IL296106A - Compositions and methods for inhibiting expression of transthyretin (ttr) - Google Patents
Compositions and methods for inhibiting expression of transthyretin (ttr)Info
- Publication number
- IL296106A IL296106A IL296106A IL29610622A IL296106A IL 296106 A IL296106 A IL 296106A IL 296106 A IL296106 A IL 296106A IL 29610622 A IL29610622 A IL 29610622A IL 296106 A IL296106 A IL 296106A
- Authority
- IL
- Israel
- Prior art keywords
- ttr
- rnai agent
- fixed dose
- double stranded
- phosphate
- Prior art date
Links
- 238000000034 method Methods 0.000 title claims description 110
- 230000014509 gene expression Effects 0.000 title claims description 57
- 230000002401 inhibitory effect Effects 0.000 title claims description 13
- 108010071690 Prealbumin Proteins 0.000 title description 163
- 239000000203 mixture Substances 0.000 title description 12
- 102000009190 Transthyretin Human genes 0.000 title 1
- 239000003795 chemical substances by application Substances 0.000 claims description 227
- 102100029290 Transthyretin Human genes 0.000 claims description 201
- 230000009368 gene silencing by RNA Effects 0.000 claims description 196
- 125000003729 nucleotide group Chemical group 0.000 claims description 98
- 239000002773 nucleotide Substances 0.000 claims description 87
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 claims description 82
- 201000010099 disease Diseases 0.000 claims description 76
- 230000000692 anti-sense effect Effects 0.000 claims description 61
- 206010002022 amyloidosis Diseases 0.000 claims description 50
- 239000003446 ligand Substances 0.000 claims description 44
- 101000772194 Homo sapiens Transthyretin Proteins 0.000 claims description 43
- 108091081021 Sense strand Proteins 0.000 claims description 42
- 101150091380 TTR gene Proteins 0.000 claims description 41
- 230000008859 change Effects 0.000 claims description 38
- 108090000623 proteins and genes Proteins 0.000 claims description 33
- 108020004999 messenger RNA Proteins 0.000 claims description 31
- 102000004169 proteins and genes Human genes 0.000 claims description 29
- 102000039446 nucleic acids Human genes 0.000 claims description 24
- 108020004707 nucleic acids Proteins 0.000 claims description 24
- 201000001119 neuropathy Diseases 0.000 claims description 21
- 230000007823 neuropathy Effects 0.000 claims description 21
- 208000033808 peripheral neuropathy Diseases 0.000 claims description 21
- -1 thymidine-glycol nucleic acid Chemical class 0.000 claims description 21
- OVRNDRQMDRJTHS-KEWYIRBNSA-N N-acetyl-D-galactosamine Chemical class CC(=O)N[C@H]1C(O)O[C@H](CO)[C@H](O)[C@@H]1O OVRNDRQMDRJTHS-KEWYIRBNSA-N 0.000 claims description 19
- 208000028389 Nerve injury Diseases 0.000 claims description 19
- 230000008764 nerve damage Effects 0.000 claims description 19
- 208000024891 symptom Diseases 0.000 claims description 19
- 230000006735 deficit Effects 0.000 claims description 18
- 229910052760 oxygen Inorganic materials 0.000 claims description 17
- 238000012360 testing method Methods 0.000 claims description 17
- 238000007920 subcutaneous administration Methods 0.000 claims description 14
- 208000031229 Cardiomyopathies Diseases 0.000 claims description 12
- 206010035226 Plasma cell myeloma Diseases 0.000 claims description 12
- 208000022256 primary systemic amyloidosis Diseases 0.000 claims description 12
- 229910052717 sulfur Inorganic materials 0.000 claims description 12
- 230000009885 systemic effect Effects 0.000 claims description 11
- 208000012902 Nervous system disease Diseases 0.000 claims description 10
- 101000979333 Homo sapiens Neurofilament light polypeptide Proteins 0.000 claims description 8
- 102100023057 Neurofilament light polypeptide Human genes 0.000 claims description 8
- 206010036105 Polyneuropathy Diseases 0.000 claims description 8
- NPRNBAPBGVERRF-ZOQUXTDFSA-N [(2r,3r,4r,5r)-5-(2,4-dioxopyrimidin-1-yl)-2-(hydroxymethyl)-4-methoxyoxolan-3-yl] dihydrogen phosphate Chemical compound CO[C@@H]1[C@H](OP(O)(O)=O)[C@@H](CO)O[C@H]1N1C(=O)NC(=O)C=C1 NPRNBAPBGVERRF-ZOQUXTDFSA-N 0.000 claims description 8
- IATVSXSQCCSKNS-ZOQUXTDFSA-N [(2r,3r,4r,5r)-5-(4-amino-2-oxopyrimidin-1-yl)-2-(hydroxymethyl)-4-methoxyoxolan-3-yl] dihydrogen phosphate Chemical compound CO[C@@H]1[C@H](OP(O)(O)=O)[C@@H](CO)O[C@H]1N1C(=O)N=C(N)C=C1 IATVSXSQCCSKNS-ZOQUXTDFSA-N 0.000 claims description 8
- VXTNWQOOBHQEDS-IOSLPCCCSA-N [(2r,3r,4r,5r)-5-(6-aminopurin-9-yl)-2-(hydroxymethyl)-4-methoxyoxolan-3-yl] dihydrogen phosphate Chemical compound CO[C@@H]1[C@H](OP(O)(O)=O)[C@@H](CO)O[C@H]1N1C2=NC=NC(N)=C2N=C1 VXTNWQOOBHQEDS-IOSLPCCCSA-N 0.000 claims description 8
- SQXGRYKQXLPVFG-AEHJODJJSA-N [(2r,3r,4s,5r)-5-(2-amino-6-oxo-3h-purin-9-yl)-4-fluoro-4-hydroxy-2-(hydroxymethyl)oxolan-3-yl] dihydrogen phosphate Chemical compound C1=NC=2C(=O)NC(N)=NC=2N1[C@@H]1O[C@H](CO)[C@@H](OP(O)(O)=O)[C@]1(O)F SQXGRYKQXLPVFG-AEHJODJJSA-N 0.000 claims description 8
- PTESHFKYBQMNFR-FJGDRVTGSA-N [(2r,3r,4s,5r)-5-(4-amino-2-oxopyrimidin-1-yl)-4-fluoro-4-hydroxy-2-(hydroxymethyl)oxolan-3-yl] dihydrogen phosphate Chemical compound O=C1N=C(N)C=CN1[C@H]1[C@](F)(O)[C@H](OP(O)(O)=O)[C@@H](CO)O1 PTESHFKYBQMNFR-FJGDRVTGSA-N 0.000 claims description 8
- KLVQSNKMLGBSHV-GIWSHQQXSA-N [(2r,3r,4s,5r)-5-(6-aminopurin-9-yl)-4-fluoro-4-hydroxy-2-(hydroxymethyl)oxolan-3-yl] dihydrogen phosphate Chemical compound C1=NC=2C(N)=NC=NC=2N1[C@@H]1O[C@H](CO)[C@@H](OP(O)(O)=O)[C@]1(O)F KLVQSNKMLGBSHV-GIWSHQQXSA-N 0.000 claims description 8
- 230000007824 polyneuropathy Effects 0.000 claims description 8
- 206010007509 Cardiac amyloidosis Diseases 0.000 claims description 7
- QXWJIJPOAPZARX-KQYNXXCUSA-N [(2r,3r,4r,5r)-5-(2-amino-6-oxo-3h-purin-9-yl)-2-(hydroxymethyl)-4-methoxyoxolan-3-yl] dihydrogen phosphate Chemical compound CO[C@@H]1[C@H](OP(O)(O)=O)[C@@H](CO)O[C@H]1N1C(N=C(N)NC2=O)=C2N=C1 QXWJIJPOAPZARX-KQYNXXCUSA-N 0.000 claims description 7
- 238000011161 development Methods 0.000 claims description 7
- 230000002526 effect on cardiovascular system Effects 0.000 claims description 7
- 101800000407 Brain natriuretic peptide 32 Proteins 0.000 claims description 6
- 206010016202 Familial Amyloidosis Diseases 0.000 claims description 6
- 230000036541 health Effects 0.000 claims description 6
- 238000001990 intravenous administration Methods 0.000 claims description 6
- 230000001404 mediated effect Effects 0.000 claims description 6
- 102100023708 Coiled-coil domain-containing protein 80 Human genes 0.000 claims description 5
- 101000978383 Homo sapiens Coiled-coil domain-containing protein 80 Proteins 0.000 claims description 5
- 101000679907 Homo sapiens Tumor necrosis factor receptor superfamily member 27 Proteins 0.000 claims description 5
- 102100022202 Tumor necrosis factor receptor superfamily member 27 Human genes 0.000 claims description 5
- 239000002131 composite material Substances 0.000 claims description 5
- 239000003814 drug Substances 0.000 claims description 5
- 201000003968 hyperthyroxinemia Diseases 0.000 claims description 5
- 230000002861 ventricular Effects 0.000 claims description 5
- 206010064571 Gene mutation Diseases 0.000 claims description 4
- 101000825960 Homo sapiens R-spondin-3 Proteins 0.000 claims description 4
- 102100022766 R-spondin-3 Human genes 0.000 claims description 4
- QGZHQRNBDZDKFW-FJGDRVTGSA-N [(2r,3r,4s,5r)-5-(2,4-dioxopyrimidin-1-yl)-4-fluoro-4-hydroxy-2-(hydroxymethyl)oxolan-3-yl] dihydrogen phosphate Chemical compound O[C@]1(F)[C@H](OP(O)(O)=O)[C@@H](CO)O[C@H]1N1C(=O)NC(=O)C=C1 QGZHQRNBDZDKFW-FJGDRVTGSA-N 0.000 claims description 4
- 230000002567 autonomic effect Effects 0.000 claims description 4
- 230000000926 neurological effect Effects 0.000 claims description 4
- 229940071643 prefilled syringe Drugs 0.000 claims description 4
- 108090000751 Ceramidases Proteins 0.000 claims description 3
- 102100023996 Neutral ceramidase Human genes 0.000 claims description 3
- 229940090047 auto-injector Drugs 0.000 claims description 3
- 229940124597 therapeutic agent Drugs 0.000 claims description 3
- 208000027121 wild type ATTR amyloidosis Diseases 0.000 claims description 3
- 108091030071 RNAI Proteins 0.000 claims 32
- 210000003169 central nervous system Anatomy 0.000 claims 2
- 108010017384 Blood Proteins Proteins 0.000 claims 1
- 102000004506 Blood Proteins Human genes 0.000 claims 1
- 208000032131 Diabetic Neuropathies Diseases 0.000 claims 1
- 102400001263 NT-proBNP Human genes 0.000 claims 1
- 230000003862 health status Effects 0.000 claims 1
- 239000000041 non-steroidal anti-inflammatory agent Substances 0.000 claims 1
- 229940021182 non-steroidal anti-inflammatory drug Drugs 0.000 claims 1
- 230000036470 plasma concentration Effects 0.000 claims 1
- 108010008064 pro-brain natriuretic peptide (1-76) Proteins 0.000 claims 1
- 239000003381 stabilizer Substances 0.000 claims 1
- 238000012228 RNA interference-mediated gene silencing Methods 0.000 description 164
- 238000011282 treatment Methods 0.000 description 70
- 125000005647 linker group Chemical group 0.000 description 68
- 108091032973 (ribonucleotides)n+m Proteins 0.000 description 63
- 102000040650 (ribonucleotides)n+m Human genes 0.000 description 45
- 210000004027 cell Anatomy 0.000 description 42
- 208000034846 Familial Amyloid Neuropathies Diseases 0.000 description 37
- 201000007905 transthyretin amyloidosis Diseases 0.000 description 36
- 230000007423 decrease Effects 0.000 description 32
- 230000004048 modification Effects 0.000 description 27
- 238000012986 modification Methods 0.000 description 27
- 206010019889 Hereditary neuropathic amyloidosis Diseases 0.000 description 25
- 230000035772 mutation Effects 0.000 description 24
- 235000000346 sugar Nutrition 0.000 description 23
- 238000003776 cleavage reaction Methods 0.000 description 22
- 230000007017 scission Effects 0.000 description 22
- 210000002966 serum Anatomy 0.000 description 22
- 150000001875 compounds Chemical class 0.000 description 21
- 239000000090 biomarker Substances 0.000 description 20
- 230000000295 complement effect Effects 0.000 description 20
- 230000000747 cardiac effect Effects 0.000 description 19
- 210000003205 muscle Anatomy 0.000 description 19
- 230000005764 inhibitory process Effects 0.000 description 18
- 239000002253 acid Substances 0.000 description 17
- 210000004369 blood Anatomy 0.000 description 17
- 239000008280 blood Substances 0.000 description 17
- 230000006872 improvement Effects 0.000 description 17
- 230000003247 decreasing effect Effects 0.000 description 15
- 239000000902 placebo Substances 0.000 description 15
- 229940068196 placebo Drugs 0.000 description 15
- 239000008194 pharmaceutical composition Substances 0.000 description 14
- 230000009467 reduction Effects 0.000 description 14
- 108091034117 Oligonucleotide Proteins 0.000 description 13
- 208000027697 autoimmune lymphoproliferative syndrome due to CTLA4 haploinsuffiency Diseases 0.000 description 12
- 235000014633 carbohydrates Nutrition 0.000 description 12
- 150000007523 nucleic acids Chemical class 0.000 description 12
- 206010061818 Disease progression Diseases 0.000 description 11
- 150000001720 carbohydrates Chemical class 0.000 description 11
- 230000005750 disease progression Effects 0.000 description 11
- 230000003278 mimic effect Effects 0.000 description 11
- 108090000765 processed proteins & peptides Proteins 0.000 description 11
- 230000011514 reflex Effects 0.000 description 11
- 208000037259 Amyloid Plaque Diseases 0.000 description 10
- OPTASPLRGRRNAP-UHFFFAOYSA-N cytosine Chemical compound NC=1C=CNC(=O)N=1 OPTASPLRGRRNAP-UHFFFAOYSA-N 0.000 description 10
- 230000000977 initiatory effect Effects 0.000 description 10
- 125000000325 methylidene group Chemical group [H]C([H])=* 0.000 description 10
- XUIIKFGFIJCVMT-GFCCVEGCSA-N D-thyroxine Chemical compound IC1=CC(C[C@@H](N)C(O)=O)=CC(I)=C1OC1=CC(I)=C(O)C(I)=C1 XUIIKFGFIJCVMT-GFCCVEGCSA-N 0.000 description 9
- FPIPGXGPPPQFEQ-OVSJKPMPSA-N all-trans-retinol Chemical compound OC\C=C(/C)\C=C\C=C(/C)\C=C\C1=C(C)CCCC1(C)C FPIPGXGPPPQFEQ-OVSJKPMPSA-N 0.000 description 9
- 125000004429 atom Chemical group 0.000 description 9
- 230000008901 benefit Effects 0.000 description 9
- 229910052799 carbon Inorganic materials 0.000 description 9
- 230000000694 effects Effects 0.000 description 9
- 150000002148 esters Chemical class 0.000 description 9
- 239000002777 nucleoside Substances 0.000 description 9
- 230000002829 reductive effect Effects 0.000 description 9
- 210000001519 tissue Anatomy 0.000 description 9
- 102000004190 Enzymes Human genes 0.000 description 8
- 108090000790 Enzymes Proteins 0.000 description 8
- 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 description 8
- 238000003556 assay Methods 0.000 description 8
- 210000003414 extremity Anatomy 0.000 description 8
- 230000006870 function Effects 0.000 description 8
- 210000004185 liver Anatomy 0.000 description 8
- 230000033001 locomotion Effects 0.000 description 8
- 239000000178 monomer Substances 0.000 description 8
- 239000000523 sample Substances 0.000 description 8
- 206010002023 Amyloidoses Diseases 0.000 description 7
- 229910019142 PO4 Inorganic materials 0.000 description 7
- 108020004459 Small interfering RNA Proteins 0.000 description 7
- 125000000217 alkyl group Chemical group 0.000 description 7
- 230000003941 amyloidogenesis Effects 0.000 description 7
- 208000003295 carpal tunnel syndrome Diseases 0.000 description 7
- 238000000338 in vitro Methods 0.000 description 7
- 238000001727 in vivo Methods 0.000 description 7
- 125000002496 methyl group Chemical group [H]C([H])([H])* 0.000 description 7
- 210000005036 nerve Anatomy 0.000 description 7
- 230000007830 nerve conduction Effects 0.000 description 7
- 210000000056 organ Anatomy 0.000 description 7
- 229950005564 patisiran Drugs 0.000 description 7
- 239000010452 phosphate Substances 0.000 description 7
- 230000035807 sensation Effects 0.000 description 7
- 230000001953 sensory effect Effects 0.000 description 7
- BGRJTUBHPOOWDU-UHFFFAOYSA-N sulpiride Chemical compound CCN1CCCC1CNC(=O)C1=CC(S(N)(=O)=O)=CC=C1OC BGRJTUBHPOOWDU-UHFFFAOYSA-N 0.000 description 7
- 230000008685 targeting Effects 0.000 description 7
- 229940034208 thyroxine Drugs 0.000 description 7
- XUIIKFGFIJCVMT-UHFFFAOYSA-N thyroxine-binding globulin Natural products IC1=CC(CC([NH3+])C([O-])=O)=CC(I)=C1OC1=CC(I)=C(O)C(I)=C1 XUIIKFGFIJCVMT-UHFFFAOYSA-N 0.000 description 7
- FPIPGXGPPPQFEQ-UHFFFAOYSA-N 13-cis retinol Natural products OCC=C(C)C=CC=C(C)C=CC1=C(C)CCCC1(C)C FPIPGXGPPPQFEQ-UHFFFAOYSA-N 0.000 description 6
- 102000035195 Peptidases Human genes 0.000 description 6
- 108091005804 Peptidases Proteins 0.000 description 6
- 102000000574 RNA-Induced Silencing Complex Human genes 0.000 description 6
- 108010016790 RNA-Induced Silencing Complex Proteins 0.000 description 6
- 208000034700 Vitreous opacities Diseases 0.000 description 6
- 229940024606 amino acid Drugs 0.000 description 6
- 150000001413 amino acids Chemical class 0.000 description 6
- 230000015572 biosynthetic process Effects 0.000 description 6
- HVYWMOMLDIMFJA-DPAQBDIFSA-N cholesterol Chemical group 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 description 6
- 208000035475 disorder Diseases 0.000 description 6
- UYTPUPDQBNUYGX-UHFFFAOYSA-N guanine Chemical compound O=C1NC(N)=NC2=C1N=CN2 UYTPUPDQBNUYGX-UHFFFAOYSA-N 0.000 description 6
- 210000003583 retinal pigment epithelium Anatomy 0.000 description 6
- 102000029752 retinol binding Human genes 0.000 description 6
- 108091000053 retinol binding Proteins 0.000 description 6
- 239000004576 sand Substances 0.000 description 6
- 102000009091 Amyloidogenic Proteins Human genes 0.000 description 5
- 108010048112 Amyloidogenic Proteins Proteins 0.000 description 5
- 108090000371 Esterases Proteins 0.000 description 5
- 230000002378 acidificating effect Effects 0.000 description 5
- 150000001408 amides Chemical group 0.000 description 5
- 230000003942 amyloidogenic effect Effects 0.000 description 5
- 210000001175 cerebrospinal fluid Anatomy 0.000 description 5
- 229940104302 cytosine Drugs 0.000 description 5
- 230000001419 dependent effect Effects 0.000 description 5
- 238000001514 detection method Methods 0.000 description 5
- AEUTYOVWOVBAKS-UWVGGRQHSA-N ethambutol Natural products CC[C@@H](CO)NCCN[C@@H](CC)CO AEUTYOVWOVBAKS-UWVGGRQHSA-N 0.000 description 5
- 102000056556 human TTR Human genes 0.000 description 5
- 238000009396 hybridization Methods 0.000 description 5
- 238000005259 measurement Methods 0.000 description 5
- 239000001301 oxygen Substances 0.000 description 5
- 125000000548 ribosyl group Chemical group C1([C@H](O)[C@H](O)[C@H](O1)CO)* 0.000 description 5
- 230000000630 rising effect Effects 0.000 description 5
- 102200150628 rs151220873 Human genes 0.000 description 5
- 230000001225 therapeutic effect Effects 0.000 description 5
- QPHRQMAYYMYWFW-FJGDRVTGSA-N 1-[(2r,3s,4r,5r)-3-fluoro-3,4-dihydroxy-5-(hydroxymethyl)oxolan-2-yl]pyrimidine-2,4-dione Chemical compound O[C@]1(F)[C@H](O)[C@@H](CO)O[C@H]1N1C(=O)NC(=O)C=C1 QPHRQMAYYMYWFW-FJGDRVTGSA-N 0.000 description 4
- IJGRMHOSHXDMSA-UHFFFAOYSA-N Atomic nitrogen Chemical group N#N IJGRMHOSHXDMSA-UHFFFAOYSA-N 0.000 description 4
- 206010019280 Heart failures Diseases 0.000 description 4
- 102100020870 La-related protein 6 Human genes 0.000 description 4
- 108050008265 La-related protein 6 Proteins 0.000 description 4
- 208000002193 Pain Diseases 0.000 description 4
- 241000282320 Panthera leo Species 0.000 description 4
- NBIIXXVUZAFLBC-UHFFFAOYSA-N Phosphoric acid Chemical compound OP(O)(O)=O NBIIXXVUZAFLBC-UHFFFAOYSA-N 0.000 description 4
- 206010040030 Sensory loss Diseases 0.000 description 4
- 241000384008 Sisis Species 0.000 description 4
- 230000036982 action potential Effects 0.000 description 4
- 125000000304 alkynyl group Chemical group 0.000 description 4
- 125000003118 aryl group Chemical group 0.000 description 4
- DDRJAANPRJIHGJ-UHFFFAOYSA-N creatinine Chemical compound CN1CC(=O)NC1=N DDRJAANPRJIHGJ-UHFFFAOYSA-N 0.000 description 4
- 206010012601 diabetes mellitus Diseases 0.000 description 4
- 238000009826 distribution Methods 0.000 description 4
- 230000004064 dysfunction Effects 0.000 description 4
- 238000011156 evaluation Methods 0.000 description 4
- 238000002347 injection Methods 0.000 description 4
- 239000007924 injection Substances 0.000 description 4
- 208000017169 kidney disease Diseases 0.000 description 4
- 150000002632 lipids Chemical group 0.000 description 4
- 210000003141 lower extremity Anatomy 0.000 description 4
- 238000012544 monitoring process Methods 0.000 description 4
- 230000008035 nerve activity Effects 0.000 description 4
- 150000003833 nucleoside derivatives Chemical class 0.000 description 4
- 125000003835 nucleoside group Chemical group 0.000 description 4
- 210000004345 peroneal nerve Anatomy 0.000 description 4
- 239000012071 phase Substances 0.000 description 4
- NBIIXXVUZAFLBC-UHFFFAOYSA-K phosphate Chemical compound [O-]P([O-])([O-])=O NBIIXXVUZAFLBC-UHFFFAOYSA-K 0.000 description 4
- 125000004437 phosphorous atom Chemical group 0.000 description 4
- 229910052698 phosphorus Inorganic materials 0.000 description 4
- 102000040430 polynucleotide Human genes 0.000 description 4
- 108091033319 polynucleotide Proteins 0.000 description 4
- 239000002157 polynucleotide Substances 0.000 description 4
- 230000008569 process Effects 0.000 description 4
- 230000004044 response Effects 0.000 description 4
- 238000012552 review Methods 0.000 description 4
- RWQNBRDOKXIBIV-UHFFFAOYSA-N thymine Chemical compound CC1=CNC(=O)NC1=O RWQNBRDOKXIBIV-UHFFFAOYSA-N 0.000 description 4
- GFFGJBXGBJISGV-UHFFFAOYSA-N Adenine Chemical compound NC1=NC=NC2=C1N=CN2 GFFGJBXGBJISGV-UHFFFAOYSA-N 0.000 description 3
- 102000001049 Amyloid Human genes 0.000 description 3
- 108010094108 Amyloid Proteins 0.000 description 3
- 206010010904 Convulsion Diseases 0.000 description 3
- HMFHBZSHGGEWLO-SOOFDHNKSA-N D-ribofuranose Chemical compound OC[C@H]1OC(O)[C@H](O)[C@@H]1O HMFHBZSHGGEWLO-SOOFDHNKSA-N 0.000 description 3
- 206010012289 Dementia Diseases 0.000 description 3
- 238000012286 ELISA Assay Methods 0.000 description 3
- 206010028570 Myelopathy Diseases 0.000 description 3
- 108091093037 Peptide nucleic acid Proteins 0.000 description 3
- 206010034620 Peripheral sensory neuropathy Diseases 0.000 description 3
- 239000004365 Protease Substances 0.000 description 3
- PYMYPHUHKUWMLA-LMVFSUKVSA-N Ribose Natural products OC[C@@H](O)[C@@H](O)[C@@H](O)C=O PYMYPHUHKUWMLA-LMVFSUKVSA-N 0.000 description 3
- FAPWRFPIFSIZLT-UHFFFAOYSA-M Sodium chloride Chemical compound [Na+].[Cl-] FAPWRFPIFSIZLT-UHFFFAOYSA-M 0.000 description 3
- 208000007107 Stomach Ulcer Diseases 0.000 description 3
- ISAKRJDGNUQOIC-UHFFFAOYSA-N Uracil Natural products O=C1C=CNC(=O)N1 ISAKRJDGNUQOIC-UHFFFAOYSA-N 0.000 description 3
- FPIPGXGPPPQFEQ-BOOMUCAASA-N Vitamin A Natural products OC/C=C(/C)\C=C\C=C(\C)/C=C/C1=C(C)CCCC1(C)C FPIPGXGPPPQFEQ-BOOMUCAASA-N 0.000 description 3
- 230000002159 abnormal effect Effects 0.000 description 3
- 125000003342 alkenyl group Chemical group 0.000 description 3
- 125000002947 alkylene group Chemical group 0.000 description 3
- HMFHBZSHGGEWLO-UHFFFAOYSA-N alpha-D-Furanose-Ribose Natural products OCC1OC(O)C(O)C1O HMFHBZSHGGEWLO-UHFFFAOYSA-N 0.000 description 3
- VREFGVBLTWBCJP-UHFFFAOYSA-N alprazolam Chemical compound C12=CC(Cl)=CC=C2N2C(C)=NN=C2CN=C1C1=CC=CC=C1 VREFGVBLTWBCJP-UHFFFAOYSA-N 0.000 description 3
- QVGXLLKOCUKJST-UHFFFAOYSA-N atomic oxygen Chemical compound [O] QVGXLLKOCUKJST-UHFFFAOYSA-N 0.000 description 3
- 125000002619 bicyclic group Chemical group 0.000 description 3
- 125000004432 carbon atom Chemical group C* 0.000 description 3
- 235000012000 cholesterol Nutrition 0.000 description 3
- 230000003413 degradative effect Effects 0.000 description 3
- 230000008021 deposition Effects 0.000 description 3
- 238000003745 diagnosis Methods 0.000 description 3
- 238000002405 diagnostic procedure Methods 0.000 description 3
- 239000000539 dimer Substances 0.000 description 3
- 230000003467 diminishing effect Effects 0.000 description 3
- 210000001163 endosome Anatomy 0.000 description 3
- 239000012530 fluid Substances 0.000 description 3
- 201000005917 gastric ulcer Diseases 0.000 description 3
- 230000007160 gastrointestinal dysfunction Effects 0.000 description 3
- 230000030279 gene silencing Effects 0.000 description 3
- 230000005484 gravity Effects 0.000 description 3
- 230000004217 heart function Effects 0.000 description 3
- 229910052739 hydrogen Inorganic materials 0.000 description 3
- 239000001257 hydrogen Substances 0.000 description 3
- 150000002500 ions Chemical class 0.000 description 3
- 210000005229 liver cell Anatomy 0.000 description 3
- 150000002772 monosaccharides Chemical group 0.000 description 3
- 210000000653 nervous system Anatomy 0.000 description 3
- 208000035824 paresthesia Diseases 0.000 description 3
- 230000037361 pathway Effects 0.000 description 3
- 210000002381 plasma Anatomy 0.000 description 3
- 102000004196 processed proteins & peptides Human genes 0.000 description 3
- 235000019833 protease Nutrition 0.000 description 3
- 238000011160 research Methods 0.000 description 3
- 230000029058 respiratory gaseous exchange Effects 0.000 description 3
- 229960003471 retinol Drugs 0.000 description 3
- 235000020944 retinol Nutrition 0.000 description 3
- 239000011607 retinol Substances 0.000 description 3
- 201000005572 sensory peripheral neuropathy Diseases 0.000 description 3
- 238000010254 subcutaneous injection Methods 0.000 description 3
- 239000007929 subcutaneous injection Substances 0.000 description 3
- 150000008163 sugars Chemical class 0.000 description 3
- 238000003786 synthesis reaction Methods 0.000 description 3
- 230000008719 thickening Effects 0.000 description 3
- 210000002303 tibia Anatomy 0.000 description 3
- 238000011830 transgenic mouse model Methods 0.000 description 3
- 235000019155 vitamin A Nutrition 0.000 description 3
- 239000011719 vitamin A Substances 0.000 description 3
- 229940045997 vitamin a Drugs 0.000 description 3
- XLYOFNOQVPJJNP-UHFFFAOYSA-N water Chemical compound O XLYOFNOQVPJJNP-UHFFFAOYSA-N 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
- FZWGECJQACGGTI-UHFFFAOYSA-N 2-amino-7-methyl-1,7-dihydro-6H-purin-6-one Chemical compound NC1=NC(O)=C2N(C)C=NC2=N1 FZWGECJQACGGTI-UHFFFAOYSA-N 0.000 description 2
- OVONXEQGWXGFJD-UHFFFAOYSA-N 4-sulfanylidene-1h-pyrimidin-2-one Chemical compound SC=1C=CNC(=O)N=1 OVONXEQGWXGFJD-UHFFFAOYSA-N 0.000 description 2
- HCGHYQLFMPXSDU-UHFFFAOYSA-N 7-methyladenine Chemical compound C1=NC(N)=C2N(C)C=NC2=N1 HCGHYQLFMPXSDU-UHFFFAOYSA-N 0.000 description 2
- KDCGOANMDULRCW-UHFFFAOYSA-N 7H-purine Chemical compound N1=CNC2=NC=NC2=C1 KDCGOANMDULRCW-UHFFFAOYSA-N 0.000 description 2
- LRFVTYWOQMYALW-UHFFFAOYSA-N 9H-xanthine Chemical compound O=C1NC(=O)NC2=C1NC=N2 LRFVTYWOQMYALW-UHFFFAOYSA-N 0.000 description 2
- 229930024421 Adenine Natural products 0.000 description 2
- IYMAXBFPHPZYIK-BQBZGAKWSA-N Arg-Gly-Asp Chemical compound NC(N)=NCCC[C@H](N)C(=O)NCC(=O)N[C@@H](CC(O)=O)C(O)=O IYMAXBFPHPZYIK-BQBZGAKWSA-N 0.000 description 2
- 206010003840 Autonomic nervous system imbalance Diseases 0.000 description 2
- 229940122361 Bisphosphonate Drugs 0.000 description 2
- 206010010774 Constipation Diseases 0.000 description 2
- 206010012735 Diarrhoea Diseases 0.000 description 2
- RTZKZFJDLAIYFH-UHFFFAOYSA-N Diethyl ether Chemical compound CCOCC RTZKZFJDLAIYFH-UHFFFAOYSA-N 0.000 description 2
- MYMOFIZGZYHOMD-UHFFFAOYSA-N Dioxygen Chemical group O=O MYMOFIZGZYHOMD-UHFFFAOYSA-N 0.000 description 2
- 208000010228 Erectile Dysfunction Diseases 0.000 description 2
- 208000010412 Glaucoma Diseases 0.000 description 2
- 108090000288 Glycoproteins Proteins 0.000 description 2
- 102000003886 Glycoproteins Human genes 0.000 description 2
- UFHFLCQGNIYNRP-UHFFFAOYSA-N Hydrogen Chemical compound [H][H] UFHFLCQGNIYNRP-UHFFFAOYSA-N 0.000 description 2
- 208000004044 Hypesthesia Diseases 0.000 description 2
- 208000001953 Hypotension Diseases 0.000 description 2
- 241000282567 Macaca fascicularis Species 0.000 description 2
- 101100154773 Macaca fascicularis TTR gene Proteins 0.000 description 2
- 241001465754 Metazoa Species 0.000 description 2
- 241000699660 Mus musculus Species 0.000 description 2
- 101100154776 Mus musculus Ttr gene Proteins 0.000 description 2
- OHLUUHNLEMFGTQ-UHFFFAOYSA-N N-methylacetamide Chemical compound CNC(C)=O OHLUUHNLEMFGTQ-UHFFFAOYSA-N 0.000 description 2
- 108091028043 Nucleic acid sequence Proteins 0.000 description 2
- 206010033799 Paralysis Diseases 0.000 description 2
- 206010034568 Peripheral coldness Diseases 0.000 description 2
- 241000288906 Primates Species 0.000 description 2
- 208000001647 Renal Insufficiency Diseases 0.000 description 2
- 241000283984 Rodentia Species 0.000 description 2
- 229910020008 S(O) Inorganic materials 0.000 description 2
- 241000527181 Sisor Species 0.000 description 2
- 206010043248 Tendon rupture Diseases 0.000 description 2
- IQFYYKKMVGJFEH-XLPZGREQSA-N Thymidine Chemical compound O=C1NC(=O)C(C)=CN1[C@@H]1O[C@H](CO)[C@@H](O)C1 IQFYYKKMVGJFEH-XLPZGREQSA-N 0.000 description 2
- ATJFFYVFTNAWJD-UHFFFAOYSA-N Tin Chemical compound [Sn] ATJFFYVFTNAWJD-UHFFFAOYSA-N 0.000 description 2
- 241000335654 Uracis Species 0.000 description 2
- DRTQHJPVMGBUCF-XVFCMESISA-N Uridine Chemical compound O[C@@H]1[C@H](O)[C@@H](CO)O[C@H]1N1C(=O)NC(=O)C=C1 DRTQHJPVMGBUCF-XVFCMESISA-N 0.000 description 2
- 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 description 2
- 230000003187 abdominal effect Effects 0.000 description 2
- MKUXAQIIEYXACX-UHFFFAOYSA-N aciclovir Chemical compound N1C(N)=NC(=O)C2=C1N(COCCO)C=N2 MKUXAQIIEYXACX-UHFFFAOYSA-N 0.000 description 2
- 150000007513 acids Chemical class 0.000 description 2
- 238000007792 addition Methods 0.000 description 2
- 229960000643 adenine Drugs 0.000 description 2
- 230000002411 adverse Effects 0.000 description 2
- 230000002776 aggregation Effects 0.000 description 2
- 238000004220 aggregation Methods 0.000 description 2
- 125000003368 amide group Chemical group 0.000 description 2
- 238000004458 analytical method Methods 0.000 description 2
- 108010072041 arginyl-glycyl-aspartic acid Proteins 0.000 description 2
- 206010003119 arrhythmia Diseases 0.000 description 2
- 125000005018 aryl alkenyl group Chemical group 0.000 description 2
- 125000003710 aryl alkyl group Chemical group 0.000 description 2
- 230000001174 ascending effect Effects 0.000 description 2
- 150000004663 bisphosphonates Chemical class 0.000 description 2
- 230000037396 body weight Effects 0.000 description 2
- 210000000988 bone and bone Anatomy 0.000 description 2
- 239000000872 buffer Substances 0.000 description 2
- 230000015556 catabolic process Effects 0.000 description 2
- 230000021615 conjugation Effects 0.000 description 2
- 230000002596 correlated effect Effects 0.000 description 2
- 230000000875 corresponding effect Effects 0.000 description 2
- 229940109239 creatinine Drugs 0.000 description 2
- 125000004122 cyclic group Chemical group 0.000 description 2
- 125000000753 cycloalkyl group Chemical group 0.000 description 2
- 238000006731 degradation reaction Methods 0.000 description 2
- HUPFGZXOMWLGNK-UHFFFAOYSA-N diflunisal Chemical compound C1=C(O)C(C(=O)O)=CC(C=2C(=CC(F)=CC=2)F)=C1 HUPFGZXOMWLGNK-UHFFFAOYSA-N 0.000 description 2
- 229960000616 diflunisal Drugs 0.000 description 2
- VHJLVAABSRFDPM-QWWZWVQMSA-N dithiothreitol Chemical compound SC[C@@H](O)[C@H](O)CS VHJLVAABSRFDPM-QWWZWVQMSA-N 0.000 description 2
- 229940079593 drug Drugs 0.000 description 2
- 238000002592 echocardiography Methods 0.000 description 2
- 238000002565 electrocardiography Methods 0.000 description 2
- 210000002683 foot Anatomy 0.000 description 2
- 238000009472 formulation Methods 0.000 description 2
- 229930182830 galactose Natural products 0.000 description 2
- 230000002068 genetic effect Effects 0.000 description 2
- 125000001475 halogen functional group Chemical group 0.000 description 2
- 125000005842 heteroatom Chemical group 0.000 description 2
- 125000004029 hydroxymethyl group Chemical group [H]OC([H])([H])* 0.000 description 2
- 208000034783 hypoesthesia Diseases 0.000 description 2
- 230000036543 hypotension Effects 0.000 description 2
- FDGQSTZJBFJUBT-UHFFFAOYSA-N hypoxanthine Chemical compound O=C1NC=NC2=C1NC=N2 FDGQSTZJBFJUBT-UHFFFAOYSA-N 0.000 description 2
- 230000001771 impaired effect Effects 0.000 description 2
- 201000001881 impotence Diseases 0.000 description 2
- 230000003993 interaction Effects 0.000 description 2
- 201000006370 kidney failure Diseases 0.000 description 2
- 210000003712 lysosome Anatomy 0.000 description 2
- 230000001868 lysosomic effect Effects 0.000 description 2
- 238000012423 maintenance Methods 0.000 description 2
- 238000004519 manufacturing process Methods 0.000 description 2
- 230000003340 mental effect Effects 0.000 description 2
- 230000004630 mental health Effects 0.000 description 2
- 125000000956 methoxy group Chemical group [H]C([H])([H])O* 0.000 description 2
- 125000000740 n-pentyl group Chemical group [H]C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])* 0.000 description 2
- 229910052757 nitrogen Inorganic materials 0.000 description 2
- 210000000496 pancreas Anatomy 0.000 description 2
- 230000001575 pathological effect Effects 0.000 description 2
- 210000001428 peripheral nervous system Anatomy 0.000 description 2
- 125000002467 phosphate group Chemical group [H]OP(=O)(O[H])O[*] 0.000 description 2
- 230000001144 postural effect Effects 0.000 description 2
- 239000002243 precursor Substances 0.000 description 2
- 238000002360 preparation method Methods 0.000 description 2
- 230000000750 progressive effect Effects 0.000 description 2
- 235000019419 proteases Nutrition 0.000 description 2
- 210000001747 pupil Anatomy 0.000 description 2
- 210000001525 retina Anatomy 0.000 description 2
- 229920002477 rna polymer Polymers 0.000 description 2
- 150000003839 salts Chemical class 0.000 description 2
- 238000012216 screening Methods 0.000 description 2
- 239000007790 solid phase Substances 0.000 description 2
- 239000000243 solution Substances 0.000 description 2
- 230000006641 stabilisation Effects 0.000 description 2
- 238000011105 stabilization Methods 0.000 description 2
- 239000000126 substance Substances 0.000 description 2
- 238000006467 substitution reaction Methods 0.000 description 2
- 239000000758 substrate Substances 0.000 description 2
- 125000004434 sulfur atom Chemical group 0.000 description 2
- RAHZWNYVWXNFOC-UHFFFAOYSA-N sulfur dioxide Inorganic materials O=S=O RAHZWNYVWXNFOC-UHFFFAOYSA-N 0.000 description 2
- 230000001629 suppression Effects 0.000 description 2
- 208000011580 syndromic disease Diseases 0.000 description 2
- TXEIIPDJKFWEEC-UHFFFAOYSA-N tafamidis Chemical compound O1C2=CC(C(=O)O)=CC=C2N=C1C1=CC(Cl)=CC(Cl)=C1 TXEIIPDJKFWEEC-UHFFFAOYSA-N 0.000 description 2
- 229960001353 tafamidis Drugs 0.000 description 2
- RYYWUUFWQRZTIU-UHFFFAOYSA-K thiophosphate Chemical compound [O-]P([O-])([O-])=S RYYWUUFWQRZTIU-UHFFFAOYSA-K 0.000 description 2
- 229940113082 thymine Drugs 0.000 description 2
- 230000032258 transport Effects 0.000 description 2
- VLPFTAMPNXLGLX-UHFFFAOYSA-N trioctanoin Chemical compound CCCCCCCC(=O)OCC(OC(=O)CCCCCCC)COC(=O)CCCCCCC VLPFTAMPNXLGLX-UHFFFAOYSA-N 0.000 description 2
- 210000000623 ulna Anatomy 0.000 description 2
- 229940035893 uracil Drugs 0.000 description 2
- PMBSJRVVKHNWGX-GIWSHQQXSA-N (2r,3s,4r,5r)-2-(6-aminopurin-9-yl)-4-dihydroxyphosphinothioyloxy-3-fluoro-5-(hydroxymethyl)oxolan-3-ol Chemical compound C1=NC=2C(N)=NC=NC=2N1[C@@H]1O[C@H](CO)[C@@H](OP(O)(O)=S)[C@]1(O)F PMBSJRVVKHNWGX-GIWSHQQXSA-N 0.000 description 1
- HSINOMROUCMIEA-FGVHQWLLSA-N (2s,4r)-4-[(3r,5s,6r,7r,8s,9s,10s,13r,14s,17r)-6-ethyl-3,7-dihydroxy-10,13-dimethyl-2,3,4,5,6,7,8,9,11,12,14,15,16,17-tetradecahydro-1h-cyclopenta[a]phenanthren-17-yl]-2-methylpentanoic acid Chemical compound C([C@@]12C)C[C@@H](O)C[C@H]1[C@@H](CC)[C@@H](O)[C@@H]1[C@@H]2CC[C@]2(C)[C@@H]([C@H](C)C[C@H](C)C(O)=O)CC[C@H]21 HSINOMROUCMIEA-FGVHQWLLSA-N 0.000 description 1
- BHQCQFFYRZLCQQ-UHFFFAOYSA-N (3alpha,5alpha,7alpha,12alpha)-3,7,12-trihydroxy-cholan-24-oic acid Natural products OC1CC2CC(O)CCC2(C)C2C1C1CCC(C(CCC(O)=O)C)C1(C)C(O)C2 BHQCQFFYRZLCQQ-UHFFFAOYSA-N 0.000 description 1
- QGVQZRDQPDLHHV-DPAQBDIFSA-N (3s,8s,9s,10r,13r,14s,17r)-10,13-dimethyl-17-[(2r)-6-methylheptan-2-yl]-2,3,4,7,8,9,11,12,14,15,16,17-dodecahydro-1h-cyclopenta[a]phenanthrene-3-thiol Chemical compound C1C=C2C[C@@H](S)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 QGVQZRDQPDLHHV-DPAQBDIFSA-N 0.000 description 1
- 125000004400 (C1-C12) alkyl group Chemical group 0.000 description 1
- 125000003837 (C1-C20) alkyl group Chemical group 0.000 description 1
- FYADHXFMURLYQI-UHFFFAOYSA-N 1,2,4-triazine Chemical class C1=CN=NC=N1 FYADHXFMURLYQI-UHFFFAOYSA-N 0.000 description 1
- KLEGMTRDCCDFJK-XDQSQZFTSA-N 1-[(2R,4S,5R)-4-[[(2R,3S,5R)-3-[[(2R,3S,5R)-5-(4-amino-5-methyl-2-oxopyrimidin-1-yl)-3-[[(2R,3S,5R)-3-[[(2R,3S,5R)-3-[[(2R,3S,5R)-3-[[(2R,3S,5R)-3-[[(2R,3S,5R)-3-[[(2R,3R,4R,5R)-3-[[(2R,3R,4R,5R)-3-[[(2R,3R,4R,5R)-5-(4-amino-5-methyl-2-oxopyrimidin-1-yl)-3-[[(2R,3R,4R,5R)-5-(4-amino-5-methyl-2-oxopyrimidin-1-yl)-3-[[(2R,3R,4R,5R)-5-(4-amino-5-methyl-2-oxopyrimidin-1-yl)-3-hydroxy-4-(2-methoxyethoxy)oxolan-2-yl]methoxy-hydroxyphosphinothioyl]oxy-4-(2-methoxyethoxy)oxolan-2-yl]methoxy-hydroxyphosphinothioyl]oxy-4-(2-methoxyethoxy)oxolan-2-yl]methoxy-hydroxyphosphinothioyl]oxy-4-(2-methoxyethoxy)-5-(5-methyl-2,4-dioxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphinothioyl]oxy-5-(6-aminopurin-9-yl)-4-(2-methoxyethoxy)oxolan-2-yl]methoxy-hydroxyphosphinothioyl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphinothioyl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphinothioyl]oxy-5-(2-amino-6-oxo-1H-purin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphinothioyl]oxy-5-(5-methyl-2,4-dioxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphinothioyl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphinothioyl]oxyoxolan-2-yl]methoxy-hydroxyphosphinothioyl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphinothioyl]oxy-5-[[[(2R,3S,5R)-2-[[[(2R,3S,5R)-2-[[[(2R,3R,4R,5R)-2-[[[(2R,3R,4R,5R)-2-[[[(2R,3R,4R,5R)-2-[[[(2R,3R,4R,5R)-5-(4-amino-5-methyl-2-oxopyrimidin-1-yl)-2-[[hydroxy-[(2R,3R,4R,5R)-2-(hydroxymethyl)-4-(2-methoxyethoxy)-5-(5-methyl-2,4-dioxopyrimidin-1-yl)oxolan-3-yl]oxyphosphinothioyl]oxymethyl]-4-(2-methoxyethoxy)oxolan-3-yl]oxy-sulfanylphosphoryl]oxymethyl]-4-(2-methoxyethoxy)-5-(5-methyl-2,4-dioxopyrimidin-1-yl)oxolan-3-yl]oxy-hydroxyphosphinothioyl]oxymethyl]-4-(2-methoxyethoxy)-5-(5-methyl-2,4-dioxopyrimidin-1-yl)oxolan-3-yl]oxy-hydroxyphosphinothioyl]oxymethyl]-5-(2-amino-6-oxo-1H-purin-9-yl)-4-(2-methoxyethoxy)oxolan-3-yl]oxy-hydroxyphosphinothioyl]oxymethyl]-5-(2-amino-6-oxo-1H-purin-9-yl)oxolan-3-yl]oxy-hydroxyphosphinothioyl]oxymethyl]-5-(5-methyl-2,4-dioxopyrimidin-1-yl)oxolan-3-yl]oxy-hydroxyphosphinothioyl]oxymethyl]oxolan-2-yl]-5-methylpyrimidine-2,4-dione Chemical compound COCCO[C@@H]1[C@H](O)[C@@H](COP(O)(=S)O[C@@H]2[C@@H](COP(O)(=S)O[C@@H]3[C@@H](COP(O)(=S)O[C@@H]4[C@@H](COP(O)(=S)O[C@@H]5[C@@H](COP(O)(=S)O[C@H]6C[C@@H](O[C@@H]6COP(O)(=S)O[C@H]6C[C@@H](O[C@@H]6COP(O)(=S)O[C@H]6C[C@@H](O[C@@H]6COP(O)(=S)O[C@H]6C[C@@H](O[C@@H]6COP(O)(=S)O[C@H]6C[C@@H](O[C@@H]6COP(O)(=S)O[C@H]6C[C@@H](O[C@@H]6COP(O)(=S)O[C@H]6C[C@@H](O[C@@H]6COP(O)(=S)O[C@H]6C[C@@H](O[C@@H]6COP(O)(=S)O[C@H]6C[C@@H](O[C@@H]6COP(O)(=S)O[C@H]6C[C@@H](O[C@@H]6COP(O)(=S)O[C@@H]6[C@@H](COP(O)(=S)O[C@@H]7[C@@H](COP(O)(=S)O[C@@H]8[C@@H](COP(S)(=O)O[C@@H]9[C@@H](COP(O)(=S)O[C@@H]%10[C@@H](CO)O[C@H]([C@@H]%10OCCOC)n%10cc(C)c(=O)[nH]c%10=O)O[C@H]([C@@H]9OCCOC)n9cc(C)c(N)nc9=O)O[C@H]([C@@H]8OCCOC)n8cc(C)c(=O)[nH]c8=O)O[C@H]([C@@H]7OCCOC)n7cc(C)c(=O)[nH]c7=O)O[C@H]([C@@H]6OCCOC)n6cnc7c6nc(N)[nH]c7=O)n6cnc7c6nc(N)[nH]c7=O)n6cc(C)c(=O)[nH]c6=O)n6cc(C)c(=O)[nH]c6=O)n6cnc7c(N)ncnc67)n6cc(C)c(N)nc6=O)n6cnc7c(N)ncnc67)n6cc(C)c(=O)[nH]c6=O)n6cnc7c6nc(N)[nH]c7=O)n6cnc7c(N)ncnc67)n6cnc7c(N)ncnc67)O[C@H]([C@@H]5OCCOC)n5cnc6c(N)ncnc56)O[C@H]([C@@H]4OCCOC)n4cc(C)c(=O)[nH]c4=O)O[C@H]([C@@H]3OCCOC)n3cc(C)c(N)nc3=O)O[C@H]([C@@H]2OCCOC)n2cc(C)c(N)nc2=O)O[C@H]1n1cc(C)c(N)nc1=O KLEGMTRDCCDFJK-XDQSQZFTSA-N 0.000 description 1
- UIFJBEWVRSHKCF-ZOQUXTDFSA-N 1-[(2r,3r,4r,5r)-4-dihydroxyphosphinothioyloxy-5-(hydroxymethyl)-3-methoxyoxolan-2-yl]pyrimidine-2,4-dione Chemical compound CO[C@@H]1[C@H](OP(O)(O)=S)[C@@H](CO)O[C@H]1N1C(=O)NC(=O)C=C1 UIFJBEWVRSHKCF-ZOQUXTDFSA-N 0.000 description 1
- QXMLYBQBMGZPTE-LTNPLRIYSA-N 1-[(2r,3s,4r,5r)-4-dihydroxyphosphinothioyloxy-3-fluoro-3-hydroxy-5-(hydroxymethyl)oxolan-2-yl]-5-methylpyrimidine-2,4-dione Chemical compound O=C1NC(=O)C(C)=CN1[C@H]1[C@](F)(O)[C@H](OP(O)(O)=S)[C@@H](CO)O1 QXMLYBQBMGZPTE-LTNPLRIYSA-N 0.000 description 1
- XKKRMVGRQHZUHY-FJGDRVTGSA-N 1-[(2r,3s,4r,5r)-4-dihydroxyphosphinothioyloxy-3-fluoro-3-hydroxy-5-(hydroxymethyl)oxolan-2-yl]pyrimidine-2,4-dione Chemical compound O[C@]1(F)[C@H](OP(O)(O)=S)[C@@H](CO)O[C@H]1N1C(=O)NC(=O)C=C1 XKKRMVGRQHZUHY-FJGDRVTGSA-N 0.000 description 1
- UHUHBFMZVCOEOV-UHFFFAOYSA-N 1h-imidazo[4,5-c]pyridin-4-amine Chemical compound NC1=NC=CC2=C1N=CN2 UHUHBFMZVCOEOV-UHFFFAOYSA-N 0.000 description 1
- IHPYMWDTONKSCO-UHFFFAOYSA-N 2,2'-piperazine-1,4-diylbisethanesulfonic acid Chemical compound OS(=O)(=O)CCN1CCN(CCS(O)(=O)=O)CC1 IHPYMWDTONKSCO-UHFFFAOYSA-N 0.000 description 1
- PXSCCKWNFZTKMS-UHFFFAOYSA-N 2-(2-aminopropyl)-7H-purin-6-amine 5-prop-1-ynyl-1H-pyrimidine-2,4-dione Chemical compound C(#CC)C=1C(NC(NC1)=O)=O.NC(CC1=NC(=C2NC=NC2=N1)N)C PXSCCKWNFZTKMS-UHFFFAOYSA-N 0.000 description 1
- PIINGYXNCHTJTF-UHFFFAOYSA-N 2-(2-azaniumylethylamino)acetate Chemical group NCCNCC(O)=O PIINGYXNCHTJTF-UHFFFAOYSA-N 0.000 description 1
- BVQKNSCKIBSOGA-KQYNXXCUSA-N 2-amino-9-[(2r,3r,4r,5r)-4-dihydroxyphosphinothioyloxy-5-(hydroxymethyl)-3-methoxyoxolan-2-yl]-3h-purin-6-one Chemical compound CO[C@@H]1[C@H](OP(O)(O)=S)[C@@H](CO)O[C@H]1N1C(NC(N)=NC2=O)=C2N=C1 BVQKNSCKIBSOGA-KQYNXXCUSA-N 0.000 description 1
- QFOCLINPINTVJI-UUOKFMHZSA-N 2-amino-9-[(2r,3r,4s,5r)-4-dihydroxyphosphinothioyloxy-3-hydroxy-5-(hydroxymethyl)oxolan-2-yl]-3h-purin-6-one Chemical compound C1=2NC(N)=NC(=O)C=2N=CN1[C@@H]1O[C@H](CO)[C@@H](OP(O)(O)=S)[C@H]1O QFOCLINPINTVJI-UUOKFMHZSA-N 0.000 description 1
- PLWXKAOZQSMLOO-AEHJODJJSA-N 2-amino-9-[(2r,3s,4r,5r)-4-dihydroxyphosphinothioyloxy-3-fluoro-3-hydroxy-5-(hydroxymethyl)oxolan-2-yl]-3h-purin-6-one Chemical compound C1=2NC(N)=NC(=O)C=2N=CN1[C@@H]1O[C@H](CO)[C@@H](OP(O)(O)=S)[C@]1(O)F PLWXKAOZQSMLOO-AEHJODJJSA-N 0.000 description 1
- 125000004200 2-methoxyethyl group Chemical group [H]C([H])([H])OC([H])([H])C([H])([H])* 0.000 description 1
- UOOOPKANIPLQPU-XVFCMESISA-N 3'-CMP Chemical compound O=C1N=C(N)C=CN1[C@H]1[C@H](O)[C@H](OP(O)(O)=O)[C@@H](CO)O1 UOOOPKANIPLQPU-XVFCMESISA-N 0.000 description 1
- FOGRQMPFHUHIGU-XVFCMESISA-N 3'-UMP Chemical compound O[C@@H]1[C@H](OP(O)(O)=O)[C@@H](CO)O[C@H]1N1C(=O)NC(=O)C=C1 FOGRQMPFHUHIGU-XVFCMESISA-N 0.000 description 1
- ZCFXTYHOHYXCTE-FJGDRVTGSA-N 4-amino-1-[(2r,3s,4r,5r)-4-dihydroxyphosphinothioyloxy-3-fluoro-3-hydroxy-5-(hydroxymethyl)oxolan-2-yl]pyrimidin-2-one Chemical compound O=C1N=C(N)C=CN1[C@H]1[C@](F)(O)[C@H](OP(O)(O)=S)[C@@H](CO)O1 ZCFXTYHOHYXCTE-FJGDRVTGSA-N 0.000 description 1
- ZLAQATDNGLKIEV-UHFFFAOYSA-N 5-methyl-2-sulfanylidene-1h-pyrimidin-4-one Chemical compound CC1=CNC(=S)NC1=O ZLAQATDNGLKIEV-UHFFFAOYSA-N 0.000 description 1
- LRSASMSXMSNRBT-UHFFFAOYSA-N 5-methylcytosine Chemical compound CC1=CNC(=O)N=C1N LRSASMSXMSNRBT-UHFFFAOYSA-N 0.000 description 1
- VPIAFVALSSSQJN-RGURZIINSA-N 6-amino-1-[(2s)-4-hydroxy-2-(hydroxymethyl)pyrrolidin-1-yl]hexan-1-one Chemical compound NCCCCCC(=O)N1CC(O)C[C@H]1CO VPIAFVALSSSQJN-RGURZIINSA-N 0.000 description 1
- DCPSTSVLRXOYGS-UHFFFAOYSA-N 6-amino-1h-pyrimidine-2-thione Chemical compound NC1=CC=NC(S)=N1 DCPSTSVLRXOYGS-UHFFFAOYSA-N 0.000 description 1
- QNNARSZPGNJZIX-UHFFFAOYSA-N 6-amino-5-prop-1-ynyl-1h-pyrimidin-2-one Chemical compound CC#CC1=CNC(=O)N=C1N QNNARSZPGNJZIX-UHFFFAOYSA-N 0.000 description 1
- LOSIULRWFAEMFL-UHFFFAOYSA-N 7-deazaguanine Chemical compound O=C1NC(N)=NC2=C1CC=N2 LOSIULRWFAEMFL-UHFFFAOYSA-N 0.000 description 1
- HRYKDUPGBWLLHO-UHFFFAOYSA-N 8-azaadenine Chemical compound NC1=NC=NC2=NNN=C12 HRYKDUPGBWLLHO-UHFFFAOYSA-N 0.000 description 1
- LPXQRXLUHJKZIE-UHFFFAOYSA-N 8-azaguanine Chemical compound NC1=NC(O)=C2NN=NC2=N1 LPXQRXLUHJKZIE-UHFFFAOYSA-N 0.000 description 1
- 229960005508 8-azaguanine Drugs 0.000 description 1
- 206010069754 Acquired gene mutation Diseases 0.000 description 1
- 108700028369 Alleles Proteins 0.000 description 1
- 108090000531 Amidohydrolases Proteins 0.000 description 1
- 102000004092 Amidohydrolases Human genes 0.000 description 1
- 241000554155 Andes Species 0.000 description 1
- 102100034608 Angiopoietin-2 Human genes 0.000 description 1
- 102000007592 Apolipoproteins Human genes 0.000 description 1
- 108010071619 Apolipoproteins Proteins 0.000 description 1
- 206010061666 Autonomic neuropathy Diseases 0.000 description 1
- 102100031500 Beta-1,4-glucuronyltransferase 1 Human genes 0.000 description 1
- 102100026031 Beta-glucuronidase Human genes 0.000 description 1
- NLZUEZXRPGMBCV-UHFFFAOYSA-N Butylhydroxytoluene Chemical compound CC1=CC(C(C)(C)C)=C(O)C(C(C)(C)C)=C1 NLZUEZXRPGMBCV-UHFFFAOYSA-N 0.000 description 1
- 102100036849 C-C motif chemokine 24 Human genes 0.000 description 1
- 102100036170 C-X-C motif chemokine 9 Human genes 0.000 description 1
- 102100027207 CD27 antigen Human genes 0.000 description 1
- 102100029380 CMRF35-like molecule 2 Human genes 0.000 description 1
- 101100241173 Caenorhabditis elegans dat-1 gene Proteins 0.000 description 1
- OKTJSMMVPCPJKN-UHFFFAOYSA-N Carbon Chemical compound [C] OKTJSMMVPCPJKN-UHFFFAOYSA-N 0.000 description 1
- 108010078791 Carrier Proteins Proteins 0.000 description 1
- 102100032219 Cathepsin D Human genes 0.000 description 1
- GXCRUTWHNMMJEK-WYUVZMMLSA-M Cefacetrile sodium Chemical compound [Na+].S1CC(COC(=O)C)=C(C([O-])=O)N2C(=O)[C@@H](NC(=O)CC#N)[C@@H]12 GXCRUTWHNMMJEK-WYUVZMMLSA-M 0.000 description 1
- 241000282693 Cercopithecidae Species 0.000 description 1
- 239000004380 Cholic acid Substances 0.000 description 1
- 102100021864 Cocaine esterase Human genes 0.000 description 1
- 241000518994 Conta Species 0.000 description 1
- 206010011224 Cough Diseases 0.000 description 1
- UOOOPKANIPLQPU-UHFFFAOYSA-N Cytidylic acid B Natural products O=C1N=C(N)C=CN1C1C(O)C(OP(O)(O)=O)C(CO)O1 UOOOPKANIPLQPU-UHFFFAOYSA-N 0.000 description 1
- SHZGCJCMOBCMKK-UHFFFAOYSA-N D-mannomethylose Natural products CC1OC(O)C(O)C(O)C1O SHZGCJCMOBCMKK-UHFFFAOYSA-N 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
- 108020004414 DNA Proteins 0.000 description 1
- 102100026662 Delta and Notch-like epidermal growth factor-related receptor Human genes 0.000 description 1
- 102100037986 Dickkopf-related protein 4 Human genes 0.000 description 1
- 101710099554 Dickkopf-related protein 4 Proteins 0.000 description 1
- 108010016626 Dipeptides Proteins 0.000 description 1
- 102100040502 Draxin Human genes 0.000 description 1
- 101150086587 Draxin gene Proteins 0.000 description 1
- 101100234002 Drosophila melanogaster Shal gene Proteins 0.000 description 1
- KCXVZYZYPLLWCC-UHFFFAOYSA-N EDTA Chemical compound OC(=O)CN(CC(O)=O)CCN(CC(O)=O)CC(O)=O KCXVZYZYPLLWCC-UHFFFAOYSA-N 0.000 description 1
- 208000027534 Emotional disease Diseases 0.000 description 1
- 108010042407 Endonucleases Proteins 0.000 description 1
- 102000004533 Endonucleases Human genes 0.000 description 1
- 241000736355 Euthyroides Species 0.000 description 1
- 102100023590 Fibroblast growth factor-binding protein 1 Human genes 0.000 description 1
- 101710094971 Fibroblast growth factor-binding protein 1 Proteins 0.000 description 1
- PNNNRSAQSRJVSB-SLPGGIOYSA-N Fucose Natural products C[C@H](O)[C@@H](O)[C@H](O)[C@H](O)C=O PNNNRSAQSRJVSB-SLPGGIOYSA-N 0.000 description 1
- 208000034826 Genetic Predisposition to Disease Diseases 0.000 description 1
- 229920002527 Glycogen Polymers 0.000 description 1
- 102100034190 Glypican-1 Human genes 0.000 description 1
- 101150085568 HSPB6 gene Proteins 0.000 description 1
- 102100039170 Heat shock protein beta-6 Human genes 0.000 description 1
- 101000924533 Homo sapiens Angiopoietin-2 Proteins 0.000 description 1
- 101000729794 Homo sapiens Beta-1,4-glucuronyltransferase 1 Proteins 0.000 description 1
- 101000933465 Homo sapiens Beta-glucuronidase Proteins 0.000 description 1
- 101000713078 Homo sapiens C-C motif chemokine 24 Proteins 0.000 description 1
- 101000947172 Homo sapiens C-X-C motif chemokine 9 Proteins 0.000 description 1
- 101000914511 Homo sapiens CD27 antigen Proteins 0.000 description 1
- 101000990046 Homo sapiens CMRF35-like molecule 2 Proteins 0.000 description 1
- 101000869010 Homo sapiens Cathepsin D Proteins 0.000 description 1
- 101000898006 Homo sapiens Cocaine esterase Proteins 0.000 description 1
- 101001054266 Homo sapiens Delta and Notch-like epidermal growth factor-related receptor Proteins 0.000 description 1
- 101001070736 Homo sapiens Glypican-1 Proteins 0.000 description 1
- 101001034314 Homo sapiens Lactadherin Proteins 0.000 description 1
- 101001051272 Homo sapiens Layilin Proteins 0.000 description 1
- 101000938676 Homo sapiens Liver carboxylesterase 1 Proteins 0.000 description 1
- 101001051093 Homo sapiens Low-density lipoprotein receptor Proteins 0.000 description 1
- 101000585663 Homo sapiens Myocilin Proteins 0.000 description 1
- 101000617708 Homo sapiens Pregnancy-specific beta-1-glycoprotein 1 Proteins 0.000 description 1
- 101000617725 Homo sapiens Pregnancy-specific beta-1-glycoprotein 2 Proteins 0.000 description 1
- 101000650117 Homo sapiens Protein Wnt-9a Proteins 0.000 description 1
- 101000825954 Homo sapiens R-spondin-1 Proteins 0.000 description 1
- 101000651309 Homo sapiens Retinoic acid receptor responder protein 1 Proteins 0.000 description 1
- 101000617796 Homo sapiens SPARC-related modular calcium-binding protein 1 Proteins 0.000 description 1
- 101000708790 Homo sapiens SPARC-related modular calcium-binding protein 2 Proteins 0.000 description 1
- 101000785978 Homo sapiens Sphingomyelin phosphodiesterase Proteins 0.000 description 1
- 101000642262 Homo sapiens Spondin-1 Proteins 0.000 description 1
- 101000669511 Homo sapiens T-cell immunoglobulin and mucin domain-containing protein 4 Proteins 0.000 description 1
- 101000893741 Homo sapiens Tissue alpha-L-fucosidase Proteins 0.000 description 1
- 101000831496 Homo sapiens Toll-like receptor 3 Proteins 0.000 description 1
- 101000801227 Homo sapiens Tumor necrosis factor receptor superfamily member 19 Proteins 0.000 description 1
- 101000617921 Homo sapiens VPS10 domain-containing receptor SorCS2 Proteins 0.000 description 1
- 101000851018 Homo sapiens Vascular endothelial growth factor receptor 1 Proteins 0.000 description 1
- UGQMRVRMYYASKQ-UHFFFAOYSA-N Hypoxanthine nucleoside Natural products OC1C(O)C(CO)OC1N1C(NC=NC2=O)=C2N=C1 UGQMRVRMYYASKQ-UHFFFAOYSA-N 0.000 description 1
- 206010022061 Injection site erythema Diseases 0.000 description 1
- 206010022086 Injection site pain Diseases 0.000 description 1
- 102000004372 Insulin-like growth factor binding protein 2 Human genes 0.000 description 1
- 108090000964 Insulin-like growth factor binding protein 2 Proteins 0.000 description 1
- 102100039340 Interleukin-18 receptor 1 Human genes 0.000 description 1
- 101710184759 Interleukin-18 receptor 1 Proteins 0.000 description 1
- 102100039078 Interleukin-4 receptor subunit alpha Human genes 0.000 description 1
- 101710169536 Interleukin-4 receptor subunit alpha Proteins 0.000 description 1
- 241000764238 Isis Species 0.000 description 1
- 102100034872 Kallikrein-4 Human genes 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
- SHZGCJCMOBCMKK-DHVFOXMCSA-N L-fucopyranose Chemical compound C[C@@H]1OC(O)[C@@H](O)[C@H](O)[C@@H]1O SHZGCJCMOBCMKK-DHVFOXMCSA-N 0.000 description 1
- 102100039648 Lactadherin Human genes 0.000 description 1
- 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 1
- 102100024621 Layilin Human genes 0.000 description 1
- 108090001090 Lectins Proteins 0.000 description 1
- 102000004856 Lectins Human genes 0.000 description 1
- 102100024640 Low-density lipoprotein receptor Human genes 0.000 description 1
- 102100020983 Lysosome membrane protein 2 Human genes 0.000 description 1
- 101000845005 Macrovipera lebetina Disintegrin lebein-2-alpha Proteins 0.000 description 1
- 241000124008 Mammalia Species 0.000 description 1
- 241000283956 Manis Species 0.000 description 1
- 101710151321 Melanostatin Proteins 0.000 description 1
- 241001068914 Melicope knudsenii Species 0.000 description 1
- LSDPWZHWYPCBBB-UHFFFAOYSA-N Methanethiol Chemical compound SC LSDPWZHWYPCBBB-UHFFFAOYSA-N 0.000 description 1
- 102000015728 Mucins Human genes 0.000 description 1
- 108010063954 Mucins Proteins 0.000 description 1
- 208000001089 Multiple system atrophy Diseases 0.000 description 1
- 241000699670 Mus sp. Species 0.000 description 1
- 102100029839 Myocilin Human genes 0.000 description 1
- 102100036836 Natriuretic peptides B Human genes 0.000 description 1
- 101710187802 Natriuretic peptides B Proteins 0.000 description 1
- 206010028813 Nausea Diseases 0.000 description 1
- 102400000064 Neuropeptide Y Human genes 0.000 description 1
- 241001316086 Novocrania Species 0.000 description 1
- 229910004679 ONO2 Inorganic materials 0.000 description 1
- REYJJPSVUYRZGE-UHFFFAOYSA-N Octadecylamine Chemical compound CCCCCCCCCCCCCCCCCCN REYJJPSVUYRZGE-UHFFFAOYSA-N 0.000 description 1
- 206010030113 Oedema Diseases 0.000 description 1
- 241000083552 Oligomeris Species 0.000 description 1
- 108010038807 Oligopeptides Proteins 0.000 description 1
- 102000015636 Oligopeptides Human genes 0.000 description 1
- 206010031127 Orthostatic hypotension Diseases 0.000 description 1
- 239000007990 PIPES buffer Substances 0.000 description 1
- 208000037273 Pathologic Processes Diseases 0.000 description 1
- 108090000608 Phosphoric Monoester Hydrolases Proteins 0.000 description 1
- 102000004160 Phosphoric Monoester Hydrolases Human genes 0.000 description 1
- ABLZXFCXXLZCGV-UHFFFAOYSA-N Phosphorous acid Chemical class OP(O)=O ABLZXFCXXLZCGV-UHFFFAOYSA-N 0.000 description 1
- 239000002202 Polyethylene glycol Substances 0.000 description 1
- 102100022019 Pregnancy-specific beta-1-glycoprotein 2 Human genes 0.000 description 1
- 102100027503 Protein Wnt-9a Human genes 0.000 description 1
- 101710150593 Protein beta Proteins 0.000 description 1
- 108010026552 Proteome Proteins 0.000 description 1
- 208000003251 Pruritus Diseases 0.000 description 1
- 235000013929 Psidium pyriferum Nutrition 0.000 description 1
- 244000236580 Psidium pyriferum Species 0.000 description 1
- 102000007615 Pulmonary Surfactant-Associated Protein A Human genes 0.000 description 1
- 108010007100 Pulmonary Surfactant-Associated Protein A Proteins 0.000 description 1
- CZPWVGJYEJSRLH-UHFFFAOYSA-N Pyrimidine Chemical compound C1=CN=CN=C1 CZPWVGJYEJSRLH-UHFFFAOYSA-N 0.000 description 1
- 102100022762 R-spondin-1 Human genes 0.000 description 1
- 101000633881 Rattus norvegicus Transthyretin Proteins 0.000 description 1
- 101100208299 Rattus norvegicus Ttr gene Proteins 0.000 description 1
- 102100027682 Retinoic acid receptor responder protein 1 Human genes 0.000 description 1
- 108091005488 SCARB2 Proteins 0.000 description 1
- 102100021995 SPARC-related modular calcium-binding protein 1 Human genes 0.000 description 1
- 102100032724 SPARC-related modular calcium-binding protein 2 Human genes 0.000 description 1
- 235000015076 Shorea robusta Nutrition 0.000 description 1
- 244000166071 Shorea robusta Species 0.000 description 1
- 208000010261 Small Fiber Neuropathy Diseases 0.000 description 1
- 206010073928 Small fibre neuropathy Diseases 0.000 description 1
- 102100026263 Sphingomyelin phosphodiesterase Human genes 0.000 description 1
- 102100036428 Spondin-1 Human genes 0.000 description 1
- 229920002472 Starch Polymers 0.000 description 1
- 101100054666 Streptomyces halstedii sch3 gene Proteins 0.000 description 1
- NINIDFKCEFEMDL-UHFFFAOYSA-N Sulfur Chemical compound [S] NINIDFKCEFEMDL-UHFFFAOYSA-N 0.000 description 1
- UCKMPCXJQFINFW-UHFFFAOYSA-N Sulphide Chemical compound [S-2] UCKMPCXJQFINFW-UHFFFAOYSA-N 0.000 description 1
- 102100039367 T-cell immunoglobulin and mucin domain-containing protein 4 Human genes 0.000 description 1
- 101150057833 THEG gene Proteins 0.000 description 1
- RYYWUUFWQRZTIU-UHFFFAOYSA-N Thiophosphoric acid Chemical class OP(O)(S)=O RYYWUUFWQRZTIU-UHFFFAOYSA-N 0.000 description 1
- 102000011923 Thyrotropin Human genes 0.000 description 1
- 108010061174 Thyrotropin Proteins 0.000 description 1
- 102100040526 Tissue alpha-L-fucosidase Human genes 0.000 description 1
- 102100024324 Toll-like receptor 3 Human genes 0.000 description 1
- 108091032917 Transfer-messenger RNA Proteins 0.000 description 1
- 102000004338 Transferrin Human genes 0.000 description 1
- 108090000901 Transferrin Proteins 0.000 description 1
- 108050000089 Transthyretin Proteins 0.000 description 1
- 102000014456 Trefoil Factor-3 Human genes 0.000 description 1
- 108010078184 Trefoil Factor-3 Proteins 0.000 description 1
- 239000007983 Tris buffer Substances 0.000 description 1
- 102100033760 Tumor necrosis factor receptor superfamily member 19 Human genes 0.000 description 1
- FOGRQMPFHUHIGU-UHFFFAOYSA-N Uridylic acid Natural products OC1C(OP(O)(O)=O)C(CO)OC1N1C(=O)NC(=O)C=C1 FOGRQMPFHUHIGU-UHFFFAOYSA-N 0.000 description 1
- 102100021938 VPS10 domain-containing receptor SorCS2 Human genes 0.000 description 1
- 102100033178 Vascular endothelial growth factor receptor 1 Human genes 0.000 description 1
- 229930003779 Vitamin B12 Natural products 0.000 description 1
- 102000021095 WAP Four-Disulfide Core Domain Protein 2 Human genes 0.000 description 1
- 108091002660 WAP Four-Disulfide Core Domain Protein 2 Proteins 0.000 description 1
- 239000005862 Whey Substances 0.000 description 1
- 108010046377 Whey Proteins Proteins 0.000 description 1
- 102000007544 Whey Proteins Human genes 0.000 description 1
- VZKAGFPIGOSWMA-IOSLPCCCSA-N [(2r,3r,4r,5r)-5-(6-aminopurin-9-yl)-3-dihydroxyphosphinothioyloxy-4-methoxyoxolan-2-yl]methanol Chemical compound CO[C@@H]1[C@H](OP(O)(O)=S)[C@@H](CO)O[C@H]1N1C2=NC=NC(N)=C2N=C1 VZKAGFPIGOSWMA-IOSLPCCCSA-N 0.000 description 1
- DUDZRBVSHCOKLU-LTNPLRIYSA-N [(2r,3r,4s,5r)-4-fluoro-4-hydroxy-2-(hydroxymethyl)-5-(5-methyl-2,4-dioxopyrimidin-1-yl)oxolan-3-yl] dihydrogen phosphate Chemical compound O=C1NC(=O)C(C)=CN1[C@H]1[C@](F)(O)[C@H](OP(O)(O)=O)[C@@H](CO)O1 DUDZRBVSHCOKLU-LTNPLRIYSA-N 0.000 description 1
- XVIYCJDWYLJQBG-UHFFFAOYSA-N acetic acid;adamantane Chemical compound CC(O)=O.C1C(C2)CC3CC1CC2C3 XVIYCJDWYLJQBG-UHFFFAOYSA-N 0.000 description 1
- 230000004913 activation Effects 0.000 description 1
- 125000002252 acyl group Chemical group 0.000 description 1
- 125000001931 aliphatic group Chemical group 0.000 description 1
- 125000005217 alkenylheteroaryl group Chemical group 0.000 description 1
- 125000003545 alkoxy group Chemical group 0.000 description 1
- 125000005083 alkoxyalkoxy group Chemical group 0.000 description 1
- 125000002877 alkyl aryl group Chemical group 0.000 description 1
- 125000005213 alkyl heteroaryl group Chemical group 0.000 description 1
- 125000004419 alkynylene group Chemical group 0.000 description 1
- 239000012491 analyte Substances 0.000 description 1
- 238000013459 approach Methods 0.000 description 1
- 230000006793 arrhythmia Effects 0.000 description 1
- 125000005015 aryl alkynyl group Chemical group 0.000 description 1
- 229940009098 aspartate Drugs 0.000 description 1
- 230000001746 atrial effect Effects 0.000 description 1
- 230000037424 autonomic function Effects 0.000 description 1
- 230000009910 autonomic response Effects 0.000 description 1
- 230000037429 base substitution Effects 0.000 description 1
- DRTQHJPVMGBUCF-PSQAKQOGSA-N beta-L-uridine Natural products O[C@H]1[C@@H](O)[C@H](CO)O[C@@H]1N1C(=O)NC(=O)C=C1 DRTQHJPVMGBUCF-PSQAKQOGSA-N 0.000 description 1
- 230000002146 bilateral effect Effects 0.000 description 1
- 239000003613 bile acid Substances 0.000 description 1
- 239000013060 biological fluid Substances 0.000 description 1
- 238000001574 biopsy Methods 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
- 230000036772 blood pressure Effects 0.000 description 1
- 210000004556 brain Anatomy 0.000 description 1
- 150000001721 carbon Chemical group 0.000 description 1
- 210000004671 cell-free system Anatomy 0.000 description 1
- 230000001413 cellular effect Effects 0.000 description 1
- 229920002678 cellulose Polymers 0.000 description 1
- 239000001913 cellulose Substances 0.000 description 1
- 238000007385 chemical modification Methods 0.000 description 1
- 239000003153 chemical reaction reagent Substances 0.000 description 1
- BHQCQFFYRZLCQQ-OELDTZBJSA-N cholic acid Chemical compound C([C@H]1C[C@H]2O)[C@H](O)CC[C@]1(C)[C@@H]1[C@@H]2[C@@H]2CC[C@H]([C@@H](CCC(O)=O)C)[C@@]2(C)[C@@H](O)C1 BHQCQFFYRZLCQQ-OELDTZBJSA-N 0.000 description 1
- 229960002471 cholic acid Drugs 0.000 description 1
- 235000019416 cholic acid Nutrition 0.000 description 1
- AGVAZMGAQJOSFJ-WZHZPDAFSA-M cobalt(2+);[(2r,3s,4r,5s)-5-(5,6-dimethylbenzimidazol-1-yl)-4-hydroxy-2-(hydroxymethyl)oxolan-3-yl] [(2r)-1-[3-[(1r,2r,3r,4z,7s,9z,12s,13s,14z,17s,18s,19r)-2,13,18-tris(2-amino-2-oxoethyl)-7,12,17-tris(3-amino-3-oxopropyl)-3,5,8,8,13,15,18,19-octamethyl-2 Chemical compound [Co+2].N#[C-].[N-]([C@@H]1[C@H](CC(N)=O)[C@@]2(C)CCC(=O)NC[C@@H](C)OP(O)(=O)O[C@H]3[C@H]([C@H](O[C@@H]3CO)N3C4=CC(C)=C(C)C=C4N=C3)O)\C2=C(C)/C([C@H](C\2(C)C)CCC(N)=O)=N/C/2=C\C([C@H]([C@@]/2(CC(N)=O)C)CCC(N)=O)=N\C\2=C(C)/C2=N[C@]1(C)[C@@](C)(CC(N)=O)[C@@H]2CCC(N)=O AGVAZMGAQJOSFJ-WZHZPDAFSA-M 0.000 description 1
- 230000001268 conjugating effect Effects 0.000 description 1
- 239000013068 control sample Substances 0.000 description 1
- 210000003792 cranial nerve Anatomy 0.000 description 1
- 239000013078 crystal Substances 0.000 description 1
- ZDPUTNZENXVHJC-UHFFFAOYSA-N cumingianoside D Natural products C1=2NC(N)=NC(=O)C=2N=CN1C1OC(CO)C(OP(O)(O)=O)C1O ZDPUTNZENXVHJC-UHFFFAOYSA-N 0.000 description 1
- 108010032220 cyclomaltodextrinase Proteins 0.000 description 1
- KXGVEGMKQFWNSR-UHFFFAOYSA-N deoxycholic acid Natural products C1CC2CC(O)CCC2(C)C2C1C1CCC(C(CCC(O)=O)C)C1(C)C(O)C2 KXGVEGMKQFWNSR-UHFFFAOYSA-N 0.000 description 1
- 230000000368 destabilizing effect Effects 0.000 description 1
- 235000005911 diet Nutrition 0.000 description 1
- 230000037213 diet Effects 0.000 description 1
- 208000037765 diseases and disorders Diseases 0.000 description 1
- 238000010494 dissociation reaction Methods 0.000 description 1
- 230000005593 dissociations Effects 0.000 description 1
- NAGJZTKCGNOGPW-UHFFFAOYSA-N dithiophosphoric acid Chemical class OP(O)(S)=S NAGJZTKCGNOGPW-UHFFFAOYSA-N 0.000 description 1
- 239000002934 diuretic Substances 0.000 description 1
- 229940030606 diuretics Drugs 0.000 description 1
- 231100000673 dose–response relationship Toxicity 0.000 description 1
- 125000004185 ester group Chemical group 0.000 description 1
- 125000001495 ethyl group Chemical group [H]C([H])([H])C([H])([H])* 0.000 description 1
- 206010016256 fatigue Diseases 0.000 description 1
- 239000000835 fiber 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
- 125000000524 functional group Chemical group 0.000 description 1
- 125000003843 furanosyl group Chemical group 0.000 description 1
- 230000005021 gait Effects 0.000 description 1
- 238000012226 gene silencing method Methods 0.000 description 1
- 150000004676 glycans Chemical class 0.000 description 1
- 229940096919 glycogen Drugs 0.000 description 1
- ZDPUTNZENXVHJC-UUOKFMHZSA-N guanosine 3'-monophosphate Chemical compound C1=NC=2C(=O)NC(N)=NC=2N1[C@@H]1O[C@H](CO)[C@@H](OP(O)(O)=O)[C@H]1O ZDPUTNZENXVHJC-UUOKFMHZSA-N 0.000 description 1
- 210000001255 hallux Anatomy 0.000 description 1
- 210000003128 head Anatomy 0.000 description 1
- 230000002489 hematologic effect Effects 0.000 description 1
- 125000001072 heteroaryl group Chemical group 0.000 description 1
- 125000004447 heteroarylalkenyl group Chemical group 0.000 description 1
- 125000004446 heteroarylalkyl group Chemical group 0.000 description 1
- 125000005312 heteroarylalkynyl group Chemical group 0.000 description 1
- 125000000623 heterocyclic group Chemical group 0.000 description 1
- 125000000592 heterocycloalkyl group Chemical group 0.000 description 1
- 150000007857 hydrazones Chemical class 0.000 description 1
- 150000002431 hydrogen Chemical group 0.000 description 1
- 238000003384 imaging method Methods 0.000 description 1
- 238000001427 incoherent neutron scattering Methods 0.000 description 1
- 238000010348 incorporation Methods 0.000 description 1
- 238000011534 incubation Methods 0.000 description 1
- 230000008595 infiltration Effects 0.000 description 1
- 238000001764 infiltration Methods 0.000 description 1
- 238000001802 infusion Methods 0.000 description 1
- 229950002218 inotersen Drugs 0.000 description 1
- 238000010255 intramuscular injection Methods 0.000 description 1
- 239000007927 intramuscular injection Substances 0.000 description 1
- 125000001449 isopropyl group Chemical group [H]C([H])([H])C([H])(*)C([H])([H])[H] 0.000 description 1
- 108010024383 kallikrein 4 Proteins 0.000 description 1
- 210000003292 kidney cell Anatomy 0.000 description 1
- 230000003907 kidney function Effects 0.000 description 1
- 239000008101 lactose Substances 0.000 description 1
- 239000002523 lectin Substances 0.000 description 1
- 230000000670 limiting effect Effects 0.000 description 1
- 230000003908 liver function Effects 0.000 description 1
- 210000005228 liver tissue Anatomy 0.000 description 1
- 230000005923 long-lasting effect Effects 0.000 description 1
- 206010025005 lumbar spinal stenosis Diseases 0.000 description 1
- 210000004072 lung Anatomy 0.000 description 1
- 210000002751 lymph Anatomy 0.000 description 1
- 238000007726 management method Methods 0.000 description 1
- 239000003550 marker Substances 0.000 description 1
- 230000007246 mechanism Effects 0.000 description 1
- 125000004573 morpholin-4-yl group Chemical group N1(CCOCC1)* 0.000 description 1
- 230000002107 myocardial effect Effects 0.000 description 1
- QVRVXSZKCXFBTE-UHFFFAOYSA-N n-[4-(6,7-dimethoxy-3,4-dihydro-1h-isoquinolin-2-yl)butyl]-2-(2-fluoroethoxy)-5-methylbenzamide Chemical compound C1C=2C=C(OC)C(OC)=CC=2CCN1CCCCNC(=O)C1=CC(C)=CC=C1OCCF QVRVXSZKCXFBTE-UHFFFAOYSA-N 0.000 description 1
- 230000008693 nausea Effects 0.000 description 1
- 210000004126 nerve fiber Anatomy 0.000 description 1
- 230000001537 neural effect Effects 0.000 description 1
- 230000007658 neurological function Effects 0.000 description 1
- 230000003961 neuronal insult Effects 0.000 description 1
- QJGQUHMNIGDVPM-UHFFFAOYSA-N nitrogen group Chemical group [N] QJGQUHMNIGDVPM-UHFFFAOYSA-N 0.000 description 1
- 125000001893 nitrooxy group Chemical group [O-][N+](=O)O* 0.000 description 1
- URPYMXQQVHTUDU-OFGSCBOVSA-N nucleopeptide y Chemical compound C([C@@H](C(=O)N[C@@H]([C@@H](C)CC)C(=O)N[C@@H](CC(N)=O)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H]([C@@H](C)CC)C(=O)N[C@@H]([C@@H](C)O)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CCC(N)=O)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CC=1C=CC(O)=CC=1)C(N)=O)NC(=O)[C@H](CC=1NC=NC=1)NC(=O)[C@H](CCCNC(N)=N)NC(=O)[C@H](CC(C)C)NC(=O)[C@H](C)NC(=O)[C@H](CO)NC(=O)[C@H](CC=1C=CC(O)=CC=1)NC(=O)[C@H](CC=1C=CC(O)=CC=1)NC(=O)[C@H](CCCNC(N)=N)NC(=O)[C@H](C)NC(=O)[C@H](CC(C)C)NC(=O)[C@H](CC(O)=O)NC(=O)[C@H](CCC(O)=O)NC(=O)[C@H](C)NC(=O)[C@H]1N(CCC1)C(=O)[C@H](C)NC(=O)[C@H](CC(O)=O)NC(=O)[C@H](CCC(O)=O)NC(=O)CNC(=O)[C@H]1N(CCC1)C(=O)[C@H](CC(N)=O)NC(=O)[C@H](CC(O)=O)NC(=O)[C@H]1N(CCC1)C(=O)[C@H](CCCCN)NC(=O)[C@H](CO)NC(=O)[C@H]1N(CCC1)C(=O)[C@@H](N)CC=1C=CC(O)=CC=1)C1=CC=C(O)C=C1 URPYMXQQVHTUDU-OFGSCBOVSA-N 0.000 description 1
- 235000003715 nutritional status Nutrition 0.000 description 1
- 230000009437 off-target effect Effects 0.000 description 1
- 210000003463 organelle Anatomy 0.000 description 1
- 125000001181 organosilyl group Chemical group [SiH3]* 0.000 description 1
- 230000003647 oxidation Effects 0.000 description 1
- 238000007254 oxidation reaction Methods 0.000 description 1
- 230000001590 oxidative effect Effects 0.000 description 1
- 125000000913 palmityl group Chemical group [H]C([*])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])[H] 0.000 description 1
- 230000009054 pathological process Effects 0.000 description 1
- 230000002093 peripheral effect Effects 0.000 description 1
- 239000002831 pharmacologic agent Substances 0.000 description 1
- 150000004713 phosphodiesters Chemical class 0.000 description 1
- 150000003904 phospholipids Chemical class 0.000 description 1
- PTMHPRAIXMAOOB-UHFFFAOYSA-N phosphoramidic acid Chemical compound NP(O)(O)=O PTMHPRAIXMAOOB-UHFFFAOYSA-N 0.000 description 1
- 230000026731 phosphorylation Effects 0.000 description 1
- 238000006366 phosphorylation reaction Methods 0.000 description 1
- 229920000768 polyamine Polymers 0.000 description 1
- 108010064470 polyaspartate Proteins 0.000 description 1
- 229920001223 polyethylene glycol Polymers 0.000 description 1
- 229920001184 polypeptide Polymers 0.000 description 1
- 229920001282 polysaccharide Polymers 0.000 description 1
- 239000005017 polysaccharide Substances 0.000 description 1
- 230000032361 posttranscriptional gene silencing Effects 0.000 description 1
- 238000002203 pretreatment Methods 0.000 description 1
- 230000002265 prevention Effects 0.000 description 1
- 208000017692 primary erythermalgia Diseases 0.000 description 1
- 238000004393 prognosis Methods 0.000 description 1
- 125000006239 protecting group Chemical group 0.000 description 1
- 230000009430 psychological distress Effects 0.000 description 1
- 230000010490 psychological well-being Effects 0.000 description 1
- 150000003212 purines Chemical class 0.000 description 1
- 230000001698 pyrogenic effect Effects 0.000 description 1
- 238000003753 real-time PCR Methods 0.000 description 1
- 239000002265 redox agent Substances 0.000 description 1
- 239000013074 reference sample Substances 0.000 description 1
- 125000006853 reporter group Chemical group 0.000 description 1
- 230000000717 retained effect Effects 0.000 description 1
- DWRXFEITVBNRMK-JXOAFFINSA-N ribothymidine Chemical compound O=C1NC(=O)C(C)=CN1[C@H]1[C@H](O)[C@H](O)[C@@H](CO)O1 DWRXFEITVBNRMK-JXOAFFINSA-N 0.000 description 1
- 210000003296 saliva Anatomy 0.000 description 1
- 238000013077 scoring method Methods 0.000 description 1
- 210000004911 serous fluid Anatomy 0.000 description 1
- 239000011780 sodium chloride Substances 0.000 description 1
- QPILZZVXGUNELN-UHFFFAOYSA-M sodium;4-amino-5-hydroxynaphthalene-2,7-disulfonate;hydron Chemical compound [Na+].OS(=O)(=O)C1=CC(O)=C2C(N)=CC(S([O-])(=O)=O)=CC2=C1 QPILZZVXGUNELN-UHFFFAOYSA-M 0.000 description 1
- 238000010532 solid phase synthesis reaction Methods 0.000 description 1
- 230000037439 somatic mutation Effects 0.000 description 1
- 241000894007 species Species 0.000 description 1
- 238000001228 spectrum Methods 0.000 description 1
- 208000005198 spinal stenosis Diseases 0.000 description 1
- 230000000087 stabilizing effect Effects 0.000 description 1
- 238000010561 standard procedure Methods 0.000 description 1
- 235000019698 starch Nutrition 0.000 description 1
- 238000007619 statistical method Methods 0.000 description 1
- 239000008223 sterile water Substances 0.000 description 1
- 239000008227 sterile water for injection Substances 0.000 description 1
- 150000003431 steroids Chemical class 0.000 description 1
- 238000003756 stirring Methods 0.000 description 1
- 230000035882 stress Effects 0.000 description 1
- 125000001424 substituent group Chemical group 0.000 description 1
- 125000000547 substituted alkyl group Chemical group 0.000 description 1
- BDHFUVZGWQCTTF-UHFFFAOYSA-M sulfonate Chemical compound [O-]S(=O)=O BDHFUVZGWQCTTF-UHFFFAOYSA-M 0.000 description 1
- 150000003457 sulfones Chemical group 0.000 description 1
- 150000003462 sulfoxides Chemical class 0.000 description 1
- 239000011593 sulfur Substances 0.000 description 1
- 210000001590 sural nerve Anatomy 0.000 description 1
- 238000001356 surgical procedure Methods 0.000 description 1
- 230000004083 survival effect Effects 0.000 description 1
- 210000002437 synoviocyte Anatomy 0.000 description 1
- 238000007910 systemic administration Methods 0.000 description 1
- DQJDBUPLRMRBAB-WZTVWXICSA-N tafamidis meglumine Chemical compound CNC[C@H](O)[C@@H](O)[C@H](O)[C@H](O)CO.O1C2=CC(C(=O)O)=CC=C2N=C1C1=CC(Cl)=CC(Cl)=C1 DQJDBUPLRMRBAB-WZTVWXICSA-N 0.000 description 1
- 229910052713 technetium Inorganic materials 0.000 description 1
- GKLVYJBZJHMRIY-UHFFFAOYSA-N technetium atom Chemical compound [Tc] GKLVYJBZJHMRIY-UHFFFAOYSA-N 0.000 description 1
- 125000000999 tert-butyl group Chemical group [H]C([H])([H])C(*)(C([H])([H])[H])C([H])([H])[H] 0.000 description 1
- 210000001550 testis Anatomy 0.000 description 1
- 238000002560 therapeutic procedure Methods 0.000 description 1
- 150000003568 thioethers Chemical class 0.000 description 1
- 229960000874 thyrotropin Drugs 0.000 description 1
- 230000001748 thyrotropin Effects 0.000 description 1
- 210000002972 tibial nerve Anatomy 0.000 description 1
- 210000003371 toe Anatomy 0.000 description 1
- 239000012581 transferrin Substances 0.000 description 1
- STCOOQWBFONSKY-UHFFFAOYSA-N tributyl phosphate Chemical compound CCCCOP(=O)(OCCCC)OCCCC STCOOQWBFONSKY-UHFFFAOYSA-N 0.000 description 1
- 125000000876 trifluoromethoxy group Chemical group FC(F)(F)O* 0.000 description 1
- 125000002023 trifluoromethyl group Chemical group FC(F)(F)* 0.000 description 1
- 150000004043 trisaccharides Chemical class 0.000 description 1
- 210000000658 ulnar nerve Anatomy 0.000 description 1
- 125000002948 undecyl group Chemical group [H]C([*])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])[H] 0.000 description 1
- 210000001364 upper extremity Anatomy 0.000 description 1
- 210000000689 upper leg Anatomy 0.000 description 1
- DRTQHJPVMGBUCF-UHFFFAOYSA-N uracil arabinoside Natural products OC1C(O)C(CO)OC1N1C(=O)NC(=O)C=C1 DRTQHJPVMGBUCF-UHFFFAOYSA-N 0.000 description 1
- 229940045145 uridine Drugs 0.000 description 1
- 210000002700 urine Anatomy 0.000 description 1
- 235000019163 vitamin B12 Nutrition 0.000 description 1
- 239000011715 vitamin B12 Substances 0.000 description 1
- 238000005406 washing Methods 0.000 description 1
- 229940075420 xanthine Drugs 0.000 description 1
Classifications
-
- 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
- C12N15/113—Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/70—Carbohydrates; Sugars; Derivatives thereof
- A61K31/7088—Compounds having three or more nucleosides or nucleotides
- A61K31/713—Double-stranded nucleic acids or oligonucleotides
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P25/00—Drugs for disorders of the nervous system
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P9/00—Drugs for disorders of the cardiovascular system
-
- 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
- C12N2310/32—Chemical structure of the sugar
-
- 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
- C12N2310/32—Chemical structure of the sugar
- C12N2310/321—2'-O-R Modification
-
- 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
- C12N2310/32—Chemical structure of the sugar
- C12N2310/322—2'-R Modification
-
- 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
- C12N2310/34—Spatial arrangement of the modifications
- C12N2310/344—Position-specific modifications, e.g. on every purine, at the 3'-end
-
- 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
- C12N2310/352—Nature of the modification linked to the nucleic acid via a carbon atom
- C12N2310/3521—Methyl
-
- 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
- C12N2310/353—Nature of the modification linked to the nucleic acid via an atom other than carbon
- C12N2310/3533—Halogen
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Engineering & Computer Science (AREA)
- Genetics & Genomics (AREA)
- Chemical & Material Sciences (AREA)
- Biomedical Technology (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Organic Chemistry (AREA)
- Molecular Biology (AREA)
- General Health & Medical Sciences (AREA)
- General Engineering & Computer Science (AREA)
- Wood Science & Technology (AREA)
- Zoology (AREA)
- Biotechnology (AREA)
- Biochemistry (AREA)
- Medicinal Chemistry (AREA)
- Veterinary Medicine (AREA)
- Public Health (AREA)
- Animal Behavior & Ethology (AREA)
- Pharmacology & Pharmacy (AREA)
- Biophysics (AREA)
- Plant Pathology (AREA)
- Microbiology (AREA)
- Physics & Mathematics (AREA)
- General Chemical & Material Sciences (AREA)
- Chemical Kinetics & Catalysis (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Cardiology (AREA)
- Neurology (AREA)
- Neurosurgery (AREA)
- Heart & Thoracic Surgery (AREA)
- Epidemiology (AREA)
- Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
- Saccharide Compounds (AREA)
- Heterocyclic Carbon Compounds Containing A Hetero Ring Having Oxygen Or Sulfur (AREA)
- Medicinal Preparation (AREA)
- Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
Description
COMPOSITIONS AND METHODS FOR INHIBITING EXPRESSION OF TRANSTHYRETIN (TTR) Related Applications The present application claims the benefi tof priority to U.S. Provisional Application No. 62/985,950, filed on March 6, 2020, the entire contents of which are incorporat edherein by reference.
Sequence Listing The instant application contains a Sequence Listing which has been submitted electronica inlly ASCII forma tand is hereby incorporat edby reference in its entirety. Said ASCII copy, created on February 26, 2021, is named 121301_12320_SL.txt and is 44,455 bytes in size.
Background Transthyretin (TTR) (also known as prealbumin) is found in serum and cerebrospinal fluid (CSF). TTR transports retinol-bindi ngprotein (RBP) and thyroxine (T4) and also acts as a carrier of retinol (vitamin A) through its association with RBP in the blood and the CSF. Transthyretin is named for its transport of thyroxine and retinol. TTR also functions as a protease and can cleave proteins including apoA-I (the major HDL apolipoprotein), amyloid -peptide, and neuropeptide Y. See Liz, M.A. et al. (2010) lUBMBLife, 62(6):429-435.
TTR is a tetramer of four identical 127-amino acid subunits (monomers) that are rich in beta sheet structure. Each monomer has two 4-stranded beta sheets and the shape of a prolate ellipsoi d.
Antiparall beta-sheetel interactions link monomers into dimers. A short loop from each monome formr s the main dimer-dimer interaction. These two pairs of loops separate the opposed, convex beta-sheet ofs the dimers to form an internal channel.
The liver is the major site of TTR expression. Other significant sites of expression include the choro idplexus, retina (particularl they retinal pigment epithelium )and pancreas.
Transthyretin is one of at leas 27t distinct types of proteins that is a precursor protein in the formation of amyloid fibrils. See Guan, J. et al. (Nov. 4, 2011) Current perspectives on cardiac amyloidosi Ams, J Physiol Heart Circ Physiol, doi: 10.1152/ajpheart.00815.2011. Extracellular deposition of amyloid fibrils in organs and tissues is the hallmark of amyloidosi Amyls. oid fibrils are compose dof misfolded protein aggregates ,which may result from either excess production of or specific mutations in precurso rproteins. The amyloidogenic potential of TTR may be related to its extensive beta sheet structure; X-ray crystallographi studiec s indicate that certain amyloidogenic mutations destabilize the tetrameric structure of the protein. See, e.g., Saraiva M.J.M. (2002) Expert Reviews in Molecular Medicine, 4(12):1-11.
Amyloidosi iss a general term for the group of amyloid diseases that are characterized by amyloid deposits. Amyloid diseases are classified based on their precursor protein; for example, the name starts with "A" for amyloid and is followed by an abbreviation of the precurso rprotein, e.g., ATTR for amloidogenic transthyretin. Ibid. 1 There are numerous TTR-associate dised ases ,most of which are amyloid diseases. Normal- sequenc eTTR is associated with cardiac amyloidos inis peopl ewho are elderly and is termed senil e systemic amyloido sis(SSA) (also called senile cardiac amyloido sis(SCA) or cardiac amyloidosis).
SSA often is accompanie byd microscopi depositsc in many othe rorgans. TTR amyloidosi manis fest s in various forms. When the peripheral nervous system is affected more prominentl y,the disease is termed familial amyloidoti polyc neuropat hy(FAP). When the heart is primaril yinvolved but the nervous system is not, the disease is called familial amyloidot cardiic omyopathy (FAC). A third major type of TTR amyloido sisis leptomeningea amyll oidosi alsos, known as leptomeningea orl meningocerebrovascul amyloar idosi cents, ral nervous system (CNS) amyloidosi ors, amyloidosi VIIs form. Mutation ins TTR may also cause amyloidotic vitreous opacitie s,carpal tunnel syndrome, and euthyroid hyperthyroxinemia, which is a non-amyloidoti diseasec thought to be secondary to an increased association of thyroxine with TTR due to a mutant TTR molecule with increased affinity for thyroxine. See, e.g., Moses et al. (1982) J. Clin. Invest., 86, 2025-2033.
Abnorma TTRl alleles may be either inherited or acquired through somatic mutations. Guan, J. et al. (Nov. 4, 2011) Current perspectives on cardiac amyloidosi s,Am J Physiol Heart Circ Physiol, doi:10.1152/ajpheart.00815.2011. Transthyretin associated ATTR is the most frequent form of hereditary systemic amyloidosi s.Lobato, L. (2003) J. Nephrol., 16:438-442. TTR mutations accelerate the process of TTR amyloid formation and are the most important risk factor for the development of ATTR. More than 85 amyloidogenic TTR variants are known to cause systemic familial amyloidos is.
TTR mutations usually give rise to systemic amyloi depositid on, with particul arinvolvement of the peripheral nervous system, although some mutations are associated with cardiomyopathy or vitreous opacities. Ibid.
The V30M mutation is the most prevalent TTR mutation. See, e.g., Lobato, L. (2003) J Nephrol, 16:438-442. The V1221 mutation is carried by 3.9% of the African American population and is the most common cause of FAC. Jacobson D.R., et al. (1997) N. Engl. J. Med. 336 (7): 466-73. It is estimated that SSA affect smore than 25% of the populatio overn age 80. Westermark, P. et al. (1990) Proc. Natl. Acad. Sci. U.S.A. 87 (7): 2843-5.
Accordingly, there is a need in the art for effective treatments for TTR-associated diseases.
Summary of the Invention The present invention provides compositio nsand methods for inhibiting expression of TTR and methods of treating or preventing a transthyretin- (TTR-) associate diseased in a human subjec tusing double stranded RNAi agents ,targeting the TTR gene.
The invention provides a double stranded RNAi agent comprising a sense strand and an antisense strand ,wherein: eac hthe sense strand and the antisense strand are independentl upy to 30 nucleotides in length; the sense strand comprises the modified nucleotid sequence e5’- usgsggauUfuCfAfUfguaaccaa‘ga-3 (SEQ ID NO: 6); and 2 the antisense strand comprises the modified nucleotid sequee nce 5’- usCfsuugGf(Tgn)uAfcaugAfaAfucccasusc-3’ (SEQ ID NO: 7), wherein a, c, g, andu are 2'-O-methyladenosine-3’-phosphate, 2'-O-methylcytidine-3’- phosphate, 2'-O-methylguanosine-3’-phosphate, and 2'-O-methyluridine-3’-phosphat respectie, vely; Af, Cf, Gf, and Uf are 2‘-fluoroadenosine-3‘ -phosphate, 2‘-fluorocytidine-3‘ -phosphate, 2’- fluoroguanosine-3’-phosphat e,and 2’-fluorouridine’-3-phosphat e,respectively; (Tgn) is thymidine-glycol nucleic acid (GNA) S-Isomer; and s is a phosphorothioa linteker.
In certai nembodiments, the sense strand of the double stranded RNAi agent is conjugat edto at least one ligand. In certain embodiments, the ligand is one or more GalNAc derivatives attached through a bivalent or tri valent branched linker. In certain embodiments, the ligand is In certai nembodiments, the ligand is attache tod the 3' end of the sense strand.
In certai nembodiments, the double stranded RNAi agent is conjugat edto the ligand as shown in the followin schematg ic: 3' wherein X is O or S.
In certai nembodiments, the sense strand is 21 nucleotide ins lengt hand the antisense strand is 23 nucleotide ins length.
The invention provides a use of a double stranded RNAi agen tin a method of treating a human subject suffering from a TTR-associated disease, comprising administration of a fixed dose of about 25 mg to about 1000 mg of a double stranded RNAi agent, wherein: eac hthe sense strand and the antisense strand are independentl upy to 30 nucleotides in length; 3 the sense strand comprises the modified nucleotid sequence e5’- usgsggauUfuCfAfUfguaaccaaga-3’ (SEQ ID NO: 6); and the antisense strand comprises the modified nucleotid sequee nce 5’- usCfsuugGf(Tgn)uAfcaugAfaAfucccasusc-3’ (SEQ ID NO: 7), wherein a, c, g, andu are 2'-O-methyladenosine-3’ -phosphate, 2'-O-methylcytidine-3’- phosphate, 2'-O-methylguanosine-3’ -phosphate, and 2'-O-methyluridine-3’-phosphat respectie, vely; Af, Cf, Of, and Uf are 2‘-fluoroadenosine-3‘ -phosphate, 2‘-fluorocytidine-3‘ -phosphate, 2’- fluoroguanosine-3’-phosphat e,and 2’-fluorouridine’-3-phosphat e,respectively; (Tgn) is thymidine-glycol nucleic acid (GNA) S-Isomer; and s is a phosphorothioa linteker.
The invention also provides a use of a double stranded RNAi agen tin a method of inhibiting expression of TTR in a human subject who does not meet diagnosti cric teria of a TTR-associate d disease ,comprising administration of a fixed dose of about 25 mg to about 1000 mg of a double stranded RNAi agent ,wherein: eac hthe sense strand and the antisense strand are independentl upy to 30 nucleotides in length; the sense strand comprises the modified nucleotid sequence e5’- usgsggauUfuCfAfUfguaaccaaga-3‘ (SEQ ID NO: 6); and the antisense strand comprises the modified nucleotid sequee nce 5’- usCfsuugGf(Tgn)uAfcaugAfaAfucccasusc-3’ (SEQ ID NO: 7), wherein a, c, g, andu are 2'-O-methyladenosine-3’-phosphate, 2'-O-methylcytidine-3’- phosphate, 2'-O-methylguanosine-3’ -phosphate, and 2'-O-methyluridine-3’-phosphat respectie, vely; Af, Cf, Gf, and Uf are 2‘-fluoroadenosine-3‘ -phosphate, 2‘-fluorocytidine-3‘ -phosphate, 2’- fluoroguanosine-3’-phosphat e,and 2’-fluorouridine’-3-phosphat e,respectively; (Tgn) is thymidine-glycol nucleic acid (GNA) S-Isomer; and s is a phosphorothioa linteker.
In certai nembodiments, the sense strand of the double stranded RNAi agent is conjugat edto at least one ligand. In certain embodiments, the ligand is one or more GalNAc derivatives attached through a bivalent or tri valent branched linker. In certai nembodiments, the ligand is HO Z°H 4 In certai nembodiments, the ligand is attached to the 3' end of the sense strand. In certain embodiments, the double stranded RNAi agen tis conjugat edto the ligand as shown in the following schematic 3' wherein X is O or S.
In certai nembodiments, the sense strand is 21 nucleotide ins lengt hand the antisense strand is 23 nucleotide ins length.
In certain embodiments, the uses of the invention comprise improving at least one indici aof neurological impairement, qualit ofy life, nerve damage, cardiovascula health.r In certain embodiments, the indici aassessed is a neurologic impaal irment, for example, using a Neuropathy Impairment (NIS) score or a modified NIS (mNIS+7) score. In certai nembodiments, the indici ais a qualit ofy life indicia assessed, for example, using a SF-36® health survey score, a Norfolk Qualit yof Life-Diabetic Neuropath y(Norfolk QOL-DN) score, a NIS-W score, a Rasch-built Overall Disabilit yScale (R-ODS) score, a composit eautonom icsymptom score (COMPASS-31), a median body mass index (mBMI) score, a 6-minute wal ktest (6MWT) score, and a 10-meter walk test score. In certain embodiments, the indicia is nerve damage assessed, for example, a change in the level of one or more proteins selected from the group neurofilament light chai n(NfL), RSPO3, CCDC80, EDA2R, NT-proBNP, and N- CDase, such as a human blood sample, or serum or plasm aderived therefrom. In certai nembodiments, the indici aof nerve damage is a change from baseline in the level of neurofilament light chai n(NfL) protein level In. certain embodiments, the indici aof cardiovascular impairment is cardiovascular hospitalizatio usinn, g Kansas City Cardiomyopathy Questionnaire Overall Summary (KCCQ-OS) with an increased score indicative of better healt stah tus, change from baseline in mean left !ventricular (LV) wall thickness by echocardiographic assessment ,change from baseline in globa longl itudin alstrain by echocardiographic assessment ,and change from baseline in N-terminal prohormone B-type Natriuretic Peptide (NTproBNP).
In certai nembodiments, the human subject carries a TTR gene mutation that is associated with the development of a TTR-associated disease, e.g., senile systemic amyloidosi (SSA),s systemic familial amyloidosi famis, lial amyloidoti polyc neuropat hy(FAP), familial amyloidoti cardic omyopat hy(FAC), leptomeningeal/Cent Nervousral System (CNS) amyloidosi ands, hyperthyroxinemia.
In certai nembodiments, the human subject has a transthyretin-mediated amyloidos (ATTRis amyloidos is)and the use of the double stranded RNAi agent reduces an amyloid TTR deposit in the human subject .In certai nembodiments, the ATTR amyloido sisis hereditary ATTR (h-ATTR) amyloidosi Ins. certain embodiments, the ATTR amyloidosi iss non-heriditary ATTR (wt ATTR) amyloidosis.
In certai nembodiments, the double stranded RNAi agent is administered to the human subject by subcutaneousl ory intravenously. In certain embodiments, the subcutaneous administration is self administration. In certain embodiments, the self-administration is via a pre-filled syringe or auto- injector device.
In certai nembodiments, the use further comprises assessing the level of TTR mRNA expression or TTR protein expression in a sample derived from the human subject, such as a human blood sample, or serum or plasm aderived therefrom.
In certai nembodiments, the double stranded RNAi agent is administered to the human subject once every month, once every othe rmonth, once every three months, once every four months, once every five months, or once every six months. In certain embodiments, the fixed dose of the double stranded RNAi agent is administere dto the human subject once about every three months In. certain embodiments, the fixed dose of the double stranded RNAi agent is administered to the human subject once about every six months.
In certai nembodiments, the double stranded RNAi agent is chronica admilly nistered to the human subject.
In certai nembodiments, the double stranded RNAi agent is administered to the human subject about once per quarter to about once per year. In certai nembodiments, the double stranded RNAi agent is administered to the human subject about once per quarter, about once every six months, or about once per year.
In certai nembodiments, the double stranded RNAi agent is administered to the human subject at a fixed dose of about 25 mg to about 300 mg. In certain embodiments, the double stranded RNAi agent is administered to the human subject at a fixed dose of about 25 mg to about 200 mg. In certain embodiments, the double stranded RNAi agen tis administered to the human subject at a fixed dose of about 75 mg to about 200 mg. In certai nembodiments, the double stranded RNAi agent is administered to the human subject at a fixed dose of about 25 mg. In certain embodiments, the double stranded RNAi agent is administered to the human subject at a fixed dose of about 50 mg. In certai nembodiments, the double stranded RNAi agent is administered to the human subject at a fixed dose of about 75 mg. In certain embodiments, the double stranded RNAi agent is administered to the human subject at a fixed dose of about 100 mg. In certai nembodiments, the double stranded RNAi agen tis administered to the human subject at a fixed dose of about 200 mg. In certain embodiments, the double stranded RNAi agent is administered to the human subject at a fixed dose of about 25 mg to about 300 mg; about 25 mg to about 200 mg; about 75 mg to about 200 mg; about 25 mg; about 50 mg; about 75 mg; about 100 mg; about 200mg; or about 300 mg once per quarter, i.e., about once every three months.
In certai nembodiments, the double stranded RNAi agent is administered to the human subject at a fixed dose of about 400 mg to about 600 mg. In certai nembodiments, the double stranded RNAi agent is administered to the human subject at a fixed dose of about 400 mg or about 600 mg about once 6 every six months to about once per year. In certain embodiments, the double stranded RNAi agent is administered to the human subject at a fixed dose of about 400 mg or about 600 mg about once every six months or about once per year.
In certai nembodiments, the double stranded RNAi agent is administered to the human subject at a fixed dose of about 700 mg to about 1000 mg or about 700 mg to about 900 mg. In certain embodiments, the double stranded RNAi agen tis administered to the human subject at a fixed dose of about 700 mg, about 800 mg, about 900mg, or about 1000 mg about once per year.
In certai nembodiments, the use further comprises administering to the human subject an additional therapeuti cagent, e.g., a TTR tetramer stabilize orr a non-steroida antil -inflammator agenty .
The present invention also provides kits for performing any of the methods of the invention.
The kits may include the double stranded RNAi agent; and a label comprising instructions for use.
The present invention is further illustrated by the following detailed description and drawings.
Brief Description of the Drawings Figure 1 is a graph depicting relative serum TTR protein levels in V30M transgenic mice (n=3 per group) after administration of a singl e1 mg/kg dose of the indicated double stranded RNAi agent s on Day 0.
Figure 2 is a graph depicting relative serum TTR protein levels in cynomolgus monkeys (n=3 per group) after administration of a singl e1 mg/kg dose or 3 mg/kg dose of the indicated double stranded RNAi agents on Day 0. Results shown are from three independent studies.
Detailed Description of the Invention The present invention provides methods of inhibiting expression of TTR, including inhibiting TTR expression in a human subjec twho does not meet diagnosti cric teria of a TTR-associated disease and methods of treating a human subject with a Transthyretin- (TTR-) associated disease using double stranded RNAi agents ,targeting the TTR gene wherein the sense strand comprises the modified nucleotid sequee nce 5’-usgsggauUfuCfAfUfguaaccaaga-3’ (SEQ ID NO: 6); and the antisense strand comprises the modified nucleotide sequence 5’-usCfsuugGf(Tgn)uAfcaugAfaAfucccasusc-3’ (SEQ ID no: ר.
The following detailed description discloses how to make and use compositio nscontaining iRNA agents to selectively inhibit the expression of a TTR gene, as well as compositions, uses, and methods for treating subject shaving diseases and disorders that would benefi tfrom inhibition or reduction of the expression of a TTR gene.
I. Definitions In order that the present invention may be more readily understood, certain terms are first defined. In addition, it shoul bed noted that whenever a valu eor range of values of a parameter are 7 recited, it is intended that values and ranges intermediate to the recited values are also intended to be part of this invention.
The article "sa" and "an" are used herein to refer to one or to more than one (i.e., to at least one) of the grammatica objectl of the article. By way of example, "an element" means one element or more than one element ,e.g., a pluralit ofy elements.
The term "including" is used herein to mean, and is used interchangeabl wityh, the phrase "including, but not limited to".
The term "or" is used herein to mean, and is used interchangeably with, the term "and/or," unless context clearly indicates otherwise.
The term "about" is used herein to mean within the typical ranges of tolerances in the art, e.g., acceptable variation in time between doses, acceptable variation in dosage unit amount. For example, "about" can be understood as within about 2 standard deviations from the mean. In certain embodiments, about means +10%. In certain embodiments, about means +5%. When about is present before a series of numbers or a range, it is understood that "about" can modify each of the numbers in the series or range.
The term "at least" , "no less than", or "or more" prior to a number or series of numbers is understood to include the number adjacent to the term "at least", and all subsequent number sor integers that coul logicald bely included, as clear from context For. example, the number of nucleotides in a nucleic acid molecu lemust be an integer. For example, "at least 18 nucleotides of a 21 nucleotide nucleic acid molecule" means that 18, 19, 20, or 21 nucleotides have the indicated property. When at least is present before a series of number sor a range ,it is understood that "at leas"t can modify each of the number sin the series or range.
As used herein, "no more than" or "less than" is understood as the valu eadjacent to the phrase and logical lower values or integers, as logical from context to, zero. For example, a duplex with an overhang of "no more than 2 nucleotides" has a 2, 1, or 0 nucleotid overhang.e When "no more than" is present before a series of numbers or a range ,it is understood that "no more than" can modify each of the number sin the series or range.
As used herein, methods of detection can include determination that the amount of analyt e present is belo wthe level of detection of the method.
As used herein, a "transthyreti"n ("TTR") refers to the well-known gene and protein. TTR is also known as prealbumin, HsT2651, PALB, and TBPA. TTR functions as a transporter of retinol- binding protein (RBP), thyroxine (T4) and retinol and, it also acts as a protease. The liver secretes TTR into the bloo d,and the choro idplexus secretes TTR into the cerebrospinal fluid. TTR is also expressed in the pancreas and the retinal pigment epithelium. The greatest clinic relalevanc ofe TTR is that both normal (wild type) and mutant TTR protein can form amyloid fibrils that aggregate into extracellula r deposits, causin gamyloidosi s.See, e.g., Saraiva M.J.M. (2002) Expert Reviews in Molecular Medicine, 4(12): 1-11 for a review. The molecula clonir ngand nucleotide sequenc eof rat transthyretin, as wel las the distribution of mRNA expression, was described by Dickson, P.W. et al. (1985) J. Biol. Chem. 260(13)8214-8219. The X-ray crystal structure of human TTR was described in Blake, C.C. et al. 8 (1974) J Mol Biol 88, 1-12. The sequenc eof a human TTR mRNA transcript can be found at National Center for Biotechnology Information (NCBI) RefSeq accession number NM_000371 (e.g., SEQ ID NOs:l and 5). The sequenc eof mouse TTR mRNA can be found at RefSeq accession number NM_013697.2, and the sequence of rat TTR mRNA can be found at RefSeq accession number NM_012681.1. Additional example sof TTR mRNA sequences are readil yavailable using publicl y available database e.g.,s, GenBank, UniProt, and OMIM.
A "TTR-associate dised ase," as used herein, is intended to include any disease associated with the TTR gene or protein. Such a disease may be caused, for example, by excess production of the TTR protein, by TTR gene mutations, by abnorm alcleavage of the TTR protein, instabilit ofy TTR tetramers, by abnorm alinteractions between TTR and othe rproteins or other endogenous or exogenous substances. A "TTR-associated disease" includes any type of transthyretin-mediated amyloidosi s (ATTR amyloidosis) wherein TTR plays a role in the formation of abnorm alextracellul aggregatar es or amyloid deposits, e.g., either herititary ATTR (h-ATTR) amyloido sisor non-heriditary ATTR (ATTR) amyloidosi s.TTR-associate dised ases include senile systemic amyloidos (SSAis ), systemic familial amyloidosi famis, lial amyloidoti polyc neuropat hy(FAP), familial amyloidoti cardic omyopat hy(FAC), leptomeningeal/Cent Nervousral System (CNS) amyloidosi amyls, oidoti vitrec ous opacities carpal, tunnel syndrome, and hyperthyroxinemia. Symptoms of TTR amyloido sisinclude sensory neuropathy (e.g., paresthesia, hypesthesia in distal limbs), autonom icneuropathy (e.g., gastrointestinal dysfunction, such as gastric ulcer, or orthostati hypotensic on), motor neuropathy, seizures, dementia, myelopathy, polyneuropathy, carpal tunnel syndrome, autonomic insufficienc y,cardiomyopathy, vitreous opacities, renal insufficiency, nephropathy, substantially reduced mBMI (modified Body Mass Index), crania l nerve dysfunction, and corneal lattic dystrophy.e As used herein, the term "strand comprising a sequence" refers to an oligonucleot ide comprising a chai nof nucleotides that is described by the sequence referred to using the standard nucleotid nomene clature.
The terms "iRNA," "RNAi agent," "iRNA agent,", "RNA interference agen"t as used interchangeabl herein,y refer to an agent that contains RNA as that term is defined herein, and which mediates the targeted cleavage of an RNA transcript via an RNA-induced silencing complex (RISC) pathway. iRNA directs the sequence-specifi degradationc of mRNA through a process known as RNA interference (RNAi). The iRNA modulates e.g.,, inhibits, the expression of a TTR gene in a cell e.g.,, a cel witl hin a subject, such as a mammalian subject.
As used herein, an "iRNA" for use in the compositions, uses, and methods of the invention is a double stranded RNA and is referred to herein as a "double stranded RNAi agent," "double stranded RNA (dsRNA) molecul" e,"dsRNA agent," or "dsRNA". The term "dsRNA" refers to a complex of ribonucl eicacid molecule havings, a duplex structure comprising two anti-paralle andl substantiall y complementary nucleic acid strands ,referred to as having "sense" and "antisense" orientations with respect to a target RNA, i.e., a TTR gene. A double stranded RNAi agent triggers the degradation of a target RNA, e.g., an mRNA, through a post-transcriptional gene-silencing mechanism referred to herein as RNA interference or RNAi. 9 As used herein, the term "modified nucleotid" erefers to a nucleotid havine g, independentl y,a modified sugar moiety, a modified internucleot idelinkage, or a modified nucleobase. Thus, the term modified nucleotid encompasse essubstitutions, additions or removal of, e.g., a functional group or atom ,to internucleosi lidenkages, sugar moieties, or nucleobases. The modifications suitable for use in the agents of the invention include all types of modifications disclose hereind or known in the art. Any such modifications, as used in a siRNA type molecule are, encompassed by "RNAi agen"t for the purposes of this specification and claims.
The duplex region may be of any lengt hthat permits specific degradation of a desired target RNA through a RISC pathway, and may range from about 21 to 36 base pairs in length, e.g., about 21- base pairs in length, for example, about 21-30, 21-29, 21-28, 21-27, 21-26, 21-25, 21-24, 21-23, or 21-22 base pairs in length. In one embodiment, an RNAi agent of the invention is a dsRNA agent ,each strand of which comprises 21-23 nucleotides that interacts with a TTR mRNA sequence to direct the cleavage of the targe tmRNA. Without wishing to be bound by theory, long double stranded RNA introduced into cells is broken down into siRNA by a Type III endonuclea knownse as Dicer (Shar pet al. (2001) Genes Dev. 15:485). Dicer, a ribonuclease-III-like enzyme, processes the dsRNA into 19-23 base pair short interfering RNAs with characteristi twoc base 3’ overhangs (Bernstein, et al., (2001) Nature 409:363). The siRNAs are then incorporat edinto an RNA-induced silencing complex (RISC) where one or more helicas esunwind the siRNA duplex, enabling the complementary antisense strand to guide target recognition (Nykanen, et al., (2001) Cell 107:309). Upon binding to the appropriate target mRNA, one or more endonucleases within the RISC cleave the targe tto induce silencing (Elbashir, et al., (2001) Genes Dev. 15:188). In one embodiment, an RNAi agent of the invention is a dsRNA of 24- nucleotide thats interact wis th a TTR mRNA sequence to direct the cleavage of the targe tRNA.
As used herein, the term "nucleotid overhange " refers to at least one unpaired nucleotid thate protrudes from the duplex structure of an iRNA, e.g., a dsRNA. For example, when a 3'-end of one strand of a dsRNA extends beyond the 5'-end of the other strand ,or vice versa, there is a nucleotide overhang. A dsRNA can comprise an overhang of at leas onet nucleotide; alternatively, the overhang can compris eat least two nucleotides, at least three nucleotides, at least four nucleotides, at least five nucleotides or more. A nucleotide overhang can comprise or consist of a nucleotide/nucleosid analog,e including a deoxynucleotide/nucleoside. The overhang(s) can be on the sense strand ,the antisense strand or any combinati onthereof. Furthermore, the nucleotide(s) of an overhang can be present on the '-end, 3'-end or both ends of either an antisense or sense strand of a dsRNA. In one embodiment of the dsRNA, at least one strand comprise sa 3’ overhang of at leas t1 nucleotide In. another embodiment, at least one strand comprises a 3’ overhang of at leas 2t nucleotides, e.g., 2, 3, 4, 5, 6, 7, 8, or 9 nucleotides. In othe rembodiments, at leas onet strand of the RNAi agen tcomprises a 5’ overhang of at least 1 nucleotide. In certain embodiments, at leas onet strand comprises a 5’ overhang of at least 2 nucleotides, e.g., 2, 3, 4, 5, 6, 7, 8, or 9 nucleotides. In still othe rembodiments, both the 3’ and the 5’ end of one strand of the RNAi agen tcomprise an overhang of at leas 1t nucleotide.
In one embodiment, the antisense strand of a dsRNA has a 1-9 nucleotide, e.g., 0-3, 1-3, 2-4, 2- , 4-9, 5-9, e.g., a 1, 2, 3, 4, 5, 6, 7, 8, or 9 nucleotide overhang, at the 3’-end or the 5’-end. In one embodiment, the sense strand of a dsRNA has a 1-9 nucleotide e.g.,, a 1, 2, 3, 4, 5, 6, 7, 8, or 9 nucleotide overhang, at the 3’-end or the 5’-end. In another embodiment, one or more of the nucleotides in the overhang is replaced with a nucleoside thiophosphate.
"Blunt" or "blunt end" means that there are no unpaired nucleotides at that end of the double stranded RNAi agent ,i.e., no nucleotid overhang.e A "blunt ended" RNAi agent is a dsRNA that is double stranded over its entire length, i.e., no nucleotide overhang at either end of the molecul e.The RNAi agents of the invention include RNAi agents with nucleotid overhangse at one end (i.e., agent s with one overhang and one blunt end) or with nucleotide overhangs at both ends.
The term "antisense strand" or "guide strand" refers to the strand of an iRNA, e.g., a dsRNA, which includes a region that is substantially complementary to a target sequence, e.g., a TTR mRNA.
As used herein, the term "region of complementari" tyrefers to the region on the antisense strand that is substantially complementary to a sequence, for example a target sequence, e.g., a TTR nucleotide sequence, as defined herein. Where the region of complementari isty not fully complementary to the target sequence, the mismatches can be in the internal or terminal regions of the molecul e.Generally, the most tolerated mismatches are in the terminal regions, e.g., within 5, 4, 3, 2, or 1 nucleotides of the ’ - or 3’-terminus of the iRNA. In one embodiment, a double stranded RNAi agent of the invention include aa nucleotide mismatch in the antisense strand. In another embodiment, a double stranded RNAi agent of the invention includes a nucleotide mismatch in the sense strand. In one embodiment , the nucleotide mismatch is, for example, within 5, 4, 3, 2, or 1 nucleotides from the 3’-terminus of the iRNA. In anothe embodir ment the, nucleotid mise match is, for example, in the 3’-terminal nucleoti de of the iRNA.
The term "sense strand," or "passenger strand" as used herein, refers to the strand of an iRNA that includes a region that is substantially complementary to a region of the antisense strand as that term is defined herein.
As used herein, the term "cleavage region" refers to a region that is locate immd ediately adjacen tot the cleavage site. The cleavage site is the site on the target at which cleavage occurs. In some embodiments, the cleavage region comprises three base son either end of, and immediately adjacen to,t the cleavage site. In some embodiments, the cleavage region comprises two bases on either end of, and immediatel yadjacent to, the cleavage site. In some embodiments, the cleavage site specifical occursly at the site bound by nucleotides 10 and 11 of the antisense strand ,and the cleavage region comprises nucleotides 11, 12 and 13.
As used herein, and unles sotherwise indicated, the term "complementary," when used to describe a first nucleotid sequence ein relation to a secon dnucleotid sequence,e refers to the abilit ofy an oligonucleoti orde polynucleot compide rising the first nucleotid sequee nce to hybridize and form a duplex structure under certain conditions with an oligonucleoti orde polynucleot compide rising the second nucleotide sequence, as will be understood by the skilled person. Such conditions can be, for example, "stringent conditions", where stringent conditions can include: 400 mM NaCl 40, mM PIPES pH 6.4, 1 mM EDTA, 50 oC or 70 oC for 12-16 hours followed by washing (see, e.g., "Molecul ar Clonin g:A Laboratory Manual, Sambrook, et al. (1989) Col dSpring Harbor Laboratory Press). Other 11 conditions, such as physiological relevantly conditions as can be encountered inside an organism, can apply. The skilled person wil lbe able to determine the set of conditions most appropriate for a test of complementari ofty two sequence sin accordance with the ultimate application of the hybridized nucleotides.
Complementa rysequence swithin an iRNA, e.g., within a dsRNA as described herein, include base-pairing of the oligonucleoti orde polynucleot compide rising a first nucleotid sequee nce to an oligonucleoti orde polynucleot compide rising a second nucleotid sequence eover the entire lengt hof one or both nucleotide sequences .Such sequences can be referred to as "fully complementary" with respect to each other herein. However, where a first sequenc eis referred to as "substantiall compley mentary" with respect to a secon dsequence herein, the two sequences can be full ycomplementary, or they can form one or more, but generall noty more than 5, 4, 3 or 2 mismatched base pairs upon hybridization for a duplex up to 30 base pairs ,whil eretaining the abilit toy hybridize under the conditions most relevant to their ultimate application, e.g., inhibition of gene expression, in vitro or in vivo.. However, where two oligonucleotide ares designed to form, upon hybridization, one or more single stranded overhangs, such overhangs shal notl be regarded as mismatches with regard to the determination of complementarity. For example, a dsRNA comprising one oligonucleoti 21de nucleotides in lengt hand anothe olir gonucleoti 23de nucleotides in length, wherein the longer oligonucleoti comprisesde a sequenc eof 21 nucleotide thats is fully complementa tory the shorter oligonucleoti cande, yet be referred to as "full ycomplementary" for the purposes described herein.
"Complementa"ry sequences, as used herein, can also include, or be formed entirely from, non- Watson-Crick base pairs or base pairs formed from non-natura andl modified nucleotides, in so far as the above requirements with respect to their abilit toy hybridize are fulfilled. Such non-Watson-Cri ck base pairs include, but are not limited to, G:U Wobble or Hoogstein base pairing.
The terms "complementary," "fully complementary" and "substantially complementary" herein can be used with respect to the base matchin betwg een the sense strand and the antisense strand of a dsRNA, or between two oligonucleotides or polynucleotides such, as the antisense strand of an iRNA agent and a target sequence, as will be understood from the context of their use.
As used herein, a polynucleot thatide is "substantially complementa tory at least part of’ a messenger RNA (mRNA) refers to a polynucleot thatide is substantially complementa tory a contiguous portion of the mRNA of interest (e.g., an mRNA encoding a TTR gene). For example, a polynucleotide is complementa tory at least a part of a TTR mRNA if the sequenc eis substantiall compley mentary to a non-interrupted portion of an mRNA encoding a TTR gene.
Accordingly, in some embodiments, the antisense polynucleotide discls osed herein are full y complementary to the targe tTTR sequence. In othe rembodiments, the antisense polynucleoti des disclosed herein are fully complementary to SEQ ID NO:8 (5’- UGGGAUUUCAUGUAACCAAGA - 3’). In one embodiment, the antisense polynucleot sequeide nce is 5’- UCUUGGUUACAUGAAAUCCCAUC -3’ (SEQ ID NO:9), wherein the U at position 7 of the antisense strand can be a T. 12 As used herein, a "reference level" is understood as a predetermined level to which a level obtained from an assay, e.g., a biomarke lever l, e.g., a protein biomarke lever l, is compared. In certain embodiments, a reference level can be a control level determined for a healthy population, e.g., a population that does not have a disease or condition associated with a changed level of the biomarker and does not have a predisposition, e.g., genetic predisposition, to a disease or condition associate witd h a changed level of the biomarker. In certain embodiments, the population shoul bed matched for certain criteria ,e.g., age, sex. In certai nembodiments, the reference level of the biomarke isr a level from the same subjec tat an earlier time, e.g., before the development of symptomatic disease or before the start of treatment. Typically sampl, es are obtained from the subject at clinical rellyevant intervals e.g.,, at intervals sufficiently separated in time that a change in the biomarke couldr be observed ,e.g., at leas at three month interval, at least a six month interval or, at leas at nine month interval. When more than two samples are obtained from a subject over time, it is understood that any of the prior samples can act as a reference level.
As used herein, a "change as compared to a reference level" and the like is understood as a statisticall ory clinical signily ficant change in the biomarke lever l, e.g., the change in the protein biomarker level as, compare dto the reference level is, greater than the typical standard deviation of the assay method. Moreover, the change shoul bed clinical relevly ant. The change as compare dto a reference level can be determined as a percent change. For example, if a reference level is 100 pg/ml for biomarker X, and the level of biomarker X in the subject is 150 pg/ml, the level is increased by 50% calcula byted ((150 pg/ml -100 pg/ml)/100 pg/ml) X 100% = 50%. If the level of biomarker X in the subject is 300 pg/ml, the level is increased by 300%. If the level of biomarker X in the subject is 50 pg/ml, the level is decreased by 50%. In certain embodiments, the change as compared to a reference level is increased by at least 50%. In certain embodiments, the change as compare dto a reference leve l is increased by at least 100%, at leas 200%,t or at leas 300%.t In certain embodiments, the change as compared to a reference sampl eis decreased by at least 25%. In certai nembodiments, the change as compared to a reference sampl eis decreased by at least 50%.
A "biologic samal ple from a subjec"t or a "sampl efrom a subjec"t as used herein, includes one or more fluids, cells or, tissues isolated from a subject. Examples of biologic fluidsal include blood, serum, serosal fluids, plasma, cerebrospinal fluid, ocula fluidsr ,lymph, urine, saliva, and the like.
Tissue samples may include samples from tissues, organs, or localized regions. For example, samples may be derived from particul arorgans, parts of organs, or fluids or cell wits hin those organs. In certain embodiments, samples may be liver tissue or be derived from the liver. In some embodiments, a "biologic sampal le from a subjec"t can refer to blood or blood derived serum or plasm afrom the subject. In some embodiments, the fluid is substantially free of cells e.g.,, is free of cells.
As used herein, a "clinical relevantly difference" is understood as at leas greatt er difference than typical interobserver variatio forn the assessment, wherein the observer may be a trained healt careh professional a caregi, ver, or the patient, performing the same assessment on the same individual at around the same time, e.g., within a week, such as within consecutive days. It is understood that certain patien tobservations are subjective and shoul bed nearly identical when performed by different 13 observers within a short time frame, e.g., body weight, heart rate . Other qualitative measures, such as some aspect sof mNIS+7 (e.g., response to touch-pressure, vibration, joint position and motion) and Norfolk-Quali ofty Life (e.g., pain level, hot or cold hands and feet, steadines swhile standing) may vary more from day to day, and from one observer to another. Therefore ,composite scores are used to aggregate observations and larger variations between observers are expected without any indication of clinical relevantly change. Assays for biomarker levels have known level sof variations within and between samples. Determination of a clinical relevantly difference is within the ability of one of skil lin the art, e.g., a healt careh professional experienced in treating patients with TTR associated diseases , clinic laboal rato professry ional.
As used herein, "chronica admilly nistered" is understood as administration for an indefinite interval e.g.,, for the remainder of the life of the subject, until liver transplant.
As used herein, a "therapeuti cagent that stabilizes TTR" or "that stabilizes a TTR tetramer" is an agent that reduces or prevents the dissociatio ofn the subunits of a TTR tetramer ,e.g., into monomers. In some embodiments, the agent reduces the formation of TTR amyloid plaque s,e.g., by reducing the level of TTR monomers or proteolyti frac gment sof TTR monomer thats form TTR amyloid plaques. Such agents include, but are not limited to, tafamidis ,diflunisal, and AGIO.
As used herein, the term "administering a therapeutic agent" is understood as providing a therapeuti cagent to a subject. In embodiments, the therapeuti cagent is provided at an appropriate dosage and by a route of administration for the agent as provided, for example, by the label of the therapeuti cagent.
II. Methods for Treating a TTR-Associated Disease The present invention provides double stranded RNAi agents and their use in methods for treating a TTR-associated disease in a human subject, such as a transthyretin-mediated amyloidosi s (ATTR amyloidosis e.g.,), hereditary ATTR (h-ATTR) amyloido sisor non-heriditar ATTRy (wt ATTR) amyloidosis or; for inhibiting expression of TTR in a subject that does not yet meet the diagnostic criteria of a TTR-associated disease, but who is at risk for developing a TTR-associate diseasd e, e.g., a subject with a TTR mutation associate wid th TTR amyloidosi as, subject with some indicia of TTR amyloido siswho does not yet meet the diagnostic criteria of TTR amyloidosi as, subjec twith altered biomarker levels associate wid th TTR amyloidosi s.The methods include administering to the subject a therapeuticall effectiy ve amoun oft an RNAi agent of the invention.
In one aspect, the present invention provides methods of treating a human subject suffering from a TTR-associated disease or at risk for developing a TTR-associated disease. The methods include administering to the human subjec ta fixed dose of about 25 mg to about 1000 mg of a double stranded RNAi agent wherein the sense strand comprises the modified nucleotide sequenc e5’- usgsggauUfuCfAfUfguaaccaaga-3‘ (SEQ ID NO: 6); and the antisense strand comprises the modified nucleotid sequee nce 5’-usCfsuugGf(Tgn)uAfcaugAfaAfucccasusc-3’ (SEQ ID NO: 7), 14 wherein a, c, g, andu are 2'-O-methyladenosine-3’-phosphate, 2'-O-methylcytidine-3’- phosphate, 2'-O-methylguanosine-3’-phosphate, and 2'-O-methyluridine-3’-phosphat respectie, vely; Af, Cf, Gf, and Uf are 2‘-fluoroadenosine-3‘ -phosphate, 2‘-fluorocytidine-3‘ -phosphate, 2’- fluoroguanosine-3’-phosphat e,and 2’-fluorouridine’-3-phosphat e,respectively; (Tgn) is thymidine-glycol nucleic acid (GNA) S-Isomer; and s is a phosphorothioa linteker.
In another aspect, the present invention provides methods of improving at least one indici aof neurological impairement or quali tyof life in a human subjec tsuffering from a TTR-associate disead se or at risk for developing a TTR-associate disead se. The methods include administerin gto the human subject a fixed dose of about 25 mg to about 1000 mg of a double stranded RNAi agent wherein the sense strand comprises the modified nucleotid sequee nce 5’-usgsggauUfuCfAfUfguaaccaaga-3’ (SEQ ID NO: 6); and the antisense strand comprises the modified nucleotide sequenc e5’- usCfsuugGf(Tgn)uAfcaugAfaAfucccasusc-3’ (SEQ ID NO: 7).
In another aspect, the present invention provides methods of reducing, slowing, or arresting a Neuropath yImpairment Score (NIS) or a modified NIS (mNIS+7) in a human subject suffering from a TTR-associate diseased or at risk for developing a TTR-associated disease. The methods include administering to the human subjec ta fixed dose of about 25 mg to about 1000 mg of a double stranded RNAi agent wherein the sense strand comprises the modified nucleotid sequence e5’- usgsggauUfuCfAfUfguaaccaaga-3‘ (SEQ ID NO: 6); and the antisense strand comprises the modified nucleotid sequee nce 5’-usCfsuugGf(Tgn)uAfcaugAfaAfucccasusc-3’ (SEQ ID NO: 7).
In another aspect, the present invention provides methods of increasin ag 6-minute wal ktest (6MWT) in a human subject suffering from a TTR-associated disease or at risk for developing a TTR- associate disead se. The methods include administering to the human subject a fixed dose of about 25 mg to about 1000 mg of a double stranded RNAi agent wherein the sense strand comprises the modified nucleotid sequee nce 5’-usgsggauUfuCfAfUfguaaccaaga-3’ (SEQ ID NO: 6); and the antisense strand comprises the modified nucleotide sequence 5’-usCfsuugGf(Tgn)uAfcaugAfaAfucccasusc-3’ (SEQ ID NO: 7).
In certai nembodiments, the double stranded RNAi agent is administered to the human subject about once per quarter to about once per year. In certai nembodiments, the double stranded RNAi agent is administered to the human subject about once per quarter, about once every six months, or about once per year.
In certai nembodiments, the double stranded RNAi agent is administered to the human subject at a fixed dose of about 25 mg to about 300 mg. In certain embodiments, the double stranded RNAi agent is administered to the human subject at a fixed dose of about 25 mg to about 200 mg. In certain embodiments, the double stranded RNAi agen tis administered to the human subject at a fixed dose of about 75 mg to about 200 mg. In certai nembodiments, the double stranded RNAi agent is administered to the human subject at a fixed dose of about 25 mg. In certain embodiments, the double stranded RNAi agent is administered to the human subject at a fixed dose of about 50 mg. In certai nembodiments, the double stranded RNAi agent is administered to the human subject at a fixed dose of about 75 mg. In certain embodiments, the double stranded RNAi agent is administered to the human subject at a fixed dose of about 100 mg. In certai nembodiments, the double stranded RNAi agen tis administered to the human subject at a fixed dose of about 200 mg. In certain embodiments, the double stranded RNAi agent is administered to the human subject at a fixed dose of about 25 mg to about 300 mg; about 25 mg to about 200 mg; about 50 mg to about 300 mg; about 25 mg; about 50 mg; about 100 mg; about 200mg; or about 300 mg once per quarter, i.e., about once every three months.
In certai nembodiments, the double stranded RNAi agent is administered to the human subject at a fixed dose of about 400 mg to about 600 mg. In certai nembodiments, the double stranded RNAi agent is administered to the human subject at a fixed dose of about 400 mg or about 600 mg about once every six months to about once per year. In certain embodiments, the double stranded RNAi agent is administered to the human subject at a fixed dose of about 400 mg or about 600 mg about once every six months or about once per year.
In certai nembodiments, the double stranded RNAi agent is administered to the human subject at a fixed dose of about 700 mg to about 1000 mg or about 700 mg to about 900 mg. In certain embodiments, the double stranded RNAi agen tis administered to the human subject at a fixed dose of about 700 mg, about 800 mg, about 900mg, or about 1000 mg about once per year.
In an embodiment, the subject is a human being treated or assessed for a disease, disorder or condition that would benefit from reduction in TTR gene expression; a human at risk for a disease, disorder or condition that would benefit from reduction in TTR gene expression, e.g., a human who does not meet the diagnosti cric teria of a TTR-associated disease, but who demonstrates at least one sign or symptom of a TTR-associated disease or has at least one risk factor of developing a TTR associate d disease; a human having a disease, disorder or condition that would benefit from reduction in TTR gene expression; or human being treated for a disease, disorder or condition that would benefit from reduction in TTR gene expression, as described herein.
In some embodiments, the human subject is suffering from a TTR-associated disease. In other embodiments, the subject is a subject at risk for developing a TTR-associated disease, e.g., a subject with a TTR gene mutation that is associated with the development of a TTR associate dised ase ,a subject with a family history of TTR-associated disease, or a subject who has signs or symptoms suggesting the development of TTR associated disease without meeting the diagnosti cric teria for a TTR-associate dised ase.
A "TTR-associate dised ase," as used herein, includes any disease caused by or associated with the formation of amyloid deposits in which the fibril precurosor sconsist of variant or wild-type TTR protein. Mutant and wild-type TTR give rise to various forms of amyloid deposition (amyloidosis ).
Amyloidosi involves thes formation and aggregation of misfolded proteins, resulting in extracellula r deposits that impair organ function. Climical syndromes associate wid th TTR aggregation include, for example, senile systemic amyloido sis(SSA); systemic familial amyloidosi famis; lial amyloidoti c polyneuropat hy(FAP); familial amyloidoti cardic omyopathy (FAC); and leptomeningea amyll oidosi s, also known as leptomeningeal or meningocerebrovascul amyloar idosi cents, ral nervous system (CNS) amyloidosi ors, amyloido sisVII form. 16 In one embodiment, the RNAi agents of the invention are administered to subject ssuffering from familial amyloidotic cardiomyopat hy(FAC). In anoth erembodiment, the RNAi agents of the invention are administered to subject ssuffering from FAC with a mixed phenotype, i.e., a subject having both cardiac and neurological impairements. In yet anothe embor diment, the RNAi agents of the invention are administered to subject ssuffering from FAP with a mixed phenotype, i.e., a subject having both neurologic andal cardiac impairements. In one embodiment, the RNAi agents of the invention are administered to subject ssuffering from FAP that has been treated with an orthotopic liver transplantati (OLonT).
In another embodiment, the RNAi agents of the invention are administered to subject ssuffering from senile systemic amyloidos (SSAis ). In othe rembodiments of the methods of the invention, RNAi agents of the invention are administered to subject ssuffering from familial amyloidoti cardiomyopathyc (FAC) and senil esystemic amyloido sis(SSA). Normal-sequence TTR cause scardiac amyloido sisin peopl ewho are elderly and is termed senil esystemic amyloido sis(SSA) (als ocalled senil ecardiac amyloido sis(SCA) or cardiac amyloidosis). SSA often is accompanie byd microscopic deposits in many othe rorgans. TTR mutations accelerate the process of TTR amyloid formation and are the most important risk factor for the development of clinical signifily cant TTR amyloido sis(also call edATTR (amyloidosis-transthyret type)).in More than 85 amyloidogenic TTR variants are known to cause systemic familial amyloidosis.
In some embodiments of the methods of the invention, RNAi agents of the invention are administered to subject ssuffering from transthyretin (TTR)-related familial amyloidoti polyneurc opathy (FAP). Such subject smay suffer from ocular manifestations, such as vitreous opacity and glaucoma. It is known to one of skil lin the art that amyloidogenic transthyretin (ATTR) synthesized by retinal pigment epithelium (RPE) plays important roles in the progression of ocular amyloidosi s.Previous studies have shown that panretinal laser photocoagulat whichion, reduced the RPE cells, prevented the progression of amyloid deposition in the vitreous, indicating that the effective suppression of ATTR expression in RPE may become a novel therapy for ocular amyloido sis(see, e.g., Kawaji T.,, et al., Ophthalmology. (2010) 117: 552-555). Another TTR-associated disease is hyperthyroxinemia, als o known as "dystransthyretinem ichyperthyroxinem"ia or "dysprealbuminemic hyperthyroxinem"ia. This type of hyperthyroxinem iamay be secondary to an increased association of thyroxine with TTR due to a mutant TTR molecul wite h increased affinity for thyroxine. See, e.g., Moses et al. (1982) J. Clin.
Invest., 86, 2025-2033.
The RNAi agents of the invention may be administered to a subject using any mode of administration known in the art, including, but not limited to subcutaneous, intravenou s,and intramuscular injection, and any combinations thereof.
In some embodiments, the agents are administered to the subject subcutaneously.
In some embodiments, a subject is administered a singl edose of an RNAi agent via subcutaneous injection, e.g., abdominal, thigh, or upper arm injection. In othe rembodiments, a subject is administered a spli tdose of an RNAi agent via subcutaneous injection. In one embodiment, the spli t dose of the RNAi agent is administered to the subject via subcutaneous injection at two different 17 anatomical locations on the subject. For example, the subjec tmay be subcutaneousl injey cted subcutaneously at a dose of 25 mg to 1000 mg. In some embodiments of the invention, the subcutaneous administration is self-administrati onvia, e.g., a pre-fille dsyringe or auto-inject orsyringe.
In some embodiments, a dose of the RNAi agent for subcutaneous administration is containe ind a volume of less than or equal to one ml of, e.g., a pharmaceutical acceptly able carrier. In some embodiments, the RNAi agent is in a non-pyrogenic formulation.
In some embodiments, the RNAi agent is administered to a subject in an amount effective to inhibit TTR expression in a cell within the subject. The amount effective to inhibit TTR expression in a cel witl hin a subject may be assessed using methods discussed below including, methods that involve assessment of the inhibition of TTR mRNA, TTR protein, or related variable suchs, as amyloid deposits.
In some embodiments, the RNAi agent is administered to a subject in a therapeuticall effey ctive amount.
"Therapeutical effectily ve amount," as used herein, is intended to include the amoun oft an RNAi agent that ,when administered to a patient for treating a TTR associated disease, is sufficient to effect treatment of the disease (e.g., by diminishing, maintaining, or slowing the progression of the existing disease as compared to an appropriate control or; diminishing, maintaining, or slowing one or more symptoms of disease as compared to an appropriate control The). "therapeuticall effey ctive amount" may vary depending on the RNAi agent ,how the agent is administered, the disease and its severity and the history, age, weight, family history, genetic makeup, stage of pathological processes mediated by TTR expression, the types of preceding or concomitant treatments, if any, and other individual characteristi ofcs the patient to be treated. Diagnost iccriteria for TTR amyloidosis, polyneuropathi andes, cardiomyopathie ares discussed further below.
"Therapeutical effectily ve amount," as used herein, is intended to include the amount of an RNAi agent that ,when administered to a subject who does not yet meet the diagnositi ccriteria of a TTR-associate disease,d e.g., a subject who has not been diagnosed with hTTR amyloidosi s polyneuropathy; a subject who does not meet the diagnosti cric teria of Stage 1 FAP, but who may be predisposed to the disease ,e.g., a subject with a TTR mutation associate wid th TTR amyloidosi as, subject suffering from one or more of orthostati hypotension,c heart failure cardiac, arrhythmia, left ventricula wallr thickness, interventricular septal wall thickness, cardiac posterior wal thil cknes s diarrhea constipation,, erectile dysfunction, glaucom inta, ravitreal deposition, scalloped pupils ;carpal tunnel syndrome, lumbar spina lstenosis, and bicep tendon rupture; a subject with an elevate d neurofilament light chai n(NfL) level as compared to a reference sample, e.g., a Nfl level of at leas 37t pg/ml in serum, is sufficient to prevent or ameliorate the disease or one or more symptoms of the disease. Symptoms that may be ameliorat edinclude sensory neuropathy (e.g., paresthesia hypesthesi, a in distal limbs), autonom icneuropathy (e.g., gastrointestinal dysfunction, such as gastric ulcer, or orthostatic hypotension), motor neuropathy, seizures, dementia, myelopathy, polyneuropat hy,carpal tunnel syndrome, autonomic insufficienc y,cardiomyopathy, vitreous opacitie s,renal insufficienc y, nephropathy, substantially reduced mBMI (modified Body Mass Index), crani alnerve dysfunction, corneal lattic dystrophy,e left !ventricular (LV) wall thickening by echocardiograph assessic ment, 18 increased global longitudinal strain by echocardiographic assessment ,increased N-terminal prohormone B-type Natriuretic Peptide (NTproBNP), and hospitalizati dueon to cardiac event. Diminishing the disease includes slowing the course of the disease or reducing the severity of later-developin diseag se.
The dose may vary depending on the RNAi agent, how the agent is administered, the degree of risk of disease ,and the history, age, weight, family history, genetic makeup, the types of preceding or concomitant treatments, if any, and othe rindividual characteristic ofs the patient to be treated.
A "therapeutically-effect amountive " also includes an amoun oft an RNAi agen tthat produces some desired local or systemic effect at a reasonable benefit/risk ratio applicable to any treatment. RNAi agents employed in the methods of the present invention may be administered in a sufficient amount to produce a reasonable benefit/risk ratio applicable to such treatment.
As used herein, the phrase "therapeutical effectily ve amount" also includes an amount that provides a benefit in the treatment, prevention, or management of pathologic processesal or symptom(s) of pathologic procesal ses mediated by TTR expression. Symptoms of TTR amyloido sisinclude sensory neuropathy (e.g. paresthesia, hypesthesia in distal limbs), autonom icneuropathy (e.g., gastrointestinal dysfunction, such as gastric ulcer, or orthostati hypotensic on), motor neuropathy, seizures, dementia, myelopathy, polyneuropathy, carpal tunnel syndrome, autonom icinsufficiency, cardiomyopathy, vitreous opacitie s,renal insufficiency, nephropathy, substantially reduced mBMI (modified Body Mass Index), cranial nerve dysfunction, corneal lattice dystrophy, left !ventricular (LV) wall thickening by echocardiographic assessment ,increased global longitudin alstrain by echocardiograph assessmic ent, increased N-terminal prohormone B-type Natriuretic Peptide (NTproBNP), and hospitalizati dueon to cardiac event.
In one embodiment, for example, when the subject has FAP, FAP with mixed phenotype, FAC with mixed phenotype, or FAP and has had an OLT, treatment of the subject with a dsRNA agent of the invention slows the progression of neuropathy. In anoth erembodiment, for example, when the subject has FAP, FAP with mixed phenotype, FAC with mixed phenotype SSA,, or FAP and has had an OLT, treatment of the subject with a dsRNA agent of the invention slow sthe progression of neuropathy and cardiomyopathy. In anothe embor diment, for example, when the subject has cardiac involvement, the method of the invention improve cardiac structure and function, including, for example, the methods reduce the mean left ventricula wallr thickness and longitudin alstrain, and reduce the expression level of the cardiac stress biomarker N-te, rminal pro b-type natriureti cpeptide (NT-proBNP).
Adminsitration of a therapeuticall or yprophylacticall effectiy ve amount of the RNAi agen tof the invention is also useful in methodsfor improving at leas onet indicia of neurological impairment or qualit ofy life in a subject suffering from or at risk of developing a TTR-associated disease.
For example, in one embodiment, the methods of the invention improve at least indicia of neurological impairment in the subject ."Improving at leas onet indici aof neurological impairment" in the subject refers to the ability of the methods of the invention to slow reduc, e, or arrest neurological impairment, or improve any symptom associated with neurologic imalpairment. Any suitable measure of neurological impairment can be used to determine whether a subject has reduced, slowed, or arrested, neurological impairment, or an improvement of a symptom associate wid th neurological impairment. 19 One suitable measure is a Neuropath yImpairment Score (NIS). NIS refers to a scoring system that measures weakness, sensation, and reflexes, especiall wiyth respect to peripheral neuropathy. The NIS score evaluate as standard group of muscles for weaknes s(1 is 25% weak, 2 is 50% weak, 3 is 75% weak, 3.25 is movement agains gravity,t 3.5 is movement with gravity eliminate d,3.75 is muscle flicker without movement, and 4 is paralyzed), a standard group of muscle stretch reflexes (0 is normal, 1 is decreased, 2 is absent) , and touch-pressure, vibration, joint position and motion, and pinprick (all graded on index finger and big toe: 0 is normal 1 ,is decreased, 2 is absent). Evaluations are correcte d for age, gender, and physical fitness.
In one embodiment, the methods of the invention reduce a NIS by at leas 5t points at 18 months from the start of dosing. In othe rembodiments, the methods of the invention result in a stabilization of NIS at 18 months from the star tof treatment with an RNAi agent provided herein. In othe r embodiments, the methods slow an increasing NIS score as compared to an appropriate control group showing the natura histol ry of the disease, e.g., a placebo control group as provided, for example in Adams et al., N Engl J Med 2018;379:11-21. It is understood that the rate of disease progression is dependen tupon a number of factors including, but not limited to, the severity of disease in the subject at the initiation of treatment, the duration of treatment, prior treatments, and the specific TTR mutation present, if any.
Methods for determining a NIS in a human subject are well known to one of skil lin the art and can be found in, for example, Dyck, PJ et al., (1997) Neurology 1997. 49(1): pgs. 229-239); Dyck PJ. (1988) Muscle Nerve. Jan; 1 l(l):21-32.
Another suitable measurement of neurological impairment is a Modified Neuropathy Impairment Score (mNIS+7). As known to one of ordinary skil lin the art, mNIS+7 refers to a clinical exam-base dassessment of neurologic impairment (NIS) combine wid th electrophysiologic measures of smal andl large nerve fiber function (NCS and QST), and measurement of autonom icfunction (postural blood pressure). The mNIS+7 score is a modificatio ofn the NIS+7 score (which represents NIS plus seven tests). NIS+7 analyzes weaknes sand muscle stretch reflexes. Five of the seven tests include attributes of nerve conduction. These attribute sare the peroneal nerve compound muscle action potential amplitude, motor nerve conduction velocity and motor nerve distal latency (MNDL), tibia l MNDL, and sural sensory nerve action potential amplitudes. These values are corrected for variables of age, gender, height, and weight. The remaining two of the seven tests include vibratory detection threshol andd heart rate decrease with deep breathing.
The mNIS+7 score modifies NIS+7 to take into account the use of Smar tSomatotopic Quantitative Sensation Testing, new autonom icassessments ,and the use of compound muscle action potential of amplitudes of the ulnar, peroneal, and tibial nerves, and sensory nerve action potential ofs the ulna andr sural nerves (Suanpraser t,N. et al., (2014) J. Neurol. Sci., 344(1-2): pgs. 121-128).
In one embodiment, the methods of the invention reduce a mNIS+7 by at leas 5t points at 18 months from the start of dosing. In othe rembodiments, the methods of the invention result in a stabilization of mNIS+7 at 18 months from the start of treatment with an RNAi agent provided herein.
In othe rembodiments, the methods slow an increasin mNIg S+7 score as compare dto an appropriate control group showing the natura hisl tory of the disease, e.g., a placebo control group as provided, for example in Adams et al., N Engl J Med 2018;379:11-21. It is understood that the rate of disease progression is dependent upon a number of factors including, but not limited to, the severity of disease in the subject at the initiation of treatment ,the duration of treatment, prior treatments, and the specific TTR mutation present, if any.
In another embodiment, the methods of the invention improve at least one indici aof quali tyof life in the subject ."Improving at least one indici aof qualit ofy life" in the subject refers to the abilit ofy the methods of the invention to slow or arrest quali tyof life worsening or improve qualit ofy life. Any suitable measure of quality of life can be used to determine whether a subject has slowed or arrested qualit ofy life worsening, or improved qualit ofy life.
For example, the SF-36® healt surveyh provides a self-reporting mult, i-item scal meae suring eight healt parah meters: physical functioning, role limitations due to physical healt problems,h bodily pain, general health, vitality (energy and fatigue), social functioning, role limitations due to emotional problems, and mental health (psychological distress and psychological well-being). Each scal ise directly transformed into a 0-100 scal one the assumption that eac hquestion carries equal weight. The lower the score the more disabilit y.The highe rthe score the less disability i.e., a score of zero is equivalent to maximum disability and a score of 100 is equivalent to no disability. The survey also provides a physical component summary and a mental compone ntsummary.
In one embodiment, the methods of the invention provide to the subject an improvement versus baseline in at least one of the SF- 36 physical healt relath ed parameter s(physical health, role-physical, bodil ypain or general health) or in at least one of the SF-36 mental healt relath ed parameters (vitality, social functioning, role-emotional or mental health). Such an improvement can take the form of an increas of,e for exampl eat least 2 or at leas 3t points ,on the scal fore any one or more parameter sat 9 months from the start of the dosing.
In othe rembodiments, the methods of the invention arrest a decreasing SF-36 parameter score at 9 months from the start of dosing for any one or more parameters, e.g., the methods result in no clinical signily ficant change of the SF-36 e.g., within the variation observed for individuals performing an SF-36 assessment. In yet othe rembodiments, the methods of the invention slow the rate at which a SF-36 score decreases at 9 months from the start of dosing, e.g., the rate of decrease of an SF-36 score in a subject treated with an RNAi agent of the invention as compared to the rate of decrease of an SF-36 score as compared to an appropriate control group showing the natural history of the disease, e.g., a placebo control group as provided, for example in Adams et al., N Engl J Med 2018;379:11-21. It is understood that the rate of disease progression is dependent upon a number of factor incls uding, but not limited to, the severity of disease in the subject at the initiation of treatment, the duration of treatment, prior treatments, and the specific TTR mutation present, if any.
Another suitable measurement of qualit ofy life is the Norfolk Qualit yof Life-Diabetic Neuropath y(Norfolk QOL-DN) questionnair e.The Norfolk QOL-DN is a validated comprehensive questionna iredesigned to capture the entire spectrum of DN related to large fiber, small fiber, and autonomic neuropathy not captured in existing instruments. 21 In one embodiment, the methods of the invention improve a subjec’ts Norfolk QOL-DN score from baseline, e.g., a change of about -2.5, -3.0, -3.5, -4.0, -4.5, or -5.0 at 9 months from the start of treatment with an RNAi agent provided herein. In othe rembodiments, the methods arrest an increasing Norfolk QOL-DN score ,e.g., the methods result in no clinical signily ficant change of theNorfolk QOL- DN score ,e.g., within the variation observed between individual perfos rming a QOL-DN assessment.
In yet othe rembodiments, the methods of the invention slow the rate at which an QOL-DN score increase s,e.g., the rate of increase of a QOL-DN score in a subject treated with an RNAi agent of the invention as compared to the rate of increase of a QOL-DN score as compared to an appropriate control group showing the natural history of the disease, e.g., a placebo control group as provided, for exampl e in Adams et al., N Engl J Med 2018;379:11-21. It is understood that the rate of disease progression is dependen tupon a number of factors including, but not limited to, the severity of disease in the subject at the initiation of treatment, the duration of treatment, prior treatments, and the specific TTR mutation present, if any.
Another suitable measurement of qualit ofy life is motor strength as assessed by, for example, a NIS-W score. A NIS-W score is a composite score that summates the weaknes sof head, trunk, and limb muscles. Using the NIS (W) (referring to the portion of the scal mease uring weakness), muscl e power is assessed as norma (0)l or complete paralysis (4) with intermediate grades; 1 representing a muscle that is deemed 25% weak by clinical strength testing, 2 as 50% weak, 3 as 75% weak, 3.25 as movement agains gravitt y, 3.50 as movement with gravity eliminated, and 3.75 as muscl eflicker.
In one embodiment, the methods of the invention provide to the subject an improvement versus baseline in an NIS-W score Such an improvement can take the form of a decrease of at least 5, 6, 7, 8, 9, or 10 points of the subjec’ts NIS-W score at 18 months from the start of treatment with an RNAi agent provided herein. In othe rembodiments, the methods arrest a decrease NIS-W score, e.g., the methods result in no clinical signily ficant increas ofe the NIS-W score, or a slowing in the rate of increas ofe NIS-W score as compared to an appropriate control group showing the natural history of the disease ,e.g., a placebo control group as provided, for example in Adams et al., N Engl J Med 2018;379:11-21. It is understood that the rate of disease progression is dependent upon a number of factors including, but not limited to, the severity of disease in the subject at the initiation of treatment, the duratio nof treatment ,prior treatments, and the specific TTR mutation present, if any.
Yet anoth ersuitable indicia of quality of life is the Rasch-built Overal Disl abilit yScale (R- ODS), which is a a patient questionna iredesigned to capture activity and social participation limitations in patients. In one embodiment, the methods of the invention provide to the subject an improvement versus baseline in an R-ODS score. Such an improvement can take the form of an increas ofe at leas 2,t for example at least 2, 3, 4, or 5 points of the subject’s R-ODS score at 18 months from the start of treatment with an RNAi agent provided herein. In othe rembodiments, the methods arrest a decreasing R-ODS score ,e.g., the methods result in no clinical signily ficant decrease of the R-ODS score at 18 months from the start of treatment with an RNAi agent provided herein. In yet othe rembodiments, the methods of the invention slow the rate at which a R-ODS score decreases at 18 months from the start of treatment with an RNAi agent provided herein, e.g., the rate of decrease of a R-ODS score in a subject 22 treated with an RNAi agent of the invention as compare dto the rate of decrease of a R-ODS score as compared to an appropriate control group showing the natura histl ory of the disease, e.g., a placebo control group as provided, for example in Adams et al., N Engl J Med 2018;379:11-21. It is understood that the rate of disease progression is dependent upon a number of factors including, but not limited to, the severity of disease in the subject at the initiation of treatment, the duration of treatment, prior treatments, and the specific TTR mutation present, if any.
The composit eautonom icsymptom score (COMPASS-31), a patient questionna irethat assesses symptoms of dysautonomi autonoma icwhich provides a symptom score from 0 to 100, is another suitable indici aof quality of life. In one embodiment, the methods of the invention provide to the subject an improvement versus baseline in a COMPASS-31 score. Such an improvement can take the form of an increas ofe at least 5, for example at least 5, 6, 7, 8, 9, or 10, point sof the subjec’ts COMPASS-31 score at 18 months from the start of treatment with an RNAi agent provided herein. In othe rembodiments, the methods arrest a decreasing COMPASS-31 score, e.g., the methods result in no clinical relevantly change of the COMPASS-31 score at 18 months from the start of treatment with an RNAi agent provided herein. In yet othe rembodiments, the methods of the invention slow the rate at which a COMPASS-31 score decreases, e.g., the rate of decrease of a COMPASS-31 score at 18 months from the start of treatment with an RNAi agen tprovided herein in a subject treated with an RNAi agent of the invention as compare dto the rate of decrease of a COMPASS-31 score as compare dto an appropriate control group showing the natural history of the disease, e.g., a placebo control group as provided, for example in Adams et al., N Engl J Med 2018;379:11-21. It is understood that the rate of disease progression is dependen tupon a number of factors including, but not limited to, the severity of disease in the subject at the initiation of treatment, the duration of treatment, prior treatments, and the specific TTR mutation present, if any.
Other qualit ofy life indicia may include nutritional status (e.g., as assessed by change in median body mass index (mBMI). In one embodiment the, methods of the invention provide to the subject an improvement versus baseline in mBMI. Such an improvement can take the form of a mBMI score increas ofe at least 2, 3, 4, 5, or more at 18 months from the start of treatment with an RNAi agent provided herein. In othe rembodiments, the methods arrest a decreasing mBMI index score, e.g., the methods result in no clinical signily ficant change of the mBMI score at 18 months from the start of treatment with an RNAi agent provided herein. In yet othe rembodiments, the methods of the invention slow the rate at which mBMI score decreases, e.g., the rate of decrease of a mBMI score at 18 months from the start of treatment with an RNAi agen tprovided herein in a subject treated with an RNAi agent of the invention as compare dto the rate of decrease of a mBMI score in a subject as compared to an appropriate control group showing the natural history of the disease, e.g., a placebo control group as provided, for example in Adams et al., N Engl J Med 2018;379:11-21. It is understood that the rate of disease progression is dependen tupon a number of factors including, but not limited to, the severity of disease in the subject at the initiation of treatment, the duration of treatment, prior treatments, and the specific TTR mutation present, if any. 23 Another quality of life indicia includes assessment of exercise capacity. One suitable measure of exercise capacity is a 6-minute walk test (6MWT), which measures how far the subject can wal kin 6 minutes, i.e., the 6-minute walk distance (6MWD). In one embodiment, the methods of the invention provide to the subject an increase from baseline in the 6MWD by at least 10 meters, e.g., at least 10, 15, , or about 30 meters at 18 months from the start of treatment with an RNAi agent provided herein.
Another suitable measure is the 10-meter walk test which measures gait speed. In one embodiment, the methods of the invention provide to the subject an increas frome baseline in the 10- meter wal ktest by at least 0.025 meters/second, e.g., at leas 0.025,t 0.03, 0.04, 0.05, 0.06, 0.07, 0.08, 0.09, 1.0, 1.5, 2.0, 2.5, 3.0, 3.5, 4.0. 4.5, or about 5.0 meters/second at 18 months from the start of treatment with an RNAi agent provided herein.
In some embodiments, a change in a plasm abiomarker level is an indici aof a decreas ein ongoing nerve damage or progression of polyneuropathy in ATTR amyloidosi s.For example, a decrease in the level of neurofilament light chai n(NfL) at 9 months as compared to an NfL level at the start of treatment can be an indicia of a decrease in ongoing nerve damage or progression of polyneuropat hyin ATTR amyloidosi s.In certai nembodiments, a decrease in the level of othe rproteins, especial lyRSPO3, CCDC80, EDA2R, and NT-proBNP, either alone or in combinati onwith a decrease in NfL level at 9 months from the start of treatment as compared to its corresponding level at the start of treatment can be an indici aof a decrease in ongoing nerve damage or progression of polyneuropat hyin ATTR amyloidosi s.In certai nembodiments, an increas ine the level of N-CDase ,either alone or in combinati onwith the othe rmarkers listed above, at 9 months from the start of treatment as compare dto an its corresponding level at the start of treatment can be an indicia of a decrease in ongoing nerve damage or progression of polyneuropat hyin ATTR amyloidosi s.Further biomarkers that can act as indicia of a decrease in nerve damage or polyneuropat inhy ATTR amyloidosi suchs, as at 9 months from the initiation of treatment with an RNAi agent provided herein, are provided in Table 1. A decrease in ongoing nerve damage or progression of polyneuropat hyin ATTR amyloidosi iss correlated with a decrease in the proteins having a positive beta coefficient. A decrease in ongoing nerve damage or progression of polyneuropat hyin ATTR amyloido sisis correlated with an increas ine proteins having a negative beta coefficient. It is understood that the change in the biomarker level is a statistical ly significant change, i.e., a change larger than the inherent variabili ofty the assay.
In certai nembodiments, the methods of the invention provide an improvement in cardiovascular indicia e.g.,, increase in Kansas City Cardiomyopathy Questionnaire Overall Summary (KCCQ-OS), decreased left !ventricular (LV) wall thickening by echocardiographic assessment as compared to baseline, decreased global longitudinal strain by echocardiographic assessment as compared to baseline, decreased N-terminal prohormone B-type Natriuretic Peptide (NTproBNP) as compared to baseline, and decrease in hospitalizati dueon to cardiac event.
The methods of the present invention may also improve the prognosi sof the subject being treated. For example, the methods of the invention may provide to the subject a reduction in probability of a clinica worsenil ng event during the treatment period, or an increased longevity, or decreased hospitalizati ason compare dto an appropriate control group showing the natural history of the disease, 24 e.g., a placebo control group as provided, for example in Adams et al., N Engl J Med 2018;379:11-21.
In certai nembodiments, a reduction in the probabil itofy a clinic alworsening event during the treatment can include a decrease in all-cause mortali tyor rates of cardiovascular-relat hospited alizati ason assessed, e.g., according to the Finkelstein-Schoenfe metldhod, as compared to an appropriate control group, as provided, for example, in Maurer et al., N Engl J Med 2018:379:11-21. It is understood that the rate of disease progression is dependent upon a number of factors including, but not limited to, the severity of disease in the subject at the initiation of treatment ,the duration of treatment, prior treatments, and the specific TTR mutation present, if any.
The dose of an RNAi agent that is administered to a subject may be tailored to balance the risks and benefits of a particul ardose, for example, to achieve a desired level of inhibition of TTR gene expression (as assessed, e.g., based on TTR mRNA expression, TTR protein expression, or a reduction in an amyloid deposit, as defined above) or a desired therapeutic effect, while at the same time avoiding undesirable side effects.
In one embodiment, an iRNA agent of the invention is administered to a subject as a "fixed dose" (e.g., a dose in mg) means that one dose of an iRNA agent is used for all subject sregardless of any specific subject-relate factd ors, such as weight.
In some embodiments, the RNAi agent is administered as a fixed dose of about 25 mg to about 1000 mg, e.g., about 25 mg, about 50 mg, about 100 mg, about 200 mg, about 300 mg, about 400 mg, about 500 mg, about 600 mg, about 700 mg, about 800 mg, about 900 mg, or about 1000 mg.
In certai nembodiments, the double stranded RNAi agent is administered to the human subject about once per quarter to about once per year. In certai nembodiments, the double stranded RNAi agent is administered to the human subject about once per quarter, about once every six months, or about once per year.
In certai nembodiments, the double stranded RNAi agent is administered to the human subject at a fixed dose of about 25 mg to about 300 mg. In certain embodiments, the double stranded RNAi agent is administered to the human subject at a fixed dose of about 25 mg to about 200 mg. In certain embodiments, the double stranded RNAi agen tis administered to the human subject at a fixed dose of about 75 mg to about 200 mg. In certai nembodiments, the double stranded RNAi agent is administered to the human subject at a fixed dose of about 25 mg. In certain embodiments, the double stranded RNAi agent is administered to the human subject at a fixed dose of about 50 mg. In certai nembodiments, the double stranded RNAi agent is administered to the human subject at a fixed dose of about 75 mg. In certain embodiments, the double stranded RNAi agent is administered to the human subject at a fixed dose of about 100 mg. In certai nembodiments, the double stranded RNAi agen tis administered to the human subject at a fixed dose of about 200 mg. In certain embodiments, the double stranded RNAi agent is administered to the human subject at a fixed dose of about 25 mg to about 300 mg; about 25 mg to about 200 mg; about 75 mg to about 200 mg; about 25 mg; about 50 mg; about 100 mg; about 200mg; or about 300 mg once per quarter, i.e., about once every three months.
In certai nembodiments, the double stranded RNAi agent is administered to the human subject at a fixed dose of about 400 mg to about 600 mg. In certai nembodiments, the double stranded RNAi agent is administered to the human subject at a fixed dose of about 400 mg or about 600 mg about once every six months to about once per year. In certain embodiments, the double stranded RNAi agent is administered to the human subject at a fixed dose of about 400 mg or about 600 mg about once every six months or about once per year.
In certai nembodiments, the double stranded RNAi agent is administered to the human subject at a fixed dose of about 700 mg to about 1000 mg or about 700 mg to about 900 mg. In certain embodiments, the double stranded RNAi agen tis administered to the human subject at a fixed dose of about 700 mg, about 800 mg, about 900mg, or about 1000 mg about once per year.
In certai nembodiments, the administration is subcutaneous administration, e.g., self- administration via, e.g., a pre-fille dsyringe or auto-inject orsyringe. In some embodiments, a dose of the RNAi agent for subcutaneous administration is contain edin a volume of less than or equal to one ml of, e.g., a pharmaceutical accly eptable carrier.
Any of these schedules may optional bely repeated for one or more iterations .The number of iterations may depend on the achievement of a desired effect, e.g., the suppression of a TTR gene, retinol binding protein level vita, min A level, or the achievement of a therapeuti ceffect, e.g., reducing an amyloid deposit or reducing a symptom of a TTR-associate disead se. In certain embodiments, the iRNA agent is administered chronically, for an indefinit eperiod of time, e.g., throughout the life of the patient.
In some embodiments, the RNAi agent is administered with othe rtherapeuti cagents or other therapeuti cregimens. For example, othe ragents or other therapeuti cregimens suitable for treating a TTR-associate diseased may include a liver transplant a heart, transplant impl, antation of a pacemaker, an agent which can reduce monome TTRr levels in the body; Tafamidi s(Vyndaqel® or Vyndamax® or) AGIO, which kineticall stay bilizes the TTR tetramer preventing tetramer dissociation required for TTR amyloidogenesi nonsts; eroidal anti-inflammat drugsory (NSAIDS), e.g., diflunisal, and diuretics, which may be employed, for example, to reduce edema in TTR amyloido siswith cardiac involvement.
In one embodiment, a subject is administered an initial dose and one or more maintenance doses of an RNAi agent. The maintenance dose or doses can be the same or lower than the initial dose, e.g., one-hal off the initia ldose. Followi ngtreatment, the patient can be monitore dfor chang esin his/her condition.
In some embodiments of the methods of the invention, expression of a TTR gene as assessed by serum or plasm aTTR levels is inhibited by at least 85%, in some embodiments at least 90%. It is understood that inhibition of TTR expression using the iRNA agents provided herein would inhibi t expression of TTR in the liver and not substantially in othe rtissues, e.g., TTR expression in the eye.
The term "inhibiting," as used herein, is used interchangeabl withy "reducing," "silencing," "downregulati"ng, "suppressing", and othe rsimilar terms, and includes any level of inhibition. In some embodiments, inhibiting includes a statistical signily ficant or clinical signily ficant inhibition.
The phrase "inhibiting expression of a TTR" is intended to refer to inhibition of expression of any TTR gene including variants or mutants of a TTR gene. Thus, the TTR gene may be a wild-type TTR gene, a mutant TTR gene (such as a mutant TTR gene giving rise to amyloid deposition). 26 Inhibition may be assessed by a decrease in an absolute or relative level of one or more variables that are associated with TTR expression compare dwith a control level. The control level may be any type of control level that is utilized in the art, e.g., a pre-dose baseline level, or a level determined from a similar subject, cell, or sample that is untreated or treated with a control (such as, e.g., buffer only control or inactive agent control). As used herein, inhibition of TTR expression is typical lyassessed by determining a TTR level in an appropriate sample (e.g., historical control sample, level determined in a normal sample or clinic trialal) or before treatment of the subject with an iRNA agent, such as those provided herein or in PCT publications WO2010048228, WO2013075035, and WO2017023660, or othe ragent, e.g., antisense oligonucleot agent,ide dicer substrat eagent ,that inhibit s the expression of TTR, see, e.g., WO2011139917 and WO2015085158; and after treatment with an iRNA agent provided herein. It is understood that the iRNA agents provided herein are durabl bute slow acting. Therefore ,the level of knockdown is determined after sufficient time to reac hnadir, e.g., at least 3 weeks after first dose of the iRNA agent in a human subject, or when steady state of TTR knockdown has been achieved, e.g., after multiple doses with an iRNA agent provided herein.
Inhibition of the expression of a TTR gene may be manifeste dby a reduction of the amoun oft mRNA expressed by a first cell or group of cells (such cell mays be present, for example, in a sampl e derived from a subject) in which a TTR gene is transcribed and which has or have been treated (e.g., by contacti ngthe cel orl cell wis th an RNAi agen tof the invention, or by administering an RNAi agent of the invention to a subject in which the cells are or were present) such that the expression of a TTR gene is inhibited, as compared to a second cel orl group of cell substas ntially identical to the first cel orl group of cell buts which has not or have not been so treated (control cell(s)). In some embodiments, the percent inhibition is assessed by expressing the level of mRNA in treated cells as a percentage of the level of mRNA in control cells using, the following formula: (mRNA in control cells - )(mRNA in treated cell s) ------------------------------------------------------------------•100% (mRNA in control cells) A similar calculati mayon be performed on serum TTR protein concentrations, e.g., in a blood sample obtained froma subject ,to determine percent inhibition of expression. If no TTR is detected in the serum or plasm asampl eafter treatment ,the amoun oft TTR present is considered to be the at the lower limit of detection of the assay used.
In some embodiments, the percent inhibition is determined using a validated and clinically acceptable method.
Alternatively, inhibition of the expression of a TTR gene may be assessed in terms of a reduction of a parameter that is functiona lllinkedy to TTR gene expression, e.g., TTR protein expression, retinol binding protein level, vitamin A level or, presence of amyloid deposits comprising TTR. TTR gene silencing may be determined in any cell expressing TTR, either constitutively or by genomic engineering, and by any assay known in the art. The liver is the major site of TTR gene expression. Other significant sites of expression include the retina and choro idplexus. 27 III. iRNAs of the Invention Suitable iRNAs for use in the methods of the present invention include double stranded ribonucl eicacid (dsRNA) molecul fores inhibiting the expression of a TTR gene in a cell such, as a cel l within a subject, e.g., a mammal such, as a human having a TTR-associated disease. The dsRNA includes an antisense strand having a region of complementari whichty is complementary to at least a part of an mRNA formed in the expression of a TTR gene. The region of complementarit isy about 21- nucleotide ors less in lengt h(e.g., about 30, 29, 28, 27, 26, 25, 24, 23, 22, or 21 nucleotides in length). Upon contact with a cell expressing the TTR gene, the iRNA selectively inhibits the expression of the TTR gene (e.g., a human, a non-human primate, or a non-primate mammal TTR gene) by at least about 70% as assayed by, for example, real time PCR using the method provided in Exampl e4 of WO2013075035 when Hep3B cell ares transfected with 10 nM of the iRNA agent using the method provided therein.
A dsRNA includes two RNA strands that are complementary and hybridize to form a duplex structure under conditions in which the dsRNA will be used. One strand of a dsRNA (the antisense strand) includes a region of complementari thatty is substantially complementary, and generall fully y complementary, to a target sequence. The target sequence can be derived from the sequence of an mRNA formed during the expression of a TTR gene. The othe rstrand (the sense strand) includes a region that is complementary to the antisense strand ,such that the two strands hybridize and form a duplex structure when combined under suitable conditions. As described elsewhere herein and as known in the art, the complementary sequence sof a dsRNA can also be contain edas self- complementary regions of a singl enucleic acid molecule as, opposed to being on separate oligonucleotide. s Generally, the duplex structure is 21 to 30 base pairs in length. Similarly, the region of complementari toty the target sequenc eis 22 and 30 nucleotides in length.
A dsRNA can be synthesized by standard methods known in the art as further discussed below. iRNA compounds of the invention may be prepared using a two-step procedure. First, the individual strands of the double stranded RNA molecul aree prepared separately. Then, the component strands are annealed. The individual strands of the siRNA compound can be prepared using solution- phase or solid-phase organi csynthesi sor both. Organic synthesis offers the advantage that the oligonucleoti strande ds comprising unnatural or modified nucleotides can be easily prepared. Single - stranded oligonucleotide of thes invention can be prepared using solution-phase or solid-phase organi c synthesi sor both.
IV. Modified iRNAs of the Invention The iRNA agents for use in the methods of the invention include defined chemical modifications in the sense and antisense strand. When the lengt hof either strand is extended to provide an antisense strand longe thanr 23 nucleotides and a sense strand longer than 21 nucleotides, the nucleotides can include modifications including, but not limited to, sugar modifications, backbone modifications, and base modifications.Modificat ionsinclude, for example, end modifications, e.g., 5’- 28 end modifications (phosphorylation, conjugation, inverted linkages or) 3’-end modifications (conjugation, DNA nucleotides, inverted linkage s,etc.־); base modifications, e.g., replacement with stabilizing bases ,destabilizing bases, or bases that base pair with an expanded repertoire of partners, removal of base s(abas icnucleotides), or conjugat edbases; sugar modifications (e.g., at the 2’-position or 4’-position) or replacement of the sugar; or backbo nemodifications, including modification or replacement of the phosphodiest erlinkages .Specific examples of iRNA compounds useful in the embodiments described herein include, but are not limited to, RNAs containi ngmodified backbon ores no natural internucleoside linkages. RNAs having modified backbon incles ude, among others, those that do not have a phosphorus atom in the backbone. For the purposes of this specification, and as sometimes referenced in the art, modified RNAs that do not have a phosphorus atom in their internucleoside backbone can also be considered to be oligonucleosides. In some embodiments, a modified iRNA has a phosphorus atom in its internucleoside backbone.
Modified RNA backbone incls ude, for example, phosphorothioa chites,ral phosphorothioates, phosphorodithioates phosphotries, ters, aminoalkylphosphotriest meters,hyl and other alkyl phosphonat includinges 3'-alkylene phosphonates and chiral phosphonates, phosphinates, phosphoramidat includinges 3'-amino phosphoramidate and aminoalkylphosphorami dates, thionophosphoramidat thionoalkyes, !phosphonates, thionoalkylphosphotrie andster boranophos, sphates having normal 3'-5' linkages, 2'-5'-linked analogs of these, and those having inverted polarit whey rein the adjacent pairs of nucleoside units are linked 3'-5' to 5'-3' or 2'-5' to 5'-2'. Various salts, mixed salts and free acid forms are also included.
Modified RNA backbone thats do not include a phosphorus atom therein have backbones that are formed by short chai nalkyl or cycloalkyl internucleoside linkages, mixed heteroatom ands alkyl or cycloalkyl internucleoside linkages, or one or more short chai nheteroatom icor heterocycl ic internucleoside linkages. These include those having morpholino linkages (formed in part from the sugar portion of a nucleoside); siloxane backbones; sulfide, sulfoxide and sulfone backbones; formacetyl and thioformace tylbackbones; methylene formacetyl and thioformacet backyl bones alke; ne containi ngbackbones; sulfamat backbones;e methyleneimino and methylenehydrazino backbones; sulfonate and sulfonami debackbones; amide backbones; and others having mixed N, O, S and CH2 component parts.
In othe rembodiments, suitable RNA mimetics are contemplated for use in iRNAs, in which both the sugar and the internucleoside linkage, i.e., the backbone, of the nucleotid unitse are replaced with novel groups. The base units are maintained for hybridization with an appropriate nucleic acid target compound. One such oligomer iccompound, an RNA mimetic that has been shown to have excellent hybridization properties, is referred to as a peptide nucleic acid (PNA). In PNA compounds, the sugar backbone of an RNA is replaced with an amide containing backbone, in particul aran aminoethylglycine backbone. The nucleobases are retained and are bound directly or indirectly to aza nitrogen atoms of the amide portion of the backbone.
Some embodiments featured in the disclosure include RNAs with phosphorothioa backboneste and oligonucleosides with heteroatom backbones and, in particul ar— CH2-NH—CH2-, — CH2—N(CH3)— 29 O—CH2—[known as a methylene (methylimino) or MMI backbone —], CH2-O—N(CH3)—CH2—, — CH2- N(CH3)—N(CH3)—CH2— and — N(CH3)—CH2—CH2— of the above-referenc edU.S. Patent No. ,489,677, and the amide backbones of the above-referenc edU.S. Patent No. 5,602,240. In some embodiments, the RNAs featured herein have morpholi nobackbo nestructures of the above-referenc ed U.S. Patent No. 5,034,506. The native phosphodiester backbo necan be represented as O-P(O)(OH)- OCH2-.
Modified RNAs can also conta inone or more substituted sugar moieties. The iRNAs, e.g., dsRNAs, featured herein can include one of the following at the 2'-position: OH; F; O-, S-, or N-alkyl; O-, S-, or N-alkenyl O-,; S- or N-alkynyl; or O-alkyl-O-alkyl wherein, the alkyl, alkenyl and alkynyl can be substituted or unsubstituted C! to Cw alkyl or C2 to C10 alkenyl and alkynyl. Exemplar ysuitable modifications include O[(CH2)nO] mCH3, O(CH2).nOCH3, O(CH2)nNH2, O(CH2) nCH3, O(CH2)nONH2, and O(CH2)nON[(CH2)nCH3)]2, where n and m are from 1 to about 10. In othe rembodiments, dsRNAs include one of the followi ngat the 2' position: C! to Cw lower alkyl, substituted lower alkyl alkar, yl, aralkyl, O-alkaryl or O-aralkyl SH,, SCH3, OCN, Cl, Br, CN, CF3, OCF3, SOCH3, SO2CH3, ONO2, NO2, N3, NH2, heterocycloalkyl heteroc, ycloalkaryl amin, oalkylamin polyalkylamo, ino,substituted silyl, an RNA cleaving group, a reporter group, an intercalat or,a group for improving the pharmacokine tic properties of an iRNA, or a group for improving the pharmacodynam propeic rties of an iRNA, and other substituent shaving simila propertr ies. In some embodiments, the modification includes a 2'-methoxyethoxy (2'-O—CH2CH2OCH3, also known as 2'-O-(2-methoxyethyl) or 2'-M0E) (Martin et al., Helv. Chim. Acta, 1995, 78:486-504) i.e., an alkoxy-alkoxy group. Another exemplary modification is 2'-dimethylaminooxyethoxy, i.e., a O(CH2)2ON(CH3)2 group, also known as 2'-DMAOE, as described in examples herein below, and 2'-dimethylaminoethoxyethoxy (also known in the art as 2'-O- dimethylaminoethoxye thylor 2'-DMAEOE), i.e., 2'-O—CH2—O—CH2—N(CH3)2. Further exemplary modifications include: 5’-Me-2’-F nucleotides, 5’-Me-2’-OMe nucleotides, 5’-Me-2’-deoxynucleotides, (both R and S isomers in these three families); 2’-alkoxyalkyl; and 2’-NMA (N-methylacetamide).
Other modifications include 2'-methoxy (2'-OCH3), 2'-aminopropo xy(2'-OCH2CH2CH2NH2) and 2'-fluoro (2'-F). Simila modir fications can also be made at othe rpositions on the RNA of an iRNA, particularly the 3' position of the sugar on the 3' terminal nucleotide or in 2'-5' linked dsRNAs and the 5' position of 5' terminal nucleotide. iRNAs can also have sugar mimetics such as cyclobut moiyl eties in place of the pentofuranosyl sugar.
The RNA of an iRNA of the invention can also include nucleobase (often referred to in the art simply as "base") modifications or substitutions. As used herein, "unmodified" or "natura" lnucleobas es include the purine bases adenine (A) and guanine (G), and the pyrimidine bases thymine (T), cytosine (C) and uraci l(U). Modified nucleobases include othe rsynthetic and natura nuclel obas suches as deoxythymidine (dT), 5-methylcytosine (5-me-C), 5-hydroxymethy cytosine,l xanthine, hypoxanthine, 2-aminoadeni ne,6-methyl and othe ralkyl derivatives of adenine and guanine, 2-propyl and other alky l derivatives of adenine and guanine, 2-thiouracil, 2-thiothymine and 2-thiocytosine, 5-halouraci andl cytosine, 5-propynyl uraci land cytosine, 6-azo uracil cytosi, ne and thymine, 5-uraci l(pseudouracil), 4- thiouracil, 8-halo 8-ami, no, 8-thiol, 8-thioalkyl, 8-hydroxyl anal othe r8-substituted adenine sand guanines, 5-halo, particularl 5-bromo,y 5-trifluoromethyl and othe r5-substituted uracil ands cytosines, 7-methylguanine and 7-methyladenine, 8-azaguanine and 8-azaadenine, 7-deazaguanine and 7- daazaadeni andne 3-deazaguani neand 3-deazaadenine. Certain of these nucleobases are particula rly useful for increasin theg binding affinity of the oligomeri compoundsc featured in the invention. These include 5-substituted pyrimidines, 6-azapyrimidines and N-2, N-6 and 0-6 substituted purines, including 2-aminopropyladenine 5-propynyluracil, and 5-propynylcytosine 5-methyl. cytosine substitutions have been shown to increas nucleice acid duplex stabilit byy 0.6-1.2°C (Sanghvi, Y. S., Crooke, S. T. and Lebleu, B., Eds., dsRNA Research and Applications, CRC Press, Boca Raton, 1993, pp. 276-278) and are exemplary base substitutions, even more particularl wheny combined with 2'-O-methoxyethyl sugar modifications.
An RNAi agent of the disclosur cane also be modified to include one or more bicyclic sugar moities. A "bicycl icsugar" is a furanosyl ring modified by a ring formed by the bridging of two carbons, whether adjacent or non-adjacent atoms. A "bicycl icnucleoside" ("BNA") is a nucleoside having a sugar moiety comprising a ring formed by bridging comprising a bridge connecting two carbons, whether adjacent or non-adjacent, two carbon atoms of the sugar ring, thereby forming a bicycl icring system. In certai nembodiments, the bridge connect thes 4’-carbon and the 2’-carbon of the sugar ring, optionally via, the 2’-acycli oxygenc atom .Thus, in some embodiments an agent of the disclosur maye include one or more locked nuclei acic ds (LNA). A locked nuclei acic d is a nucleotide having a modified ribose moiety in which the ribose moiety comprise san extra bridge connecti ngthe 2' and 4' carbons. In othe rwords, an LNA is a nucleotid compe rising a bicyclic sugar moiety comprising a 4'-CH2-O-2' bridge. This structure effectively "locks" the ribose in the 3'-endo structural conformation.
The addition of locked nucleic acids to siRNAs has been shown to increas siRNe A stability in serum, and to reduce off-target effects (Elmen, J. et al., (2005) Nucleic Acids Research 33(l):439-447; Mook, OR. et al., (2007) Mol Cane Ther 6(3):833-843; Grunweller, A. et al., (2003) Nucleic Acids Research 31(12):3185-3193). Examples of bicyclic nucleosides for use in the polynucleoti desof the disclosure include without limitation nucleosides comprising a bridge between the 4' and the 2' ribosyl ring atoms.
In certai nembodiments, the antisense polynucleot agentside of the disclosure include one or more bicycl icnucleosides comprising a 4' to 2' bridge.
A locked nucleoside can be represented by the structure (omitting stereochemistry), wherein B is a nucleobase or modified nucleobase and L is the linking group that joins the 2’- carbon to the 4’-carbon of the ribose ring. 31 Examples of such 4' to 2' bridged bicycl icnucleosides, include but are not limited to 4'- (CH2)—0-2' (ENA); 4׳-(CH2)—S-2'; 4׳-(CH2)2—O-2' (ENA); 4׳-CH(CH3)—0-2׳ (also referred to as "constrained ethyl" or "cEt") and 4׳-CH(CH2OCH3)—O-2' (and analogs thereof; see, e.g., U.S. Pat.
No. 7,399,845); 4׳-C(CH3)(CH3)—O-2' (and analogs thereof; see e.g., US Patent No. 8,278,283); 4׳- CH2—N(OCH3)-2׳ (and analogs thereof; see e.g., US Patent No. 8,278,425); 4׳-CH2—O—N(CH3)-2׳ (see, e.g.,U.S. Patent Publication No. 2004/0171570); 4׳-CH2—N(R)—O-2', wherein R is H, C1-C12 alkyl or, a nitrogen protecting group (see, e.g., U.S. Pat. No. 7,427,672); 4׳-CH2—C(H)(CH3)-2׳ (see, e.g., Chattopadhyaya et al., J. Org. Chem., 2009, 74, 118-134); and 4׳-CH2—C(=CH2)-2׳ (and analogs thereof; see, e.g., US Patent No. 8,278,426). The entire contents of each of the foregoin gare hereby incorporat hereined by reference.
Additional representative US Patent sand US Patent Publications that teac hthe preparation of locke nucleicd acid nucleotides include, but are not limited to, the followin USg: Patent Nos. 6,268,490; 6,525,191; 6,670,461; 6,770,748; 6,794,499; 6,998,484; 7,053,207; 7,034,133;7,084,125; 7,399,845; 7,427,672; 7,569,686; 7,741,457; 8,022,193; 8,030,467; 8,278,425; 8,278,426; 8,278,283; US 2008/0039618; and US 2009/0012281, the entire contents of each of which are hereby incorporat ed herein by reference.
Any of the foregoing bicyclic nucleosides can be prepared having one or more stereochemical sugar configurations including for exampl ea-L-ribofuranose and -D-ribofuranos (seee WO 99/14226).
An RNAi agent of the disclosur cane also be modified to include one or more constraine ethyld nucleotides. As used herein, a "constrained ethyl nucleotide" or "cEt" is a locked nucleic acid comprising a bicyclic sugar moiety comprising a 4'-CH(CH3)-O-2' bridge (i.e., L in the preceding structure). In one embodiment, a constraine ethyld nucleotide is in the S conformati onreferred to herein as "S-cEt." An iRNA of the invention may also include one or more "conformational restricly ted nucleotides" ("CRN"). CRN are nucleotide analogs with a linker connecting the C2’and C4’ carbons of ribose or the C3 and -C5׳ carbons of ribose . CRN lock the ribose ring into a stable conformati onand increas thee hybridization affinity to mRNA. The linker is of sufficien tlengt hto place the oxygen in an optimal position for stabilit yand affinity resulting in less ribose ring puckering.
One or more of the nucleotides of an iRNA of the invention may also include a hydroxymethyl substituted nucleotide. A "hydroxymethyl substituted nucleoti"de is an acycli 2c’-3’-seco-nucleoti de, also referred to as an "unlocked nucleic acid" ("UNA") modification.
Potentiall stabily izing modifications to the ends of RNA molecul escan include N- (acetylaminocaproyl)-4-hydroxyprol (Hyp-inolC6-NHAc), N-(caproyl-4-hydroxyproli (Hyp-C6),nol N- (acetyl-4-hydroxyprol inol(Hyp-NHAc), thymidine-2'-O-deoxythymidine (ether), N-(aminocaproyl)-4- hydroxyprolinol (Hyp-C6-amino), 2-docosanoyl-uridine-3’- phosphat e,inverted base dT(idT) and others .Disclosure of this modification can be found in WO 2011/005861.
Other modifications of the nucleotides of an iRNA of the invention include a 5’ phosphate or 5’ phosphat mimie c, e.g., a 5’-terminal phosphate or phosphate mimic on the antisense strand of an RNAi 32 agent. Suitable phosphat mime ics are disclosed in, for example US Patent Publication No. 2012/0157511, the entire contents of which are incorporat hereined by reference.
V. iRNAs Conjugated to Ligands Another modificatio ofn the RNA of an iRNA of the invention involves chemically linking to the RNA one or more ligands, moieties or conjugat esthat enhance the activity, cellular distribution or cellula uptar ke of the iRNA. Such moieties include but are not limited to lipid moieties such as a cholesterol moiety (Letsinger et al., Proc. Natl. Acid. Sci. USA, 1989, 86: 6553-6556), cholic acid (Manoharan et al., Biorg. Med. Chem. Let., 1994, 4:1053-1060), a thioether, e.g., beryl-S-tritylthiol (Manoharan etal.,Ann. N.Y. Acad. Sci., 1992, 660:306-309; Manohara etn al., Biorg. Med. Chem. Let., 1993, 3:2765-2770), a thiocholesterol (Oberhauser et al., Nucl. Acids Res., 1992, 20:533-538), an aliphatic chain, e.g., dodecandiol or undecyl residues (Saison-Behmoaras et al., EMBO J, 1991, :1111-1118; Kabano etv al., FEES Lett., 1990, 259:327-330; Svinarchuk et al., Biochimie, 1993, 75:49-54), a phospholipid, e.g., di-hexadecyl-rac-glyce orrol triethyl-ammoni um1,2-di-O-hexadecyl- rac-glycero-3-phosphon (Manoharanate et al., Tetrahedron Lett., 1995, 36:3651-3654; Shea et al., Nucl.
Acids Res., 1990, 18:3777-3783), a polyamine or a polyethylene glycol chai n(Manoharan et al., Nucleosides & Nucleotides, 1995, 14:969-973), or adamantane acetic acid (Manoharan et al., Tetrahedron Lett., 1995, 36:3651-3654), a palmityl moiety (Mishra et al., Biochim. Biophys. Acta, 1995, 1264:229-237), or an octadecylamine or hexylamino-carbonyloxycholes moietteroly (Crooke et al., J.
Pharmacol. Exp. Ther., 1996, 277:923-937).
In one embodiment, a ligand alters the distribution, targeting or lifetime of an iRNA agent into which it is incorporated. In some embodiments, a ligand provides an enhanced affinity for a selected target, e.g., molecule cell, or cel type,l compartment, e.g., a cellula or rorgan compartment, tissue, organ or region of the body, as, e.g., compared to a species absent such a ligand. Exemplary ligands will not take part in duplex pairing in a duplexed nucleic acid.
Ligand scan also include targeting groups, e.g., a cel orl tissue targeting agent, e.g., a lectin, glycoprotein, lipid or protein, e.g., an antibody, that binds to a specified cel typel such as a kidney cell.
A targeting group can be a thyrotropin, melanotropi lectn, in, glycoprotein, surfactant protein A, Mucin carbohydrat multe, ivalent lactose, monovalent galactose N-ac, etyl-galactosa mineN-ac, etyl - gulucoseamine multivalent mannose, multivalent fucose, glycosylat polyed aminoaci ds,multivalent galacto se,transferrin, bisphosphonate, poly glutamat e,polyaspartate a lipid,, cholesterol, a steroid, bile acid, folate, vitamin B12, vitamin A, biotin, or an RGD peptide or RGD peptide mimetic. In certain embodiments, ligands include monovale ornt multivalent galactose In. certai nembodiments, ligands include cholesterol.
Ligand-conjugat oligonucled eotide of thes invention may be synthesized by the use of an oligonucleoti thatde bears a pendant reactive functionalit suchy, as that derived from the attachment of a linking molecule onto the oligonucleoti (descride bed below) .This reactive oligonucleoti mayde be reacted directly with commerciall availy able ligand s,ligands that are synthesized bearing any of a variety of protecting groups, or ligands that have a linking moiety attache theretd o. 33 The oligonucleotide useds in the conjugat esof the present invention may be conveniently and routinely made through the well-known technique of solid-phase synthesis. Any othe rmeans for such synthesi sknown in the art may additional orly alternatively be employed. It is also known to use similar techniqu esto prepare other oligonucleotides, such as the phosphorothioa andtes alkylated derivatives.
A. Carbohydrate Conjugates In some embodiments of the compositions and methods of the invention, an iRNA oligonucleoti furtherde comprises a carbohydrat Thee. carbohydrate conjugat ediRNA are advantageous for the in vivo delivery of nucleic acids, as well as compositions suitable for in vivo therapeutic use, as described herein. As used herein, "carbohydrate" refers to a compound which is either a carbohydra te per se made up of one or more monosaccharid unitse having at least 6 carbon atoms (which can be linear, branched or cyclic) with an oxygen ,nitrogen or sulfur atom bonded to each carbon atom ;or a compound having as a part thereof a carbohydrate moiety made up of one or more monosaccharid unites eac hhaving at least six carbon atoms (which can be linea r,branched or cyclic) wit, h an oxygen, nitrogen or sulfur atom bonded to eac hcarbon atom .Representative carbohydrates include the sugars (mono-, di-, tri- and oligosacchar containiides ngfrom about 4, 5, 6, 7, 8, or 9 monosaccharide units), and polysacchari suchdes as starches, glycogen, cellulose and polysacchari gums.de Specific monosaccharides include C5 and above (e.g., C5, C6, C7, or C8) sugars ;di- and trisaccharides include sugars having two or three monosaccharid unitse (e.g., C5, C6, C7, or C8).
In one embodiment, a carbohydrate conjugate for use in the compositions and methods of the invention is a monosacchari Inde. anothe embor diment, a carbohydrate conjugate for use in the compositions and methods of the invention is selected from the group consisting of: 34 36 O Formula XVI, O Formula XVIII, 37 In one embodiment, the monosacchari is dean N-acetylgalactosa minesuch ,as Another representativ ecarbohydrate conjugate for use in the embodiments described herein includes, but is not limited to, 38 HO Z°H AcHN HO ZOH (Formula XXIII), when one of X or ¥ is an oligonucleotide, the othe ris a hydrogen.
In certai nembodiments of the invention, the GalNAc or GalNAc derivative is attached to an iRNA agent of the invention via a monovalent linker. In some embodiments, the GalNAc or GalNA c derivative is attache tod an iRNA agent of the invention via a bivalent linker. In yet other embodiments of the invention, the GalNAc or GalNAc derivative is attached to an iRNA agen tof the invention via a trivalent linker.
In one embodiment, the double stranded RNAi agents of the invention comprise one GalNAc or GalNAc derivative attached to the iRNA agent. In anothe embor diment, the double stranded RNAi agents of the invention compris ea pluralit (e.g.,y 2, 3, 4, 5, or 6) GalNAc or GalNAc derivatives, each independentl atty ache tod a plurality of nucleotides of the double stranded RNAi agent through a plurality of monovalent linkers.
In some embodiments, for example, when the two strands of an iRNA agent of the invention are part of one larger molecule connected by an uninterrupted chai nof nucleotides between the 3’-end of one strand and the 5’-end of the respective othe rstrand forming a hairpin loop comprising, a plurali ty of unpaired nucleotides, each unpaired nucleotide within the hairpin loop may independently compris ea GalNAc or GalNAc derivative attache viad a monovalent linker. The hairpin loop may also be formed by an extended overhang in one strand of the duplex.
In some embodiments, the carbohydrate conjugate further comprises one or more additiona l ligands as described above, such as, but not limited to, a PK modulato orr a cel permeatl ion peptide.
Additional carbohydrate conjugat essuitable for use in the present invention include those described in PCT Publicati onNos. WO 2014/179620 and WO 2014/179627, the entire contents of each of which are incorporate hereind by reference.
B. Linkers In some embodiments, the conjugate or ligand described herein can be attache tod an iRNA oligonucleoti wideth various linkers that can be cleavable or non-cleavable.
The term "linker "or "linking group" means an organi moiec ty that connect twos parts of a compound, e.g., covalentl attyaches two parts of a compound. Linkers typical lycomprise a direct bond or an atom such as oxygen or sulfur ,a unit such as NR8, C(O), C(O)NH, SO, SO2, SO2NH or a chai nof 39 atoms, such as, but not limited to, substituted or unsubstituted alkyl subs, tituted or unsubstituted alkenyl subs, tituted or unsubstituted alkynyl, arylalkyl, arylalkenyl, arylalkynyl, heteroarylalkyl, heteroarylalke nyl,heteroarylalkynyl, heterocyclylal hetekyl,rocyclylalke heterocyclnyl, ylalkynyl, aryl , heteroaryl heterocyclyl,, cycloalky cycloalkl, enyl, alkylarylalkyl alkyl, arylalken alkylarylyl, alkynyl, alkenylarylal alkenylarylalkenyl,kyl, alkenylarylalkynyl alkynylarylal, alkynylkyl, arylalke nyl, alkynylarylalkyn alkylheteroarylalkyl,yl, alkylheteroarylal kenyl,alkylheteroarylal kynyl, alkenylheteroarylalkyl, alkenylheteroarylal alkenylkenyl, heteroarylalkyny alkynl,ylheteroarylalkyl , alkynylheteroarylalkenyl, alkynylheteroarylalkynyl, alkylheterocyclylal alkylhetkyl, erocyclyla lkenyl, alkylhererocyclylal alkenylhetkynyl, erocyclyla alkenylheterocyclylalkenyl,lkyl, alkenylheterocyclylalkynyl, alkynylheterocyclylal alkynylheterocyclylalkenyl,kyl, alkynylheterocyclylalkynyl, alkylaryl alke, nylaryl alkyny, laryl, alkylheteroaryl, alkenylheteroaryl, alkynylhereroa whichryl, one or more methylenes can be interrupted or terminated by O, S, S(O), SO2, N(R8), C(O), substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl, substituted or unsubstituted heterocycli wherec; R8 is hydrogen, acyl, aliphati orc substituted aliphati c.In one embodiment, the linker is between about 1-24 atoms, 2-24, 3-24, 4-24, 5-24, 6-24, 6-18, 7-18, 8-18 atoms, 7-17, 8-17, 6-16, 7-16, or 8-16 atoms.
A cleavable linking group is one which is sufficiently stable outside the cell, but which upon entry into a target cel isl cleaved to release the two parts the linker is holding together. In one embodiment, the cleavable linking group is cleaved at leas aboutt 10 times, 20, times, 30 times, 40 times, 50 times, 60 times, 70 times, 80 times, 90 times or more, or at least about 100 times faster in a target cel orl under a first reference condition (which can, e.g., be selected to mimic or represent intracellul condiar tions than) in the blood of a subject, or under a second reference condition (which can, e.g., be selected to mimic or represent conditions found in the blood or serum).
Cleavabl linkinge groups are susceptibl toe cleavage agents ,e.g., pH, redox potential or the presence of degradative molecules. Generally, cleavage agents are more prevalent or found at higher levels or activities inside cells than in serum or blood. Examples of such degradative agents include : redox agents which are selected for particul arsubstrate sor which have no substrate specificity, including, e.g., oxidative or reductive enzymes or reductive agents such as mercaptan press, ent in cells, that can degrade a redox cleavable linking group by reduction; esterases; endosomes or agents that can create an acidic environment e.g.,, those that result in a pH of five or lower; enzymes that can hydrolyz e or degrade an acid cleavable linking group by acting as a general acid, peptidases (which can be substrate specific), and phosphatases.
A cleavable linkage group, such as a disulfide bond can be susceptible to pH. The pH of human serum is 7.4, while the average intracellul pHar is slightl lowey r, ranging from about 7.1-7.3.
Endosome haves a more acidic pH, in the rang eof 5.5-6.0, and lysosomes have an even more acidic pH at around 5.0. Some linkers will have a cleavable linking group that is cleaved at a selected pH, thereby releasing a cationic lipid from the ligand inside the cell, or into the desired compartment of the cell.
A linker can include a cleavable linking group that is cleavable by a particul arenzyme. The type of cleavable linking group incorporat edinto a linker can depend on the cel tol be targeted. For 40 example, a liver-targeting ligand can be linked to a cationic lipid through a linker that includes an ester group. Liver cell ares rich in esterases ,and therefore the linker wil lbe cleaved more efficiently in liver cells than in cel ltypes that are not esterase-rich. Other cell-types rich in esterases include cell ofs the lung, renal cortex, and testis.
Linkers that contai peptin de bonds can be used when targeting cel typesl rich in peptidases, such as liver cells and synoviocytes.
In general, the suitability of a candidate cleavable linking group can be evaluate byd testing the abilit ofy a degradative agent (or condition) to cleave the candidate linking group. It will also be desirable to also test the candidate cleavable linking group for the abilit toy resist cleavage in the blood or when in contact with othe rnon-target tissue. Thus, one can determine the relative susceptibility to cleavage between a first and a second condition, where the first is selected to be indicative of cleavage in a target cel andl the second is selected to be indicative of cleavage in othe rtissues or biological fluids , e.g., blood or serum. The evaluations can be carried out in cell free systems, in cells, in cel culture,l in organ or tissue culture, or in whole animals. It can be useful to make initial evaluations in cell-fre ore culture conditions and to confirm by further evaluations in whole animals. In some embodiments, useful candidate compounds are cleaved at leas aboutt 2, 4, 10, 20, 30, 40, 50, 60, 70, 80, 90, or about 100 times faster in the cel (orl under in vitro conditions selected to mimic intracellul conditar ions) as compared to blood or serum (or under in vitro conditions selected to mimic extracellul condiar tions). i. Redox cleavable linking groups In one embodiment, a cleavable linking group is a redox cleavable linking group that is cleaved upon reduction or oxidation. An example of reductivel ycleavable linking group is a disulphide linking group (-S-S-). To determine if a candidate cleavab linkingle group is a suitable "reductivel ycleavable linking group," or for example is suitable for use with a particular iRNA moiety and particul artargetin g agent one can look to methods described herein. For example, a candidate can be evaluated by incubation with dithiothreitol (DTT), or othe rreducing agent using reagents know in the art, which mimic the rate of cleavage which would be observed in a cell, e.g., a target cel l.The candidat escan also be evaluated under conditions which are selected to mimic blood or serum conditions. In one, candidate compounds are cleaved by at most about 10% in the bloo d.In othe rembodiments, useful candidate compounds are degraded at least about 2, 4, 10, 20, 30, 40, 50, 60, 70, 80, 90, or about 100 times faster in the cel (orl under in vitro conditions selected to mimic intracellul conditar ions) as compared to blood (or under in vitro conditions selected to mimic extracellul conditiar ons). The rate of cleavage of candidate compounds can be determined using standard enzyme kinetics assays under conditions chosen to mimic intracellul mediaar and compare dto conditions chosen to mimic extracellul mediaar . ii. Phosphate-based cleavable linking groups In certai nembodiments, a cleavable linker comprises a phosphate-base clead vable linking group. A phosphate-base clead vable linking group is cleaved by agents that degrade or hydrolyze the phosphat group.e An example of an agent that cleaves phosphate groups in cell ares enzymes such as phosphata sesin cells. Examples of phosphate-based linking groups are -O- 41 P(O)(ORk)-O-, -O-P(S)(ORk)-O-, -O-P(S)(SRk)-O-, -S-P(O)(ORk)-O-, -O-P(O)(ORk)-S-, -S- P(O)(ORk)-S-, -O-P(S)(ORk)-S-, -S-P(S)(ORk)-O-, -O-P(O)(Rk)-O-, -O-P(S)(Rk)-O-, -S-P(O)(Rk)-O-, -S-P(S)(Rk)-O-, -S-P(O)(Rk)-S-, -O-P(S)( Rk)-S, wherein Rk at eac hoccurrence can be, independentl y, C1-C20 alkyl, C1-C20 haloalkyl C6-C10, aryl or, C7-C12 aralkyl. Exemplar yembodiments include - O-P(O)(OH)-O-, -O-P(S)(OH)-O-, -O-P(S)(SH)-O-, -S-P(O)(OH)-O-, -O-P(O)(OH)-S-, -S-P(O)(OH)- S-, -O-P(S)(OH)-S-, -S-P(S)(OH)-O-, -O-P(O)(H)-O-, -O-P(S)(H)-O-, -S-P(O)(H)-O, -S-P(S)(H)-O-, - S-P(O)(H)-S-, and -O-P(S)(H)-S-. In one embodiment, a phosphate-base linkid ng group is -O- P(O)(OH)-O-. These candidat escan be evaluate usingd methods analogous to those described above.
Hi. Acid cleavable linking groups In certai nembodiments, a cleavable linker comprises an acid cleavable linking group. An acid cleavable linking group is a linking group that is cleaved under acidic conditions. In some embodiments acid cleavable linking groups are cleaved in an acidic environment with a pH of about 6.5 or lower (e.g., about 6.0, 5.75, 5.5, 5.25, 5.0, or lower), or by agents such as enzymes that can act as a general acid. In a cell, specific low pH organelles such, as endosomes and lysosomes can provide a cleaving environment for acid cleavable linking groups. Examples of acid cleavable linking groups include but are not limited to hydrazones, esters, and esters of amino acids. Acid cleavable groups can have the general formula -C=NN-, C(O)O, or -OC(O). An exemplary embodiment is when the carbon attached to the oxygen of the ester (the alkoxy group) is an aryl group, substituted alkyl group, or tertiary alkyl group such as dimethyl pentyl or t-butyl. These candidates can be evaluate usind g methods analogous to those described above. iv. Ester-based linking groups In another embodiment, a cleavable linker comprises an ester-based cleavable linking group.
An ester-based cleavable linking group is cleaved by enzymes such as esterases and amidases in cell s.
Examples of ester-based cleavable linking groups include but are not limited to esters of alkylen e, alkenyle neand alkynylene groups. Ester cleavable linking groups have the general formula -C(O)O-, or -OC(O)-. These candidat escan be evaluated using methods analogous to those described above. v. Peptide-based cleaving groups In yet anoth erembodiment, a cleavable linker comprises a peptide-based cleavable linking group. A peptide-based cleavable linking group is cleaved by enzymes such as peptidases and proteases in cells. Peptide-based cleavable linking groups are peptide bonds formed between amino acids to yield oligopeptides (e.g., dipeptides, tripeptides etc?) and polypeptides .Peptide-based cleavable groups do not include the amide group (-C(O)NH-). The amide group can be formed between any alkylene, alkenyle neor alkynelene. A peptide bond is a special type of amide bond formed between amino acids to yield peptides and proteins. The peptide based cleavage group is generally limited to the peptide bond (i.e., the amide bond) formed between amino acids yielding peptides and proteins and does not include the entire amide functional group. Peptide-based cleavab linkingle groups have the general formula - NHCHRAC(O)NHCHRBC(O)-, where RA and RB are the R groups of the two adjacent amino acids. These candidates can be evaluated using methods analogous to those described above. 42 In one embodiment, an iRNA of the invention is conjugat edto a carbohydrate through a linker.
Non-limiting examples of iRNA carbohydrate conjugates with linkers of the compositio nsand methods of the invention include, but are not limited to, (Formula XXVII), 43 (Formula XXVIII), (Formula XXIX), (Formula XXXI), when one of X or ¥ is an oligonucleotide, the othe ris a hydrogen.
In certai nembodiments of the compositions and methods of the invention, a ligand is one or more "GalNAc" (N-acetylgalactosam derivativesine) attached through a bivalent or trivalent branched linker.
In one embodiment, a dsRNA of the invention is conjugat edto a bivalent or trivalent branched linker selected from the group of structures shown in any of formula (XXXII) - (XXXV): 44 Femtila XXXII Fomala XXXIII Formul aXXXIV wherein: q2A, q2B, q3A, q3B, q4A, q4B, q5A, q5B and q5C represent independentl fory eac hoccurrence q2A, q2B, q3A, q3B, q4A, q4B, q5A, q5B and q5C represent independentl fory each occurrence 0-20 and wherein the repeating unit can be the same or different; p2A p2B p3A p3B p4A p4B p5A p5B p5C p2A p2B p3A p3B p4A p4B p4A 56ך־י p5C each independentl fory eac hoccurrenc absente CO,, NH, O, S, OC(O), NHC(O), CH2, CH2NH or CH,O; q2a q2b q3a q3b q4a q4b, q5a q5b q5c independentl fory each occurrence absent alkylene,, substituted alkylene wherin one or more methylenes can be interrupted or terminated by one or more of O, S, S(O), SO2, N(RN), C(R’)=C(R"), C=C or C(O); R2A, R2b, R3a, R3b, R4a, R4b, R5a, R5b, R5c are eac hindependentl fory each occurrence absent NH, , O, S, O ho-L H 1 CH2, C(O)O, C(O)NH, NHCH(Ra)C(O), -C(O)-CH(Ra)-NH-, CO, CH=N-O, O _ _ R—R s—s or heterocyclyl; L2a, p2B, l3a؛ p3B, £،4a־ p4B, j^5a, j^5b a1K، j^5c 1ep1esent the ligand; i.e. eac hindependentl fory eac hoccurrence a monosacchari (suchde as GalNAc) disac, charide, trisacchari de,tetrasaccharid e, oligosacchar oride, polysaccharide andR; a is H or amino acid side chain.Trivalent conjugating GalNAc derivatives are particularly useful for use with RNAi agents for inhibiting the expression of a targe t gene, such as those of formula (XXXVI): 45 Formula XXXVI -|5־A |_5A 5B-|5־B [_5B ____ t5C I 5C q5c י wherein L5A, L5B and L5C represent a monosaccharide, such as GalNAc derivative.
Examples of suitable bivalent and trivalent branched linker groups conjugati ngGalNA c derivatives include, but are not limited to, the structures recited above as formula II,s VII, XI, X, and XIII.
Representative U.S. patent sthat teac hthe preparation of RNA conjugates include, but are not limited to, U.S. Pat. Nos. 4,828,979; 4,948,882; 5,218,105; 5,525,465; 5,541,313; 5,545,730; 5,552,538; ,578,717, 5,580,731; 5,591,584; 5,109,124; 5,118,802; 5,138,045; 5,414,077; 5,486,603; 5,512,439; 5,578,718; 5,608,046; 4,587,044; 4,605,735; 4,667,025; 4,762,779; 4,789,737; 4,824,941; 4,835,263; 4,876,335; 4,904,582; 4,958,013; 5,082,830; 5,112,963; 5,214,136; 5,082,830; 5,112,963; 5,214,136; ,245,022; 5,254,469; 5,258,506; 5,262,536; 5,272,250; 5,292,873; 5,317,098; 5,371,241, 5,391,723; ,416,203, 5,451,463; 5,510,475; 5,512,667; 5,514,785; 5,565,552; 5,567,810; 5,574,142; 5,585,481; ,587,371; 5,595,726; 5,597,696; 5,599,923; 5,599,928 and 5,688,941; 6,294,664; 6,320,017; 6,576,752; 6,783,931; 6,900,297; 7,037,646; 8,106,022, the entire contents of eac hof which are hereby incorporat hereined by reference.
It is not necessary for all positions in a given compound to be uniforml ymodified, and in fact more than one of the aforementioned modifications can be incorporat edin a single compound or even at a singl enucleoside within an iRNA. The present invention also includes iRNA compounds that are chimeric compounds.
VI. Delivery of an iRNA of the Invention The delivery of an iRNA of the invention to a cel e.g.,l a cel witl hin a human subject (e.g., a subject in need thereof, such as a subject having a disease ,disorder or condition associated with contact activation pathway gene expression) can be achieved in a number of different ways . For example, delivery may be performed by contacti nga cel wil th an iRNA of the invention either in vitro or in vivo.
In vivo delivery may also be performed directly by administerin ga composition comprising an iRNA, e.g., a dsRNA, to a subject. Delivery may be performed, for example, by intravenous administration or subcutaneous administration. In certain embodiments, the iRNA agent is delivered by subcutaneous administration. In certain embodiments, the iRNA agent is administered by self-administrati onusing a pre-filled syringe or auto-inject ordevice.
In general any, method of delivering a nucleic acid molecu le(in vitro or in vivo) can be adapted for use with an iRNA of the invention (see e.g., Akhtar S. and Julia nRL. (1992) Trends Cell. Biol. 46 2(5): 139-144 and WO9402595, which are incorporat edherein by reference in their entireties). For in vivo delivery, factors to consider in order to deliver an iRNA molecu leinclude, for example, biological stability of the delivered molecul preventione, of non-specific effects, and accumulati ofon the delivered molecule in the target tissue.
VII. Pharmaceutical Compositions of the Invention The present invention also includes pharmaceutical compositions and formulations which include the iRNAs described herein for use in the methods of the invention. In one embodiment , provided herein are pharmaceutical compositions containi ngan iRNA, as described herein, and a pharmaceutical accly eptable carrier. The pharmaceutical compositions containing the iRNA are useful for treating a disease or disorder associate wid th the expression or activity of a TTR gene. Such pharmaceutical compositions are formulated based on the mode of delivery. One example is compositions that are formulated for systemic administration via parentera delil very, e.g., by subcutaneous (SC) or intravenous (IV) delivery. The pharmaceutical compositions of the invention may be administered in dosages sufficien tto inhibit expression of a TTR gene. In one embodiment, the iRNA agents of the invention, e.g., a dsRNA agent ,is formualted for subcutaneous administration in a pharmaceutical accly eptable carrier In othe rembodiments, a singl edose of the pharmaceutical compositions can be long lasting, such that subsequent doses are administere dat 1 month intervals at, not more than 1, 2, 3, or 4 month intervals or, at quarterl (abouty every three months) intervals. In some embodiments of the invention, a single dose of the pharmaceutical compositio nsof the invention is administered once per month .In othe rembodiments of the invention, a single dose of the pharmaceutical compositions of the invention is administered once every othe rmonth . In othe rembodiments, a single dose of the pharmaceutical compositions of the invention is administered quarterl y,i.e., about once every three months .In othe r embodiments, a singl edose of the pharmaceutical compositions of the invention is administered once every four months. In anothe embr odiment, a singl edose of the pharmaceutical compositions of the invention is administered once every five months. In othe rembodiments, a single dose of the pharmaceutical compositions of the invention is administered once every six months. In othe r embodiments, a singl edose of the pharmaceutical compositions of the invention is administered once every twelve months.
VIII. Kits The present invention also provides kits for performing any of the methods of the invention.
Such kits include one or more double stranded RNAi agent(s) and a label providing instructions for use of the double-stranded agent(s) for use in any of the methods if the invention. The kits may optionall y further compris emeans for contacti ngthe cel wil th the RNAi agent (e.g., an injection device or an infusion pump), or means for measuring the inhibition of TTR (e.g., means for measuring the inhibition of TTR mRNA or TTR protein). Such means for measuring the inhibition of TTR may comprise a 47 means for obtaining a sample from a subject, such as, e.g., a plasm asample. The kits of the invention may optional furtherly comprise means for administering the RNAi agent(s) to a subject or means for determining the therapeuticall effectiy ve or prophylacticall effectiy ve amount.
The RNAi agent may be provided in any convenient form, such as a solution in sterile water or othe rappropriate solutio n,e.g., PBS, normal saline, 5 mM phosphat buffer,e for resuspension and injection. For example, the RNAi agent may be provided as a 300 mg, 200 mg, 100 mg, or 50 mg vial with water or othe rappropriate solution in sterile water for injection. In certain embodiments, the RNAi agent is provided in a kit for self administration comprising a pre-filled syringe or autoinjector containi ng300mg, 200 mg, 100 mg, or 50 mg of the RNAi agent in an appropriate volume of excipien t for administratio n,optionall furtyher including instructions for use. In certain embodiments, the RNAi agent may be provided in multiple vial sor devices for administration of the dose by multiple injections to be given at about the same time, e.g, within one week, within one day, within one hour.
IX. Diagnosis of TTR amyloidosis polyneuropathy and assessment of disease burden TTR amyloido sisis a complex, multifactoria diseal se. An expanding list of criteria have been used to monitor the progression of TTR-FAP: neuropathy impairment score (NIS), NIS + 7, and modified NIS (mNIS) + 7 and mNIS + 71oniS. These diagnosti cric teria are wel lknown in the art and highlights of the criteria are provided below .As used herein, meeting the diagnosti cric teria of TTR- FAP is understood as meeting FAP stage 1 criteria, with or without the presence of a mutation associate wid th hereditary TTR-FAP. Progression of indicators of neuropathy is considered an increase of at least two points in modified neuropathy impairment score (mNIS) + 7.
Familial Amyloid Polyneuropathy (FAP) Stage Coutinho et al. developed a clinica staginl gsystem for the neuropathy symptoms of hATTR (formerly termed familial amyloid neuropathy). The scal rangese from 1 to 3, as follo ws(Ando et al.
Orphanct J Rare Dis. 2013;8:31): FAP Stage 1: Walking without assistance mi, ld neuropathy (sensory, autonomic, and motor )in lower limbs.
FAP Stage 2: Walking with assistance moderate, impairment in lower limbs, trunk, and upper limbs.
FAP Stage 3: wheelchair or bed-ridden, severe neuropathy.
A subjec twith no neuropathy is considered to be FAP Stage 0.
Neuropathy Impairment Scoring Methods Methods to assess neuropathies are known in the art. For example, in the Mayo Clinic Neurologi Examic nation Sheet and also in the weaknes ssubscores of Neuropath yImpairment Score (NIS), weakness (NIS-W) are scored in 25% decrements from 1 to 4 points and separately for major muscle groups of each side of the body (Dyck et al., Quantitating overal neuropathicl symptoms, impairments, and outcomes. In: Dyck PJ, Thomas PK, editors. Periphera lneuropathy. 4th ed.
Philadelphia: Elsevier; 2005. p. 1031-52). A broad group, especial lyof cranial proxima, l, and distal limb muscles, is evaluated in NIS-W with a maximum score of 192 points. A decreas eof the major 5 48 muscle stretch reflexes is usuall assy essed by neurologists and touch pressure, vibration, joint motion, and pin-prick sensation ofs feet and hands are scored in 25% decrements and from 1 to 4 in the Mayo Clinic Neurology Examination Sheet. To complete NIS of reflexes (NIS-R) and of sensation (NIS-S) Mayo Clinic record score sare transformed to NIS point score s(i.e., Mayo Clinic scores of 1 or 2 are given an NIS point score of 1 and Mayo Clinic scores of 3 or 4 are given an NIS score of 2). Therefore, the maximal NIS score sof the usual reflexes evaluated by neurologist (NISs -R) are 5 x 2 x 2 = 20 points and of the 4 modalities of sensation often evaluate byd neurologists (NIS-S) are 8 x 2 x 2 = 32 points. Therefore ,the maximum NIS score is: 192 + 20 + 32 =244 points. The NIS has been described in previous publications (Dyck et al. 2005 and Dyck et al., Neurol. 1997;49:229-39).
NIS + 7 has been used as the primary or co-primary outcome measure in the trials of diabetic sensorimotor polyneuropath TTRy, FAP, and othe rgeneralized sensorimotor polyneuropathies (N.
Suanprasert et al. J Neurol Sci 344 (2014) 121-128). NIS + 7 adequately assesses graded severities of muscle weaknes sand muscl estretch reflex abnormalit wiyth only minima lceiling effects for reflexes.
In NIS + 7, 5 of the 7 tests are attributes of nerve conduction — expressed either as normal deviates (Z scores) or points. The attributes included in NIS + 7 were chosen because their abnormal sensity itivel y detects diabetic sensorimotor polyneuropat hy(Dyck et al. Muscl eNerve 2003;27(2):202-10). The attributes included are the peroneal nerve compound muscle action potential (CMAP) amplitude, motor nerve conduction velocity (MNCV), and motor nerve distal latency (MNDL), tibia lMNDL and sural sensory nerve action potential (SNAP) amplitudes. Their measured values can be transforme dto normal deviates from percentile values correcting for applicabl varie able ofs age, gender, height, or weight as based on earlier studies of a large healthy subject reference cohort. Additional ly,these percentil valuese can be expressed as NIS points from obtained percentil valuese (i.e., N5th = 0 points; <5th- N 1st = 1 point and Assessment of weakness and reflex abnormal itassy, essment of sensation loss, autonomic dysfunction, and neurophysiologic test abnormalit areies not adequately assessed by NIS + 7 for use in trials of TTR FAP. In NIS + 7 sensation loss is not optimal lyassessed: 1) body distribution of sensation loss is not adequately taken into account, 2) large as compare dto smal fiberl sensory loss is over emphasized and 3) improved methods of testing and comparison to reference values are preferred over clinic alassessments .Also ,autonomic dysfunction is not adequately assessed by the use of only heart rate deep breathing (HRdb). The attributes of nerve conduction used to assess NIS + 7 are not ideal for the study of TTR FAP.
Modified neuropathy impairment score +7 (mNIS+7), and updated version of NIS + 7, is a composite score measuring motor strength, reflexes, sensation, nerve conduction, and autonomic function. Two versions of this composit emeasure were adapted from the NIS+7 to better reflect hATTR amyloido siswith polyneuropat hyand have been used as primary outcomes in inotersen and patisira n clinic trialal s. Key differences between these two versions, and the othe rneuropathy scoring systems, are summarized in the table below (from Adams et al., BMC Neurology, volum e17, Article number : 181 (2017)). In both scales, a lowe rscore represents better neurologic function (e.g. an increas ine score reflect sworsening of neurologic impairment). 49 Neuropathy Impairment Score Criteria NIS+7 mNIS+7Ionis MS I I NIS m-NlS+7 Total score 88 244 270 304 346.3 Assessment (score) Neurologic exam Neurologic Motor Neurologic exam Neurologic exam [lowe r exam Neurologic exam (192) strength/weakness (192) (192) limbs (192) only] (64) Neurologic exam Neurologic Neurologic exam Neurologic exam Reflexes [lowe r Neurologic exam (20) exam (20) (20) (20) limbs only] (8) QST - heat pain QST - heat pain and touch and touch pressure pressure at at multiple sites multiple sites (80) (80) Neurologic Sensation exam Neurologic Neurologic exam - [lowe r Neurologic exam (32) exam (32) (32) limbs Only] (16) Vibration detection threshold - - - (3.7) 2:5 - ulnar CMAP S5 - ulna CMAPr S5 - sural SNAP/ fibular nerve and SNAP, CMAP. tibia lmotor nerve distal and SNAP؛ Composite nerve latenc motory, nerve conduction - - peronealc CMAP, peroneal0 CMAP, conduction seore velocity motor, nerve distal tibial CMAP, tibial CMAP, sural latenc (18.6)y a sural SNAP (10) b SNAP (18.6)3 Heart rate Heart rate response to deep Postural blood Autonomic - - response:to deep pressure (2) function breathing (3.7)a breathin (3.7)g a 1. CMAP compound muscle action potential exa; m examination; mNTS+7 modified NIS+7; NIS Neuropathy Impairment Score; NIS-LL NIS based on examination of lower limbs only; QST quantitative sensory testing; SNAP sensory nerve action potential 2. 3Score expressed as normal deviates (0-3.72) based on healthy-subjec part ameters 3. ',Score graded according to defined categories: normal (95th percentile =) 0 points; mildly reduced (>95thto <99th percentile) = ! point; and very reduced (>99th percentile) = 2 points 4. cMay also be referred to as fibular Diagnosis of TTR amyloidosis cardiomyopathy (ATTR-CM) and assessment of disease burden Patients with hATTR amyloido sisand cardiomyopathy typically experience progressive symptoms of heart failure (HF) and cardiac arrhythmias, with death typical lyoccurring 2.5 to 5 years after diagnosis. Cardiac infiltration of the extracellul matar rix by TTR amyloid fibrils leads to a progressive increas ofe ventricula wallr thickness and a marked increas ine chambe stir ffness, resulting in impaired diastoli functic on. Systolic function is also impaired, typical lyreflected by abnormal longitudin alstrain despite a normal ejection fraction, which is preserved until late stages of the disease.
In patients with ATTR amyloidosi ands light-chai (AL)n cardiac amyloidosi boths, longitudin alstrain and N-terminal prohormone of brain natriureti cpeptide (NT-proBNP) have been shown to be independent predictors of survival.
Echocardiography is routinely used to assess cardiac structure and function; parameter spre- specified in the statistical analysi plans include mean left ventricula (LV)r wall thickness, LV mass , longitudin alstrain, and ejection fraction. Cardiac output, left atrial size, LV end-diastolic volume 50 (LVEDV), and LV end-systol icvolume (LVESV). Echocardiograms are routinely used for cardiac imaging. Myocardial strain can be assessed with speckl etracking using vendor-independe ntsoftware (TOMTEC, Munich, Germany). Analysis of NT-proBNP and troponi nI levels is routinely performed in clinic laboratal ories using commerciall availy able diagnostic tests, e.g., using chemiluminescence assays (Roche Diagnost icCobas, Indianapolis IN,, USA for NT-proBNP; Siemens Centaur XP, Camberley, Surrey, UK for troponi nI). Similarly, clinical practice routinely includes measurement of creatinine levels and estimated glomerul arfiltration rate (eGFR) based on creatinine level s,e.g., using the Modification of Diet in Renal Disease study formula.
A review providing screening and diagnostic methods for ATTR-CM was recently published by Witteles et al., 2019 (JACC: Heart Failure, 2019. 7:709-716) which provides information on methods of diagnos isincluding a list of "red flags" suggesting the presence of ATTR-CM and screening methods including echocardiography, electrocardiogra phy,cardiac magnetic resonance, the presence of systemic symptoms involving the peripheral or autonom icnervous system along with cardiac dysfunction including bilateral sensory motor polyneuropat hythat begins in the lower limbs and follo wsan ascending pattern, dysautonom iain the form of orthostati hypotension,c diarrhea/ constipation, and erectile dysfunction, and eye involvement such as glaucoma, intravitreal deposition, and scalloped pupils; carpal tunnel syndrome ,especial lybilateral carpal tunnel syndrome, lumbar spinal stenosis, and bicep tendon rupture. Other diagnostic methods include bone scintography with technetium (Tc)- labelled bisphosphonates localize tos TTR cardiac amyloid deposits for reasons that are not known.
Biopsy is also used to confirm the presence of TTR amyloido sisin heart.
Methods for assessment and classificat ionof cardiac function of the parameter sprovided above are known in the art. As used herein, the specific method of assessment or classificati ofon cardiac function may be any clinicall accy eptable standard to demonstrate sufficiently decreased cardiac function such that the standard of care includes a medical intervention, e.g., administration of a pharmacological agent ,surgery.
Serum Biomarkers as an Indicia of Nerve Damage and TTR Amyloidosis Progression The diagnosti cand monitoring methods set forth above are complex and often subjective.
Moreover, as TTR amyloidos isis rare, and the signs and symptoms above can be present in a number of othe rdiseases ,clinical validaly ted, non-invasive plasm abiomarkers may facilitate earlier diagnos isand aid monitoring of disease progression. In a study by Tica uet al., 2019 (see www.medrxiv.org/content/10.1101/19011155v2.full.pdf) plasm alevels of >1000 proteins were measured in patients with hATTR amyloido siswith polyneuropat hywho received either placebo or patisiran in the phase 3 APOLLO study (NCT01960348) and in a cohort of healthy individual s.The impact of treatment with patisiran, a lipid formulate RNAid agent that inhibits the hepati cexpression of TTR, on the time profile of each protein was determined by a linea mixer d model at 0, 9, and 18 months. Neurofilame ntlight chai n(NfL) protein was further assessed using an orthogonal quantitati ve approach. A significant change in the levels of 66 proteins was observed with patisiran vs placebo, with change in NfL, a marker of neuronal damage, most significant Analysis. of the changes in protein level s 51 demonstrated that the proteome of patients treated with patisira ntrended towards healthy individual ats 18 months. Plasma NfL levels in healthy controls were four-fold lower than in patients with TTR amyloido siswith polyneuropat hy(16.3 [SD 12.0] pg/mL vs 69.4 [SD 42.1] pg/mL, p< 10 16). Levels of NfL at 18 months increased with placebo (99.5 [SD 60.1] pg/mL) and decreased with patisiran treatment (48.8 [SD 29.9] pg/mL). At 18 months, improvement in modified Neuropathy Impairment Score+7 (mNIS+7) in patisiran-treat edpatients significantly correlat edwith a reduction in NfL level s (R=0.43, p<10 7). NfL reduction with patisiran treatment correlat edwith improvement in mNIS+7 suggests it may serve as a biomarker of nerve damage and polyneuropat hyin TTR amyloidosi Thiss. biomarker may enable earlier diagnos isof polyneuropat hyin patients with hATTR amyloidos andis facilitate monitoring of disease progression.
A decrease in the level of othe rproteins ,especiall RSPO3,y CCDC80, EDA2R, and NT- proBNP, were found to correlat wie th an improvement in mNIS+7, suggesting that, either alone or in combinati onwith eac hothe ror Nfl ,they may serve as a biomarker of nerve damage and polyneuropat hyin ATTR amyloidosi Ans. increas ine the level of N-CDase was found to correlat wite h an improvement in mNIS+7, suggesting that, either alone or in combinati onwith the other markers listed above, it may serve as a biomarker of nerve damage and polyneuropat hyin ATTR amyloidosis.
Further potential biomarkers are listed in the tabl belowe which were observed to change in response to treatment with patisiran. When a subject has an increas ine the level of a protein having a positive beta coefficient relative to a reference level indicative of progression of ATTR amyloidosis, and when a subject has a decrease in the level of a protein having a negative beta coefficient relative to a reference level indicative of progression of ATTR amyloidosi Changess. in the level of one or more of these markers can be indicia of improvement, stabilizatio orn, decrease in ongoing nerve damage and progression of polyneuropat hyin ATTR amyloidosis. 52 Table 1. Biomarkers with changed levels in response to patisiran treatment Protein -loglO (p-value) -!or10 (p-value) beta coeffident Protein beta coefficient 0.55 20.40 LDLR -0.20 5.85 M.
RSPO3 0.53 18.43 NOV 0.19 5.83 CCDC80 0.40 17.28 CD160 0.16 5.69 0.25 EDA2R 17.10 P13 0.19 5.61 -0.25 15.17 SMOC1 0.12 5.61 N-CPsse 0.62 13.15 TFPf-2 0.22 5.54 NT-9rQ^P. 11.43 0.46 LEP -0.38 5.49 WNT9A 0.20 10.78 TNFRSF19 0.16 5.47 SMOC2 0.14 10.33 ERB83 -0.07 5.44 PTN 0.39 9.20 DUA -0.14 5.43 HGF 0.17 9.02 IL-4RA 0.13 .33 0.14 6.13 REN -0.24 5.31 NELLI -0.16 8.98 CES2 -0.15 5.23 DCN CR2 0.10 8.90 0.15 5.23 ARSA -0.23 8.86 PSG1 0.21 5.20 KLK4 0.27 8.66 FGF-BP1 0.11 5.19 GPC1 8.57 0.18 OPN 0.18 .18 SFRP 0.21 8.41 TFF3 0.17 5.08 DRAXIN 0.20 8.11 ANGPT2 0.15 5.05 IL-18R1 -0.15 7.82 CCL24 -0.17 5.05 BNP 0.48 7.81 LAYN 0.13 5.02 GFR-alphal 0-17 7.69 FUCA1 -0.11 4.97 CXCL9 0.31 7.42 AXL 0.11 4.95 TIMD4 -0.21 7.02 TLR3 -0.10 4.94 RSPO1 0.16 6.94 SCARB2 0.12 4.90 6.94 MYOC 0.21 B4GAT1 -0.09 4.83 WFDC2 0.11 6.76 SORCS2 0.15 4.65 GUSB -0.27 6.71 CD300E 0.18 4.63 HSPB6 0.19 6.70 CD27 0.10 4.62 MFGE8 -0.21 6.68 SPON1 0.10 4.58 SMPD1 -0.15 6.59 IGFBP-2 0.15 4.58 FLT1 0.09 6.29 DNER -0.07 4.57 RARRES1 CTSD -0.13 6.06 0.09 4.57 CO209 -0.11 5.85 Dkk-4 0.15 4.49 Sequences for these biomarkers are provided herein as SEQ ID NOs: 17-34.
This invention is further illustrated by the followin examg ples which shoul notd be construe das limiting. The contents of all references and published patent sand patent applications cited throughout the application, as well the Sequence Listing are hereby incorporat edherein by reference. 53 EXAMPLES Exemplar ydouble straded RNAi agents for use in the methods of the invention are provided in Table 3 below .Tabl 2,e below, provides the abbreviation ofs the nucleotid monomere sand ligands used in nucleic acid sequenc erepresentation. It will be understood that these monomers, when present in an oligonucleotide, are mutually linked by 5'-3'-phosphodiest erbonds unless otherwise indicated.
Table 2. Abbreviations of nucleotide monomers and ligands Abbreviation Nucleotide(s) and ligands A Adenosine- ’3 -phosphate Af 2 ’ -fluoroadenosine-3 ’ -phosphate Afs 2 ’ -fluoroadenosine-3 ’ -phosphorothioate As adenosine-3 ’ -phosphorothioate C cytidine-3 ’ -phosphate Cf 2 ’ -fluorocytidine-3 ’ -phosphate Cfs 2 ’ -fluorocytidine-3 ’ -phosphorothioate Cs cytidine-3 ’ -phosphorothioate G guanosine -3’ -phosphate Gf 2 ’ -fluoroguanosine-3 ’ -phosphate Gfs 2 ’ -fluoroguanosine-3 ’ -phosphorothioate Gs guanosine -3’ -phosphorothioate T ’ -methyluridine- ’3 -phosphate Tf 2 ’ -fluoro-5 -methyluridine-3 ’ -phosphate Tfs 2 ’ -fluoro-5 -methyluridine-3 ’ -phosphorothioate Ts 5-methyluridine- ’ 3-phosphorothioate U Uridine-3 ’ -phosphate Uf 2 ’ -fluorouridine-3 ’ -phosphate Ufs 2 ’ -fluorouridine -3 ’ -phosphorothioate Us uridine -3’-phosphorothioate N any nucleotide (G, A, C, T or U) a 2'-O-methyladenosine-3 ’ -phosphate as 2'-O-methyladenosine-3 ’ - phosphorothioate c 2'-O-methylcytidine-3 ’ -phosphate cs 2'-O-methylcytidine-3 ’ - phosphorothioate 2'-O-methylguanosine-3 ’ -phosphate g 2'-O-methylguanosine-3 ’ - phosphorothioate gs t 2 ’ -O-methyl-5-methylthymine-3 ’ -phosphate ts 2’-O-methyl-5-methylthymine-3’-phosphorothioate u 2'-O-methyluridine-3 ’ -phosphate 54 Abbreviation Nucleotide(s) and ligands us 2'-O-methyluridine-3 ’ -phosphorothioate s phosphorothioat linkae ge N-[tris(GalNAc-alkyl)-amidodecanoyl)]-4-hydroxyprolinol L96 Hyp-(GalNAc-alkyl)3 HO /0H ——0 H H HO AcHN 0 1 H0'- H° <°H CL ho AcHN Q 0 00 ^־ HC^ J AcHN H H (Agn) Adenosine-glycol nucleic acid (GNA) (Cgn) Cytidine-glycol nucleic acid (GNA) (Ggn) Guanosine-glycol nucleic acid (GNA) thymidine-glycol nucleic acid (GNA) S-Isomer (Tgn) Table 3. Modified nucleotide sequences of sense and antisense strands of RNAi agents targeted to TTR SEQ ID Duplex ID Strand Modified Oligonucleotide Sequence (5’-3’) NO: AD-65492 sense usgsggauUfuCfAfUfguaaccaagaL96 10 antisense usCfsuugGfuuAfcaugAfaAfucccasusc 11 AD-87400 sense usgsggauUfuCfAfUfguaaccaagaL96 10 antisense usCfs(Tgn)ugGfuuAfcaugAfaAfucccasusc 12 AD-87401 sense usgsggauUfuCfAfUfguaaccaagaL96 10 antisense usCfsu(Tgn)gGfuuAfcaugAfaAfucccasusc 13 AD-87402 sense usgsggauUfuCfAfUfguaaccaagaL96 10 antisense usCfsuu(Ggn)GfuuAfcaugAfaAfucccasusc 14 AD-87403 sense usgsggauUfuCfAfUfguaaccaagaL96 10 antisense usCfsuug(Ggn)uuAfcaugAfaAfucccasusc AD-87404 sense usgsggauUfuCfAfUfguaaccaagaL96 10 antisense usCfsuugGf(Tgn)uAfcaugAfaAfucccasusc 7 AD-87405 sense usgsggauUfuCfAfUfguaaccaagaL96 10 antisense usCfsuugGfu(Tgn)AfcaugAfaAfucccasusc 16 55 Example 1: TTR Protein Knockdown by RNAi agents in the V30M Transgenic Mouse The V30M mutation is a common amyloidogeni mutc ation in human TTR. Transgenic mice lacking mouse TTR and expressing human TTR with a V30M mutation were used in the study. Mice (n = 3 per group) were administered a singl esubcutaneous 1 mg/kg dose of RNAi agent AD-65492, previousl ydisclosed in WO2018112320, or othe rRNAi agents based on the sequenc eand chemistry of AD-65492 in which a chemical modificatio atn a single position of the antisense strand was changed to a GNA modification as shown in Table 2 above. Bloo sampld es were obtained on days 0 (pre-dose), 3, 7, 10, 14, 21, 35, and 49. Serum was prepared and human TTR level swere determined using ELISA assay (see, e.g., Coelho, et al. (2013) N Engl J Med 369:819). Relative TTR levels as compared to Day 0 are shown in Figure 1.
Incorporation of a GNA at position 7 or 8 in the antisense strand was wel ltolerated (AD-87404 and AD-87405). Simila kiner tics and maximum protein knockdow wern e simila tor the parent RNAi agent AD-65492. Durability of knockdow byn AD-87404 was simila tor AD-65492. Incorporati onof a GNA at positions 3-6 in the antisense strand was less well tolerated.
Example 2: TTR Protein Knockdown by RNAi agents in Non-Human Primate The sequenc eof the iRNA agents in Tabl 2e is fully cross-reactive with cynomolgus monkey TTR. A singl esubcutaneous dose of AD-65492 (1 mg/kg) or AD-87404 (1 mg/kg or 3 mg/k) was administered to cynomolgus monkey (n = 3 per group) on day 0 in three separate studies. Blood samples were collect variousled ony Days -7 (7 days predose) through Day 119 as shown in Figure 2. Serum was prepared and cynomolgus monkey TTR levels were determined using ELISA assay (see, e.g., Coelho, et al. (2013) N Engl J Med 369:819). Relative TTR levels as compare dto Day 0 are shown in Figure 2.
TTR knockdown was simila inr in monkeys administered 1 mg/kg of AD-65492 and 3 mg/kg of AD- 87404.
Example 3: Administration of a Single Dose of AD-87404 to Healthy Human Subjects In a Phase I, randomized, single-blind, placebo-controlled study, AD-87404 (Sense: 5’- usgsggauUfuCfAfUfguaacca aga- 3’ (SEQ ID NO: 6), wherein an L96 ligand is conjugat edto the 3’ end of the sense strand); Antisense: 5’- usCfsuugGf(Tgn)uAfcaugAfaAfucccas -usc 3’ (SEQ ID NO: 7)) are administered to healthy human voluntee rsas a single dose of 25mg, 75 mg, 100 mg, 200 mg, or 400 mg, with possibl edose groups of 600 mg, 700 mg, 900 mg, and 1000 mg. Groups are balanced for relevant demographic characteristics, e.g., age, sex, body weight.
Demographicall maty ched control subject sare also administered a single dose of a placebo.
Plasma samples are collect anded the level of TTR protein in the samples from the subject sin the placebo group and the subject sin all of the treatment groups is determined using an ELISA assay (see, e.g., Coelho, et al. (2013) N Engl J Med 369:819) at pre-determined intervals e.g.,, days 1, 2, 3, 8, , 22, 29, 43, 57, 90, and then, for active treatment group subjects, every twenty-eighth day until the level of TTR recovers to 80% of the pre-treatment level (up through, approximatel oney year post-dose).
Level and duratio nof knockdown are determined. 56 Subjects in both AD-87404 groups and the control group are also monitored for adverse events.
Exemplar yadverse events monitored in the study include, but are not limited to, injection site erythema, injection site pain, pruritus, cough, nausea, fatigue, and abdominal painand clinica llysignifican t chang esin physical exams ,ECG, vital signs, or clinic laboal rato parametry ers, e.g., renal function, hematologic parameters, and liver function (e.g., alani neaminotransfera (ALT),se aspartate aminotransferas (ASTe )).
The results of this study demonstrate that a single subcutaneous dose of AD-87404 potentl yand durably knocks down TTR protein levels in a dose dependent manner.
Multiple dose studies and multiple ascending dose studies are also contemplated with monitoring of TTR protein knockdown in serum and adverse events.
Equivalents: Those skilled in the art will recognize, or be able to ascertain using no more than routine experimentation, many equivalent tos the specific embodiments and methods described herein. Such equivalents are intended to be encompassed by the scope of the following claims. 57
Claims (51)
1. An RNAi agent comprising a sense strand and an antisense strand, wherein: each of the sense strand and the antisense strand are independently up to 30 nucleotides in 5 length; the sense strand comprises the modified nucleotide sequence 5’- usgsggauUfuCfAfUfguaaccaaga-3‘ (SEQ ID NO: 6); and the antisense strand comprises the modified nucleotide sequence 5’- usCfsuugGf(Tgn)uAfcaugAfaAfucccasusc-3 ’ (SEQ ID NO: 7), 10 wherein a, c, g, andu are 2'-O-methyladenosine-3’ -phosphate, 2'-O-methylcytidine-3’- phosphate, 2'-O-methylguanosine-3’ -phosphate, and 2'-O-methyluridine-3’-phosphate, respectively; Af, Cf, Of, and Uf are 2‘-fluoroadenosine-3‘ -phosphate, 2‘-fluorocytidine-3‘ -phosphate, 2’- fluoroguanosine-3’-phosphate, and 2’-fluorouridine-3’-phosphate, respectively; (Tgn) is thymidine-glycol nucleic acid (GNA) S-Isomer; and 15 s is a phosphorothioate linker.
2. The RNAi agent of claim 1, wherein the sense strand of the double stranded RNAi agent is conjugated to at least one ligand. 20
3. The RNAi agent of claim 2, wherein the ligand is one or more GalNAc derivatives attached through a bivalent or trivalent branched linker.
4. The RNAi agent of claim 3, wherein the ligand is 25
5. The RNAi ligand of any one of claims 2-4, wherein the ligand is attached to the 3' end of the sense strand. 58 WO 2021/178778 PCT/US2021/021049
6. The RNAi agent of claim 5, wherein the double stranded RNAi agent is conjugated to the ligand as shown in the following schematic wherein X is O or S. 5
7. The RNAi agent of any one of claims 1-6, wherein the sense strand is 21 nucleotides in length and the antisense strand is 23 nucleotides in length.
8. A use of an RNAi agent in a method of treating a human subject suffering from a TTR- 10 associated disease, comprising administering to the subject a fixed dose of 25 mg to 1000 mg of a double stranded RNAi agent, comprising a sense strand and an antisense strand, wherein: each of the sense strand and the antisense strand are independently up to 30 nucleotides in length; the sense strand comprises the modified nucleotide sequence 5’- 15 usgsggauUfuCfAfUfguaaccaaga-3‘ (SEQ ID NO: 6); and the antisense strand comprises the modified nucleotide sequence 5’- usCfsuugGf(Tgn)uAfcaugAfaAfucccasusc-3 ’ (SEQ ID NO: 7), wherein a, c, g, andu are 2'-O-methyladenosine-3’ -phosphate, 2'-O-methylcytidine-3’- phosphate, 2'-O-methylguanosine-3’ -phosphate, and 2'-O-methyluridine-3’-phosphate, respectively; 20 Af, Cf, Of, and Uf are 2‘-fluoroadenosine-3‘ -phosphate, 2‘-fluorocytidine-3‘ -phosphate, 2’- fluoroguanosine-3’-phosphate, and 2’-fluorouridine-3’-phosphate, respectively; (Tgn) is thymidine-glycol nucleic acid (GNA) S-Isomer; and s is a phosphorothioate linker. 25
9. A use of an RNAi agent in a method of inhibiting expression of TTR in a human subject who does not meet diagnostic criteria of a TTR-associated disease, comprising administering to the subject a fixed dose of 25 mg to 1000 mg of a double stranded RNAi agent, comprising a sense strand and an antisense strand, wherein: each the sense strand and the antisense strand are independently up to 30 nucleotides in length; 59 WO 2021/178778 PCT/US2021/021049 the sense strand comprises the modified nucleotide sequence 5’- usgsggauUfuCfAfUfguaaccaaga-3’ (SEQ ID NO: 6); and the antisense strand comprises the modified nucleotide sequence 5’- usCfsuugGf(Tgn)uAfcaugAfaAfucccasusc-3 ’ (SEQ ID NO: 7), 5 wherein a, c, g, andu are 2'-O-methyladenosine-3’ -phosphate, 2'-O-methylcytidine-3’- phosphate, 2'-O-methylguanosine-3’ -phosphate, and 2'-O-methyluridine-3’-phosphate, respectively; Af, Cf, Of, and Uf are 2‘-fluoroadenosine-3‘ -phosphate, 2‘-fluorocytidine-3‘ -phosphate, 2’- fluoroguanosine-3’-phosphate, and 2’-fluorouridine-3’-phosphate, respectively; (Tgn) is thymidine-glycol nucleic acid (GNA) S-Isomer; and 10 s is a phosphorothioate linker.
10. The use of claim 8 or 9, wherein the sense strand of the double stranded RNAi agent is conjugated to at least one ligand.
11. The use of claim 10, wherein the ligand is one or more GalNAc derivatives attached 15 through a bivalent or trivalent branched linker.
12. The use of claim 11, wherein the ligand is 20
13. The use of any one of claims 9-12, wherein the ligand is attached to the 3' end of the sense strand. 25
14. The use of claim 13, wherein the double stranded RNAi agent is conjugated to the ligand as shown in the following schematic 60 WO 2021/178778 PCT/US2021/021049 wherein X is O or S.
15. The use of any one of claims 8-14, wherein the sense strand is 21 nucleotides in length and the antisense strand is 23 nucleotides in length. 5
16. A use of any one of claims 8-15, wherein the method comprises improving at least one indicia of neurological impairement, quality of life, ongoing nerve damage, or cardiovascular impairment. 10
17. The use of claim 16, wherein the indicia is a neurological impairment indicia.
18. The use of claim 17, wherein the neurological impairment indicia is a change from baseline in an indicia selected from the group of Neuropathy Impairment (NIS) score, a modified NIS 15 (mNIS+7) score, a NIS-W score, a composite autonomic symptom score (COMPASS-31), a median body mass index (mBMI) score, a 6-minute walk test (6MWT) score, and a 10-meter walk test score.
19. The use of claim 16, wherein the indicia is a quality of life indicia.
20.20. The use of claim 19, wherein the quality of life indicia is a change from baseline in an indicia selected from the group of a SF-36® health survey score, a Norfolk Quality of Life-Diabetic Neuropathy (Norfolk QOL-DN) score, and a Rasch-built Overall Disability Scale (R-ODS) score
21. The use of claim 16, wherein the indicia is ongoing nerve damage. 25
22. The use of claim 21, wherein the indicia of ongoing nerve damage is a change from baseline in a plasma protein level of one or more proteins selected from the group neurofilament light chain (NfL), RSPO3, CCDC80, EDA2R, NT-proBNP, and N-CDase. 61 WO 2021/178778 PCT/US2021/021049
23. The use of claim 21, wherein the indicia of ongoing nerve damage is a change in plasma level of neurofilament light chain (NfL) protein level.
24. The use of claim 16, wherein the indicia is a cardiovascular impairment indicia. 5
25. The use of claim 24, wherein the indicia of cardiovascular impairment is cardiovascular hospitalization, a change from baseline using Kansas City Cardiomyopathy Questionnaire Overall Summary (KCCQ-OS) with an increased score indicative of better health status, change from baseline in mean left !ventricular (LV) wall thickness by echocardiographic assessment, change from baseline in 10 global longitudinal strain by echocardiographic assessment, and change from baseline in N-terminal prohormone B-type Natriuretic Peptide (NTproBNP)
26. The use of any one of claims 8-25, wherein the human subject carries a TTR gene mutation that is associated with the development of a TTR-associated disease. 15
27. The use of any one of claims 8-26, wherein the TTR-associated disease is selected from the group consisting of senile systemic amyloidosis (SSA), systemic familial amyloidosis, familial amyloidotic polyneuropathy (FAP), familial amyloidotic cardiomyopathy (FAC), leptomeningeal/Central Nervous System (CNS) amyloidosis, hyperthyroxinemia, and cardiac amyloidosis. 20
28. The use of any one of claims 8-25, wherein the human subject has a transthyretin- mediated amyloidosis (ATTR amyloidosis) and the use of the RNAi agent method reduces an amyloid TTR deposit in the human subject. 25
29. The use of claim28, wherein the ATTR is hereditary ATTR (h-ATTR).
30. The use of claim 28, wherein the ATTR is non-heriditary ATTR (wt ATTR). 30
31. The use of any one of claims 8-30, wherein the double stranded RNAi agent is administered to the human subject by subcutaneous administration or intravenous administration.
32. The use of claim31, wherein the subcutaneous administration is self administration. 35
33. The use of claim 32, wherein the self administration is via a pre-filled syringe or auto- injector syringe. 62 WO 2021/178778 PCT/US2021/021049
34. The use of any one of claims 8-33, further comprising assessing the level of TTR mRNA expression or TTR protein expression in a sample derived from the human subject.
35. The use of any one of claims 8-34, wherein the double stranded RNAi agent is 5 administered to the human subject once every three months to once a year.
36. The use of any one of claims 8-35, wherein the fixed dose of the double stranded RNAi agent is administered to the human subject once about every three months, once every six months, or once a year. 10
37. The use of any one of claims 8-35, wherein the fixed dose of the double stranded RNAi agent is administered to the human subject at a fixed dose of 25 mg to 300 mg once every three months.
38. The use of any one of claims 8-35, wherein the fixed dose of the double stranded RNAi 15 agent is administered to the human subject at a fixed dose of 25 mg to 200 mg once every three months.
39. The use of any one of claims 8-35, wherein the fixed dose of the double stranded RNAi agent is administered to the human subject at a fixed dose of 75 mg to 200 mg once every three months.
40. The use of any one of claims 8-35, wherein the fixed dose of the double stranded RNAi 20 agent is administered to the human subject at a fixed dose of 25 mg once every three months.
41. The use of any one of claims 8-35, wherein the fixed dose of the double stranded RNAi agent is administered to the human subject at a fixed dose of 75 mg once every three months. 25
42. The use of any one of claims 8-35, wherein the fixed dose of the double stranded RNAi agent is administered to the human subject at a fixed dose of 100 mg once every three months.
43. The use of any one of claims 8-35, wherein the fixed dose of the double stranded RNAi 30 agent is administered to the human subject at a fixed dose of 200 mg once every three months.
44. The use of any one of claims 8-35, wherein the fixed dose of the double stranded RNAi agent is administered to the human subject at a fixed dose of 300 mg once every three months. 35 45. The use of any one of claims 8-35, wherein the fixed dose of the double stranded RNAi agent is administered to the human subject at a fixed dose of 400 mg to 600 mg once every six months to once every 12 months. 63
45.WO 2021/178778 PCT/US2021/021049
46. The use of any one of claims 8-35, wherein the fixed dose of the double stranded RNAi agent is administered to the human subject at a fixed dose of 400 mg to 600 mg once every six months or once every 12 months. 5
47. The use of any one of claims 8-35, wherein the fixed dose of the double stranded RNAi agent is administered to the human subject at a fixed dose of 400 mg or 600 mg once every six months or once every 12 months.
48. The use of any one of claims 8-35, wherein the fixed dose of the double stranded RNAi 10 agent is administered to the human subject at a fixed dose of 700 mg to 1000 mg once every 12 months.
49. The use of any one of claims 8-35, wherein the fixed dose of the double stranded RNAi agent is administered to the human subject at a fixed dose of 700 mg, 800 mg, 900 mg, or 1000 mg once every 12 months. 15
50. The use of any one of claims 8-49, further comprising administering to the human subject an additional therapeutic agent.
51. The use of claim 50, wherein the additional therapeutic agent is a TTR tetramer 20 stabilizer or a non-steroidal anti-inflammatory agent. 64
Applications Claiming Priority (2)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
US202062985950P | 2020-03-06 | 2020-03-06 | |
PCT/US2021/021049 WO2021178778A1 (en) | 2020-03-06 | 2021-03-05 | Compositions and methods for inhibiting expression of transthyretin (ttr) |
Publications (1)
Publication Number | Publication Date |
---|---|
IL296106A true IL296106A (en) | 2022-11-01 |
Family
ID=75252841
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
IL296106A IL296106A (en) | 2020-03-06 | 2021-03-05 | Compositions and methods for inhibiting expression of transthyretin (ttr) |
Country Status (8)
Country | Link |
---|---|
EP (1) | EP4114949A1 (en) |
JP (1) | JP2023516748A (en) |
KR (1) | KR20220152270A (en) |
CN (1) | CN115461460A (en) |
AU (1) | AU2021232029A1 (en) |
CA (1) | CA3174473A1 (en) |
IL (1) | IL296106A (en) |
WO (1) | WO2021178778A1 (en) |
Families Citing this family (5)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
IL281434B2 (en) | 2008-10-20 | 2023-09-01 | Alnylam Pharmaceuticals Inc | Compounds and methods for inhibiting transthyretin expression |
PL3329002T3 (en) | 2015-07-31 | 2021-04-19 | Alnylam Pharmaceuticals, Inc. | Transthyretin (ttr) irna compositions and methods of use thereof for treating or preventing ttr-associated diseases |
KR20200089656A (en) | 2017-09-19 | 2020-07-27 | 알닐람 파마슈티칼스 인코포레이티드 | Compositions and methods for treating transthyretin (TTR) mediated amyloidosis |
WO2023014677A1 (en) | 2021-08-03 | 2023-02-09 | Alnylam Pharmaceuticals, Inc. | Transthyretin (ttr) irna compositions and methods of use thereof |
TW202440923A (en) * | 2022-12-16 | 2024-10-16 | 美商安進公司 | Rnai constructs for inhibiting ttr expression and methods of use thereof |
Family Cites Families (84)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
JPS5927900A (en) | 1982-08-09 | 1984-02-14 | Wakunaga Seiyaku Kk | Oligonucleotide derivative and its preparation |
FR2540122B1 (en) | 1983-01-27 | 1985-11-29 | Centre Nat Rech Scient | NOVEL COMPOUNDS COMPRISING A SEQUENCE OF OLIGONUCLEOTIDE LINKED TO AN INTERCALATION AGENT, THEIR SYNTHESIS PROCESS AND THEIR APPLICATION |
US4605735A (en) | 1983-02-14 | 1986-08-12 | Wakunaga Seiyaku Kabushiki Kaisha | Oligonucleotide derivatives |
US4948882A (en) | 1983-02-22 | 1990-08-14 | Syngene, Inc. | Single-stranded labelled oligonucleotides, reactive monomers and methods of synthesis |
US4824941A (en) | 1983-03-10 | 1989-04-25 | Julian Gordon | Specific antibody to the native form of 2'5'-oligonucleotides, the method of preparation and the use as reagents in immunoassays or for binding 2'5'-oligonucleotides in biological systems |
US4587044A (en) | 1983-09-01 | 1986-05-06 | The Johns Hopkins University | Linkage of proteins to nucleic acids |
US5118802A (en) | 1983-12-20 | 1992-06-02 | California Institute Of Technology | DNA-reporter conjugates linked via the 2' or 5'-primary amino group of the 5'-terminal nucleoside |
US5258506A (en) | 1984-10-16 | 1993-11-02 | Chiron Corporation | Photolabile reagents for incorporation into oligonucleotide chains |
US5430136A (en) | 1984-10-16 | 1995-07-04 | Chiron Corporation | Oligonucleotides having selectably cleavable and/or abasic sites |
US4828979A (en) | 1984-11-08 | 1989-05-09 | Life Technologies, Inc. | Nucleotide analogs for nucleic acid labeling and detection |
US5034506A (en) | 1985-03-15 | 1991-07-23 | Anti-Gene Development Group | Uncharged morpholino-based polymers having achiral intersubunit linkages |
US4762779A (en) | 1985-06-13 | 1988-08-09 | Amgen Inc. | Compositions and methods for functionalizing nucleic acids |
US5317098A (en) | 1986-03-17 | 1994-05-31 | Hiroaki Shizuya | Non-radioisotope tagging of fragments |
JPS638396A (en) | 1986-06-30 | 1988-01-14 | Wakunaga Pharmaceut Co Ltd | Poly-labeled oligonucleotide derivative |
US4904582A (en) | 1987-06-11 | 1990-02-27 | Synthetic Genetics | Novel amphiphilic nucleic acid conjugates |
US5585481A (en) | 1987-09-21 | 1996-12-17 | Gen-Probe Incorporated | Linking reagents for nucleotide probes |
US5525465A (en) | 1987-10-28 | 1996-06-11 | Howard Florey Institute Of Experimental Physiology And Medicine | Oligonucleotide-polyamide conjugates and methods of production and applications of the same |
DE3738460A1 (en) | 1987-11-12 | 1989-05-24 | Max Planck Gesellschaft | MODIFIED OLIGONUCLEOTIDS |
US5082830A (en) | 1988-02-26 | 1992-01-21 | Enzo Biochem, Inc. | End labeled nucleotide probe |
US5109124A (en) | 1988-06-01 | 1992-04-28 | Biogen, Inc. | Nucleic acid probe linked to a label having a terminal cysteine |
US5262536A (en) | 1988-09-15 | 1993-11-16 | E. I. Du Pont De Nemours And Company | Reagents for the preparation of 5'-tagged oligonucleotides |
US5512439A (en) | 1988-11-21 | 1996-04-30 | Dynal As | Oligonucleotide-linked magnetic particles and uses thereof |
US5599923A (en) | 1989-03-06 | 1997-02-04 | Board Of Regents, University Of Tx | Texaphyrin metal complexes having improved functionalization |
US5457183A (en) | 1989-03-06 | 1995-10-10 | Board Of Regents, The University Of Texas System | Hydroxylated texaphyrins |
US5391723A (en) | 1989-05-31 | 1995-02-21 | Neorx Corporation | Oligonucleotide conjugates |
US4958013A (en) | 1989-06-06 | 1990-09-18 | Northwestern University | Cholesteryl modified oligonucleotides |
US5451463A (en) | 1989-08-28 | 1995-09-19 | Clontech Laboratories, Inc. | Non-nucleoside 1,3-diol reagents for labeling synthetic oligonucleotides |
US5254469A (en) | 1989-09-12 | 1993-10-19 | Eastman Kodak Company | Oligonucleotide-enzyme conjugate that can be used as a probe in hybridization assays and polymerase chain reaction procedures |
US5292873A (en) | 1989-11-29 | 1994-03-08 | The Research Foundation Of State University Of New York | Nucleic acids labeled with naphthoquinone probe |
US5486603A (en) | 1990-01-08 | 1996-01-23 | Gilead Sciences, Inc. | Oligonucleotide having enhanced binding affinity |
US7037646B1 (en) | 1990-01-11 | 2006-05-02 | Isis Pharmaceuticals, Inc. | Amine-derivatized nucleosides and oligonucleosides |
US6783931B1 (en) | 1990-01-11 | 2004-08-31 | Isis Pharmaceuticals, Inc. | Amine-derivatized nucleosides and oligonucleosides |
US5578718A (en) | 1990-01-11 | 1996-11-26 | Isis Pharmaceuticals, Inc. | Thiol-derivatized nucleosides |
US5214136A (en) | 1990-02-20 | 1993-05-25 | Gilead Sciences, Inc. | Anthraquinone-derivatives oligonucleotides |
AU7579991A (en) | 1990-02-20 | 1991-09-18 | Gilead Sciences, Inc. | Pseudonucleosides and pseudonucleotides and their polymers |
EP0455905B1 (en) | 1990-05-11 | 1998-06-17 | Microprobe Corporation | Dipsticks for nucleic acid hybridization assays and methods for covalently immobilizing oligonucleotides |
US5138045A (en) | 1990-07-27 | 1992-08-11 | Isis Pharmaceuticals | Polyamine conjugated oligonucleotides |
US5608046A (en) | 1990-07-27 | 1997-03-04 | Isis Pharmaceuticals, Inc. | Conjugated 4'-desmethyl nucleoside analog compounds |
US5489677A (en) | 1990-07-27 | 1996-02-06 | Isis Pharmaceuticals, Inc. | Oligonucleoside linkages containing adjacent oxygen and nitrogen atoms |
US5602240A (en) | 1990-07-27 | 1997-02-11 | Ciba Geigy Ag. | Backbone modified oligonucleotide analogs |
US5218105A (en) | 1990-07-27 | 1993-06-08 | Isis Pharmaceuticals | Polyamine conjugated oligonucleotides |
US5688941A (en) | 1990-07-27 | 1997-11-18 | Isis Pharmaceuticals, Inc. | Methods of making conjugated 4' desmethyl nucleoside analog compounds |
US5245022A (en) | 1990-08-03 | 1993-09-14 | Sterling Drug, Inc. | Exonuclease resistant terminally substituted oligonucleotides |
US5512667A (en) | 1990-08-28 | 1996-04-30 | Reed; Michael W. | Trifunctional intermediates for preparing 3'-tailed oligonucleotides |
JP3172178B2 (en) | 1990-11-08 | 2001-06-04 | ハイブライドン インコーポレイテッド | Incorporation of multiple reporter groups into synthetic oligonucleotides |
US5371241A (en) | 1991-07-19 | 1994-12-06 | Pharmacia P-L Biochemicals Inc. | Fluorescein labelled phosphoramidites |
US5565552A (en) | 1992-01-21 | 1996-10-15 | Pharmacyclics, Inc. | Method of expanded porphyrin-oligonucleotide conjugate synthesis |
US5595726A (en) | 1992-01-21 | 1997-01-21 | Pharmacyclics, Inc. | Chromophore probe for detection of nucleic acid |
US5272250A (en) | 1992-07-10 | 1993-12-21 | Spielvogel Bernard F | Boronated phosphoramidate compounds |
EP0654077A4 (en) | 1992-07-17 | 1996-03-13 | Ribozyme Pharm Inc | Method and reagent for treatment of animal diseases. |
US5574142A (en) | 1992-12-15 | 1996-11-12 | Microprobe Corporation | Peptide linkers for improved oligonucleotide delivery |
US6294664B1 (en) | 1993-07-29 | 2001-09-25 | Isis Pharmaceuticals, Inc. | Synthesis of oligonucleotides |
US5597696A (en) | 1994-07-18 | 1997-01-28 | Becton Dickinson And Company | Covalent cyanine dye oligonucleotide conjugates |
US5580731A (en) | 1994-08-25 | 1996-12-03 | Chiron Corporation | N-4 modified pyrimidine deoxynucleotides and oligonucleotide probes synthesized therewith |
US6576752B1 (en) | 1997-02-14 | 2003-06-10 | Isis Pharmaceuticals, Inc. | Aminooxy functionalized oligomers |
JP3756313B2 (en) | 1997-03-07 | 2006-03-15 | 武 今西 | Novel bicyclonucleosides and oligonucleotide analogues |
US6770748B2 (en) | 1997-03-07 | 2004-08-03 | Takeshi Imanishi | Bicyclonucleoside and oligonucleotide analogue |
US6794499B2 (en) | 1997-09-12 | 2004-09-21 | Exiqon A/S | Oligonucleotide analogues |
NZ503765A (en) | 1997-09-12 | 2002-04-26 | Exiqon As | Bi-cyclic and tri-cyclic nucleotide analogues |
US6320017B1 (en) | 1997-12-23 | 2001-11-20 | Inex Pharmaceuticals Corp. | Polyamide oligomers |
US7084125B2 (en) | 1999-03-18 | 2006-08-01 | Exiqon A/S | Xylo-LNA analogues |
CN1349541A (en) | 1999-05-04 | 2002-05-15 | 埃克西库恩公司 | L-ribo-LNA analogues |
US6525191B1 (en) | 1999-05-11 | 2003-02-25 | Kanda S. Ramasamy | Conformationally constrained L-nucleosides |
DE60119562T2 (en) | 2000-10-04 | 2007-05-10 | Santaris Pharma A/S | IMPROVED SYNTHESIS OF PURIN-BLOCKED NUCLEIC ACID ANALOGUE |
AU2003291753B2 (en) | 2002-11-05 | 2010-07-08 | Isis Pharmaceuticals, Inc. | Polycyclic sugar surrogate-containing oligomeric compounds and compositions for use in gene modulation |
AU2003290597A1 (en) | 2002-11-05 | 2004-06-03 | Isis Pharmaceuticals, Inc. | Modified oligonucleotides for use in rna interference |
WO2005021570A1 (en) | 2003-08-28 | 2005-03-10 | Gene Design, Inc. | Novel artificial nucleic acids of n-o bond crosslinkage type |
DK1984381T3 (en) | 2006-01-27 | 2010-11-01 | Isis Pharmaceuticals Inc | 6-modified bicyclic nucleic acid analogues |
US7569686B1 (en) | 2006-01-27 | 2009-08-04 | Isis Pharmaceuticals, Inc. | Compounds and methods for synthesis of bicyclic nucleic acid analogs |
CA2651453C (en) | 2006-05-11 | 2014-10-14 | Isis Pharmaceuticals, Inc. | 5'-modified bicyclic nucleic acid analogs |
US8278425B2 (en) | 2007-05-30 | 2012-10-02 | Isis Pharmaceuticals, Inc. | N-substituted-aminomethylene bridged bicyclic nucleic acid analogs |
US8278426B2 (en) | 2007-06-08 | 2012-10-02 | Isis Pharmaceuticals, Inc. | Carbocyclic bicyclic nucleic acid analogs |
CA2692579C (en) | 2007-07-05 | 2016-05-03 | Isis Pharmaceuticals, Inc. | 6-disubstituted bicyclic nucleic acid analogs |
EP4074344A1 (en) | 2007-12-04 | 2022-10-19 | Arbutus Biopharma Corporation | Targeting lipids |
IL281434B2 (en) | 2008-10-20 | 2023-09-01 | Alnylam Pharmaceuticals Inc | Compounds and methods for inhibiting transthyretin expression |
WO2011005861A1 (en) | 2009-07-07 | 2011-01-13 | Alnylam Pharmaceuticals, Inc. | Oligonucleotide end caps |
US8927513B2 (en) | 2009-07-07 | 2015-01-06 | Alnylam Pharmaceuticals, Inc. | 5′ phosphate mimics |
PL2563920T3 (en) | 2010-04-29 | 2017-08-31 | Ionis Pharmaceuticals, Inc. | Modulation of transthyretin expression |
DK3301177T3 (en) | 2011-11-18 | 2020-06-15 | Alnylam Pharmaceuticals Inc | RNAI AGENTS, COMPOSITIONS AND METHODS OF USING ITS FOR THE TREATMENT OF TRANSTHYRETINE (TTR) -related diseases |
IL296543B2 (en) | 2013-05-01 | 2025-02-01 | Ionis Pharmaceuticals Inc | Compositions and methods for modulating hbv and ttr expression |
CA2932904A1 (en) | 2013-12-06 | 2015-06-11 | Dicerna Pharmaceuticals, Inc. | Methods and compositions for the specific inhibition of transthyretin (ttr) by double-stranded rna |
IL316808A (en) * | 2014-08-20 | 2025-01-01 | Alnylam Pharmaceuticals Inc | Modified double-stranded rna agents and uses thereof |
PL3329002T3 (en) | 2015-07-31 | 2021-04-19 | Alnylam Pharmaceuticals, Inc. | Transthyretin (ttr) irna compositions and methods of use thereof for treating or preventing ttr-associated diseases |
US20190350962A1 (en) | 2016-12-16 | 2019-11-21 | Alnylam Pharmaceuticals, Inc. | METHODS FOR TREATING OR PREVENTING TTR-ASSOCIATED DISEASES USING TRANSTHYRETIN (TTR) iRNA COMPOSITIONS |
-
2021
- 2021-03-05 IL IL296106A patent/IL296106A/en unknown
- 2021-03-05 KR KR1020227034708A patent/KR20220152270A/en active Search and Examination
- 2021-03-05 JP JP2022553196A patent/JP2023516748A/en active Pending
- 2021-03-05 CA CA3174473A patent/CA3174473A1/en active Pending
- 2021-03-05 EP EP21714746.1A patent/EP4114949A1/en active Pending
- 2021-03-05 WO PCT/US2021/021049 patent/WO2021178778A1/en unknown
- 2021-03-05 AU AU2021232029A patent/AU2021232029A1/en active Pending
- 2021-03-05 CN CN202180030650.9A patent/CN115461460A/en active Pending
Also Published As
Publication number | Publication date |
---|---|
KR20220152270A (en) | 2022-11-15 |
AU2021232029A1 (en) | 2022-10-06 |
WO2021178778A1 (en) | 2021-09-10 |
CN115461460A (en) | 2022-12-09 |
EP4114949A1 (en) | 2023-01-11 |
CA3174473A1 (en) | 2021-09-10 |
JP2023516748A (en) | 2023-04-20 |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
IL296106A (en) | Compositions and methods for inhibiting expression of transthyretin (ttr) | |
US20240075052A1 (en) | Compositions and methods for treating transthyretin (ttr) mediated amyloidosis | |
US20240230644A1 (en) | Methods of treating transthyretin (ttr) mediated amyloidosis | |
ES2640260T3 (en) | Compositions and methods to inhibit gene expression alas1 | |
JP7058656B2 (en) | Methods for Treating or Preventing TTR-Related Diseases with Transthyretin (TTR) iRNA Compositions | |
US20230272382A1 (en) | COMPLEMENT COMPONENT C3 iRNA COMPOSITIONS AND METHODS OF USE THEREOF FOR TREATING OR PREVENTING COMPLEMENT COMPONENT C3-ASSOCIATED DISEASES | |
KR20230162998A (en) | Complement component c5 irna compositions and methods of use thereof | |
US20240218369A1 (en) | Compositions and methods for treating transthyretin (ttr) mediated amyloidosis | |
CN111108201A (en) | Antisense oligonucleotide that binds to exon 51 of human dystrophin pre-mRNA | |
IL301712A (en) | SNCA IRNA preparations and methods of using them to treat or prevent neurodegenerative diseases associated with SNCA | |
WO2022260939A2 (en) | Compositions and methods for treating or preventing stargardt's disease and/or retinal binding protein 4 (rbp4)-associated disorders | |
EP2594275B1 (en) | Prophylactic or therapeutic agent for diabetes | |
US11208653B2 (en) | Increasing RNAi efficiency through single nucleotide mismatches | |
WO2024216109A1 (en) | Formulations for oligonucleotide delivery | |
KR20240166577A (en) | SNCA-Targeting SIRNA Compositions for Treating SNCA-Associated Diseases | |
TW202446402A (en) | Formulations for oligonucleotide delivery | |
WO2024129931A1 (en) | ALPHA-V BETA-6(ανβ6) INTEGRIN LIGANDS FOR EXTRAHEPATIC DELIVERY | |
US20220340911A1 (en) | Rnai constructs for inhibiting slc30a8 expression and methods of use thereof | |
WO2006039792A1 (en) | Method of treating autoimmune diseases |