=CC=C2SC=NC2=C1 IOJUPLGTWVMSFF-UHFFFAOYSA-N 0.000 claims description 20
- RWRDLPDLKQPQOW-UHFFFAOYSA-N tetrahydropyrrole Natural products C1CCNC1 RWRDLPDLKQPQOW-UHFFFAOYSA-N 0.000 claims description 20
- 235000012000 cholesterol Nutrition 0.000 claims description 19
- 239000000463 material Substances 0.000 claims description 19
- UFHFLCQGNIYNRP-UHFFFAOYSA-N Hydrogen Chemical compound [H][H] UFHFLCQGNIYNRP-UHFFFAOYSA-N 0.000 claims description 18
- 201000010099 disease Diseases 0.000 claims description 18
- 210000004185 liver Anatomy 0.000 claims description 18
- 108090000765 processed proteins & peptides Proteins 0.000 claims description 18
- NBIIXXVUZAFLBC-UHFFFAOYSA-N Phosphoric acid Chemical group OP(O)(O)=O NBIIXXVUZAFLBC-UHFFFAOYSA-N 0.000 claims description 17
- 125000005161 aryl oxy carbonyl group Chemical group 0.000 claims description 17
- 125000004642 (C1-C12) alkoxy group Chemical group 0.000 claims description 16
- 229930013930 alkaloid Natural products 0.000 claims description 16
- 150000003797 alkaloid derivatives Chemical class 0.000 claims description 16
- 125000003342 alkenyl group Chemical group 0.000 claims description 16
- 125000000923 (C1-C30) alkyl group Chemical group 0.000 claims description 15
- WHTVZRBIWZFKQO-AWEZNQCLSA-N (S)-chloroquine Chemical compound ClC1=CC=C2C(N[C@@H](C)CCCN(CC)CC)=CC=NC2=C1 WHTVZRBIWZFKQO-AWEZNQCLSA-N 0.000 claims description 15
- WHTVZRBIWZFKQO-UHFFFAOYSA-N chloroquine Natural products ClC1=CC=C2C(NC(C)CCCN(CC)CC)=CC=NC2=C1 WHTVZRBIWZFKQO-UHFFFAOYSA-N 0.000 claims description 15
- 229960003677 chloroquine Drugs 0.000 claims description 15
- 239000002679 microRNA Substances 0.000 claims description 15
- 150000003431 steroids Chemical class 0.000 claims description 15
- 102000008394 Immunoglobulin Fragments Human genes 0.000 claims description 14
- 108010021625 Immunoglobulin Fragments Proteins 0.000 claims description 14
- 210000003734 kidney Anatomy 0.000 claims description 14
- 108091032973 (ribonucleotides)n+m Proteins 0.000 claims description 13
- 108020005004 Guide RNA Proteins 0.000 claims description 13
- 241000124008 Mammalia Species 0.000 claims description 13
- HYZJCKYKOHLVJF-UHFFFAOYSA-N 1H-benzimidazole Chemical class C1=CC=C2NC=NC2=C1 HYZJCKYKOHLVJF-UHFFFAOYSA-N 0.000 claims description 12
- 210000003169 central nervous system Anatomy 0.000 claims description 12
- 125000001181 organosilyl group Chemical group [SiH3]* 0.000 claims description 12
- 210000000278 spinal cord Anatomy 0.000 claims description 12
- 102000040650 (ribonucleotides)n+m Human genes 0.000 claims description 11
- 108700011259 MicroRNAs Proteins 0.000 claims description 11
- 229910019142 PO4 Inorganic materials 0.000 claims description 11
- 230000003213 activating effect Effects 0.000 claims description 11
- 210000004556 brain Anatomy 0.000 claims description 11
- 208000035475 disorder Diseases 0.000 claims description 11
- 210000002216 heart Anatomy 0.000 claims description 11
- 125000000623 heterocyclic group Chemical group 0.000 claims description 11
- 239000003446 ligand Substances 0.000 claims description 11
- 125000002560 nitrile group Chemical group 0.000 claims description 11
- QJGQUHMNIGDVPM-UHFFFAOYSA-N nitrogen group Chemical group [N] QJGQUHMNIGDVPM-UHFFFAOYSA-N 0.000 claims description 11
- 239000010452 phosphate Substances 0.000 claims description 11
- 150000004713 phosphodiesters Chemical group 0.000 claims description 11
- 125000006239 protecting group Chemical group 0.000 claims description 11
- 125000003161 (C1-C6) alkylene group Chemical group 0.000 claims description 10
- BCMCBBGGLRIHSE-UHFFFAOYSA-N 1,3-benzoxazole Chemical class C1=CC=C2OC=NC2=C1 BCMCBBGGLRIHSE-UHFFFAOYSA-N 0.000 claims description 10
- KAESVJOAVNADME-UHFFFAOYSA-N 1H-pyrrole Natural products C=1C=CNC=1 KAESVJOAVNADME-UHFFFAOYSA-N 0.000 claims description 10
- 108091023037 Aptamer Proteins 0.000 claims description 10
- 229910000147 aluminium phosphate Inorganic materials 0.000 claims description 10
- 150000001721 carbon Chemical group 0.000 claims description 10
- 125000000392 cycloalkenyl group Chemical group 0.000 claims description 10
- 150000002460 imidazoles Chemical class 0.000 claims description 10
- LPAGFVYQRIESJQ-UHFFFAOYSA-N indoline Chemical class C1=CC=C2NCCC2=C1 LPAGFVYQRIESJQ-UHFFFAOYSA-N 0.000 claims description 10
- 210000004962 mammalian cell Anatomy 0.000 claims description 10
- 150000002916 oxazoles Chemical class 0.000 claims description 10
- SXADIBFZNXBEGI-UHFFFAOYSA-N phosphoramidous acid Chemical group NP(O)O SXADIBFZNXBEGI-UHFFFAOYSA-N 0.000 claims description 10
- 150000003217 pyrazoles Chemical class 0.000 claims description 10
- 150000003218 pyrazolidines Chemical class 0.000 claims description 10
- 150000003219 pyrazolines Chemical class 0.000 claims description 10
- 150000003233 pyrroles Chemical class 0.000 claims description 10
- 150000003235 pyrrolidines Chemical class 0.000 claims description 10
- ZVJHJDDKYZXRJI-UHFFFAOYSA-N pyrroline Natural products C1CC=NC1 ZVJHJDDKYZXRJI-UHFFFAOYSA-N 0.000 claims description 10
- 150000003236 pyrrolines Chemical class 0.000 claims description 10
- 125000005471 saturated fatty acid group Chemical group 0.000 claims description 10
- 150000003384 small molecules Chemical class 0.000 claims description 10
- 150000003557 thiazoles Chemical class 0.000 claims description 10
- 125000005314 unsaturated fatty acid group Chemical group 0.000 claims description 10
- 210000003932 urinary bladder Anatomy 0.000 claims description 10
- OVRNDRQMDRJTHS-CBQIKETKSA-N N-Acetyl-D-Galactosamine Chemical compound CC(=O)N[C@H]1[C@@H](O)O[C@H](CO)[C@H](O)[C@@H]1O OVRNDRQMDRJTHS-CBQIKETKSA-N 0.000 claims description 9
- 229910052740 iodine Inorganic materials 0.000 claims description 9
- 210000004072 lung Anatomy 0.000 claims description 9
- 210000000496 pancreas Anatomy 0.000 claims description 9
- MBLBDJOUHNCFQT-UHFFFAOYSA-N N-acetyl-D-galactosamine Natural products CC(=O)NC(C=O)C(O)C(O)C(O)CO MBLBDJOUHNCFQT-UHFFFAOYSA-N 0.000 claims description 8
- 241000283984 Rodentia Species 0.000 claims description 8
- 230000000692 anti-sense effect Effects 0.000 claims description 8
- 125000002791 glucosyl group Chemical group C1([C@H](O)[C@@H](O)[C@H](O)[C@H](O1)CO)* 0.000 claims description 8
- 210000003205 muscle Anatomy 0.000 claims description 8
- 229910052698 phosphorus Inorganic materials 0.000 claims description 8
- 210000000952 spleen Anatomy 0.000 claims description 8
- DLFVBJFMPXGRIB-UHFFFAOYSA-N thioacetamide Natural products CC(N)=O DLFVBJFMPXGRIB-UHFFFAOYSA-N 0.000 claims description 8
- MSWZFWKMSRAUBD-GASJEMHNSA-N 2-amino-2-deoxy-D-galactopyranose Chemical compound N[C@H]1C(O)O[C@H](CO)[C@H](O)[C@@H]1O MSWZFWKMSRAUBD-GASJEMHNSA-N 0.000 claims description 7
- 239000002202 Polyethylene glycol Substances 0.000 claims description 7
- 102100025342 Voltage-dependent N-type calcium channel subunit alpha-1B Human genes 0.000 claims description 7
- 101710088658 Voltage-dependent N-type calcium channel subunit alpha-1B Proteins 0.000 claims description 7
- MSWZFWKMSRAUBD-UHFFFAOYSA-N beta-D-galactosamine Natural products NC1C(O)OC(CO)C(O)C1O MSWZFWKMSRAUBD-UHFFFAOYSA-N 0.000 claims description 7
- 210000000988 bone and bone Anatomy 0.000 claims description 7
- 229910052805 deuterium Inorganic materials 0.000 claims description 7
- 239000003937 drug carrier Substances 0.000 claims description 7
- 210000000936 intestine Anatomy 0.000 claims description 7
- 125000004433 nitrogen atom Chemical group N* 0.000 claims description 7
- 210000002784 stomach Anatomy 0.000 claims description 7
- 125000002103 4,4'-dimethoxytriphenylmethyl group Chemical group [H]C1=C([H])C([H])=C(C([H])=C1[H])C(*)(C1=C([H])C([H])=C(OC([H])([H])[H])C([H])=C1[H])C1=C([H])C([H])=C(OC([H])([H])[H])C([H])=C1[H] 0.000 claims description 6
- 125000004448 alkyl carbonyl group Chemical group 0.000 claims description 6
- 125000005129 aryl carbonyl group Chemical group 0.000 claims description 6
- 125000000051 benzyloxy group Chemical group [H]C1=C([H])C([H])=C(C([H])=C1[H])C([H])([H])O* 0.000 claims description 6
- 150000001975 deuterium Chemical group 0.000 claims description 6
- 210000001165 lymph node Anatomy 0.000 claims description 6
- 239000000546 pharmaceutical excipient Substances 0.000 claims description 6
- 239000000377 silicon dioxide Substances 0.000 claims description 6
- 239000002904 solvent Substances 0.000 claims description 6
- 108020005544 Antisense RNA Proteins 0.000 claims description 5
- 206010028980 Neoplasm Diseases 0.000 claims description 5
- 108091081021 Sense strand Proteins 0.000 claims description 5
- 201000011510 cancer Diseases 0.000 claims description 5
- 239000001913 cellulose Substances 0.000 claims description 5
- 229920002678 cellulose Polymers 0.000 claims description 5
- 239000003184 complementary RNA Substances 0.000 claims description 5
- 239000003085 diluting agent Substances 0.000 claims description 5
- 239000004615 ingredient Substances 0.000 claims description 5
- 229920000642 polymer Polymers 0.000 claims description 5
- 229910052796 boron Inorganic materials 0.000 claims description 4
- 239000000872 buffer Substances 0.000 claims description 4
- 239000000919 ceramic Substances 0.000 claims description 4
- 230000007423 decrease Effects 0.000 claims description 4
- 239000002270 dispersing agent Substances 0.000 claims description 4
- 239000011521 glass Substances 0.000 claims description 4
- 108091070501 miRNA Proteins 0.000 claims description 4
- 239000003755 preservative agent Substances 0.000 claims description 4
- 239000003381 stabilizer Substances 0.000 claims description 4
- ILMRJRBKQSSXGY-UHFFFAOYSA-N tert-butyl(dimethyl)silicon Chemical compound C[Si](C)C(C)(C)C ILMRJRBKQSSXGY-UHFFFAOYSA-N 0.000 claims description 4
- 125000001797 benzyl group Chemical group [H]C1=C([H])C([H])=C(C([H])=C1[H])C([H])([H])* 0.000 claims description 3
- 235000014113 dietary fatty acids Nutrition 0.000 claims description 3
- 229930195729 fatty acid Natural products 0.000 claims description 3
- 239000000194 fatty acid Substances 0.000 claims description 3
- 150000004665 fatty acids Chemical class 0.000 claims description 3
- 125000001981 tert-butyldimethylsilyl group Chemical group [H]C([H])([H])[Si]([H])(C([H])([H])[H])[*]C(C([H])([H])[H])(C([H])([H])[H])C([H])([H])[H] 0.000 claims description 3
- 150000001841 cholesterols Chemical class 0.000 claims description 2
- 210000005260 human cell Anatomy 0.000 claims description 2
- 102000004196 processed proteins & peptides Human genes 0.000 claims description 2
- 125000003453 indazolyl group Chemical class N1N=C(C2=C1C=CC=C2)* 0.000 claims 4
- 238000001727 in vivo Methods 0.000 abstract description 12
- 238000000338 in vitro Methods 0.000 abstract description 10
- 230000033228 biological regulation Effects 0.000 abstract description 2
- YMWUJEATGCHHMB-UHFFFAOYSA-N Dichloromethane Chemical compound ClCCl YMWUJEATGCHHMB-UHFFFAOYSA-N 0.000 description 218
- OKKJLVBELUTLKV-UHFFFAOYSA-N Methanol Chemical compound OC OKKJLVBELUTLKV-UHFFFAOYSA-N 0.000 description 132
- 239000000243 solution Substances 0.000 description 111
- 239000011541 reaction mixture Substances 0.000 description 110
- 125000004432 carbon atom Chemical group C* 0.000 description 101
- 239000012299 nitrogen atmosphere Substances 0.000 description 85
- 238000002360 preparation method Methods 0.000 description 75
- HEDRZPFGACZZDS-MICDWDOJSA-N Trichloro(2H)methane Chemical compound [2H]C(Cl)(Cl)Cl HEDRZPFGACZZDS-MICDWDOJSA-N 0.000 description 68
- WEVYAHXRMPXWCK-UHFFFAOYSA-N Acetonitrile Chemical compound CC#N WEVYAHXRMPXWCK-UHFFFAOYSA-N 0.000 description 67
- XLYOFNOQVPJJNP-UHFFFAOYSA-N water Substances O XLYOFNOQVPJJNP-UHFFFAOYSA-N 0.000 description 64
- 238000005160 1H NMR spectroscopy Methods 0.000 description 60
- XEKOWRVHYACXOJ-UHFFFAOYSA-N Ethyl acetate Chemical compound CCOC(C)=O XEKOWRVHYACXOJ-UHFFFAOYSA-N 0.000 description 60
- 238000006243 chemical reaction Methods 0.000 description 59
- 108020004999 messenger RNA Proteins 0.000 description 59
- 230000002829 reductive effect Effects 0.000 description 58
- 239000007832 Na2SO4 Substances 0.000 description 57
- PMZURENOXWZQFD-UHFFFAOYSA-L Sodium Sulfate Chemical compound [Na+].[Na+].[O-]S([O-])(=O)=O PMZURENOXWZQFD-UHFFFAOYSA-L 0.000 description 57
- 229910052938 sodium sulfate Inorganic materials 0.000 description 57
- RTZKZFJDLAIYFH-UHFFFAOYSA-N Diethyl ether Chemical compound CCOCC RTZKZFJDLAIYFH-UHFFFAOYSA-N 0.000 description 56
- 239000012074 organic phase Substances 0.000 description 53
- JGFZNNIVVJXRND-UHFFFAOYSA-N N,N-Diisopropylethylamine (DIPEA) Chemical compound CCN(C(C)C)C(C)C JGFZNNIVVJXRND-UHFFFAOYSA-N 0.000 description 45
- 238000003818 flash chromatography Methods 0.000 description 45
- 238000004949 mass spectrometry Methods 0.000 description 44
- 239000007787 solid Substances 0.000 description 44
- 238000011282 treatment Methods 0.000 description 42
- 239000012267 brine Substances 0.000 description 38
- 239000000047 product Substances 0.000 description 38
- HPALAKNZSZLMCH-UHFFFAOYSA-M sodium;chloride;hydrate Chemical compound O.[Na+].[Cl-] HPALAKNZSZLMCH-UHFFFAOYSA-M 0.000 description 38
- 239000012043 crude product Substances 0.000 description 37
- 239000003480 eluent Substances 0.000 description 34
- 125000003729 nucleotide group Chemical group 0.000 description 34
- WFDIJRYMOXRFFG-UHFFFAOYSA-N Acetic anhydride Chemical compound CC(=O)OC(C)=O WFDIJRYMOXRFFG-UHFFFAOYSA-N 0.000 description 33
- 210000001519 tissue Anatomy 0.000 description 33
- JUJWROOIHBZHMG-UHFFFAOYSA-N Pyridine Chemical compound C1=CC=NC=C1 JUJWROOIHBZHMG-UHFFFAOYSA-N 0.000 description 30
- 239000007924 injection Substances 0.000 description 26
- 238000002347 injection Methods 0.000 description 26
- 239000002773 nucleotide Substances 0.000 description 26
- ZMANZCXQSJIPKH-UHFFFAOYSA-N Triethylamine Chemical compound CCN(CC)CC ZMANZCXQSJIPKH-UHFFFAOYSA-N 0.000 description 24
- 230000000694 effects Effects 0.000 description 24
- 229920002477 rna polymer Polymers 0.000 description 23
- VHYFNPMBLIVWCW-UHFFFAOYSA-N 4-Dimethylaminopyridine Chemical compound CN(C)C1=CC=NC=C1 VHYFNPMBLIVWCW-UHFFFAOYSA-N 0.000 description 22
- 239000005289 controlled pore glass Substances 0.000 description 22
- 230000004048 modification Effects 0.000 description 22
- 238000012986 modification Methods 0.000 description 22
- 238000000746 purification Methods 0.000 description 21
- 101150062190 sod1 gene Proteins 0.000 description 21
- LFQSCWFLJHTTHZ-UHFFFAOYSA-N Ethanol Chemical compound CCO LFQSCWFLJHTTHZ-UHFFFAOYSA-N 0.000 description 20
- 229940024606 amino acid Drugs 0.000 description 20
- 235000019439 ethyl acetate Nutrition 0.000 description 20
- 238000003197 gene knockdown Methods 0.000 description 20
- 241000699666 Mus <mouse, genus> Species 0.000 description 19
- FAPWRFPIFSIZLT-UHFFFAOYSA-M Sodium chloride Chemical compound [Na+].[Cl-] FAPWRFPIFSIZLT-UHFFFAOYSA-M 0.000 description 19
- 150000002431 hydrogen Chemical class 0.000 description 18
- 239000011780 sodium chloride Substances 0.000 description 18
- 241001465754 Metazoa Species 0.000 description 17
- 241000699670 Mus sp. Species 0.000 description 17
- 238000011746 C57BL/6J (JAX™ mouse strain) Methods 0.000 description 16
- 102000008221 Superoxide Dismutase-1 Human genes 0.000 description 16
- 108010021188 Superoxide Dismutase-1 Proteins 0.000 description 16
- 239000012230 colorless oil Substances 0.000 description 15
- UMJSCPRVCHMLSP-UHFFFAOYSA-N pyridine Natural products COC1=CC=CN=C1 UMJSCPRVCHMLSP-UHFFFAOYSA-N 0.000 description 15
- 241000700159 Rattus Species 0.000 description 14
- 239000003981 vehicle Substances 0.000 description 13
- KDLHZDBZIXYQEI-UHFFFAOYSA-N Palladium Chemical compound [Pd] KDLHZDBZIXYQEI-UHFFFAOYSA-N 0.000 description 12
- 239000011324 bead Substances 0.000 description 12
- 238000001990 intravenous administration Methods 0.000 description 12
- 239000002924 silencing RNA Substances 0.000 description 12
- FALRKNHUBBKYCC-UHFFFAOYSA-N 2-(chloromethyl)pyridine-3-carbonitrile Chemical compound ClCC1=NC=CC=C1C#N FALRKNHUBBKYCC-UHFFFAOYSA-N 0.000 description 11
- 229960000549 4-dimethylaminophenol Drugs 0.000 description 11
- 239000003921 oil Substances 0.000 description 11
- 235000019198 oils Nutrition 0.000 description 11
- 229940014800 succinic anhydride Drugs 0.000 description 11
- 229910010084 LiAlH4 Inorganic materials 0.000 description 10
- 239000012280 lithium aluminium hydride Substances 0.000 description 10
- BWHMMNNQKKPAPP-UHFFFAOYSA-L potassium carbonate Chemical compound [K+].[K+].[O-]C([O-])=O BWHMMNNQKKPAPP-UHFFFAOYSA-L 0.000 description 10
- LJCNRYVRMXRIQR-OLXYHTOASA-L potassium sodium L-tartrate Chemical class [Na+].[K+].[O-]C(=O)[C@H](O)[C@@H](O)C([O-])=O LJCNRYVRMXRIQR-OLXYHTOASA-L 0.000 description 10
- 125000006413 ring segment Chemical group 0.000 description 10
- UIIMBOGNXHQVGW-UHFFFAOYSA-M Sodium bicarbonate Chemical class [Na+].OC([O-])=O UIIMBOGNXHQVGW-UHFFFAOYSA-M 0.000 description 9
- 230000000295 complement effect Effects 0.000 description 9
- 229910052731 fluorine Inorganic materials 0.000 description 9
- YWWARDMVSMPOLR-UHFFFAOYSA-M oxolane;tetrabutylazanium;fluoride Chemical compound [F-].C1CCOC1.CCCC[N+](CCCC)(CCCC)CCCC YWWARDMVSMPOLR-UHFFFAOYSA-M 0.000 description 9
- 102000004169 proteins and genes Human genes 0.000 description 9
- 239000003795 chemical substances by application Substances 0.000 description 8
- 210000000056 organ Anatomy 0.000 description 8
- 239000002336 ribonucleotide Substances 0.000 description 8
- JBWYRBLDOOOJEU-UHFFFAOYSA-N 1-[chloro-(4-methoxyphenyl)-phenylmethyl]-4-methoxybenzene Chemical compound C1=CC(OC)=CC=C1C(Cl)(C=1C=CC(OC)=CC=1)C1=CC=CC=C1 JBWYRBLDOOOJEU-UHFFFAOYSA-N 0.000 description 7
- 101100366159 Mus musculus Sod1 gene Proteins 0.000 description 7
- 238000002123 RNA extraction Methods 0.000 description 7
- 108091028664 Ribonucleotide Proteins 0.000 description 7
- 238000010839 reverse transcription Methods 0.000 description 7
- 125000002652 ribonucleotide group Chemical group 0.000 description 7
- 238000010254 subcutaneous injection Methods 0.000 description 7
- 239000007929 subcutaneous injection Substances 0.000 description 7
- 230000001225 therapeutic effect Effects 0.000 description 7
- 238000001644 13C nuclear magnetic resonance spectroscopy Methods 0.000 description 6
- ZCYVEMRRCGMTRW-UHFFFAOYSA-N 7553-56-2 Chemical compound [I] ZCYVEMRRCGMTRW-UHFFFAOYSA-N 0.000 description 6
- WKBOTKDWSSQWDR-UHFFFAOYSA-N Bromine atom Chemical compound [Br] WKBOTKDWSSQWDR-UHFFFAOYSA-N 0.000 description 6
- ZAMOUSCENKQFHK-UHFFFAOYSA-N Chlorine atom Chemical compound [Cl] ZAMOUSCENKQFHK-UHFFFAOYSA-N 0.000 description 6
- PXGOKWXKJXAPGV-UHFFFAOYSA-N Fluorine Chemical compound FF PXGOKWXKJXAPGV-UHFFFAOYSA-N 0.000 description 6
- OAICVXFJPJFONN-UHFFFAOYSA-N Phosphorus Chemical compound [P] OAICVXFJPJFONN-UHFFFAOYSA-N 0.000 description 6
- HEMHJVSKTPXQMS-UHFFFAOYSA-M Sodium hydroxide Chemical compound [OH-].[Na+] HEMHJVSKTPXQMS-UHFFFAOYSA-M 0.000 description 6
- 125000002252 acyl group Chemical group 0.000 description 6
- 239000012298 atmosphere Substances 0.000 description 6
- GDTBXPJZTBHREO-UHFFFAOYSA-N bromine Substances BrBr GDTBXPJZTBHREO-UHFFFAOYSA-N 0.000 description 6
- 229910052794 bromium Inorganic materials 0.000 description 6
- 238000007385 chemical modification Methods 0.000 description 6
- 239000000460 chlorine Substances 0.000 description 6
- 229910052801 chlorine Inorganic materials 0.000 description 6
- 125000004185 ester group Chemical group 0.000 description 6
- 125000001033 ether group Chemical group 0.000 description 6
- 239000011737 fluorine Substances 0.000 description 6
- 229910052736 halogen Inorganic materials 0.000 description 6
- 150000002367 halogens Chemical class 0.000 description 6
- 150000002473 indoazoles Chemical class 0.000 description 6
- 238000000185 intracerebroventricular administration Methods 0.000 description 6
- 239000011630 iodine Substances 0.000 description 6
- VLKZOEOYAKHREP-UHFFFAOYSA-N n-Hexane Chemical compound CCCCCC VLKZOEOYAKHREP-UHFFFAOYSA-N 0.000 description 6
- 239000012044 organic layer Substances 0.000 description 6
- 239000011574 phosphorus Substances 0.000 description 6
- 239000000843 powder Substances 0.000 description 6
- 238000010926 purge Methods 0.000 description 6
- 210000002027 skeletal muscle Anatomy 0.000 description 6
- RYYWUUFWQRZTIU-UHFFFAOYSA-K thiophosphate Chemical compound [O-]P([O-])([O-])=S RYYWUUFWQRZTIU-UHFFFAOYSA-K 0.000 description 6
- KOFLVDBWRHFSAB-UHFFFAOYSA-N 1,2,4,5-tetrahydro-1-(phenylmethyl)-5,9b(1',2')-benzeno-9bh-benz(g)indol-3(3ah)-one Chemical compound C1C(C=2C3=CC=CC=2)C2=CC=CC=C2C23C1C(=O)CN2CC1=CC=CC=C1 KOFLVDBWRHFSAB-UHFFFAOYSA-N 0.000 description 5
- 108020004414 DNA Proteins 0.000 description 5
- 102000053602 DNA Human genes 0.000 description 5
- 108060001084 Luciferase Proteins 0.000 description 5
- 239000005089 Luciferase Substances 0.000 description 5
- 101150014879 RpL13A gene Proteins 0.000 description 5
- VZTDIZULWFCMLS-UHFFFAOYSA-N ammonium formate Chemical compound [NH4+].[O-]C=O VZTDIZULWFCMLS-UHFFFAOYSA-N 0.000 description 5
- 210000001638 cerebellum Anatomy 0.000 description 5
- 239000003814 drug Substances 0.000 description 5
- 238000005516 engineering process Methods 0.000 description 5
- 230000000670 limiting effect Effects 0.000 description 5
- KWGKDLIKAYFUFQ-UHFFFAOYSA-M lithium chloride Chemical class [Li+].[Cl-] KWGKDLIKAYFUFQ-UHFFFAOYSA-M 0.000 description 5
- 210000002381 plasma Anatomy 0.000 description 5
- 229910000027 potassium carbonate Inorganic materials 0.000 description 5
- 235000018102 proteins Nutrition 0.000 description 5
- 125000001453 quaternary ammonium group Chemical group 0.000 description 5
- RWSOTUBLDIXVET-UHFFFAOYSA-O sulfonium Chemical compound [SH3+] RWSOTUBLDIXVET-UHFFFAOYSA-O 0.000 description 5
- 210000003462 vein Anatomy 0.000 description 5
- HTFNVAVTYILUCF-UHFFFAOYSA-N 2-[2-ethoxy-4-[4-(4-methylpiperazin-1-yl)piperidine-1-carbonyl]anilino]-5-methyl-11-methylsulfonylpyrimido[4,5-b][1,4]benzodiazepin-6-one Chemical compound CCOc1cc(ccc1Nc1ncc2N(C)C(=O)c3ccccc3N(c2n1)S(C)(=O)=O)C(=O)N1CCC(CC1)N1CCN(C)CC1 HTFNVAVTYILUCF-UHFFFAOYSA-N 0.000 description 4
- UOXJNGFFPMOZDM-UHFFFAOYSA-N 2-[di(propan-2-yl)amino]ethylsulfanyl-methylphosphinic acid Chemical compound CC(C)N(C(C)C)CCSP(C)(O)=O UOXJNGFFPMOZDM-UHFFFAOYSA-N 0.000 description 4
- QWTBDIBOOIAZEF-UHFFFAOYSA-N 3-[chloro-[di(propan-2-yl)amino]phosphanyl]oxypropanenitrile Chemical compound CC(C)N(C(C)C)P(Cl)OCCC#N QWTBDIBOOIAZEF-UHFFFAOYSA-N 0.000 description 4
- SFHYNDMGZXWXBU-LIMNOBDPSA-N 6-amino-2-[[(e)-(3-formylphenyl)methylideneamino]carbamoylamino]-1,3-dioxobenzo[de]isoquinoline-5,8-disulfonic acid Chemical compound O=C1C(C2=3)=CC(S(O)(=O)=O)=CC=3C(N)=C(S(O)(=O)=O)C=C2C(=O)N1NC(=O)N\N=C\C1=CC=CC(C=O)=C1 SFHYNDMGZXWXBU-LIMNOBDPSA-N 0.000 description 4
- 208000031648 Body Weight Changes Diseases 0.000 description 4
- WQZGKKKJIJFFOK-GASJEMHNSA-N Glucose Natural products OC[C@H]1OC(O)[C@H](O)[C@@H](O)[C@@H]1O WQZGKKKJIJFFOK-GASJEMHNSA-N 0.000 description 4
- 101000685323 Homo sapiens Succinate dehydrogenase [ubiquinone] flavoprotein subunit, mitochondrial Proteins 0.000 description 4
- 241000223503 Platysma Species 0.000 description 4
- 238000012228 RNA interference-mediated gene silencing Methods 0.000 description 4
- 102100023155 Succinate dehydrogenase [ubiquinone] flavoprotein subunit, mitochondrial Human genes 0.000 description 4
- WQZGKKKJIJFFOK-VFUOTHLCSA-N beta-D-glucose Chemical compound OC[C@H]1O[C@@H](O)[C@H](O)[C@@H](O)[C@@H]1O WQZGKKKJIJFFOK-VFUOTHLCSA-N 0.000 description 4
- 230000004579 body weight change Effects 0.000 description 4
- 210000004720 cerebrum Anatomy 0.000 description 4
- 238000009472 formulation Methods 0.000 description 4
- 210000005153 frontal cortex Anatomy 0.000 description 4
- 230000009368 gene silencing by RNA Effects 0.000 description 4
- 230000006872 improvement Effects 0.000 description 4
- CNJJSTPBUHAEFH-UHFFFAOYSA-N methyl 4-fluoro-3-nitrobenzoate Chemical compound COC(=O)C1=CC=C(F)C([N+]([O-])=O)=C1 CNJJSTPBUHAEFH-UHFFFAOYSA-N 0.000 description 4
- 230000000144 pharmacologic effect Effects 0.000 description 4
- XTRUQJBVQBUKSQ-UHFFFAOYSA-N propan-2-yl 4-[1-(2-fluoro-4-methylsulfonylphenyl)pyrazolo[3,4-d]pyrimidin-4-yl]oxypiperidine-1-carboxylate Chemical compound C1CN(C(=O)OC(C)C)CCC1OC1=NC=NC2=C1C=NN2C1=CC=C(S(C)(=O)=O)C=C1F XTRUQJBVQBUKSQ-UHFFFAOYSA-N 0.000 description 4
- 102200082402 rs751610198 Human genes 0.000 description 4
- 229940078677 sarna Drugs 0.000 description 4
- 210000000115 thoracic cavity Anatomy 0.000 description 4
- XNAMZBCMLZQBIC-UHFFFAOYSA-N 2-[2-[4-[5-[(1-ethylpyrazol-3-yl)amino]triazolo[4,5-d]pyrimidin-1-yl]phenyl]propan-2-yloxy]ethanol Chemical compound CCN1C=CC(NC=2N=C3N=NN(C3=CN=2)C=2C=CC(=CC=2)C(C)(C)OCCO)=N1 XNAMZBCMLZQBIC-UHFFFAOYSA-N 0.000 description 3
- 101000617738 Homo sapiens Survival motor neuron protein Proteins 0.000 description 3
- 101100298363 Mus musculus Ppig gene Proteins 0.000 description 3
- 108091028043 Nucleic acid sequence Proteins 0.000 description 3
- 101150071557 Ppig gene Proteins 0.000 description 3
- 241000288906 Primates Species 0.000 description 3
- DNIAPMSPPWPWGF-UHFFFAOYSA-N Propylene glycol Chemical compound CC(O)CO DNIAPMSPPWPWGF-UHFFFAOYSA-N 0.000 description 3
- 101100042953 Rattus norvegicus Sod1 gene Proteins 0.000 description 3
- 102100021947 Survival motor neuron protein Human genes 0.000 description 3
- 210000002376 aorta thoracic Anatomy 0.000 description 3
- 239000007864 aqueous solution Substances 0.000 description 3
- 230000015572 biosynthetic process Effects 0.000 description 3
- 210000004204 blood vessel Anatomy 0.000 description 3
- 230000001413 cellular effect Effects 0.000 description 3
- 231100000673 dose–response relationship Toxicity 0.000 description 3
- 230000006870 function Effects 0.000 description 3
- 239000008103 glucose Substances 0.000 description 3
- 230000002401 inhibitory effect Effects 0.000 description 3
- 238000007918 intramuscular administration Methods 0.000 description 3
- 238000007913 intrathecal administration Methods 0.000 description 3
- 238000011068 loading method Methods 0.000 description 3
- 230000007246 mechanism Effects 0.000 description 3
- 230000036515 potency Effects 0.000 description 3
- 210000001525 retina Anatomy 0.000 description 3
- 238000007920 subcutaneous administration Methods 0.000 description 3
- 239000000126 substance Substances 0.000 description 3
- 238000003786 synthesis reaction Methods 0.000 description 3
- 238000001890 transfection Methods 0.000 description 3
- YBJHBAHKTGYVGT-ZKWXMUAHSA-N (+)-Biotin Chemical compound N1C(=O)N[C@@H]2[C@H](CCCCC(=O)O)SC[C@@H]21 YBJHBAHKTGYVGT-ZKWXMUAHSA-N 0.000 description 2
- WGWCJTNWUFFGFH-UHFFFAOYSA-N 3-[tert-butyl(dimethyl)silyl]oxypropanal Chemical compound CC(C)(C)[Si](C)(C)OCCC=O WGWCJTNWUFFGFH-UHFFFAOYSA-N 0.000 description 2
- 101150087690 ACTB gene Proteins 0.000 description 2
- 241000251468 Actinopterygii Species 0.000 description 2
- 229920000936 Agarose Polymers 0.000 description 2
- GUBGYTABKSRVRQ-XLOQQCSPSA-N Alpha-Lactose Chemical compound O[C@@H]1[C@@H](O)[C@@H](O)[C@@H](CO)O[C@H]1O[C@@H]1[C@@H](CO)O[C@H](O)[C@H](O)[C@H]1O GUBGYTABKSRVRQ-XLOQQCSPSA-N 0.000 description 2
- 108020000948 Antisense Oligonucleotides Proteins 0.000 description 2
- 241000271566 Aves Species 0.000 description 2
- 241000283690 Bos taurus Species 0.000 description 2
- 102000003903 Cyclin-dependent kinases Human genes 0.000 description 2
- 108090000266 Cyclin-dependent kinases Proteins 0.000 description 2
- FBPFZTCFMRRESA-FSIIMWSLSA-N D-Glucitol Natural products OC[C@H](O)[C@H](O)[C@@H](O)[C@H](O)CO FBPFZTCFMRRESA-FSIIMWSLSA-N 0.000 description 2
- FBPFZTCFMRRESA-KVTDHHQDSA-N D-Mannitol Chemical compound OC[C@@H](O)[C@@H](O)[C@H](O)[C@H](O)CO FBPFZTCFMRRESA-KVTDHHQDSA-N 0.000 description 2
- FBPFZTCFMRRESA-JGWLITMVSA-N D-glucitol Chemical compound OC[C@H](O)[C@@H](O)[C@H](O)[C@H](O)CO FBPFZTCFMRRESA-JGWLITMVSA-N 0.000 description 2
- 238000012286 ELISA Assay Methods 0.000 description 2
- 241000282326 Felis catus Species 0.000 description 2
- PEDCQBHIVMGVHV-UHFFFAOYSA-N Glycerine Chemical compound OCC(O)CO PEDCQBHIVMGVHV-UHFFFAOYSA-N 0.000 description 2
- DHMQDGOQFOQNFH-UHFFFAOYSA-N Glycine Chemical compound NCC(O)=O DHMQDGOQFOQNFH-UHFFFAOYSA-N 0.000 description 2
- 101150034920 Hmbs gene Proteins 0.000 description 2
- 101000664887 Homo sapiens Superoxide dismutase [Cu-Zn] Proteins 0.000 description 2
- 101150003028 Hprt1 gene Proteins 0.000 description 2
- GUBGYTABKSRVRQ-QKKXKWKRSA-N Lactose Natural products OC[C@H]1O[C@@H](O[C@H]2[C@H](O)[C@@H](O)C(O)O[C@@H]2CO)[C@H](O)[C@@H](O)[C@H]1O GUBGYTABKSRVRQ-QKKXKWKRSA-N 0.000 description 2
- 108010047357 Luminescent Proteins Proteins 0.000 description 2
- 102000006830 Luminescent Proteins Human genes 0.000 description 2
- 102000043136 MAP kinase family Human genes 0.000 description 2
- 108091054455 MAP kinase family Proteins 0.000 description 2
- 229930195725 Mannitol Natural products 0.000 description 2
- 241000283973 Oryctolagus cuniculus Species 0.000 description 2
- 241000009328 Perro Species 0.000 description 2
- PYMYPHUHKUWMLA-LMVFSUKVSA-N Ribose Natural products OC[C@@H](O)[C@@H](O)[C@@H](O)C=O PYMYPHUHKUWMLA-LMVFSUKVSA-N 0.000 description 2
- CZMRCDWAGMRECN-UGDNZRGBSA-N Sucrose Chemical compound O[C@H]1[C@H](O)[C@@H](CO)O[C@@]1(CO)O[C@@H]1[C@H](O)[C@@H](O)[C@H](O)[C@@H](CO)O1 CZMRCDWAGMRECN-UGDNZRGBSA-N 0.000 description 2
- 229930006000 Sucrose Natural products 0.000 description 2
- 108010062481 Type 1 Angiotensin Receptor Proteins 0.000 description 2
- 102100026803 Type-1 angiotensin II receptor Human genes 0.000 description 2
- 238000010521 absorption reaction Methods 0.000 description 2
- DZBUGLKDJFMEHC-UHFFFAOYSA-N acridine Chemical compound C1=CC=CC2=CC3=CC=CC=C3N=C21 DZBUGLKDJFMEHC-UHFFFAOYSA-N 0.000 description 2
- 239000013543 active substance Substances 0.000 description 2
- HMFHBZSHGGEWLO-UHFFFAOYSA-N alpha-D-Furanose-Ribose Natural products OCC1OC(O)C(O)C1O HMFHBZSHGGEWLO-UHFFFAOYSA-N 0.000 description 2
- 230000009286 beneficial effect Effects 0.000 description 2
- 230000000975 bioactive effect Effects 0.000 description 2
- 230000008827 biological function Effects 0.000 description 2
- 210000005068 bladder tissue Anatomy 0.000 description 2
- 239000006143 cell culture medium Substances 0.000 description 2
- 230000004700 cellular uptake Effects 0.000 description 2
- 230000021615 conjugation Effects 0.000 description 2
- 239000006071 cream Substances 0.000 description 2
- 238000011161 development Methods 0.000 description 2
- 210000000188 diaphragm Anatomy 0.000 description 2
- UAOMVDZJSHZZME-UHFFFAOYSA-N diisopropylamine Substances CC(C)NC(C)C UAOMVDZJSHZZME-UHFFFAOYSA-N 0.000 description 2
- 229940043279 diisopropylamine Drugs 0.000 description 2
- 150000002016 disaccharides Chemical class 0.000 description 2
- 238000009826 distribution Methods 0.000 description 2
- 229940079593 drug Drugs 0.000 description 2
- 230000009977 dual effect Effects 0.000 description 2
- MMXKVMNBHPAILY-UHFFFAOYSA-N ethyl laurate Chemical compound CCCCCCCCCCCC(=O)OCC MMXKVMNBHPAILY-UHFFFAOYSA-N 0.000 description 2
- 238000002474 experimental method Methods 0.000 description 2
- 235000019688 fish Nutrition 0.000 description 2
- 230000030279 gene silencing Effects 0.000 description 2
- IPCSVZSSVZVIGE-UHFFFAOYSA-N hexadecanoic acid Chemical compound CCCCCCCCCCCCCCCC(O)=O IPCSVZSSVZVIGE-UHFFFAOYSA-N 0.000 description 2
- 102000056070 human SOD1 Human genes 0.000 description 2
- 230000002209 hydrophobic effect Effects 0.000 description 2
- 238000007912 intraperitoneal administration Methods 0.000 description 2
- 239000008101 lactose Substances 0.000 description 2
- 239000007788 liquid Substances 0.000 description 2
- 210000005228 liver tissue Anatomy 0.000 description 2
- 230000005291 magnetic effect Effects 0.000 description 2
- 239000000594 mannitol Substances 0.000 description 2
- 235000010355 mannitol Nutrition 0.000 description 2
- 125000000956 methoxy group Chemical group [H]C([H])([H])O* 0.000 description 2
- 150000002772 monosaccharides Chemical class 0.000 description 2
- 230000005298 paramagnetic effect Effects 0.000 description 2
- 238000007911 parenteral administration Methods 0.000 description 2
- 230000002093 peripheral effect Effects 0.000 description 2
- RDOWQLZANAYVLL-UHFFFAOYSA-N phenanthridine Chemical compound C1=CC=C2C3=CC=CC=C3C=NC2=C1 RDOWQLZANAYVLL-UHFFFAOYSA-N 0.000 description 2
- 229920001282 polysaccharide Polymers 0.000 description 2
- 239000005017 polysaccharide Substances 0.000 description 2
- 150000004804 polysaccharides Chemical class 0.000 description 2
- 230000000069 prophylactic effect Effects 0.000 description 2
- 229940126409 proton pump inhibitor Drugs 0.000 description 2
- 239000011734 sodium Substances 0.000 description 2
- 239000011343 solid material Substances 0.000 description 2
- 239000000600 sorbitol Substances 0.000 description 2
- 235000010356 sorbitol Nutrition 0.000 description 2
- 239000005720 sucrose Substances 0.000 description 2
- 230000004083 survival effect Effects 0.000 description 2
- 239000000725 suspension Substances 0.000 description 2
- 208000024891 symptom Diseases 0.000 description 2
- BCNZYOJHNLTNEZ-UHFFFAOYSA-N tert-butyldimethylsilyl chloride Chemical compound CC(C)(C)[Si](C)(C)Cl BCNZYOJHNLTNEZ-UHFFFAOYSA-N 0.000 description 2
- 238000004809 thin layer chromatography Methods 0.000 description 2
- APJYDQYYACXCRM-UHFFFAOYSA-N tryptamine Chemical compound C1=CC=C2C(CCN)=CNC2=C1 APJYDQYYACXCRM-UHFFFAOYSA-N 0.000 description 2
- 210000004127 vitreous body Anatomy 0.000 description 2
- HDTRYLNUVZCQOY-UHFFFAOYSA-N α-D-glucopyranosyl-α-D-glucopyranoside Natural products OC1C(O)C(O)C(CO)OC1OC1C(O)C(O)C(O)C(CO)O1 HDTRYLNUVZCQOY-UHFFFAOYSA-N 0.000 description 1
- MTCFGRXMJLQNBG-REOHCLBHSA-N (2S)-2-Amino-3-hydroxypropansäure Chemical compound OC[C@H](N)C(O)=O MTCFGRXMJLQNBG-REOHCLBHSA-N 0.000 description 1
- MDKGKXOCJGEUJW-VIFPVBQESA-N (2s)-2-[4-(thiophene-2-carbonyl)phenyl]propanoic acid Chemical compound C1=CC([C@@H](C(O)=O)C)=CC=C1C(=O)C1=CC=CS1 MDKGKXOCJGEUJW-VIFPVBQESA-N 0.000 description 1
- GOUUPUICWUFXPM-XIKOKIGWSA-N (2s,4r)-1-(9h-fluoren-9-ylmethoxycarbonyl)-4-hydroxypyrrolidine-2-carboxylic acid Chemical compound C1[C@H](O)C[C@@H](C(O)=O)N1C(=O)OCC1C2=CC=CC=C2C2=CC=CC=C21 GOUUPUICWUFXPM-XIKOKIGWSA-N 0.000 description 1
- OOKAZRDERJMRCJ-KOUAFAAESA-N (3r)-7-[(1s,2s,4ar,6s,8s)-2,6-dimethyl-8-[(2s)-2-methylbutanoyl]oxy-1,2,4a,5,6,7,8,8a-octahydronaphthalen-1-yl]-3-hydroxy-5-oxoheptanoic acid Chemical compound C1=C[C@H](C)[C@H](CCC(=O)C[C@@H](O)CC(O)=O)C2[C@@H](OC(=O)[C@@H](C)CC)C[C@@H](C)C[C@@H]21 OOKAZRDERJMRCJ-KOUAFAAESA-N 0.000 description 1
- 125000000027 (C1-C10) alkoxy group Chemical group 0.000 description 1
- 125000000008 (C1-C10) alkyl group Chemical group 0.000 description 1
- 125000006529 (C3-C6) alkyl group Chemical group 0.000 description 1
- LRANPJDWHYRCER-UHFFFAOYSA-N 1,2-diazepine Chemical compound N1C=CC=CC=N1 LRANPJDWHYRCER-UHFFFAOYSA-N 0.000 description 1
- RYHBNJHYFVUHQT-UHFFFAOYSA-N 1,4-Dioxane Chemical compound C1COCCO1 RYHBNJHYFVUHQT-UHFFFAOYSA-N 0.000 description 1
- XBCXJKGHPABGSD-UHFFFAOYSA-N 1-methyluracil Chemical compound CN1C=CC(=O)NC1=O XBCXJKGHPABGSD-UHFFFAOYSA-N 0.000 description 1
- OWEGMIWEEQEYGQ-UHFFFAOYSA-N 100676-05-9 Natural products OC1C(O)C(O)C(CO)OC1OCC1C(O)C(O)C(O)C(OC2C(OC(O)C(O)C2O)CO)O1 OWEGMIWEEQEYGQ-UHFFFAOYSA-N 0.000 description 1
- ZMZGFLUUZLELNE-UHFFFAOYSA-N 2,3,5-triiodobenzoic acid Chemical compound OC(=O)C1=CC(I)=CC(I)=C1I ZMZGFLUUZLELNE-UHFFFAOYSA-N 0.000 description 1
- VEPOHXYIFQMVHW-XOZOLZJESA-N 2,3-dihydroxybutanedioic acid (2S,3S)-3,4-dimethyl-2-phenylmorpholine Chemical compound OC(C(O)C(O)=O)C(O)=O.C[C@H]1[C@@H](OCCN1C)c1ccccc1 VEPOHXYIFQMVHW-XOZOLZJESA-N 0.000 description 1
- PIINGYXNCHTJTF-UHFFFAOYSA-N 2-(2-azaniumylethylamino)acetate Chemical group NCCNCC(O)=O PIINGYXNCHTJTF-UHFFFAOYSA-N 0.000 description 1
- BRLJKBOXIVONAG-UHFFFAOYSA-N 2-[[5-(dimethylamino)naphthalen-1-yl]sulfonyl-methylamino]acetic acid Chemical compound C1=CC=C2C(N(C)C)=CC=CC2=C1S(=O)(=O)N(C)CC(O)=O BRLJKBOXIVONAG-UHFFFAOYSA-N 0.000 description 1
- OALHHIHQOFIMEF-UHFFFAOYSA-N 3',6'-dihydroxy-2',4',5',7'-tetraiodo-3h-spiro[2-benzofuran-1,9'-xanthene]-3-one Chemical compound O1C(=O)C2=CC=CC=C2C21C1=CC(I)=C(O)C(I)=C1OC1=C(I)C(O)=C(I)C=C21 OALHHIHQOFIMEF-UHFFFAOYSA-N 0.000 description 1
- YTNNMAIKEURUTO-UHFFFAOYSA-N 5-[tert-butyl(dimethyl)silyl]oxypentanal Chemical compound CC(C)(C)[Si](C)(C)OCCCCC=O YTNNMAIKEURUTO-UHFFFAOYSA-N 0.000 description 1
- VVIAGPKUTFNRDU-UHFFFAOYSA-N 6S-folinic acid Natural products C1NC=2NC(N)=NC(=O)C=2N(C=O)C1CNC1=CC=C(C(=O)NC(CCC(O)=O)C(O)=O)C=C1 VVIAGPKUTFNRDU-UHFFFAOYSA-N 0.000 description 1
- MSSXOMSJDRHRMC-UHFFFAOYSA-N 9H-purine-2,6-diamine Chemical compound NC1=NC(N)=C2NC=NC2=N1 MSSXOMSJDRHRMC-UHFFFAOYSA-N 0.000 description 1
- 241000014654 Adna Species 0.000 description 1
- 229920001817 Agar Polymers 0.000 description 1
- 239000004475 Arginine Substances 0.000 description 1
- 102000008682 Argonaute Proteins Human genes 0.000 description 1
- 108010088141 Argonaute Proteins Proteins 0.000 description 1
- BSYNRYMUTXBXSQ-UHFFFAOYSA-N Aspirin Chemical compound CC(=O)OC1=CC=CC=C1C(O)=O BSYNRYMUTXBXSQ-UHFFFAOYSA-N 0.000 description 1
- 241000416162 Astragalus gummifer Species 0.000 description 1
- 241000282672 Ateles sp. Species 0.000 description 1
- 206010003694 Atrophy Diseases 0.000 description 1
- 241000972773 Aulopiformes Species 0.000 description 1
- 208000023275 Autoimmune disease Diseases 0.000 description 1
- 108091005950 Azurite Proteins 0.000 description 1
- 208000035143 Bacterial infection Diseases 0.000 description 1
- 108091032955 Bacterial small RNA Proteins 0.000 description 1
- 241000157302 Bison bison athabascae Species 0.000 description 1
- 241000282465 Canis Species 0.000 description 1
- 241000282461 Canis lupus Species 0.000 description 1
- 102000020313 Cell-Penetrating Peptides Human genes 0.000 description 1
- 108010051109 Cell-Penetrating Peptides Proteins 0.000 description 1
- 229930186147 Cephalosporin Natural products 0.000 description 1
- 108091005944 Cerulean Proteins 0.000 description 1
- 241000282994 Cervidae Species 0.000 description 1
- JZUFKLXOESDKRF-UHFFFAOYSA-N Chlorothiazide Chemical compound C1=C(Cl)C(S(=O)(=O)N)=CC2=C1NCNS2(=O)=O JZUFKLXOESDKRF-UHFFFAOYSA-N 0.000 description 1
- 108010077544 Chromatin Proteins 0.000 description 1
- 102100023804 Coagulation factor VII Human genes 0.000 description 1
- 108020004635 Complementary DNA Proteins 0.000 description 1
- 229920002261 Corn starch Polymers 0.000 description 1
- 241000699800 Cricetinae Species 0.000 description 1
- WQZGKKKJIJFFOK-QTVWNMPRSA-N D-mannopyranose Chemical compound OC[C@H]1OC(O)[C@@H](O)[C@@H](O)[C@@H]1O WQZGKKKJIJFFOK-QTVWNMPRSA-N 0.000 description 1
- HMFHBZSHGGEWLO-SOOFDHNKSA-N D-ribofuranose Chemical compound OC[C@H]1OC(O)[C@H](O)[C@@H]1O HMFHBZSHGGEWLO-SOOFDHNKSA-N 0.000 description 1
- 125000000824 D-ribofuranosyl group Chemical group [H]OC([H])([H])[C@@]1([H])OC([H])(*)[C@]([H])(O[H])[C@]1([H])O[H] 0.000 description 1
- YZCKVEUIGOORGS-OUBTZVSYSA-N Deuterium Chemical group [2H] YZCKVEUIGOORGS-OUBTZVSYSA-N 0.000 description 1
- 102000015554 Dopamine receptor Human genes 0.000 description 1
- 108050004812 Dopamine receptor Proteins 0.000 description 1
- 241000271571 Dromaius novaehollandiae Species 0.000 description 1
- 108091005941 EBFP Proteins 0.000 description 1
- 108091005942 ECFP Proteins 0.000 description 1
- LVGKNOAMLMIIKO-UHFFFAOYSA-N Elaidinsaeure-aethylester Natural products CCCCCCCCC=CCCCCCCCC(=O)OCC LVGKNOAMLMIIKO-UHFFFAOYSA-N 0.000 description 1
- 241000196324 Embryophyta Species 0.000 description 1
- 241000283086 Equidae Species 0.000 description 1
- 241000283073 Equus caballus Species 0.000 description 1
- 239000001856 Ethyl cellulose Substances 0.000 description 1
- ZZSNKZQZMQGXPY-UHFFFAOYSA-N Ethyl cellulose Chemical compound CCOCC1OC(OC)C(OCC)C(OCC)C1OC1C(O)C(O)C(OC)C(CO)O1 ZZSNKZQZMQGXPY-UHFFFAOYSA-N 0.000 description 1
- 108010023321 Factor VII Proteins 0.000 description 1
- 241000282324 Felis Species 0.000 description 1
- MPJKWIXIYCLVCU-UHFFFAOYSA-N Folinic acid Natural products NC1=NC2=C(N(C=O)C(CNc3ccc(cc3)C(=O)NC(CCC(=O)O)CC(=O)O)CN2)C(=O)N1 MPJKWIXIYCLVCU-UHFFFAOYSA-N 0.000 description 1
- 241000287828 Gallus gallus Species 0.000 description 1
- 241000963438 Gaussia <copepod> Species 0.000 description 1
- 108010010803 Gelatin Proteins 0.000 description 1
- WHUUTDBJXJRKMK-UHFFFAOYSA-N Glutamic acid Natural products OC(=O)C(N)CCC(O)=O WHUUTDBJXJRKMK-UHFFFAOYSA-N 0.000 description 1
- 239000004471 Glycine Substances 0.000 description 1
- 241000238631 Hexapoda Species 0.000 description 1
- 102000000543 Histamine Receptors Human genes 0.000 description 1
- 108010002059 Histamine Receptors Proteins 0.000 description 1
- 241000282412 Homo Species 0.000 description 1
- 101000780643 Homo sapiens Protein argonaute-2 Proteins 0.000 description 1
- 102000008100 Human Serum Albumin Human genes 0.000 description 1
- 108091006905 Human Serum Albumin Proteins 0.000 description 1
- 206010020772 Hypertension Diseases 0.000 description 1
- HEFNNWSXXWATRW-UHFFFAOYSA-N Ibuprofen Chemical compound CC(C)CC1=CC=C(C(C)C(O)=O)C=C1 HEFNNWSXXWATRW-UHFFFAOYSA-N 0.000 description 1
- 206010061218 Inflammation Diseases 0.000 description 1
- 102100034343 Integrase Human genes 0.000 description 1
- 101710203526 Integrase Proteins 0.000 description 1
- LKDRXBCSQODPBY-AMVSKUEXSA-N L-(-)-Sorbose Chemical compound OCC1(O)OC[C@H](O)[C@@H](O)[C@@H]1O LKDRXBCSQODPBY-AMVSKUEXSA-N 0.000 description 1
- XUJNEKJLAYXESH-REOHCLBHSA-N L-Cysteine Chemical compound SC[C@H](N)C(O)=O XUJNEKJLAYXESH-REOHCLBHSA-N 0.000 description 1
- ONIBWKKTOPOVIA-BYPYZUCNSA-N L-Proline Chemical compound OC(=O)[C@@H]1CCCN1 ONIBWKKTOPOVIA-BYPYZUCNSA-N 0.000 description 1
- QNAYBMKLOCPYGJ-REOHCLBHSA-N L-alanine Chemical compound C[C@H](N)C(O)=O QNAYBMKLOCPYGJ-REOHCLBHSA-N 0.000 description 1
- ODKSFYDXXFIFQN-BYPYZUCNSA-P L-argininium(2+) Chemical compound NC(=[NH2+])NCCC[C@H]([NH3+])C(O)=O ODKSFYDXXFIFQN-BYPYZUCNSA-P 0.000 description 1
- CKLJMWTZIZZHCS-REOHCLBHSA-N L-aspartic acid Chemical compound OC(=O)[C@@H](N)CC(O)=O CKLJMWTZIZZHCS-REOHCLBHSA-N 0.000 description 1
- WHUUTDBJXJRKMK-VKHMYHEASA-N L-glutamic acid Chemical compound OC(=O)[C@@H](N)CCC(O)=O WHUUTDBJXJRKMK-VKHMYHEASA-N 0.000 description 1
- ZDXPYRJPNDTMRX-VKHMYHEASA-N L-glutamine Chemical compound OC(=O)[C@@H](N)CCC(N)=O ZDXPYRJPNDTMRX-VKHMYHEASA-N 0.000 description 1
- HNDVDQJCIGZPNO-YFKPBYRVSA-N L-histidine Chemical compound OC(=O)[C@@H](N)CC1=CN=CN1 HNDVDQJCIGZPNO-YFKPBYRVSA-N 0.000 description 1
- AGPKZVBTJJNPAG-WHFBIAKZSA-N L-isoleucine Chemical compound CC[C@H](C)[C@H](N)C(O)=O AGPKZVBTJJNPAG-WHFBIAKZSA-N 0.000 description 1
- ROHFNLRQFUQHCH-YFKPBYRVSA-N L-leucine Chemical compound CC(C)C[C@H](N)C(O)=O ROHFNLRQFUQHCH-YFKPBYRVSA-N 0.000 description 1
- KDXKERNSBIXSRK-YFKPBYRVSA-N L-lysine Chemical compound NCCCC[C@H](N)C(O)=O KDXKERNSBIXSRK-YFKPBYRVSA-N 0.000 description 1
- FFEARJCKVFRZRR-BYPYZUCNSA-N L-methionine Chemical compound CSCC[C@H](N)C(O)=O FFEARJCKVFRZRR-BYPYZUCNSA-N 0.000 description 1
- COLNVLDHVKWLRT-QMMMGPOBSA-N L-phenylalanine Chemical compound OC(=O)[C@@H](N)CC1=CC=CC=C1 COLNVLDHVKWLRT-QMMMGPOBSA-N 0.000 description 1
- AYFVYJQAPQTCCC-GBXIJSLDSA-N L-threonine Chemical compound C[C@@H](O)[C@H](N)C(O)=O AYFVYJQAPQTCCC-GBXIJSLDSA-N 0.000 description 1
- QIVBCDIJIAJPQS-VIFPVBQESA-N L-tryptophane Chemical compound C1=CC=C2C(C[C@H](N)C(O)=O)=CNC2=C1 QIVBCDIJIAJPQS-VIFPVBQESA-N 0.000 description 1
- OUYCCCASQSFEME-QMMMGPOBSA-N L-tyrosine Chemical compound OC(=O)[C@@H](N)CC1=CC=C(O)C=C1 OUYCCCASQSFEME-QMMMGPOBSA-N 0.000 description 1
- KZSNJWFQEVHDMF-BYPYZUCNSA-N L-valine Chemical compound CC(C)[C@H](N)C(O)=O KZSNJWFQEVHDMF-BYPYZUCNSA-N 0.000 description 1
- 102000004407 Lactalbumin Human genes 0.000 description 1
- 108090000942 Lactalbumin Proteins 0.000 description 1
- ROHFNLRQFUQHCH-UHFFFAOYSA-N Leucine Natural products CC(C)CC(N)C(O)=O ROHFNLRQFUQHCH-UHFFFAOYSA-N 0.000 description 1
- DUKURNFHYQXCJG-UHFFFAOYSA-N Lewis A pentasaccharide Natural products OC1C(O)C(O)C(C)OC1OC1C(OC2C(C(O)C(O)C(CO)O2)O)C(NC(C)=O)C(OC2C(C(OC3C(OC(O)C(O)C3O)CO)OC(CO)C2O)O)OC1CO DUKURNFHYQXCJG-UHFFFAOYSA-N 0.000 description 1
- 241000254056 Luciola Species 0.000 description 1
- KDXKERNSBIXSRK-UHFFFAOYSA-N Lysine Natural products NCCCCC(N)C(O)=O KDXKERNSBIXSRK-UHFFFAOYSA-N 0.000 description 1
- 239000004472 Lysine Substances 0.000 description 1
- 241000282553 Macaca Species 0.000 description 1
- 241000282567 Macaca fascicularis Species 0.000 description 1
- GUBGYTABKSRVRQ-PICCSMPSSA-N Maltose Natural products O[C@@H]1[C@@H](O)[C@H](O)[C@@H](CO)O[C@@H]1O[C@@H]1[C@@H](CO)OC(O)[C@H](O)[C@H]1O GUBGYTABKSRVRQ-PICCSMPSSA-N 0.000 description 1
- 241000283923 Marmota monax Species 0.000 description 1
- 101100088246 Mus musculus Rpl13a gene Proteins 0.000 description 1
- 241000282339 Mustela Species 0.000 description 1
- GXCLVBGFBYZDAG-UHFFFAOYSA-N N-[2-(1H-indol-3-yl)ethyl]-N-methylprop-2-en-1-amine Chemical compound CN(CCC1=CNC2=C1C=CC=C2)CC=C GXCLVBGFBYZDAG-UHFFFAOYSA-N 0.000 description 1
- OVRNDRQMDRJTHS-FMDGEEDCSA-N N-acetyl-beta-D-glucosamine Chemical compound CC(=O)N[C@H]1[C@H](O)O[C@H](CO)[C@@H](O)[C@@H]1O OVRNDRQMDRJTHS-FMDGEEDCSA-N 0.000 description 1
- 108010077850 Nuclear Localization Signals Proteins 0.000 description 1
- 235000021314 Palmitic acid Nutrition 0.000 description 1
- 241000282579 Pan Species 0.000 description 1
- 208000030852 Parasitic disease Diseases 0.000 description 1
- 235000019483 Peanut oil Nutrition 0.000 description 1
- 208000008469 Peptic Ulcer Diseases 0.000 description 1
- PCNDJXKNXGMECE-UHFFFAOYSA-N Phenazine Natural products C1=CC=CC2=NC3=CC=CC=C3N=C21 PCNDJXKNXGMECE-UHFFFAOYSA-N 0.000 description 1
- 241000360551 Phrixothrix Species 0.000 description 1
- 239000004952 Polyamide Substances 0.000 description 1
- 229920002732 Polyanhydride Polymers 0.000 description 1
- 102100032709 Potassium-transporting ATPase alpha chain 2 Human genes 0.000 description 1
- ONIBWKKTOPOVIA-UHFFFAOYSA-N Proline Natural products OC(=O)C1CCCN1 ONIBWKKTOPOVIA-UHFFFAOYSA-N 0.000 description 1
- 108010029485 Protein Isoforms Proteins 0.000 description 1
- 102000001708 Protein Isoforms Human genes 0.000 description 1
- 102100034207 Protein argonaute-2 Human genes 0.000 description 1
- 108010083204 Proton Pumps Proteins 0.000 description 1
- 241001427617 Pyrophorus Species 0.000 description 1
- 108010052090 Renilla Luciferases Proteins 0.000 description 1
- 108091029810 SaRNA Proteins 0.000 description 1
- 235000019485 Safflower oil Nutrition 0.000 description 1
- 241000277331 Salmonidae Species 0.000 description 1
- MTCFGRXMJLQNBG-UHFFFAOYSA-N Serine Natural products OCC(N)C(O)=O MTCFGRXMJLQNBG-UHFFFAOYSA-N 0.000 description 1
- 229920002472 Starch Polymers 0.000 description 1
- 241000272534 Struthio camelus Species 0.000 description 1
- 241000282887 Suidae Species 0.000 description 1
- 108091046869 Telomeric non-coding RNA Proteins 0.000 description 1
- RYYWUUFWQRZTIU-UHFFFAOYSA-N Thiophosphoric acid Chemical group OP(O)(S)=O RYYWUUFWQRZTIU-UHFFFAOYSA-N 0.000 description 1
- AYFVYJQAPQTCCC-UHFFFAOYSA-N Threonine Natural products CC(O)C(N)C(O)=O AYFVYJQAPQTCCC-UHFFFAOYSA-N 0.000 description 1
- 239000004473 Threonine Substances 0.000 description 1
- 229920001615 Tragacanth Polymers 0.000 description 1
- 108700019146 Transgenes Proteins 0.000 description 1
- HDTRYLNUVZCQOY-WSWWMNSNSA-N Trehalose Natural products O[C@@H]1[C@@H](O)[C@@H](O)[C@@H](CO)O[C@@H]1O[C@@H]1[C@H](O)[C@@H](O)[C@@H](O)[C@@H](CO)O1 HDTRYLNUVZCQOY-WSWWMNSNSA-N 0.000 description 1
- 102000018690 Trypsinogen Human genes 0.000 description 1
- 108010027252 Trypsinogen Proteins 0.000 description 1
- QIVBCDIJIAJPQS-UHFFFAOYSA-N Tryptophan Natural products C1=CC=C2C(CC(N)C(O)=O)=CNC2=C1 QIVBCDIJIAJPQS-UHFFFAOYSA-N 0.000 description 1
- KZSNJWFQEVHDMF-UHFFFAOYSA-N Valine Natural products CC(C)C(N)C(O)=O KZSNJWFQEVHDMF-UHFFFAOYSA-N 0.000 description 1
- 241000251539 Vertebrata <Metazoa> Species 0.000 description 1
- 241000282485 Vulpes vulpes Species 0.000 description 1
- DUDJTRNGXIUJEB-UHFFFAOYSA-N [N].NCC(O)=O Chemical group [N].NCC(O)=O DUDJTRNGXIUJEB-UHFFFAOYSA-N 0.000 description 1
- DPXJVFZANSGRMM-UHFFFAOYSA-N acetic acid;2,3,4,5,6-pentahydroxyhexanal;sodium Chemical compound [Na].CC(O)=O.OCC(O)C(O)C(O)C(O)C=O DPXJVFZANSGRMM-UHFFFAOYSA-N 0.000 description 1
- 229960001138 acetylsalicylic acid Drugs 0.000 description 1
- 230000009471 action Effects 0.000 description 1
- 239000008186 active pharmaceutical agent Substances 0.000 description 1
- 239000000654 additive Substances 0.000 description 1
- 239000008272 agar Substances 0.000 description 1
- 235000004279 alanine Nutrition 0.000 description 1
- 239000000783 alginic acid Substances 0.000 description 1
- 235000010443 alginic acid Nutrition 0.000 description 1
- 229920000615 alginic acid Polymers 0.000 description 1
- 229960001126 alginic acid Drugs 0.000 description 1
- 150000004781 alginic acids Chemical class 0.000 description 1
- HDTRYLNUVZCQOY-LIZSDCNHSA-N alpha,alpha-trehalose Chemical compound O[C@@H]1[C@@H](O)[C@H](O)[C@@H](CO)O[C@@H]1O[C@@H]1[C@H](O)[C@@H](O)[C@H](O)[C@@H](CO)O1 HDTRYLNUVZCQOY-LIZSDCNHSA-N 0.000 description 1
- WQZGKKKJIJFFOK-PHYPRBDBSA-N alpha-D-galactose Chemical compound OC[C@H]1O[C@H](O)[C@H](O)[C@@H](O)[C@H]1O WQZGKKKJIJFFOK-PHYPRBDBSA-N 0.000 description 1
- WNROFYMDJYEPJX-UHFFFAOYSA-K aluminium hydroxide Chemical compound [OH-].[OH-].[OH-].[Al+3] WNROFYMDJYEPJX-UHFFFAOYSA-K 0.000 description 1
- 206010002026 amyotrophic lateral sclerosis Diseases 0.000 description 1
- 238000010171 animal model Methods 0.000 description 1
- PYKYMHQGRFAEBM-UHFFFAOYSA-N anthraquinone Natural products CCC(=O)c1c(O)c2C(=O)C3C(C=CC=C3O)C(=O)c2cc1CC(=O)OC PYKYMHQGRFAEBM-UHFFFAOYSA-N 0.000 description 1
- 150000004056 anthraquinones Chemical class 0.000 description 1
- 239000003242 anti bacterial agent Substances 0.000 description 1
- 230000000844 anti-bacterial effect Effects 0.000 description 1
- 230000003178 anti-diabetic effect Effects 0.000 description 1
- 239000003472 antidiabetic agent Substances 0.000 description 1
- ODKSFYDXXFIFQN-UHFFFAOYSA-N arginine Natural products OC(=O)C(N)CCCNC(N)=N ODKSFYDXXFIFQN-UHFFFAOYSA-N 0.000 description 1
- 235000003704 aspartic acid Nutrition 0.000 description 1
- 230000037444 atrophy Effects 0.000 description 1
- 230000001580 bacterial effect Effects 0.000 description 1
- 208000022362 bacterial infectious disease Diseases 0.000 description 1
- 229940125717 barbiturate Drugs 0.000 description 1
- HNYOPLTXPVRDBG-UHFFFAOYSA-N barbituric acid Chemical compound O=C1CC(=O)NC(=O)N1 HNYOPLTXPVRDBG-UHFFFAOYSA-N 0.000 description 1
- 230000008901 benefit Effects 0.000 description 1
- 150000001556 benzimidazoles Chemical class 0.000 description 1
- ZYGHJZDHTFUPRJ-UHFFFAOYSA-N benzo-alpha-pyrone Natural products C1=CC=C2OC(=O)C=CC2=C1 ZYGHJZDHTFUPRJ-UHFFFAOYSA-N 0.000 description 1
- JXWABCYGGVHAHB-UHFFFAOYSA-N benzyl n-(3-aminopropyl)carbamate Chemical compound NCCCNC(=O)OCC1=CC=CC=C1 JXWABCYGGVHAHB-UHFFFAOYSA-N 0.000 description 1
- OQFSQFPPLPISGP-UHFFFAOYSA-N beta-carboxyaspartic acid Natural products OC(=O)C(N)C(C(O)=O)C(O)=O OQFSQFPPLPISGP-UHFFFAOYSA-N 0.000 description 1
- GUBGYTABKSRVRQ-QUYVBRFLSA-N beta-maltose Chemical compound OC[C@H]1O[C@H](O[C@H]2[C@H](O)[C@@H](O)[C@H](O)O[C@@H]2CO)[C@H](O)[C@@H](O)[C@@H]1O GUBGYTABKSRVRQ-QUYVBRFLSA-N 0.000 description 1
- 238000005842 biochemical reaction Methods 0.000 description 1
- 230000003115 biocidal effect Effects 0.000 description 1
- 230000005540 biological transmission Effects 0.000 description 1
- 229960002685 biotin Drugs 0.000 description 1
- 235000020958 biotin Nutrition 0.000 description 1
- 239000011616 biotin Substances 0.000 description 1
- 230000000903 blocking effect Effects 0.000 description 1
- MCQRPQCQMGVWIQ-UHFFFAOYSA-N boron;methylsulfanylmethane Chemical compound [B].CSC MCQRPQCQMGVWIQ-UHFFFAOYSA-N 0.000 description 1
- 210000000133 brain stem Anatomy 0.000 description 1
- 239000008366 buffered solution Substances 0.000 description 1
- 239000006172 buffering agent Substances 0.000 description 1
- 239000001768 carboxy methyl cellulose Substances 0.000 description 1
- PUXBGTOOZJQSKH-UHFFFAOYSA-N carprofen Chemical compound C1=C(Cl)C=C2C3=CC=C(C(C(O)=O)C)C=C3NC2=C1 PUXBGTOOZJQSKH-UHFFFAOYSA-N 0.000 description 1
- 229960003184 carprofen Drugs 0.000 description 1
- 241001233037 catfish Species 0.000 description 1
- 210000000170 cell membrane Anatomy 0.000 description 1
- 229920002301 cellulose acetate Polymers 0.000 description 1
- 229940124587 cephalosporin Drugs 0.000 description 1
- 150000001780 cephalosporins Chemical class 0.000 description 1
- 210000001175 cerebrospinal fluid Anatomy 0.000 description 1
- 239000007795 chemical reaction product Substances 0.000 description 1
- 239000003153 chemical reaction reagent Substances 0.000 description 1
- 229960002155 chlorothiazide Drugs 0.000 description 1
- 125000003716 cholic acid group Chemical group 0.000 description 1
- 210000003483 chromatin Anatomy 0.000 description 1
- 210000000349 chromosome Anatomy 0.000 description 1
- 238000003776 cleavage reaction Methods 0.000 description 1
- 229940110456 cocoa butter Drugs 0.000 description 1
- 235000019868 cocoa butter Nutrition 0.000 description 1
- 238000004440 column chromatography Methods 0.000 description 1
- 238000013270 controlled release Methods 0.000 description 1
- 235000005687 corn oil Nutrition 0.000 description 1
- 239000002285 corn oil Substances 0.000 description 1
- 239000008120 corn starch Substances 0.000 description 1
- 235000012343 cottonseed oil Nutrition 0.000 description 1
- 239000002385 cottonseed oil Substances 0.000 description 1
- 235000001671 coumarin Nutrition 0.000 description 1
- 150000004775 coumarins Chemical class 0.000 description 1
- 230000008878 coupling Effects 0.000 description 1
- 238000010168 coupling process Methods 0.000 description 1
- 238000005859 coupling reaction Methods 0.000 description 1
- XUJNEKJLAYXESH-UHFFFAOYSA-N cysteine Natural products SCC(N)C(O)=O XUJNEKJLAYXESH-UHFFFAOYSA-N 0.000 description 1
- 235000018417 cysteine Nutrition 0.000 description 1
- 210000000805 cytoplasm Anatomy 0.000 description 1
- 108700007153 dansylsarcosine Proteins 0.000 description 1
- 230000003247 decreasing effect Effects 0.000 description 1
- 230000006735 deficit Effects 0.000 description 1
- 230000000593 degrading effect Effects 0.000 description 1
- 239000005547 deoxyribonucleotide Substances 0.000 description 1
- 125000002637 deoxyribonucleotide group Chemical group 0.000 description 1
- 238000013461 design Methods 0.000 description 1
- 238000003795 desorption Methods 0.000 description 1
- 238000006642 detritylation reaction Methods 0.000 description 1
- 125000004431 deuterium atom Chemical group 0.000 description 1
- 239000008121 dextrose Substances 0.000 description 1
- 238000003745 diagnosis Methods 0.000 description 1
- 230000002222 downregulating effect Effects 0.000 description 1
- 229940088679 drug related substance Drugs 0.000 description 1
- 239000000975 dye Substances 0.000 description 1
- 108010048367 enhanced green fluorescent protein Proteins 0.000 description 1
- 239000003623 enhancer Substances 0.000 description 1
- 150000002148 esters Chemical class 0.000 description 1
- 235000019441 ethanol Nutrition 0.000 description 1
- 235000019325 ethyl cellulose Nutrition 0.000 description 1
- 229920001249 ethyl cellulose Polymers 0.000 description 1
- LVGKNOAMLMIIKO-QXMHVHEDSA-N ethyl oleate Chemical compound CCCCCCCC\C=C/CCCCCCCC(=O)OCC LVGKNOAMLMIIKO-QXMHVHEDSA-N 0.000 description 1
- 229940093471 ethyl oleate Drugs 0.000 description 1
- 229940012413 factor vii Drugs 0.000 description 1
- ZPAKPRAICRBAOD-UHFFFAOYSA-N fenbufen Chemical compound C1=CC(C(=O)CCC(=O)O)=CC=C1C1=CC=CC=C1 ZPAKPRAICRBAOD-UHFFFAOYSA-N 0.000 description 1
- 229960001395 fenbufen Drugs 0.000 description 1
- 239000000945 filler Substances 0.000 description 1
- 229960000556 fingolimod Drugs 0.000 description 1
- KKGQTZUTZRNORY-UHFFFAOYSA-N fingolimod Chemical compound CCCCCCCCC1=CC=C(CCC(N)(CO)CO)C=C1 KKGQTZUTZRNORY-UHFFFAOYSA-N 0.000 description 1
- LPEPZBJOKDYZAD-UHFFFAOYSA-N flufenamic acid Chemical compound OC(=O)C1=CC=CC=C1NC1=CC=CC(C(F)(F)F)=C1 LPEPZBJOKDYZAD-UHFFFAOYSA-N 0.000 description 1
- 229960004369 flufenamic acid Drugs 0.000 description 1
- 108010021843 fluorescent protein 583 Proteins 0.000 description 1
- 108091006047 fluorescent proteins Proteins 0.000 description 1
- 102000034287 fluorescent proteins Human genes 0.000 description 1
- 239000006260 foam Substances 0.000 description 1
- 229940014144 folate Drugs 0.000 description 1
- OVBPIULPVIDEAO-LBPRGKRZSA-N folic acid Chemical compound C=1N=C2NC(N)=NC(=O)C2=NC=1CNC1=CC=C(C(=O)N[C@@H](CCC(O)=O)C(O)=O)C=C1 OVBPIULPVIDEAO-LBPRGKRZSA-N 0.000 description 1
- 235000019152 folic acid Nutrition 0.000 description 1
- 239000011724 folic acid Substances 0.000 description 1
- VVIAGPKUTFNRDU-ABLWVSNPSA-N folinic acid Chemical compound C1NC=2NC(N)=NC(=O)C=2N(C=O)C1CNC1=CC=C(C(=O)N[C@@H](CCC(O)=O)C(O)=O)C=C1 VVIAGPKUTFNRDU-ABLWVSNPSA-N 0.000 description 1
- 235000008191 folinic acid Nutrition 0.000 description 1
- 239000011672 folinic acid Substances 0.000 description 1
- 239000012634 fragment Substances 0.000 description 1
- 229930182830 galactose Natural products 0.000 description 1
- 239000008273 gelatin Substances 0.000 description 1
- 229920000159 gelatin Polymers 0.000 description 1
- 235000019322 gelatine Nutrition 0.000 description 1
- 235000011852 gelatine desserts Nutrition 0.000 description 1
- 238000012226 gene silencing method Methods 0.000 description 1
- 230000002068 genetic effect Effects 0.000 description 1
- 235000013922 glutamic acid Nutrition 0.000 description 1
- 239000004220 glutamic acid Substances 0.000 description 1
- ZDXPYRJPNDTMRX-UHFFFAOYSA-N glutamine Natural products OC(=O)C(N)CCC(N)=O ZDXPYRJPNDTMRX-UHFFFAOYSA-N 0.000 description 1
- 235000004554 glutamine Nutrition 0.000 description 1
- 235000011187 glycerol Nutrition 0.000 description 1
- 150000002334 glycols Chemical class 0.000 description 1
- 239000008187 granular material Substances 0.000 description 1
- 210000003494 hepatocyte Anatomy 0.000 description 1
- HNDVDQJCIGZPNO-UHFFFAOYSA-N histidine Natural products OC(=O)C(N)CC1=CN=CN1 HNDVDQJCIGZPNO-UHFFFAOYSA-N 0.000 description 1
- 229960001680 ibuprofen Drugs 0.000 description 1
- 230000028993 immune response Effects 0.000 description 1
- 238000000099 in vitro assay Methods 0.000 description 1
- 238000005462 in vivo assay Methods 0.000 description 1
- 230000004054 inflammatory process Effects 0.000 description 1
- 238000001802 infusion Methods 0.000 description 1
- 230000000266 injurious effect Effects 0.000 description 1
- 238000001361 intraarterial administration Methods 0.000 description 1
- 230000002427 irreversible effect Effects 0.000 description 1
- AGPKZVBTJJNPAG-UHFFFAOYSA-N isoleucine Natural products CCC(C)C(N)C(O)=O AGPKZVBTJJNPAG-UHFFFAOYSA-N 0.000 description 1
- 229960000310 isoleucine Drugs 0.000 description 1
- DKYWVDODHFEZIM-UHFFFAOYSA-N ketoprofen Chemical compound OC(=O)C(C)C1=CC=CC(C(=O)C=2C=CC=CC=2)=C1 DKYWVDODHFEZIM-UHFFFAOYSA-N 0.000 description 1
- 229960001691 leucovorin Drugs 0.000 description 1
- 238000004895 liquid chromatography mass spectrometry Methods 0.000 description 1
- 230000004807 localization Effects 0.000 description 1
- VTHJTEIRLNZDEV-UHFFFAOYSA-L magnesium dihydroxide Chemical compound [OH-].[OH-].[Mg+2] VTHJTEIRLNZDEV-UHFFFAOYSA-L 0.000 description 1
- 239000000347 magnesium hydroxide Substances 0.000 description 1
- 229910001862 magnesium hydroxide Inorganic materials 0.000 description 1
- 238000004519 manufacturing process Methods 0.000 description 1
- 238000001819 mass spectrum Methods 0.000 description 1
- 239000011159 matrix material Substances 0.000 description 1
- 230000001404 mediated effect Effects 0.000 description 1
- 229930182817 methionine Natural products 0.000 description 1
- XRTVFSTWEJOKNN-UHFFFAOYSA-N methyl 2-(4-fluoro-3-nitrophenyl)acetate Chemical compound COC(=O)CC1=CC=C(F)C([N+]([O-])=O)=C1 XRTVFSTWEJOKNN-UHFFFAOYSA-N 0.000 description 1
- 230000003278 mimic effect Effects 0.000 description 1
- 230000025608 mitochondrion localization Effects 0.000 description 1
- 239000000178 monomer Substances 0.000 description 1
- 210000000214 mouth Anatomy 0.000 description 1
- 210000003098 myoblast Anatomy 0.000 description 1
- WQEPLUUGTLDZJY-UHFFFAOYSA-N n-Pentadecanoic acid Natural products CCCCCCCCCCCCCCC(O)=O WQEPLUUGTLDZJY-UHFFFAOYSA-N 0.000 description 1
- 229950006780 n-acetylglucosamine Drugs 0.000 description 1
- 230000007935 neutral effect Effects 0.000 description 1
- 239000012457 nonaqueous media Substances 0.000 description 1
- 231100000252 nontoxic Toxicity 0.000 description 1
- 230000003000 nontoxic effect Effects 0.000 description 1
- 230000005937 nuclear translocation Effects 0.000 description 1
- 239000002674 ointment Substances 0.000 description 1
- 239000004006 olive oil Substances 0.000 description 1
- 235000008390 olive oil Nutrition 0.000 description 1
- 230000003647 oxidation Effects 0.000 description 1
- 238000007254 oxidation reaction Methods 0.000 description 1
- 230000036961 partial effect Effects 0.000 description 1
- 239000006072 paste Substances 0.000 description 1
- 239000000312 peanut oil Substances 0.000 description 1
- 150000002972 pentoses Chemical class 0.000 description 1
- 208000011906 peptic ulcer disease Diseases 0.000 description 1
- 230000002085 persistent effect Effects 0.000 description 1
- 230000003285 pharmacodynamic effect Effects 0.000 description 1
- COLNVLDHVKWLRT-UHFFFAOYSA-N phenylalanine Natural products OC(=O)C(N)CC1=CC=CC=C1 COLNVLDHVKWLRT-UHFFFAOYSA-N 0.000 description 1
- 229960002895 phenylbutazone Drugs 0.000 description 1
- VYMDGNCVAMGZFE-UHFFFAOYSA-N phenylbutazonum Chemical compound O=C1C(CCCC)C(=O)N(C=2C=CC=CC=2)N1C1=CC=CC=C1 VYMDGNCVAMGZFE-UHFFFAOYSA-N 0.000 description 1
- NBIIXXVUZAFLBC-UHFFFAOYSA-K phosphate Chemical compound [O-]P([O-])([O-])=O NBIIXXVUZAFLBC-UHFFFAOYSA-K 0.000 description 1
- 150000003904 phospholipids Chemical class 0.000 description 1
- 150000008300 phosphoramidites Chemical class 0.000 description 1
- 108010054442 polyalanine Proteins 0.000 description 1
- 229920002647 polyamide Polymers 0.000 description 1
- 229920000768 polyamine Polymers 0.000 description 1
- 229920000515 polycarbonate Polymers 0.000 description 1
- 239000004417 polycarbonate Substances 0.000 description 1
- 229920000728 polyester Polymers 0.000 description 1
- 229920000570 polyether Polymers 0.000 description 1
- 102000040430 polynucleotide Human genes 0.000 description 1
- 108091033319 polynucleotide Proteins 0.000 description 1
- 239000002157 polynucleotide Substances 0.000 description 1
- 229920005862 polyol Polymers 0.000 description 1
- 150000003077 polyols Chemical class 0.000 description 1
- 239000012286 potassium permanganate Substances 0.000 description 1
- 229920001592 potato starch Polymers 0.000 description 1
- 229960003101 pranoprofen Drugs 0.000 description 1
- 210000002442 prefrontal cortex Anatomy 0.000 description 1
- 230000002265 prevention Effects 0.000 description 1
- 230000008569 process Effects 0.000 description 1
- 230000001737 promoting effect Effects 0.000 description 1
- 238000000425 proton nuclear magnetic resonance spectrum Methods 0.000 description 1
- 239000000612 proton pump inhibitor Substances 0.000 description 1
- 230000002685 pulmonary effect Effects 0.000 description 1
- LISFMEBWQUVKPJ-UHFFFAOYSA-N quinolin-2-ol Chemical compound C1=CC=C2NC(=O)C=CC2=C1 LISFMEBWQUVKPJ-UHFFFAOYSA-N 0.000 description 1
- 239000002994 raw material Substances 0.000 description 1
- 230000008707 rearrangement Effects 0.000 description 1
- 108010054624 red fluorescent protein Proteins 0.000 description 1
- 238000010992 reflux Methods 0.000 description 1
- 230000029058 respiratory gaseous exchange Effects 0.000 description 1
- 230000004044 response Effects 0.000 description 1
- 230000002441 reversible effect Effects 0.000 description 1
- 125000000548 ribosyl group Chemical group C1([C@H](O)[C@H](O)[C@H](O1)CO)* 0.000 description 1
- 235000005713 safflower oil Nutrition 0.000 description 1
- 239000003813 safflower oil Substances 0.000 description 1
- 235000019515 salmon Nutrition 0.000 description 1
- 201000000980 schizophrenia Diseases 0.000 description 1
- 230000007017 scission Effects 0.000 description 1
- 235000004400 serine Nutrition 0.000 description 1
- 239000008159 sesame oil Substances 0.000 description 1
- 235000011803 sesame oil Nutrition 0.000 description 1
- 210000003491 skin Anatomy 0.000 description 1
- 235000019812 sodium carboxymethyl cellulose Nutrition 0.000 description 1
- 229920001027 sodium carboxymethylcellulose Polymers 0.000 description 1
- 238000010532 solid phase synthesis reaction Methods 0.000 description 1
- 239000003549 soybean oil Substances 0.000 description 1
- 235000012424 soybean oil Nutrition 0.000 description 1
- 239000007921 spray Substances 0.000 description 1
- 235000019698 starch Nutrition 0.000 description 1
- 239000007858 starting material Substances 0.000 description 1
- 239000008174 sterile solution Substances 0.000 description 1
- 238000006467 substitution reaction Methods 0.000 description 1
- 235000000346 sugar Nutrition 0.000 description 1
- 150000008163 sugars Chemical class 0.000 description 1
- 239000000829 suppository Substances 0.000 description 1
- 229960004492 suprofen Drugs 0.000 description 1
- 238000013268 sustained release Methods 0.000 description 1
- 239000012730 sustained-release form Substances 0.000 description 1
- 230000009747 swallowing Effects 0.000 description 1
- 230000009885 systemic effect Effects 0.000 description 1
- 239000000454 talc Substances 0.000 description 1
- 229910052623 talc Inorganic materials 0.000 description 1
- 238000012360 testing method Methods 0.000 description 1
- 238000002560 therapeutic procedure Methods 0.000 description 1
- 238000011285 therapeutic regimen Methods 0.000 description 1
- 150000003568 thioethers Chemical class 0.000 description 1
- 238000006177 thiolation reaction Methods 0.000 description 1
- 235000008521 threonine Nutrition 0.000 description 1
- 238000011200 topical administration Methods 0.000 description 1
- 230000000699 topical effect Effects 0.000 description 1
- 231100000419 toxicity Toxicity 0.000 description 1
- 230000001988 toxicity Effects 0.000 description 1
- 229940126672 traditional medicines Drugs 0.000 description 1
- 239000000196 tragacanth Substances 0.000 description 1
- 235000010487 tragacanth Nutrition 0.000 description 1
- 229940116362 tragacanth Drugs 0.000 description 1
- 238000013518 transcription Methods 0.000 description 1
- 230000035897 transcription Effects 0.000 description 1
- 238000013519 translation Methods 0.000 description 1
- GWBUNZLLLLDXMD-UHFFFAOYSA-H tricopper;dicarbonate;dihydroxide Chemical compound [OH-].[OH-].[Cu+2].[Cu+2].[Cu+2].[O-]C([O-])=O.[O-]C([O-])=O GWBUNZLLLLDXMD-UHFFFAOYSA-H 0.000 description 1
- 150000004043 trisaccharides Chemical class 0.000 description 1
- 239000010981 turquoise Substances 0.000 description 1
- 235000002374 tyrosine Nutrition 0.000 description 1
- OUYCCCASQSFEME-UHFFFAOYSA-N tyrosine Natural products OC(=O)C(N)CC1=CC=C(O)C=C1 OUYCCCASQSFEME-UHFFFAOYSA-N 0.000 description 1
- 239000004474 valine Substances 0.000 description 1
- 230000009385 viral infection Effects 0.000 description 1
- 230000003612 virological effect Effects 0.000 description 1
- 239000011782 vitamin Substances 0.000 description 1
- 235000013343 vitamin Nutrition 0.000 description 1
- 229940088594 vitamin Drugs 0.000 description 1
- 229930003231 vitamin Natural products 0.000 description 1
- 235000001892 vitamin D2 Nutrition 0.000 description 1
- 150000003722 vitamin derivatives Chemical class 0.000 description 1
- PJVWKTKQMONHTI-UHFFFAOYSA-N warfarin Chemical compound OC=1C2=CC=CC=C2OC(=O)C=1C(CC(=O)C)C1=CC=CC=C1 PJVWKTKQMONHTI-UHFFFAOYSA-N 0.000 description 1
- 229960005080 warfarin Drugs 0.000 description 1
- 239000001993 wax Substances 0.000 description 1
- 235000021241 α-lactalbumin Nutrition 0.000 description 1
Classifications
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P3/00—Drugs for disorders of the metabolism
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K47/00—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
- A61K47/50—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
- A61K47/51—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
- A61K47/54—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound
- A61K47/542—Carboxylic acids, e.g. a fatty acid or an amino acid
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K47/00—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
- A61K47/50—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
- A61K47/51—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
- A61K47/54—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound
- A61K47/545—Heterocyclic compounds
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K47/00—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
- A61K47/50—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
- A61K47/51—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
- A61K47/54—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound
- A61K47/549—Sugars, nucleosides, nucleotides or nucleic acids
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07D—HETEROCYCLIC COMPOUNDS
- C07D207/00—Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom
- C07D207/02—Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom
- C07D207/04—Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having no double bonds between ring members or between ring members and non-ring members
- C07D207/10—Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having no double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
- C07D207/12—Oxygen or sulfur atoms
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07D—HETEROCYCLIC COMPOUNDS
- C07D235/00—Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, condensed with other rings
- C07D235/02—Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, condensed with other rings condensed with carbocyclic rings or ring systems
- C07D235/04—Benzimidazoles; Hydrogenated benzimidazoles
- C07D235/06—Benzimidazoles; Hydrogenated benzimidazoles with only hydrogen atoms, hydrocarbon or substituted hydrocarbon radicals, directly attached in position 2
- C07D235/08—Radicals containing only hydrogen and carbon atoms
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07D—HETEROCYCLIC COMPOUNDS
- C07D235/00—Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, condensed with other rings
- C07D235/02—Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, condensed with other rings condensed with carbocyclic rings or ring systems
- C07D235/04—Benzimidazoles; Hydrogenated benzimidazoles
- C07D235/06—Benzimidazoles; Hydrogenated benzimidazoles with only hydrogen atoms, hydrocarbon or substituted hydrocarbon radicals, directly attached in position 2
- C07D235/12—Radicals substituted by oxygen atoms
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07D—HETEROCYCLIC COMPOUNDS
- C07D403/00—Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
- C07D403/02—Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
- C07D403/04—Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings directly linked by a ring-member-to-ring-member bond
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07D—HETEROCYCLIC COMPOUNDS
- C07D403/00—Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
- C07D403/14—Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing three or more hetero rings
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07H—SUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
- C07H15/00—Compounds containing hydrocarbon or substituted hydrocarbon radicals directly attached to hetero atoms of saccharide radicals
- C07H15/26—Acyclic or carbocyclic radicals, substituted by hetero rings
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07H—SUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
- C07H21/00—Compounds containing two or more mononucleotide units having separate phosphate or polyphosphate groups linked by saccharide radicals of nucleoside groups, e.g. nucleic acids
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N15/00—Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
- C12N15/09—Recombinant DNA-technology
- C12N15/11—DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
- C12N15/111—General methods applicable to biologically active non-coding nucleic acids
-
- A—HUMAN NECESSITIES
- A01—AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
- A01K—ANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
- A01K2207/00—Modified animals
- A01K2207/15—Humanized animals
-
- A—HUMAN NECESSITIES
- A01—AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
- A01K—ANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
- A01K2217/00—Genetically modified animals
- A01K2217/05—Animals comprising random inserted nucleic acids (transgenic)
-
- A—HUMAN NECESSITIES
- A01—AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
- A01K—ANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
- A01K2227/00—Animals characterised by species
- A01K2227/10—Mammal
- A01K2227/105—Murine
-
- A—HUMAN NECESSITIES
- A01—AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
- A01K—ANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
- A01K2267/00—Animals characterised by purpose
- A01K2267/03—Animal model, e.g. for test or diseases
- A01K2267/0306—Animal model for genetic diseases
- A01K2267/0318—Animal model for neurodegenerative disease, e.g. non- Alzheimer's
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2310/00—Structure or type of the nucleic acid
- C12N2310/10—Type of nucleic acid
- C12N2310/11—Antisense
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2310/00—Structure or type of the nucleic acid
- C12N2310/10—Type of nucleic acid
- C12N2310/14—Type of nucleic acid interfering N.A.
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2310/00—Structure or type of the nucleic acid
- C12N2310/30—Chemical structure
- C12N2310/31—Chemical structure of the backbone
- C12N2310/315—Phosphorothioates
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2310/00—Structure or type of the nucleic acid
- C12N2310/30—Chemical structure
- C12N2310/31—Chemical structure of the backbone
- C12N2310/318—Chemical structure of the backbone where the PO2 is completely replaced, e.g. MMI or formacetal
- C12N2310/3183—Diol linkers, e.g. glycols or propanediols
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2310/00—Structure or type of the nucleic acid
- C12N2310/30—Chemical structure
- C12N2310/33—Chemical structure of the base
- C12N2310/334—Modified C
- C12N2310/3341—5-Methylcytosine
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2310/00—Structure or type of the nucleic acid
- C12N2310/30—Chemical structure
- C12N2310/35—Nature of the modification
- C12N2310/351—Conjugate
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2310/00—Structure or type of the nucleic acid
- C12N2310/30—Chemical structure
- C12N2310/35—Nature of the modification
- C12N2310/351—Conjugate
- C12N2310/3515—Lipophilic moiety, e.g. cholesterol
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2320/00—Applications; Uses
- C12N2320/30—Special therapeutic applications
- C12N2320/32—Special delivery means, e.g. tissue-specific
Definitions
- the present disclosure relates to the technical field of genetic modulation, and in particular to unique compounds useful for enhancing delivery efficiency of oligonucleotides both in vitro and in vivo.
- Oligonucleotide-based therapeutics has been an emerging technology as its mechanisms of action provide expression regulation on almost unlimited target genes.
- the administration of oligonucleotides to patients, organs, tissues or cells may elicit or impact various biochemical reactions and thus achieve functions such as silencing, inhibiting, activating, and modulation of gene expression.
- the opportunity to use oligonucleotide-based therapy holds significant promise, providing solutions to medical problems that could not be addressed with traditional medicines.
- one of the major obstacles for the commercialization of oligonucleotide-based medicine is a lack of effective methods for delivering it to the right organ, tissue, or cell.
- Oligonucleotide therapeutics typically have a molecular weight ranging from 7 kDa to 14 kDa and comprise strong negative charge due to the inclusion of phosphodiester or phosphorothioate in the backbone thereof.
- the relatively large molecular weight and the high negative charge density restrain the transmission of oligonucleotides across cell membranes.
- Many strategies have been exploited for promoting the oligonucleotide delivery and yet, the delivery efficiency remains challenging.
- the present disclosure provides an oligonucleotide delivery enhancing compound (DEC) comprising a nitrogen-containing five membered heterocyclic ring moiety, the DEC can be directly or indirectly attached to an oligonucleotide so as to be used to enhance the delivery efficiency of oligonucleotides to a subject both in vitro and in vivo.
- the DEC has a structure represented by Formula AI or Formula AII
- the oligonucleotide delivery enhancing compound comprises a moiety represented by Formula BI
- the oligonucleotide delivery enhancing compound a structure represented by Formula BII
- an oligonucleotide delivery agent comprising a DEC moiety derived from the DEC of the present disclosure and at least one oligonucleotide, wherein the DEC moiety is directly or indirectly linked to the oligonucleotide via at least one linking moiety.
- the oligonucleotide can be antisense oligonucleotide (ASO) , antisense RNA, short interfering RNA (siRNA) , micro-RNA (miRNA) , small activating RNA (saRNA) , double-stranded RNA (dsRNA) , and small guide RNA (sgRNA) .
- composition comprising the DEC of the present disclosure
- the pharmaceutical composition may optionally comprise additional ingredients, such as pharmaceutically acceptable carrier, excipient, solvent, diluent, stabilizer, dispersant, buffer, compatibilizer, preservative agent and combinations thereof.
- Also provided herein is method of modulating the expression of a target gene in a subject in vitro or in vivo, wherein the method comprises the step of administrating the pharmaceutical composition of the present disclosure to a subject in the case of in vivo assays or therapeutic treatment, or contacting the pharmaceutical composition with a cell in the case of in vitro assays.
- FIG. 1 shows the knockdown activity of delivery enhancing compound conjugated siRNA (i.e., DEC-conjugated siRNA or DEC-siRNA) on mouse Factor VII (mFVII) mRNA expression in primary mouse hepatocytes (PMH) cells.
- PMH cells were transfected via lipofectamine TM RNAiMax with each of the indicated DEC-conjugated oligonucleotide (DCOs) (i.e., RD-12339, RD-12585, RD-12586, RD-12710, RD-12711 and RD-12712) at 0.1 nM and 1.0 nM for 24 hours.
- DCOs DEC-conjugated oligonucleotide
- dsCon2 served as a non-specific duplex control.
- mFVII mRNA levels were quantified by RT-qPCR using a gene specific primer set.
- Tbp was amplified as an internal reference. Shown are the mean values of mFVII mRNA in the cells relative to Mock treatment after normalizing to Tbp [mean ⁇ Standard Error of the Mean (SEM) of two replicated wells] .
- FIG. 2. shows the knockdown activity of DEC-conjugated siRNA on mouse FVII mRNA expression by free uptake in PMH cells.
- the indicated DCOs i.e., RD-12339, RD-12585, RD-12586, RD-12710, RD-12711 and RD-12712, as described in FIG. 1.
- DCOs were directly added to PMH cell culture media at 0.01, 0.05, 0.20, 0.78, 3.13, 12.50, 50 and 200 nM for 24 hours.
- Mock treatment was transfected in absence of oligonucleotide (not shown) .
- dsCon2 duplex served as a non-specific duplex control (not shown) .
- mFVII mRNA levels were quantified by RT-qPCR using a gene specific primer set. Tbp was amplified as an internal reference. Shown are the mean values of mFVII mRNA in the cells relative to Mock treatment after normalizing to Tbp (mean ⁇ SEM of two replicated wells) .
- FIG. 3. shows the in vivo knockdown activity of DEC-conjugated siRNAs on mouse FVII mRNA expression via subcutaneous injection (SC) injection in C57BL/6J mice.
- the indicated DCOs i.e., RD-12339, RD-12585, RD-12586, RD-12710, RD-12711 and RD-12712, as described in FIG. 1.
- PND postnatal day
- Saline was injected as a vehicle control to establish the baseline of mFVII mRNA expression.
- mice were sacrificed on 3 days post dosing and mFVII mRNA levels were quantified in total RNA isolated from liver tissue preps via RT-qPCR using a gene specific primer set. Tbp was amplified as an internal reference. Mean mFVII mRNA levels in liver tissue of each treatment group are shown relative to saline group after normalizing to Tbp (mean ⁇ SEM of 3 animals per group) .
- FIG. 4. shows the in vivo knockdown activity of DEC-conjugated siRNA on mouse FVII protein expression via SC injection in C57BL/6J mice.
- the indicated DCOs i.e., RD-12339, RD-12585 and RD-12586, as described in FIG. 1.
- Saline was injected as a vehicle control to establish the baseline of mFVII protein expression.
- Mice were sacrificed on day 3 following treatment and mFVII protein level was quantified in mice plasma by ELISA assay.
- FIG. 5. shows the duration of DEC-conjugated siRNA knockdown activity on mouse FVII protein expression via SC injection in C57BL/6J mice.
- the indicated DCOs i.e., RD-12710 and RD-12712
- RD-11706 was injected at 3 mg/kg and served as a control.
- Saline was injected as a vehicle control to establish the baseline of mFVII protein expression.
- Mouse plasmas were collected on day 10, 31, 54, 61, 80 and 89 following treatment and mFVII protein level was quantified in mouse plasma by ELISA assay.
- Mean mFVII protein levels in mouse plasma by detecting the OD values of 3 animals at each time point of per group are shown relative to saline group (mean ⁇ SEM of 3 animals at each time point of each group) .
- FIGs. 6A-6B show in vitro knockdown activity of DEC-siRNAs on mouse Sod1 (Sod1) mRNA expression by free uptake in PMH cells.
- DEC-siRNAs i.e., RD-13110, RD-13115 and RD-13118
- concentration gradient i.e. 1.56, 6.25, 25, 100, 400 and 1600 nM
- Sod1 mRNA levels were quantified via RT-qPCR using a gene specific primer set following RNA isolation and RT reactions. Tbp was amplified as an internal reference.
- Treatment with RD-12556 served as a non-conjugate control for comparison of dose dependent knockdown in absence DEC conjugation was shown in FIG. 6B (mean ⁇ SEM of two replicated wells) .
- FIG. 7 shows in vivo knockdown activity of different DEC-siRNAs on Sod1 mRNA expression via intracerebroventricular (ICV) injection in adult C57BL/6J mice.
- the indicated DEC-siRNAs i.e., RD-13110, RD-13115 and RD-13118
- ICV intracerebroventricular
- mice were sacrificed on 7 days post dosing and Sod1 mRNA knockdown was quantified in brain (i.e., brain-frontal cortex, cerebrum and brain-cerebellum) , spinal cord (i.e., cervical, thoracic and lumbar) and peripheral tissue (i.e., liver) via RT-qPCR using a gene specific primer set after RNA isolation and RT reaction.
- Tbp was amplified as an internal reference.
- Mean Sod1 mRNA levels are shown in each of the indicated tissues relative to mRNA levels in the saline group after normalizing to Tbp (mean ⁇ SEM of 2-3 animals per group) .
- FIG. 8 shows in vivo activity of different DEC-siRNAs on knocking down Sod1 mRNA expression following intravenous (IV) injection into the lateral tail vein of adult C57BL/6J mice.
- the indicated DEC-siRNAs i.e., RD-13110, RD-13115 and RD-13118
- Saline was injected as a vehicle control to establish the baseline.
- RD-12556 was injected at 20 mg/kg as a non-conjugate control.
- mice were sacrificed on day 7 following treatment and Sod1 mRNA knockdown was quantified in select peripheral tissues (i.e., heart, liver, spleen, lung, kidney and bladder) via RT-qPCR using a gene specific primer set after RNA isolation and RT reaction.
- Tbp was amplified as an internal reference. Mean Sod1 mRNA levels are shown in each of the indicated tissues relative to mRNA levels in the saline group after normalizing to Tbp (mean ⁇ SEM of 3 animals per group) .
- FIG. 9 shows in vivo knockdown activity of different DEC-siRNAs on Sod1 mRNA expression following IV injection into adult C57BL/6J mice.
- the indicated DEC-siRNAs i.e., RD-13110, RD-13115 and RD-13118
- Saline was injected as a vehicle control to establish the baseline.
- RD-12556 was injected at 20 mg/kg as a non-conjugate control.
- Sod1 mRNA levels were quantified in skeletal muscle tissue from different locations (i.e., bicep, semitendinosus, platysma and gluteus) via RT-qPCR using a gene specific primer set after RNA isolation and RT reaction.
- Tbp was amplified as an internal reference for RNA loading.
- Mean Sod1 mRNA levels in skeletal muscle tissue of 3 animals per group are shown relative to mRNA levels in the saline group after normalizing to Tbp (mean ⁇ SEM of 3 animals per group) .
- FIG. 10 shows knockdown activity of different DEC-siRNAs on rat Sod1 mRNA expression via intravitreal (IVT) injection in adult SD rats.
- the indicated DEC-siRNAs i.e., RD-13115 and RD-13118
- IVT injection 30 ⁇ g per left eye.
- the right eye of each rat served as a non-injected naive eye.
- Saline was injected as a vehicle control to establish the baseline.
- RD-12556 was injected as a non-conjugate control.
- Rats were sacrificed on day 14 following treatment and Sod1 mRNA levels were quantified in eyes via RT-qPCR using a rat gene specific primer set after RNA isolation and RT reaction.
- Geometric mean of the mRNA levels of Hprt1 and Hmbs were used as an internal reference for RNA loading.
- Mean Sod1 mRNA levels in retina tissue of 3 animalsfor each group are shown relative to mRNA levels in the saline group after normalizing to Hprt1 and Hmbs.
- the data represents mean ⁇ SEM of 3 mice per group.
- FIG. 11 shows the saRNA concentration of DEC-saRNA in adult C57BL/6J mice bladder after intravesical bladder (IVB) instillation.
- the indicated DEC-saRNA i.e., RD-13520
- RD-10773 was injected as a non-conjugate saRNA control (i.e., in absence of DEC) .
- mice were sacrificed on 2-, 6-, 12-hour, day-1 and day-4 following treatment and the concentrations of RD-13520 and RD-10773 were quantified in bladder tissue via stem-loop RT-qPCR using a gene specific primer set (R1- 40-AS-SL-RT, R1-40-AS-SL-F1 and SL-RT-qPCR-Uni-R2, see Table 13) .
- FIGs. 12A-12C show body weight change of and Sod1 knockdown activity of DEC-siRNAs via IV injection in adult C57BL/6J mice.
- the indicated siRNAs i.e., RD-15135, RD-15136, RD-15137 and RD-15138
- RD-15135 served as a non-conjugate control.
- RD-15136 was a lipid (i.e., C16) conjugated siRNA.
- RD-15137 with a typical DEC structure i.e., C5X5 served as a typical DEC-siRNA.
- FIG. 12A shows the body weight change of C57BL/6J mice out to day 28 post treatment.
- FIG. 12B shows the remaining mouse Sod1 mRNA on 14 days post dosing as quantified in tissues from periphery (i.e., heart, liver, kidney, fat tissues, pancreas, diaphragm) , blood vessel (i.e., thoracic aorta with vein) and skeletal muscle (i.e., bicep, semitendinosus, platysma and gluteus) via RT-qPCR using a gene specific primer set.
- periphery i.e., heart, liver, kidney, fat tissues, pancreas, diaphragm
- blood vessel i.e., thoracic aorta with vein
- skeletal muscle i.e., bicep, semitendinosus, platysma and gluteus
- FIG. 12C shows the remaining mouse Sod1 mRNA on 28 days post dosing as quantified in tissues from periphery (i.e., heart, liver, kidney, fat tissues, pancreas, diaphragm) , blood vessel (i.e., thoracic aorta with vein) and skeletal muscle (i.e., bicep, semitendinosus, platysma and gluteus) via RT-qPCR using a gene specific primer set.
- Rpl13a was amplified as an internal reference.
- Mean Sod1 mRNA levels in the selected mouse tissues are shown relative saline control after normalized to Rpl13a. The data represents mean ⁇ SEM of 3 mice per group.
- FIGs. 13A-13C show body weight change of and Sod1 knockdown activity of DEC-siRNAs via intrathecal (IT) injection in adult SD female rats.
- the indicated siRNAs i.e., RD-15135, RD-15136, RD-15137 and RD-15138
- RD-15136 was administered into C57BL/6J mice at 0.9 mg.RD-15135 served as a non-conjugate control.
- RD-15136 was a lipid (i.e., C16) conjugated siRNA.
- RD-15137 with a typical DEC structure i.e., C5X5 served as a typical DEC-siRNA.
- aCSF cerebrospinal fluid
- FIG. 13B shows the remaining rat Sod1 mRNA on 14 days post dosing as quantified in tissues from brain (i.e., frontal cortex, cerebellum and cerebrum) , spinal cord (i.e., cervical, thoracic and lumbar) and periphery (i.e., liver and kidney) via RT-qPCR using a gene specific primer set.
- FIG. 13C shows the remaining rat Sod1 mRNA on 28 days post dosing as quantifiedin tissues from brain (i.e., frontal cortex, cerebellum and brainstem) , spinal cord (i.e., cervical, thoracic and lumbar) and periphery (i.e., liver and kidney) .
- FIGs. 14A-14B show the in vitro knockdown activity of DEC-siRNA with accessory oligonucleotide (i.e., DEC-siRNA-ACO) on human SOD1 (i.e., SOD1) mRNA expression in SK-N-AS and T98G cells.
- the indicated DEC-siRNA-ACOs i.e., RD-16149, RD-16099, RD-16100, RD-16101, RD-16150, RD-16103, RD-16104 and RD-16105
- RD-16106 was transfected and served as a siRNA-ACO control. Mock treatments were transfected in absence of oligonucleotide (not shown) . SOD1 mRNA levels were quantified via RT-qPCR using a gene specific primer set following RNA isolation and RT reactions. B2M was amplified as an internal reference. Shown are the mean values of remaining SOD1 mRNA in the cells relative to Mock treatment after normalizing to B2M (mean ⁇ SEM of two replicated wells) .
- FIGs. 15A-15B show SOD1 knockdown activity and tissue concentration of siRNAs via ICV injection in adult hSOD1 G93A mice.
- the indicated siRNAs i.e., RD-14851, RD-12500, RD-16145 and RD-16334.
- RD-14851, RD-12500 and RD-16145 were administered as siRNA-ACOs.
- RD-16334 with a DEC structure i.e., L17
- Treatment with aCSF alone was used as a vehicle control to establish baseline expression.
- FIG. 15A shows the remaining SOD1 mRNA on 30 days post dosing as quantified in tissues from brain (i.e., frontal cortex, cerebellum and cerebrum) , spinal cord and periphery (i.e., liver) via RT-qPCR using a gene specific primer set.
- Rpl13a was amplified as an internal reference.
- Mean SOD1 mRNA levels in the selected mouse tissues are shown relative to mRNA levels in the aCSF group after normalized to Rpl13a.
- 15B shows the concentration of the antisense strand of each siRNA in tissues from brain (i.e., frontal cortex, cerebellum and cerebrum) , spinal cord and periphery (i.e., liver) via stem-loop RT-qPCR using a gene specific primer set (R17-02-AS-SL-RT, R17-02-AS-SL-F3 and SL-RT-qPCR-Uni-R1, see Table 13) 30 days posting dosing.
- FIGs. 16A-16B show splicing activity of “saRNA-ASO” DEC conjugated dual acting oligonucleotides (DEC-DAO) structure at converting SMN2 pre-mRNA to SMN2FL over the SMN2 ⁇ 7 mRNA isoform in GM03813 cells.
- DEC-DAO dual acting oligonucleotides
- DEC-DAO linkage with L21 linker i.e., RD-16939 and RD-16940
- DEC-saSMN2 i.e., RD-1642
- DEC-antisense oligonucleotide i.e., RD-14644
- FIGs. 16A-16B show the mRNA levels of SMN2FL and SMN2 ⁇ 7 as quantified by RT-qPCR using a gene specific primer set in each of the PCR reactions.
- SDHA was amplified as an internal reference used to normalize expression data. Data represents mean expression levels of SMN2FL or SMN2 ⁇ 7 relative to Mock treatment after normalized to SDHA (mean ⁇ SEM of two replicated transfection wells) .
- FIGs. 17A-17C show the effect of “saRNA-siRNA” DEC-DAO structure targeting two different human genes (SMN2 and SOD1) on the expression of SMN2 (SMN2FL and SMN2 ⁇ 7) and SOD1 in GM03813 cells.
- the indicated DEC-DAO linkage with L21 linker (i.e., RD-16941) , DEC-saSMN2 (i.e., RD-16424) and DEC-siSOD1 (i.e., RD-13115) were transfected into GM03813 cells at indicated concentrations (i.e., 0.1, 1 and 10 nM) for 3 days.
- FIGs. 17A-17B show the mRNA levels of SMN2FL and SMN2 ⁇ 7 as quantified by RT-qPCR using a gene specific primer set in each of the PCR reactions.
- 17C shows remaining SOD1 mRNA levels as quantified by RT-qPCR using a gene specific primer set in each of the PCR reactions.
- SDHA was amplified as an internal reference used to normalize expression data. Data represents mean expression levels of SMN2FL, SMN2 ⁇ 7 or SOD1 relative to Mock treatment after normalized to SDHA (mean ⁇ SEM of two replicated transfection wells) .
- FIGs. 18A-18B show knockdown activity of “siRNA-siRNA” DEC-DAO structure targeting two different mouse genes (Sod1 and Ppig) on the expression of mouse Sod1 and mouse Ppig in mouse myoblast cell line (C2C12) .
- the indicated DEC-DAO linkage with L21 linker (i.e., RD-16942) , DEC-siSod1 (i.e., RD-13115) and DEC-siPpig (i.e., RD-14672) were transfected into C2C12 cells at indicated concentrations (i.e., 0.01, 0.1 and 1 nM) for 24 hours. Mock treatments were transfected in the absence of an oligonucleotide.
- RD-16381 was transfected as a non-specific DEC control.
- Combo treatment i.e., RD-13115+RD-14672
- combo treatment control i.e., RD-16381+RD-13115, RD-16381+RD-14672 were also transfected into C2C12 cells at indicated concentrations (i.e., 0.01, 0.1 and 1 nM) for 24 hours.
- FIG. 18A shows remaining mouse Sod1 mRNA levels as quantified by RT-qPCR using a gene specific primer set in each of the PCR reactions.
- FIG. 18B shows the remaining mouse Ppig mRNA levels as quantified by RT-qPCR using a gene specific primer set in each of the PCR reactions.
- Mouse Rpl13a was amplified as an internal reference used to normalize expression data. Data represents mean expression levels of Sod1 and Ppig relative to Mock treatment after normalized to Rpl13a (mean ⁇ SEM of two replicated transfection wells) .
- FIG. 19 shows knockdown activity of “siRNA-siRNA” DEC-DAO structure targeting two different mouse genes (Sod1 and Ppig) on the expression of mouse Sod1 and mouse Ppig in adult C57BL/6J mice.
- the indicated DEC-DAO linkage with L21 linker (i.e., RD-16942) , DEC-siSod1 (i.e., RD-13115) and DEC-siPpig (i.e., RD-14672) were administered into C57BL/6J mice via IV injection at 10 mg/kg.
- Combo treatment i.e., RD-13115+RD-14672 was administered via IV injection at 10 mg/kg.
- Treatment with saline alone was used as a vehicle control to establish baseline expression.
- Remaining mouse Sod1 mRNA on 14 days post dosing were quantified in tissues from periphery (i.e., heart, liver, kidney, fat tissues, diaphragm) , blood vessel (i.e., thoracic aorta with vein) and skeletal muscle (i.e., bicep, semitendinosus, platysma and gluteus) via RT-qPCR using a gene specific primer set.
- Tbp was amplified as an internal reference used to normalize expression data.
- Mean Sod1 mRNA levels in the selected mouse tissues are shown relative to mRNA levels in the saline group after normalized to Tbp. The data represents mean ⁇ SEM of 3-4 mice per group.
- FIGs. 20A-20B compares the oligonucleotide concentration of DEC-saRNA to non- conjugated saRNA in retina and vitreous humor of adult SD rat after intravitreal (IVT) injection.
- the indicated DEC-saRNA (i.e., RD-16447) and non-conjugated saRNA (i.e., RD-12173) were administered via IVT injection at 0.03 mg.
- the rats were sacrificed on 1-hour, day-1, -3, -7, -14 and -28 following treatment and the concentrations of RD-16447 and RD-12173 were quantified in retina and vitreous humor via stem-loop RT-qPCR using a gene specific primer set of (R1C-0M4-AS-SL-RT, R1C-0M4-AS-SL-F4 and SL-RT-qPCR-Uni-R1, see Table 13) .
- Mean values of oligonucleotide concentrations of each group are shown (mean ⁇ SEM of 3 animals per group) .
- FIG. 21 shows the in vitro knockdown activity of DEC-siRNA on SOD1 mRNA expression in T98G cells.
- the indicated DEC-siRNA i.e., RD-17138
- RD-11566 was transfected and served as a non-specific duplex control.
- Mock treatments were transfected in absence of oligonucleotide.
- SOD1 mRNA levels were quantified via RT-qPCR using a gene specific primer set following RNA isolation and RT reactions.
- B2M was amplified as an internal reference used to normalize expression data. Shown are the mean values of remaining SOD1 mRNA in the cells relative to Mock treatment after normalizing to B2M (mean ⁇ SEM of four replicated wells) .
- the newly developed oligonucleotide delivery agent enables more efficient delivery of desired amount or higher level of the oligonucleotides, as compared to the oligonucleotide delivery technologies of the prior art, at the above indicated target tissues, and thus may improve bioactive and pharmacological properties (e.g., biodistribution, bioavailability, pharmacokinetics, activity, potency, etc. ) .
- bioactive and pharmacological properties e.g., biodistribution, bioavailability, pharmacokinetics, activity, potency, etc.
- the improvement of bioactive properties may cause improved cell uptake, higher potency, and longer duration/half-life.
- the improvement of pharmacological properties may also lead to lower toxicity, lower dose, less frequent administrations, and less undesired immune responses.
- the present oligonucleotide delivery agent involves a simpler synthesis process and thus has better processibility in manufacturing.
- the present oligonucleotide delivery agent possesses inherent pharmacological properties of a free benzimidazole or a benzimidazole derivative which targets proton pump (as reversible and irreversible proton pump inhibitors, PPI) , dopamine receptor (DRD) , angiotensin II type 1 receptor (AT1) , histamine receptor (HRH) , dual specific mitogen activated protein kinase (MEK) and/or cyclin dependent kinase (CDK) , etc.
- the inherent pharmacological properties enable the present oligonucleotide delivery agent to be potentially used in drugs including peptic ulcer, hypertension, schizophrenia, parasitic infection, bacterial infection, virus infection, and cancer, etc.
- the newly developed oligonucleotide delivery agent disclosed herein can facilitate delivery of an effective amount of oligonucleotides to achieve and function at target tissues or cells, e.g., in central nervous system (e.g., brain and spinal cord) , liver, lung, kidney, intestine, pancreas, cholecyst, heart, lymph nodes, spleen, stomach, bladder, muscle and bone.
- the oligonucleotide delivery agent s modulating activities, e.g., up-regulating or down-regulating, the expression of a target gene in the cells can be improved as compared to the technologies of the prior art.
- oligonucleotide delivery agent as described herein may facilitate improved bio-distribution of therapeutic oligonucleotides in various target (s) or cells as stated above for preventing, treating and/or delaying on-set of various diseases, disorders and/or conditions.
- any numerical ranges include any numerical values in the range (including the upper and lower values) , and also any sub-ranges in the range.
- the range from 1 to 3 may include any of the numerical values 1, 2 and 3, as well as sub-ranges from 1 to 2 and from 2 to 3.
- oligonucleotide refers to polymers of nucleotides, and includes, but is not limited to, single-stranded, double-stranded or partial double-stranded nucleic acid molecules of DNA, RNA, or DNA/RNA hybrid, oligonucleotide strands containing regularly and irregularly alternating deoxyribosyl portions and ribosyl portions, as well as modified and naturally or unnaturally existing frameworks for such oligonucleotides.
- the oligonucleotide can be selected from the group consisting of antisense oligonucleotide (ASO) , antisense RNA, short interfering RNA (siRNA) , micro-RNA (miRNA) , small activating RNA (saRNA) , dsRNA, and small guide RNA (sgRNA) .
- ASO antisense oligonucleotide
- siRNA short interfering RNA
- miRNA micro-RNA
- saRNA small activating RNA
- dsRNA small guide RNA
- oligonucleotide strand , “strand” and “oligonucleotide sequence” as used herein can be used interchangeably and refer to a generic term for short nucleotide sequences having less than 50 bases, such as less than 45, less than 40, less than 35 bases, such as 2-50 bases, or 5-45 bases, or 10-40 bases, or 15-35 bases, or 20-30 bases (including nucleotides in deoxyribonucleic acid (DNA) or ribonucleic acid (RNA) ) .
- DNA deoxyribonucleic acid
- RNA ribonucleic acid
- the length of a strand can be any length from 5 to 50 nucleotides, 10 to 40 nucleotides, 15 to 35 nucleotides, 18 to 30 nucleotides or 20 to 25 nucleotides.
- target gene can refer to nucleic acid sequences, transgenes, viral or bacterial sequences, chromosomes or extrachromosomal genes that are naturally present in organisms, and/or can be transiently or stably transfected or incorporated into cells and/or chromatins thereof.
- the target gene can be a protein-coding gene or a non-protein-coding gene such as a microRNA gene and a long non-coding RNA gene.
- one target gene is SOD1.
- target sequence is a sequence fragment to which the sense strand or antisense oligonucleotide of the siRNA or saRNA is homologous or complementary.
- a SOD1 siRNA is homologous or complementary to a target select sequence within human SOD1 transcript.
- the term “guide strand” or “G strand” refers to a strand in a small RNA duplex that assembles with the Argonaute (AGO) protein.
- the other strand partially or completely complementary to the guide strand is called “passenger strand” or “P strand” .
- the strand carrying the complementary sequence to the target is the antisense strand and, if properly designed, will be preferentially chosen to be the guide strand.
- the passenger strand is the sense strand.
- the sense strand can be selected as the guide strand, resultant in an antisense passenger strand.
- LNA refers to a locked nucleic acid in which the 2’ -oxygen and 4’ -carbon atoms are joined by an extra bridge.
- BNA refers to a 2′-O and 4′-aminoethylene bridged nucleic acid that can contain a five-membered or six-membered bridged structure with an N-O linkage.
- PNA refers to a nucleic acid mimic with a pseudopeptide backbone composed of N- (2-aminoethyl) glycine units with the nucleobases attached to the glycine nitrogen via carbonyl methylene linkers.
- antisense oligonucleotide refers to a single strand oligonucleotide or the like having, comprising, or consisting of a sequence of bases or the like which allow the oligonucleotide or the like to hybridize to a target molecule, such as another nucleic acid, modified nucleic acid or nucleic acid analog, e.g., by base-pairing, such as Watson-Crick base-pairing or non-Watson-Crick base pairing.
- a target molecule such as another nucleic acid, modified nucleic acid or nucleic acid analog, e.g., by base-pairing, such as Watson-Crick base-pairing or non-Watson-Crick base pairing.
- an antisense oligonucleotide is fully complementary or nearly fully complementary to the target molecule.
- any oligonucleotide of any type described herein or known in the art can be used as an antisense oligonucleotide.
- an antisense oligonucleotide can perform or participate in any of various biological functions, including RNA interference, RNase H-mediated cleavage, exon skipping, the prevention of exon skipping, the enhancement or blocking of an agent (e.g., a protein, RNA, protein-RNA complex, or any other molecule) from binding to another nucleic acid, or any other biological function performed by an antisense oligonucleotide, as described herein or known in the art.
- an agent e.g., a protein, RNA, protein-RNA complex, or any other molecule
- small interfering RNA can be used interchangeably and refer to a ribonucleic acid molecule that can down-regulate, knockdown, or silence target gene expression. It can be a double-stranded nucleic acid molecule. It interferes with the expression of specific genes with complementary nucleotide sequences by degrading mRNA after transcription, preventing translation. siRNA binds to target mRNA mainly in the cytoplasm to down-regulate gene expression post-transcriptionally via the RNA interference (RNAi) mechanism.
- RNAi RNA interference
- siRNAs may be designed to target a gene’s mRNA sequence to silence its expression via the RNAi mechanism, such as SOD1, for maximizing treatment outcomes, e.g., for ALS patients.
- siRNAs are molecules having endogenous RNA bases or chemically modified nucleotides. The modifications do not abolish cellular activity, but rather impart increased stability and/or increased cellular potency. Examples of chemical modifications include phosphorothioate groups, 2′-deoxynucleotide, 2′-OCH 3 -containing ribonucleotides, 2′-F-ribonucleotides, 2′-methoxyethyl ribonucleotides, combinations thereof and the like.
- the siRNA can have varying lengths (e.g., 10-200 bps) and structures (e.g., hairpins, single/double/partial-double strands, bulges, nicks/gaps, mismatches) and are processed in cells to provide active gene silencing.
- a double-stranded siRNA can have the same number of nucleotides on each strand (blunt ends) or asymmetric ends (overhangs) .
- An overhang of 1-2 nucleotides, for example, can be present on the sense and/or the antisense strand, as well as present on the 5′-and/or the 3′-ends of a given strand.
- the length of the siRNA molecule is typically about 10 to about 60, about 10 to about 50, about 15 to about 30, about 17 to about 29, about 18 to about 28, about 19 to about 27, about 20 to about 26, about 21 to about 25, and about 22 to about 24 base pairs, and typically about 15, about 16, about 17, about 18, about 19, about 20, about 21, about 23, about 25, about 30, about 40, or about 50 base pairs.
- the terms “small interfering RNA” , “silencing RNA” and “siRNA” also contain nucleic acids other than the ribonucleotide, including, but not limited to, modified nucleotides or analogues.
- small activating RNA As used herein, the terms “small activating RNA” , “saRNA” and “small activating ribonucleic acid” can be used interchangeably and refer to a ribonucleic acid molecule that can up-regulate target gene expression. It can be a double-stranded nucleic acid molecule composed of a first nucleic acid strand containing a ribonucleotide sequence with sequence homology with the non-coding nucleic acid sequence (such as a promoter and an enhancer) of a target gene and a second nucleic acid strand containing a nucleotide sequence complementary with the first strand.
- a ribonucleic acid molecule that can up-regulate target gene expression. It can be a double-stranded nucleic acid molecule composed of a first nucleic acid strand containing a ribonucleotide sequence with sequence homology with the non-coding nucleic acid sequence (such as a promoter and an
- the saRNA can also be comprised of a synthesized or vector-expressed single-stranded RNA molecule that prone to form a hairpin structure by two complementary regions within the molecule, wherein the first region contains a ribonucleotide sequence having sequence homology with the target sequence of a promoter of a gene, and a ribonucleotide sequence contained in the second region is complementary with the first region.
- the length of the duplex region of the saRNA molecule is typically about 10 to about 60, about 10 to about 50, about 10 to about 40, about 12 to about 30, about 14 to about 28, about 16 to about 26, about 18 to about 24, and about 20 to about 22 base pairs, and typically about 10, about 13, about 15, about 17, about 18, about 19, about 20, about 21, about 22, about 25, about 30, about 40, about 50, or about 60 base pairs.
- the terms “small activating RNA” , “saRNA” and “small activating ribonucleic acid” also contain nucleic acids other than the ribonucleotide, including, but not limited to, modified nucleotides or analogues.
- ODV oligonucleotide delivery vehicle
- oligonucleotide delivery vehicle refers to an oligonucleotide molecule comprising a duplex or double-stranded RNA (e.g., siRNA or saRNA) and an accessory oligonucleotide (ACO) which is covalently linked to the duplex RNA via a linker.
- RNA e.g., siRNA or saRNA
- ACO accessory oligonucleotide
- delivery enhancing compound and “DEC” are used interchangeably and refer to the compound of the present disclosure.
- DEC-conjugated oligonucleotide and “DCO” can be used interchangeably and refer to a combination in which at least one moiety derived from the DEC has been attached, e.g. covalently, to at least one oligonucleotide. Additionally, the DCO may further comprising at least one targeting moiety which helps the oligonucleotide in targeting to, accumulating in, or accessing to target site in a cell.
- administration refers to ordinary enteral administration, topical administration, and especially, injection administration, and may include, without limitation, intravenous, intramuscular, intraarterial, intrathecal, intracapsular, intraorbital, intracardiac, intradermal, intraperitoneal, transtracheal, subcutaneous, subcuticular, intraarticular, subcapsular, subarachnoid, intraspinal, and intrasternal injection and infusion.
- the term “pharmaceutical composition” refers to an active agent, optionally formulated together with one or more pharmaceutically acceptable carriers and other additives.
- active agent is present in unit dose amount appropriate for administration in a therapeutic regimen that shows a statistically significant probability of achieving a predetermined therapeutic effect when administered to a relevant population.
- compositions may be specially formulated for administration in solid or liquid form, including those adapted for the following: oral 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; parenteral administration, for example, by subcutaneous, intramuscular, intravenous or epidural injection as, for example, a sterile solution or suspension, or sustained-release formulation; topical application, for example, as a cream, ointment, or a controlled-release patch or spray applied to the skin, lungs, or oral cavity; intravaginally or intrarectally, for example, as a pessary, cream, or foam; sublingually; ocularly; transdermally; or nasally, pulmonary, and to other mucosal surfaces.
- oral administration for example, drenches (aqueous or non-aqueous solutions
- the term “pharmaceutically acceptable carrier” means a pharmaceutically acceptable material, composition or vehicle, such as a liquid or solid filler, diluent, excipient, or solvent encapsulating material, involved in carrying or transporting the subject compound from one organ, or portion of the body, to another organ, or portion of the body.
- a pharmaceutically acceptable material, composition or vehicle such as a liquid or solid filler, diluent, excipient, or solvent encapsulating material, involved in carrying or transporting the subject compound from one organ, or portion of the body, to another organ, or portion of the body.
- Each carrier must be “acceptable” in the sense of being compatible with the other ingredients of the formulation and not injurious to the patient.
- materials which can serve as pharmaceutically-acceptable carriers include: sugars, such as lactose, glucose and sucrose; starches, such as corn starch and potato starch; cellulose, and its derivatives, such as sodium carboxymethyl cellulose, ethyl cellulose and cellulose acetate; powdered tragacanth; malt; gelatin; talc; excipients, such as cocoa butter and suppository waxes; oils, such as peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, corn oil and soybean oil; glycols, such as propylene glycol; polyols, such as glycerin, sorbitol, mannitol and polyethylene glycol; esters, such as ethyl oleate and ethyl laurate; agar; buffering agents, such as magnesium hydroxide and aluminum hydroxide; alginic acid; pyrogen-free water; isotonic saline; Ring
- the term “subject” , “test subject” and related terms, as used herein, refer to any organism to which a provided compound or composition is administered in accordance with the present invention e.g., for experimental, diagnostic, prophylactic, and/or therapeutic purposes. Typical subjects include animals (e.g., mammals such as mice, rats, rabbits, non-human primates, and humans; fishes; birds; insects; worms; etc. ) and plants. In some embodiments, a subject may be suffering from, and/or susceptible to a disease, disorder, and/or condition. In some embodiments, a subject is a human being or other mammal. In some embodiments, a subject can be male or female.
- the animal is a vertebrate such as a primate, rodent, domestic animal or game animal.
- primates include chimpanzees, cynomolgus monkeys, spider monkeys, and macaques, e.g., Rhesus.
- Rodents include mice, rats, woodchucks, ferrets, rabbits and hamsters.
- domestic and game animals include cows, horses, pigs, deer, bison, buffalo, feline species, e.g., domestic cat, canine species, e.g., dog, fox, wolf, avian species, e.g., chicken, emu, ostrich, and fish, e.g., trout, catfish and salmon.
- the subject is a mammal, e.g., a primate, e.g., a human.
- the mammal can be a human, non-human primate, mouse, rat, dog, cat, horse, or cow, but are not limited to these examples.
- a mammal other than a human can be advantageously used as subjects that represent animal models of disorders associated with autoimmune disease or inflammation.
- a method and composition described herein can be used to treat domesticated animals and/or pets.
- a “therapeutically effective amount” of a composition is an amount sufficient to achieve a desired therapeutic effect, and therefore does not require cure or complete remission.
- therapeutic efficacy is an improvement in any of the disease indicators, and a therapeutically effective amount is sufficient to cause an improvement in a clinically significant condition/symptom in the treated individual.
- the phrases “therapeutically effective amount” and “effective amount” are used herein to mean an amount sufficient to reduce by at least about 15 percent, preferably by at least 50 percent, more preferably by at least 90 percent, or to increase at least about 50 percent, at least about 100 percent, at least about 200 percent, more preferable at least about 500 percent and can prevent a clinically significant deficit in the activity, function and response of the individual being treated.
- the effective amount may vary depending on such factors as the size and weight of the subject, the type of illness, or the particular agents of the application. For example, the choice of the agent of the application could affect what constitutes an “effective amount” .
- One of ordinary skill in the art would be able to study the factors contained herein and make the determination regarding the effective amount of the agents of the application without undue experimentation.
- the regime of administration may affect what constitutes an effective amount.
- the agent of the application can be administered to the subject either prior to or after the disease diagnosis or condition. Further, several divided dosages, as well as staggered dosages, can be administered daily or sequentially, or the dose can be continuously infused, or can be a bolus injection. Further, the dosages of the agent (s) of the application could be proportionally increased or decreased as indicated by the exigencies of the therapeutic or prophylactic situation.
- treat, ” “treated, ” “treating” , or “treatment” as used herein have the meanings commonly understood in the medical arts, and therefore do not require cure or complete remission, and include any beneficial or desired clinical results.
- beneficial or desired clinical results are prolonging survival as compared to expected survival without treatment, reduced symptoms including one or more of the followings: weakness and atrophy of proximal skeletal muscles, inability to sit or walk independently, difficulties in swallowing, breathing, etc.
- preventing or “delaying” a disease refers to inhibiting the full development of a disease.
- the present disclosure is based on an unexpected discovery that a compound having a specific structure comprising a nitrogen-containing five membered heterocyclic ring moiety may be directly or indirectly bonded to at least one oligonucleotide so as to aid the delivery of the oligonucleotide to a subject and thus achieving improved modulation of a target gene both in vitro and/or in vivo.
- the compound as disclosed herein may remarkably enhance oligonucleotide delivery efficiency to liver, combine endosomal escape and nuclear translocation design, without the need of a delivery vector or formulation.
- the nitrogen-containing five membered heterocyclic ring moiety may have a core structure shown by any of the following formulae, with all the chemical bonds, atoms and substituents attached to the ring atoms of the core structure omitted for the sake of simplicity:
- the above indicated core structures comprise different ring atoms, and the fused ring core structures may be either electrically neutral or locally charged in the form of e.g., sulphonium or quaternary ammonium. It can be seen that one or more unsaturated double bonds may shift in the fused ring core structure due to the delocalized ⁇ -electronic conjugation effect, and all the substituents, including the presence/absence of any substituents, may vary depending on the categories and valences of each ring atom to which the substituents are attached. For example, an additional substituent may be attached to a sulphonium or quaternary ammonium ion which is present as a ring atom of the fused ring core structure. All of these variations are within the concept of the present disclosure.
- the compound of the present disclosure comprises structure of Formula BI, wherein X′ is selected from the group consisting of carbon, nitrogen, oxygen and sulfur; each of F′, G′, H′ and I′ is independently selected from the group consisting of carbon, nitrogen, oxygen and sulfur; and each of the asterisks refers to a site optionally linked to at least one substituent or an oligonucleotide directly or indirectly.
- the compound of the present disclosure has a structure represented by Formula AI or Formula AII:
- X on each occurrence, is an atom selected from the group consisting of carbon, nitrogen, oxygen and sulfur
- each of F, G, H and I is independently selected from the group consisting of carbon, nitrogen, oxygen and sulfur.
- n is an integer of 1, 2 or 3
- m+n 4.
- C on each occurrence, is either absent or selected from the group consisting of hydrogen; halogen atom, such as fluorine, chlorine, bromine or iodine; hydroxyl; (C 1 -C 25 ) alkyl, such as - (C 2 -C 22 ) alkyl, or - (C 2 -C 19 ) alkyl, or - (C 3 -C 18 ) alkyl, or - (C 4 -C 16 ) alkyl, or - (C 6 -C 12 ) alkyl, or - (C 8 -C 10 ) alkyl; (C 1 -C 20 ) alkoxy, such as - (C 2 -C 19 ) alkoxy, or - (C 3 -C 18 ) alkoxy, or - (C 4 -C 16 ) alkoxy, or - (C 6 -C 12 ) alkoxy; halogenated (C 1 -C 20 ) alkyl
- each B is attached to any one of F, G, H and I, while C is attached to the rest of F, G, H and I.
- B is attached to H and three C’s are separately attached to each of F, G and I.
- B is attached to G and three C’s are separately attached to each of F, H and I.
- B is attached to F and three C’s are separately attached to each of G, H and I.
- B is attached to I and three C’s are separately attached to each of F, G and H.
- two B’s are separately attached to G and H, and two C’s are separately attached to each of F and I.
- B on each occurrence, is independently selected from the group consisting of hydroxyl; -C (O) OH; -P (O) 2 -OH; -P (O) -OH; -P (O) (S) -OH; -CN; - (C 1 -C 22 ) alkyl, such as - (C 2 -C 20 ) alkyl, or - (C 3 -C 16 ) alkyl, or - (C 6 -C 12 ) alkyl; -O- (C 1 -C 22 ) alkyl, such as -O- (C 2 -C 20 ) alkyl, or -O- (C 3 -C 16 ) alkyl, or -O- (C 6 -C 12 ) alkyl; - (C 1 -C 22 ) alkenyl, such as - (C 2 -C 20 ) alkenyl, or - (C 3 -C 16 ) alken
- Each of the (C 1 -C 22 ) alkyl or (C 1 -C 22 ) alkylene included in B can be an alkyl or alkylene comprising from 1 to 22 carbon atoms, or from 2 to 20 carbon atoms, or from 3 to 16 carbon atoms, or from 4 to 12 carbon atoms, or from 6 to 12 carbon atoms, or from 8 to 10 carbon atoms.
- each of A 1 , A 2 and A 3 is either absent or a substituent independently selected from the group consisting of -H, -OH, -O-R 1 , -SH, - (C 1 -C 25 ) alkyl, halogenated - (C 1 -C 25 ) alkyl, - (C 2 -C 22 ) alkenyl, - (C 1 -C 22 ) alkylene-OH, - (C 3 -C 22 ) cycloalkyl, - (C 3 -C 22 ) cycloalkenyl, - (C 1 -C 22 ) alkylene- (C 3 -C 22 ) cycloalkyl, - (C 1 -C 22 ) alkylene-R 1 , - (C 1 -C 22 ) alkylene-O-R 1 , - (C 1 -C 22 ) alkylene-COOR 1 , -C (O) O-R
- Y is selected from the group consisting of carbon, nitrogen, oxygen and sulfur; and each of P, Q, S and T is independently selected from the group consisting of carbon, nitrogen, oxygen and sulfur; and the asterisk refers to the site wherein the substituent represented by Formula III is linked with the structure represented by Formula I or Formula II;
- each of R 3 , R 4 and R 5 is either absent or a substituent independently selected from the group consisting of -H, -OH, -O-R 1 , -SH, - (C 1 -C 25 ) alkyl, halogenated - (C 1 -C 25 ) alkyl, - (C 2 -C 22 ) alkenyl, - (C 1 -C 22 ) alkylene-OH, - (C 3 -C 22 ) cycloalkyl, - (C 3 -C 22 ) cycloalkenyl, - (C 1 -C 22 ) alkylene- (C 3 -C 22 ) cycloalkyl, - (C 1 -C 22 ) alkylene-R 1 , - (C 1 -C 22 ) alkylene-O-R 1 , - (C 1 -C 22 ) alkylene-COOR 1 , -C (O) O-R
- R 7 on each occurrence, is attached to any one of P, Q, S and T, and is either absent or selected from the group consisting of hydrogen, halogen atom, hydroxyl, (C 1 -C 20 ) alkyl, (C 1 -C 20 ) alkoxy, halogenated (C 1 -C 20 ) alkyl and halogenated (C 1 -C 20 ) alkoxy.
- M is an integer of 0, 1, 2 or 3.
- R 6 is attached to any one of P, Q, S and T, and is selected from the group consisting of direct bond, -O-, -C (O) O-, -O-C (O) -, -P (O) 2 -O-, -O-P (O) 2 -O-, -P (O) (S) -O-, -O-P (O) (S) -O-, -O-P (O) -O-, - (C 1 -C 22 ) alkylene-, - (C 1 -C 22 ) alkylene-O-, -O- (C 1 -C 22 ) alkylene-, - (C 1 -C 22 ) alkylene-NH-, -NH- (C 1 -C 22 ) alkylene-, -C (O) - (C 1 -C 22 ) alkylene-, - (C 1 -C 22 ) alkylene-C (O)
- R 1 on each occurrence, is independently selected from the group consisting of hydrogen, hydroxyl, - (C 1 -C 22 ) alkyl, - (C 3 -C 22 ) cycloalkyl, - (C 6 -C 22 ) aryl, - (C 1 -C 22 ) alkoxy, - (C 3 -C 22 ) cycloalkoxy, - (C 6 -C 22 ) aryloxy, -C (O) - (C 1 -C 22 ) alkyl, -OC (O) (C 1 -C 22 ) alkyl, -C (O) -O- (C 1 -C 22 ) alkyl, -C (O) - (C 3 -C 22 ) cycloalkyl, -OC (O) - (C 3 -C 22 ) cycloalkyl, -OC (O) - (C 3 -C 22 ) cycloalkyl,
- R 2 on each occurrence, is independently selected from the group consisting of a halogen atom, a (C 1 -C 12 ) alkyl, a (C 1 -C 12 ) alkoxy, a (C 1 -C 12 ) alkoxycarbonyl, a (C 6 -C 16 ) aryl or a (C 6 -C 16 ) aryloxycarbonyl.
- one or more hydroxyl group, carboxyl group, amino group, nitrile group and phosphoric acid group contained in A 1 , A 2 , A 3 , B, C, R 1 , R 2 , R 3 , R 4 , R 5 and R 7 can be protected with a terminal protective group R P selected from the group consisting of (C 1 -C 22 ) alkyl, (C 1 -C 22 ) alkoxy, (C 1 -C 22 ) alkylcarbonyl, (C 1 -C 22 ) alkoxycarbonyl, (C 6 -C 22 ) aryl, (C 6 -C 22 ) aryloxy, (C 6 -C 22 ) arylcarbonyl, (C 6 -C 22 ) aryloxycarbonyl, tri ( (C 1 -C 22 ) alkyl) silyl and tri ( (C 1 -C 22 ) alkoxy) silyl, wherein the (C 1 -C 22 ) alky
- a 1 , A 2 and A 3 are not simultaneously hydrogen and R 3 , R 4 and R 5 are not simultaneously hydrogen.
- X is carbon, and all of A 1 , A 2 and A 3 are present.
- X is nitrogen, both A 1 and A 2 are present and A 3 is absent.
- X is nitrogen with positive charge (i.e. quaternary ammonium) , and all of A 1 , A 2 and A 3 are present.
- X is sulfur or oxygen, both A 1 and A 3 are absent and A 2 is present.
- X is sulfur with positive charge (i.e. sulfonium) , both A 2 and A 3 are present and A 1 is absent.
- each of the (C 1 -C 22 ) alkyl or (C 1 -C 22 ) alkylene included in A 1 , A 2 , A 3 , R 1 , R 2 , R 3 , R 4 , R 5 , R 6 and R 7 can be an alkyl or alkylene comprising from 1 to 22 carbon atoms, or from 2 to 20 carbon atoms, or from 3 to 16 carbon atoms, or from 4 to 12 carbon atoms, or from 6 to 12 carbon atoms, or from 8 to 10 carbon atoms.
- one or more hydroxyl group, carboxyl group, amino group, nitrile group and phosphoric acid group contained in A 1 , A 2 , A 3 , B, C, R 1 , R 2 , R 3 , R 4 , R 5 and R 7 can be linked with a support material selected from the group consisting of silica, silica gel, glass, ceramic, polymer, cellulose, and combinations thereof.
- the solid material is in the form of a bead.
- the bead may be made of any material including, without limitation, magnetic bead, paramagnetic bead, silica bead, an agarose bead, etc.
- the compound of the present disclosure may have a structure represented by any of Formula AIV to Formula AXIII,
- a 1 , A 2 , A 3 , B, C, F, G, H, I, m and n are as defined above.
- the compound of the present disclosure has a structure represented by Formula BII
- X′ is selected from the group consisting of carbon, nitrogen, oxygen and sulfur; and each of F′, G′, H′ and I′ is independently selected from the group consisting of carbon, nitrogen, oxygen and sulfur.
- each of A 1 ′, A 2 ′ and A 3 ′ is either absent or a substituent independently selected from the group consisting of -H, -R 1 ′, -O-R 1 ′, -S-R 1 ′, -C (O) -R 1 ′, -C (O) O-R 1 ′, -O-C (O) -R 1 ′, -C (O) NH-R 1 ′, -C (O) NR 2 ′-R 1 ′, -NH-C (O) -R 1 ′, -NR 2 ′-C (O) -R 1 ′, -O-P (O) 2 -O-R 1 ′, -OP (O) (S) -O-R 1 ′, -O-P (O) -O-R 1 ′, -NH-R 1 ′, -NR 2 ′-R 1 ′, - (CH 2 ) r′
- Each of the (C 1 -C 22 ) alkylene included in A 1 ′, A 2 ′ and A 3 ′ can be (C 2 -C 20 ) alkylene, (C 3 -C 16 ) alkylene, (C 4 -C 14 ) alkylene, (C 6 -C 12 ) alkylene, or (C 8 -C 10 ) alkylene.
- R 1 ′ and R 3 ′ is independently selected from the group consisting of hydrogen; hydroxyl; - (C 1 -C 30 ) alkyl, such as - (C 2 -C 25 ) alkyl, or - (C 2 -C 22 ) alkyl, or - (C 3 -C 18 ) alkyl, or - (C 4 -C 16 ) alkyl, or - (C 6 -C 12 ) alkyl, or - (C 8 -C 10 ) alkyl; - (C 3 -C 50 ) cycloalkyl, such as - (C 4 -C 40 ) cycloalkyl, or - (C 5 -C 30 ) cycloalkyl, or - (C 6 -C 20 cycloalkyl, or - (C 6 -C 16 ) cycloalkyl; - (C 6 -C 50 ) aryl, such as - (C 6
- one or more of the substituents A 1 ′, A 2 ′ and A 3 ′ contain at least one targeting moiety.
- targeting moiety refers to a part or segment of a molecule acting as a chemical signal that binds to a molecule or complex on a particular area of a cell, tissue, or organ.
- targeting moiety modifies one or more properties of the attached oligonucleotide of the invention including but not limited to pharmacodynamic, pharmacokinetic, binding, absorption, cellular distribution, cellular uptake, charge and clearance.
- Targeting moieties are routinely used in the arts and are linked directly or via an optional linking moiety to a parent compound such as an oligomeric compound.
- a targeting moiety is selected from one or more of a ligand, a peptide, nucleic acid, oligonucleotide, aptamer, small molecule, a polyethylene glycol, an amino acid, a cholesterol, a carbohydrate (e.g. glucose, galactosamine or N-acetyl galactosamine) , an antibody or antibody fragment, and localization signal such as a nuclear localization signal or a mitochondrial localization signal.
- a ligand e.g., a peptide, nucleic acid, oligonucleotide, aptamer, small molecule, a polyethylene glycol, an amino acid, a cholesterol, a carbohydrate (e.g. glucose, galactosamine or N-acetyl galactosamine) , an antibody or antibody fragment, and localization signal such as a nuclear localization signal or a mitochondrial localization signal.
- a targeting moiety is selected from the group consisting of intercalators, reporter molecules, polyamines, polyamides, vitamin moieties, polyethylene glycols, thioethers, polyethers, thiocholesterols, cholic acid moieties, folate, lipids, fatty acids, phospholipids, biotin, phenazine, phenanthridine, anthraquinone, adamantane, acridine, fluoresceins, rhodamines, coumarins, fluorophores, luminescent proteins and dyes.
- the targeting moiety is a fluorophore, any fluorophore deemed useful may be utilized.
- Non-limiting examples of useful fluorescent proteins include but are not limited to GFP, EBFP, Azurite, Cerulean, mCFP, , Turquoise, ECFP, mKeima-Red, TagCFP, AmCyan, mTFP, TurboGFP, TagGFP, EGFP, TagYFP, EYFP, Topaz, Venus, mCitrine, TurboYFP, mOrange, TurboRFP, tdTomato, TagRFP, dsRed2, mRFP, mCherry, mPlum mRaspberry, mScarlet, etc.
- luminescent proteins include without limitation, Cypridinia luciferase, Gaussia luciferase, Renilla luciferase, Phontinus luciferase, Luciola luciferase, Pyrophorus luciferase, Phrixothrix luciferase, etc.
- a targeting moiety is selected from: a cell-penetrating peptide, polyethylene glycol, an alkaloid, a tryptamine, a benzimidazole, a quinolone, an amino acid, a cholesterol, carbohydrate, and ligand.
- a targeting moiety is a carbohydrate.
- the carbohydrate can be selected from the group consisting of monosaccharides, disaccharides, trisaccharides, and polysaccharides.
- the carbohydrate is a monosaccharide selected from the group consisting of dextrose, glucose, galactose, mannitol, D-mannose, sorbitol, and sorbose.
- the carbohydrate is a disaccharide selected from the group consisting of lactose, maltose, sucrose, and trehalose.
- a targeting moiety is a polysaccharide.
- a targeting moiety is a N-acetyl galactosamine.
- a targeting moiety is an amino acid.
- the amino acid is a hydrophobic amino acid.
- the hydrophobic amino acid is selected from the group consisting of alanine, isoleucine, leucine, methionine, phenylalanine, proline, tryptophan, and valine.
- the amino acid is a polar amino acid.
- the amino acid is selected from the group consisting of arginine, histidine, lysine, cysteine, glycine, glutamine, serine, threonine, tyrosine, aspartic acid and glutamic acid.
- a targeting moiety is selected from the group consisting of human serum albumin, ⁇ -lactalbumin, trypsinogen, and polyalanine.
- the targeting moiety comprises an active drug substance, for example, aspirin, warfarin, phenylbutazone, ibuprofen, suprofen, fen-bufen, ketoprofen, (S) - (+) -pranoprofen, carprofen, dansylsarcosine, 2, 3, 5-triiodobenzoic acid, fingolimod, flufenamic acid, folinic acid, a benzothiadiazide, chlorothiazide, a diazepine, indo-methicin, a barbiturate, a cephalosporin, a sulfa drug, an antidiabetic, an antibacterial or an antibiotic.
- an active drug substance for example, aspirin, warfarin, phenylbutazone, ibuprofen, suprofen, fen-bufen, ketoprofen, (S) - (+) -pranoprofen, carpro
- the targeting moieties disclosed herein can be used to increase uptake, introduce, delivery and target uptake of oligonucleotide to particular cell or tissue types, e.g., cells or tissues in central nervous system (e.g., brain and spinal cord) , liver, lung, kidney, intestine, pancreas, cholecyst, heart, lymph nodes, spleen, stomach, bladder, muscle and bone.
- Preferred targeting moieties include those specifically provided in the Examples.
- R 1 ′ and R 3 ′ is the targeting moiety.
- all of the R 1 ′s contained in the A 1 ′, A 2 ′ and A 3 ′ are the targeting moiety as stated above;
- all of the R 3 ′ contained in the A 1 ′, A 2 ′ and A 3 ′ are the targeting moiety as stated above;
- all of the R 1 ′ and R 3 ′ contained in the A 1 ′, A 2 ′ and A 3 ′ are the targeting moiety as stated above.
- one or more hydroxyl group, carboxyl group and amino group contained in each of R 1 ′ and R 3 ′ can be optionally protected, e.g. with a terminal protection group R P selected from the group consisting of (C 1 -C 22 ) alkyl, (C 1 -C 22 ) alkoxy, (C 1 -C 22 ) alkylcarbonyl, (C 1 -C 22 ) alkoxycarbonyl, (C 6 -C 22 ) aryl, (C 6 -C 22 ) aryloxy, (C 6 - C 22 ) arylcarbonyl, (C 6 -C 22 ) aryloxycarbonyl, tri ( (C 1 -C 22 ) alkyl) silyl and tri ( (C 1 -C 22 ) alkoxy) silyl, wherein the (C 1 -C 22 ) alkyl contained in the protection group R p can be an alkyl comprising from 1 to 22 carbon atoms,
- each of R 2 ′, R 4 ′, R 5 ′ and R 6 ′ is independently a halogen atom, such as fluorine, chlorine, bromine or iodine; a (C 1 -C 12 ) alkyl, such as (C 1 -C 10 ) alkyl, or (C 2 -C 8 ) alkyl, or (C 3 -C 6 ) alkyl, or (C 4 -C 5 ) alkyl; a (C 1 -C 12 ) alkoxy, such as (C 1 -C 10 ) alkoxy, or (C 2 -C 8 ) alkoxy, or (C 3 -C 6 ) alkoxy, or (C 4 -C 5 ) alkoxy; a (C 1 -C 12 ) alkoxycarbonyl, such as (C 1 -C 10 ) alkoxycarbonyl, or (C 2 -C 8 ) alkoxycarbonyl, or (C 3 halogen atom
- each of r′, s′, p′ and q′ is an integer from 1 to 22, such as an integer from 2 to 20, or an integer from 3 to 18, an integer from 4 to 16, an integer from 6 to 12, an integer from 8 to 10.
- a 3 ′ is absent when X′ is oxygen, and A 1 ′, A 2 ′ and A 3 ′ are not simultaneously hydrogen.
- X′ is carbon, and all of A 1 ′, A 2 ′ and A 3 ′ are present.
- X′ is nitrogen, both A 1 ′ and A 2 ′ are present and A 3 ′ is absent.
- X′ is nitrogen with positive charge (i.e. quaternary ammonium) , and all of A 1 ′, A 2 ′ and A 3 ′ are present.
- X′ is sulfur or oxygen, both A 1 ′ and A 3 ′ are absent and A 2 ′ is present.
- X′ is sulfur with positive charge (i.e. sulfonium) , both A 2 ′ and A 3 ′ are present and A 1 ′ is absent.
- each C′ is attached to any one of F′, G′, H′ and I′, and is either absent or selected from the group consisting of hydrogen; halogen atom, such as fluorine, chlorine, bromine or iodine; hydroxyl; - (C 1 -C 20 ) alkyl, such as - (C 2 -C 19 ) alkyl, or - (C 3 -C 18 ) alkyl, or - (C 4 -C 16 ) alkyl, or - (C 6 -C 12 ) alkyl, or - (C 8 -C 10 ) alkyl; - (C 1 -C 20 ) alkoxy, such as - (C 2 -C 19 ) alkoxy, or - (C 3 -C 18 ) alkoxy, or - (C 4 -C 16 ) alkoxy, or - (C 6 -C 12 ) alkoxy; halogenated (C 1 -C 20 )
- m′ is an integer of 1, 2 or 3
- n′ is an integer of 1, 2 or 3
- m′+n′ 4.
- each B′ is attached to any one of F′, G′, H′ and I′, while C′ is attached to the rest of F′, G′, H′ and I′.
- B′ is attached to H′ and three C′s are separately attached to each of F′, G′ and I′.
- B′ is attached to G′ and three C′s are separately attached to each of F′, H′ and I′.
- B′ is attached to F′ and three C′s are separately attached to each of G′, H′ and I′.
- B′ is attached to I and three C′s are separately attached to each of F′, G′ and H′.
- two B′s are separately attached to G′ and H′, and two C′s are separately attached to each of F′ and I′.
- each of B′ is independently selected from the group consisting of hydroxyl, -C (O) OH, - (C 1 -C 30 ) alkoxy, -P (O) 2 -OH, -P (O) -OH, -P (O) (S) -OH, -CN, - (C 1 -C 30 ) alkylene-OH, - (C 3 -C 50 ) cycloalkylene-OH, - (C 6 -C 50 ) arylene-OH, -C (O) -NH- [ (C 1 -C 30 ) alkylene-O] r′ -H (wherein r′ is an integer of 1 to 22) , -C (O) -NH- [ (C 1 -C 30 ) alkylene-O] r′ - (C 1 -C 30 ) alkylene-C (O) -OH (wherein r′ is an integer of 1 to 22) , -C (O)
- Y′ is selected from the group consisting of carbon, nitrogen, oxygen and sulfur; and each of P′, Q′, S′ and T′ is independently selected from the group consisting of carbon, nitrogen, oxygen and sulfur; and the asterisk refers to the site wherein the substituent represented by Formula BIII is linked with any one of F′, G′, H′ and I′ of Formula BII.
- R 7 ′ is selected from the group consisting of -O-, -C (O) O-, -O-C (O) -, -P (O) 2 -O-, -O-P (O) 2 -O-, -P (O) (S) -O-, -O-P (O) (S) -O-, -O-P (O) -O-, - (C 1 -C 30 ) alkylene-, - (C 1 -C 30 ) alkylene-O-, -O- (C 1 -C 30 ) alkylene-, - (C 1 -C 30 ) alkylene-NH-, -NH- (C 1 -C 30 ) alkylene-, -C (O) - (C 1 -C 30 ) alkylene-, - (C 1 -C 30 ) alkylene-, - (C 1 -C 30 ) alkylene-, - (C 1
- each of R 8 ′ and R 9 ′ is either absent or is a substituent independently selected from the group consisting of -H, hydroxyl, - (C 1 -C 30 ) alkyl, - (C 3 -C 50 ) cycloalkyl, - (C 6 -C 50 ) aryl, - (C 1 -C 30 ) alkylene-OH, - (C 3 -C 50 ) cycloalkylene-OH, - (C 6 -C 50 ) arylene-OH, - (C 1 -C 30 ) alkylene-C (O) OH, - (C 3 -C 50 ) cycloalkylene-C (O) OH, - (C 6 -C 50 ) arylene-C (O) OH, - (C 1 -C 30 ) alkylene-NH 2 , - (C 3 -C 50 ) cycloalkylene-NH 2 , -
- R 8 ′ and R 9 ′ are optionally protected, e.g. with the protection group R P as defined herein.
- R 8 ′ and R 9 ′ can be linked together so that R 8 ′, R 9 ′, the carbon atom linked with R 8 ′ and the Y′ atom linked with R 9 ′ form an unsubstituted or substituted heterocyclic ring.
- R 9 ′ is absent when Y′ is oxygen.
- each R 10 ′ is attached to any one of P′, Q′, S′ and T′, and is independently selected from the group consisting of hydroxyl, -C (O) OH, -P (O) 2 -OH, -P (O) -OH, -P (O) (S) -OH, -CN, - (C 1 -C 30 ) alkylene-OH, - (C 3 -C 50 ) cycloalkylene-OH, - (C 6 -C 50 ) arylene-OH, - (C 5 -C 50 ) heteroarylene-OH, - (C 1 -C 30 ) alkylene-C (O) OH, - (C 3 -C 50 ) cycloalkylene-C (O) OH, - (C 6 -C 50 ) arylene-C (O) OH, - (C 5 -C 50 ) heteroarylene-C (O) OH, -C (O) OH
- the - (C 1 -C 30 ) alkylene-contained in B′, R 7 ′ R 8 ′, R 9 ′ and R 10 ′ may include - (C 1 -C 28 ) alkylene-, or - (C 1 -C 26 ) alkylene-, or - (C 1 -C 24 ) alkylene-, or - (C 2 -C 22 ) alkylene-, or - (C 3 -C 20 ) alkylene-, or - (C 4 -C 18 ) alkylene-, or - (C 5 -C 17 ) alkylene-, or - (C 6 -C 16 ) alkylene-, or - (C 7 -C 14 ) alkylene-, or - (C 8 -C 12 ) alkylene-.
- the - (C 3 -C 50 ) cycloalkylene-contained in B′, R 7 ′ R 8 ′, R 9 ′ and R 10 ′ may include - (C 3 -C 40 ) cycloalkylene-, or - (C 3 -C 30 ) cycloalkylene-, or - (C 3 -C 22 ) cycloalkylene-, or - (C 4 -C 20 ) cycloalkylene-, or - (C 5 -C 18 ) cycloalkylene-, or - (C 6 -C 16 ) cycloalkylene-, or - (C 7 -C 14 ) cycloalkylene-, or - (C 8 -C 12 ) cycloalkylene-.
- the - (C 6 -C 50 ) arylene-contained in B′, R 7 ′ R 8 ′, R 9 ′ and R 10 ′ may include - (C 6 -C 40 ) arylene-, or - (C 6 -C 30 ) arylene-, or - (C 6 -C 22 ) arylene-, or - (C 6 -C 20 ) arylene-, or - (C 6 -C 18 ) arylene-, or - (C 6 -C 16 ) arylene-, or - (C 6 -C 12 ) arylene-.
- the - (C 5 -C 50 ) heteroarylene-contained in B′, R 7 ′ R 8 ′, R 9 ′ and R 10 ′ may include - (C 5 -C 40 ) heteroarylene-, or - (C 5 -C 30 ) heteroarylene-, or - (C 5 -C 22 ) heteroarylene-, or - (C 5 -C 18 ) heteroarylene-, or - (C 6 -C 16 ) heteroarylene-.
- the term “- (C 1 -C 30 ) alkyl” may include - (C 2 -C 22 ) alkyl, or - (C 3 -C 18 ) alkyl, or - (C 4 -C 16 ) alkyl, or - (C 6 -C 12 ) alkyl, or - (C 8 -C 10 ) alkyl;
- the term “- (C 3 -C 50 ) cycloalkyl” may include - (C 4 -C 40 ) cycloalkyl, or - (C 5 -C 30 ) cycloalkyl, or - (C 6 -C 20 ) cycloalkyl, or - (C 6 -C 16 ) cycloalkyl;
- the term “- (C 6 -C 50 ) aryl” may include - (C 6 -C 40 ) aryl, or - (C 6
- each R 11 ′ is attached to any one of P′, Q′, S′ and T′, and is either absent or selected from the group consisting of hydrogen; hydroxyl; halogen atom, such as fluorine, chlorine, bromine or iodine; - (C 1 -C 20 ) alkyl, such as - (C 2 -C 19 ) alkyl, or - (C 3 -C 18 ) alkyl, or - (C 4 -C 16 ) alkyl, or - (C 6 -C 12 ) alkyl, or - (C 8 -C 10 ) alkyl; - (C 1 -C 20 ) alkoxy, such as - (C 2 -C 19 ) alkoxy, or - (C 3 -C 18 ) alkoxy, or - (C 4 -C 16 ) alkoxy, or - (C 6 -C 12 ) alkoxy; (C 1 -C 20 ) alk
- R 9 ′ is absent when Y′ is oxygen.
- Y′ is carbon, and both of R 8 ′ and R 9 ′ are present.
- Y′ is nitrogen, R 8 ′ is present and R 9 ′ is absent.
- Y′ is nitrogen with positive charge (i.e., quaternary ammonium) , both R 8 ′ and R 9 ′ are present.
- Y′ is sulfur or oxygen, and R 9 ′ is absent.
- Y′ is sulfur with positive charge (i.e., sulfonium) , and R 9 ′ is present.
- M′ is an integer of 1, 2 or 3
- N′ is an integer of 1, 2 or 3
- M′+N′ 4.
- each R 10 ′ is attached to any one of P′, Q′, S′ and T′, while R 11 ′ is attached to the rest of P′, Q′, S′ and T′.
- R 10 ′ is attached to S′ and three R 11 ′ are separately attached to each of P′, Q′ and T′.
- R 10 ′ is attached to Q′ and three R 11 ′ are separately attached to each of P′, S′ and T′.
- R 10 ′ is attached to P′ and three R 11 ′ are separately attached to each of Q′, S′ and T′.
- R 10 ′ is attached to T′ and three R 11 ′ are separately attached to each of P′, Q′ and S′.
- two R 10 ′ are separately attached to Q′ and S′, and two R 11 ′ are separately attached to each of P′ and T′.
- one or more hydroxyl group, carboxyl group, amino group and phosphoric acid group contained in each of R 8 ′, R 9 ′ and R 10 ′ are optionally protected with a terminal protection group R P selected from the group consisting of (C 1 -C 22 ) alkyl, (C 1 -C 22 ) alkoxy, (C 1 -C 22 ) alkylcarbonyl, (C 1 -C 22 ) alkoxycarbonyl, (C 6 -C 22 ) aryl, (C 6 -C 22 ) aryloxy, (C 6 -C 22 ) arylcarbonyl, (C 6 -C 22 ) aryloxycarbonyl, tri ( (C 1 -C 22 ) alkyl) silyl and tri ( (C 1 -C 22 ) alkoxy) silyl, wherein the (C 1 -C 22 ) alkyl contained in the protection group R p can be an alkyl comprising from 1 to 22
- the support material attached to R 10 ′ can be selected from the group consisting of silica, silica gel, glass, ceramic, polymer, cellulose, and combinations thereof.
- the solid material is in the form of a bead.
- the bead may be made out of any material including, without limitation, magnetic bead, paramagnetic bead, silica bead, an agarose bead, etc.
- the compound of the present disclosure may have a structure represented by any of Formula BIV to Formula BXIV,
- a 1 ′, A 2 ′, A 3 ′, B′, C′, F′, G′, H′, I′, R 7 ′, R 8 ′, R 10 ′, R 11 ′, P′, Q′, S′, T′, m′, n′, M′ and N′ are as defined herein.
- the RING I can be formed by linking A 1 ′ and A 2 ′ together and thus may consist of the nitrogen atom to which A 1 ′ is attached, the carbon atom to which A 2 ′ is attached, at least part of A 1 ′ and at least part of A 2 ′.
- the RING I can be a 4, 5, 6, 7, 8 or 9 member ring, in particular a ring fused to the core structure.
- the RING I may consist of a nitrogen atom, a carbon atom from the core structure and 2, 3, 4, 5, 6 or 7 additional ring atoms derived from A 1 ′ and A 2 ′, wherein each of the additional ring atoms may be selected from the group consisting of carbon, nitrogen, oxygen, sulfur, and combinations thereof.
- all of the additional ring atoms may be carbon atoms.
- a 4 ′ can be a substituent attached to any atom of RING I, and each of A 4 ′, A 5 ′ and A 6 ′ is independently selected from the group consisting of -R 1 ′, -O-R 1 ′, -S-R 1 ′, -C (O) -R 1 ′, -C (O) O-R 1 ′, -O-C (O) -R 1 ′, -C (O) NH-R 1 ′, -C (O) NR 2 ′-R 1 ′, -NH-C (O) -R 1 ′, -NR 2 ′-C (O) -R 1 ′, -O-P (O) 2 -O-R 1 ′, -OP (O) (S) -O-R 1 ′, -O-P (O) -O-R 1 ′, -NH-R 1 ′, -NR 2 ′-R 1 ′, - (CH
- the RING II can be formed by linking R 8 ′ and R 9 ′ together and thus may consist of the nitrogen atom to which R 9 ′ is attached, the carbon atom to which R 8 ′ is attached, at least part of R 8 ′ and at least part of R 9 ′.
- the RING II can be a 4, 5, 6, 7, 8 or 9 member ring, in particular a ring fused to the core structure of the substituent B′.
- the RING II may consist of a nitrogen atom, a carbon atom from the core structure of the substituent B′ and 2, 3, 4, 5, 6 or 7 additional ring atoms derived from R 8 ′ and R 9 ′, wherein each of the additional ring atoms may be selected from the group consisting of carbon, nitrogen, oxygen, sulfur, and combinations thereof. In another exemplary embodiment, all of the additional ring atoms may be carbon atoms.
- R 12 ′ is attached to any atom of RING II and is selected from the group consisting of -H, hydroxyl, - (C 1 -C 30 ) alkyl, - (C 3 -C 50 ) cycloalkyl, - (C 6 -C 50 ) aryl, - (C 1 -C 30 ) alkylene-OH, - (C 3 -C 50 ) cycloalkylene-OH, - (C 6 -C 50 ) arylene-OH, - (C 1 -C 30 ) alkylene-C (O) OH, - (C 3 -C 50 ) cycloalkylene-C (O) OH, - (C 6 -C 50 ) arylene-C (O) OH, - (C 1 -C 30 ) alkylene-NH 2 , - (C 3 -C 50 ) cycloalkylene-NH 2 , - (C 6 -C 50 )
- one or more hydroxyl group, carboxyl group, amino group and phosporic acid group contained in R 12 ′ are optionally protected, e.g. with a terminal protection group R P selected from the group consisting of (C 1 -C 22 ) alkyl, (C 1 -C 22 ) alkoxy, (C 1 -C 22 ) alkylcarbonyl, (C 1 -C 22 ) alkoxycarbonyl, (C 6 -C 22 ) aryl, (C 6 -C 22 ) aryloxy, (C 6 -C 22 ) arylcarbonyl, (C 6 -C 22 ) aryloxycarbonyl, tri ( (C 1 -C 22 ) alkyl) silyl and tri ( (C 1 -C 22 ) alkoxy) silyl, wherein the (C 1 -C 22 ) alkyl contained in the protection group R p can be an alkyl comprising from 1 to 22 carbon atoms, such as
- the protection group R P can be selected from the group consisting of benzyloxycarbonyl (Cbz) , tert-butyldimethylsilyl (TBS) , 4, 4′-dimethoxytrityl (DMTr) , t-butyloxy carbonyl (Boc) , benzyl (Bn) and benzyloxy (BnO) .
- the compound of Formula BI or Formula BII may have one or more chiral centers, and each of the chiral centers can be independently R chiral, S chiral, mesomeric or racemic form.
- a 1 ′, A 2 ′ and A 3 ′ is selected from -CH (- (CH 2 ) r′ -NH-C (O) - (CH 2 ) s′ -R 1 ′) (-NH-C (O) - (CH 2 ) q′ -R 3 ′) , -CH (- (CH 2 ) r′ -C (O) -NH- (CH 2 ) s′ -R 1 ′) (-C (O) -NH- (CH 2 ) q′ -R 3 ′) , -N (- (CH 2 ) r′ -NH-C (O) - (CH 2 ) s′ -R 1 ′) (-NH-C (O)
- the compound of the present disclosure has a structure represented by any one of Formula BXV to Formula BXX:
- each of the double bond and the atoms or moieties attached to the same may be in E form or Z form.
- the compound of the present disclosure has a structure represented by any of the following molecular formulae:
- an oligonucleotide delivery agent comprising a DEC compound disclosed by the present application and at least one oligonucleotide. It can be fully understood that when the DEC is directly linked with the oligonucleotide or indirectly linked with the oligonucleotide via at least one linking moiety, one or more terminal atoms (such as hydrogen, halogen, nitrogen, oxygen, sulfur, phosphorus, etc. ) or terminal groups (such as hydroxyl group, amino group, ester group, ether group, acyl group, etc. ) of the DEC may be detached so as to provide an active site linkable to the linking moiety or the oligonucleotide.
- terminal atoms such as hydrogen, halogen, nitrogen, oxygen, sulfur, phosphorus, etc.
- terminal groups such as hydroxyl group, amino group, ester group, ether group, acyl group, etc.
- the linking moieties when present, can be selected from the group consisting of -O-, -S-, -C (O) -, -NH-, -N ( (C 1 -C 12 ) alkyl) -, -N ( (C 1 -C 12 ) alkyl) -C (O) -O-, -O-C (O) -, -C (O) -O-, -O-C (O) -O-, -C (O) -NH-, -OP (O) 2 O-, -P (O) (O - ) O-, -OP (O) O-, -OP (O) (S) O-, -O-S (O) 2 -O-, -S (O) 2 -O-, -S (O) -O-, - (C 1 -C 22 ) alkylene-, - (C 1 -C 22 ) alkylene-NH
- Another embodiment of the present disclosure provides a delivery enhancing compound (DEC) conjugated oligonucleotide comprising a structure represented by Formula AA, which can more efficiently deliver the oligonucleotide both in vitro and in vivo.
- DEC delivery enhancing compound
- the Pdelivery enhancing compound moiety” shown in Formula AA is derived from any one of the above stated DEC compounds of the present disclosure. It can be understood that one or more terminal atoms (such as hydrogen, halogen, nitrogen, oxygen, sulfur, phosphorus, etc. ) or terminal groups (such as hydroxyl group, amino group, ester group, ether group, acyl group, etc. ) of the inventive compound have be detached so as to provide an active site linkable to the targeting moiety or the oligonucleotide, hence the “delivery enhancing compound” of Formula BA can be considered as a moiety obtained by subtracting said one or more atoms or terminal groups from the inventive compounds.
- terminal atoms such as hydrogen, halogen, nitrogen, oxygen, sulfur, phosphorus, etc.
- terminal groups such as hydroxyl group, amino group, ester group, ether group, acyl group, etc.
- the delivery enhancing compound (DEC) conjugated oligonucleotide has a structure represented by Formula BB,
- each of A 1 ′, A 2 ′, A 3 ′, X′, F′, G′H′, I′, C′, m′ and n′ is as defined above.
- at least one of A 1 ′, A 2 ′ and A 3 ′ comprises one or more targeting moieties
- the DEC is attached to at least one oligonucleotide via the B′′ group.
- the B′′ group can be derived by detaching one or more terminal atoms (such as hydrogen, halogen, nitrogen, oxygen, sulfur, phosphorus, etc. ) or one or more terminal groups (such as hydroxyl group, amino group, ester group, ether group, acyl group, etc. ) from the B′′ group contained in Formula BII.
- the delivery enhancing compound (DEC) conjugated oligonucleotide has a structure represented by Formula BC,
- each of A 1 ′, A 2 ′, A 3 ′, R 7 ′, R 9 ′, R 10 ′, R 11 ′, X′, Y′, F′, G′, H′, I′, C′, R′, Q′, S′, T′, m′, n′, M′ and N′ is as defined above.
- at least one of A 1 ′, A 2 ′ and A 3 ′ comprises one or more targeting moieties, and the DEC is attached to at least one oligonucleotide via the R 8 ′′ group.
- the R 8 ′′ group can be derived by detaching one or more terminal atoms (such as hydrogen, halogen, nitrogen, oxygen, sulfur, phosphorus, etc. ) or one or more terminal groups (such as hydroxyl group, amino group, ester group, ether group, acyl group, etc. ) from the R 8 ′ group contained in Formula BIII.
- one or more terminal atoms such as hydrogen, halogen, nitrogen, oxygen, sulfur, phosphorus, etc.
- terminal groups such as hydroxyl group, amino group, ester group, ether group, acyl group, etc.
- the delivery enhancing compound (DEC) conjugated oligonucleotide has a structure represented by any one of Formulae E1 to E15,
- each of A 1 ′, A 2 ′, R 7 ′ and R 10 ′ is as defined above.
- at least one of A 1 ′ and A 2 ′ comprises one or more targeting moieties, or A 2 ′ comprises one or more targeting moieties; and the DEC is attached to at least one oligonucleotide via the B′′ group or the R 8 ′′ group.
- the R 8 ′′ group can be derived by detaching one or more terminal atoms (such as hydrogen, halogen, nitrogen, oxygen, sulfur, phosphorus, etc. ) or one or more terminal groups (such as hydroxyl group, amino group, ester group, ether group, acyl group, etc.
- the B′′ group can be derived by detaching one or more terminal atoms (such as hydrogen, halogen, nitrogen, oxygen, sulfur, phosphorus, etc. ) or one or more terminal groups (such as hydroxyl group, amino group, ester group, ether group, acyl group, etc. ) from the B′ group.
- one or more terminal atoms such as hydrogen, halogen, nitrogen, oxygen, sulfur, phosphorus, etc.
- terminal groups such as hydroxyl group, amino group, ester group, ether group, acyl group, etc.
- the targeting moiety is contained in the molecular structure of the DEC compound, such as being as part of the A 1 ′, A 2 ′ or A 3 ′, and preferably being the R 1 ′ or R 3 ′ group.
- the targeting moiety is independent of the DEC compound and is additionally added after or during the synthesis of the DEC compound.
- the targeting moiety and the delivery enhancing compound moiety can be linked with each other by a first linking moiety.
- the delivery enhancing compound moiety and the oligonucleotide can be linked by a second linking moiety.
- Each of the first and second linking moieties can be selected from the group consisting of direct bond, -O-, -S-, -C (O) -, -NH-, -N ( (C 1 -C 12 ) alkyl) -, -N ( (C 1 -C 12 ) alkyl) -C (O) -O-, -O-C (O) -, -C (O) -O-, -O-C (O) -O-, -C (O) -O-, -C (O) -NH-, -OP (O) 2 O-, -OP (O) O-, -OP (O) (S) O-, -O-S (O) 2 -O-,
- the delivery enhancing compound moiety in delivery enhancing compound (DEC) conjugated oligonucleotide of Formula A, is directly linked with the oligonucleotide when the linking moiety is a direct bond. In one embodiment, in the delivery enhancing compound (DEC) conjugated oligonucleotide of Formula BA, the targeting moiety is directly linked with the delivery enhancing compound moiety when the linking moiety is a direct bond.
- the oligonucleotide may include those disclosed above, and particularly can be selected from the group consisting of antisense oligonucleotide (ASO) , antisense RNA, short interfering RNA (siRNA) , microRNA (miRNA) , saRNA, dsRNA, scRNA, sgRNA or any other oligonucleotide targeting at least one nucleic acid sequence.
- ASO antisense oligonucleotide
- siRNA short interfering RNA
- miRNA microRNA
- saRNA saRNA
- dsRNA dsRNA
- scRNA sgRNA
- any other oligonucleotide targeting at least one nucleic acid sequence any other oligonucleotide targeting at least one nucleic acid sequence.
- nucleotides of the oligonucleotides described herein may be natural, i.e., non-chemically modified, nucleotides or at least one nucleotide may be a chemically modified nucleotide.
- Non-limiting examples of the chemical modification include one or more of a combination of the following:
- At least one nucleotide in the oligonucleotide sequence being a locked nucleic acid
- oligonucleotide sequence being a deoxyribonucleotide (DNA) .
- modifications of nucleotides or oligonucleotides in the present disclosure are well known to those skilled in the art, and modifications of the phosphodiester bond refer to modifications of oxygen in the phosphodiester bond, including phosphorothioate modifications and boronated phosphate modifications.
- the modifications disclosed herein stabilize an oligonucleotide structure, maintaining high specificity and high affinity for base pairing.
- the modifications disclosed herein also stabilize a nucleic acid structure and maintain its delivering accessory properties including bioavailability, biodistribution, and/or cellular uptake of the oligonucleotide agent in various tissues prefrontal cortex, cerebellum, spinal cord (e.g., cervical, thoracic, lumber) , muscle, liver, and kidney.
- the chemical modification is to substitute the phosphodiester bond with phosphorothioate (PS) bond on the backbone of the oligonucleotide disclosed herein.
- the oligonucleotide disclosed herein comprises at least one PS backbone modification in one oligonucleotide strand.
- the oligonucleotide comprises at least 3, 4, 5, 6, 7, 8, 9, 10, 12, 14, 16, 18, 20, 24, 28, 32, or 40 PS backbone modifications in one oligonucleotide strand.
- the nucleotides or oligonucleotides of the present application includes at least one chemically modified nucleotide which is modified at 2′-OH in pentose of a nucleotide, i.e., the introduction of certain substituents at the hydroxyl position of the ribose, such as 2′-fluoro modification, 2′-oxymethyl modification, 2′-oxyethylidene methoxy modification, 2, 4′-dinitrophenol modification, locked nucleic acid (LNA) , 2′-amino modification or 2′-deoxy modification, e.g., a 2’-deoxy-2’-fluoro modified nucleotide, a 2’-deoxy-modified nucleotide.
- LNA locked nucleic acid
- the nucleotides or oligonucleotides of the present application includes at least one chemically modified nucleotide which is modified at the base of the nucleotide, e.g., 5 ′-bromouracil modification, 5’-iodouracil modification, N-methyluracil modification, or 2, 6-diaminopurine modification.
- the chemical modification of the nucleotides or oligonucleotides in the present application is an addition of a (E) -vinylphosphonate moiety at the 5’ end of the sense or antisense sequence.
- the chemical modification of the at least one chemically modified nucleotide is an addition of a 5′-methyl cytosine moiety at the 5’ end of the sense or antisense sequence.
- the oligonucleotides used in the present disclosure may be commercially available from various vendors, or synthesized in a lab scale or industrial scale.
- the oligonucleotides can be synthesized by using a commercialized synthesizer or a particularly customized synthesizer, such as a K&ADNA synthesizer purchased from K&ALaborgeraete GbR, Schaafheim, Germany, by using ordinary synthesis procedures, e.g. a solid phase synthesis technique comprising the steps of sequentially adding batches of raw materials (e.g. phosphoramidite monomers including various linkers and conjugates) onto a solid support known in the art and subjecting each base addition to a preparation cycle consisted of four chemical reactions of detritylation, coupling, oxidation/thiolation and capping, so as to produce the oligonucleotides with desired full-length.
- a solid phase synthesis technique comprising the steps of sequentially adding batches of raw materials (e.g. phosphoramidite monomers including various linkers and conjugates) onto a solid support known in the art and subjecting each base addition to a preparation cycle consisted of four chemical reactions of de
- the oligonucleotide delivery agent may comprise one, two, three, four, five, six or even more oligonucleotides separately linked with one, two, three, four, five, six or even more of the delivery enhancing compounds via one, two, three, four, five, six or even more linking moieties.
- the oligonucleotide delivery agent may have a structure represented by any of the following formulae AAI to AAXXIV:
- L represents the linking moiety
- the symbol epresents a double strand oligonucleotide, either symmetric or asymmetric independently on each of the ends; presents a single strand oligonucleotide, and each of a, b and c is independently an integer from 1 to 50, such as an integer from 2 to 45, or an integer from 3 to 40, or an integer from 4 to 35, or an integer from 5 to 30, or an integer from 10 to 20.
- Each of the linking moiety, and thus the delivery enhancing compound may be linked at 3’ end, 5’ end or any internal position, such as the n th nucleotide, of the double or single strand oligonucleotide.
- one or more of the substituents A 1 , A 2 , A 3 , B, C, A 1 ′, A 2 ′, A 3 ′, B′ and C′ of the delivery enhancing compound can be linked with the linking moiety or with the oligonucleotide (when the linking moiety is a direct bond) .
- the linking moiety is a direct bond
- either one of A 1 , A 2 , A 3 , B, A 1 ′, A 2 ′, A 3 ′ R 8 ′, R 12 ′and B′ preferably A 2 A 2 ′, B, B′, R 8 ′ or R 12 ′ is linked with the linking moiety or the oligonucleotide.
- one or more terminal groups of the above said substituents may be cleavaged or hydrolyzed to decap an active site (such as a - (C 1 -C 22 ) alkylene-OH group) which is then linked with the oligonucleotide via a linking moiety (such as -P (O) (O - ) O-) .
- an active site such as a - (C 1 -C 22 ) alkylene-OH group
- a linking moiety such as -P (O) (O - ) O-
- exemplary structures of the oligonucleotide delivery agents include O1 to O25 as illustrated below:
- J represents O or S.
- the delivery enhancing compounds are linked with double-stranded RNA (dsRNA) duplexes (including but not limited to siRNA or saRNA) and/or single-stranded antisense oligonucleotides (ASOs) at the 3’-terminus or 5’-terminus their passenger (P) strand via a linking moiety, such as -OP (O) 2 O- (-P (O) (O - ) -O-) , wherein P is passenger strand and G is guide strand; and electron rearrangement may also occur.
- dsRNA duplexes including but not limited to siRNA or saRNA
- ASOs single-stranded antisense oligonucleotides
- At least one hydrogen atom (i.e. H) contained in the delivery enhancing compound, the delivery enhancing compound moiety, the linking moiety and/or the oligonucleotide is substituted with deuterium atom (i.e. D) .
- at least one of the delivery enhancing compound, the delivery enhancing compound moiety, the linking moiety and the oligonucleotide comprises one to twenty, such as one to fifteen, or one to twelve, or one to ten, or one to eight, or one to six, or one to three, or one to two deuterium atoms.
- 1%to 100%, or 2%to 90%, or 5%to 80%, or 10%to 70%, or 20%to 60%, or 30%to 50%, or 40%to 45%by mole of the hydrogen atom contained in the the delivery enhancing compound, the delivery enhancing compound moiety, the linking moiety and/or the oligonucleotide is substituted with deuterium atom.
- the deuterium substitution rate can be within a numerical range obtained by combining any two of the above said end point values.
- compositions comprising the oligonucleotide delivery agent.
- the pharmaceutical composition may comprise one or more additional ingredients, such as pharmaceutically acceptable carrier, excipient, solvent, diluent, stabilizer, dispersant, buffer, compatibilizer, preservative agent and combinations thereof.
- Another embodiment of the present disclosure provides a method of modulating the expression of a target gene in vitro or in vivo, comprising the step of administrating the pharmaceutical composition to a subject, or contacting the pharmaceutical composition with cells of the subject.
- the oligonucleotide delivery agent and pharmaceutical composition can be applied in various organs, tissues and cells, such as liver, lung, kidney, intestine, pancreas, cholecyst, heart, lymp nodes, spleen, stomach, bladder, muscle, bone, central nervous system (CNS) , and modulate the expression of one or more target genes in the cell thereof.
- the present disclosure provides an oligonucleotide delivery enhancing compound comprising a nitrogen-containing five membered heterocyclic ring moiety and at least one substituent directly or indirectly attachable to an oligonucleotide.
- the oligonucleotide delivery enhancing compound has a structure represented by Formula AI or Formula AII
- X on each occurrence, is an atom selected from the group consisting of carbon, nitrogen, oxygen and sulfur
- each of F, G, H and I is independently selected from the group consisting of carbon, nitrogen, oxygen and sulfur
- C on each occurrence, is either absent or selected from the group consisting of hydrogen, halogen atom, hydroxyl, (C 1 -C 20 ) alkyl, (C 1 -C 20 ) alkoxy, halogenated (C 1 -C 20 ) alkyl and halogenated (C 1 -C 20 ) alkoxy;
- B on each occurrence, is independently selected from the group consisting of hydroxyl, -C (O) OH, -P (O) 2 -OH, -P (O) -OH, -P (O) (S) -OH, -CN, - (C 1 -C 22 ) alkyl, - (C 1 -C 22 ) alkenyl, - (C 1 -C 22 ) alkylene-OH, - (C 3 -C 22 ) cycloalkylene-OH, - (C 6 -C 22 ) arylene-OH, - (C 6 -C 22 ) heteroarylene-OH, - (C 1 -C 22 ) alkylene-C (O) OH, - (C 3 -C 22 ) cycloalkylene-C (O) OH, - (C 6 -C 22 ) arylene-C (O) OH, - (C 5 -C 22 ) heteroarylene-
- each of A 1 , A 2 and A 3 is either absent or a substituent independently selected from the group consisting of -H, -OH, -O-R 1 , -SH, - (C 1 -C 25 ) alkyl, halogenated - (C 1 -C 25 ) alkyl, - (C 2 -C 22 ) alkenyl, - (C 1 -C 22 ) alkylene-OH, - (C 3 -C 22 ) cycloalkyl, - (C 3 -C 22 ) cycloalkenyl, - (C 1 -C 22 ) alkylene- (C 3 -C 22 ) cycloalkyl, - (C 1 -C 22 ) alkylene-R 1 , - (C 1 -C 22 ) alkylene-O-R 1 , - (C 1 -C 22 ) alkylene-COOR 1 , -C (O) O-R 1 ,
- Y is selected from the group consisting of carbon, nitrogen, oxygen and sulfur; and each of P, Q, S and T is independently selected from the group consisting of carbon, nitrogen, oxygen and sulfur; and the asterisk refers to the site wherein the substituent represented by Formula AIII is linked with the structure represented by Formula AI or Formula AII;
- each of R 3 , R 4 and R 5 is either absent or a substituent independently selected from the group consisting of -H, -OH, -O-R 1 , -SH, - (C 1 -C 25 ) alkyl, halogenated - (C 1 -C 25 ) alkyl, - (C 2 -C 22 ) alkenyl, - (C 1 -C 22 ) alkylene-OH, - (C 3 -C 22 ) cycloalkyl, - (C 3 -C 22 ) cycloalkenyl, - (C 1 -C 22 ) alkylene- (C 3 -C 22 ) cycloalkyl, - (C 1 -C 22 ) alkylene-R 1 , - (C 1 -C 22 ) alkylene-O-R 1 , - (C 1 -C 22 ) alkylene-COOR 1 , -C (O) O-R 1 ,
- R 7 on each occurrence, is attached to any one of P, Q, S and T, and is either absent or selected from the group consisting of hydrogen, halogen atom, hydroxyl, (C 1 -C 20 ) alkyl, (C 1 -C 20 ) alkoxy, halogenated (C 1 -C 20 ) alkyl and halogenated (C 1 -C 20 ) alkoxy;
- M is an integer of 0, 1, 2 or 3;
- R 6 is attached to any one of P, Q, S and T, and is selected from the group consisting of direct bond, -O-, -C (O) O-, -O-C (O) -, -P (O) 2 -O-, -O-P (O) 2 -O-, -P (O) (S) -O-, -O-P (O) (S) -O-, -O-P (O) -O-, - (C 1 -C 22 ) alkylene-, - (C 1 -C 22 ) alkylene-O-, -O- (C 1 -C 22 ) alkylene-, - (C 1 -C 22 ) alkylene-NH-, -NH- (C 1 -C 22 ) alkylene-, -C (O) - (C 1 -C 22 ) alkylene-, - (C 1 -C 22 ) alkylene-C (O) - (
- R 1 on each occurrence, is independently selected from the group consisting of hydrogen, hydroxyl, - (C 1 -C 22 ) alkyl, - (C 3 -C 22 ) cycloalkyl, - (C 6 -C 22 ) aryl, - (C 1 -C 22 ) alkoxy, - (C 3 -C 22 ) cycloalkoxy, - (C 6 -C 22 ) aryloxy, -C (O) - (C 1 -C 22 ) alkyl, -OC (O) (C 1 -C 22 ) alkyl, -C (O) -O- (C 1 -C 22 ) alkyl, -C (O) - (C 3 -C 22 ) cycloalkyl, -OC (O) - (C 3 -C 22 ) cycloalkyl, -OC (O) - (C 3 -C 22 ) cycloalkyl,
- R 2 on each occurrence, is independently selected from the group consisting of a halogen atom, a (C 1 -C 12 ) alkyl, a (C 1 -C 12 ) alkoxy, a (C 1 -C 12 ) alkoxycarbonyl, a (C 6 -C 16 ) aryl or a (C 6 -C 16 ) aryloxycarbonyl;
- the oligonucleotide delivery enhancing compound according to claim 1 comprising a moiety represented by Formula BI and at least one substituent directly or indirectly attachable to an oligonucleotide,
- X′ is selected from the group consisting of carbon, nitrogen, oxygen and sulfur; each of F′, G′, H′ and I′ is independently selected from the group consisting of carbon, nitrogen, oxygen and sulfur; and each of the asterisks refers to a site optionally linked to at least one substituent or an oligonucleotide directly or indirectly.
- the oligonucleotide delivery enhancing compound has a structure represented by Formula BII
- X′ is selected from the group consisting of carbon, nitrogen, oxygen and sulfur; and each of F′, G′, H′ and I′ is independently selected from the group consisting of carbon, nitrogen, oxygen and sulfur;
- each of A 1 ′, A 2 ′ and A 3 ′ is either absent or a substituent independently selected from the group consisting of -H, -R 1 ′, -O-R 1 ′, -S-R 1 ′, -C (O) -R 1 ′, -C (O) O-R 1 ′, -O-C (O) -R 1 ′, -C (O) NH-R 1 ′, -C (O) NR 2 ′-R 1 ′, -NH-C (O) -R 1 ′, -NR 2 ′-C (O) -R 1 ′, -O-P (O) 2 -O-R 1 ′, -OP (O) (S) -O-R 1 ′, -O-P (O) -O-R 1 ′, -NH-R 1 ′, -NR 2 ′-R 1 ′, - (CH 2 ) r′ -NH
- each C′ is attached to any one of F′, G′, H′ and I′, and is either absent or selected from the group consisting of hydrogen, halogen atom, hydroxyl, (C 1 -C 20 ) alkyl, (C 1 -C 20 ) alkoxy, halogenated (C 1 -C 20 ) alkyl and halogenated (C 1 -C 20 ) alkoxy;
- n′ is an integer of 1, 2 or 3
- m′+n′ 4;
- each B′ is attached to any one of F′, G′, H′ and I′, and is independently selected from the group consisting of hydroxyl, -C (O) OH, - (C 1 -C 30 ) alkoxy, -P (O) 2 -OH, -P (O) -OH, -P (O) (S) -OH, -CN, - (C 1 -C 30 ) alkylene-OH, - (C 3 -C 50 ) cycloalkylene-OH, - (C 6 -C 50 ) arylene-OH, - (C 5 -C 50 ) heteroarylene-OH, - (C 1 -C 30 ) alkylene-C (O) OH, - (C 3 -C 50 ) cycloalkylene-C (O) OH, - (C 6 -C 50 ) arylene-C (O) OH, - (C 5 -C 50 ) heteroarylene-OH,
- Y′ is selected from the group consisting of carbon, nitrogen, oxygen and sulfur; and each of P′, Q′, S′ and T′ is independently selected from the group consisting of carbon, nitrogen, oxygen and sulfur; and the asterisk refers to the site wherein the substituent represented by Formula BIII is linked with any one of F′, G′, H′ and I′ of Formula BII;
- R 7 ′ is selected from the group consisting of -O-, -C (O) O-, -O-C (O) -, -P (O) 2 -O-, -O-P (O) 2 -O-, -P (O) (S) -O-, -O-P (O) (S) -O-, -O-P (O) -O-, - (C 1 -C 30 ) alkylene-, - (C 1 -C 30 ) alkylene-O-, -O- (C 1 -C 30 ) alkylene-, - (C 1 -C 30 ) alkylene-NH-, -NH- (C 1 -C 30 ) alkylene-, -C (O) - (C 1 -C 30 ) alkylene-, - (C 1 -C 30 ) alkylene-, - (C 1 -C 30 ) alkylene-, - (C 1
- each R 10 ′ is attached to any one of P′, Q′, S′ and T′, and is independently selected from the group consisting of hydroxyl, -C (O) OH, -P (O) 2 -OH, -P (O) -OH, -P (O) (S) -OH, -CN, - (C 1 -C 30 ) alkylene-OH, - (C 3 -C 50 ) cycloalkylene-OH, - (C 6 -C 50 ) arylene-OH, - (C 5 -C 50 ) heteroarylene-OH, - (C 1 -C 30 ) alkylene-C (O) OH, - (C 3 -C 50 ) cycloalkylene-C (O) OH, - (C 6 -C 50 ) arylene-C (O) OH, - (C 5 -C 50 ) heteroarylene- C (O) OH, -C (O) -
- each R 11 ′ is attached to any one of P′, Q′, S′ and T′, and is either absent or selected from the group consisting of hydrogen, halogen atom, hydroxyl, (C 1 -C 20 ) alkyl, (C 1 -C 20 ) alkoxy, (C 1 -C 20 ) alkoxycarbonyl, halogenated (C 1 -C 20 ) alkyl and halogenated (C 1 -C 20 ) alkoxycarbonyl; and
- M′ is an integer of 1, 2 or 3
- N′ is an integer of 1, 2 or 3
- M′+N′ 4.
- one or more hydroxyl group, carboxyl group, amino group, nitrile group and phosphoric acid group contained in each of A 1 , A 2 , A 3 , B, C, R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , R 1 ′, R 2 ′, R 3 ′, R 4 ′, R 5 ′, R 6 ′, R 7 ′, R 8 ′, R 9 ′, R 10 ′ and R 11 ′ are optionally protected with a terminal protection group R P selected from the group consisting of (C 1 -C 22 ) alkyl, (C 1 -C 22 ) alkoxy, (C 1 -C 22 ) alkylcarbonyl, (C 1 -C 22 ) alkoxycarbonyl, (C 6 -C 22 ) aryl, (C 6 -C 22 ) aryloxy, (C 6 -C 22 ) arylcarbonyl, (C 6 -
- the support material is selected from the group consisting of silica, silica gel, glass, ceramic, polymer, cellulose, and combinations thereof.
- the oligonucleotide delivery enhancing compound has a structure represented by any of Formula AIV to Formula AXIII and Formula BIV to BXIV,
- a 1 , A 2 , A 3 , A 4 , F, G, H, I, B, C, P, Q, S, T, R 6 , R 7 , m, n and M are as defined herein,
- each of RING I and RING II is a 4, 5, 6, 7, 8 or 9 member ring;
- a 4 ′ is attached to any atom of RING I, and each of A 4 ′, A 5 ′ and A 6 ′ is independently selected from the group consisting of -R 1 ′, -O-R 1 ′, -S-R 1 ′, -C (O) -R 1 ′, -C (O) O-R 1 ′, -O-C (O) -R 1 ′, -C (O) NH-R 1 ′, -C (O) NR 2 ′-R 1 ′, -NH-C (O) -R 1 ′, -NR 2 ′-C (O) -R 1 ′, -O-P (O) 2 -O-R 1 ′, -OP (O) (S) -O-R 1 ′, -O-P (O) -O-R 1 ′, -NH-R 1 ′, -NR 2 ′-R 1 ′, - (CH 2 )
- R 12 ′ is attached to any atom of RING II and is selected from the group consisting of -H, hydroxyl, - (C 1 -C 30 ) alkyl, - (C 3 -C 50 ) cycloalkyl, - (C 6 -C 50 ) aryl, - (C 1 -C 30 ) alkylene-OH, - (C 3 -C 50 ) cycloalkylene-OH, - (C 6 -C 50 ) arylene-OH, - (C 1 -C 30 ) alkylene-C (O) OH, - (C 3 -C 50 ) cycloalkylene-C (O) OH, - (C 6 -C 50 ) arylene-C (O) OH, - (C 1 -C 30 ) alkylene-NH 2 , - (C 3 -C 50 ) cycloalkylene-NH 2 , - (C 6 -C 50 )
- each of F, G, H and I is carbon, m is 1 and n is 3, B is attached to G or H, each of P, Q, S and T is carbon, R 6 is attached to any one of Q and S;
- protection group R P is selected from the group consisting of benzyloxycarbonyl (Cbz) , tert-butyldimethylsilyl (TBS) , 4, 4′-dimethoxytrityl (DMTr) , t-butyloxy carbonyl (Boc) , benzyl (Bn) and benzyloxy (BnO) ;
- C on each occurrence, is selected from the group consisting of hydrogen, halogen atom, hydroxyl, (C 1 -C 12 ) alkyl, (C 1 -C 12 ) alkoxy, halogenated (C 1 -C 12 ) alkyl and halogenated (C 1 -C 12 ) alkoxy;
- B on each occurrence, is selected from the group consisting of - (C 1 -C 22 ) alkylene-OH, -O-C (O) - (C 1 -C 16 ) alkylene-C (O) NH 2 , - (C 1 -C 16 ) alkylene-O-C (O) - (C 1 -C 16 ) alkylene-C (O) NH 2 , -O-C (O) - (C 1 -C 16 ) alkylene-C (O) OH, - (C 1 -C 16 ) alkylene-O-C (O) - (C 1 -C 16 ) alkylene-C (O) OH, -C (O) - (C 1 -C 16 ) alkylene-C (O) NH 2 , - (C 1 -C 16 ) alkylene-C (O) - (C 1 -C 16 ) alkylene-C (O) NH 2 ,
- each of A 1 , A 2 and A 3 is either absent or a substituent independently selected from the group consisting of -H, -OH, linear or branched - (C 6 -C 22 ) alkyl, linear or branched - (C 2 -C 22 ) alkenyl, - (C 1 -C 22 ) alkylene-OH, - (C 3 -C 22 ) cycloalkyl, - (C 3 -C 22 ) cycloalkenyl, - (C 1 -C 22 ) alkylene- (C 3 -C 22 ) cycloalkyl, - (C 1 -C 22 ) alkylene-R 1 , - (C 1 -C 22 ) alkylene-O-R 1 , - (C 1 -C 22 ) alkylene-COOR 1 , -O- (C 1 -C 22 ) alkyl, - (C 6 -C 22 ) alkylene-CO
- Y is selected from the group consisting of carbon, nitrogen, oxygen and sulfur; and each of P, Q, S and T is carbon;
- each of R 3 , R 4 and R 5 is either absent or a substituent independently selected from the group consisting of -H, -OH, linear or branched - (C 6 -C 22 ) alkyl, linear or branched - (C 2 -C 22 ) alkenyl, - (C 1 -C 22 ) alkylene-OH, - (C 3 -C 22 ) cycloalkyl, - (C 3 -C 22 ) cycloalkenyl, - (C 1 -C 22 ) alkylene- (C 3 -C 22 ) cycloalkyl, - (C 1 -C 22 ) alkylene-R 1 , - (C 1 -C 22 ) alkylene-O-R 1 , - (C 1 -C 22 ) alkylene-COOR 1 , -O- (C 1 -C 22 ) alkyl, - (C 6 -C 22 ) alkylene-CO
- R 7 on each occurrence, is attached to any one of P, Q, S and T, and is either absent or selected from the group consisting of hydrogen, halogen atom, hydroxyl, (C 1 -C 20 ) alkyl, (C 1 -C 20 ) alkoxy, halogenated (C 1 -C 20 ) alkyl and halogenated (C 1 -C 20 ) alkoxy;
- M is an integer of 0, 1, 2 or 3;
- R 6 is attached to any one of P, Q, S and T, and is selected from the group consisting of - (C 1 -C 16 ) alkylene-, - (C 1 -C 16 ) alkylene-O-, -O- (C 1 -C 16 ) alkylene-, - (C 1 -C 16 ) alkylene-NH-, -NH- (C 1 -C 16 ) alkylene-, -C (O) - (C 1 -C 16 ) alkylene-, - (C 1 -C 16 ) alkylene-C (O) -, -C (O) -O- (C 1 -C 16 ) alkylene-, - (C 1 -C16) alkylene-C (O) -O-, -C (O) -NH- (C 1 -C 16 ) alkylene-, -C (O) - NH- (C 1 -C 16 ) alkylene-,
- the oligonucleotide delivery enhancing compound has a structure represented by any one of Formula BXV to Formula BXXIX,
- each of A 1 ′ and A 2 ′ is a substituent independently selected from the group consisting of -R 1 ′, -O-R 1 ′, -S-R 1 ′, -C (O) -R 1 ′, -C (O) O-R 1 ′, -O-C (O) -R 1 ′, -C (O) NH-R 1 ′, -C (O) NR 2 ′-R 1 ′, -NH-C (O) -R 1 ′, -NR 2 ′-C (O) -R 1 ′, -O-P (O) 2 -O-R 1 ′, -OP (O) (S) -O-R 1 ′, -O-P (O) -O-R 1 ′, -NH-R 1 ′, -NR 2 ′-R 1 ′, - (CH 2 ) r′ -NH-R 1 ′, - (CH 2 )
- n′ is an integer of 1, 2 or 3
- m′+n′ 4;
- each B′ is independently selected from the group consisting of hydroxyl, -C (O) OH, - (C 1 -C 30 ) alkoxy, -P (O) 2 -OH, -P (O) -OH, -P (O) (S) -OH, -CN, - (C 1 -C 30 ) alkylene-OH, - (C 3 -C 50 ) cycloalkylene-OH, - (C 6 -C 50 ) arylene-OH, - (C 5 -C 50 ) heteroarylene-OH, - (C 1 -C 30 ) alkylene-C (O) OH, - (C 3 -C 50 ) cycloalkylene-C (O) OH, - (C 6 -C 50 ) arylene-C (O) OH, - (C 5 -C 50 ) heteroarylene-C (O) OH, -C (O) -NH- (C 1
- each R 7 ′ is selected from the group consisting of -O-, -C (O) O-, -O-C (O) -, -P (O) 2 -O-, -O-P (O) 2 -O-, -P (O) (S) -O-, -O-P (O) (S) -O-, -O-P (O) -O-, - (C 1 -C 30 ) alkylene-, - (C 1 -C 30 ) alkylene-O-, -O- (C 1 -C 30 ) alkylene-, - (C 1 -C 30 ) alkylene-NH-, -NH- (C 1 -C 30 ) alkylene-, -C (O) - (C 1 -C 30 ) alkylene-, - (C 1 -C 30 ) alkylene-, - (C 1 -C 30 ) alkylene-, - (C 1
- each R 10 ′ is independently selected from the group consisting of hydroxyl, -C (O) OH, -P (O) 2 -OH, -P (O) -OH, -P (O) (S) -OH, -CN, - (C 1 -C 30 ) alkylene-OH, - (C 3 -C 50 ) cycloalkylene-OH, - (C 6 -C 50 ) arylene-OH, - (C 5 -C 50 ) heteroarylene-OH, - (C 1 -C 30 ) alkylene-C (O) OH, - (C 3 -C 50 ) cycloalkylene-C (O) OH, - (C 6 -C 50 ) arylene-C (O) OH, - (C 5 -C 50 ) heteroarylene-C (O) OH, -C (O) -NH- (C 1 -C 30 ) alkylene-OH, -
- the oligonucleotide delivery enhancing compound has a structure of
- At least one hydrogen atom contained in the oligonucleotide delivery enhancing compound is substituted with deuterium atom.
- the present disclosure provides an oligonucleotide delivery agent, comprising a delivery enhancing compound (DEC) moiety derivable from the oligonucleotide delivery enhancing compound as indicated herein and at least one oligonucleotide.
- DEC delivery enhancing compound
- the oligonucleotide delivery enhancing compound moiety is linked with the oligonucleotide via at least one linking moiety selected from the group consisting of direct bond, -O-, -S-, -C (O) -, -NH-, -N ( (C 1 -C 12 ) alkyl) -, -N ( (C 1 -C 12 ) alkyl) -C (O) -O-, -O-C (O) -, -C (O) -O-, -O-C (O) -O-, -C (O) -NH-, -OP (O) 2 O-, -P (O) (O - ) O-, -OP (O) O-, -OP (O) (S) O-, -O-S (O) 2 -O-, -S (O) 2 -O-, -S (O) -O-, - (C 1
- the oligonucleotide is selected from the group consisting of antisense oligonucleotide (ASO) , antisense RNA, short interfering RNA (siRNA) , micro-RNA (miRNA) , small activating RNA (saRNA) , double-stranded RNA (dsRNA) , and small guide RNA (sgRNA) .
- ASO antisense oligonucleotide
- siRNA short interfering RNA
- miRNA micro-RNA
- saRNA small activating RNA
- dsRNA double-stranded RNA
- sgRNA small guide RNA
- the oligonucleotide comprises at least part of the sequence as set forth in SEQ ID NO 1 to 53.
- the oligonucleotide delivery agent comprises a structure represented by Formula AA
- the delivery enhancing compound (DEC) moiety is derived from the oligonucleotide delivery enhancing compound according to any one of claims 1 to 9 and is linked to at least one oligonucleotide directly or indirectly.
- the DEC is linked with the oligonucleotide via at least one first linking moiety.
- the TM is linked with the DEC via at least one second linking moiety.
- each of the first linking moiety and the second linking moiety is independently selected from the group consisting of direct bond, -O-, -S-, -C (O) -, -NH-, -N ( (C 1 -C 12 ) alkyl) -, -N ( (C 1 -C 12 ) alkyl) -C (O) -O-, -O-C (O) -, -C (O) -O-, -O-C (O) -O-, -C (O) -NH-, -OP (O) 2 O-, -OP (O) O-, -OP (O) (S) O-, -O-S (O) 2 -O-, -S (O) 2 -O-, -S (O) -O-, - (C 1 -C 22 ) alkylene-, - (C 1 -C 22 ) alkylene-, -NH- (
- oligonucleotide is selected from the group consisting of short interfering RNA (siRNA) , small activating RNA (saRNA) , microRNA (miRNA) , antisense oligonucleotide (ASO) and small guide RNA (sgRNA) .
- siRNA short interfering RNA
- siRNA small activating RNA
- miRNA microRNA
- ASO antisense oligonucleotide
- sgRNA small guide RNA
- the targeting moiety is one or more selected from the group consisting of ligands, peptides, nucleic acids, oligonucleotides, aptamers, lipids, fatty acids, small molecules, polyethylene glycols, amino acids, cholesterols, carbohydrates, and antibodies or antibody fragments.
- the oligonucleotide delivery agent has a structure represented by any of the formulae AAI to AAXXIV:
- L represents the linking moiety
- ⁇ represents the oligonucleotide delivery enhancing compound
- the symbol adouble strand oligonucleotide in which each of the strands represents interchangeably a sense strand or an antisense strand, either symmetric or asymmetric independently on each of the ends
- the symbol represents a single strand oligonucleotide
- each of a, b and c is independently an integer from 1 to 50.
- At least one hydrogen atom contained in the delivery enhancing compound moiety, the linking moiety, the targeting moiety and/or the oligonucleotide is substituted with deuterium atom.
- the present disclosure provides a pharmaceutical composition, the composition comprising: a) the oligonucleotide delivery agent as indicated herein; and b) optionally, one or more ingredients selected from the group consisting of pharmaceutically acceptable carrier, excipient, solvent, diluent, stabilizer, dispersant, buffer, compatibilizer, preservative agent and combinations thereof.
- the present disclosure provides a method of modulating the expression of a target gene in a subject, the method comprising the step of administrating the pharmaceutical composition as indicated herein to a subject.
- the oligonucleotide or the target gene comprises at least part of the sequence as set forth in at least part of the sequence as set forth in SEQ ID NO 1 to 53.
- the pharmaceutical composition increases the expression of the target gene. In another specific embodiment, the pharmaceutical composition decreases the expression of the target gene.
- the subject is a mammal. In another specific embodiment, the mammal is a rodent. In another specific embodiment, the rodent is a mouse. In another specific embodiment, the rodent is a rat. In another specific embodiment, the mammal is a non-human primate. In another specific embodiment, the mammal is a human. In another specific embodiment, the target gene is associated with a disease or disorder.
- the target gene is associated with a disease or disorder in the central nervous system (CNS) , brain, spinal cord, liver, lung, kidney, intestine, pancreas, cholecyst, heart, lymph nodes, spleen, stomach, bladder, muscle or bone.
- the disease is cancer.
- the present disclosure provides a method of modulating the expression of a target gene, the method comprising contacting a cell with the pharmaceutical composition as indicated herein.
- the oligonucleotide or the target gene comprises at least part of the sequence as set forth in at least part of the sequence as set forth in SEQ ID NO: 1 to 53.
- the pharmaceutical composition increases the expression of the target gene. In another specific embodiment, the pharmaceutical composition decreases the expression of the target gene.
- the cell is a mammalian cell. In another specific embodiment, the mammalian cell is a mouse cell. In another specific embodiment, the mammalian cell is a rat cell. In another specific embodiment, the mammalian cell is a non-human primate cell. In another specific embodiment, the mammalian cell is a human cell. In another specific embodiment, the target gene is associated with a disease or disorder.
- the target gene is associated with a disease or disorder in the central nervous system (CNS) , brain, spinal cord, liver, lung, kidney, intestine, pancreas, cholecyst, heart, lymph nodes, spleen, stomach, bladder, muscle or bone.
- the disease is cancer.
- Standard abbreviations may be used, e.g., bp, base pair (s) ; kb, kilobase (s) ; nM, nanomolar (s) ; s or sec, second (s) ; min, minute (s) ; h or hr, hour (s) ; aa, amino acid (s) ; nt, nucleotide (s) ; i. m., intramuscular (ly) ; i. p., intraperitoneal (ly) ; s. c., subcutaneous (ly) ; ivt or IVT, intravitreal; iv or IV, tail vein, intravenous; i. c. v. or icv or ICV, intracerebroventricular and the like.
- reaction products were purchased from commercial sources and used as received unless stated otherwise. Purification of reaction products was performed with a column chromatography comprising a silica gel (200-300 mesh) and eluting agents of hexane/ethyl acetate, DCM/MeOH. Thin layer chromatography (TLC) was carried out using pre-coated silica Gel GF plates and visualized using KMnO 4 stains. 1 H-NMR spectra were recorded at 400 or 500 MHz (Varian) using CDCl 3 with TMS.
- TLC Thin layer chromatography
- High-resolution mass spectra were recorded on LC/MS (Agilent Technologies 1260 Infinity II/6120 Quadrupole) and a time-of-flight mass spectrometer by ESI or matrix assisted laser desorption/ionization (MALDI) .
- Example 1 The preparation of compound A1 of the present disclosure
- Compound A1 was prepared in this Example by using the following procedures.
- Example 2 The preparation of compound A2 of the present disclosure
- Compound A2 was prepared in this Example by using the following procedures.
- reaction mixture was stirred at room temperature for 6 h, after concentrated under reduced pressure, the resultant residue was purified by flash chromatography (silica gel, gradient eluent: 1-5%of MeOH/DCM) to provide compound A74 (4.1 g, 60%yield) as yellow solid.
- the product was characterized with mass spectrometry and 1 H NMR. MW calc.: 713.31; MW Found: 303.17 [DMT] -, 412.36 [DMT off + H] + .
- Example 3 The preparation of Compound A3 of the present disclosure.
- the Compound A3 of the present disclosure was prepared in this Example by using the following procedures.
- reaction mixture was stirred at room temperature for 6 h, after which it was concentrated under reduced pressure.
- the resultant residue was purified with flash chromatography (silica gel, gradient eluent: 1-3%of MeOH/DCM) to provide compound A89 (8.1 g, 61%yield) as yellow solid.
- the product was characterized with mass spectrometry and 1 H NMR. MW calc.: 760.48; MW. Found: 761.8 [M + H] + .
- the crude product (300 mg, 0.41 mmol, 1.0 eq) was dissolved in DCM (10 mL) , then DMAP (175 g, 1.44 mmol, 3.5 eq) , succinic anhydride (123 mg, 1.23 mmol, 3.0 eq) were added under nitrogen atmosphere.
- the reaction mixture was stirred at room temperature overnight then H 2 O (10 mL) was added into the reaction, the mixture was extracted with DCM (3*20 mL) and the organic phase was combined, dried over Na 2 SO 4 , and concentrated.
- the resultant residue was purified with flash chromatography.
- the crude product (300 mg, 0.41 mmol, 1.0 eq) was dissolved in anhydrous DCM (5 mL) then DIPEA (204 ⁇ L, 1.23 mmol, 3.0 eq) , 3- ( (chloro (diisopropylamino) phosphanyl) oxy) propanenitrile (274 ⁇ L, 1.23 mmol, 3.0 eq. ) were added under nitrogen atmosphere at 25°C. The reaction mixture was stirred for 1 h. The mixture was extracted two times with DCM, then washed with brine and dried with anhydrous Na 2 SO 4 .
- Example 5 The preparation of compound A6 of the present disclosure
- Compound A6 was prepared in this Example by using the following procedures.
- reaction mixture was stirred at room temperature for 6 h, after which it was concentrated under reduced pressure.
- the resultant residue was purified with flash chromatography (silica gel, gradient eluent: 1-5%of MeOH/DCM) to provide compound A93 (6.23 g, 74%yield) as yellow solid.
- the product was characterized with mass spectrometry, 1 H NMR and 13 C NMR. MW calc.: 741.34; MW. Found: 303.11 [DMT] -, 440.14 [DMT off + H] + .
- Example 6 The preparation of compound A7 of the present disclosure
- Compound A7 was prepared in this Example by using the following procedures.
- the resultant residue (1.0 g, 2.25 mol, 1.0 eq) was dissolved in 10 mL DCM, and Et 3 N (0.47 mL, 3.37 mmol, 1.5 eq) , DMTrCl (915 mg, 2.7 mmol, 1.2 eq) were added therein.
- the reaction mixture was stirred at room temperature for 6 h, after which it was concentrated under reduced pressure.
- the resultant residue compound was directly used in the next step without further purification.
- the product A102 was characterized with mass spectrometry. MW calc.: 746.47; MW. Found: 303.2 [DMT] - .
- the crude product (500 mg, 0.70 mmol, 1.0 eq) was dissolved in DCM (10 mL) , then DMAP (298 mg, 2.45 mmol, 3.5 eq) , succinic anhydride (140 mg, 1.40 mmol, 2.0 eq) were added therein under nitrogen atmosphere.
- the reaction mixture was stirred at room temperature overnight then H 2 O (10 mL) was added into the reaction, the mixture was extracted with DCM (3*10 mL) and the organic phase was combined, dried over Na 2 SO 4 , and concentrated.
- the resultant residue was purified with flash chromatography.
- Example 7 The preparation of compound A8 of the present disclosure
- Compound A8 was prepared in this Example by using the following procedures.
- the resultant residue (1.0 g, 2.12 mol, 1.0 eq) was dissolved in 10 mL DCM, and Et 3 N (0.44 mL, 3.17 mmol, 1.5 eq) , DMTrCl (860 mg, 2.54 mmol, 1.2 eq) were added therein.
- the reaction mixture was stirred at room temperature for 6 h, after which it was concentrated under reduced pressure.
- the resultant residue was purified with flash chromatography (silica gel, gradient eluent: 1-5%of MeOH/DCM) to provide compound A107 (1.27 g, 77%yield) as yellow solid.
- the product was characterized with mass spectrometry and 1 H NMR. MW calc.: 774.50; MW.
- the crude product (500 mg, 0.67 mmol, 1.0 eq) was dissolved in DCM (10 mL) , then DMAP (286 mg, 2.34 mmol, 3.5 eq) , succinic anhydride (134 mg, 1.34 mmol, 2.0 eq) were added therein under nitrogen atmosphere.
- the reaction mixture was stirred at room temperature overnight then H 2 O (10 mL) was added into the reaction, the mixture was extracted with DCM (3*10 mL) and the organic phase was combined, dried over Na 2 SO 4 , and concentrated.
- the resultant residue was purified with flash chromatography.
- Example 8 The preparation of compound A9 of the present disclosure
- Compound A9 was prepared in this Example by using the following procedures.
- the resultant residue (1.5 g, 3.0 mol, 1.0 eq) was dissolved in 10 mL DCM, and Et 3 N (0.625 mL, 4.5 mmol, 1.5 eq) , DMTrCl (1.2 g, 3.6 mmol, 1.2 eq) were added therein.
- the reaction mixture was stirred at room temperature for 6 h, after which it was concentrated under reduced pressure.
- the resultant residue compound was directly used in the next step without further purification.
- the product A112 was characterized with mass spectrometry. MW calc.: 802.53; MW. Found: 303.2 [DMT] - .
- the crude product (500 mg, 0.65 mmol, 1.0 eq) was dissolved in DCM (10 mL) , then DMAP (276 mg, 2.27 mmol, 3.5 eq) , succinic anhydride (129 mg, 1.29 mmol, 2.0 eq) were added therein under nitrogen atmosphere.
- the reaction mixture was stirred at room temperature overnight then H 2 O (10 mL) was added into the reaction, the mixture was extracted with DCM (3*10 mL) and the organic phase was combined, dried over Na 2 SO 4 , and concentrated.
- the resultant residue was purified with flash chromatography.
- Example 9 The preparation of compound A10 and A13 of the present disclosure
- Example 10 The preparation of compounds A11 and A15 of the present disclosure
- reaction mixture was stirred at room temperature overnight then H 2 O (10 mL) was added into the reaction, the mixture was extracted with DCM (3*10 mL) and the organic phase was combined, dried over Na 2 SO 4 , and concentrated.
- the resultant residue was purified with flash chromatography.
- the resultant residue was purified with flash chromatography (silica gel, gradient eluent: 1-10%of MeOH/DCM) to provide compound A124 (126 mg, 55%yield) as colorless oil.
- the product was characterized with mass spectrometry and 1 H NMR. MW calc.: 1204.66; MW Found: 303.2 [DMT] - .
- Example 11 The preparation of compound A12 of the present disclosure
- Compound A12 was prepared in this Example by using the following procedures.
- the reaction mixture was stirred at room temperature overnight then H 2 O (10 mL) was added into the reaction, the mixture was extracted with DCM (3*10 mL) and the organic phase was combined, dried over Na 2 SO 4 , and concentrated.
- the resultant residue was purified with flash chromatography.
- the resultant residue was purified with flash chromatography (silica gel, gradient eluent: 1-10%of MeOH/DCM) to provide compound A127 (120 mg, 45%yield) as colorless oil.
- the product was characterized with mass spectrometry and 1 H NMR. MW calc.: 917.56; MW Found: 303.2 [DMT] - .
- Example 12 The preparation of compound A14 of the present disclosure
- Compound A14 was prepared in this Example by using the following procedures.
- Example 13 The preparation of compound B1 of the present disclosure
- Compound B1 was prepared in this Example by using the following procedures.
- the compound B15 (20 g, 27.5 mmol, 1.0 eq) was added into AcOH (100 mL) under nitrogen atmosphere. The reaction mixture was stirred at 95°C for 4 h. Then saturated NaHCO 3 solution (100 mL) was added therein, the mixture was extracted 3 times with ethyl acetate, then the organic phase was combined and washed with saturated NaHCO 3 solution (3*100 mL) , dried over Na 2 SO 4 , and concentrated. The resultant residue was purified by flash chromatography (silica gel, gradient eluent: 1-50%of EA/Hexane) to provide compound B16 (8.78 g, 45%yield) as white solid.
- the compound B17 was prepared by using the start material of (2S, 3R, 4R, 5R, 6R) -3-acetamido-6- (acetoxymethyl) tetrahydro-2H-pyran-2, 4, 5-triyl triacetate.
- Example 14 The preparation of compound B2 of the present disclosure
- Compound B2 was prepared in this Example by using the following procedures.
- This step comprises the preparation of compound B38 from methyl 4-fluoro-3-nitrobenzoate.
- the resultant residue (compound B40) was directly used in the next step without further purification.
- the compound B40 was characterized with mass spectrometry. MW calc.: 525.27; MW. Found: 526.59 [M+H] + .
- Controlled Pore Glass CPG
- DIPEA N, N-Diisopropylethylamine
- HBTU O- (1H-Benzotriazol-1-yl) -N, N, N′, N′-tetramethyluronium hexafluorophosphate
- Example 15 The preparation of Compound B3 of the present disclosure.
- the Compound B3 of the present disclosure was prepared in this Example by using the following procedures.
- the compound B39 was originated from the steps (1) - (2) of Example 14.
- Example 16 The preparation of Compound B4 of the present disclosure.
- the Compound B4 of the present disclosure was prepared in this Example by using the following procedures.
- Compound B17 was originated from the step (6) of Example 13.
- Example 17 The preparation of Compound B5 of the present disclosure.
- the Compound B5 of the present disclosure was prepared in this Example by using the following procedures.
- Compound B46 was originated from the steps (3) - (5) of Example 15.
- Example 18 The preparation of compound B6 of the present disclosure.
- the compound B6 of the present disclosure was prepared in this Example by using the following procedures.
- Example 19 The preparation of compound tC2 of the present disclosure.
- the dicarboxylic acid (20 g, 86.8 mmol) was dissolved/suspended in dry CH 2 Cl 2 (100 mL) . Then oxalyl chloride (16.2 mL, 190.96 mmol) and DMF (5 drops) were added to the solution. The reaction mixture was stirred at room temperature for 3 h, then concentrated under reduced pressure to provide crude compound C2, which was directly used in the next step without further purification.
- CPG Controlled Pore Glass
- DIPEA N, N-Diisopropylethylamine
- HBTU 208 mg, 0.55 mmol, 2.0 eq
- Compound D1 was prepared in this Example by using the following procedures.
- the compound 39 (2.3 g, 4.18 mmol, 1.0 eq) and Diisoropyl ammonium tetrazolide (2.15 g, 12.54 mmol, 3.0 eq) were dissolved in anhydrous DCM (40 mL) under nitrogen atmosphere was added 3- ( (Bis (diisopropylamino) phosphino) oxy) propanenitrile (3.78 g, 12.54 mmol, 3.0 eq) at room temperature. The reaction mixture was stirred for 6 h. The mixture was extracted two times with DCM, then washed with brine and dried with anhydrous Na 2 SO 4 .
- Example 21 The preparation of compound D2 of the present disclosure
- Compound D2 was prepared in this Example by using the following procedures.
- the compound 40 (1.1 g, 1.67 mmol, 1.0 eq) and Diisoropyl ammonium tetrazolide (858 mg, 5.01 mmol, 3.0 eq) were dissolved in anhydrous DCM (15 mL) under nitrogen atmosphere was added 3- ( (Bis (diisopropylamino) phosphino) oxy) propanenitrile (1.51 g, 5.01 mmol, 3.0 eq) at room temperature. The reaction mixture was stirred for 6 h. The mixture was extracted two times with DCM, then washed with brine and dried with anhydrous Na 2 SO 4 .
- Example 22 The preparation of compound D3 of the present disclosure
- Compound D3 was prepared in this Example by using the following procedures.
- the compound 41 (2 g, 3.63 mmol, 1.0 eq) and Diisoropyl ammonium tetrazolide (1.86 g, 10.89 mmol, 3.0 eq) were dissolved in anhydrous DCM (20 mL) under nitrogen atmosphere was added 3- ( (Bis (diisopropylamino) phosphino) oxy) propanenitrile (3.28 g, 10.89 mmol, 3.0 eq) at room temperature. The reaction mixture was stirred for 6 h. The mixture was extracted two times with DCM, then washed with brine and dried with anhydrous Na 2 SO 4 .
- Example 23 The preparation of compound D4 of the present disclosure
- Compound D4 was prepared in this Example by using the following procedures.
- the compound 22 (2 g, 3.14 mmol, 1.0 eq) and Diisoropyl ammonium tetrazolide (1.61 g, 9.43 mmol, 3.0 eq) were dissolved in anhydrous DCM (20 mL) under nitrogen atmosphere was added 3- ( (Bis (diisopropylamino) phosphino) oxy) propanenitrile (2.85 g, 9.43 mmol, 3.0 eq) at room temperature. The reaction mixture was stirred for 6 h. The mixture was extracted two times with DCM, then washed with brine and dried with anhydrous Na 2 SO 4 .
- Example 24 The preparation of compound D5 of the present disclosure
- Compound D5 was prepared in this Example by using the following procedures.
- the resultant residue was dissolved in 10 mL THF, then 1 M TBAF THF solution (3.39 mL, 3.39 mmol, 1.5 eq) was added under nitrogen atmosphere. The reaction mixture was stirred at room temperature for 2 h, then H 2 O (20 mL) was added. The mixture was extracted three times by ethyl acetate, then washed one time by brine, dried by anhydrous Na 2 SO 4 and concentrated under reduced pressure. The resultant residue was purified with flash chromatography (silica gel, gradient eluent: 1-5%of MeOH/DCM) to provide compound 43 (0.8 g, 46%yield) . The product was characterized with mass spectrometry and 1 H NMR.
- the compound 43 (800 mg, 1.05 mmol, 1.0 eq) and Diisoropyl ammonium tetrazolide (360 mg, 2.1 mmol, 2.0 eq) were dissolved in anhydrous DCM (10 mL) under nitrogen atmosphere was added 3- ( (Bis (diisopropylamino) phosphino) oxy) propanenitrile (634 mg, 2.1 mmol, 2.0 eq) at room temperature. The reaction mixture was stirred for 6 h. The mixture was extracted two times with DCM, then washed with brine and dried with anhydrous Na 2 SO 4 .
- Compound D6 was prepared in this Example by using the following procedures.
- Example 26 The preparation of compound D7 of the present disclosure
- Compound D7 was prepared in this Example by using the following procedures.
- Compound F69 was prepared in this example by using the following procedures.
- the resultant residue compound was purified with flash chromatography (silica gel, gradient eluent: 10-40%of ethyl acetate/petroleum ether) to provide compound 47 (1.5 g, 22%yield) as light-yellow oil.
- the product was characterized with mass spectrometry and 1 H NMR. MW calc.: 598.99; MW. Found: 599.72 [M + H] + .
- Oligonucleotide sequences used for cell or animal treatments in the following examples are listed in Table 1.
- Example 29 Characterization of the in vitro knockdown activity of DEC-conjugated siRNAs for mouse FVII gene.
- PMH cells were transfected with each of the indicated DCOs (i.e., RD-12339, RD-12585, RD-12586, RD-12710, RD-12711 and RD-12712) at 0.1 nM and 1 nM with Lipofectamine TM RNAiMAX (Thermofisher) for 24 hours according to the manufacturer’s instructions was shown in FIG. 1.
- DCOs i.e., RD-12339, RD-12585, RD-12586, RD-12710, RD-12711 and RD-12712
- Mock treatment was transfection in the absence of oligonucleotide.
- dsCon2 served as a non-specific duplex control.
- Mouse FVII mRNA levels were quantified by RT-qPCR using a gene specific primer set.
- Tbp was amplified as an internal reference for RNA loading. The mean expression values of FVII mRNA relative to Mock treatment are normalized to Tbp.
- transfection of RD-12339, RD-12585, RD-12586, RD-12710, RD-12711 and RD-12712 reduced FVII mRNA expression by 94%, 85%, 91%, 80%, 78%and 84%at 0.1 nM and 98%, 96%, 97%, 89%, 91%and 89%at 1 nM treatment, respectively. All tested DCOs achieved comparable knockdown activities in a dose-dependent manner with RD-12339, RD-12585 and RD-12586 having slightly greater maximal activities.
- RD-12339, RD-12585, RD-12586, RD-12710, RD-12711 and RD-12712 were added to the culture media of PMH cells at escalating concentrations (i.e., 0.01, 0.05, 0.20, 0.78, 3.13, 12.50, 50 and 200 nM) for 24 hours.
- concentrations i.e., 0.01, 0.05, 0.20, 0.78, 3.13, 12.50, 50 and 200 nM
- Example 30 Characterization of in vivo knockdown activity of DEC-conjugated siRNAs for mouse FVII gene expression.
- DCOs i.e., RD-12339, RD-12585, RD-12586, RD-12710, RD-12711 and RD-12712
- mice were sacrificed on 3 days post dosing and FVII mRNA level was quantified in liver tissue after RNA isolation and RT reaction via RT-qPCR using gene specific primer sets.
- RD-12339, RD-12586, RD-12710 and RD-12712 exhibited over 60%knockdown activity with RD-12339 having the greatest knockdown activity.
- RD-12339, RD-12585, RD-12586, RD-12710, RD-12711 and RD-12712 caused an 81%, 82%, 89%, 80%, 81%and 89%knockdown of FVII mRNA.
- FVII protein expression levels in the plasma of the treated mice were detected by ELISA assay.
- 3 DCOs i.e., RD-12339, RD-12585 and RD-12586
- RD-12339, RD-12585 and RD-12586 at medium dose (1 mg/kg) reduced FVII protein expression by 67%, 51%and 65%, respectively.
- knockdown activity was similar for all tested DCOs with RD-12339, RD-12585 and RD-12586 providing 89%, 87%and 93%reductions in FVII protein levels, respectively.
- Example 31 Potent and durable knockdown of DEC-conjugated siRNAs on mouse FVII protein in plasma of C57BL/6J mice
- the indicated DCOs i.e., RD-12710 and RD-12712
- RD-11706 was injected at 3 mg/kg and served as a control.
- Saline was injected as a vehicle control to establish the baseline of mFVII protein expression.
- Mouse plasmas were collected on 10, 31, 54, 61, 80 and 89 days post dosing and mFVII protein level was quantified in mouse plasma by ELISA assay. The results of potent and durable knockdown of DEC-siRNAs on mouse FVII protein in plasma are shown in FIG. 5.
- Example 32 In vitro knockdown activity of DEC-siRNA in PMH cells via free uptake.
- exemplary DEC-siRNAs i.e., RD-13110, RD-13115, and RD-13118
- PMH cells were treated at escalating concentrations (i.e., 1.56, 6.25, 25, 100, 400 and 1600 nM) in absence of any additional delivery system (i.e., free uptake) for 72 hours.
- Sod1 levels were assessed via RT-qPCR to generate dose response curves and estimate potency.
- potency dotted line
- Example 33 In vivo knockdown of DEC-siRNA on Sod1 mRNA level in CNS tissues
- mice were sacrificed on 7 days post dosing and CNS tissues from the brain (i.e., frontal cortex, cerebellum, and cerebrum) , spinal cord (i.e., cervical, thoracic, and lumbar) , and periphery (i.e., liver) were harvested for mRNA expression analysis via RT-qPCR.
- CNS tissues i.e., frontal cortex, cerebellum, and cerebrum
- spinal cord i.e., cervical, thoracic, and lumbar
- periphery i.e., liver
- RD-13115 and RD-13118 had improved knockdown across all tissues at the 200 ⁇ g dose indicating the C5x5 conjugates had better in vivo potency via ICV injection compared to the non-conjugated control (i.e., RD-12556) and C5x1 variant RD-13110.
- Analysis in periphery tissue (i.e., liver) also revealed that activity of the C5x5 conjugates (i.e., RD-13115 and RD-13118) were not well retained within the CNS in which systemic exposure via CNS drainage provided knockdown in the liver at levels similar to the CNS tissues.
- both RD-12556 and RD-13110 activity was selectively enriched across the CNS providing only an approximate 11%and 3%knockdown in the liver, respectively.
- the C5x5 conjugates have improved activity and biodistribution across all CNS tissues.
- C5x1 would be more ideal compared to other lipids (e.g., C5x5) .
- Example 34 In vivo knockdown of DEC-siRNA on Sod1 mRNA level following systemic administration
- mice were administered with DEC-siRNAs (i.e., RD-13110, RD-13115 and RD-13118) or non-conjugate control (i.e., RD-12556) at a 20 mg/kg dose via IV injection.
- Mice were sacrificed on day 7 after treatment and Sod1 knockdown was quantified via RT-qPCR in select organs (i.e., heart, liver, spleen, lung, kidney, and bladder) .
- RD-12556 only provided substantial knockdown in the kidney reducing Sod1 levels by 56%, whereas RD-13110 also had activity selectively enriched in the spleen.
- RD-13115 and RD-13118 both had broad activity across all tissues except bladder in which only RD-13118 was capable of reducing Sod1 levels by roughly 21%.
- C5x1 conjugation may have unique delivery functions via systemic administration offering selective targeting to the spleen, whereas C5x5 can provide knockdown to a broader spectrum of tissues.
- inclusion of ACO in RD-13118 was the only DEC-siRNA to provide measurable knockdown in the bladder indicating combining the DEC technology with ODV-siRNA may also have further delivery benefits when used in combination in vivo.
- Example 35 In vivo knockdown of DEC-siSOD1 on Sod1 mRNA level in skeletal muscle
- mice were treated with DEC-siRNAs (i.e., RD-13110, RD-13115 and RD-13118) or non-conjugate control (i.e., RD-12556) at a 20 mg/kg dose via IV injection.
- Mice were sacrificed on 7 days post dosing and Sod1 mRNA level was quantified via RT-qPCR in muscle tissues (i.e., bicep, semitendinosus, platysma and gluteus) .
- RD-12556 provided no significant knockdown in any muscle tissue compared to the saline treatment group.
- RD-13110 had modest activity in select tissues yet did not exceed 25%reductions in Sod1 mRNA levels at the 20 mg/kg dose.
- RD-13115 reduced Sod1 mRNA levels by 82%, 84%, 78%, and 76%in bicep, semitendinosus, platysma and gluteus tissues, respectively.
- RD-13118 reduced Sod1 mRNA levels by 76%, 84%, 61%, and 67%in bicep, semitendinosus, platysma and gluteus tissues, respectively.
- DEC conjugation provides knockdown in muscle tissue via IV injection; however, C5x5 conjugates (i.e., RD-13115 and RD-13118) enabled siRNA knockdown at a 20 mg/kg dose that has been otherwise unobtainable by conventional delivery technologies (i.e., LNP and GalNAc) .
- C5x5 conjugates i.e., RD-13115 and RD-13118
- siRNA knockdown at a 20 mg/kg dose that has been otherwise unobtainable by conventional delivery technologies (i.e., LNP and GalNAc) .
- Example 36 In vivo knockdown activity of DEC-siSOD1 on Sod1 mRNA level in retinal tissue
- DEC-siRNAs i.e., RD-13115 and RD-13118
- non-conjugate control i.e., RD-12556
- Rats were sacrificed on 14 days post dosing and Sod1 mRNA level was quantified via RT-qPCR in retinal tissue.
- RD-12556 provided only a 39%knockdown
- RD-13115 and RD-13118 reduced Sod1 mRNA levels by 74%and 71%, respectively.
- DEC-siRNA i.e., RD-13115 and RD-13118
- RD-12556 non-conjugated control
- Example 37 Pharmacokinetics of DEC-saRNA in bladder tissue from adult C57BL/6J mice following DEC-saRNA treatment via IVB instillation
- RD-13520 (DEC-saRNA) and RD-10773 (non-DEC-saRNA) via IVB instillation and bladder tissues were harvested on 2-, 6-, 12-hour, day-1 and day-4 after treatment.
- RD-10773 and RD-13520 were detected in the bladder tissue preps by stem-loop RT-qPCR.
- the concentrations of RD-13520 and RD-10773 in bladder were plotted in FIG. 11 to evaluate pharmacokinetics (PK) and summarized in Table 4.
- Example 38 Potent and durable knockdown of mouse Sod1 mRNA by DEC-conjugated siRNAs in C57BL/6J mice
- RD-15135 served as a non-conjugate control.
- RD-15136 was a lipid (i.e., C16) conjugated siRNA.
- RD-15137 with a typical DEC structure i.e., C5X5 served as a typical DEC-siRNA.
- FIG. 12A shows the body weight change of C57BL/6J mice out to day 28 post treatment.
- Sod1 mRNA expressions on 14 and 28 days post dosing in tissues from periphery i.e., heart, liver, kidney, fat tissues, pancreas, diaphragm
- tissues from periphery i.e., heart, liver, kidney, fat tissues, pancreas, diaphragm
- blood vessel i.e., thoracic aorta with vein
- skeletal muscle i.e., bicep, semitendinosus, platysma and gluteus
- Example 39 Potent and durable knockdown of rat Sod1 mRNA in SD rats by DEC-conjugated siRNAs
- RD-15135 served as a non-conjugate control.
- RD-15136 was a lipid (i.e., C16) conjugated siRNA.
- RD-15137 with a typical DEC structure i.e., C5X5 served as a typical DEC-siRNA.
- aCSF alone was used as a vehicle control to establish baseline expression.
- FIG. 13A shows the body weight change of SD rats out to day 28 post treatment.
- Sod1 mRNA expressions on 14 days post dosing in tissues from brain i.e., frontal cortex, cerebellum and cerebrum
- spinal cord i.e., cervical, thoracic and lumbar
- periphery i.e., liver and kidney
- FIG. 13B Sod1 mRNA on 28 days post dosing in tissues from brain (i.e., frontal cortex, cerebellum and brainstem) , spinal cord (i.e., cervical, thoracic and lumbar) and periphery (i.e., liver and kidney) were shown in FIG. 13C.
- Example 40 Delivery enhancing compound (DEC) of siRNA-ACO provides consistent and potent knockdown on SOD1 mRNA in SK-N-AS and T98G cells.
- DEC-siRNA-ACOs i.e., RD-16149, RD-16099, RD-16100, RD-16101, RD-16150, RD-16103, RD-16104 and RD-16105
- concentrations i.e., 1.56, 6.25, 25, 100, 400 and 1600 nM
- RD-16106 was transfected and served as a siRNA-ACO control. Mock treatments were transfected in absence of oligonucleotide (not shown) .
- SOD1 mRNA levels were quantified via RT-qPCR using a gene specific primer set.
- the remaining human SOD1 mRNA levels in SK-N-AS and T98G cells were plotted in FIG. 14A-14B.
- the EC 50 values were summarized in Table 5.
- Example 41 Knockdown activity and tissue concentration of siRNAs via ICV injection in adult hSOD1 G93A mice.
- siRNAs i.e., RD-14851, RD-12500, RD-16145 and RD-16334.
- RD-16334 with a DEC structure i.e., L17
- Treatment with aCSF alone was used as a vehicle control to establish baseline expression.
- FIG. 15A shows the remaining SOD1 mRNA on 30 days post dosing as quantified in tissues from brain (i.e., frontal cortex, cerebellum and cerebrum) , spinal cord and periphery (i.e., liver) via RT-qPCR using a gene specific primer set.
- FIG. 15B shows the concentration on 30 days post dosing as in tissues from brain (i.e., frontal cortex, cerebellum and cerebrum) , spinal cord and periphery (i.e., liver) via stem-loop RT-qPCR using a gene specific primer set.
- Serum chemistry i.e., glutamic-pyruvic transaminase (ALT) , glutamic oxalacetic transaminase (AST) , total bilirubin (TBIL) and creatinine (CRE)
- completed blood count i.e., white blood cell (WBC) , red blood cell (RBC) , blood platelet (PLT) etc.
- Example 42 Splicing activity of “saRNA-ASO” DEC-DAO structure in GM03813 cells.
- DEC-DAO DEC-DAO linkage with L21 linker (i.e., RD-16939 and RD-16940) , DEC-saSMN2 (i.e., RD-16424) and DEC-antisense oligonucleotide (DEC-ASO) (i.e., RD-14644) were transfected into GM03813 cells at indicated concentrations (i.e., 0.1, 1 and 10 nM) for 3 days.
- RD-16381 was transfected as a non-specific DEC control.
- Combo treatment i.e., RD-16424+RD-14644
- combo treatment control i.e., RD-16381+RD-16424, RD-16381+RD-14644
- concentrations i.e., 0.1, 1 and 10 nM
- saRNA-ASO DEC-DAO structure provides potent splicing activity on mRNA levels of SMN2FL and SMN2 ⁇ 7, indicating that combining a saRNA with a ASO into a DAO can largely retain and even increase the activity of both saRNA and ASO units.
- Example 43 Effect of “saRNA-siRNA” DEC-DAO structure targeting two different human genes (SMN2 and SOD1) on the expression of SMN2 (SMN2FL and SMN2 ⁇ 7) and pOD1 in GM03813 cells.
- DEC-DAO linkage with L21 linker i.e., RD-16941
- DEC-saSMN2 i.e., RD-16424
- DEC-siSOD1 i.e., RD-13115
- RD-16381 was transfected as a non-specific DEC control.
- FIGs. 17A-17B shows mRNA levels of SMN2FL and SMN2 ⁇ 7 and FIG. 17C shows remaining SOD1 mRNA levels.
- “saRNA-siRNA” DEC-DAO structure provides the potent gene induction on expression of SMN2FL and knockdown on SOD1 mRNA, respectively. This result indicates that combining a saRNA with a siRNA into a DAO can largely retain the activity for each unit and enhance the activity of the saRNA unit.
- Example 44 Effect of “siRNA-siRNA” DEC-DAO structure targeting two different genes (mouse Sod1 and Ppig) on the expression of mouse Sod1 and Ppig in C2C12 cells.
- DEC-DAO structure targeting two different genes was transfected into C2C12 cells at indicated concentrations (i.e., 0.01, 0.1 and 1 nM) for 24 hours.
- RD-16381 was transfected as a non-specific DEC control.
- Combo treatment i.e., RD-13115+RD-14672
- combo treatment control i.e., RD-16381+RD-13115, RD-16381+RD-14672
- concentrations i.e., 0.01, 0.1 and 1 nM
- siRNA-siRNA DEC-DAO structure provides potent knockdown activity on mouse Sod1 and Ppig mRNA levels, respectively. This result indicates that combining two siRNAs targeting different genes into a DAO can largely retain the activity for each siRNA unit and even enhance the activity of both siRNA units.
- Example 45 Effect of “siRNA-siRNA” DEC-DAO structure targeting two different genes (mouse Sod1 and Ppig) on the expression of mouse Sod1 and Ppig in vivo.
- Remaining mouse Sod1 mRNA on 14 days post dosing were quantified in tissues from periphery (i.e., heart, liver, kidney, fat tissues, diaphragm, thoracic aorta with vein) , and skeletal muscle (i.e., bicep, semitendinosus, platysma and gluteus) via RT-qPCR using a gene specific primer set.
- periphery i.e., heart, liver, kidney, fat tissues, diaphragm, thoracic aorta with vein
- skeletal muscle i.e., bicep, semitendinosus, platysma and gluteus
- Example 46 Pharmacokinetics of DEC-saRNA in retina and vitreous humor of adult SD rat following DEC-saRNA treatment via IVT injection
- DEC-saRNA i.e., RD-16447
- non-conjugated saRNA i.e., RD-12173
- the rats were sacrificed on 1-hour, day-1, -3, -7, -14 and -28 following treatment and the concentrations of RD-16447 and RD-12173 were quantified in retina and vitreous humor via stem-loop RT-qPCR. Concentrations of RD-16447 and RD-12173 in retina and vitreous humor were plotted in FIG. 20A-20B. Mean concentrations of oligonucleotide in retina and vitreous humor were summarized in Table 7.
- Example 47 Knockdown activity of DEC-siRNA targeting SOD1 in T98G cells.
- a new DEC structure (i.e., C5x34) was designed and conjugated to a siRNA targeting SOD1 gene, resulting in a new DEC-siRNA (i.e., RD-17138) .
- the indicated DEC-siRNA i.e., RD-17138
- T98G cell 0.1 nM and 1 nM for 24 hours.
- RD-11566 was transfected and served as a non-specific duplex control.
- Mock treatments were transfected in absence of oligonucleotide.
- SOD1 mRNA levels were quantified via RT-qPCR. As shown in FIG.
- RD-17138 showed a knockdown activity on SOD1 mRNA expression greater than 90%at 0.1 nM and 1 nM, suggesting the new DEC structure C5x34 can enhance the knockdown activity of siRNA on SOD1 mRNA level in vitro.
- oligonucleotides used in the following examples were synthesized via the following general method.
- the single strand oligonucleotide was synthesized on a K&ADNA synthesizer (K&A Laborgeraete GbR, chaafheim, Germany) by a solid phase synthesis technique.
- the starting material was universal solid support or special solid support commercially available or synthesis as disclosure in previous context.
- phosphoramidite monomers including various linkers and conjugates were added sequentially onto a solid support in the DNA synthesizer to generate the desired full-length oligonucleotides.
- each cycle of amidite addition consisted of four chemical reactions including detritylation, coupling, oxidation/thiolation and capping.
- detritylation was performed by using 3%dichloroacetic acid (TCA) in DCM for 45 seconds.
- the second step Phosphoramidite coupling was conducted for 6 minutes for all amidites by 12 eq.;
- oxidation was performed by using 0.02 M iodine in THF: pyridine: water (70: 20: 10, v/v/v) for 1 minute; if phosphorothioate modification needed then replace oxidation by thiolation which was carried out with 0.1 M solution of xanthane hydride in pyridine: ACN (50: 50, v/v) for 3 minutes;
- the capping was performed by using a THF: acetic anhydride: Pyridine (80: 10: 10, v/v/v) (CAP A) and N-methylimidazole: THF (10: 90, v/v) , (CAP B) for 20 seconds.
- the Cycles of four chemical reaction will be depended by the length of single of oligonucleotide.
- Deprotection I (Nucleobase Deprotection): After completion of the synthesis, the solid support was transferred to a screw-cap microcentrifuge tube. For a 1 ⁇ mol synthesis scale, 1 ml of a mixture of methylamine and ammonium hydroxide was added. The tube containing the solid support was then heated in an oven at 60°C to 65°C for 15 min and then allowed to cool to room temperature. The cleavage solution was collected and evaporated to dryness in a speedvac to provide crude single strand of oligonucleotide.
- Deprotection II Removal of 2’-TBDMS Group: If The crude RNA oligonucleotide, still carrying the 2’-TBDMS groups, then dissolved in 0.1 ml of DMSO. After adding 1 ml of Triethylamine trihydrofluoride, the tube was capped, and the mixture was shaken vigorously to ensure complete dissolution and then heated in an oven at 65°C for 15 minutes. The tube was removed from the oven and cooled down to room temperature. The solution containing the completely desilylated oligonucleotide was cooled on dry ice.
- oligonucleotides The purification of oligonucleotides was performed on an AKTA explorer 10 equipped with a Source 15Q 4.6/100 PE column using the following conditions: buffer A: (10 mM Tris-HCl, 1 mM EDTA, pH 7.5) , B: (10 mM Tris-HCl, 1 mM EDTA, 2M NaCl, pH 7.5) , gradient: 10%B to 60%B in 25 min, flow rate: 1 ml/min.
- buffer A (10 mM Tris-HCl, 1 mM EDTA, pH 7.5
- B (10 mM Tris-HCl, 1 mM EDTA, 2M NaCl, pH 7.5)
- gradient 10%B to 60%B in 25 min
- flow rate 1 ml/min.
- the pure oligonucleotides were collected and desalting by a HiPrep 26/10 Desalting column.
- sense strand passenger strand
- antisense strand guide strand
- PMH Primary mouse hepatocytes
- T98G cells (Cobioer, Cat#CBP60301) were cultured under the conditions of 5%CO2 and 37°C in modified MEM medium (Gibco, Thermo Fisher Scientific, Carlsbad, CA) supplemented with 10%bovine calf serum (Sigma-Aldrich) and 1%penicillin/streptomycin.
- modified MEM medium Gibco, Thermo Fisher Scientific, Carlsbad, CA
- 10%bovine calf serum Sigma-Aldrich
- SK-N-AS Procell, Wuhan, China, Cat#CL-0621
- C2C12 CBP60252, Cobioer, China
- SMA patient derived fibroblasts GM03813 cells were obtained from Coriell Institute (Camden, NJ, USA) and cultured at 5%CO2 and 37°C in modified MEM medium supplemented with 15%bovine calf serum, 1%NEAA and 1%penicillin/streptomycin. Transfection was carried out using Lipofectamine RNAiMax (ThermoFisher, Waltham, MA, USA) in growth media without antibiotics according to the manufacturer’s protocol. Free uptake was carried out by directly adding oligonucleotides into culture medium containing the PMH cells.
- C57BL/6J mice (B204, Beijing, China) were anesthetized with isoflurane and perfused by initial flushing reagent and digestion reagent successively.
- the liver was placed into a 10 cm dish and torn apart using forceps in culture medium.
- the cell suspension was collected by filtering through a 70-75-micron membrane in 50 mL conical tube, followed by centrifuging at 4°C for 2 minutes at 100 ⁇ g in a swinging-arm centrifuge. Cells were washed with 20 mL cold PBS after removing the supernatant (Repeat this step twice) . Cells with at least 80%viability were allowed to proceed with the assay. Appropriate number of cells were seeded to the cell culture plates which were pre-coated with collagen I 4-12 hrs in advance to yield a final confluence of 90-95%prior to the start of the assay.
- mice All animal procedures were conducted by certified laboratory personnel using protocols consistent with local and state regulations and approved by the Institutional Animal Care and Use Committee.
- C57BL/6J mice (4-6 weeks old) purchased from SPF Biotechnology Co., LTD (B204, Beijing, China) .
- Sprague-Dawley female rats (A102, SPF, China) were purchased from SPF (Suzhou) Biotechnology Co., LTD.
- Parental transgenic hSOD1 G93A mice (Strain ID #004435) were purchased from the Jackson Laboratory (Bar Harbor, ME, USA) and imported into China via Nantong University (Nantong City, Jiangsu province, China) .
- Formulations for in vivo studies were prepared fresh prior to use by dissolving aliquots of lyophilized oligonucleotide into saline or aCSF to create stock solutions for dilution to the intended treatment concentrations. Animals were randomly allocated into study groups based on body weight and sex.
- Avertin (1.2%) was prepared fresh and sterilized via 0.2-micron filter. Mice were dosed at 0.30-0.35 ml per 10 g body weight via intraperitoneal (IP) injection in a stereotaxic apparatus to rapidly induce anesthesia for up to 30 minutes. An approximate 11.5 mm incision was made in the animal’s scalp and a 25-gauge needle attached to a Hamilton syringe containing the appropriate siRNA formulation was placed at bregma level. The needle was moved to the appropriate anterior/posterior and medial/lateral coordinates (0.2 mm anterior/posterior and 1 mm to the right medial/lateral) . A total of 10 ⁇ L was injected into the lateral ventricle at an approximate rate of 1 ⁇ L/s. Following treatment, the needle was slowly withdrawn, and the wound sutured close.
- IP intraperitoneal
- Anesthesia was administered via 3.0%isoflurane in an induction chamber for continuous 10 mins. Hair was shaved around the injection site at the base of the tail and cleaned with 75%ethanol. The space between the L5-L6 spinous processes was identified and a 30-gauge needle attached to a microliter syringe containing the appropriate drug formulations was slowly inserted into the intradural space until a tail flick was observed. The needle position was subsequently secured in which 30 ⁇ L total volume of solution was injected over the course of 1 min.
- mice were exposed to an infrared lamp for 2-3 min to dilate the veins, and then held in the restrainer to straighten the tail.
- the tail was wiped with 75%alcohol and the needle was inserted 2 to 4 mm parallel to the tail vein into the lumen, keeping the bevel of the needle upwards.
- the preformed solution was injected slowly and should be free of resistance if administered correctly.
- the recommended injection volume for test article is 200 ⁇ L and the injection rate don’t exceed 5 ml/min.
- the injection site is pressed firmly with a cotton swab or finger to prevent backflow of the administration solution and/or blood.
- IVTT Intravitreal
- SD rats were housed in animal facility of Ractigen (Nantong, Jiangsu, China) and fed for at least three days prior to the intravitreal (IVT) injection of compounds.
- SD rats were anesthetized in an isoflurane (RWD, R510-22-16) induction chamber (5%isoflurane in 100%medical oxygen, 2 L/min) until they had no response to toe pinches.
- SD rats were transferred to the experimental operating platform and positioned for delivery of isoflurane (2%isoflurane in 100%medical oxygen, 1.5 L/min) using a homemade face mask during the procedure of IVT injection. Before the IVT injection of compounds, one drop of 0.5%alcaine as topical anesthetics was applied to the injected eye (left eye) .
- An anterior chamber paracentesis was performed using a 30-gauge needle, followed by approximately 5 ⁇ L aqueous humor were outflowed.
- Each compound for corresponding group was dissolved in 4 ⁇ L normal saline and loaded into a 30-gauge needle for IVT injection.
- the compounds were administered by inserting at a 45° angle of the needle through the sclera into the vitreous body, then injected into the posterior chamber keeping for 5 seconds to avoid the leaking.
- the injected eye was administered with antibiotics to prevent infection after the IVT injection.
- IVB Intravesical bladder
- mice were anesthetized and placed on a temperature-controlled pad at 28-31°C. Each mouse will be excreted urine from the bladder by urinary catheterization and to wash the bladder with saline before IVB instillation. A 2 cm catheter attached to a 1 ml syringe containing compound solution was intubated into bladder via urinary meatus. A total of 50 ⁇ L solution was injected into the bladder through an indwelling urinary catheterization at ⁇ 1.5 hours.
Landscapes
- Chemical & Material Sciences (AREA)
- Organic Chemistry (AREA)
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Engineering & Computer Science (AREA)
- General Health & Medical Sciences (AREA)
- Genetics & Genomics (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Molecular Biology (AREA)
- Biochemistry (AREA)
- Medicinal Chemistry (AREA)
- Biotechnology (AREA)
- Pharmacology & Pharmacy (AREA)
- Biomedical Technology (AREA)
- Animal Behavior & Ethology (AREA)
- Public Health (AREA)
- Veterinary Medicine (AREA)
- Epidemiology (AREA)
- General Engineering & Computer Science (AREA)
- Wood Science & Technology (AREA)
- Zoology (AREA)
- Biophysics (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Chemical Kinetics & Catalysis (AREA)
- Obesity (AREA)
- Plant Pathology (AREA)
- Microbiology (AREA)
- Physics & Mathematics (AREA)
- General Chemical & Material Sciences (AREA)
- Hematology (AREA)
- Diabetes (AREA)
- Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
- Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
Abstract
Disclosed is a compound having a specific structure, which can substantially enhance the delivery efficiency of oligonucleotides to various subjects both in vitro and in vivo, and thus achieving improved expression regulation of target genes. A pharmaceutical composition comprising the compound and a method of modulating the expression of a target gene by using the same are also disclosed.
Description
The present disclosure relates to the technical field of genetic modulation, and in particular to unique compounds useful for enhancing delivery efficiency of oligonucleotides both in vitro and in vivo.
Oligonucleotide-based therapeutics has been an emerging technology as its mechanisms of action provide expression regulation on almost unlimited target genes. The administration of oligonucleotides to patients, organs, tissues or cells may elicit or impact various biochemical reactions and thus achieve functions such as silencing, inhibiting, activating, and modulation of gene expression. The opportunity to use oligonucleotide-based therapy holds significant promise, providing solutions to medical problems that could not be addressed with traditional medicines. However, one of the major obstacles for the commercialization of oligonucleotide-based medicine is a lack of effective methods for delivering it to the right organ, tissue, or cell. Oligonucleotide therapeutics typically have a molecular weight ranging from 7 kDa to 14 kDa and comprise strong negative charge due to the inclusion of phosphodiester or phosphorothioate in the backbone thereof. The relatively large molecular weight and the high negative charge density restrain the transmission of oligonucleotides across cell membranes. Many strategies have been exploited for promoting the oligonucleotide delivery and yet, the delivery efficiency remains challenging.
Therefore, there continues to be a longstanding need for development of unique technologies that can deliver oligonucleotides with superior efficiency and to a wider range of organs, tissues and cells, especially to organs, tissues and cells where oligonucleotides are barely attainable in the prior art.
After persistent exploration, we have developed unique delivery enhancing compounds which can achieve the above targets.
The present disclosure provides an oligonucleotide delivery enhancing compound (DEC) comprising a nitrogen-containing five membered heterocyclic ring moiety, the DEC can be directly or indirectly attached to an oligonucleotide so as to be used to enhance the delivery efficiency of oligonucleotides to a subject both in vitro and in vivo. In a specific embodiment, the DEC has a structure represented by Formula AI or Formula AII
In another specific embodiment, the oligonucleotide delivery enhancing compound comprises a moiety represented by Formula BI
In another specific embodiment, the oligonucleotide delivery enhancing compound a structure represented by Formula BII
Also provided herein is an oligonucleotide delivery agent comprising a DEC moiety derived from the DEC of the present disclosure and at least one oligonucleotide, wherein the DEC moiety is directly or indirectly linked to the oligonucleotide via at least one linking moiety. The oligonucleotide can be antisense oligonucleotide (ASO) , antisense RNA, short interfering RNA (siRNA) , micro-RNA (miRNA) , small activating RNA (saRNA) , double-stranded RNA (dsRNA) , and small guide RNA (sgRNA) .
Also provided herein is a pharmaceutical composition comprising the DEC of the
present disclosure, the pharmaceutical composition may optionally comprise additional ingredients, such as pharmaceutically acceptable carrier, excipient, solvent, diluent, stabilizer, dispersant, buffer, compatibilizer, preservative agent and combinations thereof.
Also provided herein is method of modulating the expression of a target gene in a subject in vitro or in vivo, wherein the method comprises the step of administrating the pharmaceutical composition of the present disclosure to a subject in the case of in vivo assays or therapeutic treatment, or contacting the pharmaceutical composition with a cell in the case of in vitro assays.
It is to be understood that both the foregoing general description and the following detailed description are exemplary and explanatory only and are not restrictive of the invention, as claimed.
A better understanding of the features and advantages of the present invention will be obtained with reference to the following detailed description that sets forth illustrative embodiments, in which the principles of the invention are utilized, and the accompanying drawings of which:
FIG. 1. shows the knockdown activity of delivery enhancing compound conjugated siRNA (i.e., DEC-conjugated siRNA or DEC-siRNA) on mouse Factor VII (mFVII) mRNA expression in primary mouse hepatocytes (PMH) cells. PMH cells were transfected via lipofectamineTM RNAiMax with each of the indicated DEC-conjugated oligonucleotide (DCOs) (i.e., RD-12339, RD-12585, RD-12586, RD-12710, RD-12711 and RD-12712) at 0.1 nM and 1.0 nM for 24 hours. Mock treatments were transfected in absence of oligonucleotide. dsCon2 served as a non-specific duplex control. mFVII mRNA levels were quantified by RT-qPCR using a gene specific primer set. Tbp was amplified as an internal reference. Shown are the mean values of mFVII mRNA in the cells relative to Mock treatment after normalizing to Tbp [mean ± Standard Error of the Mean (SEM) of two replicated wells] .
FIG. 2. shows the knockdown activity of DEC-conjugated siRNA on mouse FVII mRNA expression by free uptake in PMH cells. The indicated DCOs (i.e., RD-12339, RD-12585, RD-12586, RD-12710, RD-12711 and RD-12712, as described in FIG. 1. ) were directly added to PMH cell culture media at 0.01, 0.05, 0.20, 0.78, 3.13, 12.50, 50 and 200 nM
for 24 hours. Mock treatment was transfected in absence of oligonucleotide (not shown) . dsCon2 duplex served as a non-specific duplex control (not shown) . mFVII mRNA levels were quantified by RT-qPCR using a gene specific primer set. Tbp was amplified as an internal reference. Shown are the mean values of mFVII mRNA in the cells relative to Mock treatment after normalizing to Tbp (mean ± SEM of two replicated wells) .
FIG. 3. shows the in vivo knockdown activity of DEC-conjugated siRNAs on mouse FVII mRNA expression via subcutaneous injection (SC) injection in C57BL/6J mice. The indicated DCOs (i.e., RD-12339, RD-12585, RD-12586, RD-12710, RD-12711 and RD-12712, as described in FIG. 1. ) were administered via SC injection into C57BL/6J mice on postnatal day (PND) 40 at 0.2, 1 and 5 mg/kg doses. Saline was injected as a vehicle control to establish the baseline of mFVII mRNA expression. Mice were sacrificed on 3 days post dosing and mFVII mRNA levels were quantified in total RNA isolated from liver tissue preps via RT-qPCR using a gene specific primer set. Tbp was amplified as an internal reference. Mean mFVII mRNA levels in liver tissue of each treatment group are shown relative to saline group after normalizing to Tbp (mean ± SEM of 3 animals per group) .
FIG. 4. shows the in vivo knockdown activity of DEC-conjugated siRNA on mouse FVII protein expression via SC injection in C57BL/6J mice. The indicated DCOs (i.e., RD-12339, RD-12585 and RD-12586, as described in FIG. 1. ) were administered via SC injection into C57BL/6J mice on PND 40 at 0.2, 1 and 5 mg/kg doses. Saline was injected as a vehicle control to establish the baseline of mFVII protein expression. Mice were sacrificed on day 3 following treatment and mFVII protein level was quantified in mice plasma by ELISA assay. Mean mFVII protein levels in mice plasma by detecting the OD values of each treatment group are shown relative to saline group (mean ± SEM of 3 animals per group) . *represents p < 0.05; **represents p < 0.01; ***represents p < 0.001; and ****represents p < 0.0001.
FIG. 5. shows the duration of DEC-conjugated siRNA knockdown activity on mouse FVII protein expression via SC injection in C57BL/6J mice. The indicated DCOs (i.e., RD-12710 and RD-12712) were administered via SC injection into C57BL/6J mice on PND 40 at 3 mg/kg for 89 days. RD-11706 was injected at 3 mg/kg and served as a control. Saline was injected as a vehicle control to establish the baseline of mFVII protein expression. Mouse plasmas were collected on day 10, 31, 54, 61, 80 and 89 following treatment and mFVII protein level was quantified in mouse plasma by ELISA assay. Mean mFVII protein levels in
mouse plasma by detecting the OD values of 3 animals at each time point of per group are shown relative to saline group (mean ± SEM of 3 animals at each time point of each group) .
FIGs. 6A-6B show in vitro knockdown activity of DEC-siRNAs on mouse Sod1 (Sod1) mRNA expression by free uptake in PMH cells. DEC-siRNAs (i.e., RD-13110, RD-13115 and RD-13118) were added into PMH cell culture medium at the indicated concentration gradient (i.e., 1.56, 6.25, 25, 100, 400 and 1600 nM) to generate dose response curves as shown in FIG. 6A. Treatments in absence of oligonucleotide were used to normalize expression data. Sod1 mRNA levels were quantified via RT-qPCR using a gene specific primer set following RNA isolation and RT reactions. Tbp was amplified as an internal reference. Treatment with RD-12556 served as a non-conjugate control for comparison of dose dependent knockdown in absence DEC conjugation was shown in FIG. 6B (mean ± SEM of two replicated wells) .
FIG. 7 shows in vivo knockdown activity of different DEC-siRNAs on Sod1 mRNA expression via intracerebroventricular (ICV) injection in adult C57BL/6J mice. The indicated DEC-siRNAs (i.e., RD-13110, RD-13115 and RD-13118) were administered via unilateral ICV injection at 200 μg. Saline was injected as a vehicle control to establish the baseline. RD-12556 was injected at 200 μg as a non-conjugate control. Mice were sacrificed on 7 days post dosing and Sod1 mRNA knockdown was quantified in brain (i.e., brain-frontal cortex, cerebrum and brain-cerebellum) , spinal cord (i.e., cervical, thoracic and lumbar) and peripheral tissue (i.e., liver) via RT-qPCR using a gene specific primer set after RNA isolation and RT reaction. Tbp was amplified as an internal reference. Mean Sod1 mRNA levels are shown in each of the indicated tissues relative to mRNA levels in the saline group after normalizing to Tbp (mean ± SEM of 2-3 animals per group) .
FIG. 8 shows in vivo activity of different DEC-siRNAs on knocking down Sod1 mRNA expression following intravenous (IV) injection into the lateral tail vein of adult C57BL/6J mice. The indicated DEC-siRNAs (i.e., RD-13110, RD-13115 and RD-13118) were administered via IV injection at 20 mg/kg. Saline was injected as a vehicle control to establish the baseline. RD-12556 was injected at 20 mg/kg as a non-conjugate control. Mice were sacrificed on day 7 following treatment and Sod1 mRNA knockdown was quantified in select peripheral tissues (i.e., heart, liver, spleen, lung, kidney and bladder) via RT-qPCR using a gene specific primer set after RNA isolation and RT reaction. Tbp was amplified as an internal
reference. Mean Sod1 mRNA levels are shown in each of the indicated tissues relative to mRNA levels in the saline group after normalizing to Tbp (mean ± SEM of 3 animals per group) .
FIG. 9 shows in vivo knockdown activity of different DEC-siRNAs on Sod1 mRNA expression following IV injection into adult C57BL/6J mice. The indicated DEC-siRNAs (i.e., RD-13110, RD-13115 and RD-13118) were administered via IV injection at 20 mg/kg. Saline was injected as a vehicle control to establish the baseline. RD-12556 was injected at 20 mg/kg as a non-conjugate control. Mice were sacrificed on 7 days post dosing and Sod1 mRNA levels were quantified in skeletal muscle tissue from different locations (i.e., bicep, semitendinosus, platysma and gluteus) via RT-qPCR using a gene specific primer set after RNA isolation and RT reaction. Tbp was amplified as an internal reference for RNA loading. Mean Sod1 mRNA levels in skeletal muscle tissue of 3 animals per group are shown relative to mRNA levels in the saline group after normalizing to Tbp (mean ± SEM of 3 animals per group) .
FIG. 10 shows knockdown activity of different DEC-siRNAs on rat Sod1 mRNA expression via intravitreal (IVT) injection in adult SD rats. The indicated DEC-siRNAs (i.e., RD-13115 and RD-13118) were administered via IVT injection at 30 μg per left eye. The right eye of each rat served as a non-injected naive eye. Saline was injected as a vehicle control to establish the baseline. RD-12556 was injected as a non-conjugate control. Rats were sacrificed on day 14 following treatment and Sod1 mRNA levels were quantified in eyes via RT-qPCR using a rat gene specific primer set after RNA isolation and RT reaction. Geometric mean of the mRNA levels of Hprt1 and Hmbs were used as an internal reference for RNA loading. Mean Sod1 mRNA levels in retina tissue of 3 animalsfor each group are shown relative to mRNA levels in the saline group after normalizing to Hprt1 and Hmbs. The data represents mean ± SEM of 3 mice per group.
FIG. 11 shows the saRNA concentration of DEC-saRNA in adult C57BL/6J mice bladder after intravesical bladder (IVB) instillation. The indicated DEC-saRNA (i.e., RD-13520) was administered via IVB injection at 3 mg. RD-10773 was injected as a non-conjugate saRNA control (i.e., in absence of DEC) . The mice were sacrificed on 2-, 6-, 12-hour, day-1 and day-4 following treatment and the concentrations of RD-13520 and RD-10773 were quantified in bladder tissue via stem-loop RT-qPCR using a gene specific primer set (R1-
40-AS-SL-RT, R1-40-AS-SL-F1 and SL-RT-qPCR-Uni-R2, see Table 13) . Mean concentration (ng/g) for both saRNAs in mouse bladder tissue samples from 2 animals (n=2) are shown at the indicated time points (mean ± SEM) .
FIGs. 12A-12C show body weight change of and Sod1 knockdown activity of DEC-siRNAs via IV injection in adult C57BL/6J mice. The indicated siRNAs (i.e., RD-15135, RD-15136, RD-15137 and RD-15138) were administered into C57BL/6J mice at 10 mg/kg. RD-15135 served as a non-conjugate control. RD-15136 was a lipid (i.e., C16) conjugated siRNA. RD-15137 with a typical DEC structure (i.e., C5X5) served as a typical DEC-siRNA. RD-15138 with a typical DEC structure (i.e., C5X5) and a lipid (i.e., C16) served as a combo siRNA (i.e., DEC-C16-siRNA) . Treatment with saline alone was used as a vehicle control to establish baseline expression. FIG. 12A shows the body weight change of C57BL/6J mice out to day 28 post treatment. FIG. 12B shows the remaining mouse Sod1 mRNA on 14 days post dosing as quantified in tissues from periphery (i.e., heart, liver, kidney, fat tissues, pancreas, diaphragm) , blood vessel (i.e., thoracic aorta with vein) and skeletal muscle (i.e., bicep, semitendinosus, platysma and gluteus) via RT-qPCR using a gene specific primer set. FIG. 12C shows the remaining mouse Sod1 mRNA on 28 days post dosing as quantified in tissues from periphery (i.e., heart, liver, kidney, fat tissues, pancreas, diaphragm) , blood vessel (i.e., thoracic aorta with vein) and skeletal muscle (i.e., bicep, semitendinosus, platysma and gluteus) via RT-qPCR using a gene specific primer set. Rpl13a was amplified as an internal reference. Mean Sod1 mRNA levels in the selected mouse tissues are shown relative saline control after normalized to Rpl13a. The data represents mean ± SEM of 3 mice per group.
FIGs. 13A-13C show body weight change of and Sod1 knockdown activity of DEC-siRNAs via intrathecal (IT) injection in adult SD female rats. The indicated siRNAs (i.e., RD-15135, RD-15136, RD-15137 and RD-15138) were administered into C57BL/6J mice at 0.9 mg.RD-15135 served as a non-conjugate control. RD-15136 was a lipid (i.e., C16) conjugated siRNA. RD-15137 with a typical DEC structure (i.e., C5X5) served as a typical DEC-siRNA. RD-15138 with a typical DEC structure (i.e., C5X5) and a lipid (i.e., C16) served as a combo siRNA (i.e., DEC-C16-siRNA) . Treatment with artificial cerebrospinal fluid (aCSF) alone was used as a vehicle control to establish baseline expression. FIG. 13A shows the body weight change of SD rats out to day 28 post treatment. FIG. 13B shows the remaining rat Sod1 mRNA on 14 days post dosing as quantified in tissues from brain (i.e., frontal cortex, cerebellum and
cerebrum) , spinal cord (i.e., cervical, thoracic and lumbar) and periphery (i.e., liver and kidney) via RT-qPCR using a gene specific primer set. FIG. 13C shows the remaining rat Sod1 mRNA on 28 days post dosing as quantifiedin tissues from brain (i.e., frontal cortex, cerebellum and brainstem) , spinal cord (i.e., cervical, thoracic and lumbar) and periphery (i.e., liver and kidney) . Geometric mean of the mRNA levels of Actb and B2m were used as an internal reference for RNA loading. Mean Sod1 mRNA levels in the selected rat tissues are shown relative to mRNA levels in the aCSF group after normalized to Actb and B2m. The data represents mean ± SEM of 3 mice per group.
FIGs. 14A-14B show the in vitro knockdown activity of DEC-siRNA with accessory oligonucleotide (i.e., DEC-siRNA-ACO) on human SOD1 (i.e., SOD1) mRNA expression in SK-N-AS and T98G cells. The indicated DEC-siRNA-ACOs (i.e., RD-16149, RD-16099, RD-16100, RD-16101, RD-16150, RD-16103, RD-16104 and RD-16105) were transfected into SK-N-AS and T98G cell at the indicated concentrations (i.e., 1.56, 6.25, 25, 100, 400 and 1600 nM) to generate dose response curves as shown in FIG. 14A-14B. RD-16106 was transfected and served as a siRNA-ACO control. Mock treatments were transfected in absence of oligonucleotide (not shown) . SOD1 mRNA levels were quantified via RT-qPCR using a gene specific primer set following RNA isolation and RT reactions. B2M was amplified as an internal reference. Shown are the mean values of remaining SOD1 mRNA in the cells relative to Mock treatment after normalizing to B2M (mean ± SEM of two replicated wells) .
FIGs. 15A-15B show SOD1 knockdown activity and tissue concentration of siRNAs via ICV injection in adult hSOD1G93A mice. The indicated siRNAs (i.e., RD-14851, RD-12500, RD-16145 and RD-16334) were administered into hSOD1G93A mice at 200 μg. RD-14851, RD-12500 and RD-16145 were administered as siRNA-ACOs. RD-16334 with a DEC structure (i.e., L17) was administered and served as a DEC-siRNA-ACO. Treatment with aCSF alone was used as a vehicle control to establish baseline expression. FIG. 15A shows the remaining SOD1 mRNA on 30 days post dosing as quantified in tissues from brain (i.e., frontal cortex, cerebellum and cerebrum) , spinal cord and periphery (i.e., liver) via RT-qPCR using a gene specific primer set. Rpl13a was amplified as an internal reference. Mean SOD1 mRNA levels in the selected mouse tissues are shown relative to mRNA levels in the aCSF group after normalized to Rpl13a. FIG. 15B shows the concentration of the antisense strand of each siRNA in tissues from brain (i.e., frontal cortex, cerebellum and cerebrum) , spinal cord
and periphery (i.e., liver) via stem-loop RT-qPCR using a gene specific primer set (R17-02-AS-SL-RT, R17-02-AS-SL-F3 and SL-RT-qPCR-Uni-R1, see Table 13) 30 days posting dosing. Mean values of siRNA concentrations from 5 animals (n=5) are shown in each treatment. The data represents mean ± SEM of 5 mice per group.
FIGs. 16A-16B show splicing activity of “saRNA-ASO” DEC conjugated dual acting oligonucleotides (DEC-DAO) structure at converting SMN2 pre-mRNA to SMN2FL over the SMN2Δ7 mRNA isoform in GM03813 cells. The indicated DEC-DAO linkage with L21 linker (i.e., RD-16939 and RD-16940) , DEC-saSMN2 (i.e., RD-16424) and DEC-antisense oligonucleotide (DEC-ASO) (i.e., RD-14644) were transfected into GM03813 cells at the indicated concentrations (0.1, 1 and 10 nM) for 3 days. Mock treatments were transfected in the absence of an oligonucleotide. RD-16381 was transfected as a non-specific DEC control. Combo treatment (i.e., RD-16424+RD-14644) and combo treatment control (i.e., RD-16381+RD-16424, RD-16381+RD-14644) were also transfected into GM03813 cells. FIGs. 16A-16B show the mRNA levels of SMN2FL and SMN2Δ7 as quantified by RT-qPCR using a gene specific primer set in each of the PCR reactions. SDHA was amplified as an internal reference used to normalize expression data. Data represents mean expression levels of SMN2FL or SMN2Δ7 relative to Mock treatment after normalized to SDHA (mean ± SEM of two replicated transfection wells) .
FIGs. 17A-17C show the effect of “saRNA-siRNA” DEC-DAO structure targeting two different human genes (SMN2 and SOD1) on the expression of SMN2 (SMN2FL and SMN2Δ7) and SOD1 in GM03813 cells. The indicated DEC-DAO linkage with L21 linker (i.e., RD-16941) , DEC-saSMN2 (i.e., RD-16424) and DEC-siSOD1 (i.e., RD-13115) were transfected into GM03813 cells at indicated concentrations (i.e., 0.1, 1 and 10 nM) for 3 days. Mock treatments were transfected in the absence of an oligonucleotide. RD-16381 was transfected as a non-specific DEC control. Combo treatment (i.e., RD-16424+RD-13115) and combo treatment control (i.e., RD-16381+RD-16424, RD-16381+RD-13115) also were transfected into GM03813 cells at indicated concentrations (i.e., 0.1, 1 and 10 nM) for 3 days. FIGs. 17A-17B show the mRNA levels of SMN2FL and SMN2Δ7 as quantified by RT-qPCR using a gene specific primer set in each of the PCR reactions. FIG. 17C shows remaining SOD1 mRNA levels as quantified by RT-qPCR using a gene specific primer set in each of the PCR reactions. SDHA was amplified as an internal reference used to normalize expression data. Data
represents mean expression levels of SMN2FL, SMN2Δ7 or SOD1 relative to Mock treatment after normalized to SDHA (mean ± SEM of two replicated transfection wells) .
FIGs. 18A-18B show knockdown activity of “siRNA-siRNA” DEC-DAO structure targeting two different mouse genes (Sod1 and Ppig) on the expression of mouse Sod1 and mouse Ppig in mouse myoblast cell line (C2C12) . The indicated DEC-DAO linkage with L21 linker (i.e., RD-16942) , DEC-siSod1 (i.e., RD-13115) and DEC-siPpig (i.e., RD-14672) were transfected into C2C12 cells at indicated concentrations (i.e., 0.01, 0.1 and 1 nM) for 24 hours. Mock treatments were transfected in the absence of an oligonucleotide. RD-16381 was transfected as a non-specific DEC control. Combo treatment (i.e., RD-13115+RD-14672) and combo treatment control (i.e., RD-16381+RD-13115, RD-16381+RD-14672) were also transfected into C2C12 cells at indicated concentrations (i.e., 0.01, 0.1 and 1 nM) for 24 hours. FIG. 18A shows remaining mouse Sod1 mRNA levels as quantified by RT-qPCR using a gene specific primer set in each of the PCR reactions. FIG. 18B shows the remaining mouse Ppig mRNA levels as quantified by RT-qPCR using a gene specific primer set in each of the PCR reactions. Mouse Rpl13a was amplified as an internal reference used to normalize expression data. Data represents mean expression levels of Sod1 and Ppig relative to Mock treatment after normalized to Rpl13a (mean ± SEM of two replicated transfection wells) .
FIG. 19 shows knockdown activity of “siRNA-siRNA” DEC-DAO structure targeting two different mouse genes (Sod1 and Ppig) on the expression of mouse Sod1 and mouse Ppig in adult C57BL/6J mice. The indicated DEC-DAO linkage with L21 linker (i.e., RD-16942) , DEC-siSod1 (i.e., RD-13115) and DEC-siPpig (i.e., RD-14672) were administered into C57BL/6J mice via IV injection at 10 mg/kg. Combo treatment (i.e., RD-13115+RD-14672) was administered via IV injection at 10 mg/kg. Treatment with saline alone was used as a vehicle control to establish baseline expression. Remaining mouse Sod1 mRNA on 14 days post dosing were quantified in tissues from periphery (i.e., heart, liver, kidney, fat tissues, diaphragm) , blood vessel (i.e., thoracic aorta with vein) and skeletal muscle (i.e., bicep, semitendinosus, platysma and gluteus) via RT-qPCR using a gene specific primer set. Tbp was amplified as an internal reference used to normalize expression data. Mean Sod1 mRNA levels in the selected mouse tissues are shown relative to mRNA levels in the saline group after normalized to Tbp. The data represents mean ± SEM of 3-4 mice per group.
FIGs. 20A-20B compares the oligonucleotide concentration of DEC-saRNA to non-
conjugated saRNA in retina and vitreous humor of adult SD rat after intravitreal (IVT) injection. The indicated DEC-saRNA (i.e., RD-16447) and non-conjugated saRNA (i.e., RD-12173) were administered via IVT injection at 0.03 mg. The rats were sacrificed on 1-hour, day-1, -3, -7, -14 and -28 following treatment and the concentrations of RD-16447 and RD-12173 were quantified in retina and vitreous humor via stem-loop RT-qPCR using a gene specific primer set of (R1C-0M4-AS-SL-RT, R1C-0M4-AS-SL-F4 and SL-RT-qPCR-Uni-R1, see Table 13) . Mean values of oligonucleotide concentrations of each group are shown (mean ± SEM of 3 animals per group) .
FIG. 21 shows the in vitro knockdown activity of DEC-siRNA on SOD1 mRNA expression in T98G cells. The indicated DEC-siRNA (i.e., RD-17138) were transfected into T98G cell at 0.1 nM and 1 nM for 24 hours. RD-11566 was transfected and served as a non-specific duplex control. Mock treatments were transfected in absence of oligonucleotide. SOD1 mRNA levels were quantified via RT-qPCR using a gene specific primer set following RNA isolation and RT reactions. B2M was amplified as an internal reference used to normalize expression data. Shown are the mean values of remaining SOD1 mRNA in the cells relative to Mock treatment after normalizing to B2M (mean ± SEM of four replicated wells) .
The newly developed oligonucleotide delivery agent enables more efficient delivery of desired amount or higher level of the oligonucleotides, as compared to the oligonucleotide delivery technologies of the prior art, at the above indicated target tissues, and thus may improve bioactive and pharmacological properties (e.g., biodistribution, bioavailability, pharmacokinetics, activity, potency, etc. ) . In some embodiments, the improvement of bioactive properties may cause improved cell uptake, higher potency, and longer duration/half-life. In some embodiments, the improvement of pharmacological properties may also lead to lower toxicity, lower dose, less frequent administrations, and less undesired immune responses. In some embodiments, the present oligonucleotide delivery agent involves a simpler synthesis process and thus has better processibility in manufacturing. In some embodiments, the present oligonucleotide delivery agent possesses inherent pharmacological properties of a free benzimidazole or a benzimidazole derivative which targets proton pump (as reversible and
irreversible proton pump inhibitors, PPI) , dopamine receptor (DRD) , angiotensin II type 1 receptor (AT1) , histamine receptor (HRH) , dual specific mitogen activated protein kinase (MEK) and/or cyclin dependent kinase (CDK) , etc. In some embodiments, the inherent pharmacological properties enable the present oligonucleotide delivery agent to be potentially used in drugs including peptic ulcer, hypertension, schizophrenia, parasitic infection, bacterial infection, virus infection, and cancer, etc.
In some embodiments, the newly developed oligonucleotide delivery agent disclosed herein can facilitate delivery of an effective amount of oligonucleotides to achieve and function at target tissues or cells, e.g., in central nervous system (e.g., brain and spinal cord) , liver, lung, kidney, intestine, pancreas, cholecyst, heart, lymph nodes, spleen, stomach, bladder, muscle and bone. In some embodiments, the oligonucleotide delivery agent’s modulating activities, e.g., up-regulating or down-regulating, the expression of a target gene in the cells can be improved as compared to the technologies of the prior art. Therefore, in light of the findings provided herein, those skilled in the art will appreciate that use of the oligonucleotide delivery agent as described herein may facilitate improved bio-distribution of therapeutic oligonucleotides in various target (s) or cells as stated above for preventing, treating and/or delaying on-set of various diseases, disorders and/or conditions.
Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which the invention belongs. Also, all publications, patent applications, patents, and other references mentioned herein are incorporated by reference.
As disclosed herein, singular forms, such as “a” and “this” , include plural objects, unless otherwise specified clearly in the context.
As disclosed herein, the terms “approximately, ” “about, ” “substantially” , and similar terms are intended to have a broad meaning in harmony with the common and accepted usage by those of ordinary skill in the art to which the subject matter of this disclosure pertains.
As disclosed herein, “and/or” means “and, or as an alternative” . If any numerical ranges are provided, the ranges include any numerical values in the range (including the upper and lower values) , and also any sub-ranges in the range. For example, the range from 1 to 3 may include any of the numerical values 1, 2 and 3, as well as sub-ranges from 1 to 2 and from 2 to 3.
The terms “oligonucleotide” , “polynucleotide” or “oligo” can be used interchangeably, and refer to polymers of nucleotides, and includes, but is not limited to, single-stranded, double-stranded or partial double-stranded nucleic acid molecules of DNA, RNA, or DNA/RNA hybrid, oligonucleotide strands containing regularly and irregularly alternating deoxyribosyl portions and ribosyl portions, as well as modified and naturally or unnaturally existing frameworks for such oligonucleotides. In an embodiment, the oligonucleotide can be selected from the group consisting of antisense oligonucleotide (ASO) , antisense RNA, short interfering RNA (siRNA) , micro-RNA (miRNA) , small activating RNA (saRNA) , dsRNA, and small guide RNA (sgRNA) .
The terms “oligonucleotide strand” , “strand” and “oligonucleotide sequence” as used herein can be used interchangeably and refer to a generic term for short nucleotide sequences having less than 50 bases, such as less than 45, less than 40, less than 35 bases, such as 2-50 bases, or 5-45 bases, or 10-40 bases, or 15-35 bases, or 20-30 bases (including nucleotides in deoxyribonucleic acid (DNA) or ribonucleic acid (RNA) ) . In some non-limiting examples, the length of a strand can be any length from 5 to 50 nucleotides, 10 to 40 nucleotides, 15 to 35 nucleotides, 18 to 30 nucleotides or 20 to 25 nucleotides.
The term “target gene” as used herein can refer to nucleic acid sequences, transgenes, viral or bacterial sequences, chromosomes or extrachromosomal genes that are naturally present in organisms, and/or can be transiently or stably transfected or incorporated into cells and/or chromatins thereof. The target gene can be a protein-coding gene or a non-protein-coding gene such as a microRNA gene and a long non-coding RNA gene.
In embodiments of the present application, one target gene is SOD1. By definition, “target sequence” is a sequence fragment to which the sense strand or antisense oligonucleotide of the siRNA or saRNA is homologous or complementary. For example, in certain embodiments, a SOD1 siRNA is homologous or complementary to a target select sequence within human SOD1 transcript.
As used herein, the term “guide strand” or “G strand” refers to a strand in a small RNA duplex that assembles with the Argonaute (AGO) protein. The other strand partially or completely complementary to the guide strand is called “passenger strand” or “P strand” . Without being limited to any specific theory, the strand carrying the complementary sequence to the target is the antisense strand and, if properly designed, will be preferentially chosen to
be the guide strand. In this case, the passenger strand is the sense strand. However, one cannot state that a strand is the guide strand until the 5’ end of the strand has been captured within the MID domain of AGO2. Accordingly, the sense strand can be selected as the guide strand, resultant in an antisense passenger strand.
As used herein, the term “LNA” refers to a locked nucleic acid in which the 2’ -oxygen and 4’ -carbon atoms are joined by an extra bridge. As used herein, the term “BNA” refers to a 2′-O and 4′-aminoethylene bridged nucleic acid that can contain a five-membered or six-membered bridged structure with an N-O linkage. As used herein, the term “PNA” refers to a nucleic acid mimic with a pseudopeptide backbone composed of N- (2-aminoethyl) glycine units with the nucleobases attached to the glycine nitrogen via carbonyl methylene linkers.
The terms “antisense oligonucleotide” or “ASO” , as used herein, refer to a single strand oligonucleotide or the like having, comprising, or consisting of a sequence of bases or the like which allow the oligonucleotide or the like to hybridize to a target molecule, such as another nucleic acid, modified nucleic acid or nucleic acid analog, e.g., by base-pairing, such as Watson-Crick base-pairing or non-Watson-Crick base pairing. In some embodiments, an antisense oligonucleotide is fully complementary or nearly fully complementary to the target molecule. In some embodiments, any oligonucleotide of any type described herein or known in the art can be used as an antisense oligonucleotide. In various embodiments, an antisense oligonucleotide can perform or participate in any of various biological functions, including RNA interference, RNase H-mediated cleavage, exon skipping, the prevention of exon skipping, the enhancement or blocking of an agent (e.g., a protein, RNA, protein-RNA complex, or any other molecule) from binding to another nucleic acid, or any other biological function performed by an antisense oligonucleotide, as described herein or known in the art.
As used herein, the terms “small interfering RNA” , “siRNA” and “inhibitory RNA (iRNA) ” can be used interchangeably and refer to a ribonucleic acid molecule that can down-regulate, knockdown, or silence target gene expression. It can be a double-stranded nucleic acid molecule. It interferes with the expression of specific genes with complementary nucleotide sequences by degrading mRNA after transcription, preventing translation. siRNA binds to target mRNA mainly in the cytoplasm to down-regulate gene expression post-transcriptionally via the RNA interference (RNAi) mechanism. siRNAs may be designed to target a gene’s mRNA sequence to silence its expression via the RNAi mechanism, such as
SOD1, for maximizing treatment outcomes, e.g., for ALS patients. siRNAs are molecules having endogenous RNA bases or chemically modified nucleotides. The modifications do not abolish cellular activity, but rather impart increased stability and/or increased cellular potency. Examples of chemical modifications include phosphorothioate groups, 2′-deoxynucleotide, 2′-OCH3-containing ribonucleotides, 2′-F-ribonucleotides, 2′-methoxyethyl ribonucleotides, combinations thereof and the like. The siRNA can have varying lengths (e.g., 10-200 bps) and structures (e.g., hairpins, single/double/partial-double strands, bulges, nicks/gaps, mismatches) and are processed in cells to provide active gene silencing. A double-stranded siRNA can have the same number of nucleotides on each strand (blunt ends) or asymmetric ends (overhangs) . An overhang of 1-2 nucleotides, for example, can be present on the sense and/or the antisense strand, as well as present on the 5′-and/or the 3′-ends of a given strand. The length of the siRNA molecule is typically about 10 to about 60, about 10 to about 50, about 15 to about 30, about 17 to about 29, about 18 to about 28, about 19 to about 27, about 20 to about 26, about 21 to about 25, and about 22 to about 24 base pairs, and typically about 15, about 16, about 17, about 18, about 19, about 20, about 21, about 23, about 25, about 30, about 40, or about 50 base pairs. In addition, the terms “small interfering RNA” , “silencing RNA” and “siRNA” also contain nucleic acids other than the ribonucleotide, including, but not limited to, modified nucleotides or analogues.
As used herein, the terms “small activating RNA” , “saRNA” and “small activating ribonucleic acid” can be used interchangeably and refer to a ribonucleic acid molecule that can up-regulate target gene expression. It can be a double-stranded nucleic acid molecule composed of a first nucleic acid strand containing a ribonucleotide sequence with sequence homology with the non-coding nucleic acid sequence (such as a promoter and an enhancer) of a target gene and a second nucleic acid strand containing a nucleotide sequence complementary with the first strand. The saRNA can also be comprised of a synthesized or vector-expressed single-stranded RNA molecule that prone to form a hairpin structure by two complementary regions within the molecule, wherein the first region contains a ribonucleotide sequence having sequence homology with the target sequence of a promoter of a gene, and a ribonucleotide sequence contained in the second region is complementary with the first region. The length of the duplex region of the saRNA molecule is typically about 10 to about 60, about 10 to about 50, about 10 to about 40, about 12 to about 30, about 14 to about 28, about
16 to about 26, about 18 to about 24, and about 20 to about 22 base pairs, and typically about 10, about 13, about 15, about 17, about 18, about 19, about 20, about 21, about 22, about 25, about 30, about 40, about 50, or about 60 base pairs. In addition, the terms “small activating RNA” , “saRNA” and “small activating ribonucleic acid” also contain nucleic acids other than the ribonucleotide, including, but not limited to, modified nucleotides or analogues.
As used herein, “ODV” and “oligonucleotide delivery vehicle” are used interchangeably, which refer to an oligonucleotide molecule comprising a duplex or double-stranded RNA (e.g., siRNA or saRNA) and an accessory oligonucleotide (ACO) which is covalently linked to the duplex RNA via a linker.
As used herein, the terms “delivery enhancing compound” and “DEC” are used interchangeably and refer to the compound of the present disclosure.
As used herein, the terms “DEC-conjugated oligonucleotide” and “DCO” can be used interchangeably and refer to a combination in which at least one moiety derived from the DEC has been attached, e.g. covalently, to at least one oligonucleotide. Additionally, the DCO may further comprising at least one targeting moiety which helps the oligonucleotide in targeting to, accumulating in, or accessing to target site in a cell.
The phrases “administration” , “parenteral administration” , “administrated” and “administered parenterally” as used herein have their art-understood meaning referring to ordinary enteral administration, topical administration, and especially, injection administration, and may include, without limitation, intravenous, intramuscular, intraarterial, intrathecal, intracapsular, intraorbital, intracardiac, intradermal, intraperitoneal, transtracheal, subcutaneous, subcuticular, intraarticular, subcapsular, subarachnoid, intraspinal, and intrasternal injection and infusion.
As used herein, the term “pharmaceutical composition” refers to an active agent, optionally formulated together with one or more pharmaceutically acceptable carriers and other additives. In some embodiments, active agent is present in unit dose amount appropriate for administration in a therapeutic regimen that shows a statistically significant probability of achieving a predetermined therapeutic effect when administered to a relevant population. In some embodiments, pharmaceutical compositions may be specially formulated for administration in solid or liquid form, including those adapted for the following: oral 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; parenteral administration, for example, by subcutaneous, intramuscular, intravenous or epidural injection as, for example, a sterile solution or suspension, or sustained-release formulation; topical application, for example, as a cream, ointment, or a controlled-release patch or spray applied to the skin, lungs, or oral cavity; intravaginally or intrarectally, for example, as a pessary, cream, or foam; sublingually; ocularly; transdermally; or nasally, pulmonary, and to other mucosal surfaces.
As used herein, the term “pharmaceutically acceptable carrier” means a pharmaceutically acceptable material, composition or vehicle, such as a liquid or solid filler, diluent, excipient, or solvent encapsulating material, involved in carrying or transporting the subject compound from one organ, or portion of the body, to another organ, or portion of the body. Each carrier must be “acceptable” in the sense of being compatible with the other ingredients of the formulation and not injurious to the patient. Some examples of materials which can serve as pharmaceutically-acceptable carriers include: sugars, such as lactose, glucose and sucrose; starches, such as corn starch and potato starch; cellulose, and its derivatives, such as sodium carboxymethyl cellulose, ethyl cellulose and cellulose acetate; powdered tragacanth; malt; gelatin; talc; excipients, such as cocoa butter and suppository waxes; oils, such as peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, corn oil and soybean oil; glycols, such as propylene glycol; polyols, such as glycerin, sorbitol, mannitol and polyethylene glycol; esters, such as ethyl oleate and ethyl laurate; agar; buffering agents, such as magnesium hydroxide and aluminum hydroxide; alginic acid; pyrogen-free water; isotonic saline; Ringer′s solution; ethyl alcohol; pH buffered solutions; polyesters, polycarbonates and/or polyanhydrides; and other non-toxic compatible substances employed in pharmaceutical formulations.
As used herein, the term “subject” , “test subject” and related terms, as used herein, refer to any organism to which a provided compound or composition is administered in accordance with the present invention e.g., for experimental, diagnostic, prophylactic, and/or therapeutic purposes. Typical subjects include animals (e.g., mammals such as mice, rats, rabbits, non-human primates, and humans; fishes; birds; insects; worms; etc. ) and plants. In some embodiments, a subject may be suffering from, and/or susceptible to a disease, disorder, and/or condition. In some embodiments, a subject is a human being or other mammal. In some
embodiments, a subject can be male or female. In non-limiting examples, the animal is a vertebrate such as a primate, rodent, domestic animal or game animal. In non-limiting examples, primates include chimpanzees, cynomolgus monkeys, spider monkeys, and macaques, e.g., Rhesus. Rodents include mice, rats, woodchucks, ferrets, rabbits and hamsters. In non-limiting examples, domestic and game animals include cows, horses, pigs, deer, bison, buffalo, feline species, e.g., domestic cat, canine species, e.g., dog, fox, wolf, avian species, e.g., chicken, emu, ostrich, and fish, e.g., trout, catfish and salmon. In certain embodiments of the aspects described herein, the subject is a mammal, e.g., a primate, e.g., a human. In non-limiting examples, the mammal can be a human, non-human primate, mouse, rat, dog, cat, horse, or cow, but are not limited to these examples. In some embodiments, a mammal other than a human can be advantageously used as subjects that represent animal models of disorders associated with autoimmune disease or inflammation. In some embodiments, a method and composition described herein can be used to treat domesticated animals and/or pets.
A “therapeutically effective amount” of a composition is an amount sufficient to achieve a desired therapeutic effect, and therefore does not require cure or complete remission. In embodiments of the present application, therapeutic efficacy is an improvement in any of the disease indicators, and a therapeutically effective amount is sufficient to cause an improvement in a clinically significant condition/symptom in the treated individual. The phrases “therapeutically effective amount” and “effective amount” are used herein to mean an amount sufficient to reduce by at least about 15 percent, preferably by at least 50 percent, more preferably by at least 90 percent, or to increase at least about 50 percent, at least about 100 percent, at least about 200 percent, more preferable at least about 500 percent and can prevent a clinically significant deficit in the activity, function and response of the individual being treated. The effective amount may vary depending on such factors as the size and weight of the subject, the type of illness, or the particular agents of the application. For example, the choice of the agent of the application could affect what constitutes an “effective amount” . One of ordinary skill in the art would be able to study the factors contained herein and make the determination regarding the effective amount of the agents of the application without undue experimentation.
The regime of administration may affect what constitutes an effective amount. The
agent of the application can be administered to the subject either prior to or after the disease diagnosis or condition. Further, several divided dosages, as well as staggered dosages, can be administered daily or sequentially, or the dose can be continuously infused, or can be a bolus injection. Further, the dosages of the agent (s) of the application could be proportionally increased or decreased as indicated by the exigencies of the therapeutic or prophylactic situation.
The terms “treat, ” “treated, ” “treating” , or “treatment” as used herein have the meanings commonly understood in the medical arts, and therefore do not require cure or complete remission, and include any beneficial or desired clinical results. Non-limiting examples of such beneficial or desired clinical results are prolonging survival as compared to expected survival without treatment, reduced symptoms including one or more of the followings: weakness and atrophy of proximal skeletal muscles, inability to sit or walk independently, difficulties in swallowing, breathing, etc.
As used herein, “preventing” or “delaying” a disease refers to inhibiting the full development of a disease.
The present disclosure is based on an unexpected discovery that a compound having a specific structure comprising a nitrogen-containing five membered heterocyclic ring moiety may be directly or indirectly bonded to at least one oligonucleotide so as to aid the delivery of the oligonucleotide to a subject and thus achieving improved modulation of a target gene both in vitro and/or in vivo. In an exemplary embodiment, the compound as disclosed herein may remarkably enhance oligonucleotide delivery efficiency to liver, combine endosomal escape and nuclear translocation design, without the need of a delivery vector or formulation.
In exemplary embodiments, the nitrogen-containing five membered heterocyclic ring moiety may have a core structure shown by any of the following formulae, with all the chemical bonds, atoms and substituents attached to the ring atoms of the core structure omitted for the sake of simplicity:
The above indicated core structures comprise different ring atoms, and the fused ring core structures may be either electrically neutral or locally charged in the form of e.g., sulphonium or quaternary ammonium. It can be seen that one or more unsaturated double bonds may shift in the fused ring core structure due to the delocalized π-electronic conjugation effect, and all the substituents, including the presence/absence of any substituents, may vary depending on the categories and valences of each ring atom to which the substituents are attached. For example, an additional substituent may be attached to a sulphonium or quaternary ammonium ion which is present as a ring atom of the fused ring core structure. All of these variations are within the concept of the present disclosure.
In an embodiment, the compound of the present disclosure comprises structure of Formula BI, wherein X′ is selected from the group consisting of carbon, nitrogen, oxygen and sulfur; each of F′, G′, H′ and I′ is independently selected from the group consisting of carbon, nitrogen, oxygen and sulfur; and each of the asterisks refers to a site optionally linked to at least one substituent or an oligonucleotide directly or indirectly.
In an embodiment, the compound of the present disclosure has a structure represented by Formula AI or Formula AII:
wherein each independently represents a covalent single or double bond; X, on each occurrence, is an atom selected from the group consisting of carbon, nitrogen, oxygen and sulfur; each of F, G, H and I is independently selected from the group consisting of carbon, nitrogen, oxygen and sulfur.
In an embodiment, m is an integer of 1, 2 or 3, n is an integer of 1, 2 or 3, and m+n=4. In some exemplary embodiments, m=1 and n=3; or m=2 and n=2; or m=3 and n=1.
In an embodiment, C, on each occurrence, is either absent or selected from the group consisting of hydrogen; halogen atom, such as fluorine, chlorine, bromine or iodine; hydroxyl; (C1-C25) alkyl, such as - (C2-C22) alkyl, or - (C2-C19) alkyl, or - (C3-C18) alkyl, or - (C4-C16) alkyl, or - (C6-C12) alkyl, or - (C8-C10) alkyl; (C1-C20) alkoxy, such as - (C2-C19) alkoxy, or - (C3-C18) alkoxy, or - (C4-C16) alkoxy, or - (C6-C12) alkoxy; halogenated (C1-C20) alkyl, such as halogenated (C2-C19) alkyl, or halogenated (C3-C18) alkyl, or halogenated (C4-C16) alkyl, or halogenated (C6-C12) alkyl, or halogenated (C8-C10) alkyl; and halogenated (C1-C20) alkoxy, such as halogenated (C1-C20) alkoxy, such as halogenated (C2-C19) alkoxy, or halogenated (C3-C18) alkoxy, or halogenated (C4-C16) alkoxy, or halogenated (C6-C12) alkoxy, or halogenated (C8-C10) alkoxy; wherein “halogenated” can be fluorinated, chlorinated, brominated, iodinated, or combinations thereof. In an embodiment, in the structure represented by Formula I, each B is attached to any one of F, G, H and I, while C is attached to the rest of F, G, H and I. In an embodiment, B is
attached to H and three C’s are separately attached to each of F, G and I. In another embodiment, B is attached to G and three C’s are separately attached to each of F, H and I. In another embodiment, B is attached to F and three C’s are separately attached to each of G, H and I. In another embodiment, B is attached to I and three C’s are separately attached to each of F, G and H. In another embodiment, two B’s are separately attached to G and H, and two C’s are separately attached to each of F and I.
In an embodiment, B, on each occurrence, is independently selected from the group consisting of hydroxyl; -C (O) OH; -P (O) 2-OH; -P (O) -OH; -P (O) (S) -OH; -CN; - (C1-C22) alkyl, such as - (C2-C20) alkyl, or - (C3-C16) alkyl, or - (C6-C12) alkyl; -O- (C1-C22) alkyl, such as -O- (C2-C20) alkyl, or -O- (C3-C16) alkyl, or -O- (C6-C12) alkyl; - (C1-C22) alkenyl, such as - (C2-C20) alkenyl, or - (C3-C16) alkenyl, or - (C6-C12) alkenyl; - (C1-C22) alkylene-OH, such as - (C2-C20) alkylene-OH, or - (C3-C16) alkylene-OH, or - (C6-C12) alkylene-OH; - (C3-C22) cycloalkylene-OH, such as - (C5-C16) cycloalkylene-OH, or - (C6-C12) cycloalkylene-OH, or - (C6-C10) cycloalkylene-OH; - (C6-C22) arylene-OH; - (C6-C22) heteroarylene-OH; - (C1-C22) alkylene-C (O) OH, such as - (C2-C18) alkylene-C (O) OH, or - (C3-C16) alkylene-C (O) OH, or - (C6-C12) alkylene-C (O) OH; - (C3-C22) cycloalkylene-C (O) OH; - (C6-C22) arylene-C (O) OH; - (C5-C22) heteroarylene-C (O) OH; -O-C (O) - (C1-C22) alkylene-C (O) NH2, such as -O-C (O) - (C2-C18) alkylene-C (O) NH2, or -O-C (O) - (C3-C16) alkylene-C (O) NH2, or -O-C (O) - (C6-C12) alkylene-C (O) NH2; - (C1-C22) alkylene-O-C (O) - (C1-C22) alkylene-C (O) NH2, such as - (C2-C18) alkylene-O-C (O) - (C2-C18) alkylene-C (O) NH2, or - (C3-C16) alkylene-O-C (O) - (C3-C16) alkylene-C (O) NH2, or - (C6-C12) alkylene-O-C (O) - (C6-C12) alkylene-C (O) NH2; -O-C (O) - (C1-C22) alkylene-C (O) OH; - (C1-C22) alkylene-O-C (O) - (C1-C22) alkylene-C (O) OH; -C (O) - (C1-C22) alkylene-C (O) NH2; - (C1-C22) alkylene-C (O) - (C1-C22) alkylene-C (O) NH2; -C (O) -NH- (C1-C22) alkylene-OH; -C (O) -NH- (C1-C22) alkylene-C (O) OH; - (C1-C22) alkylene-C (O) -NH- (C1-C22) alkylene-C (O) OH; - (C1-C30) alkylene-P (O) 2-OH; - (C1-C22) alkylene-O-P (-N (C1-C22 alkyl) 2) -O- (C1-C22) alkylene-CN, such as - (C2-C18) alkylene-O-P (-N (C2-C18 alkyl) 2) -O- (C2-C18) alkylene-CN, or - (C3-C16) alkylene-O-P (-N (C3-C16 alkyl) 2) -O- (C3-C16) alkylene-CN, or - (C6-C12) alkylene-O-P (-N (C6-C12 alkyl) 2) -O- (C6-C12) alkylene-CN; - (C1-C22) alkylene-O-P (-N (C1-C22 alkyl) 2) -O- (C1-C22) alkylene-OH; - (C1-C22) alkylene-O-P (-N (C1-C22 alkyl) 2) -O- (C1-C22) alkylene-NH2; - (C1-C22) alkylene-O-P (-N (C1-C22 alkyl) 2) -O- (C1-C22) alkylene-C (O) OH; -C (O) -NH- (C1-C22) alkylene-O-P (-N (C1-C22 alkyl) 2) -O- (C1-C22) alkylene-CN; -C (O) -NH- (C1-C22) alkylene-O-P (-N (C1-C22 alkyl) 2) -O- (C1-
C22) alkylene-OH; -C (O) -NH- (C1-C22) alkylene-O-P (-N (C1-C22 alkyl) 2) -O- (C1-C30) alkylene-C (O) OH; -C (O) -NH- (C1-C22) alkylene-O-P (-N (C1-C22 alkyl) 2) -O- (C1-C22) alkylene-NH2; - (C1-C22) alkylene-C (O) -NH- (C1-C22) alkylene-O-P (-N (C1-C22 alkyl) 2) -O- (C1-C22) alkylene-CN; -(C1-C22) alkylene-C (O) -NH- (C1-C22) alkylene-O-P (-N (C1-C22 alkyl) 2) -O- (C1-C22) alkylene-OH; - (C1-C22) alkylene-C (O) -NH- (C1-C22) alkylene-O-P (-N (C1-C22 alkyl) 2) -O- (C1-C22) alkylene-C (O) OH; - (C1-C22) alkylene-C (O) -NH- (C1-C22) alkylene-O-P (-N (C1-C22 alkyl) 2) -O- (C1-C22) alkylene-NH2; - (C1-C22) alkylene -P (O) -OH; - (C1-C22) alkylene-P (O) (S) -OH and - (C1-C22) alkylene-CN. Each of the (C1-C22) alkyl or (C1-C22) alkylene included in B can be an alkyl or alkylene comprising from 1 to 22 carbon atoms, or from 2 to 20 carbon atoms, or from 3 to 16 carbon atoms, or from 4 to 12 carbon atoms, or from 6 to 12 carbon atoms, or from 8 to 10 carbon atoms.
In an embodiment, each of A1, A2 and A3 is either absent or a substituent independently selected from the group consisting of -H, -OH, -O-R1, -SH, - (C1-C25) alkyl, halogenated - (C1-C25) alkyl, - (C2-C22) alkenyl, - (C1-C22) alkylene-OH, - (C3-C22) cycloalkyl, - (C3-C22) cycloalkenyl, - (C1-C22) alkylene- (C3-C22) cycloalkyl, - (C1-C22) alkylene-R1, - (C1-C22) alkylene-O-R1, - (C1-C22) alkylene-COOR1, -C (O) O-R1, -O- (C1-C22) alkyl, -S- (C1-C22) alkyl, -C (O) -R1, -C (O) - (C1-C22) alkyl, -O-C (O) - (C1-C22) alkyl, -O-C (O) -R1, - (C1-C22) alkylene-O-C (O) -R1, -C (O) - (C1-C22) alkylene-OH, -C (O) - (C1-C22) alkylene-R1, -C (O) - (C1-C22) alkylene-NH-R1, -C (O) - (C1-C22) alkylene-NR2-R1, -O-C (O) - (C1-C22) alkylene-OH, -O-C (O) - (C1-C22) alkylene-R1, -adamantyl, - (C1-C22) alkylene-adamantyl, -O-adamantly, -C (O) - (C1-C22) alkylene-adamantyl, - (C1-C22) alkylene-C (O) - (C1-C22) alkylene-adamantyl, -NH-C (O) - (C1-C22) alkylene-adamantyl, - (C1-C22) alkylene-NH-C (O) - (C1-C22) alkyl, - (C1-C22) alkylene-NH-C (O) -halogenated (C1-C22) alkyl, -CH (NH-CO- (C1-C22) alkyl) - (C1-C22) alkylene-NH-C (O) - (C1-C22) alkyl, -C (O) - (C1-C22) alkylene-C (O) -NH-C [- (C1-C22) alkylene-O- (C1-C22) alkylene-C (O) -NH- (C1-C22) alkylene-NH-C (O) - (C1-C22) alkyl] 3, -C (O) - (C1-C22) alkylene-C (O) -NH-C [- (C1-C22) alkylene-O- (C1-C22) alkylene-C (O) -NH- (C1-C22) alkylene-NH-C (O) - (C1-C22) alkylene-R1] 3, -CH (NH-CO-halogenated (C1-C22) alkyl) - (C1-C22) alkylene-NH-C (O) -halogenated (C1-C22) alkyl, - (C1-C22) alkylene-NH-C (O) - (C1-C22) alkylene-adamantyl, - (C1-C22) alkylene-NR2-C (O) - (C1-C22) alkylene-adamantyl, - (C1-C22) alkylene- (C1-C6 alkylene oxide) (1-20) -NH-C (O) - (C1-C22) alkylene-adamantyl, -C (O) NH- (C1-C22) alkyl, -C (O) NH-R1, -C (O) NR2-R1, -C (O) NH- (C1-C22) alkylene-OH, -C (O) NH- (C1-C22) alkylene-COOH, -NH-C (O) - (C1-C22) alkyl, -NH-
C (O) -R1, -NR2-C (O) -R1, -O-P (O) 2-O-R1, -OP (O) (S) -O-R1, -O-P (O) -O-R1, -NH-R1, -NR2-R1, - (C1-C22) alkylene-NH-R1, - (C1-C22) alkylene-NR2-R1, -C (O) - (C1-C22) alkylene-C (O) -R1, -C (O) - (C1-C22) alkylene-C (O) O-R1, -C (O) - (C1-C22) alkylene-NH-C (O) -R1, -C (O) - (C1-C22) alkylene-NR2-C (O) -R1, - (C1-C22) alkylene-C (O) -R1, - (C1-C22) alkylene-NH-C (O) -R1, - (C1-C22) alkylene-NH-C (O) - (C1-C22) alkylene-R1, - (C1-C22) alkylene-NH-C (O) - (C1-C22) alkylene-NH-C (O) - (C1-C22) alkylene-R1, - (C1-C22) alkylene-C (O) -NH- (C1-C22) alkylene-R1, - (C1-C22) alkylene-C (O) -NH- (C1-C22) alkylene-C (O) -NH- (C1-C22) alkylene-R1, - (C1-C22) alkylene-C (O) -NH- (C1-C22) alkylene-NH-C (O) - (C1-C22) alkylene-R1, - (C1-C22) alkylene-NR2-C (O) - (C1-C22) alkylene-R1, - (C1-C22) alkylene-O-P (-N (C1-C22 alkyl) 2) -O- (C1-C22) alkylene-CN, - (C1-C22) alkylene-O-P (-N (C1-C22 alkyl) 2) -O- (C1-C22) alkylene-OH, - (C1-C22) alkylene-O-P (-N (C1-C22 alkyl) 2) -O- (C1-C22) alkylene-NH2, - (C1-C22) alkylene-O-P (-N (C1-C22 alkyl) 2) -O- (C1-C22) alkylene-C (O) OH, -C (O) -NH- (C1-C22) alkylene-O-P (-N (C1-C22 alkyl) 2) -O- (C1-C22) alkylene-CN, -C (O) -NH- (C1-C22) alkylene-O-P (-N (C1-C22 alkyl) 2) -O- (C1-C22) alkylene-OH, -C (O) -NH- (C1-C22) alkylene-O-P (-N (C1-C22 alkyl) 2) -O- (C1-C30) alkylene-C (O) OH, -C (O) -NH- (C1-C22) alkylene-O-P (-N (C1-C22 alkyl) 2) -O- (C1-C22) alkylene-NH2, - (C1-C22) alkylene-C (O) -NH- (C1-C22) alkylene-O-P (-N (C1-C22 alkyl) 2) -O- (C1-C22) alkylene-CN, - (C1-C22) alkylene-C (O) -NH- (C1-C22) alkylene-O-P (-N (C1-C22 alkyl) 2) -O- (C1-C22) alkylene-OH, - (C1-C22) alkylene-C (O) -NH- (C1-C22) alkylene-O-P (-N (C1-C22 alkyl) 2) -O- (C1-C22) alkylene-C (O) OH, - (C1-C22) alkylene-C (O) -NH- (C1-C22) alkylene-O-P (-N (C1-C22 alkyl) 2) -O- (C1-C22) alkylene-NH2, - (C1-C22) alkylene -P (O) -OH, - (C1-C22) alkylene-P (O) (S) -OH, - (C1-C22) alkylene-CN, substituted or unsubstituted pyrrole, substituted or unsubstituted pyrroline, substituted or unsubstituted pyrrolidine, substituted or unsubstituted pyrazole, substituted or unsubstituted pyrazoline, substituted or unsubstituted pyrazolidine, substituted or unsubstituted imidazole, substituted or unsubstituted oxazole, substituted or unsubstituted thiazole, substituted or unsubstituted benzopyrrole, substituted or unsubstituted benzopyrroline, substituted or unsubstituted benzopyrrolidine, substituted or unsubstituted benzopyrazole, substituted or unsubstituted benzopyrazoline, substituted or unsubstituted benzopyrazolidine, substituted or unsubstituted benzoimidazole, substituted or unsubstituted benzooxazole, substituted or unsubstituted benzothiazole, wherein when the above indicated rings are substituted, the substituents may include hydroxyl, halogen atom (such as fluorine, chlorine, bromine or iodine) , (C1-C16) alkyl, (C1-C12) alkoxy; and a substituent represented by Formula III,
In an embodiment, Y is selected from the group consisting of carbon, nitrogen, oxygen and sulfur; and each of P, Q, S and T is independently selected from the group consisting of carbon, nitrogen, oxygen and sulfur; and the asterisk refers to the site wherein the substituent represented by Formula III is linked with the structure represented by Formula I or Formula II;
In an embodiment, each of R3, R4 and R5 is either absent or a substituent independently selected from the group consisting of -H, -OH, -O-R1, -SH, - (C1-C25) alkyl, halogenated - (C1-C25) alkyl, - (C2-C22) alkenyl, - (C1-C22) alkylene-OH, - (C3-C22) cycloalkyl, - (C3-C22) cycloalkenyl, - (C1-C22) alkylene- (C3-C22) cycloalkyl, - (C1-C22) alkylene-R1, - (C1-C22) alkylene-O-R1, - (C1-C22) alkylene-COOR1, -C (O) O-R1, -O- (C1-C22) alkyl, -S- (C1-C22) alkyl, -C (O) -R1, -C (O) - (C1-C22) alkyl, -O-C (O) - (C1-C22) alkyl, -O-C (O) -R1, - (C1-C22) alkylene-O-C (O) -R1, -C (O) - (C1-C22) alkylene-OH, -C (O) - (C1-C22) alkylene-R1, -C (O) - (C1-C22) alkylene-NH-R1, -C (O) - (C1-C22) alkylene-NR2-R1, -O-C (O) - (C1-C22) alkylene-OH, -O-C (O) - (C1-C22) alkylene-R1, -adamantyl, - (C1-C22) alkylene-adamantyl, -O-adamantly, -C (O) - (C1-C22) alkylene-adamantyl, - (C1-C22) alkylene-C (O) - (C1-C22) alkylene-adamantyl, -NH-C (O) - (C1-C22) alkylene-adamantyl, - (C1-C22) alkylene-NH-C (O) - (C1-C22) alkyl, - (C1-C22) alkylene-NH-C (O) -halogenated (C1-C22) alkyl, -CH (NH-CO- (C1-C22) alkyl) - (C1-C22) alkylene-NH-C (O) - (C1-C22) alkyl, -C (O) - (C1-C22) alkylene-C (O) -NH-C [- (C1-C22) alkylene-O- (C1-C22) alkylene-C (O) -NH- (C1-C22) alkylene-NH-C (O) - (C1-C22) alkyl] 3, -C (O) - (C1-C22) alkylene-C (O) -NH-C [- (C1-C22) alkylene-O- (C1-C22) alkylene-C (O) -NH- (C1-C22) alkylene-NH-C (O) - (C1-C22) alkylene-R1] 3, -CH (NH-CO-halogenated (C1-C22) alkyl) - (C1-C22) alkylene-NH-C (O) -halogenated (C1-C22) alkyl, - (C1-C22) alkylene-NH-C (O) - (C1-C22) alkylene-adamantyl, - (C1-C22) alkylene-NR2-C (O) - (C1-C22) alkylene-adamantyl, - (C1-C22) alkylene- (C1-C6 alkylene oxide) (1-20) -NH-C (O) - (C1-C22) alkylene-adamantyl, -C (O) NH- (C1-C22) alkyl, -C (O) NH-R1, -C (O) NR2-R1, -C (O) NH- (C1-C22) alkylene-OH, -C (O) NH- (C1-C22) alkylene-COOH, -NH-C (O) - (C1-C22) alkyl, -NH-C (O) -R1, -NR2-C (O) -R1, -O-P (O) 2-O-R1, -OP (O) (S) -O-R1, -O-P (O) -O-R1, -NH-R1, -NR2-R1, - (C1-
C22) alkylene-NH-R1, - (C1-C22) alkylene-NR2-R1, -C (O) - (C1-C22) alkylene-C (O) -R1, -C (O) - (C1-C22) alkylene-C (O) O-R1, -C (O) - (C1-C22) alkylene-NH-C (O) -R1, -C (O) - (C1-C22) alkylene-NR2-C (O) -R1, - (C1-C22) alkylene-C (O) -R1, - (C1-C22) alkylene-NH-C (O) -R1, - (C1-C22) alkylene-NH-C (O) - (C1-C22) alkylene-R1, - (C1-C22) alkylene-NH-C (O) - (C1-C22) alkylene-NH-C (O) - (C1-C22) alkylene-R1, - (C1-C22) alkylene-C (O) -NH- (C1-C22) alkylene-R1, - (C1-C22) alkylene-C (O) -NH- (C1-C22) alkylene-C (O) -NH- (C1-C22) alkylene-R1, - (C1-C22) alkylene-C (O) -NH- (C1-C22) alkylene-NH-C (O) - (C1-C22) alkylene-R1, - (C1-C22) alkylene-NR2-C (O) - (C1-C22) alkylene-R1, - (C1-C22) alkylene-O-P (-N (C1-C22 alkyl) 2) -O- (C1-C22) alkylene-CN, - (C1-C22) alkylene-O-P (-N (C1-C22 alkyl) 2) -O- (C1-C22) alkylene-OH, - (C1-C22) alkylene-O-P (-N (C1-C22 alkyl) 2) -O- (C1-C22) alkylene-NH2, - (C1-C22) alkylene-O-P (-N (C1-C22 alkyl) 2) -O- (C1-C22) alkylene-C (O) OH, -C (O) -NH- (C1-C22) alkylene-O-P (-N (C1-C22 alkyl) 2) -O- (C1-C22) alkylene-CN, -C (O) -NH- (C1-C22) alkylene-O-P (-N (C1-C22 alkyl) 2) -O- (C1-C22) alkylene-OH, -C (O) -NH- (C1-C22) alkylene-O-P (-N (C1-C22 alkyl) 2) -O- (C1-C30) alkylene-C (O) OH, -C (O) -NH- (C1-C22) alkylene-O-P (-N (C1-C22 alkyl) 2) -O- (C1-C22) alkylene-NH2, - (C1-C22) alkylene-C (O) -NH- (C1-C22) alkylene-O-P (-N (C1-C22 alkyl) 2) -O- (C1-C22) alkylene-CN, - (C1-C22) alkylene-C (O) -NH- (C1-C22) alkylene-O-P (-N (C1-C22 alkyl) 2) -O- (C1-C22) alkylene-OH, - (C1-C22) alkylene-C (O) -NH- (C1-C22) alkylene-O-P (-N (C1-C22 alkyl) 2) -O- (C1-C22) alkylene-C (O) OH, - (C1-C22) alkylene-C (O) -NH- (C1-C22) alkylene-O-P (-N (C1-C22 alkyl) 2) -O- (C1-C22) alkylene-NH2, - (C1-C22) alkylene -P (O) -OH, - (C1-C22) alkylene-P (O) (S) -OH, - (C1-C22) alkylene-CN, substituted or unsubstituted pyrrole, substituted or unsubstituted pyrroline, substituted or unsubstituted pyrrolidine, substituted or unsubstituted pyrazole, substituted or unsubstituted pyrazoline, substituted or unsubstituted pyrazolidine, substituted or unsubstituted imidazole, substituted or unsubstituted oxazole, substituted or unsubstituted thiazole, substituted or unsubstituted benzopyrrole, substituted or unsubstituted benzopyrroline, substituted or unsubstituted benzopyrrolidine, substituted or unsubstituted benzopyrazole, substituted or unsubstituted benzopyrazoline, substituted or unsubstituted benzopyrazolidine, substituted or unsubstituted benzoimidazole, substituted or unsubstituted benzooxazole, and substituted or unsubstituted benzothiazole, wherein when the above indicated rings are substituted, the substituents may include hydroxyl, halogen atom (such as fluorine, chlorine, bromine or iodine) , (C1-C16) alkyl, (C1-C12) alkoxy.
In an embodiment, R7, on each occurrence, is attached to any one of P, Q, S and T, and is either absent or selected from the group consisting of hydrogen, halogen atom, hydroxyl,
(C1-C20) alkyl, (C1-C20) alkoxy, halogenated (C1-C20) alkyl and halogenated (C1-C20) alkoxy.
In an embodiment, M is an integer of 0, 1, 2 or 3.
In an embodiment, R6 is attached to any one of P, Q, S and T, and is selected from the group consisting of direct bond, -O-, -C (O) O-, -O-C (O) -, -P (O) 2-O-, -O-P (O) 2-O-, -P (O) (S) -O-, -O-P (O) (S) -O-, -O-P (O) -O-, - (C1-C22) alkylene-, - (C1-C22) alkylene-O-, -O- (C1-C22) alkylene-, - (C1-C22) alkylene-NH-, -NH- (C1-C22) alkylene-, -C (O) - (C1-C22) alkylene-, - (C1-C22) alkylene-C (O) -, -C (O) -O- (C1-C22) alkylene-, - (C1-C30) alkylene-C (O) -O-, -C (O) -NH- (C1-C22) alkylene-, -C (O) -NH- (C1-C22) alkylene-C (O) -O-, -C (O) -NH- (C1-C22) alkylene-O-C (O) -, -C (O) -NH- (C1-C22) alkylene-O-C (O) -O-, -C (O) -NH- (C1-C22) alkylene-O-, -C (O) -N ( (C1-C22) alkyl) - (C1-C22) alkylene-, -C (O) -N ( (C1-C22) alkyl) - (C1-C22) alkylene-O-, -C (O) -NH- (C1-C22) alkylene-C (O) -NH- (C1-C22) alkylene-, -C (O) -NH- (C1-C22) alkylene-NH-C (O) - (C1-C22) alkylene-, -C (O) -NH- (C1-C22) alkylene-C (O) -NH-, -C (O) -N ( (C1-C22) alkyl) - (C1-C22) alkylene-C (O) -N ( (C1-C22) alkyl) - (C1-C22) alkylene-, -C (O) -N ( (C1-C22) alkyl) - (C1-C22) alkylene-C (O) -N ( (C1-C22) alkyl) -, - (C1-C22) alkylene-C (O) -NH- (C1-C22) alkylene-C (O) -NH- (C1-C22) alkylene-, - (C1-C22) alkylene-C (O) -NH- (C1-C22) alkylene-C (O) -NH-, -NH-C (O) - (C1-C22) alkylene-, -NH-C (O) - (C1-C22) alkylene-C (O) -O-, -NH-C (O) - (C1-C22) alkylene-C (O) -, -NH-C (O) - (C1-C22) alkylene-O-, -N ( (C1-C22) alkyl) -C (O) - (C1-C22) alkylene-, -N ( (C1-C22) alkyl) -C (O) - (C1-C22) alkylene-O-, -NH-C (O) - (C1-C22) alkylene-NH-C (O) - (C1-C22) alkylene-, -NH-C (O) - (C1-C22) alkylene-NH-C (O) -, -N ( (C1-C22) alkyl) -C (O) - (C1-C22) alkylene-N ( (C1-C22) alkyl) -C (O) - (C1-C22) alkylene-, -N ( (C1-C22) alkyl) -C (O) - (C1-C22) alkylene-N ( (C1-C22) alkyl) -C (O) -, - (C1-C22) alkylene-NH-C (O) - (C1-C22) alkylene-NH-C (O) - (C1-C22) alkylene-, - (C1-C22) alkylene-NH-C (O) - (C1-C22) alkylene-NH-C (O) -, - (C1-C22) alkylene -P (O) 2-O-, - (C1-C22) alkylene-O-P (O) 2-O-, - (C3-C22) cycloalkylene-, - (C3-C22) cycloalkylene-O-, -O- (C3-C22) cycloalkylene-, - (C6-C22) arylene-, - (C6-C22) arylene-O-, -O- (C6-C22) arylene-, - (C6-C22) arylene-NH-, -NH- (C6-C22) arylene-, -C (O) - (C6-C22) arylene-, - (C6-C22) arylene-C (O) -, -C (O) -O- (C6-C22) arylene-, - (C6-C22) arylene-C (O) -O-, -C (O) -NH- (C6-C22) arylene-and -C (O) -NH- (C6-C22) arylene-C (O) -O-.
In an embodiment, R1, on each occurrence, is independently selected from the group consisting of hydrogen, hydroxyl, - (C1-C22) alkyl, - (C3-C22) cycloalkyl, - (C6-C22) aryl, - (C1-C22) alkoxy, - (C3-C22) cycloalkoxy, - (C6-C22) aryloxy, -C (O) - (C1-C22) alkyl, -OC (O) (C1-C22) alkyl, -C (O) -O- (C1-C22) alkyl, -C (O) - (C3-C22) cycloalkyl, -OC (O) - (C3-C22) cycloalkyl, -C (O) -O- (C3-C22) cycloalkyl, -C (O) - (C6-C22) aryloxy, -OC (O) - (C6-C22) aryloxy, -C (O) -O- (C6-
C22) aryloxy, -C (O) -phosphate ester group, phosphodiester group, phosphoramidite group, saturated fatty acid group, unsaturated fatty acid group, glucosyl, acetamide glucosyl, acetylglucosamine, lipid, PEG, steroid, lipophile, carbohydrate, cholesterol, adamantane, amino acid, peptide, chloroquine and alkaloid.
In an embodiment, R2, on each occurrence, is independently selected from the group consisting of a halogen atom, a (C1-C12) alkyl, a (C1-C12) alkoxy, a (C1-C12) alkoxycarbonyl, a (C6-C16) aryl or a (C6-C16) aryloxycarbonyl.
In an embodiment, one or more hydroxyl group, carboxyl group, amino group, nitrile group and phosphoric acid group contained in A1, A2, A3, B, C, R1, R2, R3, R4, R5 and R7 can be protected with a terminal protective group RP selected from the group consisting of (C1-C22) alkyl, (C1-C22) alkoxy, (C1-C22) alkylcarbonyl, (C1-C22) alkoxycarbonyl, (C6-C22) aryl, (C6-C22) aryloxy, (C6-C22) arylcarbonyl, (C6-C22) aryloxycarbonyl, tri ( (C1-C22) alkyl) silyl and tri ( (C1-C22) alkoxy) silyl, wherein the (C1-C22) alkyl contained in the protection group Rp can be an alkyl comprising from 1 to 22 carbon atoms, such as from 2 to 20 carbon atoms, or from 3 to 18 carbon atoms, or from 4 to 16 carbon atoms, or from 6 to 12 carbon atoms; the (C1-C22) alkoxy contained in Rp can be an alkoxy comprising from 1 to 22 carbon atoms, such as from 2 to 20 carbon atoms, or from 3 to 18 carbon atoms, or from 4 to 16 carbon atoms, or from 6 to 12 carbon atoms; the (C6-C22) aryl contained in Rp can be an aryl comprising from 6 to 22 carbon atoms, such as from 6 to 20 carbon atoms, or from 6 to 18 carbon atoms, or from 6 to 16 carbon atoms, or from 6 to 12 carbon atoms, or from 8-10 atoms; and the (C6-C22) aryloxy contained in Rp can be an aryloxy comprising from 6 to 22 carbon atoms, such as from 6 to 20 carbon atoms, or from 6 to 18 carbon atoms, or from 6 to 16 carbon atoms, or from 6 to 12 carbon atoms, or from 8-10 atoms.
In an embodiment, A1, A2 and A3 are not simultaneously hydrogen and R3, R4 and R5 are not simultaneously hydrogen. In an exemplary embodiment, X is carbon, and all of A1, A2 and A3 are present. In another embodiment, X is nitrogen, both A1 and A2 are present and A3 is absent. In another embodiment, X is nitrogen with positive charge (i.e. quaternary ammonium) , and all of A1, A2 and A3 are present. In another embodiment, X is sulfur or oxygen, both A1 and A3 are absent and A2 is present. In another embodiment, X is sulfur with positive charge (i.e. sulfonium) , both A2 and A3 are present and A1 is absent.
In an embodiment, each of the (C1-C22) alkyl or (C1-C22) alkylene included in A1, A2, A3,
R1, R2, R3, R4, R5, R6 and R7 can be an alkyl or alkylene comprising from 1 to 22 carbon atoms, or from 2 to 20 carbon atoms, or from 3 to 16 carbon atoms, or from 4 to 12 carbon atoms, or from 6 to 12 carbon atoms, or from 8 to 10 carbon atoms.
In an embodiment, one or more hydroxyl group, carboxyl group, amino group, nitrile group and phosphoric acid group contained in A1, A2, A3, B, C, R1, R2, R3, R4, R5 and R7 can be linked with a support material selected from the group consisting of silica, silica gel, glass, ceramic, polymer, cellulose, and combinations thereof. In some embodiments, the solid material is in the form of a bead. The bead may be made of any material including, without limitation, magnetic bead, paramagnetic bead, silica bead, an agarose bead, etc.
In another embodiment, the compound of the present disclosure may have a structure represented by any of Formula AIV to Formula AXIII,
wherein A1, A2, A3, B, C, F, G, H, I, m and n are as defined above.
In an embodiment, the compound of the present disclosure has a structure represented by Formula BII
wherein X′ is selected from the group consisting of carbon, nitrogen, oxygen and sulfur; and each of F′, G′, H′ and I′ is independently selected from the group consisting of carbon, nitrogen, oxygen and sulfur.
In another embodiment, each of A1′, A2′ and A3′ is either absent or a substituent independently selected from the group consisting of -H, -R1′, -O-R1′, -S-R1′, -C (O) -R1′, -C (O) O-R1′, -O-C (O) -R1′, -C (O) NH-R1′, -C (O) NR2′-R1′, -NH-C (O) -R1′, -NR2′-C (O) -R1′, -O-P (O) 2-O-R1′, -OP (O) (S) -O-R1′, -O-P (O) -O-R1′, -NH-R1′, -NR2′-R1′, - (CH2) r′-NH-R1′, - (CH2) r′-NR2′-R1′, -C (O) - (CH2) r′-R1′, -C (O) - (CH2) r′-NH-R1′, -C (O) - (CH2) r′-NR2′-R1′, -C (O) - (CH2) r′-C (O) -R1′, -C (O) - (CH2) r′-C (O) O-R1′, -C (O) - (CH2) r′-NH-C (O) -R1′, -C (O) - (CH2) r′-NR2′-C (O) -R1′, - (CH2) r′-C (O) -R1′; - (CH2) r′-C (O) O-R1′; - (CH2) r′-O-C (O) -R1′, - (CH2) r′-R1′, - (CH2) r′-NH-C (O) -R1′, - (CH2) r′-NH-C (O) - (CH2) s′-R1′, - (CH2) r′-NH-C (O) - (C1-C22) alkylene-NH-C (O) - (CH2) s′-R1′, - (CH2) r′-C (O) -NH- (CH2) s′-R1′, - (CH2) r′-C (O) -NH- (C1-C22) alkylene-C (O) -NH- (CH2) s′-R1′, - (CH2) r′-C (O) -NH- (C1-C22) alkylene-NH-C (O) - (CH2) s′-R1′, - (CH2) r′-NR2′-C (O) - (CH2) s′-R1′, -CH (- (CH2) r′-NH-C (O) - (CH2) s′-R1′) (-NH-C (O) - (CH2) q′-R3′) , -CH (- (CH2) r′-C (O) -NH- (CH2) s′-R1′) (-C (O) -NH- (CH2) q′-R3′) , -N (- (CH2) r′-NH-C (O) - (CH2) s′-R1′) (-NH-C (O) - (CH2) q′-R3′) , -CH (- (CH2) r′-NH-C (O) - (CH2) s′-R1′) (- (CH2) p′-NH-C (O) - (CH2) q′-R3′) , -CR4′ (- (CH2) r′-NH-C (O) - (CH2) s′-R1′) (-NH-C (O) - (CH2) q′-R3′) , -CR4′ (- (CH2) r′-NH-C (O) - (CH2) s′-R1′) (- (CH2) p′-NH-C (O) - (CH2) q′-R3′) , -CH (- (CH2) r′-NR5′-C (O) - (CH2) s′-R1′) (-NR6′-C (O) - (CH2) q′-R3′) , -CH (- (CH2) r′-NR5′-C (O) - (CH2) s′-R1′) (- (CH2) p′-NR6′-C (O) - (CH2) q′-R3′) , -CR4′ ( (CH2) r′-NR5′-C (O) - (CH2) s′-R1′) ( (CH2) p′-NR6′-C (O) - (CH2) q′-R3′) , -CR4′ ( (CH2) r′-NR5′-C (O) - (CH2) s′-R1′) ( (CH2) p′-NR6′-C (O) - (CH2) q′-R3′) , or A1′ and A2′ can be linked together so that A1′, A2′, the nitrogen atom linked with A1′ and the carbon atom linked with A2′ form a unsubstituted or substituted heterocyclic ring. Each of the (C1-C22) alkylene included in A1′, A2′ and A3′ can be (C2-C20) alkylene, (C3-C16) alkylene, (C4-C14) alkylene, (C6-C12) alkylene, or (C8-C10) alkylene. Each of R1′ and R3′ is independently selected from the group consisting of hydrogen; hydroxyl;
- (C1-C30) alkyl, such as - (C2-C25) alkyl, or - (C2-C22) alkyl, or - (C3-C18) alkyl, or - (C4-C16) alkyl, or - (C6-C12) alkyl, or - (C8-C10) alkyl; - (C3-C50) cycloalkyl, such as - (C4-C40) cycloalkyl, or - (C5-C30) cycloalkyl, or - (C6-C20) cycloalkyl, or - (C6-C16) cycloalkyl; - (C6-C50) aryl, such as - (C6-C40) aryl, or - (C6-C30) aryl, or - (C6-C25) aryl, or - (C6-C22) aryl, or - (C6-C16) aryl, or - (C6-C12) aryl; - (C1-C30) alkoxy, such as - (C2-C22) alkoxy, or - (C3-C20) alkoxy, or - (C4-C16) alkoxy, or - (C6-C12) alkoxy; - (C3-C50) cycloalkoxy, such as - (C4-C40) cycloalkoxy, or - (C5-C30) cycloalkoxy, or - (C6-C20) cycloalkoxy, or - (C6-C16) cycloalkoxy; - (C6-C50) aryloxy, such as - (C6-C40) aryloxy, or - (C6-C30) aryloxy, or - (C6-C25) aryloxy, or - (C6-C22) aryloxy, or - (C6-C16) aryloxy, or - (C6-C12) aryloxy; -C (O) - (C1-C30) alkyl, such as -C (O) - (C2-C22) alkyl, or -C (O) - (C3-C18) alkyl, or -C (O) - (C4-C16) alkyl, or -C (O) - (C6-C12) alkyl, or -C (O) - (C8-C10) alkyl; -OC (O) (C1-C30) alkyl, such as -OC (O) (C2-C22) alkyl, or -OC (O) (C3-C18) alkyl, or -OC (O) (C4-C16) alkyl, or -OC (O) (C6-C12) alkyl, or -OC (O) (C8-C10) alkyl; -C (O) -O- (C1-C30) alkyl, such as -C (O) -O- (C2-C22) alkyl, or -C (O) -O- (C3-C18) alkyl, or -C (O) -O- (C4-C16) alkyl, or -C (O) -O- (C6-C12) alkyl, or -C (O) -O- (C8-C10) alkyl; -C (O) - (C3-C50) cycloalkyl, such as -C (O) - (C4-C40) cycloalkyl, or -C (O) - (C5-C30) cycloalkyl, or -C (O) - (C6-C20) cycloalkyl, or -C (O) - (C6-C16) cycloalkyl; -OC (O) - (C3-C50) cycloalkyl, such as -OC (O) - (C4-C40) cycloalkyl, or -OC (O) - (C5-C30) cycloalkyl, or -OC (O) - (C6-C20) cycloalkyl, or -OC (O) - (C6-C16) cycloalkyl; -C (O) -O- (C3-C50) cycloalkyl, such as -C (O) -O- (C4-C40) cycloalkyl, or -C (O) -O- (C5-C30) cycloalkyl, or -C (O) -O- (C6-C20) cycloalkyl, or -C (O) -O- (C6-C16) cycloalkyl; -C (O) - (C6-C50) aryloxy, such as -C (O) - (C6-C40) aryloxy, or -C (O) - (C6-C30) aryloxy, or -C (O) - (C6-C25) aryloxy, or -C (O) - (C6-C22) aryloxy, or -C (O) - (C6-C16) aryloxy, or -C (O) - (C6-C12) aryloxy; -OC (O) - (C6-C50) aryloxy, such as -OC (O) - (C6-C40) aryloxy, or -OC (O) - (C6-C30) aryloxy, or -OC (O) - (C6-C25) aryloxy, or -OC (O) - (C6-C22) aryloxy, or -OC (O) - (C6-C16) aryloxy, or -OC (O) - (C6-C12) aryloxy; -C (O) -O- (C6-C50) aryloxy, such as -C (O) -O- (C6-C40) aryloxy, or -C (O) -O- (C6-C30) aryloxy, or -C (O) -O- (C6-C25) aryloxy, or -C (O) -O- (C6-C22) aryloxy, or -C (O) -O- (C6-C16) aryloxy, or - (C6-C12) aryloxy; -C (O) -phosphate ester group, phosphodiester group, phosphoramidite group, saturated fatty acid group, unsaturated fatty acid group, glucosyl, N- acetamide glucosyl, acetyl galactosamine, lipid, PEG, steroid, lipophile, carbohydrate, cholesterol, adamantane, amino acid, peptide, ligand, nucleic acid, oligonucleotide, aptamer, small molecule, antibody, antibody fragment, chloroquine, alkaloid and the targeting moiety as stated herein.
In some embodiments, one or more of the substituents A1′, A2′ and A3′ contain at least
one targeting moiety. The term “targeting moiety” used herein refers to a part or segment of a molecule acting as a chemical signal that binds to a molecule or complex on a particular area of a cell, tissue, or organ. In general, targeting moiety modifies one or more properties of the attached oligonucleotide of the invention including but not limited to pharmacodynamic, pharmacokinetic, binding, absorption, cellular distribution, cellular uptake, charge and clearance. Targeting moieties are routinely used in the arts and are linked directly or via an optional linking moiety to a parent compound such as an oligomeric compound.
In certain embodiments, a targeting moiety is selected from one or more of a ligand, a peptide, nucleic acid, oligonucleotide, aptamer, small molecule, a polyethylene glycol, an amino acid, a cholesterol, a carbohydrate (e.g. glucose, galactosamine or N-acetyl galactosamine) , an antibody or antibody fragment, and localization signal such as a nuclear localization signal or a mitochondrial localization signal. In certain embodiments, a targeting moiety is selected from the group consisting of intercalators, reporter molecules, polyamines, polyamides, vitamin moieties, polyethylene glycols, thioethers, polyethers, thiocholesterols, cholic acid moieties, folate, lipids, fatty acids, phospholipids, biotin, phenazine, phenanthridine, anthraquinone, adamantane, acridine, fluoresceins, rhodamines, coumarins, fluorophores, luminescent proteins and dyes. When the targeting moiety is a fluorophore, any fluorophore deemed useful may be utilized. Non-limiting examples of useful fluorescent proteins include but are not limited to GFP, EBFP, Azurite, Cerulean, mCFP, , Turquoise, ECFP, mKeima-Red, TagCFP, AmCyan, mTFP, TurboGFP, TagGFP, EGFP, TagYFP, EYFP, Topaz, Venus, mCitrine, TurboYFP, mOrange, TurboRFP, tdTomato, TagRFP, dsRed2, mRFP, mCherry, mPlum mRaspberry, mScarlet, etc. Examples of luminescent proteins, include without limitation, Cypridinia luciferase, Gaussia luciferase, Renilla luciferase, Phontinus luciferase, Luciola luciferase, Pyrophorus luciferase, Phrixothrix luciferase, etc.
In certain embodiments, a targeting moiety is selected from: a cell-penetrating peptide, polyethylene glycol, an alkaloid, a tryptamine, a benzimidazole, a quinolone, an amino acid, a cholesterol, carbohydrate, and ligand.
In one embodiment, a targeting moiety is a carbohydrate. The carbohydrate can be selected from the group consisting of monosaccharides, disaccharides, trisaccharides, and polysaccharides. In some embodiments, the carbohydrate is a monosaccharide selected from the group consisting of dextrose, glucose, galactose, mannitol, D-mannose, sorbitol, and
sorbose. In another embodiment the carbohydrate is a disaccharide selected from the group consisting of lactose, maltose, sucrose, and trehalose. In one embodiment, a targeting moiety is a polysaccharide. In one embodiment, a targeting moiety is a N-acetyl galactosamine.
In another embodiment, a targeting moiety is an amino acid. In one embodiment, the amino acid is a hydrophobic amino acid. In some embodiments, the hydrophobic amino acid is selected from the group consisting of alanine, isoleucine, leucine, methionine, phenylalanine, proline, tryptophan, and valine. In yet another embodiment, the amino acid is a polar amino acid. In some embodiments, the amino acid is selected from the group consisting of arginine, histidine, lysine, cysteine, glycine, glutamine, serine, threonine, tyrosine, aspartic acid and glutamic acid.
In one embodiment, a targeting moiety is selected from the group consisting of human serum albumin, α-lactalbumin, trypsinogen, and polyalanine.
In certain embodiments, the targeting moiety comprises an active drug substance, for example, aspirin, warfarin, phenylbutazone, ibuprofen, suprofen, fen-bufen, ketoprofen, (S) - (+) -pranoprofen, carprofen, dansylsarcosine, 2, 3, 5-triiodobenzoic acid, fingolimod, flufenamic acid, folinic acid, a benzothiadiazide, chlorothiazide, a diazepine, indo-methicin, a barbiturate, a cephalosporin, a sulfa drug, an antidiabetic, an antibacterial or an antibiotic.
The targeting moieties disclosed herein can be used to increase uptake, introduce, delivery and target uptake of oligonucleotide to particular cell or tissue types, e.g., cells or tissues in central nervous system (e.g., brain and spinal cord) , liver, lung, kidney, intestine, pancreas, cholecyst, heart, lymph nodes, spleen, stomach, bladder, muscle and bone. Preferred targeting moieties include those specifically provided in the Examples.
In an embodiment, at least one of R1′ and R3′ is the targeting moiety. In an embodiment, all of the R1′s contained in the A1′, A2′ and A3′ are the targeting moiety as stated above; In another embodiment, all of the R3′ contained in the A1′, A2′ and A3′ are the targeting moiety as stated above; In an embodiment, all of the R1′ and R3′ contained in the A1′, A2′ and A3′ are the targeting moiety as stated above.
In some embodiments, one or more hydroxyl group, carboxyl group and amino group contained in each of R1′ and R3′ can be optionally protected, e.g. with a terminal protection group RP selected from the group consisting of (C1-C22) alkyl, (C1-C22) alkoxy, (C1-C22) alkylcarbonyl, (C1-C22) alkoxycarbonyl, (C6-C22) aryl, (C6-C22) aryloxy, (C6-
C22) arylcarbonyl, (C6-C22) aryloxycarbonyl, tri ( (C1-C22) alkyl) silyl and tri ( (C1-C22) alkoxy) silyl, wherein the (C1-C22) alkyl contained in the protection group Rp can be an alkyl comprising from 1 to 22 carbon atoms, such as from 2 to 20 carbon atoms, or from 3 to 18 carbon atoms, or from 4 to 16 carbon atoms, or from 6 to 12 carbon atoms; the (C1-C22) alkoxy contained in Rp can be an alkoxy comprising from 1 to 22 carbon atoms, such as from 2 to 20 carbon atoms, or from 3 to 18 carbon atoms, or from 4 to 16 carbon atoms, or from 6 to 12 carbon atoms; the (C6-C22) aryl contained in Rp can be an aryl comprising from 6 to 22 carbon atoms, such as from 6 to 20 carbon atoms, or from 6 to 18 carbon atoms, or from 6 to 16 carbon atoms, or from 6 to 12 carbon atoms, or from 8-10 atoms; and the (C6-C22) aryloxy contained in Rp can be an aryloxy comprising from 6 to 22 carbon atoms, such as from 6 to 20 carbon atoms, or from 6 to 18 carbon atoms, or from 6 to 16 carbon atoms, or from 6 to 12 carbon atoms, or from 8-10 atoms.
In some embodiments, each of R2′, R4′, R5′ and R6′ is independently a halogen atom, such as fluorine, chlorine, bromine or iodine; a (C1-C12) alkyl, such as (C1-C10) alkyl, or (C2-C8) alkyl, or (C3-C6) alkyl, or (C4-C5) alkyl; a (C1-C12) alkoxy, such as (C1-C10) alkoxy, or (C2-C8) alkoxy, or (C3-C6) alkoxy, or (C4-C5) alkoxy; a (C1-C12) alkoxycarbonyl, such as (C1-C10) alkoxycarbonyl, or (C2-C8) alkoxycarbonyl, or (C3-C6) alkoxycarbonyl, or (C4-C5) alkoxycarbonyl; a (C6-C16) aryl, such as - (C6-C12) aryl, or - (C6-C10) aryl; or a (C6-C16) aryloxycarbonyl, such as (C6-C12) aryloxycarbonyl, or (C6-C10) aryloxycarbonyl, or (C6-C8) aryloxycarbonyl.
In some embodiments, each of r′, s′, p′ and q′ is an integer from 1 to 22, such as an integer from 2 to 20, or an integer from 3 to 18, an integer from 4 to 16, an integer from 6 to 12, an integer from 8 to 10.
In another embodiment, A3′ is absent when X′ is oxygen, and A1′, A2′ and A3′ are not simultaneously hydrogen. In an exemplary embodiment, X′ is carbon, and all of A1′, A2′ and A3′ are present. In another embodiment, X′ is nitrogen, both A1′ and A2′ are present and A3′ is absent. In another embodiment, X′ is nitrogen with positive charge (i.e. quaternary ammonium) , and all of A1′, A2′ and A3′ are present. In another embodiment, X′ is sulfur or oxygen, both A1′ and A3′ are absent and A2′ is present. In another embodiment, X′ is sulfur with positive charge (i.e. sulfonium) , both A2′ and A3′ are present and A1′ is absent.
In another embodiment, each C′ is attached to any one of F′, G′, H′ and I′, and is either absent or selected from the group consisting of hydrogen; halogen atom, such as fluorine, chlorine, bromine or iodine; hydroxyl; - (C1-C20) alkyl, such as - (C2-C19) alkyl, or - (C3-C18) alkyl, or - (C4-C16) alkyl, or - (C6-C12) alkyl, or - (C8-C10) alkyl; - (C1-C20) alkoxy, such as - (C2-C19) alkoxy, or - (C3-C18) alkoxy, or - (C4-C16) alkoxy, or - (C6-C12) alkoxy; halogenated (C1-C20) alkyl, such as halogenated (C2-C19) alkyl, or halogenated (C3-C18) alkyl, or halogenated (C4-C16) alkyl, or halogenated (C6-C12) alkyl, or halogenated (C8-C10) alkyl; and halogenated (C1-C20) alkoxy, such as halogenated (C1-C20) alkoxy, such as halogenated (C2-C19) alkoxy, or halogenated (C3-C18) alkoxy, or halogenated (C4-C16) alkoxy, or halogenated (C6-C12) alkoxy, or halogenated (C8-C10) alkoxy; wherein “halogenated” can be fluorinated, chlorinated, brominated, iodinated, or combinations thereof.
In an embodiment, m′ is an integer of 1, 2 or 3, n′ is an integer of 1, 2 or 3, and m′+n′=4. In some exemplary embodiments, m′=1 and n′=3; or m′=2 and n′=2; or m′=3 and n′=1.
In an embodiment, each B′ is attached to any one of F′, G′, H′ and I′, while C′ is attached to the rest of F′, G′, H′ and I′. In an embodiment, B′ is attached to H′ and three C′s are separately attached to each of F′, G′ and I′. In another embodiment, B′ is attached to G′ and three C′s are separately attached to each of F′, H′ and I′. In another embodiment, B′ is attached to F′ and three C′s are separately attached to each of G′, H′ and I′. In another embodiment, B′ is attached to I and three C′s are separately attached to each of F′, G′ and H′. In another embodiment, two B′s are separately attached to G′ and H′, and two C′s are separately attached to each of F′ and I′.
In another embodiment, each of B′ is independently selected from the group consisting of hydroxyl, -C (O) OH, - (C1-C30) alkoxy, -P (O) 2-OH, -P (O) -OH, -P (O) (S) -OH, -CN, - (C1-C30) alkylene-OH, - (C3-C50) cycloalkylene-OH, - (C6-C50) arylene-OH, -C (O) -NH- [ (C1-C30) alkylene-O] r′-H (wherein r′ is an integer of 1 to 22) , -C (O) -NH- [ (C1-C30) alkylene-O] r′- (C1-C30) alkylene-C (O) -OH (wherein r′ is an integer of 1 to 22) , - (C5-C50) heteroarylene-OH, - (C1-C30) alkylene-C (O) OH, - (C3-C50) cycloalkylene-C (O) OH, - (C6-C50) arylene-C (O) OH, -C (O) -NH- (C1-C30) alkylene- (C6-C50) arylene- (C1-C30) alkylene-C (O) OH, - (C5-C50) heteroarylene-C (O) OH, -C (O) -NH- (C1-C30) alkylene-OH, -C (O) -NH- (C3-C50) cycloalkylene-OH, -C (O) -NH-
(C6-C50) arylene-OH, -C (O) -NH- (C5-C50) heteroarylene-OH, -C (O) -NH- (C1-C30) alkylene-C (O) OH, -C (O) -NH- (C3-C50) cycloalkylene-C (O) OH, -C (O) -NH- (C6-C50) arylene-C (O) OH, -C (O) -NH- (C5-C50) heteroarylene-C (O) OH, - (C1-C30) alkylene-C (O) -NH- (C1-C30) alkylene-C (O) OH, - (C1-C30) alkylene-C (O) -NH- (C3-C50) cycloalkylene-C (O) OH, - (C1-C30) alkylene-C (O) -NH- (C6-C50) arylene-C (O) OH, - (C1-C30) alkylene-C (O) -NH- (C5-C50) heteroarylene-C (O) OH, - (C1-C30) alkylene-P (O) 2-OH, - (C3-C50) cycloalkylene-P (O) 2-OH, - (C6-C50) arylene-P (O) 2-OH, - (C5-C50) heteroarylene-P (O) 2-OH, -C (O) -NH- (C1-C30) alkylene-O-P (-N (C1-C16alkyl) 2) -O- (C1-C30) alkylene-CN, -C (O) -NH- (C1-C30) alkylene-O-P (-N (C1-C16alkyl) 2) -O- (C1-C30) alkylene-OH, -C (O) -NH- (C1-C30) alkylene-O-P (-N (C1-C16alkyl) 2) -O- (C1-C30) alkylene-C (O) OH, -C (O) -NH- (C1-C30) alkylene-O-P (-N (C1-C16alkyl) 2) -O- (C1-C30) alkylene-NH2, - (C1-C30) alkylene-C (O) -NH- (C1-C30) alkylene-O-P (-N (C1-C16alkyl) 2) -O- (C1-C30) alkylene-CN, - (C1-C30) alkylene-C (O) -NH- (C1-C30) alkylene-O-P (-N (C1-C16alkyl) 2) -O- (C1-C30) alkylene-OH, - (C1-C30) alkylene-C (O) -NH- (C1-C30) alkylene-O-P (-N (C1-C16alkyl) 2) -O- (C1-C30) alkylene-C (O) OH, - (C1-C30) alkylene-C (O) -NH- (C1-C30) alkylene-O-P (-N (C1-C16alkyl) 2) -O- (C1-C30) alkylene-NH2, - (C1-C30) alkylene -P (O) -OH, - (C3-C50) cycloalkylene-P (O) -OH, - (C6-C50) arylene-P (O) -OH, - (C5-C50) -heteroarylene-P (O) -OH, - (C1-C30) alkylene-P (O) (S) -OH, - (C3-C50) cycloalkylene-P (O) (S) -OH, - (C6-C50) arylene-P (O) (S) -OH, - (C5-C50) heteroarylene-P (O) (S) -OH, - (C1-C30) alkylene-CN, - (C3-C50) cycloalkylene-CN, - (C6-C50) arylene-CN, - (C5-C50) heteroarylene-CN, lipid, PEG, steroid, lipophile, carbohydrate, cholesterol, adamantane, amino acid, peptide, chloroquine, alkaloid and a substituent represented by Formula BIII:
wherein Y′ is selected from the group consisting of carbon, nitrogen, oxygen and sulfur; and each of P′, Q′, S′ and T′ is independently selected from the group consisting of carbon, nitrogen, oxygen and sulfur; and the asterisk refers to the site wherein the substituent represented by Formula BIII is linked with any one of F′, G′, H′ and I′ of Formula BII.
In some embodiments, R7′ is selected from the group consisting of -O-, -C (O) O-, -O-C (O) -, -P (O) 2-O-, -O-P (O) 2-O-, -P (O) (S) -O-, -O-P (O) (S) -O-, -O-P (O) -O-, - (C1-C30) alkylene-,
- (C1-C30) alkylene-O-, -O- (C1-C30) alkylene-, - (C1-C30) alkylene-NH-, -NH- (C1-C30) alkylene-, -C (O) - (C1-C30) alkylene-, - (C1-C30) alkylene-C (O) -, -C (O) -O- (C1-C30) alkylene-, - (C1-C30) alkylene-C (O) -O-, -C (O) -NH- (C1-C30) alkylene-, -C (O) -NH- (C1-C30) alkylene-C (O) -O-, -C (O) -NH- (C1-C30) alkylene-O-C (O) -, -C (O) -NH- (C1-C30) alkylene-O-C (O) -O-, -C (O) -NH- (C1-C30) alkylene-O-, -C (O) -N ( (C1-C20) alkyl) - (C1-C30) alkylene-, -C (O) -N ( (C1-C20) alkyl) - (C1-C30) alkylene-O-, -C (O) -NH- (C1-C30) alkylene-C (O) -NH- (C1-C30) alkylene-, -C (O) -NH- (C1-C30) alkylene-NH-C (O) - (C1-C30) alkylene-, -C (O) -NH- (C1-C30) alkylene-C (O) -NH-, -C (O) -N ( (C1-C20) alkyl) - (C1-C30) alkylene-C (O) -N ( (C1-C20) alkyl) - (C1-C30) alkylene-, -C (O) -N ( (C1-C20) alkyl) - (C1-C30) alkylene-C (O) -N ( (C1-C20) alkyl) -, - (C1-C30) alkylene-C (O) -NH- (C1-C30) alkylene-C (O) -NH- (C1-C30) alkylene-, - (C1-C30) alkylene-C (O) -NH- (C1-C30) alkylene-C (O) -NH-, -NH-C (O) - (C1-C30) alkylene-, -NH-C (O) - (C1-C30) alkylene-C (O) -O-, -NH-C (O) - (C1-C30) alkylene-C (O) -, -NH-C (O) - (C1-C30) alkylene-O-, -N ( (C1-C20) alkyl) -C (O) - (C1-C30) alkylene-, -N ( (C1-C20) alkyl) -C (O) - (C1-C30) alkylene-O-, -NH-C (O) - (C1-C30) alkylene-NH-C (O) - (C1-C30) alkylene-, -NH-C (O) - (C1-C30) alkylene-NH-C (O) -, -N ( (C1-C20) alkyl) -C (O) - (C1-C30) alkylene-N ( (C1-C20) alkyl) -C (O) - (C1-C30) alkylene-, -N ( (C1-C20) alkyl) -C (O) - (C1-C30) alkylene-N ( (C1-C20) alkyl) -C (O) -, - (C1-C30) alkylene-NH-C (O) - (C1-C30) alkylene-NH-C (O) - (C1-C30) alkylene-, - (C1-C30) alkylene-NH-C (O) - (C1-C30) alkylene-NH-C (O) -, - (C1-C30) alkylene -P (O) 2-O-, - (C1-C30) alkylene-O-P (O) 2-O-, - (C3-C50) cycloalkylene-, - (C3-C50) cycloalkylene-O-, -O- (C3-C50) cycloalkylene-, - (C3-C50) cycloalkylene-NH-, -NH- (C3-C50) cycloalkylene-, -C (O) - (C3-C50) cycloalkylene-, - (C3-C50) cycloalkylene-C (O) -, -C (O) -O- (C3-C50) cycloalkylene-, - (C3-C50) cycloalkylene-C (O) -O-, -C (O) -NH- (C3-C50) cycloalkylene-, -C (O) -NH- (C3-C50) cycloalkylene-C (O) -O-, -C (O) -NH- (C3-C50) cycloalkylene-O-, -C (O) -N ( (C1-C20) alkyl) - (C3-C50) cycloalkylene-, -C (O) -N ( (C1-C20) alkyl) - (C3-C50) cycloalkylene-O-, -C (O) -NH- (C3-C50) cycloalkylene-C (O) -NH- (C3-C50) cycloalkylene-, -C (O) -NH- (C3-C50) cycloalkylene-C (O) -NH-, -C (O) -N ( (C1-C20) alkyl) - (C3-C50) cycloalkylene-C (O) -N ( (C1-C20) alkyl) - (C3-C50) cycloalkylene-, -C (O) -N ( (C1-C20) alkyl) - (C3-C50) cycloalkylene-C (O) -N ( (C1-C20) alkyl) -, - (C3-C50) cycloalkylene-C (O) -NH- (C3-C50) cycloalkylene-C (O) -NH- (C3-C50) cycloalkylene-, - (C3-C50) cycloalkylene-C (O) -NH- (C3-C50) cycloalkylene-C (O) -NH-, - (C3-C50) cycloalkylene -P (O) 2-O-, - (C3-C50) cycloalkylene-O-P (O) 2-O-, - (C6-C50) arylene-, - (C6-C50) arylene-O-, -O- (C6-C50) arylene-, - (C6-C50) arylene-NH-, -NH- (C6-C50) arylene-, -C (O) - (C6-C50) arylene-, - (C6-C50) arylene-C (O) -, -C (O) -O- (C6-C50) arylene-, - (C6-C50) arylene-C (O) -O-, -
C (O) -NH- (C6-C50) arylene-, -C (O) -NH- (C6-C50) arylene-C (O) -O-, -C (O) -NH- (C6-C50) arylene-O-, -C (O) -N ( (C1-C20) alkyl) - (C6-C50) arylene-, -C (O) -N ( (C1-C20) alkyl) - (C6-C50) arylene-O-, -C (O) -NH- (C6-C50) arylene-C (O) -NH- (C3-C50) cycloalkylene-, -C (O) -NH- (C6-C50) arylene-C (O) -NH-, -C (O) -N ( (C1-C20) alkyl) - (C6-C50) arylene-C (O) -N ( (C1-C20) alkyl) - (C6-C50) arylene-, -C (O) -N ( (C1-C20) alkyl) - (C6-C50) arylene-C (O) -N ( (C1-C20) alkyl) -, - (C6-C50) arylene-C (O) -NH- (C6-C50) arylene-C (O) -NH- (C6-C50) arylene-, - (C6-C50) arylene-C (O) -NH- (C6-C50) arylene-C (O) -NH-, - (C6-C50) arylene -P (O) 2-O-, - (C6-C50) arylene-O-P (O) 2-O-, - (C5-C50) heteroarylene-, - (C5-C50) heteroarylene-O-, -O- (C5-C50) heteroarylene-, - (C5-C50) heteroarylene-NH-, -NH- (C5-C50) heteroarylene-, -C (O) - (C5-C50) heteroarylene-, - (C5-C50) heteroarylene-C (O) -, -C (O) -O- (C5-C50) heteroarylene-, - (C5-C50) heteroarylene-C (O) -O-, -C (O) -NH- (C5-C50) heteroarylene-, -C (O) -NH- (C5-C50) heteroarylene-C (O) -O-, -C (O) -NH- (C5-C50) heteroarylene-O-, -C (O) -N ( (C1-C20) alkyl) - (C5-C50) heteroarylene-, -C (O) -N ( (C1-C20) alkyl) - (C5-C50) heteroarylene-O-, -C (O) -NH- (C5-C50) heteroarylene-C (O) -NH- (C3-C50) cycloalkylene-, -C (O) -NH- (C5-C50) heteroarylene-C (O) -NH-, -C (O) -N ( (C1-C20) alkyl) - (C5-C50) heteroarylene-C (O) -N ( (C1-C20) alkyl) - (C5-C50) heteroarylene-, -C (O) -N ( (C1-C20) alkyl) - (C5-C50) heteroarylene-C (O) -N ( (C1-C20) alkyl) -, - (C5-C50) heteroarylene-C (O) -NH- (C5-C50) heteroarylene-C (O) -NH- (C6-C50) arylene-, - (C5-C50) heteroarylene-C (O) -NH- (C5-C50) heteroarylene-C (O) -NH-, - (C5-C50) heteroarylene -P (O) 2-O-and - (C5-C50) heteroarylene-O-P (O) 2-O-.
In some embodiments, each of R8′ and R9′ is either absent or is a substituent independently selected from the group consisting of -H, hydroxyl, - (C1-C30) alkyl, - (C3-C50) cycloalkyl, - (C6-C50) aryl, - (C1-C30) alkylene-OH, - (C3-C50) cycloalkylene-OH, - (C6-C50) arylene-OH, - (C1-C30) alkylene-C (O) OH, - (C3-C50) cycloalkylene-C (O) OH, - (C6-C50) arylene-C (O) OH, - (C1-C30) alkylene-NH2, - (C3-C50) cycloalkylene-NH2, - (C6-C50) arylene-NH2, - (C1-C30) alkoxy, - (C3-C50) cycloalkoxy, - (C6-C50) aryloxy, -C (O) - (C1-C30) alkyl, -OC (O) (C1-C30) alkyl, -C (O) -O- (C1-C30) alkyl, -C (O) - (C3-C50) cycloalkyl, -OC (O) - (C3-C50) cycloalkyl, -C (O) -O- (C3-C50) cycloalkyl, -C (O) - (C6-C50) aryloxy, -OC (O) - (C6-C50) aryloxy, -C (O) -O- (C6-C50) aryloxy, -C (O) -NH- (C1-C30) alkyl, -C (O) -NH- (C3-C50) cycloalkyl, -C (O) -NH- (C6-C50) aryl, - (C1-C30) alkylene-phosphoric acid, - (C3-C50) cycloalkylene-phosphoric acid, - (C6-C50) arylene-phosporic acid. In some embodiments, one or more hydroxyl group, carboxyl group, amino group and phosporic acid group contained in each of R8′ and R9′ are optionally protected, e.g. with the protection group RP as defined herein. In an embodiment, R8′ and R9′
can be linked together so that R8′, R9′, the carbon atom linked with R8′ and the Y′ atom linked with R9′ form an unsubstituted or substituted heterocyclic ring. In another embodiment, R9′ is absent when Y′ is oxygen.
In an embodiment, each R10′ is attached to any one of P′, Q′, S′ and T′, and is independently selected from the group consisting of hydroxyl, -C (O) OH, -P (O) 2-OH, -P (O) -OH, -P (O) (S) -OH, -CN, - (C1-C30) alkylene-OH, - (C3-C50) cycloalkylene-OH, - (C6-C50) arylene-OH, - (C5-C50) heteroarylene-OH, - (C1-C30) alkylene-C (O) OH, - (C3-C50) cycloalkylene-C (O) OH, - (C6-C50) arylene-C (O) OH, - (C5-C50) heteroarylene-C (O) OH, -C (O) -NH- (C1-C30) alkylene-OH, -C (O) -NH- (C3-C50) cycloalkylene-OH, -C (O) -NH- (C6-C50) arylene-OH, -C (O) -NH- (C5-C50) heteroarylene-OH, -C (O) -NH- (C1-C30) alkylene-C (O) OH, -C (O) -NH- (C3-C50) cycloalkylene-C (O) OH, -C (O) -NH- (C6-C50) arylene-C (O) OH, -C (O) -NH- (C5-C50) heteroarylene-C (O) OH, - (C1-C30) alkylene-C (O) -NH- (C1-C30) alkylene-C (O) OH, - (C1-C30) alkylene-O-C (O) - (C1-C30) alkylene-C (O) NH2, - (C1-C30) alkylene-O-C (O) - (C1-C30) alkylene-C (O) OH, - (C1-C30) alkylene-O-C (O) - (C1-C30) alkylene-NH2, - (C1-C30) alkylene-O-C (O) - (C1-C30) alkylene-OH, - (C1-C30) alkylene-C (O) -NH- (C3-C50) cycloalkylene-C (O) OH, - (C1-C30) alkylene-C (O) -NH- (C6-C50) arylene-C (O) OH, - (C1-C30) alkylene-C (O) -NH- (C5-C50) heteroarylene-C (O) OH, - (C1-C30) alkylene-P (O) 2-OH, - (C3-C50) cycloalkylene-P (O) 2-OH, - (C6-C50) arylene-P (O) 2-OH, - (C5-C50) heteroarylene-P (O) 2-OH, -C (O) -NH- (C1-C30) alkylene-O-P (-N (C1-C16alkyl) 2) -O- (C1-C30) alkylene-CN, -C (O) -NH- (C1-C30) alkylene-O-P (-N (C1-C16alkyl) 2) -O- (C1-C30) alkylene-OH, -C (O) -NH- (C1-C30) alkylene-O-P (-N (C1-C16alkyl) 2) -O- (C1-C30) alkylene-C (O) OH, -C (O) -NH- (C1-C30) alkylene-O-P (-N (C1-C16alkyl) 2) -O- (C1-C30) alkylene-NH2, - (C1-C30) alkylene-C (O) -NH- (C1-C30) alkylene-O-P (-N (C1-C16alkyl) 2) -O- (C1-C30) alkylene-CN, - (C1-C30) alkylene-C (O) -NH- (C1-C30) alkylene-O-P (-N (C1-C16alkyl) 2) -O- (C1-C30) alkylene-OH, - (C1-C30) alkylene-C (O) -NH- (C1-C30) alkylene-O-P (-N (C1-C16alkyl) 2) -O- (C1-C30) alkylene-C (O) OH, - (C1-C30) alkylene-C (O) -NH- (C1-C30) alkylene-O-P (-N (C1-C16alkyl) 2) -O- (C1-C30) alkylene-NH2, - (C1-C30) alkylene -P (O) -OH, - (C3-C50) cycloalkylene-P (O) -OH, - (C6-C50) arylene-P (O) -OH, - (C5-C50) -heteroarylene-P (O) -OH, - (C1-C30) alkylene-P (O) (S) -OH, - (C3-C50) cycloalkylene-P (O) (S) -OH, - (C6-C50) arylene-P (O) (S) -OH, - (C5-C50) heteroarylene-P (O) (S) -OH, - (C1-C30) alkylene-CN, - (C3-C50) cycloalkylene-CN, - (C6-C50) arylene-CN, - (C5-C50) heteroarylene-CN, wherein one or more hydroxyl group, carboxyl group, amino group, nitrile group and phosporic acid group contained in R10′is optionally
protected with the Rp as defined herein or linked to a support material.
In an embodiment, the - (C1-C30) alkylene-contained in B′, R7′ R8′, R9′ and R10′ may include - (C1-C28) alkylene-, or - (C1-C26) alkylene-, or - (C1-C24) alkylene-, or - (C2-C22) alkylene-, or - (C3-C20) alkylene-, or - (C4-C18) alkylene-, or - (C5-C17) alkylene-, or - (C6-C16) alkylene-, or - (C7-C14) alkylene-, or - (C8-C12) alkylene-. In another embodiment, the - (C3-C50) cycloalkylene-contained in B′, R7′ R8′, R9′ and R10′ may include - (C3-C40) cycloalkylene-, or - (C3-C30) cycloalkylene-, or - (C3-C22) cycloalkylene-, or - (C4-C20) cycloalkylene-, or - (C5-C18) cycloalkylene-, or - (C6-C16) cycloalkylene-, or - (C7-C14) cycloalkylene-, or - (C8-C12) cycloalkylene-. In another embodiment, the - (C6-C50) arylene-contained in B′, R7′ R8′, R9′ and R10′ may include - (C6-C40) arylene-, or - (C6-C30) arylene-, or - (C6-C22) arylene-, or - (C6-C20) arylene-, or - (C6-C18) arylene-, or - (C6-C16) arylene-, or - (C6-C12) arylene-. In another embodiment, the - (C5-C50) heteroarylene-contained in B′, R7′ R8′, R9′ and R10′ may include - (C5-C40) heteroarylene-, or - (C5-C30) heteroarylene-, or - (C5-C22) heteroarylene-, or - (C5-C18) heteroarylene-, or - (C6-C16) heteroarylene-. In another embodiment, in each of the substituent R8′ and R9′, the term “- (C1-C30) alkyl” may include - (C2-C22) alkyl, or - (C3-C18) alkyl, or - (C4-C16) alkyl, or - (C6-C12) alkyl, or - (C8-C10) alkyl; the term “- (C3-C50) cycloalkyl” may include - (C4-C40) cycloalkyl, or - (C5-C30) cycloalkyl, or - (C6-C20) cycloalkyl, or - (C6-C16) cycloalkyl; the term “- (C6-C50) aryl” may include - (C6-C40) aryl, or - (C6-C30) aryl, or - (C6-C25) aryl, or - (C6-C22) aryl, or - (C6-C16) aryl, or - (C6-C12) aryl; and the term “- (C6-C50) aryloxy” may include - (C6-C40) aryloxy, or - (C6-C30) aryloxy, or - (C6-C25) aryloxy, or - (C6-C22) aryloxy, or - (C6-C16) aryloxy, or - (C6-C12) aryloxy.
In another embodiment, each R11′ is attached to any one of P′, Q′, S′ and T′, and is either absent or selected from the group consisting of hydrogen; hydroxyl; halogen atom, such as fluorine, chlorine, bromine or iodine; - (C1-C20) alkyl, such as - (C2-C19) alkyl, or - (C3-C18) alkyl, or - (C4-C16) alkyl, or - (C6-C12) alkyl, or - (C8-C10) alkyl; - (C1-C20) alkoxy, such as - (C2-C19) alkoxy, or - (C3-C18) alkoxy, or - (C4-C16) alkoxy, or - (C6-C12) alkoxy; (C1-C20) alkoxycarbonyl, such as (C1-C16) alkoxycarbonyl, or (C2-C12) alkoxycarbonyl, or (C3-C8) alkoxycarbonyl, or (C4-C6) alkoxycarbonyl; halogenated (C1-C20) alkyl, such as halogenated (C2-C19) alkyl, or halogenated (C3-C18) alkyl, or halogenated (C4-C16) alkyl, or halogenated (C6-C12) alkyl, or halogenated (C8-C10) alkyl; halogenated (C1-C20) alkoxy, such as halogenated (C2-C19) alkoxy, or halogenated (C3-C18) alkoxy, or halogenated (C4-C16) alkoxy, or halogenated
(C6-C12) alkoxy, or halogenated (C8-C10) alkoxy; and halogenated (C1-C20) alkoxycarbonyl, such as halogenated (C2-C19) alkoxycarbonyl, or halogenated (C3-C18) alkoxycarbonyl, or halogenated (C4-C16) alkoxycarbonyl, or halogenated (C6-C12) alkoxycarbonyl, or halogenated (C8-C10) alkoxycarbonyl; wherein “halogenated” can be fluorinated, chlorinated, brominated, iodinated, or combinations thereof.
In another embodiment, R9′ is absent when Y′ is oxygen. In an exemplary embodiment, Y′ is carbon, and both of R8′ and R9′ are present. In another embodiment, Y′ is nitrogen, R8′ is present and R9′ is absent. In another embodiment, Y′ is nitrogen with positive charge (i.e., quaternary ammonium) , both R8′ and R9′ are present. In another embodiment, Y′ is sulfur or oxygen, and R9′ is absent. In another embodiment, Y′ is sulfur with positive charge (i.e., sulfonium) , and R9′ is present.
In an embodiment, M′ is an integer of 1, 2 or 3, N′ is an integer of 1, 2 or 3, and M′+N′=4. In some exemplary embodiments, M′=1 and N′=3; or M′=2 and N′=2; or M′=3 and N′=1.
In an embodiment, each R10′ is attached to any one of P′, Q′, S′ and T′, while R11′ is attached to the rest of P′, Q′, S′ and T′. In an embodiment, R10′ is attached to S′ and three R11′ are separately attached to each of P′, Q′ and T′. In another embodiment, R10′ is attached to Q′ and three R11′ are separately attached to each of P′, S′ and T′. In another embodiment, R10′ is attached to P′ and three R11′ are separately attached to each of Q′, S′ and T′. In another embodiment, R10′ is attached to T′ and three R11′ are separately attached to each of P′, Q′ and S′. In another embodiment, two R10′ are separately attached to Q′ and S′, and two R11′ are separately attached to each of P′ and T′.
In a specific embodiment, one or more hydroxyl group, carboxyl group, amino group and phosphoric acid group contained in each of R8′, R9′ and R10′ are optionally protected with a terminal protection group RP selected from the group consisting of (C1-C22) alkyl, (C1-C22) alkoxy, (C1-C22) alkylcarbonyl, (C1-C22) alkoxycarbonyl, (C6-C22) aryl, (C6-C22) aryloxy, (C6-C22) arylcarbonyl, (C6-C22) aryloxycarbonyl, tri ( (C1-C22) alkyl) silyl and tri ( (C1-C22) alkoxy) silyl, wherein the (C1-C22) alkyl contained in the protection group Rp can be an alkyl comprising from 1 to 22 carbon atoms, such as from 2 to 20 carbon atoms, or from 3 to 18 carbon atoms, or from 4 to 16 carbon atoms, or from 6 to 12 carbon atoms; the (C1-
C22) alkoxy contained in Rp can be an alkoxy comprising from 1 to 22 carbon atoms, such as from 2 to 20 carbon atoms, or from 3 to 18 carbon atoms, or from 4 to 16 carbon atoms, or from 6 to 12 carbon atoms; the (C6-C22) aryl contained in Rp can be an aryl comprising from 6 to 22 carbon atoms, such as from 6 to 20 carbon atoms, or from 6 to 18 carbon atoms, or from 6 to 16 carbon atoms, or from 6 to 12 carbon atoms, or from 8-10 atoms; and the (C6-C22) aryloxy contained in Rp can be an aryloxy comprising from 6 to 22 carbon atoms, such as from 6 to 20 carbon atoms, or from 6 to 18 carbon atoms, or from 6 to 16 carbon atoms, or from 6 to 12 carbon atoms, or from 8-10 atoms.
In an exemplary embodiment, the support material attached to R10′can be selected from the group consisting of silica, silica gel, glass, ceramic, polymer, cellulose, and combinations thereof. In some embodiments, the solid material is in the form of a bead. The bead may be made out of any material including, without limitation, magnetic bead, paramagnetic bead, silica bead, an agarose bead, etc.
In another embodiment, the compound of the present disclosure may have a structure represented by any of Formula BIV to Formula BXIV,
wherein A1′, A2′, A3′, B′, C′, F′, G′, H′, I′, R7′, R8′, R10′, R11′, P′, Q′, S′, T′, m′, n′, M′ and N′ are as defined herein.
In an embodiment, the RING I can be formed by linking A1′ and A2′ together and thus may consist of the nitrogen atom to which A1′ is attached, the carbon atom to which A2′ is attached, at least part of A1′ and at least part of A2′. The RING I can be a 4, 5, 6, 7, 8 or 9 member ring, in particular a ring fused to the core structure. For example, the RING I may consist of a nitrogen atom, a carbon atom from the core structure and 2, 3, 4, 5, 6 or 7 additional ring atoms derived from A1′ and A2′, wherein each of the additional ring atoms may be selected from the group consisting of carbon, nitrogen, oxygen, sulfur, and combinations thereof. In another exemplary embodiment, all of the additional ring atoms may be carbon atoms.
A4′ can be a substituent attached to any atom of RING I, and each of A4′, A5′ and A6′ is independently selected from the group consisting of -R1′, -O-R1′, -S-R1′, -C (O) -R1′, -C (O) O-R1′, -O-C (O) -R1′, -C (O) NH-R1′, -C (O) NR2′-R1′, -NH-C (O) -R1′, -NR2′-C (O) -R1′, -O-P (O) 2-O-R1′, -OP (O) (S) -O-R1′, -O-P (O) -O-R1′, -NH-R1′, -NR2′-R1′, - (CH2) r′-NH-R1′, - (CH2) r′-NR2′-R1′, -C (O) - (CH2) r′-R1′, -C (O) - (CH2) r′-NH-R1′, -C (O) - (CH2) r′-NR2′-R1′, -C (O) - (CH2) r′-C (O) -R1′, -C (O) - (CH2) r′-C (O) O-R1′, -C (O) - (CH2) r′-NH-C (O) -R1′, -C (O) - (CH2) r′-NR2′-C (O) -R1′, - (CH2) r′-C (O) -R1′; - (CH2) r′-C (O) O-R1′; - (CH2) r′-O-C (O) -R1′, - (CH2) r′-R1′, - (CH2) r′-NH-C (O) -R1′, - (CH2) r′-NH-C (O) - (CH2) s′-R1′, - (CH2) r′-NR2′-C (O) - (CH2) s′-R1′, -CH (- (CH2) r′-NH-C (O) - (CH2) s′-R1′) (-NH-C (O) - (CH2) q′-R3′) , -N (- (CH2) r′-NH-C (O) - (CH2) s′-R1′) (-NH-C (O) - (CH2) q′-R3′) , -CH (- (CH2) r′-NH-C (O) - (CH2) s′-R1′) (- (CH2) p′-NH-C (O) - (CH2) q′-R3′) , -CR4′ (- (CH2) r′-NH-C (O) - (CH2) s′-R1′) (-NH-C (O) - (CH2) q′-R3′) , -CR4′ (- (CH2) r′-NH-C (O) - (CH2) s′-R1′) (- (CH2) p′-NH-C (O) - (CH2) q′-R3′) , -CH (- (CH2) r′-NR5′-C (O) - (CH2) s′-R1′) (-NR6′-C (O) - (CH2) q′-R3′) , -CH (- (CH2) r′-NR5′-C (O) - (CH2) s′-R1′) (- (CH2) p′-NR6′-C (O) - (CH2) q′-R3′) , -CR4′ ( (CH2) r′-NR5′-C (O) - (CH2) s′-
R1′) ( (CH2) p′-NR6′-C (O) - (CH2) q′-R3′) , -CR4′ ( (CH2) r′-NR5′-C (O) - (CH2) s′-R1′) ( (CH2) p′-NR6′-C (O) - (CH2) q′-R3′) ; wherein each of R1′, R2′, R3′, R4′, R5′, R6′, r′, s′, p′ and q′ are as defined herein.
In an embodiment, the RING II can be formed by linking R8′ and R9′ together and thus may consist of the nitrogen atom to which R9′ is attached, the carbon atom to which R8′ is attached, at least part of R8′ and at least part of R9′. The RING II can be a 4, 5, 6, 7, 8 or 9 member ring, in particular a ring fused to the core structure of the substituent B′. For example, the RING II may consist of a nitrogen atom, a carbon atom from the core structure of the substituent B′ and 2, 3, 4, 5, 6 or 7 additional ring atoms derived from R8′ and R9′, wherein each of the additional ring atoms may be selected from the group consisting of carbon, nitrogen, oxygen, sulfur, and combinations thereof. In another exemplary embodiment, all of the additional ring atoms may be carbon atoms.
In an embodiment, R12′ is attached to any atom of RING II and is selected from the group consisting of -H, hydroxyl, - (C1-C30) alkyl, - (C3-C50) cycloalkyl, - (C6-C50) aryl, - (C1-C30) alkylene-OH, - (C3-C50) cycloalkylene-OH, - (C6-C50) arylene-OH, - (C1-C30) alkylene-C (O) OH, - (C3-C50) cycloalkylene-C (O) OH, - (C6-C50) arylene-C (O) OH, - (C1-C30) alkylene-NH2, - (C3-C50) cycloalkylene-NH2, - (C6-C50) arylene-NH2, - (C1-C30) alkoxy, - (C3-C50) cycloalkoxy, - (C6-C50) aryloxy, -C (O) - (C1-C30) alkyl, -OC (O) (C1-C30) alkyl, -C (O) -O- (C1-C30) alkyl, -C (O) - (C3-C50) cycloalkyl, -OC (O) - (C3-C50) cycloalkyl, -C (O) -O- (C3-C50) cycloalkyl, -C (O) - (C6-C50) aryloxy, -OC (O) - (C6-C50) aryloxy, -C (O) -O- (C6-C50) aryloxy, -C (O) -NH- (C1-C30) alkyl, -C (O) -NH- (C3-C50) cycloalkyl, -C (O) -NH- (C6-C50) aryl, - (C1-C30) alkylene-phosporic acid, - (C3-C50) cycloalkylene-phosporic acid, - (C6-C50) arylene-phosporic acid; wherein each - (C1-C30) alkyl contained in R12′ may comprise - (C2-C22) alkyl, or - (C3-C18) alkyl, or - (C4-C16) alkyl, or - (C6-C12) alkyl, or - (C8-C10) alkyl; wherein each - (C3-C50) cycloalkyl contained in R12′ may comprise - (C4-C40) cycloalkyl, or - (C5-C30) cycloalkyl, or - (C6-C20) cycloalkyl, or - (C6-C16) cycloalkyl; each - (C6-C50) aryl contained in R12′ may comprise - (C6-C40) aryl, or - (C6-C30) aryl, or - (C6-C25) aryl, or - (C6-C22) aryl, or - (C6-C16) aryl, or - (C6-C12) aryl; each - (C1-C30) alkoxy contained in R12′ may comprise - (C2-C22) alkoxy, or - (C3-C20) alkoxy, or - (C4-C16) alkoxy, or - (C6-C12) alkoxy; each - (C3-C50) cycloalkoxy contained in R12′ may comprise -(C4-C40) cycloalkoxy, or - (C5-C30) cycloalkoxy, or - (C6-C20) cycloalkoxy, or - (C6-
C16) cycloalkoxy; each - (C6-C50) aryloxy contained in R12′ may comprise - (C6-C40) aryloxy, or - (C6-C30) aryloxy, or - (C6-C25) aryloxy, or - (C6-C22) aryloxy, or - (C6-C16) aryloxy, or - (C6-C12) aryloxy; each - (C1-C30) alkylene-contained in R12′ may comprise - (C1-C28) alkylene-, or - (C1-C26) alkylene-, or - (C1-C24) alkylene-, or - (C2-C22) alkylene-, or - (C3-C20) alkylene-, or - (C4-C18) alkylene-, or - (C5-C17) alkylene-, or - (C6-C16) alkylene-, or - (C7-C14) alkylene-, or - (C8-C12) alkylene-; the - (C3-C50) cycloalkylene- contained in R12′ may include - (C3-C40) cycloalkylene-, or - (C3-C30) cycloalkylene-, or - (C3-C22) cycloalkylene-, or - (C4-C20) cycloalkylene-, or - (C5-C18) cycloalkylene-, or - (C6-C16) cycloalkylene-, or - (C7-C14) cycloalkylene-, or - (C8-C12) cycloalkylene-; the - (C6-C50) arylene-contained in R12′ may include - (C6-C40) arylene-, or - (C6-C30) arylene-, or - (C6-C22) arylene-, or - (C6-C20) arylene-, or - (C6-C18) arylene-, or - (C6-C16) arylene-, or - (C6-C12) arylene-.
In an embodiment, one or more hydroxyl group, carboxyl group, amino group and phosporic acid group contained in R12′ are optionally protected, e.g. with a terminal protection group RP selected from the group consisting of (C1-C22) alkyl, (C1-C22) alkoxy, (C1-C22) alkylcarbonyl, (C1-C22) alkoxycarbonyl, (C6-C22) aryl, (C6-C22) aryloxy, (C6-C22) arylcarbonyl, (C6-C22) aryloxycarbonyl, tri ( (C1-C22) alkyl) silyl and tri ( (C1-C22) alkoxy) silyl, wherein the (C1-C22) alkyl contained in the protection group Rp can be an alkyl comprising from 1 to 22 carbon atoms, such as from 2 to 20 carbon atoms, or from 3 to 18 carbon atoms, or from 4 to 16 carbon atoms, or from 6 to 12 carbon atoms; the (C1-C22) alkoxy contained in Rp can be an alkoxy comprising from 1 to 22 carbon atoms, such as from 2 to 20 carbon atoms, or from 3 to 18 carbon atoms, or from 4 to 16 carbon atoms, or from 6 to 12 carbon atoms; the (C6-C22) aryl contained in Rp can be an aryl comprising from 6 to 22 carbon atoms, such as from 6 to 20 carbon atoms, or from 6 to 18 carbon atoms, or from 6 to 16 carbon atoms, or from 6 to 12 carbon atoms, or from 8-10 atoms; and the (C6-C22) aryloxy contained in Rp can be an aryloxy comprising from 6 to 22 carbon atoms, such as from 6 to 20 carbon atoms, or from 6 to 18 carbon atoms, or from 6 to 16 carbon atoms, or from 6 to 12 carbon atoms, or from 8-10 atoms. In a specific embodiment, the protection group RP can be selected from the group consisting of benzyloxycarbonyl (Cbz) , tert-butyldimethylsilyl (TBS) , 4, 4′-dimethoxytrityl (DMTr) , t-butyloxy carbonyl (Boc) , benzyl (Bn) and benzyloxy (BnO) .
In an embodiment, the compound of Formula BI or Formula BII may have one or more chiral centers, and each of the chiral centers can be independently R chiral, S chiral,
mesomeric or racemic form. For example, when one or more of A1′, A2′ and A3′ is selected from -CH (- (CH2) r′-NH-C (O) - (CH2) s′-R1′) (-NH-C (O) - (CH2) q′-R3′) , -CH (- (CH2) r′-C (O) -NH- (CH2) s′-R1′) (-C (O) -NH- (CH2) q′-R3′) , -N (- (CH2) r′-NH-C (O) - (CH2) s′-R1′) (-NH-C (O) - (CH2) q′-R3′) , -CH (- (CH2) r′-NH-C (O) - (CH2) s′-R1′) (- (CH2) p′-NH-C (O) - (CH2) q′-R3′) , -CR4′ (- (CH2) r′-NH-C (O) - (CH2) s′-R1′) (-NH-C (O) - (CH2) q′-R3′) , -CR4′ (- (CH2) r′-NH-C (O) - (CH2) s′-R1′) (- (CH2) p′-NH-C (O) - (CH2) q′-R3′) , -CH (- (CH2) r′-NR5′-C (O) - (CH2) s′-R1′) (-NR6′-C (O) - (CH2) q′-R3′) , -CH (- (CH2) r′-NR5′-C (O) - (CH2) s′-R1′) (- (CH2) p′-NR6′-C (O) - (CH2) q′-R3′) , -CR4′ ( (CH2) r′-NR5′-C (O) - (CH2) s′-R1′) ( (CH2) p′-NR6′-C (O) - (CH2) q′-R3′) , -CR4′ ( (CH2) r′-NR5′-C (O) - (CH2) s′-R1′) ( (CH2) p′-NR6′-C (O) - (CH2) q′-R3′) , the central carbon atoms contained therein can be chiral centers.
In some embodiments, the compound of the present disclosure has a structure represented by any one of Formula BXV to Formula BXX:
wherein each of A1′, A2′, B′, R7′, R8′ and R10′ are as defined herein.
In another embodiment, when any one or more of the above indicated substituents comprise at least one double bond, each of the double bond and the atoms or moieties attached to the same may be in E form or Z form.
In an exemplary embodiment, the compound of the present disclosure has a structure represented by any of the following molecular formulae:
wherein represents a support material as defined above.
Another embodiment of the present disclosure provides an oligonucleotide delivery agent comprising a DEC compound disclosed by the present application and at least one oligonucleotide. It can be fully understood that when the DEC is directly linked with the oligonucleotide or indirectly linked with the oligonucleotide via at least one linking moiety, one or more terminal atoms (such as hydrogen, halogen, nitrogen, oxygen, sulfur, phosphorus, etc. ) or terminal groups (such as hydroxyl group, amino group, ester group, ether group, acyl group, etc. ) of the DEC may be detached so as to provide an active site linkable to the linking moiety or the oligonucleotide.
In an embodiment, the linking moieties, when present, can be selected from the group consisting of -O-, -S-, -C (O) -, -NH-, -N ( (C1-C12) alkyl) -, -N ( (C1-C12) alkyl) -C (O) -O-, -O-C (O) -, -C (O) -O-, -O-C (O) -O-, -C (O) -NH-, -OP (O) 2O-, -P (O) (O-) O-, -OP (O) O-, -OP (O) (S) O-, -O-S (O) 2-O-, -S (O) 2-O-, -S (O) -O-, - (C1-C22) alkylene-, - (C1-C22) alkylene-NH-, -NH- (C1-C22) alkylene-, - (C1-C22) alkylene-NH-C (O) -, - (C1-C22) alkylene-C (O) -, - (C1-C22) alkylene-C (O) -
O-, -C (O) - (C1-C22) alkylene-, -NH-C (O) - (C1-C22) alkylene-, -C (O) -NH- (C1-C22) alkylene-, -C (O) - (C1-C22) alkylene-NH-, -NH- (C1-C22) alkylene-C (O) -, -C (O) - (C1-C22) alkylene-C (O) -, -NH-(C1-C22) alkylene-NH-, -C (O) - (C1-C22) alkylene-C (O) O-, -O-C (O) - (C1-C22) alkylene-C (O) -O-, -C (O) -O- (C1-C22) alkylene-O-C (O) -, -C (O) - (C1-C22) alkylene-NH-C (O) -, -NH-C (O) - (C1-C22) alkylene-C (O) -, -NH-C (O) - (C1-C22) alkylene-C (O) -NH-, -C (O) -NH- (C1-C22) alkylene-NH-C (O) -, - (C1-C22) alkylene-OP (O) 2O-, - (C1-C22) alkylene-OP (O) (O-) O-, - (C1-C22) alkylene-OP(O) (O-) O- (C1-C22) alkylene-, - (C1-C22) alkylene-OP (O) O-, - (C1-C22) alkylene-OP (O) (S) O-, - (C1-C22) alkylene-O-S (O) 2-O-, - (C1-C22) alkylene-S (O) 2-O-, - (C1-C22) alkylene-S (O) -O-, -O-P (O) 2-O- (C1-C22) alkylene-OP (O) 2O-, -O-P (O) -O- (C1-C22) alkylene-OP (O) O-, -OP (O) (S) O- (C1-C22) alkylene-OP (O) (S) O-, -O-S (O) 2-O- (C1-C22) alkylene-O-S (O) 2-O-, -S (O) 2-O- (C1-C22) alkylene-S (O) 2-O-and -O-S (O) - (C1-C22) alkylene-S (O) -O-; wherein the - (C1-C22) alkylene-contained in the linking moiety can be an alkylene group comprising from 1 to 22 carbon atoms, such as from 2 to 20 carbon atoms, or from 3 to 18 carbon atoms, or from 4 to 16 carbon atoms, or from 5 to 12 carbon atoms, or from 6 to 10 carbon atoms. In one embodiment, the delivery enhancing compound is directly linked with the oligonucleotide when the linking moiety is a direct bond.
Another embodiment of the present disclosure provides a delivery enhancing compound (DEC) conjugated oligonucleotide comprising a structure represented by Formula AA, which can more efficiently deliver the oligonucleotide both in vitro and in vivo.
The Pdelivery enhancing compound moiety” shown in Formula AA is derived from any one of the above stated DEC compounds of the present disclosure. It can be understood that one or more terminal atoms (such as hydrogen, halogen, nitrogen, oxygen, sulfur, phosphorus, etc. ) or terminal groups (such as hydroxyl group, amino group, ester group, ether group, acyl group, etc. ) of the inventive compound have be detached so as to provide an active site linkable to the targeting moiety or the oligonucleotide, hence the “delivery enhancing compound” of Formula BA can be considered as a moiety obtained by subtracting said one or more atoms or terminal groups from the inventive compounds.
In an embodiment of the present disclosure, the delivery enhancing compound (DEC) conjugated oligonucleotide has a structure represented by Formula BB,
wherein each of A1′, A2′, A3′, X′, F′, G′H′, I′, C′, m′ and n′ is as defined above. In an embodiment, at least one of A1′, A2′ and A3′ comprises one or more targeting moieties, and the DEC is attached to at least one oligonucleotide via the B″ group. As stated above, the B″ group can be derived by detaching one or more terminal atoms (such as hydrogen, halogen, nitrogen, oxygen, sulfur, phosphorus, etc. ) or one or more terminal groups (such as hydroxyl group, amino group, ester group, ether group, acyl group, etc. ) from the B″ group contained in Formula BII.
In another embodiment of the present disclosure, the delivery enhancing compound (DEC) conjugated oligonucleotide has a structure represented by Formula BC,
wherein each of A1′, A2′, A3′, R7′, R9′, R10′, R11′, X′, Y′, F′, G′, H′, I′, C′, R′, Q′, S′, T′, m′, n′, M′ and N′ is as defined above. In an embodiment, at least one of A1′, A2′ and A3′ comprises one or more targeting moieties, and the DEC is attached to at least one oligonucleotide via the R8″ group. As stated above, the R8″ group can be derived by detaching one or more terminal atoms (such as hydrogen, halogen, nitrogen, oxygen, sulfur, phosphorus, etc. ) or one or more terminal groups (such as hydroxyl group, amino group, ester group, ether
group, acyl group, etc. ) from the R8′ group contained in Formula BIII.
In some embodiments of the present disclosure, the delivery enhancing compound (DEC) conjugated oligonucleotide has a structure represented by any one of Formulae E1 to E15,
wherein each of A1′, A2′, R7′ and R10′ is as defined above. In an embodiment, at least one of A1′ and A2′ comprises one or more targeting moieties, or A2′ comprises one or more targeting moieties; and the DEC is attached to at least one oligonucleotide via the B″ group or the R8″ group. As stated above, the R8″ group can be derived by detaching one or more terminal atoms (such as hydrogen, halogen, nitrogen, oxygen, sulfur, phosphorus, etc. ) or one or more terminal groups (such as hydroxyl group, amino group, ester group, ether group, acyl group, etc. ) from the R8 group, and the B″ group can be derived by detaching one or more terminal atoms (such as hydrogen, halogen, nitrogen, oxygen, sulfur, phosphorus, etc. ) or one or more terminal groups (such as hydroxyl group, amino group, ester group, ether group, acyl group, etc. ) from the B′ group.
In an embodiment, the targeting moiety is contained in the molecular structure of the DEC compound, such as being as part of the A1′, A2′ or A3′, and preferably being the R1′ or R3′ group. In another embodiment, the targeting moiety is independent of the DEC compound and
is additionally added after or during the synthesis of the DEC compound.
In an embodiment, the targeting moiety and the delivery enhancing compound moiety can be linked with each other by a first linking moiety. In an embodiment, the delivery enhancing compound moiety and the oligonucleotide can be linked by a second linking moiety. Each of the first and second linking moieties can be selected from the group consisting of direct bond, -O-, -S-, -C (O) -, -NH-, -N ( (C1-C12) alkyl) -, -N ( (C1-C12) alkyl) -C (O) -O-, -O-C (O) -, -C (O) -O-, -O-C (O) -O-, -C (O) -NH-, -OP (O) 2O-, -OP (O) O-, -OP (O) (S) O-, -O-S (O) 2-O-, -S (O) 2-O-, -S (O) -O-, - (C1-C22) alkylene-, - (C1-C22) alkylene-NH-, -NH- (C1-C22) alkylene-, - (C1-C22) alkylene-NH-C (O) -, - (C1-C22) alkylene-C (O) -, - (C1-C22) alkylene-C (O) -O-, -C (O) - (C1-C22) alkylene-, -NH-C (O) - (C1-C22) alkylene-, -C (O) -NH- (C1-C22) alkylene-, -C (O) - (C1-C22) alkylene-NH-, -NH- (C1-C22) alkylene-C (O) -, -C (O) - (C1-C22) alkylene-C (O) -, -NH- (C1-C22) alkylene-NH-, -C (O) - (C1-C22) alkylene-C (O) O-, -O-C (O) - (C1-C22) alkylene-C (O) -O-, -C (O) -O- (C1-C22) alkylene-O-C (O) -, -C (O) - (C1-C22) alkylene-NH-C (O) -, -NH-C (O) - (C1-C22) alkylene-C (O) -, -NH-C (O) - (C1-C22) alkylene-C (O) -NH-, -NH- (C1-C22) alkylene-OP (O) 2O-, -NH- (C1-C22) alkylene-CH ( (C1-C22) alkylene-OH) -OP (O) 2O-, -NH- (C1-C22) alkylene-CH ( (C1-C22) alkylene-OH) - (C1-C22) alkylene-OP (O) 2O-, -C (O) -NH- (C1-C22) alkylene-NH-C (O) -, - (C1-C22) alkylene-OP (O) 2O-, - (C1-C22) alkylene-OP (O) O-, - (C1-C22) alkylene-OP (O) (S) O-, - (C1-C22) alkylene-O-S (O) 2-O-, - (C1-C22) alkylene-S (O) 2-O-, - (C1-C22) alkylene-S (O) -O-, -O-P (O) 2-O- (C1-C22) alkylene-OP (O) 2O-, -O-P (O) -O- (C1-C22) alkylene-OP (O) O-, -OP (O) (S) O- (C1-C22) alkylene-OP (O) (S) O-, -O-S (O) 2-O- (C1-C22) alkylene-O-S (O) 2-O-, -S (O) 2-O- (C1-C22) alkylene-S (O) 2-O-and -O-S (O) - (C1-C22) alkylene-S (O) -O-; wherein the - (C1-C22) alkylene-contained in the linking moiety can be - (C1-C20) alkylene-, or - (C1-C18) alkylene-, or - (C1-C16) alkylene-, or - (C2-C12) alkylene-, or - (C3-C10) alkylene-, or - (C4-C8) alkylene-, or - (C5-C6) alkylene-. In one embodiment, in delivery enhancing compound (DEC) conjugated oligonucleotide of Formula A, the delivery enhancing compound moiety is directly linked with the oligonucleotide when the linking moiety is a direct bond. In one embodiment, in the delivery enhancing compound (DEC) conjugated oligonucleotide of Formula BA, the targeting moiety is directly linked with the delivery enhancing compound moiety when the linking moiety is a direct bond.
The oligonucleotide may include those disclosed above, and particularly can be selected from the group consisting of antisense oligonucleotide (ASO) , antisense RNA, short
interfering RNA (siRNA) , microRNA (miRNA) , saRNA, dsRNA, scRNA, sgRNA or any other oligonucleotide targeting at least one nucleic acid sequence.
All nucleotides of the oligonucleotides described herein may be natural, i.e., non-chemically modified, nucleotides or at least one nucleotide may be a chemically modified nucleotide. Non-limiting examples of the chemical modification include one or more of a combination of the following:
1) modification of a phosphodiester bond of nucleotides in the oligonucleotide sequence;
2) modification of 2′-OH of the ribose in the nucleotide;
3) modification of a base in the nucleotide;
4) at least one nucleotide in the oligonucleotide sequence being a locked nucleic acid, and
5) at least one nucleotide in the oligonucleotide sequence being a deoxyribonucleotide (DNA) .
Chemical modifications of nucleotides or oligonucleotides in the present disclosure are well known to those skilled in the art, and modifications of the phosphodiester bond refer to modifications of oxygen in the phosphodiester bond, including phosphorothioate modifications and boronated phosphate modifications. The modifications disclosed herein stabilize an oligonucleotide structure, maintaining high specificity and high affinity for base pairing. The modifications disclosed herein also stabilize a nucleic acid structure and maintain its delivering accessory properties including bioavailability, biodistribution, and/or cellular uptake of the oligonucleotide agent in various tissues prefrontal cortex, cerebellum, spinal cord (e.g., cervical, thoracic, lumber) , muscle, liver, and kidney.
In some embodiments, the chemical modification is to substitute the phosphodiester bond with phosphorothioate (PS) bond on the backbone of the oligonucleotide disclosed herein. In some embodiments, the oligonucleotide disclosed herein comprises at least one PS backbone modification in one oligonucleotide strand. In some embodiments, the oligonucleotide comprises at least 3, 4, 5, 6, 7, 8, 9, 10, 12, 14, 16, 18, 20, 24, 28, 32, or 40 PS backbone modifications in one oligonucleotide strand.
In some embodiments, the nucleotides or oligonucleotides of the present application includes at least one chemically modified nucleotide which is modified at 2′-OH in pentose of a nucleotide, i.e., the introduction of certain substituents at the hydroxyl position of the ribose, such as 2′-fluoro modification, 2′-oxymethyl modification, 2′-oxyethylidene methoxy
modification, 2, 4′-dinitrophenol modification, locked nucleic acid (LNA) , 2′-amino modification or 2′-deoxy modification, e.g., a 2’-deoxy-2’-fluoro modified nucleotide, a 2’-deoxy-modified nucleotide.
In some embodiments, the nucleotides or oligonucleotides of the present application includes at least one chemically modified nucleotide which is modified at the base of the nucleotide, e.g., 5 ′-bromouracil modification, 5’-iodouracil modification, N-methyluracil modification, or 2, 6-diaminopurine modification.
In some embodiments, the chemical modification of the nucleotides or oligonucleotides in the present application is an addition of a (E) -vinylphosphonate moiety at the 5’ end of the sense or antisense sequence. In some embodiments, the chemical modification of the at least one chemically modified nucleotide is an addition of a 5′-methyl cytosine moiety at the 5’ end of the sense or antisense sequence. The oligonucleotides used in the present disclosure may be commercially available from various vendors, or synthesized in a lab scale or industrial scale. In an exemplary embodiment, the oligonucleotides can be synthesized by using a commercialized synthesizer or a particularly customized synthesizer, such as a K&ADNA synthesizer purchased from K&ALaborgeraete GbR, Schaafheim, Germany, by using ordinary synthesis procedures, e.g. a solid phase synthesis technique comprising the steps of sequentially adding batches of raw materials (e.g. phosphoramidite monomers including various linkers and conjugates) onto a solid support known in the art and subjecting each base addition to a preparation cycle consisted of four chemical reactions of detritylation, coupling, oxidation/thiolation and capping, so as to produce the oligonucleotides with desired full-length.
The oligonucleotide delivery agent may comprise one, two, three, four, five, six or even more oligonucleotides separately linked with one, two, three, four, five, six or even more of the delivery enhancing compounds via one, two, three, four, five, six or even more linking moieties. For example, the oligonucleotide delivery agent may have a structure represented by any of the following formulae AAI to AAXXIV:
wherein L represents the linking moiety, represents the oligonucleotide delivery enhancing compound, the symbol epresents a double strand oligonucleotide, either symmetric or asymmetric independently on each of the ends; presents a single strand oligonucleotide, and each of a, b and c is independently an integer from 1 to 50, such as an integer from 2 to 45, or an integer from 3 to 40, or an integer from 4 to 35, or an integer from 5 to 30, or an integer from 10 to 20. Each of the linking moiety, and thus the delivery enhancing compound, may be linked at 3’ end, 5’ end or any internal position, such as the nth nucleotide, of the double or single strand oligonucleotide.
In one embodiment, one or more of the substituents A1, A2, A3, B, C, A1′, A2′, A3′, B′ and C′ of the delivery enhancing compound can be linked with the linking moiety or with the oligonucleotide (when the linking moiety is a direct bond) . For example, either one of A1, A2, A3, B, A1′, A2′, A3′ R8′, R12′and B′, preferably A2 A2′, B, B′, R8′ or R12′ is linked with the linking moiety or the oligonucleotide. Without being limited to any theory, one or more terminal groups of the above said substituents (such as DMTrO-C1-C22 alkylene-or - (C1-C22) alkylene-O-P (-N (C1-C22 alkyl) 2) -O- (C1-C22) alkylene-CN) , may be cleavaged or hydrolyzed to decap an active site (such as a - (C1-C22) alkylene-OH group) which is then linked with the oligonucleotide via a linking moiety (such as -P (O) (O-) O-) . For example, exemplary structures of the oligonucleotide delivery agents include O1 to O25 as illustrated
below:
Compound O1 (RD-13110, derived from Compound A1)
Compound O2 (RD-13115, RD-13118, RD-13520, RD-15137, RD-15138, RD-16424, RD-14644, RD-14672, RD-16447, RD-16381, derived from Compound A5)
Compound O3 (derived from Compound A7)
Compound O4 (derived from Compound A8)
Compound O5 (derived from Compound A9)
Compound O6 (derived from Compound A10)
Compound O7 (derived from Compound A11)
Compound O8 (derived from Compound A12)
Compound O9 (derived from Compound A13)
Compound O10 (derived from Compound A14)
Compound O11 (derived from Compound A15)
Compound O12 (derived from Compound A16)
Compound O13 (derived from Compound A6)
wherein J represents O or S.
It can be seen that in all the structures O1 to O25, the delivery enhancing compounds are linked with double-stranded RNA (dsRNA) duplexes (including but not limited to siRNA or saRNA) and/or single-stranded antisense oligonucleotides (ASOs) at the 3’-terminus or 5’-terminus their passenger (P) strand via a linking moiety, such as -OP (O) 2O- (-P (O) (O-) -O-) , wherein P is passenger strand and G is guide strand; and electron rearrangement may also occur.
In some embodiments, at least one hydrogen atom (i.e. H) contained in the delivery enhancing compound, the delivery enhancing compound moiety, the linking moiety and/or the oligonucleotide is substituted with deuterium atom (i.e. D) . In an exemplary embodiment, at least one of the delivery enhancing compound, the delivery enhancing compound moiety, the linking moiety and the oligonucleotide comprises one to twenty, such as one to fifteen, or one to twelve, or one to ten, or one to eight, or one to six, or one to three, or one to two deuterium atoms. In an exemplary embodiment, 1%to 100%, or 2%to 90%, or 5%to 80%, or 10%to 70%, or 20%to 60%, or 30%to 50%, or 40%to 45%by mole of the hydrogen atom contained in the the delivery enhancing compound, the delivery enhancing compound moiety, the linking moiety and/or the oligonucleotide is substituted with deuterium atom. In another exemplary embodiment, the deuterium substitution rate can be within a numerical range obtained by combining any two of the above said end point values.
Another embodiment of the present disclosure provides a pharmaceutical composition comprising the oligonucleotide delivery agent. In another embodiment, the pharmaceutical composition may comprise one or more additional ingredients, such as pharmaceutically acceptable carrier, excipient, solvent, diluent, stabilizer, dispersant, buffer, compatibilizer, preservative agent and combinations thereof.
Another embodiment of the present disclosure provides a method of modulating the expression of a target gene in vitro or in vivo, comprising the step of administrating the pharmaceutical composition to a subject, or contacting the pharmaceutical composition with cells of the subject. For example, it is estimated that the oligonucleotide delivery agent and pharmaceutical composition can be applied in various organs, tissues and cells, such as liver, lung, kidney, intestine, pancreas, cholecyst, heart, lymp nodes, spleen, stomach, bladder, muscle, bone, central nervous system (CNS) , and modulate the expression of one or more target genes in the cell thereof.
Particular embodiments
In a specific embodiment, the present disclosure provides an oligonucleotide delivery enhancing compound comprising a nitrogen-containing five membered heterocyclic ring moiety and at least one substituent directly or indirectly attachable to an oligonucleotide.
In another specific embodiment, the oligonucleotide delivery enhancing compound has a structure represented by Formula AI or Formula AII
wherein eachindependently represents a covalent single or double bond; X, on each occurrence, is an atom selected from the group consisting of carbon, nitrogen, oxygen and sulfur; each of F, G, H and I is independently selected from the group consisting of carbon, nitrogen, oxygen and sulfur;
wherein m is an integer of 1, 2 or 3, n is an integer of 1, 2 or 3, and m+n=4;
wherein C, on each occurrence, is either absent or selected from the group consisting of hydrogen, halogen atom, hydroxyl, (C1-C20) alkyl, (C1-C20) alkoxy, halogenated (C1-C20) alkyl and halogenated (C1-C20) alkoxy;
wherein B, on each occurrence, is independently selected from the group consisting of hydroxyl, -C (O) OH, -P (O) 2-OH, -P (O) -OH, -P (O) (S) -OH, -CN, - (C1-C22) alkyl, - (C1-C22) alkenyl, - (C1-C22) alkylene-OH, - (C3-C22) cycloalkylene-OH, - (C6-C22) arylene-OH, - (C6-C22) heteroarylene-OH, - (C1-C22) alkylene-C (O) OH, - (C3-C22) cycloalkylene-C (O) OH, - (C6-C22) arylene-C (O) OH, - (C5-C22) heteroarylene-C (O) OH, -O-C (O) - (C1-C22) alkylene-C (O) NH2, - (C1-C22) alkylene-O-C (O) - (C1-C22) alkylene-C (O) NH2, -O-C (O) - (C1-C22) alkylene-C (O) OH, - (C1-C22) alkylene-O-C (O) - (C1-C22) alkylene-C (O) OH, -C (O) - (C1-C22) alkylene-C (O) NH2, - (C1-C22) alkylene-C (O) - (C1-C22) alkylene-C (O) NH2, -C (O) -NH- (C1-C22) alkylene-OH, -C (O) -NH- (C1-C22) alkylene-C (O) OH, - (C1-C22) alkylene-C (O) -NH- (C1-C22) alkylene-C (O) OH, - (C1-C30) alkylene-P (O) 2-OH, - (C1-C22) alkylene-O-P (-N (C1-C22 alkyl) 2) -O- (C1-C22) alkylene-CN, - (C1-C22) alkylene-O-P (-N (C1-C22 alkyl) 2) -O- (C1-C22) alkylene-OH, - (C1-C22) alkylene-O-P (-
N (C1-C22 alkyl) 2) -O- (C1-C22) alkylene-NH2, - (C1-C22) alkylene-O-P (-N (C1-C22 alkyl) 2) -O- (C1-C22) alkylene-C (O) OH, -C (O) -NH- (C1-C22) alkylene-O-P (-N (C1-C22 alkyl) 2) -O- (C1-C22) alkylene-CN, -C (O) -NH- (C1-C22) alkylene-O-P (-N (C1-C22 alkyl) 2) -O- (C1-C22) alkylene-OH, -C (O) -NH- (C1-C22) alkylene-O-P (-N (C1-C22 alkyl) 2) -O- (C1-C30) alkylene-C (O) OH, -C (O) -NH- (C1-C22) alkylene-O-P (-N (C1-C22 alkyl) 2) -O- (C1-C22) alkylene-NH2, - (C1-C22) alkylene-C (O) -NH- (C1-C22) alkylene-O-P (-N (C1-C22 alkyl) 2) -O- (C1-C22) alkylene-CN, - (C1-C22) alkylene-C (O) -NH- (C1-C22) alkylene-O-P (-N (C1-C22 alkyl) 2) -O- (C1-C22) alkylene-OH, - (C1-C22) alkylene-C (O) -NH- (C1-C22) alkylene-O-P (-N (C1-C22 alkyl) 2) -O- (C1-C22) alkylene-C (O) OH, - (C1-C22) alkylene-C (O) -NH- (C1-C22) alkylene-O-P (-N (C1-C22 alkyl) 2) -O- (C1-C22) alkylene-NH2, - (C1-C22) alkylene -P (O) -OH, - (C1-C22) alkylene-P (O) (S) -OH, - (C1-C22) alkylene-CN,
wherein each of A1, A2 and A3 is either absent or a substituent independently selected from the group consisting of -H, -OH, -O-R1, -SH, - (C1-C25) alkyl, halogenated - (C1-C25) alkyl, - (C2-C22) alkenyl, - (C1-C22) alkylene-OH, - (C3-C22) cycloalkyl, - (C3-C22) cycloalkenyl, - (C1-C22) alkylene- (C3-C22) cycloalkyl, - (C1-C22) alkylene-R1, - (C1-C22) alkylene-O-R1, - (C1-C22) alkylene-COOR1, -C (O) O-R1, -O- (C1-C22) alkyl, -S- (C1-C22) alkyl, -C (O) -R1, -C (O) - (C1-C22) alkyl, -O-C (O) - (C1-C22) alkyl, -O-C (O) -R1, - (C1-C22) alkylene-O-C (O) -R1, -C (O) - (C1-C22) alkylene-OH, -C (O) - (C1-C22) alkylene-R1, -C (O) - (C1-C22) alkylene-NH-R1, -C (O) - (C1-C22) alkylene-NR2-R1, -O-C (O) - (C1-C22) alkylene-OH, -O-C (O) - (C1-C22) alkylene-R1, -adamantyl, - (C1-C22) alkylene-adamantyl, -O-adamantly, -C (O) - (C1-C22) alkylene-adamantyl, - (C1-C22) alkylene-C (O) - (C1-C22) alkylene-adamantyl, -NH-C (O) - (C1-C22) alkylene-adamantyl, - (C1-C22) alkylene-NH-C (O) - (C1-C22) alkyl, - (C1-C22) alkylene-NH-C (O) -halogenated (C1-C22) alkyl, -CH (NH-CO- (C1-C22) alkyl) - (C1-C22) alkylene-NH-C (O) - (C1-C22) alkyl, -C (O) - (C1-C22) alkylene-C (O) -NH-C [- (C1-C22) alkylene-O- (C1-C22) alkylene-C (O) -NH- (C1-C22) alkylene-NH-C (O) - (C1-C22) alkyl] 3, -C (O) - (C1-C22) alkylene-C (O) -NH-C [- (C1-C22) alkylene-O- (C1-C22) alkylene-C (O) -NH- (C1-C22) alkylene-NH-C (O) - (C1-C22) alkylene-R1] 3, -CH (NH-CO-halogenated (C1-C22) alkyl) - (C1-C22) alkylene-NH-C (O) -halogenated (C1-C22) alkyl, - (C1-C22) alkylene-NH-C (O) - (C1-C22) alkylene-adamantyl, - (C1-C22) alkylene-NR2-C (O) - (C1-C22) alkylene-adamantyl, - (C1-C22) alkylene- (C1-C6 alkylene oxide) (1-20) -NH-C (O) - (C1-C22) alkylene-adamantyl, -C (O) NH- (C1-C22) alkyl, -C (O) NH-R1, -C (O) NR2-R1, -C (O) NH- (C1-C22) alkylene-OH, -C (O) NH- (C1-C22) alkylene-COOH, -NH-C (O) - (C1-C22) alkyl, -NH-C (O) -R1, -NR2-C (O) -R1, -O-P (O) 2-O-R1, -OP (O) (S) -O-R1, -O-P (O) -O-R1, -NH-R1, -NR2-R1, - (C1-
C22) alkylene-NH-R1, - (C1-C22) alkylene-NR2-R1, -C (O) - (C1-C22) alkylene-C (O) -R1, -C (O) - (C1-C22) alkylene-C (O) O-R1, -C (O) - (C1-C22) alkylene-NH-C (O) -R1, -C (O) - (C1-C22) alkylene-NR2-C (O) -R1, - (C1-C22) alkylene-C (O) -R1, - (C1-C22) alkylene-NH-C (O) -R1, - (C1-C22) alkylene-NH-C (O) - (C1-C22) alkylene-R1, - (C1-C22) alkylene-NH-C (O) - (C1-C22) alkylene-NH-C (O) - (C1-C22) alkylene-R1, - (C1-C22) alkylene-C (O) -NH- (C1-C22) alkylene-R1, - (C1-C22) alkylene-C (O) -NH- (C1-C22) alkylene-C (O) -NH- (C1-C22) alkylene-R1, - (C1-C22) alkylene-C (O) -NH- (C1-C22) alkylene-NH-C (O) - (C1-C22) alkylene-R1, - (C1-C22) alkylene-NR2-C (O) - (C1-C22) alkylene-R1, - (C1-C22) alkylene-O-P (-N (C1-C22 alkyl) 2) -O- (C1-C22) alkylene-CN, - (C1-C22) alkylene-O-P (-N (C1-C22 alkyl) 2) -O- (C1-C22) alkylene-OH, - (C1-C22) alkylene-O-P (-N (C1-C22 alkyl) 2) -O- (C1-C22) alkylene-NH2, - (C1-C22) alkylene-O-P (-N (C1-C22 alkyl) 2) -O- (C1-C22) alkylene-C (O) OH, -C (O) -NH- (C1-C22) alkylene-O-P (-N (C1-C22 alkyl) 2) -O- (C1-C22) alkylene-CN, -C (O) -NH- (C1-C22) alkylene-O-P (-N (C1-C22 alkyl) 2) -O- (C1-C22) alkylene-OH, -C (O) -NH- (C1-C22) alkylene-O-P (-N (C1-C22 alkyl) 2) -O- (C1-C30) alkylene-C (O) OH, -C (O) -NH- (C1-C22) alkylene-O-P (-N (C1-C22 alkyl) 2) -O- (C1-C22) alkylene-NH2, - (C1-C22) alkylene-C (O) -NH- (C1-C22) alkylene-O-P (-N (C1-C22 alkyl) 2) -O- (C1-C22) alkylene-CN, - (C1-C22) alkylene-C (O) -NH- (C1-C22) alkylene-O-P (-N (C1-C22 alkyl) 2) -O- (C1-C22) alkylene-OH, - (C1-C22) alkylene-C (O) -NH- (C1-C22) alkylene-O-P (-N (C1-C22 alkyl) 2) -O- (C1-C22) alkylene-C (O) OH, - (C1-C22) alkylene-C (O) -NH- (C1-C22) alkylene-O-P (-N (C1-C22 alkyl) 2) -O- (C1-C22) alkylene-NH2, - (C1-C22) alkylene -P (O) -OH, - (C1-C22) alkylene-P (O) (S) -OH, - (C1-C22) alkylene-CN, substituted or unsubstituted pyrrole, substituted or unsubstituted pyrroline, substituted or unsubstituted pyrrolidine, substituted or unsubstituted pyrazole, substituted or unsubstituted pyrazoline, substituted or unsubstituted pyrazolidine, substituted or unsubstituted imidazole, substituted or unsubstituted oxazole, substituted or unsubstituted thiazole, substituted or unsubstituted benzopyrrole, substituted or unsubstituted benzopyrroline, substituted or unsubstituted benzopyrrolidine, substituted or unsubstituted benzopyrazole, substituted or unsubstituted benzopyrazoline, substituted or unsubstituted benzopyrazolidine, substituted or unsubstituted benzoimidazole, substituted or unsubstituted benzooxazole, substituted or unsubstituted benzothiazole, and a substituent represented by Formula AIII,
wherein Y is selected from the group consisting of carbon, nitrogen, oxygen and sulfur; and each of P, Q, S and T is independently selected from the group consisting of carbon, nitrogen, oxygen and sulfur; and the asterisk refers to the site wherein the substituent represented by Formula AIII is linked with the structure represented by Formula AI or Formula AII;
wherein each of R3, R4 and R5 is either absent or a substituent independently selected from the group consisting of -H, -OH, -O-R1, -SH, - (C1-C25) alkyl, halogenated - (C1-C25) alkyl, - (C2-C22) alkenyl, - (C1-C22) alkylene-OH, - (C3-C22) cycloalkyl, - (C3-C22) cycloalkenyl, - (C1-C22) alkylene- (C3-C22) cycloalkyl, - (C1-C22) alkylene-R1, - (C1-C22) alkylene-O-R1, - (C1-C22) alkylene-COOR1, -C (O) O-R1, -O- (C1-C22) alkyl, -S- (C1-C22) alkyl, -C (O) -R1, -C (O) - (C1-C22) alkyl, -O-C (O) - (C1-C22) alkyl, -O-C (O) -R1, - (C1-C22) alkylene-O-C (O) -R1, -C (O) - (C1-C22) alkylene-OH, -C (O) - (C1-C22) alkylene-R1, -C (O) - (C1-C22) alkylene-NH-R1, -C (O) - (C1-C22) alkylene-NR2-R1, -O-C (O) - (C1-C22) alkylene-OH, -O-C (O) - (C1-C22) alkylene-R1, -adamantyl, - (C1-C22) alkylene-adamantyl, -O-adamantly, -C (O) - (C1-C22) alkylene-adamantyl, - (C1-C22) alkylene-C (O) - (C1-C22) alkylene-adamantyl, -NH-C (O) - (C1-C22) alkylene-adamantyl, - (C1-C22) alkylene-NH-C (O) - (C1-C22) alkyl, - (C1-C22) alkylene-NH-C (O) -halogenated (C1-C22) alkyl, -CH (NH-CO- (C1-C22) alkyl) - (C1-C22) alkylene-NH-C (O) - (C1-C22) alkyl, -C (O) - (C1-C22) alkylene-C (O) -NH-C [- (C1-C22) alkylene-O- (C1-C22) alkylene-C (O) -NH- (C1-C22) alkylene-NH-C (O) - (C1-C22) alkyl] 3, -C (O) - (C1-C22) alkylene-C (O) -NH-C [- (C1-C22) alkylene-O- (C1-C22) alkylene-C (O) -NH- (C1-C22) alkylene-NH-C (O) - (C1-C22) alkylene-R1] 3, -CH (NH-CO-halogenated (C1-C22) alkyl) - (C1-C22) alkylene-NH-C (O) -halogenated (C1-C22) alkyl, - (C1-C22) alkylene-NH-C (O) - (C1-C22) alkylene-adamantyl, - (C1-C22) alkylene-NR2-C (O) - (C1-C22) alkylene-adamantyl, - (C1-C22) alkylene- (C1-C6 alkylene oxide) (1-20) -NH-C (O) - (C1-C22) alkylene-adamantyl, -C (O) NH- (C1-C22) alkyl, -C (O) NH-R1, -C (O) NR2-R1, -C (O) NH- (C1-C22) alkylene-OH, -C (O) NH- (C1-C22) alkylene-COOH, -NH-C (O) - (C1-C22) alkyl, -NH-C (O) -
R1, -NR2-C (O) -R1, -O-P (O) 2-O-R1, -OP (O) (S) -O-R1, -O-P (O) -O-R1, -NH-R1, -NR2-R1, - (C1-C22) alkylene-NH-R1, - (C1-C22) alkylene-NR2-R1, -C (O) - (C1-C22) alkylene-C (O) -R1, -C (O) - (C1-C22) alkylene-C (O) O-R1, -C (O) - (C1-C22) alkylene-NH-C (O) -R1, -C (O) - (C1-C22) alkylene-NR2-C (O) -R1, - (C1-C22) alkylene-C (O) -R1, - (C1-C22) alkylene-NH-C (O) -R1, - (C1-C22) alkylene-NH-C (O) - (C1-C22) alkylene-R1, - (C1-C22) alkylene-NH-C (O) - (C1-C22) alkylene-NH-C (O) - (C1-C22) alkylene-R1, - (C1-C22) alkylene-C (O) -NH- (C1-C22) alkylene-R1, - (C1-C22) alkylene-C (O) -NH- (C1-C22) alkylene-C (O) -NH- (C1-C22) alkylene-R1, - (C1-C22) alkylene-C (O) -NH- (C1-C22) alkylene-NH-C (O) - (C1-C22) alkylene-R1, - (C1-C22) alkylene-NR2-C (O) - (C1-C22) alkylene-R1, - (C1-C22) alkylene-O-P (-N (C1-C22 alkyl) 2) -O- (C1-C22) alkylene-CN, - (C1-C22) alkylene-O-P (-N (C1-C22 alkyl) 2) -O- (C1-C22) alkylene-OH, - (C1-C22) alkylene-O-P (-N (C1-C22 alkyl) 2) -O- (C1-C22) alkylene-NH2, - (C1-C22) alkylene-O-P (-N (C1-C22 alkyl) 2) -O- (C1-C22) alkylene-C (O) OH, -C (O) -NH- (C1-C22) alkylene-O-P (-N (C1-C22 alkyl) 2) -O- (C1-C22) alkylene-CN, -C (O) -NH- (C1-C22) alkylene-O-P (-N (C1-C22 alkyl) 2) -O- (C1-C22) alkylene-OH, -C (O) -NH- (C1-C22) alkylene-O-P (-N (C1-C22 alkyl) 2) -O- (C1-C30) alkylene-C (O) OH, -C (O) -NH- (C1-C22) alkylene-O-P (-N (C1-C22 alkyl) 2) -O- (C1-C22) alkylene-NH2, - (C1-C22) alkylene-C (O) -NH- (C1-C22) alkylene-O-P (-N (C1-C22 alkyl) 2) -O- (C1-C22) alkylene-CN, - (C1-C22) alkylene-C (O) -NH- (C1-C22) alkylene-O-P (-N (C1-C22 alkyl) 2) -O- (C1-C22) alkylene-OH, - (C1-C22) alkylene-C (O) -NH- (C1-C22) alkylene-O-P (-N (C1-C22 alkyl) 2) -O- (C1-C22) alkylene-C (O) OH, - (C1-C22) alkylene-C (O) -NH- (C1-C22) alkylene-O-P (-N (C1-C22 alkyl) 2) -O- (C1-C22) alkylene-NH2, - (C1-C22) alkylene -P (O) -OH, - (C1-C22) alkylene-P (O) (S) -OH, - (C1-C22) alkylene-CN, substituted or unsubstituted pyrrole, substituted or unsubstituted pyrroline, substituted or unsubstituted pyrrolidine, substituted or unsubstituted pyrazole, substituted or unsubstituted pyrazoline, substituted or unsubstituted pyrazolidine, substituted or unsubstituted imidazole, substituted or unsubstituted oxazole, substituted or unsubstituted thiazole, substituted or unsubstituted benzopyrrole, substituted or unsubstituted benzopyrroline, substituted or unsubstituted benzopyrrolidine, substituted or unsubstituted benzopyrazole, substituted or unsubstituted benzopyrazoline, substituted or unsubstituted benzopyrazolidine, substituted or unsubstituted benzoimidazole, substituted or unsubstituted benzooxazole, and substituted or unsubstituted benzothiazole,
wherein R7, on each occurrence, is attached to any one of P, Q, S and T, and is either absent or selected from the group consisting of hydrogen, halogen atom, hydroxyl, (C1-C20) alkyl, (C1-C20) alkoxy, halogenated (C1-C20) alkyl and halogenated (C1-C20) alkoxy;
wherein M is an integer of 0, 1, 2 or 3;
wherein R6 is attached to any one of P, Q, S and T, and is selected from the group consisting of direct bond, -O-, -C (O) O-, -O-C (O) -, -P (O) 2-O-, -O-P (O) 2-O-, -P (O) (S) -O-, -O-P (O) (S) -O-, -O-P (O) -O-, - (C1-C22) alkylene-, - (C1-C22) alkylene-O-, -O- (C1-C22) alkylene-, - (C1-C22) alkylene-NH-, -NH- (C1-C22) alkylene-, -C (O) - (C1-C22) alkylene-, - (C1-C22) alkylene-C (O) -, -C (O) -O- (C1-C22) alkylene-, - (C1-C30) alkylene-C (O) -O-, -C (O) -NH- (C1-C22) alkylene-, -C (O) -NH-(C1-C22) alkylene-C (O) -O-, -C (O) -NH- (C1-C22) alkylene-O-C (O) -, -C (O) -NH- (C1-C22) alkylene-O-C (O) -O-, -C (O) -NH- (C1-C22) alkylene-O-, -C (O) -N ( (C1-C22) alkyl) - (C1-C22) alkylene-, -C (O) -N ( (C1-C22) alkyl) - (C1-C22) alkylene-O-, -C (O) -NH- (C1-C22) alkylene-C (O) -NH- (C1-C22) alkylene-, -C (O) -NH- (C1-C22) alkylene-NH-C (O) - (C1-C22) alkylene-, -C (O) -NH- (C1-C22) alkylene-C (O) -NH-, -C (O) -N ( (C1-C22) alkyl) - (C1-C22) alkylene-C (O) -N ( (C1-C22) alkyl) - (C1-C22) alkylene-, -C (O) -N ( (C1-C22) alkyl) - (C1-C22) alkylene-C (O) -N ( (C1-C22) alkyl) -, - (C1-C22) alkylene-C (O) -NH- (C1-C22) alkylene-C (O) -NH- (C1-C22) alkylene-, - (C1-C22) alkylene-C (O) -NH- (C1-C22) alkylene-C (O) -NH-, -NH-C (O) - (C1-C22) alkylene-, -NH-C (O) - (C1-C22) alkylene-C (O) -O-, -NH-C (O) - (C1-C22) alkylene-C (O) -, -NH-C (O) - (C1-C22) alkylene-O-, -N ( (C1-C22) alkyl) -C (O) - (C1-C22) alkylene-, -N ( (C1-C22) alkyl) -C (O) - (C1-C22) alkylene-O-, -NH-C (O) - (C1-C22) alkylene-NH-C (O) - (C1-C22) alkylene-, -NH-C (O) - (C1-C22) alkylene-NH-C (O) -, -N ( (C1-C22) alkyl) -C (O) - (C1-C22) alkylene-N ( (C1-C22) alkyl) -C (O) - (C1-C22) alkylene-, -N ( (C1-C22) alkyl) -C (O) - (C1-C22) alkylene-N ( (C1-C22) alkyl) -C (O) -, - (C1-C22) alkylene-NH-C (O) - (C1-C22) alkylene-NH-C (O) - (C1-C22) alkylene-, - (C1-C22) alkylene-NH-C (O) - (C1-C22) alkylene-NH-C (O) -, - (C1-C22) alkylene -P (O) 2-O-, - (C1-C22) alkylene-O-P (O) 2-O-, - (C3-C22) cycloalkylene-, - (C3-C22) cycloalkylene-O-, -O- (C3-C22) cycloalkylene-, - (C6-C22) arylene-, - (C6-C22) arylene-O-, -O- (C6-C22) arylene-, - (C6-C22) arylene-NH-, -NH- (C6-C22) arylene-, -C (O) - (C6-C22) arylene-, - (C6-C22) arylene-C (O) -, -C (O) -O- (C6-C22) arylene-, - (C6-C22) arylene-C (O) -O-, -C (O) -NH- (C6-C22) arylene-and -C (O) -NH- (C6-C22) arylene-C (O) -O-;
wherein R1, on each occurrence, is independently selected from the group consisting of hydrogen, hydroxyl, - (C1-C22) alkyl, - (C3-C22) cycloalkyl, - (C6-C22) aryl, - (C1-C22) alkoxy, - (C3-C22) cycloalkoxy, - (C6-C22) aryloxy, -C (O) - (C1-C22) alkyl, -OC (O) (C1-C22) alkyl, -C (O) -O- (C1-C22) alkyl, -C (O) - (C3-C22) cycloalkyl, -OC (O) - (C3-C22) cycloalkyl, -C (O) -O- (C3-C22) cycloalkyl, -C (O) - (C6-C22) aryloxy, -OC (O) - (C6-C22) aryloxy, -C (O) -O- (C6-C22) aryloxy, -C (O) -phosphate ester group, phosphodiester group, phosphoramidite group, saturated fatty
acid group, unsaturated fatty acid group, glucosyl, acetamide glucosyl, galactosamine, N-acetyl galactosamine, lipid, PEG, steroid, lipophile, carbohydrate, cholesterol, adamantane, amino acid, peptide, chloroquine and alkaloid,
wherein R2, on each occurrence, is independently selected from the group consisting of a halogen atom, a (C1-C12) alkyl, a (C1-C12) alkoxy, a (C1-C12) alkoxycarbonyl, a (C6-C16) aryl or a (C6-C16) aryloxycarbonyl;
wherein one or more hydroxyl group, carboxyl group, amino group, nitrile group and phosphoric acid group contained in A1, A2, A3, B, C, R1, R2, R3, R4, R5 and R6 are optionally linked to a support material or protected with a terminal protective group; and
with the proviso that A1, A2 and A3 are not simultaneously hydrogen and R3, R4 and R5 are not simultaneously hydrogen.
In another specific embodiment, the oligonucleotide delivery enhancing compound according to claim 1, comprising a moiety represented by Formula BI and at least one substituent directly or indirectly attachable to an oligonucleotide,
wherein X′ is selected from the group consisting of carbon, nitrogen, oxygen and sulfur; each of F′, G′, H′ and I′ is independently selected from the group consisting of carbon, nitrogen, oxygen and sulfur; and each of the asterisks refers to a site optionally linked to at least one substituent or an oligonucleotide directly or indirectly.
In another specific embodiment, the oligonucleotide delivery enhancing compound has a structure represented by Formula BII
wherein X′ is selected from the group consisting of carbon, nitrogen, oxygen and sulfur; and each of F′, G′, H′ and I′ is independently selected from the group consisting of carbon,
nitrogen, oxygen and sulfur;
wherein each of A1′, A2′ and A3′ is either absent or a substituent independently selected from the group consisting of -H, -R1′, -O-R1′, -S-R1′, -C (O) -R1′, -C (O) O-R1′, -O-C (O) -R1′, -C (O) NH-R1′, -C (O) NR2′-R1′, -NH-C (O) -R1′, -NR2′-C (O) -R1′, -O-P (O) 2-O-R1′, -OP (O) (S) -O-R1′, -O-P (O) -O-R1′, -NH-R1′, -NR2′-R1′, - (CH2) r′-NH-R1′, - (CH2) r′-NR2′-R1′, -C (O) - (CH2) r′-R1′, -C (O) - (CH2) r′-NH-R1′, -C (O) - (CH2) r′-NR2′-R1′, -C (O) - (CH2) r′-C (O) -R1′, -C (O) - (CH2) r′-C (O) O-R1′, -C (O) - (CH2) r′-NH-C (O) -R1′, -C (O) - (CH2) r′-NR2′-C (O) -R1′, - (CH2) r′-C (O) -R1′; - (CH2) r′-C (O) O-R1′; - (CH2) r′-O-C (O) -R1′, - (CH2) r′-R1′, - (CH2) r′-NH-C (O) -R1′, - (CH2) r′-NH-C (O) - (CH2) s′-R1′, - (CH2) r′-NH-C (O) - (C1-C22) alkylene-NH-C (O) - (CH2) s′-R1′, - (CH2) r′-C (O) -NH- (CH2) s′-R1′, - (CH2) r′-C (O) -NH- (C1-C22) alkylene-C (O) -NH- (CH2) s′-R1′, - (CH2) r′-C (O) -NH- (C1-C22) alkylene-NH-C (O) - (CH2) s′-R1′, - (CH2) r′-NR2′-C (O) - (CH2) s′-R1′, -CH (- (CH2) r′-NH-C (O) - (CH2) s′-R1′) (-NH-C (O) - (CH2) q′-R3′) , -CH (- (CH2) r′-C (O) -NH- (CH2) s′-R1′) (-C (O) -NH- (CH2) q′-R3′) , -N (- (CH2) r′-NH-C (O) - (CH2) s′-R1′) (-NH-C (O) - (CH2) q′-R3′) , -CH (- (CH2) r′-NH-C (O) - (CH2) s′-R1′) (- (CH2) p′-NH-C (O) - (CH2) q′-R3′) , -CR4′ (- (CH2) r′-NH-C (O) - (CH2) s′-R1′) (-NH-C (O) - (CH2) q′-R3′) , -CR4′ (- (CH2) r′-NH-C (O) - (CH2) s′-R1′) (- (CH2) p′-NH-C (O) - (CH2) q′-R3′) , -CH (- (CH2) r′-NR5′-C (O) - (CH2) s′-R1′) (-NR6′-C (O) - (CH2) q′-R3′) , -CH (- (CH2) r′-NR5′-C (O) - (CH2) s′-R1′) (- (CH2) p′-NR6′-C (O) - (CH2) q′-R3′) , -CR4′ ( (CH2) r′-NR5′-C (O) - (CH2) s′-R1′) ( (CH2) p′-NR6′-C (O) - (CH2) q′-R3′) , -CR4′ ( (CH2) r′-NR5′-C (O) - (CH2) s′-R1′) ( (CH2) p′-NR6′-C (O) - (CH2) q′-R3′) , or A1′ and A2′ are linked together so that A1′, A2′, the nitrogen atom linked with A1′ and the carbon atom linked with A2′ form a unsubstituted or substituted heterocyclic ring; wherein each of R1′ and R3′ is independently selected from the group consisting of hydrogen, hydroxyl, - (C1-C30) alkyl, - (C3-C50) cycloalkyl, - (C6-C50) aryl, - (C1-C30) alkoxy, - (C3-C50) cycloalkoxy, - (C6-C50) aryloxy, -C (O) - (C1-C30) alkyl, -OC (O) (C1-C30) alkyl, -C (O) -O- (C1-C30) alkyl, -C (O) - (C3-C50) cycloalkyl, -OC (O) - (C3-C50) cycloalkyl, -C (O) -O- (C3-C50) cycloalkyl, -C (O) - (C6-C50) aryloxy, -OC (O) - (C6-C50) aryloxy, -C (O) -O- (C6-C50) aryloxy, -C (O) -phosphate ester group, phosphodiester group, phosphoramidite group, saturated fatty acid group, unsaturated fatty acid group, glucosyl, acetamide glucosyl, galactosamine, N-acetyl galactosamine, lipid, PEG, steroid, lipophile, carbohydrate, cholesterol, adamantane, amino acid, peptide, ligand, nucleic acid, oligonucleotide, aptamer, small molecule, antibody, antibody fragment, chloroquine, alkaloid and targeting moiety, wherein one or more hydroxyl group, carboxyl group and amino
group contained in each of R1′ and R3′ are optionally protected; wherein each of R2′, R4′, R5′ and R6′ is independently a halogen atom, a (C1-C12) alkyl, a (C1-C12) alkoxy, a (C1-C12) alkoxycarbonyl, a (C6-C16) aryl or a (C6-C16) aryloxycarbonyl; wherein each of r′, s′, p′ and q′ is an integer from 1 to 22; and with the proviso that A3′ is absent when X′ is oxygen, and A1′, A2′ and A3′ are not simultaneously hydrogen;
wherein each C′ is attached to any one of F′, G′, H′ and I′, and is either absent or selected from the group consisting of hydrogen, halogen atom, hydroxyl, (C1-C20) alkyl, (C1-C20) alkoxy, halogenated (C1-C20) alkyl and halogenated (C1-C20) alkoxy;
wherein m′ is an integer of 1, 2 or 3, n′ is an integer of 1, 2 or 3, and m′+n′=4;
wherein each B′ is attached to any one of F′, G′, H′ and I′, and is independently selected from the group consisting of hydroxyl, -C (O) OH, - (C1-C30) alkoxy, -P (O) 2-OH, -P (O) -OH, -P (O) (S) -OH, -CN, - (C1-C30) alkylene-OH, - (C3-C50) cycloalkylene-OH, - (C6-C50) arylene-OH, - (C5-C50) heteroarylene-OH, - (C1-C30) alkylene-C (O) OH, - (C3-C50) cycloalkylene-C (O) OH, - (C6-C50) arylene-C (O) OH, - (C5-C50) heteroarylene-C (O) OH, -C (O) -NH- (C1-C30) alkylene-OH, -C (O) -NH- [ (C1-C30) alkylene-O] r′-H (wherein r′ is an integer of 1 to 22) , -C (O) -NH- [ (C1-C30) alkylene-O] r′- (C1-C30) alkylene-C (O) -OH (wherein r′ is an integer of 1 to 22) , -C (O) -NH- (C3-C50) cycloalkylene-OH, -C (O) -NH- (C6-C50) arylene-OH, -C (O) -NH- (C5-C50) heteroarylene-OH, -C (O) -NH- (C1-C30) alkylene-C (O) OH, -C (O) -NH- (C3-C50) cycloalkylene-C (O) OH, -C (O) -NH- (C6-C50) arylene-C (O) OH, -C (O) -NH- (C1-C30) alkylene- (C6-C50) arylene- (C1-C30) alkylene-C (O) OH, -C (O) -NH- (C5-C50) heteroarylene-C (O) OH, - (C1-C30) alkylene-C (O) -NH- (C1-C30) alkylene-C (O) OH, - (C1-C30) alkylene-C (O) -NH- (C3-C50) cycloalkylene-C (O) OH, - (C1-C30) alkylene-C (O) -NH- (C6-C50) arylene-C (O) OH, - (C1-C30) alkylene-C (O) -NH- (C5-C50) heteroarylene-C (O) OH, - (C1-C30) alkylene-P (O) 2-OH, - (C3-C50) cycloalkylene-P (O) 2-OH, - (C6-C50) arylene-P (O) 2-OH, - (C5-C50) heteroarylene-P (O) 2-OH, -C (O) -NH- (C1-C30) alkylene-O-P (-N (C1-C16alkyl) 2) -O- (C1-C30) alkylene-CN, -C (O) -NH- (C1-C30) alkylene-O-P (-N (C1-C16alkyl) 2) -O- (C1-C30) alkylene-OH, -C (O) -NH- (C1-C30) alkylene-O-P (-N (C1-C16alkyl) 2) -O- (C1-C30) alkylene-C (O) OH, -C (O) -NH- (C1-C30) alkylene-O-P (-N (C1-C16alkyl) 2) -O- (C1-C30) alkylene-NH2, - (C1-C30) alkylene-C (O) -NH- (C1-C30) alkylene-O-P (-N (C1-C16alkyl) 2) -O- (C1-C30) alkylene-CN, - (C1-C30) alkylene-C (O) -NH- (C1-C30) alkylene-O-P (-N (C1-C16alkyl) 2) -O- (C1-C30) alkylene-OH, - (C1-C30) alkylene-C (O) -NH- (C1-C30) alkylene-O-P (-N (C1-C16alkyl) 2) -O- (C1-C30) alkylene-C (O) OH, - (C1-C30) alkylene-C (O) -NH- (C1-C30) alkylene-O-P (-N (C1-
C16alkyl) 2) -O- (C1-C30) alkylene-NH2, - (C1-C30) alkylene -P (O) -OH, - (C3-C50) cycloalkylene-P (O) -OH, - (C6-C50) arylene-P (O) -OH, - (C5-C50) -heteroarylene-P (O) -OH, - (C1-C30) alkylene-P (O) (S) -OH, - (C3-C50) cycloalkylene-P (O) (S) -OH, - (C6-C50) arylene-P (O) (S) -OH, - (C5-C50) heteroarylene-P (O) (S) -OH, - (C1-C30) alkylene-CN, - (C3-C50) cycloalkylene-CN, - (C6-C50) arylene-CN, - (C5-C50) heteroarylene-CN, lipid, PEG, steroid, lipophile, carbohydrate, cholesterol, adamantane, amino acid, peptide, chloroquine, alkaloid and a substituent represented by Formula BIII:
wherein Y′ is selected from the group consisting of carbon, nitrogen, oxygen and sulfur; and each of P′, Q′, S′ and T′ is independently selected from the group consisting of carbon, nitrogen, oxygen and sulfur; and the asterisk refers to the site wherein the substituent represented by Formula BIII is linked with any one of F′, G′, H′ and I′ of Formula BII;
where R7′ is selected from the group consisting of -O-, -C (O) O-, -O-C (O) -, -P (O) 2-O-, -O-P (O) 2-O-, -P (O) (S) -O-, -O-P (O) (S) -O-, -O-P (O) -O-, - (C1-C30) alkylene-, - (C1-C30) alkylene-O-, -O- (C1-C30) alkylene-, - (C1-C30) alkylene-NH-, -NH- (C1-C30) alkylene-, -C (O) - (C1-C30) alkylene-, - (C1-C30) alkylene-C (O) -, -C (O) -O- (C1-C30) alkylene-, - (C1-C30) alkylene-C (O) -O-, -C (O) -NH- (C1-C30) alkylene-, -C (O) -NH- (C1-C30) alkylene-C (O) -O-, -C (O) -NH- (C1-C30) alkylene-O-C (O) -, -C (O) -NH- (C1-C30) alkylene-O-C (O) -O-, -C (O) -NH- (C1-C30) alkylene-O-, -C (O) -N ( (C1-C20) alkyl) - (C1-C30) alkylene-, -C (O) -N ( (C1-C20) alkyl) - (C1-C30) alkylene-O-, -C (O) -NH- (C1-C30) alkylene-C (O) -NH- (C1-C30) alkylene-, -C (O) -NH- (C1-C30) alkylene-NH-C (O) - (C1-C30) alkylene-, -C (O) -NH- (C1-C30) alkylene-C (O) -NH-, -C (O) -N ( (C1-C20) alkyl) - (C1-C30) alkylene-C (O) -N ( (C1-C20) alkyl) - (C1-C30) alkylene-, -C (O) -N ( (C1-C20) alkyl) - (C1-C30) alkylene-C (O) -N ( (C1-C20) alkyl) -, - (C1-C30) alkylene-C (O) -NH- (C1-C30) alkylene-C (O) -NH- (C1-C30) alkylene-, - (C1-C30) alkylene-C (O) -NH- (C1-C30) alkylene-C (O) -NH-, -NH-C (O) - (C1-C30) alkylene-, -NH-C (O) - (C1-C30) alkylene-C (O) -O-, -NH-C (O) - (C1-C30) alkylene-C (O) -, -NH-C (O) - (C1-C30) alkylene-O-, -N ( (C1-C20) alkyl) -C (O) - (C1-C30) alkylene-, -N ( (C1-C20) alkyl) -C (O) - (C1-C30) alkylene-O-, -NH-C (O) - (C1-C30) alkylene-NH-C (O) - (C1-C30) alkylene-, -NH-C (O) - (C1-C30) alkylene-NH-C (O) -, -N ( (C1-C20) alkyl) -C (O) - (C1-C30) alkylene-N ( (C1-C20) alkyl) -
C (O) - (C1-C30) alkylene-, -N ( (C1-C20) alkyl) -C (O) - (C1-C30) alkylene-N ( (C1-C20) alkyl) -C (O) -, - (C1-C30) alkylene-NH-C (O) - (C1-C30) alkylene-NH-C (O) - (C1-C30) alkylene-, - (C1-C30) alkylene-NH-C (O) - (C1-C30) alkylene-NH-C (O) -, - (C1-C30) alkylene -P (O) 2-O-, - (C1-C30) alkylene-O-P (O) 2-O-, - (C3-C50) cycloalkylene-, - (C3-C50) cycloalkylene-O-, -O- (C3-C50) cycloalkylene-, - (C3-C50) cycloalkylene-NH-, -NH- (C3-C50) cycloalkylene-, -C (O) - (C3-C50) cycloalkylene-, - (C3-C50) cycloalkylene-C (O) -, -C (O) -O- (C3-C50) cycloalkylene-, - (C3-C50) cycloalkylene-C (O) -O-, -C (O) -NH- (C3-C50) cycloalkylene-, -C (O) -NH- (C3-C50) cycloalkylene-C (O) -O-, -C (O) -NH- (C3-C50) cycloalkylene-O-, -C (O) -N ( (C1-C20) alkyl) - (C3-C50) cycloalkylene-, -C (O) -N ( (C1-C20) alkyl) - (C3-C50) cycloalkylene-O-, -C (O) -NH- (C3-C50) cycloalkylene-C (O) -NH- (C3-C50) cycloalkylene-, -C (O) -NH- (C3-C50) cycloalkylene-C (O) -NH-, -C (O) -N ( (C1-C20) alkyl) - (C3-C50) cycloalkylene-C (O) -N ( (C1-C20) alkyl) - (C3-C50) cycloalkylene-, -C (O) -N ( (C1-C20) alkyl) - (C3-C50) cycloalkylene-C (O) -N ( (C1-C20) alkyl) -, - (C3-C50) cycloalkylene-C (O) -NH- (C3-C50) cycloalkylene-C (O) -NH- (C3-C50) cycloalkylene-, - (C3-C50) cycloalkylene-C (O) -NH- (C3-C50) cycloalkylene-C (O) -NH-, - (C3-C50) cycloalkylene -P (O) 2-O-, - (C3-C50) cycloalkylene-O-P (O) 2-O-, - (C6-C50) arylene-, - (C6-C50) arylene-O-, -O- (C6-C50) arylene-, - (C6-C50) arylene-NH-, -NH- (C6-C50) arylene-, -C (O) - (C6-C50) arylene-, - (C6-C50) arylene-C (O) -, -C (O) -O- (C6-C50) arylene-, - (C6-C50) arylene-C (O) -O-, -C (O) -NH- (C6-C50) arylene-, -C (O) -NH- (C6-C50) arylene-C (O) -O-, -C (O) -NH- (C6-C50) arylene-O-, -C (O) -N ( (C1-C20) alkyl) - (C6-C50) arylene-, -C (O) -N ( (C1-C20) alkyl) - (C6-C50) arylene-O-, -C (O) -NH- (C6-C50) arylene-C (O) -NH- (C3-C50) cycloalkylene-, -C (O) -NH- (C6-C50) arylene-C (O) -NH-, -C (O) -N ( (C1-C20) alkyl) - (C6-C50) arylene-C (O) -N ( (C1-C20) alkyl) - (C6-C50) arylene-, -C (O) -N ( (C1-C20) alkyl) - (C6-C50) arylene-C (O) -N ( (C1-C20) alkyl) -, - (C6-C50) arylene-C (O) -NH- (C6-C50) arylene-C (O) -NH- (C6-C50) arylene-, - (C6-C50) arylene-C (O) -NH- (C6-C50) arylene-C (O) -NH-, - (C6-C50) arylene -P (O) 2-O-, - (C6-C50) arylene-O-P (O) 2-O-, - (C5-C50) heteroarylene-, - (C5-C50) heteroarylene-O-, -O- (C5-C50) heteroarylene-, - (C5-C50) heteroarylene-NH-, -NH- (C5-C50) heteroarylene-, -C (O) - (C5-C50) heteroarylene-, - (C5-C50) heteroarylene-C (O) -, -C (O) -O- (C5-C50) heteroarylene-, - (C5-C50) heteroarylene-C (O) -O-, -C (O) -NH- (C5-C50) heteroarylene-, -C (O) -NH- (C5-C50) heteroarylene-C (O) -O-, -C (O) -NH- (C5-C50) heteroarylene-O-, -C (O) -N ( (C1-C20) alkyl) - (C5-C50) heteroarylene-, -C (O) -N ( (C1-C20) alkyl) - (C5-C50) heteroarylene-O-, -C (O) -NH- (C5-C50) heteroarylene-C (O) -NH- (C3-C50) cycloalkylene-, -C (O) -NH- (C5-C50) heteroarylene-C (O) -NH-, -C (O) -N ( (C1-C20) alkyl) - (C5-C50) heteroarylene-C (O) -N ( (C1-C20) alkyl) - (C5-
C50) heteroarylene-, -C (O) -N ( (C1-C20) alkyl) - (C5-C50) heteroarylene-C (O) -N ( (C1-C20) alkyl) -, - (C5-C50) heteroarylene-C (O) -NH- (C5-C50) heteroarylene-C (O) -NH- (C6-C50) arylene-, - (C5-C50) heteroarylene-C (O) -NH- (C5-C50) heteroarylene-C (O) -NH-, - (C5-C50) heteroarylene -P (O) 2-O-, - (C5-C50) heteroarylene-O-P (O) 2-O-; wherein each of R8′ and R9′ is either absent or is a substituent independently selected from the group consisting of -H, hydroxyl, - (C1-C30) alkyl, - (C3-C50) cycloalkyl, - (C6-C50) aryl, - (C1-C30) alkylene-OH, - (C3-C50) cycloalkylene-OH, - (C6-C50) arylene-OH, - (C1-C30) alkylene-C (O) OH, - (C3-C50) cycloalkylene-C (O) OH, - (C6-C50) arylene-C (O) OH, - (C1-C30) alkylene-NH2, - (C3-C50) cycloalkylene-NH2, - (C6-C50) arylene-NH2, - (C1-C30) alkoxy, - (C3-C50) cycloalkoxy, - (C6-C50) aryloxy, -C (O) - (C1-C30) alkyl, -OC (O) (C1-C30) alkyl, -C (O) -O- (C1-C30) alkyl, -C (O) - (C3-C50) cycloalkyl, -OC (O) - (C3-C50) cycloalkyl, -C (O) -O- (C3-C50) cycloalkyl, -C (O) - (C6-C50) aryloxy, -OC (O) - (C6-C50) aryloxy, -C (O) -O- (C6-C50) aryloxy, -C (O) -NH- (C1-C30) alkyl, -C (O) -NH- (C3-C50) cycloalkyl, -C (O) -NH- (C6-C50) aryl, - (C1-C30) alkylene-phosphoric acid, - (C3-C50) cycloalkylene-phosphoric acid, - (C6-C50) arylene-phosporic acid; wherein one or more hydroxyl group, carboxyl group, amino group and phosporic acid group contained in each of R8′ and R9′ are optionally protected with a terminal protective group; or R8′ and R9′ are linked together so that R8′, R9′, the carbon atom linked with R8′ and the Y′ atom linked with R9′ form a unsubstituted or substituted heterocyclic ring; with the proviso that R9′ is absent when Y′ is oxygen or sulfur;
each R10′ is attached to any one of P′, Q′, S′ and T′, and is independently selected from the group consisting of hydroxyl, -C (O) OH, -P (O) 2-OH, -P (O) -OH, -P (O) (S) -OH, -CN, - (C1-C30) alkylene-OH, - (C3-C50) cycloalkylene-OH, - (C6-C50) arylene-OH, - (C5-C50) heteroarylene-OH, - (C1-C30) alkylene-C (O) OH, - (C3-C50) cycloalkylene-C (O) OH, - (C6-C50) arylene-C (O) OH, - (C5-C50) heteroarylene- C (O) OH, -C (O) -NH- (C1-C30) alkylene-OH, -C (O) -NH- (C3-C50) cycloalkylene-OH, -C (O) -NH- (C6-C50) arylene-OH, -C (O) -NH- (C5-C50) heteroarylene-OH, -C (O) -NH- (C1-C30) alkylene-C (O) OH, -C (O) -NH- (C3-C50) cycloalkylene-C (O) OH, -C (O) -NH- (C6-C50) arylene-C (O) OH, -C (O) -NH- (C5-C50) heteroarylene-C (O) OH, - (C1-C30) alkylene-C (O) -NH- (C1-C30) alkylene-C (O) OH, - (C1-C30) alkylene-O-C (O) - (C1-C30) alkylene-C (O) NH2, - (C1-C30) alkylene-O-C (O) - (C1-C30) alkylene-C (O) OH, - (C1-C30) alkylene-O-C (O) - (C1-C30) alkylene-NH2, - (C1-C30) alkylene-O-C (O) - (C1-C30) alkylene-OH, - (C1-C30) alkylene-C (O) -NH- (C3-C50) cycloalkylene-C (O) OH, - (C1-C30) alkylene-C (O) -NH- (C6-C50) arylene-C (O) OH, - (C1-C30) alkylene-C (O) -NH- (C5-C50) heteroarylene-C (O) OH, - (C1-C30) alkylene-P (O) 2-OH, - (C3-
C50) cycloalkylene-P (O) 2-OH, - (C6-C50) arylene-P (O) 2-OH, - (C5-C50) heteroarylene-P (O) 2-OH, -C (O) -NH- (C1-C30) alkylene-O-P (-N (C1-C16alkyl) 2) -O- (C1-C30) alkylene-CN, -C (O) -NH- (C1-C30) alkylene-O-P (-N (C1-C16alkyl) 2) -O- (C1-C30) alkylene-OH, -C (O) -NH- (C1-C30) alkylene-O-P (-N (C1-C16alkyl) 2) -O- (C1-C30) alkylene-C (O) OH, -C (O) -NH- (C1-C30) alkylene-O-P (-N (C1-C16alkyl) 2) -O- (C1-C30) alkylene-NH2, - (C1-C30) alkylene-C (O) -NH- (C1-C30) alkylene-O-P (-N (C1-C16alkyl) 2) -O- (C1-C30) alkylene-CN, - (C1-C30) alkylene-C (O) -NH- (C1-C30) alkylene-O-P (-N (C1-C16alkyl) 2) -O- (C1-C30) alkylene-OH, - (C1-C30) alkylene-C (O) -NH- (C1-C30) alkylene-O-P (-N (C1-C16alkyl) 2) -O- (C1-C30) alkylene-C (O) OH, - (C1-C30) alkylene-C (O) -NH- (C1-C30) alkylene-O-P (-N (C1-C16alkyl) 2) -O- (C1-C30) alkylene-NH2, - (C1-C30) alkylene -P (O) -OH, - (C3-C50) cycloalkylene-P (O) -OH, - (C6-C50) arylene-P (O) -OH, - (C5-C50) -heteroarylene-P (O) -OH, - (C1-C30) alkylene-P (O) (S) -OH, - (C3-C50) cycloalkylene-P (O) (S) -OH, - (C6-C50) arylene-P (O) (S) -OH, - (C5-C50) heteroarylene-P (O) (S) -OH, - (C1-C30) alkylene-CN, - (C3-C50) cycloalkylene-CN, - (C6-C50) arylene-CN, - (C5-C50) heteroarylene-CN, wherein one or more hydroxyl group, carboxyl group, amino group, nitrile group and phosporic acid group contained in R10′ is optionally linked to a support material or protected with a terminal protective group;
wherein each R11′ is attached to any one of P′, Q′, S′ and T′, and is either absent or selected from the group consisting of hydrogen, halogen atom, hydroxyl, (C1-C20) alkyl, (C1-C20) alkoxy, (C1-C20) alkoxycarbonyl, halogenated (C1-C20) alkyl and halogenated (C1-C20) alkoxycarbonyl; and
wherein M′ is an integer of 1, 2 or 3, N′ is an integer of 1, 2 or 3, and M′+N′=4.
In another specific embodiment, one or more hydroxyl group, carboxyl group, amino group, nitrile group and phosphoric acid group contained in each of A1, A2, A3, B, C, R1, R2, R3, R4, R5, R6, R1′, R2′, R3′, R4′, R5′, R6′, R7′, R8′, R9′, R10′ and R11′ are optionally protected with a terminal protection group RP selected from the group consisting of (C1-C22) alkyl, (C1-C22) alkoxy, (C1-C22) alkylcarbonyl, (C1-C22) alkoxycarbonyl, (C6-C22) aryl, (C6-C22) aryloxy, (C6-C22) arylcarbonyl, (C6-C22) aryloxycarbonyl, glucosyl, acetamide glucosyl, galactosamine, N-acetyl galactosamine, tri ( (C1-C22) alkyl) silyl and tri ( (C1-C22) alkoxy) silyl; and
wherein the support material is selected from the group consisting of silica, silica gel, glass, ceramic, polymer, cellulose, and combinations thereof.
In another specific embodiment, the oligonucleotide delivery enhancing compound has a structure represented by any of Formula AIV to Formula AXIII and Formula BIV to BXIV,
wherein A1, A2, A3, A4, F, G, H, I, B, C, P, Q, S, T, R6, R7, m, n and M are as defined herein,
wherein each of RING I and RING II is a 4, 5, 6, 7, 8 or 9 member ring;
wherein A4′ is attached to any atom of RING I, and each of A4′, A5′ and A6′ is independently selected from the group consisting of -R1′, -O-R1′, -S-R1′, -C (O) -R1′, -C (O) O-R1′, -O-C (O) -R1′, -C (O) NH-R1′, -C (O) NR2′-R1′, -NH-C (O) -R1′, -NR2′-C (O) -R1′, -O-P (O) 2-O-R1′, -OP (O) (S) -O-R1′, -O-P (O) -O-R1′, -NH-R1′, -NR2′-R1′, - (CH2) r′-NH-R1′, - (CH2) r′-NR2′-R1′, -C (O) - (CH2) r′-R1′, -C (O) - (CH2) r′-NH-R1′, -C (O) - (CH2) r′-NR2′-R1′, -C (O) - (CH2) r′-C (O) -R1′, -C (O) - (CH2) r′-C (O) O-R1′, -C (O) - (CH2) r′-NH-C (O) -R1′, -C (O) - (CH2) r′-NR2′-C (O) -R1′, - (CH2) r′-C (O) -R1′; - (CH2) r′-C (O) O-R1′; - (CH2) r′-O-C (O) -R1′, - (CH2) r′-R1′, - (CH2) r′-NH-C (O) -R1′, - (CH2) r′-NH-C (O) - (CH2) s′-R1′, - (CH2) r′-NR2′-C (O) - (CH2) s′-R1′, -CH (- (CH2) r′-NH-C (O) - (CH2) s′-R1′) (-NH-C (O) - (CH2) q′-R3′) , -N (- (CH2) r′-NH-C (O) - (CH2) s′-R1′) (-NH-C (O) - (CH2) q′-R3′) , -CH (- (CH2) r′-NH-C (O) - (CH2) s′-R1′) (- (CH2) p′-NH-C (O) - (CH2) q′-R3′) , -CR4′ (- (CH2) r′-NH-C (O) - (CH2) s′-R1′) (-NH-C (O) - (CH2) q′-R3′) , -CR4′ (- (CH2) r′-NH-C (O) - (CH2) s′-R1′) (- (CH2) p′-NH-C (O) - (CH2) q′-R3′) , -CH (- (CH2) r′-NR5′-C (O) - (CH2) s′-R1′) (-NR6′-C (O) - (CH2) q′-R3′) , -CH (- (CH2) r′-NR5′-C (O) - (CH2) s′-R1′) (- (CH2) p′-NR6′-C (O) - (CH2) q′-R3′) , -CR4′ ( (CH2) r′-NR5′-C (O) - (CH2) s′-R1′) ( (CH2) p′-NR6′-C (O) - (CH2) q′-R3′) , -CR4′ ( (CH2) r′-NR5′-C (O) - (CH2) s′-R1′) ( (CH2) p′-NR6′-C (O) - (CH2) q′-R3′) ; wherein each of R1′ and R3′ is independently selected from the group consisting of hydrogen, hydroxyl, - (C1-C30) alkyl, - (C3-C50) cycloalkyl, - (C6-C50) aryl, - (C1-
C30) alkoxy, - (C3-C50) cycloalkoxy, - (C6-C50) aryloxy, -C (O) - (C1-C30) alkyl, -OC (O) (C1-C30) alkyl, -C (O) -O- (C1-C30) alkyl, -C (O) - (C3-C50) cycloalkyl, -OC (O) - (C3-C50) cycloalkyl, -C (O) -O- (C3-C50) cycloalkyl, -C (O) - (C6-C50) aryloxy, -OC (O) - (C6-C50) aryloxy, -C (O) -O- (C6-C50) aryloxy, -C (O) -phosphate ester group, phosphodiester group, phosphoramidite group, saturated fatty acid group, unsaturated fatty acid group, lipid, PEG, steroid, lipophile, carbohydrate, cholesterol, adamantane, amino acid, peptide, ligand, nucleic acid, oligonucleotide, aptamer, small molecule, antibody, antibody fragment, polyethylene glycol, carbohydrate, antibody, antibody fragment, chloroquine, alkaloid and targeting moiety, wherein one or more hydroxyl group, carboxyl group and amino group contained in each of R1′ and R3′ are optionally protected; wherein each of R2′, R4′, R5′ and R6′ is independently a halogen atom, a (C1-C12) alkyl, a (C1-C12) alkoxy, a (C6-C16) aryl or a (C6-C16) aryloxy; wherein each of r′, s′, p′ and q′ is an integer from 1 to 22; and
wherein R12′ is attached to any atom of RING II and is selected from the group consisting of -H, hydroxyl, - (C1-C30) alkyl, - (C3-C50) cycloalkyl, - (C6-C50) aryl, - (C1-C30) alkylene-OH, - (C3-C50) cycloalkylene-OH, - (C6-C50) arylene-OH, - (C1-C30) alkylene-C (O) OH, - (C3-C50) cycloalkylene-C (O) OH, - (C6-C50) arylene-C (O) OH, - (C1-C30) alkylene-NH2, - (C3-C50) cycloalkylene-NH2, - (C6-C50) arylene-NH2, - (C1-C30) alkoxy, - (C3-C50) cycloalkoxy, - (C6-C50) aryloxy, -C (O) - (C1-C30) alkyl, -OC (O) (C1-C30) alkyl, -C (O) -O- (C1-C30) alkyl, -C (O) - (C3-C50) cycloalkyl, -OC (O) - (C3-C50) cycloalkyl, -C (O) -O- (C3-C50) cycloalkyl, -C (O) - (C6-C50) aryloxy, -OC (O) - (C6-C50) aryloxy, -C (O) -O- (C6-C50) aryloxy, -C (O) -NH- (C1-C30) alkyl, -C (O) -NH- (C3-C50) cycloalkyl, -C (O) -NH- (C6-C50) aryl, - (C1-C30) alkylene-phosporic acid, - (C3-C50) cycloalkylene-phosporic acid, - (C6-C50) arylene-phosporic acid; wherein one or more hydroxyl group, carboxyl group, amino group and phosporic acid group contained in R12′ are optionally protected.
In another specific embodiment, each of F, G, H and I is carbon, m is 1 and n is 3, B is attached to G or H, each of P, Q, S and T is carbon, R6 is attached to any one of Q and S;
wherein the protection group RP is selected from the group consisting of benzyloxycarbonyl (Cbz) , tert-butyldimethylsilyl (TBS) , 4, 4′-dimethoxytrityl (DMTr) , t-butyloxy carbonyl (Boc) , benzyl (Bn) and benzyloxy (BnO) ;
wherein C, on each occurrence, is selected from the group consisting of hydrogen, halogen atom, hydroxyl, (C1-C12) alkyl, (C1-C12) alkoxy, halogenated (C1-C12) alkyl and
halogenated (C1-C12) alkoxy;
wherein B, on each occurrence, is selected from the group consisting of - (C1-C22) alkylene-OH, -O-C (O) - (C1-C16) alkylene-C (O) NH2, - (C1-C16) alkylene-O-C (O) - (C1-C16) alkylene-C (O) NH2, -O-C (O) - (C1-C16) alkylene-C (O) OH, - (C1-C16) alkylene-O-C (O) - (C1-C16) alkylene-C (O) OH, -C (O) - (C1-C16) alkylene-C (O) NH2, - (C1-C16) alkylene-C (O) - (C1-C16) alkylene-C (O) NH2, -C (O) -NH- (C1-C16) alkylene-OH, -C (O) -NH- (C1-C16) alkylene-C (O) OH, - (C1-C16) alkylene-C (O) -NH- (C1-C16) alkylene-C (O) OH, - (C1-C16) alkylene-O-P (-N (C1-C16 alkyl) 2) -O- (C1-C16) alkylene-CN, - (C1-C16) alkylene-O-P (-N (C1-C16 alkyl) 2) -O- (C1-C16) alkylene-OH, - (C1-C16) alkylene-O-P (-N (C1-C16 alkyl) 2) -O- (C1-C16) alkylene-NH2, - (C1-C16) alkylene-O-P (-N (C1-C16 alkyl) 2) -O- (C1-C16) alkylene-C (O) OH, and -C (O) -NH- (C1-C16) alkylene-O-P (-N (C1-C16 alkyl) 2) -O- (C1-C16) alkylene-CN,
wherein each of A1, A2 and A3 is either absent or a substituent independently selected from the group consisting of -H, -OH, linear or branched - (C6-C22) alkyl, linear or branched - (C2-C22) alkenyl, - (C1-C22) alkylene-OH, - (C3-C22) cycloalkyl, - (C3-C22) cycloalkenyl, - (C1-C22) alkylene- (C3-C22) cycloalkyl, - (C1-C22) alkylene-R1, - (C1-C22) alkylene-O-R1, - (C1-C22) alkylene-COOR1, -O- (C1-C22) alkyl, - (C6-C22) alkylene-adamantyl, - (C1-C22) alkylene-NH-C (O) - (C1-C22) alkyl, - (C1-C22) alkylene-NH-C (O) - (C1-C22) alkylene-adamantyl, - (C1-C22) alkylene-NR2-C (O) - (C1-C22) alkylene-adamantyl, - (C1-C22) alkylene- (C1-C6 alkylene oxide) (1-20) -NH-C (O) - (C1-C22) alkylene-adamantyl, -C (O) NH-R1, - (C1-C22) alkylene-O-P (-N (C1-C22 alkyl) 2) -O- (C1-C22) alkylene-CN, - (C1-C22) alkylene-O-P (-N (C1-C22 alkyl) 2) -O- (C1-C22) alkylene-OH, - (C1-C22) alkylene-O-P (-N (C1-C22 alkyl) 2) -O- (C1-C22) alkylene-NH2, - (C1-C22) alkylene-O-P (-N (C1-C22 alkyl) 2) -O- (C1-C22) alkylene-C (O) OH, -C (O) -NH- (C1-C22) alkylene-O-P (-N (C1-C22 alkyl) 2) -O- (C1-C22) alkylene-CN, substituted or unsubstituted pyrrole, substituted or unsubstituted pyrroline, substituted or unsubstituted pyrrolidine, substituted or unsubstituted pyrazole, substituted or unsubstituted pyrazoline, substituted or unsubstituted pyrazolidine, substituted or unsubstituted imidazole, substituted or unsubstituted oxazole, substituted or unsubstituted thiazole, substituted or unsubstituted benzopyrrole, substituted or unsubstituted benzopyrroline, substituted or unsubstituted benzopyrrolidine, substituted or unsubstituted benzopyrazole, substituted or unsubstituted benzopyrazoline, substituted or unsubstituted benzopyrazolidine, substituted or unsubstituted benzoimidazole, substituted or unsubstituted benzooxazole, substituted or unsubstituted benzothiazole, and a
substituent represented by Formula AIII,
wherein Y is selected from the group consisting of carbon, nitrogen, oxygen and sulfur; and each of P, Q, S and T is carbon;
wherein each of R3, R4 and R5 is either absent or a substituent independently selected from the group consisting of -H, -OH, linear or branched - (C6-C22) alkyl, linear or branched - (C2-C22) alkenyl, - (C1-C22) alkylene-OH, - (C3-C22) cycloalkyl, - (C3-C22) cycloalkenyl, - (C1-C22) alkylene- (C3-C22) cycloalkyl, - (C1-C22) alkylene-R1, - (C1-C22) alkylene-O-R1, - (C1-C22) alkylene-COOR1, -O- (C1-C22) alkyl, - (C6-C22) alkylene-adamantyl, - (C1-C22) alkylene-NH-C (O) - (C1-C22) alkyl, - (C1-C22) alkylene-NH-C (O) - (C1-C22) alkylene-adamantyl, - (C1-C22) alkylene-NR2-C (O) - (C1-C22) alkylene-adamantyl, - (C1-C22) alkylene- (C1-C6 alkylene oxide) (1-20) -NH-C (O) - (C1-C22) alkylene-adamantyl, -C (O) NH-R1, - (C1-C22) alkylene-O-P (-N (C1-C22 alkyl) 2) -O- (C1-C22) alkylene-CN, - (C1-C22) alkylene-O-P (-N (C1-C22 alkyl) 2) -O- (C1-C22) alkylene-OH, - (C1-C22) alkylene-O-P (-N (C1-C22 alkyl) 2) -O- (C1-C22) alkylene-NH2, - (C1-C22) alkylene-O-P (-N (C1-C22 alkyl) 2) -O- (C1-C22) alkylene-C (O) OH, -C (O) -NH- (C1-C22) alkylene-O-P (-N (C1-C22 alkyl) 2) -O- (C1-C22) alkylene-CN, substituted or unsubstituted pyrrole, substituted or unsubstituted pyrroline, substituted or unsubstituted pyrrolidine, substituted or unsubstituted pyrazole, substituted or unsubstituted pyrazoline, substituted or unsubstituted pyrazolidine, substituted or unsubstituted imidazole, substituted or unsubstituted oxazole, substituted or unsubstituted thiazole, substituted or unsubstituted benzopyrrole, substituted or unsubstituted benzopyrroline, substituted or unsubstituted benzopyrrolidine, substituted or unsubstituted benzopyrazole, substituted or unsubstituted benzopyrazoline, substituted or unsubstituted benzopyrazolidine, substituted or unsubstituted benzoimidazole, substituted or unsubstituted benzooxazole, and substituted or unsubstituted benzothiazole,
wherein R7, on each occurrence, is attached to any one of P, Q, S and T, and is either absent or selected from the group consisting of hydrogen, halogen atom, hydroxyl, (C1-C20) alkyl, (C1-C20) alkoxy, halogenated (C1-C20) alkyl and halogenated (C1-C20) alkoxy;
wherein M is an integer of 0, 1, 2 or 3;
wherein R6 is attached to any one of P, Q, S and T, and is selected from the group consisting of - (C1-C16) alkylene-, - (C1-C16) alkylene-O-, -O- (C1-C16) alkylene-, - (C1-C16) alkylene-NH-, -NH- (C1-C16) alkylene-, -C (O) - (C1-C16) alkylene-, - (C1-C16) alkylene-C (O) -, -C (O) -O- (C1-C16) alkylene-, - (C1-C16) alkylene-C (O) -O-, -C (O) -NH- (C1-C16) alkylene-, -C (O) -
NH- (C1-C16) alkylene-C (O) -O-, -C (O) -NH- (C1-C16) alkylene-O-C (O) -, -C (O) -NH- (C1-C16) alkylene-O-C (O) -O-, -C (O) -NH- (C1-C16) alkylene-O-, -C (O) -N ( (C1-C16) alkyl) - (C1-C16) alkylene-, -C (O) -N ( (C1-C16) alkyl) - (C1-C16) alkylene-O-, -C (O) -NH- (C1-C16) alkylene-C (O) -NH- (C1-C16) alkylene-, -C (O) -NH- (C1-C16) alkylene-NH-C (O) - (C1-C16) alkylene-, -C (O) -NH- (C1-C16) alkylene-C (O) -NH-, -C (O) -N ( (C1-C16) alkyl) - (C1-C16) alkylene-C (O) -N ( (C1-C16) alkyl) - (C1-C16) alkylene-, -C (O) -N ( (C1-C16) alkyl) - (C1-C16) alkylene-C (O) -N ( (C1-C16) alkyl) -, - (C1-C16) alkylene-C (O) -NH- (C1-C16) alkylene-C (O) -NH- (C1-C16) alkylene-, - (C1-C16) alkylene-C (O) -NH- (C1-C16) alkylene-C (O) -NH-, -NH-C (O) - (C1-C16) alkylene-, -NH-C (O) - (C1-C16) alkylene-C (O) -O-, -NH-C (O) - (C1-C16) alkylene-C (O) -, -NH-C (O) - (C1-C16) alkylene-O-, -N ( (C1-C16) alkyl) -C (O) - (C1-C16) alkylene-, -N ( (C1-C16) alkyl) -C (O) - (C1-C16) alkylene-O-, -NH-C (O) - (C1-C16) alkylene-NH-C (O) - (C1-C16) alkylene-, -NH-C (O) - (C1-C16) alkylene-NH-C (O) -, -N ( (C1-C16) alkyl) -C (O) - (C1-C16) alkylene-N ( (C1-C16) alkyl) -C (O) - (C1-C16) alkylene-, -N ( (C1-C16) alkyl) -C (O) - (C1-C16) alkylene-N ( (C1-C16) alkyl) -C (O) -, - (C1-C16) alkylene-NH-C (O) - (C1-C16) alkylene-NH-C (O) - (C1-C16) alkylene-, - (C1-C16) alkylene-NH-C (O) - (C1-C16) alkylene-NH-C (O) -.
In another specific embodiment, the oligonucleotide delivery enhancing compound has a structure represented by any one of Formula BXV to Formula BXXIX,
wherein each of A1′ and A2′ is a substituent independently selected from the group consisting of -R1′, -O-R1′, -S-R1′, -C (O) -R1′, -C (O) O-R1′, -O-C (O) -R1′, -C (O) NH-R1′, -C (O) NR2′-R1′, -NH-C (O) -R1′, -NR2′-C (O) -R1′, -O-P (O) 2-O-R1′, -OP (O) (S) -O-R1′, -O-P (O) -O-R1′, -NH-R1′, -NR2′-R1′, - (CH2) r′-NH-R1′, - (CH2) r′-NR2′-R1′, -C (O) - (CH2) r′-R1′, -C (O) - (CH2) r′-NH-R1′, -C (O) - (CH2) r′-NR2′-R1′, -C (O) - (CH2) r′-C (O) -R1′, -C (O) - (CH2) r′-C (O) O-R1′, -C (O) - (CH2) r′-NH-C (O) -R1′, -C (O) - (CH2) r′-NR2′-C (O) -R1′, - (CH2) r′-C (O) -R1′; - (CH2) r′-C (O) O-R1′; - (CH2) r′-O-C (O) -R1′, - (CH2) r′-R1′, - (CH2) r′-NH-C (O) -R1′, - (CH2) r′-NH-C (O) - (CH2) s′-R1′, - (CH2) r′-NH-C (O) - (C1-C22) alkylene-NH-C (O) - (CH2) s′-R1′, - (CH2) r′-C (O) -NH- (CH2) s′-R1′, - (CH2) r′-C (O) - NH- (C1-C22) alkylene-C (O) -NH- (CH2) s′-R1′, - (CH2) r′-C (O) -NH- (C1-C22) alkylene-NH-C (O) - (CH2) s′-R1′, - (CH2) r′-NR2′-C (O) - (CH2) s′-R1′, -CH (- (CH2) r′-NH-C (O) - (CH2) s′-R1′) (-NH-C (O) - (CH2) q′-R3′) , -CH (- (CH2) r′-C (O) -NH- (CH2) s′-R1′) (-C (O) -NH- (CH2) q′-R3′) , -N (- (CH2) r′-NH-C (O) - (CH2) s′-R1′) (-NH-C (O) - (CH2) q′-R3′) , -CH (- (CH2) r′-NH-C (O) - (CH2) s′-R1′) (- (CH2) p′-NH-C (O) - (CH2) q′-R3′) , -CR4′ (- (CH2) r′-NH-C (O) - (CH2) s′-R1′) (-NH-C (O) - (CH2) q′-R3′) , -CR4′ (- (CH2) r′-NH-C (O) - (CH2) s′-R1′) (- (CH2) p′-NH-C (O) - (CH2) q′-R3′) , -CH (- (CH2) r′-NR5′-C (O) - (CH2) s′-R1′) (-NR6′-C (O) - (CH2) q′-R3′) , -CH (- (CH2) r′-NR5′-C (O) - (CH2) s′-
R1′) (- (CH2) p′-NR6′-C (O) - (CH2) q′-R3′) , -CR4′ ( (CH2) r′-NR5′-C (O) - (CH2) s′-R1′) ( (CH2) p′-NR6′-C (O) - (CH2) q′-R3′) , and -CR4′ ( (CH2) r′-NR5′-C (O) - (CH2) s′-R1′) ( (CH2) p′-NR6′-C (O) - (CH2) q′-R3′) ; wherein each of R1′ and R3′ is independently selected from the group consisting of hydrogen, hydroxyl, - (C1-C30) alkyl, - (C3-C50) cycloalkyl, - (C6-C50) aryl, - (C1-C30) alkoxy, - (C3-C50) cycloalkoxy, - (C6-C50) aryloxy, -C (O) - (C1-C30) alkyl, -OC (O) (C1-C30) alkyl, -C (O) -O- (C1-C30) alkyl, -C (O) - (C3-C50) cycloalkyl, -OC (O) - (C3-C50) cycloalkyl, -C (O) -O- (C3-C50) cycloalkyl, -C (O) - (C6-C50) aryloxy, -OC (O) - (C6-C50) aryloxy, -C (O) -O- (C6-C50) aryloxy, -C (O) -phosphate ester group, phosphodiester group, phosphoramidite group, saturated fatty acid group, unsaturated fatty acid group, lipid, PEG, steroid, lipophile, carbohydrate, cholesterol, adamantane, amino acid, peptide, ligand, nucleic acid, oligonucleotide, aptamer, small molecule, antibody, antibody fragment, polyethylene glycol, antibody, antibody fragment, chloroquine, alkaloid and targeting moiety, wherein one or more hydroxyl group, carboxyl group and amino group contained in each of R1′ and R3′ are optionally protected; wherein each of R2′, R4′, R5′ and R6′ is independently a halogen atom, a (C1-C12) alkyl, a (C1-C12) alkoxy, a (C1-C12) alkoxycarbonyl, a (C6-C16) aryl or a (C6-C16) aryloxycarbonyl; wherein each of r′, s′, p′ and q′ is an integer from 1 to 22;
wherein m′ is an integer of 1, 2 or 3, n′ is an integer of 1, 2 or 3, and m′+n′=4;
wherein each B′ is independently selected from the group consisting of hydroxyl, -C (O) OH, - (C1-C30) alkoxy, -P (O) 2-OH, -P (O) -OH, -P (O) (S) -OH, -CN, - (C1-C30) alkylene-OH, - (C3-C50) cycloalkylene-OH, - (C6-C50) arylene-OH, - (C5-C50) heteroarylene-OH, - (C1-C30) alkylene-C (O) OH, - (C3-C50) cycloalkylene-C (O) OH, - (C6-C50) arylene-C (O) OH, - (C5-C50) heteroarylene-C (O) OH, -C (O) -NH- (C1-C30) alkylene-OH, -C (O) -NH- [ (C1-C30) alkylene-O] r′-H (wherein r′ is an integer of 1 to 22) , -C (O) -NH- [ (C1-C30) alkylene-O] r′- (C1-C30) alkylene-C (O) -OH (wherein r′ is an integer of 1 to 22) , -C (O) -NH- (C3-C50) cycloalkylene-OH, -C (O) -NH- (C6-C50) arylene-OH, -C (O) -NH- (C5-C50) heteroarylene-OH, -C (O) -NH- (C1-C30) alkylene-C (O) OH, -C (O) -NH- (C3-C50) cycloalkylene-C (O) OH, -C (O) -NH- (C6-C50) arylene-C (O) OH, -C (O) -NH- (C1-C30) alkylene- (C6-C50) arylene- (C1-C30) alkylene-C (O) OH, -C (O) -NH- (C5-C50) heteroarylene-C (O) OH, - (C1-C30) alkylene-C (O) -NH- (C1-C30) alkylene-C (O) OH, - (C1-C30) alkylene-C (O) -NH- (C3-C50) cycloalkylene-C (O) OH, - (C1-C30) alkylene-C (O) -NH- (C6-C50) arylene-C (O) OH, - (C1-C30) alkylene-C (O) -NH- (C5-C50) heteroarylene-C (O) OH, - (C1-
C30) alkylene-P (O) 2-OH, - (C3-C50) cycloalkylene-P (O) 2-OH, - (C6-C50) arylene-P (O) 2-OH, - (C5-C50) heteroarylene-P (O) 2-OH, -C (O) -NH- (C1-C30) alkylene-O-P (-N (C1-C16alkyl) 2) -O- (C1-C30) alkylene-CN, -C (O) -NH- (C1-C30) alkylene-O-P (-N (C1-C16alkyl) 2) -O- (C1-C30) alkylene-OH, -C (O) -NH- (C1-C30) alkylene-O-P (-N (C1-C16alkyl) 2) -O- (C1-C30) alkylene-C (O) OH, -C (O) -NH- (C1-C30) alkylene-O-P (-N (C1-C16alkyl) 2) -O- (C1-C30) alkylene-NH2, - (C1-C30) alkylene-C (O) -NH- (C1-C30) alkylene-O-P (-N (C1-C16alkyl) 2) -O- (C1-C30) alkylene-CN, - (C1-C30) alkylene-C (O) -NH- (C1-C30) alkylene-O-P (-N (C1-C16alkyl) 2) -O- (C1-C30) alkylene-OH, - (C1-C30) alkylene-C (O) -NH- (C1-C30) alkylene-O-P (-N (C1-C16alkyl) 2) -O- (C1-C30) alkylene-C (O) OH, - (C1-C30) alkylene-C (O) -NH- (C1-C30) alkylene-O-P (-N (C1-C16alkyl) 2) -O- (C1-C30) alkylene-NH2, - (C1-C30) alkylene -P (O) -OH, - (C3-C50) cycloalkylene-P (O) -OH, - (C6-C50) arylene-P (O) -OH, - (C5-C50) -heteroarylene-P (O) -OH, - (C1-C30) alkylene-P (O) (S) -OH, - (C3-C50) cycloalkylene-P (O) (S) -OH, - (C6-C50) arylene-P (O) (S) -OH, - (C5-C50) heteroarylene-P (O) (S) -OH, - (C1-C30) alkylene-CN, - (C3-C50) cycloalkylene-CN, - (C6-C50) arylene-CN, - (C5-C50) heteroarylene-CN, lipid, PEG, steroid, lipophile, carbohydrate, cholesterol, adamantane, amino acid, peptide, chloroquine and alkaloid;
where each R7′ is selected from the group consisting of -O-, -C (O) O-, -O-C (O) -, -P (O) 2-O-, -O-P (O) 2-O-, -P (O) (S) -O-, -O-P (O) (S) -O-, -O-P (O) -O-, - (C1-C30) alkylene-, - (C1-C30) alkylene-O-, -O- (C1-C30) alkylene-, - (C1-C30) alkylene-NH-, -NH- (C1-C30) alkylene-, -C (O) - (C1-C30) alkylene-, - (C1-C30) alkylene-C (O) -, -C (O) -O- (C1-C30) alkylene-, - (C1-C30) alkylene-C (O) -O-, -C (O) -NH- (C1-C30) alkylene-, -C (O) -NH- (C1-C30) alkylene-C (O) -O-, -C (O) -NH- (C1-C30) alkylene-O-C (O) -, -C (O) -NH- (C1-C30) alkylene-O-C (O) -O-, -C (O) -NH- (C1-C30) alkylene-O-, -C (O) -N ( (C1-C20) alkyl) - (C1-C30) alkylene-, -C (O) -N ( (C1-C20) alkyl) - (C1-C30) alkylene-O-, -C (O) -NH- (C1-C30) alkylene-C (O) -NH- (C1-C30) alkylene-, -C (O) -NH- (C1-C30) alkylene-NH-C (O) - (C1-C30) alkylene-, -C (O) -NH- (C1-C30) alkylene-C (O) -NH-, -C (O) -N ( (C1-C20) alkyl) - (C1-C30) alkylene-C (O) -N ( (C1-C20) alkyl) - (C1-C30) alkylene-, -C (O) -N ( (C1-C20) alkyl) - (C1-C30) alkylene-C (O) -N ( (C1-C20) alkyl) -, - (C1-C30) alkylene-C (O) -NH- (C1-C30) alkylene-C (O) -NH- (C1-C30) alkylene-, - (C1-C30) alkylene-C (O) -NH- (C1-C30) alkylene-C (O) -NH-, -NH-C (O) - (C1-C30) alkylene-, -NH-C (O) - (C1-C30) alkylene-C (O) -O-, -NH-C (O) - (C1-C30) alkylene-C (O) -, -NH-C (O) - (C1-C30) alkylene-O-, -N ( (C1-C20) alkyl) -C (O) - (C1-C30) alkylene-, -N ( (C1-C20) alkyl) -C (O) - (C1-C30) alkylene-O-, -NH-C (O) - (C1-C30) alkylene-NH-C (O) - (C1-C30) alkylene-, -NH-C (O) - (C1-C30) alkylene-NH-C (O) -, -N ( (C1-C20) alkyl) -C (O) - (C1-C30) alkylene-N ( (C1-C20) alkyl) -
C (O) - (C1-C30) alkylene-, -N ( (C1-C20) alkyl) -C (O) - (C1-C30) alkylene-N ( (C1-C20) alkyl) -C (O) -, - (C1-C30) alkylene-NH-C (O) - (C1-C30) alkylene-NH-C (O) - (C1-C30) alkylene-, - (C1-C30) alkylene-NH-C (O) - (C1-C30) alkylene-NH-C (O) -, - (C1-C30) alkylene -P (O) 2-O-, - (C1-C30) alkylene-O-P (O) 2-O-, - (C3-C50) cycloalkylene-, - (C3-C50) cycloalkylene-O-, -O- (C3-C50) cycloalkylene-, - (C3-C50) cycloalkylene-NH-, -NH- (C3-C50) cycloalkylene-, -C (O) - (C3-C50) cycloalkylene-, - (C3-C50) cycloalkylene-C (O) -, -C (O) -O- (C3-C50) cycloalkylene-, - (C3-C50) cycloalkylene-C (O) -O-, -C (O) -NH- (C3-C50) cycloalkylene-, -C (O) -NH- (C3-C50) cycloalkylene-C (O) -O-, -C (O) -NH- (C3-C50) cycloalkylene-O-, -C (O) -N ( (C1-C20) alkyl) - (C3-C50) cycloalkylene-, -C (O) -N ( (C1-C20) alkyl) - (C3-C50) cycloalkylene-O-, -C (O) -NH- (C3-C50) cycloalkylene-C (O) -NH- (C3-C50) cycloalkylene-, -C (O) -NH- (C3-C50) cycloalkylene-C (O) -NH-, -C (O) -N ( (C1-C20) alkyl) - (C3-C50) cycloalkylene-C (O) -N ( (C1-C20) alkyl) - (C3-C50) cycloalkylene-, -C (O) -N ( (C1-C20) alkyl) - (C3-C50) cycloalkylene-C (O) -N ( (C1-C20) alkyl) -, - (C3-C50) cycloalkylene-C (O) -NH- (C3-C50) cycloalkylene-C (O) -NH- (C3-C50) cycloalkylene-, - (C3-C50) cycloalkylene-C (O) -NH- (C3-C50) cycloalkylene-C (O) -NH-, - (C3-C50) cycloalkylene -P (O) 2-O-, - (C3-C50) cycloalkylene-O-P (O) 2-O-, - (C6-C50) arylene-, - (C6-C50) arylene-O-, -O- (C6-C50) arylene-, - (C6-C50) arylene-NH-, -NH- (C6-C50) arylene-, -C (O) - (C6-C50) arylene-, - (C6-C50) arylene-C (O) -, -C (O) -O- (C6-C50) arylene-, - (C6-C50) arylene-C (O) -O-, -C (O) -NH- (C6-C50) arylene-, -C (O) -NH- (C6-C50) arylene-C (O) -O-, -C (O) -NH- (C6-C50) arylene-O-, -C (O) -N ( (C1-C20) alkyl) - (C6-C50) arylene-, -C (O) -N ( (C1-C20) alkyl) - (C6-C50) arylene-O-, -C (O) -NH- (C6-C50) arylene-C (O) -NH- (C3-C50) cycloalkylene-, -C (O) -NH- (C6-C50) arylene-C (O) -NH-, -C (O) -N ( (C1-C20) alkyl) - (C6-C50) arylene-C (O) -N ( (C1-C20) alkyl) - (C6-C50) arylene-, -C (O) -N ( (C1-C20) alkyl) - (C6-C50) arylene-C (O) -N ( (C1-C20) alkyl) -, - (C6-C50) arylene-C (O) -NH- (C6-C50) arylene-C (O) -NH- (C6-C50) arylene-, - (C6-C50) arylene-C (O) -NH- (C6-C50) arylene-C (O) -NH-, - (C6-C50) arylene -P (O) 2-O-, - (C6-C50) arylene-O-P (O) 2-O-, - (C5-C50) heteroarylene-, - (C5-C50) heteroarylene-O-, -O- (C5-C50) heteroarylene-, - (C5-C50) heteroarylene-NH-, -NH- (C5-C50) heteroarylene-, -C (O) - (C5-C50) heteroarylene-, - (C5-C50) heteroarylene-C (O) -, -C (O) -O- (C5-C50) heteroarylene-, - (C5-C50) heteroarylene-C (O) -O-, -C (O) -NH- (C5-C50) heteroarylene-, -C (O) -NH- (C5-C50) heteroarylene-C (O) -O-, -C (O) -NH- (C5-C50) heteroarylene-O-, -C (O) -N ( (C1-C20) alkyl) - (C5-C50) heteroarylene-, -C (O) -N ( (C1-C20) alkyl) - (C5-C50) heteroarylene-O-, -C (O) -NH- (C5-C50) heteroarylene-C (O) -NH- (C3-C50) cycloalkylene-, -C (O) -NH- (C5-C50) heteroarylene-C (O) -NH-, -C (O) -N ( (C1-C20) alkyl) - (C5-C50) heteroarylene-C (O) -N ( (C1-C20) alkyl) - (C5-
C50) heteroarylene-, -C (O) -N ( (C1-C20) alkyl) - (C5-C50) heteroarylene-C (O) -N ( (C1-C20) alkyl) -, - (C5-C50) heteroarylene-C (O) -NH- (C5-C50) heteroarylene-C (O) -NH- (C6-C50) arylene-, - (C5-C50) heteroarylene-C (O) -NH- (C5-C50) heteroarylene-C (O) -NH-, - (C5-C50) heteroarylene -P (O) 2-O-, - (C5-C50) heteroarylene-O-P (O) 2-O-; wherein each R8′ is a substituent independently selected from the group consisting of -H, hydroxyl, - (C1-C30) alkyl, - (C3-C50) cycloalkyl, - (C6-C50) aryl, - (C1-C30) alkylene-OH, - (C3-C50) cycloalkylene-OH, - (C6-C50) arylene-OH, - (C1-C30) alkylene-C (O) OH, - (C3-C50) cycloalkylene-C (O) OH, - (C6-C50) arylene-C (O) OH, - (C1-C30) alkylene-NH2, - (C3-C50) cycloalkylene-NH2, - (C6-C50) arylene-NH2, - (C1-C30) alkoxy, - (C3-C50) cycloalkoxy, - (C6-C50) aryloxy, -C (O) - (C1-C30) alkyl, -OC (O) (C1-C30) alkyl, -C (O) -O- (C1-C30) alkyl, -C (O) - (C3-C50) cycloalkyl, -OC (O) - (C3-C50) cycloalkyl, -C (O) -O- (C3-C50) cycloalkyl, -C (O) - (C6-C50) aryloxy, -OC (O) - (C6-C50) aryloxy, -C (O) -O- (C6-C50) aryloxy, -C (O) -NH- (C1-C30) alkyl, -C (O) -NH- (C3-C50) cycloalkyl, -C (O) -NH- (C6-C50) aryl, - (C1-C30) alkylene-phosphoric acid, - (C3-C50) cycloalkylene-phosphoric acid, - (C6-C50) arylene-phosporic acid; wherein one or more hydroxyl group, carboxyl group, amino group and phosporic acid group contained in each of R8′ are optionally protected with a terminal protective group; and
wherein each R10′ is independently selected from the group consisting of hydroxyl, -C (O) OH, -P (O) 2-OH, -P (O) -OH, -P (O) (S) -OH, -CN, - (C1-C30) alkylene-OH, - (C3-C50) cycloalkylene-OH, - (C6-C50) arylene-OH, - (C5-C50) heteroarylene-OH, - (C1-C30) alkylene-C (O) OH, - (C3-C50) cycloalkylene-C (O) OH, - (C6-C50) arylene-C (O) OH, - (C5-C50) heteroarylene-C (O) OH, -C (O) -NH- (C1-C30) alkylene-OH, -C (O) -NH- (C3-C50) cycloalkylene-OH, -C (O) -NH- (C6-C50) arylene-OH, -C (O) -NH- (C5-C50) heteroarylene-OH, -C (O) -NH- (C1-C30) alkylene-C (O) OH, -C (O) -NH- (C3-C50) cycloalkylene-C (O) OH, -C (O) -NH- (C6-C50) arylene-C (O) OH, -C (O) -NH- (C5-C50) heteroarylene-C (O) OH, - (C1-C30) alkylene-C (O) -NH- (C1-C30) alkylene-C (O) OH, - (C1-C30) alkylene-O-C (O) - (C1-C30) alkylene-C (O) NH2, - (C1-C30) alkylene-O-C (O) - (C1-C30) alkylene-C (O) OH, - (C1-C30) alkylene-O-C (O) - (C1-C30) alkylene- NH2, - (C1-C30) alkylene-O-C (O) - (C1-C30) alkylene-OH, - (C1-C30) alkylene-C (O) -NH- (C3-C50) cycloalkylene-C (O) OH, - (C1-C30) alkylene-C (O) -NH- (C6-C50) arylene-C (O) OH, - (C1-C30) alkylene-C (O) -NH- (C5-C50) heteroarylene-C (O) OH, - (C1-C30) alkylene-P (O) 2-OH, - (C3-C50) cycloalkylene-P (O) 2-OH, - (C6-C50) arylene-P (O) 2-OH, - (C5-C50) heteroarylene-P (O) 2-OH, -C (O) -NH- (C1-C30) alkylene-O-P (-N (C1-C16alkyl) 2) -O- (C1-C30) alkylene-CN, -C (O) -NH- (C1-C30) alkylene-O-P (-N (C1-C16alkyl) 2) -O- (C1-C30) alkylene-OH, -C (O) -NH- (C1-C30) alkylene-O-
P (-N (C1-C16alkyl) 2) -O- (C1-C30) alkylene-C (O) OH, -C (O) -NH- (C1-C30) alkylene-O-P (-N (C1-C16alkyl) 2) -O- (C1-C30) alkylene-NH2, - (C1-C30) alkylene-C (O) -NH- (C1-C30) alkylene-O-P (-N (C1-C16alkyl) 2) -O- (C1-C30) alkylene-CN, - (C1-C30) alkylene-C (O) -NH- (C1-C30) alkylene-O-P (-N (C1-C16alkyl) 2) -O- (C1-C30) alkylene-OH, - (C1-C30) alkylene-C (O) -NH- (C1-C30) alkylene-O-P (-N (C1-C16alkyl) 2) -O- (C1-C30) alkylene-C (O) OH, - (C1-C30) alkylene-C (O) -NH- (C1-C30) alkylene-O-P (-N (C1-C16alkyl) 2) -O- (C1-C30) alkylene-NH2, - (C1-C30) alkylene -P (O) -OH, - (C3-C50) cycloalkylene-P (O) -OH, - (C6-C50) arylene-P (O) -OH, - (C5-C50) -heteroarylene-P (O) -OH, - (C1-C30) alkylene-P (O) (S) -OH, - (C3-C50) cycloalkylene-P (O) (S) -OH, - (C6-C50) arylene-P (O) (S) -OH, - (C5-C50) heteroarylene-P (O) (S) -OH, - (C1-C30) alkylene-CN, - (C3-C50) cycloalkylene-CN, - (C6-C50) arylene-CN, - (C5-C50) heteroarylene-CN, wherein one or more hydroxyl group, carboxyl group, amino group, nitrile group and phosporic acid group contained in R10′ is optionally linked to a support material or protected with a terminal protective group.
In another specific embodiment, the oligonucleotide delivery enhancing compound has a structure of
wherein represents a support material.
In another specific embodiment, at least one hydrogen atom contained in the oligonucleotide delivery enhancing compound is substituted with deuterium atom.
In another specific embodiment, the present disclosure provides an oligonucleotide delivery agent, comprising a delivery enhancing compound (DEC) moiety derivable from the oligonucleotide delivery enhancing compound as indicated herein and at least one oligonucleotide.
In another specific embodiment, the oligonucleotide delivery enhancing compound moiety is linked with the oligonucleotide via at least one linking moiety selected from the group consisting of direct bond, -O-, -S-, -C (O) -, -NH-, -N ( (C1-C12) alkyl) -, -N ( (C1-C12) alkyl) -C (O) -O-, -O-C (O) -, -C (O) -O-, -O-C (O) -O-, -C (O) -NH-, -OP (O) 2O-, -P (O) (O-) O-, -OP (O) O-, -OP (O) (S) O-, -O-S (O) 2-O-, -S (O) 2-O-, -S (O) -O-, - (C1-C22) alkylene-, - (C1-C22) alkylene-NH-, -NH- (C1-C22) alkylene-, - (C1-C22) alkylene-NH-C (O) -, - (C1-C22) alkylene-C (O) -, - (C1-C22) alkylene-C (O) -O-, -C (O) - (C1-C22) alkylene-, -NH-C (O) - (C1-C22) alkylene-, -C (O) -NH- (C1-C22) alkylene-, -C (O) - (C1-C22) alkylene-NH-, -NH- (C1-C22) alkylene-C (O) -, -C (O) - (C1-C22) alkylene-C (O) -, -NH- (C1-C22) alkylene-NH-, -C (O) - (C1-C22) alkylene-C (O) O-, -O-C (O) - (C1-C22) alkylene-C (O) -O-, -C (O) -O- (C1-C22) alkylene-O-C (O) -, -C (O) - (C1-C22) alkylene-NH-C (O) -, -NH-C (O) - (C1-C22) alkylene-C (O) -, -NH-C (O) - (C1-C22) alkylene-C (O) -NH-, -C (O) -NH- (C1-C22) alkylene-NH-C (O) -, - (C1-C22) alkylene-OP (O) 2O-, - (C1-C22) alkylene-OP (O) (O-) O-, - (C1-C22) alkylene-OP (O) (O-) O- (C1-C22) alkylene-, - (C1-C22) alkylene-OP (O) O-, - (C1-C22) alkylene-OP (O) (S) O-, - (C1-C22) alkylene-O-S (O) 2-O-, - (C1-C22) alkylene-S (O) 2-O-, - (C1-C22) alkylene-S (O) -O-, -O-P (O) 2-O- (C1-C22) alkylene-OP (O) 2O-, -O-P (O) -O- (C1-C22) alkylene-OP (O) O-, -OP (O) (S) O- (C1-C22) alkylene-OP (O) (S) O-, -O-S (O) 2-O- (C1-C22) alkylene-O-S (O) 2-
O-, -S (O) 2-O- (C1-C22) alkylene-S (O) 2-O-and -O-S (O) - (C1-C22) alkylene-S (O) -O-; and
the oligonucleotide is selected from the group consisting of antisense oligonucleotide (ASO) , antisense RNA, short interfering RNA (siRNA) , micro-RNA (miRNA) , small activating RNA (saRNA) , double-stranded RNA (dsRNA) , and small guide RNA (sgRNA) .
In another specific embodiment, the oligonucleotide comprises at least part of the sequence as set forth in SEQ ID NO 1 to 53.
In another specific embodiment, the oligonucleotide delivery agent comprises a structure represented by Formula AA
wherein the delivery enhancing compound (DEC) moiety is derived from the oligonucleotide delivery enhancing compound according to any one of claims 1 to 9 and is linked to at least one oligonucleotide directly or indirectly.
In another specific embodiment, the DEC is linked with the oligonucleotide via at least one first linking moiety.
In another specific embodiment, the TM is linked with the DEC via at least one second linking moiety.
In another specific embodiment, each of the first linking moiety and the second linking moiety is independently selected from the group consisting of direct bond, -O-, -S-, -C (O) -, -NH-, -N ( (C1-C12) alkyl) -, -N ( (C1-C12) alkyl) -C (O) -O-, -O-C (O) -, -C (O) -O-, -O-C (O) -O-, -C (O) -NH-, -OP (O) 2O-, -OP (O) O-, -OP (O) (S) O-, -O-S (O) 2-O-, -S (O) 2-O-, -S (O) -O-, - (C1-C22) alkylene-, - (C1-C22) alkylene-NH-, -NH- (C1-C22) alkylene-, - (C1-C22) alkylene-NH-C (O) -, - (C1-C22) alkylene-C (O) -, - (C1-C22) alkylene-C (O) -O-, -C (O) - (C1-C22) alkylene-, -NH-C (O) - (C1-C22) alkylene-, -C (O) -NH- (C1-C22) alkylene-, -C (O) - (C1-C22) alkylene-NH-, -NH- (C1-C22) alkylene-C (O) -, -C (O) - (C1-C22) alkylene-C (O) -, -NH- (C1-C22) alkylene-NH-, -C (O) - (C1-C22) alkylene-C (O) O-, -O-C (O) - (C1-C22) alkylene-C (O) -O-, -C (O) -O- (C1-C22) alkylene-O-C (O) -, -C (O) - (C1-C22) alkylene-NH-C (O) -, -NH-C (O) - (C1-C22) alkylene-C (O) -, -NH-C (O) - (C1-C22) alkylene-C (O) -NH-, -NH- (C1-C22) alkylene-OP (O) 2O-, -NH- (C1-C22) alkylene-CH ( (C1-C22) alkylene-OH) -OP (O) 2O-, -NH- (C1-C22) alkylene-CH ( (C1-C22) alkylene-OH) - (C1-
C22) alkylene-OP (O) 2O-, -C (O) -NH- (C1-C22) alkylene-NH-C (O) -, - (C1-C22) alkylene-OP (O) 2O-, - (C1-C22) alkylene-OP (O) O-, - (C1-C22) alkylene-OP (O) (S) O-, - (C1-C22) alkylene-O-S (O) 2-O-, - (C1-C22) alkylene-S (O) 2-O-, - (C1-C22) alkylene-S (O) -O-, -O-P (O) 2-O- (C1-C22) alkylene-OP (O) 2O-, -O-P (O) -O- (C1-C22) alkylene-OP (O) O-, -OP (O) (S) O- (C1-C22) alkylene-OP (O) (S) O-, -O-S (O) 2-O- (C1-C22) alkylene-O-S (O) 2-O-, -S (O) 2-O- (C1-C22) alkylene-S (O) 2-O-and -O-S (O) - (C1-C22) alkylene-S (O) -O-; and/or
wherein the oligonucleotide is selected from the group consisting of short interfering RNA (siRNA) , small activating RNA (saRNA) , microRNA (miRNA) , antisense oligonucleotide (ASO) and small guide RNA (sgRNA) .
In another specific embodiment, the targeting moiety is one or more selected from the group consisting of ligands, peptides, nucleic acids, oligonucleotides, aptamers, lipids, fatty acids, small molecules, polyethylene glycols, amino acids, cholesterols, carbohydrates, and antibodies or antibody fragments.
In another specific embodiment, the oligonucleotide delivery agent has a structure represented by any of the formulae AAI to AAXXIV:
wherein L represents the linking moiety, ☆ represents the oligonucleotide delivery enhancing compound, the symbol adouble strand oligonucleotide in which each of the strands represents interchangeably a sense strand or an antisense strand, either symmetric or asymmetric independently on each of the ends; the symbol represents a single strand oligonucleotide, and each of a, b and c is independently an integer from 1 to 50.
In another specific embodiment, at least one hydrogen atom contained in the delivery enhancing compound moiety, the linking moiety, the targeting moiety and/or the oligonucleotide is substituted with deuterium atom.
In another specific embodiment, the present disclosure provides a pharmaceutical composition, the composition comprising: a) the oligonucleotide delivery agent as indicated herein; and b) optionally, one or more ingredients selected from the group consisting of pharmaceutically acceptable carrier, excipient, solvent, diluent, stabilizer, dispersant, buffer, compatibilizer, preservative agent and combinations thereof.
In another specific embodiment, the present disclosure provides a method of modulating the expression of a target gene in a subject, the method comprising the step of administrating the pharmaceutical composition as indicated herein to a subject.
In another specific embodiment, the oligonucleotide or the target gene comprises at least part of the sequence as set forth in at least part of the sequence as set forth in SEQ ID NO 1 to 53.
In another specific embodiment, the pharmaceutical composition increases the expression of the target gene. In another specific embodiment, the pharmaceutical composition decreases the expression of the target gene. In another specific embodiment, the subject is a mammal. In another specific embodiment, the mammal is a rodent. In another specific embodiment, the rodent is a mouse. In another specific embodiment, the rodent is a rat. In another specific embodiment, the mammal is a non-human primate. In another specific
embodiment, the mammal is a human. In another specific embodiment, the target gene is associated with a disease or disorder. In another specific embodiment, the target gene is associated with a disease or disorder in the central nervous system (CNS) , brain, spinal cord, liver, lung, kidney, intestine, pancreas, cholecyst, heart, lymph nodes, spleen, stomach, bladder, muscle or bone. In another specific embodiment, the disease is cancer.
In another specific embodiment, the present disclosure provides a method of modulating the expression of a target gene, the method comprising contacting a cell with the pharmaceutical composition as indicated herein.
In another specific embodiment, the oligonucleotide or the target gene comprises at least part of the sequence as set forth in at least part of the sequence as set forth in SEQ ID NO: 1 to 53.
In another specific embodiment, the pharmaceutical composition increases the expression of the target gene. In another specific embodiment, the pharmaceutical composition decreases the expression of the target gene. In another specific embodiment, the cell is a mammalian cell. In another specific embodiment, the mammalian cell is a mouse cell. In another specific embodiment, the mammalian cell is a rat cell. In another specific embodiment, the mammalian cell is a non-human primate cell. In another specific embodiment, the mammalian cell is a human cell. In another specific embodiment, the target gene is associated with a disease or disorder. In another specific embodiment, the target gene is associated with a disease or disorder in the central nervous system (CNS) , brain, spinal cord, liver, lung, kidney, intestine, pancreas, cholecyst, heart, lymph nodes, spleen, stomach, bladder, muscle or bone. In another specific embodiment, the disease is cancer.
EXAMPLES
Some embodiments of the invention will now be described in the following Examples, wherein all parts and percentages are by weight unless otherwise specified. However, the scope of the present disclosure is not, of course, limited to the formulations set forth in these examples. Rather, the Examples are merely inventive of the disclosure.
The following examples are set forth so as to provide those of ordinary skill in the art with a complete disclosure and description of how to make and use the present invention and are not intended to limit the scope of what the inventors regard as their invention nor are they
intended to represent that the experiments below are all or the only experiments performed. Efforts have been made to ensure accuracy with respect to numbers used (e.g., amounts, temperature, etc. ) but some experimental errors and deviations should be accounted for. Unless indicated otherwise, parts are parts by weight, molecular weight is weight average molecular weight, temperature is in degrees Celsius, and pressure is at or near atmospheric. Standard abbreviations may be used, e.g., bp, base pair (s) ; kb, kilobase (s) ; nM, nanomolar (s) ; s or sec, second (s) ; min, minute (s) ; h or hr, hour (s) ; aa, amino acid (s) ; nt, nucleotide (s) ; i. m., intramuscular (ly) ; i. p., intraperitoneal (ly) ; s. c., subcutaneous (ly) ; ivt or IVT, intravitreal; iv or IV, tail vein, intravenous; i. c. v. or icv or ICV, intracerebroventricular and the like.
All the starting materials, reagents and solvents used hereafter were purchased from commercial sources and used as received unless stated otherwise. Purification of reaction products was performed with a column chromatography comprising a silica gel (200-300 mesh) and eluting agents of hexane/ethyl acetate, DCM/MeOH. Thin layer chromatography (TLC) was carried out using pre-coated silica Gel GF plates and visualized using KMnO4 stains. 1H-NMR spectra were recorded at 400 or 500 MHz (Varian) using CDCl3 with TMS. High-resolution mass spectra (HRMS) were recorded on LC/MS (Agilent Technologies 1260 Infinity II/6120 Quadrupole) and a time-of-flight mass spectrometer by ESI or matrix assisted laser desorption/ionization (MALDI) .
Example 1. The preparation of compound A1 of the present disclosure
Compound A1 was prepared in this Example by using the following procedures.
(1) The preparation of compound A62
To a solution of Fmoc-L-hydroxyproline A61 (13.3 g, 37.6 mmol, 1.0 eq) in anhydrous THF (250 mL) , was added borane-methyl sulfide complex (8.0 mL of 10 M in THF, 80 mmol,
2.1 eq) slowly at room temperature. The reaction mixture was stirred for 5 min at room temperature and then heated to reflux for about 1 h. Methanol (15 mL) was carefully added to the reaction mixture, which was refluxed for 15 min. After that, the reaction mixture was concentrated under reduced pressure. Then the crude products were evaporated three times with methanol (100 mL each) . The crude product A62 was directly used in the next step without further purification.
(2) The preparation of compound A63
To a solution of compound A62 (37.6 mmol, 1.0 eq) in anhydrous pyridine (200 mL) , was added DMTrCl (14 g, 41.4 mmol, 1.1 eq) slowly at ice bath. The reaction was stirred under nitrogen atmosphere overnight and then concentrated under reduced pressure. The crude product was dissolved in dry MeCN (300 mL) then the mixture was added Et3N (15.6 mL, 113 mmol, 3.0 eq) and heated to 60 ℃ for 4 h. After concentrated under reduced pressure, the resulting residue was purified by flash chromatography (silica gel, gradient eluent: 1-8%of MeOH/DCM) to provide the desired product A63 (7.57 g, 48%yield) as yellow solid. The product was characterized with mass spectrometry and 1H NMR. MW calc.: 419.21; MW Found: 303.2 [DMT] -. 1H NMR (400 MHz, CDCl3) δ 7.41 (d, J = 7.4 Hz, 2H) , 7.30 (d, J = 8.8 Hz, 4H) , 7.28 -7.22 (m, 2H) , 7.18 (t, J = 7.2 Hz, 1H) , 6.80 (d, J = 8.8 Hz, 4H) , 4.34 (s, 1H) , 3.75 (d, J = 11.1 Hz, 6H) , 3.60 (dd, J = 12.7, 6.7 Hz, 1H) , 3.10 -2.92 (m, 5H) , 2.86 (d, J = 11.5 Hz, 1H) , 1.85 (dd, J = 13.5, 7.1 Hz, 1H) , 1.63 (ddd, J = 13.7, 7.9, 5.9 Hz, 1H) .
(3) The preparation of compound A66
To a solution of compound A64 (1.4 g, 7.2 mmol, 2.0 eq) in DCM (30 mL) , was added compound A65 (1.0 g, 3.6 mmol, 1.0 eq) , O-Benzotriazol-1-yl-N, N, N', N'-tetramethyluronium hexafluorophosphate (HBTU) (2.7 g, 7.2 mmol, 2.0 eq) and N, N-Diisopropylethylamine
(DIPEA) (0.93 g, 7.2 mmol, 2.0 eq) under nitrogen atmosphere. The reaction mixture was stirred overnight at room temperature. Then 30 mL H2O was added into the reaction mixture, and the mixture was extracted with DCM (3*30 mL) . The organic phase was combined, dried over Na2SO4, and concentrated. The resultant residue was purified by using flash chromatography (silica gel, gradient eluent: 1-5%of MeOH/DCM) to provide compound A66 (1.7 g, 94%yield) as colorless oil. The product was characterized with mass spectrometry and 1H NMR. MW calc.: 453.31; MW Found: 454.29 [M+H] +. 1H NMR (400 MHz, CDCl3) δ 3.69 -3.61 (m, 5H) , 3.59 -3.53 (m, 8H) , 3.48 (t, J = 5.2 Hz, 2H) , 3.35 (dd, J = 10.7, 5.3 Hz, 2H) , 3.15 (qd, J = 7.4, 4.4 Hz, 3H) , 2.43 (t, J = 6.4 Hz, 2H) , 1.92 -1.85 (m, 5H) , 1.54 -1.52 (m, 4H) , 1.37 (s, 12H) .
(4) The preparation of compound A67
To a solution of compound A66 (0.8 g, 1.7 mmol, 1.0 eq) in DCM (10 mL) , was added HCl/Dioxane (4M, 10 mL) . The reaction mixture was stirred for 3 h at room temperature, then concentrated under reduced pressure to provide crude product as yellow oil. The crude product was dissolved in DCM (10 mL) then added compound A63 (583 mg, 1.7 mmol, 1.0 eq) , HBTU (1.28 g, 3.4 mmol, 2.0 eq) and DIPEA (439 mg, 3.4 mmol, 2.0 eq) under nitrogen atmosphere. The reaction mixture was stirred overnight at room temperature. Then 10 mL H2O was added into the reaction mixture, and the mixture was extracted with DCM (3*20 mL) . The organic phase was combined, dried over Na2SO4, and concentrated. The resultant residue was purified by using flash chromatography (silica gel, gradient eluent: 10%of MeOH/DCM) to provide compound A67 (261 mg, 21%yield) as yellow solid. The product was characterized with mass spectrometry and 1H NMR. MW calc.: 798.45; MW Found: 303.31 [DMT] -, 519.62 [DMT off + Na] +. 1H NMR (400 MHz, CDCl3) δ 7.36 (d, J = 7.7 Hz, 2H) , 7.26 -7.23 (m, 6H) , 7.17 (t, J = 7.0 Hz, 1H) , 6.81 (dd, J = 11.4, 5.1 Hz, 4H) , 3.85 -3.81 (m, 1H) , 3.77 (s, 6H) ,
3.62 -3.56 (m, 8H) , 3.53 (dd, J = 9.1, 4.5 Hz, 2H) , 3.44 -3.32 (m, 4H) , 2.73 (q, J = 7.2 Hz, 6H) , 2.45 -2.33 (m, 1H) , 2.26 -2.16 (m, 1H) , 2.15 -2.02 (m, 1H) , 1.99 -1.90 (m, 5H) , 1.72 -1.66 (m, 3H) , 1.63 -1.58 (m, 9H) .
(5) The preparation of compound A68
To a solution of compound A67 (666 mg, 0.92 mmol, 1.0 eq) , DMAP (393 mg, 3.22 mmol, 3.5 eq) in DCM (6 mL) , succinic anhydride (282 mg, 2.76 mmol, 3.0 eq) were added under nitrogen atmosphere. The reaction mixture was stirred at room temperature overnight, and then H2O (10 mL) was added into the reaction. The mixture was extracted with DCM (3*10 mL) . The organic phase was combined, dried over Na2SO4, and concentrated. The resultant residue was purified with flash chromatography (silica gel, gradient eluent: 1-10%of MeOH/DCM) to provide compound A68 (447 mg, 59%yield) as colorless oil. The product was characterized with mass spectrometry and 1H NMR. MW calc.: 898.46; MW Found: 303.15 [DMT] -, 597.98 [DMT off + H] +. 1H NMR (400 MHz, CDCl3) δ 7.36 -7.32 (m, 2H) , 7.27 -7.25 (m, 6H) , 7.19 -7.16 (m, 1H) , 6.85 -6.76 (m, 4H) , 5.46 -5.32 (m, 1H) , 3.82 (d, J = 6.3 Hz, 1H) , 3.77 (s, 6H) , 3.60 (dd, J = 9.3, 4.5 Hz, 8H) , 3.53 (t, J = 5.0 Hz, 2H) , 3.44 -3.40 (m, 2H) , 3.14 (dd, J = 9.2, 2.9 Hz, 1H) , 2.99 -2.93 (m, 6H) , 2.62 -2.45 (m, 5H) , 2.34 -2.28 (m, 1H) , 2.19 -2.04 (m, 1H) , 1.96 -1.92 (m, 5H) , 1.69 -1.66 (m, 3H) , 1.62 -1.58 (m, 9H) .
(6) The preparation of compound A1
To a solution of compound A68 (200 mg, 0.223 mmol, 1.0 eq) , Controlled Pore Glass (CPG) (3.9 g) , DIPEA (111 μL, 0.669 mmol, 3.0 eq) in acetonitrile (30 mL) was added HBTU (169 mg, 0.446 mmol, 2.0 eq) under nitrogen atmosphere. The reaction mixture was stirred at
25 ℃ overnight and then sequentially washed with DCM and ethyl ether to produce the crude solid support. To a solution of acetic anhydride (12 mL) , pyridine (28 mL) , NEt3 (401 μL) in acetonitrile (18 mL) was added the crude solid support under nitrogen atmosphere. The reaction mixture was stirred at 25℃ overnight and then sequentially washed with DCM and ethyl ether to produce Compound A1 of the present disclosure (3.67 g) .
Example 2. The preparation of compound A2 of the present disclosure
Compound A2 was prepared in this Example by using the following procedures.
(1) The preparation of compound A70
To a solution of methyl 4-fluoro-3-nitrobenzoate A69 (8.0 g, 40.2 mmol, 1.0 eq) and K2CO3 (5.5 g, 40.2 mmol) in anhydrous DMF (100 mL) , under nitrogen atmosphere, was added benzyl (3-aminopropyl) carbamate (8.3 g, 40.2 mmol, 1.0 eq) . The reaction mixture was stirred at 25 ℃ for 6 h, then cold water (100 mL) was added. The mixture was extracted 3 times by ethyl acetate, then the organic phase was washed three times by saturated LiCl solution and one time by brine. Then dried by anhydrous Na2SO4 and concentrated under reduced pressure to form yellow solid compound A70 which was directly used in the next step without further purification.
(2) The preparation of compound A72
To a solution of compound A70 (10 g, 25.84 mmol, 1.0 eq) in THF/H2O (9: 1, 100 mL) , under ice bath, was added HCOONH4 (9.78 g, 155.04 mmol, 6.0 eq) and Zn powder (10.14 g, 155.04 mmol, 6.0 eq) . After 10 minutes, the reaction mixture was moved to room temperature and stirred overnight. Then the reaction mixture was filtered and concentrated under reduced pressure. After that, water (100 mL) was added into the mixture, then extracted three times by ethyl acetate, the organic phase was washed one time by brine. After dried by anhydrous Na2SO4 and concentrated under reduced pressure, the light red solid compound A72 formed then was directly used in the next step without further purification.
(3) The preparation of compound A73.
To a solution of compound A72 (8.0 g, 22.5 mmol, 1.0 eq) in EtOH (200 mL) under nitrogen atmosphere, was added 3- ( (tert-butyldimethylsilyl) oxy) propanal (4.2 g, 22.5 mmol, 1.0 eq) and AcOH (5.1 mL, 90 mmol, 4.0 eq) . The reaction mixture was stirred at 80℃ overnight, and then concentrated under reduced pressure. Then a saturated NaHCO3 solution (100 mL) was added, the mixture was extracted three times by ethyl acetate, then the organic phase was combined and washed by brine, dried over Na2SO4, and concentrated. The resultant residue compound A73 was directly used in the next step without further purification. The compound A73 was characterized with mass spectrometry. MW calc.: 525.27; MW. Found: 526.59 [M+H] +.
(4) The preparation of compound A74
To a solution of compound A73 (5.0 g, 9.5 mmol, 1.0 eq) in anhydrous THF (50 mL) , under nitrogen atmosphere, was added 1 M TBAF THF solution (14.25 mL, 14.25 mmol, 1.5 eq) . The reaction mixture was stirred at room temperature for 1 h, and then concentrated under
reduced pressure. Then water (100 mL) was added, the mixture was extracted three times by DCM, then the organic phase was combined and washed by brine, dried over Na2SO4, and concentrated. The resultant residue was dissolved in 50 mL pyridine, and DMTrCl (3.86 g, 11.4 mmol, 1.2 eq) was added. The reaction mixture was stirred at room temperature for 6 h, after concentrated under reduced pressure, the resultant residue was purified by flash chromatography (silica gel, gradient eluent: 1-5%of MeOH/DCM) to provide compound A74 (4.1 g, 60%yield) as yellow solid. The product was characterized with mass spectrometry and 1H NMR. MW calc.: 713.31; MW Found: 303.17 [DMT] -, 412.36 [DMT off + H] +. 1H NMR (400 MHz, CDCl3) δ 8.39 (d, J = 1.1 Hz, 1H) , 7.96 (dd, J = 8.5, 1.3 Hz, 1H) , 7.38 -7.27 (m, 8H) , 7.20 (dq, J = 6.7, 2.5 Hz, 7H) , 6.76 -6.70 (m, 4H) , 5.10 (s, 2H) , 4.19 (t, J = 7.3 Hz, 2H) , 3.94 (s, 3H) , 3.74 (s, 6H) , 3.65 (t, J = 6.6 Hz, 2H) , 3.20 (d, J = 6.3 Hz, 2H) , 3.09 (t, J = 6.4 Hz, 2H) , 2.00 -1.90 (m, 2H) .
(5) The preparation of compound A75
To a solution of compound A74 (4.0 g, 5.6 mmol, 1.0 eq) in anhydrous THF (50 mL) under nitrogen atmosphere and ice bath, was added LiAlH4 (319 mg, 8.4 mmol, 1.5 eq) . The mixture was moved to room temperature after 10 minutes and stirred for 1 h. Then the reaction was moved to ice bath, saturated potassium sodium tartrate solution (30 mL) was added slowly into the mixture. After 30 minutes, the reaction was extracted three times with Et2O, then the organic phase was combined and washed by brine, dried over Na2SO4, and concentrated. The resultant residue was dissolved in 50 mL DMF, then imidazole (572 mg, 8.4 mmol, 1.5 eq) and TBSCl (1.27 g, 8.4 mmol, 1.5 eq) was added. The reaction mixture was stirred at room temperature for 1 h, after concentrated under reduced pressure, the resultant residue was purified by flash chromatography (silica gel, gradient eluent: 1-5%of MeOH/DCM) to provide compound A75 (4.03 g, 90%yield) as yellow solid. The product was characterized with mass spectrometry and 1H NMR. MW calc.: 799.40; MW. Found: 303.17 [DMT] . 1H NMR (400 MHz, CDCl3) δ 7.65 (s, 1H) , 7.38 -7.29 (m, 7H) , 7.19 (ddd, J = 8.4, 7.9, 3.7 Hz, 9H) , 6.78 -6.71 (m, 4H) , 5.10 (s, 2H) , 4.84 (s, 2H) , 4.16 (t, J = 7.2 Hz, 2H) , 3.75 (s, 6H) , 3.61
(t, J = 6.8 Hz, 2H) , 3.18 (d, J = 6.3 Hz, 2H) , 3.07 (t, J = 6.4 Hz, 2H) , 2.95 (s, 1H) , 2.88 (s, 1H) , 0.94 (s, 9H) , 0.10 (s, 6H) .
(6) The preparation of compound A78
To a solution of compound A75 (220 mg, 0.276 mmol, 1.0 eq) in MeOH (8 mL) , was added Pd/C (22 mg) under nitrogen atmosphere slowly. Then the reaction atmosphere was replaced with hydrogen three times. After that, the reaction mixture was stirred by purging with a hydrogen balloon at room temperature overnight. Then the reaction mixture was filtered and concentrated under reduced pressure to provide crude product. The crude product was dissolved in DCM (8 mL) , then compound A77 (64 mg, 0.33 mmol, 1.2 eq) , HBTU (209 mg, 0.552 mmol, 2.0 eq) and DIPEA (155 μL, 0.938 mmol, 3.4 eq) were added into the reaction mixture under nitrogen atmosphere. The reaction mixture was stirred overnight at room temperature. Then H2O (10 mL) was added into the reaction, the mixture was extracted with DCM (3*10 mL) . The organic phase was combined, dried over Na2SO4, and concentrated. The resultant residue was purified with flash chromatography (silica gel, gradient eluent: 1-5%of MeOH/DCM) to provide compound A78 (199 mg, 86%yield) as yellow oil. The product was characterized by mass spectrometry and 1H NMR. MW calc.: 841.48; MW Found: 303.2 [DMT] -, 540.3 [DMT off + H] +. 1H NMR (400 MHz, CDCl3) δ 7.64 (d, J = 6.5 Hz, 1H) , 7.34 -7.27 (m, 4H) , 7.22 -7.17 (m, 6H) , 6.83 (dd, J = 8.1, 2.7 Hz, 1H) , 6.81 -6.65 (m, 4H) , 4.84 (d, J = 3.3 Hz, 2H) , 4.23 -4.15 (m, 2H) , 3.87 -3.78 (m, 2H) , 3.77 (s, 6H) , 3.72 -3.48 (m, 2H) , 3.29 -3.19 (m, 2H) , 3.17 -3.06 (m, 2H) , 1.87 -1.81 (m, 6H) , 1.68 -1.57 (m, 11H) , 0.95 (s, 9H) , 0.11 (s, 6H) .
(7) The preparation of compound A79
To a solution of compound A78 (165 mg, 0.196 mmol, 1.0 eq) in anhydrous THF (5 mL) , under nitrogen atmosphere, was added 1 M TBAF THF solution (1.18 mL, 1.18 mmol, 6.0 eq) . The reaction mixture was stirred at room temperature for 1 h, and then concentrated under reduced pressure to provide crude product. The crude product was dissolved in DCM (8 mL) , then DMAP (84 mg, 0.686 mmol, 3.5 eq) , succinic anhydride (39 mg, 0.392 mmol, 2.0 eq) were added under nitrogen atmosphere. The reaction mixture was stirred at room temperature overnight then H2O (10 mL) was added into the reaction, the mixture was extracted with DCM (3*10 mL) and the organic phase was combined, dried over Na2SO4, and concentrated. The resultant residue was purified with flash chromatography. The resultant residue was purified with flash chromatography (silica gel, gradient eluent: 1-8%of MeOH/DCM) to provide compound A79 (97 mg, 60%yield) as colorless oil. The product was characterized with mass spectrometry and 1H NMR. MW calc.: 827.41; MW Found: 303.2 [DMT] -, 527.3 [DMT off + H] +. 1H NMR (400 MHz, CDCl3) δ 7.66 (d, J = 4.4 Hz, 1H) , 7.33 -7.29 (m, 2H) , 7.24 (s, 2H) , 7.23 -7.14 (m, 7H) , 6.75 (dd, J = 8.1, 5.6 Hz, 4H) , 5.22 (s, 2H) , 4.18 (s, 2H) , 3.75 (s, 6H) , 3.61 (t, J = 6.7 Hz, 2H) , 3.23 (s, 2H) , 3.08 (d, J = 13.2 Hz, 2H) , 2.95 (dd, J = 14.6, 7.3 Hz, 2H) , 2.71 -2.55 (m, 4H) , 1.98 -1.92 (m, 4H) , 1.85 (d, J = 3.6 Hz, 2H) , 1.70 -1.59 (m, 6H) , 1.33 -1.16 (m, 5H) .
(8) The preparation of compound A2
To a solution of the compound A79 (85 mg, 0.103 mmol, 1.0 eq) , Controlled Pore Glass (CPG) (1.8 g) , DIPEA (51 μL, 0.31 mmol, 3.0 eq) in acetonitrile (14 mL) was added HBTU (78 mg, 0.206 mmol, 2.0 eq) under nitrogen atmosphere. The reaction mixture was stirred at 25℃ overnight and then subsequently washed with DCM and ethyl ether to generate crude solid support. To a solution of acetic anhydride (6.2 mL) , pyridine (12 mL) , NEt3 (186
μL) in acetonitrile (7.9 mL) was added the crude solid support under nitrogen atmosphere. The reaction mixture was stirred at 25 ℃ overnight and then subsequently washed with DCM and ethyl ether to produce the Compound A2 of the present disclosure (1.62 g) .
Example 3. The preparation of Compound A3 of the present disclosure.
The Compound A3 of the present disclosure was prepared in this Example by using the following procedures.
(1) The preparation of compound A81
To a solution of compound A75 (1.44 g, 1.8 mmol, 1.0 eq) in MeOH (30 mL) , was added Pd/C (144 mg) under nitrogen atmosphere slowly. Then the reaction atmosphere was replaced with hydrogen three times. After that, the reaction mixture was stirred by purging with a hydrogen balloon at room temperature overnight. Then the reaction mixture was filtered and concentrated under reduced pressure to provide crude product. The crude product was dissolved in DCM (40 mL) , then palmitic acid (508 mg, 1.98 mmol, 1.1 eq) , HBTU (1.36 g, 3.6 mmol, 2.0 eq) and DIPEA (1.01 mL, 6.12 mmol, 3.4 eq) were added into the reaction mixture under nitrogen atmosphere. The reaction mixture was stirred overnight at room temperature. Then H2O (30 mL) was added into the reaction, the mixture was extracted with DCM (3*40 mL) . The organic phase was combined, dried over Na2SO4, and concentrated. The resultant residue was purified with flash chromatography (silica gel, gradient eluent: 1-5%of MeOH/DCM) to provide compound A81 (1.43 g, 88%yield) as yellow oil. The product was characterized with mass spectrometry and 1H NMR. MW calc.: 903.59; MW Found: 303.2 [DMT] -, 488.4 [DMT and TBS off + H] +. 1H NMR (400 MHz, CDCl3) δ 7.62 (d, J = 4.7 Hz, 1H) , 7.33 -7.27 (m, 4H) , 7.22 -7.16 (m, 7H) , 6.74 (dd, J = 8.1, 5.6 Hz, 4H) , 4.83 (s, 2H) ,
4.23 -4.16 (m, 2H) , 3.76 (s, 6H) , 3.62 (dd, J = 11.9, 5.3 Hz, 2H) , 3.27 -3.21 (m, 2H) , 3.15 (d, J = 6.9 Hz, 2H) , 2.11 -2.05 (m, 2H) , 2.03 -1.90 (m, 2H) , 1.34 -1.25 (m, 29H) , 0.95 (s, 9H) , 0.11 (s, 6H) .
(2) The preparation of compound A82
To a solution of compound A81 (1.4 g, 1.55 mmol, 1.0 eq) in anhydrous THF (30 mL) , under nitrogen atmosphere, was added 1 M TBAF THF solution (4.65 mL, 4.65 mmol, 3.0 eq) . The reaction mixture was stirred at room temperature for 1 h, and then concentrated under reduced pressure to provide crude product. The crude product was dissolved in DCM (40 mL) , then DMAP (660 mg, 5.4 mmol, 3.5 eq) , succinic anhydride (465 mg, 4.65 mmol, 3.0 eq) were added therein under nitrogen atmosphere. The reaction mixture was stirred at room temperature overnight then H2O (30 mL) was added into the reaction, the mixture was extracted with DCM (3*30 mL) and the organic phase was combined, dried over Na2SO4, and concentrated. The resultant residue was purified with flash chromatography. The resultant residue was purified with flash chromatography (silica gel, gradient eluent: 1-8%of MeOH/DCM) to provide compound A82 (827 mg, 60%yield) as colorless oil. The product was characterized with mass spectrometry and 1H NMR. MW calc.: 889.52; MW Found: 303.1 [DMT] -, 588.3 [DMT off + H] +. 1H NMR (400 MHz, CDCl3) δ 7.65 (d, J = 4.5 Hz, 1H) , 7.34 -7.28 (m, 2H) , 7.25 -7.14 (m, 9H) , 6.74 (dd, J = 8.1, 5.5 Hz, 4H) , 5.21 (s, 2H) , 4.34 -3.90 (m, 2H) , 3.74 (s, 6H) , 3.60 (dd, J = 12.1, 5.4 Hz, 2H) , 3.22 -3.16 (m, 2H) , 3.09 -3.04 (m, 2H) , 2.63 -2.60 (m, 4H) , 2.16 -2.01 (m, 2H) , 1.97 -1.84 (m, 2H) , 1.33 -1.23 (m, 26H) , 0.87 (t, J = 6.7 Hz, 3H) .
(3) The preparation of compound A3
To a solution of the compound A82 (120 mg, 0.13 mmol, 1.0 eq) , Controlled Pore Glass (CPG) (2.29 g) , DIPEA (65 μL, 0.39 mmol, 3.0 eq) in acetonitrile (15 mL) was added HBTU (99 mg, 0.26 mmol, 2.0 eq) under nitrogen atmosphere. The reaction mixture was stirred at 25℃ overnight and then subsequently washed with DCM and ethyl ether to generate crude solid support. To a solution of acetic anhydride (7.8 mL) , pyridine (15 mL) , NEt3 (234 μL) in acetonitrile (10 mL) was added the crude solid support under nitrogen atmosphere. The reaction mixture was stirred at 25 ℃ overnight and then subsequently washed with DCM and ethyl ether to produce the Compound A3 of the present disclosure (2.1 g) .
Example 4. The preparation of compound A4 and compound A5 of the present disclosure
Compound A4 and compound A5 were prepared in this Example by using the following procedures.
(1) The preparation of compound A85
To a solution of methyl 2- (4-fluoro-3-nitrophenyl) acetate A84 (17.3 g, 81 mmol, 1.0 eq) and K2CO3 (11.2 g, 81 mmol) in anhydrous DMF (200 mL) , under nitrogen atmosphere, was added compound A83 (19.56 g, 81 mmol, 1.0 eq) . The reaction mixture was stirred at 55 ℃ for 6 h, then cold water (100 mL) was added. The mixture was extracted three times by ethyl acetate, then the organic phase was washed three times by saturated LiCl solution and one time by brine. Then dried by anhydrous Na2SO4 and concentrated under reduced pressure to form yellow oil compound A85 which was directly used in the next step without further purification.
(2) The preparation of compound A86
To a solution of compound A85 (35.18 g, 81 mmol, 1.0 eq) in THF/H2O (9: 1, 280 mL) , under ice bath, was added HCOONH4 (30.67 g, 486 mmol, 6.0 eq) and Zn powder (31.78 g, 486 mmol, 6.0 eq) . After 10 minutes, the reaction mixture was moved to room temperature and stirred overnight. Then the reaction mixture was filtered and concentrated under reduced pressure. After that, water (200 mL) was added into the mixture, then extracted three times by ethyl acetate, the organic phase was washed one time by brine. After dried by anhydrous Na2SO4 and concentrated under reduced pressure, the product A86 formed then was directly used in the next step without further purification. The compound A86 was characterized with mass spectrometry. MW calc.: 404.34; MW. Found: 405.3 [M+H] +.
(3) The preparation of compound A88
To a solution of compound A86 (19.36 g, 48 mmol, 1.0 eq) in EtOH (200 mL) under nitrogen atmosphere, was added 3- ( (tert-butyldimethylsilyl) oxy) propanal A87 (9.0 g, 48 mmol, 1.0 eq) and AcOH (11 mL, 192 mmol, 4.0 eq) . The reaction mixture was stirred at 80℃
overnight, and then concentrated under reduced pressure. Then a saturated NaHCO3 solution (100 mL) was added, the mixture was extracted three times by ethyl acetate, then the organic phase was combined and washed by brine, dried over Na2SO4, and concentrated. The resultant residue compound A88 was directly used in the next step without further purification.
(4) The preparation of compound A89
To a solution of compound A88 (10 g, 17.5 mmol, 1.0 eq) in anhydrous THF (50 mL) under nitrogen atmosphere, was added 1 M TBAF THF solution (26.3 mL, 26.3 mmol, 1.5 eq) . The reaction mixture was stirred at room temperature for 1 h, and then concentrated under reduced pressure. Then water (100 mL) was added, the mixture was extracted three times with DCM, then the organic phase was combined and washed with brine, dried over Na2SO4, and concentrated. The resultant residue was dissolved in 50 mL pyridine, and DMTrCl (7.12 g, 21 mmol, 1.2 eq) was added therein. The reaction mixture was stirred at room temperature for 6 h, after which it was concentrated under reduced pressure. The resultant residue was purified with flash chromatography (silica gel, gradient eluent: 1-3%of MeOH/DCM) to provide compound A89 (8.1 g, 61%yield) as yellow solid. The product was characterized with mass spectrometry and 1H NMR. MW calc.: 760.48; MW. Found: 761.8 [M + H] +. 1H NMR (400 MHz, CDCl3) δ 7.59 -7.53 (m, 1H) , 7.38 -7.31 (m, 2H) , 7.23 -7.19 (m, 6H) , 7.18 -7.14 (m, 3H) , 6.76 (dd, J = 7.8, 5.6 Hz, 4H) , 4.17 -4.02 (m, 2H) , 3.76 (s, 6H) , 3.73 (s, 2H) , 3.67 (s, 3H) , 3.59 (t, J = 7.0 Hz, 2H) , 3.19 -3.05 (m, 2H) , 1.29 -1.25 (m, 28H) , 0.88 (t, J = 6.5 Hz, 3H) .
(5) The preparation of compound A90
To a solution of compound A89 (2.7 g, 3.55 mmol, 1.0 eq) in anhydrous THF (20 mL) under nitrogen atmosphere and ice bath, was added LiAlH4 (202 mg, 5.33 mmol, 1.5 eq) . The mixture was moved to room temperature after 10 minutes and stirred for 1 h. Then the reaction
was moved to ice bath, saturated potassium sodium tartrate solution (20 mL) was added slowly into the mixture. After 30 minutes, the reaction was extracted three times with Et2O, then the organic phase was combined and washed by brine, dried over Na2SO4, and concentrated. The crude product (300 mg, 0.41 mmol, 1.0 eq) was dissolved in DCM (10 mL) , then DMAP (175 g, 1.44 mmol, 3.5 eq) , succinic anhydride (123 mg, 1.23 mmol, 3.0 eq) were added under nitrogen atmosphere. The reaction mixture was stirred at room temperature overnight then H2O (10 mL) was added into the reaction, the mixture was extracted with DCM (3*20 mL) and the organic phase was combined, dried over Na2SO4, and concentrated. The resultant residue was purified with flash chromatography. The resultant residue was purified with flash chromatography (silica gel, gradient eluent: 1-10%of MeOH/DCM) to provide compound A90 (113 mg, 33%yield) as colorless oil. The product was characterized with mass spectrometry and 1H NMR. MW calc.: 832.50; MW Found: 303.2 [DMT] -, 531.3 [DMT off + H] +. 1H NMR (400 MHz, CDCl3) δ 7.63 (s, 1H) , 7.31 (dd, J = 7.6, 4.1 Hz, 2H) , 7.28 -7.11 (m, 9H) , 6.74 (dd, J = 7.9, 5.6 Hz, 4H) , 4.35 -4.32 (m, 2H) , 4.12 -4.08 (m, 2H) , 3.75 (s, 6H) , 3.59 (dd, J = 11.8, 5.1 Hz, 2H) , 3.24 -3.10 (m, 2H) , 3.09 -2.96 (m, 2H) , 2.63 (dd, J = 7.2, 3.0 Hz, 4H) , 1.37 -1.25 (m, 28H) , 0.88 (t, J = 6.5 Hz, 3H) .
(6) The preparation of compound A4
To a solution of the compound A90 (90 mg, 0.108 mmol, 1.0 eq) , Controlled Pore Glass (CPG) (1.89 g) , DIPEA (54 μL, 0.324 mmol, 3.0 eq) in acetonitrile (14 mL) was added HBTU (82 mg, 0.216 mmol, 2.0 eq) under nitrogen atmosphere. The reaction mixture was stirred at 25℃ overnight and then subsequently washed with DCM and ethyl ether to generate crude solid support. To a solution of acetic anhydride (6.5 mL) , pyridine (12.6 mL) , NEt3 (195 μL) in acetonitrile (8.3 mL) was added the crude solid support under nitrogen atmosphere. The reaction mixture was stirred at 25 ℃ overnight and then subsequently washed with DCM and ethyl ether to produce the Compound A4 of the present disclosure (1.7 g) .
(7) The preparation of compound A5
To a solution of compound A89 (2.7 g, 3.55 mmol, 1.0 eq) in anhydrous THF (20 mL) under nitrogen atmosphere and ice bath, was added LiAlH4 (202 mg, 5.33 mmol, 1.5 eq) . The mixture was moved to room temperature after 10 minutes and stirred for 1 h. Then the reaction was moved to ice bath, saturated potassium sodium tartrate solution (20 mL) was added slowly into the mixture. After 30 minutes, the reaction was extracted three times with Et2O, then the organic phase was combined and washed by brine, dried over Na2SO4, and concentrated. The crude product (300 mg, 0.41 mmol, 1.0 eq) was dissolved in anhydrous DCM (5 mL) then DIPEA (204 μL, 1.23 mmol, 3.0 eq) , 3- ( (chloro (diisopropylamino) phosphanyl) oxy) propanenitrile (274 μL, 1.23 mmol, 3.0 eq. ) were added under nitrogen atmosphere at 25℃. The reaction mixture was stirred for 1 h. The mixture was extracted two times with DCM, then washed with brine and dried with anhydrous Na2SO4. The organic layer was concentrated under reduced pressure and the resultant residue was purified with flash chromatography (silica gel, gradient eluent: 1-5%of MeOH/DCM, 1%Et3N) to provide compound A5 (299 mg, 78%yield) as colorless oil. The product was characterized with mass spectrometry and 1H NMR. MW calc.: 946.61; MW Found: 303.2 [DMT] -. 1H NMR (400 MHz, CDCl3) δ 7.62 (s, 1H) , 7.32 (dd, J = 7.6, 4.1 Hz, 2H) , 7.27 -7.11 (m, 9H) , 6.73 (dd, J = 7.9, 5.6 Hz, 4H) , 4.34 -4.31 (m, 2H) , 4.10 -4.06 (m, 2H) , 3.75 (s, 6H) , 3.64 -3.61 (m, 2H) , 3.58 (dd, J = 11.8, 5.1 Hz, 2H) , 3.24 -3.18 (m, 2H) , 3.09 -2.88 (m, 4H) , 2.65 -2.55 (m, 4H) , 1.37 -1.28 (m, 28H) , 1.22 (dd, J = 6.8, 3.2 Hz, 12H) , 0.88 (t, J = 6.5 Hz, 3H) .
Example 5. The preparation of compound A6 of the present disclosure
Compound A6 was prepared in this Example by using the following procedures.
(1) The preparation of compound A92
To a solution of compound A72 (8.87 g, 24.84 mmol, 1.0 eq) in EtOH (220 mL) under nitrogen atmosphere, was added 5- ( (tert-butyldimethylsilyl) oxy) pentanal A91 (5.4 g, 24.84 mmol, 1.0 eq) and AcOH (5.73 mL, 99.36 mmol, 4.0 eq) . The reaction mixture was stirred at 80℃ overnight, and then concentrated under reduced pressure. Then saturated NaHCO3 solution (100 mL) was added therein, the mixture was extracted three times with ethyl acetate. The organic phase was combined and washed by brine, dried over Na2SO4, and concentrated. The resultant residue was purified with flash chromatography (silica gel, gradient eluent: 1-5%of MeOH/DCM) to provide the compound A92 (6.3 g, 46%yield) as red solid. The product was characterized with mass spectrometry, 1H NMR and 13C NMR. MW calc.: 553.30; MW Found: 554.29 [M+H] +. 1H NMR (400 MHz, CDCl3) δ 8.42 (d, J = 0.8 Hz, 1H) , 7.95 (dd, J = 8.5, 1.1 Hz, 1H) , 7.44 -7.28 (m, 5H) , 7.26 (d, J = 6.8 Hz, 1H) , 5.12 (s, 2H) , 4.16 (dd, J = 12.6, 5.3 Hz, 2H) , 3.93 (s, 3H) , 3.67 (t, J = 6.3 Hz, 2H) , 3.28 (d, J = 6.2 Hz, 2H) , 2.87 (t, J = 7.6 Hz, 2H) , 2.04 -1.88 (m, 4H) , 1.67 (dd, J = 14.7, 6.6 Hz, 2H) , 0.88 (s, 9H) , 0.04 (s, 6H) . 13C NMR (100 MHz, CDCl3) δ 167.72, 156.63, 142.35, 138.19, 136.30, 128.60, 128.24, 124.10, 123.87, 121.63, 108.66, 66.96, 62.71, 60.41, 52.06, 41.31, 38.57, 32.46, 30.43, 27.30, 25.97, 24.15, 21.07, 18.34, 14.21.
(2) The preparation of compound A93
To a solution of compound A92 (6.3 g, 11.4 mmol, 1.0 eq) in anhydrous THF (50 mL)
under nitrogen atmosphere, was added 1 M TBAF THF solution (17.1 mL, 17.1 mmol, 1.5 eq) . The reaction mixture was stirred at room temperature for 1 h, and then concentrated under reduced pressure. Then water (100 mL) was added, the mixture was extracted three times with DCM, then the organic phase was combined and washed with brine, dried over Na2SO4, and concentrated. The resultant residue was dissolved in 50 mL pyridine, and DMTrCl (4.6 g, 13.68 mmol, 1.2 eq) was added therein. The reaction mixture was stirred at room temperature for 6 h, after which it was concentrated under reduced pressure. The resultant residue was purified with flash chromatography (silica gel, gradient eluent: 1-5%of MeOH/DCM) to provide compound A93 (6.23 g, 74%yield) as yellow solid. The product was characterized with mass spectrometry, 1H NMR and 13C NMR. MW calc.: 741.34; MW. Found: 303.11 [DMT] -, 440.14 [DMT off + H] +. 1H NMR (400 MHz, CDCl3) δ 8.63 (dd, J = 5.7, 1.5 Hz, 1H) , 8.43 (d, J = 1.0 Hz, 1H) , 7.97 (dd, J = 8.5, 1.1 Hz, 1H) , 7.45 (d, J = 7.4 Hz, 2H) , 7.37 -7.28 (m, 11H) , 7.21 (dd, J = 8.1, 6.1 Hz, 1H) , 6.83 (t, J = 5.9 Hz, 4H) , 5.13 (s, 2H) , 4.12 (t, J = 7.2 Hz, 2H) , 3.95 (s, 3H) , 3.79 (s, 6H) , 3.24 (d, J = 6.2 Hz, 2H) , 3.14 (t, J = 6.2 Hz, 2H) , 2.83 (t, J = 7.4 Hz, 2H) , 2.01 (dd, J = 16.0, 8.5 Hz, 4H) , 1.79 (dd, J = 14.1, 6.6 Hz, 2H) . 13C NMR (100 MHz, CDCl3) δ 167.81, 158.43, 156.64, 149.95, 145.34, 142.43, 138.26, 136.57, 136.05, 130.11, 128.69, 128.39, 128.26, 128.23, 127.84, 126.75, 124.17, 123.89, 121.70, 113.11, 108.75, 85.93, 67.02, 62.90, 55.31, 52.16, 41.37, 38.64, 30.50, 29.76, 27.30, 24.53.
(3) The preparation of compound A94
To a solution of compound A93 (5.0 g, 6.74 mmol, 1.0 eq) in anhydrous THF (60 mL) under nitrogen atmosphere and ice bath, was added LiAlH4 (384 mg, 10.11 mmol, 1.5 eq) . The mixture was then transferred to room temperature after 10 minutes and stirred for about 1 h. Then the reaction was transferred to ice bath, and saturated potassium sodium tartrate solution (30 mL) was added slowly into the mixture. After reacting for 30 minutes, the reaction was extracted three times with Et2O, then the organic phase was combined and washed with brine, dried over Na2SO4, and concentrated. The resultant residue was dissolved in 20 mL DMF, then
imidazole (688 mg, 10.11 mmol, 1.5 eq) and TBSCl (1.524 g, 10.11 mmol, 1.5 eq) were added. The reaction mixture was stirred at room temperature for 1 h. After being concentrated under reduced pressure, the resultant residue was purified with flash chromatography (silica gel, gradient eluent: 1-5%of MeOH/DCM) to provide compound A94 (4.78 g, 86%yield) as yellow solid. The product was characterized with mass spectrometry, 1H NMR and 13C NMR. MW calc.: 827.43; MW. Found: 303.16 [DMT] -, 526.50 [DMT off + H] +. 1H NMR (400 MHz, CDCl3) δ 7.69 (s, 1H) , 7.47 -7.42 (m, 2H) , 7.40 -7.28 (m, 11H) , 7.22 (t, J = 3.5 Hz, 3H) , 6.86 -6.80 (m, 4H) , 5.13 (s, 2H) , 4.86 (s, 2H) , 4.15 (dd, J = 14.3, 7.2 Hz, 2H) , 3.79 (s, 6H) , 3.13 (t, J = 6.3 Hz, 2H) , 2.82 (t, J = 7.5 Hz, 2H) , 2.05 -1.94 (m, 4H) , 1.81 -1.74 (m, 2H) , 1.29 (t, J = 7.1 Hz, 2H) , 0.97 (s, 9H) , 0.13 (s, 6H) . 13C NMR (100 MHz, CDCl3) δ 171.27, 158.45, 156.60, 154.95, 145.40, 142.93, 136.66, 135.52, 134.14, 130.14, 128.70, 128.37, 128.28, 127.85, 126.74, 120.93, 117.18, 113.13, 108.73, 85.93, 67.00, 65.53, 63.00, 60.51, 55.32, 53.55, 41.19, 38.74, 30.51, 29.85, 27.33, 26.14, 24.78, 21.17, 18.57, 14.33.
(4) The preparation of compound A96
To a solution of compound A94 (264 mg, 0.319 mmol, 1.0 eq) in MeOH (5 mL) , was added Pd/C (16 mg) under nitrogen atmosphere slowly. Then the reaction atmosphere was replaced with hydrogen three times. After that, the reaction mixture was stirred by purging with a hydrogen balloon at room temperature overnight. Then the reaction mixture was filtered and concentrated under reduced pressure to provide crude product. The crude product was dissolved in DCM (8 mL) , then compound A77 (74 mg, 0.38 mmol, 1.2 eq) , HBTU (242 mg, 0.64 mmol, 2.0 eq) and DIPEA (179 μL, 1.08 mmol, 3.4 eq) were added into the reaction mixture under nitrogen atmosphere. The reaction mixture was stirred overnight at room temperature. Then H2O (10 mL) was added into the reaction, the mixture was extracted with DCM (3*10 mL) . The organic phase was combined, dried over Na2SO4, and concentrated. The
resultant residue was purified with flash chromatography (silica gel, gradient eluent: 1-5%of MeOH/DCM) to provide compound A96 (236 mg, 85%yield) as yellow solid. The product was characterized with mass spectrometry and 1H NMR. MW calc.: 869.52; MW. Found: 303.2 [DMT] -, 568.4 [DMT off + H] +. 1H NMR (400 MHz, CDCl3) δ 7.67 (s, 1H) , 7.44 -7.39 (m, 2H) , 7.32 -7.27 (m, 5H) , 7.26 -7.14 (m, 4H) , 6.83 -6.78 (m, 4H) , 4.83 (s, 2H) , 4.11 (t, J = 7.4 Hz, 2H) , 3.78 (s, 6H) , 3.25 (dd, J = 13.2, 6.7 Hz, 2H) , 3.10 (t, J = 6.2 Hz, 2H) , 2.86 (dd, J = 9.5, 5.9 Hz, 2H) , 1.97 (d, J = 13.0 Hz, 6H) , 1.85 (s, 2H) , 1.74 -1.65 (m, 6H) , 1.59 (dd, J = 13.7, 7.1 Hz, 9H) , 0.94 (s, 9H) , 0.10 (s, 6H) .
(5) The preparation of compound A97
To a solution of compound A96 (223 mg, 0.26 mmol, 1.0 eq) in anhydrous THF (7 mL) , under nitrogen atmosphere, was added 1 M TBAF THF solution (1.54 mL, 1.54 mmol, 6.0 eq) . The reaction mixture was stirred at room temperature for 1 h, and then concentrated under reduced pressure to provide crude product. The crude product was dissolved in DCM (10 mL) , then DMAP (110 mg, 0.90 mmol, 3.5 eq) , succinic anhydride (52 mg, 0.52 mmol, 2.0 eq) were added therein under nitrogen atmosphere. The reaction mixture was stirred at room temperature overnight then H2O (10 mL) was added into the reaction, the mixture was extracted with DCM (3*10 mL) and the organic phase was combined, dried over Na2SO4, and concentrated. The resultant residue was purified with flash chromatography. The resultant residue was purified with flash chromatography (silica gel, gradient eluent: 1-10%of MeOH/DCM) to provide compound A97 (116 mg, 53%yield) as colorless oil. The product was characterized with mass spectrometry and 1H NMR. MW calc.: 855.45; MW. Found: 303.2 [DMT] -, 554.3 [DMT off + H] +. 1H NMR (400 MHz, CDCl3) δ 7.62 (s, 1H) , 7.42 (dd, J = 7.0, 1.5 Hz, 2H) , 7.37 (s, 1H) , 7.32 -7.26 (m, 6H) , 7.18 (t, J = 7.2 Hz, 2H) , 6.82 -6.79 (m, 4H) , 5.19 (s, 2H) , 4.28 -4.12 (m, 2H) , 3.78 (s, 6H) , 3.24 (s, 2H) , 3.09 (t, J = 6.2 Hz, 2H) , 2.88 -2.83 (m, 2H) , 2.64 (t, J = 3.9 Hz, 4H) , 1.96 (d, J = 13.0 Hz, 6H) , 1.84 (s, 2H) , 1.73 -1.66 (m, 6H) , 1.57 (dd, J = 13.7, 7.1 Hz, 9H) .
(6) The preparation of compound A6
To a solution of the compound A97 (100 mg, 0.117 mmol, 1.0 eq) , Controlled Pore Glass (CPG) (2.05 g) , DIPEA (58 μL, 0.35 mmol, 3.0 eq) in acetonitrile (16 mL) was added HBTU (89 mg, 0.234 mmol, 2.0 eq) under nitrogen atmosphere. The reaction mixture was stirred at 25℃ overnight and then subsequently washed with DCM and ethyl ether to generate crude solid support. To a solution of acetic anhydride (7.02 mL) , pyridine (13.5 mL) , NEt3 (211 μL) in acetonitrile (9 mL) was added the crude solid support under nitrogen atmosphere. The reaction mixture was stirred at 25 ℃ overnight and then subsequently washed with DCM and ethyl ether to produce the Compound A6 of the present disclosure (1.76 g) .
Example 6. The preparation of compound A7 of the present disclosure
Compound A7 was prepared in this Example by using the following procedures.
(1) The preparation of compound A99
To a solution of methyl 4-fluoro-3-nitrobenzoate A69 (8.0 g, 40.2 mmol, 1.0 eq) and K2CO3 (5.5 g, 40.2 mmol, 1.0 eq) in anhydrous DMF (100 mL) , under nitrogen atmosphere, was added compound A98 (8.57 g, 40.2 mmol, 1.0 eq) . The reaction mixture was stirred at 25 ℃ for 6 h, then cold water (100 mL) was added. The mixture was extracted three times by
ethyl acetate, then the organic phase was washed three times by saturated LiCl solution and one time by brine. Then dried by anhydrous Na2SO4 and concentrated under reduced pressure to form brown oil compound A99 which was directly used in the next step without further purification.
(2) The preparation of compound A100
To a solution of compound A99 (15.76 g, 40.17 mmol, 1.0 eq) in THF/H2O (9: 1, 165 mL) , under ice bath, was added HCOONH4 (15.21 g, 241 mmol, 6.0 eq) and Zn powder (15.76 g, 241 mmol, 6.0 eq) . After 10 minutes, the reaction mixture was moved to room temperature and stirred overnight. Then the reaction mixture was filtered and concentrated under reduced pressure. After that, water (100 mL) was added into the mixture, then extracted three times by ethyl acetate, the organic phase was washed one time by brine. After dried by anhydrous Na2SO4 and concentrated under reduced pressure, the brown solid compound A100 formed then was directly used in the next step without further purification. The compound was characterized with mass spectrometry. MW calc.: 362.29; MW. Found: 363.4 [M+H] +.
(3) The preparation of compound A102
To a solution of compound A100 (6.0 g, 16.56 mmol, 1.0 eq) in EtOH (100 mL) under nitrogen atmosphere, was added compound A101 (1.69 g, 16.56 mmol, 1.0 eq) and AcOH (3.8 mL, 66.24 mmol, 4.0 eq) . The reaction mixture was stirred at 80℃ overnight, and then concentrated under reduced pressure. Then a saturated NaHCO3 solution (100 mL) was added, the mixture was extracted three times by ethyl acetate, then the organic phase was combined and washed by brine, dried over Na2SO4, and concentrated. The resultant residue compound was directly used in the next step without further purification. The compound was characterized with mass spectrometry. MW calc.: 444.34; MW. Found: 445.4 [M+H] +.
The resultant residue (1.0 g, 2.25 mol, 1.0 eq) was dissolved in 10 mL DCM, and Et3N (0.47 mL, 3.37 mmol, 1.5 eq) , DMTrCl (915 mg, 2.7 mmol, 1.2 eq) were added therein. The
reaction mixture was stirred at room temperature for 6 h, after which it was concentrated under reduced pressure. The resultant residue compound was directly used in the next step without further purification. The product A102 was characterized with mass spectrometry. MW calc.: 746.47; MW. Found: 303.2 [DMT] -.
(4) The preparation of compound A103
To a solution of compound A102 (1.6 g, 2.16 mmol, 1.0 eq) in anhydrous THF (13 mL) under nitrogen atmosphere and ice bath, was added LiAlH4 (123 mg, 3.24 mmol, 1.5 eq) . The mixture was moved to room temperature after 10 minutes and stirred for 1 h. Then the reaction was moved to ice bath, saturated potassium sodium tartrate solution (10 mL) was added slowly into the mixture. After 30 minutes, the reaction was extracted three times with Et2O, then the organic phase was combined and washed by brine, dried over Na2SO4, and concentrated. The crude product (500 mg, 0.70 mmol, 1.0 eq) was dissolved in DCM (10 mL) , then DMAP (298 mg, 2.45 mmol, 3.5 eq) , succinic anhydride (140 mg, 1.40 mmol, 2.0 eq) were added therein under nitrogen atmosphere. The reaction mixture was stirred at room temperature overnight then H2O (10 mL) was added into the reaction, the mixture was extracted with DCM (3*10 mL) and the organic phase was combined, dried over Na2SO4, and concentrated. The resultant residue was purified with flash chromatography. The resultant residue was purified with flash chromatography (silica gel, gradient eluent: 1-10%of MeOH/DCM) to provide compound A103 (285 mg, 53%yield) as colorless oil. The product was characterized with mass spectrometry and 1H NMR. MW calc.: 818.49; MW. Found: 303.2 [DMT] -, 517.4 [DMT off + H] +. 1H NMR (400 MHz, CDCl3) δ 7.81 (s, 1H) , 7.41 (d, J = 7.6 Hz, 2H) , 7.30 (d, J = 8.8 Hz, 5H) , 7.25 -7.15 (m, 4H) , 6.80 (d, J = 8.8 Hz, 4H) , 5.26 (s, 2H) , 4.04 (d, J = 7.2 Hz, 2H) , 3.77 (s, 6H) , 3.11 (t, J = 6.1 Hz, 2H) , 2.90 (t, J = 7.7 Hz, 2H) , 2.69 (dd, J = 8.4, 4.5 Hz, 4H) , 2.02 -1.95 (m, 2H) , 1.74 (dd, J = 13.4, 6.9 Hz, 4H) , 1.28 -1.24 (m, 22H) , 0.87 (t, J = 6.7 Hz, 3H) .
(5) The preparation of compound A7
To a solution of the compound A103 (124 mg, 0.15 mmol, 1.0 eq) , Controlled Pore Glass (CPG) (2.66 g) , DIPEA (75 μL, 0.45 mmol, 3.0 eq) in acetonitrile (20 mL) was added HBTU (115 mg, 0.30 mmol, 2.0 eq) under nitrogen atmosphere. The reaction mixture was stirred at 25℃ overnight and then subsequently washed with DCM and ethyl ether to generate crude solid support. To a solution of acetic anhydride (9.06 mL) , pyridine (17.4 mL) , NEt3 (272 μL) in acetonitrile (11.6 mL) was added the crude solid support under nitrogen atmosphere. The reaction mixture was stirred at 25 ℃ overnight and then subsequently washed with DCM and ethyl ether to produce the Compound A7 of the present disclosure (2.4 g) .
Example 7. The preparation of compound A8 of the present disclosure
Compound A8 was prepared in this Example by using the following procedures.
(1) The preparation of compound A105
To a solution of methyl 4-fluoro-3-nitrobenzoate A104 (15 g, 75.3 mmol, 1.0 eq) and K2CO3 (10.4 g, 75.3 mmol, 1.0 eq) in anhydrous DMF (240 mL) , under nitrogen atmosphere, was added compound 69 (18.18 g, 75.3 mmol, 1.0 eq) . The reaction mixture was stirred at 25 ℃ for 6 h, then cold water (100 mL) was added. The mixture was extracted three times by ethyl acetate, then the organic phase was washed three times by saturated LiCl solution and one time by brine. Then dried by anhydrous Na2SO4 and concentrated under reduced pressure to form brown oil compound A105 which was directly used in the next step without further
purification.
(2) The preparation of compound A106
To a solution of compound A105 (31.65 g, 75.3 mmol, 1.0 eq) in THF/H2O (9∶1, 300 mL) , under ice bath, was added HCOONH4 (28.5 g, 451.8 mmol, 6.0 eq) and Zn powder (29.5 g, 451.8 mmol, 6.0 eq) . After 10 minutes, the reaction mixture was moved to room temperature and stirred overnight. Then the reaction mixture was filtered and concentrated under reduced pressure. After that, water (100 mL) was added into the mixture, then extracted three times by ethyl acetate, the organic phase was washed one time by brine. After dried by anhydrous Na2SO4 and concentrated under reduced pressure, the brown solid compound A106 formed then was directly used in the next step without further purification.
(3) The preparation of compound A107
To a solution of compound A106 (6.0 g, 15.4 mmol, 1.0 eq) in EtOH (90 mL) under nitrogen atmosphere, was added compound A101 (1.57 g, 15.4 mmol, 1.0 eq) and AcOH (3.54 mL, 61.6 mmol, 4.0 eq) . The reaction mixture was stirred at 80℃ overnight, and then concentrated under reduced pressure. Then a saturated NaHCO3 solution (80 mL) was added, the mixture was extracted three times by ethyl acetate, then the organic phase was combined and washed by brine, dried over Na2SO4, and concentrated. The resultant residue was directly used in the next step without further purification. The resultant residue was characterized with mass spectrometry. MW calc.: 472.37; MW. Found: 473.4 [M+H] +.
The resultant residue (1.0 g, 2.12 mol, 1.0 eq) was dissolved in 10 mL DCM, and Et3N (0.44 mL, 3.17 mmol, 1.5 eq) , DMTrCl (860 mg, 2.54 mmol, 1.2 eq) were added therein. The reaction mixture was stirred at room temperature for 6 h, after which it was concentrated under reduced pressure. The resultant residue was purified with flash chromatography (silica gel, gradient eluent: 1-5%of MeOH/DCM) to provide compound A107 (1.27 g, 77%yield) as
yellow solid. The product was characterized with mass spectrometry and 1H NMR. MW calc.: 774.50; MW. Found: 303.2 [DMT] -, 473.4 [DMT off + H] +. 1H NMR (400 MHz, CDCl3) δ 8.42 (d, J = 0.9 Hz, 1H) , 7.97 (dd, J = 8.5, 1.4 Hz, 1H) , 7.42 (d, J = 7.4 Hz, 2H) , 7.33 -7.28 (m, 5H) , 7.25 (d, J = 6.8 Hz, 2H) , 7.19 (t, J = 7.3 Hz, 1H) , 6.80 (dd, J = 9.4, 2.4 Hz, 4H) , 4.05 (t, J = 7.5 Hz, 2H) , 3.93 (s, 3H) , 3.78 (s, 6H) , 3.12 (t, J = 6.3 Hz, 2H) , 2.92 -2.80 (m, 2H) , 2.07 -1.97 (m, 2H) , 1.82 -1.71 (m, 4H) , 1.28 -1.24 (m, 26H) , 0.88 (t, J = 6.8 Hz, 3H) .
(4) The preparation of compound A108
To a solution of compound A107 (1.27 g, 1.64 mmol, 1.0 eq) in anhydrous THF (10 mL) under nitrogen atmosphere and ice bath, was added LiAlH4 (93 mg, 2.46 mmol, 1.5 eq) . The mixture was moved to room temperature after 10 minutes and stirred for 1 h. Then the reaction was moved to ice bath, saturated potassium sodium tartrate solution (10 mL) was added slowly into the mixture. After 30 minutes, the reaction was extracted three times with Et2O, then the organic phase was combined and washed by brine, dried over Na2SO4, and concentrated. The crude product (500 mg, 0.67 mmol, 1.0 eq) was dissolved in DCM (10 mL) , then DMAP (286 mg, 2.34 mmol, 3.5 eq) , succinic anhydride (134 mg, 1.34 mmol, 2.0 eq) were added therein under nitrogen atmosphere. The reaction mixture was stirred at room temperature overnight then H2O (10 mL) was added into the reaction, the mixture was extracted with DCM (3*10 mL) and the organic phase was combined, dried over Na2SO4, and concentrated. The resultant residue was purified with flash chromatography. The resultant residue was purified with flash chromatography (silica gel, gradient eluent: 1-10%of MeOH/DCM) to provide compound A108 (425 mg, 62%yield) as colorless oil. The product was characterized with mass spectrometry and 1H NMR. MW calc.: 846.52; MW. Found: 303.2 [DMT] -, 545.4 [DMT off + H] +. 1H NMR (400 MHz, CDCl3) δ 7.81 (s, 1H) , 7.42 (d, J = 7.5 Hz, 2H) , 7.30 (d, J = 8.8 Hz, 4H) , 7.25 (d, J = 7.6 Hz, 3H) , 7.19 (dd, J = 14.9, 7.9 Hz, 2H) , 6.80 (d, J = 8.8 Hz, 4H) , 5.26 (s, 2H) , 4.03 (t, J = 7.4 Hz, 2H) , 3.77 (s, 6H) , 3.11 (t, J = 6.1 Hz, 2H) , 2.90 (t, J = 7.7 Hz, 2H) , 2.73 -2.66 (m, 4H) , 1.99 (dt, J = 15.3, 7.7 Hz, 2H) , 1.74 (dd, J =
13.5, 6.9 Hz, 4H) , 1.29 -1.25 (m, 26H) , 0.87 (t, J = 6.7 Hz, 3H) .
(5) The preparation of compound A8
To a solution of the compound A108 (114 mg, 0.134 mmol, 1.0 eq) , Controlled Pore Glass (CPG) (2.36 g) , DIPEA (67 μL, 0.4 mmol, 3.0 eq) in acetonitrile (18 mL) was added HBTU (102 mg, 0.268 mmol, 2.0 eq) under nitrogen atmosphere. The reaction mixture was stirred at 25℃ overnight and then subsequently washed with DCM and ethyl ether to generate crude solid support. To a solution of acetic anhydride (8.04 mL) , pyridine (15.5 mL) , NEt3 (241 μL) in acetonitrile (10 mL) was added the crude solid support under nitrogen atmosphere. The reaction mixture was stirred at 25 ℃ overnight and then subsequently washed with DCM and ethyl ether to produce the Compound A8 of the present disclosure (2.2 g) .
Example 8. The preparation of compound A9 of the present disclosure
Compound A9 was prepared in this Example by using the following procedures.
(1) The preparation of compound A110
To a solution of methyl 4-fluoro-3-nitrobenzoate A69 (8.0 g, 40.2 mmol, 1.0 eq) and
K2CO3 (5.55 g, 40.2 mmol) in anhydrous DMF (128 mL) , under nitrogen atmosphere, was added compound A109 (10.83 g, 40.2 mmol, 1.0 eq) . The reaction mixture was stirred at 25 ℃ for 6 h, then cold water (100 mL) was added. The mixture was extracted three times by ethyl acetate, then the organic phase was washed three times by saturated LiCl solution and one time by brine. Then dried by anhydrous Na2SO4 and concentrated under reduced pressure to form brown oil compound A110 which was directly used in the next step without further purification.
(2) The preparation of compound A111
To a solution of compound A110 (13 g, 29 mmol, 1.0 eq) in THF/H2O (9∶1, 120 mL) , under ice bath, was added HCOONH4 (10.98 g, 174 mmol, 6.0 eq) and Zn powder (11.38 g, 174 mmol, 6.0 eq) . After 10 minutes, the reaction mixture was moved to room temperature and stirred overnight. Then the reaction mixture was filtered and concentrated under reduced pressure. After that, water (100 mL) was added into the mixture, then extracted three times by ethyl acetate, the organic phase was washed one time by brine. After dried by anhydrous Na2SO4 and concentrated under reduced pressure, the brown solid compound A111 formed then was directly used in the next step without further purification.
(3) The preparation of compound A112
To a solution of compound A111 (6.0 g, 14.34 mmol, 1.0 eq) in EtOH (80 mL) under nitrogen atmosphere, was added compound A101 (1.46 g, 14.34 mmol, 1.0 eq) and AcOH (3.3 mL, 57.36 mmol, 4.0 eq) . The reaction mixture was stirred at 80℃ overnight, and then concentrated under reduced pressure. Then a saturated NaHCO3 solution (80 mL) was added, the mixture was extracted three times by ethyl acetate, then the organic phase was combined and washed by brine, dried over Na2SO4, and concentrated. The resultant residue was directly used in the next step without further purification. The resultant residue was characterized with mass spectrometry. MW calc.: 500.40; MW. Found: 501.4 [M+H] +.
The resultant residue (1.5 g, 3.0 mol, 1.0 eq) was dissolved in 10 mL DCM, and Et3N
(0.625 mL, 4.5 mmol, 1.5 eq) , DMTrCl (1.2 g, 3.6 mmol, 1.2 eq) were added therein. The reaction mixture was stirred at room temperature for 6 h, after which it was concentrated under reduced pressure. The resultant residue compound was directly used in the next step without further purification. The product A112 was characterized with mass spectrometry. MW calc.: 802.53; MW. Found: 303.2 [DMT] -.
(4) The preparation of compound A113
To a solution of compound A112 (1.7 g, 2.12 mmol, 1.0 eq) in anhydrous THF (15 mL) under nitrogen atmosphere and ice bath, was added LiAlH4 (121 mg, 3.18 mmol, 1.5 eq) . The mixture was moved to room temperature after 10 minutes and stirred for 1 h. Then the reaction was moved to ice bath, saturated potassium sodium tartrate solution (10 mL) was added slowly into the mixture. After 30 minutes, the reaction was extracted three times with Et2O, then the organic phase was combined and washed by brine, dried over Na2SO4, and concentrated. The crude product (500 mg, 0.65 mmol, 1.0 eq) was dissolved in DCM (10 mL) , then DMAP (276 mg, 2.27 mmol, 3.5 eq) , succinic anhydride (129 mg, 1.29 mmol, 2.0 eq) were added therein under nitrogen atmosphere. The reaction mixture was stirred at room temperature overnight then H2O (10 mL) was added into the reaction, the mixture was extracted with DCM (3*10 mL) and the organic phase was combined, dried over Na2SO4, and concentrated. The resultant residue was purified with flash chromatography. The resultant residue was purified with flash chromatography (silica gel, gradient eluent: 1-10%of MeOH/DCM) to provide compound A113 (340 mg, 60%yield) as colorless oil. The product was characterized with mass spectrometry and 1H NMR. MW calc.: 874.55; MW. Found: 303.2 [DMT] -, 573.4 [DMT off + H] +. 1H NMR (400 MHz, CDCl3) δ 7.79 (s, 1H) , 7.42 (d, J = 7.5 Hz, 2H) , 7.30 (d, J = 8.8 Hz, 4H) , 7.26 -7.22 (m, 4H) , 7.17 (d, J = 8.0 Hz, 1H) , 6.81 (t, J = 5.8 Hz, 4H) , 5.26 (s, 2H) , 4.02 (t, J = 7.4 Hz, 2H) , 3.77 (s, 6H) , 3.11 (t, J = 6.1 Hz, 2H) , 2.88 (t, J = 7.7 Hz, 2H) , 2.69 (dt, J = 10.5, 5.3 Hz, 4H) , 1.97 (dd, J = 15.0, 7.6 Hz, 2H) , 1.74 (dd, J = 13.5, 6.9 Hz, 4H) , 1.28 -1.25 (m, 30H) , 0.87 (t, J = 6.8 Hz, 3H) .
(5) The preparation of compound A9
To a solution of the compound A113 (100 mg, 0.114 mmol, 1.0 eq) , Controlled Pore Glass (CPG) (2.0 g) , DIPEA (57 μL, 0.342 mmol, 3.0 eq) in acetonitrile (15 mL) was added HBTU (86 mg, 0.228 mmol, 2.0 eq) under nitrogen atmosphere. The reaction mixture was stirred at 25℃ overnight and then subsequently washed with DCM and ethyl ether to generate crude solid support. To a solution of acetic anhydride (6.8 mL) , pyridine (13 mL) , NEt3 (210 μL) in acetonitrile (9 mL) was added the crude solid support under nitrogen atmosphere. The reaction mixture was stirred at 25 ℃ overnight and then subsequently washed with DCM and ethyl ether to produce the compound A9 of the present disclosure (1.87 g) .
Example 9. The preparation of compound A10 and A13 of the present disclosure
Compounds A10 and A13 were prepared in this Example by using the following procedures.
(1) The preparation of compound A115
To a solution of compound A106 (2.0 g, 5.12 mmol, 1.0 eq) in EtOH (30 mL) under nitrogen atmosphere, was added compound A114 (790 mg, 5.12 mmol, 1.0 eq) and AcOH (1.18 mL, 20.48 mmol, 4.0 eq) . The reaction mixture was stirred at 80℃ overnight, and then concentrated under reduced pressure. Then a saturated NaHCO3 solution (20 mL) was added, the mixture was extracted three times by ethyl acetate, then the organic phase was combined and washed by brine, dried over Na2SO4, and concentrated. The resultant residue was purified with flash chromatography. The resultant residue was purified with flash chromatography (silica gel, gradient eluent: 1-25%of EA/Hexane) to provide compound A115 (1.46 g, 54%yield) as yellow oil. The product was characterized with mass spectrometry. MW calc.: 524.43; MW Found: 525.5 [M+H] +.
(2) The preparation of compound A116
To a solution of compound A115 (500 mg, 0.95 mmol, 1.0 eq) in MeOH (18 mL) at room temperature, was added 18 mL 0.5 M NaOH aqueous solution. The mixture was stirred overnight. Then the reaction was acidified by 2M HCl and extracted three times with Et2O, then the organic phase was combined and washed by brine, dried over Na2SO4, and concentrated. The organic layer was concentrated under reduced pressure and the resultant residue was purified with flash chromatography (silica gel, gradient eluent: 1-5%of MeOH/DCM) to provide compound A116 of the present disclosure (400 mg, 82%yield) as white solid. The product was characterized with mass spectrometry. MW calc.: 510.42; MW Found: 511.4 [M + H] +.
(3) The preparation of compound A117
To a solution of compound A94 (298 mg, 0.36 mmol, 1.0 eq) in MeOH (5 mL) , was added Pd/C (30 mg) under nitrogen atmosphere slowly. Then the reaction atmosphere was replaced with hydrogen three times. After that, the reaction mixture was stirred by purging with a hydrogen balloon at room temperature overnight. Then the reaction mixture was filtered and concentrated under reduced pressure to provide crude product A95. The crude product A95 was dissolved in DCM (5mL) , then compound A116 (220 mg, 0.432 mmol, 1.2 eq) , HBTU (273 mg, 0.72 mmol, 2.0 eq) and DIPEA (203 μL, 1.224 mmol, 3.4 eq) were added into the reaction mixture under nitrogen atmosphere. The reaction mixture was stirred overnight at room temperature. Then H2O (10 mL) was added into the reaction, the mixture was extracted with DCM (3*10 mL) . The organic phase was combined, dried over Na2SO4, and concentrated. The resultant residue was purified with flash chromatography (silica gel, gradient eluent: 1-5%of MeOH/DCM) to provide compound A117 (400 mg, 94%yield) as yellow oil. The product was characterized with mass spectrometry and 1H NMR. MW calc.: 1185.80; MW. Found: 303.2 [DMT] -. 1H NMR (400 MHz, CDCl3) δ 8.05 (d, J = 1.3 Hz, 1H) , 7.43 -7.40 (m, 2H) , 7.32 -7.26 (m, 8H) , 7.23 (dd, J = 11.0, 4.3 Hz, 3H) , 7.18 (dd, J = 7.9, 1.8 Hz, 1H) , 6.81 -6.78 (m, 4H) , 6.38 (t, J = 5.8 Hz, 1H) , 5.09 (t, J = 7.0 Hz, 1H) , 4.83 (s, 2H) , 4.18 (t, J = 7.3 Hz, 2H) , 4.11 -4.07 (m, 2H) , 3.76 (s, 6H) , 3.49 (dd, J = 13.0, 6.6 Hz, 2H) , 3.09 (t, J = 6.2 Hz, 2H) , 2.91 -2.83 (m, 4H) , 2.14 -2.01 (m, 5H) , 1.80 -1.73 (m, 4H) , 1.67 (s, 3H) , 1.59 (s, 3H) , 1.32 -1.25 (m, 30H) , 0.99 (d, J = 6.6 Hz, 3H) , 0.94 (s, 9H) , 0.87 (d, J = 4.6 Hz, 3H) , 0.10 (s, 6H) .
(4) The preparation of compound A118
To a solution of compound A117 (400 mg, 0.337 mmol, 1.0 eq) in anhydrous THF (8 mL) , under nitrogen atmosphere, was added 1M TBAF THF solution (2.02 mL, 2.02 mmol, 6.0 eq) . The reaction mixture was stirred at room temperature for 1 h, and then concentrated under reduced pressure to provide crude product. The crude product was dissolved in DCM (10 mL) , then DMAP (144 mg, 1.18 mmol, 3.5 eq) , succinic anhydride (67 mg, 0.674 mmol, 2.0 eq) were added therein under nitrogen atmosphere. The reaction mixture was stirred at room temperature overnight then H2O (10 mL) was added into the reaction, the mixture was extracted with DCM (3*20 mL) and the organic phase was combined, dried over Na2SO4, and concentrated. The resultant residue was purified with flash chromatography. The resultant residue was purified with flash chromatography (silica gel, gradient eluent: 1-10%of MeOH/DCM) to provide compound A118 (235 mg, 60%yield) as white solid. The product was characterized with mass spectrometry and 1H NMR. MW calc.: 1171.73; MW Found: 303.2 [DMT] -, 871.3 [DMT off + H] +. 1H NMR (400 MHz, CDCl3) δ 8.11 (s, 1H) , 7.65 (s, 1H) , 7.41 (d, J = 7.6 Hz, 2H) , 7.31 -7.24 (m, 9H) , 7.18 (dd, J = 7.9, 5.6 Hz, 2H) , 6.80 (d, J = 8.8 Hz, 4H) , 5.20 (s, 2H) , 5.07 (d, J = 6.3 Hz, 1H) , 4.13 (dt, J = 15.1, 7.1 Hz, 4H) , 3.76 (s, 6H) , 3.33 (dd, J = 11.3, 5.6 Hz, 4H) , 3.09 (t, J = 6.2 Hz, 2H) , 2.83 (t, J = 7.6 Hz, 2H) , 2.72 -2.68 (m, 4H) , 2.15 -2.04 (m, 5H) , 1.76 (dd, J = 15.1, 7.9 Hz, 4H) , 1.67 (s, 3H) , 1.60 (s, 3H) , 1.31 -1.26 (m, 30H) , 0.96 (s, 3H) , 0.87 (d, J = 7.0 Hz, 3H) .
(5) The preparation of compound A10
To a solution of the compound A118 (170 mg, 0.145 mmol, 1.0 eq) , Controlled Pore Glass (CPG) (2.54 g) , DIPEA (72 μL, 0.435 mmol, 3.0 eq) in acetonitrile (15 mL) was added HBTU (110 mg, 0.29 mmol, 2.0 eq) under nitrogen atmosphere. The reaction mixture was stirred at 25℃ overnight and then subsequently washed with DCM and ethyl ether to generate crude solid support. To a solution of acetic anhydride (8.67 mL) , pyridine (16.8 mL) , NEt3 (260 μL) in acetonitrile (11 mL) was added the crude solid support under nitrogen atmosphere. The reaction mixture was stirred at 25 ℃ overnight and then subsequently washed with DCM and ethyl ether to produce the Compound A10 of the present disclosure (2.38 g) .
(6) The preparation of compound A13
To a solution of compound A116 (400 mg, 0.76 mmol, 1.0 eq) in anhydrous THF (5 mL) under nitrogen atmosphere and ice bath, was added LiAlH4 (43 mg, 1.14 mmol, 1.5 eq) . The mixture was moved to room temperature after 10 minutes and stirred for 1 h. Then the reaction was moved to ice bath, saturated potassium sodium tartrate solution (10 mL) was added slowly into the mixture. After 30 minutes, the reaction was extracted three times with Et2O, then the organic phase was combined and washed by brine, dried over Na2SO4, and concentrated to generate crude product. To a solution of crude compound and DIPEA (378 μL, 2.28 mmol, 3.0 eq) in anhydrous DCM (5 mL) under nitrogen atmosphere, was added 3- ( (chloro (diisopropylamino) phosphanyl) oxy) propanenitrile (540 mg, 2.28 mmol, 3.0 eq. ) at 25℃. The reaction mixture was stirred for 1 h. The mixture was extracted two times with DCM, then washed with brine and dried with anhydrous Na2SO4. The organic layer was
concentrated under reduced pressure and the resultant residue was purified with flash chromatography (silica gel, gradient eluent: 1-5%of MeOH/DCM, 1%NEt3) to provide compound A13 (397 mg, 75%yield) as colorless oil. The product was characterized with mass spectrometry and 1H NMR. MW calc.: 696.55; MW Found: 614.4 [M -diisopropylamine + 2H] +. 1H NMR (400 MHz, CDCl3) δ 7.68 (s, 1H) , 7.25 (dd, J = 8.6, 3.9 Hz, 2H) , 4.85 (dd, J = 11.9, 8.3 Hz, 1H) , 4.75 (dd, J = 11.9, 9.0 Hz, 1H) , 4.10 -4.04 (m, 2H) , 3.90 -3.78 (m, 2H) , 3.73 -3.60 (m, 2H) , 2.73 (d, J = 7.2 Hz, 2H) , 2.61 (td, J = 6.5, 1.4 Hz, 2H) , 1.98 -1.92 (m, 1H) , 1.88 -1.52 (m, 8H) , 1.36 -1.23 (m, 31H) , 1.21 (dd, J = 6.8, 0.7 Hz, 12H) , 1.15 -0.98 (m, 3H) , 0.88 (t, J = 6.8 Hz, 3H) .
Example 10. The preparation of compounds A11 and A15 of the present disclosure
Compounds A11 and A15 were prepared in this Example by using the following procedures.
(1) The preparation of compound A120
To a solution of compound A106 (2.0 g, 5.12 mmol, 1.0 eq) in EtOH (30 mL) under nitrogen atmosphere, was added compound A119 (958 mg, 5.12 mmol, 1.0 eq) and AcOH
(1.18 mL, 20.48 mmol, 4.0 eq) . The reaction mixture was stirred at 80℃ overnight, and then concentrated under reduced pressure. Then a saturated NaHCO3 solution (30 mL) was added, the mixture was extracted three times by ethyl acetate, then the organic phase was combined and washed by brine, dried over Na2SO4, and concentrated. The resultant residue compound A120 was directly used in the next step without further purification. The compound A120 was characterized with mass spectrometry. MW calc.: 557.36; MW. Found: 558.4 [M + H] +.
(2) The preparation of compound A122
To a solution of compound A121 (500 mg, 0.9 mmol, 1.0 eq) in MeOH (18 mL) at room temperature, was added 18 mL 0.5 M NaOH aqueous solution. The mixture was stirred overnight. Then the reaction was acidified by 2M HCl and extracted three times with Et2O, then the organic phase was combined and washed by brine, dried over Na2SO4, and concentrated. The organic layer was concentrated under reduced pressure and the resultant residue was purified with flash chromatography (silica gel, gradient eluent: 1-5%of MeOH/DCM) to provide compound A122 of the present disclosure (330 mg, 68%yield) as white solid. The product was characterized by mass spectrometry. MW calc.: 543.35; MW. Found: 544.5 [M + H] +.
(3) The preparation of compound A123
To a solution of compound A94 (273 mg, 0.33 mmol, 1.0 eq) in MeOH (5 mL) , was added Pd/C (27 mg) under nitrogen atmosphere slowly. Then the reaction atmosphere was replaced with hydrogen three times. After that, the reaction mixture was stirred by purging with a hydrogen balloon at room temperature overnight. Then the reaction mixture was filtered and concentrated under reduced pressure to provide crude product A95. The crude product
A95 was dissolved in DCM (5mL) , then compound A122 (229 mg, 0.33 mmol, 1.0 eq) , HBTU (250 mg, 0.66 mmol, 2.0 eq) and DIPEA (186 μL, 1.12 mmol, 3.4 eq) were added into the reaction mixture under nitrogen atmosphere. The reaction mixture was stirred overnight at room temperature. Then H2O (5 mL) was added into the reaction, the mixture was extracted with DCM (3*10 mL) . The organic phase was combined, dried over Na2SO4, and concentrated. The resultant residue was purified with flash chromatography (silica gel, gradient eluent: 1-5%of MeOH/DCM) to provide compound A123 (249 mg, 62%yield) as white solid. The product was characterized with mass spectrometry and 1H NMR. MW calc.: 1218.73; MW Found: 303.2 [DMT] -. 1H NMR (400 MHz, CDCl3) δ 9.19 (s, 1H) , 8.28 (s, 1H) , 7.73 -7.69 (m, 2H) , 7.56 (s, 1H) , 7.29 (d, J = 7.0 Hz, 3H) , 7.18 -7.15 (m, 6H) , 7.07 (dt, J = 9.2, 8.5 Hz, 6H) , 6.67 (d, J = 8.9 Hz, 4H) , 4.72 (s, 2H) , 4.12 -4.05 (m, 2H) , 3.96 (t, J = 7.0 Hz, 2H) , 3.64 (s, 6H) , 3.12 (d, J = 6.4 Hz, 2H) , 2.95 (t, J = 6.3 Hz, 2H) , 2.70 -2.63 (m, 7H) , 1.82 (dd, J = 17.3, 10.5 Hz, 4H) , 1.59 (dd, J = 12.8, 6.9 Hz, 4H) , 1.18 -1.04 (m, 24H) , 0.83 (s, 9H) , 0.75 (d, J = 7.0 Hz, 3H) , 0.03 (s, 6H) .
(4) The preparation of compound A124
To a solution of compound A123 (230 mg, 0.19 mmol, 1.0 eq) in anhydrous THF (5 mL) , under nitrogen atmosphere, was added 1M TBAF THF solution (1.13 mL, 1.13 mmol, 6.0 eq) . The reaction mixture was stirred at room temperature for 1 h, and then concentrated under reduced pressure to provide crude product. The crude product was dissolved in DCM (6 mL) , then DMAP (81 mg, 0.66 mmol, 3.5 eq) , succinic anhydride (38 mg, 0.38 mmol, 2.0 eq) were added therein under nitrogen atmosphere. The reaction mixture was stirred at room temperature overnight then H2O (10 mL) was added into the reaction, the mixture was extracted with DCM (3*10 mL) and the organic phase was combined, dried over Na2SO4, and
concentrated. The resultant residue was purified with flash chromatography. The resultant residue was purified with flash chromatography (silica gel, gradient eluent: 1-10%of MeOH/DCM) to provide compound A124 (126 mg, 55%yield) as colorless oil. The product was characterized with mass spectrometry and 1H NMR. MW calc.: 1204.66; MW Found: 303.2 [DMT] -. 1H NMR (400 MHz, CDCl3) δ 8.55 (s, 1H) , 8.07 (d, J = 8.2 Hz, 2H) , 7.73 (d, J = 8.1 Hz, 1H) , 7.63 (d, J = 10.8 Hz, 2H) , 7.38 (d, J = 11.5 Hz, 4H) , 7.28 (t, J = 8.1 Hz, 6H) , 7.16 (t, J = 6.8 Hz, 4H) , 6.79 (d, J = 8.7 Hz, 4H) , 5.16 (s, 2H) , 4.30 -4.20 (m, 4H) , 3.75 (s, 6H) , 3.58 (d, J = 5.1 Hz, 2H) , 3.43 -3.41 (m, 7H) , 3.07 (s, 2H) , 2.86 (t, J = 7.2 Hz, 2H) , 2.61 (d, J = 3.4 Hz, 4H) , 2.21 -2.15 (m, 2H) , 1.98 -1.93 (m, 2H) , 1.27 -1.20 (m, 26H) , 0.86 (d, J = 7.0 Hz, 3H) .
(5) The preparation of compound A11
To a solution of the compound A124 (126 mg, 0.104 mmol, 1.0 eq) , Controlled Pore Glass (CPG) (1.83 g) , DIPEA (52 μL, 0.312 mmol, 3.0 eq) in acetonitrile (12 mL) was added HBTU (79 mg, 0.208 mmol, 2.0 eq) under nitrogen atmosphere. The reaction mixture was stirred at 25℃ overnight and then subsequently washed with DCM and ethyl ether to generate crude solid support. To a solution of acetic anhydride (6.2 mL) , pyridine (12 mL) , NEt3 (186 μL) in acetonitrile (7.9 mL) was added the crude solid support under nitrogen atmosphere. The reaction mixture was stirred at 25 ℃ overnight and then subsequently washed with DCM and ethyl ether to produce the Compound A11 of the present disclosure (1.7 g) .
(6) The preparation of compound A15
To a solution of compound A121 (300 mg, 0.538 mmol, 1.0 eq) in anhydrous THF (5 mL) under nitrogen atmosphere and ice bath, was added LiAlH4 (31 mg, 0.8 mmol, 1.5 eq) . The mixture was moved to room temperature after 10 minutes and stirred for 1 h. Then the reaction was moved to ice bath, saturated potassium sodium tartrate solution (5 mL) was added slowly into the mixture. After 30 minutes, the reaction was extracted three times with Et2O, then the organic phase was combined and washed by brine, dried over Na2SO4, and concentrated to generate crude product. To a solution of crude compound and DIPEA (265 μL, 1.6 mmol, 3.0 eq) in anhydrous DCM (5 mL) under nitrogen atmosphere, was added 3- ( (chloro (diisopropylamino) phosphanyl) oxy) propanenitrile (379 mg, 1.6 mmol, 3.0 eq. ) at 25℃. The reaction mixture was stirred for 1 h. The mixture was extracted two times with DCM, then washed with brine and dried with anhydrous Na2SO4. The organic layer was concentrated under reduced pressure and the resultant residue was purified with flash chromatography (silica gel, gradient eluent: 1-5%of MeOH/DCM, 1%NEt3) to provide compound A15 (274 mg, 70%yield) as colorless oil. The product was characterized with mass spectrometry and 1H NMR. MW calc.: 729.47; MW Found: 669.5 [M -diisopropylamine + Na] +. 1H NMR (400 MHz, CDCl3) δ 8.28 (s, 1H) , 7.73 -7.68 (m, 2H) , 7.56 (s, 1H) , 7.18 -7.15 (m, 4H) , 4.72 (s, 2H) , 4.12 -4.05 (m, 4H) , 2.70 -2.63 (m, 7H) , 1.92 -1.88 (m, 2H) , 1.27 -1.23 (m, 26H) , 1.21 (dd, J = 6.8, 0.7 Hz, 12H) , 0.88 (t, J = 6.8 Hz, 3H) .
Example 11. The preparation of compound A12 of the present disclosure
Compound A12 was prepared in this Example by using the following procedures.
(1) The preparation of compound A126
To a solution of compound A94 (298 mg, 0.36 mmol, 1.0 eq) in MeOH (5 mL) , was added Pd/C (30 mg) under nitrogen atmosphere slowly. Then the reaction atmosphere was replaced with hydrogen three times. After that, the reaction mixture was stirred by purging with a hydrogen balloon at room temperature overnight. Then the reaction mixture was filtered and concentrated under reduced pressure to provide crude product A95. The crude product A95 was dissolved in DCM (5mL) , then compound A125 (111 mg, 0.43 mmol, 1.2 eq) , HBTU (273 mg, 0.72 mmol, 2.0 eq) and DIPEA (203 μL, 1.22 mmol, 3.4 eq) were added into the reaction mixture under nitrogen atmosphere. The reaction mixture was stirred overnight at room temperature. Then H2O (5 mL) was added into the reaction, the mixture was extracted with DCM (3*10 mL) . The organic phase was combined, dried over Na2SO4, and concentrated. The resultant residue was purified with flash chromatography (silica gel, gradient eluent: 1-5%of MeOH/DCM) to provide compound A126 (295 mg, 88%yield) as yellow oil. The product was characterized with mass spectrometry and 1H NMR. MW calc.: 931.63; MW Found: 303.2 [DMT] -. 1H NMR (400 MHz, CDCl3) δ 7.68 (s, 1H) , 7.47 -7.39 (m, 2H) , 7.32 -7.23 (m, 9H) , 6.83 -6.77 (m, 4H) , 4.83 (s, 2H) , 4.10 (t, J = 7.3 Hz, 2H) , 3.78 (s, 6H) , 3.24 (dd, J = 13.2, 6.7 Hz, 2H) , 3.11 (t, J = 6.2 Hz, 2H) , 2.06 (dd, J = 12.4, 4.5 Hz, 2H) , 2.02 -1.93 (m, 4H) , 1.79 -1.72 (m, 2H) , 1.56 (d, J = 7.0 Hz, 2H) , 1.29 -1.23 (m, 26H) , 0.94 (s, 9H) , 0.87 (d, J = 7.0 Hz, 3H) , 0.10 (s, 6H) .
(2) The preparation of compound A127
To a solution of compound A126 (270 mg, 0.29 mmol, 1.0 eq) in anhydrous THF (6 mL) , under nitrogen atmosphere, was added 1M TBAF THF solution (1.74 mL, 1.74 mmol, 6.0 eq) . The reaction mixture was stirred at room temperature for 1 h, and then concentrated under reduced pressure to provide crude product. The crude product was dissolved in DCM (10 mL) , then DMAP (124 mg, 1.02 mmol, 3.5 eq) , succinic anhydride (58 mg, 0.58 mmol, 2.0 eq) were added therein under nitrogen atmosphere. The reaction mixture was stirred at room temperature overnight then H2O (10 mL) was added into the reaction, the mixture was extracted with DCM (3*10 mL) and the organic phase was combined, dried over Na2SO4, and concentrated. The resultant residue was purified with flash chromatography. The resultant residue was purified with flash chromatography (silica gel, gradient eluent: 1-10%of MeOH/DCM) to provide compound A127 (120 mg, 45%yield) as colorless oil. The product was characterized with mass spectrometry and 1H NMR. MW calc.: 917.56; MW Found: 303.2 [DMT] -. 1H NMR (400 MHz, CDCl3) δ 7.64 (s, 1H) , 7.46 -7.40 (m, 3H) , 7.31 (d, J = 8.8 Hz, 4H) , 7.25 (d, J = 7.8 Hz, 2H) , 7.20 (t, J = 7.7 Hz, 2H) , 6.80 (d, J = 8.8 Hz, 4H) , 5.18 (s, 2H) , 4.31 -4.18 (m, 2H) , 3.78 (s, 6H) , 3.42 -3.40 (m, 4H) , 3.09 (t, J = 6.3 Hz, 2H) , 2.87 (t, J = 7.5 Hz, 2H) , 2.67 -2.62 (m, 4H) , 2.34 (t, J = 7.6 Hz, 2H) , 2.05 -1.92 (m, 4H) , 1.29 -1.23 (m, 26H) , 0.88 (t, J = 6.8 Hz, 3H) .
(3) The preparation of compound A12
To a solution of the compound A127 (120 mg, 0.13 mmol, 1.0 eq) , Controlled Pore Glass (CPG) (2.29 g) , DIPEA (65 μL, 0.39 mmol, 3.0 eq) in acetonitrile (15 mL) was added
HBTU (99 mg, 0.26 mmol, 2.0 eq) under nitrogen atmosphere. The reaction mixture was stirred at 25℃ overnight and then subsequently washed with DCM and ethyl ether to generate crude solid support. To a solution of acetic anhydride (7.8 mL) , pyridine (15 mL) , NEt3 (234 μL) in acetonitrile (10 mL) was added the crude solid support under nitrogen atmosphere. The reaction mixture was stirred at 25 ℃ overnight and then subsequently washed with DCM and ethyl ether to produce the Compound A12 of the present disclosure (2.1 g) .
Example 12. The preparation of compound A14 of the present disclosure
Compound A14 was prepared in this Example by using the following procedures.
(1) The preparation of compound A129.
To a solution of compound A106 (1.55 g, 3.96 mmol, 1.0 eq) in EtOH (20 mL) under nitrogen atmosphere, was added compound A128 (500 mg, 3.96 mmol, 1.0 eq) and AcOH (913 μL, 15.84 mmol, 4.0 eq) . The reaction mixture was stirred at 80℃ overnight, and then concentrated under reduced pressure. Then a saturated NaHCO3 solution (20 mL) was added, the mixture was extracted three times by ethyl acetate, then the organic phase was combined and washed by brine, dried over Na2SO4, and concentrated. The resultant residue compound A129 was directly used in the next step without further purification. The compound A129 was characterized with mass spectrometry. MW calc.: 496.40; MW. Found: 497.5 [M+H] +.
(2) The preparation of compound A14
To a solution of compound A129 (400 mg, 0.806 mmol, 1.0 eq) in anhydrous THF (5 mL) under nitrogen atmosphere and ice bath, was added LiAlH4 (46 mg, 1.2 mmol, 1.5 eq) . The mixture was moved to room temperature after 10 minutes and stirred for 1 h. Then the reaction was moved to ice bath, saturated potassium sodium tartrate solution (5 mL) was added slowly into the mixture. After 30 minutes, the reaction was extracted three times with Et2O, then the organic phase was combined and washed by brine, dried over Na2SO4, and concentrated to generate crude product. To a solution of crude compound and DIPEA (401 μL, 2.42 mmol, 3.0 eq) in anhydrous DCM (5 mL) under nitrogen atmosphere, was added 3- ( (chloro (diisopropylamino) phosphanyl) oxy) propanenitrile (573 mg, 2.42 mmol, 3.0 eq. ) at 25℃. The reaction mixture was stirred for 1 h. The mixture was extracted two times with DCM, then washed with brine and dried with anhydrous Na2SO4. The organic layer was concentrated under reduced pressure and the resultant residue was purified with flash chromatography (silica gel, gradient eluent: 1-3%of MeOH/DCM, 1%NEt3) to provide compound A14 (339 mg, 63%yield) as colorless oil. The product was characterized with mass spectrometry and 1H NMR. MW calc.: 668.52; MW Found: 607.4 [M -diisopropylamine + Na] +. 1H NMR (400 MHz, CDCl3) δ 7.62 (s, 1H) , 7.19 (dd, J = 6.9, 2.8 Hz, 2H) , 5.03 (t, J = 7.0 Hz, 1H) , 4.81 -4.79 (m, 1H) , 4.71 -4.66 (m, 1H) , 4.04 -3.98 (m, 2H) , 3.79 (dd, J = 6.9, 2.8 Hz, 1H) , 3.60 (ddd, J = 13.6, 6.8, 3.4 Hz, 2H) , 2.82 -2.78 (m, 1H) , 2.61 -2.53 (m, 3H) , 1.63 -1.60 (m, 3H) , 1.54 -1.52 (m, 3H) , 1.33 -1.17 (m, 30H) , 1.14 (dd, J = 6.8, 2.0 Hz, 12H) , 0.92 (d, J = 6.6 Hz, 3H) , 0.82 (t, J = 6.8 Hz, 3H) .
Example 13. The preparation of compound B1 of the present disclosure
Compound B1 was prepared in this Example by using the following procedures.
(1) Preparation of compound B12 from 4-fluoro-3-nitrobenzoic acid.
To a solution of 4-fluoro-3-nitrobenzoic acid (35 g, 189.1 mmol, 1.0 eq) and Na2CO3 (30.6 g, 283.6 mmol, 1.5 eq) in anhydrous DMF (500 mL) under nitrogen atmosphere, was added benzyl bromide (35.6 g, 207.98 mmol, 1.1 eq) slowly. The reaction mixture was stirred at 45℃ for 3 h, then cold water (200 mL) was added therein. The mixture was extracted 3 times with ethyl acetate, then the organic phase was washed 3 times with saturated LiCl solution and 1 time with brine. Then the organic phase was dried with anhydrous Na2SO4 and concentrated under reduced pressure to form a yellow oil (compound B12) which was directly used in the next step without further purification.
(2) Preparation of compound B13 from compound B12 by SNAr reaction of tert-butyl (3-aminopropyl) carbamate.
To a solution of compound B12 (52 g, 189.1 mmol, 1.0 eq) and K2CO3 (26.13 g, 189.1 mmol, 1.0 eq) in anhydrous DMF (500 mL) under nitrogen atmosphere, was added tert-butyl (3-aminopropyl) carbamate (32.9 g, 189.1 mmol, 1.0 eq) . The reaction mixture was stirred at room temperature for 3 h, then cold water (200 mL) was added. The mixture was extracted 3 times by ethyl acetate, then the organic phase was washed 3 times by saturated LiCl solution and 1 time by brine. After dried by anhydrous Na2SO4 and concentrated under reduced pressure, the yellow solid B13 was formed and then directly used in the next step without further purification.
(3) Compound B14 was generated by reduction of compound B13.
To a solution of compound B13 (81.16 g, 189.1 mmol, 1.0 eq) in THF/H2O (9: 1, 666 mL) under ice bath, was added HCOONH4 (71.58 g, 1.134 mol, 6.0 eq) and Zn powder (74.19 g, 1.134 mol, 6.0 eq) . After 10 minutes, the reaction mixture was moved to room temperature and stirred overnight. Then the reaction mixture was filtered and concentrated under reduced pressure. After that, water (200 mL) was added into the mixture, then the mixture was extracted 3 times with ethyl acetate, the organic phase was washed 1 time with brine. The organic brine was dried with anhydrous NaSO4 and concentrated under reduced pressure, a light red solid (compound B14) was formed and was directly used in the next step without further purification.
(4) Compound B15 was generated by acylation of compound B14 with N2, N6-bis (tert-butoxycarbonyl) -L-lysine.
To a solution of compound B14 (16.8 g, 42 mmol, 1.0 eq) in DCM (300 mL) , was added N2, N6-bis (tert-butoxycarbonyl) -L-lysine (21.82 g, 63 mmol, 1.5 eq) , EDCI (12.08 g, 63 mmol, 1.5 eq) and DMAP (2.565 g, 21 mmol, 0.5 eq) under nitrogen atmosphere. The reaction mixture was stirred overnight at room temperature. Then 200 mL H2O was added into the reaction mixture, and the mixture was extracted with DCM (3*100 mL) . The organic phase was combined, dried over Na2SO4, and concentrated. The resultant residue was purified by using flash chromatography (silica gel, gradient eluent: 1-50%of EA/Hexane) to provide compound 15 (27.5 g, 90%yield) as yellow solid. The product was characterized with mass spectrometry and 1H NMR. MW calc.: 727.42; MW Found: 728.47 [M+H] +. 1H NMR (400 MHz, CDCl3) δ 7.88 (dd, J = 8.6, 1.9 Hz, 1H) , 7.79 (s, 1H) , 7.35 (ddt, J = 9.8, 7.1, 5.5 Hz, 5H) , 6.62 (d, J = 8.6 Hz, 1H) , 5.29 (s, 2H) , 4.16 (s, 1H) , 3.37 -2.97 (m, 6H) , 1.78 (d, J = 25.4 Hz, 4H) , 1.51 (s, 2H) , 1.48 -1.35 (m, 27H) , 1.36 -1.13 (m, 2H) .
(5) The compound B16, which comprises a benzimidazole structure was prepared from compound B15.
The compound B15 (20 g, 27.5 mmol, 1.0 eq) was added into AcOH (100 mL) under nitrogen atmosphere. The reaction mixture was stirred at 95℃ for 4 h. Then saturated NaHCO3 solution (100 mL) was added therein, the mixture was extracted 3 times with ethyl acetate, then the organic phase was combined and washed with saturated NaHCO3 solution (3*100 mL) , dried over Na2SO4, and concentrated. The resultant residue was purified by flash chromatography (silica gel, gradient eluent: 1-50%of EA/Hexane) to provide compound B16 (8.78 g, 45%yield) as white solid. The product was characterized with mass spectrometry and
1H NMR. MW calc.: 709.41; MW Found: 710.57 [M+H] +. 1H NMR (400 MHz, CDCl3) δ 8.50 (s, 1H) , 8.04 (dd, J = 8.5, 1.4 Hz, 1H) , 7.46 (d, J = 7.0 Hz, 2H) , 7.41 (s, 1H) , 7.38 -7.34 (m, 3H) , 5.39 (s, 2H) , 5.02 (dd, J = 15.8, 7.7 Hz, 1H) , 3.20 -3.10 (m, 6H) , 2.08 -1.96 (m, 4H) , 1.57 -1.48 (m, 4H) , 1.41 (d, J = 8.6 Hz, 27H) .
(6) The compound B17 was prepared by using the start material of (2S, 3R, 4R, 5R, 6R) -3-acetamido-6- (acetoxymethyl) tetrahydro-2H-pyran-2, 4, 5-triyl triacetate.
To a solution of (2S, 3R, 4R, 5R, 6R) -3-acetamido-6- (acetoxymethyl) tetrahydro-2H-pyran-2, 4, 5-triyl triacetate (30 g, 77 mmol, 1.0 eq) in DCM (300 mL) , was added TMSOTf (15.3 mL, 84.7 mmol, 1.1 eq) under nitrogen atmosphere. The reaction mixture was stirred at room temperature for 6 h. Then NaHCO3 solution (21 g NaHCO3 in 200 mL water) was added under ice bath and the reaction was transferred to room temperature. After 30 minutes of stirring, the mixture was extracted 3 times with DCM, then the organic phase was combined and washed with brine, dried over Na2SO4, and concentrated. The resultant residue was purified with flash chromatography (silica gel, gradient eluent: 1-50%of EA/Hexane) to provide compound B17 (19.8 g, 78%yield) . The product was characterized with mass spectrometry and 1H NMR. MW calc.: 329.11; MW Found: 330.33 [M+H] +. 1H NMR (400 MHz, CDCl3) δ 5.97 (d, J = 9.5 Hz, 1H) , 5.39 (s, 1H) , 5.26 (dd, J = 11.4, 3.2 Hz, 1H) , 4.53 (td, J = 11.4, 3.4 Hz, 1H) , 4.43 (t, J = 6.5 Hz, 1H) , 4.27 (s, 1H) , 4.11 -4.08 (m, 1H) , 2.17 (s, 3H) , 2.05 (s, 3H) , 2.00 (d, J = 7.3 Hz, 6H) .
(7) An alkoxy group terminated with vinyl group was attached to compound B17.
To a solution of compound B17 (15 g, 45.6 mmol, 1.0 eq) in DCE (200 mL) , was added dec-9-en-1-ol (8.5 g, 54.7 mmol, 1.2 eq) and TMSOTf (1.6 mL, 9.1 mmol, 0.2 eq) under nitrogen atmosphere. The reaction mixture was stirred at room temperature for 6 h. Then
saturated NaHCO3 solution (100 mL) was added therein, the mixture was extracted 3 times with DCM, then the organic phase was combined and washed with brine, dried over Na2SO4, and concentrated. The resultant residue was purified with flash chromatography (silica gel, gradient eluent: 1-5%of MeOH/DCM) to provide compound B20 (19.6 g, 89%yield) . The product was characterized with mass spectrometry and 1H NMR. MW calc.: 485.26; MW Found: 486.48 [M+H] +. 1H NMR (400 MHz, CDCl3) δ 5.84 -5.74 (m, 1H) , 5.49 (d, J = 8.5 Hz, 1H) , 5.35 (d, J = 2.7 Hz, 1H) , 5.33 -5.25 (m, 2H) , 5.01 -4.88 (m, 2H) , 4.70 (d, J = 8.4 Hz, 1H) , 4.20 -4.07 (m, 2H) , 3.94 -3.80 (m, 3H) , 3.47 (dt, J = 9.5, 6.9 Hz, 1H) , 2.13 (s, 3H) , 2.03 (s, 3H) , 1.98 (d, J = 9.7 Hz, 3H) , 1.95 (d, J = 10.1 Hz, 3H) , 1.56 (d, J = 6.4 Hz, 2H) , 1.35 (d, J = 6.7 Hz, 2H) , 1.27 (s, 8H) .
(8) The terminal vinyl group of compound B20 was oxidized to a carboxyl group.
To a solution of the compound B20 (19.6 g, 40.4 mmol, 1.0 eq) in ACN/DCM/H2O (50mL/50mL/60mL) under ice bath, was added NaIO4 (34.6 g, 161.6 mol, 4.0 eq) and RuCl3·3H2O (1.66 g, 8.0 mol, 0.2 eq) . After 10 minutes, the reaction mixture was transferred to room temperature and stirred overnight. Then the reaction mixture was filtered and concentrated under reduced pressure. After that, water (50 mL) was added into the mixture, then the mixture extracted 3 times with DCM, the organic phase was washed 1 time with brine. After being dried with anhydrous Na2SO4 and concentrated under reduced pressure, the resultant residue was purified with flash chromatography (silica gel, gradient eluent: 1-8%of MeOH/DCM) to provide compound B21 (17 g, 84%yield) . The product was characterized with mass spectrometry and 1H NMR. MW calc.: 503.24; MW Found: 504.49 [M+H] +. 1H NMR (400 MHz, CDCl3) δ 5.65 (d, J = 8.7 Hz, 1H) , 5.35 (d, J = 2.8 Hz, 1H) , 5.31 (d, J = 6.1 Hz, 1H) , 4.68 (d, J = 8.3 Hz, 1H) , 4.18 -4.12 (m, 2H) , 3.91 (dd, J = 11.8, 5.4 Hz, 2H) , 3.48 (dd, J = 9.7, 6.7 Hz, 1H) , 2.35 (t, J = 7.2 Hz, 2H) , 2.15 (s, 3H) , 2.05 (s, 3H) , 2.01 (s, 3H) , 1.96 (s, 3H) , 1.63 (s, 2H) , 1.32 (s, 10H) .
(9) The compounds B16 and B21 were linked together.
To a solution of compound B16 (4.56 g, 6.4 mmol, 1.0 eq) in DCM (10 mL) , was added HCl/Dioxane (4M, 30 mL) . The reaction mixture was stirred for 3 h at room temperature, then concentrated under reduced pressure to provide a crude product as yellow solid. The crude product was dissolved in 50 mL DCM, then compound 21 (9.66 g, 19.2 mmol, 3.0 eq) , HBTU (8.49 g, 22.4 mmol, 3.5 eq) and DIPEA (12.72 mL, 76.8 mmol, 12.0 eq) were added into the reaction mixture under nitrogen atmosphere. The reaction mixture was stirred overnight at room temperature. Then 50 mL H2O was added into the reaction mixture, the mixture was extracted with DCM (3*50 mL) . The organic phase was combined, dried over Na2SO4, and concentrated. The resultant residue was purified with flash chromatography (silica gel, gradient eluent: 1-8%of MeOH/DCM) to provide compound B22 (10.4 g, 87%yield) as yellow solid. The product was characterized with mass spectrometry and 1H NMR.
MW calc.: 1864.93; MW Found: 1895.54 [M+H] +. 1H NMR (400 MHz, CDCl3) δ 8.45 (s, 1H) , 8.04 (dd, J = 8.5, 1.4 Hz, 1H) , 7.46 (d, J = 7.1 Hz, 2H) , 7.36 (dd, J = 13.2, 7.3 Hz, 4H) , 5.39 (s, 2H) , 5.35 (d, J = 3.2 Hz, 4H) , 5.29 (s, 6H) , 4.75 J 4.63 (m, 4H) , 4.14 (qd, J = 11.1, 6.8 Hz, 9H) , 3.92 (dd, J = 13.0, 6.4 Hz, 6H) , 3.64 (dd, J = 13.0, 6.6 Hz, 4H) , 3.49 -3.42 (m, 4H) , 3.12 -3.06 (m, 3H) , 2.19 (dd, J = 11.6, 7.5 Hz, 6H) , 2.13 (s, 9H) , 2.03 (s, 9H) , 1.99 (s, 9H) , 1.93 (d, J = 2.6 Hz, 9H) , 1.54 -1.50 (m, 12H) , 1.45 (s, 6H) , 1.25 (s, 18H) .
(10) The compound B22 was reduced to produce compound B1 of the present disclosure.
To a solution of the compound B22 (1.4 g, 0.75 mmol, 1.0 eq) in MeOH (25 mL) , was added Pd/C (100 mg) under nitrogen atmosphere slowly. Then the reaction atmosphere was replaced with hydrogen gas for three times. After that, the reaction mixture was stirred by purging with a hydrogen balloon at room temperature overnight. Then the reaction mixture was filtered and concentrated under reduced pressure, the resultant residue was purified with flash chromatography (silica gel, gradient eluent: 1-15%of MeOH/DCM) to provide compound B1 (1.2 g, 90%yield) as colorless oil. The product was characterized with mass spectrometry and 1H NMR. The product was characterized with mass spectrometry and 1H NMR. MW calc.: 1774.88; MW Found: 1775.32 [M+H] +. 1H NMR (400 MHz, CDCl3) δ 8.73 (s, 1H) , 8.18 (d, J = 8.4 Hz, 1H) , 7.55 (d, J = 8.6 Hz, 1H) , 5.36 -5.27 (m, 6H) , 4.69 (d, J = 7.9 Hz, 2H) , 4.56 -4.37 (m, 2H) , 4.17 -4.08 (m, 6H) , 4.00 -3.88 (m, 6H) , 3.88 -3.77 (m, 3H) , 3.65 (dd, J = 13.3, 6.6 Hz, 2H) , 3.50 -3.39 (m, 4H) , 3.11 -3.03 (m, 2H) , 2.20 (dd, J = 17.0, 9.2 Hz, 8H) , 2.13 (d, J = 1.5 Hz, 9H) , 2.04 -2.02 (m, 9H) , 1.98 (d, J = 1.9 Hz, 9H) , 1.94 -1.89 (m, 9H) , 1.60 -1.52 (m, 16H) , 1.44 (d, J = 6.4 Hz, 6H) , 1.31 -1.20 (m, 24H) .
Example 14. The preparation of compound B2 of the present disclosure
Compound B2 was prepared in this Example by using the following procedures.
(1) This step comprises the preparation of compound B38 from methyl 4-fluoro-3-nitrobenzoate.
To a solution of methyl 4-fluoro-3-nitrobenzoate (8.0 g, 40.2 mmol, 1.0 eq) and K2CO3 (5.5 g, 40.2 mmol) in anhydrous DMF (100 mL) , under nitrogen atmosphere, was added benzyl (3-aminopropyl) carbamate (8.3 g, 40.2 mmol, 1.0 eq) . The reaction mixture was stirred at 25 ℃ for 6 h, then cold water (100 mL) was added. The mixture was extracted 3 times by ethyl acetate, then the organic phase was washed 3 times by saturated LiCl solution and 1 time by brine. Then dried by anhydrous Na2SO4 and concentrated under reduced pressure to form yellow solid (compound B38) which was directly used in the next step without further purification.
(2) The compound B38 was converted to compound B39.
To a solution of compound B38 (10 g, 25.84 mmol, 1.0 eq) in THF/H2O (9: 1, 100 mL) , under ice bath, was added HCOONH4 (9.78 g, 155.04 mmol, 6.0 eq) and Zn powder (10.14 g, 155.04 mmol, 6.0 eq) . After 10 minutes, the reaction mixture was moved to room temperature and stirred overnight. Then the reaction mixture was filtered and concentrated under reduced pressure. After that, water (100 mL) was added into the mixture, then extracted 3 times by ethyl acetate, the organic phase was washed 1 time by brine. After dried by anhydrous Na2SO4 and concentrated under reduced pressure, the light red solid (compound B39) formed then was directly used in the next step without further purification.
(3) The compound B40, which comprises a benzimidazole structure was prepared from compound B39.
To a solution of compound B39 (8.0 g, 22.5 mmol, 1.0 eq) in EtOH (200 mL) under nitrogen atmosphere, was added 3- ( (tert-butyldimethylsilyl) oxy) propanal (4.2 g, 22.5 mmol, 1.0 eq) and AcOH (5.1 mL, 90 mmol, 4.0 eq) . The reaction mixture was stirred at 80℃ overnight, and then concentrated under reduced pressure. Then a saturated NaHCO3 solution (100 mL) was added, the mixture was extracted 3 times by ethyl acetate, then the organic phase was combined and washed by brine, dried over Na2SO4, and concentrated. The resultant residue (compound B40) was directly used in the next step without further purification. The compound B40 was characterized with mass spectrometry. MW calc.: 525.27; MW. Found: 526.59 [M+H] +.
(4) The tert-butyldimethylsilyl (TBS) protectinggroup contained in compound B40 was replaced with 4, 4′-dimethoxytrityl (DMT) group.
To a solution of compound B40 (5.0 g, 9.5 mmol, 1.0 eq) in anhydrous THF (50 mL) , under nitrogen atmosphere, was added 1 M TBAF THF solution (14.25 mL, 14.25 mmol, 1.5 eq) . The reaction mixture was stirred at room temperature for about 1 h, and then concentrated under reduced pressure. Then water (100 mL) was added, the mixture was extracted 3 times by DCM, then the organic phase was combined and washed by brine, dried over Na2SO4, and concentrated. The resultant residue was dissolved in 50 mL pyridine, and DMTrCl (3.86 g, 11.4 mmol, 1.2 eq) was added. The reaction mixture was stirred at room temperature for 6 h, after concentrated under reduced pressure, the resultant residue was purified by flash chromatography (silica gel, gradient eluent: 1-5%of MeOH/DCM) to provide compound B41 (4.1 g, 60%yield) as yellow solid. The product was characterized with mass spectrometry and 1H NMR. MW calc.: 713.31; MW Found: 303.17 [DMT] -, 412.36 [DMT off + H] +. 1H NMR (400 MHz, CDCl3) δ 8.39 (d, J = 1.1 Hz, 1H) , 7.96 (dd, J = 8.5, 1.3 Hz, 1H) , 7.38 -7.27 (m, 8H) , 7.20 (dq, J = 6.7, 2.5 Hz, 7H) , 6.76 -6.70 (m, 4H) , 5.10 (s, 2H) , 4.19 (t, J = 7.3 Hz, 2H) , 3.94 (s, 3H) , 3.74 (s, 6H) , 3.65 (t, J = 6.6 Hz, 2H) , 3.20 (d, J = 6.3 Hz, 2H) , 3.09 (t, J = 6.4 Hz, 2H) , 2.00 -1.90 (m, 2H) .
(5) The carbonyloxy group contained in compound B41 was reduced to methylene-oxy group so as to produce compound B42.
To a solution of compound B41 (4.0 g, 5.6 mmol, 1.0 eq) in anhydrous THF (50 mL) under nitrogen atmosphere and ice bath, was added LiAlH4 (319 mg, 8.4 mmol, 1.5 eq) . The mixture was moved to room temperature after 10 minutes and stirred for 1 h. Then the reaction was moved to ice bath, saturated potassium sodium tartrate solution (30 mL) was added slowly into the mixture. After 30 minutes, the reaction was extracted 3 times with Et2O, then the
organic phase was combined and washed by brine, dried over Na2SO4, and concentrated. The resultant residue was dissolved in 50 mL DMF, then imidazole (572 mg, 8.4 mmol, 1.5 eq) and TBSCl (1.27 g, 8.4 mmol, 1.5 eq) was added. The reaction mixture was stirred at room temperature for 1 h, after concentrated under reduced pressure, the resultant residue was purified by flash chromatography (silica gel, gradient eluent: 1-5%of MeOH/DCM) to provide compound B42 (4.03 g, 90%yield) as yellow solid. The product was characterized with mass spectrometry and 1H NMR. MW calc.: 799.40; MW. Found: 303.17 [DMT] . 1H NMR (400 MHz, CDCl3) δ 7.65 (s, 1H) , 7.38 -7.29 (m, 7H) , 7.19 (ddd, J = 8.4, 7.9, 3.7 Hz, 9H) , 6.78 -6.71 (m, 4H) , 5.10 (s, 2H) , 4.84 (s, 2H) , 4.16 (t, J = 7.2 Hz, 2H) , 3.75 (s, 6H) , 3.61 (t, J = 6.8 Hz, 2H) , 3.18 (d, J = 6.3 Hz, 2H) , 3.07 (t, J = 6.4 Hz, 2H) , 2.95 (s, 1H) , 2.88 (s, 1H) , 0.94 (s, 9H) , 0.10 (s, 6H) .
(6) The compound B22 was originated from the steps (1) - (9) of Example 13
(7) Compound B43 originated from reduction of compound B42 was linked with compound B22.
To a solution of compound B22 (1.36 g, 0.732 mmol, 1.1 eq) in MeOH (15 mL) , was added Pd/C (136 mg) under nitrogen atmosphere slowly. Then the reaction atmosphere was replaced with hydrogen three times. After that, the reaction mixture was stirred by purging with a hydrogen balloon at room temperature overnight. Then the reaction mixture was filtered and concentrated under reduced pressure to provide crude product. The crude product was dissolved in DCM (15mL) , then HBTU (508 mg, 1.34 mmol, 2.0 eq) and DIPEA (375 μL, 2.28 mmol, 3.4 eq) were added into the reaction mixture under nitrogen atmosphere. Five minutes later, compound B43 (446 mg, 0.67 mmol, 1.0eq) , which was produced by reducing compound B42, was added into the reaction. The reaction mixture was stirred overnight at room temperature. Then H2O (20 mL) was added into the reaction, the mixture was extracted with DCM (3*50 mL) . The organic phase was combined, dried over Na2SO4, and concentrated. The resultant residue was purified with flash chromatography (silica gel, gradient eluent: 1-5%of MeOH/DCM) to provide compound B24 (1.5 g, 93%yield) as yellow solid. The product was characterized with mass spectrometry and 1H NMR. MW calc.: 2422.23; MW Found: 1212.47 [M+2] /2. 1H NMR (400 MHz, CDCl3) δ 8.96 (s, 1H) , 8.81 (d, J = 2.8 Hz, 1H) , 8.13 (s, 1H) , 8.02 (d, J = 7.9 Hz, 2H) , 7.63 (d, J = 7.6 Hz, 2H) , 7.54 (t, J = 7.1 Hz, 2H) , 7.29 (s, 2H) , 7.18 (s, 4H) , 6.72 (d, J = 8.8 Hz, 4H) , 5.35 (s, 2H) , 5.29 (d, J = 11.0 Hz, 3H) , 4.83 (s, 2H) , 4.70 -4.67 (m, 2H) , 4.28 (t, J = 7.2 Hz, 3H) , 4.17 -4.09 (m, 6H) , 3.94 -3.80 (m, 8H) , 3.74 (s, 6H) , 3.70 -3.57 (m, 8H) , 3.46 (d, J = 4.9 Hz, 4H) , 3.12 -3.06 (m, 4H) , 2.78 -2.65 (m, 3H) , 2.20 (d, J = 8.1 Hz, 3H) , 2.14 (d, J = 14.6 Hz, 6H) , 2.12 (s, 9H) , 2.02 (s, 9H) , 1.98 (s, 9H) , 1.92 (s, 9H) , 1.54 -1.50 (m, 12H) , 1.28 (s, 6H) , 1.22 -1.16 (m, 24H) , 0.93 (s, 9H) , 0.10 (s, 6H) .
(8) Compound B25 was generated by deprotecting TBS group of compound B24 then acylated with succinic anhydride.
To a solution of compound B24 (1.5 g, 0.62 mmol, 1.0 eq) in anhydrous THF (35 mL) , under nitrogen atmosphere, was added 1 M TBAF THF solution (1.87 mL, 1.87 mmol, 3.0 eq) . The reaction mixture was stirred at room temperature for 1 h, and then concentrated under reduced pressure to provide crude product. The crude product was dissolved in DCM (15 mL) , then DMAP (265 mg, 2.17 mmol, 3.5 eq) , succinic anhydride (124 mg, 1.24 mmol, 2.0 eq) were added therein under nitrogen atmosphere. The reaction mixture was stirred at room temperature overnight then H2O (20 mL) was added into the reaction, the mixture was extracted with DCM (3*50 mL) and the organic phase was combined, dried over Na2SO4, and concentrated. The resultant residue was purified with flash chromatography. The resultant residue was purified with flash chromatography (silica gel, gradient eluent: 1-10%of MeOH/DCM) to provide compound B25 (794 mg, 53%yield) as colorless oil. The product was characterized with mass spectrometry and 1H NMR. MW calc.: 2408.12; MW Found: 1205.60 [M+2] /2. 1H NMR (400 MHz, CDCl3) δ 8.30 (s, 1H) , 7.92 -7.84 (m, 2H) , 7.59 (s, 2H) , 7.31 (d, J = 6.9 Hz, 2H) , 7.22 -7.14 (m, 8H) , 6.74 (d, J = 8.8 Hz, 4H) , 5.32 -5.25 (m, 3H) , 5.20 (s, 2H) , 4.77 -4.71 (m, 2H) , 4.28 (s, 2H) , 4.13 (dd, J = 9.6, 4.8 Hz, 6H) , 4.01 (dd, J
= 8.7, 6.2 Hz, 3H) , 3.92 (d, J = 6.6 Hz, 3H) , 3.88 -3.83 (m, 2H) , 3.75 (s, 6H) , 3.61 (t, J = 6.7 Hz, 2H) , 3.39 -3.30 (m, 14H) , 3.13 (d, J = 6.1 Hz, 4H) , 2.70 (dd, J = 19.6, 12.8 Hz, 4H) , 2.34 -2.20 (m, 6H) , 2.12 (s, 9H) , 2.02 (s, 9H) , 1.98 (s, 9H) , 1.93 (s, 9H) , 1.68 (dd, J = 10.1, 6.1 Hz, 12H) , 1.48 -1.43 (m, 12H) , 1.31 -1.22 (m, 24H) .
(9) The compound B25 was linked with a solid support, Controlled Pore Glass (CPG) , to produce Compound B2 of the present disclosure.
To a solution of the compound B25 (754 mg, 0.33 mmol, 1.0 eq) , Controlled Pore Glass (CPG) (5.58 g) , N, N-Diisopropylethylamine (DIPEA) (160 uL, 0.99 mmol, 3.0 eq) in acetonitrile (40 mL) was added O- (1H-Benzotriazol-1-yl) -N, N, N′, N′-tetramethyluronium hexafluorophosphate (HBTU) (250 mg, 0.66 mmol, 2.0 eq) under nitrogen atmosphere. The reaction mixture was stirred at 25℃ overnight and then subsequently washed with DCM and ethyl ether to generate crude solid support. To a solution of acetic anhydride (17.7 mL) , pyridine (40 mL) , NEt3 (580 μL) in acetonitrile (26.2 mL) was added the crude solid support under nitrogen atmosphere. The reaction mixture was stirred at 25 ℃ overnight and then subsequently washed with DCM and ethyl ether to produce the Compound B2 of the present
disclosure (5.3 g) .
Example 15. The preparation of Compound B3 of the present disclosure.
The Compound B3 of the present disclosure was prepared in this Example by using the following procedures.
(1) The compound B39 was originated from the steps (1) - (2) of Example 14.
(2) The compound B44, which comprises a benzimidazole structure was prepared from the compound B39.
To a solution of compound B39 (8.87 g, 24.84 mmol, 1.0 eq) in EtOH (220 mL) under nitrogen atmosphere, was added 5- ( (tert-butyldimethylsilyl) oxy) pentanal B11 (5.4 g, 24.84 mmol, 1.0 eq) and AcOH (5.73 mL, 99.36 mmol, 4.0 eq) . The reaction mixture was stirred at 80℃ overnight, and then concentrated under reduced pressure. Then saturated NaHCO3 solution (100 mL) was added therein, the mixture was extracted 3 times with ethyl acetate. The organic phase was combined and washed by brine, dried over Na2SO4, and concentrated. The resultant residue was purified with flash chromatography (silica gel, gradient eluent: 1-5%of MeOH/DCM) to provide the compound B44 (6.3 g, 46%yield) as red solid. The product
was characterized with mass spectrometry, 1H NMR and 13C NMR. MW calc.: 553.30; MW Found: 554.29 [M+H] +. 1H NMR (400 MHz, CDCl3) δ 8.42 (d, J = 0.8 Hz, 1H) , 7.95 (dd, J = 8.5, 1.1 Hz, 1H) , 7.44 -7.28 (m, 5H) , 7.26 (d, J = 6.8 Hz, 1H) , 5.12 (s, 2H) , 4.16 (dd, J = 12.6, 5.3 Hz, 2H) , 3.93 (s, 3H) , 3.67 (t, J = 6.3 Hz, 2H) , 3.28 (d, J = 6.2 Hz, 2H) , 2.87 (t, J = 7.6 Hz, 2H) , 2.04 -1.88 (m, 4H) , 1.67 (dd, J = 14.7, 6.6 Hz, 2H) , 0.88 (s, 9H) , 0.04 (s, 6H) . 13C NMR (100 MHz, CDCl3) δ 167.72, 156.63, 142.35, 138.19, 136.30, 128.60, 128.24, 124.10, 123.87, 121.63, 108.66, 66.96, 62.71, 60.41, 52.06, 41.31, 38.57, 32.46, 30.43, 27.30, 25.97, 24.15, 21.07, 18.34, 14.21.
(3) The tert-butyldimethylsilyl (TBS) protecting group contained in compound B44 was replaced with 4, 4′-dimethoxytrityl (DMT) group.
To a solution of compound B44 (6.3 g, 11.4 mmol, 1.0 eq) in anhydrous THF (50 mL) under nitrogen atmosphere, was added 1 M TBAF THF solution (17.1 mL, 17.1 mmol, 1.5 eq) . The reaction mixture was stirred at room temperature for 1 h, and then concentrated under reduced pressure. Then water (100 mL) was added, the mixture was extracted 3 times with DCM, then the organic phase was combined and washed with brine, dried over Na2SO4, and concentrated. The resultant residue was dissolved in 50 mL pyridine, and DMTrCl (4.6 g, 13.68 mmol, 1.2 eq) was added therein. The reaction mixture was stirred at room temperature for 6 h, after which it was concentrated under reduced pressure. The resultant residue was purified with flash chromatography (silica gel, gradient eluent: 1-5%of MeOH/DCM) to provide compound B45 (6.23 g, 74%yield) as yellow solid. The product was characterized with mass spectrometry, 1H NMR and 13C NMR. MW calc.: 741.34; MW. Found: 303.11 [DMT] -, 440.14 [DMT off + H] +. 1H NMR (400 MHz, CDCl3) δ 8.63 (dd, J = 5.7, 1.5 Hz, 1H) , 8.43 (d, J = 1.0 Hz, 1H) , 7.97 (dd, J = 8.5, 1.1 Hz, 1H) , 7.45 (d, J = 7.4 Hz, 2H) , 7.37 -7.28 (m, 11H) , 7.21 (dd, J = 8.1, 6.1 Hz, 1H) , 6.83 (t, J = 5.9 Hz, 4H) , 5.13 (s, 2H) , 4.12 (t, J = 7.2 Hz, 2H) , 3.95 (s, 3H) , 3.79 (s, 6H) , 3.24 (d, J = 6.2 Hz, 2H) , 3.14 (t, J = 6.2 Hz, 2H) , 2.83 (t, J = 7.4 Hz, 2H) , 2.01 (dd, J = 16.0, 8.5 Hz, 4H) , 1.79 (dd, J = 14.1, 6.6 Hz, 2H) . 13C NMR (100
MHz, CDCl3) δ 167.81, 158.43, 156.64, 149.95, 145.34, 142.43, 138.26, 136.57, 136.05, 130.11, 128.69, 128.39, 128.26, 128.23, 127.84, 126.75, 124.17, 123.89, 121.70, 113.11, 108.75, 85.93, 67.02, 62.90, 55.31, 52.16, 41.37, 38.64, 30.50, 29.76, 27.30, 24.53.
(4) The carbonyloxy group contained in compound B45 was reduced to methylene-oxy group so as to produce compound B46.
To a solution of compound B45 (5.0 g, 6.74 mmol, 1.0 eq) in anhydrous THF (60 mL) under nitrogen atmosphere and ice bath, was added LiAlH4 (384 mg, 10.11 mmol, 1.5 eq) . The mixture was then transferred to room temperature after 10 minutes and stirred for about 1 h. Then the reaction was transferred to ice bath, and saturated potassium sodium tartrate solution (30 mL) was added slowly into the mixture. After reacting for 30 minutes, the reaction was extracted 3 times with Et2O, then the organic phase was combined and washed with brine, dried over Na2SO4, and concentrated. The resultant residue was dissolved in 20 mL DMF, then imidazole (688 mg, 10.11 mmol, 1.5 eq) and TBSCl (1.524 g, 10.11 mmol, 1.5 eq) were added. The reaction mixture was stirred at room temperature for 1 h. After being concentrated under reduced pressure, the resultant residue was purified with flash chromatography (silica gel, gradient eluent: 1-5%of MeOH/DCM) to provide compound B46 (4.78 g, 86%yield) as yellow solid. The product was characterized with mass spectrometry, 1H NMR and 13C NMR. MW calc.: 827.43; MW. Found: 303.16 [DMT] -, 526.50 [DMT off + H] +. 1H NMR (400 MHz, CDCl3) δ 7.69 (s, 1H) , 7.47 -7.42 (m, 2H) , 7.40 -7.28 (m, 11H) , 7.22 (t, J = 3.5 Hz, 3H) , 6.86 -6.80 (m, 4H) , 5.13 (s, 2H) , 4.86 (s, 2H) , 4.15 (dd, J = 14.3, 7.2 Hz, 2H) , 3.79 (s, 6H) , 3.13 (t, J = 6.3 Hz, 2H) , 2.82 (t, J = 7.5 Hz, 2H) , 2.05 -1.94 (m, 4H) , 1.81 -1.74 (m, 2H) , 1.29 (t, J = 7.1 Hz, 2H) , 0.97 (s, 9H) , 0.13 (s, 6H) . 13C NMR (100 MHz, CDCl3) δ 171.27, 158.45, 156.60, 154.95, 145.40, 142.93, 136.66, 135.52, 134.14, 130.14, 128.70, 128.37, 128.28, 127.85, 126.74, 120.93, 117.18, 113.13, 108.73, 85.93, 67.00, 65.53, 63.00, 60.51, 55.32, 53.55, 41.19, 38.74, 30.51, 29.85, 27.33, 26.14, 24.78, 21.17, 18.57, 14.33.
(5) The compound B22 was originated from the steps (1) - (9) of Example 13.
(6) The compound B22 was linked with compound B47, which was derived from compound B46, to form compound B27.
To a solution of compound B22 (2.24 g, 1.2 mmol, 1.0 eq) in MeOH (20 mL) , was added Pd/C (224 mg) under nitrogen atmosphere slowly. Then the reaction atmosphere was replaced with hydrogen gas for three times. After that, the reaction mixture was stirred by purging with a hydrogen balloon at room temperature overnight. Then the reaction mixture was filtered and concentrated under reduced pressure to provide crude product. The crude product was dissolved in DCM (20 mL) , then HBTU (910 mg, 2.4 mmol, 2.0) and DIPEA (676 μL, 4.08 mmol, 3.4 eq) were added into the reaction under nitrogen atmosphere. Five minutes later, compound B47 (849 mg, 1.2 mmol) , which was produced by reducing compound B46, was added into the reaction. The reaction mixture was stirred overnight at room temperature. Then H2O (20 mL) was added into the reaction, and the mixture was extracted with DCM (3*50 mL) . The organic phase was combined, dried over Na2SO4, and concentrated. The resultant residue was purified with flash chromatography (silica gel, gradient eluent: 1-8%of MeOH/DCM) to provide compound B27 (1.48 g, 50%yield) as white
solid. The product was characterized with mass spectrometry and 1H NMR. MW calc.: 2450.26; MW. Found: 1074.76 [DMT off] -/2. 1H NMR (400 MHz, CDCl3) δ 8.09 (s, 1H) , 7.73 -7.59 (m, 2H) , 7.39 (t, J = 10.1 Hz, 3H) , 7.29 (d, J = 8.6 Hz, 5H) , 7.25 -7.15 (m, 4H) , 6.78 (t, J = 8.2 Hz, 4H) , 5.42 -5.16 (m, 9H) , 4.83 (s, 2H) , 4.68 (d, J = 7.6 Hz, 2H) , 4.42 -4.25 (m, 2H) , 4.22 -4.06 (m, 8H) , 4.02 -3.79 (m, 10H) , 3.80 -3.74 (m, 6H) , 3.70 -3.30 (m, 8H) , 3.21 (s, 2H) , 3.09 (t, J = 5.9 Hz, 2H) , 2.86 (d, J = 7.5 Hz, 1H) , 2.31 -2.14 (m, 10H) , 2.12 (s, 9H) , 2.05 -2.02 (m, 9H) , 1.99 (s, 9H) , 1.93 (s, 9H) , 1.75 (d, J = 6.8 Hz, 2H) , 1.60 -1.50 (m, 12H) , 1.26 (d, J = 12.4 Hz, 24H) , 0.93 (s, 9H) , 0.10 (s, 6H) .
(7) Compound B28 was generated by deprotecting TBS group of compound B27 then acylated with succinic anhydride.
To a solution of compound B27 (1.22 g, 0.5 mmol, 1.0 eq) in anhydrous THF (10 mL) under nitrogen atmosphere, was added 1 M TBAF THF solution (1.5 mL, 1.5 mmol, 3 eq) . The reaction mixture was stirred at room temperature for 1 h, and then concentrated under reduced pressure to provide crude product. The crude product was dissolved in DCM (10 mL) then DMAP (212 mg, 1.75 mmol, 3.5 eq) , succinic anhydride (150 mg, 1.5 mmol, 3.0 eq) were added therein under nitrogen atmosphere. The reaction mixture was stirred at room temperature overnight and then H2O (10 mL) was added into the reaction. The mixture was
extracted with DCM (3*20 mL) . The organic phase was combined, dried over Na2SO4, and concentrated. The resulting residue was purified with flash chromatography (silica gel, gradient eluent: 1-10%of MeOH/DCM) to provide compound B28 (380 mg, 31%yield) as colorless oil. The product was characterized with mass spectrometry. MW calc.: 2436.19; MW. Found: 1067.52 [DMT off] -/2.
(8) The compound B28 was linked with a solid support, Controlled Pore Glass (CPG) , to produce Compound B3 of the present disclosure.
To a solution of compound B28 (250 mg, 0.103 mmol, 1.0 eq) , Controlled Pore Glass (CPG) (1.8 g) , N, N-Diisopropylethylamine (DIPEA) (51 μL, 0.309 mmol, 3.0 eq) in acetonitrile (14 mL) was added HBTU (78 mg, 0.206 mmol, 2.0 eq) under nitrogen atmosphere. The reaction mixture was stirred at 25 ℃ overnight and then sequentially washed with DCM and ethyl ether to produce the crude solid support. To a solution of acetic anhydride (6.2 mL) , pyridine (12 mL) , NEt3 (186 uL) in acetonitrile (7.9 mL) was added the crude solid support under nitrogen atmosphere. The reaction mixture was stirred at 25℃ overnight and then sequentially washed with DCM and ethyl ether to produce Compound B3
of the present disclosure (1.84 g) .
Example 16: The preparation of Compound B4 of the present disclosure.
The Compound B4 of the present disclosure was prepared in this Example by using the following procedures.
(1) Compound B17 was originated from the step (6) of Example 13.
(2) An alkoxy group terminated with vinyl group was attached to compound B17.
To a solution of compound B17 (19.7 g, 59.8 mmol, 1.0 eq) in DCE (260 mL) , was added hex-5-en-1-ol (7.2 g, 71.8 mmol, 1.2 eq) and TMSOTf (2.2 mL, 12 mmol, 0.2 eq) under nitrogen atmosphere. The reaction mixture was stirred at room temperature for 6 h. Then saturated NaHCO3 solution (100 mL) was added, the mixture was extracted 3 times with DCM. The organic phase was combined and washed with brine, dried over Na2SO4, and concentrated. The resultant residue was purified with flash chromatography (silica gel, gradient eluent: 1-5%of MeOH/DCM) to provide compound B18 (18.7 g, 73%yield) .
(3) The terminal vinyl group of compound B18 was oxidized to a carboxyl group.
To a solution of compound B18 (18.7g, 43.6 mmol, 1.0 eq) in ACN/DCM/H2O (140mL/140mL/210mL) under ice bath, was added NaIO4 (37.3 g, 174.4 mol, 4.0 eq) and RuCl3·3H2O (1.8 g, 8.72 mol, 0.2 eq) . After 10 minutes, the reaction mixture was transferred to room temperature and stirred overnight. Then the reaction mixture was filtered and concentrated under reduced pressure. After that, water (100 mL) was added into the mixture, then the mixture was extracted 3 times with DCM, the organic phase was washed 1 time with brine. After being dried with anhydrous Na2SO4 and concentrated under reduced pressure, the resultant residue was purified with flash chromatography (silica gel, gradient eluent: 1-8%of MeOH/DCM) to provide compound B19 (19 g, 97%yield) . The product was characterized with mass spectrometry and 1H NMR. MW calc.: 447.17; MW Found: 448.36 [M+H] +. 1H NMR (400 MHz, CDCl3) δ 6.15 (d, J = 8.7 Hz, 1H) , 5.33 (d, J = 3.0 Hz, 1H) , 5.29 -5.26 (m, 1H) , 4.66 (d, J = 8.4 Hz, 1H) , 4.13 (dd, J = 9.3, 6.8 Hz, 2H) , 3.92 (dd, J = 8.4, 5.4 Hz, 2H) , 3.50 (dd, J = 5.7, 4.0 Hz, 1H) , 2.35 (dd, J = 14.3, 7.0 Hz, 2H) , 2.13 (s, 3H) , 2.03 (s, 3H) , 1.98 (s, 3H) , 1.95 (s, 3H) , 1.69 -1.61 (m, 4H) .
(4) Compound B16 was originated from the steps (1) - (5) of Example 13.
(5) The compound B16 was linked with compound B19 to produce compound B30.
To a solution of compound B16 (1.8 g, 2.54 mmol, 1.0 eq) in DCM (10 mL) , was added HCl/Dioxane (4M, 20 mL) . The reaction mixture was stirred for 3 h at room temperature, then concentrated under reduced pressure to provide crude product as yellow
solid. After that, the crude product was dissolved in 25 mL DCM, then compound B19 (3.4 g, 7.62 mmol, 3.0 eq) , HBTU (3.37 g, 8.89 mmol, 3.5 eq) and DIPEA (5.05 mL, 30.48 mmol, 12.0 eq) were added into the reaction under nitrogen atmosphere. The reaction mixture was stirred overnight at room temperature. Then 20 mL H2O was added into the reaction mixture. The mixture was extracted with DCM (3*30 mL) . The organic phase was combined, dried over Na2SO4, and concentrated. The resultant residue was purified with flash chromatography (silica gel, gradient eluent: 1-8%of MeOH/DCM) to provide compound B30 (3.67 g, 85%yield) as yellow solid. The product was characterized with mass spectrometry and 1H NMR. MW calc.: 1697.80; MW Found: 1698.53 [M+H] +. 1H NMR (400 MHz, CDCl3) δ 8.41 (d, J = 22.5 Hz, 1H) , 8.04 (d, J = 8.5 Hz, 1H) , 7.44 (d, J = 7.0 Hz, 2H) , 7.40 -7.31 (m, 4H) , 5.42 -5.26 (m, 7H) , 5.19 (td, J = 11.0, 3.3 Hz, 2H) , 4.64 -4.55 (m, 2H) , 4.24 -3.99 (m, 10H) , 3.94 -3.75 (m, 6H) , 3.68 -3.62 (m, 1H) , 3.35 -3.16 (m, 4H) , 3.11 -3.06 (m, 1H) , 2.35 -2.26 (m, 3H) , 2.23 -2.15 (m, 6H) , 2.12 (d, J = 6.9 Hz, 9H) , 2.03 (s, 9H) , 1.99 (s, 9H) , 1.92 (dd, J = 11.6, 5.2 Hz, 9H) , 1.78 (s, 2H) , 1.71 -1.61 (m, 6H) , 1.51 (dd, J = 12.6, 6.5 Hz, 8H) , 1.42 (d, J = 6.6 Hz, 4H) .
(6) Compound B47 was originated from the steps (3) - (5) of Example 15.
(7) The compound B30 was linked with compound B47, which was derived from compound B46, to form compound B31.
To a solution of compound B30 (1.85 g, 1.09 mmol, 1.0 eq) in MeOH (20 mL) , was added Pd/C (185 mg) under nitrogen atmosphere slowly. Then the reaction atmosphere was replaced with hydrogen gas for three times. After that, the reaction mixture was stirred by purging with a hydrogen balloon at room temperature overnight. Then the reaction mixture was filtered and concentrated under reduced pressure to provide crude product.
The crude product was dissolved in DCM (20 mL) , then HBTU (826 mg, 2.18 mmol, 2.0 eq) and DIPEA (614 μL, 3.7 mmol, 3.4 eq) were added into the reaction under nitrogen atmosphere. Five minutes later, compound 47 (849 mg, 1.2 mmol, 1.1 eq) , which was produced by reducing compound B46, was added into the reaction mixture. The reaction mixture was stirred overnight at room temperature. Then H2O (20 mL) was added into the reaction, the mixture was extracted with DCM (3*50 mL) . The organic phase was combined, dried over Na2SO4, and concentrated. The resultant residue was purified with flash chromatography (silica gel, gradient eluent: 1-8%of MeOH/DCM) to provide compound B31 (1.8 g, 72%yield) as white solid. The product was characterized with mass spectrometry and 1H NMR. MW calc.: 2282.08; MW Found: 1142.33 [M+2H] +/2. 1H NMR (400 MHz, CDCl3) δ 8.08 (s, 1H) , 7.64 (s, 1H) , 7.38 (dd, J = 12.7, 8.1 Hz, 4H) , 7.28 (d, J = 8.8 Hz, 5H) , 7.23 (d, J
= 7.8 Hz, 2H) , 7.20 -7.15 (m, 2H) , 6.78 (d, J = 8.8 Hz, 4H) , 5.33 (s, 3H) , 5.23 -5.11 (m, 2H) , 4.81 (s, 2H) , 4.58 (dd, J = 8.4, 2.1 Hz, 2H) , 4.18 (d, J = 10.6 Hz, 3H) , 4.17 -4.04 (m, 8H) , 3.96 -3.80 (m, 6H) , 3.75 (s, 6H) , 3.63 (dd, J = 13.3, 6.6 Hz, 1H) , 3.57 -3.40 (m, 5H) , 3.22 (s, 3H) , 3.07 (dd, J = 10.9, 4.7 Hz, 2H) , 2.85 (t, J = 7.5 Hz, 2H) , 2.42 (s, 6H) , 2.20 (dd, J = 34.7, 6.1 Hz, 8H) , 2.10 (d, J = 7.6 Hz, 9H) , 2.06 (d, J = 9.1 Hz, 2H) , 2.01 (d, J = 3.3 Hz, 9H) , 1.97 (dd, J = 7.5, 2.4 Hz, 9H) , 1.89 (dd, J = 28.9, 13.9 Hz, 9H) , 1.80 -1.64 (m, 6H) , 1.62 -1.46 (m, 12H) , 1.40 (d, J = 6.0 Hz, 3H) , 0.92 (s, 9H) , 0.08 (s, 6H) .
(8) Compound B32 was generated by deprotecting TBS group of compound B31 then acylated with succinic anhydride.
To a solution of compound B31 (1.57 g, 0.687 mmol, 1.0 eq) in anhydrous THF (10 mL) under nitrogen atmosphere, was added 1 M TBAF THF solution (2.06 mL, 2.06 mmol, 3.0 eq) . The reaction mixture was stirred at room temperature for 1 h, and then concentrated under reduced pressure to produce the crude product.
The crude product was dissolved in DCM (15 mL) then DMAP (587 mg, 4.8 mmol, 3.5 eq) , succinic anhydride (137 mg, 1.374 mmol, 2.0 eq) were added under nitrogen atmosphere. The reaction mixture was stirred at room temperature overnight, then H2O (20 mL) was added into the reaction mixture. The mixture was extracted with DCM (3*50 mL) . The
organic phase was combined, dried over Na2SO4, and concentrated. The resultant residue was purified with flash chromatography. The resultant residue was purified with flash chromatography (silica gel, gradient eluent: 1-10%of MeOH/DCM) to provide compound B32 (1.0 g, 64%yield) as colorless oil. The product was characterized with mass spectrometry and 1H NMR. MW calc.: 2268.01; MW Found: 1135.22 [M+2H] +/2. 1H NMR (400 MHz, CDCl3) δ 8.17 (d, J = 6.6 Hz, 2H) , 7.40 (d, J = 7.6 Hz, 2H) , 7.30 -7.26 (m, 5H) , 7.24 (s, 2H) , 7.18 (d, J = 6.8 Hz, 2H) , 6.79 (d, J = 8.8 Hz, 4H) , 6.50 (s, 2H) , 5.32 (s, 2H) , 5.29 (s, 5H) , 5.25 -5.12 (m, 4H) , 4.60 (dd, J = 15.7, 8.3 Hz, 2H) , 4.17 (d, J = 10.0 Hz, 2H) , 4.14 -3.96 (m, 8H) , 3.87 (dd, J = 15.1, 6.5 Hz, 5H) , 3.76 (s, 6H) , 3.45 (s, 6H) , 3.19 (d, J = 8.7 Hz, 2H) , 3.08 (t, J = 6.0 Hz, 2H) , 3.01 (s, 10H) , 2.90 -2.77 (m, 2H) , 2.63 (d, J = 17.4 Hz, 4H) , 2.19 (dd, J = 24.8, 17.2 Hz, 9H) , 2.12 -2.08 (m, 9H) , 2.01 (d, J = 3.7 Hz, 9H) , 1.98 -1.94 (m, 9H) , 1.87 (dd, J = 24.8, 13.3 Hz, 9H) , 1.76 -1.62 (m, 6H) , 1.55 (d, J = 15.4 Hz, 12H) .
(9) The compound B32 was linked with a solid support, Controlled Pore Glass (CPG) , to produce Compound B4 of the present disclosure.
To a solution of compound B32 (233 mg, 0.103 mmol, 1.0 eq) , Controlled Pore Glass (CPG) (1.8 g) , N, N-diisopropylethylamine (DIPEA) (51 μL, 0.309 mmol, 3.0 eq) in acetonitrile (14 mL) was added HBTU (78 mg, 0.206 mmol, 2.0 eq) under nitrogen
atmosphere. The reaction mixture was stirred at 25 ℃ overnight and then washed with DCM and ethyl ether to generate crude solid support.
To a solution of acetic anhydride (6.2 mL) , pyridine (12 mL) , NEt3 (186 uL) in acetonitrile (7.9 mL) was added the crude solid support under nitrogen atmosphere. The reaction mixture was stirred at 25 ℃ overnight and then washed with DCM and ethyl ether to produce Compound B4 of the present disclosure (1.84 g) .
Example 17: The preparation of Compound B5 of the present disclosure.
The Compound B5 of the present disclosure was prepared in this Example by using the following procedures.
(1) Compound B46 was originated from the steps (3) - (5) of Example 15.
(2) Compound B48 was generated by reduction of compound B46 then acylation with 6- ( ( (benzyloxy) carbonyl) amino) hexanoic acid.
To a solution of compound B46 (1.0 g, 1.2 mmol, 1.0 eq) in MeOH (30 mL) , was added Pd/C (100 mg) under nitrogen atmosphere slowly. Then the reaction atmosphere was replaced with hydrogen for three times. After that, the reaction mixture was stirred by purging with a hydrogen balloon at room temperature overnight. Then the reaction mixture was filtered
and concentrated under reduced pressure to provide crude product. After that, the crude product was dissolved in 20 mL DCM, then 6- ( ( (benzyloxy) carbonyl) amino) hexanoic acid (350 mg, 1.32 mmol, 1.1 eq) , HBTU (682 mg, 1.8 mmol, 1.5 eq) and DIPEA (675 μL, 4.08 mmol, 3.4 eq) were added into the reaction mixture under nitrogen atmosphere. The reaction mixture was stirred overnight at room temperature. Then 20 mL H2O was added into the reaction mixture, the mixture was extracted with DCM (3*50 mL) . The organic phase was combined, dried over Na2SO4, and concentrated. The resultant residue was purified with flash chromatography (silica gel, gradient eluent: 1-5%of MeOH/DCM) to provide compound B48 (1.0 g, 88%yield) as yellow oil. The product was characterized with mass spectrometry and 1H NMR. MW calc.: 940.52; MW. Found: 303.29 [DMT] -, 639.64 [DMT off + H] +. 1H NMR (400 MHz, CDCl3) δ 7.62 (d, J = 4.0 Hz, 1H) , 7.40 (dd, J = 5.3, 3.3 Hz, 2H) , 7.29 (dtd, J = 7.8, 4.7, 2.2 Hz, 14H) , 6.80 (dt, J = 9.0, 5.7 Hz, 4H) , 5.06 (s, 2H) , 4.81 (s, 2H) , 4.11 (t, J = 7.4 Hz, 2H) , 3.77 (s, 6H) , 3.18 -3.08 (m, 4H) , 2.88 (t, J = 7.6 Hz, 2H) , 2.09 -1.92 (m, 6H) , 1.60 -1.25 (m, 10H) , 0.93 (s, 9H) , 0.10 (s, 6H) .
(3) Compound B22 was originated from the steps (1) - (9) of Example 13.
(4) Compound B22 was linked with compound B49, which was derived from compound B48, to form compound B34.
To a solution of compound B22 (2.4 g, 1.28 mmol) in MeOH (20 mL) , was added Pd/C (240 mg) under nitrogen atmosphere slowly. Then the reaction atmosphere was replaced with hydrogen gas for three times. After that, the reaction mixture was stirred by purging with a hydrogen balloon at room temperature overnight. Then the reaction mixture was filtered and concentrated under reduced pressure to provide crude product.
The crude product was dissolved in DCM (20 mL) , then HBTU (971 mg, 2.56 mmol, 2.0 eq) and DIPEA (721 μL, 4.352 mmol, 3.4 eq) were added into the reaction under nitrogen atmosphere. After five minutes, the compound B49 (1.04 g, 1.27 mmol, 1.0 eq) , which was obtained by reducing the compound B48, was added into the reaction mixture. The reaction mixture was stirred overnight at room temperature. Then H2O (20 mL) was added into the reaction mixture. The mixture was extracted with DCM (3*30 mL) . The organic phase was combined, dried over Na2SO4, and concentrated. The resultant residue was purified with flash chromatography (silica gel, gradient eluent: 1-8%of MeOH/DCM) to provide compound B34 (1.07 g, 33%yield) as white solid. The product was characterized with mass spectrometry and 1H NMR. MW calc.: 2563.35; MW Found: 1283.20 [M+2H] +/2. 1H NMR (400 MHz, CDCl3) δ 8.10 (s, 1H) , 7.73 (t, J = 7.7 Hz, 1H) , 7.62 (s, 1H) , 7.39 (d, J = 7.4 Hz, 2H) , 7.28 (d, J = 8.9
Hz, 5H) , 7.25 -7.21 (m, 3H) , 7.19 -7.15 (m, 2H) , 6.78 (d, J = 8.8 Hz, 4H) , 5.34 (s, 4H) , 5.29 (s, 2H) , 5.27 (s, 2H) , 4.80 (s, 2H) , 4.68 (t, J = 7.5 Hz, 3H) , 4.13 (dt, J = 17.5, 10.8 Hz, 9H) , 4.03 -3.96 (m, 3H) , 3.92 (d, J = 6.2 Hz, 4H) , 3.76 (s, 6H) , 3.67 -3.63 (m, 2H) , 3.44 (d, J = 5.4 Hz, 6H) , 3.08 (t, J = 6.1 Hz, 3H) , 2.79 (t, J = 7.6 Hz, 2H) , 2.15 (d, J = 7.7 Hz, 12H) , 2.12 (s, 9H) , 2.07 (s, 3H) , 2.02 (d, J = 3.1 Hz, 9H) , 1.98 (d, J = 1.8 Hz, 9H) , 1.97 (s, 5H) , 1.91 (s, 9H) , 1.56 -1.49 (m, 18H) , 1.42 (d, J = 6.6 Hz, 6H) , 1.24 (d, J = 15.2 Hz, 24H) , 0.92 (s, 9H) , 0.08 (s, 6H) .
(5) Compound B35 was generated by deprotecting TBS group of compound B34 then acylated with succinic anhydride.
To a solution of compound B34 (873 mg, 0.346 mmol, 1.0 eq) in anhydrous THF (10 mL) under nitrogen atmosphere, was added 1 M TBAF THF solution (2.07 mL, 2.07 mmol, 6.0 eq) . The reaction mixture was stirred at room temperature for 1 h, and then concentrated under reduced pressure to generate the crude product.
The crude product was dissolved in DCM (10 mL) then DMAP (148 mg, 1.21 mmol, 3.5 eq) , succinic anhydride (104 mg, 1.038 mmol, 3.0 eq) were added under nitrogen atmosphere. The reaction mixture was stirred at room temperature overnight, and then H2O (20 mL) was added into the reaction. The mixture was extracted with DCM (3*30 mL) . The
organic phase was combined, dried over Na2SO4, and concentrated. The resultant residue was purified with flash chromatography (silica gel, gradient eluent: 1-10%of MeOH/DCM) to provide compound B35 (293 mg, 33%yield) as colorless oil. The product was characterized with mass spectrometry and 1H NMR. MW calc.: 2549.28; MW Found: 1276.36 [M+2H] +/2. 1H NMR (400 MHz, CDCl3) δ 8.10 (d, J = 7.1 Hz, 2H) , 7.39 (d, J = 7.5 Hz, 2H) , 7.28 (s, 4H) , 7.25 -7.10 (m, 5H) , 6.78 (d, J = 8.6 Hz, 4H) , 6.70 (d, J = 7.3 Hz, 2H) , 5.33 (s, 2H) , 5.29 (s, 6H) , 4.70 (d, J = 6.1 Hz, 3H) , 4.17 -3.98 (m, 11H) , 3.87 (d, J = 35.7 Hz, 6H) , 3.75 (s, 6H) , 3.46 (s, 4H) , 3.31 -3.24 (m, 2H) , 3.20 (s, 8H) , 3.07 (s, 4H) , 2.72 (d, J = 51.3 Hz, 4H) , 2.14 (s, 6H) , 2.09 (d, J = 16.8 Hz, 12H) , 2.01 (s, 9H) , 1.96 (s, 12H) , 1.92 (s, 9H) , 1.67 (dd, J = 16.4, 7.7 Hz, 6H) , 1.61 -1.40 (m, 18H) , 1.37 -1.03 (m, 30H) .
(6) The compound B35 was linked with a solid support, Controlled Pore Glass (CPG) , to produce compound B5 of the present disclosure.
To a solution of compound B35 (262 mg, 0.103 mmol, 1.0 eq) , Controlled Pore Glass (CPG) (1.8 g) , N, N-Diisopropylethylamine (DIPEA) (51 μL, 0.309 mmol, 3.0 eq) in acetonitrile (14 mL) was added HBTU (78 mg, 0.206 mmol, 2.0 eq) under nitrogen atmosphere. The reaction mixture was stirred at 25 ℃ overnight and then washed with DCM and ethyl ether to produce crude solid support.
To a solution of acetic anhydride (6.2 mL) , pyridine (12 mL) , NEt3 (186 μL) in acetonitrile (7.9 mL) was added the crude solid support under nitrogen atmosphere. The reaction mixture was stirred at 25 ℃ overnight and then washed with DCM and ethyl ether to produce compound B5 of the present disclosure (1.78 g) .
Example 18: The preparation of compound B6 of the present disclosure.
The compound B6 of the present disclosure was prepared in this Example by using the following procedures.
(1) Compound B22 was originated from the steps (1) - (9) of Example 13.
(2) Compound B37 was generated by reduction of compound B22 and then acylation with 6-aminohexan-1-ol
To a solution of compound B22 (1.0 g, 0.54 mmol, 1.0 eq) in MeOH (25 mL) , was added Pd/C (100 mg) under nitrogen atmosphere slowly. Then the reaction atmosphere was
replaced with hydrogen gas for three times. After that, the reaction mixture was stirred by purging with a hydrogen balloon at room temperature overnight. Then the reaction mixture was filtered and concentrated under reduced pressure to provide a crude product.
The crude product was dissolved in DCM (20 mL) , then HBTU (410 mg, 1.08 mmol, 2.0 eq) and DIPEA (305 μL, 1.84 mmol, 3.4 eq) were added into the reaction mixture under nitrogen atmosphere. Five minutes later, 6-aminohexan-1-ol (70 mg, 0.594 mmol, 1.1 eq) was added into the reaction mixture. The reaction mixture was stirred overnight at room temperature. Then H2O (20 mL) was added into the reaction mixture, and the mixture was extracted with DCM (3*50 mL) . The organic phase was combined, dried over Na2SO4, and concentrated. The resultant residue was purified with flash chromatography (silica gel, gradient eluent: 1-10%of MeOH/DCM) to provide compound B37 (0.9 g, 90%yield) as white solid. The product was characterized with mass spectrometry and 1H NMR. MW calc.: 1873.98; MW Found: 1874.16 [M+H] +. 1H NMR (400 MHz, CDCl3) δ 8.11 (s, 1H) , 7.76 (d, J = 8.5 Hz, 1H) , 7.40 (d, J = 8.4 Hz, 1H) , 5.39 -5.26 (m, 9H) , 4.73 -4.66 (m, 3H) , 4.14 (dt, J = 18.0, 11.1 Hz, 8H) , 3.95 -3.88 (m, 4H) , 3.64 (d, J = 4.4 Hz, 4H) , 3.47 (dd, J = 14.8, 8.0 Hz, 6H) , 3.25 (dd, J = 13.8, 6.8 Hz, 6H) , 2.14 (s, 9H) , 2.04 (d, J = 3.4 Hz, 9H) , 2.00 -1.98 (m, 9H) , 1.93 (s, 9H) , 1.68 (s, 10H) , 1.60 -1.53 (m, 14H) , 1.44 (s, 8H) , 1.27 (d, J = 12.4 Hz, 24H) .
(3) The compound B6 of the present disclosure was prepared with compound B37.
To a solution of compound B37 (0.9 g, 0.48 mmol, 1.0 eq) and DIPEA (238 μL, 1.44 mmol, 3.0 eq) in anhydrous DCM (10 mL) under nitrogen atmosphere, was added 3- ( (chloro (diisopropylamino) phosphanyl) oxy) propanenitrile (341 mg, 1.44 mmol, 3.0 eq. ) at 25℃. The reaction mixture was stirred for 1 h. The mixture was extracted two times with DCM, then washed with brine and dried with anhydrous Na2SO4. The organic layer was
concentrated under reduced pressure and the resultant residue was purified with flash chromatography (silica gel, gradient eluent: 1-8%of MeOH/DCM, 1%Et3N) to provide compound B6 of the present disclosure (495 mg, 50%yield) as white solid. The product was characterized with mass spectrometry and 1H NMR. MW calc.: 2074.09; MW Found: 987.54 [M -diisopropylamine] /2. 1H NMR (400 MHz, CDCl3) δ 8.11 (s, 1H) , 7.75 (d, J = 8.4 Hz, 1H) , 7.40 (d, J = 8.5 Hz, 1H) , 5.38 -5.18 (m, 8H) , 4.74 -4.63 (m, 3H) , 4.23 -4.06 (m, 8H) , 4.05 -3.94 (m, 4H) , 3.95 -3.88 (m, 4H) , 3.87 -3.73 (m, 5H) , 3.64 -3.54 (m, 4H) , 3.47 (dd, J = 13.8, 6.6 Hz, 5H) , 2.64 (t, J = 6.4 Hz, 6H) , 2.17 (d, J = 5.8 Hz, 6H) , 2.13 (s, 9H) , 2.03 (d, J = 2.9 Hz, 9H) , 2.00 -1.98 (m, 9H) , 1.93 (s, 9H) , 1.56 (dd, J = 26.2, 20.3 Hz, 16H) , 1.42 (d, J = 7.3 Hz, 6H) , 1.27 (d, J = 11.1 Hz, 24H) , 1.18 (d, J = 2.3 Hz, 6H) , 1.16 (d, J = 2.3 Hz, 6H) , 1.10 (t, J = 7.1 Hz, 6H) .
Example 19: The preparation of compound tC2 of the present disclosure.
(1) Compound C3 was prepared according to the following procedures:
The dicarboxylic acid (20 g, 86.8 mmol) was dissolved/suspended in dry CH2Cl2 (100 mL) . Then oxalyl chloride (16.2 mL, 190.96 mmol) and DMF (5 drops) were added to the solution. The reaction mixture was stirred at room temperature for 3 h, then concentrated under reduced pressure to provide crude compound C2, which was directly used in the next step without further purification.
A solution of benzyl alcohol (9.39 g, 86.8 mmol) and Et3N (12 mL, 86.8 mmol) in THF (200 mL) was added dropwise to an ice-cold solution of compound C2 (86.8 mmol) in THF (100 mL) over 2 hours. Then the solution was warmed to room temperature and stirred overnight. A mixture of H2O (80 mL) , Et3N (12 mL, 86.8 mmol) and THF (80 mL) was added slowly to the solution over 1 hour, and the stirring was continued for 2 hours. THF was then removed and 20 mL H2O was added to the residue. The mixture was extracted by ethyl ether (3*100 mL) and the organic phase was combined, dried over Na2SO4, and concentrated. Ethyl acetate (20 mL) was added to the residue and the suspension was filtered to remove dodecanedioic acid. Concentration of the filtrate followed by chromatography (Hexane: Ethyl
acetate = 4 : 1 to 2: 1) afforded 12.5 g (45%yield) compound C3 as white solid. MW calc.: 320.20; MW Found: 319.02 [M-H] -.
(2) Compound C6 was synthesized by the following procedures:
To a solution of Fmoc-L-hydroxyproline compound C4 (13.3 g, 37.6 mmol) in anhydrous THF (250 mL) , was added borane-methyl sulfide complex (6.16 g, 80 mmol) slowly at room temperature. The reaction mixture was stirred for 5 min at room temperature and then heated to reflux for about 1 h. Methanol (15 mL) was carefully added to the reaction mixture, after which the substance in the vessel was refluxed for 15 min. After that, the reaction mixture was concentrated under reduced pressure. Then the crude products were evaporated three times with methanol (100 mL each) to remove boron-related impurity. The obtained crude product compound C5 was directly used in the next step without further purification.
To a solution of compound C5 (37.6 mmol) in anhydrous pyridine (200 mL) , was added DMTrCl (14 g, 41.4 mmol) slowly at ice bath. The reaction mixture was stirred at nitrogen atmosphere overnight and then concentrated under reduced pressure. The crude product was dissolved in dry MeCN (300 mL) , after that, Et3N (112.8 mmol, 15.6 mL) was added into the mixture and then the reaction mixture was heated to 60 ℃ and kept at that temperature for 4 h. After concentrated under reduced pressure, the resulting residue was purified by flash chromatography (silica gel, gradient eluent: 1-8%of MeOH/DCM) to provide compound C6 (7.57 g, 48%yield) as yellow solid. 1H NMR (400 MHz, CDCl3) δ 7.41 (d, J = 7.4 Hz, 2H) , 7.30 (d, J = 8.8 Hz, 4H) , 7.28 -7.22 (m, 2H) , 7.18 (t, J = 7.2 Hz, 1H) , 6.80 (d, J = 8.8 Hz, 4H) , 4.34 (s, 1H) , 3.75 (d, J = 11.1 Hz, 6H) , 3.60 (dd, J = 12.7, 6.7 Hz, 1H) , 3.10 -2.92 (m, 5H) , 2.86 (d, J = 11.5 Hz, 1H) , 1.85 (dd, J = 13.5, 7.1 Hz, 1H) , 1.63 (ddd, J = 13.7, 7.9, 5.9 Hz, 1H) .
(3) The compound C9 was prepared by the following procedures:
To a solution of 2-amino-2- (hydroxymethyl) propane-1, 3-diol compound C7 (25 g, 206 mmol) in DMSO (50 mL) , was added sodium hydroxide solution (0.83 g NaOH in 4 mL H2O) slowly. Then the 3- (tert-butoxy) -3-oxoprop-1-en-1-ylium (90 g, 700 mmol) was added slowly under nitrogen. The reaction was stirred at room temperature for 2 days. Then 100 mL H2O was added into the reaction, the mixture was extracted by ethyl acetate (3*150 mL) and the organic phase was combined, dried over Na2SO4, and concentrated. The resulting residue was purified by flash chromatography (silica gel, gradient eluent: 1-5%of MeOH/DCM) to provide compound C9 (42 g, 40%yield) as colorless oil.
(4) The Compound C10 was prepared by the following procedures:
To a solution of 12- (benzyloxy) -12-oxododecanoic acid compound C3 (3.3 g, 10.3 mmol) in DCM (50 mL) , was added HBTU (7.81 g, 20.6 mmol) and DIPEA (5.1 mL, 30.9 mmol) under nitrogen atmosphere. 5 minutes later, compound C9 (7.8 g, 15.45 mmol) was added into the reaction system. After that, the reaction mixture was stirred at room temperature for 4 h. Then 50 mL H2O was added into the reaction system, the mixture was extracted with DCM (3*100 mL) and the organic phase was combined, dried over Na2SO4, and concentrated. The resulting residue was purified with flash chromatography (silica gel, gradient eluent: 1-30%of EA/HX) to provide compound C10 (7.16 g, 86%yield) as colorless oil. 1H NMR (400 MHz, CDCl3) δ 7.42 -7.27 (m, 5H) , 6.00 (s, 1H) , 5.11 (s, 2H) , 3.70 (s, 6H) , 3.64 (t, J = 6.3 Hz, 6H) ,
2.44 (t, J = 6.3 Hz, 6H) , 2.34 (t, J = 7.5 Hz, 2H) , 2.17 -2.09 (m, 2H) , 1.59 (d, J = 11.6 Hz, 4H) , 1.45 (s, 27H) , 1.26 (s, 12H) .
(5) The compound 12 was prepared by the following procedures:
To a solution of compound C10 (7.0 g, 8.67 mmol) in DCM (100 mL) , was added CF3COOH (50 mL) under nitrogen atmosphere. The reaction mixture was stirred overnight at room temperature, then concentrated under reduced pressure to provide crude product as colorless oil. After that, the colorless oil was dissolved in 100 mL DCM, then HBTU (19.73 g, 52.02 mmol) and DIPEA (13 mL, 78.03 mmol) were added into the reaction system under nitrogen atmosphere. After Five minutes, tert-butyl (3-aminopropyl) carbamate compound C11 (6.8 g, 39 mmol) was added into the reaction system. The reaction mixture was stirred overnight at room temperature. Then 50 mL H2O was added into the reaction system, the mixture was extracted with DCM (3*100 mL) and the organic phase was combined, dried over Na2SO4, and concentrated. The resulting residue was purified with flash chromatography (silica gel, gradient eluent: 1-8%of MeOH/DCM) to provide compound C12 (6.7 g, 70%yield) as colorless oil.
(6) The compound C14 was prepared by using the following procedures:
To a solution of the compound C12 (1.0 g, 0.9 mmol, 1.0 eq) in DCM (8 mL) , was added HCl/Dioxane (4M, 8 mL) . The reaction mixture was stirred at room temperature for 3 h, then concentrated under reduced pressure to provide crude product as yellow solid. After that, the crude product was dissolved in 15 mL DCM, then compound C13 (1.81 g, 4.05 mmol, 4.5 eq) , HBTU (1.53 g, 4.05 mmol, 4.5 eq) and DIPEA (1.79 mL, 10.8 mmol, 12.0 eq) were added into the reaction system under nitrogen atmosphere. The reaction mixture was stirred overnight at room temperature. Then 10 mL H2O was added into the reaction mixture. The mixture was extracted with DCM (3*20 mL) . The organic phase was combined, dried over Na2SO4, and concentrated. The resultant residue was purified with flash chromatography (silica gel, gradient eluent: 1-8%of MeOH/DCM) to provide compound C14 (0.99 g, 52%yield) as white solid. The product was characterized with 1H NMR. 1H NMR (400 MHz, CDCl3) 1H NMR (400 MHz, CDCl3) δ 7.37 -7.32 (m, 5H) , 5.34 (d, J = 2.9 Hz, 3H) , 5.29 (s, 2H) , 5.19 (dd, J = 11.2, 3.2 Hz, 3H) , 5.10 (s, 2H) , 4.59 (d, J = 8.4 Hz, 3H) , 4.12 (dd, J = 13.4, 5.8 Hz, 6H) , 3.90 (d, J = 6.2 Hz, 4H) , 3.67 (s, 12H) , 3.47 (s, 6H) , 3.26 (d, J = 5.3 Hz, 10H) , 3.06 (q, J = 7.4 Hz, 4H) , 2.42 (t, J = 5.4 Hz, 6H) , 2.34 (t, J = 7.5 Hz, 3H) , 2.22 (dd, J = 16.2, 7.7 Hz, 5H) , 2.13 (s, 9H) , 2.03 (s, 9H) , 1.98 (s, 9H) , 1.94 (s, 9H) , 1.40 -1.35 (m, 22H) , 1.27 -1.22 (m, 12H) .
(7) The compound C15 was prepared by using the following procedures:
To a solution of compound C14 (0.98 g, 0.47 mmol) in MeOH (10 mL) , was added Pd/C (98 mg) under nitrogen atmosphere slowly. Then the reaction atmosphere was replaced with hydrogen gas for three times. After that, the reaction mixture was stirred by purging with a hydrogen balloon at room temperature overnight. Then the reaction mixture was filtered and
concentrated under reduced pressure to provide crude product. The crude product was dissolved in DCM (20 mL) , then HBTU (267 mg, 0.705 mmol, 1.5 eq) and DIPEA (265 μL, 1.598 mmol, 3.4 eq) were added into the reaction system under nitrogen atmosphere. After five minutes, compound C6 (236 mg, 0.564 mmol, 1.2 eq) was added into the reaction mixture. The reaction mixture was stirred overnight at room temperature. Then H2O (10 mL) was added into the reaction mixture. The mixture was extracted with DCM (3*20 mL) . The organic phase was combined, dried over Na2SO4, and concentrated. The resultant residue was purified with flash chromatography (silica gel, gradient eluent: 1-10%of MeOH/DCM) to provide compound C15 (0.79 g, 69%yield) as yellow solid. The product was characterized with 1H NMR. 1H NMR (400 MHz, CDCl3) δ 7.34 (d, J = 8.5 Hz, 2H) , 7.25 -7.20 (m, 5H) , 6.97 -6.89 (m, 2H) , 6.80 (t, J = 8.6 Hz, 4H) , 5.33 (s, 3H) , 5.17 (ddd, J = 11.0, 7.7, 3.2 Hz, 3H) , 4.60 (t, J = 9.1 Hz, 3H) , 4.19 -4.06 (m, 9H) , 3.95 -3.85 (m, 6H) , 3.78 (d, J = 3.5 Hz, 6H) , 3.67 (s, 12H) , 3.48 (t, J = 10.8 Hz, 4H) , 3.26 (s, 12H) , 2.42 (t, J = 5.2 Hz, 6H) , 2.24 (dd, J = 26.4, 11.2 Hz, 11H) , 2.14 (s, 9H) , 2.03 (s, 9H) , 1.98 (s, 9H) , 1.93 (s, 9H) , 1.72 -1.51 (m, 22H) , 1.38 -1.31 (m, 6H) , 1.27 (d, J = 8.6 Hz, 12H) .
(8) The compound C16 was prepared by using the following procedures:
To a solution of compound C15 (780 mg, 0.324 mmol, 1.0 eq) in anhydrous DCM (6 mL) were added DMAP (138 mg, 1.13 mmol, 3.5 eq) and succinic anhydride (97 mg, 0.97 mmol, 3.0 eq) under nitrogen atmosphere. The reaction mixture was stirred at room temperature overnight, and then H2O (10 mL) was added into the reaction system. The mixture was extracted with DCM (3*20 mL) . The organic phase was combined, dried over Na2SO4, and concentrated. The resultant residue was purified with flash chromatography (silica gel,
gradient eluent: 1-15%of MeOH/DCM) to provide compound C16 (690 mg, 85%yield) as white solid. The product was characterized with 1H NMR. 1H NMR (400 MHz, CDCl3) δ 7.38 -7.31 (m, 4H) , 7.21 -7.12 (m, 5H) , 6.80 (dd, J = 13.7, 5.2 Hz, 4H) , 5.33 (d, J = 2.7 Hz, 3H) , 5.19 (d, J = 11.2 Hz, 3H) , 4.63 (dd, J = 8.2, 4.9 Hz, 3H) , 4.16 -4.07 (m, 9H) , 3.78 (s, 6H) , 3.77 (s, 6H) , 3.67 (s, 12H) , 3.51 -3.45 (m, 4H) , 3.25 (s, 12H) , 2.58 (d, J = 7.7 Hz, 4H) , 2.43 (t, J = 5.2 Hz, 6H) , 2.25 (td, J = 15.1, 7.7 Hz, 11H) , 2.13 (s, 9H) , 2.03 (s, 9H) , 1.97 (s, 9H) , 1.94 (s, 9H) , 1.83 -1.51 (m, 24H) , 1.46 -1.29 (m, 6H) , 1.26 (d, J = 12.0 Hz, 12H) .
(9) The compound tC2 was prepared by using the following procedures:
To a solution of compound C16 (690 mg, 0.275 mmol, 1.0 eq) , Controlled Pore Glass (CPG) (Code: C3006-1000, Hebei DNA chem Biotechnology Co., Ltd, China) (4.8 g) , N, N-Diisopropylethylamine (DIPEA) (137 μL, 0.825 mmol, 3.0 eq) in acetonitrile (40 mL) and HBTU (208 mg, 0.55 mmol, 2.0 eq) were added under nitrogen atmosphere. The reaction mixture was shaked at 25 ℃ overnight and then washed with DCM and ethyl ether to produce crude support material. To a solution of acetic anhydride (15 mL) , pyridine (34 mL) , NEt3 (500 μL) in acetonitrile (22 mL) was added the crude support material under nitrogen atmosphere. The reaction mixture was shaked at 25 ℃ 1h and then washed with DCM and ethyl ether in sequence to produce Compound tC2 of the present disclosure (4.7 g) .
Example 20. The preparation of compound D1 of the present disclosure
Compound D1 was prepared in this Example by using the following procedures.
(1) The preparation of compound 2
To a solution of methyl 2- (4-fluoro-3-nitrophenyl) acetate 1 (5 g, 23.4 mmol, 1.0 eq) and K2CO3 (4.85 g, 35.1 mmol, 1.5 eq) in anhydrous DMF (40 mL) , under nitrogen atmosphere, was added 25 wt%methylamine in water (4.36 mL, 35.1 mmol, 1.5 eq) . The reaction mixture was stirred at room temperature for 3 h, then cold water (80 mL) was added. The mixture was extracted three times with ethyl acetate, then the organic phase was washed three times with saturated LiCl solution and one time with brine. Then the organic phase was dried over anhydrous Na2SO4 and concentrated under reduced pressure to form compound 2 as red solid which was directly used in the next step without further purification. The compound 2 was characterized with mass spectrometry. MW calc.: 224.08; MW Found: 225.17 [M+H] +.
(2) The preparation of compound 3
To a solution of compound 2 (23.4 mmol, 1.0 eq) in THF/H2O (9: 1, 111 mL) , under ice bath, was added HCOONH4 (8.86 g, 140.4 mmol, 6.0 eq) and Zn powder (9.18 g, 140.4 mmol, 6.0 eq) . After stirred for 10 minutes, the reaction mixture was moved from the ice bath and stirred at room temperature overnight. Then the reaction mixture was filtered and concentrated under reduced pressure. After that, water (100 mL) was added into the mixture, then the mixture was extracted three times with ethyl acetate, the organic phase was washed one time with brine. After dried over anhydrous Na2SO4 and concentrated under reduced pressure, the product 3 formed then was directly used in the next step without further purification. The
compound 3 was characterized with mass spectrometry. MW calc.: 194.11; MW Found: 195.26 [M+H] +.
(3) The preparation of compound 5
To a solution of compound 3 (23.4 mmol, 1.0 eq) in EtOH (50 mL) under nitrogen atmosphere, was added compound 4 (2.39 g, 23.4 mmol, 1.0 eq) and AcOH (5.39 mL, 93.6 mmol, 4.0 eq) . The reaction mixture was stirred at 80℃ overnight, and then concentrated under reduced pressure. Then a saturated NaHCO3 solution (50 mL) was added, the mixture was extracted three times with ethyl acetate, then the organic phase was combined and washed with brine, dried over Na2SO4, and concentrated. The resultant residue was purified with flash chromatography (silica gel, gradient eluent: 1-5%of MeOH/DCM) to provide compound 5 (2.64 g, 41%yield) . The product was characterized with mass spectrometry and 1H NMR. MW calc.: 276.15; MW Found: 277.25 [M + H] +. 1H NMR (400 MHz, CDCl3) δ 7.58 (s, 1H) , 7.30 -7.20 (m, 1H) , 7.17 (d, J = 8.3 Hz, 1H) , 3.73 (s, 2H) , 3.70 -3.63 (m, 8H) , 2.90 (t, J = 7.2 Hz, 2H) , 2.09 -1.94 (m, 2H) , 1.71 (dt, J = 13.0, 6.3 Hz, 2H) .
(4) The preparation of compound 6
To a solution of compound 5 (2.6 g, 9.41 mmol, 1.0 eq) and triethylamine (TEA, 1.43 g, 14.12 mmol, 1.5 eq) in DCM (20 mL) under nitrogen atmosphere, was added DMTrCl (3.5 g, 10.35 mmol, 1.1 eq) . The reaction mixture was stirred at room temperature overnight, after which it was concentrated under reduced pressure. Then a saturated NaHCO3 solution (50 mL) was added, the mixture was extracted three times with ethyl acetate, then the organic phase was combined and washed with brine, dried over Na2SO4, and concentrated. The resultant residue 6 was directly used in the next step without further purification. The product was characterized with mass spectrometry. MW calc.: 578.28; MW. Found: 579.30 [M + H] +.
(5) The preparation of compound 39
To a solution of compound 6 (9.41 mmol, 1.0 eq) in anhydrous THF (50 mL) under nitrogen atmosphere and ice bath, was added LiAlH4 (536 mg, 14.12 mmol, 1.5 eq) . The mixture was moved to room temperature after 10 minutes and stirred for 1 h. Then the reaction was moved to ice bath, saturated potassium sodium tartrate solution (10 mL) was added slowly into the mixture. After 30 minutes, the reaction was extracted three times with ethyl acetate, then the organic phase was combined and washed with brine, dried over Na2SO4, and concentrated. The resultant residue was purified with flash chromatography (silica gel, gradient eluent: 1-5%of MeOH/DCM) to provide compound 39 (2.4 g, 46%yield) . The product was characterized with mass spectrometry and 1H NMR. MW calc.: 550.28; MW Found: 551.63 [M + H] +. 1H NMR (400 MHz, CDCl3) δ 7.54 (s, 1H) , 7.46 -7.40 (m, 2H) , 7.34 -7.28 (m, 4H) , 7.25 (dd, J = 8.3, 5.7 Hz, 2H) , 7.19 (d, J = 8.4 Hz, 2H) , 7.10 (dd, J = 8.2, 1.3 Hz, 1H) , 6.84 -6.76 (m, 4H) , 3.87 (t, J = 6.7 Hz, 2H) , 3.76 (s, 6H) , 3.62 (s, 3H) , 3.11 (t, J = 6.3 Hz, 2H) , 2.96 (t, J = 6.7 Hz, 2H) , 2.87 -2.80 (m, 2H) , 1.99 -1.89 (m, 2H) , 1.83 -1.70 (m, 2H) .
(6) The preparation of compound D1
The compound 39 (2.3 g, 4.18 mmol, 1.0 eq) and Diisoropyl ammonium tetrazolide (2.15 g, 12.54 mmol, 3.0 eq) were dissolved in anhydrous DCM (40 mL) under nitrogen atmosphere was added 3- ( (Bis (diisopropylamino) phosphino) oxy) propanenitrile (3.78 g, 12.54 mmol, 3.0 eq) at room temperature. The reaction mixture was stirred for 6 h. The mixture was extracted two times with DCM, then washed with brine and dried with anhydrous Na2SO4. The organic layer was concentrated under reduced pressure and the resultant residue was purified with flash chromatography (silica gel, gradient eluent: 1-5%of MeOH/DCM, 1%Et3N) to
provide compound D1 (2.9 g, 91%yield) as yellow oil. The product was characterized with mass spectrometry and 1H NMR. MW calc.: 750.39; MW Found: 303.22 [DMT] -. 1H NMR (400 MHz, CDCl3) δ 7.52 (s, 1H) , 7.46 -7.40 (m, 2H) , 7.34 -7.29 (m, 4H) , 7.28 -7.23 (m, 2H) , 7.22 -7.16 (m, 2H) , 7.11 (dd, J = 8.2, 1.4 Hz, 1H) , 6.83 -6.77 (m, 4H) , 3.78 (s, 6H) , 3.76 -3.72 (m, 2H) , 3.64 (s, 3H) , 3.61 -3.48 (m, 4H) , 3.11 (t, J = 6.3 Hz, 2H) , 3.04 (t, J = 7.3 Hz, 2H) , 2.87 -2.80 (m, 2H) , 2.57 -2.47 (m, 2H) , 2.03 -1.90 (m, 2H) , 1.79 -1.72 (m, 2H) , 1.19 (s, 3H) , 1.18 (s, 3H) , 1.16 (s, 3H) , 1.14 (s, 3H) .
Example 21. The preparation of compound D2 of the present disclosure
Compound D2 was prepared in this Example by using the following procedures.
(1) The preparation of compound 8
To a solution of methyl 2- (4-fluoro-3-nitrophenyl) acetate 1 (5 g, 23.4 mmol, 1.0 eq) and K2CO3 (3.56 g, 25.74 mmol, 1.1 eq) in anhydrous DMF (40 mL) , under nitrogen atmosphere, was added compound 7 (3.58 g, 25.74 mmol, 1.1 eq) . The reaction mixture was stirred at room temperature overnight, then cold water (100 mL) was added. The mixture was extracted three times with ethyl acetate, then the organic phase was washed three times with saturated LiCl solution and one time with brine. Then the organic phase was dried over anhydrous Na2SO4 and concentrated under reduced pressure to form compound 8 as yellow oil which was directly used in the next step without further purification. The product was characterized with mass spectrometry. MW calc.: 332.15; MW Found: 333.22 [M + H] +.
(2) The preparation of compound 9
To a solution of compound 8 (23.4 mmol, 1.0 eq) in THF/H2O (9: 1, 111 mL) , under ice bath, was added HCOONH4 (8.86 g, 140.4 mmol, 6.0 eq) and Zn powder (9.18 g, 140.4 mmol, 6.0 eq) . After stirred for 10 minutes, the reaction mixture was moved from the ice bath and stirred at room temperature overnight. Then the reaction mixture was filtered and concentrated under reduced pressure. After that, water (200 mL) was added into the mixture, then the mixture was extracted three times with ethyl acetate, the organic phase was washed one time with brine. After dried over anhydrous Na2SO4 and concentrated under reduced pressure, the product 9 formed then was directly used in the next step without further purification. The compound 9 was characterized with mass spectrometry. MW calc.: 302.17; MW Found: 303.22 [M + H] +.
(3) The preparation of compound 10
To a solution of compound 9 (23.4 mmol, 1.0 eq) in EtOH (50 mL) under nitrogen atmosphere, was added 5-Hydroxypentanal 4 (2.39 g, 23.4 mmol, 1.0 eq) and AcOH (5.39 mL, 93.6 mmol, 4.0 eq) . The reaction mixture was stirred at 80℃ for 4 h, and then concentrated under reduced pressure. Then a saturated NaHCO3 solution (100 mL) was added, the mixture was extracted three times with ethyl acetate, then the organic phase was combined and washed with brine, dried over Na2SO4, and concentrated. The product 10 formed then was directly used in the next step without further purification. The product was characterized with mass spectrometry. MW calc.: 384.22; MW Found: 385.30 [M + H] +.
(4) The preparation of compound 11
To a solution of compound 10 (8.27 mmol, 1.0 eq) and triethylamine (TEA, 1.26 g, 12.41 mmol, 1.5 eq) in DCM (20 mL) under nitrogen atmosphere, was added DMTrCl (3.36 g, 9.92 mmol, 1.2 eq) . The reaction mixture was stirred at room temperature for 2 h, after which it was concentrated under reduced pressure. Then a saturated NaHCO3 solution (50 mL) was added, the mixture was extracted three times with ethyl acetate, then the organic phase was combined and washed with brine, dried over Na2SO4, and concentrated. The resultant residue 11 was directly used in the next step without further purification. The product was characterized with mass spectrometry. MW calc.: 686.35; MW. Found: 385.27 [M -DMT + H] +.
(5) The preparation of compound 40
To a solution of compound 11 (8.27 mmol, 1.0 eq) in anhydrous THF (40 mL) under nitrogen atmosphere and ice bath, was added LiAlH4 (471 mg, 12.4 mmol, 1.5 eq) . The mixture was moved to room temperature after 10 minutes and stirred for 1 h. Then the reaction was moved to ice bath, saturated potassium sodium tartrate solution (20 mL) was added slowly into the mixture. After 30 minutes, the reaction was extracted three times with ethyl acetate, then the organic phase was combined and washed with brine, dried over Na2SO4, and concentrated. The resultant residue was purified with flash chromatography (silica gel, gradient eluent: 1-8%of MeOH/DCM) to provide compound 40 (1.15 g, 21%yield) . The
product was characterized with mass spectrometry and 1H NMR. MW calc.: 658.35; MW Found: 357.27 [M -DMT + H] +. 1H NMR (400 MHz, CDCl3) δ 7.56 (s, 1H) , 7.44 -7.40 (m, 2H) , 7.32 -7.28 (m, 4H) , 7.26 (s, 2H) , 7.19 (dd, J = 7.8, 3.7 Hz, 1H) , 7.10 (dd, J = 8.2, 1.4 Hz, 1H) , 7.01 (d, J = 8.2 Hz, 1H) , 6.95 (d, J = 8.3 Hz, 1H) , 6.82 -6.78 (m, 4H) , 6.77 (d, J = 1.3 Hz, 1H) , 4.07 -4.00 (m, 2H) , 3.88 (t, J = 6.6 Hz, 2H) , 3.81 (d, J = 7.0 Hz, 2H) , 3.77 (s, 6H) , 3.11 (t, J = 6.2 Hz, 2H) , 2.97 (t, J = 6.6 Hz, 2H) , 2.78 -2.70 (m, 2H) , 2.25 (s, 3H) , 2.22 -2.15 (m, 2H) , 1.98 (dd, J = 15.2, 7.4 Hz, 2H) , 1.80 -1.70 (m, 2H) .
(6) The preparation of compound D2
The compound 40 (1.1 g, 1.67 mmol, 1.0 eq) and Diisoropyl ammonium tetrazolide (858 mg, 5.01 mmol, 3.0 eq) were dissolved in anhydrous DCM (15 mL) under nitrogen atmosphere was added 3- ( (Bis (diisopropylamino) phosphino) oxy) propanenitrile (1.51 g, 5.01 mmol, 3.0 eq) at room temperature. The reaction mixture was stirred for 6 h. The mixture was extracted two times with DCM, then washed with brine and dried with anhydrous Na2SO4. The organic layer was concentrated under reduced pressure and the resultant residue was purified with flash chromatography (silica gel, gradient eluent: 1-5%of MeOH/DCM, 1%Et3N) to provide compound D2 (1.15 g, 80%yield) . The product was characterized with mass spectrometry and 1H NMR. MW calc.: 858.46; MW Found: 303.22 [DMT] -. 1H NMR (400 MHz, CDCl3) δ 7.54 (s, 1H) , 7.45 -7.39 (m, 2H) , 7.33 -7.28 (m, 4H) , 7.27 (d, J = 8.0 Hz, 2H) , 7.22 -7.17 (m, 1H) , 7.10 (dd, J = 8.2, 1.4 Hz, 1H) , 7.02 (d, J = 8.2 Hz, 1H) , 6.95 (d, J = 8.3 Hz, 1H) , 6.85 -6.78 (m, 4H) , 6.78 (d, J = 1.3 Hz, 1H) , 4.21 -4.11 (m, 2H) , 4.07 -4.00 (m, 2H) , 3.81 (d, J = 7.0 Hz, 2H) , 3.78 (s, 6H) , 3.57 -3.51 (m, 2H) , 3.11 (t, J = 6.3 Hz, 2H) , 3.03 (t, J = 7.2 Hz, 2H) , 2.79 -2.72 (m, 4H) , 2.61 -2.51 (m, 2H) , 2.26 (s, 3H) , 2.22 -2.13 (m, 2H) , 2.02 -1.95 (m, 2H) , 1.80 -1.69 (m, 2H) , 1.19 -1.11 (m, 12H) .
Example 22. The preparation of compound D3 of the present disclosure
Compound D3 was prepared in this Example by using the following procedures.
(1) The preparation of compound 13
To a solution of methyl 2- (4-fluoro-3-nitrophenyl) acetate 1 (5 g, 23.4 mmol, 1.0 eq) and K2CO3 (3.56 g, 25.74 mmol, 1.1 eq) in anhydrous DMF (40 mL) , under nitrogen atmosphere, was added compound 12 (2.65 g, 25.74 mmol, 1.1 eq) . The reaction mixture was stirred at 50 ℃ overnight, then cold water (100 mL) was added. The mixture was extracted three times with ethyl acetate, then the organic phase was washed three times with saturated LiCl solution and one time with brine. Then the organic phase was dried over anhydrous Na2SO4 and concentrated under reduced pressure to form compound 13 as yellow oil which was directly used in the next step without further purification. The product was characterized with mass spectrometry. MW calc.: 296.14; MW Found: 297.77 [M + H] +.
(2) The preparation of compound 14
To a solution of compound 47 (23.4 mmol, 1.0 eq) and triethylamine (TEA, 3.55 g, 35.1 mmol, 1.5 eq) in DCM (50 mL) under nitrogen atmosphere, was added DMTrCl (8.72 g, 25.74 mmol, 1.1 eq) . The reaction mixture was stirred at room temperature for 3 h, after which it was concentrated under reduced pressure. Then a saturated NaHCO3 solution (50 mL) was added, the mixture was extracted three times with ethyl acetate, then the organic phase was combined and washed with brine, dried over Na2SO4, and concentrated. The resultant residue 14 was directly used in the next step without further purification. The product was
characterized with mass spectrometry. MW calc.: 598.27; MW. Found: 303.12 [DMT] -.
(3) The preparation of compound 15
To a solution of compound 14 (23.4 mmol, 1.0 eq) in THF/H2O (9: 1, 111 mL) , under ice bath, was added HCOONH4 (8.86 g, 140.4 mmol, 6.0 eq) and Zn powder (9.18 g, 140.4 mmol, 6.0 eq) . After stirred for 10 minutes, the reaction mixture was moved from the ice bath and stirred at room temperature overnight. Then the reaction mixture was filtered and concentrated under reduced pressure. After that, water (100 mL) was added into the mixture, then the mixture was extracted three times with ethyl acetate, the organic phase was washed one time with brine. After dried over anhydrous Na2SO4 and concentrated under reduced pressure, the product 15 formed then was directly used in the next step without further purification. The compound 15 was characterized with mass spectrometry. MW calc.: 568.29; MW Found: 267.19 [M -DMT + H] +.
(4) The preparation of compound 16
To a solution of compound 15 (23.4 mmol, 1.0 eq) in EtOH (50 mL) under nitrogen atmosphere, was added Paraformaldehyde (2.1 g, 23.4 mmol, 1.0 eq) and AcOH (5.4 mL, 93.6 mmol, 4.0 eq) . The reaction mixture was stirred at 80 ℃ for 3 h, and then concentrated under reduced pressure. Then a saturated NaHCO3 solution (50 mL) was added, the mixture was extracted three times with ethyl acetate, then the organic phase was combined and washed with brine, dried over Na2SO4, and concentrated. The resultant residue was purified with flash chromatography (silica gel, gradient eluent: 1-5%of MeOH/DCM) to provide compound 16 (6.6 g, 49%yield) . The product was characterized with mass spectrometry and 1H NMR. MW calc.: 578.28; MW Found: 579.40 [M + H] +. 1H NMR (400 MHz, CDCl3) δ 7.83 (s, 1H) , 7.69 (s, 1H) , 7.44 -7.37 (m, 2H) , 7.34 -7.24 (m, 7H) , 7.23 -7.17 (m, 2H) , 6.84 -6.77 (m, 4H) ,
4.10 (d, J = 7.2 Hz, 2H) , 3.78 (s, 6H) , 3.75 (s, 2H) , 3.68 (s, 3H) , 3.04 (t, J = 6.4 Hz, 2H) , 1.88 -1.78 (m, 2H) , 1.69 -1.58 (m, 2H) , 1.50 -1.36 (m, 2H) .
(5) The preparation of compound 41
To a solution of compound 16 (6.6 g, 9.32 mmol, 1.0 eq) in anhydrous THF (50 mL) under nitrogen atmosphere and ice bath, was added LiAlH4 (531 mg, 14 mmol, 1.5 eq) . The mixture was moved to room temperature after 10 minutes and stirred for 1 h. Then the reaction was moved to ice bath, saturated potassium sodium tartrate solution (20 mL) was added slowly into the mixture. After 30 minutes, the reaction was extracted three times with ethyl acetate, then the organic phase was combined and washed with brine, dried over Na2SO4, and concentrated. The resultant residue was purified with flash chromatography (silica gel, gradient eluent: 1-5%of MeOH/DCM) to provide compound 41 (5.72 g, 90%yield) . The product was characterized with mass spectrometry and 1H NMR. MW calc.: 550.28; MW Found: 551.46 [M + H] +. 1H NMR (400 MHz, CDCl3) δ 7.82 (s, 1H) , 7.65 (d, J = 0.7 Hz, 1H) , 7.40 (dd, J = 8.3, 3.3 Hz, 2H) , 7.30 -7.24 (m, 7H) , 7.22 -7.12 (m, 2H) , 6.84 -6.77 (m, 4H) , 4.12 (t, J = 7.1 Hz, 2H) , 3.90 (t, J = 6.5 Hz, 2H) , 3.78 (s, 6H) , 3.02 (dt, J = 16.5, 6.5 Hz, 4H) , 1.84 (dt, J = 15.0, 7.3 Hz, 2H) , 1.63 (dd, J = 14.4, 6.8 Hz, 2H) , 1.49 -1.35 (m, 2H) .
(6) The preparation of compound D3
The compound 41 (2 g, 3.63 mmol, 1.0 eq) and Diisoropyl ammonium tetrazolide (1.86 g, 10.89 mmol, 3.0 eq) were dissolved in anhydrous DCM (20 mL) under nitrogen atmosphere
was added 3- ( (Bis (diisopropylamino) phosphino) oxy) propanenitrile (3.28 g, 10.89 mmol, 3.0 eq) at room temperature. The reaction mixture was stirred for 6 h. The mixture was extracted two times with DCM, then washed with brine and dried with anhydrous Na2SO4. The organic layer was concentrated under reduced pressure and the resultant residue was purified with flash chromatography (silica gel, gradient eluent: 1-5%of MeOH/DCM, 1%Et3N) to provide compound D3 (2.48 g, 91%yield) . The product was characterized with mass spectrometry and 1H NMR. MW calc.: 750.39; MW Found: 303.21 [DMT] -. 1H NMR (400 MHz, CDCl3) δ 7.82 (s, 1H) , 7.63 (s, 1H) , 7.42 -7.38 (m, 2H) , 7.30 -7.26 (m, 7H) , 7.20 (dd, J = 7.8, 3.7 Hz, 1H) , 7.17 -7.13 (m, 1H) , 6.83 -6.78 (m, 4H) , 4.11 (t, J = 7.2 Hz, 2H) , 3.78 (s, 6H) , 3.60 -3.50 (m, 4H) , 3.04 (t, J = 6.5 Hz, 4H) , 2.75 (td, J = 6.3, 2.0 Hz, 1H) , 2.55 (td, J = 6.6, 3.5 Hz, 2H) , 1.84 (dt, J = 15.0, 7.4 Hz, 3H) , 1.69 -1.58 (m, 2H) , 1.50 -1.37 (m, 2H) , 1.18 -1.13 (m, 12H) .
Example 23. The preparation of compound D4 of the present disclosure
Compound D4 was prepared in this Example by using the following procedures.
(1) The preparation of compound 18
To a solution of compound 17 (5 g, 25.12 mmol, 1.0 eq) and K2CO3 (3.8 g, 27.6 mmol, 1.1 eq) in anhydrous DMF (40 mL) , under nitrogen atmosphere, was added compound 12 (2.84 g, 27.6 mmol, 1.1 eq) . The reaction mixture was stirred at room temperature overnight, then cold water (100 mL) was added. The mixture was extracted three times with ethyl acetate,
then the organic phase was washed three times with saturated LiCl solution and one time with brine. Then the organic phase was dried over anhydrous Na2SO4 and concentrated under reduced pressure to form compound 18 as yellow oil which was directly used in the next step without further purification. The product was characterized with mass spectrometry. MW calc.: 282.12; MW Found: 283.86 [M + H] +.
(2) The preparation of compound 19
To a solution of compound 18 (25.12 mmol, 1.0 eq) and triethylamine (TEA, 3.81 g, 37.68 mmol, 1.5 eq) in DCM (50 mL) under nitrogen atmosphere, was added DMTrCl (8.51 g, 27.63 mmol, 1.1 eq) . The reaction mixture was stirred at room temperature for 4 h, after which it was concentrated under reduced pressure. Then a saturated NaHCO3 solution (50 mL) was added, the mixture was extracted three times with ethyl acetate, then the organic phase was combined and washed with brine, dried over Na2SO4, and concentrated. The resultant residue 19 was directly used in the next step without further purification. The product was characterized with mass spectrometry. MW calc.: 584.25; MW. Found: 303.12 [DMT] -.
(3) The preparation of compound 20
To a solution of compound 19 (25.12 mmol, 1.0 eq) in THF/H2O (9: 1, 111 mL) , under ice bath, was added HCOONH4 (9.51 g, 150.72 mmol, 6.0 eq) and Zn powder (9.86 g, 150.72 mmol, 6.0 eq) . After stirred for 10 minutes, the reaction mixture was moved from the ice bath and stirred at room temperature overnight. Then the reaction mixture was filtered and concentrated under reduced pressure. After that, water (100 mL) was added into the mixture, then the mixture was extracted three times with ethyl acetate, the organic phase was washed one time with brine. After dried over anhydrous Na2SO4 and concentrated under reduced pressure, the product 20 formed then was directly used in the next step without further purification. The compound 20 was characterized with mass spectrometry. MW calc.: 554.28;
MW Found: 253.19 [M -DMT + H] +.
(4) The preparation of compound 22
To a solution of compound 20 (25.12 mmol, 1.0 eq) in EtOH (140 mL) under nitrogen atmosphere, was added 5-Hydroxypentanal 4 (2.57 g, 25.12 mmol, 1.0 eq) and AcOH (5.8 mL, 100.48 mmol, 4.0 eq) . The reaction mixture was stirred at 80 ℃ for 6 h, and then concentrated under reduced pressure. Then a saturated NaHCO3 solution (100 mL) was added, the mixture was extracted three times with ethyl acetate, then the organic phase was combined and washed with brine, dried over Na2SO4, and concentrated. The resultant residue was purified with flash chromatography (silica gel, gradient eluent: 1-5%of MeOH/DCM) to provide compound 22 (9.4 g, 60%yield) . The product was characterized with mass spectrometry and 1H NMR. MW calc.: 636.32; MW Found: 637.51 [M + H] +. 1H NMR (400 MHz, CDCl3) δ 8.41 (d, J = 1.2 Hz, 1H) , 7.94 (dd, J = 8.5, 1.5 Hz, 1H) , 7.43 -7.37 (m, 2H) , 7.30 -7.25 (m, 6H) , 7.24 (d, J = 1.6 Hz, 1H) , 7.22 -7.16 (m, 1H) , 6.83 -6.78 (m, 4H) , 4.08 (dd, J = 10.1, 4.7 Hz, 2H) , 3.92 (s, 3H) , 3.77 (s, 6H) , 3.67 (t, J = 6.1 Hz, 2H) , 3.04 (t, J = 6.3 Hz, 2H) , 2.88 (t, J = 7.2 Hz, 2H) , 1.78 -1.70 (m, 4H) , 1.66 -1.61 (m, 2H) , 1.53 -1.41 (m, 2H) , 1.25 (t, J = 7.1 Hz, 2H) .
(5) The preparation of compound D4
The compound 22 (2 g, 3.14 mmol, 1.0 eq) and Diisoropyl ammonium tetrazolide (1.61 g, 9.43 mmol, 3.0 eq) were dissolved in anhydrous DCM (20 mL) under nitrogen atmosphere was added 3- ( (Bis (diisopropylamino) phosphino) oxy) propanenitrile (2.85 g, 9.43 mmol, 3.0 eq)
at room temperature. The reaction mixture was stirred for 6 h. The mixture was extracted two times with DCM, then washed with brine and dried with anhydrous Na2SO4. The organic layer was concentrated under reduced pressure and the resultant residue was purified with flash chromatography (silica gel, gradient eluent: 1-5%of MeOH/DCM, 1%Et3N) to provide compound D4 (2.4 g, 93%yield) . The product was characterized with mass spectrometry and 1H NMR. MW calc.: 836.43; MW Found: 303.70 [DMT] -. 1H NMR (400 MHz, CDCl3) δ 8.41 (d, J = 1.2 Hz, 1H) , 7.94 (dd, J = 8.5, 1.5 Hz, 1H) , 7.42 -7.37 (m, 2H) , 7.30 -7.26 (m, 7H) , 7.23 -7.16 (m, 1H) , 6.83 -6.78 (m, 4H) , 4.10 (t, J = 7.4 Hz, 2H) , 3.93 (s, 3H) , 3.78 (s, 6H) , 3.64 -3.49 (m, 5H) , 3.04 (t, J = 6.3 Hz, 2H) , 2.91 -2.85 (m, 2H) , 2.65 -2.55 (m, 3H) , 2.00 (dt, J = 15.3, 7.6 Hz, 2H) , 1.82 -1.76 (m, 4H) , 1.64 (dd, J = 14.2, 6.7 Hz, 2H) , 1.51 -1.44 (m, 2H) , 1.17 -1.13 (m, 12H) .
Example 24. The preparation of compound D5 of the present disclosure
Compound D5 was prepared in this Example by using the following procedures.
(1) The preparation of compound 24
To a solution of compound 1 (50.77 g, 238.3 mmol, 1.0 eq) and K2CO3 (32.9 g, 238.3 mmol, 1.0 eq) in anhydrous DMF (700 mL) , under nitrogen atmosphere, was added compound 23 (57.5 g, 238.3 mmol, 1.0 eq) . The reaction mixture was stirred at room temperature overnight, then cold water (500 mL) was added. The mixture was extracted three times with ethyl acetate, then the organic phase was washed three times with saturated LiCl solution and one time with brine. Then the organic phase was dried over anhydrous Na2SO4 and concentrated under reduced pressure to form compound 24 as yellow oil which was directly
used in the next step without further purification.
(2) The preparation of compound 25
To a solution of compound 24 (15.6 g, 35.87 mmol, 1.0 eq) in anhydrous THF (160 mL) under nitrogen atmosphere and ice bath, was added LiAlH4 (1.36 g, 35.87 mmol, 1.0 eq) . The mixture was moved to room temperature after 10 minutes and stirred for 1 h. Then the reaction was moved to ice bath, saturated potassium sodium tartrate solution (50 mL) was added slowly into the mixture. After 30 minutes, the reaction was extracted three times with ethyl acetate, then the organic phase was combined and washed with brine, dried over Na2SO4, and concentrated.
The resultant residue was dissolved in 140 mL DMF, then imidazole (3.66 g, 53.8 mmol, 1.5 eq) and TBSCl (6.5 g, 43.04 mmol, 1.0 eq) were added under nitrogen atmosphere. The reaction mixture was stirred at room temperature for 2 h, then H2O (100 mL) was added. The mixture was extracted three times by ethyl acetate, then the organic phase was washed three times by saturated LiCl solution and one time by brine and then dried by anhydrous Na2SO4 and concentrated under reduced pressure to form compound 25 as yellow oil which was directly used in the next step without further purification. The product was characterized with mass spectrometry. MW calc.: 520.41; MW Found: 521.31 [M + H] +.
(3) The preparation of compound 26
To a solution of compound 25 (35.87 mmol, 1.0 eq) in THF/H2O (9: 1, 178 mL) , under ice bath, was added HCOONH4 (13.58 g, 215.22 mmol, 6.0 eq) and Zn powder (14.1 g, 215.22 mmol, 6.0 eq) . After stirred for 10 minutes, the reaction mixture was moved from the ice bath and stirred at room temperature overnight. Then the reaction mixture was filtered and concentrated under reduced pressure. After that, water (150 mL) was added into the mixture, then the mixture was extracted three times with ethyl acetate, the organic phase was washed one time with brine. After dried over anhydrous Na2SO4 and concentrated under reduced pressure, the product 26 formed then was directly used in the next step without further purification. The compound 26 was characterized with mass spectrometry. MW calc.: 490.43;
MW Found: 491.40 [M + H] +.
(4) The preparation of compound 42
To a solution of compound 26 (2.0 g, 4.08 mmol, 1.0 eq) in EtOH (20 mL) under nitrogen atmosphere, was added 5-Hydroxypentanal 4 (417 mg, 6.12 mmol, 1.0 eq) and AcOH (0.94 mL, 16.32 mmol, 4.0 eq) . The reaction mixture was stirred at 80 ℃ for 6 h, and then concentrated under reduced pressure. Then a saturated NaHCO3 solution (50 mL) was added, the mixture was extracted three times with ethyl acetate, then the organic phase was combined and washed with brine, dried over Na2SO4, and concentrated. The resultant residue was purified with flash chromatography (silica gel, gradient eluent: 1-5%of MeOH/DCM) to provide compound 42 (1.3 g, 56%yield) . The product was characterized with mass spectrometry. MW calc.: 572.47; MW Found: 573.31 [M + H] +.
(5) The preparation of compound 43
To a solution of compound 42 (1.3 g, 2.26 mmol, 1.0 eq) and triethylamine (TEA, 343 mg, 3.39 mmol, 1.5 eq) in DCM (15 mL) under nitrogen atmosphere, was added DMTrCl (919 mg, 2.71 mmol, 1.2 eq) . The reaction mixture was stirred at room temperature overnight, after which it was concentrated under reduced pressure. Then a saturated NaHCO3 solution (30 mL) was added, the mixture was extracted three times with ethyl acetate, then the organic phase was combined and washed with brine, dried over Na2SO4, and concentrated.
The resultant residue was dissolved in 10 mL THF, then 1 M TBAF THF solution (3.39 mL, 3.39 mmol, 1.5 eq) was added under nitrogen atmosphere. The reaction mixture was stirred at room temperature for 2 h, then H2O (20 mL) was added. The mixture was extracted three times by ethyl acetate, then washed one time by brine, dried by anhydrous Na2SO4 and concentrated under reduced pressure. The resultant residue was purified with flash chromatography (silica gel, gradient eluent: 1-5%of MeOH/DCM) to provide compound 43 (0.8 g, 46%yield) . The product was characterized with mass spectrometry and 1H NMR. MW calc.: 760.52; MW Found: 303.05 [DMT] -. 1H NMR (400 MHz, CDCl3) δ 7.55 (s, 1H) , 7.42 (d,
J = 7.3 Hz, 2H) , 7.33 -7.29 (m, 4H) , 7.26 -7.26 (m, 2H) , 7.24 (d, J = 3.4 Hz, 1H) , 7.21 (d, J = 3.0 Hz, 1H) , 7.09 (dd, J = 8.3, 1.4 Hz, 1H) , 6.80 (d, J = 8.9 Hz, 4H) , 4.05 -3.99 (m, 2H) , 3.92 -3.85 (m, 2H) , 3.78 (s, 6H) , 3.11 (t, J = 6.3 Hz, 2H) , 2.98 (t, J = 6.5 Hz, 2H) , 2.85 -2.79 (m, 2H) , 2.04 -1.94 (m, 2H) , 1.76 (dd, J = 13.9, 7.1 Hz, 4H) , 1.30 -1.25 (m, 27H) , 0.89 -0.85 (m, 3H) .
(6) The preparation of compound D5
The compound 43 (800 mg, 1.05 mmol, 1.0 eq) and Diisoropyl ammonium tetrazolide (360 mg, 2.1 mmol, 2.0 eq) were dissolved in anhydrous DCM (10 mL) under nitrogen atmosphere was added 3- ( (Bis (diisopropylamino) phosphino) oxy) propanenitrile (634 mg, 2.1 mmol, 2.0 eq) at room temperature. The reaction mixture was stirred for 6 h. The mixture was extracted two times with DCM, then washed with brine and dried with anhydrous Na2SO4. The organic layer was concentrated under reduced pressure and the resultant residue was purified with flash chromatography (silica gel, gradient eluent: 1-5%of MeOH/DCM, 1%Et3N) to provide compound D5 (660 mg, 65%yield) . The product was characterized with mass spectrometry and 1H NMR. MW calc.: 960.63; MW Found: 303.04 [DMT] -. 1H NMR (400 MHz, CDCl3) δ 7.52 (s, 1H) , 7.46 -7.40 (m, 2H) , 7.34 -7.28 (m, 4H) , 7.26 (s, 2H) , 7.19 (t, J = 7.3 Hz, 2H) , 7.11 -7.05 (m, 1H) , 6.85 -6.75 (m, 4H) , 4.22 -4.12 (m, 2H) , 4.01 (t, J = 7.4 Hz, 2H) , 3.78 (s, 6H) , 3.61 -3.48 (m, 4H) , 3.11 (t, J = 6.2 Hz, 2H) , 2.85 -2.79 (m, 2H) , 2.76 (td, J = 6.3, 2.0 Hz, 2H) , 2.56 (td, J = 6.6, 3.7 Hz, 1H) , 1.98 (dt, J = 15.3, 7.7 Hz, 2H) , 1.83 -1.67 (m, 4H) , 1.27 -1.21 (m, 27H) , 1.20 -1.13 (m, 12H) , 0.90 -0.84 (m, 3H) .
Example 25. The preparation of compound D6
Compound D6 was prepared in this Example by using the following procedures.
(1) Preparation of compound 5
To a solution of methyl 2- (4-fluoro-3-nitrophenyl) acetate 4 (9.9 g, 46.4 mmol, 1.0 eq) and K2CO3 (6.4 g, 46.4 mmol, 1.0 eq) in anhydrous DMF (80 mL) , under nitrogen atmosphere, was added compound 3 (11.2 g, 46.4 mmol, 1.0 eq) . The reaction mixture was stirred at 55 ℃ for 8 h, then cold water (100 mL) was added. The mixture was extracted three times with ethyl acetate, then the organic phase was washed three times with saturated LiCl solution and one time with brine. Then the organic phase was dried over anhydrous Na2SO4 and concentrated under reduced pressure to form compound 5 as yellow oil which was directly used in the next step without further purification.
(2) Preparation of compound 16
To a solution of compound 5 (15.58 g, 35.87 mmol, 1.0 eq) in anhydrous THF (160 mL) under nitrogen atmosphere and ice bath, was added LiAlH4 (1.36 g, 35.87 mmol, 1.0 eq) slowly. The mixture was moved to room temperature after 10 minutes and stirred for 1 h. Then the reaction was moved to ice bath, saturated potassium sodium tartrate solution (30 mL) was added slowly into the mixture. After 30 minutes, the reaction was extracted three times with EtOAc, then the organic phase was combined and washed with brine, dried over Na2SO4, and concentrated. The crude residue was then dissolved in anhydrous DMF (140 mL) . Then imidazole (3.66 g, 53.8 mmol, 1.5 eq) and TBSCl (6.5 g, 43.04 mmol, 1.2 eq) were added under nitrogen atmosphere. The reaction mixture was stirred at room temperature for 2 h, then
H2O (100 mL) was added. The mixture was extracted three times by ethyl acetate, then the organic phase was washed three times by saturated LiCl solution and one time by brine and then dried by anhydrous Na2SO4 and concentrated under reduced pressure. The resultant residue 16 was directly used in the next step without further purification. The product was characterized with mass spectrometry. MW calc.: 520.41; MW Found: 521.4 [M + H] +.
(3) Preparation of compound 17
To a solution of compound 16 (35.87 mmol, 1.0 eq) in THF/H2O (9: 1, 178 mL) , under ice bath, was added HCOONH4 (13.58 g, 215.2 mmol, 6.0 eq) and Zn powder (14.1 g, 215.2 mmol, 6.0 eq) . After stirred for 10 minutes, the reaction mixture was moved from the ice bath and stirred at room temperature overnight. Then the reaction mixture was filtered and concentrated under reduced pressure. After that, water (200 mL) was added into the mixture, then the mixture was extracted three times with ethyl acetate, the organic phase was washed one time with brine. After dried over anhydrous Na2SO4 and concentrated under reduced pressure, the product 17 formed then was directly used in the next step without further purification. The compound 17 was characterized with mass spectrometry. MW calc.: 490.43; MW Found: 491.40 [M+H] +.
(4) Preparation of compound 18
To a solution of compound 17 (5 g, 10.2 mmol, 1.0 eq) in EtOH (25 mL) under nitrogen atmosphere, was added 5-Hydroxypentanal (1.04 g, 10.2 mmol, 1.0 eq) and AcOH (2.35 mL, 40.8 mmol, 4.0 eq) . The reaction mixture was stirred at 80℃ overnight, and then concentrated under reduced pressure. Then a saturated NaHCO3 solution (25 mL) was added, the mixture was extracted three times with ethyl acetate, then the organic phase was combined and washed with brine, dried over Na2SO4, and concentrated. The resultant residue was purified with flash chromatography (silica gel, gradient eluent: 1-5%of MeOH/DCM) to provide compound 18 (1.9 g, 33%yield) . The product was characterized with mass spectrometry and 1H NMR. MW calc.: 572.47; MW Found: 573.61 [M + H] +. 1H NMR (400 MHz, CDCl3) δ 7.53 (s, 1H) , 7.21 (d, J = 8.2 Hz, 1H) , 7.11 (dd, J = 8.2, 1.4 Hz, 1H) , 4.07 (t, J = 7.5 Hz, 2H) , 3.84 (dd, J = 9.3,
5.6 Hz, 2H) , 3.69 (t, J = 6.1 Hz, 2H) , 2.93 (dt, J = 19.8, 7.3 Hz, 4H) , 2.13 -2.03 (m, 2H) , 1.84 -1.70 (m, 4H) , 1.40 -1.21 (m, 26H) , 0.93 -0.85 (m, 12H) , 0.03 (s, 6H) .
(5) Preparation of compound 19
To a solution of compound 18 (1.9 g, 3.32 mmol, 1.0 eq) in DCM (10 mL) , was added Dess-Martin periodinane (DMP) (1.69 g, 3.98 mmol, 1.2 eq) under nitrogen atmosphere. The reaction mixture was stirred for 6 h. After reaction, NaHCO3 solution (20 mL) was added into the mixture and the mixture was stirred for 30 min. The mixture was extracted three times by DCM and then the organic phase was washed one time by brine. After dried by anhydrous Na2SO4 and concentrated under reduced pressure, the resultant residue was purified with flash chromatography (silica gel, gradient eluent: 1-5%of MeOH/DCM) to provide compound 19 (619 mg, 33%yield) as brown oil. The product was characterized with mass spectrometry and 1H NMR. MW calc.: 570.46; MW Found: 571.61 [M + H] +. 1H NMR (400 MHz, CDCl3) δ 7.98 (s, 1H) , 7.35 (d, J = 8.2 Hz, 2H) , 7.30 (s, 1H) , 4.25 (t, J = 7.5 Hz, 2H) , 3.89 (t, J = 6.9 Hz, 2H) , 3.18 -3.12 (m, 2H) , 3.01 (t, J = 6.8 Hz, 2H) , 2.55 -2.50 (m, 2H) , 2.32 -2.24 (m, 2H) , 1.85 (dd, J = 14.3, 7.2 Hz, 2H) , 1.36 -1.28 (m, 26H) , 0.93 -0.86 (m, 12H) , 0.03 (s, 6H) .
(6) Preparation of compound 20
To a solution of compound 8 (733 mg, 1.16 mmol, 1.2 eq) in anhydrous THF (5 mL) under nitrogen atmosphere at ice bath, was added NaH (60%dispersion in mineral oil, 77 mg, 1.93 mmol, 2.0 eq) . The reaction mixture was stirred at ice bath for 15 min, and then compound 19 (550 mg, 0.964 mmol, 1.0 eq) was added to the reaction mixture. The mixture was stirred at room temperature for 2 h and then the reaction was moved to ice bath, saturated ammonium chloride (5 mL) was added slowly into the mixture. After 5 minutes, the reaction was extracted two times with ethyl acetate, the organic phase was washed one time by brine. Then dried by anhydrous Na2SO4 and concentrated under reduced pressure. The resultant
residue was purified with flash chromatography (silica gel, gradient eluent: 1-5%of MeOH/DCM) to provide compound 20 (699 mg, 81%yield) . The product was characterized with mass spectrometry and 1H NMR. MW calc.: 876.58; MW. Found: 877.96 [M + H] +. 1H NMR (400 MHz, CDCl3) δ 7.54 (s, 1H) , 7.22 (d, J = 8.2 Hz, 1H) , 7.15 -7.10 (m, 1H) , 6.97 -6.84 (m, 1H) , 5.73 -5.65 (m, 5H) , 4.07 (t, J = 7.5 Hz, 2H) , 3.85 (t, J = 7.4 Hz, 2H) , 2.96 (t, J = 7.4 Hz, 2H) , 2.88 (t, J = 7.5 Hz, 2H) , 2.18 -2.06 (m, 2H) , 1.85 -1.70 (m, 3H) , 1.33 -1.18 (m, 48H) , 0.90 (s, 9H) , 0.03 (s, 6H) .
(7) Preparation of compound 21
To a solution of compound 20 (690 mg, 0.77 mmol, 1.0 eq) in THF (2 mL) was added 2 M HCl (2 mL) . The reaction mixture was stirred at room temperature for 4 h. 5 mL H2O was added into the reaction. Then the mixture was extracted two times with ethyl acetate, washed with brine and dried with anhydrous Na2SO4. The organic layer was concentrated under reduced pressure and the resultant residue was purified with flash chromatography (silica gel, gradient eluent: 1-5%of MeOH/DCM) to provide compound 21 (488 mg, 82%yield) . The product was characterized with mass spectrometry and 1H NMR. MW calc.: 762.49; MW Found: 763.76 [M+H] +. 1H NMR (400 MHz, CDCl3) δ 7.72 (s, 1H) , 7.33 (d, J = 8.3 Hz, 1H) , 7.24 (d, J = 8.0 Hz, 1H) , 6.87 -6.74 (m, 1H) , 5.75 -5.59 (m, 5H) , 4.14 (t, J = 7.4 Hz, 2H) , 3.91 (t, J = 6.4 Hz, 2H) , 3.09 -2.95 (m, 4H) , 2.46 (d, J = 6.8 Hz, 2H) , 2.17 (dt, J = 14.4, 7.3 Hz, 2H) , 1.87 -1.75 (m, 3H) , 1.31 -1.17 (m, 44H) , 0.88 (t, J = 6.8 Hz, 3H) .
(8) Preparation of compound D6
To a solution of compound 21 (480 mg, 0.62 mmol, 1.0 eq) and Diisoropyl ammonium tetrazolide (212 mg, 1.24 mmol, 2.0 eq) in anhydrous DCM (5 mL) under nitrogen atmosphere
was added 3- ( (Bis (diisopropylamino) phosphino) oxy) propanenitrile (374 mg, 1.24 mmol, 2.0 eq) at room temperature. The reaction mixture was stirred for 4 h. The mixture was extracted two times with DCM, then washed with brine and dried with anhydrous Na2SO4. The organic layer was concentrated under reduced pressure and the resultant residue was purified with flash chromatography (silica gel, gradient eluent: 1-3%of MeOH/DCM, 1%Et3N) to provide compound D6 (514 mg, 84%yield) as yellow oil. The product was characterized with mass spectrometry and 1H NMR. MW calc.: 962.60; MW Found: 880.82 [M -83+ H] +. 1H NMR (400 MHz, CDCl3) δ 7.53 (s, 1H) , 7.22 (d, J = 8.2 Hz, 1H) , 7.11 (d, J = 8.2, 1H) , 6.95 -6.82 (m, 1H) , 5.83 -5.59 (m, 5H) , 4.05 (t, J = 7.5 Hz, 2H) , 3.83 -3.73 (m, 2H) , 3.51 -3.46 (m, 2H) , 3.04 (t, J = 7.3 Hz, 2H) , 2.86 (t, J = 7.5 Hz, 2H) , 2.76 (td, J = 6.2, 1.9 Hz, 5H) , 2.10 (dd, J = 15.1, 7.6 Hz, 2H) , 1.81 -1.72 (m, 3H) , 1.26 -1.24 (m, 26H) , 1.21 (s, 18H) , 1.18 -1.34 (m, 12H) , 0.88 (t, J = 6.8 Hz, 3H) .
Example 26. The preparation of compound D7 of the present disclosure
Compound D7 was prepared in this Example by using the following procedures.
(1) The preparation of compound 24
To a solution of compound 1 (2.24 g, 10.50 mmol, 0.9 eq) and K2CO3 (4.84 g, 35.01 mmol, 3.0 eq) in anhydrous DMF (40 mL) , under nitrogen atmosphere, was added compound 27 (3.8 g, 11.67 mmol, 1.0 eq) . The reaction mixture was stirred at 60 ℃ overnight, then cold water (120 mL) was added. The mixture was extracted three times with ethyl acetate, then the
organic phase was washed three times with saturated LiCl solution and one time with brine. Then the organic phase was dried with anhydrous Na2SO4 and concentrated under reduced pressure to form compound 28 as yellow oil which was directly used in the next step without further purification.
(2) The preparation of compound 29
To a solution of compound 28 (6.15 g, 11.96 mmol) in MeOH (60 mL) under was added 10%Pd/C (615 mg) . The reaction mixture was stirred room temperature under hydrogen atmosphere for 16 h, and then filtered. The filtrate was concentrated under reduced pressure. The resultant residue 29 was directly used in next step without further purification. The resultant residue 29 was characterized with mass spectrometry. MW calc.: 488.43; MW. Found: 489.54 [M + H] +.
(3) The preparation of compound 31
To a solution of compound 29 (5.79 g, 11.86 mmol, 1.0 eq) in EtOH (59 mL) under nitrogen atmosphere, was added 30 (2.23 g, 11.86 mmol, 1.0 eq) and AcOH (2.73 mL, 47.44 mmol, 4.0 eq) . The reaction mixture was stirred at 80 ℃ for 2 h, and then concentrated under reduced pressure. Then a saturated NaHCO3 solution (100 mL) was added, the mixture was extracted three times with ethyl acetate, then the organic phase was combined and washed with brine, dried with anhydrous Na2SO4, and concentrated. The resultant residue was purified with flash chromatography (silica gel, gradient eluent: 10-50%of ethyl acetate/petroleum ether) to provide compound 31 (1.8 g, 26%yield) . The product was characterized with mass spectrometry. MW calc.: 656.53; MW Found: 657.70 [M + H] +.
(4) The preparation of compound 32
To a solution of compound 31 (1.80 g, 2.74 mmol, 1.0 eq) in THF (14 mL) under nitrogen atmosphere, was added 1 M TBAF THF solution (4.11 mL, 4.11 mmol, 1.5 eq) . The reaction mixture was stirred at room temperature for 2 h, then H2O (20 mL) was added. The mixture was extracted three times by ethyl acetate, then washed one time by brine, dried by anhydrous Na2SO4 and concentrated under reduced pressure.
The resultant residue and triethylamine (TEA, 0.57 mL, 4.11 mmol, 1.5 eq) in DCM (14 mL) under nitrogen atmosphere, was added DMTrCl (1.11 g, 3.29 mmol, 1.2 eq) . The reaction mixture was stirred for 6 h at room temperature. The mixture was extracted two times with DCM, then washed with NaHCO3, brine and dried with anhydrous Na2SO4. The organic layer was concentrated under reduced pressure and the resultant residue was purified with flash chromatography (silica gel, gradient eluent: 10-50%of ethyl acetate/petroleum ether) to provide compound 32 (1.44 g, 62%yield) . The product was characterized with 1H NMR. 1H NMR (400 MHz, CDCl3) δ 7.56 (s, 1H) , 7.37 -7.32 (m, 2H) , 7.27 -7.22 (m, 6H) , 7.21 -7.13 (m, 3H) , 6.78 -6.72 (m, 4H) , 4.14 -4.06 (m, 2H) , 3.76 (s, 6H) , 3.73 (s, 2H) , 3.67 (s, 3H) , 3.59 (t, J = 6.9 Hz, 2H) , 3.11 (t, J = 7.0 Hz, 2H) , 1.77 -1.68 (m, 2H) , 1.30 -1.20 (m, 38H) , 0.87 (t, 3H) .
(5) The preparation of compound 43
To a solution of compound 32 (1.34 g, 1.59 mmol, 1.0 eq) in anhydrous THF (8 mL) under nitrogen atmosphere and ice bath, was added LiAlH4 (90 mg, 2.38 mmol, 1.5 eq) . The mixture was moved to room temperature after 10 minutes and stirred for 1 h. Then the reaction was moved to ice bath, saturated potassium sodium tartrate solution (10 mL) was added slowly
into the mixture. After 30 minutes, the reaction was extracted three times with ethyl acetate, then the organic phase was combined and washed with brine, dried anhydrous Na2SO4, and concentrated.
The crude product and Diisoropyl ammonium tetrazolide (817 mg, 4.77 mmol, 3.0 eq) were dissolved in anhydrous DCM (8 mL) under nitrogen atmosphere was added 3- ( (Bis (diisopropylamino) phosphino) oxy) propanenitrile (1.44 g, 4.77 mmol, 3.0 eq) at room temperature. The reaction mixture was stirred for 6 h. The mixture was extracted two times with DCM, then washed with brine and dried with anhydrous Na2SO4. The organic layer was concentrated under reduced pressure and the resultant residue was purified with flash chromatography (silica gel, gradient eluent: 10-50%of ethyl acetate/petroleum ether, 1%Et3N) to provide compound D7 (1.45 g, 90%yield) as light-yellow oil. The product was characterized with 1H NMR. 1H NMR (400 MHz, CDCl3) δ 7.50 (s, 1H) , 7.37 -7.31 (m, 2H) , 7.29 -7.21 (m, 6H) , 7.18 (d, J = 11.0 Hz, 2H) , 7.12 -7.07 (m, 1H) , 6.78 -6.71 (m, 4H) , 4.16 -4.05 (m, 4H) , 3.76 (s, 6H) , 3.62 -3.56 (m, 4H) , 3.11 (t, J = 7.0 Hz, 2H) , 3.03 (t, J = 7.4 Hz, 2H) , 2.61 (t, J = 6.4 Hz, 2H) , 2.57 -2.49 (m, 2H) , 1.74 -1.68 (m, 2H) , 1.27 -1.23 (m, 38H) , 1.18 -1.16 (m, 12H) , 0.88 (t, J = 6.7 Hz, 3H) .
Example 27. Preparation of compound F69
Compound F69 was prepared in this example by using the following procedures.
(1) The preparation of compound 45
To a solution of methyl 2- (4-fluoro-3-nitrophenyl) acetate 4 (5 g, 23.5 mmol, 1.0 eq) and K2CO3 (4.9 g, 35.3 mmol, 1.5 eq) in anhydrous DMF (80 mL) , under nitrogen atmosphere, was added compound 51 (6.9 g, 25.8 mmol, 1.1 eq) . The reaction mixture was stirred at 55 ℃ for 6 h, then cold water (200 mL) was added. The mixture was extracted three times with ethyl
acetate, then the organic phase was washed three times with saturated LiCl solution and one time with brine. Then the organic phase was dried over anhydrous Na2SO4 and concentrated under reduced pressure to form compound 45 as yellow oil which was directly used in the next step without further purification.
(2) The preparation of compound 46
To a solution of compound 45 (10.8 g, 23.5 mmol, 1.0 eq) in THF/H2O (9: 1, 130 mL) , under ice bath, was added HCOONH4 (8.9 g, 141 mmol, 6.0 eq) and Zn powder (9.2 g, 141 mmol, 6.0 eq) . After stirred for 10 minutes, the reaction mixture was moved from the ice bath and stirred at room temperature overnight. Then the reaction mixture was filtered and concentrated under reduced pressure. After that, water (200 mL) was added into the mixture, then the mixture was extracted three times with ethyl acetate, the organic phase was washed one time with brine. After dried over anhydrous Na2SO4 and concentrated under reduced pressure, the product 46 formed then was directly used in the next step without further purification.
(3) The preparation of compound 47
To a solution of compound 46 (5 g, 11.6 mmol, 1.0 eq) in EtOH (60 mL) under nitrogen atmosphere, was added 3- ( (tert-butyldimethylsilyl) oxy) propanal 7 (3.3 g, 17.4 mmol, 1.5 eq) and AcOH (2.7 mL, 46.4 mmol, 4.0 eq) . The reaction mixture was stirred at 80℃ overnight, and then concentrated under reduced pressure. Then a saturated NaHCO3 solution (100 mL) was added, the mixture was extracted three times with ethyl acetate, then the organic phase was combined and washed with brine, dried over Na2SO4, and concentrated. The resultant residue compound was purified with flash chromatography (silica gel, gradient eluent: 10-40%of ethyl acetate/petroleum ether) to provide compound 47 (1.5 g, 22%yield) as light-yellow oil. The product was characterized with mass spectrometry and 1H NMR. MW calc.: 598.99; MW. Found: 599.72 [M + H] +. 1H NMR (400 MHz, CDCl3) δ 7.59 (s, 1H) , 7.24 (d, J = 8.3 Hz, 1H) , 7.16 (d, J = 8.3 Hz, 1H) , 5.40 -5.31 (m, 2H) , 4.17 -4.09 (m, 4H) , 3.74 (s, 2H) , 3.68 (s, 3H) , 3.09 (t, J = 6.9 Hz, 2H) , 2.06 -1.92 (m, 4H) , 1.81 -1.72 (m, 2H) , 1.39 -1.19 (m,
22H) , 0.88 (t, J = 6.5 Hz, 3H) , 0.86 (s, 9H) , 0.01 (s, 6H) .
(4) The preparation of compound 48
To a solution of compound 47 (1.5 g, 2.5 mmol, 1.0 eq) in THF (10 mL) under nitrogen atmosphere, was added 2 M HCl solution (10 mL) . The reaction mixture was stirred at room temperature for 2 h. 20 mL saturated NaHCO3 solution was added into the reaction. Then the mixture was extracted two times with ethyl acetate, washed with brine and dried with anhydrous Na2SO4. The organic layer was concentrated under reduced pressure. The resultant residue was dissolved in DCM (10 mL) , triethylamine (0.4 g, 3.8 mmol, 1.5 eq) and DMTrCl (1.1 g, 3.3 mmol, 1.3 eq) was added therein. The reaction mixture was stirred at room temperature for 6 h, after which it was concentrated under reduced pressure. The resultant residue was purified with flash chromatography (silica gel, gradient eluent: 20-50%of ethyl acetate/petroleum ether) to provide compound 48 (1.5 g, 76%yield) as light-yellow oil. The product was characterized with mass spectrometry and 1H NMR. MW calc.: 787.1; MW. Found: 788.1 [M + H] +. 1H NMR (400 MHz, CDCl3) δ 7.56 (s, 1H) , 7.34 (d, J = 7.1 Hz, 2H) , 7.25 -7.13 (m, 9H) , 6.75 (d, J = 8.8 Hz, 4H) , 5.41 -5.30 (m, 2H) , 4.13 -4.07 (m, 2H) , 3.76 (s, 6H) , 3.73 (s, 2H) , 3.67 (s, 3H) , 3.60 (t, J = 7.0 Hz, 2H) , 3.11 (t, J = 6.9 Hz, 2H) , 1.99 (t, J = 15.9 Hz, 4H) , 1.77 -1.65 (m, 2H) , 1.33 -1.23 (m, 22H) , 0.87 (t, J = 6.8 Hz, 3H) .
(5) The preparation of compound F69
To a solution of compound 48 (1.5 g, 1.9 mmol, 1.0 eq) in anhydrous THF (20 mL) under nitrogen atmosphere and ice bath, was added LiAlH4 (73 mg, 1.9 mmol, 1.0 eq) . The mixture was moved to room temperature after 10 minutes and stirred for 1 h. Then the reaction was moved to ice bath, saturated potassium sodium tartrate solution (10 mL) was added slowly into the mixture. After 30 minutes, the reaction was extracted three times with ethyl acetate, then the organic phase was combined and washed with brine, dried over Na2SO4, and concentrated. The crude product (1.3 g, 1.7 mmol, 1.0 eq) and Diisoropyl ammonium
tetrazolide (587 mg, 3.4 mmol, 2 eq) were dissolved in anhydrous DCM (10 mL) under nitrogen atmosphere was added 3- ( (Bis (diisopropylamino) phosphino) oxy) propanenitrile (1.0 g, 3.4 mmol, 2 eq) at room temperature. The reaction mixture was stirred for 6 h. The mixture was extracted two times with DCM, then washed with brine and dried with anhydrous Na2SO4. The organic layer was concentrated under reduced pressure and the resultant residue was purified with flash chromatography (silica gel, gradient eluent: 10-30%of ethyl acetate/petroleum ether, 1%Et3N) to provide compound F69 (1.2 g, 78%yield) as light-yellow oil. The product was characterized with mass spectrometry and 1H NMR. MW calc.: 959.31; MW Found: 876.75 [M-diisopropyl + H] +. 1H NMR (400 MHz, CDCl3) δ 7.50 (s, 1H) , 7.34 (d, J = 7.2 Hz, 2H) , 7.25 -7.14 (m, 8H) , 7.09 (d, J = 8.2 Hz, 1H) , 6.75 (d, J = 8.8 Hz, 4H) , 5.41 -5.31 (m, 2H) , 4.13 -4.05 (m, 2H) , 3.93 -3.83 (m, 1H) , 3.81 -3.70 (m, 9H) , 3.64 -3.54 (m, 4H) , 3.11 (t, J = 7.0 Hz, 2H) , 3.03 (t, J = 7.3 Hz, 2H) , 2.57 -2.47 (m, 2H) , 2.07 -1.92 (m, 4H) , 1.77 -1.66 (m, 2H) , 1.34 -1.24 (m, 22H) , 1.18 -1.12 (m, 12H) , 0.87 (t, J = 6.7 Hz, 3H) .
Example 28. Design and synthesis of oligonucleotides
Oligonucleotide sequences used for cell or animal treatments in the following examples are listed in Table 1.
Example 29. Characterization of the in vitro knockdown activity of DEC-conjugated siRNAs for mouse FVII gene.
In vitro knockdown activity of DEC-conjugated siRNAs (i.e., RD-12339, RD-12585, RD-12586, RD-12710, RD-12711 and RD-12712) for mouse FVII mRNA was characterized in this Example.
PMH cells were transfected with each of the indicated DCOs (i.e., RD-12339, RD-12585, RD-12586, RD-12710, RD-12711 and RD-12712) at 0.1 nM and 1 nM with LipofectamineTM RNAiMAX (Thermofisher) for 24 hours according to the manufacturer’s instructions was shown in FIG. 1. DCOs (i.e., RD-12339, RD-12585, RD-12586, RD-12710, RD-12711 and RD-12712) were also directly added into culture medium containing PMH cells at 0.01, 0.05, 0.20, 0.78, 3.13, 12.50, 50 and 200 nM was shown in FIG. 2. Mock treatment was transfection in the absence of oligonucleotide. dsCon2 served as a non-specific duplex control. Mouse FVII mRNA levels were quantified by RT-qPCR using a gene specific primer set. Tbp was amplified as an internal reference for RNA loading. The mean expression values of FVII mRNA relative to Mock treatment are normalized to Tbp.
As shown in FIG. 1, transfection of RD-12339, RD-12585, RD-12586, RD-12710, RD-12711 and RD-12712 reduced FVII mRNA expression by 94%, 85%, 91%, 80%, 78%and 84%at 0.1 nM and 98%, 96%, 97%, 89%, 91%and 89%at 1 nM treatment, respectively. All tested DCOs achieved comparable knockdown activities in a dose-dependent manner with RD-12339, RD-12585 and RD-12586 having slightly greater maximal activities.
For free uptake in absence of lipofectamine, RD-12339, RD-12585, RD-12586, RD-12710, RD-12711 and RD-12712 were added to the culture media of PMH cells at escalating concentrations (i.e., 0.01, 0.05, 0.20, 0.78, 3.13, 12.50, 50 and 200 nM) for 24 hours. The treated cells were harvested to isolated total cellular RNA which was used to detect FVII mRNA expression by RT-qPCR after cDNA conversion by reverse transcription (RT) reaction. As shown in FIG. 2, all the DCOs caused a dose-dependent knockdown on FVII mRNA level with a calculated IC50 ranging from 0.2308 to 0.6005 nM and RD-12585 having the lowest IC50 at 0.2308 nM were summarized in Table 2.
Table 2. DCO IC50 values for FVII mRNA knockdown in PMH cells
Overall, these results indicate that the DCOs have in vitro target gene knockdown activity with and without lipofectamineTM RNAiMax in PMH cells.
Example 30. Characterization of in vivo knockdown activity of DEC-conjugated siRNAs for mouse FVII gene expression.
To characterize the in vivo delivery efficiency (knockdown activity) of DEC-conjugated siRNAs in liver tissue, the indicated DCOs (i.e., RD-12339, RD-12585, RD-12586, RD-12710, RD-12711 and RD-12712) were administered via SC injection into adult C57BL/6J mouse on PND 40 at 0.2, 1 and 5 mg/kg. All test DCOs were dissolved in saline. Saline alone was injected as a vehicle control to establish baseline expression in liver tissues. Mice were sacrificed on 3 days post dosing and FVII mRNA level was quantified in liver tissue after RNA isolation and RT reaction via RT-qPCR using gene specific primer sets.
As shown in FIG. 3, treatments of RD-12339, RD-12585, RD-12586, RD-12710, RD-12711 and RD-12712 at low dose (0.2 mg/kg) reduced FVII mRNA levels by 25%, 0%, 2%, 22%, 0%and 0%, among which only RD-12339 and RD-12710 showed knockdown activity. At medium dose (1 mg/kg) , all siRNA conjugates exhibited substantial knockdown of FVII mRNA. RD-12339, RD-12585, RD-12586, RD-12710, RD-12711 and RD-12712 caused a 67%, 47%, 64%, 60%, 43%and 62%knockdown of FVII mRNA, respectively. RD-12339, RD-12586, RD-12710 and RD-12712 exhibited over 60%knockdown activity with RD-12339 having the greatest knockdown activity. At high dose (5 mg/kg) , RD-12339, RD-12585, RD-12586, RD-12710, RD-12711 and RD-12712 caused an 81%, 82%, 89%, 80%, 81%and 89%knockdown of FVII mRNA.
Further, FVII protein expression levels in the plasma of the treated mice were detected by ELISA assay. As shown in FIG. 4, 3 DCOs (i.e., RD-12339, RD-12585 and RD-12586) at low dose (0.2 mg/kg) reduced FVII protein expression by 44%, 3%and 21%, respectively. RD-12339, RD-12585 and RD-12586 at medium dose (1 mg/kg) reduced FVII protein
expression by 67%, 51%and 65%, respectively. At the high dose (5 mg/kg) , knockdown activity was similar for all tested DCOs with RD-12339, RD-12585 and RD-12586 providing 89%, 87%and 93%reductions in FVII protein levels, respectively.
Overall, the data indicates that DCOs can be successfully delivered to the liver and cause significant knockdown of target gene FVII mRNA and protein expression in a dose-dependent manner.
Example 31. Potent and durable knockdown of DEC-conjugated siRNAs on mouse FVII protein in plasma of C57BL/6J mice
The indicated DCOs (i.e., RD-12710 and RD-12712) were administered via SC injection into C57BL/6J mice on PND 40 at 3 mg/kg for 89 days. RD-11706 was injected at 3 mg/kg and served as a control. Saline was injected as a vehicle control to establish the baseline of mFVII protein expression. Mouse plasmas were collected on 10, 31, 54, 61, 80 and 89 days post dosing and mFVII protein level was quantified in mouse plasma by ELISA assay. The results of potent and durable knockdown of DEC-siRNAs on mouse FVII protein in plasma are shown in FIG. 5.
Example 32. In vitro knockdown activity of DEC-siRNA in PMH cells via free uptake.
To test in vitro knockdown activity of exemplary DEC-siRNAs (i.e., RD-13110, RD-13115, and RD-13118) , PMH cells were treated at escalating concentrations (i.e., 1.56, 6.25, 25, 100, 400 and 1600 nM) in absence of any additional delivery system (i.e., free uptake) for 72 hours. Sod1 levels were assessed via RT-qPCR to generate dose response curves and estimate potency. As shown in FIG. 6A, all DEC-siRNAs demonstrated dose dependent knockdown in which potency (dotted line) is depicted at concentrations associated with 50%Sod1 levels relative to basal expression. In comparison, treatment with non-conjugate control (i.e., RD-12556) did not demonstrate knockdown until the highest tested dose in which activity did not exceed 50%reductions in Sod1 levels (FIG. 6B) . Table 3 summarizes both potency and knockdown activity at the highest tested dose for each test article. In general, all DEC-siRNAs (i.e., RD-13110, RD-13115, and RD-13118) regardless of conjugate had better potencies than the non-conjugate control RD-12556. C5x5 conjugates (i.e., RD-13115 and RD-13118) provided knockdown at potencies roughly 11-fold lower than the C5x1 variant RD-13110. Inclusion of ACO in RD-13118 also provided an approximate 2.5-fold improvement in potency indicating combining the DEC technology with ODV-siRNA
(PCT/CN2022/104037) may have delivery benefits when used in combination.
Table 3. The knockdown activity of DEC-siRNA in PMH cells via free uptake
*ND; not detected (knockdown did not exceed 50%Sod1)
*ND; not detected (knockdown did not exceed 50%Sod1)
Example 33. In vivo knockdown of DEC-siRNA on Sod1 mRNA level in CNS tissues
To test the in vivo knockdown activity of DEC-siRNAs (i.e., RD-13110, RD-13115 and RD-13118) in comparison to non-conjugate control (i.e., RD-12556) , adult C57BL/6J mice were administered at a 200 μg total dose via unilateral ICV injection. All test articles were formulated in saline in which treatment with saline alone functioned as a procedural control to establish baseline expression in CNS tissues. Mice were sacrificed on 7 days post dosing and CNS tissues from the brain (i.e., frontal cortex, cerebellum, and cerebrum) , spinal cord (i.e., cervical, thoracic, and lumbar) , and periphery (i.e., liver) were harvested for mRNA expression analysis via RT-qPCR. As shown in FIG. 7, RD-12556 and RD-13110 had similar activities in the indicated CNS tissues ranging between 43-73%and 40-63%knockdown, respectively. Whereas RD-13115 and RD-13118 had improved knockdown across all tissues at the 200 μg dose indicating the C5x5 conjugates had better in vivo potency via ICV injection compared to the non-conjugated control (i.e., RD-12556) and C5x1 variant RD-13110. Analysis in periphery tissue (i.e., liver) also revealed that activity of the C5x5 conjugates (i.e., RD-13115 and RD-13118) were not well retained within the CNS in which systemic exposure via CNS drainage provided knockdown in the liver at levels similar to the CNS tissues. In comparison, both RD-12556 and RD-13110 activity was selectively enriched across the CNS providing only an approximate 11%and 3%knockdown in the liver, respectively. Overall, the C5x5 conjugates have improved activity and biodistribution across all CNS tissues. For indication in which confinement to the CNS may be important to limit systemic toxicities, C5x1 would be more ideal compared to other lipids (e.g., C5x5) .
Example 34. In vivo knockdown of DEC-siRNA on Sod1 mRNA level following systemic administration
To test in vivo knockdown following systemic exposure, adult C57BL/6J mice were administered with DEC-siRNAs (i.e., RD-13110, RD-13115 and RD-13118) or non-conjugate control (i.e., RD-12556) at a 20 mg/kg dose via IV injection. Mice were sacrificed on day 7 after treatment and Sod1 knockdown was quantified via RT-qPCR in select organs (i.e., heart, liver, spleen, lung, kidney, and bladder) . As shown in FIG. 8, RD-12556 only provided substantial knockdown in the kidney reducing Sod1 levels by 56%, whereas RD-13110 also had activity selectively enriched in the spleen. RD-13115 and RD-13118 both had broad activity across all tissues except bladder in which only RD-13118 was capable of reducing Sod1 levels by roughly 21%. Overall, these results indicate that C5x1 conjugation may have unique delivery functions via systemic administration offering selective targeting to the spleen, whereas C5x5 can provide knockdown to a broader spectrum of tissues. Notably, inclusion of ACO in RD-13118 was the only DEC-siRNA to provide measurable knockdown in the bladder indicating combining the DEC technology with ODV-siRNA may also have further delivery benefits when used in combination in vivo.
Example 35. In vivo knockdown of DEC-siSOD1 on Sod1 mRNA level in skeletal muscle
To test in vivo knockdown activity in skeletal muscle tissues, adult C57BL/6J mice were treated with DEC-siRNAs (i.e., RD-13110, RD-13115 and RD-13118) or non-conjugate control (i.e., RD-12556) at a 20 mg/kg dose via IV injection. Mice were sacrificed on 7 days post dosing and Sod1 mRNA level was quantified via RT-qPCR in muscle tissues (i.e., bicep, semitendinosus, platysma and gluteus) . As shown in FIG. 9, RD-12556 provided no significant knockdown in any muscle tissue compared to the saline treatment group. RD-13110 had modest activity in select tissues yet did not exceed 25%reductions in Sod1 mRNA levels at the 20 mg/kg dose. In contrast, RD-13115 reduced Sod1 mRNA levels by 82%, 84%, 78%, and 76%in bicep, semitendinosus, platysma and gluteus tissues, respectively. Similarly, RD-13118 reduced Sod1 mRNA levels by 76%, 84%, 61%, and 67%in bicep, semitendinosus, platysma and gluteus tissues, respectively. In general, DEC conjugation provides knockdown in muscle tissue via IV injection; however, C5x5 conjugates (i.e., RD-13115 and RD-13118)
enabled siRNA knockdown at a 20 mg/kg dose that has been otherwise unobtainable by conventional delivery technologies (i.e., LNP and GalNAc) .
Example 36. In vivo knockdown activity of DEC-siSOD1 on Sod1 mRNA level in retinal tissue
To test in vivo knockdown activity of DEC-siRNAs in the eye, adult SD rats were treated with DEC-siRNAs (i.e., RD-13115 and RD-13118) or non-conjugate control (i.e., RD-12556) at a 30 μg dose via local IVT injection into the eye. Rats were sacrificed on 14 days post dosing and Sod1 mRNA level was quantified via RT-qPCR in retinal tissue. As shown in FIG. 10, RD-12556 provided only a 39%knockdown, whereas RD-13115 and RD-13118 reduced Sod1 mRNA levels by 74%and 71%, respectively. DEC-siRNA (i.e., RD-13115 and RD-13118) provided nearly twice the knockdown activity at a 30 μg dose compared to non-conjugated control (i.e., RD-12556) indicating the DEC technology has benefit for delivering oligonucleotides to the eye to manipulate gene expression.
Example 37. Pharmacokinetics of DEC-saRNA in bladder tissue from adult C57BL/6J mice following DEC-saRNA treatment via IVB instillation
Adult C57BL/6J mice were treated with 3 mg RD-13520 (DEC-saRNA) and RD-10773 (non-DEC-saRNA) via IVB instillation and bladder tissues were harvested on 2-, 6-, 12-hour, day-1 and day-4 after treatment. RD-10773 and RD-13520 were detected in the bladder tissue preps by stem-loop RT-qPCR. The concentrations of RD-13520 and RD-10773 in bladder were plotted in FIG. 11 to evaluate pharmacokinetics (PK) and summarized in Table 4.
Table 4. Mean saRNA concentration in bladder tissue of C57BL/6J mice
Example 38. Potent and durable knockdown of mouse Sod1 mRNA by DEC-conjugated siRNAs in C57BL/6J mice
To further assess knockdown durability of DEC-siRNA in comparison to C16-siRNA, the indicated siRNAs (i.e., RD-15135, RD-15136, RD-15137 and RD-15138) were administered into C57BL/6J mice at 10 mg/kg. RD-15135 served as a non-conjugate control. RD-15136 was a lipid (i.e., C16) conjugated siRNA. RD-15137 with a typical DEC structure (i.e., C5X5) served as a typical DEC-siRNA. RD-15138 with a typical DEC structure (i.e., C5X5) and a lipid (i.e., C16) served as a combo siRNA (i.e., DEC-C16-siRNA) . Treatment with saline alone was used as a vehicle control to establish baseline expression. Mouse Sod1 mRNA levels on 14 and 28 days post dosing were quantified via RT-qPCR using a gene specific primer set, respectively. FIG. 12A shows the body weight change of C57BL/6J mice out to day 28 post treatment. Sod1 mRNA expressions on 14 and 28 days post dosing in tissues from periphery (i.e., heart, liver, kidney, fat tissues, pancreas, diaphragm) , blood vessel (i.e., thoracic aorta with vein) and skeletal muscle (i.e., bicep, semitendinosus, platysma and gluteus) were shown in FIG. 12B-12C.
Example 39. Potent and durable knockdown of rat Sod1 mRNA in SD rats by DEC-conjugated siRNAs
To further assess knockdown durability of DEC-siRNA in comparison to C16-siRNA, the indicated siRNAs (i.e., RD-15135, RD-15136, RD-15137 and RD-15138) were administered into adult SD female rats at 0.9 mg. RD-15135 served as a non-conjugate control. RD-15136 was a lipid (i.e., C16) conjugated siRNA. RD-15137 with a typical DEC structure (i.e., C5X5) served as a typical DEC-siRNA. RD-15138 with a typical DEC structure (i.e., C5X5) and a lipid (i.e., C16) served as a combo siRNA (i.e., DEC-C16-siRNA) . Treatment with aCSF alone was used as a vehicle control to establish baseline expression. Rat Sod1 mRNA levels on 14 and 28 days post dosing were quantified via RT-qPCR using a gene specific primer set, respectively. FIG. 13A shows the body weight change of SD rats out to day 28 post treatment. Sod1 mRNA expressions on 14 days post dosing in tissues from brain (i.e., frontal cortex, cerebellum and cerebrum) , spinal cord (i.e., cervical, thoracic and lumbar) and periphery (i.e., liver and kidney) were shown in FIG. 13B. Sod1 mRNA on 28 days post dosing in tissues from brain (i.e., frontal cortex, cerebellum and brainstem) , spinal cord (i.e., cervical, thoracic and lumbar) and periphery (i.e., liver and kidney) were shown in FIG. 13C.
Example 40. Delivery enhancing compound (DEC) of siRNA-ACO provides
consistent and potent knockdown on SOD1 mRNA in SK-N-AS and T98G cells.
To assess in vitro knockdown activity of DEC-siRNA-ACOs, the indicated DEC-siRNA-ACOs (i.e., RD-16149, RD-16099, RD-16100, RD-16101, RD-16150, RD-16103, RD-16104 and RD-16105) were transfected into SK-N-AS and T98G cell at the indicated concentrations (i.e., 1.56, 6.25, 25, 100, 400 and 1600 nM) to generate dose response curves. RD-16106 was transfected and served as a siRNA-ACO control. Mock treatments were transfected in absence of oligonucleotide (not shown) . SOD1 mRNA levels were quantified via RT-qPCR using a gene specific primer set. The remaining human SOD1 mRNA levels in SK-N-AS and T98G cells were plotted in FIG. 14A-14B. The EC50 values were summarized in Table 5.
Table 5. EC50 values for SOD1 knockdown by DEC-siRNA-ACOs in SK-N-AS and T98G cells.
Example 41. Knockdown activity and tissue concentration of siRNAs via ICV injection in adult hSOD1G93A mice.
To assess the knockdown activity and pharmacokinetics of siRNAs and, the indicated siRNAs (i.e., RD-14851, RD-12500, RD-16145 and RD-16334) were administered into hSOD1G93A mice at 200 μg. RD-14851, RD-12500 and RD-16145 were administered as siRNA-ACOs. RD-16334 with a DEC structure (i.e., L17) was administered and served as a DEC-siRNA-ACO. Treatment with aCSF alone was used as a vehicle control to establish baseline expression. FIG. 15A shows the remaining SOD1 mRNA on 30 days post dosing as quantified in tissues from brain (i.e., frontal cortex, cerebellum and cerebrum) , spinal cord and periphery (i.e., liver) via RT-qPCR using a gene specific primer set. FIG. 15B shows the
concentration on 30 days post dosing as in tissues from brain (i.e., frontal cortex, cerebellum and cerebrum) , spinal cord and periphery (i.e., liver) via stem-loop RT-qPCR using a gene specific primer set. Mean concentration in tissues from frontal cortex, cerebellum, cerebrum, spinal cord and liver to evaluate pharmacokinetics and are summarized in Table 6. Serum chemistry [i.e., glutamic-pyruvic transaminase (ALT) , glutamic oxalacetic transaminase (AST) , total bilirubin (TBIL) and creatinine (CRE) ] and completed blood count [i.e., white blood cell (WBC) , red blood cell (RBC) , blood platelet (PLT) etc. ] were detected and demonstrated safety.
Table 6. Mean concentration of siRNA in hSOD1G93A mouse tissues by ICV injection
Example 42. Splicing activity of “saRNA-ASO” DEC-DAO structure in GM03813 cells.
To assess the splicing activity of “saRNA-ASO” DEC-DAO structure at converting SMN2 pre-mRNA to SMN2FL over the SMN2Δ7 mRNA isoform in GM03813 cells. The indicated DEC-DAO linkage with L21 linker (i.e., RD-16939 and RD-16940) , DEC-saSMN2 (i.e., RD-16424) and DEC-antisense oligonucleotide (DEC-ASO) (i.e., RD-14644) were transfected into GM03813 cells at indicated concentrations (i.e., 0.1, 1 and 10 nM) for 3 days. RD-16381 was transfected as a non-specific DEC control. Combo treatment (i.e., RD-16424+RD-14644) and combo treatment control (i.e., RD-16381+RD-16424, RD-16381+RD-14644) were also transfected into GM03813 cells at indicated concentrations (i.e., 0.1, 1 and 10 nM) for 3 days. As shown in FIGs. 16A-16B, “saRNA-ASO” DEC-DAO structure provides potent splicing activity on mRNA levels of SMN2FL and SMN2Δ7, indicating that combining a saRNA with a ASO into a DAO can largely retain and even increase the activity of both saRNA and ASO units.
Example 43. Effect of “saRNA-siRNA” DEC-DAO structure targeting two different human genes (SMN2 and SOD1) on the expression of SMN2 (SMN2FL and SMN2Δ7) and pOD1 in GM03813 cells.
To assess the effect of “saRNA-siRNA” DEC-DAO structure targeting two different genes, the indicated DEC-DAO linkage with L21 linker (i.e., RD-16941) , DEC-saSMN2 (i.e., RD-16424) and DEC-siSOD1 (i.e., RD-13115) were transfected into GM03813 cells at indicated concentrations (i.e., 0.1, 1 and 10 nM) for 3 days. RD-16381 was transfected as a non-specific DEC control. Combo treatment (i.e., RD-16424+RD-13115) and combo treatment control (i.e., RD-16381+RD-16424, RD-16381+RD-13115) also were transfected into GM03813 cells at indicated concentrations (i.e., 0.1, 1 and 10 nM) for 3 days. FIGs. 17A-17B shows mRNA levels of SMN2FL and SMN2Δ7 and FIG. 17C shows remaining SOD1 mRNA levels. As shown FIGs. 17A-17C, “saRNA-siRNA” DEC-DAO structure provides the potent gene induction on expression of SMN2FL and knockdown on SOD1 mRNA, respectively. This result indicates that combining a saRNA with a siRNA into a DAO can largely retain the activity for each unit and enhance the activity of the saRNA unit.
Example 44. Effect of “siRNA-siRNA” DEC-DAO structure targeting two different genes (mouse Sod1 and Ppig) on the expression of mouse Sod1 and Ppig in C2C12 cells.
To assess the effect of “siRNA-siRNA” DEC-DAO structure targeting two different genes (mouse Sod1 and Ppig) , the indicated DEC-DAO linkage with L21 linker (i.e., RD-16942) , DEC-siSod1 (i.e., RD-13115) and DEC-siPpig (i.e., RD-14672) were transfected into C2C12 cells at indicated concentrations (i.e., 0.01, 0.1 and 1 nM) for 24 hours. RD-16381 was transfected as a non-specific DEC control. Combo treatment (i.e., RD-13115+RD-14672) and combo treatment control (i.e., RD-16381+RD-13115, RD-16381+RD-14672) were also transfected into C2C12 cells at indicated concentrations (i.e., 0.01, 0.1 and 1 nM) for 24 hours. As shown in FIG. 18A-18B, “siRNA-siRNA” DEC-DAO structure provides potent knockdown activity on mouse Sod1 and Ppig mRNA levels, respectively. This result indicates that combining two siRNAs targeting different genes into a DAO can largely retain the activity for each siRNA unit and even enhance the activity of both siRNA units.
Example 45. Effect of “siRNA-siRNA” DEC-DAO structure targeting two different
genes (mouse Sod1 and Ppig) on the expression of mouse Sod1 and Ppig in vivo.
To assess the in vivo effect of “siRNA-siRNA” DEC-DAO structure targeting two different genes (mouse Sod1 and Ppig) , the indicated DEC-DAO linkage with L21 linker (i.e., RD-16942) , DEC-siSod1 (i.e., RD-13115) and DEC-siPpig (i.e., RD-14672) were administered into C57BL/6J mice via IV injection at 10 mg/kg. Combo treatment (i.e., RD-13115+RD-14672) was administered via IV injection at 10 mg/kg. Treatment with saline alone was used as a vehicle control to establish baseline expression. Remaining mouse Sod1 mRNA on 14 days post dosing were quantified in tissues from periphery (i.e., heart, liver, kidney, fat tissues, diaphragm, thoracic aorta with vein) , and skeletal muscle (i.e., bicep, semitendinosus, platysma and gluteus) via RT-qPCR using a gene specific primer set. As shown in FIG. 19, “siRNA-siRNA” DEC-DAO structure (i.e., RD-16942) presented similar knockdown activity on Sod1 mRNA expression. This result indicates that combining two siRNAs targeting different genes into a DAO structure can retain the knockdown activity of the units used as single entity in peripheral and skeletal muscle tissues.
Example 46. Pharmacokinetics of DEC-saRNA in retina and vitreous humor of adult SD rat following DEC-saRNA treatment via IVT injection
To assess the pharmacokinetics of DEC-saRNA, adult SD rats were administered with 0.03 mg DEC-saRNA (i.e., RD-16447) and non-conjugated saRNA (i.e., RD-12173) via IVT injection. The rats were sacrificed on 1-hour, day-1, -3, -7, -14 and -28 following treatment and the concentrations of RD-16447 and RD-12173 were quantified in retina and vitreous humor via stem-loop RT-qPCR. Concentrations of RD-16447 and RD-12173 in retina and vitreous humor were plotted in FIG. 20A-20B. Mean concentrations of oligonucleotide in retina and vitreous humor were summarized in Table 7.
Table 7. Mean oligonucleotide concentration in retina and vitreous humor of SD rats
Example 47. Knockdown activity of DEC-siRNA targeting SOD1 in T98G cells.
A new DEC structure (i.e., C5x34) was designed and conjugated to a siRNA targeting SOD1 gene, resulting in a new DEC-siRNA (i.e., RD-17138) . To assess in vitro knockdown activity of DEC-siRNA, the indicated DEC-siRNA (i.e., RD-17138) were transfected into T98G cell at 0.1 nM and 1 nM for 24 hours. RD-11566 was transfected and served as a non-specific duplex control. Mock treatments were transfected in absence of oligonucleotide. SOD1 mRNA levels were quantified via RT-qPCR. As shown in FIG. 21, RD-17138 showed a knockdown activity on SOD1 mRNA expression greater than 90%at 0.1 nM and 1 nM, suggesting the new DEC structure C5x34 can enhance the knockdown activity of siRNA on SOD1 mRNA level in vitro.
Materials and Methods
General synthesis method of oligonucleotides
The oligonucleotides used in the following examples were synthesized via the following general method.
Single strand synthesis method
The single strand oligonucleotide was synthesized on a K&ADNA synthesizer (K&A Laborgeraete GbR, chaafheim, Germany) by a solid phase synthesis technique.
The starting material was universal solid support or special solid support commercially available or synthesis as disclosure in previous context. In general, phosphoramidite monomers including various linkers and conjugates (0.1M in acetonitrile or dichloromethane) , were added sequentially onto a solid support in the DNA synthesizer to generate the desired full-length oligonucleotides.
Amidite addition: each cycle of amidite addition consisted of four chemical reactions including detritylation, coupling, oxidation/thiolation and capping. In first step, the detritylation was performed by using 3%dichloroacetic acid (TCA) in DCM for 45 seconds. In the second step, Phosphoramidite coupling was conducted for 6 minutes for all amidites by 12 eq.; In the third step, oxidation was performed by using 0.02 M iodine in THF: pyridine: water (70: 20: 10, v/v/v) for 1 minute; if phosphorothioate modification needed then replace oxidation by thiolation which was carried out with 0.1 M solution of xanthane hydride in pyridine: ACN (50: 50, v/v) for 3 minutes; In the fourth step, the capping was
performed by using a THF: acetic anhydride: Pyridine (80: 10: 10, v/v/v) (CAP A) and N-methylimidazole: THF (10: 90, v/v) , (CAP B) for 20 seconds. The Cycles of four chemical reaction will be depended by the length of single of oligonucleotide.
Deprotection I (Nucleobase Deprotection) : After completion of the synthesis, the solid support was transferred to a screw-cap microcentrifuge tube. For a 1 μmol synthesis scale, 1 ml of a mixture of methylamine and ammonium hydroxide was added. The tube containing the solid support was then heated in an oven at 60℃ to 65℃ for 15 min and then allowed to cool to room temperature. The cleavage solution was collected and evaporated to dryness in a speedvac to provide crude single strand of oligonucleotide.
Deprotection II (Removal of 2’-TBDMS Group) : If The crude RNA oligonucleotide, still carrying the 2’-TBDMS groups, then dissolved in 0.1 ml of DMSO. After adding 1 ml of Triethylamine trihydrofluoride, the tube was capped, and the mixture was shaken vigorously to ensure complete dissolution and then heated in an oven at 65℃ for 15 minutes. The tube was removed from the oven and cooled down to room temperature. The solution containing the completely desilylated oligonucleotide was cooled on dry ice. 2 ml of ice-cold n-butanol (-20℃) were carefully added in 0.5 ml portions to precipitate the oligonucleotides. The precipitate was filtered, washed with 1 ml ice-cold n-butanol, and subsequently dissolved in 0.01 M Tris (hydroxymethyl) aminomethanol hydrochloride buffer.
(2) Single strand purification
The purification of oligonucleotides was performed on an AKTA explorer 10 equipped with a Source 15Q 4.6/100 PE column using the following conditions: buffer A: (10 mM Tris-HCl, 1 mM EDTA, pH 7.5) , B: (10 mM Tris-HCl, 1 mM EDTA, 2M NaCl, pH 7.5) , gradient: 10%B to 60%B in 25 min, flow rate: 1 ml/min. The pure oligonucleotides were collected and desalting by a HiPrep 26/10 Desalting column.
(3) Annealing to form duplex
For duplex, after the generation of desalted purified single strand solutions, sense strand (passenger strand) and antisense strand (guide strand) were mixed by equal volumes at equimolar concentration in the tube. Place the tube in a heat block at 95℃ for 5 min and then cool to room temperature then were subsequently lyophilized to powder.
Cell culture and treatment
Primary mouse hepatocytes (PMH) were isolated from the liver of C57BL/6J mice (Beijing Vital River Laboratory Animal Technology Co., Ltd. ) and cultured under the conditions of 5%CO2 and 37℃ in modified Willian’s Medium E (WME) medium (A12176-01, Gibco, Thermo Fisher Scientific, Carlsbad, CA) supplemented with 1%Insulin (S6955, Selleck, US) and 1%penicillin/streptomycin (Gibco) . T98G cells (Cobioer, Cat#CBP60301) were cultured under the conditions of 5%CO2 and 37℃ in modified MEM medium (Gibco, Thermo Fisher Scientific, Carlsbad, CA) supplemented with 10%bovine calf serum (Sigma-Aldrich) and 1%penicillin/streptomycin. SK-N-AS (Procell, Wuhan, China, Cat#CL-0621) and C2C12 (CBP60252, Cobioer, China) cells were maintained in DMEM medium supplemented with 10%FBS, and 1%penicillin/streptomycin. SMA patient derived fibroblasts GM03813 cells were obtained from Coriell Institute (Camden, NJ, USA) and cultured at 5%CO2 and 37℃ in modified MEM medium supplemented with 15%bovine calf serum, 1%NEAA and 1%penicillin/streptomycin. Transfection was carried out using Lipofectamine RNAiMax (ThermoFisher, Waltham, MA, USA) in growth media without antibiotics according to the manufacturer’s protocol. Free uptake was carried out by directly adding oligonucleotides into culture medium containing the PMH cells.
Primary mouse hepatocyte (PMH) isolation
C57BL/6J mice (B204, Beijing, China) were anesthetized with isoflurane and perfused by initial flushing reagent and digestion reagent successively. The liver was placed into a 10 cm dish and torn apart using forceps in culture medium. The cell suspension was collected by filtering through a 70-75-micron membrane in 50 mL conical tube, followed by centrifuging at 4℃ for 2 minutes at 100 × g in a swinging-arm centrifuge. Cells were washed with 20 mL cold PBS after removing the supernatant (Repeat this step twice) . Cells with at least 80%viability were allowed to proceed with the assay. Appropriate number of cells were seeded to the cell culture plates which were pre-coated with collagen I 4-12 hrs in advance to yield a final confluence of 90-95%prior to the start of the assay.
Animal studies
All animal procedures were conducted by certified laboratory personnel using protocols consistent with local and state regulations and approved by the Institutional Animal Care and Use Committee. C57BL/6J mice (4-6 weeks old) purchased from SPF Biotechnology Co., LTD (B204, Beijing, China) . Sprague-Dawley female rats (A102, SPF, China) were
purchased from SPF (Suzhou) Biotechnology Co., LTD. Parental transgenic hSOD1G93A mice (Strain ID #004435) were purchased from the Jackson Laboratory (Bar Harbor, ME, USA) and imported into China via Nantong University (Nantong City, Jiangsu Province, China) . Formulations for in vivo studies were prepared fresh prior to use by dissolving aliquots of lyophilized oligonucleotide into saline or aCSF to create stock solutions for dilution to the intended treatment concentrations. Animals were randomly allocated into study groups based on body weight and sex.
Intracerebroventricular (ICV) injection
Avertin (1.2%) was prepared fresh and sterilized via 0.2-micron filter. Mice were dosed at 0.30-0.35 ml per 10 g body weight via intraperitoneal (IP) injection in a stereotaxic apparatus to rapidly induce anesthesia for up to 30 minutes. An approximate 11.5 mm incision was made in the animal’s scalp and a 25-gauge needle attached to a Hamilton syringe containing the appropriate siRNA formulation was placed at bregma level. The needle was moved to the appropriate anterior/posterior and medial/lateral coordinates (0.2 mm anterior/posterior and 1 mm to the right medial/lateral) . A total of 10 μL was injected into the lateral ventricle at an approximate rate of 1 μL/s. Following treatment, the needle was slowly withdrawn, and the wound sutured close.
Intrathecal (IT) injection
Anesthesia was administered via 3.0%isoflurane in an induction chamber for continuous 10 mins. Hair was shaved around the injection site at the base of the tail and cleaned with 75%ethanol. The space between the L5-L6 spinous processes was identified and a 30-gauge needle attached to a microliter syringe containing the appropriate drug formulations was slowly inserted into the intradural space until a tail flick was observed. The needle position was subsequently secured in which 30 μL total volume of solution was injected over the course of 1 min.
Intravenous (IV) injection into the tail vein
Mice were exposed to an infrared lamp for 2-3 min to dilate the veins, and then held in the restrainer to straighten the tail. The tail was wiped with 75%alcohol and the needle was inserted 2 to 4 mm parallel to the tail vein into the lumen, keeping the bevel of the needle upwards. The preformed solution was injected slowly and should be free of resistance if administered correctly. The recommended injection volume for test article is 200 μL and the
injection rate don’t exceed 5 ml/min. At the end of administration, the injection site is pressed firmly with a cotton swab or finger to prevent backflow of the administration solution and/or blood.
Intravitreal (IVT) injection
SD rats were housed in animal facility of Ractigen (Nantong, Jiangsu, China) and fed for at least three days prior to the intravitreal (IVT) injection of compounds. SD rats were anesthetized in an isoflurane (RWD, R510-22-16) induction chamber (5%isoflurane in 100%medical oxygen, 2 L/min) until they had no response to toe pinches. SD rats were transferred to the experimental operating platform and positioned for delivery of isoflurane (2%isoflurane in 100%medical oxygen, 1.5 L/min) using a homemade face mask during the procedure of IVT injection. Before the IVT injection of compounds, one drop of 0.5%alcaine as topical anesthetics was applied to the injected eye (left eye) . An anterior chamber paracentesis was performed using a 30-gauge needle, followed by approximately 5 μL aqueous humor were outflowed. Each compound for corresponding group was dissolved in 4 μL normal saline and loaded into a 30-gauge needle for IVT injection. The compounds were administered by inserting at a 45° angle of the needle through the sclera into the vitreous body, then injected into the posterior chamber keeping for 5 seconds to avoid the leaking. At the end of administration, the injected eye was administered with antibiotics to prevent infection after the IVT injection.
Intravesical bladder (IVB) instillation
Mice were anesthetized and placed on a temperature-controlled pad at 28-31℃. Each mouse will be excreted urine from the bladder by urinary catheterization and to wash the bladder with saline before IVB instillation. A 2 cm catheter attached to a 1 ml syringe containing compound solution was intubated into bladder via urinary meatus. A total of 50 μL solution was injected into the bladder through an indwelling urinary catheterization at ~1.5 hours.
RNA Isolation and reverse transcription-quantitative polymerase chain reaction (RT-qPCR)
Total cellular RNA was isolated from cell culture using the RNeasy Plus Mini kit (Qiagen, Hilden, Germany) according to the manufacturer’s protocol. Animal tissues were stored in RNAlaterTM (AM7021, Thermo Fisher, Carlsbad, CA, USA) prior to sample
processing in which total RNA samples were isolated using the MagPure Total RNA Micro LQ kit (Magen, R6621, Guangzhou, Guangdong, China) in conjunction with the auto-pure96 machine (ALLSHENG, Hangzhou, Zhejiang, China) . The resultant RNA (1 μg) was reverse transcribed into cDNA by using a PrimeScript RT kit containing gDNA Eraser (Takara, Shlga, Japan) . The resultant cDNA was amplified in a Roche LightCycler 480 Multiwell Plate 384 (Roche, ref: 4729749001, US) using SYBR Premix Ex Taq II (Takara, Shlga, Japan) reagents and primers specific for amplifying target genes of interest. Melting curves were performed after amplification to confirm primer specificity. Reaction conditions were as follows: Reverse transcription reaction (stage 1) : 42℃ for 5 min, 95℃ for 10 sec; PCR reaction (stage 2) : 95℃ for 5 sec, 60℃ for 30 sec, 72℃ for 10 sec; 40 cycles of amplification; Melting curve (stage 3) . PCR reaction conditions are shown in Table 8 and Table 9. Primer sequences are listed in Table 10.
Table 8: RT reaction
Table 9: RT-qPCR reaction
Table 10. Primer sequences for RT-qPCR assay
One reference gene
To calculate the expression level (Erel) of target gene mRNA in an siRNA-and saRNA-transfected sample relative to control treatment (Mock) , the averaged Ct values of the target gene and the internal reference gene were substituted into the following Formula
Erel= 2 (CtTm-CtTs) /2 (CtRm-CtRs)
Erel= 2 (CtTm-CtTs) /2 (CtRm-CtRs)
wherein CtTm was the Ct value of the target gene from the mock-treated sample; CtTs was the Ct value of the target gene from the siRNA-treated sample; CtRm was the Ct value of the internal reference gene from the mock-treated sample; CtRs was the Ct value of the internal reference gene from the siRNA-treated sample.
Two reference genes
To calculate the expression level (Erel) of target gene mRNA in an siRNA-and saRNA-transfected sample relative to control treatment (Mock) , the Ct values of the target gene and the two internal reference genes were substituted into the following Formula
Erel=2 (CtTm-CtTs) / ( (2 (CtR1m-CtR1s) *2 (CtR2m-CtR2s) ) (1/2) )
Erel=2 (CtTm-CtTs) / ( (2 (CtR1m-CtR1s) *2 (CtR2m-CtR2s) ) (1/2) )
wherein CtTm was the Ct value of the target gene from the mock-treated sample; CtTs was the Ct value of the target gene from the siRNA-treated sample; CtR1m was the Ct value of the internal reference gene 1 from the mock-treated sample; CtR1s was the Ct value of the internal reference gene 1 from the siRNA-treated sample; CtR2m was the Ct value of the internal reference gene 2 from the mock-treated sample; and CtR2s was the Ct value of the internal reference gene 2 from the siRNA-treated sample.
Stem-loop RT-qPCR
To quantify oligonucleotides in biological samples, animal tissues were harvested using Heat-Lysis method and stored lysate at -80℃. Duplex, antisense strand and formulated saRNAs were used for the preparation of serial 10-fold dilutions into 95℃ boiled tissue (100 mg/mL) or plasma (1: 10 diluted) in 1x lysis buffer. saRNA concentrations in nM were converted to ng/g using the corresponding molecular weights. Two non-template controls were included in all experiments. The first control contains the water used to prepare the transcription master mix and the second contains the lysis buffer used as diluent for samples and standards.
Reverse transcription reactions were performed using a Takara Reverse Transcription kit (Takara, RR037A) . A total of 4 μL of cDNA (1: 40 dilution) from the previous step was added into the PCR amplification reaction mix (0.5 μM forward primer, 0.5 μM reverse primer, 2 × TG Green premix Ex Taq II) . The qPCR reaction was run with the option ‘Standard Curve’ in a Light cycler 480. Stem-loop RT-qPCR reaction conditions are shown in Table 11 and Table 12. Stem-loop primer sequences are listed in Table 13.
Table 11. Stem-loop RT reaction
Table 12. Stem-loop RT-qPCR reaction
Table 13. Primer sequences for stem-loop RT-qPCR assay
ELISA assay
Plasma was prepared by collecting 225 μL blood per mouse into 1.5 mL EP tube with 25 μL 3.2%citrate as an anticoagulant, followed by mixing and centrifuging 15 min at 2500 g within 30 minutes of collection. The samples were tested immediately or aliquoted and stored at -80℃ without repeated freeze-thaw cycles. A suggested 1000-fold dilution was diluted with Tris-BSA buffer (Cord: 221304, BioMed, France) , mixed well and started assay. FVII Standard (Normal Mouse Plasma) was prepared by taking one tube of pool plasma which is made by pooling plasma samples of five normal mice (C57BL/6J) , stored at -80℃ and was used as an initiation of a standard plasma. 20 μL pool plasma was pipetted to 480 μL Tris-BSA buffer (R4) to obtain a 1: 25 Solution A in a 1.5 mL tube. Then 50 μL Solution A was pipetted to 950 μL Tris-BSA buffer to obtain a 1: 500 stock solution. This stock solution was used to produce a dilution series of standard plasma. Each tube was mixed thoroughly before the next transfer.
In this assay, a standard plasma dilution of 1: 1000 was assigned value of 100 %FVII activity. By definition, the stock solution was assigned 200%FVII activity. The dynamic range was from 0 to 200%FVII activity. The stock solution (i.e., 200%FVII activity) was served as the high value standard. Tris-BSA buffer (R4) was served as the zero-value standard. FVII protein expression levels were detected by the OD values using BIOPHEN FVII ELISA kits (Cord : 221304, BioMed, France) following the instructions provided by the manufacturer of the kits.
Statistical analysis
Differences between groups of continuous variables were compared using one-way analysis of variance (ANOVA) followed by Dunnett’s multiple comparisons. A P value of less than 0.05 was considered statistically significant between two groups. *represents p < 0.05, **represents p < 0.01, ***represents p < 0.001, ****represents p < 0.0001.
While the invention has been particularly shown and described with reference to a preferred embodiment and various alternate embodiments, it is understood by persons skilled in the relevant art that various changes in form and details can be made therein without departing from the spirit and scope of the invention.
Claims (46)
- An oligonucleotide delivery enhancing compound comprising a nitrogen-containing five membered heterocyclic ring moiety and at least one substituent directly or indirectly attachable to an oligonucleotide.
- The oligonucleotide delivery enhancing compound according to claim 1, having a structure represented by Formula AI or Formula AII
wherein each independently represents a covalent single or double bond; X, on each occurrence, is an atom selected from the group consisting of carbon, nitrogen, oxygen and sulfur; each of F, G, H and I is independently selected from the group consisting of carbon, nitrogen, oxygen and sulfur;wherein m is an integer of 1, 2 or 3, n is an integer of 1, 2 or 3, and m+n=4;wherein C, on each occurrence, is either absent or selected from the group consisting of hydrogen, halogen atom, hydroxyl, (C1-C20) alkyl, (C1-C20) alkoxy, halogenated (C1-C20) alkyl and halogenated (C1-C20) alkoxy;wherein B, on each occurrence, is independently selected from the group consisting of hydroxyl, -C (O) OH, -P (O) 2-OH, -P (O) -OH, -P (O) (S) -OH, -CN, - (C1-C22) alkyl, - (C1-C22) alkenyl, - (C1-C22) alkylene-OH, - (C3-C22) cycloalkylene-OH, - (C6-C22) arylene-OH, - (C6-C22) heteroarylene-OH, - (C1-C22) alkylene-C (O) OH, - (C3-C22) cycloalkylene-C (O) OH, - (C6-C22) arylene-C (O) OH, - (C5-C22) heteroarylene-C (O) OH, -O-C (O) - (C1-C22) alkylene-C (O) NH2, - (C1-C22) alkylene-O-C (O) - (C1-C22) alkylene-C (O) NH2, -O-C (O) - (C1-C22) alkylene-C (O) OH, - (C1-C22) alkylene-O-C (O) - (C1-C22) alkylene-C (O) OH, -C (O) - (C1-C22) alkylene-C (O) NH2, - (C1-C22) alkylene-C (O) - (C1-C22) alkylene-C (O) NH2, -C (O) -NH- (C1-C22) alkylene-OH, -C (O) -NH- (C1-C22) alkylene-C (O) OH, - (C1-C22) alkylene-C (O) -NH- (C1-C22) alkylene-C (O) OH, - (C1- C30) alkylene-P (O) 2-OH, - (C1-C22) alkylene-O-P (-N (C1-C22 alkyl) 2) -O- (C1-C22) alkylene-CN, - (C1-C22) alkylene-O-P (-N (C1-C22 alkyl) 2) -O- (C1-C22) alkylene-OH, - (C1-C22) alkylene-O-P (-N (C1-C22 alkyl) 2) -O- (C1-C22) alkylene-NH2, - (C1-C22) alkylene-O-P (-N (C1-C22 alkyl) 2) -O- (C1-C22) alkylene-C (O) OH, -C (O) -NH- (C1-C22) alkylene-O-P (-N (C1-C22 alkyl) 2) -O- (C1-C22) alkylene-CN, -C (O) -NH- (C1-C22) alkylene-O-P (-N (C1-C22 alkyl) 2) -O- (C1-C22) alkylene-OH, -C (O) -NH- (C1-C22) alkylene-O-P (-N (C1-C22 alkyl) 2) -O- (C1-C30) alkylene-C (O) OH, -C (O) -NH- (C1-C22) alkylene-O-P (-N (C1-C22 alkyl) 2) -O- (C1-C22) alkylene-NH2, - (C1-C22) alkylene-C (O) -NH-(C1-C22) alkylene-O-P (-N (C1-C22 alkyl) 2) -O- (C1-C22) alkylene-CN, - (C1-C22) alkylene-C (O) -NH- (C1-C22) alkylene-O-P (-N (C1-C22 alkyl) 2) -O- (C1-C22) alkylene-OH, - (C1-C22) alkylene-C (O) -NH- (C1-C22) alkylene-O-P (-N (C1-C22 alkyl) 2) -O- (C1-C22) alkylene-C (O) OH, - (C1-C22) alkylene-C (O) -NH- (C1-C22) alkylene-O-P (-N (C1-C22 alkyl) 2) -O- (C1-C22) alkylene-NH2, - (C1-C22) alkylene -P (O) -OH, - (C1-C22) alkylene-P (O) (S) -OH, - (C1-C22) alkylene-CN,wherein each of A1, A2 and A3 is either absent or a substituent independently selected from the group consisting of -H, -OH, -O-R1, -SH, - (C1-C25) alkyl, halogenated - (C1-C25) alkyl, - (C2-C22) alkenyl, - (C1-C22) alkylene-OH, - (C3-C22) cycloalkyl, - (C3-C22) cycloalkenyl, - (C1-C22) alkylene- (C3-C22) cycloalkyl, - (C1-C22) alkylene-R1, - (C1-C22) alkylene-O-R1, - (C1-C22) alkylene-COOR1, -C (O) O-R1, -O- (C1-C22) alkyl, -S- (C1-C22) alkyl, -C (O) -R1, -C (O) - (C1-C22) alkyl, -O-C (O) - (C1-C22) alkyl, -O-C (O) -R1, - (C1-C22) alkylene-O-C (O) -R1, -C (O) - (C1-C22) alkylene-OH, -C (O) - (C1-C22) alkylene-R1, -C (O) - (C1-C22) alkylene-NH-R1, -C (O) - (C1-C22) alkylene-NR2-R1, -O-C (O) - (C1-C22) alkylene-OH, -O-C (O) - (C1-C22) alkylene-R1, -adamantyl, - (C1-C22) alkylene-adamantyl, -O-adamantly, -C (O) - (C1-C22) alkylene-adamantyl, - (C1-C22) alkylene-C (O) - (C1-C22) alkylene-adamantyl, -NH-C (O) - (C1-C22) alkylene-adamantyl, - (C1-C22) alkylene-NH-C (O) - (C1-C22) alkyl, - (C1-C22) alkylene-NH-C (O) -halogenated (C1-C22) alkyl, -CH (NH-CO- (C1-C22) alkyl) - (C1-C22) alkylene-NH-C (O) - (C1-C22) alkyl, -C (O) - (C1-C22) alkylene-C (O) -NH-C [- (C1-C22) alkylene-O- (C1-C22) alkylene-C (O) -NH- (C1-C22) alkylene-NH-C (O) - (C1-C22) alkyl] 3, -C (O) - (C1-C22) alkylene-C (O) -NH-C [- (C1-C22) alkylene-O- (C1-C22) alkylene-C (O) -NH- (C1-C22) alkylene-NH-C (O) - (C1-C22) alkylene-R1] 3, -CH (NH-CO-halogenated (C1-C22) alkyl) - (C1-C22) alkylene-NH-C (O) -halogenated (C1-C22) alkyl, - (C1-C22) alkylene-NH-C (O) - (C1-C22) alkylene-adamantyl, - (C1-C22) alkylene-NR2-C (O) - (C1-C22) alkylene-adamantyl, - (C1-C22) alkylene- (C1-C6 alkylene oxide) (1-20) -NH-C (O) - (C1-C22) alkylene-adamantyl, -C (O) NH- (C1-C22) alkyl, -C (O) NH-R1, -C (O) NR2-R1, -C (O) NH- (C1- C22) alkylene-OH, -C (O) NH- (C1-C22) alkylene-COOH, -NH-C (O) - (C1-C22) alkyl, -NH-C (O) -R1, -NR2-C (O) -R1, -O-P (O) 2-O-R1, -OP (O) (S) -O-R1, -O-P (O) -O-R1, -NH-R1, -NR2-R1, - (C1-C22) alkylene-NH-R1, - (C1-C22) alkylene-NR2-R1, -C (O) - (C1-C22) alkylene-C (O) -R1, -C (O) - (C1-C22) alkylene-C (O) O-R1, -C (O) - (C1-C22) alkylene-NH-C (O) -R1, -C (O) - (C1-C22) alkylene-NR2-C (O) -R1, - (C1-C22) alkylene-C (O) -R1, - (C1-C22) alkylene-NH-C (O) -R1, - (C1-C22) alkylene-NH-C (O) - (C1-C22) alkylene-R1, - (C1-C22) alkylene-NH-C (O) - (C1-C22) alkylene-NH-C (O) - (C1-C22) alkylene-R1, - (C1-C22) alkylene-C (O) -NH- (C1-C22) alkylene-R1, - (C1-C22) alkylene-C (O) -NH- (C1-C22) alkylene-C (O) -NH- (C1-C22) alkylene-R1, - (C1-C22) alkylene-C (O) -NH- (C1-C22) alkylene-NH-C (O) - (C1-C22) alkylene-R1, - (C1-C22) alkylene-NR2-C (O) - (C1-C22) alkylene-R1, - (C1-C22) alkylene-O-P (-N (C1-C22 alkyl) 2) -O- (C1-C22) alkylene-CN, - (C1-C22) alkylene-O-P (-N (C1-C22 alkyl) 2) -O- (C1-C22) alkylene-OH, - (C1-C22) alkylene-O-P (-N (C1-C22 alkyl) 2) -O- (C1-C22) alkylene-NH2, - (C1-C22) alkylene-O-P (-N (C1-C22 alkyl) 2) -O- (C1-C22) alkylene-C (O) OH, -C (O) -NH- (C1-C22) alkylene-O-P (-N (C1-C22 alkyl) 2) -O- (C1-C22) alkylene-CN, -C (O) -NH- (C1-C22) alkylene-O-P (-N (C1-C22 alkyl) 2) -O- (C1-C22) alkylene-OH, -C (O) -NH- (C1-C22) alkylene-O-P (-N (C1-C22 alkyl) 2) -O- (C1-C30) alkylene-C (O) OH, -C (O) -NH- (C1-C22) alkylene-O-P (-N (C1-C22 alkyl) 2) -O- (C1-C22) alkylene-NH2, - (C1-C22) alkylene-C (O) -NH- (C1-C22) alkylene-O-P (-N (C1-C22 alkyl) 2) -O- (C1-C22) alkylene-CN, - (C1-C22) alkylene-C (O) -NH- (C1-C22) alkylene-O-P (-N (C1-C22 alkyl) 2) -O- (C1-C22) alkylene-OH, - (C1-C22) alkylene-C (O) -NH- (C1-C22) alkylene-O-P (-N (C1-C22 alkyl) 2) -O- (C1-C22) alkylene-C (O) OH, - (C1-C22) alkylene-C (O) -NH- (C1-C22) alkylene-O-P (-N (C1-C22 alkyl) 2) -O- (C1-C22) alkylene-NH2, - (C1-C22) alkylene -P (O) -OH, - (C1-C22) alkylene-P (O) (S) -OH, - (C1-C22) alkylene-CN, substituted or unsubstituted pyrrole, substituted or unsubstituted pyrroline, substituted or unsubstituted pyrrolidine, substituted or unsubstituted pyrazole, substituted or unsubstituted pyrazoline, substituted or unsubstituted pyrazolidine, substituted or unsubstituted imidazole, substituted or unsubstituted oxazole, substituted or unsubstituted thiazole, substituted or unsubstituted benzopyrrole, substituted or unsubstituted benzopyrroline, substituted or unsubstituted benzopyrrolidine, substituted or unsubstituted benzopyrazole, substituted or unsubstituted benzopyrazoline, substituted or unsubstituted benzopyrazolidine, substituted or unsubstituted benzoimidazole, substituted or unsubstituted benzooxazole, substituted or unsubstituted benzothiazole, and a substituent represented by Formula AIII,
wherein Y is selected from the group consisting of carbon, nitrogen, oxygen and sulfur; and each of P, Q, S and T is independently selected from the group consisting of carbon, nitrogen, oxygen and sulfur; and the asterisk refers to the site wherein the substituent represented by Formula AIII is linked with the structure represented by Formula AI or Formula AII;wherein each of R3, R4 and R5 is either absent or a substituent independently selected from the group consisting of -H, -OH, -O-R1, -SH, - (C1-C25) alkyl, halogenated - (C1-C25) alkyl, - (C2-C22) alkenyl, - (C1-C22) alkylene-OH, - (C3-C22) cycloalkyl, - (C3-C22) cycloalkenyl, - (C1-C22) alkylene- (C3-C22) cycloalkyl, - (C1-C22) alkylene-R1, - (C1-C22) alkylene-O-R1, - (C1-C22) alkylene-COOR1, -C (O) O-R1, -O- (C1-C22) alkyl, -S- (C1-C22) alkyl, -C (O) -R1, -C (O) - (C1-C22) alkyl, -O-C (O) - (C1-C22) alkyl, -O-C (O) -R1, - (C1-C22) alkylene-O-C (O) -R1, -C (O) - (C1-C22) alkylene-OH, -C (O) - (C1-C22) alkylene-R1, -C (O) - (C1-C22) alkylene-NH-R1, -C (O) - (C1-C22) alkylene-NR2-R1, -O-C (O) - (C1-C22) alkylene-OH, -O-C (O) - (C1-C22) alkylene-R1, -adamantyl, - (C1-C22) alkylene-adamantyl, -O-adamantly, -C (O) - (C1-C22) alkylene-adamantyl, - (C1-C22) alkylene-C (O) - (C1-C22) alkylene-adamantyl, -NH-C (O) - (C1-C22) alkylene-adamantyl, - (C1-C22) alkylene-NH-C (O) - (C1-C22) alkyl, - (C1-C22) alkylene-NH-C (O) -halogenated (C1-C22) alkyl, -CH (NH-CO- (C1-C22) alkyl) - (C1-C22) alkylene-NH-C (O) - (C1-C22) alkyl, -C (O) - (C1-C22) alkylene-C (O) -NH-C [- (C1-C22) alkylene-O- (C1-C22) alkylene-C (O) -NH- (C1-C22) alkylene-NH-C (O) - (C1-C22) alkyl] 3, -C (O) - (C1-C22) alkylene-C (O) -NH-C [- (C1-C22) alkylene-O- (C1-C22) alkylene-C (O) -NH- (C1-C22) alkylene-NH-C (O) - (C1-C22) alkylene-R1] 3, -CH (NH-CO-halogenated (C1-C22) alkyl) - (C1-C22) alkylene-NH-C (O) -halogenated (C1-C22) alkyl, - (C1-C22) alkylene-NH-C (O) - (C1-C22) alkylene-adamantyl, - (C1-C22) alkylene-NR2-C (O) - (C1-C22) alkylene-adamantyl, - (C1-C22) alkylene- (C1-C6 alkylene oxide) (1-20) -NH-C (O) - (C1-C22) alkylene-adamantyl, -C (O) NH- (C1-C22) alkyl, -C (O) NH-R1, -C (O) NR2-R1, -C (O) NH- (C1-C22) alkylene-OH, -C (O) NH- (C1-C22) alkylene-COOH, -NH-C (O) - (C1-C22) alkyl, -NH-C (O) - R1, -NR2-C (O) -R1, -O-P (O) 2-O-R1, -OP (O) (S) -O-R1, -O-P (O) -O-R1, -NH-R1, -NR2-R1, - (C1-C22) alkylene-NH-R1, - (C1-C22) alkylene-NR2-R1, -C (O) - (C1-C22) alkylene-C (O) -R1, -C (O) - (C1-C22) alkylene-C (O) O-R1, -C (O) - (C1-C22) alkylene-NH-C (O) -R1, -C (O) - (C1-C22) alkylene-NR2-C (O) -R1, - (C1-C22) alkylene-C (O) -R1, - (C1-C22) alkylene-NH-C (O) -R1, - (C1-C22) alkylene-NH-C (O) - (C1-C22) alkylene-R1, - (C1-C22) alkylene-NH-C (O) - (C1-C22) alkylene-NH-C (O) - (C1-C22) alkylene-R1, - (C1-C22) alkylene-C (O) -NH- (C1-C22) alkylene-R1, - (C1-C22) alkylene-C (O) -NH-(C1-C22) alkylene-C (O) -NH- (C1-C22) alkylene-R1, - (C1-C22) alkylene-C (O) -NH- (C1-C22) alkylene-NH-C (O) - (C1-C22) alkylene-R1, - (C1-C22) alkylene-NR2-C (O) - (C1-C22) alkylene-R1, - (C1-C22) alkylene-O-P (-N (C1-C22 alkyl) 2) -O- (C1-C22) alkylene-CN, - (C1-C22) alkylene-O-P (-N (C1-C22 alkyl) 2) -O- (C1-C22) alkylene-OH, - (C1-C22) alkylene-O-P (-N (C1-C22 alkyl) 2) -O- (C1-C22) alkylene-NH2, - (C1-C22) alkylene-O-P (-N (C1-C22 alkyl) 2) -O- (C1-C22) alkylene-C (O) OH, -C (O) -NH- (C1-C22) alkylene-O-P (-N (C1-C22 alkyl) 2) -O- (C1-C22) alkylene-CN, -C (O) -NH- (C1-C22) alkylene-O-P (-N (C1-C22 alkyl) 2) -O- (C1-C22) alkylene-OH, -C (O) -NH- (C1-C22) alkylene-O-P (-N (C1-C22 alkyl) 2) -O- (C1-C30) alkylene-C (O) OH, -C (O) -NH- (C1-C22) alkylene-O-P (-N (C1-C22 alkyl) 2) -O- (C1-C22) alkylene-NH2, - (C1-C22) alkylene-C (O) -NH- (C1-C22) alkylene-O-P (-N (C1-C22 alkyl) 2) -O- (C1-C22) alkylene-CN, - (C1-C22) alkylene-C (O) -NH- (C1-C22) alkylene-O-P (-N (C1-C22 alkyl) 2) -O- (C1-C22) alkylene-OH, - (C1-C22) alkylene-C (O) -NH- (C1-C22) alkylene-O-P (-N (C1-C22 alkyl) 2) -O- (C1-C22) alkylene-C (O) OH, - (C1-C22) alkylene-C (O) -NH- (C1-C22) alkylene-O-P (-N (C1-C22 alkyl) 2) -O- (C1-C22) alkylene-NH2, - (C1-C22) alkylene -P (O) -OH, - (C1-C22) alkylene-P (O) (S) -OH, - (C1-C22) alkylene-CN, substituted or unsubstituted pyrrole, substituted or unsubstituted pyrroline, substituted or unsubstituted pyrrolidine, substituted or unsubstituted pyrazole, substituted or unsubstituted pyrazoline, substituted or unsubstituted pyrazolidine, substituted or unsubstituted imidazole, substituted or unsubstituted oxazole, substituted or unsubstituted thiazole, substituted or unsubstituted benzopyrrole, substituted or unsubstituted benzopyrroline, substituted or unsubstituted benzopyrrolidine, substituted or unsubstituted benzopyrazole, substituted or unsubstituted benzopyrazoline, substituted or unsubstituted benzopyrazolidine, substituted or unsubstituted benzoimidazole, substituted or unsubstituted benzooxazole, and substituted or unsubstituted benzothiazole,wherein R7, on each occurrence, is attached to any one of P, Q, S and T, and is either absent or selected from the group consisting of hydrogen, halogen atom, hydroxyl, (C1-C20) alkyl, (C1-C20) alkoxy, halogenated (C1-C20) alkyl and halogenated (C1-C20) alkoxy;wherein M is an integer of 0, 1, 2 or 3;wherein R6 is attached to any one of P, Q, S and T, and is selected from the group consisting of direct bond, -O-, -C (O) O-, -O-C (O) -, -P (O) 2-O-, -O-P (O) 2-O-, -P (O) (S) -O-, -O-P (O) (S) -O-, -O-P (O) -O-, - (C1-C22) alkylene-, - (C1-C22) alkylene-O-, -O- (C1-C22) alkylene-, - (C1-C22) alkylene-NH-, -NH- (C1-C22) alkylene-, -C (O) - (C1-C22) alkylene-, - (C1-C22) alkylene-C (O) -, -C (O) -O- (C1-C22) alkylene-, - (C1-C30) alkylene-C (O) -O-, -C (O) -NH- (C1-C22) alkylene-, -C (O) -NH-(C1-C22) alkylene-C (O) -O-, -C (O) -NH- (C1-C22) alkylene-O-C (O) -, -C (O) -NH- (C1-C22) alkylene-O-C (O) -O-, -C (O) -NH- (C1-C22) alkylene-O-, -C (O) -N ( (C1-C22) alkyl) - (C1-C22) alkylene-, -C (O) -N ( (C1-C22) alkyl) - (C1-C22) alkylene-O-, -C (O) -NH- (C1-C22) alkylene-C (O) -NH- (C1-C22) alkylene-, -C (O) -NH- (C1-C22) alkylene-NH-C (O) - (C1-C22) alkylene-, -C (O) -NH- (C1-C22) alkylene-C (O) -NH-, -C (O) -N ( (C1-C22) alkyl) - (C1-C22) alkylene-C (O) -N ( (C1-C22) alkyl) - (C1-C22) alkylene-, -C (O) -N ( (C1-C22) alkyl) - (C1-C22) alkylene-C (O) -N ( (C1-C22) alkyl) -, - (C1-C22) alkylene-C (O) -NH- (C1-C22) alkylene-C (O) -NH- (C1-C22) alkylene-, - (C1-C22) alkylene-C (O) -NH- (C1-C22) alkylene-C (O) -NH-, -NH-C (O) - (C1-C22) alkylene-, -NH-C (O) - (C1-C22) alkylene-C (O) -O-, -NH-C (O) - (C1-C22) alkylene-C (O) -, -NH-C (O) - (C1-C22) alkylene-O-, -N ((C1-C22) alkyl) -C (O) - (C1-C22) alkylene-, -N ( (C1-C22) alkyl) -C (O) - (C1-C22) alkylene-O-, -NH-C (O) - (C1-C22) alkylene-NH-C (O) - (C1-C22) alkylene-, -NH-C (O) - (C1-C22) alkylene-NH-C (O) -, -N ((C1-C22) alkyl) -C (O) - (C1-C22) alkylene-N ( (C1-C22) alkyl) -C (O) - (C1-C22) alkylene-, -N ( (C1-C22) alkyl) -C (O) - (C1-C22) alkylene-N ( (C1-C22) alkyl) -C (O) -, - (C1-C22) alkylene-NH-C (O) - (C1-C22) alkylene-NH-C (O) - (C1-C22) alkylene-, - (C1-C22) alkylene-NH-C (O) - (C1-C22) alkylene-NH-C (O) -, - (C1-C22) alkylene -P (O) 2-O-, - (C1-C22) alkylene-O-P (O) 2-O-, - (C3-C22) cycloalkylene-, - (C3-C22) cycloalkylene-O-, -O- (C3-C22) cycloalkylene-, - (C6-C22) arylene-, - (C6-C22) arylene-O-, -O- (C6-C22) arylene-, - (C6-C22) arylene-NH-, -NH- (C6-C22) arylene-, -C (O) - (C6-C22) arylene-, - (C6-C22) arylene-C (O) -, -C (O) -O- (C6-C22) arylene-, - (C6-C22) arylene-C (O) -O-, -C (O) -NH- (C6-C22) arylene-and -C (O) -NH- (C6-C22) arylene-C (O) -O-;wherein R1, on each occurrence, is independently selected from the group consisting of hydrogen, hydroxyl, - (C1-C22) alkyl, - (C3-C22) cycloalkyl, - (C6-C22) aryl, - (C1-C22) alkoxy, - (C3-C22) cycloalkoxy, - (C6-C22) aryloxy, -C (O) - (C1-C22) alkyl, -OC (O) (C1-C22) alkyl, -C (O) -O- (C1-C22) alkyl, -C (O) - (C3-C22) cycloalkyl, -OC (O) - (C3-C22) cycloalkyl, -C (O) -O- (C3-C22) cycloalkyl, -C (O) - (C6-C22) aryloxy, -OC (O) - (C6-C22) aryloxy, -C (O) -O- (C6-C22) aryloxy, -C (O) -phosphate ester group, phosphodiester group, phosphoramidite group, saturated fatty acid group, unsaturated fatty acid group, glucosyl, acetamide glucosyl, galactosamine, N-acetyl galactosamine, lipid, PEG, steroid, lipophile, carbohydrate, cholesterol, adamantane, amino acid, peptide, chloroquine and alkaloid,wherein R2, on each occurrence, is independently selected from the group consisting of a halogen atom, a (C1-C12) alkyl, a (C1-C12) alkoxy, a (C1-C12) alkoxycarbonyl, a (C6-C16) aryl or a (C6-C16) aryloxycarbonyl;wherein one or more hydroxyl group, carboxyl group, amino group, nitrile group and phosphoric acid group contained in A1, A2, A3, B, C, R1, R2, R3, R4, R5 and R6 are optionally linked to a support material or protected with a terminal protective group; andwith the proviso that A1, A2 and A3 are not simultaneously hydrogen and R3, R4 and R5 are not simultaneously hydrogen. - The oligonucleotide delivery enhancing compound according to claim 1, comprising a moiety represented by Formula BI and at least one substituent directly or indirectly attachable to an oligonucleotide,
wherein X′ is selected from the group consisting of carbon, nitrogen, oxygen and sulfur; each of F′, G′, H′ and I′ is independently selected from the group consisting of carbon, nitrogen, oxygen and sulfur; and each of the asterisks refers to a site optionally linked to at least one substituent or an oligonucleotide directly or indirectly. - The oligonucleotide delivery enhancing compound according to claim 3, having a structure represented by Formula BII
wherein X′ is selected from the group consisting of carbon, nitrogen, oxygen and sulfur; and each of F′, G′, H′ and I′ is independently selected from the group consisting of carbon, nitrogen, oxygen and sulfur;wherein each of A1′, A2′ and A3′ is either absent or a substituent independently selected from the group consisting of -H, -R1′, -O-R1′, -S-R1′, -C (O) -R1′, -C (O) O-R1′, -O-C (O) -R1′, -C (O) NH-R1′, -C (O) NR2′-R1′, -NH-C (O) -R1′, -NR2′-C (O) -R1′, -O-P (O) 2-O-R1′, -OP (O) (S) -O-R1′, -O-P (O) -O-R1′, -NH-R1′, -NR2′-R1′, - (CH2) r′-NH-R1′, - (CH2) r′-NR2′-R1′, -C (O) - (CH2) r′-R1′, -C (O) - (CH2) r′-NH-R1′, -C (O) - (CH2) r′-NR2′-R1′, -C (O) - (CH2) r′-C (O) -R1′, -C (O) - (CH2) r′-C (O) O-R1′, -C (O) - (CH2) r′-NH-C (O) -R1′, -C (O) - (CH2) r′-NR2′-C (O) -R1′, - (CH2) r′-C (O) -R1′; - (CH2) r′-C (O) O-R1′; - (CH2) r′-O-C (O) -R1′, - (CH2) r′-R1′, - (CH2) r′-NH-C (O) -R1′, - (CH2) r′-NH-C (O) - (CH2) s′-R1′, - (CH2) r′-NH-C (O) - (C1-C22) alkylene-NH-C (O) - (CH2) s′-R1′, - (CH2) r′-C (O) -NH- (CH2) s′-R1′, - (CH2) r′-C (O) -NH- (C1-C22) alkylene-C (O) -NH- (CH2) s′-R1′, - (CH2) r′-C (O) -NH- (C1-C22) alkylene-NH-C (O) - (CH2) s′-R1′, - (CH2) r′-NR2′-C (O) - (CH2) s′-R1′, -CH (- (CH2) r′-NH-C (O) - (CH2) s′-R1′) (-NH-C (O) - (CH2) q′-R3′) , -CH (- (CH2) r′-C (O) -NH- (CH2) s′-R1′) (-C (O) -NH- (CH2) q′-R3′) , -N (- (CH2) r′-NH-C (O) - (CH2) s′-R1′) (-NH-C (O) - (CH2) q′-R3′) , -CH (- (CH2) r′-NH-C (O) - (CH2) s′-R1′) (- (CH2) p′-NH-C (O) - (CH2) q′-R3′) , -CR4′ (- (CH2) r′-NH-C (O) - (CH2) s′-R1′) (-NH-C (O) - (CH2) q′-R3′) , -CR4′ (- (CH2) r′-NH-C (O) - (CH2) s′-R1′) (- (CH2) p′-NH-C (O) - (CH2) q′-R3′) , -CH (- (CH2) r′-NR5′-C (O) - (CH2) s′-R1′) (-NR6′-C (O) - (CH2) q′-R3′) , -CH (- (CH2) r′-NR5′-C (O) - (CH2) s′-R1′) (- (CH2) p′-NR6′-C (O) - (CH2) q′-R3′) , -CR4′ ( (CH2) r′-NR5′-C (O) - (CH2) s′-R1′) ( (CH2) p′-NR6′-C (O) - (CH2) q′-R3′) , -CR4′ ( (CH2) r′-NR5′-C (O) - (CH2) s′-R1′) ( (CH2) p′-NR6′-C (O) - (CH2) q′-R3′) , or A1′ and A2′ are linked together so that A1′, A2′, the nitrogen atom linked with A1′ and the carbon atom linked with A2′ form a unsubstituted or substituted heterocyclic ring; wherein each of R1′ and R3′ is independently selected from the group consisting of hydrogen, hydroxyl, - (C1-C30) alkyl, - (C3-C50) cycloalkyl, - (C6-C50) aryl, - (C1-C30) alkoxy, - (C3-C50) cycloalkoxy, - (C6-C50) aryloxy, -C (O) - (C1-C30) alkyl, -OC (O) (C1-C30) alkyl, -C (O) -O- (C1-C30) alkyl, -C (O) - (C3-C50) cycloalkyl, -OC (O) - (C3-C50) cycloalkyl, -C (O) -O- (C3-C50) cycloalkyl, -C (O) - (C6-C50) aryloxy, -OC (O) - (C6-C50) aryloxy, -C (O) -O- (C6-C50) aryloxy, -C (O) -phosphate ester group, phosphodiester group, phosphoramidite group, saturated fatty acid group, unsaturated fatty acid group, glucosyl, acetamide glucosyl, galactosamine, N-acetyl galactosamine, lipid, PEG, steroid, lipophile, carbohydrate, cholesterol, adamantane, amino acid, peptide, ligand, nucleic acid, oligonucleotide, aptamer, small molecule, antibody, antibody fragment, chloroquine, alkaloid and targeting moiety, wherein one or more hydroxyl group, carboxyl group and amino group contained in each of R1′ and R3′ are optionally protected; wherein each of R2′, R4′, R5′ and R6′ is independently a halogen atom, a (C1-C12) alkyl, a (C1-C12) alkoxy, a (C1-C12) alkoxycarbonyl, a (C6-C16) aryl or a (C6-C16) aryloxycarbonyl; wherein each of r′, s′, p′ and q′ is an integer from 1 to 22; and with the proviso that A3′ is absent when X′ is oxygen, and A1′, A2′ and A3′ are not simultaneously hydrogen;wherein each C′ is attached to any one of F′, G′, H′ and I′, and is either absent or selected from the group consisting of hydrogen, halogen atom, hydroxyl, (C1-C20) alkyl, (C1-C20) alkoxy, halogenated (C1-C20) alkyl and halogenated (C1-C20) alkoxy;wherein m′ is an integer of 1, 2 or 3, n′ is an integer of 1, 2 or 3, and m′+n′=4;wherein each B′ is attached to any one of F′, G′, H′ and I′, and is independently selected from the group consisting of hydroxyl, -C (O) OH, - (C1-C30) alkoxy, -P (O) 2-OH, -P (O) -OH, -P (O) (S) -OH, -CN, - (C1-C30) alkylene-OH, - (C3-C50) cycloalkylene-OH, - (C6-C50) arylene-OH, - (C5-C50) heteroarylene-OH, - (C1-C30) alkylene-C (O) OH, - (C3-C50) cycloalkylene-C (O) OH, - (C6-C50) arylene-C (O) OH, - (C5-C50) heteroarylene-C (O) OH, -C (O) -NH- (C1-C30) alkylene-OH, -C (O) -NH- [ (C1-C30) alkylene-O] r′-H (wherein r′ is an integer of 1 to 22) , -C (O) -NH- [ (C1-C30) alkylene-O] r′- (C1-C30) alkylene-C (O) -OH (wherein r′ is an integer of 1 to 22) , -C (O) -NH- (C3-C50) cycloalkylene-OH, -C (O) -NH- (C6-C50) arylene-OH, -C (O) -NH- (C5-C50) heteroarylene-OH, -C (O) -NH- (C1-C30) alkylene-C (O) OH, -C (O) -NH- (C3-C50) cycloalkylene-C (O) OH, -C (O) -NH- (C6-C50) arylene-C (O) OH, -C (O) -NH- (C1-C30) alkylene- (C6-C50) arylene- (C1-C30) alkylene-C (O) OH, -C (O) -NH- (C5-C50) heteroarylene-C (O) OH, - (C1-C30) alkylene-C (O) -NH- (C1-C30) alkylene-C (O) OH, - (C1-C30) alkylene-C (O) -NH- (C3-C50) cycloalkylene-C (O) OH, - (C1-C30) alkylene-C (O) -NH- (C6-C50) arylene-C (O) OH, - (C1-C30) alkylene-C (O) -NH- (C5-C50) heteroarylene-C (O) OH, - (C1-C30) alkylene-P (O) 2-OH, - (C3-C50) cycloalkylene-P (O) 2-OH, - (C6-C50) arylene-P (O) 2-OH, - (C5-C50) heteroarylene-P (O) 2-OH, -C (O) -NH- (C1-C30) alkylene-O-P (-N (C1-C16alkyl) 2) -O- (C1-C30) alkylene-CN, -C (O) -NH- (C1-C30) alkylene-O-P (-N (C1-C16alkyl) 2) -O- (C1-C30) alkylene-OH, -C (O) -NH- (C1-C30) alkylene-O-P (-N (C1-C16alkyl) 2) -O- (C1-C30) alkylene-C (O) OH, -C (O) -NH- (C1-C30) alkylene-O-P (-N (C1-C16alkyl) 2) -O- (C1-C30) alkylene-NH2, - (C1-C30) alkylene-C (O) -NH- (C1-C30) alkylene-O-P (-N (C1-C16alkyl) 2) -O- (C1-C30) alkylene-CN, - (C1-C30) alkylene-C (O) -NH- (C1-C30) alkylene-O-P (-N (C1-C16alkyl) 2) -O- (C1-C30) alkylene-OH, - (C1-C30) alkylene-C (O) -NH- (C1-C30) alkylene-O-P (-N (C1-C16alkyl) 2) -O- (C1-C30) alkylene-C (O) OH, - (C1-C30) alkylene-C (O) -NH- (C1-C30) alkylene-O-P (-N (C1-C16alkyl) 2) -O- (C1-C30) alkylene-NH2, - (C1-C30) alkylene -P (O) -OH, - (C3-C50) cycloalkylene-P (O) -OH, - (C6-C50) arylene-P (O) -OH, - (C5-C50) -heteroarylene-P (O) -OH, - (C1-C30) alkylene-P (O) (S) -OH, - (C3-C50) cycloalkylene-P (O) (S) -OH, - (C6-C50) arylene-P (O) (S) -OH, - (C5-C50) heteroarylene-P (O) (S) -OH, - (C1-C30) alkylene-CN, - (C3-C50) cycloalkylene-CN, - (C6-C50) arylene-CN, - (C5-C50) heteroarylene-CN, lipid, PEG, steroid, lipophile, carbohydrate, cholesterol, adamantane, amino acid, peptide, chloroquine, alkaloid and a substituent represented by Formula BIII:
wherein Y′ is selected from the group consisting of carbon, nitrogen, oxygen and sulfur; and each of P′, Q′, S′ and T′ is independently selected from the group consisting of carbon, nitrogen, oxygen and sulfur; and the asterisk refers to the site wherein the substituent represented by Formula BIII is linked with any one of F′, G′, H′and I′ of Formula BII;where R7′ is selected from the group consisting of -O-, -C (O) O-, -O-C (O) -, -P (O) 2-O-, -O-P (O) 2-O-, -P (O) (S) -O-, -O-P (O) (S) -O-, -O-P (O) -O-, - (C1-C30) alkylene-, - (C1-C30) alkylene-O-, -O- (C1-C30) alkylene-, - (C1-C30) alkylene-NH-, -NH- (C1-C30) alkylene-, -C (O) - (C1-C30) alkylene-, - (C1-C30) alkylene-C (O) -, -C (O) -O- (C1-C30) alkylene-, - (C1-C30) alkylene-C (O) -O-, -C (O) -NH- (C1-C30) alkylene-, -C (O) -NH- (C1-C30) alkylene-C (O) -O-, -C (O) -NH- (C1-C30) alkylene-O-C (O) -, -C (O) -NH- (C1-C30) alkylene-O-C (O) -O-, -C (O) -NH- (C1-C30) alkylene-O-, -C (O) -N ( (C1-C20) alkyl) - (C1-C30) alkylene-, -C (O) -N ( (C1-C20) alkyl) - (C1-C30) alkylene-O-, -C (O) -NH- (C1-C30) alkylene-C (O) -NH- (C1-C30) alkylene-, -C (O) -NH- (C1-C30) alkylene-NH-C (O) - (C1-C30) alkylene-, -C (O) -NH- (C1-C30) alkylene-C (O) -NH-, -C (O) -N ( (C1-C20) alkyl) - (C1-C30) alkylene-C (O) -N ( (C1-C20) alkyl) - (C1-C30) alkylene-, -C (O) -N ( (C1-C20) alkyl) - (C1-C30) alkylene-C (O) -N ( (C1-C20) alkyl) -, - (C1-C30) alkylene-C (O) -NH- (C1-C30) alkylene-C (O) -NH-(C1-C30) alkylene-, - (C1-C30) alkylene-C (O) -NH- (C1-C30) alkylene-C (O) -NH-, -NH-C (O) - (C1-C30) alkylene-, -NH-C (O) - (C1-C30) alkylene-C (O) -O-, -NH-C (O) - (C1-C30) alkylene-C (O) -, -NH-C (O) - (C1-C30) alkylene-O-, -N ( (C1-C20) alkyl) -C (O) - (C1-C30) alkylene-, -N ( (C1-C20) alkyl) - C (O) - (C1-C30) alkylene-O-, -NH-C (O) - (C1-C30) alkylene-NH-C (O) - (C1-C30) alkylene-, -NH-C (O) - (C1-C30) alkylene-NH-C (O) -, -N ( (C1-C20) alkyl) -C (O) - (C1-C30) alkylene-N ( (C1-C20) alkyl) -C (O) - (C1-C30) alkylene-, -N ( (C1-C20) alkyl) -C (O) - (C1-C30) alkylene-N ( (C1-C20) alkyl) -C (O) -, - (C1-C30) alkylene-NH-C (O) - (C1-C30) alkylene-NH-C (O) - (C1-C30) alkylene-, - (C1-C30) alkylene-NH-C (O) - (C1-C30) alkylene-NH-C (O) -, - (C1-C30) alkylene -P (O) 2-O-, - (C1-C30) alkylene-O-P (O) 2-O-, - (C3-C50) cycloalkylene-, - (C3-C50) cycloalkylene-O-, -O- (C3-C50) cycloalkylene-, - (C3-C50) cycloalkylene-NH-, -NH- (C3-C50) cycloalkylene-, -C (O) - (C3-C50) cycloalkylene-, - (C3-C50) cycloalkylene-C (O) -, -C (O) -O- (C3-C50) cycloalkylene-, - (C3-C50) cycloalkylene-C (O) -O-, -C (O) -NH- (C3-C50) cycloalkylene-, -C (O) -NH- (C3-C50) cycloalkylene-C (O) -O-, -C (O) -NH- (C3-C50) cycloalkylene-O-, -C (O) -N ( (C1-C20) alkyl) - (C3-C50) cycloalkylene-, -C (O) -N ( (C1-C20) alkyl) - (C3-C50) cycloalkylene-O-, -C (O) -NH- (C3-C50) cycloalkylene-C (O) -NH- (C3-C50) cycloalkylene-, -C (O) -NH- (C3-C50) cycloalkylene-C (O) -NH-, -C (O) -N ( (C1-C20) alkyl) - (C3-C50) cycloalkylene-C (O) -N ( (C1-C20) alkyl) - (C3-C50) cycloalkylene-, -C (O) -N ( (C1-C20) alkyl) - (C3-C50) cycloalkylene-C (O) -N ( (C1-C20) alkyl) -, - (C3-C50) cycloalkylene-C (O) -NH- (C3-C50) cycloalkylene-C (O) -NH- (C3-C50) cycloalkylene-, - (C3-C50) cycloalkylene-C (O) -NH- (C3-C50) cycloalkylene-C (O) -NH-, - (C3-C50) cycloalkylene -P (O) 2-O-, - (C3-C50) cycloalkylene-O-P (O) 2-O-, - (C6-C50) arylene-, - (C6-C50) arylene-O-, -O- (C6-C50) arylene-, - (C6-C50) arylene-NH-, -NH- (C6-C50) arylene-, -C (O) - (C6-C50) arylene-, - (C6-C50) arylene-C (O) -, -C (O) -O- (C6-C50) arylene-, - (C6-C50) arylene-C (O) -O-, -C (O) -NH- (C6-C50) arylene-, -C (O) -NH- (C6-C50) arylene-C (O) -O-, -C (O) -NH- (C6-C50) arylene-O-, -C (O) -N ( (C1-C20) alkyl) - (C6-C50) arylene-, -C (O) -N ( (C1-C20) alkyl) - (C6-C50) arylene-O-, -C (O) -NH- (C6-C50) arylene-C (O) -NH- (C3-C50) cycloalkylene-, -C (O) -NH- (C6-C50) arylene-C (O) -NH-, -C (O) -N ( (C1-C20) alkyl) - (C6-C50) arylene-C (O) -N ( (C1-C20) alkyl) - (C6-C50) arylene-, -C (O) -N ( (C1-C20) alkyl) - (C6-C50) arylene-C (O) -N ( (C1-C20) alkyl) -, - (C6-C50) arylene-C (O) -NH- (C6-C50) arylene-C (O) -NH- (C6-C50) arylene-, - (C6-C50) arylene-C (O) -NH- (C6-C50) arylene-C (O) -NH-, - (C6-C50) arylene -P (O) 2-O-, - (C6-C50) arylene-O-P (O) 2-O-, - (C5-C50) heteroarylene-, - (C5-C50) heteroarylene-O-, -O- (C5-C50) heteroarylene-, - (C5-C50) heteroarylene-NH-, -NH- (C5-C50) heteroarylene-, -C (O) - (C5-C50) heteroarylene-, - (C5-C50) heteroarylene-C (O) -, -C (O) -O- (C5-C50) heteroarylene-, - (C5-C50) heteroarylene-C (O) -O-, -C (O) -NH- (C5-C50) heteroarylene-, -C (O) -NH- (C5-C50) heteroarylene-C (O) -O-, -C (O) -NH- (C5-C50) heteroarylene-O-, -C (O) -N ( (C1-C20) alkyl) - (C5-C50) heteroarylene-, -C (O) -N ( (C1-C20) alkyl) - (C5-C50) heteroarylene-O-, -C (O) -NH- (C5- C50) heteroarylene-C (O) -NH- (C3-C50) cycloalkylene-, -C (O) -NH- (C5-C50) heteroarylene-C (O) -NH-, -C (O) -N ( (C1-C20) alkyl) - (C5-C50) heteroarylene-C (O) -N ( (C1-C20) alkyl) - (C5-C50) heteroarylene-, -C (O) -N ( (C1-C20) alkyl) - (C5-C50) heteroarylene-C (O) -N ( (C1-C20) alkyl) -, - (C5-C50) heteroarylene-C (O) -NH- (C5-C50) heteroarylene-C (O) -NH- (C6-C50) arylene-, - (C5-C50) heteroarylene-C (O) -NH- (C5-C50) heteroarylene-C (O) -NH-, - (C5-C50) heteroarylene -P (O) 2-O-, - (C5-C50) heteroarylene-O-P (O) 2-O-; wherein each of R8′ and R9′ is either absent or is a substituent independently selected from the group consisting of -H, hydroxyl, - (C1-C30) alkyl, - (C3-C50) cycloalkyl, - (C6-C50) aryl, - (C1-C30) alkylene-OH, - (C3-C50) cycloalkylene-OH, - (C6-C50) arylene-OH, - (C1-C30) alkylene-C (O) OH, - (C3-C50) cycloalkylene-C (O) OH, - (C6-C50) arylene-C (O) OH, - (C1-C30) alkylene-NH2, - (C3-C50) cycloalkylene-NH2, - (C6-C50) arylene-NH2, - (C1-C30) alkoxy, - (C3-C50) cycloalkoxy, - (C6-C50) aryloxy, -C (O) - (C1-C30) alkyl, -OC (O) (C1-C30) alkyl, -C (O) -O- (C1-C30) alkyl, -C (O) - (C3-C50) cycloalkyl, -OC (O) - (C3-C50) cycloalkyl, -C (O) -O- (C3-C50) cycloalkyl, -C (O) - (C6-C50) aryloxy, -OC (O) - (C6-C50) aryloxy, -C (O) -O- (C6-C50) aryloxy, -C (O) -NH- (C1-C30) alkyl, -C (O) -NH- (C3-C50) cycloalkyl, -C (O) -NH- (C6-C50) aryl, - (C1-C30) alkylene-phosphoric acid, - (C3-C50) cycloalkylene-phosphoric acid, - (C6-C50) arylene-phosporic acid; wherein one or more hydroxyl group, carboxyl group, amino group and phosporic acid group contained in each of R8′ and R9′ are optionally protected with a terminal protective group; or R8′ and R9′ are linked together so that R8′, R9′, the carbon atom linked with R8′ and the Y′ atom linked with R9′ form a unsubstituted or substituted heterocyclic ring; with the proviso that R9′ is absent when Y′ is oxygen or sulfur;each R10′ is attached to any one of P′, Q′, S′ and T′, and is independently selected from the group consisting of hydroxyl, -C (O) OH, -P (O) 2-OH, -P (O) -OH, -P (O) (S) -OH, -CN, - (C1-C30) alkylene-OH, - (C3-C50) cycloalkylene-OH, - (C6-C50) arylene-OH, - (C5-C50) heteroarylene-OH, - (C1-C30) alkylene-C (O) OH, - (C3-C50) cycloalkylene-C (O) OH, - (C6-C50) arylene-C (O) OH, - (C5-C50) heteroarylene- C (O) OH, -C (O) -NH- (C1-C30) alkylene-OH, -C (O) -NH- (C3-C50) cycloalkylene-OH, -C (O) -NH- (C6-C50) arylene-OH, -C (O) -NH- (C5-C50) heteroarylene-OH, -C (O) -NH- (C1-C30) alkylene-C (O) OH, -C (O) -NH- (C3-C50) cycloalkylene-C (O) OH, -C (O) -NH- (C6-C50) arylene-C (O) OH, -C (O) -NH- (C5-C50) heteroarylene-C (O) OH, - (C1-C30) alkylene-C (O) -NH-(C1-C30) alkylene-C (O) OH, - (C1-C30) alkylene-O-C (O) - (C1-C30) alkylene-C (O) NH2, - (C1-C30) alkylene-O-C (O) - (C1-C30) alkylene-C (O) OH, - (C1-C30) alkylene-O-C (O) - (C1-C30) alkylene-NH2, - (C1-C30) alkylene-O-C (O) - (C1-C30) alkylene-OH, - (C1-C30) alkylene-C (O) -NH- (C3- C50) cycloalkylene-C (O) OH, - (C1-C30) alkylene-C (O) -NH- (C6-C50) arylene-C (O) OH, - (C1-C30) alkylene-C (O) -NH- (C5-C50) heteroarylene-C (O) OH, - (C1-C30) alkylene-P (O) 2-OH, - (C3-C50) cycloalkylene-P (O) 2-OH, - (C6-C50) arylene-P (O) 2-OH, - (C5-C50) heteroarylene-P (O) 2-OH, -C (O) -NH- (C1-C30) alkylene-O-P (-N (C1-C16alkyl) 2) -O- (C1-C30) alkylene-CN, -C (O) -NH- (C1-C30) alkylene-O-P (-N (C1-C16alkyl) 2) -O- (C1-C30) alkylene-OH, -C (O) -NH- (C1-C30) alkylene-O-P (-N (C1-C16alkyl) 2) -O- (C1-C30) alkylene-C (O) OH, -C (O) -NH- (C1-C30) alkylene-O-P (-N (C1-C16alkyl) 2) -O- (C1-C30) alkylene-NH2, - (C1-C30) alkylene-C (O) -NH- (C1-C30) alkylene-O-P (-N (C1-C16alkyl) 2) -O- (C1-C30) alkylene-CN, - (C1-C30) alkylene-C (O) -NH- (C1-C30) alkylene-O-P (-N (C1-C16alkyl) 2) -O- (C1-C30) alkylene-OH, - (C1-C30) alkylene-C (O) -NH- (C1-C30) alkylene-O-P (-N (C1-C16alkyl) 2) -O- (C1-C30) alkylene-C (O) OH, - (C1-C30) alkylene-C (O) -NH- (C1-C30) alkylene-O-P (-N (C1-C16alkyl) 2) -O- (C1-C30) alkylene-NH2, - (C1-C30) alkylene -P (O) -OH, - (C3-C50) cycloalkylene-P (O) -OH, - (C6-C50) arylene-P (O) -OH, - (C5-C50) -heteroarylene-P (O) -OH, - (C1-C30) alkylene-P (O) (S) -OH, - (C3-C50) cycloalkylene-P (O) (S) -OH, - (C6-C50) arylene-P (O) (S) -OH, - (C5-C50) heteroarylene-P (O) (S) -OH, - (C1-C30) alkylene-CN, - (C3-C50) cycloalkylene-CN, - (C6-C50) arylene-CN, - (C5-C50) heteroarylene-CN, wherein one or more hydroxyl group, carboxyl group, amino group, nitrile group and phosporic acid group contained in R10′ is optionally linked to a support material or protected with a terminal protective group;wherein each R11′ is attached to any one of P′, Q′, S′ and T′, and is either absent or selected from the group consisting of hydrogen, halogen atom, hydroxyl, (C1-C20) alkyl, (C1-C20) alkoxy, (C1-C20) alkoxycarbonyl, halogenated (C1-C20) alkyl and halogenated (C1-C20) alkoxycarbonyl; andwherein M′ is an integer of 1, 2 or 3, N′ is an integer of 1, 2 or 3, and M′+N′=4. - The oligonucleotide delivery enhancing compound according to claim 2 or 4, wherein one or more hydroxyl group, carboxyl group, amino group, nitrile group and phosphoric acid group contained in each of A1, A2, A3, B, C, R1, R2, R3, R4, R5, R6, R1′, R2′, R3′, R4′, R5′, R6′, R7′, R8′, R9′, R10′ and R11′ are optionally protected with a terminal protection group RP selected from the group consisting of (C1-C22) alkyl, (C1-C22) alkoxy, (C1-C22) alkylcarbonyl, (C1-C22) alkoxycarbonyl, (C6-C22) aryl, (C6-C22) aryloxy, (C6-C22) arylcarbonyl, (C6-C22) aryloxycarbonyl, glucosyl, acetamide glucosyl, galactosamine, N-acetyl galactosamine, tri ( (C1-C22) alkyl) silyl and tri ( (C1-C22) alkoxy) silyl; andwherein the support material is selected from the group consisting of silica, silica gel, glass, ceramic, polymer, cellulose, and combinations thereof.
- The oligonucleotide delivery enhancing compound according to claim 2 or 4, having a structure represented by any of Formula AIV to Formula AXIII and Formula BIV to BXIV,
wherein A1, A2, A3, A4, F, G, H, I, B, C, P, Q, S, T, R6, R7, m, n and M are as defined in claim 2,wherein each of RING I and RING II is a 4, 5, 6, 7, 8 or 9 member ring;wherein A4′ is attached to any atom of RING I, and each of A4′, A5′ and A6′ is independently selected from the group consisting of -R1′, -O-R1′, -S-R1′, -C (O) -R1′, -C (O) O-R1′, -O-C (O) -R1′, -C (O) NH-R1′, -C (O) NR2′-R1′, -NH-C (O) -R1′, -NR2′-C (O) -R1′, -O-P (O) 2-O-R1′, -OP (O) (S) -O-R1′, -O-P (O) -O-R1′, -NH-R1′, -NR2′-R1′, - (CH2) r′-NH-R1′, - (CH2) r′-NR2′-R1′, -C (O) - (CH2) r′-R1′, -C (O) - (CH2) r′-NH-R1′, -C (O) - (CH2) r′-NR2′-R1′, -C (O) - (CH2) r′-C (O) -R1′, -C (O) - (CH2) r′-C (O) O-R1′, -C (O) - (CH2) r′-NH-C (O) -R1′, -C (O) - (CH2) r′-NR2′-C (O) -R1′, - (CH2) r′-C (O) -R1′; - (CH2) r′-C (O) O-R1′; - (CH2) r′-O-C (O) -R1′, - (CH2) r′-R1′, - (CH2) r′-NH-C (O) -R1′, - (CH2) r′-NH-C (O) - (CH2) s′-R1′, - (CH2) r′-NR2′-C (O) - (CH2) s′-R1′, -CH (- (CH2) r′-NH-C (O) - (CH2) s′-R1′) (-NH-C (O) - (CH2) q′-R3′) , -N (- (CH2) r′-NH-C (O) - (CH2) s′-R1′) (-NH-C (O) - (CH2) q′-R3′) , -CH (- (CH2) r′-NH-C (O) - (CH2) s′-R1′) (- (CH2) p′-NH-C (O) - (CH2) q′-R3′) , -CR4′ (- (CH2) r′-NH-C (O) - (CH2) s′-R1′) (-NH-C (O) - (CH2) q′-R3′) , -CR4′ (- (CH2) r′-NH-C (O) - (CH2) s′-R1′) (- (CH2) p′-NH-C (O) - (CH2) q′-R3′) , -CH (- (CH2) r′-NR5′-C (O) - (CH2) s′-R1′) (-NR6′-C (O) - (CH2) q′-R3′) , -CH (- (CH2) r′-NR5′-C (O) - (CH2) s′-R1′) (- (CH2) p′-NR6′-C (O) - (CH2) q′-R3′) , -CR4′ ( (CH2) r′-NR5′-C (O) - (CH2) s′-R1′) ( (CH2) p′-NR6′-C (O) - (CH2) q′-R3′) , -CR4′ ( (CH2) r′-NR5′-C (O) - (CH2) s′-R1′) ( (CH2) p′-NR6′-C (O) - (CH2) q′-R3′) ; wherein each of R1′ and R3′ is independently selected from the group consisting of hydrogen, hydroxyl, - (C1-C30) alkyl, - (C3-C50) cycloalkyl, - (C6-C50) aryl, - (C1-C30) alkoxy, - (C3-C50) cycloalkoxy, - (C6-C50) aryloxy, -C (O) - (C1-C30) alkyl, -OC (O) (C1-C30) alkyl, -C (O) -O- (C1-C30) alkyl, -C (O) - (C3-C50) cycloalkyl, -OC (O) - (C3-C50) cycloalkyl, -C (O) -O- (C3-C50) cycloalkyl, -C (O) - (C6-C50) aryloxy, -OC (O) - (C6-C50) aryloxy, -C (O) -O- (C6-C50) aryloxy, -C (O) -phosphate ester group, phosphodiester group, phosphoramidite group, saturated fatty acid group, unsaturated fatty acid group, lipid, PEG, steroid, lipophile, carbohydrate, cholesterol, adamantane, amino acid, peptide, ligand, nucleic acid, oligonucleotide, aptamer, small molecule, antibody, antibody fragment, polyethylene glycol, carbohydrate, antibody, antibody fragment, chloroquine, alkaloid and targeting moiety, wherein one or more hydroxyl group, carboxyl group and amino group contained in each of R1′ and R3′ are optionally protected; wherein each of R2′, R4′, R5′ and R6′ is independently a halogen atom, a (C1-C12) alkyl, a (C1-C12) alkoxy, a (C6-C16) aryl or a (C6-C16) aryloxy; wherein each of r′, s′, p′ and q′ is an integer from 1 to 22; andwherein R12′ is attached to any atom of RING II and is selected from the group consisting of -H, hydroxyl, - (C1-C30) alkyl, - (C3-C50) cycloalkyl, - (C6-C50) aryl, - (C1-C30) alkylene-OH, - (C3-C50) cycloalkylene-OH, - (C6-C50) arylene-OH, - (C1-C30) alkylene-C (O) OH, - (C3-C50) cycloalkylene-C (O) OH, - (C6-C50) arylene-C (O) OH, - (C1-C30) alkylene-NH2, - (C3-C50) cycloalkylene-NH2, - (C6-C50) arylene-NH2, - (C1-C30) alkoxy, - (C3-C50) cycloalkoxy, - (C6-C50) aryloxy, -C (O) - (C1-C30) alkyl, -OC (O) (C1-C30) alkyl, -C (O) -O- (C1-C30) alkyl, -C (O) - (C3-C50) cycloalkyl, -OC (O) - (C3-C50) cycloalkyl, -C (O) -O- (C3-C50) cycloalkyl, -C (O) - (C6-C50) aryloxy, -OC (O) - (C6-C50) aryloxy, -C (O) -O- (C6-C50) aryloxy, -C (O) -NH- (C1-C30) alkyl, -C (O) -NH- (C3-C50) cycloalkyl, -C (O) -NH- (C6-C50) aryl, - (C1-C30) alkylene-phosporic acid, - (C3-C50) cycloalkylene-phosporic acid, - (C6-C50) arylene-phosporic acid; wherein one or more hydroxyl group, carboxyl group, amino group and phosporic acid group contained in R12′ are optionally protected. - The oligonucleotide delivery enhancing compound according to claim 2, wherein each of F, G, H and I is carbon, m is 1 and n is 3, B is attached to G or H, each of P, Q, S and T is carbon, R6 is attached to any one of Q and S;wherein the protection group RP is selected from the group consisting of benzyloxycarbonyl (Cbz) , tert-butyldimethylsilyl (TBS) , 4, 4′-dimethoxytrityl (DMTr) , t-butyloxy carbonyl (Boc) , benzyl (Bn) and benzyloxy (BnO) ;wherein C, on each occurrence, is selected from the group consisting of hydrogen, halogen atom, hydroxyl, (C1-C12) alkyl, (C1-C12) alkoxy, halogenated (C1-C12) alkyl and halogenated (C1-C12) alkoxy;wherein B, on each occurrence, is selected from the group consisting of - (C1-C22) alkylene-OH, -O-C (O) - (C1-C16) alkylene-C (O) NH2, - (C1-C16) alkylene-O-C (O) - (C1-C16) alkylene-C (O) NH2, -O-C (O) - (C1-C16) alkylene-C (O) OH, - (C1-C16) alkylene-O-C (O) - (C1-C16) alkylene-C (O) OH, -C (O) - (C1-C16) alkylene-C (O) NH2, - (C1-C16) alkylene-C (O) - (C1-C16) alkylene-C (O) NH2, -C (O) -NH- (C1-C16) alkylene-OH, -C (O) -NH- (C1-C16) alkylene-C (O) OH, - (C1-C16) alkylene-C (O) -NH- (C1-C16) alkylene-C (O) OH, - (C1-C16) alkylene-O-P (-N (C1-C16 alkyl) 2) -O- (C1-C16) alkylene-CN, - (C1-C16) alkylene-O-P (-N (C1-C16 alkyl) 2) -O- (C1-C16) alkylene-OH, - (C1-C16) alkylene-O-P (-N (C1-C16 alkyl) 2) -O- (C1-C16) alkylene-NH2, - (C1-C16) alkylene-O-P (-N (C1-C16 alkyl) 2) -O- (C1-C16) alkylene-C (O) OH, and -C (O) -NH- (C1-C16) alkylene-O-P (-N (C1-C16 alkyl) 2) -O- (C1-C16) alkylene-CN,wherein each of A1, A2 and A3 is either absent or a substituent independently selected from the group consisting of -H, -OH, linear or branched - (C6-C22) alkyl, linear or branched - (C2-C22) alkenyl, - (C1-C22) alkylene-OH, - (C3-C22) cycloalkyl, - (C3-C22) cycloalkenyl, - (C1-C22) alkylene- (C3-C22) cycloalkyl, - (C1-C22) alkylene-R1, - (C1-C22) alkylene-O-R1, - (C1-C22) alkylene-COOR1, -O- (C1-C22) alkyl, - (C6-C22) alkylene-adamantyl, - (C1-C22) alkylene-NH-C (O) - (C1-C22) alkyl, - (C1-C22) alkylene-NH-C (O) - (C1-C22) alkylene-adamantyl, - (C1-C22) alkylene-NR2-C (O) - (C1-C22) alkylene-adamantyl, - (C1-C22) alkylene- (C1-C6 alkylene oxide) (1-20) -NH-C (O) - (C1-C22) alkylene-adamantyl, -C (O) NH-R1, - (C1-C22) alkylene-O-P (-N (C1-C22 alkyl) 2) -O- (C1-C22) alkylene-CN, - (C1-C22) alkylene-O-P (-N (C1-C22 alkyl) 2) -O- (C1-C22) alkylene-OH, - (C1-C22) alkylene-O-P (-N (C1-C22 alkyl) 2) -O- (C1-C22) alkylene-NH2, - (C1-C22) alkylene-O-P (-N (C1-C22 alkyl) 2) -O- (C1-C22) alkylene-C (O) OH, -C (O) -NH- (C1-C22) alkylene-O-P (-N (C1-C22 alkyl) 2) -O- (C1-C22) alkylene-CN, substituted or unsubstituted pyrrole, substituted or unsubstituted pyrroline, substituted or unsubstituted pyrrolidine, substituted or unsubstituted pyrazole, substituted or unsubstituted pyrazoline, substituted or unsubstituted pyrazolidine, substituted or unsubstituted imidazole, substituted or unsubstituted oxazole, substituted or unsubstituted thiazole, substituted or unsubstituted benzopyrrole, substituted or unsubstituted benzopyrroline, substituted or unsubstituted benzopyrrolidine, substituted or unsubstituted benzopyrazole, substituted or unsubstituted benzopyrazoline, substituted or unsubstituted benzopyrazolidine, substituted or unsubstituted benzoimidazole, substituted or unsubstituted benzooxazole, substituted or unsubstituted benzothiazole, and a substituent represented by Formula AIII,wherein Y is selected from the group consisting of carbon, nitrogen, oxygen and sulfur; and each of P, Q, S and T is carbon;wherein each of R3, R4 and R5 is either absent or a substituent independently selected from the group consisting of -H, -OH, linear or branched - (C6-C22) alkyl, linear or branched - (C2-C22) alkenyl, - (C1-C22) alkylene-OH, - (C3-C22) cycloalkyl, - (C3-C22) cycloalkenyl, - (C1-C22) alkylene- (C3-C22) cycloalkyl, - (C1-C22) alkylene-R1, - (C1-C22) alkylene-O-R1, - (C1-C22) alkylene-COOR1, -O- (C1-C22) alkyl, - (C6-C22) alkylene-adamantyl, - (C1-C22) alkylene-NH-C (O) - (C1-C22) alkyl, - (C1-C22) alkylene-NH-C (O) - (C1-C22) alkylene-adamantyl, - (C1-C22) alkylene-NR2-C (O) - (C1-C22) alkylene-adamantyl, - (C1-C22) alkylene- (C1-C6 alkylene oxide) (1-20) -NH-C (O) - (C1-C22) alkylene-adamantyl, -C (O) NH-R1, - (C1-C22) alkylene-O-P (-N (C1-C22 alkyl) 2) -O- (C1-C22) alkylene-CN, - (C1-C22) alkylene-O-P (-N (C1-C22 alkyl) 2) -O- (C1-C22) alkylene-OH, - (C1-C22) alkylene-O-P (-N (C1-C22 alkyl) 2) -O- (C1-C22) alkylene-NH2, - (C1-C22) alkylene-O-P (-N (C1-C22 alkyl) 2) -O- (C1-C22) alkylene-C (O) OH, -C (O) -NH- (C1-C22) alkylene-O-P (-N (C1-C22 alkyl) 2) -O- (C1-C22) alkylene-CN, substituted or unsubstituted pyrrole, substituted or unsubstituted pyrroline, substituted or unsubstituted pyrrolidine, substituted or unsubstituted pyrazole, substituted or unsubstituted pyrazoline, substituted or unsubstituted pyrazolidine, substituted or unsubstituted imidazole, substituted or unsubstituted oxazole, substituted or unsubstituted thiazole, substituted or unsubstituted benzopyrrole, substituted or unsubstituted benzopyrroline, substituted or unsubstituted benzopyrrolidine, substituted or unsubstituted benzopyrazole, substituted or unsubstituted benzopyrazoline, substituted or unsubstituted benzopyrazolidine, substituted or unsubstituted benzoimidazole, substituted or unsubstituted benzooxazole, and substituted or unsubstituted benzothiazole,wherein R7, on each occurrence, is attached to any one of P, Q, S and T, and is either absent or selected from the group consisting of hydrogen, halogen atom, hydroxyl, (C1-C20) alkyl, (C1-C20) alkoxy, halogenated (C1-C20) alkyl and halogenated (C1-C20) alkoxy;wherein M is an integer of 0, 1, 2 or 3;wherein R6 is attached to any one of P, Q, S and T, and is selected from the group consisting of - (C1-C16) alkylene-, - (C1-C16) alkylene-O-, -O- (C1-C16) alkylene-, - (C1-C16) alkylene-NH-, -NH- (C1-C16) alkylene-, -C (O) - (C1-C16) alkylene-, - (C1-C16) alkylene-C (O) -, -C (O) -O- (C1-C16) alkylene-, - (C1-C16) alkylene-C (O) -O-, -C (O) -NH- (C1-C16) alkylene-, -C (O) -NH-(C1-C16) alkylene-C (O) -O-, -C (O) -NH- (C1-C16) alkylene-O-C (O) -, -C (O) -NH- (C1-C16) alkylene-O-C (O) -O-, -C (O) -NH- (C1-C16) alkylene-O-, -C (O) -N ( (C1-C16) alkyl) - (C1-C16) alkylene-, -C (O) -N ( (C1-C16) alkyl) - (C1-C16) alkylene-O-, -C (O) -NH- (C1-C16) alkylene-C (O) -NH- (C1-C16) alkylene-, -C (O) -NH- (C1-C16) alkylene-NH-C (O) - (C1-C16) alkylene-, -C (O) -NH- (C1-C16) alkylene-C (O) -NH-, -C (O) -N ( (C1-C16) alkyl) - (C1-C16) alkylene-C (O) -N ( (C1-C16) alkyl) - (C1-C16) alkylene-, -C (O) -N ( (C1-C16) alkyl) - (C1-C16) alkylene-C (O) -N ( (C1-C16) alkyl) -, - (C1-C16) alkylene-C (O) -NH- (C1-C16) alkylene-C (O) -NH- (C1-C16) alkylene-, - (C1-C16) alkylene-C (O) -NH- (C1-C16) alkylene-C (O) -NH-, -NH-C (O) - (C1-C16) alkylene-, -NH-C (O) - (C1-C16) alkylene-C (O) -O-, -NH-C (O) - (C1-C16) alkylene-C (O) -, -NH-C (O) - (C1-C16) alkylene-O-, -N ((C1-C16) alkyl) -C (O) - (C1-C16) alkylene-, -N ( (C1-C16) alkyl) -C (O) - (C1-C16) alkylene-O-, -NH-C (O) - (C1-C16) alkylene-NH-C (O) - (C1-C16) alkylene-, -NH-C (O) - (C1-C16) alkylene-NH-C (O) -, -N ((C1-C16) alkyl) -C (O) - (C1-C16) alkylene-N ( (C1-C16) alkyl) -C (O) - (C1-C16) alkylene-, -N ( (C1-C16) alkyl) -C (O) - (C1-C16) alkylene-N ( (C1-C16) alkyl) -C (O) -, - (C1-C16) alkylene-NH-C (O) - (C1-C16) alkylene-NH-C (O) - (C1-C16) alkylene-, - (C1-C16) alkylene-NH-C (O) - (C1-C16) alkylene-NH-C (O) -.
- The oligonucleotide delivery enhancing compound according to claim 4, having a structure represented by any one of Formula BXV to Formula BXXIX,
wherein each of A1′ and A2′ is a substituent independently selected from the group consisting of -R1′, -O-R1′, -S-R1′, -C (O) -R1′, -C (O) O-R1′, -O-C (O) -R1′, -C (O) NH-R1′, -C (O) NR2′-R1′, -NH-C (O) -R1′, -NR2′-C (O) -R1′, -O-P (O) 2-O-R1′, -OP (O) (S) -O-R1′, -O-P (O) -O-R1′, -NH-R1′, -NR2′-R1′, - (CH2) r′-NH-R1′, - (CH2) r′-NR2′-R1′, -C (O) - (CH2) r′-R1′, -C (O) - (CH2) r′-NH-R1′, -C (O) - (CH2) r′-NR2′-R1′, -C (O) - (CH2) r′-C (O) -R1′, -C (O) - (CH2) r′-C (O) O-R1′, -C (O) - (CH2) r′-NH-C (O) -R1′, -C (O) - (CH2) r′-NR2′-C (O) -R1′, - (CH2) r′-C (O) -R1′; - (CH2) r′-C (O) O-R1′; - (CH2) r′-O-C (O) -R1′, - (CH2) r′-R1′, - (CH2) r′-NH-C (O) -R1′, - (CH2) r′-NH-C (O) - (CH2) s′-R1′, - (CH2) r′-NH-C (O) - (C1-C22) alkylene-NH-C (O) - (CH2) s′-R1′, - (CH2) r′-C (O) -NH- (CH2) s′-R1′, - (CH2) r′-C (O) - NH- (C1-C22) alkylene-C (O) -NH- (CH2) s′-R1′, - (CH2) r′-C (O) -NH- (C1-C22) alkylene-NH-C (O) - (CH2) s′-R1′, - (CH2) r′-NR2′-C (O) - (CH2) s′-R1′, -CH (- (CH2) r′-NH-C (O) - (CH2) s′-R1′) (-NH-C (O) - (CH2) q′-R3′) , -CH (- (CH2) r′-C (O) -NH- (CH2) s′-R1′) (-C (O) -NH- (CH2) q′-R3′) , -N (- (CH2) r′-NH-C (O) - (CH2) s′-R1′) (-NH-C (O) - (CH2) q′-R3′) , -CH (- (CH2) r′-NH-C (O) - (CH2) s′-R1′) (- (CH2) p′-NH-C (O) - (CH2) q′-R3′) , -CR4′ (- (CH2) r′-NH-C (O) - (CH2) s′-R1′) (-NH-C (O) - (CH2) q′-R3′) , -CR4′ (- (CH2) r′-NH-C (O) - (CH2) s′-R1′) (- (CH2) p′-NH-C (O) - (CH2) q′-R3′) , -CH (- (CH2) r′-NR5′-C (O) - (CH2) s′-R1′) (-NR6′-C (O) - (CH2) q′-R3′) , -CH (- (CH2) r′-NR5′-C (O) - (CH2) s′-R1′) (- (CH2) p′-NR6′-C (O) - (CH2) q′-R3′) , -CR4′ ( (CH2) r′-NR5′-C (O) - (CH2) s′-R1′) ( (CH2) p′-NR6′-C (O) - (CH2) q′-R3′) , and -CR4′ ( (CH2) r′-NR5′-C (O) - (CH2) s′-R1′) ( (CH2) p′-NR6′-C (O) - (CH2) q′-R3′) ; wherein each of R1′ and R3′ is independently selected from the group consisting of hydrogen, hydroxyl, - (C1-C30) alkyl, - (C3-C50) cycloalkyl, - (C6-C50) aryl, - (C1-C30) alkoxy, - (C3-C50) cycloalkoxy, - (C6-C50) aryloxy, -C (O) - (C1-C30) alkyl, -OC (O) (C1-C30) alkyl, -C (O) -O- (C1-C30) alkyl, -C (O) - (C3-C50) cycloalkyl, -OC (O) - (C3-C50) cycloalkyl, -C (O) -O- (C3-C50) cycloalkyl, -C (O) - (C6-C50) aryloxy, -OC (O) - (C6-C50) aryloxy, -C (O) -O- (C6-C50) aryloxy, -C (O) -phosphate ester group, phosphodiester group, phosphoramidite group, saturated fatty acid group, unsaturated fatty acid group, lipid, PEG, steroid, lipophile, carbohydrate, cholesterol, adamantane, amino acid, peptide, ligand, nucleic acid, oligonucleotide, aptamer, small molecule, antibody, antibody fragment, polyethylene glycol, antibody, antibody fragment, chloroquine, alkaloid and targeting moiety, wherein one or more hydroxyl group, carboxyl group and amino group contained in each of R1′ and R3′ are optionally protected; wherein each of R2′, R4′, R5′ and R6′ is independently a halogen atom, a (C1-C12) alkyl, a (C1-C12) alkoxy, a (C1-C12) alkoxycarbonyl, a (C6-C16) aryl or a (C6-C16) aryloxycarbonyl; wherein each of r′, s′, p′ and q′ is an integer from 1 to 22;wherein m′ is an integer of 1, 2 or 3, n′ is an integer of 1, 2 or 3, and m′+n′=4;wherein each B′ is independently selected from the group consisting of hydroxyl, -C (O) OH, - (C1-C30) alkoxy, -P (O) 2-OH, -P (O) -OH, -P (O) (S) -OH, -CN, - (C1-C30) alkylene-OH, - (C3-C50) cycloalkylene-OH, - (C6-C50) arylene-OH, - (C5-C50) heteroarylene-OH, - (C1- C30) alkylene-C (O) OH, - (C3-C50) cycloalkylene-C (O) OH, - (C6-C50) arylene-C (O) OH, - (C5-C50) heteroarylene-C (O) OH, -C (O) -NH- (C1-C30) alkylene-OH, -C (O) -NH- [ (C1-C30) alkylene-O] r′-H (wherein r′ is an integer of 1 to 22) , -C (O) -NH- [ (C1-C30) alkylene-O] r′- (C1-C30) alkylene-C (O) -OH (wherein r′ is an integer of 1 to 22) , -C (O) -NH- (C3-C50) cycloalkylene-OH, -C (O) -NH-(C6-C50) arylene-OH, -C (O) -NH- (C5-C50) heteroarylene-OH, -C (O) -NH- (C1-C30) alkylene-C (O) OH, -C (O) -NH- (C3-C50) cycloalkylene-C (O) OH, -C (O) -NH- (C6-C50) arylene-C (O) OH, -C (O) -NH- (C1-C30) alkylene- (C6-C50) arylene- (C1-C30) alkylene-C (O) OH, -C (O) -NH- (C5-C50) heteroarylene-C (O) OH, - (C1-C30) alkylene-C (O) -NH- (C1-C30) alkylene-C (O) OH, - (C1-C30) alkylene-C (O) -NH- (C3-C50) cycloalkylene-C (O) OH, - (C1-C30) alkylene-C (O) -NH- (C6-C50) arylene-C (O) OH, - (C1-C30) alkylene-C (O) -NH- (C5-C50) heteroarylene-C (O) OH, - (C1-C30) alkylene-P (O) 2-OH, - (C3-C50) cycloalkylene-P (O) 2-OH, - (C6-C50) arylene-P (O) 2-OH, - (C5-C50) heteroarylene-P (O) 2-OH, -C (O) -NH- (C1-C30) alkylene-O-P (-N (C1-C16alkyl) 2) -O- (C1-C30) alkylene-CN, -C (O) -NH- (C1-C30) alkylene-O-P (-N (C1-C16alkyl) 2) -O- (C1-C30) alkylene-OH, -C (O) -NH- (C1-C30) alkylene-O-P (-N (C1-C16alkyl) 2) -O- (C1-C30) alkylene-C (O) OH, -C (O) -NH- (C1-C30) alkylene-O-P (-N (C1-C16alkyl) 2) -O- (C1-C30) alkylene-NH2, - (C1-C30) alkylene-C (O) -NH- (C1-C30) alkylene-O-P (-N (C1-C16alkyl) 2) -O- (C1-C30) alkylene-CN, - (C1-C30) alkylene-C (O) -NH- (C1-C30) alkylene-O-P (-N (C1-C16alkyl) 2) -O- (C1-C30) alkylene-OH, - (C1-C30) alkylene-C (O) -NH- (C1-C30) alkylene-O-P (-N (C1-C16alkyl) 2) -O- (C1-C30) alkylene-C (O) OH, - (C1-C30) alkylene-C (O) -NH- (C1-C30) alkylene-O-P (-N (C1-C16alkyl) 2) -O- (C1-C30) alkylene-NH2, - (C1-C30) alkylene -P (O) -OH, - (C3-C50) cycloalkylene-P (O) -OH, - (C6-C50) arylene-P (O) -OH, - (C5-C50) -heteroarylene-P (O) -OH, - (C1-C30) alkylene-P (O) (S) -OH, - (C3-C50) cycloalkylene-P (O) (S) -OH, - (C6-C50) arylene-P (O) (S) -OH, - (C5-C50) heteroarylene-P (O) (S) -OH, - (C1-C30) alkylene-CN, - (C3-C50) cycloalkylene-CN, - (C6-C50) arylene-CN, - (C5-C50) heteroarylene-CN, lipid, PEG, steroid, lipophile, carbohydrate, cholesterol, adamantane, amino acid, peptide, chloroquine and alkaloid;where each R7′ is selected from the group consisting of -O-, -C (O) O-, -O-C (O) -, -P (O) 2-O-, -O-P (O) 2-O-, -P (O) (S) -O-, -O-P (O) (S) -O-, -O-P (O) -O-, - (C1-C30) alkylene-, - (C1-C30) alkylene-O-, -O- (C1-C30) alkylene-, - (C1-C30) alkylene-NH-, -NH- (C1-C30) alkylene-, -C (O) - (C1-C30) alkylene-, - (C1-C30) alkylene-C (O) -, -C (O) -O- (C1-C30) alkylene-, - (C1-C30) alkylene-C (O) -O-, -C (O) -NH- (C1-C30) alkylene-, -C (O) -NH- (C1-C30) alkylene-C (O) -O-, -C (O) -NH- (C1-C30) alkylene-O-C (O) -, -C (O) -NH- (C1-C30) alkylene-O-C (O) -O-, -C (O) -NH- (C1-C30) alkylene- O-, -C (O) -N ( (C1-C20) alkyl) - (C1-C30) alkylene-, -C (O) -N ( (C1-C20) alkyl) - (C1-C30) alkylene-O-, -C (O) -NH- (C1-C30) alkylene-C (O) -NH- (C1-C30) alkylene-, -C (O) -NH- (C1-C30) alkylene-NH-C (O) - (C1-C30) alkylene-, -C (O) -NH- (C1-C30) alkylene-C (O) -NH-, -C (O) -N ( (C1-C20) alkyl) - (C1-C30) alkylene-C (O) -N ( (C1-C20) alkyl) - (C1-C30) alkylene-, -C (O) -N ( (C1-C20) alkyl) - (C1-C30) alkylene-C (O) -N ( (C1-C20) alkyl) -, - (C1-C30) alkylene-C (O) -NH- (C1-C30) alkylene-C (O) -NH- (C1-C30) alkylene-, - (C1-C30) alkylene-C (O) -NH- (C1-C30) alkylene-C (O) -NH-, -NH-C (O) - (C1-C30) alkylene-, -NH-C (O) - (C1-C30) alkylene-C (O) -O-, -NH-C (O) - (C1-C30) alkylene-C (O) -, -NH-C (O) - (C1-C30) alkylene-O-, -N ( (C1-C20) alkyl) -C (O) - (C1-C30) alkylene-, -N ( (C1-C20) alkyl) -C (O) - (C1-C30) alkylene-O-, -NH-C (O) - (C1-C30) alkylene-NH-C (O) - (C1-C30) alkylene-, -NH-C (O) - (C1-C30) alkylene-NH-C (O) -, -N ( (C1-C20) alkyl) -C (O) - (C1-C30) alkylene-N ( (C1-C20) alkyl) -C (O) - (C1-C30) alkylene-, -N ( (C1-C20) alkyl) -C (O) - (C1-C30) alkylene-N ( (C1-C20) alkyl) -C (O) -, - (C1-C30) alkylene-NH-C (O) - (C1-C30) alkylene-NH-C (O) - (C1-C30) alkylene-, - (C1-C30) alkylene-NH-C (O) - (C1-C30) alkylene-NH-C (O) -, - (C1-C30) alkylene -P (O) 2-O-, - (C1-C30) alkylene-O-P (O) 2-O-, - (C3-C50) cycloalkylene-, - (C3-C50) cycloalkylene-O-, -O- (C3-C50) cycloalkylene-, - (C3-C50) cycloalkylene-NH-, -NH- (C3-C50) cycloalkylene-, -C (O) - (C3-C50) cycloalkylene-, - (C3-C50) cycloalkylene-C (O) -, -C (O) -O- (C3-C50) cycloalkylene-, - (C3-C50) cycloalkylene-C (O) -O-, -C (O) -NH- (C3-C50) cycloalkylene-, -C (O) -NH- (C3-C50) cycloalkylene-C (O) -O-, -C (O) -NH- (C3-C50) cycloalkylene-O-, -C (O) -N ( (C1-C20) alkyl) - (C3-C50) cycloalkylene-, -C (O) -N ( (C1-C20) alkyl) - (C3-C50) cycloalkylene-O-, -C (O) -NH- (C3-C50) cycloalkylene-C (O) -NH- (C3-C50) cycloalkylene-, -C (O) -NH- (C3-C50) cycloalkylene-C (O) -NH-, -C (O) -N ( (C1-C20) alkyl) - (C3-C50) cycloalkylene-C (O) -N ( (C1-C20) alkyl) - (C3-C50) cycloalkylene-, -C (O) -N ( (C1-C20) alkyl) - (C3-C50) cycloalkylene-C (O) -N ( (C1-C20) alkyl) -, - (C3-C50) cycloalkylene-C (O) -NH- (C3-C50) cycloalkylene-C (O) -NH- (C3-C50) cycloalkylene-, - (C3-C50) cycloalkylene-C (O) -NH- (C3-C50) cycloalkylene-C (O) -NH-, - (C3-C50) cycloalkylene -P (O) 2-O-, - (C3-C50) cycloalkylene-O-P (O) 2-O-, - (C6-C50) arylene-, - (C6-C50) arylene-O-, -O- (C6-C50) arylene-, - (C6-C50) arylene-NH-, -NH- (C6-C50) arylene-, -C (O) - (C6-C50) arylene-, - (C6-C50) arylene-C (O) -, -C (O) -O- (C6-C50) arylene-, - (C6-C50) arylene-C (O) -O-, -C (O) -NH- (C6-C50) arylene-, -C (O) -NH- (C6-C50) arylene-C (O) -O-, -C (O) -NH- (C6-C50) arylene-O-, -C (O) -N ( (C1-C20) alkyl) - (C6-C50) arylene-, -C (O) -N ( (C1-C20) alkyl) - (C6-C50) arylene-O-, -C (O) -NH- (C6-C50) arylene-C (O) -NH- (C3-C50) cycloalkylene-, -C (O) -NH- (C6-C50) arylene-C (O) -NH-, -C (O) -N ( (C1-C20) alkyl) - (C6-C50) arylene-C (O) -N ( (C1-C20) alkyl) - (C6-C50) arylene-, -C (O) -N ( (C1-C20) alkyl) - (C6-C50) arylene- C (O) -N ( (C1-C20) alkyl) -, - (C6-C50) arylene-C (O) -NH- (C6-C50) arylene-C (O) -NH- (C6-C50) arylene-, - (C6-C50) arylene-C (O) -NH- (C6-C50) arylene-C (O) -NH-, - (C6-C50) arylene -P (O) 2-O-, - (C6-C50) arylene-O-P (O) 2-O-, - (C5-C50) heteroarylene-, - (C5-C50) heteroarylene-O-, -O- (C5-C50) heteroarylene-, - (C5-C50) heteroarylene-NH-, -NH- (C5-C50) heteroarylene-, -C (O) - (C5-C50) heteroarylene-, - (C5-C50) heteroarylene-C (O) -, -C (O) -O- (C5-C50) heteroarylene-, - (C5-C50) heteroarylene-C (O) -O-, -C (O) -NH- (C5-C50) heteroarylene-, -C (O) -NH- (C5-C50) heteroarylene-C (O) -O-, -C (O) -NH- (C5-C50) heteroarylene-O-, -C (O) -N ( (C1-C20) alkyl) - (C5-C50) heteroarylene-, -C (O) -N ( (C1-C20) alkyl) - (C5-C50) heteroarylene-O-, -C (O) -NH- (C5-C50) heteroarylene-C (O) -NH- (C3-C50) cycloalkylene-, -C (O) -NH- (C5-C50) heteroarylene-C (O) -NH-, -C (O) -N ( (C1-C20) alkyl) - (C5-C50) heteroarylene-C (O) -N ( (C1-C20) alkyl) - (C5-C50) heteroarylene-, -C (O) -N ( (C1-C20) alkyl) - (C5-C50) heteroarylene-C (O) -N ( (C1-C20) alkyl) -, - (C5-C50) heteroarylene-C (O) -NH- (C5-C50) heteroarylene-C (O) -NH- (C6-C50) arylene-, - (C5-C50) heteroarylene-C (O) -NH- (C5-C50) heteroarylene-C (O) -NH-, - (C5-C50) heteroarylene -P (O) 2-O-, - (C5-C50) heteroarylene-O-P (O) 2-O-; wherein each R8′ is a substituent independently selected from the group consisting of -H, hydroxyl, - (C1-C30) alkyl, - (C3-C50) cycloalkyl, - (C6-C50) aryl, - (C1-C30) alkylene-OH, - (C3-C50) cycloalkylene-OH, - (C6-C50) arylene-OH, - (C1-C30) alkylene-C (O) OH, - (C3-C50) cycloalkylene-C (O) OH, - (C6-C50) arylene-C (O) OH, - (C1-C30) alkylene-NH2, - (C3-C50) cycloalkylene-NH2, - (C6-C50) arylene-NH2, - (C1-C30) alkoxy, - (C3-C50) cycloalkoxy, - (C6-C50) aryloxy, -C (O) - (C1-C30) alkyl, -OC (O) (C1-C30) alkyl, -C (O) -O- (C1-C30) alkyl, -C (O) - (C3-C50) cycloalkyl, -OC (O) - (C3-C50) cycloalkyl, -C (O) -O- (C3-C50) cycloalkyl, -C (O) - (C6-C50) aryloxy, -OC (O) - (C6-C50) aryloxy, -C (O) -O- (C6-C50) aryloxy, -C (O) -NH- (C1-C30) alkyl, -C (O) -NH- (C3-C50) cycloalkyl, -C (O) -NH- (C6-C50) aryl, - (C1-C30) alkylene-phosphoric acid, - (C3-C50) cycloalkylene-phosphoric acid, - (C6-C50) arylene-phosporic acid; wherein one or more hydroxyl group, carboxyl group, amino group and phosporic acid group contained in each of R8′ are optionally protected with a terminal protective group; andwherein each R10′ is independently selected from the group consisting of hydroxyl, -C (O) OH, -P (O) 2-OH, -P (O) -OH, -P (O) (S) -OH, -CN, - (C1-C30) alkylene-OH, - (C3-C50) cycloalkylene-OH, - (C6-C50) arylene-OH, - (C5-C50) heteroarylene-OH, - (C1-C30) alkylene-C (O) OH, - (C3-C50) cycloalkylene-C (O) OH, - (C6-C50) arylene-C (O) OH, - (C5-C50) heteroarylene-C (O) OH, -C (O) -NH- (C1-C30) alkylene-OH, -C (O) -NH- (C3-C50) cycloalkylene-OH, -C (O) -NH- (C6-C50) arylene-OH, -C (O) -NH- (C5-C50) heteroarylene-OH, -C (O) -NH- (C1-C30) alkylene- C (O) OH, -C (O) -NH- (C3-C50) cycloalkylene-C (O) OH, -C (O) -NH- (C6-C50) arylene-C (O) OH, -C (O) -NH- (C5-C50) heteroarylene-C (O) OH, - (C1-C30) alkylene-C (O) -NH- (C1-C30) alkylene-C (O) OH, - (C1-C30) alkylene-O-C (O) - (C1-C30) alkylene-C (O) NH2, - (C1-C30) alkylene-O-C (O) - (C1-C30) alkylene-C (O) OH, - (C1-C30) alkylene-O-C (O) - (C1-C30) alkylene- NH2, - (C1-C30) alkylene-O-C (O) - (C1-C30) alkylene-OH, - (C1-C30) alkylene-C (O) -NH- (C3-C50) cycloalkylene-C (O) OH, - (C1-C30) alkylene-C (O) -NH- (C6-C50) arylene-C (O) OH, - (C1-C30) alkylene-C (O) -NH- (C5-C50) heteroarylene-C (O) OH, - (C1-C30) alkylene-P (O) 2-OH, - (C3-C50) cycloalkylene-P (O) 2-OH, - (C6-C50) arylene-P (O) 2-OH, - (C5-C50) heteroarylene-P (O) 2-OH, -C (O) -NH- (C1-C30) alkylene-O-P (-N (C1-C16alkyl) 2) -O- (C1-C30) alkylene-CN, -C (O) -NH- (C1-C30) alkylene-O-P (-N (C1-C16alkyl) 2) -O- (C1-C30) alkylene-OH, -C (O) -NH- (C1-C30) alkylene-O-P (-N (C1-C16alkyl) 2) -O- (C1-C30) alkylene-C (O) OH, -C (O) -NH- (C1-C30) alkylene-O-P (-N (C1-C16alkyl) 2) -O- (C1-C30) alkylene-NH2, - (C1-C30) alkylene-C (O) -NH- (C1-C30) alkylene-O-P (-N (C1-C16alkyl) 2) -O- (C1-C30) alkylene-CN, - (C1-C30) alkylene-C (O) -NH- (C1-C30) alkylene-O-P (-N (C1-C16alkyl) 2) -O- (C1-C30) alkylene-OH, - (C1-C30) alkylene-C (O) -NH- (C1-C30) alkylene-O-P (-N (C1-C16alkyl) 2) -O- (C1-C30) alkylene-C (O) OH, - (C1-C30) alkylene-C (O) -NH- (C1-C30) alkylene-O-P (-N (C1-C16alkyl) 2) -O- (C1-C30) alkylene-NH2, - (C1-C30) alkylene -P (O) -OH, - (C3-C50) cycloalkylene-P (O) -OH, - (C6-C50) arylene-P (O) -OH, - (C5-C50) -heteroarylene-P (O) -OH, - (C1-C30) alkylene-P (O) (S) -OH, - (C3-C50) cycloalkylene-P (O) (S) -OH, - (C6-C50) arylene-P (O) (S) -OH, - (C5-C50) heteroarylene-P (O) (S) -OH, - (C1-C30) alkylene-CN, - (C3-C50) cycloalkylene-CN, - (C6-C50) arylene-CN, - (C5-C50) heteroarylene-CN, wherein one or more hydroxyl group, carboxyl group, amino group, nitrile group and phosporic acid group contained in R10′ is optionally linked to a support material or protected with a terminal protective group. - The oligonucleotide delivery enhancing compound according to claim 2 or claim 4, having a structure of
wherein represents a support material. - The oligonucleotide delivery enhancing compound according to any of claims 1 to 9, wherein at least one hydrogen atom contained in the oligonucleotide delivery enhancing compound is substituted with deuterium atom.
- An oligonucleotide delivery agent, comprising a delivery enhancing compound (DEC) moiety derivable from the oligonucleotide delivery enhancing compound according to any of claims 1 to 10 and at least one oligonucleotide.
- The oligonucleotide delivery agent according to claim 11, wherein the oligonucleotide delivery enhancing compound moiety is linked with the oligonucleotide via at least one linking moiety selected from the group consisting of direct bond, -O-, -S-, -C (O) -, -NH-, -N ( (C1-C12) alkyl) -, -N ( (C1-C12) alkyl) -C (O) -O-, -O-C (O) -, -C (O) -O-, -O-C (O) -O-, -C (O) -NH-, -OP (O) 2O-, -P (O) (O-) O-, -OP (O) O-, -OP (O) (S) O-, -O-S (O) 2-O-, -S (O) 2-O-, -S (O) - O-, - (C1-C22) alkylene-, - (C1-C22) alkylene-NH-, -NH- (C1-C22) alkylene-, - (C1-C22) alkylene-NH-C (O) -, - (C1-C22) alkylene-C (O) -, - (C1-C22) alkylene-C (O) -O-, -C (O) - (C1-C22) alkylene-, -NH-C (O) - (C1-C22) alkylene-, -C (O) -NH- (C1-C22) alkylene-, -C (O) - (C1-C22) alkylene-NH-, -NH- (C1-C22) alkylene-C (O) -, -C (O) - (C1-C22) alkylene-C (O) -, -NH- (C1-C22) alkylene-NH-, -C (O) - (C1-C22) alkylene-C (O) O-, -O-C (O) - (C1-C22) alkylene-C (O) -O-, -C (O) -O- (C1-C22) alkylene-O-C (O) -, -C (O) - (C1-C22) alkylene-NH-C (O) -, -NH-C (O) - (C1-C22) alkylene-C (O) -, -NH-C (O) - (C1-C22) alkylene-C (O) -NH-, -C (O) -NH- (C1-C22) alkylene-NH-C (O) -, - (C1-C22) alkylene-OP (O) 2O-, - (C1-C22) alkylene-OP (O) (O-) O-, - (C1-C22) alkylene-OP (O) (O-) O- (C1-C22) alkylene-, - (C1-C22) alkylene-OP (O) O-, - (C1-C22) alkylene-OP (O) (S) O-, - (C1-C22) alkylene-O-S (O) 2-O-, - (C1-C22) alkylene-S (O) 2-O-, - (C1-C22) alkylene-S (O) -O-, -O-P (O) 2-O- (C1-C22) alkylene-OP (O) 2O-, -O-P (O) -O- (C1-C22) alkylene-OP (O) O-, -OP (O) (S) O- (C1-C22) alkylene-OP (O) (S) O-, -O-S (O) 2-O- (C1-C22) alkylene-O-S (O) 2-O-, -S (O) 2-O- (C1-C22) alkylene-S (O) 2-O- and -O-S (O) - (C1-C22) alkylene-S (O) -O-; andthe oligonucleotide is selected from the group consisting of antisense oligonucleotide (ASO) , antisense RNA, short interfering RNA (siRNA) , micro-RNA (miRNA) , small activating RNA (saRNA) , double-stranded RNA (dsRNA) , and small guide RNA (sgRNA) .
- The oligonucleotide delivery agent according to claim 11, wherein the oligonucleotide comprises at least part of the sequence as set forth in SEQ ID NO 1 to 53.
- The oligonucleotide delivery agent according to claim 11, comprising a structure represented by Formula AA
wherein the delivery enhancing compound (DEC) moiety is derived from the oligonucleotide delivery enhancing compound according to any one of claims 1 to 9 and is linked to at least one oligonucleotide directly or indirectly. - The oligonucleotide delivery agent according to claim 14, wherein the DEC is linked with the oligonucleotide via at least one first linking moiety.
- The oligonucleotide delivery agent according to claim 14, wherein the TM is linked with the DEC via at least one second linking moiety.
- The oligonucleotide delivery agent according to any one of claims 14-16, wherein each of the first linking moiety and the second linking moiety is independently selected from the group consisting of direct bond, -O-, -S-, -C (O) -, -NH-, -N ( (C1-C12) alkyl) -, -N ( (C1-C12) alkyl) -C (O) -O-, -O-C (O) -, -C (O) -O-, -O-C (O) -O-, -C (O) -NH-, -OP (O) 2O-, -OP (O) O-, -OP (O) (S) O-, -O-S (O) 2-O-, -S (O) 2-O-, -S (O) -O-, - (C1-C22) alkylene-, - (C1-C22) alkylene-NH-, -NH-(C1-C22) alkylene-, - (C1-C22) alkylene-NH-C (O) -, - (C1-C22) alkylene-C (O) -, - (C1-C22) alkylene-C (O) -O-, -C (O) - (C1-C22) alkylene-, -NH-C (O) - (C1-C22) alkylene-, -C (O) -NH- (C1-C22) alkylene-, -C (O) - (C1-C22) alkylene-NH-, -NH- (C1-C22) alkylene-C (O) -, -C (O) - (C1-C22) alkylene-C (O) -, -NH- (C1-C22) alkylene-NH-, -C (O) - (C1-C22) alkylene-C (O) O-, -O-C (O) - (C1-C22) alkylene-C (O) -O-, -C (O) -O- (C1-C22) alkylene-O-C (O) -, -C (O) - (C1-C22) alkylene-NH-C (O) -, -NH-C (O) - (C1-C22) alkylene-C (O) -, -NH-C (O) - (C1-C22) alkylene-C (O) -NH-, -NH- (C1-C22) alkylene-OP (O) 2O-, -NH- (C1-C22) alkylene-CH ( (C1-C22) alkylene-OH) -OP (O) 2O-, -NH- (C1-C22) alkylene-CH ( (C1-C22) alkylene-OH) - (C1-C22) alkylene-OP (O) 2O-, -C (O) -NH- (C1-C22) alkylene-NH-C (O) -, - (C1-C22) alkylene-OP (O) 2O-, - (C1-C22) alkylene-OP (O) O-, - (C1-C22) alkylene-OP (O) (S) O-, - (C1-C22) alkylene-O-S (O) 2-O-, - (C1-C22) alkylene-S (O) 2-O-, - (C1-C22) alkylene-S (O) -O-, -O-P (O) 2-O- (C1-C22) alkylene-OP (O) 2O-, -O-P (O) -O- (C1-C22) alkylene-OP (O) O-, -OP (O) (S) O- (C1-C22) alkylene-OP (O) (S) O-, -O-S (O) 2-O- (C1-C22) alkylene-O-S (O) 2-O-, -S (O) 2-O- (C1-C22) alkylene-S (O) 2-O-and -O-S (O) - (C1-C22) alkylene-S (O) -O-; and/orwherein the oligonucleotide is selected from the group consisting of short interfering RNA (siRNA) , small activating RNA (saRNA) , microRNA (miRNA) , antisense oligonucleotide (ASO) and small guide RNA (sgRNA) .
- The oligonucleotide delivery agent according to any of claims 14-16, the targeting moiety is one or more selected from the group consisting of ligands, peptides, nucleic acids, oligonucleotides, aptamers, lipids, fatty acids, small molecules, polyethylene glycols, amino acids, cholesterols, carbohydrates, and antibodies or antibody fragments.
- The oligonucleotide delivery agent according to claim 13, having a structure represented by any of the formulae AAI to AAXXIV:
wherein L represents the linking moiety, represents the oligonucleotide delivery enhancing compound, the symbolepresents a double strand oligonucleotide in which each of the strands represents interchangeably a sense strand or an antisense strand, either symmetric or asymmetric independently on each of the ends; the symbolrepresents a single strand oligonucleotide, and each of a, b and c is independently an integer from 1 to 50. - The oligonucleotide delivery agent according to any of claims 11 to 19, wherein at least one hydrogen atom contained in the delivery enhancing compound moiety, the linking moiety, the targeting moiety and/or the oligonucleotide is substituted with deuterium atom.
- A pharmaceutical composition, the composition comprising:a) the oligonucleotide delivery agent according to any of claims 11 to 20; andb) optionally, one or more ingredients selected from the group consisting of pharmaceutically acceptable carrier, excipient, solvent, diluent, stabilizer, dispersant, buffer, compatibilizer, preservative agent and combinations thereof.
- A method of modulating the expression of a target gene in a subject, the method comprising the step of administrating the pharmaceutical composition according to claim 21 to a subject.
- The method of claim 22, wherein the oligonucleotide or the target gene comprises at least part of the sequence as set forth in at least part of the sequence as set forth in SEQ ID NO 1 to 53.
- The method of claim 22, wherein the pharmaceutical composition increases the expression of the target gene.
- The method of claim 22, wherein the pharmaceutical composition decreases the expression of the target gene.
- The method of any of claims 22-24, wherein the subject is a mammal.
- The method of claim 26, wherein the mammal is a rodent.
- The method of claim 27, wherein the rodent is a mouse.
- The method of claim 27, wherein the rodent is a rat.
- The method of claim 26, wherein the mammal is a non-human primate.
- The method of claim 26, wherein the mammal is a human.
- The method of any of claims 22-24, wherein the target gene is associated with a disease or disorder.
- The method of claim 32, wherein the target gene is associated with a disease or disorder in the central nervous system (CNS) , brain, spinal cord, liver, lung, kidney, intestine, pancreas, cholecyst, heart, lymph nodes, spleen, stomach, bladder, muscle or bone.
- The method of claim 33, wherein the disease is cancer.
- A method of modulating the expression of a target gene, the method comprising contacting a cell with the pharmaceutical composition of claim 21.
- The method of claim 35, wherein the oligonucleotide or the target gene comprises at least part of the sequence as set forth in at least part of the sequence as set forth in SEQ ID NO: 1 to 53.
- The method of claim 35, wherein the pharmaceutical composition increases the expression of the target gene.
- The method of claim 35, wherein the pharmaceutical composition decreases the expression of the target gene.
- The method of claim 35, wherein the cell is a mammalian cell.
- The method of claim 39, wherein the mammalian cell is a mouse cell.
- The method of claim 39, wherein the mammalian cell is a rat cell.
- The method of claim 39, wherein the mammalian cell is a non-human primate cell.
- The method of claim 39, wherein the mammalian cell is a human cell.
- The method of any of claims 35-43, wherein the target gene is associated with a disease or disorder.
- The method of claim 44, wherein the target gene is associated with a disease or disorder in the central nervous system (CNS) , brain, spinal cord, liver, lung, kidney, intestine, pancreas, cholecyst, heart, lymph nodes, spleen, stomach, bladder, muscle or bone.
- The method of claim 44, wherein the disease is cancer.
Applications Claiming Priority (4)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
CN2022101636 | 2022-06-27 | ||
CNPCT/CN2022/101636 | 2022-06-27 | ||
CN2022111702 | 2022-08-11 | ||
CNPCT/CN2022/111702 | 2022-08-11 |
Publications (1)
Publication Number | Publication Date |
---|---|
WO2024002046A1 true WO2024002046A1 (en) | 2024-01-04 |
Family
ID=89382932
Family Applications (2)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
PCT/CN2023/072927 WO2024001172A1 (en) | 2022-06-27 | 2023-01-18 | Oligonucleotide modulators activating complement factor h expression |
PCT/CN2023/102623 WO2024002046A1 (en) | 2022-06-27 | 2023-06-27 | Oligonucleotide delivery enhancing compounds, pharmaceutical compositions and methods using the same |
Family Applications Before (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
PCT/CN2023/072927 WO2024001172A1 (en) | 2022-06-27 | 2023-01-18 | Oligonucleotide modulators activating complement factor h expression |
Country Status (2)
Country | Link |
---|---|
TW (1) | TW202400190A (en) |
WO (2) | WO2024001172A1 (en) |
Cited By (1)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
CN118290282A (en) * | 2024-03-20 | 2024-07-05 | 北京剂泰医药科技有限公司 | Ionizable lipid compound, preparation method and application thereof |
Citations (4)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
CN105392488A (en) * | 2013-05-01 | 2016-03-09 | Isis制药公司 | Compositions and methods for modulating apolipoprotein c-iii expression |
US20160376585A1 (en) * | 2013-07-11 | 2016-12-29 | Alnylam Pharmaceuticals, Inc. | Oligonucleotide-ligand conjugates and process for their preparation |
CN106459969A (en) * | 2014-05-01 | 2017-02-22 | Ionis制药公司 | Compositions and methods for modulating growth hormone receptor expression |
CN112400018A (en) * | 2018-05-07 | 2021-02-23 | 阿尔尼拉姆医药品有限公司 | Extrahepatic delivery |
Family Cites Families (6)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
EP2021499A4 (en) * | 2005-12-16 | 2010-02-17 | Univ Leland Stanford Junior | Functional arrays for high throughput characterization of gene expression regulatory elements |
US20180305689A1 (en) * | 2015-04-22 | 2018-10-25 | Mina Therapeutics Limited | Sarna compositions and methods of use |
US11162099B2 (en) * | 2017-09-08 | 2021-11-02 | Mina Therapeutics Limited | HNF4A saRNA compositions and methods of use |
US20200208152A1 (en) * | 2017-09-08 | 2020-07-02 | Mina Therapeutics Limited | Stabilized sarna compositions and methods of use |
KR20200140377A (en) * | 2018-04-10 | 2020-12-15 | 락티젠 세러퓨틱스 | New small active RNA |
WO2021026476A1 (en) * | 2019-08-08 | 2021-02-11 | Mpeg La, L.L.C. | Complement targeting with multimeric oligonucleotides |
-
2023
- 2023-01-18 WO PCT/CN2023/072927 patent/WO2024001172A1/en unknown
- 2023-03-13 TW TW112109234A patent/TW202400190A/en unknown
- 2023-06-27 WO PCT/CN2023/102623 patent/WO2024002046A1/en unknown
Patent Citations (4)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
CN105392488A (en) * | 2013-05-01 | 2016-03-09 | Isis制药公司 | Compositions and methods for modulating apolipoprotein c-iii expression |
US20160376585A1 (en) * | 2013-07-11 | 2016-12-29 | Alnylam Pharmaceuticals, Inc. | Oligonucleotide-ligand conjugates and process for their preparation |
CN106459969A (en) * | 2014-05-01 | 2017-02-22 | Ionis制药公司 | Compositions and methods for modulating growth hormone receptor expression |
CN112400018A (en) * | 2018-05-07 | 2021-02-23 | 阿尔尼拉姆医药品有限公司 | Extrahepatic delivery |
Cited By (1)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
CN118290282A (en) * | 2024-03-20 | 2024-07-05 | 北京剂泰医药科技有限公司 | Ionizable lipid compound, preparation method and application thereof |
Also Published As
Publication number | Publication date |
---|---|
WO2024001172A1 (en) | 2024-01-04 |
TW202400190A (en) | 2024-01-01 |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
US20220195429A1 (en) | Oligonucleotide compositions and methods thereof | |
US10724035B2 (en) | Oligonucleotide compositions and methods thereof | |
AU2016334232B2 (en) | Oligonucleotide compositions and methods thereof | |
EP3010514B1 (en) | Double-stranded antisense nucleic acid with exon-skipping effect | |
AU2016280709B2 (en) | Defined multi-conjugate oligonucleotides | |
WO2021234459A9 (en) | Double stranded oligonucleotide compositions and methods relating thereto | |
JP2022519019A (en) | Oligonucleotide composition and its method | |
EP4022059A1 (en) | Oligonucleotide compositions and methods of use thereof | |
KR102713731B1 (en) | Novel compounds and their uses | |
JP6808710B2 (en) | A composition that stably contains a nucleic acid molecule | |
KR20100106314A (en) | Lipid-modified stranded rna having potent rna interference effect | |
US20230277675A1 (en) | Systemic delivery of oligonucleotides | |
KR20170098914A (en) | Native miRNAs for gene expression control and uses thereof | |
WO2024002046A1 (en) | Oligonucleotide delivery enhancing compounds, pharmaceutical compositions and methods using the same | |
JP2023509870A (en) | Pharmaceutical combinations of therapeutic oligonucleotides targeting HBV and TLR7 agonists for the treatment of HBV | |
JP2024505035A (en) | Compositions and methods for inhibiting gene expression in the central nervous system | |
AU2020398192A1 (en) | Peptide docking vehicle for targeted nucleic acid delivery | |
JP2023509872A (en) | Pharmaceutical combinations of antiviral agents targeting HBV and/or immunomodulatory agents for the treatment of HBV | |
TW202406576A (en) | Oligonucleotide delivery enhancing compounds, pharmaceutical compositions and methods using the same | |
WO2024002045A1 (en) | Oligonucleotide delivery agents, pharmaceutical compositions and methods using the same | |
US20240026358A1 (en) | Oligonucleotide compositions and methods thereof | |
US20230392137A1 (en) | Oligonucleotide compositions and methods thereof | |
US20230220390A1 (en) | Nucleic acid molecule having improved stability, and use thereof | |
RU2797833C1 (en) | Oligonucleotide compositions and methods related to them | |
TW202417049A (en) | Oligonucleotide delivery agents, pharmaceutical compositions and methods using the same |
Legal Events
Date | Code | Title | Description |
---|---|---|---|
121 | Ep: the epo has been informed by wipo that ep was designated in this application |
Ref document number: 23830223 Country of ref document: EP Kind code of ref document: A1 |